<<

Pediatric hematology Inherited marrow failure syndromes

I. Dokal ABSTRACT The inherited/constitutional (BM) failure syndromes are a diverse group of disorders Centre for Paediatrics, characterized by BM failure usually in association with one or more extra-hematopoietic abnormality. Blizard Institute, The BM failure, which can involve all or a single lineage, often presents in childhood but this may not Barts and The London School of be until adulthood in some cases. Furthermore, some patients initially labeled as “idiopathic aplastic Medicine and Dentistry, ” are cryptic presentations of these genetic syndromes. Significant advances in the genetics of Queen Mary University of London, these syndromes have been made with more than fifty disease identified to date. These Barts Health NHS Trust, advances have provided an insight into normal hematopoiesis and how this is disrupted in patients London with BM failure. They have also provided important information on fundamental biological pathways: DNA repair-Fanconi genes; maintenance-dyskeratosis congenita genes; ribosome biogene - Correspondence: sis-Shwachman Diamond syndrome and Diamond-Blackfan anemia genes. Additionally, as these dis - Inderjeet Dokal orders are usually associated with developmental abnormalities and an increased risk of , they E-mail: [email protected] are providing an insight into human development and the genesis of cancer. In the clinic, genetic tests Acknowledgments stemming from the recent advances are facilitating diagnosis and personalized management. I would like to thank my current col - Hematopoietic stem cell transplantation using fludarabine-based protocols has improved outcomes leagues (Laura Collopy, Upal significantly; management of some of the other complications remains a challenge. Hossain, Hemanth Tummala, Tom Learning goals Vulliamy and Amanda Walne) and past colleagues (Richard Beswick, At the conclusion of this activity, participants should know that: Michael Kirwan, Stuart Knight, Anna - the inherited BM failure syndromes are a very heterogeneous group of disorders characterized by Marrone, Philip Mason and David inability to make an adequate number of mature blood cells, usually in association with one or Stevens) whose contribution has more extra-hematopoietic abnormalities; been important to our research pro - - more than fifty disease genes have been identified and these have provided an important insight gram over the years. Hemanth into basic cell biology and hematopoiesis; Tummala provided useful comments - the genetic advances are facilitating accurate diagnosis and personalized management; on the manuscript. I am also grate - - hematopoietic stem cell transplantation using low-intensity fludarabine-based protocols is asso - ful to the patients and all our col - ciated with low toxicity and greater efficacy. leagues (doctors and nurses) for their support and to The Wellcome Trust and Medical Research Council for financial support over the years. Introduction atresia) and neurological abnormalities. Approximately 30% of FA patients have no Hematology Education: overt somatic abnormalities. The majority of the education program for the The inherited bone marrow (BM) failure syndromes are a diverse group of life-threaten - patients present towards the end of the first annual congress of the European 1 decade of life. However, increasingly some Hematology Association ing disorders. The features of the recognized inherited BM syndromes are summarized in patients are being diagnosed in adulthood and 2014;8:299-308 Tables 1 and 2. An outline of each syndrome is many patients diagnosed in childhood are sur - given below. viving into adulthood. FA cells display a high frequency of sponta - neous chromosomal breakage and hypersensi - tivity to DNA cross-linking agents such as diepoxybutane and mitomycin-C. This “FA Fanconi anemia (FA) was first described by cell hallmark” led to the development of a 2 Fanconi in 1927. It is usually inherited as an diagnostic test over two decades ago and has autosomal recessive (AR) trait, but in a small facilitated many advances, including clarifica - subset of patients it can be an X-linked reces - tion of the genetics, with 16 subtypes/comple - sive (XLR) disorder. FA patients are clinically mentation groups currently characterized. 4-19 3 heterogeneous. Characteristic features The encoded by the FA genes (Table include the progressive development of BM 2) participate in a complicated network impor - failure and an increased predisposition to tant in DNA repair. 20-22 Specifically, eight of malignancy (hematologic and non-hematolog - the FA proteins (FANCA, FANCB, FANCC, ic). Affected individuals may also have one or FANCE, FANCF, FANCG, FANCL and more developmental/somatic abnormality FANCM) interact with each other and form a including skin (e.g. cafe au lait spots), skeletal nuclear complex called the “FA core com - (e.g. radial hypoplasia), genitourinary (e.g. plex”. The FA core complex is required for the single kidney), gastrointestinal (e.g. duodenal activation of the FANCI-FANCD2 Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) | 299 | 19 th Congress of the European Hematology Association

complex to a monoubiquitinated form (FANCI-FANCD2- highly conserved nucleolar protein called . Ub). FANCI-FANCD2-Ub then interacts with DNA repair Dyskerin associates with the H/ACA class of small nucle - proteins (including BRCA2, BRCA1 and RAD51) leading olar RNAs in small nucleolar ribonucleoprotein particles to repair of the DNA damage. FA-D1 patients have biallel - (snoRNPs), which are important in guiding the conversion ic in BRCA2. These observations have linked of uracil to pseudouracil during the maturation of riboso - the FA proteins with BRCA1 and BRCA2 (FANCD1) in a mal RNA. Dyskerin also associates with the RNA compo - DNA damage response pathway: “the FA/BRCA path - nent of (TERC) where it is important in stabi - way”. The BRCA2 protein is important in the repair of lizing the telomerase complex, which is critical in the DNA damage by homologous recombination. Cells lack - maintenance of . 42-44 Heterozygous mutations in ing BRCA2 inaccurately repair damaged DNA and are TERC and TERT (telomerase ) have hypersensitive to DNA cross-linking agents. It has been been found in patients with AD-DC 29-31 and in some established that FANCJ is BRIP1 (partner of BRCA1), patients with aplastic anemia (AA), myelodysplasia FANCN is PALB2 (partner of BRCA2), and that SLX4 is (MDS), acute leukemia, pulmonary and liver fibrosis. 45-50 also a FA protein. These findings further strengthen the A subset of patients with the multi-system disorder connection between FA and DNA repair; specifically it Hoyeraal-Hreidarsson (HH) syndrome have been found to appears that the FA pathway orchestrates incisions at sites have DKC1 mutations. 51 It has also been established that of cross-linked DNA. 21 Recent studies suggest the FA pro - AR-DC is genetically heterogeneous with seven subtypes teins may be important in counteracting aldehyde-induced due to biallelic mutations in NHP2 , NOP10 , TERT, genotoxicity in hematopoietic stem cells. 23-24 In addition to TCAB1 , USB1 , CTC1 and RTEL1 . One AD-DC subtype their role in DNA repair, they also have other functions. was found to be due to mutations in TINF2 that encodes a For example, there is evidence of mitochondrial dysfunc - component of the shelterin complex that protects telom - tion and impaired ROS (reactive oxygen species) detoxi - eres and controls access of telomerase to the telomere. fying machinery in FA cells. 25 Collectively, these findings have demonstrated that classi - cal DC, HH, a subset of AA and MDS/AML are principal - ly due to a defect in telomere maintenance, and cells from Dyskeratosis congenita these patients have very short/and or abnormal telom - eres. 42,52 The multi-system abnormalities seen in these Classical dyskeratosis congenita (DC), first described in patients, including the increased incidence of malignancy, 1910, is an inherited BM failure syndrome characterized have highlighted the critical role of telomeres and led to by the muco-cutaneous triad of abnormal skin pigmenta - 26,27 the recognition of a new category of human diseases: “the tion, dystrophy and mucosal . A variety telomereopathies”. of other abnormalities have also been reported: dental (e.g. severe caries), gastrointestinal (e.g. esophageal stenosis), genitourinary (e.g. phimosis), neurological (e.g. cerebellar Shwachman-Diamond syndrome hypoplasia), ophthalmic (e.g. naso-lacrymal duct narrow - ing), pulmonary (e.g. fibrosis) and skeletal (e.g. osteo - Shwachman-Diamond syndrome (SDS), first described porosis). BM failure is the major cause of mortality with in 1964, is an AR disorder characterized by exocrine pan - an additional predisposition to malignancy (hematologic creatic insufficiency, BM failure and other somatic abnor - and non-hematologic) and fatal pulmonary complications. malities (particularly metaphyseal dysostosis). 53-54 XLR, autosomal dominant (AD) and AR subtypes of DC Features of pancreatic insufficiency are apparent early in are recognized. Ten DC genes ( DKC1 , TERC , TERT , infancy. The spectrum of hematologic abnormalities NOP10 , NHP 2, TINF2 , TCAB1 , USB1 , CTC1 and includes neutropenia, (approx. 20%), MDS RTEL1 )28-41 have been identified to date. and leukemia (approx. 25%). The majority (>90%) of SDS The mutated in X-linked DC ( DKC1 ) encodes a patients have biallelic mutations in the SBDS gene. 55 The

