1
|
DeMauro SB, Kirpalani H, Ziolkowski K, Hintz S, Watterberg K, Lowe J, Shankaran S, Chawla S, Vohr B, Msall M, D'Angio C, Yoder BA, Lai K, Winter S, Colaizy T, Merhar S, Bann CM, Trotta M, Newman J, Natarajan A, Das A. The HYdrocortisone for Bronchopulmonary Dysplasia Respiratory and Developmental (HYBRiD) outcomes study: protocol for a longitudinal cohort study. BMC Pediatr 2024; 24:737. [PMID: 39543521 PMCID: PMC11562334 DOI: 10.1186/s12887-024-05198-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) affects up to half of extremely preterm infants, and is associated with adverse long-term respiratory, neurodevelopmental, and educational sequelae and costly health service and family economic outcomes. The NICHD Neonatal Research Network Hydrocortisone for Bronchopulmonary Dysplasia (BPD) Trial evaluated the efficacy and safety of hydrocortisone treatment to prevent BPD in high-risk infants. The trial enrolled 800 very preterm infants with respiratory failure and followed the participants until 2 years corrected age to assess safety of the trial intervention. Longer-term impacts of hydrocortisone exposure and severity of BPD on functional outcomes of high-risk infants remain unknown. The HYdrocortisone for BPD Respiratory and Developmental (HYBRiD) Outcomes Study extends follow-up of all surviving children enrolled in the Hydrocortisone for BPD Trial until early school age. It aims to characterize the childhood functional motor, cognitive, academic, and pulmonary outcomes of this large, well-phenotyped trial cohort. METHODS Parents of surviving trial participants complete telephone questionnaires when their children are 3 and 4 years corrected age. A single in-person study visit takes place at early school age (5 years, 0 months to 7 years, 11 months corrected age). Children undergo a multidimensional assessment of functional outcomes and parents complete a battery of questionnaires. In 5 of 19 participating centers, respiratory mechanics are evaluated with impulse oscillometry. DISCUSSION The HYBRiD Outcomes Study will be the largest and most comprehensive evaluation to date of the functional early school age outcomes of children with a history of severe neonatal lung disease and of children exposed to HC during infancy. This will substantially improve understanding of the longer-term implications of severe neonatal lung disease; provide data to facilitate the development of future randomized intervention trials in this population; and inform public policy by enhancing knowledge about school age resource requirements in children with a history of prematurity and lung disease. TRIAL REGISTRATION clinicaltrials.gov ID NCT01353313. Primary trial registration 5/11/11 modified to include followup through school age 12/13/17. This manuscript reflects version 3 of the trial manuscript, dated 10/12/2020.
Collapse
Affiliation(s)
- Sara B DeMauro
- Children's Hospital of Philadelphia, Philadelphia, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | - Haresh Kirpalani
- Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | | | - Susan Hintz
- School of Medicine, Stanford University, Stanford, USA
| | | | - Jean Lowe
- University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Seetha Shankaran
- University of Texas at Austin, Austin, USA
- Wayne State University, Detroit, USA
| | - Sanjay Chawla
- Wayne State University, Detroit, USA
- Children's Hospital of Michigan and Central Michigan University, Mount Pleasant, USA
| | - Betty Vohr
- Women & Infants Hospital of Rhode Island and Warren Alpert Medical School of Brown University, Providence, USA
| | - Michael Msall
- Comer Children's Hospital and Kennedy Research Center on Intellectual and Developmental Disabilities of the University of Chicago School of Medicine, Chicago, USA
| | - Carl D'Angio
- University of Rochester School of Medicine and Dentistry, Rochester, USA
| | | | - Khanh Lai
- University of Utah School of Medicine, Salt Lake City, USA
| | - Sarah Winter
- University of Utah School of Medicine, Salt Lake City, USA
| | | | - Stephanie Merhar
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, USA
| | - Carla M Bann
- Analytics Division, RTI International, North Carolina, USA
| | - Marissa Trotta
- Analytics Division, RTI International, North Carolina, USA
| | - Jamie Newman
- Analytics Division, RTI International, North Carolina, USA
| | | | - Abhik Das
- National Heart Lung and Blood Institute, Bethesda, USA
| |
Collapse
|
2
|
Kimoto Y, Hirata K, Hirano S, Wada K, Moriichi A, Ito Y, Cho K, Mizuno K, Toyoshima K, Kushima R, Ishii N, Yamaguchi T, Hashimoto K, Yada Y, Kono Y. Post-Discharge Growth Among Extremely Preterm Infants With or Without Bronchopulmonary Dysplasia. Pediatr Pulmonol 2024. [PMID: 39513637 DOI: 10.1002/ppul.27388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024]
Abstract
OBJECTIVE This study aimed to evaluate whether bronchopulmonary dysplasia (BPD) and home respiratory care (HRC) after discharge, including home oxygen therapy (HOT) or home mechanical ventilation (HMV), were associated with the growth of extremely preterm infants. STUDY DESIGN This secondary study uses data from a study on the infant and early childhood development survey of low-birth-weight infants in the Health, Labor, and Welfare Science Research. We enrolled 788 singleton extremely preterm infants appropriate for gestational age (GA). BPD was defined as the requirement of supplemental oxygen or respiratory support at 36 weeks corrected GA. We compared trends of anthropometric measurements from age 0 to 5 years among infants with non-BPD, BPD without HRC, and BPD with HRC. RESULTS We obtained 4113 weight and length measurements. At age 0-5 years, the BPD with the HRC group weighed less and was shorter than the other two groups. Lower GA and BPD status were significantly associated with growth failure of weight at 3 and 5 years after adjustment for potential risk factors. Lower GA was significantly associated with growth failure of length at 3 and 5 years. Conversely, although BPD status was significantly associated with growth failure of length at 3 years, it was not at 5 years. CONCLUSION Among extremely preterm infants, BPD with HRC was associated with a significant growth delay at 0-5 years compared with non-BPD and BPD without HRC. However, the effect of different severities of BPD on growth failure of length was attenuated with age.
Collapse
Affiliation(s)
- Yasuka Kimoto
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Katsuya Hirata
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Shinya Hirano
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Kazuko Wada
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Akinori Moriichi
- Division of Information for Specific Pediatric Chronic Diseases, Research Institute, National Center for Child Health and Development, Tokyo, Japan
| | - Yoshiya Ito
- Division of Clinical Medicine, Japanese Red Cross Hokkaido College of Nursing, Kitami, Japan
| | - Kazutoshi Cho
- Department of Pediatrics, Japan Community Healthcare Organization Hokkaido Hospital, Sapporo, Japan
| | - Katsumi Mizuno
- Department of Pediatrics, School of Medicine, Showa University, Tokyo, Japan
| | - Katsuaki Toyoshima
- Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Kanagawa, Japan
| | - Reiko Kushima
- Department of Neonatology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Nozomi Ishii
- Department of Newborn Medicine, Aiiku Hospital, Tokyo, Japan
| | - Takeshi Yamaguchi
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan
| | - Keiji Hashimoto
- Department of Rehabilitation Medicine, Showa University School of Medicine, Yokohama, Japan
| | - Yukari Yada
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Yumi Kono
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
3
|
Oh WO, Heo YJ. Early Parenting Interventions to Enhance Development in Infants and Children Born Prematurely: A Systematic Review and Meta-Analysis. J Pediatr Health Care 2024; 38:e1-e20. [PMID: 39023458 DOI: 10.1016/j.pedhc.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/08/2024] [Accepted: 06/08/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Parenting interventions have the potential to become effective strategies for improving the developmental trajectories of infants and children born prematurely. However, the effectiveness of parenting interventions is not well understood. METHODS A literature search was conducted in five databases. A total of 24 studies involving 3,636 participants were included for review. RESULTS The results showed a significant effect in cognition, language, motor development, and behavioral problems of children born prematurely. Parenting stress, anxiety, and interactive behaviors showed significant effect size. DISCUSSION This review focuses on interventions that employ scaffolding parenting strategies to enhance the development of children born prematurely. Efforts should continue to empower parents through effective and sustainable parenting interventions to improve the quality of life of preterm children.
Collapse
Affiliation(s)
- Won-Oak Oh
- Won-Oak Oh, Professor and Yoo-Jin Heo, Post-Doctoral Researcher, College of Nursing, Korea University, Seoul, South Korea
| | - Yoo-Jin Heo
- Won-Oak Oh, Professor and Yoo-Jin Heo, Post-Doctoral Researcher, College of Nursing, Korea University, Seoul, South Korea.
| |
Collapse
|
4
|
Gallipoli A, Unger S, El Shahed A, Fan CPS, Signorile M, Wilson D, Hoban R. Outcomes after intranasal human milk therapy in preterm infants with intraventricular hemorrhage. J Perinatol 2024:10.1038/s41372-024-02147-3. [PMID: 39384614 DOI: 10.1038/s41372-024-02147-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
OBJECTIVE Intraventricular hemorrhage (IVH) is a common cause of brain injury in preterm infants. Fresh human milk (HM) contains stem cells (SCs) that could potentially be delivered via intranasal HM (IHM). In this IHM pilot study, we describe outcomes. STUDY DESIGN Infants <33 weeks gestation with IVH were given IHM until maximum 28 days of age. Short-term neurologic outcomes and follow-up testing were compared to historic HM-fed infants. Longitudinal outcomes were plotted using linear mixed models. Weighted G-computation quantified treatment effects. Propensity score models calculated inverse probability weights for IVH grade, gestational age, and sex. RESULT 37 infants (35.1% grade 3-4 IVH) were compared to 191 historic controls (17.8% grade 3-4 IVH). Post-hemorrhagic ventricular dilatation was common (25.7% IHM patients). Most weighted outcomes, although not significant, favored IHM at 4-12 and 18 months corrected age. CONCLUSION This phase 1 study suggests powered trials of IHM for brain injury are needed. CLINICAL TRIAL REGISTRY NAME: clinicaltrials.gov identifier NCT04225286.
Collapse
Affiliation(s)
- Alessia Gallipoli
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sharon Unger
- Department of Paediatrics, Izaak Walton Killam Hospital, Halifax, NS, Canada
| | - Amr El Shahed
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Chun-Po Steve Fan
- Ted Rogers Computational Program, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Marisa Signorile
- Ted Rogers Computational Program, Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Diane Wilson
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rebecca Hoban
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada.
- Division of Neonatology, Seattle Children's Hospital, Seattle, WA, USA.
- University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Johnson ED, Keppel K, McNamara L, Collaco JM, Boss RD. Continuous Neuromuscular Blockade for Bronchopulmonary Dysplasia. Am J Perinatol 2024; 41:1848-1857. [PMID: 38447952 DOI: 10.1055/s-0044-1782180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
OBJECTIVE Bronchopulmonary dysplasia (BPD) is the most common late morbidity for premature infants. Continuous neuromuscular blockade (CNMB) is suggested for the most unstable phase of BPD, despite no outcome data. We explored the association between duration of CNMB for severe BPD and mortality. DESIGN Medical record review of children <5 years old admitted from 2016 to 2022 with BPD and one or more course of CNMB for ≥14 days. RESULTS Twelve children received a total of 20 episodes of CNMB for ≥14 days (range 14-173 d) during their hospitalization. Most (10/12) were born at <28 weeks' gestation and most (11/12) with birth weight <1,000 g; 7/12 were of Black race/ethnicity. All were hospitalized since birth. Most (10/12) were initially transferred from an outside neonatal intensive care unit (ICU), typically after a >60-day hospitalization (9/12). Half (6/12) of them had a ≥60-day stay in our neonatal ICU before transferring to our pediatric ICU for, generally, ≥90 days (8/12). The primary study outcome was survival to discharge: 2/12 survived. Both had shorter courses of CNMB (19 and 25 d); only one child who died had a course ≤25 days. Just two infants had increasing length Z-scores during hospitalization; only one infant had a final length Z-score > - 2. CONCLUSION In this case series of infants with severe BPD, there were no survivors among those receiving ≥25 days of CNMB. Linear growth, an essential growth parameter for infants with BPD, decreased in most patients. These data do not support the use of ≥25 days of CNMB to prevent mortality in infants with severe BPD. KEY POINTS · This is a case series of neuromuscular blockade for severe BPD.. · Neuromuscular blockade did not improve linear growth.. · Ten out of 12 infants who were on prolonged neuromuscular blockade died..
