1
|
Enzer KG, Baker CD, Wisniewski BL. Bronchopulmonary Dysplasia. Clin Chest Med 2024; 45:639-650. [PMID: 39069327 DOI: 10.1016/j.ccm.2024.03.007] [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] [Indexed: 07/30/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease, associated with premature birth, that arises during the infantile period. It is an evolving disease process with an unchanged incidence due to advancements in neonatal care which allow for the survival of premature infants of lower gestational ages and birth weights. Currently, there are few effective interventions to prevent BPD. However, careful attention to BPD phenotypes and comprehensive care provided by an interdisciplinary team have improved care. Interventions early in the disease course hold promise for improving long-term survival and outcomes in adulthood for this high-risk population.
Collapse
Affiliation(s)
- Katelyn G Enzer
- Department of Pediatrics, Section of Pulmonary and Sleep Medicine, University of Colorado School of Medicine, 13123 East 16th Avenue Box B-395, Aurora, CO 80045, USA.
| | - Christopher D Baker
- Department of Pediatrics, Section of Pulmonary and Sleep Medicine, University of Colorado School of Medicine, 13123 East 16th Avenue Box B-395, Aurora, CO 80045, USA
| | - Benjamin L Wisniewski
- Department of Pediatrics, Section of Pulmonary and Sleep Medicine, University of Colorado School of Medicine, 13123 East 16th Avenue Box B-395, Aurora, CO 80045, USA
| |
Collapse
|
2
|
O'Shea TM, Jensen ET, Yi JX, Lester B, Aschner JL, Stroustrup A, Zhang X, McGrath M, Sanderson K, Joseph RM, Singh R, Thompson AL, Hofheimer J, Vohr B, McGowan E, Santos H, Fry RC. Association of Growth During Infancy with Neurodevelopment and Obesity in Children Born Very Preterm: The Environmental Influences on Child Health Outcomes Cohort. J Pediatr 2024; 271:114050. [PMID: 38641165 PMCID: PMC11239281 DOI: 10.1016/j.jpeds.2024.114050] [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: 12/30/2023] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVE To evaluate associations between change in weight z score after neonatal intensive care unit (NICU) discharge and neurodevelopmental outcomes and obesity at 12-48 months of age among individuals born very preterm. STUDY DESIGN This secondary analysis used data from infants born very preterm participating in the Environmental influences on Child Health Outcomes cohort (n = 1400). Growth during infancy was calculated as change in weight z score between NICU discharge and follow-up at a mean of 27 months of age. Very low weight gain was defined as a change in weight z score <-1.67; very high weight gain was a change in weight z score >1.67. Neurodevelopmental outcomes included the Bayley Scales of Infant and Toddler Development, Child Behavior Checklist 1.5-5 years, and Modified Checklist for Autism in Toddlers. Multivariable linear regression was used to estimate associations between increase in weight z score and neurodevelopmental outcomes. RESULTS Very low weight gain between NICU discharge and follow-up (experienced by 6.4% of participants) was associated with lower scores on cognitive (adjusted mean difference: -4.26; 95% CI: -8.55, -0.04) and language (adjusted mean difference: -4.80; 95% CI: -9.70, -0.11) assessments. Very high weight gain (experienced by 13.6% of participants) was associated with an increased obesity risk (adjusted relative risk: 6.20; 95% CI: 3.99, 9.66) but not with neurodevelopmental outcomes. CONCLUSIONS Very high weight gain in the first 12-48 months after NICU discharge was associated with a higher risk of obesity at follow-up; very low weight gain was associated with lower scores on cognitive and language assessments.
Collapse
Affiliation(s)
- T Michael O'Shea
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC.
| | - Elizabeth T Jensen
- Department of Epidemiology and Prevention, Wake Forest University, Winston-Salem, NC
| | - Joe X Yi
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Barry Lester
- Department of Psychiatry and Human Behavior, The Alpert Medical School of Brown University, Providence, RI
| | - Judy L Aschner
- Pediatrics, Albert Einstein College of Medicine, Bronx, NY; Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ
| | - Annemarie Stroustrup
- Pediatrics, Northwell Health, Cohen Children's Medical Center and the Departments of Pediatrics and Occupational Medicine, Epidemiology & Prevention, Zucker School of Medicine at Hofstra, New Hyde Park, NY
| | - Xueying Zhang
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Monica McGrath
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD
| | - Keia Sanderson
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC; Department of Internal Medicine, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Robert M Joseph
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Rachana Singh
- Department of Pediatrics, Tufts University School of Medicine, Boston, MA
| | - Amanda L Thompson
- Department of Anthropology, University of North Carolina, Chapel Hill, NC
| | - Julie Hofheimer
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Betty Vohr
- Department of Pediatrics, Brown University, Providence, RI
| | | | - Hudson Santos
- University of Miami School of Nursing and Health Studies, Coral Gables, FL
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina, Chapel Hill, NC
| |
Collapse
|
3
|
House M, Lagoski M, DiGeronimo R, Eldredge LC, Manimtim W, Baker CD, Coghill C, Fernandes CJ, Griffiths P, Ibrahim J, Kielt MJ, Lagatta J, Machry JS, Mikhael M, Vyas-Read S, Weems MF, Yallapragada SG, Murthy K, Nelin LD. Interdisciplinary clinical bronchopulmonary dysplasia programs: development, evolution, and maturation. J Perinatol 2024:10.1038/s41372-024-02049-4. [PMID: 39020027 DOI: 10.1038/s41372-024-02049-4] [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: 01/24/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024]
Abstract
Multidisciplinary bronchopulmonary dysplasia (BPD) programs provide improved and consistent medical management, care of the developing infant, family support, and smoother transitions in care resulting in improved survival, pulmonary, and extra-pulmonary outcomes. This review summarizes the benefits of interdisciplinary BPD management, as well as strategies for initial programmatic development, program growth, and maintenance at centers across the United States factoring in institutional, provider, and parent reported goals that were derived from a consensus conference on BPD management.
Collapse
Affiliation(s)
- Melissa House
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Megan Lagoski
- Ann & Robert H. Lurie Children's Hospitals of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert DiGeronimo
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - Laurie C Eldredge
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - Winston Manimtim
- Divison of Neonatology, Children's Mercy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Christopher D Baker
- Section of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Carl Coghill
- Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caraciolo J Fernandes
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - John Ibrahim
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew J Kielt
- Comprehensive Center for BPD, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Joana Silva Machry
- Division of Neonatology, Maternal Fetal & Neonatal Institute at Johns Hopkins All Children's Hospital St. Petersburg, St. Petersburg, FL, USA
| | - Michel Mikhael
- Division of Neonatology, Children's Hospital of Orange County, Orange, CA, USA
| | - Shilpa Vyas-Read
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mark F Weems
- Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Karna Murthy
- Ann & Robert H. Lurie Children's Hospitals of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Children's Hospitals Neonatal Consortium, Dover, DE, USA
| | - Leif D Nelin
- Comprehensive Center for BPD, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
4
|
Collaco JM, Eldredge LC, McGrath-Morrow SA. Long-term pulmonary outcomes in BPD throughout the life-course. J Perinatol 2024:10.1038/s41372-024-01957-9. [PMID: 38570594 DOI: 10.1038/s41372-024-01957-9] [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: 02/10/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Respiratory disease is one of the most common complications of preterm birth. Survivors of prematurity have increased risks of morbidities and mortalities independent of prematurity, and frequently require multiple medications, home respiratory support, and subspecialty care to maintain health. Although advances in neonatal and pulmonary care have improved overall survival, earlier gestational age, lower birth weight, chorioamnionitis and late onset sepsis continue to be major factors in the development of bronchopulmonary dysplasia. These early life events associated with prematurity can have respiratory consequences that persist into adulthood. Furthermore, after initial hospital discharge, air pollution, respiratory tract infections and socioeconomic status may modify lung growth trajectories and influence respiratory outcomes in later life. Given that the incidence of respiratory disease associated with prematurity remains stable or increased, there is a need for pediatric and adult providers to be familiar with the natural history, manifestations, and common complications of disease.
Collapse
Affiliation(s)
- Joseph M Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Laurie C Eldredge
- Division of Pediatric Pulmonology, Seattle Children's Hospital, Seattle, WA, USA
| | - Sharon A McGrath-Morrow
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Levin JC, Sheils CA, Hayden LP. Pre-Flight Hypoxemia Challenge Testing in Bronchopulmonary Dysplasia. Pediatrics 2023; 152:e2022061001. [PMID: 37503557 PMCID: PMC10389772 DOI: 10.1542/peds.2022-061001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Former premature infants with bronchopulmonary dysplasia (BPD) are at risk for hypoxemia during air travel, but it is unclear until what age. We aimed to determine pass rates for high altitude simulation testing (HAST) by age in children with BPD and identify risks for failure. METHODS Retrospective, observational analysis of HAST in children with BPD at Boston Children's Hospital, using interval censoring to estimate the time-to-event curve of first pass. Curves were stratified by neonatal risk factors. Pass was considered lowest Spo2 ≥ 90%, or ≥94% for subjects with ongoing pulmonary hypertension (PH). RESULTS Ninety four HAST studies were analyzed from 63 BPD subjects; 59 studies (63%) were passed. At 3 months corrected gestational age (CGA), 50% of subjects had passed; at 6 months CGA, 67% has passed; at 12 and 18 months CGA, 72% had passed; and at 24 months CGA, 85% had passed. Neonatal factors associated with delayed time-to-pass included postnatal corticosteroid use, respiratory support at NICU discharge, and tracheostomy. BPD infants who did not require respiratory support at 36 weeks were likely to pass (91%) at 6 months CGA. At 24 months, children least likely to pass included those with a history of PH (63%) and those discharged from the NICU with oxygen or respiratory support (71%). CONCLUSIONS Children with BPD on respiratory support at 36 weeks should be considered for preflight hypoxemia challenges through at least 24 months CGA, and longer if they had PH or went home from NICU on respiratory support.
Collapse
Affiliation(s)
- Jonathan C. Levin
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
- Division of Pulmonary Medicine, Boston Children’s Hospital, Boston, Massachusetts
| | - Catherine A. Sheils
- Division of Pulmonary Medicine, Boston Children’s Hospital, Boston, Massachusetts
| | - Lystra P. Hayden
- Division of Pulmonary Medicine, Boston Children’s Hospital, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
6
|
Stroustrup A, Zhang X, Spear E, Bandyopadhyay S, Narasimhan S, Meher AK, Choi J, Qi G, Poindexter BB, Teitelbaum SL, Andra SS, Gennings C, Aschner JL. Phthalate exposure in the neonatal intensive care unit is associated with development of bronchopulmonary dysplasia. ENVIRONMENT INTERNATIONAL 2023; 178:108117. [PMID: 37517179 PMCID: PMC10581357 DOI: 10.1016/j.envint.2023.108117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
OBJECTIVE Bronchopulmonary dysplasia (BPD) is a serious yet common morbidity of preterm birth. Although prior work suggests a possible role for phthalate exposure in the development of BPD, no study has rigorously evaluated this. Our objective was to determine whether hospital-based phthalate exposure is associated with the development of BPD and to identify developmental windows sensitive to exposure. STUDY DESIGN This is a prospective multicenter cohort study of 360 preterm infants born at 23-33 weeks gestation participating in the Developmental Impact of NICU Exposures (DINE) cohort. 939 urine specimens collected during the NICU stay were analyzed for biomarkers of phthalate exposure by liquid chromatography with tandem mass spectrometry. The modified Shennan definition was used to diagnose bronchopulmonary dysplasia. Reverse distributed-lag modeling identified developmental windows sensitive to specific phthalate exposure, controlling for relevant covariates including sex and respiratory support. RESULTS Thirty-five percent of participants were diagnosed with BPD. Exposure to specific phthalate mixtures at susceptible points in preterm infant development are associated with later diagnosis of BPD in models adjusted for use of respiratory support. The weighted influence of specific phthalate metabolites in the mixtures varied by sex. Metabolites of di(2-ethylhexyl) phthalate, a phthalate previously linked to neonatal respiratory support equipment, drove this association, particularly among female infants, at 26- to 30-weeks post-menstrual age. CONCLUSIONS This is the largest and only multi-site study of NICU-based phthalate exposure and clinical impact yet reported. In well-constructed models accounting for infant sex and respiratory support, we found a significant positive association between ultimate diagnosis of BPD and prior exposure to phthalate mixtures with DEHP predominance at 26- to 30-weeks PMA or 34-36-weeks PMA. This information is critically important as it identifies a previously unrecognized and modifiable contributing factor to BPD.
Collapse
Affiliation(s)
- Annemarie Stroustrup
- Department of Pediatrics and Department of Occupational Medicine, Epidemiology and Prevention, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, New Hyde Park, NY, United States; Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Xueying Zhang
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Emily Spear
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sanjukta Bandyopadhyay
- Clinical and Translational Science Institute, University of Rochester, Rochester, NY, United States
| | - Srinivasan Narasimhan
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anil K Meher
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jaeun Choi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Gao Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Brenda B Poindexter
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Susan L Teitelbaum
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Syam S Andra
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chris Gennings
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Judy L Aschner
- Department of Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ, United States; Department of Pediatrics and Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
7
|
O'Shea TM, McGrath M, Aschner JL, Lester B, Santos HP, Marsit C, Stroustrup A, Emmanuel C, Hudak M, McGowan E, Patel S, Fry RC. Environmental influences on child health outcomes: cohorts of individuals born very preterm. Pediatr Res 2023; 93:1161-1176. [PMID: 35948605 PMCID: PMC9363858 DOI: 10.1038/s41390-022-02230-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/27/2022] [Accepted: 07/19/2022] [Indexed: 12/05/2022]
Abstract
The National Institutes of Health's Environmental influences on Child Health Outcomes (ECHO) Program was designed to address solution-oriented research questions about the links between children's early life environment and their risks of pre-, peri-, and post-natal complications, asthma, obesity, neurodevelopmental disorders, and positive health. Children born very preterm are at increased risk for many of the outcomes on which ECHO focuses, but the contributions of environmental factors to this risk are not well characterized. Three ECHO cohorts consist almost exclusively of individuals born very preterm. Data provided to ECHO from cohorts can be used to address hypotheses about (1) differential risks of chronic health and developmental conditions between individuals born very preterm and those born at term; (2) health disparities across social determinants of health; and (3) mechanisms linking early-life exposures and later-life outcomes among individuals born very preterm. IMPACT: The National Institutes of Health's Environmental Influences on Child Health Outcomes Program is conducting solution-oriented research on the links between children's environment and health. Three ECHO cohorts comprise study participants born very preterm; these cohorts have enrolled, to date, 1751 individuals born in 14 states in the U.S. in between April 2002 and March 2020. Extensive data are available on early-life environmental exposures and child outcomes related to neurodevelopment, asthma, obesity, and positive health. Data from ECHO preterm cohorts can be used to address questions about the combined effects of preterm birth and environmental exposures on child health outcomes.
