Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-75dct Total loading time: 0 Render date: 2024-06-12T05:05:02.391Z Has data issue: false hasContentIssue false

Section III - Erythrocyte Disorders

Published online by Cambridge University Press:  30 January 2021

Pedro A. de Alarcón
Affiliation:
University of Illinois College of Medicine
Eric J. Werner
Affiliation:
Children's Hospital of the King's Daughters
Robert D. Christensen
Affiliation:
University of Utah
Martha C. Sola-Visner
Affiliation:
Harvard University, Massachusetts
Get access

Summary

The newborn screening (NBS) program is a well-established comprehensive public health initiative with the main goal of identifying newborns affected by genetic disorders, for whom early interventions may prevent disease morbidity and mortality. The early-in-life screening for genetic conditions not only permits early institution of specific therapeutic measures for those affected, but also creates the opportunity for genetic counselling for carriers (e.g., parents). Several hematologic conditions have benefited from NBS, most notably hemoglobinopathies, particularly sickle cell disease (SCD), for which early diagnosis with preemptive penicillin initiation has substantively reduced pediatric mortality [1, 2]. The inclusion of severe combined immune deficiency (SCID) in the panel of screened genetic disorders has allowed for early referral to hematopoietic stem cell transplantation and the soon-to-be scaled up, gene therapy [3, 4].

Type
Chapter
Information
Neonatal Hematology
Pathogenesis, Diagnosis, and Management of Hematologic Problems
, pp. 93 - 200
Publisher: Cambridge University Press
Print publication year: 2021

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

Gaston, MH, Verter, JI, Woods, G, et al. Prophylaxis with oral penicillin in children with sickle cell anemia. A randomized trial. N Engl J Med 1986;314(25):1593–9.Google Scholar
Vichinsky, E, Hurst, D, Earles, A, Kleman, K, Lubin, B. Newborn screening for sickle cell disease: Effect on mortality. Pediatrics 1988;81(6):749–55.CrossRefGoogle ScholarPubMed
Cowan, MJ, Neven, B, Cavazanna-Calvo, M, Fischer, A, Puck, J. Hematopoietic stem cell transplantation for severe combined immunodeficiency diseases. Biol Blood Marrow Transplant 2008;14(1 Suppl 1):73–5.CrossRefGoogle ScholarPubMed
Myers, LA, Patel, DD, Puck, JM, Buckley, RH. Hematopoietic stem cell transplantation for severe combined immunodeficiency in the neonatal period leads to superior thymic output and improved survival. Blood 2002;99(3):872–8.Google Scholar
Guthrie, R, Susi, A. A simple phenylalanine method for detecting phenylketonuria in large populations of newborn infants. Pediatrics 1963;32:338–43.CrossRefGoogle ScholarPubMed
Levy, HL. Genetic screening. Adv Hum Genet 1973;4:1104.Google ScholarPubMed
Dussault, JH, Coulombe, P, Laberge, C, et al. Preliminary report on a mass screening program for neonatal hypothyroidism. J Pediatr 1975;86(5):670–4.CrossRefGoogle ScholarPubMed
Itano, HA, Pauling, L. A rapid diagnostic test for sickle cell anemia. Blood 1949;4(1):66–8.Google Scholar
Nalbandian, RM, Nichols, BM, Camp, FR, Jr., et al. Dithionite tube test – a rapid, inexpensive technique for the detection of hemoglobin S and non-S sickling hemoglobin. Clin Chem 1971;17(10):1028–32.Google Scholar
Nichols, BM, Nalbandian, RM, Henry, RL, Wolf, PL, Camp, FR, Jr. Murayama test for hemoglobin S: Simplification in technique. Clin Chem 1971;17(10):1059–60.Google Scholar
Hicksg, EJ, Griep, JA, Nordschow, CD. Comparison of results for three method of hemoglobin S identification. Clin Chem 1973;19(5):533–5.Google Scholar
Garrick, MD, Dembure, P, Guthrie, R. Sickle-cell anemia and other hemoglobinopathies: Procedures and strategy for screening employing spots of blood on filter paper as specimens. N Engl J Med 1973;288(24):1265–8.CrossRefGoogle ScholarPubMed
Kohn, J. Separation of haemoglobins on cellulose acetate. J Clin Pathol 1969;22(1):109–11.Google Scholar
Benson, JM, Therrell, BL, Jr. History and current status of newborn screening for hemoglobinopathies. Semin Perinatol 2010;34(2):134–44.Google Scholar
Rashed, MS, Ozand, PT, Bucknall, MP, Little, D. Diagnosis of inborn errors of metabolism from blood spots by acylcarnitines and amino acids profiling using automated electrospray tandem mass spectrometry. Pediatr Res 1995;38(3):324–31.Google Scholar
Moat, SJ, Rees, D, King, L, et al. Newborn blood spot screening for sickle cell disease by using tandem mass spectrometry: Implementation of a protocol to identify only the disease states of sickle cell disease. Clin Chem 2014;60(2):373–80.Google Scholar
Schmidt, RM, Holland, S. Standardization in abnormal hemoglobin detection. An evaluation of hemoglobin electrophoresis kits. Clin Chem 1974;20(5):591–4.Google Scholar
Schmidt, RM, Brosious, EM. Evaluation of proficiency in the performance of tests for abnormal hemoglobins. Am J Clin Pathol 1974;62(5):664–9.CrossRefGoogle ScholarPubMed
De Jesus, VR, Mei, JV, Bell, CJ, Hannon, WH. Improving and assuring newborn screening laboratory quality worldwide: 30-year experience at the Centers for Disease Control and Prevention. Semin Perinatol 2010;34(2):125–33.Google Scholar
De Jesus, VR, Mei, JV, Cordovado, SK, Cuthbert, CD. The Newborn Screening Quality Assurance Program at the Centers for Disease Control and Prevention: Thirty-five year experience assuring newborn screening laboratory quality. Int J Neonatal Screen 2015;1(1):1326.Google Scholar
Gaston, M, Rosse, WF. The cooperative study of sickle cell disease: Review of study design and objectives. Am J Pediatr Hematol Oncol 1982;4(2):197201.Google Scholar
Consensus conference. Newborn screening for sickle cell disease and other hemoglobinopathies. JAMA 1987;258(9):1205–9.Google Scholar
Therrell, BL, Jr., Lloyd-Puryear, MA, Eckman, JR, Mann, MY. Newborn screening for sickle cell diseases in the United States: A review of data spanning 2 decades. Semin Perinatol 2015;39(3):238–51.Google Scholar
Watson, MS, Mann, MY, Lloyd, -Puryear, MA, et al., American College of Medical Genetics Newborn Screening Expert Group. Newborn screening: Toward a uniform screening panel and system: Executive summary. Pediatrics 2006; 117 (5 Pt 2): S296307.Google Scholar
Koopmans, J, Hiraki, S, Ross, LF. Attitudes and beliefs of pediatricians and genetic counselors regarding testing and screening for CF and G6PD: Implications for policy. Am J Med Genet A 2006;140(21):2305–11.Google Scholar
Natowicz, M. Newborn screening: Setting evidence-based policy for protection. N Engl J Med 2005;353(9):867–70.CrossRefGoogle ScholarPubMed
Botkin, JR, Clayton, EW, Fost, NC, et al. Newborn screening technology: Proceed with caution. Pediatrics 2006;117(5):1793–9.Google Scholar
Heredero-Baute, L. Community-based program for the diagnosis and prevention of genetic disorders in Cuba: Twenty years of experience.Community Genet 2004;7(2–3):130–6.Google ScholarPubMed
Therrell, BL, Padilla, CD, Loeber, JG, et al. Current status of newborn screening worldwide: 2015.Semin Perinatol 2015;39(3):171–87.CrossRefGoogle ScholarPubMed
Fernandes, AP, Januario, JN, Cangussu, CB, Macedo, DL, Viana, MB. Mortality of children with sickle cell disease: A population study. J Pediatr 2010;86(4):279–84.Google Scholar
Sabarense, AP, Lima, GO, Silva, LM, Viana, MB. Survival of children with sickle cell disease in the comprehensive newborn screening programme in Minas Gerais, Brazil. Paediatr Int Child Health 2015;35(4):329–32.Google Scholar
Silva-Pinto, AC, Alencar de Queiroz, MC, Antoniazzo, Zamaro, PJ, Arruda, M, Pimentel, dos Santos, H. The neonatal screening program in Brazil, focus on sickle cell disease (SCD).Int J Neonatal Screen 2019;5(1):11.Google Scholar
Piel, FB, Hay, SI, Gupta, S, Weatherall, DJ, Williams, TN. Global burden of sickle cell anaemia in children under five, 2010–2050: Modelling based on demographics, excess mortality, and interventions. PLoS Med 2013;10(7):e1001484.CrossRefGoogle ScholarPubMed
Upadhye, DS, Jain, DL, Trivedi, YL, et al. Neonatal screening and the clinical outcome in children with sickle cell disease in central India. PLoS One 2016;11(1):e0147081.CrossRefGoogle ScholarPubMed
Dixit, S, Sahu, P, Kar, SK, Negi, S. Identification of the hot-spot areas for sickle cell disease using cord blood screening at a district hospital: An Indian perspective. J Community Genet 2015;6(4):383–7.Google Scholar
Upadhye, D, Das, RS, Ray, J, et al. Newborn screening for hemoglobinopathies and red cell enzymopathies in Tripura State: A malaria-endemic state in northeast India. Hemoglobin 2018;42(1):43–6.Google Scholar
Panigrahi, S, Patra, PK, Khodiar, PK. Neonatal screening of sickle cell anemia: A preliminary report. Indian J Pediatr 2012;79(6):747–50.Google Scholar
Howson, CP, Cedergren, B, Giugliani, R, et al. Universal newborn screening: A roadmap for action. Mol Genet Metab 2018;124(3):177–83.Google Scholar
McGann, PT, Ferris, MG, Ramamurthy, U, et al. A prospective newborn screening and treatment program for sickle cell anemia in Luanda, Angola. Am J Hematol 2013;88(12):984–9.Google Scholar
McGann, PT, Grosse, SD, Santos, B, et al. A cost-effectiveness analysis of a pilot neonatal screening program for sickle cell anemia in the Republic of Angola. J Pediatr 2015;167(6):1314–9.Google Scholar
Ndeezi, G, Kiyaga, C, Hernandez, AG, et al. Burden of sickle cell trait and disease in the Uganda Sickle Surveillance Study (US3): A cross-sectional study. Lancet Glob Health 2016;4(3):e195200.Google Scholar
McGann, PT, Hernandez, AG, Ware, RE. Sickle cell anemia in sub-Saharan Africa: Advancing the clinical paradigm through partnerships and research. Blood 2017;129(2):155–61.Google Scholar
Streetly, A, Sisodia, R, Dick, M, et al. Evaluation of newborn sickle cell screening programme in England: 2010–2016. Arch Dis Child 2018;103(7):648–53.Google Scholar
Holtzman, NA. Newborn screening for severe combined immunodeficiency: Progress and challenges. JAMA 2014;312(7):701–2.Google Scholar
Adams, SP, Rashid, S, Premachandra, T, et al. Screening of neonatal UK dried blood spots using a duplex TREC screening assay. J Clin Immunol 2014;34(3):323–30.CrossRefGoogle ScholarPubMed
Audrain, M, Thomas, C, Mirallie, S, et al. Evaluation of the T-cell receptor excision circle assay performances for severe combined immunodeficiency neonatal screening on Guthrie cards in a French single centre study. Clin Immunol 2014;150(2):137–9.Google Scholar
Somech, R, Lev, A, Simon, AJ, et al. Newborn screening for severe T and B cell immunodeficiency in Israel: A pilot study. Isr Med Assoc J 2013;15(8):404–9.Google Scholar
Chien, YH, Chiang, SC, Chang, KL, et al. Incidence of severe combined immunodeficiency through newborn screening in a Chinese population. J Formos Med Assoc 2015;114(1):12–6.CrossRefGoogle ScholarPubMed
Kwan, A, Church, JA, Cowan, MJ, et al. Newborn screening for severe combined immunodeficiency and T-cell lymphopenia in California: Results of the first 2 years. J Allergy Clin Immunol 2013;132(1):140–50.CrossRefGoogle ScholarPubMed
Hassell, KL. Population estimates of sickle cell disease in the US. Am J Prev Med 2010;38(4 Suppl):S512–21.CrossRefGoogle Scholar
Modell, B, Darlison, M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ 2008;86(6):480–7.Google Scholar
Weatherall, DJ. The inherited diseases of hemoglobin are an emerging global health burden. Blood 2010;115(22):4331–6.Google Scholar
Pasvol, G, Weatherall, DJ, Wilson, RJ. Cellular mechanism for the protective effect of haemoglobin S against P. falciparum malaria.Nature 1978;274(5672):701–3.Google Scholar
Allen, SJ, O’Donnell, A, Alexander, ND, et al. Alpha+–thalassemia protects children against disease caused by other infections as well as malaria. Proc Natl Acad Sci USA 1997;94(26):14736–41.Google Scholar
Taylor, SM, Parobek, CM, Fairhurst, RM. Haemoglobinopathies and the clinical epidemiology of malaria: A systematic review and meta-analysis. Lancet Infect Dis 2012;12(6):457–68.Google Scholar
Williams, TN, Mwangi, TW, Wambua, S, et al. Negative epistasis between the malaria-protective effects of alpha+–thalassemia and the sickle cell trait. Nat Genet 2005;37(11):1253–7.Google Scholar
Sundd, P, Gladwin, MT, Novelli, EM. Pathophysiology of sickle cell disease. Annu Rev Pathol 2019;14:263–92.Google Scholar
Serjeant, GR. Natural history and determinants of clinical severity of sickle cell disease. Curr Opin Hematol 1995;2(2):103–8.Google Scholar
Chui, DH, Waye, JS. Hydrops fetalis caused by alpha-thalassemia: An emerging health care problem. Blood 1998;91(7):2213–22.Google Scholar
Fucharoen, S, Viprakasit, V. Hb H disease: Clinical course and disease modifiers. Hematology Am Soc Hematol Educ Program 2009;2009(1):2634.Google Scholar
Lal, A, Goldrich, ML, Haines, DA, et al. Heterogeneity of hemoglobin H disease in childhood. N Engl J Med 2011;364(8):710–18.Google Scholar
Menzel, S, Garner, C, Rooks, H, Spector, TD, Thein, SL. HbA2 levels in normal adults are influenced by two distinct genetic mechanisms. Br J Haematol 2013;160(1):101–5.Google Scholar
Olivieri, NF. The beta-thalassemias. New Engl J Med 1999;341(2):99109.Google Scholar
Ho, PJ, Hall, GW, Luo, LY, Weatherall, DJ, Thein, SL. Beta-thalassaemia intermedia: Is it possible consistently to predict phenotype from genotype? Br J Haematol 1998;100(1):70–8.Google Scholar
Ho, PJ, Wickramasinghe, SN, Rees, DC, et al. Erythroblastic inclusions in dominantly inherited beta thalassemias. Blood 1997;89(1):322–8.Google Scholar
Efremov, DG, Efremov, GD, Zisovski, N, et al. Variation in clinical severity among patients with Hb Lepore-Boston-beta-thalassaemia is related to the type of beta-thalassaemia. Br J Haematol 1988;68(3):351–5.Google Scholar
Thein, SL. Genetic modifiers of beta-thalassemia. Haematologica 2005;90(5):649–60.Google Scholar
Higgs, DR, Clegg, JB, Weatherall, DJ, Serjeant, BE, Serjeant, GR. Interaction of the alpha alpha alpha globin gene haplotype and sickle haemoglobin. Br J Haematol 1984;57(4):671–8.Google Scholar
Cappellini, MD, Fiorelli, G. Glucose-6-phosphate dehydrogenase deficiency. Lancet 2008;371(9606):6474.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(22):12128–32.Google Scholar
Frank, JE. Diagnosis and management of G6PD deficiency. Am Fam Physician 2005;72(7):1277–82.Google Scholar
Kaplan, M, Hammerman, C. Glucose-6-phosphate dehydrogenase deficiency: A hidden risk for kernicterus. Semin Perinatol 2004;28(5):356–64.Google Scholar
Slusher, TM, Vreman, HJ, McLaren, DW, et al. Glucose-6-phosphate dehydrogenase deficiency and carboxyhemoglobin concentrations associated with bilirubin-related morbidity and death in Nigerian infants. J Pediatr 1995;126(1):102–8.Google Scholar
Al-Mousa, H, Al-Saud, B. Primary immunodeficiency diseases in highly consanguineous populations from Middle East and North Africa: Epidemiology, diagnosis, and care. Front Immunol 2017;8:678.Google Scholar
Griffith, LM, Cowan, MJ, Notarangelo, LD, et al. Improving cellular therapy for primary immune deficiency diseases: Recognition, diagnosis, and management. J Allergy Clin Immunol 2009;124(6):1152–60 e12.Google Scholar
Cossu, F. Genetics of SCID. Ital J Pediatr 2010;36:76.CrossRefGoogle ScholarPubMed
Shearer, WT, Dunn, E, Notarangelo, LD, et al. Establishing diagnostic criteria for severe combined immunodeficiency disease (SCID), leaky SCID, and Omenn syndrome: The Primary Immune Deficiency Treatment Consortium experience. J Allergy Clin Immunol 2014;133(4):1092–8.Google Scholar
Pai, SY, Logan, BR, Griffith, LM, et al. Transplantation outcomes for severe combined immunodeficiency, 2000–2009. N Engl J Med 2014;371(5):434–46.Google Scholar
Sarmiento, JD, Villada, F, Orrego, JC, Franco, JL, Trujillo-Vargas, CM. Adverse events following immunization in patients with primary immunodeficiencies. Vaccine 2016;34(13):1611–6.Google Scholar
Chong, SS, Boehm, CD, Higgs, DR, Cutting, GR. Single-tube multiplex-PCR screen for common deletional determinants of alpha-thalassemia. Blood 2000;95(1):360–2.Google Scholar
Harteveld, CL, Refaldi, C, Cassinerio, E, Cappellini, MD, Giordano, PC. Segmental duplications involving the alpha-globin gene cluster are causing beta-thalassemia intermedia phenotypes in beta-thalassemia heterozygous patients. Blood Cells Mol Dis 2008;40(3):312–16.Google Scholar
Old, JM. Screening and genetic diagnosis of haemoglobinopathies. Scand J Clin Lab Invest 2007;67(1):7186.CrossRefGoogle ScholarPubMed
Bond, M, Hunt, B, Flynn, B, et al. Towards a point-of-care strip test to diagnose sickle cell anemia. PLoS One 2017;12(5):e0177732.Google Scholar
Kumar, AA, Chunda-Liyoka, C, Hennek, JW, et al. Evaluation of a density-based rapid diagnostic test for sickle cell disease in a clinical setting in Zambia. PLoS One 2014;9(12):e114540.CrossRefGoogle Scholar
Yang, X, Kanter, J, Piety, NZ, et al. A simple, rapid, low-cost diagnostic test for sickle cell disease. Lab Chip 2013;13(8):1464–7.Google Scholar
Kanter, J, Telen, MJ, Hoppe, C, et al. Validation of a novel point of care testing device for sickle cell disease. BMC Med 2015;13:225.Google Scholar
McGann, PT, Schaefer, BA, Paniagua, M, Howard, TA, Ware, RE. Characteristics of a rapid, point-of-care lateral flow immunoassay for the diagnosis of sickle cell disease. Am J Hematol 2016;91(2):205–10.Google Scholar
Quinn, CT, Paniagua, MC, DiNello, RK, Panchal, A, Geisberg, M. A rapid, inexpensive and disposable point-of-care blood test for sickle cell disease using novel, highly specific monoclonal antibodies. Br J Haematol 2016;175(4):724–32.Google Scholar
Ung, R, Alapan, Y, Hasan, MN, et al. Point-of-care screening for sickle cell disease by a mobile micro-electrophoresis platform. Blood 2015;126(23).Google Scholar
Grosveld, F, Antoniou, M, Berry, M, et al. The regulation of human globin gene switching. Philos Trans R Soc Lond B Biol Sci 1993;339(1288):183–91.Google Scholar
Colombo, B, Kim, B, Atencio, RP, Molina, C, Terrenato, L. The pattern of fetal haemoglobin disappearance after birth. Br J Haematol 1976;32(1):7987.Google Scholar
Davis, LR. Changing blood picture in sickle-cell anaemia from shortly after birth to adolescence. J Clin Pathol 1976;29(10):898901.Google Scholar
Hustace, T, Fleisher, JM, Sanchez Varela, AM, Podda, A, Alvarez, O. Increased prevalence of false positive hemoglobinopathy newborn screening in premature infants. Pediatr Blood Cancer 2011;57(6):1039–43.Google Scholar
Hoppe, CC. Newborn screening for non-sickling hemoglobinopathies. Hematology Am Soc Hematol Educ Program 2009; 2009:1925.Google Scholar
Reed, W, Lane, PA, Lorey, F, et al. Sickle-cell disease not identified by newborn screening because of prior transfusion. J Pediatr 2000;136(2):248–50.Google Scholar
Yates, AM, Mortier, NA, Hyde, KS, Hankins, JS, Ware, RE. The diagnostic dilemma of congenital unstable hemoglobinopathies. Pediatr Blood Cancer 2010;55(7):1393–5.Google Scholar
Alauddin, H, Langa, M, Mohd Yusoff, M, et al. Detection of alpha-thalassaemia in neonates on cord blood and dried blood spot samples by capillary electrophoresis. Malays J Pathol 2017;39(1):1723.Google Scholar
Frempong, T, Pearson, HA. Newborn screening coupled with comprehensive follow-up reduced early mortality of sickle cell disease in Connecticut. Conn Med 2007;71(1):912.Google Scholar
Okpala, I, Thomas, V, Westerdale, N, et al. The comprehensiveness care of sickle cell disease. Eur J Haematol 2002;68(3):157–62.Google Scholar
Fogel, BN, Nguyen, HLT, Smink, G, Sekhar, DL. Variability in state-based recommendations for management of alpha thalassemia trait and silent carrier detected on the newborn screen. J Pediatr 2018;195:283–7.CrossRefGoogle ScholarPubMed
Michlitsch, J, Azimi, M, Hoppe, C, et al. Newborn screening for hemoglobinopathies in California. Pediatr Blood Cancer 2009;52(4):486–90.Google Scholar
Watchko, JF, Kaplan, M, Stark, AR, Stevenson, DK, Bhutani, VK. Should we screen newborns for glucose-6-phosphate dehydrogenase deficiency in the United States? J Perinatol 2013;33(7):499504.Google Scholar
WHO Working Group. Glucose-6-phosphate dehydrogenase deficiency. Bull World Health Organ 1989;67(6):601–11.Google Scholar
Nock, ML, Johnson, EM, Krugman, RR, et al. Implementation and analysis of a pilot in-hospital newborn screening program for glucose-6-phosphate dehydrogenase deficiency in the United States. J Perinatol 2011;31(2):112–17.Google Scholar
Watchko, JF. Hyperbilirubinemia in African American neonates: Clinical issues and current challenges. Semin Fetal Neonatal Med 2010;15(3):176–82.Google Scholar
Kaplan, M, Hammerman, C, Vreman, HJ, Stevenson, DK, Beutler, E. Acute hemolysis and severe neonatal hyperbilirubinemia in glucose-6-phosphate dehydrogenase-deficient heterozygotes. J Pediatr 2001;139(1):137–40.Google Scholar
Herschel, M, Ryan, M, Gelbart, T, Kaplan, M. Hemolysis and hyperbilirubinemia in an African American neonate heterozygous for glucose-6-phosphate dehydrogenase deficiency. J Perinatol 2002;22(7):577–9.Google Scholar
Beutler, E, Baluda, MC. The separation of glucose-6-phosphate-dehydrogenase-deficient erythrocytes from the blood of heterozygotes for glucose-6-phosphate-dehydrogenase deficiency. Lancet 1964;1(7326):189–92.Google Scholar
Beutler, E, Blume, KG, Kaplan, JC, et al. International Committee for Standardization in Haematology: Recommended screening test for glucose-6-phosphate dehydrogenase (G-6-PD) deficiency. Br J Haematol 1979;43(3):465–7.Google Scholar
Luzzatto, L. Glucose 6-phosphate dehydrogenase deficiency: From genotype to phenotype. Haematologica 2006;91(10):1303–6.Google Scholar
Beutler, E. G6PD deficiency. Blood 1994;84(11):3613–36.Google Scholar
Lin, Z, Fontaine, JM, Freer, DE, Naylor, EW. Alternative DNA-based newborn screening for glucose-6-phosphate dehydrogenase deficiency. Mol Genet Metab 2005;86(1–2):212–19.Google Scholar
Kaplan, M, Hammerman, C. Neonatal screening for glucose-6-phosphate dehydrogenase deficiency: Biochemical versus genetic technologies. Semin Perinatol 2011;35(3):155–61.Google Scholar
Carpenter, KH, Wiley, V. Application of tandem mass spectrometry to biochemical genetics and newborn screening. Clin Chim Acta 2002;322(1–2):110.Google Scholar
Millington, DS, Sista, R, Eckhardt, A, et al. Digital microfluidics: A future technology in the newborn screening laboratory? Semin Perinatol 2010;34(2):163–9.Google Scholar
Heimall, J, Logan, BR, Cowan, MJ, et al. Immune reconstitution and survival of 100 SCID patients post-hematopoietic cell transplant: A PIDTC natural history study. Blood 2017;130(25):2718–27.Google Scholar
Griffith, LM, Cowan, MJ, Kohn, DB, et al. Allogeneic hematopoietic cell transplantation for primary immune deficiency diseases: Current status and critical needs. J Allergy Clin Immunol 2008;122(6):1087–96.Google Scholar
Gaspar, HB, Qasim, W, Davies, EG, et al. How I treat severe combined immunodeficiency. Blood 2013;122(23):3749–58.Google Scholar
Dvorak, CC, Cowan, MJ. Hematopoietic stem cell transplantation for primary immunodeficiency disease. Bone Marrow Transplant 2008;41(2):119–26.Google Scholar
Wahlstrom, JT, Dvorak, CC, Cowan, MJ. Hematopoietic stem cell transplantation for severe combined immunodeficiency. Curr Pediatr Rep 2015;3(1):110.Google Scholar
Malacarne, F, Benicchi, T, Notarangelo, LD, et al. Reduced thymic output, increased spontaneous apoptosis and oligoclonal B cells in polyethylene glycol-adenosine deaminase-treated patients. Eur J Immunol 2005;35(11):3376–86.CrossRefGoogle ScholarPubMed
Gaspar, HB, Aiuti, A, Porta, F, et al. How I treat ADA deficiency. Blood 2009;114(17):3524–32.Google Scholar
Ferrua, F, Aiuti, A. Twenty-five years of gene therapy for ADA-SCID: From bubble babies to an approved drug. Hum Gene Ther 2017;28(11):972–81.Google Scholar
Cavazzana, M, Six, E, Lagresle-Peyrou, C, Andre-Schmutz, I, Hacein-Bey-Abina, S. Gene therapy for X-linked severe combined immunodeficiency: Where do we stand? Hum Gene Ther 2016;27(2):108–16.Google Scholar
Hacein-Bey-Abina, S, Pai, SY, Gaspar, HB, et al. A modified gamma-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med 2014;371(15):1407–17.Google Scholar
De Ravin, SS, Wu, X, Moir, S, et al. Lentiviral hematopoietic stem cell gene therapy for X-linked severe combined immunodeficiency. Sci Transl Med 2016;8(335):335ra57.Google Scholar
Mamcarz, E, Zhou, S, Lockey, T, et al. Lentiviral gene therapy with low dose busulfan for infants with SCID-X1 (revised). N Engl J Med 2019;380:1525–34.Google Scholar
Aiuti, A, Roncarolo, MG, Naldini, L. Gene therapy for ADA-SCID, the first marketing approval of an ex vivo gene therapy in Europe: Paving the road for the next generation of advanced therapy medicinal products. EMBO Mol Med 2017;9(6):737–40.Google Scholar
Punwani, D, Kawahara, M, Yu, J, et al. Lentivirus mediated correction of Artemis-deficient severe combined immunodeficiency. Hum Gene Ther 2017;28(1):112–24.Google Scholar
Atlas, SA. The sickle cell trait and surgical complications: A matched-pair patient analysis. JAMA 1974;229(8):1078–80.Google Scholar
Metras, D, Coulibaly, AO, Ouattara, K, et al. Open-heart surgery in sickle-cell haemoglobinopathies: Report of 15 cases. Thorax 1982;37(7):486–91.Google Scholar
Naik, RP, Haywood, C, Jr. Sickle cell trait diagnosis: Clinical and social implications. Hematol Am Soc Hematol Educ Prog 2015;2015:160–7.Google Scholar
Holtkamp, KCA, Mathijssen, IB, Lakeman, P, et al. Factors for successful implementation of population-based expanded carrier screening: Learning from existing initiatives. Eur J Public Health 2017;27(2):372–7.Google Scholar
Kaback, MM. Population-based genetic screening for reproductive counseling: The Tay–Sachs disease model. Eur J Pediatr 2000;159 Suppl 3:S192–5.Google Scholar
De Wert, GM, Dondorp, WJ, Knoppers, BM. Preconception care and genetic risk: Ethical issues. J Community Genet 2012;3(3):221–8.Google Scholar
Henneman, L, Borry, P, Chokoshvili, D, et al. Responsible implementation of expanded carrier screening. Eur J Hum Genet 2016;24(6):e1e12.Google Scholar
Bombard, Y, Miller, FA, Hayeems, RZ, Avard, D, Knoppers, BM. Reconsidering reproductive benefit through newborn screening: A systematic review of guidelines on preconception, prenatal and newborn screening. Eur J Hum Genet 2010;18(7):751–60.Google Scholar
Mitchell, JJ, Capua, A, Clow, C, Scriver, CR. Twenty-year outcome analysis of genetic screening programs for Tay–Sachs and beta-thalassemia disease carriers in high schools. Am J Hum Genet 1996;59(4):793–8.Google Scholar
Lena-Russo, D, Badens, C, Aubinaud, M, et al. Outcome of a school screening programme for carriers of haemoglobin disease. J Med Screen 2002;9(2):67–9.Google Scholar
Amato, A, Cappabianca, MP, Lerone, M, et al. Carrier screening for inherited haemoglobin disorders among secondary school students and young adults in Latium, Italy. J Community Genet 2014;5(3):265–8.Google Scholar
van Elderen, T, Mutlu, D, Karstanje, J, et al. Turkish female immigrants’ intentions to participate in preconception carrier screening for hemoglobinopathies in the Netherlands: An empirical study. Public Health Genomics 2010;13(7–8):415–23.Google Scholar
Saffi, M, Howard, N. Exploring the effectiveness of mandatory premarital screening and genetic counselling programmes for beta-thalassaemia in the Middle East: A scoping review. Public Health Genomics 2015;18(4):193203.Google Scholar
Kihlbom, U. Ethical issues in preconception genetic carrier screening. Ups J Med Sci 2016:14.Google Scholar
Langlois, S, Benn, P, Wilkins-Haug, L. Current controversies in prenatal diagnosis 4: Pre-conception expanded carrier screening should replace all current prenatal screening for specific single gene disorders. Prenat Diagn 2015;35(1):23–8.Google Scholar
Nazareth, SB, Lazarin, GA, Goldberg, JD. Changing trends in carrier screening for genetic disease in the United States. Prenat Diagn 2015;35(10):931–5.Google Scholar
Lazarin, GA, Haque, IS. Expanded carrier screening: A review of early implementation and literature. Semin Perinatol 2016;40(1):2934.Google Scholar
Ciske, DJ, Haavisto, A, Laxova, A, Rock, LZ, Farrell, PM. Genetic counseling and neonatal screening for cystic fibrosis: An assessment of the communication process. Pediatrics 2001;107(4):699705.Google Scholar
Lewis, S, Curnow, L, Ross, M, Massie, J. Parental attitudes to the identification of their infants as carriers of cystic fibrosis by newborn screening. J Paediatr Child Health 2006;42(9):533–7.Google Scholar
Hayeems, RZ, Bytautas, JP, Miller, FA. A systematic review of the effects of disclosing carrier results generated through newborn screening. J Genet Couns 2008;17(6):538–49.Google Scholar
Omran, AR. The epidemiologic transition. A theory of the epidemiology of population change. Milbank Mem Fund Q 1971;49(4):509–38.Google Scholar
Darlison, MW, Modell, B. Sickle-cell disorders: Limits of descriptive epidemiology. Lancet 2013;381(9861):98–9.Google Scholar

