Neuroprotective therapies in the NICU in term infants: present and future

Abstract

Outcomes of neonatal encephalopathy (NE) have improved since the widespread implementation of therapeutic hypothermia (TH) in high-resource settings. While TH for NE in term and near-term infants has proven beneficial, 30–50% of infants with moderate-tosevere NE treated with TH still suffer death or significant impairments. There is therefore a critical need to find additional pharmacological and non-pharmacological interventions that improve the outcomes for these children. There are many potential candidates; however, it is unclear whether these interventions have additional benefits when used with TH. Although primary and delayed (secondary) brain injury starting in the latent phase after HI are major contributors to neurodisability, the very late evolving effects of tertiary brain injury likely require different interventions targeting neurorestoration. Clinical trials of seizure management and neuroprotection bundles are needed, in addition to current trials combining erythropoietin, stem cells, and melatonin with TH.

Molloy E.J., El-Dib M., Juul S.E., et al. Neuroprotective therapies in the NICU in term infants: present and future. Pediatr Res (2022). DOI: https://doi.org/10.1038/s41390-022-02295-2

PMID: 36195634.

* A list of authors and their affiliations appears at the end of the paper.



REFERENCES

1. Liu L., et al. Global, regional, and national causes of under-5 mortality in 2000–15: an updated systematic analysis with implications for the sustainable development goals. Lancet. 2016; 388: 3027–35.

2. Fleiss B., Gressens P. Tertiary mechanisms of brain damage: a new hope for treatment of cerebral palsy? Lancet Neurol. 2012; 11: 556–66.

3. Zareen Z., et al. Cytokine dysregulation persists in childhood post neonatal encephalopathy. BMC Neurol. 2020; 20: 115.

4. Azzopardi D., et al. Implementation and conduct of therapeutic hypothermia for perinatal asphyxial encephalopathy in the UK – analysis of national data. PLoS One. 2012; 7: e38504.

5. Shankaran S., et al. Neonatal magnetic resonance imaging pattern of brain injury as a biomarker of childhood outcomes following a trial of hypothermia for neonatal hypoxic-ischemic encephalopathy. J Pediatr. 2015; 167: 987.e3–93.e3.

6. Jacobs S.E., et al. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev. 2013. 2013: CD003311.

7. Natarajan G., Pappas A., Shankaran S. Outcomes in childhood following therapeutic hypothermia for neonatal hypoxic-ischemic encephalopathy (HIE). Semin Perinatol. 2016; 40: 549–55.

8. Wassink G., Gunn E.R., Drury P.P., Bennet L., Gunn A.J. The mechanisms and treatment of asphyxial encephalopathy. Front Neurosci. 2014; 8: 40.

9. Davidson J.O., et al. How long is sufficient for optimal neuroprotection with cerebral cooling after ischemia in fetal sheep? J Cereb Blood Flow Metab. 2018; 38: 1047–59.

10. Alonso-Alconada D., et al. Brain cell death is reduced with cooling by 3.5°C to 5°C but increased with cooling by 8.5°C in a piglet asphyxia model. Stroke. 2015; 46: 275–8.

11. Davidson J.O., et al. How long is too long for cerebral cooling after ischemia in fetal sheep? J Cereb Blood Flow Metab. 2015; 35: 751–8.

12. Shankaran S., et al. Effect of depth and duration of cooling on death or disability at age 18 months among neonates with hypoxic-ischemic encephalopathy: a randomized clinical trial. JAMA. 2017; 318: 57–67.

13. Edwards A.D., et al. Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of trial data. BMJ. 2010; 340: c363.

14. Laptook A.R., et al. Effect of therapeutic hypothermia initiated after 6 h of age on death or disability among newborns with hypoxic-ischemic encephalopathy: a randomized clinical trial. JAMA. 2017; 318: 1550–60.

15. Thayyil S., et al. Hypothermia for moderate or severe neonatal encephalopathy in low-income and middle-income countries (HELIX): a randomised controlled trial in India, Sri Lanka, and Bangladesh. Lancet Glob Health. 2021; 9: e1273–85.

