1
|
Van Der Heijden JEM, Van Hove H, Van Elst NM, Van Den Broek P, Van Drongelen J, Scheepers HCJ, De Wildt SN, Greupink R. Optimization of the betamethasone and dexamethasone dosing regimen during pregnancy: a combined placenta perfusion and pregnancy physiologically based pharmacokinetic modeling approach. Am J Obstet Gynecol 2025; 232:228.e1-228.e9. [PMID: 38763343 DOI: 10.1016/j.ajog.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Antenatal betamethasone and dexamethasone are prescribed to women who are at high risk of premature birth to prevent neonatal respiratory distress syndrome (RDS). The current treatment regimens, effective to prevent neonatal RDS, may be suboptimal. Recently, concerns have been raised regarding possible adverse long-term neurological outcomes due to high fetal drug exposures. Data from nonhuman primates and sheep suggest maintaining a fetal plasma concentration above 1 ng/mL for 48 hours to retain efficacy, while avoiding undesirable high fetal plasma levels. OBJECTIVE We aimed to re-evaluate the current betamethasone and dexamethasone dosing strategies to assess estimated fetal exposure and provide new dosing proposals that meet the efficacy target but avoid excessive peak exposures. STUDY DESIGN A pregnancy physiologically based pharmacokinetic (PBPK) model was used to predict fetal drug exposures. To allow prediction of the extent of betamethasone and dexamethasone exposure in the fetus, placenta perfusion experiments were conducted to determine placental transfer. Placental transfer rates were integrated in the PBPK model to predict fetal exposure and model performance was verified using published maternal and fetal pharmacokinetic data. The verified pregnancy PBPK models were then used to simulate alternative dosing regimens to establish a model-informed dose. RESULTS Ex vivo data showed that both drugs extensively cross the placenta. For betamethasone 15.7±1.7% and for dexamethasone 14.4±1.5%, the initial maternal perfusate concentration reached the fetal circulations at the end of the 3-hour perfusion period. Pregnancy PBPK models that include these ex vivo-derived placental transfer rates accurately predicted maternal and fetal exposures resulting from current dosing regimens. The dose simulations suggest that for betamethasone intramuscular, a dose reduction from 2 dosages 11.4 mg, 24 hours apart, to 4 dosages 1.425 mg, 12 hours apart would avoid excessive peak exposures and still meet the fetal response threshold. For dexamethasone, the dose may be reduced from 4 times 6 mg every 12 hours to 8 times 1.5 mg every 6 hours. CONCLUSION A combined placenta perfusion and pregnancy PBPK modeling approach adequately predicted both maternal and fetal drug exposures of 2 antenatal corticosteroids (ACSs). Strikingly, our PBPK simulations suggest that drug doses might be reduced drastically to still meet earlier proposed efficacy targets and minimize peak exposures. We propose the provided model-informed dosing regimens are used to support further discussion on an updated ACS scheme and design of clinical trials to confirm the effectiveness and safety of lower doses.
Collapse
Affiliation(s)
- Joyce E M Van Der Heijden
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Hedwig Van Hove
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niki M Van Elst
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Petra Van Den Broek
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joris Van Drongelen
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hubertina C J Scheepers
- Department of Obstetrics and Gynecology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Saskia N De Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Pediatric and Neonatal Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Rick Greupink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Baud O, Sentilhes L, Ursino M, Doret-Dion M, Alberti C, Aupiais C, Schmitz T. Survival without severe neonatal morbidity after antenatal betamethasone dose reduction: a post hoc analysis of a randomized non-inferiority trial. Am J Obstet Gynecol 2024; 231:458.e1-458.e16. [PMID: 38341166 DOI: 10.1016/j.ajog.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/29/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Antenatal betamethasone is recommended before preterm delivery to accelerate fetal lung maturation. However, its optimal dose remains unknown. A 50% dose reduction was proposed to decrease the potential dose-related long-term neurodevelopmental side effects, including psychological development, sleep, and emotional disorders. Because noninferiority of the half dose in terms of the need for exogenous surfactant was not shown in the primary analysis, its impact on survival without major neonatal morbidity needs to be investigated. OBJECTIVE This study aimed to investigate the impact of antenatal betamethasone dose reduction on survival of very preterm infants without severe neonatal morbidity, a factor known to have a strong correlation with long-term outcomes. STUDY DESIGN We performed a post hoc secondary analysis of a randomized, multicenter, double-blind, placebo-controlled, noninferiority trial, testing half (11.4 mg once; n=1620) vs full (11.4 mg twice, 24 hours apart; n=1624) antenatal betamethasone doses in women at risk of preterm delivery. To measure survival without severe neonatal morbidity at hospital discharge among neonates born before 32 weeks of gestation, we used the definition of the French national prospective study on preterm children, EPIPAGE 2, comprising 1 of the following morbidities: grade 3 to 4 intraventricular hemorrhage, cystic periventricular leukomalacia, necrotizing enterocolitis stage ≥2, retinopathy of prematurity requiring anti-vascular endothelial growth factor therapy or laser, and moderate-to-severe bronchopulmonary dysplasia. RESULTS After exclusion of women who withdrew consent or had pregnancy termination and of participants lost to follow-up (8 in the half-dose and 10 in the full-dose group), the rate of survival without severe neonatal morbidity among neonates born before 32 weeks of gestation was 300 of 451 (66.5%) and 304 of 462 (65.8%) in the half-dose and full-dose group, respectively (risk difference, +0.7%; 95% confidence interval, -5.6 to +7.1). There were no significant between-group differences in the cumulative number of neonatal morbidities. Results were similar when using 2 other internationally recognized definitions of severe neonatal morbidity and when considering the overall population recruited in the trial. CONCLUSION In the BETADOSE trial, severe morbidity at discharge of newborns delivered before 32 weeks of gestation was found to be similar among those exposed to 11.4-mg and 22.8-mg antenatal betamethasone. Additional studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Olivier Baud
- Division of Neonatology and Pediatric Intensive Care, Children's University Hospital of Geneva, University of Geneva, Geneva, Switzerland; Université Paris Cité, Inserm U1141, Paris, France.
| | - Loic Sentilhes
- Department of Obstetrics and Gynecology, Bordeaux University Hospital, Bordeaux, France
| | - Moreno Ursino
- Clinical Epidemiology Unit, CIC-EC 1426, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Centre de Recherche des Cordeliers, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France; HeKA, Inria Paris, Paris, France
| | - Muriel Doret-Dion
- Department of Obstetrics and Gynecology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Corinne Alberti
- Clinical Epidemiology Unit, CIC-EC 1426, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, Inserm U1123, ECEVE, Paris, France
| | - Camille Aupiais
- Université Paris Cité, Inserm U1123, ECEVE, Paris, France; Pediatric Emergency Department, Jean Verdier Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne Paris Nord University, Paris, France
| | - Thomas Schmitz
- Department of Obstetrics and Gynecology, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, Centre for Research in Epidemiology and Statistics (CRESS), Inserm, INRAE, Paris, France
| |
Collapse
|
3
|
Chawanpaiboon S, Chukaew R, Pooliam J. A comparison of 2 doses of antenatal dexamethasone for the prevention of respiratory distress syndrome: an open-label, noninferiority, pragmatic randomized trial. Am J Obstet Gynecol 2024; 230:260.e1-260.e19. [PMID: 37442247 DOI: 10.1016/j.ajog.2023.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Antenatal corticosteroids have been used for the prevention of respiratory complications, intraventricular hemorrhage, necrotizing enterocolitis, and other adverse neonatal outcomes for over 50 years, with limited evidence about their optimal doses. Higher steroid doses or frequencies of antenatal corticosteroids in preterm newborns pose adverse effects such as prolonged adrenal suppression, negative effects on fetal programming and metabolism, and increased risks of neurodevelopmental and neuropsychological impairments. Conversely, lower doses of antenatal corticosteroids may be an effective alternative to induce fetal lung maturation with less risk to the fetus. Late preterm births represent the largest population of all preterm neonates, with a respiratory distress syndrome risk of 8.83%. Therefore, determining the optimal antenatal corticosteroid dosage is of particular importance for this population. OBJECTIVE This study aimed to compare the efficacy of 5-mg and 6-mg dexamethasone in preventing neonatal respiratory distress syndrome in women with preterm births at 320 to 366 weeks of gestation. STUDY DESIGN This was an open-label, randomized, controlled, noninferiority trial. Singleton pregnant women (n=370) at 320 to 366 weeks of gestation with spontaneous preterm labor or preterm premature rupture of membranes were enrolled. They were randomly assigned (1:1) to a 5-mg or 6-mg dexamethasone group. Dexamethasone was administered intramuscularly every 12 hours for 4 doses or until delivery. The primary outcome was the reduction in neonatal respiratory distress syndrome cases, whereas the secondary outcomes were any adverse maternal or neonatal events. RESULTS Between December 2020 and April 2022, 370 eligible women, anticipating deliveries within the gestational range of 32 0/7 to 36 6/7 weeks, willingly participated in the study. They were evenly split, with 185 women assigned to the 5-mg group and 185 to the 6-mg group. The study revealed that the demographic profiles of the participants in the 2 groups were remarkably similar, with no statistically significant disparities (P>.05). It is noteworthy that most of these women gave birth after 34 weeks of gestation. Despite a substantial proportion not completing the full course of steroid treatment, the 5-mg dose exhibited noninferiority compared with the 6-mg dose of dexamethasone, as indicated by a modest proportional difference of 0.5% (95% confidence interval, -2.8 to 43.9). Neonatal respiratory distress syndrome occurred in a relatively low percentage of newborns in both groups, affecting 2.2% in the 5-mg group and 1.6% in the 6-mg group. Notably, the risk difference of 0.6% fell comfortably within the predefined noninferiority threshold of 10%. CONCLUSION Our study suggests that a 5-mg dexamethasone dose is noninferior to a standard 6-mg dose in preventing neonatal respiratory distress syndrome in preterm births.
