Clinical characteristics and outcomes in neonates with perinatal acute respiratory distress syndrome in China: A national, multicentre, cross-sectional study

Long Chen, Jie Li, Yuan Shi, Chinese Neonatal ARDS (ChiNARDS) study group, Long Chen, Jie Li, Yuan Shi, Chinese Neonatal ARDS (ChiNARDS) study group

Abstract

Background: Neonatal acute respiratory distress syndrome (NARDS) was defined in 2017 and the epidemiological data remain unknown. Our objective was to explore aetiological factors, clinical characteristics and outcomes in patients with perinatal NARDS.

Methods: A multicentre, prospective, cross-sectional study was performed in 58 tertiary neonatal intensive care units in China from Jan 1, 2018 to June 30, 2019. Neonates diagnosed with NARDS were included. Primary outcomes were aetiological factors, clinical characteristics and outcomes. Binary logistic regression and multivariate cox proportional regression were performed to identify independent predictors for bronchopulmonary dysplasia (BPD) and/or death or single death. This study was registered with ClinicalTrials.Gov, NCT03311165.

Findings: Among 70,013 admitted neonates, the incidence of NARDS was 1.44% (1005). The cumulative incidences were 65.6%, 86.7%, 94.1% within one, two and three days after birth. The median gestational age and birth weight were 36.4 weeks and 2700 g. Three main aetiological triggers included pneumonia (58.1%), asphyxia (24.3%) and early-onset sepsis (EOS) (21.3%). BPD and/or death was observed in 213 (21.2%) infants, consisting 104 (10.3%) BPD and 126 (12.6%) deaths. The numbers of mild, moderate and severe NARDS were 537 (53.4%), 286 (28.4%) and 182 (18.2%). Two or more doses of surfactant was associated with increased mortality as compared with one or less doses of surfactant (odds ratio [OR] 1.93, 95% confidence interval [CI] 1.20-3.10, P = 0.006). Similarity also appeared in the comparison between EOS and non-EOS triggers (OR 1.57, 95% CI 1.06-2.33, P = 0.023).

Interpretation: NARDS incidence was 1.44% and the three main aetiologies were pneumonia, asphyxia and EOS. The cumulative incidences were 65.6%, 86.7%, and 94.1% within one, two and three days after birth. Our results suggested that two or more doses of surfactant increased mortality compared with one or less doses of surfactant.

Funding: The National Clinical Research Center of China and Clinical Medical Study Program of Children's Hospital of Chongqing Medical University (NCRC-2019-GP-13) and Natural Science Foundation of Chongqing (cstc2020jcyj-msxmX0197).

Keywords: ARDS, acute respiratory distress syndrome; Acute respiratory distress syndrome; BIPAP, bi-level positive airway pressure; BPD, bronchopulmonary dysplasia; Bronchopulmonary dysplasia; CI, confidence interval; CMV, conventional mechanical ventiation; Epidemiology; GA, gestational age; GDM, gestational diabetes mellitus; HDCP, hypertensive disorder complicating pregnancy; HFOV, high-frequency oscillatory ventilation; ICP, intrahepatic cholestasis of pregnancy; IQR, interquartile range; IV, invasive ventilation; IVF, in vitro fertilization; MAS, meconium aspiration syndrome; Mortality; NARDS, neonatalacute respiratory distress syndrome; NCPAP, nasal continuous positive airway pressure; NHFOV, nasal high-frequency oscillatory ventilation; NIPPV, nasal intermittent positive pressure ventilation; NIV, noninvasive ventilation; NIV-I, intubation after NIV failure; Neonate; OR, odds ratio; PH, pulmonary hemorrhage; PPHN, persistent pulmonary hypertension of newborn; PROM, premature rupture of the membrane; iNO, inhaled nitric oxide.

Conflict of interest statement

No financial disclosure, and the other authors also have no financial disclosures relevant to this article. No financial or non-financial benefits have been received or will be received from any party related directly or indirectly to the individuals of this article.

© 2022 The Author(s).