16. Wassink G., et al. A working model for hypothermic neuroprotection. J Physiol. 2018; 596: 5641–54.

17. Aslam S., Strickland T., Molloy E.J. Neonatal encephalopathy: need for recognition of multiple etiologies for optimal management. Front Pediatr. 2019; 7: 142.

18. Calkavur S., et al. Genetic factors that influence short-term neurodevelopmental outcome in term hypoxic-ischaemic encephalopathic neonates. J Int Med Res. 2011; 39: 1744–56.

19. Tan E.S. Inborn errors of metabolism presenting as neonatal encephalopathy: practical tips for clinicians. Ann Acad Med Singapore. 2008; 37: 94–3.

20. Wood S., Crawford S., Hicks M., Mohammad K. Hospital-related, maternal, and fetal risk factors for neonatal asphyxia and moderate or severe hypoxicischemic encephalopathy: a retrospective cohort study. J Matern Fetal Neonatal Med. 2021; 34: 1448–53.

21. Tann C.J., et al. Perinatal risk factors for neonatal encephalopathy: an unmatched case-control study. Arch Dis Child Fetal Neonatal Ed. 2018; 103: F250–6.

22. Rossi A.C., Prefumo F. Antepartum and intrapartum risk factors for neonatal hypoxic-ischemic encephalopathy: a systematic review with meta-analysis. Curr Opin Obstet Gynecol. 2019; 31: 410–7.

23. Oliveira V., et al. Hypothermia for encephalopathy in low-income and middle-income countries: feasibility of whole-body cooling using a low-cost servo-controlled device. BMJ Paediatr Open. 2018; 2: e000245.

24. Kofke W.A. Incrementally applied multifaceted therapeutic bundles in neuroprotection clinical trials… time for change. Neurocrit Care. 2010; 12: 438–44.

25. Williams K.P., Galerneau F. Intrapartum fetal heart rate patterns in the prediction of neonatal acidemia. Am J Obstet Gynecol. 2003; 188: 820–3.

26. Lingappan K., Kaiser J.R., Srinivasan C., Gunn A.J. Relationship between PCO2 and unfavorable outcome in infants with moderate-to-severe hypoxic ischemic encephalopathy. Pediatr Res. 2016; 80: 204–8.

27. Ivy A.S., Clark C.L., Bahm S.M., Meurs K.P., Wusthoff C.J. Improving the identification of neonatal encephalopathy: utility of a web-based video tool. Am J Perinatol. 2017; 34: 520–2.

28. Mohammad K., et al. Hemodynamic instability associated with increased risk of death or brain injury in neonates with hypoxic ischemic encephalopathy. J Neonatal Perinat Med. 2017; 10: 363–70.

29. Ilves P., Kiisk M., Soopõld T., Talvik T. Serum total magnesium and ionized calcium concentrations in asphyxiated term newborn infants with hypoxicischaemic encephalopathy. Acta Paediatr. 2000; 89: 680–5.

30. Wintermark P., Mohammad K., Bonifacio S.L. Proposing a care practice bundle for neonatal encephalopathy during therapeutic hypothermia. Semin Fetal Neonatal Med. 2021; 26: 101303.

31. Roychoudhury S., et al. Implementation of neonatal neurocritical care program improved short-term outcomes in neonates with moderate-to-severe hypoxic ischemic encephalopathy. Pediatr Neurol. 2019; 101: 64–70.

32. Bashir R.A., et al. Implementation of a neurocritical care program: improved seizure detection and decreased antiseizure medication at discharge in neonates with hypoxic-ischemic encephalopathy. Pediatr Neurol. 2016; 64: 38–43.

33. Goswami I., et al. Introduction of continuous video EEG monitoring into 2 different NICU models by training neonatal nurses. Adv Neonatal Care. 2018; 18: 250–9.

34. Mohammad K., et al. Impact of outreach education program on outcomes of neonates with hypoxic ischemic encephalopathy. Paediatr Child Health. 2020; 26: e215–21.

