Myelodysplastic syndrome (MDS) typically presents in older adults with the acquisition of age-related somatic mutations, whereas MDS presenting in children and younger adults is more frequently associated with germline genetic predisposition. Germline predisposition is increasingly recognized in MDS presenting at older ages as well. Although each individual genetic disorder is rare, as a group, the genetic MDS disorders account for a significant subset of MDS in children and young adults. Because many patients lack overt syndromic features, genetic testing plays an important role in the diagnostic evaluation. This review provides an overview of syndromes associated with genetic predisposition to MDS, discusses implications for clinical evaluation and management, and explores scientific insights gleaned from the study of MDS predisposition syndromes. The effects of germline genetic context on the selective pressures driving somatic clonal evolution are explored. Elucidation of the molecular and genetic pathways driving clonal evolution may inform surveillance and risk stratification, and may lead to the development of novel therapeutic strategies.

Myelodysplastic syndrome (MDS) is defined by the World Health Organization as a clonal hematopoietic disorder characterized by ineffective hematopoiesis, cytopenias, single- or multilineage dysplasia, and an increased propensity to evolve to acute myeloid leukemia (AML).1  Primary MDS typically presents in older adults, with median age of disease onset of 72 to 75 years in the context of somatic mutations acquired with age.2,3  MDS presenting in children and younger adults is more frequently associated with germline genetic predisposition.4  In recognition of the growing subgroup of MDS or acute leukemia associated with germline mutations or familial cases, the revised World Health Organization classification of myeloid neoplasms now includes a new category: myeloid neoplasms with germline predisposition.1 

A high risk for MDS in childhood and early adulthood is characteristic of certain genetic bone marrow failure syndromes.5  Although physical stigmata and family history provide important clues to an underlying genetic MDS predisposition syndrome, it is now clear that clinically silent phenotypes with cryptic presentations are common. Several genomic studies demonstrated that a subset of pediatric and young adult patients younger than 40 years presenting with MDS have unrecognized bone marrow failure and genetic MDS syndromes.4,6,7  Although many published studies on germline genetic predisposition thus far have typically used the adolescent and young adult upper age range of 40 years, it is critical to note that genetic MDS predisposition syndromes also present in older adulthood. In particular, germline mutations in DDX41 present with myeloid malignancies in older adults. Germline telomere biology disorders, GATA2, thrombocytopenia-associated disorders, and others may present across all ages. Older age at presentation is insufficient to rule out a germline predisposition disorder. Indeed, before the availability of genetic testing, diagnosis of MDS predisposition was skewed toward patients presenting with overt syndromic features in childhood, whereas nonsyndromic patients presenting with MDS in adulthood were previously missed. Additional data are needed to inform evidence-based determination of age of presentation for each MDS predisposition disorder in the era of genetic testing. Recent studies have shown that although each individual genetic disorder is rare, as a group they account for approximately 4% to 15% of patients with MDS.4,6,7  These findings elucidate the broad phenotypic spectrum of these genetic syndromes and highlight the importance of screening for these disorders.

The diagnosis of a germline MDS predisposition disorder carries profound implications for medical management and treatment. Baseline dysplasia associated with some MDS predisposition disorders must not be confused with frank MDS. Prompt diagnosis of these disorders allows surveillance to detect early signs of disease progression and timely hematopoietic stem cell transplant (HSCT) before progression to leukemia, which often carries a poor prognosis. Many of these genes are associated with additional medical complications or comorbidities that may require modified conditioning regimens for HSCT. Genetic testing of potential family member donors is essential before HSCT to avoid choosing a donor carrying the same germline mutations as the proband. Diagnosis also allows family counseling.

This review provides an overview of genetic predisposition to MDS, discusses clinical evaluation and management, and explores scientific insights garnered from the study of MDS germline genetics. The germline genetic MDS disorders advance our understanding of the genes that govern hematopoiesis, as well as the cellular and molecular perturbations that lead to malignant transformation. Last, we explore the emerging hypothesis that somatic mutations may result in hematopoietic stem cell adaptation or oncogenic transformation driven by specific selective pressures exerted by the germline genetic context of the hematopoietic stem cell. Because of space limitations, only a subset of disorders in which MDS predisposition is a predominant feature are included here. This focused review of MDS predisposition does not review the broader classification of leukemia predisposition disorders such as lymphoid malignancies, myeloproliferative syndromes, chronic myelomonocytic leukemia, or RASopathies, so the reader is referred to the other articles within this issue, as well as excellent recent reviews of these topics and the references therein.8-11 

GATA2

GATA2 encodes a zinc finger transcription factor critical for hematopoiesis, hematopoietic stem cell homeostasis, and lymphatic development (see Spinner et al12  and McReynolds et al13  and references therein). The phenotypic spectrum of germline GATA2 mutations is broad and may present with MonoMac syndrome14  or Emberger syndrome15  (Table 1). However, germline GATA2 mutations may also present with isolated neutropenia or bone marrow failure without syndromic features or family history.16-18  The bone marrow histology in GATA2 deficiency is typically hypocellular and may manifest characteristic megakaryocyte dysmorphologies with micronuclei or splayed nuclei19  (Figure 1). Additional findings include monocytopenia and immunologic abnormalities.

Table 1.

