1
|
Collaco JM, Eldredge LC, McGrath-Morrow SA. Long-term pulmonary outcomes in BPD throughout the life-course. J Perinatol 2024:10.1038/s41372-024-01957-9. [PMID: 38570594 DOI: 10.1038/s41372-024-01957-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Respiratory disease is one of the most common complications of preterm birth. Survivors of prematurity have increased risks of morbidities and mortalities independent of prematurity, and frequently require multiple medications, home respiratory support, and subspecialty care to maintain health. Although advances in neonatal and pulmonary care have improved overall survival, earlier gestational age, lower birth weight, chorioamnionitis and late onset sepsis continue to be major factors in the development of bronchopulmonary dysplasia. These early life events associated with prematurity can have respiratory consequences that persist into adulthood. Furthermore, after initial hospital discharge, air pollution, respiratory tract infections and socioeconomic status may modify lung growth trajectories and influence respiratory outcomes in later life. Given that the incidence of respiratory disease associated with prematurity remains stable or increased, there is a need for pediatric and adult providers to be familiar with the natural history, manifestations, and common complications of disease.
Collapse
Affiliation(s)
- Joseph M Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Laurie C Eldredge
- Division of Pediatric Pulmonology, Seattle Children's Hospital, Seattle, WA, USA
| | - Sharon A McGrath-Morrow
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Chou HC, Chen CM. Hyperoxia Induces Ferroptosis and Impairs Lung Development in Neonatal Mice. Antioxidants (Basel) 2022; 11:antiox11040641. [PMID: 35453326 PMCID: PMC9032171 DOI: 10.3390/antiox11040641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Oxygen is often required to treat newborns with respiratory disorders, and prolonged exposure to high oxygen concentrations impairs lung development. Ferroptosis plays a vital role in the development of many diseases and has become the focus of treatment and prognosis improvement for related diseases, such as neurological diseases, infections, cancers, and ischemia-reperfusion injury. Whether ferroptosis participates in the pathogenesis of hyperoxia-induced lung injury remains unknown. The aims of this study are to determine the effects of hyperoxia on lung ferroptosis and development in neonatal mice. Newborn C57BL/6 mice were reared in either room air (RA) or hyperoxia (85% O2) at postnatal days 1–7. On postnatal days 3 and 7, the lungs were harvested for histological and biochemical analysis. The mice reared in hyperoxia exhibited significantly higher Fe2+, malondialdehyde, and iron deposition and significantly lower glutathione, glutathione peroxidase 4, and vascular density than did those reared in RA on postnatal days 3 and 7. The mice reared in hyperoxia exhibited a comparable mean linear intercept on postnatal day 3 and a significantly higher mean linear intercept than the mice reared in RA on postnatal day 7. These findings demonstrate that ferroptosis was induced at a time point preceding impaired lung development, adding credence to the hypothesis that ferroptosis is involved in the pathogenesis of hyperoxia-induced lung injury and suggest that ferroptosis inhibitors might attenuate hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence:
| |
Collapse
|
3
|
Abstract
In evaluating vitamin E (VE) nutritional status of preterm infants, it is essential that any data should be compared with those of healthy term infants, and never with those of adults. Moreover, it should be evaluated in terms of gestational age (GA), not birth weight (BW), because placental transfer of most nutrients from mother to fetus is dependent on GA, not BW. Judging from the limited data during the last 75 years, there was no significant correlation between GA and VE concentrations in circulation or in the red blood cells (RBCs), leukocytes, and buccal mucosal cells. In addition, the oxidizability of polyunsaturated fatty acids (PUFAs) in plasma or RBCs, as targets for protection by VE chain-breaking ability, was lower in preterm infants. However, because of the minimal information available about hepatic VE levels, which is considered a key determinant of whole body VE status, the decision on whether VE status of preterm infants is comparable with that of term infants should be postponed. Clinical trials of VE supplementation in preterm infants were repeatedly undertaken to investigate whether VE reduces severity or inhibits development of several diseases specific to preterm infants, namely retinopathy of prematurity (ROP), bronchopulmonary dysplasia (BPD), and germinal matrix hemorrhage - intraventricular hemorrhage (GMH-IVH). Most of these trials resulted in a misfire, with a few exceptions for IVH prevention. However, almost all these studies were performed from 1980s to early 1990s, in the pre-surfactant era, and the study populations were composed of mid-preterm infants with GAs of approximately 30 weeks (wks). There is considerable difference in 'preterm infants' between the pre- and post-surfactant eras; modern neonatal medicine mainly treats preterm infants of 28 wks GA or less. Therefore, these results are difficult to apply in modern neonatal care. Before considering new trials of VE supplementation, we should fully understand modern neonatal medicine, especially the recent method of oxygen supplementation. Additionally, a deeper understanding of recent progress in pathophysiology and therapies for possible target diseases is necessary to decide whether VE administration is still worth re-challenging in modern neonatal intensive care units (NICUs). In this review, we present recent concepts and therapeutic trends in ROP, BPD, and GMH-IVH for those unfamiliar with neonatal medicine. Numerous studies have reported the possible involvement of reactive oxygen species (ROS)-induced damage in relation to supplemental oxygen use, inflammation, and immature antioxidant defense in the development of both BPD and ROP. Various antioxidants effectively prevented the exacerbation of BPD and ROP in animal models. In the future, VE should be re-attempted as a complementary factor in combination with various therapies for BPD, ROP, and GMH-IVH. Because VE is a natural and safe supplement, we are certain that it will attract attention again in preterm medicine.
