1
|
Pharande P, Sehgal A, Menahem S. Cardiovascular Sequelae of Bronchopulmonary Dysplasia in Preterm Neonates Born before 32 Weeks of Gestational Age: Impact of Associated Pulmonary and Systemic Hypertension. J Cardiovasc Dev Dis 2024; 11:233. [PMID: 39195141 DOI: 10.3390/jcdd11080233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common respiratory disorder of prematurity for infants born before 32 weeks of gestational age (GA). Early and prolonged exposure to chronic hypoxia and inflammation induces pulmonary hypertension (PH) with the characteristic features of a reduced number and increased muscularisation of the pulmonary arteries resulting in an increase in the pulmonary vascular resistance (PVR) and a fall in their compliance. BPD and BPD-associated pulmonary hypertension (BPD-PH) together with systemic hypertension (sHTN) are chronic cardiopulmonary disorders which result in an increased mortality and long-term problems for these infants. Previous studies have predominantly focused on the pulmonary circulation (right ventricle and its function) and developing management strategies accordingly for BPD-PH. However, recent work has drawn attention to the importance of the left-sided cardiac function and its impact on BPD in a subset of infants arising from a unique pathophysiology termed postcapillary PH. BPD infants may have a mechanistic link arising from chronic inflammation, cytokines, oxidative stress, catecholamines, and renin-angiotensin system activation along with systemic arterial stiffness, all of which contribute to the development of BPD-sHTN. The focus for the treatment of BPD-PH has been improvement of the right heart function through pulmonary vasodilators. BPD-sHTN and a subset of postcapillary PH may benefit from afterload reducing agents such as angiotensin converting enzyme inhibitors. Preterm infants with BPD-PH are at risk of later cardiac and respiratory morbidities as young adults. This paper reviews the current knowledge of the pathophysiology, diagnosis, and treatment of BPD-PH and BPD-sHTN. Current knowledge gaps and emerging new therapies will also be discussed.
Collapse
Affiliation(s)
- Pramod Pharande
- Monash Newborn, Monash Children's Hospital, 246 Clayton Road, Clayton, Melbourne, VIC 3168, Australia
- Department of Pediatrics, Monash University, Melbourne, VIC 3800, Australia
| | - Arvind Sehgal
- Monash Newborn, Monash Children's Hospital, 246 Clayton Road, Clayton, Melbourne, VIC 3168, Australia
- Department of Pediatrics, Monash University, Melbourne, VIC 3800, Australia
| | - Samuel Menahem
- Department of Pediatrics, Monash University, Melbourne, VIC 3800, Australia
- Paediatric and Foetal Cardiac Units, Monash Medical Centre, Melbourne, VIC 3168, Australia
- Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
2
|
Chang YT, Liu JR, Chen WM, Tseng CN, See LC. First-year outcomes of very low birth weight preterm singleton infants with hypoxemic respiratory failure treated with milrinone and inhaled nitric oxide (iNO) compared to iNO alone: A nationwide retrospective study. PLoS One 2024; 19:e0297137. [PMID: 38722851 PMCID: PMC11081351 DOI: 10.1371/journal.pone.0297137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/28/2023] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Inhaled nitric oxide (iNO) has a beneficial effect on hypoxemic respiratory failure. The increased use of concurrent iNO and milrinone was observed. We aimed to report the trends of iNO use in the past 15 years in Taiwan and compare the first-year outcomes of combining iNO and milrinone to the iNO alone in very low birth weight preterm (VLBWP) infants under mechanical ventilation. METHODS This nationwide cohort study enrolled preterm singleton infants with birth weight <1500g treated with iNO from 2004 to 2019. Infants were divided into two groups, with a combination of intravenous milrinone (Group 2, n = 166) and without milrinone (Group 1, n = 591). After propensity score matching (PSM), each group's sample size is 124. The primary outcomes were all-cause mortality and the respiratory condition, including ventilator use and duration. The secondary outcomes were preterm morbidities within one year after birth. RESULTS After PSM, more infants in Group 2 needed inotropes. The mortality rate was significantly higher in Group 2 than in Group 1 from one month after birth till 1 year of age (55.1% vs. 13.5%) with the adjusted hazard ratio of 4.25 (95%CI = 2.42-7.47, p <0.001). For infants who died before 36 weeks of postmenstrual age (PMA), Group 2 had longer hospital stays compared to Group 1. For infants who survived after 36 weeks PMA, the incidence of moderate and severe bronchopulmonary dysplasia (BPD) was significantly higher in Group 2 than in Group 1. For infants who survived until one year of age, the incidence of pneumonia was significantly higher in Group 2 (28.30%) compared to Group 1 (12.62%) (p = 0.0153). CONCLUSION Combined treatment of iNO and milrinone is increasingly applied in VLBWP infants in Taiwan. This retrospective study did not support the benefits of combining iNO and milrinone on one-year survival and BPD prevention. A future prospective study is warranted.
Collapse
Affiliation(s)
- Ya-Ting Chang
- Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Jia-Rou Liu
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Wei-Min Chen
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chi-Nan Tseng
- Department of Cardiac Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Lai-Chu See
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
- Biostatistics Core Laboratory, Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
3
|
Gurram Venkata SKR, Lodha A, Hicks M, Jain A, Lapointe A, Makary H, Kanungo J, Lee KS, Ye X, Shah PS, Soraisham AS. Neurodevelopmental outcomes of preterm neonates receiving rescue inhaled nitric oxide in the first week of age: a cohort study. Arch Dis Child Fetal Neonatal Ed 2024; 109:211-216. [PMID: 37890983 DOI: 10.1136/archdischild-2023-325418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/11/2023] [Indexed: 10/29/2023]
Abstract
OBJECTIVE To assess the neurodevelopmental outcomes of preterm neonates who received inhaled nitric oxide (iNO) in the first week of age for hypoxaemic respiratory failure (HRF). METHODS In this retrospective cohort study, we included neonates born at <29 weeks gestational age (GA) between January 2010 and December 2018 who had a neurodevelopmental assessment at 18-24 months corrected age (CA) at one of the Canadian Neonatal Follow-Up Network clinics. The primary outcome was neurodevelopmental impairment (NDI). We performed propensity score-matched analysis to compare the outcomes of those who received and did not receive iNO. RESULTS Of the 5612 eligible neonates, 460 (8.2%) received iNO in the first week of age. Maternal age, receipt of antenatal corticosteroids, GA and birth weight were lower in the iNO group compared with the no-iNO group. Neonates in the iNO group had higher illness severity scores and higher rates of preterm prolonged rupture of membranes and were small for GA. Severe brain injury, bronchopulmonary dysplasia and mortality were higher in the iNO group. Of the 4889 survivors, 3754 (77%) neonates had follow-up data at 18-24 months CA. After propensity score matching, surviving infants who received rescue iNO were not associated with higher odds of NDI (adjusted OR 1.34; 95% CI 0.85 to 2.12). CONCLUSIONS In preterm neonates <29 weeks GA with HRF, rescue iNO use was not associated with worse neurodevelopmental outcomes among survivors who were assessed at 18-24 months CA.
Collapse
Affiliation(s)
| | - Abhay Lodha
- Pediatrics, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Matthew Hicks
- Pediatrics, University of Alberta Faculty of Medicine & Dentistry, Edmonton, Alberta, Canada
| | - Amish Jain
- Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anie Lapointe
- Pediatrics, University of Montreal Faculty of Medicine, Montreal, Quebec, Canada
| | - Hala Makary
- Pediatrics, Dalhousie University Faculty of Medicine, Halifax, Nova Scotia, Canada
| | - Jaideep Kanungo
- Pediatrics, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Kyong-Soon Lee
- Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiang Ye
- Maternal-Infant Care Research Centre, Mount Sinai Hospital Pediatrics, Toronto, Ontario, Canada
| | - Prakesh S Shah
- Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Amuchou S Soraisham
- Pediatrics, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Feng Z, Wu X, Xu X, Cui Q, Wu F. Efficacy of inhaled nitric oxide in preterm infants ≤ 34 weeks: a systematic review and meta-analysis of randomized controlled trials. Front Pharmacol 2024; 14:1268795. [PMID: 38273818 PMCID: PMC10808707 DOI: 10.3389/fphar.2023.1268795] [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: 07/28/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Background: The effect of inhaled nitric oxide (iNO) in neonates >34 weeks on improving respiration is well documented. However, the efficacy of iNO in preterm infants ≤34 weeks remains controversial. Objectives: The main purpose of this review is to assess the effectiveness and safety of iNO treatment in preterm infants ≤34 weeks. Search methods: We systematically searched PubMed, Embase and Cochrane Libraries from their inception to 1 June 2023. We also reviewed the reference lists of retrieved studies. Selection criteria: Our study involved randomized controlled trials on preterm infants ≤34 weeks, especially those receiving iNO treatment, and mainly assessed outcomes such as bronchopulmonary dysplasia (BPD) and mortality. Two authors independently reviewed these trials, extracted data, and evaluated study biases. Disagreements were resolved by consensus. We used the GRADE method to assess evidence quality. Results: Our research included a total of 17 studies involving 4,080 neonates and 7 follow-up studies. The synthesis of results showed that in neonates, iNO treatment reduced the incidence of BPD (RR: 0.92; 95% CI: 0.86-0.98). It also decreased the composite outcome of death or BPD (RR: 0.94; 95% CI: 0.90-0.98), without increasing the risk of short-term (such as intraventricular hemorrhage, periventricular leukomalacia) and long-term neurological outcomes (including Bayley mental developmental index <70, cerebral palsy and neurodevelopmental impairment). Furthermore, iNO did not significantly affect other neonatal complications like sepsis, pulmonary hemorrhage, necrotizing enterocolitis, and symptomatic patent ductus arteriosus. Subgroup analysis revealed that iNO significantly reduced BPD incidence in neonates at 36 weeks under specific intervention conditions, including age less than 3 days, birth weight over 1,000 g, iNO dose of 10 ppm or higher, or treatment duration exceeding 7 days (p < 0.05). Conclusion: Inhaled NO reduced the incidence of BPD in neonates at 36 weeks of gestation, and the effect of the treatment depended on neonatal age, birth weight, duration and dose of iNO. Therefore, iNO can be considered a promising treatment for the potential prevention of BPD in premature infants. More data, however, would be needed to support nitric oxide registration in this specific patient population, to minimize its off-label use.
Collapse
Affiliation(s)
- Zhoushan Feng
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
| | - Xiaohong Wu
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
| | - Xiaona Xu
- Department of Obstetrics and Gynecology, School of Medicine, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qiliang Cui
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
| | - Fan Wu
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China
| |
Collapse
|
5
|
Dumpa V, Avulakunta I, Bhandari V. Respiratory management in the premature neonate. Expert Rev Respir Med 2023; 17:155-170. [PMID: 36803028 DOI: 10.1080/17476348.2023.2183843] [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: 02/23/2023]
Abstract
INTRODUCTION Advances in neonatal care have made possible the increased survival of extremely preterm infants. Even though there is widespread recognition of the harmful effects of mechanical ventilation on the developing lung, its use has become imperative in the management of micro-/nano-preemies. There is an increased emphasis on the use of less-invasive approaches such as minimally invasive surfactant therapy and non-invasive ventilation that have been proven to result in improved outcomes. AREAS COVERED Here, we review the evidence-based practices surrounding the respiratory management of extremely preterm infants including delivery room interventions, invasive and non-invasive ventilation approaches, and specific ventilator strategies in respiratory distress syndrome and bronchopulmonary dysplasia. Adjuvant relevant respiratory pharmacotherapies used in preterm neonates are also discussed. EXPERT OPINION Early use of non-invasive ventilation and use of less invasive surfactant administration are key strategies in the management of respiratory distress syndrome in preterm infants. Ventilator management in bronchopulmonary dysplasia must be tailored according to the individual phenotype. There is strong evidence to start caffeine early to improve respiratory outcomes, but evidence is lacking on the use of other pharmacological agents in preterm neonates, and an individualized approach has to be considered for their use.