Figures

Fig. 1
Fig. 1
Flow chart of studied neonates. ARDS: acute respiratory distress syndrome; NICU: neonatal intensive care unit.
Fig. 2
Fig. 2
Changing trends of chest X-rays in neonates with perinatal NARDS. (2A–2C) 30 + 3 weeks. (2A) The chest X-ray at 5 h after birth after the first administration of surfactant with NCPAP; (2B) The chest X-ray before the second administration of surfactant after invasive ventilation on the second day, OI: 22; now the antibiotic was used; (2C) the chest X-ray before extubation on the fifth day. Diagnosis: EOS related NARDS. (2D–2F) 36 + 6 weeks. (2D) the chest X-ray at 10 h after birth after the first administration of surfactant with NCPAP; (2E) the chest X-ray before the second administration of surfactant on the second day, OI: 10; now the antibiotic was used; (2F) the chest X-ray on the third day. Diagnosis: EOS related NARDS. (2G–2J) 30 + 1 weeks. (2G) the chest X-ray at 2 h after birth before the first administration of surfactant with NIPPV, OI:12; (2H) the chest X-ray on the third day, OI: 3; (2I) the chest X-ray on the eighth day; (2J) the chest X-ray on the fifth day, now the antibiotic was used; Diagnosis: LOS related NARDS. (2K–2M) 26 + 3 weeks. (2K) the chest X-ray at 4 h after birth before the first administration of surfactant with NIPPV, OI:16; (2L) the chest X-ray on the second day before extubation, OI: 4; (2M) the chest X-ray on the tenth day, now the antibiotic was used, OI:15. Diagnosis: LOS related NARDS. NCPAP: nasal continuous positive airway pressure; OI: oxygenation index; EOS: early onset sepsis; NARDS: neonatal acute respiratory distress syndrome; NIPPV: nasal intermittent positive pressure ventilation; LOS: late onset sepsis.
Fig. 3
Fig. 3
Kaplan–Meier analyses for in-hospital survival for respiratory modes of NIV, NIV-I and IV (Fig 3A) (P = 0.356), mild, moderate and severe NARDS (Fig 3B) (P = 0.032), direct vs indirect insult (Fig 3C) (HR 2.55, 95% CI 1.71–3.58, P < 0.001) and EOS vs non-EOS (Fig 3D) (HR 1.72, 95% CI 1.21–2.91, P = 0.006). ARDS plus RDS vs ARDS (Fig 3E) (HR 0.95, 95% CI 0.64–1.41, P = 0.817) and non-perinatal-onset ARDS vs late-onset ARDS (Fig 3F) (HR 2.89, 95% CI 0.62–6.50, P = 0.259). NIV: noninvasive ventilation; NIV-I: intubation after NIV failure; IV: invasive ventilation; NARDS: neonatal acute respiratory distress syndrome; HR:Hazard ratio; CI:confidence interval; OS: early onset sepsis; ARDS: acute respiratory distress syndrome; RDS: respiratory distress syndrome.