Collapse
Affiliation(s)
- Saifon Chawanpaiboon
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Ronnakorn Chukaew
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Julaporn Pooliam
- Clinical Epidemiological Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
4
|
Du R, Zhao X, Song L, Wang H, Liu D, Wang Q. A physiologically based toxicokinetic model of P-glycoprotein transporter-mediated placenta perfusion of dexamethasone in the pregnant rat. Food Chem Toxicol 2024; 183:114213. [PMID: 38052401 DOI: 10.1016/j.fct.2023.114213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/09/2023] [Accepted: 11/19/2023] [Indexed: 12/07/2023]
Abstract
The present dosage of Dexamethasone (DEX) administered to pregnant women may pose a risk of toxicity to their unborn offspring. We aimed to develop a maternal-fetal physiologically based toxicokinetic (PBTK) model for DEX in pregnant rats, with a specific focus on the role of the P-glycoprotein (P-gp) transporter in placenta perfusion, and finally facilitate the optimization of clinical DEX dosage. We conducted animal experiments to determine DEX concentrations in various rat tissues, and constructed the PBTK model using MATLAB software. Sensitivity analysis was performed to assess input parameters and the model stability, with fold error (FE) values serving as evaluation indices. Our results indicate the successful construction of the PBTK model, with the fitting key parameters such as the absorption rate constant (Ka), intrinsic hepatic clearance (CLh,int) and intrinsic P-gp clearance (CLint,P-gp). The median concentration of DEX in maternal plasma, fetal plasma, fetal lung, and fetal brain were determined, which allowed us to fit the tissue-to-plasma partition coefficients for the fetal lung (Kp,lung,f) and fetal brain (Kp,brain,f). After making adjustments, all calculated FE values were found to be less than 2, demonstrating the acceptability and accuracy of our model's predictions. Our model integrated external literature data and internal animal experimentation to comprehensively evaluate the maternal-fetal PK characteristics of DEX. These findings provide valuable support for the optimization of clinical DEX dosing.
Collapse
Affiliation(s)
- Ruihu Du
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, China
| | - Xiaoqi Zhao
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Ling Song
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China; Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, 100191, China; Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China.
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, Beijing, 100191, China; Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, China.
| |
Collapse
|
5
|
Sultana S, Vogel JP, Oladapo OT. The efficacy of antenatal corticosteroids to improve preterm newborn outcomes in low-resource countries: Are we there yet? BJOG 2023; 130 Suppl 3:84-91. [PMID: 37530472 DOI: 10.1111/1471-0528.17611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/24/2023] [Indexed: 08/03/2023]
Affiliation(s)
- Saima Sultana
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Victoria, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Joshua P Vogel
- Maternal, Child and Adolescent Health Program, Burnet Institute, Melbourne, Victoria, Australia
| | - Olufemi T Oladapo
- UNDP-UNFPA-UNICEF-WHO-World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| |
Collapse
|
6
|
Chawanpaiboon S, Pooliam J, Chuchotiros M. A case-control study on the effects of incomplete, one, and more than one dexamethasone course on acute respiratory problems in preterm neonates born between 28 0 and 36 6 weeks of gestation. BMC Pregnancy Childbirth 2022; 22:880. [PMID: 36443697 PMCID: PMC9703789 DOI: 10.1186/s12884-022-05209-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To compare the effects of an incomplete course and more than 1 course of dexamethasone, relative to a control of a single complete course, on foetal respiratory problems and other adverse outcomes of preterm birth. METHODS This was a retrospective chart review of 1800 women with preterm delivery. Data were collected on newborns whose mothers administered 1 full course of dexamethasone (916/1800; 50.9%), a partial course (716/1800; 39.8%) and more than 1 course (168/1800; 9.3%). Demographic data and adverse maternal and neonatal outcomes were recorded. RESULTS Preterm singleton newborns whose mothers received several steroid hormone courses were significantly more likely to have adverse outcomes than newborns of mothers given 1 course. The negative outcomes were the need for positive pressure ventilation ([aOR] 1.831; 95% CI, (1.185,2.829); P = 0.019), ventilator support ([aOR] 1.843; 95% CI, (1.187,2.861); P = 0.011), and phototherapy ([aOR] 1.997; 95% CI, (1.378,2.895); P < 0.001), transient tachypnoea of the newborn ([aOR] 1.801; 95% CI, (1.261,2.571); P = 0.002), intraventricular haemorrhage ([aOR] 2.215; 95% CI, (1.159, 4.233); P = 0.027), sepsis ([aOR] 1.737; 95% CI, (1.086, 2.777); P = 0.007), and admission to neonatal intensive care ([aOR] 1.822; 95% CI, (1.275,2.604); P = 0.001). In the group of very preterm infants, newborns of mothers administered an incomplete course had developed respiratory distress syndrome (RDS) ([aOR] 3.177; 95% CI, (1.485, 6.795); P = 0.006) and used ventilatory support ([aOR] 3.565; 95% CI, (1.912, 6.650); P < 0.001) more than those of mothers receiving a single course. CONCLUSIONS Preterm singleton newborns whose mothers were given multiple courses of dexamethasone had an increased incidence of RDS and other adverse outcomes than those of mothers receiving a full course. However, very preterm newborns whose mothers were administered 1 full dexamethasone course had a significantly lower incidence of RDS than those whose mothers were given partial courses.
Collapse
Affiliation(s)
- Saifon Chawanpaiboon
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Julaporn Pooliam
- Clinical Epidemiological Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Monsak Chuchotiros
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynaecology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| |
Collapse
|
7
|
Zafran N, Massalha M, Suleiman A, Massalha R, Mahagna L, Weiner SA, Romano S, Shalev E, Salim R. Association between betamethasone levels and respiratory distress syndrome in preterm births: A prospective cohort study. Clin Transl Sci 2022; 15:2528-2537. [PMID: 35923139 PMCID: PMC9579395 DOI: 10.1111/cts.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 01/25/2023] Open
Abstract
The recommended fixed dosage of betamethasone for pregnancies at risk of preterm birth was determined in the 1970s, regardless of gestational age (GA), number of fetuses, and maternal weight. We aimed to examine the association between maternal and neonatal betamethasone serum levels and neonatal respiratory distress syndrome (RDS) and to examine whether levels correlate with maternal weight, GA, or number of fetuses. A prospective study was conducted at a single academic medical center between August 2016 and February 2019. Women received betamethasone and delivered between 28+0 and 34+6 weeks were included. Maternal serum levels (MSLs), and neonatal serum levels (NSLs) of betamethasone at delivery were analyzed using Corticosteroid enzyme-linked immunosorbent assay kit. RDS was diagnosed according to clinical and radiographic findings. We assumed that the sensitivity of NSLs to detect RDS is 95%; hence, 150 neonates were needed (power 80%, alpha 0.05). Overall, 124 women were included; including 96 (77.4%) singletons, 26 (21.0%) twins, and 2 (1.6%) triplets, corresponding to 154 neonates. RDS was diagnosed in 35 neonates (22.7%). After adjusting for GA, time elapsed from the last dose, and number of doses, NSLs were associated with RDS (relative risk: 0.97, 95% confidence interval: 0.94-0.99, p = 0.011). A level of 6.00 ng/ml predicted RDS with a sensitivity of 80.0% and specificity of 64.7%. Adjusted MSLs were not associated with RDS. Both maternal and neonatal serum levels were not associated with the number of fetuses and maternal weight. In conclusion, NSLs are associated with RDS whereas MSLs are not.