Genes involved in predisposition to myelodysplastic syndrome

Cellular function, syndrome, geneInheritanceHematologic malignancyOther malignancyVariable associated phenotypes
DNA repair     
Fanconi anemia     
  FANCA AR MDS, AML Squamous cell carcinoma: oral, gastrointestinal, genitourinary; FANCD1/BRCA2 subtype also presents with other solid tumors and ALL. Bone marrow failure, short stature, hyper/hypopigmentation, facial dysmorphologies, skeletal, digits, hypogonadism, cardiopulmonary, gastrointestinal, endocrinopathies, CNS 
  FANCB XLR 
  FANCC AR 
  FANCD1/BRCA2 AR 
  FANCD2 AR 
  FANCE AR 
  FANCF AR 
  FANCG AR 
  FANCI AR 
  FANCJ/BRIP1/BACH1 AR 
  FANCL AR 
  FANCM AR 
  FANCN/PALB2 AR 
  FANCO/RAD51C AR 
  FANCP/SLX4 AR 
  FANCQ/ERCC4 AR 
  FANCR/RAD51 AD 
  FANCS/BRCA1 AR 
  FANCT/UBE2T AR 
  FANCU/XRCC2 AR 
  FANCV/REV7 AR 
  FANCW/RFWD3 AR    
Transcription factor     
 Familial platelet disorder with propensity to myeloid malignancy     
  RUNX1 AD MDS, AML, T-ALL, hairy cell leukemia  Thrombocytopenia, platelet dysfunction 
 GATA2-spectrum disorders     
  GATA2 AD MDS, AML, CMML, JMML  Emberger syndrome, MonoMac syndrome, immunodeficiencies/immune dysregulation (monocytopenia, B-cell deficiencies, NK cell deficiencies), hearing loss, warts, atypical mycobacterial infections, lymphedema, monocytopenia, marrow failure 
 Thrombocytopenia 5     
  ETV6 AD ALL, MDS, AML, CMML, MM  Thrombocytopenia, platelet dysfunction 
Ribosomal abnormalities     
 Diamond Blackfan anemia     
  GATA1 AD MDS, AML Osteosarcoma, soft tissue sarcomas Bone marrow failure, short stature, radial ray abnormalities, cardiac, renal anomalies, facial dysmorphisms 
  RPL5 AD 
  RPL11 AD 
  RPL15 AD 
  RPL23 AD 
  RPL26 AD 
  RPL27 AD 
  RPL31 AD 
  RPL35a AD 
  RPL36 AD 
  RPS7 AD 
  RPS10 AD 
  RPS15 AD 
  RPS17 AD 
  RPS19 AD 
  RPS24 AD 
  RPS26 AD 
  RPS27 AD 
  RPS27A AD 
  RPS28 AD 
  RPS29 AD 
  TSR2 X-linked 
 Shwachman Diamond syndrome     
  SBDS AR MDS, AML  Bone marrow failure, short stature, exocrine pancreatic dysfunction, skeletal dysplasias, immunodeficiencies, neurocognitive, other congenital anomalies 
  DNAJC21 AR AML  
  EFL1 AR  
Telomere maintenance     
 DC/TBD     
  DKC1 XLR MDS, AML Oral and gastrointestinal squamous cell carcinoma Bone marrow failure, nail dystrophy, skin pigmentation abnormalities, oral leukoplakia, idiopathic pulmonary fibrosis, liver disease, short telomeres, immunodeficiencies, vascular anomalies, other congenital anomalies 
  TERC AD 
  TERT AD, AR 
  NOLA3/NOP10 AR 
  NOLA2/NHP2 AR 
  TINF2 AD 
  WRAP53/TCAB1 AR 
  CTC1 AR 
  RTEL1 AD, AR 
  ACD/ TPP1 AD, AR 
  PARN AD, AR 
  NAF1 AD 
  STN1 AD 
RAS pathway     
 Noonan syndrome     
  PTPN11, SOS1, RAF1, KRAS, NRAS, BRAF, MAP2K1 AD JMML, MDS, AML, MPN Solid tumors Facial dysmorphism, short stature, cardiac, broad neck, thoracic, cryptorchidism, coagulopathy (Noonan) 
 Noonan-like    
  CBL AD JMML, MDS  
Other     
 Severe congenital neutropenia     
  ELANE AD MDS, AML (ALL?)  Osteopenia 
  HAX1 AR MDS, AML (ALL?)  Seizures, neurologic abnormalities 
  GFI1 AD MDS, AML  Lymphopenia 
  G6PC3 AR MDS, AML  Neutropenia, congenital heart malformations, prominent superficial veins, urogenital abnormalities, Dursun syndrome 
  JAGN1 AR AML  Skeletal and dental abnormalities 
 Thrombocytopenia 2     
  ANKRD26 AD MDS, AML, CLL, CMML  Thrombocytopenia, platelet dysfunction 
 X-linked neutropenia     
  WAS X-linked MDS, AML  Immunologic abnormalities, monocytopenia 
 MIRAGE syndrome, (myelodysplasia, infection, restriction of growth, adrenal insufficiency, genital phenotypes, and enteropathy)     
  SAMD9 AD MDS, AML  Cytopenias, immunologic abnormalities, short stature, adrenal hypoplasia, infections, gastrointestinal, genitourinary skeletal, CNS, monosomy 7 
 Ataxia-pancytopenia syndrome     
  SAMD9L AD MDS, AML  Bone marrow failure, ataxia, monosomy 7 
 Neurofibromatosis 1     
  NF1 AD JMML, MDS Optic gliomas, CNS tumors, malignant peripheral nerve sheath tumors, other solid tumors Café au lait spots, axillary freckling, Lisch nodules, neurofibromas 
 Bloom syndrome     
  BLM AR Lymphoma, ALL, MDS, AML  Short stature, sun-sensitive rash, pulmonary disease, immunodeficiency 
 Constitutional mismatch repair deficiency     
  MLH1 AR ALL, Lymphoma, MDS, AML Gastrointestinal (colon), ovarian, uterine, CNS, other  
  MSH2 AR 
  MSH6 AR 
  EPCAM AR 
  PMS2 AR 
 Li-Fraumeni     
  TP53 AD ALL, MDS, AML, lymphoma Breast, osteosarcoma, soft tissue sarcoma, CNS, adrenocortical carcinoma, other  
 Susceptibility to acute myeloid leukemia     
  DDX41 AD MDS, AML, CML, lymphoma  Malignancies present in older age (>50 y) 
 Familial myeloproliferative neoplasms     
  JAK2 haplotype 46/1 [GGCC], TERT rs2736100_c AD PV, ET, PMF   
  ATG2B, GSKIP AD PV, ET, PMF, CML, CMML, AML 
  RBBP6 AD PV, ET, PMF 
 Myeloid neoplasms associated with trisomy 8 mosaicism     
  Trisomy 8 Variable MDS, AML, CML   
 Congenital neutropenia or SDS-Like disorder     
  SRP54 AR  Bone marrow failure, short stature, exocrine pancreatic dysfunction, skeletal dysplasias, immunodeficiencies 
 Bone Marrow Failure Syndrome 1     
  SRP72 AD MDS  Bone marrow failure, sensorineural hearing loss 
Cellular function, syndrome, geneInheritanceHematologic malignancyOther malignancyVariable associated phenotypes
DNA repair     
Fanconi anemia     
  FANCA AR MDS, AML Squamous cell carcinoma: oral, gastrointestinal, genitourinary; FANCD1/BRCA2 subtype also presents with other solid tumors and ALL. Bone marrow failure, short stature, hyper/hypopigmentation, facial dysmorphologies, skeletal, digits, hypogonadism, cardiopulmonary, gastrointestinal, endocrinopathies, CNS 
  FANCB XLR 
  FANCC AR 
  FANCD1/BRCA2 AR 
  FANCD2 AR 
  FANCE AR 
  FANCF AR 
  FANCG AR 
  FANCI AR 
  FANCJ/BRIP1/BACH1 AR 
  FANCL AR 
  FANCM AR 
  FANCN/PALB2 AR 
  FANCO/RAD51C AR 
  FANCP/SLX4 AR 
  FANCQ/ERCC4 AR 
  FANCR/RAD51 AD 
  FANCS/BRCA1 AR 
  FANCT/UBE2T AR 
  FANCU/XRCC2 AR 
  FANCV/REV7 AR 
  FANCW/RFWD3 AR    
Transcription factor     
 Familial platelet disorder with propensity to myeloid malignancy     
  RUNX1 AD MDS, AML, T-ALL, hairy cell leukemia  Thrombocytopenia, platelet dysfunction 
 GATA2-spectrum disorders     
  GATA2 AD MDS, AML, CMML, JMML  Emberger syndrome, MonoMac syndrome, immunodeficiencies/immune dysregulation (monocytopenia, B-cell deficiencies, NK cell deficiencies), hearing loss, warts, atypical mycobacterial infections, lymphedema, monocytopenia, marrow failure 
 Thrombocytopenia 5     
  ETV6 AD ALL, MDS, AML, CMML, MM  Thrombocytopenia, platelet dysfunction 
Ribosomal abnormalities     
 Diamond Blackfan anemia     
  GATA1 AD MDS, AML Osteosarcoma, soft tissue sarcomas Bone marrow failure, short stature, radial ray abnormalities, cardiac, renal anomalies, facial dysmorphisms 
  RPL5 AD 
  RPL11 AD 
  RPL15 AD 
  RPL23 AD 
  RPL26 AD 
  RPL27 AD 
  RPL31 AD 
  RPL35a AD 
  RPL36 AD 
  RPS7 AD 
  RPS10 AD 
  RPS15 AD 
  RPS17 AD 
  RPS19 AD 
  RPS24 AD 
  RPS26 AD 
  RPS27 AD 
  RPS27A AD 
  RPS28 AD 
  RPS29 AD 
  TSR2 X-linked 
 Shwachman Diamond syndrome     
  SBDS AR MDS, AML  Bone marrow failure, short stature, exocrine pancreatic dysfunction, skeletal dysplasias, immunodeficiencies, neurocognitive, other congenital anomalies 
  DNAJC21 AR AML  
  EFL1 AR  
Telomere maintenance     
 DC/TBD     
  DKC1 XLR MDS, AML Oral and gastrointestinal squamous cell carcinoma Bone marrow failure, nail dystrophy, skin pigmentation abnormalities, oral leukoplakia, idiopathic pulmonary fibrosis, liver disease, short telomeres, immunodeficiencies, vascular anomalies, other congenital anomalies 
  TERC AD 
  TERT AD, AR 
  NOLA3/NOP10 AR 
  NOLA2/NHP2 AR 
  TINF2 AD 
  WRAP53/TCAB1 AR 
  CTC1 AR 
  RTEL1 AD, AR 
  ACD/ TPP1 AD, AR 
  PARN AD, AR 
  NAF1 AD 
  STN1 AD 
RAS pathway     
 Noonan syndrome     
  PTPN11, SOS1, RAF1, KRAS, NRAS, BRAF, MAP2K1 AD JMML, MDS, AML, MPN Solid tumors Facial dysmorphism, short stature, cardiac, broad neck, thoracic, cryptorchidism, coagulopathy (Noonan) 
 Noonan-like    
  CBL AD JMML, MDS  
Other     
 Severe congenital neutropenia     
  ELANE AD MDS, AML (ALL?)  Osteopenia 
  HAX1 AR MDS, AML (ALL?)  Seizures, neurologic abnormalities 
  GFI1 AD MDS, AML  Lymphopenia 
  G6PC3 AR MDS, AML  Neutropenia, congenital heart malformations, prominent superficial veins, urogenital abnormalities, Dursun syndrome 
  JAGN1 AR AML  Skeletal and dental abnormalities 
 Thrombocytopenia 2     
  ANKRD26 AD MDS, AML, CLL, CMML  Thrombocytopenia, platelet dysfunction 
 X-linked neutropenia     
  WAS X-linked MDS, AML  Immunologic abnormalities, monocytopenia 
 MIRAGE syndrome, (myelodysplasia, infection, restriction of growth, adrenal insufficiency, genital phenotypes, and enteropathy)     
  SAMD9 AD MDS, AML  Cytopenias, immunologic abnormalities, short stature, adrenal hypoplasia, infections, gastrointestinal, genitourinary skeletal, CNS, monosomy 7 
 Ataxia-pancytopenia syndrome     
  SAMD9L AD MDS, AML  Bone marrow failure, ataxia, monosomy 7 
 Neurofibromatosis 1     
  NF1 AD JMML, MDS Optic gliomas, CNS tumors, malignant peripheral nerve sheath tumors, other solid tumors Café au lait spots, axillary freckling, Lisch nodules, neurofibromas 
 Bloom syndrome     
  BLM AR Lymphoma, ALL, MDS, AML  Short stature, sun-sensitive rash, pulmonary disease, immunodeficiency 
 Constitutional mismatch repair deficiency     
  MLH1 AR ALL, Lymphoma, MDS, AML Gastrointestinal (colon), ovarian, uterine, CNS, other  
  MSH2 AR 
  MSH6 AR 
  EPCAM AR 
  PMS2 AR 
 Li-Fraumeni     
  TP53 AD ALL, MDS, AML, lymphoma Breast, osteosarcoma, soft tissue sarcoma, CNS, adrenocortical carcinoma, other  
 Susceptibility to acute myeloid leukemia     
  DDX41 AD MDS, AML, CML, lymphoma  Malignancies present in older age (>50 y) 
 Familial myeloproliferative neoplasms     
  JAK2 haplotype 46/1 [GGCC], TERT rs2736100_c AD PV, ET, PMF   
  ATG2B, GSKIP AD PV, ET, PMF, CML, CMML, AML 
  RBBP6 AD PV, ET, PMF 
 Myeloid neoplasms associated with trisomy 8 mosaicism     
  Trisomy 8 Variable MDS, AML, CML   
 Congenital neutropenia or SDS-Like disorder     
  SRP54 AR  Bone marrow failure, short stature, exocrine pancreatic dysfunction, skeletal dysplasias, immunodeficiencies 
 Bone Marrow Failure Syndrome 1     
  SRP72 AD MDS  Bone marrow failure, sensorineural hearing loss 

AD, autosomal dominant; AR, autosomal recessive; CMML, chronic myelomonocytic leukemia; ET, essential thrombocythemia; JMML, juvenile myelomonocytic leukemia; PMF, primary myelofibrosis; PV, polycythemia vera.

Because of space limitations, this table is not exhaustive and readers are referred to additional references cited in the text.

Figure 1.

Selected bone marrow histology and physical findings associated with genetic MDS predisposition syndromes. (A) Hematoxylin and eosin (left panel) or May-Grunwald Giemsa staining of abnormal bone marrow megakaryocytes present in GATA2 predisposition disorders. (B) May-Grunwald Giemsa staining of bone marrow aspirate from a patient with SCN demonstrating myeloid maturation arrest (marrow images courtesy of M. Fleming). (C) Patient features characteristic of diagnostic triad of TBDs, including lacy/reticular pigmentation on neck and trunk, oral leukoplakia, and dystrophic fingernails (images courtesy of S. Agarwal).

Figure 1.

Selected bone marrow histology and physical findings associated with genetic MDS predisposition syndromes. (A) Hematoxylin and eosin (left panel) or May-Grunwald Giemsa staining of abnormal bone marrow megakaryocytes present in GATA2 predisposition disorders. (B) May-Grunwald Giemsa staining of bone marrow aspirate from a patient with SCN demonstrating myeloid maturation arrest (marrow images courtesy of M. Fleming). (C) Patient features characteristic of diagnostic triad of TBDs, including lacy/reticular pigmentation on neck and trunk, oral leukoplakia, and dystrophic fingernails (images courtesy of S. Agarwal).

Close modal

MDS with germline GATA2 mutations is frequently associated with monosomy 7/del7q (-7) or trisomy 8, particularly in children and younger adults.20,21  A study of 426 cases of pediatric MDS identified germline GATA2 mutations in 37% of patients with primary MDS with -7 and 16% of MDS cases with trisomy 8.20  Germline GATA2 mutations were identified in 72% of adolescents with -7 MDS. In contrast, germline GATA2 mutations were absent in therapy-related MDS.