35. Arriagada S., Huang H., Fletcher K., Giannone P. Prevention of excessive hypothermia in infants with hypoxic ischemic encephalopathy prior to admission to a quaternary care center: a Neonatal Outreach Educational Project. J Perinatol. 2019; 39: 1417–27.

36. Cramer S.C., et al. Harnessing neuroplasticity for clinical applications. Brain. 2011; 134: 1591–609.

37. O’Brien K., et al. Effectiveness of family integrated care in neonatal intensive care units on infant and parent outcomes: a multicentre, multinational, cluster-randomised controlled trial. Lancet Child Adolesc Health, 2018; 2: 245–54.

38. DeMaster D., et al. Nurturing the preterm infant brain: leveraging neuroplasticity to improve neurobehavioral outcomes. Pediatr Res. 2019; 85: 166–75.

39. Maguire C.M., et al. Effects of individualized developmental care in a randomized trial of preterm infants <32 weeks. Pediatrics. 2009; 124: 1021–30.

40. Procianoy R.S., Mendes E.W., Silveira R.C. Massage therapy improves neurodevelopment outcome at two years corrected age for very low birth weight infants. Early Hum Dev. 2010; 86: 7–11.

41. Feldman R., Rosenthal Z., Eidelman A.I. Maternal-preterm skin-to-skin contact enhances child physiologic organization and cognitive control across the first 10 years of life. Biol Psychiatry. 2014; 75: 56–64.

42. Chorna O., et al. Neuroprocessing mechanisms of music during fetal and neonatal development: a role in neuroplasticity and neurodevelopment. Neural Plast. 2019. 2019; 3972918.

43. Braid S., Bernstein J. Improved cognitive development in preterm infants with shared book reading. Neonatal Netw. 2015; 34: 10–7.

44. Shellhaas R.A., Burns J.W., Barks J.D.E., Hassan F., Chervin R.D. Maternal voice and infant sleep in the neonatal intensive care unit. Pediatrics. 2019; 144: e20190288.

45. Smith S.W., Ortmann A.J., Clark W.W. Noise in the neonatal intensive care unit: a new approach to examining acoustic events. Noise Health. 2018; 20: 121–30.

46. Shellhaas R.A., et al. Neonatal sleep-wake analyses predict 18-month neurodevelopmental outcomes. Sleep. 2017; 40: zsx144.

47. Forcada-Guex M., Pierrehumbert B., Borghini A., Moessinger A., Muller-Nix C. Early dyadic patterns of mother-infant interactions and outcomes of prematurity at 18 months. Pediatrics. 2006; 118: e107–14.

48. Osmond E., et al. Neonatal seizures: magnetic resonance imaging adds value in the diagnosis and prediction of neurodisability. Acta Paediatr. 2014; 103: 820–6.

49. Soul J.S. Acute symptomatic seizures in term neonates: etiologies and treatments. Semin Fetal Neonatal Med. 2018; 23: 183–90.

50. Glass H.C., et al. Contemporary profile of seizures in neonates: a prospective cohort study. J Pediatr. 2016; 174: 98–103.e101.

51. Shellhaas R.A., et al. The American Clinical Neurophysiology Society’s Guideline on continuous electroencephalography monitoring in neonates. J Clin Neurophysiol. 2011; 28: 611–7.

52. Massey S.L., Jensen F.E., Abend N.S. Electroencephalographic monitoring for seizure identification and prognosis in term neonates. Semin Fetal Neonatal Med. 2018; 23: 168–74.

53. Pavel A.M., et al. A machine-learning algorithm for neonatal seizure recognition: a multicentre, randomised, controlled trial. Lancet Child Adolesc Health. 2020; 4: 740–9.

54. Hellström-Westas L., Boylan G., Agren J. Systematic review of neonatal seizure management strategies provides guidance on anti-epileptic treatment. Acta Paediatr. 2015; 104: 123–9.

55. Guidotti I., et al. Hypothermia reduces seizure burden and improves neurological outcome in severe hypoxic-ischemic encephalopathy: an observational study. Dev Med Child Neurol. 2016; 58: 1235–41.

