3,158
Views
0
CrossRef citations to date
0
Altmetric
Review

The prevention and management strategies for neonatal chronic lung disease

&
Pages 143-154 | Received 07 Nov 2022, Accepted 20 Feb 2023, Published online: 28 Feb 2023

ABSTRACT

Introduction

Survival from even very premature birth is improving, but long-term respiratory morbidity following neonatal chronic lung disease (bronchopulmonary dysplasia (BPD)) has not reduced. Affected infants may require supplementary oxygen at home, because they have more hospital admissions particularly due to viral infections and frequent, troublesome respiratory symptoms requiring treatment. Furthermore, adolescents and adults who had BPD have poorer lung function and exercise capacity.

Areas covered

Antenatal and postnatal preventative strategies and management of infants with BPD. A literature review was undertaken using PubMed and Web of Science.

Expert opinion

There are effective preventative strategies which include caffeine, postnatal corticosteroids, vitamin A, and volume guarantee ventilation. Side-effects, however, have appropriately caused clinicians to reduce use of systemically administered corticosteroids to infants only at risk of severe BPD. Promising preventative strategies which need further research are surfactant with budesonide, less invasive surfactant administration (LISA), neurally adjusted ventilatory assist (NAVA) and stem cells. The management of infants with established BPD is under-researched and should include identifying the optimum form of respiratory support on the neonatal unit and at home and which infants will most benefit in the long term from pulmonary vasodilators, diuretics, and bronchodilators.

1. Introduction

Survival from even very premature birth is improving in many countries, but long-term respiratory morbidity following neonatal chronic lung disease or bronchopulmonary dysplasia (BPD) has not reduced [Citation1]. BPD was originally described in infants with severe respiratory disease who required high peak pressures and levels of supplemental oxygen during mechanical ventilation resulting in fibrosis and emphysematous alveolar areas. Routine use of antenatal steroids and postnatal surfactant decreased the incidence of respiratory distress syndrome (RDS) and mortality [Citation2,Citation3], but not BPD. The subsequent description of ‘new’ BPD highlighted that prematurely infants who did not receive artificial ventilation could require supplementary oxygen at 36 weeks post-menstrual age (PMA) [Citation4]. There have been many definitions of BPD, a consensus statement in 2001 defined BPD as oxygen dependence at 28 days and then the severity was determined at 36 weeks PMA as mild, moderate, or severe depending on the level of respiratory support then required [Citation4]. The cardinal feature of new BPD is arrested development with simplification of the lungs – impairment of both alveolarization and vascularization of the pulmonary vessels. These changes are in part driven by a systemic inflammatory cascade. BPD is most commonly seen in infants born prematurely but can also occur in infants with conditions associated with pulmonary hypoplasia, such as congenital diaphragmatic or anterior wall defect. Poor intrauterine growth is associated with an increased risk of BPD [Citation5] and worse lung function at follow-up [Citation6,Citation7]. There, however, have been conflicting results from studies investigating whether chorioamnionitis increases the risk of BPD. In some, the risk disappeared once the earlier gestational age at birth of the chorioamnionitis exposed infants was taken into account. A large meta-analysis [Citation8] demonstrated a significant relationship between chorioamnionitis and BPD, and there is some evidence that wheezing may be increased at follow-up of infants exposed to chorioamnionitis [Citation9]. Males have a higher incidence of BPD, which relates to their more immature lung structure at birth, and they have poorer lung function in childhood and adulthood [Citation10,Citation11].

A worldwide review showed that BPD was a global problem, but the incidence reported by different countries varied between 10% and 89% [Citation12]. In Europe and North America, the incidence has been reported to be as high as 80% in those born at the lowest gestations.

BPD occurs more commonly in those infants born prematurely (less than 37 weeks of gestational age) or very low birthweight (VLBW less than 2500 gms) than those born at term. It particularly occurs in those born extremely premature (less than 29 weeks of gestational age) or extremely low birthweight (ELBW less than 1000gms).

The chronic lung disease related to prematurity has been shown to have consequences beyond discharge from the neonatal unit. Infants may require supplementary oxygen at home, because they have more hospital admissions than those without BPD particularly due to viral infections and have frequent, troublesome respiratory symptoms requiring treatment. Furthermore, studies of lung function in adolescents [Citation10] and adults [Citation13] have shown that those who had BPD had poorer lung function compared to those who did not. Adults born prematurely who developed BPD had poorer airway function compared to those without BPD and those born at term [Citation11]. At 35 years, those born extremely preterm or extremely low birthweight had six times the risk for chronic obstructive pulmonary disease [Citation14,Citation15]. In addition, they have poorer exercise capacity [Citation16]; 39 young people born before 29 weeks of gestation had an abnormal forced expiratory flow at 75% (FEF75) (<5th percentile) with a reduced sprint distance of 114 m and 27 having a very abnormal FEF75 (<2.5 percentile) had a reduced sprint distance of 159 m. Putting those results in context, adults in a cardiac rehabilitation program undertaking regular modified shuttle sprint tests had a modified shuttle sprint test difference of 70 m [Citation17], thus a difference of 159 m is clinically significant [Citation17]. Puberty is the last positive effector of lung function with rapid growth of the airways and lung parenchyma [Citation18], yet, studies have demonstrated that lung function may deteriorate during puberty in those who had BPD [Citation19,Citation20], particularly if they had received postnatal corticosteroids [Citation21]. It is important to reduce this respiratory burden. Furthermore, BPD has been associated with adverse neurodevelopmental outcomes. In a case series of 250 infants from a single center, those who had developed grade two or three BPD had worse neurodevelopmental outcomes in a grade-dependent manner. In another single center, retrospective study increased BPD severity was associated with an increased risk of neurodevelopmental delay at both 2 and 5 years [Citation22,Citation23]. The aim of this review, therefore, is to discuss possible preventative and management BPD strategies.

2. Methods

A literature review was undertaken using PubMed and the Web of Science with the search terms in the subheadings of the review, adding ‘neonatal,’ ‘infant,’ ‘premature,’ or ‘bronchopulmonary dysplasia’ as appropriate. Article abstracts were then screened to identify relevant articles and those with relevant findings pertinent to the expert review included ‘chronic lung disease’ or ‘bronchopulmonary dysplasia’ as appropriate. Article abstracts were then screened to identify relevant articles and those with relevant findings pertinent to the expert review included. Further articles were added to the review if cited in articles identified from the literature search. Seminal articles were included where appropriate. Meta-analyses and Cochrane reviews were preferred in the context of this review; however, individual studies were examined where data were of relevance or conflicting. Recent studies involving infants exposed to modern neonatal interventions such as antenatal corticosteroids and postnatal surfactant were used in preference. The literature search was performed independently by both authors and the results then shared in the context of writing this review.

3. Antenatal preventative strategies

3.1. Antenatal corticosteroids and thyroid stimulating hormone

Antenatal corticosteroids administered to mothers at risk of preterm delivery reduce infant mortality. A Cochrane review, however, demonstrated no significant difference in the risk of BPD (risk ratio (RR), 0.86; 95% confidence interval, 0.61–1.22) [Citation24]. This may reflect that antenatal corticosteroids, as intra-amniotic endotoxin, impairs secondary septation [Citation25]. Thyroid releasing hormone showed promising results in lambs, resulting in increased surfactant production and lung compliance. In human studies, no such benefit was seen and there was a suggestion of delayed motor development [Citation26].

3.2. Sildenafil

Sildenafil has been trialed as an intervention to improve fetal growth and reduce the associated neonatal morbidity. In a meta-analysis of eight small, randomized controlled trials (RCTs), enrolling 576 infants in total, doses between 3 and 20 g per day of sildenafil citrate were given to mothers in whom poor intrauterine fetal growth was identified. Growth was improved (Standardized Mean Difference 0.41, 95% CI 0.24 to 0.58) and the gestation of pregnancy lengthened (SMD 0.30, 95% CI 0.07, 0.51) [Citation27]. There were, however, no significant differences in the infant clinical outcomes investigated (mortality and respiratory distress syndrome); rates of BPD were not reported. An interim analysis of a large randomized trial of Sildenafil (the STRIDER trial [Citation28]) led to the halting of the trial after 11 infants died in the treatment arm compared to none in the placebo arm [Citation29].

4. Postnatal preventative strategies

4.1. Surfactant therapy

Surfactant production is detected in the fetal lung from the 22 weeks of gestation, but only reaches maturity at 33 to 34 weeks [Citation30]. Surfactant deficiency can result in severe RDS and death in prematurely born infants [Citation31]. Surfactant administration to treat RDS reduces mortality, but not BPD [Citation2]. A comparison of prophylactic and selective surfactant administration [Citation32] demonstrated a non-significant trend in reducing the requirement for oxygen at 28 days in favor of selective use, but there was no significant difference in the requirement for oxygen at 36 weeks corrected weeks gestational age. In the TOLSURF randomized trial of infants less than 29 days of gestational age who continued to require mechanical ventilation between 7 and 14 days after birth, infants received inhaled nitric oxide with either surfactant or a placebo every 1 to 3 days up to a maximum of five doses. There was no significant effect of late surfactant use on survival without BPD development [Citation33], but at 1 year of age infants who received late surfactant required fewer post-discharge hospital admissions for respiratory problems [Citation34].

