Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-qlrfm Total loading time: 0 Render date: 2024-07-08T20:42:57.413Z Has data issue: false hasContentIssue false

Section 4 - Specific Conditions Associated with Fetal and Neonatal Brain Injury

Published online by Cambridge University Press:  13 December 2017

David K. Stevenson
Affiliation:
Stanford University, California
William E. Benitz
Affiliation:
Stanford University, California
Philip Sunshine
Affiliation:
Stanford University, California
Susan R. Hintz
Affiliation:
Stanford University, California
Maurice L. Druzin
Affiliation:
Stanford University, California
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2017

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Elwood, JM, Little, J, Elwood, JH. Epidemiology and Control of Neural Tube Defects. Oxford University Press, 1992.CrossRefGoogle Scholar
Warkany, J, Lemire, RJ, Cohen, MM. Mental Retardation and Congenital Malformations of the Central Nervous System. Chicago: Year Book, 1981.Google Scholar
Golden, JA. Towards a greater understanding of the pathogenesis of holoprosencephaly. Brain Dev 1999; 21: 513–21.CrossRefGoogle ScholarPubMed
Matsunaga, E, Shiota, K. Holoprosencephaly in human embryos: epidemiologic studies of 150 cases. Teratology 1977; 16: 261–72.CrossRefGoogle ScholarPubMed
Cohen, MM. Perspectives on holoprosencephaly: Part III. Spectra, distinctions, continuities, and discontinuities. Am J Med Genet 1989; 34: 271–88.Google Scholar
Croen, LA, Shaw, GM, Lammer, EJ. Holoprosencephaly: epidemiologic and clinical characteristics of a California population. Am J Med Genet 1996; 64: 465–72.3.0.CO;2-O>CrossRefGoogle ScholarPubMed
Rasmussen, SA, Moore, CA, Khoury, MJ, et al. Descriptive epidemiology of holoprosencephaly and arhinencephaly in metropolitan Atlanta, 1968–1992. Am J Med Genet 1996; 66: 320–33.3.0.CO;2-O>CrossRefGoogle ScholarPubMed
Bullen, PJ, Rankin, JM, Robson, SC. Investigation of the epidemiology and prenatal diagnosis of holoprosencephaly in the North of England. Am J Obstet Gynecol 2001; 184: 1256–62.Google Scholar
Barkovich, AJ, Quint, DJ. Middle interhemispheric fusion: an unusual variant of holoprosencephaly. AJNR Am J Neuroradiol 1993; 14: 431–40.Google ScholarPubMed
Simon, EM, Hevner, RF, Pinter, JD, et al. The middle interhemispheric variant of holoprosencephaly. AJNR Am J Neuroradiol 2002; 23: 151–5.Google ScholarPubMed
Plawner, LL, Delgado, MR, Miller, VS, et al. Neuroanatomy of holoprosencephaly as predictor of function: beyond the face predicting the brain. Neurology 2002; 59: 1058–66.CrossRefGoogle ScholarPubMed
Barr, M, Cohen, MM. Holoprosencephaly survival and performance. Am J Med Genet 1999; 89: 116–20.3.0.CO;2-4>CrossRefGoogle ScholarPubMed
Lewis, AJ, Simon, EM, Barkovich, AJ, et al. Middle interhemispheric variant of holoprosencephaly: a distinct cliniconeuroradiologic subtype. Neurology 2002; 59: 1860–5.Google Scholar
Taylor, AI. Autosomal trisomy syndromes: a detailed study of 27 cases of Edwards’ syndrome and 27 cases of Patau’s syndrome. J Med Genet 1968; 5: 227–52.Google Scholar
Olsen, CL, Hughes, JP, Youngblood, LG, et al. Epidemiology of holoprosencephaly and phenotypic characteristics of affected children: New York State, 1984–1989. Am J Med Genet 1997; 73: 217–26.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
Ming, JE, Muenke, M. Multiple hits during early embryonic development: digenic diseases and holoprosencephaly. Am J Hum Genet 2002; 71: 1017–32.Google Scholar
Roessler, E, Belloni, E, Gaudenz, K, et al. Mutations in the human Sonic Hedgehog gene cause holoprosencephaly. Nat Genet 1996; 14: 357–60.CrossRefGoogle ScholarPubMed
Ming, JE, Kaupas, ME, Roessler, E, et al. Mutations in PATCHED-1, the receptor for SONIC HEDGEHOG, are associated with holoprosencephaly. Hum Genet 2002; 110: 297301.CrossRefGoogle ScholarPubMed
Barr, M Jr., Hanson, JW, Currey, K, et al. Holoprosencephaly in infants of diabetic mothers. J Pediatr 1983; 102: 565–8.Google Scholar
Malinger, G, Lev, D, Kidron, D, et al. Differential diagnosis in fetuses with absent septum pellucidum. Ultrasound Obstet Gynecol 2005; 25: 42–9.CrossRefGoogle ScholarPubMed
Carmichael, J, Woods, C. Genetic defects of human brain development. Curr Neurol Neurosci Rep 2006; 6: 437–46.Google Scholar
Mirzaa, GM, Poduri, A. Megalencephaly and hemimegalencephaly: breakthroughs in molecular etiology. Am J Med Genet C Semin Med Genet 2014; 166C: 156–72.Google ScholarPubMed
Guerrini, R, Dobyns, WB. Malformations of cortical development: clinical features and genetic causes. Lancet Neurol 2014; 13: 710–26.CrossRefGoogle ScholarPubMed
Barkovich, AJ, Guerrini, R, Kuzniecky, RI, et al. A developmental and genetic classification for malformations of cortical development: update 2012. Brain 2012; 135: 1348–69.Google Scholar
Kerjan, G, Gleeson, JG. Genetic mechanisms underlying abnormal neuronal migration in classical lissencephaly. Trends Genet 2007; 23: 623–30.CrossRefGoogle ScholarPubMed
Prayson, RA. Classification and pathologic characteristics of the cortical dysplasias. Childs Nerv Sys 2014; 30: 1805–12.Google Scholar
Lim, KC, Crido, PB. Focal malformations of cortical development: new vistas for molecular pathogenesis. Neurosciences 2013; 252: 262–76.Google ScholarPubMed
Glenn, OA, Goldstein, RB, Li, KC, et al. Fetal magnetic resonance imaging in the evaluation of fetuses referred for sonographically suspected abnormalities of the corpus callosum. J Ultrasound Med 2005; 24: 791804.Google Scholar
Edwards, TJ, Sherr, EH, Barkovich, AJ, Richards, LJ. Clinical, genetic and imaging findings identify new causes for corpus callosum development syndromes. Brain 2014; 217: 1579–613.Google Scholar
Aicardi, J. Aicardi syndrome. Brain Dev 2005; 27: 164–71.Google Scholar
Girard, N, Chaumoitre, K, Confort-Gouny, S, et al. Magnetic resonance imaging and the detection of fetal brain anomalies, injury, and physiologic adaptations. Curr Opin Obstet Gynecol 2006; 18: 164–76.CrossRefGoogle ScholarPubMed
Haverkamp, F, Zerres, K, Ostertun, B, et al. Familial schizencephaly: further delineation of a rare disorder. J Med Genet 1995; 32: 242–4.CrossRefGoogle ScholarPubMed
Brunelli, S, Faiella, A, Capra, V, et al. Germline mutations in the homeobox gene EMX2 in patients with severe schizencephaly. Nat Genet 1996; 12: 94–6.CrossRefGoogle ScholarPubMed
Robinson, AJ. Inferior vermian hypoplasia: preconception, misconceptions. Ultrasound Obstet Gynecol 2014; 43: 123–36.CrossRefGoogle Scholar
Grinberg, I, Northrup, H, Ardinger, H, et al. Heterozygous deletion of the linked genes ZIC1 and ZIC4 is involved in Dandy-Walker malformation. Nat Genet 2004; 36: 1053–5.CrossRefGoogle ScholarPubMed
Ecker, JL, Shipp, TD, Bromley, B, et al. The sonographic diagnosis of Dandy-Walker and Dandy-Walker variant: associated findings and outcomes. Prenat Diagn 2000; 20: 328–32.3.0.CO;2-O>CrossRefGoogle ScholarPubMed
Romani, M, Micalizzi, A, Valente, EM. Joubert syndrome: congenital cerebellar ataxia with “molar tooth.” Lancet Neurol 2013; 12: 894905.CrossRefGoogle ScholarPubMed
Rudnik-Schöneborg, S, Barth, PG, Zerres, K. Pontocerebellar hypoplasia. Am J Med Genet C Semin Med Genet 2014; 166C: 173–83.Google Scholar
Kanemura, Y, Okamoto, N, Sakamoto, H, et al. Molecular mechanisms and neuroimaging criteria for severe L1 syndrome with X-linked hydrocephalus. J Neurosurg 2006; 105: 403–12.Google Scholar
Alvarez, H, Garcia Monaco, R, Rodesch, G, et al. Vein of Galen aneurysmal malformations. Neuroimag Clin North Am 2007; 17:189206.CrossRefGoogle ScholarPubMed
Glenn, OA, Barkovich, AJ. Magnetic resonance imaging of the fetal brain and spine: an increasingly important tool in prenatal diagnosis, part 1. AJNR Am J Neuroradiol 2006; 27: 1604–11.Google ScholarPubMed
Glenn, OA, Barkovich, J. Magnetic resonance imaging of the fetal brain and spine: an increasingly important tool in prenatal diagnosis: part 2. AJNR Am J Neuroradiol 2006; 27: 1807–14.Google ScholarPubMed

References

Jones, K.L., Jones, MC, Campo, MD. Smith’s Recognizable Patterns of Human Malformation, 7th edn. Philadelphia: Elsevier Saunders, 2013: xiii.Google Scholar
Yang, Y, et al. Molecular findings among patients referred for clinical whole-exome sequencing. JAMA 2014; 312(18): 1870–9.Google Scholar
Lee, H, et al. Clinical exome sequencing for genetic identification of rare Mendelian disorders. JAMA 2014; 312(18): 1880–7.CrossRefGoogle ScholarPubMed
Iglesias, A, et al. The usefulness of whole-exome sequencing in routine clinical practice. Genet Med 2014; 16(12): 922–31.CrossRefGoogle ScholarPubMed
Yang, Y, et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med 2013; 369(16): 1502–11.Google Scholar
Reuter, JA, Spacek, DV, Snyder, MP. High-throughput sequencing technologies. Mol Cell 2015; 58(4): 586–97.Google Scholar
Driscoll, DJ, et al. Prader-Willi syndrome. GeneReviews. Seattle: University of Washington, 1998. Available from www.ncbi.nlm.nih.gov/books/NBK1330/.Google Scholar
Smith, A, et al. Birth prevalence of Prader-Willi syndrome in Australia. Arch Dis Child 2003; 88(3): 263–4.CrossRefGoogle ScholarPubMed
Vogels, A, et al. Minimum prevalence, birth incidence and cause of death for Prader-Willi syndrome in Flanders. Eur J Hum Genet 2004; 12(3): 238–40.Google Scholar
Kubota, T, et al. Methylation-specific PCR simplifies imprinting analysis. Nature Genet 1997; 16(1): 1617.CrossRefGoogle ScholarPubMed
Cassidy, SB, McCandless, SE. Prader-Willi syndrome. In Management of Genetic Syndromes. Hoboken, NJ: Wiley-Liss, 2010:xvii.Google Scholar
Arnold, WD, Kassar, D, Kissel, JT. Spinal muscular atrophy: diagnosis and management in a new therapeutic era. Muscle Nerve 2015; 51(2): 5767.Google Scholar
Prior, TW, Russman, BS. Spinal muscular atrophy. GeneReviews. Seattle: University of Washington, 2000. Available from www.ncbi.nlm.nih.gov/books/NBK1352/.Google Scholar
Scriver, CR. The Metabolic and Molecular Bases of Inherited Disease, 8th edn. New York: McGraw-Hill, 2001: xlvii, I140.Google Scholar
Bird, TD. Myotonic dystrophy type 1. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Harper, PS. Myotonic dystrophy. In Major Problems in Neurology, vol. 21, 2nd edn. London: Saunders, 1989: xi.Google Scholar
Bachmann, G, et al. The clinical and genetic correlates of MRI findings in myotonic dystrophy. Neuroradiology 1996; 38(7): 629–35.Google Scholar
Wan, M, et al. Rett syndrome and beyond: recurrent spontaneous and familial MECP2 mutations at CpG hotspots. Am J Hum Genet 1999; 65(6): 1520–9.CrossRefGoogle ScholarPubMed
Christodoulou, J, Ho, G. MECP2-related disorders. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Leslie, N, Tinkle, BT. Glycogen storage disease type II (Pompe disease). In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Martiniuk, F, et al. Carrier frequency for glycogen storage disease type II in New York and estimates of affected individuals born with the disease. Am J Med Genet 1998; 79(1): 6972.Google Scholar
Kishnani, PS, et al. A retrospective, multinational, multicenter study on the natural history of infantile-onset Pompe disease. J Pediatr 2006; 148(5): 671–6.CrossRefGoogle ScholarPubMed
Kishnani, PS, et al. Pompe disease diagnosis and management guideline. Genet Med 2006; 8(5): 267–88.CrossRefGoogle ScholarPubMed
Goebel, HH. Congenital myopathies in the new millennium. J Child Neurol 2005; 20(2): 94101.CrossRefGoogle ScholarPubMed
Maggi, L, et al. Congenital myopathies–clinical features and frequency of individual subtypes diagnosed over a 5-year period in the United Kingdom. Neuromus Disord: NMD 2013; 23(3): 195205.Google Scholar
North, KN, et al. Approach to the diagnosis of congenital myopathies. Neuromusc Disord NMD 2014; 24(2): 97116.Google Scholar
Sparks, S, et al. Congenital muscular dystrophy overview. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Mendell, JR, Boue, DR, Martin, PT. The congenital muscular dystrophies: recent advances and molecular insights. Pediatr Dev Pathol 2006; 9(6): 427–43.Google Scholar
Mostacciuolo, ML, et al. Genetic epidemiology of congenital muscular dystrophy in a sample from north-east Italy. Hum Genet 1996; 97(3): 277–9.Google Scholar
Peat, RA, et al. Diagnosis and etiology of congenital muscular dystrophy. Neurology 2008; 71(5): 312–21.CrossRefGoogle ScholarPubMed
Plante-Bordeneuve, V, Said, G. Dejerine-Sottas disease and hereditary demyelinating polyneuropathy of infancy. Muscle Nerve 2002; 26(5): 608–21.CrossRefGoogle ScholarPubMed
Baets, J, et al. Genetic spectrum of hereditary neuropathies with onset in the first year of life. Brain: A Journal of Neurology 2011; 134(Pt 9): 2664–76.Google Scholar
Abicht, A, Muller, JS, Lochmuller, H. Congenital myasthenic syndromes. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Engel, AG, et al. Congenital myasthenic syndromes: pathogenesis, diagnosis, and treatment. Lancet. Neurol 2015; 14(5): 461.Google Scholar
Crow, YJ. Aicardi-Goutieres syndrome. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Rice, G, et al. Clinical and molecular phenotype of Aicardi-Goutieres syndrome. Am J Hum Genet 2007; 81(4): 713–25.Google Scholar
Cohn, RD, et al. Intracranial hemorrhage as the initial manifestation of a congenital disorder of glycosylation. Pediatrics 2006; 118(2): e514–21.Google Scholar
McDonald, J, Pyeritz, RE. Hereditary hemorrhagic telangiectasia. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Marchuk, DA, et al. Report on the workshop on hereditary hemorrhagic telangiectasia, July 10–11, 1997. Am J Med Genet 1998; 76(3): 269–73.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Morgan, T, et al. Intracranial hemorrhage in infants and children with hereditary hemorrhagic telangiectasia (Osler-Weber-Rendu syndrome). Pediatrics 2002; 109(1): e12.Google Scholar
Shovlin, CL, et al. Diagnostic criteria for hereditary hemorrhagic telangiectasia (Rendu-Osler-Weber syndrome). Am J Med Genet 2000; 91(1): 66–7.3.0.CO;2-P>CrossRefGoogle ScholarPubMed
McDonald, J, et al. Molecular diagnosis in hereditary hemorrhagic telangiectasia: findings in a series tested simultaneously by sequencing and deletion/duplication analysis. Clin Genet 2011; 79(4): 335–44.CrossRefGoogle Scholar
Porteous, ME, Berg, JN. Hereditary hemorrhagic telangiectasia. In Management of Genetic Syndromes. Hoboken, NJ: Wiley-Liss, 2005: xvii.Google Scholar
Baethmann, M, et al. Hydrocephalus internus in two patients with 5,10-methylenetetrahydrofolate reductase deficiency. Neuropediatrics 2000; 31(6): 314–7.CrossRefGoogle ScholarPubMed
Longo, D, et al. MRI and 1H-MRS findings in early-onset cobalamin C/D defect. Neuropediatrics 2005; 36(6): 366–72.Google Scholar
Stumpel, C, Vos, YJ., L1 syndrome. GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Halliday, J, et al. X linked hydrocephalus: a survey of a 20 year period in Victoria, Australia. J Med Genet 1986; 23(1): 2331.Google Scholar
Chow, CW, et al. Congenital absence of pyramids and its significance in genetic diseases. Acta Neuropathol 1985; 65(3–4): 313–7.Google Scholar
Weese-Mayer, DE, et al. Congenital central hypoventilation syndrome. In GeneReviews, Pagon, RA, et al., eds. Seattle: University of Washington, 1993.Google Scholar
Todd, ES, et al. Facial phenotype in children and young adults with PHOX2B-determined congenital central hypoventilation syndrome: quantitative pattern of dysmorphology. Pediatr Res 2006; 59(1): 3945.Google Scholar
Berry-Kravis, EM, et al. Congenital central hypoventilation syndrome: PHOX2B mutations and phenotype. Am J Respir Crit Care Med 2006; 174(10): 1139–44.Google Scholar
Trochet, D, et al. PHOX2B genotype allows for prediction of tumor risk in congenital central hypoventilation syndrome. Am J Hum Genet 2005; 76(3): 421–6.Google Scholar

References

Del Bigio, MR. Cell proliferation in human ganglionic eminence and suppression after prematurity-associated haemorrhage. Brain 2011; 134(Pt 5): 1344–61.CrossRefGoogle ScholarPubMed
Paneth, N, Rudelli, R, Kazam, E, et al. Brain Damage in the Preterm Infant (Clinics in Developmental Medicine 131). London: MacKeith Press, 1994.Google Scholar
Parodi, A, Rossi, A, Severino, M et al. Accuracy of ultrasound in assessing cerebellar haemorrhages in very low birthweight babies. Arch Dis Child Fetal Neonatal Ed 2015; 100(4): F289–92.Google Scholar
Tortora, D, Severino, M, Malova, M et al. Differences in subependymal vein anatomy may predispose preterm infants to GMH-IVH Arch Dis Child Fetal Neonatal Ed. 2017 Jun 6. pii: fetalneonatal-2017-312710. doi: 10.1136/archdischild-2017-312710. [Epub ahead of print]CrossRefGoogle Scholar
Takashima, S, Takashi, M, Ando, Y. Pathogenesis of periventricular white matter haemorrhage in preterm infants. Brain Dev 1986; 8: 2530.CrossRefGoogle ScholarPubMed
Gould, SJ, Howard, S, Hope, PL, et al. Periventricular intraparenchymal cerebral haemorrhage in preterm infants: the role of venous infarction. J Pathol 1987; 151: 197202.Google Scholar
Ghazi-Birry, HS, Brown, WR, Moody, DM, et al. Human germinal matrix: venous origin of hemorrhage and vascular characteristics. AJNR Am J Neuroradiol 1997; 18: 219–29.Google Scholar
Pape, KE, Wigglesworth, JS. Haemorrhage, Ischaemia and Perinatal Brain (Clinics in Developmental Medicine 69/70). London: SIMP/Heinemann, 1979: 133–48.Google Scholar
Ment, LR, Stewart, WB, Ardito, TA, et al. Germinal matrix microvascular maturation correlates inversely with the risk period for neonatal intraventricular hemorrhage. Brain Res Dev Brain Res 1995; 84: 142–9.Google Scholar
Pinto Cardoso, G, Abily-Donval, L, Chadie, A, et al. Le réseau de périnatalité de Haute-Normandie [Epidemiological study of very preterm infants at Rouen University Hospital: changes in mortality, morbidity, and care over 11 years]. Arch Pediatr 2013; 20(2): 156–63Google Scholar
Horbar, JD, Carpenter, JH, Badger, GJ, et al. Mortality and neonatal morbidity among infants 501 to 1500 grams from 2000 to 2009. Pediatrics 2012; 129: 1019–26Google Scholar
Larroque, B, Marret, S, Ancel, PY, et al. White matter damage and intraventricular hemorrhage in very preterm infants: the EPIPAGE study. J Pediatr 2003; 143: 477–83.CrossRefGoogle ScholarPubMed
Hamrick, SE, Miller, SP, Leonard, C, et al. Trends in severe brain injury and neurodevelopmental outcome in premature newborn infants: the role of cystic periventricular leukomalacia. J Pediatr 2004; 145: 593–9.Google Scholar
Thorp, JA, Jones, PG, Clark, RH, et al. Perinatal factors associated with severe intracranial hemorrhage. Am J Obstet Gynecol 2001; 185: 859–62.Google Scholar
Yanowitz, TD, Jordan, JA, Gilmour, CH, et al. Hemodynamic disturbances in premature infants born after chorioamnionitis: association with cord blood cytokine concentrations. Pediatr Res 2002; 51: 310–6.Google Scholar
Haque, KN, Hayes, AM, Ahmed, Z, et al. Caesarean or vaginal delivery for preterm very-low-birth weight (≤1,250 g) infant: experience from a district general hospital in UK. Arch Gynecol Obstet 2008; 277: 207–12.Google Scholar
Riskin, A, Riskin-Mashiah, S, Bader, D et al. Delivery mode and severe intraventricular hemorrhage in single, very low birth weight, vertex infants. Obstet Gynecol 2008; 112: 21–8.Google Scholar
Herbst, A, Källén, K. Influence of mode of delivery on neonatal mortality and morbidity in spontaneous preterm breech delivery. Eur J Obstet Gynecol Reprod Biol 2007; 133: 25–9.Google Scholar
Jensen, EA, Lorch, SA. Association between Off-Peak Hour Birth and Neonatal Morbidity and Mortality among Very Low Birth Weight Infants. J Pediatr. 2017 Jul; 186: 4148.e4Google Scholar
Mercer, JS, Vohr, BR, McGrath, MM, et al. Delayed cord clamping in very preterm infants reduces the incidence of intraventricular hemorrhage and late-onset sepsis: a randomized, controlled trial. Pediatrics 2006; 117: 1235–42.Google Scholar
Rabe, H, Diaz-Rossello, JL, Duley, L, Dowswell, T. Effect of timing of umbilical cord clamping and other strategies to influence placental transfusion at preterm birth on maternal and infant outcomes. Cochrane Database Syst Rev 2012; 15(8): CD003248.Google Scholar
Heuchan, AM, Evans, N, Henderson Smart, DJ. Perinatal risk factors for major intraventricular haemorrhage in the Australian and New Zealand Neonatal Network, 1995–97. Arch Dis Child Fetal Neonatal Ed 2002; 86: F8690.Google Scholar
Palmer, KG, Kronsberg, SS, Barton, BA, et al. Effect of inborn versus outborn delivery on clinical outcomes in ventilated preterm neonates: secondary results from the NEOPAIN trial. J Perinatol 2005; 25: 270–5.Google Scholar
Mohamed, MA, Aly, H. Transport of premature infants is associated with increased risk for intraventricular haemorrhage. Arch Dis Child Fetal Neonatal Ed 2010; 95: F403–7.CrossRefGoogle ScholarPubMed
Limperopoulos, C, Gauvreau, KK, O’Leary, H, et al. Cerebral hemodynamic changes during intensive care of preterm infants. Pediatrics 2008; 122(5): e1006–13.CrossRefGoogle ScholarPubMed
Vela-Huerta, MM, Amador-Licona, M, Medina-Ovando, N, Aldana-Valenzuela, C. Factors associated with early severe intraventricular haemorrhage in very low birth weight infants. Neuropediatrics 2009; 40(5): 224–7.Google Scholar
Osborn, DA, Evans, N, Kluckow, M. Hemodynamic and antecedent risk factors of early and late periventricular/intraventricular hemorrhage in premature infants. Pediatrics 2003; 112: 33–9.CrossRefGoogle ScholarPubMed
Tsuji, M, Saul, P, du Plessis, A, et al. Cerebral intravascular oxygenation correlates with mean arterial pressure in critically ill premature infants. Pediatrics 2000; 106: 625–32.CrossRefGoogle ScholarPubMed
Soul, JS, Hammer, PE, Tsuji, M, et al. Fluctuating pressure-passivity is common in the cerebral circulation of sick premature infants. Pediatr Res 2007; 61: 467–73.CrossRefGoogle ScholarPubMed
Alderliesten, T, Lemmers, PM, Smarius, JJ, et al. Cerebral oxygenation, extraction, and autoregulation in very preterm infants who develop peri-intraventricular hemorrhage. J Pediatr 2013; 162(4): 698704.Google Scholar
Noori, S, McCoy, M, Anderson, MP, et al. Changes in cardiac function and cerebral blood flow in relation to peri/intraventricular hemorrhage in extremely preterm infants. J Pediatr 2014; 164(2): 264–70.Google Scholar
Ikeda, T, Amizuka, T, Ito, Y, et al. Changes in the perfusion waveform of the internal cerebral vein and intraventricular hemorrhage in the acute management of extremely low-birth-weight infants. Eur J Pediatr 2015; 174(3): 331–8Google Scholar
Fabres, J, Carlo, WA, Phillips, V, et al. Both extremes of arterial carbon dioxide pressure and the magnitude of fluctuations in arterial carbon dioxide pressure are associated with severe intraventricular hemorrhage in preterm infants. Pediatrics 2007; 119: 299305.CrossRefGoogle ScholarPubMed
Dalton, J, Dechert, RE, Sarkar, S. Assessment of association between rapid fluctuations in serum sodium and intraventricular hemorrhage in hypernatremic preterm infants. Am J Perinatol 2015; 32(8): 795802.Google Scholar
Barnette, AR, Myers, BJ, Berg, CS, Inder, TE. Sodium intake and intraventricular hemorrhage in the preterm infant. Ann Neurol 2010; 67(6): 817–23Google Scholar
Ryckman, KK, Dagle, JM, Kelsey, K, et al. Replication of genetic associations in the inflammation, complement, and coagulation pathways with intraventricular hemorrhage in LBW preterm neonates. Pediatr Res 2011; 70: 90–5.CrossRefGoogle ScholarPubMed
Ment, LR, Adén, U, Lin, A et al. Gene Targets for IVH Study Group. Gene-environment interactions in severe intraventricular hemorrhage of preterm neonates. Pediatr Res 2014; 75(1–2): 241–50.CrossRefGoogle Scholar
Harteman, JC, Groenendaal, F, van Haastert, IC, et al. Atypical timing and presentation of periventricular haemorrhagic infarction in preterm infants: the role of thrombophilia. Dev Med Child Neurol 2012; 54(2): 140–7Google Scholar
Harding, DR, Dhamrait, S, Whitelaw, A, et al. Does interleukin-6 genotype influence cerebral injury or developmental progress after preterm birth? Pediatrics 2004; 114(4): 941–7Google Scholar
Göpel, W, Härtel, C, Ahrens, P, et al. Interleukin-6–174-genotype, sepsis and cerebral injury in very low birth weight infants. Genes Immun 2006; 7: 65–8.Google Scholar
Kallankari, H, Kaukola, T, Ojaniemi, M, et al. Chemokine CCL18 predicts intraventricular hemorrhage in very preterm infants. Ann Med 2010; 42: 416–25.CrossRefGoogle ScholarPubMed
de Vries, LS, Koopman, C, Groenendaal, F, et al. COL4A1 mutation in two preterm siblings with antenatal onset of parenchymal haemorrhage. Ann Neurol 2009; 65: 12–8.Google Scholar
Meuwissen, ME, Halley, DJ, Smit, LS, et al. The expanding phenotype of COL4A1 and COL4A2 mutations: clinical data on 13 newly identified families and a review of the literature. Genet Med 2015; 7(11): 843–53.Google Scholar
Shankaran, S, Bauer, CR, Bain, R, et al. Prenatal and perinatal risk and protective factors for neonatal intracranial hemorrhage. Arch Pediatr Adolesc Med 1996; 150: 491–7.Google Scholar
Gagliardi, L, Rusconi, F, Da Frè, M, et al. Pregnancy disorders leading to very preterm birth influence neonatal outcomes: results of the population-based ACTION cohort study. Pediatr Res 2013; 73: 794801.Google Scholar
Andre, P, Thebaud, B, Delavaucoupet, J, et al. Late-onset cystic periventricular leukomalacia in premature infants: a threat until term. Am J Perinatol 2001; 18: 7986.CrossRefGoogle ScholarPubMed
de Vries, LS, Rademaker, KJ, Roelantsvan-Rijn, AM, et al. Unilateral haemorrhagic parenchymal infarction in the preterm infant. Eur J Pediatr Neurol 2001; 5: 139–49.Google Scholar
Volpe, JJ. Neonatal Neurology, 4th edn. Philadelphia: Saunders, 2001.Google Scholar
Correa, F, Enríquez, G, Rosselló, J, et al. Posterior fontanelle sonography: an acoustic window into the neonatal brain. AJNR Am J Neuroradiol 2004; 25: 1274–82.Google Scholar
Maalouf, EF, Duggan, PJ, Counsell, SJ, et al. Comparison of findings on cranial ultrasound and magnetic resonance imaging in preterm infants. Pediatrics 2001; 107: 719–27.Google Scholar
Parodi, A, Morana, G, Severino, MS, et al. Low-grade intraventricular hemorrhage: is ultrasound good enough? J Matern Fetal Neonatal Med 2015; Suppl 1:2261–4.Google Scholar
Dudink, J, Lequin, M, Weisglas-Kuperus, N, et al. Venous subtypes of preterm periventricular haemorrhagic infarction. Arch Dis Child Fetal Neonatal Ed 2007; 93: F201–6.Google Scholar
Bassan, H, Benson, CB, Limperopoulos, C, et al. Ultrasonographic features and severity scoring of periventricular hemorrhagic infarction in relation to risk factors and outcome. Pediatrics 2006; 117: 2111–18.Google Scholar
Bassan, H, Limperopoulos, C, Visconti, K, et al. Neurodevelopmental outcome in survivors of periventricular hemorrhagic infarction. Pediatrics 2007; 120: 785–92.Google Scholar
Limperopoulos, C, Benson, CB, Bassan, H, et al. Cerebellar hemorrhage in the preterm infant: ultrasonographic findings and risk factors. Pediatrics 2005; 116: 717–24.Google Scholar
Steggerda, SJ, Leijser, LM, Wiggers-de Bruïne, FT, et al. Cerebellar injury in preterm infants: incidence and findings on US and MR images. Radiology 2009; 252: 190–9.Google Scholar
Ecury-Goossen, GM, Dudink, J, Lequin, M, et al. The clinical presentation of preterm cerebellar haemorrhage. Eur J Pediatr 2010; 169(10): 1249–53.CrossRefGoogle ScholarPubMed
Tam, EW, Miller, SP, Studholme, C, et al. Differential effects of intraven- tricular hemorrhage and white matter injury on preterm cerebellar growth. J Pediatr 2011; 158(3): 366–71.CrossRefGoogle Scholar
Steggerda, SJ, De Bruïne, FT, van den Berg-Huysmans, AA, et al. Small cerebellar hemorrhage in preterm infants: perinatal and postnatal factors and outcome. Cerebellum 2013; 12(6): 794801Google Scholar
Limperopoulos, C, Bassan, H, Gauvreau, K, et al. Does cerebellar injury in premature infants contribute to the high prevalence of long-term cognitive, learning, and behavioral disability in survivors? Pediatrics 2007; 120: 584–93.Google Scholar
Srinivasan, L, Allsop, J, Counsell, SJ, et al. Smaller cerebellar volumes in very preterm infants at term-equivalent age are associated with the presence of supratentorial lesions. AJNR Am J Neuroradiol 2006; 27: 573–9.Google ScholarPubMed
Morita, T, Morimoto, M, Yamada, K, et al. Low-grade intraventricular hemorrhage disrupts cerebellar white matter in preterm infants: evidence from diffusion tensor imaging. Neuroradiology 2015; 57(5): 507–14Google Scholar
McLendon, D, Check, J, Carteaux, P, et al. Implementation of potentially better practices for the prevention of brain hemorrhage and ischemic brain injury in very low birth weight infants. Pediatrics 2003; 111: e497503.CrossRefGoogle ScholarPubMed
Olischar, M, Klebermass, K, Waldhoer, T, et al. Background patterns and sleep-wake cycles on amplitude-integrated electroencephalography in preterms younger than 30 weeks gestational age with peri-/intraventricular haemorrhage. Acta Paediatr 2007; 96: 1743–50.Google Scholar
Murphy, BP, Inder, TE, Rooks, V, et al. Posthaemorrhagic ventricular dilatation in the premature infant: natural history and predictors of outcome. Arch Dis Child Fetal Neonatal Ed 2002; 87: F3741.Google Scholar
Ingram, MC, Huguenard, AL, Miller, BA, Chern, JJ. Poor correlation between head circumference and cranial ultrasound findings in premature infants with intraventricular hemorrhage. J Neurosurg Pediatr 2014; 14: 184–9.CrossRefGoogle ScholarPubMed
Levene, MI, Starte, DR. A longitudinal study of posthaemorrhagic ventricular dilatation in the newborn. Arch Dis Child 1981; 56: 905–10.Google Scholar
Davies, MW, Swaminathan, M, Chuang, SI, et al. Reference ranges for the linear dimensions of the intracranial ventricles in preterm neonates. Arch Dis Child Fetal Neonatol Ed 2000; 82:F219–23.CrossRefGoogle ScholarPubMed
Brouwer, MJ, de Vries, LS, Groenendaal, F, et al. New reference values for the neonatal cerebral ventricles. Radiology 2012; 262(1): 224–33.Google Scholar
Kaiser, A, Whitelaw, A. Cerebrospinal fluid pressure during posthaemorrhagic ventricular dilatation in newborn. Arch Dis Child 1985; 60: 920–4.Google Scholar
Soul, JS, Eichenwald, E, Walter, G, et al. CSF removal in infantile posthemorrhagic hydrocephalus results in significant improvement in cerebral hemodynamics. Pediatr Res 2004; 55: 872–6.Google Scholar
Klebermass-Schrehof, K, Rona, Z, Waldhör, T, et al. Can neurophysiological assessment improve timing of intervention in posthaemorrhagic ventricular dilatation? Arch Dis Child Fetal Neonatal Ed 2013; 98(4): F291–7.Google Scholar
Sävman, K, Blennow, M, Hagberg, H, et al. Cytokine response in cerebrospinal fluid from preterm infants with posthaemorrhagic ventricular dilatation. Acta Paediatr 2002; 91: 1357–63.Google Scholar
Felderhoff-Mueser, U, Buhrer, C, Groneck, P, et al. Soluble Fas (CD95/Apo-1), soluble Fas ligand and activated caspase 3 in the cerebrospinal fluid of infants with posthemorrhagic and nonhemorrhagic hydrocephalus. Pediatr Res 2003; 54: 5964.CrossRefGoogle ScholarPubMed
Heep, A, Stoffel-Wagner, B, Bartmann, P, et al. Vascular endothelial growth factor and transforming growth factor-β1 are highly expressed in the cerebrospinal fluid of premature infants with posthemorrhagic hydrocephalus. Pediatr Res 2004; 56: 768–74.Google Scholar
Schmitz, T, Heep, A, Groenendaal, F, et al. Interleukin-1β, interleukin-18, and interferon-γ expression in the cerebrospinal fluid of premature infants with posthemorrhagic hydrocephalus–markers of white matter damage? Pediatr Res 2007; 61: 722–6.CrossRefGoogle ScholarPubMed
Whitelaw, A, Lee-Kelland, R. Repeated lumbar or ventricular punctures in newborns with intraventricular haemorrhage. Cochrane Database Syst Rev. 2017 Apr 6; 4: CD000216.Google Scholar
Ventriculomegaly Trial Group. Randomized trial of early tapping in neonatal posthaemorrhagic ventricular dilatation: results at 30 months. Arch Dis Child Fetal Neonatal Ed 1994; 70: F129–36.Google Scholar
Whitelaw, A, Evans, D, Carter, M, et al. Randomized clinical trial of prevention of hydrocephalus after intraventricular hemorrhage in preterm infants: brainwashing versus tapping fluid. Pediatrics 2007; 119: e1071–8.CrossRefGoogle ScholarPubMed
Brouwer, AJ, Groenendaal, F, van Haastert, IC, et al. Neurodevelopmental outcome of preterm infants with severe intraventricular hemorrhage and therapy for post-hemorrhagic ventricular dilatation. J Pediatr 2008; 152: 648–54.CrossRefGoogle ScholarPubMed
Bassan, H, Eshel, R, Golan, I, et al. External Ventricular Drainage Study Investigators. Timing of external ventricular drainage and neurodevelopmental outcome in preterm infants with posthemorrhagic hydrocephalus. Eur J Paediatr Neurol 2012; 16(6): 662–70.CrossRefGoogle Scholar
Vasileiadis, GT, Gelman, N, Han, VK, et al. Uncomplicated intraventricular hemorrhage is followed by reduced cortical volume at near-term age. Pediatrics 2004; 114: e367–72.CrossRefGoogle ScholarPubMed
Patra, K, Wilson-Costello, D, Taylor, HG, et al. Grades I–II intraventricular hemorrhage in extremely low birth weight infants: effects on neurodevelopment. J Pediatr 2006; 149: 169–73.CrossRefGoogle ScholarPubMed
Bolisetty, S, Dhawan, A, Abdel-Latif, M, et al. New South Wales and Australian Capital Territory Neonatal Intensive Care Units’ Data Collection. Intraventricular hemorrhage and neurodevelopmental outcomes in extreme preterm infants. Pediatrics 2014; 133(1): 5562.CrossRefGoogle ScholarPubMed
Payne, AH, Hintz, SR, Hibbs, AM, et al. Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Neurodevelopmental outcomes of extremely low-gestational-age neonates with low-grade periventricular-intraventricular hemorrhage. JAMA Pediatr 2013; 167(5): 451–9.Google Scholar
Radic, JAE, Vincer, M, McNeely, PD. Outcomes of intraventricular hemorrhage and posthemorrhagic hydrocephalus in a population-based cohort of very preterm infants born to residents of Nova Scotia from 1993 to 2010. J Neurosurg Pediatr 2015; 15: 580–8.CrossRefGoogle Scholar
Reubsaet, P, Brouwer, AJ, van Haastert, IC, et al The Impact of Low-Grade Germinal Matrix-Intraventricular Hemorrhage on Neurodevelopmental Outcome of Very Preterm Infants. Neonatology 2017 Jul 14; 112(3): 203210CrossRefGoogle Scholar
Kuban, K, Sanocka, U, Leviton, A, et al. White matter disorders of prematurity: association with intraventricular hemorrhage and ventriculomegaly. The Developmental Epidemiology Network. J Pediatr 1999; 134: 539–46.CrossRefGoogle ScholarPubMed
Brouwer, MJ, van Kooij, BJ, van Haastert, IC, et al. Sequential cranial ultrasound and cerebellar diffusion weighted imaging contribute to the early prognosis of neurodevelopmental outcome in preterm infants. PLoS One. 2014; 9(10): e109556Google Scholar
Fernell, E, Hagberg, G, Hagberg, B. Infantile hydrocephalus in preterm, low-birth-weight infants: a nationwide Swedish cohort study 1979–1988. Acta Paediatr 1993; 82: 45–8.Google Scholar
Persson, EK, Hagberg, G, Uvebrant, P. Disabilities in children with hydrocephalus: a population-based study of children aged between four and twelve years. Neuropediatrics 2006; 37: 330–6.Google Scholar
Sherlock, RL, Synnes, AR, Grunau, RE, et al. Long-term outcome after neonatal intraparenchymal echodensities with porencephaly. Arch Dis Child Fetal Neonatal Ed 2008; 93: F127–31.Google Scholar
De Vries, LS, Groenendaal, F, Eken, P, et al. Asymmetrical myelination of the posterior limb of the internal capsule: an early predictor of hemiplegia. Neuropediatrics 1999; 30: 314–19.Google Scholar
Roze, E, Benders, MJ, Kersbergen, KJ, et al. Neonatal DTI early after birth predicts motor outcome in preterm infants with periventricular hemorrhagic infarction. Pediatr Res 2015; 78(3): 298303.CrossRefGoogle ScholarPubMed
Counsell, SJ, Dyet, LE, Larkman, DJ, et al. Thalamo-cortical connectivity in children born preterm mapped using probabilistic magnetic resonance tractography. Neuroimage 2007; 34: 896904.Google Scholar
Roberts, D, Dalziel, S. Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev 2006; 3: CD004454.Google Scholar
Brownfoot, FC, Gagliardi, DI, Bain, E, et al. Different corticosteroids and regimens for accelerating fetal lung maturation for women at risk of preterm birth. Cochrane Database Syst Rev. 2013; 8: CD006764.Google Scholar
Baud, O, Foix-L’Helias, L, Kaminski, M, et al. Antenatal glucocorticoid treatment and cystic periventricular leukomalacia in very premature infants. N Engl J Med 1999; 341: 1190–6.Google Scholar
Modi, N, Lewis, H, Al-Naqeeb, N, et al. The effects of repeated antenatal glucocorticoid therapy on the brain. Pediatr Res 2001; 50: 581–5.Google Scholar
Wapner, RJ, Sorokin, Y, Mele, L, et al. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Long-term outcomes after repeat doses of antenatal corticosteroids. N Engl J Med 2007; 357(12): 1190–8.Google Scholar
Crowther, CA, Hiller, JE, Doyle, LW, et al. Effect of magnesium sulfate given for neuroprotection before preterm birth. JAMA 2003; 290: 2669–76.Google Scholar
Rouse, DJ, Hirtz, DG, Thom, E, et al. A randomized controlled trial of magnesium sulfate for the prevention of cerebral palsy. N Engl J Med 2008; 359: 895905.CrossRefGoogle ScholarPubMed
Doyle, LW, Crowther, CA, Middleton, P, Marret, S. Antenatal magnesium sulfate and neurologic outcome in preterm infants: a systematic review. Obstet Gynecol 2009; 113: 1327–33.Google Scholar
Stark, MJ, Hodyl, NA, Andersen, CC. Effects of antenatal magnesium sulphate treatment for neonatal neuro-protection on cerebral oxygen kinetics. Pediatr Res 2015; 78(3): 310–4.Google Scholar
Kamyar, M, Manuck, TA, Stoddard, GJ, et al. Magnesium sulfate, chorioamnionitis, and neurodevelopment after preterm birth. BJOG 2016; 123(7): 1161–6.Google Scholar
Crowther, CA, Crosby, DD, Henderson-Smart, DJ. Vitamin K prior to preterm birth for preventing neonatal periventricular haemorrhage. Cochrane Database Syst Rev 2010; 20(1): CD000229.Google Scholar
Smit, E, Odd, D, Whitelaw, A. Postnatal phenobarbital for the prevention of intraventricular haemorrhage in preterm infants. Cochrane Database Syst Rev 2013; 13(8): CD001691.Google Scholar
Fowlie, PW, Davis, PG, McGuire, W. Prophylactic intravenous indomethacin for preventing mortality and morbidity in preterm infants. Cochrane Database Syst Rev 2010; 7(7): CD000174.Google Scholar
Schmidt, B, Davis, P, Moddeman, D, et al. Trial of indomethacin prophylaxis in preterm investigators: long-term effects of indomethacin prophylaxis in extremely-low-birth-weight infants. N Eng J Med 2001; 344: 1966–72.Google Scholar
Ment, LR, Peterson, BS, Meltzer, JA, et al. A functional magnetic resonance imaging study of the long-term influences of early indomethacin exposure on language processing in the brains of prematurely born children. Pediatrics 2006; 118: 961–70.Google Scholar
Van Overmeire, B, Allegaert, K, Casaer, A, et al. Prophylactic ibuprofen in premature infants: a multicentre, randomised, double-blind, placebo-controlled trial. Lancet 2004; 364(9449): 1945–9.Google Scholar
Shah, SS, Ohlsson, A. Ibuprofen for the prevention of patent ductus arteriosus in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2006; 1: CD004213.Google Scholar
Synnes, AR, Macnab, YC, Qiu, Z, et al. Neonatal intensive care unit characteristics affect the incidence of severe intraventricular hemorrhage. Med Care 2006; 44: 754–9.CrossRefGoogle ScholarPubMed
Wu, YW, Hamrick, SEG, Miller, SP, et al. Intraventricular hemorrhage in term neonates caused by sinovenous thrombosis. Ann Neurol 2003; 54: 123–6.Google Scholar
Roland, EH, Flodmark, O, Hill, A. Thalamic hemorrhagic with intraventricular hemorrhage in the full term newborn. Pediatrics 1990; 85: 737–42.Google Scholar
Jocelyn, LJ, Casiro, OG. Neurodevelopmental outcome of term infants with intraventricular hemorrhage. Am J Dis Child 1992; 146: 194–7.Google Scholar
Whitby, EH, Griffiths, PD, Rutter, S, et al. Frequency and natural history of subdural haemorrhages in babies and relation to obstetric factors. Lancet 2004; 363: 846–51.Google Scholar
Looney, CB, Smith, JK, Merck, LH, et al. Intracranial hemorrhage in asymptomatic neonates: prevalence on MR images and relationship to obstetric and neonatal risk factors. Radiology 2007; 242: 535–41.Google Scholar
Hofmeyr, GJ, Hannah, ME. Planned caesarean section for term breech delivery. Cochrane Database Syst Rev 2003; 3: CD000166.Google Scholar
Chamnanvanakij, S, Rollins, N, Perlman, JM. Subdural hematoma in term infants. Pediatr Neurol 2002; 26: 301–14.Google Scholar
Govaert, P, Vanhaesebrouck, P, de Praeter, C. Traumatic neonatal intracranial bleeding and stroke. Arch Dis Child 1992; 67: 840–5.CrossRefGoogle ScholarPubMed
Brouwer, AJ, Groenendaal, F, Koopman, C, et al. Intracranial hemorrhage in full-term newborns: a hospital-based cohort study. Neuroradiology 2010; 52(6): 567–76CrossRefGoogle ScholarPubMed
Vinchon, M, Pierrat, V, Tchofo, PJ, et al. Traumatic intracranial hemorrhage in newborns. Childs Nerv Syst 2005; 21: 1042–8.Google Scholar
Kilani, RA, Wetmore, J. Neonatal subgaleal hematoma:presentation and outcome. Radiological findings and factors associated with mortality. Am J Perinatol 2006; 23: 41–8.Google Scholar
Chang, HY, Peng, CC, Kao, HA, et al. Neonatal subgaleal hemorrhage: clinical presentation, treatment, and predictors of poor prognosis. Pediatr Int 2007; 49: 903–7.Google Scholar
Dale, ST, Coleman, LT. Neonatal alloimmune thrombocytopenia: antenatal and postnatal imaging findings in the pediatric brain. AJNR Am J Neuroradiol 2002; 23: 1457–65.Google Scholar
Berkowitz, RL, Bussel, JB, McFarland, JG. Alloimmune thrombocytopenia: state of the art 2006. Am J Obstet Gynecol 2006; 195(4): 907–13.Google Scholar
Hardart, GE, Hardart, MKM, Arnold, JH. Intracranial hemorrhage in premature neonates treated with extracorporeal membrane oxygenation correlates with conceptional age. J Pediatr 2004; 145: 184–9.Google Scholar
Gannon, CM, Kornhauser, MS, Gross, GW, et al. When combined, early bedside head ultrasound and electroencephalography predict abnormal computerized tomography or magnetic resonance brain images obtained after extracorporeal membrane oxygenation treatment. J Perinatol 2001; 21: 451–5.Google Scholar
Bulas, DI, Glass, P, O’Donnell, RM, et al. Neonates treated with ECMO: predictive value of early CT and US neuroimaging findings on short-term neurodevelopmental outcome. Radiology 1995; 195: 407–12.Google Scholar
Bulas, D, Glass, P. Neonatal ECMO: neuroimaging and neurodevelopmental outcome. Semin Perinatol 2005; 29(1): 5865.Google Scholar
de Mol, AC, Gerrits, LC, van Heijst, AF, et al. Intravascular volume administration: a contributing risk factor for intracranial hemorrhage during extracorporeal membrane oxygenation? Pediatrics 2008; 121(6): e1599–603.Google Scholar

References

Mercuri, E, Barnett, A, Rutherford, M, et al. Neonatal cerebral infarction and neuromotor outcome at school age. Pediatrics 2004; 113: 95100.Google Scholar
Mercuri, E, Anker, S, Guzzetta, A, et al. Neonatal cerebral infarction and visual function at school age. Arch Dis Child Fetal Neonatal Ed 2003; 88:F487–91.Google Scholar
Mercuri, E, Rutherford, M, Cowan, F, et al. Early prognostic indicators of outcome in infants with neonatal cerebral infarction: a clinical, electroencephalogram, and magnetic resonance imaging study. Pediatrics 1999; 103:3946.Google Scholar
deVeber, GA, MacGregor, D, Curtis, R, et al. Neurologic outcome in survivors of childhood arterial ischemic stroke and sinovenous thrombosis. J Child Neurol 2000; 15: 316–24.CrossRefGoogle ScholarPubMed
McLinden, A, Baird, AD, Westmacott, R, et al. Early cognitive outcome after neonatal stroke. J Child Neurol 2007; 22: 1111–16.Google Scholar
Sran, SK, Baumann, RJ. Outcome of neonatal strokes. Am J Dis Child 1988; 142: 1086–8.Google ScholarPubMed
Sreenan, C, Bhargava, R, Robertson, CM. Cerebral infarction in the term newborn: clinical presentation and long-term outcome. J Pediatr 2000; 137: 351–5.CrossRefGoogle ScholarPubMed
Lee, J, Croen, LA, Lindan, C, et al. Predictors of outcome in perinatal arterial stroke: a population-based study. Ann Neurol 2005; 58: 303–8.CrossRefGoogle ScholarPubMed
Schulzke, S, Weber, P, Luetschg, J, Fahnenstich, H. Incidence and diagnosis of unilateral arterial cerebral infarction in newborn infants. J Perinat Med 2005; 33: 170–5.Google Scholar
Raju, TN, Nelson, KB, Ferriero, D, et al. Ischemic perinatal stroke: summary of a workshop sponsored by the National Institute of Child Health and Human Development and the National Institute of Neurological Disorders and Stroke. Pediatrics 2007; 120: 609–16.Google Scholar
Laugesaar, R, Kolk, A, Tomberg, T, et al. Acutely and retrospectively diagnosed perinatal stroke: a population-based study. Stroke 2007; 38: 2234–40.Google Scholar
Lee, J, Croen, LA, Backstrand, KH, et al. Maternal and infant characteristics associated with perinatal arterial stroke in the infant. JAMA 2005; 293: 723–9.CrossRefGoogle ScholarPubMed
Laugesaar, R, Kolk, A, Tomberg, T, et al. Acutely and retrospectively diagnosed perinatal stroke: a population-based study. Stroke 2007; 38: 2234–40.Google Scholar
Wu, YW, March, WM, Croen, LA, et al. Perinatal stroke in children with motor impairment: a population-based study. Pediatrics 2004; 114: 612–19.Google Scholar
Ozduman, K, Pober, BR, Barnes, P, et al. Fetal stroke. Pediatr Neurol 2004; 30: 151–62.Google Scholar
Levy, SR, Abroms, IF, Marshall, PC, et al. Seizures and cerebral infarction in the full-term newborn. Ann Neurol 1985; 17: 366–70.Google Scholar
Clancy, R, Malin, S, Laraque, D, et al. Focal motor seizures heralding stroke in full-term neonates. Am J Dis Child 1985; 139: 601–6.Google Scholar
Ramaswamy, V, Miller, SP, Barkovich, AJ, et al. Perinatal stroke in term infants with neonatal encephalopathy. Neurology 2004; 62: 2088–91.Google Scholar
Guzzetta, A, Mercuri, E, Rapisardi, G, et al. General movements detect early signs of hemiplegia in term infants with neonatal cerebral infarction. Neuropediatrics 2003; 34: 61–6.Google Scholar
Kirton, A, Armstrong-Wells, J, Chang, T, et al. Symptomatic neonatal arterial ischemic stroke: The international pediatric stroke study. Pediatrics 2011; 128:e1402–10.CrossRefGoogle ScholarPubMed
Ecury-Goossen, GM, Raets, MM, Lequin, M, et al. Risk factors, clinical presentation, and neuroimaging findings of neonatal perforator stroke. Stroke 2013; 44: 2115–20.Google Scholar
Fitzgerald, KC, Williams, LS, Garg, BP, et al. Cerebral sinovenous thrombosis in the neonate. Arch Neurol 2006; 63: 405–9.Google Scholar
Wu, YW, Miller, SP, Chin, K, et al. Multiple risk factors in neonatal sinovenous thrombosis. Neurology 2002; 59: 438–40.Google Scholar
Fitzgerald, KC, Golomb, MR. Neonatal arterial ischemic stroke and sinovenous thrombosis associated with meningitis. J Child Neurol 2007; 22: 818–22.Google Scholar
deVeber, G, Andrew, M, Adams, C, et al. Cerebral sinovenous thrombosis in children. N Engl J Med 2001; 345: 417–23.Google Scholar
Roland, EH, Flodmark, O, Hill, A. Thalamic hemorrhage with intraventricular hemorrhage in the full-term newborn. Pediatrics 1990; 85: 737–42.Google Scholar
Wu, YW, Hamrick, SE, Miller, SP, et al. Intraventricular hemorrhage in term neonates caused by sinovenous thrombosis. Ann Neurol 2003; 54: 123–6.Google Scholar
Benders, MJ, Groenendaal, F, Uiterwaal, CS, et al. Maternal and infant characteristics associated with perinatal arterial stroke in the preterm infant. Stroke 2007; 38: 1759–65.Google Scholar
de Vries, LS, Groenendaal, F, Eken, P, et al. Infarcts in the vascular distribution of the middle cerebral artery in preterm and fullterm infants. Neuropediatrics 1997; 28:8896.Google Scholar
Golomb, MR, MacGregor, DL, Domi, T, et al. Presumed pre- or perinatal arterial ischemic stroke: risk factors and outcomes. Ann Neurol 2001; 50: 163–8.Google Scholar
Kirton, A, Shroff, M, Pontigon, AM, et al. Risk factors and presentations of periventricular venous infarction vs. arterial presumed perinatal ischemic stroke. Arch Neurol 2010; 67: 842–8.Google Scholar
Takanashi, J, Tada, H, Barkovich, AJ, et al. Magnetic resonance imaging confirms periventricular venous infarction in a term-born child with congenital hemiplegia. Dev Med Child Neurol 2005; 47: 706–8.Google Scholar
Armstrong-Wells, J, Johnston, CS, Wu, YW, et al. Prevalence and predictors of perinatal hemorrhagic stroke: results from the Kaiser pediatric stroke study. Pediatrics 2009; 123: 823–8.Google Scholar
Gould, DB, Phalan, FC, Breedveld, GJ, et al. Mutations in Col4a1 cause perinatal cerebral hemorrhage and porencephaly. Science 2005; 308: 1167–71.Google Scholar
Breedveld, G, de Coo, IF, Lequin, MH, et al. Novel mutations in three families confirm a major role of COL4A1 in hereditary porencephaly. J Med Genet 2006; 43: 490–5.Google Scholar
de Vries, LS, Koopman, C, Groenendaal, F, et al. COL4A1 mutation in two preterm siblings with antenatal onset of parenchymal hemorrhage. Ann Neurol 2009; 65: 12–8.Google Scholar
Meuwissen, MEC Halley, DJJ, Smit, LS, et al. The expanding phenotype of COL4A1 and COL4A2 mutations: clinical data on 13 newly identified families and a review of the literature. Genet Med 2015; 17(11):843–53.Google Scholar
Arkel, YS, Ku, DH. Thrombophilia and pregnancy: review of the literature and some original data. Clin Appl Thromb Hemost 2001; 7: 259–68.Google Scholar
Silver, RK, MacGregor, SN, Pasternak, JF, et al. Fetal stroke associated with elevated maternal anticardiolipin antibodies. Obstet Gynecol 1992; 80: 497–9.Google Scholar
Akanli, LF, Trasi, SS, Thuraisamy, K, et al. Neonatal middle cerebral artery infarction: association with elevated maternal anticardiolipin antibodies. Am J Perinatol 1998; 15:399402.Google Scholar
Gunther, G, Junker, R, Strater, R, et al. Symptomatic ischemic stroke in full-term neonates: role of acquired and genetic prothrombotic risk factors. Stroke 2000; 31: 2437–41.Google Scholar
Ballem, P. Acquired thrombophilia in pregnancy. Semin Thromb Hemost 1998; 24: 41–7.Google Scholar
Rigo, J, Nagy, B, Fintor, L, et al. Maternal and neonatal outcome of preeclamptic pregnancies: the potential roles of factor V Leiden mutation and 5,10 methylenetetrahydrofolate reductase. Hypertens Pregnancy 2000; 19: 163–72.Google Scholar
Hague, WM, Dekker, GA. Risk factors for thrombosis in pregnancy. Best Pract Res Clin Haematol 2003; 16:197210.Google Scholar
Chasnoff, IJ, Bussey, ME, Savich, R, et al. Perinatal cerebral infarction and maternal cocaine use. J Pediatr 1986; 108: 456–9.Google Scholar
Heier, LA, Carpanzano, CR, Mast, J, et al. Maternal cocaine abuse: the spectrum of radiologic abnormalities in the neonatal CNS. AJNR Am J Neuroradiol 1991; 12: 951–6.Google Scholar
Ment, LR, Ehrenkranz, RA, Duncan, CC. Bacterial meningitis as an etiology of perinatal cerebral infarction. Pediatr Neurol 1986; 2: 276–9.Google Scholar
Amit, M, Camfield, PR. Neonatal polycythemia causing multiple cerebral infarcts. Arch Neurol 1980; 37: 109–10.Google Scholar
Jarjour, IT, Ahdab-Barmada, M. Cerebrovascular lesions in infants and children dying after extracorporeal membrane oxygenation. Pediatr Neurol 1994; 10:1319.Google Scholar
Pellicer, A, Cabanas, F, Garcia-Alix, A, et al. Stroke in neonates with cardiac right-to-left shunt. Brain Dev 1992; 14: 381–5.Google Scholar
Brenner, B, Fishman, A, Goldsher, D, et al. Cerebral thrombosis in a newborn with a congenital deficiency of antithrombin III. Am J Hematol 1988; 27: 209–11.Google Scholar
Hogeveen, M, Blom, HJ, Van Amerongen, M, et al. Hyperhomocysteinemia as risk factor for ischemic and hemorrhagic stroke in newborn infants. J Pediatr 2002; 141: 429–31.Google Scholar
Garoufi, AJ, Prassouli, AA, Attilakos, AV, et al. Homozygous MTHFR C677T gene mutation and recurrent stroke in an infant. Pediatr Neurol 2006; 35:4951.Google Scholar
Curry, CJ, Bhullar, S, Holmes, J, et al. Risk factors for perinatal arterial stroke: a study of 60 mother-child pairs. Pediatr Neurol 2007; 37:99107.Google Scholar
Lynch, JK, Han, CJ, Nee, LE, et al. Prothrombotic factors in children with stroke or porencephaly. Pediatrics 2005; 116: 447–53.Google Scholar
Kurnik, K, Kosch, A, Strater, R, et al. Recurrent thromboembolism in infants and children suffering from symptomatic neonatal arterial stroke: a prospective follow-up study. Stroke 2003; 34: 2887–92.Google Scholar
Miller, SP, Wu, YW, Lee, J, et al. Candidate gene polymorphisms do not differ between newborns with stroke and normal controls. Stroke 2006; 37: 2678–83.Google Scholar
Hernanz-Schulman, M, Cohen, W, Genieser, NB. Sonography of cerebral infarction in infancy. AJR Am J Roentgenol 1988; 150:897902.Google Scholar
Messer, J, Haddad, J, Casanova, R. Transcranial Doppler evaluation of cerebral infarction in the neonate. Neuropediatrics 1991; 22: 147–51.Google Scholar
Golomb, MR, Dick, PT, MacGregor, DL, et al. Cranial ultrasonography has a low sensitivity for detecting arterial ischemic stroke in term neonates. J Child Neurol 2003; 18:98103.Google Scholar
Cowan, F, Mercuri, E, Groenendaal, F, et al. Does cranial ultrasound imaging identify arterial cerebral infarction in term neonates? Arch Dis Child Fetal Neonatal Ed 2005; 90:F252–6.Google Scholar
Mader, I, Schoning, M, Klose, U, et al. Neonatal cerebral infarction diagnosed by diffusion-weighted MRI: pseudonormalization occurs early. Stroke 2002; 33: 1142–5.CrossRefGoogle ScholarPubMed
Kuker, W, Mohrle, S, Mader, I, et al. MRI for the management of neonatal cerebral infarctions: importance of timing. Childs Nerv Syst 2004; 20: 742–8.Google Scholar
Shroff, M, deVeber, G. Sinovenous thrombosis in children. Neuroimag Clin North Am 2003; 13: 115–38.Google Scholar
De Vries, LS, Van der Grond, J, Van Haastert, IC, et al. Prediction of outcome in new-born infants with arterial ischaemic stroke using diffusion-weighted magnetic resonance imaging. Neuropediatrics 2005; 36:1220.Google Scholar
Groenendaal, F, Benders, MJ, de Vries, LS. Pre-Wallerian degeneration in the neonatal brain following perinatal cerebral hypoxia-ischemia demonstrated with MRI. Semin Perinatol 2006; 30: 146–50.Google Scholar
Kirton, A, Deveber, G, Pontigon, AM, et al. Presumed perinatal ischemic stroke: vascular classification predicts outcomes. Ann Neurol 2008; 63: 436–43.Google Scholar
Weiner, SP, Painter, MJ, Geva, D, et al. Neonatal seizures: electroclinical dissociation. Pediatr Neurol 1991; 7: 363–8.Google Scholar
Koelfen, W, Freund, M, Varnholt, V. Neonatal stroke involving the middle cerebral artery in term infants: clinical presentation, EEG and imaging studies, and outcome. Dev Med Child Neurol 1995; 37: 204–12.Google Scholar
Chalmers, EA. Perinatal stroke: risk factors and management. Br J Haematol 2005; 130: 333–43.Google Scholar
Elbers, J, Viero, S, MacGregor, D, et al. Placental pathology in neonatal stroke. Pediatrics 2011; 127:e722–9.Google Scholar
Baird, TA, Parsons, MW, Phanh, T, et al. Persistent poststroke hyperglycemia is independently associated with infarct expansion and worse clinical outcome. Stroke 2003; 34: 2208–14.Google Scholar
Vannucci, RC, Mujsce, DJ. Effect of glucose on perinatal hypoxic-ischemic brain damage. Biol Neonate 1992; 62: 215–24.Google Scholar
Clancy, RR. Prolonged electroencephalogram monitoring for seizures and their treatment. Clin Perinatol 2006; 33: 649–65, vi.Google Scholar
Monagle, P, Chan, A, Massicotte, P, et al. Antithrombotic therapy in children: the Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy. Chest 2004; 126: 645S–87S.Google Scholar
Jones, MW, Morgan, E, Shelton, JE. Primary care of the child with cerebral palsy: a review of systems (part II). J Pediatr Health Care 2007; 21: 226–37.Google Scholar
Juenger, H, Linder-Lucht, M, Walther, M, et al. Cortical neuromodulation by constraint-induced movement therapy in congenital hemiparesis: an FMRI study. Neuropediatrics 2007; 38: 130–6.Google Scholar
Sutcliffe, TL, Gaetz, WC, Logan, WJ, et al. Cortical reorganization after modified constraint-induced movement therapy in pediatric hemiplegic cerebral palsy. J Child Neurol 2007; 22: 1281–7.Google Scholar
Hoare, BJ, Wasiak, J, Imms, C, et al. Constraint-induced movement therapy in the treatment of the upper limb in children with hemiplegic cerebral palsy. Cochrane Database Syst Rev 2007; 2:CD004149.Google Scholar
Rajapakse, T, Kirton, A. Non-invasive brain stimulation in children: applications and future directions. Transl Neurosci 2013; 4: 217–33.Google Scholar
Trauner, DA, Chase, C, Walker, P, et al. Neurologic profiles of infants and children after perinatal stroke. Pediatr Neurol 1993; 9: 383–6.Google Scholar
Wulfeck, BB, Trauner, DA, Tallal, PA. Neurologic, cognitive, and linguistic features of infants after early stroke. Pediatr Neurol 1991; 7: 266–9.Google Scholar
Tillema, JM, Byars, AW, Jacola, LM, et al. Cortical reorganization of language functioning following perinatal left MCA stroke. Brain Lang 2008; 105:99111.Google Scholar
Jacola, LM, Schapiro, MB, Schmithorst, VJ, et al. Functional magnetic resonance imaging reveals atypical language organization in children following perinatal left middle cerebral artery stroke. Neuropediatrics 2006; 37:4652.Google Scholar
Adhikari, MH, Beharelle, AR, Griffa, A, et al. Computational modeling of resting-state activity demonstrates markers of normalcy in children with prenatal or perinatal stroke. J Neurosci 2015; 35: 8914–24.Google Scholar
Golomb, MR, deVeber, GA, MacGregor, DL, et al. Independent walking after neonatal arterial ischemic stroke and sinovenous thrombosis. J Child Neurol 2003; 18: 530–6Google Scholar
Kirton, A, Shroff, M, Visvanathan, T, et al. Quantified corticospinal tract diffusion restriction predicts neonatal stroke outcome. Stroke 2007; 38: 974–80.Google Scholar
Estan, J, Hope, P. Unilateral neonatal cerebral infarction in full term infants. Arch Dis Child Fetal Neonatal Ed 1997; 76:F8893.Google Scholar
Golomb, MR, Garg, BP, Carvalho, KS, et al. Perinatal stroke and the risk of developing childhood epilepsy. J Pediatr 2007; 151:409–13.e2.Google Scholar
Hetherington, R, Tuff, L, Anderson, P, et al. Short-term intellectual outcome after arterial ischemic stroke and sinovenous thrombosis in childhood and infancy. J Child Neurol 2005; 20: 553–9.Google Scholar
Trauner, DA, Nass, R, Ballantyne, A. Behavioural profiles of children and adolescents after pre- or perinatal unilateral brain damage. Brain 2001; 124:9951002.CrossRefGoogle ScholarPubMed
Mercuri, E, Spano, M, Bruccini, G, et al. Visual outcome in children with congenital hemiplegia: correlation with MRI findings. Neuropediatrics 1996; 27: 184–8.Google Scholar
Guzzetta, A, Fazzi, B, Mercuri, E, et al. Visual function in children with hemiplegia in the first years of life. Dev Med Child Neurol 2001; 43: 321–9.Google Scholar
Wen, TC, Rogido, M, Gressens, P, et al. A reproducible experimental model of focal cerebral ischemia in the neonatal rat. Brain Res Brain Res Protoc 2004; 13:7683.Google Scholar
Derugin, N, Ferriero, DM, Vexler, ZS. Neonatal reversible focal cerebral ischemia: a new model. Neurosci Res 1998; 32: 349–53.CrossRefGoogle ScholarPubMed
Renolleau, S, Aggoun-Zouaoui, D, Ben-Ari, Y, et al. A model of transient unilateral focal ischemia with reperfusion in the P7 neonatal rat: morphological changes indicative of apoptosis. Stroke 1998; 29: 1454–61.Google Scholar
Wendland, M, Manabat, C, Fox, CK, et al. The blood–brain barrier is more preserved in neonatal versus adult rats following transient focal cerebral ischemia. J Cereb Blood Flow Metab 2003; 23:169.Google Scholar
Manabat, C, Han, BH, Wendland, M, et al. Reperfusion differentially induces caspase-3 activation in ischemic core and penumbra after stroke in immature brain. Stroke 200(34): 207–13.Google Scholar
Gonzalez, FF, Larpthaveesarp, A, McQuillen, P, et al. Erythropoietin increases neurogenesis and oligodendrogliosis of subventricular zone precursor cells after neonatal stroke. Stroke 2013; 44: 753–8.Google Scholar
Gonzalez, FF, Abel, R, Almli, CR, et al. Erythropoietin sustains cognitive function and brain volume after neonatal stroke. Dev Neurosci 2009; 31: 403–11.Google Scholar