References

Beutler, E, Waalen, J. The definition of anemia: What is the lower limit of normal of the blood hemoglobin concentration? Blood 2006;107:1747–50.Google Scholar
Henry, E, Christensen, RD. Reference intervals in neonatal hematology. Clin Perinatol 2015;42:483–97.Google Scholar
Wintrobe, MM. Blood of normal men and women. Bull John Hopkins Hosp 1933;53:118–40.Google Scholar
Jopling, J, Henry, E, Wiedmeier, SE, Christensen, RD. Reference ranges for hematocrit and blood hemoglobin concentration during the neonatal period: Data from a multihospital health care system. Pediatrics 2009;123:e3337.Google Scholar
Ruíz-Argüelles, GJ, Sanchez-Medal, L, Loria, A, et al. Red cell indices in normal adults residing at altitude from sea level to 2670 meters. Am J Hematol 1980;8:265–71.Google Scholar
Oh, W, Lind, J. Venous and capillary hematocrit in newborn infants and placental transfusion. Acta Paediatr Scand 1966;55:3842.Google Scholar
Linderkamp, O, Versmold, HT, Strohhacker, I, et al. Capillary–venous hematocrit differences in newborn infants. 1. Relationship to blood volume, peripheral blood flow, and acid-base parameters. Eur J Pediatr 1977:127;914.Google Scholar
Judkins, AJ, MacQueen, BC, Christensen, RD, et al. Automated quantification of fragmented red blood cells: Neonatal reference intervals and clinical disorders of neonatal intensive care unit patients with high values. Neonatology. 2018;115:512.Google Scholar
Christensen, RD, Lambert, DK, Richards, DS. Estimating the nucleated red blood cell ‘emergence time’ in neonates. J Perinatol 2014;34:116–19.Google Scholar
Christensen, RD, Albertine, KH, Dahl, MJ, et al. Nucleated red blood cell counts in term neonates following fetal hypoxia. Neonatology (in press).Google Scholar
Kim, YA, Makar, RS. Detection of fetomaternal hemorrhage. Am J Hematol 2012;87:417–23.Google Scholar
Badior, KE, Casey, JR. Molecular mechanism for the red blood cell senescence clock. IUBMB Life 2018;70:3240.Google Scholar
Christensen, RD, Yaish, HM. Hemolytic disorders causing severe neonatal hyperbilirubinemia. Clin Perinatol 2015;42:515–27.Google Scholar
Christensen, RD, Lambert, DK, Henry, E, et al. End-tidal carbon monoxide as an indicator of the hemolytic rate. Blood Cells Mol Dis 2015;54:292–6.Google Scholar
Christensen, RD, Malleske, DT, Lambert, DK, Baer, VL, et al. Measuring end-tidal carbon monoxide of jaundiced neonates in the birth hospital to identify those with hemolysis. Neonatology 2016;109:15.Google Scholar
Bhutani, VK, Maisels, MJ, Schutzman, DL, Castillo Cuadrado, ME, et al. Identification of risk for neonatal haemolysis. Acta Paediatr 2018;107:1350–6.CrossRefGoogle ScholarPubMed
Tidmarsh, GF, Wong, RJ, Stevenson, DK. End-tidal carbon monoxide and hemolysis. J Perinatol 2014;34:577–81.Google Scholar
Christensen, RD, Yaish, HM, Lemons, RS. Neonatal hemolytic jaundice: Morphologic features of erythrocytes that will help you diagnose the underlying condition. Neonatology 2014;105:243–9.Google Scholar
MacQueen, BC, Christensen, RD, Ward, DM, et al. The iron status at birth of neonates with risk factors for developing iron deficiency: A pilot study. J Perinatol 2017;37:436–40.Google Scholar
MacQueen, BC, Robert, D. Christensen, RD, Baer, VL, Ward, DM, Snow, GL. Screening umbilical cord blood for congenital iron deficiency. Blood Cells Mol Dis 2019;77:95100.Google Scholar
Yoon, D, Ponka, P, Prchal, JT. Hypoxia 5. Hypoxia and hematopoiesis. Am J Physiol Cell Physiol 2011;300:C1215C1222.Google Scholar
Shih, HM, Wu, CJ, Lin, SL. Physiology and pathophysiology of renal erythropoietin-producing cells. J Formos Med Assoc 2018;117:955–63.Google Scholar

