1
|
Fraga MV, Dysart KC, Stoller JZ, Huber M, Fedec A, Mercer-Rosa L, Kirpalani H. Echocardiographic Assessment of Pulmonary Arterial Hypertension Following Inhaled Nitric Oxide in Infants with Severe Bronchopulmonary Dysplasia. Neonatology 2023; 120:633-641. [PMID: 37573771 DOI: 10.1159/000531586] [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: 02/06/2023] [Accepted: 06/08/2023] [Indexed: 08/15/2023]
Abstract
OBJECTIVES Inhaled nitric oxide (iNO) is an effective pulmonary vasodilator. However, the efficacy of iNO in former premature infants with established bronchopulmonary dysplasia (BPD) has not been studied. This study aimed to determine the efficacy of iNO in reducing pulmonary artery pressure in infants with severe BPD as measured by echocardiography. STUDY DESIGN Prospective, observational study enrolling infants born at less than 32 weeks gestation and in whom (1) iNO therapy was initiated after admission to our institution, or (2) at the outside institution less than 48 h before transfer and received an echocardiogram prior to iNO initiation, and (3) had severe BPD. Data were collected at three time-points: (1) before iNO; (2) 12-48 h after initiation of iNO; and (3) 48-168 h after initiation of iNO. The primary outcome was the effect of iNO on pulmonary artery pressure measured by echocardiography in patients with severe BPD between 48 and 168 h after initiating iNO therapy. RESULTS Of 37 enrolled, 81% had echocardiographic evidence of pulmonary arterial hypertension (PAH) before iNO and 56% after 48 h of iNO (p = 0.04). FiO2 requirements were significantly different between time-points (1) and (3) (p = 0.05). There were no significant differences between Tricuspid Annular Plane Systolic Excursion (TAPSE) Z-Scores, time to peak velocity: right ventricular ejection time (TPV:RVET), and ventilator changes. CONCLUSIONS Although we found a statistically significant reduction of PAH between time-point (1) and (3), future trials are needed to further guide clinical care.
Collapse
Affiliation(s)
- María V Fraga
- Division of Neonatology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kevin C Dysart
- Division of Neonatology, Department of Pediatrics, Nemours Children's Health, duPont Hospital for Children, Wilmington, Delaware, USA
| | - Jason Z Stoller
- Division of Neonatology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Matthew Huber
- Division of Neonatology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Anysia Fedec
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Laura Mercer-Rosa
- Division of Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Haresh Kirpalani
- Emeritus Professor of Pediatrics, Division of Neonatology, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Emeritus Professor of Pediatrics, Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
2
|
Mitra S, Altit G. L'utilisation du monoxyde d'azote inhalé chez les nouveau-nés. Paediatr Child Health 2023; 28:119-127. [PMID: 37151927 PMCID: PMC10156931 DOI: 10.1093/pch/pxac108] [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: 01/19/2022] [Accepted: 06/29/2022] [Indexed: 05/09/2023] Open
Abstract
Le monoxyde d'azote inhalé (NOi), un vasodilatateur pulmonaire sélectif, est utilisé pour le traitement des nouveau-nés en insuffisance respiratoire hypoxémique (IRH) associée à une hypertension pulmonaire persistante du nouveau-né. Idéalement, il doit commencer à être administré après la confirmation échocardiographique de ce type d'hypertension. L'utilisation de NOi est recommandée chez les nouveau-nés peu prématurés ou à terme chez qui survient une IRH malgré des stratégies d'oxygénation ou de ventilation optimales. Cependant, il n'est pas recommandé d'y recourir systématiquement chez les nouveau-nés prématurés sous assistance respiratoire. On peut l'envisager comme traitement de secours chez les nouveau-nés prématurés en IRH précoce associée à une rupture prolongée des membranes ou à un oligoamnios, ou en IRH tardive en cas d'hypertension pulmonaire liée à une dysplasie bronchopulmonaire et accompagnée d'une insuffisance ventriculaire droite marquée. On peut aussi l'envisager chez les nouveau-nés atteints d'une hernie diaphragmatique congénitale qui présentent une IRH persistante, malgré un recrutement pulmonaire optimal, des signes échocardiographiques d'hypertension pulmonaire suprasystémique et un fonctionnement ventriculaire gauche approprié.
Collapse
Affiliation(s)
- Souvik Mitra
- Société canadienne de pédiatrie, comité d'étude du fœtus et du nouveau-né, Ottawa (Ontario)Canada
| | - Gabriel Altit
- Société canadienne de pédiatrie, comité d'étude du fœtus et du nouveau-né, Ottawa (Ontario)Canada
| |
Collapse
|
3
|
Mitra S, Altit G. Inhaled nitric oxide use in newborns. Paediatr Child Health 2023; 28:119-127. [PMID: 37151928 PMCID: PMC10156933 DOI: 10.1093/pch/pxac107] [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: 01/19/2022] [Accepted: 06/29/2022] [Indexed: 05/09/2023] Open
Abstract
Inhaled nitric oxide (iNO), a selective pulmonary vasodilator, is used as a therapeutic modality in infants with hypoxemic respiratory failure (HRF) associated with persistent pulmonary hypertension of the newborn (PPHN). iNO should ideally be initiated following echocardiographic confirmation of PPHN. Use of iNO is recommended in late preterm and term infants who develop HRF despite optimal oxygenation and ventilation strategies. However, routine iNO use in preterm infants on respiratory support is not recommended. iNO may be considered as a rescue modality in preterm infants with early-onset HRF when associated with prolonged rupture of membranes or oligohydramnios, or late-onset HRF in the context of bronchopulmonary dysplasia-associated pulmonary hypertension (PH) with severe right ventricular failure. A trial of iNO may also be considered for infants with congenital diaphragmatic hernia with persistent HRF despite optimal lung recruitment, and with echocardiographic evidence of supra-systemic PH and adequate left ventricular function.
Collapse
Affiliation(s)
- Souvik Mitra
- Canadian Paediatric Society, Fetus and Newborn Committee, Ottawa, Ontario, Canada
| | - Gabriel Altit
- Canadian Paediatric Society, Fetus and Newborn Committee, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Fike CD, Aschner JL. Pharmacotherapy for Pulmonary Hypertension in Infants with Bronchopulmonary Dysplasia: Past, Present, and Future. Pharmaceuticals (Basel) 2023; 16:503. [PMID: 37111262 PMCID: PMC10141152 DOI: 10.3390/ph16040503] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
Approximately 8-42% of premature infants with chronic lung disease of prematurity, bronchopulmonary dysplasia (BPD), develop pulmonary hypertension (PH). Infants with BPD-PH carry alarmingly high mortality rates of up to 47%. Effective PH-targeted pharmacotherapies are desperately needed for these infants. Although many PH-targeted pharmacotherapies are commonly used to treat BPD-PH, all current use is off-label. Moreover, all current recommendations for the use of any PH-targeted therapy in infants with BPD-PH are based on expert opinion and consensus statements. Randomized Control Trials (RCTs) are needed to determine the efficacy of PH-targeted treatments in premature infants with or at risk of BPD-PH. Prior to performing efficacy RCTs, studies need to be conducted to obtain pharmacokinetic, pharmacodynamic, and safety data for any pharmacotherapy used in this understudied and fragile patient population. This review will discuss current and needed treatment strategies, identify knowledge deficits, and delineate both challenges to be overcome and approaches to be taken to develop effective PH-targeted pharmacotherapies that will improve outcomes for premature infants with or at risk of developing BPD-PH.
Collapse
Affiliation(s)
- Candice D. Fike
- Department of Pediatrics, University of Utah Health, Salt Lake City, UT 84108, USA
| | - Judy L. Aschner
- Department of Pediatrics, Joseph M. Sanzari Children’s Hospital at Hackensack University Medical Center, Hackensack, NJ 07601, USA
- Department of Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| |
Collapse
|
5
|
Fike CD, Avachat C, Birnbaum AK, Aschner JL, Sherwin CM. Pharmacokinetics of L-Citrulline in Neonates at Risk of Developing Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension. Paediatr Drugs 2023; 25:87-96. [PMID: 36316628 PMCID: PMC10039462 DOI: 10.1007/s40272-022-00542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Options to treat pulmonary hypertension (PH) in neonates with bronchopulmonary dysplasia (BPD) are few and largely ineffective. Improving the bioavailability of nitric oxide (NO) might be an efficacious treatment for BPD-PH. When administered orally, the NO-L-arginine precursor, L-citrulline, increases NO production in children and adults, however, pharmacokinetic (PK) studies of oral L-citrulline have not been performed in infants and children. OBJECTIVES This study characterized the PK of enterally administered L-citrulline in neonates at risk of developing BPD-PH to devise a model-informed dosing strategy. METHODS AND RESULTS Ten premature neonates (≤ 28 weeks gestation) were administered a single dose of 150 mg/kg (powder form solubilized in sterile water) oral L-citrulline at 32 ± 1 weeks postmenstrual age. Due to the need to limit blood draws, time windows were used to maximize the sampling over the dosing interval by assigning neonates to one of two groups (ii) samples collected pre-dose and at 1- and 2.5-h post-dose, and (ii) pre-dose and 0.25- and 3-h post-dose. The L-arginine concentrations (µmol/L) and the L-citrulline (µmol/L) plasma concentration-time data were evaluated using non-compartmental analysis (Phoenix WinNonlin version 8.1). Optimal dosage strategies were derived using a simulation-based methodology. Simulated doses of 51.5 mg or 37.5 mg/kg given four times a day produced steady-state concentrations close to a target of 50 µmol/L. The volume of distribution (V/F) and clearance (CL/F) were 302.89 ml and 774.96 ml/h, respectively, with the drug exhibiting a half-life of 16 minutes. The AUC from the time of dosing to the time of last concentration was 1473.3 h*μmol/L, with Cmax and Tmax of 799 μmol/L and 1.55 h, respectively. CONCLUSION This is the first PK study in neonates presenting data that can be used to inform dosing strategies in future randomized controlled trials evaluating enteral L-citrulline as a potential treatment to reduce PH associated with BPD in premature neonates. REGISTRATION Clinical trials.gov Identifier: NCT03542812.