References

Tamaki, M, Fujitani, Y, Hara, A, et al. The diabetes-susceptible gene SLC30A8/ZnT8 regulates hepatic insulin clearance. J Clin Invest 2013; 123(10): 4513–24.Google Scholar
Thorsson, AV, Hintz, RL. Insulin receptors in the newborn: increase in receptor affinity and number. N Engl J Med 1977; 297(17): 908–12.Google Scholar
Chernausek, SD, Beach, DC, Banach, W, Sperling, MA. Characteristics of hepatic receptors for somatomedin-C/insulin-like growth factor I and insulin in the developing human. J Clin Endocrinol Metab 1987; 64(4): 737–43.Google Scholar
Harken, AH, Filler, RM, AvRuskin, TW, Crigler, JF Jr. The role of “total” pancreatectomy in the treatment of unremitting hypoglycemia of infancy. J Pediatr Surg 1971; 6(3): 284–9.Google Scholar
Menni, F, de Lonlay, P, Sevin, C, et al. Neurologic outcomes of 90 neonates and infants with persistent hyperinsulinemic hypoglycemia. Pediatrics 2001; 107(3): 476–9.Google Scholar
Meissner, T, Wendel, U, Burgard, P, et al. Long-term follow-up of 114 patients with congenital hyperinsulinism. Eur J Endocrinol 2003; 149(1):4351.Google Scholar
Steinkrauss, L, Lipman, TH, Hendell, CD, et al. Effects of hypoglycemia on developmental outcome in children with congenital hyperinsulinism. J Pediatr Nurs 2005; 20(2): 109–18.Google Scholar
Avatapalle, HB, Banerjee, I, Shah, S, et al. Abnormal neurodevelopmental outcomes are common in children with transient congenital hyperinsulinism. Front Endocrinol (Lausanne) 2013; 4:60.Google Scholar
Lord, K, Radcliffe, J, Gallagher, PR, et al. High risk of diabetes and neurobehavioral deficits in individuals with surgically treated hyperinsulinism. J Clin Endocrinol Metab 2015; 100(11): 4133–9.Google Scholar
Hoe, FM, Thornton, PS, Wanner, LA, et al. Clinical features and insulin regulation in infants with a syndrome of prolonged neonatal hyperinsulinism. J Pediatr 2006; 148(2): 207–12.Google Scholar
Hume, R, McGeechan, A, Burchell, A. Failure to detect preterm infants at risk of hypoglycemia before discharge. J Pediatr 1999; 134(4):499502.Google Scholar
Lucas, A, Morley, R, Cole, TJ. Adverse neurodevelopmental outcome of moderate neonatal hypoglycemia. BMJ 1988; 297(6659): 1304–8.Google Scholar
Cornblath, M, Schwartz, R. Outcome of neonatal hypoglycaemia: complete data are needed. BMJ 1999; 318(7177): 194–5.Google Scholar
Tin, W, Brunskill, G, Kelly, T, Fritz, S. 15-year follow-up of recurrent “hypoglycemia” in preterm infants. Pediatrics 2012; 130(6): e1497–503.Google Scholar
Kerstjens, JM, Bocca-Tjeertes, IF, de Winter, AF, et al. Neonatal morbidities and developmental delay in moderately preterm-born children. Pediatrics 2012:130(2): e265–72.Google Scholar
McIntyre, S, Taitz, D, Keogh, J, et al. A systemic review of risk factors for cerebral palsy in children born at term in developed countries. Dev Med Child Neurol 2013; 55(6): 499508.Google Scholar
Tam, EW, Haeusslein, LA, Bonifacio, SL, et al. Hypoglycemia is associated with increased risk for brain injury and adverse neurodevelopmental outcome in neonates at risk for encephalopathy. J Pediatr 2012; 161(1): 8893.Google Scholar
Kaiser, JR, Bai, S, Gibson, N, et al. Association between transient newborn hypoglycemia and fourth-grade achievement test proficiency: a population-based study. JAMA Pediatr 2015; 169(10): 913–21.Google Scholar
McKinlay, CJ, Harding, JE. Revisiting transitional hypoglycemia: only time will tell. JAMA Pediatr 2015; 169(10): 892–4.Google Scholar
Boluyt, N, van Kempen, A, Offringa, M. Neurodevelopment after neonatal hypoglycemia: a systemic review and design of an optimal future study. Pediatrics 2006; 117(6): 2231–43.Google Scholar
Waisbren, SE, Landau, Y, Wilson, J, Vockley, J. Neuropsychological outcomes in fatty acid oxidation disorders: 85 cases detected by newborn screening. Dev Disabil Res Rev 2013; 17(3): 260–8.Google Scholar
Signorini, SG, Decio, A, Fedeli, C, et al. Septo-optic dysplasia in childhood: the neurological, cognitive and neuro-ophthalmological perspective. Dev Med Child Neurol 2012; 54(11): 1018–24.Google Scholar
Stanley, CA, Rozance, PJ, Thornton, PS, et al. Re-evaluating “transitional neonatal hypoglycemia”: mechanism and implications for management. J Pediatr 2015; 166(6): 1520–5.Google Scholar
Thornton, PS, Stanley, CA, De León, DD, et al. Recommendations from the Pediatric Endocrine Society for evaluation and management of persistent hypoglycemia in neonates, infants, and children. J Pediatr 2015; 167(2): 238–45.Google Scholar
Bier, DM, Leake, RD, Haymond, MW, et al. Measurement of “true” glucose production rates in infancy and childhood with 6,6-dideuteroglucose. Diabetes 1977; 26(11): 1016–23.Google Scholar
Kalhan, SC, D’Angelo, LJ, Savin, SM, Adam, PA. Glucose production in pregnant women at term gestation: sources of glucose for human fetus. J Clin Invest 1979; 63(3): 388–94.Google Scholar
Bougneres, PF, Lemmel, C, Ferré, P, Bier, DM. Ketone body transport in the human neonate and infant. J Clin Invest 1986; 77(1): 42–8.Google Scholar
Boardman, JP, Hawdon, JM. Hypoglycemia and hypoxic-ischaemic encephalopathy. Dev Med Child Neurol 2015; 57(Suppl 3):2933.Google Scholar
Sperling, MA, DeLamater, PV, Phelps, D, et al. Spontaneous and amino acid-stimulated glucagon secretion in the immediate postnatal period: relation to glucose and insulin. J Clin Invest 1974; 53(4): 1159–66.Google Scholar
Guemes, M, Rahman, SA, Hussain, K. What is a normal blood glucose? Arch Dis Child 2016; 101(6): 569–74.Google Scholar
Dekelbab, BH, Sperling, MA. Recent advances in hyperinsulinemic hypoglycemia of infancy. Acta Paediatr 2006; 95(10): 1157–64.Google Scholar
McKinlay, CJ, Alsweiler, JM, Ansell, JM, et al. Neonatal glycemia and neurodevelopmental outcomes at 2 years. N Engl J Med 2015; 373(16): 1507–18.Google Scholar
Auer, RN. Hypoglycemic brain damage. Metab Brain Dis 2004; 19(3–4): 169–75.Google Scholar
Turner, CP, Blackburn, MR, Rivkees, SA. A1 adenosine receptors mediate hypoglycemia-induced neuronal injury. J Mol Endocrinol 2004; 32(1): 129–44.Google Scholar
Kim, M, Yu, ZX, Fredholm, BB, Rivkees, SA. Susceptibility of the developing brain to acute hypoglycemia involving A1 adenosine receptor activation. Am J Physiol Endocrinol Metab 2005; 289(4): E562–9.Google Scholar
Mujsce, DJ, Christensen, MA, Vannucci, RC. Regional cerebral blood flow and glucose utilization during hypoglycemia in newborn dogs. Am J Physiol 1989; 256(6 Pt 2): H1659–66.Google Scholar
Karaoğlu, P, Polat, AI, Yiş, U, Hiz, S. Parieto-occipital encephalomalacia in children: clinical and electrophysiological features of twenty-seven cases. J Pediatr Neurosci 2015; 10(2): 103–7.Google Scholar
LaManna, JC, Harik, SI. Regional comparisons of brain glucose influx. Brain Res 1985; 326(2): 299305.Google Scholar
Sperling, MA, Menon, RK. Differential diagnosis and management of neonatal hypoglycemia. Pediatr Clin North Am 2004; 51(3): 703–23.Google Scholar
Duvanel, CB, Fawer, CL, Cotting, J, et al. Long-term effects of neonatal hypoglycemia on brain growth and psychomotor development in small-for-gestational-age preterm infants. J Pediatr 1999; 134(4): 492–8.Google Scholar
Rozance, PJ. Update on neonatal hypoglycemia. Curr Opin Endocrinol Diabetes Obes 2014; 21(1):4550.Google Scholar
Ferrara, C, Patel, P, Becker, S, et al. Biomarkers of insulin for the diagnosis of hyperinsulinemic hypoglycemia in infants. J Pediatr 2016; 168: 212–9.Google Scholar
Kelly, A, Tang, R, Becker, S, Stanley, CA. Poor specificity of low growth hormone and cortisol levels during fasting hypoglycemia for the diagnoses of growth hormone deficiency and adrenal insufficiency. Pediatrics 2008; 122(3): e522–8.Google Scholar
Cornblath, M, Parker, ML, Reisner, SH, et al. Secretion and metabolism of growth hormone in premature and full-term infants. J Clin Endocrinol Metab 1965; 25: 209–18.Google Scholar
Kaplan, SL, Grumbach, MM, Shepard, TH. The ontogenesis of human fetal hormones. I. Growth hormone and insulin. J Clin Invest 1972; 51(12): 3080–93.Google Scholar
Lanes, R, Nieto, C, Bruguera, C, et al. Growth hormone release in response to growth hormone-releasing hormone in term and preterm neonates. Biol Neonate 1989; 56(5): 252–6Google Scholar
Secco, A, di lorgi, N, Napoli, F, et al. The glucagon test in the diagnosis of growth hormone deficiency in children with short stature younger than 6 years. J Clin Endocrinol Metab 2009; 94(11): 4251–7.Google Scholar
Finegold, DN, Stanely, CA, Baker, L. Glycemic response to glucagon during fasting hypoglycemia: an aid in the diagnosis of hyperinsulinism. J Pediatr 1980; 96(2): 257–9.Google Scholar
Rahman, SA, Nessa, A, Hussain, K. Molecular mechanisms of congenital hyperinsulinism. J Mol Endocrinol 2015; 54(2): R119–29.Google Scholar
Barkovich, AJ, Ali, FA, Rowley, HA, Bass, N. Imaging patterns of neonatal hypoglycemia. AJNR Am J Neuroradiol 1998; 19(3): 523–8.Google Scholar
Alkalay, AL, Flores-Sarnat, L, Sarnat, HB, et al. Brain imaging findings in neonatal hypoglycemia: case report and review of 23 cases. Clin Pediatr (Phila) 2005; 44(9): 783–90.Google Scholar
Alkalay, AL, Flores-Sarnat, L, Sarnat, HB, et al. Plasma glucose concentrations in profound neonatal hypoglycemia. Clin Pediatr (Phila) 2006:45(6): 550–8.Google Scholar
Wong, DS, Poskitt, KJ, Chau, V, et al. Brain injury patterns in hypoglycemia in neonatal encephalopathy. AJNR Am J Neuroradiol 2013; 34(7): 1456–61.Google Scholar
Burns, CM, Rutherford, MA, Boardman, JP, Cowan, FM. Patterns of cerebral injury and neurodevelopmental outcomes after symptomatic neonatal hypoglycemia. Pediatrics 2008; 122(1): 6574.Google Scholar
Bozzola, M, Adamsbaum, C, Biscaldi, I, et al. Role of magnetic resonance imaging in the diagnosis and prognosis of growth hormone deficiency. Clin Endocrinol (Oxf) 1996; 45(1): 21–6.Google Scholar
Kornreich, L, Horev, G, Lazar, L, et al. MR findings in growth hormone deficiency: correlation with severity of hypopituitarism. AJNR Am J Neuroradiol 1998; 19(8): 1495–9.Google Scholar
Dutta, P, Bhansali, A, Singh, P, et al. Congenital hypopituitarism: clinico-radiological correlation. J Pediatr Endocrinol Metab 2009; 22(10): 921–8.Google Scholar
Saudubray, JM, Martin, D, de Lonlay, P, et al. Recognition and management of fatty acid oxidation defects: a series of 107 patients. J Inherit Metab Dis 1999; 22(4): 488502.Google Scholar
Shulman, DI, Palmert, MR, Kemp, SF, Lawson Wilkins Drug and Therapeutics Committee. Adrenal insufficiency: still a cause of morbidity and death in childhood. Pediatrics 2007; 119(2): e484–94.Google Scholar
Grimberg, A, DiVall, SA, Polychronakos, C, et al.; Drug and Therapeutics Committee and Ethics Committee of the Pediatric Endocrine Society. Guidelines for growth hormone and insulin-like growth factor-I treatment in children and adolescents: growth hormone deficiency, idiopathic short stature, and primary insulin-like growth factor-I deficiency. Horm Res Paediatr. 2016; 86(6): 361–97.Google Scholar
Tas, E, Mahmood, B, Garibaldi, L, Sperling, M. Liver injury may increase the risk of diazoxide toxicity: a case report. Eur J Pediatr 2015; 174(3): 403–6.Google Scholar
Sperling, MA, Menon, R. Hypoglycemia in the Newborn. In Stevenson, DK, Cohen, RS, Sunshine, P, eds., Neonatology: Clinical Practice and Procedures, 1st ed. New York: McGraw-Hill Education, 2015: 649–64.Google Scholar
Palladino, AA, Bennett, MJ, Stanley, CA. Hyperinsulinism in infancy and childhood: when an insulin level is not always enough. Clin Chem 2008; 54(2): 256–63.Google Scholar
Laje, P, Halaby, L, Adzick, NS, Stanley, CA. Necrotizing enterocolitis in neonates receiving octreotide for the management of congenital hyperinsulinism. Pediatr Diabetes 2010; 11(2): 142–7.Google Scholar
Demirbilek, H, Shah, P, Arya, VB, et al. Long-term follow-up of children with congenital hyperinsulinism on octreotide therapy. J Clin Endocrinol Metab 2014; 99(10): 3660–7.Google Scholar
Senniappan, S, Alexandrescu, S, Tatevian, N, et al. Sirolimus therapy in infants with severe hyperinsulinemic hypoglycemia. N Engl J Med 2014; 370(12): 1131–7.Google Scholar
Gopal-Kothandapani, JS, Hussain, K. Congenital hyperinsulinism: role of fluorine-18 L-3,4 hydroxyphenylalanine positron emission tomography scanning. World J Radiol 2014; 6(6): 252–60.Google Scholar
Suchi, M, MacMullen, CM, Thornton, PS, et al. Molecular and immunohistochemical analyses of the focal form of congenital hyperinsulinism. Mod Pathol 2006; 19(1): 122–9.Google Scholar
Adzick, NS, Thornton, PS, Stanley, CA, et al. A multidisciplinary approach to the focal form of congenital hyperinsulinism leads to successful treatment by partial pancreatectomy. J Pediatr Surg 2004; 39(3): 270–5.Google Scholar
Laje, P, Stanley, CA, Palladino, AA, et al. Pancreatic head resection and roux-en-Y pancreaticojejunostomy for the treatment of the focal form of congenital hyperinsulinism. J Pediatr Surg 2012:47(1): 130–5.Google Scholar

References

Schmorl, G. Zur kenntis des icterus neonatorum. Verh Dtsch Ges Pathol 1903; 6: 109.Google Scholar
American Academy of Pediatrics. Management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation. Pediatrics 2004; 114: 297–16.Google Scholar
Bhutani, VK, Wong, R. Bilirubin-induced neurologic dysfunction (BIND). Semin Fetal Neonatal Med 2015; 20:1.Google Scholar
Johnson, L, Bhutani, VK. The clinical syndrome of bilirubin-induced neurologic dysfunction. Semin Perinatol 2011; 35: 101–13.Google Scholar
Good, WV, Hou, C. Visuocortical bilirubin-induced neurological dysfunction. Semin Fetal Neonatal Med 2015; 20:3741.Google Scholar
Olds, C, Oghalai, JS. Audiologic impairment associated with bilirubin-induced neurologic damage. Semin Fetal Neonatal Med 2015; 20: 42–6.Google Scholar
Rose, J, Vassar, R. Movement disorders due to bilirubin toxicity. Semin Fetal Neonatal Med 2015; 20: 20–5.Google Scholar
Wusthoff, CJ, Loe, IM. Impact of bilirubin-induced neurologic dysfunction on neurodevelopmental outcomes. Semin Fetal Neonatal Med 2015; 20: 52–7.Google Scholar
Govaert, P, Lequin, M, Swarte, R, et al. Changes in globus pallidus with (pre)term kernicterus. Pediatrics 2003; 112: 1256–63.Google Scholar
Penn, AA, Enzmann, DR, Hahn, JS, Stevenson, DK. Kernicterus in a full term infant. Pediatrics 1994; 93: 1003–6.Google Scholar
Wisnowski, JL, Panigrahy, A, Painter, MJ, Watchko, JF. Magnetic resonance imaging of bilirubin encephalopathy: current limitations and future promise. Semin Perinatol 2014; 38: 422–8.Google Scholar
Stevenson, DK, Bartoletti, AL, Ostrander, CR, Johnson, JD. Pulmonary excretion of carbon monoxide in the human newborn infant as an index of bilirubin production. III. Measurement of pulmonary excretion of carbon monoxide after the first postnatal week in premature infants. Pediatrics 1979; 64:598600.Google Scholar
Bhutani, VK, Johnson, L, Sivieri, EM. Predictive ability of a predischarge hour-specific serum bilirubin for subsequent significant hyperbilirubinemia in healthy term and near-term newborns. Pediatrics 1999; 103:614.Google Scholar
Volpe, JJ. Bilirubin and brain injury. In Volpe, JJ, ed., Neurology of the Newborn, 2nd edn. Philadelphia: Saunders, 2000:490514.Google Scholar
Amato, M. Mechanisms of bilirubin toxicity. Eur J Pediatr 1995; 154:S54–9.Google Scholar
Amit, Y, Chan, G, Fedunec, S, et al. Bilirubin toxicity in a neuroblastoma cell line N-115. I. Effects on Na+K+-ATPase, [3H]-thymidine uptake, l-[35S]-methionine incorporation, and mitochondrial function. Pediatr Res 1989; 25: 364–8.Google Scholar
Brito, MA, Silva, RF, Brites, D. Bilirubin induces loss of membrane lipids and exposure of phosphatidylserine in human erythrocytes. Cell Biol Toxicol 2002; 18: 181–92.Google Scholar
Hanko, E, Hansen, TW, Almaas, R, et al. Bilirubin induces apoptosis and necrosis in human NT2-N neurons. Pediatr Res 2005; 57: 179–84.Google Scholar
Wennberg, RP. Cellular basis of bilirubin toxicity. NY State J Med 1991; 91: 493–6.Google Scholar
Wennberg, RP, Gospe, SM Jr., Rhine, WD, et al. Brainstem bilirubin toxicity in the newborn primate may be promoted and reversed by modulating PCO2. Pediatr Res 1993; 34:69.Google Scholar
Brites, D, Fernandes, A. Bilirubin-induced neural impairment:a special focus on myelination, age-related windows of susceptibility and associated co-morbidities. Semin Fetal Neonatal Med 2015; 20: 14–9.Google Scholar
Watchko, JF, Painter, MJ, Panigrahy, A. Are the neuromotor disabilities of bilirubin-induced neurologic dysfunction disorders related to the cerebellum and its connections? Semin Fetal Neonatal Med 2015; 20:4751.Google Scholar
Ahlfors, CE. Measurement of plasma unbound unconjugated bilirubin. Anal Biochem 2000; 279: 130–5.Google Scholar
Ahlfors, CE. Unbound bilirubin associated with kernicterus: a historical approach. J Pediatr 2000; 137: 540–4.Google Scholar
Ahlfors, CE, Wennberg, RP. Bilirubin-albumin binding and neonatal jaundice. Semin Perinatol 2004; 28: 334–9.Google Scholar
Lamola, AA, Bhutani, VK, Du, L, et al. Neonatal bilirubin binding capacity discerns risk of neurological dysfunction. Pediatr Res 2015; 77: 334–9.Google Scholar
Morioka, I, Nakamura, H, Koda, T, et al. Serum unbound bilirubin as a predictor for clinical kernicterus in extremely low birth weight infants at a late age in the neonatal intensive care unit. Brain Dev 2015; 37(8):753–7.Google Scholar
Levine, RL, Fredericks, WR, Rapoport, SI. Clearance of bilirubin from rat brain after reversible osmotic opening of the blood-brain barrier. Pediatr Res 1985; 19: 1040–3.Google Scholar
Dennery, PA, McDonagh, AF, Spitz, DR, et al. Hyperbilirubinemia results in reduced oxidative injury in neonatal Gunn rats exposed to hyperoxia. Free Radic Biol Med 1995; 19: 395–04.Google Scholar
Stocker, R, Yamamoto, Y, McDonagh, AF, et al. Bilirubin is an antioxidant of possible physiological importance. Science 1987; 235: 1043–6.Google Scholar
Berardi, A, Lugli, L, Ferrari, F, et al. Kernicterus associated with hereditary spherocytosis and UGT1A1 promoter polymorphism. Biol Neonate 2006; 90: 243–6.Google Scholar
Kaplan, M, Hammerman, C, Rubaltelli, FF, et al. Hemolysis and bilirubin conjugation in association with UDP-glucuronosyltransferase 1A1 promoter polymorphism. Hepatology 2002; 35: 905–11.Google Scholar
Kaplan, M, Renbaum, P, Levy-Lahad, E, et al. Gilbert syndrome and glucose-6-phosphate dehydrogenase deficiency: a dose-dependent genetic interaction crucial to neonatal hyperbilirubinemia. Proc Natl Acad Sci USA 1997; 94: 12128–32.Google Scholar
Beutler, E, Gelbart, T, Demina, A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter:A balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci USA 1998; 95: 8170–4.Google Scholar
Akaba, K, Kimura, T, Sasaki, A, et al. Neonatal hyperbilirubinemia and mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene: a common missense mutation among Japanese, Koreans and Chinese. Biochem Mol Biol Int 1998; 46: 21–6.Google Scholar
Kaplan, M, Hammerman, C, Renbaum, P, et al. Differing pathogenesis of perinatal bilirubinemia in glucose-6-phosphate dehydrogenase-deficient versus-normal neonates. Pediatr Res 2001; 50: 532–7.Google Scholar
Kaplan, M, Renbaum, P, Vreman, HJ, et al. (TA)n UGT 1A1 promoter polymorphism: a crucial factor in the pathophysiology of jaundice in G-6-PD deficient neonates. Pediatr Res 2007; 61: 727–31.Google Scholar
Kaplan, M, Renbaum, P, Hammerman, C, et al. Heme oxygenase-1 promoter polymorphisms and neonatal jaundice. Neonatology 2014; 106: 323–9.Google Scholar
Denschlag, D, Marculescu, R, Unfried, G, et al. The size of a microsatellite polymorphism of the haem oxygenase 1 gene is associated with idiopathic recurrent miscarriage. Mol Hum Reprod 2004; 10: 211–4.Google Scholar
Exner, M, Minar, E, Wagner, O, Schillinger, M. The role of heme oxygenase-1 promoter polymorphisms in human disease. Free Radic Biol Med 2004; 37: 1097–104.Google Scholar
Takeda, M, Kikuchi, M, Ubalee, R, et al. Microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with susceptibility to cerebral malaria in Myanmar. Jpn J Infect Dis 2005; 58: 268–71.Google Scholar
Yamada, N, Yamaya, M, Okinaga, S, et al. Microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with susceptibility to emphysema. Am J Hum Genet 2000; 66: 187–95.Google Scholar
Katayama, Y, Yokota, T, Zhao, H, et al. Association of HMOX1 gene promoter polymorphisms with hyperbilirubinemia in the early neonatal period. Pediatr Int 2015; 57(4):645–9.Google Scholar
Huang, MJ, Kua, KE, Teng, HC, et al. Risk factors for severe hyperbilirubinemia in neonates. Pediatr Res 2004; 56: 682–9.Google Scholar
Watchko, JF, Daood, MJ, Hansen, TW. Brain bilirubin content is increased in P-glycoprotein-deficient transgenic null mutant mice. Pediatr Res 1998; 44: 763–6.Google Scholar
Mollison, PL, Cutbush, M. Haemaolytic disease of the newborn. In Gairdner, D, ed., Recent Advances in Pediatrics. New York: Blakinston, 1954:110.Google Scholar
Watchko, JF. Vigintiphobia revisited. Pediatrics 2005; 115: 1747–53.Google Scholar
Watchko, JF, Oski, FA. Bilirubin 20 mg/dL = vigintiphobia. Pediatrics 1983; 71: 660–3.Google Scholar
Andersen, DH, Blanc, WA, Crozier, DN, Silverman, WA. A difference in mortality rate and incidence of kernicterus among premature infants allotted to two prophylactic antibacterial regimens. Pediatrics 1956; 18: 614–25.Google Scholar
Stevenson, DK, Fanaroff, AA, Maisels, MJ, et al. Prediction of hyperbilirubinemia in near-term and term infants. Pediatrics 2001; 108: 31–9.Google Scholar
Vreman, HJ, Stevenson, DK, Oh, W, et al. Semiportable electrochemical instrument for determining carbon monoxide in breath. Clin Chem 1994; 40: 1927–33.Google Scholar
Castillo Cuadrado, ME, Bhutani, VK, Aby, JL, et al. Evaluation of a new end-tidal carbon monoxide monitor from the bench to the bedside. Acta Paediatr 2015; 104:e279–82.Google Scholar
Brodersen, R, Stern, L. Deposition of bilirubin acid in the central nervous system: a hypothesis for the development of kernicterus. Acta Paediatr Scand 1990; 79: 12–9.Google Scholar
Bratlid, D, Cashore, WJ, Oh, W. Effect of acidosis on bilirubin deposition in rat brain. Pediatrics 1984; 73: 431–4.Google Scholar
Herschel, M, Karrison, T, Wen, M, et al. Evaluation of the direct antiglobulin (Coombs’) test for identifying newborns at risk for hemolysis as determined by end-tidal carbon monoxide concentration (ETCOc); and comparison of the Coombs’ test with ETCOc for detecting significant jaundice. J Perinatol 2002; 22: 341–7.Google Scholar
Tidmarsh, GF, Wong, RJ, Stevenson, DK. End-tidal carbon monoxide and hemolysis. J Perinatol 2014; 34: 577–81.Google Scholar
Gartner, LM, Snyder, RN, Chabon, RS, Bernstein, J. Kernicterus: high incidence in premature infants with low serum bilirubin concentrations. Pediatrics 1970; 45: 906–17.Google Scholar
Morris, BH, Oh, W, Tyson, JE, et al. Aggressive vs. conservative phototherapy for infants with extremely low birth weight. N Engl J Med 2008; 359: 1885–96.Google Scholar
Tyson, JE, Pedroza, C, Langer, J, et al. Does aggressive phototherapy increase mortality while decreasing profound impairment among the smallest and sickest newborns? J Perinatol 2012; 32: 677–84.Google Scholar
Vreman, HJ, Knauer, Y, Wong, RJ, et al. Dermal carbon monoxide excretion in neonatal rats during light exposure. Pediatr Res 2009; 66: 66–9.Google Scholar
Lamola, AA, Bhutani, VK, Wong, RJ, et al. The effect of hematocrit on the efficacy of phototherapy for neonatal jaundice. Pediatr Res 2013; 74:5460.Google Scholar
Ebbesen, F. Recurrence of kernicterus in term and near-term infants in Denmark. Acta Paediatr 2000; 89: 1213–7.Google Scholar
Ebbesen, F, Andersson, C, Verder, H, et al. Extreme hyperbilirubinaemia in term and near-term infants in Denmark. Acta Paediatr 2005; 94:5964.Google Scholar
Maisels, MJ, Newman, TB. Kernicterus in otherwise healthy, breast-fed term newborns. Pediatrics 1995; 96: 730–3.Google Scholar
Newman, TB, Maisels, MJ. Less aggressive treatment of neonatal jaundice and reports of kernicterus: lessons about practice guidelines. Pediatrics 2000; 105: 242–5.Google Scholar
Perlman, JM, Rogers, BB, Burns, D. Kernicteric findings at autopsy in two sick near term infants. Pediatrics 1997; 99: 612–5.Google Scholar
Fischer, AF, Nakamura, H, Uetani, Y, et al. Comparison of bilirubin production in Japanese and Caucasian infants. J Pediatr Gastroenterol Nutr 1988; 7: 27–9.Google Scholar
Stevenson, DK, Vreman, HJ, Oh, W, et al. Bilirubin production in healthy term infants as measured by carbon monoxide in breath. Clin Chem 1994; 40: 1934–9.Google Scholar

References

Wood, JL. Plethora in the newborn infant associated with cyanosis and convulsions. J Pediatr 1952; 54: 143–51.Google Scholar
Michael, A, Mauer, AM. Maternal-fetal transfusion as a cause of plethora in the neonatal period. Pediatrics 1961; 28: 458–61.Google Scholar
Minkowski, A. Acute cardiac failure in connection with neonatal polycythemia in monovular twins and single newborn infants. Biol Neonate 1962; 4: 6174.Google Scholar
Danks, DM, Stevens, LH. Neonatal respiratory distress associated with a high haematocrit reading. Lancet 1964; 2: 499500.Google Scholar
Gross, CP, Hathaway, WE, McCaughey, HR. Hyperviscosity in the neonate. J Pediatr 1973; 82: 1004–12.Google Scholar
Oh, W, Oh, MA, Lind, J. Renal function and blood volume in newborn infants related to placental transfusion. Acta Paediatr Scand 1966; 56: 197210.Google Scholar
Nowicki, P, Oh, W, Yao, A, et al. Effect of polycythemia on gastrointestinal blood flow and oxygenation in piglets. Am J Physiol 1984; 247: G220–5.Google Scholar
Surjadhana, A, Rouleau, J, Boerboom, L, et al. Myocardial blood flow and its distribution in anesthetized polycythemic dogs. Circ Res 1978; 43: 619–31.Google Scholar
Rosenkrantz, TS, Oh, W. Cerebral blood flow velocity in infants with polycythemia and hyperviscosity: effects of partial exchange transfusion with plasmanate. J Pediatr 1982; 101: 94–8.Google Scholar
Fouron, JC, Hebert, F. The circulatory effects of hematocrit variations in normovolemic newborn lambs. J Pediatr 1973; 82: 9951003.Google Scholar
Gatti, RA, Muster, AJ, Cole, RB, et al. Neonatal polycythemia with transient cyanosis and cardiorespiratory abnormalities. J Pediatr 1966; 69: 1063–72.Google Scholar
Kotagal, VR, Keenan, WJ, Reuter, JH, et al. Regional blood flow in polycythemia and hypervolemia. Pediatr Res 1977; 11: 394.Google Scholar
Kotagal, VR, Kleinman, LI. Effect of acute polycythemia on newborn renal hemodynamics and function. Pediatr Res 1982; 16: 148–51.Google Scholar
Bergqvist, G, Zetterman, R. Blood viscosity and peripheral circulation in newborn infants. Acta Paediatr Scand 1974; 63: 865–8.Google Scholar
Linderkamp, O, Strohhacker, I, Versmold, HT, et al. Peripheral circulation in the newborn: interaction of peripheral blood flow, blood pressure, blood volume and blood viscosity. J Pediatr 1978; 129: 7381.Google Scholar
Gustafsson, L, Applegren, L, Myrvold, HE. The effect of polycythemia on blood flow in working and non-working skeletal muscle. Acta Physiol Scand 1980; 109: 143–8.Google Scholar
Waffarn, F, Cole, CD, Huxtable, RF. Effects of polycythemia on cutaneous blood flow and transcutaneous PO2 and PCO2 in the hyperviscosity neonate. Pediatrics 1984; 74: 389–94.Google Scholar
Jones, MD, Traystman, RJ, Simmons, MA, et al. Effects of changes in arterial O2 content on cerebral blood flow in the lamb. Am J Physiol 1981; 240: H209–15.Google Scholar
Rosenkrantz, TS, Stonestreet, BS, Hansen, NB, et al. Cerebral blood flow in the newborn lamb with polycythemia and hyperviscosity. J Pediatr 1984; 104: 276–80.Google Scholar
Rosenkrantz, TS, Philipps, AF, Skrzypczak, PS, et al. Cerebral metabolism in the newborn lamb with polycythemia. Pediatr Res 1988; 23: 329–33.Google Scholar
Oh, W, Wallgren, G, Hanson, JS, et al. The effects of placental transfusion on respiratory mechanics of normal term newborn infants. Pediatrics 1967; 40: 612.Google Scholar
Hakanson, DO, Oh, W. Necrotizing enterocolitis and hyperviscosity in the newborn infant. J Pediatr 1977; 90: 458–61.Google Scholar
LeBlanc, MH, D’Cruz, C, Pate, K. Necrotizing enterocolitis can be caused by polycythemic hyperviscosity in the newborn dog. J Pediatr 1984; 105: 804–9.Google Scholar
Black, VD, Rumack, CM, Lubchenco, LO, et al. Gastrointestinal injury in polycythemic term infants. Pediatrics 1985; 76: 225–31.Google Scholar
Herson, VC, Raye, JR, Rowe, JC, et al. Acute renal failure associated with polycythemia in a neonate. J Pediatr 1982; 100: 137–9.Google Scholar
Leake, RD, Chan, GM, Zakauddin, S, et al. Glucose perturbation in experimental hyperviscosity. Pediatr Res 1980; 14: 1320–3.Google Scholar
Creswell, JS, Warburton, D, Susa, JB, et al. Hyperviscosity in the newborn lamb produces perturbation in glucose homeostasis. Pediatrics 1981; 15: 1348–50.Google Scholar
Rivers, RPA. Coagulation changes associated with a high haematocrit in the newborn infant. Acta Paediatr Scand 1975; 64: 449–56.Google Scholar
Katz, J, Rodriquez, E, Mandani, G, et al. Normal coagulation findings, thrombocytopenia, and peripheral hemoconcentration in neonatal polycythemia. J Pediatr 1982; 101: 99102.Google Scholar
Henriksson, P. Hyperviscosity of the blood and haemostasis in the newborn infant. Acta Paediatr Scand 1979; 68: 701–4.Google Scholar
Shaikh, BS, Erslev, AJ. Thrombocytopenia in polycythemic mice. J Lab Clin Med 1978; 92: 765–71.Google Scholar
Jackson, CW, Smith, PJ, Edwards, CC, et al. Relationship between packed cell volume platelets and platelet survival in red blood cell–hypertransfused mice. J Lab Clin Med 1979; 94: 500–9.Google Scholar
Meberg, A. Transitory thrombocytopenia in newborn mice after intrauterine hypoxia. Pediatr Res 1980; 14: 1071–3.Google Scholar
Rosenkrantz, TS, Oh, W. Neonatal polycythemia and hyperviscosity. In Milunsky, A, Friedman, EA, Gluck, L, eds., Advances in Perinatal Medicine, vol. 5. New York: Plenum, 1986: 93123.Google Scholar
Cornbleet, J. Spurious results from automated hematology cell counters. Lab Med 1983; 14: 509–14.Google Scholar
Penn, D, Williams, PR, Dutcher, TF, et al. Comparison of hematocrit determination by microhematocrit electronic particle counter. Am J Clin Pathol 1979; 72: 71–4.Google Scholar
Pearson, TC, Guthrie, L. Trapped plasma in the microhematocrit. Am J Clin Pathol 1982; 78: 770–2.Google Scholar
Oh, W, Lind, J. Venous and capillary hematocrit in newborn infants and placental transfusion. Acta Paediatr Scand 1966; 55: 3840.Google Scholar
Shohat, M, Reisner, SH, Mimouni, F, et al. Neonatal polycythemia II. Definition related to time of sampling. Pediatrics 1984; 73: 1113.Google Scholar
Wirth, FH, Goldberg, KE, Lubchenco, LO. Neonatal hyperviscosity. I. Incidence. Pediatrics 1979; 63: 833–6.Google Scholar
Stevens, K, Wirth, FH. Incidence of neonatal hyperviscosity at sea level. J Pediatr 1980; 97: 118–19.Google Scholar
Ghavam, S, Batra, D, Mercer, J, et al. Effects of placental transfusion in extremely low birthweight infants: meta-analysis of long- and short-term outcomes. Transfusion 2014; 54(4): 1192–8.Google Scholar
Christensen, R, Baer, V, Gerday, E, et al. Whole blood viscosity in the neonate: effects of gestational age, hematocrit, mean corpuscular volume, and umbilical cord milking. J Perinatol 2014; 34(1): 1621.Google Scholar
Hutton, EK, Hassan, ES: Late vs early clamping of the umbilical cord in full-term neonates: systematic review and meta-analysis of controlled trials. JAMA. 2007; 297(11): 1241–52.Google Scholar
Oh, W, Blankenship, W, Lind, J. Further study of neonatal blood volume in relation to placental transfusion. Ann Paediatr 1966; 207: 147–59.Google Scholar
Yao, AC, Moinian, M, Lind, J. Distribution of blood between infants and placenta after birth. Lancet 1969; 2: 871–3.Google Scholar
Linderkamp, O. Placental transfusion: determinants and effects. Clin Perinatol 1981; 9: 559–92.Google Scholar
Saigal, S, Usher, RH. Symptomatic neonatal plethora. Biol Neonate 1977; 32: 6272.Google Scholar
Philip, AGS, Yee, AB, Rosy, M, et al. Placental transfusion as an intrauterine phenomenon in deliveries complicated by fetal distress. BMJ 1969; 2: 1113.Google Scholar
Flod, NE, Ackerman, BD. Perinatal asphyxia and residual placenta blood volume. Acta Paediatr Scand 1971; 60: 433–6.Google Scholar
Yao, AC, Lind, J. Effect of gravity on placental transfusion. Lancet 1969; 2: 505–8.Google Scholar
Oh, W, Omori, K, Emmanouilides, GC, et al. Placenta to lamb fetus transfusion in utero during acute hypoxia. Am J Obstet Gynecol 1975; 122: 316–21.Google Scholar
Sacks, MO. Occurrence of anemia and polycythemia in phenotypically dissimilar single ovum human twins. Pediatrics 1959; 24: 604–8.Google Scholar
Schwartz, JL, Maniscalco, WM, Lane, AT, et al. Twin transfusion syndrome causing cutaneous erythropoiesis. Pediatrics 1984; 74: 527–9.Google Scholar
Humbert, JR, Abelson, H, Hathaway, WE, et al. Polycythemia in small for gestational age infants. J Pediatr 1969; 75: 812–19.Google Scholar
Widness, JA, Garcia, JA, Oh, W, et al. Cord serum erythropoietin values and disappearance rates after birth in polycythemic newborns. Pediatr Res 1982; 16: 218A.Google Scholar
Philipps, AF, Dubin, JW, Matty, PJ, et al. Arterial hypoxemia and hyperinsulinemia in the chronically hyperglycemic fetal lamb. Pediatr Res 1982; 16: 653–8.Google Scholar
Wells, RE, Penton, R, Merrill, EW. Measurements of viscosity of biologic fluids by core plate viscometer. J Lab Clin Med 1961; 57: 646–56.Google Scholar
Linderkamp, O, Versmold, HT, Riegel, KP, et al. Contributions of red cells and plasma to blood viscosity in preterm and full-term infants and adults. Pediatrics 1984; 74: 4551.Google Scholar
Poiseuille, JLM. Recherches expérimentales sur le mouvement des liquides dans les tubes de très petits diameters. C R Acad Sci 1840; 11: 9611041.Google Scholar
van der Elst, CW, Malan, AF, de Heese, HV. Blood viscosity in modern medicine. S Afr Med J 1977; 52: 526–8.Google Scholar
Dintenfass, L. Blood viscosity, internal fluidity of the red cell, dynamic coagulation and the critical capillary radius as factors in the physiology and pathology of circulation and microcirculation. Med J Aust 1968; 1: 688–96.Google Scholar
Wells, RE, Merrill, EW. Influence of flow properties of blood upon viscosity-hematocrit relationships. J Clin Invest 1962; 41: 1591–8.Google Scholar
Bergqvist, G. Viscosity of the blood in the newborn infants. Acta Paediatr Scand 1974; 63: 858–64.Google Scholar
Wells, R. Syndromes of hyperviscosity. N Engl J Med 1970; 283: 183–6.Google Scholar
Smith, CM, Prasler, WJ, Tukey, DP, et al. Fetal red cells are more deformable than adult red cells. Blood 1981; 58: 35a.Google Scholar
Lichtman, MA. Cellular deformability during maturation of the myeloblast: possible role in marrow egress. N Engl J Med 1970; 283: 943–8.Google Scholar
Lichtman, MA. Rheology of leukocytes, leukocyte suspensions, and blood in leukemia. J Clin Invest 1973; 52: 350.Google Scholar
Miller, ME. Developmental maturation of human neutrophil motility and its relationship to membrane deformability. In Bellanti, UA, Dayton, DH, eds., The Phagocytic Cell in Host Resistance. New York: Raven Press, 1975: 295.Google Scholar
Burton, AC. Role of geometry, of size and shape, in the microcirculation. Fed Proc 1966; 25: 1753–60.Google Scholar
Fahraeus, R, Lindqvist, T. The viscosity of the blood in narrow capillary tubes. Am J Physiol 1931; 96: 561–8.Google Scholar
Goldstein, M, Stonestreet, BS, Brann, BS, et al. Cerebral cortical blood flow and oxygen metabolism in normocythemic hyperviscous newborn piglets. Pediatr Res 1988; 24: 486–9.Google Scholar
Massik, J, Tang, YL, Hudak, ML, et al. Effect of hematocrit on cerebral blood flow with induced polycythemia. J Appl Physiol 1987; 62: 1090–6.Google Scholar
Rosenkrantz, TS, Philipps, AF, Knox, I, et al. Regulation of cerebral glucose metabolism in normal and polycythemic newborns. J Cereb Blood Flow Metab 1992; 12: 856–65.Google Scholar
LeBlanc, MH, Kotagal, UR, Kleinman, LI. Physiological effects of hypervolemic polycythemia in newborn dogs. J Appl Physiol 1982; 53: 865–72.Google Scholar
Murphy, DJ, Reller, MD, Meyer, RA, et al. Effects of neonatal polycythemia and partial exchange transfusion on cardiac function: an echocardiographic study. Pediatrics 1985; 76: 909–13.Google Scholar
Brashear, RE. Effects of acute plasma for blood exchange in experimental polycythemia. Respiration 1980; 40: 297306.Google Scholar
Bada, HS, Korones, SB, Pourcyrous, M, et al. Asymptomatic syndrome of polycythemic hyperviscosity: effect of partial plasma exchange transfusion. J Pediatr 1992; 120: 579–85.Google Scholar
Ramamurthy, RS, Brans, YW. Neonatal polycythemia. I. Criteria for diagnosis and treatment. Pediatrics 1981; 68: 168–74.Google Scholar
Goldberg, K, Wirth, FH, Hathaway, WE, et al. Neonatal hyperviscosity. II. Effect of partial plasma exchange transfusion. Pediatrics 1982; 69: 419–25.Google Scholar
van der Elst, CW, Molteno, CD, Malan, AF, et al. The management of polycythemia in the newborn infant. Early Hum Dev 1980; 4: 393403.Google Scholar
Høst, A, Ulrich, M. Late prognosis in untreated neonatal polycythemia with minor or no symptoms. Acta Paediatr Scand 1982; 71: 629–33.Google Scholar
Black, VD, Lubchenco, LD, Luckey, DW, et al. Developmental and neurologic sequelae of neonatal hyperviscosity syndrome. Pediatrics 1982; 69: 426–31.Google Scholar
Hakanson, DO, Oh, W. Hyperviscosity in the small-for-gestationa1 age infant. Biol Neonate 1980; 37: 109–12.Google Scholar
Black, VD, Lubchenco, LO, Koops, BL, et al. Neonatal hyperviscosity: randomized study of effect of partial plasma exchange on long-term outcome. Pediatrics 1985; 75: 1048–53.Google Scholar
Black, VD, Camp, BW, Lubchenco, LO, et al. Neonatal hyperviscosity is associated with lower achievement and IQ scores at school age. Pediatr Res 1988; 23: 442A.Google Scholar
Dempsey, EM, Barrington, K. Short and long term outomes following partial exchange transfusion in the polycythemic newborn: a systematic review. Arch Dis Child Fetal Neonatal Ed 2006; 91: F26.Google Scholar
American Academy of Pediatrics Committee on the Fetus and Newborn. Routine evaluation of blood pressure, hematocrit and glucose in newborns. Pediatrics 1993; 92: 474–6.Google Scholar
Rosenkrantz, TS. Polycythemia and hyperviscosity in the newborn. Semin Thromb Hemost 2003; 29: 515–27.Google Scholar
Bowman, JM, Pollock, JM, Penston, LE. Fetomaternal transplacental hemorrhage during pregnancy and after delivery. Vox Sang 1986; 51: 117–21.Google Scholar
Scott, JR, Warenski, JC. Tests to detect and quantitate fetal maternal bleeding. Clin Obstet Gynecol 1982; 25: 277.Google Scholar
Sebring, ES, Polesky, HF. Fetomaternal hemorrhage: Incidence, risk factors, time of occurrence and clinical effects. Transfusion 1990; 30: 344–57.Google Scholar
Pollack, W, Ascari, WQ, Kochesky, RJ et al. Studies on Rh prophylaxis. 1. Relationship between dose of anti-Rh and size of antigenic stimulus. Transfusion 1971; 11: 333–9.Google Scholar
De Almeida, V, Bowman, JM. Massive fetomaternal hemorrhage: Manitoba experience. Obstet Gynecol 1994; 83: 323–8.Google Scholar
Rubod, C, Deruelle, P, Le Goueff, F, et al. Long term prognosis for infants after massive fetomaternal hemorrhage. Obstet Gynecol 2007; 110(2 pt 1): 256–60.Google Scholar
Giacoia, GP. Severe fetomaternal hemorrhage: a review. Obstet Gynecol Surv 1997; 52: 372.Google Scholar
Salim, R, Ben- Shlomo, I, Nachum, Z et al. The incidence of large fetomaternal hemorrhage and Kleihauer-Betke test. Obstet Gynecol 2005; 105(5 Pt 1): 1039–44.Google Scholar
Kosasa, T, Ebesugawa, I, Nakayama, R, Hale, R. Massive fetomaternal hemorrhage precedes by decreased fetal movement and a nonreactive fetal heart rate pattern. Obstet Gynecol 1993; 82: 711–14Google Scholar
Markham, LA, Charsha, DS, Peelmuter, B. Case report of massive fetomaternal hemorrhage and a guideline for acute neonatal management. Adv Neonat Care 2006, 6: 197207.Google Scholar
Devi, B, Jennison, RF, Langley, FA. Significance of placental pathology in transplacental hemorrhage. J Clin Pathol 1968; 21: 322–31.Google Scholar
Ness, PM, Baldwin, ML, Niebyl, JR. Clinical high risk designation does not predict excess fetal-maternal hemorrhage. Am J Obstet Gynecol 1987; 156: 154–8.Google Scholar
Banninger, V, Schmid, J. External cephalic version close to term in the management of breech presentation. Z Geburtshilfe Perinatol 1977; 181: 189192.Google Scholar
Blom, AH, Gevers, RH. Een patient met diffuse choriangiomatosis placenta. Ned Tijdschr Geneeskd 1974; 118: 710.Google Scholar
Blackburn, GK. Massive fetomaternal hemorrhage due to choricarcinoma of the uterus. J Pediatr 1976; 89: 680681.Google Scholar
Shankar, M, Gough, GW, Chakravarti, S, Vellacott, ID. Massive feto-maternal haemorrhage with good perinatal outcome following failed external cephalic version. Fetal Diang Ther 2004; 19: 6871.Google Scholar
Lam, CM, Wong, SF, Lee, DW, et al. Massive feto-maternal hemorrhage: an early presentation of women with gestational choriocarcinoma. Acta Obstet Gynecol Scand 2002; 81: 573–76.Google Scholar
Kleihauer, E, Betke, K. Practical use of the demonstration of cells containing hemoglobin F in fixed blood smears (in German). Izv Mikrobiol Inst 1960; 1: 292–5.Google Scholar
Polesky, HF, Sebring, ES. Evaluation of methods for detection and quantitation of fetal cells and their effect on RhIG usage. Am J Clin Pathol 1981; 76: 525–9.Google Scholar
Cunningham, FG, Gant, NF, Leveno, KJ et al. Disease and injuries of the fetus and newborn. In Williams Obstetrics, 21st ed. New York: McGraw- Hill, 2001: 1039–91.Google Scholar
Chen, JC, Bigelelow, NC, Davis, BH. Proposed flow cytometric reference method for the determination of erythroid F-cell counts. Cytometry 2000; 42: 239–46.Google Scholar
Porra, V, Bernaud, J, Gueret, P, et al. Identification and quantification of fetal RBCs in maternal blood by a dual-color flow cytometric method: evaluation of fetal cell count kit. Transfusion 2007; 47: 1281–9.Google Scholar
Bromilow, IM, Duguid, JK. Measurement of feto-maternal haemorrhage: a comparative study of three Kleihauer techniques and two flow cytometry methods. Clin Lab Haemtol 1997; 19: 137–42.Google Scholar
Fong, EA, Davies, JI, Grey, DE, et al. Detection of massive transplacental haemorrhage by flow cytometry. Clin Lab Haemtol 2000; 22: 325–7.Google Scholar
Murphy, KW, Venkatraman, N, Stevens, J. Limitations of ultrasound in the diagnosis of fetomaternal hemorrhage. BJOG 2000; 107: 1317.Google Scholar
Glasser, L, West, JH, et al. Incompatible fetomaternal transfusion with maternal intravascular lysis. Transfusion 1970; 10: 322.Google Scholar
Malcus, P, Bjorklund, LJ, Lilja, M, et al. Massive feto-maternal Hemorrhage: diagnosis by cardiotocography, Doppler ultrasonography and ST waveform analysis of fetal electrcardiography. Fetal Diagn Ther 2006; 21: 812.Google Scholar
Moise, KJ Jr. The usefulness of middle cerebral artery Doppler assessment in the treatment of the fetus at risk for anemia. Am J Obstet Gynecol 2008; 161: el4.Google Scholar
Rubod, C, Houfflin, V, Belot, F, et al. Successful in utero treatment of chronic and massive fetomaternal hemorrhage with fetal hydrops. Fetal Diagn Ther 2006; 21: 410–13.Google Scholar
Wylie, JW, D’Alton, ME. Fetomaternal hemorrhage. Obstet Gynecol 2010; 115: 1039–51.Google Scholar
Kecskes, Z. Large fetomaternal hemorrhage: clinical presentation and outcome. J Maternal-Fetal and Neonatal Medicine. 2003; 13: 128–32.Google Scholar

References

Bukowski, R, Saade, GR. Hydrops fetalis. Clin Perinatol 2000; 27: 1007–31.Google Scholar
Bellini, C, Hennekam, RCM, Fulcheri, E, et al. Etiology of nonimmune hydrops fetalis: a systematic review. Am J Med Genet A 2010; 152A: 1189–96.Google Scholar
Bellini, C. Immune hydrops fetalis. Available at www.orpha.net/consor/cgi-bin/OC_Exp.php?lng=EN&Expert=364013.Google Scholar
Bellini, C. Nonimmune hydrops fetalis. Available at www.orpha.net/consor/cgi-bin/OC_Exp.php?lng=EN&Expert=363999.Google Scholar
Bellini, C, Donarini, G, Paladini, D, et al. Etiology of non-immune hydrops fetalis: an update. Am J Med Genet A 2015; 167: 1082–8.Google Scholar
Rodríguez, MM, Chaves, F, Romaguera, RL, et al. Value of autopsy in nonimmune hydrops fetalis: series of 51 stillborn fetuses. Pediatr Dev Pathol 2002; 5: 365–74.Google Scholar
Bellini, C, Boccardo, F, Bonioli, E, et al. Lymphodynamics in the fetus and newborn. Lymphology 2006; 39: 110–11.Google Scholar
Randenberg, AL. Nonimmune hydrops fetalis. I. Etiology and pathophysiology. Neonatal Netw 2010; 29: 281–95.Google Scholar
Randenberg, AL. Nonimmune hydrops fetalis. II. Does etiology influence mortality? Neonatal Netw 2010; 29: 367–80.Google Scholar
Bellini, C, Hennekam, RC. Non-immune hydrops fetalis: a short review of etiology and pathophysiology. Am J Med Genet A 2012; 158A: 597605.Google Scholar
Bauer, k, Brace, RA, Stonestreet, BS. Fluid distribution in the fetus and neonate. In Polin, RA, Fox, WW, Abman, SH, eds., Fetal and Neonatal Physiology, 4th edn. Philadelphia: Elsevier Saunders, 2011: 1436–44.Google Scholar
Lorenz, JM. Fetal and neonatal body water compartment volumes with references to growth and development. In Polin, RA, Fox, WW, Abman, SH, eds., Fetal and Neonatal Physiology, 4th edn. Philadelphia: Elsevier Saunders, 2011: 1445–50.Google Scholar
Louveau, A, Smirnov, I, Keyes, TJ, et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015; 523(7560):337–41.Google Scholar
Brace, RA. Fetal thoracic duct lymph flow response to intravascular saline infusion. Am J Physiol 1988; 254: 1007–10.Google Scholar
Levick, JR 1, Michel, CC. Microvascular fluid exchange and the revised Starling principle. Cardiovasc Res 2010; 87: 198210.Google Scholar
Gilbert, WM, Brace, RA. Amniotic fluid volume and normal flows to and from the amniotic cavity. Semin Perinatol 1993; 17: 150–7.Google Scholar
Magann, EF, Sandlin, AT, Ounpraseuth, ST. Amniotic fluid and the clinical relevance of the sonographically estimated amniotic fluid volume: oligohydramnios. J Ultrasound Med 2011; 30: 1573–85.Google Scholar
Kiserud, T. Physiology of the fetal circulation. Semin Fetal Neonatal Med 2005; 10: 493503.Google Scholar
Rudolph, AM. Congenital cardiovascular malformations and the fetal circulation. Arch Dis Child Fetal Neonatal Ed. 2010:95(2): F132–6.Google Scholar
Lumbers, ER, Gunn, AJ, Zhang, DY, et al. Nonimmune hydrops fetalis and activation of the renin-angiotensin system after asphyxia in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol 2001; 280: R1045–51.Google Scholar
Keunen, H, Blanco, CE, van Reempts, JL, et al. Absence of neuronal damage after umbilical cord occlusion of 10, 15, and 20 minutes in midgestation fetal sheep. Am J Obstet Gynecol 1997; 176: 515–20.Google Scholar
O’Connell, AE, Boyce, AC, Lumbers, ER, et al. The effects of asphyxia on renal function in fetal sheep at midgestation. J Physiol 2003; 552: 933–43.Google Scholar
Nathanielsz, PW, Hanson, MA. The fetal dilemma: spare the brain and spoil the liver. J Physiol 2003; 548: 333.Google Scholar
Tchirikov, M, Kertschanska, S, Schröder, HJ. Differential effects of catecholamines on vascular rings from ductus venosus and intrahepatic veins of fetal sheep. J Physiol 2003; 548: 519–26.Google Scholar
Bellini, C, Hennekam, RC, Bonioli, E. A diagnostic flow chart for non-immune hydrops fetalis. Am J Med Genet A 2009; 149A: 852–53.Google Scholar
Norton, ME, Chauhan, SP, Dashe, JS. Society for maternal-fetal medicine (SMFM) clinical guideline 7: nonimmune hydrops fetalis. Am J Obstet Gynecol 2015; 212: 127–39.Google Scholar
Désilets, V1, Audibert, F. Investigation and management of non-immune fetal hydrops. Society of Obstetrician and Gynaecologists of Canada. J Obstet Gynaecol Can 2013; 35: 923–38.Google Scholar
Jaeggi, E, Laskin, C, Hamilton, R, et al. The importance of the level of maternal anti-Ro/SSA antibodies as a prognostic marker of the development of cardiac neonatal lupus erythematosus a prospective study of 186 antibody-exposed fetuses and infants. J Am Coll Cardiol 2010; 55: 2778–84.Google Scholar
Winichagoon, P, Kumpan, P, Holmes, P, et al. Validation of the immunochromatographic strip for α-thalassemia screening: a multicenter study. Transl Res 2014; 5: S1931–52.Google Scholar
Izmirly, PM, Saxena, A, Kim, MY, et al. Maternal and fetal factors associated with mortality and morbidity in a multi-racial/ethnic registry of anti-SSA/Ro-associated cardiac neonatal lupus. Circulation. 2011; 124: 1927–35.Google Scholar
Bebbington, M. Twin-to-twin transfusion syndrome: current understanding of pathophysiology, in-utero therapy and impact for future development. Semin Fetal Neonatal Med 2010; 15: 1520.Google Scholar
Mosquera, C1, Miller, RS, Simpson, LL. Twin-twin transfusion syndrome. Semin Perinatol 2012; 36: 182–9.Google Scholar
Benoit, RM, Baschat, AA. Twin-to-twin transfusion syndrome: prenatal diagnosis and treatment. Am J Perinatol 2014; 31: 583–94.Google Scholar
Berry, SM, Stone, J, Norton, ME, et al. Fetal blood sampling. Society for Maternal-Fetal Medicine (SMFM). Am J Obstet Gynecol 2013; 209: 170–80.Google Scholar
Tudor Car, L, van-Velthoven, MH, Brusamento, S, et al. Integrating prevention of mother-to-child HIV transmission (PMTCT) programmes with other health services for preventing HIV infection and improving HIV outcomes in developing countries. Cochrane Database Syst Rev 2011; 6: CD008741.Google Scholar
Shahrook, S, Mori, R, Ochirbat, T, et al. Strategies of testing for syphilis during pregnancy. Cochrane Database Syst Rev 2014; 10: CD010385.Google Scholar
Neu, N, Duchon, J, Zachariah, P. TORCH infections. Clin Perinatol 2015; 42: 77103.Google Scholar
Rodríguez, MM, Chaves, F, Romaguera, RL, et al. Value of autopsy in nonimmune hydrops fetalis: series of 51 stillborn fetuses. Pediatr Dev Pathol 2002; 5: 365–74.Google Scholar
Wainwright, HC. My approach to performing a perinatal or neonatal autopsy. J Clin Pathol 2006; 59: 673–80.Google Scholar
Bellini, C, Fulcheri, E, Rutigliani, M, et al. Immunohistochemistry in non-immune hydrops fetalis: a single center experience in 79 fetuses. Am J Med Genet A 2010; 152A: 1189–96.Google Scholar
Désilets, V, Oligny, LL. Fetal and perinatal autopsy in prenatally diagnosed fetal abnormalities with normal karyotype. J Obstet Gynaecol Can 2011; 33: 1047–57.Google Scholar
Heller, DS, Faye-Petersen, OM. Pathology of the stillborn infant for the general pathologist, part 1. Adv Anat Pathol 2015; 22: 128.Google Scholar
Faye-Petersen, OM, Heller, DS. Pathology of the stillborn infant for the general pathologist, part 2. Adv Anat Pathol 2015; 22: 7193.Google Scholar
Dreux, S, Salomon, LJ, Rosenblatt, J, et al. Biochemical analysis of ascites fluid as an aid to etiological diagnosis: a series of 100 cases of nonimmune fetal ascites. Prenat Diagn. 2015; 35: 214–20.Google Scholar
Bellini, C, Rutigliani, M, Boccardo, FM, et al. Nuchal translucency and lymphatic system maldevelopment. J Perinat Med 2009; 37: 673–6.Google Scholar
Bellini, C, Rutigliani, M, Boccardo, F, et al. Perinatal deaths and lymphatic system involvement: a diagnostic flow-chart applying immunohistochemical methods. Lymphology 2011; 44: 131–3.Google Scholar
Bellini, C, Hennekam, RC. Clinical disorders of primary malfunctioning of the lymphatic system. Adv Anat Embryol Cell Biol 2014; 214: 187204.Google Scholar
Kooper, AJ, Janssens, PM, de Groot, AN, et al. Lysosomal storage diseases in non-immune hydrops fetalis pregnancies. Clin Chim Acta 2006; 371: 176–82.Google Scholar
Gort, L, Granell, MR, Fernández, G, et al. Fast protocol for the diagnosis of lysosomal diseases in nonimmune hydrops fetalis. Prenat Diagn 2012; 32: 1139–42.Google Scholar
Whybra, C, Mengel, E, Russo, A, et al. Lysosomal storage disorder in non-immunological hydrops fetalis (NIHF): more common than assumed? Report of four cases with transient NIHF and a review of the literature. Orphanet J Rare Dis 2012; 7: 86.Google Scholar
Surmeli-Onay, O, Yakarisik, S, Korkmaz, A, et al. Prenatal-onset Niemann-Pick type C disease with nonimmune hydrops fetalis. Pediatr Neonatol 2013; 54: 344–47.Google Scholar
Adams, LL, Gungor, S, Turan, S, et al. When are amniotic fluid viral PCR studies indicated in prenatal diagnosis? Prenat Diagn 2012; 32: 8893.Google Scholar
Andescavage, NN, du Plessis, A, Limperopoulos, C. Advanced MR imaging of the placenta: exploring the in utero placenta-brain connection. Semin Perinatol 2015; 39: 113–23.Google Scholar
Bellini, C, Boccardo, F, Taddei, G, et al. Diagnostic protocol for lymphoscintigraphy in newborns. Lymphology 2005; 38: 915.Google Scholar
Bellini, C, Boccardo, F, Campisi, C, et al. Lymphatic dysplasias in newborns and children: the role of lymphoscintigraphy. J Pediatr 2008; 152: 587–9.Google Scholar
Bellini, C, Di Battista, E, Boccardo, F, et al. The role of lymphoscintigraphy in the diagnosis of lymphedema in Turner syndrome. Lymphology 2009; 42: 123–9.Google Scholar
Bellini, C, Ergaz, Z, Radicioni, M, et al. Congenital fetal and neonatal visceral chylous effusions: neonatal chylothorax and chylous ascites revisited. A multicenter retrospective study. Lymphology 2012; 45: 91102.Google Scholar
Bellini, C, Villa, G, Sambuceti, G, et al. Lymphoscintigraphy patterns in newborns and children with congenital lymphatic dysplasia. Lymphology 2014; 47: 2839.Google Scholar
Brouillard, P, Boon, L, Vikkula, M. Genetics of lymphatic anomalies. J Clin Invest 2014; 124: 898904.Google Scholar
Adzick, NS. Prospects for fetal surgery. Early Hum Dev 2013; 89: 881–6.Google Scholar
Van Mieghem, T, Al-Ibrahim, A, Deprest, J, et al. Minimally invasive therapy for fetal sacrococcygeal teratoma: case series and systematic review of the literature. Ultrasound Obstet Gynecol 2014; 43: 611–9.Google Scholar
Wenstrom, KD, Carr, SR. Fetal surgery: principles, indications, and evidence. Obstet Gynecol 2014; 124: 817–35.Google Scholar
Carlton, DP. Hydrops fetalis. In Stevenson, DK, Benitz, WE, Sunshine, P, et al., eds., Fetal and Neonatal Brain Injury, 4th edn. Cambridge University Press, 2009: 325–30.Google Scholar
Bellini, C, Hennekam, RC, Boccardo, F, et al. Nonimmune idiopathic hydrops fetalis and congenital lymphatic dysplasia. Am J Med Genet A 2006; 140A: 678–84.Google Scholar
Thoresen, M. Who should we cool after perinatal asphyxia? Semin Fetal Neonatal Med 2015; 20: 6671.Google Scholar

