1
|
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
|
2
|
Gosavi A, Amin Z, Carter SWD, Choolani MA, Fee EL, Milad MA, Jobe AH, Kemp MW. Antenatal corticosteroids in Singapore: a clinical and scientific assessment. Singapore Med J 2024; 65:479-487. [PMID: 36254928 PMCID: PMC11479002 DOI: 10.4103/singaporemedj.smj-2022-014] [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: 02/03/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
ABSTRACT Preterm birth (PTB; delivery prior to 37 weeks' gestation) is the leading cause of early childhood death in Singapore today. Approximately 9% of Singaporean babies are born preterm; the PTB rate is likely to increase given the increased use of assisted reproduction technologies, changes in the incidence of gestational diabetes/high body mass index and the ageing maternal population. Antenatal administration of dexamethasone phosphate is a key component of the obstetric management of Singaporean women who are at risk of imminent preterm labour. Dexamethasone improves preterm outcomes by crossing the placenta to functionally mature the fetal lung. The dexamethasone regimen used in Singapore today affords a very high maternofetal drug exposure over a brief period of time. Drawing on clinical and experimental data, we reviewed the pharmacokinetic profile and pharmacodynamic effects of dexamethasone treatment regimen in Singapore, with a view to creating a development pipeline for optimising this critically important antenatal therapy.
Collapse
Affiliation(s)
- Arundhati Gosavi
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zubair Amin
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sean William David Carter
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Mahesh Arjandas Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Erin Lesley Fee
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Mark Amir Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, Michigan, USA
| | - Alan Hall Jobe
- Perinatal Research, Department of Pediatrics, Cincinnati Children’s Hospital Medical Centre, University of Cincinnati, Cincinnati, Ohio, USA
| | - Matthew Warren Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
- School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
3
|
Carter SWD, Fee EL, Usuda H, Oguz G, Ramasamy A, Amin Z, Agnihotri B, Wei Q, Xiawen L, Takahashi T, Takahashi Y, Ikeda H, Kumagai Y, Saito Y, Saito M, Mattar C, Evans MI, Illanes SE, Jobe AH, Choolani M, Kemp MW. Antenatal steroids elicited neurodegenerative-associated transcriptional changes in the hippocampus of preterm fetal sheep independent of lung maturation. BMC Med 2024; 22:338. [PMID: 39183288 PMCID: PMC11346182 DOI: 10.1186/s12916-024-03542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Antenatal steroid therapy for fetal lung maturation is routinely administered to women at risk of preterm delivery. There is strong evidence to demonstrate benefit from antenatal steroids in terms of survival and respiratory disease, notably in infants delivered at or below 32 weeks' gestation. However, dosing remains unoptimized and lung benefits are highly variable. Current treatment regimens generate high-concentration, pulsatile fetal steroid exposures now associated with increased risk of childhood neurodevelopmental diseases. We hypothesized that damage-associated changes in the fetal hippocampal transcriptome would be independent of preterm lung function. METHODS Date-mated ewes carrying a single fetus at 122 ± 2dGA (term = 150dGA) were randomized into 4 groups: (i) Saline Control Group, 4×2ml maternal saline intramuscular(IM) injections at 12hr intervals (n = 11); or (ii) Dex High Group, 2×12mg maternal IM dexamethasone phosphate injections at 12hr intervals followed by 2×2ml IM saline injections at 12hr intervals (n = 12; representing a clinical regimen used in Singapore); or (iii) Dex Low Group, 4×1.5mg maternal IM dexamethasone phosphate injections 12hr intervals (n = 12); or (iv) Beta-Acetate Group, 1×0.125mg/kg maternal IM betamethasone acetate injection followed by 3×2ml IM sterile normal saline injections 12hr intervals (n = 8). Lambs were surgically delivered 48hr after first maternal injection at 122-125dGA, ventilated for 30min to establish lung function, and euthanised for necropsy and tissue collection. RESULTS Preterm lambs from the Dex Low and Beta-Acetate Groups had statistically and biologically significant lung function improvements (measured by gas exchange, lung compliance). Compared to the Saline Control Group, hippocampal transcriptomic data identified 879 differentially significant expressed genes (at least 1.5-fold change and FDR < 5%) in the steroid-treated groups. Pulsatile dexamethasone-only exposed groups (Dex High and Dex Low) had three common positively enriched differentially expressed pathways related in part to neurodegeneration ("Prion Disease", "Alzheimer's Disease", "Arachidonic Acid metabolism"). Adverse changes were independent of respiratory function during ventilation. CONCLUSIONS Our data suggests that exposure to antenatal steroid therapy is an independent cause of damage- associated transcriptomic changes in the brain of preterm, fetal sheep. These data highlight an urgent need for careful reconsideration and balancing of how antenatal steroids are used, both for patient selection and dosing regimens.
Collapse
Affiliation(s)
- Sean W D Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Gokce Oguz
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome #02-01, Singapore, 138632, Republic of Singapore
| | - Zubair Amin
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Biswas Agnihotri
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Neonatology Khoo Teck Puat, National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Qin Wei
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Liu Xiawen
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Hideyuki Ikeda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yusaku Kumagai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuya Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Citra Mattar
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Mark I Evans
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Fetal Medicine Foundation of America, New York, NY, USA
| | - Sebastián E Illanes
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Reproductive Biology Program, Center for Biomedical Research and Innovation, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Alan H Jobe
- Centre for Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
| | - Matthew W Kemp
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road NUHS Tower Block, Level 12, Singapore, 119228, Singapore
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Women and Infants Research Foundation, Perth, WA, Australia
| |
Collapse
|
4
|
Paping A, Ehrlich L, Melchior K, Ziska T, Wippermann W, Starke A, Heinichen K, Henrich W, Braun T. A Sustainable Translational Sheep Model for Planned Cesarean Delivery of Contraction-Free Ewes. Reprod Sci 2024; 31:791-802. [PMID: 37848643 PMCID: PMC10912125 DOI: 10.1007/s43032-023-01365-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023]
Abstract
We evaluated whether the sheep constitutes a useful translational model to evaluate anatomical and surgical aspects of cesarean delivery (CD) from a human medical perspective with the aim of both maternal and neonatal well-being. Our hypothesis was that CD in contraction-free ewes is not associated with major complications. Primary endpoint was the transferability of anatomical conditions and surgical techniques of CD from the ewe to the human. Secondary endpoints were maternal and fetal survival, occurrence of retained fetal membranes, metritis, mastitis, or wound infections. Forty-eight Merino ewes were delivered by CD after 95% gestation (142-144 days). Both ewes and newborn lambs were cared for intensively after the delivery. Ovine uterine anatomy during CD appeared slightly different but comparable to the human uterus. Uterine incisions were mostly performed in the uterine horns, not in the uterine corpus. The ovine uterine wall is thinner than in humans. All ewes survived without any major complications. Seventy-seven (88.5%) out of 87 live-born lambs survived without any complications. The contraction-free ewe constitutes an appropriate and safe model to evaluate anatomical and surgical aspects of CD from a human medical perspective. We present a step-by-step manual for successfully planned cesarean delivery for sheep including the perioperative management illustrated with photographs and a five-minute video. With adequate planning and a reasonable number of staff, it is possible to safeguard both maternal and neonatal survival. This sustainable translational medicine model offers additional potential for the offspring to be used for further research studies (e.g., transgenerational inheritance research).
Collapse
Affiliation(s)
- Alexander Paping
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Obstetrics, Augustenburger Platz 1, 13353, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Division of 'Experimental Obstetrics', Berlin, Germany.
| | - Loreen Ehrlich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Division of 'Experimental Obstetrics', Berlin, Germany
| | - Kerstin Melchior
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Division of 'Experimental Obstetrics', Berlin, Germany
| | - Thomas Ziska
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Division of 'Experimental Obstetrics', Berlin, Germany
| | - Wolf Wippermann
- Clinic for Ruminants and Swine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Alexander Starke
- Clinic for Ruminants and Swine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Karin Heinichen
- Oberholz Farm for Teaching and Research, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Wolfgang Henrich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Obstetrics, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Thorsten Braun
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Obstetrics, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Division of 'Experimental Obstetrics', Berlin, Germany
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
Kumagai Y, Kemp MW, Usuda H, Takahashi T, Takahashi Y, Hamada H, Schmidt AF, Hanita T, Watanabe S, Sato S, Ikeda H, Fee EL, Furfaro L, Newnham JP, Jobe AH, Yaegashi N, Saito M. A Reduction in Antenatal Steroid Dose Was Associated with Reduced Cardiac Dysfunction in a Sheep Model of Pregnancy. Reprod Sci 2023; 30:3222-3234. [PMID: 37264260 PMCID: PMC10643432 DOI: 10.1007/s43032-023-01264-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Abstract
Despite widespread use, dosing regimens for antenatal corticosteroid (ACS) therapy are poorly unoptimized. ACS therapy exerts a programming effect on fetal development, which may be associated with an increased risk of cardiovascular disease. Having demonstrated that low-dose steroid therapy is an efficacious means of maturing the preterm lung, we hypothesized that a low-dose steroid exposure would exert fewer adverse functional and transcriptional changes on the fetal heart. We tested this hypothesis using low-dose steroid therapy (10 mg delivered to the ewe over 36 h via constant infusion) and compared cardiac effects with those of a higher dose treatment (30 mg delivered to the ewe over 24 h by intramuscular injection; simulating currently employed clinical ACS regimens). Fetal cardiac function was assessed by ultrasound on the day of ACS treatment initiation. Transcriptomic analyses were performed on fetal myocardial tissue. Relative to saline control, fetuses in the higher-dose clinical treatment group had significantly lower ratios between early diastolic ventricular filling and ventricular filling during atrial systole, and showed the differential expression of myocardial hypertrophy-associated transcripts including βMHC, GADD45γ, and PPARγ. The long-term implications of these changes remain unstudied. Irrespective, optimizing ACS dosing regimens to maximize respiratory benefit while minimizing adverse effects on key organ systems, such as the heart, offers a means of improving the acute and long-term outcomes associated with this important obstetric therapy.