References

Zanjani, ED, Ascensao, JL, McGlave, PB, Banisadre, M, Ash, RC. Studies in the liver to kidney switch of erythropoietin production. J Clin Invest 1981;67:1183–8.Google Scholar
Eckardt, K-U, Ratcliffe, PJ, Tan, CC, Bauer, C, Kurtz, A. Age-dependent expression of the erythropoietin gene in rat liver and kidneys. J. Clin. Invest 1992;89:753–60.Google Scholar
Kling, PJ, Schmidt, RL, Roberts, RA, Widness, JA. Serum erythropoietin levels during infancy: Associations with erythropoiesis. J Pediatr 1996;128:791–6.Google Scholar
Stockman, JA, III, Garcia, JF, Oski, FA. The anemia of prematurity: Factors governing the erythropoietin response. N Engl J Med 1977;296:647–50.Google Scholar
Ohls, RK. Evaluation and treatment of anemia in the neonate. In Christensen, RD, ed. Hematologic Problems of the Neonate (Philadelphia, PA: WB Saunders, 2000). pp. 137–69.Google Scholar
Lundstrom, U, Siimes, MA. At what age does iron supplementation become necessary in low-birth weight infants? J Pediatr 1977;91:878–83.CrossRefGoogle ScholarPubMed
Ohls, RK. Erythropoietin to prevent and treat the anemia of prematurity. Curr Opin Pediatr 1999;11:108–14.Google Scholar
Maier, RF, Obladen, M, Messinger, D, Wardrop, CAJ. Factors related to transfusion in very low birthweight infants treated with erythropoietin. Arch Dis Child 1996;74:F182F186.Google Scholar
Ohls, RK. Erythropoietin treatment in extremely low birth weight infants: Blood in versus blood out. J Pediatr 2002;141:36.Google Scholar
Kling, PJ, Sullivan, TM, Leftwich, ME, Roe, DJ. Score for neonatal acute physiology predicts erythrocyte transfusions in premature infants. Arch Dis Pediatr Adolesc Med 1997;151:2731.Google Scholar
Ekhaguere, OA, Morriss, FH, Jr., Bell, EF, Prakash, N, Widness, JA. Predictive factors and practice trends in red blood cell transfusions for very-low-birth-weight infants. Pediatr Res 2016;79:736–41.Google Scholar
Shannon, KM, Keith, JM, Mentzer, WC, et al. Recombinant human erythropoietin stimulates erythropoiesis and reduces erythrocyte transfusions in very low birth weight preterm infants. Pediatrics 1995;95:110.Google Scholar
Widness, JA, Seward, VJ, Kromer, IJ, et al. Changing patterns of red blood cell transfusion in very low birth weight infants. J Pediatr 1996;129:680–7.Google Scholar
Hume, H. Red blood cell transfusions for preterm infants: The role of evidence-based medicine. Sem Perinatol 1997;21:819.Google Scholar
Bednarek, FJ, Weisberger, S, Richardson, DK, et al. Variations in blood transfusions among newborn intensive care units. J Pediatr 1998;133:601–7.Google Scholar
Ringer, SA, Richardson, DK, Sacher, RA, Keszler, M, Churchill, WH. Variations in transfusion practice in neonatal intensive care. Pediatrics 1998;101:194200.Google Scholar
Bell, EF, Strauss, RG, Widness, JA, et al. Randomized trial of liberal versus restrictive guidelines for red blood cell transfusion in preterm infants. Pediatrics 2005;115:1685–91.Google Scholar
Luban, NL. Management of anemia in the newborn. Early Hum Dev 2008;84:493–8.Google Scholar
Becquet, O, Guyot, D, Kuo, P, et al. Respective effects of phlebotomy losses and erythropoietin treatment on the need for blood transfusion in very premature infants. BMC Pediatr 2013;13:176.Google Scholar
Maier, RF, Sonntag, J, Walka, MM, et al. Changing practices of red blood cell transfusions in infants with birth weights less than 1000 g. J Pediatr 2000;136:220–4.Google Scholar
Donato, H, Vain, N, Rendo, P, et al. Effect of early versus late administration of human recombinant erythropoietin on transfusion requirements in premature infants: Results of a randomized, placebo-controlled, multicenter trial. Pediatrics 2000;105:10661072.Google Scholar
Widness, JA, Kulhavy, JC, Johnson, KJ, et al. Clinical performance of an in-line point-of-care monitor in neonates. Pediatrics 2000;106:497504.Google Scholar
Moya, MP, Clark, RH, Nicks, J, Tanaka, DT. The effects of bedside blood gas monitoring on blood loss and ventilator management. Biol Neonate 2001;80:257–61.Google Scholar
Lin, JC, Strauss, RG, Kulhavy, JC, et al. Phlebotomy overdraw in the neonatal intensive care nursery. Pediatrics.2000;106:e19.Google Scholar
Bishara, N, Ohls, RK. Current controversies in the management of the anemia of prematurity. Semin Perinatol 2009;33:2934.Google Scholar
Kinmond, S, Aitchison, TC, Holland, BM, et al. Umbilical cord clamping and preterm infants: A randomised trial. Brit Med J 1993;306:172–5.Google Scholar
Wardrop, CAJ, Holland, BM. The roles and vital importance of placental blood to the newborn infant. J Perinat Med 1995;23:139–43.Google Scholar
Ibrahim, HM, Krouskop, RW, Lewis, DF, Dhanireddy, R. Placental transfusion: Umbilical cord clamping and preterm infants. J Perinatol.2000;20:351–4.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;8:CD003248.Google Scholar
Committee on Obstetric Practice. Committee opinion no. 684: Delayed umbilical cord clamping after birth. Obstet Gynecol 2017;129:e5e10.Google Scholar
Strauss, RG. How I transfuse red blood cells and platelets to infants with the anemia and thrombocytopenia of prematurity. Transfusion 2008;48:209–17.Google Scholar
Aladangady, N, Aitchison, TC, Beckett, C, et al. Is it possible to predict the blood volume of a sick preterm infant? Arch Dis Child Fetal Neonatal Ed 2004;89:F344–7.Google Scholar
Backes, CH, Rivera, BK, Haque, U, et al. Placental transfusion strategies in very preterm neonates: A systematic review and meta-analysis. Obstet Gynecol 2014;124:4756.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;297:1241–52.Google Scholar
Chaparro, CM, Neufeld, LM, Alvarez, GT, Cedillo, RE-L, Dewey, KG. Effect of timing of umbilical cord clamping on iron status in Mexican infants: A randomised controlled trial. Lancet 2006;367:19972004.Google Scholar
Ohls, RK, Ehrenkranz, RA, Wright, LL, et al. The effects of early erythropoietin therapy on the transfusion requirements of preterm infants below 1250 grams birthweight: A multicenter, randomized controlled trial. Pediatrics 2001;108:934–42.Google Scholar
Roseff, SD, Luban, NL, Manno, CS. Guidelines for assessing appropriateness of pediatric transfusion. Transfusion 2002;42:1398–413.Google Scholar
Strauss, RG. Practical issues in neonatal transfusion practice. Am J Clin Path 1997;107:S57S63.Google Scholar
Simon, TL, Alverson, DC, AuBuchon, J, et al. Practice parameter for the use of red blood cell transfusions. Arch Pathol Lab Med 1998;122:130–8.Google Scholar
Kirpalani, H, Whyte, RK, Andersen, C, et al. The Premature Infants in Need of Transfusion (PINT) study: A randomized, controlled trial of a restrictive (low) versus liberal (high) transfusion threshold for extremely low birth weight infants. J Pediatr 2006;149:301–7.Google Scholar
Whyte, R, Kirpalani, H. Low versus high haemoglobin concentration threshold for blood transfusion for preventing morbidity and mortality in very low birth weight infants. Cochrane Database Syst Rev 2011;11:CD000512.Google Scholar
Ibrahim, M, Ho, SK, Yeo, CL. Restrictive versus liberal red blood cell transfusion thresholds in very low birth weight infants: A systematic review and meta-analysis. J Paediatr Child Health 2014;50:122–30.Google Scholar
McCoy, TE, Conrad, AL, Richman, LC, et al. Neurocognitive profiles of preterm infants randomly assigned to lower or higher hematocrit thresholds for transfusion. Child Neuropsychol 2011;17:347–67.Google Scholar
McCoy, TE, Conrad, AL, Richman, LC, et al. The relationship between brain structure and cognition in transfused preterm children at school age. Dev Neuropsychol 2014;39:226–32.Google Scholar
Whyte, RK, Kirpalani, H, Asztalos, EV, et al. Neurodevelopmental outcome of extremely low birth weight infants randomly assigned to restrictive or liberal hemoglobin thresholds for blood transfusion. Pediatrics 2009;123:207–13.Google Scholar
Warwood, TL, Ohls, RK, Lambert, DK, et al. Intravenous administration of darbepoetin to NICU patients. J Perinatol 2006;26:296300.Google Scholar
Vamvakas, EC, Strauss, RG. Meta-analysis of controlled clinical trials studying the efficacy of rHuEPO in reducing blood transfusions in the anemia of prematurity. Transfusion 2001;41:406–15.Google Scholar
Ohlsson, A, Aher, SM. Early erythropoiesis-stimulating agents in preterm or low birth weight infants. Cochrane Database Syst Rev 2017;11:CD004863.Google Scholar
Aher, SM, Ohlsson, A. Late erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2014;4:CD004868.Google Scholar
Aher, SM, Ohlsson, A. Early versus late erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2012;10:CD004865.Google Scholar
Maier, RF, Obladen, M, Kattner, E, et al. High- versus low-dose erythropoietin in extremely low birth weight infants. J Pediatr 1998;132:866–70.Google Scholar
Carnielli, VP, D Riol, R, Montini, G. Iron supplementation enhances response to high doses of recombinant human erythropoietin in preterm infants. Arch Dis Child 1998;79:F44F48.Google Scholar
Brown, MS, Jones, MA, Ohls, RK, Christensen, RD. Single-dose pharmacokinetics of recombinant human erythropoietin in preterm infants after intravenous and subcutaneous administration. J Pediatr 1993;122:655–7.Google Scholar
Widness, JA, Veng-Pedersen, P, Peters, C, et al. Erythropoietin pharmacokinetics in premature infants: Developmental, non-linearity, and treatment effects. J Appl Physiol 1996;80:140–8.Google Scholar
Ohls, RK, Veerman, MW, Christensen, RD. Pharmacokinetics and effectiveness of recombinant erythropoietin administered to preterm infants by continuous infusion in total parenteral nutrition solution. J Pediatr 1996;128:518–23.Google Scholar
Brown, MS, Keith, JF. Comparison between two and five doses a week of recombinant human erythropoietin for anemia of prematurity: A randomized trial. Pediatrics 1999;104:210–15.Google Scholar
Bechensteen, AG, Håga, P, Halvorsen, S, et al. Erythropoietin, protein, and iron supplementation and the prevention of anaemia of prematurity. Arch Dis Child 1993;69:1923.Google Scholar
Messer, J, Haddad, J, Donato, L, Astruc, D, Matis, J. Early treatment of premature infants with recombinant human erythropoietin. Pediatrics 1993;92:519–23.Google Scholar
Bechensteen, AG, Halvorsen, S, Haga, P, PM C, Liestøl, K. Erythropoietin (EPO), protein and iron supplementation and the prevention of anaemia of prematurity: Effects on serum immunoreactive Epo, growth and protein and iron metabolism. Acta Paediatr 1996;85:490–5.Google Scholar
Worthington-White, D, Behnke, M, Gross, S. Premature infants require additional folate and vitamin B-12 to reduce the severity of the anemia of prematurity. Am J Clin Nutr 1994;60:930–5.Google Scholar
Haiden, N, Schwindt, J, Cardona, F, et al. Effects of a combined therapy of erythropoietin, iron, folate, and vitamin B12 on the transfusion requirements of extremely low birth weight infants. Pediatrics 2006;118:2004–13.Google Scholar
Carnielli, VP, Montini, G, Da Riol, R, Dall’Amico, R, Cantarutti, F. Effect of high doses of human recombinant erythropoietin on the need for blood transfusions in preterm infants. J Pediatr 1992;121:98102.Google Scholar
Meyer, MP, Haworth, C, Meyer, JH, Commerford, A. A comparison of oral and intravenous iron supplementation in preterm infants receiving recombinant erythropoietin. J Pediatr 1996;129:258–63.Google Scholar
Kivivuori, SM, Virtanen, M, Raivio, KO, Viinikka, L, Siimes, MA. Oral iron is sufficient for erythropoietin treatment of very low birth-weight infants. Eur J Pediatr 1999;158:147–51.Google Scholar
Pollak, A, Hayde, M, Hayn, M, et al. Effect of intravenous iron supplementation on erythropoiesis in erythropoietin-treated premature infants. Pediatrics 2001;107:7885.Google Scholar
Ohls, RK, Harcum, J, Schibler, KR, Christensen, RD. The effect of erythropoietin on the transfusion requirements of preterm infants weighing 750 grams or less: A randomized, double-blind, placebo-controlled study. J Pediatr 1997;131:661–5.Google Scholar
Kumar, P, Shankaran, S, Krishnan, RG. Recombinant human erythropoietin therapy for treatment of anemia of prematurity in very low birth weight infants: A randomized, double-blind, placebo-controlled trial. J Perinatol 1998;18:173–7.Google Scholar
Ohls, RK, Ehrenkranz, RA, Lemons, JA, et al. A multicenter randomized double-masked placebo-controlled trial of early erythropoietin and iron administration to preterm infants. Pediatr Res 1999;45:216A.Google Scholar
Zipursky, A. The risk of hematopoietic growth factor therapy in newborn infants. Pediatr Res 2002;51:549.Google Scholar
Ohls, RK, Christensen, RD. Recombinant erythropoietin compared with erythrocyte transfusion in the treatment of anemia of prematurity. J Pediatr 1991;119:781–8.Google Scholar
Beck, D, Masserey, E, Meyer, M, Calame, A. Weekly intravenous administration of recombinant human erythropoietin in infants with the anaemia of prematurity. Eur J Pediatr 1991;150:767–72.Google Scholar
Halperin, DS, Wacker, P, Lacourt, G, et al. Effects of recombinant human erythropoietin in infants with anemia of prematurity: A pilot study. J Pediatr 1990;116:779–86.Google Scholar
Emmerson, A. Double blind trial of recombinant human erythropoietin in preterm infants: Comment. Archives Dis Child 1993;69:542.Google Scholar
Brown, MS, Baron, AE, France, EK, Hamman, RF. Association between higher cumulative doses of recombinant erythropoietin and risk for retinopathy of prematurity. J AAPOS 2006;10:143–9.Google Scholar
Ashley, RA, Dubuque, SH, Dvorak, B, et al. Erythropoietin stimulates vasculogenesis in neonatal rat mesenteric microvascular endothelial cells. Pediatr Res 2002;51:472–8.Google Scholar
Rao, R, Georgieff, MK. Iron in fetal and neonatal nutrition. Semin Fetal Neonatal Med 2007;12:5463.Google Scholar
Ledbetter, DJ, Juul, SE. Erthropoietin and the incidence of necrotizing enterocolitis in infants with very low birth weight. J Pediatr Surg 2000;35:178–82.Google Scholar
Ohls, RK, Osborne, KA, Chrstensen, RD. Efficacy and cost analysis of treating very low birth weight infants with erythropoietin during their first two weeks of life: A randomized, placebo-controlled trial. J Pediatr 1995;126:421–6.Google Scholar
Bierer, R, Peceny, MC, Hartenberger, CH, Ohls, RK. Erythropoietin concentrations and neurodevelopmental outcome in preterm infants. Pediatrics. 2006;118:e635-640.Google Scholar
Ohls, RK, Cannon, DC, Phillips, J, et al. Preschool assessment of preterm infants treated with darbepoetin and erythropoietin. Pediatrics 2016;137:e20153859.Google Scholar
Neubauer, AP, Voss, W, Wachtendorf, M, Jungmann, T. Erythropoietin improves neurodevelopmental outcome of extremely preterm infants. Ann Neurol 2010;67:657–66.Google Scholar
Massaro, AN, Wu, YW, Bammler, TK, et al. Plasma biomarkers of brain injury in neonatal hypoxic-ischemic encephalopathy. J Pediatr 2018;194:6775.Google Scholar
Juul, SE, Comstock, BA, Heagerty, PJ, et al. High-dose erythropoietin for asphyxia and encephalopathy (HEAL): A randomized controlled trial – background, aims, and study protocol. Neonatology 2018;113:331–8.Google Scholar
Garg, B, Sharma, D, Bansal, A. Systematic review seeking erythropoietin role for neuroprotection in neonates with hypoxic ischemic encephalopathy: Presently where do we stand. J Matern Fetal Neonatal Med 2018;31:3214–24.Google Scholar
Fischer, HS, Reibel, NJ, Buhrer, C, Dame, C. Prophylactic early erythropoietin for neuroprotection in preterm infants: A meta-analysis. Pediatrics 2017;139:e20164317.Google Scholar
Juul, SE, Mayock, DE, Comstock, BA, Heagerty, PJ. Neuroprotective potential of erythropoietin in neonates; design of a randomized trial. Matern Health Neonatol Perinatol 2015;1:27.Google Scholar
Fauchere, JC, Koller, BM, Tschopp, A, et al. Swiss erythropoietin neuroprotection trial g. safety of early high-dose recombinant erythropoietin for neuroprotection in very preterm infants. J Pediatr 2015;167:52–7 e53.Google Scholar
Natalucci, G, Latal, B, Koller, B, et al. Swiss EPONTG. Effect of early prophylactic high-dose recombinant human erythropoietin in very preterm infants on neurodevelopmental outcome at 2 years: A randomized clinical trial. JAMA 2016;315:2079–85.Google Scholar
Shireman, TI, Hilsenrath, PE, Strauss, RG, Widness, JA, Mutnick, AH. Recombinant human erythropoietin vs transfusions in the treatment of anemia of prematurity. Arch Pediatr Adolesc Med 1994;148:582–8.Google Scholar
Maier, RF, Obladen, M, Scigalla, P, et al. The effect of epoetin beta (recombinant human erythropoietin) on the need for transfusion in very-low-birth-weight infants. N Engl J Med 1994;330:1173–8.Google Scholar
Fain, J, Hilsenrath, P, Widness, JA, Strauss, RG, Mutnick, AH. A cost analysis comparing erythropoietin and red cell transfusions in the treatment of anemia of prematurity. Transfusion 1995;35:936–43.Google Scholar
Yeo, CL, Choo, S, Ho, LY. Effect of recombinant human erythropoietin on transfusion needs in preterm infants. J Paediatr Child Health 2001;37:352–8.Google Scholar
Atasay, B, Gunlemez, A, Akar, N, Arsan, S. Does early erythropoietin therapy decrease transfusions in anemia of prematurity. Indian J Pediatr 2002;69:389–91.Google Scholar
Avent, M, Cory, BJ, Galpin, J, et al. A comparison of high versus low dose recombinant human erythropoietin versus blood transfusion in the management of anaemia of prematurity in a developing country. J Tropical Pediatr 2002;48:227–33.Google Scholar
Oski, FA. Iron deficiency in infancy and childhood. New Engl J Med 1993;329:190–3.Google Scholar
Oski, FA. Differential diagnosis of anemia. In Nathan, DG, Oski, FA, eds. 4th ed. Hematology of Infancy and Childhood (Philadelphia, PA: W.B. Saunders, 1993). pp. 346–53.Google Scholar
Dallman, PR. Iron deficiency in the weanling: A nutritional problem on the way to resolution. Acta Paediatr Scand Suppl 1986;323:5967.Google Scholar
Harthoorn-Lasthuizen, EJ, Lindemans, J, Langenhuijsen, MM. Does iron-deficient erythropoiesis in pregnancy influence fetal iron supply? Acta Obstet Gynecol Scand 2001;80:392–6.Google Scholar
McCarthy, PJ, Zundel, HR, Johnson, KR, Blohowiak, SE, Kling, PJ. Impact of growth restriction and other prenatal risk factors on cord blood iron status in prematurity. J Pediatr Hematol Oncol 2016;38(3):210–15.Google Scholar
Mukhopadhyay, K, Yadav, RK, Kishore, SS, et al. Iron status at birth and at 4 weeks in preterm-SGA infants in comparison with preterm and term-AGA infants. J Matern Fetal Neonatal Med 2012;25:1474–8.Google Scholar
McLimore, HM, Phillips, AK, Blohowiak, SE, et al. Impact of multiple prenatal risk factors on newborn iron status at delivery. J Pediatr Hematol Oncol 2013;35:473–77.Google Scholar
Jones, AD, Zhao, G, Jiang, YP, et al. Maternal obesity during pregnancy is negatively associated with maternal and neonatal iron status. Eur J Clin Nutr 2016;70(8):918–24.Google Scholar
MacQueen, BC, Christensen, RD, Ward, DM, et al. The iron status at birth of neonates with risk factors for developing iron deficiency: A pilot study. J Perinatol 2017;37:43640.Google Scholar
McCarthy, EK, Kenny, LC, Hourihane, JOB, et al. Impact of maternal, antenatal and birth-associated factors on iron stores at birth: Data from a prospective maternal-infant birth cohort. Eur J Clin Nutr 2017;71:782–7.Google Scholar
Fomon, SJ. Iron. In Fomon, S, ed. Nutrition of Normal Infants (St. Louis, MO: Mosby, 1993). pp. 239–60.Google Scholar
Ehrenkranz, RA. Iron requirements of preterm infants. Nutrition 1994;10:77–8.Google Scholar
Fomon, SJ, Ziegler, EE, Nelson, SE, Serfass, RE, Frantz, JA. Erythrocyte incorporation of iron by 56-day-old infants fed a 58Fe-labeled supplement. Pediatr Res 1995;38:373–8.Google Scholar
Widness, JA, Lombard, KA, Ziegler, EE, et al. Erythroctye incorporation and absorption of 58Fe in premature infants treated with erythropoietin. Pediatr Res 1997;41:416–23.Google Scholar
Georgieff, MK. Long-term brain and behavioral consequences of early iron deficiency. Nutr Rev 2011;69 Suppl 1:S4348.Google Scholar
Saarinen, UM, Siimes, MA. Serum ferritin in assessment of iron nutrition in healthy infants. Acta Paediatr Scand 1978;67:745–51.Google Scholar
Al-Kharfy, T, Smyth, JA, Wadsworth, L, et al. Erythropoietin therapy in neonates at risk of having bronchopulmonary dysplasia and requiring multiple transfusions. J Pediatr 1996;129:8996.Google Scholar
Bader, D, Blondheim, O, Jonas, R, et al. Decreased ferritin levels, despite iron supplementation, during erythropoietin therapy in anaemia of prematurity. Acta Paediatr 1996;85:496501.Google Scholar
Bechensteen, AG, Halvorsen, S, Haga, P, Cotes, PM, Liestol, K. Erythropoietin, protein and iron supplementation and the prevention of anaemia of prematurity: Effects on serum immunoreactive Epo, growth and protein and iron metabolism. Acta Paediatr. 1996;85:490–5.Google Scholar
Emmerson, A. Role of erythropoietin in the newborn. Archives Dis Child 1993;69:273–5.Google Scholar
Meyer, MP, Meyer, JH, Commerford, A, et al. Recombinant human erythropoietin in the treatment of the anemia of prematurity: Results of a double-blind, placebo-controlled study. Pediatrics 1994;93:918–23.Google Scholar
Morris, KP, Watson, S, Reid, MM, Hamilton, PJ, Coulthard, MG. Assessing iron status in children with chronic renal failure on erythropoietin: Which measurements should we use? Pediatr Nephrol 1994;8:51–6.Google Scholar
Goodnough, LT, Skikne, B, Brugnara, C. Erythropoietin, iron and erythropoiesis. Blood 2000;96:823–33.Google Scholar
Akkermans, MD, Uijterschout, L, Abbink, M, et al. Predictive factors of iron depletion in late preterm infants at the postnatal age of 6 weeks. Eur J Clin Nutr 2016;70:941–6.Google Scholar
Taetle, R. The role of transferrin receptors in hemopoietic cell growth. Exp Hematol 1990;18:360–5.Google Scholar
Kivivuori, SM, Heikinheimo, M, Teppo, A-M, Siimes, MA. Early rise in serum concentration of transferrin receptor induced by recombinant human erythropoietin in very-low-birth-weight infants Pediatr Res. 1994;36:85–9.Google Scholar
Bechensteen, AG, Haga, P, Halvorsen, S, et al. Effect of low and moderate doses of recombinant human erythropoietin on the haematological response in premature infants on a high protein and iron intake. Eur J Pediatr 1997;156:5661.Google Scholar
Kohgo, Y, Niitsu, Y, Kondo, H, et al. Serum transferrin receptor as a new index of erythropoiesis. Blood 1987;70:1955–8.Google Scholar
Beguin, Y, Clemons, GK, Pootrakul, P, Fillet, G. Quantitative assessment of erythropoiesis and functional classification of anemia based on measurements of serum transferrin receptor and erythropoietin. Blood 1993;81:1067–76.Google Scholar
Beguin, Y, Loo, M, S. RZ, et al. Early prediction of response to recombinant human erythropoietin in patients with the anemia of renal failure by serum transferrin receptor and fibrinogen. Blood 1993;82:2010–16.Google Scholar
Rusia, U, Flowers, C, Madan, N, Agarwal, N, Sood, SK. Serum transferrin receptor levels in the evaluation of iron deficiency in the neonate. Acta Paediatr Japon 1995;38:455–9.Google Scholar
Kling, PJ, Widness, JA. Transfusions, (RBC Tx) and erythropoiesis indicators influence serum transferrin receptors (TfR) levels in premature infants. Pediatr Res 1995;37:282A.Google Scholar
Kling, PJ, Roberts, RA, Widness, JA. Plasma transferrin receptor levels and indices of erythropoiesis and iron status in healthy term infants. Am J Pediatr Hem/Onc 1997;20:309–14.Google Scholar
Krallis, N, Cholevas, V, Mavridis, A, et al. Effect of recombinant human erythropoietin in preterm infants. Eur J Haematol 1999;63:71–6.Google Scholar
Kasper, DC, Widness, JA, Haiden, N, Berger, A, et al. Characterization and differentiation of iron status in anemic very low birth weight infants using a diagnostic nomogram. Neonatology 2009;95:164–71.Google Scholar
Braun, J, Lindner, K, Schreiber, M, Heidler, RA, Horl, WH. Percentage of hypochromic red blood cells as predictor of erythropoietic and iron response after i.v. iron supplementation in maintenance haemodialysis patients. Nephrol Dial Transplan 1997;12:1173–81.Google Scholar
Rettmer, RL, Carlson, TH, Origenes, ML, Jack, RM, Labbe, RF. Zinc protoporphyrin/heme ratio for diagnosis of preanemic iron deficiency. Pediatrics 1999;104:e37.Google Scholar
Kaltwasser, JP, Gottschalk, R. Erythropoietin and iron. Kidney Int 1999;55:S49S56.Google Scholar
National Kidney Foundation. Clinical Practice Guidelines for the Treatment of Anemia of Chronic Renal Failure (New York: National Kidney Foundation; 1999), available online at https://bit.ly/33S1B23.Google Scholar
Lott, DG, Zimmerman, MB, Labbe’, RF, Kling, PJ, Widness, JA. Erythrocyte zinc protoporphyrin ratios are elevated with prematurity and with fetal hypoxia. Pediatrics 2005;116:414–22.Google Scholar
Miller, SM, McPherson, RJ, Juul, SE. Iron sulfate supplementation decreases zinc protoporphyrin to heme ratio in premature infants. J Pediatr 2006;148:44–8.Google Scholar
Winzerling, JJ, Kling, PJ. Iron deficient erythropoiesis in premature infants measured by blood zinc protoporphyrin/heme. J Pediatr 2001;139:134–6.Google Scholar
Blohowiak, SE, Chen, ME, Repyak, KS, et al. Reticulocyte enrichment of zinc protoporphyrin/heme discriminates impaired iron supply during early development. Pediatr Res 2008;64:63–7.Google Scholar
Shaw, JCL. Iron absorption by the premature infant: The effect of transfusion and iron supplements on the serum ferritin levels. Acta Paediatr Scand Suppl 1982;299:83–9.Google Scholar
Arad, I, Konijn, AM, Linder, N, Goldstein, MDM, Kaufmann, NA. Serum ferritin levels in preterm infants after multiple blood transfusions. Am J Perinatol 1988;5:40–3.Google Scholar
Brown, MS. Effect of transfusion and phlebotomy on serum ferritin levels in low birth weight infants. J Perinatol 1996;16:3942.Google Scholar
Soubasi, V, Kremenopoulos, G, Diamanti, E, et al. Follow-up of very low birth weight infants after erythrpoietin treatment to prevent anemia of prematurity. J Pediatr 1995;127:291–7.Google Scholar
Sullivan, JL. Iron, plasma antioxidants and the ‘oxygen radical disease of prematurity’. Am J Dis Child 1988;142:1341–4.Google Scholar
Evans, PJ, Evans, R, Kovar, IZ, Holton, AF, Halliwell, B. Bleomycin-detectable iron in the plasma of premature and full-term neonates. FEBS Letters 1992;303:210–12.Google Scholar
Miller, NJ, Rice-Evans, C, Davies, MJ, Gopinathan, V, Milner, A. A novel method for measuring antioxidant capacity and its application to monitoring the antioxidant status in premature neonates. Clin Sci 1993;84:407–12.Google Scholar
Humphrey, MJ, Harrell-Bean, HA, Eskelson, C, Corrigan, JJ. Blood transfusion in the neonate: Effects of dilution and age of blood on hemolysis. J Pediatr 1982;101:605–7.Google Scholar
Kling, PJ, Reichard, RD, Roberts, RA, Winzerling, JJ, Woodward, SS. The effects of transfusions on oxidative stress and plasma erythropoietin levels in premature infants. Ann Hematol 2000;79:B13.Google Scholar
Obladen, M, Maier, R, Grauel, L, et al. Recombinant human erythropoietin (rhEPO) for prevention of anaemia of prematurity: A randomized multicentre trial. Pediatr Res 1990;28:287A(Abstr).Google Scholar
Shannon, KM, Mentzer, WC, Abels, RI, et al. Recombinant human erythropoietin in anemia of prematurity: Preliminary results of a double-blind placebo controlled pilot study. J Pediatr 1991;118:949–55.Google Scholar
Shannon, KM, Mentzer, WC, Abels, RI, et al. Enhancement of erythropoiesis by recombinant human erythropoietin in low birth weight infants: A pilot study. J Pediatr 1992;120:586–92.Google Scholar
Soubasi, V, Kremenpoulos, G, Diamandi, E, Tsantali, C, Tsakiris, D. In which neonates does early recombinant human erythropoietin treatment prevent anemia of prematurity? Results of a randomized, controlled study. Pediatr Res 1993;34:675–9.Google Scholar
Ronnestad, A, Moe, PJ, Breivik, N. Enhancement of erythropoiesis by erythropoietin, bovine protein and energy fortified mother’s milk during anaemia of prematurity. Acta Paediatrica 1995;84:809–11.Google Scholar
Samanci, N, Ovali, F, Dagoglu, T. Effects of recombinant human erythropoietin in infants with very low birth weights. J Int Med Res 1996;24:190–8.Google Scholar
Giannakopoulou, C, Bolonaki, I, Stiakaki, E, et al. Erythropoietin (rHuEPO) administration to premature infants for the treatment of their anemia. Pediatr Hematol Oncol 1998;15:3743.Google Scholar
Maier, RF, Obladen, M, Mueller-Hansen, I, et al. Early treatment with erythropoietin beta ameliorates anemia and reduces transfusion requirements in infants with birth weights below 1000 g. J Pediatr 2002;141:815.Google Scholar