Collapse
Affiliation(s)
- Noah Zafran
- Department of Obstetrics and GynecologyEmek Medical CenterAfulaIsrael,The Ruth and Bruce Rappaport Faculty of Medicine, TechnionHaifaIsrael
| | - Manal Massalha
- Department of Obstetrics and GynecologyEmek Medical CenterAfulaIsrael
| | - Abeer Suleiman
- Department of Obstetrics and GynecologyEmek Medical CenterAfulaIsrael
| | | | - Lila Mahagna
- Endocrinology LaboratoryEmek Medical CenterAfulaIsrael
| | | | - Shabtai Romano
- Department of Obstetrics and GynecologyEmek Medical CenterAfulaIsrael,The Ruth and Bruce Rappaport Faculty of Medicine, TechnionHaifaIsrael
| | - Eliezer Shalev
- Department of Obstetrics and GynecologyEmek Medical CenterAfulaIsrael,The Ruth and Bruce Rappaport Faculty of Medicine, TechnionHaifaIsrael
| | - Raed Salim
- Department of Obstetrics and GynecologyEmek Medical CenterAfulaIsrael,The Ruth and Bruce Rappaport Faculty of Medicine, TechnionHaifaIsrael
| |
Collapse
|
8
|
Schmitz T, Doret-Dion M, Sentilhes L, Parant O, Claris O, Renesme L, Abbal J, Girault A, Torchin H, Houllier M, Le Saché N, Vivanti AJ, De Luca D, Winer N, Flamant C, Thuillier C, Boileau P, Blanc J, Brevaut V, Bouet PE, Gascoin G, Beucher G, Datin-Dorriere V, Bounan S, Bolot P, Poncelet C, Alberti C, Ursino M, Aupiais C, Baud O. Neonatal outcomes for women at risk of preterm delivery given half dose versus full dose of antenatal betamethasone: a randomised, multicentre, double-blind, placebo-controlled, non-inferiority trial. Lancet 2022; 400:592-604. [PMID: 35988568 DOI: 10.1016/s0140-6736(22)01535-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 07/10/2022] [Accepted: 08/04/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Antenatal betamethasone is recommended before preterm delivery to accelerate fetal lung maturation. However, reports of growth and neurodevelopmental dose-related side-effects suggest that the current dose (12 mg plus 12 mg, 24 h apart) might be too high. We therefore investigated whether a half dose would be non-inferior to the current full dose for preventing respiratory distress syndrome. METHODS We designed a randomised, multicentre, double-blind, placebo-controlled, non-inferiority trial in 37 level 3 referral perinatal centres in France. Eligible participants were pregnant women aged 18 years or older with a singleton fetus at risk of preterm delivery and already treated with the first injection of antenatal betamethasone (11·4 mg) before 32 weeks' gestation. We used a computer-generated code producing permuted blocks of varying sizes to randomly assign (1:1) women to receive either a placebo (half-dose group) or a second 11·4 mg betamethasone injection (full-dose group) 24 h later. Randomisation was stratified by gestational age (before or after 28 weeks). Participants, clinicians, and study staff were masked to the treatment allocation. The primary outcome was the need for exogenous intratracheal surfactant within 48 h after birth. Non-inferiority would be shown if the higher limit of the 95% CI for the between-group difference between the half-dose and full-dose groups in the primary endpoint was less than 4 percentage points (corresponding to a maximum relative risk of 1·20). Four interim analyses monitoring the primary and the secondary safety outcomes were done during the study period, using a sequential data analysis method that provided futility and non-inferiority stopping rules and checked for type I and II errors. Interim analyses were done in the intention-to-treat population. This trial was registered with ClinicalTrials.gov, NCT02897076. FINDINGS Between Jan 2, 2017, and Oct 9, 2019, 3244 women were randomly assigned to the half-dose (n=1620 [49·9%]) or the full-dose group (n=1624 [50·1%]); 48 women withdrew consent, 30 fetuses were stillborn, 16 neonates were lost to follow-up, and 9 neonates died before evaluation, so that 3141 neonates remained for analysis. In the intention-to-treat analysis, the primary outcome occurred in 313 (20·0%) of 1567 neonates in the half-dose group and 276 (17·5%) of 1574 neonates in the full-dose group (risk difference 2·4%, 95% CI -0·3 to 5·2); thus non-inferiority was not shown. The per-protocol analysis also did not show non-inferiority (risk difference 2·2%, 95% CI -0·6 to 5·1). No between-group differences appeared in the rates of neonatal death, grade 3-4 intraventricular haemorrhage, stage ≥2 necrotising enterocolitis, severe retinopathy of prematurity, or bronchopulmonary dysplasia. INTERPRETATION Because non-inferiority of the half-dose compared with the full-dose regimen was not shown, our results do not support practice changes towards antenatal betamethasone dose reduction. FUNDING French Ministry of Health.
Collapse
Affiliation(s)
- Thomas Schmitz
- Department of Obstetrics and Gynaecology, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, Centre for Research in Epidemiology and Statistics, INSERM U1153, INRA, Paris, France.