Collapse
Affiliation(s)
- T Michael O'Shea
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| | - Monica McGrath
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Judy L Aschner
- Department of Pediatrics, Joseph M. Sanzari Children's Hospital at Hackensack University Medical Center, Hackensack, NJ, USA
- Department of Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Barry Lester
- Department of Pediatrics, Women & Infants Hospital, Brown University, Providence, RI, USA
- Brown Center for the Study of Children at Risk, Warren Alpert Medical School of Brown University, Women & Infants Hospital, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Hudson P Santos
- Biobehavioral Laboratory, School of Nursing, The University of North Carolina, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina, Chapel Hill, NC, USA
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Annemarie Stroustrup
- Departments of Pediatrics and Occupational Medicine, Epidemiology and Prevention, Zucker School of Medicine at Hofstra, Northwell Health, Cohen Children's Medical Center, New Hyde Park, NY, USA
| | - Crisma Emmanuel
- Biobehavioral Laboratory, School of Nursing, The University of North Carolina, Chapel Hill, NC, USA
| | - Mark Hudak
- Department of Pediatrics, University of Florida College of Medicine - Jacksonville, Jacksonville, FL, USA
| | - Elisabeth McGowan
- Women & Infants Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Simran Patel
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
8
|
Lagatta JM, Zhang L, Yan K, Dawson S, Msall ME, Ambalavanan N, Brousseau DC. Prospective Risk Stratification Identifies Healthcare Utilization Associated with Home Oxygen Therapy for Infants with Bronchopulmonary Dysplasia. J Pediatr 2022; 251:105-112.e1. [PMID: 35934128 DOI: 10.1016/j.jpeds.2022.07.040] [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: 01/20/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To test whether prospective classification of infants with bronchopulmonary dysplasia identifies lower-risk infants for discharge with home oxygen who have fewer rehospitalizations by 1 year after neonatal intensive care unit discharge. STUDY DESIGN This is a prospective single-center cohort that included infants from 2016 to 2019 with bronchopulmonary dysplasia, defined as receiving respiratory support at 36 weeks of postmenstrual age. "Lower-risk" infants were receiving ≤2 L/min nasal cannula flow, did not have pulmonary hypertension or airway comorbidities, and had blood gas partial pressure of carbon dioxide <70 mm Hg. We compared 3 groups by discharge status: lower-risk room air, lower-risk home oxygen, and higher-risk home oxygen. The primary outcome was rehospitalization at 1 year postdischarge, and the secondary outcomes were determined by the chart review and parent questionnaire. RESULTS Among 145 infants, 32 (22%) were lower-risk discharged in room air, 49 (32%) were lower-risk using home oxygen, and 64 (44%) were higher-risk. Lower-risk infants using home oxygen had rehospitalization rates similar to those of lower-risk infants on room air (18% vs 16%, P = .75) and lower rates than higher-risk infants (39%, P = .018). Lower-risk infants using home oxygen had more specialty visits (median 10, IQR 7-14 vs median 6, IQR 3-11, P = .028) than those on room air. Classification tree analysis identified risk status as significantly associated with rehospitalization, along with distance from home to hospital, inborn, parent-reported race, and siblings in the home. CONCLUSIONS Prospectively identified lower-risk infants discharged with home oxygen had fewer rehospitalizations than higher-risk infants and used more specialty care than lower-risk infants discharged in room air.
Collapse
Affiliation(s)
- Joanne M Lagatta
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI.
| | - Liyun Zhang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Ke Yan
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Sara Dawson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Michael E Msall
- Department of Pediatrics, University of Chicago, Chicago, IL
| | | | - David C Brousseau
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
9
|
Cheema Z, Kwinta P, Moreira A, Tovar M, Mustafa SB. Big Data for Tiny Patients: A Precision Medicine Approach to Bronchopulmonary Dysplasia. Pediatr Ann 2022; 51:e396-e404. [PMID: 36215088 DOI: 10.3928/19382359-20220803-06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease of extreme prematurity. Despite more than 50 years of research, current treatments are ineffective, and clinicians are largely unable to accurately predict which neonates the condition will develop in. A deeper understanding of the molecular mechanisms underlying the characteristic arrest in lung development are warranted. Integrating high-fidelity technology from precision medicine approaches may fill this gap and provide the tools necessary to identify biomarkers and targetable pathways. In this review, we describe insights garnered from current studies using omics for BPD prediction and stratification. We conclude by describing novel programs that will integrate multi-omics in efforts to better understand and treat the pathogenesis of BPD. [Pediatr Ann. 2022;51(10):e396-e404.].
Collapse
|
10
|
Torres J, Malla SD, Silveira V, Mainero L, Czeisler C, Díaz-Rossello JL, Maccarrone A, Medoro A, Sanchez P, Blasina F, Otero JJ. Local clinical informatics investments are required for in silico biomarker generation across the globe: lessons learned from a secondary analysis of the PROP trial. JOURNAL OF GLOBAL HEALTH REPORTS 2022. [DOI: 10.29392/001c.37938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Background Advances in statistical modelling and machine learning approaches, which can be deployed locally using open source programming languages, represent a unique opportunity to improve workflows and lower costs in health care across the globe through the creation of in silico biomarkers. The goal of this study was to extract meaningful data from the publicly available Prematurity and Respiratory Outcomes Program (PROP) trial data that could help generate useful clinical diagnostic aids with minimal cost for deployment in global healthcare settings. Methods A cluster analysis of the PROP dataset was conducted. We generated a simple model using an open-source software platform that generates a growth prediction of patients born less than 30 weeks. We then obtained validation data from a Uruguayan hospital to test the capacity for deployment of the models. Results Analysis revealed two main clusters of patients in the trial, with differentiation mainly based on the clinical and anthropomorphic measurements of birth gestational age, birth weight, and head circumference. The anthropometric measurements of daily weight, birth weight, head circumference, and birth gestational age were highly correlated with respiratory dysfunction and co-morbidities We note that deviation from this predicted growth curve in PROP patients was associated with culture-proven sepsis, and may represent a more sensitive anthropomorphic biomarker than the weight percentile systems routinely used globally such as Fenton curves. We found that early deviation from our projected growth model was highly associated with patient fatality. However, over long-term predictions, models trained on PROP clinical trial patients showed significantly more error in the Uruguayan patients. Conclusions Although these prediction models built upon PROP data were not generalizable to Uruguayan patients, our data suggest that prediction models using simple anthropomorphic measurements, if trained on local patients, may be able to provide value as a low-cost in silico biomarker. We concluded that local investment in clinical informatics infrastructure is needed to train models based on locally extracted clinical data.
Collapse
Affiliation(s)
- Juliet Torres
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Valentina Silveira
- Division of Neonatology, Department of Pediatrics, Hospital de Clinicas, Montevideo, Uruguay
| | - Luis Mainero
- Pan-American Health Organization, Montevideo, Uruguay
| | - Catherine Czeisler
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - José L. Díaz-Rossello
- Division of Neonatology, Department of Pediatrics, Hospital de Clinicas, Montevideo, Uruguay
| | - Alejandro Maccarrone
- Servicio de Neonatologia, Hospital Provincial del Centenario, Rosario, Santa Fe, Argentina
| | - Alexandria Medoro
- Division of Neonatology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Pablo Sanchez
- Division of Neonatology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Fernanda Blasina
- Division of Neonatology, Department of Pediatrics, Hospital de Clinicas, Montevideo, Uruguay
| | - Jose J. Otero
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
11
|
McDavid A, Laniewski N, Grier A, Gill AL, Kessler HA, Huyck H, Carbonell E, Holden-Wiltse J, Bandyopadhyay S, Carnahan J, Dylag AM, Topham DJ, Falsey AR, Caserta MT, Pryhuber GS, Gill SR, Scheible KM. Aberrant newborn T cell and microbiota developmental trajectories predict respiratory compromise during infancy. iScience 2022; 25:104007. [PMID: 35310935 PMCID: PMC8931366 DOI: 10.1016/j.isci.2022.104007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/30/2021] [Accepted: 02/25/2022] [Indexed: 11/11/2022] Open
Abstract
Neonatal immune-microbiota co-development is poorly understood, yet age-appropriate recognition of - and response to - pathogens and commensal microbiota is critical to health. In this longitudinal study of 148 preterm and 119 full-term infants from birth through one year of age, we found that postmenstrual age or weeks from conception is a central factor influencing T cell and mucosal microbiota development. Numerous features of the T cell and microbiota functional development remain unexplained; however, by either age metric and are instead shaped by discrete perinatal and postnatal events. Most strikingly, we establish that prenatal antibiotics or infection disrupt the normal T cell population developmental trajectory, influencing subsequent respiratory microbial colonization and predicting respiratory morbidity. In this way, early exposures predict the postnatal immune-microbiota axis trajectory, placing infants at later risk for respiratory morbidity in early childhood.
Collapse
Affiliation(s)
- Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Nathan Laniewski
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Alex Grier
- Genomics Research Center, University of Rochester, Rochester, NY, USA
| | - Ann L. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Haeja A. Kessler
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Heidie Huyck
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | | | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Sanjukta Bandyopadhyay
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Jennifer Carnahan
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Andrew M. Dylag
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - David J. Topham
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Ann R. Falsey
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Mary T. Caserta
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | | | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
12
|
Bogdan RD, Bohiltea RE, Toma AI. Respiratory Follow Up of the Premature Neonates-Rationale and Practical Issues. J Clin Med 2022; 11:1746. [PMID: 35330070 PMCID: PMC8955296 DOI: 10.3390/jcm11061746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/13/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
The aim of the review was to present the state of knowledge about the respiratory pathology in former premature neonates (children that were born preterm-before 37 weeks of gestation-and are examined and evaluated after 40 weeks corrected age) other than chronic lung disease, in order to provide reasons for a respiratory follow-up program for this category of patients. After a search of the current evidence, we found that premature infants are prone to long-term respiratory consequences due to several reasons: development of the lung outside of the uterus, leading to dysmaturation of the structures, pulmonary pathology due to immaturity, infectious agents or mechanical ventilation and deficient control of breathing. The medium- to long-term respiratory consequences of being born before term are represented by an increased risk of respiratory infections (especially viral) during the first years of life, a risk of recurrent wheezing and asthma and a decrease in pulmonary volumes and airway flows. Late preterm infants have risks of pulmonary long-term consequences similar to other former premature infants. Due to all the above risks, premature neonates should be followed in an organized fashion, being examined at regular time intervals from discharge from the maternity hospital until adulthood-this could lead to an early detection of the risks and preventive therapies in order to improve their prognosis and assure a normal and productive life. The difficulties related to establishing such programs are represented by the insufficient standardization of the data gathering forms, clinical examinations and lung function tests, but it is our belief that if more premature infants are followed, the experience will allow standards to be established in these fields and the methods of data gathering and evaluation to be unified.
Collapse
Affiliation(s)
- Raluca Daniela Bogdan
- Pediatrics Department, Medicover Hospital, Str. Pechea No. 8, Sector 1, 031056 Bucharest, Romania;
| | - Roxana Elena Bohiltea
- Department of Obstetrics and Gynecology, Carol Davila University of Medicine and Pharmacy, Bd Eroii Sanitari Nr 8, 050471 Bucharest, Romania
| | - Adrian Ioan Toma
- Neonatology Department, Life Memorial Hospital, Calea Grivitei No. 365, Sector 1, 010719 Bucharest, Romania
- Faculty of Medicine, University “Titu Maiorescu”, Str. Gh Petrascu 67, Sector 3, 031593 Bucharest, Romania
| |
Collapse
|
13
|
Voynow JA, Feng R, Ren CL, Dylag AM, Kemp JS, McDowell K, Sharp J, Moore PE, Eichenwald E, Panitch H, Clem C, Johnson R, Davis SD. Pulmonary function tests in extremely low gestational age infants at one year of age. Pediatr Pulmonol 2022; 57:435-447. [PMID: 34779149 DOI: 10.1002/ppul.25757] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 11/08/2022]
Abstract
RATIONALE Identifying neonatal and post-discharge exposures among extremely low gestational age newborns (ELGANs) that drive increased pulmonary morbidity and abnormal lung function at 1 year of age proves challenging. OBJECTIVE The NIH-sponsored Prematurity and Respiratory Outcomes Program (PROP), evaluated infant pulmonary function tests (iPFTs) at 1 year corrected age to determine which demographic and clinical factors are associated with abnormal lung function. METHODS iPFTs were performed on a PROP subcohort of 135 participants following Institutional Review Board (IRB)-approved written consent. Demographic data, Neonatal Intensive Care Unit (NICU) clinical care, and post-NICU exposures were analyzed for association with iPFTs. MAIN RESULTS A significant decrease in forced expiratory volume at 0.5 s (FEV0.5 ) and/or forced expiratory flows at 75% of forced vital capacity (FEF75 ), were associated with male sex and African American race. Clinical factors including longer duration of ventilatory support, exposure to systemic steroids, and weight less than the 10th percentile at 36 weeks postmenstrual age were also associated with airflow obstruction, whereas supplemental oxygen requirement and bronchopulmonary dysplasia were not. Additionally, the need for respiratory medications, technology, or hospitalizations during the first year, ascertained by a quarterly survey, were the only post-NICU factors associated with decreased FEV0.5 and FEF75 . Only 7% of infants had reversible airflow obstruction. CONCLUSIONS Neonatal demographic factors, respiratory support in the NICU, and a history of greater post-NICU medical utilization for respiratory disease had the strongest association with lower lung function at 1 year in ELGANs.