Collapse
Affiliation(s)
- Emily D Johnson
- Department of Nursing, Johns Hopkins Hospital, Baltimore, Maryland
| | - Kristopher Keppel
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - LeAnn McNamara
- Department of Pharmacy, Johns Hopkins Hospital, Baltimore, Maryland
| | - Joseph M Collaco
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Renee D Boss
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
- Berman Institute of Bioethics, Baltimore, Maryland
| |
Collapse
|
6
|
Notz L, Adams M, Bassler D, Boos V. Association between early metabolic acidosis and bronchopulmonary dysplasia/death in preterm infants born at less than 28 weeks' gestation: an observational cohort study. BMC Pediatr 2024; 24:605. [PMID: 39342228 PMCID: PMC11438188 DOI: 10.1186/s12887-024-05077-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Metabolic acidosis occurs frequently during the first postnatal days in extremely preterm infants and is mainly attributed to renal immaturity. Recent studies suggested a link between metabolic acidosis and the development of BPD. The aim of this study was to systematically investigate the association between severe metabolic acidosis during the first two weeks of life and bronchopulmonary dysplasia (BPD) / mortality among preterm infants born before 28 weeks' gestation. METHODS Monocentric observational cohort study including 1748 blood gas samples of 138 extremely preterm infants born 2020-2022. Metabolic acidosis was defined as pH < 7.2 with base excess (BE) < -10 mmol/L or standard bicarbonate (SBC) < 12 mmol/L. Primary outcome was BPD and/or death at 36 weeks postmenstrual age. RESULTS Fifty-six (40.6%) infants had BPD/death. Metabolic acidosis occurred in 50.0% of infants with BPD/death, compared to 22.0% of BPD-free survivors (p = 0.001) during the first 14 postnatal days. Minimum pH (median 7.12 vs. 7.19, p < 0.001), BE (median -10.9 vs. -9.5 mmol/L, p = 0.005), SBC (median 14.7 vs. 16.1 mmol/L, p < 0.001) were different between the two groups. After adjusting for confounders, pH (postnatal days 2-6), BE (postnatal day 3) and SBC (postnatal days 2-4) were significantly lower in infants with BPD/death. Metabolic acidosis on postnatal days 1-7 was associated with higher odds of BPD (adjusted Odds Ratio (aOR) 3.461, 95% CI 1.325-9.042) and BPD/death (aOR 3.087, 95% CI 1.225-7.778). CONCLUSIONS Metabolic acidosis during the first week of life was associated with higher odds of BPD/death in extremely preterm infants.
Collapse
Affiliation(s)
- Laura Notz
- Department of Neonatology, Newborn Research, University Hospital Zurich (USZ), University of Zurich (UZH), Frauenklinikstrasse 10, Zurich, 8091, Switzerland
| | - Mark Adams
- Department of Neonatology, Newborn Research, University Hospital Zurich (USZ), University of Zurich (UZH), Frauenklinikstrasse 10, Zurich, 8091, Switzerland
| | - Dirk Bassler
- Department of Neonatology, Newborn Research, University Hospital Zurich (USZ), University of Zurich (UZH), Frauenklinikstrasse 10, Zurich, 8091, Switzerland
| | - Vinzenz Boos
- Department of Neonatology, Newborn Research, University Hospital Zurich (USZ), University of Zurich (UZH), Frauenklinikstrasse 10, Zurich, 8091, Switzerland.
| |
Collapse
|
7
|
Luo L, Li Y, Long Z, Jiang F, Wu F, Wang Q. Exploring research trends and hotspots on oxidative stress and bronchopulmonary dysplasia: Insights from bibliometric and visualized study. Pediatr Pulmonol 2024. [PMID: 39264135 DOI: 10.1002/ppul.27268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/18/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a severe chronic lung disease primarily affecting premature infants, often resulting from prolonged mechanical ventilation and oxygen therapy. Oxidative stress plays a critical role in the pathogenesis of BPD, contributing to lung injury, inflammation, and impaired lung development. Despite extensive research, there is a need to systematically map out the research trends and hotspots in this field to inform future studies and therapeutic strategies. METHODS This study utilized bibliometric and visualized analysis to explore global research trends and hotspots on oxidative stress and BPD from 2004 to 2024. A comprehensive literature search was conducted in the Web of Science Core Collection, focusing on publications related to oxidative stress and BPD. Tools such as VOSviewer, Citespace, and the R package Bibliometrix were employed to analyze Coauthorship, co-citation, and keyword co-occurrence networks, as well as to identify emerging research fronts and influential studies. RESULTS The analysis identified 597 relevant publications, showing a steady increase in research output over the 20-year period, with a significant surge in the last decade. The United States led in research contributions, followed by China and Germany, with notable collaborations among these countries. Coauthorship analysis highlighted key research institutions, such as Harvard University and the University of California, as central nodes in the research network. Thematic clustering revealed five major research areas: antioxidant mechanisms, inflammation, molecular pathways, lung development, and therapeutic interventions. The keyword co-occurrence analysis showed a shift in research focus over time. Early studies concentrated on basic pathophysiological mechanisms, while recent research has increasingly focused on advanced molecular techniques, such as gene expression and targeted therapies. Notably, the study identified emerging research hotspots, including the role of extracellular vesicles and cellular senescence in BPD, as well as the potential therapeutic applications of antioxidants like superoxide dismutase mimetics. CONCLUSION This bibliometric study provides a comprehensive overview of the research landscape on oxidative stress and BPD, identifying key trends, influential authors, and emerging research topics. The findings underscore the importance of continued research in this field, particularly in translating basic scientific insights into clinical applications to improve outcomes for infants affected by BPD. The study also highlights potential areas for future investigation, including the development of novel therapeutic strategies targeting oxidative stress in BPD.
Collapse
Affiliation(s)
- Liyan Luo
- Department of Neonatology, Dali Bai Autonomous Prefecture Maternal and Child Health Care Hospital, Dali, China
| | - Yuan Li
- Department of Dermatology, The Fifth People's Hospital of Hainan Province, Haikou, China
| | - Zhi Long
- Department of Neonatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Fang Wu
- Department of Neonatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Wang
- Department of Neonatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
He Y, Li D, Zhang M, Li F. Bioinformatic analysis reveals the relationship between macrophage infiltration and Cybb downregulation in hyperoxia-induced bronchopulmonary dysplasia. Sci Rep 2024; 14:20089. [PMID: 39209930 PMCID: PMC11362550 DOI: 10.1038/s41598-024-70877-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common sequela of prematurity and is characterized by alveolar simplification and lung angiogenesis failure. The aim of this study was to explore the immune signatures of BPD. Differentially expressed gene analysis and immune infiltration analysis were conducted to identify key immune cell types and related genes by using the mRNA-seq dataset GSE25286. The expression patterns of key genes were validated in the scRNA-seq dataset GSE209664 and in experiments. The cell-cell crosstalk of key immune cells was explored with CellChat. We found that differentially expressed genes between BPD mice and controls were mostly enriched in leukocyte migration and M1 macrophages were highly enriched in BPD lungs. Hub genes (Cybb, Papss2, F7 and Fpr2) were validated at the single-cell level, among which the downregulation of Cybb was most closely related to macrophage infiltration. The reduced mRNA and protein levels of Cybb were further validated in animal experiments. Colocalization analysis of Cybb and macrophage markers demonstrated a significant decrease of Cybb in M1 macrophages. Cell-cell crosstalk found that alveolar epithelial cells interacted actively with macrophages through MIF-(CD74 + CD44) signalling. In conclusion, M1 macrophages played important roles in promoting BPD-like lung injury, which was correlated with a specific reduction of Cybb in macrophages and the potential activation of MIF signalling.
Collapse
Affiliation(s)
- Yi He
- Department of Pediatrics, Women and Children's Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children; Chongqing Research Center for Prevention & Control of Maternal and Child Diseases and Public Health, Chongqing, 401147, China
| | - Decai Li
- Department of Pediatrics, Women and Children's Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children; Chongqing Research Center for Prevention & Control of Maternal and Child Diseases and Public Health, Chongqing, 401147, China
| | - Meiyu Zhang
- Department of Neonatal Diagnosis and Treatment Center, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400015, China
| | - Fang Li
- Department of Pediatrics, Women and Children's Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children; Chongqing Research Center for Prevention & Control of Maternal and Child Diseases and Public Health, Chongqing, 401147, China.
| |
Collapse
|
9
|
Ford N, Bloomfield FH, Jiang Y, Cormack BE. Early hypophosphataemia and refeeding syndrome in extremely low birthweight babies and outcomes to 2 years of age: secondary cohort analysis from the ProVIDe trial. Arch Dis Child Fetal Neonatal Ed 2024:fetalneonatal-2024-327029. [PMID: 39122562 DOI: 10.1136/archdischild-2024-327029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE To investigate in extremely low birthweight (ELBW; <1000 g) babies the associations between refeeding syndrome (serum phosphate <1.4 mmol·L-1 and serum total calcium>2.8 mmol·L-1) and hypophosphataemia in the first week and death or neurodisability at 2 years' corrected age (CA). DESIGN Secondary cohort analysis of the ProVIDe trial participants with serum biochemistry within 7 days of birth. At 2 years' CA, neurodisability was assessed by Bayley Scales of Infant Development Edition III and neurological examination. Associations between neurodisability and other variables were analysed using t-tests and logistic regression adjusted for sex and smallness-for-gestational age. SETTING Six tertiary neonatal intensive care units (NICUs) in New Zealand. PARTICIPANTS 352 ELBW babies born between 29 April 2014 and 30 October 2018. MAIN OUTCOME MEASURE Death or neurodisability at 2 years' CA. RESULTS Fifty-nine babies died, two after discharge from the NICU. Of the 336 babies who survived to 2 years' CA, 277 had neurodevelopmental assessment and 107 (39%) had a neurodisability. Death or neurodisability was more likely in babies who had refeeding syndrome (aOR 1.96 (95% CI 1.09 to 3.53), p=0.02) and in babies who had hypophosphataemia (aOR 1.74 (95% CI 1.09 to 2.79), p=0.02). Hypophosphataemia was associated with increased risk of death (aOR 2.07 (95% CI 1.09 to 3.95), p=0.03)) and severe hypophosphataemia (<0.9 mmol·L-1) with increased risk of death (aOR 2.67 (95% CI 1.41 to 5.00), p=0.002) and neurodisability (aOR 2.31 (95% CI 1.22 to 4.35), p=0.01). CONCLUSIONS In ELBW babies, refeeding syndrome and hypophosphataemia in the first week are associated with death or neurodisability. Until optimal phosphate requirements are determined through further research, monitoring for hypophosphataemia and mitigation strategies are indicated. TRIAL REGISTRATION NUMBER ACTRN12612001084875.
Collapse
Affiliation(s)
- Nadia Ford
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Yannan Jiang
- Department of Statisitics, University of Auckland, Auckland, New Zealand
| | - Barbara Elizabeth Cormack
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Starship Child Health, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
10
|
Wang L, Xiao J, Zhang B, Hou A. Epigenetic modifications in the development of bronchopulmonary dysplasia: a review. Pediatr Res 2024; 96:632-642. [PMID: 38570557 DOI: 10.1038/s41390-024-03167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
While perinatal medicine advancements have bolstered survival outcomes for premature infants, bronchopulmonary dysplasia (BPD) continues to threaten their long-term health. Gene-environment interactions, mediated by epigenetic modifications such as DNA methylation, histone modification, and non-coding RNA regulation, take center stage in BPD pathogenesis. Recent discoveries link methylation variations across biological pathways with BPD. Also, the potential reversibility of histone modifications fuels new treatment avenues. The review also highlights the promise of utilizing mesenchymal stem cells and their exosomes as BPD therapies, given their ability to modulate non-coding RNA, opening novel research and intervention possibilities. IMPACT: The complexity and universality of epigenetic modifications in the occurrence and development of bronchopulmonary dysplasia were thoroughly discussed. Both molecular and cellular mechanisms contribute to the diverse nature of epigenetic changes, suggesting the need for deeper biochemical techniques to explore these molecular alterations. The utilization of innovative cell-specific drug delivery methods like exosomes and extracellular vesicles holds promise in achieving precise epigenetic regulation.
Collapse
Affiliation(s)
- Lichuan Wang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Jun Xiao
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Bohan Zhang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Ana Hou
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Pharande P, Sehgal A, Menahem S. Cardiovascular Sequelae of Bronchopulmonary Dysplasia in Preterm Neonates Born before 32 Weeks of Gestational Age: Impact of Associated Pulmonary and Systemic Hypertension. J Cardiovasc Dev Dis 2024; 11:233. [PMID: 39195141 DOI: 10.3390/jcdd11080233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common respiratory disorder of prematurity for infants born before 32 weeks of gestational age (GA). Early and prolonged exposure to chronic hypoxia and inflammation induces pulmonary hypertension (PH) with the characteristic features of a reduced number and increased muscularisation of the pulmonary arteries resulting in an increase in the pulmonary vascular resistance (PVR) and a fall in their compliance. BPD and BPD-associated pulmonary hypertension (BPD-PH) together with systemic hypertension (sHTN) are chronic cardiopulmonary disorders which result in an increased mortality and long-term problems for these infants. Previous studies have predominantly focused on the pulmonary circulation (right ventricle and its function) and developing management strategies accordingly for BPD-PH. However, recent work has drawn attention to the importance of the left-sided cardiac function and its impact on BPD in a subset of infants arising from a unique pathophysiology termed postcapillary PH. BPD infants may have a mechanistic link arising from chronic inflammation, cytokines, oxidative stress, catecholamines, and renin-angiotensin system activation along with systemic arterial stiffness, all of which contribute to the development of BPD-sHTN. The focus for the treatment of BPD-PH has been improvement of the right heart function through pulmonary vasodilators. BPD-sHTN and a subset of postcapillary PH may benefit from afterload reducing agents such as angiotensin converting enzyme inhibitors. Preterm infants with BPD-PH are at risk of later cardiac and respiratory morbidities as young adults. This paper reviews the current knowledge of the pathophysiology, diagnosis, and treatment of BPD-PH and BPD-sHTN. Current knowledge gaps and emerging new therapies will also be discussed.