Collapse
Affiliation(s)
- Yusaku Kumagai
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan.
| | - Matthew W Kemp
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
- College of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haruo Usuda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Tsukasa Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Yuki Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Hirotaka Hamada
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | | | - Takushi Hanita
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shimpei Watanabe
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shinichi Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hideyuki Ikeda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Erin L Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Lucy Furfaro
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - John P Newnham
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
- Cincinnati Children's Hospital Medical Centre, Cincinnati, OH, USA
| | - Nobuo Yaegashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Masatoshi Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
9
|
Razaz N, Allen VM, Fahey J, Joseph KS. Antenatal Corticosteroid Prophylaxis at Late Preterm Gestation: Clinical Guidelines vs Clinical Practice. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2023; 45:319-326. [PMID: 36933800 DOI: 10.1016/j.jogc.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVES We investigated how the Antenatal Late Preterm Steroids (ALPS) trial findings have been translated into clinical practice in Canada and the United States (U.S.). METHODS The study included all live births in Nova Scotia, Canada and the United States from 2007 to 2020. ACS administration within specific categories of gestational age was assessed by calculating rates per 100 live births, and temporal changes were quantified using odds ratios (OR) and 95% CI. Temporal trends in optimal and suboptimal ACS use were also assessed. RESULTS In Nova Scotia, the rate of any ACS administration increased significantly among women delivering at 350 to 366 weeks, from 15.2% in 2007-2016 to 19.6% in 2017-2020 (OR 1.36, 95% CI 1.14-1.62). Overall, the U.S. rates were lower than the rates in Nova Scotia. In the U.S., rates of any ACS administration increased significantly across all gestational age categories: among live births at 350 to 366 weeks gestation, any ACS use increased from 4.1% in 2007-2016 to 18.5% in 2017-2020 (OR 5.33, 95% CI 5.28-5.38). Among infants between 240 and 346 weeks gestation in Nova Scotia, 32% received optimally timed ACS, while 47% received ACS with suboptimal timing. Of the women who received ACS in 2020, 34% in Canada and 20% in the U.S. delivered at ≥37 weeks. CONCLUSION Publication of the ALPS trial resulted in increased ACS administration at late preterm gestation in Nova Scotia, Canada and the U.S. However, a significant fraction of women receiving ACS prophylaxis delivered at term gestation.
Collapse
Affiliation(s)
- Neda Razaz
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Victoria M Allen
- Department of Obstetrics and Gynaecology, Dalhousie University and the IWK Health Centre, Halifax, Nova Scotia, Canada
| | - John Fahey
- Reproductive Care Program of Nova Scotia, Halifax, Nova Scotia, Canada
| | - K S Joseph
- Department of Obstetrics and Gynaecology, University of British Columbia, and the Children's and Women's Hospital and Health Centre of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Liu L, Liu C, Li T, Xu B, Xiao Y, Wang Y. Acute Type A Aortic Dissection and Late Pregnancy: What Should We Do? Braz J Cardiovasc Surg 2023; 38:183-190. [PMID: 35675490 PMCID: PMC10010727 DOI: 10.21470/1678-9741-2020-0670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Acute type A aortic dissection (AAAD) in late pregnancy is a rare but severe disease. Lack of clinical experience is the main cause of high mortality. This study tries to investigate the multidisciplinary therapeutic strategy for these patients. CASE PRESENTATION We reported three patients with AAAD in late pregnancy. Sudden chest pain was the main clinical symptom before operation. All three patients and their newborns survived through multidisciplinary approach in diagnosis and treatment. No serious complications occurred during the mid-term follow-up. CONCLUSION Multidisciplinary diagnosis and treatment strategy play a crucial role in saving the lives of pregnant women with AAAD.
Collapse
Affiliation(s)
- Lingchao Liu
- Institute of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chencheng Liu
- Institute of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Tianbo Li
- Institute of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Bo Xu
- Institute of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yingbin Xiao
- Institute of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yong Wang
- Institute of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
11
|
Usuda H, Fee EL, Carter S, Furfaro L, Takahashi T, Takahashi Y, Newnham JP, Milad MA, Saito M, Jobe AH, Kemp MW. Low-dose antenatal betamethasone treatment achieves preterm lung maturation equivalent to that of the World Health Organization dexamethasone regimen but with reduced endocrine disruption in a sheep model of pregnancy. Am J Obstet Gynecol 2022; 227:903.e1-903.e16. [PMID: 35792176 DOI: 10.1016/j.ajog.2022.06.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/08/2022] [Accepted: 06/28/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND The intramuscular administration of antenatal steroids to women at risk of preterm delivery achieves high maternal and fetal plasma steroid concentrations, which are associated with adverse effects and may reduce treatment efficacy. We have demonstrated that antenatal steroid efficacy is independent of peak maternofetal steroid levels once exposure is maintained above a low threshold. OBJECTIVE This study aimed to test, using a sheep model of pregnancy, whether the low-dose antenatal steroid regimen proposed as part of the Antenatal Corticosteroids for Improving Outcomes in Preterm Newborns trial would achieve preterm lung maturation equivalent to that of the existing World Health Organization dexamethasone treatment regimen, but with reduced risk of adverse outcomes. STUDY DESIGN Following ethical review and approval, date-mated ewes with single fetuses received intramuscular injections of either (1) four 6-mg maternal intramuscular injections of dexamethasone phosphate every 12 hours (n=22), (2) 4 2-mg maternal intramuscular injections of betamethasone phosphate every 12 hours (n=21), or (3) 4 2-mL maternal intramuscular injections of saline every 12 hours (n=16). Of note, 48 hours after first injection, (124±1 day), lambs were delivered, ventilated for 30 minutes, and euthanized for sampling. Arterial blood gas, respiratory, hematological, and biochemical data were analyzed for between-group differences with analysis of variance according to distribution and variance, with P<.05 taken as significant. RESULTS After 30 minutes of ventilation, lambs from both steroid-treated groups had significant and equivalent improvements in lung function relative to saline control (P<.05). There was no significant difference in arterial blood pH, pO2, pCO2, lung compliance, ventilator efficiency index, or lung volume at necropsy with a static pressure of 40 cmH2O. The messenger RNA expression of surfactant protein (Sp)a, Spb, Spc, Spd, aquaporin (Aqp)1, Aqp5, and sodium channel epithelial 1 subunit beta (Scnn1b) was equivalent between both steroid groups. Maternal and fetal plasma neutrophil, glucose, and fetal plasma C-peptide levels were significantly elevated in the dexamethasone group, relative to the betamethasone group. Fetal plasma insulin-like growth factor 1 was significantly reduced in the dexamethasone group compared with the betamethasone group (P<0.05). Fetal adrenocorticotropic hormone (r=0.53), maternal glucose value (r=-0.52), and fetal glucose values (r=-0.42) were correlated with maternal weight in the betamethasone group (P<.05), whereas fetal pCO2 and pO2 were not correlated. There was no significant difference between male and female lamb outcomes in any groups for any of the items evaluated. CONCLUSION This study reported that in preterm lambs, a low-dose treatment regimen of 8 mg betamethasone achieves lung maturation equivalent to that of a 24-mg dexamethasone-based regimen, but with smaller perturbations to the maternofetal hypothalamic-pituitary-adrenal axis. These data suggested that given steroid pharmacokinetic differences between sheep and humans, a betamethasone dose of 2 mg may remain above the minimum dose necessary for robust maturation of the preterm lung. Maternal weight-adjusted betamethasone doses might also be a key to reducing perturbations to the maternofetal hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Erin L Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Sean Carter
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Lucy Furfaro
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - John P Newnham
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; School of Veterinary and Life Sciences, Murdoch University, Western Australia, Australia
| | - Mark A Milad
- School of Veterinary and Life Sciences, Murdoch University, Western Australia, Australia
| | - Masatoshi Saito
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Alan H Jobe
- Milad Pharmaceutical Consulting LLC, Plymouth, MI
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, Western Australia, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan; Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati Medical School, Cincinnati, OH; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, Singapore.
| |
Collapse
|
12
|
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
|
13
|
Takahashi T, Jobe AH, Fee EL, Newnham JP, Schmidt AF, Usuda H, Kemp MW. The complex challenge of antenatal steroid therapy nonresponsiveness. Am J Obstet Gynecol 2022; 227:696-704. [PMID: 35932879 DOI: 10.1016/j.ajog.2022.07.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/01/2022]
Abstract
Antenatal steroid therapy is standard care for women at imminent risk of preterm delivery. When deliveries occur within 7 days of treatment, antenatal steroid therapy reduces the risk of neonatal death and improves preterm outcomes by exerting diverse developmental effects on the fetal organs, in particular the preterm lung and cardiovascular system. There is, however, sizable variability in antenatal steroid treatment efficacy, and an important percentage of fetuses exposed to antenatal steroid therapy do not respond sufficiently to derive benefit. Respiratory distress syndrome, for example, is a central metric of clinical trials to assess antenatal steroid outcomes. In the present analysis, we addressed the concept of antenatal steroid nonresponsiveness, and defined a failed or suboptimal response to antenatal steroids as death or a diagnosis of respiratory distress syndrome following treatment. For deliveries at 24 to 35 weeks' gestation, the number needed to treat to prevent 1 case of respiratory distress syndrome was 19 (95% confidence interval, 14-28). Reflecting gestation-dependent risk, for deliveries at >34 weeks' gestation the number needed to treat was 55 (95% confidence interval, 30-304), whereas for elective surgical deliveries at term this number was 106 (95% confidence interval, 61-421). We reviewed data from clinical and animal studies investigating antenatal steroid therapy to highlight the significant incidence of antenatal steroid therapy nonresponsiveness (ie, residual mortality or respiratory distress syndrome after treatment), and the potential mechanisms underpinning this outcome variability. The origins of this variability may be related to both the manner in which the therapy is applied (ie, the treatment regimen itself) and factors specific to the individual (ie, genetic variation, stress, infection). The primary aims of this review were: (1) to emphasize to the obstetrical and neonatal communities the extent of antenatal steroid response variability and its potential impact; (2) to propose approaches by which antenatal steroid therapy may be better applied to improve overall benefit; and (3) to stimulate further research toward the empirical optimization of this important antenatal therapy.
Collapse
Affiliation(s)
- Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Alan H Jobe
- Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - John P Newnham
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | | | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Matthew W Kemp
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia; Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan; School of Veterinary and Life Sciences, Murdoch University, Perth, Australia; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
14
|
Ozdemir F, Acmaz G, Madendag Y, Madendag IC, Muderris II. The effect of steroid administration on fetal diaphragm function. BMC Pregnancy Childbirth 2022; 22:762. [PMID: 36224559 PMCID: PMC9555001 DOI: 10.1186/s12884-022-05074-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 09/16/2022] [Indexed: 11/22/2022] Open
Abstract
Background Antenatal steroid administrations lead to not only accelerated lung maturation, improved blood gas measurements but also lung dynamics and lung compliance. This study aimed to investigate structural and functional changes in diaphragm after antenatal steroid administration. Methods The 79 volunteers were divided into 2 groups according to presence of preterm delivery. Betamethasone (CelestoneR) 12 mg intramuscularly was routinely administered to pregnancies complicated with preterm delivery between 28th -34th weeks of gestation. Same dose was repeated 24 h later. In all patients, diaphragm thickness, diaphragmatic excursion and costophrenic angle were measured in both the inspirium and expirium stage of respiration. This is an observational cross-sectional study. Results Diaphragm thickness, diaphragmatic excursion and diaphragm thickening fraction parameters were improved but costophrenic angle was not different 7 days after steroid administration. Diaphragm thickness, diaphragmatic excursion and costophrenic angle changes during inspiration and expiration stage after 7 days of betamethasone treatment reflects the effect of steroid administration on diaphragm muscle. Comparisons of the differences that occur after steroid rescue protocol were done by subtracting the diaphragm thickness, diaphragmatic excursion and costophrenic angle parameters before the treatment from the diaphragm thickness, diaphragmatic excursion and costophrenic angle parameters 7 days after steroid treatment respectively. Conclusion Diaphragm became more mobile in patients with preterm labor, 7 days after steroid administration. This situation reflects positive effect of steroid administration on diaphragm function.