Collapse
Affiliation(s)
- Judith A Voynow
- Division of Pediatric Pulmonology, Children's Hospital of Richmond at VCU, Richmond, Virginia, USA
| | - Rui Feng
- Department of Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Clement L Ren
- Division of Pediatric Pulmonology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Pediatric Pulmonology, University of Rochester, Rochester, New York, USA.,Division of Pediatric Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, USA
| | - Andrew M Dylag
- Division of Neonatology, University of Rochester, Rochester, New York, USA
| | - James S Kemp
- Division of Pediatric Pulmonology, Washington University Medical Center, St. Louis, Missouri, USA
| | - Karen McDowell
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jack Sharp
- Division of Pediatric Pulmonology, Duke University Medical Center, Durham, North Carolina, USA.,Division of Pediatric Pulmonology, Texas Children's Hospital, Woodlands, Texas, USA
| | - Paul E Moore
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eric Eichenwald
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Division of Neonatology, University of Texas Health Science Center- Houston, Houston, Texas, USA
| | - Howard Panitch
- Division of Pediatric Pulmonology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Charles Clem
- Division of Pediatric Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, USA
| | - Robin Johnson
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Stephanie D Davis
- Division of Pediatric Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, USA.,Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
14
|
Cristea AI, Ren CL, Amin R, Eldredge LC, Levin JC, Majmudar PP, May AE, Rose RS, Tracy MC, Watters KF, Allen J, Austin ED, Cataletto ME, Collaco JM, Fleck RJ, Gelfand A, Hayes D, Jones MH, Kun SS, Mandell EW, McGrath-Morrow SA, Panitch HB, Popatia R, Rhein LM, Teper A, Woods JC, Iyer N, Baker CD. Outpatient Respiratory Management of Infants, Children, and Adolescents with Post-Prematurity Respiratory Disease: An Official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2021; 204:e115-e133. [PMID: 34908518 PMCID: PMC8865713 DOI: 10.1164/rccm.202110-2269st] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Premature birth affects millions of neonates each year, placing them at risk for respiratory disease due to prematurity. Bronchopulmonary dysplasia is the most common chronic lung disease of infancy, but recent data suggest that even premature infants who do not meet the strict definition of bronchopulmonary dysplasia can develop adverse pulmonary outcomes later in life. This post-prematurity respiratory disease (PPRD) manifests as chronic respiratory symptoms, including cough, recurrent wheezing, exercise limitation, and reduced pulmonary function. This document provides an evidence-based clinical practice guideline on the outpatient management of infants, children, and adolescents with PPRD. Methods: A multidisciplinary panel of experts posed questions regarding the outpatient management of PPRD. We conducted a systematic review of the relevant literature. The Grading of Recommendations, Assessment, Development, and Evaluation approach was used to rate the quality of evidence and the strength of the clinical recommendations. Results: The panel members considered the strength of each recommendation and evaluated the benefits and risks of applying the intervention. In formulating the recommendations, the panel considered patient and caregiver values, the cost of care, and feasibility. Recommendations were developed for or against three common medical therapies and four diagnostic evaluations in the context of the outpatient management of PPRD. Conclusions: The panel developed recommendations for the outpatient management of patients with PPRD on the basis of limited evidence and expert opinion. Important areas for future research were identified.
Collapse
|
15
|
Collaco JM, McGrath-Morrow SA. Bronchopulmonary dysplasia as a determinant of respiratory outcomes in adult life. Pediatr Pulmonol 2021; 56:3464-3471. [PMID: 33730436 PMCID: PMC8446084 DOI: 10.1002/ppul.25301] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/05/2021] [Accepted: 01/22/2021] [Indexed: 12/13/2022]
Abstract
Respiratory disease is unfortunately common in preterm infants with the archetype being bronchopulmonary dysplasia (BPD). BPD affects approximately 50,000 preterm infants in the U.S. annually with substantial morbidity and mortality related to its pathology (alveolar, airway, and pulmonary vasculature maldevelopment). Predicting the likelihood and severity of chronic respiratory disease in these children as they age is difficult and compounded by the lack of consistent phenotyping. Barriers to understanding the actual scope of this problem include few longitudinal studies, information limited by small retrospective studies and the ever-changing landscape of therapies in the NICU that affect long-term respiratory outcomes. Thus, the true burden of adult respiratory disease caused by premature birth is currently unknown. Nevertheless, limited data suggest that a substantial percentage of children with a history of BPD have long-term respiratory symptoms and persistent airflow obstruction associated with altered lung function trajectories into adult life. Small airway disease with variable bronchodilator responsiveness, is the most common manifestation of lung dysfunction in adults with a history of BPD. The etiology of this is unclear however, developmental dysanapsis may underlie the airflow obstruction in some adults with a history of BPD. This type of flow limitation resembles that of aging adults with chronic obstructive lung disease with no history of smoking. It is also unclear whether lung function abnormalities in people with a history of BPD are static or if these individuals with BPD have a more accelerated decline in lung function as they age compared to controls. While some of the more significant mediators of lung function, such as tobacco smoke and respiratory infections have been identified, more work is necessary to identify the best means of preserving lung function for individuals born prematurely throughout their lifespan.
Collapse
Affiliation(s)
- Joseph M Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sharon A McGrath-Morrow
- Division of Pulmonary and Sleep, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Mammel D, Kemp J. Prematurity, the diagnosis of bronchopulmonary dysplasia, and maturation of ventilatory control. Pediatr Pulmonol 2021; 56:3533-3545. [PMID: 34042316 DOI: 10.1002/ppul.25519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 11/10/2022]
Abstract
Infants born before 32 weeks gestational age and receiving respiratory support at 36 weeks postmenstrual age (PMA) are diagnosed with bronchopulmonary dysplasia (BPD). This label suggests that their need for supplemental oxygen (O2 ) is primarily due to acquired dysplasia of airways and airspaces, and that the supplemental O2 is treating residual parenchymal lung disease. However, emerging evidence suggests that immature ventilatory control may also contribute to the need for supplemental O2 at 36 weeks PMA. In all newborns, maturation of ventilatory control continues ex utero and is a plastic process. Among premature infants, supplemental O2 mitigates the hypoxemic effects of delayed maturation of ventilatory control, as well as reduces the duration and frequency of periodic breathing events. Nevertheless, prematurity is associated with altered and occasionally aberrant maturation of ventilatory control. Infants born prematurely, with or without a diagnosis of BPD, are more prone to long-lasting effects of dysfunctional ventilatory control. This review addresses normal and abnormal maturation of ventilatory control and suggests how aberrant maturation complicates assigning the diagnosis of BPD. Greater awareness of the interaction between parenchymal lung disease and delayed maturation of ventilatory control is essential to understanding why a given premature infant requires and is benefitting from supplemental O2 at 36 weeks PMA.
Collapse
Affiliation(s)
- Daniel Mammel
- Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, USA
| | - James Kemp
- Department of Pediatrics, Allergy and Pulmonary Medicine, Division of Allergy, Immunology, and Pulmonary Medicine, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, USA
| |
Collapse
|
17
|
El-Khuffash A, Lewandowski AJ, Jain A, Hamvas A, Singh GK, Levy PT. Cardiac Performance in the First Year of Age Among Preterm Infants Fed Maternal Breast Milk. JAMA Netw Open 2021; 4:e2121206. [PMID: 34448867 PMCID: PMC8397926 DOI: 10.1001/jamanetworkopen.2021.21206] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE There is a beneficial association between human breast milk exposure in the neonatal period and cardiac mechanics in adults who were born preterm. It is unknown whether this benefit is apparent in infants in the first year of age. OBJECTIVE To test the hypothesis that higher consumption of mother's own milk in preterm infants is associated with enhanced cardiac performance during the first year of age. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study of cardiac and nutritional data at an academic medical center included 80 individuals born preterm and 100 individuals in the control group born full-term. All births were between 2011 and 2013. Two-dimensional echocardiograms were performed at 32 weeks' and 36 weeks' postmenstrual age and at 1 year's corrected age in individuals born preterm and at 1 month and 1 year of age in individuals born full-term. Statistical analysis was performed from January to May 2021. EXPOSURES Consumption of mother's own milk. MAIN OUTCOMES AND MEASURES Main study outcomes included echocardiography measures of right and left ventricle longitudinal strain (function), left ventricle mass index and right ventricular areas (morphology), and pulmonary vascular resistance (pulmonary hemodynamics) at age 1 year. RESULTS Of 180 infants included in the study, 97 (54%) were Black infants and 89 (49%) were female infants. Among the 80 infants born in the preterm cohort, 43 (54%) were female infants and 43 (54%) were Black infants. The median gestational age at birth of the preterm infants was 27.0 weeks (interquartile range, 26.0-28.0 weeks) and the median birth weight was 960 g (interquartile range, 800-1138). For each week of exposure to mother's own milk, preterm infants had greater magnitudes of right ventricular strain (eg, right longitudinal strain: β, 0.021; 95% CI, 0.002-0.041; P < .001) and left ventricular strain (eg, left longitudinal strain: β, 0.065; 95% CI, 0.049-0.080; P = .01), larger right ventricle areas (eg, systolic area: β, 0.026; 95% CI, 0.011-0.042; P = .009), larger left ventricle mass index (β, 0.045; 95% CI, 0.024-0.073; P = .003), and decreased pulmonary vascular resistance (eg, pulmonary artery acceleration time: β, 0.041; 95% CI, 0.018-0.063; P < .001) at 1 year's corrected age, even after accounting for gestational age and common neonatal morbidities. Cardiac values approached those seen in controls born full-term with increased mother's own milk exposure. There were no differences in any of the cardiac indices at 32 weeks' postmenstrual age, but with each week of exposure, right ventricle function (eg, right longitudinal strain: β, 0.016; 95% CI, 0.002-0.031; P < .001) was greater and pulmonary pressured (eg, pulmonary artery acceleration time: β, 0.0032; 95% CI, 0.0013-0.0062; P < .001). CONCLUSIONS AND RELEVANCE This study found that preterm infants with higher consumption of mother's own milk had enhanced cardiac performance at age 1 year, suggesting that mother's own milk consumption may play a dynamic modulator role on cardiac mechanics in preterm-born infants and help in normalization of the preterm cardiac phenotype.
Collapse
Affiliation(s)
- Afif El-Khuffash
- Department of Neonatology, The Rotunda Hospital and School of Medicine (Pediatrics), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Adam J Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe, Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Amish Jain
- Department of Paediatrics, University of Toronto and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Aaron Hamvas
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Gautam K Singh
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, Missouri
- Department of Pediatrics, Central Michigan University School of Medicine, Children's Hospital of Michigan, Detroit
| | - Philip T Levy
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
18
|
Left Ventricle Phenotyping Utilizing Tissue Doppler Imaging in Premature Infants with Varying Severity of Bronchopulmonary Dysplasia. J Clin Med 2021; 10:jcm10102211. [PMID: 34065264 PMCID: PMC8160781 DOI: 10.3390/jcm10102211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by alveolar-capillary simplification and is associated with pulmonary hypertension (PH) in preterm infants. The contribution of left ventricle (LV) disease towards this severe BPD-PH phenotype is not well established. We aimed to describe the longitudinal trajectory of the LV function as measured by tissue Doppler imaging (TDI) and its association with BPD-PH. We retrospectively assessed prospectively acquired clinical and echocardiographic data from 77 preterm infants born between 2011 and 2013. We characterized the LV function by measuring systolic and diastolic myocardial velocities (s’, e’, a’), isovolumetric relaxation time (IVRT), and myocardial performance index with TDI at three time periods from 32 and 36 weeks, postmenstrual age through one year of age. We also measured post systolic motion (PSM), a marker of myocardial dysfunction that results from asynchronous movement of the ventricular walls, and not previously described in preterm infants. Patients were stratified into groups according to BPD severity and the presence of PH and compared over time. Conventional TDI measures of the LV function were similar between groups, but the septal PSM was significantly prolonged over the first year of age in patients with BPD-PH. PSM provides a novel objective way to assess the hemodynamic impact of lung and pulmonary vascular disease severity on LV function in preterm infants with BPD and PH.