Table 1. Characteristics of the inherited bone marrow failure syndromes.

FA DC SDS DBA CDA CAMT SCN Other Inheritance pattern AR, XLR XLR, AR AR AD AR AR AD AR, AD AD AD AR Somatic abnormalities Yes Yes Yes Yes Rare Rare Rare Rare Bone marrow failure AA (90%) AA (80%) AA (20%) RCA a Dysery Meg b Neut c AA Short telomeres Yes Yes d Yes No No No ?? Cancer Yes Yes Yes Yes No Yes Yes ?Yes Chromosome instability Yes Yes Yes ?? No ?? Genes identified 16 10 1 12 41 5 3 FA: Fanconi anemia; DC: dyskeratosis congenita; SDS: Shwachman-Diamond syndrome; DBA: Diamond-Blackfan anemia; CDA: congenital dyserythropoietic anemia; CAMT: congenital amegakaryocytic ; SCN: severe congenital neutropenia; AD: autosomal dominant; AR: autosomal recessive; XLR: X-linked recessive; RCA a: red cell aplasia although some patients can develop global BM failure; Dysery: usually dyserythro - poiesis; Meg b: low megakaryocytes which can progress to global BM failure; Neut C: usually low neutrophils; Yes d: usually very short.

| 300 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) Milan, Italy, June 12-15, 2014

Table 2. Inherited BM failure genetic subtypes.

(i) FA Complementation group/gene Approximate % of FA patients Chromosome location Gene product Exons A ( FANCA ) 65 16q24.3 FANCA 43 B ( FANCB a) <1 Xp22.2 FANCB 10 C ( FANCC ) 12 9q22.3 FANCC 14 D1 ( FANCD1 b) <1 13q12.3 FANCD1 27 D2 ( FANCD2 ) <1 3p25.3 FANCD2 44 E ( FANCE )46p21.3 FANCE 10 F ( FANCF )411p15 FANCF 1 G ( FANCG ) 12 9p13 FANCG 14 I ( FANCI ) <1 15q26.1 FANCI 35 J ( FANCJ/BRIP1 c) <5 17q23.1 FANCJ 20 L ( FANCL ) <1 2p16.1 FANCL 14 M ( FANCM ) <1 14q21.3 FANCM 23 N ( FANCN/PALB2 d) <1 16p12.1 FANCN 13 O ( FANCO/RAD51C ) <1 17q25.1 FANCO 9 P ( FANCP/SLX4 )216p13.3 FANCP 15 Q ( FANCQ/ERCC4 ) <1 16p13.12 FANCQ 11 aFANCB is on the X-chromosome; bFANCD1 is BRCA2; cFANCJ is BRIP1 (partner of BRCA1); dFANCN is PALB2 (partner of BRCA2). (ii) DC DC subtype Approximate % of DC patients Chromosome location Gene product Exons X-linked recessive 25 Xq28 DKC1 (dyskerin) 15 Autosomal dominant 5 3q26 TERC 1 3 5p15 TERT 16 12 14q11 TIN2 6 Autosomal recessive <1 15q14 NOP10 2 <1 5p15 TERT 16 <1 5q35 NHP2 4 <1 17p13.1 TCAB1 13 <2 16q13 USB1 7 <1 17p13.1 CTC1 23 2 20q13.3 RTEL1 35 Uncharacterized >30 ??? Data based on the Dyskeratosis Congenita Registry in London. (iii) SDS SDS subtype Approximate % of SDS patients Chromosome location Gene product Exons Autosomal recessive >90 7q11 SBDS 5 Uncharacterized <10 ??? (iv) DBA DBA subtype Approximate % of DBA patients Chromosome location Gene product Exons Autosomal dominant 25 19q13.2 RPS19 6 2 10q22-23 RPS24 6 1 15q25.2 RPS17 5 7 1p22.1 RPL5 8 5 1p35-36.1 RPL11 6 3 3q29 RPL35A 6 1 2p RPS7 7 7 6q21.31 RPS10 6 3 12q13.2 RPS26 4 1 17p13.1 RPL26 4 <1 3p24.2 RPL15 4 X-linked recessive <1 Xp11.23 GATA1 6 Uncharacterized ~30 ???

Continued on next page

Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) | 301 | 19 th Congress of the European Hematology Association

Table 2. Continued from previous page.

(v) CDA CDA subtype Approximate % of CDA patients Chromosome location Gene product Exons Type I (AR) Majority 15q15.2 CDAN1 15 Subset 15q14 C15ORF41 11 Type II (AR) Majority 20p11.23 SEC23B 8 Type III (AD) Majority 15q21 KIF23 23 Other variants 19p13.2 KLF1 3 (vi) CAMT CAMT subtype Approximate % of CAMT patients Chromosome location Gene product Exons Autosomal recessive Majority 1p34 MPL 12 Uncharacterized ???? (vii): SCN Subtype Approximate % of patients Chromosome location Gene product Exons Autosomal dominant 50-60 19p13.3 ELANE 6 < 2 1p22 GFI1 3 Autosomal recessive 15 1q21.3 HAX1 7 ?5 17q21.31 G6PC3 6 Rare 1q21.2 VPS45 4 Miscellaneous syndromes* *This is a heterogeneous group and includes patients with neutropenia as part of a broader syndrome such as Wiskot-Aldrich syndrome (WAS) and Shwachman-Diamond syndrome (SDS).