References

Diamond, LK, Blackfan, KD, Baty, JM. Erythroblastosis fetalis and its association with universal edema of the fetus, icterus gravis neonatorum and anemia of the newborn. J Pediatr 1932;1:269309.Google Scholar
Landsteiner, K, Weiner, A. An agglutinable factor in human blood recognized by immune sera for Rhesus blood. Pro Soc Exp Biol Med 1940;43:223.Google Scholar
Mollison, PL. Methods of determining the posttransfusion survival of stored red cells. Transfusion 1984;24:93–6.Google Scholar
Bowman, JM. Treatment options for the fetus with alloimmune hemolytic disease. Transfus Med Rev 1990;4:191207.Google Scholar
Chavez, GF, Mulinare, J, Edmonds, LD. Epidemiology of Rh hemolytic disease of the newborn in the United States. JAMA 1991;265:3270–4.Google Scholar
Murphy, SL. Deaths: Final data for 1998. Natl Vital Stat Rep 2000;48:1105.Google Scholar
Liumbruno, GM, D’Alessandro, A, Rea, F, et al. The role of antenatal immunoprophylaxis in the prevention of maternal-foetal anti-Rh(D) alloimmunisation. Blood Transfus 2010;8:816.Google Scholar
Moise, KJ, Jr. Non-anti-D antibodies in red-cell alloimmunization. Eur J Obstet Gynecol Reprod Biol 2000;92:7581.Google Scholar
Bowman, JM, Pollock, JM, Penston, LE. Fetomaternal transplacental hemorrhage during pregnancy and after delivery. Vox Sang 1986;51:117–21.Google Scholar
Cohen, F, Zuelzer, WW. Mechanisms of isoimmunization. II. Transplacental passage and postnatal survival of fetal erythrocytes in heterospecific pregnancies. Blood 1967;30:796804.Google Scholar
Huchet, J, Defossez, Y, Brossard, Y. Detection of transplacental hemorrhage during the last trimester of pregnancy. Transfusion 1988;28:506.Google Scholar
Feldman, N, Skoll, A, Sibai, B. The incidence of significant fetomaternal hemorrhage in patients undergoing cesarean section. Am J Obstet Gynecol 1990;163:855–8.Google Scholar
McKenna, DS, Nagaraja, HN, O’Shaughnessy, R. Management of pregnancies complicated by anti-Kell isoimmunization. Obstet Gynecol 1999;93:667–73.Google Scholar
Howard, H, Martlew, V, McFadyen, I, et al. Consequences for fetus and neonate of maternal red cell allo-immunisation. Arch Dis Child Fetal Neonatal Ed 1998;78:F626.Google Scholar
Clarke, CA. Preventing rhesus babies: The Liverpool research and follow up. Arch Dis Child 1989;64:1734–40.Google Scholar
Doyle, B, Quigley, J, Allen, C, Fitzgerald, J. Homozygous expression of fetal red cell antigen in donor oocyte pregnancy complicated by allo-immunisation: Are current antibody thresholds to trigger increased monitoring relevant? Transfus Med 2014;24:182–3.Google Scholar
Alsaati, G, Sandler, SG. Assisted reproductive technology: An uncommon, but increasing, cause of parent–child ABO discrepancy. Transfusion 2015;55:2048–9.Google Scholar
Simister, NE, Story, CM. Human placental Fc receptors and the transmission of antibodies from mother to fetus. J Reprod Immunol 1997;37:123.Google Scholar
Palfi, M, Selbing, A. Placental transport of maternal immunoglobulin G. Am J Reprod Immunol. 1998;39:24–6.Google Scholar
Devey, ME, Voak, D. A critical study of the IgG subclasses of Rh anti-D antibodies formed in pregnancy and in immunized volunteers. Immunology 1974;27:1073–9.Google Scholar
Palfi, M, Hilden, JO, Gottvall, T, Selbing, A. Placental transport of maternal immunoglobulin G in pregnancies at risk of Rh (D) hemolytic disease of the newborn. Am J Reprod Immunol 1998;39:323–8.Google Scholar
Yamamoto, F, Clausen, H, White, T, Marken, J, Hakomori, S. Molecular genetic basis of the histo-blood group ABO system. Nature 1990;345:229–33.Google Scholar
Ozolek, JA, Watchko, JF, Mimouni, F. Prevalence and lack of clinical significance of blood group incompatibility in mothers with blood type A or B. J Pediatr 1994;125:8791.Google Scholar
Maisels, MJ, Stevenson, DK, Watchko, JF. Care of the Jaundiced Neonate (New York: McGraw Hill, 2012).Google Scholar
Kaplan, M, Hammerman, C, Vreman, HJ, Wong, RJ, Stevenson, DK. Hemolysis and hyperbilirubinemia in antiglobulin positive, direct ABO blood group heterospecific neonates. J Pediatr 2010;157:772–7.Google Scholar
Naiman, JL. Erythroblastosis Fetalis (Philadelphia: W.B. Saunders, 1982).Google Scholar
Adewuyi, JO, Gwanzura, C, Mvere, D. Characteristics of anti-A and anti-B in black Zimbabweans. Vox Sang 1994;67:307–9.Google Scholar
Adewuyi, JO, Gwanzura, C. Racial difference between white and black Zimbabweans in the haemolytic activity of A, B, O antibodies. Afr J Med Med Sci 2001;30:71–4.Google Scholar
Murray, NA, Roberts, IA. Haemolytic disease of the newborn. Arch Dis Child Fetal Neonatal Ed 2007;92:F83-88.Google Scholar
Watchko, JF. Common hematologic problems in the newborn nursery. Pediatr Clin North Am 2015;62:509–24.Google Scholar
Maisels, MJ, Watchko, JF. Routine blood typing and DAT in infants of group O mothers. J Perinatol 2013;33:579.Google Scholar
Judd, WJ. Practice guidelines for prenatal and perinatal immunohematology, revisited. Transfusion 2001;41:1445–52.Google Scholar
AAP Subcommittee on Hyperbilirubinemia. Management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation. Pediatrics 2004;114:297316.Google Scholar
Avent, ND, Reid, ME. The Rh blood group system: A review. Blood. 2000;95:375–87.Google Scholar
Westhoff, CM. The Rh blood group system in review: A new face for the next decade. Transfusion 2004;44:1663–73.Google Scholar
Avent, ND. New insight into the Rh system: Structure and function. ISBT Science Series 2007;2:3543.Google Scholar
Colin, Y, Cherif-Zahar, B, Le Van Kim, C, et al. Genetic basis of the RhD-positive and RhD-negative blood group polymorphism as determined by Southern analysis. Blood 1991;78:2747–52.Google Scholar
Avent, ND, Martin, PG, Armstrong-Fisher, SS, et al. Evidence of genetic diversity underlying Rh D-, weak D (Du), and partial D phenotypes as determined by multiplex polymerase chain reaction analysis of the RHD gene. Blood 1997;89:2568–77.Google Scholar
Daniels, G, Green, C, Smart, E. Differences between RhD-negative Africans and RhD-negative Europeans. Lancet 1997;350:862–3.Google Scholar
Okuda, H, Kawano, M, Iwamoto, S, et al. The RHD gene is highly detectable in RhD-negative Japanese donors. J Clin Invest 1997;100:373–9.Google Scholar
Singleton, BK, Green, CA, Avent, ND, et al. The presence of an RHD pseudogene containing a 37 base pair duplication and a nonsense mutation in Africans with the Rh D-negative blood group phenotype. Blood 2000;95:1218.Google Scholar
Sun, CF, Chou, CS, Lai, NC, Wang, WT. RHD gene polymorphisms among RhD-negative Chinese in Taiwan. Vox Sang 1998;75:52–7.Google Scholar
Winters, JL, Pineda, AA, Gorden, LD, et al. RBC alloantibody specificity and antigen potency in Olmsted County, Minnesota. Transfusion 2001;41:1413–20.Google Scholar
Jakobowicz, R, Williams, L, Silberman, F. Immunization of Rh-negative volunteers by repeated injections of very small amounts of Rh-positive blood. Vox Sang 1972;23:376–81.Google Scholar
Bowman, JM, Chown, B, Lewis, M, Pollock, JM. Rh isoimmunization during pregnancy: Antenatal prophylaxis. Can Med Assoc J 1978;118:623–7.Google Scholar
Tovey, LA, Townley, A, Stevenson, BJ, Taverner, J. The Yorkshire antenatal anti-D immunoglobulin trial in primigravidae. Lancet 1983;2:244–6.Google Scholar
Nevanlinna, HR, Anttinen, EE, Vainio, T. Hemolytic disease of newborn due to Rh isoimmunization: Considerations on therapy and prognosis. Duodecim 1956;72:354–69.Google Scholar
Clarke, C, Finn, R, McConnell, R, Sheppard, P. The protection afforded by ABO incompatibility against erythroblastosis due to rhesus anti-D. Int Arch Allergy Immunol 1958;13:377–81.Google Scholar
Ascari, WQ, Levine, P, Pollack, W. Incidence of maternal Rh immunization by ABO compatible and incompatible pregnancies. Br Med J 1969;1:399401.Google Scholar
Murray, S, Knox, EG, Walker, W. Rhesus haemolytic disease of the newborn and the ABO groups. Vox Sang 1965;10:631.Google Scholar
Bollason, G, Hjartardottir, H, Jonsson, T, et al. Red blood cell alloimmunization in pregnancy during the years 1996–2015 in Iceland: A nationwide population study. Transfusion 2017;57:2578–85.Google Scholar
Awowole, I, Cohen, K, Rock, J, Sparey, C. Prevalence and obstetric outcome of women with red cell antibodies in pregnancy at the Leeds Teaching Hospitals NHS Trust, West Yorkshire, England. Eur J Obstet Gynecol Reprod Biol 2019;237:8992.Google Scholar
Moran, P, Robson, SC, Reid, MM. Anti-E in pregnancy. BJOG 2000;107:1436–8.Google Scholar
McAdams, RM, Dotzler, SA, Winter, LW, Kerecman, JD. Severe hemolytic disease of the newborn from anti-e. J Perinatol 2008;28:230–2.Google Scholar
Sturgeon, P. Hematological observations on the anemia associated with blood type Rhnull. Blood 1970;36:310–20.Google Scholar
Bowell, PJ, Allen, DL, Entwistle, CC. Blood group antibody screening tests during pregnancy. Br J Obstet Gynaecol 1986;93:1038–43.Google Scholar
Filbey, D, Hanson, U, Wesstrom, G. The prevalence of red cell antibodies in pregnancy correlated to the outcome of the newborn: A 12 year study in central Sweden. Acta Obstet Gynecol Scand 1995;74:687–92.Google Scholar
Kozlowski, CL, Lee, D, Shwe, KH, Love, EM. Quantification of anti-c in haemolytic disease of the newborn. Transfus Med 1995;5:3742.Google Scholar
Wenk, RE, Goldstein, P, Felix, JK. Alloimmunization by hr’(c), hemolytic disease of newborns, and perinatal management. Obstet Gynecol 1986;67:623–6.Google Scholar
Bowell, PJ, Brown, SE, Dike, AE, Inskip, MJ. The significance of anti-c alloimmunization in pregnancy. Br J Obstet Gynaecol 1986;93:1044–8.Google Scholar
Babinszki, A, Berkowitz, RL. Haemolytic disease of the newborn caused by anti-c, anti-E and anti-Fya antibodies: Report of five cases. Prenat Diagn 1999;19:533–6.Google Scholar
Mouro, I, Colin, Y, Cherif-Zahar, B, Cartron, JP, Le Van Kim, C. Molecular genetic basis of the human Rhesus blood group system. Nat Genet 1993;5:62–5.Google Scholar
Moncharmont, P, Juron-Dupraz, F, Rigal, D, Vignal, M, Meyer, F. Haemolytic disease of two newborns in a Rhesus anti-e alloimmunized woman. Review of literature.Haematologia (Budap) 1990;23:97100.Google Scholar
Coombs, RR, Mourant, AE, Race, RR. A new test for the detection of weak and incomplete Rh agglutinins. Br J Exp Pathol 1945;26:255–66.Google Scholar
Bowman, JM, Pollock, JM, Manning, FA, Harman, CR, Menticoglou, S. Maternal Kell blood group alloimmunization. Obstet Gynecol 1992;79:239–44.Google Scholar
Babinszki, A, Lapinski, RH, Berkowitz, RL. Prognostic factors and management in pregnancies complicated with severe Kell alloimmunization: Experiences of the last 13 years. Am J Perinatol 1998;15:695701.Google Scholar
Southcott, MJ, Tanner, MJ, Anstee, DJ. The expression of human blood group antigens during erythropoiesis in a cell culture system. Blood. 1999;93:442535.Google Scholar
Weiner, CP, Widness, JA. Decreased fetal erythropoiesis and hemolysis in Kell hemolytic anemia. Am J Obstet Gynecol 1996;174:547–51.Google Scholar
Vaughan, JI, Manning, M, Warwick, RM, et al. Inhibition of erythroid progenitor cells by anti-Kell antibodies in fetal alloimmune anemia. N Engl J Med 1998;338:798803.Google Scholar
Vaughan, JI, Warwick, R, Letsky, E, et al. Erythropoietic suppression in fetal anemia because of Kell alloimmunization. Am J Obstet Gynecol 1994;171:247–52.Google Scholar
Mayne, KM, Bowell, PJ, Pratt, GA. The significance of anti-Kell sensitization in pregnancy. Clin Lab Haematol 1990;12:379–85.Google Scholar
Redman, CM, Marsh, WL. The Kell blood group system and the McLeod phenotype. Semin Hematol 1993;30:209–18.Google Scholar
Gorlin, JB, Kelly, L. Alloimmunisation via previous transfusion places female Kpb-negative recipients at risk for having children with clinically significant hemolytic disease of the newborn. Vox Sang 1994;66:46–8.Google Scholar
Lowe, RF, Musengezi, AT, Moores, P. Severe hemolytic disease of the newborn associated with anti-JSb. Transfusion 1978;18:466–8.Google Scholar
Gordon, MC, Kennedy, MS, O’Shaughnessy, RW, Waheed, A. Severe hemolytic disease of the newborn due to anti-Js(b). Vox Sang 1995;69:140–1.Google Scholar
Miller, LH, Mason, SJ, Clyde, DF, McGinniss, MH. The resistance factor to Plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy.N Engl J Med 1976;295:302–4.Google Scholar
Hadley, TJ, Peiper, SC. From malaria to chemokine receptor: The emerging physiologic role of the Duffy blood group antigen. Blood 1997;89:3077–91.Google Scholar
Horuk, R, Chitnis, CE, Darbonne, WC, et al. A receptor for the malarial parasite Plasmodium vivax: The erythrocyte chemokine receptor. Science 1993;261:1182–4.Google Scholar
Sim, BK, Chitnis, CE, Wasniowska, K, Hadley, TJ, Miller, LH. Receptor and ligand domains for invasion of erythrocytes by Plasmodium falciparum. Science 1994;264:1941–4.Google Scholar
Tournamille, C, Le Van Kim, C, Gane, P, Cartron, JP, Colin, Y. Molecular basis and PCR-DNA typing of the Fya/fyb blood group polymorphism. Hum Genet 1995;95:407–10.Google Scholar
Tournamille, C, Colin, Y, Cartron, JP, Le Van Kim C. Disruption of a GATA motif in the Duffy gene promoter abolishes erythroid gene expression in Duffy-negative individuals. Nat Genet 1995;10:224–8.Google Scholar
Chaudhuri, A, Polyakova, J, Zbrzezna, V, Pogo, AO. The coding sequence of Duffy blood group gene in humans and simians: Restriction fragment length polymorphism, antibody and malarial parasite specificities, and expression in nonerythroid tissues in Duffy-negative individuals. Blood 1995;85:615–21.Google Scholar
Weatherall, DJ. Host genetics and infectious disease. Parasitology 1996;112 Suppl:S2329.Google Scholar
Peiper, SC, Wang, ZX, Neote, K, et al. The Duffy antigen/receptor for chemokines (DARC) is expressed in endothelial cells of Duffy negative individuals who lack the erythrocyte receptor. J Exp Med 1995;181:1311–17.Google Scholar
Sosler, SD, Perkins, JT, Fong, K, Saporito, C. The prevalence of immunization to Duffy antigens in a population of known racial distribution. Transfusion 1989;29:505–7.Google Scholar
Vescio, LA, Farina, D, Rogido, M, Sola, A. Hemolytic disease of the newborn caused by anti-Fyb. Transfusion 1987;27:366.Google Scholar
Shah, VP, Gilja, BK. Hemolytic disease of newborn due to anti-Duffy (Fya). NY State J Med 1983;83:244–5.Google Scholar
Weinstein, L, Taylor, ES. Hemolytic disease of the neonate secondary to anti-Fya. Am J Obstet Gynecol 1975;121:643–5.Google Scholar
Goodrick, MJ, Hadley, AG, Poole, G. Haemolytic disease of the fetus and newborn due to anti-Fy(a) and the potential clinical value of Duffy genotyping in pregnancies at risk. Transfus Med 1997;7:301–4.Google Scholar
Mallinson, G, Soo, KS, Schall, TJ, Pisacka, M, Anstee, DJ. Mutations in the erythrocyte chemokine receptor (Duffy) gene: The molecular basis of the Fya/Fyb antigens and identification of a deletion in the Duffy gene of an apparently healthy individual with the Fy(a-b-) phenotype. Br J Haematol 1995;90:823–9.Google Scholar
Buchanan, DI, Sinclair, M, Sanger, R, Gavin, J, Teesdale, P. An Alberta Cree Indian with a rare Duffy antibody, anti-Fy 3. Vox Sang 1976;30:114–21.Google Scholar
Lawicki, S, Covin, RB, Powers, AA. The Kidd (JK) Blood Group System. Transfus Med Rev 2017;31:165–72.Google Scholar
Olives, B, Merriman, M, Bailly, P, et al. The molecular basis of the Kidd blood group polymorphism and its lack of association with type 1 diabetes susceptibility. Hum Mol Genet 1997;6:1017–20.Google Scholar
Woodfield, DG, Douglas, R, Smith, J, Simpson, A, Pinder, L, Staveley, JM. The Jk(a-b-) phenotype in New Zealand Polynesians. Transfusion 1982;22:276–8.Google Scholar
Dorner, I, Moore, JA, Chaplin, H, Jr. Combined maternal erythrocyte autosensitization and materno-fetal Jk incompatibility. Transfusion 1974;14:212–19.Google Scholar
Merlob, P, Litwin, A, Reisner, SH, Cohen, IJ, Zaizov, R. Hemolytic disease of the newborn caused by anti-Jkb. Pediatr Hematol Oncol 1987;4:357–60.Google Scholar
Thakral, B, Malhotra, S, Saluja, K, Kumar, P, Marwaha, N. Hemolytic disease of newborn due to anti-Jk b in a woman with high risk pregnancy. Transfus Apher Sci 2010;43:41–3.Google Scholar
Pierce, SR, Hardman, JT, Steele, S, Beck, ML. Hemolytic disease of the newborn associated with anti-Jk3. Transfusion 1980;20:189–91.Google Scholar
Bruce, LJ, Pan, RJ, Cope, DL, et al. Altered structure and anion transport properties of band 3 (AE1, SLC4A1) in human red cells lacking glycophorin A. J Biol Chem 2004;279:2414–40.Google Scholar
Tomita, M, Furthmayr, H, Marchesi, VT. Primary structure of human erythrocyte glycophorin A. Isolation and characterization of peptides and complete amino acid sequence. Biochemistry 1978;17:4756–70.Google Scholar
Geifman-Holtzman, O, Wojtowycz, M, Kosmas, E, Artal, R. Female alloimmunization with antibodies known to cause hemolytic disease. Obstet Gynecol 1997;89:272–5.Google Scholar
Pal, M, Williams, B. Prevalence of maternal red cell alloimmunisation: A population study from Queensland, Australia. Pathology 2015;47:151–5.Google Scholar
Yasuda, H, Ohto, H, Nollet, KE, et al. Hemolytic disease of the fetus and newborn with late-onset anemia due to anti-M: A case report and review of the Japanese literature. Transfus Med Rev 2014;28:16.Google Scholar
De Young-Owens, A, Kennedy, M, Rose, RL, Boyle, J, O’Shaughnessy, R. Anti-M isoimmunization: Management and outcome at the Ohio State University from 1969 to 1995. Obstet Gynecol 1997;90:962–6.Google Scholar
Duguid, JK, Bromilow, IM, Entwistle, GD, Wilkinson, R. Haemolytic disease of the newborn due to anti-M. Vox Sang 1995;68:195–6.Google Scholar