The spectrum of pathogenic germline GATA2 mutations includes missense, nonsense, frameshift, duplication, regulatory region alterations, and partial or whole gene deletions.20,22  Mutations result in haploinsufficiency or impairment of DNA binding or transactivation.

SAMD9/SAMD9L

Initial reports associating refractory anemia, monosomy 7, and AML were described by E.J. Freireich in the 1960s.23  Subsequent studies of the recurrent common microdeletion region at 7q22 led to discovery of a region containing 2 genes harboring the sterile α motif (SAM) domain-9 (SAMD9) and SAMD9L (SAMD9-like; see Wlodarski et al21  and Davidsson et al24  and references therein).

Homozygous germline loss-of-function mutations in SAMD9L are associated with normophosphatemic familial tumoral calcinosis,25  whereas heterozygous gain-of-function missense mutations in SAMD9 or SAMD9L are associated with cytopenias and a high risk of developing MDS with monosomy 7/del7q, with variable additional clinical phenotypes (Table 1). Germline mutations in SAMD9 and SAMD9L were initially identified in MIRAGE syndrome26  and ataxia-pancytopenia syndrome,27,28  respectively (Table 1). Patients frequently present with bone marrow failure and a high risk for monosomy 7 MDS at a young age.29-31  Patients with germline SAMD9 and SAMD9L may present with MDS or AML without syndromic features.

The molecular functions of these genes remain poorly understood; both genes exert negative effects on cell proliferation consistent with tumor suppressors.28,31 SAMD9/SAMD9L share a common evolutionary history and encode proteins with 60% homology. Murine studies indicate that SAMD9L functions as an endosomal fusion facilitator and, in the haploinsufficient state, leads to MDS.32  Studies of clonal evolution in these disorders are discussed here.

Fanconi anemia

A century ago, Swiss pediatrician Guido Fanconi described a family with 3 sons with various physical abnormalities and pernicious anemia.33  Now known as Fanconi anemia (FA), this disorder is characterized by genomic instability, hypersensitivity to DNA cross-linking agents, and predisposition to marrow failure, as well as hematologic malignancies and solid tumors.34-36  The congenital anomalies variably associated with FA are numerous (Table 1). A significant subset (25%-30%) of patients lack these physical findings.37  Hematologic abnormalities are variable and include cytopenias, red cell macrocytosis, hypocellular marrow with mild dysplasia, and bone marrow failure with increased risk for MDS or AML. The cumulative incidence for MDS was 40% by age 50 years and 20% by age 40 years for AML.34  The incidence of leukemia is even higher in the FANCD1/BRCA2 subtype of FA, with many cases presenting at younger than 5 years.38 

This clinically and genetically diverse syndrome is caused by germline mutations in any of at least 23 FA genes (FANCA-FANCW) that function coordinately in DNA repair.39  Cells from patients with FA manifest increased chromosomal breakage and radial formation after exposure to mitomycin C or diepoxybutane.40,41 

Of particular note in FA is the importance of reduced-intensity conditioning for HSCT because of the heightened toxicity of radiation and alkylator-based conditioning regimens in these patients.42,43  Unlike other MDS syndromes that can be cured by HSCT, these patients have increased morbidity posttransplantation and have a higher risk for solid tumors compared with nontransplanted patients.44-46 

Diamond-Blackfan anemia

Diamond-Blackfan anemia (DBA) was first described by Diamond and Blackfan in the 1930s as a congenital hypoplastic anemia.47  DBA registries across the world are elucidating the clinical and molecular features of DBA (see Da Costa et al,48  Vlachos et al,49  and Vlachos et al50  and references therein) Approximately half of patients with DBA exhibit physical abnormalities (Table 1). The majority of DBA cases are associated with increased levels of erythrocyte adenosine deaminase activity.51  Patients typically present in infancy with macrocytic anemia and reticulocytopenia. The bone marrow histopathology typically shows red cell aplasia in an otherwise normocellular marrow. Major causes of morbidity and mortality are related to treatment adverse effects and long-term risks for malignancy, including MDS and solid tumors.52,53  In a study from the DBA Registry of North America, 2% of patients with DBA had developed AML by age 45 years.52 

DBA is most frequently caused by autosomal dominant mutations, resulting in haploinsufficiency of genes encoding ribosomal proteins, most commonly RPS19.54  Around 50% of ribosomal gene mutations arise de novo.48  X-linked mutations in GATA1, which encodes a transcription factor critical for erythropoiesis, also cause DBA.55  X-linked mutations in TSR2 that binds RPS26 have also been reported. How mutations in ubiquitously expressed genes encoding an essential organelle result in tissue-specific phenotypes has been a subject of intense interest. Disease mechanisms include p53-mediated apoptosis induced by ribosomal stress,56  increased cell death from free excess heme with delayed globin production,57  increased autophagy,58  and altered translation of select erythroid-specific transcripts such as GATA1.59,60  In adult 5q- syndrome MDS, an acquired deletion resulting in haploinsufficiency of the RPS14 gene is associated with MDS61  and sensitivity to lenalidomide.62 

Dyskeratosis congenita/telomere biology disorders

Dyskeratosis congenita (DC) or telomere biology disorders (TBDs) encompass genetically heterogeneous disorders related to impaired telomere maintenance (see Townsley et al,63  Bertuch et al,64  and Agarwal et al65  and references therein). These deficiencies lead to a broad and variable clinical phenotype (Table 1; Figure 1), but patients often present without overt syndromic features.66  DC/TBDs are frequently associated with hematologic complications including bone marrow failure, MDS, and AML.65,67  The actuarial risk (absent competing risks) of MDS in the patients with DC reached a plateau of 3% at 29 years of age.68  The cumulative incidence of MDS in DC has been estimated to be 2% by age 50 years.69  MDS may be the sole presenting clinical feature. Screening for TBDs involves evaluation of lymphocyte telomere lengths, which are typically below the first percentile for age across multiple lymphocyte subsets.70,71  The sensitivity of this test may be lower in adults as a result of decreased telomere lengths with increasing age. DC/TBD is associated with many extrahematologic complications, particularly pulmonary fibrosis, hepatic abnormalities, and vascular anomalies.72,73  Patients are sensitive to toxicities from chemotherapy and radiation and warrant specially tailored transplant regimens.65,74,75 

DC/TBDs are caused by mutations in at least 13 genes involved in telomere maintenance with variable inheritance patterns (Table1).64  Genetic testing is helpful diagnostically, as shortened telomeres are sometimes noted in other disorders.71  Telomeres are specialized protein:DNA complexes protecting chromosome ends that prevent genomic instability resulting from free DNA ends (see Townsley et al63  and references therein). Telomeres shorten over progressive cycles of DNA replication. Critically short telomere lengths trigger senescence and cell death.

Shwachman Diamond syndrome

In 1964, Shwachman and Diamond observed children in a cystic fibrosis clinic who had pancreatic insufficiency and neutropenia and normal sweat tests, without pulmonary manifestations.76  Separately, Bodian described cases of familial exocrine pancreatic insufficiency.77  Shwachman Diamond syndrome (SDS) is characterized by exocrine pancreatic dysfunction and bone marrow failure, as well as additional physical findings (Table 1). However, these features are not present in all patients,78  and patients may present with MDS as the first observed feature of disease.4  The most common hematologic abnormality is neutropenia, which may be mild or severe and may be transient or persistent. Additional hematologic complications include bone marrow failure, MDS, and AML. In a French cohort of 102 patients with SDS, the cumulative incidence of MDS/AML was 18.8% at 20 and 36.1% at 30 years of age.79  The bone marrow is typically hypocellular for age, with mild dysgranulopoietic features. The most sensitive test of exocrine pancreatic dysfunction in SDS is serum trypsinogen below age 3 years or pancreatic isoamylase at or above age 3 years.80 

SDS is most commonly caused by autosomal recessive mutations in the eponymous SBDS gene, leading to low levels of SDS protein.81  SDS is involved in the joining of the large (60S) and small (40S) ribosomal subunits and functions as a cofactor for elongation factor-like (EFL1) to remove the antiassociation factor eIF6 from the large (60S) subunit.82-85  SDS also plays a role in mitotic spindle stabilization.86  Data from the North American SDS Registry reveal a spectrum of SBDS mutations including missense, splice site, nonsense, frameshift, and partial or whole gene deletions. Recently, autosomal recessive mutations in DNAJC2187,88  and EFL1,89  which also function in ribosome maturation, have been found in patients with variable clinical features of SDS. Autosomal dominant mutations in SRP54 have also been found in patients with SDS-like features.90  SRP54 protein is a component of the ribonucleoprotein complex that mediates cotranslational targeting of secretory and membrane proteins to the endoplasmic reticulum. Patients with SRP54 mutations may have exocrine pancreatic insufficiency or severe neutropenia as a presenting symptom.91  Thus far, AML has been described in a patient with DNAJC21 mutations,90  and no cases of MDS or AML have been described in patients with EFL1 or SRP54 mutations.

Familial MDS associated with thrombocytopenia

RUNX1

RUNX1 deficiency is the cause of an autosomal dominant familial platelet disorder with predisposition to myeloid leukemia (see Bellissimo et al92  and Schlegelberger et al93  and references therein). Familial platelet disorder with predisposition to myeloid leukemia typically presents with mild to moderate thrombocytopenia with normal-sized platelets, functional platelet defects leading to excessive bleeding, and an increased risk of developing MDS, AML, or lymphoid malignancies such as T-cell acute lymphoblastic leukemia (T-ALL) or B-cell malignancies. The thrombocytopenia may be mild and asymptomatic. Qualitative platelet abnormalities may cause bleeding in excess of that predicted by the platelet count. Although longitudinal data are limited, lifetime risk for myeloid malignancy in patients with germline RUNX1 mutations appears to be highly variable and has been estimated to be around 44%, but ranges from 11% to 100% (reviewed in Godley et al22 ). Disease anticipation whereby the clinical manifestations arise at younger ages in subsequent generations has been described in families with germline RUNX1 mutations.

RUNX1 encodes a heterodimeric transcription factor essential for hematopoiesis, megakaryopoiesis, and platelet function. RUNX1 functions as a transcriptional activator for some genes and a transcriptional repressor for others. Germline mutations in RUNX1 include missense, nonsense, frameshift, duplication, partial or full gene deletion mutations, or gene rearrangements.22  Missense point mutations often occur in the highly conserved RUNT homology domain at sites affecting DNA binding or heterodimerization. Truncations within the RUNT domain may result in hypomorphic mutations.