SBDS gene product (SBDS) has an important role in the with heterozygous RPL11 mutations. Interestingly, a rare maturation of the 60S ribosomal subunit and, therefore, DBA subgroup has been recently found to be due to ribosome biogenesis. 56 SDS is thus principally a disorder GATA1 (encoding an erythroid transcriptional factor) of defective ribosome biogenesis. mutations, 67 highlighting the heterogeneity in the molecu - lar pathways that can give rise to a DBA . In Japanese populations, RPS19 mutations only account Diamond-Blackfan anemia for approximately 13% of DBA patients and there are also some differences in the clinical associated Diamond-Blackfan anemia (DBA), initially described in 57 with the different DBA genes compared to Western popu - 1934, usually presents in early infancy, with features of 68 58 lations. This suggests ethnic differences in phenotypic anemia. The hallmark of classical DBA is a selective expression, a feature that has been observed previously in decrease in erythroid precursors (reduced reticulocyte other genetic diseases, including FA. count) and normochromic macrocytic anemia associated with a variable number of somatic abnormalities such as craniofacial (e.g. high arched palate), thumb, cardiac and Congenital dyserythropietic urogenital malformations. MDS and AML have been reported in a few patients, suggesting an increased predis - The congenital dyserythropietic anemias (CDAs) com - position to hematologic cancer. There are also cases that prise a heterogeneous group of inherited disorders of ery - have evolved into AA. Thus, although DBA has been thropoiesis characterized by anemia, ineffective erythro - regarded classically as a pure red cell aplasia, a more glob - poiesis and morphological evidence of dyserythropoei - al hematopoietic defect can be observed in some patients. sis. 69,70 The first description of this group of disorders was The first DBA gene ( RPS19 ) was identified in 1999 59 in 1966 by Crookston and colleagues. In 1968, Wendt and and in Western populations accounts for approximately Heimpel classified the CDAs into three types: I, II and III. 25% of DBA patients. Subsequently heterozygous muta - Over the years, many additional subtypes (IV, V, VI and tions in other genes encoding proteins of the small VII) have been added to the list, often based on case report (RPS24, RPS17, RPS7, RPS10, RPS26) and large (RPL5, studies. With the emerging genetics it will be interesting to RPL11, RPL15, RPL26, RPL35A) ribosomal subunits see how this classification evolves. have also been reported; collectively the genetic basis of CDA type 1: the majority of patients present with approximately 60-70% of DBA patients can now be estab - splenomegaly and mild to moderate anemia (usually lished. 60-66 These observations have also demonstrated that macrocytic). In some cases, non-hematologic features DBA is principally a disorder of ribosome biogenesis. It is (e.g. skeletal abnormalities, abnormal skin pigmentation) noteworthy that patients with mutations in the RPL5 gene have been observed. Ineffective erythropoiesis is evi - tend to have multiple physical abnormalities, including denced by peripheral (anisocytosis) and marrow (mega - craniofacial, thumb and heart anomalies, whereas isolated loblastic erythroid precursors, internuclear chromatin thumb malformations are predominantly seen in patients bridging and binuclearity affecting 3-7% of erythroblasts)

| 302 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) Milan, Italy, June 12-15, 2014 abnormalities as well as increased markers of hemolysis ated with a variable number of non-hematologic abnor - (elevated LDH and bilirubin). The defining ultrastructural malities. It is also important to note there are a number of feature is a spongy (‘Swiss cheese’) appearance of the het - other syndromes (reviewed by Hauck and Klein 77 ) that erochromatin in the majority of erythroblasts on electron have neutropenia as part of the broader syndrome. microscopy (EM). Recognized to be an AR disorder, the first gene ( CDAN1 ) responsible for CDA type 1 was iden - 71 Congenital amegakaryocytic thrombocytopenia tified in 2002. Recently, a second gene, C15ORF41 , was found to be responsible for some cases of CDA type 1. 72 CDA type II: this is the most common subtype of CDA Congenital amegakaryocytic thrombocytopenia and was initially described as hereditary erythroblastic (CAMT) usually presents in infancy and is characterized multinuclearity with a positive acidified serum lysis test by isolated thrombocytopenia, reduction/absence of (HEMPAS) in 1969. It is inherited as an AR trait. The ane - megakaryocytes in the BM with usually no somatic abnor - mia is variable (8-11 g/dL); approximately 10% of cases malities. Approximately 50% of patients will develop AA, require regular transfusions. The clinical presentations usually by the age of five years. They can also evolve into include a variable degree of jaundice, hepatomegaly and MDS or leukemia. CAMT patients have biallelic muta - tions in the gene ( MPL ) encoding the thrombopoietin splenomegaly, and cirrhosis. Mental retardation has been 83 reported in some cases. Peripheral blood morphology receptor. shows moderate to marked red cell anisocytosis. BM fea - tures include normoblastic erythroid hyperplasia with usu - Other subtypes of inherited bone marrow failure ally more than 10% binucleate erythroblasts. At the EM syndromes level, the erythroid cells have characteristic peripheral arrangement of the endoplasmic reticulum giving the There are cases of BM failure that are familial and/or appearance of ‘double membrane’. Red cells are hemol - have one or more extra-hematopoietic abnormality but ysed by some acidified sera but not by the patients own which do not fit into the recognized entities listed above. serum. In 2009, the gene encoding the secretory COPII The recent availability of next generation sequencing tech - component SEC23B was shown to be responsible for 73 nology is making it possible to clarify the genetic basis CDAII. and pathophysiology of these cases. Examples of such CDA type III: this subtype is rare. In one of the largest cases that have been recently characterized include the (Swedish) families investigated, the disease was character - familial aplastic anemia associated with constitutional ized by giant multinucleated erythroblasts in the marrow. thrombopoietin receptor (MPL), thrombopoietin ligand There appears to be an increased prevalence of lympho - and SRP72 mutations. 84-86 It is likely that more such cases proliferative disorders in CDA type III. CDA III exhibits of familial/constitutional BM failure will be characterized AD transmission and is caused by mutations in the KIF23 74 in the near future. They are also likely to provide new gene. KIF23 encodes mitotic kinesin-like protein 1, insights into hematopoiesis and how this might be disrupt - which plays a critical role in cytokinesis during cell divi - ed in BM failure. sion. The functional role of the proteins encoded by CDAN1 , C15ORF41 and SEC23B in CDA disease pheno - type remains unknown. 70 CDA variants due to mutations Epidemiology in erythroid factor genes ( GATA-1 and KLF1 )75 have also been recently identified. The true incidence and natural history of these disorders remains largely unknown. It is generally believed that SCN and DBA are amongst the most prevalent of these Severe congenital neutropenia (including disorders; the estimated annual birth incidence of DBA is Kostmann syndrome) 5 cases per 10 6. Tamary et al. reported on a retrospective population-based registry of inherited BM failure syn - Very severe peripheral neutropenia (frequently 87 9 dromes in Israel. One hundred and twenty-seven patients <0.2x10 /L) characterizes severe congenital neutropenia diagnosed between 1966 and 2007 were registered; 52% 76,77 (SCN). These patients usually present with recurrent had FA, 17% SCN, 14% DBA, 6% CAMT, 5% DC, 2% life-threatening infections in infancy. BM examination SDS, and 2% thrombocytopenia with absent radii. This typically shows a maturation arrest in the myeloid lineage. report represented the first comprehensive population- These patients can progress to MDS and leukemia, usually based study evaluating the incidence and complications of with acquisition of secondary mutations, including in the the different inherited BM failure syndromes. The most G-CSF receptor. In the majority of patients, heterozygous common disease was FA, which also carried the worst mutations in neutrophil elastase gene ( ELANE ) have been prognosis, with severe BM failure and cancer develop - 78 demonstrated. These mutations are thought to lead to ment. It is noteworthy that these data are probably only accumulation of the non-functional protein which in turn relevant to Israel. For example, based on the data from this triggers an unfolded protein response leading to matura - registry, the annual incidence of FA was calculated to be tional arrest. The original family described by Kostmann, approximately 2 per 100,000 live births. This is 7-fold had AR SCN, and has been shown to be associated with higher than expected from the world-wide carrier frequen - biallelic mutations in the HAX1 gene, 79 predicted to lead to cy of 1:300 and probably reflects the high rate of consan - defects in cell death. Mutations in other genes ( GFI1, guinity in Israel. G6PC3 and VPS45 )80-82 are also associated with SCN. In a recent report from the Canadian registry, 88 the most Whilst ELANE mutations tend to produce an isolated neu - common disease was DBA followed by FA; this gave an tropenia, mutations in some of the other genes are associ - approximate incidence for FA of 11.4 cases per 10 6 births.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) | 303 | 19 th Congress of the European Hematology Association