Collapse
Affiliation(s)
- Candice D Fike
- Department of Pediatrics, The University of Utah Health, Salt Lake City, UT, USA
| | - Charul Avachat
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Angela K Birnbaum
- Department of Pediatrics, The University of Utah Health, Salt Lake City, UT, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Judy L Aschner
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - Catherine M Sherwin
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA.
- Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton, OH, USA.
- Dayton Children's Hospital, Dayton, OH, 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: 3.0] [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
|
Sakaria RP, Dhanireddy R. Pharmacotherapy in Bronchopulmonary Dysplasia: What Is the Evidence? Front Pediatr 2022; 10:820259. [PMID: 35356441 PMCID: PMC8959440 DOI: 10.3389/fped.2022.820259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
Bronchopulmonary Dysplasia (BPD) is a multifactorial disease affecting over 35% of extremely preterm infants born each year. Despite the advances made in understanding the pathogenesis of this disease over the last five decades, BPD remains one of the major causes of morbidity and mortality in this population, and the incidence of the disease increases with decreasing gestational age. As inflammation is one of the key drivers in the pathogenesis, it has been targeted by majority of pharmacological and non-pharmacological methods to prevent BPD. Most extremely premature infants receive a myriad of medications during their stay in the neonatal intensive care unit in an effort to prevent or manage BPD, with corticosteroids, caffeine, and diuretics being the most commonly used medications. However, there is no consensus regarding their use and benefits in this population. This review summarizes the available literature regarding these medications and aims to provide neonatologists and neonatal providers with evidence-based recommendations.
Collapse
Affiliation(s)
- Rishika P. Sakaria
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ramasubbareddy Dhanireddy
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
8
|
Malloy KW, Austin ED. Pulmonary hypertension in the child with bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:3546-3556. [PMID: 34324276 PMCID: PMC8530892 DOI: 10.1002/ppul.25602] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/25/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease of prematurity resulting from complex interactions of perinatal factors that often lead to prolonged respiratory support and increased pulmonary morbidity. There is also growing appreciation for the dysmorphic pulmonary bed characterized by vascular growth arrest and remodeling, resulting in pulmonary vascular disease and its most severe form, pulmonary hypertension (PH) in children with BPD. In this review, we comprehensively discuss the pathophysiology of PH in children with BPD, evaluate the current recommendations for screening and diagnosis of PH, discern associated comorbid conditions, and outline the current treatment options.
Collapse
Affiliation(s)
- Kelsey W Malloy
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eric D Austin
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Roberts K, Stepanovich G, Bhatt-Mehta V, Donn SM. New Pharmacologic Approaches to Bronchopulmonary Dysplasia. J Exp Pharmacol 2021; 13:377-396. [PMID: 33790663 PMCID: PMC8006962 DOI: 10.2147/jep.s262350] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary Dysplasia is the most common long-term respiratory morbidity of preterm infants, with the risk of development proportional to the degree of prematurity. While its pathophysiologic and histologic features have changed over time as neonatal demographics and respiratory therapies have evolved, it is now thought to be characterized by impaired distal lung growth and abnormal pulmonary microvascular development. Though the exact sequence of events leading to the development of BPD has not been fully elucidated and likely varies among patients, it is thought to result from inflammatory and mechanical/oxidative injury from chronic ventilatory support in fragile, premature lungs susceptible to injury from surfactant deficiency, structural abnormalities, inadequate antioxidant defenses, and a chest wall that is more compliant than the lung. In addition, non-pulmonary issues may adversely affect lung development, including systemic infections and insufficient nutrition. Once BPD has developed, its management focuses on providing adequate gas exchange while promoting optimal lung growth. Pharmacologic strategies to ameliorate or prevent BPD continue to be investigated. A variety of agents, to be reviewed henceforth, have been developed or re-purposed to target different points in the pathways that lead to BPD, including anti-inflammatories, diuretics, steroids, pulmonary vasodilators, antioxidants, and a number of molecules involved in the cell signaling cascade thought to be involved in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Katelyn Roberts
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gretchen Stepanovich
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Varsha Bhatt-Mehta
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- College of Pharmacy, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Steven M Donn
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Tracy MC, Cornfield DN. Bronchopulmonary Dysplasia: Then, Now, and Next. PEDIATRIC ALLERGY, IMMUNOLOGY, AND PULMONOLOGY 2020; 33:99-109. [PMID: 35922031 PMCID: PMC9354034 DOI: 10.1089/ped.2020.1205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/14/2020] [Indexed: 06/12/2023]
Abstract
Bronchopulmonary dysplasia (BPD) has evolved considerably since its first description over 50 years ago. This review aims to provide a historical framework for conceptualizing BPD and a current understanding of the changing definition, epidemiology, pathophysiology, treatment, and outcomes of BPD. The transdisciplinary approach that led to the initial phenotypic description of BPD continues to hold promise today. Investigators are refining the definition of BPD in light of changes in clinical care and increasing survival rates of very preterm infants. Despite improvements in perinatal care the incidence of BPD continues to increase. There is growing recognition that antenatal risk factors play a key role in the development of BPD. Strategies designed to prevent or limit neonatal lung injury continue to evolve. Defining the phenotype of infants with BPD can meaningfully direct treatment. Infants with BPD benefit from an interdisciplinary approach to longitudinal care with a focus on growth and neurocognitive development. While the ultimate impact of BPD on long-term pulmonary morbidity remains an active area of investigation, current data indicate that most children and adolescents with a history of BPD have a quality of life comparable to that of other preterm infants.
Collapse
Affiliation(s)
- Michael C. Tracy
- Center for Excellence in Pulmonary Biology, Division of Pediatric Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - David N. Cornfield
- Center for Excellence in Pulmonary Biology, Division of Pediatric Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
11
|
Baczynski M, Bell EF, Finan E, McNamara PJ, Jain A. Survey of practices in relation to chronic pulmonary hypertension in neonates in the Canadian Neonatal Network and the National Institute of Child Health and Human Development Neonatal Research Network. Pulm Circ 2020; 10:2045894020937126. [PMID: 32728420 PMCID: PMC7366415 DOI: 10.1177/2045894020937126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/03/2020] [Indexed: 11/17/2022] Open
Abstract
Current knowledge gaps pertaining to diagnosis and management of neonatal chronic
pulmonary hypertension (cPH) may result in significant variability in clinical practice.
The objective of the study is to understand cPH management practices in neonatal intensive
care units affiliated with the Canadian Neonatal Network (CNN) and National Institute of
Child Health and Human Development Neonatal Research Network (NRN). A 32-question survey
seeking practice details for cPH evaluation, diagnostic criteria, conservative measures,
pharmacotherapeutics, and follow-up was e-mailed to a designated physician at each center.
Responses were described as frequency (percentage) and compared between CNN and NRN, where
appropriate. Overall response rate was 67% (CNN 20/28 (71%), NRN 9/15 (60%)). While 8
(28%) centers had standardized management protocols, 17 (59%) routinely evaluate high-risk
patients; moderate-severe chronic lung disease being the commonest indication. While
interventricular septal flattening on echocardiography was the commonest listed diagnostic
criterion, several adjunctive indices were also identified. Asymptomatic neonates with cPH
were managed expectantly (routine care) in 50% of sites, and using various conservative
measures in others. Pulmonary vasodilators were prescribed for symptomatic cases, with 60%
of sites using them early (86% reporting any use). Seventy-five percent of sites use
inhaled nitric oxide and sildenafil citrate as first- and second-line agents,
respectively. Use of standard protocols, cardiac catheterization, and conservative
measures for asymptomatic cases was more common in NRN units
(p < 0.05). While there is relative homogeneity in patient
identification and diagnostic criteria used for neonatal cPH, significant interunit
inconsistencies still exists in routine evaluation, use of additional investigations,
management of asymptomatic cases, frequency and type of conservative measures, and choice
of pulmonary vasodilators.