References

Freedman, RM, Ingram, DL, Gross, I, et al. A half century of neonatal sepsis at Yale: 1928 to 1978. Am J Dis Child 1981; 135: 140–4.Google Scholar
Bizzarro, MJ, Dembry, LM, Baltimore, RS, Gallagher, PG. Changing patterns in neonatal Escherichia coli sepsis and ampicillin resistance in the era of intrapartum antibiotic prophylaxis. Pediatrics 2008; 121: 689–96.Google Scholar
Bizzarro, MJ, Raskind, C, Baltimore, RS, Gallagher, PG. Seventy-five years of neonatal sepsis at Yale: 1928–2003. Pediatrics 2005; 116: 595602.Google Scholar
Bizzarro, MJ, Shabanova, V, Baltimore, RS, et al. Neonatal sepsis 2004–2013: the rise and fall of coagulase-negative staphylococci. J Pediatr 2015; 166: 1193–9.Google Scholar
Gladstone, IM, Ehrenkranz, RA, Edberg, SC, Baltimore, RS. A ten-year review of neonatal sepsis and comparison with the previous fifty-year experience. Pediatr Infect Dis J 1990; 9: 819–25.Google Scholar
Baltimore, RS, Huie, SM, Meek, JI, et al. Early-onset neonatal sepsis in the era of group B streptococcal prevention. Pediatrics 2001; 108: 1094–8.Google Scholar
Chen, KT, Puopolo, KM, Eichenwald, EC, et al. No increase in rates of early-onset neonatal sepsis by antibiotic-resistant group B Streptococcus in the era of intrapartum antibiotic prophylaxis. Am J Obstet Gynecol 2005; 192: 1167–71.Google Scholar
Daley, AJ, Isaacs, D. Ten-year study on the effect of intrapartum antibiotic prophylaxis on early onset group B streptococcal and Escherichia coli neonatal sepsis in Australasia. Pediatr Infect Dis J 2004; 23: 630–4.Google Scholar
Edwards, RK, Jamie, WE, Sterner, D, et al. Intrapartum antibiotic prophylaxis and early-onset neonatal sepsis patterns. Infect Dis Obstet Gynecol 2003; 11: 221–6.Google Scholar
Moore, MR, Schrag, SJ, Schuchat, A. Effects of intrapartum antimicrobial prophylaxis for prevention of group-B streptococcal disease on the incidence and ecology of early-onset neonatal sepsis. Lancet Infect Dis 2003; 3: 201–13.Google Scholar
Schrag, SJ, Stoll, BJ. Early-onset neonatal sepsis in the era of widespread intrapartum chemoprophylaxis. Pediatr Infect Dis J 2006; 25: 939–40.Google Scholar
Baker, CJ, Halsey, NA, Schuchat, A. 1997 AAP guidelines for prevention of early-onset group B streptococcal disease. Pediatrics 1999; 103: 701.Google Scholar
Eichenwald, EC. Perinatally transmitted neonatal bacterial infections. Infect Dis Clin North Am 1997; 11: 223–39.Google Scholar
Hyde, TB, Hilger, TM, Reingold, A, et al. Trends in incidence and antimicrobial resistance of early-onset sepsis: population-based surveillance in San Francisco and Atlanta. Pediatrics 2002; 110: 690–5.Google Scholar
Schrag, S, Schuchat, A. Prevention of neonatal sepsis. Clin Perinatol 2005; 32: 601–15.Google Scholar
Schuchat, A, Zywicki, SS, Dinsmoor, MJ, et al. Risk factors and opportunities for prevention of early-onset neonatal sepsis: a multicenter case-control study. Pediatrics 2000; 105: 21–6.Google Scholar
Vesikari, T, Janas, M, Gronroos, P, et al. Neonatal septicaemia. Arch Dis Child 1985; 60: 542–6.Google Scholar
Edwards, MS, Nizet, V, Baker, CJ. Group B streptococcal infections. In Infectious Diseases of the Fetus and Newborn Infant, 6th edn. Philadelphia: Saunders, 2006: 403–64.Google Scholar
Lukacs, SL, Schoendorf, KC. National estimates of newborn sepsis rates in the United States, 1990–2001. Presented at the Annual Meeting of the Society for Pediatric and Perinatal Epidemioloic Research, Salt Lake City, UT, 2004.Google Scholar
Lukacs, SL, Schoendorf, KC, Schuchat, A. Trends in sepsis-related neonatal mortality in the United States, 1985–1998. Pediatr Infect Dis J 2004; 23: 599603.Google Scholar
Schrag, SJ, Hadler, JL, Arnold, KE, et al. Risk factors for invasive, early-onset Escherichia coli infections in the era of widespread intrapartum antibiotic use. Pediatrics 2006; 118: 570–6.Google Scholar
Seale, AC, Blencowe, H, Zaidi, A, et al. Neonatal severe bacterial infection impairment estimates in South Asia, sub-Saharan Africa, and Latin America for 2010. Pediatr Res 2013; 74: 7385.Google Scholar
Ganatra, HA, Zaidi, AK. Neonatal infections in the developing world. In Seminars in Perinatology. New York: Elsevier, 2010: 416–25.Google Scholar
Thaver, D, Zaidi, AK. Burden of neonatal infections in developing countries: a review of evidence from community-based studies. Pediatr Infect Dis 2009; 28: S39.Google Scholar
Zaidi, AKM, Darmstadt, GL, Stoll, B. Neonatial infections: a global perspective. In Infectious Disease of the Fetus and Newborn Infant, 8th edn. Philadelphia: Saunders, 2014.Google Scholar
Darmstadt, GL, Saha, SK, Choi, Y, et al. Population-based incidence and etiology of community-acquired neonatal bacteremia in Mirzapur, Bangladesh: an observational study. J Infect Dis 2009; 200: 906–15.Google Scholar
Seale, AC, Blencowe, H, Manu, AA, et al. Estimates of possible severe bacterial infection in neonates in sub-Saharan Africa, South Asia, and Latin America for 2012: a systematic review and meta-analysis. Lancet Infect Dis 2014; 14: 731–41.Google Scholar
Darmstadt, GL, Ahmed, S, Ahmed, ANU, Saha, SK. Mechanism for prevention of infection in preterm neonates by topical emollients: a randomized, controlled clinical trial. Pediatr Infect Dis J 2014; 33: 1124–7.Google Scholar
Darmstadt, GL, Saha, SK, Ahmed, S, et al. Effect of topical emollient treatment of preterm neonates in Bangladesh on invasion of pathogens into the bloodstream. Pediatr Res 2007; 61: 588–93.Google Scholar
Arachaisri, T, Ballow, M. Developmental immunology of the newborn. Immunol Allergy Clin North Am 1999; 19: 253–79.Google Scholar
Lewis, D, Wilson, C. Developmental Immunology and Role of the Host Defenses in Neonatal Susceptibility to Infection, 4th edn. Philadelphia: Saunders, 1995.Google Scholar
Georgeson, GD, Szony, BJ, Streitman, K, et al. Natural killer cell cytotoxicity is deficient in newborns with sepsis and recurrent infections. Eur J Pediatr 2001; 160: 478–82.Google Scholar
Wilson, DC, Edgar, JD. Predictors of bacterial infection in neonates. J Pediatr 1997; 130: 166.Google Scholar
Herbst, A, Källén, K. Time between membrane rupture and delivery and septicemia in term neonates. Obstet Gynecol 2007; 110: 612–18.Google Scholar
Gerdes, JS. Diagnosis and management of bacterial infections in the neonate. Pediatr Clin North Am 2004; 51: 939–59.Google Scholar
Chen, KT, Ringer, S, Cohen, AP, Lieberman, E. The role of intrapartum fever in identifying asymptomatic term neonates with early-onset neonatal sepsis. J Perinatol 2002; 22: 653–7.Google Scholar
Schrag, SJ, Zell, ER, Lynfield, R, et al. A population-based comparison of strategies to prevent early-onset group B streptococcal disease in neonates. N Engl J Med 2002; 347: 233–9.Google Scholar
Towers, CV, Suriano, K, Asrat, T. The capture rate of at-risk term newborns for early-onset group B streptococcal sepsis determined by a risk factor approach. Am J Obstet Gynecol 1999; 181: 1243–9.Google Scholar
Yancey, MK, Duff, P, Kubilis, P, et al. Risk factors for neonatal sepsis. Obstet Gynecol 1996; 87: 188–94.Google Scholar
Glasgow, TS, Young, PC, Wallin, J, et al. Association of intrapartum antibiotic exposure and late-onset serious bacterial infections in infants. Pediatrics 2005; 116: 696702.Google Scholar
Taukobong, HF, Kincaid, MM, Levy, JK, et al. Does addressing gender inequalities and empowering women and girls improve health and development programme outcomes? Health Policy Plan 2016; 10: 14921514.Google Scholar
Lawn, JE, Blencowe, H, Oza, S, et al. Every newborn: progress, priorities, and potential beyond survival. Lancet 2014; 384: 189205.Google Scholar
Darmstadt, GL, Syed, U, Patel, Z, Kabir, N. Review of domiciliary newborn-care practices in Bangladesh. J Health Popul Nutr 2006; 24: 380–93.Google Scholar
Kumar, V, Kumar, A, Ghosh, AK, et al. Enculturating science: community-centric design of behavior change interactions for accelerating health impact. Semin Perinatol 2015; 39: 393415.Google Scholar
Kumar, V, Mohanty, S, Kumar, A, et al. Effect of community-based behaviour change management on neonatal mortality in Shivgarh, Uttar Pradesh, India: a cluster-randomised controlled trial. Lancet 2008; 372: 1151–62.Google Scholar
Mullany, LC, Darmstadt, GL, Katz, J, et al. Risk factors for umbilical cord infection among newborns of southern Nepal. Am J Epidemiol 2007; 165: 203–11.Google Scholar
Mullany, LC, Katz, J, Li, YM, et al. Breast-feeding patterns, time to initiation, and mortality risk among newborns in southern Nepal. J Nutrition 2008; 138: 599603.Google Scholar
Winch, PJ, Alam, MA, Akther, A, et al. Local understandings of vulnerability and protection during the neonatal period in Sylhet district, Bangladesh: a qualitative study. Lancet 2005; 366: 478–85.Google Scholar
Weston, EJ, Pondo, T, Lewis, MM, et al. The burden of invasive early-onset neonatal sepsis in the United States, 2005–2008. Pediatr Infect Dis J 2011; 30: 937.Google Scholar
Stoll, BJ, Hansen, NI, Sánchez, PJ, et al. Early onset neonatal sepsis: the burden of group B streptococcal and E. coli disease continues. Pediatrics 2011; 127:817–26.Google Scholar
Denniston, S, Riordan, FA. Staphylococcus aureus bacteraemia in children and neonates: a 10 year retrospective review. J Infect 2006; 53: 387–93.Google Scholar
Hakim, H, Mylotte, JM, Faden, H. Morbidity and mortality of staphylococcal bacteremia in children. Am J Infect Control 2007; 35: 102–5.Google Scholar
Verani, JR, McGee, L, Schrag, SJ. Prevention of perinatal group B streptococcal disease: Revised guidelines from CDC, 2010. Department of Health and Human Services, Centers for Disease Control and Prevention, Atlanta, GA, 2010.Google Scholar
Polin, RA, Papile, L-A, Baley, JE, et al. Management of neonates with suspected or proven early-onset bacterial sepsis. Pediatrics 2012; 129: 1006–15.Google Scholar
Hamer, DH, Darmstadt, GL, Carlin, JB, et al. Etiology of bacteremia in young infants in six countries. Pediatr Infect Dis J 2015; 34: e18.Google Scholar
Waters, D, Jawad, I, Ahmad, A, et al. Aetiology of community-acquired neonatal sepsis in low and middle income countries. J Global Health 2011; 1: 154–70.Google Scholar
Schiano, MA, Hauth, JC, Gilstrap, LC 3rd. Second-stage fetal tachycardia and neonatal infection. Am J Obstet Gynecol 1984; 148: 779–81.Google Scholar
Soman, M, Green, B, Daling, J. Risk factors for early neonatal sepsis. Am J Epidemiol 1985; 121: 712–19.Google Scholar
Escobar, GJ, Li, DK, Armstrong, MA, et al. Neonatal sepsis workups in infants weighing less than 2,000 grams at birth: a population-based study. Pediatrics 2000; 106: 256–63.Google Scholar
Baker, CJ. Group B streptococcal infections. Clin Perinatol 1997; 24: 5970.Google Scholar
Ottolini, MC, Lundgren, K, Mirkinson, LJ, et al. Utility of complete blood count and blood culture screening to diagnose neonatal sepsis in the asymptomatic at risk newborn. Pediatr Infect Dis J 2003; 22: 430–4.Google Scholar
Wortham, JM, Hansen, NI, Schrag, SJ, et al. Chorioamnionitis and culture-confirmed, early-onset neonatal infections. Pediatrics 2016; 137(1): e20152323.Google Scholar
World Health Organization (WHO). Pocket Book of Hospital Care for Children: Guidelines for the Management of Common Illnesses with Limited Resources: Geneva: WHO, 2005.Google Scholar
UNICEF, Organization WH. Caring for Newborns and Children in the Community: A Training Course for Community Health Workers. Caring for the Newborn at Home. New York: United Nations, 2015.Google Scholar
Group YICSS. Clinical signs that predict severe illness in children under age 2 months: a multicentre study. Lancet 2008; 371: 135–42.Google Scholar
Bomela, HN, Ballot, DE, Cory, BJ, Cooper, PA. Use of C-reactive protein to guide duration of empiric antibiotic therapy in suspected early neonatal sepsis. Pediatr Infect Dis J 2000; 19: 531–5.Google Scholar
Townsend, TR, Shapiro, M, Rosner, B, Kass, EH. Use of antimicrobial drugs in general hospitals. IV. Infants and children. Pediatrics 1979; 64: 573–8.Google Scholar
Paerregaard, A, Bruun, B, Andersen, GE, Witt, J. No advantage of capillary blood compared with venous blood for culture in neonates. Pediatr Infect Dis J 1989; 8: 659–60.Google Scholar
Wiswell, TE, Hachey, WE. Multiple site blood cultures in the initial evaluation for neonatal sepsis during the first week of life. Pediatr Infect Dis J 1991; 10: 365–9.Google Scholar
Sarkar, S, Bhagat, I, DeCristofaro, J, et al. A study of the role of multiple site blood cultures in the evaluation of neonatal sepsis. J Perinatol 2006; 26: 1822.Google Scholar
Yaacobi, N, Bar-Meir, M, Shchors, I, Bromiker, R. A prospective controlled trial of the optimal volume for neonatal blood cultures. Pediatr Infect Dis J 2015; 34: 351–4.Google Scholar
Jardine, L, Davies, MW, Faoagali, J. Incubation time required for neonatal blood cultures to become positive. J Paediatr Child Health 2006; 42: 797802.Google Scholar
Kumar, Y, Qunibi, M, Neal, T, Yoxall, C. Time to positivity of neonatal blood cultures. Arch Dis Child Fetal Neonat Ed 2001; 85: F182–6.Google Scholar
Liu, C, Ai, H, Wang, W, et al. Comparison of 16S rRNA gene PCR and blood culture for diagnosis of neonatal sepsis. Arch Pediatr 2014; 21: 162–9.Google Scholar
Visser, VE, Hall, RT. Urine culture in the evaluation of suspected neonatal sepsis. J Pediatr 1979; 94: 635–8.Google Scholar
DiGeronimo, RJ. Lack of efficacy of the urine culture as part of the initial workup of suspected neonatal sepsis. Pediatr Infect Dis J 1992; 11: 764–6.Google Scholar
Evans, ME, Schaffner, W, Federspiel, CF, et al. Sensitivity, specificity, and predictive value of body surface cultures in a neonatal intensive care unit. JAMA 1988; 259: 248–52.Google Scholar
Kite, P, Millar, MR, Gorham, P, Congdon, P. Comparison of five tests used in diagnosis of neonatal bacteraemia. Arch Dis Child 1988; 63: 639–43.Google Scholar
Schouten-Van Meeteren, NY, Rietveld, A, Moolenaar, AJ, Van Bel, F. Influence of perinatal conditions on C-reactive protein production. J Pediatr 1992; 120: 621–4.Google Scholar
Benitz, WE, Han, MY, Madan, A, Ramachandra, P. Serial serum C-reactive protein levels in the diagnosis of neonatal infection. Pediatrics 1998; 102: E41.Google Scholar
Jaye, DL, Waites, KB. Clinical applications of C-reactive protein in pediatrics. Pediatr Infect Dis J 1997; 16: 735–46; quiz 746–7.Google Scholar
Chiesa, C, Signore, F, Assumma, M, et al. Serial measurements of C-reactive protein and interleukin-6 in the immediate postnatal period: reference intervals and analysis of maternal and perinatal confounders. Clin Chem 2001; 47: 1016–22.Google Scholar
Laborada, G, Rego, M, Jain, A, et al. Diagnostic value of cytokines and C-reactive protein in the first 24 hours of neonatal sepsis. Am J Perinatol 2003; 20: 491501.Google Scholar
Hofer, N, Zacharias, E, Müller, W, Resch, B. An update on the use of C-reactive protein in early-onset neonatal sepsis: current insights and new tasks. Neonatology 2012; 102: 2536.Google Scholar
Bhandari, V, Wang, C, Rinder, C, Rinder, H. Hematologic profile of sepsis in neonates: neutrophil CD64 as a diagnostic marker. Pediatrics 2008; 121: 129–34.Google Scholar
Kocabas, E, Sarikcioglu, A, Aksaray, N, et al. Role of procalcitonin, C-reactive protein, interleukin-6, interleukin-8 and tumor necrosis factor-alpha in the diagnosis of neonatal sepsis. Turk J Pediatr 2007; 49: 720.Google Scholar
Ng, PC, Li, G, Chui, KM, et al. Neutrophil CD64 is a sensitive diagnostic marker for early-onset neonatal infection. Pediatr Res 2004; 56: 796803.Google Scholar
Santana Reyes, C, Garcia-Munoz, F, Reyes, D, et al. Role of cytokines (interleukin-1β, 6, 8, tumour necrosis factor-α, and soluble receptor of interleukin-2) and C-reactive protein in the diagnosis of neonatal sepsis. Acta Paediatr 2003; 92: 221–7.Google Scholar
Franz, AR, Steinbach, G, Kron, M, Pohlandt, F. Reduction of unnecessary antibiotic therapy in newborn infants using interleukin-8 and C-reactive protein as markers of bacterial infections. Pediatrics 1999; 104: 447–53.Google Scholar
Mohsen, AHA, Kamel, BA. Predictive values for procalcitonin in the diagnosis of neonatal sepsis. Electron Phys 2015; 7: 1190.Google Scholar
Sakha, K, Husseini, M, Seyyedsadri, N. The role of the procalcitonin in diagnosis of neonatal sepsis and correlation between procalcitonin and C-reactive protein in these patients. Pakistan J Biol Sci: PJBS 2008; 11: 1785–90.Google Scholar
Nupponen, I, Andersson, S, Jarvenpaa, AL, et al. Neutrophil cd11b expression and circulating interleukin-8 as diagnostic markers for early-onset neonatal sepsis. Pediatrics 2001; 108: E12.Google Scholar
Weirich, E, Rabin, RL, Maldonado, Y, et al. Neutrophil CD11b expression as a diagnostic marker for early-onset neonatal infection. J Pediatr 1998; 132: 445–1.Google Scholar
Edgar, JD, Wilson, DC, McMillan, SA, et al. Predictive value of soluble immunological mediators in neonatal infection. Clin Sci (Colch) 1994; 87: 165–71.Google Scholar
Kennon, C, Overturf, G, Bessman, S, et al. Granulocyte colony-stimulating factor as a marker for bacterial infection in neonates. J Pediatr 1996; 128: 765–9.Google Scholar
Backes, RJ, Rouse, MS, Henry, NK, et al. Activity of penicillin combined with an aminoglycoside against group B streptococci in vitro and in experimental endocarditis. J Antimicrob Chemother 1986; 18: 491–8.Google Scholar
Starr, SE. Antimicrobial therapy of bacterial sepsis in the newborn infant. J Pediatr 1985; 106: 1043–8.Google Scholar
Jenson, HB, Pollock, BH. Meta-analyses of the effectiveness of intravenous immune globulin for prevention and treatment of neonatal sepsis. Pediatrics 1997; 99: E2.Google Scholar
Manroe, BL, Rosenfeld, CR, Weinberg, AG, Browne, R. The differential leukocyte count in the assessment and outcome of early-onset neonatal group B streptococcal disease. J Pediatr 1977; 91: 632–7.Google Scholar
Bilgin, K, Yaramis, A, Haspolat, K, et al. A randomized trial of granulocyte-macrophage colony-stimulating factor in neonates with sepsis and neutropenia. Pediatrics 2001; 107: 3641.Google Scholar
Gillan, ER, Christensen, RD, Suen, Y, et al. A randomized, placebo-controlled trial of recombinant human granulocyte colony-stimulating factor administration in newborn infants with presumed sepsis: significant induction of peripheral and bone marrow neutrophilia. Blood 1994; 84: 1427–33.Google Scholar
Schibler, KR, Osborne, KA, Leung, LY, et al. A randomized, placebo-controlled trial of granulocyte colony-stimulating factor administration to newborn infants with neutropenia and clinical signs of early-onset sepsis. Pediatrics 1998; 102: 613.Google Scholar
Tshefu, A, Lokangaka, A, Ngaima, S, et al. Simplified antibiotic regimens compared with injectable procaine benzylpenicillin plus gentamicin for treatment of neonates and young infants with clinical signs of possible serious bacterial infection when referral is not possible: a randomised, open-label, equivalence trial. Lancet 2015; 385: 1767–76.Google Scholar
Bennet, R, Eriksson, M, Zetterstrom, R. Neonatal septicemia: comparison of onset and risk factors during three consecutive 5-year periods. Acta Paediatr Scand 1987; 76: 361–2.Google Scholar
Mok, PM, Reilly, BJ, Ash, JM. Osteomyelitis in the neonate: clinical aspects and the role of radiography and scintigraphy in diagnosis and management. Radiology 1982; 145: 677–82.Google Scholar
Bennet, R, Bergdahl, S, Eriksson, M, Zetterstrom, R. The outcome of neonatal septicemia during fifteen years. Acta Paediatr Scand 1989; 78: 40–3.Google Scholar
Alfven, G, Bergqvist, G, Bolme, P, Eriksson, M. Longterm follow-up of neonatal septicemia. Acta Paediatr Scand 1978; 67: 769–73.Google Scholar
Sehdev, HM, Stamilio, DM, Macones, GA, et al. Predictive factors for neonatal morbidity in neonates with an umbilical arterial cord pH less than 7.00. Am J Obstet Gynecol 1997; 177: 1030–4.Google Scholar
Wu, YW. Systematic review of chorioamnionitis and cerebral palsy. Ment Retard Dev Disabil Res Rev 2002; 8: 25–9.Google Scholar
Arifeen, SE, Mullany, LC, Shah, R, et al. The effect of cord cleansing with chlorhexidine on neonatal mortality in rural Bangladesh: a community-based, cluster-randomised trial. Lancet 2012; 379: 1022–8.Google Scholar
Imdad, A, Mullany, LC, Baqui, AH, et al. The effect of umbilical cord cleansing with chlorhexidine on omphalitis and neonatal mortality in community settings in developing countries: a meta-analysis. BMC Public Health 2013; 13: S15.Google Scholar
Mullany, LC, Darmstadt, GL, Khatry, SK, et al. Topical applications of chlorhexidine to the umbilical cord for prevention of omphalitis and neonatal mortality in southern Nepal: a community-based, cluster-randomised trial. Lancet 2006; 367: 910–18.Google Scholar
World Health Organization (WHO). WHO Model List of Essential Medicines, 19th list (updated). Geneva: WHO, April 2015.Google Scholar
Darmstadt, GL, Badrawi, N, Law, PA, et al. Topically applied sunflower seed oil prevents invasive bacterial infections in preterm infants in Egypt: a randomized, controlled clinical trial. Pediatr Infect Dis J 2004; 23: 719–25.Google Scholar
Darmstadt, GL, Saha, SK, Ahmed, AS, et al. Effect of topical treatment with skin barrier-enhancing emollients on nosocomial infections in preterm infants in Bangladesh: a randomised controlled trial. Lancet 2005; 365: 1039–45.Google Scholar
Salam, RA, Darmstadt, GL, Bhutta, ZA. Effect of emollient therapy on clinical outcomes in preterm neonates in Pakistan: a randomised controlled trial. Arch Dis Child Fetal Neonat Ed 2015; 100: F210–15.Google Scholar
Salam, RA, Das, JK, Darmstadt, GL, Bhutta, ZA. Emollient therapy for preterm newborn infants: evidence from the developing world. BMC Public Health 2013; 13: 17.Google Scholar
Conde-Agudelo, A, Díaz-Rossello, J. Kangaroo mother care to reduce morbidity and mortality in low birth weight infants. Cochrane Database Syst Rev 2014; 4: CD002771.Google Scholar
Engmann, C, Wall, S, Darmstadt, G, et al. Consensus on kangaroo mother care acceleration. Lancet 2013; 382: e26–7.Google Scholar
Schuchat, A. Group B streptococcal disease: from trials and tribulations to triumph and trepidation. Clin Infect Dis 2001; 33: 751–6.Google Scholar
Schuchat, A. Epidemiology of group B streptococcal disease in the United States: shifting paradigms. Clin Microbiol Rev 1998; 11: 497513.Google Scholar
Davies, HD, Adair, C, McGeer, A, et al. Antibodies to capsular polysaccharides of group B Streptococcus in pregnant Canadian women: relationship to colonization status and infection in the neonate. J Infect Dis 2001; 184: 285–91.Google Scholar
Blumberg, HM, Stephens, DS, Modansky, M, et al. Invasive group B streptococcal disease: the emergence of serotype V. J Infect Dis 1996; 173: 365–73.Google Scholar
Klegerman, ME, Boyer, KM, Papierniak, CK, Gotoff, SP. Estimation of the protective level of human IgG antibody to the type-specific polysaccharide of group B Streptococcus type Ia. J Infect Dis 1983; 148: 648–55.Google Scholar
Schuchat, A, Deaver-Robinson, K, Plikaytis, BD, et al. Multistate case-control study of maternal risk factors for neonatal group B streptococcal disease. The Active Surveillance Study Group. Pediatr Infect Dis J 1994; 13: 623–9.Google Scholar
Boyer, KM, Gotoff, SP. Prevention of early-onset neonatal group B streptococcal disease with selective intrapartum chemoprophylaxis. N Engl J Med 1986; 314: 1665–9.Google Scholar
CDC. Decreasing incidence of perinatal group B streptococcal disease – United States, 1993–1995. MMWR Morb Mortal Wkly Rep 1997; 46: 473.Google Scholar
Whitney, CG, Plikaytis, BD, Gozansky, WS, et al. Prevention practices for perinatal group B streptococcal disease: a multi-state surveillance analysis. Neonatal Group B Streptococcal Disease Study Group. Obstet Gynecol 1997; 89: 2832.Google Scholar
Schrag, S, Gorwitz, R, Fultz-Butts, K, Schuchat, A. Prevention of perinatal group B streptococcal disease. MMWR Recomm Rep 2002; 51: 122.Google Scholar
Centers for Disease Control and Prevention (CDC). Prevention of perinatal group B streptococcal disease: a public health perspective. Centers for Disease Control and Prevention. MMWR Morb Mortal Wkly Rep 1996; 45: 124.Google Scholar
CDC. Diminishing racial disparities in early-onset neonatal group B streptococcal disease – United States, 2000–2003. MMWR Morb Mortal Wkly Rep 2004; 53: 502.Google Scholar
CDC. Perinatal group B streptococcal disease after universal screening recommendations–United States, 2003–2005. MMWR Morb Mortal Wkly Rep 2007; 56: 701–5.Google Scholar
CDC. Active bacterial core surveillance report: Streptococcus pneumoniae, 2009.Google Scholar
Ekelund, K, Konradsen, H. Invasive group B streptococcal disease in infants: a 19-year nationwide study. Serotype distribution, incidence and recurrent infection. Epidemiol Infect 2004; 132: 1083–90.Google Scholar
Phares, CR, Lynfield, R, Farley, MM, et al. Epidemiology of invasive group B streptococcal disease in the United States, 1999–2005. JAMA 2008; 299: 2056–65.Google Scholar
Katz, V, Bowes, WA Jr. Perinatal group B streptococcal infections across intact amniotic membranes. J Reprod Med 1988; 33: 445–9.Google Scholar
Dillon, HC Jr, Khare, S, Gray, BM. Group B streptococcal carriage and disease: a 6-year prospective study. J Pediatr 1987; 110: 31–6.Google Scholar
Trager, JD, Martin, JM, Barbadora, K, et al. Probable community acquisition of group B Streptococcus in an infant with late-onset disease: demonstration using field inversion gel electrophoresis. Arch Pediatr Adolesc Med 1996; 150: 766–8.Google Scholar
Ancona, RJ, Ferrieri, P, Williams, PP. Maternal factors that enhance the acquisition of group-B streptococci by newborn infants. J Med Microbiol 1980; 13: 273–80.Google Scholar
Regan, JA, Klebanoff, MA, Nugent, RP. The epidemiology of group B streptococcal colonization in pregnancy. Vaginal Infections and Prematurity Study Group. Obstet Gynecol 1991; 77: 604–10.Google Scholar
Curtis, J, Kim, G, Wehr, NB, Levine, RL. Group B streptococcal phospholipid causes pulmonary hypertension. Proc Natl Acad Sci USA 2003; 100: 5087–90.Google Scholar
CDC. Hospital-based policies for prevention perinatal group B streptococcal disease – United States, 1999. MMWR Morbid Mortal Wkly Rep 2000; 49: 936–40.Google Scholar
Bromberger, P, Lawrence, JM, Braun, D, et al. The influence of intrapartum antibiotics on the clinical spectrum of early-onset group B streptococcal infection in term infants. Pediatrics 2000; 106: 244–50.Google Scholar
Pulver, L, Hopfenbeck, M, Young, P, et al. Continued early onset group B streptococcal infections in the era of intrapartum prophylaxis. J Perinatol 2009; 29: 20–5.Google Scholar
Puopolo, KM, Madoff, LC, Eichenwald, EC. Early-onset group B streptococcal disease in the era of maternal screening. Pediatrics 2005; 115: 1240–6.Google Scholar
Maayan-Metzger, A, Barzilai, A, Keller, N, Kuint, J. Are the “good old” antibiotics still appropriate for early-onset neonatal sepsis? A 10 year survey. Israel Med Assoc J 2009; 11: 138.Google Scholar
Glass, P, Wagner, AE, Papero, PH, et al. Neurodevelopmental status at age five years of neonates treated with extracorporeal membrane oxygenation. J Pediatr 1995; 127: 447–57.Google Scholar
American Academy of Pediatrics Committee on Infectious Diseases and Committee on Fetus and Newborn. Revised guidelines for prevention of early-onset group B streptococcal (GBS) infection. Pediatrics 1997; 99: 489–96.Google Scholar
American College of Obstetricians and Gynecologists. Prevention of Early-Onset Group B Streptococcal Disease in Newborns [Opinion 173]. Washington, DC: American College of Obstetricians and Gynecologists, 1996.Google Scholar
Rosenstein, NE, Schuchat, A. Opportunities for prevention of perinatal group B streptococcal disease: a multistate surveillance analysis. The Neonatal Group B Streptococcal Disease Study Group. Obstet Gynecol 1997; 90: 901–6.Google Scholar
Cueto, M, Sanchez, M, Sanpedro, A. Timing of intrapartum ampicillin and prevention of vertical transmission of group B streptococci. Obstet Gynecol 1998; 91: 112–14.Google Scholar
Hall, C, Easton, J, Granoff, D, et al. Guidelines for prevention of group B streptococcal (GBS) infection by chemoprophylaxis. Pediatrics 1992; 90: 775–8.Google Scholar
Gotoff, SP, Boyer, KM. Prevention of early-onset neonatal group B streptococcal disease. Pediatrics 1997; 99: 866–9.Google Scholar
Siegel, JD, Cushion, NB. Prevention of early-onset group B streptococcal disease: another look at single-dose penicillin at birth. Obstet Gynecol 1996; 87: 692–8.Google Scholar
Benitz, WE. The neonatal group B streptococcal debate. Pediatrics 1998; 101: 494–6.Google Scholar
Lieu, TA, Mohle-Boetani, JC, Ray, GT, et al. Neonatal group B streptococcal infection in a managed care population. Perinatal Group B Streptococcal Infection Study Group. Obstet Gynecol 1998; 92: 21–7.Google Scholar
Yow, MD, Mason, EO, Leeds, LJ, et al. Ampicillin prevents intrapartum transmission of group B Streptococcus. JAMA 1979; 241: 1245–7.Google Scholar
Heim, K, Alge, A, Marth, C. Anaphylactic reaction to ampicillin and severe complication in the fetus. Lancet 1991; 337: 859–60.Google Scholar
Towers, CV, Carr, MH, Padilla, G, Asrat, T. Potential consequences of widespread antepartal use of ampicillin. Am J Obstet Gynecol 1998; 179: 879–83.Google Scholar
Pylipow, M, Gaddis, M, Kinney, JS. Selective intrapartum prophylaxis for group B Streptococcus colonization: management and outcome of newborns. Pediatrics 1994; 93: 631–5.Google Scholar
Davis, RL, Hasselquist, MB, Cardenas, V, et al. Introduction of the new Centers for Disease Control and Prevention group B streptococcal prevention guideline at a large West Coast health maintenance organization. Am J Obstet Gynecol 2001; 184: 603–10.Google Scholar
Jaureguy, F, Carton, M, Panel, P, et al. Effects of intrapartum penicillin prophylaxis on intestinal bacterial colonization in infants. J Clin Microbiol 2004; 42: 5184–8.Google Scholar
Borchardt, SM, DeBusscher, JH, Tallman, PA, et al. Frequency of antimicrobial resistance among invasive and colonizing group B streptococcal isolates. BMC Infect Dis 2006; 6: 1.Google Scholar
Castor, ML, Whitney, CG, Como-Sabetti, K, et al. Antibiotic resistance patterns in invasive group B streptococcal isolates. Infect Dis Obstet Gynecol 2008; 2008: 727505.Google Scholar
Kasper, DL, Paoletti, LC, Wessels, MR, et al. Immune response to type III group B streptococcal polysaccharide-tetanus toxoid conjugate vaccine. J Clin Invest 1996; 98: 2308–14.Google Scholar
Baker, CJ, Paoletti, LC, Rench, MA, et al. Immune response of healthy women to two different group B streptococcal type V capsular polysaccharide-protein conjugate vaccines. J Infect Dis 2004; 189: 1103–12.Google Scholar
Guttormsen, H-K, Liu, Y, Paoletti, LC. Functional activity of antisera to group B streptococcal conjugate vaccines measured with an opsonophagocytosis assay and HL-60 effector cells. Human Vaccines 2008; 4: 370–4.Google Scholar
Buccato, S, Maione, D, Rinaudo, CD, et al. Use of Lactococcus lactis expressing pili from group B Streptococcus as a broad-coverage vaccine against streptococcal disease. J Infect Dis 2006; 194: 331–40.Google Scholar
Rosini, R, Rinaudo, CD, Soriani, M, et al. Identification of novel genomic islands coding for antigenic pilus‐like structures in Streptococcus agalactiae. Mol Microbiol 2006; 61: 126–41.Google Scholar
Hillier, S, Ferris, D, Fine, D, Ferrieri, P. Women receiving group B Streptococcus serotype III tetanus toxoid (GBS III-TT) vaccine have reduced vaginal and rectal acquisition of GBS type III. Presented at the 47th Annual Meeting of the IDSA, 2009.Google Scholar
Le Doare, K, Heath, PT. An overview of global GBS epidemiology. Vaccine 2013; 31(Suppl 4): D712.Google Scholar
Edmond, KM, Kortsalioudaki, C, Scott, S, et al. Group B streptococcal disease in infants aged younger than 3 months: systematic review and meta-analysis. Lancet 2012; 379: 547–56.Google Scholar
Bingen, E, Picard, B, Brahimi, N, et al. Phylogenetic analysis of Escherichia coli strains causing neonatal meningitis suggests horizontal gene transfer from a predominant pool of highly virulent B2 group strains. J Infect Dis 1998; 177: 642–50.Google Scholar
Sarff, LD, McCracken, GH, Schiffer, MS, et al. Epidemiology of Escherichia coli K1 in healthy and diseased newborns. Lancet 1975; 1: 1099–104.Google Scholar
McCracken, GH Jr, Sarff, LD, Glode, MP, et al. Relation between Escherichia coli K1 capsular polysaccharide antigen and clinical outcome in neonatal meningitis. Lancet 1974; 2: 246–50.Google Scholar
Guerina, NG, Kessler, TW, Guerina, VJ, et al. The role of pili and capsule in the pathogenesis of neonatal infection with Escherichia coli K1. J Infect Dis 1983; 148: 395405.Google Scholar
Mayor-Lynn, K, Gonzalez-Quintero, VH, O’Sullivan, MJ, et al. Comparison of early-onset neonatal sepsis caused by Escherichia coli and group B Streptococcus. Am J Obstet Gynecol 2005; 192: 1437–9.Google Scholar
Alarcon, A, Pena, P, Salas, S, et al. Neonatal early onset Escherichia coli sepsis: trends in incidence and antimicrobial resistance in the era of intrapartum antimicrobial prophylaxis. Pediatr Infect Dis J 2004; 23: 295–9.Google Scholar
Charles, D, Larsen, B. Streptococcal puerperal sepsis and obstetric infections: a historical perspective. Rev Infect Dis 1986; 8: 411–22.Google Scholar
Chuang, I, Van Beneden, C, Beall, B, et al. Population-based surveillance for postpartum invasive group A Streptococcus infections, 1995–2000. Clin Infect Dis 2002; 35: 665–70.Google Scholar
Miyairi, I, Berlingieri, D, Protic, J, Belko, J. Neonatal invasive group A streptococcal disease: case report and review of the literature. Pediatr Infect Dis J 2004; 23: 161–5.Google Scholar
Greenberg, D, Leibovitz, E, Shinnwell, ES, et al. Neonatal sepsis caused by Streptococcus pyogenes: resurgence of an old etiology? Pediatr Infect Dis J 1999; 18: 479–81.Google Scholar
Macris, MH, Hartman, N, Murray, B, et al. Studies of the continuing susceptibility of group A streptococcal strains to penicillin during eight decades. Pediatr Infect Dis J 1998; 17: 377–81.Google Scholar
Kaplan, EL, Johnson, DR, Del Rosario, MC, Horn, DL. Susceptibility of group A beta-hemolytic streptococci to thirteen antibiotics: examination of 301 strains isolated in the United States between 1994 and 1997. Pediatr Infect Dis J 1999; 18: 1069–72.Google Scholar
Norrby-Teglund, A, Ihendyane, N, Kansal, R, et al. Relative neutralizing activity in polyspecific IgM, IgA, and IgG preparations against group A streptococcal superantigens. Clin Infect Dis 2000; 31: 1175–82.Google Scholar
Zimbelman, J, Palmer, A, Todd, J. Improved outcome of clindamycin compared with beta-lactam antibiotic treatment for invasive Streptococcus pyogenes infection. Pediatr Infect Dis J 1999; 18: 1096–100.Google Scholar
Dobson, SR, Baker, CJ. Enterococcal sepsis in neonates: features by age at onset and occurrence of focal infection. Pediatrics 1990; 85: 165–71.Google Scholar
McNeeley, DF, Saint-Louis, F, Noel, GJ. Neonatal enterococcal bacteremia: an increasingly frequent event with potentially untreatable pathogens. Pediatr Infect Dis J 1996; 15: 800–5.Google Scholar
Murray, BE. The life and times of the Enterococcus. Clin Microbiol Rev 1990; 3: 4665.Google Scholar

References

Klein, JO, Feigin, RD, McCracken, GH. Report on the task force on diagnosis and management of meningitis. Pediatrics 1986; 78: 959–82.Google Scholar
Philip, AGS. Neonatal Sepsis and Meningitis. Boston: G.K. Hall, 1985: Appendix.Google Scholar
Philip, AGS. The changing face of neonatal infection: experience at a regional medical center. Pediatr Infect Dis J 1994; 13: 1098–102.Google Scholar
Greenberg, D, Shinwell, ES, Yagupsky, P, et al. A prospective study of neonatal sepsis and meningitis in southern Israel. Pediatr Infect Dis J 1997; 16: 768–73.Google Scholar
Moreno, MT, Vargas, S, Poveda, R, et al. Neonatal sepsis and meningitis in a developing Latin American country. Pediatr Infect Dis J 1994; 13: 516–20.Google Scholar
Laving, AM, Musoke, RN, Wasunno, AO, et al. Neonatal bacterial meningitis at the newborn unit of Kenyatta National Hospital. East Afr Med J 2003; 80: 456–62.Google Scholar
Trijbels-Smeulders, M, de Jonge, GA, Pasker-de Jonge, PC, et al. Epidemiology of neonatal group B streptococcal disease in the Netherlands before and after introduction of guidelines for prevention. Arch Dis Child Fetal Neonatal Ed 2007; 92: F271–6.Google Scholar
Mulder, CJJ, Zanen, HC. A study of 280 cases of neonatal meningitis in the Netherlands. J Infect 1984; 9: 177–84.Google Scholar
De Louvois, J, Halket, S, Harvey, D. Neonatal meningitis in England and Wales: sequelae at 5 years of age. Eur J Pediatr 2005; 164: 730–4.Google Scholar
Isaacs, D, Royle, JA. Intrapartum antibiotics and early onset neonatal sepsis caused by group B Streptococcus and by other organisms in Australia. Australasian Study Group for Neonatal Infections. Pediatr Infect Dis J 1999; 18: 524–8.Google Scholar
Francis, BM, Gilbert, GL. Survey of neonatal meningitis in Australia:1987–1989. Med J Aust 1992; 156: 240–3.Google Scholar
Gordon, A, Isaacs, D. Late onset neonatal gram-negative bacillary infection in Australia and New Zealand, 1992–2002. Pediatr Infect Dis J 2006; 25: 25–9.Google Scholar
Schrag, SJ, Stoll, BJ. Early-onset neonatal sepsis in the era of widespread intrapartum chemoprophylaxis. Pediatr Infect Dis J 2006; 25: 939–40.Google Scholar
May, M, Daley, AJ, Donarth, S, et al. Early onset neonatal meningitis in Australia and New Zealand, 1992–2002. Arch Dis Child Fetal Neonatal Ed 2005; 90: F324–7.Google Scholar
Okike, IO, Johnson, AP, Henderson, KL, et al. Incidence, etiology and outcome of bacterial meningitis in infants aged <90 days in the United Kingdom and the Republic of Ireland: prospective, enhanced national population-based surveillance. Clin Infect Dis 2014; 59: e150–7.Google Scholar
Darlow, BA, Voss, L, Lennon, DR, Grimwood, K. Early-onset neonatal group B Streptococcus sepsis following national risk-based prevention guidelines. Austr NZ Obstet Gynaecol 2016; 56: 6074.Google Scholar
Longe, AC, Omene, JA, Okolo, AA. Neonatal meningitis in Nigerian infants. Acta Paediatr Scand 1984; 73: 477–81.Google Scholar
Tessin, I, Trollfors, B, Thiringer, K. Incidence and etiology of neonatal septicemia and meningitis in western Sweden 1975–1986. Acta Paediatr Scand 1990; 79: 1023–30.Google Scholar
Hristeva, L, Booy, R, Bowler, I, et al. Prospective surveillance of neonatal meningitis. Arch Dis Child 1993; 69: 1418.Google Scholar
Shiva, F, Mosaffa, N, Khabbaz, R, et al. Lumbar puncture in neonates under and over 72 hours of age. J Coll Phys Surg Pak 2006; 16: 525–8.Google Scholar
Weiss, MG, Ionides, SP, Anderson, CL. Meningitis in premature infants with respiratory distress: role of admission lumbar puncture. J Pediatr 1991; 119: 973–5.Google Scholar
Sanghvi, KP, Tudehope, DI. Neonatal bacterial sepsis in a neonatal intensive care unit: a 5 year analysis. J Paediatr Child Health 1996; 32: 333–8.Google Scholar
Johnson, CE, Whitwell, JK, Pethe, K, et al. Term newborns who are at risk for sepsis: are lumbar punctures necessary? Pediatrics 1997; 99: E10.Google Scholar
Pong, A, Bradley, JS. Bacterial meningitis and the newborn infant. Infect Dis Clin North Am 1999; 13: 711–33.Google Scholar
Albanyan, EA, Baker, CJ. Is lumbar puncture necessary to exclude meningitis in neonates and young infants: lessons from the group B Streptococcus cellulitis-adenitis syndrome. Pediatrics 1998; 102: 985–6.Google Scholar
Flidel-Rimon, O, Leibovitz, E, Eventor Friedman, S, et al: Is lumbar puncture (LP) required in every work-up for suspected late-onset sepsis in neonates? Acta Paediatr 2011; 100: 303–4.Google Scholar
Stoll, BJ, Hansen, N, Fanaroff, AA, et al. To tap or not to tap:high likelihood of meningitis without sepsis among very low birth weight infants. Pediatrics 2004; 113: 1181–6.Google Scholar
Franco, SM, Cornelius, VE, Andrews, BF. Long-term outcome of neonatal meningitis. Am J Dis Child 1992; 146: 567–71.Google Scholar
Unhanand, M, Mustafa, MM, McCracken, GH Jr, et al. Gram-negative enteric bacillary meningitis: a twenty-one-year experience. J Pediatr 1993; 122: 1521.Google Scholar
Edwards, MS, Rench, MA, Haffar, AAM, et al. Long-term sequelae of group B streptococcal meningitis in infants. J Pediatr 1985; 106: 717–22.Google Scholar
Klinger, G, Chin, CN, Beyene, J, et al. Predicting the outcome of neonatal bacterial meningitis. Pediatrics 2000; 106: 477–82.Google Scholar
Dellagrammaticas, HD, Christodoulou, C, Megaloyanni, E, et al. Treatment of gram-negative bacterial meningitis in term neonates with third generation cephalosporins plus amikacin. Biol Neonate 2000; 77: 139–46.Google Scholar
Harvey, D, Holt, DE, Bedford, H. Bacterial meningitis in the newborn: a prospective study of mortality and morbidity. Semin Perinatol 1999; 23: 218–25.Google Scholar
Chang, CJ, Chang, WN, Huang, LT, et al. Neonatal bacterial meningitis in southern Taiwan. Pediatr Neurol 2003; 29: 288–94.Google Scholar
Daoud, AS, al-Sheyyab, M, Abu-Ekteish, F, et al. Neonatal meningitis in northern Jordan. J Trop Pediatr 1996; 42: 267–70.Google Scholar
Molyneux, E, Walsh, A, Phiri, A, et al. Acute bacterial meningitis in children admitted to the Queen Elizabeth Central Hospital, Blantyre, Malawi in 1996–97. Trop Med Int Health 1998; 3: 610–18.Google Scholar
Kamoun, F, Dowlut, MB, Ameur, SB, et al. Neonatal purulent meningitis in southern Tunisia: Epidemiology, bacteriology, risk factors and prognosis. Fetal Pediatr Pathol 2015; 34: 233–40.Google Scholar
Gaschignard, J, Levy, O, Olivier, R, et al. Neonatal bacterial meningitis: 444 cases in 7 years. Pediatr Infect Dis J 2011; 30: 212–17.Google Scholar
Thigpen, MC, Whitney, CG, Massonnier, NE, et al. Bacterial meningitis in the United States, 1998–2007. N Engl J Med 2011; 364: 2016–25.Google Scholar
Synnott, MB, Morse, DL, Hall, SM. Neonatal meningitis in England and Wales: a review of routine national data. Arch Dis Child 1994; 71: F7580.Google Scholar
Gebremariam, A. Neonatal meningitis in Addis Ababa: a 10 year review. Ann Trop Paediatr 1998; 18: 279–83.Google Scholar
Berardi, A, Lugli, L, Baronciani, D, et al. Group B streptococcal infections in a northern region of Italy. Pediatrics 2007; 120: e487–93.Google Scholar
Mulder, CJJ, Zanen, HC. Listeria monocytogenes neonatal meningitis in the Netherlands. Eur J Pediatr 1986; 145: 60–2.Google Scholar
Linnan, MJ, Mascola, L, Lou, XD, et al. Epidemic listeriosis associated with Mexican-style cheese. N Engl J Med 1988; 319: 823–8.Google Scholar
Zaki, M, Daoud, AS, al Saleh, Q, et al. Bacterial meningitis in the newborn: a Kuwaiti experience. J Trop Pediatr 1990; 36: 62–5.Google Scholar
Baker, CJ. Group B streptococcal infections in neonates. Pediatr Rev 1979; 1: 515.Google Scholar
Carstensen, H, Henrichsen, J, Jepsen, OB. A national survey of severe group B streptococcal infections in neonates and young infants in Denmark, 1978–83. Acta Paediatr Scand 1985; 74: 934–41.Google Scholar
Joubrel, C, Tazi, A. Six, A, et al. Group B Streptococcus neonatal invasive infections, France 2007–2012. Clin Microbiol Infect 2015; 21: 910–16.Google Scholar
Domelier, AS, van der Mee-Marquet, N, Grandet, A, et al. Loss of catabolic function in Streptococcus agalactiae strains and its association with neonatal meningitis. J Clin Microbiol 2006; 44: 3245–50.Google Scholar
Brandolini, M, Corbella, M, Cambieri, P, et al. Late-onset neonatal group B streptococcal disease associated with breast-milk transmission: molecular typing using RAPD-PCR. Early Hum Dev 2014; 90(Suppl 1): S84–6.Google Scholar
Mulder, CJJ, van Alphen, L, Zanen, HC. Neonatal meningitis caused by Escherichia coli in the Netherlands. J Infect Dis 1984; 150: 935–40.Google Scholar
Basmaci, R, Bonacorsi, S, Bidot, P, et al. Escherichia coli meningitis features in 325 children from 2001 to 2013 in France. Clin Infect Dis 2015; 61: 779–86.Google Scholar
Ignjatovic, M. Bacterial causes of meningitis in newborns [in Serbian]. Srp Arh Calok Lek 2001; 129: 3641.Google Scholar
Davies, PA, Rudd, PT. Neonatal Meningitis (Clinics in Developmental Medicine 132). London: MacKeith Press, 1994.Google Scholar
Arango, CA, Rathore, MH. Neonatal meningococcal meningitis: case reports and review of literature. Pediatr Infect Dis J 1996; 15: 1134–6.Google Scholar
Doctor, BA, Newman, N, Minich, NM, et al. Clinical outcomes of neonatal meningitis in very low-birth-weight infants. Clin Pediatr 2001; 40: 473–80.Google Scholar
Tse, G, Silver, M, Whyte, H, et al. Neonatal meningitis and multiple brain abscesses due to Citrobacter diversus. Pediatr Pathol Lab Med 1997; 17: 977–82.Google Scholar
Goldenberg, RL, Hauth, JC, Andrews, WW. Intrauterine infection and preterm delivery. N Engl J Med 2000; 342: 1500–7.Google Scholar
Edwards, MS, Jackson, CV, Baker, CJ. Increased risk of group B streptococcal disease in twins. JAMA 1981; 245: 2044–6.Google Scholar
Levy, HL, Sepe, SJ, Shih, VE, et al. Sepsis due to Escherichia coli in neonates with galactosemia. N Engl J Med 1977; 297: 823–5.Google Scholar
Bingen, E, Bonacorsi, S, Brahimi, N, et al. Virulence patterns of Escherichia coli K1 strains associated with neonatal meningitis. J Clin Microbiol 1997; 35: 2981–2.Google Scholar
Öhman, L, Tullus, K, Katouli, M, et al. Correlation between susceptibility of infants to infection and interaction with neurotrophils of Escherichia coli strains, causing neonatal and infantile septicemia. J Infect Dis 1995; 171: 128–33.Google Scholar
Berman, P, Banker, B. Neonatal meningitis:a clinical and pathological study of 29 cases. Pediatrics 1966; 38: 624.Google Scholar
Bell, WE, McGuinness, GA. Suppurative central nervous system infections in the neonate. Semin Perinatol 1982; 6: 124.Google Scholar
Polin, RA, Harris, MC. Neonatal bacterial meningitis. Semin Neonatol 2001; 6: 157–72.Google Scholar
Anagnostakis, D, Messaritakis, J, Damianos, D, et al. Blood-brain barrier permeability in “healthy,” infected and stressed neonates. J Pediatr 1992; 121: 291–4.Google Scholar
Temesvari, P, Abraham, CS, Speer, CP, et al. Escherichia coli O111 B4 lipopolysaccharide given intracisternally induces blood-brain barrier opening during experimental neonatal meningitis in piglets. Pediatr Res 1993; 34: 182–6.Google Scholar
Huang, S, Stins, MF, Kim, KS. Bacterial penetration across the blood-brain barrier during the development of neonatal meningitis. Microbes Infect 2000; 2: 1237–44.Google Scholar
Chang Chien, HY, Chiu, NC, Li, WC, et al. Characteristics of neonatal bacterial meningitis in a teaching hospital in Taiwan from 1984–1997. J Microbiol Immunol Infect 2000; 33: 100–4.Google Scholar
Smith, PB, Cotton, CM, Garges, HP, et al. A comparison of neonatal gram-negative rod and gram-positive cocci meningitis. J Perinatol 2006; 26: 111–14.Google Scholar
Garges, HP, Moody, MA, Cotten, CM, et al. Neonatal meningitis:what is the correlation among cerebrospinal fluid cultures, blood cultures and cerebrospinal fluid parameters? Pediatrics 2006; 117: 1094–100.Google Scholar
Sarman, G, Moise, AA, Edwards, MS. Meningeal inflammation in neonatal gram-negative bacteremia. Pediatr Infect Dis J 1995; 14: 701–4.Google Scholar
Porter, FL, Miller, JP, Cole, FS, et al. A controlled clinical trial of local anesthesia for lumbar puncture in newborns. Pediatrics 1991; 88: 663–9.Google Scholar
Halcrow, SJ, Crawford, PJ, Craft, AW. Epidermoid spinal cord tumour after lumbar puncture. Arch Dis Child 1985; 60: 978–9.Google Scholar
McCracken, GH, Mize, SG. A controlled study of intrathecal antibiotic therapy in gram-negative enteric meningitis of infancy: report of the Neonatal Meningitis Co-operative Study Group. J Pediatr 1976; 89: 6672.Google Scholar
Philip, AGS, Baker, CJ. Cerebrospinal fluid C-reactive protein in neonatal meningitis. J Pediatr 1983; 102: 715–17.Google Scholar
Martinez de Tejada, B, Stan, CM, Boulvain, M, et al: Development of rapid PCR assay for screening maternal colonization by group B Streptococcus and neonatal invasive Escherichia coli during labor. Gynecol Obstet Invest 2010; 70: 250–5.Google Scholar
Philip, AGS. Response of C-reactive protein in neonatal group B streptococcal infection. Pediatr Infect Dis J 1985; 4: 145–8.Google Scholar
Sabel, KG, Hanson, LA. The clinical usefulness of C-reactive protein (CRP) determinations in bacterial meningitis and septicemia in infancy. Acta Paediatr Scand 1974; 63: 381–8.Google Scholar
Sabel, KG, Wadsworth, C. C-reactive protein (CRP) in early diagnosis of neonatal septicemia. Acta Paediatr Scand 1979; 68: 825–31.Google Scholar
Pourcyrous, M, Bada, HS, Korones, SB, et al. Significance of serial C-reactive protein responses in neonatal infection and other disorders. Pediatrics 1993; 92: 431–5.Google Scholar
Andersen, J, Christensen, R, Hartel, J. Clinical features and epidemiology of septicemia and meningitis in neonates due to Streptococcus agalactiae in Copenhagen County, Denmark: a 10 year survey from 1992 to 2001. Acta Paediatr 2004; 93: 1334–9.Google Scholar
Putto, A, Ruuskanen, O, Meurman, O, et al. C-reactive protein in the evaluation of febrile illness. Arch Dis Child 1986; 61: 24–9.Google Scholar
Raju, VSN, Rao, MN, Rao, VSRM. Cranial sonography in pyogenic meningitis in neonates and infants. J Trop Pediatr 1995; 41: 6873.Google Scholar
Hill, A, Shackelford, GD, Volpe, JJ. Ventriculitis with neonatal bacterial meningitis: identification by real-time ultrasound. J Pediatr 1981; 99: 133–6.Google Scholar
Trivedi, R, Malik, GK, Gupta, RK, et al. Increased anisotropy in neonatal meningitis: an indicator of meningeal inflammation. Neuroradiology 2007; 49: 767–75.Google Scholar
Tibussek, D. Sinclair, A. Yau, I, et al: Late-onset group B streptococcal meningitis has cerebrovascular complications. J Pediatr 2015; 166: 1187–92.Google Scholar
Vogel, LC, Boyer, KM, Gadzala, CA, et al. Prevalence of type-specific group B streptococcal antibody in pregnant women. J Pediatr 1980; 96: 1047–51.Google Scholar
Sidiropoulos, D, Hermann, U, Morell, A, et al. Transplacental passage of intravenous immunoglobulin in the last trimester of pregnancy. J Pediatr 1986; 109: 505–8.Google Scholar
Edwards, MS, Hall, MA, Rench, MA, et al. Patterns of immune response among survivors of group B streptococcal meningitis. J Infect Dis 1990; 161: 6570.Google Scholar
Baker, CJ, Rench, MA, Edwards, MS, et al. Immunization of pregnant women with a polysaccharide vaccine of group B Streptococcus. N Engl J Med 1988; 319: 1180–5.Google Scholar
Sennhauser, FH, Balloch, A, MacDonald, RA, et al. Materno-fetal transfer of IgG anti-Escherichia coli antibodies with enhanced avidity and opsonic activity in very premature neonates. Pediatr Res 1990; 27: 365–71.Google Scholar
Ment, LR, Ehrenkranz, RA, Duncan, CC. Bacterial meningitis as an etiology of perinatal cerebral infarction. Pediatr Neurol 1986; 2: 276–9.Google Scholar
Cohen, C, Rice, EN, Thomas, DE, et al. Diabetes insipidus as a hallmark neuroendocrine complication of neonatal meningitis. Curr Opin Pediatr 1998; 10: 449–52.Google Scholar
McCracken, GH, Mize, SG, Threlkeld, N. Intraventricular gentamicin therapy in gram-negative bacillary meningitis of infancy: report of the Second Neonatal Meningitis Co-operative Study Group. Lancet 1980; i: 787–91.Google Scholar
Renier, D, Flandin, C, Hirsch, E, et al. Brain abscesses in neonates: a study of 30 cases. J Neurosurg 1988; 69: 877–82.Google Scholar
Burdette, JH, Santos, C. Enterobacter sakazakii brain abscess in the neonate: the importance of neuroradiologic imaging. Pediatr Radiol 2000; 30: 33–4.Google Scholar
Isaacs, D. Unnatural selection: reducing antibiotic resistance in neonatal units. Arch Dis Child Fetal Neonatal Ed 2006; 91: F72–4.Google Scholar
Saez-Llorens, X, McCracken, GH. Antimicrobial and anti-inflammatory treatment of bacterial meningitis. Infect Dis Clin North Am 1999; 13: 619–36.Google Scholar
Puopolo, K. Eichenwald, EC: No change in incidence of ampicillin-resistant neonatal early-onset sepsis over 18 years. Pediatrics 2010; 125: e1031–8.Google Scholar
Heath, PT, Nik Yussoff, NK, Baker, CJ. Neonatal meningitis. Arch Dis Child Fetal Neonatal Ed 2003; 88: F137–8.Google Scholar
Tessin, I, Trollfors, B, Thiringer, K, et al. Ampicillin-aminoglycoside combination as initial treatment for neonatal septicaemia or meningitis: a retrospective evaluation of 12 years experience. Acta Paediatr Scand 1991; 80: 911–16.Google Scholar
Adelman, RD, Wirth, F, Rubio, T. A controlled study of the nephrotoxicity of mezlocillin and gentamicin plus ampicillin in the neonate. J Pediatr 1987; 111: 888–93.Google Scholar
Tullus, K, Olsson-Liljequist, B, Lundstrom, G, et al. Antibiotic susceptibility of 629 bacterial blood and CSF isolates from Swedish infants and the therapeutic implications. Acta Paediatr Scand 1991; 80: 205–12.Google Scholar
Quinn, JP, Rodvold, KA. Antibiotic policies in neonatal intensive-care units. Lancet 2000; 355: 946–7.Google Scholar
Lebel, MH, Hoy, MJ, McCracken, GH. Comparative efficacy of ceftriaxone and cefuroxime for treatment of bacterial meningitis. J Pediatr 1989; 114: 1049–54.Google Scholar
Black, SB, Levine, P, Shinefield, HR. The necessity for monitoring chloramphenicol levels when treating neonatal meningitis. J Pediatr 1978; 92: 235–6.Google Scholar
Ahmed, A. A critical evaluation of vancomycin for treatment of bacterial meningitis. Pediatr Infect Dis J 1997; 16: 895–3.Google Scholar
Haimi-Cohen, Y, Amir, J, Weinstock, A, et al. The use of imipenem-cilastatin in neonatal meningitis caused by Citrobacter diversus. Acta Paediatr Int J Paediatr 1993; 82: 530–2.Google Scholar
Leggiadro, RJ. Favorable outcome possible in Citrobacter brain abscess. Pediatr Infect Dis J 1996; 15: 557.Google Scholar
Radetsky, M. Duration of treatment in bacterial meningitis: a historical inquiry. Pediatr Infect Dis J 1990; 9: 29.Google Scholar
Peltola, H, Luhtala, K, Valmari, P. C-reactive protein as a detector of organic complications during recovery from childhood purulent meningitis. J Pediatr 1984; 104: 869–72.Google Scholar
Astruc, J, Taillebois, L, Rodière, F, et al. Raccourcissement du traitment antibiotiques des méningites bactériennes de l’enfant:interêt de la surveillance de la C-réactive protéine. Arch Franc Pédiatr 1990; 47: 637–40.Google Scholar
Mathur, NB Kharod, P, Kumar, S: Evaluation of duration of antibiotic therapy in neonatal bacterial meningitis: a randomized, controlled trial. J Trop Pediatr 2015; 61: 119–25.Google Scholar
Prober, CG. The role of steroids in the management of children with bacterial meningitis. Pediatrics 1995; 95: 2931.Google Scholar
Daoud, AS, Batieha, A, Al-Sheyyab, M, et al. Lack of effectiveness of dexamethasone in neonatal bacterial meningitis. Eur J Pediatr 1999; 158: 230–3.Google Scholar
Ogunlesi, TA. Odigwe, CC, Oladpo, OT. Adjuvant corticosteroids for reducing death in neonatal bacterial meningitis. Cochrane Database Syst Rev 2015; 11: CD010435.Google Scholar
McCracken, GH, Sarff, LD, Glode, MP, et al. Relation between Escherichia coli K1 capsular polysaccharide antigen and clinical outcome in neonatal meningitis. Lancet 1974; ii: 246–50.Google Scholar
Levent, F, Baker, CJ, Rench, MA, Edwards, MS. Early outcomes of group B streptococcal meningitis in the 21st century. Pediatr Infect Dis J 2010; 29: 1009–12.Google Scholar
Bennet, R, Bergdahl, S, Eriksson, M, et al. The outcome of neonatal septicemia during fifteen years. Acta Paediatr Scand 1989; 78: 40–3.Google Scholar
Moffett, KS, Berkowitz, FE. Quadriplegia complicating Escherichia coli meningitis in a newborn infant: case report and review of 22 cases of spinal cord dysfunction in patients with acute bacterial meningitis. Clin Infect Dis 1997; 25: 211–14.Google Scholar
Klinger, G, Chin, CN, Otsobu, H, et al. Prognostic value of EEG in neonatal bacterial meningitis. Pediatr Neurol 2001; 24: 2831.Google Scholar
Siegel, JD, McCracken, GH, Threlkeld, N, et al. Single-dose penicillin prophylaxis of neonatal group B streptococcal disease: conclusion of a 41 month controlled trial. Lancet 1982; i: 1426–30.Google Scholar
Pyati, SP, Pildes, RS, Jacobs, NM, et al. Penicillin in infants weighing two kilograms or less with early-onset group B streptococcal disease. N Engl J Med 1983; 308: 1383–9.Google Scholar
Baker, CJ, Melish, ME, Hall, RT, et al. Intravenous immune globulin for the prevention of nosocomial infection in low-birth-weight neonates. N Engl J Med 1992; 327: 213–19.Google Scholar
Fanaroff, AA, Korones, SB, Wright, LL, et al. A controlled trial of intravenous immune globulin to reduce nosocomial infections in very-low-birth-weight infants. National Institute of Child Health and Human Development Neonatal Research Network. N Engl J Med 1994; 330: 1107–13.Google Scholar
Weisman, LE, Stoll, BJ, Kueser, TJ, et al. Intravenous immune globulin prophylaxis of late-onset sepsis in premature neonates. J Pediatr 1994; 125: 922–30.Google Scholar
Weisman, LE, Cruess, DF, Fischer, GW. Opsonic activity of commercially available standard intravenous immunoglobulin preparations. Pediatr Infect Dis J 1994; 13: 1122–5.Google Scholar
Boyer, KM, Gotoff, SP. Prevention of early-onset neonatal group B streptococcal disease with selective intrapartum chemoprophylaxis. N Engl J Med 1986; 314: 1665–9.Google Scholar
Ascher, DP, Becker, JA, Yoder, BA, et al. Failure of intrapartum antibiotics to prevent culture-proved neonatal group B streptococcal sepsis. J Perinatol 1992; 13: 212–16.Google Scholar
Centers for Disease Control and Prevention. Prevention of perinatal group B streptococcal disease:a public health perspective. MMWR Recomm Rep 1996; 45: 124.Google Scholar
Schrag, SJ, Zywicki, S, Farley, MM, et al. Group B streptococcal disease in the era of intrapartum antibiotic prophylaxis. N Engl J Med 2000; 342: 1520.Google Scholar
Kalliola, S, Vuopio-Varkilla, J, Takala, AK, et al. Neonatal group B streptococcal disease in Finland:a ten-year nationwide study. Pediatr Infect Dis J 1999; 18: 806–10.Google Scholar
Puopolo, KM, Madoff, LC, Eichenwald, EC. Early-onset group B streptococcal disease in the era of maternal screening. Pediatrics 2005; 115: 1240–6.Google Scholar
Philip, AGS, Mills, PC. Use of C-reactive protein in minimizing antibiotic exposure: experience with infants initially admitted to a well-baby nursery. Pediatrics 2000; 106: e4.Google Scholar
Paoletti, LC, Madoff, LC. Vaccines to prevent neonatal GBS infection. Semin Neonatol 2002; 7: 315–23.Google Scholar
Baker, CJ, Paoletti, LC, Rench, MA, et al. Immune response of healthy women to two different group B streptococcal type V capsular polysaccharide-protein conjugate vaccines. J Infect Dis 2004; 189: 1103–12.Google Scholar
Baker, CJ, Carrey, VJ, Rench, MA, et al. Maternal antibody at delivery protects neonates from early-onset streptococcal disease. J Infect Dis 2014; 209: 781–8.Google Scholar
Bonadio, WA, Stanco, L, Bruce, R, et al. Reference values of normal cerebrospinal fluid composition in infants ages 0 to 8 weeks. Pediatr Infect Dis J 1992; 11: 589–91.Google Scholar
Ahmed, A, Hickey, SM, Ehrett, S, et al. Cerebrospinal fluid values in the term neonate. Pediatr Infect Dis J 1996; 15: 298303.Google Scholar

