Scolaris Content Display Scolaris Content Display

Spät (nach ≥ 7 Tagen) verabreichte systemische postnatale Kortikoide zur Vorbeugung der bronchopulmonalen Dysplasie bei Frühgeborenen

Collapse all Expand all

Background

Many infants born preterm develop bronchopulmonary dysplasia (BPD), with lung inflammation playing a role. Corticosteroids have powerful anti‐inflammatory effects and have been used to treat individuals with established BPD. However, it is unclear whether any beneficial effects outweigh the adverse effects of these drugs.

Objectives

To examine the relative benefits and adverse effects of late (starting at seven or more days after birth) systemic postnatal corticosteroid treatment for preterm infants with evolving or established BPD.

Search methods

We ran an updated search on 25 September 2020 of the following databases: CENTRAL via CRS Web and MEDLINE via OVID. We also searched clinical trials databases and reference lists of retrieved articles for randomised controlled trials (RCTs). We did not include quasi‐RCTs.

Selection criteria

We selected for inclusion in this review RCTs comparing systemic (intravenous or oral) postnatal corticosteroid treatment versus placebo or no treatment started at seven or more days after birth for preterm infants with evolving or established BPD. We did not include trials of inhaled corticosteroids.

Data collection and analysis

We used standard Cochrane methods. We extracted and analysed data regarding clinical outcomes that included mortality, BPD, and cerebral palsy. We used the GRADE approach to assess the certainty of evidence.

Main results

Use of the GRADE approach revealed that the certainty of evidence was high for most of the major outcomes considered, except for BPD at 36 weeks for all studies combined and for the dexamethasone subgroup, which were downgraded one level to moderate because of evidence of publication bias, and for the combined outcome of mortality or BPD at 36 weeks for all studies combined and for the dexamethasone subgroup, which were downgraded one level to moderate because of evidence of substantial heterogeneity.

We included 23 RCTs (1817 infants); 21 RCTS (1382 infants) involved dexamethasone (one also included hydrocortisone) and two RCTs (435 infants) involved hydrocortisone only. The overall risk of bias of included studies was low; all were RCTs and most trials used rigorous methods.

Late systemic corticosteroids overall reduce mortality to the latest reported age (risk ratio (RR) 0.81, 95% confidence interval (CI) 0.66 to 0.99; 21 studies, 1428 infants; high‐certainty evidence). Within the subgroups by drug, neither dexamethasone (RR 0.85, 95% CI 0.66 to 1.11; 19 studies, 993 infants; high‐certainty evidence) nor hydrocortisone (RR 0.74, 95% CI 0.54 to 1.02; 2 studies, 435 infants; high‐certainty evidence) alone clearly reduce mortality to the latest reported age. We found little evidence for statistical heterogeneity between the dexamethasone and hydrocortisone subgroups (P = 0.51 for subgroup interaction).

Late systemic corticosteroids overall probably reduce BPD at 36 weeks' postmenstrual age (PMA) (RR 0.89, 95% CI 0.80 to 0.99; 14 studies, 988 infants; moderate‐certainty evidence). Dexamethasone probably reduces BPD at 36 weeks' PMA (RR 0.76, 95% CI 0.66 to 0.87; 12 studies, 553 infants; moderate‐certainty evidence), but hydrocortisone does not (RR 1.10, 95% CI 0.92 to 1.31; 2 studies, 435 infants; high‐certainty evidence) (P < 0.001 for subgroup interaction).

Late systemic corticosteroids overall probably reduce the combined outcome of mortality or BPD at 36 weeks' PMA (RR 0.85, 95% CI 0.79 to 0.92; 14 studies, 988 infants; moderate‐certainty evidence). Dexamethasone probably reduces the combined outcome of mortality or BPD at 36 weeks' PMA (RR 0.75, 95% CI 0.67 to 0.84; 12 studies, 553 infants; moderate‐certainty evidence), but hydrocortisone does not (RR 0.98, 95% CI 0.88 to 1.09; 2 studies, 435 infants; high‐certainty evidence) (P < 0.001 for subgroup interaction).

Late systemic corticosteroids overall have little to no effect on cerebral palsy (RR 1.17, 95% CI 0.84 to 1.61; 17 studies, 1290 infants; high‐certainty evidence). We found little evidence for statistical heterogeneity between the dexamethasone and hydrocortisone subgroups (P = 0.63 for subgroup interaction).

Late systemic corticosteroids overall have little to no effect on the combined outcome of mortality or cerebral palsy (RR 0.90, 95% CI 0.76 to 1.06; 17 studies, 1290 infants; high‐certainty evidence). We found little evidence for statistical heterogeneity between the dexamethasone and hydrocortisone subgroups (P = 0.42 for subgroup interaction).

Studies had few participants who were not intubated at enrolment; hence, it is not possible to make any meaningful comments on the effectiveness of late corticosteroids in preventing BPD in non‐intubated infants, including those who might in the present day be supported by non‐invasive techniques such as nasal continuous positive airway pressure or high‐flow nasal cannula oxygen/air mixture, but who might still be at high risk of later BPD.

Results of two ongoing studies are awaited.

Authors' conclusions

Late systemic postnatal corticosteroid treatment (started at seven days or more after birth) reduces the risks of mortality and BPD, and the combined outcome of mortality or BPD, without evidence of increased cerebral palsy. However, the methodological quality of studies determining long‐term outcomes is limited, and no studies were powered to detect increased rates of important adverse long‐term neurodevelopmental outcomes. This review supports the use of late systemic corticosteroids for infants who cannot be weaned from mechanical ventilation. The role of late systemic corticosteroids for infants who are not intubated is unclear and needs further investigation. Longer‐term follow‐up into late childhood is vital for assessment of important outcomes that cannot be assessed in early childhood, such as effects of late systemic corticosteroid treatment on higher‐order neurological functions, including cognitive function, executive function, academic performance, behaviour, mental health, motor function, and lung function. Further RCTs of late systemic corticosteroids should include longer‐term survival free of neurodevelopmental disability as the primary outcome.

PICOs

Population
Intervention
Comparison
Outcome

The PICO model is widely used and taught in evidence-based health care as a strategy for formulating questions and search strategies and for characterizing clinical studies or meta-analyses. PICO stands for four different potential components of a clinical question: Patient, Population or Problem; Intervention; Comparison; Outcome.

See more on using PICO in the Cochrane Handbook.

Die späte (ab dem Alter von sieben Tagen begonnene) Behandlung von Frühgeborenen mit systemischen Kortikoiden zur Vorbeugung der Entwicklung einer bronchopulmonalen Dysplasie

Fragestellung des Reviews: Die Ermittlung des Nutzens und schädlicher Wirkungen der Behandlung mit entzündungshemmenden, als Kortikoide (oder Kortikosteroide) bezeichneten, Medikamenten ab dem siebten Lebenstag oder später zur Vorbeugung oder Behandlung einer bronchopulmonalen Dysplasie (einer chronischen Lungenerkrankung) bei Frühgeborenen.

Hintergrund: Kortikosteroide können das Auftreten von Lungenentzündungen bei Neugeborenen mit bronchopulmonaler Dysplasie verringern, können jedoch möglicherweise auch schädliche Wirkungen haben. Die Entwicklung einer bronchopulmonalen Dysplasie ist ein großes Problem für Frühgeborene auf Neugeborenen‐Intensivstationen und ist mit einer höheren Sterblichkeit und schwerwiegenderen langfristigen Folgen für die betroffenen Kinder verbunden. Eine anhaltende Entzündung der Lungen ist die wahrscheinlichste Ursache für die Entwicklung der bronchopulmonalen Dysplasie. Kortikosteroid‐haltige Medikamente haben eine starke entzündungshemmende Wirkung und werden daher zur Vorbeugung oder Behandlung der bronchopulmonalen Dysplasie eingesetzt, insbesondere bei Babys, die nicht von der mechanischen (künstlichen) Beatmung entwöhnt werden können.

Studienmerkmale: Wir begutachteten alle verfügbaren klinischen Studien zur Behandlung von Frühgeborenen, in denen den Frühgeborenen ab dem siebten Lebenstag Kortikoide systemisch, d. h. über eine Injektion (Spritze) oder in Form eines Medikaments verabreicht wurden und in denen Daten zur Häufigkeit der Entwicklung einer bronchopulmonalen Dysplasie im späteren Verlauf der Neugeborenenphase berichtet wurden. Wir schlossen 23 Studien mit insgesamt 1817 Neugeborenen in den Review ein. Die Suche ist auf dem Stand vom 25. September 2020.

Hauptergebnisse: Die Ergebnisse des Reviews weisen darauf hin, dass die Verabreichung von systemischen Kortikoiden an Neugeborene ab dem siebten Tag nach der Geburt oder später das Risiko von Todesfällen und der Entwicklung einer bronchopulmonalen Dysplasie verringert, ohne die Häufigkeit des Auftretens einer Zerebralparese (einer durch eine Hirnschädigung bedingten Beeinträchtigung der Bewegungsfähigkeit) in der späteren Kindheit zu erhöhen. Die längerfristigen Wirkungen sind jedoch bislang nicht ausreichend untersucht worden. Es erscheint sinnvoll, die späte Behandlung mit systemischen Kortikoiden bei Neugeborenen, die nicht von der mechanischen Beatmung entwöhnt werden können, begrenzt zu halten und die Dosierung und Dauer einer Behandlung so gering wie möglich zu halten.

Die Ergebnisse von zwei laufenden (noch nicht abgeschlossenen) Studien sind noch nicht verfügbar.

Vertrauenswürdigkeit der Evidenz: Die Vertrauenswürdigkeit der Evidenz, die unsere Schlussfolgerungen für die Hauptergebnisse stützt, ist insgesamt hoch.

Authors' conclusions

Implications for practice

The condition of the ventilator‐dependent infant with evolving BPD from the age of seven days or later may be at least transiently improved by a course of systemic corticosteroids. High‐quality or moderate‐quality evidence shows that such treatment reduces rates of mortality, BPD, and the combined outcome of mortality or BPD, without evidence of increasing rates of cerebral palsy. However, the methodological quality of studies determining long‐term outcomes is limited: in most reports, surviving children have been assessed predominantly before school age, and no study has been sufficiently powered to detect important adverse long‐term neurodevelopmental outcomes. Given evidence of both benefits and harms of treatment, and limitations of the evidence for long‐term outcomes at present, it appears prudent to reserve the use of late systemic corticosteroids for infants who cannot be weaned from mechanical ventilation via an endotracheal tube from the age of seven days of age, and to minimise the dose and duration of any course of treatment.

Evidence is insufficient to guide the use of late systemic corticosteroids to prevent BPD among infants who are not intubated.

Implications for research

Studies are needed to examine the lowest safe dose of corticosteroid. One large ongoing placebo‐controlled trial of systemic hydrocortisone in ventilator‐dependent infants beyond the first week after birth may help to establish the role of hydrocortisone, if any, in intubated infants (NCT01353313). Hydrocortisone at more physiological doses should be compared with dexamethasone at lower doses for ventilator‐dependent infants. Studies of other corticosteroids, such as betamethasone or methylprednisolone, might be worthwhile. Review authors have noted a compelling need for long‐term follow‐up studies among all children who have been enrolled in randomised controlled trials of postnatal corticosteroids. Investigators must examine a broad range of adverse neurodevelopmental outcomes, which include major neurosensory (including cerebral palsy, visual and auditory function) as well as cognitive and behavioural outcomes that may be more evident at older ages. New studies should be designed to assess overall risks and benefits of corticosteroids and should be sufficiently powered to detect important adverse long‐term neurodevelopmental sequelae.

Despite increasing use of non‐invasive ventilation, infants still become oxygen‐dependent and may develop BPD. Outcomes of systemic corticosteroids given to infants on non‐invasive ventilation remain to be investigated.

Summary of findings

Open in table viewer
Summary of findings 1. Systemic corticosteroids (dexamethasone or hydrocortisone) compared with control (placebo or nothing) for chronic lung disease in preterm infants

Systemic corticosteroids (dexamethasone or hydrocortisone) compared with control (placebo or nothing) for chronic lung disease in preterm infants

Patient or population: preterm infants with chronic lung disease
Setting: multiple neonatal intensive care units from high‐income countries
Intervention: systemic corticosteroids (dexamethasone or hydrocortisone)
Comparison: control (placebo or nothing)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with control (placebo or nothing)

Risk with systemic corticosteroids (dexamethasone or hydrocortisone)

Mortality at latest reported age

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.81
(0.66 to 0.99)

1428
(21 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

228 per 1000

185 per 1000
(151 to 226)

Study population (subgroup of studies treating with dexamethasone)

RR 0.85
(0.66 to 1.11)

993
(19 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

193 per 1000

164 per 1000
(128 to 215)

Study population (subgroup of studies treating with hydrocortisone)

RR 0.74
(0.54 to 1.02)

435
(2 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

305 per 1000

226 per 1000
(165 to 311)

BPD at 36 weeks' PMA

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.89
(0.80 to 0.99)

988
(14 RCTs)

⊕⊕⊕⊝
MODERATEa

Strong evidence for subgroup differences (interaction P < 0.001)

594 per 1000

529 per 1000
(475 to 588)

Study population (subgroup of studies treating with dexamethasone)

RR 0.76
(0.66 to 0.87)

553
(12 RCTs)

⊕⊕⊕⊝
MODERATEa

659 per 1000

501 per 1000
(435 to 573)

Study population (subgroup of studies treating with hydrocortisone)

RR 1.10
(0.92 to 1.31)

435
(2 RCTs)

⊕⊕⊕⊕
HIGH

516 per 1000

567 per 1000
(474 to 676)

Mortality or BPD at 36 weeks' PMA

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.85
(0.79 to 0.92)

988
(14 RCTs)

⊕⊕⊕⊝
MODERATEb

Strong evidence for subgroup differences (interaction P < 0.001)

771 per 1000

656 per 1000
(609  to 710)

Study population (subgroup of studies treating with dexamethasone)

RR 0.75
(0.67 to 0.84)

553
(12 RCTs)

⊕⊕⊕⊝
MODERATEb

787 per 1000

590 per 1000
(527to 661)

Study population (subgroup of studies treating with hydrocortisone)

RR 0.98
(0.88 to 1.09)

435
(2 RCTs)

⊕⊕⊕⊕
HIGH

753 per 1000

738 per 1000
(663 to 832)

Cerebral palsy ‐ at latest reported age

Study population (studies treating with dexamethasone or hydrocortisone)

RR 1.17
(0.84 to 1.61)

1290
(17 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome.

93 per 1000

109 per 1000
(78 to 150)

Study population (subgroup of studies treating with dexamethasone)

RR 1.17
(0.84 to 1.61

1290
(17 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

121 per 1000

135 per 1000 (95 to 193)

Study population (subgroup of studies treating with hydrocortisone)

RR 1.40

(0.60 to 3.26)

435 (2 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

40 per 1000

57 per 1000

Mortality or cerebral palsy ‐ at latest reported age

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.90
(0.76 to 1.06)

1290
(17 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome.

324 per 1000

291 per 1000
(246 to 343)

Study population (subgroup of studies treating with dexamethasone)

RR 0.95 (0.77 to 1.16)

855 (15 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

312 per 1000

296 per 1000 (240 to 362)

Study population (subgroup of studies treating with hydrocortisone)

RR 0.82 (0.62 to 1.08)

435 (2 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

345 per 1000

283 per 1000

*The risk in the intervention group (and its 95% confidence interval) is based on assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

BPD: bronchopulmonary dysplasia; CI: confidence interval; PMA: postmenstrual age; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.

High certainty: further research is very unlikely to change our confidence in the estimate of effect.

Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.

Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.

Very low certainty: we are very uncertain about the estimate.

aDowngraded one level because publication bias was suspected.

bDowngraded one level for moderate heterogeneity.

Background

Description of the condition

Surfactant therapy has improved outcomes for preterm infants with respiratory distress syndrome but has only modestly reduced the risk of bronchopulmonary dysplasia (BPD) (Egberts 1997). Recent data suggest approximately 50% of infants born at < 28 weeks' gestation who survive to 36 weeks' gestation have BPD, with rates remaining stubbornly high, even since exogenous surfactant and more non‐invasive ventilation have been introduced into clinical care over the past 30 years (Cheong 2020). Management BPD in infants is both time‐consuming and costly. The term 'bronchopulmonary dysplasia' describes injury with maldevelopment of the lung that follows preterm birth and is a major problem in neonatal intensive care units. Persistent lung inflammation is the most likely underlying pathogenesis.

Description of the intervention

Postnatal corticosteroid treatment has been shown to have some acute effects on lung function in infants with established BPD, especially among those who are ventilator‐dependent (CDTG 1991; Mammel 1983). Corticosteroids may be given parenterally or enterally. Investigators have expressed concern that the benefits of corticosteroids might not outweigh their adverse effects, which include hypertension, hyperglycaemia, intestinal perforation, and extreme catabolism (Anonymous 1991; Ng 1993). Animal studies have also raised concerns about adverse effects on the central nervous system of corticosteroids given perinatally to immature offspring (Flagel 2002; Gramsbergen 1998).

How the intervention might work

Corticosteroids might prevent or treat BPD through their potent anti‐inflammatory effects.

Why it is important to do this review

Multiple published systematic reviews have examined the use of systemic postnatal corticosteroids in infants with or at risk of BPD (Arias‐Camison 1999; Bhuta 1998; Doyle 2000; Doyle 2010a; Doyle 2010b; Doyle 2010c; Doyle 2014a; Doyle 2014b; Doyle 2017b; Halliday 1997; Halliday 1999; Tarnow‐Mordi 1999). Other systematic reviews have addressed early versus late use of inhaled corticosteroids for preventing BPD (Shah 2017a), or for treating BPD (Onland 2017a), as well as use of systemic versus inhaled corticosteroids for preventing BPD (Shah 2017b), or for treating BPD (Shah 2017c). Another review compared different systemic corticosteroid regimens (Onland 2017b).

Two existing Cochrane Reviews have explored trials in which systemic postnatal corticosteroids were started within seven days of birth (Doyle 2017a), or were started more than seven days after birth (Doyle 2017b). The present systematic review updates the review of systemic corticosteroids started seven days or more after birth.

Objectives

To examine the relative benefits and adverse effects of late (starting at seven or more days after birth) systemic postnatal corticosteroid treatment for preterm infants with evolving or established BPD.

Methods

Criteria for considering studies for this review

Types of studies

We included randomised controlled trials (RCTs) of late (seven days or more) systemic postnatal corticosteroid treatment for preterm infants with evolving or established BPD that reported clinically important outcome variables. We did not include cluster randomised, cross‐over, or quasi‐randomised controlled trials.

Types of participants

We included preterm infants with evolving or established BPD, defined as oxygen‐dependent, ventilator‐dependent, or both, with or without radiographic changes of BPD.

Types of interventions

Treatment with systemic corticosteroids (dexamethasone or hydrocortisone) versus control (placebo or nothing).

Types of outcome measures

These are divided into primary and secondary outcomes.

Primary outcomes

  • Mortality at various ages (including at 28 days after birth, at 36 weeks' postmenstrual age, at discharge home after primary hospitalisation, and at latest age reported)

  • BPD (including at 28 days after birth, at 36 weeks' postmenstrual age, and at 36 weeks' postmenstrual age among survivors)

  • Mortality or BPD (at 28 days after birth and at 36 weeks' postmenstrual age)

  • Longer‐term outcomes into childhood (including blindness, deafness, cerebral palsy, and major neurosensory disability)

Secondary outcomes

  • Failure to extubate

  • Late rescue with corticosteroids

  • Need for home oxygen therapy

  • Complications during primary hospitalisation (including infection, hyperglycaemia, hypertension, pulmonary air leak, patent ductus arteriosus, severe intraventricular haemorrhage, periventricular leukomalacia, necrotising enterocolitis, sepsis, gastrointestinal bleeding, intestinal perforation, and severe retinopathy of prematurity)

  • Later childhood outcomes, including respiratory function, blood pressure, and growth

Search methods for identification of studies

Electronic searches

We conducted a comprehensive updated search in September 2020 including Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 9), in the Cochrane Library; and OVID MEDLINE(R) and Epub Ahead of Print, In‐Process & Other Non‐Indexed Citations, Daily and Versions(R) (1 January 2016 to 25 September 2020). We have included the search strategies for each database in Appendix 1. We did not apply language restrictions.