Collapse
Affiliation(s)
- Fatma Ozdemir
- Department of Obstetrics and Gynecology, Erciyes University Faculty of Medicine, Yenidogan District, Turhan Baytop Street No:1, 38280, Kayseri, Melikgazi Kayseri, Turkey
| | - Gokhan Acmaz
- Department of Obstetrics and Gynecology, Erciyes University Faculty of Medicine, Yenidogan District, Turhan Baytop Street No:1, 38280, Kayseri, Melikgazi Kayseri, Turkey.
| | - Yusuf Madendag
- Department of Obstetrics and Gynecology, Erciyes University Faculty of Medicine, Yenidogan District, Turhan Baytop Street No:1, 38280, Kayseri, Melikgazi Kayseri, Turkey
| | - Ilknur Col Madendag
- Obstetrics and Gynecology Department, Kayseri City Hospital, Kayseri, Turkey
| | - Iptisam Ipek Muderris
- Department of Obstetrics and Gynecology, Erciyes University Faculty of Medicine, Yenidogan District, Turhan Baytop Street No:1, 38280, Kayseri, Melikgazi Kayseri, Turkey
| |
Collapse
|
15
|
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: 16] [Impact Index Per Article: 8.0] [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
|
16
|
Takahashi T, Takahashi Y, Fee EL, Saito M, Yaegashi N, Usuda H, Bridges JP, Milad MA, Furfaro L, Carter S, Schmidt AF, Newnham JP, Jobe AH, Kemp MW. Continuous but not pulsed low-dose fetal betamethasone exposures extend the durability of antenatal steroid therapy. Am J Physiol Lung Cell Mol Physiol 2022; 322:L784-L793. [PMID: 35380907 DOI: 10.1152/ajplung.00018.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antenatal steroid (ANS) therapy is standard care for women at imminent risk of preterm labor. Despite extensive and long-standing use, 40-50% of babies exposed antenatally to steroids do not derive benefit; remaining undelivered 7d or more after ANS treatment is associated with a lack of treatment benefit, and increased risk of harms. We used a pregnant sheep model to evaluate the impact of continuous vs. pulsed ANS treatments on fetal lung maturation at an extended, eight-day treatment to delivery interval. Continuous low-dose ANS treatments for more than 72 hours in duration improved fetal lung maturation at eight days after treatment initiation. If fetal ANS exposure was interrupted, the beneficial ANS effect was lost. Truncated treatments, including that simulating the current clinical treatment regimen, did not improve lung function. Variable fetal lung maturation was correlated to the amount of saturated phosphatidylcholine present in the lung fluid. These data demonstrate that: i) the durability of ANS therapy may be enhanced by employing an extended, low-dose treatment regimen with reducing total dose; and ii) interrupting the continuity of fetal exposure by allowing it to fall below a minimal threshold was associated with comparably poor functional maturation of the preterm ovine lung.
Collapse
Affiliation(s)
- Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yuki Takahashi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Nobuo Yaegashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - James P Bridges
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mark A Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, MI, United States
| | - Lucy Furfaro
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Sean Carter
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Augusto F Schmidt
- Department of Neonatology, Pulmonary Biology and Pediatrics, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, Cincinnati, OH, United States.,Miller School of Medicine, University of Miami, Miami, FL, United States
| | - John P Newnham
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Alan Hall Jobe
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,Department of Neonatology, Pulmonary Biology and Pediatrics, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew W Kemp
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.,School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
17
|
Asztalos EV, Murphy KE, Matthews SG. A Growing Dilemma: Antenatal Corticosteroids and Long-Term Consequences. Am J Perinatol 2022; 39:592-600. [PMID: 33053595 DOI: 10.1055/s-0040-1718573] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE A single course of synthetic antenatal corticosteroids is standard care for women considered to be at risk for preterm birth before 34 weeks of gestation. While the intended target is the fetal lung, the fetal brain contains remarkably high levels of glucocorticoid receptors in structures critical in the regulation of behavior and endocrine function. Negative programming signals may occur which can lead to permanent maladaptive changes and predispose the infant/child to an increased risk in physical, mental, and developmental disorders. METHODS Framed around these areas of concerns for physical, mental, and developmental disorders, this narrative review drew on studies (animal and clinical), evaluating the long-term effects of antenatal corticosteroids to present the case that a more targeted approach to the use of antenatal corticosteroids for the betterment of the fetus urgently needed. RESULTS Studies raised concerns about the potential negative long-term consequences, especially for the exposed fetus who was born beyond the period of the greatest benefit from antenatal corticosteroids. The long-term consequences are more subtle in nature and usually manifest later in life, often beyond the scope of most clinical trials. CONCLUSION Continued research is needed to identify sufficient safety data, both short term and long term. Caution in the use of antenatal corticosteroids should be exercised while additional work is undertaken to optimize dosing strategies and better identify women at risk of preterm birth prior to administration of antenatal corticosteroids. KEY POINTS · A single-course ACS is a remarkable therapy with substantial benefits.. · There is a potential of long-term neurodevelopmental consequences in the ACS-exposed fetus.. · There is a need to improve dosing strategies and identification of appropriate at risk women..
Collapse
Affiliation(s)
- Elizabeth V Asztalos
- Department of Newborn and Developmental Paediatrics, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kellie E Murphy
- Department of Obstetrics and Gynecology, Sinai Health Systems, University of Toronto, Toronto, Ontario, Canada
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Takahashi T, Fee EL, Takahashi Y, Saito M, Yaegashi N, Usuda H, Furfaro L, Carter S, Schmidt AF, Newnham JP, Jobe AH, Kemp MW. Betamethasone phosphate reduces the efficacy of antenatal steroid therapy and is associated with lower birthweights when administered to pregnant sheep in combination with betamethasone acetate. Am J Obstet Gynecol 2022; 226:564.e1-564.e14. [PMID: 34626553 DOI: 10.1016/j.ajog.2021.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Antenatal corticosteroid therapy is a standard of care for women at imminent risk of preterm labor. However, the optimal (maximum benefit and minimal risk of side effects) antenatal corticosteroid dosing strategy remains unclear. Although conveying overall benefit when given to the right patient at the right time, antenatal corticosteroid treatment efficacy is highly variable and is not risk-free. Building on earlier findings, we hypothesized that when administered in combination with slow-release betamethasone acetate, betamethasone phosphate and the high maternal-fetal betamethasone concentrations it generates are redundant for fetal lung maturation. OBJECTIVE Using an established sheep model of prematurity and postnatal ventilation of the preterm lamb, we aimed to compare the pharmacodynamic effects of low-dosage treatment with betamethasone acetate only against a standard dosage of betamethasone phosphate and betamethasone acetate as recommended by the American College of Obstetricians and Gynecologists for women at risk of imminent preterm delivery between 24 0/7 and 35 6/7 weeks' gestation. STUDY DESIGN Ewes carrying a single fetus at 122±1 days' gestation (term=150 days) were randomized to receive either (1) maternal intramuscular injections of sterile saline (the saline negative control group, n=12), (2) 2 maternal intramuscular injections of 0.25 mg/kg betamethasone phosphate+betamethasone acetate administered at 24-hour dosing intervals (the betamethasone phosphate+betamethasone acetate group, n=12); or (3) 2 maternal intramuscular injections of 0.125 mg/kg betamethasone acetate administered at 24-hour dosing intervals (the betamethasone acetate group, n=11). The fetuses were surgically delivered 48 hours after treatment initiation and ventilated for 30 minutes to determine functional lung maturation. The fetuses were euthanized after ventilation, and the lungs were collected for analysis using quantitative polymerase chain reaction and Western blot assays. Fetal plasma adrenocorticotropic hormone levels were measured in the cord blood samples taken at delivery. RESULTS Preterm lambs were defined as either antenatal corticosteroid treatment responders or nonresponders using an arbitrary cutoff, being a PaCO2 level at 30 minutes of ventilation being more extreme than 2 standard deviations from the mean value of the normally distributed saline control group values. Compared with the animals in the saline control group, the animals in the antenatal corticosteroid treatment groups showed significantly improved lung physiological responses (blood gas and ventilation data) and had a biochemical signature (messenger RNA and surfactant protein assays) consistent with functional maturation. However, the betamethasone acetate group had a significantly higher treatment response rate than the betamethasone phosphate+betamethasone acetate group. These physiological results were strongly correlated to the amount of surfactant protein A. Birthweight was lower in the betamethasone phosphate+betamethasone acetate group and the fetal hypothalamic-pituitary-adrenal axis was suppressed to a greater extent in the betamethasone phosphate+betamethasone acetate group. CONCLUSION Low-dosage antenatal corticosteroid therapy solely employing betamethasone acetate was sufficient for fetal lung maturation. The elevated maternal-fetal betamethasone concentrations associated with the coadministration of betamethasone phosphate did not in addition improve lung maturation but were associated with greater fetal hypothalamic-pituitary-adrenal axis suppression, a lower antenatal corticosteroid treatment response rate, and lower birthweight-outcomes not desirable in a clinical setting. These data warranted a clinical investigation of sustained low-dosage antenatal corticosteroid treatments that avoid high maternal-fetal betamethasone exposures.
Collapse
Affiliation(s)
- Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Yuki Takahashi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Nobuo Yaegashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Lucy Furfaro
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Sean Carter
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Augusto F Schmidt
- Perinatal Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, Cincinnati, OH; Miller School of Medicine, University of Miami, Miami, FL
| | - John P Newnham
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Perinatal Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, Cincinnati, OH
| | - Matthew W Kemp
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan; School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
19
|
Shinwell ES, Gurevitz P, Portnov I. Current evidence for prenatal and postnatal corticosteroids in preterm infants. Arch Dis Child Fetal Neonatal Ed 2022; 107:121-125. [PMID: 33658282 DOI: 10.1136/archdischild-2020-319706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/16/2021] [Accepted: 02/17/2021] [Indexed: 01/08/2023]
Abstract
Antenatal corticosteroids undoubtedly save many lives and improve the quality of many others. However, the currently accepted dosage schedule has been in place since 1972, and recent studies have suggested that beneficial effects may be seen with less. Most but not all studies of long-term outcome show no adverse effects. The use of antenatal corticosteroids in women with COVID-19 raises important questions regarding potential risks and benefits. However, currently, most authorities recommend continuing according to published guidelines. With regard to postnatal corticosteroids, alternatives to systemic dexamethasone, the somewhat tainted standard of care, show promise in preventing bronchopulmonary dysplasia without adverse effects. Systemic hydrocortisone and inhaled corticosteroids are of note. The mixture of surfactant and corticosteroids deserves particular attention in the coming years.