Collapse
Affiliation(s)
- Pramod Pharande
- Monash Newborn, Monash Children's Hospital, 246 Clayton Road, Clayton, Melbourne, VIC 3168, Australia
- Department of Pediatrics, Monash University, Melbourne, VIC 3800, Australia
| | - Arvind Sehgal
- Monash Newborn, Monash Children's Hospital, 246 Clayton Road, Clayton, Melbourne, VIC 3168, Australia
- Department of Pediatrics, Monash University, Melbourne, VIC 3800, Australia
| | - Samuel Menahem
- Department of Pediatrics, Monash University, Melbourne, VIC 3800, Australia
- Paediatric and Foetal Cardiac Units, Monash Medical Centre, Melbourne, VIC 3168, Australia
- Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
12
|
Butler V, Treluyer L, Patkaï J, Biset A, Jarreau PH, Ancel PY, Rozé JC, Marchand-Martin L, Durox M, Lapillonne A, Picaud JC, Mitanchez D, Tscherning C, Biran V, Cambonie G, Lopez E, Hascoet JM, Desfrere L, Chollat C, Zana-Taïeb E, Torchin H. Mortality and neurodevelopmental outcomes at 2 years' corrected age of very preterm infants with necrotising enterocolitis or spontaneous intestinal perforation: The EPIPAGE-2 cohort study. Eur J Pediatr 2024:10.1007/s00431-024-05675-4. [PMID: 38955846 DOI: 10.1007/s00431-024-05675-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE The primary objective was to evaluate the impact of necrotising enterocolitis (NEC) and spontaneous intestinal perforation (SIP) on mortality and neurodevelopmental outcomes at 2 years' corrected age (CA) in infants born before 32 weeks' gestation (WG). METHODS We studied neurodevelopment at 2 years' CA of infants with NEC or SIP who were born before 32 WG from the EPIPAGE-2 cohort study. The primary outcome was death or the presence of moderate-to-severe motor or sensory disability defined by moderate-to-severe cerebral palsy or hearing or visual disability. The secondary outcome was developmental delay defined by a score < 2 SDs below the mean for any of the five domains of the Ages and Stages Questionnaire. RESULTS At 2 years' CA, 46% of infants with SIP, 34% of infants with NEC, and 14% of control infants died or had a moderate-to-severe sensorimotor disability (p < 0.01). This difference was mainly due to an increase in in-hospital mortality in the infants with SIP or NEC. Developmental delay at 2 years' CA was more frequent for infants with SIP than controls (70.8% vs 44.0%, p = 0.02) but was similar for infants with NEC and controls (49.3% vs 44.0%, p = 0.5). On multivariate analysis, the likelihood of developmental delay was associated with SIP (adjusted odds ratio = 3.0, 95% CI 1.0-9.1) but not NEC as compared with controls. CONCLUSION NEC and SIP significantly increased the risk of death or sensorimotor disability at 2 years' CA. SIP was also associated with risk of developmental delay at 2 years' CA.
Collapse
Grants
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- ANR-11-EQPX-0038 and ANR-19-COHO001 Centre National de la Recherche Scientifique
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- 11779 Fondation de France
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- SPF20160936356 Fondation pour la Recherche Médicale
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- DGOS13-040 Epinutri
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- G13129KK Ministère de l'Enseignement Supérieur, De La Recherche et de L'Innovation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
- R20065KK Apicil Foundation
Collapse
Affiliation(s)
- Victoria Butler
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Paris Cité University, Cochin Hospital, 53 Avenue de L'Observatoire, Paris, 75014, France.
| | - Ludovic Treluyer
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Paris Cité University, Cochin Hospital, 53 Avenue de L'Observatoire, Paris, 75014, France
- Paris Cité University, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, Paris, 75006, France
| | - Juliana Patkaï
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Paris Cité University, Cochin Hospital, 53 Avenue de L'Observatoire, Paris, 75014, France
| | - Aline Biset
- Department of Neonatal Paediatrics, Trousseau Hospital, Assistance Publique - Hôpitaux de Paris, Sorbonne University, Paris, France
| | - Pierre-Henri Jarreau
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Paris Cité University, Cochin Hospital, 53 Avenue de L'Observatoire, Paris, 75014, France
- Paris Cité University, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, Paris, 75006, France
- FHU Prema, Paris, France
| | - Pierre-Yves Ancel
- Paris Cité University, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, Paris, 75006, France
- FHU Prema, Paris, France
- Clinical Investigation Center P1419, Assistance Publique - Hôpitaux de Paris, Paris, France
- Nutrition EPIPAGE-2 Study Group, Paris, France
| | - Jean-Christophe Rozé
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Department of Neonatology, CHU Nantes, Nantes, France
- UMR PhAN 1280 INRAE, CIC004 INSERM, Nantes, France
| | - Laetitia Marchand-Martin
- Paris Cité University, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, Paris, 75006, France
- Nutrition EPIPAGE-2 Study Group, Paris, France
| | - Mélanie Durox
- Paris Cité University, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, Paris, 75006, France
- Nutrition EPIPAGE-2 Study Group, Paris, France
| | - Alexandre Lapillonne
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Necker-Enfants Malades Hospital, Paris, France
- URP 7328, Paris Cité University, Paris, France
| | - Jean-Charles Picaud
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Department of Neonatology, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, 69677, France
- CarMen, INSERM, INRA, Université Claude Bernard Lyon 1, Pierre-Bénite, Lyon, 69310, France
| | - Delphine Mitanchez
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Department of Neonatology, Bretonneau Hospital, François Rabelais University, Tours, 37000, France
| | - Charlotte Tscherning
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Division of Neonatology, Oslo University Hospital, Oslo, 0372, Norway
- Infinity, University of Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Valérie Biran
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Hôpital Robert Debré, Paris, France
- INSERM U1141, Paris Cité University, Paris, France
| | - Gilles Cambonie
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Department of Neonatology, CHU Montpellier, Montpellier University, Montpellier, France
- INSERM UMR 1058, University of Montpellier, Montpellier, France
| | | | - Jean-Michel Hascoet
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Neonatology Department, Maternité Régionale Universitaire Adolphe Pinard - Nancy University, Nancy, France
- DevAH, University of Lorraine, Vandoeuvre-Les-Nancy, France
| | - Luc Desfrere
- Nutrition EPIPAGE-2 Study Group, Paris, France
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Louis Mourier Hospital, Colombes, France
| | - Clément Chollat
- Department of Neonatal Paediatrics, Trousseau Hospital, Assistance Publique - Hôpitaux de Paris, Sorbonne University, Paris, France
- NeuroDiderot, INSERM, Paris Cité University, Paris, France
| | - Elodie Zana-Taïeb
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Paris Cité University, Cochin Hospital, 53 Avenue de L'Observatoire, Paris, 75014, France
- INSERM U955, IMRB, Créteil, France
| | - Héloïse Torchin
- Neonatal Intensive Care Unit, Assistance Publique - Hôpitaux de Paris, Paris Cité University, Cochin Hospital, 53 Avenue de L'Observatoire, Paris, 75014, France
- Paris Cité University, Centre of Research in Epidemiology and StatisticS (CRESS), Obstetrical Perinatal and Pediatric Epidemiology Research Team (EPOPé), INSERM, INRAE, Paris, 75006, France
| |
Collapse
|
13
|
Chong D, Belteki G. Detection and quantitative analysis of patient-ventilator interactions in ventilated infants by deep learning networks. Pediatr Res 2024; 96:418-426. [PMID: 38316942 DOI: 10.1038/s41390-024-03064-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND The study of patient-ventilator interactions (PVI) in mechanically ventilated neonates is limited by the lack of unified PVI definitions and tools to perform large scale analyses. METHODS An observational study was conducted in 23 babies randomly selected from 170 neonates who were ventilated with SIPPV-VG, SIMV-VG or PSV-VG mode for at least 12 h. 500 breaths were randomly selected and manually annotated from each recording to train convolutional neural network (CNN) models for PVI classification. RESULTS The average asynchrony index (AI) over all recordings was 52.5%. The most frequently occurring PVIs included expiratory work (median: 28.4%, interquartile range: 23.2-40.2%), late cycling (7.6%, 2.8-10.2%), failed triggering (4.6%, 1.2-6.2%) and late triggering (4.4%, 2.8-7.4%). Approximately 25% of breaths with a PVI had two or more PVIs occurring simultaneously. Binary CNN classifiers were developed for PVIs affecting ≥1% of all breaths (n = 7) and they achieved F1 scores of >0.9 on the test set except for early triggering where it was 0.809. CONCLUSIONS PVIs occur frequently in neonates undergoing conventional mechanical ventilation with a significant proportion of breaths containing multiple PVIs. We have developed computational models for seven different PVIs to facilitate automated detection and further evaluation of their clinical significance in neonates. IMPACT The study of patient-ventilator interactions (PVI) in mechanically ventilated neonates is limited by the lack of unified PVI definitions and tools to perform large scale analyses. By adapting a recent taxonomy of PVI definitions in adults, we have manually annotated neonatal ventilator waveforms to determine prevalence and co-occurrence of neonatal PVIs. We have also developed binary deep learning classifiers for common PVIs to facilitate their automatic detection and quantification.
Collapse
Affiliation(s)
- David Chong
- Neonatal Intensive Care Unit, The Rosie Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Gusztav Belteki
- Neonatal Intensive Care Unit, The Rosie Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
14
|
Hannan KE, Bourque SL, Ross E, Wymore EM, Kinsella JP, Mandell EW, Houin SS. Successful and Rapid Reduction in Neurosedative and Analgesic Medications in Complex Infants with Severe Bronchopulmonary Dysplasia After Tracheostomy Placement: Experience with 24-hour Propofol Infusions. J Pediatr 2024; 270:114040. [PMID: 38554746 DOI: 10.1016/j.jpeds.2024.114040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Infants with severe bronchopulmonary dysplasia may require high doses of neurosedative medications to ensure pain control and stability following tracheostomy placement. Subsequent weaning of these medications safely and rapidly is a challenge. We describe a 24-hour propofol infusion to reduce neurosedative medications in 3 high-risk infants following tracheostomy placement.
Collapse
Affiliation(s)
- Kathleen E Hannan
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO.
| | - Stephanie L Bourque
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Emma Ross
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Erica M Wymore
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - John P Kinsella
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Erica W Mandell
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Satya S Houin
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
15
|
Taha A, Akangire G, Noel-Macdonnell J, Gladdis T, Manimtim W. The impact of early tracheostomy on neurodevelopmental outcomes of infants with severe bronchopulmonary dysplasia exposed to postnatal corticosteroids. J Perinatol 2024; 44:979-987. [PMID: 38158399 DOI: 10.1038/s41372-023-01864-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE To compare the cognitive, language and motor scores of infants with severe BPD exposed to postnatal corticosteroids (PCS) and had early (ET), late (LT) or no tracheostomy (NT). METHODS Retrospective study was designed to compare the developmental outcomes of 71 infants born between 2010 and 2017 with severe BPD exposed to PCS and had ET (≤122 days), LT (>122 days), or NT. RESULTS Cognitive scores were lower in LT versus NT and ET (p = 0.050); motor scores were worse in LT versus NT and ET (p = 0.004). Dexamethasone use was higher in LT versus NT and ET (p = 0.040). Adjusted for PCS, odds for major cognitive impairment were 90% less in ET versus LT. Trend for improved language and motor outcomes was seen in ET versus LT. CONCLUSION Infants with severe BPD exposed to PCS and had ET had significantly better cognitive, and trend toward improved language and motor outcomes.
Collapse
Affiliation(s)
- Amjad Taha
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Gangaram Akangire
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA.
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.
| | - Janelle Noel-Macdonnell
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
- Department of Health Services and Outcomes Research, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Tiffany Gladdis
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
- Department of Developmental and Behavioral Health, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Winston Manimtim
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| |
Collapse
|
16
|
Callahan KP, Farrell K, Gibbs K, Kielt MJ, Morris H, Nilan K, Thomas S, DeMauro SB. Childhood outcomes following discharge from a referral bronchopulmonary dysplasia program. J Perinatol 2024:10.1038/s41372-024-02035-w. [PMID: 38937610 DOI: 10.1038/s41372-024-02035-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
OBJECTIVES The primary objective of this study was to profile the childhood health, development, and health-related quality of life (HR QoL) for children with the most severe bronchopulmonary dysplasia (BPD), those discharged from a quaternary referral program. STUDY DESIGN We collected cross-sectional data through telephone interviews with 282 families of children ages 18 months to 11 years who had been discharged from a BPD referral program. RESULTS Respiratory morbidities were near universal, with 42% of children ever having required a tracheostomy and severity of these morbidities correlated with parent-reported health and QoL. Developmental morbidities were also marked: 97% required an individualized educational plan. While respiratory morbidities and overall health improved over time, developmental morbidities were increasingly prominent, resulting in lower quality of life. CONCLUSIONS Among children referred to a quaternary BPD program, respiratory and developmental morbidities are on numerous counts more severe than any reported in the literature.