Collapse
|
19
|
Namba F, Tanaka K, Omori S, Ikeda K, Kawabata K, Sato H, Honda M, Ichikawa T, Minosaki Y, Michikawa T, Oka S, Kabe K. Extreme Prematurity and Pulmonary Outcomes Program in Saitama: Protocol for a Prospective Multicenter Cohort Study in Japan. JMIR Res Protoc 2021; 10:e22948. [PMID: 33666556 PMCID: PMC7980118 DOI: 10.2196/22948] [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: 07/27/2020] [Revised: 12/25/2020] [Accepted: 01/16/2021] [Indexed: 11/21/2022] Open
Abstract
Background Because of the improvements in survival rates for preterm infants, not only the rates of bronchopulmonary dysplasia (BPD) but also those of long-term respiratory complications of premature birth are increasing, resulting in financial and health burdens in developed countries. Thus far, the risk factors of respiratory morbidities in extremely preterm infants remain unknown. Furthermore, the definition and the predictive ability of BPD for long-term respiratory outcomes are yet to be determined. Objective The objective of our study, Extreme Prematurity and Pulmonary Outcomes Program in Saitama, is to develop the diagnostic criteria for BPD and to determine the prognostic factors contributing to the long-term pulmonary outcomes manifesting in extremely preterm infants. Methods The Extreme Prematurity and Pulmonary Outcomes Program in Saitama is an observational prospective cohort study performed by a consortium of six neonatal intensive care units (NICUs) in Saitama, Japan. The subjects included in this study are infants (from each clinical center) with gestational ages 22 to 27 weeks. The target is 400 subjects. This study aims to determine the definition of BPD and other perinatal factors that accurately predict the long-term pulmonary outcomes in survivors of extreme prematurity. Moreover, the association between BPD and postprematurity respiratory disease will be investigated using generalized linear models. Results The protocol and consent forms were evaluated and approved on September 5, 2019, by the Ethics Committee of Saitama Medical Center, Saitama Medical University. Enrollment began on April 1, 2020. It is expected to end on March 31, 2023. The follow-up for 1 year corrected age is expected to continue through the middle of 2024. Conclusions The Extreme Prematurity and Pulmonary Outcomes Program in Saitama incorporates aspects of neonatal care in secondary- and tertiary-level NICUs to develop existing research studies on the definition of BPD, objective biomarkers, and outcome measures of respiratory morbidity in extremely preterm infants beyond NICU hospitalization, thereby leading to a novel understanding of the nature and natural history of BPD and potential mechanistic and therapeutic targets in at-risk subjects. International Registered Report Identifier (IRRID) DERR1-10.2196/22948
Collapse
Affiliation(s)
- Fumihiko Namba
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Kosuke Tanaka
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Sayu Omori
- Division of Neonatology, Department of Pediatrics, Saitama City Hospital, Saitama, Japan
| | - Kazushige Ikeda
- Division of Neonatology, Department of Pediatrics, Saitama City Hospital, Saitama, Japan
| | - Ken Kawabata
- Division of Neonatology, Saitama Children's Medical Center, Saitama, Japan
| | - Hiroaki Sato
- Division of Neonatology, Department of Perinatal and Neonatal Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Masakazu Honda
- Division of Neonatal Medicine, Department of Pediatrics, Saitama Medical University Hospital, Saitama, Japan
| | - Tomonori Ichikawa
- Neonatal Intensive Care Unit, Kawaguchi Municipal Medical Center, Saitama, Japan
| | - Yoshihiro Minosaki
- Neonatal Intensive Care Unit, Kawaguchi Municipal Medical Center, Saitama, Japan
| | - Takehiro Michikawa
- Department of Environmental and Occupational Health, School of Medicine, Toho University, Tokyo, Japan
| | - Shuntaro Oka
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Kazuhiko Kabe
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| |
Collapse
|
20
|
Bhattacharya S, Mereness JA, Baran AM, Misra RS, Peterson DR, Ryan RM, Reynolds AM, Pryhuber GS, Mariani TJ. Lymphocyte-Specific Biomarkers Associated With Preterm Birth and Bronchopulmonary Dysplasia. Front Immunol 2021; 11:563473. [PMID: 33552042 PMCID: PMC7859626 DOI: 10.3389/fimmu.2020.563473] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 12/07/2020] [Indexed: 01/11/2023] Open
Abstract
Many premature babies who are born with neonatal respiratory distress syndrome (RDS) go on to develop Bronchopulmonary Dysplasia (BPD) and later Post-Prematurity Respiratory Disease (PRD) at one year corrected age, characterized by persistent or recurrent lower respiratory tract symptoms frequently related to inflammation and viral infection. Transcriptomic profiles were generated from sorted peripheral blood CD8+ T cells of preterm and full-term infants enrolled with consent in the NHLBI Prematurity and Respiratory Outcomes Program (PROP) at the University of Rochester and the University at Buffalo. We identified outcome-related gene expression patterns following standard methods to identify markers for oxygen utilization and BPD as outcomes in extremely premature infants. We further identified predictor gene sets for BPD based on transcriptomic data adjusted for gestational age at birth (GAB). RNA-Seq analysis was completed for CD8+ T cells from 145 subjects. Among the subjects with highest risk for BPD (born at <29 weeks gestational age (GA); n=72), 501 genes were associated with oxygen utilization. In the same set of subjects, 571 genes were differentially expressed in subjects with a diagnosis of BPD and 105 genes were different in BPD subjects as defined by physiologic challenge. A set of 92 genes could predict BPD with a moderately high degree of accuracy. We consistently observed dysregulation of TGFB, NRF2, HIPPO, and CD40-associated pathways in BPD. Using gene expression data from both premature and full-term subjects (n=116), we identified a 28 gene set that predicted the PRD status with a moderately high level of accuracy, which also were involved in TGFB signaling. Transcriptomic data from sort-purified peripheral blood CD8+ T cells from 145 preterm and full-term infants identified sets of molecular markers of inflammation associated with independent development of BPD in extremely premature infants at high risk for the disease and of PRD among the preterm and full-term subjects.
Collapse
Affiliation(s)
- Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Jared A. Mereness
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Andrea M. Baran
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Ravi S. Misra
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Derick R. Peterson
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Rita M. Ryan
- Department of Pediatrics, University at Buffalo, Buffalo, NY, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | | | - Gloria S. Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Thomas J. Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| |
Collapse
|
21
|
Echocardiographic Assessment of Right Ventricular Afterload in Preterm Infants: Maturational Patterns of Pulmonary Artery Acceleration Time Over the First Year of Age and Implications for Pulmonary Hypertension. J Am Soc Echocardiogr 2020; 32:884-894.e4. [PMID: 31272593 DOI: 10.1016/j.echo.2019.03.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Assessment of pulmonary hemodynamics is critical in the diagnosis and management of cardiopulmonary disease of premature infants, but reliable noninvasive indices of pulmonary hemodynamics in preterm infants are lacking. Because pulmonary artery acceleration time (PAAT) is a validated noninvasive method to assess right ventricular (RV) afterload in infants and children, the aim of this study was to investigate the maturational changes of PAAT measures in preterm infants over the first year of age and to discern the impact of typical cardiopulmonary abnormalities on these measures. METHODS In a prospective multicenter study of 239 preterm infants (<29 weeks at birth), PAAT was assessed at days 1, 2, and 5 to 7, at 32 and 36 weeks' postmenstrual age, and at 1-year corrected age. To account for heart rate variability, PAAT was adjusted for RV ejection time. Premature infants who developed bronchopulmonary dysplasia or had echocardiographic findings of pulmonary hypertension were analyzed separately. Intra- and interobserver reproducibility analysis was performed. RESULTS PAAT was feasible in 95% of the image acquisitions, and there was high intra- and interobserver agreement (intraclass correlation coefficients > 0.9 and coefficients of variation < 6%). In uncomplicated preterm infants (n = 103 [48%]) PAAT and PAAT adjusted for RV ejection time increased longitudinally from birth to 1-year corrected age (P < .001) and were linearly associated with gestational age at birth (r = 0.81 and r = 0.82, P < .001) and increasing postnatal weight and postnatal age (r > 0.81, P < .001). PAAT measures were significantly reduced (P < .001) in infants with bronchopulmonary dysplasia and/or pulmonary hypertension (n = 119 [51%]) beyond 1 week of age. CONCLUSIONS PAAT measures increase in preterm infants from birth to 1-year corrected age, reflective of the physiologic postnatal drop in RV afterload. Bronchopulmonary dysplasia and pulmonary hypertension have a negative impact on PAAT measures. By demonstrating excellent reliability and establishing reference patterns of PAAT in preterm infants, this study suggests that PAAT and PAAT adjusted for RV ejection time can be used as complementary parameters to assess physiologic and pathologic changes in pulmonary hemodynamics in neonates.
Collapse
|
22
|
Mandell EW, Ryan S, Seedorf GJ, Gonzalez T, Smith BJ, Fleet JC, Abman SH. Maternal Vitamin D Deficiency Causes Sustained Impairment of Lung Structure and Function and Increases Susceptibility to Hyperoxia-induced Lung Injury in Infant Rats. Am J Respir Cell Mol Biol 2020; 63:79-91. [PMID: 32135073 DOI: 10.1165/rcmb.2019-0295oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vitamin D deficiency (VDD) during pregnancy is associated with increased respiratory morbidities and risk for chronic lung disease after preterm birth. However, the direct effects of maternal VDD on perinatal lung structure and function and whether maternal VDD increases the susceptibility of lung injury due to hyperoxia are uncertain. In the present study, we sought to determine whether maternal VDD is sufficient to impair lung structure and function and whether VDD increases the impact of hyperoxia on the developing rat lung. Four-week-old rats were fed VDD chow and housed in a room shielded from ultraviolet A/B light to achieve 25-hydroxyvitamin D concentrations <10 ng/ml at mating and throughout lactation. Lung structure was assessed at 2 weeks for radial alveolar count, mean linear intercept, pulmonary vessel density, and lung function (lung compliance and resistance). The effects of hyperoxia for 2 weeks after birth were assessed after exposure to fraction of inspired oxygen of 0.95. At 2 weeks, VDD offspring had decreased alveolar and vascular growth and abnormal airway reactivity and lung function. Impaired lung structure and function in VDD offspring were similar to those observed in control rats exposed to postnatal hyperoxia alone. Maternal VDD causes sustained abnormalities of distal lung growth, increases in airway hyperreactivity, and abnormal lung mechanics during infancy. These changes in VDD pups were as severe as those measured after exposure to postnatal hyperoxia alone. We speculate that antenatal disruption of vitamin D signaling increases the risk for late-childhood respiratory disease.
Collapse
Affiliation(s)
| | - Sharon Ryan
- Pediatric Heart Lung Center.,Section of Neonatology, and
| | - Gregory J Seedorf
- Pediatric Heart Lung Center.,Section of Pulmonary Medicine, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Tania Gonzalez
- Pediatric Heart Lung Center.,Section of Neonatology, and
| | - Bradford J Smith
- Department of Bioengineering, College of Engineering and Applied Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado; and
| | - James C Fleet
- Department of Foods and Nutrition, and.,Interdepartmental Nutrition Program, Purdue University, West Lafayette, Indiana
| | - Steven H Abman
- Pediatric Heart Lung Center.,Section of Pulmonary Medicine, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado
| |
Collapse
|
23
|
Maturational patterns of left ventricular rotational mechanics in pre-term infants through 1 year of age. Cardiol Young 2020; 30:1238-1246. [PMID: 32665043 DOI: 10.1017/s1047951120001912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pre-mature birth impacts left ventricular development, predisposing this population to long-term cardiovascular risk. The aims of this study were to investigate maturational changes in rotational properties from the neonatal period through 1 year of age and to discern the impact of cardiopulmonary complications of pre-maturity on these measures. METHODS Pre-term infants (<29 weeks at birth, n = 117) were prospectively enrolled and followed to 1-year corrected age. Left ventricular basal and apical rotation, twist, and torsion were measured by two-dimensional speckle-tracking echocardiography and analysed at 32 and 36 weeks post-menstrual age and 1-year corrected age. A mixed random effects model with repeated measures analysis was used to compare rotational mechanics over time. Torsion was compared in infants with and without complications of cardiopulmonary diseases of pre-maturity, specifically bronchopulmonary dysplasia, pulmonary hypertension, and patent ductus arteriosus. RESULTS Torsion decreased from 32 weeks post-menstrual age to 1-year corrected age in all pre-term infants (p < 0.001). The decline from 32 to 36 weeks post-menstrual age was more pronounced in infants with cardiopulmonary complications, but was similar to healthy pre-term infants from 36 weeks post-menstrual age to 1-year corrected age. The decline was due to directional and magnitude changes in apical rotation over time (p < 0.05). CONCLUSION This study tracks maturational patterns of rotational mechanics in pre-term infants and reveals torsion declines from the neonatal period through 1 year. Cardiopulmonary diseases of pre-maturity may negatively impact rotational mechanics during the neonatal period, but the myocardium recovers by 1-year corrected age.
Collapse
|
24
|
Dylag AM, Kopin HG, O’Reilly MA, Wang H, Davis SD, Ren CL, Pryhuber GS. Early Neonatal Oxygen Exposure Predicts Pulmonary Morbidity and Functional Deficits at 1 Year. J Pediatr 2020; 223:20-28.e2. [PMID: 32711747 PMCID: PMC9337224 DOI: 10.1016/j.jpeds.2020.04.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To evaluate the predictive value of cumulative oxygen exposure thresholds over the first 2 postnatal weeks, linking them to bronchopulmonary dysplasia (BPD) and 1-year pulmonary morbidity and lung function in extremely low gestational age newborns. STUDY DESIGN Infants (N = 704) enrolled in the Prematurity and Respiratory Outcomes Program, a multicenter prospective cohort study, that survived to discharge were followed through their neonatal intensive care unit hospitalization to 1-year corrected age. Cumulative oxygen exposure (OxygenAUC14) thresholds were derived from univariate models of BPD, stratifying infants into high-, intermediate-, and low-oxygen exposure groups. These groups were then used in multivariate logistic regressions to prospectively predict post-prematurity respiratory disease (PRD), respiratory morbidity score (RMS) in the entire cohort, and pulmonary function z scores (N = 108 subset of infants) at 1-year corrected age. RESULTS Over the first 14 postnatal days, infants exposed to high oxygen averaged ≥33.1% oxygen, infants exposed to intermediate oxygen averaged 29.1%-33.1%, and infants exposed to low oxygen were below both cutoffs. In multivariate models, infants exposed to high oxygen showed increased PRD and RMS, whereas infants exposed to intermediate oxygen demonstrated increased moderate/severe RMS. Infants in the high/intermediate groups had decreased forced expiratory volume at 0.5 seconds/forced vital capacity ratio. CONCLUSIONS OxygenAUC14 establishes 3 thresholds of oxygen exposure that risk stratify infants early in their neonatal course, thereby predicting short-term (BPD) and 1-year (PRD, RMS) respiratory morbidity. Infants with greater OxygenAUC14 have altered pulmonary function tests at 1 year of age, indicating early evidence of obstructive lung disease and flow limitation, which may predispose extremely low gestational age newborns to increased long-term pulmonary morbidity. TRIAL REGISTRATION ClinicalTrials.gov: NCT01435187.
Collapse
Affiliation(s)
- Andrew M. Dylag
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY
| | - Hannah G. Kopin
- School of Medicine, School of Public Health Sciences, University of Rochester, Rochester, NY
| | - Michael A. O’Reilly
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY
| | - Hongyue Wang
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY
| | - Stephanie D. Davis
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Clement L. Ren
- Division of Pediatric Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indiana University, Indianapolis, IN
| | - Gloria S. Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY
| |
Collapse
|
25
|
McDavid A, Corbett AM, Dutra JL, Straw AG, Topham DJ, Pryhuber GS, Caserta MT, Gill SR, Scheible KM, Holden-Wiltse J. Eight practices for data management to enable team data science. J Clin Transl Sci 2020; 5:e14. [PMID: 33948240 PMCID: PMC8057476 DOI: 10.1017/cts.2020.501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION In clinical and translational research, data science is often and fortuitously integrated with data collection. This contrasts to the typical position of data scientists in other settings, where they are isolated from data collectors. Because of this, effective use of data science techniques to resolve translational questions requires innovation in the organization and management of these data. METHODS We propose an operational framework that respects this important difference in how research teams are organized. To maximize the accuracy and speed of the clinical and translational data science enterprise under this framework, we define a set of eight best practices for data management. RESULTS In our own work at the University of Rochester, we have strived to utilize these practices in a customized version of the open source LabKey platform for integrated data management and collaboration. We have applied this platform to cohorts that longitudinally track multidomain data from over 3000 subjects. CONCLUSIONS We argue that this has made analytical datasets more readily available and lowered the bar to interdisciplinary collaboration, enabling a team-based data science that is unique to the clinical and translational setting.