Mayne, KM, Bowell, PJ, Green, SJ, Entwistle, CC. The significance of anti-S sensitization in pregnancy. Clin Lab Haematol 1990;12:105–7.Google Scholar
Smith, G, Knott, P, Rissik, J, de la Fuente, J, Win, N. Anti-U and haemolytic disease of the fetus and newborn. Br J Obstet Gynaecol 1998;105:1318–21.Google Scholar
Novaretti, MC, Jens, E, Pagliarini, T, et al. Hemolytic disease of the newborn due to anti-U.Rev Hosp Clin Fac Med Sao Paulo 2003;58:320–3.Google Scholar
Zipursky, A, Paul, VK. The global burden of Rh disease. Arch Dis Child Fetal Neonatal Ed 2011;96:F8485.Google Scholar
Clarke, CA, Mollison, PL. Deaths from Rh haemolytic disease of the fetus and newborn, 1977–87. J R Coll Physicians Lond 1989;23:181–4.Google Scholar
McMaster Conference on Prevention of Rh Immunization 28–30 September, 1977. Vox Sang 1979;36:5064.Google Scholar
Joseph, KS, Kramer, MS. The decline in Rh hemolytic disease: Should Rh prophylaxis get all the credit? Am J Public Health 1998;88:209–15.Google Scholar
Trolle, B. Prenatal Rh-immune prophylaxis with 300 micrograms immune globulin anti-D in the 28th week of pregnancy. Acta Obstet Gynecol Scand 1989;68:45–7.Google Scholar
Queenan, JT, Gadow, EC, Lopes, AC. Role of spontaneous abortion in Rh immunization. Am J Obstet Gynecol 1971;110:128–30.Google Scholar
Queenan, JT, Kubarych, SF, Shah, S, Holland, B. Role of induced abortion in rhesus immunisation. Lancet 1971;1:815–17.Google Scholar
Tabsh, KM, Lebherz, TB, Crandall, BF. Risks of prophylactic anti-D immunoglobulin after second-trimester amniocentesis. Am J Obstet Gynecol 1984;149:225–6.Google Scholar
Brandenburg, H, Jahoda, MG, Pijpers, L, Wladimiroff, JW. Rhesus sensitization after midtrimester genetic amniocentesis. Am J Med Genet 1989;32:225–6.Google Scholar
Daffos, F, Capella-Pavlovsky, M, Forestier, F. Fetal blood sampling during pregnancy with use of a needle guided by ultrasound: A study of 606 consecutive cases. Am J Obstet Gynecol 1985;153:655–60.Google Scholar
Bowman, JM, Pollock, JM, Peterson, LE, et al. Fetomaternal hemorrhage following funipuncture: Increase in severity of maternal red-cell alloimmunization. Obstet Gynecol 1994;84:839–43.Google Scholar
Blakemore, KJ, Baumgarten, A, Schoenfeld-Dimaio, M, et al. Rise in maternal serum alpha-fetoprotein concentration after chorionic villus sampling and the possibility of isoimmunization. Am J Obstet Gynecol 1986;155:988–93.Google Scholar
Jansen, MW, Brandenburg, H, Wildschut, HI, et al. The effect of chorionic villus sampling on the number of fetal cells isolated from maternal blood and on maternal serum alpha-fetoprotein levels. Prenat Diagn 1997;17:953–9.Google Scholar
Rose, PG, Strohm, PL, Zuspan, FP. Fetomaternal hemorrhage following trauma. Am J Obstet Gynecol 1985;153:844–7.Google Scholar
Boucher, M, Marquette, GP, Varin, J, Champagne, J, Bujold, E. Fetomaternal hemorrhage during external cephalic version. Obstet Gynecol 2008;112:7984.Google Scholar
An assessment of the hazards of amniocentesis. Report to the Medical Research Council by their Working Party on Amniocentesis. Br J Obstet Gynaecol 1978;85 Suppl 2:141.Google Scholar
Simonovits, I, Timar, I, Bajtai, G. Rate of Rh immunization after induced abortion. Vox Sang 1980;38:161–4.Google Scholar
USPST Task Force. Screening for D (Rh) Incompatibility. In USPST Task Force Guide to Clinical Preventive Services 2nd ed. (Washington, DC: Office of Disease Prevention and Health Promotion, 1996).Google Scholar
Committee on Practice Bulletins-Obstetrics. Practice Bulletin No. 181: Prevention of Rh D alloimmunization. Obstet Gynecol 2017;130:e57e70.Google Scholar
Moise KJ, Jr. , Gandhi, M, Boring, NH, et al. Circulating cell-free DNA to determine the fetal RHD status in all three trimesters of pregnancy. Obstet Gynecol 2016;128:1340–6.Google Scholar
Vivanti, A, Benachi, A, Huchet, FX, et al. Diagnostic accuracy of fetal rhesus D genotyping using cell-free fetal DNA during the first trimester of pregnancy. Am J Obstet Gynecol 2016;215:606e601–606 e605.Google Scholar
de Haas, M, Thurik, FF, van der Ploeg, CP, et al. Sensitivity of fetal RHD screening for safe guidance of targeted anti-D immunoglobulin prophylaxis: Prospective cohort study of a nationwide programme in the Netherlands. BMJ 2016;355:i5789.Google Scholar
Sandler, SG, Flegel, WA, Westhoff, CM, et al. It’s time to phase in RHD genotyping for patients with a serologic weak D phenotype. College of American Pathologists Transfusion Medicine Resource Committee Work Group.Transfusion 2015;55:680–9.Google Scholar
Liley, AW. Liquor amnil analysis in the management of the pregnancy complicated by resus sensitization. Am J Obstet Gynecol 1961;82:1359–70.Google Scholar
Queenan, JT, Tomai, TP, Ural, SH, King, JC. Deviation in amniotic fluid optical density at a wavelength of 450 nm in Rh-immunized pregnancies from 14 to 40 weeks’ gestation: A proposal for clinical management. Am J Obstet Gynecol 1993;168:1370–6.Google Scholar
Mari, G, Deter, RL, Carpenter, RL, et al. Noninvasive diagnosis by Doppler ultrasonography of fetal anemia due to maternal red-cell alloimmunization. Collaborative Group for Doppler Assessment of the Blood Velocity in Anemic Fetuses. N Engl J Med 2000;342:914.Google Scholar
Zwiers, C, van Kamp, I, Oepkes, D, Lopriore, E. Intrauterine transfusion and non-invasive treatment options for hemolytic disease of the fetus and newborn: Review on current management and outcome. Expert Rev Hematol 2017;10:337–44.Google Scholar
Lindenburg, IT, Smits-Wintjens, VE, van Klink, JM, et al. Long-term neurodevelopmental outcome after intrauterine transfusion for hemolytic disease of the fetus/newborn: The LOTUS study. Am J Obstet Gynecol 2012;206:141 e141-148.Google Scholar
Watson, WJ, Wax, JR, Miller, RC, Brost, BC. Prevalence of new maternal alloantibodies after intrauterine transfusion for severe Rhesus disease. Am J Perinatol 2006;23:189–92.Google Scholar
Nicolaides, KH, Soothill, PW, Clewell, WH, et al. Fetal haemoglobin measurement in the assessment of red cell isoimmunisation. Lancet 1988;1:1073–5.Google Scholar
Scheier, M, Hernandez-Andrade, E, Fonseca, EB, Nicolaides, KH. Prediction of severe fetal anemia in red blood cell alloimmunization after previous intrauterine transfusions. Am J Obstet Gynecol 2006;195:1550–6.Google Scholar
Papantoniou, N, Sifakis, S, Antsaklis, A. Therapeutic management of fetal anemia: Review of standard practice and alternative treatment options. J Perinat Med 2013;41:7182.Google Scholar
Schumacher, B, Moise, KJ, Jr. Fetal transfusion for red blood cell alloimmunization in pregnancy. Obstet Gynecol 1996;88:137–50.Google Scholar
Lindenburg, IT, van Kamp, IL, van Zwet, EW, et al. Increased perinatal loss after intrauterine transfusion for alloimmune anaemia before 20 weeks of gestation. BJOG 2013;120:847–52.Google Scholar
Vats, K, Watchko, JF. Coordinating care across the perinatal continuum in hemolytic disease of the fetus and newborn: The timely handoff of a positive maternal anti-erythrocyte antibody Screen. J Pediatr 2019;214:212–16.Google Scholar
Goplerud, CP, White, CA, Bradbury, JT, Briggs, TL. The first Rh-isoimmunized pregnancy. Am J Obstet Gynecol 1973;115:632–8.Google Scholar
Herschel, M, Karrison, T, Wen, M, Caldarelli, L, Baron, B. Isoimmunization is unlikely to be the cause of hemolysis in ABO-incompatible but direct antiglobulin test-negative neonates. Pediatrics 2002;110:127–30.Google Scholar
Cashore, WJ. Neonatal hyperbilirubinemia. In McMillan, JA, Feigin, RD, DeAngelis, CD, Jones, MDJ, eds. Oski’s Pediatrics 4th ed. (Philadelphia, PA: Lippincott, Williams & Wilkins; 2006), pp. 235–45.Google Scholar
Christensen, RD, Agarwal, AM, George, TI, Bhutani, VK, Yaish, HM. Acute neonatal bilirubin encephalopathy in the State of Utah 2009–2018. Blood Cells Mol Dis 2018;72:1013.Google Scholar
American Academy of Pediatrics Subcommittee on Hyperbilirubinemia. Management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation [published correction appears in Pediatrics 2004;114(4):1138]. Pediatrics 2004;114(1):297316.Google Scholar
Alcock, GS, Liley, H. Immunoglobulin infusion for isoimmune haemolytic jaundice in neonates. Cochrane Database of Systematic Reviews 2002;3:CD003313.Google Scholar
Zwiers, C, Scheffer-Rath, MEA, Lopriore, E, de Haas, M, Liley, HG. Immunoglobulin for alloimmune hemolytic disease in neonates. Cochrane Database of Systematic Reviews 2018;3:CD003313.Google Scholar
Watchko, JF. Emergency release uncross-matched packed red blood cells for immediate double volume exchange transfusion in neonates with intermediate to advanced acute bilirubin encephalopathy: Timely but insufficient? J Perinatol 2018;38:947–53.Google Scholar
Blackall, DP, Pesek, GD, Montgomery, MM, et al. Hemolytic disease of the fetus and newborn due to anti-Ge3: Combined antibody-dependent hemolysis and erythroid precursor cell growth inhibition. Am J Perinatol 2008;25:541–5.Google Scholar
Dorn, I, Schlenke, P, Hartel, C. Prolonged anemia in an intrauterine-transfused neonate with Rh-hemolytic disease: No evidence for anti-D-related suppression of erythropoiesis in vitro. Transfusion 2010;50:1064–70.Google Scholar
Rath, ME, Smits-Wintjens, VE, Lindenburg, IT, et al. Exchange transfusions and top-up transfusions in neonates with Kell haemolytic disease compared to Rh D haemolytic disease. Vox Sang 2011;100:312–16.Google Scholar
Bhutani, VK, Zipursky, A, Blencowe, H, et al. Neonatal hyperbilirubinemia and Rhesus disease of the newborn: Incidence and impairment estimates for 2010 at regional and global levels. Pediatr Res 2013;74 Suppl 1:86100.Google Scholar
Osaro, E, Charles, AT. Rh isoimmunization in sub-Saharan Africa indicates need for universal access to anti-RhD immunoglobulin and effective management of D-negative pregnancies. Int J Womens Health 2010;2:429–37.Google Scholar
Kancherla, V, Oakley, GP, Jr., Brent, RL. Urgent global opportunities to prevent birth defects. Semin Fetal Neonatal Med 2014;19:153–60.Google Scholar
Waldron, P, de Alarcón, P. ABO hemolytic disease of the newborn: A unique constellation of findings in siblings and review of protective mechanisms in the fetal-maternal system. Am J Perinatol 1999;16:391–8.Google Scholar
Sherer, DM, Abramowicz, JS, Ryan, RM, et al. Severe fetal hydrops resulting from ABO incompatibility. Obstet Gynecol 1991;78:897–9.Google Scholar
Lacey, PA, Caskey, CR, Werner, DJ, Moulds, JJ. Fatal hemolytic disease of a newborn due to anti-D in an Rh-positive Du variant mother. Transfusion 1983;23:91–4.Google Scholar
Bowman, JM, Pollock, JM, Manning, FA, Harman, CR. Severe anti-C hemolytic disease of the newborn. Am J Obstet Gynecol 1992;166:1239–43.Google Scholar
Macher, S, Wagner, T, Rosskopf, K, et al. Severe case of fetal hemolytic disease caused by anti-C(w) requiring serial intrauterine transfusions complicated by pancytopenia and cholestasis. Transfusion 2016;56:80–3.Google Scholar
Malik, S, Moiz, B. Clinical significance of maternal anti-Cw antibodies: A review of three cases and literature. J Pak Med Assoc 2012;62:620–1.Google Scholar
Grobel, RK, Cardy, JD. Hemolytic disease of the newborn due to anti-EW. A fourth example of the Rh antigen, EW. Transfusion 1971;11:77–8.Google Scholar
Jakobowicz, R, Whittingham, S, Barrie, JU, Simmons, RT. A further investigation on polyvalent anti-C (rh’) and anti-G (rhg) antibodies produced by iso-immunization in pregnancy. Med J Aust 1962;49(1):896–7.Google Scholar
Li, BJ, Jiang, YJ, Yuan, F, Ye, HX. Exchange transfusion of least incompatible blood for severe hemolytic disease of the newborn due to anti-Rh17. Transfus Med 2010;20:66–9.Google Scholar
Vaughan, JI, Warwick, R, Welch, CR, Letsky, EA. Anti-Kell in pregnancy. Br J Obstet Gynaecol 1991;98:944–5.Google Scholar
Moncharmont, P, Juron-Dupraz, F, Doillon, M, Vignal, M, Debeaux, P. A case of hemolytic disease of the newborn infant due to anti-K (Cellano). Acta Haematol 1991;85:45–6.Google Scholar
Bowman, JM, Harman, FA, Manning, CR, Pollock, JM. Erythroblastosis fetalis produced by anti-k. Vox Sang 1989;56:187–9.Google Scholar
Duguid, JKM, Bromilow, IM. Haemolytic disease of the newborn due to anti-k. Vox Sang 1990;58:6972.Google Scholar
Moulds, JM, Persa, R, Rierson, D, et al. Three novel alleles in the Kell blood group system resulting in the Knull phenotype and the first in a Native American. Transfusion 2013;53:2867–71.Google Scholar
Jovanovic-Srzentic, S, Djokic, M, Tijanic, N, et al. Antibodies detected in samples from 21,730 pregnant women. Immunohematology 2003;19:8992.Google Scholar
Mittal, K, Sood, T, Bansal, N, et al. Clinical significance of rare maternal anti Jk(a) antibody. Indian J Hematol Blood Transfus. 2016;32:497–9.Google Scholar
Ishida, A, Ohto, H, Yasuda, H, et al. Anti-M antibody induced prolonged anemia following hemolytic disease of the newborn due to erythropoietic suppression in 2 siblings. J Pediatr Hematol Oncol 2015;37:e375377.Google Scholar
Wu, KH, Chu, SL, Chang, JG, Shih, MC, Peng, CT. Haemolytic disease of the newborn due to maternal irregular antibodies in the Chinese population in Taiwan. Transfus Med 2003;13:311–14.Google Scholar
Telischi, M, Behzad, O, Issitt, PD, Pavone, BG. Hemolytic disease of the newborn due to anti-N. Vox Sang 1976;31:109–16.Google Scholar
Moncharmont, P, Buclet, D, Trouilloud, C, Peyrard, T, Rigal, D. Severe hemolytic disease of the fetus and the newborn associated with anti-Vw (Vw). J Matern Fetal Neonatal Med 2010;23:1066–8.Google Scholar
Field, TE, Wilson, TE, Dawes, BJ, Giles, CM. Haemolytic disease of the newborn due to anti-Mt a. Vox Sang 1972;22:432–7.Google Scholar
Cheung, CC, Challis, D, Fisher, G, et al. Anti-Mta associated with three cases of hemolytic disease of the newborn. Immunohematology 2002;18:37–9.Google Scholar
Wu, KH, Chang, JG, Lin, M, et al. Hydrops foetalis caused by anti-Mur in first pregnancy–a case report. Transfus Med 2002;12:325–7.Google Scholar
Brooks, S, Squires, JE. Hemolytic disease of the fetus and newborn caused by anti-Lan. Transfusion. 2014;54:1317–20.Google Scholar
Davies, J, Day, S, Milne, A, Roy, A, Simpson, S. Haemolytic disease of the foetus and newborn caused by auto anti-LW. Transfus Med 2009;19:218–19.Google Scholar
Francis, BJ, Hatcher, DE. Hemolytic disease of the newborn apparently caused by anti-Lu-a. Transfusion 1961;1:248–50.Google Scholar
Scheffer, H, Tamaki, HT. Anti-Lu-b and mild hemolytic disease of the newborn: A case report. Transfusion 1966;6:497–8.Google Scholar
Dube, VE, Zoes, CS. Subclinical hemolytic disease of the newborn associated with IgG anti-Lub. Transfusion 1982;22:251–3.Google Scholar
Abhyankar, S, Silfen, S, Rao, SP, Vinciguerra, C, Dimiaio, TM. Positive cord blood “DAT” due to anti-Le(a): Absence of hemolytic disease of the newborn. Am J Pediatr Hematol Oncol 1989;11:184–5.Google Scholar
Carreras Vescio, LA, Torres, OW, Virgilio, OS, Pizzolato, M. Mild hemolytic disease of the newborn due to anti-Lewis(a). Vox Sang 1993;64:194–5.Google Scholar
Bharucha, ZS, Joshi, SR, Bhatia, HM. Hemolytic disease of the newborn due to anti-Le. Vox Sang 1981;41:36–9.Google Scholar
Reid, ME, Ellisor, SS. Hemolytic disease of newborn due to anti-Leb. Vox Sang 1982;42:278.Google Scholar
Joshi, SR, Wagner, FF, Vasantha, K, Panjwani, SR, Flegel, WA. An AQP1 null allele in an Indian woman with Co(a-b-) phenotype and high-titer anti-Co3 associated with mild HDN. Transfusion 2001;41:1273–8.Google Scholar
Sacks, DA, Johnson, CS, Platt, LD. Isoimmunization in pregnancy to Gerbich antigen. Am J Perinatol 1985;2:208–10.Google Scholar
Pate, LL, Myers, JC, Palma, JP, et al. Anti-Ge3 causes late-onset hemolytic disease of the newborn: The fourth case in three Hispanic families. Transfusion 2013;53:2152–7.Google Scholar
DeMarco, M, Uhl, L, Fields, L, et al. Hemolytic disease of the newborn due to the Scianna antibody, anti-Sc2. Transfusion 1995;35:5860.Google Scholar
Rauch, S, Ritgen, J, Wisskirchen, M, et al. A case of anti-Rd causing fetal anemia. Transfusion 2017;57:1485–7.Google Scholar
Nakajima, H, Ito, K. An example of anti-Jra causing hemolytic disease of the newborn and frequency of Jra antigen in the Japanese population. Vox Sang 1978;35:265–7.Google Scholar
Masumoto, A, Masuyama, H, Sumida, Y, Segawa, T, Hiramatsu, Y. Successful management of anti-Jra alloimmunization in pregnancy: A case report. Gynecol Obstet Invest 2010;69:81–3.Google Scholar
Takabayashi, T, Murakami, M, Yajima, H, et al. Influence of maternal antibody anti-Jra on the baby: A case report and pedigree chart. Tohoku J Exp Med 1985;145:97101.Google Scholar
Ishihara, Y, Miyata, S, Chiba, Y, Kawai, T. Successful treatment of extremely severe fetal anemia due to anti-Jra alloimmunization. Fetal Diagn Ther 2006;21:269–71.Google Scholar
Braschler, T, Vokt, CA, Hustinx, H, et al. Management of a pregnant woman with anti-holley alloantibody. Transfus Med Hemother 2015;42:129–30.Google Scholar
Ferguson, SJ, Boyce, F, Blajchman, MA. Anti-Ytb in pregnancy. Transfusion 1979;19:581–2.Google Scholar
Moulds, MK. Serological investigation and clinical significance of high-titer, low-avidity (HTLA) antibodies. Am J Med Technol 1981;47:789–95.Google Scholar
Maayan-Metzger, A, Schwartz, T, Sulkes, J, Merlob, P. Maternal anti-D prophylaxis during pregnancy does not cause neonatal haemolysis. Arch Dis Child Fetal Neonatal Ed 2001;84:F6062.Google Scholar
Cohen, DN, Johnson, MS, Liang, WH, McDaniel, HL, Young, PP. Clinically significant hemolytic disease of the newborn secondary to passive transfer of anti-D from maternal RhIG. Transfusion 2014;54:2863–6.Google Scholar