Collapse
Affiliation(s)
- Katharine P Callahan
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Medical Ethics and Health Policy, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Kathryn Farrell
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Gibbs
- Department of Medical Ethics and Health Policy, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Heidi Morris
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen Nilan
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Sara B DeMauro
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Ransom MA, Blatt AM, Pua HH, Sucre JMS. The emerging role of extracellular vesicles in bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2024; 326:L517-L523. [PMID: 38469633 PMCID: PMC11380955 DOI: 10.1152/ajplung.00244.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/13/2024] Open
Abstract
Extracellular vesicle (EV) biology in neonatal lung development and disease is a rapidly growing area of investigation. Although EV research in the neonatal population lags behind EV research in adult lung diseases, recent discoveries demonstrate promise in furthering our understanding of the pathophysiology of bronchopulmonary dysplasia and the potential use of EVs in the clinical setting, as both biomarkers and therapeutic agents. This review article explores some of the recent advances in this field and our evolving knowledge of the role of EVs in bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Meaghan A Ransom
- Department of Pediatrics, Vanderbilt University Medical Center; Nashville, Tennessee, United States
| | - Alexander M Blatt
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center; Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
18
|
Solís-García G, Elias S, Dunn M, Jasani B. Late surfactant administration after 48 hours of age in preterm neonates with respiratory insufficiency: a systematic review and meta-analysis. Arch Dis Child Fetal Neonatal Ed 2024; 109:301-307. [PMID: 38071552 DOI: 10.1136/archdischild-2023-326333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/02/2023] [Indexed: 04/20/2024]
Abstract
OBJECTIVE To systematically review and meta-analyse the effect of late surfactant administration versus placebo in reducing the incidence of death or bronchopulmonary dysplasia (BPD) in preterm infants. DESIGN PubMed, EMBASE, CINAHL and Cochrane CENTRAL were searched until 30 May 2023, for randomised controlled trials (RCTs) comparing administration of surfactant after 48 hours of age versus placebo in preterm ventilator-dependent neonates. The primary outcome was incidence of death or BPD at 36 weeks' postmenstrual age (PMA). Secondary outcomes included incidence of BPD at 36 weeks PMA, pre-discharge mortality, use of postnatal steroids, post-discharge respiratory support, treatment with steroids or hospitalisation prior to 1-year corrected age. RESULTS Pooled analyses of four RCTs (N=850) showed no statistically significant difference between groups in the incidence of death or BPD at 36 weeks' PMA (relative risk (RR) 0.99; 95% CI 0.90 to 1.10; Grades of Recommendation, Assessment, Development and Evaluation (GRADE): moderate). Late surfactant administration significantly decreased the need for post-discharge respiratory support prior to 1-year corrected age (two RCTs; N=522; RR 0.72; 95% CI 0.59 to 0.89; GRADE: low). Other secondary outcomes did not differ significantly between the groups. CONCLUSIONS Administration of late surfactant does not improve the rates of death or BPD at 36 weeks when administered to preterm infants with prolonged respiratory insufficiency. Additional adequately powered trials are needed to establish the efficacy of late surfactant therapy in preterm infants. PROSPERO REGISTRATION NUMBER CRD42023432463.
Collapse
Affiliation(s)
- Gonzalo Solís-García
- Neonatology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neonatology, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Sara Elias
- Neonatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Dunn
- Neonatology, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | - Bonny Jasani
- Neonatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
19
|
van de Loo M, van Kaam A, Offringa M, Doyle LW, Cooper C, Onland W. Corticosteroids for the prevention and treatment of bronchopulmonary dysplasia: an overview of systematic reviews. Cochrane Database Syst Rev 2024; 4:CD013271. [PMID: 38597338 PMCID: PMC11005325 DOI: 10.1002/14651858.cd013271.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) remains an important complication of prematurity. Pulmonary inflammation plays a central role in the pathogenesis of BPD, explaining the rationale for investigating postnatal corticosteroids. Multiple systematic reviews (SRs) have summarised the evidence from numerous randomised controlled trials (RCTs) investigating different aspects of administrating postnatal corticosteroids. Besides beneficial effects on the outcome of death or BPD, potential short- and long-term harms have been reported. OBJECTIVES The primary objective of this overview was to summarise and appraise the evidence from SRs regarding the efficacy and safety of postnatal corticosteroids in preterm infants at risk of developing BPD. METHODS We searched the Cochrane Database of Systematic Reviews, MEDLINE, Embase, CINAHL, and Epistemonikos for SRs in April 2023. We included all SRs assessing any form of postnatal corticosteroid administration in preterm populations with the objective of ameliorating pulmonary disease. All regimens and comparisons were included. Two review authors independently checked the eligibility of the SRs comparing corticosteroids with placebo, and corticosteroids with different routes of administration and regimens. The included outcomes, considered key drivers in the decision to administer postnatal corticosteroids, were the composite outcome of death or BPD at 36 weeks' postmenstrual age (PMA), its individual components, long-term neurodevelopmental sequelae, sepsis, and gastrointestinal tract perforation. We independently assessed the methodological quality of the included SRs by using AMSTAR 2 (A Measurement Tool to Assess Systematic Reviews) and ROBIS (Risk Of Bias In Systematic reviews) tools. We assessed the certainty of the evidence using GRADE. We provided a narrative description of the characteristics, methodological quality, and results of the included SRs. MAIN RESULTS We included nine SRs (seven Cochrane, two non-Cochrane) containing 87 RCTs, 1 follow-up study, and 9419 preterm infants, investigating the effects of postnatal corticosteroids to prevent or treat BPD. The quality of the included SRs according to AMSTAR 2 varied from high to critically low. Risk of bias according to ROBIS was low. The certainty of the evidence according to GRADE ranged from very low to moderate. Early initiated systemic dexamethasone (< seven days after birth) likely has a beneficial effect on death or BPD at 36 weeks' PMA (risk ratio (RR) 0.88, 95% confidence interval (CI) 0.81 to 0.95; number needed to treat for an additional beneficial outcome (NNTB) 16, 95% CI 10 to 41; I2 = 39%; 17 studies; 2791 infants; moderate-certainty evidence) and on BPD at 36 weeks' PMA (RR 0.72, 95% CI 0.63 to 0.82; NNTB 13, 95% CI 9 to 21; I2 = 39%; 17 studies; 2791 infants; moderate-certainty evidence). Early initiated systemic hydrocortisone may also have a beneficial effect on death or BPD at 36 weeks' PMA (RR 0.90, 95% CI 0.82 to 0.99; NNTB 18, 95% CI 9 to 594; I2 = 43%; 9 studies; 1376 infants; low-certainty evidence). However, these benefits are likely accompanied by harmful effects like cerebral palsy or neurosensory disability (dexamethasone) or gastrointestinal perforation (both dexamethasone and hydrocortisone). Late initiated systemic dexamethasone (≥ seven days after birth) may have a beneficial effect on death or BPD at 36 weeks' PMA (RR 0.75, 95% CI 0.67 to 0.84; NNTB 5, 95% CI 4 to 9; I2 = 61%; 12 studies; 553 infants; low-certainty evidence), mostly contributed to by a beneficial effect on BPD at 36 weeks' PMA (RR 0.76, 95% CI 0.66 to 0.87; NNTB 6, 95% CI 4 to 13; I2 = 14%; 12 studies; 553 infants; low-certainty evidence). No harmful side effects were shown in the outcomes chosen as key drivers to the decision to start or withhold late systemic dexamethasone. No effects, either beneficial or harmful, were found in the subgroup meta-analyses of late hydrocortisone studies. Early initiated inhaled corticosteroids probably have a beneficial effect on death and BPD at 36 weeks' PMA (RR 0.86, 95% CI 0.75 to 0.99; NNTB 19, 95% CI not applicable; I2 = 0%; 6 studies; 1285 infants; moderate-certainty evidence), with no apparent adverse effects shown in the SRs. In contrast, late initiated inhaled corticosteroids do not appear to have any benefits or harms. Endotracheal instillation of corticosteroids (budesonide) with surfactant as a carrier likely has a beneficial effect on death or BPD at 36 weeks' PMA (RR 0.60, 95% CI 0.49 to 0.74; NNTB 4, 95% CI 3 to 6; I2 = 0%; 2 studies; 381 infants; moderate-certainty evidence) and on BPD at 36 weeks' PMA. No evidence of harmful effects was found. There was little evidence for effects of different starting doses or timing of systemic corticosteroids on death or BPD at 36 weeks' PMA, but potential adverse effects were observed for some comparisons. Lowering the dose might result in a more unfavourable balance of benefits and harms. Moderately early initiated systemic corticosteroids, compared with early systemic corticosteroids, may result in a higher incidence of BPD at 36 weeks' PMA. Pulse dosing instead of continuous dosing may have a negative effect on death and BPD at 36 weeks' PMA. We found no differences for the comparisons of inhaled versus systemic corticosteroids. AUTHORS' CONCLUSIONS This overview summarises the evidence of nine SRs investigating the effect of postnatal corticosteroids in preterm infants at risk for BPD. Late initiated (≥ seven days after birth) systemic administration of dexamethasone is considered an effective intervention to reduce the risk of BPD in infants with a high risk profile for BPD, based on a favourable balance between benefits and harms. Endotracheal instillation of corticosteroids (budesonide) with surfactant as a carrier is a promising intervention, based on the beneficial effect on desirable outcomes without (so far) negative side effects. Pending results of ongoing large, multicentre RCTs investigating both short- and long-term effects, endotracheal instillation of corticosteroids (budesonide) with surfactant as a carrier is not appropriate for clinical practice at present. Early initiated (< seven days after birth) systemic dexamethasone and hydrocortisone and late initiated (≥ seven days after birth) hydrocortisone are considered ineffective interventions, because of an unfavourable balance between benefits and harms. No conclusions are possible regarding early and late inhaled corticosteroids, as more research is needed.
Collapse
Affiliation(s)
- Moniek van de Loo
- Department of Neonatology, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
- Amsterdam Reproduction & Development, Amsterdam, Netherlands
| | - Anton van Kaam
- Department of Neonatology, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
- Amsterdam Reproduction & Development, Amsterdam, Netherlands
| | - Martin Offringa
- Child Health Evaluative Sciences, Hospital for Sick Children, Toronto, Canada
| | - Lex W Doyle
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Parkville, Australia
- Newborn Research, The Royal Women's Hospital, Parkville, Australia
| | - Chris Cooper
- Cochrane Neonatal Group, Vermont Oxford Network, Burlington, USA
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Wes Onland
- Department of Neonatology, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
- Amsterdam Reproduction & Development, Amsterdam, Netherlands
| |
Collapse
|
20
|
Halbmeijer NM, Onland W, Dudink J, Cools F, Debeer A, van Kaam AH, Benders MJNL, van der Aa NE. Effect of Systemic Hydrocortisone on Brain Abnormalities and Regional Brain Volumes in Ventilator-dependent Infants Born Preterm: Substudy of the SToP-BPD Study. J Pediatr 2024; 265:113807. [PMID: 37923196 DOI: 10.1016/j.jpeds.2023.113807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/04/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVE To evaluate whether a high cumulative dose of systemic hydrocortisone affects brain development compared with placebo when initiated between 7 and 14 days after birth in ventilated infants born preterm. STUDY DESIGN A double-blind, placebo-controlled, randomized trial was conducted in 16 neonatal intensive care units among infants born at <30 weeks of gestation or with a birth weight of <1250 g who were ventilator-dependent in the second week after birth. Three centers performed MRI at term-equivalent age. Brain injury was assessed on MRI using the Kidokoro scoring system and compared between the 2 treatment groups. Both total and regional brain volumes were calculated using an automatic segmentation method and compared using multivariable regression analysis adjusted for baseline variables. RESULTS From the 3 centers, 78 infants participated in the study and 59 had acceptable MRI scans (hydrocortisone group, n = 31; placebo group, n = 28). Analyses of the median global brain abnormality score of the Kidokoro score showed no difference between the hydrocortisone and placebo groups (median, 7; IQR, 5-9 vs median, 8, IQR, 4-10, respectively; P = .92). In 39 infants, brain tissue volumes were measured, showing no differences in the adjusted mean total brain tissue volumes, at 352 ± 32 mL in the hydrocortisone group and 364 ± 51 mL in the placebo group (P = .80). CONCLUSIONS Systemic hydrocortisone started in the second week after birth in ventilator-dependent infants born very preterm was not found to be associated with significant differences in brain development compared with placebo treatment. TRIAL REGISTRATION The SToP-BPD study was registered with the Netherlands Trial Register (NTR2768; registered on 17 February 2011; https://www.trialregister.nl/trial/2640) and the European Union Clinical Trials Register (EudraCT, 2010-023777-19; registered on 2 November 2010; https://www.clinicaltrialsregister.eu/ctr-search/trial/2010-023777-19/NL).