| | - Muriel Doret-Dion
- Department of Obstetrics and Gynaecology, Hospital Femme-Mère-Enfant, Hospices Civils de Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Loic Sentilhes
- Department of Obstetrics and Gynaecology, Bordeaux University Hospital, Bordeaux, France
| | - Olivier Parant
- Department of Obstetrics and Gynaecology, Toulouse University Hospital, Toulouse, France
| | - Olivier Claris
- Department of Neonatology, Hospital Femme-Mère-Enfant, Hospices Civils de Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Laurent Renesme
- Department of Neonatology, Bordeaux University Hospital, Bordeaux, France
| | - Julie Abbal
- Department of Neonatology, Toulouse University Hospital, Toulouse, France
| | - Aude Girault
- Université Paris Cité, Centre for Research in Epidemiology and Statistics, INSERM U1153, INRA, Paris, France; MaternitéPort-Royal, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Héloïse Torchin
- Université Paris Cité, Centre for Research in Epidemiology and Statistics, INSERM U1153, INRA, Paris, France; Department of Neonatology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marie Houllier
- Department of Obstetrics and Gynaecology, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nolwenn Le Saché
- Department of Neonatology, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alexandre J Vivanti
- Department of Obstetrics and Gynaecology, Antoine Béclère Hospital, Assistance Publique-Hôpitaux de Paris, Paris, Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France
| | - Daniele De Luca
- Department of Neonatology, Antoine Béclère Hospital, Assistance Publique-Hôpitaux de Paris, Paris, Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France
| | - Norbert Winer
- Department of Obstetrics and Gynaecology, University Medical Centre of Nantes, Centre d'Investigation Clinique CIC Mere enfant, Nantes, France; National Institute of Agricultural Research, UMR 1280, Physiology of Nutritional Adaptations, University of Nantes, IMAD and CRNH-Ouest, Nantes, France
| | - Cyril Flamant
- Department of Neonatology, Nantes University Hospital, Nantes, France
| | - Claire Thuillier
- Department of Obstetrics and Gynaecology, Poissy Hospital Centre, Poissy, France
| | - Pascal Boileau
- Department of Neonatology, Poissy Hospital Centre, Poissy, France
| | - Julie Blanc
- Department of Obstetrics and Gynaecology, Marseille Nord University Hospital, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Véronique Brevaut
- Department of Neonatology, Marseille Nord University Hospital, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Pierre-Emmanuel Bouet
- Department of Obstetrics and Gynaecology, Angers University Hospital, Angers, France
| | - Géraldine Gascoin
- Department of Neonatology, Angers University Hospital, Angers, France
| | - Gaël Beucher
- Department of Obstetrics and Gynaecology, Caen University Hospital, Caen, France
| | | | - Stéphane Bounan
- Department of Obstetrics and Gynaecology, Saint-Denis Hospital, Saint-Denis, France
| | - Pascal Bolot
- Department of Neonatology, Saint-Denis Hospital, Saint-Denis, France
| | - Christophe Poncelet
- Department of Obstetrics and Gynaecology, Pontoise Hospital, Pontoise, France
| | - Corinne Alberti
- Clinical Epidemiology Unit, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, INSERM U1123, ECEVE, Paris, France
| | - Moreno Ursino
- Clinical Epidemiology Unit, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Centre de Recherche des Cordeliers, Université Paris Cité, INSERM U1138, Inria, HeKA, Paris, France
| | - Camille Aupiais
- Université Paris Cité, INSERM U1123, ECEVE, Paris, France; Paediatric Emergency Department, Jean Verdier Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne Paris Nord University, Paris, France
| | - Olivier Baud
- Université Paris Cité, INSERM U1141, Paris, France; Division of Neonatology and Paediatric Intensive Care, Children's University Hospital of Geneva and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
9
|
Prenatal or postnatal corticosteroids favor clinical, respiratory, metabolic outcomes and oxidative balance of preterm lambs corticotherapy for premature neonatal lambs. Theriogenology 2022; 182:129-137. [DOI: 10.1016/j.theriogenology.2022.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022]
|
10
|
Foissac F, Zheng Y, Hirt D, Lui G, Bouazza N, Ville Y, Goffinet F, Rozenberg P, Kayem G, Mandelbrot L, Benaboud S, Jarreau PH, Tréluyer JM. Maternal Betamethasone for Prevention of Respiratory Distress Syndrome in Neonates: Population Pharmacokinetic and Pharmacodynamic Approach. Clin Pharmacol Ther 2020; 108:1026-1035. [PMID: 32394434 DOI: 10.1002/cpt.1887] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/25/2020] [Indexed: 02/02/2023]
Abstract
Despite antenatal corticosteroids therapy, respiratory distress syndrome (RDS) is still a leading cause of neonatal morbidity and mortality in premature newborns. To date, the relationship between in utero fetal drug exposure and occurrence of RDS remains poorly evaluated. This study aims to describe the pharmacokinetics of betamethasone in pregnant women and to evaluate the transplacental drug transfer and administration scheme for the prevention of RDS. Pregnant women > 27 weeks' gestation and who received at least a single dose of betamethasone for prevention of RDS were enrolled. Maternal, cord blood, and amniotic fluid betamethasone time-courses were analyzed using the Monolix software. A total of 220 maternal blood, 56 cord blood, and 26 amniotic fluid samples were described by a two-compartment model with two effect compartments linked by rate transfer constants. Apparent clearances and volumes of distribution parameters were allometrically scaled for a 70 kg third trimester pregnant woman. The impact of a twin pregnancy was found to increase maternal clearance by 28%. Using a fetal-to-mother exposure ratio, the median (95% confidence interval (CI)) transplacental transfer of betamethasone was estimated to 35% (95% CI 0.11-0.67). After adjustment for gestational age and twin pregnancy, RDS was found to be associated to the time spent in utero below quantifiable concentrations (i.e., < 1 ng/mL): odds ratio of 1.10 (95% CI 1.01-1.19) per day increase (P < 0.05). Trying to take into account both efficacy and safety, we simulated different dosing schemes in order to maintain a maximum of fetuses above 1 ng/mL without exceeding the total standard dose.
Collapse
Affiliation(s)
- Frantz Foissac
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,URC/CIC Paris Descartes Necker Cochin, Necker-Enfants Malades Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | - Yi Zheng
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,Clinical Pharmacology Department, AP-HP Paris Centre Hospital Group, Paris, France
| | - Déborah Hirt
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,Clinical Pharmacology Department, AP-HP Paris Centre Hospital Group, Paris, France
| | - Gabrielle Lui
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,Clinical Pharmacology Department, AP-HP Paris Centre Hospital Group, Paris, France
| | - Naïm Bouazza
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,URC/CIC Paris Descartes Necker Cochin, Necker-Enfants Malades Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | - Yves Ville
- Maternity, Hôpital Necker - Enfants Malades, AP-HP, Paris, France.,EA 7328, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - François Goffinet
- Port-Royal Maternity Unit, Cochin Hospital, AP-HP, Paris, France.,Obstetrical, Perinatal, and Pediatric Epidemiology Team and Biostatistics Sorbonne Paris Cité Research Center (U1153), INSERM and Université Paris Descartes, Paris, France
| | - Patrick Rozenberg
- Department of Gynecology and Obstetrics, Poissy-Saint Germain Hospital, Poissy, France.,EA 7285, Paris Saclay University, Montigny-Le-Bretonneux, France
| | - Gilles Kayem
- Obstetrical, Perinatal, and Pediatric Epidemiology Team and Biostatistics Sorbonne Paris Cité Research Center (U1153), INSERM and Université Paris Descartes, Paris, France.,Department of Obstetrics and Gynecology, Trousseau Hospital, AP-HP, Paris, France
| | - Laurent Mandelbrot
- Department of Obstetrics and Gynecology, Louis Mourier Hospital, AP-HP, Colombes, France.,INSERM, IAME, UMR 1137, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sihem Benaboud
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,Clinical Pharmacology Department, AP-HP Paris Centre Hospital Group, Paris, France
| | - Pierre-Henri Jarreau
- Université de Paris, Neonatal Intensive Care Unit of Port-Royal, Paris Centre University Hospitals, APHP, Paris, France.,Université de Paris, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, INSERM, INRA, Paris, France
| | - Jean-Marc Tréluyer
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université de Paris, Paris, France.,URC/CIC Paris Descartes Necker Cochin, Necker-Enfants Malades Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France.,Clinical Pharmacology Department, AP-HP Paris Centre Hospital Group, Paris, France
| |
Collapse
|
11
|
Morphometric and functional pulmonary changes of premature neonatal puppies after antenatal corticoid therapy. Theriogenology 2020; 153:19-26. [PMID: 32417607 DOI: 10.1016/j.theriogenology.2020.05.005] [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: 08/12/2019] [Revised: 10/24/2019] [Accepted: 05/04/2020] [Indexed: 11/22/2022]
Abstract
Among prematurity complications, the most important disorder is structural immaturity and inadequate production of pulmonary surfactant. Betamethasone is the drug of choice to artificially improve pulmonary capacity, thus we aimed to verify the effect of prenatal maternal treatment on lung development of premature puppies. Pregnant bitches were allocated in Term Group (n = 7), Preterm-Treated Group (interrupted pregnancies with maternal administration of betamethasone; n = 7), Preterm-Control Group (untreated interrupted pregnancies; n = 7), Extremely-Preterm Group (interrupted pregnancies at 55d; n = 6). Puppies were subjected to chest radiographic at birth, morphometric description of pulmonary structures and immunohistochemical analysis of surfactant protein B, proliferating cell nuclear antigen and cytokeratin were performed. In Preterm-Treated Group it was possible to more clearly identify cardiac silhouette and lung parenchyma by X-Ray. Saccular formation was higher in Preterm Groups, while Term Group had higher subsaccular development. Lung septation was higher in Treated and Term Groups. Term Group had higher number of cells marked for SP-B, whereas higher proliferation was observed in Extreme-Preterm and Preterm-Control Groups. Preterm Treated and Term Groups had higher tissue differentiation. In conclusion, antenatal maternal corticotherapy in dogs acted by increasing lung morphology and development of areas of gas exchange, regulate metabolism of pulmonary fluids rather than stimulate surfactant production.