We searched clinical trial registries for ongoing and recently completed trials. We searched the World Health Organization’s International Clinical Trials Registry Platform (ICTRP) (www.who.int/ictrp/search/en/) and the US National Library of Medicine’s ClinicalTrials.gov (clinicaltrials.gov), via Cochrane CENTRAL. Additionally, we searched the International Standard Randomized Controlled Trials Number (ISRCTN) Registry (http://www.isrctn.com/) for any unique trials not found through the Cochrane CENTRAL search.

This is the fifth update of this review. Our previous search details are listed in Appendix 2 and Appendix 3.

Searching other resources

We also searched the reference lists of all identified publications for additional references not identified by the electronic literature search.

Data collection and analysis

We used the methods of Cochrane Neonatal for data collection and analysis.

Selection of studies

We included all RCTS that fulfilled the selection criteria presented in the previous section. Two review authors (LWD and JC) independently reviewed results of the updated search and selected studies for inclusion. We resolved disagreements by discussion.

Data extraction and management

For each included trial, we sought information regarding methods of randomisation, blinding, and stratification, and whether the trial was single‐ or multi‐centred. Information on trial participants included birth weight, gestational age, and sex. We analysed information on the following clinical outcomes: mortality, BPD (including BPD at 28 days after birth, BPD at 36 weeks' postmenstrual age, BPD at 36 weeks' postmenstrual age in survivors, late rescue with corticosteroids (among all infants and survivors), and need for home oxygen therapy), mortality or BPD (at 28 days after birth and at 36 weeks' postmenstrual age), and long‐term outcomes (including blindness, deafness, cerebral palsy, and major neurosensory disability). Secondary outcomes included failure to extubate, complications during primary hospitalisation (including infection, hyperglycaemia, glycosuria, hypertension, echodensities on ultrasound scan of brain, necrotising enterocolitis, gastrointestinal bleeding, gastrointestinal perforation, and severe retinopathy of prematurity), and longer‐term outcomes of cognitive delay, respiratory health and function, blood pressure, and growth during childhood.

For each study, one review author (LWD) entered final data into Review Manager (RevMan) 5 (Review Manager 2020); a second review author (SH) then checked the data for accuracy. We resolved discrepancies through discussion or through consultation with a third assessor (HLH).

We attempted to contact the authors of original reports to request further details when information regarding any of the above was unclear.

Assessment of risk of bias in included studies

Two review authors (LWD and JC) independently assessed risk of bias (low, high, or unclear) of all included trials using the Cochrane ‘Risk of bias’ tool for the following domains (Higgins 2011).

  • Sequence generation (selection bias).

  • Allocation concealment (selection bias).

  • Blinding of participants and personnel (performance bias).

  • Blinding of outcome assessment (detection bias).

  • Incomplete outcome data (attrition bias).

  • Selective reporting (reporting bias).

  • Any other bias.

We resolved disagreements by discussion or by consultation with a third assessor. See Appendix 4 for a more detailed description of risk of bias for each domain. 

Measures of treatment effect

We used the standard methods of Cochrane Neonatal to analyse data.

We performed statistical analyses using Review Manager (RevMan) 5 (Review Manager 2020). We analysed dichotomous data using risk ratio (RR), risk difference (RD), and the number needed to treat for an additional beneficial outcome (NNTB), or the number needed to treat for an additional harmful outcome (NNTH). We reported the 95% confidence interval (CI) for all estimates.

We analysed continuous data using mean difference (MD), or standardised mean difference (SMD) to combine trials that measured the same outcome using different methods.

Unit of analysis issues

For clinical outcomes such as episodes of sepsis, we analysed the data as proportions of neonates having one or more episodes.

Dealing with missing data

For included studies, we noted levels of attrition. When we had concern regarding the impact of including studies with high levels of missing data in the overall assessment of treatment effect, we planned to explore this concern using sensitivity analysis.

We performed all outcome analyses on an intention‐to‐treat basis (i.e. we included in the analyses all participants randomised to each group). The denominator for each outcome in each trial was the number randomised minus any participants whose outcomes were known to be missing.

Assessment of heterogeneity

We examined heterogeneity between trials by inspecting forest plots and quantifying the impact of heterogeneity using the I² statistic. If noted, we planned to explore possible causes of statistical heterogeneity using prespecified subgroup analysis (e.g. differences in study quality, participants, intervention regimens, outcome assessments).

Assessment of reporting biases

We assessed possible publication bias and other biases by examining symmetry/asymmetry on funnel plots. In addition, we computed Egger's statistic on funnel plots to assess the strength of the evidence for publication bias.

For included trials that were recently performed (and therefore prospectively registered), we used the websites www.clinicaltrials.gov and www.controlled-trials.com to explore possible selective reporting of study outcomes by comparing primary and secondary outcomes for reports in which primary and secondary outcomes were proposed at trial registration. If we found such discrepancies, we planned to contact the primary investigators to request missing data on outcomes prespecified at trial registration.

Data synthesis

When we judged meta‐analysis to be appropriate, we carried out the analysis using Review Manager (RevMan) 5, supplied by Cochrane (Review Manager 2020). We used the Mantel‐Haenszel method for estimates of typical RR and RD. We analysed continuous measures using the inverse variance method, and we computed MDs or SMDs. 

We used the fixed‐effect model for all meta‐analyses.

Subgroup analysis and investigation of heterogeneity

We included subgroup analyses by type of corticosteroid used (dexamethasone or hydrocortisone) if we identified a sufficient number of trials to make such subgroup analyses meaningful.

Sensitivity analysis

We planned to perform sensitivity analyses for situations where this might affect interpretation of significant results (e.g. when risk of bias was associated with the quality of some of included trials).

Summary of findings and assessment of the certainty of the evidence

We used the GRADE approach, as outlined in the GRADE Handbook (Schünemann 2013), to assess the certainty of evidence for the following (clinically relevant) outcomes: mortality, BPD (including BPD at 28 days after birth, BPD at 36 weeks' postmenstrual age, BPD at 36 weeks' postmenstrual age in survivors, late rescue with corticosteroids (among all infants and survivors), and need for home oxygen therapy), mortality or BPD (at 28 days after birth and at 36 weeks' postmenstrual age), and long‐term outcomes (including blindness, deafness, cerebral palsy, and major neurosensory disability).

Two review authors (LWD and JC) independently assessed the certainty of evidence for each of the outcomes above. We considered evidence from RCTs as high certainty but downgraded the evidence one level for serious (or two levels for very serious) limitations based upon the following: design (risk of bias), consistency across studies, directness of evidence, precision of estimates, and presence of publication bias. We used the GRADEpro GDT Guideline Development Tool to create summary of findings Table 1 to report the certainty of evidence.

The GRADE approach results in an assessment of the certainty of a body of evidence as one of four grades.

  • High certainty: further research is very unlikely to change our confidence in the estimate of effect.

  • Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.

  • Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.

  • Very low certainty: we are very uncertain about the estimate.

Results

Description of studies

Up to the 2014 update of this review, 21 studies had been included.

For the 2017 update of this review, database searches for the calendar years 2013‐2017 identified 4930 references; other search sources identified 102 additional references. After removing 852 duplicate records, 4180 title abstracts were screened, 4166 full texts were excluded, 14 full text articles were assessed for eligibility, and no new studies were included at that time.

Results of the search

For the current update of this review, database searches for the calendar years 2016‐2020 identified 3760 references; other search sources identified no additional references. After removing 404 duplicate records, 3356 title abstracts were screened, 3351 full texts were excluded, five full text articles were assessed for eligibility, and two new studies were included. See Figure 1.


Study flow diagram: review update.

Study flow diagram: review update.

 With addition of the two new RCTs, we have included in this updated review 23 RCTs, in total, that have recruited 1817 infants. These trials enrolled preterm infants who were oxygen‐ or ventilator‐dependent (or both) beyond six days of age. Investigators typically used dexamethasone at an initial dose of 0.5 to 1.0 mg/kg/d, with initial duration of therapy ranging from three days to six weeks. In two studies, the corticosteroid was hydrocortisone (Onland 2019Parikh 2013). Details are given below and in the Characteristics of included studies table.

We discuss the excluded trials below and in the Characteristics of excluded studies table.

We identified two ongoing RCTs of hydrocortisone to prevent or treat BPD (He 2020NCT01353313). See Characteristics of ongoing studies.

Included studies

We included 23 RCTs (1817 infants) in this updated review.

Ariagno 1987 was updated with more data provided by investigators in September 2000. Investigators randomised 34 preterm infants of less than 1501 grams birth weight who were ventilator‐dependent and were not weaning from mechanical ventilation at three weeks of age to parenteral dexamethasone (n = 17) or placebo (n = 17) groups. Treated babies received one of two regimens: a 10‐day course of 1.0 mg/kg/d for four days and 0.5 mg/kg/d for six days ((total dose 7 mg/kg dexamethasone over 10 days), or a seven‐day course of 1.0 mg/kg/d for three days followed by 0.5 mg/kg/d for four days (total dose 8.5 mg/kg dexamethasone over 10 days). Researchers calculated total respiratory system compliance from a pneumotachometer and made airway pressure measurements during mechanical inflation before and after seven days of treatment. Outcomes included mortality, duration of ventilation and oxygen therapy, and complications of prematurity and treatment. Country: USA.

Avery 1985 enrolled 16 infants with birth weight less than 1500 grams, a clinical and radiographic diagnosis of respiratory distress syndrome, inability to be weaned from the ventilator after two weeks, and radiological evidence of stage II or III BPD (Northway 1967). Researchers excluded babies if they had patent ductus arteriosus, congenital heart disease, sepsis, or pneumonia; had received intravenous lipids for at least 24 hours; and were over six weeks of age. To those randomised to receive dexamethasone (n = 8), investigators gave 0.5 mg/kg/d intravenously in two divided doses for three days, followed by 0.3 mg/kg/d for a further three days, thereafter decreased by 10% of the current dose every three days until a dose of 0.1 mg/kg/d was reached. At that point, they gave the drug on alternate days for one week, then discontinued (total dose approximately 8 mg/kg dexamethasone over 42 days). Control infants (n = 8) did not receive a placebo. Country: USA.

Brozanski 1995 was a prospective randomised double‐blind trial conducted to assess the efficacy and safety of pulse doses of dexamethasone for survival without supplemental oxygen given to very low birth weight infants at high risk of BPD. Trial authors randomly assigned 78 infants with birth weight less than 1501 grams, who were ventilator‐dependent at seven days, to receive pulse doses of dexamethasone 0.5 mg/kg/d 12‐hourly or an equivalent volume of a saline placebo for three days at 10‐day intervals, until they no longer required supplemental oxygen or mechanical ventilation, or had reached 36 weeks' postmenstrual age (total minimum dose 3 mg/kg dexamethasone over three days). Infants were excluded from the study if they had complex congenital anomalies, pulmonary hypoplasia, or haemodynamic instability. Country: USA. Participants were recruited between March 1991 and April 1993. Supported by grants from the Magee‐Women's Health Foundation Research Fund and the GCRC/National Institutes of Health 5M0 IRR00084.

CDTG 1991 (Collaborative Dexamethasone Trial Group 1991) was a multi‐centre trial conducted at 31 centres in six countries (UK, Ireland, Belgium, Germany, Canada, and USA) over a period of two and a half years from August 1986 to January 1989. A total of 287 infants who were oxygen‐dependent and had been in a static or deteriorating condition over the preceding week were eligible for trial entry from around three weeks of age. Study authors excluded infants with major malformations (n = 2), and they delayed trial entry to allow treatment of any intercurrent infection or heart failure. Infants did not have to require mechanical ventilation ‐ at the time of entry approximately two‐thirds of infants were intubated and one‐third were not. Those allocated to the dexamethasone group (n = 143) received 0.6 mg/kg/d intravenously (or orally if there was no intravenous line) for one week (total dose 4.2 mg/kg dexamethasone over seven days). There was an option to give a second tapering nine‐day course (0.6, 0.4, and 0.2 mg/kg/d for three days each) if, after initial improvement, relapse occurred. Control infants (n = 142) received an equivalent volume of saline placebo. Supported by Action Research for Crippled Children.

Cummings 1989 randomised 36 preterm infants with birth weight less than 1251 grams and gestational age less than 31 weeks, who were dependent on oxygen (> 29%) and mechanical ventilation (rate > 14 per minute with no evidence of weaning during the previous 72 hours) at two weeks of age, to receive a 42‐day course of dexamethasone or an 18‐day course of dexamethasone or saline placebo. They did not include infants with symptomatic patent ductus arteriosus, renal failure, or sepsis. To infants in the 42‐day group (n = 13), researchers administered dexamethasone at a dose of 0.5 mg/kg/d for three days and 0.3 mg/kg/d for the next three days. They then reduced the dose by 10% every three days until a dose of 0.1 mg/kg was reached on Day 34. After three days at this dose, the drug was given on alternate days for one week and then was stopped (total dose 7.9 mg/kg dexamethasone over 42 days). Infants in the 18‐day dexamethasone group (n = 12) received the same initial dose of 0.5 mg/kg/d for three days, but their dose was then decreased more rapidly by 50% every three days until a dose of 0.06 mg/kg was reached on Day 10. After three days at this dose, study authors gave the drug on alternate days for one week and then stopped (total dose 3 mg/kg dexamethasone over 18 days). For the remaining four treatment days, those infants received saline placebo. Infants in the control group (n = 11) received saline placebo for 42 days. Researchers combined the two treatment groups for the purposes of this meta‐analysis and provided additional data on some short‐term and long‐term outcomes for inclusion in this review. Country: USA. Participants were recruited between January 1986 and June 1987.

Doyle 2006 included a total of 70 infants of less than 1000 grams birth weight or born at less than 28 weeks' gestation, who were at least seven days of age and were ventilator‐dependent and considered eligible for postnatal corticosteroids. Exclusions were few and comprised only those with congenital anomalies likely to adversely affect long‐term neurological outcomes. Trialists worked at 11 collaborating centres within Australia, New Zealand, and Canada and performed stratification by centre. They randomly allocated infants to twice‐daily doses of a 10‐day tapering course of dexamethasone sodium phosphate (0.15 mg/kg/d for three days, 0.10 mg/kg/d for three days, 0.05 mg/kg/d for two days, 0.02 mg/kg/d for two days (total dose 0.89 mg/kg dexamethasone over 10 days) (n = 35 infants)) or to an equivalent volume of 0.9% saline placebo (n = 35 infants). A repeat course of the same blinded drug was a therapeutic option for attending physicians. The dexamethasone preparation did not contain bisulphite preservative. Researchers based the sample size calculation for the original trial on detecting improvement in survival free of major neurosensory disability from 50% to 60%, with a two‐sided type I error rate of 5% and 80% power, and required that a total of 814 infants be recruited. This study was stopped early at 70 infants, not only because less than 10% of the initial sample had been recruited after 2.5 years (March 2000 to October 2002), making it unlikely that the total sample size of 814 would be achieved within a reasonable time, but also because the rate of recruitment had fallen ‐ not increased ‐ even though more centres had entered the study from the time of its inception. Countries: Australia, New Zealand, Canada. Supported by the National Health ad Medical Research Council of Australia (Project Grant 108700).

Durand 1995 was a prospective randomised trial of 44 infants of birth weight 501 grams to 1500 grams and gestational age between 24 and 32 weeks who failed to be weaned from the ventilator at 7 to 14 days; one infant was excluded after randomisation because of birth weight < 500 grams, and hence data were reported for 43 infants. Their oxygen requirement was > 29% and ventilator rate > 14 per minute. Investigators excluded infants with documented sepsis, evidence of systemic hypertension, congenital heart disease, renal failure, intraventricular haemorrhage (grade IV), and multiple congenital anomalies. Infants in the treatment group (n = 23) received dexamethasone 0.5 mg/kg/d 12‐hourly intravenously for the first three days, 0.25 mg/kg/d for the next three days, and 0.10 mg/kg/d on the seventh day of treatment (total dose 2.35 mg/kg dexamethasone over seven days). Controls (n = 20) received no placebo and no dexamethasone during the seven‐day study period. At the end of the study week, the attending clinician could start dexamethasone treatment for controls. Country: USA. Participants were recruited between December 1990 and November 1992.

Harkavy 1989 randomised 21 preterm infants who were ventilator‐ and oxygen‐dependent at 30 days of age to receive dexamethasone or placebo. They gave dexamethasone 0.5 mg/kg/d in two or more doses either intravenously or by mouth (total dose 1 mg/kg dexamethasone or more over 2 days), and they gave an equivalent volume of saline to controls. Country: USA. Participants were recruited between April 1983 and July 1987. Supported by a grant from the Columbia Hospital for Women Research Foundation.

Kari 1993 was a randomised double‐blind placebo‐controlled trial that enrolled 41 infants with birth weight less than 1501 grams, gestational age greater than 23 weeks, dependence on mechanical ventilation at 10 days, and no signs of patent ductus arteriosus, sepsis, gastrointestinal bleeding, or major malformations. Infants in the dexamethasone group (n = 17) received 0.5 mg/kg/d intravenously in two doses for seven days (total dose 3.5 mg/kg dexamethasone over seven days), whereas the placebo group (n = 24) received normal saline. Country: Finland. Participants were recruited between January 1989 and February 1991. Supported by The Foundation for Pediatric Research, The Academy of Finland, and The Sigfrid Juselius Foundation.

In Kazzi 1990, 23 preterm infants with birth weight less than 1500 grams and radiological findings consistent with a diagnosis of BPD who were ventilator‐dependent at three to four weeks of age were eligible for study entry provided they needed more than 34% oxygen and had a ventilator rate greater than 14 per minute or peak inspiratory pressure > 17 cmH₂O. Infants had to show lack of improvement in ventilator dependency during the preceding five days. Infants in the treatment group (n = 12) received dexamethasone 0.50 mg/kg/d for three days, given as a single daily dose by nasogastric tube. The dose was tapered to 0.40 mg/kg/d for two days, then to 0.25 mg/kg/d for two days (total dose 2.8 mg/kg dexamethasone over seven days). Thereafter, infants received hydrocortisone administered in four divided doses every six hours, beginning with 8 mg/kg/d for two days and tapered by 50% of the dose every other day until 0.5 mg/kg/d was reached (total dose 31 mg/kg hydrocortisone over 10 days). After a total of 17 days, treatment was discontinued. Infants in the control group (n = 11) received equal volumes of saline. In subgroup analysis by type of corticosteroid, this study is included in the dexamethasone subgroup because the dose of corticosteroid received comprised more dexamethasone than it did hydrocortisone. Country: USA. Participants were recruited between August 1986 and February 1989.

Kothadia 1999 randomly allocated 118 preterm infants (birth weight < 1501 grams) between 15 and 25 days of age who were ventilator‐dependent to receive a 42‐day tapering course of dexamethasone (n = 57) or saline placebo (n = 61). The dosage schedule was 0.25 mg/kg 12‐hourly for three days and 0.15 mg/kg 12‐hourly for three days, followed by a 10% reduction in dose every three days until a dose of 0.1 mg/kg had been received for three days, from which time they received 0.1 mg/kg every other day until 42 days after entry (total dose 7.9 mg/kg dexamethasone over 42 days). Study authors provided additional data on some short‐term outcomes for inclusion in this review. Country: USA. Participants were recruited between April 1992 and May 1995. Supported by the Brenner Children's Hospital, Winston‐Salem, North Carolina.

