Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-75dct Total loading time: 0 Render date: 2024-06-08T04:22:28.891Z Has data issue: false hasContentIssue false

Chapter 4 - Bone Marrow Failure Syndromes

from Section II - Bone Marrow Failure and Immune 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

Inherited bone marrow failure syndromes (IBMFS) are a rare but important consideration in the differential diagnosis of cytopenias in childhood [1]. However, diagnosis of IBMFS in the newborn period can be challenging because many of the manifestations considered typical for a specific disorder may not yet be present, and in many cases children will not be recognized until later in life. Young children with IBMFS may have one or more cytopenias, congenital anomalies, both, or neither. A high index of suspicion for an IBMFS is required in order to establish the correct diagnosis, determine appropriate clinical management and follow up plans, and provide the family with genetic counseling. Some IBMFS predispose to leukemia or solid tumors; while the development of cancer is uncommon in the newborn period, this risk is an important determinant of subsequent follow up for the child and any affected family members.

Type
Chapter
Information
Neonatal Hematology
Pathogenesis, Diagnosis, and Management of Hematologic Problems
, pp. 43 - 61
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

Khincha, PP, Savage, SA. Neonatal manifestations of inherited bone marrow failure syndromes. Semin Fetal Neonatal Med 2016;21(1):5765.CrossRefGoogle ScholarPubMed
Giri, N, Reed, HD, Stratton, P, Savage, SA, Alter, BP. Pregnancy outcomes in mothers of offspring with inherited bone marrow failure syndromes. Pediatr Blood Cancer 2018;65(1).CrossRefGoogle ScholarPubMed
Kutler, DI, Auerbach, AD. Fanconi anemia in Ashkenazi Jews. Fam Cancer 2004;3(3–4):241–8.Google Scholar
Vlachos, A, Klein, GW, Lipton, JM. The Diamond–Blackfan Anemia Registry: Tool for investigating the epidemiology and biology of Diamond–Blackfan anemia. J Pediatr Hematol Oncol 2001;23(6):377–82.CrossRefGoogle ScholarPubMed
Farrar, JE, Dahl, N. Untangling the phenotypic heterogeneity of Diamond Blackfan anemia. Semin Hematol 2011;48(2):124–35.Google Scholar
Vlachos, A, Rosenberg, PS, Atsidaftos, E, Alter, BP, Lipton, JM. Incidence of neoplasia in Diamond–Blackfan anemia: A report from the Diamond–Blackfan Anemia Registry. Blood 2012;119(16):3815–9.Google Scholar
Vlachos, A, Rosenberg, PS, Atsidaftos, E, et al. Increased risk of colon cancer and osteogenic sarcoma in Diamond–Blackfan anemia. Blood 2018;132(20):2205–8.Google Scholar
Boria, I, Garelli, E, Gazda, HT, et al. The ribosomal basis of Diamond–Blackfan anemia: Mutation and database update. Hum Mutat 2010;31(12):1269–79.Google Scholar
Dianzani, I, Loreni, F. Diamond–Blackfan anemia: A ribosomal puzzle. Haematologica 2008;93(11):1601–4.CrossRefGoogle ScholarPubMed
Da Costa, L, Narla, A, Mohandas, N. An update on the pathogenesis and diagnosis of Diamond–Blackfan anemia. F1000Res 2018;7.CrossRefGoogle Scholar
Ulirsch, JC, Verboon, JM, Kazerounian, S, et al. The genetic landscape of Diamond–Blackfan anemia. Am J Hum Genet 2018;103(6):930–47.CrossRefGoogle ScholarPubMed
Sankaran, VG, Ghazvinian, R, Do, R, et al. Exome sequencing identifies GATA1 mutations resulting in Diamond–Blackfan anemia. J Clin Invest 2012;122(7):2439–43.CrossRefGoogle ScholarPubMed
Parrella, S, Aspesi, A, Quarello, P, et al. Loss of GATA-1 full length as a cause of Diamond-Blackfan anemia phenotype. Pediatr Blood Cancer 2014;61(7):1319–21.Google Scholar
Vlachos, A, Ball, S, Dahl, N, et al. Diagnosing and treating Diamond–Blackfan anaemia: Results of an international clinical consensus conference. Br J Haematol 2008;142(6):859–76.CrossRefGoogle ScholarPubMed