The mode of surfactant delivery has also been examined [Citation35,Citation36]. In a meta-analysis of six trials recruiting in total 895 infants born between 23 and 34 weeks of gestation, who were randomized to either intubation (including INSURE only and those ventilated beyond the delivery room) versus less invasive surfactant administration (LISA), showed that LISA undertaken once infants were admitted to the neonatal unit was associated with a reduced risk ratio of the combined outcome of BPD or death [Citation37]. In a subsequent study of 485 infants with a gestational age of 25 to 28 weeks with RDS, minimally invasive surfactant administration when the infants required 30% or more of supplemental oxygen was not associated with a significant reduction in death or BPD. In another study, infants given LISA in the delivery suit had a lower respiratory rate and oxygen requirement following surfactant administration compared to historical controls, suggesting that LISA reduced respiratory distress [Citation38]. LISA in delivery suite was also demonstrated to reduce days of ventilation and BPD compared to routine care [Citation39]. No trials, however, have compared delivery suite LISA to later administration. In addition, there remains significant variation in LISA techniques, premedication use, and subsequent respiratory management [Citation40].

4.2. Respiratory support

Invasive mechanical respiratory support can be lifesaving but can increase the risk of BPD, particularly if high pressures and/or high-inspired oxygen levels are used. Studies, therefore, have focused on providing alternative treatments to invasive mechanical support or by limiting the damage done by such support.

4.2.1. Noninvasive mechanical ventilation

Continuous positive airway pressure as an alternative to intubation and ventilation was not shown to reduce the incidence of BPD in one systematic review [Citation41]. A more recent review, however, demonstrated that although there was insufficient evidence to evaluate prophylactic continuous positive airway pressure (CPAP) compared to oxygen therapy, when compared to mechanical ventilation, it reduced BPD and the combined outcome of death and BPD [Citation42]. Synchronized noninvasive positive pressure ventilation (sNIPPV) reduced the need for reintubation following extubation from mechanical ventilation compared to CPAP alone [Citation43], as well as reducing BPD, although this outcome was reported in only three small trials.

Heated humidified high flow nasal cannula (HHHFNC) oxygen has increased in popularity within neonatal units over recent years. A systematic review of 13 studies, however, showed no benefit in the prevention of BPD when using HHHFNC compared to CPAP when used as the primary mode of respiratory support or after extubation [Citation44]. HHHFNC can facilitate earlier feeding with faster weight gain and weaning from supplementary oxygen in those infants requiring support beyond 32 weeks [Citation45].

4.2.2. Volume targeted ventilation (VTV)

In vitro and pre-clinical studies showed that high tidal volumes during mechanical ventilation led to release of inflammatory interleukins and subsequent damage to the lung parenchyma. Tidal volumes of 4 to 7 ml/kg are normally targeted during VTV, whereas without volume targeting, volumes of up to 10 ml/kg can be delivered [Citation46]. In a Cochrane review, VTV compared to pressure-limited, time-cycled ventilation reduced the incidence of BPD within the prematurely born population [Citation47]. In infants with a birthweight less than one kilo, however, there was no significant difference in the rate of BPD. The range of tidal volumes used in the studies varied significantly and lower tidal volumes can be pro-inflammatory as can higher tidal volumes [Citation48]. Furthermore, during acute respiratory distress, higher rather than lower volumes within the tidal volume range were associated with a lower work of breathing [Citation49,Citation50]. Among infants with evolving or established BPD, a target tidal volume of 7mls/kg was associated with a reduced work of breathing significantly below baseline [Citation51], likely reflecting the increased physiological dead space of infants with BPD.

4.2.3. High frequency oscillation ventilation

High-frequency oscillation ventilation (HFOV) results in smaller tidal volumes being delivered to an infant than during conventional ventilation [Citation52]. In vitro studies have shown that the inflammatory interleukin release from cells undergoing oscillation-like stretch was lower than cells stretched in a way that mimicked conventional ventilation [Citation53]. A Cochrane review highlighted that prophylactic HFOV was associated with a significant, but borderline reduction in moderate-to-severe BPD [Citation54], but the study designs differed, as did the oscillators and most importantly the control respiratory support used. A subsequent individual patient meta-analysis found no significant difference in the rates of BPD between those receiving HFOV or conventional mechanical ventilation (CMV) [Citation55]. The UKOS trial randomized 797 infants born at less than 29 weeks of gestation to receive either conventional or oscillatory ventilation within an hour of birth [Citation56]. The study found no significant difference in the primary combined outcome of BPD or death, however, subsequent follow-up at 11 to 14 years of age showed better airway function in those who received HFOV [Citation57]. When examined again at 16 to 19 years of age, the differences were smaller and no longer statistically significant suggesting that pubertal growth may have caused the conventionally supported group’s lung function to catch up [Citation58].

The use of volume-targeted high-frequency oscillation (HFOV-VG) has been suggested to reduce the inflammatory effect of mechanical ventilation. There are, however, no randomized studies comparing HFOV-VG with conventional ventilation, but one retrospective study highlighted a reduction in BPD [Citation59].

4.2.4. Neurally adjusted ventilator assist

In neurally adjusted ventilator assist (NAVA), the ventilator trigger and ‘assistance’ are governed by the electrical activity of the diaphragm measured by electrodes on a modified nasogastric tube. Crossover studies of infants with evolving or established BPD have demonstrated that NAVA, when compared to conventional ventilation, resulted in a lower mean airway pressure requirement, a better oxygen index, and a better oxygenation as measured by the alveolar-arterial gradient [Citation60,Citation61]. A Cochrane review comparing NAVA to other triggered modes of ventilation found only one eligible randomized trial [Citation62]. In that study of 60 infants, whilst ventilator parameters improved on NAVA used as rescue support compared to the control group, there was no significant difference seen in the rate of BPD [Citation63]. A single center study comparing the outcomes of infants who received noninvasive or invasive NAVA with that of historical controls reported no significant difference in the incidence of BPD, but there was a reduction in the overall length of stay on the neonatal unit (112 days versus 140 days, p = 0.019) [Citation61].

4.3. Supplementary oxygen

Hyperoxia has been shown to induce abnormal vascular growth lung tissue in vitro and in animal studies [Citation64]. Abnormal vascular growth has been shown to be a key component of the pathophysiology of new BPD [Citation65]. It has been suggested that this abnormal vascularization mirrors changes seen in retinopathy of prematurity [Citation66], which is reduced by reducing the exposure of preterm infants to supplemental oxygen [Citation67]. It is, therefore, reasonable to suspect that reducing the exposure of infants born prematurely to supplemental oxygen might reduce the incidence of BPD; however, this has not been shown in randomized trials. In a meta-analysis examining exposure to high versus low levels of supplementary oxygen during resuscitation, there was no significant difference in rates of BPD [Citation68]. Furthermore, in a multicenter study randomizing infants born at less than 28 weeks gestation to higher or lower oxygen saturation targets on the neonatal unit, there were no significant differences seen in rates of BPD; however, more infants died in the group with lower oxygen saturation targets [Citation69]. Thus, despite the benefits seen in vitro and in animal studies, reducing the exposure of preterm infants to supplemental oxygen requires a cautious approach given the risk of increased mortality.

4.4. Inhaled nitric oxide

A Cochrane review of inhaled nitric oxide use in preterm infants with respiratory failure examined three different timings of the intervention: early treatment (within 3 days) for impaired oxygenation (eight trials), routine use in infants requiring respiratory support (four trials), and later use in those at an increased risk of BPD (three trials) [Citation70]. No benefits to receiving iNO were seen regarding any of the timings of iNO administration. There were, however, trends toward positive outcomes, and it could be argued that more select entry criteria to better define a high-risk population might result in more positive results. A trial of 800 infants born between 24 and 28 + 6 weeks of gestational age included those with mild disease, that is requiring surfactant or CPAP for RDS within the first 24 hours of birth. They were randomized to inhaled nitric oxide (iNO) or placebo at 5 ppm for a minimum of 7 days and a maximum of 21 days. No significant differences were found in BPD development or death [Citation71]. At 7-year follow-up, rates of rehospitalisation and use of respiratory medications were similar between the two groups [Citation72].

4.5. Postnatal sildenafil

The use of sildenafil to prevent BPD has been poorly studied. The effects of sildenafil in a rat model were encouraging with increased alveolarisation, improved pulmonary vascularization, reduced inflammation, and reduced right ventricular hypertrophy [Citation73]. Whilst there is some evidence for the use of sildenafil in term infants with pulmonary hypertension where inhaled nitric oxide therapy is unavailable [Citation74], a systematic review of its use in pulmonary hypertension associated with BPD failed to show any benefit beyond a short-term reduction in oxygen requirement [Citation75]. In 40 infants born between 24.0 and 29.7 weeks of gestation randomized to receive oral sildenafil (0.5 mg/kg every 6 hours or a placebo), no significant differences were seen between the two groups in mortality, respiratory support at 36 weeks or side-effects [Citation76]. Concerns had been raised regarding the use of sildenafil in children after a trial recruiting children aged between 1 and 17 years with pulmonary hypertension demonstrated an increased mortality in those who received high-dose sildenafil [Citation77]. A randomized control trial is currently underway to investigate the safety of sildenafil in the neonatal population at different doses [Citation78]. The trial will investigate respiratory outcomes as a secondary outcome measure.

