Antenatal melatonin as an antioxidant in human pregnancies complicated by fetal growth restriction--a phase I pilot clinical trial: study protocol

Nicole O Alers, Graham Jenkin, Suzanne L Miller, Euan M Wallace, Nicole O Alers, Graham Jenkin, Suzanne L Miller, Euan M Wallace

Abstract

Background: Fetal growth restriction complicates about 5% of pregnancies and is commonly caused by placental dysfunction. It is associated with increased risks of perinatal mortality and short-term and long-term morbidity, such as cerebral palsy. Chronic in utero hypoxaemia, inflammation and oxidative stress are likely culprits contributing to the long-term neurological sequelae of fetal growth restriction. In this regard, we propose that melatonin, a powerful antioxidant, might mitigate morbidity and/or mortality associated with fetal growth restriction. Melatonin has an excellent biosafety profile and crosses the placenta and blood-brain barrier. We present the protocol for a phase I clinical trial to investigate the efficacy of maternal oral melatonin administration in women with a pregnancy complicated by fetal growth restriction.

Methods and analysis: The proposed trial is a single-arm, open-label clinical trial involving 12 women. Severe, early onset fetal growth restriction will be diagnosed by an estimated fetal weight ≤10th centile in combination with abnormal fetoplacental Doppler studies, occurring before 34 weeks of pregnancy. Baseline measurements of maternal and fetal well-being, levels of oxidative stress and ultrasound and Doppler measurements will be obtained at the time of diagnosis of fetal growth restriction. Women will then start melatonin treatment (4 mg) twice daily until birth. The primary outcomes are the levels of oxidative stress in the maternal and fetal circulation and placenta. Secondary outcomes are fetoplacental Doppler studies (uterine artery, umbilical artery middle cerebral artery and ductus venosus), fetal biometry, fetal biophysical profile and a composite determination of neonatal outcome. A historical cohort of gestational-matched fetal growth restriction and a healthy pregnancy cohort will be used as comparators.

Ethics and dissemination: Ethical approval has been obtained from Monash Health Human Research Ethics Committee B (HREC12133B). Data will be presented at international conferences and published in peer-reviewed journals.

Trial registration number: Clinical Trials, protocol registration system: NCT01695070.

Keywords: OBSTETRICS.

