Hostname: page-component-5cf477f64f-54txb Total loading time: 0 Render date: 2025-03-29T17:50:35.889Z Has data issue: false hasContentIssue false

A review of the potential off-target effects of antenatal steroid exposures on fetal development

Published online by Cambridge University Press:  26 March 2025

Sean W.D. Carter*
Affiliation:
Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore King Edward Memorial Hospital, Perth, Western Australia, Australia Women and Infants Research Foundation, Perth, Western Australia, Australia
Matthew W. Kemp
Affiliation:
Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore Women and Infants Research Foundation, Perth, Western Australia, Australia Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
*
Corresponding author: Sean W.D. Carter; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Antenatal steroids (ANS) are one of the most widely prescribed medications in pregnancy, being administered to women at risk of preterm delivery. In the setting of preterm delivery at or below 35 weeks’ gestation, systematic review data show ANS reduce perinatal morbidity and mortality, primarily by promoting fetal lung maturation. However, with the expanding use of this intervention has come a growing appreciation for the potential off-target, adverse effects of ANS therapy on wider fetal development. We undertook a narrative literature review of the animal and clinical literature to assess current evidence for adverse effects of ANS exposure and fetal development. This review presents a summary of the evidence relating to the potential for wide-ranging, off-target, adverse effects of ANS therapy on fetal development and programming. We highlight an urgent need for further animal and clinical studies investigating the effects of ANS on the fetal immune, cardiovascular, renal and hepatic systems given a current sparsity of evidence. We also strongly suggest an emphasis on open disclosure, discussion and education of clinicians and patients with regard to the potential benefits and risks of ANS therapy, particularly in late preterm and term gestations where infants derive relatively few benefits from these drugs. We also propose further studies on the optimisation of ANS therapy through improved patient selection and improved dosing regimens based on a pharmacokinetic-pharmacodynamic informed understanding of ANS action on the fetal lung.

Topics structure

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press in association with The International Society for Developmental Origins of Health and Disease (DOHaD)

Introduction

Antenatal steroids (ANS) are one of the most widely prescribed off-label medications in obstetric practice today. Since the first randomised control trial (RCT) of ANS reported in 1972 by Liggins and Howie, these drugs have become routinely administered to women at risk of preterm birth (birth <37 completed weeks’ gestation) due to their recognised benefits of reducing perinatal morbidity and mortality, primarily by promoting fetal lung maturation. Reference McGoldrick, Stewart, Parker and Dalziel1– Reference Roberts, Brown, Medley and Dalziel3 Additional anecdotal evidence suggests an improvement in cardiovascular stability, although these effects are difficult to isolate from improved lung maturation in the clinical setting. Of interest, however, is that the original Auckland Steroid Trial (AST) by Liggins and Howie only reported on a small portion of the entire cohort of patients (n = 287 of 1115 patients) when they published their findings of a significant reduction in respiratory distress syndrome (RDS) in those born under 32 weeks’ gestation and a survival benefit only under 30 weeks’ gestation. Reference Liggins and Howie4 Remarkably, it took over 50 years for the findings from the entire AST cohort (n = 1115) to be published, which revealed a reduction in RDS, but no survival benefit of ANS at any gestation nor a reduction in the rates of intraventricular haemorrhage and no benefit of doubling the ANS dose. Reference Walters, Lin, Crowther, Gamble, Dalziel and Harding5 At present, ANS dosing remains largely unchanged around the world since the first publication of the AST.

There are two fluorinated steroids, each with distinct pharmacokinetic profiles in common use for ANS therapy: betamethasone (either solely as the rapidly released phosphate ester or as the phosphate ester in combination with the slow release acetate ester) or rapidly released dexamethasone phosphate. Reference Jobe, Kemp, Schmidt, Takahashi, Newnham and Milad6,Reference Jobe, Milad, Peppard and Jusko7 Today, all ANS are administered via maternal intramuscular injection. Hydrocortisone is not used for this indication due to its very short half-life and placental inactivation. Despite the findings of the 1994 NIH Consensus Development Panel on ANS administration, Reference Gilstrap, Christensen and Clewell8 sizable variation exists worldwide regarding ANS dosing regimens.

Improved preterm lung function is likely achieved by steroids crossing the placenta and activating the glucocorticoid receptor (GR) in fetal lung tissue resulting in both genomic and non-genomic downstream signalling. Reference Kemp, Newnham, Challis, Jobe and Stock9 The exact molecular mechanisms by which ANS achieve accelerated preterm lung maturation are not yet fully understood. However, there is evidence that ANS therapy results in reduced mesenchymal and epithelial cell proliferation, increased fluid clearance, extracellular remodelling with increased elastin expression and thinning of the lung septae, as well as enhanced maturation of alveolar type I and type II epithelial cells with the initiation of surfactant production. Reference Bridges, Sudha and Lipps10 These changes all contribute to functional maturation via improved lung expansion and gas exchange. However, concerns have also been raised about the potential pluripotent, off-target, adverse effects of ANS exposure on wider fetal development beyond lung maturation, given that corticosteroids may impact the regulation of as much as 20% of the human transcriptome (Fig. 1). Reference Kemp, Newnham, Challis, Jobe and Stock9

Figure 1. Graphical abstract of the short and long term off target effects of antenatal corticosteroid therapy on fetal development. Created in BioRender.com.

The seventh article of the Declaration of Helsinki notes that “Even the best proven interventions must be evaluated continually through research for their safety, effectiveness, efficiency, accessibility and quality”. 11 ANS therapy is an obvious candidate for careful evaluation according to these principles given its extensive use, poorly understood mode of action and lack of dose optimisation. This is especially true given that, since the introduction of ANS into clinical practice in 1972 and widespread global use from the 1990s, there have been significant other advances in the obstetric management of high-risk patients and in the neonatal care of preterm infants including the use of endogenous surfactant and improved neonatal ventilation techniques. Reference Lawn, Davidge and Paul12,Reference Usuda, Carter and Takahashi13 Indeed, the vast majority of improvements in perinatal outcomes (perhaps with the exception of peri-viable deliveries) seen today were achieved prior to the widespread introduction of ANS therapy (i.e. post-1994). Reference Lawn, Davidge and Paul12 These advances in neonatal care have contributed in their own right to a reduction in preterm morbidity and mortality irrespective of whether an infant has been exposed to ANS. As such, it is important that, rather than solely focusing on short-term outcomes (i.e. RDS risk), there is consideration given to long-term outcomes such as neurodevelopment and adult chronic disease risk.

It is also important to assess benefits and risks as a function of gestational age. This is important due to the increased use of ANS at peri-viable gestations (with clear survival benefits) and in older or late preterm gestations (>35 weeks’ gestation) where the respiratory and survival benefits of ANS are, at best, far more modest. For example, based on systematic review data, the number needed to treat to prevent one case of RDS at <34 weeks’ gestation is 19, compared to 55 at >35 weeks’ gestation and 106 at term prior to Elective Lower Uterine Segment Caesarean Section. Reference Takahashi, Jobe and Fee14 Supporting these observations are the findings of the ALPS trial, which demonstrated no significant reduction in RDS risk with ANS compared to placebo after 34 weeks’ gestation. Reference Gyamfi-Bannerman and Thom15 Of further concern is that a large proportion (up to 44%) of infants who are exposed to ANS due to an incorrect assessment of preterm birth risk go on to deliver at term. Reference Rodriguez, Wang, Ali Khan, Cartwright, Gissler and Järvelin16 Thereby both mother and fetus are exposed to a therapy conveying them no benefit but potential harms.

In light of the above, this review examined the short- and long-term, off-target, developmental implications of ANS exposure by examining both animal and clinical studies.

Methods

We performed a narrative review of the animal and clinical literature focusing on the short- and long-term effects of ANS on fetal development and the origins of disease. Search terms included “antenatal steroids”, “glucocorticoid”, “synthetic glucocorticoids”, “fetal”, “neonatal”, “infant”, “childhood”, “adult”, “development”, “respiratory”, “lung maturation”, “cardiovascular”, “brain”, “neurodevelopment”, “programming”, “hippocampus”, “renal”, “kidney”, “hepatic”, “liver”, “metabolic”, “immune”, “HPA axis”, “animal”, “human” in the following databases: SCOPUS, PUBMED, MEDLINE, EMBASE and Google Scholar. Searches were conducted between July and October 2024. Data from approximately 200 papers was reviewed with randomized control trials, large animal studies (particularly non-human primates) and systematic reviews being prioritised. A focus was placed on literature from 2010 to 2024, with the inclusion of articles outside this range for historically important studies or those addressing specific knowledge gaps. Articles included in this review were full-text, English language papers.

Results and discussion

Neurodevelopmental and behavioural outcomes

Multiple factors predispose the developing fetal brain to short and long-term development risk from ANS exposure. Firstly, endogenous glucocorticoids such as cortisol are integral to driving normal neurodevelopment by inducing remodelling, programmed apoptosis and proliferation of axons and dendrites in a time-critical manner. Reference Seckl and Meaney17 Animal models (rodents) have demonstrated that the placental expression of 11β-HSD-2 (which rapidly converts active cortisol to its inactive form) is highly expressed in midgestation, potentially protecting the brain from premature remodelling effects of endogenous glucocorticoids (cortisol). Reference Seckl and Meaney17 However, at the end of midgestation, placental 11β-HSD-2 is dramatically reduced in both mice and rats leading to cortisol-induced terminal neuronal differentiation as described above. Reference Seckl and Meaney17 Importantly, unlike cortisol, the exogenous glucocorticoids, dexamethasone and betamethasone are resistant to 11β-HSD-2 and thus readily cross the placenta in their active form to activate the GR in fetal tissues, including the developing brain. Reference Seckl and Meaney17 Secondly, evidence suggests that the fetal hippocampus and hypothalamus both have a high density of GR expression making these structures potentially susceptible to off-target effects of ANS. Reference Seckl and Meaney17Reference Whitelaw and Thoresen20 Furthermore, both clinical and animal models of chronic stress (a condition marked by chronic glucocorticoid elevation) and clinical depression have demonstrated downregulation of GR expression in the hippocampus on pathological examination. These findings indicate an important link between glucocorticoid exposure and long-term behavioural risks. Reference McGowan, Sasaki and D”alessio21 Finally, the third trimester (wherein the majority of ANS exposure occurs) corresponds to a critical stage of fetal neurodevelopment, where there is a dramatic increase in myelination of axons and occipital white matter as well as neural synapse formation. Reference Connors, Levitt and Matthews22,Reference Buss, Entringer, Swanson and Wadhwa23 Therefore, a comprehensive understanding of the effects of ANS exposures on the developing fetal brain is of high importance given the potential long-term implications on mental, behavioural and learning issues for exposed individuals.