References

Pearson, HA. Life-span of the fetal red blood cell. J Pediatr 1967;70(2):166–71.Google Scholar
Oski, FA, Naiman, JL. Hematologic Problems in the Newborn, 3rd ed. (Philadelphia, PA: W. B. Saunders, 1982), pp. 1360.Google Scholar
Matovcik, LM, Mentzer, WC. The membrane of the human neonatal red cell. Clin Haematol 1985;14(1):203–21.Google Scholar
Matovcik, LM, Chiu, D, Lubin, B, et al. The aging process of human neonatal erythrocytes. Pediatr Res 1986;20(11):1091–6.Google Scholar
Advani, R, Mentzer, W, Andrews, D, Schrier, S. Oxidation of hemoglobin F is associated with the aging process of neonatal red blood cells. Pediatr Res 1992;32(2):165–8.Google Scholar
Widness, JA, Kuruvilla, DJ, Mock, DM, et al. autologous infant and allogeneic adult red cells demonstrate similar concurrent post-transfusion survival in very low birth weight neonates. J Pediatr 2015;167(5):1001–6.Google Scholar
Kuruvilla, DJ, Widness, JA, Nalbant, D, et al. Estimation of adult and neonatal RBC lifespans in anemic neonates using RBCs labeled at several discrete biotin densities. Pediatr Res 2017;81(6):905–10.Google Scholar
Geaghan, SM. Hematologic values and appearances in the healthy fetus, neonate, and child. Clin Lab Med 1999;19(1):137, v.Google Scholar
Holroyde, CP, Oski, FA, Gardner, FH. The pocked erythrocyte red-cell surface alterations in reticuloendothelial immaturity of the neonate. N Engl J Med 1969;281(10):516–20.Google Scholar
Padmanabhan, J, Risemberg, HM, Rowe, RD Howell-Jolly bodies in the peripheral blood of full-term and premature neonates. Johns Hopkins Med J 1973;132(3):146–50.Google Scholar
Maisels, MJ, Pathak, ANelson, NM, Nathan, DG, Smith, CA. Endogenous production of carbon monoxide in normal and erythroblastotic newborn infants. J Clin Invest 1971;50(1):18.Google Scholar
Arias, IM. The pathogenesis of physiologic jaundice of the newborn: A reevaluation. Birth Defects Orig Artic Ser 1970;6(2):55–9.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(1):614.Google Scholar
Necheles, TF, Rai, US, Valaes, T. The role of haemolysis in neonatal hyperbilirubinaemia as reflected in carboxyhaemoglobin levels. Acta Paediatr Scand 1976;65(3):361–7.Google Scholar
Coburn, RF, Williams, WJ, Kahn, SB. Endogenous carbon monoxide production in patients with hemolytic anemia. J Clin Invest 1966;45(4):460–8.Google Scholar
Coburn, RF. Endogenous carbon monoxide production. N Engl J Med 1970;282(4):207–9.Google Scholar
Smith, DW, Inguillo, D, Martin, D, et al. Use of noninvasive tests to predict significant jaundice in full-term infants: Preliminary studies. Pediatrics 1985;75(2):278–80.Google Scholar
Stevenson, DK, Vreman, HJ. Carbon monoxide and bilirubin production in neonates. Pediatrics 1997;100(2 Pt 1):252–4.Google Scholar
Salmi, TT Haptoglobin levels in the plasma of newborn infants with special reference to infections. Acta Paediatr Scand Suppl 1973;241:155.Google Scholar
Freda, VJ, Gorman, JG, Pollack, W, Bowe, E. Prevention of Rh hemolytic disease–ten years’ clinical experience with Rh immune globulin. N Engl J Med 1975;292(19):1014–16.Google Scholar
Baumann, R, Rubin, H. Autoimmune hemolytic anemia during pregnancy with hemolytic disease in the newborn. Blood 1973;41(2):293–7.Google Scholar
Hocking, DR. Neonatal haemolytic disease due to dapsone. Med J Aust 1968;1(26):1130–1.Google Scholar
Durocher, JR, Payne, RC, Conrad, ME. Role of sialic acid in erythrocyte survival. Blood 1975;45(1):1120.Google Scholar
Batton, DG, Amanullah, A, Comstock, C. Fetal schistocytic hemolytic anemia and umbilical vein varix. J Pediatr Hematol Oncol 2000;22(3):259–61.Google Scholar
Oski, FA, Barness, LA. Vitamin E deficiency:a previously unrecognized cause of hemolytic anemia in the premature infant. J Pediatr 1967;70(2):211–20.Google Scholar
Ritchie, JH, Fish, MB, McMasters, V, Grossman, M. Edema and hemolytic anemia in premature infants A vitamin E deficiency syndrome. N Engl J Med 1968;279(22):1185–90.Google Scholar
Gomez-Pomar, E, Hatfield, E, Garlitz, K, Westgate, PM, Bada, HS. Vitamin E in the preterm infant: A forgotten cause of hemolytic anemia. Am J Perinatol 2018;35(3):305–10.Google Scholar
Smith, H. Normal Values and Appearances: Diagnosis in Paediatric Haematology (New York: Churchill Livingstone, 1996) p. 338.Google Scholar
Tuffy, P, Brown, AK, Zuelzer, WW. Infantile pyknocytosis: A common erythrocyte abnormality of the first trimester. AMA J Dis Child 1959;98(2):227–41.Google Scholar
Zannos-Mariolea, L, Kattamis, C, Paidoucis, M. Infantile pyknocytosis and glucose-6-phosphate dehydrogenase deficiency. Br J Haematol 1962;8:258–65.Google Scholar
Keimowitz, R, Desforges, JF. Infantile pyknocytosis. N Engl J Med 1965;273(21):1152–4.Google Scholar
Ackerman, BD. Infantile pyknocytosis in Mexican-American infants. Am J Dis Child 1969;117(4):417–23.Google Scholar
Dabbous, IA, El Bahlawan, L. Infantile pyknocytosis: A forgotten or a dead diagnosis? J Pediatr Hematol Oncol 2002;24(6):507.Google Scholar
Eyssette-Guerreau, S, Bader-Meunier, B, Garcon, L, Guitton, C, Cynober, T. Infantile pyknocytosis: A cause of haemolytic anaemia of the newborn. Br J Haematol 2006; 133(4):439–2.Google Scholar
Dahoui, HA, Abboud, MR, Saab, R, et al. Familial infantile pyknocytosis in association with pulmonary hypertension. Pediatr Blood Cancer 2008;51(2):290–2.Google Scholar
Kraus, D, Yacobovich, J, Hoffer, V, et al. Infantile pyknocytosis: A rare form of neonatal anemia. Isr Med Assoc J 2010;12(3):188–9.Google Scholar
Vos, MJ, Martens, D, van de Leur, SJ, van Wijk, R. Neonatal hemolytic anemia due to pyknocytosis. Eur J Pediatr 2014;173(12):1711–14.Google Scholar
Rees, C, Lund, K, Bain, BJ. Infantile pyknocytosis. Am J Hematol 2019;94(4):489–90.Google Scholar
Gallagher, PG. Abnormalities of the erythrocyte membrane. Pediatr Clin North Am 2013;60(6):1349–62.Google Scholar
Gallagher, PG, Glader, B. Hereditary spherocytosis, hereditary elliptocytosis, and other disorders associated with abnormalities of the erythrocyte membrane. In Greer, JP, Rodgers, GM, Glader, B, et al., eds. Wintrobe’s Clinical Hematology (Philadelphia PA: Wolters Kluwer/Lippincott Williams & Wilkins, 2019), pp. 720–41.Google Scholar
Morton, NE, Mackinney, AA, Kosower, N, Schilling, RF, Gray, MP. Genetics of spherocytosis. Am J Hum Genet 1962;14:170–84.Google Scholar
Perrotta, S, Gallagher, PG, Mohandas, N . Hereditary spherocytosis. Lancet 2008;372(9647):1411–26.Google Scholar
Agre, P, Asimos, A, Casella, JF McMillan, C. Inheritance pattern and clinical response to splenectomy as a reflection of erythrocyte spectrin deficiency in hereditary spherocytosis. N Engl J Med 1986;315(25):1579–83.Google Scholar
Eber, SW, Pekrun, A, Neufeldt, A, Schroter, W. Prevalence of increased osmotic fragility of erythrocytes in German blood donors: Screening using a modified glycerol lysis test. Ann Hematol 1992;64(2):8892.Google Scholar
Palek, J, Jarolim, P. Clinical expression and laboratory detection of red blood cell membrane protein mutations. Semin Hematol 1993;30(4):249–83.Google Scholar
Stamey, CC, Diamond, LK. Congenital hemolytic anemia in the newborn: Relationship to kernicterus. AMA J Dis Child 1957;94(6):616–22.Google Scholar
Trucco, JI, Brown, AK. Neonatal manifestations of hereditary spherocytosis. Am J Dis Child 1967;113(2):263–70.Google Scholar
Rubins, J, Young, LE. Hereditary spherocytosis and glucose-6-phosphate dehydrogenase deficiency. JAMA 1977;237(8):797–8.Google Scholar
49. Christensen, RD, Yaish, HM, Gallagher, PG. A pediatrician’s practical guide to diagnosing and treating hereditary spherocytosis in neonates. Pediatrics 2015;135(6):1107–14.Google Scholar
King, MJ, Behrens, J, Rogers, C, et al. Rapid flow cytometric test for the diagnosis of membrane cytoskeleton-associated haemolytic anaemia. Br J Haematol 2000;111(3):924–33.Google Scholar
Christensen, RD, Agarwal, AM, Nussenzveig, RH, et al. Evaluating eosin-5-maleimide binding as a diagnostic test for hereditary spherocytosis in newborn infants. J Perinatol 2015;35(5):357–61.Google Scholar
Agarwal, AM, Nussenzveig, RH, Reading, NS, et al. Clinical utility of next-generation sequencing in the diagnosis of hereditary haemolytic anaemias. Br J Haematol 2016;174(5):806–14.Google Scholar
Andolfo, I, Russo, R, Gambale, A, Iolascon, A. New insights on hereditary erythrocyte membrane defects. Haematologica 2016;101(11):1284–94.Google Scholar
Iolascon, A, Faienza, MF, Moretti, A, Perrotta, S, Miraglia del Giudice, E. UGT1 promoter polymorphism accounts for increased neonatal appearance of hereditary spherocytosis. Blood 1998;91(3):1093.Google Scholar
Gallagher, PG, Petruzzi, MJ, Weed, SA, et al. Mutation of a highly conserved residue of betaI spectrin associated with fatal and near-fatal neonatal hemolytic anemia. J Clin Invest 1997;99(2):267277.Google Scholar
Delhommeau, F, Cynober, T, Schischmanoff, PO, et al. Natural history of hereditary spherocytosis during the first year of life. Blood 2000;95(2):393–7.Google Scholar
Diamond, LK. Splenectomy in childhood and the hazard of overwhelming infection. Pediatrics 1969;43(5):886–9.Google Scholar
Tracy, ET, Rice, HE. Partial splenectomy for hereditary spherocytosis. Pediatr Clin North Am 2008;55(2):503–19, x.Google Scholar
Niss, O, Chonat, S, Dagaonkar, N, et al. Genotype-phenotype correlations in hereditary elliptocytosis and hereditary pyropoikilocytosis. Blood Cells Mol Dis 2016;61:49.Google Scholar
Austin, RF, Desforges, JF. Hereditary elliptocytosis: An unusual presentation of hemolysis in the newborn associated with transient morphologic abnormalities. Pediatrics 1969;44(2):196200.Google Scholar
MacDougall, LG, Moodley, G, Quirk, M. The pyropoikilocytosis-elliptocytosis syndrome in a black South African infant: Clinical and hematological features. Am J Pediatr Hematol Oncol 1982;4(3):344–9.Google Scholar
Mentzer, WC, Jr, Iarocci, TA, Mohandas, N, et al. Modulation of erythrocyte membrane mechanical stability by 2,3-diphosphoglycerate in the neonatal poikilocytosis/elliptocytosis syndrome. J Clin Invest 1987;79(3):943–9.Google Scholar
Luzzatto, L, Nannelli, C, Notaro, R. Glucose-6-phosphate dehydrogenase deficiency. Hematol Oncol Clin North Am 2016;30(2):373–93.Google Scholar
Grace, RF, Glader, B. Red blood cell enzyme disorders. Pediatr Clin North Am 2018;65(3):579–95.Google Scholar
Glader, B, Grace, RF. Hereditary hemolytic anemias due to red blood cell enzyme disorders. In Greer, JP, Rodgers, GM, Glader, B, et al., eds. Wintrobe’s Clinical Hematology (Philadelphia PA: Wolters Kluwer/Lippincott Williams & Wilkins, 2019), pp. 742–61.Google Scholar
Glader, B. Hereditary hemolytic anemias due to red blood cell enzyme disorders. In Greer, JP, Foerster, J, Lukens, JN, et al., eds. Wintrobe’s Clinical Hematology (Philadelphia PA: Wolters Kluwer/Lippincott Williams & Wilkins Health, 2003).Google Scholar
Cappellini, MD, Fiorelli, G Glucose-6-phosphate dehydrogenase deficiency. Lancet 2008;371(9606):6474.Google Scholar
WHO Working Group. Glucose-6-phosphate dehydrogenase deficiency. Bull World Health Organ 1989;67:601–11.Google Scholar
Beutler, E. The molecular biology of enzymes of erythrocyte metabolism. In Stamatoyannopoulos, G, ed. The Molecular Basis of Blood Diseases (Philadelphia PA: W.B. Saunders, 2001).Google Scholar
Jiang, W, Yu, G, Liu, P, et al. Structure and function of glucose-6-phosphate dehydrogenase-deficient variants in Chinese population. Hum Genet 2006;119(5):463–78.Google Scholar
Valaes, T. Severe neonatal jaundice associated with glucose-6-phosphate dehydrogenase deficiency: Pathogenesis and global epidemiology. Acta Paediatr Suppl 1994;394:5876.Google Scholar
Kaplan, M, Algur, N, Hammerman, C. Onset of jaundice in glucose-6-phosphate dehydrogenase-deficient neonates. Pediatrics 2001;108(4):9569.Google Scholar
Johnson, L, Bhutani, VK, Karp, K, Sivieri, EM, Shapiro, SM. Clinical report from the pilot USA Kernicterus Registry (1992 to 2004). J Perinatol 2009;29(Suppl 1):S2545.Google Scholar
Bienzle, U, Effiong, C, Luzzatto, L. Erythrocyte glucose 6-phosphate dehydrogenase deficiency (G6PD type A–) and neonatal jaundice. Acta Paediatr Scand 1976;65(6):701–3.Google Scholar
Kaplan, M, Hammerman, C, Feldman, R, Brisk, R. Predischarge bilirubin screening in glucose-6-phosphate dehydrogenase-deficient neonates. Pediatrics 2000;105(3 Pt 1):533–7.Google Scholar
Kaplan, M, Hammerman, C, Beutler, E. Hyperbilirubinaemia, glucose-6-phosphate dehydrogenase deficiency and Gilbert syndrome. Eur J Pediatr 2001;160(3):195.Google Scholar
Slusher, TM, Vreman, HJ, McLaren, DW, et al.Glucose-6-phosphate dehydrogenase deficiency and carboxyhemoglobin concentrations associated with bilirubin-related morbidity and death in Nigerian infants. J Pediatr 1995;126(1):102–8.Google Scholar
Oyebola, DD. Care of the neonate and management of neonatal jaundice as practised by Yoruba traditional healers of Nigeria. J Trop Pediatr 1983;29(1):1822.Google Scholar
Mentzer, WC, Collier, E. Hydrops fetalis associated with erythrocyte G-6-PD deficiency and maternal ingestion of fava beans and ascorbic acid. J Pediatr 1975;86(4):565–7.Google Scholar
MacDonald, MG. Hidden risks: Early discharge and bilirubin toxicity due to glucose 6-phosphate dehydrogenase deficiency. Pediatrics 1995;96(4 Pt 1):734–8.Google Scholar
Kaplan, M, Hammerman, C. The need for neonatal glucose-6-phosphate dehydrogenase screening: A global perspective. J Perinatol 2009;29(Suppl1):S4652.Google Scholar
Meloni, T, Forteleoni, G, Meloni, GF. Marked decline of favism after neonatal glucose-6-phosphate dehydrogenase screening and health education: The northern Sardinian experience. Acta Haematol 1992;87(1–2):2931.Google Scholar
American Academy of Pediatrics Subcommittee on Hyperbilirubinemia Management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation. Pediatrics 2004;114(1):297316.Google Scholar
Lin, Z, Fontaine, JM, Freer, DE, Naylor, EW. Alternative DNA-based newborn screening for glucose-6-phosphate dehydrogenase deficiency. Mol Genet Metab 2005;86(1–2):212–19.Google Scholar
Watchko, JF, Kaplan, M, Stark, AR, Stevenson, DK, Bhutani, VK. Should we screen newborns for glucose-6-phosphate dehydrogenase deficiency in the United States? J Perinatol 2013;33:499.Google Scholar
Bhutani, VK, Kaplan, M, Glader, B, et al. Point-of-care quantitative measure of glucose-6-phosphate dehydrogenase enzyme deficiency. Pediatrics 2015;136(5):e1268–75.Google Scholar
Kaplan, M, Vreman, HJ, Hammerman, C, et al. Contribution of haemolysis to jaundice in Sephardic Jewish glucose-6-phosphate dehydrogenase deficient neonates. Br J Haematol 1996;93(4):822–7.Google Scholar
Kappas, A, Drummond, GS, Valaes, T. A single dose of Sn-mesoporphyrin prevents development of severe hyperbilirubinemia in glucose-6-phosphate dehydrogenase-deficient newborns. Pediatrics 2001;108(1):2530.Google Scholar
McCurdy, PR, Morse, EE. Glucose-6-phosphate dehydrogenase deficiency and blood transfusion. Vox Sang 1975;28(3):230–7.Google Scholar
Mimouni, F, Shohat, S, Reisner, SH. G6PD-deficient donor blood as a cause of hemolysis in two preterm infants. Isr J Med Sci 1986;22(2):120–2.Google Scholar
Kumar, P, Sarkar, S, Narang, A. Acute intravascular haemolysis following exchange transfusion with G-6-PD deficient blood. Eur J Pediatr 1994;153(2):98–9.Google Scholar
Grace, RF, Zanella, A, Neufeld, EJ, et al. Erythrocyte pyruvate kinase deficiency: 2015 status report. Am J Hematol 2015;90(9):825–30.Google Scholar
Grace, RF, Bianchi, P, van Beers, EJ, et al. Clinical spectrum of pyruvate kinase deficiency: Data from the Pyruvate Kinase Deficiency Natural History Study. Blood 2018;131(20):2183–92.Google Scholar
Beutler, E, Gelbart, T. Estimating the prevalence of pyruvate kinase deficiency from the gene frequency in the general white population. Blood 2000;95(11):3585–8.Google Scholar
Carey, PJ, Chandler, J, Hendrick, A, et al. Prevalence of pyruvate kinase deficiency in northern European population in the north of England Northern Region Haematologists Group. Blood 2000;96(12):4005–6.Google Scholar
Zanella, A, Fermo, E, Bianchi, P, Chiarelli, LR, Valentini, G. Pyruvate kinase deficiency: The genotype-phenotype association. Blood Rev 2007;21(4):217–31.Google Scholar