Collapse
Affiliation(s)
- Nienke M Halbmeijer
- Department of Neonatology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands.
| | - Wes Onland
- Department of Neonatology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Filip Cools
- Department of Neonatology, University Hospital Brussel, Brussel, Belgium
| | - Anne Debeer
- Department of Neonatology, University Hospital Leuven, Leuven, Belgium
| | - Anton H van Kaam
- Department of Neonatology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Manon J N L Benders
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Niek E van der Aa
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
21
|
Msall ME, Lagatta JM, Bora S. Optimizing trajectories of social adaptive competencies after extreme prematurity during the first 1000 days. Semin Fetal Neonatal Med 2024; 29:101531. [PMID: 38632009 PMCID: PMC11156543 DOI: 10.1016/j.siny.2024.101531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Over 75% of surviving extremely preterm infants do not have major neurodevelopmental disabilities; however, more than half face difficulties with communication, coordination, attention, learning, social, and executive function abilities. These "minor" challenges can have a negative impact on educational and social outcomes, resulting in physical, behavioral, and social health problems in adulthood. We will review assessment tools for social-emotional and adaptive functional skills in early childhood as these determine family and early childhood supports. We highlight bronchopulmonary dysplasia as an example of the critical intersections of parental wellbeing, medical and developmental adaptive trajectories in infancy and early childhood, and partnerships between child neurologists and community medical and developmental professionals. We examine studies of engaging parents to promote developmental trajectories, with a focus on supporting parent-child interactions that underlie communication, social-adaptive behaviors, and learning in the first 1000 days of life. Recommendations for neurodevelopmental surveillance and screening of extremely preterm infants can also be applied to infants with other risk factors for altered neurodevelopment.
Collapse
Affiliation(s)
- Michael E Msall
- Department of Pediatrics, Section of Developmental and Behavioral Pediatrics and Kennedy Research Center on Intellectual and Developmental Disabilities, University of Chicago Medicine, Chicago, IL, USA.
| | - Joanne M Lagatta
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Samudragupta Bora
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
22
|
Higgins BV, Baer RJ, Steurer MA, Karvonen KL, Oltman SP, Jelliffe-Pawlowski LL, Rogers EE. Resuscitation, survival and morbidity of extremely preterm infants in California 2011-2019. J Perinatol 2024; 44:209-216. [PMID: 37689808 PMCID: PMC10844092 DOI: 10.1038/s41372-023-01774-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/21/2023] [Accepted: 08/31/2023] [Indexed: 09/11/2023]
Abstract
OBJECTIVE To describe changes over time in resuscitation, survival, and morbidity of extremely preterm infants in California. STUDY DESIGN This population-based, retrospective cohort study includes infants born ≤28 weeks. Linked birth certificates and hospital discharge records were used to evaluate active resuscitation, survival, and morbidity across two epochs (2011-2014, 2015-2019). RESULTS Of liveborn infants, 0.6% were born ≤28 weeks. Active resuscitation increased from 16.9% of 22-week infants to 98.1% of 25-week infants and increased over time in 22-, 23-, and 25-week infants (p-value ≤ 0.01). Among resuscitated infants, survival to discharge increased from 33.2% at 22 weeks to 96.1% at 28 weeks. Survival without major morbidity improved over time for 28-week infants (p-value < 0.01). CONCLUSION Among infants ≤28 weeks, resuscitation and survival increased with gestational age and morbidity decreased. Over time, active resuscitation of periviable infants and morbidity-free survival of 28-week infants increased. These trends may inform counseling around extremely preterm birth.
Collapse
Affiliation(s)
- Brennan V Higgins
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| | - Rebecca J Baer
- California Preterm Birth Initiative, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Martina A Steurer
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- California Preterm Birth Initiative, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Kayla L Karvonen
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Scott P Oltman
- California Preterm Birth Initiative, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Laura L Jelliffe-Pawlowski
- California Preterm Birth Initiative, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Elizabeth E Rogers
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Munoz FA, Carter EH, Edwards EM, Jerome M, Litt JS. Does faster weight trajectory lead to improved neurodevelopmental outcomes in ELBW infants with bronchopulmonary dysplasia? J Perinatol 2024; 44:301-306. [PMID: 37898685 DOI: 10.1038/s41372-023-01808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/25/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
OBJECTIVE Examine the relationship between weight trajectory and 2-year neurodevelopmental outcomes for extremely low birthweight (ELBW) infants with BPD. STUDY DESIGN Secondary analysis of infants born from 2010 to 2019. The predictor was BPD severity and the outcome was neurodevelopmental impairment, defined as any Bayley Scales of Infant Development (BSID) III score <70 at 24 months' corrected age. Repeated measures logistic regression was performed. RESULTS In total, 5042 infants were included. Faster weight trajectory was significantly associated with a decreased probability of having at least one BSID III score <70 for infants with grade 1-2 BPD (p < 0.0001) and an increased probability of at least one BSID III score <70 for infants with grade 3 BPD (p < 0.009). There was no significant association between weight trajectory and BSID III score <70 for infants with grade 0 BPD. CONCLUSION The association between postnatal weight trajectory and neurodevelopmental outcome in this study differs by BPD severity.
Collapse
Affiliation(s)
- Fernando A Munoz
- Division of Neonatology, Oregon Health & Science University, Portland, OR, USA.
| | | | - Erika M Edwards
- Vermont Oxford Network, Burlington, VT, USA
- College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
- Robert Larner, MD, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Maggie Jerome
- Graduate Programs in Human Nutrition, Oregon Health & Science University, Portland, OR, USA
| | - Jonathan S Litt
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Social and Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
24
|
Marks JD, Schreiber MD. A Randomized Clinical Trial of Inhaled Nitric Oxide Treatment in Premature Infants Reveals the Effect of Maternal Racial Identity on Efficacy. J Clin Med 2023; 12:7567. [PMID: 38137636 PMCID: PMC10743643 DOI: 10.3390/jcm12247567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Respiratory distress syndrome increases the risk of death and bronchopulmonary dysplasia (BPD) in premature infants. Inhaled nitric oxide (iNO) may reduce these risks. Recent meta-analyses have suggested that iNO is effective only at doses higher than 5 ppm and in infants born to Black mothers. In a randomized, double-blinded, controlled trial, infants born before 32 0/7 weeks gestation, weighing <1500 g, and requiring respiratory support were assigned to receive iNO for either seven days (short iNO), or until 33 0/7 weeks PMA (long iNO). The primary outcome was death or BPD. A total of 273 patients were enrolled, of whom 83 receiving long iNO (61.5%) experienced the primary outcome, compared with 65 (47.1%) receiving short iNO (relative risk (RR) 1.37; 95% confidence interval (CI), 1.06-1.79; p = 0.017). This increase was due solely to increased BPD in infants weighing 750-999 g (RR 1.33, 95% CI 1.07-1.66, p = 0.009). However, there was no difference in the numbers of infants requiring supplemental oxygen at 40 weeks PMA. Among infants < 750 g, long-iNO-treated infants had a lower cumulative probability of death (χ2 5.12, p = 0.02). Long iNO increased the primary outcome in non-Black infants (RR 1.93, 95% CI 1.20-3.24) but not in Black infants. Understanding how maternal racial identity determines responses of premature infants to iNO may help narrow the gap in health outcomes between Black and non-Black infants.
Collapse
Affiliation(s)
| | - Michael D. Schreiber
- Departments of Pediatrics and Neurology, University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
25
|
Li W, Wang Y, Song J, Zhang C, Xu Y, Xu F, Wang X, Zhu C. Association between bronchopulmonary dysplasia and death or neurodevelopmental impairment at 3 years in preterm infants without severe brain injury. Front Neurol 2023; 14:1292372. [PMID: 38033771 PMCID: PMC10684711 DOI: 10.3389/fneur.2023.1292372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Objective We investigated the association between bronchopulmonary dysplasia (BPD) and 3 years death or neurodevelopmental impairment (NDI) in very preterm infants without severe brain injury. Method Our prospective cohort study recruited preterm infants who were born prior to 32 weeks of gestational age and survived in the neonatal intensive care unit until 36 weeks of corrected age. Upon reaching 3 years of age, each infant was assessed for death or NDI such as cerebral palsy, cognitive deficit, hearing loss, and blindness. Correlations between BPD and death or NDI were determined using multiple logistic regression analyses adjusted for confounding factors. Result A total of 1,417 infants without severe brain injury who survived until 36 weeks of corrected age were initially enrolled in the study. Over the study period, 201 infants were lost to follow-up and 5 infants were excluded. Our final dataset, therefore, included 1,211 infants, of which 17 died after 36 weeks of corrected age and 1,194 were followed up to 3 years of age. Among these infants, 337 (27.8%) developed BPD. Interestingly, by 3 years of age, BPD was demonstrated to be independently associated with death or NDI, with an adjusted odds ratio of 1.935 (95% confidence interval: 1.292-2.899, p = 0.001), in preterm infants without severe neonatal brain injury. Conclusion Our findings indicate that BPD is strongly associated with death or NDI in preterm infants without severe neonatal brain injury at 3 years of age. Further research is needed to understand the mechanisms linking the development of BPD with death or NDI and whether appropriate treatment of BPD may ameliorate or prevent the development of neurological complications.
Collapse
Affiliation(s)
- Wenli Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Department of Neonatology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Department of Neonatology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Song
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Department of Neonatology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Department of Neonatology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Falin Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Department of Neonatology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Center for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Goteborg, Sweden
| |
Collapse
|
26
|
Basu SK, Kapse KJ, Murnick J, Pradhan S, Spoehr E, Zhang A, Andescavage N, Nino G, du Plessis AJ, Limperopoulos C. Impact of bronchopulmonary dysplasia on brain GABA concentrations in preterm infants: Prospective cohort study. Early Hum Dev 2023; 186:105860. [PMID: 37757548 PMCID: PMC10843009 DOI: 10.1016/j.earlhumdev.2023.105860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is associated with cognitive-behavioral deficits in very preterm (VPT) infants, often in the absence of structural brain injury. Advanced GABA-editing techniques like Mescher-Garwood point resolved spectroscopy (MEGA-PRESS) can quantify in-vivo gamma-aminobutyric acid (GABA+, with macromolecules) and glutamate (Glx, with glutamine) concentrations to investigate for neurophysiologic perturbations in the developing brain of VPT infants. OBJECTIVE To investigate the relationship between the severity of BPD and basal-ganglia GABA+ and Glx concentrations in VPT infants. METHODS MRI studies were performed on a 3 T scanner in a cohort of VPT infants [born ≤32 weeks gestational age (GA)] without major structural brain injury and healthy-term infants (>37 weeks GA) at term-equivalent age. MEGA-PRESS (TE68ms, TR2000ms, 256averages) sequence was acquired from the right basal-ganglia voxel (∼3cm3) and metabolite concentrations were quantified in institutional units (i.u.). We stratified VPT infants into no/mild (grade 0/1) and moderate-severe (grade 2/3) BPD. RESULTS Reliable MEGA-PRESS data was available from 63 subjects: 29 healthy-term and 34 VPT infants without major structural brain injury. VPT infants with moderate-severe BPD (n = 20) had the lowest right basal-ganglia GABA+ (median 1.88 vs. 2.28 vs. 2.12 i.u., p = 0.025) and GABA+/choline (0.73 vs. 0.99 vs. 0.88, p = 0.004) in comparison to infants with no/mild BPD and healthy-term infants. The GABA+/Glx ratio was lower (0.34 vs. 0.44, p = 0.034) in VPT infants with moderate-severe BPD than in infants with no/mild BPD. CONCLUSIONS Reduced GABA+ and GABA+/Glx in VPT infants with moderate-severe BPD indicate neurophysiologic perturbations which could serve as early biomarkers of future cognitive deficits.
Collapse
Affiliation(s)
- Sudeepta K Basu
- Neonatology, Children's National Hospital, Washington, D.C., USA; Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA
| | - Kushal J Kapse
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA
| | - Jonathan Murnick
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Diagnostic Imaging and Radiology, Children's National Hospital, Washington, D.C., USA
| | - Subechhya Pradhan
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA
| | - Emma Spoehr
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA
| | - Anqing Zhang
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Biostatistics and Epidemiology, Children's National Hospital, Washington, D.C., USA
| | - Nickie Andescavage
- Neonatology, Children's National Hospital, Washington, D.C., USA; Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA; Division of Neurology, Children's National Hospital, Washington, D.C., USA
| | - Gustavo Nino
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Pulmonary and Sleep Medicine, Children's National Hospital, Washington, D.C., USA
| | - Adre J du Plessis
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Neurology, Children's National Hospital, Washington, D.C., USA; Perinatal Pediatrics institute, Children's National Hospital, Washington, D.C., USA
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA; Division of Diagnostic Imaging and Radiology, Children's National Hospital, Washington, D.C., USA; Division of Neurology, Children's National Hospital, Washington, D.C., USA.
| |
Collapse
|
27
|
Ahn SY, Chang YS, Lee MH, Sung S, Kim AR, Park WS. Five-year follow-up of phase II trial of stromal cells for bronchopulmonary dysplasia. Thorax 2023; 78:1105-1110. [PMID: 37604693 DOI: 10.1136/thorax-2022-219622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 07/03/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND We previously performed a phase II randomised double-blind clinical trial of mesenchymal stromal cell (MSCs) transplantation to prevent bronchopulmonary dysplasia in extremely premature infants. Subsequently, we followed the infants enrolled in this clinical trial to determine the safety and effectiveness of MSCs against bronchopulmonary dysplasia at 5-year follow-up. METHODS We evaluated infants at 5 years of age receiving placebo or MSCs in a prospective follow-up study. RESULTS In terms of the primary end point of composite respiratory morbidities, including respiratory problem-related readmission, emergency department visits or oxygen therapy, the MSC group had a rate of 60.7% for composite morbidities, while the control group showed a tendency of higher rate of 83.9% for the same outcomes without statistical significance. In terms of the secondary outcomes, the MSC group infants showed a tendency of being less likely to visit emergency department (control 67.7% vs MSC 35.7%) and to receive oxygen therapy (control 29.0% vs MSC 3.6%). No difference was observed in the incidence of respiratory problem-related hospital readmission or wheezing episodes between the groups. CONCLUSION Intratracheally instilled MSCs showed the possibility of potential to decrease respiratory symptom-related emergency department visits and oxygen therapy episodes in infants born extremely preterm during the 5 years after a phase II randomised controlled, double-blind trial of MSCs transplantation for bronchopulmonary dysplasia. This small size study suggests preliminary insights that can be further tested using larger sample sizes. TRIAL REGISTRATION NUMBER NCT01897987.