Kovacs 1998 was a double‐blind RCT conducted to assess the efficacy of a combination of prophylactic systemic dexamethasone and nebulised budesonide in reducing the incidence and severity of BPD in infants at less than 30 weeks' gestation and weighing less than 1501 grams who were ventilator‐dependent at the age of seven days. Thirty infants received dexamethasone 0.25 mg/kg twice daily for three days (total dose 1.5 mg/kg dexamethasone over three days), followed by nebulised budesonide 500 µg twice daily for 18 days. Thirty control infants received systemic and inhaled saline. Study authors provided additional data on some short‐term and long‐term outcomes for inclusion in this review. Country: Canada. Participants were recruited between March 1993 and October 1995.

Noble‐Jamieson 1989 enrolled 18 infants over four weeks of age who required more than 30% oxygen delivered by a headbox (oxyhood). Congenital infection, gastric erosion, and necrotising enterocolitis were absolute contraindications to trial entry; investigators excluded one infant because of necrotising enterocolitis. Entry was postponed if an infant had a central venous catheter, active infection, untreated patent ductus arteriosus, glucose intolerance, or major segmental pulmonary collapse. Trial entry was postponed for 11 infants, mainly because of suspected sepsis. Researchers randomly allocated infants to receive either dexamethasone (n = 9) or saline (n = 9). They gave dexamethasone orally or intravenously at a dose of 0.25 mg/kg twice daily for the first week, 0.125 mg/kg twice daily for the second week, and 0.10 mg/kg daily for the third week (total dose 3.325 mg/kg dexamethasone over 21 days). Twice‐weekly cranial ultrasound scans were performed on all infants, with scans analysed blinded to treatment allocation after completion of the study. Country: England.

Ohlsson 1992 enrolled 25 infants with birth weight less than 1501 grams after receiving parental informed consent, if the following criteria were met: postnatal age 21 to 35 days, inspired oxygen greater than 29%, chest radiograph consistent with BPD, and treatment with diuretics resulting in no signs of improvement in ventilator requirements during the previous 72 hours. Researchers excluded infants if they had a diagnosis of suspected or proven infection, significant congenital malformation, or clinical evidence of patent ductus arteriosus, necrotising enterocolitis, and gastrointestinal haemorrhage or perforation. The treatment group (n = 12) received dexamethasone 0.50 mg/kg 12‐hourly for three days, 0.25 mg/kg 12‐hourly for three days, 0.125 mg/kg 12‐hourly for three days, and 0.125 mg/kg daily for three days (total dose 5.625 mg/kg dexamethasone over 12 days). Investigators gave dexamethasone intravenously at a standard volume of 1 mL. The Research Ethical Committee did not permit use of an intravenous placebo, so a physician not involved in subsequent care of the infant gave a sham injection of 1 mL of normal saline into the bed in the control group (n = 13). Study authors provided additional data for some short‐term outcomes for inclusion in this review. Country: Canada. Participants were recruited between April 1986 and June 1988. Supported by a grant from the Dean's Fund of the University of Toronto.

Onland 2019 was a double‐blind RCT conducted to compare hydrocortisone with placebo in infants < 1250 grams birth weight or < 30 weeks' gestational age who were ventilator‐dependent between 7 and 14 days of age and at high risk of developing BPD. Infants were ineligible if they had chromosomal defects or major congenital malformations or had received corticosteroids for improving lung function in the first week of life. A total of 181 infants received a total dose of 72.5 mg/kg of hydrocortisone over 22 days, and 190 infants received placebo. The primary endpoint was mortality or BPD at 36 weeks' postmenstrual age. Countries: Netherlands and Belgium. Participants were recruited from 15 November 2011 to 23 December 2016. Supported by a project grant from The Netherlands Organization for Health Research and Development (priority medicines for children grant 11‐32010‐02).

Papile 1998 was a double‐blind RCT conducted to compare the benefits and hazards of initiating dexamethasone therapy at two weeks of age versus four weeks of age to 371 ventilator‐dependent very low birth weight (501 grams to 1500 grams) infants who had respiratory index scores (mean airway pressure (MAP) × fraction of inspired oxygen) ≥ 2.4 at 13 to 15 days of age that had been increasing or minimally decreasing during the previous 48 hours, or a score of 4.0, even if there had been improvement during the preceding 48 hours; had received no glucocorticoid treatment after birth; had no evidence or suspicious signs of sepsis as judged by the treating physician; and had no major congenital anomaly of the cardiovascular, pulmonary, or central nervous system. A total of 182 infants received dexamethasone for two weeks followed by placebo for two weeks, and 189 infants received placebo for two weeks followed by either dexamethasone (those with a respiratory index score ≥ 2.4 on treatment Day 14) or additional placebo for two weeks. The dexamethasone dose was 0.5 mg/kg/d intravenously or orally for five days, then 0.3 mg/kg for three days, 0.14 mg/kg for three days, and 0.06 mg/kg for three days (total dose 4.0 mg/kg dexamethasone over 14 days). Only outcome data at 28 days were eligible for inclusion in this review (see below). Country: USA. Participants were recruited from September 1992 to July 1995. Supported by cooperative agreements (U10 HD27881, U10 HD21373, U10 HD27851, U10 HD27853, U10 HD21397, U10 HD19897, U10 HD21415, U10 HD27856, U10 HD21364, U10 HD27880, U10 HD27904, U10 HD27871, and U10 HD21385) with the National Institute of Child Health and Human Development and by General Clinical Research Center grants MO1 RR 00997, MO1 RR 08084, MO1 RR 00750, MO1 RR 00070, and MO1 RR 06022. Dexamethasone was provided by Merck Sharp & Dohme.

Parikh 2013 was a double‐blind RCT of hydrocortisone versus saline placebo given to 64 infants with birth weight ≤ 1000 grams who were ventilator‐dependent between 10 and 21 days of age and at high risk of developing BPD, with the primary outcome of differences in brain tissue volumes on magnetic resonance imaging (MRI) at term‐equivalent age. Infants were excluded if they were at < 23 weeks' gestation at birth, were previously treated with corticosteroids, were receiving indomethacin treatment or were likely to receive it within seven days, had presumed sepsis or necrotising enterocolitis, or had a major congenital anomaly of the cardiopulmonary or central nervous system. Thirty‐one infants received a total of 17 mg/kg of hydrocortisone over seven days, and 33 infants received an identical volume of saline placebo. This trial included follow‐up at 18 to 22 months of age, corrected for prematurity. Country: USA. Participants were recruited between 11 October 2005 and 8 September 2008. Supported by National Institutes of Neurological Disorders and Stroke (K23‐NS048152) and National Center for Research Resources (UL1RR024148 to University of Texas Health Science Center at Houston Center for Clinical and Translational Sciences).

Romagnoli 1997 was a randomised trial of 30 preterm infants who were ventilator‐ and oxygen‐dependent at 10 days and were at 90% risk of developing BPD based on the trial authors' own scoring system. Fifteen infants received dexamethasone 0.5 mg/kg/d for six days, 0.25 mg/kg/d for six days, and 0.125 mg/kg/d for two days (total dose 4.75 mg/kg dexamethasone over 14 days). Control infants (n = 15) did not receive any steroid. Study authors provided additional data on some short‐term outcomes for inclusion in this review. Country: Italy. Participants were recruited between April 1996 and June 1997.

Scott 1997 was a double‐blind RCT of dexamethasone versus saline placebo given to 15 infants who were ventilator‐dependent between 11 and 14 days of age with an inspired oxygen requirement greater than 60%. The primary outcome was cortisol response to adrenocorticotrophic hormone (ACTH). Infants with lethal anomalies were excluded. Ten infants received a total of 1.9 mg/kg of dexamethasone over five days, and five infants received an identical volume of saline placebo. Country: USA. Participants were recruited between 11 October 2005 and 8 September 2008. Supported by a Bristol‐Myers Research Grant and the General Clinical Research Center of the University of New Mexico, Program DRR (NIH 5M01RR00997‐14‐18).

Vento 2004 was a randomised trial of 20 neonates with birth weight < 1251 grams and gestation < 33 weeks who were oxygen‐ and ventilator‐dependent on the 10th day of life. Infants received dexamethasone 0.5 mg/kg/d for three days, 0.25 mg/kg/d for three days, and 0.125 mg/kg/d for one day (total dose 2.375 mg/kg dexamethasone over seven days) (n = 10), or they received no steroid treatment (n = 10). Country: Italy. Participants were recruited between August 1998 and July 2000.

In Vincer 1998, researchers randomly assigned 20 very low birth weight infants who were ventilator‐dependent at 28 days to receive either a six‐day course of intravenous dexamethasone 0.5 mg/kg/d for three days followed by 0.3 mg/kg/d for the final three days (total dose 2.4 mg/kg dexamethasone over six days) (n = 11), or an equal volume of saline placebo (n = 9). This trial included a two‐year follow‐up. Study authors provided additional data on some short‐term outcomes for inclusion in this review. Country: Canada.

Walther 2003 was a double‐blind randomised clinical trial involving 36 preterm infants with birth weight > 599 grams, gestation of 24 to 32 weeks, and respiratory distress syndrome requiring mechanical ventilation with oxygen > 29% or respiratory index > 1.7 between 7 and 14 days of life. Exclusion criteria were sepsis or other documented infection, congenital heart disease, systemic hypertension, unstable clinical status (renal failure, grade IV intraventricular haemorrhage), or multiple congenital anomalies. Eligible infants received dexamethasone 0.2 mg/kg/d for four days, 0.15 mg/kg/d for four days, 0.1 mg/kg for four days, and 0.05 mg/kg for two days (total dose 1.9 mg/kg over 14 days) (n = 19 infants), or saline placebo (n = 17 infants). Country: USA. Participants were recruited between December 1996 and November 1999. Supported by NIH grant P20 RR11145 and GCRC grant M01 RR00425.

Yates 2019 was a multi‐centre randomised blinded parallel‐group placebo‐controlled feasibility study, designed to be the forerunner of a larger RCT. Eligible infants were at < 30 weeks’ gestational age with postnatal age from 10 to 24 days, were ventilator‐dependent and receiving at least 30% inspired oxygen, and were at high risk of developing BPD. Excluded were infants who had received previous courses of postnatal steroids for respiratory disease; and those who had a severe congenital anomaly affecting the lungs, heart, or central nervous system; who had a surgical abdominal procedure or patent ductus arteriosus ligation; or who had an illness or medication for which postnatal corticosteroid would be contraindicated. Eligible infants were randomised to very low‐dose dexamethasone (0.05 mg/kg daily for 10 days, then every second day on Days 12, 14, and 16 after trial entry (total dose 0.65 mg/kg dexamethasone over 16 days) (n = 12 infants)), or saline placebo (n = 10 infants). Country: England. Participants were recruited between 21 July 2017 and 14 April 2018. Supported by the Efficacy and Mechanism Evaluation (EME) programme ‐ a Medical Research Council and National Institute for Health Research (NIHR) partnership.

Excluded studies

In total, we excluded 46 studies. Vento 2004 was listed as excluded and provided data for two separate cohorts of infants ‐ the first cohort randomised at 10 days of age (those data are included in this "Late" review), and the second cohort randomised at four days of age (hence these data are included in the "Early" review (Doyle 2021)).

We excluded 32 studies that were included in Doyle 2021. That Cochrane Review addressed the use of postnatal corticosteroids commenced in the first six days after birth to prevent BPD in preterm infants (Anttila 2005Baden 1972Batton 2012Baud 2016Biswas 2003Bonsante 2007Efird 2005Garland 1999Halac 1990Hochwald 2014Kopelman 1999Lauterbach 2006Lin 1999Mukhopadhyay 1998Ng 2006Peltoniemi 2005Rastogi 1996Romagnoli 1999Sanders 1994Shinwell 1996Sinkin 2000Soll 1999Stark 2001Subhedar 1997Suske 1996Tapia 1998Vento 2004aWang 1996Watterberg 1999Watterberg 2004Yeh 1990Yeh 1997).

We excluded an additional study because it compared two different dosages of dexamethasone only (Marr 2019). We excluded other studies for a variety of reasons. See Characteristics of excluded studies.

Risk of bias in included studies

Overall most studies had low risk of bias (Figure 2; Figure 3). All were RCTs, although the method of random allocation was not always clear. Allocation concealment applied to most studies. Blinding of investigators and others was achieved most often with the use of placebo, usually saline solution. Follow‐up reporting for short‐term outcomes during primary hospitalisation most often was complete but was more variable for long‐term outcomes beyond discharge and later into childhood.


Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Ariagno 1987 was a double‐blind trial in which the pharmacist performed randomisation. Trialists provided outcomes for all enrolled infants. Follow‐up consisted of the following: investigators assessed surviving children at 12, 24, and 36 months of age, corrected for prematurity, in the High‐Risk Follow‐Up Clinic. Data included cerebral palsy and auditory status, but criteria were not defined. Personnel involved and blinding of assessors to treatment groups were unclear. The follow‐up rate of survival was 96% (23/24) (Ariagno 2000).

Avery 1985 paired and compared treatment and control infants for success in weaning. Investigators stratified infants at entry by weight into three categories: less than 1000 grams, 1000 grams to 1250 grams, and 1251 grams to 1500 grams. Within each weight group, equal numbers of treatment cards and control cards were placed into envelopes for random selection. The first treated infant and the first control infant within a given weight category made the first pair, and researchers considered in the sequential analysis only infants who were paired for weaning success. If both infants in a pair were successful or had treatment failure, the result was a tie and the pair was discarded. If one infant weaned and the other did not, the pair was scored as favouring treatment or control. The study was stopped when significance was reached from weaning from the ventilator in the sequential analysis of untied pairs. At that time, 16 infants had been studied and 14 had been matched to form seven pairs. Study authors reported no follow‐up component.

In Brozanski 1995, researchers achieved randomisation by using a random numbers table and stratified infants according to sex and birth weight (< 1000 grams versus > 999 grams). They reported treatment allocation on cards inside sequentially numbered envelopes that were kept in the pharmacy where randomisation took place. Investigators enrolled 88 infants but provided outcome data, apart from survival without supplemental oxygen at 36 weeks' postmenstrual age, for only 78 infants. They withdrew 10 infants during the study because of pharmacy error (dexamethasone group two infants, placebo group one infant), parental choice (placebo group two infants), or attending physician request (dexamethasone group one infant, placebo group four infants). All five infants withdrawn from the study by the attending physician subsequently received an extended course of dexamethasone. Follow‐up consisted of the following (Hofkosh 1995): unknown observer(s) blinded to treatment group allocation saw survivors at 12 months of age, corrected for prematurity. The follow‐up rate of survivors was 68% (44/65). Study authors did not specify criteria for the diagnosis of cerebral palsy. Psychological assessment included the Mental Developmental Index (MDI) of the Bayley Scales of Infant Development (BSID). Study authors provided no data on major neurodevelopmental disability.

CDTG 1991 assigned groups by telephone call to the Clinical Trial Service Unit in Oxford. Investigators stratified infants by clinical centre and by whether or not they were ventilator‐dependent. After completion of the trial, clinicians could give open steroids if this was clinically indicated because of life‐threatening deterioration. Researchers retained infants in the group to which they had been allocated for the purpose of analysis. They enrolled 287 infants in the trial; two were ineligible because of major malformations (Fallot's tetralogy, oesophageal atresia), leaving 285 infants included in the analysis. Follow‐up consisted of the following (Jones 1995): researchers provided data on survivors at 36 months of age, not corrected for prematurity. Primary sources of data, obtained in the UK and in Ireland, were healthcare provider visitors, who provided data on major neurosensory diagnoses or other chronic problems, and general practitioners, who provided data on health and hospitalisations. Parents completed questionnaires, including the Minnesota Child Development Inventory (CDI). Parents, healthcare visitors, and general practitioners (GPs) were unaware of treatment group allocation. In some countries, investigators sought data from paediatricians only (< 10% cases). The follow‐up rate of survivors was 94% (209/223). Trialists did not specify criteria for the diagnosis of cerebral palsy or blindness, but they defined severe hearing loss (deafness) as hearing loss requiring either hearing aids or special schooling. Major disability comprised any types of non‐ambulant cerebral palsy at three years of age, < 50% of age level on the CDI, or predicted special schooling for sensory or other impairment. Further follow‐up at 13 to 17 years of age consisted of the following (Jones 2005a; Jones 2005b): assessors who were blinded to treatment group allocation assessed surviving children from the 25 individual British and Irish study centres at 13 to 17 years of age. Families completed a questionnaire on functional status, diagnoses of potentially disabling conditions (visual or hearing impairment, learning disabilities, cerebral palsy, and epilepsy), and the child's schooling. Study authors asked GPs to complete a questionnaire to report known functional problems, diagnoses, and hospital admissions. The paediatrician responsible for each child's care made the diagnosis of cerebral palsy. One of three research nurses blinded to the children's original treatment allocation visited surviving children at home. They administered a non‐verbal reasoning test and the British Picture Vocabulary Scale and averaged these scores as a proxy for IQ. Investigators defined moderate disability as consisting of one or two of the following: IQ 2 to 3 standard deviations (SD) below the mean, ambulatory cerebral palsy, hearing deficits corrected with hearing aids, impaired vision, or a behaviour disorder with a major impact on schooling. They defined severe disability as any of the following: IQ > 3 SDs below the mean, wheelchair‐dependent cerebral palsy, uncorrectable hearing loss, blindness (perception of light only), or three moderate disabilities. Respiratory function included spirometry to measure forced expiratory volume in one second (FEV₁), forced vital capacity (FVC), the FEV₁/FVC ratio, and forced expired flow from 25% to 75% (FEF)25%-75%; study authors expressed results and growth measurements as standardised scores (z‐scores). They assessed other outcomes, but we did not include them in the review. These included data on types of schooling, teacher questionnaires on a child's ability, and the Strengths and Difficulties Questionnaire. The follow‐up rate of eligible survivors at 13 to 17 years was 77% (150/195), including data from five severely disabled children at three years of age who were not contacted as teenagers.

In Cummings 1989, investigators achieved randomisation by sequential assignment from a table of random numbers known only to a pharmacist who had no knowledge of the clinical status of infants. Study authors present outcome data for all 36 infants enrolled in the study. This trial included two experimental groups: one treated for 18 days, and the other treated for 42 days, compared with a single control group. For these analyses, we combined treatment groups (n = 25) and compared the combined data with data from the control group (n = 11). Follow‐up consisted of the following: a paediatrician and an occupational therapist saw survivors at 15 months of age, corrected for prematurity. Observers were blinded to treatment group allocation. The follow‐up rate of survivors was 100% (23/23). Researchers specified criteria for the diagnosis of cerebral palsy but did not specify criteria for blindness or deafness. Psychological assessment included the MDI and the Psychomotor Developmental Index (PDI) of the BSID. Major disability comprised any of the following: cerebral palsy or MDI or PDI < 1 SD. Investigators later assessed survivors at four years of age and confirmed neurological status for all participants (Cummings 2002 (personal communication follow‐up of Cummings 1989)). Researchers provided further follow‐up at 15 years of age (Gross 2005). Assessors were blinded to treatment group allocation. Outcomes included growth (body size converted to z‐scores), general health, respiratory morbidity, and respiratory function testing. Cognition was assessed by the Wechsler Intelligence Scale for Children ‐ Third Edition (WISC‐III). Teachers completed data on class repetition, performance, and behaviour. Pulmonary function testing included spirometry to measure FEV₁, FVC, and FEF25%-75%, along with measurement of lung volumes (total lung capacity (TLC) and residual volume (RV)) by nitrogen washout; study authors expressed results as % predicted for age, height, and sex. Trial authors reported numbers of surviving children with ongoing respiratory symptoms of wheezing or congestion and interpreted these as a diagnosis of asthma for meta‐analysis. They defined intact survival as a normal neurological examination, IQ > 70, and receiving education in a normal classroom. For the meta‐analysis, investigators defined major neurological disability as any of an abnormal neurological examination (i.e. cerebral palsy), cognitive delay (IQ < 71), or not in a regular classroom (with or without additional help). They did not measure blood pressure.

