498
Views
3
CrossRef citations to date
0
Altmetric
Editorial

Does the intrauterine growth-restricted fetus benefit from antenatal glucocorticoids?

&
Pages 149-152 | Published online: 10 Jan 2014

In Australia, 8% of the 280,000 annual births are preterm; specifically, prior to 37 weeks completed gestation Citation[1]. Preterm infants represent 75% of all neonatal deaths in Australia, with the vast majority of these deaths due to pulmonary disease Citation[2]. The costs of caring for preterm infants are enormous – US$5.8 billion in the USA, representing 57% of neonatal care in that country Citation[3]. The cost to support a single infant born at 25 weeks is estimated to be at US$250,000 Citation[4]. Since Liggins’ proposal that glucocorticoids (GCs), including cortisol, β-methasone and dexamethasone, specifically induce pulmonary surfactant synthesis Citation[5], it has been shown that synthetic GCs reduce the incidence of respiratory distress syndrome by approximately 50% in preterm infants Citation[6]. Interestingly, not all babies may benefit from antenatal GC; for example, the growth-restricted fetus. Although the size of this subset of preterm babies is not clear, 7% of Australian babies are born intrauterine growth restricted (IUGR) Citation[1]. This editorial discusses the evidence for and against the use of antenatal GC, particularly in relation to the complex cardiovascular and respiratory physiology of the IUGR infant.

Benefits of antenatal GC treatment

The use of a single course of antenatal GCs, either dexamethasone or β-methasone, is recommended for women at risk of preterm labor between 24 and 34 weeks, and is associated with positive outcomes for the infant in that they reduce the incidence of neonatal complications associated with preterm birth; for example, neonatal respiratory distress Citation[6], intraventricular hemorrhage Citation[7] and neonatal death Citation[7–9]. Experimental studies show an improvement in fetal lung mechanics after very short treatment-to-delivery times Citation[10], with concomitant alterations in lung structure Citation[11]. Antenatal GC is most effective when administered between 7 days and 24 h before delivery Citation[8]. As 50% of women in threatened preterm labor do not deliver within 7–14 days of initial antenatal GC Citation[12], it has, in previous years, been common practice to repeat the course of antenatal GC at weekly intervals until the time of delivery Citation[13–15]. The latest Cochrane review on antenatal GCs in women at risk of preterm labor concludes that a single course of antenatal GC is recommended to accelerate fetal lung maturation Citation[16,17]. However, further research is required concerning the optimal dose-to-delivery interval, optimal corticosteroid to use and the effects of multiple pregnancies, as well as to confirm the long-term effects into adulthood, especially of multiple doses Citation[16]. A recent, randomized controlled trial shows that multiple doses every 14 days do not improve survival in preterm babies but do decrease fetal growth Citation[18].

Antenatal GCs alter lung structure, as they result in a thinning of the alveolar walls, a higher proportion of alveolar ducts and a lower alveolar wall fraction in preterm sheep Citation[19]. These structural indices result in a higher average alveolar volume and improved lung function within 2 days after GCs Citation[19]. Improved lung function is measured by improved lung compliance Citation[20,21], greater pulmonary surfactant stability (i.e., sustained ability to lower surface tension) and the ability of the surfactant to improve lung compliance in surfactant-deficient animals Citation[20]. These effects occurred after a single treatment 48 h before delivery Citation[20,21]. On the other hand, increased surfactant lipid content and saturation, as well as an increased expression of surfactant proteins (SPs) in alveolar (i.e., lavageable) surfactant, is only manifest after repeated doses over a prolonged period of time (7–21 days) Citation[19,21]. In sheep, repetitive doses at 7-day intervals improve fetal lung function cumulatively when delivered preterm Citation[22].

Costs of antenatal GC treatment

Despite the benefits of antenatal GC treatment for neonatal lung function, antenatal GCs are also associated with several negative birth outcomes, such as adrenal suppression, neonatal sepsis, and increased risk of neonatal and maternal infections Citation[8,23]. Of greatest concern is that antenatal GCs reduce fetal growth, resulting in both lower body weight, but also in a smaller and less mature brain Citation[12,23]. In fact, these children remain smaller at school age Citation[24]. In addition, animal studies show that antenatal GCs reduce cerebral blood flow due to increased cerebral vascular resistance with a decrease in oxygen delivery Citation[25]. Moreover, there are deleterious effects on cerebral myelination Citation[26] and significant alterations in the function of the hypothalamic–pituitary–adrenal axis Citation[27,28]. These changes may affect later neurodevelopment Citation[29]. There is an increase in umbilical blood flow in response to β-methasone Citation[30]. In pregnant ewes infused for 48 h with β-methasone, fetal cerebral blood flow and oxygen delivery are reduced due to increased cerebrovascular resistance. This may account for the decreased brain growth observed at term in human fetuses following either single or repeated GCs Citation[31]. GCs act to increase vascular resistance through a variety of mechanisms, including enhanced sensitivity to vasoconstrictors and decreased sensitivity to vasodilators Citation[32].