Fargo, JH, Kratz, CP, Giri, N, et al. Erythrocyte adenosine deaminase: Diagnostic value for Diamond–Blackfan anaemia. Br J Haematol 2013;160(4):547–54.CrossRefGoogle ScholarPubMed
Da Costa, L, O’Donohue, MF, van Dooijeweert, B, et al. Molecular approaches to diagnose Diamond–Blackfan anemia: The EuroDBA experience. Eur J Med Genet 2018;61(11):664–73.CrossRefGoogle ScholarPubMed
Bartels, M, Bierings, M. How I manage children with Diamond–Blackfan anaemia. Br J Haematol 2019;184(2):123–33.CrossRefGoogle Scholar
Lahoti, A, Harris, YT, Speiser, PW, et al. Endocrine dysfunction in Diamond–Blackfan anemia (DBA): A report from the DBA Registry (DBAR). Pediatr Blood Cancer 2016;63(2):306–12.CrossRefGoogle ScholarPubMed
Vlachos, A, Muir, E. How I treat Diamond–Blackfan anemia. Blood 2010;116(19):3715–23.CrossRefGoogle Scholar
Venugopal, P, Moore, S, Lawrence, DM, et al. Self-reverting mutations partially correct the blood phenotype in a Diamond–Blackfan anemia patient. Haematologica 2017;102(12):e506–e9.Google Scholar
Garelli, E, Quarello, P, Giorgio, E, et al. Spontaneous remission in a Diamond–Blackfan anaemia patient due to a revertant uniparental disomy ablating a de novo RPS19 mutation. Br J Haematol 2019;185(5):994–8.Google Scholar
Jongmans, MCJ, Diets, IJ, Quarello, P, et al. Somatic reversion events point towards. Haematologica 2018;103(12):e607–e9.Google Scholar
Ben-Ami, T, Revel-Vilk, S, Brooks, R, et al. Extending the clinical phenotype of adenosine deaminase 2 deficiency. J Pediatr 2016;177:316–20.Google Scholar
Hashem, H, Egler, R, Dalal, J. Refractory pure red cell aplasia manifesting as deficiency of adenosine deaminase 2. J Pediatr Hematol Oncol 2017;39(5):e293–e6.Google Scholar
Meyts, I, Aksentijevich, I. Deficiency of adenosine deaminase 2 (DADA2): Updates on the phenotype, genetics, pathogenesis, and treatment. J Clin Immunol 2018;38(5):569–78.Google Scholar
Pearson, HA, Lobel, JS, Kocoshis, SA, et al. A new syndrome of refractory sideroblastic anemia with vacuolization of marrow precursors and exocrine pancreatic dysfunction. J Pediatr 1979;95(6):976–84.CrossRefGoogle ScholarPubMed
Rotig, A, Colonna, M, Bonnefont, JP, et al. Mitochondrial DNA deletion in Pearson’s marrow/pancreas syndrome. Lancet 1989;1(8643):902–3.Google Scholar
Rötig, A, Cormier, V, Blanche, S, et al. Pearson’s marrow-pancreas syndrome: A multisystem mitochondrial disorder in infancy. J Clin Invest 1990;86(5):1601–8.Google Scholar
Shanske, S, Tang, Y, Hirano, M, et al. Identical mitochondrial DNA deletion in a woman with ocular myopathy and in her son with Pearson syndrome. Am J Hum Genet 2002;71(3):679–83.CrossRefGoogle Scholar
Superti-Furga, A, Schoenle, E, Tuchschmid, P, et al. Pearson bone marrow-pancreas syndrome with insulin-dependent diabetes, progressive renal tubulopathy, organic aciduria and elevated fetal haemoglobin caused by deletion and duplication of mitochondrial DNA. Eur J Pediatr 1993;152(1):4450.Google Scholar
Tadiotto, E, Maines, E, Degani, D, et al. Bone marrow features in Pearson syndrome with neonatal onset: A case report and review of the literature. Pediatr Blood Cancer 2018;65(4): 10.1002/pbc.26939.Google Scholar
Roy, NBA, Babbs, C. The pathogenesis, diagnosis and management of congenital dyserythropoietic anaemia type I. Br J Haematol 2019;185(3):436–49.CrossRefGoogle ScholarPubMed
Moreno-Carralero, MI, Horta-Herrera, S, Morado-Arias, M, et al. Clinical and genetic features of congenital dyserythropoietic anemia (CDA). Eur J Haematol 2018;101(3):368–78.Google Scholar