4.6. Fluid restriction

Fluid restriction has been suggested to result in a reduction in the incidence of BPD in that when reduced volumes of fluid were given to ventilated infants as part of a package of respiratory care, the incidence of BPD was lower [Citation79]. In a RCT including infants at high risk of developing BPD, routinely exposed to antenatal corticosteroids and postnatal surfactant, fluid restriction was not associated with a reduction in BPD, but fewer of the fluid restricted group required treatment with postnatal corticosteroids. Colloid infusion, however, was associated with an increased duration of oxygen dependency [Citation80]. A Cochrane review highlighted the lack of studies investigating the effects of fluid restriction on the development of BPD [Citation81].

4.7. Patent ductus arteriosus closure

A patent ductus arteriosus (PDA) for more than 10 days has been shown to increase the risk of BPD [Citation82]. Whilst administering ibuprofen has been shown to successfully close PDAs, a Cochrane review did not find any evidence that it reduced the incidence of BPD when given prophylactically [Citation83]. A multicentre, randomized trial attempted to recruit only those infants whose ducts were judged as haemodynamically significant enrolled 337 infants to receive either ibuprofen or no treatment [Citation84]. The trial did not demonstrate any improvement in respiratory status with regard to ventilator days or the durations of noninvasive respiratory support or supplementary oxygen. There remains controversy regarding PDA management and its role in the pathogenesis of respiratory disease [Citation85]. In a recently published multicentre RCT which included 273 infants, expectant management for PDA in infants born at less than 28 weeks of gestation was non-inferior to early ibuprofen treatment with respect to BPD [Citation86].

4.8. Caffeine

Caffeine acts by increasing central respiratory drive stimulating diaphragmatic contractility has a diuretic action [Citation87] and has anti-inflammatory properties [Citation88,Citation89]. In the Caffeine for Apnea of Prematurity (CAP) trial, infants were randomized if methylxanthines were thought to be required for prevention or treatment of apnea and/or facilitation of removal of an endotracheal tube in the first 10 days after birth. The primary outcome of death or neurodevelopmental disability (NDI) determined at 18 to 21 months was significantly lower in the caffeine group who also had a lower BPD rate [Citation90]. Caffeine use was associated with a lower PDA incidence [Citation91], which may in part have been responsible for the reduction in BPD. Early initiation of caffeine therapy was associated with a greater reduction in ventilation duration [Citation92].

4.9. Vitamin A

Vitamin A is a group of fat-soluble compounds which maintain the integrity of epithelial cells of the respiratory tract. Premature infants are vitamin A deficient [Citation93], and lower vitamin A levels have been documented in those who subsequently developed BPD [Citation94]. A Cochrane review of vitamin A supplementation in very low birth weight infants or those born before 32 weeks of gestational age demonstrated Vitamin A administration was associated with a small, but significant reduction in BPD. This, however, requires repeated intramuscular injections [Citation95]. Furthermore, a systematic review of 17 studies suggested that the benefit of vitamin A supplementation is likely to be limited to infants with baseline vitamin A intake less than 1500 IU per kg per day [Citation96].

5. Macrolides

The inflammatory cascade has been shown to have an important role in the development of BPD with higher levels of inflammatory cytokines such as IL-6 and TNF-α in those who went on to develop severe BPD or die [Citation97]. Colonization of the respiratory tract with Ureaplasma urealyticum has been associated with an increased incidence of BPD [Citation98]. Ureaplasma urealyticum has been shown to induce inflammatory cytokine production including IL-6 and TNF-α both systemically and within the lungs [Citation99,Citation100]. It has also been shown to promote apoptosis of lung epithelial cells [Citation101]. Azithromycin inhibits neutrophil migration, chemotaxis [Citation102] and the subsequent release of inflammatory interleukins such as IL-6 and TNF-α [Citation103]. It is effective in reducing the bacterial load of Ureaplasma urealyticum in preterm infants [Citation104]. A meta-analysis of three RCTs involving 319 premature infants randomized to either azithromycin or placebo showed that prophylactic azithromycin resulted in a borderline reduction of BPD. A further meta-analysis of 529 patients randomized to azithromycin or placebo failed to demonstrate a reduction in BPD but did demonstrate a significant reduction in the length of supplemental oxygen requirement [Citation105]. The meta-analysis, however, was very heterogenous, including trials utilizing short or long courses of azithromycin (less than or more than 7 days), and infants who did not require ventilation. Only one study reported longer-term respiratory outcomes and found no significant differences in respiratory or neurodevelopmental outcomes at 2 years of age [Citation106]. In a trial of 220 infants, a subgroup analysis of the effect of azithromycin in those colonized with Ureaplasma urealyticum (76 patients) [Citation107] demonstrated that there was a reduction in BPD but not a difference in Ureaplasma urealyticum colonization between the two groups. A larger multicentre, randomized trial of azithromycin versus placebo (AZTEC trial) has completed recruitment, and the results are awaited [Citation108]. A single center trial of another macrolide, clarithromycin, in infants colonized by Ureaplasma urealyticum on day 3 after birth, showed a benefit in reducing BPD (2.9% versus 36.4%) [Citation109].

5.1. Antioxidants

One of the multifactorial causes implicated in BPD development has been oxygen toxicity, not least as prematurely born infants have low antioxidant defenses. The enzyme superoxide dismutase (SOD) dismutates the toxic superoxide radical into the less toxic hydrogen peroxide. Nevertheless, a Cochrane review highlighted that, in two RCTs, SOD did not significantly reduce death or oxygen dependency at 28 days or 36 weeks, but, in one, there was a lower frequency of respiratory problems after discharge [Citation110]. A systematic review of 26 trials demonstrated that vitamin E supplementation reduced the risk of intracranial hemorrhage but increased the risk of sepsis so is not recommended [Citation111]. Further RCTs are required to identify if antioxidant supplementation will reduce BPD and more importantly improve long-term respiratory outcomes in prematurely born infants.

5.2. Postnatal corticosteroids

5.2.1. Dexamethasone

Corticosteroids are frequently administered to ventilator-dependent infants with the hope of improving respiratory function and facilitating extubation. Whilst given early (before 7 days after birth) corticosteroids reduced the incidence of BPD or death, there was an increased risk of hyperglycemia, hypertension, gastrointestinal bleeding, gastrointestinal perforation and growth failure [Citation112]. Late administration of postnatal corticosteroids was also associated with a reduction of BPD or death [Citation113] and whilst the incidence of hyperglycemia was increased, there were no significant differences in gastrointestinal complications or growth. It should be noted that a number of those in the placebo arms of the included studies subsequently received corticosteroids. The duration of the courses included in the meta-analysis varied, but one study demonstrated that oxygen diffusion continued to improve throughout a 9-day course; however, immature infants were less responsive to postnatal corticosteroids [Citation114]. In an RCT which included infants of less than 28 weeks of gestational age, a 42-day tapering course compared to a 9-day course with rapid tapering was associated with decreased hospital morbidities and increased rate of survival without handicap. Only 59 infants, however, were included in the study and given the known side-effects of corticosteroids, it would be important to further explore the effects of a prolonged duration of corticosteroids before this regime is introduced into routine clinical practice [Citation115]. Ventilated infants who go onto develop BPD frequently receive multiple courses, but there is a paucity of data on the efficacy of a second course of systemic postnatally administered corticosteroids. In a retrospective study, a second course was associated with improvements in oxygenation and successful weaning from mechanical ventilation [Citation116]. Acutely, growth impairment can occur as a side-effect of systemically administered corticosteroids. In a whole population study of 6,104 preterm infants with BPD, of whom 28.3% received corticosteroids no significant differences were seen in weight gain or head circumference growth from birth to discharge in those that did or did not receive corticosteroids [Citation117]. Lung function, however, was worse at 11 to 14 years and 16 to 19 years in those that received corticosteroids while on the NICU. The mean lung function was lower as the number of courses of dexamethasone increased [Citation118]. In the unexposed group, lung function improved between 11 to 14 and 16 to 19 years but forced expiratory flow at 75% of the expired vital capacity and forced expiratory volume in 1 second deteriorated in those who received postnatal corticosteroids likely increasing their risk of the premature onset of chronic obstructive pulmonary disease [Citation118]. Secondary analysis of the Preterm Erythropoietin Neuroprotective (PENUT) RCT demonstrated that long duration and higher cumulative dose of dexamethasone were associated with worse neurodevelopmental scores at a corrected age of 2 years [Citation119].

5.2.2. Hydrocortisone

Hydrocortisone has been investigated as an alternative to dexamethasone due to the improved neurological side effect profile associated with hydrocortisone in laboratory studies [Citation120]. Studies of hydrocortisone in prematurely born infants, however, have yielded conflicting results [Citation121]. A study of hydrocortisone within French neonatal units suggested a significant benefit in survival without BPD among the 256 infants randomized to low-dose prophylactic hydrocortisone compared to the 257 who were not [Citation122]. A subsequent trial of 800 infants in the US, who were randomized to low-dose hydrocortisone on day 14 or placebo, showed no benefit associated with receiving low-dose hydrocortisone and a higher incidence of hypertension [Citation123]. A meta-analysis of studies utilizing hydrocortisone highlighted an increased risk of late onset sepsis and gastrointestinal perforation [Citation124].