Effects of antenatal GC treatment in the IUGR fetus

There are conflicting data on whether antenatal GCs are Citation[9] or are not Citation[7] associated with a reduction in the complications associated with preterm delivery in IUGR fetuses. A large study of 19,759 very low-birthweight neonates found that antenatal GCs lowered the risk of respiratory distress, intraventricular hemorrhage and death in both normal and growth-restricted fetuses Citation[9]. On the other hand, a study of 1148 neonates found that there was no difference in the incidence of respiratory distress, intraventricular hemorrhage or necrotizing enterocolitis in growth-restricted fetuses whether they were treated with antenatal GCs or not Citation[7]. The already higher circulating plasma cortisol concentrations in the IUGR fetus may have already affected lung growth and surfactant maturation such that exposure to higher doses may have no or a detrimental effect. In addition, the effects of GCs are widespread, not limited to the lung, and as such, actions of GCs at other sites may be deleterious for the IUGR fetus. The complexity of the interactions between the factors that regulate SP synthesis is exemplified in the case of the IUGR fetus.

Impact of IUGR, hypoxemia & cortisol on the surfactant system

Fetal sheep models have been utilized to investigate the effects of IUGR on the lung and surfactant system and antenatal GCs, either separately or additively, with conflicting but interesting mechanistic results. IUGR is most commonly caused by placental insufficiency leading to chronic hypoxemia and hypoglycemia Citation[33,34]. The fetus responds to the reduced substrate supply with a range of adaptations. Neuroendocrine adaptations include a reduction in plasma IGF-1 concentration Citation[35] and an augmentation of the normal prepartum surge in cortisol Citation[36–38]. There are also changes in lung structure in the IUGR fetus, which include structural deficits to both the alveolar cells and the lung parenchyma Citation[39,40].

One study attempted to address the impact of hypoxemia directly, without the complications of nutrient restriction and/or hypoglycemia. In this case, only older fetuses responded to hypoxemia with an increase in SP mRNA expression correlating with the prepartum cortisol surge Citation[41], a result very similar to that observed with the uteroplacental embolization model of IUGR by Gagnon et al.Citation[42]. On the other hand, in an umbilical cord-occlusion model Citation[43], there was a significant reduction in SP-A, -B and -C mRNA expression between 130 and 133 days Citation[43]. This decrease in SP mRNA expression occurred despite an increase in cortisol at the end of the 4-day experimental period. The authors argue that their model causes a much more severe hypoxemia (partial pressure of oxygen in the blood decreased by 70% as opposed to 30–50% in other studies), and is associated with both hypercapnia and a transient acidosis Citation[43].

In contrast, we have used the carunclectomy model to induce chronic fetal hypoxemia and hypoglycemia across the last 4 weeks of gestation in the sheep Citation[36,44,45]. In this model, we have shown that fetal hypoxemia decreases gene and protein expression of SP-A, -B and -C Citation[46]. Hypoxia stabilizes a potent transcription factor, HIF-1α, which is essential for the differentiation of type II alveolar epithelial cells and SP expression Citation[47]. However, it appears that chronic, as opposed to acute hypoxia, have differing effects on the stability of the HIF-1 complex Citation[48]. This may explain differences in SP expression in the uteroplacental embolization and carunclectomy sheep models of IUGR. It is therefore possible that the different causes of IUGR and/or hypoxemia induce different fetal adaptations that lead to different changes in SP expression. Clearly the timing of the hypoxemic insult relative to gestational age, the duration of hypoxemia and the impact on plasma cortisol or exogenous GCs are all crucial in eliciting the SP or SP mRNA expression response, and hence lung maturation.