Collapse
Affiliation(s)
- Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Anthony M. Corbett
- Clinical and Translational Science Institute, University of Rochester, Rochester, NY, USA
| | - Jennifer L. Dutra
- Clinical and Translational Science Institute, University of Rochester, Rochester, NY, USA
| | - Andrew G. Straw
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - David J. Topham
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | | - Mary T. Caserta
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
- Clinical and Translational Science Institute, University of Rochester, Rochester, NY, USA
| |
Collapse
|
26
|
Sahoo D, Zaramela LS, Hernandez GE, Mai U, Taheri S, Dang D, Stouch AN, Medal RM, McCoy AM, Aschner JL, Blackwell TS, Zengler K, Prince LS. Transcriptional profiling of lung macrophages identifies a predictive signature for inflammatory lung disease in preterm infants. Commun Biol 2020; 3:259. [PMID: 32444859 PMCID: PMC7244484 DOI: 10.1038/s42003-020-0985-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Lung macrophages mature after birth, placing newborn infants, particularly those born preterm, within a unique window of susceptibility to disease. We hypothesized that in preterm infants, lung macrophage immaturity contributes to the development of bronchopulmonary dysplasia (BPD), the most common serious complication of prematurity. By measuring changes in lung macrophage gene expression in preterm patients at risk of BPD, we show here that patients eventually developing BPD had higher inflammatory mediator expression even on the first day of life. Surprisingly, the ex vivo response to LPS was similar across all samples. Our analysis did however uncover macrophage signature genes whose expression increased in the first week of life specifically in patients resilient to disease. We propose that these changes describe the dynamics of human lung macrophage differentiation. Our study therefore provides new mechanistic insight into both neonatal lung disease and human developmental immunology.
Collapse
Affiliation(s)
- Debashis Sahoo
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Livia S Zaramela
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gilberto E Hernandez
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Uyen Mai
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sahar Taheri
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Dharanidhar Dang
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ashley N Stouch
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rachel M Medal
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Alyssa M McCoy
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Judy L Aschner
- Department of Pediatrics, Joseph M Sanzari Children's Hospital, Hackensack University Medical Center, Hackensack Meridian School of Medicine at Seton Hall, Hackensack, NJ, 07110, USA
| | - Timothy S Blackwell
- Departments of Medicine, Cancer Biology, and Developmental Cell Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Karsten Zengler
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Lawrence S Prince
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
27
|
Voynow JA, Fisher K, Sunday ME, Cotten CM, Hamvas A, Hendricks-Muñoz KD, Poindexter BB, Pryhuber GS, Ren CL, Ryan RM, Sharp JK, Young SP, Zhang H, Greenberg RG, Herring AH, Davis SD. Urine gastrin-releasing peptide in the first week correlates with bronchopulmonary dysplasia and post-prematurity respiratory disease. Pediatr Pulmonol 2020; 55:899-908. [PMID: 31995668 PMCID: PMC7071969 DOI: 10.1002/ppul.24665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/04/2020] [Indexed: 11/07/2022]
Abstract
RATIONALE Bronchopulmonary dysplasia (BPD) is associated with post-prematurity respiratory disease (PRD) in survivors of extreme preterm birth. Identifying early biomarkers that correlate with later development of BPD and PRD may provide insights for intervention. In a preterm baboon model, elevated gastrin-releasing peptide (GRP) is associated with BPD, and GRP inhibition mitigates BPD occurrence. OBJECTIVE We performed a prospective cohort study to investigate whether urine GRP levels obtained in the first postnatal week were associated with BPD, PRD, and other urinary biomarkers of oxidative stress. METHODS Extremely low gestational age infants (23-28 completed weeks) were enrolled in a US multicenter observational study, The Prematurity and Respiratory Outcomes Program (http://clinicaltrials.gov/ct2/show/NCT01435187). We used multivariable logistic regression to examine the association between urine GRP in the first postnatal week and multiple respiratory outcomes: BPD, defined as supplemental oxygen use at 36 + 0 weeks postmenstrual age, and post-PRD, defined by positive quarterly surveys for increased medical utilization over the first year (PRD score). RESULTS A total of 109 of 257 (42%) infants had BPD, and 120 of 217 (55%) had PRD. On adjusted analysis, GRP level more than 80 was associated with BPD (adjusted odds ratio [aOR], 1.83; 95% confidence interval [CI], 1.03-3.25) and positive PRD score (aOR, 2.46; 95% CI, 1.35-4.48). Urine GRP levels correlated with duration of NICU ventilatory and oxygen support and with biomarkers of oxidative stress: allantoin and 8-hydroxydeoxyguanosine. CONCLUSIONS Urine GRP in the first postnatal week was associated with concurrent urine biomarkers of oxidative stress and with later diagnoses of BPD and PRD.
Collapse
Affiliation(s)
- Judith A Voynow
- Division of Pediatric Pulmonology, Duke University, Durham, North Carolina.,Division of Pediatric Pulmonology, Children's Hospital of Richmond, Richmond, Virginia
| | - Kimberley Fisher
- Division of Neonatology, Duke University, Durham, North Carolina
| | - Mary E Sunday
- Department of Pathology, Duke University, Durham, North Carolina
| | - Charles M Cotten
- Division of Neonatology, Duke University, Durham, North Carolina
| | - Aaron Hamvas
- Division of Neonatology, Washington University, St Louis, Missouri.,Division of Neonatology, Northwestern University, Chicago, Illinois
| | | | - Brenda B Poindexter
- Division of Neonatology, Indiana University, Indianapolis, Indiana.,Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Clement L Ren
- Division of Pediatric Pulmonology, University of Rochester, Rochester, New York.,Division of Pediatric Pulmonology, Indiana University, Indianapolis, Indiana
| | - Rita M Ryan
- Division of Neonatology, State University of New York at Buffalo, Buffalo, New York.,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Jack K Sharp
- Division of Pediatric Pulmonology, Duke University, Durham, North Carolina.,Division of Pediatric Pulmonology, State University of New York at Buffalo, Buffalo, New York.,Division of Pediatric Pulmonology, Baylor College of Medicine, Houston, Texas
| | - Sarah P Young
- Division of Medical Genetics, Duke University, Durham, North Carolina
| | - Haoyue Zhang
- Division of Medical Genetics, Duke University, Durham, North Carolina
| | - Rachel G Greenberg
- Division of Neonatology and The Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Amy H Herring
- Duke Global Health Institute, Duke University, Durham, North Carolina
| | - Stephanie D Davis
- Division of Pediatric Pulmonology, Indiana University, Indianapolis, Indiana.,Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
28
|
Greenberg JM, Poindexter BB, Shaw PA, Bellamy SL, Keller RL, Moore PE, McPherson C, Ryan RM. Respiratory medication use in extremely premature (<29 weeks) infants during initial NICU hospitalization: Results from the prematurity and respiratory outcomes program. Pediatr Pulmonol 2020; 55:360-368. [PMID: 31794157 DOI: 10.1002/ppul.24592] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND The use of medications to treat respiratory conditions of extreme prematurity is often based upon studies of adults or children over 2 years of age. Little is known about the spectrum of medications used or dosing ranges. To inform the design of future studies, we conducted a prospective analysis of respiratory medication exposure among 832 extremely low gestational age neonates. METHODS The prematurity and respiratory outcomes program (PROP) enrolled neonates less than 29-week gestation from 6 centers incorporating 13 clinical sites. We collected recorded daily "respiratory" medications given along with dosing information through 40-week postmenstrual age or neonatal intensive care unit discharge if earlier. RESULTS PROP participants were exposed to a wide range of respiratory medications, often at doses beyond published recommendations. Nearly 50% received caffeine and furosemide beyond published recommendations for cumulative dose. Those who developed bronchopulmonary dysplasia were more likely to receive treatment with respiratory medications. However, more than 30% of PROP subjects that did not develop bronchopulmonary dysplasia also were treated with diuretics, systemic steroids, and other respiratory medications. CONCLUSION Extremely preterm neonates in PROP were exposed to high doses of medications at levels known to generate significant adverse effects. With limited evidence for efficacy, there is an urgent need for controlled trials in this vulnerable patient population.
Collapse
Affiliation(s)
- James M Greenberg
- Departments of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Brenda B Poindexter
- Departments of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Pamela A Shaw
- Department of Biostatistics, Epidemiology and Informatics, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scarlett L Bellamy
- Department of Biostatistics, Epidemiology and Informatics, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roberta L Keller
- Department of Pediatrics, University of California-San Francisco, San Francisco, California
| | - Paul E Moore
- Departments of Pediatrics and Pharmacology, Vanderbilt University, Nashville, Tennessee
| | | | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
29
|
Edwards BA, Nava-Guerra L, Kemp JS, Carroll JL, Khoo MC, Sands SA, Terrill PI, Landry SA, Amin RS. Assessing ventilatory instability using the response to spontaneous sighs during sleep in preterm infants. Sleep 2019; 41:5077835. [PMID: 30137560 DOI: 10.1093/sleep/zsy161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Study Objectives Periodic breathing (PB) is common in newborns and is an obvious manifestation of ventilatory control instability. However, many infants without PB may still have important underlying ventilatory control instabilities that go unnoticed using standard clinical monitoring. Methods to detect infants with "subclinical" ventilatory control instability are therefore required. The current study aimed to assess the degree of ventilatory control instability using simple bedside recordings in preterm infants. Methods Respiratory inductance plethysmography (RIP) recordings were analyzed from ~20 minutes of quiet sleep in 20 preterm infants at 36 weeks post-menstrual age (median [range]: 36 [34-40]). The percentage time spent in PB was also calculated for each infant (%PB). Spontaneous sighs were identified and breath-by-breath measurements of (uncalibrated) ventilation were derived from RIP traces. Loop gain (LG, a measure of ventilatory control instability) was calculated by fitting a simple ventilatory control model (gain, time-constant, delay) to the post-sigh ventilatory pattern. For comparison, periodic inter-breath variability was also quantified using power spectral analysis (ventilatory oscillation magnitude index [VOMI]). Results %PB was strongly associated with LG (r2 = 0.77, p < 0.001) and moderately with the VOMI (r2 = 0.21, p = 0.047). LG (0.52 ± 0.05 vs. 0.30 ± 0.03; p = 0.0025) and the VOMI (-8.2 ± 1.1 dB vs. -11.8 ± 0.9 dB; p = 0.026) were both significantly higher in infants that displayed PB vs. those without. Conclusions LG and VOMI determined from the ventilatory responses to spontaneous sighs can provide a practical approach to assessing ventilatory control instability in preterm infants. Such simple techniques may help identify infants at particular risk for ventilatory instabilities with concomitant hypoxemia and its associated consequences.
Collapse
Affiliation(s)
- Bradley A Edwards
- Sleep and Circadian Medicine Laboratory, Department of Physiology, Monash University, Melbourne, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.,Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Leonardo Nava-Guerra
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - James S Kemp
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - John L Carroll
- Division of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Michael C Khoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA
| | - Scott A Sands
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Philip I Terrill
- School of Information Technology and Electrical Engineering, University of Queensland, Brisbane, Australia
| | - Shane A Landry
- Sleep and Circadian Medicine Laboratory, Department of Physiology, Monash University, Melbourne, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia
| | - Raouf S Amin
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
30
|
Benjamin JT, van der Meer R, Slaughter JC, Steele S, Plosa EJ, Sucre JM, Moore PE, Aschner JL, Blackwell TS, Young LR. Inverse Relationship between Soluble RAGE and Risk for Bronchopulmonary Dysplasia. Am J Respir Crit Care Med 2019; 197:1083-1086. [PMID: 29035091 DOI: 10.1164/rccm.201707-1445le] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | | | | | - Steven Steele
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Erin J Plosa
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | | | - Paul E Moore
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Judy L Aschner
- 2 Children's Hospital at Montefiore/Albert Einstein College of Medicine Bronx, New York and
| | - Timothy S Blackwell
- 1 Vanderbilt University Medical Center Nashville, Tennessee.,3 Nashville Veterans Affairs Medical Center Nashville, Tennessee
| | - Lisa R Young
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
31
|
The discovery BPD (D-BPD) program: study protocol of a prospective translational multicenter collaborative study to investigate determinants of chronic lung disease in very low birth weight infants. BMC Pediatr 2019; 19:227. [PMID: 31279333 PMCID: PMC6612113 DOI: 10.1186/s12887-019-1610-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/02/2019] [Indexed: 01/21/2023] Open
Abstract
Background Premature birth is a growing and serious public health problem affecting more than one of every ten infants worldwide. Bronchopulmonary dysplasia (BPD) is the most common neonatal morbidity associated with prematurity and infants with BPD suffer from increased incidence of respiratory infections, asthma, other forms of chronic lung illness, and death (Day and Ryan, Pediatr Res 81: 210–213, 2017; Isayama et la., JAMA Pediatr 171:271–279, 2017). BPD is now understood as a longitudinal disease process influenced by the intrauterine environment during gestation and modulated by gene-environment interactions throughout the neonatal and early childhood periods. Despite of this concept, there remains a paucity of multidisciplinary team-based approaches dedicated to the comprehensive study of this complex disease. Methods The Discovery BPD (D-BPD) Program involves a cohort of infants < 1,250 g at birth prospectively followed until 6 years of age. The program integrates analysis of detailed clinical data by machine learning, genetic susceptibility and molecular translation studies. Discussion The current gap in understanding BPD as a complex multi-trait spectrum of different disease endotypes will be addressed by a bedside-to-bench and bench-to-bedside approach in the D-BPD program. The D-BPD will provide enhanced understanding of mechanisms, evolution and consequences of lung diseases in preterm infants. The D-BPD program represents a unique opportunity to combine the expertise of biologists, neonatologists, pulmonologists, geneticists and biostatisticians to examine the disease process from multiple perspectives with a singular goal of improving outcomes of premature infants. Trial registration Does not apply for this study. Electronic supplementary material The online version of this article (10.1186/s12887-019-1610-8) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Erickson CT, Patel MD, Choudhry S, Bisselou KS, Sekarski T, Craft M, Li L, Khuffash AE, Hamvas A, Kutty S, Singh GK, Levy PT. Persistence of right ventricular dysfunction and altered morphometry in asymptomatic preterm Infants through one year of age: Cardiac phenotype of prematurity. Cardiol Young 2019; 29:945-953. [PMID: 31287038 PMCID: PMC6715519 DOI: 10.1017/s1047951119001161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Prematurity impacts myocardial development and may determine long-term outcomes. The objective of this study was to test the hypothesis that preterm neonates develop right ventricle dysfunction and adaptive remodelling by 32 weeks post-menstrual age that persists through 1 year corrected age. MATERIALS AND METHODS A subset of 80 preterm infants (born <29 weeks) was selected retrospectively from a prospectively enrolled cohort and measures of right ventricle systolic function and morphology by two-dimensional echocardiography were assessed at 32 weeks post-menstrual age and at 1 year of corrected age. Comparisons were made to 50 term infants at 1 month and 1 year of age. Sub-analyses were performed in preterm-born infants with bronchopulmonary dysplasia and/or pulmonary hypertension. RESULT In both term and preterm infants, right ventricle function and morphology increased over the first year (p < 0.01). The magnitudes of right ventricle function measures were lower in preterm-born infants at each time period (p < 0.01 for all) and right ventricle morphology indices were wider in all preterm infants by 1 year corrected age, irrespective of lung disease. Measures of a) right ventricle function were further decreased and b) morphology increased through 1 year in preterm infants with bronchopulmonary dysplasia and/or pulmonary hypertension (p < 0.01). CONCLUSION Preterm infants exhibit abnormal right ventricle performance with remodelling at 32 weeks post-menstrual age that persists through 1 year corrected age, suggesting a less developed intrinsic myocardial function response following preterm birth. The development of bronchopulmonary dysplasia and pulmonary hypertension leave a further negative impact on right ventricle mechanics over the first year of age.