Collapse
Affiliation(s)
- Vikramaditya Dumpa
- Division of Neonatology, Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Indirapriya Avulakunta
- Division of Neonatology, Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Vineet Bhandari
- Division of Neonatology, Department of Pediatrics, Cooper Medical School of Rowan University, the Children's Regional Hospital at Cooper, Camden, NJ, USA
| |
Collapse
|
6
|
Oka S, Nishimura E, Ozawa J, Haga M, Miyahara N, Sakatani S, Minamitani Y, Namba F. Therapeutic response of iNO in preterm infants with hypoxemic respiratory failure. Pediatr Int 2023; 65:e15423. [PMID: 36412230 DOI: 10.1111/ped.15423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Inhaled nitric oxide (iNO) has been used as a rescue treatment for preterm infants with hypoxemic respiratory failure (HRF). However, its effectiveness remains debatable. Thus, in this study, we aimed to examine the impact of iNO therapy on HRF in extremely preterm infants. METHODS A retrospective observational study was performed. Extremely preterm infants admitted to our neonatal intensive care unit who received iNO therapy later in their postnatal life were included. The oxygen saturation index (OSI) was used as an index of the severity of respiratory failure. RESULTS In total, 30 extremely preterm infants were included in this study. Oxygenation was enhanced after the administration of iNO in infants with HRF. The OSI decreased by more than 20% in 12 patients (40%, positive responder) and did not decrease in 17 patients (57%, negative responder) within the first 6 h of treatment. The iNO initiation day was the significant independent factor associated with a positive response to iNO therapy in extremely preterm infants with HRF. CONCLUSIONS iNO therapy was effective in enhancing oxygenation in extremely preterm infants with HRF. Earlier use of iNO was the significant factor associated with a positive therapeutic response to iNO, implying that iNO may be more effective in pulmonary vessels which are less damaged by shorter-term mechanical ventilation.
Collapse
Affiliation(s)
- Shuntaro Oka
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Eri Nishimura
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Junichi Ozawa
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Mitsuhiro Haga
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Naoyuki Miyahara
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Shun Sakatani
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan.,Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Yohei Minamitani
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Fumihiko Namba
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| |
Collapse
|
7
|
Nakanishi H, Isayama T, Kokubo M, Hirano S, Kusuda S. Inhaled Nitric Oxide Therapy in the Post-Acute Phase in Extremely Preterm Infants: A Japanese Cohort Study. J Pediatr 2023; 252:61-67.e5. [PMID: 36116533 DOI: 10.1016/j.jpeds.2022.07.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the trends in inhaled nitric oxide (iNO) utilization in the late phase of hospitalization in a large Japanese cohort of extremely preterm infants and evaluate its benefit on long-term outcomes. STUDY DESIGN This was a retrospective multicenter cohort study of 15 977 extremely preterm infants born at <28 weeks of gestational age between 2003 and 2016, in the Neonatal Research Network, Japan. Demographic characteristics, morbidity, and mortality were compared between extremely preterm infants with and without post-acute iNO therapy. Multivariable logistic analysis was performed to determine factors associated with post-acute iNO and its impact on neurodevelopmental outcomes at 3 years of age. RESULTS Post-acute iNO utilization rates increased from 0.3% in 2009 to 1.9% in 2016, even under strict insurance coverage rules starting in 2009. Gestational age (1-week increment; aOR 0.82, 95% CI 0.76-0.88), small for gestational age (1.47, 1.08-1.99), histologic chorioamnionitis (1.50, 1.21-1.86), 5-minute Apgar score <4 (1.51, 1.10-2.07), air leak (1.92, 1.30-2.83), and bubbly/cystic appearance on chest X-Ray (1.68, 1.37-2.06) were associated with post-acute iNO. Post-acute iNO was not associated with neurodevelopmental outcomes at 3 years of age. CONCLUSIONS The increasing post-acute iNO utilization rate among extremely preterm infants has been concurrent with improved survival rates of extremely preterm infants in Japan. Infants treated with post-acute iNO had more severe disease and complications than the comparison group, but there were no differences in neurodevelopmental outcome at 3 years. This suggests post-acute iNO may benefit extremely preterm infants.
Collapse
Affiliation(s)
- Hidehiko Nakanishi
- Research and Development Center for New Medical Frontiers, Department of Advanced Medicine, Division of Neonatal Intensive Care Medicine, Kitasato University School of Medicine, Kanagawa, Japan.
| | - Tetsuya Isayama
- Division of Neonatology, National Center for Child Health and Development, Tokyo, Japan
| | - Masayo Kokubo
- Division of Neonatology, Nagano Children's Hospital, Nagano, Japan
| | - Shinya Hirano
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Satoshi Kusuda
- Department of Pediatrics, Kyorin University, Tokyo, Japan
| |
Collapse
|
8
|
Extracellular Signal-Regulated Kinase 1 Alone Is Dispensable for Hyperoxia-Mediated Alveolar and Pulmonary Vascular Simplification in Neonatal Mice. Antioxidants (Basel) 2022; 11:antiox11061130. [PMID: 35740027 PMCID: PMC9219973 DOI: 10.3390/antiox11061130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 02/04/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a morbid lung disease distinguished by lung alveolar and vascular simplification. Hyperoxia, an important BPD causative factor, increases extracellular signal-regulated kinases (ERK)-1/2 expression, whereas decreased lung endothelial cell ERK2 expression reduces angiogenesis and potentiates hyperoxia-mediated BPD in mice. However, ERK1′s role in experimental BPD is unclear. Thus, we hypothesized that hyperoxia-induced experimental BPD would be more severe in global ERK1-knockout (ERK1-/-) mice than their wild-type (ERK1+/+ mice) littermates. We determined the extent of lung development, ERK1/2 expression, inflammation, and oxidative stress in ERK1-/- and ERK1+/+ mice exposed to normoxia (FiO2 21%) or hyperoxia (FiO2 70%). We also quantified the extent of angiogenesis and hydrogen peroxide (H2O2) production in hyperoxia-exposed neonatal human pulmonary microvascular endothelial cells (HPMECs) with normal and decreased ERK1 signaling. Compared with ERK1+/+ mice, ERK1-/- mice displayed increased pulmonary ERK2 activation upon hyperoxia exposure. However, the extent of hyperoxia-induced inflammation, oxidative stress, and interrupted lung development was similar in ERK1-/- and ERK1+/+ mice. ERK1 knockdown in HPMECs increased ERK2 activation at baseline, but did not affect in vitro angiogenesis and hyperoxia-induced H2O2 production. Thus, we conclude ERK1 is dispensable for hyperoxia-induced experimental BPD due to compensatory ERK2 activation.
Collapse
|
9
|
Siahanidou T, Spiliopoulou C. Pharmacological Neuroprotection of the Preterm Brain: Current Evidence and Perspectives. Am J Perinatol 2022; 39:479-491. [PMID: 32961562 DOI: 10.1055/s-0040-1716710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite improvements in viability, the long-term neurodevelopmental outcomes of preterm babies remain serious concern as a significant percentage of these infants develop neurological and/or intellectual impairment, and they are also at increased risk of psychiatric illnesses later in life. The current challenge is to develop neuroprotective approaches to improve adverse outcomes in preterm survivors. The purpose of this review was to provide an overview of the current evidence on pharmacological agents targeting the neuroprotection of the preterm brain. Among them, magnesium sulfate, given antenatally to pregnant women with imminent preterm birth before 30 to 34 weeks of gestation, as well as caffeine administered to preterm infants after birth, exhibited neuroprotective effects for human preterm brain. Erythropoietin treatment of preterm infants did not result in neuroprotection at 2 years of age in two out of three published large randomized controlled trials; however, long-term follow-up of these infants is needed to come to definite conclusions. Further studies are also required to assess whether melatonin, neurosteroids, inhaled nitric oxide, allopurinol, or dietary supplements (omega-3 fatty acids, choline, curcumin, etc.) could be implemented as neuroprotectants in clinical practice. Furthermore, other pharmacological agents showing promising signs of neuroprotective efficacy in preclinical studies (growth factors, hyaluronidase inhibitors or treatment, antidiabetic drugs, cannabidiol, histamine-H3 receptor antagonists, etc.), as well as stem cell- or exosomal-based therapies and nanomedicine, may prove useful in the future as potential neuroprotective approaches for human preterm brain. KEY POINTS: · Magnesium and caffeine have neuroprotective effects for the preterm brain.. · Follow-up of infants treated with erythropoietin is needed.. · Neuroprotective efficacy of several drugs in animals needs to be shown in humans..
Collapse
Affiliation(s)
- Tania Siahanidou
- Neonatal Unit of the First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
10
|
魏 思. Recent research on the effect of common treatments given in the perinatal period on neurodevelopment in offspring. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:332-338. [PMID: 35351267 PMCID: PMC8974644 DOI: 10.7499/j.issn.1008-8830.2111002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
The perinatal period is the key period for the development of brain and central nervous system, and different events in this period will have a profound influence on brain development. Glucocorticoids, antibiotics, magnesium sulfate, caffeine, pulmonary surfactant, and mild hypothermia treatment are commonly used drugs or treatment methods in the perinatal period and are closely associated with the prognosis of neonatal neurodevelopment. This article reviews the latest research on the effect of perinatal treatments on neonatal neurodevelopment, so as to provide a reference for clinical decision making.
Collapse
|
11
|
Peluso AM, Othman HF, Karnati S, Sammour I, Aly HZ. Epidemiologic evaluation of inhaled nitric oxide use among neonates with gestational age less than 35 weeks. Pediatr Pulmonol 2022; 57:427-434. [PMID: 34842352 DOI: 10.1002/ppul.25775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/16/2021] [Accepted: 11/26/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND AND OBJECTIVES The use of inhaled nitric oxide (iNO) in +late preterm and term infants with pulmonary hypertension is Food and Drug Administration (FDA) approved and has improved outcomes and survival. iNO use is not FDA approved for preterm infants and previous studies show no mortality benefit. The objectives were 1) to determine the usage of iNO among preterm neonates <35 weeks before and after the 2010 National Institutes of Health consensus statement and 2) to evaluate characteristics and outcomes among preterm neonates who received iNO. METHODS This is a population-based cross-sectional study. Billing and procedure codes were used to determine iNO usage. Data were queried from the National Inpatient Sample from 2004 to 2016. Neonates were included if gestational age was <35 weeks. The epochs were spilt into 2004-2010 (Epoch 1) and 2011-2016 (Epoch 2). Prevalence of iNO use, mortality, bronchopulmonary dysplasia (BPD), intraventricular hemorrhage, length of stay, mechanical ventilation, and cost of hospitalization. RESULTS There were 4865 preterm neonates <35 weeks who received iNO. There was a significant increase in iNO use during Epoch 2 (p < 0.001). There was significantly higher use in Epoch 2 among neonates small for gestational age (SGA) 2.3% versus 7.2%, congenital heart disease (CHD) 11.1% versus 18.6%, and BPD 35.2% versus 46.8%. Mortality was significantly lower in Epoch 2 19.8% versus 22.7%. CONCLUSION Usage of iNO was higher after the release of the consensus statement. The increased use of iNO among preterm neonates may be targeted at specific high-risk populations such as SGA and CHD neonates. There was lower mortality in Epoch 2; however, the cost was doubled.