Collapse
Affiliation(s)
- Eric S Shinwell
- Neonatology, Ziv Medical Center, Tzfat, Israel .,Azrieli Faculty of Medicine, Bar-Ilan University, Tzfat, Israel
| | | | | |
Collapse
|
20
|
Berger R, Kyvernitakis I, Maul H. Administration of Antenatal Corticosteroids: Current State of Knowledge. Geburtshilfe Frauenheilkd 2022; 82:287-296. [PMID: 35250378 PMCID: PMC8893986 DOI: 10.1055/a-1555-3444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022] Open
Abstract
The administration of a single course of corticosteroids before week 34 + 0 of gestation in cases with impending preterm birth is now standard procedure in obstetric care and firmly
established in the guidelines of different countries. But despite the apparently convincing data, numerous aspects of this intervention have not yet been properly studied. It is still not
clear which corticosteroid achieves the best results. There are very few studies on what constitutes an appropriate dose, circadian rhythms, the time frame in which corticosteroids are
effective, and the balance between the risks and benefits of repeat administration. As the existing studies have rarely included patients before week 24 + 0 of gestation, we have very little
information on the possible benefits of administering corticosteroids before this timepoint. If corticosteroids are administered antenatally after week 34 + 0 of gestation, the short-term
benefit may be offset by the long-term adverse effect on psychomotor development. This present study summarizes the current state of knowledge regarding these issues.
Collapse
Affiliation(s)
- Richard Berger
- Marienhaus Klinikum St. Elisabeth, Klinik für Gynäkologie und Geburtshilfe, Neuwied, Germany
| | - Ioannis Kyvernitakis
- Asklepios Kliniken Barmbek, Wandsbek und Nord-Heidberg, Frauenkliniken, Hamburg, Germany
| | - Holger Maul
- Asklepios Kliniken Barmbek, Wandsbek und Nord-Heidberg, Frauenkliniken, Hamburg, Germany
| |
Collapse
|
21
|
Ryan RM, Paintlia MK, Newton DA, Spyropoulos DD, Kemp M, Jobe AH, Baatz JE. Oxygen and steroids affect the regulatory role of natriuretic peptide receptor-C on surfactant secretion by type II cells. Am J Physiol Lung Cell Mol Physiol 2022; 322:L13-L22. [PMID: 34668435 PMCID: PMC8721905 DOI: 10.1152/ajplung.00300.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Atrial natriuretic peptide (ANP) and its receptors natriuretic peptide receptor (NPR)-A and NPR-C are all highly expressed in alveolar epithelial type II cells (AEC2s) in the late-gestation ovine fetal lung and are dramatically decreased postnatally. However, of all the components, NPR-C stimulation inhibits ANP-mediated surfactant secretion. Since alveolar oxygen increases dramatically after birth, and steroids are administered to mothers antenatally to enhance surfactant lung maturity, we investigated the effects of O2 concentration and steroids on NPR-C-mediated surfactant secretion in AEC2s. NPR-C expression was highest at 5% O2 while being suppressed by 21% O2, in cultured mouse lung epithelial cells (MLE-15s) and/or human primary AEC2s. Surfactant protein-B (SP-B) was significantly elevated in media from both in vitro and ex vivo culture at 13% O2 versus 21% O2 in the presence of ANP or terbutaline (TER). Both ANP and C-ANP (an NPR-C agonist) attenuated TER-induced SP-B secretion; this effect was reversed by dexamethasone (DEX) pretreatment in AEC2s and by transfection with NPR-C siRNA in MLE-15 cells. DEX markedly reduced AEC2 NPR-C expression, and pregnant ewes treated with betamethasone showed reduced ANP in fetal sheep lung fluid. These data suggest that elevated O2 downregulates AEC2 NPR-C and that steroid-mediated NPR-C downregulation in neonatal lungs may provide a novel mechanism for their effect on perinatal surfactant production.
Collapse
Affiliation(s)
- Rita M. Ryan
- 1Case Western Reserve University, UH Rainbow Babies and Children’s Hospital, Cleveland, Ohio,2Darby Children’s Research Institute, Department of Pediatrics,
Medical University of South Carolina, Charleston, South Carolina
| | - Manjeet K. Paintlia
- 2Darby Children’s Research Institute, Department of Pediatrics,
Medical University of South Carolina, Charleston, South Carolina
| | - Danforth A. Newton
- 2Darby Children’s Research Institute, Department of Pediatrics,
Medical University of South Carolina, Charleston, South Carolina
| | - Demetri D. Spyropoulos
- 3Department of Pathology and Laboratory Medicine, Medical
University of South Carolina, Charleston, South Carolina
| | - Matthew Kemp
- 4Department of Obstetrics and Gynaecology, University of
Western Australia, Perth, Western Australia, Australia,5Department of Obstetrics and Gynecology, Yong Loo Lin School of
Medicine, National University of Singapore, Singapore
| | - Alan H. Jobe
- 6Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - John E. Baatz
- 2Darby Children’s Research Institute, Department of Pediatrics,
Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
22
|
Abstract
Prematurity remains a leading cause of perinatal morbidity and mortality, and also has significant implications for long-term health. Obstetricians have a key role to play in improving outcomes for infants born at extremely preterm gestations. This review explores the evidence for interventions available to obstetricians caring for women at risk of birthing at extremely preterm gestations, including antenatal corticosteroids, magnesium sulfate, tocolysis and antibiotics. It also addresses the importance of strategies to facilitate safe in-utero transfer, to maximise the chance of extremely preterm births occurring in tertiary centers, and the clinical value of strategies by which preterm birth can be predicted. The paper concludes with an appraisal of evidence for different modes of birth at extremely preterm gestations, and for delayed cord clamping.
Collapse
|
23
|
Anoshchenko O, Milad MA, Unadkat JD. Estimating fetal exposure to the P-gp substrates, corticosteroids, by PBPK modeling to inform prevention of neonatal respiratory distress syndrome. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1057-1070. [PMID: 34273255 PMCID: PMC8452292 DOI: 10.1002/psp4.12674] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 11/08/2022]
Abstract
We have previously developed a maternal-fetal physiologically-based pharmacokinetic (m-f PBPK) model to dynamically predict (and verify) fetal-maternal exposure to drugs that passively diffuse across the placenta. Here, we extended the application of this model to dynamically predict fetal exposure to drugs which are effluxed by placental P-glycoprotein, namely the antenatal corticosteroids (ACS; dexamethasone [DEX], and betamethasone [BET]). To do so, we estimated both the placental P-gp mediated efflux clearance (CL) and the passive diffusion CL of the ACS. The efficacy and toxicity of the currently used maternal ACS dosing regimens to prevent neonatal respiratory distress syndrome could be improved by altering their dosing regimens. Therefore, to illustrate the utility of our m-f PBPK model, we used it to design alternative dosing regimens of DEX and BET that could potentially improve their efficacy and reduce their toxicity. The redesigned dosing regimens are convenient to administer, maintain maternal-fetal exposure (area under the concentration-time curve [AUC]) or maximum plasma concentration (Cmax ) or both (DEX and BET) or minimize maternal exposure while maintaining fetal drug plasma concentrations above the minimum therapeutic threshold of 1 ng/ml for 48 h (BET only; based on efficacy data in sheep). To our knowledge, this is the first study to dynamically predict fetal plasma concentrations of placental P-gp effluxed drugs. Our approach and our m-f PBPK model could be used in the future to predict maternal-fetal exposure to any drug and to design alternative dosing regimens of the drug.
Collapse
Affiliation(s)
- Olena Anoshchenko
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Washington, Seattle, USA
| | - Mark A Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, Michigan, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Washington, Seattle, USA
| |
Collapse
|
24
|
Jobe AH, Schmidt AF. Chapter for antenatal steroids - Treatment drift for a potent therapy with unknown long-term safety seminars in fetal and neonatal medicine. Semin Fetal Neonatal Med 2021; 26:101231. [PMID: 33773951 DOI: 10.1016/j.siny.2021.101231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter on therapeutic drift with antenatal steroids will make the case that this pilar of treatment to improve the outcomes of preterm infants, despite multiple Randomized Control Trials (RCTs) and meta-analysis, has multiple gaps in solid clinical data to support any expanded use of Antenatal Corticosteroids (ACS). A basic problem is that agents used for ACS have never been evaluated to minimize fetal exposures. Based on the premise that all drug exposure to the fetus should be minimized and only used when necessary, ACS is a potent developmental modulator that has never been evaluated to minimize the dose and duration of fetal exposure. The use of ACS is expanding to late preterm infants where the benefit is modest, to elective C-sections, and periviable fetuses, with minimal RCT data of long-term benefit. Relevant animal experiments demonstrate that much lower doses will induce lung maturation in sheep and primates. Another area of drift in the use of ACS is based on the assumption that the old RCT data accurately predict the magnitude of benefit when ACS is used today with entirely different OB and neonatal care strategies to improve outcomes. We do not have data that demonstrate the effectiveness of ACS in very low resource environments, where most of the preterm mortality occurs. The final concern is the risk of ACS to the infant and child. Short-term risks are minimal but dysmaturation effects of ACS on multiple organ systems (lung, heart, brain, and kidney) may result in disease presentation in later life.
Collapse
Affiliation(s)
- Alan H Jobe
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| | - Augusto F Schmidt
- The University of Miami, Miller School of Medicine, Coral Gables, FL, 33124, USA.
| |
Collapse
|
25
|
Ali H, Salama H, Robertson N, Olukade T, Al-Obaidly S, Al-Qubaisi M, Al Rifai H. Antenatal corticosteroids and short-term neonatal outcomes in term and near-term infants of diabetic mothers. Analysis of the Qatar PEARL-peristat registry. J Perinat Med 2021; 49:377-382. [PMID: 33098633 DOI: 10.1515/jpm-2020-0249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/02/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVES A recent discussion surrounding the extension of antenatal corticosteroid (ACS) use beyond 34 weeks of gestation did not include the subgroup of infants of diabetic mothers (IDM). We aimed to examine the association between ACS exposure and outcomes in neonates born at term and at near-term gestation in a large cohort of IDMs. METHODS We selected 13976 eligible near-term and term infants who were included in the PEARL-Peristat Perinatal Registry Study (PPS). We assessed the association of ACS exposure with neonatal outcomes in a multivariate regression model that controlled for diabetes mellitus (DM) and other perinatal variables. RESULTS The incidence of DM was 28% (3,895 of 13,976) in the cohort. Caesarean section was performed in one-third of the study population. The incidence of ACS exposure was low (1.8%) and typically occurred>2 weeks before delivery. The incidence rates of respiratory distress syndrome (RDS)/ transient tachypnoea of newborns (TTN), all-cause neonatal intensive care unit (NICU) admissions, NICU admissions for hypoglycaemia, and low 5-min Apgar scores were 3.5, 8.8, 1.3, and 0.1%, respectively. In a multivariate regression model, ACS was associated with a slight increase in NICU admissions (OR: 1.44; 95% CI: 1.04-2.03; p=0.028), but not with RDS/TTN. CONCLUSIONS Although the low exposure rate was a limitation, ACS administration did not reduce respiratory morbidity in near-term or term IDMs. It was independently associated with an increase in NICU admissions. Randomized controlled trials are required to assess the efficacy and safety of ACS administration in diabetic mothers at late gestation.