Doyle 2006 was a double‐blind trial with randomisation performed centrally by non‐clinical staff independent of the chief investigators, with random variation in block sizes of two to eight for each centre. Syringes were prepared and labelled identically within the pharmacy department at the centre, concealing treatment allocation from study site investigators and the infant's caregivers. They discouraged but did not prohibit open‐label corticosteroids after randomisation; some infants may have received both a second course of their initially allocated study drug and open‐label corticosteroids. No one apart from the pharmacists at individual study sites had access to the treatment code. Trial authors reported short‐term outcomes for all enrolled infants. Follow‐up included the following (Doyle 2007 (follow‐up publication of Doyle 2006)): paediatricians and psychologists who were blinded to treatment group allocation assessed surviving children at 24 months of age, corrected for prematurity, at individual study sites. They considered children to have a neurosensory impairment if they had cerebral palsy (criteria included abnormalities of tone and motor dysfunction), blindness (bilateral vision worse than 6/60), deafness requiring hearing aids or worse, or developmental delay (defined as a MDI on the BSID < 85 (< ‐1 SD) (Bayley 1993). Researchers graded severity of the neurosensory disability imposed by the impairment as follows: severe ‐ bilateral blindness, cerebral palsy with the child unlikely ever to walk, or MDI < 55 (< ‐3 SD); moderate ‐ deafness, cerebral palsy in children not walking at two years but expected to walk, or MDI from 55 to < 70 (‐3 SD to < ‐2 SD); mild ‐ cerebral palsy but walking at two years with only minimal limitation of movement or MDI 70 to < 85 (< ‐2 SD to ‐1 SD). They considered the remaining children to have no neurosensory disability. Major neurosensory disability comprised moderate or severe disability. The follow‐up rate of survivors at two years was 98% (58/59).

In Durand 1995, investigators performed randomisation via blind drawing of random cards contained in sealed envelopes. Clinical personnel were not aware of the group assignment of any infant. Study authors present outcome data for 43 of the 44 enrolled infants. They excluded one infant in the control group from all analyses as the result of birth weight < 500 grams. Follow‐up consisted of the following (Durand 2012 (personal communication follow‐up of Durand 1995)): a developmental paediatrician, a paediatric neurologist, and other specialised personnel (including a psychologist) assessed surviving children at 12 months of age, corrected for prematurity. A paediatric ophthalmologist performed all eye examinations. All staff were blinded to treatment group allocation. Children were considered to have a neurosensory impairment if they had cerebral palsy (defined as non‐progressive motor impairment with abnormal muscle tone and decreased range of movement), blindness (bilateral vision worse than 6/60), deafness requiring hearing aids or worse, or developmental delay (defined as MDI < 70 on the BSID). The follow‐up rate of survivors at 12 months was 78% (29/37).

Harkavy 1989 achieved randomisation by using random numbers held in the pharmacy. Clinicians and investigators were unaware of treatment assignments. Study authors provided outcome data for 21 of the 22 enrolled infants. One infant died after consent but before random assignment to a treatment group. Follow‐up consisted of the following (Harkavy 2002 (personal communication follow‐up of Harkavy 1989)): a neonatologist and an occupational therapist saw survivors at ages ranging from 6 to 24 months, corrected for prematurity. Observers were blinded to treatment group allocation. The follow‐up rate of survivors was 32% (6/19). Trialists did not specify criteria for the diagnosis of cerebral palsy, blindness, or deafness. Psychological assessment included the MDI of the BSID. Study authors did not define major disability.

In Kari 1993, researchers performed randomisation in blocks of 10 for each participating hospital. Clinicians and investigators were unaware of treatment assignments. Study authors present outcomes for all 41 infants enrolled in the trial. The number of infants recruited was only 25% of the estimate required for the sample size. Therefore, the study was discontinued after 26 months. Follow‐up consisted of the following (Mieskonen 2003): only one of four centres in this multi‐centre study provided follow‐up; this centre contributed 23 of the 41 participants to the original study. Three infants died before discharge (one dexamethasone; two placebo). No late deaths in childhood are known. Survivors were followed in the hospital's outpatient clinic. One child in the dexamethasone group had deafness requiring a hearing aid, seizures treated with anticonvulsants, and attention deficit hyperactivity disorder, and required assistance with schooling but did not have cerebral palsy at 7.8 years of age. This child would not co‐operate with the respiratory component of the study. Another child in the dexamethasone group had no confirmed cerebral palsy at 2.6 years of age and was not traced at school age but was said to be attending normal school. One child in the placebo group had multiple difficulties in speech and cognitive function at five years of age and was expected to require extra help at school but refused further follow‐up. Another child in the placebo group had minor difficulties in comprehension at five years of age but was lost to further follow‐up. In total, 16 children participated in the follow‐up study at seven to nine years of age. Neurological status at five years of age was obtained from hospital records, including assessments for cerebral palsy (abnormal muscle tone, increased tendon reflexes and positive Babinski sign, or persistent or exaggerated primitive reflexes, dyskinesia, or ataxia), visual or hearing deficits, and school maturity (details of testing not given). Severe disability comprised any of more than mild cerebral palsy, severe global delay (not defined), or sensory or other impairment requiring special schooling; moderate disability comprised any of mild cerebral palsy, severe deafness, moderate global delay (extra help needed at school, assessment of global retardation or language problems), or home oxygen beyond three years of age. For this meta‐analysis, we have extracted data for major neurological disability for those with more than mild cerebral palsy, blindness, or deafness, or needing extra help with schooling. One investigator blinded to neonatal details then assessed children at 7.8 to 9.2 years of age, including presumably treatment group allocation. Age was not corrected for prematurity. Study authors measured children for height and weight and performed lung function tests, electrocardiography (ECG), and echocardiography.

In Kazzi 1990, randomisation was assigned by drawing a pre‐coded card prepared from a table of random numbers. Infants were stratified by birth weight into three groups: less than 1000 grams, 1000 grams to 1250 grams, and 1251 grams to 1500 grams. The pharmacist drew the card from the appropriate group, and neither investigators nor nursery staff were aware of the treatment group. Study authors provided outcome data for all 23 enrolled infants and reported no follow‐up component.

In Kothadia 1999, researchers randomised infants within six strata, defined in terms of birth weight (500 grams to 800 grams, 801 grams to 1100 grams, and 1101 grams to 1500 grams) and sex, with a block size of eight. They did not describe the exact method of randomisation. Control infants were given an equal volume of normal saline. Investigators assessed outcome data in a blinded fashion. Study authors initially described zero cross‐over in this trial, but review of data at age 19 years revealed that one child who was randomised to placebo received a 42‐day tapering course of placebo, then subsequently a 12‐day tapering course of dexamethasone. In addition, three of the children randomised to placebo received 24‐hour courses of dexamethasone for upper airway oedema. Follow‐up consisted of the following: a developmental paediatrician or one of two neonatologists and a physical therapist saw survivors at 12 months of age, corrected for prematurity, if any neurological abnormality was detected. Observers were blind to treatment group allocation. The follow‐up rate of survivors at 12 months of age was 98% (93/95). Trialists specified criteria for the diagnosis of cerebral palsy. Paediatric ophthalmologists diagnosed blindness. Study authors did not define deafness. Psychological assessment included the MDI of the BSID; investigators assessed the first 10 infants using the original Bayley Scales, and the remainder using BSID‐II. Major disability comprised any of cerebral palsy, blindness, or an MDI < ‐2 SD. Children were assessed again at 4 to 6 years of age and at 8 to 11 years of age (Nixon 2007; O'Shea 2007; Washburn 2006). Parents, children, and follow‐up examiners were not aware of children's randomisation assignment. Investigators diagnosed cerebral palsy at four to six years if the child had a neuromotor abnormality detected on neurological examination by a nurse with specialised training in neurodevelopmental follow‐up, and if the parent reported that the child was receiving treatment for cerebral palsy. A parent was interviewed again at the 8‐ to 11‐year visit as to whether a diagnosis of cerebral palsy had ever been made. For intelligence and academic achievement, at the four‐ to six‐year visit, a child psychologist assessed the child using the Differential Abilities Scales (DAS), the Kaufman Survey of Early Academic and Language Skills (K‐SEALS), and the Vineland Adaptive Behavioral Scales (VABS). At the 8‐ to 11‐year visit, a child psychologist assessed the child using the Wechsler Individual Achievement Tests (WIAT), the Wechsler Intelligence Scale for Children ‐ Third Edition (WISC‐III), and the Vineland Adaptive Behavior Scale (VABS). Investigators defined a major neurodevelopmental impairment at 4 to 6 years and/or at 8 to 11 years as cerebral palsy, and at 4 to 6 years of age as mental retardation (IQ < 70 on either the DAS (n = 11 participants) or the WISC‐III (n = 71 participants) and a VABS composite score < 70) at last follow‐up. For five dexamethasone‐treated and eight placebo‐treated children who did not undergo intelligence testing at 4 to 6 years or at 8 to 11 years of age, they defined major neurodevelopmental impairment as blindness, cerebral palsy (at the most recent visit), or a Bayley MDI < 70 for adjusted age. All survivors were assessed at least once at or beyond one year of age. The follow‐up rate at 4 to 11 years of age was 88% (84/95). Respiratory data were collected at 8 to 11 years of age via pulmonary function testing. Researchers obtained forced expiratory flow rates and volumes (FVC, FEV₁, FEV₁/FVC ratio, and FEF25% -75%) expressed as % of predicted as appropriate, and considered abnormal if below the fifth percentile. They determined TLC and RV from body plethysmography and expressed these as a ratio (RV/TLC), as well as pulmonary diffusing capacity (diffusing capacity of the lungs for carbon monoxide (DLCO)) via the single‐breath carbon monoxide technique. However, most children could not cope with plethysmography and the single‐breath diffusion manoeuvre, hence study authors did not analyse TLC, RV, and diffusing capacity data. Investigators also assessed asthma diagnosis and airway reactivity. They categorised children as having asthma if the parent or guardian reported that the child had asthma, had used medications for asthma treatment, or both. A sub‐sample of children underwent maximal progressive exercise testing on a cycle ergometer as part of the larger study. Researchers repeated spirometry immediately and five minutes post exercise, as well as 20 minutes following three puffs of albuterol delivered with a spacer. They used a 15% decrease in FEV₁ from pre‐exercise values as the criterion to define exercise‐induced bronchoconstriction, and they considered a 12% increase in FEV₁ from pre‐exercise levels to be a positive bronchodilator response. The follow‐up rate at 8 to 11 years of age for respiratory data was 72% (68/95) but was 66% (63/95) for respiratory function testing. 

Kovacs 1998 assigned eligible infants using a "blocked" randomisation procedure, and only the designated pharmacist who prepared all study medications was aware of group assignments. Researchers stratified infants before randomisation into two categories according to gestational age (22 to 26 weeks versus 27 to 29 weeks). Follow‐up consisted of the following (Kovacs 2002 (personal communication follow‐up of Kovacs 1998)): study authors obtained data from the regular follow‐up clinic at ages up to 90 months in 70% (33/47) of survivors and did not specify personnel involved, blinding of assessors to treatment group, and criteria for various diagnoses, including cerebral palsy and major disability.

Noble‐Jamieson 1989 did not describe the method of randomisation. Medical and nursing staff were unaware of the drug given. Study authors provided outcome data for all 18 enrolled infants and reported no follow‐up component.

Ohlsson 1992 performed randomisation by using computer‐generated random numbers and wrote down allocation groups on cards enclosed in opaque envelopes and kept under lock in the pharmacy. Envelopes were available only to the pharmacist who drew the appropriate card and distributed the study drug. We have described under Description of studies the problem of administering placebo. Trialists discontinued treatment for suspected infection in one infant in each group and treatment for blood transfusion‐derived cytomegalovirus in one infant in the study group. They provided outcome data for all enrolled infants. Follow‐up consisted of the following (Ohlsson 1990 (additional publication of Ohlsson 1992)): researchers saw survivors in the regular follow‐up clinic up to at least 18 months of age in 96% (23/24) of cases; the remaining survivor was developing normally when last seen at 12 months of age. Age was probably not corrected for prematurity. Study authors did not specify personnel involved nor blinding of observers, neither did they specify criteria for the diagnoses of cerebral palsy and blindness. Psychological assessment included the MDI of the BSID.

In Onland 2019, the randomisation schedule was computer‐generated, with stratification for study centre and gestational age in two groups (< 27 weeks and > 26 weeks), The allocation ratio was 1:1 with block randomisation using variable block sizes. Multiple birth infants were randomised independently, unless parents or caretakers explicitly demanded that siblings should be in the same treatment arm. Infants’ parents and all members of the child's medical team and investigators were blinded to group allocation throughout the study. Survivors were assessed at two years' corrected age. Neurodevelopmental impairment was defined as presence of one or more of the following: cognitive and/or motor composite score less than 85 on the Bayley Scales of Infant and Toddler Development Third Edition (Dutch version), cerebral palsy greater than level II in the Gross Motor Function Classification System, or hearing or visual impairment. Additional data were obtained from the authors on the rates of cerebral palsy (personal communication, September 2021).

In Papile 1998, random assignment took place at each centre's pharmacy via the urn method ‐ a procedure that promotes equal distribution of participants among treatment groups. To blind clinical staff to treatment group assignment, investigators prepared different volumes of placebo (saline) to match the various doses of dexamethasone. They reported no follow‐up component.

In Parikh 2013, an individual not involved in the study generated the randomisation sequence, but study authors did not specify the precise method. They described two strata ‐ one for birth weight (< 751 grams versus 751 to 1000 grams) and one for respiratory index score (2 to 4 versus > 4). Access to the randomisation assignment was limited to two study pharmacists, and blinding was maintained by using an identical volume of saline placebo. Follow‐up consisted of the following (Parikh 2013; Parikh 2015): certified examiners assessed survivors at 18 to 22 months' corrected age and were blinded to group allocation. Certified examiners diagnosed cerebral palsy and specified the criteria for diagnosis. Study authors defined bilateral deafness as bilateral hearing loss requiring amplification, and bilateral blindness as bilateral vision loss with only form or shadow vision or no useful vision. Psychological assessment included the Bayley Scales of Infant and Toddler Development ‐ Third Edition (Bayley III). Investigators defined any neurodevelopmental impairment as any of cerebral palsy, cognitive delay, language delay, blindness, or deafness.

Romagnoli 1997 achieved random allocation by opening numbered, sealed envelopes. Researchers did not give placebo to control infants. They reported outcome measures for all 30 infants included in the study. Follow‐up consisted of the following (Romagnoli 2002): one paediatrician and one neurologist saw survivors at 36 to 42 months of age, corrected for prematurity, with observers blinded to treatment group allocation. The follow‐up rate of survivors was 100% (30/30). The neurologist made the diagnosis of cerebral palsy, but study authors did not specify the criteria used and reported no specific criteria for blindness and deafness. Psychological assessment included the Stanford Binet Test ‐ Third Revision. Study authors provided no data on major disability.

Scott 1997 achieved randomisation using a random numbers table. Blinding was maintained by using an identical volume of saline placebo. There was no follow‐up component.

Vento 2004 did not state the method of randomisation. It is not clear whether clinicians caring for infants or those assessing outcomes were blinded to treatment group assignment. The control group did not receive a placebo. Follow‐up consisted of the following (Vento 2012 (personal communication follow‐up of Vento 2004)): a paediatric neurologist who was blinded to treatment group allocation assessed surviving children between one and four years of age, corrected for prematurity up to two years. They considered children to have a major neurosensory impairment if they had non‐ambulant cerebral palsy, blindness (bilateral vision worse than 6/60), deafness requiring hearing aids or worse, or severe cognitive delay (defined as IQ < 55). The follow‐up rate of survivors at a mean age of 26 months was 100% (18/18).

Vincer 1998 achieved random allocation but did not describe in the abstract the method used. Control infants were given equal volumes of saline placebo, which concealed treatment allocation. Follow‐up consisted of the following (Vincer 2002 (personal communication follow‐up of Vincer 1998)): one of two neonatologists saw survivors at 24 months of age, corrected for prematurity. They referred children with a developmental abnormality to a neurologist. Observers were blind to treatment group allocation. The follow‐up rate of survivors was 100% (17/17). Study authors specified criteria for the diagnosis of cerebral palsy, but not for blindness or deafness. Psychological assessment included the MDI of the BSID. Major disability comprised any of moderate or severe cerebral palsy, bilateral blindness, deafness, or MDI < 2 SD.

In Walther 2003, a staff pharmacist was in charge of randomisation and drug preparation. Investigators and clinical caregivers were unaware of treatment allocation. Infants in the control group received a saline placebo. Open‐label steroid therapy was used only if it became essential for management of ventilator dependency, ideally seven days after completion of therapy and at the discretion of the attending neonatologist. Follow‐up consisted of the following (Walther 2012 (personal communication follow‐up of Walther 2003)): surviving children were assessed at between one and four years of age with no details provided about correction for prematurity and personnel involved; however, trial personnel were blinded to knowledge of treatment group allocation. They defined developmental delay as MDI < 70 on the BSID. The follow‐up rate of survivors was 78% (25/32).

In Yates 2019, randomisation was managed via a secure web‐based randomisation facility hosted centrally, with continuous telephone backup available. The randomisation programme used a minimisation algorithm to ensure balance between trial groups, with respect to collaborating hospital, sex, multiple births, gestational age at birth, and existing diuretic therapy for the 24 hours before randomisation. Multiple births from the same family were randomised individually. Dexamethasone and saline placebo were supplied in identical vials, and hence staff were unaware of treatment allocation. Open‐label treatment was allowed after the 16‐day intervention period had elapsed; however open‐label could start earlier at clinician discretion. There was no long‐term follow‐up component.

Allocation

We found little evidence of allocation bias overall; most studies had no evidence of allocation bias, and in a small minority the risk was unclear.

Blinding

We found little evidence of blinding bias overall; most studies had no evidence of blinding bias, but small minorities had unclear or high risk of blinding bias.

Incomplete outcome data

We found little evidence of attrition bias overall; most studies had no evidence of attrition bias, and a small minority had unclear risk.

Selective reporting

Just over one‐half of studies had no evidence of selective reporting bias, and the remainder had unclear risk of selective reporting bias.

Other potential sources of bias

A majority of studies used a valid method of random sequence generation, but in approximately 40% of studies, the methods used for randomisation were unclear.

Effects of interventions

See: Summary of findings 1 Systemic corticosteroids (dexamethasone or hydrocortisone) compared with control (placebo or nothing) for chronic lung disease in preterm infants

Results of meta‐analysis

Meta‐analysis of these 23 studies yielded the following results.

Mortality

Evidence indicates that late systemic corticosteroid treatment was associated with reduced mortality at all ages: 28 days (typical risk ratio (RR) 0.60, 95% confidence interval (CI) 0.40 to 0.89; typical risk difference (RD) ‐0.04, 95% CI ‐0.09 to ‐0.00; 7 studies, 970 infants; Analysis 1.1), 36 weeks' postmenstrual age (typical RR 0.70, 95% CI 0.52 to 0.94; typical RD ‐0.05, 95% CI ‐0.10 to ‐0.01; 15 studies, 1029 infants; Analysis 1.2), before hospital discharge (typical RR 0.79, 95% CI 0.63 to 0.98; typical RD ‐0.04, 95% CI ‐0.08 to ‐0.00; 20 studies, 1406 infants; Analysis 1.3), and at latest reported age (RR 0.81, 95% CI 0.66 to 0.99; RD ‐0.05, 95% CI ‐0.09 to ‐0.00; 21 studies, 1428 infants; Analysis 1.4).