56. Srinivasakumar P., et al. Therapeutic hypothermia in neonatal hypoxic ischemic encephalopathy: electrographic seizures and magnetic resonance imaging evidence of injury. J Pediatr. 2013; 163: 465–70.

57. Low E., et al. Cooling and seizure burden in term neonates: an observational study. Arch Dis Child Fetal Neonatal Ed. 2012; 97: F267.

58. Davidson J.O., et al. Non-additive effects of delayed connexin hemichannel blockade and hypothermia after cerebral ischemia in near-term fetal sheep. J Cereb Blood Flow Metab. 2015; 35: 2052–61.

59. Holmes G.L. The long-term effects of neonatal seizures. Clin Perinatol. 2009; 36: 901–4.

60. Wirrell E.C., Armstrong E.A., Osman L.D., Yager J.Y. Prolonged seizures exacerbate perinatal hypoxic-ischemic brain damage. Pediatr Res. 2001; 50: 445–54.

61. Yager J.Y., Armstrong E.A., Jaharus C., Saucier D.M., Wirrell E.C. Preventing hyperthermia decreases brain damage following neonatal hypoxic-ischemic seizures. Brain Res. 2004; 1011: 48–57.

62. Wyatt J.S., et al. Determinants of outcomes after head cooling for neonatal encephalopathy. Pediatrics. 2007; 119: 912–21.

63. Laptook A., et al. Elevated temperature after hypoxic-ischemic encephalopathy: risk factor for adverse outcomes. Pediatrics. 2008; 122: 491–9.

64. Tan W.K., Williams C.E., Gunn A.J., Mallard C.E., Gluckman P.D. Suppression of postischemic epileptiform activity with MK-801 improves neural outcome in fetal sheep. Ann Neurol. 1992; 32: 677–82.

65. Kharoshankaya L. et al. Seizure burden and neurodevelopmental outcome in neonates with hypoxic-ischemic encephalopathy. Dev Med Child Neurol. 2016; 58: 1242–8.

66. Glass H.C., et al. Clinical neonatal seizures are independently associated with outcome in infants at risk for hypoxic-ischemic brain injury. J Pediatr. 2009; 155: 318–23.

67. Srinivasakumar P., et al. Treating EEG seizures in hypoxic ischemic encephalopathy: a randomized controlled trial. Pediatrics. 2015; 136: e1302–9.

68. van Rooij L.G., et al. Effect of treatment of subclinical neonatal seizures detected with aEEG: randomized, controlled trial. Pediatrics. 2010; 125: e358–66.

69. Soul J.S., et al. Recommendations for the design of therapeutic trials for neonatal seizures. Pediatr Res. 2019; 85: 943–54.

70. Painter M.J., et al. Phenobarbital compared with phenytoin for the treatment of neonatal seizures. N Engl J Med. 1999; 341: 485–9.

71. El-Dib M., Soul J.S. The use of phenobarbital and other anti-seizure drugs in newborns. Semin Fetal Neonatal Med. 2017; 22: 321–7.

72. Soul J.S., et al. A pilot randomized, controlled, double-blind trial of bumetanide to treat neonatal seizures. Ann Neurol. 2021; 89: 327–40.

73. Sharpe C., et al. Levetiracetam versus phenobarbital for neonatal seizures: a randomized controlled trial. Pediatrics. 2020; 145: e20193182.

74. Boylan G.B., et al. Second-line anticonvulsant treatment of neonatal seizures: a video-EEG monitoring study. Neurology. 2004; 62: 486–8.

75. Castro Conde J.R., Hernández Borges A.A., Doménech Martínez E., González Campo C., Perera Soler R. Midazolam in neonatal seizures with no response to phenobarbital. Neurology. 2005; 64: 876–9.

76. Weeke L.C., et al. Lidocaine response rate in aEEG-confirmed neonatal seizures: retrospective study of 413 full-term and preterm infants. Epilepsia. 2016; 57: 233–42.

77. Pressler R.M., et al. Bumetanide for the treatment of seizures in newborn babies with hypoxic ischaemic encephalopathy (NEMO): an open-label, dose finding, and feasibility phase 1/2 trial. Lancet Neurol. 2015; 14: 469–77.