Heimpel, H. Congenital dyserythropoietic anemias: Epidemiology, clinical significance, and progress in understanding their pathogenesis. Ann Hematol 2004;83(10):613–21.Google Scholar
Heimpel, H, Matuschek, A, Ahmed, M, et al. Frequency of congenital dyserythropoietic anemias in Europe. Eur J Haematol 2010;85(1):20–5.Google Scholar
Parez, N, Dommergues, M, Zupan, V, et al. Severe congenital dyserythropoietic anaemia type I: Prenatal management, transfusion support and alpha-interferon therapy. Br J Haematol 2000;110(2):420–3.CrossRefGoogle ScholarPubMed
Kato, K, Sugitani, M, Kawataki, M, et al. Congenital dyserythropoietic anemia type 1 with fetal onset of severe anemia. J Pediatr Hematol Oncol 2001;23(1):63–6.Google Scholar
Chin, HL, Lee, LY, Koh, PL. Fetal-onset congenital dyserythropoietic anemia type 1 due to a novel mutation with severe iron overload and severe cholestatic liver disease. J Pediatr Hematol Oncol 2019;41(1):e51–e3.CrossRefGoogle ScholarPubMed
Liu, S, Liu, YN, Zhen, L, Li, DZ. Fetal-onset congenital dyserythropoietic anemia type 1 due to CDAN1 mutations presenting as hydrops fetalis. Pediatr Hematol Oncol 2018;35(7–8):447–50.CrossRefGoogle ScholarPubMed
Shalev, H, Moser, A, Kapelushnik, J, et al. Congenital dyserythropoietic anemia type I presenting as persistent pulmonary hypertension of the newborn. J Pediatr 2000;136(4):553–5.Google Scholar
Wickramasinghe, SN. Congenital dyserythropoietic anaemias: Clinical features, haematological morphology and new biochemical data. Blood Rev 1998;12(3):178200.Google Scholar
Dgany, O, Avidan, N, Delaunay, J, et al. Congenital dyserythropoietic anemia type I is caused by mutations in codanin-1. Am J Hum Genet 2002;71(6):1467–74.CrossRefGoogle ScholarPubMed
Babbs, C, Roberts, NA, Sanchez-Pulido, L, et al. Homozygous mutations in a predicted endonuclease are a novel cause of congenital dyserythropoietic anemia type I. Haematologica 2013;98(9):1383–7.CrossRefGoogle Scholar
Heimpel, H, Schwarz, K, Ebnöther, M, et al. Congenital dyserythropoietic anemia type I (CDA I): Molecular genetics, clinical appearance, and prognosis based on long-term observation. Blood 2006;107(1):334–40.CrossRefGoogle ScholarPubMed
Marwaha, RK, Bansal, D, Trehan, A, Garewal, G. Interferon therapy in congenital dyserythropoietic anemia type I/II. Pediatr Hematol Oncol 2005;22(2):133–8.CrossRefGoogle ScholarPubMed
Bader-Meunier, B, Leverger, G, Tchernia, G, et al. Clinical and laboratory manifestations of congenital dyserythropoietic anemia type I in a cohort of French children. J Pediatr Hematol Oncol 2005;27(8):416–9.CrossRefGoogle Scholar
Rathe, M, Møller, MB, Greisen, PW, Fisker, N. Successful management of transfusion-dependent congenital dyserythropoietic anemia type 1b with interferon alfa-2a. Pediatr Blood Cancer 2018;65(3): e26866.Google Scholar
Bianchi, P, Fermo, E, Vercellati, C, et al. Diagnostic power of laboratory tests for hereditary spherocytosis: A comparison study in 150 patients grouped according to molecular and clinical characteristics. Haematologica 2012;97(4):516–23.Google Scholar
Iolascon, A, Heimpel, H, Wahlin, A, Tamary, H. Congenital dyserythropoietic anemias: Molecular insights and diagnostic approach. Blood 2013;122(13):2162–6.Google Scholar
Ayas, M, al-Jefri, A, Baothman, A, et al. Transfusion-dependent congenital dyserythropoietic anemia type I successfully treated with allogeneic stem cell transplantation. Bone Marrow Transplant 2002;29(8):681–2.CrossRefGoogle ScholarPubMed
Donadieu, J, Fenneteau, O, Beaupain, B, Mahlaoui, N, Chantelot, CB. Congenital neutropenia: Diagnosis, molecular bases and patient management. Orphanet J Rare Dis 2011;6:26.Google Scholar
Dale, DC. How I manage children with neutropenia. Br J Haematol 2017;178(3):351–63.Google Scholar