Collapse
Affiliation(s)
| | - Edward F Bell
- Division of Neonatology, University of Iowa Stead Family Children's Hospital, Iowa City, IA, USA
| | - Emer Finan
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Canada.,Lunnenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Patrick J McNamara
- Division of Neonatology, University of Iowa Stead Family Children's Hospital, Iowa City, IA, USA.,Physiology, University of Toronto, Toronto, Canada
| | - Amish Jain
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Canada.,Lunnenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,Department of Paediatrics, University of Toronto, Toronto, Canada
| |
Collapse
|
12
|
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: 1.3] [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
|
13
|
El-Saie A, Shivanna B. Novel Strategies to Reduce Pulmonary Hypertension in Infants With Bronchopulmonary Dysplasia. Front Pediatr 2020; 8:201. [PMID: 32457857 PMCID: PMC7225259 DOI: 10.3389/fped.2020.00201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/02/2020] [Indexed: 01/10/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a developmental lung disorder of preterm infants primarily caused by the failure of host defense mechanisms to prevent tissue injury and facilitate repair. This disorder is the most common complication of premature birth, and its incidence remains unchanged over the past few decades. Additionally, BPD increases long-term cardiopulmonary and neurodevelopmental morbidities of preterm infants. Pulmonary hypertension (PH) is a common morbidity of BPD. Importantly, the presence of PH increases both the short- and long-term morbidities and mortality in BPD infants. Further, there are no curative therapies for this complex disease. Besides providing an overview of the pathogenesis and diagnosis of PH associated with BPD, we have attempted to comprehensively review and summarize the current literature on the interventions to prevent and/or mitigate BPD and PH in preclinical studies. Our goal was to provide insight into the therapies that have a high translational potential to meaningfully manage BPD patients with PH.
Collapse
Affiliation(s)
- Ahmed El-Saie
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Binoy Shivanna
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
14
|
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: 16] [Impact Index Per Article: 3.2] [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
|
15
|
Abstract
Introduction: Bronchopulmonary dysplasia (BPD) is a common long-term adverse complication of very premature delivery. Affected infants can suffer chronic respiratory morbidities including lung function abnormalities and reduced exercise capacity even as young adults. Many studies have investigated possible preventative strategies; however, it is equally important to identify optimum management strategies for infants with evolving or established BPD. Areas covered: Respiratory support modalities and established and novel pharmacological treatments. Expert opinion: Respiratory support modalities including proportional assist ventilation and neurally adjusted ventilatory assist are associated with short term improvements in oxygenation indices. Such modalities need to be investigated in appropriate RCTs. Many pharmacological treatments are routinely used with a limited evidence base, for example diuretics. Stem cell therapies in small case series are associated with promising results. More research is required before it is possible to determine if such therapies should be investigated in large RCTs with long-term outcomes.
Collapse
Affiliation(s)
- Emma Williams
- a Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London , UK.,b The Asthma UK Centre for Allergic Mechanisms in Asthma, King's College London , UK
| | - Anne Greenough
- a Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London , UK.,c NIHR Biomedical Research Centre based at Guy's and St Thomas' NHS Foundation Trust and King's College London , London , UK
| |
Collapse
|
16
|
Jankov RP, Daniel KL, Iny S, Kantores C, Ivanovska J, Ben Fadel N, Jain A. Sodium nitrite augments lung S-nitrosylation and reverses chronic hypoxic pulmonary hypertension in juvenile rats. Am J Physiol Lung Cell Mol Physiol 2018; 315:L742-L751. [DOI: 10.1152/ajplung.00184.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Deficient nitric oxide (NO) signaling plays a critical role in the pathogenesis of chronic neonatal pulmonary hypertension (PHT). Physiological NO signaling is regulated by S-nitrosothiols (SNOs), which act both as a reservoir for NO and as a reversible modulator of protein function. We have previously reported that therapy with inhaled NO (iNO) increased peroxynitrite-mediated nitration in the juvenile rat lung, although having minimal reversing effects on vascular remodeling. We hypothesized that sodium nitrite (NaNO2) would be superior to iNO in enhancing lung SNOs, thereby contributing to reversal of chronic hypoxic PHT. Rat pups were exposed to air or hypoxia (13% O2) from postnatal days 1 to 21. Dose-response prevention studies were conducted from days 1–21 to determine the optimal dose of NaNO2. Animals then received rescue therapy with daily subcutaneous NaNO2 (20 mg/kg), vehicle, or were continuously exposed to iNO (20 ppm) from days 14–21. Chronic PHT secondary to hypoxia was both prevented and reversed by treatment with NaNO2. Rescue NaNO2 increased lung NO and SNO contents to a greater extent than iNO, without causing nitration. Seven lung SNO proteins upregulated by treatment with NaNO2 were identified by multiplex tandem mass tag spectrometry, one of which was leukotriene A4 hydrolase (LTA4H). Rescue therapy with a LTA4H inhibitor, SC57461A (10 mg·kg−1·day−1 sc), partially reversed chronic hypoxic PHT. We conclude that NaNO2 was superior to iNO in increasing tissue NO and SNO generation and reversing chronic PHT, in part via upregulated SNO-LTA4H.
Collapse
Affiliation(s)
- Robert P. Jankov
- Molecular Biomedicine Program, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Kathrine L. Daniel
- Molecular Biomedicine Program, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Shira Iny
- Molecular Biomedicine Program, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Crystal Kantores
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Julijana Ivanovska
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Nadya Ben Fadel
- Faculty of Medicine, Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Amish Jain
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
17
|
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: 33] [Impact Index Per Article: 4.7] [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
|
18
|
|
19
|
Abstract
PURPOSE OF REVIEW Bronchopulmonary dysplasia (BPD) or chronic lung disease of infancy BPD was originally described 50 years ago, in 1967 by Northway et al. This article possesses two fundamental objectives to provide: a brief historical perspective on BPD; and an update relative to current notions of epidemiology, pathophysiology, evaluation, and clinical management of BPD complicated by vascular disease. The review highlights areas of consensus and ongoing uncertainty. RECENT FINDINGS The clinical cause and presentation of infants with BPD has evolved over the past several decades. Considerable improvements in neonatal care, including surfactant replacement therapies, antenatal steroids, nutritional support, ventilator management, and attention to the potential of oxygen toxicity, underlie the evolution of BPD. Most children with BPD improve over time. However, in the presence of vascular disease, the morbidity and mortality associated with BPD increases considerably. Though recent recommendations include procuring an echocardiogram to screen for pulmonary hypertension in infants with established BPD, there is less agreement surrounding the additional diagnostic and putative treatment modalities for infants with BPD and pulmonary hypertension. The indications, rationale, potential benefits, and risks of vasodilator therapy in BPD are discussed. SUMMARY The pediatric community has 50 years of experience with BPD. Past experience should be used to inform present and future diagnostic and treatment strategies. This review seeks to arm the clinician with evidence that motivates a physiology-based approach to the management of infants with BPD and pulmonary hypertension.
Collapse
|
20
|
|
21
|
Dikalova A, Aschner JL, Kaplowitz MR, Summar M, Fike CD. Tetrahydrobiopterin oral therapy recouples eNOS and ameliorates chronic hypoxia-induced pulmonary hypertension in newborn pigs. Am J Physiol Lung Cell Mol Physiol 2016; 311:L743-L753. [PMID: 27542807 PMCID: PMC5142125 DOI: 10.1152/ajplung.00238.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/14/2016] [Indexed: 01/21/2023] Open
Abstract
We previously showed that newborn piglets who develop pulmonary hypertension during exposure to chronic hypoxia have diminished pulmonary vascular nitric oxide (NO) production and evidence of endothelial NO synthase (eNOS) uncoupling (Fike CD, Dikalova A, Kaplowitz MR, Cunningham G, Summar M, Aschner JL. Am J Respir Cell Mol Biol 53: 255-264, 2015). Tetrahydrobiopterin (BH4) is a cofactor that promotes eNOS coupling. Current clinical strategies typically invoke initiating treatment after the diagnosis of pulmonary hypertension, rather than prophylactically. The major purpose of this study was to determine whether starting treatment with an oral BH4 compound, sapropterin dihydrochloride (sapropterin), after the onset of pulmonary hypertension would recouple eNOS in the pulmonary vasculature and ameliorate disease progression in chronically hypoxic piglets. Normoxic (control) and hypoxic piglets were studied. Some hypoxic piglets received oral sapropterin starting on day 3 of hypoxia and continued throughout an additional 7 days of hypoxic exposure. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess eNOS dimer-to-monomer ratios (a measure of eNOS coupling), NO production, and superoxide (O2·-) generation. Although higher than in normoxic controls, pulmonary vascular resistance was lower in sapropterin-treated hypoxic piglets than in untreated hypoxic piglets. Consistent with eNOS recoupling, eNOS dimer-to-monomer ratios and NO production were greater and O2·- generation was less in pulmonary arteries from sapropterin-treated than untreated hypoxic animals. When started after disease onset, oral sapropterin treatment inhibits chronic hypoxia-induced pulmonary hypertension at least in part by recoupling eNOS in the pulmonary vasculature of newborn piglets. Rescue treatment with sapropterin may be an effective strategy to inhibit further development of pulmonary hypertension in newborn infants suffering from chronic cardiopulmonary conditions associated with episodes of prolonged hypoxia.