Collapse
Affiliation(s)
- Tohru Ogihara
- Division of Neonatology, Department of Pediatrics, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan.
| | - Makoto Mino
- Division of Neonatology, Department of Pediatrics, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| |
Collapse
|
4
|
Tong Y, Zuo J, Yue D. Application Prospects of Mesenchymal Stem Cell Therapy for Bronchopulmonary Dysplasia and the Challenges Encountered. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9983664. [PMID: 33997051 PMCID: PMC8110410 DOI: 10.1155/2021/9983664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in premature babies, especially affecting those with very low or extremely low birth weights. Survivors experience adverse lung and neurological defects including cognitive dysfunction. This impacts the prognosis of children with BPD and may result in developmental delays. The currently available options for the treatment of BPD are limited owing to low efficacy or several side effects; therefore, there is a lack of effective treatments for BPD. The treatment for BPD must help in the repair of damaged lung tissue and promote further growth of the lung tissue. In recent years, the emergence of stem cell therapy, especially mesenchymal stem cell (MSC) therapy, has improved the treatment of BPD to a great extent. This article briefly reviews the advantages, research progress, and challenges faced with the use of MSCs in the treatment of BPD. Stem cell therapy is beneficial as it repairs damaged tissues by reducing inflammation, fibrosis, and by acting against oxidative stress damage. Experimental trials have also proven that MSCs provide a promising avenue for BPD treatment. However, there are challenges such as the possibility of MSCs contributing to tumorous growths, the presence of heterogeneous cell populations resulting in variable efficacy, and the ethical considerations regarding the use of this treatment in humans. Therefore, more research must be conducted to determine whether MSC therapy can be approved as a treatment option for BPD.
Collapse
Affiliation(s)
- Yajie Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| | - Jingye Zuo
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| | - Dongmei Yue
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| |
Collapse
|
5
|
Porzionato A, Zaramella P, Dedja A, Guidolin D, Bonadies L, Macchi V, Pozzobon M, Jurga M, Perilongo G, De Caro R, Baraldi E, Muraca M. Intratracheal administration of mesenchymal stem cell-derived extracellular vesicles reduces lung injuries in a chronic rat model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2021; 320:L688-L704. [PMID: 33502939 DOI: 10.1152/ajplung.00148.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Early therapeutic effect of intratracheally (IT)-administered extracellular vesicles secreted by mesenchymal stem cells (MSC-EVs) has been demonstrated in a rat model of bronchopulmonary dysplasia (BPD) involving hyperoxia exposure in the first 2 postnatal weeks. The aim of this study was to evaluate the protective effects of IT-administered MSC-EVs in the long term. EVs were produced from MSCs following GMP standards. At birth, rats were distributed in three groups: (a) animals raised in ambient air for 6 weeks (n = 10); and animals exposed to 60% hyperoxia for 2 weeks and to room air for additional 4 weeks and treated with (b) IT-administered saline solution (n = 10), or (c) MSC-EVs (n = 10) on postnatal days 3, 7, 10, and 21. Hyperoxia exposure produced significant decreases in total number of alveoli, total surface area of alveolar air spaces, and proliferation index, together with increases in mean alveolar volume, mean linear intercept and fibrosis percentage; all these morphometric changes were prevented by MSC-EVs treatment. The medial thickness index for <100 µm vessels was higher for hyperoxia-exposed/sham-treated than for normoxia-exposed rats; MSC-EV treatment significantly reduced this index. There were no significant differences in interstitial/alveolar and perivascular F4/8-positive and CD86-positive macrophages. Conversely, hyperoxia exposure reduced CD163-positive macrophages both in interstitial/alveolar and perivascular populations and MSC-EV prevented these hyperoxia-induced reductions. These findings further support that IT-administered EVs could be an effective approach to prevent/treat BPD, ameliorating the impaired alveolarization and pulmonary artery remodeling also in a long-term model. M2 macrophage polarization could play a role through anti-inflammatory and proliferative mechanisms.