Rosenberg, PS, Zeidler, C, Bolyard, AA, et al. Stable long-term risk of leukaemia in patients with severe congenital neutropenia maintained on G-CSF therapy. Br J Haematol 2010;150(2):196–9.Google Scholar
Xia, J, Bolyard, AA, Rodger, E, et al. Prevalence of mutations in ELANE, GFI1, HAX1, SBDS, WAS and G6PC3 in patients with severe congenital neutropenia. Br J Haematol 2009;147(4):535–42.CrossRefGoogle ScholarPubMed
Donadieu, J, Leblanc, T, Bader Meunier, B, et al. Analysis of risk factors for myelodysplasias, leukemias and death from infection among patients with congenital neutropenia: Experience of the French Severe Chronic Neutropenia Study Group. Haematologica 2005;90(1):4553.Google Scholar
Nanua, S, Murakami, M, Xia, J, et al. Activation of the unfolded protein response is associated with impaired granulopoiesis in transgenic mice expressing mutant Elane. Blood 2011;117(13):3539–47.Google Scholar
Nayak, RC, Trump, LR, Aronow, BJ, et al. Pathogenesis of ELANE-mutant severe neutropenia revealed by induced pluripotent stem cells. J Clin Invest 2015;125(8):3103–16.CrossRefGoogle ScholarPubMed
Nasri, M, Ritter, M, Mir, P, et al. CRISPR/Cas9 mediated ELANE knockout enables neutrophilic maturation of primary hematopoietic stem and progenitor cells and induced pluripotent stem cells of severe congenital neutropenia patients. Haematologica 2020;105(3):598609.CrossRefGoogle ScholarPubMed
Donadieu, J, Beaupain, B, Fenneteau, O, Bellanné-Chantelot, C. Congenital neutropenia in the era of genomics: Classification, diagnosis, and natural history. Br J Haematol 2017;179(4):557–74.Google Scholar
Yılmaz Karapınar, D, Patıroğlu, T, Metin, A, et al. Homozygous c.130–1 ins A (pW44X) mutation in the HAX1 gene as the most common cause of congenital neutropenia in Turkey: Report from the Turkish Severe Congenital Neutropenia Registry. Pediatr Blood Cancer 2019;66(10):e27923.CrossRefGoogle ScholarPubMed
Dale, DC, Bolyard, AA, Schwinzer, BG, et al. The Severe Chronic Neutropenia International Registry: 10-year follow-up report. Support Cancer Ther 2006;3(4):220–31.Google Scholar
Touw, IP. Game of clones: The genomic evolution of severe congenital neutropenia. Hematology Am Soc Hematol Educ Program 2015;2015:17.Google Scholar
Fioredda, F, Iacobelli, S, van Biezen, A, et al. Stem cell transplantation in severe congenital neutropenia: An analysis from the European Society for Blood and Marrow Transplantation. Blood 2015;126(16):1885–92; quiz 970.CrossRefGoogle ScholarPubMed
Dale, DC, Welte, K. Cyclic and chronic neutropenia. Cancer Treat Res 2011;157:97108.Google Scholar
Ginzberg, H, Shin, J, Ellis, L, et al. Shwachman syndrome: Phenotypic manifestations of sibling sets and isolated cases in a large patient cohort are similar. J Pediatr 1999;135(1):81–8.CrossRefGoogle Scholar
Goobie, S, Popovic, M, Morrison, J, et al. Shwachman-Diamond syndrome with exocrine pancreatic dysfunction and bone marrow failure maps to the centromeric region of chromosome 7. Am J Hum Genet 2001;68(4):1048–54.Google Scholar
Myers, KC, Bolyard, AA, Otto, B, et al. Variable clinical presentation of Shwachman–Diamond syndrome: update from the North American Shwachman–Diamond Syndrome Registry. J Pediatr 2014;164(4):866–70.CrossRefGoogle ScholarPubMed
Boocock, GR, Morrison, JA, Popovic, M, et al. Mutations in SBDS are associated with Shwachman-Diamond syndrome. Nat Genet 2003;33(1):97101.Google Scholar
Finch, AJ, Hilcenko, C, Basse, N, et al. Uncoupling of GTP hydrolysis from eIF6 release on the ribosome causes Shwachman-Diamond syndrome. Genes Dev 2011;25(9):917–29.Google Scholar
Carapito, R, Konantz, M, Paillard, C, et al. Mutations in signal recognition particle SRP54 cause syndromic neutropenia with Shwachman–Diamond-like features. J Clin Invest 2017;127(11):4090–103.Google Scholar