Collapse
Affiliation(s)
- Anna Dikalova
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Judy L Aschner
- Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, New York, New York
| | - Mark R Kaplowitz
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pediatrics, the University of Utah School of Medicine, Salt Lake City, Utah; and
| | - Marshall Summar
- Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia
| | - Candice D Fike
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pediatrics, the University of Utah School of Medicine, Salt Lake City, Utah; and
| |
Collapse
|
22
|
Abstract
Bronchopulmonary dysplasia is the most common morbidity among surviving premature infants. Injury to the developing lung is the result of the interaction between a susceptible host and a number of contributing factors such as mechanical ventilation and infection. The resulting persistent impairment of pulmonary function and need for ongoing therapy are the underlying characteristics of bronchopulmonary dysplasia. Important insights into the pathogenesis of bronchopulmonary dysplasia have led to numerous therapies and preventive approaches. Although significant progress has been made, in order to further affect the incidence and severity of the disease, we need to further study (a) the genetically determined predisposing factors, (b) the relative contribution of the various pathogenetic pathways, and, most important, (c) how to best translate the knowledge gained from these studies into effective clinical approaches.
Collapse
Affiliation(s)
- Helen Christou
- Division of Newborn Medicine, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
23
|
Abman SH, Grenolds A, Mourani P. Pulmonary Vascular Disease in Bronchopulmonary Dysplasia. ACTA ACUST UNITED AC 2016. [DOI: 10.21693/1933-088x-15.2.92] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Pulmonary vascular disease and pulmonary hypertension (PH) contributes significantly to the pathogenesis, pathophysiology, and clinical course of infants with bronchopulmonary dysplasia (BPD). This article briefly reviews the impact of premature birth on the developing lung circulation, mechanisms that contribute to the development of PH in premature newborns, and the diagnostic evaluation and management of severe PH in infants with BPD.
Collapse
Affiliation(s)
- Steven H. Abman
- Pediatric Heart Lung Center, Section of Pediatric Pulmonary Medicine, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| | - Alicia Grenolds
- Pediatric Heart Lung Center, Section of Pediatric Pulmonary Medicine, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| | - Peter Mourani
- Pediatric Heart Lung Center, Section of Critical Care Medicine, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| |
Collapse
|
24
|
Abman SH, Hansmann G, Archer SL, Ivy DD, Adatia I, Chung WK, Hanna BD, Rosenzweig EB, Raj JU, Cornfield D, Stenmark KR, Steinhorn R, Thébaud B, Fineman JR, Kuehne T, Feinstein JA, Friedberg MK, Earing M, Barst RJ, Keller RL, Kinsella JP, Mullen M, Deterding R, Kulik T, Mallory G, Humpl T, Wessel DL. Pediatric Pulmonary Hypertension: Guidelines From the American Heart Association and American Thoracic Society. Circulation 2015; 132:2037-99. [PMID: 26534956 DOI: 10.1161/cir.0000000000000329] [Citation(s) in RCA: 706] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension is associated with diverse cardiac, pulmonary, and systemic diseases in neonates, infants, and older children and contributes to significant morbidity and mortality. However, current approaches to caring for pediatric patients with pulmonary hypertension have been limited by the lack of consensus guidelines from experts in the field. In a joint effort from the American Heart Association and American Thoracic Society, a panel of experienced clinicians and clinician-scientists was assembled to review the current literature and to make recommendations on the diagnosis, evaluation, and treatment of pediatric pulmonary hypertension. This publication presents the results of extensive literature reviews, discussions, and formal scoring of recommendations for the care of children with pulmonary hypertension.
Collapse
MESH Headings
- Cardiovascular Agents/therapeutic use
- Child
- Child, Preschool
- Combined Modality Therapy
- Diagnostic Imaging/methods
- Disease Management
- Extracorporeal Membrane Oxygenation
- Genetic Counseling
- Heart Defects, Congenital/complications
- Heart Defects, Congenital/therapy
- Hernias, Diaphragmatic, Congenital/complications
- Hernias, Diaphragmatic, Congenital/therapy
- Humans
- Hypertension, Pulmonary/diagnosis
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/therapy
- Infant
- Infant, Newborn
- Lung/embryology
- Lung Transplantation
- Nitric Oxide/administration & dosage
- Nitric Oxide/therapeutic use
- Oxygen Inhalation Therapy
- Persistent Fetal Circulation Syndrome/diagnosis
- Persistent Fetal Circulation Syndrome/therapy
- Postoperative Complications/therapy
- Respiration, Artificial/adverse effects
- Respiration, Artificial/methods
- Ventilator-Induced Lung Injury/prevention & control
Collapse
|
25
|
Fike CD, Dikalova A, Kaplowitz MR, Cunningham G, Summar M, Aschner JL. Rescue Treatment with L-Citrulline Inhibits Hypoxia-Induced Pulmonary Hypertension in Newborn Pigs. Am J Respir Cell Mol Biol 2015; 53:255-64. [PMID: 25536367 DOI: 10.1165/rcmb.2014-0351oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Infants with cardiopulmonary disorders associated with hypoxia develop pulmonary hypertension. We previously showed that initiation of oral L-citrulline before and continued throughout hypoxic exposure improves nitric oxide (NO) production and ameliorates pulmonary hypertension in newborn piglets. Rescue treatments, initiated after the onset of pulmonary hypertension, better approximate clinical strategies. Mechanisms by which L-citrulline improves NO production merit elucidation. The objective of this study was to determine whether starting L-citrulline after the onset of pulmonary hypertension inhibits disease progression and improves NO production by recoupling endothelial NO synthase (eNOS). Hypoxic and normoxic (control) piglets were studied. Some hypoxic piglets received oral L-citrulline starting on Day 3 of hypoxia and continuing throughout the remaining 7 days of hypoxic exposure. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess NO production and eNOS dimer-to-monomer ratios (a measure of eNOS coupling). Pulmonary vascular resistance was lower in L-citrulline-treated hypoxic piglets than in untreated hypoxic piglets but was higher than in normoxic controls. NO production and eNOS dimer-to-monomer ratios were greater in pulmonary arteries from L-citrulline-treated than from untreated hypoxic animals but were lower than in normoxic controls. When started after disease onset, oral L-citrulline treatment improves NO production by recoupling eNOS and inhibits the further development of chronic hypoxia-induced pulmonary hypertension in newborn piglets. Oral L-citrulline may be a novel strategy to halt or reverse pulmonary hypertension in infants suffering from cardiopulmonary conditions associated with hypoxia.
Collapse
Affiliation(s)
- Candice D Fike
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.,2 Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Anna Dikalova
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark R Kaplowitz
- 1 Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gary Cunningham
- 3 Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia; and
| | - Marshall Summar
- 3 Division of Genetics and Metabolism, Children's National Medical Center, Washington, District of Columbia; and
| | - Judy L Aschner
- 4 Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, New York, New York
| |
Collapse
|
26
|
Kumarathasan P, Blais E, Saravanamuthu A, Bielecki A, Mukherjee B, Bjarnason S, Guénette J, Goegan P, Vincent R. Nitrative stress, oxidative stress and plasma endothelin levels after inhalation of particulate matter and ozone. Part Fibre Toxicol 2015. [PMID: 26376633 DOI: 10.1186/s12989‐015‐0103‐7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND While exposure to ambient air contaminants is clearly associated with adverse health outcomes, disentangling mechanisms of pollutant interactions remains a challenge. OBJECTIVES We aimed at characterizing free radical pathways and the endothelinergic system in rats after inhalation of urban particulate matter, ozone, and a combination of particles plus ozone to gain insight into pollutant-specific toxicity mechanisms and any effect modification due to air pollutant mixtures. METHODS Fischer 344 rats were exposed for 4 h to a 3 × 3 concentration matrix of ozone (0, 0.4, 0.8 ppm) and EHC-93 particles (0, 5, 50 mg/m(3)). Bronchoalveolar lavage fluid (BALF), BAL cells, blood and plasma were analysed for biomarkers of effects immediately and 24 h post-exposure. RESULTS Inhalation of ozone increased (p < 0.05) lipid oxidation products in BAL cells immediately post-exposure, and increased (p < 0.05) total protein, neutrophils and mature macrophages in the BALF 24 h post-exposure. Ozone increased (p < 0.05) the formation of reactive oxygen species (ROS), assessed by m-, p-, o-tyrosines in BALF (Ozone main effects, p < 0.05), while formation of reactive nitrogen species (RNS), indicated by 3-nitrotyrosine, correlated with dose of urban particles (EHC-93 main effects or EHC-93 × Ozone interactions, p < 0.05). Carboxyhemoglobin levels in blood exhibited particle exposure-related increase (p < 0.05) 24 h post recovery. Plasma 3-nitrotyrosine and o-tyrosine were increased (p < 0.05) after inhalation of particles; the effect on 3-nitrotyrosine was abrogated after exposure to ozone plus particles (EHC-93 × Ozone, p < 0.05). Big endothelin-1 (BET-1) and ET-1 were increased in plasma after inhalation of particles or ozone alone, but the effects appeared to be attenuated by co-exposure to contaminants (EHC-93 × Ozone, p < 0.05). Plasma ET levels were positively correlated (p < 0.05) with BALF m- and o-tyrosine levels. CONCLUSIONS Pollutant-specific changes can be amplified or abrogated following multi-pollutant exposures. Oxidative and nitrative stress in the lung compartment may contribute to secondary extra-pulmonary ROS/RNS formation. Nitrative stress and endothelinergic imbalance emerge as potential key pathways of air pollutant health effects, notably of ambient particulate matter.