Grace, RF, Layton, DM, Barcellini, W. How we manage patients with pyruvate kinase deficiency. Br J Haematol 2019;189:721–34.Google Scholar
Bowman, HS, McKusick, VA, Dronamraju, KR. Pyruvate kinase deficient hemolytic anemia in an Amish isolate. Am J Hum Genet 1965;17:18.Google Scholar
Rider, NL, Strauss, KA Brown, K, et al. Erythrocyte pyruvate kinase deficiency in an old-order Amish cohort: Longitudinal risk and disease management. Am J Hematol 2011;86(10):827–34.Google Scholar
Gallagher, PG, Glader, B. Diagnosis of pyruvate kinase deficiency. Pediatr Blood Cancer 2016;63(5):771–2.Google Scholar
Bianchi, P, Fermo, E, Glader, B, et al. Addressing the diagnostic gaps in pyruvate kinase deficiency: Consensus recommendations on the diagnosis of pyruvate kinase deficiency. Am J Hematol 2019;94(1):149–61.Google Scholar
Hutton, JJ, Chilcote, RR, Glucose phosphate isomerase deficiency with hereditary nonspherocytic hemolytic anemia. J Pediatr 1974;85(4):494–7.Google Scholar
Schroter, W, Koch, HH, Wonneberger, B, et al. Glucose phosphate isomerase deficiency with congenital nonspherocytic hemolytic anemia: A new variant (type Nordhorn;IClinical and genetic studies. Pediatr Res 1974;8(1):1825.Google Scholar
Van Biervliet, JP, Van Milligen-Boersma, L, Staal, GE. A new variant of glucosephosphate isomerase deficiency (GPI-Utrecht). Clin Chim Acta 1975;65(2):157–65.Google Scholar
Ravindranath, Y, Paglia, DE, Warrier, I, et al. Glucose phosphate isomerase deficiency as a cause of hydrops fetalis. N Engl J Med 1987;316(5):258–61.Google Scholar
Xu, W, Beutler, E. The characterization of gene mutations for human glucose phosphate isomerase deficiency associated with chronic hemolytic anemia. J Clin Invest 1994;94(6):2326–9.Google Scholar
Vora, S. Isozymes of phosphofructokinase. Isozymes Curr Top Biol Med Res 1982;6:119–67.Google Scholar
Vora, S, DiMauro, S, Spear, D, Harker, D, Danon, MJ. Characterization of the enzymatic defect in late-onset muscle phosphofructokinase deficiency: New subtype of glycogen storage disease type VII. J Clin Invest 1987;80(5):1479–85.Google Scholar
Schneider, AS, Valentine, WN, Hattori, M, Heins, HL Jr. Hereditary hemolytic anemia with triosephosphate isomerase deficiency. N Engl J Med 1965;272:229–35.Google Scholar
Valentine, WN, Hsieh, HS, Paglia, DE, et al. Hereditary hemolytic anemia: Association with phosphoglycerate kinase deficiency in erythrocytes and leukocytes. Trans Assoc Am Physicians 1968;81:4965.Google Scholar
Schneider, A, Westwood, B, Yim, C, et al. Triosephosphate isomerase deficiency: Repetitive occurrence of point mutation in amino acid 104 in multiple apparently unrelated families. Am J Hematol 1995;50(4):263–8.Google Scholar
112. Valentine, WN, Oski, FA, Paglia, DE, et al. Hereditary hemolytic anemia with hexokinase deficiency: Role of hexokinase in erythrocyte aging. N Engl J Med 1967;276(1):111.Google Scholar
Kanno, H. Hexokinase: Gene structure and mutations. Baillieres Best Pract Res Clin Haematol 2000;13(1):838.Google Scholar
Beutler, E. Red cell enzyme defects as nondiseases and as diseases. Blood 1979;54(1):17.Google Scholar
Beutler, E, Baranko, PV, Feagler, J, et al. Hemolytic anemia due to pyrimidine-5’-nucleotidase deficiency: Report of eight cases in six families. Blood 1980;56(2):251–5.Google Scholar
Paglia, DE, Valentine, WN. Hereditary and acquired defects in the pyrimidine nucleotidase of human erythrocytes. Curr Top Hematol 1980;3:75109.Google Scholar
Paglia, DE, Valentine, WN, Keitt, AS, Brockway, RA, Nakatani, M. Pyrimidine nucleotidase deficiency with active dephosphorylation of dTMP: Evidence for existence of thymidine nucleotidase in human erythrocytes. Blood 1983;62(5):1147–9.Google Scholar
Taher, AT, Weatherall, DJ, Cappellini, MD. Thalassaemia. Lancet 2018;391(10116):155–67.Google Scholar
Vichinsky, E. Advances in the treatment of alpha-thalassemia. Blood Rev 2012;26 Suppl 1:S3134.Google Scholar
Piel, FB, Weatherall, DJ. The alpha-thalassemias. N Engl J Med 2014;371(20):1908–16.Google Scholar
Singh, SA, Sarangi, S, Appiah-Kubi, A, et al. Hb Adana (HBA2 or HBA1:c.179 G > A) and alpha thalassemia: Genotype-phenotype correlation. Pediatr Blood Cancer 2018;65(9):e27220.Google Scholar
Chui, DH, Waye, JS. Hydrops fetalis caused by alpha-thalassemia: An emerging health care problem. Blood 1998;91(7):2213–22.Google Scholar
Liang, ST, Wong, VC, So, WW, et al. Homozygous alpha-thalassaemia: Clinical presentation, diagnosis and management: A review of 46 cases. Br J Obstet Gynaecol 1985;92(7):680–4.Google Scholar
Beaudry, MA, Ferguson, DJ, Pearse, K, et al. Survival of a hydropic infant with homozygous alpha-thalassemia-1. J Pediatr 1986;108(5 Pt 1):713–16.Google Scholar
Bianchi, DW, Beyer, EC, Stark, AR, et al. Normal long-term survival with alpha-thalassemia. J Pediatr 1986;108 (5 Pt 1):716–18.Google Scholar
Singer, ST, Styles, L, Bojanowski, J, et al. Changing outcome of homozygous alpha-thalassemia:cautious optimism. J Pediatr Hematol Oncol 2000;22(6):539–42.Google Scholar
Chui, DH. Alpha-thalassemia: Hb H disease and Hb Barts hydrops fetalis. Ann N Y Acad Sci 2005;1054:2532.Google Scholar
Hsieh, FJ, Ko, TM, Chen, HY. Hydrops fetalis caused by severe alpha-thalassemia. Early Hum Dev 1992;29(1–3):233–6.Google Scholar
Guy, G, Coady, DJ, Jansen, V, Snyder, J, Zinberg, S. Alpha-thalassemia hydrops fetalis: Clinical and ultrasonographic considerations. Am J Obstet Gynecol 1985;153(5):500–4.Google Scholar
Stein, J, Berg, C, Jones, JA, Detter, JC. A screening protocol for a prenatal population at risk for inherited hemoglobin disorders: Results of its application to a group of Southeast Asians and blacks. Am J Obstet Gynecol 1984;150(4):333–41.Google Scholar
Glader, BE. Screening for anemia and erythrocyte disorders in children. Pediatrics 1986;78(2):368–9.Google Scholar
Chan, V, Ghosh, A, Chan, TK, Wong, V, Todd, D. Prenatal diagnosis of homozygous alpha thalassaemia by direct DNA analysis of uncultured amniotic fluid cells. Br Med J (Clin Res Ed) 1984;288(6427):1327–9.Google Scholar
Fucharoen, S, Winichagoon, P, Thonglairoam, V, et al. Prenatal diagnosis of thalassemia and hemoglobinopathies in Thailand: Experience from 100 pregnancies. Southeast Asian J Trop Med Public Health 1991;22(1):1629.Google Scholar
Hsieh, FJ, Chang, FM, Ko, TM, Chen, HY Percutaneous ultrasound-guided fetal blood sampling in the management of nonimmune hydrops fetalis. Am J Obstet Gynecol 1987;157(1):44–9.Google Scholar
Jelin, AC, Sagaser, KG, Wilkins-Haug, L. Prenatal genetic testing options. Pediatr Clin North Am 2019;66(2):281–93.Google Scholar
Winichagoon, P, Sithongdee, S, Kanokpongsakdi, S, et al. Noninvasive prenatal diagnosis for hemoglobin Bart’s hydrops fetalis. Int J Hematol 2005;81(5):396–9.Google Scholar
Tungwiwat, W, Fucharoen, S, Fucharoen, G, Ratanasiri, T, Sanchaisuriya, K. Development and application of a real-time quantitative PCR for prenatal detection of fetal alpha(0)-thalassemia from maternal plasma. Ann N Y Acad Sci 2006;1075:103–7.Google Scholar
Ho, SS, Chong, SS, Koay, ES, et al. Noninvasive prenatal exclusion of haemoglobin Bart’s using foetal DNA from maternal plasma. Prenat Diagn 2009;30(1):6573.Google Scholar
Hudecova, I, Chiu, RW. Non-invasive prenatal diagnosis of thalassemias using maternal plasma cell free DNA. Best Pract Res Clin Obstet Gynaecol 2017;39:6373.Google Scholar
Yates, A. Prenatal Screening and Testing for Hemoglobinopathy (Philadelphia PA: Wolters Kluwer, 2019).Google Scholar
Chik, KW, Shing, MM, Li, CK, et al. Treatment of hemoglobin Bart’s hydrops with bone marrow transplantation. J Pediatr 1998;132(6):1039–42.Google Scholar
Thornley, I, Lehmann, L, Ferguson, WS, et al. Homozygous alpha-thalassemia treated with intrauterine transfusions and postnatal hematopoietic stem cell transplantation. Bone Marrow Transplant 2003;32(3):341–2.Google Scholar
Lucke, T, Pfister, S Durken, M. Neurodevelopmental outcome and haematological course of a long-time survivor with homozygous alpha-thalassaemia: Case report and review of the literature. Acta Paediatr 2005;94(9):1330–3.Google Scholar
Yi, JS, Moertel, CL, Baker, KS. Homozygous alpha-thalassemia treated with intrauterine transfusions and unrelated donor hematopoietic cell transplantation. J Pediatr 2009;154(5):766–8.Google Scholar
Derderian, SC, Jeanty, C, Walters, MC, Vichinsky, E, MacKenzie, TC. In utero hematopoietic cell transplantation for hemoglobinopathies. Front Pharmacol 2014;5:278.Google Scholar
Higgs, DR, Weatherall, DJ. The alpha thalassaemias. Cell Mol Life Sci 2009;66(7):1154–62.Google Scholar
Jomoui, W, Fucharoen, G, Sanchaisuriya, K, Nguyen, VH, Fucharoen, S. Hemoglobin Constant Spring among Southeast Asian populations: Haplotypic heterogeneities and phylogenetic analysis. PLoS One 2015;10(12):e0145230.Google Scholar
Sirilert, S, Charoenkwan, P, Sirichotiyakul, S, et al. Prenatal diagnosis and management of homozygous hemoglobin Constant Spring disease. J Perinatol 2019;39(7):927–33.Google Scholar
Olivieri, NF. The beta-thalassemias. N Engl J Med 1999;341(2):99109.Google Scholar
Cunningham, MJ, Sankaran, VG, Nathan, DG Orkin, SH. The thalassemias. In Orkin, SH, Nathan, DG, Ginsburg, D, et al., eds. Nathan and Oski’s Hematology of Infancy and Childhood (Philadelphia PA: Saunders/Elsevier,2009).Google Scholar
Cao, A, Rosatelli, MC, Monni, G, Galanello, R. Screening for thalassemia: A model of success. Obstet Gynecol Clin North Am 2002;29(2):305–28, vi–vii.Google Scholar
Olivieri, NF, Muraca, GM, O’Donnell, A, et al.Studies in haemoglobin E beta-thalassaemia. Br J Haematol 2008;141(3):388–97.Google Scholar
Sirichotiyakul, S, Saetung, R, Sanguansermsri, T. Prenatal diagnosis of beta-thalassemia/Hb E by hemoglobin typing compared to DNA analysis. Hemoglobin 2009;33(1):1723.Google Scholar
Vichinsky, EP, MacKlin, EA, Waye, J S, Lorey, F, Olivieri, NF. Changes in the epidemiology of thalassemia in North America: A new minority disease. Pediatrics 2005;116(6):e818825.Google Scholar
Lorey, F, Cunningham, G, Shafer, F, Lubin, B, Vichinsky, E. Universal screening for hemoglobinopathies using high-performance liquid chromatography: Clinical results of 2.2 million screens. Eur J Hum Genet 1994;2(4):262–71.Google Scholar
Lenfant, C. The Management of Sickle Cell Disease (Bethesda MD: National Insitutes of Health, 2002).Google Scholar
Gaston, MH, Verter, J I, Woods, G, et al. Prophylaxis with oral penicillin in children with sickle cell anemia. A randomized trial. N Engl J Med 1986;314(25):1593–99.Google Scholar
Michlitsch, J, Azimi, M, Hoppe, C, et al. Newborn screening for hemoglobinopathies in California. Pediatr Blood Cancer 2009;52(4):486–90.Google Scholar
Wethers, D, Pearson, HA, Gaston, M. Newborn screening for sickle cell disease and other hemoglobinopathies. Pediatrics 1989;89(5):813–14.Google Scholar
Koshy, M, Burd, L. Obstetric and gynecologic issues. In Embury, S, Hebbel, RP eds. Sickle Cell Disease: Basic Principles and Clinical Practice (New York: Raven Press, 1995), p. 689.Google Scholar
Villers, MS, Jamison, MG, De Castro, LM, James, AH. Morbidity associated with sickle cell disease in pregnancy. Am J Obstet Gynecol 2008;199(2):125,e121–5.Google Scholar
Williamson, D. The unstable haemoglobins. Blood Rev 1993;7(3):146–63.Google Scholar
Carrell, RW, Kay, R. A simple method for the detection of unstable haemoglobins. Br J Haematol 1972;23(5):615–19.Google Scholar
Lee-Potter, JP, Deacon-Smith, RA, Simpkiss, MJ, Kamuzora, H, Lehmann, H. A new cause of haemolytic anaemia in the newborn. A description of an unstable fetal haemoglobin: F Poole, alpha2-G-gamma2 130 tryptophan yields glycine. J Clin Pathol 1975;28(4):317–20.Google Scholar
Charache, S, Mondzac, AM, Gessner, U. Hemoglobin Hasharon (alpha-2–47 his(CD5;beta-2): A hemoglobin found in low concentration. J Clin Invest 1969;48(5):834–47.Google Scholar
Levine, RL, Lincoln, DR, Buchholz, WM, Gribble, TJ Schwartz, HC. Hemoglobin Hasharon in a premature infant with hemolytic anemia. Pediatric Research 1975;9(1):711.Google Scholar
Bender, JW, Reilly, MP Asakura, T. Molecular stability and function of hemoglobins Hasharon (alpha(2)47 (CD5)Asp–His beta 2) and Hasharon (alpha(2)47 (CD5)Asp–His delta 2). Hemoglobin 1984;8(1):6173.Google Scholar
Martin, H, Huisman, TH. Formation of ferrihaemoglobin of isolated human haemoglobin types by sodium nitrite. Nature 1963;200:898–9.Google Scholar
Bartos, HR, Desforges, JF. Erythrocyte DPNH dependent diaphorase levels in infants. Pediatrics 1966;37(6):991–3.Google Scholar
Comly, HH. Cyanosis in infants caused by nitrates in well water. J Am Med Assoc 1945;129(2):112–16.Google Scholar
Gelperin, A, Jacobs, EE, Kletke, LS. The development of methemoglobin in mothers and newborn infants from nitrate in water supplies. IMJ Ill Med J 1971;140(1):42–4 passim.Google Scholar
Keating, JP, Lell, ME, Strauss, AW, Zarkowsky, H, Smith, GE. Infantile methemoglobinemia caused by carrot juice. N Engl J Med 1973;288(16):824–6.Google Scholar
Yano, SS, Danish, EH, Hsia, YE. Transient methemoglobinemia with acidosis in infants. J Pediatr 1982;100(3):415–18.Google Scholar
Avner, JR, Henretig, FM, McAneney, CM. Acquired methemoglobinemia: The relationship of cause to course of illness. Am J Dis Child 1990;144(11):1229–30.Google Scholar
Kay, MA, O’Brien, W, Kessler, B, et al. Transient organic aciduria and methemoglobinemia with acute gastroenteritis. Pediatrics 1990;85(4):589–92.Google Scholar
Pollack, ES, Pollack, CV, Jr. Incidence of subclinical methemoglobinemia in infants with diarrhea. Ann Emerg Med 1994;24(4):652–6.Google Scholar
Hanukoglu, A, Danon, PN. Endogenous methemoglobinemia associated with diarrheal disease in infancy. J Pediatr Gastroenterol Nutr 1996;23(1):17.Google Scholar
Wessel, DL, Adatia, I, Van Marter, LJ, et al. Improved oxygenation in a randomized trial of inhaled nitric oxide for persistent pulmonary hypertension of the newborn. Pediatrics 1997;100(5):E7.Google Scholar
Climie, CR, McLean, S, Starmer, GA, Thomas, J. Methaemoglobinaemia in mother and foetus following continuous epidural analgesia with prilocaine: Clinical and experimental data. Br J Anaesth 1967;39(2):155160.Google Scholar
Law, RM, Halpern, S, Martins, RF, et al. Measurement of methemoglobin after EMLA analgesia for newborn circumcision. Biol Neonate 1996;70(4):213–17.Google Scholar
Tush, GM, Kuhn, RJ. Methemoglobinemia induced by an over-the-counter medication. Ann Pharmacother 1996;30(11):1251–4.Google Scholar
Brisman, M, Ljung, BM, Otterbom, I, Larsson, LE, Andreasson, SE. Methaemoglobin formation after the use of EMLA cream in term neonates. Acta Paediatr 1998;87(11):1191–4.Google Scholar
Essink-Tebbes, CM, Wuis, EW, Liem, KD, van Dongen, RT, Hekster, YA. Safety of lidocaine-prilocaine cream application four times a day in premature neonates:a pilot study. Eur J Pediatr 1999;158(5):421–3.Google Scholar
Kearns, GL, Fiser, DH. Metoclopramide-induced methemoglobinemia. Pediatrics 1988;82(3):364–6.Google Scholar
Hjelt, K, Lund, JT, Scherling, B, et al. Methaemoglobinaemia among neonates in a neonatal intensive care unit. Acta Paediatr 1995;84(4):365–70.Google Scholar
Percy, MJ, McFerran, NV, Lappin, TR. Disorders of oxidised haemoglobin. Blood Rev 2005;19(2):61–8.Google Scholar
Percy, MJ, Lappin, TR. Recessive congenital methaemoglobinaemia: Cytochrome b(5; reductase deficiency. Br J Haematol 2008;141(3):298308.Google Scholar
Kutlar, F, Hilliard, LM, Zhuang, L, et al. Hb M Dothan [beta 25/26 (B7/B8)/(GGT/GAG-->GAG//Gly/Glu-->Glu]; a new mechanism of unstable methemoglobin variant and molecular characteristics. Blood Cells Mol Dis 2009;43(3):235–8.Google Scholar
Hayashi, A, Fujita, T, Fujimura, M, Titani, K. A new abnormal fetal hemoglobin, Hb FM-Osaka (alpha 2 gamma 2 63His replaced by Tyr). Hemoglobin 1980;4(3–4):4478.Google Scholar
Glader, BE, Zwerdling, D, Kutlar, F, et al. Hb F-M-Osaka or alpha 2 G gamma 2 (63)(E7)His ----Tyr in a Caucasian male infant. Hemoglobin 1989;13(7–8):769–73.Google Scholar
Priest, JR, Watterson, J, Jones, RT, Faassen, AE, Hedlund, BE. Mutant fetal hemoglobin causing cyanosis in a newborn. Pediatrics 1989;83(5):734–6.Google Scholar
Harley, JD, Celermajer, JM. Neonatal methaemoglobinaemia and the red-brown screening-test. Lancet 1970;2(7685):1223–5.Google Scholar