Dhanraj, S, Matveev, A, Li, H, et al. Biallelic mutations in DNAJC21 cause Shwachman–Diamond syndrome. Blood 2017;129(11):1557–62.Google Scholar
Tan, S, Kermasson, L, Hoslin, A, et al. EFL1 mutations impair eIF6 release to cause Shwachman-Diamond syndrome. Blood 2019;134(3):277–90.Google Scholar
Dror, Y, Donadieu, J, Koglmeier, J, et al. Draft consensus guidelines for diagnosis and treatment of Shwachman–Diamond syndrome. Ann N Y Acad Sci 2011;1242:4055.Google Scholar
Keogh, SJ, McKee, S, Smithson, SF, Grier, D, Steward, CG. Shwachman–Diamond syndrome: A complex case demonstrating the potential for misdiagnosis as asphyxiating thoracic dystrophy (Jeune syndrome). BMC Pediatr 2012;12:48.Google Scholar
Rosenberg, PS, Alter, BP, Bolyard, AA, et al. The incidence of leukemia and mortality from sepsis in patients with severe congenital neutropenia receiving long-term G-CSF therapy. Blood 2006;107(12):4628–35.Google Scholar
Link, DC. Mechanisms of leukemic transformation in congenital neutropenia. Curr Opin Hematol 2019;26(1):3440.Google Scholar
Toiviainen-Salo, S, Pitkänen, O, Holmström, M, et al. Myocardial function in patients with Shwachman–Diamond syndrome: Aspects to consider before stem cell transplantation. Pediatr Blood Cancer 2008;51(4):461–7.CrossRefGoogle ScholarPubMed
Chandler, KE, Kidd, A, Al-Gazali, L, et al. Diagnostic criteria, clinical characteristics, and natural history of Cohen syndrome. J Med Genet 2003;40(4):233–41.CrossRefGoogle ScholarPubMed
Kishnani, PS, Austin, SL, Abdenur, JE, et al. Diagnosis and management of glycogen storage disease type I: a practice guideline of the American College of Medical Genetics and Genomics. Genet Med 2014;16(11):e1.Google Scholar
Bachelerie, F. CXCL12/CXCR4-axis dysfunctions: Markers of the rare immunodeficiency disorder WHIM syndrome. Dis Markers 2010;29(3–4):189–98.Google Scholar
Kawai, T, Malech, HL. WHIM syndrome: Congenital immune deficiency disease. Curr Opin Hematol 2009;16(1):20–6.Google Scholar
McDermott, DH, Pastrana, DV, Calvo, KR, et al. Plerixafor for the treatment of WHIM syndrome. N Engl J Med 2019;380(2):163–70.Google Scholar
Mamrak, NE, Shimamura, A, Howlett, NG. Recent discoveries in the molecular pathogenesis of the inherited bone marrow failure syndrome Fanconi anemia. Blood Rev 2017;31(3):93–9.Google Scholar
Fiesco-Roa, MO, Giri, N, McReynolds, LJ, Best, AF, Alter, BP. Genotype-phenotype associations in Fanconi anemia: A literature review. Blood Rev 2019;37:100589.Google Scholar
Alter, BP, Giri, N. Thinking of VACTERL-H? Rule out Fanconi Anemia according to PHENOS. Am J Med Genet A 2016;170(6):1520–4.Google Scholar
Petryk, A, Kanakatti Shankar, R, Giri, N, et al. Endocrine disorders in Fanconi anemia: Recommendations for screening and treatment. J Clin Endocrinol Metab 2015;100(3):803–11.Google Scholar
Kutler, DI, Singh, B, Satagopan, J, et al. A 20-year perspective on the International Fanconi Anemia Registry (IFAR). Blood 2003;101(4):1249–56.Google Scholar
Brosh, RM, Bellani, M, Liu, Y, Seidman, MM. Fanconi anemia: A DNA repair disorder characterized by accelerated decline of the hematopoietic stem cell compartment and other features of aging. Ageing Res Rev 2017;33:6775.Google Scholar
Rosenberg, PS, Alter, BP, Ebell, W. Cancer risks in Fanconi anemia: Findings from the German Fanconi Anemia Registry. Haematologica 2008;93(4):511–7.Google Scholar
Alter, BP, Giri, N, Savage, SA, Rosenberg, PS. Cancer in the National Cancer Institute inherited bone marrow failure syndrome cohort after fifteen years of follow-up. Haematologica 2018;103(1):30–9.Google Scholar