References

Kenneson, A, Cannon, MJ. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol 2007; 17: 253–76.Google Scholar
Manicklal, S, Emery, VC, Lazzarotto, T, et al. The “silent” global burden of congenital cytomegalovirus. Clin Microbiol Rev 2013; 26: 86102.Google Scholar
Dollard, SC, Grosse, SD, Ross, DS. New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection. Rev Med Virol 2007; 17: 355–63.Google Scholar
Stagno, S, Reynolds, DW, Huang, ES, et al. Congenital cytomegalovirus infection. N Engl J Med 1977; 296: 1254–8.Google Scholar
Saigal, S, Lunyk, O, Larke, RP, Chernesky, MA. The outcome in children with congenital cytomegalovirus infection: a longitudinal follow-up study. Am J Dis Child 1982; 136: 896901.Google Scholar
Fowler, KB, McCollister, FP, Dahle, AJ, et al. Progressive and fluctuating sensorineural hearing loss in children with asymptomatic congenital cytomegalovirus infection. J Pediatr 1997; 130: 624–30.Google Scholar
Boppana, SB, Ross, SA, Fowler, KB. Congenital cytomegalovirus infection: clinical outcome. Clin Infect Dis 2013; 57(Suppl 4): S178–81.Google Scholar
Pass, RF, Stagno, S, Myers, GJ, Alford, CA. Outcome of symptomatic congenital cytomegalovirus infection: results of long-term longitudinal follow-up. Pediatrics 1980; 66: 758–62.Google Scholar
Boppana, SB, Fowler, KB, Vaid, Y, et al. Neuroradiographic findings in the newborn period and long-term outcome in children with symptomatic congenital cytomegalovirus infection. Pediatrics 1997; 99: 409–14.Google Scholar
Pass, RF, Anderson, B. Mother-to-child transmission of cytomegalovirus and prevention of congenital infection. J Pediatr Infect Dis Soc 2014; 3: S26.Google Scholar
Boppana, SB, Rivera, LB, Fowler, KB, et al. Intrauterine transmission of cytomegalovirus to infants of women with preconceptional immunity. N Engl J Med 2001; 344: 1366–71.Google Scholar
Monif, GR, Egan, EA 2nd, Held, B, Eitzman, DV. The correlation of maternal cytomegalovirus infection during varying stages in gestation with neonatal involvement. J Pediatr 1972; 80: 1720.Google Scholar
Reynolds, DW, Stagno, S, Stubbs, KG, et al. Inapparent congenital cytomegalovirus infection with elevated cord IgM levels: casual relation with auditory and mental deficiency. N Engl J Med 1974; 290: 291–6.Google Scholar
Hanshaw, JB, Scheiner, AP, Moxley, AW, et al. School failure and deafness after “silent” congenital cytomegalovirus infection. N Engl J Med 1976; 295: 468–70.Google Scholar
Conboy, TJ, Pass, RF, Stagno, S, et al. Intellectual development in school-aged children with asymptomatic congenital cytomegalovirus infection. Pediatrics 1986; 77: 801–6.Google Scholar
Williamson, WD, Demmler, GJ, Percy, AK, Catlin, FI. Progressive hearing loss in infants with asymptomatic congenital cytomegalovirus infection. Pediatrics 1992; 90: 862–6.Google Scholar
Dahle, AJ, Fowler, KB, Wright, JD, et al. Longitudinal investigation of hearing disorders in children with congenital cytomegalovirus. J Am Acad Audiol 2000; 11: 283–90.Google Scholar
Boppana, SB, Fowler, KB, Pass, RF, et al. Congenital cytomegalovirus infection: association between virus burden in infancy and hearing loss. J Pediatr 2005; 146: 817–23.Google Scholar
Forner, G, Abate, D, Mengoli, C, et al. High cytomegalovirus (CMV) DNAemia predicts CMV sequelae in asymptomatic congenitally infected newborns born to women with primary infection during pregnancy. J Infect Dis 2015; 212: 6771.Google Scholar
Stagno, S, Pass, RF, Cloud, G, et al. Primary cytomegalovirus infection in pregnancy: incidence, transmission to fetus, and clinical outcome. JAMA 1986; 256: 1904–8.Google Scholar
Boppana, SB, Pass, RF, Britt, WJ, et al. Symptomatic congenital cytomegalovirus infection: neonatal morbidity and mortality. Pediatr Infect Dis J 1992; 11: 93–9.Google Scholar
Stagno, S, Pass, RF, Alford, CA. Perinatal infections and maldevelopment. Birth Defects Original Article Series 1981; 17: 3150.Google Scholar
McCracken, GH Jr, Shinefield, HM, Cobb, K, et al. Congenital cytomegalic inclusion disease: a longitudinal study of 20 patients. Am J Dis Child 1969; 117: 522–39.Google Scholar
Butt, W, Mackay, RJ, de Crespigny, LC, et al. Intracranial lesions of congenital cytomegalovirus infection detected by ultrasound scanning. Pediatrics 1984; 73: 611–4.Google Scholar
James, SH, Kimberlin, DW, Whitley, RJ. Antiviral therapy for herpesvirus central nervous system infections: neonatal herpes simplex virus infection, herpes simplex encephalitis, and congenital cytomegalovirus infection. Antiviral Res 2009; 83: 207–13.Google Scholar
Troendle Atkins, J, Demmler, GJ, Williamson, WD, et al. Polymerase chain reaction to detect cytomegalovirus DNA in the cerebrospinal fluid of neonates with congenital infection. J Infect Dis 1994; 169: 1334–7.Google Scholar
Noyola, DE, Demmler, GJ, Nelson, CT, et al. Early predictors of neurodevelopmental outcome in symptomatic congenital cytomegalovirus infection. J Pediatr 2001; 138: 325–31.Google Scholar
Williamson, WD, Desmond, MM, LaFevers, N, et al. Symptomatic congenital cytomegalovirus: disorders of language, learning, and hearing. Am J Dis Child 1982; 136: 902–5.Google Scholar
Rivera, LB, Boppana, SB, Fowler, KB, et al. Predictors of hearing loss in children with symptomatic congenital cytomegalovirus infection. Pediatrics 2002; 110: 762–7.Google Scholar
Kimberlin, DW, Lin, CY, Sanchez, PJ, et al. Effect of ganciclovir therapy on hearing in symptomatic congenital cytomegalovirus disease involving the central nervous system: a randomized, controlled trial. J Pediatr 2003; 143: 1625.Google Scholar
Kimberlin, DW, Jester, PM, Sanchez, PJ, et al. Valganciclovir for symptomatic congenital cytomegalovirus disease. N Engl J Med 2015; 372: 933–43.Google Scholar
Wolf, A, Cowen, D, Paige, BH. Toxoplasmic encephalomyelitis. III. A new case of granulomatous encephalomyelitis due to a protozoon. Am J Pathol 1939; 15: 657–94.Google Scholar
McAuley, JB. Congenital toxoplasmosis. J Pediatr Infect Dis Soc 2014; 3: S30–5.Google Scholar
Lynfield, R, Guerina, NG. Toxoplasmosis. Pediatr Rev 1997; 18: 7583.Google Scholar
American Academy of Pediatrics. Toxoplasma gondii Infections. In Kimberlin, DW, Brady, MT, Jackson, MA, Long, SS, eds., Red Book: 2015 Report of the Committee on Infectious Diseases. Elk Grive Village, IL: American Academy of Pediatrics, 2015: 787–96.Google Scholar
Vogel, N, Kirisits, M, Michael, E, et al. Congenital toxoplasmosis transmitted from an immunologically competent mother infected before conception. Clin Infect Dis 1996; 23: 1055–60.Google Scholar
Lindsay, DS, Dubey, JP. Toxoplasma gondii: the changing paradigm of congenital toxoplasmosis. Parasitology 2011; 138: 1829–31.Google Scholar
Hohlfeld, P, Daffos, F, Thulliez, P, et al. Fetal toxoplasmosis: outcome of pregnancy and infant follow-up after in utero treatment. J Pediatr 1989; 115: 765–9.Google Scholar
Foulon, W, Villena, I, Stray-Pedersen, B, et al. Treatment of toxoplasmosis during pregnancy: a multicenter study of impact on fetal transmission and children’s sequelae at age 1 year. Am J Obstet Gynecol 1999; 180: 410–5.Google Scholar
SYROCOT (Systematic Review on Congenital Toxoplasmosis) Study Group, Thiebaut, R, Leproust, S, et al. Effectiveness of prenatal treatment for congenital toxoplasmosis: a meta-analysis of individual patients’ data. Lancet 2007; 369: 115–22.Google Scholar
Prusa, AR, Kasper, DC, Pollak, A, et al. The Austrian Toxoplasmosis Register, 1992–2008. Clin Infect Dis 2015; 60: e410.Google Scholar
Hotop, A, Hlobil, H, Gross, U. Efficacy of rapid treatment initiation following primary Toxoplasma gondii infection during pregnancy. Clin Infect Dis 2012; 54: 1545–52.Google Scholar
Cortina-Borja, M, Tan, HK, Wallon, M, et al. Prenatal treatment for serious neurological sequelae of congenital toxoplasmosis: an observational prospective cohort study. PLoS Med 2010; 7.Google Scholar
Wallon, M, Peyron, F, Cornu, C, et al. Congenital toxoplasma infection: monthly prenatal screening decreases transmission rate and improves clinical outcome at age 3 years. Clin Infect Dis 2013; 56: 1223–31.Google Scholar
Koppe, JG, Loewer-Sieger, DH, de Roever-Bonnet, H. Results of 20-year follow-up of congenital toxoplasmosis. Lancet 1986; 1: 254–6.Google Scholar
Stagno, S, Reynolds, DW, Amos, CS, et al. Auditory and visual defects resulting from symptomatic and subclinical congenital cytomegaloviral and Toxoplasma infections. Pediatrics 1977; 59: 669–78.Google Scholar
Wilson, CB, Remington, JS, Stagno, S, Reynolds, DW. Development of adverse sequelae in children born with subclinical congenital Toxoplasma infection. Pediatrics 1980; 66: 767–74.Google Scholar
Guerina, NG, Hsu, HW, Meissner, HC, et al. Neonatal serologic screening and early treatment for congenital Toxoplasma gondii infection. The New England Regional Toxoplasma Working Group. N Engl J Med 1994; 330: 1858–63.Google Scholar
Stillwaggon, E, Carrier, CS, Sautter, M, McLeod, R. Maternal serologic screening to prevent congenital toxoplasmosis: a decision-analytic economic model. PLoS Neglect Trop Dis 2011; 5: e1333.Google Scholar
Wong, SY, Remington, JS. Toxoplasmosis in pregnancy. Clin Infect Dis 1994; 18: 853–61; quiz 62.Google Scholar
McLeod, R, Boyer, K, Karrison, T, et al. Outcome of treatment for congenital toxoplasmosis, 1981–2004: the National Collaborative Chicago-Based, Congenital Toxoplasmosis Study. Clin Infect Dis 2006; 42: 1383–94.Google Scholar
McAuley, J, Boyer, KM, Patel, D, et al. Early and longitudinal evaluations of treated infants and children and untreated historical patients with congenital toxoplasmosis: the Chicago Collaborative Treatment Trial. Clin Infect Dis 1994; 18: 3872.Google Scholar
Roizen, N, Swisher, CN, Stein, MA, et al. Neurologic and developmental outcome in treated congenital toxoplasmosis. Pediatrics 1995; 95: 1120.Google Scholar
Patel, DV, Holfels, EM, Vogel, NP, et al. Resolution of intracranial calcifications in infants with treated congenital toxoplasmosis. Radiology 1996; 199: 433–40.Google Scholar
Naessens, A, Jenum, PA, Pollak, A, et al. Diagnosis of congenital toxoplasmosis in the neonatal period: a multicenter evaluation. J Pediatr 1999; 135: 714–9.Google Scholar
Gilbert, RE, Thalib, L, Tan, HK, et al. Screening for congenital toxoplasmosis: accuracy of immunoglobulin M and immunoglobulin A tests after birth. J Med Screen 2007; 14: 813.Google Scholar
Olariu, TR, Remington, JS, McLeod, R, et al. Severe congenital toxoplasmosis in the United States: clinical and serologic findings in untreated infants. Pediatr Infect Dis J 2011; 30: 1056–61.Google Scholar
Foulon, W, Pinon, JM, Stray-Pedersen, B, et al. Prenatal diagnosis of congenital toxoplasmosis: a multicenter evaluation of different diagnostic parameters. Am J Obstet Gynecol 1999; 181: 843–7.Google Scholar
Wallon, M, Franck, J, Thulliez, P, et al. Accuracy of real-time polymerase chain reaction for Toxoplasma gondii in amniotic fluid. Obstet Gynecol 2010; 115: 727–33.Google Scholar
Dudgeon, JA. Congenital rubella. J Pediatr 1975; 87: 1078–86.Google Scholar
Reef, SE, Frey, TK, Theall, K, et al. The changing epidemiology of rubella in the 1990s: on the verge of elimination and new challenges for control and prevention. JAMA 2002; 287: 464–72.Google Scholar
Centers for Disease Control and Prevention. Increase in rubella and congenital rubella syndrome – United States, 1988–1990. MMWR Morbid Mortal Wkly Rep 1991; 40: 93–9.Google Scholar
Achievements in public health: elimination of rubella and congenital rubella syndrome – United States, 1969–2004. Ann Pharmacother 2005; 39: 1151–2.Google Scholar
Centers for Disease Control and Prevention. Three cases of congenital rubella syndrome in the postelimination era – Maryland, Alabama, and Illinois, 2012. MMWR Morbid Mortal Wkly Rep 2013; 62:226–9.Google Scholar
Lambert, N, Strebel, P, Orenstein, W, et al. Rubella. Lancet 2015; 385: 2297–307.Google Scholar
Ujiie, M, Nabae, K, Shobayashi, T. Rubella outbreak in Japan. Lancet 2014; 383: 1460–1.Google Scholar
Miller, E, Cradock-Watson, JE, Pollock, TM. Consequences of confirmed maternal rubella at successive stages of pregnancy. Lancet 1982; 2: 781–4.Google Scholar
Neu, N, Duchon, J, Zachariah, P. TORCH infections. Clin Perinatol 2015; 42: 77103.Google Scholar
Meitsch, K, Enders, G, Wolinsky, JS, et al. The role of rubella-immunoblot and rubella-peptide-EIA for the diagnosis of the congenital rubella syndrome during the prenatal and newborn periods. J Med Virol 1997; 51: 280–3.Google Scholar
Mace, M, Cointe, D, Six, C, et al. Diagnostic value of RT-PCR of amniotic fluid for prenatal diagnosis of congenital rubella infection in pregnant women with confirmed primary rubella infection. J Clin Microbiol 2004; 42: 4818–20.Google Scholar
Ueda, K, Nishida, Y, Oshima, K, Shepard, TH. Congenital rubella syndrome: correlation of gestational age at time of maternal rubella with type of defect. J Pediatr 1979; 94: 763–5.Google Scholar
Peckham, CS. Clinical and laboratory study of children exposed in utero to maternal rubella. Arch Dis Childhood 1972; 47: 571–7.Google Scholar
Frey, TK. Neurological aspects of rubella virus infection. Intervirology 1997; 40: 167–75.Google Scholar
Weil, ML, Itabashi, H, Cremer, NE, et al. Chronic progressive panencephalitis due to rubella virus simulating subacute sclerosing panencephalitis. N Engl J Med 1975; 292: 994–8.Google Scholar
Macfarlane, DW, Boyd, RD, Dodrill, CB, Tufts, E. Intrauterine rubella, head size, and intellect. Pediatrics 1975; 55: 797801.Google Scholar
Chang, YC, Huang, CC, Liu, CC. Frequency of linear hyperechogenicity over the basal ganglia in young infants with congenital rubella syndrome. Clin Infect Dis 1996; 22: 569–71.Google Scholar
Lane, B, Sullivan, EV, Lim, KO, et al. White matter MR hyperintensities in adult patients with congenital rubella. Am J Neuroradiol 1996; 17: 99103.Google Scholar
Townsend, JJ, Baringer, JR, Wolinsky, JS, et al. Progressive rubella panencephalitis: late onset after congenital rubella. N Engl J Med 1975; 292: 990–3.Google Scholar
Desmond, MM, Fisher, ES, Vorderman, AL, et al. The longitudinal course of congenital rubella encephalitis in nonretarded children. J Pediatr 1978; 93: 584–91.Google Scholar
McIntosh, ED, Menser, MA. A fifty-year follow-up of congenital rubella. Lancet 1992; 340: 414–5.Google Scholar
Chess, S, Fernandez, P, Korn, S. Behavioral consequences of congenital rubella. J Pediatr 1978; 93: 699703.Google Scholar
Brown, AS, Cohen, P, Greenwald, S, Susser, E. Nonaffective psychosis after prenatal exposure to rubella. Am J Psychiatry 2000; 157: 438–43.Google Scholar
Tanemura, M, Suzumori, K, Yagami, Y, Katow, S. Diagnosis of fetal rubella infection with reverse transcription and nested polymerase chain reaction: a study of 34 cases diagnosed in fetuses. Am J Obstet Gynecol 1996; 174: 578–82.Google Scholar
Reef, SE, Redd, SB, Abernathy, E, et al. The epidemiological profile of rubella and congenital rubella syndrome in the United States, 1998–2004: the evidence for absence of endemic transmission. Clin Infect Dis 2006; 43(Suppl 3): S126–32.Google Scholar
Papania, MJ, Wallace, GS, Rota, PA, et al. Elimination of endemic measles, rubella, and congenital rubella syndrome from the Western hemisphere: the US experience. JAMA Pediatr 2014; 168: 148–55.Google Scholar
McLean, HQ, Fiebelkorn, AP, Temte, JL, Wallace, GS. Prevention of measles, rubella, congenital rubella syndrome, and mumps, 2013: summary recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recommend Rep 2013; 62: 134.Google Scholar
Klass, PE, Brown, ER, Pelton, SI. The incidence of prenatal syphilis at the Boston City Hospital: a comparison across four decades. Pediatrics 1994; 94: 24–8.Google Scholar
Wharton, M, Chorba, TL, Vogt, RL, et al. Case definitions for public health surveillance. MMWR Recommend Rep 1990; 39: 143.Google Scholar
Kwak, J, Lamprecht, C. A review of the guidelines for the evaluation and treatment of congenital syphilis. Pediatr Ann 2015; 44: e108–14.Google Scholar
Sung, L, MacDonald, NE. Syphilis: a pediatric perspective. Pediatr Rev 1998; 19: 1722.Google Scholar
Sanchez, PJ. Laboratory tests for congenital syphilis. Pediatr Infect Dis J 1998; 17: 70–1.Google Scholar
American Academy of Pediatrics. Syphilis. In: Kimberlin, DW, Brady, MT, Jackson, MA, Long, SS, eds., Red Book: 2015 Report of the Committee on Infectious Diseases. Elk Grove, IL: American Academy of Pediatrics, 2015: 755–68.Google Scholar
Stoll, BJ. Congenital syphilis: evaluation and management of neonates born to mothers with reactive serologic tests for syphilis. Pediatr Infect Dis J 1994; 13: 845–52; quiz 53.Google Scholar
Beeram, MR, Chopde, N, Dawood, Y, et al. Lumbar puncture in the evaluation of possible asymptomatic congenital syphilis in neonates. J Pediatr 1996; 128: 125–9.Google Scholar
Srinivasan, G, Ramamurthy, RS, Bharathi, A, et al. Congenital syphilis: a diagnostic and therapeutic dilemma. Pediatr Infect Dis 1983; 2: 436–41.Google Scholar
Sanchez, PJ, Wendel, GD Jr, Grimprel, E, et al. Evaluation of molecular methodologies and rabbit infectivity testing for the diagnosis of congenital syphilis and neonatal central nervous system invasion by Treponema pallidum. J Infect Dis 1993; 167: 148–57.Google Scholar
Michelow, IC, Wendel, GD Jr, Norgard, MV, et al. Central nervous system infection in congenital syphilis. N Engl J Med 2002; 346: 1792–8.Google Scholar
Fiumara, NJ, Lessell, S. Manifestations of late congenital syphilis: an analysis of 271 patients. Arch Dermatol 1970; 102: 7883.Google Scholar
Workowski, KA, Bolan, GA, CDC. Sexually transmitted diseases treatment guidelines, 2015. MMWR Recommend Rep 2015; 64: 1137.Google Scholar
Bialas, KM, Swamy, GK, Permar, SR. Perinatal cytomegalovirus and varicella zoster virus infections: epidemiology, prevention, and treatment. Clin Perinatol 2015; 42: 6175.Google Scholar
American College of Obstetricians and Gynecologists. Cytomegalovirus, parvovirus B19, varicella zoster, and toxoplasmosis in pregnancy (Practice Bulletin No. 151). Obstet Gynecol 2015; 125: 1510–25.Google Scholar
Laforet, EG, Lynch, CL Jr. Multiple congenital defects following maternal varicella: report of a case. N Engl J Med 1947; 236: 534–7.Google Scholar
Pastuszak, AL, Levy, M, Schick, B, et al. Outcome after maternal varicella infection in the first 20 weeks of pregnancy. N Engl J Med 1994; 330: 901–5.Google Scholar
Alkalay, AL, Pomerance, JJ, Rimoin, DL. Fetal varicella syndrome. J Pediatr 1987; 111: 320–3.Google Scholar
Enders, G, Miller, E, Cradock-Watson, J, et al. Consequences of varicella and herpes zoster in pregnancy: prospective study of 1739 cases. Lancet 1994; 343: 1548–51.Google Scholar
Jones, KL, Johnson, KA, Chambers, CD. Offspring of women infected with varicella during pregnancy: a prospective study. Teratology 1994; 49: 2932.Google Scholar
Scharf, A, Scherr, O, Enders, G, Helftenbein, E. Virus detection in the fetal tissue of a premature delivery with a congenital varicella syndrome: a case report. J Perinatal Med 1990; 18: 317–22.Google Scholar
Brunell, PA. Fetal and neonatal varicella-zoster infections. Semin Perinatol 1983; 7: 4756.Google Scholar
Wilson, E, Goss, MA, Marin, M, et al. Varicella vaccine exposure during pregnancy: data from 10 years of the pregnancy registry. J Infect Dis 2008; 197(Suppl 2): S178–84.Google Scholar
Ogilvie, MM. Antiviral prophylaxis and treatment in chickenpox: a review prepared for the UK Advisory Group on Chickenpox on behalf of the British Society for the Study of Infection. J Infect 1998; 36(Suppl 1): 31–8.Google Scholar
Smith, CK, Arvin, AM. Varicella in the fetus and newborn. Semin Fetal Neonat Med 2009; 14: 209–17.Google Scholar
Meyers, JD. Congenital varicella in term infants: risk reconsidered. J Infect Dis 1974; 129: 215–7.Google Scholar
Meyberg-Solomayer, GC, Fehm, T, Muller-Hansen, I, et al. Prenatal ultrasound diagnosis, follow-up, and outcome of congenital varicella syndrome. Fetal Diagn Ther 2006; 21: 296301.Google Scholar
Mouly, F, Mirlesse, V, Meritet, JF, et al. Prenatal diagnosis of fetal varicella-zoster virus infection with polymerase chain reaction of amniotic fluid in 107 cases. Am J Obstet Gynecol 1997; 177: 894–8.Google Scholar
Brown, ZA, Wald, A, Morrow, RA, et al. Effect of serologic status and cesarean delivery on transmission rates of herpes simplex virus from mother to infant. JAMA 2003; 289: 203–9.Google Scholar
Kimberlin, DW. Herpes simplex virus infections of the newborn. Semin Perinatol 2007; 31: 1925.Google Scholar
James, SH, Kimberlin, DW. Neonatal herpes simplex virus infection: epidemiology and treatment. Clin Perinatol 2015; 42: 4759.Google Scholar
Jones, CA, Raynes-Greenow, C, Issacs, D. Population-based surveillance of neonatal HSV infection in Australia (1997–2011). Clin Infect Dis 2014; 59(4): 525–31.Google Scholar
James, SH, Sheffield, JS, Kimberlin, DW. Mother-to-child transmission of herpes simplex virus. J Pediatr Infect Dis Soc 2014; 3: S1923.Google Scholar
Brown, ZA, Benedetti, J, Ashley, R, et al. Neonatal herpes simplex virus infection in relation to asymptomatic maternal infection at the time of labor. N Engl J Med 1991; 324: 1247–52.Google Scholar
Hutto, C, Arvin, A, Jacobs, R, et al. Intrauterine herpes simplex virus infections. J Pediatr 1987; 110: 97101.Google Scholar
Vasileiadis, GT, Roukema, HW, Romano, W, et al. Intrauterine herpes simplex infection. Am J Perinatol 2003; 20: 55–8.Google Scholar
Kimberlin, DW, Lin, CY, Jacobs, RF, et al. Natural history of neonatal herpes simplex virus infections in the acyclovir era. Pediatrics 2001; 108: 223–9.Google Scholar
Kimberlin, DW. Management of HSV encephalitis in adults and neonates: diagnosis, prognosis and treatment. Herpes 2007; 14: 11–6.Google Scholar
Whitley, R, Arvin, A, Prober, C, et al. A controlled trial comparing vidarabine with acyclovir in neonatal herpes simplex virus infection. N Engl J Med 1991; 324: 444–9.Google Scholar
Whitley, R, Arvin, A, Prober, C, et al. Predictors of morbidity and mortality in neonates with herpes simplex virus infections. N Engl J Med 1991; 324: 450–4.Google Scholar
Kimberlin, D. Herpes simplex virus, meningitis and encephalitis in neonates. Herpes 2004; 11(Suppl 2): 65A76A.Google Scholar
Lakeman, FD, Whitley, RJ, National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. Diagnosis of herpes simplex encephalitis: application of polymerase chain reaction to cerebrospinal fluid from brain-biopsied patients and correlation with disease. J Infect Dis 1995; 171: 857–63.Google Scholar
Kimberlin, DW, Lakeman, FD, Arvin, AM, et al. Application of the polymerase chain reaction to the diagnosis and management of neonatal herpes simplex virus disease. J Infect Dis 1996; 174: 1162–7.Google Scholar
Kimberlin, DW, Whitley, RJ, Wan, W, et al. Oral acyclovir suppression and neurodevelopment after neonatal herpes. N Engl J Med 2011; 365: 1284–92.Google Scholar
Whitley, RJ, Nahmias, AJ, Soong, SJ, et al. Vidarabine therapy of neonatal herpes simplex virus infection. Pediatrics 1980; 66: 495501.Google Scholar
Kimberlin, DW, Lin, CY, Jacobs, RF, et al. Safety and efficacy of high-dose intravenous acyclovir in the management of neonatal herpes simplex virus infections. Pediatrics 2001; 108: 230–8.Google Scholar
James, SH, Kimberlin, DW. Neonatal herpes simplex virus infection. Infect Dis Clin North Am 2015; 29(3): 391400.Google Scholar
American College of Obstetricians and Gynecologists. Clinical management guidelines for obstetrician-gynecologists: management of herpes in pregnancy (Practice Bulletin No. 82). Obstet Gynecol 2007; 109: 1489–98.Google Scholar
Arvin, AM, Hensleigh, PA, Prober, CG, et al. Failure of antepartum maternal cultures to predict the infant’s risk of exposure to herpes simplex virus at delivery. N Engl J Med 1986; 315: 796800.Google Scholar
American Academy of Pediatrics. Herpes Simplex. In Kimberlin, DW, Brady, MT, Jackson, MA, Long, SS, eds., Red Book: 2015 Report of the Committee on Infectious Diseases. Oak Grove, IL: American Academy of Pediatrics, 2015: 432–45.Google Scholar

References

Centers for Disease Control and Prevention. Unexplained immunodeficiency and opportunistic infections in infants – New York, New Jersey, California. MMWR Morbid Mortal Wkly Rep 1982; 31: 665–7.Google Scholar
Prendergast, AJ, Essajee, S, Penazzato, M. See comment in PubMed Commons below HIV and the Millennium Development Goals. Arch Dis Child 2015; 100(Suppl 1):S4852.Google Scholar
Abrams, EJ, Myer, L. Can we achieve an AIDS-free generation? Perspectives on the global campaign to eliminate new pediatric HIV infections. J Acquir Immune Defic Syndr 2013; 63(Suppl 2): S208–12.Google Scholar
Hurst, SA, Appelgren, KE, Kourtis, AP. Prevention of mother-to-child transmission of HIV type 1: the role of neonatal and infant prophylaxis. Expert Rev Anti-infect Ther 2015; 13: 169–81.Google Scholar
Mofenson, LM. Prevention in neglected subpopulations: prevention of mother-to-child transmission of HIV infection. Clin Infect Dis 2010; 50(Suppl 3): S130–48.Google Scholar
See comment in PubMed Commons below Mofenson, LM, Cotton, MF. The challenges of success: adolescents with perinatal HIV infection. J Int AIDS Soc 2013; 16: 18650.Google Scholar
Rakhmanina, NY, van den Anker, JN. Pharmacologic prevention of perinatal HIV infection. Early Hum Dev 2014; 90(Suppl 1):S13–5.Google Scholar
White, AB, Mirjahangir, JF, Horvath, H, et al. Antiretroviral interventions for preventing breast milk transmission of HIV. Cochrane Database Syst Rev 2014; 10: CD011323.Google Scholar
Tenthani, L, Haas, AD, Tweya, H, et al. Retention in care under universal antiretroviral therapy for HIV-infected pregnant and breastfeeding women (“Option B+”) in Malawi. AIDS 2014; 28: 589–98.Google Scholar
Shaffer, N, Abrams, EJ, Becquet, R, et al. Option B+ for prevention of mother-to-child transmission of HIV in resource-constrained settings: great promise but some early caution. AIDS 2014; 28: 599601Google Scholar
Colvin, CJ, Konopka, S, Chalker, JC, et al. A systematic review of health system barriers and enablers for antiretroviral therapy (ART) for HIV-infected pregnant and postpartum women. PLoS One 2014; 9: e108150.Google Scholar
Tardieu, M, Tejiokem, M, Nguefack, S. Virus-induced lesions and the fetal brain: examples of the transmission of HIV-1 and CMV from mother to offspring. Handb Clin Neurol 2013; 112: 1103–8.Google Scholar
Wilmshurst, JM, Donald, KA, Eley, B. Update on the key developments of the neurologic complications in children infected with HIV. Curr Opin HIV AIDS 2014; 9: 533–8.Google Scholar
Donald, KA, Hoare, J, Eley, B, Wilmshurst, JM. Neurologic complications of pediatric human immunodeficiency virus: implications for clinical practice and management challenges in the African setting. Semin Pediatr Neurol 2014; 21:311.Google Scholar
Samia, P, Petersen, R, Walker, KG, et al. Prevalence of seizures in children infected with human immunodeficiency virus. J Child Neurol 2013; 28: 297302.Google Scholar
Donald, KA, Walker, KG, Kilborn, T, et al. HIV Encephalopathy: pediatric case series description and insights from the clinic coalface. AIDS Res Ther 2015; 12: 2.Google Scholar
Govender, R, Eley, B, Walker, K, et al. Neurologic and neurobehavioral sequelae in children with human immunodeficiency virus (HIV-1) infection. J Child Neurol 2015; 26: 1355–64.Google Scholar
Recommendations of the US Public Health Service Task Force on the use of zidovudine to reduce perinatal transmission of human immunodeficiency virus, 2014. Available at www.cdc.gov/mmwr/preview/mmwrhtml/00032271.htm (accessed August 30, 2015).Google Scholar
Centers for Disease Control and Prevention. Recommendations for assisting in the prevention of perinatal transmission of human T-lymphotropic virus type III/lymphadenopathy-associated virus and acquired immunodeficiency syndrome. MMWR Morbid Mortal Wkly Rep 1985; 34: 721–6.Google Scholar
Public Health Service Task Force recommendations for use of antiretroviral drugs in pregnant HIV-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States, 2014. Available at www.aidsinfo.nih.gov/contentfiles/lvguidelines/perinatalgl.pdf (accessed August 30, 2015).Google Scholar
American Academy of Pediatrics. Human immunodeficiency virus infection, in Kimberlin, DW, Brady, MT, Jackson, MA, Long, SS, eds., Red Book, 30th edn. Elk Grove, IL: American Academy of Pediatrics, 2015:453–76.Google Scholar
Centers for Disease Control and Prevention. HIV among pregnant women, infants and children, 2017. Available at www.cdc.gov/hiv/risk/gender/pregnant women/facts/index.html (accessed July 28, 2017).Google Scholar
Centers for Disease Control and Prevention. HIV surveillance report, 2010. Available at www.cdc.gov/hiv/topics/surveillance/resources/reports/ (accessed August 21, 2015).Google Scholar
Centers for Disease Control and Prevention. Racial/ethnic disparities among children with diagnoses of perinatal HIV infection – 34 states, 2004–2007. MMWR Morbid Mortal Wkly Rep 2010; 59:97101.Google Scholar
Cooper, ER, Charurat, M, Mofenson, L, et al. Combination antiretroviral strategies for the treatment of pregnant HIV-1-infected women and prevention of perinatal HIV-1 transmission. J Acquir Immune Defic Syndr 2002; 29: 484–94.Google Scholar
Townsend, CL, Byrne, L, Cortina-Borja, M, et al. Earlier initiation of ART and further decline in mother-to-child HIV transmission rates, 2000–2011. AIDS 2014; 28: 1049–57.Google Scholar
Nesheim, S, Taylor, A, Lampe, MA, et al. A framework for elimination of perinatal transmission of HIV in the United States. Pediatrics 2012; 130: 738–44.Google Scholar
Whitmore, SK, Taylor, AW, Espinoza, L, et al. Correlates of mother-to-child transmission of HIV in the United States and Puerto Rico. Pediatrics 2012; 129:e7481.Google Scholar
Camacho-Gonzalez, AF, Kingbo, MH, Boylan, A, et al. Missed opportunities for prevention of mother-to-child transmission in the United States. AIDS 2015; 29: 1511–15.Google Scholar
UNAIDS. 2014 Progress Report on the Global Plan towards the Elimination of New HIV Infections among Children by 2015 and Keeping Their Mothers Alive. Geneva: Joint United Nations Programme on HIV/AIDS, 2014.Google Scholar
UNAIDS. Fact sheet - Latest statistics on the status of the AIDS epidemic. Geneva: Joint United Nations Programme on HIV/AIDS, 2017. Available at http://www.unaids.org/en/resources/fact-sheet (accessed July 28, 2017).Google Scholar
Piot, P, Abdool Karim, SS, Hecht, R, et al. Defeating AIDS: advancing global health. Lancet 2015; 386: 171218.Google Scholar
Newell, ML, Coovadia, H, Cortina-Borja, M, et al. Ghent International AIDS Society (IAS) Working Group on HIV Infection in Women and Children. Mortality of infected and uninfected infants born to HIV-infected mothers in Africa: a pooled analysis. Lancet 2004; 364: 1236–43.Google Scholar
Working Group on Mother-to-Child Transmission of HIV Infection. Rates of mother-to-child transmission of HIV-1 in Africa, America, and Europe: results from 13 perinatal studies. J Acquir Immune Defic Syndr 1995; 8: 506–10.Google Scholar
European Collaborative Study. Vertical transmission of HIV-1: maternal immune status and obstetric factors. AIDS 1996; 10: 1675–81.Google Scholar
Shetty, AK, Maldonado, YA. Human immunodeficiency virus/Acquired immunodeficiency syndrome in the infant. In Wilson, C, Nizet, V, Maldonado, YA, Remington, JS, Klein, JO (eds.), Remington and Klein’s Infectious Diseases of the Fetus and Newborn Infant, 8th edn. Philadelphia: Elsevier/Saunders, 2015: 619–74.Google Scholar
Connor, EM, Sperling, RS, Gelber, R, et al. Reduction of maternal-infant transmission of human immunodeficiency virus type 1 with zidovudine treatment. N Engl J Med 1994; 331: 1173–80.Google Scholar
DeCock, KM, Fowler, MG, Mercier, E, et al. Prevention of mother-to-child HIV transmission in resource-poor countries: translating research into policy and practice. JAMA 2000; 283: 1175–82.Google Scholar
Thorne, C, Newell, ML. Prevention of mother-to-child transmission of HIV infection. Curr Opin Infect Dis 2004; 17: 247–52.Google Scholar
Stevens, J, Hermione, L. Mother to child transmission of HIV: what works and how much is enough? J Infect 2014; 69(Suppl 1): S5662.Google Scholar
Magder, LS, Mofenson, L, Paul, ME, et al. Risk factors for in utero and intrapartum transmission of HIV. J Acquir Immune Defic Syndr 2005; 38: 8795.Google Scholar
Sperling, RS, Shapiro, DE, Coombs, RW, et al. Maternal viral load, zidovudine treatment, and the risk of transmission of human immunodeficiency virus type 1 from mother to infant. N Engl J Med 1996; 335: 1621–9.Google Scholar
International Perinatal HIV Group. The mode of delivery and the risk of vertical transmission of human immunodeficiency virus type 11: a meta-analysis of 15 prospective studies. N Engl J Med 1999; 340: 977–87.Google Scholar
European Mode of Delivery Collaboration. Elective cesarean section versus vaginal delivery in prevention of vertical HIV-1 transmission: a randomized clinical trial. Lancet 1999; 353: 1035–9.Google Scholar
Kuhn, L, Sinkala, M, Kankasa, C, et al. High uptake of exclusive breastfeeding and reduced early post-natal HIV transmission. PLoS One 2007; 2: e1363.Google Scholar
Rollins, N, Coovadia, HM. Breastfeeding and HIV transmission in the developing world: past, present, future. Curr Opin HIV AIDS 2013; 8: 467–73.Google Scholar
McArthur, JC, Brew, BJ, Nath, A. Neurological complications of HIV infection. Lancet 2005; 4: 543–55.Google Scholar
Van Rie, HPR, Dow, A, Robertson, K. Neurologic and neurodevelopmental manifestations of pediatric HIV/AIDS: a global perspective. Eur J Paediatr Neurol 2007; 11: 19.Google Scholar
Pearson, DA, McGrath, NM, Nozycee, M, et al. Predicting HIV disease progression in children using measures of neuropsychological and neurological functioning. Pediatric AIDS Clinical Trials 152 Study Team. Pediatrics 2000; 106: e76.Google Scholar
UNAIDS. Report on the Global AIDS Epidemic 2013. Geneva: Joint United Nations Programme on HIV/AIDS, 2013.Google Scholar
van Arnhem, LA, Bunders, MJ, Scherpbier, HJ, et al. Neurologic abnormalities in HIV-1 infected children in the era of combination antiretroviral therapy. PLoS One 2013; 8: e64398.Google Scholar
Foster, CJ, Biggs, RL, Melvin, D, et al. Neurodevelopmental outcomes in children with HIV infection under 3 years of age. Dev Med Child Neurol 2006; 48: 677–82.Google Scholar
Crowell, CS, Malee, KM, Yogev, R, Muller, WJ. Neurologic disease in HIV-infected children and the impact of combination antiretroviral therapy. Rev Med Virol 2014; 24: 316–31.Google Scholar
Tardieu, M, Le, CJ, Persoz, A, et al. HIV-1-related encephalopathy in infants compared with children and adults. French Pediatric HIV Infection Study and the SEROCO Group. Neurology 2000; 54: 1089–95.Google Scholar
Sturdevant, CB, Dow, A, Jabara, CB, et al. Central nervous system compartmentalization of HIV-1 subtype C variants early and late in infection in young children. PLOS Pathog 2012; 8: e1003094.Google Scholar
Gorry, PR, Bristol, G, Zack, JA, et al. Macrophage tropism of human immunodeficiency virus type 1 isolates from brain and lymphoid tissues predict neurotropism independent of coreceptor specificity. J Virol 2001; 75: 10073–89.Google Scholar
Cooper, ER, Hanson, C, Diaz, C, et al. Encephalopathy and progression of human immunodeficiency virus disease in a cohort of children with perinatally acquired human immunodeficiency virus infection. J Pediatr 1998; 132: 808–12.Google Scholar
Labato, MN, Caldwell, MB, Ng, P, et al. Encephalopathy in children with perinatally acquired human immunodeficiency virus infection. J Pediatr 1995; 126: 710–15.Google Scholar
Chiriboga, CA, Fleishman, S, Champion, S, et al. Incidence and prevalence of HIV encephalopathy in children with HIV infection receiving highly active anti-retroviral therapy (HAART). J Pediatr 2005; 146: 402–7.Google Scholar
Belman, AL, Ultmann, MH, Horoupian, D, et al. Neurologic complications in infants and children with acquired immune deficiency syndrome. Ann Neurol 1985; 18: 560–6.Google Scholar
Epstein, LG, Sharer, LR, Oleske, JM, et al. Neurologic manifestations of human immunodeficiency virus infection in children. Pediatrics 1986; 78: 678–87.Google Scholar
Mintz, M, Rapaport, R, Oleske, JM, et al. Progressive encephalopathy in children with acquired immunodeficiency syndrome. Am J Dis Child 1989; 143: 771–4.Google Scholar
Mitchell, CD, HIV-1 encephalopathy among perinatally infected children: neuropathogenesis and response to highly active antiretroviral therapy. Ment Retard Dev Disabil Res Rev 2006; 12: 216–22.Google Scholar
Nachman, SA, et al., Incidence of noninfectious conditions in perinatally HIV-infected children and adolescents in the HAART era. Arch Pediatr Adolesc Med 2009; 163: 164–71.Google Scholar
Patel, K, et al., Impact of HAART and CNS-penetrating antiretroviral regimens on HIV encephalopathy among perinatally infected children and adolescents. AIDS 2009; 23: 1893–901.Google Scholar
Laughton, B, Cornell, M, Boivin, M, Van Rie, A. Neurodevelopment in perinatally HIV-infected children: a concern for adolescence. J Int AIDS Soc 2013; 16:18603.Google Scholar
Heather, C, Swang, M, Lawrence, J, et al. Of mice and monkeys: can animal models be utilized to study neurological consequences of pediatric HIV-1 infection. ACS Chem Neurosci 2015; 6: 1276–89.Google Scholar
Bissel, SJ, Wiley, CA. Human immunodeficiency virus infection of the brain: pitfalls in evaluating infected/affected cell populations. Brain Pathol 2004; 14:97108.Google Scholar
Persidsky, Y, Zheng, J, Miller, D, Gendelman, HE. Mononuclear phagocytes mediate blood-brain barrier compromise and neuronal injury during HIV-1-associated dementia. J Leukocyt Biol 2001; 68(3): 413–22.Google Scholar
Blumberg, BM, Gelbard, HA, Epstein, LG. HIV-1 infection of the developing nervous system: central role of astrocytes in pathogenesis. Virus Res 1994; 32: 253–67.Google Scholar
Tornatore, C, Chandra, R, Berger, JR, Major, EO. HIV-1 infection of subcortical astrocytes in the pediatric central nervous system. Neuolrogy 1994; 44: 481–7.Google Scholar
Canto-Nogues, C, Sanchez-Ramon, S, Alvarez, S, et al. HIV-1 infection of neurons might account for progressive HIV-1-associated encephalopathy in children. J Mol Neurosci 2005; 27:7989.Google Scholar
Chen, MF, Gill, AJ, Kolson, DL. Neuropathogenesis of HIV-associated neurocognitive disorders: roles for immune activation, HIV blipping and viral tropism. Curr Opin HIV AIDS 2014; 9: 559–64.Google Scholar
Kaul, M. HIV’s double strike at the brain: neuronal toxicity and compromised neurogenesis. Front Biosci 2008; 13: 2484–94.Google Scholar
Foster, SB, Lu, M, Glaze, DG, et al. Associations of cytokines, sleep patterns, and neurocognitive function in youth with HIV infection. Clin Immunol 2012; 144:1323.Google Scholar
Epstein, LG, Gelbard, HA. HIV-1-induced neuronal injury in the developing brain. J Leukocyt Biol 1999; 65: 453–7.Google Scholar
Schwartz, L, Major, EO. Neural progenitors and HIV-1-associated central nervous system disease in adults and children. Curr HIV Res 2006; 4: 319–27.Google Scholar
Medders, KE, Kaul, M. Mitogen-activated protein kinase p38 in HIV infection and associated brain injury. J Neuroimmun Pharmacol 2011; 6: 202–15.Google Scholar
Rodriguez-Franco, EJ, Cantres-Rosario, YM, Plaud-Valentin, M, et al. Dysregulation of macrophage-secreted cathepsin B contributes to HIV-1-linked neuronal apoptosis. PLoS One 2012; 7:e36571.Google Scholar
Teodorof, C, Divakar, S, Soontornniyomkij, B, et al. Intracellular mannose binding lectin mediates subcellular trafficking of HIV-1 gp120 in neurons. Neurobiol Dis 2014; 69:5464.Google Scholar
Singh, KK, Wang, Y, Gray, KP, et al. Genetic variants in the host restriction factor APOBEC3G are associated with HIV-1-related disease progression and central nervous system impairment in children. J Acquir Immune Defic Syndr 2013; 62:197203.Google Scholar
Tornatore, C, Chandra, R, Berger, JR, Major, EO. HIV-1 infection of subcortical astrocytes in the pediatric central nervous system. Neurology 1994; 44: 481–7.Google Scholar
da Cunha, A, Mintz, M, Eiden, LE, Sharer, LR. A neuronal and neuroanatomical correlate of HIV-1 encephalopathy relative to HIV-1 encephalitis in HIV-1 infected children. J Neuropathol Exp Neurol 1997; 56: 974–87.Google Scholar
Lyman, YD, Kress, Y, Kure, K, et al. Detection of HIV in fetal central nervous tissue. AIDS 1990; 4: 917–20.Google Scholar
Levy, JA, Shimabukuro, J, Hollander, H, et al. Isolation of AIDS-associated retroviruses from cerebrospinal fluid and brain of patients with neurological symptoms. Lancet 1985; 2: 586–8.Google Scholar
European Collaborative Study. Neurologic signs in young children with human immunodeficiency virus infection. Pediatr Infect Dis J 1990; 9: 402–6.Google Scholar
George, R, Andronikou, S, du Plessis, AM, et al. Central nervous system manifestations of HIV infection in children. Pediatr Radiol 2009; 39: 575–85.Google Scholar
Hoare, J, Ransford, GL, Phillips, N, et al. Systematic review of neuroimaging studies in vertically transmitted HIV positive children and adolescents. Metab Brain Dis 2014; 29: 221–9.Google Scholar
Ackermann, C, Andronikou, S, Laughton, B, et al. White matter signal abnormalities in children with suspected HIV-related neurologic disease on early combination antiretroviral therapy. Pediatr Infect Dis J 2014; 33:e207–12.Google Scholar
Hoare, J, Fouche, JP, Phillips, N, et al. White matter micro-structural changes in ART-naïve and ART-treated children and adolescents infected with HIV in South Africa. AIDS 2015; 29: 1793–801.Google Scholar
Hoare, J, Fouche, JP, Spottiswoode, B, et al. A diffusion tensor imaging and neurocognitive study of HIV-positive children who are HAART-naive “slow progressors.” J Neurovirol 2012; 18: 205–12.Google Scholar
Panel on Antiretroviral Therapy and Medical Management of HIV-Infected Children. Guidelines for the use of antiretroviral agents in pediatric HIV infection. Available at http://aidsinfo.nih.gov/contentfiles/lvguidelines/pediatricguidelines.pdf (accessed July 28, 2017).Google Scholar
Abrams, EJ, Kuhn, L. Should treatment be started among all HIV-infected children and then stopped?, Lancet 2003; 362: 1595–6.Google Scholar
Violari, A, Cotton, MF, Gibb, DM, et al. Early antiretroviral therapy and mortality among HIV-infected infants. N Engl J Med 2008; 359: 2233–44.Google Scholar
Saavedra-Lozano, J, Ramos, JT, Sanz, F, et al. Salvage therapy with abacavir and other reverse transcriptase inhibitors for human immunodeficiency-associated encephalopathy. Pediatr Infect Dis J 2006; 25: 1142–52.Google Scholar
DeCarli, C, Fugate, L, Falloon, J, et al. Brain growth and cognitive improvement in children with human immunodeficiency virus-induced encephalopathy after 6 months of continuous infusion zidovudine therapy. J Acquir Immune Defic Syndr 1991; 4: 585–92.Google Scholar
McCoig, C, Castrejon, MM, Castano, E, et al. Effects of combination antiretroviral therapy on cerebrospinal fluid HIV RNA, HIV resistance, and clinical manifestations of encephalopathy. J Pediatr 2002; 141:3644.Google Scholar
Laughton, B, Cornell, M. Grove, D, et al. Early antiretroviral therapy improves neurodevelopmental outcomes in infants. AIDS 2012; 26: 1685–90.Google Scholar
Cohen, S, Ter Stege, JA, Geurtsen, GJ, et al. Poorer cognitive performance in perinatally HIV-infected children versus healthy socioeconomically matched controls. Clin Infect Dis 2015; 60: 1111–19.Google Scholar
Nichols, SL, Brummel, SS, Smith, RA, et al. Executive functioning in children and adolescents with perinatal HIV infection. Pediatr Infect Dis J 2015; 34: 969–75.Google Scholar

References

American College of Obstetricians and Gynecologists’ Task Force on Neonatal Encephalopathy. Executive summary: neonatal encephalopathy and neurologic outcome. Obstet Gynecol, 2014; 123(4): 896901.Google Scholar
Biagas, K., Hypoxic-ischemic brain injury: advancements in the understanding of mechanisms and potential avenues for therapy. Curr Opin Pediatr 1999; 11(3): 223–8.Google Scholar
Greene, CL, Goodman, SI. Catastrophic metabolic encephalopathies in the newborn period: evaluation and management. Clin Perinatol 1997; 24(4): 773–86.Google Scholar
Khong, PL, et al. MRI of neonatal encephalopathy. Clin Radiol 2003; 58(11): 833–44.Google Scholar
Vannucci, RC. Hypoxic-ischemic encephalopathy. Am J Perinatol 2000; 17(3): 113–20.Google Scholar
Barkovich, AJ, Sargent, SK. Profound asphyxia in the premature infant: imaging findings. AJNR Am J Neuroradiol 1995; 16(9): 1837–46.Google Scholar
Barkovich, AJ, et al. Perinatal asphyxia: MR findings in the first 10 days. AJNR Am J Neuroradiol 1995; 16(3): 427–38.Google Scholar
Roland, EH, et al. Perinatal hypoxic-ischemic thalamic injury: clinical features and neuroimaging. Ann Neurol 1998; 44(2): 161–6.Google Scholar
Hoon, AH Jr, et al. Brain magnetic resonance imaging in suspected extrapyramidal cerebral palsy: observations in distinguishing genetic-metabolic from acquired causes. J Pediatr 1997; 131(2): 240–5.Google Scholar
Schwartz, ES, Barkovich, AJ. Brain and spine injuries in infancy and childhood. In Pediatric Neuroimaging. Philadelphia: Lippincott Williams & Wilkins, 2012: 240366.Google Scholar
Barkovich, AJ, Patay, Z. Metabolic, toxic, and inflammatory brain disorders. In Barkovich, AJ, Raybaud, C, eds., Pediatric Neuroimaging. Philadelphia: Lippincott Williams & Wilkins, 2012: 81239.Google Scholar
Haas, RH, et al. Acute basal ganglia infarction in propionic acidemia. J Child Neurol 1995; 10(1): 1822.Google Scholar
Greenamyre, T, et al. Evidence for transient perinatal glutamatergic innervation of globus pallidus. J Neurosci 1987; 7(4): 1022–30.Google Scholar
Penn, AA, et al. Kernicterus in a full term infant. Pediatrics 1994; 93(6 Pt 1): 1003–6.Google Scholar
Ankarcrona, M. et al. Glutamate-induced neuronal death: a succession of necrosis or apoptosis depending on mitochondrial function. Neuron 1995; 15(4): 961–73.Google Scholar
Eliasson, MJ, et al. Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med 1997; 3(10): 1089–95.Google Scholar
Johnston, MV. Hypoxic and ischemic disorders of infants and children: lecture for 38th Meeting of Japanese Society of Child Neurology, Tokyo, Japan, July 1996. Brain Dev 1997; 19(4): 235–9.Google Scholar
Johnston, MV, Hoon, AH Jr. Possible mechanisms in infants for selective basal ganglia damage from asphyxia, kernicterus, or mitochondrial encephalopathies. J Child Neurol 2000; 15(9): 588–91.Google Scholar
Shi, Y, et al. Role of carbon monoxide and nitric oxide in newborn infants with postasphyxial hypoxic-ischemic encephalopathy. Pediatrics 2000; 106(6): 1447–51.Google Scholar
Blomgren, K, Hagberg, H. Free radicals, mitochondria, and hypoxia-ischemia in the developing brain. Free Radic Biol Med 2006; 40(3): 388–97.Google Scholar
Rousset, CI, et al. Mitochondria and perinatal brain injury. J Mater Fetal Neonatal Med 2012; 25(Suppl 1): 35–8.Google Scholar
Agani, FH, et al. The role of mitochondria in the regulation of hypoxia-inducible factor 1 expression during hypoxia. J Biol Chem 2000; 275(46): 35863–7.Google Scholar
Goldberg, RN, et al. Hyperammonemia associated with perinatal asphyxia. Pediatrics 1979; 64(3): 336–41.Google Scholar
Sheth, RD, Hobbs, GR, Mullett, M. Neonatal seizures: incidence, onset, and etiology by gestational age. J Perinatol 1999; 19(1): 40–3.Google Scholar
Vasudevan, C, Levene, M. Epidemiology and aetiology of neonatal seizures. Semin Fetal Neonatal Med 2013; 18(4): 185–91.Google Scholar
Van Hove, JL, Lohr, NJ. Metabolic and monogenic causes of seizures in neonates and young infants. Mol Genet Metab 2011; 104(3): 214–30.Google Scholar
Noh, GJ, Tavyev Asher, YJ, Graham, JM Jr. Clinical review of genetic epileptic encephalopathies. Eur J Med Genet 2012; 55(5): 281–98.Google Scholar
Papetti, L, et al. Metabolic epilepsy: an update. Brain Dev 2013; 35(9): 827–41.Google Scholar
Takahashi, Y, et al. Epilepsy in peroxisomal diseases. Epilepsia 1997; 38(2): 182–8.Google Scholar
Gire, C, et al. Clinical features and neuroradiological findings of mitochondrial pathology in six neonates. Childs Nerv Syst 2002; 18(11): 621–8.Google Scholar
von Kleist-Retzow, JC, et al. Antenatal manifestations of mitochondrial respiratory chain deficiency. J Pediatr 2003; 143(2): 208–12.Google Scholar
Enns, GM, et al. Clinical and molecular features of congenital disorder of glycosylation in patients with type 1 sialotransferrin pattern and diverse ethnic origins. J Pediatr 2002; 141(5): 695700.Google Scholar
Hoffmann, GF, Surtees, RA, Wevers, RA. Cerebrospinal fluid investigations for neurometabolic disorders. Neuropediatrics 1998; 29(2): 5971.Google Scholar
Hyland, K, Arnold, LA. Value of lumbar puncture in the diagnosis of infantile epilepsy and folinic acid-responsive seizures. J Child Neurol 2002; 17(Suppl 3): 3S48–55; discussion 3S56.Google Scholar
Mastrangelo, M, Leuzzi, V. Genes of early-onset epileptic encephalopathies: from genotype to phenotype. Pediatr Neurol 2012; 46(1): 2431.Google Scholar
Nordli, DR Jr. Epileptic encephalopathies in infants and children. J Clin Neurophysiol 2012; 29(5): 420–4.Google Scholar
Epi, KC, et al. De novo mutations in epileptic encephalopathies. Nature 2013; 501(7466): 217–21.Google Scholar
Veeramah, KR, et al. Exome sequencing reveals new causal mutations in children with epileptic encephalopathies. Epilepsia 2013; 54(7): 1270–81.Google Scholar
Bekiesiniska-Figatowska, M, Rokicki, D, Walecki, J. MRI in nonketotic hyperglycinaemia: case report. Neuroradiology 2001; 43(9): 792–3.Google Scholar
del Toro, M, et al. Progressive vacuolating glycine leukoencephalopathy with pulmonary hypertension. Ann Neurol 2006; 60(1): 148–52.Google Scholar
Mourmans, J, et al. Sequential MR imaging changes in nonketotic hyperglycinemia. Am J Neuroradiol 2006; 27(1): 208–11.Google Scholar
Roldan, A, et al. Glycine and other neurotransmitter amino acids in cerebrospinal fluid in perinatal asphyxia and neonatal hypoxic-ischaemic encephalopathy. Acta Paediatr 1999; 88(10): 1137–41.Google Scholar
Meyer, S, et al. A novel missense mutation in a neonate with nonketotic hyperglycinemia. Pediatr Neurol 2010; 43(5): 363–7.Google Scholar
Eyaid, WM, et al. An inborn error of metabolism presenting as hypoxic-ischemic insult. Pediatr Neurol 2005; 32(2): 134–6.Google Scholar
Topcu, M, et al. Molybdenum cofactor deficiency: report of three cases presenting as hypoxic-ischemic encephalopathy. J Child Neurol 2001; 16(4): 264–70.Google Scholar
Bayram, E, et al. Molybdenum cofactor deficiency: review of 12 cases (MoCD and review). Eur J Paediatr Neurol 2013; 17(1): 16.Google Scholar
Schwahn, BC, et al. Efficacy and safety of cyclic pyranopterin monophosphate substitution in severe molybdenum cofactor deficiency type A: a prospective cohort study. Lancet 2015; 386(10007): 1955–63.Google Scholar
Plecko, B, et al. Biochemical and molecular characterization of 18 patients with pyridoxine-dependent epilepsy and mutations of the antiquitin (ALDH7A1) gene. Hum Mutat 2007; 28(1): 1926.Google Scholar
Scharer, G, et al. The genotypic and phenotypic spectrum of pyridoxine-dependent epilepsy due to mutations in ALDH7A1. J Inherit Metab Dis 2010; 33(5): 571–81.Google Scholar
Baxter, P. Epidemiology of pyridoxine dependent and pyridoxine responsive seizures in the UK. Arch Dis Child 1999; 81(5): 431–3.Google Scholar
Baxter, P, et al. Pyridoxine-dependent seizures: demographic, clinical, MRI and psychometric features, and effect of dose on intelligence quotient. Dev Med Child Neurol 1996; 38(11): 9981006.Google Scholar
Haenggeli, CA, Girardin, E, Paunier, L. Pyridoxine-dependent seizures, clinical and therapeutic aspects. Eur J Pediatr 1991; 150(7): 452–5.Google Scholar
Hyland, K, et al. Folinic acid responsive seizures: a new syndrome? J Inherit Metab Dis 1995; 18(2): 177–81.Google Scholar
Torres, OA, et al. Folinic acid-responsive neonatal seizures. J Child Neurol 1999; 14(8): 529–32.Google Scholar
Gallagher, RC, et al. Folinic acid-responsive seizures are identical to pyridoxine-dependent epilepsy. Ann Neurol 2009; 65(5): 550–6.Google Scholar
Hoffmann, GF, et al. Pyridoxal 5’-phosphate may be curative in early-onset epileptic encephalopathy. J Inherit Metab Dis 2007; 30(1): 96–9.Google Scholar
Mills, PB, et al. Neonatal epileptic encephalopathy caused by mutations in the PNPO gene encoding pyridox(am)ine 5’-phosphate oxidase. Hum Mol Genet 2005; 14(8): 1077–86.Google Scholar
Jaeken, J. Genetic disorders of gamma-aminobutyric acid, glycine, and serine as causes of epilepsy. J Child Neurol 2002; 17(Suppl 3): 3S84–7; discussion 3S88.Google Scholar
Kraoua, I, et al. 3-Phosphoglycerate dehydrogenase deficiency: description of two new cases in Tunisia and review of the literature. Neuropediatrics 2013; 44(5): 281–5.Google Scholar
Haberle, J, et al. Congenital glutamine deficiency with glutamine synthetase mutations. N Engl J Med 2005; 353(18): 1926–33.Google Scholar
Haberle, J, et al. Glutamine supplementation in a child with inherited GS deficiency improves the clinical status and partially corrects the peripheral and central amino acid imbalance. Orphanet J Rare Dis 2012; 7: 48.Google Scholar
Ruzzo, EK, et al. Deficiency of asparagine synthetase causes congenital microcephaly and a progressive form of encephalopathy. Neuron 2013; 80(2): 429–41.Google Scholar
Alfadhel, M, et al. Asparagine synthetase deficiency: new inborn errors of metabolism. JIMD Rep 2015; 22: 11–6.Google Scholar
Medina-Kauwe, LK, et al. 4-Aminobutyrate aminotransferase (GABA-transaminase) deficiency. J Inherit Metab Dis 1999; 22(4): 414–27.Google Scholar
Parviz, M, et al. Disorders of GABA metabolism: SSADH and GABA-transaminase deficiencies. J Pediatr Epilepsy 2014; 3(4): 217–27.Google Scholar
Au, KM, et al. Diagnosis of dihydropyrimidine dehydrogenase deficiency in a neonate with thymine-uraciluria. Hong Kong Med J 2003; 9(2): 130–2.Google Scholar
Enns, GM, et al. Head imaging abnormalities in dihydropyrimidine dehydrogenase deficiency. J Inherit Metab Dis 2004; 27(4): 513–22.Google Scholar
Marie, S, et al. AICA-ribosiduria: a novel, neurologically devastating inborn error of purine biosynthesis caused by mutation of ATIC. Am J Hum Genet 2004; 74(6): 1276–81.Google Scholar
Jurecka, A, et al. Adenylosuccinate lyase deficiency. J Inherit Metab Dis 2015; 38(2): 231–42.Google Scholar
Stockler, S, et al. Guanidinoacetate methyltransferase deficiency: the first inborn error of creatine metabolism in man. Am J Hum Genet 1996; 58(5): 914–22.Google Scholar
Gordon, N. Guanidinoacetate methyltransferase deficiency (GAMT). Brain Dev 2010; 32(2): 7981.Google Scholar
De Vivo, DC, et al. Defective glucose transport across the blood-brain barrier as a cause of persistent hypoglycorrhachia, seizures, and developmental delay. N Engl J Med 1991; 325(10): 703–9.Google Scholar
De Vivo, DC, Leary, L, Wang, D. Glucose transporter 1 deficiency syndrome and other glycolytic defects. J Child Neurol 2002; 17(Suppl 3): S1523; discussion S24–5.Google Scholar
Barkovich, AJ. Pediatric Neuroimaging, 3rd edn. Philadelphia: Lippincott, Williams & Wilkins, 2000: 71156, 162205.Google Scholar
Norman, MG, et al. Perinatal hemorrhagic and hypoxic-ischemic lesions. In Norman, MG, ed., Congenital Malformations of the Brain: Pathological and Genetic Aspects. Oxford University Press, 1995: 419–23.Google Scholar
Birch-Machin, MA, et al. Fatal lactic acidosis in infancy with a defect of complex III of the respiratory chain. Pediatr Res 1989; 25(5): 553–9.Google Scholar
von Dobeln, U, et al. Fatal neonatal lactic acidosis with respiratory insufficiency due to complex I and IV deficiency. Acta Paediatr 1993; 82(12): 1079–81.Google Scholar
Zeviani, M, Bertagnolio, B, Uziel, G. Neurological presentations of mitochondrial diseases. J Inherit Metab Dis 1996; 19(4): 504–20.Google Scholar
Muraki, K, et al. Severe lactic acidosis and neonatal death in Pearson syndrome. J Inherit Metab Dis 1997; 20(1): 43–8.Google Scholar
Procaccio, V, et al. Nuclear DNA origin of mitochondrial complex I deficiency in fatal infantile lactic acidosis evidenced by transnuclear complementation of cultured fibroblasts. J Clin Invest 1999; 104(1): 8392.Google Scholar
Willis, TA, et al. Cytochrome oxidase deficiency presenting as birth asphyxia. Dev Med Child Neurol 2000; 42(6): 414–7.Google Scholar
Molinari, F, et al. Impaired mitochondrial glutamate transport in autosomal recessive neonatal myoclonic epilepsy. Am J Hum Genet 2005; 76(2): 334–9.Google Scholar
Muntau, AC, et al. Combined D-2- and L-2-hydroxyglutaric aciduria with neonatal onset encephalopathy: a third biochemical variant of 2-hydroxyglutaric aciduria? Neuropediatrics 2000; 31(3): 137–40.Google Scholar
Hoffmann, GF, et al. Clinical and biochemical phenotype in 11 patients with mevalonic aciduria. Pediatrics 1993; 91(5): 915–21.Google Scholar
Bakkeren, JA, et al. Organic aciduria in hypoxic premature newborns simulating an inborn error of metabolism. Eur J Pediatr 1977; 127(1): 41–7.Google Scholar
Martin, HC, et al. Clinical whole-genome sequencing in severe early-onset epilepsy reveals new genes and improves molecular diagnosis. Hum Mol Genet 2014; 23(12): 3200–11.Google Scholar
Ohba, C, et al. GRIN1 mutations cause encephalopathy with infantile-onset epilepsy, and hyperkinetic and stereotyped movement disorders. Epilepsia 2015; 56(6): 841–8.Google Scholar
Nicita, F, et al. The genetics of monogenic idiopathic epilepsies and epileptic encephalopathies. Seizure 2012; 21(1): 311.Google Scholar
Tavyev Asher, YJ, Scaglia, F. Molecular bases and clinical spectrum of early infantile epileptic encephalopathies. Eur J Med Genet 2012; 55(5): 299306.Google Scholar
Pavone, P, et al. Ohtahara syndrome with emphasis on recent genetic discovery. Brain Dev 2012; 34(6): 459–68.Google Scholar
Dedek, K, et al. Neonatal convulsions and epileptic encephalopathy in an Italian family with a missense mutation in the fifth transmembrane region of KCNQ2. Epilepsy Res 2003; 54(1): 21–7.Google Scholar
Tang, S, et al. Encephalopathy and SCN1A mutations. Epilepsia 2011; 52(4): e2630.Google Scholar
Schanen, NC, et al. Neonatal encephalopathy in two boys in families with recurrent Rett syndrome. J Child Neurol 1998; 13(5): 229–31.Google Scholar
Mirzaa, GM, et al. CDKL5 and ARX mutations in males with early-onset epilepsy. Pediatr Neurol 2013; 48(5): 367–77.Google Scholar
Mencarelli, MA, et al. Novel FOXG1 mutations associated with the congenital variant of Rett syndrome. J Med Genet 2010; 47(1): 4953.Google Scholar
Byrd, DJ, et al. Neonatal pyruvate dehydrogenase deficiency with lipoate responsive lactic acidaemia and hyperammonaemia. Eur J Pediatr 1989; 148(6): 543–7.Google Scholar
Robinson, BH, et al. Disorders of pyruvate carboxylase and the pyruvate dehydrogenase complex. J Inherit Metab Dis 1996; 19(4): 452–62.Google Scholar
Bonnefont, JP, et al. Alpha-ketoglutarate dehydrogenase deficiency presenting as congenital lactic acidosis. J Pediatr 1992; 121(2): 255–8.Google Scholar
Haworth, JC, et al. Lactic acidosis in three sibs due to defects in both pyruvate dehydrogenase and alpha-ketoglutarate dehydrogenase complexes. Pediatrics 1976; 58(4): 564–72.Google Scholar
Kerrigan, JF, et al. Fumaric aciduria: clinical and imaging features. Ann Neurol 2000; 47(5): 583–8.Google Scholar
Wilcken, B, et al. Outcome of neonatal screening for medium-chain acyl-CoA dehydrogenase deficiency in Australia: a cohort study. Lancet 2007; 369(9555): 3742.Google Scholar
Duran, M, et al. 3-Hydroxydicarboxylic aciduria due to long-chain 3-hydroxyacyl-coenzyme A dehydrogenase deficiency associated with sudden neonatal death: protective effect of medium-chain triglyceride treatment. Eur J Pediatr 1991; 150(3): 190–5.Google Scholar
Stanley, CA, et al. Brief report: a deficiency of carnitine-acylcarnitine translocase in the inner mitochondrial membrane. N Engl J Med 1992; 327(1): 1923.Google Scholar
Bertrand, C, et al. Very long chain acyl-CoA dehydrogenase deficiency: identification of a new inborn error of mitochondrial fatty acid oxidation in fibroblasts. Biochim Biophys Acta 1993; 1180(3): 327–9.Google Scholar
Hintz, SR, et al. Early neonatal diagnosis of long-chain 3-hydroxyacyl coenzyme a dehydrogenase and mitochondrial trifunctional protein deficiencies. Mol Genet Metab 2002; 75(2): 120–7.Google Scholar
Ibdah, JA, et al. A fetal fatty-acid oxidation disorder as a cause of liver disease in pregnant women. N Engl J Med 1999; 340(22): 1723–31.Google Scholar
Agsteribbe, E, et al. A fatal, systemic mitochondrial disease with decreased mitochondrial enzyme activities, abnormal ultrastructure of the mitochondria and deficiency of heat shock protein 60. Biochem Biophys Res Commun 1993; 193(1): 146–54.Google Scholar
Briones, P, et al. A new case of multiple mitochondrial enzyme deficiencies with decreased amount of heat shock protein 60. J Inherit Metab Dis 1997; 20(4): 569–77.Google Scholar
Poggi, GM, et al. Fatal neonatal outcome in a case of muscular mitochondrial DNA depletion. J Inherit Metab Dis 2000; 23(7): 755–7.Google Scholar
Valnot, I, et al. Mutations of the SCO1 gene in mitochondrial cytochrome c oxidase deficiency with neonatal-onset hepatic failure and encephalopathy. Am J Hum Genet 2000; 67(5): 1104–9.Google Scholar
Seyda, A, et al. A novel syndrome affecting multiple mitochondrial functions, located by microcell-mediated transfer to chromosome 2p14-2p13. Am J Hum Genet 2001; 68(2): 386–96.Google Scholar
Mayr, JA, et al. Lipoic acid synthetase deficiency causes neonatal-onset epilepsy, defective mitochondrial energy metabolism, and glycine elevation. Am J Hum Genet 2011; 89(6): 792–7.Google Scholar
Huemer, M, et al. Clinical, morphological, biochemical, imaging and outcome parameters in 21 individuals with mitochondrial maintenance defect related to FBXL4 mutations. J Inherit Metab Dis 2015; 38(5): 905–14.Google Scholar
Nakano, Y, et al. Fatal case of mitochondrial DNA depletion with severe asphyxia in a newborn. Pediatr Int 2011; 53(2): 240–2.Google Scholar
Chitayat, D, et al. Brain dysgenesis and congenital intracerebral calcification associated with 3-hydroxyisobutyric aciduria. J Pediatr 1992; 121(1): 86–9.Google Scholar
Burlina, AB, Bonafe, L, Zacchello, F. Clinical and biochemical approach to the neonate with a suspected inborn error of amino acid and organic acid metabolism. Semin Perinatol 1999; 23(2): 162–73.Google Scholar
Livingston, JH, et al. Intracranial calcification in childhood: a review of aetiologies and recognizable phenotypes. Dev Med Child Neurol 2014; 56(7): 612–26.Google Scholar
Rice, G, et al. Clinical and molecular phenotype of Aicardi-Goutieres syndrome. Am J Hum Genet 2007; 81(4): 713–25.Google Scholar
Mochida, GH, et al. A homozygous mutation in the tight-junction protein JAM3 causes hemorrhagic destruction of the brain, subependymal calcification, and congenital cataracts. Am J Hum Genet 2010; 87(6): 882–9.Google Scholar
O’Driscoll, MC, et al. Recessive mutations in the gene encoding the tight junction protein occludin cause band-like calcification with simplified gyration and polymicrogyria. Am J Hum Genet 2010; 87(3): 354–64.Google Scholar
Escobar, LF, et al. Neonatal presentation of lethal neuromuscular glycogen storage disease type IV. J Perinatol 2012; 32(10): 810–3.Google Scholar
Tein, I. Neonatal metabolic myopathies. Semin Perinatol 1999; 23(2): 125–51.Google Scholar
Garcia, CA, et al. Neurological involvement in the Smith-Lemli-Opitz syndrome: clinical and neuropathological findings. Dev Med Child Neurol 1973; 15(1): 4855.Google Scholar
Siintola, E, et al. Cathepsin D deficiency underlies congenital human neuronal ceroid-lipofuscinosis. Brain 2006; 129(Pt 6): 1438–45.Google Scholar
Simpson, MA, et al. Infantile-onset symptomatic epilepsy syndrome caused by a homozygous loss-of-function mutation of GM3 synthase. Nat Genet 2004; 36(11): 1225–9.Google Scholar
Jain, P, et al. Menkes disease: an important cause of early onset refractory seizures. J Pediatr Neurosci 2014; 9(1): 11–6.Google Scholar
Kurian, MA, et al. The monoamine neurotransmitter disorders: an expanding range of neurological syndromes. Lancet Neurol 2011; 10(8): 721–33.Google Scholar