Collapse
Affiliation(s)
- Allison M Peluso
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, Ohio, USA
| | - Hasan F Othman
- Department of Pediatrics, Michigan State University/Sparrow Health System, Lansing, Michigan, USA
| | - Sreenivas Karnati
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, Ohio, USA
| | - Ibrahim Sammour
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, Ohio, USA
| | - Hany Z Aly
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, Ohio, USA
| |
Collapse
|
12
|
Muehlbacher T, Bassler D, Bryant MB. Evidence for the Management of Bronchopulmonary Dysplasia in Very Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2021; 8:298. [PMID: 33924638 PMCID: PMC8069828 DOI: 10.3390/children8040298] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Very preterm birth often results in the development of bronchopulmonary dysplasia (BPD) with an inverse correlation of gestational age and birthweight. This very preterm population is especially exposed to interventions, which affect the development of BPD. OBJECTIVE The goal of our review is to summarize the evidence on these daily procedures and provide evidence-based recommendations for the management of BPD. METHODS We conducted a systematic literature research using MEDLINE/PubMed on antenatal corticosteroids, surfactant-replacement therapy, caffeine, ventilation strategies, postnatal corticosteroids, inhaled nitric oxide, inhaled bronchodilators, macrolides, patent ductus arteriosus, fluid management, vitamin A, treatment of pulmonary hypertension and stem cell therapy. RESULTS Evidence provided by meta-analyses, systematic reviews, randomized controlled trials (RCTs) and large observational studies are summarized as a narrative review. DISCUSSION There is strong evidence for the use of antenatal corticosteroids, surfactant-replacement therapy, especially in combination with noninvasive ventilation strategies, caffeine and lung-protective ventilation strategies. A more differentiated approach has to be applied to corticosteroid treatment, the management of patent ductus arteriosus (PDA), fluid-intake and vitamin A supplementation, as well as the treatment of BPD-associated pulmonary hypertension. There is no evidence for the routine use of inhaled bronchodilators and prophylactic inhaled nitric oxide. Stem cell therapy is promising, but should be used in RCTs only.
Collapse
Affiliation(s)
- Tobias Muehlbacher
- Department of Neonatology, University Hospital Zurich, 8091 Zurich, Switzerland; (D.B.); (M.B.B.)
| | | | | |
Collapse
|
13
|
Nitric oxide and the brain. Part 2: Effects following neonatal brain injury-friend or foe? Pediatr Res 2021; 89:746-752. [PMID: 32563184 DOI: 10.1038/s41390-020-1021-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/30/2020] [Accepted: 06/02/2020] [Indexed: 12/30/2022]
Abstract
Nitric oxide (NO) has critical roles in a wide variety of key biologic functions and has intricate transport mechanisms for delivery to key distal tissues under normal conditions. However, NO also plays important roles during disease processes, such as hypoxia-ischemia, asphyxia, neuro-inflammation, and retinopathy of prematurity. The effects of exogenous NO on the developing neonatal brain remain controversial. Inhaled NO (iNO) can be neuroprotective or toxic depending on a variety of factors, including cellular redox state, underlying disease processes, duration of treatment, and dose. This review identifies key gaps in knowledge that should prompt further investigation into the possible role of iNO as a therapeutic agent after injury to the brain. IMPACT: NO is a key signal mediator in the neonatal brain with neuroprotective and neurotoxic properties. iNO, a commonly used medication, has significant effects on the neonatal brain. Dosing, duration, and timing of administration of iNO can affect the developing brain. This review article summarizes the roles of NO in association with various disease processes that impact neonates, such as brain hypoxia-ischemia, asphyxia, retinopathy of prematurity, and neuroinflammation. The impact of this review is that it clearly describes gaps in knowledge, and makes the case for further, targeted studies in each of the identified areas.
Collapse
|
14
|
Qasim A, Jain SK. Milrinone Use in Persistent Pulmonary Hypertension of the Newborn. Neoreviews 2021; 21:e165-e178. [PMID: 32123121 DOI: 10.1542/neo.21-3-e165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Failure of the normal transition from in utero to ex utero physiology leads to "persistent" pulmonary hypertension of the newborn (PPHN). PPHN is frequently associated with low systemic blood pressure and low cardiac output because of increased right ventricular afterload and myocardial dysfunction. The general management of newborns with PPHN is geared toward maintenance of normothermia, normal serum electrolytes, normal intravascular volume, correction of acidosis, adequate sedation/analgesia, adequate ventilation and oxygenation with optimal lung recruitment, and avoidance of hyperoxia. Inotropic and vasoactive agents are commonly initiated early to increase cardiac output, maintain adequate systemic blood pressure, and enhance oxygen delivery to the tissue. Unfortunately, there is not much evidence on the choice, timing of initiation, dosing, monitoring, and titrating of vasoactive agents in this patient population. In this review, we will discuss the pathophysiology of PPHN and review the use of inotropic, lusitropic, and vasoactive agents in the management of PPHN, with particular attention to milrinone.
Collapse
Affiliation(s)
- Amna Qasim
- Department of Pediatrics, Division of Pediatric Cardiology, Texas Children's Hospital/Baylor College of Medicine, Houston, TX
| | - Sunil K Jain
- Department of Pediatrics, Division of Neonatology, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
15
|
A quality improvement project improving the value of iNO utilization in preterm and term infants. J Perinatol 2021; 41:164-172. [PMID: 32770031 DOI: 10.1038/s41372-020-0768-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/26/2020] [Accepted: 07/28/2020] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Inhaled NO (iNO) is used in the NICU for management of hypoxemic respiratory failure. The cost of iNO is significant and does not consistently improve outcomes in infants <34 weeks. PROJECT DESIGN Our team used The Model for Improvement to design a quality improvement project to utilize iNO for appropriate indications, ensure response to therapy and initiate timely weaning. The project was carried out at a Level IV NICU and successful interventions spread to a smaller Level III NICU. RESULTS This project demonstrated significant improvement in all measures; total iNO hours per month, average iNO hours per patient, and the percentage of prolonged iNO courses. With an estimated cost of $115/h, the cost per patient for iNO use declined by half from $21,620 to $10,580. CONCLUSIONS Our team improved the value of iNO utilization at our institution and spread successful interventions to another NICU in our network.
Collapse
|
16
|
Menon RT, Shrestha AK, Barrios R, Reynolds C, Shivanna B. Tie-2 Cre-Mediated Deficiency of Extracellular Signal-Regulated Kinase 2 Potentiates Experimental Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension in Neonatal Mice. Int J Mol Sci 2020; 21:ijms21072408. [PMID: 32244398 PMCID: PMC7177249 DOI: 10.3390/ijms21072408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 01/09/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH) is a significant lung morbidity of infants, and disrupted lung angiogenesis is a hallmark of this disease. We observed that extracellular signal-regulated kinases (ERK) 1/2 support angiogenesis in vitro, and hyperoxia activates ERK1/2 in fetal human pulmonary microvascular endothelial cells (HPMECs) and in neonatal murine lungs; however, their role in experimental BPD and PH is unknown. Therefore, we hypothesized that Tie2 Cre-mediated deficiency of ERK2 in the endothelial cells of neonatal murine lungs would potentiate hyperoxia-induced BPD and PH. We initially determined the role of ERK2 in in vitro angiogenesis using fetal HPMECs. To disrupt endothelial ERK2 signaling in the lungs, we decreased ERK2 expression by breeding ERK2flox/flox mice with Tie-Cre mice. One-day-old endothelial ERK2-sufficient (eERK2+/+) or –deficient (eERK2+/−) mice were exposed to normoxia or hyperoxia (FiO2 70%) for 14 d. We then performed lung morphometry, gene and protein expression studies, and echocardiography to determine the extent of inflammation, oxidative stress, and development of lungs and PH. The knockdown of ERK2 in HPMECs decreased in vitro angiogenesis. Hyperoxia increased lung inflammation and oxidative stress, decreased lung angiogenesis and alveolarization, and induced PH in neonatal mice; however, these effects were augmented in the presence of Tie2-Cre mediated endothelial ERK2 deficiency. Therefore, we conclude that endothelial ERK2 signaling is necessary to mitigate hyperoxia-induced experimental BPD and PH in neonatal mice. Our results indicate that endothelial ERK2 is a potential therapeutic target for the management of BPD and PH in infants.
Collapse
Affiliation(s)
- Renuka T. Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.M.); (A.K.S.)
| | - Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.M.); (A.K.S.)
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA;
| | - Corey Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.M.); (A.K.S.)
- Correspondence: ; Tel.: +1-832-824-6474; Fax: +1-832-825-3204
| |
Collapse
|
17
|
Lin H, Wang X. The effects of gasotransmitters on bronchopulmonary dysplasia. Eur J Pharmacol 2020; 873:172983. [PMID: 32017936 DOI: 10.1016/j.ejphar.2020.172983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023]
Abstract
Bronchopulmonary dysplasia (BPD), which remains a major clinical problem for preterm infants, is caused mainly by hyperoxia, mechanical ventilation and inflammation. Many approaches have been developed with the aim of decreasing the incidence of or alleviating BPD, but effective methods are still lacking. Gasotransmitters, a type of small gas molecule that can be generated endogenously, exert a protective effect against BPD-associated lung injury; nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) are three such gasotransmitters. The protective effects of NO have been extensively studied in animal models of BPD, but the results of these studies are inconsistent with those of clinical trials. NO inhalation seems to have no effect on BPD, although side effects have been reported. NO inhalation is not recommended for BPD treatment in preterm infants, except those with severe pulmonary hypertension. Both CO and H2S decreased lung injury in BPD rodent models in preclinical studies. Another small gas molecule, hydrogen, exerts a protective effect against BPD. The nuclear factor erythroid-derived 2 (Nrf2)/heme oxygenase-1 (HO-1) axis seems to play a central role in the protective effect of these gasotransmitters on BPD. Gasotransmitters play important roles in mammals, but further clinical trials are needed to explore their effects on BPD.
Collapse
Affiliation(s)
- Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Xinbao Wang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
18
|
Sherlock LG, Wright CJ, Kinsella JP, Delaney C. Inhaled nitric oxide use in neonates: Balancing what is evidence-based and what is physiologically sound. Nitric Oxide 2019; 95:12-16. [PMID: 31866361 DOI: 10.1016/j.niox.2019.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Inhaled nitric oxide is a powerful therapeutic used in neonatology. Its use is evidenced-based for term and near-term infants with persistent pulmonary hypertension; however, it is frequently used off-label both in term and preterm babies. This article reviews the off-label uses of iNO in infants. Rationale is discussed for a selective application of iNO based on physiologically guided principles, and new research avenues are considered.
Collapse
Affiliation(s)
- Laurie G Sherlock
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - John P Kinsella
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Cassidy Delaney
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
19
|
Abstract
Rates of bronchopulmonary dysplasia (BPD) are increasing. After preterm birth, there are important developmental periods in which neonates are more vulnerable to stressful events. These periods are opportunities for pharmacologic interventions. Many drugs remain inadequately tested and no new drugs have been approved in more than 25 years for BPD prevention or therapy. More progress is needed in defining appropriate end points based on the pathophysiology of BPD and postdischarge chronic pulmonary insufficiency of prematurity and to develop effective new drugs. In addition, much work is needed to better define perinatal factors, early postnatal findings, and physiologic phenotypes or endotypes.