Notably, 53.5% of the cases fell into an uncharacterized and avoidance of sunlight is important. Subjects should category. also avoid occupations that expose them to hazardous chemicals or repeated physical trauma. When doing domestic chores such as washing up, use of protective General principles of diagnosis and management gloves is advisable (particularly in DC). Extremes of tem - perature, both hot and cold, should be avoided as the skin A diagnosis of an inherited BM failure should be con - is usually fragile compared to the normal population. sidered when there is one or more extra-hematopoietic Liver disease is more common in FA and DC patients features associated with the BM failure. In the context of than in the normal population. Alcohol consumption children presenting upfront with aplastic anemia (global should, therefore, be kept to a minimum, and all drug trilineage BM failure), a diagnosis of an inherited BM fail - administrations require close monitoring. ure also needs to be on the differential list. The specific Drugs also need to be used carefully, as patients with extra-hematopoietic abnormalities help in diagnosing a inherited BM failure syndromes tend to be small in stature recognized syndrome; however, this is not always possible and more sensitive to many drugs. This is particularly on clinical features alone. important in FA and DC patients undergoing allogeneic The chromosomal breakage analysis test on peripheral hematopoietic stem cell transplantation (SCT). blood lymphocytes following exposure to diepoxybutane or mitomycin-C remains a very useful way of diagnosing FA. All children presenting with AA should be tested for Management of the hematologic complications FA. Genetic testing for the FA genes is possible, but this is not straightforward and not always routinely available. Major advances in supportive treatments 90 have led to Telomere length, particularly using flow fluorescence in considerable improvement in the outcome of these situ hybridization (flow-FISH) can be a very useful initial patients. Blood product support has been a key component screening technique 89 in the diagnosis of DC. Patients with of this effort. Packed red cell transfusions should be given DC frequently (but not always) have very short telomeres to maintain the hemoglobin at a level that allows the compared to age-matched controls. Genetic testing for the patient to be asymptomatic (typically >8 g/dL), platelets DC genes ( DKC1 , TERC , TERT , NOP10 , NHP2 , TINF2 , should be maintained above 10x10 9/L. It is reasonable to USB1 , TCAB1 , CTC1 and RTEL1 ) can be helpful in sub - give prophylactic antimicrobials to patients with a neu - stantiating the diagnosis. However, as in FA, this is not trophil count consistently below 0.5x10 9/L. As a general straightforward (as many patients will have private muta - rule, all neutropenic patients need prompt therapy with tions and some of the genes are large), and in approxi - broad-spectrum intravenous antibiotics if they develop an mately one-third of patients the genetic basis will remain infection. Leuco-depleted and, where appropriate, CMV- unknown even after testing for all 10 DC genes. negative blood products should be chosen to prevent In patients with global BM failure, the other genetic development of HLA-antibodies and reduce the risk of tests to consider are those for the MPL and SBDS genes. CMV disease. For patients presenting with an isolated neutropenia, Inherited BM failure syndromes do respond to specific analysis for the ELANE , GFI1 and HAX1 may help sub - interventions. In patients with FA and DC who have sig - stantiate the underlying diagnosis. For those with an iso - nificant peripheral cytopenia (Hb < 8 g/dL, neutrophils lated anemia, testing for the DBA and CDA genes is indi - <0.5x10 9/L, platelets <20x10 9/L), the first-line medical cated. Once diagnosis of an inherited syndrome has been therapy is usually with oxymetholone that should be start - made (clinically and/or genetically) it is important to ed at a dose of 0.5-1.0 mg/kg/day and gradually increased, explain to the patient/family the chronic nature of these if necessary, to a maximal dose of 5 mg/kg/day. DC disorders. In general, patients need to be followed-up patients are usually more sensitive to oxymetholone than throughout life and will need to be monitored for compli - FA patients. There is also increasing experience in the use cations. The frequency of monitoring investigations (such of danazol in these patients. 91,92 Approximately 70% of as blood tests, BM examinations, pulmonary function patients with DC and FA will have a hematologic response tests) is difficult to define precisely in view of the consid - to anabolic steroids; this can be durable for years in some erable clinical heterogeneity. However, regular (possibly patients. Patients with severe BM failure and with HLA- on an annual basis) follow up is advisable, with more fre - compatible donors can be cured of their hematologic com - quent monitoring (and appropriate investigations) being plications. Over the years, it has been recognized that implemented if there is a specific problem. patients with inherited BM failure syndromes have greater In view of the significant risk of cancer in many of these efficacy and lower toxicity using low-intensity fludara - syndromes, patients should be advised (according to age) bine-based protocols. There is now considerable experi - to avoid active and passive smoking. They should also ence using such protocols in both FA and DC patients, 93-97 avoid sunbathing and keep alcohol intake to a minimum as representing a major advance in outcome following SCT they go through adolescence and adulthood. Treatment for in FA and DC patients. cancer depends on the specific cancer but consideration In patients with DBA, the first-line therapy remains cor - has to be given to the underlying genetic defect (i.e. more ticosteroids (usually prednisolone). Up to 80% of patients supportive care, reduce drug doses). With regards to pul - respond to steroids as a single agent. The dose and fre - monary disease in FA and DC, patients should be encour - quency of prednisolone is titrated to the lowest amount aged to avoid smoking. Medical treatment is usually diffi - required to maintain a hemoglobin response to minimize cult in severe lung disease; lung transplant may be an side-effects. In the minority of patients who are refractory option in some cases. to steroids, or when patients lose steroid responsiveness, Advice on skin care (e.g. use of moisturising creams) supportive treatment with regular red blood transfusions is

| 304 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) Milan, Italy, June 12-15, 2014