No evidence suggests publication bias for mortality at latest reported age upon examination of a funnel plot (Egger test, P = 0.78) (Figure 4).


Funnel plot of comparison: 1 Mortality, outcome: 1.4 Mortality at latest reported age.

Funnel plot of comparison: 1 Mortality, outcome: 1.4 Mortality at latest reported age.

Bronchopulmonary dysplasia

The incidence of BPD was significantly decreased at 28 days of life (typical RR 0.90, 95% CI 0.84 to 0.95; typical RD ‐0.11, 95% CI ‐0.17 to ‐0.05; 7 studies, 994 infants; Analysis 2.1), and at 36 weeks' postmenstrual age (typical RR 0.89, 95% CI 80 to 0.99; typical RD ‐0.07, 95% CI ‐0.13 to ‐0.01; 14 studies, 988 infants; Analysis 2.2), but the evidence was not as strong at 36 weeks' postmenstrual age among survivors (typical RR 0.91, 95% CI 0.82 to 1.01; typical RD ‐0.06, 95% CI ‐0.13 to 0.01; 9 studies, 624 infants; Analysis 2.3). We noted strong evidence of publication bias upon examining a funnel plot for BPD at 36 weeks (Egger test, P < 0.001) (Figure 5). Data show reduced need for late rescue with corticosteroids (typical RR 0.48, 95% CI 0.41 to 0.57; typical RD ‐0.20, 95% CI ‐0.24 to ‐0.16; 15 studies, 1489 infants; Analysis 2.4) and reduced need for home oxygen both overall (typical RR 0.71, 95% CI 0.54 to 0.94; typical RD ‐0.08, 95% CI ‐0.14 to ‐0.01; 7 studies, 611 infants; Analysis 2.5) and for survivors only (typical RR 0.69, 95% CI 0.51 to 0.94; typical RD ‐0.13, 95% CI ‐0.24 to ‐0.03; 6 studies, 277 infants; Analysis 2.6).


Funnel plot of comparison: 2 Bronchopulmonary dysplasia (BPD), outcome: 2.2 BPD at 36 weeks' postmenstrual age.

Funnel plot of comparison: 2 Bronchopulmonary dysplasia (BPD), outcome: 2.2 BPD at 36 weeks' postmenstrual age.

Mortality or bronchopulmonary dysplasia

Strong evidence indicates that the combined outcome of mortality or BPD was decreased both at 28 days of life (typical RR 0.87, 95% CI 0.83 to 0.91; typical RD ‐0.12 to 95% CI ‐0.16 to ‐0.08; 6 studies, 934 infants; Analysis 3.1) and at 36 weeks' postmenstrual age (RR 0.85, 95% CI 0.79 to 0.92; RD ‐0.12, 95% CI ‐0.17 to ‐0.07; 14 studies, 988 infants; Analysis 3.2). We found little evidence of publication bias upon examining a funnel plot for mortality or BPD at 36 weeks (Egger test, P = 0.33) (Figure 6).


Funnel plot of comparison: 3 Mortality or BPD, outcome: 3.2 Mortality or BPD at 36 weeks' postmenstrual age.

Funnel plot of comparison: 3 Mortality or BPD, outcome: 3.2 Mortality or BPD at 36 weeks' postmenstrual age.

Failure to extubate

Failure to extubate was significantly decreased at three days (typical RR 0.83, 95% CI 0.78 to 0.88; typical RD ‐0.16, 95% CI ‐0.21 to ‐0.11; 10 studies, 764 infants; Analysis 4.1), at seven days (typical RR 0.67, 95% CI 0.62 to 0.73; typical RD ‐0.27, 95% CI ‐0.32 to ‐0.22; 17 studies, 1130 infants; Analysis 4.2), at 14 days (typical RR 0.65, 95% CI 0.53 to 0.80; typical RD ‐0.19, 95% CI ‐0.28 to ‐0.10; 5 studies, 458 infants; Analysis 4.3), and at 28 days (typical RR 0.57, 95% CI 0.37 to 0.89; typical RD ‐0.14, 95% CI ‐0.25 to ‐0.03; 3 studies, 236 infants; Analysis 4.4).

Complications during primary hospitalisation
Metabolic complications

Results show increased risks of hyperglycaemia (typical RR 1.59, 95% CI 1.34 to 1.89; typical RD 0.10, 95% CI 0.07 to 0.14; 19 studies, 1684 infants; Analysis 5.2) and glycosuria (typical RR 8.03, 95% CI 2.43 to 26.5; typical RD 0.72, 95% CI 0.52 to 0.91; 2 studies, 48 infants; Analysis 5.3), as well as increased risk of hypertension (typical RR 1.67, 95% CI 1.19 to 2.33; typical RD 0.04, 95% CI 0.01 to 0.06; 17 studies, 1628 infants; Analysis 5.4).

Gastrointestinal complications

We found little evidence for differences in gastrointestinal complications: necrotising enterocolitis (typical RR 0.92, 95% CI 0.62 to 1.38; 11 studies, 1409 infants; Analysis 5.6), gastrointestinal bleeding (typical RR 1.33, 95% CI 0.97 to 1.83; 9 studies, 1385 infants; Analysis 5.7), or gastrointestinal perforation (RR 0.67, 95% CI 0.26 to 1.70; 5 studies, 552 infants; Analysis 5.8).

Other complications

No evidence suggests that infection rates were different between groups (typical RR 1.03, 95% CI 0.91 to 1.16; 20 studies, 1742 infants; Analysis 5.1). Evidence indicates an increase in severe retinopathy of prematurity overall (typical RR 1.27, 95% CI 1.03 to 1.58; typical RD 0.06, 95% CI 0.01 to 0.12; 13 studies, 929 infants; Analysis 5.9), but not among survivors (typical RR 1.17, 95% CI 0.94 to 1.45; 10 studies, 697 infants; Analysis 5.10). Evidence shows an increase in hypertrophic cardiomyopathy (typical RR 2.76, 95% CI 1.33 to 5.74; typical RD 0.13, 95% CI 0.05 to 0.20; 4 studies, 238 infants; Analysis 5.11). We found little evidence for real reductions in pneumothorax (typical RR 0.89, 95% CI 0.53 to 1.49; 3 studies, 157 infants; Analysis 5.12) or in severe intraventricular haemorrhage (typical RR 0.54, 95% CI 0.26 to 1.11; 7 studies, 639 infants; Analysis 5.13).

Follow‐up data

  • Rates of children with low cut‐off scores for the Mental Developmental Index on the Bayley Scales were little affected overall (typical RR 0.81, 95% CI 0.47 to 1.38; 7 studies, 333 infants; Analysis 6.1) or among survivors assessed (typical RR 0.74, 95% CI 0.45 to 1.22; 7 studies, 232 infants; Analysis 6.2). Rates of children with low cut‐off scores for the Psychomotor Developmental Index on the Bayley Scales were little affected overall (typical RR 0.78, 95% CI 0.34 to 1.80; 1 study, 118 infants; Analysis 6.3) or among survivors assessed (typical RR 0.67, 95% CI 0.30 to 1.50; 1 study, 90 infants; Analysis 6.4)

  • The increase in retinopathy of prematurity did not translate into a significant increase in blindness overall (typical RR 0.78, 95% CI 0.35 to 1.73; 13 studies, 784 infants; Analysis 6.5) or among survivors assessed (typical RR 0.77, 95% CI 0.35 to 1.67; 13 studies, 539 infants; Analysis 6.6)

  • We found little evidence for a difference in rates of deafness overall (typical RR 0.56, 95% CI 0.26 to 1.27; 9 studies, 936 infants; Analysis 6.7) or among survivors assessed (typical RR 0.62, 95% CI 0.29 to 1.36; 9 studies, 616 infants; Analysis 6.8)

  • We found little evidence for a difference in rates of cerebral palsy at the latest reported age overall (typical RR 1.17, 95% CI 0.84 to 1.61; 17 studies, 1290 infants; Analysis 6.10) or among survivors assessed at the latest reported age (typical RR 1.15, 95% CI 0.81 to 1.61; 16 studies, 628 infants; Analysis 6.16). A funnel plot for the outcome of cerebral palsy at latest reported age provided little evidence of publication bias (Egger test, P = 0.13) (Figure 7). Cerebral palsy was not significantly increased in studies limited to the first three years after birth (typical RR 1.11, 95% CI 0.81 to 1.51; 16 studies, 1311 infants; Analysis 6.9), or among survivors assessed at 1‐3 years of age (typical RR 1.08, 95% CI 0.79 to 1.47; 16 studies, 923 infants; Analysis 6.15. The combined rate of mortality or cerebral palsy was little affected in studies limited to the first three years after birth (typical RR 0.89, 95% CI 0.76 to 1.04; 16 studies, 1311 infants; Analysis 6.13). The combined rate of either mortality or cerebral palsy at latest reported age was not significantly different (typical RR 0.90, 95% CI 0.76 to 1.06; 16 studies, 1290 infants; Analysis 6.14). A funnel plot for the outcome mortality or cerebral palsy at latest reported age provided little evidence of publication bias (Egger test, P = 0.99) (Figure 8)

  • Major neurosensory disability was not significantly different overall (typical RR 1.09, 95% CI 0.88 to 1.34; 10 studies, 1090 infants; Analysis 6.17) or among survivors assessed (typical RR 1.01, 95% CI 0.83 to 1.22; 10 studies, 778 infants; Analysis 6.20). The combined rate of mortality or major neurosensory disability was not significantly different (typical RR 0.96, 95% CI 0.85 to 1.08; 10 studies, 1090 infants; Analysis 6.19)

  • The rate of abnormal neurological examination overall was increased (typical RR 1.81, 95% CI 1.05 to 3.11; typical RD 0.13, 95% CI 0.02 to 0.24; 4 studies, 200 infants; Analysis 6.21), but the clinical importance of this finding is unclear in the absence of important increases in cerebral palsy or major neurosensory disability. Rates of the combined outcome of mortality or abnormal neurological examination were not significantly different between groups (typical RR 0.96, 95% CI 0.71 to 1.31; 4 studies, 200 infants; Analysis 6.23)

  • The only study reporting re‐hospitalisation rates over the first five years noted little evidence of a difference between groups (Analysis 6.25Analysis 6.26). The same study with follow‐up of survivors to five years noted little evidence for increased maternal reports of wheezing (RR 1.47, 95% CI 0.82 to 2.64; 1 study, 74 infants; Analysis 7.1), need for corrective lenses (RR 1.61, 95% CI 0.82 to 3.13; 1 study, 74 infants; Analysis 7.2), and need for physical therapy (RR 1.49, 95% CI 0.71 to 3.11; 1 study, 74 infants; Analysis 7.3), and a non‐significant decrease in the need for speech therapy (RR 0.46, 95% CI 0.21 to 1.02; 1 study, 74 infants; Analysis 7.4)

  • Data show no substantial differences between groups for other outcomes in later childhood, including IQ, respiratory health or function, blood pressure, and growth, with the exceptions of a significant reduction in rates of children with FEV₁ < ‐2 SD (typical RR 0.58, 95% CI 0.36 to 0.94; 2 studies, 187 infants; Analysis 8.2), increased standardised mean difference for FEV₁ (standardised mean difference (SMD) 0.28, 95% CI 0.01 to 0.55; 4 studies, 222 participants; Analysis 8.5), and increased mean difference for forced vital capacity (MD 7.68% predicted, 95% CI 1.69 to 13.68; 3 studies, 98 participants; Analysis 8.7).


Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.10 Cerebral palsy at latest reported age.

Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.10 Cerebral palsy at latest reported age.


Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.14 Mortality or cerebral palsy at latest reported age.

Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.14 Mortality or cerebral palsy at latest reported age.

Differences by type of corticosteroid used

When such comparisons were possible, there were few outcomes for which evidence shows differences in treatment effects between RCTs involving dexamethasone and those involving only hydrocortisone. Notable exceptions were that effects of corticosteroids in reducing BPD at 36 weeks (Analysis 2.2), in reducing mortality or BPD at 36 weeks (Analysis 3.2), and in increasing the rate of hypertension (Analysis 5.4) all arose from treatment with dexamethasone ‐ not with hydrocortisone. 

Sensitivity analysis, excluding studies with higher risk of bias

Two studies had higher risk of bias largely because they included no control groups, and hence blinding to knowledge of treatment allocation was not possible (Durand 1995Romagnoli 1997). Both studies involved dexamethasone only. Excluding these two studies from major outcomes of mortality at latest age, BPD at 36 weeks, combined mortality or BPD at 36 weeks, cerebral palsy, or mortality or cerebral palsy had little effect on most odds ratios or on any CIs, and altered no conclusions, with one exception; for outcomes of BPD at 36 weeks for all studies combined, the typical RR was 0.89 (95% CI 0.80 to 0.99) with all studies included (Analysis 2.2), but was changed slightly to typical RR 0.93 (95% CI 0.83 to 1.03) when the two studies were excluded.

Results of individual trials

Ariagno 1987: total respiratory system compliance improved in the dexamethasone group (P < 0.05). Time from initiation of treatment to first extubation was shorter for the dexamethasone group (6 versus 45 days; P = 0.0006), but time to final extubation was not significantly different (30 versus 48 days). Data show 10 deaths ‐ five in the dexamethasone group and five in the control group ‐ all occurring after the treatment period. Proportionate weight gain was greater among control infants (P < 0.003) during treatment. Five dexamethasone‐treated infants had infection, as did two in the control group. Hyperglycaemia and hypertension were similar between groups. At follow‐up, cerebral palsy was detected in one child in the dexamethasone group at 36 months of age and in three controls at 12 months of age.

Avery 1985: sequential analysis exceeded the criterion (P < 0.005) when seven consecutive untied pairs showed weaning with dexamethasone and failure to wean in control infants. Pulmonary compliance improved by 64% in the treated group and by 5% in the control group (P < 0.01). Results show no significant intergroup differences in mortality, length of hospital stay, sepsis, hypertension, hyperglycaemia, or electrolyte abnormalities.

Brozanski 1995: at 36 weeks' postmenstrual age, results show a significant increase in survival rates without oxygen supplementation (17/39 versus 7/39; P = 0.03) and a significant decrease in the incidence of BPD (46% versus 23%; P = 0.047) in the group that received pulse dexamethasone therapy. Supplemental oxygen requirements were less throughout the study period in the dexamethasone group (P = 0.013). Mortality and durations of supplemental oxygen, ventilator support, and hospital stay did not differ significantly between groups. The need for insulin therapy for hyperglycaemia was increased in the dexamethasone group (P < 0.05). At follow‐up, data show no significant differences between groups in rate of cerebral palsy among survivors assessed (20% versus 21%). Rate of death or survival among randomised children with cerebral palsy was lower in the dexamethasone group (23% versus 33%), but this difference was not statistically significant. The mean Mental Developmental Index (MDI) was 89.5 (SD 23.7) in the dexamethasone group and 80.8 (SD 26.0) in the control group ‐ a non‐significant difference.

CDTG 1991: dexamethasone treatment significantly reduced the duration of mechanical ventilation among infants who were ventilator‐dependent at entry (median days for survivors, 11 versus 17.5). Data show no statistically significant differences between total groups of survivors in time receiving supplemental oxygen and length of stay in hospital, although trends favoured the dexamethasone group. Twenty‐five infants in each group died before hospital discharge; most were ventilator‐dependent at trial entry. Open treatment with steroids was later given to 18% of the dexamethasone group and 43% of the placebo group (P < 0.001). We found little evidence of serious side effects and noted that infection rates in particular were similar in the two groups. At follow‐up, results show no clear differences between randomised groups in the original study for outcomes at three years. This conclusion held when data for cerebral palsy, blindness, and deafness were updated on the basis of results obtained at 13 to 17 years of age. Rates of intellectual impairment and moderate and severe disability at 13 to 17 years of age were similar in both groups, and data reveal no substantial differences in lung function or growth z‐scores, nor in proportions with high blood pressure.

Cummings 1989: infants in the 42‐day dexamethasone group, but not those in the 18‐day group, were weaned from mechanical ventilation significantly faster than controls (median 29, 73, and 84 days, respectively; P < 0.05) and from supplemental oxygen (medians 65, 190, and 136 days, respectively; P < 0.05). No clinical complications of steroid administration were noted. At follow‐up, combining both dexamethasone groups revealed no significant differences between dexamethasone‐treated and control children for rates of cerebral palsy, blindness, deafness, developmental delay, or major neurosensory disability among survivors, or for death or survival with cerebral palsy, or for death or survival with major disability among those randomised. Neurological status was confirmed at four years of age for all children (Cummings 2002 (personal communication follow‐up of Cummings 1989)). Data show no significant differences in psychometric test scores at 15 months or 4 years of age. Between 4 and 15 years, one child in the 18‐day group had died, leaving 22 survivors, all of whom (100%) were assessed at 15 years of age. There were no significant differences between dexamethasone groups combined and the placebo group for any of the major neurological outcomes, nor for growth or respiratory function.

Doyle 2006: substantially more infants were extubated successfully by 10 days in the dexamethasone group than in the control group (odds ratio (OR) 11.2, 95% confidence interval (CI) 3.2 to 39.0; P < 0.001). Twelve of 21 dexamethasone‐treated infants were re‐intubated after initial extubation compared with one of four placebo‐treated infants. Mortality was reduced but not significantly in the dexamethasone group (OR 0.52, 95% CI 0.14 to 1.95; P = 0.33), and the same was true for BPD among survivors (OR 0.58, 95% CI 0.08 to 3.32; P = 0.71). Combined rates of death or BPD (86% versus 91%; P = 0.45) and death or severe BPD (34% versus 46%; P = 0.33) were not different between groups. During the first 10 days, mean airway pressure (MAP), peak inspiratory pressure, and inspired oxygen concentration all decreased significantly in the dexamethasone group compared with the placebo group. Data show no differences between groups in rates of high blood glucose levels or high blood pressure. Open‐label use of corticosteroids, sepsis, necrotising enterocolitis, patent ductus arteriosus, and severe retinopathy of prematurity were similar for the two groups. No infant had gastrointestinal perforation or bleeding. One infant in the placebo group had cardiac hypertrophy, but none in the dexamethasone group. At follow‐up, rates of cerebral palsy, blindness, and deafness, of Bayley MDI or PDI < ‐1 SD, or of major neurological disability were similar in the two groups, as were combined rates of death or cerebral palsy, or death or major disability.

Durand 1995: data show significant differences in compliance and tidal volume in the dexamethasone group compared with the control group (P < 0.001). Dexamethasone also significantly decreased inspired oxygen concentration and MAP (both P < 0.001) and facilitated successful weaning from mechanical ventilation. BPD (supplemental oxygen at 36 weeks' postmenstrual age, chest radiograph changes) was significantly decreased in the dexamethasone group (2/21 versus 8/17; P < 0.01). Survival with BPD was also better in the dexamethasone group (19/23 versus 9/20; P < 0.02). Except for transient increases in blood pressure and plasma glucose, we found no evidence of adverse effects of treatment and no significant differences in rates of infection, intraventricular haemorrhage, and retinopathy of prematurity. Thirteen infants in the control group subsequently received dexamethasone.

Harkavy 1989: dexamethasone treatment reduced age at extubation (39.4 days versus 57.2 days) compared with placebo. Average oxygen requirements for the steroid‐treated group were significantly lower during the first 10 days of treatment, but data show no significant differences between groups in age of weaning to room air (74.9 days versus 95.5 days), age at discharge (111 days versus 119 days), or number of deaths (1 (11%) versus 2 (17%)). Dexamethasone therapy was associated with a significantly increased incidence of hyperglycaemia (89% versus 8%; P = 0.01) but did not influence significantly the incidence of hypertension, intraventricular haemorrhage, infection, or retinopathy of prematurity. Steroid‐treated infants had a significant delay in weight gain (P < 0.02) during the first three weeks of treatment. Among the small number of children followed up, cerebral palsy was diagnosed in one of three in the dexamethasone group and in two of three controls.