Animal studies

Taking into account variability in the ontogeny of animal model neurodevelopment (especially with regard to rodent models vs. the human), evidence from a range of animal studies has demonstrated alterations in the fetal nervous system following ANS exposures. Reference Back, Riddle, Dean and Hohimer24 An early study by Uno et al. demonstrated that both term and preterm fetal rhesus monkeys exposed to antenatal dexamethasone had a reduction in neuron density as well as evidence of neurodegeneration characterised by shrinkage of pyramidal neurons and marked atrophy of the zona lucidum within the hippocampus and dentate gyrus. Reference Uno, Lohmiller and Thieme18 These observed neurodegenerative effects were found to be dose-dependent. Reference Uno, Lohmiller and Thieme18 Further, work by Dunlop et al. in a sheep model of pregnancy also demonstrated that repeat administration of ANS delayed myelination of the fetal optic nerve. Reference Dunlop, Archer, Quinlivan, Beazley and Newnham25 The same group then later showed that single and repeat doses of ANS resulted in reduced whole brain weight in the same sheep model of pregnancy. Reference Huang, Beazley, Quinlivan, Evans, Newnham and Dunlop26 Similar findings have been observed in rats and mice exposed to single and repeat antenatal dexamethasone demonstrating a dose-dependent reduction in brain weight as well as evidence of reduced neurogenesis within the forebrain, hippocampus and cortex. Reference Kanagawa, Tomimatsu and Hayashi27Reference Noorlander, Tijsseling and Hessel29 Studies of ANS therapy (betamethasone) in juvenile baboons have also shown evidence of impaired cognition including decreased motivation in both sexes and sex-specific differences with females demonstrating impaired learning outcomes. Reference Rodriguez, Zürcher, Keenan, Bartlett, Nathanielsz and Nijland30 A similar study in baboons investigated structural brain effects of ANS and showed that repeat ANS exposure resulted in reduced myelination within the corpus callosum, the subcortical and cortical deep white matter. Reference Shields, Thomson, Winter, Coalson and Rees31 Furthermore, evidence of increased astrogliosis indicating possible neuronal injury and damage was also observed in these animals. Reference Shields, Thomson, Winter, Coalson and Rees31 Despite the limitation that many of these studies were conducted on term-born animals, the findings from these non-human primate studies add considerable translational weight to human outcomes, given the similarities in neurodevelopment and cognitive ability. Our group recently demonstrated that preterm lambs delivered at 122 ± 2 d gestation (term = 150 d gestation) exposed to either dexamethasone or betamethasone had differential gene expression in the fetal hippocampus related to neuropsychiatric and neurostructural disorders. Reference Carter, Fee and Usuda32 Additionally, lambs exposed to dexamethasone at both a clinical dose (utilised as a first-line treatment in Singapore) or significant dose reduction (75%) had upregulation of transcriptomic pathways within the hippocampus related to neurodegenerative disorders such as Parkinson’s, Alzheimer’s and Prion disease, a profile which was not seen in betamethasone exposed animals. Reference Carter, Fee and Usuda32 Schmidt et al. reported similar findings of abnormal neurodevelopment in rhesus macaques with ANS (dexamethasone or betamethasone) exposure resulting in alteration of the fetal hippocampal transcriptome, notably in genes related to synaptic transmission, neurogenesis and neuronal maturation. Reference Schmidt, Kannan and Bridges33,Reference Schmidt, Schnell and Eaton34 Other studies of ANS exposure on pregnant guinea pigs have shown altered DNA methylation patterns, altered GR binding and transcriptomic profiles in the fetal hippocampus. Reference Crudo, Petropoulos and Suderman35Reference Sasaki, Eng, Lee, Kostaki and Matthews37 These findings suggest the potential for long-term adverse neurodevelopmental changes with ANS exposure. Concerningly many of these changes were seen across multiple generations suggesting that ANS also exert epigenetic neurodevelopmental influences on future offspring. Reference Constantinof, Moisiadis, Kostaki, Szyf and Matthews38,Reference Constantinof, Boureau, Moisiadis, Kostaki, Szyf and Matthews39 .

Clinical studies

Clinical and large population cohort studies examining the impact of ANS on neurodevelopment have also raised concerns. Tijsseling et al. demonstrated a reduction in neuron density within the hippocampi of ANS-exposed neonates (n = 10) compared to non-ANS-exposed neonates (n = 11) that delivered preterm. Reference Tijsseling, Wijnberger and Derks40 Davis et al. reported structural brain changes on MRI examination of healthy, term-born pre-adolescents exposed to betamethasone ANS compared to controls. Reference Davis, Sandman, Buss, Wing and Head41 The MRI findings included bilateral cortical thinning, seen maximally (30% reduction) in the rostral anterior cingulate cortex, which corresponded to an increased risk of affective disorder problems in that cohort. Reference Davis, Sandman, Buss, Wing and Head41

Of additional interest are the findings from two large population cohort studies demonstrating associations between ANS exposure and mental and behavioural issues later in life. The first, from a population cohort study of >674,000 children born in Finland between 2006 and 2017, showed that ANS exposure was associated with a higher risk of mental and behavioural disorders in the entire cohort (12% vs. 6.45%, HR 1.33), in term-born children ( 8.89% vs. 6.31%, HR 1.47) and in preterm-born children (14.59% vs. 10.71%, HR 1.00). Reference Räikkönen, Gissler and Kajantie42 A second population cohort study by Lin et al. of all infants born in Taiwan between 2004 and 2010 (>1,160,000 infants) demonstrated a significant association between ANS exposure and childhood mental disorders in the entire cohort (HR 1.13), those born at term (HR 1.11), in late preterm (34–37 weeks gestation) born children (HR 1.15) and in children born <28 weeks’ gestation (HR 1.22). Reference Lin, Lin, Lin, Hsu and Hsu43 They noted a particular increased risk for disorders such as ADHD and developmental delay. Reference Lin, Lin, Lin, Hsu and Hsu43 Whilst a smaller Chinese cohort study by Tao et al. reported the findings of 1759 infants (710 of which were exposed to ANS) and demonstrated increased risk of cognitive delay measured using the Bayley Scales and Toddler Development in 1-year-olds exposed to dexamethasone ANS after cofounders were taken into account (n = 710). Reference Tao, Du and Chi44 Reassurance regarding the generalisability of these observed associations can be found given that these studies draw from ethnically and geographically distinct populations (i.e. Northern Europe vs. Asia).

The 5 year follow-up data of 80% of the children from the MACS trial (comparing single to repeat course of ANS) demonstrated that term-born children exposed to repeat courses of ANS had a 3.7-fold increased odds of neurosensory disability, which was not observed in the preterm cohort. Reference Asztalos, Murphy and Willan45 However, follow-up of children who were enrolled in a similar trial of repeat compared to single-dose ANS by the Maternal Fetal Medicine Unit network showed no difference in neurocognition (assessed by the Bayley Testing) between groups at age 2–3 years of age. Reference Wapner, Sorokin and Mele46 Similarly, participants from the ALPS trial of ANS versus placebo between 34 and <37 weeks gestation showed no adverse neurocognitive or behavioural outcomes at 6 years of age. However, only 42% of participants were included in the analysis. Reference Gyamfi-Bannerman, Clifton and Tita47 Interestingly, the main findings of the original ALPS trial demonstrated a reduction in transient tachypnoea of the newborn but no significant effect on RDS rates or total duration of hospital stay for infants, suggesting that any benefit from ANS therapy was transient. Reference Gyamfi-Bannerman and Thom15 Concerningly though, this same trial reported a significant increased risk of neonatal hypoglycaemia with ANS exposure, which has been linked to poor neurodevelopmental outcomes later in life including impaired executive and motor functioning. Reference Gyamfi-Bannerman and Thom15,Reference McKinlay, Alsweiler and Anstice48

Follow-up data from 51% of the children enrolled in the ASTECS trial (administration of ANS vs. placebo prior to term elective caesarean section) demonstrated that those children exposed to ANS were more likely (17.7% vs. 8.5% – number needed to harm = 11) to be in the lowest quartile of academic ability at school at age 8–15 years old. Reference Stutchfield, Whitaker, Gliddon, Hobson, Kotecha and Doull49 Similarly, follow-up of children at 6 years of age from the original AST (n = 250 of 318 enrolled at the start of the trial) demonstrated that ANS-exposed children scored lower in tests of cognitive development (Ravens Progressive Matrices Test) and visual memory (particularly in males) indicating evidence of potential cognitive delay. Reference MacArthur, Howie, Dezoete and Elkins50 Supporting these findings is evidence from a cohort of 19-year-olds that demonstrated ANS exposure was associated with poorer IQ scores and more behavioural problems, particularly in individuals with GR subtypes that convey an increased sensitivity to ANS. Reference van der Voorn, Wit, van der Pal, Rotteveel and Finken51

Perhaps the longest follow-up of ANS-exposed infants has been reported by Savoy et al. This longitudinal study followed up 142 infants who were born at extremely low birthweights and either received ANS (n = 63) or were untreated (n = 79). Comparisons were made between groups and also matched to normal birthweight, term-born subjects. Confounders such as socio-economic status, neurosensory impairment and postnatal steroid exposure were considered. Concerningly, at 22–26 years of age, those exposed to ANS had a 3–5-fold increase in the odds of having a diagnosed mood disturbance (anxiety or depression), which persisted in later follow-up at 29–36 years of age. Reference Savoy, Ferro, Schmidt, Saigal and Van Lieshout52 In contrast, a long-term follow-up of data from the original AST by Liggins and Howie of ANS versus placebo was published in 2005. They reported no differences between groups in neurocognition or mental and behavioural disorders; however, it is important to note that only 27% of the original trial participants were followed up and reported on. Reference Dalziel, Lim and Lambert53 Interestingly, a similar population cohort study from New Zealand of all the babies born at a very low birthweight (<1500 g) in 1986 revealed that on follow-up (n = 250 of the original 413 trial participants included), ANS exposure was associated with double the risk of major depression at 26–30 years of age. Reference Darlow, Harris and Horwood54

Two clear themes emerge from our analysis of the literature in relation to the risk of adverse brain effects in association with ANS exposures. Firstly, from animal models (wherein tissue for histological, proteomic and molecular studies are readily available), it is clear that doses of steroids used clinically elicit both acute and lasting changes in transcriptome, proteome and central nervous system architecture and growth. The molecular and structural changes reported are also consistent with observations from human cohort studies. Although it is indeed reasonable to assess rodent and small animal data with caution due to developmental differences, it is more difficult to dismiss extensive data from longer gestation non-human primates or from sheep studies.

Secondly, greater caution than presently exercised is warranted regarding the use of ANS and neurodevelopmental outcomes. The presently available evidence is of sufficient strength that it should be considered (and disclosed) when consenting patients at risk of preterm delivery for ANS therapy, especially over 35 weeks’ gestation.

Hypothalamic pituitary adrenal axis effects

The hypothalamic pituitary adrenal (HPA) axis functions to regulate corticosteroid metabolism and secretion, which is critical for normal homeostasis and the body’s ability to respond to stress. Reference Moisiadis and Matthews55 The hypothalamus produces corticotropin-releasing hormone (CRH) and vasopressin which in turn stimulates adrenocorticotropic hormone (ACTH) release from the anterior pituitary. ACTH then stimulates the release of cortisol from the adrenal cortex. The HPA axis is then regulated via the negative feedback of cortisol on glucocorticoid and mineralocorticoid receptors (MR) within the hippocampus, hypothalamus and anterior pituitary. Endogenous glucocorticoids have wide-ranging, time-critical effects on fetal development, which include fetal growth, cardiovascular, immune, metabolic and neurobehavioral programming. Reference Moisiadis and Matthews55 Unlike the endogenous glucocorticoid cortisol, exogenous glucocorticoids (ANS) have a much higher affinity for the GR and exert a larger and more sustained negative feedback effect on the HPA axis. Reference Sánchez, Young, Plotsky and Insel56 Therefore, disruption of the HPA axis via ANS has the potential to exert wide-ranging acute and chronic fetal programming effects, thereby providing a possible link to the origins of childhood and adult disease related to ANS exposure. Reference Waffarn and Davis57Reference de Vries, Holmes and Heijnis60 Data describing the effects of ANS on the HPA axis from animal and clinical studies are outlined below.