References

Hickling, KG, Henderson, SJ, Jackson, R. Low mortality associated with low volume pressure limited ventilation with permissive hypercapnia in severe adult respiratory distress syndrome. Intensive Care Med 1990; 16: 372–7.Google Scholar
Mariani, G, Cifuentes, J, Carlo, WA. Randomized trial of permissive hypercapnia in preterm infants. Pediatrics 1999; 104: 1082–8.Google Scholar
Ni Chonghaile, M, Higgins, B, Laffey, JG. Permissive hypercapnia: role in protective lung ventilatory strategies. Curr Opin Crit Care 2005; 11: 5662.Google Scholar
Carlo, WA. Permissive hypercapnia and permissive hypoxemia in neonates. J Perinatol 2007; 27:S6470.Google Scholar
Thome, UH, Ambalavanan, A. Permissive hypercapnia to decrease lung injury in ventilated preterm neonates. Semin Fetal Neonatal Med 2009; 14: 21–7.Google Scholar
Hernandez, MJ, Brennan, RW, Vannucci, RC, et al. Cerebral blood flow and oxygen consumption in the newborn dog. Am J Physiol 1978; 234: R209–15.Google Scholar
Reivich, M, Brann, AW, Shapiro, H. Reactivity of cerebral vessels to CO2 in the newborn rhesus monkey. Eur Neurol 1971; 6: 132–6.Google Scholar
Rosenberg, AA, Jones, MD, Traystman, RJ, et al. Response of cerebral blood flow to changes in pCO2 in fetal, newborn, and adult sheep. Am J Physiol 1982; 242: H862–6.Google Scholar
Griesen, G. Cerebral blood flow and energy metabolism in the newborn. Clin Perinatol 1997; 24: 531–46.Google Scholar
Dietz, V, Wolf, M, Keel, M, et al. CO2 reactivity of the cerebral hemoglobin concentration in healthy term newborns measured by near infrared spectrophotometry. Biol Neonate 1999; 75: 8590.Google Scholar
Jayasinghe, D, Gill, AB, Levene, MI. CBF reactivity in hypotensive and normotensive preterm infants. Pediatr Res 2003; 54: 848–53.Google Scholar
Kaiser, JR, Gauss, CH, Williams, DK. The effects of hypercapnia on cerebral autoregulation in ventilated very low birth weight infants. Pediatr Res 2005; 58: 931–5.Google Scholar
Kenny, JD, Garcia-Prats, JA, Hilliard, JL, et al. Hypercarbia at birth:a possible role in the pathogenesis of intraventricular hemorrhage. Pediatrics 1978; 62: 465–7.Google Scholar
Levene, MI, Fawer, CL, Lamont, RF. Risk factors in the development of intraventricular haemorrhage in the preterm neonate. Arch Dis Child 1982; 57: 410–17.Google Scholar
Kaiser, JR, Gauss, CH, Pont, MJ, et al. Hypercapnia during the first 3 days of life is associated with severe intraventricular hemorrhage in very low birth weight infants. J Perinatol 2006; 26: 279–85.Google Scholar
Kaiser, JR, Gauss, CH, Williams, DK. Surfactant administration acutely affects cerebral and systemic hemodynamics and gas exchange in very low birth weight infants. J Pediatr 2004; 144: 809–14.Google Scholar
Lightbum, MH, Gauss, CH, Williams, DK, Kaiser, JR. Observational study of cerebral hemodynamics during dopamine treatment in hypotensive ELBW infants on the first day of life. J Perinatol 2013; 33: 698702.Google Scholar
Zayek, MM, Alrifai, W, Whitehurst, RM Jr, et al. Acidemia versus hypercapnia and risk for severe intraventricular hemorrhage. Am J Perinatol 2014; 31: 345–52.Google Scholar
Gleason, CA, Short, BL, Jones, MD Jr. Cerebral blood flow and metabolism during and after prolonged hypocapnia in newborn lambs. J Pediatr 1989; 115: 309–14.Google Scholar
Fabres, J, Carlo, WA, Phillips, V, et al. Both extremes of arterial carbon dioxide pressure and the magnitude of fluctuations in arterial carbon dioxide pressure are associated with severe intraventricular hemorrhage in preterm infants. J Pediatr 2007; 119: 299305.Google Scholar
Noori, S, Anderson, M, Soleymani, S, Seri, I. Effect of carbon dioxide on cerebral blood flow velocity in preterm infants during postnatal transition. Acta Paediatr 2014; 103:e344–9.Google Scholar
Vannucci, RC, Towfighi, J, Heitjan, DF, et al. Carbon dioxide protects the perinatal rat brain from hypoxic–ischemic damage. Pediatrics 1995; 95: 868–74.Google Scholar
Vannucci, RC, Towfighi, J, Brucklacher, RM, et al. Effect of extreme hypercapnia on hypoxic–ischemic brain damage in the immature rat. Pediatr Res 2001; 49: 799803.Google Scholar
Komori, M, Takada, K, Tomizawa, Y, et al. Permissive range of hypercapnia for improved peripheral microcirculation and cardiac output in rabbits. Crit Care Med 2007; 35: 2171–5.Google Scholar
Xu, L, Glassford, AJM, Giaccia, AJ, et al. Acidosis reduces neuronal apoptosis. NeuroReport 1998; 9: 875–9.Google Scholar
Laffey, JG, Tanaka, M, Engelberts, D, et al. Therapeutic hypercapnia reduces pulmonary and systemic injury following in vivo lung reperfusion. Am J Respir Crit Care Med 2000; 162: 2287–94.Google Scholar
Laffey, JG, Engelberts, D, Kavanagh, BP. Buffering hypercapnic acidosis worsens acute lung injury. Am J Respir Crit Care Med 2000; 161: 141–6.Google Scholar
Fujita, M, Asanuma, H, Hirata, A, et al. Prolonged transient acidosis during early reperfusion contributes to the cardioprotective effects of postconditioning. Am J Physiol Heart Circ Physiol 2007; 292: H2004–8.Google Scholar
Carlo, WA, Stark, AR, Wright, LL, et al. Minimal ventilation to prevent bronchopulmonary dysplasia in extremely low-birth-weight infants. J Pediatr 2002; 141: 370–5.Google Scholar
Thome, UH, Carroll, W, Wu, T-J, et al. Outcome of extremely preterm infants randomized at birth to different PaCO2 targets during the first seven days of life. Biol Neonate 2006; 90: 218–25.Google Scholar
Miller, JD, Carlo, WA. Safety and effectiveness of permissive hypercapnia in the preterm infant. Curr Opin Pediatr 2007; 19: 142–4.Google Scholar
Ou, X, Glasier, CM, Ramakrishnaiah, RH, et al. Diffusion tensor imaging in extremely low birth weight infants managed with hypercapnic vs. normocapnic ventilation. Pediatr Radiol 2014; 44: 980–6.Google Scholar
van Kaam, AH, De Jaegere, AP, Rimensberger, PC, et al. Incidence of hypo- and hypercapnia in a cross-sectional European cohort of ventilated newborn infants. Arch Dis Child Fetal Neonatal Ed 2013; 98:F323–6.Google Scholar
American College of Obstetricians and Gynecologists, Committee on Obstetric Practice. Umbilical cord blood gas and acid-base analysis. Obstet Gynecol 2006; 108: 1319–22.Google Scholar
Evans, OB. Lactic acidosis in childhood, part I. Pediatr Neurol 1985; 1: 325–8.Google Scholar
Bar, A, Riskin, A, Iancu, T, et al. A newborn infant with protracted diarrhea and metabolic acidosis. J Pediatr 2007; 150: 198201.Google Scholar
Debray, FG, Lamber, M, Chevalier, I, et al. Long-term outcome and clinical spectrum of 73 pediatric patients with mitochondrial diseases. Pediatrics 2007; 119: 722–33.Google Scholar
Bhati, RS, Sheridan, BC, Mill, MR, et al. Heart transplantation for progressive cardiomyopathy as a manifestation of MELAS syndrome. J Heart Lung Transplant 2005; 24: 2286–9.Google Scholar
Schurr, A. Lactate: the ultimate cerebral oxidative energy substrate? J Cerebral Blood Flow Metabol 2006; 26: 142–52.Google Scholar
Bergersen, LH. Is lactate food for neurons? Comparison of monocarboxylate transporter subtypes in brain and muscle. Neuroscience 2007; 145:1119.Google Scholar
Boumezbeur, F, Petersen, KF, Cline, GW, et al. The contribution of blood lactate to brain energy metabolism in humans measured by dynamic 13C nuclear magnetic resonance spectroscopy. J Neurosci 2010; 30: 13983–191.Google Scholar
Overgaard, M, Rasmussen, P, Bohm, AM, et al. Hypoxia and exercise provoke both lactate release and lactate oxidation by the human brain. FASEB J 2012; 26: 3012–20.Google Scholar
Pirchl, M, Marksteiner, J, Humpel, C. Effects of acidosis on brain capillary endothelial cells and cholinergic neurons: relevance to vascular dementia and Alzheimer’s disease. Neurol Res 2006; 28: 657–64.Google Scholar
Gilbert, E, Tang, JM, Ludvig, N, et al. Elevated lactate suppresses neuronal firing in vivo and inhibits glucose metabolism in hippocampal slice cultures. Brain Res 2006; 1117: 213–23.Google Scholar
Ammari, AN, Schulze, KF. Uses and abuses of sodium bicarbonate in the neonatal intensive care unit. Curr Opin Pediatr 2002; 14: 151–6.Google Scholar
Murki, S, Kumar, P, Lingappa, L, et al. Effect of a single dose of sodium bicarbonate given during neonatal resuscitation at birth on the acid-base status on first day of life. J Perinatol 2004; 24: 696–9.Google Scholar
Lokesh, L, Kumar, P, Murki, S, et al. A randomized, controlled trial of sodium bicarbonate in neonatal resuscitation: effect on immediate outcome. Resuscitation 2000; 60: 219–23.Google Scholar
Beveridge, CJE, Wilkinson, AR. Sodium bicarbonate infusion during resuscitation of infants at birth. Cochrane Database Syst Rev 2006; 1:CD004864.Google Scholar
Mintzer, JP, Parvez, B, Alpan G, LaGamma, EF. Effects of sodium bicarbonate correction of metabolic acidosis on regional tissue oxygenation in very low birth weight neonates. J Perinatol 2015; 35(8): 601–6.Google Scholar
Laffey, JG, Kavanagh, BP. Hypocapnia. N Engl J Med 2002; 347:4353.Google Scholar
Cassin, S, Dawes, GS, Mott, JC, et al. The vascular resistance of the foetal and newly ventilated lung of the lamb. J Physiol 1964; 171:6179.Google Scholar
Rudolph, AM, Yuan, S. Response of the pulmonary vasculature to hypoxia and Hþ ion concentration changes. J Clin Invest 1966; 45:399411.Google Scholar
Peckham, GJ, Fox, WW. Physiologic factors affecting pulmonary artery pressure in infants with persistent pulmonary hypertension. J Pediatr 1978; 93: 1005–10.Google Scholar
Fox, WW, Duara, S. Persistent pulmonary hypertension in the neonate: diagnosis and management. J Pediatr 1983; 103: 505–14.Google Scholar
Marron, MJ, Crisafi, MA, Driscoll, JM, et al. Hearing and neurodevelopmental outcome in survivors of persistent pulmonary hypertension of the newborn. Pediatrics 1992; 90: 392–6.Google Scholar
Brett, C, Dekle, M, Leonard, CH, et al. Developmental follow-up of hyperventilated neonates: preliminary observations. Pediatrics 1981; 68: 588–91.Google Scholar
Ferrara, B, Johnson, DE, Chang, PN, et al. Efficacy and neurologic outcome of profound hypocapneic alkalosis for the treatment of persistent pulmonary hypertension in infancy. J Pediatr 1984; 105: 457–61.Google Scholar
Bernbaum, JC, Russell, P, Sheridan, PH, et al. Long-term follow-up of newborns with persistent pulmonary hypertension. Crit Care Med 1984; 12: 579–83.Google Scholar
Ballard, RA, Leonard, CH. Developmental follow-up of infants with persistent pulmonary hypertension of the newborn. Clin Perinatol 1984; 11: 737–44.Google Scholar
Leavitt, AM, Watchko, JF, Bennett, FC, et al. Neurodevelopmental outcome following persistent pulmonary hypertension of the neonate. J Perinatol 1987; 7: 288–91.Google Scholar
Bifano, EM, Pfannenstsiel, A. Duration of hyperventilation and outcome in infants with persistent pulmonary hypertension. Pediatrics 1988; 81: 657–61.Google Scholar
Cohen, RS, Stevenson, D, Malachowski, N, et al. Late morbidity among survivors on respiratory failure treated with tolazoline. J Pediatr 1980; 97: 644–7.Google Scholar
Graziani, LJ, Baumgart, S, Desai, S, et al. Clinical antecedents of neurologic and audiologic abnormalities in survivors of extracorporeal membrane oxygenation. J Child Neurol 1997; 12: 415–22.Google Scholar
Lyrene, Rk, Welch, KA, Godoy, G, et al. Alkalosis attenuates hypoxic pulmonary vasoconstriction in neonatal lambs. Pediatr Res 1985; 19: 1268–71.Google Scholar
Schreiber, MD, Heymann, MA, Soifer, SJ. Increased arterial pH, not decreased PaCO2, attenuates hypoxia-induced pulmonary vasoconstriction in newborn lambs. Pediatr Res 1986; 20: 113–17.Google Scholar
Chang, AC, Zucker, HA, Hickey, PR, et al. Pulmonary vascular resistance in infants after cardiac surgery: role of carbon dioxide and hydrogen ion. Crit Care Med 1995; 23: 568–74.Google Scholar
Lee, KJ, Hernandez, G, Gordon, JB. Hypercapnic acidosis and compensated hypercapnia in control and pulmonary hypertensive piglets. Pediatr Pulmonol 2003; 36:94101.Google Scholar
Walsh-Sukys, MC, Tyson, JE, Wright, LL, et al. Persistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomes. Pediatrics 2000; 105:1420.Google Scholar
Hansen, NB, Nowicki, PT, Miller, RR, et al. Alterations in cerebral blood flow and oxygen consumption during prolonged hypocarbia. Pediatr Res 1986; 20: 147–50.Google Scholar
Kusada, S, Shisida, N, Miyagi, N, et al. Cerebral blood flow during treatment for pulmonary hypertension. Arch Dis Child Fetal Neonatal Ed 1999; 80:F30–3.Google Scholar
Menke, J, Michel, E, Rabe, H, et al. Simultaneous influence of blood pressure, PCO2, and PO2 on cerebral blood flow velocity in preterm infants of less than 33 weeks’ gestation. Pediatr Res 1993; 24: 173–7.Google Scholar
Kaiser, JR, Gauss, CH, Williams, DK. The effects of hypercapnia on cerebral autoregulation in ventilated very low birth weight infants. Pediatr Res 2005; 58: 931–5.Google Scholar
Soul, JS, Hammer, PE, Tsuji, P, et al. Fluctuating pressure-passivity is common in the cerebral circulation of sick premature infants. Pediatr Res 2007; 61: 467–73.Google Scholar
Kennedy, C, Sakurada, O, Shinohara, M, et al. Local cerebral glucose utilization in the newborn macaque monkey. Ann Neurol 1982; 12: 333–40.Google Scholar
Vannucci, RC, Vannucci, SJ. Perinatal brain metabolism. In Polin, RA, Fox, WW, Abman, SH, eds., Fetal and Neonatal Physiology, 3rd edn. Philadelphia: Saunders, 2004: 1713–25.Google Scholar
Graham, EM, Apostolou, M, Mishra, OP, et al. Modification of the N-methyl-d-aspartate (NMDA) receptor in the brain of newborn piglets following hyperventilation induced ischemia. Neurosci Lett 1996; 218:2932.Google Scholar
Fritz, KI, Zubrow, AB, Ashraf, QM, et al. The effect of moderate hypocapnic ventilation on nuclear Ca2+-ATPase activity, nuclear Ca2+ flux, and Ca2+/calmodulin kinase IV activity in the cerebral cortex of newborn piglets. Neurochem Res 2004; 29: 791–6.Google Scholar
Naulty, CM, Weiss, IP, Herer, GR. Progressive sensorineural hearing loss in survivors of persistent fetal circulation. Ear Hear 1986; 7: 74–7.Google Scholar
Hendricks-Munoz, KD, Walton, JP. Hearing loss in infants with persistent fetal circulation. Pediatrics 1988; 81: 650–6.Google Scholar
Calvert, SA, Hoskins, EM, Fong, KW, et al. Etiological factors associated with the development of periventricular leukomalacia. Acta Paediatr Scand 1987; 76: 254–9.Google Scholar
Graziani, LJ, Spitzer, AR, Mitchell, DG, et al. Mechanical ventilation in preterm infants: neurosonographic and developmental studies. Pediatrics 1992; 90: 515–22.Google Scholar
Wiswell, TE, Graziani, LJ, Kornhauser, MS, et al. Effects of hypocarbia on the development of cystic periventricular leukomalacia in premature infants treated with high-frequency jet ventilation. Pediatrics 1996; 98: 918–24.Google Scholar
Shankaran, S, Langer, JC, Kazzi, SN, et al. Cumulative index of exposure to hypocarbia and hyperoxia as risk factors for periventricular leukomalacia in low birth weight infants. Pediatrics 2006; 118: 1654–9.Google Scholar
HiFO Study Group. Randomized study of high-frequency oscillatory ventilation in infants with severe respiratory distress syndrome. J Pediatr 1993; 122: 609–19.Google Scholar
Wiswell, TE, Graziani, LJ, Kornhauser, MS, et al. High-frequency jet ventilation in the early management of respiratory distress syndrome is associated with a greater risk for adverse outcomes. Pediatrics 1996; 98: 1035–43.Google Scholar
Keszler, M, Modanlou, HD, Brudno, DS, et al. Multicenter controlled clinical trial of high-frequency jet ventilation in preterm infants with uncomplicated respiratory distress syndrome. Pediatrics 1997; 100: 593–9.Google Scholar
Klinger, G, Beyene, J, Shah, P, et al. Do hyperoxaemia and hypocapnia add to the risk of brain injury after intrapartum asphyxia? Arch Dis Child Fetal Neonatal Ed 2005; 90:F4952.Google Scholar
Vento, M, Asensi, M, Sastre, J, et al. Oxidative stress in asphyxiated term infants resuscitated with 100% oxygen. J Pediatr 2003; 142: 240–6.Google Scholar
Curley, G, Kavanagh, BP, Laffey, JG. Hypocapnia and the injured brain: more harm than benefit. Crit Care Med 2010; 38: 1348–59.Google Scholar
Laski, ME, Sabatini, S. Metabolic alkalosis, bedside and bench. Semin Nephrol 2006; 26: 404–21.Google Scholar
Shaer, AJ. Inherited primary renal tubular hypokalemic alkalosis: a review of Gitelman and Bartter syndromes. Am J Med Sci 2001; 322: 316–32.Google Scholar
Naesens, M, Steels, P, Verberckmoes, R, et al. Bartter’s and Gitelman’s syndromes: from gene to clinic. Nephron Physiol 2004; 96:6578.Google Scholar
Kagalwalla, AF. Congenital chloride diarrhea:a study in Arab children. J Clin Gastroenterol 1994; 19:3640.Google Scholar
Hihnala, S, Höglund, P, Lammi, L, et al. Long-term clinical outcome in patients with congenital chloride diarrhea. J Pediatr Gastroenterol Nutr 2006; 42: 369–75.Google Scholar
Sasse, S, Kribs, A, Vierzig, A, et al. A staged protocol for the treatment of persistent pulmonary hypertension of the newborn. Klin Padiatr 1997; 209: 301–7.Google Scholar
Adeva-Andany, MM, Fernandez-Fernandez, C, Mourino-Bayolo, D, et al. Sodium bicarbonate therapy in patients with metabolic acidosis. Sci World J 2014; available at http://dx.doi.org/10.1155/2014/627673.Google Scholar

References

Walsh-Sukys, MC, Tyson, JE, Wright, LL, et al. Persistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomes. Pediatrics 2000; 105: 1420Google Scholar
Kumar, VH, Hutchison, AA, Lakshminrusimha, S, et al. Characteristics of pulmonary hypertension in preterm neonates. J Perinatol 2007; 27: 214–9.Google Scholar
Aikio, O, Metsola, J, Vuolteenaho, R, et al. Transient defect in nitric oxide generation after rupture of fetal membranes and responsiveness to inhaled nitric oxide in very preterm infants with hypoxic respiratory failure. J Pediatr 2012; 161: 397403.Google Scholar
Cerro, MJ, Abman, S, Diaz, G, et al. A consensus approach to the classification of pediatric pulmonary hypertensive vascular disease: report from the PVRI Pediatric Taskforce, Panama 2011. Pulm Circ 2011; 1: 286–98.Google Scholar
Ivy, DD, Abman, SH, Barst, RJ, et al. Pediatric pulmonary hypertension. J Am Coll Cardiol 2013; 62: D117–26.Google Scholar
Teitel, DF, Iwamoto, HS, Rudolph, AM. Changes in the pulmonary circulation during birth-related events. Pediatr Res 1990; 27: 372–8.Google Scholar
Bloch, KD, Filippov, G, Sanchez, LS, et al. Pulmonary soluble guanylate cyclase, a nitric oxide receptor, is increased during the perinatal period. Am J Physiol 1997; 272: L400–6.Google Scholar
Hanson, KA, Ziegler, JW, Rybalkin, SD, et al. Chronic pulmonary hypertension increases fetal lung cGMP phosphodiesterase activity. Am J Physiol 1998; 275: L931–41.Google Scholar
Sanchez, LS, de la Monte, SM, Filippov, G, et al. Cyclic-GMP-binding, cyclic-GMP-specific phosphodiesterase (PDE5) gene expression is regulated during rat pulmonary development. Pediatr Res 1998; 43: 163–8.Google Scholar
Hernandez-Diaz, S, Van Marter, LJ, Werler, MM, et al. Risk factors for persistent pulmonary hypertension of the newborn. Pediatrics 2007; 120: e272–82.Google Scholar
Delaney, C, Gien, J, Grover, TR, et al. Pulmonary vascular effects of serotonin and selective serotonin reuptake inhibitors in the late-gestation ovine fetus. Am J Physiol Lung Cell Mol Physiol 2011; 301: L937–44.Google Scholar
Delaney, C, Gien, J, Roe, G, et al. Serotonin contributes to high pulmonary vascular tone in a sheep model of persistent pulmonary hypertension of the newborn. Am J Physiol Lung Cell Mol Physiol 2013; 304: L894901.Google Scholar
Belik, J. Fetal and neonatal effects of maternal drug treatment for depression. Semin Perinatol 2008; 32: 350–4.Google Scholar
Fornaro, E, Li, D, Pan, J, Belik, J. Prenatal exposure to fluoxetine induces fetal pulmonary hypertension in the rat. Am J Respir Crit Care Med 2007; 176: 1035–40.Google Scholar
Hybrechts, KF, Bateman, BT, Palmsten, K, et al. Antidepressant use late in pregnancy and risk of persistent pulmonary hypertension of the newborn. JAMA 2015; 313: 2142–51.Google Scholar
Kieler, H, Artama, M, Engeland, A, et al. Selective serotonin reuptake inhibitors during pregnancy and risk of persistent pulmonary hypertension in the newborn: population based cohort study from the five Nordic countries. BMJ 2012; 344: d8012.Google Scholar
Grigoriadis, S, Vonderporten, EH, Mamisashvili, L, et al. Prenatal exposure to antidepressants and persistent pulmonary hypertension of the newborn: systematic review and meta-analysis. BMJ (Clin Res Ed) 2014; 348: f6932.Google Scholar
Wilson, KL, Zelig, CM, Harvey, JP, et al. Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am J Perinatol 2011; 28: 1924.Google Scholar
Andrade, SE, McPhillips, H, Loren, D, et al. Antidepressant medication use and risk of persistent pulmonary hypertension of the newborn. Pharmacoepidemiol Drug Saf 2009; 18: 246–52.Google Scholar
Lim, K, Sanders, A, Brain, U, et al. Third trimester fetal pulmonary artery Doppler blood flow velocity characteristics following prenatal selective serotonin reuptake inhibitor (SSRI) exposure. Early Hum Dev 2012; 88: 609–15.Google Scholar
Talati, AJ, Salim, MA, Korones, SB. Persistent pulmonary hypertension after maternal naproxen ingestion in a term newborn: a case report. Am J Physiol 2000; 17: 6971.Google Scholar
Wild, LM, Nickerson, PA, Morin, FC 3rd. Ligating the ductus arteriosus before birth remodels the pulmonary vasculature of the lamb. Pediatr Res 1989; 25: 251–7.Google Scholar
Van Marter, LJ, Hernandez-Diaz, S, Werler, MM, et al. Nonsteroidal antiinflammatory drugs in late pregnancy and persistent pulmonary hypertension of the newborn. Pediatrics 2013; 131: 7987.Google Scholar
Shah, PS, Hellmann, J, Adatia, I. Clinical characteristics and follow up of Down syndrome infants without congenital heart disease who presented with persistent pulmonary hypertension of newborn. J Perinat Med 2004; 32: 168–70.Google Scholar
Cua, CL, Blankenship, A, North, AL, et al. Increased incidence of idiopathic persistent pulmonary hypertension in Down syndrome neonates. Pediatr Cardiol 2007; 28: 250–4.Google Scholar
Weijerman, ME, van Furth, AM, van der Mooren, MD, et al. Prevalence of congenital heart defects and persistent pulmonary hypertension of the neonate with Down syndrome. Eur J Pediatr 2010; 169: 1195–9.Google Scholar
Southgate, WM, Annibale, DJ, Hulsey, TC, Purohit, DM. International experience with trisomy 21 infants placed on extracorporeal membrane oxygenation. Pediatrics 2001; 107: 549–52.Google Scholar
Byers, HM, Dagle, JM, Klein, JM, et al. Variations in CRHR1 are associated with persistent pulmonary hypertension of the newborn. Pediatr Res 2012; 71: 162–7.Google Scholar
Chandrasekar, I, Eis, A, Konduri, GG. Betamethasone attenuates oxidant stress in endothelial cells from fetal lambs with persistent pulmonary hypertension. Pediatr Res 2008; 63: 6772.Google Scholar
Perez, M, Lakshminrusimha, S, Wedgwood, S, et al. Hydrocortisone normalizes oxygenation and cGMP regulation in lambs with persistent pulmonary hypertension of the newborn. Am J Physiol Lung Cell Mol Physiol 2011; 302(6): L595603.Google Scholar
Lapointe, A, Barrington, KJ. Pulmonary hypertension and the asphyxiated newborn. J Pediatr 2011; 158: e1924.Google Scholar
Cornish, JD, Dreyer, GL, Snyder, GE, et al. Failure of acute perinatal asphyxia or meconium aspiration to produce persistent pulmonary hypertension in a neonatal baboon model. Am J Obstet Gynecol 1994; 171: 43–9.Google Scholar
Murphy, JD, Vawter, GF, Reid, LM. Pulmonary vascular disease in fatal meconium aspiration. J Pediatr 1984; 104: 758–62.Google Scholar
Toubas, PL, Hof, RP, Heymann, MA, Rudolph, AM. Effects of hypothermia and rewarming on the neonatal circulation. Arch Fr Pediatr 1978; 35: 8492.Google Scholar
Thoresen, M. Hypothermia after perinatal asphyxia: selection for treatment and cooling protocol. J Pediatr 2011; 158: e45–9.Google Scholar
Sarkar, S, Barks, JD, Bhagat, I, et al. Pulmonary dysfunction and therapeutic hypothermia in asphyxiated newborns: whole body versus selective head cooling. Am J Perinatol 2009; 26: 265–70.Google Scholar
Shankaran, S, Laptook, AR, Pappas, A, et al. Effect of depth and duration of cooling on deaths in the NICU among neonates with hypoxic ischemic encephalopathy: a randomized clinical trial. JAMA 2014; 312: 2629–39.Google Scholar
Dodson, RB, Morgan, M, Galambos, C, et al. Chronic intrauterine pulmonary hypertension increases main pulmonary artery stiffness and adventitial remodeling in fetal sheep. Am J Physiol Lung Cell Mol Physiol 2014; 307(11): L822–8.Google Scholar
Stenmark, KR, Davie, N, Frid, M, et al. Role of the adventitia in pulmonary vascular remodeling. Physiology 2006; 21: 134–45.Google Scholar
Stenmark, KR, Yeager, ME, El Kasmi, KC, et al. The adventitia: essential regulator of vascular wall structure and function. Annu Rev Physiol 2013; 75: 2347.Google Scholar
Soifer, SJ, Kaslow, D, Roman, C, Heymann, MA. Umbilical cord compression produces pulmonary hypertension in newborn lambs: a model to study the pathophysiology of persistent pulmonary hypertension in the newborn. J Dev Physiol 1987; 9: 239–52.Google Scholar
Villaneuva, ME, Zaher, FM, Svinarich, DM, Konduri, GG. Decreased gene expression of endothelial nitric oxide synthase in newborns with persistent pulmonary hypertension. Pediatr Res 1998; 44: 338–43.Google Scholar
Zagariya, A, Doherty, J, Bhat, R, et al. Elevated immunoreactive endothelin-1 levels in newborn rabbit lungs after meconium aspiration. Pediatr Crit Care Med 2002; 3: 297302.Google Scholar
Soukka, H, Jalonen, J, Kero, P, Kaapa, P. Endothelin-1, atrial natriuretic peptide and pathophysiology of pulmonary hypertension in porcine meconium aspiration. Acta Paediatr 1998; 87: 424–8.Google Scholar
Soukka, H, Viinikka, L, Kaapa, P. Involvement of thromboxane A2 and prostacyclin in the early pulmonary hypertension after porcine meconium aspiration. Pediatr Res 1998; 44: 838–42.Google Scholar
Davis, AS, RW, D., Van Meurs, KP. Persistent pulmonary hypertension of the newborn. In Stevenson, DK, Benitz, WE, Sunshine, P, et al., eds., Fetal and Neonatal Brain Injury, 4th edn. Cambridge University Press, 2009: 419–42.Google Scholar
Steinhorn, RH, Russell, JA, Morin, FC 3rd. Disruption of cGMP production in pulmonary arteries isolated from fetal lambs with pulmonary hypertension. Am J Physiol 1995; 268: H1483–9.Google Scholar
de Buys Roessingh, A, Fouquet, V, Aigrain, Y, et al. Nitric oxide activity through guanylate cyclase and phosphodiesterase modulation is impaired in fetal lambs with congenital diaphragmatic hernia. J Pediatr Surg 2011; 46: 1516–22.Google Scholar
Chester, M, Seedorf, G, Tourneux, P, et al. Cinaciguat, a soluble guanylate cyclase activator, augments cGMP after oxidative stress and causes pulmonary vasodilation in neonatal pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2011; 301: L755–64.Google Scholar
Kinsella, JP, Truog, WE, Walsh, WF, et al. Randomized, multicenter trial of inhaled nitric oxide and high-frequency oscillatory ventilation in severe, persistent pulmonary hypertension of the newborn. J Pediatr 1997; 131: 5562.Google Scholar
Wung, JT, James, LS, Kilchevsky, E, James, E. Management of infants with severe respiratory failure and persistence of the fetal circulation, without hyperventilation. Pediatrics 1985; 76: 488–94.Google Scholar
Lotze, A, Mitchell, BR, Bulas, DI, et al. Multicenter study of surfactant (beractant) use in the treatment of term infants with severe respiratory failure. Survanta in Term Infants Study Group. J Pediatr 1998; 132: 40–7.Google Scholar
Konduri, GG, Sokol, GM, Van Meurs, KP, et al. Impact of early surfactant and inhaled nitric oxide therapies on outcomes in term/late preterm neonates with moderate hypoxic respiratory failure. J Perinatol 2013; 33: 944–9.Google Scholar
Walsh-Sukys, MC, Tyson, JE, Wright, LL, et al. Persistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomes. Pediatrics 2000; 105: 1420.Google Scholar
Laffey, JG, Engelberts, D, Kavanaugh, BP. Inurious effects of hypocapnic alkalosis in the isolated lung. Am J Respir Crit Care Med 2000; 162: 399405.Google Scholar
Marron, MJ, Crisafi, MA, Driscoll, JM Jr, et al. Hearing and neurodevelopmental outcome in survivors of persistent pulmonary hypertension of the newborn. Pediatrics 1992; 90: 392–6.Google Scholar
Hendricks-Munoz, KD, Walton, JP. Hearing loss in infants with persistent fetal circulation. Pediatrics 1988; 81: 650–6.Google Scholar
Lakshminrusimha, S, Russell, JA, Steinhorn, RH, et al. Pulmonary hemodynamics in neonatal lambs resuscitated with 21%, 50%, and 100% oxygen. Pediatr Res 2007; 62: 313–8.Google Scholar
Lakshminrusimha, S, Swartz, DD, Gugino, SF, et al. Oxygen concentration and pulmonary hemodynamics in newborn lambs with pulmonary hypertension. Pediatr Res 2009; 66: 539–44.Google Scholar
Clark, RH, Kueser, TJ, Walker, MW, et al. Low-dose nitric oxide therapy for persistent pulmonary hypertension of the newborn. Clinical Inhaled Nitric Oxide Research Group. N Engl J Med 2000; 342: 469–74.Google Scholar
Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in full-term and nearly full-term infants with hypoxic respiratory failure. N Engl J Med 1997; 336: 597604.Google Scholar
Neonatal Inhaled Nitric Oxide Study Group. Inhaled nitric oxide in term and near-term infants: neurodevelopmental follow-up of the neonatal inhaled nitric oxide study group (NINOS). J Pediatr 2000; 136: 611–7.Google Scholar
Clark, RH, Huckaby, JL, Kueser, TJ, et al. Low-dose nitric oxide therapy for persistent pulmonary hypertension: 1-year follow-up. J Perinatol 2003; 23: 300–3.Google Scholar
Farrow, KN, Lakshminrusimha, S, Czech, L, et al. SOD and inhaled nitric oxide normalize phosphodiesterase 5 expression and activity in neonatal lambs with persistent pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2010; 299: L109–16.Google Scholar
Farrow, KN, Groh, BS, Schumacker, PT, et al. Hyperoxia increases phosphodiesterase 5 expression and activity in ovine fetal pulmonary artery smooth muscle cells. Circ Res 2008; 102: 226–33.Google Scholar
Farrow, KN, Lee, KJ, Perez, M, et al. Brief hyperoxia increases mitochondrial oxidation and increases phosphodiesterase 5 activity in fetal pulmonary artery smooth muscle cells. Antioxid Redox Signal 2012; 17: 460–70.Google Scholar
Baquero, H, Soliz, A, Neira, F, et al. Oral sildenafil in infants with persistent pulmonary hypertension of the newborn: a pilot randomized blinded study. Pediatrics 2006; 117: 1077–83.Google Scholar
Steinhorn, RH, Kinsella, JP, Pierce, C, et al. Intravenous sildenafil in the treatment of neonates with persistent pulmonary hypertension. J Pediatr 2009; 155: 841–7.Google Scholar
Keller, RL, Moore, P, Teitel, D, et al. Abnormal vascular tone in infants and children with lung hypoplasia: findings from cardiac catheterization and the response to chronic therapy. Pediatr Crit Care Med 2006; 7: 589–94.Google Scholar
Brannon, TS, MacRitchie, AN, Jaramillo, MA, et al. Ontogeny of cyclooxygenase-1 and cyclooxygenase-2 gene expression in ovine lung. Am J Physiol 1998; 274: L6671.Google Scholar
Brannon, TS, North, AJ, Wells, LB, Shaul, PW. Prostacyclin synthesis in ovine pulmonary artery is developmentally regulated by changes in cyclooxygenase-1 gene expression. J Clin Invest 1994; 93: 2230–5.Google Scholar
Christman, BW, McPherson, CD, Newman, JH, et al. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med 1992; 327: 70–5.Google Scholar
Lakshminrusimha, S, Porta, NF, Farrow, KN, et al. Milrinone enhances relaxation to prostacyclin and iloprost in pulmonary arteries isolated from lambs with persistent pulmonary hypertension of the newborn. Pediatr Crit Care Med 2009; 10: 106–12.Google Scholar
Walmrath, D, Schneider, T, Schermuly, R, et al. Direct comparison of inhaled nitric oxide and aerosolized prostacyclin in acute respiratory distress syndrome. Am J Respir Crit Care Med 1996; 153: 991–6.Google Scholar
Bindl, L, Fahnenstich, H, Peukert, U. Aerosolised prostacyclin for pulmonary hypertension in neonates. Arch Dis Child Fetal Neonatal Ed 1994; 71: F214–6.Google Scholar
Soditt, V, Aring, C, Groneck, P. Improvement of oxygenation induced by aerosolized prostacyclin in a preterm infant with persistent pulmonary hypertension of the newborn. Intensive Care Med 1997; 23: 1275–8.Google Scholar
Olmsted, K, Oluola, O, Parthiban, A, Raghuveer, T. Can inhaled prostacyclin stimulate surfactant in ELBW infants? J Perinatol 2007; 27: 724–6.Google Scholar
Kelly, LK, Porta, NF, Goodman, DM, et al. Inhaled prostacyclin for term infants with persistent pulmonary hypertension refractory to inhaled nitric oxide. J Pediatr 2002; 141: 830–2.Google Scholar
Porta, NF, Steinhorn, RH. Pulmonary vasodilator therapy in the NICU: inhaled nitric oxide, sildenafil, and other pulmonary vasodilating agents. Clin Perinatol 2012; 39: 149–64.Google Scholar
Ferdman, DJ, Rosenzweig, EB, Zuckerman, WA, Krishnan, U. Subcutaneous treprostinil for pulmonary hypertension in chronic lung disease of infancy. Pediatrics 2014; 134: e274–8.Google Scholar
Luscher, TF. Endothelin: systemic arterial and pulmonary effects of a new peptide with potent biologic properties. Am Rev Respir Dis 1992; 146: S5660.Google Scholar
Perreault, T, Coceani, F. Endothelin in the perinatal circulation. Can J Physiol Pharmacol 2003; 81: 644–53.Google Scholar
Mann, J, Farrukh, IS, Michael, JR. Mechanisms by which endothelin 1 induces pulmonary vasoconstriction in the rabbit. J Appl Physiol 1991; 71: 410–6.Google Scholar
Ivy, DD, Kinsella, JP, Abman, SH. Physiologic characterization of endothelin A and B receptor activity in the ovine fetal pulmonary circulation. J Clin Invest 1994; 93: 2141–8.Google Scholar
Ivy, DD, Le Cras, TD, Horan, MP, Abman, SH. Chronic intrauterine pulmonary hypertension increases preproendothelin-1 and decreases endothelin B receptor mRNA expression in the ovine fetal lung. Chest 1998; 114: 65S.Google Scholar
Gien, J, Tseng, N, Seedorf, G, et al. Endothelin-1 impairs angiogenesis in vitro through Rho-kinase activation after chronic intrauterine pulmonary hypertension in fetal sheep. Pediatr Res 2013; 73: 252–62.Google Scholar
Ivy, DD, Parker, TA, Ziegler, JW, et al. Prolonged endothelin A receptor blockade attenuates chronic pulmonary hypertension in the ovine fetus. J Clin Invest 1997; 99: 1179–86.Google Scholar
Mohamed, WA, Ismail, M. A randomized, double-blind, placebo-controlled, prospective study of bosentan for the treatment of persistent pulmonary hypertension of the newborn. J Perinatol 2012; 32(8): 608–13.Google Scholar
Steinhorn, RH, Fineman, J, Kusic-Pajic, A, et al. Bosentan as adjunctive therapy for persistent pulmonary hypertension of the newborn: results of the FUTURE-4 study. Circulation 2014; 130: A13503.Google Scholar
Reller, MD, Morton, MJ, Reid, DL, Thornburg, KL. Fetal lamb ventricles respond differently to filling and arterial pressures and to in utero ventilation. Pediatr Res 1987; 22: 621–6.Google Scholar
Cheung, PY, Barrington, KJ. The effects of dopamine and epinephrine on hemodynamics and oxygen metabolism in hypoxic anesthetized piglets. Crit Care 2001; 5: 158–66.Google Scholar
James, AT, Corcoran, JD, McNamara, PJ, et al. The effect of milrinone on right and left ventricular function when used as a rescue therapy for term infants with pulmonary hypertension. Cardiol Young 2015:1–10.Google Scholar
McNamara, PJ, Shivananda, SP, Sahni, M, et al. Pharmacology of milrinone in neonates with persistent pulmonary hypertension of the newborn and suboptimal response to inhaled nitric oxide. Pediatr Crit Care Med 2013; 14: 7484.Google Scholar
Chen, B, Lakshminrusimha, S, Czech, L, et al. Regulation of phosphodiesterase 3 in the pulmonary arteries during the perinatal period in sheep. Pediatr Res 2009; 66: 682–7.Google Scholar
Mohamed, A, Nasef, N, Shah, V, McNamara, PJ. Vasopressin as a rescue therapy for refractory pulmonary hypertension in neonates: case series. Pediatr Crit Care Med 2014; 15: 148–54.Google Scholar
Paden, ML, Rycus, PT, Thiagarajan, RR. Update and outcomes in extracorporeal life support. Semin Perinatol 2014; 38: 6570.Google Scholar
Bifano, EM, Pfannenstiel, A. Duration of hyperventilation and outcome in infants with persistent pulmonary hypertension. Pediatrics 1988; 81: 657–61.Google Scholar
Hageman, JR, Dusik, J, Keuler, H, et al. Outcome of persistent pulmonary hypertension in relation to severity of presentation. Am J Dis Child 1988; 142: 293–6.Google Scholar
Lipkin, PH, Davidson, D, Spivak, L, et al. Neurodevelopmental and medical outcomes of persistent pulmonary hypertension in term newborns treated with nitric oxide. J Pediatr 2002; 140: 306–10.Google Scholar
Konduri, GG, Vohr, B, Robertson, C, et al. Early inhaled nitric oxide therapy for term and near-term newborn infants with hypoxic respiratory failure: neurodevelopmental follow-up. J Pediatr 2007; 150: 235–40.Google Scholar
Fliman, PJ, deRegnier, RA, Kinsella, JP, et al. Neonatal extracorporeal life support: impact of new therapies on survival. J Pediatr 2006; 148: 595–9.Google Scholar
Massaro, AN. MRI for neurodevelopmental prognostication in the high-risk term infant. Semin Perinatol 2015; 39: 159–67.Google Scholar
Rollins, MD, Yoder, BA, Moore, KR, et al. Utility of neuroradiographic imaging in predicting outcomes after neonatal extracorporeal membrane oxygenation. J Pediatr Surg 2012; 47: 7680.Google Scholar
Gannon, CM, Kornhauser, MS, Gross, GW, et al. When combined, early bedside head ultrasound and electroencephalography predict abnormal computerized tomography or magnetic resonance brain images obtained after extracorporeal membrane oxygenation treatment. J Perinatol 2001; 21: 451–5.Google Scholar
Pappas, A, Shankaran, S, Stockmann, PT, Bara, R. Changes in amplitude-integrated electroencephalography in neonates treated with extracorporeal membrane oxygenation: a pilot study. J Pediatr 2006; 148: 125–7.Google Scholar
van Heijst, AF, de Mol, AC, Ijsselstijn, H. ECMO in neonates: neuroimaging findings and outcome. Semin Perinatol 2014; 38: 104–13.Google Scholar
UK Collaborative ECMO Trial Group. The collaborative UK ECMO (Extracorporeal Membrane Oxygenation) trial: follow-up to 1 year of age. Pediatrics 1998; 101: E1.Google Scholar
Bennett, CC, Johnson, A, Field, DJ, Elbourne, D. UK collaborative randomised trial of neonatal extracorporeal membrane oxygenation: follow-up to age 4 years. Lancet 2001; 357: 1094–6.Google Scholar
McNally, H, Bennett, CC, Elbourne, D, Field, DJ. United Kingdom collaborative randomized trial of neonatal extracorporeal membrane oxygenation: follow-up to age 7 years. Pediatrics 2006; 117: e845–54.Google Scholar
Ijsselstijn, H, van Heijst, AF. Long-term outcome of children treated with neonatal extracorporeal membrane oxygenation: increasing problems with increasing age. Semin Perinatol 2014; 38: 114–21.Google Scholar
Paden, ML, Conrad, SA, Rycus, PT, Thiagarajan, RR. Extracorporeal life support organization registry report 2012. ASAIO J 2013; 59: 202–10.Google Scholar
Parish, AP, Bunyapen, C, Cohen, MJ, et al. Seizures as a predictor of long-term neurodevelopmental outcome in survivors of neonatal extracorporeal membrane oxygenation (ECMO). J Child Neurol 2004; 19: 930–4.Google Scholar
Campbell, LR, Bunyapen, C, Gangarosa, ME, et al. Significance of seizures associated with extracorporeal membrane oxygenation. J Pediatr 1991; 119: 789–92.Google Scholar
Vaucher, YE, Dudell, GG, Bejar, R, Gist, K. Predictors of early childhood outcome in candidates for extracorporeal membrane oxygenation. J Pediatr 1996; 128: 109–17.Google Scholar
Kornhauser, MS, Baumgart, S, Desai, SA, et al. Adverse neurodevelopmental outcome after extracorporeal membrane oxygenation among neonates with bronchopulmonary dysplasia. J Pediatr 1998; 132: 307–11.Google Scholar
Van Meurs, KP, Nguyen, HT, Rhine, WD, et al. Intracranial abnormalities and neurodevelopmental status after venovenous extracorporeal membrane oxygenation. J Pediatr 1994; 125: 304–7.Google Scholar
Desai, SA, Stanley, C, Gringlas, M, et al. Five-year follow-up of neonates with reconstructed right common carotid arteries after extracorporeal membrane oxygenation. J Pediatr 1999; 134: 428–33.Google Scholar
Desai, S, Kollros, PR, Graziani, LJ, et al. Sensitivity and specificity of the neonatal brain-stem auditory evoked potential for hearing and language deficits in survivors of extracorporeal membrane oxygenation. J Pediatr 1997; 131: 233–9.Google Scholar
Cheung, PY, Robertson, CM. Sensorineural hearing loss in survivors of neonatal extracorporeal membrane oxygenation. Pediatr Rehabil 1997; 1: 127–30.Google Scholar
Joint Committee on Infant Hearing. Year 2007 position statement: principles and guidelines for early hearing detection and intervention programs. Pediatrics 2007; 120: 898921.Google Scholar
Beardsmore, C, Dundas, I, Poole, K, et al. Respiratory function in survivors of the UK Extracorporeal Membrane Oxygenation Trial. Am J Respir Crit Care Med 2000; 161: 1129–35.Google Scholar
Majaesic, CM, Jones, R, Dinu, IA, et al. Clinical correlations and pulmonary function at 8 years of age after severe neonatal respiratory failure. Pediatr Pulmonol 2007; 42: 829–37.Google Scholar

References

Bellinger, DC, Wypij, D, Kuban, KC, et al. Developmental and neurological status of children at 4 years of age after heart surgery with hypothermic circulatory arrest or low-flow cardiopulmonary bypass. Circulation 1999; 100: 526–32.Google Scholar
Majnemer, A, Limperopoulos, C. Developmental progress of children with congenital heart defects requiring open heart surgery. Semin Pediatr Neurol 1999; 6:1219.Google Scholar
Mahle, WT, Wernovsky, G. Neurodevelopmental outcomes in hypoplastic left heart syndrome. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2004; 7:3947.Google Scholar
Trittenwein, G, Nardi, A, Pansi, H, et al. Early postoperative prediction of cerebral damage after pediatric cardiac surgery. Ann Thorac Surg 2003; 76: 576–80.Google Scholar
Menache, CC, du Plessis, AJ, Wessel, DL, et al. Current incidence of acute neurologic complications after open heart operations in children. Ann Thorac Surg 2002; 73: 1752–8.Google Scholar
Pradat, P. Epidemiology of major congenital heart defects in Sweden, 1981–1986. J Epidemiol Community Health 1992; 46: 211–15.Google Scholar
Caputo, S, Capozzi, G, Russo, MG, et al. Familial recurrence of congenital heart disease in patients with ostium secundum atrial septal defect. Eur Heart J 2005; 26: 2179–84.Google Scholar
Caputo, S, Russo, MG, Capozzi, G, et al. Congenital heart disease in a population of dizygotic twins: an echocardiographic study. Int J Cardiol 2005; 102: 293–6.Google Scholar
Cymbron, T, Anjos, R, Cabral, R, et al. Epidemiological characterization of congenital heart disease in Sao Miguel Island, Azores, Portugal. Community Genet 2006; 9: 107–12.Google Scholar
Stollenberg, C, Magnus, P, Skrondal, A, et al. Consanguinity and recurrence risk of birth defects: a population-based study. Am J Med Genet 1999; 82: 423–8.Google Scholar
Williams, GD, Ramamoorthy, C. Brain monitoring and protection during pediatric cardiac surgery. Semin Cardiothorac Vasc Anesth 2007; 11:23.Google Scholar
Sullivan, KE, Jawad, KF, Randall, P, et al. Lack of correlation between impaired T cell production, immunodeficiency, and other phenotypic features in chromosome 22q11.2 deletion syndromes. Clin Immunol Immunopathol 1998; 86: 141–6.Google Scholar
Gothelf, D. Velocardiofacial syndrome. Child Adolesc Psychiatr Clin North Am 2007; 16: 677–93.Google Scholar
Mokhtar, MM, Abdel-Fattah, M. Major birth defects amongst infants with Down syndrome in Alexandria, Egypt (1995–2000): trends and risk factors. East Mediterr Health J 2001; 7: 441–51.Google Scholar
Ashok, M, Thangavel, G, Indrani, S, et al. Atrioventricular septal defect: associated anomalies and aneuploidy in prenatal life. Indian Pediatr 2003; 40: 659–64.Google Scholar
Gaynor, JW, Gerdes, M, Zackai, EH, et al. Apolipoprotein E genotype and neurodevelopment sequelae on infant cardiac surgery. J Thorac Cardiovasc Surg 2003; 126: 1736–45.Google Scholar
Mahle, WT, Tavani, F, Zimmerman, RA, et al. An MRI study of neurological injury before and after congenital heart surgery. Circulation 2002; 106:I109–14.Google Scholar
Limperopoulos, C, Majnemer, A, Shevell, MI, et al. Predictors of developmental disabilities after open heart surgery in young children with congenital heart defects. J Pediatr 2002; 141: 51–8.Google Scholar
Miller, SP, McQuillen, PS. Neurology of congenital heart disease: insight from brain imaging. Arch Dis Child Fetal Neonatal Ed 2007; 92:F435–7.Google Scholar
Gaynor, JW. Periventricular leukomalacia following neonatal and infant cardiac surgery. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2004; 7: 133–40.Google Scholar
Galli, KK, Zimmerman, RA, Jarvik, GP, et al. Periventricular leukomalacia is common after neonatal cardiac surgery. J Thorac Cardiovasc Surg 2004; 127:692704.Google Scholar
Hsia, TY, Gruber, PJ. Factors influencing neurological outcome after neonatal cardiopulmonary bypass: what we can and cannot control. Ann Thorac Surg 2006; 81:S2381–8.Google Scholar
Licht, DJ, Brain maturation is delayed in infants with complex congenital heart defects. J Thorac Cardiovasc Surg. 2009; 137(3): 529–37.Google Scholar
Glauser, TA, Rorke, LB, Weinberg, PM, et al. Congenital brain anomalies associated with the hypoplastic left heart syndrome. Pediatrics 1990; 85: 984–90.Google Scholar
Hinton, RB, et al. Prenatal head growth and white matter injury in hypoplastic left heart syndrome. Pediatr Res 2008; 64(4): 364–9Google Scholar
Te Pas, AB, van Wezel-Meijler, G, Bokenkamp-Gramann, R, et al. Preoperative cranial ultrasound findings in infants with major congenital heart disease. Acta Paediatr 2005; 94: 1597–603.Google Scholar
Miller, SP, McQuillen, PS, Hamrick, S, et al. Abnormal brain development in newborns with congenital heart disease. N Engl J Med 2007; 357: 1928–38.Google Scholar
McQuillen, PS, Hamrick, SEG, Perez, MJ, et al. Balloon atrial septostomy is associated with preoperative stroke in neonates with transposition of the great arteries. Circulation 2006; 113: 280–5.Google Scholar
McQuillen, PS, Barkovic, AJ, Hamrick, SEG, et al. Temporal and anatomic risk profile of brain injury with neonatal repair of congenital heart defects. Stroke 2007; 38: 736–41.Google Scholar
Bellinger, DC, Wypij, D, duPlessis, AJ, et al. Neurodevelopmental status at eight years in children with Dextro-transposition of the great arteries: the Boston Circulatory Arrest Trial. J Thorac Cardiovasc Surg 2003; 126: 1385–96.Google Scholar
Kurth, CD, Priestley, M, Watzman, HM, et al. Desflurane confers neurologic protection for deep hypothermic circulatory arrest in newborn pigs. Anesthesiology 2001; 95: 959–64.Google Scholar
Kawaguchi, M, Furuya, H, Patel, PM. Neuroprotective effects of anesthetic agents. J Anesth 2005; 19: 150–6.Google Scholar
Reid, RW, Warner, DS. Pro: arguments for use of barbiturates in infants and children undergoing deep hypothermic circulatory arrest. J Cardiovasc Anesth 1998; 12: 591–4.Google Scholar
Peek, GJ, Firmin, RK. The inflammatory and coagulative response to prolonged extracorporeal membrane oxygenation, a review. ASAIO J 1999; 45: 250–63.Google Scholar
Westaby, S, Saatvedt, K, White, S, et al. Is there a relationship between cognitive dysfunction and systemic inflammatory response after cardiopulmonary bypass? Ann Thorac Surg 2001; 71: 667–72.Google Scholar
Stump, DA. Embolic factors associated with cardiac surgery. Semin Cardiothorac Vasc Anesth 2005; 9: 151–2.Google Scholar
Prasongsukarn, K, Borger, MA. Reducing cerebral emboli during cardiopulmonary bypass. Semin Cardiothorac Vasc Anesth 2005; 9: 153–8.Google Scholar
Ehrlich, MP, McCullough, JN, Zhang, N, et al. Effect of hypothermia on cerebral blood flow and metabolism in the pig. Ann Thorac Surg 2002; 73: 191–7.Google Scholar
Neptune, WB, Bougas, JA, Panico, FG. Open-heart surgery without the need for donor-blood priming in the pump oxygenator. N Engl J Med 1960; 263: 111–15.Google Scholar
Jonas, RA, Wypij, D, Roth, SJ, et al. The influence of hemodilution on outcome after hypothermic cardiopulmonary bypass: results of a randomized trial in infants. J Thorac Cardiovasc Surg 2003; 126: 1765–74.Google Scholar
Jaggers, JJ, Ungerleider, RM. Cardiopulmonary bypass in infants and children. In Mavroudis, C, Backer, CL, eds., Pediatric Cardiac Surgery. St. Louis: Mosby, 2003: 171–93.Google Scholar
del Nido, PJ, McGowan, FX. Surgical approaches, cardiopulmonary bypass in pediatric cardiac surgery. In Sellke, FW, Swanson, SCJ, del Nido, P, eds., Surgery of the Chest. Philadelphia: Elsevier Saunders, 2005: 1821–58.Google Scholar
du Plessis, AJ, Jonas, RA, Wypij, D, et al. Perioperative effects of alpha-stat versus pH-stat strategies for deep hypothermic cardiopulmonary bypass in infants. J Thorac Cardiovasc Surg 1997; 114:9911001.Google Scholar
Hiramatsu, T, Miura, T, Forbess, JM, et al. pH strategies and cerebral energetics before and after circulatory arrest. J Thorac Cardiovasc Surg 1995; 109: 948–57.Google Scholar
Jonas, RA. Optimal pH strategy for CPB in neonates, infants and children. Perfusion 1998; 13: 377–87.Google Scholar
Scallan, MJH. Cerebral injury during pediatric heart surgery: perfusion issues. Perfusion 2004; 19: 221–8.Google Scholar
Gravlee, GP, Davis, RF, Kurusz, M, et al. Cardiopulmonary Bypass: Principles and Practice, 2nd edn. Philadelphia: Lippincott Williams & Wilkins, 2000:chap. 30.Google Scholar
Pigula, FA, Siewers, RD, Nemoto, EM. Hypothermic cardio-pulmonary bypass alters oxygen/glucose uptake in the pediatric brain. J Thorac Cardiovasc Surg 2001; 121: 366 –73Google Scholar
Schmid, FX, Phillip, A, Foltan, M, et al. Adequacy of perfusion during hypothermia: regional distribution of cardiopulmonary bypass flow, mixed venous and regional venous oxygen saturation-hypothermia and distribution of flow and oxygen. Thorac Cardiovasc Surg 2003; 51: 306 –11.Google Scholar
Kurth, CD, Steven, JM, Nicolson, SC, Jacobs, ML. Cerebral oxygenation during cardiopulmonary bypass in children. J Thorac Cardiovasc Surg 1997; 113: 71–9Google Scholar
Hanley, FL. Religion, politics … deep hypothermic circulatory arrest. J Thorac Cardiovasc Surg 2005; 130:1236.Google Scholar
Gaynor, JW, Nicholson, SC, Jarvik, GP, et al. Increasing duration of deep hypothermic circulatory arrest is associated with an increased incidence of postoperative electroencephalographicseizures. J Thorac Cardiovasc Surg 2005; 130: 1278–86.Google Scholar
Newburger, JW, Jonas, RA, Wernozsky, G, et al. A comparison of the perioperative neurologic effects of hypothermic circulatory arrest versus low-flow cardiopulmonary bypass in infant heart surgery. N Engl J Med 1993; 329: 1057–64.Google Scholar
Ungerleider, RM, Gaynor, JW. The Boston Circulatory Arrest Study: an analysis. J Thorac Cardiovasc Surg 2004; 127: 1256–61.Google Scholar
Wypij, D, Newburger, JW, Rappaport, LA, et al. The effect of duration of deep hypothermic circulatory arrest in infant heart surgery on late neurodevelopment: the Boston Circulatory Arrest Trial. J Thorac Cardiovasc Surg 2003; 126: 1397–403.Google Scholar
Amir, G, Ramamoorthy, C, Riemer, RK, et al. Neonatal brain protection and deep hypothermic circulatory arrest: pathophysiology of ischemic neuronal injury and protective strategies. Ann Thorac Surg 2005; 80: 1955–64.Google Scholar
Algra, S, et al. Neurological injury after neonatal cardiac surgery: a randomized, controlled trial of 2 perfusion techniques. Pediatr Cardiol 2014; 129: 224–33Google Scholar
Ly, M. et al. Continuous cerebral perfusion for aortic arch repair: hypothermia versus normothermia. Ann Thorac Surg 2011; 92: 942–8Google Scholar
Austin, EH, Edmonds, HL, Auden, SM, et al. Benefit of neurophysiologic monitoring for pediatric cardiac surgery. J Thorac Cardiovasc Surg 1997; 114: 707–16.Google Scholar
Sakamoto, T, Duebener, LF, Laussen, PC, et al. Cerebral ischemia caused by obstructed superior vena cava cannula is detected by near-infrared spectroscopy. J Cardiothorac Vasc Anesth 2004; 18:293303.Google Scholar
Kussman, BD, Wypij, D, Laussen, PC, et al. Relationship of intraoperative cerebral oxygen saturation to neurodevelopmental outcome and brain magnetic resonance imaging at 1 year of age in infants undergoing biventricular repair. Circulation. 2010; 122(3): 245–54.Google Scholar
Svenarud, P, Persson, M, van der Linden, J. Effect of CO2 insufflation on the number and behaviour of air microemboli in open-heart surgery: a randomized clinical trial. Circulation 2004; 109: 1127–32.Google Scholar
Bronicki, RA, Backer, CL, Baden, HP, et al. Dexamethasone reduces the inflammatory response to cardiopulmonary bypass in children. Ann Thorac Surg 2000; 69: 1490–5.Google Scholar
Checchia, PA, Bronicki, RA, Costello, JM, et al. Steroid use before pediatric cardiac operations using cardiopulmonary bypass: an international survey of 36 centers. Pediatr Crit Care Med 2005; 6: 441–4.Google Scholar
Elliott, M. Modified ultrafiltration and open heart surgery in children. Pediatr Anaesth 1999; 9:15.Google Scholar
Rodriguez, RA, Ruel, M, Broecker, L, et al. High flow rates during modified ultrafiltration decrease cerebral blood flow velocity and venous oxygen saturation in infants. Ann Thorac Surg 2005; 80: 22–8.Google Scholar
Ungerleider, RM, Shen, I, Yeh, T, et al. Routine mechanical ventricular assist following the Norwood procedure improved neurologic outcome and excellent hospital survival. Ann Thorac Surg 2004; 77:1822.Google Scholar
Peek, GJ, Firmin, RK. Extracorporeal membrane oxygenation for pediatric cardiac support. Coron Artery Dis 1997; 8: 371–89.Google Scholar
Hetzer, R, Loebe, M, Potapov, EV, et al. Circulatory support with pneumatic paracorporeal ventricular assist device in infants and children. Ann Thorac Surg 1998; 66: 1498–506.Google Scholar
Edwards, AD, Azzopardi, D. Hypothermic neural rescue treatment: from laboratory to cotside? Arch Dis Child Fetal Neonatal Ed 1998; 78:F8891.Google Scholar
Liu, WG, Qiu, WS, Zhang, Y, et al. Effects of selective brain cooling in patients with severe traumatic brain injury: a preliminary study. J Int Med Res 2006; 34:5864.Google Scholar
Gunn, AJ, Gunn, TR, de Haan, HH, et al. Dramatic neuronal rescue with prolonged selective head cooling after ischemia in fetal lambs. J Clin Invest 1997; 99: 248–56.Google Scholar
Paret, G, Tirosh, R, Lotan, D, et al. Early prediction of neurological outcome after falls in children: metabolic and clinical markers. J Accid Emerg Med 1999; 16: 186–8.Google Scholar
Chiaretti, A, De Benedictis, R, Langer, A, et al. Prognostic implications of hyperglycaemia in paediatric head injury. Childs Nerv Syst 1998; 14: 455–9.Google Scholar
Macrae, D. A randomized trial of hyperglycemic control in the pediatric intensive care unit. N Engl J Med 2014; 370(2): 107–18Google Scholar
Steward, DJ, DaSilva, CA, Flegel, T. Elevated blood glucose levels may increase the danger of neurological deficit following profound hypothermic cardiac arrest. Anesthesiology 1998; 68:653.Google Scholar
Ballweg, JA, Wernovsky, G, Gaynor, JW. Neurodevelopmental outcomes following congenital heart surgery. Pediatr Cardiol 2007; 28: 126–33.Google Scholar
Mahle, WT. Neurologic and cognitive outcomes in children with congenital heart disease. Curr Opin Pediatr 2001; 13: 482–6.Google Scholar
Gaynor, JW, Wernovsky, G, Jarvik, GP, et al. Patient characteristics are important determinants of neurodevelopmental outcome at one year of age after neonatal and infant cardiac surgery. J Thorac Cardiovasc Surg 2007; 133: 1344–53.Google Scholar
Robertson, CMT, Joffe, AR, Sauve, RS, et al. Outcomes from an interprovincial program of newborn open heart surgery. J Pediatr 2004; 144:8692.Google Scholar
Atallah, J, Joffe, AR, Robertson, CMT, et al. Two-year general and neurodevelopmental outcomes after neonatal complex cardiac surgery in patients with deletion 22q11.2: a comparative study. J Thorac Cardiovasc Surg 2007; 134: 772–9.Google Scholar
Forbess, JM, Visconti, KJ, Hancock-Friesen, C, et al. Neurodevelopmental outcome after congenital heart surgery: results from an institutional registry. Circulation 2002; 106:195202.Google Scholar
Hack, M, Wilson-Costello, D, Friedman, H, et al. Neurodevelopment and predictors of outcomes of children with birth weights of less than 1000 g:1992–1995. Arch Pediatr Adolesc Med 2000; 154: 725–31.Google Scholar
Hintz, SR, Kendrick, DE, Vohr, BR, et al. Changes in neurodevelopmental outcomes at 18–22 months corrected age among infants born at less than 25 weeks’ gestation 1993–1999. Pediatrics 2005; 115: 1645–51.Google Scholar
Wernovsky, G, Stiles, KM, Gauvreau, K, et al. Cognitive development after Fontan operation. Circulation 2000; 102: 883–9.Google Scholar
Mahle, WT, Viconti, KJ, Freier, C, et al. Relationship of surgical approach to neurodevelopmental outcomes in hypoplastic left heart syndrome. Pediatrics 2006; 117:e90–7.Google Scholar
Licht, DJ, Wang, J, Silvestre, DW, et al. Preoperative cerebral blood flow is diminished in neonates with severe congenital heart defects. J Thorac Cardiovasc Surg 2004; 128: 841–9.Google Scholar
Ilke, L, Hale, K, Fashaw, L, et al. Developmental outcome of patients with hypoplastic left heart syndrome treated with heart transplantation. J Pediatr 2003; 142: 20–5.Google Scholar
Mahle, WT, Clancy, RR, Moss, EM, et al. Neurodevelopmental outcome and lifestyle assessment in school-aged and adolescent children with hypoplastic left heart syndrome. Pediatrics 2000; 105: 1082–9.Google Scholar
Hack, M, Taylor, HG, Drotar, D, et al. Poor predictive validity of the Bayley Scales of Infant Development for cognitive function of extremely low birth weight children at school age. Pediatrics 2005; 116: 333–41.Google Scholar
McGrath, E, Wypij, D, Rappaport, LA, et al. Prediction of IQ and achievement at age 8 years from neurodevelopmental status at age 1 year in children with D-transposition of the great arteries. Pediatrics 2004; 114:e572–6.Google Scholar
Alton, GY, Robertson, CMT, Sauve, R, et al. Early childhood health, growth, and neurodevelopmental outcomes after complete repair of total anomalous pulmonary venous connection at 6 weeks or younger. J Thorac Cardiovasc Surg 2007; 133: 905–11.Google Scholar
Wypij, D, Jonas, RA, Bellinger, DC, et al. The effect of hematocrit during hypothermic cardiopulmonary bypass in infant heart surgery: results from the combined Boston hematocrit trials. J Thorac Cardiovasc Surg 2008; 135: 355–60.Google Scholar
Newburger, JW, Wypij, D, Bellinger, DC, et al. Length of stay after infant heart surgery is related to cognitive outcome at age 8 years. J Pediatr 2003; 143:6773.Google Scholar

References

Joint Commission. Sentinel Event Alert 30, July 21, 2004. Available at www.jointcommission.org/sentinelevents/sentineleventalert/sea_30.htm (accessed February 2008).Google Scholar
Perlman, JM, Wyllie, J, Kattwinkel, J, et al. Neonatal resuscitation: 2015 international consensus on cardiopulmonary resuscitation and emergency cardiovascular care science with treatment recommendations, part 7. Circulation 2015; 132(16 Suppl 1): S204–41.Google Scholar
Wyllie, J, Perlman, JM, Kattwinkel, J, et al. Neonatal resuscitation: 2015 international consensus on cardiopulmonary resuscitation and emergency cardiovascular care science with treatment recommendations, part 7. Resuscitation 2015; 95: e169201.Google Scholar
Perlman, JM, Wyllie, J, Kattwinkel, J, et al. Neonatal resuscitation: 2015 international consensus on cardiopulmonary resuscitation and emergency cardiovascular care science with treatment recommendations, part 7. Pediatrics 2015; 136(Suppl 2): S120–66.Google Scholar
Weiner, GM. Neonatal Resuscitation Textbook, 7th edn. Elk Grove Village, IL: American Academy of Pediatrics, American Heart Association, 2016.Google Scholar
Lockwood, CJ, Lemons, JA, eds. Guidelines for Perinatal Care, 6th edn. Elk Grove Village, IL: American Academy of Pediatrics, American College of Obstetricians and Gynecologists, 2007.Google Scholar
Oh, W, Fanaroff, AA, Carlo, WA, et al. Effects of delayed cord clamping in very-low-birth-weight infants. J Perinatol 2011; 31(Suppl 1): S6871.Google Scholar
Strauss, RG, Mock, DM, Johnson, KJ, et al. A randomized clinical trial comparing immediate versus delayed clamping of the umbilical cord in preterm infants: short-term clinical and laboratory endpoints. Transfusion 2008; 48(4): 658–65.Google Scholar
March, MI, Hacker, MR, Parson, AW, et al. The effects of umbilical cord milking in extremely preterm infants: a randomized controlled trial. J Perinatol 2013; 33(10): 763–7.Google Scholar
Katheria, AC, Leone, TA, Woelkers, D, et al. The effects of umbilical cord milking on hemodynamics and neonatal outcomes in premature neonates. J Pediatr 2014; 164(5): 1045–50.Google Scholar
Mullany, LC, Katz, J, Khatry, SK, et al. Risk of mortality associated with neonatal hypothermia in southern Nepal. Arch Pediatr & Adolesc Med 2010; 164(7): 650–6.Google Scholar
Laptook, AR, Salhab, W, Bhaskar, B. Admission temperature of low birth weight infants: predictors and associated morbidities. Pediatrics 2007; 119(3): e643–9.Google Scholar
Halbower, AC, Jones, MD. Physiologic reflexes and their impact on resuscitation of the newborn. Clin Perinatol 1999; 26: 621–7.Google Scholar
Vain, NE, Szyld, EG, Prudent, LM, et al. Oropharyngeal and nasopharyngeal suctioning of meconium-stained neonates before delivery of their shoulders: multicentre, randomized controlled trial. Lancet 2004; 364: 597602.Google Scholar
Wiswell, TE, Gannon, CM, Jacob, J, et al. Delivery room management of the apparently vigorous meconium-stained neonate: results of the multicenter, international collaborative trial. Pediatrics 2000; 106: 17.Google Scholar
Halliday, HL. Endotracheal intubation at birth for preventing morbidity and mortality in vigorous, meconium-stained infants born at term. Cochrane Database Syst Rev 2001; 1: CD000500.Google Scholar
Manganaro, R, Mami, C, Palmara, A, et al. Incidence of meconium aspiration syndrome in term meconium-stained babies managed at birth with selective tracheal intubation. J Perinat Med 2001; 29(6): 465–8.Google Scholar
Summary AAP/AHA 2015 guidelines for cardiopulmonary resuscitation and emergency cardiovascular care of the neonate, 2015. Available at www2.aap.org/nrp/docs/15535_NRP%20Guidelines%20Flyer_English_FINAL.pdf (accessed January 2016).Google Scholar
Bennett, S, Finer, NN, Rich, W, et al. A comparison of three neonatal resuscitation devices. Resuscitation 2005; 67: 113–18.Google Scholar
Aziz, HF, Martin, JB, Moore, JJ. The pediatric disposable end-tidal carbon dioxide detector role in endotracheal intubation in newborns. J Perinatol 1999; 19: 110–13.Google Scholar
Repetto, JE, Donohue, PCP, Baker, SF, et al. Use of capnography in the delivery room for assessment of endotracheal tube placement. J Perinatol 2001; 21: 284–7.Google Scholar
Shankaran, S, Laptook, AR, Pappas, A, et al. Effect of depth and duration of cooling on deaths in the NICU among neonates with hypoxic ischemic encephalopathy: a randomized clinical trial. JAMA 2014; 312(24): 2629–39.Google Scholar
Esmail, N, Saleh, M, Ali, A. Laryngeal mask airway versus endotracheal intubation for Apgar score improvement in neonatal resuscitation. Eg J Anesth 2002; 18: 115–21.Google Scholar
Milner, AD, Saunders, RA. Pressure and volume changes during the first breath of human neonates. Arch Dis Child 1977; 52: 918–24.Google Scholar
Avery, ME, Mead, J. Surface properties in relation to atelectasis and hyaline membrane disease. Am J Dis Child 1959; 97: 517–23.Google Scholar
Gerhardt, T, Bancalari, E. Chest wall compliance in full term and preterm infants. Acta Paediatr Scand 1980; 69: 359–64.Google Scholar
Heldt, G, McIlroy, MB. Dynamics of chest wall in preterm infants. J Appl Physiol 1987; 62: 170–4.Google Scholar
Morley, CJ. Continuous distending pressure. Arch Dis Child Fetal Neonatal Ed 1999; 81: F152–6.Google Scholar
Saunders, RA, Milner, AD, Hopkin, IE. The effects of continuous positive airway pressure on lung mechanics and lung volumes in the neonate. Biol Neonate 1976; 29: 178–86.Google Scholar
Richardson, CP, Jung, AL. Effects of continuous positive airway pressure on pulmonary function and blood gases of infants with respiratory distress syndrome. Pediatr Res 1978; 12: 771–4.Google Scholar
Cotton, RB, Lindstom, DP, Kanarek, KS, et al. Effect of positive-end-expiratory-pressure on right ventricular output in lambs with hyaline membrane disease. Acta Paediatr Scand 1980; 69: 603–6.Google Scholar
Ramji, S, Ahuja, S, Thirupuram, S, et al. Resuscitation of asphyxic newborn infants with room air or 100% oxygen. Pediatr Res 1993; 34: 809–12.Google Scholar
Saugstad, OD, Rootwelt, T, Aalen, O. Resuscitation of asphyxiated newborn infants with room air or oxygen: an international controlled trial. The Resair 2 Study. Pediatrics 1998; 102: e1.Google Scholar
Ramji, S, Rasaily, R, Mishra, PK, et al. Resuscitation of asphyxiated newborns with room air or 100% oxygen at birth: a multicentric trial. Indian Pediatr 2003; 40: 510–17.Google Scholar
Saugstad, OD, Ramji, S, Irani, SF, et al. Resuscitation of newborn infants with 21% or 100% oxygen: follow-up at 18 to 24 months. Pediatrics 2003; 112: 296300.Google Scholar
Vento, M, Asensi, M, Sastre, J, et al. Resuscitation with room air instead of 100% oxygen prevents oxidative stress in moderately asphyxiated term neonates. Pediatrics 2001; 107: 642–7.Google Scholar
Vento, M, Asensi, M, Sastre, J, et al. Oxidative stress in asphyxiated term infants resuscitated with 100% oxygen. J Pediatr 2003; 142: 240–6.Google Scholar
Kapadia, VS, Chalak, LF, Sparks, JE, et al. Resuscitation of preterm neonates with limited versus high oxygen strategy. Pediatrics. 2013; 132(6): e1488–96.Google Scholar
Rabi, Y, Singhal, N, Nettel-Aguirre, A. Room-air versus oxygen administration for resuscitation of preterm infants: the ROAR study. Pediatrics. 2011; 128(2): e374–81.Google Scholar
Rook, D, Schierbeek, H, Vento, M, et al. Resuscitation of preterm infants with different inspired oxygen fractions. J Pediatr 2014; 164(6): 1322–6.Google Scholar
Vento, M, Moro, M, Escrig, R, et al. Preterm resuscitation with low oxygen causes less oxidative stress, inflammation, and chronic lung disease. Pediatrics 2009; 124(3): e439–49.Google Scholar
Wang, CL, Anderson, C, Leone, TA, et al. Resuscitation of preterm neonates by using room air or 100% oxygen. Pediatrics 2008; 121(6): 1083–9.Google Scholar
Rajani, AK, Chitkara, R, Oehlert, J, Halamek, LP. Comparison of umbilical venous and intraosseous access during simulated neonatal resuscitation. Pediatrics. 2011; 128(4): e954–8.Google Scholar
Kitterman, JA, Phibbs, RH, Tooley, WH. Catheterization of umbilical vessels in newborn infants. Pediatr Clin North Am. 1970; 17: 895912.Google Scholar
Heinild, S, Søndergaard, T, Tudvad, F. Bone marrow infusion in childhood: experiences from a thousand infusions. J Pediatr 1947; 30: 400–12.Google Scholar
Glaeser, PW, Losek, JD, Nelson, DB, et al. Pediatric intraosseous infusions: impact on vascular access time. Am J Emerg Med 1988; 6: 330–2.Google Scholar
Ellemunter, H, Burkhart, S, Trawoger, R, et al. Intraosseous lines in preterm and full term neonates. Arch Dis Child Fetal Neonatal Ed 1999; 80: F74–5.Google Scholar
International Liaison Committee on Resuscitation. The International Liaison Committee on Resuscitation (ILCOR) consensus on science with treatment recommendations for pediatric and neonatal patients: neonatal resuscitation. Pediatrics 2006; 117: e978–88.Google Scholar
Romero, T, Friedman, WF. Limited left ventricular response to volume overload in the neonatal period: a comparative study with the adult animal. Pediatr Res 1979; 13: 910–15.Google Scholar
So, KW, Fok, TF, Ng, PC, et al. Randomized, controlled trial of colloid or crystalloid in hypotensive preterm infants. Arch Dis Child Fetal Neonatal Ed 1997; 76: F43–6.Google Scholar
Oca, MJ, Nelson, M, Donn, SM. Randomized trial of normal saline versus 5% albumin for the treatment of neonatal hypotension. J Perinatol 2003; 23: 473–6.Google Scholar
Emery, EF, Greenough, A, Gamsu, HR. Randomised, controlled trial of colloid infusions in hypotensive preterm infants. Arch Dis Child 1992; 67: 1185–8.Google Scholar
Srinivasan, G, Pildes, RS, Caughey, M, et al. Plasma glucose values in normal neonates: a new look. J Pediatr 1986; 109: 114–17.Google Scholar
Bishop, RL, Weisfeldt, ML. Sodium bicarbonate administration during cardiac arrest: effect on arterial pH, PCO2 and osmolality. JAMA 1976; 235: 506–9.Google Scholar
Berenyi, KJ, Wolk, M, Killip, T. Cerebrospinal fluid acidosis complicating therapy of experimental cardiopulmonary arrest. Circulation 1975; 52: 319–24.Google Scholar
Simmons, MA, Adcock, EW, Bard, H, et al. Hypernatremia and intracranial hemorrhages in neonates. N Engl J Med 1974; 291: 610.Google Scholar
Papile, L, Burnstein, J, Burnstein, R, et al. Relationship of intravenous sodium bicarbonate infusions and cerebral intraventricular hemorrhage. J Pediatr 1978; 93: 834–6.Google Scholar
Johnson, PJ. Sodium bicarbonate use in the treatment of acute neonatal lactic acidosis:benefit or harm? Neonatal Network 2011; 30(3): 199205.Google Scholar
Chernick, V, Manfreda, J, De Booy, V, et al. Clinical trial of naloxone in birth asphyxia. J Pediatr 1988; 113: 519–25.Google Scholar
Halamek, LP. The advantages of prenatal consultation by a neonatologist. J Perinatol 2001; 21: 116–20.Google Scholar
Halamek, LP. Prenatal consultation at the limits of viability. NeoReviews 2003; 4: e153–6.Google Scholar
Saigal, S. The limits of viability. Pediatr Res 2001; 49: 451.Google Scholar
Wood, NS, Marlow, N, Costeloe, K, et al. Neurologic and developmental disability after extremely preterm birth. EPICure Study Group. N Engl J Med 2000; 343: 378–84.Google Scholar
Costeloe, K, Hennessy, E, Gibson, AT, et al. The EPICure Study: outcomes to discharge from hospital for infants born at the threshold of viability. Pediatrics 2000; 106: 659–71.Google Scholar
deLeeuw, R, Cuttini, M, Nadai, M, et al. Treatment choices for extremely preterm infants: an international perspective. J Pediatr 2000; 137: 608–15.Google Scholar
Vohr, BR, Wright, LL, Dusick, AM, et al. Neurodevelopment and functional outcomes of extremely low birth weight infants in the National Institute of Child Health and Human Development Neonatal Research Network, 1993–1994. Pediatrics 2000; 105: 1216–26.Google Scholar
El-Metwally, D, Vohr, B, Tucker, R. Survival and neonatal morbidity at the limits of viability in the mid 1900s: 22 to 25 weeks. J Pediatr 2000; 137: 616–22.Google Scholar
Watts, J, Saigal, S. Replies to malcontent: fumes from the spleen. Pediatr Perinat Epidemiol 1995; 9: 375–9.Google Scholar
Saigal, S, Stoskopf, BL, Feeny, D, et al. Differences in preferences for neonatal outcomes among health care professionals, parents and adolescents. JAMA 1999; 281: 1991–7.Google Scholar
Lorenz, JM, Paneth, N. Treatment decisions for the extremely premature infant. J Pediatr 2000; 137: 593–5.Google Scholar
Stevenson, DK, Goldworth, A. Ethical dilemmas in the delivery room. Semin Perinatol 1998; 22: 198206.Google Scholar