Collapse
Affiliation(s)
- Haytham Ali
- Sidra Medical and Research Center, Doha, Qatar
| | - Husam Salama
- Department of Neonatalogy, Hamad Medical Corp, Doha, Qatar
| | - Nicola Robertson
- Department of Obstetrics and Gynaeocology, University College Hospital, London, United Kingdom of Great Britain and Northern Ireland
| | - Tawa Olukade
- Department of Obstetrics and Gynecology, Hamad Medical Corp, Doha, Qatar
| | - Sawsan Al-Obaidly
- Department of Obstetrics and Gynecology, Hamad Medical Corp, Doha, Qatar
| | - Mai Al-Qubaisi
- Department of Obstetrics and Gynecology, Hamad Medical Corp, Doha, Qatar
| | - Hilal Al Rifai
- Department of Neonatology, Hamad Medical Corp, Doha, Qatar
| |
Collapse
|
26
|
Jobe AH, Kemp M, Schmidt A, Takahashi T, Newnham J, Milad M. Antenatal corticosteroids: a reappraisal of the drug formulation and dose. Pediatr Res 2021; 89:318-325. [PMID: 33177675 PMCID: PMC7892336 DOI: 10.1038/s41390-020-01249-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 12/22/2022]
Abstract
We review the history of antenatal corticosteroid therapy (ACS) and present recent experimental data to demonstrate that this, one of the pillars of perinatal care, has been inadequately evaluated to minimize fetal exposure to these powerful medications. There have been concerns since 1972 that fetal exposures to ACS convey risk. However, this developmental modulator, with its multiple widespread biologic effects, has not been evaluated for drug choice, dose, or duration of treatment, despite over 30 randomized trials. The treatment used in the United States is two intramuscular doses of a mixture of 6 mg betamethasone phosphate (Beta P) and 6 mg betamethasone acetate (Beta Ac). To optimize outcomes with ACS, the goal should be to minimize fetal drug exposure. We have determined that the minimum exposure needed for fetal lung maturation in sheep, monkeys, and humans (based on published cord blood corticosteroid concentrations) is about 1 ng/ml for a 48-h continuous exposure, far lower than the concentration reached by the current dosing. Because the slowly released Beta Ac results in prolonged fetal exposure, a drug containing Beta Ac is not ideal for ACS use. IMPACT: Using sheep and monkey models, we have defined the minimum corticosteroid exposure for a fetal lung maturation. These results should generate new clinical trials of antenatal corticosteroids (ACS) at much lower fetal exposures to ACS, possibly given orally, with fewer risks for the fetus.
Collapse
Affiliation(s)
- Alan H. Jobe
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,grid.24827.3b0000 0001 2179 9593Perinatal Institute, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH USA
| | - Matthew Kemp
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,grid.412757.20000 0004 0641 778XCentre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan ,grid.1025.60000 0004 0436 6763School of Veterinary and Life Sciences, Murdoch University, Perth, WA Australia
| | - Augusto Schmidt
- grid.26790.3a0000 0004 1936 8606Division of Neonatology, Department of Pediatrics, University of Miami, Miami, FL USA
| | - Tsukasa Takahashi
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,grid.412757.20000 0004 0641 778XCentre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - John Newnham
- grid.1012.20000 0004 1936 7910Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia
| | - Mark Milad
- Milad Pharmaceutical Consulting, Plymouth, MI USA
| |
Collapse
|
27
|
Schmidt AF, Kannan PS, Bridges J, Presicce P, Jackson CM, Miller LA, Kallapur SG, Chougnet CA, Jobe AH. Prenatal inflammation enhances antenatal corticosteroid-induced fetal lung maturation. JCI Insight 2020; 5:139452. [PMID: 33328385 PMCID: PMC7819743 DOI: 10.1172/jci.insight.139452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/11/2020] [Indexed: 11/17/2022] Open
Abstract
Respiratory complicˆations are the major cause of morbidity and mortality among preterm infants, which is partially prevented by the administration of antenatal corticosteroids (ACS). Most very preterm infants are exposed to chorioamnionitis, but short- and long-term effects of ACS treatment in this setting are not well defined. In low-resource settings, ACS increased neonatal mortality by perhaps increasing infection. We report that treatment with low-dose ACS in the setting of inflammation induced by intraamniotic lipopolysaccharide (LPS) in rhesus macaques improves lung compliance and increases surfactant production relative to either exposure alone. RNA sequencing shows that these changes are mediated by suppression of proliferation and induction of mesenchymal cellular death via TP53. The combined exposure results in a mature-like transcriptomic profile with inhibition of extracellular matrix development by suppression of collagen genes COL1A1, COL1A2, and COL3A1 and regulators of lung development FGF9 and FGF10. ACS and inflammation also suppressed signature genes associated with proliferative mesenchymal progenitors similar to the term gestation lung. Treatment with ACS in the setting of inflammation may result in early respiratory advantage to preterm infants, but this advantage may come at a risk of abnormal extracellular matrix development, which may be associated with increased risk of chronic lung disease.
Collapse
Affiliation(s)
- Augusto F. Schmidt
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Paranthaman S. Kannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James Bridges
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Pietro Presicce
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Courtney M. Jackson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lisa A. Miller
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UCD, Davis, California, USA
| | - Suhas G. Kallapur
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alan H. Jobe
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Variability in the efficacy of a standardized antenatal steroid treatment was independent of maternal or fetal plasma drug levels: evidence from a sheep model of pregnancy. Am J Obstet Gynecol 2020; 223:921.e1-921.e10. [PMID: 32445634 DOI: 10.1016/j.ajog.2020.05.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Administration of antenatal steroids is standard of care for women assessed to be at imminent risk of preterm delivery. There is a marked variation in antenatal steroid dosing strategy, selection for treatment criteria, and agent choice worldwide. This, combined with very limited optimization of antenatal steroid use per se, means that treatment efficacy is highly variable, and the rate of respiratory distress syndrome is decreased to perhaps as low as 40%. In some cases, antenatal steroid use is associated with limited benefit and potential harm. OBJECTIVE We hypothesized that individual differences in maternofetal steroid exposure would contribute to observed variability in antenatal steroid treatment efficacy. Using a chronically catheterized sheep model of pregnancy, we aimed to explore the relationship between maternofetal steroid exposure and antenatal steroid treatment efficacy as determined by functional lung maturation in preterm lambs undergoing ventilation. STUDY DESIGN Ewes carrying a single fetus underwent surgery to catheterize a fetal and maternal jugular vein at 119 days' gestation. Animals recovered for 24 hours before being randomized to either (1) a single maternal intramuscular injection of 2 mL saline (negative control group, n=10) or (2) a single maternal intramuscular injection of 0.25 mg/kg betamethasone phosphate plus acetate (antenatal steroid group, n=20). Serial maternal and fetal plasma samples were collected from each animal after 48 hours before fetuses were delivered and ventilated for 30 minutes. Total and free plasma betamethasone concentration was measured by mass spectrometry. Fetal lung tissue was collected for analysis using quantitative polymerase chain reaction. RESULTS One animal from the control group and one animal from the antenatal steroid group did not complete their treatment protocol and were removed from analyses. Animals in the antenatal steroid group were divided into a responder subgroup (n=12/19) and a nonresponder subgroup (n=7/19) using a cutoff of partial pressure of arterial CO2 at 30-minute ventilation within 2 standard deviations of the mean value from saline-treated negative control group animals. Although antenatal steroid improved fetal lung maturation in the undivided antenatal steroid group and in the responder subgroup both physiologically (blood gas- and ventilation-related data) and biochemically (messenger ribonucleic acid expression related to fetal lung maturation), these values did not improve relative to saline-treated control group animals in the antenatal steroid nonresponder subgroup. No differences in betamethasone distribution, clearance, or protein binding were identified between the antenatal steroid responder and nonresponder subgroups. CONCLUSION This study correlated individual maternofetal steroid exposures with preterm lung maturation as determined by pulmonary ventilation. Herein, approximately 40% of preterm lambs exposed to antenatal steroids had lung maturation that was not significantly different to saline-treated control group animals. These nonresponsive animals received maternal and fetal betamethasone exposures identical to animals that had a significant improvement in functional lung maturation. These data suggest that the efficacy of antenatal steroid therapy is not solely determined by maternofetal drug levels and that individual fetal or maternal factors may play a role in determining treatment outcomes in response to glucocorticoid signaling.
Collapse
|
29
|
Yadav E, Yadav N, Hus A, Yadav JS. Aquaporins in lung health and disease: Emerging roles, regulation, and clinical implications. Respir Med 2020; 174:106193. [PMID: 33096317 DOI: 10.1016/j.rmed.2020.106193] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/17/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
Abstract
Aquaporins (AQPs) aka water channels are a family of conserved transmembrane proteins (~30 kDa monomers) expressed in various organ systems. Of the 13 AQPs (AQP0 through AQP12) in the human body, four (AQPs 1, 3, 4, and 5) are expressed in the respiratory system. These channels are conventionally known for mediating transcellular fluid movements. Certain AQPs (aquaglyceroporins) have the capability to transport glycerol and potentially other solutes. There is an emerging body of literature unveiling the non-conventional roles of AQPs such as in cell proliferation and migration, gas permeation, signal potentiation, etc. Initial gene knock-out studies established a physiological role for lung AQPs, particularly AQP5, in maintaining homeostasis, by mediating fluid secretion from submucosal glands onto the airway surface liquid (ASL) lining. Subsequent studies have highlighted the functional significance of AQPs, particularly AQP1 and AQP5 in lung pathophysiology and diseases, including but not limited to chronic and acute lung injury, chronic obstructive pulmonary disease (COPD), other inflammatory lung conditions, and lung cancer. AQP1 has been suggested as a potential prognostic marker for malignant mesothelioma. Recent efforts are directed toward exploiting AQPs as targets for diagnosis, prevention, intervention, and/or treatment of various lung conditions. Emerging information on regulatory pathways and directed mechanistic research are posited to unravel novel strategies for these clinical implications. Future considerations should focus on development of AQP inhibitors, blockers, and modulators for therapeutic needs, and better understanding the role of lung-specific AQPs in inter-individual susceptibility to chronic lung diseases such as COPD and cancer.