Animal studies

Studies using pregnant sheep have shown both immediate and prolonged maternal and fetal HPA axis suppression with ANS exposure. Suppression of both ACTH and cortisol has been observed in pregnant ewes and fetal lambs exposed to ANS therapies at current clinical doses and in regimens with significant dose reduction (∼75%). Reference Carter, Fee and Usuda32,Reference Usuda, Fee and Carter61Reference Fee, Takahashi and Takahashi64 HPA axis suppression persisted in these animals even when steroid concentration was undetectable in both maternal and fetal plasma circulation. Reference Carter, Fee and Usuda32 Further studies in guinea pigs have also shown dose-dependent suppression of the fetal HPA axis, including downregulation of CRH mRNA in the fetal hypothalamus, providing further evidence that ANS pass the blood-brain barrier and act directly on the developing fetal brain through the GR. Reference McCabe, Marash, Li and Matthews65 A study by de Vries et al. in non-human primates also showed dose-dependent maternal HPA axis suppression in pregnancy and elevated plasma cortisol concentrations to minor stress insults (blood sampling) in the 8-month-old offspring. Reference de Vries, Holmes and Heijnis60 Similarly, Uno et al. demonstrated dose-dependent maternal and fetal HPA axis suppression in rhesus macaques exposed to single and repeat courses of antenatal dexamethasone. Reference Uno, Lohmiller and Thieme18

Clinical studies

Clinical studies have also demonstrated evidence of acute HPA axis dysregulation in ANS-exposed neonates at birth. Findings included reduced basal and stress-induced cortisol concentrations. Reference Waffarn and Davis57,Reference Niwa, Kawai and Kanazawa66Reference Weiss, Keeton, Richoux, Cooper and Niemann69 In contrast, one study of term neonates exposed to ANS demonstrated elevated cortisol concentrations in response to stress. Reference Davis, Waffarn and Sandman70 Multiple other studies have suggested that ANS-induced HPA axis suppression at birth is acute and does not persist past the neonatal period. Reference Gover, Brummelte and Synnes71,Reference Ashwood, Crowther and Willson72 However, one high-quality study clearly demonstrated ongoing blunted basal and stress-induced cortisol concentrations at 12 months of age in those children exposed to ANS. Reference Weiss, Keeton, Richoux, Cooper and Niemann69 Additionally, follow-up studies of children aged 6–11 years Reference Alexander, Rosenlöcher and Stalder73 and adolescents Reference Ilg, Kirschbaum, Li, Rosenlöcher, Miller and Alexander74 exposed to ANS have both demonstrated ongoing HPA axis dysregulation and reprogramming effects including significantly higher cortisol response to stress testing. Reference Alexander, Rosenlöcher and Stalder73,Reference Ilg, Kirschbaum, Li, Rosenlöcher, Miller and Alexander74

The impact of ANS on the maternal HPA axis is also another question. A study in non-pregnant women administered a 6 mg IM dose of either dexamethasone phosphate, betamethasone phosphate or combined betamethasone phosphate and acetate demonstrated cortisol suppression up to 60 hours, 72 hours and >4 days, respectively. Reference Jobe, Milad, Peppard and Jusko7 Notably, the prolonged HPA axis suppression identified in the betamethasone acetate and phosphate group occurred at maternal dosing substantially lower than that used clinically and persisted at plasma concentrations now understood to be sub-therapeutic for fetal lung maturation. McKenna et al. reported the findings of a prospective case-control trial in pregnant patients between 24 and 34 weeks’ gestation, investigating the effects of repeat ANS (at least 2 × weekly course of betamethasone phosphate and acetate, n = 18) on maternal HPA axis suppression, demonstrating secondary adrenal insufficiency after corticotropin stimulation test over 48 hours post-ANS exposure. Reference McKenna, Wittber, Nagaraja and Samuels75 Given that the maternal HPA axis is closely related to fetal HPA axis regulation through the transplacental passage of cortisol and CRH, these findings are of concern. Furthermore, secondary adrenal insufficiency in pregnancy may put patients at risk of falls related to postural hypotension, fatigue and weakness as well as infection related to immune system dysregulation and intrapartum morbidity due to inadequate stress response similar to Addison’s disease (adrenal insufficiency).

Immune system modulation

Previous studies have established that corticosteroids exert dose-dependent, immune-modulating and anti-inflammatory effects Reference Sarnes, Crofford, Watson, Dennis, Kan and Bass76 including increased leukocyte release from bone marrow, impaired leukocyte migration into affected tissue from circulating plasma Reference Nakagawa, Terashima, D’yachkova, Bondy, Hogg and van Eeden77,Reference Waisman, Van Eeden, Hogg, Solimano, Massing and Bondy78 and inhibition of neutrophil apoptosis. Reference Cox79

Animal studies

Our group has demonstrated in pregnant sheep that ANS exposure at both current clinical doses and with 75% dose reduction, results in rapid maternal and fetal plasma neutrophilia and lymphocytopenia. Reference Carter, Fee and Usuda32,Reference Usuda, Fee and Carter61 Similar findings have also been seen in rat models, demonstrating reduced maternal and fetal lymphocyte proliferation and lymphocyte interleukin-2 production after antenatal betamethasone exposure. Reference Murthy and Moya80 Mice exposed to antenatal betamethasone also exhibit significant reductions in thymus size and a large reduction in CD4+/CD8+ T-cell counts at birth. Reference Diepenbruck, Much and Krumbholz81 As the thymus is integral to T-cell production and acts as a primary immune organ, these findings have implications for adaptive immune system functioning.

Clinical studies

Animal data showing ANS-associated immunological disruption are supported by data from clinical studies suggesting potentially serious ANS related immune modulation. For example, Smolders et al. reported ANS-exposed infants had an increased rate of hospital admission due to infection during the first year of life. Reference Smolders-de Haas, Neuvel, Schmand, Treffers, Koppe and Hoeks82 Whilst a secondary analysis of the ACT trial showed a significant association between severe bacterial infection and neonatal mortality in infants exposed to ANS in African nations. Reference Klein, McClure and Colaci83 Furthermore, a Finnish population cohort study demonstrated that ANS exposure was associated with a higher need for treatment of infectious diseases (respiratory, gastrointestinal and urinary tract) up to 4 years of life, in the entire cohort, term and late preterm-born infants compared to non-exposed infants. Reference Räikkönen, Gissler, Kajantie and Tapiainen84 Finally, a recent large cohort study in Taiwan demonstrated ANS exposure was associated with a small but significant increased risk of serious childhood infection. Reference Liang, Tsai, Kuo and Tsai85 At present, there is a lack of evidence on the long-term immune modulation effects of ANS besides the few studies outlined above. However the evidence of increased risk of infection is concerning especially given that many preterm infants are exposed to both intrauterine infection (chorioamnionitis is a leading cause of preterm birth) and ANS. Reference Carter, Neubronner and Su86

Growth restriction

Animal studies

Evidence from studies of multiple different species demonstrates that ANS exposure is associated with reduced intrauterine fetal growth. Extensive work has been performed in sheep models of pregnancy on the effects of ANS on fetal growth. Huang et al. utilised single or repeat weekly courses (up to 4 courses) of 0.5 mg/kg betamethasone (equivalent to double the current standard clinical dose delivered in a single injection) in preterm and term animals with delivery occurring 24 hours after the last dose of ANS. Analyses showed reduced birthweight in all groups compared to saline control. Reference Huang, Beazley, Quinlivan, Evans, Newnham and Dunlop26 The largest reduction in growth was seen with repeat courses and in the term gestation groups exposed to ANS. Reference Huang, Beazley, Quinlivan, Evans, Newnham and Dunlop26 The same group then showed that 1, 2 or 3–4 weekly courses of ANS administration resulted in a 15%, 19% and 27% reduction in birthweight, respectively. Reference Ikegami, Jobe, Newnham, Polk, Willet and Sly87 Similar findings were reported in a subsequent sheep study showing both single and repeat ANS dosing resulted in reduced lung, heart, liver, thymus and kidney weight in addition to a reduced overall birthweight. Reference Newnham, Evans, Godfrey, Huang, Ikegami and Jobe88 Interestingly the study team also administered direct fetal ANS (single and repeat courses) at the same dose which did not affect birthweight, such as that seen in maternal ANS dosing. Although speculative, this may be due to placental export of the drug back across into maternal circulation, fetal hepatic drug metabolism and a lower or shortened overall steroid exposure. Reference Newnham, Evans, Godfrey, Huang, Ikegami and Jobe88 Following on from this earlier work, Takahashi et al. using a reduced dose of antenatal betamethasone (2 × 0.25 mg/kg given 24 hours apart) in a sheep model, demonstrated reduced birthweights of ∼ 15% in preterm lambs exposed to ANS compared to saline control. Reference Takahashi, Fee and Takahashi63 A potential mechanism for ANS-induced growth restriction has been postulated in the same sheep-based model by Usuda et al., who observed that fetal plasma concentrations of insulin-like growth factor 1 (IGF-1) were reduced at birth in lambs exposed to dexamethasone (4 × 6 mg maternal intramuscular injections of dexamethasone phosphate 12 hourly) compared to both saline control and a reduced dose betamethasone regimen (4 × 2 mg maternal intramuscular injections of betamethasone phosphate 12 hourly). Reference Usuda, Fee and Carter61 This is a significant finding as IGF-1 is associated with fetal growth. Reference Netchine, Azzi, Le Bouc and Savage89,Reference Agrogiannis, Sifakis, Patsouris and Konstantinidou90

Rodent models have also been used to explore a potential link between ANS exposure and intrauterine growth restriction. Noting material differences in gestational length between rodents and humans (and thus a proportionally longer exogenous steroid exposure in rodents), Ozdemir et al. showed that mouse pups exposed antenatally to single and repeat courses of betamethasone or dexamethasone had smaller lung, liver and birthweights compared to control. Reference Ozdemir, Guvenal, Cetin, Kaya and Cetin91 Interestingly, dexamethasone pups exhibited a larger reduction in growth than the betamethasone groups. Reference Ozdemir, Guvenal, Cetin, Kaya and Cetin91 A study of single-dose antenatal betamethasone or dexamethasone in rats also showed similar findings including reduced birthweights but only in the dexamethasone group. Whilst at 3 weeks of age (time of weening), both ANS-exposed groups had reduced body weights. Reference Abrantes, Valencia, Bany-Mohammed, Aranda and Beharry92 Another study of rats exposed to a single course of antenatal betamethasone demonstrated reduced birthweights and lower serum glucose, insulin, IGF-1 and leptin levels compared to saline control. Reference Abrantes, Valencia, Bany-Mohammed, Aranda and Beharry93 Furthermore, males exposed to antenatal betamethasone who then mated with control (unexposed) females gave birth to offspring with lower birthweights, suggesting a possible male-linked, transgenerational epigenetic effect from ANS exposure on fetal growth. Reference Abrantes, Valencia, Bany-Mohammed, Aranda and Beharry93

Finally, three different studies in rabbits have investigated the effect of ANS on fetal growth. Sun et al. utilised a reduced dose of betamethasone (0.1 mg/kg) compared to clinically utilised doses and demonstrated that two doses (given 24 hours apart) resulted in a 20% reduction in birthweight, whilst a single dose (48 hours prior to delivery) reduced birthweight by 9.4% but significantly did not functionally mature the fetal lung. Reference Sun, Jobe, Rider and Ikegami94 A study in pregnant rabbits by Pratt et al. utilising 1, 2 or 3 courses of betamethasone (2 × 0.1 mg/kg 24 hours apart) demonstrated a progressive reduction in birthweight with increasing ANS courses, representing a clear dose-dependent effect of ANS on fetal growth. Reference Pratt, Magness, Phernetton, Hendricks, Abbott and Bird95 Interestingly, the same group also gave a later term course (single and double) of betamethasone and showed a greater reduction in birthweight in these animals compared to preterm gestations. Reference Pratt, Magness, Phernetton, Hendricks, Abbott and Bird95 A third study in rabbits utilising the same dose of betamethasone demonstrated that a single course resulted in reduced birthweight that persisted up to 7 weeks of age. Concerningly, these animals also demonstrated evidence of impaired neurocognition on functional testing. Reference Van Der Merwe, Van Der Veeken and Inversetti96

The findings outlined above of reduced fetal growth with ANS exposure are particularly troubling, given they are observed across multiple different species and persist despite significant ANS dose reduction.