Collapse
|
12
|
Schmitz T, Alberti C, Ursino M, Baud O, Aupiais C. Full versus half dose of antenatal betamethasone to prevent severe neonatal respiratory distress syndrome associated with preterm birth: study protocol for a randomised, multicenter, double blind, placebo-controlled, non-inferiority trial (BETADOSE). BMC Pregnancy Childbirth 2019; 19:67. [PMID: 30755164 PMCID: PMC6373166 DOI: 10.1186/s12884-019-2206-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/28/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although antenatal betamethasone is recommended worldwide for women at risk of preterm delivery, concerns persist regarding the long-term effects associated with this treatment. Indeed, adverse events, mainly dose-related, have been reported. The current recommended dose of antenatal betamethasone directly derives from sheep experiments performed in the late 60's and has not been challenged in 45 years. Therefore, randomized trials evaluating novel dose regimens are urgently needed. METHODS A randomised, double blind, placebo-controlled, non-inferiority trial will be performed in 37 French level 3 maternity units. Women with a singleton pregnancy at risk of preterm delivery before 32 weeks of gestation having already received a first 11.4 mg injection of betamethasone will be randomised to receive either a second injection of 11.4 mg betamethasone (full dose arm) or placebo (half dose arm) administered intramuscularly 24 h after the first injection. The primary binary outcome will be the occurrence of severe respiratory distress syndrome (RDS), defined as the need for exogenous intra-tracheal surfactant in the first 48 h of life. Considering that 20% of the pregnant women receiving the full dose regimen would have a neonate with severe RDS, 1571 patients in each treatment group are required to show that the half dose regimen is not inferior to the full dose, that is the difference in severe RDS rate do not exceed 4% (corresponding to a Relative Risk of 20%), with a 1-sided 2.5% type-1 error and a 80% power. Interim analyses will be done after every 300 neonates who reach the primary outcome on the basis of intention-to-treat, using a group-sequential non-inferiority design. DISCUSSION If the 50% reduced antenatal betamethasone dose is shown to be non-inferior to the full dose to prevent severe RDS associated with preterm birth, then it should be used consistently in women at risk of preterm delivery and would be of great importance to their children. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT 02897076 (registration date 09/13/2016).
Collapse
Affiliation(s)
- Thomas Schmitz
- Service de Gynécologie Obstétrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, 48 boulevard Sérurier, 75019 Paris, France
- Université Paris Diderot, Site Villemin, 10 avenue de Verdun, 75010 Paris, France
- Inserm, U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Research Center, Obstetrical, Perinatal and Pediatric Epidemiology Team, 53 avenue de l’observatoire, 75014 Paris, France
| | - Corinne Alberti
- Université Paris Diderot, Site Villemin, 10 avenue de Verdun, 75010 Paris, France
- Unité d’épidémiologie clinique, CIC-EC 1426, Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
- Inserm, U1123, ECEVE, 10 avenue de Verdun, 75010 Paris, France
| | - Moreno Ursino
- Inserm, U1138, Equipe 22, Sorbonne Université, Université Paris Descartes, 75006 Paris, France
| | - Olivier Baud
- Service de néonatalogie, Hôpitaux universitaires de Genève, 32 boulevard de la Cluse, 1205 Genève, Switzerland
- Inserm, U1141, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
| | - Camille Aupiais
- Université Paris Diderot, Site Villemin, 10 avenue de Verdun, 75010 Paris, France
- Inserm, U1123, ECEVE, 10 avenue de Verdun, 75010 Paris, France
- Inserm, U1138, Equipe 22, Sorbonne Université, Université Paris Descartes, 75006 Paris, France
- Service d’Accueil des Urgences Pédiatriques, Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
| | - for the BETADOSE study group and the GROG (Groupe de Recherche en Gynécologie Obstétrique)
- Service de Gynécologie Obstétrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, 48 boulevard Sérurier, 75019 Paris, France
- Université Paris Diderot, Site Villemin, 10 avenue de Verdun, 75010 Paris, France
- Inserm, U1153, Epidemiology and Biostatistics Sorbonne Paris Cité Research Center, Obstetrical, Perinatal and Pediatric Epidemiology Team, 53 avenue de l’observatoire, 75014 Paris, France
- Unité d’épidémiologie clinique, CIC-EC 1426, Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
- Inserm, U1123, ECEVE, 10 avenue de Verdun, 75010 Paris, France
- Inserm, U1138, Equipe 22, Sorbonne Université, Université Paris Descartes, 75006 Paris, France
- Service de néonatalogie, Hôpitaux universitaires de Genève, 32 boulevard de la Cluse, 1205 Genève, Switzerland
- Inserm, U1141, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
- Service d’Accueil des Urgences Pédiatriques, Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, 48 boulevard Sérurier, 75019 Paris, France
| |
Collapse
|
13
|
Mottet N, Aubry S, Vidal C, Boiteux G, Metz JP, Riethmuller D, Pazart L, Ramanah R. Feasibility of 2-D ultrasound shear wave elastography of fetal lungs in case of threatened preterm labour: a study protocol. BMJ Open 2017; 7:e018130. [PMID: 29282263 PMCID: PMC5770838 DOI: 10.1136/bmjopen-2017-018130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION 2-D ultrasound shear wave elastography (SWE) could be considered as a new noninvasive tool for monitoring fetal lung development based on evaluation of mechanical properties during pregnancy. Interesting results are available concerning the use of SWE on developing organs, especially on premature infants and animal models. The main objective in this study is to evaluate the feasibility of 2-D SWE in human fetal lungs between 24 and 34 weeks of gestation (WG). The secondary objective is to modellise fetal lung-to-liver elastography ratio (LLE ratio) and to assess variations between normal lung and lung surfactant-enriched after a corticosteroids course indicated for a threatened preterm labour (TPL). METHODS/DESIGN A prospective case-control study will be performed between 24 and 34 WG. Fetal lungs and liver will be explored by SWE into two groups: fetuses of women with an uncomplicated pregnancy (control group) and fetuses of women with a TPL requiring administration of corticosteroids (cases group). LLE ratio will be defined as the value of the lung elasticity divided by the value of the liver elasticity.Primary judgement criterion is the value of elasticity modulus expressed in kilopascal. Lungs and liver will be explored through three measurements to define the most reproducible regions with the lowest intra- and inter-observer variability. Feasibility will be evaluated by assessing the number of examinations performed and the number of examinations with interpretable results. Intra- and inter-observer reproducibility will be evaluated by means of the intra-class correlation coefficient. ETHICS AND DISSEMINATION Approval of the study protocol was obtained from the human ethical research committee (Comité de Protection des Personnes EST II, process number 15/494) and the French National Agency for Medicines and Health Products Safety (process number 2015-A01575-44). All participants will sign a statement of informed consent. TRIAL REGISTRATION NUMBER NCT02870608; Recruiting.