Figure 1. Interconnections of molecules mutated in different bone marrow failure syndromes. Images (A-F) are derived using the STRING database: http://string-db.org/newstring_cgi/show_input_page.pl STRING is a database of known and predicted protein interactions. The key protein (red at the center node) and its asso - ciated interactions (blue lines) are achieved through confidence view, which include direct (physical) and indirect (func - tional) associations. Variability in confidence of protein-protein interactions are determined by increasing thickness of blue line annotated. These interactions are derived from four sources such as genomic context, high-throughput experiments, conserved co-expression studies and previous knowledge (databases). (A) In this image FANCA (mutated in FA-A) is used as the reference and shows all its associations with other proteins both FA (FANCB, FANCC, FANCE, FANCF, FANCG, FANCL and FANCM) and non-FA (e.g. BLM). (B) Image is based around DKC1 (mutated in X-linked DC); it associates with several other DC proteins (e.g. NOP10, NHP2, TERT) but also others that are linked with other BM failure syndromes (e.g. EIF6 which has a strong connection with SBDS, shown in E). (C) This image is based around FANCD2. It shows its associations with several FA proteins (FANCA FANCB, FANCC, FANCE, FANCI, and BRCA2) and BRCA1. (D) Associations are based around TINF2 that is mutated in AD-DC. TINF2 interacts with several proteins important in telomere maintenance (POT1, TERF1, and TERF2) but also with BRCA1 (important in DNA repair and in the FA pathway, denoted with **). (E) Associations are focused on SBDS (mutated in SDS). It shows its interesting connection with EIF6 that also interacts with DKC1 (mutated in DC, denoted with *). (F) Image based around RPS19; it shows its associations with several other ribosomal proteins mutated in DBA. Gene names for those mutated as well as for several key interconnecting bone marrow failure proteins are as follows: RP: ribosomal protein; FANC: Fanconi anemia complementation group; DKC1: dyskeratosis congenita 1; NOP10: nucleolar protein 10 homolog; NHP2; non-histone ribonucleoprotein 2 homolog; GAR1: and rich ribonucleoprotein 1 homolog; TERF1: telomeric repeat binding factor 1; TERF2: telomeric repeat binding factor 2; TINF2: TERF1-interacting nuclear factor 2; TERF2IP: TERF2 interacting protein (RAP1); POT1: protection of telomeres 1 homolog; TPP1: TIN2 interacting protein 1 (=ACD, adrenocortical dysplasia homolog); PINX1: PIN2 (=TERF1) interacting protein; ATM: ataxia telangiectasia mutated; MRE1: meiotic recombination 11 homolog; BRCA1: breast cancer 1; BRIP1: BRCA1 interacting protein C-terminal helicase 1; PALB2: partner and localizer of BRCA2; BLM: protein.

Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) | 305 | 19 th Congress of the European Hematology Association instituted. This should be accompanied with a comprehen - Hematology of Infancy and Childhood. DG Nathan, HS Orkin (eds.) WB Saunders, Philadelphia; 1998:237-335. sive iron chelation program to prevent iron overload. At 2. Fanconi G. Familiare infantile pernziosaartige anaemie this stage, for DBA patients who have a compatible sibling (pernizioses Blutbild und Konstitution). Jahrb Kinderheilkd. BM donor, hematopoietic SCT may become appropriate 1927;117:257-80. 3. Auerbach AD, Buchwald M, Joenje H. In: The Metabolic and and is potentially curative. Molecular Basis of Inherited Disease. CR Scriver, et al. (eds.) CDA patients with mild anemia require no major inter - McGraw-Hill, New York; 2001:753-68. vention. Folate supplementation should be given to pre - 4. Strathdee CA, Gavish H, Shannon WR, Buchwald M. Cloning vent folate deficiency. If regular transfusions are neces - of cDNAs for Fanconi’s anaemia by functional complementa - tion. Nature. 1992;356:763-7. sary, early attention to iron chelation is essential. 5. Lo Ten Foe JR, Rooimans MA, Bosnoyan-Collins L, Alon N, Splenectomy may be of benefit in some patients (CDA Wijker M, Parker L, et al. Expression cloning of a cDNA for type II) and there are case reports of successful the major Fanconi anaemia gene, FAA. Nat Genet. 1996;14:320-3. hematopoietic SCT. In CDA type I, there are also case 6. de Winter JP, Waisfisz Q, Rooimans MA, Van Berkel CGM, reports of improvement in the Hb after treatment with a Bosnoyan-Collins L, Alon N, et al. The Fanconi anaemia interferon- The mechanism of this therapeutic benefit group G gene FANCG is identical with XRCC9. Nat Genet. 1998;20:281-3. remains unclear. 7. de Winter JP, Leveille F, van Berkel CG, Rooimans MA, van The mainstay of management of neutropenia in SCN Der Weel L, Steltenpool J, et al. Isolation of a cDNA repre - patients is with G-CSF. More than 90% respond to treat - senting the Fanconi anemia complementation group E gene. ment and the dose is adjusted to maintain an absolute neu - Am J Hum Genet. 2000;67:1306-8. 9 8. de Winter JP, Rooimans, MA, van Der Weel L, van Berkel CG, trophil count of more than 1.5-2.0x10 cells/L. On an aver - Alon N, Bosnoyan-Collins L, et al. The Fanconi anaemia gene age, the G-CSF responsiveness is usually maintained and FANCF encodes a novel protein with homology to ROM. Nat sustained for at least 6-10 years. Other measures to pre - Genet. 2000;24:15-6. 9. Timmers C, Taniguchi T, Hejna J, Reifsteck C, Lucas L, Bruun vent infection are also instituted and any evidence of D, et al. Positional cloning of a novel Fanconi anemia gene, infection should be promptly treated. For patients with FANCD2. Mol Cell. 2001;7:241-8. compatible donors, SCT may be appropriate if they have a 10. Howlett NG, Taniguchi T, Olson S, Cox B, Waisfisz Q, De Die-Smulders C, et al. Biallelic inactivation of BRCA2 in poor response to G-CSF or if there is evolution to Fanconi Anemia. Science. 2002;297:606-9. MDS/leukemia. 11. Meetei AR, de Winter JP, Medhurst AL, Wallisch M, Waisfisz Q, van de Vrugt HJ, et al. A novel ubiquitin ligase is deficient in Fanconi anemia. Nat Genet. 2003;35:165-70. Conclusions 12. Meetei AR, Levitus M, Xue Y, Medhurst AL, Zwaan M, Ling C, et al. X-linked inheritance of Fanconi anemia complemen - tation group B. Nat Genet. 2004;36:1219-24. The significant advances in the molecular basis of the 13. Meetei AR, Medhurst AL, Ling C, Xue Y, Singh TR, Bier P, et al. A human ortholog of archaeal DNA repair protein Hef is inherited BM failure syndromes have provided insights defective in Fanconi Anemia complementation group M. Nat into several biological pathways, such as DNA repair (FA Genet. 2005;37:958-63. genes) and telomere maintenance (DC genes), important 14. Levitus M, Waisfisz Q, Godthelp BC, de Vries Y, Hussain S, in human physiology. They have also provided interesting Wiegant WW, et al. The DNA helicase BRIP1 is defective in Fanconi anemia complementation group J. Nat Genet. connections between inherited (e.g. DBA and SDS) and 2005;37:934-5. acquired (MDS, 5q – syndrome) hematologic disorders by 15. Reid S, Schindler D, Hanenberg H, Barker K, Hanks S, Kalb identifying links in unifying biological pathways (e.g. R, et al. Biallelic mutations in PALB2 cause Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet. ribosome biogenesis). 2007;39:142-3. Clinical similarities (BM failure, developmental anom - 16. Smogorzewska A, Matsuoka S, Vinciguerra P, McDonald ER, alies and cancer) between these syndromes have been Hurov KE, Luo J, et al. Identification of the FANCI protein, a monoubiquitinated FANCD2 paralog required for DNA repair. acknowledged for several years, and it is no surprise that Cell. 2007;129:289-301. some overlap is also observed at the level of molecular 17. Vaz F, Hanenberg H, Schuster B, Barker K, Wiek C, Erven V, pathology. For example, SDS and DBA both appear to be et al. of the RAD51C gene in a Fanconi anemia-like disorder. Nat Genet. 2010;42:406-9. disorders of ribosomal biogenesis; FA, DC and SDS all 18. Stoepker C, Hain K, Schuster B, Hilhorst-Hofstee Y, have short telomeres. It is possible that further Rooimans MA, Steltenpool J, et al. SLX4, a coordinator of overlap/connections in these pathways will emerge. This structure-specific endonucleases, is mutated in a new Fanconi anemia subtype. Nat Genet. 2011;43:138-41. is highlighted by the complex interactions between the dif - 19. Bogliolo M, Schuster B, Stoepker C, Derkunt B, Su Y, Raams ferent molecules encoded by the genes mutated in the BM A, et al. Mutations in ERCC4, encoding the DNA-repair failure syndromes (Figure 1). For example, many of the endonuclease XPF, cause Fanconi anemia. Am J Hum Genet. FA proteins interact with BRCA1, and BRCA1 also inter - 2013;92:800-6. 20. Wang W. Emergence of a DNA-damage response network acts with several components involved in telomere main - consisting of Fanconi anaemia and BRCA proteins. Nat Rev tenance that are important in DC. The genetic advances Genet. 2007;6:735-48. have already led to improved diagnosis (particularly 21. Crossa GP, Patel KJ. The Fanconi anemia pathway orches - trates incisions at sites of crosslinked DNA. J Pathol. where presentation is atypical) and are allowing improve - 2012;226:326-37. ments to be made in personalized management. This 22. Kottemann MC, Smogorzewska A. Fanconi anaemia and the includes the use of low-intensity fludarabine-based proto - repair of Watson and Crick DNA crosslinks. Nature. 2013;493:356-63. cols that have resulted in major improvements in outcome 23. Garaycoechea JI, Crossan GP, Langevin F, Daly M, Arends following hematopoietic SCT. New targeted therapies for MJ, Patel KJ. Genotoxic consequences of endogenous aldehy - the different syndromes may emerge in the future. des on mouse haematopoietic stem cell function. Nature. 2012;489:571-5. 24. Hira A, Yabe H, Yoshida K, Okuno Y, Shiraishi Y, Chiba K, et al. Variant ALDH2 is associated with accelerated progression References of bone marrow failure in Japanese Fanconi anemia patients. Blood. 2013;122: 3206-9. 1. Alter BP, Young NS. The bone marrow failure syndromes. In: 25. Kumari U, Ya Jun W, Huat Bay B, Lyakhovich A. Evidence of