Clinical studies

The evidence from clinical studies of ANS-induced growth restriction is unclear, with the most recent Cochrane review of ANS by McGoldrick et al. stating that ANS result in little or no difference in mean birthweight and with an uncertain effect on the incidence of small for gestational age (<10th centile) at birth. Reference McGoldrick, Stewart, Parker and Dalziel1 However, two high-quality clinical trials have demonstrated clear evidence of fetal growth restriction with repeat ANS exposure. Firstly, the ACTORDS trial comparing repeat dosing to single course of ANS demonstrated small reductions in z-scores of birthweight and head circumference in infants exposed to repeat courses of ANS. Reference Crowther, Haslam, Hiller, Doyle and Robinson97 Secondly, the MACS trial comparing single to repeat courses of ANS showed no improvement of perinatal morbidity or mortality with repeat ANS dosing but demonstrated a reduction in birthweight, length and head circumference with repeat ANS dosing. These findings remained true in a secondary analysis of the MACS trial data when gestational age was accounted for. Reference Murphy, Hannah and Willan98,Reference Murphy, Willan and Hannah99

Cohort studies have also demonstrated an association between ANS exposure and evidence of reduced fetal growth. Term-born infants exposed to a single course of ANS had reduced birthweights, lengths and head circumferences compared to matched unexposed controls. Reference Davis, Waffarn, Uy, Hobel, Glynn and Sandman100 A cohort study of 477 infants exposed to repeat ANS and born preterm (<33 weeks’ gestation) also had reductions in birthweight (up to 9%) and reduced head circumferences. Reference French, Hagan, Evans, Godfrey and Newnham101 Further, Bloom et al. reported a retrospective cohort study of preterm infants exposed to a dexamethasone (four × 6 mg maternal intramuscular injections of dexamethasone phosphate 12 hourly) single or repeat regimen (n = 961). Dexamethasone-exposed infants demonstrated reduced birthweight compared to a reference population of matched controls from the same institution that did not receive ANS (n = 2808). The study also demonstrated significantly reduced birthweight with ANS exposure of up to −161 g (at 30–32 weeks’ gestation) when compared to a historically matched cohort (n = 444) from the same institution who on retrospective analysis had the same indications for ANS but were born a year before the introduction of ANS. Reference Bloom, Sheffield, McIntire and Leveno102 Braun et al. published the findings of a retrospective cohort study from all births at an institution in Germany from 1996 to 2008 and showed that preterm antenatal betamethasone exposure (n = 1799) was associated with a dose-dependent reduction in birthweight compared to matched controls (n = 42,240) after confounders were accounted for including gestational age. Reference Braun, Sloboda and Tutschek103 Those infants exposed to a dose of 24 mg of betamethasone (equivalent to a single course) had a reduction in birthweight of 523g, whilst any dose of betamethasone >24 mg (repeat courses) was associated with an 811g reduction in birthweight. Reference Braun, Sloboda and Tutschek103 Findings of reduced fetal growth were also detected on routine antenatal ultrasound scan prior to delivery with a dose-dependent reduction in estimated fetal weight (EFW). Reference Braun, Sloboda and Tutschek103 However, no difference was detected in neonatal morbidity or mortality. Reference Braun, Sloboda and Tutschek103 Similar findings have been reported from a study performed in Rome, which showed evidence of reduced growth velocity (head circumference, abdominal circumference, EFW) on antenatal ultrasound with a corresponding reduction in birthweight at delivery for a cohort of infants (n = 262) exposed to a single course of betamethasone (2 × 12 mg IM 24 hours apart) compared to controls (n = 270). Reference Rizzo, Mappa, Bitsadze, Khizroeva, Makatsariya and D’Antonio104 However, there was a significant difference in gestational age of delivery between the ANS cohort (mean 37.6 weeks) and control (mean 39.7 weeks) which may have contributed to the observed findings. Reference Rizzo, Mappa, Bitsadze, Khizroeva, Makatsariya and D’Antonio104 The longer-term impact of ANS on childhood growth is relatively unexplored. Osteen et al. reported the results of follow-up from 3,556 term-born children at 5 years of age, of which 629 children were exposed to ANS (betamethasone) and 2927 children were not (controls). Reference Osteen, Yang and McKinzie105 All pregnancies, including the controls were at some point assessed for threatened preterm labour. Those exposed to ANS had higher rates (21.8% vs. 16.4%) of being a small for gestational age infant (birthweight <10th centile) compared to controls which persisted at 5 years of age (13.7% vs. 7.1%, OR 2.00 95% CI 1.22–3.25). Reference Osteen, Yang and McKinzie105 However, ANS-exposed mothers were more likely to have diabetes or hypertensive disorders which could have affected fetal growth. Further, it is noted that there was a difference in gestational age at delivery (38.5 vs. 39.0 weeks); however, this is unlikely to be clinically significant. Reference Osteen, Yang and McKinzie105

Finally, a large population cohort study of all babies born in Finland between 2006 and 2010 (n = 278,508 singleton births, 4,887 of which were exposed to ANS) showed an association between ANS exposure and reduced birthweight, head circumference and length in babies born at preterm gestations up to term. Reference Rodriguez, Wang, Ali Khan, Cartwright, Gissler and Järvelin16

The longer-term implications of ANS-induced growth restriction observed in these studies are not yet fully understood but warrant further investigation. Especially so, as many ANS trial follow-up studies (including ASTECS and ALPS trials) have not reported on childhood or adolescent growth. Intrauterine growth restriction (IUGR) is however known to be a long-term risk factor for metabolic syndrome and associated cardiovascular disease. Therefore, it is not unreasonable to extrapolate the findings of IUGR to the observed effects of ANS on fetal growth. Reference Longo, Bollani, Decembrino, Di Comite, Angelini and Stronati106

Cardiovascular effects

Fetal cardiovascular system maturation is likely a result of both mechanical and hormonal influences. Reference Jellyman, Fletcher, Fowden and Giussani107 However, the exact mechanisms behind fetal cardiovascular maturation and the programming effects of glucocorticoids on fetal heart development are not yet fully understood. Studies have however shown that the GR and MR are highly expressed in the myocardium, endothelium and vascular smooth muscle. Reference Walker108

Animal studies

Key insights into the effects of glucocorticoids on the development of the cardiovascular system have been made in animal models and are explored below. Firstly, a study utilising a mouse model overexpressing cardiomyocyte GR showed no major structural abnormalities within the fetal heart but demonstrated an increased incidence of bradycardia and conduction defects. Reference Sainte-Marie, Cat and Perrier109 Other rodent studies have demonstrated that GR knockout results in a small but structurally normal heart, which exhibits diastolic dysfunction and early heart failure in midgestation. Reference Rog-Zielinska, Thomson and Kenyon110 On a cellular level, GR knockout mice display features of cardiac immaturity including poorly aligned cardiomyocytes, short, disorganised myofibrils as well as impaired cellular calcium homeostasis, which is important for normal cardiac contraction. Reference Rog-Zielinska, Thomson and Kenyon110 The importance of MR activation by glucocorticoids was demonstrated in a study that generated mice that overexpressed cardiomyocyte MR activity. Data showed that these mice had structurally normal hearts but demonstrated early and sudden death due to major arrhythmias. Reference Ouvrard-Pascaud, Sainte-Marie and Bénitah111 When the cardiomyocyte MR was knocked out, there was no major functional or morphological impact on the heart, demonstrating the variable effect of the MR on heart development. Reference Fraccarollo, Berger and Galuppo112

Rodent models of the cardiovascular effects of ANS show mixed results. Sakurai et al. demonstrated evidence of fetal cardiac maturation in rats exposed to antenatal dexamethasone. Histologically, dexamethasone-exposed hearts displayed organisation of ventricular myofibrils, cardiomyocyte hyperplasia and proliferation as well as an increase in cross-sectional area of the myocardium. Reference Sakurai, Osada and Takeba113 In contrast, a study in rats exposed to antenatal dexamethasone demonstrated impaired cardiac maturation with an increased heart/body ratio and lack of transition from hyperplastic growth to hypertrophic growth within the ventricles (higher cellular proliferation and lower extracellular matrix composition). Reference Torres, Belser, Umeda and Tucker114 De Vries et al. reported that adult rats exposed to antenatal dexamethasone had decreased heart weights but with increased cardiomyocyte size suggesting hypertrophy and increased collagen content. The authors suggested these changes were consistent with signs of early onset degeneration, which may cause permanent abnormalities to the cardiac structure. Reference dDe Vries, van der Leij and Bakker115

Sheep studies from the late 1990s have demonstrated that a direct in utero fetal infusion for 48 hours of betamethasone or dexamethasone at concentrations similar to clinical exposures results in an increase in fetal blood pressure and femoral vascular resistance. Reference Derks, Giussani and Jenkins116 Similar findings were demonstrated by Koenen et al. in fetal baboons exposed to maternal antenatal betamethasone. They showed an increase in fetal blood pressure without a change in fetal heart rate, maternal heart rate or maternal blood pressure. Reference Koenen, Mecenas, Smith, Jenkins and Nathanielsz117 Other non-human primate studies have demonstrated persistent cardiovascular effects after ANS exposure including increased blood pressure in juveniles. Reference de Vries, Holmes and Heijnis60,Reference Smith, Altamirano, Ervin, Seidner and Jobe118 Our group has demonstrated that fetal sheep exposed to an equivalent clinical dose of antenatal betamethasone have evidence of abnormal cardiac compliance on cardiac ultrasound examination (reduced mitral and tricuspid valve E/A ratios) which was associated with differential expression of genes related to myocardial hypertrophy within the fetal heart. Reference Kumagai, Kemp and Usuda119

A study using preterm pigs exposed to antenatal betamethasone compared to preterm controls demonstrated findings of cardiac maturation including increased atrial weight as well as evidence of cardiomyocyte terminal differentiation in the left and right ventricles as demonstrated by a shift from mononucleated cells to multinucleated. These findings indicate possible preterm cardiac hypertrophy resulting in acutely improved preterm cardiac function; however, the long-term implications of ANS-induced cardiac hypertrophy are unknown. Reference Kim, Eiby and Lumbers120

Clinical studies

Evidence from cohort studies on the effects of ANS with regard to the cardiovascular system is mixed. Dalzeil et al. have reported on the cardiovascular outcomes of individuals from the original AST (ANS vs. placebo) and showed no differences in blood pressure at 6 years of age and no difference in blood pressure, plasma lipids, cardiovascular disease or diabetes at 30 years of age. Reference Dalziel, Walker and Parag121 They did, however, report an increased insulin response to oral glucose tolerance test at 30 years of age, which may indicate a higher risk of developing diabetes (a main cardiovascular disease risk factor) due to insulin resistance later in life. Reference Dalziel, Walker and Parag121 A major limitation of the cohort data from the AST is that it includes only 19% of the study population at 6 years of age and 46% at 30 years of age. The 50-year cardiovascular follow-up of this same cohort was published recently by Walters et al. and showed no difference in cardiovascular risk factors (hypertension, hyperlipidaemia, diabetes, pre-diabetes) or major cardiovascular events between the exposed and unexposed cohorts. Reference Walters, Gamble and Crowther122 However, again follow-up rates were low (46% of the original cohort), and outcomes were measured by a patient questionnaire alone which has the potential for limitations and bias.