Impact of IUGR & antenatal GCs on the cardiovascular system

The fetus adapts to a decreased substrate supply by slowing its growth, as well as undergoing important cardiovascular adaptations Citation[44,45]. In particular, there is a redistribution of blood flow to the brain, adrenals and heart at the expense of the peripheral organs in order to survive in a suboptimal environment. This results in relative brain sparing Citation[45,49–51]. However, the important issue is that antenatal GCs cause cardiovascular changes in normally grown fetuses, including decreased fetal cerebral blood flow Citation[25], decreased brain growth and increased blood pressure, that are contrary to the cardiovascular adaptations that the IUGR fetus must make to survive. Hence, there is a possibility that antenatal GC in IUGR infants may compromise cardiovascular development. In fact, in a recent study in a single umbilical artery ligation sheep model of IUGR, β-methasone caused an equivalent fall in carotid artery blood flow in IUGR fetuses, but a larger rebound increase that was not observed in control fetuses, and ultimately led to increased cerebral oxidative damage in IUGR fetuses Citation[52]. In addition, studies in humans suggest that IUGR fetuses with absent end-diastolic blood flow have no change in middle cerebral artery blood flow in the 48 h after β-methasone treatment Citation[53].

Conclusion

The ability of the IUGR fetus to spare brain growth is key to surviving in an intrauterine environment with a reduced substrate supply. Exposure to GCs, which alter the fetus’ ability to regulate blood pressure and, thus, blood flow, are likely to cause further decreases in fetal body and brain growth. Evidence from sheep models of IUGR suggest that the timing, duration and severity of placental insufficiency may be key indicators in the fetal surfactant response to a reduced substrate supply and the effectiveness of antenatal GCs in improving lung function and, thus, reducing the incidence of neonatal morbidity due to respiratory distress syndrome. The impact of single versus multiple doses of antenatal GC in the IUGR fetus is, for the aforementioned reasons, an even greater concern. These questions must be interrogated with randomized controlled studies to determine both short- and long-term outcomes in infants, and with mechanistic studies in animal models of human IUGR.