References

Kohn, LT, Corrigan, JM, Donaldson, MS, eds. To Err Is Human: Building a Safer Health System. Washington, DC: National Academy of Sciences, 2000.Google Scholar
Brennan, TA, Leape, LL, Laird, NM, et al. Incidence of adverse events and negligence in hospitalized patients: results of the Harvard Medical Practice Study I. Quality & Safety in Health Care 2004; 13: 145–51; discussion 151–2.Google Scholar
Leape, LL, Brennan, TA, Laird, N, et al. The nature of adverse events in hospitalized patients: results of the Harvard Medical Practice Study II. N Engl J Med 1991; 324: 377–84.Google Scholar
Thomas, EJ, Studdert, DM, Burstin, HR, et al. Incidence and types of adverse events and negligent care in Utah and Colorado. Med Care 2000; 38: 261–71.Google Scholar
Crossing the Quality Chasm: A New Health System for the 21st Century. Washington, DC: National Academy of Sciences, 2001.Google Scholar
Preventing infant death and injury during delivery. Sentinel Event Alert 2004: 13.Google Scholar
Aspden, P, Corrigan, JM, Wolcott, J, Erickson, SM, eds. Patient Safety: Achieving a New Standard for Care. Washington, DC: National Academy of Sciences, 2004.Google Scholar
Billings, CE, Reynard, WD. Human factors in aircraft incidents: results of a 7-year study. Aviat Space Environ Med 1984; 55: 960–5.Google Scholar
Cockpit Resource Management. San Diego: Academic Press, 1993.Google Scholar
Chitkara, R, Rajani, AK, Oehlert, JW, et al. The accuracy of human senses in the detection of neonatal heart rate during standardized simulated resuscitation: implications for delivery of care, training and technology design. Resuscitation 2013; 84: 369–72.Google Scholar
Rajani, AK, Chitkara, R, Oehlert, J, Halamek, LP. Comparison of umbilical venous and intraosseous access during simulated neonatal resuscitation. Pediatrics 2011; 128: e954–8.Google Scholar
Yamada, NK, Yaeger, KA, Halamek, LP. Analysis and classification of errors made by teams during neonatal resuscitation. Resuscitation 2015; 96: 109–13.Google Scholar
Thomas, EJ, Sexton, JB, Lasky, RE, et al. Teamwork and quality during neonatal care in the delivery room. J Perinatol 2006; 26: 163–9.Google Scholar
Thomas, EJ, Taggart, B, Crandell, S, et al. Teaching teamwork during the Neonatal Resuscitation Program: a randomized trial. J Perinatol 2007; 27: 409–14.Google Scholar
Thomas, EJ, Williams, AL, Reichman, EF, et al. Team training in the neonatal resuscitation program for interns: teamwork and quality of resuscitations. Pediatrics 2010; 125: 539–46.Google Scholar
International Ergonomics Association. Available at www.iea.cc/whats/index.html (accessed July 14, 2015).Google Scholar
Gawron, VJ, Drury, CG, Fairbanks, RJ, Berger, RC. Medical error and human factors engineering: where are we now? Am J Med Qual 2006; 21: 5767.Google Scholar
Gosbee, J. Human factors engineering and patient safety. Quality & Safety in Health Care 2002; 11: 352–4.Google Scholar
Walsh, T, Beatty, PC. Human factors error and patient monitoring. Physiol Meas 2002; 23: R111–32.Google Scholar
Hunt, EA, Nelson, KL, Shilkofski, NA. Simulation in medicine: addressing patient safety and improving the interface between healthcare providers and medical technology. Biomed Instrum Technol 2006; 40: 399404.Google Scholar
Fuerch, JH, Yamada, NK, Coelho, PR, et al. Impact of a novel decision support tool on adherence to Neonatal Resuscitation Program algorithm. Resuscitation 2015; 88: 52–6.Google Scholar

References

Benitz, WE, Frankel, LR, Stevenson, DK. The pharmacology of neonatal resuscitation and cardiopulmonary intensive care. II. Extended intensive care. West J Med 1986; 145: 4751.Google Scholar
van den Broek, MP, Groenendaal, F, Egberts, AC, Rademaker, CM. Effects of hypothermia on pharmacokinetics and pharmacodynamics: a systematic review of preclinical and clinical studies. Clin Pharmacokinet 2010; 49: 277–94.Google Scholar
Zanelli, S, Buck, M, Fairchild, K. Physiologic and pharmacologic considerations for hypothermia therapy in neonates. J Perinatol 2011; 31: 377–86.Google Scholar
Stevenson, DK, Benitz, WE. A practical approach to diagnosis and immediate care of the cyanotic neonate: stabilization and preparation for transfer to level III nursery. Clin Pediatr (Phila) 1987; 26: 325–31.Google Scholar
Bruce, DA. Effects of hyperventilation on cerebral blood flow and metabolism. Clin Perinatol 1984; 11: 673–80.Google Scholar
Walsh-Sukys, MC, Tyson, JE, Wright, LL, et al. Persistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomes. Pediatrics 2000; 105: 1420.Google Scholar
Kusuda, S, Shishida, N, Miyagi, N, et al. Cerebral blood flow during treatment for pulmonary hypertension. Arch Dis Child Fetal Neonatal Ed 1999; 80: F30–3.Google Scholar
Gleason, CA, Short, BL, Jones, MD Jr. Cerebral blood flow and metabolism during and after prolonged hypocapnia in newborn lambs. J Pediatr 1989; 115: 309–14.Google Scholar
Liem, KD, Hopman, JC, Oeseburg, B, et al. Cerebral oxygenation and hemodynamics during induction of extracorporeal membrane oxygenation as investigated by near infrared spectrophotometry. Pediatrics 1995; 95: 555–61.Google Scholar
Toft, PB, Leth, H, Lou, HC, et al. Local vascular CO2 reactivity in the infant brain assessed by functional MRI. Pediatr Radiol 1995; 25: 420–4.Google Scholar
Chalak, LF, Tarumi, T, Zhang, R. The “neurovascular unit approach” to evaluate mechanisms of dysfunctional autoregulation in asphyxiated newborns in the era of hypothermia therapy. Early Hum Dev 2014; 90: 687–94.Google Scholar
Bifano, EM, Pfannenstiel, A. Duration of hyperventilation and outcome in infants with persistent pulmonary hypertension. Pediatrics 1988; 81: 657–61.Google Scholar
Hendricks-Munoz, KD, Walton, JP. Hearing loss in infants with persistent fetal circulation. Pediatrics 1988; 81: 650–6.Google Scholar
Leavitt, AM, Watchko, JF, Bennett, FC, Folsom, RC. Neurodevelopmental outcome following persistent pulmonary hypertension of the neonate. J Perinatol 1987; 7: 288–91.Google Scholar
Walsh-Sukys, MC, Cornell, DJ, Houston, LN, et al. Treatment of persistent pulmonary hypertension of the newborn without hyperventilation: an assessment of diffusion of innovation. Pediatrics 1994; 94: 303–6.Google Scholar
Wung, JT, James, LS, Kilchevsky, E, James, E. Management of infants with severe respiratory failure and persistence of the fetal circulation, without hyperventilation. Pediatrics 1985; 76: 488–94.Google Scholar
Clark, RH, Yoder, BA, Sell, MS. Prospective, randomized comparison of high-frequency oscillation and conventional ventilation in candidates for extracorporeal membrane oxygenation. J Pediatr 1994; 124: 447–54.Google Scholar
Baumgart, S, Hirschl, RB, Butler, SZ, et al. Diagnosis-related criteria in the consideration of extracorporeal membrane oxygenation in neonates previously treated with high-frequency jet ventilation. Pediatrics 1992; 89: 491–4.Google Scholar
deLemos, R, Yoder, B, McCurnin, D, et al. The use of high-frequency oscillatory ventilation (HFOV) and extracorporeal membrane oxygenation (ECMO) in the management of the term/near term infant with respiratory failure. Early Hum Dev 1992; 29: 299303.Google Scholar
Bhuta, T, Henderson-Smart, DJ. Rescue high frequency oscillatory ventilation versus conventional ventilation for pulmonary dysfunction in preterm infants. Cochrane Database Syst Rev 1998; 2: CD000438.Google Scholar
Hintz, SR, Suttner, DM, Sheehan, AM, et al. Decreased use of neonatal extracorporeal membrane oxygenation (ECMO): how new treatment modalities have affected ECMO utilization. Pediatrics 2000; 106: 1339–43.Google Scholar
Cools, F, Offringa, M. Meta-analysis of elective high frequency ventilation in preterm infants with respiratory distress syndrome. Arch Dis Child Fetal Neonatal Ed 1999; 80: F1520.Google Scholar
Henderson-Smart, DJ, Bhuta, T, Cools, F, Offringa, M. Elective high frequency oscillatory ventilation versus conventional ventilation for acute pulmonary dysfunction in preterm infants. Cochrane Database Syst Rev 2003; 4: CD000104.Google Scholar
Crone, RK, Favorito, J. The effects of pancuronium bromide on infants with hyaline membrane disease. J Pediatr 1980; 97: 991–3.Google Scholar
Goudsouzian, NG, Liu, LM, Savarese, JJ. Metocurine in infants and children: neuromuscular and clinical effects. Anesthesiology 1978; 49: 266–9.Google Scholar
Anand, KJ, Barton, BA, McIntosh, N, et al. Analgesia and sedation in preterm neonates who require ventilatory support: results from the NOPAIN trial: neonatal outcome and prolonged analgesia in neonates. Arch Pediatr Adolesc Med 1999; 153: 331–8.Google Scholar
Grunau, RE, Whitfield, MF, Petrie-Thomas, J, et al. Neonatal pain, parenting stress and interaction, in relation to cognitive and motor development at 8 and 18 months in preterm infants. Pain 2009; 143: 138–46.Google Scholar
Jobe, AH. Pulmonary surfactant therapy. N Engl J Med 1993; 328: 861–8.Google Scholar
Kendig, JW, Ryan, RM, Sinkin, RA, et al. Comparison of two strategies for surfactant prophylaxis in very premature infants: a multicenter randomized trial. Pediatrics 1998; 101: 1006–12.Google Scholar
Soll, RF. Synthetic surfactant for respiratory distress syndrome in preterm infants. Cochrane Database Syst Rev 1998; 3: CD001149.Google Scholar
Bancalari, E, del Moral, T. Bronchopulmonary dysplasia and surfactant. Biol Neonate 2001; 80(Suppl 1): 713.Google Scholar
Greenough, A. Expanded use of surfactant replacement therapy. Eur J Pediatr 2000; 159: 635–40.Google Scholar
Lotze, A, Mitchell, BR, Bulas, DI, et al. Multicenter study of surfactant (beractant) use in the treatment of term infants with severe respiratory failure. Survanta in Term Infants Study Group. J Pediatr 1998; 132: 40–7.Google Scholar
Neonatal Inhaled Nitric Oxide Study Group (NINOS). Inhaled nitric oxide in full-term and nearly full-term infants with hypoxic respiratory failure. N Engl J Med 1997; 336: 597604.Google Scholar
Neonatal Inhaled Nitric Oxide Study Group (NINOS). Inhaled nitric oxide and hypoxic respiratory failure in infants with congenital diaphragmatic hernia. Pediatrics 1997; 99: 838–45.Google Scholar
Van Meurs, KP, Wright, LL, Ehrenkranz, RA, et al. Inhaled nitric oxide for premature infants with severe respiratory failure. N Engl J Med 2005; 353: 1322.Google Scholar
Breitweser, JA, Meyer, RA, Sperling, MA, et al. Cardiac septal hypertrophy in hyperinsulinemic infants. J Pediatr 1980; 96: 535–9.Google Scholar
Seri, I. Systemic and pulmonary effects of vasopressors and inotropes in the neonate. Biol Neonate 2006; 89: 340–2.Google Scholar
Zaritsky, A, Chernow, B. Use of catecholamines in pediatrics. J Pediatr 1984; 105: 341–50.Google Scholar
Friedman, WF, George, BL. Treatment of congestive heart failure by altering loading conditions of the heart. J Pediatr 1985; 106: 697706.Google Scholar
Bard, H. Hemoglobin synthesis and metabolism during the neonatal period. In Christensen, RD, ed., Hematologic Problems of the Neonate. Philadelphia: Saunders, 2000: 374–7.Google Scholar
Yeh, TF, Shibli, A, Leu, ST, et al. Early furosemide therapy in premature infants (less than or equal to 2000 g) with respiratory distress syndrome: a randomized, controlled trial. J Pediatr 1984; 105: 603–9.Google Scholar
Benitz, WE. Treatment of persistent patent ductus arteriosus in preterm infants: time to accept the null hypothesis? J Perinatol 2010; 30: 241–52.Google Scholar
Van Overmeire, B, Smets, K, Lecoutere, D, et al. A comparison of ibuprofen and indomethacin for closure of patent ductus arteriosus. N Engl J Med 2000; 343: 674–81.Google Scholar
Fowlie, PW. Intravenous indomethacin for preventing mortality and morbidity in very low birth weight infants. Cochrane Database Syst Rev 2000; 2: CD000174.Google Scholar
Schmidt, B, Davis, P, Moddemann, D, et al. Long-term effects of indomethacin prophylaxis in extremely-low-birth-weight infants. N Engl J Med 2001; 344: 1966–72.Google Scholar
Ment, LR, Vohr, B, Allan, W, et al. Outcome of children in the indomethacin intraventricular hemorrhage prevention trial. Pediatrics 2000; 105: 485–91.Google Scholar
Ment, LR, Vohr, BR, Makuch, RW, et al. Prevention of intraventricular hemorrhage by indomethacin in male preterm infants. J Pediatr 2004; 145: 832–4.Google Scholar
Clyman, RI. Recommendations for the postnatal use of indomethacin: an analysis of four separate strategies. J Pediatr 1996; 128: 601–7.Google Scholar
Watterberg, KL, Gerdes, JS, Cole, CH, et al. Prophylaxis of early adrenal insufficiency to prevent bronchopulmonary dysplasia: a multicenter trial. Pediatrics 2004; 114: 1649–57.Google Scholar
Heymann, MA. Pharmacologic use of prostaglandin E1 in infant with congenital heart disease. Am Heart J 1981; 101: 837–43.Google Scholar
Brann, AW Jr., Myers, RE. Central nervous system findings in the newborn monkey following severe in utero partial asphyxia. Neurology 1975; 25: 327–38.Google Scholar
Myers, RE. Two patterns of perinatal brain damage and their conditions of occurrence. Am J Obstet Gynecol 1972; 112: 246–76.Google Scholar
Myers, RE. Experimental models of perinatal brain damage: relevance to human pathology. In Gluck, L, ed., Intrauterine Asphyxia and the Developing Fetal Brain. Chicago: Year Book, 1977: 3797.Google Scholar
Mujsce, DJ, Christensen, MA, Vannucci, RC. Cerebral blood flow and edema in perinatal hypoxic-ischemic brain damage. Pediatr Res 1990; 27: 450–3.Google Scholar
Young, RS, Yagel, SK. Cerebral physiological and metabolic effects of hyperventilation in the neonatal dog. Ann Neurol 1984; 16: 337–42.Google Scholar
Hill, A. Current concepts of hypoxic-ischemic cerebral injury in the term newborn. Pediatr Neurol 1991; 7: 317–25.Google Scholar
Lupton, BA, Hill, A, Roland, EH, et al. Brain swelling in the asphyxiated term newborn: pathogenesis and outcome. Pediatrics 1988; 82: 139–46.Google Scholar
Volpe, JJ. Hypoxic-ischemic encephalopathy. In Volpe, JJ, ed., Neurology of the Newborn. Philadelphia: Saunders, 2001: 217394.Google Scholar
Levene, MI, Evans, DH. Continuous measurement of subarachnoid pressure in the severely asphyxiated newborn. Arch Dis Child 1983; 58: 1013–5.Google Scholar
Levene, MI. Management and outcome of birth asphyxia. In Levene, MI, Lilforde, RJ, eds., Fetal and Neonatal Neurology and Neurosurgery. Edinburgh: Churchill Livingston, 1995: 427–42.Google Scholar
Levene, MI, Evans, DH, Forde, A, Archer, LN. Value of intracranial pressure monitoring of asphyxiated newborn infants. Dev Med Child Neurol 1987; 29: 311–9.Google Scholar
Rosenberg, AA, Jones, MD Jr, Traystman, RJ, et al. Response of cerebral blood flow to changes in PCO2 in fetal, newborn, and adult sheep. Am J Physiol 1982; 242: H862–6.Google Scholar
Bernbaum, JC, Russell, P, Sheridan, PH, et al. Long-term follow-up of newborns with persistent pulmonary hypertension. Crit Care Med 1984; 12: 579–83.Google Scholar
Wiswell, TE, Graziani, LJ, Kornhauser, MS, et al. Effects of hypocarbia on the development of cystic periventricular leukomalacia in premature infants treated with high-frequency jet ventilation. Pediatrics 1996; 98: 918–24.Google Scholar
Dammann, O, Allred, EN, Kuban, KC, et al. Hypocarbia during the first 24 postnatal hours and white matter echolucencies in newborns ≤28 weeks gestation. Pediatr Res 2001; 49: 388–93.Google Scholar
Vannucci, RC, Towfighi, J, Heitjan, DF, Brucklacher, RM. Carbon dioxide protects the perinatal brain from hypoxic-ischemic damage: an experimental study in the immature rat. Pediatrics 1995; 95: 868–74.Google Scholar
Vannucci, RC, Towfighi, J, Brucklacher, RM, Vannucci, SJ. Effect of extreme hypercapnia on hypoxic-ischemic brain damage in the immature rat. Pediatr Res 2001; 49: 799803.Google Scholar
Cooper, PR, Moody, S, Clark, WK, et al. Dexamethasone and severe head injury: a prospective double-blind study. J Neurosurg 1979; 51: 307–16.Google Scholar
Dearden, NM, Gibson, JS, McDowall, DG, et al. Effect of high-dose dexamethasone on outcome from severe head injury. J Neurosurg 1986; 64: 81–8.Google Scholar
Levene, MI, Evans, DH. Medical management of raised intracranial pressure after severe birth asphyxia. Arch Dis Child 1985; 60: 12–6.Google Scholar
Barks, JD, Post, M, Tuor, UI. Dexamethasone prevents hypoxic-ischemic brain damage in the neonatal rat. Pediatr Res 1991; 29: 558–63.Google Scholar
Tuor, UI, Simone, CS, Barks, JD, Post, M. Dexamethasone prevents cerebral infarction without affecting cerebral blood flow in neonatal rats. Stroke 1993; 24: 452–7.Google Scholar
National Institutes of Health Consensus Development Panel on the Effect of Corticosteroids for Fetal Maturation of Perinatal Outcomes. Effect of corticosteroids for fetal maturation on perinatal outcomes. JAMA 1994; 273: 413–8.Google Scholar
Adhikari, M, Moodley, M, Desai, PK. Mannitol in neonatal cerebral oedema. Brain Dev 1990; 12: 349–51.Google Scholar
Marchal, C, Costagliolu, P, Leaveau, P, Wong, RJ. Treatment de la souffrance cerebrale neonatale d’orisivie anoxique par le mannitol. Rev Pediatr 1974; 9: 581–9.Google Scholar
Adamson, SJ, Alessandri, LM, Badawi, N, et al. Predictors of neonatal encephalopathy in full-term infants. BMJ 1995; 311: 598602.Google Scholar
Badawi, N, Kurinczuk, JJ, Keogh, JM, et al. Intrapartum risk factors for newborn encephalopathy: the Western Australian case-control study. BMJ 1998; 317: 1554–8.Google Scholar
Shankaran, S. The postnatal management of the asphyxiated term infant. Clin Perinatol 2002; 29: 675–92.Google Scholar
Shankaran, S, Laptook, AR, Pappas, A, et al. Effect of depth and duration of cooling on deaths in the NICU among neonates with hypoxic ischemic encephalopathy: a randomized clinical trial. JAMA 2014; 312: 2629–39.Google Scholar
Svenningsen, NW, Blennow, G, Lindroth, M, et al. Brain-orientated intensive care treatment in severe neonatal asphyxia: effects of phenobarbitone protection. Arch Dis Child 1982; 57: 176–83.Google Scholar
Hall, RT, Hall, FK, Daily, DK. High-dose phenobarbital therapy in term newborn infants with severe perinatal asphyxia: a randomized, prospective study with three-year follow-up. J Pediatr 1998; 132: 345–8.Google Scholar
Sarkar, S, Barks, JD, Bapuraj, JR, et al. Does phenobarbital improve the effectiveness of therapeutic hypothermia in infants with hypoxic-ischemic encephalopathy? J Perinatol 2012; 32: 1520.Google Scholar
Meyn, DF Jr, Ness, J, Ambalavanan, N, Carlo, WA. Prophylactic phenobarbital and whole-body cooling for neonatal hypoxic-ischemic encephalopathy. J Pediatr 2010; 157: 334–6.Google Scholar
Diaz, J, Schain, RJ. Phenobarbital: effects of long-term administration on behavior and brain of artificially reared rats. Science 1978; 199: 90–1.Google Scholar
Bittigau, P, Sifringer, M, Genz, K, et al. Antiepileptic drugs and apoptotic neurodegeneration in the developing brain. Proc Natl Acad Sci USA 2002; 99: 15089–94.Google Scholar
Miller, SP, Weiss, J, Barnwell, A, et al. Seizure-associated brain injury in term newborns with perinatal asphyxia. Neurology 2002; 58: 542–8.Google Scholar
Evans, DJ, Levene, MI, Tsakmakis, M. Anticonvulsants for preventing mortality and morbidity in full term newborns with perinatal asphyxia. Cochrane Database Syst Rev 2007; 3: D001240.Google Scholar
Giacoia, GP. Asphyxial brain damage in the newborn: new insights into pathophysiology and possible pharmacologic interventions. South Med J 1993; 86: 676–82.Google Scholar
Muir, KW, Lees, KR. Clinical experience with excitatory amino acid antagonist drugs. Stroke 1995; 26: 503–13.Google Scholar
Levene, M. Role of excitatory amino acid antagonists in the management of birth asphyxia. Biol Neonate 1992; 62: 248–51.Google Scholar
Steinberg, GK, Bell, TE, Yenari, MA. Dose escalation safety and tolerance study of the N-methyl-d-aspartate antagonist dextromethorphan in neurosurgery patients. J Neurosurg 1996; 84: 860–6.Google Scholar
Davis, SM, Lees, KR, Albers, GW, et al. Selfotel in acute ischemic stroke: possible neurotoxic effects of an NMDA antagonist. Stroke 2000; 31: 347–54.Google Scholar
Parikka, H, Toivonen, L, Naukkarinen, V, et al. Decreases by magnesium of QT dispersion and ventricular arrhythmias in patients with acute myocardial infarction. Eur Heart J 1999; 20: 111–20.Google Scholar
Lampl, Y, Gilad, R, Geva, D, et al. Intravenous administration of magnesium sulfate in acute stroke: a randomized double-blind study. Clin Neuropharmacol 2001; 24: 11–5.Google Scholar
Lucas, MJ, Leveno, KJ, Cunningham, FG. A comparison of magnesium sulfate with phenytoin for the prevention of eclampsia. N Engl J Med 1995; 333: 201–5.Google Scholar
de Haan, HH, Gunn, AJ, Williams, CE, et al. Magnesium sulfate therapy during asphyxia in near-term fetal lambs does not compromise the fetus but does not reduce cerebral injury. Am J Obstet Gynecol 1997; 176: 1827.CrossRefGoogle Scholar
Marret, S, Gressens, P, Gadisseux, JF, Evrard, P. Prevention by magnesium of excitotoxic neuronal death in the developing brain: an animal model for clinical intervention studies. Dev Med Child Neurol 1995; 37: 473–84.Google Scholar
McDonald, JW, Silverstein, FS, Johnston, MV. Magnesium reduces N-methyl-d-aspartate (NMDA)–mediated brain injury in perinatal rats. Neurosci Lett 1990; 109: 234–8.Google Scholar
Penrice, J, Amess, PN, Punwani, S, et al. Magnesium sulfate after transient hypoxia-ischemia fails to prevent delayed cerebral energy failure in the newborn piglet. Pediatr Res 1997; 41: 443–7.Google Scholar
Levene, M, Blennow, M, Whitelaw, A, et al. Acute effects of two different doses of magnesium sulphate in infants with birth asphyxia. Arch Dis Child Fetal Neonatal Ed 1995; 73: F174–7.Google Scholar
Robertson, NJ, Edwards, AD. Recent advances in developing neuroprotective strategies for perinatal asphyxia. Curr Opin Pediatr 1998; 10: 575–80.CrossRefGoogle ScholarPubMed
Marret, S, Doyle, LW, Crowther, CA, Middleton, P. Antenatal magnesium sulphate neuroprotection in the preterm infant. Semin Fetal Neonatal Med 2007; 12: 311–7.Google Scholar
Crowther, CA, Hiller, JE, Doyle, LW, Haslam, RR. Effect of magnesium sulfate given for neuroprotection before preterm birth: a randomized, controlled trial. JAMA 2003; 290: 2669–76.CrossRefGoogle ScholarPubMed
Rouse, DJ, Hirtz, DG, Thom, E, et al. A randomized, controlled trial of magnesium sulfate for the prevention of cerebral palsy. N Engl J Med 2008; 359: 895905.Google Scholar
American College of Obstetricians and Gynecologists Committee on Obstetric Practice Society for Maternal-Fetal Medicine. Magnesium sulfate before anticipated preterm birth for neuroprotection (Committee Opinion No. 455). Obstet Gynecol 2010; 115: 669–71.Google Scholar
Gunn, AJ, Mydlar, T, Bennet, L, et al. The neuroprotective actions of a calcium channel antagonist, flunarizine, in the infant rat. Pediatr Res 1989; 25: 573–6.CrossRefGoogle ScholarPubMed
Gunn, AJ, Williams, CE, Mallard, EC, et al. Flunarizine, a calcium channel antagonist, is partially prophylactically neuroprotective in hypoxic-ischemic encephalopathy in the fetal sheep. Pediatr Res 1994; 35: 657–63.Google Scholar
Levene, MI, Gibson, NA, Fenton, AC, et al. The use of a calcium-channel blocker, nicardipine, for severely asphyxiated newborn infants. Dev Med Child Neurol 1990; 32: 567–74.Google Scholar
Buonocore, G, Groenendaal, F. Anti-oxidant strategies. Semin Fetal Neonatal Med 2007; 12: 287–95.Google Scholar
Palmer, C, Towfighi, J, Roberts, RL, Heitjan, DF. Allopurinol administered after inducing hypoxia-ischemia reduces brain injury in 7-day-old rats. Pediatr Res 1993; 33: 405–11.Google Scholar
Van Bel, F, Shadid, M, Moison, RM, et al. Effect of allopurinol on postasphyxial free radical formation, cerebral hemodynamics, and electrical brain activity. Pediatrics 1998; 101: 185–93.Google Scholar
Benders, MJ, Bos, AF, Rademaker, CM, et al. Early postnatal allopurinol does not improve short term outcome after severe birth asphyxia. Arch Dis Child Fetal Neonatal Ed 2006; 91: F163–5.Google Scholar
Gunes, T, Ozturk, MA, Koklu, E, et al. Effect of allopurinol supplementation on nitric oxide levels in asphyxiated newborns. Pediatr Neurol 2007; 36: 1724.Google Scholar
Welin, AK, 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.CrossRefGoogle ScholarPubMed
Robertson, NJ, Faulkner, S, Fleiss, B, et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain 2013; 136: 90105.Google Scholar
Aly, H, Elmahdy, H, El-Dib, M, et al. Melatonin use for neuroprotection in perinatal asphyxia: a randomized controlled pilot study. J Perinatol 2015; 35: 186–91.Google Scholar
Tan, WK, Williams, CE, Mallard, CE, Gluckman, PD. Monosialoganglioside GM1 treatment after a hypoxic-ischemic episode reduces the vulnerability of the fetal sheep brain to subsequent injuries. Am J Obstet Gynecol 1994; 170: 663–9.Google Scholar
Hall, ED. The neuroprotective pharmacology of methylprednisolone. J Neurosurg 1992; 76: 1322.Google Scholar
Amar, AP, Levy, ML. Pathogenesis and pharmacological strategies for mitigating secondary damage in acute spinal cord injury. Neurosurgery 1999; 44: 1027–39; discussion 3940.Google Scholar
Hall, ED, McCall, JM, Means, ED. Therapeutic potential of the lazaroids (21-aminosteroids) in acute central nervous system trauma, ischemia and subarachnoid hemorrhage. Adv Pharmacol 1994; 28: 221–68.Google Scholar
Kavanagh, RJ, Kam, PC. Lazaroids: efficacy and mechanism of action of the 21-aminosteroids in neuroprotection. Br J Anaesth 2001; 86: 110–9.Google Scholar
McPherson, RJ, Juul, SE. Recent trends in erythropoietin-mediated neuroprotection. Int J Dev Neurosci 2008; 26: 103–11.Google Scholar
Gonzalez, FF, McQuillen, P, Mu, D, et al. Erythropoietin enhances long-term neuroprotection and neurogenesis in neonatal stroke. Dev Neurosci 2007; 29: 321–30.Google Scholar
Wu, YW, Bauer, LA, Ballard, RA, et al. Erythropoietin for neuroprotection in neonatal encephalopathy: safety and pharmacokinetics. Pediatrics 2012; 130: 683–91.Google Scholar
Sabir, H, Bishop, S, Cohen, N, et al. Neither xenon nor fentanyl induces neuroapoptosis in the newborn pig brain. Anesthesiology 2013; 119: 345–57.Google Scholar
Ma, D, Hossain, M, Chow, A, et al. Xenon and hypothermia combine to provide neuroprotection from neonatal asphyxia. Ann Neurol 2005; 58: 182–93.Google Scholar
Hobbs, C, Thoresen, M, Tucker, A, et al. Xenon and hypothermia combine additively, offering long-term functional and histopathologic neuroprotection after neonatal hypoxia/ischemia. Stroke 2008; 39: 1307–13.Google Scholar
Chakkarapani, E, Dingley, J, Aquilina, K, et al. Effects of xenon and hypothermia on cerebrovascular pressure reactivity in newborn global hypoxic-ischemic pig model. J Cereb Blood Flow Metab 2013; 33: 1752–60.Google Scholar
Dingley, J, Tooley, J, Liu, X, et al. Xenon ventilation during therapeutic hypothermia in neonatal encephalopathy: a feasibility study. Pediatrics 2014; 133: 809–18.Google Scholar
Osredkar, D, Toet, MC, van Rooij, LG, et al. Sleep-wake cycling on amplitude-integrated electroencephalography in term newborns with hypoxic-ischemic encephalopathy. Pediatrics 2005; 115: 327–32.Google Scholar
Hellstrom-Westas, L, Rosen, I, Svenningsen, NW. Predictive value of early continuous amplitude integrated EEG recordings on outcome after severe birth asphyxia in full term infants. Arch Dis Child Fetal Neonatal Ed 1995; 72: F34–8.Google Scholar
Toet, MC, Hellstrom-Westas, L, Groenendaal, F, et al. Amplitude integrated EEG 3 and 6 hours after birth in full term neonates with hypoxic-ischaemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 1999; 81: F1923.Google Scholar
Thoresen, M, Hellstrom-Westas, L, Liu, X, de Vries, LS. Effect of hypothermia on amplitude-integrated electroencephalogram in infants with asphyxia. Pediatrics 2010; 126: e131–9.Google Scholar
Ancora, G, Maranella, E, Grandi, S, et al. Early predictors of short term neurodevelopmental outcome in asphyxiated cooled infants: a combined brain amplitude integrated electroencephalography and near infrared spectroscopy study. Brain Dev 2013; 35: 2631.Google Scholar
Toet, MC, Lemmers, PM, van Schelven, LJ, van Bel, F. Cerebral oxygenation and electrical activity after birth asphyxia: their relation to outcome. Pediatrics 2006; 117: 333–9.Google Scholar

References

Low, JA, Lindsay, BG, Derrick, EJ. Threshold of metabolic acidosis associated with newborn complications. Am J Obstet Gynecol 1997; 177: 1391–4.Google Scholar
Gunn, AJ, Parer, JT, Mallard, EC, et al. Cerebral histologic and electrocorticographic changes after asphyxia in fetal sheep. Pediatr Res 1992; 31: 486–91.Google Scholar
George, S, Gunn, AJ, Westgate, JA, et al. Fetal heart rate variability and brainstem injury after asphyxia in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol 2004; 287: R925–33.Google Scholar
Gunn, AJ, Gunn, TR, de Haan, HH, et al. Dramatic neuronal rescue with prolonged selective head cooling after ischemia in fetal lambs. J Clin Invest 1997; 99: 248–56.Google Scholar
Tan, WK, Williams, CE, During, MJ, et al. Accumulation of cytotoxins during the development of seizures and edema after hypoxic-ischemic injury in late gestation fetal sheep. Pediatr Res 1996; 39: 791–7.Google Scholar
Azzopardi, D, Wyatt, JS, Cady, EB, et al. Prognosis of newborn infants with hypoxic-ischemic brain injury assessed by phosphorus magnetic resonance spectroscopy. Pediatr Res 1989; 25: 445–51.Google Scholar
Roth, SC, Baudin, J, Cady, E, et al. Relation of deranged neonatal cerebral oxidative metabolism with neurodevelopmental outcome and head circumference at 4 years. Dev Med Child Neurol 1997; 39: 718–25.Google Scholar
Lorek, A, Takei, Y, Cady, EB, et al. Delayed (“secondary”) cerebral energy failure after acute hypoxia-ischemia in the newborn piglet: continuous 48-hour studies by phosphorus magnetic resonance spectroscopy. Pediatr Res 1994; 36: 699706.Google Scholar
Blumberg, RM, Cady, EB, Wigglesworth, JS, et al. Relation between delayed impairment of cerebral energy metabolism and infarction following transient focal hypoxia-ischaemia in the developing brain. Exp Brain Res 1997; 113: 130–7.Google Scholar
Bennet, L, Roelfsema, V, Pathipati, P, et al. Relationship between evolving epileptiform activity and delayed loss of mitochondrial activity after asphyxia measured by near-infrared spectroscopy in preterm fetal sheep. J Physiol 2006; 572: 141–54.Google Scholar
Vannucci, RC, Towfighi, J, Vannucci, SJ. Secondary energy failure after cerebral hypoxia-ischemia in the immature rat. J Cereb Blood Flow Metab 2004; 24: 1090–7.Google Scholar
Tsuji, M, Naruse, H, Volpe, J, Holtzman, D. Reduction of cytochrome aa3 measured by near-infrared spectroscopy predicts cerebral energy loss in hypoxic piglets. Pediatr Res 1995; 37: 253–9.Google Scholar
Keogh, MJ, Drury, PP, Bennet, L, et al. Limited predictive value of early changes in EEG spectral power for neural injury after asphyxia in preterm fetal sheep. Pediatr Res 2012; 71: 345–53.Google Scholar
Williams, CE, Gunn, A, Gluckman, PD. Time course of intracellular edema and epileptiform activity following prenatal cerebral ischemia in sheep. Stroke 1991; 22: 516–21.Google Scholar
Gunn, AJ, Thoresen, M. Hypothermic neuroprotection. NeuroRx 2006; 3: 154–69.Google Scholar
Jacobs, SE, Berg, M, Hunt, R, et al. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2013; 1: CD003311.Google Scholar
Guillet, R, Edwards, AD, Thoresen, M, et al. Seven- to eight-year follow-up of the CoolCap trial of head cooling for neonatal encephalopathy. Pediatr Res 2012; 71: 205–9.Google Scholar
Azzopardi, D, Strohm, B, Marlow, N, et al. Effects of hypothermia for perinatal asphyxia on childhood outcomes. N Engl J Med 2014; 371: 140–9.Google Scholar
Shankaran, S, Pappas, A, McDonald, SA, et al. Childhood outcomes after hypothermia for neonatal encephalopathy. N Engl J Med 2012; 366: 2085–92.Google Scholar
Gunn, AJ, Gunn, TR, Gunning, MI, et al. Neuroprotection with prolonged head cooling started before postischemic seizures in fetal sheep. Pediatrics 1998; 102: 1098–106.Google Scholar
Gunn, AJ, Bennet, L, Gunning, MI, et al. Cerebral hypothermia is not neuroprotective when started after postischemic seizures in fetal sheep. Pediatr Res 1999; 46: 274–80.Google Scholar
Sirimanne, ES, Blumberg, RM, Bossano, D, et al. The effect of prolonged modification of cerebral temperature on outcome after hypoxic-ischemic brain injury in the infant rat. Pediatr Res 1996; 39: 591–7.Google Scholar
Thoresen, M, Penrice, J, Lorek, A, et al. Mild hypothermia after severe transient hypoxia-ischemia ameliorates delayed cerebral energy failure in the newborn piglet. Pediatr Res 1995; 37: 667–70.Google Scholar
Tooley, JR, Satas, S, Porter, H, et al. Head cooling with mild systemic hypothermia in anesthetized piglets is neuroprotective. Ann Neurol 2003; 53: 6572.Google Scholar
Thoresen, M, Tooley, J, Liu, X, et al. Time is brain: starting therapeutic hypothermia within three hours after birth improves motor outcome in asphyxiated newborns. Neonatology 2013; 104: 228–33.Google Scholar
Gluckman, PD, Wyatt, JS, Azzopardi, D, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial. Lancet 2005; 365: 663–70.Google Scholar
Westgate, JA, Gunn, AJ, Gunn, TR. Antecedents of neonatal encephalopathy with fetal acidaemia at term. Br J Obstet Gynaecol 1999; 106: 774–82.Google Scholar
Colbourne, F, Auer, RN, Sutherland, GR. Characterization of postischemic behavioral deficits in gerbils with and without hypothermic neuroprotection. Brain Res 1998; 803: 6978.Google Scholar
Alonso-Alconada, D, Broad, KD, Bainbridge, A, 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.Google Scholar
Colbourne, F, Corbett, D. Delayed postischemic hypothermia: a six month survival study using behavioral and histological assessments of neuroprotection. J Neurosci 1995; 15: 7250–60.Google Scholar
Colbourne, F, Corbett, D, Zhao, Z, et al. Prolonged but delayed postischemic hypothermia: a long-term outcome study in the rat middle cerebral artery occlusion model. J Cereb Blood Flow Metab 2000; 20: 1702–8.Google Scholar
Davidson, JO, Wassink, G, Yuill, CA, et al. How long is too long for cerebral cooling after ischemia in fetal sheep? J Cereb Blood Flow Metab 2015; 35: 751–8.Google Scholar
Shankaran, S, Laptook, AR, Pappas, A, et al. Effect of depth and duration of cooling on deaths in the NICU among neonates with hypoxic ischemic encephalopathy: a randomized clinical trial. JAMA 2014; 312: 2629–39.Google Scholar
Johnston, MV. Excitotoxicity in perinatal brain injury. Brain Pathol 2005; 15: 234–40.Google Scholar
Bruno, VM, Goldberg, MP, Dugan, LL, et al. Neuroprotective effect of hypothermia in cortical cultures exposed to oxygen-glucose deprivation or excitatory amino acids. J Neurochem 1994; 63: 1398–406.CrossRefGoogle ScholarPubMed
Wassink, G, Gunn, ER, Drury, PP, et al. The mechanisms and treatment of asphyxial encephalopathy. Front Neurosci 2014; 8: 40.Google Scholar
Northington, FJ, Zelaya, ME, O’Riordan, DP, et al. Failure to complete apoptosis following neonatal hypoxia-ischemia manifests as “continuum” phenotype of cell death and occurs with multiple manifestations of mitochondrial dysfunction in rodent forebrain. Neuroscience 2007; 149: 822–33.Google Scholar
Edwards, AD, Yue, X, Squier, MV, et al. Specific inhibition of apoptosis after cerebral hypoxia-ischaemia by moderate post-insult hypothermia. Biochem Biophys Res Commun 1995; 217: 1193–9.Google Scholar
Bennet, L, Roelfsema, V, George, S, et al. The effect of cerebral hypothermia on white and grey matter injury induced by severe hypoxia in preterm fetal sheep. J Physiol 2007; 578: 491506.Google Scholar
Hagberg, H, Mallard, C, Jacobsson, B. Role of cytokines in preterm labour and brain injury. BJOG. 2005; 112(Suppl 1): 1618.Google Scholar
Roelfsema, V, Bennet, L, George, S, et al. The window of opportunity for cerebral hypothermia and white matter injury after cerebral ischemia in near-term fetal sheep. J Cereb Blood Flow Metab 2004; 24: 877–86.Google Scholar
Welin, AK, Sandberg, M, Lindblom, A, et al. White matter injury following prolonged free radical formation in the 0.65 gestation fetal sheep brain. Pediatr Res 2005; 58: 100–5.CrossRefGoogle ScholarPubMed
Fraser, M, Bennet, L, van Zijl, PL, et al. Extracellular amino acids and peroxidation products in the periventricular white matter during and after cerebral ischemia in preterm fetal sheep. J Neurochem 2008; 105: 2214–23.Google Scholar
Tan, WK, Williams, CE, Gunn, AJ, et al. Suppression of postischemic epileptiform activity with MK-801 improves neural outcome in fetal sheep. Ann Neurol 1992; 32: 677–82.Google Scholar
George, SA, Barrett, RD, Bennet, L, et al. Nonadditive neuroprotection with early glutamate receptor blockade and delayed hypothermia after asphyxia in preterm fetal sheep. Stroke 2012; 43: 3114–7.Google Scholar
Bennet, L, Dean, JM, Wassink, G, Gunn, AJ. Differential effects of hypothermia on early and late epileptiform events after severe hypoxia in preterm fetal sheep. J Neurophysiol 2007; 97: 572–8.Google Scholar
Biagioni, E, Bartalena, L, Boldrini, A, et al. Electroencephalography in infants with periventricular leukomalacia: prognostic features at preterm and term age. J Child Neurol 2000; 15: 16.Google Scholar
Dean, JM, George, SA, Wassink, G, et al. Suppression of post hypoxic-ischemic EEG transients with dizocilpine is associated with partial striatal protection in the preterm fetal sheep. Neuropharmacology 2006; 50: 491503.Google Scholar
Bennet, L, Booth, L, Gunn, AJ. Potential biomarkers for hypoxic-ischemic encephalopathy. Semin Fetal Neonatal Med 2010; 15: 253–60.Google Scholar
Jensen, EC, Bennet, L, Hunter, CJ, et al. Post-hypoxic hypoperfusion is associated with suppression of cerebral metabolism and increased tissue oxygenation in near-term fetal sheep. J Physiol 2006; 572: 131–9.Google Scholar
Elstad, M, Whitelaw, A, Thoresen, M. Cerebral resistance index is less predictive in hypothermic encephalopathic newborns. Acta Paediatr 2011; 100: 1344–9.Google Scholar
Laptook, AR, Corbett, RJ, Sterett, R, et al. Quantitative relationship between brain temperature and energy utilization rate measured in vivo using 31P and 1H magnetic resonance spectroscopy. Pediatr Res 1995; 38: 919–25.Google Scholar
Lotgering, FK, Bishai, JM, Struijk, PC, et al. Ten-minute umbilical cord occlusion markedly reduces cerebral blood flow and heat production in fetal sheep. Am J Obstet Gynecol 2003; 189: 233–8.Google Scholar
Hunter, CJ, Bennet, L, Power, GG, et al. Key neuroprotective role for endogenous adenosine A1 receptor activation during asphyxia in the fetal sheep. Stroke 2003; 34: 2240–5.Google Scholar
Mortola, JP. Implications of hypoxic hypometabolism during mammalian ontogenesis. Respir Physiol Neurobiol 2004; 141: 345–56.Google Scholar
Guan, J, Bennet, L, Gluckman, PD, Gunn, AJ. Insulin-like growth factor-1 and post-ischemic brain injury. Prog Neurobiol 2003; 70: 443–62.Google Scholar
Clawson, TF, Vannucci, SJ, Wang, GM, et al. Hypoxia-ischemia-induced apoptotic cell death correlates with IGF-I mRNA decrease in neonatal rat brain. Biol Signals Recept 1999; 8: 281–93.Google Scholar
Guan, J, Bennet, L, George, S, et al. Insulin-like growth factor-1 reduces postischemic white matter injury in fetal sheep. J Cereb Blood Flow Metab 2001; 21: 493502.Google Scholar
George, SA, Bennet, L, Weaver-Mikaere, L, et al. White matter protection with insulin like-growth factor 1 (IGF-1) and hypothermia is not additive after severe reversible cerebral ischemia in term fetal sheep. Dev Neurosci 2011; 33: 280–7.Google Scholar
Cao, Y, Gunn, AJ, Bennet, L, et al. Insulin-like growth factor (IGF)-1 suppresses oligodendrocyte caspase-3 activation and increases glial proliferation after ischemia in near-term fetal sheep. J Cereb Blood Flow Metab 2003; 23: 739–47.Google Scholar
Corley, SM, Ladiwala, U, Besson, A, Yong, VW. Astrocytes attenuate oligodendrocyte death in vitro through an alpha(6) integrin-laminin-dependent mechanism. Glia 2001; 36: 281–94.Google Scholar
Ohlsson, A, Aher, SM. Early erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2006; 3: CD004863.Google Scholar
Digicaylioglu, M, Lipton, SA. Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-κB signalling cascades. Nature 2001; 412: 641–7.Google Scholar
Statler, PA, McPherson, RJ, Bauer, LA, et al. Pharmacokinetics of high-dose recombinant erythropoietin in plasma and brain of neonatal rats. Pediatr Res 2007; 61: 671–5.Google Scholar
Robertson, NJ, Tan, S, Groenendaal, F, et al. Which neuroprotective agents are ready for bench to bedside translation in the newborn infant? J Pediatr 2012; 160: 544–52.Google Scholar
Nadam, J, Navarro, F, Sanchez, P, et al. Neuroprotective effects of erythropoietin in the rat hippocampus after pilocarpine-induced status epilepticus. Neurobiol Dis 2006; 2: 412–26.Google Scholar
Drury, PP, Davidson, JO, van den Heuij, LG, et al. Status epilepticus after prolonged umbilical cord occlusion is associated with greater neural injury in fetal sheep at term-equivalent. PLoS One 2014; 9: e96530.Google Scholar
Fang, AY, Gonzalez, FF, Sheldon, RA, Ferriero, DM. Effects of combination therapy using hypothermia and erythropoietin in a rat model of neonatal hypoxia-ischemia. Pediatr Res 2013; 73: 12–7.Google Scholar
Fan, X, van Bel, F, van der Kooij, MA, et al. Hypothermia and erythropoietin for neuroprotection after neonatal brain damage. Pediatr Res 2013; 73: 1823.Google Scholar
Traudt, CM, McPherson, RJ, Bauer, LA, et al. Concurrent erythropoietin and hypothermia treatment improve outcomes in a term nonhuman primate model of perinatal asphyxia. Dev Neurosci 2013; 35: 491503.Google Scholar
Wang, H, Zhang, L, Jin, Y. A meta-analysis of the protective effect of recombinant human erythropoietin (rhEPO) for neurodevelopment in preterm infants. Cell Biochem Biophys 2015; 71: 795802.Google Scholar
Zhu, C, Kang, W, Xu, F, et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics 2009; 124: e218–26.CrossRefGoogle ScholarPubMed
Elmahdy, H, El-Mashad, AR, El-Bahrawy, H, et al. Human recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics 2010; 125: e1135–42.Google Scholar
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.Google Scholar
Drury, PP, Davidson, JO, Bennet, L, et al. Partial neural protection with prophylactic low-dose melatonin after asphyxia in preterm fetal sheep. J Cereb Blood Flow Metab 2014; 34: 126–35.Google Scholar
Welin, AK, 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.Google Scholar
Robertson, NJ, Faulkner, S, Fleiss, B, et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain 2013; 136: 90105.Google Scholar
Gitto, E, Romeo, C, Reiter, RJ, et al. Melatonin reduces oxidative stress in surgical neonates. J Pediatr Surg 2004; 39: 184–9; discussion 189.Google Scholar
Bennet, L, Tan, S, Van den Heuij, L, et al. Cell therapy for neonatal hypoxia-ischemia and cerebral palsy. Ann Neurol 2012; 71: 589600.Google Scholar
Drobyshevsky, A, Cotten, CM, Shi, Z, et al. Human umbilical cord blood cells ameliorate motor deficits in rabbits in a cerebral palsy model. Dev Neurosci 2015; 37(4–5): 349–62.Google Scholar
Park, WS, Sung, SI, Ahn, SY, et al. Hypothermia augments neuroprotective activity of mesenchymal stem cells for neonatal hypoxic-ischemic encephalopathy. PLoS One 2015; 10: e0120893.Google Scholar
Min, K, Song, J, Kang, JY, et al. Umbilical cord blood therapy potentiated with erythropoietin for children with cerebral palsy: a double-blind, randomized, placebo-controlled trial. Stem Cells 2013; 31: 581–91.Google Scholar
Bae, SH, Lee, HS, Kang, MS, et al. The levels of pro-inflammatory factors are significantly decreased in cerebral palsy patients following an allogeneic umbilical cord blood cell transplant. Int J Stem Cells 2012; 5: 31–8.Google Scholar
Dickinson, R, Peterson, BK, Banks, P, et al. Competitive inhibition at the glycine site of the N-methyl-d-aspartate receptor by the anesthetics xenon and isoflurane: evidence from molecular modeling and electrophysiology. Anesthesiology 2007; 107: 756–67.Google Scholar
Lobo, N, Yang, B, Rizvi, M, Ma, D. Hypothermia and xenon: novel noble guardians in hypoxic-ischemic encephalopathy? J Neurosci Res 2013; 91: 473–8.Google Scholar
Chakkarapani, E, Dingley, J, Liu, X, et al. Xenon enhances hypothermic neuroprotection in asphyxiated newborn pigs. Ann Neurol 2010; 68: 330–41.Google Scholar
Faulkner, S, Bainbridge, A, Kato, T, et al. Xenon augmented hypothermia reduces early lactate/N-acetylaspartate and cell death in perinatal asphyxia. Ann Neurol 2011; 70: 133–50.Google Scholar
Faulkner, SD, Downie, NA, Mercer, CJ, et al. A xenon recirculating ventilator for the newborn piglet: developing clinical applications of xenon for neonates. Eur J Anaesthesiol 2012; 29: 577–85.Google Scholar
Galinsky, R, Bennet, L, Groenendaal, F, et al. Magnesium is not consistently neuroprotective for perinatal hypoxia-ischemia in term-equivalent models in preclinical studies: a systematic review. Dev Neurosci 2014; 36: 7382.Google Scholar
Doyle, LW, Crowther, CA, Middleton, P, Marret, S. Antenatal magnesium sulfate and neurologic outcome in preterm infants: a systematic review. Obstet Gynecol 2009; 113: 1327–33.Google Scholar
Doyle, LW, Anderson, PJ, Haslam, R, et al. School-age outcomes of very preterm infants after antenatal treatment with magnesium sulfate vs placebo. JAMA 2014; 312: 1105–13.Google Scholar
Thornton, JS, Ordidge, RJ, Penrice, J, et al. Temporal and anatomical variations of brain water apparent diffusion coefficient in perinatal cerebral hypoxic-ischemic injury: relationships to cerebral energy metabolism. Magn Reson Med 1998; 39: 920–7.Google Scholar
Davidson, JO, Green, CR, Bennet, L, Gunn, AJ. Battle of the hemichannels: connexins and pannexins in ischemic brain injury. Int J Dev Neurosci 2014; 45: 6674.Google Scholar
Orellana, JA, Hernandez, DE, Ezan, P, et al. Hypoxia in high glucose followed by reoxygenation in normal glucose reduces the viability of cortical astrocytes through increased permeability of connexin 43 hemichannels. Glia 2010; 58: 329–43.Google Scholar
O’Carroll, SJ, Alkadhi, M, Nicholson, LF, Green, CR. Connexin 43 mimetic peptides reduce swelling, astrogliosis, and neuronal cell death after spinal cord injury. Cell Commun Adhes 2008; 15: 2742.Google Scholar
Davidson, JO, Green, CR, Nicholson, LF, et al. Connexin hemichannel blockade improves outcomes in a model of fetal ischemia. Ann Neurol 2012; 71: 121–32.Google Scholar
Davidson, JO, Drury, PP, Green, CR, et al. Connexin hemichannel blockade is neuroprotective after asphyxia in preterm fetal sheep. PLoS One. 2014; 9: e96558.Google Scholar
Davidson, JO, Green, CR, Nicholson, LF, et al. Connexin hemichannel blockade is neuroprotective after, but not during, global cerebral ischemia in near-term fetal sheep. Exp Neurol 2013; 248: 301–8.Google Scholar
Gunn, AJ, Thoresen, M. Animal studies of neonatal hypothermic neuroprotection have translated well in to practice. Resuscitation 2015; 97: 8890.Google Scholar
Dean, JM, Gunn, AJ, Wassink, G, et al. Endogenous α(2)-adrenergic receptor–mediated neuroprotection after severe hypoxia in preterm fetal sheep. Neuroscience 2006; 142: 615–28.Google Scholar
Guan, J, Bennet, L, George, S, et al. Selective neuroprotective effects with insulin-like growth factor-1 in phenotypic striatal neurons following ischemic brain injury in fetal sheep. Neuroscience 2000; 95: 831–9.Google Scholar