Collapse
Affiliation(s)
- Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Patrizia Zaramella
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Arben Dedja
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Diego Guidolin
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Luca Bonadies
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Michela Pozzobon
- Institute of Pediatric Research, Padua, Italy.,Stem Cell and Regenerative Medicine Laboratory, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Marcin Jurga
- The Cell Factory BVBA (Esperite NV), Niel, Belgium
| | - Giorgio Perilongo
- Institute of Pediatric Research, Padua, Italy.,Pediatric Clinic, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy.,Institute of Pediatric Research, Padua, Italy
| | - Maurizio Muraca
- Institute of Pediatric Research, Padua, Italy.,Stem Cell and Regenerative Medicine Laboratory, Department of Women's and Children's Health, University of Padova, Padua, Italy
| |
Collapse
|
6
|
Ding Y, Chen Z, Lu Y. Vitamin A supplementation prevents the bronchopulmonary dysplasia in premature infants: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e23101. [PMID: 33545924 PMCID: PMC7837939 DOI: 10.1097/md.0000000000023101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/16/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND It is necessary to evaluate the effectiveness and safety of vitamin A supplementation on the bronchopulmonary dysplasia (BPD) in premature infants. METHODS Randomized controlled trials (RCTs) on the role of supplemental vitamin A in preterm infants were searched. The Medline et al databases were manually searched from inception to April 30, 2020. Related outcomes including incidence of BPD, retinopathy of prematurity (ROP), necrotizing enterocolitis (NEC), intraventricular hemorrhage (IVH), sepsis and mortality were assessed with Review Manager 5.3 software, and Random-effect model was applied for all conditions. RESULTS A total of 9 RCTs with 1409 patients were included. The analyzed results showed that the incidence of BPD in vitamin A group was significantly less than that of control group (OR = 0.67, 95%CI [0.52-0.88]). There was no significant difference in the incidence of ROP (OR = 0.65, 95%CI [0.29-1.48]), NEC (OR = 0.88, 95%CI [0.59-1.30]), IVH (OR = 0.90, 95%CI [0.65-1.25]), sepsis (OR = 0.84, 95%CI [0.64-1.09]) and mortality (OR = 0.98, 95%CI [0.72-1.34]) among two groups. CONCLUSION Vitamin A supplementation is beneficial to the prophylaxis of BPD in premature infants, further studies on the administration approaches and dosages of vitamin A in premature infants are warranted.
Collapse
|
7
|
Chen CM, Hwang J, Chou HC, Chen C. Anti-Tn Monoclonal Antibody Attenuates Hyperoxia-Induced Lung Injury by Inhibiting Oxidative Stress and Inflammation in Neonatal Mice. Front Pharmacol 2020; 11:568502. [PMID: 33013407 PMCID: PMC7506044 DOI: 10.3389/fphar.2020.568502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Maternal immunization with Tn vaccine increases serum anti-Tn antibody titers and attenuates hyperoxia-induced lung injury in neonatal rats. This study determined whether anti-Tn monoclonal antibody can protect against hyperoxia-induced lung injury in neonatal mice. Newborn BALB/c mice were exposed to room air (RA) or normobaric hyperoxia (85% O2) for 1 week, creating four study groups as follows: RA + phosphate-buffered saline (PBS), RA + anti-Tn monoclonal antibody, O2 + PBS, and O2 + anti-Tn monoclonal antibody. The anti-Tn monoclonal antibody at 25 μg/g body weight in 50 μl PBS was intraperitoneally injected on postnatal days 2, 4, and 6. Hyperoxia reduced body weight and survival rate, increased mean linear intercept (MLI) and lung tumor necrosis factor-α, and decreased vascular endothelial growth factor (VEGF) expression and vascular density on postnatal day 7. Anti-Tn monoclonal antibody increased neonatal serum anti-Tn antibody titers, reduced MLI and cytokine, and increased VEGF expression and vascular density to normoxic levels. The attenuation of lung injury was accompanied by a reduction in lung oxidative stress and nuclear factor-κB activity. Anti-Tn monoclonal antibody improves alveolarization and angiogenesis in hyperoxia-injured newborn mice lungs through the suppression of oxidative stress and inflammation.