Collapse
Affiliation(s)
- Prem Kumarathasan
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada.
| | - Erica Blais
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Anushuyadevi Saravanamuthu
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Agnieszka Bielecki
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Ballari Mukherjee
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Stephen Bjarnason
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| | - Josée Guénette
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| | - Patrick Goegan
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| | - Renaud Vincent
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| |
Collapse
|
27
|
Kumarathasan P, Blais E, Saravanamuthu A, Bielecki A, Mukherjee B, Bjarnason S, Guénette J, Goegan P, Vincent R. Nitrative stress, oxidative stress and plasma endothelin levels after inhalation of particulate matter and ozone. Part Fibre Toxicol 2015; 12:28. [PMID: 26376633 PMCID: PMC4573945 DOI: 10.1186/s12989-015-0103-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/02/2015] [Indexed: 01/08/2023] Open
Abstract
Background While exposure to ambient air contaminants is clearly associated with adverse health outcomes, disentangling mechanisms of pollutant interactions remains a challenge. Objectives We aimed at characterizing free radical pathways and the endothelinergic system in rats after inhalation of urban particulate matter, ozone, and a combination of particles plus ozone to gain insight into pollutant-specific toxicity mechanisms and any effect modification due to air pollutant mixtures. Methods Fischer 344 rats were exposed for 4 h to a 3 × 3 concentration matrix of ozone (0, 0.4, 0.8 ppm) and EHC-93 particles (0, 5, 50 mg/m3). Bronchoalveolar lavage fluid (BALF), BAL cells, blood and plasma were analysed for biomarkers of effects immediately and 24 h post-exposure. Results Inhalation of ozone increased (p < 0.05) lipid oxidation products in BAL cells immediately post-exposure, and increased (p < 0.05) total protein, neutrophils and mature macrophages in the BALF 24 h post-exposure. Ozone increased (p < 0.05) the formation of reactive oxygen species (ROS), assessed by m-, p-, o-tyrosines in BALF (Ozone main effects, p < 0.05), while formation of reactive nitrogen species (RNS), indicated by 3-nitrotyrosine, correlated with dose of urban particles (EHC-93 main effects or EHC-93 × Ozone interactions, p < 0.05). Carboxyhemoglobin levels in blood exhibited particle exposure-related increase (p < 0.05) 24 h post recovery. Plasma 3-nitrotyrosine and o-tyrosine were increased (p < 0.05) after inhalation of particles; the effect on 3-nitrotyrosine was abrogated after exposure to ozone plus particles (EHC-93 × Ozone, p < 0.05). Big endothelin-1 (BET-1) and ET-1 were increased in plasma after inhalation of particles or ozone alone, but the effects appeared to be attenuated by co-exposure to contaminants (EHC-93 × Ozone, p < 0.05). Plasma ET levels were positively correlated (p < 0.05) with BALF m- and o-tyrosine levels. Conclusions Pollutant-specific changes can be amplified or abrogated following multi-pollutant exposures. Oxidative and nitrative stress in the lung compartment may contribute to secondary extra-pulmonary ROS/RNS formation. Nitrative stress and endothelinergic imbalance emerge as potential key pathways of air pollutant health effects, notably of ambient particulate matter. Electronic supplementary material The online version of this article (doi:10.1186/s12989-015-0103-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Prem Kumarathasan
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada.
| | - Erica Blais
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Anushuyadevi Saravanamuthu
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Agnieszka Bielecki
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Ballari Mukherjee
- Analytical Biochemistry and Proteomics Laboratory, Environmental Health Centre, Room 233A, 0803C Tunney's Pasture, Ottawa, K1A 0 K9, ON, Canada
| | - Stephen Bjarnason
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| | - Josée Guénette
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| | - Patrick Goegan
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| | - Renaud Vincent
- Inhalation Toxicology Laboratory, Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0 K9, ON, Canada
| |
Collapse
|
28
|
Ballard PL, Keller RL, Black DM, Durand DJ, Merrill JD, Eichenwald EC, Truog WE, Mammel MC, Steinhorn R, Ryan RM, Courtney SE, Horneman H, Ballard RA. Inhaled nitric oxide increases urinary nitric oxide metabolites and cyclic guanosine monophosphate in premature infants: relationship to pulmonary outcome. Am J Perinatol 2015; 32:225-32. [PMID: 24968129 PMCID: PMC5032843 DOI: 10.1055/s-0034-1382255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Inhaled nitric oxide (iNO) has been tested to prevent bronchopulmonary dysplasia (BPD) in premature infants, however, the role of cyclic guanosine monophosphate (cGMP) is not known. We hypothesized that levels of NO metabolites (NOx) and cGMP in urine, as a noninvasive source for biospecimen collection, would reflect the dose of iNO and relate to pulmonary outcome. STUDY DESIGN Studies were performed on 125 infants who required mechanical ventilation at 7 to 14 days and received 24 days of iNO at 20-2 ppm. A control group of 19 infants did not receive iNO. RESULTS In NO-treated infants there was a dose-dependent increase of both NOx and cGMP per creatinine (maximal 3.1- and 2-fold, respectively, at 10-20 ppm iNO) compared with off iNO. NOx and cGMP concentrations at both 2 ppm and off iNO were inversely related to severity of lung disease during the 1st month, and the NOx levels were lower in infants who died or developed BPD at term. NOx was higher in Caucasian compared with other infants at all iNO doses. CONCLUSION Urinary NOx and cGMP are biomarkers of endogenous NO production and lung uptake of iNO, and some levels reflect the severity of lung disease. These results support a role of the NO-cGMP pathway in lung development.
Collapse
Affiliation(s)
- Philip L. Ballard
- Department of Pediatrics, University of California, San Francisco, California
| | - Roberta L. Keller
- Department of Pediatrics, University of California, San Francisco, California
| | - Dennis M. Black
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - David J. Durand
- Division of Neonatology, Children’s Hospital and Research Center Oakland, Oakland, California
| | - Jeffrey D. Merrill
- Division of Neonatology, Children’s Hospital and Research Center Oakland, Oakland, California
| | - Eric C. Eichenwald
- Department of Pediatrics, University of Texas Medical School, Houston, Texas
| | - William E. Truog
- Department of Pediatrics, Children’s Mercy Hospitals and Clinics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Mark C. Mammel
- Department of Pediatrics, Children’s Hospital and Clinics of Minnesota, St. Paul, Minnesota
| | - Robin Steinhorn
- Department of Pediatrics, University of California Davis Children’s Hospital, Sacramento, California
| | - Rita M. Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Sherry E. Courtney
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Hart Horneman
- Department of Pediatrics, University of California, San Francisco, California
| | - Roberta A. Ballard
- Department of Pediatrics, University of California, San Francisco, California
| | | |
Collapse
|
29
|
Rossor T, Greenough A. Advances in paediatric pulmonary vascular disease associated with bronchopulmonary dysplasia. Expert Rev Respir Med 2014; 9:35-43. [PMID: 25426585 DOI: 10.1586/17476348.2015.986470] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pulmonary hypertension (PH) is a common finding in infants with bronchopulmonary dysplasia (BPD). The aim of this review is to describe recent advances in the diagnosis and treatment of PH and discuss whether they will benefit infants and children with BPD related PH. Echocardiography remains the mainstay of diagnosis but has limitations, further developments in diagnostic techniques and identification of biomarkers are required. There are many potential therapies for PH associated with BPD. Inhaled nitric oxide has been shown to improve short term outcomes only. Sidenafil in resource limited settings was shown in three randomized trials to significantly reduce mortality. The efficacy of other therapies including prostacyclin, PDE3 inhibitors and endothelin receptor blockers has only been reported in case reports or case series. Randomized controlled trials with long term follow up are required to appropriately assess the efficacy of therapies aimed at improving the outcome of children with PH.
Collapse
Affiliation(s)
- Thomas Rossor
- Division of Asthma, Allergy and Lung Biology, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, England, UK
| | | |
Collapse
|
30
|
Keszler M. Guidelines for Rational and Cost-Effective Use of iNO Therapy in Term and Preterm Infants. J Clin Neonatol 2014; 1:59-63. [PMID: 24027689 PMCID: PMC3743146 DOI: 10.4103/2249-4847.96739] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Inhaled nitric oxide (iNO) is an effective but costly therapy for infants with hypoxemic respiratory failure. The approved and solidly evidence-based indication is for treatment of late preterm and term infants with persistent pulmonary hypertension of the newborn (PPHN); however, off-label use of iNO has become widespread. Although iNO treatment of infants with congenital diaphragmatic hernia constitutes one of the approved indications, available evidence from randomized trials suggests marginal if any efficacy. Rescue therapy in preterm infants with severe respiratory failure has been studied extensively and is not supported by data from a number of controlled trials. Such use is widespread, but should be discouraged. There may be a subgroup of such infants with pulmonary hypoplasia and documented PPHN who may benefit from this treatment, but the data are limited. Several studies have examined the use of iNO for prevention of chronic lung disease with inconsistent results. This promising application requires more study before it can be recommended. There may be a role of iNO in treating infants with pulmonary hypertension complicating severe bronchopulmonary dysplasia, but there are limited data on long term outcomes. Alternate therapies such as sildenafil may be beneficial in this specific population as well as in other causes of pulmonary hypertension. Rational use of this expensive treatment will maximize cost:benefit and avoid potential exposure to unknown adverse effects not balanced by documentable benefits.