Collapse
Affiliation(s)
- Nicolas Mottet
- Department of Obstetrics and Gynaecology, Pôle Mère-Femme, University Hospital of Besancon, University of Franche-Comte, Besançon, France
- Nanomedecine Laboratory, INSERM EA4662, University of Franche-Comte, Besancon, France
| | - Sébastien Aubry
- Nanomedecine Laboratory, INSERM EA4662, University of Franche-Comte, Besancon, France
- Department of Musculoskeletal Imaging, University Hospital of Besancon, Besancon, France
| | - Chrystelle Vidal
- Centre d’investigation Clinique-Innovation Technologique, INSERM, University Hospital of Besançon, Besançon, France
| | - Guillaume Boiteux
- Centre d’investigation Clinique-Innovation Technologique, INSERM, University Hospital of Besançon, Besançon, France
| | - Jean-Patrick Metz
- Department of Obstetrics and Gynaecology, Pôle Mère-Femme, University Hospital of Besancon, University of Franche-Comte, Besançon, France
| | - Didier Riethmuller
- Department of Obstetrics and Gynaecology, Pôle Mère-Femme, University Hospital of Besancon, University of Franche-Comte, Besançon, France
| | - Lionel Pazart
- Centre d’investigation Clinique-Innovation Technologique, INSERM, University Hospital of Besançon, Besançon, France
| | - Rajeev Ramanah
- Department of Obstetrics and Gynaecology, Pôle Mère-Femme, University Hospital of Besancon, University of Franche-Comte, Besançon, France
- Nanomedecine Laboratory, INSERM EA4662, University of Franche-Comte, Besancon, France
| |
Collapse
|
14
|
Influence of prenatal maternal corticosteroid therapy on clinical and metabolic features and pulmonary function of preterm newborn puppies. Theriogenology 2017; 97:179-185. [DOI: 10.1016/j.theriogenology.2017.04.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/24/2017] [Accepted: 04/24/2017] [Indexed: 11/17/2022]
|
15
|
Kemp MW, Saito M, Usuda H, Molloy TJ, Miura Y, Sato S, Watanabe S, Clarke M, Fossler M, Scmidt A, Kallapur SG, Kramer BW, Newnham JP, Jobe AH. Maternofetal pharmacokinetics and fetal lung responses in chronically catheterized sheep receiving constant, low-dose infusions of betamethasone phosphate. Am J Obstet Gynecol 2016; 215:775.e1-775.e12. [PMID: 27555319 DOI: 10.1016/j.ajog.2016.08.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/24/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Antenatal steroids are standard of care for cases of anticipated preterm labor to improve neonatal outcomes. However, steroids are potent drugs, and their use in pregnancy remains largely unoptimized. OBJECTIVE The objective of the study was to measure the maternofetal pharmacokinetics of constant, low-dose intravenous betamethasone phosphate infusions and correlate these data with the transcriptional effect exerted by subclinical betamethasone exposures on the ovine fetal lung. STUDY DESIGN Thirty-two ewes carrying a single fetus had surgery to catheterize fetal and maternal jugular veins at 116 days of gestation (term, 150 days). Animals were recovered for 2 days and then were randomized to receive 2 sequential maternal intravenous infusions of either (n = 4/group) of the following: 1) saline, 0.125, 0.04, or 0.0125 mg/kg betamethasone phosphate over 3 hours; or 2) saline, 0.25, 0.08, or 0.025 mg/kg betamethasone phosphate over 12 hours. Each infusion was separated by 2 days. Fetal lung tissue was collected for analysis using quantitative polymerase chain reaction and an ovine-specific microarray. Plasma betamethasone levels from time-course catheter samples were determined by mass spectrometry. Data were assessed for distribution, variance, and tested by an analysis of variance. RESULTS Betamethasone was detectable (>1 ng/mL) in fetal plasma only in animals randomized to 0.125 mg/kg 3 hour or 0.250 mg/kg 12 hour infusions. Fetal betamethasone half-lives were 1.7-2.8 times greater than maternal values. At maximum concentration, fetal plasma betamethasone levels were approximately 10% of maternal levels. Compared with saline control, all animals, other than those receiving 0.0125 mg/kg 3 hour betamethasone phosphate infusions, had evidence of dose-dependent glucocorticoid transcriptional responses in the fetal lung. CONCLUSION Constant maternal betamethasone infusions delivering substantially lower fetal and maternal betamethasone maximal concentrations than those achieved with current clinical treatment protocols were associated with dose-dependent changes in glucocorticoid-response markers in the fetal lung. Further studies to determine the minimally efficacious dose of steroids for improving outcomes in preterm infants should be viewed as a priority.
Collapse
|
16
|
Perez M, Wisniewska K, Lee KJ, Cardona HJ, Taylor JM, Farrow KN. Dose-dependent effects of glucocorticoids on pulmonary vascular development in a murine model of hyperoxic lung injury. Pediatr Res 2016; 79:759-65. [PMID: 26756781 PMCID: PMC4853243 DOI: 10.1038/pr.2016.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/31/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Exposure of neonatal mice to hyperoxia results in pulmonary vascular remodeling and aberrant phosphodiesterase type 5 (PDE5) signaling. Although glucocorticoids are frequently utilized in the NICU, little is known about their effects on the developing pulmonary vasculature and on PDE5. We sought to determine the effects of hydrocortisone (HC) on pulmonary vascular development and on PDE5 in a neonatal mouse model of hyperoxic lung injury. METHODS C57BL/6 mice were placed in 21% O2 or 75% O2 within 24 h of birth and received HC (1, 5, or 10 mg/kg subcutaneously every other day) or vehicle. At 14 d, right ventricular hypertrophy (RVH), medial wall thickness (MWT), lung morphometry, and pulmonary artery (PA) PDE5 activity were assessed. PDE5 activity was measured in isolated pulmonary artery smooth muscle cells exposed to 21 or 95% O2 ± 100 nmol/l HC for 24 h. RESULTS Hyperoxia resulted in alveolar simplification, RVH, increased MWT, and increased PA PDE5 activity. HC decreased hyperoxia-induced RVH and attenuated MWT. HC had dose-dependent effects on alveolar simplification. HC decreased hyperoxia-induced PDE5 activity both in vivo and in vitro. CONCLUSIONS HC decreases hyperoxia-induced pulmonary vascular remodeling and attenuates PDE5 activity. These findings suggest that HC may protect against hyperoxic injury in the developing pulmonary vasculature.
Collapse
Affiliation(s)
- Marta Perez
- Department of Pediatrics, Northwestern University, Chicago, IL, USA,Corresponding author: Marta Perez, MD, Assistant Professor of Pediatrics, Northwestern University Feinberg School of Medicine, 310 E. Superior St., Morton 4-410, Chicago, IL 60611, Phone: 312-503-2385, Fax: 312-503-1181,
| | | | - Keng Jin Lee
- Department of Pediatrics, Northwestern University, Chicago, IL, USA
| | | | - Joann M. Taylor
- Department of Pediatrics, Northwestern University, Chicago, IL, USA
| | | |
Collapse
|
17
|
Salim R, Suleiman A, Colodner R, Nachum Z, Goldstein LH, Shalev E. Measurement of betamethasone concentration in maternal serum treated for fetal lung maturity; Is it feasible? Reprod Biol Endocrinol 2016; 14:7. [PMID: 26860904 PMCID: PMC4748478 DOI: 10.1186/s12958-016-0142-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The association between maternal serum concentration of betamethasone given for fetal lung maturity and perinatal outcome has not been investigated. This may be due to an absence of a reliable method for measuring serum betamethasone concentrations. We aimed in the current study to assess the feasibility of a specific ELISA kit to measure the concentrations of betamethasone in maternal serum and to examine the trend of sequential measurements after a course of betamethasone for fetal lung maturity. METHODS Pregnant women at risk for preterm birth who received betamethasone between 24 and 34 weeks of gestation were prospectively included. Serum concentrations were determined before administering betamethasone (baseline), and 36 hours, 48 hours, 72 hours, and 5 to 7 days after the 1(st) dose. Betamethasone concentration in samples was determined using Corticosteroid ELISA kit. The Friedman test was used to test whether there were significant differences between the measurements. RESULTS Five singleton pregnancies were included. Using the ELISA kit, betamethasone concentration in maternal serum samples was obtained for all women. Among the five measurements performed, the concentration was highest at 36 hours after the 1(st) dose and close to baseline at the 5(th) measurement performed after 5 to 7 days (p < 0.05). Serum concentration varied at each time point between the five women but similar trend was observed. CONCLUSION Betamethasone concentration is measurable in the serum of pregnant women with this ELISA kit.
Collapse
Affiliation(s)
- Raed Salim
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, 18101, Israel.
- Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | - Abeer Suleiman
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, 18101, Israel.
| | - Raul Colodner
- Clinical Microbiology Laboratory, Emek Medical Center, Afula, Israel.
| | - Zohar Nachum
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, 18101, Israel.
- Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | - Lee H Goldstein
- Rappaport Faculty of Medicine, Technion, Haifa, Israel.
- Department of Internal Medicine C, Clinical Pharmacology and toxicology Unit, Emek Medical Center, Afula, Israel.
| | - Eliezer Shalev
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, 18101, Israel.
- Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
18
|
Fetal and neonatal outcomes after term and preterm delivery following betamethasone administration. Int J Gynaecol Obstet 2015; 130:64-9. [DOI: 10.1016/j.ijgo.2015.01.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/05/2015] [Accepted: 03/18/2015] [Indexed: 11/20/2022]
|
19
|
Abstract
Since their introduction more than forty years ago, antenatal glucocorticoids have become a cornerstone in the management of preterm birth and have been responsible for substantial reductions in neonatal mortality and morbidity. Clinical trials conducted over the past decade have shown that these benefits may be increased further through administration of repeat doses of antenatal glucocorticoids in women at ongoing risk of preterm and in those undergoing elective cesarean at term. At the same time, a growing body of experimental animal evidence and observational data in humans has linked fetal overexposure to maternal glucocorticoids with increased risk of cardiovascular, metabolic and other disorders in later life. Despite these concerns, and somewhat surprisingly, there has been little evidence to date from randomized trials of longer-term harm from clinical doses of synthetic glucocorticoids. However, with wider clinical application of antenatal glucocorticoid therapy there has been greater need to consider the potential for later adverse effects. This paper reviews current evidence for the short- and long-term health effects of antenatal glucocorticoids and discusses the apparent discrepancy between data from randomized clinical trials and other studies.
Collapse
|
20
|
Braun T, Challis JR, Newnham JP, Sloboda DM. Early-life glucocorticoid exposure: the hypothalamic-pituitary-adrenal axis, placental function, and long-term disease risk. Endocr Rev 2013; 34:885-916. [PMID: 23970762 DOI: 10.1210/er.2013-1012] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
An adverse early-life environment is associated with long-term disease consequences. Adversity early in life is hypothesized to elicit developmental adaptations that serve to improve fetal and postnatal survival and prepare the organism for a particular range of postnatal environments. These processes, although adaptive in their nature, may later prove to be maladaptive or disadvantageous if the prenatal and postnatal environments are widely discrepant. The exposure of the fetus to elevated levels of either endogenous or synthetic glucocorticoids is one model of early-life adversity that contributes substantially to the propensity of developing disease. Moreover, early-life glucocorticoid exposure has direct clinical relevance because synthetic glucocorticoids are routinely used in the management of women at risk of early preterm birth. In this regard, reports of adverse events in human newborns have raised concerns about the safety of glucocorticoid treatment; synthetic glucocorticoids have detrimental effects on fetal growth and development, childhood cognition, and long-term behavioral outcomes. Experimental evidence supports a link between prenatal exposure to synthetic glucocorticoids and alterations in fetal development and changes in placental function, and many of these alterations appear to be permanent. Because the placenta is the conduit between the maternal and fetal environments, it is likely that placental function plays a key role in mediating effects of fetal glucocorticoid exposure on hypothalamic-pituitary-adrenal axis development and long-term disease risk. Here we review recent insights into how the placenta responds to changes in the intrauterine glucocorticoid environment and discuss possible mechanisms by which the placenta mediates fetal hypothalamic-pituitary-adrenal development, metabolism, cardiovascular function, and reproduction.
Collapse
Affiliation(s)
- Thorsten Braun
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, 1280 Main Street West, HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1.
| | | | | | | |
Collapse
|
21
|
Elevated glucocorticoids during ovine pregnancy increase appetite and produce glucose dysregulation and adiposity in their granddaughters in response to ad libitum feeding at 1 year of age. Am J Obstet Gynecol 2013; 209:353.e1-9. [PMID: 23727517 DOI: 10.1016/j.ajog.2013.05.051] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 05/03/2013] [Accepted: 05/29/2013] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Synthetic glucocorticoids (sGCs) are administered to women threatening preterm labor. We have shown multigenerational endocrine and metabolic effects of fetal sGC exposure. We hypothesized that sGC exposure would alter the second filial generation (F2) offspring neonatal leptin peak that controls development of appetitive behavior with metabolic consequences. STUDY DESIGN F0 nulliparous ewes were bred to a single ram. Beginning at day 103 of gestation (term 150 days), dexamethasone (DEX) ewes received 4 injections of 2 mg DEX intramuscularly, 12 hours apart. Control ewes received saline. Ewes lambed naturally. At 22 months of age, F1 offspring were mated to produce F2 offspring. At 10 months of age, F2 female offspring were placed on an ad libitum feeding challenge for 12 weeks. RESULTS DEX F2 female offspring did not show a postnatal leptin peak and their plasma cortisol concentration was elevated in the first days of life. During the feeding challenge, DEX F2 offspring consumed 10% more feed and gained 20% more weight compared with control F2 offspring. At the end of the feeding challenge, DEX F2 offspring had greater adiposity compared with control F2 offspring. F2 sGC offspring showed impaired insulin secretion in response to an intravenous glucose tolerance test. CONCLUSION sGC administration to F0 mothers eliminates the neonatal leptin peak in F2 female offspring potentially by inhibition caused by elevated cortisol in the DEX F2 offspring. F2 offspring showed increased appetite, weight gain, and adiposity during an ad libitum feeding challenge accompanied by decreased insulin response to an intravenous glucose tolerance test.
Collapse
|
22
|
Niu Y, Herrera EA, Evans RD, Giussani DA. Antioxidant treatment improves neonatal survival and prevents impaired cardiac function at adulthood following neonatal glucocorticoid therapy. J Physiol 2013; 591:5083-93. [PMID: 23940378 DOI: 10.1113/jphysiol.2013.258210] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Glucocorticoids are widely used to treat chronic lung disease in premature infants but their longer-term adverse effects on the cardiovascular system raise concerns. We reported that neonatal dexamethasone treatment in rats induced in the short term molecular indices of cardiac oxidative stress and cardiovascular tissue remodelling at weaning, and that neonatal combined antioxidant and dexamethasone treatment was protective at this time. In this study, we investigated whether such effects of neonatal dexamethasone have adverse consequences for NO bioavailability and cardiovascular function at adulthood, and whether neonatal combined antioxidant and dexamethasone treatment is protective in the adult. Newborn rat pups received daily i.p. injections of a human-relevant tapering dose of dexamethasone (D; n = 8; 0.5, 0.3, 0.1 μg g(-1)) or D with vitamins C and E (DCE; n = 8; 200 and 100 mg kg(-1), respectively) on postnatal days 1-3 (P1-3); vitamins were continued from P4 to P6. Controls received equal volumes of vehicle from P1 to P6 (C; n = 8). A fourth group received vitamins alone (CCE; n = 8). At P100, plasma NO metabolites (NOx) was measured and isolated hearts were assessed under both Working and Langendorff preparations. Relative to controls, neonatal dexamethasone therapy increased mortality by 18% (P < 0.05). Surviving D pups at adulthood had lower plasma NOx concentrations (10.6 ± 0.8 vs. 28.0 ± 1.5 μM), an increased relative left ventricular (LV) mass (70 ± 2 vs. 63 ± 1%), enhanced LV end-diastolic pressure (14 ± 2 vs. 8 ± 1 mmHg) and these hearts failed to adapt output with increased preload (cardiac output: 2.9 ± 2.0 vs. 10.6 ± 1.2 ml min(-1)) or afterload (cardiac output: -5.3 ± 2.0 vs.1.4 ± 1.2 ml min(-1)); all P < 0.05. Combined neonatal dexamethasone with antioxidant vitamins improved postnatal survival, restored plasma NOx and protected against cardiac dysfunction at adulthood. In conclusion, neonatal dexamethasone therapy promotes cardiac dysfunction at adulthood. Combined neonatal treatment with antioxidant vitamins is an effective intervention.
Collapse
Affiliation(s)
- Youguo Niu
- D. A. Giussani: Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | | | | | | |
Collapse
|
23
|
Boudaa N, Samson N, Carrière V, Germim PS, Pasquier JC, Bairam A, Praud JP. Effects of caffeine and/or nasal CPAP treatment on laryngeal chemoreflexes in preterm lambs. J Appl Physiol (1985) 2013; 114:637-46. [PMID: 23305977 DOI: 10.1152/japplphysiol.00599.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Current knowledge suggests that laryngeal chemoreflexes (LCR) are involved in the occurrence of certain neonatal apneas/bradycardias, especially in the preterm newborn. While caffeine and/or nasal continuous positive airway pressure (nCPAP) are the most frequent options used for treating apneas in preterm newborns, their effects on LCR-related apneas/bradycardias are virtually unknown. The aim of the present study was to test the hypothesis that caffeine and/or nCPAP decreases LCR-related cardiorespiratory inhibition in a preterm ovine model. Seven preterm lambs were born vaginally on gestational day 133 (normal gestation: 147 days) after intramuscular injections of betamethasone and mifepristone. Five days after birth, a chronic surgical instrumentation was performed to record states of alertness, electrocardiogram, systemic arterial pressure, and electromyographic activity of a laryngeal constrictor muscle, as well as to insert a transcutaneous supraglottal catheter. LCR were induced in quiet sleep under four conditions: 1) control (without caffeine or nCPAP); 2) nCPAP (5 cmH2O, without caffeine); 3) caffeine (10 mg/kg infused intravenously for 30 min, without nCPAP); and 4) nCPAP + caffeine. Our results showed that nCPAP consistently blunted LCR-related cardiorespiratory inhibition vs. control condition, contrary to caffeine whose overall effect was nonsignificant. In addition, nCPAP condition was characterized by a more consistent and rapid arousal after HCl injection. No significant differences were observed between all tested conditions with regard to swallowing and cough. It is concluded that nCPAP should be further assessed for its usefulness in treating neonatal apneas linked to LCR.