ETV6

Autosomal dominant germline mutations in ETV6 (also known as TEL) are associated with inherited thrombocytopenia and increase risk for both myeloid and lymphoid hematologic malignancies (see Hock et al94  and references therein). Clinical features of germline ETV6 mutations include thrombocytopenia, platelet function abnormalities, macrocytosis, and increased bleeding tendency. The marrow may be hypocellular with dysmegakaryopoiesis, variable megakaryocyte numbers, and eosinophilia.95  Germline ETV6 mutations are predominantly found in ALL but are also reported in myeloid malignancies including MDS and AML.96-101  Data are scant to inform disease penetrance. Thus far, the numbers of total patients reported is too few to ascertain associated syndromic features.

ETV6 encodes a transcriptional repressor critical for hematopoiesis, megakaryopoiesis, and embryonic development.94  Germline ETV6 mutations are typically located in the DNA binding ETS domain and result in autosomal dominant inhibition of ETV6 function through dimerization. A P214L mutation located in the linker region implicated in release of an autoinhibitory domain has also been found in multiple unrelated kindreds.

ANKRD26

An autosomal dominant disorder characterized by quantitative and qualitative platelet disorders and increased risk for MDS and AML is also caused by mutations in ANKRD26 (see Noris et al102  and references therein). This disorder is characterized by moderate thrombocytopenia with normal platelet size, increased bleeding, and abnormal platelet function and aggregation.103,104  Bone marrow exam is notable for megakaryocyte dysplasia with small megakaryocytes containing hypolobated nuclei. Thus far, of the reported cases, about 5% developed acute leukemia (2.2% had MDS and 1.3% had CML), which is notably higher than expected in the general population.102 

ANKRD26 encodes a protein with N-terminal ankyrin repeat domains believed to function in protein-protein interactions. Although the function of ANKRD26 is unknown, expression profiling demonstrates a presence in megakaryocytes, and less so in erythroid cells.105  Mutations in ANRK26 are typically point mutations located in the 5′ untranslated region of the gene, although deletions and point mutations within the coding region have been reported.22  The 5′UTR mutations impair the binding of repressive transcription factors such as RUNX1 and FLI1 to this regulatory region, resulting in aberrantly increased ANKRD26 expression, which in turn disrupts platelet production.106 

DDX41

Germline mutations in DDX41 have been associated with autosomal dominant familial MDS/AML presenting in older age.107-110  Patients typically present with an increased risk for normal karyotype MDS or AML. Additional hematologic malignancies have also been described (Table 1). In comparison with other germline MDS syndromes, those associated with DDX41 mutation present at older age. The bone marrow is typically hypocellular. Some cases with antecedent cytopenias and macrocytosis preceding presentation with myeloid malignancy have been reported. No common nonhematologic phenotype has been associated with DDX41 germline mutations, and family history is often the sole clinical clue to DDX41 germline predisposition. DDX41 encodes a helicase possibly functioning in RNA splicing. Germline mutations in DDX41 include frameshift, missense, nonsense, and whole-gene deletion/contiguous gene mutations.111  In contrast, somatic mutations associated with malignancy are typically missense mutations.

SRP72

Germline mutations in the ribonuclear protein complex gene SRP72 have been described with autosomal-dominant MDS and bone marrow failure. Two families were found to have heterozygous mutations in SRP72 associated with autosomal dominant aplastic anemia, and the patients with MDS presented in their third and fifth decade of life.112  The SRP72 gene encodes a subunit of the signal recognition particle complex that is responsible for the trafficking to the endoplasmic reticulum. Mutations thus far described are frameshift and missense mutations.112 

ELANE

Severe congenital neutropenia (SCN) is a genetically heterogeneous disorder characterized by neutropenia typically less the 500/μL (see Skokowa et al113  and references therein). SCN is caused by mutations in several different genes, most commonly ELANE, which encodes neutrophil elastase114  (NE; Table 1). These patients usually present in early infancy with severe neutropenia and infectious complications, although an antecedent normal neutrophil count does not rule out a genetic neutropenia disorder. Bone marrow findings with ELANE mutations include maturation arrest of neutrophil precursors at the promyelocyte stage in the bone marrow (Figure 1).115  Overall cumulative incidence of sepsis death in SCN is 10%, and 22% for development of MDS/AML after 15 years of treatment with granulocyte colony-stimulating factor (G-CSF).116  The overall hazard of MDS/AML is 2.3%/year after 10 years receiving G-CSF.116  A poor absolute neutrophil count response despite high doses of G-CSF is associated with an increased risk for development of MDS/AML likely related to disease severity.116  Mutations are scattered throughout the ELANE gene and include missense, nonsense, frameshift, splice, insertion, and deletion mutations.117 

The mechanism by which ELANE mutations lead to the phenotype of SCN is not well understood. Data support multiple etiologies for hematopoietic cellular stressors such as misfolding of NE protein leading to activation of the unfolded protein response,118,119  mislocalized NE,120  or altered substrates of mutant NE protein121  with increased apoptosis.

Li Fraumeni Syndrome/TP53

First described by Frederick Li and Joseph Fraumeni, this autosomal-dominant disorder is characterized by a high risk for a spectrum of solid tumors and hematologic malignancies at a young age. Germline mutations in TP53 are associated with Li Fraumeni syndrome (LFS). The tumor spectrum includes soft tissue sarcomas, premenopausal breast cancer, central nervous system (CNS) tumors, adrenocortical carcinomas, pancreatic tumors, and others, as well as MDS and lymphoid or myeloid malignancies (see Quinn et al,9  Guha et al,122  and Kratz et al123  and references therein). LFS is highly penetrant, with a cumulative cancer incidence, mostly of solid tumors, of 50% by age 31 years for females and 50% by age 46 years for males, and nearly 100% by age 70 years for either sex.124  Leukemia, typically hypodiploid ALL but also AML and CML, occur in around 2% to 4% of patients with LFS.122  MDS in patients with LFS tends to arise secondary to treatment of another primary neoplasm, although primary MDS is also seen. The p53 protein plays a central role in checkpoint controls of cell growth, survival, and stress responses involving cell cycle arrest, apoptosis, senescence, DNA repair, and metabolic stressors. Missense mutations are the most common type of germline TP53 mutation (74%), often located within exons 5 to 8 in the cord DNA-binding domain, and may result in either dominant-negative or loss-of-function mutations. Deletions involving all or part of TP53, nonsense mutations, and splice site mutations are also found.

Genetic predisposition to MDS should be considered in a patient presenting at a young age with bone marrow failure, MDS, or AML, or with unexpected hematologic toxicity during treatment of a malignancy at a young age. Clinical clues include physical anomalies, endocrinopathies, short stature or failure to thrive, or immune deficiencies in a patient with hematologic abnormalities such as cytopenias, unexplained red cell macrocytosis, or frank malignancy (Table 1). Family history of first- or second-degree relatives with malignancy, cytopenias, congenital anomalies, or excessive toxicity with chemotherapy or radiation also raise concern for a genetic MDS predisposition disorder. The lack of characteristic clinical features or negative family history does not exclude the presence of a germline MDS syndrome. Germline mutations can arise de novo or may result from parental gonadal mosaicism. In addition, several of these germline MDS syndromes have no overt syndromic features or have variable penetrance or delayed expressivity. Cytogenetic clonal abnormalities common to certain genetic MDS disorders may prompt further investigation. For example, MDS with monosomy 7 frequently arises in patients with germline mutations in GATA2, SAMD9, SAMD9L, or genetic bone marrow failure syndromes.125  Del20q or isochromosome 7 are common in SDS.126  1q+ and 3q26q29 amplifications are common in FA.127-129 

Patients and families with suspected germline genetic MDS syndromes should receive genetic counseling regarding the indication for sending genetic testing, limitations of such testing, and family counseling. The strengths and weaknesses of different sequencing options were recently discussed.130  The clinician should be aware of which genes are included and whether all regions of interest within those genes are covered by the genetic tests ordered. With features noted previously or a strong clinical suspicion, testing from a somatic tissue such as fibroblasts should be sent. Targeted panels designed for evaluation of somatic MDS mutations should not be used to evaluate germline mutations for a given gene, as the somatic mutation panels may be missing coverage at key regions of genes of interest or at mutation sites that differ in the acquired vs the germline setting. Because of the frequency of mutations that may be gene deletions or duplications, clinicians should also be sure that testing methods detect copy number variations.

Surveillance

Diagnosis of genetic predisposition to MDS provides an opportunity for prospective clinical surveillance and intervention before the development of advanced disease. AML carries an especially poor prognosis for many of these disorders, so early diagnosis offers an opportunity for timely bone marrow transplant before development of leukemia. The reader is referred to recent discussions of surveillance and management of patients with genetic predisposition to MDS.131,132 

Treatment

Curative treatment of MDS for patients with germline MDS predisposition is a hematopoietic stem cell transplant. It is challenging that many genetic bone marrow failure syndromes are associated with excessive conditioning regimen-related toxicities and may require specialized reduced intensity conditioning regimens to minimize treatment-related toxicities that may, however, compromise ablation of the malignant clones.44,133-135  In addition, extrahematopoietic manifestations of germline predisposition disorders (Table 1) may further increase transplant risk. It is imperative to ensure that a related stem cell donor does not have the same genetic MDS predisposition disorder. Close monitoring for late effects is important in many of these patients, given the unique toxicities particular to their syndrome.136 

Clonal evolution: cytogenetics

The genomic evolution of clonal expansion within germline MDS syndromes is an area of intense clinical and scientific interest. These disorders allow study of clonal evolution before the development of MDS, enabling researchers to investigate somatic events prospectively as they arise. This provides a unique opportunity to gain insights into the molecular events driving MDS and a clinical opportunity to potentially identify prognostic mutations, intervention strategies, and targeted therapies.

Cytogenetically abnormal clones frequently arise in these genetic predisposition disorders. Some of these clones may wax and wane, and their clinical significance is unclear.131,137  Critical to the workup of pediatric and young adult patients who present with hypocellular bone marrows is evaluation of both cytogenetics and fluorescence in situ hybridization studies, as the failing marrow often yields few metaphases, many of which may arise from lymphocytes rather than the marrow.131  This differs from aging-related sporadic MDS in which the marrow is often hypercellular and cytogenetics studies are often considered sufficient.

Clonal cytogenetics in germline MDS have provided unique insights into potential drivers of clonality that may be adaptive or maladaptive (Figure 2). Adaptive mutations provide a fitness advantage by complementing the intrinsic hematopoietic defect, whereas maladaptive mutations promote malignant transformation.

Figure 2.

Model of germline genetic context in clonal evolution. Acquired somatic clonal mutations may be adaptive (improve hematopoiesis) or maladaptive (lead to transformation), depending on the underlying genetic MDS predisposition. In SBDS-mutant hematopoietic stem/progenitor cells, iso7q or del20q are common adaptive clonal aberrations, whereas TP53 somatic mutation is maladaptive. Fanconi anemia development of +3q is associated with progression to malignancy, whereas somatic reversion is not. In SAMD9/9L disorders, correction of the mutation by reversion or loss of mutant via truncation is adaptive, whereas development of monosomy is associated with MDS or AML. TERC-mutant HSPCs may undergo somatic reversion to regain 2 wild-type TERC alleles. GOF, gain of function; LOF, loss of function; HSPC, hematopoietic stem/progenitor cells; WT, wild type. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 2.