Collapse
|
20
|
Preventing bronchopulmonary dysplasia: new tools for an old challenge. Pediatr Res 2019; 85:432-441. [PMID: 30464331 DOI: 10.1038/s41390-018-0228-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/12/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most prevalent chronic lung disease in infants and presents as a consequence of preterm birth. Due to the lack of effective preventive and treatment strategies, BPD currently represents a major therapeutic challenge that requires continued research efforts at the basic, translational, and clinical levels. However, not all very low birth weight premature babies develop BPD, which suggests that in addition to known gestational age and intrauterine and extrauterine risk factors, other unknown factors must be involved in this disease's development. One of the main goals in BPD research is the early prediction of very low birth weight infants who are at risk of developing BPD in order to initiate the adequate preventive strategies. Other benefits of determining the risk of BPD include providing prognostic information and stratifying infants for clinical trial enrollment. In this article, we describe new opportunities to address BPD's complex pathophysiology by identifying prognostic biomarkers and develop novel, complex in vitro human lung models in order to develop effective therapies. These therapies for protecting the immature lung from injury can be developed by taking advantage of recent scientific progress in -omics, 3D organoids, and regenerative medicine.
Collapse
|
21
|
Bancalari E, Jain D. Bronchopulmonary Dysplasia: 50 Years after the Original Description. Neonatology 2019; 115:384-391. [PMID: 30974430 DOI: 10.1159/000497422] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 11/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the few diseases in neonatal medicine that has continued to evolve since its first description about 50 years ago. Over these years, advancements in neonatal medicine such as antenatal steroids and exogenous surfactant therapy have significantly reduced neonatal mortality and lowered the limits of viability for preterm infants. Although the incidence of BPD continues to be high, especially in extremely low birth weight infants, the clinical picture has evolved into a milder disease with low mortality or significant morbidities. This new BPD is the result of complex interactions between altered alveolar and vascular development, injury by ante- and postnatal pathogenic factors, and reparative processes in the lung. There has been significant progress in our understanding of risk factors for BPD, but challenges persist in its definition, and in finding effective preventive strategies. There are promising developments with newer preventive interventions such as mesenchymal stem cells, exosomes, immunomodulators, and growth factors, but they are still in preclinical stage. The future challenges include finding ways to define BPD based on the severity of lung pathology, which can better predict long-term outcomes, development of early predictors of lung disease, and finding innovative and evidence-based preventive and management strategies.
Collapse
Affiliation(s)
- Eduardo Bancalari
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA,
| | - Deepak Jain
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
22
|
Collura CA, Mara KC, Weaver AL, Clark RH, Carey WA. Outcomes of early inhaled nitric oxide use in premature African American neonates. J Perinatol 2018; 38:1657-1665. [PMID: 30275545 DOI: 10.1038/s41372-018-0232-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/02/2018] [Accepted: 08/15/2018] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Meta-analysis of individual-patient clinical trial data suggests that inhaled nitric oxide (iNO) improves respiratory outcomes in premature African American neonates. We hypothesized that early iNO therapy would be associated with lower mortality and less chronic lung disease (CLD) in extremely premature African American neonates. STUDY DESIGN We conducted a retrospective cohort study of propensity score- and race-matched neonates 22-29 weeks gestation who were mechanically ventilated for treatment of respiratory distress and associated pulmonary hypertension (RDS + PPHN). We evaluated the association of iNO within 7 days of life with in-hospital mortality and CLD, using Cox proportional hazards regression and logistic regression, respectively. RESULT Among 178 matched pairs of African American patients, iNO was not associated with lower mortality (HR = 0.94, 95% CI 0.69-1.30) or less CLD (OR = 0.94, 95% CI 0.47-1.87). CONCLUSIONS Early, off-label iNO use is not associated with improved outcomes in premature African American neonates with RDS + PPHN.
Collapse
Affiliation(s)
| | - Kristin C Mara
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Amy L Weaver
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Reese H Clark
- Center for Research, Education & Quality, Pediatrix Medical Group, Sunrise, FL, USA
| | - William A Carey
- Division of Neonatal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
23
|
Inhaled nitric oxide as a rescue therapy in a preterm neonate with severe pulmonary hypertension: a case report. Ital J Pediatr 2018; 44:55. [PMID: 29764471 PMCID: PMC5952598 DOI: 10.1186/s13052-018-0494-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/08/2018] [Indexed: 11/19/2022] Open
Abstract
Background Inhaled nitric oxide (iNO) has been approved for the treatment of persistent pulmonary hypertension of the newborn (PPHN) in term and near-term newborns. Its role in the management of persistent pulmonary hypertension in preterm infants is not clear. Although guidelines do not exist, some studies have shown that iNO could be used as a rescue therapy in preterm neonate with severe pulmonary hypertension. Case presentation We describe the case of a preterm neonate, born at 30 + 1 weeks of gestation, with hypoxic respiratory failure not responding to maximal conventional therapy. On the third day of life echocardiography showed severe pulmonary hypertension with right to left shunt and therapy with iNO was started. We achieved a rapid improvement in clinical conditions and pulmonary pressure normalized after 42 h of treatment. Conclusions Moving on a case by case basis, treatment with iNO should be considered as a rescue therapy in preterm newborns with acute hypoxic respiratory failure caused by severe pulmonary hypertension.
Collapse
|
24
|
Rallis D, Deierl A, Atreja G, Chaban B, Banerjee J. The efficacy of inhaled nitric oxide treatment in premature infants with acute pulmonary hypertension. Early Hum Dev 2018; 127:1-5. [PMID: 30205295 DOI: 10.1016/j.earlhumdev.2018.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although inhaled nitric oxide (iNO) therapy in term infants with pulmonary hypertension (PHT) has demonstrated definite benefit, the use of iNO in preterm infants remains inconclusive. AIMS To evaluate the impact of iNO treatment in premature infants with acute PHT. STUDY DESIGN Retrospective cohort. SUBJECTS Infants < 34 weeks' gestational age, admitted during 2010-2016 to two neonatal units, having treated with iNO for confirmed PHT. A positive response was defined by FiO2 reduction ≥20% within 3-h post iNO initiation. Early PHT was defined when developed within the first 72 h of age. OUTCOME MEASURES The primary outcome was the evaluation of the acute response to iNO administration. Secondary outcomes included the comparison of neonatal characteristics and outcomes between positive and negative responders, and early or late PHT infants. RESULTS Of the 55 infants of our cohort, 39 (71%) had a positive response to iNO administration. No differences noted regarding bronchopulmonary dysplasia, intraventricular haemorrhage or other morbidities; however, positive responders had significantly higher survival rate in overall (77 vs 21%, p = 0.001) and within early PHT subgroup (74 vs 33%, p = 0.044). Regression analysis revealed that oligohydramnios (OR 2.834, 95%CI 1.652-6.070) and early PHT (OR 1.953, 95%CI 1.377-2.930) were significantly related with a positive response. CONCLUSIONS Preterm infants with confirmed acute PHT respond in significant proportion to the iNO administration, especially in the background of oligohydramnios or the development of early PHT.
Collapse
Affiliation(s)
- Dimitrios Rallis
- Imperial College Healthcare NHS Trust, Neonatal Unit, London, UK.
| | - Aniko Deierl
- Imperial College Healthcare NHS Trust, Neonatal Unit, London, UK
| | - Gaurav Atreja
- Imperial College Healthcare NHS Trust, Neonatal Unit, London, UK
| | - Badr Chaban
- Imperial College Healthcare NHS Trust, Neonatal Unit, London, UK
| | - Jayanta Banerjee
- Imperial College Healthcare NHS Trust, Neonatal Unit, London, UK
| |
Collapse
|
25
|
Qasim A, Dasgupta S, Aly AM, Jain SK. Sildenafil Use in the Treatment of Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension: A Case Series. AJP Rep 2018; 8:e219-e222. [PMID: 30345157 PMCID: PMC6188884 DOI: 10.1055/s-0038-1673343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/18/2018] [Indexed: 11/02/2022] Open
Abstract
Objective This article studies the role of sildenafil in reducing myocardial stress (measured by serial N-terminal pro b-type natriuretic peptide [NTproBNP] levels) secondary to bronchopulmonary dysplasia-associated pulmonary hypertension (BPD-PH). Study Design This is a case series of three extremely low birth weight infants with severe BPD at 36 weeks' postmenstrual age. All infants had very elevated NTproBNP (> 2,000 ng/dL) levels and echocardiographic evidence of BPD-PH. Sildenafil was started and infants were followed up every 2 weeks clinically along with NTproBNP levels and echocardiograms. Results After 4 weeks of sildenafil treatment, NTproBNP levels decreased significantly in all infants, echocardiograms showed significant improvement in one infant, and respiratory severity score improved significantly in one infant. All infants tolerated sildenafil. Conclusion Sildenafil reduced NTproBNP levels in all infants with BPD-PH but the echocardiographic findings and respiratory scores did not improve consistently. We speculate that this may be due to a delay in diagnosis and initiation of therapy after irreversible pulmonary changes have set in.
Collapse
Affiliation(s)
- Amna Qasim
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Soham Dasgupta
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Ashraf M Aly
- Division of Pediatric Cardiology, University of Texas Medical Branch, Galveston, Texas
| | - Sunil K Jain
- Division of Neonatology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
26
|
de Wijs‐Meijler DPM, Duncker DJ, Danser AHJ, Reiss IKM, Merkus D. Changes in the nitric oxide pathway of the pulmonary vasculature after exposure to hypoxia in swine model of neonatal pulmonary vascular disease. Physiol Rep 2018; 6:e13889. [PMID: 30375198 PMCID: PMC6205946 DOI: 10.14814/phy2.13889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 09/12/2018] [Accepted: 09/16/2018] [Indexed: 11/24/2022] Open
Abstract
Neonatal pulmonary vascular disease (PVD) is increasingly recognized as a disease that complicates the cardiopulmonary adaptations after birth and predisposes to long-term cardiopulmonary disease. There is growing evidence that PVD is associated with disruptions in the nitric oxide (NO)-cGMP-phosphodiesterase 5 (PDE5) pathway. Examination of the functionality of different parts of this pathway is required for better understanding of the pathogenesis of neonatal PVD. For this purpose, the role of the NO-cGMP-PDE5 pathway in regulation of pulmonary vascular function was investigated in vivo, both at rest and during exercise, and in isolated pulmonary small arteries in vitro, in a neonatal swine model with hypoxia-induced PVD. Endothelium-dependent vasodilatation was impaired in piglets with hypoxia-induced PVD both in vivo at rest and in vitro. Moreover, the responsiveness to the NO-donor SNP was reduced in hypoxia-exposed piglets in vivo, while the relaxation to SNP and 8-bromo-cyclicGMP in vitro were unaltered. Finally, PDE5 inhibition-induced pulmonary vasodilatation was impaired in hypoxia-exposed piglets both in vitro and in vivo at rest. During exercise, however, the pulmonary vasodilator effect of PDE5 inhibition was significantly larger in hypoxia-exposed as compared to normoxia-exposed piglets. In conclusion, the impaired endothelium-dependent vasodilatation in piglets with hypoxia-induced PVD was accompanied by reduced responsiveness to NO, potentially caused by altered sensitivity and/or activity of soluble guanylyl cyclase (sGC), resulting in an impaired cGMP production. Our findings in a newborn animal model for neonatal PVD suggests that sGC stimulators/activators may be a novel treatment strategy to alleviate neonatal PVD.