In comparison, a cohort study of 210 children born prematurely and with a birthweight <1500 g was reported at 14 years of age (>84% follow-up). Reference Doyle, Ford, Davis and Callanan123 Those exposed to ANS (n = 89) had an elevated systolic and diastolic blood pressure (albeit not in the hypertensive range) when compared to a matched unexposed group (n = 88). Reference Doyle, Ford, Davis and Callanan123 Furthermore, a prospective cohort study of 23–28-year-olds showed that individuals exposed to ANS had evidence of increased aortic arch stiffness on MRI assessment that was equivalent to adults decades older. Reference Kelly, Lewandowski and Worton124 This is a concerning finding as increased aortic arch stiffness is associated with an increased risk of developing hypertension, stroke and coronary artery disease later in life. Reference Kelly, Lewandowski and Worton124

Hepatic and metabolic effects

The programming effects of glucocorticoids on fetal and neonatal liver function is an area that is poorly understood at present. However, early studies have shown the importance of cortisol on fetal hepatic glycogen storage. In 1977, Barnes et al. showed that in utero control lambs had a rapid rise in hepatic glycogen concentration at 130d gestation (term 150d), whilst hepatic glycogen storage was markedly reduced in lambs that had undergone surgical removal of the adrenal and pituitary glands (removing endogenous fetal glucocorticoid exposure). Reference Barnes, Comline and Silver125 A direct fetal infusion of cortisol via a fetal catheter was then seen to restore normal hepatic glycogen concentrations. Reference Barnes, Comline and Silver125 Similar studies using glucocorticoid knock-out mice have shown impaired hepatic gluconeogenesis and glycogen storage after birth. Reference Monica Shih, Huang, Chu, Hsu and B-c126 This is significant as after birth and prior to the onset of established breastfeeding the infant must rely on endogenous glucose for metabolism. Therefore, any reduction in an infant’s ability to maintain glucose homeostasis immediately after birth (i.e. reduced gluconeogenesis and glycogen storage) may predispose it to hypoglycaemia, a finding that was seen in ANS-exposed infants in the ALPS trial. Reference Gyamfi-Bannerman and Thom15

Animal studies

Two studies (one in rhesus macaques, one in mice) have reported that antenatal betamethasone exposure resulted in an increase in fetal hepatic glycogen content and hepatic weight. Reference Ozdemir, Guvenal, Cetin, Kaya and Cetin91,Reference Epstein, Farrell, Sparks, Pepe, Driscoll and Chez127 Supporting this is evidence from rat models of antenatal exposure to dexamethasone showing an increase in hepatic steatosis without obesity, possibly due to the suppression of key genes (AMPK, PGC1α) involved in hepatic fat metabolism. Reference Drake, Raubenheimer, Kerrigan, McInnes, Seckl and Walker128,Reference Carbone, Zuloaga, Hiroi, Foradori, Legare and Handa129 An important study in sheep by Franko et al. demonstrated a mechanistic link between ANS exposure and increased hepatic glycogen storage. This study showed that a clinically relevant dose of antenatal dexamethasone increased the expression of the fetal hepatic gluconeogenic enzyme glucose-6-phosphatase (G6Pase), which was associated with increased fetal hepatic glycogen storage (2–3-fold), fetal plasma glucose and insulin levels. Reference Franko, Giussani, Forhead and Fowden130 Another study by de Vries et al. in non-human primates (African vervet) exposed to antenatal dexamethasone showed impaired glucose tolerance at 8 months of age based on higher fasting plasma insulin levels and abnormal (slower) glucose clearance on oral glucose tolerance test. Reference de Vries, Holmes and Heijnis60 At 12–14 months of age, the dexamethasone-exposed non-human primates from this study had a reduction in the number and size of pancreatic β cells (which are responsible for insulin production), as well as a reduction in hepatic expression and mRNA for a key gluconeogenic enzyme phosphoenol-pyruvate carboxykinase. Reference de Vries, Holmes and Heijnis60 These findings add further weight to the potential issues of glucose homeostasis and adaptation that infants exposed to ANS may face in the postnatal and infant periods. These changes may also predispose to long-term glucose insensitivity and increased risk of type II diabetes mellitus. Furthermore, Kuo et al. demonstrated that 10-year-old baboons (equivalent to a 40-year-old human) that were exposed to ANS had increased pericardial fat and hepatic lipid accumulation at a normal weight, indicating evidence of long-term metabolic reprogramming. Reference Kuo, Li and Li131 Conversely, a study utilising a rat model of pregnancy by Zhang et al. reported evidence of liver dysplasia, inhibition of hepatocyte proliferation and dose-dependent reduction in liver weight after antenatal dexamethasone exposure. Reference Zhang, Liu and Hu132 In sheep, our group has shown that preterm lambs exposed to ANS (both dexamethasone and betamethasone) demonstrate elevated neonatal plasma GGT levels, Reference Carter, Fee and Usuda32 which may be an indication of potential hepatobiliary dysfunction. Reference Cabrera-Abreu and Green133

In conclusion, normal hepatic homeostasis appears to be significantly affected in these animal models with ANS exposure, resulting in an increased risk of developing hepatic steatosis at a normal body weight (without obesity) in both early and later life.

Clinical studies

The exact mechanism of how ANS-induced fetal hepatic steatosis occurs remains relatively unknown. This is possibly because GR/MR expression and function within the developing fetal liver is poorly understood. Reference Chen, Xia, Shen, Xu, Guo and Wang134 However, mechanisms may be postulated from findings in adults, in which exogenous glucocorticoids increase hepatic lipid synthesis through increased expression of fatty acid synthase as well as an increase in hydrolysis of circulating triglycerides leading to a corresponding rise in free fatty acids. Reference Peckett, Wright and Riddell135 Unfortunately, there are very little clinical data at present on the short and long-term effects of ANS on human fetal and adult hepatic functioning. This is of concern given the rising incidence and earlier age of onset of non-alcoholic fatty liver disease around the world in conjunction with evidence in this review of a potential fetal origin for hepatic steatosis. Reference Chen, Xia, Shen, Xu, Guo and Wang134 Furthermore, the link between ANS exposure and disordered glucose metabolism outlined in this article suggests a possible link to childhood and adult-onset type II diabetes mellitus. This is important given that the Auckland Steroids Trial Cohort also demonstrated evidence of insulin resistance in later life. Reference Dalziel, Walker and Parag121

Renal system effects

Endogenous glucocorticoids (cortisol) demonstrate both glucocorticoid and modest MR activity. Presently, the impact of cortisol on fetal renal system development is not fully understood. Rat models have however revealed that pharmacological blockade of 11β-HSD-2 (the placental enzyme that protects the fetus from high levels of maternal glucocorticoids) results in adult-onset hypertension. Reference Lindsay, Lindsay, Edwards and Seckl136 Further, clinical studies of individuals with a severe genetic deficiency of 11β-HSD-2 display a phenotype of juvenile-onset hypertension. Reference Dave-Sharma, Wilson and Harbison137 Therefore, it is evident that fetal programming of the renal system must occur in relation to specific endogenous glucocorticoid exposures.

The impact of exogenous glucocorticoids (betamethasone and dexamethasone) on fetal renal programming is similarly not well characterised but requires further investigation especially given that these drugs demonstrate no direct MR action but cause suppression of cortisol and therefore impede cortisol’s action on the MR.

Animal studies

Animal models have in part helped to shed some light on the fetal programming effects of ANS exposure. Ortiz et al. utilising a rat model of pregnancy demonstrated a 20%–30% reduction in glomerular number as well as hypertension in juveniles who were exposed to two doses of antenatal dexamethasone compared to control. Reference Ortiz, Quan, Weinberg and Baum138 Similar findings have been demonstrated in sheep studies of ANS exposure showing a reduction in nephron and glomeruli counts at two months, six months and seven years of age which was accompanied by the development of hypertension. Reference Wintour, Moritz, Johnson, Ricardo, Samuel and Dodic139Reference Tang, Bi and Valego142 Fetal kidney development in sheep closely resembles that of humans, making this animal model highly translatable. Persistent abnormal renal morphology has also been demonstrated in ANS-exposed adult sheep at 7 years of age in the study by Wintour et al. Reference Wintour, Moritz, Johnson, Ricardo, Samuel and Dodic139 Findings from this study included grossly enlarged and dilated proximal renal tubules as well as abnormal collagen accumulation in the renal tubular interstitium and renal cortical vessels. The authors postulated that these findings, particularly increased collagen within cortical vessels, may account for the development of hypertension seen in those animals. Reference Wintour, Moritz, Johnson, Ricardo, Samuel and Dodic139 Furthermore, other studies in sheep exposed to antenatal betamethasone exhibited hypertension and abnormal angiotensin receptor expression at 1–1.5 years of age. Reference Gwathmey, Shaltout, Rose, Diz and Chappell143

These findings provide a link between ANS exposure, abnormal renal development and risk of hypertension in adult life . This is a potentially significant finding given the increasing incidence of renal disease and hypertension in human populations today. The exact mechanism by which ANS might cause hypertension remains unknown. However, human adults with essential hypertension have been shown to have a significant reduction in nephron counts similar to that observed in ANS animal studies. Reference Keller, Zimmer, Mall, Ritz and Amann144 Therefore, a reduction in nephron count may impair renal sodium excretion leading to increased blood volume and subsequent blood pressure.

Clinical studies

Evidence from two cohort studies investigating the effect of ANS on renal function and development is unclear. A cohort (n = 173) of 14-year-olds who were born at a mean gestation of ∼ 28 weeks’ and with a very low birth weight <1500 g showed that those exposed to ANS compared to unexposed exhibited alterations in the renin-angiotensin-aldosterone system (RAAS) as measured by urinary and plasma biomarkers. Reference South, Nixon and Chappell145 Dysregulation of the RAAS was more marked for those adolescents of black ethnicity. Reference South, Nixon and Chappell145 The authors concluded that these changes may increase the risk of developing hypertension and renal inflammation/fibrosis later in life. Reference South, Nixon and Chappell145 By comparison, a very similar cohort (n = 162) of 14-year-olds who were also born premature with a very low birthweight (<1500 g) showed that ANS exposure was not associated with abnormal kidney function as measured by eGFR. Reference Floyd, Beavers, Jensen, Washburn and South146 However, biomarkers of the RAAS were not measured in this cohort.

The impact of ANS on mineralocorticoid expression is not well characterised. Kessel et al. showed that in a clinical study of women exposed to antenatal betamethasone, plasma aldosterone levels were lower in both mother and infant at delivery compared to controls, which took up to 3–7 days to recover. Reference Kessel, Cale, Verbrick, Parker, Carlton and Bird147 Aldosterone plays a critical role in the homeostasis of electrolyte (potassium/sodium) excretion and reabsorption in the kidney, which therefore affects blood volume and pressure. Reference Kessel, Cale, Verbrick, Parker, Carlton and Bird147 The long-term programming effects of aldosterone suppression on the fetus remain unknown and require further investigation in clinical studies. However, this is another potential mechanism by which ANS may contribute to the observed development of hypertension in some individuals.

There is a current paucity of evidence from recent RCT follow-up studies (including ALPS/ASTECs or AST) on the long-term effects of ANS on renal functioning, therefore highlighting an area of need for further investigation.

Conclusion

ANS therapy undoubtedly improves perinatal outcomes when administered to the right patient at the right time. However, in order to maximise the potential benefits and minimise the potential harms of ANS therapy, a comprehensive understanding of the on and off-target effects of ANS on fetal development is required. Improving the current understanding of the potential off-target effects of ANS therapy on fetal development was the aim of this review. In addition, we highlight the urgent need for increased animal and clinical studies investigating fetal developmental programming after ANS exposure, particularly with regard to the immune, cardiovascular, renal and hepatic systems given the current sparsity of high-quality, long-term evidence. We also advocate for increased education, open disclosure and discussion between clinicians and patients on the current evidence of the off-target effects of ANS. We place particular emphasis on the late preterm and term gestations wherein the observed benefits of ANS therapy are very modest. Finally, we urge further studies on the optimisation of ANS therapies through better patient selection and reduced dose regimens based on the current pharmacokinetic understanding.

Acknowledgements

The authors would like to acknowledge Professor Alan Jobe for his extensive contribution to the current understanding of both the benefits and risks of ANS therapy as well as his mentorship and friendship.

Financial support

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Competing interests

None.