Model of germline genetic context in clonal evolution. Acquired somatic clonal mutations may be adaptive (improve hematopoiesis) or maladaptive (lead to transformation), depending on the underlying genetic MDS predisposition. In SBDS-mutant hematopoietic stem/progenitor cells, iso7q or del20q are common adaptive clonal aberrations, whereas TP53 somatic mutation is maladaptive. Fanconi anemia development of +3q is associated with progression to malignancy, whereas somatic reversion is not. In SAMD9/9L disorders, correction of the mutation by reversion or loss of mutant via truncation is adaptive, whereas development of monosomy is associated with MDS or AML. TERC-mutant HSPCs may undergo somatic reversion to regain 2 wild-type TERC alleles. GOF, gain of function; LOF, loss of function; HSPC, hematopoietic stem/progenitor cells; WT, wild type. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

These adaptive clones are illustrated by 2 different alterations in SDS: isochromosome 7q and del20q. The iso7q region of duplication has been observed to duplicate the hypomorphic SBDS allele, which is predicted to potentially increase SDS protein and has not been associated with increased leukemia risk.138  Del20q clones also frequently arise in SDS. The deleted region spans EIF6, resulting in haploinsufficiency, and may provide a clonal fitness advantage by reducing translational stress.139 

Adaptive somatic clones have also been described in patients with germline mutations in SAMD9/SAMD9L which reside on chromosome 7 (reviewed in Wlodarski et al21  and Davidsson et al24 ). Germline SAMD9 or SAMD9L gain-of-function mutations cause growth suppression of hematopoietic cells and resulting marrow failure.28,31  Patients with germline SAMD9 or SAMD9L mutations have a propensity to develop clones that have lost or inactivated the mutant SAMD9/SAMD9L allele. The mechanisms of this adaptation can be through truncating or loss of function mutations in cis with the mutant SAMD9 or SAMD9L allele, or through genetic reversion through duplication of the wild-type allele. Improvement in blood counts has been observed to accompany this somatic inactivation of the mutant SAMD9/SAMD9L allele. A second strategy to eliminate the mutant gene is to delete all or part of chromosome 7 carrying the mutant SAMD9/SAMD9L allele. Although this is predicted to result in improved cell growth, this comes at the cost of promoting development of monosomy 7 MDS.

The somatic reversion of FA genes also results in disease-context-specific adaptive clones. Somatic reversion occurs through point mutation, gene conversion, or recombination resulting in outgrowth of cells that have restored a wild-type FA allele.140,141  Somatic reversion is clinically important because it can lead to false-negative chromosomal breakage testing of lymphocytes. Mechanistically, the reversion event corrects the DNA repair phenotype and confers a growth advantage over the remaining germline mutant cells, but does not seem to drive myeloid malignancy. However, malignant clones may still arise from the remaining uncorrected marrow cells.140  In addition, somatic reversion via mitotic recombination has also been documented in several cases of DC with TERC mutation.142 

Clonal evolution: somatic genomics

The availability of deep genomic sequencing allows previously unprecedented detailed and comprehensive analysis of the somatic mutational landscape in MDS. Patterns of somatic mutations differ between genetic MDS predisposition disorders, suggesting that germline genetic context may drive different mechanisms of clonal hematopoiesis.143  Recent studies have reported clonal hematopoiesis in young patients with germline GATA2, RUNX1, and SBDS mutations and SCN.144-146  Recent studies examining the molecular mechanisms underlying reduced penetrance in GATA2-mutated MDS and AML may involve epigenetic changes.147  Somatic mutations, similar to cytogenetic changes, may be adaptive or oncogenic. For example, patients with SCN frequently acquire mutations in the G-CSF receptor, CSF3R.113,145,148,149 CSF3R mutations were detected both in the neutropenic and AML phase of the disease. The majority of mutations caused truncations of the cytoplasmic domain of the CSF3R receptor, resulting in cytokine-independent signaling.150,151  Although CSF3R clonal mutations are prevalent in AML in patients with SCN, they have also been observed for many years in patients who have not transformed to AML, indicating that such mutations alone are not sufficient to cause AML.148,152 

In contrast to SCN, clonal somatic mutations in SDS commonly involve TP53 and, when present in MDS, are associated with poor prognosis.4,145  Functional studies are needed to determine whether these mutations may relieve the translational stress of the ribosomal abnormality or provide a growth advantage. Longitudinal studies of TP53 mutations in SDS are lacking, so the prognostic significance of TP53 mutation arising outside the context of MDS is currently unclear.

Data are lacking to inform the clinical implications of somatic clonal abnormalities in germline MDS syndromes. Somatic clonal mutations should be considered within the context of the blood counts, marrow morphology, and clonal dynamics. Longitudinal studies integrating somatic alterations of serial samples with clinical outcomes are needed to understand the prognostic significance of somatic clonal abnormalities and potential targeted therapies toward premalignant clones.

As with any rare disease, collaborative studies, longitudinal disease registries and clinically annotated sample repositories are essential to advance our understanding with the goal of improving clinical outcomes. Disease penetrance and cumulative cancer risk remain to be determined for many of these disorders. Major challenges include the elucidation of additional genes causing MDS, identification of prognostic biomarkers for risk stratification to inform surveillance and timing of HSCT, and development of personalized treatments for patients with genetic predisposition to MDS.

The authors apologize to all those whose work could not be cited because of space limitations. Recent reviews were cited in each section, and readers are referred to primary references therein. The authors are grateful to Mark Fleming and Suneet Agarwal for their contribution of images for figures.

The authors gratefully acknowledge support from the National Institute of Diabetes and Digestive and Kidney Diseases at the National Institutes of Health (R24DK099808) (A.S.).

Contribution: All authors provided conception and design, manuscript writing, and final approval of manuscript, and all authors are accountable for all aspects of the work.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Akiko Shimamura, Karp 8210, 300 Longwood Ave, Boston, MA 08115; e-mail: akiko.shimamura@childrens.harvard.edu.