Kari 1993: at 28 days of life, pulmonary outcome was significantly better among girls treated with dexamethasone but not in all infants. Data show no significant differences between groups in long‐term outcomes, except a shorter duration of supplemental oxygen among dexamethasone‐treated female infants. After one week of dexamethasone treatment, results show significant but short‐lived suppression of basal cortisol concentrations and of the adrenal response to ACTH. Investigators observed no serious side effects. At follow‐up, the only hospital providing follow‐up data reported no significant differences between dexamethasone and control children in rates of mortality, cerebral palsy, blindness, deafness, or major neurological disability, nor of death or survival with cerebral palsy or death or survival with major neurological disability, among those randomised. At seven to nine years of age, data show some improvement in lung function among eight steroid‐treated children compared with seven controls, and no substantial differences in height or weight between steroid and placebo groups, but data were not reported in a form that would allow meta‐analysis. No children had hypertrophic cardiomyopathy.

Kazzi 1990: infants who received dexamethasone required less oxygen on Days 8 and 17 (P < 0.005) and were more likely to be extubated eight days after therapy (8/12 versus 3/11; P < 0.05, P = 0.12 after Yates correction) compared with infants in the control group. Dexamethasone significantly shortened the duration of mechanical ventilation (median 4 versus 22 days; P < 0.05), but we found no evidence of effects on duration of oxygen therapy, hospitalisation, or home oxygen therapy, nor on the occurrence and severity of retinopathy of prematurity, rate of growth, or mortality.

Kothadia 1999: infants treated with dexamethasone were on mechanical ventilation and supplemental oxygen for fewer days after study entry (median days on ventilator: 5th and 95th centiles, 13 (1 to 64) versus 25 (6 to 104); days on oxygen: 59 (6 to 247) versus 100 (11 to 346)). Fewer infants in the dexamethasone group had failed to be extubated by the third day (82% versus 97%) or the seventh day (63% versus 90%). Data show no significant differences in rates of death, infection, or severe retinopathy of prematurity. At one‐year follow‐up, more surviving dexamethasone‐treated infants had cerebral palsy (24% versus 7%) and abnormal findings on neurological examination (42% versus 18%). However, deaths before one year were more frequent in the placebo group (26%) than in the dexamethasone group (12%); thus, rates of the combined outcome, death or cerebral palsy at one year, were not significantly different (dexamethasone 33% versus placebo 31%). An additional child in the placebo group was reported to have cerebral palsy at age four to six years. Risk of cerebral palsy was higher among surviving dexamethasone‐treated children at four to six years of age, although cognitive, functional, and medical outcomes were not significantly different between treated and non‐treated survivors. The combined outcome, death or cerebral palsy, was also similar at four‐ to six‐year follow‐up. Results show no substantial differences in rates of asthma, nor in blood pressure or growth. Fewer participants in the dexamethasone group had a low value for FEV₁ at between 8 and 11 years (dexamethasone 40% versus placebo 68%).

Kovacs 1998: mortality in hospital was not significantly different in the two groups (27% dexamethasone versus 17% controls). It is not possible to determine precisely when infants died, and hence this study cannot contribute to mortality at 28 days or at 36 weeks. Steroid‐treated infants required less ventilatory support between 9 and 17 days of age, and less supplemental oxygen between 8 and 10 days of age. Fewer infants in the dexamethasone group had failed to be extubated by the seventh day (73% versus 93%). Infants in this group also had better pulmonary compliance at 10 days, but comparison with controls revealed that all improvements were not maintained over ensuing weeks. Incidences of BPD at 28 days of life and at 36 weeks' postmenstrual age among survivors were not significantly different between groups (80% versus 87% at 28 days of life; 45% versus 56% at 36 weeks' postmenstrual age). We found no evidence of steroid‐related adverse effects, other than transient glycosuria. At follow‐up, data show no significant differences between dexamethasone‐treated and control children in rates of cerebral palsy, blindness, deafness, developmental delay, or major neurosensory disability among survivors assessed, nor in death or survival with cerebral palsy or death or survival with major disability among those randomised.

Noble‐Jamieson 1989: dexamethasone‐treated infants showed more rapid improvement in ventilation requirements during the first week of treatment, although the overall duration of oxygen therapy was similar in both groups. Cranial ultrasound examination revealed new periventricular abnormalities in three out of five dexamethasone‐treated infants with previous normal scans, compared with none of four placebo‐treated infants.

Ohlsson 1992: dexamethasone facilitated weaning from mechanical ventilation (P = 0.015). The incidence of infection was not significantly increased, although glycosuria (P = 0.0002) and systolic blood pressure (P = 0.003) were increased and heart rate (P = 0.0001) and weight gain (P = 0.0002) were decreased in the dexamethasone‐treated group. At follow‐up among survivors, cerebral palsy was diagnosed in one of 11 children in the dexamethasone group, and in three of 13 controls.

Onland 2019: hydrocortisone was associated with reduction in mortality at 36 weeks' postmenstrual age (15% versus 24%; P = 0.048) but a higher rate of BPD (55% versus 50%; P = 0.31) and no substantial effect on the primary endpoint of death or BPD (71% versus 74%; P = 0.54). Short‐term benefits included higher rates of successful extubation over the first 14 days of treatment and a reduction in treatment with open‐label steroids (28% versus 57%; P < 0.001), but adverse effects included more hyperglycaemia requiring insulin treatment (18% versus 8%; P = 0.004). At follow‐up, rates of neurodevelopmental impairment, cerebral palsy, and visual and hearing impairment were similar in the two groups, as were combined rates of death or cerebral palsy, or death or neurodevelopmental impairment.

Papile 1998: as infants in the early group were given dexamethasone from 14 days, they can be considered as having been treated late by our definition (> 7 days of age). Upon examination of only 28‐day outcomes, babies in this study's late group can be considered as controls, as they did not receive dexamethasone until after 28 days. Mortality at 28 days was 7/182 in the early group (treated) compared with 16/189 in the late group (controls). Oxygen was required on Day 28 in 141/182 versus 168/189, and the combination of 28‐day mortality or oxygen requirement was evident in 147/182 versus 184/189; the latter was significant (P < 0.001). It is not possible to use long‐term follow‐up data in this meta‐analysis, as all infants were eligible for dexamethasone after 28 days.

Parikh 2013: data show no substantial differences in brain tissue volume between groups. Low‐dose hydrocortisone had little effect on any other reported outcomes, including mortality, BPD, and acute complications. The follow‐up rate of survivors was 86% overall (37/43). Cerebral palsy was diagnosed in 15% (3/20) of survivors in the steroid group and in 6% (1/17) of those in the placebo group. Rates of cognitive and language delay, defined as < 80 on the Bayley III, were 21% versus 47% and 50% versus 59% in steroid and placebo groups, respectively. Rates of cognitive and language delay could not be pooled with others in the meta‐analysis that used earlier versions of the Bayley Scales, because tests and definitions were different. Rates of any neurodevelopmental impairment were similar between the two groups.

Romagnoli 1997: treated infants showed an increase in dynamic respiratory compliance and a decreased incidence of BPD at 28 days of life and at 36 weeks' postmenstrual age. Fewer infants in the dexamethasone group had failed to be extubated by the seventh day (40% versus 87%). Dexamethasone‐treated infants had lower weight gain during treatment and a significantly higher incidence of hypertrophic cardiomyopathy compared with controls. Data show no significant differences between groups regarding incidence of hypertension, sepsis, necrotising enterocolitis, or hyperglycaemia. At follow‐up, data show no significant differences between dexamethasone‐treated and control children for rates of cerebral palsy, blindness, deafness, or intellectual impairment among survivors assessed, or for death or survival with cerebral palsy among those randomised.

Scott 1997: cortisol responses to ACTH were lower in the dexamethasone group than in the placebo group. On Day 28 of life, eight of 10 infants in the dexamethasone group no longer required mechanical ventilation, compared with none of five infants in the placebo group (P = 0.04, as reported by trial authors).

Vento 2004: six dexamethasone‐treated infants and five control infants were extubated within seven days. Data show no significant differences between groups regarding respiratory distress syndrome, patent ductus arteriosus, or severe intraventricular haemorrhage (grade 3 or 4), as well as lower absolute cell counts (P ≤ 0.05) and proportions of polymorphonuclear cells (P < 0.001) in tracheal aspirate fluid in the treated group on Day 7. Treated infants also had an increase in dynamic pulmonary compliance, which was significant compared with the control group at seven days (P < 0.05). We noted no significant differences between groups regarding inspired oxygen concentration but found that infants in the dexamethasone group had significantly lower MAP on Day 7 (P < 0.05).

Vincer 1998: two of 11 dexamethasone‐treated infants died before hospital discharge compared with one of nine control infants. The number of days when infants had apnoeic spells (14 versus 2; P = 0.005) was greater in the dexamethasone‐treated group. Fewer infants in the dexamethasone group had failed to be extubated by the third day (27% versus 100%) or the seventh day (27% versus 100%). Data show a trend towards more retinopathy of prematurity in the dexamethasone group (64% versus 22%; P = 0.064) but similarities in all other outcome variables between groups. At follow‐up among survivors, cerebral palsy was diagnosed in four of nine children in the dexamethasone group and in two of eight controls.

Walther 2003: MAP on the first day of life was higher in the control group than in the dexamethasone group (9.1 versus 7.5 cm H₂O; P < 0.05). More infants in the dexamethasone group were successfully extubated within 7 to 14 days than in the placebo group (P < 0.05). Hyperglycaemia occurred more frequently in the dexamethasone group (P < 0.05), and infants in the control group more often received open‐label dexamethasone (P < 0.05). Incidences of hypertension, sepsis, necrotising enterocolitis, spontaneous gastrointestinal perforation, gastrointestinal bleeding, intraventricular haemorrhage, or periventricular leukomalacia were not significantly different between groups. Similarly, data show no significant differences in duration of ventilation or oxygen, BPD, nor mortality or survival without BPD between groups. Two infants in the control group were discharged home while on oxygen.

Yates 2019: two of the 12 infants in the dexamethasone group had received hydrocortisone prior to study entry, compared with none of 10 infants in the placebo group. In infants who remained in the study seven days after trial entry, five of eight dexamethasone‐treated infants were extubated by Day 7, compared with four of six control infants. Two of 12 dexamethasone‐treated infants died before 36 weeks' postmenstrual age compared with one of 10 control infants; 100% of survivors in both groups had BPD at 36 weeks' postmenstrual age or at discharge (if sooner) (dexamethasone 10/10; placebo 9/9).

Discussion

In this review, we found reductions in mortality at all ages (to 28 days after birth, to 36 weeks' corrected age, to hospital discharge, and at latest reported age) when systemic corticosteroids were started after seven days of age. Both dexamethasone and hydrocortisone contributed to the reduction in mortality, but evidence for either of them alone in reducing mortality was weaker than when they were combined. Systemic corticosteroids show other clear benefits after seven days of age in reducing rates of BPD at 28 days after birth and at 36 weeks' postmenstrual age, and in reducing the combined outcome of mortality or BPD at both time points; these benefits were attributable to dexamethasone ‐ not to hydrocortisone. Systemic corticosteroids also facilitated extubation at multiple time points. These benefits of corticosteroids came at the cost of higher rates of short‐term complications of hypertension, hyperglycaemia, glycosuria, and hypertrophic cardiomyopathy. An increase in severe retinopathy of prematurity was not accompanied by significant increases in blindness or in the need for corrective lenses at follow‐up.

Corticosteroids may have other significant effects. They can cause weight loss or poor weight gain (Ariagno 1987; Harkavy 1989; Ohlsson 1992). Although catchup growth after corticosteroid therapy has been reported (Gibson 1993), worries about reduced brain growth have been noted in animal studies (Gramsbergen 1998; Weichsel 1977), as well as in human studies (Papile 1998). Animal studies have shown abnormal lung growth (Tschanz 1995).

For this review, data on long‐term neurosensory follow‐up were available from 17 studies comprising 1348 randomised infants, but these studies were of varying methodological quality. The significant increase in abnormal neurological examination among those randomised is of potential concern; however, this is tempered by data showing that cerebral palsy and major neurosensory disability, both overall and among survivors, were not significantly increased, and that abnormal neurological examination findings were reported in only four of the 17 follow‐up studies, and among only 200 randomised participants. It should also be noted that some of the studies reporting cerebral palsy as an outcome did so when children were aged less than five years, an age when a diagnosis of cerebral palsy is not certain in all cases (Stanley 1982). Moreover, only one study was designed primarily to test effects of postnatal corticosteroids on adverse long‐term neurosensory outcomes; that study was terminated for futility with < 10% of the projected sample size recruited (Doyle 2006). All studies were underpowered to detect clinically important differences in long‐term neurosensory outcomes. Researchers performing animal studies have expressed concern about possible adverse effects of corticosteroids used at these doses during early postnatal life on the neurodevelopment of very immature infants (Weichsel 1977). Clearly, more information on long‐term outcomes of survivors is needed.

Clinicians must weigh the benefits of acute improvement in respiratory function and increased chances of extubation, along with improved survival, against potential detrimental effects, both metabolic and neurological. Dexamethasone may be a harmful drug for the immature brain, and clinicians must consider limiting its use to situations in which it is essential to achieve weaning from the ventilator. Lower doses and shorter courses should be considered for these infants; the DART study (an RCT of low‐dose, short‐course dexamethasone in ventilator‐dependent infants), which provided a total dose of only 0.89 mg/kg over 10 days, reported short‐term benefits of extubation and reduced respiratory support (Doyle 2006). Additional studies of low‐dose systemic corticosteroids for infants at high risk of developing BPD beyond the first week of life are warranted.

Summary of main results

In this review, we found evidence for reductions in mortality, BPD, and the combined outcome of mortality or BPD, without evidence for an effect on cerebral palsy, or the combined outcome of mortality or cerebral palsy. Whether systemic corticosteroids given starting from seven days of age improve survival free of long‐term neurodevelopmental disability among infants with evolving BPD remains to be confirmed.

Overall completeness and applicability of evidence

Data on in‐hospital outcomes were relatively complete, but data on longer‐term outcomes were incomplete. Results are applicable to ventilator‐dependent infants at high risk of developing BPD.

Quality of the evidence

Review authors assessed the certainty of evidence for five major outcomes: mortality at latest reported age, BPD at 36 weeks, mortality or BPD at 36 weeks, cerebral palsy at latest reported age, and mortality or cerebral palsy at latest reported age (summary of findings Table 1). We assessed the certainty of evidence for most outcomes as high, except for BPD at 36 weeks, which we downgraded to moderate quality because we noted risk of publication bias in these studies, and for the combined outcome of mortality or BPD at 36 weeks because of moderate heterogeneity, particularly among those treated with dexamethasone. Excluding two studies at higher risk of bias from a sensitivity analysis altered no conclusions.

Potential biases in the review process

Although Embase records are indexed in CENTRAL, we acknowledge that the omission of a search of Embase in 2020 may have reduced the sensitivity of our search.

Agreements and disagreements with other studies or reviews

Evidence showing benefit from systemic corticosteroids started from the age of seven days of age for reducing the rate of mortality among infants with evolving BPD is stronger than in earlier versions of this review (Doyle 2017b). Other main conclusions concerning BPD, extubation, and short‐term complications are consistent with earlier reviews.

Study flow diagram: review update.

Figures and Tables -
Figure 1

Study flow diagram: review update.

original image

Figures and Tables -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Figures and Tables -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Funnel plot of comparison: 1 Mortality, outcome: 1.4 Mortality at latest reported age.

Figures and Tables -
Figure 4

Funnel plot of comparison: 1 Mortality, outcome: 1.4 Mortality at latest reported age.

Funnel plot of comparison: 2 Bronchopulmonary dysplasia (BPD), outcome: 2.2 BPD at 36 weeks' postmenstrual age.

Figures and Tables -
Figure 5

Funnel plot of comparison: 2 Bronchopulmonary dysplasia (BPD), outcome: 2.2 BPD at 36 weeks' postmenstrual age.

Funnel plot of comparison: 3 Mortality or BPD, outcome: 3.2 Mortality or BPD at 36 weeks' postmenstrual age.

Figures and Tables -
Figure 6

Funnel plot of comparison: 3 Mortality or BPD, outcome: 3.2 Mortality or BPD at 36 weeks' postmenstrual age.

Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.10 Cerebral palsy at latest reported age.

Figures and Tables -
Figure 7

Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.10 Cerebral palsy at latest reported age.

Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.14 Mortality or cerebral palsy at latest reported age.

Figures and Tables -
Figure 8

Funnel plot of comparison: 6 Long‐term follow‐up, outcome: 6.14 Mortality or cerebral palsy at latest reported age.

Comparison 1: Mortality at different ages, Outcome 1: Neonatal mortality before 28 days after birth

Figures and Tables -
Analysis 1.1

Comparison 1: Mortality at different ages, Outcome 1: Neonatal mortality before 28 days after birth

Comparison 1: Mortality at different ages, Outcome 2: Mortality at 36 weeks' postmenstrual age

Figures and Tables -
Analysis 1.2

Comparison 1: Mortality at different ages, Outcome 2: Mortality at 36 weeks' postmenstrual age

Comparison 1: Mortality at different ages, Outcome 3: Mortality to hospital discharge

Figures and Tables -
Analysis 1.3

Comparison 1: Mortality at different ages, Outcome 3: Mortality to hospital discharge

Comparison 1: Mortality at different ages, Outcome 4: Mortality at latest reported age

Figures and Tables -
Analysis 1.4

Comparison 1: Mortality at different ages, Outcome 4: Mortality at latest reported age

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 1: BPD at 28 days after birth

Figures and Tables -
Analysis 2.1

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 1: BPD at 28 days after birth

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 2: BPD at 36 weeks' postmenstrual age

Figures and Tables -
Analysis 2.2

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 2: BPD at 36 weeks' postmenstrual age

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 3: BPD at 36 weeks in survivors

Figures and Tables -
Analysis 2.3

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 3: BPD at 36 weeks in survivors

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 4: Late rescue with corticosteroids

Figures and Tables -
Analysis 2.4

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 4: Late rescue with corticosteroids

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 5: Home on oxygen

Figures and Tables -
Analysis 2.5

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 5: Home on oxygen

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 6: Survivors discharged home on oxygen

Figures and Tables -
Analysis 2.6

Comparison 2: Bronchopulmonary dysplasia (BPD), Outcome 6: Survivors discharged home on oxygen

Comparison 3: Mortality or BPD, Outcome 1: Mortality or BPD at 28 days after birth

Figures and Tables -
Analysis 3.1

Comparison 3: Mortality or BPD, Outcome 1: Mortality or BPD at 28 days after birth

Comparison 3: Mortality or BPD, Outcome 2: Mortality or BPD at 36 weeks' postmenstrual age

Figures and Tables -
Analysis 3.2

Comparison 3: Mortality or BPD, Outcome 2: Mortality or BPD at 36 weeks' postmenstrual age

Comparison 4: Failure to extubate, Outcome 1: Failure to extubate by 3rd day after treatment

Figures and Tables -
Analysis 4.1

Comparison 4: Failure to extubate, Outcome 1: Failure to extubate by 3rd day after treatment

Comparison 4: Failure to extubate, Outcome 2: Failure to extubate by 7th day after treatment

Figures and Tables -
Analysis 4.2

Comparison 4: Failure to extubate, Outcome 2: Failure to extubate by 7th day after treatment

Comparison 4: Failure to extubate, Outcome 3: Failure to extubate by 14th day after treatment

Figures and Tables -
Analysis 4.3

Comparison 4: Failure to extubate, Outcome 3: Failure to extubate by 14th day after treatment

Comparison 4: Failure to extubate, Outcome 4: Failure to extubate by 28th day after treatment

Figures and Tables -
Analysis 4.4

Comparison 4: Failure to extubate, Outcome 4: Failure to extubate by 28th day after treatment