Collapse
Affiliation(s)
- Daphne P. M. de Wijs‐Meijler
- Division of Experimental CardiologyDepartment of CardiologyUniversity Medical Center RotterdamErasmus MCRotterdamThe Netherlands
- Division of NeonatologyDepartment of PediatricsSophia Children's HospitalErasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Dirk J. Duncker
- Division of Experimental CardiologyDepartment of CardiologyUniversity Medical Center RotterdamErasmus MCRotterdamThe Netherlands
| | - A. H. Jan Danser
- Division of PharmacologyDepartment of Internal MedicineErasmus MC University Medical Center RotterdamRotterdamThe Netherlands
| | - Irwin K. M. Reiss
- Division of NeonatologyDepartment of PediatricsSophia Children's HospitalErasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Daphne Merkus
- Division of Experimental CardiologyDepartment of CardiologyUniversity Medical Center RotterdamErasmus MCRotterdamThe Netherlands
| |
Collapse
|
27
|
Donda K, Zambrano R, Moon Y, Percival J, Vaidya R, Dapaah-Siakwan F, Luo S, Duncan MR, Bao Y, Wang L, Qin L, Benny M, Young K, Wu S. Riociguat prevents hyperoxia-induced lung injury and pulmonary hypertension in neonatal rats without effects on long bone growth. PLoS One 2018; 13:e0199927. [PMID: 29990355 PMCID: PMC6038999 DOI: 10.1371/journal.pone.0199927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common and serious chronic lung disease of premature infants. Severe BPD complicated with pulmonary hypertension (PH) increases the mortality of these infants. Riociguat is an allosteric soluble guanylate cyclase stimulator and is approved by the FDA for treating PH in adults. However, it has not been approved for use in neonates due to concern for adverse effects on long bone growth. To address this concern we investigated if administration of riociguat is beneficial in preventing hyperoxia-induced lung injury and PH without side effects on long bone growth in newborn rats. Newborn rats were randomized to normoxia (21% O2) or hyperoxia (85% O2) exposure groups within 24 hours of birth, and received riociguat or placebo by once daily intraperitoneal injections during continuous normoxia or hyperoxia exposure for 9 days. In the hyperoxia control group, radial alveolar count, mean linear intercept and vascular density were significantly decreased, the pathological hallmarks of BPD, and these were accompanied by an increased inflammatory response. There was also significantly elevated vascular muscularization of peripheral pulmonary vessels, right ventricular systolic pressure and right ventricular hypertrophy indicating PH. However, administration of riociguat significantly decreased lung inflammation, improved alveolar and vascular development, and decreased PH during hyperoxia by inducing cGMP production. Additionally, riociguat did not affect long bone growth or structure. These data indicate that riociguat is beneficial in preventing hyperoxia-induced lung injury and PH without affecting long bone growth and structure and hence, suggests riociguat may be a potential novel agent for preventing BPD and PH in neonates.
Collapse
Affiliation(s)
- Keyur Donda
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Ronald Zambrano
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Younghye Moon
- Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Justin Percival
- Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ruben Vaidya
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Fredrick Dapaah-Siakwan
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Shihua Luo
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Matthew R. Duncan
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Yong Bao
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Luqing Wang
- Department of Orthopedic Surgery, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ling Qin
- Department of Orthopedic Surgery, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Merline Benny
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Karen Young
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Shu Wu
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
28
|
Ellsworth KR, Ellsworth MA, Weaver AL, Mara KC, Clark RH, Carey WA. Association of Early Inhaled Nitric Oxide With the Survival of Preterm Neonates With Pulmonary Hypoplasia. JAMA Pediatr 2018; 172:e180761. [PMID: 29800952 PMCID: PMC6137510 DOI: 10.1001/jamapediatrics.2018.0761] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
IMPORTANCE Pulmonary hypoplasia affects a very small percentage of preterm neonates, but its presence is associated with high rates of mortality. OBJECTIVE To determine whether treatment with inhaled nitric oxide during the first week of life was associated with improved in-hospital survival in a cohort of extremely preterm neonates with pulmonary hypoplasia. DESIGN, SETTING, AND PARTICIPANTS This cohort study used data from the Pediatrix Medical Group's Clinical Data Warehouse, a data set containing information from more than 350 neonatal intensive care units in 35 US states and Puerto Rico. Since inhaled nitric oxide was not randomly prescribed, we used 1-to-1 propensity score matching to reduce the imbalance of measured covariates between the 2 treatment groups. The initial, unmatched cohort included singleton neonates who were born between 22 and 29 weeks' gestation, had a birth weight of 400 g or more, were diagnosed with pulmonary hypoplasia as a cause of their respiratory distress, remained free of major anomalies, and were discharged between January 1, 2000, and December 31, 2014. We defined exposure as the initiation of inhaled nitric oxide on day t in days 0 to 7 of the life of a neonate. Each exposed neonate was matched 1-to-1 to a neonate who had not initiated inhaled nitric oxide on a given day. MAIN OUTCOMES AND MEASURES The primary outcome was mortality defined as death prior to transfer or discharge home. Secondary outcomes were any-stage necrotizing enterocolitis, retinopathy of prematurity requiring treatment, chronic lung disease, and periventricular leukomalacia. RESULTS Among 92 635 neonates in our study sample, we identified 767 (0.8%) with pulmonary hypoplasia who met all study inclusion criteria, of whom 185 (0.2%) were exposed to inhaled nitric oxide. Among 151 matched pairs of exposed and unexposed neonates, we did not identify a significant association between inhaled nitric oxide use and mortality (hazard ratio [HR], 0.79; 95% CI, 0.57-1.11). Subgroup analyses of neonates with and without persistent pulmonary hypertension (PPHN) likewise revealed no significant association between inhaled nitric oxide use and mortality (pulmonary hypoplasia with PPHN: HR, 0.67; 95% CI, 0.45-1.01; pulmonary hypoplasia without PPHN: HR, 1.11; 95% CI, 0.61-2.02), but these findings may have been influenced by ascertainment bias. CONCLUSIONS AND RELEVANCE Early treatment with inhaled nitric oxide is not associated with improved survival among extremely preterm neonates with pulmonary hypoplasia. Clinical trials are warranted to clarify the matter.
Collapse
Affiliation(s)
| | | | - Amy L. Weaver
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Kristin C. Mara
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Reese H. Clark
- Center for Research, Education and Quality, Pediatrix Medical Group, Sunrise, Florida
| | - William A. Carey
- Division of Neonatal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
29
|
Hyperoxia Disrupts Extracellular Signal-Regulated Kinases 1/2-Induced Angiogenesis in the Developing Lungs. Int J Mol Sci 2018; 19:ijms19051525. [PMID: 29783779 PMCID: PMC5983575 DOI: 10.3390/ijms19051525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 11/17/2022] Open
Abstract
Hyperoxia contributes to the pathogenesis of bronchopulmonary dysplasia (BPD), a chronic lung disease of infants that is characterized by interrupted alveologenesis. Disrupted angiogenesis inhibits alveologenesis, but the mechanisms of disrupted angiogenesis in the developing lungs are poorly understood. In pre-clinical BPD models, hyperoxia increases the expression of extracellular signal-regulated kinases (ERK) 1/2; however, its effects on the lung endothelial ERK1/2 signaling are unclear. Further, whether ERK1/2 activation promotes lung angiogenesis in infants is unknown. Hence, we tested the following hypotheses: (1) hyperoxia exposure will increase lung endothelial ERK1/2 signaling in neonatal C57BL/6J (WT) mice and in fetal human pulmonary artery endothelial cells (HPAECs); (2) ERK1/2 inhibition will disrupt angiogenesis in vitro by repressing cell cycle progression. In mice, hyperoxia exposure transiently increased lung endothelial ERK1/2 activation at one week of life, before inhibiting it at two weeks of life. Interestingly, hyperoxia-mediated decrease in ERK1/2 activation in mice was associated with decreased angiogenesis and increased endothelial cell apoptosis. Hyperoxia also transiently activated ERK1/2 in HPAECs. ERK1/2 inhibition disrupted angiogenesis in vitro, and these effects were associated with altered levels of proteins that modulate cell cycle progression. Collectively, these findings support our hypotheses, emphasizing that the ERK1/2 pathway is a potential therapeutic target for BPD infants with decreased lung vascularization.
Collapse
|
30
|
Carey WA, Weaver AL, Mara KC, Clark RH. Inhaled Nitric Oxide in Extremely Premature Neonates With Respiratory Distress Syndrome. Pediatrics 2018; 141:peds.2017-3108. [PMID: 29439205 DOI: 10.1542/peds.2017-3108] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Inhaled nitric oxide (iNO) is increasingly prescribed to extremely premature neonates with respiratory distress syndrome (RDS). Most of this off-label use occurs during the first week of life. We studied this practice, hypothesizing that it would not be associated with improved survival. METHODS We queried the Pediatrix Medical Group Clinical Data Warehouse to identify all neonates born at 22 to 29 weeks' gestation from 2004 to 2014. In our study sample, we included singletons who required mechanical ventilation for treatment of RDS and excluded those with anomalies. The primary outcome was death before discharge. Through a sequential risk set approach, each patient who received iNO during the first 7 days of life ("case patient") was matched by using propensity scores to a patient who had not received iNO at a chronological age before the case patient's iNO initiation age (defined as the index age for the matched pair). The association between iNO status and in-hospital mortality was evaluated in a Cox proportional hazards regression model by using age as the time scale with patients entering the risk set at their respective index age. RESULTS Among 37 909 neonates in our study sample, we identified 993 (2.6%) who received iNO. The 2 matched cohorts each contained 971 patients. We did not observe a significant association between iNO exposure and mortality (hazard ratio, 1.08; 95% confidence interval, 0.94-1.25; P = .29). CONCLUSIONS Off-label prescription of iNO is not associated with reduced in-hospital mortality among extremely premature neonates with RDS.
Collapse
Affiliation(s)
| | - Amy L Weaver
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota; and
| | - Kristin C Mara
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota; and
| | - Reese H Clark
- Center for Research, Education and Quality, Pediatrix Medical Group, Sunrise, Florida
| |
Collapse
|
31
|
Tal A, Greenberg D, Av-Gay Y, Golan-Tripto I, Feinstein Y, Ben-Shimol S, Dagan R, Goldbart AD. Nitric oxide inhalations in bronchiolitis: A pilot, randomized, double-blinded, controlled trial. Pediatr Pulmonol 2018; 53:95-102. [PMID: 29178284 DOI: 10.1002/ppul.23905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/11/2017] [Indexed: 11/08/2022]
Abstract
AIM The aims of this pilot study were to determine safety, tolerability (primary outcome) and efficacy (secondary outcome) of high-dose inhaled nitric oxide for the treatment of infants with moderately severe bronchiolitis. METHODS This was a pilot, double-blinded, randomized controlled study (phase IIa). Intermittent inhalations of nitric oxide 160 ppm for 30 min or oxygen/air (control) were given 5 times/day to hospitalized infants (2-11 months) with acute bronchiolitis. Oxygen saturation, methemoglobin, and nitric dioxide (NO2 ) levels and vital signs were monitored. RESULTS Forty-three infants were enrolled. Baseline characteristics were comparable in both study groups. Mean clinical score, comprised of four components: respiratory rate, use of accessory muscles, wheezes and crackles, and % room-air oxygen saturation, was 7.86 (±1.1) and 8.09 (±1.2) in the NO and control groups, respectively, consistent with moderate severity. The overall frequency of adverse events was similar between the groups. Repeated nitric oxide inhalations did not result in increased inhaled NO2 levels or cumulative effect on methemoglobin levels. Secondary outcomes of efficacy were measured by length of hospitalization (LOS) in hours: LOS did not differ between groups. However, in a post-hoc analysis of a subgroup of infants hospitalized for >24 h (n = 24), the median LOS was shorter in the nitric oxide (41.9 h) than in the control group (62.5 h) (P = 0.014). CONCLUSION Our study was unable to detect a difference in side effects using intermittent high-dose nitric-oxide inhalation or supportive treatment alone, in infants with moderate bronchiolitis. Preliminary efficacy outcomes are encouraging.