1.
Arber
DA
,
Orazi
A
,
Hasserjian
R
, et al
.
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia
.
Blood
.
2016
;
127
(
20
):
2391
-
2405
.
2.
da Silva-Coelho
P
,
Kroeze
LI
,
Yoshida
K
, et al
.
Clonal evolution in myelodysplastic syndromes
.
Nat Commun
.
2017
;
8
:
15099
.
3.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
, et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
.
2014
;
371
(
26
):
2488
-
2498
.
4.
Lindsley
RC
,
Saber
W
,
Mar
BG
, et al
.
Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation
.
N Engl J Med
.
2017
;
376
(
6
):
536
-
547
.
5.
Shimamura
A
,
Alter
BP
.
Pathophysiology and management of inherited bone marrow failure syndromes
.
Blood Rev
.
2010
;
24
(
3
):
101
-
122
.
6.
Keel
SB
,
Scott
A
,
Sanchez-Bonilla
M
, et al
.
Genetic features of myelodysplastic syndrome and aplastic anemia in pediatric and young adult patients
.
Haematologica
.
2016
;
101
(
11
):
1343
-
1350
.
7.
Schwartz
JR
,
Ma
J
,
Lamprecht
T
, et al
.
The genomic landscape of pediatric myelodysplastic syndromes
.
Nat Commun
.
2017
;
8
(
1
):
1557
.
8.
McReynolds
LJ
,
Savage
SA
.
Pediatric leukemia susceptibility disorders: manifestations and management
.
Hematology Am Soc Hematol Educ Program
.
2017
;
2017
:
242
-
250
.
9.
Quinn
E
,
Nichols
KE
.
Cancer predisposition syndromes associated with myeloid malignancy
.
Semin Hematol
.
2017
;
54
(
2
):
115
-
122
.
10.
Feurstein
S
,
Drazer
MW
,
Godley
LA
.
Genetic predisposition to leukemia and other hematologic malignancies
.
Semin Oncol
.
2016
;
43
(
5
):
598
-
608
.
11.
Vainchenker
W
,
Kralovics
R
.
Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms
.
Blood
.
2017
;
129
(
6
):
667
-
679
.
12.
Spinner
MA
,
Sanchez
LA
,
Hsu
AP
, et al
.
GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity
.
Blood
.
2014
;
123
(
6
):
809
-
821
.
13.
McReynolds
LJ
,
Calvo
KR
,
Holland
SM
.
Germline GATA2 mutation and bone marrow failure
.
Hematol Oncol Clin North Am
.
2018
;
32
(
4
):
713
-
728
.
14.
Vinh
DC
,
Patel
SY
,
Uzel
G
, et al
.
Autosomal dominant and sporadic monocytopenia with susceptibility to mycobacteria, fungi, papillomaviruses, and myelodysplasia
.
Blood
.
2010
;
115
(
8
):
1519
-
1529
.
15.
Ostergaard
P
,
Simpson
MA
,
Connell
FC
, et al
.
Mutations in GATA2 cause primary lymphedema associated with a predisposition to acute myeloid leukemia (Emberger syndrome)
.
Nat Genet
.
2011
;
43
(
10
):
929
-
931
.
16.
Zhang
MY
,
Keel
SB
,
Walsh
T
, et al
.
Genomic analysis of bone marrow failure and myelodysplastic syndromes reveals phenotypic and diagnostic complexity
.
Haematologica
.
2015
;
100
(
1
):
42
-
48
.
17.
Collin
M
,
Dickinson
R
,
Bigley
V
.
Haematopoietic and immune defects associated with GATA2 mutation
.
Br J Haematol
.
2015
;
169
(
2
):
173
-
187
.
18.
Pasquet
M
,
Bellanné-Chantelot
C
,
Tavitian
S
, et al
.
High frequency of GATA2 mutations in patients with mild chronic neutropenia evolving to MonoMac syndrome, myelodysplasia, and acute myeloid leukemia
.
Blood
.
2013
;
121
(
5
):
822
-
829
.
19.
Ganapathi
KA
,
Townsley
DM
,
Hsu
AP
, et al
.
GATA2 deficiency-associated bone marrow disorder differs from idiopathic aplastic anemia
.
Blood
.
2015
;
125
(
1
):
56
-
70
.
20.
Wlodarski
MW
,
Hirabayashi
S
,
Pastor
V
, et al
;
EWOG-MDS
.
Prevalence, clinical characteristics, and prognosis of GATA2-related myelodysplastic syndromes in children and adolescents
.
Blood
.
2016
;
127
(
11
):
1387
-
1397, quiz 1518
.
21.
Wlodarski
MW
,
Sahoo
SS
,
Niemeyer
CM
.
Monosomy 7 in pediatric myelodysplastic syndromes
.
Hematol Oncol Clin North Am
.
2018
;
32
(
4
):
729
-
743
.
22.
Godley
LA
.
Inherited predisposition to acute myeloid leukemia
.
Semin Hematol
.
2014
;
51
(
4
):
306
-
321
.
23.
Freireich
E
,
Whang
J
,
Tijo
JH
,
Levin
RH
,
Brittin
GM
,
Frei
IE
.
Refractory anemia, granulocytic hyperplasia of bone marrow, and a missing chromosome in marrow cells. A new clinical syndrome?
Clin Res
.
1964
;
12
:
284
.
24.
Davidsson
J
,
Puschmann
A
,
Tedgård
U
,
Bryder
D
,
Nilsson
L
,
Cammenga
J
.
SAMD9 and SAMD9L in inherited predisposition to ataxia, pancytopenia, and myeloid malignancies
.
Leukemia
.
2018
;
32
(
5
):
1106
-
1115
.
25.
Topaz
O
,
Indelman
M
,
Chefetz
I
, et al
.
A deleterious mutation in SAMD9 causes normophosphatemic familial tumoral calcinosis
.
Am J Hum Genet
.
2006
;
79
(
4
):
759
-
764
.
26.
Narumi
S
,
Amano
N
,
Ishii
T
, et al
.
SAMD9 mutations cause a novel multisystem disorder, MIRAGE syndrome, and are associated with loss of chromosome 7
.
Nat Genet
.
2016
;
48
(
7
):
792
-
797
.
27.
Chen
DH
,
Below
JE
,
Shimamura
A
, et al
.
Ataxia-pancytopenia syndrome is caused by missense mutations in SAMD9L
.
Am J Hum Genet
.
2016
;
98
(
6
):
1146
-
1158
.
28.
Tesi
B
,
Davidsson
J
,
Voss
M
, et al
.
Gain-of-function SAMD9L mutations cause a syndrome of cytopenia, immunodeficiency, MDS, and neurological symptoms
.
Blood
.
2017
;
129
(
16
):
2266
-
2279
.
29.
Buonocore
F
,
Kühnen
P
,
Suntharalingham
JP
, et al
.
Somatic mutations and progressive monosomy modify SAMD9-related phenotypes in humans
.
J Clin Invest
.
2017
;
127
(
5
):
1700
-
1713
.
30.
Pastor
VB
,
Sahoo
SS
,
Boklan
J
, et al
.
Constitutional SAMD9L mutations cause familial myelodysplastic syndrome and transient monosomy 7
.
Haematologica
.
2018
;
103
(
3
):
427
-
437
.
31.
Wong
JC
,
Bryant
V
,
Lamprecht
T
, et al
.
Germline SAMD9 and SAMD9L mutations are associated with extensive genetic evolution and diverse hematologic outcomes
.
JCI Insight
.
2018
;
3
(
14
):
121086
.
32.
Nagamachi
A
,
Matsui
H
,
Asou
H
, et al
.
Haploinsufficiency of SAMD9L, an endosome fusion facilitator, causes myeloid malignancies in mice mimicking human diseases with monosomy 7
.
Cancer Cell
.
2013
;
24
(
3
):
305
-
317
.
33.
Lobitz
S
,
Velleuer
E
.
Guido Fanconi (1892-1979): a jack of all trades
.
Nat Rev Cancer
.
2006
;
6
(
11
):
893
-
898
.
34.
Alter
BP
.
Fanconi anemia and the development of leukemia
.
Best Pract Res Clin Haematol
.
2014
;
27
(
3-4
):
214
-
221
.
35.
Bagby
G
.
Recent advances in understanding hematopoiesis in Fanconi anemia
.
F1000 Res
.
2018
;
7
:
105
.
36.
Savage
SA
,
Walsh
MF
.
Myelodysplastic syndrome, acute myeloid leukemia, and cancer surveillance in Fanconi anemia
.
Hematol Oncol Clin North Am
.
2018
;
32
(
4
):
657
-
668
.
37.
Giampietro
PF
,
Verlander
PC
,
Davis
JG
,
Auerbach
AD
.
Diagnosis of Fanconi anemia in patients without congenital malformations: an international Fanconi Anemia Registry Study
.
Am J Med Genet
.
1997
;
68
(
1
):
58
-
61
.
38.
Wagner
JE
,
Tolar
J
,
Levran
O
, et al
.
Germline mutations in BRCA2: shared genetic susceptibility to breast cancer, early onset leukemia, and Fanconi anemia
.
Blood
.
2004
;
103
(
8
):
3226
-
3229
.
39.
Ceccaldi
R
,
Sarangi
P
,
D’Andrea
AD
.
The Fanconi anaemia pathway: new players and new functions
.
Nat Rev Mol Cell Biol
.
2016
;
17
(
6
):
337
-
349
.
40.
Sasaki
MS
,
Tonomura
A
.
A high susceptibility of Fanconi’s anemia to chromosome breakage by DNA cross-linking agents
.
Cancer Res
.
1973
;
33
(
8
):
1829
-
1836
.
41.
Auerbach
AD
,
Adler
B
,
Chaganti
RS
.
Prenatal and postnatal diagnosis and carrier detection of Fanconi anemia by a cytogenetic method
.
Pediatrics
.
1981
;
67
(
1
):
128
-
135
.
42.
Ebens
CL
,
MacMillan
ML
,
Wagner
JE
.
Hematopoietic cell transplantation in Fanconi anemia: current evidence, challenges and recommendations
.
Expert Rev Hematol
.
2017
;
10
(
1
):
81
-
97
.
43.
Peffault de Latour
R
,
Soulier
J
.
How I treat MDS and AML in Fanconi anemia
.
Blood
.
2016
;
127
(
24
):
2971
-
2979
.
44.
Rosenberg
PS
,
Socié
G
,
Alter
BP
,
Gluckman
E
.
Risk of head and neck squamous cell cancer and death in patients with Fanconi anemia who did and did not receive transplants
.
Blood
.
2005
;
105
(
1
):
67
-
73
.
45.
Rosenberg
PS
,
Greene
MH
,
Alter
BP
.
Cancer incidence in persons with Fanconi anemia
.
Blood
.
2003
;
101
(
3
):
822
-
826
.
46.
Anur
P
,
Friedman
DN
,
Sklar
C
, et al
.
Late effects in patients with Fanconi anemia following allogeneic hematopoietic stem cell transplantation from alternative donors
.
Bone Marrow Transplant
.
2016
;
51
(
7
):
938
-
944
.
47.
Diamond
LK
,
Blackfan
KD
.
Hypoplastic anemia
.
Am J Dis Child
.
1938
;
56
:
464
-
467
.
48.
Da Costa
L
,
Narla
A
,
Mohandas
N
.
An update on the pathogenesis and diagnosis of Diamond-Blackfan anemia
.
F1000Res
.
2018
;7:F1000 Faculty Rev-1350.
49.
Vlachos
A
,
Blanc
L
,
Lipton
JM
.
Diamond Blackfan anemia: a model for the translational approach to understanding human disease
.
Expert Rev Hematol
.
2014
;
7
(
3
):
359
-
372
.
50.
Vlachos
A
,
Ball
S
,
Dahl
N
, et al
;
Participants of Sixth Annual Daniella Maria Arturi International Consensus Conference
.
Diagnosing and treating Diamond Blackfan anaemia: results of an international clinical consensus conference
.
Br J Haematol
.
2008
;
142
(
6
):
859
-
876
.
51.
Glader
BE
,
Backer
K
,
Diamond
LK
.
Elevated erythrocyte adenosine deaminase activity in congenital hypoplastic anemia
.
N Engl J Med
.
1983
;
309
(
24
):
1486
-
1490
.
52.
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
-
3819
.
53.