References

McGoldrick, E, Stewart, F, Parker, R, Dalziel, SR. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2020; 12(12), CD004454.Google ScholarPubMed
Roberts, D, Dalziel, S. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2007; 4, 1141.Google Scholar
Roberts, D, Brown, J, Medley, N, Dalziel, SR. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2017; 3, CD004454(1–191).Google ScholarPubMed
Liggins, GC, Howie, RN. A controlled trial of antepartum glucocorticoid treatment for prevention of the respiratory distress syndrome in premature infants. Pediatrics. 1972; 50(4), 515525.CrossRefGoogle ScholarPubMed
Walters, AG, Lin, L, Crowther, CA, Gamble, GD, Dalziel, SR, Harding, JE. Betamethasone for preterm birth: Auckland steroid trial full results and new insights 50 years on. J Pediatr. 2023; 255, 8088.e5.Google Scholar
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(2), 318325.CrossRefGoogle ScholarPubMed
Jobe, AH, Milad, MA, Peppard, T, Jusko, WJ. Pharmacokinetics and pharmacodynamics of intramuscular and oral betamethasone and dexamethasone in reproductive age women in India. Clin Transl Sci. 2020; 13(2), 391399.CrossRefGoogle ScholarPubMed
Gilstrap, LC, Christensen, R, Clewell, WH, et al. Effect of corticosteroids for fetal maturation on perinatal outcomes: NIH consensus development panel on the effect of corticosteroids for fetal maturation on perinatal outcomes. JAMA. 1995; 273(5), 413418.CrossRefGoogle Scholar
Kemp, MW, Newnham, JP, Challis, J, Jobe, AH, Stock, S. The clinical use of corticosteroids in pregnancy. Hum Reprod Update. 2016; 22(2), 240259.Google ScholarPubMed
Bridges, JP, Sudha, P, Lipps, D, et al. Glucocorticoid regulates mesenchymal cell differentiation required for perinatal lung morphogenesis and function. Am J Physiol Lung Cell Mol Physiol. 2020; 319(2), L239L255.Google ScholarPubMed
World Medical Association. Declaration of Helsinki. Ethical principles for medical research involving human subjects. 1964 [updated 28th September 2024, cited 2024 7th August]; Available from: https://www.wma.net/policies-post/wma-declaration-of-helsinki-ethical-principles-for-medical-research-involving-human-subjects/ Google Scholar
Lawn, JE, Davidge, R, Paul, VK, et al. Born too soon: care for the preterm baby. Reprod Health. 2013; 10, 119.CrossRefGoogle ScholarPubMed
Usuda, H, Carter, S, Takahashi, T, et al. Perinatal care for the extremely preterm infant. Semin Fetal Neonatal Med. 2022; 27(2), 101334.Google ScholarPubMed
Takahashi, T, Jobe, AH, Fee, EL, et al. The complex challenge of antenatal steroid therapy non-responsiveness. Am J Obstet Gynecol. 2022; 227(5), 696704.Google Scholar
Gyamfi-Bannerman, C, Thom, EA. Antenatal Betamethasone for women at risk for late preterm delivery. N Engl J Med. 2016; 375(5), 486487.Google ScholarPubMed
Rodriguez, A, Wang, Y, Ali Khan, A, Cartwright, R, Gissler, M, Järvelin, M-R. Antenatal corticosteroid therapy (ACT) and size at birth: a population-based analysis using the Finnish medical birth register. Plos Med. 2019; 16(2), e1002746.CrossRefGoogle ScholarPubMed
Seckl, JR, Meaney, MJ. Glucocorticoid programming. Ann N Y Acad Sci. 2004; 1032(1), 6384.CrossRefGoogle ScholarPubMed
Uno, H, Lohmiller, L, Thieme, C, et al. Brain damage induced by prenatal exposure to dexamethasone in fetal rhesus macaques. I. Hippocampus. Dev Brain Res. 1990; 53(2), 157167.CrossRefGoogle ScholarPubMed
Velísek, L. Prenatal corticosteroid impact on hippocampus: implications for postnatal outcomes. Epilepsy Behav. 2005; 7(1), 5767.CrossRefGoogle ScholarPubMed
Whitelaw, A, Thoresen, M. Antenatal steroids and the developing brain. Arch Dis Child Fetal Neonatal Ed. 2000; 83(2), F154F157.CrossRefGoogle ScholarPubMed
McGowan, PO, Sasaki, A, D”alessio, AC, et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci. 2009; 12(3), 342348.CrossRefGoogle Scholar
Connors, SL, Levitt, P, Matthews, SG, et al. Fetal mechanisms in neurodevelopmental disorders. Pediatr Neurol. 2008; 38(3), 163176.CrossRefGoogle ScholarPubMed
Buss, C, Entringer, S, Swanson, JM, Wadhwa, PD. The role of stress in brain development: the gestational environment’s long-term effects on the brain. Cerebrum. 2012; 2012, 4.Google ScholarPubMed
Back, SA, Riddle, A, Dean, J, Hohimer, AR. The instrumented fetal sheep as a model of cerebral white matter injury in the premature infant. Neurotherapeutics. 2012; 9(2), 359370.Google Scholar
Dunlop, SA, Archer, MA, Quinlivan, JA, Beazley, LD, Newnham, JP. Repeated prenatal corticosteroids delay myelination in the ovine central nervous system. J Matern Fetal Med. 1997; 6(6), 309313.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
Huang, W, Beazley, L, Quinlivan, J, Evans, S, Newnham, J, Dunlop, S. Effect of corticosteroids on brain growth in fetal sheep. Obstet Gynecol. 1999; 94(2), 213218.Google ScholarPubMed
Kanagawa, T, Tomimatsu, T, Hayashi, S, et al. The effects of repeated corticosteroid administration on the neurogenesis in the neonatal rat. Am J Obstet Gynecol. 2006; 194(1), 231238.Google ScholarPubMed
Tsiarli, MA, Rudine, A, Kendall, N, et al. Antenatal dexamethasone exposure differentially affects distinct cortical neural progenitor cells and triggers long-term changes in murine cerebral architecture and behavior. Transl Psychiatry. 2017; 7(6), e1153.Google ScholarPubMed
Noorlander, CW, Tijsseling, D, Hessel, EVS, et al. Antenatal glucocorticoid treatment affects hippocampal development in mice. PLoS ONE. 2014; 9(1), e85671.CrossRefGoogle ScholarPubMed
Rodriguez, JS, Zürcher, NR, Keenan, KE, Bartlett, TQ, Nathanielsz, PW, Nijland, MJ. Prenatal betamethasone exposure has sex specific effects in reversal learning and attention in juvenile baboons. Am J Obstet Gynecol. 2011; 204(6), 545.e1545.e10.CrossRefGoogle ScholarPubMed
Shields, A, Thomson, M, Winter, V, Coalson, J, Rees, S. Repeated courses of antenatal corticosteroids have adverse effects on aspects of brain development in naturally delivered baboon infants. Pediatr Res. 2012; 71(6), 661667.CrossRefGoogle ScholarPubMed
Carter, SW, Fee, EL, Usuda, H, et al. Antenatal steroids elicited neurodegenerative-associated transcriptional changes in the hippocampus of preterm fetal sheep independent of lung maturation. BMC Med. 2024; 22(1), 338.CrossRefGoogle ScholarPubMed
Schmidt, AF, Kannan, PS, Bridges, JP, et al. Dosing and formulation of antenatal corticosteroids for fetal lung maturation and gene expression in rhesus macaques. Sci Rep-UK. 2019; 9(1), 110.Google ScholarPubMed
Schmidt, AF, Schnell, DJ, Eaton, KP, et al. Fetal maturation revealed by amniotic fluid cell-free transcriptome in rhesus macaques. JCI Insight. 2022; 7(18), e162101.CrossRefGoogle ScholarPubMed
Crudo, A, Petropoulos, S, Suderman, M, et al. Effects of antenatal synthetic glucocorticoid on glucocorticoid receptor binding, DNA methylation, and genome-wide mRNA levels in the fetal male hippocampus. Endocrinology. 2013; 154(11), 41704181.Google ScholarPubMed
Crudo, A, Suderman, M, Moisiadis, VG, et al. Glucocorticoid programming of the fetal male hippocampal epigenome. Endocrinology. 2013; 154(3), 11681180.Google ScholarPubMed
Sasaki, A, Eng, ME, Lee, AH, Kostaki, A, Matthews, SG. DNA methylome signatures of prenatal exposure to synthetic glucocorticoids in hippocampus and peripheral whole blood of female guinea pigs in early life. Transl Psychiat. 2021; 11(1), 63.CrossRefGoogle ScholarPubMed
Constantinof, A, Moisiadis, VG, Kostaki, A, Szyf, M, Matthews, SG. Antenatal glucocorticoid exposure results in sex-specific and transgenerational changes in prefrontal cortex gene transcription that relate to behavioural outcomes. Sci Rep-UK. 2019; 9(1), 764.Google ScholarPubMed
Constantinof, A, Boureau, L, Moisiadis, VG, Kostaki, A, Szyf, M, Matthews, SG. Prenatal glucocorticoid exposure results in changes in gene transcription and DNA methylation in the female juvenile guinea pig hippocampus across three generations. Sci Rep-UK. 2019; 9(1), 18211.CrossRefGoogle ScholarPubMed
Tijsseling, D, Wijnberger, LDE, Derks, JB, et al. Effects of antenatal glucocorticoid therapy on hippocampal histology of preterm infants. PLoS ONE. 2012; 7(3), e33369.CrossRefGoogle Scholar
Davis, EP, Sandman, CA, Buss, C, Wing, DA, Head, K. Fetal glucocorticoid exposure is associated with preadolescent brain development. Biol Psychiat. 2013; 74(9), 647655.CrossRefGoogle ScholarPubMed
Räikkönen, K, Gissler, M, Kajantie, E. Associations between maternal antenatal corticosteroid treatment and mental and behavioral disorders in children. JAMA. 2020; 323(19), 19241933.Google ScholarPubMed
Lin, Y-H, Lin, C-H, Lin, M-C, Hsu, Y-C, Hsu, C-T. Antenatal corticosteroid exposure is associated with childhood mental disorders in late preterm and term infants. J Pediatr. 2022; 253, 245251.Google ScholarPubMed
Tao, S, Du, J, Chi, X, et al. Associations between antenatal corticosteroid exposure and neurodevelopment in infants. Am J Obstet Gynecol. 2022; 227(5), 759.e1759.e15.CrossRefGoogle ScholarPubMed
Asztalos, EV, Murphy, KE, Willan, AR, et al. Multiple courses of antenatal corticosteroids for preterm birth study: outcomes in children at 5 years of age (MACS-5). JAMA Pediatr. 2013; 167(12), 11021110.Google ScholarPubMed
Wapner, RJ, Sorokin, Y, Mele, L, et al. Long-term outcomes after repeat doses of antenatal corticosteroids. New Engl J Med. 2007; 357(12), 11901198.CrossRefGoogle ScholarPubMed
Gyamfi-Bannerman, C, Clifton, RG, Tita, AT, et al. Neurodevelopmental outcomes after late preterm antenatal corticosteroids: the ALPS follow-up study. JAMA. 2024; 331(19), 1629.CrossRefGoogle ScholarPubMed
McKinlay, CJD, Alsweiler, JM, Anstice, NS, et al. Association of neonatal glycemia with neurodevelopmental outcomes at 4.5 Years. JAMA Pediatr. 2017; 171(10), 972983.Google ScholarPubMed
Stutchfield, PR, Whitaker, R, Gliddon, AE, Hobson, L, Kotecha, S, Doull, IJ. Behavioural, educational and respiratory outcomes of antenatal betamethasone for term caesarean section (ASTECS trial). Arch Dis Child Fetal Neonatal Ed. 2013; 98(3), F195F200.CrossRefGoogle ScholarPubMed
MacArthur, B, Howie, R, Dezoete, J, Elkins, J. School progress and cognitive development of 6-year-old children whose mothers were treated antenatally with betamethasone. Pediatrics. 1982; 70(1), 99105.CrossRefGoogle ScholarPubMed
van der Voorn, B, Wit, JM, van der Pal, SM, Rotteveel, J, Finken, MJJ. Antenatal glucocorticoid treatment and polymorphisms of the glucocorticoid and mineralocorticoid receptors are associated with IQ and behavior in young adults born very preterm. J Clin Endocrinol Metab. 2015; 100(2), 500507.CrossRefGoogle ScholarPubMed