Collapse
Affiliation(s)
- Asher Tal
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Advanced Inhalation Therapies (A.I.T.) Ltd., Rehovot, Israel
| | - David Greenberg
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Advanced Inhalation Therapies (A.I.T.) Ltd., Rehovot, Israel.,Pediatric Infectious Diseases Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Yossef Av-Gay
- Advanced Inhalation Therapies (A.I.T.) Ltd., Rehovot, Israel.,Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Inbal Golan-Tripto
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yael Feinstein
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Shalom Ben-Shimol
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Pediatric Infectious Diseases Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Ron Dagan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Aviv D Goldbart
- Department of Pediatrics, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
32
|
Stark A, Dammann C, Nielsen HC, Volpe MV. A Pathogenic Relationship of Bronchopulmonary Dysplasia and Retinopathy of Prematurity? A Review of Angiogenic Mediators in Both Diseases. Front Pediatr 2018; 6:125. [PMID: 29951473 PMCID: PMC6008318 DOI: 10.3389/fped.2018.00125] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/16/2018] [Indexed: 01/11/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are common and significant morbidities of prematurely born infants. These diseases have in common altered and pathologic vascular formation in the face of incomplete organ development. Therefore, it is reasonable to question whether factors affecting angiogenesis could have a joint pathogenic role for both diseases. Inhibition or induced expression of a single angiogenic factor is unlikely to be 100% causative or protective of either of BPD or ROP. It is more likely that interactions of multiple factors leading to disordered angiogenesis are present, increasing the likelihood of common pathways in both diseases. This review explores this possibility by assessing the evidence showing involvement of specific angiogenic factors in the vascular development and maldevelopment in each disease. Theoretical interactions of specific factors mutually contributing to BPD and ROP are proposed and, where possible, a timeline of the proposed relationships between BPD and ROP is developed. It is hoped that future research will be inspired by the theories put forth in this review to enhance the understanding of the pathogenesis in both diseases.
Collapse
Affiliation(s)
- Ashley Stark
- Tufts University School of Medicine, Boston, MA, United States
| | - Christiane Dammann
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States.,Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Heber C Nielsen
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States.,Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - MaryAnn V Volpe
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
33
|
Dani C, Corsini I, Cangemi J, Vangi V, Pratesi S. Nitric oxide for the treatment of preterm infants with severe RDS and pulmonary hypertension. Pediatr Pulmonol 2017; 52:1461-1468. [PMID: 29058384 DOI: 10.1002/ppul.23843] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/05/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Inhaled nitric oxide (iNO) cannot be recommended for the routine treatment of respiratory failure in premature neonates, but it has been suggested that the effectiveness of iNO therapy should be further studied in more select preterm infants, such as those with persistent pulmonary hypertension of the newborn (PPHN). OBJECTIVE To evaluate the frequency of PPHN in very preterm infants with severe respiratory distress syndrome (RDS), to assess the effectiveness of iNO in these patients, and to individuate possible predictive factors for the response to iNO in preterm infants with RDS. STUDY DESIGN We retrospectively studied infants <30 weeks of gestational age or birth weight <1250 g, who were affected by severe RDS and treated with iNO during the first week of life. Clinical characteristics of infants with or without echocardiographic diagnosis of PPHN were compared, as well as those of responder or no responder to iNO therapy. Effectiveness of iNO was evaluated by recording changes of MAP, FiO2 , SpO2 /FiO2 ratio, and oxygenation index (OI) before, and 3 ± 1, 6 ± 1, 12 ± 3, 24 ± 6, 48 ± 6, and 72 ± 12 h after beginning therapy. RESULTS We studied 42 (4.6%) infants, of whom 28 (67%) had PPHN and 14 (33%) did not. iNO therapy was associated with improved oxygenation in both the groups but it was quicker in the PPHN than in the no PPHN group. Multivariate analysis showed that FiO2 >0.65, diagnosis of PPHN, and birth weight >750 g independently predicts effectiveness of iNO in very preterm infants with RDS. CONCLUSION We found that PPHN is a frequent complication of severe RDS in very preterm infants and iNO therapy can improve their oxygenation earlier than in infants without PPHN. iNO therapy is not recommended for the routinely treatment of RDS in premature neonates but in cases of concurrent diagnosis of PPHN it should be considered carefully.
Collapse
Affiliation(s)
- Carlo Dani
- Division of Neonatology, Careggi University Hospital of Florence, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Iuri Corsini
- Division of Neonatology, Careggi University Hospital of Florence, Florence, Italy
| | - Jessica Cangemi
- Division of Neonatology, Careggi University Hospital of Florence, Florence, Italy
| | - Venturella Vangi
- Division of Neonatology, Careggi University Hospital of Florence, Florence, Italy
| | - Simone Pratesi
- Division of Neonatology, Careggi University Hospital of Florence, Florence, Italy
| |
Collapse
|
34
|
Baczynski M, Ginty S, Weisz DE, McNamara PJ, Kelly E, Shah P, Jain A. Short-term and long-term outcomes of preterm neonates with acute severe pulmonary hypertension following rescue treatment with inhaled nitric oxide. Arch Dis Child Fetal Neonatal Ed 2017; 102:F508-F514. [PMID: 28483819 DOI: 10.1136/archdischild-2016-312409] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/13/2017] [Accepted: 03/17/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To describe short-term and long-term outcomes of preterm neonates with severe acute pulmonary hypertension (aPHT) in relation to response to rescue inhaled nitric oxide (iNO) therapy. DESIGN Retrospective cohort studyover a 6 year period. SETTING Tertiary neonatal intensive care unit. PATIENTS 89 neonates <35 weeks gestational age (GA) who received rescue iNO for aPHT, including 62 treated at ≤3 days of age (early aPHT). INTERVENTIONS iNO ≥ 1 hour. MAIN OUTCOME MEASURES Positive responders (reduction in fraction of inspired oxygen (FiO2) ≥0.20 within 1 hour of iNO) were compared with non-responders. Primary outcome was survival without moderate-to-severe disability at 18 months of age. RESULTS Mean (SD) GA and birth weight was 27.7 (3.0) weeks and 1077 (473) gm, respectively. Median (IQR) pre-iNO FiO2 was 1.0 (1.0, 1.0). Positive response rate to iNO was 46%. Responders showed improved survival without disability (51% vs 15%; p<0.01), lower mortality (34% vs 71%; p<0.01) and disability among survivors (17% vs 50%; p=0.06). Higher GA (adjusted OR: 1.44 (95% CI 1.10 to 1.89)), aPHT in context of preterm prolonged rupture of membranes (6.26 (95% CI 1.44 to 27.20)) and positive response to rescue iNO (5.81 (95% CI 1.29 to, 26.18)) were independently associated with the primary outcome. Compared with late cases (>3 days of age), early aPHT had a higher response rate to iNO (61% vs 11%; p<0.01) and lower mortality (43% vs 78%; p<0.01). CONCLUSION A positive response to rescue iNO in preterm infants with aPHT is associated with survival benefit, which is not offset by long-term disability.
Collapse
Affiliation(s)
- Michelle Baczynski
- Department of Respiratory Therapy, Mount Sinai Hospital, Toronto, Canada
| | - Shannon Ginty
- Department of Respiratory Therapy, Mount Sinai Hospital, Toronto, Canada
| | - Dany E Weisz
- Department of Newborn and Developmental Paediatrics, Sunnybrook Health Science Center, Toronto, Canada.,Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Patrick J McNamara
- Department of Paediatrics, University of Toronto, Toronto, Canada.,Division of Neonatology, Hospital for Sick Children, Toronto, Canada.,Physiology, University of Toronto, Toronto, Canada
| | - Edmond Kelly
- Department of Paediatrics, University of Toronto, Toronto, Canada.,Department of Paediatrics, Mount Sinai Hospital, Toronto, Canada
| | - Prakeshkumar Shah
- Department of Paediatrics, University of Toronto, Toronto, Canada.,Department of Paediatrics, Mount Sinai Hospital, Toronto, Canada.,Lunnenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Amish Jain
- Department of Paediatrics, University of Toronto, Toronto, Canada.,Physiology, University of Toronto, Toronto, Canada.,Department of Paediatrics, Mount Sinai Hospital, Toronto, Canada.,Lunnenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| |
Collapse
|
35
|
Pulmonary hypertension associated with bronchopulmonary dysplasia in preterm infants. J Reprod Immunol 2017; 124:21-29. [PMID: 29035757 DOI: 10.1016/j.jri.2017.09.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 09/11/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) and BPD-associated pulmonary hypertension (BPD-PH) are chronic inflammatory cardiopulmonary diseases with devastating short- and long-term consequences for infants born prematurely. The immature lungs of preterm infants are ill-prepared to achieve sufficient gas exchange, thus usually necessitating immediate commencement of respiratory support and oxygen supplementation. These therapies are life-saving, but they exacerbate the tissue damage that is inevitably inflicted on a preterm lung forced to perform gas exchange. Together, air-breathing and necessary therapeutic interventions disrupt normal lung development by aggravating pulmonary inflammation and vascular remodelling, thus frequently precipitating BPD and PH via an incompletely understood pathogenic cascade. BPD and BPD-PH share common risk factors, such as low gestational age at birth, fetal growth restriction and perinatal maternal inflammation; however, these risk factors are not unique to BPD or BPD-PH. Occurring in 17-24% of BPD patients, BPD-PH substantially worsens the morbidity and mortality attributable to BPD alone, thus darkening their outlook; for example, BPD-PH entails a mortality of up to 50%. The absence of a safe and effective therapy for BPD and BPD-PH renders neonatal cardiopulmonary disease an area of urgent unmet medical need. Besides the need to develop new therapeutic strategies, a major challenge for clinicians is the lack of a reliable method for identifying babies at risk of developing BPD and BPD-PH. In addition to discussing current knowledge on pathophysiology, diagnosis and treatment of BPD-PH, we highlight emerging biomarkers that could enable clinicians to predict disease-risk and also optimise treatment of BPD-PH in our tiniest patients.
Collapse
|
36
|
Aschner JL, Bancalari EH, McEvoy CT. Can We Prevent Bronchopulmonary Dysplasia? J Pediatr 2017; 189:26-30. [PMID: 28947055 PMCID: PMC5657541 DOI: 10.1016/j.jpeds.2017.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/25/2017] [Accepted: 08/02/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Judy L. Aschner
- Dept of Pediatrics, Albert Einstein College of Medicine and the Children’s Hospital at Montefiore, Bronx NY 10467, USA
| | - Eduardo H. Bancalari
- Dept of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Cindy T. McEvoy
- Dept of Pediatrics, Oregon Health & Science University, Portland, OR 97239-3098, USA
| |
Collapse
|
37
|
Mueller M, Kramer BW. Stem cells and Bronchopulmonary Dysplasia - The five questions: Which cells, when, in which dose, to which patients via which route? Paediatr Respir Rev 2017; 24:54-59. [PMID: 28162941 DOI: 10.1016/j.prrv.2016.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022]
Abstract
Preterm birth is the leading cause of death in newborns and children. Despite advances in perinatology, immature infants continue to face serious risks such chronic respiratory impairment from bronchopulmonary dysplasia (BPD). Current treatment options are insufficient and novel approaches are desperately needed. In recent years stem cells have emerged as potential candidates to treat BPD with mesenchymal stem/stromal cells (MSCs) being particularly promising. MSCs originate from several stem cell niches including bone marrow, skin, or adipose, umbilical cord, and placental tissues. Although the first MSCs clinical trials in BPD are ongoing, multiple questions remain open. In this review, we discuss the question of the optimal cell source (live cells or cell products), route and timing of the transplantation. Furthermore, we discuss MSCs possible capacities including migration, homing, pro-angiogenesis, anti-inflammatory, and tissue-regenerative potential as well.