Collapse
MESH Headings
- Bronchopulmonary Dysplasia/complications
- Bronchopulmonary Dysplasia/pathology
- Echocardiography
- Female
- Humans
- Hypertension, Pulmonary/complications
- Hypertension, Pulmonary/pathology
- Infant
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/diagnostic imaging
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/pathology
- Male
- Retrospective Studies
- Ventricular Dysfunction, Right/diagnostic imaging
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/pathology
- Ventricular Remodeling
Collapse
Affiliation(s)
- Collin T. Erickson
- Department of Pediatric Cardiology and Cardiovascular Surgery, University of Nebraska College of Medicine and Children’s Hospital and Medical Center, Omaha, Nebraska
| | - Meghna D. Patel
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Swati Choudhry
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
- Section of Pediatric Cardiology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Karl Stessy Bisselou
- Department of Pediatric Cardiology and Cardiovascular Surgery, University of Nebraska College of Medicine and Children’s Hospital and Medical Center, Omaha, Nebraska
| | - Tim Sekarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Mary Craft
- Department of Pediatric Cardiology and Cardiovascular Surgery, University of Nebraska College of Medicine and Children’s Hospital and Medical Center, Omaha, Nebraska
| | - Ling Li
- Department of Pediatric Cardiology and Cardiovascular Surgery, University of Nebraska College of Medicine and Children’s Hospital and Medical Center, Omaha, Nebraska
| | - Afif El Khuffash
- Department of Neonatology, The Rotunda Hospital & School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aaron Hamvas
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shelby Kutty
- Department of Pediatric Cardiology and Cardiovascular Surgery, University of Nebraska College of Medicine and Children’s Hospital and Medical Center, Omaha, Nebraska
- Division of Pediatric Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Gautam K. Singh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Philip T. Levy
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Dennery PA, Di Fiore JM, Ambalavanan N, Bancalari E, Carroll JL, Claure N, Hamvas A, Hibbs AM, Indic P, Kemp J, Krahn KN, Lake D, Laposky A, Martin RJ, Natarajan A, Rand C, Schau M, Weese-Mayer DE, Zimmet AM, Moorman JR. Pre-Vent: the prematurity-related ventilatory control study. Pediatr Res 2019; 85:769-776. [PMID: 30733614 PMCID: PMC6503843 DOI: 10.1038/s41390-019-0317-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND The increasing incidence of bronchopulmonary dysplasia in premature babies may be due in part to immature ventilatory control, contributing to hypoxemia. The latter responds to ventilation and/or oxygen therapy, treatments associated with adverse sequelae. This is an overview of the Prematurity-Related Ventilatory Control Study which aims to analyze the under-utilized cardiorespiratory continuous waveform monitoring data to delineate mechanisms of immature ventilatory control in preterm infants and identify predictive markers. METHODS Continuous ECG, heart rate, respiratory, and oxygen saturation data will be collected throughout the NICU stay in 500 infants < 29 wks gestation across 5 centers. Mild permissive hypercapnia, and hyperoxia and/or hypoxia assessments will be conducted in a subcohort of infants along with inpatient questionnaires, urine, serum, and DNA samples. RESULTS Primary outcomes will be respiratory status at 40 wks and quantitative measures of immature breathing plotted on a standard curve for infants matched at 36-37 wks. Physiologic and/or biologic determinants will be collected to enhance the predictive model linking ventilatory control to outcomes. CONCLUSIONS By incorporating bedside monitoring variables along with biomarkers that predict respiratory outcomes we aim to elucidate individualized cardiopulmonary phenotypes and mechanisms of ventilatory control contributing to adverse respiratory outcomes in premature infants.
Collapse
Affiliation(s)
- Phyllis A. Dennery
- Brown University, Warren Alpert School of Medicine, Hasbro Children’s Hospital, Providence, RI,Address for Correspondence Phyllis A. Dennery, MD, Department of Pediatrics, Warren Alpert School of Medicine at Brown University, Office of the Chair, Hasbro Children’s Hospital, 593 Eddy Street, Suite 125 / Providence, RI 02903, (401) 444-5648,
| | - Juliann M. Di Fiore
- Case Western Reserve University, School of Medicine, Department of Pediatrics, University Hospitals: Rainbow Babies & Children’s Hospital, Division of Neonatology, Cleveland, OH
| | - Namasivayam Ambalavanan
- University of Alabama at Birmingham School of Med, Div. of Neonatology, Dept. Pediatrics Molecular and Cellular Pathology, and Cell, Developmental, and Integrative Biology, Birmingham, AL
| | - Eduardo Bancalari
- University of Miami Miller School of Medicine, Holtz Children’s Hospital - Jackson Memorial Medical Center, Division of Neonatology, Department of Pediatrics, Miami, FL
| | - John L. Carroll
- University of Arkansas for Medical Science, Department of Pediatrics, Little Rock, AR
| | - Nelson Claure
- University of Miami Miller School of Medicine, Holtz Children’s Hospital - Jackson Memorial Medical Center, Division of Neonatology, Department of Pediatrics, Miami, FL
| | - Aaron Hamvas
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Chicago, IL
| | - Anna Maria Hibbs
- Case Western Reserve University, School of Medicine, Department of Pediatrics, University Hospitals: Rainbow Babies & Children’s Hospital, Division of Neonatology, Cleveland, OH
| | - Premananda Indic
- University of Texas Tyler, Department of Electrical Engineering, University of Alabama at Birmingham School of Med, Div. of Neonatology, Dept. Pediatrics, Tyler, TX
| | - James Kemp
- Washington University School of Medicine in St. Louis, Department of Pediatrics, St. Louis, MO
| | - Katy N. Krahn
- University of Virginia School of Medicine, Division of Cardiovascular Medicine, Charlottesville, VA
| | - Douglas Lake
- University of Virginia School of Medicine, Division of Cardiovascular Medicine, Charlottesville, VA
| | - Aaron Laposky
- National Institute of Health, National Center of Sleep Disorders Research, Bethesda, MD
| | - Richard J. Martin
- Case Western Reserve University, School of Medicine, Department of Pediatrics, University Hospitals: Rainbow Babies & Children’s Hospital, Division of Neonatology, Cleveland, OH
| | - Aruna Natarajan
- Division of Lung Diseases, National Heart Lung and Blood Institute, National Institute of Health, Bethesda, MD
| | - Casey Rand
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Chicago, IL
| | - Molly Schau
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Chicago, IL
| | - Debra E. Weese-Mayer
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Stanley Manne Children’s Research Institute, Chicago, IL
| | - Amanda M. Zimmet
- University of Virginia School of Medicine, Division of Cardiovascular Medicine, Charlottesville, VA
| | - J. Randall Moorman
- University of Virginia School of Medicine, Division of Cardiovascular Medicine, Charlottesville, VA
| |
Collapse
|
34
|
Respiratory Medications in Infants <29 Weeks during the First Year Postdischarge: The Prematurity and Respiratory Outcomes Program (PROP) Consortium. J Pediatr 2019; 208:148-155.e3. [PMID: 30857774 PMCID: PMC6486865 DOI: 10.1016/j.jpeds.2018.12.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/10/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To determine patterns of respiratory medications used in neonatal intensive care unit graduates. STUDY DESIGN The Prematurity Respiratory Outcomes Program enrolled 835 babies <29 weeks of gestation in the first week. Of 751 survivors, 738 (98%) completed at least 1, and 85% completed all 4, postdischarge medication usage in-person/telephone parental questionnaires requested at 3, 6, 9, and 12 months of corrected age. Respiratory drug usage over the first year of life after in neonatal intensive care unit discharge was analyzed. RESULTS During any given quarter, 66%-75% of the babies received no respiratory medication and 45% of the infants received no respiratory drug over the first year. The most common postdischarge medication was the inhaled bronchodilator albuterol; its use increased significantly from 13% to 31%. Diuretic usage decreased significantly from 11% to 2% over the first year. Systemic steroids (prednisone, most commonly) were used in approximately 5% of subjects in any one quarter. Inhaled steroids significantly increased over the first year from 9% to 14% at 12 months. Drug exposure changed significantly based on gestational age with 72% of babies born at 23-24 weeks receiving at least 1 respiratory medication but only 40% of babies born at 28 weeks. Overall, at some time in the first year, 55% of infants received at least 1 drug including an inhaled bronchodilator (45%), an inhaled steroid (22%), a systemic steroid (15%), or diuretic (12%). CONCLUSION Many babies born at <29 weeks have no respiratory medication exposure postdischarge during the first year of life. Inhaled medications, including bronchodilators and steroids, increase over the first year.
Collapse
|
35
|
Ryan RM, Feng R, Bazacliu C, Ferkol TW, Ren CL, Mariani TJ, Poindexter BB, Wang F, Moore PE. Black Race Is Associated with a Lower Risk of Bronchopulmonary Dysplasia. J Pediatr 2019; 207:130-135.e2. [PMID: 30612812 PMCID: PMC6458591 DOI: 10.1016/j.jpeds.2018.11.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 10/22/2018] [Accepted: 11/09/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To use a large current prospective cohort of infants <29 weeks to compare bronchopulmonary dysplasia (BPD) rates in black and white infants. STUDY DESIGN The Prematurity and Respiratory Outcome Program (PROP) enrolled 835 infants born in 2011-2013 at <29 weeks of gestation; 728 black or white infants survived to 36 weeks postmenstrual age (PMA). Logistic regression was used to compare BPD outcomes (defined as supplemental oxygen requirement at 36 weeks PMA) between the races, adjusted for gestational age (GA), antenatal steroid use, intubation at birth, and surfactant use at birth. RESULTS Of 707 black or white infants with available BPD outcomes, BPD was lower in black infants (38% vs 45%), even though they were of significantly lower GA. At every GA, BPD was more common in white infants. The aOR for BPD was 0.60 (95% CI, 0.42-0.85; P = .004) for black infants compared with white infants after adjusting for GA. Despite the lower rate of BPD, black infants had a higher rate of first-year post-prematurity respiratory disease (black, 79%; white, 63%). CONCLUSIONS In this large cohort of recently born preterm infants at <29 weeks GA, compared with white infants, black infants had a lower risk of BPD but an increased risk of persistent respiratory morbidity.
Collapse
Affiliation(s)
- Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC.
| | - Rui Feng
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA
| | | | - Thomas W Ferkol
- Department of Pediatrics, Washington University, St. Louis, MO
| | - Clement L Ren
- Division of Pediatric Pulmonology, Allergy and Sleep Medicine, Department of Pediatrics, Indiana University, Indianapolis, IN
| | - Thomas J Mariani
- Department of Pediatrics, University of Rochester, Rochester, NY
| | - Brenda B Poindexter
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Fan Wang
- Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH
| | - Paul E Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN
| |
Collapse
|
36
|
Burris HH, Hwang SS, Collins JW, Kirpalani H, Wright CJ. Re-conceptualizing Associations between Race and Morbidities of Extreme Prematurity. J Pediatr 2019; 207:10-14.e1. [PMID: 30723017 DOI: 10.1016/j.jpeds.2018.12.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 12/21/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Heather H Burris
- Division of Neonatology, The Children's Hospital of Philadelphia at the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunah S Hwang
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado
| | - James W Collins
- Division of Neonatology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Haresh Kirpalani
- Emeritus, Division of Neonatology, The Children's Hospital of Philadelphia at the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado.
| |
Collapse
|
37
|
Ballard PL, Keller RL, Truog WE, Chapin C, Horneman H, Segal MR, Ballard RA. Surfactant status and respiratory outcome in premature infants receiving late surfactant treatment. Pediatr Res 2019; 85:305-311. [PMID: 30140069 PMCID: PMC6377352 DOI: 10.1038/s41390-018-0144-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/21/2018] [Accepted: 07/27/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Many premature infants with respiratory failure are deficient in surfactant, but the relationship to occurrence of bronchopulmonary dysplasia (BPD) is uncertain. METHODS Tracheal aspirates were collected from 209 treated and control infants enrolled at 7-14 days in the Trial of Late Surfactant. The content of phospholipid, surfactant protein B, and total protein were determined in large aggregate (active) surfactant. RESULTS At 24 h, surfactant treatment transiently increased surfactant protein B content (70%, p < 0.01), but did not affect recovered airway surfactant or total protein/phospholipid. The level of recovered surfactant during dosing was directly associated with content of surfactant protein B (r = 0.50, p < 0.00001) and inversely related to total protein (r = 0.39, p < 0.0001). For all infants, occurrence of BPD was associated with lower levels of recovered large aggregate surfactant, higher protein content, and lower SP-B levels. Tracheal aspirates with lower amounts of recovered surfactant had an increased proportion of small vesicle (inactive) surfactant. CONCLUSIONS We conclude that many intubated premature infants are deficient in active surfactant, in part due to increased intra-alveolar metabolism, low SP-B content, and protein inhibition, and that the severity of this deficit is predictive of BPD. Late surfactant treatment at the frequency used did not provide a sustained increase in airway surfactant.