| 306 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) Milan, Italy, June 12-15, 2014

mitochondrial dysfunction and impaired ROS detoxifying 48. Calado RT, Regal JA, Kleiner DE, Schrump DS, Peterson NR, machinery in Fanconi Anemia cells. Oncogene. Pons V, et al. A spectrum of severe familial liver disorders 2013;33(2):165-72. associate with telomerase mutations. PLoS One. 26. Zinsser F. Atrophia Cutis Reticularis cum Pigmentations, 2009;4:e7926. Dystrophia Unguium et Leukoplakis oris (Poikioodermia 49. Calado RT, Regal JA, Hills M, Yewdell WT, Dalmazzo LF, atrophicans vascularis Jacobi.). Ikonographia Dermatologica. Zago MA, et al. Constitutional hypomorphic telomerase muta - 1910;5:219-23. tions in patients with acute myeloid leukemia. Proc Natl Acad 27. Dokal I. Dyskeratosis congenita in all its forms. Br J Sci USA. 2009;106:1187-92. Haematol. 2000;110:768-79. 50. Kirwan M, Vulliamy T, Marrone A, Walne AJ, Beswick B, 28. Heiss NS, Knight SW, Vulliamy TJ, Klauck SM, Wiemann S, Hillmen P, et al. Defining the pathogenic role of telomerase Mason PJ, et al. X-linked dyskeratosis congenita is caused by mutations in myelodysplastic syndrome and acute myeloid mutations in a highly conserved gene with putative nucleolar leukaemia. Hum Mutat. 2009;30:1567-73. functions. Nature Genet. 1998;19:32-8. 51. Knight SW, Heiss NS, Vulliamy TJ, Aalfs CM, McMahon C, 29. Vulliamy T, Marrone A, Goldman F, Dearlove A, Bessler M, Richmond P, et al. Unexplained aplastic anaemia, immunode - Mason PJ, et al. The RNA component of telomerase is mutated ficiency, and cerebellar hypoplasia (Hoyeraal–Hreidarsson in autosomal dominant dyskeratosis congenita. Nature. syndrome) due to mutations in the dyskeratosis congenita 2001;413:432-5. gene, DKC1. Br J Haematol. 1999;107:335-9. 30. Vulliamy TJ, Walne A, Baskaradas A, Mason PJ, Marrone A, 52. Vulliamy TJ, Knight SW, Mason PJ, Dokal I. Very short Dokal I. Mutations in the reverse transcriptase component of telomeres in the peripheral blood of patients with X-linked and telomerase (TERT) in patients with bone marrow failure. autosomal dyskeratosis congenita. Blood Cells Mol Dis. Blood Cells Mol Dis. 2005;34:257-63. 2001;27:353-7. 31. Armanios M, Chen JL, Chang YP, Brodsky RA, Hawkins A, 53. Shwachman H, Diamond LK, Oski FA, Khaw KT. The syn - Griffin CA, et al. of telomerase reverse drome of pancreatic insufficiency and bone marrow dysfunc - transcriptase leads to in autosomal dominant tion. J Pediatr. 1964;65:645-63. dyskeratosis congenita. Proc Natl Acad Sci USA. 2005;102: 54. Dror Y, Freedman MH. Shwachman-Diamond syndrome. Br J 15960-4. Haematol. 2002;118:701-13. 32. Marrone A, Walne A, Tamary H, Masunari Y, Kirwan M, 55. Boocock GRB, Morrison JA, Popovic M, Richards N, Ellis L, Beswick R, et al. Telomerase reverse transcriptase homozy - Durie PR, et al. Mutations in SBDS are associated with gous mutations in autosomal recessive dyskeratosis congenita Shwachman-Diamond syndrome. Nat Genet. 2003;33:97-101. and Hoyeraal-Hreidarsson syndrome. Blood. 2007;110:4198- 56. Menne TF, Goyenechea B, Sanchez-Puig N, Wong CC, 205. Tonkin LM, Ancliff PJ, et al. The Shwachman-Bodian 33. Walne AJ, Vulliamy T, Marrone A, Beswick R, Kirwan M, Diamond syndrome protein mediates translational activation Masunari Y, et al. Genetic Heterogeneity in autosomal reces - of ribosomes in yeast. Nat Genet. 2007;39:486-96. sive dyskeratosis congenita with one subtype due to mutations 57. Josephs HW. Anemia of infancy and early childhood. in the telomerase-associated protein NOP10. Hum Mol Genet. Medicine. 1936;15:307. 2007;16:1619-29. 58. Lipton JM, Astidaaftos E, Zyskind I, Vlachos A. Improving 34. Vulliamy T, Beswick R, Kirwan M, Marrone A, Digweed M, clinical care and elucidating the pathophysiology of Diamond Walne A, et al. Mutations in the telomerase component NHP2 Blackfan anemia: An update from the Diamond Blackfan cause the premature ageing syndrome dyskeratosis congenita. Anemia Registry. Pediatr Blood Cancer. 2006;46:558-64. Proc Natl Acad Sci USA. 2008;105:8073-8. 59. Draptchinskaia N, Gustavsson P, Andersson B, Pettersson M, 35. Savage SA, Giri N, Baerlocher GM, Orr N, Lansdorp PM, Willig TN, Dianzani I, et al. The gene encoding ribosomal pro - Alter BP. TINF2, a component of the shelterin telomere pro - tein S19 is mutated in Diamond-Blackfan anaemia. Nat Genet. tection complex, is mutated in dyskeratosis congenita. Am J 1999;21:169-75. Hum Genet. 