Alter, BP, Rosenberg, PS, Brody, LC. Clinical and molecular features associated with biallelic mutations in FANCD1/BRCA2. J Med Genet 2007;44(1):19.Google Scholar
Reid, S, Schindler, D, Hanenberg, H, et al. Biallelic mutations in PALB2 cause Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet 2007;39(2):162–4.Google Scholar
Gueiderikh, A, Rosselli, F, Neto, JBC. A never-ending story: The steadily growing family of the FA and FA-like genes. Genet Mol Biol 2017;40(2):398407.Google Scholar
Lo Ten Foe, JR, Kwee, ML, Rooimans, MA, et al. Somatic mosaicism in Fanconi anemia: Molecular basis and clinical significance. Eur J Hum Genet 1997;5(3):137–48.Google Scholar
Gross, M, Hanenberg, H, Lobitz, S, et al. Reverse mosaicism in Fanconi anemia: Natural gene therapy via molecular self-correction. Cytogenet Genome Res 2002;98(2–3):126–35.Google Scholar
Hira, A, Yabe, H, Yoshida, K, et al. Variant ALDH2 is associated with accelerated progression of bone marrow failure in Japanese Fanconi anemia patients. Blood 2013;122(18):3206–9.Google Scholar
Ebens, CL, MacMillan, ML, Wagner, JE. Hematopoietic cell transplantation in Fanconi anemia: Current evidence, challenges and recommendations. Expert Rev Hematol 2017;10(1):8197.CrossRefGoogle ScholarPubMed
Dufour, C. How I manage patients with Fanconi anaemia. Br J Haematol 2017;178(1):3247.Google Scholar
Calado, RT, Clé, DV. Treatment of inherited bone marrow failure syndromes beyond transplantation. Hematology Am Soc Hematol Educ Program 2017;2017(1):96101.Google Scholar
Paustian, L, Chao, MM, Hanenberg, H, et al. Androgen therapy in Fanconi anemia: A retrospective analysis of 30 years in Germany. Pediatr Hematol Oncol 2016;33(1):512.Google Scholar
Río, P, Navarro, S, Wang, W, et al. Successful engraftment of gene-corrected hematopoietic stem cells in non-conditioned patients with Fanconi anemia. Nat Med 2019;25(9):1396–401.Google Scholar
Glousker, G, Touzot, F, Revy, P, Tzfati, Y, Savage, SA. Unraveling the pathogenesis of Hoyeraal–Hreidarsson syndrome, a complex telomere biology disorder. Br J Haematol 2015;170(4):457–71.Google Scholar
Hoyeraal, HM, Lamvik, J, Moe, PJ. Congenital hypoplastic thrombocytopenia and cerebral malformations in two brothers. Acta Paediatr Scand 1970;59(2):185–91.Google Scholar
Knight, SW, Heiss, NS, Vulliamy, TJ, et al. Unexplained aplastic anaemia, immunodeficiency, and cerebellar hypoplasia (Hoyeraal–Hreidarsson syndrome) due to mutations in the dyskeratosis congenita gene, DKC1. Br J Haematol 1999;107(2):335–9.Google Scholar
Revesz, T, Fletcher, S, al-Gazali, LI, DeBuse, P. Bilateral retinopathy, aplastic anaemia, and central nervous system abnormalities: A new syndrome? J Med Genet 1992;29(9):673–5.Google Scholar
Niewisch, MR, Savage, SA. An update on the biology and management of dyskeratosis congenita and related telomere biology disorders. Expert Rev Hematol 2019;12(12):1037–52.Google Scholar
Vulliamy, TJ, Marrone, A, Knight, SW, et al. Mutations in dyskeratosis congenita: Their impact on telomere length and the diversity of clinical presentation. Blood 2006;107(7):2680–5.Google Scholar
Alter, BP, Giri, N, Savage, SA, Rosenberg, PS. Telomere length in inherited bone marrow failure syndromes. Haematologica 2015;100(1):4954.Google Scholar
Alter, BP, Giri, N, Savage, SA, Rosenberg, PS. Cancer in dyskeratosis congenita. Blood 2009;113(26):6549–57.Google Scholar
Islam, A, Rafiq, S, Kirwan, M, et al. Haematological recovery in dyskeratosis congenita patients treated with danazol. Br J Haematol 2013;162(6):854–6.Google Scholar
Khincha, PP, Wentzensen, IM, Giri, N, Alter, BP, Savage, SA. Response to androgen therapy in patients with dyskeratosis congenita. Br J Haematol 2014;165(3):349–57.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
×