References

    1. Robert R. Intrauterine growth restriction. Obstet Gynecol 2002;99:490–6
    1. Warkany J, Monroe BB, Sutherland BS. Intrauterine growth retardation. Am J Dis Child 1961;102:249–79
    1. Chase HC. A study of infant mortality from linked records: comparison of neonatal mortality from two cohort studies. United States, January-March 1950 and 1960. Vital Health Stat 20 Data Natl Vital Stat Syst 1972;13:1–99
    1. Ghidini A. Doppler of the ductus venosus in severe preterm fetal growth restriction. Obstet Gynecol 2007;109(2 Pt 1):250–2
    1. Bernstein IM, Horbar JD, Badger GJ, et al. Morbidity and mortality among very-low-birth-weight neonates with intrauterine growth restriction. The Vermont Oxford Network. Am J Obstet Gynecol 2000;182(1 Pt 1):198–206
    1. Ross MG, Beall MH. Adult sequelae of intrauterine growth restriction. Semin Perinatol 2008;32:213–18
    1. Barker DJ. In utero programming of chronic disease. Clin Sci Lond Engl 1979 1998;95:115–28
    1. Miller SL, Wallace EM. Effect of antenatal steroids on haemodynamics in the normally grown and growth restricted fetus. Curr Pediatr Rev 2013;9:67–74
    1. Miller SL, Wallace EM, Walker DW. Antioxidant therapies: a potential role in perinatal medicine. Neuroendocrinology. 2012;96:13–23
    1. Love S. Oxidative stress in brain ischemia. Brain Pathol Zurich Switz 1999;9:119–31
    1. Halliwell B. Reactive oxygen species and the central nervous system. J Neurochem 1992;59:1609–23
    1. Morrow JD, Minton TA, Roberts LJ., II The F2-isoprostane, 8-epi-prostaglandin F2α, a potent agonist of the vascular thromboxane/endoperoxide receptor, is a platelet thromboxane/endoperoxide receptor antagonist. Prostaglandins 1992;44:155–63
    1. Schmidt H, Grune T, Müller R, et al. Increased levels of lipid peroxidation products malondialdehyde and 4-hydroxynonenal after perinatal hypoxia. Pediatr Res 1996;40:15–20
    1. Wentzel P, Eriksson UJ. 8-Iso-PGF2α administration generates dysmorphogenesis and increased lipid peroxidation in rat embryos in vitro. Teratology 2002;66:164–8
    1. Marnett LJ. Lipid peroxidation–DNA damage by malondialdehyde. Mutat Res Mol Mech Mutagen 1999;424:83–95
    1. Inder T, Mocatta T, Darlow B, et al. Elevated free radical products in the cerebrospinal fluid of VLBW infants with cerebral white matter injury. Pediatr Res 2002;52:213–18
    1. Haynes RL, Folkerth RD, Keefe RJ, et al. Nitrosative and oxidative injury to premyelinating oligodendrocytes in periventricular leukomalacia. J Neuropathol Exp Neurol 2003;62:441–50
    1. Pablos MI, Chuang J, Reiter RJ, et al. Time course of the melatonin-induced increase in glutathione peroxidase activity in chick tissues. Biol Signals 1995;4:325–30
    1. Pozo D, Reiter RJ, Calvo JR, et al. Inhibition of cerebellar nitric oxide synthase and cyclic GMP production by melatonin via complex formation with calmodulin. J Cell Biochem 1997;65:430–42
    1. García JJ, Reiter RJ, Guerrero JM, et al. Melatonin prevents changes in microsomal membrane fluidity during induced lipid peroxidation. FEBS Lett 1997;408:297–300
    1. Lanoix D, Lacasse A-A, Reiter RJ, et al. Melatonin: The watchdog of villous trophoblast homeostasis against hypoxia/reoxygenation-induced oxidative stress and apoptosis. Mol Cell Endocrinol 2013;381:35–45
    1. Okatani Y, Okamoto K, Hayashi K, et al. Maternal-fetal transfer of melatonin in pregnant women near term. J Pineal Res 1998;25: 129–34
    1. Okatani Y, Wakatsuki A, Kaneda C. Melatonin increases activities of glutathione peroxidase and superoxide dismutase in fetal rat brain. J Pineal Res 2000;28:89–96
    1. Torres-Farfan C, Valenzuela FJ, Mondaca M, et al. Evidence of a role for melatonin in fetal sheep physiology: direct actions of melatonin on fetal cerebral artery, brown adipose tissue and adrenal gland. J Physiol 2008;586:4017–27
    1. Thakor AS, Herrera EA, Serón Ferré M, et al. Melatonin and vitamin C increase umbilical blood flow via nitric oxide dependent mechanisms. J Pineal Res 2010;49:399–406
    1. Miller SL, Yan EB, Castillo-Melendez M, et al. Melatonin provides neuroprotection in the late-gestation fetal sheep brain in response to umbilical cord occlusion. Dev Neurosci 2005;27:200–10
    1. Yawno T, Castillo-Melendez M, Jenkin G, et al. Mechanisms of melatonin-induced protection in the brain of late gestation fetal sheep in response to hypoxia. Dev Neurosci 2012;34:543–51
    1. Welin A-K, Svedin P, Lapatto R, et al. Melatonin reduces inflammation and cell death in white matter in the mid-gestation fetal sheep following umbilical cord occlusion. Pediatr Res 2007;61:153–8
    1. Supramaniam VG, Jenkin G, Loose J, et al. Chronic fetal hypoxia increases activin A concentrations in the late-pregnant sheep. BJOG Int J Obstet Gynaecol 2006;113:102–9
    1. Unfer V, Raffone E, Rizzo P, et al. Effect of a supplementation with myo-inositol plus melatonin on oocyte quality in women who failed to conceive in previous in vitro fertilization cycles for poor oocyte quality: a prospective, longitudinal, cohort study. Gynecol Endocrinol 2011;27:857–61
    1. Tamura H, Nakamura Y, Terron MP, et al. Melatonin and pregnancy in the human. Reprod Toxicol 2008;25:291–303
    1. Benedetto PPRERV Effect of the treatment with myo-inositol plus folic acid plus melatonin in comparison with a treatment with myo-inositol plus folic acid on oocyte quality and pregnancy outcome in IVF cycles. A prospective, clinical trial. Eur Rev Med Pharmacol Sci 2010;14:555.
    1. Barchas J, Dacosta F, Spector S. Acute pharmacology of melatonin. Nature 1967;214:919–20
    1. Silman RE. Melatonin: a contraceptive for the nineties. Eur J Obstet Gynecol Reprod Biol 1993;49:3–9
    1. Jahnke G, Marr M, Myers C, et al. Maternal and developmental toxicity evaluation of melatonin administered orally to pregnant Sprague-Dawley rats. Toxicol Sci Off J Soc Toxicol 1999;50:271–9
    1. U.S. Food and Drug Administration, Guidance for Industry Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers, Pharmacology and Toxicology, 2005,
    1. Gitto E, Reiter RJ, Cordaro SP, et al. Oxidative and inflammatory parameters in respiratory distress syndrome of preterm newborns: beneficial effects of melatonin. Am J Perinatol 2004;21:209–16
    1. Gitto E, Romeo C, Reiter RJ, et al. Melatonin reduces oxidative stress in surgical neonates. J Pediatr Surg 2004;39:184–9; discussion 184–9
    1. Gitto E, Karbownik M, Reiter RJ, et al. Effects of melatonin treatment in septic newborns. Pediatr Res 2001;50:756.
    1. Fulia F, Gitto E, Cuzzocrea S, et al. Increased levels of malondialdehyde and nitrite/nitrate in the blood of asphyxiated newborns: reduction by melatonin. J Pineal Res 2001;31:343–9
    1. Gitto E, Aversa S, Salpietro CD, et al. Pain in neonatal intensive care: role of melatonin as an analgesic antioxidant. J Pineal Res.2012;52:291–5
    1. Tamura H, Takasaki A, Miwa I, et al. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. J Pineal Res 2008;44:280–7
    1. Hobson SR, Lim R, Gardiner EE, et al. Phase I pilot clinical trial of antenatal maternally administered melatonin to decrease the level of oxidative stress in human pregnancies affected by pre-eclampsia (PAMPR): study protocol. BMJ Open 2013;3:e003788

Source: PubMed

3
Tilaa