2008;82:501-9. 60. Cmejla R, Cmejlova J, Handrkova H, Petrak J, Pospisilova D. 36. Walne AJ, Vulliamy T, Beswick R, Kirwan M, Dokal I. Ribosomal protein S17 gene (RPS17) is mutated in Diamond- Mutations in C16orf57 and normal length telomeres unify a Blackfan anemia. Hum Mutat. 2007;28:1178-82. subset of patients with dyskeratosis congenita, 61. Gazda HT, Sheen MR, Vlachos A, Choesmel V, O’Donohue with neutropenia and Rothmund-Thomson syndrome. Hum MF, Schneider H, et al. Ribosomal protein L5 and L11 muta - Mol Genet. 2010;19:4453-61. tions are associated with cleft palate and abnormal thumbs in 37. Zhong F, Savage SA, Shkreli M, Giri N, Jessop L, Myers T, et Diamond-Blackfan anemia patients. Am J Hum Genet. al. Disruption of telomerase trafficking by TCAB1 mutation 2008;83:769-80. causes dyskeratosis congenita. Genes Dev. 2011;25:11-6. 62. Farrar JE, Nater M, Caywood E, McDevitt MA, Kowalski J, 38. Keller RB, Gagne KE, Usmani GN, Asdourian GK, Williams Takemoto CM, et al. Abnormalities of the large ribosomal sub - DA, Hofmann I, et al. CTC1 Mutations in a patient with unit protein, Rpl35a, in Diamond-Blackfan anemia. Blood. dyskeratosis congenita. Pediatr Blood Cancer. 2012;59:311-4. 2008;112:1582-92. 39. Walne AJ, Bhagat T, Kirwan M, Gitaux C, Desguerre I, 63. Choesmel V, Fribourg S, Aguissa-Touré AH, Pinaud N, Legrand Leonard N, et al. Mutations in the telomere capping complex P, Gazda HT, et al. Mutation of ribosomal protein RPS24 in in bone marrow failure and related syndromes. Diamond-Blackfan anemia results in a ribosome biogenesis Haematologica. 2013;98:334-8. disorder. Hum Mol Genet. 2008;17:1253-63. 40. Ballew BJ, Yeager M, Jacobs K, Giri N, Boland J, Burdett L, 64. Doherty L, Sheen MR, Vlachos A, Choesmel V, O’Donohue et al. Germline mutations of regulator of telomere elongation MF, Clinton C, et al. Ribosomal protein genes RPS10 and helicase 1, RTEL1, in Dyskeratosis congenita. Hum Genet. RPS26 are commonly mutated in Diamond-Blackfan anemia. 2013;132:473-80. Am J Hum Genet. 2010;86:222-8. 41. Walne AJ, Vulliamy T, Kirwan M, Plagnol V, Dokal I. 65. Gazda HT, Preti M, Sheen MR, O’Donohue MF, Vlachos A, Constitutional mutations in RTEL1cause severe dyskeratosis Davies SM, et al. Frameshift mutation in p53 regulator RPL26 congenita. Am J Hum Genet. 2013;92(3):448-53. is associated with multiple physical abnormalities and a spe - 42. Mitchell JR, Wood E, Collins K. A telomerase component is cific pre-ribosomal RNA processing defect in diamond-black - defective in the human disease dyskeratosis congenita. Nature. fan anemia. Hum Mutat. 2012;33:1037-44. 1999;402:551-5. 66. Landowski M, O’Donohue MF, Buros C, Ghazvinian R, 43. Cohen SB, Graham ME, Lovrecz GO, Bache N, Robinson PJ, Montel-Lehry N, Vlachos A, et al., Novel deletion of RPL15 Reddel RR. Protein composition of catalytically active human identified by array-comparative genomic hybridization in telomerase from immortal cells. Science. 2007;315:1850-3. Diamond-Blackfan anemia. Hum Genet. 2013;132:1265-74. 44. Blasco MA. Telomere length, stem cells and aging. Nat Chem 67. Sankaran VG, Ghazvinian R, Do R, Thiru P, Vergilio JA, Biol. 2007;3:640-9. Beggs AH, et al. Exome sequencing identifies GATA1 muta - 45. Vulliamy T, Marrone A, Dokal I, Mason PJ. Association tions resulting in Diamond-Blackfan anemia. J Clin Invest. between aplastic anaemia and mutations in telomerase RNA. 2012;122:2439-43. Lancet. 2002;359:2168-70. 68. Konno Y, Toki T, Tandai S, Xu G, Wang R, Terui K, et al. 46. Yamaguchi H, Calado RT, Ly H, Kajigaya S, Baerlocher GM, Mutations in the ribosomal protein genes in Japanese patients Chanock SJ, et al. Mutations in TERT, the gene for reverse with Diamond-Blackfan anemia. Haematologica. transcriptase, in aplastic anemia. N Engl J Med. 2010;95:1293-9. 2005;352:1413-24. 69. Wickramasinghe SN, Wood WG. Advances in the understand - 47. Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, ing of congenital Dyserythropoietic anaemia. Br J Haematol. Markin C, et al. Telomerase mutations in families with idio - 2005;131:431-6. pathic pulmonary fibrosis. N Engl J Med. 2007;356:1317-26. 70. Iolascon A, Heimpel H, Wahlin A, Tamary H. Congenital

Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1) | 307 | 19 th Congress of the European Hematology Association

dyserythropoietic anemias: molecular insights and diagnostic thrombopoietin ligand mutation in a Micronesian family with approach. Blood. 2013;122:2162-6. autosomal recessive aplastic anemia. Blood. 2013 Oct 1 [Epub 71. Dgany O, Avidan N, Delaunay J, Krasnov T, Shalmon L, ahead of print] Shalev H, et al. Congenital dyserythropoietic anemia type I is 86. Kirwan M, Walne AJ, Plagnol V, Velangi M, Ho A, Hossain U, caused by mutations in codanin-1. Am J Hum Genet. 2002;71:1467-74. et al. Exome sequencing identifies autosomal-dominant 72. Babbs C, Roberts NA, Sanchez-Pulido L, McGowan SJ, SRP72 mutations associated with familial aplasia and Ahmed MR, Brown JM, et al. Homozygous mutations in a myelodysplasia. Am J Hum Genet. 2012;90:888-92. predicted endonuclease are a novel cause of congenital 87. Tamary H, Nishri D, Yacobovich J, Zilber R, Dgany O, dyserythropoietic anemia type I. Haematologica. Krasnov T, et al. Frequency and natural history of inherited 2013;98:1383-7. bone marrow failure syndromes: the Israeli Inherited Bone 73. Schwarz K, Iolascon A, Verissimo F, Trede NS, Horsley W, Chen W, et al. Mutations affecting the secretory COPII coat Marrow Failure Registry. Haematologica. 2010;95:1300-7. component SEC23B cause congenital dyserythropoietic ane - 88. Tsangaris E, Klaassen R, Fernandez CV, Yanofsky R, Shereck mia type II. Nat Genet. 2009;41:936-40. E, Champagne J, et al. Genetic analysis of inherited bone mar - 74. Liljeholm M, Irvine AF, Vikberg AL, Norberg A, Month S, row failure syndromes from one prospective, comprehensive Sandström H, et al. Congenital dyserythropoietic anemia type and population-based cohort and identification of novel muta - III (CDA III) is caused by a mutation in kinesin family mem - tions. J Med Genet. 2011;48:618-28. ber, KIF23. Blood. 2013;121:4791-9. 89. Alter BP, Baerlocher GM, Savage SA, Chanock SJ, Weksler 75. Arnaud L, Saison C, Helias V, Lucien N, Steschenko D, Giarratana MC, et al. A dominant mutation in the gene encod - BB, Willner JP, et al. Very short telomere length by flow fluo - ing the erythroid transcription factor KLF1 causes a congenital rescence in situ hybridization identifies patients with dysker - dyserythropoietic anemia. Am J Hum Genet. 2010;87:721-7. atosis congenita. Blood. 2007;110:1439-47. 76. Welte K, Zeidler C. Severe Congenital Neutropenia. Hematol 90. Ahmed M, Dokal I. Understanding aplastic anaemia/bone Oncol Clin North Am. 2009;23:307-20. marrow failure syndromes. Paediatrics and Child Health. 77. Hauck F, Klein C. Pathogenic mechanisms and clinical impli - 2009;19:351-7. cations of congenital neutropenia syndromes. Curr Opin 91. Scheckenbach K, Morgan M, Filger-Brillinger J, Sandmann Allergy Clin Immunol. 2013;13:596-606. 78. Dale DC, Person RE, Bolyard AA, Aprikyan AG, Bos C, M, Strimling B, Scheurlen W, et al. Treatment of the bone Bonilla MA, et al. Mutations in the gene encoding neutrophil marrow failure in Fanconi anemia patients with danazol. elastase in congenital and cyclic neutropenia. Blood. Blood Cells Mol Dis. 2012;48:128-31. 2000;96:2317-22. 92. Islam A, Rafiq S, Kirwan M, Walne A, Cavenagh J, Vulliamy 79. Klein C, Grudzien M, Appaswamy G, Germeshausen M, T, et al. Haematological recovery in dyskeratosis congenita Sandrock I, Schäffer AA, et al. HAX1 deficiency causes auto - patients treated with danazol. Br J Haematol. 2013;162:854-6. somal recessive severe congenital neutropenia (Kostmann dis - 93. de la Fuente J, Reiss S, McCloy M, Vulliamy T, Roberts IA, ease). Nat Genet. 2007;39:86-92. 80. Person RE, Li FQ, Duan Z, Benson KF, Wechsler J, Papadaki Rahemtulla A, et al. Non-TBI stem cell transplantation proto - HA, et al Mutations in proto-oncogene GFI1 cause human col for Fanconi anaemia using HLA-compatible sibling and neutropenia and target ELA2. Nat Genet. 2003;34:308-12. unrelated donors. Bone Marrow Transplant. 2003;32:653-6. 81. Boztug K, Appaswamy G, Ashikov A, Schäffer AA, Salzer U, 94. Gluckman E, Wagner JE. Hematopoietic stem cell transplan - Diestelhorst J, et al. A syndrome with congenital neutropenia tation in childhood inherited bone marrow failure syndrome. and mutations in G6PC3. N Engl J Med. 2009;360:32-43. Bone Marrow Transplant. 2008;41:127-32. 82. Vilboux T, Lev A, Malicdan MC, Simon AJ, Järvinen P, Racek T, et al. A congenital neutrophil defect syndrome associated 95. Peffault de Latour R, Porcher R, Dalle JH, Aljurf M, Korthof with mutations in VPS45. N Engl J Med. 2013;369:54-65. ET, et al. Allogeneic hematopoietic stem cell transplantation in 83. Ihara K, Ishii E, Eguchi M, Takada H, Suminoe A, Good RA, Fanconi anemia: the EBMT experience. Blood. et al. Identification of mutations in the c-mpl gene in congen - 2013;122:4279-86. ital amegakaryocytic thrombocytopenia. Proc Natl Acad Sci 96. de la Fuente J, Dokal I. Dyskeratosis congenita: advances in USA. 1999;96:3132-6. the understanding of the telomerase defect and the role of stem 84. Walne AJ, Dokal A, Plagnol V, Beswick R, Kirwan M, de la cell transplantation. Pediatr Transplant. 2007;11:584-94. Fuente J, et al. Exome sequencing identifies MPL as a causative gene in familial aplastic anemia. Haematologica. 97. Dietz AC, Orchard PJ, Baker KS, Giller RH, Savage SA, Alter 2012;97:524-8. BP, et al. Disease-specific hematopoietic cell transplantation: 85. Dasouki MJ, Rafi SK, Olm-Shipman AJ, Wilson NR, non myeloablative conditioning regimen for dyskeratosis con - Abhyankar S, Ganter B, et al. Exome sequencing reveals a genita. Bone Marrow Transplant. 2011;46:98-104.

| 308 | Hematology Education: the education program for the annual congress of the European Hematology Association | 2014; 8(1)