Collapse
Affiliation(s)
- Ekta Yadav
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Niket Yadav
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, 22908-0738, USA
| | - Ariel Hus
- Department of Biology, University of Miami, Coral Gables, Florida, 33146, USA
| | - Jagjit S Yadav
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
30
|
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: 12] [Impact Index Per Article: 3.0] [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
|
31
|
Thevathasan I, Said JM. Controversies in antenatal corticosteroid treatment. Prenat Diagn 2020; 40:1138-1149. [PMID: 32157719 DOI: 10.1002/pd.5664] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/17/2019] [Accepted: 12/08/2019] [Indexed: 12/17/2022]
Abstract
Antenatal corticosteroids are now established as one of the cornerstones of therapy in the prevention of neonatal morbidity and mortality prior to preterm birth. Although this practice is widely accepted, a significant number of controversies exist. This review explores the knowledge gaps regarding the use of antenatal corticosteroids in the preterm, late preterm and term populations. Furthermore, the role of antenatal corticosteroids in special populations, such as diabetes, multiple pregnancies and periviable gestations, where high-quality data from randomized controlled trials are lacking, is also considered.
Collapse
Affiliation(s)
- Iniyaval Thevathasan
- Maternal Fetal Medicine, Joan Kirner Women's & Children's Sunshine Hospital, Western Health, St Albans, Victoria, Australia
| | - Joanne M Said
- Maternal Fetal Medicine, Joan Kirner Women's & Children's Sunshine Hospital, Western Health, St Albans, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
32
|
Hrabalkova L, Takahashi T, Kemp MW, Stock SJ. Antenatal Corticosteroids for Fetal Lung Maturity - Too Much of a Good Thing? Curr Pharm Des 2020; 25:593-600. [PMID: 30914016 DOI: 10.2174/1381612825666190326143814] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Between 5-15% of babies are born prematurely worldwide, with preterm birth defined as delivery before 37 completed weeks of pregnancy (term is at 40 weeks of gestation). Women at risk of preterm birth receive antenatal corticosteroids as part of standard care to accelerate fetal lung maturation and thus improve neonatal outcomes in the event of delivery. As a consequence of this treatment, the entire fetal organ system is exposed to the administered corticosteroids. The implications of this exposure, particularly the long-term impacts on offspring health, are poorly understood. AIMS This review will consider the origins of antenatal corticosteroid treatment and variations in current clinical practices surrounding the treatment. The limitations in the evidence base supporting the use of antenatal corticosteroids and the evidence of potential harm to offspring are also summarised. RESULTS Little has been done to optimise the dose and formulation of antenatal corticosteroid treatment since the first clinical trial in 1972. International guidelines for the use of the treatment lack clarity regarding the recommended type of corticosteroid and the gestational window of treatment administration. Furthermore, clinical trials cited in the most recent Cochrane Review have limitations which should be taken into account when considering the use of antenatal corticosteroids in clinical practice. Lastly, there is limited evidence regarding the long-term effects on the different fetal organ systems exposed in utero, particularly when the timing of corticosteroid administration is sub-optimal. CONCLUSION Further investigations are urgently needed to determine the most safe and effective treatment regimen for antenatal corticosteroids, particularly regarding the type of corticosteroid and optimal gestational window of administration. A clear consensus on the use of this common treatment could maximise the benefits and minimise potential harms to offspring.
Collapse
Affiliation(s)
- Lenka Hrabalkova
- Tommy's Centre for Maternal and Fetal Health at the MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Matthew W Kemp
- Tohoku University Hospital, Sendai, Miyagi, Japan.,Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Australia
| | - Sarah J Stock
- Tommy's Centre for Maternal and Fetal Health at the MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom.,Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Kemp MW, Saito M, Schmidt AF, Usuda H, Watanabe S, Sato S, Hanita T, Kumagai Y, Takahashi T, Musk GC, Furfaro L, Stinson L, Fee EL, Eddershaw PJ, Payne MS, Smallwood K, Bridges J, Newnham JP, Jobe AH. The duration of fetal antenatal steroid exposure determines the durability of preterm ovine lung maturation. Am J Obstet Gynecol 2020; 222:183.e1-183.e9. [PMID: 31494126 DOI: 10.1016/j.ajog.2019.08.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/14/2019] [Accepted: 08/23/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Antenatal corticosteroids (ACS) are the standard of care for maturing the fetal lung and improving outcomes for preterm infants. Antenatal corticosteroid dosing remains nonoptimized, and there is little understanding of how different treatment-to-delivery intervals may affect treatment efficacy. The durability of a lung maturational response is important because the majority of women treated with antenatal corticosteroids do not deliver within the widely accepted 1- to 7-day window of treatment efficacy. OBJECTIVE We used a sheep model to test the duration of fetal exposures for efficacy at delivery intervals from 1 to 10 days. MATERIALS AND METHODS For infusion studies, ewes with single fetuses were randomized to receive an intravenous bolus and maintenance infusion of betamethasone phosphate to target 1-4 ng/mL fetal plasma betamethasone for 36 hours, with delivery at 2, 4 ,or 7 days posttreatment or sterile saline solution as control. Animals receiving the clinical treatment were randomised to receive either a single injection of 0.25 mg/kg with a 1:1 mixture of betamethasone phosphate + betamethasone acetate with delivery at either 1 or 7 days posttreatment, or 2 treatments of 0.25 mg/kg betamethasone phosphate + betamethasone acetate spaced at 24 hours (giving ∼48 hours of fetal steroid exposure) with delivery at 2, 5, 7, or 10 days posttreatment. Negative control animals were treated with saline solution. All lambs were delivered at 121 ± 3 days gestational age and ventilated for 30 minutes to assess lung function. RESULTS Preterm lambs delivered at 1 or 2 days post-antenatal corticosteroid treatment had significant improvements in lung maturation for both intravenous and single-dose intramuscular treatments. After 2 days, the efficacy of 36-hour betamethasone phosphate infusions was lost. The single dose of 1:1 betamethasone phosphate + betamethasone acetate also was ineffective at 7 days. In contrast, animals treated with 2 doses had significant improvements in lung maturation at 2, 5, and 7 days, with treatment efficacy reduced by 10 days. CONCLUSION In preterm lambs, the durability of antenatal corticosteroids treatment depends on the duration of fetal exposure and is independent of the intravenous or intramuscular maternal route of administration. For acute 24- to 48-hour posttreatment deliveries, a 24-hour fetal antenatal corticosteroids exposure was sufficient for lung maturation. A fetal exposure duration of at least 48 hours was necessary to maintain long-term treatment durability. A single-dose ACS treatment should be sufficient for women delivering within <48 hours of antenatal corticosteroids treatment.
Collapse
Affiliation(s)
- Matthew W Kemp
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan; School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia.
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | | | - Haruo Usuda
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Shimpei Watanabe
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shinichi Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Takushi Hanita
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Yusaku Kumagai
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Tsukasa Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Gabrielle C Musk
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Lucy Furfaro
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Lisa Stinson
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | | | - Matthew S Payne
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Kiara Smallwood
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - James Bridges
- Cincinnati Children's Hospital Medical Centre, Cincinnati, OH
| | - John P Newnham
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Cincinnati Children's Hospital Medical Centre, Cincinnati, OH
| |
Collapse
|
34
|
Kumagai Y, Kemp MW, Yaegashi N, Saito M. Contemporary Challenges and Developments: Antenatal Corticosteroid Therapy. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2019. [DOI: 10.1007/s13669-019-00270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
35
|
Xing Y, Zhang W, Zhao H, Shen Z, Liang W, Zhou J, Shi L, Chen J, Zhong X, Tang S. Multi‑organ assessment via a 9.4‑Tesla MRS evaluation of metabolites during the embryonic development of cleft palate induced by dexamethasone. Mol Med Rep 2019; 20:3326-3336. [PMID: 31432193 PMCID: PMC6755240 DOI: 10.3892/mmr.2019.10558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 06/19/2019] [Indexed: 02/05/2023] Open
Abstract
The aim of the present study was to determine the association between maternal metabolism and development of the fetal palate, and to suggest a potential non‑invasive prenatal diagnostic method for fetal cleft palate (CP). Dexamethasone (DXM) was used to create a CP mouse model. A 9.4‑Tesla (T) magnetic resonance spectroscopy (MRS) imager was used to measure an array of metabolites in the maternal serum, placental tissue, amniotic fluid and fetal palates. Multivariate statistical analysis was performed using SIMCA‑P 14.1 software. Following DXM treatment, variations were detected in multiple metabolites in the female mice and their fetuses based on 9.4T MRS. It was indicated that in the experimental group during CP formation, leucine, valine, creatine, acetate and citrate levels in the palatal tissue were lower, whereas lactate, alanine, proline/inositol and glutamate‑containing metabolite levels were higher, compared with the levels in the control group. In placental tissue and amniotic fluid, succinate and choline levels were lower in the experimental group. The relative concentrations of cholesterol and lipids in palatal tissues from mice treated with DXM were higher compared with the concentrations in tissues from mice in the control group, with the exception of (CH2)n lipids. In the placental tissue, the alteration in cholesterol level exhibited the opposite trend. Lipid levels for the different lipid forms varied and most of them were unsaturated lipids.