78. Koh S., Tibayan F.D., Simpson J.N., Jensen F.E. Nbqx or topiramate treatment after perinatal hypoxia-induced seizures prevents later increases in seizure-induced neuronal injury. Epilepsia. 2004; 45: 569–75.

79. Glass H.C., Poulin C., Shevell M.I. Topiramate for the treatment of neonatal seizures. Pediatr Neurol. 2011; 44: 439–42.

80. Nuñez-Ramiro A., et al. Topiramate plus cooling for hypoxic-ischemic encephalopathy: a randomized, controlled, multicenter, double-blinded trial. Neonatology. 2019; 116: 76–84.

81. van den Broek M.P., et al. Pharmacokinetics and clinical efficacy of phenobarbital in asphyxiated newborns treated with hypothermia: a thermopharmacological approach. Clin Pharmacokinet. 2012; 51: 671–9.

82. Shellhaas R.A., Ng C.M., Dillon C.H., Barks J.D., Bhatt-Mehta V. Population pharmacokinetics of phenobarbital in infants with neonatal encephalopathy treated with therapeutic hypothermia. Pediatr Crit Care Med. 2013; 14: 194–202.

83. Bittigau P., et al. Antiepileptic drugs and apoptotic neurodegeneration in the developing brain. Proc Natl Acad Sci USA. 2002; 99: 15 089–94.

84. Forcelli P.A., Kim J., Kondratyev A., Gale K. Pattern of antiepileptic drug-induced cell death in limbic regions of the neonatal rat brain. Epilepsia. 2011; 52: e207–11.

85. Zhou K.Q., et al. Treating seizures after hypoxic-ischemic encephalopathy-current controversies and future directions. Int J Mol Sci. 2021; 22: 7121.

86. Kilicdag H., et al. The effect of levetiracetam on neuronal apoptosis in neonatal ratmodel of hypoxic ischemic brain injury. Early Hum Dev. 2013; 89: 355–60.

87. Komur M., et al. Neuroprotective effect of levetiracetam on hypoxic ischemic brain injury in neonatal rats. Childs Nerv Syst. 2014; 30: 1001–9.

88. Gonzalez F.F., et al. Erythropoietin increases neurogenesis and oligodendrogliosis of subventricular zone precursor cells after neonatal stroke. Stroke. 2013; 44: 753–8.

89. Shingo T., Sorokan S.T., Shimazaki T., Weiss S. Erythropoietin regulates the in vitro and in vivo production of neuronal progenitors by mammalian forebrain neural stem cells. J Neurosci. 2001; 21: 9733–43.

90. O’Dea M.I. et al. Altered cytokine endotoxin responses in neonatal encephalopathy predict MRI outcomes. Front Pediatr. 2021; 9: 734540.

91. O’Hare F.M., et al. Serial cytokine alterations and abnormal neuroimaging in newborn infants with encephalopathy. Acta Paediatr. 2017; 106: 561–7.

92. Ruth V., Widness J.A., Clemons G., Raivio K.O. Postnatal changes in serum immunoreactive erythropoietin in relation to hypoxia before and after birth. J Pediatr. 1990; 116: 950–4.

93. Hagag A.A., El Frargy M.S., Abd El-Latif A.E. Study of cord blood erythropoietin, leptin and adiponectin levels in neonates with hypoxic ischemic encephalopathy. Endocr Metab Immune Disord Drug Targets. 2020; 20: 213–20.

94. Teramo K.A., Klemetti M.M., Widness J.A. Robust increases in erythropoietin production by the hypoxic fetus is a response to protect the brain and other vital organs. Pediatr Res. 2018; 84: 807–12.

95. van der Kooij M.A., Groenendaal F., Kavelaars A., Heijnen C.J., van Bel F. Neuroprotective properties and mechanisms of erythropoietin in in vitro and in vivo experimental models for hypoxia/ischemia. Brain Res Rev. 2008; 59: 22–33.