Collapse
Affiliation(s)
- So Yoon Ahn
- Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, South Korea
| | - Yun Sil Chang
- Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, South Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Myung Hee Lee
- Social Information Research Institute, Seoul, South Korea
| | - Sein Sung
- Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ai-Rhan Kim
- Pediatrics, University of Ulsan, Ulsan, South Korea
| | - Won Soon Park
- Pediatrics, Gangnam CHA Hospital, CHA University School of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
Zhang F, Wang M, Li Z, Deng J, Fan Y, Gou Z, Zhou Y, Huang L, Lu L. Rapamycin attenuates pyroptosis by suppressing mTOR phosphorylation and promoting autophagy in LPS-induced bronchopulmonary dysplasia. Exp Lung Res 2023; 49:178-192. [PMID: 37874145 DOI: 10.1080/01902148.2023.2266236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 09/27/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE/AIM Bronchopulmonary dysplasia (BPD) is associated with poor survival in preterm infants. Intrauterine infection can aggravate the degree of obstruction of alveolar development in premature infants; however, the pathogenic mechanism remains unclear. In this study, we sought to determine whether pyroptosis could be inhibited by downregulating mammalian target of rapamycin (mTOR) activation and inducing autophagy in BPD-affected lung tissue. MATERIALS AND METHODS We established a neonatal rat model of BPD induced by intrauterine infection via intraperitoneally injecting pregnant rats with lipopolysaccharide (LPS). Subsequently, mTOR levels and pyroptosis were evaluated using immunohistochemistry, immunofluorescence, TUNEL staining, and western blotting. The Shapiro-Wilk test was employed to assess the normality of the experimental data. Unpaired t-tests were used to compare the means between two groups, and comparisons between multiple groups were performed using analysis of variance. RESULTS Pyroptosis of lung epithelial cells increased in BPD lung tissues. After administering an mTOR phosphorylation inhibitor (rapamycin) to neonatal rats with BPD, the level of autophagy increased, while the expression of autophagy cargo adaptors, LC3 and p62, did not differ. Following rapamycin treatment, NLRP3, Pro-caspase-1, caspase-1, pro-IL-1β, IL-1β, IL-18/Pro-IL-18, N-GSDMD/GSDMD, Pro-caspase-11, and caspase-11 were negatively regulated in BPD lung tissues. The opposite results were observed after treatment with the autophagy inhibitor MHY1485, showing an increase in pyroptosis and a significant decrease in the number of alveoli in BPD. CONCLUSIONS Rapamycin reduces pyroptosis in neonatal rats with LPS-induced BPD by inhibiting mTOR phosphorylation and inducing autophagy; hence, it may represent a potential therapeutic for treating BPD.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
- Clinic Medical College, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Minrong Wang
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
- Clinic Medical College, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Zhongni Li
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
- Clinic Medical College, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Jiehong Deng
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
- Clinic Medical College, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Yang Fan
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Zhixian Gou
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Yue Zhou
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Li Huang
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| | - Liqun Lu
- Department of Pediatrics, The First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
- Clinic Medical College, Chengdu Medical College, Chengdu, Sichuan Province, P.R. China
| |
Collapse
|
29
|
Katz TA, van Kaam AH, Mugie SM, Aarnoudse-Moens CSH, de Groof F, van Kempen AAMW, van den Heuvel MEN, Vogelzang J, Rijpert M, Schiering IA, Koomen-Botman I, Visser F, Leemhuis AG, Onland W. Risk Factors for Neurodevelopmental Impairment at 2- and 5-Years Corrected Age in Preterm Infants with Established Bronchopulmonary Dysplasia. Neonatology 2023; 121:125-132. [PMID: 37852207 PMCID: PMC10836742 DOI: 10.1159/000533653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/11/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION The objective of this study was to identify risk factors for neurodevelopmental impairment (NDI) at 2- and 5-years corrected age (CA) in a cohort of preterm infants with established bronchopulmonary dysplasia (BPD). METHODS This single-center retrospective cohort study included infants born between 2009 and 2016 at a gestational age (GA) <30 weeks with moderate or severe BPD at 36 weeks' postmenstrual age. Perinatal characteristics, (social) demographics, and comorbidities were collected from the electronic patient records. Odds ratios for NDI were calculated with univariate and multivariate logistic regression analyses adjusting for potential confounders. RESULTS Of the 602 eligible infants, 123 infants were diagnosed with BPD. NDI was present in 30.3% and 56.1% at 2- and 5-years CA, respectively. The only independent risk factors associated with NDI in the multivariate analyses were birthweight (adjusted odds ratio [aOR] 0.74, 95% CI 0.57-0.95; aOR 0.70, 95% CI 0.54-0.91, respectively), small for GA (SGA) (aOR 3.25, 95% CI 1.09-9.61; aOR 5.44, 95% CI 1.62-18.2, respectively) at both time points, and male gender at 5-years CA (OR 2.49, 95% CI 1.11-5.57). CONCLUSION Birthweight and SGA are independent risk factors for NDI at 2- and 5-years CA and male gender at 5-years CA in preterm infants with BPD. In contrast, well-known other risk factors for NDI in the general population of preterm infants, such as GA, maternal education, and neonatal comorbidities were not independently associated with NDI.
Collapse
Affiliation(s)
- Trixie A Katz
- Department of Neonatology, Amsterdam University Medical Centers, Emma Children's Hospital, Amsterdam, The Netherlands,
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands,
| | - Anton H van Kaam
- Department of Neonatology, Amsterdam University Medical Centers, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Suzanne M Mugie
- Department of Neonatology, Amsterdam University Medical Centers, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Cornelieke S H Aarnoudse-Moens
- Department of Neonatology, Amsterdam University Medical Centers, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Femke de Groof
- Department of Pediatrics, North West Hospital Group, Alkmaar, The Netherlands
| | | | | | - Judith Vogelzang
- Department of Pediatrics, Flevo Hospital, Almere, The Netherlands
| | - Maarten Rijpert
- Department of Pediatrics, Zaans Medical Center, Zaandam, The Netherlands
| | - Irene A Schiering
- Department of Pediatrics, Spaarne Hospital, Haarlem, The Netherlands
| | | | - Fenna Visser
- Department of Pediatrics, Amstelland Hospital, Amstelveen, The Netherlands
| | - Aleid G Leemhuis
- Department of Neonatology, Amsterdam University Medical Centers, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Wes Onland
- Department of Neonatology, Amsterdam University Medical Centers, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Miller AN, Shepherd EG, Manning A, Shamim H, Chiang T, El-Ferzli G, Nelin LD. Tracheostomy in Severe Bronchopulmonary Dysplasia-How to Decide in the Absence of Evidence. Biomedicines 2023; 11:2572. [PMID: 37761012 PMCID: PMC10526913 DOI: 10.3390/biomedicines11092572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Infants with the most severe forms of bronchopulmonary dysplasia (BPD) may require long-term invasive positive pressure ventilation for survival, therefore necessitating tracheostomy. Although life-saving, tracheostomy has also been associated with high mortality, postoperative complications, high readmission rates, neurodevelopmental impairment, and significant caregiver burden, making it a highly complex and challenging decision. However, for some infants tracheostomy may be necessary for survival and the only way to facilitate a timely and safe transition home. The specific indications for tracheostomy and the timing of the procedure in infants with severe BPD are currently unknown. Hence, centers and clinicians display broad variations in practice with regard to tracheostomy, which presents barriers to designing evidence-generating studies and establishing a consensus approach. As the incidence of severe BPD continues to rise, the question remains, how do we decide on tracheostomy to provide optimal outcomes for these patients?
Collapse
Affiliation(s)
- Audrey N. Miller
- Comprehensive Center for Bronchopulmonary Dysplasia, Department of Pediatrics, Division of Neonatology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.N.M.); (E.G.S.); (G.E.-F.)
| | - Edward G. Shepherd
- Comprehensive Center for Bronchopulmonary Dysplasia, Department of Pediatrics, Division of Neonatology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.N.M.); (E.G.S.); (G.E.-F.)
| | - Amy Manning
- Department of Otolaryngology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.M.); (H.S.); (T.C.)
| | - Humra Shamim
- Department of Otolaryngology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.M.); (H.S.); (T.C.)
| | - Tendy Chiang
- Department of Otolaryngology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.M.); (H.S.); (T.C.)
| | - George El-Ferzli
- Comprehensive Center for Bronchopulmonary Dysplasia, Department of Pediatrics, Division of Neonatology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.N.M.); (E.G.S.); (G.E.-F.)
| | - Leif D. Nelin
- Comprehensive Center for Bronchopulmonary Dysplasia, Department of Pediatrics, Division of Neonatology, Nationwide Children’s Hospital, Ohio State University College of Medicine, Columbus, OH 43205, USA; (A.N.M.); (E.G.S.); (G.E.-F.)
| |
Collapse
|
31
|
Miller AN, Shepherd EG, El-Ferzli G, Nelin LD. Multidisciplinary bronchopulmonary dysplasia care. Expert Rev Respir Med 2023; 17:989-1002. [PMID: 37982177 DOI: 10.1080/17476348.2023.2283120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease in neonates and infants, which often presents with multisystem organ involvement, co-morbidities, and prolonged hospital stays. Therefore, a multidisciplinary chronic care approach is needed in the severest forms of BPD to optimize outcomes. However, this approach can be challenging to implement. The objective of this article is to review and synthesize the available literature regarding multidisciplinary care in infants and children with established BPD, and to provide a framework that can guide clinical practice and future research. AREAS COVERED A literature search was conducted using Ovid MEDLINE, CINAHL, and Embase and several components of multidisciplinary management of BPD were identified and reviewed, including chronic care, team development, team members, discharge planning, and outpatient care. EXPERT OPINION Establishing a core multidisciplinary group familiar with the chronicity of established BPD is recommended as best practice for this population. Acknowledging this is not feasible for all individual centers, it is important for clinical practice and future research to focus on the development and incorporation of national consulting services, telemedicine, and educational resources.
Collapse
Affiliation(s)
- Audrey N Miller
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital and Division of Neonatology, Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA
| | - Edward G Shepherd
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital and Division of Neonatology, Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA
| | - George El-Ferzli
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital and Division of Neonatology, Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA
| | - Leif D Nelin
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital and Division of Neonatology, Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
32
|
Altit G, Cayouette F, Dorval V, Lapointe A. Systemic hypertension in preterm infants and neurodevelopmental outcomes. J Perinatol 2023; 43:943-945. [PMID: 36609483 DOI: 10.1038/s41372-022-01577-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/13/2022] [Accepted: 11/30/2022] [Indexed: 01/08/2023]
Affiliation(s)
- Gabriel Altit
- Department of Pediatrics, Division of Neonatology, Montreal Children's Hospital, McGill University, Montréal, QC, Canada
| | - Florence Cayouette
- Department of Pediatrics, CHU Ste-Justine, Université de Montréal, Montréal, QC, Canada
| | - Véronique Dorval
- Department of Pediatrics, Division of Neonatology, CHU Sainte-Justine, Université de Montréal, 3175 chemin de la Côte Ste-Catherine, Montréal, QC, Canada
| | - Anie Lapointe
- Department of Pediatrics, Division of Neonatology, CHU Sainte-Justine, Université de Montréal, 3175 chemin de la Côte Ste-Catherine, Montréal, QC, Canada.
| |
Collapse
|
33
|
Sanlorenzo LA, Hatch LD. Developing a Respiratory Quality Improvement Program to Prevent and Treat Bronchopulmonary Dysplasia in the Neonatal Intensive Care Unit. Clin Perinatol 2023; 50:363-380. [PMID: 37201986 DOI: 10.1016/j.clp.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Improvements in respiratory care have resulted in improved outcomes for preterm infants over the past three decades. To target the multifactorial nature of neonatal lung diseases, neonatal intensive care units (NICUs) should consider developing comprehensive respiratory quality improvement programs that address all drivers of neonatal respiratory disease. This article presents a potential framework for developing a quality improvement program to prevent bronchopulmonary dysplasia in the NICU. Drawing on available research and quality improvement reports, the authors discuss key components, measures, drivers, and interventions that should be considered when building a respiratory quality improvement program devoted to preventing and treating bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Lauren A Sanlorenzo
- Department of Pediatrics, Division of Neonatology, Columbia University Medical Center, 3959 Broadway Avenue, New York, NY 10032, USA
| | - Leon Dupree Hatch
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, 4413 VCH, 2200 Children's Way, Nashville, TN 37232, USA; Center for Child Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA; Critical Illness, Brain Dysfunction, and Survivorship Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
34
|
Protective Effects of Early Caffeine Administration in Hyperoxia-Induced Neurotoxicity in the Juvenile Rat. Antioxidants (Basel) 2023; 12:antiox12020295. [PMID: 36829854 PMCID: PMC9952771 DOI: 10.3390/antiox12020295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
High-risk preterm infants are affected by a higher incidence of cognitive developmental deficits due to the unavoidable risk factor of oxygen toxicity. Caffeine is known to have a protective effect in preventing bronchopulmonary dysplasia associated with improved neurologic outcomes, although very early initiation of therapy is controversial. In this study, we used newborn rats in an oxygen injury model to test the hypothesis that near-birth caffeine administration modulates neuronal maturation and differentiation in the hippocampus of the developing brain. For this purpose, newborn Wistar rats were exposed to 21% or 80% oxygen on the day of birth for 3 or 5 days and treated with vehicle or caffeine (10 mg/kg/48 h). Postnatal exposure to 80% oxygen resulted in a drastic reduction of associated neuronal mediators for radial glia, mitotic/postmitotic neurons, and impaired cell-cycle regulation, predominantly persistent even after recovery to room air until postnatal day 15. Systemic caffeine administration significantly counteracted the effects of oxygen insult on neuronal maturation in the hippocampus. Interestingly, under normoxia, caffeine inhibited the transcription of neuronal mediators of maturing and mature neurons. The early administration of caffeine modulated hyperoxia-induced decreased neurogenesis in the hippocampus and showed neuroprotective properties in the neonatal rat oxygen toxicity model.