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
-
2208
.
54.
Draptchinskaia
N
,
Gustavsson
P
,
Andersson
B
, et al
.
The gene encoding ribosomal protein S19 is mutated in Diamond-Blackfan anaemia
.
Nat Genet
.
1999
;
21
(
2
):
169
-
175
.
55.
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
-
2443
.
56.
Dutt
S
,
Narla
A
,
Lin
K
, et al
.
Haploinsufficiency for ribosomal protein genes causes selective activation of p53 in human erythroid progenitor cells
.
Blood
.
2011
;
117
(
9
):
2567
-
2576
.
57.
Yang
Z
,
Keel
SB
,
Shimamura
A
, et al
.
Delayed globin synthesis leads to excess heme and the macrocytic anemia of Diamond Blackfan anemia and del(5q) myelodysplastic syndrome
.
Sci Transl Med
.
2016
;
8
(
338
):
338ra67
.
58.
Heijnen
HF
,
van Wijk
R
,
Pereboom
TC
, et al
.
Ribosomal protein mutations induce autophagy through S6 kinase inhibition of the insulin pathway
.
PLoS Genet
.
2014
;
10
(
5
):
e1004371
.
59.
Ludwig
LS
,
Gazda
HT
,
Eng
JC
, et al
.
Altered translation of GATA1 in Diamond-Blackfan anemia
.
Nat Med
.
2014
;
20
(
7
):
748
-
753
.
60.
Khajuria
RK
,
Munschauer
M
,
Ulirsch
JC
, et al
.
Ribosome levels selectively regulate translation and lineage commitment in human hematopoiesis
.
Cell
.
2018
;
173
(
1
):
90
-
103
.
61.
Ebert
BL
,
Pretz
J
,
Bosco
J
, et al
.
Identification of RPS14 as a 5q- syndrome gene by RNA interference screen
.
Nature
.
2008
;
451
(
7176
):
335
-
339
.
62.
Krönke
J
,
Fink
EC
,
Hollenbach
PW
, et al
.
Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS
.
Nature
.
2015
;
523
(
7559
):
183
-
188
.
63.
Townsley
DM
,
Dumitriu
B
,
Young
NS
.
Bone marrow failure and the telomeropathies
.
Blood
.
2014
;
124
(
18
):
2775
-
2783
.
64.
Bertuch
AA
.
The molecular genetics of the telomere biology disorders
.
RNA Biol
.
2016
;
13
(
8
):
696
-
706
.
65.
Agarwal
S
.
Evaluation and management of hematopoietic failure in dyskeratosis congenita
.
Hematol Oncol Clin North Am
.
2018
;
32
(
4
):
669
-
685
.
66.
Savage
SA
.
Beginning at the ends: telomeres and human disease
.
F1000Res
.
2018
;7:F1000 Faculty Rev-524.
67.
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
-
39
.
68.
Alter
BP
,
Giri
N
,
Savage
SA
,
Rosenberg
PS
.
Cancer in dyskeratosis congenita
.
Blood
.
2009
;
113
(
26
):
6549
-
6557
.
69.
Alter
BP
,
Giri
N
,
Savage
SA
, et al
.
Malignancies and survival patterns in the National Cancer Institute inherited bone marrow failure syndromes cohort study
.
Br J Haematol
.
2010
;
150
(
2
):
179
-
188
.
70.
Alter
BP
,
Baerlocher
GM
,
Savage
SA
, et al
.
Very short telomere length by flow fluorescence in situ hybridization identifies patients with dyskeratosis congenita
.
Blood
.
2007
;
110
(
5
):
1439
-
1447
.
71.
Alter
BP
,
Giri
N
,
Savage
SA
,
Rosenberg
PS
.
Telomere length in inherited bone marrow failure syndromes
.
Haematologica
.
2015
;
100
(
1
):
49
-
54
.
72.
Higgs
C
,
Crow
YJ
,
Adams
DM
, et al
;
Clinical Care Consortium for Telomere-associated Ailments (CCCTAA)
.
Understanding the evolving phenotype of vascular complications in telomere biology disorders [published online ahead of print 25 August 2018]
.
Angiogenesis
.
doi:10.1007/s10456-018-9640-7
.
73.
Armanios
M
.
Syndromes of telomere shortening
.
Annu Rev Genomics Hum Genet
.
2009
;
10
(
1
):
45
-
61
.
74.
Fioredda
F
,
Iacobelli
S
,
Korthof
ET
, et al
.
Outcome of haematopoietic stem cell transplantation in dyskeratosis congenita
.
Br J Haematol
.
2018
;
183
(
1
):
110
-
118
.
75.
Gadalla
SM
,
Sales-Bonfim
C
,
Carreras
J
, et al
.
Outcomes of allogeneic hematopoietic cell transplantation in patients with dyskeratosis congenita
.
Biol Blood Marrow Transplant
.
2013
;
19
(
8
):
1238
-
1243
.
76.
Shwachman
H
,
Diamond
LK
,
Oski
FA
,
Khaw
KT
.
The syndrome of pancreatic insufficiency and bone marrow dysfunction
.
J Pediatr
.
1964
;
65
(
5
):
645
-
663
.
77.
Bodian
M
,
Sheldon
W
,
Lightwood
R
.
Congenital hypoplasia of the exocrine pancreas
.
Acta Paediatr (Stockh)
.
1964
;
53
(
3
):
282
-
293
.
78.
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
-
870
.
79.
Donadieu
J
,
Fenneteau
O
,
Beaupain
B
, et al
;
Associated investigators of the French Severe Chronic Neutropenia Registry
.
Classification of and risk factors for hematologic complications in a French national cohort of 102 patients with Shwachman-Diamond syndrome
.
Haematologica
.
2012
;
97
(
9
):
1312
-
1319
.
80.
Ip
WF
,
Dupuis
A
,
Ellis
L
, et al
.
Serum pancreatic enzymes define the pancreatic phenotype in patients with Shwachman-Diamond syndrome
.
J Pediatr
.
2002
;
141
(
2
):
259
-
265
.
81.
Boocock
GR
,
Morrison
JA
,
Popovic
M
, et al
.
Mutations in SBDS are associated with Shwachman-Diamond syndrome
.
Nat Genet
.
2003
;
33
(
1
):
97
-
101
.
82.
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
-
929
.
83.
Wong
CC
,
Traynor
D
,
Basse
N
,
Kay
RR
,
Warren
AJ
.
Defective ribosome assembly in Shwachman-Diamond syndrome
.
Blood
.
2011
;
118
(
16
):
4305
-
4312
.
84.
Menne
TF
,
Goyenechea
B
,
Sánchez-Puig
N
, et al
.
The Shwachman-Bodian-Diamond syndrome protein mediates translational activation of ribosomes in yeast
.
Nat Genet
.
2007
;
39
(
4
):
486
-
495
.
85.
Warren
AJ
.
Molecular basis of the human ribosomopathy Shwachman-Diamond syndrome
.
Adv Biol Regul
.
2018
;
67
:
109
-
127
.
86.
Austin
KM
,
Gupta
ML
Jr
,
Coats
SA
, et al
.
Mitotic spindle destabilization and genomic instability in Shwachman-Diamond syndrome
.
J Clin Invest
.
2008
;
118
(
4
):
1511
-
1518
.
87.
Dhanraj
S
,
Matveev
A
,
Li
H
, et al
.
Biallelic mutations in DNAJC21 cause Shwachman-Diamond syndrome
.
Blood
.
2017
;
129
(
11
):
1557
-
1562
.
88.
Tummala
H
,
Walne
AJ
,
Williams
M
, et al
.
DNAJC21 mutations link a cancer-prone bone marrow failure syndrome to corruption in 60S ribosome subunit maturation
.
Am J Hum Genet
.
2016
;
99
(
1
):
115
-
124
.
89.
Stepensky
P
,
Chacón-Flores
M
,
Kim
KH
, et al
.
Mutations in EFL1, an SBDS partner, are associated with infantile pancytopenia, exocrine pancreatic insufficiency and skeletal anomalies in aShwachman-Diamond like syndrome
.
J Med Genet
.
2017
;
54
(
8
):
558
-
566
.
90.
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
-
4103
.
91.
Bellanné-Chantelot
C
,
Schmaltz-Panneau
B
,
Marty
C
, et al
.
Mutations in the SRP54 gene cause severe congenital neutropenia as well as Shwachman-Diamond-like syndrome
.
Blood
.
2018
;
132
(
12
):
1318
-
1331
.
92.
Bellissimo
DC
,
Speck
NA
.
RUNX1 mutations in inherited and sporadic leukemia
.
Front Cell Dev Biol
.
2017
;
5
:
111
.
93.
Schlegelberger
B
,
Heller
PG
.
RUNX1 deficiency (familial platelet disorder with predisposition to myeloid leukemia, FPDMM)
.
Semin Hematol
.
2017
;
54
(
2
):
75
-
80
.
94.
Hock
H
,
Shimamura
A
.
ETV6 in hematopoiesis and leukemia predisposition
.
Semin Hematol
.
2017
;
54
(
2
):
98
-
104
.
95.
Kanagal-Shamanna
R
,
Loghavi
S
,
DiNardo
CD
, et al
.
Bone marrow pathologic abnormalities in familial platelet disorder with propensity for myeloid malignancy and germline RUNX1 mutation
.
Haematologica
.
2017
;
102
(
10
):
1661
-
1670
.
96.
Zhang
MY
,
Churpek
JE
,
Keel
SB
, et al
.
Germline ETV6 mutations in familial thrombocytopenia and hematologic malignancy
.
Nat Genet
.
2015
;
47
(
2
):
180
-
185
.
97.
Noetzli
L
,
Lo
RW
,
Lee-Sherick
AB
, et al
.
Germline mutations in ETV6 are associated with thrombocytopenia, red cell macrocytosis and predisposition to lymphoblastic leukemia
.
Nat Genet
.
2015
;
47
(
5
):
535
-
538
.
98.
Topka
S
,
Vijai
J
,
Walsh
MF
, et al
.
Germline ETV6 mutations confer susceptibility to acute lymphoblastic leukemia and thrombocytopenia
.
PLoS Genet
.
2015
;
11
(
6
):
e1005262
.
99.
Moriyama
T
,
Metzger
ML
,
Wu
G
, et al
.
Germline genetic variation in ETV6 and risk of childhood acute lymphoblastic leukaemia: a systematic genetic study
.
Lancet Oncol
.
2015
;
16
(
16
):
1659
-
1666
.
100.
Melazzini
F
,
Palombo
F
,
Balduini
A
, et al
.
Clinical and pathogenic features of ETV6-related thrombocytopenia with predisposition to acute lymphoblastic leukemia
.
Haematologica
.
2016
;
101
(
11
):
1333
-
1342
.
101.
Poggi
M
,
Canault
M
,
Favier
M
, et al
.
Germline variants in ETV6 underlie reduced platelet formation, platelet dysfunction and increased levels of circulating CD34+ progenitors
.
Haematologica
.
2017
;
102
(
2
):
282
-
294
.
102.
Noris
P
,
Favier
R
,
Alessi
MC
, et al
.
ANKRD26-related thrombocytopenia and myeloid malignancies
.
Blood
.
2013
;
122
(
11
):
1987
-
1989
.
103.
Pippucci
T
,
Savoia
A
,
Perrotta
S
, et al
.
Mutations in the 5′ UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2
.
Am J Hum Genet
.
2011
;
88
(
1
):
115
-
120
.
104.
Noris
P
,
Perrotta
S
,
Seri
M
, et al
.
Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families
.
Blood
.
2011
;
117
(
24
):
6673
-
6680
.
105.
Macaulay
IC
,
Tijssen
MR
,
Thijssen-Timmer
DC
, et al
.
Comparative gene expression profiling of in vitro differentiated megakaryocytes and erythroblasts identifies novel activatory and inhibitory platelet membrane proteins
.
Blood
.
2007
;
109
(
8
):
3260
-
3269
.
106.
Bluteau
D
,
Balduini
A
,
Balayn
N
, et al
.