References

Wood, JL. Plethora in the newborn infant associated with cyanosis and convulsions. J Pediatr 1952;54:143–51.Google Scholar
Michael, AF, 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 with a high hematocrit. Lancet 1964;2:499500.Google Scholar
Gross, GP, Hathaway, WE, McGaughey, HR. Hyperviscosity in the neonate. J Pediatr 1973;82:1004–12.Google Scholar
Ramamurthy, RS, Brans, YW. Neonatal polycythemia. I. Criteria for diagnosis and treatment. Pediatrics 1981;68:168–74.Google Scholar
Oh, W, Lind, J. Venous and capillary hematocrit in newborn infants and placental transfusion. Acta Paediatr Scand 1966;55:3840.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
Brooks, GI, Backes, CR. Hyperviscosity secondary to polycythemia in the appropriate for gestational age neonate. J Am Obstet Assoc 1981;80:415–18.Google Scholar
Reisner, SH, Mor, N, Levy, Y, Merlob, P. Incidence of neonatal polycythemia. Isr J Med Sci 1983;19:848–9.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
Oh, W, Blankenship, W, Lind, J. Further study of neonatal blood volume in relation to placental transfusion. Ann Paediatr 1996;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 1982; 9:559–92.Google Scholar
McDonald, SJ, Midddleton, P, Dowswell, T, Morris, PS. Cochrane in context: Effect of timing of umbilical cord clamping in term infants on maternal and neonatal outcomes. Evidence-based child Health: A Cochrane Review Journal 2014:9:198400.Google Scholar
Shohat, M, Teisner, SH, Mimoini, F, Merlob, P. Neonatal polycythemia. II. Definition related to time of sampling. Pediatrics 1984;73:1113.Google Scholar
Poiseuille, JLM. Recherches experimentales sur le mouvement des liquides dans les tubes de tres petits diametres. C R Acad Sci 1840;11:9611041.Google Scholar
Van der Elst, CW, Malan, AF, de V Heese H, . Blood viscosity in modern medicine. S Afr Med J 1977;52:526–8.Google Scholar
Wells, RE, Pento, R, Merrill, EW. Measurements of viscosity of biologic fluids by core plate viscometer. J Lab Clin Med 1961;57:646–56.Google Scholar
Wells, RE, Merrill, EW. Influence of flow properties of blood upon viscosity–hematocrit relationships. J Clin Invest 1961;41:1591–8.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
Linderkamp, O, Versmold, HT, Riegel, KP, Betke, K. Contributions of red cells and plasma to blood viscosity in preterm and full-term infants and adults. Pediatrics 1984;74:4551.Google Scholar
Burton, AC. Role of geometry, of size and shape, in the microcirculation. Fed Proc 1966;25:1753–60.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
Somer, T, Ditze, J. Clinical and rheological studies in a patient with hyperviscosity syndrome due to Waldenstrom’s macroglobulinemia. Bibl Haematol 1981;47:242–6.Google Scholar
Charm, SE, Kurland, GS. Blood Flow and Microcirculation (New York: John Wiley & Sons, 1974).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
Linderkamp, O, Wu, PYK, Meiselman, HJ. Deformability of density separated red blood cells in normal newborn infants and adults. Pediatr Res 1982;16:964–8.Google Scholar
Lichtman, MA. Cellular deformability during maturation of the myeloblast. 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;52:350–8.Google Scholar
Miller, ME. Developmental maturation of human neutrophil motility and its relationship to membrane deformability. In Bellanti, JA, Dayton, DH, eds. The Phagocytic Cell in Host Resistance (New York: Raven Press, 1975), p. 295.Google Scholar
Rand, PW, Austin, WH, Lacombe, E, Barker, N. pH and blood viscosity. J Appl Physiol 1968;25:550–9.Google Scholar
Fahraeus, R, Lindqvist, T. The viscosity of the blood in narrow capillary tubes. Am J Physiol 1931;96:562–8.Google Scholar
Nowicki, P, Oh, W, Yao, A, Hansen, NB, Stonestreet, SS. Effect of polycythemia on gastrointestinal blood flow and oxygenation in piglets. Am J Physiol 1984;247:G220G225.Google Scholar
LeBlanc, MH, Kotagal, VR, Kleinman, LI. Physiological effects of hypervolemic polycythemia in newborn dogs. J Appl Physiol 1982;53:865–72.Google Scholar
Surjadhana, A, Rouleau, J, Boerboom, L, Hoffman, JIE. Myocardial blood flow and its distribution in anesthetized polycythemic dogs. Circ Res 1978;43:619–31.Google Scholar
Brashear, RE. Effects of acute plasma for blood exchange in experimental polycythemia. Respiration 1980;40:297306.Google Scholar
Rosenkrantz, TS, Oh, W. Cerebral blood flow velocity in infants with polycythemia and hyperviscosity: Effects of partial exchange transfusion with Plamanate. 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, Muister, AJ, Cole, RB, Paul, MH. 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;11:394A.Google Scholar
Swetnam, SM, Yabek, SM, Alverson, DC. Hemodynamic consequences of neonatal polycythemia. J Pediatr 1987;110:443–7.Google Scholar
Murphy, DJ, Jr., Reller, MD, Meyer, RA, Kaplan, S. Effects of neonatal polycythemia and partial exchange transfusion on cardiac function: An echocardiographic study. Pediatrics 1985;76:90913.Google Scholar
Boehm, G, Delitzsch, AK, Senger, H, et al. Postnatal development of liver and exocrine pancreas in polycythemic newborn infants. J Pediatr Gastroenterol Nutr 1992; 15:310–14.Google Scholar
Kotagal, VR, Kleinman, LI. Effect of acute polycythemia on newborn renal hemodynamics and function. Pediatr Res 1982;16:14851.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. Eur 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, Huxtabl, RF. Effects of polycythemia and hyperviscosity on cutaneous blood flow and transcutaneous pO2 and pCO2 in neonate. Pediatrics 1984;74:389–94.Google Scholar
Jones, MD, Traystman, RJ, Simmons, MA, Molteni, RA. Effects of changes in arterial O2 content on cerebral blood flow in the lamb. Am J Physiol 1981;240:H209H215.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
Goldstein, M, Stonestreet, BS, Brann, BS, 4th, Oh, W. Cerebral cortical blood flow and oxygen metabolism in normocythemic hyperviscous newborn piglets. Pediatr Res 1988;24:486–9.Google Scholar
Rosenkrantz, TS, Philipps, AF, Skrzypczak, PS, Raye, JR. Cerebral metabolism in the newborn lamb with polythemia. Pediatr Res 1985;23:329–33.Google Scholar
Tenenbaum, DG, Piasecki, GJ, Oh, W, Rosenkrantz, TS, Jackson, BT. Fetal polycythemia and hyperviscosity: effect in umbilical blood flow and fetal oxygen consumption. Am J Obstet Gynecol 1983;147:4851.Google Scholar
Saigal, S, Usher, RH. Symptomatic neonatal plethora. Biol Neonate 1977;32:6272.Google Scholar
Sacks, MO. Occurrence of anemia and polycythemia in phenotypically dissimilar single ovum human twins. Pediatrics 1959;24:6048.Google Scholar
Schwartz, JL, Maniscalco, WM, Lane, AT, Currao, WJ. Twin transfusion syndrome causing cutaneous erythropoiesis. Pediatrics 1984;74:527–9.Google Scholar
Philip, AGS, Yee, AB, Rosy, M, et al. Placental transfusion as an intrauterine phenomenon in deliveries complicated by fetal distress. Br Med J 1969;2:1113.Google Scholar
Flod, NE, Ackerman, BD. Perinatal asphyxia and residual placental blood volume. Acta Paediatr Scand 1971;60:433–6.Google Scholar
Yao, AC, Lind, J. Effect of gravity on placental transfusion. Lancet 1969;2:505–6.Google Scholar
Oh, W, Omori, K, Emmanouilides, GC, Phelps, DI. Placenta to lamb fetus transfusion in utero during acute hypoxia. Am J Obstet Gynecol 1975;122:316–21.Google Scholar
Humbert, JR, Abelson, H, Hathaway, WE, Battaglia, FC. Polycythemia in small for gestational age infants. J Pediatr 1969;75:812–19.Google Scholar
Widness, JA, Garcia, JA, Oh, W, Schwartz, R. 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, Raye, JR. Arterial hypoxemia and hyperinsulinemia in the chronically hyperglycemic fetal lamb. Pediatr Res 1982;16:653–8.Google Scholar
Hod, M, Merlob, P, Friedman, S. Prevalence of congenital anomalies and neonatal complications in the offspring of diabetic mothers in Israel. Isr J Med Sci 1991;27:498502.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 Medical Book Co., 1986), pp. 93123.Google Scholar
Hakanson, DO, Oh, W. Hyperviscosity in the small-for-gestational age infant. Biol Neonate 1980;37:109–12.Google Scholar
Rawlings, JS, Pettet, G, Wiswell, TE, Clapper, J. Estimated blood volumes in polycythemic neonates as a function of birth weight. J Pediatr 1982;101:594.Google Scholar
Brans, YW, Shannon, DL, Ramamurthy, RS. Neonatal polycythemia. II. Plasma, blood and red cell volume estimates in relation to hematocrit levels and quality of intrauterine growth. Pediatrics 1981;68:175–82.Google Scholar
Thorton, CJ, Shanno, DL, Hunter, MA, Ramamurthy, RS, Brans, YW. Body water estimates in neonatal polycythemia. J Pediatr 1983;102:113–17.Google Scholar
Oh, W, Wallgren, G, Hanson, JS, Lind, J. The effects of placental transfusion on respiratory mechanics of normal term newborn infants. Pediatrics 1967;40:612.Google Scholar
Scott, F, Evans, N. Distal gangrene in a polycythemic recipient fetus in twin-twin transfusion. Obstet Gynecol 1995;86:677–9.Google Scholar
Van der Elst, CW, Moteno, CD, Malan, AF, de V Heese, H. The management of polycythemia in the newborn infant. Early Hum Dev 1980;4:393403.Google Scholar
Host, A, Ulrich, M. Late prognosis in untreated neonatal polycythemia with minor or no symptoms. Acta Paediatr Scand 1982;71:629–33.Google Scholar
Leake, RD, Thanopoulos, B, Nieberg, R. Hyperviscosity syndrome associated with necrotizing enterocolitis. Am J Dis Child 1975;129:1192–4.Google Scholar
Hakanson, DO, Oh, W. Necrotizing enterocolitis and hyperviscosity in the newborn infant. J Pediatr 1977;90:458–61.Google Scholar
Martinez-Tallo, E, Claure, N, Bancalari, E. Necrotizing entercolitis in full term or near term infants: Risk factors. Biol Neonate 1997;71:292–8.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, Koops, BL. Gastrointestinal injury in polycythemic term infants. Pediatrics 1985;76:225–31.Google Scholar
Herson, VC, Raye, JR, Rowe, JC, Philipps, AF. 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
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
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, Mandini, G, Branson, HE. 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, Whidden, MA. 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
Voorhies, TM, Lipper, EG, Lee, BCP, et al. Occlusive vascular disease in asphyxiated newborn infants. J Pediatr 1984;105:92.Google Scholar
Peters, M, Ten Cate, JW, Koo, LH, Breederveld, C. Persistent antithrombin III deficiency: Risk factor for thromboembolic complication in neonates small for gestational age. J Pediatr 1984;105:310–14.Google Scholar
Merchant, RH, Agarwal, MB, Joshi, NC, Parekh, SR. Neonatal polycythemia: A potentially serious disorder. Indian J Pediatr 1983;50:149–52.Google Scholar
Amit, M, Camfield, PR. Neonatal polycythemia causing multiple cerebral infarcts. Arch Neurol 1980;37:109–10.Google Scholar
Saggese, G, Bertelloni, S, Baroncelli, GI, et al. Elevated calcitonin gene related peptide in polycythemic newborn infants. Acta Pediatr 1992;81:966–8.Google Scholar
Alkalay, A, Pomerance, JJ, Prause, J, et al. Cholecalciferol metabolites in polycythemic newborns. Isr J Med Sci 1985;21:95–7.Google Scholar
Goldberg, K, Wirth, FH, Hathaway, WE. Neonatal hyperviscosity. II. Effect of partial plasma exchange transfusion. Pediatrics 1982;69:419–25.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
Black, VD, Lubchenco, LO, Koops, BL, Poland, RL, Powell, DP. 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
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
Dempsey, EM, Barrington, K. Short and long term outcomes following partial exchange transfusion in the polycythemic newborn: A systematic review. Arch Dis Child Fetal Neonatal Ed 2006;91(1):F2F6.Google Scholar
Supapannachart, S, Siripoonya, P, Boonwattanasoontorn, W, Kanjanavanit, S. Neonatal polycythemia: Effects of partial exchange transfusion using fresh frozen plasma, Haemaccel and normal saline. Med Assoc Thai 1999;82(Suppl1):S82S86.Google Scholar
Wong, W, Fok, TF, Lee, CH, et al. Randomised controlled trial: comparison of colloid or crystalloid for partial exchange transfusion for treatment of neonatal polycythaemia. Arch Dis Child Fetal Neonatal Ed 1997;77:F115F118.Google Scholar
Glader, B. Erythrocyte disorders in infancy. In Schaffer, AJ, Avery, ME, eds. Diseases of the Newborn (Philadelphia, PA: W. B. Saunders, 1977), p. 625.Google Scholar
American Academy of Pediatrics Committee on Fetus and Newborn. Routine evaluation of blood pressure, hematocrit, and glucose in newborns. Pediatrics 1993;92:474–6.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
×