Collapse
|
35
|
张 晨, 李 文, 陆 林, 朱 楚, 秦 璠, 苑 孟, 薛 倩, 徐 发. [Influence of bronchopulmonary dysplasia on cerebral blood flow in preterm infants: a prospective study based on arterial spin labeling]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:31-37. [PMID: 36655661 PMCID: PMC9893825 DOI: 10.7499/j.issn.1008-8830.2208068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/27/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVES To investigate local cerebral blood perfusion in preterm infants with bronchopulmonary dysplasia (BPD) based on cerebral blood flow (CBF) values of arterial spin labeling (ASL). METHODS A prospective study was conducted on 90 preterm infants with a gestational age of <32 weeks and a birth weight of <1 500 g who were born in the Department of Obstetrics and admitted to the Department of Neonatology in the Third Affiliated Hospital of Zhengzhou University from August 2021 to June 2022. All of the infants underwent cranial MRI and ASL at the corrected gestational age of 35-40 weeks. According to the presence or absence of BPD, they were divided into a BPD group with 45 infants and a non-BPD group with 45 infants. The two groups were compared in terms of the CBF values of the same regions of interest (frontal lobe, temporal lobe, parietal lobe, occipital lobe, thalamus, and basal ganglia) on ASL image. RESULTS Compared with the non-BPD group, the BPD group had a significantly lower 1-minute Apgar score, a significantly longer duration of assisted ventilation, and a significantly higher incidence rate of fetal distress (P<0.05). After control for the confounding factors such as corrected age and age at the time of cranial MRI by multiple linear regression analysis, compared with the non-BPD group, the BPD group still had higher CBF values of the frontal lobe, temporal lobe, parietal lobe, occipital lobe, basal ganglia, and thalamus at both sides (P<0.05). CONCLUSIONS BPD can increase cerebral blood perfusion in preterm infants, which might be associated with hypoxia and a long duration of assisted ventilation in the early stage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - 发林 徐
- 河南省儿科疾病临床医学研究中心,河南郑州450052
- 郑州大学先进医学研究中心,河南郑州450052
| |
Collapse
|
36
|
Halbmeijer NM, Onland W, Cools F, Swarte RM, van der Heide-Jalving M, Dijk P, Mulder-de Tollenaer S, Tan RNGB, Mohns T, Bruneel E, van Heijst AFJ, Kramer B, Debeer A, van Weissenbruch MM, Marechal Y, Blom H, Plaskie K, Offringa M, van Wassenaer-Leemhuis AG, van Kaam AH, Aarnoudse-Moens CSH. Effect of systemic hydrocortisone in ventilated preterm infants on parent-reported behavioural outcomes at 2 years' corrected age: follow-up of a randomised clinical trial. Arch Dis Child Fetal Neonatal Ed 2023:archdischild-2022-324179. [PMID: 36593110 DOI: 10.1136/archdischild-2022-324179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To report the parent-reported behavioural outcomes of infants included in the Systemic Hydrocortisone To Prevent Bronchopulmonary Dysplasia in preterm infants study at 2 years' corrected age (CA). DESIGN Randomised placebo-controlled trial. SETTING Dutch and Belgian neonatal intensive care units. PATIENTS Infants born <30 weeks' gestation and/or birth weight <1250 g, and ventilator dependent in the second week of life. INTERVENTION Infants were randomly assigned to a 22-day course of systemic hydrocortisone (cumulative dose 72.5 mg/kg; n=182) or placebo (n=190). MAIN OUTCOME MEASURES Parent-reported behavioural outcomes at 2 years' CA assessed with the Child Behavior Checklist (CBCL 1½-5). RESULTS Parents completed the CBCL of 183 (70% (183/262)) infants (hydrocortisone group, n=96; placebo group, n=87). Multiple imputation was used to account for missing data. Infants with critically elevated T-scores (>55) were found in 22.9%, 19.1% and 29.4% of infants for total, internalising and externalising problems, respectively; these scores were not significantly different between groups (mean difference -1.52 (95% CI -4.00 to 0.96), -2.40 (95% CI -4.99 to 0.20) and -0.81 (95% CI -3.40 to 1.77), respectively). In the subscales, we found a significantly lower T-score for anxiety problems in the hydrocortisone group (mean difference -1.26, 95% CI -2.41 to -0.12). CONCLUSION This study found high rates of behaviour problems at 2 years' CA following very preterm birth, but these problems were not associated with hydrocortisone treatment initiated between 7 and 14 days after birth in ventilated preterm infants. TRIAL REGISTRATION NUMBER NTR2768; EudraCT 2010-023777-19.
Collapse
Affiliation(s)
- Nienke Marjolein Halbmeijer
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Wes Onland
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Filip Cools
- Neonatology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Renate M Swarte
- Section of Neonatology, Pediatrics Department, Erasmus MC Sophia Children's Hospital, Rotterdam, Netherlands
| | | | - Peter Dijk
- Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, Netherlands
| | | | - Ratna N G B Tan
- Neonatology, Leiden University Medical Center, Leiden, Netherlands
| | - Thilo Mohns
- NICU, Woman-Mother-Child Center, Màxima Medical Centre, Veldhoven, Netherlands
| | - Els Bruneel
- Department of Neonatology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Arno F J van Heijst
- Department of Neonatology, Amalia Children's Hospital, Radboud Universiteit Nijmegen, Nijmegen, Netherlands
| | - Boris Kramer
- Pediatrics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Anne Debeer
- Neonatology, KU Leuven University Hospitals, Leuven, Belgium
| | - Mirjam M van Weissenbruch
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands.,Neonatology, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Yoann Marechal
- Department of Neonatology, Centre Hospitalier Universitaire de Charleroi, Charleroi, Belgium
| | - Henry Blom
- Neonatology, Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | - Katleen Plaskie
- Department of Neonatology, GZA Ziekenhuizen Campus Sint-Augustinus, Wilrijk, Belgium
| | - Martin Offringa
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Child Health Evaluative Sciences, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aleid G van Wassenaer-Leemhuis
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Anton H van Kaam
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Cornelieke S H Aarnoudse-Moens
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands .,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands.,Psychosocial Department, Universiteit van Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
37
|
Yang L, Bao Z, Zhang L, Lei X, Zhang L. Position management on pulmonary function and bronchopulmonary dysplasia in premature infants: study protocol for a randomised controlled trial. BMJ Open 2022; 12:e062291. [PMID: 36521889 PMCID: PMC9756205 DOI: 10.1136/bmjopen-2022-062291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Bronchopulmonary dysplasia (BPD) is a common disease caused by various factors and mechanisms in premature infants. Owing to lung hypoplasia and the lack of alveolar surfactants in premature infants, oxygen therapy is often needed to maintain adequate breathing. Nevertheless, prolonged oxygen therapy can easily induce BPD, and there is currently no effective treatment. Therefore, the prevention of BPD in premature infants during hospitalisation is essential. Studies have revealed that the prone position can effectively improve the oxygenation of premature infants. However, a few studies have reported whether prone positioning can improve lung function and reduce BPD incidence. This trial will determine whether the prone position, compared with the supine position, can reduce BPD incidence and improve lung function in preterm infants. METHODS AND ANALYSIS This study protocol is for a single-centre, single-blind, randomised controlled trial of the prone position in premature infants. Following daily feeding, premature infants will be placed in the lateral position for 30 min; then they will be turned to the supine position (control group) or prone position (intervention group) for 2 hours each in the morning and afternoon. Moreover, infants in both groups will be placed in the supine or lateral position alternately according to their medical needs for the remaining time. The study begins when the premature infants are stable within 5 days after admission and ends when they are discharged from the hospital or at 36 weeks postmenstrual age. The primary outcome is the survival rate without BPD. The secondary outcomes include lung function parameters and lung oxygen saturation. ETHICS AND DISSEMINATION This trial is approved by the ethics committee of the Affiliated Hospital of Southwest Medical University, (ref approval no.KY2021186). The results will be published in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ChiCTR2100049847.
Collapse
Affiliation(s)
- Liu Yang
- School of Nursing, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhengrong Bao
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lianyu Zhang
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Clinical Nursing Research Institute, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoping Lei
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Perinatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Lingping Zhang
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Clinical Nursing Research Institute, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
38
|
Bronchopulmonary dysplasia is not related to neurofilament light for neuroaxonal damage in preterm infants. Pediatr Res 2022:10.1038/s41390-022-02365-5. [PMID: 36344694 DOI: 10.1038/s41390-022-02365-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Neurofilament light (NfL) has been identified as a biomarker for neuroaxonal damage in preterm infants, but its relation with bronchopulmonary dysplasia (BPD) has not been established. We hypothesized that BPD is associated with increased NfL levels at an early stage, indicative of early neuroaxonal damage. METHODS We included preterm infants born <30 weeks of gestation for assessment of NfL levels from cord blood and blood obtained at postnatal days 3, 7, 14, and 28. We used linear regression analysis to compare NfL levels between infants with moderate/severe BPD and infants with no/mild BPD, and linear mixed model analysis to compare the effect of time on NfL levels between groups. RESULTS Sixty-seven infants with a gestational age (GA) of 27 ± 1.3 weeks were included for analysis, of whom 19 (28%) developed moderate/severe BPD. Although NfL levels were higher at every time point in infants with BPD, statistical significance was lost after adjustment for GA, small for gestational age (SGA) and intraventricular hemorrhage (IVH). Groups did not differ in NfL change over time. CONCLUSIONS The positive association between BPD and NfL in the first weeks of life could be explained by GA, SGA and IVH rather than by development of BPD. IMPACT Neurofilament light chain (NfL) is a known biomarker for neuroaxonal damage. Biomarkers for brain damage during the first weeks of life in preterm infants developing BPD are lacking. NfL levels obtained during the first weeks of life did not differ between infants with and without BPD in analyses adjusted for GA, SGA, and IVH.
Collapse
|
39
|
deRegnier RA. Disparities in Neurodevelopmental Services Between Children with Congenital Heart Disease and Children Born Very Preterm. J Pediatr 2022; 250:9-10. [PMID: 35944712 DOI: 10.1016/j.jpeds.2022.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Raye-Ann deRegnier
- Neonatology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.
| |
Collapse
|
40
|
Mohammadi A, Higazy R, Gauda EB. PGC-1α activity and mitochondrial dysfunction in preterm infants. Front Physiol 2022; 13:997619. [PMID: 36225305 PMCID: PMC9548560 DOI: 10.3389/fphys.2022.997619] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Extremely low gestational age neonates (ELGANs) are born in a relatively hyperoxic environment with weak antioxidant defenses, placing them at high risk for mitochondrial dysfunction affecting multiple organ systems including the nervous, respiratory, ocular, and gastrointestinal systems. The brain and lungs are highly affected by mitochondrial dysfunction and dysregulation in the neonate, causing white matter injury (WMI) and bronchopulmonary dysplasia (BPD), respectively. Adequate mitochondrial function is important in providing sufficient energy for organ development as it relates to alveolarization and axonal myelination and decreasing oxidative stress via reactive oxygen species (ROS) and reactive nitrogen species (RNS) detoxification. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a master regulator of mitochondrial biogenesis and function. Since mitochondrial dysfunction is at the root of WMI and BPD pathobiology, exploring therapies that can regulate PGC-1α activity may be beneficial. This review article describes several promising therapeutic agents that can mitigate mitochondrial dysfunction through direct and indirect activation and upregulation of the PGC-1α pathway. Metformin, resveratrol, omega 3 fatty acids, montelukast, L-citrulline, and adiponectin are promising candidates that require further pre-clinical and clinical studies to understand their efficacy in decreasing the burden of disease from WMI and BPD in preterm infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
| | - Estelle B. Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Estelle B. Gauda,
| |
Collapse
|
41
|
The Conflicting Role of Caffeine Supplementation on Hyperoxia-Induced Injury on the Cerebellar Granular Cell Neurogenesis of Newborn Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5769784. [PMID: 35693697 PMCID: PMC9175096 DOI: 10.1155/2022/5769784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/04/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022]
Abstract
Preterm birth disrupts cerebellar development, which may be mediated by systemic oxidative stress that damages neuronal developmental stages. Impaired cerebellar neurogenesis affects several downstream targets important for cognition, emotion, and speech. In this study, we demonstrate that oxidative stress induced with high oxygen (80%) for three or five postnatal days (P3/P5) could significantly damage neurogenesis and proliferative capacity of granular cell precursor and Purkinje cells in rat pups. Reversal of cellular neuronal damage after recovery to room air (P15) was augmented by treatment with caffeine. However, downstream transcripts important for migration and differentiation of postmitotic granular cells were irreversibly reduced by hyperoxia, without rescue by caffeine. Protective effects of caffeine in the cerebellum were limited to neuronal survival but failed to restore important transcript signatures.