5.2.3. Inhaled corticosteroids

Locally acting corticosteroids delivered by inhalation or nebulization have been suggested to reduce the incidence of BPD. A study of 863 infants randomized to either budesonide or placebo, however, showed that whilst BPD was reduced in those treated with budesonide, there was a non-significant increase in mortality [Citation125]. Follow-up of the cohort demonstrated that the increase in mortality among those who received budesonide had become statistically significant [Citation126].

Budesonide given with surfactant at delivery has also been studied. The rationale for combining budesonide with surfactant is related to the increase in inflammatory interleukins in those who go on to develop BPD [Citation97] and the increased solubility of budesonide when combined with surfactant [Citation127]. In a randomized, multicentre study in which 265 infants born at less than 29 weeks gestation were randomized to either surfactant combined with budesonide or surfactant alone, the incidence of death or BPD was reduced in those who received the combination therapy [Citation128]. In the trial, tracheal aspirates were sent from a subset of 40 infants and lower levels of inflammatory interleukins (IL-1, IL-6 and IL-8) were observed in the intervention arm [Citation128]. At 2 years of age, however, there were no significant differences in the number of infants with respiratory symptoms or requiring respiratory medications [Citation128]. In a study in which the outcomes of infants who received budesonide and surfactant were compared to historical controls, there were no significant differences in neurodevelopmental outcomes at 4 to 6 months or 18 to 22 months corrected age [Citation129].

5.3. Stem cells

The lungs’ endogenous stem and progenitor cells which regenerate normal alveolarization and pulmonary vascular development are dysfunctional in BPD [Citation130]. Mesenchymal stem cells (MSCs) are thought to possess potent immunomodulatory and secretory properties acting in a paracrine manner to modulate endogenous responses to injury and repair [Citation131]. In a phase 1 clinical trial, the safety and feasibility of MSC transplantation was reported [Citation132]. In a subsequent randomized controlled phase 11 trial, infants born between 23 and 28 weeks of gestation deteriorating on mechanical ventilation at 5 to 14 days were randomly allocated to receive stem cells or placebo. The primary outcome of death or BPD was not significantly improved with MSC transplantation, but in a secondary analysis, the incidence of severe BPD in the infants born at 23 to 24 weeks gestation was significantly lower, 19% compared to 53% [Citation133].

6. Management

6.1. Diuretics

Diuretics have been studied in those developing or with established BPD. Two Cochrane reviews highlighted that whilst a transient improvement in respiratory status was seen up to a week after starting diuretics, there was no evidence that diuretics improved respiratory outcomes beyond this time [Citation134,Citation135]. A retrospective review over a five-year period in a single center in the era of routine antenatal corticosteroids and postnatal surfactant showed that diuretic use was associated with electrolyte abnormalities and reduction in weight gain with only slight improvement in short term respiratory status [Citation136]. A systematic review included eight randomized studies of preterm infants less than 3 weeks of age with evolving or established BPD randomly allocated to receive an aerosolized loop diuretic or not. A single dose transiently improved respiratory mechanics, but there was no information available on the effects of chronic administration effects [Citation137].

6.2. Inhaled bronchodilators

In a retrospective review of 1429 infants born at less than 29 weeks of gestational age, 33% had received bronchodilators. The frequency and duration of the courses were variable between centers [Citation138], likely reflecting the lack of evidence of positive long-term benefit. Studies in ventilated infants have shown only short-term benefits, yet prolonged ventilation (>54 days) best predicted bronchodilator use [Citation138].

6.3. Leukotriene receptor blockade

Montelukast is a selective leukotriene receptor antagonist of cysteinyl leukotriene, which causes bronchoconstriction, mucus secretion, airway hyperactivity, and increased vascular permeability [Citation139]. In a non-randomized study, montelukast given daily for at least 3 weeks in 11 infants was associated with improved survival compared to controls and the pulmonary severity score and the duration of ventilation were lower [Citation140]. A subsequent prospective, multicentre, RCT of 66 infants born at less than 32 weeks of gestational age with a postnatal age greater than 14 days, all ventilator or supplementary oxygen dependent demonstrated no significant difference in the incidence of moderate-to-severe BPD (43.4% versus 52.8%, p = 0.912) [Citation141].

6.4. Anti-hypertensives

The incidence of systemic hypertension has been reported to be between 7% and 43% [Citation142]. Infants have increased aorta wall thickness and stiffness [Citation143], hypertrophy of the heart and reduced cardiac function [Citation144]. Captopril, an angiotensin-converting enzyme inhibitor, may improve endothelial function and nitric oxide release [Citation142]. In a case series of six infants with severe BPD unresponsive to sildenafil and diuretics, 5 weeks after commencing treatment with captopril, there was a significant reduction in the mean fraction of inspired oxygen and ventilatory requirements with a reduction in aorta intima media thickness [Citation142]. Clearly, further studies are required to determine the optimum management of hypertension in this population.

6.5. Pulmonary hypertension

The incidence of pulmonary hypertension (PH) in infants with BPD ranges from 17% to 37%. In a single center study, the development of PH was associated with a lower gestational age and ventilatory support 28 days after birth. Mortality was increased in that group, as was length and healthcare cost of stay, after adjusting for gestational age [Citation145]. The treatment of PH is based on consensus opinion, and few randomized trials have been undertaken [Citation146]. It has been suggested that the mainstay of treatment of PH in BPD is pulmonary vascular dilatation, prevention of further lung damage, offloading the right ventricle, prevention of coronary ischemia, and improved quality of life [Citation147]. Whilst some treatments such as sildenafil have been shown to reduce the degree of pulmonary hypertension [Citation148], there is little evidence that this has improved overall outcomes or reduced the incidence of BPD. Indeed, one study of sildenafil showed that in the 21 infants with pulmonary hypertension, although the degree of pulmonary hypertension improved when infants received sildenafil, there was no improvement in pulmonary gas exchange or respiratory severity score [Citation149].

A historical cohort study of 20 infants evaluated to a median age of 2 years gave an overall survival rate of 95% in those treated with either sildenafil or Bosentan [Citation150]. Whilst the authors identified this was a lower mortality rate than other reported observational studies, they acknowledged that there were significant differences in their population, in particular the degree of severity of pulmonary hypertension.

These data would suggest that pulmonary hypertension occurs as a result of the lung damage associated with BPD but altering the degree of hypertension does not reverse or treat the underlying chronic lung disease. There is insufficient evidence to state that longer term outcomes are improved.

6.6. Nutrition

Infants with significant respiratory morbidity have an increased basal metabolic rate requiring increased calorie intake to preserve normal growth velocities [Citation151]. Furthermore, it has been shown that delayed introduction of proteins and nutrition in a preterm population is associated with poor growth [Citation152]. In one trial investigating the effect of early standardized, concentrated, macronutrients, parenteral nutrition in infants born less than 29 weeks, there was no significant difference reported in the rates of BPD at 36 weeks between the 74 infants in the intervention arm and the 76 infants in the standard care arm [Citation153]. The intervention group, however, had better growth at 36 weeks PMA (mean difference 0.40, 95% CI 0.005 to 0.79). The role of nutrition in the development of BPD is multifactorial [Citation154], but those data suggest that simply improving growth may not improve respiratory outcomes.

6.7. Home respiratory support

The majority of infants are usually able to be weaned off home oxygen before 12 months of age [Citation154–158]. Exposure to secondhand smoke, however, has been associated with a trend for longer duration of supplemental home oxygen [Citation154]. Infants with severe BPD when ventilated at home have to be able to trigger a portable ventilator [Citation159]. Portable ventilators may have less rapid response times. It is, therefore, important to transition infants early on to portable home ventilators so that the most appropriate ventilator can be identified. Studies are required to develop devices to optimize home ventilation.

6.8. Immunization

Respiratory syncytial virus (RSV) is an important cause of readmission, morbidity, and mortality among preterm infants [Citation160]. A monoclonal antibody, palivizumab, has been shown to reduce hospital admissions and admissions to intensive care [Citation161]. The cost of immunization is substantial, and therefore cost analyses have been performed to mitigate the costs by targeting those most at risk of the severe consequences of RSV infection, for example, infants requiring home oxygen at discharge [Citation162].

7. Expert opinion

Bronchopulmonary dysplasia (BPD) is the most common long-term adverse outcome of very premature delivery and is associated with chronic respiratory morbidity. It should be noted, however, that extremely prematurely born infants who did not develop BPD may also suffer such a long-term adverse respiratory outcome. As a consequence, it is important to assess the long-term efficacy of all treatment strategies in extremely preterm infants regardless of the development of BPD. Prediction of infants at high risk of chronic respiratory morbidity could facilitate targeting of interventions within and out with randomized controlled trials. Cluster analysis may provide a method of identifying discrete groups of prematurely born infants with differing respiratory outcomes during the first 7 years. In a study of 168 infants, readily available clinical data were used to classify them into hierarchical agglomerative clusters according to their birth weight and duration of neonatal ventilation into the risk of subsequent viral infections [Citation163]. This approach merits further study as it is possible that interventions such as iNO if delivered to a high-risk population might improve respiratory outcomes.