Savoy, C, Ferro, MA, Schmidt, LA, Saigal, S, Van Lieshout, RJ. Prenatal betamethasone exposure and psychopathology risk in extremely low birth weight survivors in the third and fourth decades of life. Psychoneuroendocrino. 2016; 74, 278285.Google ScholarPubMed
Dalziel, SR, Lim, VK, Lambert, A, et al. Antenatal exposure to betamethasone: psychological functioning and health related quality of life 31 years after inclusion in randomised controlled trial. BMJ. 2005; 331(7518), 665.CrossRefGoogle ScholarPubMed
Darlow, BA, Harris, SL, Horwood, LJ. Little evidence for long-term harm from antenatal corticosteroids in a population-based very low birthweight young adult cohort. Paediatr Perinat Epidemiol. 2022; 36(5), 631639.CrossRefGoogle Scholar
Moisiadis, VG, Matthews, SG. Glucocorticoids and fetal programming part 1: outcomes. Nat Rev Endocrinol. 2014; 10(7), 391402.CrossRefGoogle ScholarPubMed
Sánchez, MM, Young, LJ, Plotsky, PM, Insel, TR. Distribution of corticosteroid receptors in the rhesus brain: relative absence of glucocorticoid receptors in the hippocampal formation. J Neurosci. 2000; 20(12), 46574668.CrossRefGoogle ScholarPubMed
Waffarn, F, Davis, EP. Effects of antenatal corticosteroids on the hypothalamic-pituitary-adrenocortical axis of the fetus and newborn: experimental findings and clinical considerations. Am J Obstet Gynecol. 2012; 207(6), 446454.CrossRefGoogle ScholarPubMed
Seckl, JR. Prenatal glucocorticoids and long-term programming. Eur J Endocrinol. 2004; 151(3), U49U62.CrossRefGoogle ScholarPubMed
Matthews, SG. Antenatal glucocorticoids and programming of the developing CNS. Pediatr Res. 2000; 47(3), 291300.Google ScholarPubMed
de Vries, A, Holmes, MC, Heijnis, A, et al. Prenatal dexamethasone exposure induces changes in nonhuman primate offspring cardiometabolic and hypothalamic-pituitary-adrenal axis function. J Clin Invest. 2007; 117(4), 10581067.CrossRefGoogle ScholarPubMed
Usuda, H, Fee, EL, Carter, S, et al. 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(6), 903.e1903.e16.CrossRefGoogle Scholar
Fee, EL, Takahashi, T, Takahashi, Y, et al. One percent of the clinical dose used for antenatal steroid therapy is sufficient to induce lung maturation when administered directly to the preterm ovine fetus. Am J Physiol Lung Cell Mol Physiol. 2022; 322(6), L853L865.CrossRefGoogle Scholar
Takahashi, T, Fee, EL, Takahashi, Y, et al. 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(4), 564.e1564.e14.Google ScholarPubMed
Fee, EL, Takahashi, T, Takahashi, Y, et al. Respiratory benefit in preterm lambs is progressively lost when the concentration of fetal plasma betamethasone is titrated below two nanograms per milliliter. Am J Physiol Lung Cell Mol Physiol. 2023; 325(5), L628L637.Google ScholarPubMed
McCabe, L, Marash, D, Li, A, Matthews, S. Repeated antenatal glucocorticoid treatment decreases hypothalamic corticotropin releasing hormone mRNA but not corticosteroid receptor mRNA expression in the fetal guinea-pig brain. J Neuroendocrinol. 2001; 13(5), 425431.Google Scholar
Niwa, F, Kawai, M, Kanazawa, H, et al. Limited response to CRH stimulation tests at 2 weeks of age in preterm infants born at less than 30 weeks of gestational age. Clin Endocrinol. 2013; 78(5), 724729.Google ScholarPubMed
Schäffer, L, Luzi, F, Burkhardt, T, Rauh, M, Beinder, E. Antenatal betamethasone administration alters stress physiology in healthy neonates. Obstet Gynecol. 2009; 113(5), 10821088.CrossRefGoogle ScholarPubMed
Tegethoff, M, Pryce, C, Meinlschmidt, G. Effects of intrauterine exposure to synthetic glucocorticoids on fetal, newborn, and infant hypothalamic-pituitary-adrenal axis function in humans: a systematic review. Endocr Rev. 2009; 30(7), 753789.Google ScholarPubMed
Weiss, SJ, Keeton, V, Richoux, S, Cooper, B, Niemann, S. Exposure to antenatal corticosteroids and infant cortisol regulation. Psychoneuroendocrino. 2023; 147, 105960.CrossRefGoogle ScholarPubMed
Davis, EP, Waffarn, F, Sandman, CA. Prenatal treatment with glucocorticoids sensitizes the hpa axis response to stress among full-term infants. Dev Psychobiol. 2011; 53(2), 175183.CrossRefGoogle ScholarPubMed
Gover, A, Brummelte, S, Synnes, AR, et al. Single course of antenatal steroids did not alter cortisol in preterm infants up to 18 months. Acta Paediatr. 2012; 101(6), 604608.Google Scholar
Ashwood, PJ, Crowther, CA, Willson, KJ, et al. Neonatal adrenal function after repeat dose prenatal corticosteroids: a randomized controlled trial. Am J Obstet Gynecol. 2006; 194(3), 861867.Google ScholarPubMed
Alexander, N, Rosenlöcher, F, Stalder, T, et al. Impact of antenatal synthetic glucocorticoid exposure on endocrine stress reactivity in term-born children. J Clin Endocrinol Metab. 2012; 97(10), 35383544.CrossRefGoogle ScholarPubMed
Ilg, L, Kirschbaum, C, Li, S-C, Rosenlöcher, F, Miller, R, Alexander, N. Persistent effects of antenatal synthetic glucocorticoids on endocrine stress reactivity from childhood to adolescence. J Clin Endocrinol Metab. 2018; 104(3), 827834.Google Scholar
McKenna, DS, Wittber, GM, Nagaraja, H, Samuels, P. The effects of repeat doses of antenatal corticosteroids on maternal adrenal function. Am J Obstet Gynecol. 2000; 183(3), 669673.Google ScholarPubMed
Sarnes, E, Crofford, L, Watson, M, Dennis, G, Kan, H, Bass, D. Incidence and US costs of corticosteroid-associated adverse events: a systematic literature review. Clin Ther. 2011; 33(10), 14131432.Google ScholarPubMed
Nakagawa, M, Terashima, T, D’yachkova, Y, Bondy, GP, Hogg, JC, van Eeden, SF. Glucocorticoid-induced granulocytosis: contribution of marrow release and demargination of intravascular granulocytes. Circulation. 1998; 98(21), 23072313.Google ScholarPubMed
Waisman, D, Van Eeden, SF, Hogg, JC, Solimano, A, Massing, B, Bondy, GP. L-selectin expression on polymorphonuclear leukocytes and monocytes in premature infants: reduced expression after dexamethasone treatment for bronchopulmonary dysplasia. J Pediatr. 1998; 132(1), 5356.CrossRefGoogle ScholarPubMed
Cox, G. Glucocorticoid treatment inhibits apoptosis in human neutrophils. Separation of survival and activation outcomes. J Immunol. 1995; 154(9), 47194725.CrossRefGoogle Scholar
Murthy, KK, Moya, FR. Effect of betamethasone on maternal, fetal and neonatal rat cellular immunity. Early Hum Dev. 1994; 36(1), 111.CrossRefGoogle ScholarPubMed
Diepenbruck, I, Much, CC, Krumbholz, A, et al. Effect of prenatal steroid treatment on the developing immune system. J Mol Med. 2013; 91(11), 12931302.Google ScholarPubMed
Smolders-de Haas, H, Neuvel, J, Schmand, B, Treffers, PE, Koppe, J, Hoeks, J. Physical development and medical history of children who were treated antenatally with corticosteroids to prevent respiratory distress syndrome: a 10-to 12-year follow-up. Pediatrics. 1990; 86(1), 6570.Google ScholarPubMed
Klein, K, McClure, EM, Colaci, D, et al. The antenatal corticosteroids trial (ACT): a secondary analysis to explore site differences in a multi-country trial. Reprod Health. 2016; 13(1), 64.CrossRefGoogle Scholar
Räikkönen, K, Gissler, M, Kajantie, E, Tapiainen, T. Antenatal corticosteroid treatment and infectious diseases in children: a nationwide observational study. Lancet Reg Health Eur. 2023; 35, 100750.CrossRefGoogle ScholarPubMed
Liang, FW, Tsai, HF, Kuo, PL, Tsai, PY. Antenatal corticosteroid therapy in late preterm delivery: a nationwide population-based retrospective study in Taiwan. BJOG. 2021; 128(9), 14971502.Google ScholarPubMed
Carter, SW, Neubronner, S, Su, LL, et al. Chorioamnionitis: an update on diagnostic evaluation. Biomedicines. 2023; 11(11), 2922.CrossRefGoogle ScholarPubMed
Ikegami, M, Jobe, AH, Newnham, J, Polk, DH, Willet, KE, Sly, P. Repetitive prenatal glucocorticoids improve lung function and decrease growth in preterm lambs. Am J Resp Crit Care. 1997; 156(1), 178184.Google ScholarPubMed
Newnham, JP, Evans, SF, Godfrey, M, Huang, W, Ikegami, M, Jobe, A. Maternal, but not fetal, administration of corticosteroids restricts fetal growth. J Matern Fetal Med. 1999; 8(3), 8187.Google Scholar
Netchine, I, Azzi, S, Le Bouc, Y, Savage, MO. IGF1 molecular anomalies demonstrate its critical role in fetal, postnatal growth and brain development. Best Pract Res Clin Endocrinol Metab. 2011; 25(1), 181190.Google ScholarPubMed
Agrogiannis, GD, Sifakis, S, Patsouris, ES, Konstantinidou, AE. Insulin-like growth factors in embryonic and fetal growth and skeletal development (Review). Mol Med Rep. 2014; 10(2), 579584.Google ScholarPubMed
Ozdemir, H, Guvenal, T, Cetin, M, Kaya, T, Cetin, A. A placebo-controlled comparison of effects of repetitive doses of betamethasone and dexamethasone on lung maturation and lung, liver, and body weights of mouse pups. Pediatr Res. 2003; 53(1), 98103.Google ScholarPubMed
Abrantes, MA, Valencia, AM, Bany-Mohammed, F, Aranda, JV, Beharry, KD. Combined antenatal and postnatal steroid effects on fetal and postnatal growth, and neurological outcomes in neonatal rats. Am J Transl Res. 2019; 11(3), 16971710.Google ScholarPubMed
Abrantes, MA, Valencia, AM, Bany-Mohammed, F, Aranda, JV, Beharry, KD. Intergenerational influence of antenatal betamethasone on growth, growth factors, and neurological outcomes in rats. Reprod Sci. 2020; 27(1), 418431.CrossRefGoogle ScholarPubMed
Sun, B, Jobe, A, Rider, E, Ikegami, M. Single dose versus two doses of betamethasone for lung maturation in preterm rabbits. Pediatr Res. 1993; 33(3), 256260.Google ScholarPubMed
Pratt, L, Magness, RR, Phernetton, T, Hendricks, SK, Abbott, DH, Bird, IM. Repeated use of betamethasone in rabbits: effects of treatment variation on adrenal suppression, pulmonary maturation, and pregnancy outcome. Am J Obstet Gynecol. 1999; 180(4), 9951005.Google ScholarPubMed
Van Der Merwe, J, Van Der Veeken, L, Inversetti, A, et al. Neurocognitive sequelae of antenatal corticosteroids in a late preterm rabbit model. Am J Obstet Gynecol. 2022; 226(6), 850.e1850.e21.CrossRefGoogle Scholar
Crowther, CA, Haslam, RR, Hiller, JE, Doyle, LW, Robinson, JS. Neonatal respiratory distress syndrome after repeat exposure to antenatal corticosteroids: a randomised controlled trial. Lancet. 2006; 367(9526), 19131919.Google ScholarPubMed
Murphy, KE, Hannah, ME, Willan, AR, et al. Multiple courses of antenatal corticosteroids for preterm birth (MACS): a randomised controlled trial. Lancet. 2008; 372(9656), 21432151.Google Scholar