References

Sheth, RD, Hobbs, GR, Mullett, M. Neonatal seizures: incidence, onset, and etiology by gestational age. J Perinatol 1999; 19(1): 40–3.Google Scholar
Abend, NS, Wusthoff, CJ, Goldberg, EM, Dlugos, DJ. Electrographic seizures and status epilepticus in critically ill children and neonates with encephalopathy. Lancet Neurol 2013; 12(12): 1170–9.Google Scholar
Shellhaas, RA, Chang, T, Tsuchida, T, et al. The American Clinical Neurophysiology Society’s guideline on continuous electroencephalography monitoring in neonates. J Clin Neurophysiol 2011; 28(6): 611–7.Google Scholar
Wusthoff, CJ. Diagnosing neonatal seizures and status epilepticus. J Clin Neurophysiol 2013; 30(2): 115–21.Google Scholar
Tsuchida, TN, Wusthoff, CJ, Shellhaas, RA, et al. American Clinical Neurophysiology Society standardized EEG terminology and categorization for the description of continuous EEG monitoring in neonates: report of the American Clinical Neurophysiology Society Critical Care Monitoring Committee. J Clin Neurophysiol 2013; 30(2): 161–73.Google Scholar
Oliveira, AJ, Nunes, ML, Haertel, LM, et al. Duration of rhythmic EEG patterns in neonates: new evidence for clinical and prognostic significance of brief rhythmic discharges. Clin Neurophysiol 2000; 111(9): 1646–53.Google Scholar
Nagarajan, L, Palumbo, L, Ghosh, S. Brief electroencephalography rhythmic discharges (BERDs) in the neonate with seizures: their significance and prognostic implications. J Child Neurol 2011; 26(12): 1529–33.Google Scholar
Berg, AT, Berkovic, SF, Brodie, MJ, et al. Revised terminology and concepts for organization of seizures and epilepsies: report of the ILAE Commission on Classification and Terminology, 2005–2009. Epilepsia 2010; 51(4): 676–85.Google Scholar
Bye, AM, Flanagan, D. Spatial and temporal characteristics of neonatal seizures. Epilepsia 1995; 36(10): 1009–16.Google Scholar
Clancy, RR. The contribution of EEG to the understanding of neonatal seizures. Epilepsia 1996; 37(Suppl 1):S52–9.Google Scholar
Bye, AM, Cunningham, CA, Chee, KY, Flanagan, D. Outcome of neonates with electrographically identified seizures, or at risk of seizures. Pediatr Neurol 1997; 16(3): 225–31.Google Scholar
Pisani, F, Copioli, C, Di Gioia, C, et al. Neonatal seizures: relation of ictal video-electroencephalography (EEG) findings with neurodevelopmental outcome. J Child Neurol 2008; 23(4): 394–8.Google Scholar
Scher, MS. Neonatal seizures: an expression of fetal or neonatal brain disorders. In Stevenson, DK, Benitz, WE, Sunshine, P, et al., eds., Fetal and Neonatal Brain Injury, 4th edn. Cambridge University Press, 2009:499526.Google Scholar
Nagarajan, L, Palumbo, L, Ghosh, S. Classification of clinical semiology in epileptic seizures in neonates. Eur J Paediatr Neurol 2012; 16(2): 118–25.Google Scholar
Ronen, GM, Buckley, D, Penney, S, Streiner, DL. Long-term prognosis in children with neonatal seizures: a population-based study. Neurology 2007; 69(19): 1816–22.Google Scholar
Garfinkle, J, Shevell, MI. Prognostic factors and development of a scoring system for outcome of neonatal seizures in term infants. Eur J Paediatr Neurol 2011; 15(3): 222–9.Google Scholar
Clancy, RR. Prolonged electroencephalogram monitoring for seizures and their treatment. Clin Perinatol 2006; 33(3): 649–65.Google Scholar
Lawrence, R, Mathur, A, Nguyen, T, et al. A pilot study of continuous limited-channel aEEG in term infants with encephalopathy. J Pediatr 2009; 154(6): 835–41.Google Scholar
Murray, DM, Boylan, GB, Ali, I, et al. Defining the gap between electrographic seizure burden, clinical expression and staff recognition of neonatal seizures. Arch Dis Child Fetal Neonatal Ed 2008; 93(3):F187–91.CrossRefGoogle ScholarPubMed
Malone, A, Ryan, CA, Fitzgerald, A, et al. Interobserver agreement in neonatal seizure identification. Epilepsia 2009; 50(9): 2097–101.Google Scholar
Orivoli, S, Facini, C, Pisani, F. Paroxysmal nonepileptic motor phenomena in newborn. Brain Dev 2015; 37(9): 833–9.Google Scholar
Connell, J, Oozeer, R, de Vries, L, et al. Clinical and EEG response to anticonvulsants in neonatal seizures. Arch Dis Child 1989; 64(4): 459–64.CrossRefGoogle ScholarPubMed
Scher, MS, Alvin, J, Gaus, L, et al. Uncoupling of EEG-clinical neonatal seizures after antiepileptic drug use. Pediatr Neurol 2003; 28(4): 277–80.Google Scholar
Parker, S, Zuckerman, B, Bauchner, H, et al. Jitteriness in full-term neonates: prevalence and correlates. Pediatrics 1990; 85(1):1723.Google Scholar
Shuper, A, Zalzberg, J, Weitz, R, Mimouni, M. Jitteriness beyond the neonatal period: a benign pattern of movement in infancy. J Child Neurol 1991; 6(3): 243–5.Google Scholar
Sims, M, Artal, R, Quach, H, Wu, PY. Neonatal jitteriness of unknown origin and circulating catecholamines. J Perinat Med 1986; 14(2): 123–6.CrossRefGoogle ScholarPubMed
Rosman, NP, Donnelly, JH, Braun, MA. The jittery newborn and infant: a review. J Dev Behav Pediatr 1984; 5(5): 263–73.Google Scholar
Coulter, DL, Allen, RJ. Benign neonatal sleep myoclonus. Arch Neurol 1982; 39(3): 191–2.Google Scholar
Resnick, TJ, Moshé, SL, Perotta, L, Chambers, HJ. Benign neonatal sleep myoclonus: relationship to sleep states. Arch Neurol 1986; 43(3): 266–8.Google Scholar
Scher, MS. Pathologic myoclonus of the newborn: electrographic and clinical correlations. Pediatr Neurol 1985; 1(6): 342–8.Google Scholar
Huntsman, RJ, Lowry, NJ, Sankaran, K. Nonepileptic motor phenomena in the neonate. Paediatr Child Health 2008; 13(8): 680–4.CrossRefGoogle ScholarPubMed
Bakker, MJ, van Dijk, JG, van den Maagdenberg, AM, Tijssen, MA. Startle syndromes. Lancet Neurol 2006; 5(6): 513–24.Google Scholar
Volpe, JJ. Neurology of the Newborn, 5th edn. Philadelphia: Saunders Elsevier Health Sciences, 2008.Google Scholar
Ronen, GM, Penney, S, Andrews, W. The epidemiology of clinical neonatal seizures in Newfoundland: a population-based study. J Pediatr 1999; 134(1): 71–5.Google Scholar
Wusthoff, CJ, Dlugos, DJ, Gutierrez-Colina, A, et al. Electrographic seizures during therapeutic hypothermia for neonatal hypoxic-ischemic encephalopathy. J Child Neurol 2011; 26(6): 724–8.CrossRefGoogle ScholarPubMed
Glass, HC, Wusthoff, CJ, Shellhaas, RA, et al. Risk factors for EEG seizures in neonates treated with hypothermia: a multicenter cohort study. Neurology 2014; 82(14): 1239–44.Google Scholar
Isaeva, E, Isaev, D, Holmes, GL. Alteration of synaptic plasticity by neonatal seizures in rat somatosensory cortex. Epilepsy Res 2013; 106(1–2): 280–3.Google Scholar
Miller, SP, Weiss, J, Barnwell, A, et al. Seizure-associated brain injury in term newborns with perinatal asphyxia. Neurology 2002; 58(4): 542–8.Google Scholar
McCabe, BK, Silveira, DC, Cilio, MR, et al. Reduced neurogenesis after neonatal seizures. J Neurosci 2001; 21(6): 2094–103.Google Scholar
Lynch, M, Sayin, U, Bownds, J, et al. Long-term consequences of early postnatal seizures on hippocampal learning and plasticity. Eur J Neurosci 2000; 12(7): 2252–64.Google Scholar
Glass, HC, Glidden, D, Jeremy, RJ, et al. Clinical neonatal seizures are independently associated with outcome in infants at risk for hypoxic-ischemic brain injury. J Pediatr 2009; 155(3): 318–23.Google Scholar
Shankaran, S, Laptook, AR, Ehrenkranz, RA, et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med 2005; 353(15): 1574–84.Google Scholar
Gluckman, PD, Wyatt, JS, Azzopardi, D, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial. Lancet 2005; 365(9460): 663–70.Google Scholar
Murray, DM, Ryan, CA, Boylan, GB, et al. Prediction of seizures in asphyxiated neonates: correlation with continuous video-electroencephalographic monitoring. Pediatrics 2006; 118(1): 41–6.Google Scholar
Glass, HC, Nash, KB, Bonifacio, SL, et al. Seizures and magnetic resonance imaging–detected brain injury in newborns cooled for hypoxic-ischemic encephalopathy. J Pediatr 2011; 159(5): 731–5.Google Scholar
Srinivasakumar, P, Zempel, J, Wallendorf, M, et al. Therapeutic hypothermia in neonatal hypoxic ischemic encephalopathy: electrographic seizures and magnetic resonance imaging evidence of injury. J Pediatr 2013; 163(2): 465–70.Google Scholar
Tekgul, H, Gauvreau, K, Soul, J, et al. The current etiologic profile and neurodevelopmental outcome of seizures in term newborn infants. Pediatrics 2006; 117(4): 1270–80.Google Scholar
Lynch, JK. Epidemiology and classification of perinatal stroke. Semin Fetal Neonatal Med 2009; 14(5): 245–9.Google Scholar
Grunt, S, Mazenauer, L, Buerki, SE, et al. Incidence and outcomes of symptomatic neonatal arterial ischemic stroke. Pediatrics 2015; 135(5):e1220–8.Google Scholar
Golomb, MR, Garg, BP, Carvalho, KS, et al. Perinatal stroke and the risk of developing childhood epilepsy. J Pediatr 2007; 151(4): 409–13.Google Scholar
Suppiej, A, Mastrangelo, M, Mastella, L, et al. Pediatric epilepsy following neonatal seizures symptomatic of stroke. Brain Dev 2015; 38(1):2731.Google Scholar
Wusthoff, CJ, Kessler, SK, Vossough, A, et al. Risk of later seizure after perinatal arterial ischemic stroke: a prospective cohort study. Pediatrics 2011; 127(6):e1550–7.Google Scholar
deVeber, G, Andrew, M, Adams, C, et al. Cerebral sinovenous thrombosis in children. N Engl J Med 2001; 345(6): 417–23.Google Scholar
Berfelo, FJ, Kersbergen, KJ, van Ommen, CH, et al. Neonatal cerebral sinovenous thrombosis from symptom to outcome. Stroke 2010; 41(7): 1382–8.Google Scholar
Scher, MS, Aso, K, Beggarly, ME, et al. Electrographic seizures in preterm and full-term neonates: clinical correlates, associated brain lesions, and risk for neurologic sequelae. Pediatrics 1993; 91(1): 128–34.Google Scholar
Davis, AS, Hintz, SR, Van Meurs, KP, et al. Seizures in extremely low birth weight infants are associated with adverse outcome. J Pediatr 2010; 157(5): 720–5.Google Scholar
Vasudevan, C, Levene, M. Epidemiology and aetiology of neonatal seizures. Semin Fetal Neonatal Med 2013; 18(4): 185–91.Google Scholar
Rozance, PJ, Hay, WW. Hypoglycemia in newborn infants: features associated with adverse outcomes. Biol Neonate 2006; 90(2):7486.Google Scholar
Kotulska, K, Jurkiewicz, E, Domańska-Pakieła, D, et al. Epilepsy in newborns with tuberous sclerosis complex. Eur J Paediatr Neurol 2014; 18(6): 714–21.Google Scholar
Van Hove, JL, Lohr, NJ. Metabolic and monogenic causes of seizures in neonates and young infants. Mol Genet Metab 2011; 104(3): 214–30.Google Scholar
Ficicioglu, C, Bearden, D. Isolated neonatal seizures: when to suspect inborn errors of metabolism. Pediatr Neurol 2011; 45(5): 283–91.Google Scholar
Wusthoff, C. Benign familial neonatal epilepsy. J Pediatr Epilepsy 2012; 2012(1): 143–9.Google Scholar
Olson, HE, Poduri, A, Pearl, PL. Genetic forms of epilepsies and other paroxysmal disorders. Semin Neurol 2014; 34(3): 266–79.Google Scholar
Pryor, DS, Don, N, Macourt, DC. Fifth day fits: a syndrome of neonatal convulsions. Arch Dis Child 1981; 56(10): 753–8.Google Scholar
Kato, M, Saitoh, S, Kamei, A, et al. A longer polyalanine expansion mutation in the ARX gene causes early infantile epileptic encephalopathy with suppression-burst pattern (Ohtahara syndrome). Am J Hum Genet 2007; 81(2): 361–6.CrossRefGoogle ScholarPubMed
Yamamoto, H, Okumura, A, Fukuda, M. Epilepsies and epileptic syndromes starting in the neonatal period. Brain Dev 2011; 33(3): 213–20.Google Scholar
Tavyev Asher, YJ, Scaglia, F. Molecular bases and clinical spectrum of early infantile epileptic encephalopathies. Eur J Med Genet 2012; 55(5):299306.Google Scholar
WHO, ILAE, IRCCS. Guidelines on Neonatal Seizures. Geneva: World Health Organization, 2011.Google Scholar
Shellhaas, RA, Clancy, RR. Characterization of neonatal seizures by conventional EEG and single-channel EEG. Clin Neurophysiol 2007; 118(10): 2156–61.Google Scholar
Frenkel, N, Friger, M, Meledin, I, et al. Neonatal seizure recognition: comparative study of continuous-amplitude integrated EEG versus short conventional EEG recordings. Clin Neurophysiol 2011; 122(6): 1091–7.CrossRefGoogle ScholarPubMed
Shellhaas, RA, Soaita, AI, Clancy, RR. Sensitivity of amplitude-integrated electroencephalography for neonatal seizure detection. Pediatrics 2007; 120(4): 770–7.Google Scholar
Osmond, E, Billetop, A, Jary, S, et al. Neonatal seizures: magnetic resonance imaging adds value in the diagnosis and prediction of neurodisability. Acta Paediatr 2014; 103(8): 820–6.Google Scholar
van Rooij, LG, Toet, MC, van Huffelen, AC, et al. Effect of treatment of subclinical neonatal seizures detected with aEEG: randomized, controlled trial. Pediatrics 2010; 125(2):e358–66.CrossRefGoogle ScholarPubMed
van Rooij, LG, Hellström-Westas, L, de Vries, LS. Treatment of neonatal seizures. Semin Fetal Neonatal Med 2013; 18(4): 209–15.Google Scholar
Wietstock, SO, Bonifacio, SL, McCulloch, CE, et al. Neonatal neurocritical care service is associated with decreased administration of seizure medication. J Child Neurol 2015; 30(9): 1135–41.Google Scholar
Bittigau, P, Sifringer, M, Genz, K, et al. Antiepileptic drugs and apoptotic neurodegeneration in the developing brain. Proc Natl Acad Sci USA 2002; 99(23): 15089–94.Google Scholar
Ikonomidou, C, Turski, L. Antiepileptic drugs and brain development. Epilepsy Res 2010; 88(1):1122.Google Scholar
Booth, D, Evans, DJ. Anticonvulsants for neonates with seizures. Cochrane Database Syst Rev. 2004; 4:CD004218.Google Scholar
Cobo, NH, Sankar, R, Murata, KK, et al. The ketogenic diet as broad-spectrum treatment for super-refractory pediatric status epilepticus: challenges in implementation in the pediatric and neonatal intensive care units. J Child Neurol 2015; 30(2): 259–66.Google Scholar
Painter, MJ, Scher, MS, Stein, AD, et al. Phenobarbital compared with phenytoin for the treatment of neonatal seizures. N Engl J Med 1999; 341(7): 485–9.Google Scholar
Painter, MJ, Pippenger, C, MacDonald, H, Pitlick, W. Phenobarbital and diphenylhydantoin levels in neonates with seizures. J Pediatr 1978; 92(2): 315–9.Google Scholar
Van Orman, CB, Darwish, HZ. Efficacy of phenobarbital in neonatal seizures. Can J Neurol Sci 1985; 12(2): 95–9.Google ScholarPubMed
Gilman, JT, Gal, P, Duchowny, MS, et al. Rapid sequential phenobarbital treatment of neonatal seizures. Pediatrics 1989; 83(5): 674–8.Google Scholar
Pressler, RM, Boylan, GB, Marlow, N, 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(5): 469–77.Google Scholar
Vento, M, de Vries, LS, Alberola, A, et al. Approach to seizures in the neonatal period: a European perspective. Acta Paediatr 2010; 99(4):497501.Google Scholar
Glass, HC. Neonatal seizures: advances in mechanisms and management. Clin Perinatol 2014; 41(1): 177–90.Google Scholar
Mruk, AL, Garlitz, KL, Leung, NR. Levetiracetam in neonatal seizures: a review. J Pediatr Pharmacol Ther 2015; 20(2):7689.Google Scholar
Khan, O, Chang, E, Cipriani, C, et al. Use of intravenous levetiracetam for management of acute seizures in neonates. Pediatr Neurol 2011; 44(4): 265–9.CrossRefGoogle ScholarPubMed
Abend, NS, Gutierrez-Colina, AM, Monk, HM, et al. Levetiracetam for treatment of neonatal seizures. J Child Neurol 2011; 26(4): 465–70.Google Scholar
Pearl, PL, Gospe, SM. Pyridoxal phosphate dependency, a newly recognized treatable catastrophic epileptic encephalopathy. J Inherit Metab Dis 2007; 30(1):24.Google Scholar
Pisani, F, Cerminara, C, Fusco, C, Sisti, L. Neonatal status epilepticus vs recurrent neonatal seizures: clinical findings and outcome. Neurology 2007; 69(23): 2177–85.Google Scholar
McBride, MC, Laroia, N, Guillet, R. Electrographic seizures in neonates correlate with poor neurodevelopmental outcome. Neurology 2000; 55(4): 506–13.Google Scholar
Rosenbaum, P, Paneth, N, Leviton, A, et al. A report: the definition and classification of cerebral palsy April 2006. Dev Med Child Neurol Suppl 2007; 109:814.Google Scholar
Uria-Avellanal, C, Marlow, N, Rennie, JM. Outcome following neonatal seizures. Semin Fetal Neonatal Med 2013; 18(4): 224–32.Google Scholar
Fisher, RS, Acevedo, C, Arzimanoglou, A, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia 2014; 55(4): 475–82.Google Scholar
Sillanpää, M, Camfield, P, Camfield, C. Predicting long-term outcome of childhood epilepsy in Nova Scotia, Canada, and Turku, Finland: validation of a simple scoring system. Arch Neurol 1995; 52(6): 589–92.Google Scholar
Teune, MJ, van Wassenaer, AG, van Dommelen, P, et al. Perinatal risk indicators for long-term neurological morbidity among preterm neonates. Am J Obstet Gynecol 2011; 204(5):396.Google Scholar
Pisani, F, Spagnoli, C. Neonatal seizures: a review of outcomes and outcome predictors. Neuropediatrics 2015; 47(1):1219.Google Scholar
Glass, HC, Hong, KJ, Rogers, EE, et al. Risk factors for epilepsy in children with neonatal encephalopathy. Pediatr Res 2011; 70(5): 535–40.Google Scholar
Bellinger, DC, Wypij, D, Rivkin, MJ, et al. Adolescents with d-transposition of the great arteries corrected with the arterial switch procedure: neuropsychological assessment and structural brain imaging. Circulation 2011; 124(12): 1361–9.Google Scholar
Legido, A, Clancy, RR, Berman, PH. Neurologic outcome after electroencephalographically proven neonatal seizures. Pediatrics 1991; 88(3): 583–96.Google Scholar
deVeber, GA, MacGregor, D, Curtis, R, Mayank, S. Neurologic outcome in survivors of childhood arterial ischemic stroke and sinovenous thrombosis. J Child Neurol 2000; 15(5): 316–24.Google Scholar

References

Committee on Nutrition. Nutritional needs of low birth weight infants. Pediatrics 1985; 75; 976–86.Google Scholar
Grand, RJ, Watkins, JB, Torti, FM. Development of the human gastrointestinal tract. Gastroenterology 1976; 70:790810.Google Scholar
Lebenthal, E, Le, PC. Interactions of determinants in the ontogeny of the gastrointestinal tract: a unified concept. Pediatr Res 1983; 17:1924.Google Scholar
Milla, PJ. Development of intestinal structure and function. In Tanner, MS, Stocks, RJ, eds., Neonatal Gastroenterology: Contemporary Issues. Newcastle upon Tyne: Scholium International, 1984:120.Google Scholar
Cavell, B. Gastric emptying in preterm infants. Acta Paediatr Scand 1979; 68: 725–30.Google Scholar
Yu, VYH. Effect of body position on gastric emptying in the neonate. Arch Dis Child 975; 50: 500–4.Google Scholar
Herbst, JJ, Minton, SD, Book, LS. Gastroesophageal reflux causing respiratory distress and apnea in newborn infants. J Pediatr 1979; 95: 763–8.Google Scholar
Pitcher-Wilmott, R, Shurack, JG, Fox, WW. Decreased lung volume after nasogastric feeding of neonates recovering from respiratory distress. J Pediatr 1979; 96: 914–16.Google Scholar
Toce, SS, Keenan, WJ, Homan, SM. Enteral feeding in very low-birth-weight infants: a comparison of two nasogastric methods. Am J Dis Child 1987; 141: 439–44.Google Scholar
Narayanan, I, Singh, B, Harvey, D. Fat loss during feeding of human milk. Arch Dis Child 1984; 59: 475–7.Google Scholar
Moyer, L, Chan, GM. Clotted feeding tubes with transpyloric feeding of premature infant formula. J Pediatr Gastroenterol Nutr 1982; 1: 55–7.Google Scholar
Lucas, A, Bloom, SR, Aynsley-Green, A. Metabolic and endocrine events at the time of the first feed of human milk in preterm and term infants. Arch Dis Child 1978; 53: 731–6.Google Scholar
Bremer, HJ, Brooke, OG, Orzalesi, M, et al. Nutrition and Feeding of Preterm Infants. Oxford: Blackwell, 1987: 197–8.Google Scholar
Van Caillie, M, Powell, GK. Nasoduodenal versus nasogastric feeding in the very low birth weight infant. Pediatrics 1975; 56: 1065–72.Google Scholar
Whitfield, MF. Poor weight gain of the low birth weight infant fed nasojejunally. Arch Dis Child 1982; 57:597601.Google Scholar
Roy, RN, Pollnitz, RP, Hamilton, JR, et al. Impaired assimilation of nasojejunal feeds in healthy low-birth-weight infants. J Pediatr 1977; 90: 431–4.Google Scholar
Beddis, I, McKenzie, S. Transpyloric feeding in the very low birth weight (1500 gm and below) infant. Arch Dis Child 1979; 54: 213–7.Google Scholar
Whittfield, MF. Transpyloric feeding in infants undergoing intensive care. Arch Dis Child 1980; 55:571.Google Scholar
Lucas, A. Enteral nutrition. In Tsang, RC, Lucas, A, Uauy, R, et al., eds., Nutritional Needs of the Premature Infant. Baltimore: Williams & Wilkins, 1993: 209–23.Google Scholar
Hayhurst, EG, Wyman, M. Morbidity associated with prolonged use of polyvinyl feeding tubes. Am J Dis Child 1975; 129: 72–4.Google Scholar
Rodriguez, JP, Guero, J, Frias, EG, et al. Duodenorenal perforation in a neonate by a tube of silicone rubber during transpyloric feeding. J Pediatr 1978; 92: 113–16.Google Scholar
Dellagrammaticus, HD, Duerden, BI, Milner, RDG. Upper intestinal bacterial flora during transpyloric feeding. Arch Dis Child 1983; 58: 115–19.Google Scholar
Vazquez, C, Arroyos, A, Valls, A. Necrotizing enterocolitis: increased incidence in infants receiving nasoduodenal feeding. Arch Dis Child 1980; 55:826.Google Scholar
Pereira, GR, Lemons, JA. Controlled study of transpyloric and intermittent gavage feeding in the small preterm infant. Pediatrics 1981; 67:6872.Google Scholar
Schanler, RJ, Schulman, RJ, Lau, C, et al. Feeding strategies for premature infants: randomized trial of gastrointestinal priming and tube-feeding method. Pediatrics 1999; 103: 434–9.Google Scholar
Lucas, A, Bloom, SR, Aynsley-Green, A. Gut hormones and “minimal enteral feeding.” Acta Paediatr Scand 1986; 75: 719–23.Google Scholar
Dunn, L, Hulman, S, Weiner, J, et al. Beneficial effects of early hypocaloric enteral feeding on neonatal gastrointestinal function: preliminary report of a randomized trial. J Pediatr 1988; 112: 622–9.Google Scholar
Slagle, TA, Gross, SJ. Effect of early low-volume enteral substrate on subsequent feeding tolerance in very low birth weight infants. J Pediatr 1988; 113: 526–31.Google Scholar
Kliegman, RM. Experimental validation of neonatal feeding practices. Pediatrics 1999; 103: 492–3.Google Scholar
Schulman, RJ, Schanler, RJ, Lau, C, et al. Early feeding, antenatal glucocorticoids, and human milk decrease intestinal permeability in preterm infants. Pediatr Res 1998; 44: 519–23.Google Scholar
Schulman, RJ, Schanler, RJ, Laue, C, et al. Early feeding, feeding tolerance, and lactase activity in premature infants. J Pediatr 1998; 133: 645–9.Google Scholar
Schanler, RJ. Feeding strategies in premature infants. Presented as a lecture at Advances in Perinatal and Pediatric Nutrition Conference, Stanford University, July 1999. Published in course syllabus, 1999: 454–61.Google Scholar
Moyer-Mileur, L, Chan, GM. Nutritional support of very-low-birth-weight infants requiring prolonged assisted ventilation. Am J Dis Child 1986; 140: 929–32.Google Scholar
Unger, A, Goetzman, BW, Chan, C, et al. Nutritional practices and outcome of extremely premature infants. Am J Dis Child 1986; 140: 1027–33.Google ScholarPubMed
Gunn, T, Reaman, G, Outerbridge, EW, et al. Peripheral total parenteral nutrition for premature infants with the respiratory distress syndrome: a controlled study. J Pediatr 1978; 92: 608–13.Google Scholar
Yu, VYH, James, B, Hendry, P, et al. Total parenteral nutrition in very low birthweight infants: a controlled trial. Arch Dis Child 1979; 54: 653–61.Google Scholar
Kerner, JA, Hattner, JAT, Trautman, MS, et al. Postnatal somatic growth in very low birth weight infants on peripheral parenteral nutrition. J Pediatr Perinat Nutr 1988; 2:2734.Google Scholar
Glass, EJ, Hume, R, Lang, MA, et al. Parenteral nutrition compared with transpyloric feeding. Arch Dis Child 1984; 59: 131–5.Google Scholar
Dryburgh, E. Transpyloric feeding in 49 infants undergoing intensive care. Arch Dis Child 1980; 55: 879–2.Google Scholar
Zlotkin, SH, Stallings, VA, Pencharz, PB. Total parenteral nutrition in children. Pediatr Clin North Am 1985; 32:381400.Google Scholar
Cashore, WJ, Sedaghatian, MR, Usher, RH. Nutritional supplements with intravenously administered lipid, protein hydrolysate, and glucose in small premature infants. Pediatrics 1975; 56:816.Google Scholar
Churella, HR, Bachhuber, BS, MacLean, WC. Survey: methods of feeding low-birth-weight infants. Pediatrics 1985; 76: 243–9.Google Scholar
Adamkin, DA. Nutrition in very very low birth weight infants. Clin Perinatol 1986; 13: 419–43.Google Scholar
Higgs, SC, Malan, AF, Heese, HDeV. A comparison of oral feeding and total parenteral nutrition in infants of very low birthweight. S Afr Med J 1974; 48: 2169–73.Google Scholar
Hall, RT, Rhodes, PG. Total parenteral alimentation via indwelling umbilical catheters in the newborn period. Arch Dis Child 1976; 51: 929–34.Google Scholar
Merritt, RJ. Neonatal nutritional support. Clin Consult Nutr Support 1981; 1:10.Google Scholar
Coran, AG. Parenteral nutritional support of the neonate. Tele Session (a group telephone workshop, August 17), Tele Session Corporation, New York, 1981.Google Scholar
Kerner, JA. The use of umbilical catheters for parenteral nutrition. In Kerner, JA, ed., Manual of Pediatric Parenteral Nutrition. New York: Wiley, 1983: 303–6.Google Scholar
Sadig, HF. Broviac catheterization in low birth weight infants: incidence and treatment of associated complications. Crit Care Med 1987; 15:4750.Google Scholar
Kanarek, KS, Kuznicki, MB, Blair, RC. Infusion of total parenteral nutrition via the umbilical artery. J Parenter Enteral Nutr 1991; 15: 71–4.Google Scholar
Nakamura, KT, Sato, Y, Erenberg, A. Evaluation of a percutaneously placed 27-gauge central venous catheter in neonates weighing less than 1200 grams. J Parenter Enteral Nutr 1990; 14: 295–9.Google Scholar
Abdulla, F, Dietrich, KA, Pramanik, AK. Percutaneous femoral venous catheterization in preterm neonates. J Pediatr 1990; 117: 788–91.Google Scholar
Sunshine, P. Fetal gastrointestinal physiology. In Eden, RD, Boehm, FH, eds., Assessment and Care of the Fetus: Physiological, Clinical and Medicolegal Principles. East Norwalk, CT: Appleton & Lange, 1990:93111.Google Scholar
Koldovsky, O, Sunshine, P, Kretchmer, N. Cellular migration of intestinal epithelia in suckling and weaned rats. Nature 1966; 212: 1389–90.Google Scholar
Herbst, JJ, Sunshine, P. Postnatal development of the small intestine of the rat. Pediatr Res 1969; 3:2733.Google Scholar
Sunshine, P, Herbst, JJ, Koldovsky, O, et al. Adaptation of the gastrointestinal tract to extrauterine life. Ann NY Acad Sci 1971; 176:1629.Google Scholar
Herbst, JJ, Sunshine, P, Kretchmer, N. Intestinal malabsorption in infancy and childhood. Adv Pediatr 1969; 16:1164.Google Scholar
Kliegman, RM, Fanaroff, AN. Necrotizing enterocolitis. N Engl J Med 1984; 310: 1093–103.Google Scholar
Kosloske, AM. Pathogenesis and prevention of necrotizing enterocolitis: a hypothesis based on personal observation and a review of the literature. Pediatrics 1984; 74: 1086–92.Google Scholar
Neu, J. Necrotizing enterocolitis:an update. Acta Paediatr 2005; 94: 100–5.Google Scholar
Stevenson, DK, Blakely, ML. Historical perspectives: necrotizing enterocolitis: an inherited or acquired condition? NeoReviews 2006; 7:e125–34.Google Scholar
Gephar, SM, McGrath, JM, Effken, JA, Halpern, MD. Nectrotizing enterocolitis risk: state of the science. Adv Neotatal Care 2012; 12:7789.Google Scholar
Brown, E, Sweet, AY. Preventing necrotizing enterocolitis in neonates. JAMA 1978; 240: 2452–4.Google Scholar
Frantz, ID, L’Heureux, P, Engel, RR, et al. Necrotizing enterocolitis. J Pediatr 1975; 56: 259–63.Google Scholar
Walsh, MC, Kliegman, R, Fanaroff, A. Necrotizing enterocolitis: a practitioner’s perspective. Pediatr Rev 1988; 9: 219–26.Google Scholar
Stoll, BJ, Kanto, WP, Glass, RI, et al. Epidemiology of necrotizing enterocolitis: a case control study. J Pediatr 1980; 96: 447–51.Google Scholar
Holman, RC, Stoll, BJ, Curns, AT, et al. Enterocolitis hospitalizations among neonates in the United States. Paediatr Perinat Epidemiol 2006; 20:498506.Google Scholar
Moonijian, AS, Peckham, G, Fox, W, et al. Necrotizing enterocolitis: endemic vs. epidemic. Pediatr Res 1978; 12:530.Google Scholar
Walsh, MC, Kliegman, RM. Necrotizing enterocolitis: treatment based on staging criteria. Pediatr Clin North Am 1986; 33:179200.Google Scholar
Mata, AG, Rosenpart, RM. Intraobserver variability in the radiographic diagnosis of necrotizing enterocolitis. Pediatrics 1980; 66:6871.Google Scholar
Kirschner, B, Lahr, C, Lahr, D. Detection of increased breath hydrogen in infants with necrotizing enterocolitis. Gastroenterology 1980; 72: A57/1080.Google Scholar
Stevenson, DK, Shahin, SM, Ostrander, CR, et al. Breath hydrogen in preterm infants: correlation with changes in bacterial colonization of the gastrointestinal tract. J Pediatr 1982; 101: 607–10.Google Scholar
Garcia, J, Smith, FR, Cucinelli, SA. Urinary D-lactate in infants with necrotizing enterocolitis. J Pediatr 1984; 104: 268–70.Google Scholar
Rivas, Y, Solans, C, Spivak, W. Serum D-lactic acid level, a new marker for necrotizing enterocolitis. J Pediatr Gastroenterol Nutr 2000; 31:S236.Google Scholar
Bell, MJ, Ternberg, JL, Feigin, RD, et al. Neonatal necrotizing enterocolitis: therapeutic decisions based upon clinical staging. Ann Surg 1978; 187:17.Google Scholar
Kliegman, RM, Hack, M, Jones, P, et al. Epidemiologic study of necrotizing enterocolitis among low-birth-weight infants: absence of identifiable risk factors. J Pediatr 1982; 100: 440–4.Google Scholar
Kerner, JA, Hartman, GE, Sunshine, P. The medical and surgical management of infants with the short bowel syndrome. J Perinatol 1985; 5: 517–21.Google Scholar
Raine, PAM. Neonatal necrotizing enterocolitis. In Reed, GB, Claireaux, AE, Cockburn, F, eds., Diseases of the Fetus and Newborn, 2nd edn. London: Chapman & Hall, 1995: 1485–91.Google Scholar
Dimmitt, RA, Moss, RL. Clinical management of necrotizing enterocolitis. NeoReviews 2001; 2:e110–17.Google Scholar
Israel, EJ, Morera, C. Necrotizing enterocolitis. In Walker, WA, Durie, PR, Hamilton, JR, et al., eds., Pediatric Gastrointestinal Disease, 3rd edn. Ontario: Decker, 2000: 665–76.Google Scholar
Moss, RL, Dimmit, RA, Barnhart, DC, et al. Laparotomy versus peritoneal drainage for necrotizing enterocolitis and perforation. N Engl J Med 2006; 354: 2225–34.Google Scholar
Flake, AW. Necrotizing enterocolitis in preterm infants: is laparotomy necessary? N Engl J Med 2006; 354:2275.Google Scholar
Eyal, F, Sagi, E, Avital, A. Necrotizing enterocolitis in the very low birth weight infant: expressed breast milk feeding compared with parenteral feeding. Arch Dis Child 1982; 57: 274–6.Google Scholar
Ostertag, SG, LaGamma, EF, Reisen, CE, et al. Early enteral feeding does not affect the incidence of necrotizing enterocolitis. Pediatrics 1986; 77: 275–80.Google Scholar
Krousop, RW. The influences of feeding practices. In Brown, EG, Sweet, AY, eds., Necrotizing Enterocolitis. New York:Grune & Stratton, 1980:57.Google Scholar
Book, LS, Herbst, JJ, Jung, AL. Comparison of fast and slow-feeding rate schedules to the development of necrotizing enterocolitis. J Pediatr 1976; 89: 463–6.Google Scholar
Goldman, HI. Feeding and necrotizing enterocolitis. Am J Dis Child 1980; 134: 553–5.Google Scholar
Anderson, DM, Rome, ES, Kliegman, RM. Relationship of endemic necrotizing enterocolitis to alimentation. Pediatr Res 1985; 19:331A.Google Scholar
Book, LS, Herbst, JJ, Jung, AL. Carbohydrate malabsorption in necrotizing enterocolitis. Pediatrics 1976; 57: 201–4.Google Scholar
Sweet, AY. Necrotizing enterocolitis: feeding the neonate weighing less than 1500 grams – nutrition and beyond. In Sunshine, P, ed., Report of the 79th Ross Conference on Pediatric Research. Columbus, OH: Ross Products Division, 1980.Google Scholar
Caple, J, Armentrout, D, Huseby, V, et al. Randomized, controlled trial of slow versus rapid feeding volume advancement in preterm infants. Pediatrics 2004; 114: 1597–600.Google Scholar
Kennedy, KA, Tyson, JE, Chamnanvanikij, S. Early versus delayed initiation of progressive enteral feeding for parenterally fed low birth weight or preterm infants. Cochrane Database Syst Rev 2000; 2:CD001970.Google Scholar
DeLemos, RA, Rogers, JH, McLaughlin, GW. Experimental production of necrotizing enterocolitis in newborn goats. Pediatr Res 1974; 8:380.Google Scholar
Book, LS, Herbst, JJ, Atherton, SO, et al. Necrotizing enterocolitis in low-birth-weight infants fed an elemental formula. J Pediatr 1975; 87: 602–5.Google Scholar
Willis, DM, Chabot, J, Radde, IC, et al. Unsuspected hyperosmolality of oral solutions contributing to necrotizing enterocolitis in very-low-birth-weight infants. Pediatrics 1977; 60: 535–8.Google Scholar
AAP Committee on Nutrition. Commentary on breast feeding and infant formulas including proposed standards for formulas. Pediatrics 1976; 57: 278–85.Google Scholar
White, KC, Harkavy, KZ. Hypertonic formula resulting from added oral medications. Am J Dis Child 1982; 136: 931–3.Google Scholar
Ernst, JA, Williams, JM, Glick, MR. Osmolality of substances used in the intensive care nursery. Pediatrics 1983; 72: 347–52.Google Scholar
Barnard, J, Greene, H, Cotton, R. Necrotizing enterocolitis. In Kretchmer, N, Minkowski, A, eds., Nutritional Adaptation of the Gastrointestinal Tract of the Newborn. Nestle Nutrition, vol. 3. New York: Raven Press, 1983: 103–28.Google Scholar
Udall, JN. Gastrointestinal host defense and necrotizing enterocolitis. J Pediatr 1990; 117:3343.Google Scholar
Eibl, MM, Wolf, HM, Furnkranz, H, et al. Prevention of necrotizing enterocolitis in low-birth-weight infants by IgA-lgG feeding. N Engl J Med 1988; 319:17.Google Scholar
Foster, J, Cole, M. Oral immunoglobulin for preventing necrotizing enterocolitis in preterm and low-birth-weight neonates. Cochrane Database Syst Rev 2004; 1:CD001816.Google Scholar
Barlow, B, Santulli, TV, Heird, WC, et al. An experimental study of acute neonatal necrotizing enterocolitis: the importance of breast milk. J Pediatr Surg 1974; 9: 587–95.Google Scholar
Reisner, SH, Garty, B. Necrotizing enterocolitis despite breast-feeding. Lancet 1977; ii:507.Google Scholar
Kliegman, RM, Pittard, WB, Fanaroff, AA. Necrotizing enterocolitis in neonates fed human milk. J Pediatr 1979; 95: 450–3.Google Scholar
Stevenson, DK, Yang, C, Kerner, JA, et al. Intestinal flora in the second week of life in hospitalized preterm infants fed stored frozen breast milk or a proprietary formula. Clin Pediatr 1985; 24: 338–41.Google Scholar
Lucas, A, Cole, TJ. Breast milk and neonatal necrotising enterocolitis. Lancet 1990; 336: 1519–23.Google Scholar
Neu, J. Necrotizing enterocolitis: the search for a unifying pathogenic theory leading to prevention. Pediatr Clin North Am 1996; 43: 409–32.Google Scholar
Sisk, PM, Lovelady, CA, Dillard, RG, et al. Early human milk feeding is associated with a lower risk of necrotizing enterocolitis in very low birth weight infants. J Perinatol 2007; 27: 428–33.Google Scholar
Kliegman, RM, Fanaroff, AA. Neonatal necrotizing enterocolitis: a nine-year experience. Epidemiology and uncommon observations. Am J Dis Child 1981; 135: 603–14.Google Scholar
Lehmiller, DH, Kanto, WF. Relationship of mesenteric thromboembolism, oral feeding and necrotizing enterocolitis. J Pediatr 1978; 92:96100.Google Scholar
Hillman, LS, Goodwin, SL, Sherman, WR. Identification and measurement of plasticiser in neonatal tissues after umbilical catheters and blood products. N Engl J Med 1975; 292: 381–6.Google Scholar
Touloukian, RJ, Kadaw, A, Spencer, RP. The gastrointestinal complications of umbilical venous exchange transfusion: a clinical and experimental study. Pediatrics 1973; 51:3642.Google Scholar
Davey, AM, Wagner, CL, Cox, C, et al. Feeding premature infants while low umbilical artery catheters are in place: a prospective, randomized trial. J Pediatr 1994; 124: 795–9.Google Scholar
Johnson, L, Bowen, FW, Abbasi, S, et al. Relationship of prolonged pharmacologic serum levels of vitamin E to incidence of sepsis and necrotizing enterocolitis in infants with birth weight 1500 grams or less. Pediatrics 1985; 75: 619–38.Google Scholar
Kerner, JA, Poole, RL, Sunshine, P, et al. High serum vitamin E levels in premature infants receiving MVI-Pediatric. J Pediatr Perinat Nutr 1987; 1:7582.Google Scholar
Neu, J, Masi, M, Stevenson, DK, et al. Effects of asphyxia and oral gentamicin on intestinal lactase in the suckling rat. Pediatr Pharmacol 1981; 1: 215–20.Google Scholar
Czyrko, C, Del Pin, CA, O’Neill, JA, et al. Maternal cocaine abuse and necrotizing enterocolitis: outcome and survival. J Pediatr Surg 1991; 26: 414–18.Google Scholar
Lopez, SL. Time of onset of necrotizing enterocolitis in newborn infants with known prenatal cocaine exposure. Clin Pediatr 1995; 34: 424–9.Google Scholar
Bauer, CR, Morrison, JC, Poole, WK, et al. A decreased incidence of necrotizing enterocolitis after prenatal glucocorticoid therapy. Pediatrics 1984; 73: 682–8.Google Scholar
Halac, E, Halac, J, Begue, EF, et al. Prenatal and postnatal corticosteroid therapy to prevent neonatal necrotizing enterocolitis: a controlled trial. J Pediatr 1990; 117: 132–8.Google Scholar
Muguruma, K, Gray, PW, Tjoelker, LW, et al. The central role of PAF in necrotizing enterocolitis development. Adv Exp Med Biol 1997; 407: 379–82.Google Scholar
Thebaud, B, Lacaze-Masmonteil, T, Watterberg, K. Postnatal glucocorticoids in very preterm infants: “the good, the bad, and the ugly.” Pediatrics 2001; 107: 413–15.Google Scholar
Crissinger, KD. Pathogenesis of necrotizing enterocolitis. Workshop presented at the 25th Clinical Congress of A.S.P.E.N. Chicago: Clinical Congress of the American Society for Parenteral and Enteral Nutrition, 2001: 203–4.Google Scholar
Crissinger, K. Understanding necrotizing enterocolitis: promising directions. Pathophysiology 1999; 5: 247–56.Google Scholar
Kliegman, R, Walker, W, Yolken, R. Necrotizing enterocolitis research agenda for a disease of unknown etiology and pathogenesis. Pediatr Res 1993; 34: 701–8.Google Scholar
Caplan, MS, Jilling, T. The pathophysiology of necrotizing enterocolitis. NeoReviews 2001; 2:e103–9.Google Scholar
Rabinowitz, SS, Dzakpasu, P, Piecuch, S, et al. Platelet-activating factor in infants at risk for necrotizing enterocolitis. J Pediatr 2001; 138: 81–6.Google Scholar
Gonzalez-Crussi, F, Hsueh, W. Experimental model of ischemic bowel necrosis: the role of platelet activating factor and endotoxin. Am J Pathol 1983; 112: 127–35.Google Scholar
Chang, H, Gonzalez-Crussi, F, Hsueh, W, et al. Prevention of experimental necrotizing enterocolitis with intestinal trefoil factor (ITF). Gastroenterology 2000; 118:A197.Google Scholar
Becker, RM, Wue, G, Galanko, JA, et al. Reduced serum amino acid concentrations in infants with necrotizing enterocolitis. J Pediatr 2000; 137: 785–93.Google Scholar
Caplan, MS, Miller-Catchpole, R, Kaup, S, et al. Bifidobacterial supplementation reduces the incidence of necrotizing enterocolitis in a neonatal rat model. Gastroenterology 1999; 117: 577–83.Google Scholar
Butel, MJ, Roland, N, Hilbert, A, et al. Clostridial pathogenicity in experimental necrotizing enterocolitis in gnotobiotic quails and protective role of bifidobacteria. J Med Microbiol 1998; 47: 391–9.Google Scholar
Catala, I, Butel, MJ, Bensaada, M, et al. Oligofructose contributes to the protective role of bifidobacteria in experimental necrotizing enterocolitis in quails. J Med Microbiol 1999; 48:8994.Google Scholar
Garg, P. Necrotizing enterocolitis: newer insights. J Clin Diagn Res 2007; 1:90103.Google Scholar
Lin, HC, Su, BH, Chen, AC, et al. Oral probiotics reduce the incidence and severity of necrotizing enterocolitis in very low birth weight infants. Pediatrics 2005; 115:14.Google Scholar
Bin-Nun, A, Bromiker, R, Wilschanski, M, et al. Oral probiotics prevent necrotizing enterocolitis in very low birth weight neonates. J Pediatr 2005; 147: 192–6.Google Scholar
Kunz, AN, Noel, JM, Fairchok, MP. Two cases of Lactobacillus bacteremia during probiotic treatment of short gut syndrome. J Pediatr Gastoenterol Nutr 2004; 38: 457–8.Google Scholar
Munoz, P, Bouza, E, Cuenca-Estrella, M, et al. Saccharomyces cerevisiae fungemia: an emerging infectious disease. Clin Infect Dis 2005; 40:1625.Google Scholar
Barclay, AR, Stenson, B, Simpson, JH, et al. Probiotics for necrotizing enterocolitis: a systematic review. J Pediatr Gastroenterol Nutr 2007; 45: 569–72.Google Scholar
Hunter, CJ, Upperman, JS, Ford, HR, et al. Understanding the susceptibility of the premature infant to necrotizing enterocolitis (NEC). Pediatr Res 2008; 63: 117–23.Google Scholar
Carlson, SE, Montalto, MB, Ponder, DL, et al. Lower incidence of necrotizing enterocolitis in infants fed a preterm formula with egg phospholipids. Pediatr Res 1998; 44: 491–8.Google Scholar
Sonntag, J, Grimmer, I, Scholz, T, et al. Growth and neurodevelopmental outcome of very low birthweight infants with necrotizing enterocolitis. Acta Paediatr 2000; 89: 528–32.Google Scholar
Vohr, BR, Wright, LL, Dusick, AM, et al. Neurodevelopmental and functional outcomes of extremely low birth weight infants in the National Institute of Child Health and Human Developmental Neonatal Research Network, 1993–1994. Pediatrics 2000; 105: 1216–26.Google Scholar
Akisu, M, Baka, M, Coker, I, et al. Effect of dietary n-3 fatty acids on hypoxia-induced necrotizing enterocolitis in young mice: n-3 fatty acids alter platelet-activating factor and leukotriene B4 production in the intestine. Biol Neonate 1998; 74: 31–8.Google Scholar
Rayyis, SF, Ambalavanan, N, Wright, L, et al. Randomized trial of “slow” versus “fast” feed advancements on the incidence of necrotizing enterocolitis in very low birth weight infants. J Pediatr 1999; 134: 293–7.Google Scholar
La Gamma, E, Browne, L. Feeding practices for infants less than 1500 grams at birth and the pathogenesis of necrotizing enterocolitis. Clin Perinatol 1994; 21:271306.Google Scholar
Brown, E, Sweet, A. Neonatal necrotizing enterocolitis. Pediatr Clin North Am 1982; 29: 114–70.Google Scholar
Schmolzer, G, Urlesberger, B, Haim, M, et al. Multi-modal approach to prophylaxis of necrotizing enterocolitis: a clinical report and review of literature. Pediatr Surg Int 2006; 22: 573–80.Google Scholar
Quan, R, Yang, C, Rubinstein, S, et al. The effect of nutritional additives on anti-infective factors in human milk. Clin Pediatr 1994; 33: 325–8.Google Scholar
Aynsley-Green, A. Metabolic and endocrine interrelation in the human fetus and neonate. Am J Clin Nutr 1985; 41:399417.Google Scholar
Feng, JJ, Kwong, LK, Kerner, JA, et al. Resumption of intestinal maturation upon reintroduction of intraluminal nutrients: functional and biochemical correlations. Clin Res 1987; 35:228A.Google Scholar
Mascarenhas, MR, Kerner, JA, Stallings, VA. Parenteral and enteral nutrition. In Walker, WA, Durie, PR, Hamilton, JR, et al., eds., Pediatric Gastrointestinal Disease, 3rd edn. Ontario: Decker, 2000: 1705–52.Google Scholar
Zenk, KE, Sills, JH, Koeppel, RM. Neonatal Medications and Nutrition: A Comprehensive Guide, 2nd edn. Santa Rosa, CA: NICU Ink Book, 2000: 527–8.Google Scholar
Bin-Nun, A, Booms, C, Sabag, N, et al. Rapid fecal calprotectin (FC) analysis: point of care testing for diagnosing early necrotizing enterocolitis. Am J Perinatol 2015; 32(04):337342.Google Scholar
Fallon, EM, Nehra, D, Potemkin, AK, et al. A.S.P.E.N. clinical guidelines: nutrition support of neonatal patients at risk for necrotizing enterocolitis. J Parenteral Enteral Nutr 2012; 36(5):5–6-523.Google Scholar
Abrams, SA, Schanler, RJ, Lee, ML, et al. Greater mortality and morbidity in extremely preterm infants fed a diet containing cow milk protein products. Breastfeed Med 2014; 9(6): 281–5.Google Scholar
Cristofalo, EA, Schanler, RJ, Blanco, CL, et al. Randomized trial of exclusive human milk versus preterm formula diets in extremely premature infants. J Pediatr 2013; 163(6): 1592–5.Google Scholar
Sullivan, S, Schanler, RJ, Kim, JH, et al. An exclusively human milk-based diet is associated with a lower rate of necrotizing enterocolitis than a diet of human milk and bovine milk-based products. J Pediatr 2010; 156(4): 562–7.Google Scholar
Bertelli, C, Pillonel, T, Torregrossa, A, et al. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clin Infect Dis 2015; 60(6): 924–7.Google Scholar
Repa, A, Thanhaeuser, M, Endress, D, et al. Probiotics (Lactobacillus acidophilus and Bifidobacterium bifidum) prevent NEC in VLBW infants fed breast milk but not formula. Clin Invest 2015; 77(2): 381–8.Google Scholar
Hartel, C, Pagel, J, Rupp, J, et al. Prophylactic use of Lactobacillus acidophilus/Bifidobacterium infantis probiotics and outcome in very low birth weight infants. J Pediatr 2014; 165(2): 285–9.Google Scholar
Jacobs, SE, Tobin, JM, Opie, GF et al. Probiotic effects on late-onset sepsis in very preterm infants: a randomized controlled trial. J Pediatr 2013; 132(6): 1055–62.Google Scholar
Athalye-Jape, G, Rao, S, Patole, S. Lactobacillus reuteri DSM 17938 as a probiotic for preterm neonates: a strain-specific systematic review. J Parenter Enteral Nutr 2015; 40(6): 783–94.Google Scholar

References

Blencowe, H, Cousens, S, Oestergaard, M, et al. National, regional and worldwide estimates of preterm birth. Lancet 2012; 379(9832): 2162–72.Google Scholar
Martin, JA, Hamilton, BE, Osterman, MJK, et al. Births: final data for 2013. National Vital Statistics Reports 2015; 64(1); available at www.cdc.gov/nchs/data/nvsr/nvsr64/nvsr64_01.pdf.Google Scholar
March of Dimes. Peristats, 2015. Available at www.marchofdimes.org/peristats/Peristats.aspx (acessed December 10, 2015).Google Scholar
Raju, TNK, Mercer, BM, Burchfield, DJ, et al. Birth: executive summary of a joint workshop by the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Society for Maternal-Fetal Medicine, American Academy of Pediatrics, and American College of Obstetricians and Gynecologists. Obstet Gynecol 2014; 123: 1083–96.Google Scholar
Stoll, BJ, Hansen, NI, Bell, EF, et al. Trends in care practices, morbidity and mortality of extremely preterm neonates, 1993–2012. JAMA 2015; 314(10): 1039–51.Google Scholar
Zegers, MJ, Hukkelhoven, CWPM, Uiterwaal, CSPM, et al. Changing Dutch approach and trends in short-term outcome of periviable preterms. Arch Dis Child Fetal Neonatal Ed 2016; 101: F391–6.Google Scholar
Anthony, S, den Ouden, L, Brand, R, et al. Changes in perinatal care and survival in very preterm and extremely preterm infants in the Netherlands between 1983–1995. Eur J Obstet Gyn Reprod Biol 2004; 112: 170–7.Google Scholar
Finnstrom, O, Olausson, PO, Sedin, G, et al. The Swedish national prospective study on extremely low birth weight (ELBW) infants: incidence, mortality, morbidity, and survival in relation to level of care. Acta Paediatr 1997; 86: 501–11.Google Scholar
Serenius, F, Ewald, U, Farooqui, A, et al. Short-term outcome after active perinatal management at 23–25 weeks of gestation: a study from two Swedish tertiary care centres. 2. Infant survival. Acta Paediatr 2004; 93: 1081–9.Google Scholar
The EXPRESS Group. One-year survival of extremely preterm infants after active perinatal care in Sweden. JAMA 2009; 301(21): 2225–33.Google Scholar
Costeloe, K, Hennessy, E, Gibson, AT, et al. The EPICure Study: outcomes to discharge from hospital for infants born at the threshold of viability. Pediatrics 2000; 106: 659–71.Google Scholar
Costeloe, K, Henessy, EM, Haider, S, et al. Short term outcomes after extreme preterm birth in England: comparison of two birth cohorts in 1995 and 2006 (the EPICure studies). BMJ 2012; 345: e7976.Google Scholar
Ancel, P-Y, Goffinet, F, EPIPAGE-2 Writing Group. Survival and morbidity of preterm children born at 22 through 34 weeks’ gestation in France in 2011: results of the EPIPAGE-2 Cohort Study. JAMA Pediatr 2015; 169: 230–8.Google Scholar
Doyle, LW, VICS Group. Neonatal intensive care at borderline viability: is it worth it? Early Hum Dev 2004; 80: 103–13.Google Scholar
Doyle, LW. Evaluation of neonatal intensive care for extremely-low-birth-weight infants. Semin Fetal Neonatal Med 2006; 11: 139–45.Google Scholar
Doyle, LW, VICS Group. Evaluation of neonatal intensive care for extremely low birth weight infants in Victoria over two decades. II. Efficiency. Pediatrics 2004; 113: 511–14.Google Scholar
Doyle, LW, Roberts, G, Anderson, PJ, VICS Group. Outcomes at age 2 years of infants < 28 weeks’ gestational age born in Victoria in 2005. J Pediatr 2010; 156: 4953.Google Scholar
Ishii, N, Kono, Y, Yoneoto, N, et al. Outcomes of infants born at 22 and 23 weeks’ gestation. Pediatrics 2013; 132: 6271.Google Scholar
Marlow, N, Bennett, C, Draper, ES, et al. Perinatal outcomes for extremely preterm babies in relation to place of birth in England: the EPICure 2 study. Arch Dis Child Fetal Neonatal Ed 2014; 99: F181–8.Google Scholar
Ochiai, M, Kinjo, T, Takahata, Y, et al. Survival and neurodevelopmental outcome of preterm infants born at 22–24 weeks of gestational age. Neonatology 2014; 105: 7984.Google Scholar
Tyson, JE, Parikh, NA, Langer, J, et al. Intensive care for extreme prematurity: moving beyond gestational age. N Engl J Med 2008; 358(16): 1672–81.Google Scholar
Lee, HC, Green, C, Hintz, SR, et al. Prediction of death for extremely preterm infants in a population-based cohort. Pediatrics 2010; 126: e644–50.Google Scholar
Rysavy, MA, Li, L, Bell, EF, et al. Eunice Kennedy Shriver NICHD Neonatal Research Network: between-hospital variation in treatment and outcomes in extremely preterm infants. N Engl J Med 2015; 372: 1801–11.Google Scholar
Hintz, SR, Bann, CM, Ambalavanan, N, et al. Predicting time to discharge for extremely preterm infants. Pediatrics 2010; 125: e146–54.Google Scholar
Ambalavanan, N, Carlo, WA, Tyson, JE, et al. Outcome trajectories in extremely preterm infants. Pediatrics 2012; 130: e115–25.Google Scholar
Mangham, LJ, Petrou, S, Doyle, LW, et al. The cost of preterm birth throughout childhood in England and Wales. Pediatrics 2009; 123(2):e312–27.Google Scholar
McCabe, ERB, Carrino, GE, Russell, RB, Howse, JL. Fighting for the next generation: US prematurity in 2030. Pediatrics 2014; 134: 1193–9.Google Scholar
Janvier, A, Lantos, J, Deschenes, M, et al. Caregivers attitudes for very premature infants: what if they knew? Acta Pediatr 2008; 97(3): 276–9.Google Scholar
Aylward, GP, Aylward, BS. The changing yardstick in measurement of cognitive abilities in infancy. J Dev Behav Pediatr 2011; 32: 465–8.Google Scholar
Anderson, PJ, De Luca, CR, Hutchinson, E, et al. Underestimation of developmental delay by the new Bayley-III scales. Arch Pediatr Adolesc Med 2010; 164(4): 352–6.Google Scholar
Msall, ME. Measuring outcomes after extreme prematurity with the Bayley-III scales of infant and toddler development: a cautionary tale from Australia. Arch Pediatr Adolesc Med 2010; 164: 391–3.Google Scholar
Marlow, N, Wolke, D, Bracewell, M, et al. Neurologic and developmental disability at six years of age after extremely preterm birth. N Engl J Med 2005; 352: 919.Google Scholar
Saigal, S, Rosenbaum, P. What matters in the long term: reflections on the context of adult outcomes versus detailed measures in childhood. Semin Fetal Neonatal Med 2007; 12: 417–22.Google Scholar
Saigal, S, Tyson, J. Measurement of quality of life of survivors of neonatal intensive care: critique and implications. Semin Perinatol 2008; 32: 5966.Google Scholar
Saigal, S. Premie Voices:Young Men and Women Born Very Prematurely Describe Their Lives, Challenges, and Achievements. 1st edn. Victoria, BC: Friesen Press, 2014.Google Scholar
Hack, M, Flannery, DJ, Schluchter, M, et al. Outcomes in young adulthood for very low birth weight infants. N Engl J Med 2002; 346: 149–57.Google Scholar
Hack, M, Schluchter, M, Forrest, CB, et al. Self-reported adolescent health status of extremely low birth weight children born 1992–1995. Pediatrics 2012; 130(1):4653.Google Scholar
Litt, JS, Gerry Taylor, H, Margevicius, S, et al. Academic achievement of adolescents born with extremely low birth weight. Acta Paediatr 2012; 101(12): 1240–5.Google Scholar
Hack, M. Adult outcomes of preterm children. J Dev Behav Pediatr 2009; 30(5): 460–70.Google Scholar
Hack, M, Taylor, HG, Drotar, D, et. al. Poor predictive validity of the Bayley scales of infant development for cognitive function of extremely low birth weight children at school age. Pediatrics 2005; 116: 333–41.Google Scholar
Ment, LR, Vohr, B, Allan, W, et. al. Change in cognitive function over time in very low-birth-weight infants. JAMA 2003; 289: 705–11.Google Scholar
Spittle, A, Orton, J, Anderson, PJ, et al. Early developmental intervention programmes provided post hospital discharge to prevent motor and cognitive impairment in preterm infants. Cochrane Database Syst Rev 2015; 11: CD005495.Google Scholar
Davis, NM, Ford, GW, Anderson, PJ, Doyle, LW. Developmental coordination disorder at 8 years of age in a regional cohort of extremely-low-birthweight or very preterm infants. Dev Med Child Neurol 2007; 49: 325–30.Google Scholar
Roberts, G, Anderson, PJ, Davis, N, et al. Developmental coordination disorder in geographic cohorts of 8-year-old children born extremely preterm or extremely low birth weight in the 1990s. Dev Med Child Neurol 2011; 53: 5560.Google Scholar
Bayley, N. Bayley Scales of Infant and Toddler Development, 3rd edn. San Antonio, TX: Harcourt Assessment, 2006.Google Scholar
Vohr, BR, Stephens, BE, Higgins, RD, et al. Are outcomes of extremely preterm infants improving? Impact of Bayley assessment on outcomes. J Pediatr 2012; 161: 222–8.Google Scholar
Duncan, AF, Bann, C, Boatman, C, et al. Do currently recommended Bayley-III cutoffs overestimate motor impairment in infants born < 27 weeks gestation? J Perinatol 2015; 35: 516–21.Google Scholar
Spittle, AJ, Spencer-Smith, MM, Eeles, AL, et al. Does the Bayley-III mkotor scale at 2 years predict motor outcome at 4 years on very preterm children? Dev Med Child Neurol 2013; 55: 448–52.Google Scholar
Spencer-Smith, MM, Spittle, AJ, Lee, KJ, et al. Bayley-III cognitive and language scales in preterm children. Pediatrics 2015; 135: e1258–65.Google Scholar
Johnson, S, Moore, T, Marlow, N. Using the Bayley-III to assess neurodevelopmental delay: which cut-off should be used? Pediatr Res 2014; 75: 670–4.Google Scholar
Palisano, R, Rosenbaum, P, Walter, S, et al. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Dev Med Child Neurol 1997; 39: 214–23.Google Scholar
Victorian Infant Collaborative Study Group. Economic outcome for intensive care of infants of birthweight 500–999 g born in Victoria in the post-surfactant era. J Paediatr Child Health 1997; 33: 202–8.Google Scholar
Hutchinson, EA, De Luca, CR, Doyle, LW, et al. School age outcomes of extremely preterm or extremely low birth weight children. Pediatrics 2013; 131(4): e1053–61.Google Scholar
Burnett, AC, Scratch, SE, Lee, KJ, et al. Executive function in adolescents born < 1000 g or <28 weeks: a prospective cohort study. Pediatrics. 2015; 135(4):e826–34Google Scholar
Burnett, A, Davey, CG, Wood, SJ, et al. Extremely preterm birth and adolescent mental health in a geographical cohort born in the late 1990s. Psychol Med 2014; 44(7): 1533–44.Google Scholar
Roberts, G, Burnett, AC, Lee, KJ, et al. Quality of life at age 18 years after extremely preterm birth in the post-surfactant era. J Pediatr 2013; 163(4): 1008–13.Google Scholar
Moore, T, Hennessy, EM, Myles, J, et al. Neurological and developmental outcome in extremely preterm children born in England in 1995 and 2006: the EPICure studies. BMJ 2012; 345: e7961.Google Scholar
Moore, T, Johnson, S, Haider, S, et al. Interpreting developmental test scores over time: using the scond and third editions of the Bayley scales of infant and toddler development. J Pediatr 2012; 160: 553–8.Google Scholar
Moore, T, Johnson, S, Hennessy, E, Marlow, N. Screening for autism in extremely preterm infants: problems in interpretation. Dev Med Child Neurol 2012; 54(6): 514–20.Google Scholar
Kuban, KC, O’Shea, TM, Allred, EN, et al. Positive screening on the Modified Checklist for Autism in Toddlers (M-CHAT) in extremely low gestational age newborns. J Pediatr 2009; 154: 535–40.Google Scholar
Limperopoulos, C, Bassan, H, Sullivan, NR, et al. Positive screening for autism in ex-preterm infants: prevalence and risk factors. Pediatrics 2008; 121: 758–65.Google Scholar
Serenius, F, Kallen, K, Blennow, M, et al. Neurodevelopmental outcome in extremely preterm infants at 2.5 years after active perinatal care in Sweden. JAMA 2013; 309: 1810–20.Google Scholar
Serenius, F, Blennow, M, Marsal, K, et al. Intensity of perinatal care for extremely preterm infants: outcomes at 2.5 years. Pediatrics 2015; 135: e1163–72.Google Scholar
Hintz, SR, Kendrick, DE, Wilson-Costello, D, et al. Neurodevelopmental outcomes at 18–22 months are not improving for <25 week EGA infants. Pediatrics 2011; 127: 6270.Google Scholar
Vohr, BR, Wright, LL, Dusick, AM, et al. Center differences and outcomes of extremely low birth weight infants. Pediatrics 2004; 113: 781–9.Google Scholar
Schlapbach, LJ, Adams, M, Prioletti, E, et al. Outcome at two years of age in a Swiss national cohort of extremely preterm infants born between 2000 and 2008. BMC Pediatr 2012; 12: 198.Google Scholar
Schlapbach, LJ, Aebischer, M, Adams, M, et al. Impact of sepsis on neurodevelopmental outcome in a Swiss national cohort of extremely premature infants. Pediatrics 2011; 128: e348–57.Google Scholar
Pape, KE, Blackwell, RJ, Cusick, G, et al. Ultrasound detection of brain damage in preterm infants. Lancet 1979; 1(8129): 1261–4.Google Scholar
Slovis, TL, Kuhn, LR. Real-time sonography of the brain through the anterior fontanelle. AJR 1981; 136: 277–86.Google Scholar
Papile, L-A, Burstein, J, Burstein, R, Koffler, H. Incidence and evolution of subependymal and intraventricular hemorrhage: a study of infants with birth weight less than 1500 gm. J Pediatr 1978; 92: 529–34.Google Scholar
Ment, LR, Bada, HS, Barnes, P, et al. Practice parameter: neuroimaging of the neonate. Report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 2002; 58: 1726–38.Google Scholar
Ancel, P-Y, Livinec, F, Larroque, B, et al. Cerebral palsy among very preterm children in relation to gestational age and neonatal ultrasound abnormalities: the EPIPAGE Cohort Study. Pediatrics 2006; 117: 828–35.Google Scholar
de Vries, LS, Van Haastert, IC, Rademaker, KJ, et al. Ultrasound abnormalities preceding cerebral palsy in high-risk preterm infants. J Pediatr 2004; 144: 815–20.Google Scholar
de Vries, LS, van Haastert, IC, Benders, MJNL, Groenendaal, F. Myth: cerebral palsy cannot be predicted by neonatal brain imaging. Semin Fetal Neonatal Med. 2011; 16(5): 279–87.Google Scholar
O’Shea, TM, Allred, EN, Dammann, O, et al. The ELGAN study of the brain and related disorders in extremely low gestational age newborns. Early Hum Dev 2009; 85: 719–25.Google Scholar
Kuban, KCK, Allred, EN, O’Shea, TM, et al. Cranial ultrasound lesion in the NICU predict cerebral palsy at age 2 years in children born at extremely low gestational age. J Child Neurol 2009; 24: 6372.Google Scholar
Wood, NS, Costeloe, K, Gibson, AT, et al. The EPICure Study: association and antecedents of neurological and developmental disability at 30 months of age following extremely preterm birth. Arch Dis Child Fetal Neonatal Ed 2005; 90: F134–40.Google Scholar
Hack, M, Wilson-Costello, D, Friedman, H, et al. Neurodevelopment and predictors of outcomes of children with birth weights of less than 1000 g, 1992–1995. Arch Pediatr Adolesc Med 2000; 154: 725–31.Google Scholar
Broitman, E, Ambalavanan, N, Higgins, R, et al. Clinical data predict neurodevelopmental outcome better than head ultrasound in extremely low birth weight infants. J Pediatr 2007; 151: 500–5.Google Scholar
Laptook, AR, O’Shea, RM, Shankaran, S, et al. Adverse neurodevelopmental outcomes among extremely low birth weight infants with a normal head ultrasound: prevalence and antecedents. Pediatrics 2005; 115: 673–80.Google Scholar
O’Shea, TM, Allred, EN, Kuban, KCK, et al. Intraventricular hemorrhage and developmental outcomes at 24 months of age in extremely preterm infants. J Child Neurol 2012; 27(1): 22–9.Google Scholar
Beaino, G, Khoshnood, B, Kaminski, M, et al. Predictors of cerebral palsy in very preterm infants: the EPIPAGE prospective population-based cohort study. Dev Med Child Neurol 2010; 52: e119–25.Google Scholar
Marret, S, Marchand-Martin, L, Picaud, J-C, et al. Brain injury in very preterm children and neurosensory and cognitive disabilities during childhood: the EPIPAGE Cohort Study. PLoS One 2013; 8: e62683.Google Scholar
Hintz, SR, Slovis, T, Bulas, D, et al. Interobserver reliability and accuracy of cranial ultrasound scanning in premature infants. J Pediatr 2007; 150: 592–6.Google Scholar
Limperopoulos, C, Bassan, H, Gauvreau, K, et al. Does cerebellar injury in premature infants contribute to the high prevalence of long-term cognitive, learning and behavioral disability in survivors? Pediatrics 2007; 120: 584–93.Google Scholar
Limperopoulos, C, Chilingaryan, G, Sullivan, N, et al. Injury to the premature cerebellum: outcome is related to remote cortical development. Cereb Cortex 2014; 24(3): 728–36.Google Scholar
Pierrat, V, Duquennoy, C, van Haastert, IC, et al. Ultrasound diagnosis and neurodevelopmental outcome of localized and extensive cystic periventricular leucomalacia. Arch Dis Child Fetal Neonatal Ed 2001; 84: F151–6.Google Scholar
Bassan, H, Limperopoulos, C, Visconti, K, et al. Neurodevelopmental outcome in survivors of periventricular hemorrhagic infarction. Pediatrics 2007; 120: 785–92.Google Scholar
Davis, AS, Hintz, SR, Bann, CM, et al. Characteristics of severe intracranial hemorrhage and other factors in predicting early childhood outcomes of extremely preterm infants. J Perinatol 2014; 34(3): 203–8.Google Scholar
Merhar, SL, Tabangin, ME, Meinzen-Derr, J, Schibler, KR. Fetal and postnatal brain MRI in premature infants with twin-twin transfusion syndrome. J Perinatol 2013; 33: 112–18.Google Scholar
Volpe, JJ, Kinney, HC, Jensen, FE, Rosenberg, PA. The developing oligodendrocyte: key cellular target in brain injury in the premature infant. Int J Dev Neurosci 2011; 29: 423–40.Google Scholar
Dyet, LE, Kennea, N, Counsell, SJ, et al. Natural history of brain lesions in extremely preterm infants studied with serial magnetic resonance imaging from birth and neurodevelopmental assessment. Pediatrics 2005; 118: 536–48.Google Scholar
Inder, TE, Warfield, SK, Wang, H, et al. Abnormal cerebral structure is present at term in premature infants. Pediatrics 2005; 115: 286–94.Google Scholar
Inder, TE, Huppi, PS, Warfield, S, et al. Periventricular white matter injury in the premature infant is followed by reduced cerebral cortical gray matter volume at term. Ann Neurol 1999; 46: 755–60.Google Scholar
Boardman, JP, Counsell, SJ, Rueckert, D, et al. Abnormal deep grey matter development following preterm birth detected using deformation-based morphometry. NeuroImage 2006; 32: 70–8.Google Scholar
Thompson, DK, Warfield, SK, Carlin, JB, et al. Perinatal risk factors altering regional brain structure in the preterm infant. Brain 2007; 130: 667–77.Google Scholar
Peterson, BS, Anderson, AW, Ehrenkranz, R, et al. Regional brain volumes and their later neurodevelopmental correlates in term and preterm infants. Pediatrics 2003; 111: 939–48.Google Scholar
Dean, JM, Bennet, L, Back, SA, et al. What brakes the preterm brain? An arresting story. Pediatric Res 2014; 75: 227–33.Google Scholar
Roelants-van Rijn, AM, Groenendaal, F, Beek, FJA, et al. Parenchymal brain injury in the preterm infant: comparison of cranial ultrasound, MRI and neurodevelopmental outcome. Neuropediatrics 2001; 32: 80–9.Google Scholar
Mirmiran, M, Barnes, PD, Keller, K, et al. Neonatal brain magnetic resonance imaging before discharge is better than serial cranial ultrasound in predicting cerebral palsy in very low birth weight preterm infants. Pediatrics 2004; 114: 992–8.Google Scholar
Sie, LTL, Hart, AAM, van Hof, J, et al. Predictive value of neonatal MRI with respect to late MRI findings and clinical outcome: a study in infants with periventricular densities on neonatal ultrasound. Neuropediatrics 2005; 36: 7889.Google Scholar
Woodward, LJ, Anderson, PJ, Austin, NC, et al. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med 2006; 355: 685–94.Google Scholar
Hintz, SR, Barnes, PD, Bulas, D, et al. Neuroimaging and neurodevelopmental outcome in extremely preterm infants. Pediatrics 2015; 135: e3242.Google Scholar
Skiöld, B, Eriksson, C, Eliasson, A-C, et al. General movements and magnetic resonance imaging in the prediction of neuromotor outcome in children born extremely preterm. Early Hum Dev 2013; 89(7): 467–72.Google Scholar
Jeon, TY, Kim, JH, Yoo, SY, et al. Neurodevelopmental outcomes in preterm infants: comparison of infants with and without diffuse excessive high signal intensity on MR images at near-term-equivalent age. Radiology 2012; 263(2): 518–26.Google Scholar
Skiöld, B, Vollmer, B, Böhm, B, et al. Neonatal magnetic resonance imaging and outcome at age 30 months in extremely preterm infants. J Pediatr 2012; 160(4): 559–66.Google Scholar
Janvier, A, Barrington, K. Trying to predict the future of ex-preterm infants: who benefits from a brain MRI at term? Acta Paediatr 2012; 101(10): 1016–17.Google Scholar
Ho, T, Dukhovny, D, Zupancic, JAF, et al. Choosing wisely in newborn medicine: five opportunities to increase value. Pediatrics 2015; 136: e482–9.Google Scholar
Tam, EW, Rosenbluth, G, Rogers, EE, et al. Cerebellar hemorrhage on magnetic resonance imaging in preterm newborns associated with abnormal neurologic outcome. J Pediatr 2011; 158(2): 245–50.Google Scholar
Steggerda, SJ, de Bruine, FT, van den Berg-Huysmans, AA, et al. Small cerebellar hemorrhage in preterm infants: perinatal and postnatal factors and outcome. Cerebellum 2013; 12(6):794801.Google Scholar
Sie, LTL, Hart, AAM, van Hof, J, et al. Predictive value of neonatal MRI with respect to late MRI findings and clinical outcome: a study in infants with periventricular densities on neonatal ultrasound. Neuropediatrics 2005; 36(2):7889.Google Scholar
Spittle, AJ, Boyd, RN, Inder, TE, Doyle, LW. Predicting motor development in very preterm infants at 12 months’ corrected age: the role of qualitative magnetic resonance imaging and general movements assessments. Pediatrics 2009; 123(2): 512–17.Google Scholar
Skiöld, B, Eriksson, C, Eliasson, A-C, et al. General movements and magnetic resonance imaging in the prediction of neuromotor outcome in children born extremely preterm. Early Hum Dev 2013; 89(7): 467–72.Google Scholar
Glass, HC, Bonifacio, SL, Chau, V, et al. Recurrent postnatal infections are associated with progressive white matter injury in premature infants. Pediatrics 2008; 122: 299305.Google Scholar
Shah, DK, Doyle, LW, Anderson, PJ, et al. Adverse neurodevelopment in preterm infants with postnatal sepsis or necrotizing enterocolitis is mediated by white matter abnormalities on magnetic resonance imaging at term. J Pediatr 2008; 153: 170–5.Google Scholar
Mathur, AM, Neil, JJ, Inder, TE. Understanding brain injury and neurodevelopmental disabilities in the preterm infant: the evolving role of advanced magnetic resonance imaging. Semin Perinatol 2010; 34: 5766.Google Scholar
Doyle, LW, Clucas, L, Roberts, G, et al. The cost of long-term follow up of high-risk infants for research studies. J Paediatr Child Health 2015; 51: 1012–16.Google Scholar