Collapse
Affiliation(s)
- Nadia Boudaa
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Physiology, Université de Sherbrooke, Quebec, Canada J1H 5N4
| | | | | | | | | | | | | |
Collapse
|
24
|
Collins JJP, Kuypers E, Nitsos I, Jane Pillow J, Polglase GR, Kemp MW, Newnham JP, Cleutjens JP, Frints SGM, Kallapur SG, Jobe AH, Kramer BW. LPS-induced chorioamnionitis and antenatal corticosteroids modulate Shh signaling in the ovine fetal lung. Am J Physiol Lung Cell Mol Physiol 2012; 303:L778-87. [PMID: 22962010 DOI: 10.1152/ajplung.00280.2011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chorioamnionitis and antenatal corticosteroids mature the fetal lung functionally but disrupt late-gestation lung development. Because Sonic Hedgehog (Shh) signaling is a major pathway directing lung development, we hypothesized that chorioamnionitis and antenatal corticosteroids modulated Shh signaling, resulting in an altered fetal lung structure. Time-mated ewes with singleton ovine fetuses received an intra-amniotic injection of lipopolysaccharide (LPS) and/or maternal intramuscular betamethasone 7 and/or 14 days before delivery at 120 days gestational age (GA) (term = 150 days GA). Intra-amniotic LPS exposure decreased Shh mRNA levels and Gli1 protein expression, which was counteracted by both betamethasone pre- or posttreatment. mRNA and protein levels of fibroblast growth factor 10 and bone morphogenetic protein 4, which are important mediators of lung development, increased 2-fold and 3.5-fold, respectively, 14 days after LPS exposure. Both 7-day and 14-day exposure to LPS changed the mRNA levels of elastin (ELN) and collagen type I alpha 1 (Col1A1) and 2 (Col1A2), which resulted in fewer elastin foci and increased collagen type I deposition in the alveolar septa. Corticosteroid posttreatment prevented the decrease in ELN mRNA and increased elastin foci and decreased collagen type I deposition in the fetal lung. In conclusion, fetal lung exposure to LPS was accompanied by changes in key modulators of lung development resulting in abnormal lung structure. Betamethasone treatment partially prevented the changes in developmental processes and lung structure. This study provides new insights into clinically relevant prenatal exposures and fetal lung development.
Collapse
Affiliation(s)
- Jennifer J P Collins
- Department of Pediatrics, School for Oncology and Developmental Biology, School for Mental Health and Neuroscience, Maastricht University Medical Center, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Long NM, Shasa DR, Ford SP, Nathanielsz PW. Growth and insulin dynamics in two generations of female offspring of mothers receiving a single course of synthetic glucocorticoids. Am J Obstet Gynecol 2012; 207:203.e1-8. [PMID: 22939726 DOI: 10.1016/j.ajog.2012.06.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/07/2012] [Accepted: 06/13/2012] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Synthetic glucocorticoid administration to women threatening preterm delivery increases neonatal survival. However, mounting evidence shows that fetal exposure to glucocorticoid levels higher than appropriate for current maturation adversely programs offspring development. We examined fetal synthetic glucocorticoid multigenerational metabolic effects on F1 and F2 female offspring. STUDY DESIGN At 0.7 gestation, pregnant F0 ewes received 4 injections of dexamethasone (2 mg, approximately 60 ug.kg(-1) day(-1) 12 hours apart) or saline (control). F1 female offspring were bred to produce F2 female offspring. Postpubertal pancreatic β-cell function was tested in F1 and F2 by intravenous glucose tolerance test. RESULTS F1 and F2 ewe lambs showed reduced birthweight and morphometrics, and similar increased fasting glucose and decreased intravenous glucose tolerance test β-cell response. CONCLUSION This is the first demonstration of multigenerational programming of later life β-cell response by clinically relevant doses of synthetic glucocorticoid indicating the need for study of long-term effects of fetal exposure to synthetic glucocorticoid.
Collapse
Affiliation(s)
- Nathan M Long
- The Center for the Study of Fetal Programming, Laramie, WY, USA
| | | | | | | |
Collapse
|
26
|
Abstract
There is no controversy that women at risk of preterm delivery before 32 to 34 weeks' gestational age should be treated with antenatal steroids. Three recent meta-analyses by the Cochrane Collaboration on the benefits of antenatal steroids, the choice of steroid and dosing, and repeat doses of corticosteroids comprehensively summarize the available clinical information to about 2007. However, there are many unanswered questions about which steroid and dose to use and about their use in selected populations. This review focuses on those areas of uncertainty.
Collapse
Affiliation(s)
- Ronald Wapner
- Department of Obstetrics and Gynecology, Columbia University Medical Center
| | - Alan H. Jobe
- Cincinnati Children’s Hospital Medical Center, Division of Pulmonary Biology, The University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, TEL: (513) 636-8563, FAX: (513) 636-8691
| |
Collapse
|
27
|
Albertine KH, Dahl MJ, Gonzales LW, Wang ZM, Metcalfe D, Hyde DM, Plopper CG, Starcher BC, Carlton DP, Bland RD. Chronic lung disease in preterm lambs: effect of daily vitamin A treatment on alveolarization. Am J Physiol Lung Cell Mol Physiol 2010; 299:L59-72. [PMID: 20382748 DOI: 10.1152/ajplung.00380.2009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neonatal chronic lung disease is characterized by failed formation of alveoli and capillaries, and excessive deposition of matrix elastin, which are linked to lengthy mechanical ventilation (MV) with O(2)-rich gas. Vitamin A supplementation has improved respiratory outcome of premature infants, but there is little information about the structural and molecular manifestations in the lung that occur with vitamin A treatment. We hypothesized that vitamin A supplementation during prolonged MV, without confounding by antenatal steroid treatment, would improve alveolar secondary septation, decrease thickness of the mesenchymal tissue cores between distal air space walls, and increase alveolar capillary growth. We further hypothesized that these structural advancements would be associated with modulated expression of tropoelastin and deposition of matrix elastin, phosphorylated Smad2 (pSmad2), cleaved caspase 3, proliferating cell nuclear antigen (PCNA), VEGF, VEGF-R2, and midkine in the parenchyma of the immature lung. Eight preterm lambs (125 days' gestation, term approximately 150 days) were managed by MV for 3 wk: four were treated with daily intramuscular Aquasol A (vitamin A), 5,000 IU/kg, starting at birth; four received vehicle alone. Postmortem lung assays included quantitative RT-PCR and in situ hybridization, immunoblot and immunohistochemistry, and morphometry and stereology. Daily vitamin A supplementation increased alveolar secondary septation, decreased thickness of the mesenchymal tissue cores between the distal air space walls, and increased alveolar capillary growth. Associated molecular changes were less tropoelastin mRNA expression, matrix elastin deposition, pSmad2, and PCNA protein localization in the mesenchymal tissue core of the distal air space walls. On the other hand, mRNA expression and protein abundance of VEGF, VEGF-R2, midkine, and cleaved caspase 3 were increased. We conclude that vitamin A treatment partially improves lung development in chronically ventilated preterm neonates by modulating expression of tropoelastin, deposition of elastin, and expression of vascular growth factors.
Collapse
Affiliation(s)
- Kurt H Albertine
- Department of Pediatrics, University of Utah Health Sciences Center, Salt Lake City, Utah 84158, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|