A number of strategies have been shown to reduce the incidence of BPD, but given its multifactorial nature, there is unlikely to be a single magic bullet to prevent BPD. It will be therefore important to investigate a combination of such strategies. In the meanwhile, it is important to introduce those preventative strategies without adverse effects into routine care such as caffeine and volume targeted ventilation (VTV) but with the appropriate VT levels. Other preventative strategies require further research before they should be introduced into routine care such as NAVA, stem cells and surfactant with budesonide. Stem cell administration has delivered promising results, but there are possible long-term adverse outcomes and therefore this needs particularly investigating.

Very few strategies have been robustly assessed in infants with evolving or established BPD. The growing number of BPD infants means this is now a crucial priority. Appropriate multicentre RCTs of novel modes of ventilation will be needed to establish the optimum mode of respiratory support for that population. It is now clear that many such babies have pulmonary hypertension and mandate that infants with evolving and established BPD should be routinely and regularly screened and the optimum treatment identified. Diuretics and bronchodilators are over-prescribed, and evidence-based guidelines need to be produced. Quality improvement projects will hopefully result in the appropriate use of diuretics, bronchodilators, and corticosteroids.

The chronic respiratory morbidity of infants who develop BPD and those with pulmonary hypoplasia, for example infants with congenital diaphragmatic hernia, means it is essential that it is the primary outcome rather than BPD or survival without BPD. Funders of randomized trials need to be further persuaded of the importance of this outcome. Equally, it will be important to define chronic respiratory morbidity with regard to what matters to the infant, their parents and healthcare utilization.

Article highlights

BPD preventative strategies:

  • There are a number of preventative strategies which have been shown in randomized trials to reduce BPD, and these include systemically administered corticosteroids, vitamin A, caffeine, and volume guarantee ventilation.

  • Promising interventions or those where further evidence is required are surfactant administered with budesonide, LISA, inhaled nitric oxide (iNO), NAVA and stem cells.

Management of infants with evolving or established BPD:

  • Should be screened for pulmonary hypertension.

  • Systematic corticosteroids should only be considered in those who remain ventilator dependent on high pressures and oxygen levels who have made no progress over the first 2 weeks despite the absence of a patent ductus arteriosus (PDA) or infection.

  • Diuretics should be given to those not tolerating standard fluid volumes with poor growth.

  • Regular bronchodilators should only be given to those infants who have shown a positive response, i.e. a reduction in respiratory support.