Comparison 5: Complications during primary hospitalisation, Outcome 1: Infection

Figures and Tables -
Analysis 5.1

Comparison 5: Complications during primary hospitalisation, Outcome 1: Infection

Comparison 5: Complications during primary hospitalisation, Outcome 2: Hyperglycaemia

Figures and Tables -
Analysis 5.2

Comparison 5: Complications during primary hospitalisation, Outcome 2: Hyperglycaemia

Comparison 5: Complications during primary hospitalisation, Outcome 3: Glycosuria

Figures and Tables -
Analysis 5.3

Comparison 5: Complications during primary hospitalisation, Outcome 3: Glycosuria

Comparison 5: Complications during primary hospitalisation, Outcome 4: Hypertension

Figures and Tables -
Analysis 5.4

Comparison 5: Complications during primary hospitalisation, Outcome 4: Hypertension

Comparison 5: Complications during primary hospitalisation, Outcome 5: New cranial echodensities

Figures and Tables -
Analysis 5.5

Comparison 5: Complications during primary hospitalisation, Outcome 5: New cranial echodensities

Comparison 5: Complications during primary hospitalisation, Outcome 6: Necrotising enterocolitis (NEC)

Figures and Tables -
Analysis 5.6

Comparison 5: Complications during primary hospitalisation, Outcome 6: Necrotising enterocolitis (NEC)

Comparison 5: Complications during primary hospitalisation, Outcome 7: Gastrointestinal bleeding

Figures and Tables -
Analysis 5.7

Comparison 5: Complications during primary hospitalisation, Outcome 7: Gastrointestinal bleeding

Comparison 5: Complications during primary hospitalisation, Outcome 8: Gastrointestinal perforation

Figures and Tables -
Analysis 5.8

Comparison 5: Complications during primary hospitalisation, Outcome 8: Gastrointestinal perforation

Comparison 5: Complications during primary hospitalisation, Outcome 9: Severe retinopathy of prematurity (ROP)

Figures and Tables -
Analysis 5.9

Comparison 5: Complications during primary hospitalisation, Outcome 9: Severe retinopathy of prematurity (ROP)

Comparison 5: Complications during primary hospitalisation, Outcome 10: Severe ROP in survivors

Figures and Tables -
Analysis 5.10

Comparison 5: Complications during primary hospitalisation, Outcome 10: Severe ROP in survivors

Comparison 5: Complications during primary hospitalisation, Outcome 11: Hypertrophic cardiomyopathy

Figures and Tables -
Analysis 5.11

Comparison 5: Complications during primary hospitalisation, Outcome 11: Hypertrophic cardiomyopathy

Comparison 5: Complications during primary hospitalisation, Outcome 12: Pneumothorax

Figures and Tables -
Analysis 5.12

Comparison 5: Complications during primary hospitalisation, Outcome 12: Pneumothorax

Comparison 5: Complications during primary hospitalisation, Outcome 13: Severe intraventricular haemorrhage (IVH)

Figures and Tables -
Analysis 5.13

Comparison 5: Complications during primary hospitalisation, Outcome 13: Severe intraventricular haemorrhage (IVH)

Comparison 5: Complications during primary hospitalisation, Outcome 14: Cystic periventricular leukomalacia

Figures and Tables -
Analysis 5.14

Comparison 5: Complications during primary hospitalisation, Outcome 14: Cystic periventricular leukomalacia

Comparison 6: Long‐term follow‐up, Outcome 1: Bayley Mental Developmental Index (MDI) < ‐2 SD

Figures and Tables -
Analysis 6.1

Comparison 6: Long‐term follow‐up, Outcome 1: Bayley Mental Developmental Index (MDI) < ‐2 SD

Comparison 6: Long‐term follow‐up, Outcome 2: Bayley MDI < ‐2 SD in survivors tested

Figures and Tables -
Analysis 6.2

Comparison 6: Long‐term follow‐up, Outcome 2: Bayley MDI < ‐2 SD in survivors tested

Comparison 6: Long‐term follow‐up, Outcome 3: Bayley Psychomotor Developmental Index (PDI) < ‐2 SD

Figures and Tables -
Analysis 6.3

Comparison 6: Long‐term follow‐up, Outcome 3: Bayley Psychomotor Developmental Index (PDI) < ‐2 SD

Comparison 6: Long‐term follow‐up, Outcome 4: Bayley PDI < ‐2 SD in survivors tested

Figures and Tables -
Analysis 6.4

Comparison 6: Long‐term follow‐up, Outcome 4: Bayley PDI < ‐2 SD in survivors tested

Comparison 6: Long‐term follow‐up, Outcome 5: Blindness

Figures and Tables -
Analysis 6.5

Comparison 6: Long‐term follow‐up, Outcome 5: Blindness

Comparison 6: Long‐term follow‐up, Outcome 6: Blindness in survivors assessed

Figures and Tables -
Analysis 6.6

Comparison 6: Long‐term follow‐up, Outcome 6: Blindness in survivors assessed

Comparison 6: Long‐term follow‐up, Outcome 7: Deafness

Figures and Tables -
Analysis 6.7

Comparison 6: Long‐term follow‐up, Outcome 7: Deafness

Comparison 6: Long‐term follow‐up, Outcome 8: Deafness in survivors assessed

Figures and Tables -
Analysis 6.8

Comparison 6: Long‐term follow‐up, Outcome 8: Deafness in survivors assessed

Comparison 6: Long‐term follow‐up, Outcome 9: Cerebral palsy at 1 to 3 years of age

Figures and Tables -
Analysis 6.9

Comparison 6: Long‐term follow‐up, Outcome 9: Cerebral palsy at 1 to 3 years of age

Comparison 6: Long‐term follow‐up, Outcome 10: Cerebral palsy at latest reported age

Figures and Tables -
Analysis 6.10

Comparison 6: Long‐term follow‐up, Outcome 10: Cerebral palsy at latest reported age

Comparison 6: Long‐term follow‐up, Outcome 11: Mortality before follow‐up in trials assessing cerebral palsy at 1‐3 years of age

Figures and Tables -
Analysis 6.11

Comparison 6: Long‐term follow‐up, Outcome 11: Mortality before follow‐up in trials assessing cerebral palsy at 1‐3 years of age

Comparison 6: Long‐term follow‐up, Outcome 12: Mortality before follow‐up in trials assessing cerebral palsy at latest reported age

Figures and Tables -
Analysis 6.12

Comparison 6: Long‐term follow‐up, Outcome 12: Mortality before follow‐up in trials assessing cerebral palsy at latest reported age

Comparison 6: Long‐term follow‐up, Outcome 13: Mortality or cerebral palsy at 1 to 3 years

Figures and Tables -
Analysis 6.13

Comparison 6: Long‐term follow‐up, Outcome 13: Mortality or cerebral palsy at 1 to 3 years

Comparison 6: Long‐term follow‐up, Outcome 14: Mortality or cerebral palsy at latest reported age

Figures and Tables -
Analysis 6.14

Comparison 6: Long‐term follow‐up, Outcome 14: Mortality or cerebral palsy at latest reported age

Comparison 6: Long‐term follow‐up, Outcome 15: Cerebral palsy in survivors assessed at 1‐3 years of age

Figures and Tables -
Analysis 6.15

Comparison 6: Long‐term follow‐up, Outcome 15: Cerebral palsy in survivors assessed at 1‐3 years of age

Comparison 6: Long‐term follow‐up, Outcome 16: Cerebral palsy in survivors assessed at latest age

Figures and Tables -
Analysis 6.16

Comparison 6: Long‐term follow‐up, Outcome 16: Cerebral palsy in survivors assessed at latest age

Comparison 6: Long‐term follow‐up, Outcome 17: Major neurosensory disability (variable criteria ‐ see individual studies)

Figures and Tables -
Analysis 6.17

Comparison 6: Long‐term follow‐up, Outcome 17: Major neurosensory disability (variable criteria ‐ see individual studies)

Comparison 6: Long‐term follow‐up, Outcome 18: Mortality before follow‐up in trials assessing major neurosensory disability (variable criteria)

Figures and Tables -
Analysis 6.18

Comparison 6: Long‐term follow‐up, Outcome 18: Mortality before follow‐up in trials assessing major neurosensory disability (variable criteria)

Comparison 6: Long‐term follow‐up, Outcome 19: Mortality or major neurosensory disability (variable criteria)

Figures and Tables -
Analysis 6.19

Comparison 6: Long‐term follow‐up, Outcome 19: Mortality or major neurosensory disability (variable criteria)

Comparison 6: Long‐term follow‐up, Outcome 20: Major neurosensory disability (variable criteria) in survivors assessed

Figures and Tables -
Analysis 6.20

Comparison 6: Long‐term follow‐up, Outcome 20: Major neurosensory disability (variable criteria) in survivors assessed

Comparison 6: Long‐term follow‐up, Outcome 21: Abnormal neurological exam (variable criteria ‐ see individual studies)

Figures and Tables -
Analysis 6.21

Comparison 6: Long‐term follow‐up, Outcome 21: Abnormal neurological exam (variable criteria ‐ see individual studies)

Comparison 6: Long‐term follow‐up, Outcome 22: Mortality before follow‐up in trials assessing abnormal neurological exam (variable criteria)

Figures and Tables -
Analysis 6.22

Comparison 6: Long‐term follow‐up, Outcome 22: Mortality before follow‐up in trials assessing abnormal neurological exam (variable criteria)

Comparison 6: Long‐term follow‐up, Outcome 23: Mortality or abnormal neurological exam (variable criteria)

Figures and Tables -
Analysis 6.23

Comparison 6: Long‐term follow‐up, Outcome 23: Mortality or abnormal neurological exam (variable criteria)

Comparison 6: Long‐term follow‐up, Outcome 24: Abnormal neurological exam (variable criteria) in survivors assessed

Figures and Tables -
Analysis 6.24

Comparison 6: Long‐term follow‐up, Outcome 24: Abnormal neurological exam (variable criteria) in survivors assessed

Comparison 6: Long‐term follow‐up, Outcome 25: Re‐hospitalisation

Figures and Tables -
Analysis 6.25

Comparison 6: Long‐term follow‐up, Outcome 25: Re‐hospitalisation

Comparison 6: Long‐term follow‐up, Outcome 26: Re‐hospitalisation in survivors seen at follow‐up

Figures and Tables -
Analysis 6.26

Comparison 6: Long‐term follow‐up, Outcome 26: Re‐hospitalisation in survivors seen at follow‐up

Comparison 7: Later childhood outcomes, Outcome 1: Recurrent wheezing in survivors examined at 5 years

Figures and Tables -
Analysis 7.1

Comparison 7: Later childhood outcomes, Outcome 1: Recurrent wheezing in survivors examined at 5 years

Comparison 7: Later childhood outcomes, Outcome 2: Use of corrective lenses in survivors examined at 5 years

Figures and Tables -
Analysis 7.2

Comparison 7: Later childhood outcomes, Outcome 2: Use of corrective lenses in survivors examined at 5 years

Comparison 7: Later childhood outcomes, Outcome 3: Use of physical therapy in survivors examined at 5 years

Figures and Tables -
Analysis 7.3

Comparison 7: Later childhood outcomes, Outcome 3: Use of physical therapy in survivors examined at 5 years

Comparison 7: Later childhood outcomes, Outcome 4: Use of speech therapy in survivors examined at 5 years

Figures and Tables -
Analysis 7.4

Comparison 7: Later childhood outcomes, Outcome 4: Use of speech therapy in survivors examined at 5 years

Comparison 7: Later childhood outcomes, Outcome 5: Intellectual impairment in survivors tested at 5 or more years

Figures and Tables -
Analysis 7.5

Comparison 7: Later childhood outcomes, Outcome 5: Intellectual impairment in survivors tested at 5 or more years

Comparison 7: Later childhood outcomes, Outcome 6: IQ

Figures and Tables -
Analysis 7.6

Comparison 7: Later childhood outcomes, Outcome 6: IQ

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 1: Asthma in survivors assessed

Figures and Tables -
Analysis 8.1

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 1: Asthma in survivors assessed

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 2: Forced expired volume in 1 second < ‐2 SD

Figures and Tables -
Analysis 8.2

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 2: Forced expired volume in 1 second < ‐2 SD

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 3: Forced expired volume in 1 second ‐ z‐score

Figures and Tables -
Analysis 8.3

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 3: Forced expired volume in 1 second ‐ z‐score

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 4: Forced expired volume in 1 second ‐ % predicted

Figures and Tables -
Analysis 8.4

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 4: Forced expired volume in 1 second ‐ % predicted

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 5: Forced expired volume in 1 second ‐ standardised mean difference

Figures and Tables -
Analysis 8.5

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 5: Forced expired volume in 1 second ‐ standardised mean difference

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 6: Forced vital capacity ‐ z‐score

Figures and Tables -
Analysis 8.6

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 6: Forced vital capacity ‐ z‐score

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 7: Forced vital capacity ‐ % predicted

Figures and Tables -
Analysis 8.7

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 7: Forced vital capacity ‐ % predicted

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 8: Forced vital capacity ‐ standardised mean difference

Figures and Tables -
Analysis 8.8

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 8: Forced vital capacity ‐ standardised mean difference

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 9: FEV₁/FVC %

Figures and Tables -
Analysis 8.9

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 9: FEV₁/FVC %

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 10: FEV₁/FVC < ‐2 SD

Figures and Tables -
Analysis 8.10

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 10: FEV₁/FVC < ‐2 SD

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 11: FEF25% -75% ‐ % predicted

Figures and Tables -
Analysis 8.11

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 11: FEF25% -75% ‐ % predicted

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 12: Positive bronchodilator response

Figures and Tables -
Analysis 8.12

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 12: Positive bronchodilator response

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 13: Forced vital capacity < ‐2 SD

Figures and Tables -
Analysis 8.13

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 13: Forced vital capacity < ‐2 SD

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 14: Exercise‐induced bronchoconstriction

Figures and Tables -
Analysis 8.14

Comparison 8: Respiratory outcomes in childhood ‐ after 5 years, Outcome 14: Exercise‐induced bronchoconstriction

Comparison 9: Growth in childhood, Outcome 1: Height ‐ z‐score

Figures and Tables -
Analysis 9.1

Comparison 9: Growth in childhood, Outcome 1: Height ‐ z‐score

Comparison 9: Growth in childhood, Outcome 2: Height < ‐2 SD

Figures and Tables -
Analysis 9.2

Comparison 9: Growth in childhood, Outcome 2: Height < ‐2 SD

Comparison 9: Growth in childhood, Outcome 3: Weight ‐ z‐score

Figures and Tables -
Analysis 9.3

Comparison 9: Growth in childhood, Outcome 3: Weight ‐ z‐score

Comparison 9: Growth in childhood, Outcome 4: Weight < ‐2 SD

Figures and Tables -
Analysis 9.4

Comparison 9: Growth in childhood, Outcome 4: Weight < ‐2 SD

Comparison 9: Growth in childhood, Outcome 5: Body mass index (BMI) ‐ z‐score

Figures and Tables -
Analysis 9.5

Comparison 9: Growth in childhood, Outcome 5: Body mass index (BMI) ‐ z‐score

Comparison 9: Growth in childhood, Outcome 6: BMI < ‐2 SD

Figures and Tables -
Analysis 9.6

Comparison 9: Growth in childhood, Outcome 6: BMI < ‐2 SD

Comparison 10: Blood pressure in childhood, Outcome 1: Systolic blood pressure > 95th centile

Figures and Tables -
Analysis 10.1

Comparison 10: Blood pressure in childhood, Outcome 1: Systolic blood pressure > 95th centile

Comparison 10: Blood pressure in childhood, Outcome 2: Systolic blood pressure z‐score

Figures and Tables -
Analysis 10.2

Comparison 10: Blood pressure in childhood, Outcome 2: Systolic blood pressure z‐score

Comparison 10: Blood pressure in childhood, Outcome 3: Diastolic blood pressure > 95th centile

Figures and Tables -
Analysis 10.3

Comparison 10: Blood pressure in childhood, Outcome 3: Diastolic blood pressure > 95th centile

Comparison 10: Blood pressure in childhood, Outcome 4: Diastolic blood pressure z‐score

Figures and Tables -
Analysis 10.4

Comparison 10: Blood pressure in childhood, Outcome 4: Diastolic blood pressure z‐score

Summary of findings 1. Systemic corticosteroids (dexamethasone or hydrocortisone) compared with control (placebo or nothing) for chronic lung disease in preterm infants

Systemic corticosteroids (dexamethasone or hydrocortisone) compared with control (placebo or nothing) for chronic lung disease in preterm infants

Patient or population: preterm infants with chronic lung disease
Setting: multiple neonatal intensive care units from high‐income countries
Intervention: systemic corticosteroids (dexamethasone or hydrocortisone)
Comparison: control (placebo or nothing)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No. of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with control (placebo or nothing)

Risk with systemic corticosteroids (dexamethasone or hydrocortisone)

Mortality at latest reported age

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.81
(0.66 to 0.99)

1428
(21 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

228 per 1000

185 per 1000
(151 to 226)

Study population (subgroup of studies treating with dexamethasone)

RR 0.85
(0.66 to 1.11)

993
(19 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

193 per 1000

164 per 1000
(128 to 215)

Study population (subgroup of studies treating with hydrocortisone)

RR 0.74
(0.54 to 1.02)

435
(2 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

305 per 1000

226 per 1000
(165 to 311)

BPD at 36 weeks' PMA

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.89
(0.80 to 0.99)

988
(14 RCTs)

⊕⊕⊕⊝
MODERATEa

Strong evidence for subgroup differences (interaction P < 0.001)

594 per 1000

529 per 1000
(475 to 588)

Study population (subgroup of studies treating with dexamethasone)

RR 0.76
(0.66 to 0.87)

553
(12 RCTs)

⊕⊕⊕⊝
MODERATEa

659 per 1000

501 per 1000
(435 to 573)

Study population (subgroup of studies treating with hydrocortisone)

RR 1.10
(0.92 to 1.31)

435
(2 RCTs)

⊕⊕⊕⊕
HIGH

516 per 1000

567 per 1000
(474 to 676)

Mortality or BPD at 36 weeks' PMA

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.85
(0.79 to 0.92)

988
(14 RCTs)

⊕⊕⊕⊝
MODERATEb

Strong evidence for subgroup differences (interaction P < 0.001)

771 per 1000

656 per 1000
(609  to 710)

Study population (subgroup of studies treating with dexamethasone)

RR 0.75
(0.67 to 0.84)

553
(12 RCTs)

⊕⊕⊕⊝
MODERATEb

787 per 1000

590 per 1000
(527to 661)

Study population (subgroup of studies treating with hydrocortisone)

RR 0.98
(0.88 to 1.09)

435
(2 RCTs)

⊕⊕⊕⊕
HIGH

753 per 1000

738 per 1000
(663 to 832)

Cerebral palsy ‐ at latest reported age

Study population (studies treating with dexamethasone or hydrocortisone)

RR 1.17
(0.84 to 1.61)

1290
(17 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome.

93 per 1000

109 per 1000
(78 to 150)

Study population (subgroup of studies treating with dexamethasone)

RR 1.17
(0.84 to 1.61

1290
(17 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

121 per 1000

135 per 1000 (95 to 193)

Study population (subgroup of studies treating with hydrocortisone)

RR 1.40

(0.60 to 3.26)

435 (2 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

40 per 1000

57 per 1000

Mortality or cerebral palsy ‐ at latest reported age

Study population (studies treating with dexamethasone or hydrocortisone)

RR 0.90
(0.76 to 1.06)

1290
(17 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome.