Financial & competing interests disclosure

Sandra Orgeig acknowledges research funding from the Australian Research Council (DP0666086). Janna L Morrison was supported by fellowships from the Heart Foundation (PF 03A 1283 and CR 07A 3328), and the National Health and Medical Research Council (Biomedical CDA 511341). Janna L Morrison acknowledges research funding from the National Health and Medical Research Council of Australia (456418 and 456421). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Laws PJ, Hilder L. Australia’s mothers and babies 2006. Perinatal Statistics Series (AIHW cat. no. PER 46), 1–104 (2008).
  • Moss TJ, Doherty DA, Nitsos I, Sloboda DM, Harding R, Newnham JP. Effects into adulthood of single or repeated antenatal corticosteroids in sheep. Am. J. Obstet. Gynecol.192(1), 146–152 (2005).
  • St John EB, Nelson KG, Cliver SP, Bishnoi RR, Goldenberg RL. Cost of neonatal care according to gestational age at birth and survival status. Am. J. Obstet. Gynecol.182(1 Pt 1), 170–175 (2000).
  • Gilbert WM, Danielsen B. Pregnancy outcomes associated with intrauterine growth restriction. Am. J. Obstet. Gynecol.188(6), 1596–1599; discussion 1599–1601 (2003).
  • Liggins GC. Premature delivery of foetal lambs infused with glucocorticoids. Endocrinology45, 515–523 (1969).
  • Crowley PA. Antenatal corticosteroid therapy: a meta-analysis of the randomized trials, 1972 to 1994. Am. J. Obstet. Gynecol.173(1), 322–335 (1995).
  • Elimian A, Verma U, Canterino J, Shah J, Visintainer P, Tejani N. Effectiveness of antenatal steroids in obstetric subgroups. Obstet. Gynecol.93(2), 174–179 (1999).
  • National Institutes of Health Consensus Development Panel. Antenatal Corticosteroids Revisited: Repeat Courses – National Institutes of Health Consensus Development Conference Statement, August 17–18, 2000. Obstet. Gynecol.98(1), 144–150 (2001).
  • Bernstein IM, Horbar JD, Badger GJ, Ohlsson A, Golan A. Morbidity and mortality among very-low-birth-weight neonates with intrauterine growth restriction. The Vermont Oxford Network. Am. J. Obstet. Gynecol.182(1 Pt 1), 198–206 (2000).
  • Ikegami M, Polk D, Jobe A. Minimum interval from fetal β-methasone treatment to postnatal lung responses in preterm lambs. Am. J. Obstet. Gynecol.174(5), 1408–1413 (1996).
  • Jobe AH, Ikegami M. Lung development and function in preterm infants in the surfactant treatment era. Annu. Rev. Physiol.62(1), 825–846 (2000).
  • Modi N, Lewis H, Al-Naqeeb N, Ajayi-Obe M, Dore CJ, Rutherford M. The effects of repeated antenatal glucocorticoid therapy on the developing brain. Pediatr. Res.50(5), 581–585 (2001).
  • Brocklehurst P, Gates S, McKenzie-McHarg K, Alfirevic Z, Chamberlain G. Are we prescribing multiple courses of antenatal corticosteroids? A survey of practice in the UK. Br. J. Obstet. Gynaecol.106(9), 977–979 (1999).
  • Quinlivan JA, Evans SF, Dunlop SA, Beazley LD, Newnham JP. Use of corticosteroids by Australian obstetricians – a survey of clinical practice. Aust. N. Z. J. Obstet. Gynaecol.38(1), 1–7 (1998).
  • Crowther CA, Haslam RR, Hiller JE, Doyle LW, Robinson JS. Neonatal respiratory distress syndrome after repeat exposure to antenatal corticosteroids: a randomised controlled trial. Lancet367(9526), 1913–1919 (2006).
  • Roberts D, Dalziel S. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst. Rev.3, CD004454 (2006).
  • Crowther CA, Harding J. Repeat doses of prenatal corticosteroids for women at risk of preterm birth for preventing neonatal respiratory disease. Cochrane Database Syst. Rev. (3), CD003935 (2003).
  • Murphy KE, Hannah ME, Willan AR et al. Multiple courses of antenatal corticosteroids for preterm birth (MACS): a randomised controlled trial. Lancet372(9656), 2143–2151 (2008).
  • Polglase GR, Nitsos I, Jobe AH, Newnham JP, Moss TJ. Maternal and intra-amniotic corticosteroid effects on lung morphometry in preterm lambs. Pediatr. Res.62(1), 32–36 (2007).
  • Ueda T, Ikegami M, Polk D, Mizuno K, Jobe A. Effects of fetal corticosteroid treatments on postnatal surfactant function in preterm lambs. J. Appl. Physiol.79, 846–851 (1995).
  • Ballard PL, Ning Y, Polk D, Ikegami M, Jobe AH. Glucocorticoid regulation of surfactant components in immature lambs. Am. J. Physiol.273(5 Pt 1), L1048–L1057 (1997).
  • Willet KE, Jobe AH, Ikegami M, Kovar J, Sly PD. Lung morphometry after repetitive antenatal glucocorticoid treatment in preterm sheep. Am. J. Respir. Crit. Care Med.163(6), 1437–1443 (2001).
  • Walfisch A, Hallak M, Mazor M. Multiple courses of antenatal steroids: risks and benefits. Obstet. Gynecol.98(3), 491–497 (2001).
  • Schaap AH, Wolf H, Bruinse HW, Smolders-De Haas H, Van Ertbruggen I, Treffers PE. Effects of antenatal corticosteroid administration on mortality and long-term morbidity in early preterm, growth-restricted infants. Obstet. Gynecol.97(6), 954–960 (2001).
  • Schwab M, Roedel M, Anwar MA et al. Effects of bmethasone administration to the fetal sheep in late gestation on fetal cerebral blood flow. J. Physiol.528(Pt 3), 619–632 (2000).
  • Huang WL, Harper CG, Evans SF, Newnham JP, Dunlop SA. Repeated prenatal corticosteroid administration delays myelination of the corpus callosum in fetal sheep. Int. J. Dev. Neurosci.19(4), 415–425 (2001).