Declaration of interest

A Greenough has held grants from various manufacturers (Abbott Laboratories, MedImmune) and ventilator manufacturers (SLE). A Greenough has received honoraria for giving lectures and advising various manufacturers (Abbott Laboratories, MedImmune) and ventilator manufacturers (SLE). A Greenough has received a non-conditional educational grant from SLE. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Costeloe KL, Hennessy EM, Haider S, et al. Short term outcomes after extreme preterm birth in England: comparison of two birth cohorts in 1995 and 2006 (the EPICure studies). BMJ. 2012;345(dec04 3):e7976.
  • Soll RF. Prophylactic natural surfactant extract for preventing morbidity and mortality in preterm infants. Cochrane Database Syst Rev. 2000;2:CD000511.
  • Crowley PA. Antenatal corticosteroid therapy: a meta-analysis of the randomized trials, 1972 to 1994. Am J Obstet Gynecol. 1995;173:322–335.
  • Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2001;163:1723–1729.
  • Charles E, Hunt KA, Harris C, et al. Small for gestational age and extremely low birth weight infant outcomes. J Perinat Med. 2019;47:247–251.
  • Harris C, Lunt A, Bisquera A, et al. Intrauterine growth retardation and lung function of very prematurely born young people. Pediatr Pulmonol. 2021;56:2284–2291.
  • Greenough A, Yuksel B, Cheeseman P. Effect of in utero growth retardation on lung function at follow up of prematurely born infants. Eur Respir J. 2004;24:731–733.
  • Villamor-Martinez E, Alvarez-Fuente M, Ghazi AM, et al. Association of chorioamnionitis with bronchopulmonary dysplasia among preterm infants: a systematic review, meta-analysis and metaregression. JAMA Network Open. 2019;2:e1914611.
  • Wang X, Zhang LH, Zhu SQ, et al. Histological chorioamnionitis is associated with an increased risk of wheezing in preterm children less than 34 gestational weeks. BMC Pediatr. 2021;21:104.
  • Harris C, Morris S, Lunt A, et al. Influence of bronchopulmonary dysplasia on lung function in adolescents who were born extremely prematurely. Pediatr Pulmonol. 2022;57:3151–3157.
  • Hurst JR, Beckmann J, Ni Y, et al. Respiratory and cardiovascular outcomes in survivors of extremely preterm birth at 19 years. Am J Respir Crit Care Med. 2020;202:422–432.
  • Siffel C, Kistler KD, Lewis JFM, et al. Global incidence of bronchopulmonary dysplasia among extremely preterm infants: a systematic literature review. J Matern Fetal Neonatal Med. 2021;34:1721–1731.
  • Yang J, Kingsford RA, Horwood J, et al. Lung function of adults born at very low birth weight. Pediatrics. 2020;145:e20192359.
  • Saarenpaa H-K, Tikanmaki M, Sipola-Leppanen M, et al. Lung function in very low birth weight adults. Pediatrics. 2015;136:642–650.
  • Bardsen T, Roksund OD, Benestad MR, et al. Tracking of lung function from 10 to 35 years after being born extremely preterm or with extremely low birth weight. Thorax. 2022;77:790–798.
  • Harris C, Lunt A, Bisquera A, et al. Lung function and exercise capacity in prematurely born young people. Pediatr Pulmonol. 2020;55:2289–2295.
  • Houchen-Wolloff L, Boyce S, Singh S. The minimum clinically important improvement in the incremental shuttle walk test following cardiac rehabilitation. Eur J Prev Cardiol. 2015;22:972–978.
  • Smith J, Emerson S, Kurti S, et al. Lung volume and expiratory flow rates from pre- to post-puberty. Eur J Appl Physiol. 2015;115:1645–1652.
  • Doyle LW, Adams A-M, Robertson C, et al. Increasing airway obstruction from 8 to 18 years in extremely preterm/low-birthweight survivors born in the surfactant era. Thorax. 2017;72:712–719.
  • Bisquera A, Harris C, Lunt A, et al. Longitudinal changes in lung function in very prematurely born young people receiving high-frequency oscillation or conventional ventilation from birth. Pediatr Pulmonol. 2022;57:1489–1496.
  • Harris C, Bisquera A, Zivanovic S, et al. Postnatal dexamethasone exposure and lung function in adolescents born very prematurely. PloS One. 2020;15:e0237080.
  • Katz TA, Vliegenthart RJ, Aarnoudse-Moens CS, et al. Severity of bronchopulmonary dysplasia and neurodevelopmental outcome at 2 and 5 years corrected age. J Pediatr. 2022;234:40–46.
  • Oluwole I, Tan JB, DeSouza S, et al. The association between bronchopulmonary dysplasia grade and risks of adverse neurodevelopmental outcomes among preterm infants born at less than 30 weeks of gestation. J Matern Fetal Neonatal Med. 2023;36:2167074.
  • Roberts D, Dalziel S. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2006;3:CD004454.
  • Willet KE, Jobe AH, Ikegami M, et al. Antenatal endotoxin and glucocorticoid effects on lung morphometry in preterm lambs. Pediatr Res. 2000;48:782–788.
  • Crowther CA, Alfirevic Z, Han S, et al. Thyrotropin‐releasing hormone added to corticosteroids for women at risk of preterm birth for preventing neonatal respiratory disease. Cochrane Database Syst Rev. 2013;11:CD000019.
  • Chen J, Gong X, Chen P, et al. Effect of L-arginine and sildenafil citrate on intrauterine growth restriction fetuses: a meta-analysis. BMC Pregnancy Childbirth. 2016;16:225.
  • Sharp A, Cornforth C, Jackson R, et al. Maternal sildenafil for severe fetal growth restriction (STRIDER): a multicentre, randomised, placebo-controlled, double-blind trial. Lancet Child Adolesc Health. 2018;2:93–102.
  • Hawkes N. Trial of Viagra for fetal growth restriction is halted after baby deaths. BMJ. 2018;362:k3247.
  • Fleisher B, Kulovich MV, Hallman M, et al. Lung profile: sex differences in normal pregnancy. Obstet Gynecol. 1985;66:327–330.
  • Avery ME, Mead J. Surface properties in relation to atelectasis and hyaline membrane disease. AMA J Dis Child. 1959;97:517–523.
  • Rojas-Reyes MX, Morley CJ, Soll R. Prophylactic versus selective use of surfactant in preventing morbidity and mortality in preterm infants. Cochrane Database Syst Rev. 2012;3:CD000510.
  • Ballard RA, Keller RL, Black DM, et al. Randomized trial of late surfactant treatment in ventilated preterm infants receiving inhaled nitric oxide. J Pediatr. 2016;168:23–29.
  • Hascoet JM, Picaud JC, Ligi I, et al. Late surfactant administration in very preterm neonates with prolonged respiratory distress and pulmonary outcome at 1 year of age: a randomized clinical trial. JAMA Pediatr. 2016;170:365–372.
  • Ambulkar H, Dassios T, Greenough A. Evaluation of methods of surfactant administration in the delivery suite? Arch Med Sci. 2023;19:116–121.
  • Dargaville PA, Carlin JB, Davis PG. OPTIMIST A trial investigators. Minimally invasive surfactant therapy vs sham treatment and death or bronchopulmonary dysplasia in preterm infants with respiratory distress syndrome. JAMA. 2022;327:1614–1615.
  • Aldana-Aguirre JC, Pinto M, Featherstone RM, et al. Less invasive surfactant administration versus intubation for surfactant delivery in preterm infants with respiratory distress syndrome: a systematic review and meta-analysis. Arch Dis Child Fetal Neonatal Ed. 2017;102:F17.
  • Ambulkar H, Williams EE, Hickey A, et al. Respiratory monitoring during less invasive surfactant administration in the delivery suite. Early Hum Dev. 2021;154:105311.
  • Fms AT, Ambulkar H, Williams E, et al. Outcomes following less-invasive-surfactant-administration in the delivery-room. Early Hum Dev. 2022;167:105562.
  • Jeffreys E, Hunt K, Dassios T, et al. UK survey of less invasive surfactant administration. Arch Dis Child Fetal Neonatal Ed. 2019;104:F567.
  • Schmolzer GM, Kumar M, Pichler G, et al. Non-invasive versus invasive respiratory support in preterm infants at birth: systematic review and meta-analysis. BMJ. 2013;347:F5980.
  • Subramaniam P, Ho JJ, Davis PG. Prophylactic or very early initiation of continuous positive airway pressure (CPAP) for preterm infants. Cochrane Database Syst Rev. 2021;10:CD001243.
  • Lemyre B, Davis PG, De Paoli AG, et al. Nasal intermittent positive pressure ventilation (NIPPV) versus nasal continuous positive airway pressure (NCPAP) for preterm neonates after extubation. Cochrane Database Syst Rev. 2014;9:CD003212.
  • de Jesus Brito S, Tsopanoglou SP, Galvão EL, et al. Can high-flow nasal cannula reduce the risk of bronchopulmonary dysplasia compared with CPAP in preterm infants? A systematic review and meta-analysis. BMC Pediatr. 2021;21:407.
  • Shetty S, Hunt K, Douthwaite A, et al. High-flow nasal cannula oxygen and nasal continuous positive airway pressure and full oral feeding in infants with bronchopulmonary dysplasia. Arch Dis Child Fetal Neonatal Ed. 2016;101:F408–F411.
  • Peng W, Zhu H, Shi H, et al. Volume-targeted ventilation is more suitable than pressure-limited ventilation for preterm infants: a systematic review and meta-analysis. Arch Dis Child Fetal Neonatal Ed. 2014;99:F158–F165.
  • Klingenberg C, Wheeler KI, McCallion N, et al. Volume‐targeted versus pressure‐limited ventilation in neonates. Cochrane Database Syst Rev. 2017;10:CD003666.
  • Lista G, Castoldi F, Fontana P, et al. Lung inflammation in preterm infants with respiratory distress syndrome: effects of ventilation with different tidal volumes. Pediatr Pulmonol. 2006;41:357–363.
  • Patel DS, Rafferty GF, Lee S, et al. Work of breathing and volume targeted ventilation in respiratory distress. Arch Dis Child Fetal Neonatal Ed. 2010;95:F443–F446.
  • Patel DS, Sharma A, Prendergast M, et al. Work of breathing and different levels of volume-targeted ventilation. Pediatrics. 2009;123:e679–e684.
  • Hunt K, Dassios T, Ali K, et al. Volume targeting levels and work of breathing in infants with evolving or established bronchopulmonary dysplasia. Arch Dis Child Fetal Neonatal Ed. 2019;104:F46–F49.
  • Dimitriou G, Cheeseman P, Greenough A. Lung volume and the response to high volume strategy, high frequency oscillation. Acta Paediatr. 2004;93:613–617.
  • Harris C, Thorpe SD, Rushwan S, et al. An in vitro investigation of the inflammatory response to the strain amplitudes which occur during high frequency oscillation ventilation and conventional mechanical ventilation. J Biomech. 2019;88:186–189.