Collapse
Affiliation(s)
- Philip L Ballard
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| | - Roberta L. Keller
- Department of Pediatrics, University of California, San Francisco, San Francisco CA
| | - William E. Truog
- Department of Pediatrics, Children’s Mercy Hospitals and Clinics/University of Missouri-Kansas City School of Medicine, Kansas City MO
| | - Cheryl Chapin
- Department of Pediatrics, University of California, San Francisco, San Francisco CA
| | - Hart Horneman
- Department of Pediatrics, University of California, San Francisco, San Francisco CA
| | - Mark R. Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco CA
| | - Roberta A Ballard
- Department of Pediatrics, University of California, San Francisco, San Francisco CA
| | | |
Collapse
|
38
|
DeMauro SB, Bellamy SL, Fernando M, Hoffmann J, Gratton T, Schmidt B. Patient, Family, and Center-Based Factors Associated with Attrition in Neonatal Clinical Research: A Prospective Study. Neonatology 2019; 115:328-334. [PMID: 30836358 PMCID: PMC6657796 DOI: 10.1159/000494105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 09/27/2018] [Indexed: 01/25/2023]
Abstract
BACKGROUND Attrition, or loss to follow-up, presents a significant threat to the integrity and validity of longitudinal clinical research. Little is known about predictors of attrition in neonatal clinical research, and no prior studies have examined how families' experiences participating in research with their infants influences study compliance. OBJECTIVE To identify novel factors that were associated with attrition over 1 year of study follow-up among preterm infants enrolled in the multicenter Prematurity and Respiratory Outcomes Program (PROP) observational study. METHODS At discharge, research coordinators estimated the likelihood of attrition. The parents completed questionnaires about their experience with the study at discharge and at 1 year corrected age. The primary endpoint was completion of 4 PROP interviews during the first year. Logistic models were used to evaluate the associations between infant, family, and center-based characteristics and attrition. RESULTS Among 318 children, 283 (89%) met the primary endpoint. In bivariate analyses, lower maternal education, more people in the household, public insurance, and site were associated with attrition (p < 0.05). Parent survey responses, infant characteristics, and site characteristics were unrelated to attrition. Coordinators' prediction of attrition was associated with completion of early study interviews; this effect waned over time. In multivariable analyses, lower maternal education and more people in the household were the factors most strongly associated with attrition. CONCLUSION Future neonatal research should evaluate novel strategies to decrease the burden associated with study participation and reinforcement of study goals with families who have lower educational levels to facilitate participation and decrease attrition bias.
Collapse
Affiliation(s)
- Sara B DeMauro
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA, .,University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,
| | - Scarlett L Bellamy
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Dornsife School of Public Health, Drexel University, Philadelphia, Pennsylvania, USA
| | - Melissa Fernando
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Julie Hoffmann
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Teresa Gratton
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Barbara Schmidt
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
39
|
Ren CL, Feng R, Davis SD, Eichenwald E, Jobe A, Moore PE, Panitch HB, Sharp JK, Kisling J, Clem C. Tidal Breathing Measurements at Discharge and Clinical Outcomes in Extremely Low Gestational Age Neonates. Ann Am Thorac Soc 2018; 15:1311-1319. [PMID: 30088802 PMCID: PMC6322016 DOI: 10.1513/annalsats.201802-112oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/07/2018] [Indexed: 11/20/2022] Open
Abstract
RATIONALE The relationship between respiratory function at hospital discharge and the severity of later respiratory disease in extremely low gestational age neonates is not well defined. OBJECTIVES To test the hypothesis that tidal breathing measurements near the time of hospital discharge differ between extremely premature infants with bronchopulmonary dysplasia (BPD) or respiratory disease in the first year of life and those without these conditions. METHODS Study subjects were part of the PROP (Prematurity and Respiratory Outcomes Program) study, a longitudinal cohort study of infants born at less than 29 gestational weeks followed from birth to 1 year of age. Respiratory inductance plethysmography was used for tidal breathing measurements before and after inhaled albuterol 1 week before anticipated hospital discharge. Infants were breathing spontaneously and were receiving less than or equal to 1 L/min nasal cannula flow at 21% to 100% fraction of inspired oxygen. A survey of respiratory morbidity was administered to caregivers at 3, 6, 9, and 12 months corrected age to assess for respiratory disease. We compared tidal breathing measurements in infants with and without BPD (oxygen requirement at 36 wk) and with and without respiratory disease in the first year of life. Measurements were also performed in a comparison cohort of term infants. RESULTS A total of 765 infants survived to 36 weeks postmenstrual age, with research-quality tidal breathing data in 452 out of 564 tested (80.1%). Among these 452 infants, the rate of postdischarge respiratory disease was 65.7%. Compared with a group of 18 term infants, PROP infants had abnormal tidal breathing patterns. However, there were no clinically significant differences in tidal breathing measurements in PROP infants who had BPD or who had respiratory disease in the first year of life compared with those without these diagnoses. Bronchodilator response was not significantly associated with respiratory disease in the first year of life. CONCLUSIONS Extremely premature infants receiving less than 1 L/min nasal cannula support at 21% to 100% fraction of inspired oxygen have tidal breathing measurements that differ from term infants, but these measurements do not differentiate those preterm infants who have BPD or will have respiratory disease in the first year of life from those who do not. Clinical trial registered with www.clinicaltrials.gov (NCT01435187).
Collapse
Affiliation(s)
- Clement L. Ren
- Riley Hospital for Children and Indiana University, Indianapolis, Indiana
| | - Rui Feng
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Stephanie D. Davis
- Riley Hospital for Children and Indiana University, Indianapolis, Indiana
| | - Eric Eichenwald
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Alan Jobe
- Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Paul E. Moore
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | - Jeff Kisling
- Riley Hospital for Children and Indiana University, Indianapolis, Indiana
| | - Charles Clem
- Riley Hospital for Children and Indiana University, Indianapolis, Indiana
| | - on behalf of the Prematurity and Respiratory Outcomes Program*
- Riley Hospital for Children and Indiana University, Indianapolis, Indiana
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Children’s Hospital Medical Center, Cincinnati, Ohio
- Vanderbilt University School of Medicine, Nashville, Tennessee
- Texas Children’s Hospital, Houston, Texas; and
- Washington University, St. Louis, Missouri
| |
Collapse
|
40
|
Torgerson DG, Ballard PL, Keller RL, Oh SS, Huntsman S, Hu D, Eng C, Burchard EG, Ballard RA. Ancestry and genetic associations with bronchopulmonary dysplasia in preterm infants. Am J Physiol Lung Cell Mol Physiol 2018; 315:L858-L869. [PMID: 30113228 PMCID: PMC6295513 DOI: 10.1152/ajplung.00073.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 01/19/2023] Open
Abstract
Bronchopulmonary dysplasia in premature infants is a common and often severe lung disease with long-term sequelae. A genetic component is suspected but not fully defined. We performed an ancestry and genome-wide association study to identify variants, genes, and pathways associated with survival without bronchopulmonary dysplasia in 387 high-risk infants treated with inhaled nitric oxide in the Trial of Late Surfactant study. Global African genetic ancestry was associated with increased survival without bronchopulmonary dysplasia among infants of maternal self-reported Hispanic white race/ethnicity [odds ratio (OR) = 4.5, P = 0.01]. Admixture mapping found suggestive outcome associations with local African ancestry at chromosome bands 18q21 and 10q22 among infants of maternal self-reported African-American race/ethnicity. For all infants, the top individual variant identified was within the intron of NBL1, which is expressed in midtrimester lung and is an antagonist of bone morphogenetic proteins ( rs372271081 , OR = 0.17, P = 7.4 × 10-7). The protective allele of this variant was significantly associated with lower nitric oxide metabolites in the urine of non-Hispanic white infants ( P = 0.006), supporting a role in the racial differential response to nitric oxide. Interrogating genes upregulated in bronchopulmonary dysplasia lungs indicated association with variants in CCL18, a cytokine associated with fibrosis and interstitial lung disease, and pathway analyses implicated variation in genes involved in immune/inflammatory processes in response to infection and mechanical ventilation. Our results suggest that genetic variation related to lung development, drug metabolism, and immune response contribute to individual and racial/ethnic differences in respiratory outcomes following inhaled nitric oxide treatment of high-risk premature infants.
Collapse
Affiliation(s)
- Dara G Torgerson
- Department of Pediatrics, University of California , San Francisco, California
| | - Philip L Ballard
- Department of Pediatrics, University of California , San Francisco, California
| | - Roberta L Keller
- Department of Pediatrics, University of California , San Francisco, California
| | - Sam S Oh
- Department of Medicine, University of California , San Francisco, California
| | - Scott Huntsman
- Department of Medicine, University of California , San Francisco, California
| | - Donglei Hu
- Department of Medicine, University of California , San Francisco, California
| | - Celeste Eng
- Department of Medicine, University of California , San Francisco, California
| | - Esteban G Burchard
- Department of Medicine, University of California , San Francisco, California
- Department of Bioengineering and Therapeutic Sciences, University of California , San Francisco, California
| | - Roberta A Ballard
- Department of Pediatrics, University of California , San Francisco, California
| |
Collapse
|
41
|
Hamvas A, Feng R, Bi Y, Wang F, Bhattacharya S, Mereness J, Kaushal M, Cotten CM, Ballard PL, Mariani TJ. Exome sequencing identifies gene variants and networks associated with extreme respiratory outcomes following preterm birth. BMC Genet 2018; 19:94. [PMID: 30342483 PMCID: PMC6195962 DOI: 10.1186/s12863-018-0679-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 10/01/2018] [Indexed: 12/28/2022] Open
Abstract
Background Previous studies have identified genetic variants associated with bronchopulmonary dysplasia (BPD) in extremely preterm infants. However, findings with genome-wide significance have been rare, and not replicated. We hypothesized that whole exome sequencing (WES) of premature subjects with extremely divergent phenotypic outcomes could facilitate the identification of genetic variants or gene networks contributing disease risk. Results The Prematurity and Respiratory Outcomes Program (PROP) recruited a cohort of > 765 extremely preterm infants for the identification of markers of respiratory morbidity. We completed WES on 146 PROP subjects (85 affected, 61 unaffected) representing extreme phenotypes of early respiratory morbidity. We tested for association between disease status and individual common variants, screened for rare variants exclusive to either affected or unaffected subjects, and tested the combined association of variants across gene loci. Pathway analysis was performed and disease-related expression patterns were assessed. Marginal association with BPD was observed for numerous common and rare variants. We identified 345 genes with variants unique to BPD-affected preterm subjects, and 292 genes with variants unique to our unaffected preterm subjects. Of these unique variants, 28 (19 in the affected cohort and 9 in unaffected cohort) replicate a prior WES study of BPD-associated variants. Pathway analysis of sets of variants, informed by disease-related gene expression, implicated protein kinase A, MAPK and Neuregulin/epidermal growth factor receptor signaling. Conclusions We identified novel genes and associated pathways that may play an important role in susceptibility/resilience for the development of lung disease in preterm infants. Electronic supplementary material The online version of this article (10.1186/s12863-018-0679-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aaron Hamvas
- Department of Pediatrics, Northwestern University, Chicago, IL, USA. .,Ann and Robert H. Lurie Children's Hospital of Chicago and Northwestern University, Chicago, IL, USA.
| | - Rui Feng
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yingtao Bi
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Fan Wang
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jared Mereness
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Madhurima Kaushal
- Center for Biomedical Informatics, Washington University, St. Louis, MO, USA
| | | | - Philip L Ballard
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Thomas J Mariani
- Department of Pediatrics, University of Rochester, Rochester, NY, USA. .,Division of Neonatology and Pediatric Molecular and Personalized Medicine Program University of Rochester Medical Center, 601 Elmwood Ave, Box 850, Rochester, NY, 14642, USA.
| | | |
Collapse
|
42
|
Higgins RD, Jobe AH, Koso-Thomas M, Bancalari E, Viscardi RM, Hartert TV, Ryan RM, Kallapur SG, Steinhorn RH, Konduri GG, Davis SD, Thebaud B, Clyman RI, Collaco JM, Martin CR, Woods JC, Finer NN, Raju TNK. Bronchopulmonary Dysplasia: Executive Summary of a Workshop. J Pediatr 2018; 197:300-308. [PMID: 29551318 PMCID: PMC5970962 DOI: 10.1016/j.jpeds.2018.01.043] [Citation(s) in RCA: 552] [Impact Index Per Article: 78.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/27/2017] [Accepted: 01/12/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Rosemary D Higgins
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD.
| | - Alan H Jobe
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH
| | - Marion Koso-Thomas
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD
| | - Eduardo Bancalari
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL
| | - Rose M Viscardi
- Department of Pediatrics, University of Maryland Baltimore, Baltimore, MD
| | - Tina V Hartert
- Department of Medicine, Division of Allergy, Pulmonology and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology Professor of Pediatrics David Geffen School of Medicine at UCLA Mattel Children's Hospital UCLA, Los Angeles, CA
| | - Robin H Steinhorn
- Children's National Health System, Department of Pediatrics, Washington, DC
| | - Girija G Konduri
- Department of Pediatrics, Cardiovascular Research Center and Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI
| | - Stephanie D Davis
- Section of Pediatric Pulmonology, Allergy and Sleep Medicine, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN
| | - Bernard Thebaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ontario, Canada; Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ontario, Canada; Department of Cellular and Molecular Biology, University of Ottawa, Ontario, Canada
| | - Ronald I Clyman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Joseph M Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Camilia R Martin
- Department of Neonatology and Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jason C Woods
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Neil N Finer
- Department of Pediatrics, University of California at San Diego, San Diego, CA
| | - Tonse N K Raju
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD
| |
Collapse
|
43
|
Stone C, Qiu Y, Kurland IJ, Slaughter JC, Moore P, Cook-Mills J, Hartert T, Aschner JL. Effect of Maternal Smoking on Plasma and Urinary Measures of Vitamin E Isoforms in the First Month after Extreme Preterm Birth. J Pediatr 2018; 197:280-285.e3. [PMID: 29398053 PMCID: PMC5971015 DOI: 10.1016/j.jpeds.2017.12.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022]
Abstract
We examined the effect of maternal smoking on plasma and urinary levels of vitamin E isoforms in preterm infants. Maternal smoking during pregnancy decreased infant plasma alpha- and gamma-tocopherol concentrations at 1 week and 4 weeks, with 45% of infants of smokers deficient in alpha-tocopherol at 1 month after birth.