96. Kellert B.A., McPherson R.J., Juul S.E. A comparison of high-dose recombinant erythropoietin treatment regimens in brain-injured neonatal rats. Pediatr Res. 2007; 61: 451–5.

97. Weber A., et al. Neuronal damage after moderate hypoxia and erythropoietin. Neurobiol Dis. 2005; 20: 594–600.

98. Juul S.E., et al. Erytropoietin concentrations in cerebrospinal fluid of nonhuman primates and fetal sheep following high-dose recombinant erythropoietin. Biol Neonate. 2004; 85: 138–44.

99. Statler P.A., McPherson R.J., Bauer L.A., Kellert B.A., Juul S.E. Pharmacokinetics of high-dose recombinant erythropoietin in plasma and brain of neonatal rats. Pediatr Res. 2007; 61: 671–5.

100. Wassink G., et al. Recombinant erythropoietin does not augment hypothermic white matter protection after global cerebral ischaemia in near-term fetal sheep. Brain Commun. 2021; 3: fcab172.

101. Wassink G., et al. Non-additive effects of adjunct erythropoietin therapy with therapeutic hypothermia after global cerebral ischaemia in near-term fetal sheep. J Physiol. 2020; 598: 999–1015.

102. Malla R.R., Asimi R., Teli M.A., Shaheen F., Bhat M.A. Erythropoietin monotherapy in perinatal asphyxia with moderate to severe encephalopathy: a randomized placebo-controlled trial. J Perinatol. 2017; 37: 596–601.

103. Zhu C., et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics. 2009; 124: e218–26.

104. Wu Y.W., et al. High-dose erythropoietin and hypothermia for hypoxic-ischemic encephalopathy: a phase II trial. Pediatrics. 2016; 137: e20160191.

105. Massaro A.N., et al. Plasma biomarkers of brain injury in neonatal hypoxicischemic encephalopathy. J Pediatr. 2018; 194: 67.e61–75.e61.

106. Wu Y.W., et al. Trial of erythropoietin for hypoxic-ischemic encephalopathy in newborns. N Engl J Med. 2022; 387: 148–59.

107. University of Sidney, National Health and Medical Research Council, Australia. PAEAN – Erythropoietin for Hypoxic Ischaemic Encephalopathy in Newborns. 2016. URL: https://clinicaltrials.gov/ct2/show/NCT03079167

108. University of Utah, Darbe Administration in Newborns Undergoing Cooling for Encephalopathy. 2015. URL: https://clinicaltrials.gov/ct2/show/NCT01471015

109. University of New M. & University of U. Mild Encephalopathy in the Newborn Treated With Darbepoetin. 2021. URL: https://clinicaltrials.gov/ct2/show/NCT03071861

110. Kurtzberg J. A history of cord blood banking and transplantation. Stem Cells Transl Med. 2017; 6: 1309–11.

111. Wang X., et al. Effect of umbilical cord mesenchymal stromal cells on motor functions of identical twins with cerebral palsy: pilot study on the correlation of efficacy and hereditary factors. Cytotherapy. 2015; 17: 224–31.

112. Wang Y., Chen X., Cao W., Shi Y. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol. 2014; 15: 1009–16.

113. Cunningham C.J., Redondo-Castro E., Allan S.M. The therapeutic potential of the mesenchymal stem cell secretome in ischaemic stroke. J Cereb Blood Flow Metab. 2018; 38: 1276–92.

114. Fisher S.A., et al. Mesenchymal stromal cells as treatment or prophylaxis for acute or chronic graft-versus-host disease in haematopoietic stem cell transplant (HSCT) recipients with a haematological condition. Cochrane Database Syst Rev. 2019; 1: CD009768.

115. Vaes J.E.G., et al. The potential of stem cell therapy to repair white matter injury in preterm infants: lessons learned from experimental models. Front Physiol. 2019; 10: 540.

116. Vu Q., Xie K., Eckert M., Zhao W., Cramer S.C. Meta-analysis of preclinical studies of mesenchymal stromal cells for ischemic stroke. Neurology. 2014; 82: 1277–86.