324 per 1000

291 per 1000
(246 to 343)

Study population (subgroup of studies treating with dexamethasone)

RR 0.95 (0.77 to 1.16)

855 (15 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

312 per 1000

296 per 1000 (240 to 362)

Study population (subgroup of studies treating with hydrocortisone)

RR 0.82 (0.62 to 1.08)

435 (2 RCTs)

⊕⊕⊕⊕
HIGH

Critical outcome

345 per 1000

283 per 1000

*The risk in the intervention group (and its 95% confidence interval) is based on assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

BPD: bronchopulmonary dysplasia; CI: confidence interval; PMA: postmenstrual age; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.

High certainty: further research is very unlikely to change our confidence in the estimate of effect.

Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.

Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.

Very low certainty: we are very uncertain about the estimate.

aDowngraded one level because publication bias was suspected.

bDowngraded one level for moderate heterogeneity.

Figures and Tables -
Summary of findings 1. Systemic corticosteroids (dexamethasone or hydrocortisone) compared with control (placebo or nothing) for chronic lung disease in preterm infants
Comparison 1. Mortality at different ages

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Neonatal mortality before 28 days after birth Show forest plot

7

970

Risk Ratio (M‐H, Fixed, 95% CI)

0.60 [0.39, 0.92]

1.1.1 Dexamethasone

6

599

Risk Ratio (M‐H, Fixed, 95% CI)

0.46 [0.24, 0.86]

1.1.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.43, 1.40]

1.2 Mortality at 36 weeks' postmenstrual age Show forest plot

15

1029

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.52, 0.94]

1.2.1 Dexamethasone

13

594

Risk Ratio (M‐H, Fixed, 95% CI)

0.68 [0.43, 1.08]

1.2.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.49, 1.04]

1.3 Mortality to hospital discharge Show forest plot

20

1406

Risk Ratio (M‐H, Fixed, 95% CI)

0.79 [0.63, 0.98]

1.3.1 Dexamethasone

18

971

Risk Ratio (M‐H, Fixed, 95% CI)

0.82 [0.62, 1.10]

1.3.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.53, 1.03]

1.4 Mortality at latest reported age Show forest plot

21

1428

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.66, 0.99]

1.4.1 Dexamethasone

19

993

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.66, 1.11]

1.4.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.54, 1.02]

Figures and Tables -
Comparison 1. Mortality at different ages
Comparison 2. Bronchopulmonary dysplasia (BPD)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 BPD at 28 days after birth Show forest plot

7

994

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.84, 0.95]

2.1.1 Dexamethasone

6

623

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.81, 0.94]

2.1.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.83, 1.04]

2.2 BPD at 36 weeks' postmenstrual age Show forest plot

14

988

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.80, 0.99]

2.2.1 Dexamethasone

12

553

Risk Ratio (M‐H, Fixed, 95% CI)

0.76 [0.66, 0.87]

2.2.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.92, 1.31]

2.3 BPD at 36 weeks in survivors Show forest plot

9

624

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.82, 1.01]

2.3.1 Dexamethasone

7

278

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.69, 0.93]

2.3.2 Hydrocortisone

2

346

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.88, 1.17]

2.4 Late rescue with corticosteroids Show forest plot

15

1489

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.41, 0.57]

2.4.1 Dexamethasone

13

1054

Risk Ratio (M‐H, Fixed, 95% CI)

0.46 [0.37, 0.57]

2.4.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.52 [0.40, 0.67]

2.5 Home on oxygen Show forest plot

7

611

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.54, 0.94]

2.6 Survivors discharged home on oxygen Show forest plot

6

277

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.51, 0.94]

Figures and Tables -
Comparison 2. Bronchopulmonary dysplasia (BPD)
Comparison 3. Mortality or BPD

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 Mortality or BPD at 28 days after birth Show forest plot

6

934

Risk Ratio (M‐H, Fixed, 95% CI)

0.87 [0.83, 0.91]

3.1.1 Dexamethasone

5

563

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.79, 0.90]

3.1.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.84, 0.98]

3.2 Mortality or BPD at 36 weeks' postmenstrual age Show forest plot

14

988

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.79, 0.92]

3.2.1 Dexamethasone

12

553

Risk Ratio (M‐H, Fixed, 95% CI)

0.75 [0.67, 0.84]

3.2.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.88, 1.09]

Figures and Tables -
Comparison 3. Mortality or BPD
Comparison 4. Failure to extubate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Failure to extubate by 3rd day after treatment Show forest plot

10

764

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.78, 0.88]

4.1.1 Dexamethasone

9

408

Risk Ratio (M‐H, Fixed, 95% CI)

0.76 [0.69, 0.84]

4.1.2 Hydrocortisone

1

356

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.84, 0.98]

4.2 Failure to extubate by 7th day after treatment Show forest plot

17

1130

Risk Ratio (M‐H, Fixed, 95% CI)

0.67 [0.62, 0.73]

4.2.1 Dexamethasone

16

783

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.60, 0.73]

4.2.2 Hydrocortisone

1

347

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.60, 0.82]

4.3 Failure to extubate by 14th day after treatment Show forest plot

5

458

Risk Ratio (M‐H, Fixed, 95% CI)

0.65 [0.53, 0.80]

4.3.1 Dexamethasone

4

124

Risk Ratio (M‐H, Fixed, 95% CI)

0.63 [0.45, 0.90]

4.3.2 Hydrocortisone

1

334

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.51, 0.85]

4.4 Failure to extubate by 28th day after treatment Show forest plot

3

236

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.37, 0.89]

Figures and Tables -
Comparison 4. Failure to extubate
Comparison 5. Complications during primary hospitalisation

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Infection Show forest plot

20

1742

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.91, 1.16]

5.1.1 Dexamethasone

18

1307

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.96, 1.35]

5.1.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.87 [0.73, 1.04]

5.2 Hyperglycaemia Show forest plot

19

1684

Risk Ratio (M‐H, Fixed, 95% CI)

1.59 [1.34, 1.89]

5.2.1 Dexamethasone

17

1249

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [1.26, 1.85]

5.2.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

1.85 [1.23, 2.77]

5.3 Glycosuria Show forest plot

2

48

Risk Ratio (M‐H, Fixed, 95% CI)

8.03 [2.43, 26.52]

5.4 Hypertension Show forest plot

17

1628

Risk Ratio (M‐H, Fixed, 95% CI)

1.67 [1.19, 2.33]

5.4.1 Dexamethasone

15

1193

Risk Ratio (M‐H, Fixed, 95% CI)

2.45 [1.48, 4.06]

5.4.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.70, 1.73]

5.5 New cranial echodensities Show forest plot

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

7.00 [0.41, 118.69]

5.6 Necrotising enterocolitis (NEC) Show forest plot

11

1409

Risk Ratio (M‐H, Fixed, 95% CI)

0.92 [0.62, 1.38]

5.6.1 Dexamethasone

9

974

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.54, 1.63]

5.6.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.51, 1.63]

5.7 Gastrointestinal bleeding Show forest plot

9

1385

Risk Ratio (M‐H, Fixed, 95% CI)

1.33 [0.97, 1.83]

5.7.1 Dexamethasone

8

1014

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [0.99, 1.93]

5.7.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.40, 2.74]

5.8 Gastrointestinal perforation Show forest plot

5

552

Risk Ratio (M‐H, Fixed, 95% CI)

0.67 [0.26, 1.70]

5.8.1 Dexamethasone

3

117

Risk Ratio (M‐H, Fixed, 95% CI)

0.36 [0.02, 8.05]

5.8.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.72 [0.27, 1.92]

5.9 Severe retinopathy of prematurity (ROP) Show forest plot

13

929

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [1.03, 1.58]

5.9.1 Dexamethasone

12

558

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [1.07, 1.79]

5.9.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.76, 1.59]

5.10 Severe ROP in survivors Show forest plot

10

697

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.94, 1.45]

5.10.1 Dexamethasone

9

416

Risk Ratio (M‐H, Fixed, 95% CI)

1.31 [0.99, 1.74]

5.10.2 Hydrocortisone

1

281

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.69, 1.40]

5.11 Hypertrophic cardiomyopathy Show forest plot

4

238

Risk Ratio (M‐H, Fixed, 95% CI)

2.76 [1.33, 5.74]

5.12 Pneumothorax Show forest plot

3

157

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.53, 1.49]

5.13 Severe intraventricular haemorrhage (IVH) Show forest plot

7

639

Risk Ratio (M‐H, Fixed, 95% CI)

0.54 [0.26, 1.11]

5.13.1 Dexamethasone

6

268

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.23, 1.13]

5.13.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.12, 4.14]

5.14 Cystic periventricular leukomalacia Show forest plot

2

392

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.30, 1.84]

5.14.1 Dexamethasone

1

21

Risk Ratio (M‐H, Fixed, 95% CI)

0.31 [0.01, 6.74]

5.14.2 Hydrocortisone

1

371

Risk Ratio (M‐H, Fixed, 95% CI)

0.82 [0.31, 2.15]

Figures and Tables -
Comparison 5. Complications during primary hospitalisation
Comparison 6. Long‐term follow‐up

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Bayley Mental Developmental Index (MDI) < ‐2 SD Show forest plot

7

333

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.47, 1.38]

6.2 Bayley MDI < ‐2 SD in survivors tested Show forest plot

7

232

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.45, 1.22]

6.3 Bayley Psychomotor Developmental Index (PDI) < ‐2 SD Show forest plot

1

118

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.34, 1.80]

6.4 Bayley PDI < ‐2 SD in survivors tested Show forest plot

1

90

Risk Ratio (M‐H, Fixed, 95% CI)

0.67 [0.30, 1.50]

6.5 Blindness Show forest plot

13

784

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.35, 1.73]

6.5.1 Dexamethasone

12

720

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.35, 1.73]

6.5.2 Hydrocortisone

1

64

Risk Ratio (M‐H, Fixed, 95% CI)

Not estimable

6.6 Blindness in survivors assessed Show forest plot

13

539

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.35, 1.67]

6.6.1 Dexamethasone

12

502

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.35, 1.67]

6.6.2 Hydrocortisone

1

37

Risk Ratio (M‐H, Fixed, 95% CI)

Not estimable

6.7 Deafness Show forest plot

9

936

Risk Ratio (M‐H, Fixed, 95% CI)

0.57 [0.26, 1.27]

6.7.1 Dexamethasone

7

501

Risk Ratio (M‐H, Fixed, 95% CI)

0.56 [0.22, 1.44]

6.7.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.59 [0.13, 2.71]

6.8 Deafness in survivors assessed Show forest plot

9

616

Risk Ratio (M‐H, Fixed, 95% CI)

0.62 [0.29, 1.36]

6.8.1 Dexamethasonep

7

325

Risk Ratio (M‐H, Fixed, 95% CI)

0.67 [0.27, 1.66]

6.8.2 Hydrocortisone

2

291

Risk Ratio (M‐H, Fixed, 95% CI)

0.52 [0.11, 2.36]

6.9 Cerebral palsy at 1 to 3 years of age Show forest plot

16

1311

Risk Ratio (M‐H, Fixed, 95% CI)

1.11 [0.81, 1.52]

6.9.1 Dexamethasone

14

876

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.76, 1.50]

6.9.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [0.60, 3.26]

6.10 Cerebral palsy at latest reported age Show forest plot

17

1290

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.84, 1.61]

6.10.1 Dexamethasone

15

855

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.79, 1.60]

6.10.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [0.60, 3.26]

6.11 Mortality before follow‐up in trials assessing cerebral palsy at 1‐3 years of age Show forest plot

16

1746

Risk Ratio (M‐H, Fixed, 95% CI)

0.78 [0.66, 0.93]

6.11.1 Dexamethasone

16

1311

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.65, 0.99]

6.11.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.54, 1.02]

6.12 Mortality before follow‐up in trials assessing cerebral palsy at latest reported age Show forest plot

17

1290

Odds Ratio (M‐H, Fixed, 95% CI)

0.73 [0.56, 0.97]

6.12.1 Dexamethasone

15

855

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.55, 1.12]

6.12.2 Hydrocortisone

2

435

Odds Ratio (M‐H, Fixed, 95% CI)

0.67 [0.43, 1.03]

6.13 Mortality or cerebral palsy at 1 to 3 years Show forest plot

16

1311

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.76, 1.04]

6.13.1 Dexamethasone

14

876

Risk Ratio (M‐H, Fixed, 95% CI)

0.92 [0.76, 1.12]

6.13.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.82 [0.62, 1.08]

6.14 Mortality or cerebral palsy at latest reported age Show forest plot

17

1290

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.76, 1.06]

6.14.1 Dexamethasone

15

855

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.77, 1.16]

6.14.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.82 [0.62, 1.08]

6.15 Cerebral palsy in survivors assessed at 1‐3 years of age Show forest plot

16

923

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.79, 1.47]

6.15.1 Dexamethasone

14

631

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.75, 1.47]

6.15.2 Hydrocortisone

2

292

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [0.55, 2.93]

6.16 Cerebral palsy in survivors assessed at latest age Show forest plot

17

883

Odds Ratio (M‐H, Fixed, 95% CI)

1.17 [0.80, 1.71]

6.16.1 Dexamethasone

15

591

Odds Ratio (M‐H, Fixed, 95% CI)

1.14 [0.75, 1.74]

6.16.2 Hydrocortisone

2

292

Odds Ratio (M‐H, Fixed, 95% CI)

1.29 [0.53, 3.17]

6.17 Major neurosensory disability (variable criteria ‐ see individual studies) Show forest plot

10

1090

Risk Ratio (M‐H, Fixed, 95% CI)

1.09 [0.88, 1.34]

6.17.1 Dexamethasone

8

655

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.85, 1.60]

6.17.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.78, 1.35]

6.18 Mortality before follow‐up in trials assessing major neurosensory disability (variable criteria) Show forest plot

10

1090

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.64, 1.00]

6.18.1 Dexamethasone

8

655

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.63, 1.15]

6.18.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.54, 1.02]

6.19 Mortality or major neurosensory disability (variable criteria) Show forest plot

10

1090

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.85, 1.08]

6.19.1 Dexamethasone

8

655

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.86, 1.26]

6.19.2 Hydrocortisone

2

435

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.75, 1.04]

6.20 Major neurosensory disability (variable criteria) in survivors assessed Show forest plot

10

778

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.83, 1.22]

6.20.1 Dexamethasone

8

480

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.81, 1.50]

6.20.2 Hydrocortisone

2

298

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.73, 1.19]

6.21 Abnormal neurological exam (variable criteria ‐ see individual studies) Show forest plot

4

200

Risk Ratio (M‐H, Fixed, 95% CI)

1.81 [1.05, 3.11]

6.22 Mortality before follow‐up in trials assessing abnormal neurological exam (variable criteria) Show forest plot

4

200

Risk Ratio (M‐H, Fixed, 95% CI)

0.57 [0.33, 0.99]

6.23 Mortality or abnormal neurological exam (variable criteria) Show forest plot

4

200

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.71, 1.31]

6.24 Abnormal neurological exam (variable criteria) in survivors assessed Show forest plot

4

145

Risk Ratio (M‐H, Fixed, 95% CI)

1.62 [0.96, 2.73]

6.25 Re‐hospitalisation Show forest plot

1

118

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.79, 1.66]

6.26 Re‐hospitalisation in survivors seen at follow‐up Show forest plot

1

92

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.72, 1.34]

Figures and Tables -
Comparison 6. Long‐term follow‐up
Comparison 7. Later childhood outcomes

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

7.1 Recurrent wheezing in survivors examined at 5 years Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.47 [0.82, 2.64]

7.2 Use of corrective lenses in survivors examined at 5 years Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.61 [0.82, 3.13]

7.3 Use of physical therapy in survivors examined at 5 years Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

1.49 [0.71, 3.11]

7.4 Use of speech therapy in survivors examined at 5 years Show forest plot

1

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.46 [0.21, 1.02]

7.5 Intellectual impairment in survivors tested at 5 or more years Show forest plot

3

254

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.71, 1.52]

7.6 IQ Show forest plot

2

92

Std. Mean Difference (IV, Fixed, 95% CI)

0.08 [‐0.35, 0.51]

Figures and Tables -
Comparison 7. Later childhood outcomes
Comparison 8. Respiratory outcomes in childhood ‐ after 5 years

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

8.1 Asthma in survivors assessed Show forest plot

2

213

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.44, 1.16]

8.2 Forced expired volume in 1 second < ‐2 SD Show forest plot

2

187

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.36, 0.94]

8.3 Forced expired volume in 1 second ‐ z‐score Show forest plot

1

124

Mean Difference (IV, Fixed, 95% CI)

0.28 [‐0.14, 0.70]

8.4 Forced expired volume in 1 second ‐ % predicted Show forest plot

3

98

Mean Difference (IV, Fixed, 95% CI)

5.87 [‐1.26, 13.00]

8.5 Forced expired volume in 1 second ‐ standardised mean difference Show forest plot

4

222

Std. Mean Difference (IV, Fixed, 95% CI)

0.29 [0.02, 0.56]

8.6 Forced vital capacity ‐ z‐score Show forest plot

1

120

Mean Difference (IV, Fixed, 95% CI)

0.09 [‐0.31, 0.49]

8.7 Forced vital capacity ‐ % predicted Show forest plot

3

98

Mean Difference (IV, Fixed, 95% CI)

7.77 [1.79, 13.74]

8.8 Forced vital capacity ‐ standardised mean difference Show forest plot

4

218

Std. Mean Difference (IV, Fixed, 95% CI)

0.25 [‐0.02, 0.52]

8.9 FEV₁/FVC % Show forest plot

1

63

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐3.70, 5.70]

8.10 FEV₁/FVC < ‐2 SD Show forest plot

1

63

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.44, 1.77]

8.11 FEF25% -75% ‐ % predicted Show forest plot

1

63

Mean Difference (IV, Fixed, 95% CI)

7.00 [‐5.40, 19.40]

8.12 Positive bronchodilator response Show forest plot

1

55

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.42, 3.23]

8.13 Forced vital capacity < ‐2 SD Show forest plot

2

183

Risk Ratio (M‐H, Fixed, 95% CI)

0.57 [0.24, 1.34]

8.14 Exercise‐induced bronchoconstriction Show forest plot

1

56

Risk Ratio (M‐H, Fixed, 95% CI)

0.87 [0.13, 5.73]

Figures and Tables -
Comparison 8. Respiratory outcomes in childhood ‐ after 5 years
Comparison 9. Growth in childhood

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

9.1 Height ‐ z‐score Show forest plot

2

208

Mean Difference (IV, Fixed, 95% CI)

0.14 [‐0.18, 0.46]

9.2 Height < ‐2 SD Show forest plot

3

229

Risk Ratio (M‐H, Fixed, 95% CI)

0.76 [0.36, 1.60]

9.3 Weight ‐ z‐score Show forest plot

2

207

Mean Difference (IV, Fixed, 95% CI)

0.03 [‐0.35, 0.40]

9.4 Weight < ‐2 SD Show forest plot

2

90

Risk Ratio (M‐H, Fixed, 95% CI)

0.64 [0.17, 2.37]

9.5 Body mass index (BMI) ‐ z‐score Show forest plot

2

205

Mean Difference (IV, Fixed, 95% CI)

0.02 [‐0.34, 0.38]

9.6 BMI < ‐2 SD Show forest plot

1

67

Risk Ratio (M‐H, Fixed, 95% CI)

0.49 [0.13, 1.87]

Figures and Tables -
Comparison 9. Growth in childhood
Comparison 10. Blood pressure in childhood

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

10.1 Systolic blood pressure > 95th centile Show forest plot

2

207

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.49, 1.45]

10.2 Systolic blood pressure z‐score Show forest plot

1

67

Mean Difference (IV, Fixed, 95% CI)

0.04 [‐0.43, 0.52]

10.3 Diastolic blood pressure > 95th centile Show forest plot

2

206

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.23, 4.60]

10.4 Diastolic blood pressure z‐score Show forest plot

1

67

Mean Difference (IV, Fixed, 95% CI)

0.01 [‐0.32, 0.34]

Figures and Tables -
Comparison 10. Blood pressure in childhood