Collapse
|
42
|
McKenzie K, Lynch E, Msall ME. Scaffolding Parenting and Health Development for Preterm Flourishing Across the Life Course. Pediatrics 2022; 149:186921. [PMID: 35503323 PMCID: PMC9847416 DOI: 10.1542/peds.2021-053509k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 01/21/2023] Open
Abstract
Advances in obstetrics and neonatal medicine have resulted in improved survival rates for preterm infants. Remarkably, >75% extremely (<28 weeks) preterm infants who leave the NICU do not experience major neurodevelopmental disabilities, although >50% experience more minor challenges in communication, perception, cognition, attention, regulatory, and executive function that can adversely impact educational and social function resulting in physical, behavioral, and social health issues in adulthood. Even late premature (32-36 weeks) infants have more neurodevelopmental challenges than term infants. Although early intervention and educational programs can mitigate risks of prematurity for children's developmental trajectories, restrictive eligibility requirement and limitations on frequency and intensity mean that many premature infants must "fail first" to trigger services. Social challenges, including lack of family resources, unsafe neighborhoods, structural racism, and parental substance use, may compound biological vulnerabilities, yet existing services are ill-equipped to respond. An intervention system for premature infants designed according to Life Course Health Development principles would instead focus on health optimization from the start; support emerging developmental capabilities such as self-regulation and formation of reciprocal secure early relationships; be tailored to each child's unique neurodevelopmental profile and social circumstances; and be vertically, horizontally, and longitudinally integrated across levels (individual, family, community), domains (health, education), and time. Recognizing the increased demands placed on parents, it would include parental mental health supports and provision of trauma-informed care. This developmental scaffolding would incorporate parenting, health, and developmental interventions, with the aim of improved health trajectories across the whole of the life course.
Collapse
Affiliation(s)
- Kamryn McKenzie
- University of Chicago Kennedy Research Center on Intellectual and Neurodevelopmental Disabilities, Chicago, Illinois
| | - Emma Lynch
- University of Chicago Kennedy Research Center on Intellectual and Neurodevelopmental Disabilities, Chicago, Illinois
| | - Michael E. Msall
- Address correspondence to Address correspondence to: Michael E. Msall, MD, Section of Developmental and Behavioral Pediatrics, University of Chicago Kennedy Research Center and Comer Children's Hospital, 936 East 61 St Street, Room 207, Chicago, IL 60637. E-mail:
| |
Collapse
|
43
|
Katz TA, Vliegenthart RJS, Aarnoudse-Moens CSH, Leemhuis AG, Beuger S, Blok GJ, van Brakel MJM, van den Heuvel MEN, van Kempen AAMW, Lutterman C, Rijpert M, Schiering IA, Ran NC, Visser F, Wilms J, van Kaam AH, Onland W. Severity of Bronchopulmonary Dysplasia and Neurodevelopmental Outcome at 2 and 5 Years Corrected Age. J Pediatr 2022; 243:40-46.e2. [PMID: 34929243 DOI: 10.1016/j.jpeds.2021.12.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/16/2021] [Accepted: 12/05/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate the association between bronchopulmonary dysplasia (BPD) severity and risk of neurodevelopmental impairment (NDI) at 2 years and 5 years corrected age and to examine whether this association changes over time. STUDY DESIGN This single-center retrospective cohort study included patients with a gestational age <30 weeks surviving to 36 weeks postmenstrual age, divided into groups according to BPD severity. NDI was defined as having cognitive or motor abilities below -1 SD, cerebral palsy, or a hearing or a visual impairment. The association was assessed using a multivariate logistic regression model analysis, adjusting for known confounders for NDI, and mixed-model analysis. RESULTS Of the 790 surviving infants (15% diagnosed with mild BPD, 9% with moderate BPD, and 10% with severe BPD), 88% and 82% were longitudinally assessed at 2 years and 5 years corrected age, respectively. The mixed-model analysis showed a statistically significant increase in NDI at all levels of BPD severity compared with infants with no BPD, and a 5-fold increased risk in NDI was seen from 2 years to 5 years corrected age in all degrees of BPD severity. The strength of this association between NDI and BPD severity did not change over time. CONCLUSIONS Increased BPD severity is associated with increased risk of NDI at both 2 years and 5 years corrected age. The absolute incidence of NDI increased significantly from 2 years to 5 years corrected age for all BPD severity categories, but this increased risk was similar at both time points in each category.
Collapse
Affiliation(s)
- Trixie A Katz
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, University of Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Roseanne J S Vliegenthart
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, University of Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Cornelieke S H Aarnoudse-Moens
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, University of Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Aleid G Leemhuis
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, University of Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Sabine Beuger
- Northwest Clinics, Department of Pediatrics, Alkmaar, the Netherlands
| | - Geert Jan Blok
- Northwest Clinics, Department of Pediatrics, Alkmaar, the Netherlands
| | | | | | | | - Claire Lutterman
- Department of Pediatrics, Flevoziekenhuis, Almere, the Netherlands
| | - Maarten Rijpert
- Department of Pediatrics, Zaans Medisch Centrum, Zaandam, the Netherlands
| | - Irene A Schiering
- Department of Pediatrics, Spaarne Gasthuis, Haarlem, the Netherlands
| | - Nicolien C Ran
- Department of Pediatrics, Red Cross Hospital, Beverwijk, the Netherlands
| | - Fenna Visser
- Department of Pediatrics, Amstellandziekenhuis, Amstelveen, the Netherlands
| | - Janneke Wilms
- Department of Pediatrics, Bovenij Ziekenhuis, Amsterdam, the Netherlands
| | - Anton H van Kaam
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, University of Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Wes Onland
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, University of Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
44
|
Reduction in Mechanical Ventilation: Necessary but Not Sufficient to Prevent Bronchopulmonary Dysplasia in Infants Born Extremely Preterm? J Pediatr 2022; 243:12-13. [PMID: 34974061 DOI: 10.1016/j.jpeds.2021.12.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 11/23/2022]
|
45
|
Lin H, Chen X, Ge J, Shi L, Du L, Ma X. Home oxygen use and 1-year outcome among preterm infants with bronchopulmonary dysplasia discharged from a Chinese regional NICU. Front Pediatr 2022; 10:978743. [PMID: 36160774 PMCID: PMC9500185 DOI: 10.3389/fped.2022.978743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE This study aims to compare the clinical characteristics and 1-year outcomes of preterm infants with bronchopulmonary dysplasia (BPD) who were discharged on supplemental oxygen or room air. MATERIALS AND METHODS The preterm infants (born <32 weeks' gestation, birth weight ≤1,250 g) diagnosed with BPD and admitted between January 2020 and December 2020 were enrolled. The clinical data during hospitalization were collected through the hospital's electronic record system. The outcomes after discharge were acquired from the outpatient system and through telephonic interviews. RESULTS Of the 87 preterm infants diagnosed with BPD, 81 infants survived until discharge. The 81 infants were divided into the home oxygen group (n = 29) and room air group (n = 52) according to supplemental oxygen or not at discharge. Infants in the home oxygen group were more likely to receive postnatal systemic steroids and higher ventilation settings at 36 weeks' PMA. There was one patient in each group who died before 1 year corrected age, respectively. All the infants had successfully weaned off oxygen eventually during the first year. The median duration of home oxygen therapy was 25 (7,42) days. Readmission occurred in 49 (64.5%) infants. Readmissions for infants with home oxygen were more often related to respiratory disease. In addition, wheezing disorders and home inhalation occurred more frequently in the home oxygen group (p = 0.022, p = 0.004). Although the incidence of underweight at 1 year corrected age was higher in the room air group (10.0 vs. 3.8%), there was no significant difference (p = 0.620). The rate of neurodevelopmental impairment was similar between these two groups (26.0 vs. 30.8%, p = 0.659). CONCLUSIONS It was the first study focused on preterm infants with BPD receiving home oxygen in China. Infants with home oxygen were more likely to have respiratory problems after discharge from NICU. Home oxygen use was not associated with more readmission for infants with BPD, and no difference was found in neurodevelopmental impairment and growth outcome.
Collapse
Affiliation(s)
- Huijia Lin
- Department of NICU, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuefeng Chen
- Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiajing Ge
- Department of NICU, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Liping Shi
- Department of NICU, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Lizhong Du
- Department of NICU, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaolu Ma
- Department of NICU, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
46
|
Yitayew M, Chahin N, Rustom S, Thacker LR, Hendricks-Muñoz KD. Fenton vs. Intergrowth-21st: Postnatal Growth Assessment and Prediction of Neurodevelopment in Preterm Infants. Nutrients 2021; 13:nu13082841. [PMID: 34445001 PMCID: PMC8400500 DOI: 10.3390/nu13082841] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
Although the survival rate of preterm infants has improved over the years, growth failure and associated impaired neurodevelopmental outcome remains a significant morbidity. Optimal nutrition plays an important role in achieving adequate postnatal growth. Accurate growth monitoring of preterm infants is critical in guiding nutritional protocols. Currently, there is no consensus regarding which growth assessment tool is suitable for monitoring postnatal growth of preterm infants to foster optimal neurodevelopmental outcomes while avoiding future consequences of aggressive nutritional approaches including increased risk for cardiovascular disease and metabolic syndrome. A retrospective single center cohort study was conducted to compare the performance of two growth-assessment tools, Fenton and Intergrowth-21st (IG-21st) in the classification of size at birth, identification of impaired growth and predicting neurodevelopment. A total of 340 infants with mean gestational age of 30 weeks were included. Proportion of agreement between the two tools for identification of small for gestational age (SGA) was high 0.94 (0.87, 0.1) however, agreement for classification of postnatal growth failure at discharge was moderate 0.6 (0.52, 0.69). Growth failure at discharge was less prevalent using IG-21st. There was significant association between weight-based growth failure and poor neurodevelopmental outcomes at 12 and 24 months of age.
Collapse
Affiliation(s)
- Miheret Yitayew
- Department of Pediatrics, Children’s Hospital of Richmond, Virginia Commonwealth University, Richmond, VA 23298, USA; (N.C.); (K.D.H.-M.)
- Division of Neonatal Medicine, Department of Pediatrics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence:
| | - Nayef Chahin
- Department of Pediatrics, Children’s Hospital of Richmond, Virginia Commonwealth University, Richmond, VA 23298, USA; (N.C.); (K.D.H.-M.)
| | - Salem Rustom
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.R.); (L.R.T.)
| | - Leroy R. Thacker
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA; (S.R.); (L.R.T.)
| | - Karen D. Hendricks-Muñoz
- Department of Pediatrics, Children’s Hospital of Richmond, Virginia Commonwealth University, Richmond, VA 23298, USA; (N.C.); (K.D.H.-M.)
- Division of Neonatal Medicine, Department of Pediatrics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
47
|
Association of Severe Retinopathy of Prematurity and Bronchopulmonary Dysplasia with Adverse Neurodevelopmental Outcomes in Preterm Infants without Severe Brain Injury. Brain Sci 2021; 11:brainsci11060699. [PMID: 34073292 PMCID: PMC8226991 DOI: 10.3390/brainsci11060699] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022] Open
Abstract
Although impaired neurodevelopment is strongly associated with severe brain injury, most preterm infants survive without severe brain injury. In this study, the association of impaired neurodevelopment and neonatal morbidities of preterm infants was assessed after excluding those with severe brain injury. This was a retrospective study of very low birthweight infants in a single tertiary center. After excluding infants with severe brain injury, the study population was categorized as infants without intraventricular hemorrhage (IVH) and with low-grade IVH. Neurodevelopmental outcomes at a corrected age (CA) of 18-24 months were evaluated using the Bayley Scales of Infant and Toddler Development 3rd Edition (Bayley-III). Cerebral palsy (CP), hearing impairment and blindness were also assessed and compared. Of 240 infants, 25 (11.6%) infants had combined neurodevelopmental impairment (NDI). In the multivariate analysis for combined NDI, small for gestational age (SGA) (adjusted OR 6.820, 95% confidence intervals (CI) 1.770-26.307), moderate to severe bronchopulmonary dysplasia (BPD) (aOR 3.21, 95% CI 1.032-9.999) and severe retinopathy of prematurity (ROP) (aOR 5.669, 95% CI 1.132-28.396) were associated with combined NDI. Among neonatal morbidities, moderate to severe BPD and severe ROP were associated with adverse neurodevelopmental outcomes in preterm infants without severe brain injury.
Collapse
|