117. Meier C., et al. Spastic paresis after perinatal brain damage in rats is reduced by human cord blood mononuclear cells. Pediatr Res. 2006; 59: 244–9.

118. Rosenkranz K., Meier C. Umbilical cord blood cell transplantation after brain ischemia – from recovery of function to cellular mechanisms. Ann Anat. 2011; 193: 371–9.

119. Rosenkranz K., et al. Transplantation of human umbilical cord blood cells mediated beneficial effects on apoptosis, angiogenesis and neuronal survival after hypoxic-ischemic brain injury in rats. Cell Tissue Res. 2012; 348: 429–38.

120. McDonald C.A., et al. Effects of umbilical cord blood cells, and subtypes, to reduce neuroinflammation following perinatal hypoxic-ischemic brain injury. J Neuroinflamm. 2018; 15: 47.

121. van Velthoven C.T., Kavelaars A., van Bel F., Heijnen C.J. Mesenchymal stem cell treatment after neonatal hypoxic-ischemic brain injury improves behavioral outcome and induces neuronal and oligodendrocyte regeneration. Brain Behav Immun. 2010; 24: 387–93.

122. van Velthoven C.T., Kavelaars A., van Bel F., Heijnen C.J. Repeated mesenchymal stem cell treatment after neonatal hypoxia-ischemia has distinct effects on formation and maturation of new neurons and oligodendrocytes leading to restoration of damage, corticospinal motor tract activity, and sensorimotor function. J Neurosci. 2010; 30: 9603–11.

123. Nair S., et al. Neuroprotection offered by mesenchymal stem cells in perinatal brain injury: role of mitochondria, inflammation, and reactive oxygen species. J Neurochem. 2021; 158: 59–73.

124. Kaminski N., et al. Mesenchymal stromal cell-derived extracellular vesicles reduce neuroinflammation, promote neural cell proliferation and improve oligodendrocyte maturation in neonatal hypoxic-ischemic brain injury. Front Cell Neurosci. 2020; 14: 601176.

125. Herz J., et al. Interaction between hypothermia and delayed mesenchymal stem cell therapy in neonatal hypoxic-ischemic brain injury. Brain Behav Immun. 2018; 70: 118–30.

126. Robertson N.J., et al. Human umbilical cord mesenchymal stromal cells as an adjunct therapy with therapeutic hypothermia in a piglet model of perinatal asphyxia. Cytotherapy. 2021; 23: 521–35.

127. Bernardo M.E., Fibbe W.E. Mesenchymal stromal cells and hematopoietic stem cell transplantation. Immunol Lett. 2015; 168: 215–21.

128. Cotten C.M., et al. Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr. 2014; 164: 973.e1–9.e1.

129. Tsuji M., et al. Autologous cord blood cell therapy for neonatal hypoxicischaemic encephalopathy: a pilot study for feasibility and safety. Sci Rep. 2020; 10: 4603.

130. Reiter R.J. Pineal melatonin: cell biology of its synthesis and of its physiological interactions. Endocr Rev. 1991; 12: 151–80.

131. Tarocco A., et al. Melatonin as a master regulator of cell death and inflammation: molecular mechanisms and clinical implications for newborn care. Cell Death Dis. 2019; 10: 317.

132. Ng K.Y., Leong M.K., Liang H., Paxinos G. Melatonin receptors: distribution in mammalian brain and their respective putative functions. Brain Struct Funct. 2017; 222: 2921–39.

133. Calvo J.R., González-Yanes C., Maldonado M.D. The role of melatonin in the cells of the innate immunity: a review. J Pineal Res. 2013; 55: 103–20.

134. Hardeland R. Melatonin and inflammation-story of a double-edged blade. J Pineal Res. 2018; 65: e12525.

135. Reiter R.J., et al. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016; 61: 253–78.

136. Sun F.Y., et al. Neuroprotection by melatonin against ischemic neuronal injury associated with modulation of DNA damage and repair in the rat following a transient cerebral ischemia. J Pineal Res. 2002; 33: 48–56.