Collapse
Affiliation(s)
- Martin Keszler
- Department of Pediatrics, Women and Infants Hospital, Providence, RI USA
| |
Collapse
|
31
|
Madurga A, Mižíková I, Ruiz-Camp J, Vadász I, Herold S, Mayer K, Fehrenbach H, Seeger W, Morty RE. Systemic hydrogen sulfide administration partially restores normal alveolarization in an experimental animal model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2014; 306:L684-97. [PMID: 24508731 DOI: 10.1152/ajplung.00361.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Arrested alveolarization is the pathological hallmark of bronchopulmonary dysplasia (BPD), a complication of premature birth. Here, the impact of systemic application of hydrogen sulfide (H2S) on postnatal alveolarization was assessed in a mouse BPD model. Exposure of newborn mice to 85% O2 for 10 days reduced the total lung alveoli number by 56% and increased alveolar septal wall thickness by 29%, as assessed by state-of-the-art stereological analysis. Systemic application of H2S via the slow-release H2S donor GYY4137 for 10 days resulted in pronounced improvement in lung alveolarization in pups breathing 85% O2, compared with vehicle-treated littermates. Although without impact on lung oxidative status, systemic H2S blunted leukocyte infiltration into alveolar air spaces provoked by hyperoxia, and restored normal lung interleukin 10 levels that were otherwise depressed by 85% O2. Treatment of primary mouse alveolar type II (ATII) cells with the rapid-release H2S donor NaHS had no impact on cell viability; however, NaHS promoted ATII cell migration. Although exposure of ATII cells to 85% O2 caused dramatic changes in mRNA expression, exposure to either GYY4137 or NaHS had no impact on ATII cell mRNA expression, as assessed by microarray, suggesting that the effects observed were independent of changes in gene expression. The impact of NaHS on ATII cell migration was attenuated by glibenclamide, implicating ion channels, and was accompanied by activation of Akt, hinting at two possible mechanisms of H2S action. These data support further investigation of H2S as a candidate interventional strategy to limit the arrested alveolarization associated with BPD.
Collapse
Affiliation(s)
- Alicia Madurga
- Dept. of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, D-61231 Bad Nauheim, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease in infants and children that is associated with significant morbidity and mortality. The disease is characterized by progressive pulmonary vascular functional and structural changes resulting in increased pulmonary vascular resistance and eventual right heart failure and death. In the majority of pediatric patients, PAH is idiopathic or associated with congenital heart disease and rarely is associated with other conditions such as connective tissue or thromboembolic disease. Although treatment of the underlying disease and reversal of advanced structural changes has not yet been achieved with current therapy, quality of life and survival have been improved significantly. Targeted pulmonary vasodilator therapies, including endothelin receptor antagonists, prostacyclin analogs, and phosphodiesterase type 5 inhibitors, have demonstrated hemodynamic and functional improvement in children. The management of pediatric PAH remains challenging, as treatment decisions continue to depend largely on results from evidence-based adult studies and the clinical experience of pediatric experts. This article reviews the current drug therapies and their use in the management of PAH in children.
Collapse
Affiliation(s)
- Erika E Vorhies
- Division of Pediatric Cardiology, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, C.S. Mott Children's Hospital, Ann Arbor, MI, USA
| | | |
Collapse
|
33
|
Fike CD, Aschner JL. Looking beyond PPHN: the unmet challenge of chronic progressive pulmonary hypertension in the newborn. Pulm Circ 2013; 3:454-66. [PMID: 24618533 DOI: 10.1086/674438] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Abstract Infants with forms of pulmonary hypertension (PH) that persist or develop beyond the first week of life are an understudied group of patients with up to 40%-60% mortality. The clinical management of the progressive PH that develops in these infants is challenging because of the nonspecific signs and symptoms of clinical presentation, the limited diagnostic sensitivity of standard echocardiographic techniques, and the lack of proven therapies. The signaling mechanisms that underlie the structural and functional abnormalities in the pulmonary circulation of these infants are not yet clear. The ability to improve outcomes for these patients awaits technological advances to improve diagnostic capabilities and therapeutic discoveries made in basic science laboratories that can be tested in randomized clinical trials.
Collapse
Affiliation(s)
- Candice D Fike
- 1 Department of Pediatrics, Vanderbilt University School of Medicine, and Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee, USA
| | | |
Collapse
|
34
|
Fike CD, Dikalova A, Slaughter JC, Kaplowitz MR, Zhang Y, Aschner JL. Reactive oxygen species-reducing strategies improve pulmonary arterial responses to nitric oxide in piglets with chronic hypoxia-induced pulmonary hypertension. Antioxid Redox Signal 2013; 18:1727-38. [PMID: 23244497 PMCID: PMC3619184 DOI: 10.1089/ars.2012.4823] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS There are no effective treatments for chronic pulmonary hypertension in infants with cardiopulmonary disorders associated with hypoxia, such as those with chronic lung disease. These patients often have poor or inconsistent pulmonary dilator responses to inhaled nitric oxide (iNO) therapy for unknown reasons. One possible explanation for poor responsiveness to iNO is reduced NO bioavailability caused by interactions between reactive oxygen species (ROS) and NO. Our major aim was to determine if strategies to reduce ROS improve dilator responses to the NO donor, S-nitroso-N-acetyl-penicillamine (SNAP), in resistance pulmonary arteries (PRAs) from a newborn piglet model of chronic pulmonary hypertension. RESULTS The dilation to SNAP was significantly impaired in PRAs from piglets with chronic hypoxia-induced pulmonary hypertension. ROS scavengers, including cell-permeable and impermeable agents to degrade hydrogen peroxide (H(2)O(2)), improved dilation to SNAP in PRAs from chronically hypoxic piglets. Treatment with agents to inhibit nitric oxide synthase and NADPH oxidase, potential enzymatic sources of ROS, also improved dilation to SNAP in PRAs from hypoxic piglets. INNOVATION Our studies are the first to utilize a newborn model of chronic pulmonary hypertension to evaluate the impact of a number of potential therapeutic strategies for ROS removal on responses to exogenous NO in the vessels most relevant to the regulation of pulmonary vascular resistance (PRA). CONCLUSIONS Strategies aimed at reducing ROS merit further evaluation and consideration as therapeutic approaches to improve responses to iNO in infants with chronic pulmonary hypertension.
Collapse
Affiliation(s)
- Candice D Fike
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Bronchopulmonary dysplasia (BPD) is among the most common chronic lung diseases in infants in the US. Improved survival of preterm infants who developed BPD is becoming increasingly important because of the high risk for persistent pulmonary morbidities such as poor respiratory gas exchange, pulmonary hypertension, and excess airway expiratory resistance later in life. This review focuses on unique insights provided by the two large-animal, physiological models of neonatal chronic lung disease: preterm baboons and preterm lambs. The models' are valuable because they contribute to better understanding of the underlying molecular pathogenic mechanisms. An epigenetic hypothesis is proposed as a pathogenic mechanism for BPD and its persistent pulmonary morbidities.
Collapse
Affiliation(s)
- Kurt H. Albertine
- Departments of Pediatrics, Medicine, Neurobiology & Anatomy, University of Utah, School of Medicine, Salt Lake City, Utah 84158-1289
| |
Collapse
|
36
|
Papoff P, Cerasaro C, Caresta E, Barbàra CS, Midulla F, Moretti C. Current strategies for treating infants with severe bronchopulmonary dysplasia. J Matern Fetal Neonatal Med 2012; 25 Suppl 3:15-20. [DOI: 10.3109/14767058.2012.712352] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Abstract
The pulmonary circulation rapidly adapts at birth to establish lungs as the site of gas exchange. Abnormal transition at birth and/or parenchymal lung disease can result in neonatal hypoxemic respiratory failure. This article reviews the functional changes in pulmonary hemodynamics and structural changes in pulmonary vasculature secondary to (1) normal and abnormal transition at birth, and (2) diseases associated with neonatal hypoxemic respiratory failure. Various management strategies to correct respiratory failure are also discussed.