  • Cools F, Offringa M, Askie LM. Elective high frequency oscillatory ventilation versus conventional ventilation for acute pulmonary dysfunction in preterm infants. Cochrane Database Syst Rev. 2015;3:CD000104.
  • Cools F, Askie LM, Offringa M, et al. Elective high-frequency oscillatory versus conventional ventilation in preterm infants: a systematic review and meta-analysis of individual patients’ data. Lancet. 2010;375:2082–2091.
  • Johnson AH, Peacock JL, Greenough A, et al. High-frequency oscillatory ventilation for the prevention of chronic lung disease of prematurity. N Engl J Med. 2002;347:633–642.
  • Zivanovic S, Peacock J, Alcazar-Paris M, et al. Late outcomes of a randomized trial of high-frequency oscillation in neonates. N Engl J Med. 2014;370:1121–1130.
  • Harris C, Bisquera A, Lunt A, et al. Outcomes of the neonatal trial of high-frequency oscillation at 16 to 19 years. N Engl J Med. 2020;383:689–691.
  • Ramos-Navarro C, González-Pacheco N, Rodríguez-Sánchez de la BA, et al. Effect of a new respiratory care bundle on bronchopulmonary dysplasia in preterm neonates. Eur J Pediatr. 2020;179:1833–1842.
  • Hunt KA, Dassios T, Greenough A. Proportional assist ventilation (PAV) versus neurally adjusted ventilator assist (NAVA): effect on oxygenation in infants with evolving or established bronchopulmonary dysplasia. Eur J Pediatr. 2020;179:901–908.
  • Shetty S, Hunt K, Peacock J, et al. Crossover study of assist control ventilation and neurally adjusted ventilatory assist. Eur J Pediatr. 2017;176:509–513.
  • Rossor TE, Hunt KA, Shetty S, et al. Neurally adjusted ventilatory assist compared to other forms of triggered ventilation for neonatal respiratory support. Cochrane Database Syst Rev. 2017;10:CD012251.
  • Kallio M, Koskela U, Peltoniemi O, et al. Neurally adjusted ventilatory assist (NAVA) in preterm newborn infants with respiratory distress syndrome—a randomized controlled trial. Eur J Pediatr. 2016;175:1175–1183.
  • Wickramasinghe L, Tsantikos E, Lau M, et al. Lung and eye pathology develop concurrently in neonatal supplemental oxygen model. Eur Respir J. 2019;54:OA274.
  • Silva DM, Nardiello C, Pozarska A, et al. Recent advances in the mechanisms of lung alveolarization and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2015;309:L1239–L1272.
  • Wickramasinghe LC, van Wijngaarden P, Tsantikos E, et al. The immunological link between neonatal lung and eye disease. Clin Transl Immunol. 2021;10:e1322.
  • Flynn JT, Bancalari E, Snyder ES, et al. A Cohort study of transcutaneous oxygen tension and the incidence and severity of retinopathy of prematurity. N Engl J Med. 1992;326:1050–1054.
  • Welsford M, Nishiyama C, Shortt C, et al. Initial oxygen use for preterm newborn resuscitation: a systematic review with meta-analysis. Pediatrics. 2019;143:e20181828.
  • Stenson BJ, Tarnow-Mordi WO, Darlow BA, et al. Oxygen saturation and outcomes in preterm infants. N Engl J Med. 2013;368:2094–2104.
  • Barrington KJ, Finer N, Pennaforte T. Inhaled nitric oxide for respiratory failure in preterm infants. Cochrane Database Syst Rev. 2017;1:CD000509.
  • Mercier JC, Hummler H, Durrmeyer X, et al. Inhaled nitric oxide for prevention of bronchopulmonary dysplasia in premature babies (EUNO): a randomised controlled trial. Lancet. 2010;376:346–354.
  • Greenough A, Decobert F, Field D, et al. Inhaled nitric oxide (iNO) for preventing prematurity-related bronchopulmonary dysplasia (BPD): 7-year follow-up of the European Union Nitric Oxide (EUNO) trial. J Perinat Med. 2020;49:104–110.
  • de Visser YP, Walther FJ, Laghmani El H, et al. Sildenafil attenuates pulmonary inflammation and fibrin deposition, mortality and right ventricular hypertrophy in neonatal hyperoxic lung injury. Respir Res. 2009;10:30.
  • Kelly LE, Ohlsson A, Shah PS. Sildenafil for pulmonary hypertension in neonates. Cochrane Database Syst Rev. 2017;8:CD005494.
  • van der Graaf M, Rojer LA, Helbing W, et al. EXPRESS: sildenafil for bronchopulmonary dysplasia and pulmonary hypertension: a meta-analysis. Pulm Circ. 2019;9:2045894019837875.
  • Abounahia FF, Abu-Jarir R, Abounahia MF, et al. prophylactic sildenafil in preterm infants at risk of bronchopulmonary dysplasia: a pilot randomized, double-blinded, placebo-controlled trial. Clin Drug Investig. 2019;39:1093–1107.
  • Barst RJ, Ivy DD, Gaitan G, et al. A randomized, double-blind, placebo-controlled, dose-ranging study of oral sildenafil citrate in treatment-naive children with pulmonary arterial hypertension. Circulation. 2012;125:324–334.
  • Schneider S, Bailey M, Spears T, et al. Safety of sildenafil in premature infants with severe bronchopulmonary dysplasia (SILDI-SAFE): a multicenter, randomized, placebo-controlled, sequential dose-escalating, double-masked, safety study. BMC Pediatr. 2020;20:559.
  • Tooley WH. Epidemiology of bronchopulmonary dysplasia. J Pediatr. 1979;95:851–858.
  • Kavvadia V, Greenough A, Dimitriou G, et al. Randomised trial of fluid restriction in ventilated very low birthweight infants. Arch Dis Child Fetal Neonatal Ed. 2000;83:F91–F96.
  • Barrington KJ, Fortin-Pellerin E, Pennaforte T. Fluid restriction for treatment of preterm infants with chronic lung disease. Cochrane Database Syst Rev. 2017;2:CD005389.
  • Clyman RI, Hills NK, Cambonie G, et al. Patent ductus arteriosus, tracheal ventilation, and the risk of bronchopulmonary dysplasia. Pediatr Res. 2022;91:652–658.
  • Ohlsson A, Walia R, Shah SS. Ibuprofen for the treatment of patent ductus arteriosus in preterm or low birth weight (or both) infants. Cochrane Database Syst Rev. 2020;2:CD003481.
  • J-C R, Cambonie G, Le Thuaut A, et al. Effect of early targeted treatment of ductus arteriosus with ibuprofen on survival without cerebral palsy at 2 years in infants with exer: a randomized clinical trial. J Pediatr. 2021;233:33–42.e2.
  • Hundscheid T, El-Khuffash A, McNamara PJ, et al. Survey highlighting the lack of consensus on diagnosis and treatment of patent ductus arteriosus in prematurity. Eur J Pediatr. 2022;181:2459–2468.
  • Hundscheid T, Onland W, Kooi EM, et al. Expectant management of early Ibuprofen for patent ductus arteriosus. N Engl J Med. 2022. DOI:10.1056/NEJMoa2207418.
  • Nobile S, Carnielli VP. Caffeine for preterm infants: current indications and uncertainties. Acta Biomed. 2015;86:32–35.
  • Weichelt U, Cay R, Schmitz T, et al. Prevention of hyperoxia-mediated pulmonary inflammation in neonatal rats by caffeine. Eur Respir J. 2013;41:966–973.
  • Fehrholz M, Speer CP, Kunzmann S. Caffeine and rolipram affect Smad signalling and TGF-β1 stimulated CTGF and transgelin expression in lung epithelial cells. PLoS One. 2014;9:e97357.
  • Schmidt B, Roberts RS, Davis P, et al. Long-term effects of caffeine therapy for apnea of prematurity. N Engl J Med. 2007;357:1893–1902.
  • Patel RM, Leong T, Carlton DP, et al. Early caffeine therapy and clinical outcomes in extremely preterm infants. J Perinatol. 2013;33:134–140.
  • Davis PG, Schmidt B, Roberts RS, et al. Caffeine for Apnea of Prematurity trial: benefits may vary in subgroups. J Pediatr. 2010;156:382–387.
  • Shenai JP, Chytil F, Stahlman MT. Vitamin A status of neonates with bronchopulmonary dysplasia. Pediatr Res. 1985;19:185–188.
  • EAG AG, Alsharany W, Ali AA, et al. Anti-oxidant profiles and markers of oxidative stress in preterm neonates. Paediatr Int Child Health. 2016;36:134–140.
  • Darlow BA, Graham PJ, Rojas-Reyes MX. Vitamin A supplementation to prevent mortality and short and long-term morbidity in very low birth weight infants. Cochrane Database Syst Rev. 2016;8:CD000501.
  • Rakshasbhuvankar AA, Pillow J, Simmer KN, et al. Vitamin A supplementation in very preterm or very low birthweight infants to prevent morbidity and mortality: a systematic review and meta-analysis of randomised trials. Am J Clin Nutr. 2021;114:2084–2096.
  • Ambalavanan N, Carlo WA, D’Angio CT, et al. Cytokines associated with bronchopulmonary dysplasia or death in extremely low birth weight infants. Pediatrics. 2009;123:1132–1141.
  • Lowe J, Watkins WJ, Edwards MO, et al. Association between pulmonary ureaplasma colonization and bronchopulmonary dysplasia in preterm infants: updated systematic review and meta-analysis. Pediatr Infect Dis J. 2014;33:697–702.
  • Cheah FC, Winterbourn CC, Darlow BA, et al. Nuclear factor kappaB activation in pulmonary leukocytes from infants with hyaline membrane disease: associations with chorioamnionitis and Ureaplasma urealyticum colonization. Pediatr Res. 2005;57:616–623.
  • Kotecha S, Hodge R, Schaber JA, et al. Pulmonary Ureaplasma urealyticum is associated with the development of acute lung inflammation and chronic lung disease in preterm infants. Pediatr Res. 2004;55(1):61–68.
  • Li Y-H, Chen M, Brauner A, et al. Ureaplasma urealyticum induces apoptosis in human lung epithelial cells and macrophages. Biol Neonate. 2002;82(3):166–173.
  • Parnham MJ, Culic O, Erakovic V, et al. Modulation of neutrophil and inflammation markers in chronic obstructive pulmonary disease by short-term azithromycin treatment. Eur J Pharmacol. 2005;517(1–2):132–143.
  • Aghai ZH, Kode A, Saslow JG, et al. Azithromycin suppresses activation of nuclear factor-kappa B and synthesis of pro-inflammatory cytokines in tracheal aspirate cells from premature infants. Pediatr Res. 2007;62(4):483–488.
  • Viscardi RM, Terrin ML, Magder LS, et al. Randomised trial of azithromycin to eradicate Ureaplasma in preterm infants. Arch Dis Child Fetal Neonatal Ed. 2020;105(6):615–622.
  • Razak A, Alshehri N. Azithromycin for preventing bronchopulmonary dysplasia in preterm infants: a systematic review and meta-analysis. Pediatr Pulmonol. 2021;56(5):957–966.
  • Viscardi RM, Terrin ML, Magder LS, et al. Randomized trial of azithromycin to eradicate Ureaplasma respiratory colonization in preterm infants: 2-year outcomes. Pediatr Res. 2022;91(1):178–187.
  • Ballard HO, Shook LA, Bernard P, et al. Use of azithromycin for the prevention of bronchopulmonary dysplasia in preterm infants: a randomized, double-blind, placebo controlled trial. Pediatr Pulmonol. 2011;46(2):111–118.