Collapse
Affiliation(s)
- Yue Xing
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Hanxing Zhao
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Zhiwei Shen
- Department of Medical Imaging, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Weijie Liang
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, Central South University Third Xiangya Hospital, Changsha, Hunan 410013, P.R. China
| | - Lungang Shi
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jiasheng Chen
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiaoping Zhong
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Dr Shijie Tang, Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia Road, Shantou, Guangdong 515041, P.R. China, E-mail: ;
| |
Collapse
|
36
|
Schmidt AF, Kemp MW, Milad M, Miller LA, Bridges JP, Clarke MW, Kannan PS, Jobe AH. Oral dosing for antenatal corticosteroids in the Rhesus macaque. PLoS One 2019; 14:e0222817. [PMID: 31536601 PMCID: PMC6752828 DOI: 10.1371/journal.pone.0222817] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/06/2019] [Indexed: 02/04/2023] Open
Abstract
Antenatal corticosteroids (ACS) are standard of care for women at risk of preterm delivery, although choice of drug, dose or route have not been systematically evaluated. Further, ACS are infrequently used in low resource environments where most of the mortality from prematurity occurs. We report proof of principle experiments to test betamethasone-phosphate (Beta-P) or dexamethasone-phosphate (Dex-P) given orally in comparison to the clinical treatment with the intramuscular combination drug beta-phosphate plus beta-acetate in a Rhesus Macaque model. First, we performed pharmacokinetic studies in non-pregnant monkeys to compare blood levels of the steroids using oral dosing with Beta-P, Dex-P and an effective maternal intramuscular dose of the beta-acetate component of the clinical treatment. We then evaluated maternal and fetal blood steroid levels with limited fetal sampling under ultrasound guidance in pregnant macaques. We found that oral Beta is more slowly cleared from plasma than oral Dex. The blood levels of both drugs were lower in maternal plasma of pregnant than in non-pregnant macaques. Using the pharmacokinetic data, we treated groups of 6–8 pregnant monkeys with oral Beta-P, oral Dex-P, or the maternal intramuscular clinical treatment and saline controls and measured pressure-volume curves to assess corticosteroid effects on lung maturation at 5d. Oral Beta-P improved the pressure-volume curves similarly to the clinical treatment. Oral Dex-P gave more variable and nonsignificant responses. We then compared gene expression in the fetal lung, liver and hippocampus between oral Beta-P and the clinical treatment by RNA-sequencing. The transcriptomes were largely similar with small gene expression differences in the lung and liver, and no differences in the hippocampus between the groups. As proof of principle, ACS therapy can be effective using inexpensive and widely available oral drugs. Clinical dosing strategies must carefully consider the pharmacokinetics of oral Beta-P or Dex-P to minimize fetal exposure while achieving the desired treatment responses.
Collapse
Affiliation(s)
- Augusto F. Schmidt
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | | | - Mark Milad
- Milad Pharmaceutical consulting LLC, Plymouth, Michigan, United States of America
| | - Lisa A. Miller
- California National Primate Research Center, University of California, Davis, Davis, California, United States of America
| | - James P. Bridges
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Michael W. Clarke
- Metabolomics Australia, Centre for Microscopy, Characterization and Analysis, The University of Western Australia, Perth, WA, Australia
| | - Paranthaman S. Kannan
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Alan H. Jobe
- Department of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
- University of Western Australia, Perth, Australia
- * E-mail:
| |
Collapse
|
37
|
Dosing and formulation of antenatal corticosteroids for fetal lung maturation and gene expression in rhesus macaques. Sci Rep 2019; 9:9039. [PMID: 31227752 PMCID: PMC6588577 DOI: 10.1038/s41598-019-45171-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/15/2019] [Indexed: 02/07/2023] Open
Abstract
Antenatal corticosteroids (ANS) are the major intervention to decrease respiratory distress syndrome and mortality from premature birth and are standard of care. The use of ANS is expanding to include new indications and gestational ages, although the recommended dosing was never optimized. The most widely used treatment is two intramuscular doses of a 1:1 mixture of betamethasone-phosphate (Beta-P) and betamethasone-acetate (Beta-Ac) - the clinical drug. We tested in a primate model the efficacy of the slow release Beta-Ac alone for enhancing fetal lung maturation and to reduce fetal corticosteroid exposure and potential toxic effects. Pregnant rhesus macaques at 127 days of gestation (80% of term) were treated with either the clinical drug (0.25 mg/kg) or Beta-Ac (0.125 mg/kg). Beta-Ac alone increased lung compliance and surfactant concentration in the fetal lung equivalently to the clinical drug. By transcriptome analyses the early suppression of genes associated with immune responses and developmental pathways were less affected by Beta-Ac than the clinical drug. Promoter and regulatory analysis prediction identified differentially expressed genes targeted by the glucocorticoid receptor in the lung. At 5 days the clinical drug suppressed genes associated with neuronal development and differentiation in the fetal hippocampus compared to control, while low dose Beta-Ac alone did not. A low dose ANS treatment with Beta-Ac should be assessed for efficacy in human trials.
Collapse
|
38
|
Abstract
Treatment with antenatal corticosteroids (ACS) is standard of care for women at risk of preterm birth between 24 and 34 weeks' gestation. ACS are increasingly used for other indications, including threated or indicated late preterm birth, elective cesarean, and in at-risk pregnancies for periviable gestations. However, the various drugs and doses being used worldwide have not been rigorously evaluated to optimize clinical responses and to minimize potential risks. Translational research in animal models indicate that a constant, low concentration fetal exposure to ACS is sufficient for lung maturation, resulting in lower fetal exposures. Clinical trials to develop dosing strategies with inexpensive and widely available drugs could promote greater use in low resource environments.
Collapse
Affiliation(s)
- Matthew W Kemp
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan; School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia.
| | - Augusto F Schmidt
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue
- MLC 7029, Cincinnati, OH, 45229, USA.
| | - Alan H Jobe
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue
- MLC 7029, Cincinnati, OH, 45229, USA.
| |
Collapse
|
39
|
McKinlay CJ, Manley BJ. Antenatal and postnatal corticosteroids: A swinging pendulum. Semin Fetal Neonatal Med 2019; 24:167-169. [PMID: 31147160 DOI: 10.1016/j.siny.2019.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Christopher Jd McKinlay
- Liggins Institute, University of Auckland, Auckland, New Zealand; Kidz First Neonatal Care, Counties Manukau Health, Auckland, New Zealand.
| | - Brett J Manley
- Neonatal Services and Newborn Research Centre, The Royal Women's Hospital, Melbourne, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Australia; Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
40
|
Abstract
Pregnancy profoundly alters a woman's physiology. These changes alter drug absorption, distribution, metabolism, and elimination and emphasize the pharmacologic complexity of pregnancy. They also emphasize the dangers of extrapolating pharmacologic expectations from nonpregnant populations to pregnant women and their fetuses. Although concerns about fetal safety have historically limited pharmacokinetic studies during pregnancy, it is important to recognize that many medications are clinically indicated for various maternal or fetal conditions, and it is particularly important that these therapies be evidence-based with appropriate study, including short-term and long-term outcomes data.
Collapse
Affiliation(s)
- Robert M Ward
- Pediatrics, Pediatric Clinical Pharmacology, University of Utah, University of Utah School of Medicine, 295 Chipeta Way, Salt Lake City, UT 84108, USA.
| | - Michael W Varner
- Department of Obstetrics and Gynecology, University of Utah, 30 North 1900 East, Room 2B 200, Salt Lake City, UT 84132, USA
| |
Collapse
|
41
|
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: 4.0] [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
|
42
|
Schmidt AF, Jobe AH, Kannan PS, Bridges JP, Newnham JP, Saito M, Usuda H, Kumagai Y, Fee EL, Clarke M, Kemp MW. Oral antenatal corticosteroids evaluated in fetal sheep. Pediatr Res 2019; 86:589-594. [PMID: 31365919 PMCID: PMC6848022 DOI: 10.1038/s41390-019-0519-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/24/2019] [Accepted: 06/08/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The use of antenatal corticosteroids (ACS) in low-resource environments is sporadic. Further, drug choice, dose, and route of ACS are not optimized. We report the pharmacokinetics and pharmacodynamics of oral dosing of ACS using a preterm sheep model. METHODS We measured pharmacokinetics of oral betamethasone-phosphate (Beta-P) and dexamethasone-phosphate (Dex-P) using catheterized pregnant sheep. We compared fetal lung maturation responses of oral Beta-P and Dex-P to the standard treatment with 2 doses of the i.m. mixture of Beta-P and betamethasone-acetate at 2, 5, and 7 days after initiation of ACS. RESULTS Oral Dex-P had lower bioavailability than Beta-P, giving a lower maximum maternal and fetal concentration. A single oral dose of 0.33 mg/kg of Beta-P was equivalent to the standard clinical treatment assessed at 2 days; 2 doses of 0.16 mg/kg of oral Beta-P were equivalent to the standard clinical treatment at 7 days as assessed by lung mechanics and gas exchange after preterm delivery and ventilation. In contrast, oral Dex-P was ineffective because of its decreased bioavailability. CONCLUSION Using a sheep model, we demonstrate the use of pharmacokinetics to develop oral dosing strategies for ACS. Oral dosing is feasible and may facilitate access to ACS in low-resource environments.
Collapse
Affiliation(s)
- Augusto F. Schmidt
- 0000 0001 2179 9593grid.24827.3bDivision of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH USA ,0000 0004 1936 8606grid.26790.3aPresent Address: Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL USA
| | - Alan H. Jobe
- 0000 0001 2179 9593grid.24827.3bDivision of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH USA
| | - Paranthaman S. Kannan
- 0000 0001 2179 9593grid.24827.3bDivision of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH USA
| | - James P. Bridges
- 0000 0001 2179 9593grid.24827.3bDivision of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH USA
| | - John P. Newnham
- 0000 0004 1936 7910grid.1012.2Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia
| | - Masatoshi Saito
- 0000 0004 1936 7910grid.1012.2Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,0000 0004 0641 778Xgrid.412757.2Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574 Japan
| | - Haruo Usuda
- 0000 0004 1936 7910grid.1012.2Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,0000 0004 0641 778Xgrid.412757.2Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574 Japan
| | - Yusaku Kumagai
- 0000 0004 0641 778Xgrid.412757.2Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574 Japan
| | - Erin L. Fee
- 0000 0004 1936 7910grid.1012.2Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia
| | - Michael Clarke
- 0000 0004 1936 7910grid.1012.2Metabolomics Australia, Centre for Microscopy, Characterization and Analysis, The University of Western Australia, Perth, WA Australia
| | - Matthew W. Kemp
- 0000 0004 1936 7910grid.1012.2Division of Obstetrics and Gynecology, The University of Western Australia, Perth, WA Australia ,0000 0004 0641 778Xgrid.412757.2Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi 980-8574 Japan ,0000 0004 0436 6763grid.1025.6School of Veterinary and Life Sciences, Murdoch University, Perth, WA Australia
| |
Collapse
|
43
|
Morsi A, DeFranco D, Witchel SF. The Hypothalamic-Pituitary-Adrenal Axis and the Fetus. Horm Res Paediatr 2018; 89:380-387. [PMID: 29874660 DOI: 10.1159/000488106] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 02/28/2018] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids (GCs), cortisol in humans, influence multiple essential maturational events during gestation. In the human fetus, fetal hypothalamic-pituitary-adrenal (HPA) axis function, fetal adrenal steroidogenesis, placental 11β- hydroxysteroid dehydrogenase type 2 activity, maternal cortisol concentrations, and environmental factors impact fetal cortisol exposure. The beneficial effects of synthetic glucocorticoids (sGCs), such as dexamethasone and betamethasone, on fetal lung maturation have significantly shifted the management of preterm labor and threatened preterm birth. Accumulating evidence suggests that exposure to sGCs in utero at critical developmental stages can alter the function of organ systems and that these effects may have sequelae that extend into adult life. Maternal stress and environmental influences may also impact fetal GC exposure. This article explores the vulnerability of the fetal HPA axis to endogenous GCs and exogenous sGCs.