Collapse
Affiliation(s)
- Martin Mueller
- Department of Obstetrics and Gynecology, University Hospital Bern, Bern, Switzerland; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA; Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands; School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands; School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands.
| |
Collapse
|
38
|
Deepak J, Eduardo B. Prevention of bronchopulmonary dysplasia: current strategies. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:841-851. [PMID: 28774356 PMCID: PMC7390045 DOI: 10.7499/j.issn.1008-8830.2017.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/28/2017] [Indexed: 06/07/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the few diseases affecting premature infants that have continued to evolve since its first description about half a century ago. The current form of BPD, a more benign and protracted respiratory failure in extremely preterm infants, is in contrast to the original presentation of severe respiratory failure with high mortality in larger premature infants. This new BPD is end result of complex interplay of various antenatal and postnatal factors causing lung injury and subsequent abnormal repair leading to altered alveolar and vascular development. The change in clinical and pathologic picture of BPD over time has resulted in new challenges in developing strategies for its prevention and management. While some of these strategies like Vitamin A supplementation, caffeine and volume targeted ventilation have stood the test of time, others like postnatal steroids are being reexamined with great interest in last few years. It is quite clear that BPD is unlikely to be eliminated unless some miraculous strategy cures prematurity. The future of BPD prevention will probably be a combination of antenatal and postnatal strategies acting on multiple pathways to minimize lung injury and abnormal repair as well as promote normal alveolar and vascular development.
Collapse
Affiliation(s)
- Jain Deepak
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Bancalari Eduardo
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| |
Collapse
|
39
|
Ma L, Zhou P, Neu J, Lin HC. Potential Nutrients for Preventing or Treating Bronchopulmonary Dysplasia. Paediatr Respir Rev 2017; 22:83-88. [PMID: 27843119 DOI: 10.1016/j.prrv.2016.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/01/2016] [Accepted: 08/29/2016] [Indexed: 10/21/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a frequent complication occurring in extremely preterm infants. Despite recent advances in newborn medicine, the incidence of BPD does not appear to have changed markedly, and specific treatments and prevention strategies are still lacking. Nutrition plays an important role in normal lung development and maturation. Malnutrition may delay somatic growth and new alveoli development, thus aggravating pulmonary injury involved in the pathogenesis of BPD. However, few nutrients have been investigated for their potential to mitigate the pathogenesis of BPD. In this article, we reviewed the recent progress in research on potential nutrients useful for the prevention or treatment of BPD, including glutamine, cysteine and N-acetylcysteine, L-arginine and L-citrulline, long chain polyunsaturated fatty acids (LCPUFAs), inositol, selenium, and some antioxidant vitamins including vitamin A. Current evidence shows that vitamin A and LCPUFA can prevent BPD, and that L-citrulline might provide a new method to treat chronic pulmonary hypertension associated with BPD in premature infants. The effects of other nutrients on BPD prevention need to be further studied.
Collapse
Affiliation(s)
- Liya Ma
- Department of Neonatology, Shenzhen Baoan Maternal and Child Health Hospital, China.
| | - Ping Zhou
- Department of Neonatology, Shenzhen Baoan Maternal and Child Health Hospital, China.
| | - Josef Neu
- Department of Pediatrics, University of Florida, U.S.A..
| | - Hung-Chih Lin
- Department of Pediatrics, Children's Hospital and School of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
40
|
Mueller M, Wolfs TGA, Schoeberlein A, Gavilanes AWD, Surbek D, Kramer BW. Mesenchymal stem/stromal cells-a key mediator for regeneration after perinatal morbidity? Mol Cell Pediatr 2016; 3:6. [PMID: 26869264 PMCID: PMC4751100 DOI: 10.1186/s40348-016-0034-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Perinatal complications in both term- and preterm-born infants are a leading cause of neonatal morbidities and mortality. Infants face different challenges in the neonatal intensive care unit with long-term morbidities such as perinatal brain injury and bronchopulmonary dysplasia being particularly devastating. While advances in perinatal medicine have improved our understanding of the pathogenesis, effective therapies to prevent and/or reduce the severity of these disorders are still lacking. The potential of mesenchymal stem/stromal cell (MSC) therapy has emerged during the last two decades, and an increasing effort is conducted to address brain- and lung-related morbidities in neonates at risk. Various studies support the notion that MSCs have protective effects. MSCs are an easy source and may be readily available after birth in a clinical setting. MSCs' mechanisms of action are diverse, including migration and homing, release of growth factors and immunomodulation, and the potential to replace injured cells. Here, we review the pathophysiology of perinatally acquired brain and lung injuries and focus on MSCs as potential candidates for therapeutic strategies summarizing preclinical and clinical evidence.
Collapse
Affiliation(s)
- Martin Mueller
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Tim G A Wolfs
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
| | - Andreina Schoeberlein
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Antonio W D Gavilanes
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- Institute of Biomedicine, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador.
- Department of Neuropsychology, Division Neuroscience, School of Mental Health and neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| | - Daniel Surbek
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
- Department of Neuropsychology, Division Neuroscience, School of Mental Health and neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
41
|
Guo X, Sun Y, Miao J, Cui M, Wang J, Han S. No inhalation in combination with high frequency ventilation treatment in the treatment of neonatal severe respiratory failure. Pak J Med Sci 2016; 32:1218-1223. [PMID: 27882025 PMCID: PMC5103137 DOI: 10.12669/pjms.325.10682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective: To discuss over NO inhalation (iNO) in combination with high frequency ventilation treatment in relieving clinical symptoms and respiratory state of patients with neonatal severe respiratory failure. Methods: Ninety newborns with severe respiratory failure who received treatment in our hospital were selected for this study. They were divided into research group and control group according to visiting time. Patients in the control group were given conventional treatment in combination with high-frequency oscillatory ventilation, while patients in the research group were given iNO for treatment additionally besides the treatment the same as the control group. Changes of respiratory function indexes and arterial blood gas indexes of patients in the two groups were compared. Mechanical ventilation time, time of oxygen therapy and the length of hospital stay were recorded. Besides, postoperative outcome and the incidence of complications were analyzed. Results: After treatment, the level of PaO2 of both groups significantly improved, and respiratory function indexes such as partial pressure of carbon dioxide in artery (PaCO2), oxygenation index (OI), fraction of inspiration O2 (FiO2) and mean arterial pressure (MAP) decreased (P<0.05); the improvement of various indexes of the research group was more obvious than that of the control group (P<0.05). Mechanical ventilation time, oxygen therapy time and the length of hospital stay of the research group was much shorter than those of the control group. The incidence of complications in the two groups had no statistically significant difference (P>0.05), but the clinical outcome of the research group was better than that of the control group. Conclusion: NO inhalation in combination with high frequency ventilation for treating neonatal severe respiratory failure is effective in improving blood gas index and respiratory function, enhance cure rate, and reduce the incidence of complications and mortality; hence it is safe and effective and worth clinical promotion.
Collapse
Affiliation(s)
- Xiaohui Guo
- Xiaohui Guo, Department of Pediatrics, Binzhou People's Hospital, Shandong 256610, China
| | - Yanfeng Sun
- Yanfeng Sun, Department of Oncology, Binzhou People's Hospital, Shandong 256610, China
| | - Jing Miao
- Jing Miao, Department of Pediatrics, Binzhou People's Hospital, Shandong 256610, China
| | - Min Cui
- Min Cui, Department of Pediatrics, Binzhou People's Hospital, Shandong 256610, China
| | - Jiangbo Wang
- Jiangbo Wang, Department of Pediatrics, Binzhou People's Hospital, Shandong 256610, China
| | - Shuzhen Han
- Shuzhen Han, Department of Pediatrics, Binzhou People's Hospital, Shandong 256610, China
| |
Collapse
|
42
|
Hilgendorff A, Apitz C, Bonnet D, Hansmann G. Pulmonary hypertension associated with acute or chronic lung diseases in the preterm and term neonate and infant. The European Paediatric Pulmonary Vascular Disease Network, endorsed by ISHLT and DGPK. Heart 2016; 102 Suppl 2:ii49-56. [PMID: 27053698 DOI: 10.1136/heartjnl-2015-308591] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 10/14/2015] [Indexed: 11/04/2022] Open
Abstract
Persistent pulmonary hypertension of the newborn (PPHN) is the most common neonatal form and mostly reversible after a few days with improvement of the underlying pulmonary condition. When pulmonary hypertension (PH) persists despite adequate treatment, the severity of parenchymal lung disease should be assessed by chest CT. Pulmonary vein stenosis may need to be ruled out by cardiac catheterisation and lung biopsy, and genetic workup is necessary when alveolar capillary dysplasia is suspected. In PPHN, optimisation of the cardiopulmonary situation including surfactant therapy should aim for preductal SpO2between 91% and 95% and severe cases without post-tricuspid-unrestrictive shunt may receive prostaglandin E1 to maintain ductal patency in right heart failure. Inhaled nitric oxide is indicated in mechanically ventilated infants to reduce the need for extracorporal membrane oxygenation (ECMO), and sildenafil can be considered when this therapy is not available. ECMO may be indicated according to the ELSO guidelines. In older preterm infant, where PH is mainly associated with bronchopulmonary dysplasia (BPD) or in term infants with developmental lung anomalies such as congenital diaphragmatic hernia or cardiac anomalies, left ventricular diastolic dysfunction/left atrial hypertension or pulmonary vein stenosis, can add to the complexity of the disease. Here, oral or intravenous sildenafil should be considered for PH treatment in BPD, the latter for critically ill patients. Furthermore, prostanoids, mineralcorticoid receptor antagonists, and diuretics can be beneficial. Infants with proven or suspected PH should receive close follow-up, including preductal/postductal SpO2measurements, echocardiography and laboratory work-up including NT-proBNP, guided by clinical improvement or lack thereof.