References

    1. Ashbaugh D.G., Boyd B.D., Petty T.L., Levine B.E. Acute respiratory distress in adults. Lancet. 1967;290:319–323.
    1. ARDS Definition Task Force. Ranieri V.M., Rubenfeld G.D., et al. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307:2526–2533.
    1. Khemani R.G., Smith L.S., Zimmerman J.J., Erickson S., For the Pediatric Acute Lung Injury Consensus Conference Group Pediatric acute respiratory distress syndrome: definition, incidence, and epidemiology: proceedings from the pediatric acute lung injury consensus Conference. Pediatr Crit Care Med. 2015;16:S23–S40.
    1. De Luca D., van Kaam A.H., Tingay D.G., et al. The Montreux definition of neonatal ARDS: biological and clinical background behind the description of a new entity. Lancet Respir Med. 2017;5:657–666.
    1. the Chinese Neonatal ARDS(CNARDS) study group The protocol for neonatal acute respiratory distress syndrome epidemiology: a multicenter observational study. Chin J Evid based Pediatr. 2018;13:70–74. (in Chinese)
    1. Jobe A.H., Bancalari E. Bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2001;163:1723–1729.
    1. International Committee for the Classification of Retinopathy of Prematurity The international Classification of retinopathy of prematurity revisited. Arch Ophthalmol. 2005;123(7):991–999.
    1. Stoll B.J. Epidemiology of necrotizing enterocolitis. Clin Perinatol. 1994;21(2):205–218.
    1. Papile L.A., Munsick-Bruno G., Schaefer A. Relationship of cerebral intraventricular hemorrhage and early childhood neurologic handicaps. J Pediatr. 1983;103(2):273–277.
    1. Kidokoro H., Anderson P.J., Doyle L.W., et al. Brain injury and altered brain growth in preterm infants: predictors and prognosis. Pediatrics. 2014;134(2):e444–e453.
    1. Cochi S.E., Kempker J.A., Annangi S., Kramer M.R., Martin G.S. Mortality trends of acute respiratory distress syndrome in the United States from 1999-2013. Ann Am Thorac Soc. 2016;13:1742–1751.
    1. Khemani R.G., Smith L., Lopez-Fernandez Y.M., et al. Paediatric acute respiratory distress syndrome incidence and epidemiology (PARDIE): an international, observational study. Lancet Respir Med. 2019;7:115–128.
    1. Wen-Liang Yu, Lu Zhu-Jin, Wang Ying, et al. The epidemiology of acute respiratory distress syndrome in pediatric intensive care units in China. Intensive Care Med. 2009;35:136–143.
    1. Zecca E., De Luca D., Marras M., Caruso A., Bernardini T., Romagnoli C. Intrahepatic cholestasis of pregnancy and neonatal respiratory distress syndrome. Pediatrics. 2006;117:1669–1672.
    1. Tang Shuo, Lei Bao. Clinical characteristics and prognosis-related factors of neonatal acute respiratory distress syndrome. J Third Mil Med Univ. 2019;41:898–902. (in Chinese)
    1. Shi Y., De Luca D. NASal OscillatioN post-Extubation (NASONE) study group. Continuous positive airway pressure (CPAP) vs noninvasive positive pressure ventilation (NIPPV) vs noninvasive high frequency oscillation ventilation (NHFOV) as post-extubation support in preterm neonates: protocol for an assessor-blinded, multicenter, randomized controlled trial. BMC Pediatr. 2019;19(1):256.
    1. De Luca D., Tingay D.G., van Kaam A.H., et al. Epidemiology of neonatal acute respiratory distress syndrome: prospective, multicenter, international Cohort study. Pediatr Crit Care Med. 2022;23(7):524–534.
    1. Hudson L.D., Milberg J.A., Anardi D., Maunder R.J. Clinical risks for development of the acute respiratory distress syndrome. Am J Respir Crit Care Med. 1995;151:293–301.
    1. Pediatric Acute Lung Injury Consensus Conference Group Pediatric acute respiratory distress syndrome: consensus recommendations from the pediatric acute lung injury consensus Conference. Pediatr Crit Care Med. 2015;16:428–439.
    1. Anzueto A., Baughman R.P., Guntupalli K.K., et al. Aerosolized surfactant in adults with sepsis-induced acute respiratory distress syndrome. Exosurf acute respiratory distress syndrome sepsis study group. N Engl J Med. 1996;334:1417–1421.
    1. Sweet D.G., Carnielli V., Greisen G., et al. European consensus guidelines on the management of respiratory distress syndrome-2019 update. Neonatology. 2019;115:432–450.
    1. Zhangxue H., Min G., Jinning Z., et al. Glycochenodeoxycholate induces rat alveolar epithelial type II cell death and inhibits surfactant secretion in vitro. Free Radic Biol Med. 2012;53:122–128.
    1. Chen X., Shan Q., Jiang L., Zhu B., Xi X. Quantitative proteomic analysis by iTRAQ for identification of candidate biomarkers in plasma from acute respiratory distress syndrome patients. Biochem Biophys Res Commun. 2013;441:1–6.
    1. Thompson B.T., Chambers R.C., Liu K.D. Acute respiratory distress syndrome. N Engl J Med. 2017;377:562–572.
    1. El Shahed A.I., Dargaville P.A., Ohlsson A., Roger S. Surfactant for meconium aspiration syndrome in term and late preterm infants. Cochrane Database Syst Rev. 2014;2014:CD002054.
    1. Natarajan C.K., Sankar M.J., Jain K., Agarwal R., Paul V.K. Surfactant therapy and antibody in neonates with meconium aspiration syndrome: a systematic review and meta-analysis. J Perinatol. 2016;36:S49–S54.
    1. Bellani G., Laffey J.G., Pham T., et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 Countries. JAMA. 2016;315:788–800.
    1. Sheu C.C., Gong M.N., Zhai R., et al. Clinical characteristics and outcomes of sepsis-related vs non-sepsis-related ARDS. Chest. 2010;138:559–567.
    1. Yehya N., Keim G., Thomas J.N. Subtypes of pediatric acute respiratory distress syndrome have different predictors of mortality. Intensive Care Med. 2018;44:1230–1239.
    1. Pneumonia Etiology Research for Child Health (PERCH) Study Group Causes of severe pneumonia requiring hospital admission in children without HIV infection from Africa and Asia: the PERCH multi-country case-control study. Lancet. 2019;394:757–779.
    1. Cao Y., Jiang S., Sun J., et al. Assessment of neonatal intensive care unit practices, morbidity, and mortality among very preterm infants in China. JAMA Netw Open. 2021;4(8) doi: 10.1001/jamanetworkopen.2021.18904.

Source: PubMed

3
Suscribir