  • Sloboda DM, Newnham JP, Challis JR. Effects of repeated maternal β-methasone administration on growth and hypothalamic–pituitary–adrenal function of the ovine fetus at term. J. Endocrinol.165(1), 79–91 (2000).
  • Sloboda DM, Newnham JP, Challis JR. Repeated maternal glucocorticoid administration and the developing liver in fetal sheep. J. Endocrinol.175(2), 535–543 (2002).
  • Baud O, Sola A. Corticosteroids in perinatal medicine: how to improve outcomes without affecting the developing brain? Semin. Fetal Neonatal Med.12(4), 273–279 (2007).
  • Schwab M, Coksaygan T, Nathanielsz PW. bmethasone effects on ovine uterine and umbilical placental perfusion at the dose used to enhance fetal lung maturation. Am. J. Obstet. Gynecol.194(2), 572–579 (2006).
  • Huang WL, Beazley LD, Quinlivan JA, Evans SF, Newnham JP, Dunlop SA. Effect of corticosteroids on brain growth in fetal sheep. Obstet. Gynecol.94(2), 213–218 (1999).
  • Anwar MA, Schwab M, Poston L, Nathanielsz PW. bmethasone-mediated vascular dysfunction and changes in hematological profile in the ovine fetus. Am. J. Physiol.276(4 Pt 2), H1137–H1143 (1999).
  • Edwards LJ, Simonetta G, Owens JA, Robinson JS, McMillen IC. Restriction of placental and fetal growth in sheep alters fetal blood pressure responses to angiotensin II and captopril. J. Physiol.515(Pt 3), 897–904 (1999).
  • Economides DL, Nicolaides KH. Blood glucose and oxygen tension levels in small-for-gestational-age fetuses. Am. J. Obstet. Gynecol.160(2), 385–389 (1989).
  • Owens JA, Kind KL, Carbone F, Robinson JS, Owens PC. Circulating insulin-like growth factors-I and -II and substrates in fetal sheep following restriction of placental growth. J. Endocrinol.140(1), 5–13 (1994).
  • Morrison JL, Botting KJ, Dyer JL et al.Restriction of placental function alters heart development in the sheep fetus. Am. J. Physiol. Regul. Integr. Comp. Physiol.293(1), R306–R313 (2007).
  • Economides DL, Nicolaides KH, Linton EA, Perry LA, Chard T. Plasma cortisol and adrenocorticotropin in appropriate and small for gestational age fetuses. Fetal Ther.3(3), 158–164 (1988).
  • Phillips ID, Simonetta G, Owens JA, Robinson JS, Clarke IJ, McMillen IC. Placental restriction alters the functional development of the pituitary–adrenal axis in the sheep fetus during late gestation. Pediatr. Res.40(6), 861–866 (1996).
  • Lipsett J, Tamblyn M, Madigan K et al. Restricted fetal growth and lung development: a morphometric analysis of pulmonary structure. Pediatr. Pulmonol.41(12), 1138–1145 (2006).
  • Rees S, Ng J, Dickson K, Nicholas T, Harding R. Growth retardation and the development of the respiratory system in fetal sheep. Early Hum. Dev.26(1), 13–27 (1991).
  • Braems GA, Yao LJ, Inchley K et al. Ovine surfactant protein cDNAs: use in studies on fetal lung growth and maturation after prolonged hypoxemia. Am. J. Physiol.278(4), L754–L764 (2000).
  • Gagnon R, Langridge J, Inchley K, Murotsuki J, Possmayer F. Changes in surfactant-associated protein mRNA profile in growth-restricted fetal sheep. Am. J. Physiol.276(3 Pt 1), L459–L465 (1999).
  • Nardo L, Zhao L, Green L, Possmayer F, Richardson BS, Bocking AD. The effect of repeated umbilical cord occlusions on pulmonary surfactant protein mRNA levels in the ovine fetus. J. Soc. Gynecol. Investig.12(7), 510–517 (2005).
  • Morrison JL. Sheep models of intrauterine growth restriction: fetal adaptations and consequences. Clin. Exp. Pharmacol. Physiol.35(7), 730–743 (2008).
  • McMillen IC, Adams MB, Ross JT et al. Fetal growth restriction: adaptations and consequences. Reproduction122(2), 195–204 (2001).
  • Orgeig S, Crittenden TA, Marchant C, McMillen IC, Morrison JL. Intrauterine growth restriction delays surfactant protein maturation in the sheep fetus. Am. J. Physiol. Lung Cell. Mol. Physiol. (2010) (Epub ahead of print).
  • Saini Y, Harkema JR, LaPres JJ. HIF1a is essential for normal intrauterine differentiation of alveolar epithelium and surfactant production in the newborn lung of mice. J. Biol. Chem.283(48), 33650–33657 (2008).
  • Ginouves A, Ilc K, Macias N, Pouyssegur J, Berra E. PHDs overactivation during chronic hypoxia ‘desensitizes’ HIFα and protects cells from necrosis. Proc. Natl Acad. Sci. USA105(12), 4745–4750 (2008).
  • Nathanielsz PW, Hanson MA. The fetal dilemma: spare the brain and spoil the liver. J. Physiol.548(Pt 2), 333 (2003).
  • Dubiel M, Gunnarsson GO, Gudmundsson S. Blood redistribution in the fetal brain during chronic hypoxia. Ultrasound Obstet. Gynecol.20(2), 117–121 (2002).
  • Cheema R, Dubiel M, Gudmundsson S. Fetal brain sparing is strongly related to the degree of increased placental vascular impedance. J. Perinat. Med.34(4), 318–322 (2006).
  • Miller SL, Chai M, Loose J et al. The effects of maternal β-methasone administration on the intrauterine growth-restricted fetus. Endocrinology148(3), 1288–1295 (2007).
  • Nozaki AM, Francisco RP, Fonseca ES, Miyadahira S, Zugaib M. Fetal hemodynamic changes following maternal bmethasone administration in pregnancies with fetal growth restriction and absent end-diastolic flow in the umbilical artery. Acta Obstet. Gynecol. Scand.88(3), 350–354 (2009).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.