Thrombocytopenia-associated mutations in the ANKRD26 regulatory region induce MAPK hyperactivation
.
J Clin Invest
.
2014
;
124
(
2
):
580
-
591
.
107.
Polprasert
C
,
Schulze
I
,
Sekeres
MA
, et al
.
Inherited and somatic defects in DDX41 in myeloid neoplasms
.
Cancer Cell
.
2015
;
27
(
5
):
658
-
670
.
108.
Cardoso
SR
,
Ryan
G
,
Walne
AJ
, et al
.
Germline heterozygous DDX41 variants in a subset of familial myelodysplasia and acute myeloid leukemia
.
Leukemia
.
2016
;
30
(
10
):
2083
-
2086
.
109.
Lewinsohn
M
,
Brown
AL
,
Weinel
LM
, et al
.
Novel germ line DDX41 mutations define families with a lower age of MDS/AML onset and lymphoid malignancies
.
Blood
.
2016
;
127
(
8
):
1017
-
1023
.
110.
Maciejewski
JP
,
Padgett
RA
,
Brown
AL
,
Müller-Tidow
C
.
DDX41-related myeloid neoplasia
.
Semin Hematol
.
2017
;
54
(
2
):
94
-
97
.
111.
Cheah
JJC
,
Hahn
CN
,
Hiwase
DK
,
Scott
HS
,
Brown
AL
.
Myeloid neoplasms with germline DDX41 mutation
.
Int J Hematol
.
2017
;
106
(
2
):
163
-
174
.
112.
Kirwan
M
,
Walne
AJ
,
Plagnol
V
, et al
.
Exome sequencing identifies autosomal-dominant SRP72 mutations associated with familial aplasia and myelodysplasia
.
Am J Hum Genet
.
2012
;
90
(
5
):
888
-
892
.
113.
Skokowa
J
,
Dale
DC
,
Touw
IP
,
Zeidler
C
,
Welte
K
.
Severe congenital neutropenias
.
Nat Rev Dis Primers
.
2017
;
3
:
17032
.
114.
Dale
DC
,
Person
RE
,
Bolyard
AA
, et al
.
Mutations in the gene encoding neutrophil elastase in congenital and cyclic neutropenia
.
Blood
.
2000
;
96
(
7
):
2317
-
2322
.
115.
Ancliff
PJ
.
Congenital neutropenia
.
Blood Rev
.
2003
;
17
(
4
):
209
-
216
.
116.
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
-
199
.
117.
Makaryan
V
,
Zeidler
C
,
Bolyard
AA
, et al
.
The diversity of mutations and clinical outcomes for ELANE-associated neutropenia
.
Curr Opin Hematol
.
2015
;
22
(
1
):
3
-
11
.
118.
Grenda
DS
,
Murakami
M
,
Ghatak
J
, et al
.
Mutations of the ELA2 gene found in patients with severe congenital neutropenia induce the unfolded protein response and cellular apoptosis
.
Blood
.
2007
;
110
(
13
):
4179
-
4187
.
119.
Köllner
I
,
Sodeik
B
,
Schreek
S
, et al
.
Mutations in neutrophil elastase causing congenital neutropenia lead to cytoplasmic protein accumulation and induction of the unfolded protein response
.
Blood
.
2006
;
108
(
2
):
493
-
500
.
120.
Benson
KF
,
Li
FQ
,
Person
RE
, et al
.
Mutations associated with neutropenia in dogs and humans disrupt intracellular transport of neutrophil elastase
.
Nat Genet
.
2003
;
35
(
1
):
90
-
96
.
121.
Horwitz
MS
,
Corey
SJ
,
Grimes
HL
,
Tidwell
T
.
ELANE mutations in cyclic and severe congenital neutropenia: genetics and pathophysiology
.
Hematol Oncol Clin North Am
.
2013
;
27
(
1
):
19
-
41, vii
.
122.
Guha
T
,
Malkin
D
.
Inherited TP53 Mutations and the Li-Fraumeni Syndrome
.
Cold Spring Harb Perspect Med
.
2017
;
7
(
4
):
a026187
.
123.
Kratz
CP
,
Achatz
MI
,
Brugieres
L
, et al
.
Cancer screening recommendations for individuals with Li-Fraumeni Syndrome
.
Clin Cancer Res
.
2017
;
23
(11):e38-e45.
124.
Mai
PL
,
Best
AF
,
Peters
JA
, et al
.
Risks of first and subsequent cancers among TP53 mutation carriers in the National Cancer Institute Li-Fraumeni syndrome cohort
.
Cancer
.
2016
;
122
(
23
):
3673
-
3681
.
125.
Inaba
T
,
Honda
H
,
Matsui
H
.
The enigma of monosomy 7
.
Blood
.
2018
;
131
(
26
):
2891
-
2898
.
126.
Maserati
E
,
Minelli
A
,
Pressato
B
, et al
.
Shwachman syndrome as mutator phenotype responsible for myeloid dysplasia/neoplasia through karyotype instability and chromosomes 7 and 20 anomalies
.
Genes Chromosomes Cancer
.
2006
;
45
(
4
):
375
-
382
.
127.
Cioc
AM
,
Wagner
JE
,
MacMillan
ML
,
DeFor
T
,
Hirsch
B
.
Diagnosis of myelodysplastic syndrome among a cohort of 119 patients with fanconi anemia: morphologic and cytogenetic characteristics
.
Am J Clin Pathol
.
2010
;
133
(
1
):
92
-
100
.
128.
Quentin
S
,
Cuccuini
W
,
Ceccaldi
R
, et al
.
Myelodysplasia and leukemia of Fanconi anemia are associated with a specific pattern of genomic abnormalities that includes cryptic RUNX1/AML1 lesions
.
Blood
.
2011
;
117
(
15
):
e161
-
e170
.
129.
Tönnies
H
,
Huber
S
,
Kuhl
JS
,
Gerlach
A
,
Ebell
W
,
Neitzel
H
.
Clonal chromosomal aberrations in bone marrow cells of Fanconi anemia patients: gains of the chromosomal segment 3q26q29 as an adverse risk factor
.
Blood
.
2003
;
101
(
10
):
3872
-
3874
.
130.
Furutani
E
,
Shimamura
A
.
Germline genetic predisposition to hematologic malignancy
.
J Clin Oncol
.
2017
;
35
(
9
):
1018
-
1028
.
131.
Godley
LA
,
Shimamura
A
.
Genetic predisposition to hematologic malignancies: management and surveillance
.
Blood
.
2017
;
130
(
4
):
424
-
432
.
132.
Porter
CC
,
Druley
TE
,
Erez
A
, et al
.
Considerations for surveillance for children with leukemia predisposing conditions
.
Clin Cancer Res
.
2017
;
23
(
11
):
e14
-
e22
.
133.
Locatelli
F
,
Strahm
B
.
How I treat myelodysplastic syndromes of childhood
.
Blood
.
2018
;
131
(
13
):
1406
-
1414
.
134.
Burroughs
LM
,
Shimamura
A
,
Talano
JA
, et al
.
Allogeneic hematopoietic cell transplantation using treosulfan-based conditioning for treatment of marrow failure disorders
.
Biol Blood Marrow Transplant
.
2017
;
23
(
10
):
1669
-
1677
.
135.
Vlachos
A
,
Muir
E
.
How I treat Diamond-Blackfan anemia
.
Blood
.
2010
;
116
(
19
):
3715
-
3723
.
136.
Dietz
AC
,
Duncan
CN
,
Alter
BP
, et al
.
The Second Pediatric Blood and Marrow Transplant Consortium International Consensus Conference on Late Effects after Pediatric Hematopoietic Cell Transplantation: Defining the Unique Late Effects of Children Undergoing Hematopoietic Cell Transplantation for Immune Deficiencies, Inherited Marrow Failure Disorders, and Hemoglobinopathies
.
Biol Blood Marrow Transplant
.
2017
;
23
(
1
):
24
-
29
.
137.
Babushok
DV
,
Bessler
M
,
Olson
TS
.
Genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia in children and young adults
.
Leuk Lymphoma
.
2016
;
57
(
3
):
520
-
536
.
138.
Minelli
A
,
Maserati
E
,
Nicolis
E
, et al
.
The isochromosome i(7)(q10) carrying c.258+2t>c mutation of the SBDS gene does not promote development of myeloid malignancies in patients with Shwachman syndrome
.
Leukemia
.
2009
;
23
(
4
):
708
-
711
.
139.
Pressato
B
,
Valli
R
,
Marletta
C
, et al
.
Deletion of chromosome 20 in bone marrow of patients with Shwachman-Diamond syndrome, loss of the EIF6 gene and benign prognosis
.
Br J Haematol
.
2012
;
157
(
4
):
503
-
505
.
140.
Gregory
JJ
Jr
,
Wagner
JE
,
Verlander
PC
, et al
.
Somatic mosaicism in Fanconi anemia: evidence of genotypic reversion in lymphohematopoietic stem cells
.
Proc Natl Acad Sci USA
.
2001
;
98
(
5
):
2532
-
2537
.
141.
Waisfisz
Q
,
Morgan
NV
,
Savino
M
, et al
.
Spontaneous functional correction of homozygous fanconi anaemia alleles reveals novel mechanistic basis for reverse mosaicism
.
Nat Genet
.
1999
;
22
(
4
):
379
-
383
.
142.
Jongmans
MC
,
Verwiel
ET
,
Heijdra
Y
, et al
.
Revertant somatic mosaicism by mitotic recombination in dyskeratosis congenita
.
Am J Hum Genet
.
2012
;
90
(
3
):
426
-
433
.
143.
Schaefer
EJ
,
Lindsley
RC
.
Significance of clonal mutations in bone marrow failure and inherited myelodysplastic syndrome/acute myeloid leukemia predisposition syndromes
.
Hematol Oncol Clin North Am
.
2018
;
32
(
4
):
643
-
655
.
144.
Churpek
JE
,
Pyrtel
K
,
Kanchi
KL
, et al
.
Genomic analysis of germ line and somatic variants in familial myelodysplasia/acute myeloid leukemia
.
Blood
.
2015
;
126
(
22
):
2484
-
2490
.
145.
Xia
J
,
Miller
CA
,
Baty
J
, et al
.
Somatic mutations and clonal hematopoiesis in congenital neutropenia
.
Blood
.
2018
;
131
(
4
):
408
-
416
.
146.
Beekman
R
,
Valkhof
MG
,
Sanders
MA
, et al
.
Sequential gain of mutations in severe congenital neutropenia progressing to acute myeloid leukemia
.
Blood
.
2012
;
119
(
22
):
5071
-
5077
.
147.
Al Seraihi
AF
,
Rio-Machin
A
,
Tawana
K
, et al
.
GATA2 monoallelic expression underlies reduced penetrance in inherited GATA2-mutated MDS/AML
.
Leukemia
.
2018
;
32
(
11
):
2502
-
2507
.
148.
Link
DC
,
Kunter
G
,
Kasai
Y
, et al
.
Distinct patterns of mutations occurring in de novo AML versus AML arising in the setting of severe congenital neutropenia
.
Blood
.
2007
;
110
(
5
):
1648
-
1655
.
149.
Touw
IP
.
Game of clones: the genomic evolution of severe congenital neutropenia
.
Hematology Am Soc Hematol Educ Program
.
2015
;
2015
:
1
-
7
.
150.
Fukunaga
R
,
Ishizaka-Ikeda
E
,
Nagata
S
.
Growth and differentiation signals mediated by different regions in the cytoplasmic domain of granulocyte colony-stimulating factor receptor
.
Cell
.
1993
;
74
(
6
):
1079
-
1087
.
151.
Dong
F
,
van Buitenen
C
,
Pouwels
K
,
Hoefsloot
LH
,
Löwenberg
B
,
Touw
IP
.
Distinct cytoplasmic regions of the human granulocyte colony-stimulating factor receptor involved in induction of proliferation and maturation
.
Mol Cell Biol
.
1993
;
13
(
12
):
7774
-
7781
.
152.
Germeshausen
M
,
Ballmaier
M
,
Welte
K
.
Incidence of CSF3R mutations in severe congenital neutropenia and relevance for leukemogenesis: Results of a long-term survey
.
Blood
.
2007
;
109
(
1
):
93
-
99
.
Sign in via your Institution