References

Colver, A, Fairhurst, C, Pharoah, PO. Cerebral palsy. Lancet 2014; 383: 1240–9.Google Scholar
Pakula, AT, Van Naarden Braun, K, Yeargin-Allsopp, M. Cerebral palsy: classification and epidemiology. Phys Med Rehabil Clin North Am 2009; 20: 425–52.Google Scholar
Bax, MC. Terminology and classification of cerebral palsy. Dev Med Child Neurol 1964; 6:295.Google Scholar
Rosenbaum, P, Paneth, N, Leviton, A, et al. A report: the definition and classification of cerebral palsy April 2006. Dev Med Child Neurol Suppl 2007; 109:9.Google Scholar
World Health Organization. International classification of functioning, disability, and health, 2001. Available at http://who.int/classifications/icf/icf_more/en/ (accessed May 23, 2015).Google Scholar
Rosenbaum, P, Stewart, D. World Health Organization International Classification of Functioning, Disability, and Health: a model to guide clinical thinking, practice and research in the field of cerebral palsy. Semin Pediatr Neurol 2004; 11:510.Google Scholar
Chan, G, Miller, F. Assessment and treatment of children with cerebral palsy. Orthop Clin North Am 2014; 45: 313–25.Google Scholar
Foran, JR, Steinman, S, Barash, I, et al. Structural and mechanical alterations in spastic skeletal muscle. Dev Med Child Neurol 2005; 47: 713–7.Google Scholar
Bax, M, Tydeman, C, Flodmark, O. Clinical and MRI correlates of cerebral palsy: the European Cerebral Palsy Study. JAMA 2006; 296: 1602–8.Google Scholar
Shevell, MI, Dagenais, L, Hall, N. Comorbidities in cerebral palsy and their relationship to neurologic subtype and GMFCS level. Neurology 2009; 72: 2090–6.Google Scholar
Novak, I. Evidence-based diagnosis, health care, and rehabilitation for children with cerebral palsy. J Child Neurol 2014; 29: 1141–56.Google Scholar
MacLennan, AH, Thompson, SC, Gecz, J. Cerebral palsy: causes, pathways, and the role of genetic variants. Am J Obstet Gynecol 2015; 213(6): 779–88.Google Scholar
Ellenberg, JH, Nelson, KB. The association of cerebral palsy with birth asphyxia: a definitional quagmire. Dev Med Child Neurol 2013; 55: 210–6.Google Scholar
Rosenbaum, PL, Palisano, RJ, Bartlett, DJ, et al. Development of the gross motor function classification system for cerebral palsy. Dev Med Child Neurol 2008; 50: 249–53.Google Scholar
Palisano, R, Rosenbaum, P, Walter, S, et al. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Dev Med Child Neurol 1997; 39: 214–23.Google Scholar
Palisano, RJ, Rosenbaum, P, Bartlett, D, Livingston, MH. Content validity of the expanded and revised Gross Motor Function Classification System. Dev Med Child Neurol 2008; 50: 744–50.Google Scholar
Gross Motor Function Classification System, 2015. Available at https://canchild.ca/system/tenon/assets/attachments/000/000/058/original/GMFCS-ER_English.pdf (accessed May 27, 2015).Google Scholar
Manual Abilities Classification System, 2015. Available at www.macs.nu/files/Mini-MACS_English_2016.pdf (accessed May 27, 2015).Google Scholar
Ohrvall, AM, Krumlinde-Sundholm, L, Eliasson, AC. The stability of the Manual Ability Classification System over time. Dev Med Child Neurol 2014; 56: 185–9.Google Scholar
Hidecker, MJ, Paneth, N, Rosenbaum, PL, et al. Developing and validating the Communication Function Classification System for individuals with cerebral palsy. Dev Med Child Neurol 2011; 53: 704–10.Google Scholar
Oskoui, M, Coutinho, F, Dykeman, J, et al. An update on the prevalence of cerebral palsy: a systematic review and meta-analysis. Dev Med Child Neurol 2013; 55: 509–19.Google Scholar
Yeargin-Allsopp, M, Van Naarden Braun, K, Doernberg, NS, et al. Prevalence of cerebral palsy in 8-year-old children in three areas of the United States in 2002: a multisite collaboration. Pediatrics 2008; 121: 547–54.Google Scholar
Durkin, MS, Maenner, MJ, Benedict, RE, et al. The role of socio-economic status and perinatal factors in racial disparities in the risk of cerebral palsy. Dev Med Child Neurol 2015; 57(9): 835–32.Google Scholar
Benfer, KA, Jordan, R, Bandaranayake, S, et al. Motor severity in children with cerebral palsy studied in a high-resource and low-resource country. Pediatrics 2014; 134:e1594–602.Google Scholar
McIntyre, S, Taitz, D, Keogh, J, et al. A systematic review of risk factors for cerebral palsy in children born at term in developed countries. Dev Med Child Neurol 2013; 55:499508.Google Scholar
Van Naarden Braun, K, Christensen, D, Doernberg, N, et al. Trends in the prevalence of autism spectrum disorder, cerebral palsy, hearing loss, intellectual disability, and vision impairment, metropolitan atlanta, 1991–2010. PLoS One 2015; 10:e0124120.Google Scholar
Aisen, ML, Kerkovich, D, Mast, J, et al. Cerebral palsy: clinical care and neurological rehabilitation. Lancet Neurol 2011; 10: 844–52.Google Scholar
Jacobs, SE, Berg, M, Hunt, R, et al. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2013; 1:CD003311.Google Scholar
Jacquemyn, Y, Zecic, A, Van Laere, D, Roelens, K. The use of intravenous magnesium in non-preeclamptic pregnant women: fetal/neonatal neuroprotection. Arch Gynecol Obstet 2015; 291: 969–75.Google Scholar
Sotiriadis, A, Tsiami, A, Papatheodorou, S, et al. Neurodevelopmental outcome after a single course of antenatal steroids in children born preterm: a systematic review and meta-analysis. Obstet Gynecol 2015; 125: 1385–96.Google Scholar
Ashwal, S, Russman, BS, Blasco, PA, et al. Practice parameter: diagnostic assessment of the child with cerebral palsy: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 2004; 62: 851–63.Google Scholar
Bosanquet, M, Copeland, L, Ware, R, Boyd, R. A systematic review of tests to predict cerebral palsy in young children. Dev Med Child Neurol 2013; 55: 418–26.Google Scholar
Reid, SM, Dagia, CD, Ditchfield, MR, et al. Population-based studies of brain imaging patterns in cerebral palsy. Dev Med Child Neurol 2014; 56: 222–32.Google Scholar
Yin Foo, R, Guppy, M, Johnston, LM. Intelligence assessments for children with cerebral palsy: a systematic review. Dev Med Child Neurol 2013; 55: 911–8.Google Scholar
Novak, I, Hines, M, Goldsmith, S, Barclay, R. Clinical prognostic messages from a systematic review on cerebral palsy. Pediatrics 2012; 130:e1285–312.Google Scholar
Odding, E, Roebroeck, ME, Stam, HJ. The epidemiology of cerebral palsy: incidence, impairments and risk factors. Disabil Rehabil 2006; 28: 183–91.Google Scholar
Brossard-Racine, M, Hall, N, Majnemer, A, et al. Behavioural problems in school age children with cerebral palsy. Eur J Paediatr Neurol 2012; 16:3541.Google Scholar
Parkes, J, White-Koning, M, Dickinson, HO, et al. Psychological problems in children with cerebral palsy: a cross-sectional European study. J Child Psychol Psychiatry 2008; 49: 405–13.Google Scholar
Fazzi, E, Signorini, SG, La Piana, R, et al. Neuro-ophthalmological disorders in cerebral palsy: ophthalmological, oculomotor, and visual aspects. Dev Med Child Neurol 2012; 54: 730–6.Google Scholar
Dufresne, D, Dagenais, L, Shevell, MI. Spectrum of visual disorders in a population-based cerebral palsy cohort. Pediatr Neurol 2014; 50: 324–8.Google Scholar
Dufresne, D, Dagenais, L, Shevell, MI. Epidemiology of severe hearing impairment in a population-based cerebral palsy cohort. Pediatr Neurol 2014; 51: 641–4.Google Scholar
Cockerill, H, Elbourne, D, Allen, E, et al. Speech, communication and use of augmentative communication in young people with cerebral palsy: the SH&PE population study. Child Care Health Dev 2014; 40: 149–57.Google Scholar
Andrew, MJ, Parr, JR, Sullivan, PB. Feeding difficulties in children with cerebral palsy. Arch Dis Child Educ Pract Ed 2012; 97: 222–9.Google Scholar
Parkinson, KN, Dickinson, HO, Arnaud, C, et al. Pain in young people aged 13 to 17 years with cerebral palsy: cross-sectional, multicentre European study. Arch Dis Child 2013; 98: 434–40.Google Scholar
Parkinson, KN, Gibson, L, Dickinson, HO, Colver, AF. Pain in children with cerebral palsy: a cross-sectional multicentre European study. Acta Paediatr 2010; 99: 446–51.Google Scholar
Simard-Tremblay, E, Constantin, E, Gruber, R, et al. Sleep in children with cerebral palsy: a review. J Child Neurol 2011; 26: 1303–10.Google Scholar
Romeo, DM, Brogna, C, Musto, E, et al. Sleep disturbances in preschool age children with cerebral palsy: a questionnaire study. Sleep Med 2014; 15: 1089–93.Google Scholar
Bjornson, KF, Belza, B, Kartin, D, et al. Self-reported health status and quality of life in youth with cerebral palsy and typically developing youth. Arch Phys Med Rehabil 2008; 89: 121–7.Google Scholar
Colver, A, Rapp, M, Eisemann, N, et al. Self-reported quality of life of adolescents with cerebral palsy: a cross-sectional and longitudinal analysis. Lancet 2015; 385: 705–16.Google Scholar
Nieuwenhuijsen, C, Donkervoort, M, Nieuwstraten, W, et al. Experienced problems of young adults with cerebral palsy: targets for rehabilitation care. Arch Phys Med Rehabil 2009; 90: 1891–7.Google Scholar
Verhoef, JA, Bramsen, I, Miedema, HS, et al. Development of work participation in young adults with cerebral palsy: a longitudinal study. J Rehabil Med 2014; 46: 648–55.Google Scholar
Wiegerink, DJ, Roebroeck, ME, van der Slot, WM, et al. Importance of peers and dating in the development of romantic relationships and sexual activity of young adults with cerebral palsy. Dev Med Child Neurol 2010; 52: 576–82.Google Scholar
Moster, D, Lie, RT, Markestad, T. Long-term medical and social consequences of preterm birth. N Engl J Med 2008; 359: 262–73.Google Scholar
Nelson, KB, Grether, JK. Can magnesium sulfate reduce the risk of cerebral palsy in very low birthweight infants? Pediatrics 1995; 95: 263–9.Google Scholar
Magnesium sulfate use in obstetrics (Committee Opinion No. 573). Obstet Gynecol 2013; 122: 727–8.Google Scholar
Fauchere, JC, Koller, BM, Tschopp, A, et al. Safety of early high-dose recombinant erythropoietin for neuroprotection in very preterm infants. J Pediatr 2015; 167(1): 52–7.e13.Google Scholar
Marks, JD, Schreiber, MD. Inhaled nitric oxide and neuroprotection in preterm infants. Clin Perinatol 2008; 35:793807.Google Scholar
Schmidt, B, Anderson, PJ, Doyle, LW, et al. Survival without disability to age 5 years after neonatal caffeine therapy for apnea of prematurity. JAMA 2012; 307: 275–82.Google Scholar
Novak, I, McIntyre, S, Morgan, C, et al. A systematic review of interventions for children with cerebral palsy: state of the evidence. Dev Med Child Neurol 2013; 55:885910.Google Scholar
Huang, HH, Fetters, L, Hale, J, McBride, A. Bound for success: a systematic review of constraint-induced movement therapy in children with cerebral palsy supports improved arm and hand use. Phys Ther 2009; 89: 1126–41.Google Scholar
Delgado, MR, Hirtz, D, Aisen, M, et al. Practice parameter: pharmacologic treatment of spasticity in children and adolescents with cerebral palsy (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology 2010; 74: 336–43.Google Scholar
Robb, JE, Hagglund, G. Hip surveillance and management of the displaced hip in cerebral palsy. J Child Orthop 2013; 7: 407–13.Google Scholar
Min, K, Song, J, Kang, JY, et al. Umbilical cord blood therapy potentiated with erythropoietin for children with cerebral palsy: a double-blind, randomized, placebo-controlled trial. Stem Cells 2013; 31: 581–91.Google Scholar
Ruff, CA, Faulkner, SD, Fehlings, MG. The potential for stem cell therapies to have an impact on cerebral palsy: opportunities and limitations. Dev Med Child Neurol 2013; 55: 689–97.Google Scholar
Baird, G, McConachie, H, Scrutton, D. Parents’ perceptions of disclosure of the diagnosis of cerebral palsy. Arch Dis Child 2000; 83: 475–80.Google Scholar
Orioles, A, Miller, VA, Kersun, LS, et al. “To be a phenomenal doctor you have to be the whole package”: physicians’ interpersonal behaviors during difficult conversations in pediatrics. J Palliat Med 2013; 16: 929–33.Google Scholar
Shevell, AH, Shevell, M. Doing the “talk”: disclosure of a diagnosis of cerebral palsy. J Child Neurol 2013; 28: 230–5.Google Scholar
Feldman, HM. Redesigning Health Care for Children with Disabilities: Strengthening Inclusion, Contribution, and Health. Baltimore: Brookes Publishing, 2013.Google Scholar

References

Volpe, JJ. Brain injury in premature infants: a complex amalgam of destructive and developmental disturbances. Lancet Neurol 2009; 8(1): 110–24. PubMed PMID: 19081519.Google Scholar
Aylward, GP. Neurodevelopmental outcomes of infants born prematurely. J Dev Behav Pediatr 2014; 35(6): 394407. PubMed PMID: 25007063.Google Scholar
Saigal, S, Tyson, J. Measurement of quality of life of survivors of neonatal intensive care: critique and implications. Semin Perinatol 2008; 32(1): 5966. PubMed PMID: 18249241.Google Scholar
Bhutta, AT, Cleves, MA, Casey, PH, et al. Cognitive and behavioral outcomes of school-aged children who were born preterm: a meta-analysis. JAMA 2002; 288(6): 728–37. PubMed PMID: 12169077.Google Scholar
Kerr-Wilson, CO, Mackay, DF, Smith, GCS, Pell, JP. Meta-analysis of the association between preterm delivery and intelligence. J Public Health 2012; 34(2): 209–16.Google Scholar
Luu, TM, Ment, LR, Schneider, KC, et al. Lasting effects of preterm birth and neonatal brain hemorrhage at 12 years of age. Pediatrics 2009; 123(3): 1037–44.Google Scholar
Miyake, A, Friedman, NP, Emerson, MJ, et al. The unity and diversity of executive functions and their contributions to complex “frontal lobe” tasks: a latent variable analysis. Cognitive Psychol 2000; 41(1): 49100.Google Scholar
Anderson, PJ, Reidy, N. Assessing executive function in preschoolers. Neuropsychol Rev 2012; 22(4): 345–60. PubMed PMID: 2012-32535-004.Google Scholar
Loe, IM, Feldman, HM, Huffman, LC. Executive function mediates effects of gestational age on functional outcomes and behavior in preschoolers. J Dev Behav Pediatr 2014; 35(5): 323–33. PubMed PMID: 24906034. Pubmed Central PMCID: NIHMS642002 (available on 06/01/15) and PMC4247205 (available on 06/01/15).Google Scholar
Loe, IM, Lee, ES, Luna, B, Feldman, HM. Executive function skills are associated with reading and parent-rated child function in children born prematurely. Early Hum Dev 2012; 88(2): 111–8.Google Scholar
Mulder, H, Pitchford, NJ, Hagger, MS, Marlow, N. Development of executive function and attention in preterm children: a systematic review. Dev Neuropsychol 2009; 34(4): 393421.Google Scholar
Aarnoudse-Moens, CS, Weisglas-Kuperus, N, van Goudoever, JB, Oosterlaan, J. Meta-analysis of neurobehavioral outcomes in very preterm and/or very low birth weight children. Pediatrics 2009; 124(2): 717–28. PubMed PMID: 19651588.Google Scholar
Anderson, PJ. Neuropsychological outcomes of children born very preterm. Semin Fetal Neonatal Med 2014; 19(2): 90–6. PubMed PMID: 24361279.Google Scholar
van de Weijer-Bergsma, E, Wijnroks, L, Jongmans, MJ. Attention development in infants and preschool children born preterm: a review. Infant Behav Dev 2008; 31(3): 333–51. PubMed PMID: 18294695.Google Scholar
Anderson, PJ, De Luca, CR, Hutchinson, E, et al. Attention problems in a representative sample of extremely preterm/extremely low birth weight children. Dev Neuropsychol 2011; 36(1): 5773. PubMed PMID: 21253991.Google Scholar
Wilson-Ching, M, Molloy, CS, Anderson, VA, et al. Attention difficulties in a contemporary geographic cohort of adolescents born extremely preterm/extremely low birth weight. J Int Neuropsychol Soc 2013; 19(10): 1097–108. PubMed PMID: 24050646.Google Scholar
Kail, R, Salthouse, TA. Processing speed as a mental capacity. Acta Psychol 1994; 86(2): 199225.Google Scholar
Rose, SA, Feldman, JF. Memory and speed: their role in the relation of infant information processing to later IQ. Child Dev 1997; 68(4): 630–41.Google Scholar
Mulder, H, Pitchford, NJ, Marlow, N. Processing speed and working memory underlie academic attainment in very preterm children. Arch Dis Child Fetal Neonatal Ed 2010; 95(4): F267–72.Google Scholar
Eilers, RE, Oller, D, Levine, S, Basinger, D, et al. The role of prematurity and socioeconomic status in the onset of canonical babbling in infants. Infant Behav Dev 1993; 16(3): 297315. PubMed PMID: Peer Reviewed Journal: 1994–05051–001.Google Scholar
Reilly, S, Eadie, P, Bavin, EL, et al. Growth of infant communication between 8 and 12 months: a population study. J Paediatr Child Health 2006; 42(12): 764–70. PubMed PMID: 17096710.Google Scholar
Stolt, S, Klippi, A, Launonen, K, et al. Size and composition of the lexicon in prematurely born very-low-birth-weight and full-term Finnish children at two years of age. J Child Language 2007; 34(2): 283310.Google Scholar
Menyuk, P, Liebergott, J, Schultz, M. Early Language Development in Full-Term and Premature Infants. Hillsdale, NJ: Erlbaum Associates, 1995.Google Scholar
Aarnoudse-Moens, CS, Oosterlaan, J, Duivenvoorden, HJ, et al. Development of preschool and academic skills in children born very preterm. J Pediatr 2011; 158(1): 51–6. PubMed PMID: 20708749.Google Scholar
Barre, N, Morgan, A, Doyle, LW, Anderson, PJ. Language abilities in children who were very preterm and/or very low birth weight: a meta-analysis. J Pediatr 2011; 158(5): 766–74.Google Scholar
van Noort-van der Spek, IL, Franken, M-CJP, Weisglas-Kuperus, N. Language functions in preterm-born children: a systematic review and meta-analysis. Pediatrics 2012; 129(4): 745–54.Google Scholar
Lee, ES, Yeatman, JD, Luna, B, Feldman, HM. Specific language and reading skills in school-aged children and adolescents are associated with prematurity after controlling for IQ. Neuropsychologia 2011; 49(5): 906–13.Google Scholar
Northam, GB, Liegeois, F, Chong, WK, et al. Speech and oromotor outcome in adolescents born preterm: relationship to motor tract integrity. J Pediatr 2012; 160(3): 402–8. PubMed PMID: 22000302. Pubmed Central PMCID: PMC3657185.Google Scholar
Lewis, BA, Singer, LT, Fulton, S, et al. Speech and language outcomes of children with bronchopulmonary dysplasia. J Commun Disord 2002; 35(5): 393406.Google Scholar
Wolke, D, Meyer, R. Cognitive status, language attainment, and prereading skills of 6-year-old very preterm children and their peers: the Bavarian Longitudinal Study. Dev Med Child Neurol 1999; 41(02): 94109.Google Scholar
McMahon, E, Wintermark, P, Lahav, A. Auditory brain development in premature infants: the importance of early experience. Ann NY Acad Sci 2012; 1252(1): 1724.Google Scholar
Rand, K, Lahav, A. Impact of the NICU environment on language deprivation in preterm infants. Acta Paediatr 2014; 103(3): 243–8.Google Scholar
Caskey, M, Stephens, B, Tucker, R, Vohr, B. Importance of parent talk on the development of preterm infant vocalizations. Pediatrics 2011; 128(5): 910–6.Google Scholar
Simms, V, Gilmore, C, Cragg, L, et al. Nature and origin of mathematics difficulties in very preterm children: a different etiology than developmental dyscalculia. Pediatr Res 2015; 77(2): 389–95.Google Scholar
Anderson, P, Doyle, LW, Callanan, C, et al. Neurobehavioral outcomes of school-age children born extremely low birth weight or very preterm in the 1990s. JAMA 2003; 289(24): 3264–72. PubMed PMID: Peer Reviewed Journal: 2003–06381–001.Google Scholar
Johnson, S, Hennessy, E, Smith, R, et al. Academic attainment and special educational needs in extremely preterm children at 11 years of age: the EPICure study. Arch Dis Child Fetal Neonatal Ed 2009; 94(4): F283–9. PubMed PMID: 19282336.Google Scholar
Taylor, HG, Espy, KA, Anderson, PJ. Mathematics deficiencies in children with very low birth weight or very preterm birth. Dev Disabil Res Rev 2009; 15(1): 52–9. PubMed PMID: 19213016.Google Scholar
Litt, J, Taylor, HG, Klein, N, Hack, M. Learning disabilities in children with very low birthweight: prevalence, neuropsychological correlates, and educational interventions. J Learning Disabil 2005; 38(2): 130–41. PubMed PMID: 15813595.Google Scholar
Simms, V, Cragg, L, Gilmore, C, et al. Mathematics difficulties in children born very preterm: current research and future directions. Arch Dis Child Fetal Neonatal Ed 2013; 98(5): F457–63. PubMed PMID: 23759519.Google Scholar
Guarini, A, Sansavini, A, Giovanelli, G, et al. Basic numberical processes in preterms. World J Pediatr 2006; 2(2): 102–8.Google Scholar
Guarini, A, Sansavini, A, Fabbri, M, et al. Basic numerical processes in very preterm children: a critical transition from preschool to school age. Early Hum Dev 2014; 90(3): 103–11. PubMed PMID: 24331582.Google Scholar
Stjernqvist, K, Svenningsen, N. Ten‐year follow‐up of children born before 29 gestational weeks: health, cognitive development, behaviour and school achievement. Acta Paediatr 1999; 88(5): 557–62.Google Scholar
Saigal, S, Ouden, Ld, Wolke, D, et al. School-age outcomes in children who were extremely low birth weight from four international population-based cohorts. Pediatrics 2003; 112(4): 943–50.Google Scholar
Hack, M, Flannery, DJ, Schluchter, M, et al. Outcomes in young adulthood for very-low-birth-weight infants [see comment]. N Engl J Med 2002; 346(3): 149–57. PubMed PMID: 11796848.Google Scholar
Spittle, AJ, Treyvaud, K, Doyle, LW, et al. Early emergence of behavior and social-emotional problems in very preterm infants. J Am Acad Child Adolesc Psychiatry 2009; 48(9): 909–18. PubMed PMID: 19633579.Google Scholar
Arpi, E, Ferrari, F. Preterm birth and behaviour problems in infants and preschool-age children: a review of the recent literature. Dev Med Child Neurol 2013; 55(9): 788–96. PubMed PMID: 23521214.Google Scholar
Johnson, S, Marlow, N. Growing up after extremely preterm birth: lifespan mental health outcomes. Semin Fetal Neonatal Med 2014; 19(2): 97104.Google Scholar
Johnson, S, Marlow, N. Preterm birth and childhood psychiatric disorders. Pediatr Res 2011; 69(5 Part 2): 11R–8R.Google Scholar
Woodward, LJ, Moor, S, Hood, KM, et al. Very preterm children show impairments across multiple neurodevelopmental domains by age 4 years. Arch Dis Child Fetal Neonatal Ed 2009; 94(5): F339–44. PubMed PMID: 19307223.Google Scholar
Elgen, SK, Leversen, KT, Grundt, JH, et al. Mental health at 5 years among children born extremely preterm: a national population-based study. Eur Child Adolesc Psychiatry 2012; 21(10): 583–9. PubMed PMID: 22752364.Google Scholar
Scott, MN, Taylor, HG, Fristad, MA, et al. Behavior disorders in extremely preterm/extremely low birth weight children in kindergarten. J Dev Behav Pediatr 2012; 33(3): 202–13.Google Scholar
Burnett, A, Davey, CG, Wood, SJ, et al. Extremely preterm birth and adolescent mental health in a geographical cohort born in the 1990s. Psychol Med 2014; 44(7): 1533–44. PubMed PMID: 23981686.Google Scholar
Botting, N, Powls, A, Cooke, RW, et al. Attention deficit hyperactivity disorders and other psychiatric outcomes in very low birthweight children at 12 years. J Child Psychol Psychiatry 1997; 38(8): 931–41. PubMed PMID: 9413793.Google Scholar
Indredavik, MS, Vik, T, Heyerdahl, S, et al. Psychiatric symptoms and disorders in adolescents with low birth weight. Arch Dis Child Fetal Neonat Ed. 2004; 89(5): F445–50. PubMed PMID: 15321968.Google Scholar
Johnson, S, Hollis, C, Kochhar, P, et al. Psychiatric disorders in extremely preterm children: longitudinal finding at age 11 years in the EPICure study. J Am Acad Child Adolesc Psychiatry 2010; 49(5): 453–63. PubMed PMID: 2010–09510–007.Google Scholar
Saigal, S, Stoskopf, B, Streiner, D, et al. Transition of extremely low-birth-weight infants from adolescence to young adulthood: comparison with normal birth-weight controls [see comment]. JAMA 2006; 295(6): 667–75. PubMed PMID: 16467235.Google Scholar
Burnett, A, Anderson, P, Cheong, J, et al. Prevalence of psychiatric diagnoses in preterm and full-term children, adolescents and young adults: a meta-analysis. Psychol Med 2011; 41(12): 2463–74.Google Scholar
Loe, I, Lee, E, Luna, B, Feldman, H. Behavior problems of 9–16 year old preterm children: biological, sociodemograhic, and intellectual contributions. Early Hum Dev 2011; 87(4): 247–52.Google Scholar
Kuban, KC, O’Shea, TM, Allred, EN, et al. Positive screening on the Modified Checklist for Autism in Toddlers (M-CHAT) in extremely low gestational age newborns. J Pediatr 2009; 154(4): 535–40. PubMed PMID: 19185317. PubMed Central PMCID: NIHMS106672 PMC2693887.Google Scholar
Luyster, RJ, Kuban, KC, O’Shea, TM, et al. The Modified Checklist for Autism in Toddlers in extremely low gestational age newborns: individual items associated with motor, cognitive, vision and hearing limitations. Paediatr Perinat Epidemiol 2011; 25(4): 366–76. PubMed PMID: 21649679.Google Scholar
Moore, T, Johnson, S, Hennessy, E, Marlow, N. Screening for autism in extremely preterm infants: problems in interpretation. Dev Med Child Neurol 2012; 54(6): 514–20. PubMed PMID: 22458327.Google Scholar
Guy, A, Seaton, SE, Boyle, EM, Draper, ES, Field, DJ, Manktelow, BN, et al. Infants born late/moderately preterm are at increased risk for a positive autism screen at 2 years of age. Journal of Pediatrics. 2015 Feb; 166(2): 269–75.e3. PubMed PMID: 25477165. English.Google Scholar
Hack, M, Taylor, HG, Schluchter, M, Andreias, L, Drotar, D, Klein, N. Behavioral outcomes of extremely low birth weight children at age 8 years. Journal of Developmental & Behavioral Pediatrics. 2009 Apr; 30(2): 122–30. PubMed PMID: 19322106. Pubmed Central PMCID: NIHMS273114 PMC3074440. English.Google Scholar
Johnson, S, Hollis, C, Kochhar, P, Hennessy, E, Wolke, D, Marlow, N. Autism spectrum disorders in extremely preterm children. The Journal of Pediatrics. 2010 Apr; 156(4): 525–31. PubMed PMID: Peer-Reviewed Status-Unknown: 2010–07245-017. English.Google Scholar
Perlman, JM. Neurobehavioral deficits in premature graduates of intensive care: potential medical and neonatal environmental risk factors. Pediatrics 2001; 108(6): 1339–48.Google Scholar
Gano, D, Andersen, SK, Glass, HC, et al. Impaired cognitive performance in premature newborns with two or more surgeries prior to term-equivalent age. Pediatr Res 2015; 78(3): 323–9.Google Scholar
Brummelte, S, Grunau, RE, Chau, V, et al. Procedural pain and brain development in premature newborns. Ann Neurol 2012; 71(3): 385–96.Google Scholar
Goldstein, RF. Developmental care for premature infants: a state of mind. Pediatrics 2012; 129(5): e1322–3.Google Scholar
Parker, JD, Schoendorf, KC, Kiely, JL. Associations between measures of socioeconomic status and low birth weight, small for gestational age, and premature delivery in the United States. Ann Epidemiol 1994; 4(4): 271–8.Google Scholar
Joseph, K, Liston, RM, Dodds, L, et al. Socioeconomic status and perinatal outcomes in a setting with universal access to essential health care services. Can Med Assoc J 2007; 177(6): 583–90.Google Scholar
Aylward, GP. Neurodevelopmental outcomes of infants born prematurely. J Dev Behavl Pediatr 2005; 26(6): 427–40.Google Scholar
Mulder, H, Pitchford, NJ, Marlow, N. Processing speed mediates executive function difficulties in very preterm children in middle childhood. J Int Neuropsychol Soc 2011; 17(03): 445–54.Google Scholar
Rose, SA, Feldman, JF, Jankowski, JJ, Van Rossem, R. Pathways from prematurity and infant abilities to later cognition. Child Dev 2005; 76(6): 1172–84. PubMed PMID: 16274433.Google Scholar
Rose, SA, Feldman, JF, Jankowski, JJ. Processing speed in the 1st year of life: a longitudinal study of preterm and full-term infants. Dev Psychol 2002; 38(6): 895902. PubMed PMID: 12428702.Google Scholar
Marchman, V, Adams, K, Loi, E, et al. Early language processing efficiency predicts later receptive vocabulary outcomes in children born preterm. Child Neuropsychol 2015; 22(6): 649–65.Google Scholar
Back, SA, Miller, SP. Brain injury in premature neonates: a primary cerebral dysmaturation disorder? Ann Neurol 2014; 75(4): 469–86. PubMed PMID: 24615937.Google Scholar
Back, SA. Perinatal white matter injury: the changing spectrum of pathology and emerging insights into pathogenetic mechanisms. Ment Retard Dev Disabil Res Rev 2006; 12(2): 129–40. PubMed PMID: 16807910.Google Scholar
Pannek, K, Scheck, SM, Colditz, PB, et al. Magnetic resonance diffusion tractography of the preterm infant brain: a systematic review. Dev Med Child Neurol 2014; 56(2): 113–24. PubMed PMID: 24102176.Google Scholar
Eikenes, L, Lohaugen, GC, Brubakk, AM, et al. Young adults born preterm with very low birth weight demonstrate widespread white matter alterations on brain DTI. Neuroimaging 2011; 54(3): 1774–85. PubMed PMID: 20965255.Google Scholar
Allin, MPG, Kontis, D, Walshe, M, et al. White matter and cognition in adults who were born preterm. PLoS One 2011; 6(10): e24525.Google Scholar
Feldman, HM, Lee, ES, Loe, IM, et al. White matter microstructure on diffusion tensor imaging is associated with conventional MRI findings and cognitive function in adolescents born preterm. Dev Med Child Neurol 2012; 54(9): 775868.Google Scholar
Woodward, LJ, Anderson, PJ, Austin, NC, et al. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med 2006; 355(7): 685–94. PubMed PMID: 16914704.Google Scholar
van Kooij, BJ, van Pul, C, Benders, MJ, et al. Fiber tracking at term displays gender differences regarding cognitive and motor outcome at 2 years of age in preterm infants. Pediatr Res 2011; 70(6): 626–32. PubMed PMID: 21857376.Google Scholar
Foster-Cohen, SH, Friesen, MD, Champion, PR, Woodward, LJ. High prevalence/low severity language delay in preschool children born very preterm. J Dev BehavPediatr 2010; 31(8): 658–67. PubMed PMID: 20613625.Google Scholar
Groeschel, S, Tournier, J-D, Northam, GB, et al. Identification and interpretation of microstructural abnormalities in motor pathways in adolescents born preterm. NeuroImage 2014; 87: 209–19.Google Scholar
Northam, GB, Liégeois, F, Chong, WK, et al. Total brain white matter is a major determinant of IQ in adolescents born preterm. Ann Neurol 2011; 69(4): 702–11.Google Scholar
Skranes, J, Lohaugen, GC, Martinussen, M, et al. White matter abnormalities and executive function in children with very low birth weight. NeuroReport 2009; 20(3): 263–6. PubMed PMID: 19444947.Google Scholar
Feldman, HM, Lee, ES, Yeatman, JD, Yeom, KW. Language and reading skills in school-aged children and adolescents born preterm are associated with white matter properties on diffusion tensor imaging. Neuropsychologia 2012; 50(14): 3348–62. PubMed PMID: 23088817. Pubmed Central PMCID: NIHMS419970 PMC3631607.Google Scholar
Northam, GB, Liegeois, F, Tournier, JD, et al. Interhemispheric temporal lobe connectivity predicts language impairment in adolescents born preterm. Brain 2012; 135(Pt 12): 3781–98. PubMed PMID: 23144265. Pubmed Central PMCID: PMC4031625.Google Scholar
Mullen, KM, Vohr, BR, Katz, KH, et al. Preterm birth results in alterations in neural connectivity at age 16 years. Neuroimage 2011; 54(4): 2563–70. PubMed PMID: 21073965.Google Scholar
Starke, M, Kiechl-Kohlendorfer, U, Kucian, K, et al. Brain structure, number magnitude processing, and math proficiency in 6- to 7-year-old children born prematurely: a voxel-based morphometry study. NeuroReport. 2013; 24(8): 419–24. PubMed PMID: 23587788.Google Scholar
Loe, IM, Lee, ES, Feldman, HM. Attention and internalizing behaviors in relation to white matter in children born preterm. J Dev Behav Pediatr 2013; 34(3): 156.Google Scholar
Fischi-Gómez, E, Vasung, L, Meskaldji, D-E, et al. Structural brain connectivity in school-age preterm infants provides evidence for impaired networks relevant for higher order cognitive skills and social cognition. Cerebral Cortex 2015; 25(9): 2793–805.Google Scholar
Abernethy, LJ, Cooke, RW, Foulder-Hughes, L. Caudate and hippocampal volumes, intelligence, and motor impairment in 7-year-old children who were born preterm. Pediatr Res 2004; 55(5): 884–93.Google Scholar
Thompson, DK, Wood, SJ, Doyle, LW, et al. Neonate hippocampal volumes: Prematurity, perinatal predictors, and 2-year outcome. Ann Neurol 2008; 63(5): 642–51.Google Scholar
Kilbride, HW, Thorstad, K, Daily, DK. Preschool outcome of less than 801-gram preterm infants compared with full-term siblings. Pediatrics 2004; 113(4): 742–7.Google Scholar
Peterson, J, Taylor, HG, Minich, N, et al. Subnormal head circumference in very low birth weight children: neonatal correlates and school-age consequences. Early Hum Dev 2006; 82(5): 325–34. PubMed PMID: 16360293.Google Scholar
Allen, MC. Neurodevelopmental outcomes of preterm infants. Curr Opin Neurol 2008; 21(2): 123–8.Google Scholar
Nosarti, C, Al‐Asady, MHS, Frangou, S, et al. Adolescents who were born very preterm have decreased brain volumes. Brain 2002; 125(7): 1616–23.Google Scholar
Kesler, SR, Ment, LR, Vohr, B, et al. Volumetric analysis of regional cerebral development in preterm children. Pediatr Neurol 2004; 31(5): 318–25. PubMed PMID: 15519112.Google Scholar
Kesler, SR, Vohr, B, Schneider, KC, et al. Increased temporal lobe gyrification in preterm children. Neuropsychologia 2006; 44(3): 445–53. PubMed PMID: 15985272.Google Scholar
Limperopoulos, C, Bassan, H, Gauvreau, K, et al. Does cerebellar injury in premature infants contribute to the high prevalence of long-term cognitive, learning, and behavioral disability in survivors? Pediatrics 2007; 120(3): 584–93.Google Scholar
Wilson, SL, Cradock, MM. Review: accounting for prematurity in developmental assessment and the use of age-adjusted scores. J Pediatr Psychol 2004; 29(8): 641–9.Google Scholar
World Health Organization. International Classification of Functioning, Disability and Health. Geneva: WHO, 2001.Google Scholar

References

Volpe, J. Neurology of the Newborn, 4th edn. Philadelphia: Saunders, 2001.Google Scholar
Miller, SP, Latal, B, Clark, H, et al. Clinical signs predict 30-month neurodevelopmental outcome after neonatal encephalopathy. Am J Obstet Gynecol 2004; 190: 93–9.Google Scholar
Ferriero, DM. Neonatal brain injury. N Engl J Med 2004; 351: 1985–95.Google Scholar
Vannucci, RC, Perlman, JM. Interventions for perinatal hypoxic-ischemic encephalopathy. Pediatrics 1997; 100: 1004–14.Google Scholar
Finer, NN, Robertson, CM, Richards, RT, et al. Hypoxic-ischemic encephalopathy in term neonates: perinatal factors and outcome. J Pediatr 1981; 98: 112–17.Google Scholar
Nelson, KB, Lynch, JK. Stroke in newborn infants. Lancet Neurol 2004; 3: 150–8.Google Scholar
Edwards, AD, Brocklehurst, P, Gunn, AJ, 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.Google Scholar
Jacobs, SE, Berg, M, Hunt, R, et al. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2013; 1:CD003311.Google Scholar
Tagin, MA, Woolcott, CG, Vincer, MJ, et al. Hypothermia for neonatal hypoxic ischemic encephalopathy: an updated systematic review and meta-analysis. Arch Pediatr Adolesc Med 2012; 166: 558–66.Google Scholar
Juul, SE, Ferriero, DM. Pharmacologic neuroprotective strategies in neonatal brain injury. Clin Perinatol 2014; 41: 119–31.Google Scholar
Thoresen, M. Cooling after perinatal asphyxia. Semin Fetal Neonatal Med 2015; 20:65.Google Scholar
Dammann, O, Ferriero, D, Gressens, P. Neonatal encephalopathy or hypoxic-ischemic encephalopathy? Appropriate terminology matters. Pediatr Res 2011; 70:12.Google Scholar
Volpe, JJ. Neonatal encephalopathy: an inadequate term for hypoxic-ischemic encephalopathy. Ann Neurol 2012; 72: 156–66.Google Scholar
Badawi, N, Kurinczuk, JJ, Keogh, JM, et al. Intrapartum risk factors for newborn encephalopathy: the Western Australian case-control study. BMJ 1998; 317: 1554–8.Google Scholar
Cowan, F, Rutherford, M, Groenendaal, F, et al. Origin and timing of brain lesions in term infants with neonatal encephalopathy. Lancet 2003; 361: 736–42.Google Scholar
Gano, D1, Chau, V, Poskitt, KJ, Hill, A, Roland, E, Brant, R, Chalmers, M, Miller, SP. Evolution of pattern of injury and quantitative MRI on days 1 and 3 in term newborns with hypoxic-ischemic encephalopathy. Pediatr Res 2013 Jul; 74(1): 82–7. doi: 10.1038/pr.2013.69. Epub 2013 Apr 25.Google Scholar
Barkovich, AJ, Miller, SP, Bartha, A, et al. MR imaging, MR spectroscopy, and diffusion tensor imaging of sequential studies in neonates with encephalopathy. AJNR Am J Neuroradiol 2006; 27: 533–47.Google Scholar
McKinstry, RC, Miller, JH, Snyder, AZ, et al. A prospective, longitudinal diffusion tensor imaging study of brain injury in newborns. Neurology 2002; 59: 824–33.Google Scholar
American College of Obstetricians and Gynecologists’ Task Force on Neonatal Encephalopathy. Executive summary: Neonatal encephalopathy and neurologic outcome, second edition. Report of the American College of Obstetricians and Gynecologists’ Task Force on Neonatal Encephalopathy. Obstet Gynecol 2014; 123:896901.Google Scholar
Myers, RE. Four patterns of perinatal brain damage and their conditions of occurrence in primates. Adv Neurol 1975; 10: 223–34.Google Scholar
Myers, RE. Two patterns of perinatal brain damage and their conditions of occurrence. Am J Obstet Gynecol 1972; 112: 246–76.Google Scholar
Childs, AM, Cornette, L, Ramenghi, LA, et al. Magnetic resonance and cranial ultrasound characteristics of periventricular white matter abnormalities in newborn infants. Clin Radiol 2001; 56: 647–55.Google Scholar
Felderhoff-Mueser, U, Rutherford, MA, Squier, WV, et al. Relationship between MR imaging and histopathologic findings of the brain in extremely sick preterm infants. AJNR Am J Neuroradiol 1999; 20: 1349–57.Google Scholar
Hope, PL, Gould, SJ, Howard, S, et al. Precision of ultrasound diagnosis of pathologically verified lesions in the brains of very preterm infants. Dev Med Child Neurol 1988; 30: 457–71.Google Scholar
Schouman-Claeys, E, Henry-Feugeas, MC, Roset, F, et al. Periventricular leukomalacia: correlation between MR imaging and autopsy findings during the first 2 months of life. Radiology 1993; 189:5964.Google Scholar
Barkovich, AJ, Hajnal, BL, Vigneron, D, et al. Prediction of neuromotor outcome in perinatal asphyxia: evaluation of MR scoring systems. AJNR Am J Neuroradiol 1998; 19: 143–9.Google Scholar
McQuillen, PS, Ferriero, DM. Selective vulnerability in the developing central nervous system. Pediatr Neurol 2004; 30: 227–35.Google Scholar
Sie, LT, van der Knaap, MS, Oosting, J, et al. MR patterns of hypoxic-ischemic brain damage after prenatal, perinatal or postnatal asphyxia. Neuropediatrics 2000; 31: 128–36.Google Scholar
Miller, SP, Ramaswamy, V, Michelson, D, et al. Patterns of brain injury in term neonatal encephalopathy. J Pediatr 2005; 146: 453–60.Google Scholar
Gadian, DG, Aicardi, J, Watkins, KE, et al. Developmental amnesia associated with early hypoxic-ischaemic injury. Brain 2000; 123(Pt 3):499507.Google Scholar
Ramaswamy, V, Miller, SP, Barkovich, AJ, et al. Perinatal stroke in term infants with neonatal encephalopathy. Neurology 2004; 62: 2088–91.Google Scholar
Lee, J, Croen, LA, Backstrand, KH, et al. Maternal and infant characteristics associated with perinatal arterial stroke in the infant. JAMA 2005; 293: 723–9.Google Scholar
Perez, A, Ritter, S, Brotschi, B, et al. Long-term neurodevelopmental outcome with hypoxic-ischemic encephalopathy. J Pediatr 2013; 163: 454–9.Google Scholar
Steinman, KJ, Gorno-Tempini, ML, Glidden, DV, et al. Neonatal watershed brain injury on magnetic resonance imaging correlates with verbal IQ at 4 years. Pediatrics 2009; 123: 1025–30.Google Scholar
Miller, SP, Newton, N, Ferriero, DM, et al. Predictors of 30-month outcome after perinatal depression: role of proton MRS and socioeconomic factors. Pediatr Res 2002; 52: 71–7.Google Scholar
Gonzalez, FF, Miller, SP. Does perinatal asphyxia impair cognitive function without cerebral palsy? Arch Dis Child Fetal Neonatal Ed 2006; 91:F454–9.Google Scholar
Barnett, A, Mercuri, E, Rutherford, M, et al. Neurological and perceptual-motor outcome at 5–6 years of age in children with neonatal encephalopathy: relationship with neonatal brain MRI. Neuropediatrics 2002; 33: 242–8.Google Scholar
Dixon, G, Badawi, N, Kurinczuk, JJ, et al. Early developmental outcomes after newborn encephalopathy. Pediatrics 2002; 109:2633.Google Scholar
Marlow, N, Rose, AS, Rands, CE, Draper, ES. Neuropsychological and educational problems at school age associated with neonatal encephalopathy. Arch Dis Child Fetal Neonatal Ed 2005; 90:F380–7.Google Scholar
Moster, D, Lie, RT, Markestad, T. Joint association of Apgar scores and early neonatal symptoms with minor disabilities at school age. Arch Dis Child Fetal Neonatal Ed 2002; 86:F1621.Google Scholar
Robertson, CM, Finer, NN, Grace, MG. School performance of survivors of neonatal encephalopathy associated with birth asphyxia at term. J Pediatr 1989; 114: 753–60.Google Scholar
Goodwin, TM, Belai, I, Hernandez, P, et al. Asphyxial complications in the term newborn with severe umbilical acidemia. Am J Obstet Gynecol 1992; 167: 1506–12.Google Scholar
Nelson, KB, Ellenberg, JH. Apgar scores as predictors of chronic neurologic disability. Pediatrics 1981; 68:3644.Google Scholar
Sarnat, HB, Sarnat, MS. Neonatal encephalopathy following fetal distress: a clinical and electroencephalographic study. Arch Neurol 1976; 33:696705.Google Scholar
Robertson, CM, Finer, NN. Long-term follow-up of term neonates with perinatal asphyxia. Clin Perinatol 1993; 20:483500.Google Scholar
Gunn, AJ, Wyatt, JS, Whitelaw, A, et al. Therapeutic hypothermia changes the prognostic value of clinical evaluation of neonatal encephalopathy. J Pediatr 2008; 152: 55–8.Google Scholar
de Vries, LS, Jongmans, MJ. Long-term outcome after neonatal hypoxic-ischaemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 2010; 95:F220–4.Google Scholar
Diamond, A. Executive functions. Annu Rev Psychol 2013; 64: 135–68.Google Scholar
van Handel, M, Swaab, H, de Vries, LS, Jongmans, MJ. Long-term cognitive and behavioral consequences of neonatal encephalopathy following perinatal asphyxia: a review. Eur J Paediatr 2007; 166: 645–54.Google Scholar
Kjellmer, I, Beijer, E, Carlsson, G, et al. Follow-up into young adulthood after cardiopulmonary resuscitation in term and near-term newborn infants. I. Educational achievements and social adjustment. Acta Paediatr 2002; 91: 1212–17.Google Scholar
Lindstrom, K, Lagerroos, P, Gillberg, C, Fernell, E. Teenage outcome after being born at term with moderate neonatal encephalopathy. Pediatr Neurol 2006; 35: 268–74.Google Scholar
Viggedal, G, Lundalv, E, Carlsson, G, Kjellmer, I. Follow-up into young adulthood after cardiopulmonary resuscitation in term and near-term newborn infants. II. Neuropsychological consequences. Acta Paediatr 2002; 91: 1218–26.Google Scholar
van Handel, M, de Sonneville, L, de Vries, LS, et al. Specific memory impairment following neonatal encephalopathy in term-born children. Dev Neuropsychol 2012; 37:3050.Google Scholar
Dilenge, ME, Majnemer, A, Shevell, MI. Long-term developmental outcome of asphyxiated term neonates. J Child Neurol 2001; 16: 781–92.Google Scholar
Badawi, N, Felix, JF, Kurinczuk, JJ, et al. Cerebral palsy following term newborn encephalopathy: a population-based study. Dev Med Child Neurol 2005; 47: 293–8.Google Scholar
Volpe, JJ. Neonatal Neurology, 4th edn. Philadelphia: Saunders, 2001.Google Scholar
Handley-Derry, M, Low, JA, Burke, SO, et al. Intrapartum fetal asphyxia and the occurrence of minor deficits in 4- to 8-year-old children. Dev Med Child Neurol 1997; 39: 508–14.Google Scholar
van Kooij, B, van Handel, M, Uiterwaal, C, et al. Corpus callosum size in relation to motor performance in 9- to 10- year-old children with neonatal encephalopathy. Pediatr Res 2008; 63:16.Google Scholar
van Schie, PE, Schijns, J, Becher, JG, et al. Long-term motor and behavioral outcome after perinatal hypoxic-ischemic encephalopathy. Eur J Paediatr Neurol. 2015; 19: 354–9.Google Scholar
Van Hof-van Duin, J, Mohn, G. Visual defects in children after cerebral hypoxia. Behav Brain Res 1984; 14: 147–55.Google Scholar
Mercuri, E, Atkinson, J, Braddick, O, et al. Basal ganglia damage and impaired visual function in the newborn infant. Arch Dis Child Fetal Neonatal Ed 1997; 77:F111–14.Google Scholar
Robertson, C, Finer, N. Term infants with hypoxic-ischemic encephalopathy: outcome at 3.5 years. Dev Med Child Neurol 1985; 27: 473–84.Google Scholar
Shankaran, S, Woldt, E, Koepke, T, et al. Acute neonatal morbidity and long-term central nervous system sequelae of perinatal asphyxia in term infants. Early Hum Dev 1991; 25: 135–48.Google Scholar
Mercuri, E, Anker, S, Guzzetta, A, et al. Visual function at school age in children with neonatal encephalopathy and low Apgar scores. Arch Dis Child Fetal Neonatal Ed 2004; 89:F258–62.Google Scholar
D’Souza, SW, McCartney, E, Nolan, M, Taylor, IG. Hearing, speech, and language in survivors of severe perinatal asphyxia. Arch Dis Child 1981; 56: 245–52.Google Scholar
Roland, EH, Hill, A, Norman, MG, et al. Selective brainstem injury in an asphyxiated newborn. Ann Neurol 1988; 23:8992.Google Scholar
Brunquell, PJ, Glennon, CM, DiMario, FJ Jr, et al. Prediction of outcome based on clinical seizure type in newborn infants. J Pediatr 2002; 140: 707–12.Google Scholar
Clancy, RR, Legido, A. Postnatal epilepsy after EEG-confirmed neonatal seizures. Epilepsia 1991; 32:6976.Google Scholar
Hellstrom-Westas, L, Blennow, G, Lindroth, M, et al. Low risk of seizure recurrence after early withdrawal of antiepileptic treatment in the neonatal period. Arch Dis Child Fetal Neonatal Ed 1995; 72:F97101.Google Scholar
Toet, MC, Groenendaal, F, Osredkar, D, et al. Postneonatal epilepsy following amplitude-integrated EEG-detected neonatal seizures. Pediatr Neurol 2005; 32: 241–7.Google Scholar
Jung, DE, Ritacco, DG, Nordli, DR, et al. Early Anatomical Injury Patterns Predict Epilepsy in Head Cooled Neonates With Hypoxic-Ischemic Encephalopathy. Pediatr Neurol 2015; 53(2): 135–40.Google Scholar
Robertson, CM, Finer, NN. Educational readiness of survivors of neonatal encephalopathy associated with birth asphyxia at term. J Dev Behav Pediatr 1988; 9:298306.Google Scholar
Stuart, A, Otterblad Olausson, P, Kallen, K. Apgar scores at 5 minutes after birth in relation to school performance at 16 years of age. Obstet Gynecol 2011; 118: 201–8.Google Scholar
Koot, HM. The study of quality of life: concepts and methods. In: Koot, HM, Wallander, JL, eds., Quality of Life in Child and Adolescent Illness. Hove: Bunner-Routledge, 2001.Google Scholar
Azzopardi, DV, Strohm, B, Edwards, AD, et al. Moderate hypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med 2009; 361: 1349–58.Google Scholar
Thornberg, E, Thiringer, K, Odeback, A, Milsom, I. Birth asphyxia: incidence, clinical course and outcome in a Swedish population. Acta Paediatr 1995; 84: 927–32.Google Scholar
Roland, EH, Poskitt, K, Rodriguez, E, et al. Perinatal hypoxic-ischemic thalamic injury: clinical features and neuroimaging. Ann Neurol 1998; 44: 161–6.Google Scholar
Rutherford, MA, Pennock, JM, Counsell, SJ, et al. Abnormal magnetic resonance signal in the internal capsule predicts poor neurodevelopmental outcome in infants with hypoxic-ischemic encephalopathy. Pediatrics 1998; 102: 323–8.Google Scholar
Krageloh-Mann, I, Helber, A, Mader, I, et al. Bilateral lesions of thalamus and basal ganglia: origin and outcome. Dev Med Child Neurol 2002; 44: 477–84.Google Scholar
Le Strange, E, Saeed, N, Cowan, FM, et al. MR imaging quantification of cerebellar growth following hypoxic-ischemic injury to the neonatal brain. AJNR Am J Neuroradiol 2004; 25: 463–8.Google Scholar
Sargent, MA, Poskitt, KJ, Roland, EH, et al. Cerebellar vermian atrophy after neonatal hypoxic-ischemic encephalopathy. AJNR Am J Neuroradiol 2004; 25: 1008–15.Google Scholar
Bonifacio, SL, Saporta, A, Glass, HC, et al. Therapeutic hypothermia for neonatal encephalopathy results in improved microstructure and metabolism in the deep gray nuclei. AJNR Am J Neuroradiol 2012; 33: 2050–5.Google Scholar
Nagy, Z, Lindstrom, K, Westerberg, H, et al. Diffusion tensor imaging on teenagers, born at term with moderate hypoxic-ischemic encephalopathy. Pediatr Res 2005; 58: 936–40.Google Scholar
Rutherford, M, Counsell, S, Allsop, J, et al. Diffusion-weighted magnetic resonance imaging in term perinatal brain injury: a comparison with site of lesion and time from birth. Pediatrics 2004; 114: 1004–14.Google Scholar
Thayyil, S, Chandrasekaran, M, Taylor, A, et al. Cerebral magnetic resonance biomarkers in neonatal encephalopathy: a meta-analysis. Pediatrics 2010; 125:e382–95.Google Scholar
Alderliesten, T, de Vries, LS, Khalil, Y, et al. Therapeutic hypothermia modifies perinatal asphyxia-induced changes of the corpus callosum and outcome in neonates. PLoS One 2015; 10:e0123230.Google Scholar
Bonifacio, SL, deVries, LS, Groenendaal, F. Impact of hypothermia on predictors of poor outcome: how do we decide to redirect care? Semin Fetal Neonatal Med 2015; 20: 122–7.Google Scholar
Massaro, AN. MRI for neurodevelopmental prognostication in the high-risk term infant. Semin Perinatol 2015; 39: 159–67.Google Scholar
Merchant, N, Azzopardi, D. Early predictors of outcome in infants treated with hypothermia for hypoxic-ischaemic encephalopathy. Dev Med Child Neurol 2015; 57(Suppl 3):816.Google Scholar
Sabir, H, Cowan, FM. Prediction of outcome methods assessing short- and long-term outcome after therapeutic hypothermia. Semin Fetal Neonatal Med 2015; 20: 115–21.Google Scholar
Shankaran, S. Outcomes of hypoxic-ischemic encephalopathy in neonates treated with hypothermia. Clin Perinatol 2014; 41: 149–9.Google Scholar
Shankaran, S, Pappas, A, McDonald, SA, et al. Predictive value of an early amplitude integrated electroencephalogram and neurologic examination. Pediatrics 2011; 128:e112–20.Google Scholar
Simbruner, G, Mittal, RA, Rohlmann, F, Muche, R. Systemic hypothermia after neonatal encephalopathy: outcomes of neo.nEURO.network RCT. Pediatrics 2010; 126:e771–8.Google Scholar
Shankaran, S, Laptook, AR, Tyson, JE, et al. Evolution of encephalopathy during whole body hypothermia for neonatal hypoxic-ischemic encephalopathy. J Pediatr 2012; 160: 567–72.Google Scholar
Thoresen, M, Hellstrom-Westas, L, Liu, X, de Vries, LS. Effect of hypothermia on amplitude-integrated electroencephalogram in infants with asphyxia. Pediatrics 2010; 126:e131–9.Google Scholar
Cheong, JL, Coleman, L, Hunt, RW, et al. Prognostic utility of magnetic resonance imaging in neonatal hypoxic-ischemic encephalopathy: substudy of a randomized trial. Arch Pediatr Adolesc Med 2012; 166: 634–40.Google Scholar
Rutherford, M, Ramenghi, LA, Edwards, AD, et al. Assessment of brain tissue injury after moderate hypothermia in neonates with hypoxic-ischaemic encephalopathy: a nested substudy of a randomised controlled trial. Lancet Neurol 2010; 9:3945.Google Scholar

References

Placencia, FX, McCullough, LB. The history of ethical decision making in neonatal intensive care. J Intensive Care Med 2011; 26(6): 368–84.Google Scholar
Section 504 of the Rehabilitation Act of 1983, Pub. L. No. 93–112, 87 Stat. 355. 29 USC § 701. Available at www.usbr.gov/cro/pdfsplus/rehabact.pdf (accessed June 8, 2015).Google Scholar
Child Welfare Information Gateway. What is child abuse and neglect? Recognizing the signs and symptoms, 2015. Available at www.childwelfare.gov/pubpdfs/whatiscan.pdf (accessed July 8, 2015).Google Scholar
Elsasser, G. Reagan loses Baby Doe case. Chicago Tribune, June 1986. Available at http://articles.chicagotribune.com/1986–06-10/news/8602110374_1_handicapped-infant-rehabilitation-act (accessed July 8, 2015).Google Scholar
USC, Title 42 – The Public Health and Welfare, Chapter 67 – Child Abuse Prevention and Treatment and Adoption Reform. Available at www.gpo.gov/fdsys/pkg/USCODE-2010-title42/html/USCODE-2010-title42-chap67.htm (accessed July 8, 2015).Google Scholar
Kopelman, L. Rejecting the Baby Doe rules and defending a “negative” analysis of the best interests standard. J Med Philos 2005; 30(4): 331–52.Google Scholar
In re: Baby “K,” 16 F.3d 590 (4th Cir.), cert. denied, 115 S. Ct. 91, 1994.Google Scholar
Brown, K. In the matter of Baby K: the Fourth Circuit stretches EMTALA even further. Mercer Law Rev 1996; 47: 1173–79.Google Scholar
Texas Health and Safety Code, Chapter 166, Section 46, 1999. Available at www.statutes.legis.state.tx.us/Docs/HS/htm/HS.166.htm (accessed August 5, 2015).Google Scholar
Bassel, A. Order at the end of life: establishing a clear and fair mechanism for the resolution of futility disputes. Vanderbilt Law Rev Notes 2010; 63(2). Available at www.vanderbiltlawreview.org/2010/03/order-at-the-end-of-life-establishing-a-clear-and-fair-mechanism-for-the-resolution-of-futility-disputes/ (accessed August 5, 2015).Google Scholar
California Probate Code, Sections 4735 and 4736.Google Scholar
Chabot, S., H.R. 2175 – 107th Congress (2001–2): Born-Alive Infants Protection Act of 2002. Available at www.congress.gov/bill/107th-congress/house-bill/2175/text (accessed July 8, 2015).Google Scholar
Centers for Medicare and Medicaid Services. Memo 05–26: Interaction of the EMTALA and the Born-Alive Infants Protection Act of 2002, April 2005. Available at www.cms.gov/Medicare/Provider-Enrollment-and-Certification/SurveyCertificationGenInfo/Downloads/SCletter05-26.pdf.Google Scholar
American Academy of Pediatrics Neonatal Resuscitation Program Steering Committee. Resources: Born-Alive Infants Protection Act of 2001, Public Law No. 107–207, January 2003. Available at www2.aap.org/nrp/inst_resources-resources-bornalive_act.html (accessed August 5, 2015).Google Scholar
A definition of irreversible coma. Report of the Ad Hoc Committee of the Harvard Medical School to Examine the Definition of Brain Death. JAMA 1968; 205(6): 337–40.Google Scholar
Debolt, D. Jahi McMath: family breaks silence on brain-dead girl’s condition. San Jose Mercury News, October 2014. Available at www.mercurynews.com/ci_26659381/jahi-mcmath-family-breaks-silence-brain-dead-girls (accessed August 5, 2015).Google Scholar
Bernat, JL. Whither brain death? Am J Bioethics 2014; 14(8).Google Scholar
Truog, RD, Miller, FG. Changing the conversation about brain death. Am J Bioethics 2014; 14(8).Google Scholar
Magnus, D, Wilfond, B, Caplan, AL., Perspective: Accepting Brain Death. N Engl J Med 2014; 370(10): 891–4.Google Scholar
Singh, J, Fanaroff, J, Andrews, B, et al. Resuscitation in the “gray zone” of viability: determining physician preferences and predicting infant outcomes. Pediatrics 2007; 120(3): 519–26.Google Scholar
Di Pietro, NC, Whiteley, L, Mizgalewicz, A, Illes, J. Treatments for neurodevelopmental disorders: evidence, advocacy, and the Internet. J Autism Dev Disord 2013; 43(1): 122–33.Google Scholar
Vavasseur, C, Foran, A, Murphy, JFA. Consensus statements on the borderlands of neonatal viability: from uncertainty to grey areas. Ir Med J 2007; 100(8): 561–4.Google Scholar
Verhagen, AAE, Janvier, A, Leuthner, SR, et al. Categorizing neonatal deaths: a cross-cultural study in the United States, Canada, and The Netherlands. J Pediatr 2010; 156(1): 33–7.Google Scholar
Weiner, J, Sharma, J, Lantos, J, Kilbride, H. How infants die in the neonatal intensive care unit: trends from 1999 through 2008. Arch Pediatr Adolesc Med 2011; 165(7): 630–4.Google Scholar
Kuschel, CA, Kent, A. Improved neonatal survival and outcomes at borderline viability brings increasing ethical dilemmas. J Paediatr Child Health 2011; 47(9): 585–9.Google Scholar
Kelly, CE, Cheong, JLY, Gabra Fam, L, et al. Moderate and late preterm infants exhibit widespread brain white matter microstructure alterations at term-equivalent age relative to term-born controls. Brain Imaging Behav 2015; 10(1): 41–9.Google Scholar
Gigerenzer, G, Gaissmaier, W, Kurz-Milcke, E, et al. Helping doctors and patients make sense of health statistics. Psychol Sci Public Interest 2007; 8(2): 5396.Google Scholar
Singh, J, Fanaroff, J, Andrews, B, et al. Resuscitation in the “gray zone” of viability: determining physician preferences and predicting infant outcomes. Pediatrics 2007; 120(3): 519–26.Google Scholar
Tennant, PWG, Pearce, MS, Bythell, M, Rankin, J. 20-year survival of children born with congenital anomalies: a population-based study. Lancet 2010; 375(9715): 649–56.Google Scholar
Wang, Y, Hu, J, Druschel, CM, Kirby, RS. Twenty-five-year survival of children with birth defects in New York State: a population-based study. Birth Defects Res A Clin Mol Teratol 2011; 91(12): 9951003.Google Scholar
Wilkinson, DJC, Thiele, P, Watkins, A, De Crespigny, L. Fatally flawed? A review and ethical analysis of lethal congenital malformations. BJOG 2012; 119(11): 1302–8.Google Scholar
Di Pietro, NC, Whiteley, L, Mizgalewicz, A, Illes, J. Treatments for neurodevelopmental disorders: evidence, advocacy, and the Internet. J Autism Dev Disord 2013; 43(1): 122–33.Google Scholar
Akins, RS, Angkustsiri, K, Hansen, RL. Complementary and alternative medicine in autism: an evidence-based approach to negotiating safe and efficacious interventions with families. Neurother J Am Soc Exp Neurother 2010; 7(3): 307–19.Google Scholar
Bell, E, Wallace, T, Chouinard, I, et al. Responding to requests of families for unproven interventions in neurodevelopmental disorders: hyperbaric oxygen “treatment” and stem cell “therapy” in cerebral palsy. Dev Disabil Res Rev 2011; 17(1): 1926.Google Scholar
Janvier, A, Lantos, J, POST Investigators. Ethics and etiquette in neonatal intensive care. JAMA Pediatr 2014; 168(9): 857–8.Google Scholar
Ali, Z. Selection criteria in the NICU: who should get effective critical care? Indian J Med Ethics 2009; 6(2): 103–4.Google Scholar
Verhagen, AAE, Janvier, A, Leuthner, SR, et al. Categorizing neonatal deaths: a cross-cultural study in the United States, Canada, and the Netherlands. J Pediatr 2010; 156(1): 33–7.Google Scholar
Wilkinson, DJ, Fitzsimons, JJ, Dargaville, PA, et al. Death in the neonatal intensive care unit: changing patterns of end of life care over two decades. Arch Dis Child Fetal Neonatal Ed 2006; 91(4): F268–71.Google Scholar
Brecht, M, Wilkinson, DJC. The outcome of treatment limitation discussions in newborns with brain injury. Arch Dis Child Fetal Neonatal Ed 2015; 100(2): F155–60.Google Scholar
Yu, VYH. Is neonatal intensive care justified in all preterm infants? Croat Med J. 2005; 46(5): 744–50.Google Scholar
Swamy, R, Mohapatra, S, Bythell, M, Embleton, ND. Survival in infants live born at less than 24 weeks’ gestation: the hidden morbidity of non-survivors. Arch Dis Child Fetal Neonatal Ed 2010; 95(4): F293–4.Google Scholar
Char, D, Cho, M, Magnus, D. Whole genome sequencing in critically ill children. Lancet Respir Med 2015; 3(4): 264–6.Google Scholar
Richards, C, Crawley, L, Magnus, D. Use of neurodevelopmental delay in pediatric solid organ transplant listing decisions: inconsistencies in standards across major pediatric transplant centers. Pediatr Transplant 2009; 13(7): 843–50.Google Scholar
American Academy of Pediatrics Infant Bioethics Task Force and Consultants. Guidelines for infant bioethics committees. Pediatrics 1984; 74(2): 306–10.Google Scholar
McGee, G, Spanogle, J, Caplan, A, et al. Successes and failures of hospital ethics committees: a national survey of ethics committee chairs. Camb Q Healthc Ethics 2002; 11(1): 8793.Google Scholar
Molloy, J, Evans, M, Coughlin, K. Moral distress in the resuscitation of extremely premature infants. Nurs Ethics 2015; 22(1): 5263.Google Scholar
Hack, M, Horbar, JD, Malloy, MH, et al. Very low birth weight outcomes of the National Institute of Child Health and Human Development Neonatal Network. Pediatrics 1991; 87(5): 587–97.Google Scholar
What is Vermont Oxford Network? Available at https://public.vtoxford.org/about-us/ (accessed August 4, 2015))Google Scholar
Malkar, MB, Gardner, WP, Mandy, GT, et al. Respiratory severity score on day of life 30 is predictive of mortality and the length of mechanical ventilation in premature infants with protracted ventilation. Pediatr Pulmonol 2015; 50(4): 363–9.Google Scholar
Bassler, D, Stoll, BJ, Schmidt, B, et al. Using a count of neonatal morbidities to predict poor outcome in extremely low birth weight infants: added role of neonatal infection. Pediatrics 2009; 123(1): 313–18.Google Scholar
Hoffman, L, Bann, C, Higgins, R, et al. Developmental outcomes of extremely preterm infants born to adolescent mothers. Pediatrics 2015; 135(6): 1082–92.Google Scholar
Magnus, D. The SUPPORT controversy and the debate over research within the standard of care. Am J Bioeth 2013; 13(12): 12Google Scholar
Tin, W, Milligan, D, Pennefather, P, Hey, E. Pulse oximetry, severe retinopathy, and outcome at one year in babies of less than 28 weeks gestation. Arch Dis Child Fetal Neonatal Ed 2001; 84(2): F106–10.Google Scholar
Anderson, CG, Benitz, WE, Madan, A. Retinopathy of prematurity and pulse oximetry: a national survey of recent practices. J Perinatol 2004; 24(3): 164–8.Google Scholar
Magnus, D, Caplan, AL. Perspective: risk, consent, and SUPPORT. N Engl J Med 2013; 368(20): 1864–5.Google Scholar
Drazen, JM, et al. Support for SUPPORT. N Engl J Med 2015; 373: 1469–70.Google Scholar
Wilfond, B, Magnus, D, et al. Letter: The OHRP and SUPPORT. N Engl J Med 2013; 368: e36.Google Scholar
Wilfond, B, Magnus, D, et al. Letter: “The OHRP and SUPPORT.” N Engl J Med. 2013; 368: e36Google Scholar
Macklin, R, Shepherd, L, Dreger, A. The OHRP and SUPPORT: another view. N Engl J Med 2013; 369(2):e3.Google Scholar
Lantos, JD. Vindication for SUPPORT. N Engl J Med 2015; 373: 1393–5.Google Scholar
Magnus, D, Wilfond, B. Research on medical practices and the ethics of disclosure. Pediatrics 2015; 135(2): 208–10.Google Scholar
Cho, MK, Magnus, D, Constantine, M, et al. Attitudes toward risk and informed consent for research on medical practices: a cross-sectional survey. Ann Intern Med 2015; 162(10): 690–6.Google Scholar

References

California BAJI (Book of Approved Jury Instructions), 6.30.Google Scholar
Ibid., 6.05.Google Scholar
Ibid., 6.00.1, 6.02.Google Scholar
Ibid., 6.03.Google Scholar
Ibid., 3.76, 3.77.Google Scholar
Altshuler, G. Placental insights into neurodevelopmental and other childhood diseases. Semin Pediatr Neurol 1995; 2: 90–9.Google Scholar
Truman v. Thomas (1980) 27 Cal. 3d 285, California Supreme Court.Google Scholar
American College of Obstetricians and Gynecologists. Fetal heart rate patterns: monitoring, interpretation, and management (ACOG Technical Bulletin No. 207, July 1995 (replaces No. 132, September 1989). Int J Gynaecol Obstet 1995; 51: 6574.Google Scholar
American College of Obstetricians and Gynecologists. Intrapartum Fetal Heart Rate Monitoring (ACOG Technical Bulletin No. 132). Washington, DC: American College of Obstetricians and Gynecologists, 1989.Google Scholar
American College of Obstetricians and Gynecologists. The Intrauterine Device (ACOG Technical Bulletin No. 104). Washington, DC: American College of Obstetricians and Gynecologists, 1987.Google Scholar
American College of Obstetricians and Gynecologists. Fetal and Neonatal Neurologic Injury (ACOG Technical Bulletin No. 163). Washington, DC: American College of Obstetricians and Gynecologists, 1992.Google Scholar
American Academy of Pediatrics, American College of Obstetricians and Gynecologists. Relationship between perinatal factors and neurologic outcome. In Poland, RL, Freeman, RK, eds., Guidelines for Perinatal Care, 3rd edn. Elk Grove Village, IL: AAP, 1992: 221–4.Google Scholar
Nelson, KB. Is it HIE? And why that matters. Acta Paediatr 2007; 96: 1113–14.Google Scholar
California BAJI (Book of Approved Jury Instructions). 6.11 (revised), Duty of Disclosure.Google Scholar
Ibid., 6.28, Duty of Patient.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×