Collapse
Affiliation(s)
- Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jaulang Hwang
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chinde Chen
- Department of Research and Development, Taivital Biopharmaceutical Co. LTD, Taoyuan, Taiwan
| |
Collapse
|
8
|
Willis GR, Fernandez-Gonzalez A, Reis M, Yeung V, Liu X, Ericsson M, Andrews NA, Mitsialis SA, Kourembanas S. Mesenchymal stromal cell-derived small extracellular vesicles restore lung architecture and improve exercise capacity in a model of neonatal hyperoxia-induced lung injury. J Extracell Vesicles 2020; 9:1790874. [PMID: 32939235 PMCID: PMC7480622 DOI: 10.1080/20013078.2020.1790874] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Early administration of mesenchymal stromal cell (MSC)-derived small extracellular vesicles (MEx) has shown considerable promise in experimental models of bronchopulmonary dysplasia (BPD). However, the ability of MEx to reverse the long-term pulmonary complications associated with established BPD remains unknown. In this study, MEx were isolated from media conditioned by human Wharton’s Jelly-derived MSC cultures. Newborn mice (FVB strain) were exposed to hyperoxia (HYRX (75% O2)) before returning to room air at postnatal day 14 (PN14). Following prolonged HYRX-exposure, animals received a single MEx dose at PN18 or serial MEx treatments at PN18-39 (“late” intervention). This group was compared to animals that received an early single MEx dose at PN4 (“early” intervention). Animals were harvested at PN28 or 60 for assessment of pulmonary parameters. We found that early and late MEx interventions effectively ameliorated core features of HYRX-induced neonatal lung injury, improving alveolar simplification, pulmonary fibrosis, vascular remodelling and blood vessel loss. Exercise capacity testing and assessment of pulmonary hypertension (PH) showed functional improvements following both early and late MEx interventions. In conclusion, delivery of MEx following prolonged HYRX-exposure improves core features of experimental BPD, restoring lung architecture, decreasing pulmonary fibrosis and vascular muscularization, ameliorating PH and improving exercise capacity. Taken together, delivery of MEx may not only be effective in the immediate neonatal period to prevent the development of BPD but may provide beneficial effects for the management and potentially the reversal of cardiorespiratory complications in infants and children with established BPD.
Collapse
Affiliation(s)
- Gareth R Willis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Monica Reis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Vincent Yeung
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Xianlan Liu
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Maria Ericsson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Nick A Andrews
- F.M. Kirby Center for Neurobiology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Lignelli E, Palumbo F, Myti D, Morty RE. Recent advances in our understanding of the mechanisms of lung alveolarization and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L832-L887. [PMID: 31596603 DOI: 10.1152/ajplung.00369.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common cause of morbidity and mortality in preterm infants. A key histopathological feature of BPD is stunted late lung development, where the process of alveolarization-the generation of alveolar gas exchange units-is impeded, through mechanisms that remain largely unclear. As such, there is interest in the clarification both of the pathomechanisms at play in affected lungs, and the mechanisms of de novo alveoli generation in healthy, developing lungs. A better understanding of normal and pathological alveolarization might reveal opportunities for improved medical management of affected infants. Furthermore, disturbances to the alveolar architecture are a key histopathological feature of several adult chronic lung diseases, including emphysema and fibrosis, and it is envisaged that knowledge about the mechanisms of alveologenesis might facilitate regeneration of healthy lung parenchyma in affected patients. To this end, recent efforts have interrogated clinical data, developed new-and refined existing-in vivo and in vitro models of BPD, have applied new microscopic and radiographic approaches, and have developed advanced cell-culture approaches, including organoid generation. Advances have also been made in the development of other methodologies, including single-cell analysis, metabolomics, lipidomics, and proteomics, as well as the generation and use of complex mouse genetics tools. The objective of this review is to present advances made in our understanding of the mechanisms of lung alveolarization and BPD over the period 1 January 2017-30 June 2019, a period that spans the 50th anniversary of the original clinical description of BPD in preterm infants.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
10
|
Arsan S, Korkmaz A, Oğuz S. Turkish Neonatal Society guideline on prevention and management of bronchopulmonary dysplasia. TURK PEDIATRI ARSIVI 2018; 53:S138-S150. [PMID: 31236027 PMCID: PMC6568289 DOI: 10.5152/turkpediatriars.2018.01814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Scientific and technological advances in perinatology and neonatology have led to an increased rate of survival and decreased incidences of various neonatal morbidities. However, the incidence of bronchopulmonary dysplasia has remained almost the same for years in very-low-birth-weight preterm infants. Although bronchopulmonary dysplasia is the leading cause of chronic respiratory morbidity in small preterms, no substantial improvement has been achieved in prevention and treatment strategies to date. Currently, postnatal very-low-dose corticosteroids, caffeine, and vitamin A seem to be the drugs of choice, and stem cell therapy appears to be the most promising treatment modality for the future. In this guideline, which was prepared by the Turkish Neonatal Society, recent evidence-based recommendations for the prevention and treatment of bronchopulmonary dysplasia are summarized.