  • Kotecha SJ, Course CW, Jones KE, et al. Follow-up study of infants recruited to the randomised, placebo-controlled trial of azithromycin for the prevention of chronic lung disease of prematurity in preterm infants-study protocol for the AZTEC-FU study. Trials. 2022;23:796.
  • Ozdemir R, Erdeve O, Dizdar EA, et al. Clarithromycin in preventing bronchopulmonary dysplasia in Ureaplasma urealyticum-positive preterm infants. Pediatrics. 2011;128:e1496–e1501.
  • Suresh GK, Davis JM, Soll RF. Superoxide dismutase for preventing chronic lung disease in mechanically ventilated preterm infants. Cochrane Database Syst Rev. 2001;1:CD001968.
  • Brion LP, Bell EF, Raghuveer TS. Vitamin E supplementation for prevention of morbidity and mortality in preterm infants. Cochrane Database Syst Rev. 2003;4:CD003665.
  • Doyle LW, Cheong JL, Hay S, et al. Early (< 7 days) systemic postnatal corticosteroids for prevention of bronchopulmonary dysplasia in preterm infants. Cochrane Database Syst Rev. 2021;10:CD001146.
  • Doyle LW, Cheong JL, Hay S, et al. Late (>/= 7 days) systemic postnatal corticosteroids for prevention of bronchopulmonary dysplasia in preterm infants. Cochrane Database Syst Rev. 2021;11:CD001145.
  • Dassios T, Kaltsogianni O, Hickey A, et al. Chronology and determinants of respiratory function changes following administration of systemic postnatal corticosteroids in extremely preterm infants. J Pediatr. 2019;215:17–23.
  • Marr BL, Mettelman BB, Bode MM, et al. Randomized trial of a 42 day compared with a 9 day courses of dexamethasone for the treatment of evolving bronchopulmonary dysplasia in extremely preterm infants. J Pediatr. 2019;211:20–26.
  • Dassios T, Kaltsogianni O, Greenough A. Second course of systemic dexamethasone: efficacy and respiratory function changes. J Matern Fetal Neonatal Med. 2022;35:1401–1404.
  • Williams EE, Dassios T, Mann M, et al. The effect of postnatal corticosteroids on growth parameters in infants with bronchopulmonary dysplasia. J Perinat Med. 2021;49:1141–1144.
  • Harris C, Crichton S, Zivanovic S, et al. Effect of dexamethasone exposure on the neonatal unit on the school age lung function of children born very prematurely. PloS One. 2018;13:e0200243.
  • Puia-Dumitrescu M, Wood TR, Comstock BA, et al. Dexamethasone, prednisolone and methylprednisolone use and 2 year neurodevelopmental outcomes in extremely preterm infants. JAMA Netw Ope. 2022;5:e221947.
  • Gruver-Yates AL, Cidlowski JA. Tissue-specific actions of glucocorticoids on apoptosis: a double-edged sword. Cells. 2013;2:202–223.
  • Greenough A. Hydrocortisone to prevent bronchopulmonary dysplasia — not a silver bullet. New Engl J Med. 2022;386:1181–1183.
  • Baud O, Maury L, Lebail F, et al. Effect of early low-dose hydrocortisone on survival without bronchopulmonary dysplasia in extremely preterm infants (PREMILOC): a double-blind, placebo-controlled, multicentre, randomised trial. Lancet. 2016;387:1827–1836.
  • Watterberg KL, Walsh MC, Li L, et al. Hydrocortisone to improve survival without bronchopulmonary dysplasia. New Engl J Med. 2022;386:1121–1131.
  • Shaffer ML, Baud O, Lacaze-Masmonteil T, et al. Effect of prophylaxis for early adrenal insufficiency using low-dose hydrocortisone in very preterm infants: an individual patient data meta-analysis. J Pediatr. 2019;207:136–142.e5.
  • Bassler D, Plavka R, Shinwell ES, et al. Early inhaled budesonide for the prevention of bronchopulmonary dysplasia. New Engl J Med. 2015;373:1497–1506.
  • Bassler D, Shinwell ES, Hallman M, et al. Long-term effects of inhaled budesonide for bronchopulmonary dysplasia. New Engl J Med. 2018;378:148–157.
  • Wiedmann TS, Bhatia R, Wattenberg LW. Drug solubilization in lung surfactant. J Control Release. 2000;65:43–47.
  • Yeh TF, Chen CM, Wu SY, et al. Intratracheal administration of budesonide/surfactant to prevent bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2016;193:86–95.
  • Anderson CD, Brett Kothe T, Josephsen JB, et al. Budesonide mixed with surfactant did not affect neurodevelopmental outcomes at 6 or 18 months corrected age in observational cohorts. J Perinatol. 2021;41:1681–1689.
  • Mobius MA, Thebaud B. Bronchopulmonary dysplasia: where have all the stem cells gone? Chest. 2017;152:1043–1052.
  • Collins A. Stem cell therapy for bronchopulmonary dysplasia. Curr Opin Pediatr. 2020;32:210–215.
  • Ahn SY, Chang YS, Kim JH, et al. Two-year follow-up outcomes of premature infants enrolled in the phase i trial of mesenchymal stem cells transplantation for bronchopulmonary dysplasia. J Pediatr. 2017;185:49–54 e2.
  • Ahn SY, Chang YS, Lee MH, et al. Stem cells for bronchopulmonary dysplasia in preterm infants: a randomized controlled phase II trial. Stem Cells Transl Med. 2021;10:1129–1137.
  • Stewart A, Brion LP, Ambrosio‐Perez I. Diuretics acting on the distal renal tubule for preterm infants with (or developing) chronic lung disease. Cochrane Database Syst Rev. 2011;9:CD001817.
  • Stewart A, Brion LP. Intravenous or enteral loop diuretics for preterm infants with (or developing) chronic lung disease. Cochrane Database Syst Rev. 2011;9:CD001453.
  • Tan C, Sehgal K, Sehgal K, et al. Diuretic use in infants with developing or established chronic lung disease: a practice looking for evidence. J Paediatr Child Health. 2020;56:1189–1193.
  • Brion LP, Primhak RA, Yong W. Aerosolized diuretics for preterm infants with (or developing) chronic lung disease. Cochrane Database Syst Rev. 2006;3:CD001694.
  • Slaughter JL, Stenger MR, Reagan PB, et al. Inhaled bronchodilator use for infants with bronchopulmonary dysplasia. J Perinatol. 2015;35:61–66.
  • Peters-Golden M, Henderson WR Jr. Leukotrienes. N Engl J Med. 2007;357:1841–1854.
  • Rupprecht T, Rupprecht C, Harms D, et al. Leukotriene receptor blockade as a life-saving treatment in severe bronchopulmonary dysplasia. Respiration. 2014;88:285–290.
  • Kim SB, Lee JH, Lee J, et al. The efficacy and safety of Montelukast sodium in the prevention of bronchopulmonary dysplasia. Korean J Pediatr. 2015;58:347–353.
  • Sehgal A, Krishnamurthy MB, Clark M, et al. ACE inhibition for severe bronchopulmonary dysplasia - an approach based on physiology. Physiol Rep. 2018;6:e13821.
  • Sehgal A, Malikiwi A, Paul E, et al. Systemic arterial stiffness in infants with bronchopulmonary dysplasia: potential cause of systemic hypertension. J Perinatol. 2016;36:564–569.
  • Seghal A, Malikiwi A, Paul E, et al. A new look at bronchopulmonary dysplasia: postcapillary pathophysiology and cardiac dysfunction. Pulm Circ. 2016;6:508–515.
  • Arattu Thodika FM, Nanjundappa M, Dassios T, et al. Pulmonary hypertension in infants with bronchopulmonary dysplasia: risk factors, mortality and duration of hospitalisation. J Perinat Med. 2021;50:327–333.
  • Hansmann G, Koestenberger M, Alastalo TP, et al. updated consensus statement on the diagnosis and treatment of pediatric pulmonary hypertension: the European pediatric pulmonary vascular disease network (EPPVDN), endorsed by AEPC, ESPR and ISHLT. J Heart Lung Transplant. 2019;38:879–901.
  • Hansmann G, Sallmon H, Roehr CC, et al. Pulmonary hypertension in bronchopulmonary dysplasia. Pediatr Res. 2021;89:446–455.
  • Kadmon G, Schiller O, Dagan T, et al. Pulmonary hypertension specific treatment in infants with bronchopulmonary dysplasia. Pediatr Pulmonol. 2017;52:77–83.
  • Nyp M, Sandritter T, Poppinga N, et al. Sildenafil citrate, bronchopulmonary dysplasia and disordered pulmonary gas exchange: any benefits? J Perinatol. 2012;32:64–69.
  • Admon G, Schiller O, Dagan T, et al. Pulmonary hypertension specific treatment in infants with bronchopulmonary dysplasia. Pediatr Pulmonol. 2017;52:77–83.
  • Kurzner SI, Garg M, Bautista DB, et al. Growth failure in infants with bronchopulmonary dysplasia: nutrition and elevated resting metabolic expenditure. Pediatrics. 1988;81:379–384.
  • Biniwale MA, Ehrenkranz RA. The role of nutrition in the prevention and management of bronchopulmonary dysplasia. Semin Perinatol. 2006;30:200–208.
  • Morgan C, McGowan P, Herwitker S, et al. Postnatal head growth in preterm infants: a randomized controlled parenteral nutrition study. Pediatrics. 2014;133:e120–e128.
  • Silva DT, Hagan R, Sly PD. Home oxygen management of neonatal chronic lung disease in Western Australia. J Paediatr Child Health. 1995;31:185–188.
  • Doyle LW, Carse E, Adams AM, et al. Ventilation in extremely preterm infants and respiratory function at 8 years. New Engl J Med. 2017;377:329–337.
  • Bertrand P, Alvarez C, Fabres J, et al. Home oxygen therapy in children with chronic respiratory failure]. Revista Medica de Chile. 1998;126:284–292.
  • Norzila MZ, Azizi BH, Norrashidah AW, et al. Home oxygen therapy for children with chronic lung diseases. Med J Malaysia. 2001;56:151–157.
  • Batey N, Batra D, Dorling J, et al. Impact of a protocol-driven unified service for neonates with bronchopulmonary dysplasia. ERJ Open Res. 2019;5:00183–02018.
  • DiBlasi RM, Crotwell DN, Poli J, et al. A pilot study to assess short-term physiologic outcomes of transitioning infants with severe bronchopulmonary dysplasia from ICU to two subacute ventilators. Can J Respir Ther. 2018;54:6–11.
  • Na’amnih W, Kassem E, Tannous S, et al. Incidence and risk factors of hospitalisations for respiratory syncytial virus among children aged less than 2 years. Epidemiol Infect. 2022;150:e45.
  • Andabaka T, Nickerson JW, Rojas‐Reyes MX, et al. Monoclonal antibody for reducing the risk of respiratory syncytial virus infection in children. Cochrane Database Syst Rev. 2013;4:CD006602.
  • Wang D, Bayliss S, Meads C. Palivizumab for immunoprophylaxis of respiratory syncytial virus (RSV) bronchiolitis in high-risk infants and young children: a systematic review and additional economic modelling of subgroup analyses. Health Technol Assess. 2011;15:1.
  • MacBean V, Lunt A, Drysdale SB, et al. Predicting healthcare outcomes in prematurely born infants using cluster analysis. Pediatr Pulmonol. 2018;53:1067–1072.