Collapse
Affiliation(s)
- Anne Hilgendorff
- Department of Neonatology, Dr von Haunersche Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Christian Apitz
- Department of Paediatric Cardiology, Children's Hospital, University of Ulm, Ulm, Germany
| | - Damien Bonnet
- Unité Médico-Chirurgicale de Cardiologie Congénitale et Pédiatrique, Centre de référence Malformations Cardiaques Congénitales Complexes-M3C, Hôpital Necker Enfants Malades, APHP, Université Paris Descartes, Sorbonne Paris, Paris, France
| | - Georg Hansmann
- Department of Paediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| |
Collapse
|
43
|
Villalón H, Peñaloza G, Tuma D. TERAPIA REGENERATIVA EN NEONATOLOGÍA. REVISTA MÉDICA CLÍNICA LAS CONDES 2016. [DOI: 10.1016/j.rmclc.2016.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
44
|
Shahzad T, Radajewski S, Chao CM, Bellusci S, Ehrhardt H. Pathogenesis of bronchopulmonary dysplasia: when inflammation meets organ development. Mol Cell Pediatr 2016; 3:23. [PMID: 27357257 PMCID: PMC4927524 DOI: 10.1186/s40348-016-0051-9] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/20/2016] [Indexed: 01/12/2023] Open
Abstract
Bronchopulmonary dysplasia is a chronic lung disease of preterm infants. It is caused by the disturbance of physiologic lung development mainly in the saccular stage with lifelong restrictions of pulmonary function and an increased risk of abnormal somatic and psychomotor development. The contributors to this disease’s entity are multifactorial with pre- and postnatal origin. Central to the pathogenesis of bronchopulmonary is the induction of a massive pulmonary inflammatory response due to mechanical ventilation and oxygen toxicity. The extent of the pro-inflammatory reaction and the disturbance of further alveolar growth and vasculogenesis vary largely and can be modified by prenatal infections, antenatal steroids, and surfactant application. This minireview summarizes the important recent research findings on the pulmonary inflammatory reaction obtained in patient cohorts and in experimental models. Unfortunately, recent changes in clinical practice based on these findings had only limited impact on the incidence of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Tayyab Shahzad
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany.,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Sarah Radajewski
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany.,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Cho-Ming Chao
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany.,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Saverio Bellusci
- University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Harald Ehrhardt
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany. .,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany.
| |
Collapse
|
45
|
Akter F, Coghlan G, de Mel A. Nitric oxide in paediatric respiratory disorders: novel interventions to address associated vascular phenomena? Ther Adv Cardiovasc Dis 2016; 10:256-70. [PMID: 27215618 DOI: 10.1177/1753944716649893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) has a significant role in modulating the respiratory system and is being exploited therapeutically. Neonatal respiratory failure can affect around 2% of all live births and is responsible for over one third of all neonatal mortality. Current treatment method with inhaled NO (iNO) has demonstrated great benefits to patients with persistent pulmonary hypertension, bronchopulmonary dysplasia and neonatal respiratory distress syndrome. However, it is not without its drawbacks, which include the need for patients to be attached to mechanical ventilators. Notably, there is also a lack of identification of subgroups amongst abovementioned patients, and homogeneity in powered studies associated with iNO, which is one of the limitations. There are significant developments in drug delivery methods and there is a need to look at alternative or supplementary methods of NO delivery that could reduce current concerns. The addition of NO-independent activators and stimulators, or drugs such as prostaglandins to work in synergy with NO donors might be beneficial. It is of interest to consider such delivery methods within the respiratory system, where controlled release of NO can be introduced whilst minimizing the production of harmful byproducts. This article reviews current therapeutic application of iNO and the state-of-the-art technology methods for sustained delivery of NO that may be adapted and developed to address respiratory disorders. We envisage this perspective would prompt active investigation of such systems for their potential clinical benefit.
Collapse
Affiliation(s)
- Farhana Akter
- UCL Centre for Nanotechnology and Regenerative Medicine; Division of Surgery and Interventional Science, UCL, UK
| | - Gerry Coghlan
- Pulmonary Hypertension Unit, Royal Free London NHS Foundation Trust, UK
| | - Achala de Mel
- Lecturer in Regenerative Medicine, UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery and Interventional Science, University College London, Royal Free NHS Trust Hospital, 9th Floor, Room 355, Pond Street, London NW3 2QG, UK
| |
Collapse
|
46
|
Combined iNO and endothelial progenitor cells improve lung alveolar and vascular structure in neonatal rats exposed to prolonged hyperoxia. Pediatr Res 2015; 77:784-92. [PMID: 25742118 DOI: 10.1038/pr.2015.39] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 11/06/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Stem cells or inhaled nitric oxide (iNO) are reported to improve lung structures in bronchopulmonary dysplasia (BPD) models. We hypothesized that combined iNO and transplanted endothelial progenitor cells (EPCs) might restore lung structure in rats after neonatal hyperoxia. METHODS Litters were separated into eight groups: room air, hyperoxia, hyperoxia + iNO, hyperoxia + iNO + L-NAME, hyperoxia + EPCs, hyperoxia + EPCs + L-NAME, hyperoxia + EPCs + iNO, and hyperoxia + EPCs + iNO + L-NAME. Litters were exposed to hyperoxia from the 21st day, then, sacrificed. EPCs were injected on the 21st day. L-NAME was injected daily for 7 d from the 21st day. Serum vascular endothelial growth factor (VEGF), radial alveolar count (RAC), VIII factor, EPCs engraftment, lung VEGF, VEGFR2, endothelial nitric oxide (eNOS) and SDF-1 expression, and NO production were examined. RESULTS Hyperoxia exposure led to air space enlargement, loss of lung capillaries, and low expression of VEGF and eNOS. Transplanted EPCs, when combined with iNO, had significantly increased engraftment in lungs, compared to EPCs alone, upon hyperoxia exposure. There was improvement in alveolarization, microvessel density, and upregulation of VEGF and eNOS proteins in the hyperoxia-exposed EPCs with iNO group, compared to hyperoxia alone. CONCLUSION Combined EPCs and iNO improved lung structures after neonatal hyperoxia. This was associated with the upregulation of VEGF and eNOS expression.
Collapse
|
47
|
Ellsworth MA, Harris MN, Carey WA, Spitzer AR, Clark RH. Off-label use of inhaled nitric oxide after release of NIH consensus statement. Pediatrics 2015; 135:643-8. [PMID: 25755237 DOI: 10.1542/peds.2014-3290] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2015] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Inhaled nitric oxide (iNO) therapy is an off-label medication in infants <34 weeks' gestational age. In 2011, the National Institutes of Health released a statement discouraging routine iNO use in premature infants. The objective of this study was to describe utilization patterns of iNO in American NICUs in the years surrounding the release of the National Institutes of Health statement. We hypothesized that iNO prescription rates in premature infants have remained unchanged since 2011. METHODS The Pediatrix Medical Group Clinical Data Warehouse was queried for the years 2009-2013 to describe first exposure iNO use among all admitted neonates stratified by gestational age. RESULTS Between 2009 and 2013, the rate of iNO utilization in 23- to 29-week neonates increased from 5.03% to 6.19%, a relative increase of 23% (confidence interval: 8%-40%; P = .003). Of all neonates who received iNO therapy in 2013, nearly half were <34 weeks' gestation, with these infants accounting for more than half of all first exposure iNO days each year of the study period. CONCLUSIONS The rates of off-label iNO use in preterm infants continue to rise despite evidence revealing no clear benefit in this population. This pattern of iNO prescription is not benign and comes with economic consequences.
Collapse
Affiliation(s)
- Marc A Ellsworth
- Division of Neonatal Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Malinda N Harris
- Division of Neonatal Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - William A Carey
- Division of Neonatal Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Alan R Spitzer
- Center for Research, Education and Quality, Pediatrix Medical Group, Sunrise, Florida
| | - Reese H Clark
- Center for Research, Education and Quality, Pediatrix Medical Group, Sunrise, Florida
| |
Collapse
|
48
|
Iyengar A, Davis JM. Drug therapy for the prevention and treatment of bronchopulmonary dysplasia. Front Pharmacol 2015; 6:12. [PMID: 25762933 PMCID: PMC4329808 DOI: 10.3389/fphar.2015.00012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/13/2015] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION As more infants are surviving at younger gestational ages, bronchopulmonary dysplasia (BPD) remains as a frequent neonatal complication occurring after preterm birth. The multifactorial nature of the disease process makes BPD a challenging condition to treat. While multiple pharmacologic therapies have been investigated over the past two decades, there have been limited advances in the field. Often multiple therapies are used concurrently without clear evidence of efficacy, with potential for significant side effects from drug-drug interactions. METHODS Systematic literature review. CONCLUSION Although there is physiologic rationale for the use of many of these therapies, none of them has single-handedly altered the incidence, severity, or progression of BPD. Future research should focus on developing clinically significant end-points (short and long term respiratory assessments), investigating biomarkers that accurately predict risk and progression of disease, and creating appropriate stratification models of BPD severity. Applying a multi-modal approach to the study of new and existing drugs should be the most effective way of establishing the optimal prevention and treatment regimens for BPD.
Collapse
Affiliation(s)
- Anjali Iyengar
- Department of Pediatrics, Floating Hospital for Children at Tufts Medical CenterBoston, MA, USA
| | | |
Collapse
|
49
|
Jensen EA, Kirpalani H. Inhaled nitric oxide to prevent bronchopulmonary dysplasia in preterm infants--less than a silver bullet. J Pediatr 2014; 165:1079-81. [PMID: 25266344 DOI: 10.1016/j.jpeds.2014.08.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 08/20/2014] [Indexed: 11/16/2022]
Affiliation(s)
- Erik A Jensen
- Division of Neonatology Department of Pediatrics, The Children's Hospital of Philadelphia The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Haresh Kirpalani
- Division of Neonatology Department of Pediatrics, The Children's Hospital of Philadelphia The University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
50
|
Bhattacharya S, Zhou Z, Yee M, Chu CY, Lopez AM, Lunger VA, Solleti SK, Resseguie E, Buczynski B, Mariani TJ, O'Reilly MA. The genome-wide transcriptional response to neonatal hyperoxia identifies Ahr as a key regulator. Am J Physiol Lung Cell Mol Physiol 2014; 307:L516-23. [PMID: 25150061 DOI: 10.1152/ajplung.00200.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Premature infants requiring supplemental oxygen are at increased risk for developing bronchopulmonary dysplasia (BPD). Rodent models involving neonatal exposure to excessive oxygen concentrations (hyperoxia) have helped to identify mechanisms of BPD-associated pathology. Genome-wide assessments of the effects of hyperoxia in neonatal mouse lungs could identify novel BPD-related genes and pathways. Newborn C57BL/6 mice were exposed to 100% oxygen for 10 days, and whole lung tissue RNA was used for high-throughput, sequencing-based transcriptomic analysis (RNA-Seq). Significance Analysis of Microarrays and Ingenuity Pathway Analysis were used to identify genes and pathways affected. Expression patterns for selected genes were validated by qPCR. Mechanistic relationships between genes were further tested in cultured mouse lung epithelial cells. We identified 300 genes significantly and substantially affected following acute neonatal hyperoxia. Canonical pathways dysregulated in hyperoxia lungs included nuclear factor (erythryoid-derived-2)-like 2-mediated oxidative stress signaling, p53 signaling, eNOS signaling, and aryl hydrocarbon receptor (Ahr) pathways. Cluster analysis identified Ccnd1, Cdkn1a, and Ahr as critical regulatory nodes in the response to hyperoxia, with Ahr serving as the major effector node. A mechanistic role for Ahr was assessed in lung epithelial cells, and we confirmed its ability to regulate the expression of multiple hyperoxia markers, including Cdkn1a, Pdgfrb, and A2m. We conclude that a global assessment of gene regulation in the acute neonatal hyperoxia model of BPD-like pathology has identified Ahr as one driver of gene dysregulation.
Collapse
Affiliation(s)
- Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - Zhongyang Zhou
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Min Yee
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Perinatal and Pediatric Origins of Disease Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Chin-Yi Chu
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - Ashley M Lopez
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - Valerie A Lunger
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - Siva Kumar Solleti
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - Emily Resseguie
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Perinatal and Pediatric Origins of Disease Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Bradley Buczynski
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Perinatal and Pediatric Origins of Disease Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - Michael A O'Reilly
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Perinatal and Pediatric Origins of Disease Program, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|