Murphy, KE, Willan, AR, Hannah, ME, et al. Effect of antenatal corticosteroids on fetal growth and gestational age at birth. Obstet Gynecol. 2012; 119(5), 917923.CrossRefGoogle ScholarPubMed
Davis, E, Waffarn, F, Uy, C, Hobel, C, Glynn, L, Sandman, C. Effect of prenatal glucocorticoid treatment on size at birth among infants born at term gestation. J Perinatol. 2009; 29(11), 731737.CrossRefGoogle ScholarPubMed
French, NP, Hagan, R, Evans, SF, Godfrey, M, Newnham, JP. Repeated antenatal corticosteroids: size at birth and subsequent development. Am J Obstet Gynecol. 1999; 180(1), 114121.CrossRefGoogle ScholarPubMed
Bloom, SL, Sheffield, JS, McIntire, DD, Leveno, KJ. Antenatal dexamethasone and decreased birth weight. Obstet Gynecol. 2001; 97(4), 485490.Google ScholarPubMed
Braun, T, Sloboda, DM, Tutschek, B, et al. Fetal and neonatal outcomes after term and preterm delivery following betamethasone administration. Int J Gynecol Obstet. 2015; 130(1), 6469.CrossRefGoogle ScholarPubMed
Rizzo, G, Mappa, I, Bitsadze, V, Khizroeva, J, Makatsariya, A, D’Antonio, F. Administration of antenatal corticosteroid is associated with reduced fetal growth velocity: a longitudinal study. J Matern Fetal Neonatal Med. 2022; 35(14), 27752780.CrossRefGoogle ScholarPubMed
Osteen, SJ, Yang, Z, McKinzie, AH, et al. Long-term childhood outcomes for babies born at term who were exposed to antenatal corticosteroids. Am J Obstet Gynecol. 2023; 228(1), 80.e8180.e86.CrossRefGoogle ScholarPubMed
Longo, S, Bollani, L, Decembrino, L, Di Comite, A, Angelini, M, Stronati, M. Short-term and long-term sequelae in intrauterine growth retardation (IUGR). J Matern Fetal Neonatal Med. 2013; 26(3), 222225.Google ScholarPubMed
Jellyman, JK, Fletcher, AJ, Fowden, AL, Giussani, DA. Glucocorticoid maturation of fetal cardiovascular function. Trends Mol Med. 2020; 26(2), 170184.Google ScholarPubMed
Walker, BR. Glucocorticoids and cardiovascular disease*. Eur J Endocrinol. 2007; 157(5), 545559.CrossRefGoogle ScholarPubMed
Sainte-Marie, Y, Cat, AND, Perrier, R, et al. Conditional glucocorticoid receptor expression in the heart induces atrio-ventricular block. FASEBJ. 2007; 21(12), 31333141.Google ScholarPubMed
Rog-Zielinska, EA, Thomson, A, Kenyon, CJ, et al. Glucocorticoid receptor is required for foetal heart maturation. Hum Mol Genet. 2013; 22(16), 32693282.CrossRefGoogle ScholarPubMed
Ouvrard-Pascaud, A, Sainte-Marie, Y, Bénitah, J-P, et al. Conditional mineralocorticoid receptor expression in the heart leads to life-threatening arrhythmias. Circulation. 2005; 111(23), 30253033.CrossRefGoogle ScholarPubMed
Fraccarollo, D, Berger, S, Galuppo, P, et al. Deletion of cardiomyocyte mineralocorticoid receptor ameliorates adverse remodeling after myocardial infarction. Circulation. 2011; 123(4), 400408.Google ScholarPubMed
Sakurai, K, Osada, Y, Takeba, Y, et al. Exposure of immature rat heart to antenatal glucocorticoid results in cardiac proliferation. Pediatr Int. 2019; 61(1), 3142.CrossRefGoogle ScholarPubMed
Torres, A, Belser, WW, Umeda, PK, Tucker, D. Indicators of delayed maturation of rat heart treated prenatally with dexamethasone. Pediatr Res. 1997; 42(2), 139144.CrossRefGoogle ScholarPubMed
dDe Vries, WB, van der Leij, FR, Bakker, JM, et al. Alterations in adult rat heart after neonatal dexamethasone therapy. Pediatr Res. 2002; 52(6), 900906.Google Scholar
Derks, JB, Giussani, DA, Jenkins, SL, et al. A comparative study of cardiovascular, endocrine and behavioural effects of betamethasone and dexamethasone administration to fetal sheep. J Physiol. 1997; 499(1), 217226.CrossRefGoogle ScholarPubMed
Koenen, S, Mecenas, C, Smith, G, Jenkins, S, Nathanielsz, P. Effects of maternal betamethasone administration on fetal and maternal blood pressure and heart rate in the baboon at 0.7 of gestation. Am J Obstet Gynecol. 2002; 186(4), 812817.CrossRefGoogle ScholarPubMed
Smith, LM, Altamirano, AK, Ervin, MG, Seidner, SR, Jobe, AH. Prenatal glucocorticoid exposure and postnatal adaptation in premature newborn baboons ventilated for six days. Am J Obstet Gynecol. 2004; 191(5), 16881694.Google ScholarPubMed
Kumagai, Y, Kemp, MW, Usuda, H, et al. A reduction in antenatal steroid dose was associated with reduced cardiac dysfunction in a sheep model of pregnancy. Reprod Sci. 2023; 30(11), 32223234.CrossRefGoogle Scholar
Kim, MY, Eiby, YA, Lumbers, ER, et al. Effects of glucocorticoid exposure on growth and structural maturation of the heart of the preterm piglet. PLoS One. 2014; 9(3), e93407.CrossRefGoogle ScholarPubMed
Dalziel, SR, Walker, NK, Parag, V, et al. Cardiovascular risk factors after antenatal exposure to betamethasone: 30-year follow-up of a randomised controlled trial. Lancet. 2005; 365(9474), 18561862.CrossRefGoogle ScholarPubMed
Walters, AG, Gamble, GD, Crowther, CA, et al. Cardiovascular outcomes 50 years after antenatal exposure to betamethasone: follow-up of a randomised double-blind, placebo-controlled trial. Plos Med. 2024; 21(4), e1004378.Google ScholarPubMed
Doyle, L, Ford, G, Davis, N, Callanan, C. Antenatal corticosteroid therapy and blood pressure at 14 years of age in preterm children. Clin Sci. 2000; 98(2), 137142.Google ScholarPubMed
Kelly, BA, Lewandowski, AJ, Worton, SA, et al. Antenatal glucocorticoid exposure and long-term alterations in aortic function and glucose metabolism. Pediatrics. 2012; 129(5), e1282e1290.CrossRefGoogle ScholarPubMed
Barnes, R, Comline, R, Silver, M. Effect of cortisol on liver glycogen concentrations in hypophysectomized, adrenalectomized and normal foetal lambs during late or prolonged gestation. J Physiol. 1978; 275(1), 567579.CrossRefGoogle ScholarPubMed
Monica Shih, M-C, Huang, C-CJ, Chu, H-P, Hsu, N-C, B-c, Chung. Embryonic steroids control developmental programming of energy balance. Endocrinology. 2021; 162(12), bqab196.CrossRefGoogle ScholarPubMed
Epstein, MF, Farrell, PM, Sparks, JW, Pepe, G, Driscoll, SG, Chez, RA. Maternal betamethasone and fetal growth and development in the monkey. Am J Obstet Gynecol. 1977; 127(3), 261263.CrossRefGoogle ScholarPubMed
Drake, AJ, Raubenheimer, PJ, Kerrigan, D, McInnes, KJ, Seckl, JR, Walker, BR. Prenatal dexamethasone programs expression of genes in liver and adipose tissue and increased hepatic lipid accumulation but not obesity on a high-fat diet. Endocrinology. 2010; 151(4), 15811587.CrossRefGoogle Scholar
Carbone, DL, Zuloaga, DG, Hiroi, R, Foradori, CD, Legare, ME, Handa, RJ. Prenatal dexamethasone exposure potentiates diet-induced hepatosteatosis and decreases plasma IGF-I in a sex-specific fashion. Endocrinology. 2012; 153(1), 295306.CrossRefGoogle Scholar
Franko, K, Giussani, D, Forhead, A, Fowden, A. Effects of dexamethasone on the glucogenic capacity of fetal, pregnant, and non-pregnant adult sheep. J Endocrinol. 2007; 192(1), 6773.Google ScholarPubMed
Kuo, AH, Li, J, Li, C, et al. Prenatal steroid administration leads to adult pericardial and hepatic steatosis in male baboons. Int J Obesity. 2017; 41(8), 12991302.CrossRefGoogle ScholarPubMed
Zhang, D, Liu, K, Hu, W, et al. Prenatal dexamethasone exposure caused fetal rats liver dysplasia by inhibiting autophagy-mediated cell proliferation. Toxicology. 2021; 449, 152664.Google ScholarPubMed
Cabrera-Abreu, JC, Green, A. γ-Glutamyltransferase: value of its measurement in paediatrics. Ann Clin Biochem. 2002; 39(1), 2225.Google ScholarPubMed
Chen, Z, Xia, L-P, Shen, L, Xu, D, Guo, Y, Wang, H. Glucocorticoids and intrauterine programming of nonalcoholic fatty liver disease. Metabolis. 2024; 150, 155713.CrossRefGoogle ScholarPubMed
Peckett, AJ, Wright, DC, Riddell, MC. The effects of glucocorticoids on adipose tissue lipid metabolism. Metabolis. 2011; 60(11), 15001510.Google ScholarPubMed
Lindsay, RS, Lindsay, RM, Edwards, CR, Seckl, JR. Inhibition of 11β-hydroxysteroid dehydrogenase in pregnant rats and the programming of blood pressure in the offspring. Hypertension. 1996; 27(6), 12001204.Google ScholarPubMed
Dave-Sharma, S, Wilson, RC, Harbison, MD, et al. Examination of genotype and phenotype relationships in 14 patients with apparent mineralocorticoid excess. J Clin Endocrinol Metab. 1998; 83(7), 22442254.Google ScholarPubMed
Ortiz, LA, Quan, A, Weinberg, A, Baum, M. Effect of prenatal dexamethasone on rat renal development. Kidney Int. 2001; 59(5), 16631669.CrossRefGoogle ScholarPubMed
Wintour, EM, Moritz, KM, Johnson, K, Ricardo, S, Samuel, CS, Dodic, M. Reduced nephron number in adult sheep, hypertensive as a result of prenatal glucocorticoid treatment. J Physiol. 2003; 549(3), 929935.CrossRefGoogle ScholarPubMed
Moritz, KM, De Matteo, R, Dodic, M, et al. Prenatal glucocorticoid exposure in the sheep alters renal development in utero: implications for adult renal function and blood pressure control. Am J Physiol Regul Integr Comp Physiol. 2011; 301(2), R500R509.CrossRefGoogle ScholarPubMed
Figueroa, JP, Rose, JC, Massmann, GA, Zhang, J, Acuña, G. Alterations in fetal kidney development and elevations in arterial blood pressure in young adult sheep after clinical doses of antenatal glucocorticoids. Pediatr Res. 2005; 58(3), 510515.CrossRefGoogle Scholar
Tang, L, Bi, J, Valego, N, et al. Prenatal betamethasone exposure alters renal function in immature sheep: sex differences in effects. Am J Physiol Regul Integr Comp Physiol. 2010; 299(3), R793R803.Google ScholarPubMed
Gwathmey, TM, Shaltout, HA, Rose, JC, Diz, DI, Chappell, MC. Glucocorticoid-induced fetal programming alters the functional complement of angiotensin receptor subtypes within the kidney. Hypertension. 2011; 57(3), 620626.Google ScholarPubMed
Keller, G, Zimmer, G, Mall, G, Ritz, E, Amann, K. Nephron number in patients with primary hypertension. New Engl J Med. 2003; 348(2), 101108.Google ScholarPubMed
South, AM, Nixon, PA, Chappell, MC, et al. Antenatal corticosteroids and the renin-angiotensin-aldosterone system in adolescents born preterm. Pediatr Res. 2017; 81(1), 8893.CrossRefGoogle ScholarPubMed
Floyd, WN, Beavers, DP, Jensen, ET, Washburn, LK, South, AM. Association of antenatal corticosteroids with kidney function in adolescents born preterm with very low birth weight. J Perinatol. 2023; 43(8), 10381044.CrossRefGoogle ScholarPubMed
Kessel, JM, Cale, JM, Verbrick, E, Parker, CR Jr, Carlton, DP, Bird, IM. Antenatal betamethasone depresses maternal and fetal aldosterone levels. Reprod Sci. 2009; 16(1), 94104.(Thousand Oaks, Calif)CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Graphical abstract of the short and long term off target effects of antenatal corticosteroid therapy on fetal development. Created in BioRender.com.