Collapse
Affiliation(s)
- Satyan Lakshminrusimha
- Division of Neonatology, Women and Children's Hospital of Buffalo, State University of New York at Buffalo, 219 Bryant Street, Buffalo, NY 14222, USA.
| |
Collapse
|
38
|
Bhat R, Salas AA, Foster C, Carlo WA, Ambalavanan N. Prospective analysis of pulmonary hypertension in extremely low birth weight infants. Pediatrics 2012; 129:e682-9. [PMID: 22311993 PMCID: PMC3289526 DOI: 10.1542/peds.2011-1827] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES Pulmonary hypertension is associated with bronchopulmonary dysplasia in extremely low birth weight (ELBW) infants and contributes to morbidity and mortality. The objective was to determine the prevalence of pulmonary hypertension among ELBW infants by screening echocardiography and evaluate subsequent outcomes. METHODS All ELBW infants admitted to a regional perinatal center were evaluated for pulmonary hypertension with echocardiography at 4 weeks of age and subsequently if clinical signs suggestive of right-sided heart failure or severe lung disease were evident. Management was at discretion of the clinician, and infants were evaluated until discharge from the hospital or pre-discharge death occurred. RESULTS One hundred forty-five ELBW infants (birth weight: 755 ± 144 g; median gestational age: 26 weeks [interquartile range: 24-27]) were screened from December 2008 to February 2011. Overall, 26 (17.9%) were diagnosed with pulmonary hypertension at any time during hospitalization (birth weight: 665 ± 140 g; median gestational age: 26 weeks [interquartile range: 24-27]): 9 (6.2%) by initial screening (early pulmonary hypertension) and 17 (11.7%) who were identified later (late pulmonary hypertension). Infants with pulmonary hypertension were more likely to receive oxygen treatment on day 28 compared with those without pulmonary hypertension (96% vs 75%, P < .05). Of the 26 infants, 3 died (all in the late group because of cor pulmonale) before being discharged from the hospital. CONCLUSIONS Pulmonary hypertension is relatively common, affecting at least 1 in 6 ELBW infants, and persists to discharge in most survivors. Routine screening of ELBW infants with echocardiography at 4 weeks of age identifies only one-third of the infants diagnosed with pulmonary hypertension. Further research is required to determine optimal detection and intervention strategies.
Collapse
Affiliation(s)
- Ramachandra Bhat
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Ariel A. Salas
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | | | - Waldemar A. Carlo
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | | |
Collapse
|
39
|
Tropea K, Christou H. Current pharmacologic approaches for prevention and treatment of bronchopulmonary dysplasia. Int J Pediatr 2012; 2012:598606. [PMID: 22262977 PMCID: PMC3259479 DOI: 10.1155/2012/598606] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 11/04/2011] [Indexed: 11/23/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a major complication of preterm birth and has serious adverse long-term health consequences. The etiology of BPD is complex, multifactorial, and incompletely understood. Contributing factors include ventilator-induced lung injury, exposure to toxic oxygen levels, and infection. Several preventive and therapeutic strategies have been developed with variable success. These include lung protective ventilator strategies and pharmacological and nutritional interventions. These strategies target different components and stages of the disease process and they are commonly used in combination. The purpose of this review is to discuss the evidence for current pharmacological interventions and identify future therapeutic modalities that appear promising in the prevention and management of BPD. Continued improved understanding of BPD pathogenesis leads to opportunities for newer preventive approaches. These will need to be evaluated in the setting of current clinical practice in order to assess their efficacy.
Collapse
Affiliation(s)
- Kristen Tropea
- Division of Newborn Medicine, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
- Division of Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Helen Christou
- Division of Newborn Medicine, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
- Division of Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
40
|
Kim GB. Pulmonary hypertension in infants with bronchopulmonary dysplasia. KOREAN JOURNAL OF PEDIATRICS 2010; 53:688-93. [PMID: 21189939 PMCID: PMC2994133 DOI: 10.3345/kjp.2010.53.6.688] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Accepted: 05/13/2010] [Indexed: 11/27/2022]
Abstract
An increase in the number of preterm infants and a decrease in the gestational age at birth have resulted in an increase in the number of patients with significant bronchopulmonary dysplasia (BPD) and secondary pulmonary hypertension (PH). PH contributes significantly to the high morbidity and mortality in the BPD patients. Therefore, regular monitoring for PH by using echocardiography and B-type natriuretic peptide (BNP) or N-terminal-proBNP must be conducted in the BPD patients with greater than moderate degree to prevent PH and to ensure early treatment if PH is present. In the BPD patients with significant PH, multi-modality treatment, including treatment for correcting an underlying disease, oxygen supply, use of diverse selective pulmonary vasodilators (inhaled nitric oxide, inhaled prostacyclins, sildenafil, and endothelin-receptor antagonist) and other methods, is mandatory.
Collapse
Affiliation(s)
- Gi Beom Kim
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| |
Collapse
|
41
|
Johnston LC, Gonzales LW, Lightfoot RT, Guttentag SH, Ischiropoulos H. Opposing regulation of human alveolar type II cell differentiation by nitric oxide and hyperoxia. Pediatr Res 2010; 67:521-5. [PMID: 20098340 PMCID: PMC3066065 DOI: 10.1203/pdr.0b013e3181d4f20f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Clinical trials demonstrated decreasing rates of bronchopulmonary dysplasia in preterm infants with hypoxic respiratory failure treated with inhaled nitric oxide (iNO). However, the molecular and biochemical effects of iNO on developing human fetal lungs remain vastly unknown. By using a well-characterized model of human fetal alveolar type II cells, we assessed the effects of iNO and hyperoxia, independently and concurrently, on NO-cGMP signaling pathway and differentiation. Exposure to iNO increased cGMP levels by 40-fold after 3 d and by 8-fold after 5 d despite constant expression of phosphodiesterase-5 (PDE5). The levels of cGMP declined significantly on exposure to iNO and hyperoxia at 3 and 5 d, although expression of soluble guanylyl cyclase (sGC) was sustained. Surfactant proteins B and C (SP-B, SP-C) and thyroid transcription factor (TTF)-1 mRNA levels increased in cells exposed to iNO in normoxia but not on exposure to iNO plus hyperoxia. Collectively, these data indicate an increase in type II cell markers when undifferentiated lung epithelial cells are exposed to iNO in room air. However, hyperoxia overrides these potentially beneficial effects of iNO despite sustained expression of sGC.
Collapse
Affiliation(s)
- Lindsay C Johnston
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
42
|
Inhaled nitric oxide prevents 3-nitrotyrosine formation in the lungs of neonatal mice exposed to >95% oxygen. Lung 2010; 188:217-27. [PMID: 20237791 DOI: 10.1007/s00408-010-9235-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 03/02/2010] [Indexed: 10/19/2022]
Abstract
Inhaled nitric oxide is being evaluated as a preventative therapy for patients at risk for bronchopulmonary dysplasia (BPD). Nitric oxide (NO), in the presence of superoxide, forms peroxynitrite, which reacts with tyrosine residues on proteins to form 3-nitrotyrosine (3-NT). However, NO can also act as an antioxidant and was recently found to improve the oxidative balance in preterm infants. Thus, we tested the hypothesis that the addition of a therapeutically relevant concentration (10 ppm) of NO to a hyperoxic exposure would lead to decreased 3-NT formation in the lung. FVB mouse pups were exposed to either room air (21% O(2)) or >95% O(2) with or without 10 ppm NO within 24 h of birth. In the first set of studies, body weights and survival were monitored for 7 days, and exposure to >95% O(2) resulted in impaired weight gain and near 100% mortality by 7 days. However, the mortality occurred earlier in pups exposed to >95% O(2) + NO than in pups exposed to >95% O(2) alone. In a second set of studies, lungs were harvested at 72 h. Immunohistochemistry of the lungs at 72 h revealed that the addition of NO decreased alveolar, bronchial, and vascular 3-NT staining in pups exposed to both room air and hyperoxia. The lung nitrite levels were higher in animals exposed to >95% oxygen + NO than in animals exposed to >95% oxygen alone. The protein levels of myeloperoxidase, monocyte chemotactic protein-1, and intracellular adhesion molecule-1 were assessed after 72 h of exposure and found to be greatest in the lungs of pups exposed to >95% O(2). This hyperoxia-induced protein expression was significantly attenuated by the addition of 10 ppm NO. We propose that in the presence of >95% O(2), peroxynitrite formation results in protein nitration; however, adding excess NO to the >95% O(2) exposure prevents 3-NT formation by NO reacting with peroxynitrite to produce nitrite and NO(2). We speculate that the decreased protein nitration observed with the addition of NO may be a potential mechanism limiting hyperoxic lung injury.
Collapse
|
43
|
|
44
|
Inhaled nitric oxide to prevent bronchopulmonary dysplasia in preterm neonates. Semin Fetal Neonatal Med 2009; 14:28-34. [PMID: 18986855 DOI: 10.1016/j.siny.2008.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bronchopulmonary dysplasia is a chronic lung disease that affects premature infants and contributes to their morbidity and mortality. With the advent of prenatal steroids and postnatal exogenous surfactant and less aggressive respiratory support, premature infants can develop chronic oxygen dependency without even acute respiratory distress. This 'new bronchopulmonary dysplasia' could be the result of impaired postnatal growth. Several experimental studies have suggested a possible role of the vascular endothelial growth factor/nitric oxide (VEGF/NO) pathway in restoring pulmonary angiogenesis and enhancing distal lung growth. The results of the clinical studies are, however, inconclusive, and it is currently unclear which subsets of premature infants might benefit from inhaled nitric oxide. Besides, severe intracranial haemorrhage and/or cystic periventricular leucomalacia may affect the most immature babies, many of whom are spared from severe initial respiratory disease. Recently, inhaled nitric oxide was shown to significantly decrease the incidence of these neurological events, and to improve the long-term outcome in a few clinical trials. At times neuroprotective, at times neurotoxic, nitric oxide is capable of divergent effects depending upon the extent of cerebral damage, the redox state of the cell, and the experimental model used. Recently, our group found that inhaled nitric oxide had remote effects including angiogenesis and maturation on the developing brain in rodent pups. Thus, we await the results of the recently completed randomised clinical trial of inhaled nitric oxide to prevent bronchopulmonary dysplasia (the European Nitric Oxide or 'EUNO' trial) where, besides the primary endpoint of chronic oxygen dependency reduction at 36 weeks' postconceptional age, long-term lung and brain will be followed-up until 7 years of age.