Collapse
Affiliation(s)
- Amr Morsi
- Division of Pediatric Endocrinology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donald DeFranco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Selma F Witchel
- Division of Pediatric Endocrinology, Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
44
|
Kemp MW, Jobe AH, Usuda H, Nathanielsz PW, Li C, Kuo A, Huber HF, Clarke GD, Saito M, Newnham JP, Stock SJ. Efficacy and safety of antenatal steroids. Am J Physiol Regul Integr Comp Physiol 2018; 315:R825-R839. [PMID: 29641233 DOI: 10.1152/ajpregu.00193.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antenatal steroids (ANS) are among the most important and widely utilized interventions to improve outcomes for preterm infants. A significant body of evidence demonstrates improved outcomes in preterm infants (24-34 wk) delivered between 1 and 7 days after the administration of a single course of ANS. Moreover, ANS have the advantage of being widely available, low cost, and easily administered via maternal intramuscular injection. The use of ANS to mature the fetal lung is, however, not without contention. Their use in pregnancy is not FDA approved, and treatment doses and regimens remain largely unoptimized. Their mode of use varies considerably between countries, and there are lingering concerns regarding the safety of exposing the fetus to high doses of exogenous steroids. A significant proportion of women deliver outside the 1- to 7-day therapeutic window after ANS treatment, and this delay may be associated with an increased risk of adverse outcomes for both mother and baby. Today, animal-based studies are one means by which key questions of dosing and safety relating to ANS may be resolved, allowing for further refinement(s) of this important therapy. Complementary approaches using nonhuman primates, sheep, and rodents have provided invaluable advances to our understanding of how exogenous steroid exposure impacts fetal development. Focusing on these three major model groups, this review highlights the role of three key animal models (sheep, nonhuman primates, rodents) in the development of antenatal steroid therapy, and provides an up-to-date synthesis of current efforts to refine this therapy in an era of personalised medicine.
Collapse
Affiliation(s)
- Matthew W Kemp
- Division of Obstetrics and Gynaecology, University of Western Australia , Perth , Australia
- Tohoku University Hospital, Sendai, Miyagi , Japan
| | - Alan H Jobe
- Division of Obstetrics and Gynaecology, University of Western Australia , Perth , Australia
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Centre , Cincinnati, Ohio
| | - Haruo Usuda
- Division of Obstetrics and Gynaecology, University of Western Australia , Perth , Australia
- Tohoku University Hospital, Sendai, Miyagi , Japan
| | | | - Cun Li
- Department of Animal Science, University of Wyoming , Laramie, Wyoming
| | - Anderson Kuo
- Department of Radiology, University of Texas Health Science Center San Antonio , San Antonio, Texas
| | - Hillary F Huber
- Department of Animal Science, University of Wyoming , Laramie, Wyoming
| | - Geoffrey D Clarke
- Department of Radiology, University of Texas Health Science Center San Antonio , San Antonio, Texas
| | - Masatoshi Saito
- Division of Obstetrics and Gynaecology, University of Western Australia , Perth , Australia
- Tohoku University Hospital, Sendai, Miyagi , Japan
| | - John P Newnham
- Division of Obstetrics and Gynaecology, University of Western Australia , Perth , Australia
| | - Sarah J Stock
- Division of Obstetrics and Gynaecology, University of Western Australia , Perth , Australia
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
45
|
The efficacy of antenatal steroid therapy is dependent on the duration of low-concentration fetal exposure: evidence from a sheep model of pregnancy. Am J Obstet Gynecol 2018; 219:301.e1-301.e16. [PMID: 29758177 DOI: 10.1016/j.ajog.2018.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Antenatal corticosteroids are among the most important and widely used interventions to improve outcomes for preterm infants. Antenatal corticosteroid dosing regimens remain unoptimized and without maternal weight-adjusted dosing. We, and others, have hypothesized that, once a low concentration of maternofetal steroid exposure is achieved and maintained, the duration of the steroid exposure determines treatment efficacy. Using a sheep model of pregnancy, we tested the relationship among steroid dose, duration of exposure, and treatment efficacy. OBJECTIVE The study was conducted to investigate the relative importance of duration and magnitude of fetal corticosteroid exposure to mature the preterm fetal ovine lung. STUDY DESIGN Ewes with single fetuses at 120 days gestation received an intravenous bolus (loading dose) followed by a maintenance infusion of betamethasone phosphate to target 12-hour fetal plasma betamethasone concentrations of (1) 20 ng/mL, (2) 10 ng/mL, or (3) 2 ng/mL. In a subsequent experiment, fetal plasma betamethasone concentrations were targeted at 2 ng/mL for 26 hours. Negative control animals received sterile saline solution. Positive control animals received 2 intramuscular injections of 0.25 mg/kg Celestone Chronodose (betamethasone phosphate + betamethasone acetate) spaced at 24 hours. Preterm lambs were delivered surgically and ventilated 48 hours after treatment commenced. Maternal and fetal plasma betamethasone concentrations were confirmed by mass spectrometry in a parallel study of chronically catheterized, corticosteroid-treated ewes and fetuses. RESULTS The loading and maintenance doses were achieved and maintained the desired fetal plasma betamethasone concentrations of approximately 20, 10, and 2 ng/mL for 12 hours. Compared with the 12-hour infusion-treated animals, lambs from the positive control (2 intramuscular doses of 0.25 mg/kg Celestone Chronodose) group had the greatest functional lung maturation (compliance, gas exchange, arterial pH) and molecular evidence of maturation (glucocorticoid receptor signaling activation), despite having maximum fetal plasma betamethasone concentrations 2.5 times lower than animals in the 20 ng/mL betamethasone infusion group. Lambs from the 12-hour 2-ng/mL betamethasone infusion group had little functional lung maturation. In contrast, lambs from the 26-hour 2-ng/mL betamethasone infusion group had functional lung maturation equivalent to lambs from the positive control group. CONCLUSION In preterm lambs that were exposed to antenatal corticosteroids, high maternofetal plasma betamethasone concentrations did not correlate with improved lung maturation. The largest and most consistent improvements in lung maturation were in animals that were exposed to either the clinical course of Celestone Chronodose or a low-dose betamethasone phosphate infusion to achieve a fetal plasma betamethasone concentration of approximately 2 ng/mL for 26 hours. The duration of low-concentration maternofetal steroid exposure, not total dose or peak drug exposure, is a key determinant for antenatal corticosteroids efficacy. These findings underscore the need to develop an optimized steroid dosing regimen that may improve both the efficacy and safety of antenatal corticosteroids therapy.
Collapse
|
46
|
Jobe AH, Goldenberg RL. Antenatal corticosteroids: an assessment of anticipated benefits and potential risks. Am J Obstet Gynecol 2018; 219:62-74. [PMID: 29630886 DOI: 10.1016/j.ajog.2018.04.007] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 12/20/2022]
Abstract
Antenatal corticosteroids are standard of care for pregnancies at risk of preterm delivery between 24-34 weeks' gestational age. Recent trials demonstrate modest benefits from antenatal corticosteroids for late preterm and elective cesarean deliveries, and antenatal corticosteroids for periviable deliveries should be considered with family discussion. However, many women with threatened preterm deliveries receive antenatal corticosteroids but do not deliver until >34 weeks or at term. The net effect is that a substantial fraction of the delivery population will be exposed to antenatal corticosteroids. There are gaps in accurate assessments of benefits of antenatal corticosteroids because the randomized controlled trials were performed prior to about 1990 in pregnancies generally >28 weeks. The care practices for the mother and infant survival were different than today. The randomized controlled trial data also do not strongly support the optimal interval from antenatal corticosteroid treatment to delivery of 1-7 days. Epidemiology-based studies using large cohorts with >85% of at-risk pregnancies treated with antenatal corticosteroids probably overestimate the benefits of antenatal corticosteroids. Although most of the prematurity-associated mortality is in low-resource environments, the efficacy and safety of antenatal corticosteroids in those environments remain to be evaluated. The short-term benefits of antenatal corticosteroids for high-risk pregnancies in high-resource environments certainly justify antenatal corticosteroids as few risks have been identified over many years. However, cardiovascular and metabolic abnormalities have been identified in large animal models and cohorts of children exposed to antenatal corticosteroids that are consistent with fetal programming for adult diseases. These late effects of antenatal corticosteroids suggest caution for the expanded use of antenatal corticosteroids beyond at-risk pregnancies at 24-34 weeks. A way forward is to develop noninvasive fetal assessments to identify pregnancies across a wider gestational age that could benefit from antenatal corticosteroids.
Collapse
|
47
|
Visconti K, Senthamaraikannan P, Kemp MW, Saito M, Kramer BW, Newnham JP, Jobe AH, Kallapur SG. Extremely preterm fetal sheep lung responses to antenatal steroids and inflammation. Am J Obstet Gynecol 2018; 218:349.e1-349.e10. [PMID: 29274832 DOI: 10.1016/j.ajog.2017.12.207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/27/2017] [Accepted: 12/14/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND The efficacy of antenatal steroids for fetal lung maturation in the periviable period is not fully understood. OBJECTIVE We sought to determine the lung maturational effects of antenatal steroids and inflammation in early gestation sheep fetuses, similar to the periviable period in human beings. STUDY DESIGN Date-mated ewes with singleton fetuses were randomly assigned to 1 of 4 treatment groups (n = 8/group): (1) maternal intramuscular injection of betamethasone; (2) intraamniotic lipopolysaccharide; (3) betamethasone + lipopolysaccharide; and (4) intraamniotic + intramuscular saline (controls). Fetuses were delivered surgically 48 hours later at 94 days' gestation (63% term gestation) for comprehensive evaluations of lung maturation, and lung and systemic inflammation. RESULTS Relative to controls, first, betamethasone increased the fetal lung air space to mesenchymal area ratio by 47% but did not increase the messenger RNAs for the surfactant proteins-B and -C that are important for surfactant function or increase the expression of pro-surfactant protein-C in the alveolar type II cells. Second, betamethasone increased expression of 1 of the 4 genes in surfactant lipid synthetic pathways. Third, betamethasone increased genes involved in epithelium sodium channel transport, but not sodium-potassium adenosine triphosphatase or Aquaporin 5. Fourth, lipopolysaccharide increased proinflammatory genes in the lung but did not effectively recruit activated inflammatory cells. Last, betamethasone incompletely suppressed lipopolysaccharide-induced lung inflammation. In the liver, betamethasone when given alone increased the expression of serum amyloid A3 and C-reactive protein messenger RNAs. CONCLUSION Compared the more mature 125-day gestation sheep, antenatal steroids do not induce pulmonary surfactants during the periviable period, indicating a different response.
Collapse
|