Collapse
Affiliation(s)
- Cosby Stone
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN.
| | - Yunping Qiu
- Department of Medicine, Stable Isotope and Metabolomics Core Facility, Diabetes Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Irwin J. Kurland
- Department of Medicine, Stable Isotope and Metabolomics Core Facility, Diabetes Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - James C Slaughter
- Vanderbilt University School of Medicine, Department of Biostatistics, Nashville, Tennessee, USA
| | - Paul Moore
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Joan Cook-Mills
- Division of Allergy-Immunology, Department of Medicine, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Tina Hartert
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Judy L. Aschner
- Division of Neonatology, Department of Pediatrics, Albert Einstein College of Medicine and the Children’s Hospital at Montefiore, Bronx, New York, USA
| |
Collapse
|
44
|
Levy PT, Patel MD, Choudhry S, Hamvas A, Singh GK. Evidence of Echocardiographic Markers of Pulmonary Vascular Disease in Asymptomatic Infants Born Preterm at One Year of Age. J Pediatr 2018; 197:48-56.e2. [PMID: 29625733 PMCID: PMC5970955 DOI: 10.1016/j.jpeds.2018.02.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To test the hypothesis that echocardiographic markers of pulmonary vascular disease (PVD) exist in asymptomatic infants born preterm at 1-year corrected age. STUDY DESIGN We conducted a prospective cohort study of 80 infants born preterm (<29 weeks of gestation) and 100 age- and weight-matched infants born at term and compared broad-based conventional and quantitative echocardiographic measures of pulmonary hemodynamics at 1-year corrected age. Pulmonary artery acceleration time (PAAT), a validated index of pulmonary vascular resistance, arterial pressure, and compliance, was used to assess pulmonary hemodynamics. Lower PAAT is indicative of PVD. Subanalyses were performed in infants with bronchopulmonary dysplasia (BPD, n = 48, 59%) and/or late-onset pulmonary hypertension (n = 12, 15%). RESULTS At 1 year, there were no differences between conventional measures of pulmonary hypertension in the infants born at term and preterm. All infants born preterm had significantly lower values of PAAT than infants born at term (73 ± 8 milliseconds vs 98 ± 5 milliseconds, P < .001). Infants born preterm with BPD had even lower PAAT than those without BPD (69 ± 5 milliseconds vs 79 ± 4 milliseconds, P < .01). The degree of PVD at 1-year corrected age was inversely related to gestation in all infants born preterm. Data analysis included adjustment for ventricular function and other confounding factors. CONCLUSIONS In comparison with infants born at term, infants born preterm exhibit abnormal PAAT at 1-year corrected age irrespective of neonatal lung disease status, suggesting the existence of PVD beyond infancy. PAAT measurements offer a reliable, noninvasive tool for screening and longitudinal monitoring of pulmonary hemodynamics in infants.
Collapse
Affiliation(s)
- Philip T Levy
- Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, MO.
| | - Meghna D Patel
- Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, MO
| | - Swati Choudhry
- Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, MO
| | - Aaron Hamvas
- Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Gautam K Singh
- Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, MO
| |
Collapse
|
45
|
Blaisdell CJ, Troendle J, Zajicek A. Acute Responses to Diuretic Therapy in Extremely Low Gestational Age Newborns: Results from the Prematurity and Respiratory Outcomes Program Cohort Study. J Pediatr 2018; 197:42-47.e1. [PMID: 29599068 PMCID: PMC5970973 DOI: 10.1016/j.jpeds.2018.01.066] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/06/2018] [Accepted: 01/24/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine if daily respiratory status improved more in extremely low gestational age (GA) premature infants after diuretic exposure compared with those not exposed in modern neonatal intensive care units. STUDY DESIGN The Prematurity and Respiratory Outcomes Program (PROP) was a multicenter observational cohort study of 835 extremely premature infants, GAs of 230/7-286/7 weeks, enrolled in the first week of life from 13 US tertiary neonatal intensive care units. We analyzed the PROP study daily medication and respiratory support records of infants ≤34 weeks postmenstrual age. We determined whether there was a temporal association between the administration of diuretics and an acute change in respiratory status in premature infants in the neonatal intensive care unit, using an ordered categorical ranking of respiratory status. RESULTS Infants in the diuretic exposed group of PROP were of lower mean GA and lower mean birth weight (P < .0001). Compared with infants unexposed to diuretics, the probability (adjusted for infant characteristics including GA, birth weight, sex, and respiratory status before receiving diuretics) that the exposed infants were on a higher level of respiratory support was significantly greater (OR, >1) for each day after the initial day of diuretic exposure. CONCLUSIONS Our analysis did not support the ability of diuretics to substantially improve the extremely premature infant's respiratory status. Further study of both safety and efficacy of diuretics in this setting are warranted. TRIAL REGISTRATION Clinicaltrials.gov: NCT01435187.
Collapse
Affiliation(s)
- Carol J. Blaisdell
- Division of Lung Diseases, National, Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD. Dr. Blaisdell is now Senior Program Official of the Environmental influences on Child Health Outcomes (ECHO) Program Office, Office of the Director, National Institutes of Health, 6011 Executive Boulevard, Suite 305, Bethesda, MD
| | - James Troendle
- Office of Biostatistics Research, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Anne Zajicek
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6100 Executive Boulevard, Suite 4A01, MSC 7510, Bethesda, MD 20892-7510. Dr. Zajicek is now the Deputy Director of the Office of Clinical Research, Office of the Director, National Institutes of Health, 1 Center Drive Room 208A, Bethesda, MD 20892-0155
| | | |
Collapse
|
46
|
Stone CA, McEvoy CT, Aschner JL, Kirk A, Rosas-Salazar C, Cook-Mills JM, Moore PE, Walsh WF, Hartert TV. Update on Vitamin E and Its Potential Role in Preventing or Treating Bronchopulmonary Dysplasia. Neonatology 2018; 113:366-378. [PMID: 29514147 PMCID: PMC5980725 DOI: 10.1159/000487388] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022]
Abstract
Vitamin E is obtained only through the diet and has a number of important biological activities, including functioning as an antioxidant. Evidence that free radicals may contribute to pathological processes such as bronchopulmonary dysplasia (BPD), a disease of prematurity associated with increased lung injury, inflammation and oxidative stress, led to trials of the antioxidant vitamin E (α-tocopherol) to prevent BPD with variable results. These trials were all conducted at supraphysiologic doses and 2 of these trials utilized a formulation containing a potentially harmful excipient. Since 1991, when the last of these trials was conducted, both neonatal management strategies for minimizing oxygen and ventilator-related lung injury and our understanding of vitamin E isoforms in respiratory health have advanced substantially. It is now known that there are differences between the effects of vitamin E isoforms α-tocopherol and γ-tocopherol on the development of respiratory morbidity and inflammation. What is not known is whether improvements in physiologic concentrations of individual or combinations of vitamin E isoforms during pregnancy or following preterm birth might prevent or reduce BPD development. The answers to these questions require adequately powered studies targeting pregnant women at risk of preterm birth or their premature infants immediately following birth, especially in certain subgroups that are at increased risk of vitamin E deficiency (e.g., smokers). The objective of this review is to compile, update, and interpret what is known about vitamin E isoforms and BPD since these first studies were conducted, and suggest future research directions.
Collapse
Affiliation(s)
- Cosby A Stone
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cindy T McEvoy
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Judy L Aschner
- Division of Neonatology, Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, Bronx, New York, USA
| | - Ashudee Kirk
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christian Rosas-Salazar
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joan M Cook-Mills
- Division of Allergy-Immunology, Department of Medicine, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Paul E Moore
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William F Walsh
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tina V Hartert
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
47
|
O'Connor MG, Suthar D, Vera K, Slaughter JC, Maitre NL, Steele S, Beller A, Fike CD, Aschner JL, Moore PE, Austin ED. Pulmonary hypertension in the premature infant population: Analysis of echocardiographic findings and biomarkers. Pediatr Pulmonol 2018; 53:302-309. [PMID: 29168320 PMCID: PMC5815883 DOI: 10.1002/ppul.23913] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/18/2017] [Accepted: 10/22/2017] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Extremely low gestational age neonates (ELGANs) are at risk for pulmonary hypertension (PH). We hypothesized that PH, defined by echocardiogram at 36 weeks gestational age (GA), would associate with respiratory morbidity, increased oxidant stress, and reduced nitric oxide production. STUDY DESIGN ELGANs in the Vanderbilt fraction of the Prematurity and Respiratory Outcomes Program (PROP) who had echocardiograms at 36 ± 1 weeks GA were studied. Echocardiogram features of PH were compared with clinical characteristics as well as markers of oxidant stress and components of the nitric oxide pathway. Biomarkers were obtained at enrollment (median day 3), 7, 14, and 28 days of life. RESULTS Sixty of 172 infants had an echocardiogram at 36 weeks; 11 had evidence of PH. Infants did not differ by PH status in regards to demographics, respiratory morbidity, or oxidant stress. However, odds of more severe PH were significantly higher in infants with higher nitric oxide metabolites (NOx) at enrollment and with a lower citrulline level at day 7. CONCLUSIONS Respiratory morbidity may not always associate with PH at 36 weeks among ELGANs. However, components of nitric oxide metabolism are potential biologic markers of PH in need of further study.
Collapse
Affiliation(s)
- Michael G O'Connor
- Division of Pediatric Pulmonary, Allergy, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Divya Suthar
- Division of Pediatric Cardiology, Vanderbilt University, Nashville, Tennessee
| | - Kimberly Vera
- Division of Pediatric Cardiology, Vanderbilt University, Nashville, Tennessee
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | | | - Steven Steele
- Division of Neonatology, Vanderbilt University, Nashville, Tennessee
| | - Amy Beller
- Division of Neonatology, Vanderbilt University, Nashville, Tennessee
| | - Candice D Fike
- Division of Neonatology, University of Utah, Salt Lake City, Utah
| | - Judy L Aschner
- Divisionof Neonatology, Albert Einstein College of Medicine, Bronx, New York
| | - Paul E Moore
- Division of Pediatric Pulmonary, Allergy, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Eric D Austin
- Division of Pediatric Pulmonary, Allergy, and Immunology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
48
|
Scheible KM, Emo J, Laniewski N, Baran AM, Peterson DR, Holden-Wiltse J, Bandyopadhyay S, Straw AG, Huyck H, Ashton JM, Tripi KS, Arul K, Werner E, Scalise T, Maffett D, Caserta M, Ryan RM, Reynolds AM, Ren CL, Topham DJ, Mariani TJ, Pryhuber GS. T cell developmental arrest in former premature infants increases risk of respiratory morbidity later in infancy. JCI Insight 2018; 3:96724. [PMID: 29467329 DOI: 10.1172/jci.insight.96724] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/17/2018] [Indexed: 12/31/2022] Open
Abstract
The inverse relationship between gestational age at birth and postviral respiratory morbidity suggests that infants born preterm (PT) may miss a critical developmental window of T cell maturation. Despite a continued increase in younger PT survivors with respiratory complications, we have limited understanding of normal human fetal T cell maturation, how ex utero development in premature infants may interrupt normal T cell development, and whether T cell development has an effect on infant outcomes. In our longitudinal cohort of 157 infants born between 23 and 42 weeks of gestation, we identified differences in T cells present at birth that were dependent on gestational age and differences in postnatal T cell development that predicted respiratory outcome at 1 year of age. We show that naive CD4+ T cells shift from a CD31-TNF-α+ bias in mid gestation to a CD31+IL-8+ predominance by term gestation. Former PT infants discharged with CD31+IL8+CD4+ T cells below a range similar to that of full-term born infants were at an over 3.5-fold higher risk for respiratory complications after NICU discharge. This study is the first to our knowledge to identify a pattern of normal functional T cell development in later gestation and to associate abnormal T cell development with health outcomes in infants.
Collapse
Affiliation(s)
| | | | | | - Andrea M Baran
- Department of Biostatistics and Computational Biology, and
| | | | | | | | - Andrew G Straw
- Department of Biostatistics and Computational Biology, and
| | | | | | | | - Karan Arul
- Undergraduate Campus, University of Rochester, Rochester, New York, USA
| | | | | | | | | | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anne Marie Reynolds
- Department of Pediatrics, State University of New York, University at Buffalo, Buffalo, New York, USA
| | - Clement L Ren
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | |
Collapse
|
49
|
Baker CD. Biomarkers of pulmonary hypertension after preterm birth: The IMPROV study. Pediatr Pulmonol 2018; 53:124-125. [PMID: 29266849 DOI: 10.1002/ppul.23929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Christopher D Baker
- Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
50
|
|