Collapse
|
45
|
Richir MC, van Leeuwen PAM, van den Berg A, Wessels R, Twisk JWR, Rauwerda JA, Teerlink T, de Vries TPGM, van Elburg RM. Plasma ADMA concentrations at birth and mechanical ventilation in preterm infants: a prospective pilot study. Pediatr Pulmonol 2008; 43:1161-6. [PMID: 18991340 DOI: 10.1002/ppul.20886] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
RATIONALE Nitric oxide (NO) produced in the lung is an important mediator of normal lung development, vascular smooth muscle relaxation, and ventilation perfusion matching. NO is synthesized from arginine by the action of NO-synthase (NOS). Asymmetric dimethylarginine (ADMA), an endogenous derivate of arginine, inhibits NOS and is thereby a determinant of NO synthesis. We compared ADMA and arginine levels in preterm infants requiring mechanical ventilation with preterm infants who did not require mechanical ventilation and determined the relation between ADMA and the length of mechanical ventilation in these infants. METHODS Thirty preterm infants, mean (SD) gestational age 29.3 (1.7) weeks and birth weight 1,340 (350) gram, of the Neonatal Intensive Care Unit of the VU University Medical Center were included. ADMA and arginine were measured in umbilical cord blood and the length of mechanical ventilation (days) was registered. RESULTS Gestational age and birth weight were significantly smaller in infants requiring mechanical ventilation, but were not significantly correlated with plasma ADMA concentration after birth. Plasma ADMA concentrations were significantly higher in infants who required mechanical ventilation than in infants who did not require mechanical ventilation (1.53 +/- 0.23 and 1.37 +/- 0.14 micromol/L, respectively; P = 0.036). ADMA concentration was significantly related to length of mechanical ventilation (B = 3.4; 95% CI: 1.1-5.6; P = 0.006), also after adjustment for gestational age (B = 2.3; 95% CI: 0.4-4.2; P = 0.024). CONCLUSIONS Preterm infants who require mechanical ventilation have increased ADMA levels compared to non-ventilated preterm infants. ADMA levels at birth are related to the length of mechanical ventilation. An increased ADMA concentration could reduce NO synthesis, which could lead to insufficient gas exchange and, consequently, a longer period of mechanical ventilation.
Collapse
Affiliation(s)
- Milan C Richir
- Department of Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Inhaled nitric oxide (iNO) has quickly become a standard therapy for term and near-term infants with hypoxic respiratory failure and persistent pulmonary hypertension. Its effect on the lung is believed to be through the stimulation of soluble guanylyl cyclase and the increased production of cyclic guanosine 3',5'-monophosphate (cGMP). However, in addition to pulmonary vasodilation and a decrease in pulmonary vascular resistance, nitric oxide (NO) shows several additional potential beneficial effects on the lung. This article reviews NO mechanisms of action, early clinical trial of iNO and clinical aspects for the use of iNO in acute respiratory failure of the term and near-tem neonates.
Collapse
|
47
|
MESH Headings
- Brain Diseases/etiology
- Bronchopulmonary Dysplasia/etiology
- Bronchopulmonary Dysplasia/prevention & control
- Developmental Disabilities/etiology
- Ductus Arteriosus, Patent/epidemiology
- Enterocolitis, Necrotizing/etiology
- Humans
- Infant Care
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/epidemiology
- Infant, Premature, Diseases/etiology
- Infant, Premature, Diseases/prevention & control
- Infant, Very Low Birth Weight
- Neonatology
- Prognosis
- Respiration, Artificial
Collapse
Affiliation(s)
- Eric C Eichenwald
- Department of Pediatrics and the Section of Neonatology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA.
| | | |
Collapse
|
48
|
Tin W, Wiswell TE. Adjunctive therapies in chronic lung disease: examining the evidence. Semin Fetal Neonatal Med 2008; 13:44-52. [PMID: 17983879 DOI: 10.1016/j.siny.2007.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Chronic lung disease (CLD) or bronchopulmonary dysplasia (BPD) is one of the most common long-term complications in very premature infants. The incidence of CLD has been increasing over the past two decades in parallel with an improvement in the survival of this population. We have witnessed a revolution in the therapies that are used, either to manage these infants' respiratory distress syndrome (RDS) with an aim to prevent CLD or to manage the established condition. Several devices and strategies have been developed to provide respiratory support with minimal risk of lung injuries. Multiple adjunctive agents have also been used either to reduce the risk of CLD or to mitigate its course. There is considerable evidence supporting the use of exogenous surfactant, but unfortunately many other therapies currently used for CLD, either preventative or as a treatment, are based on very little or no evidence. The gold standard to assess a given therapy is the randomised controlled trial (RCT), designed to look at clinically meaningful outcomes and long-term safety. In this context, we discuss the support - or lack thereof - for the adjunctive therapies used in relation to CLD. Many of the therapies have been examined as systematic reviews by the Cochrane Neonatal Review Group. These reviews are noted in the references and can be easily accessed at the following website sponsored by the National Institute of Child Health and Human Development: www.nichd.nih.gov/cochrane/default.cfm.
Collapse
Affiliation(s)
- Win Tin
- The James Cook University Hospital, Marton Road, Middlesbrough, TS4 3BW, UK.
| | | |
Collapse
|
49
|
Abstract
OBJECTIVE Elevated pulmonary vascular resistance and poor ventilation-perfusion matching are commonly found in preterm infants with severe respiratory distress syndrome (RDS) and respiratory failure. Inhaled nitric oxide (iNO) can improve gas exchange and decrease pulmonary vascular resistance. This study was conducted to determine whether iNO therapy improves oxygenation in such infants. STUDY DESIGN Between July 2000 and 2006, 65 preterm infants (birth weight, <1500 g; gestational age, <31 weeks) with severe RDS and respiratory failure requiring mechanical ventilation and an oxygenation index (OI)>or=25 were randomly divided into two groups. Group A infants (n=32) received iNO therapy. iNO was started at a dose of five parts per million (p.p.m.). The maximal dose of NO was 20 p.p.m. Group B infants (n=33) did not receive iNO therapy, receive inhaled oxygen placebo only, was served as control group. Mechanical ventilation and iNO therapy were managed by neonatologists who were not involved in safety monitoring, data analysis and interpretation, or manuscript preparation. This study was randomized but not blinded. RESULT The OI was significantly lower (P<0.01) in the iNO therapy group than in the control group at 30 min, 3, 12 and 24 h after initiating iNO therapy. Six infants in the iNO-treated group and 10 infants in the control group died. Post hoc analyses did not reveal any significant differences in the incidences of chronic lung disease (CLD), intracranial hemorrhage (ICH), patent ductus arteriosus (PDA), retinopathy of prematurity (ROP) or duration of intubation between the iNO-treated and the control groups. CONCLUSION We conclude that iNO therapy leads to an improvement in oxygenation without short-term side effects (such as pulmonary hemorrhage, intracranial hemorrhage, pneumothorax or acute deterioration) in premature infants with severe RDS and respiratory failure. However, iNO therapy does not significantly reduce mortality rate or the incidences of CLD, ICH, PDA or ROP.
Collapse
|
50
|
Trevisanuto D, Doglioni N, Micaglio M, Zanardo V. Feasibility of nitric oxide administration by neonatal helmet-CPAP: a bench study. Paediatr Anaesth 2007; 17:851-5. [PMID: 17683403 DOI: 10.1111/j.1460-9592.2007.02297.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Inhaled nitric oxide (NO) may have a role in the treatment of preterm infants with respiratory failure. We evaluated the feasibility of administering NO therapy by a new continuous positive airway pressure (CPAP) system (neonatal helmet-CPAP). METHODS While maintaining a constant total flow of 8, 10, and 12 l.min(-1), NO concentrations were progressively increased to 5, 10, 20, and 40 p.p.m. in the neonatal helmet-CPAP pressure chamber (5 cmH2O). NO, NO2, and O2 concentrations were measured in the pressure chamber and the immediate external environment. RESULTS In the chamber, NO2 levels remained low (<or=0.8 p.p.m.) at inhaled therapeutic NO concentrations (5, 10, 20, and 40 p.p.m.). The lower O2 concentrations were 95% at 40 p.p.m. NO levels. Leakage of NO and NO2 to the surrounding environment was negligible. CONCLUSIONS NO administration is safe and feasible using the neonatal helmet-CPAP system. This method allows the delivery of accurate NO levels and high O2 concentrations avoiding NO2 accumulation. Further experimental and clinical studies are needed.
Collapse
Affiliation(s)
- Daniele Trevisanuto
- Pediatric Department, Neonatal Intensive Care Unit, Medical School, University of Padova, Azienda Ospedaliera di Padova, Padova, Italy.
| | | | | | | |
Collapse
|