Collapse
Affiliation(s)
- Saadet Arsan
- Division of Neonatology, Department of Pediatrics, Ankara University, Faculty of Medicine, Ankara, Turkey
| | - Ayşe Korkmaz
- Division of Neonatology, Department of Pediatrics, Acıbadem University, Faculty of Medicine, İstanbul, Turkey
| | - Suna Oğuz
- Zekai Tahir Burak Women’s Health Practice and Research Center, Health Sciences University, Ankara, Turkey
| |
Collapse
|
11
|
Guillot M, Offringa M, Lacaze-Masmonteil T, Thébaud B. Cell-based therapy for bronchopulmonary dysplasia in preterm infants 1. Can J Physiol Pharmacol 2018; 97:232-234. [PMID: 30290122 DOI: 10.1139/cjpp-2018-0342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of extreme prematurity. Currently, there is no specific treatment available. Preclinical studies support cell therapy as a promising therapy for BPD in preterm infants. A successful translation to a safe and effective clinical intervention depends on multiple factors including the perspective of neonatal health care providers. A 2-hour workshop with 40 Canadian neonatologists was held to enhance the design of a phase II trial of stem cells for babies at risk for BPD, with a focus on the population to target and the outcomes to measure in such a trial. The consensus was that infants recruited in an early trial of stem cells should be the ones with the highest risk of developing severe BPD. This risk should be established based on known antenatal, perinatal, and postnatal risk factors. The primary outcome in a phase II trial will be focussed on a non-clinical outcome (e.g., a dose-finding study or a safety study). With other aspects of a translational study discussed, this workshop contributed to accelerate the design of a first Canadian clinical cell-therapy study for BPD in preterm infants.
Collapse
Affiliation(s)
- Mireille Guillot
- a Children's Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada.,b The Ottawa Hospital, General Campus, Ottawa, ON K1H 8L6, Canada.,c University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Martin Offringa
- d The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada.,e Child Health Evaluative Sciences, Sick Kids Research Institute, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Thierry Lacaze-Masmonteil
- f Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1 Canada.,g Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Bernard Thébaud
- a Children's Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada.,b The Ottawa Hospital, General Campus, Ottawa, ON K1H 8L6, Canada.,c University of Ottawa, Ottawa, ON K1N 6N5, Canada.,h Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW This review provides a concise summary of recent literature pertaining to emerging therapies for bronchopulmonary dysplasia (BPD). To provide context for the presented therapies, a brief overview of recently proposed changes to the definition of BPD and the concept of expanded respiratory outcomes is included. RECENT FINDINGS New or redefined respiratory outcomes are required to improve accuracy in evaluating new therapies and correlating results with long-term clinical outcomes of importance. Dexamethasone is no longer the only steroid-based therapy showing promise for impacting BPD. Early trials indicate hydrocortisone, inhaled budesonide, and a budesonide-surfactant combination may be of benefit to preterm infants. Additionally, simple approaches like increasing utilization of mother's own milk may deserve more emphasis. Of significant interest is the traction stem cell therapies are acquiring as one of the more anticipated treatments for BPD. A new preclinical meta-analysis demonstrates the benefits of mesenchymal stromal cell therapy in animal models while the results of early clinical trials remain eagerly awaited. SUMMARY BPD continues to be the most frequently occurring significant morbidity for extremely preterm infants, yet highly effective therapies remain elusive. Promising new treatments are on the horizon, but only continued efforts to complete well-designed clinical trials will determine the true impact of these emerging therapies.
Collapse
|