137. Vriend J., Reiter R.J. Melatonin feedback on clock genes: a theory involving the proteasome. J Pineal Res. 2015; 58: 1–11.

138. Curtis A.M., Bellet M.M., Sassone-Corsi P., O’Neill L.A. Circadian clock proteins and immunity. Immunity. 2014; 40: 178–86.

139. Robertson N.J., et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain. 2013; 136: 90–105.

140. Aridas J.D., et al. Melatonin augments the neuroprotective effects of hypothermia in lambs following perinatal asphyxia. J Pineal Res. 2021; 71: e12744.

141. Pang R., et al. Melatonin and/or erythropoietin combined with hypothermia in a piglet model of perinatal asphyxia. Brain Commun. 2020; 3: fcaa211.

142. Robertson N.J., et al. Melatonin as an adjunct to therapeutic hypothermia in a piglet model of neonatal encephalopathy: a translational study. Neurobiol Dis. 2019; 121: 240–51.

143. Robertson N.J., et al. High-dose melatonin and ethanol excipient combined with therapeutic hypothermia in a newborn piglet asphyxia model. Sci Rep. 2020; 10: 3898.

144. Jerez-Calero A., et al. Hypothermia plus melatonin in asphyctic newborns: a randomized-controlled pilot study. Pediatr Crit Care Med. 2020; 21: 647–55.

145. Aly H., et al. Melatonin use for neuroprotection in perinatal asphyxia: a randomized controlled pilot study. J Perinatol. 2015; 35: 186–91.

146. Fulia F., 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.

147. Ahmad Q.M., Chishti A.L., Waseem N. Role of melatonin in management of hypoxic ischaemic encephalopathy in newborns: a randomized control trial. J Pak Med Assoc. 2018; 68: 1233–7.

148. Ahmed J., Pullattayil S.A., Robertson N.J., More K. Melatonin for neuroprotection in neonatal encephalopathy: a systematic review & meta-analysis of clinical trials. Eur J Paediatr Neurol. 2021; 31: 38–45.

149. Moreira G.A., Moraes Neto R., Ribeiro R.G., Crippa A.C.S. Cannabidiol for the treatment of refractory epilepsy in children: a critical review of the literature. Rev Paul Pediatr. 2022; 41: e2021197.

150. Lafuente H., et al. Effects of cannabidiol and hypothermia on short-term brain damage in new-born piglets after acute hypoxia-ischemia. Front Neurosci. 2016; 10: 323.

151. Barata L., et al. Neuroprotection by cannabidiol and hypothermia in a piglet model of newborn hypoxic-ischemic brain damage. Neuropharmacology. 2019; 146: 1–11.

152. Garberg H.T., et al. Short-term effects of cannabidiol after global hypoxiaischemia in newborn piglets. Pediatr Res. 2016; 80: 710–8.

153. Garberg H.T., et al. High-dose cannabidiol induced hypotension after global hypoxia-ischemia in piglets. Neonatology. 2017; 112: 143–9.

154. Maiwald C.A., et al. Effect of allopurinol in addition to hypothermia treatment in neonates for hypoxic-ischemic brain injury on neurocognitive outcome (Albino): study protocol of a blinded randomized placebo-controlled parallel group multicenter trial for superiority (phase III). BMC Pediatr. 2019; 19: 210.

All articles in our journal are distributed under the Creative Commons Attribution 4.0 International License (CC BY 4.0 license)

CHIEF EDITOR
CHIEF EDITOR
Degtyarev Dmitriy Nikolaevich
Doctor of Medical Sciences, Professor, Deputy Director for Scientific Research of the V.I. Kulakov Obstetrics, Gynecology and Perinatology National Medical Research Center of Ministry of Healthсаre of the Russian Federation, Head of the Chair of Neonatology at the Clinical Institute of Children's Health named after N.F. Filatov, I.M. Sechenov First Moscow State Medical University, Chairman of the Ethics Committee of the Russian Society of Neonatologists, Moscow, Russian Federation

ORCID iD 0000-0001-8975-2425

Journals of «GEOTAR-Media»