All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know AML.

The AML Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your AML Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The AML Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the AML Hub cannot guarantee the accuracy of translated content. The AML Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2020-10-14T08:17:48.000Z

Inherited hematologic malignancies predisposition syndromes

Oct 14, 2020
Share:

Bookmark this article

Hereditary predispositions to hematologic malignancies occur in approximately 5─10% of cases. During the Society of Hematologic Oncology (SOHO) Eighth Annual Meeting, Courtney DiNardo gave a talk about the importance of recognizing patients with inherited cancer predisposition syndromes.1

The identification of an inherited predisposition allows optimized treatment of the patient, more informed donor decisions (avoiding a related donor with the same predisposition), and opportunities for identification and screening of family members.1

Inherited predisposition syndromes associated with leukemia and myeloproliferative neoplasms (MPN) are reported in Table 1.

Table 1. Leukemia and MPN predisposition syndromes1

ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; B-ALL, B-cell ALL; CLL, chronic lymphocytic leukemia; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasms; T-ALL, T-cell ALL.

Hematologic malignancy

Predisposition syndrome

Adult bone marrow failure/ hypocellular MDS/AML

Fanconi anemia

Dyskeratosis congenita

Shwachman–Diamond syndrome

Diamond Blackfan anemia

Familial aplastic anemia and hypocellular MDS

Familial MDS/acute leukemia

RUNX1 familial platelet disorder/AML (and T-ALL)

GATA2 familial MDS/AML (MonoMAC syndrome)

CEBPα familial AML

ANKRD26 familial platelet disorder with propensity to MDS/AML

DDX41 familial MDS/AML

ETV6 familial MDS/AML/lymphoid malignancies

SAMD9/SAMD9L familial MDS & immunodeficiency syndrome

Familial ALL

Li–Fraumeni syndrome: germline TP53 mutations

PAX5: familial B-cell lymphoid leukemias/lymphomas

IKZF1-associated predisposition to B-ALL

Biallelic SH2B3 mutations (also MPN risk?)

Familial CLL

POT1 familial CLL/melanoma/cardiac myxomas

Familial MPN

JAK2 V617I, R564Q & R867Q/N, others

 

MPL S505N, W515R, P106L, others

 

TET2 D1858fs

 

700 kb duplication, with ATG2B and GSKIP overexpression, leads to increased progenitor sensitivity to thrombopoietin

In 15─20% of cases, patients with either aplastic anemia, hypocellular myelodysplastic syndrome (MDS), or bone marrow failure have an inherited predisposition syndrome, such as Fanconi anemia, dyskeratosis congenita (DC), Shwachman–Diamond syndrome, Diamond Blackfan anemia, or others. These patients should be recognized and referred for genetic testing in order to provide optimal treatment strategies.1

Genetic profiling of patients with inherited bone marrow failure syndromes is important because they

  • respond poorly to anti-thymocyte globulin and cyclosporine (a shorter telomere length by flow-FISH [fluorescent in situ hybridization] in patients with aplastic anemia correlates with poor response to standard immune suppressive therapy);
  • respond well to growth factors and androgens (effective in ˃ 50% of patients);
  • should be treated with specialized reduced-intensity regimens to avoid excessive morbidity (avoid alkylating agents); and
  • need surveillance for solid tumors.

Clinical evaluation1

Dyskeratosis congenita

DC (also called short telomere syndrome, telomere biology disorder, or telomeropathy) is caused by mutations in genes important for telomere maintenance. Telomeres protect chromosome ends from degradation, and their length regulates the ability of a cell to duplicate. The instability resulting from an inadequate telomere maintenance can lead to uncontrolled cell growth and development of cancer.

Telomere flow-FISH is the technique used to detect DC. With telomere flow-FISH it is possible to distinguish DC from other causes of bone marrow failure.

Familial GATA2 (MonoMAC) syndrome

Familial GATA2 syndrome, also known as MonoMAC or Emberger syndrome, is due to autosomal dominant mutations or deletion of GATA2 and is associated with immunodeficiency and frequent infections. The median age at diagnosis is 18 years, and the lifetime risk of MDS/acute myeloid leukemia (AML) is 75–90%. It is particularly common in younger patients with MDS, with 10─15% of them having germline GATA2 mutations, and occurs frequently in those with MDS carrying monosomy of chromosome 7:

  • 27% of children and young adults
  • 72% of patients aged 12─19

In cases of transplants from a related donor, it is important to test family members to be sure that they are not GATA2 mutation-positive. However, overall survival and stem cell transplant outcomes appear to be independent of GATA2 status.

RUNX1, ANKRD26, and ETV6 variants

MDS and acute leukemia predisposition syndromes include autosomal dominant pathogenic variants in RUNX1 (21q), ANKRD26 (10p12), and ETV6 (12p13), which are associated with mild to moderate thrombocytopenia, platelet aggregation defects, and abnormal megakaryocytes.

RUNX1 was the first described, and about 40% of patients with germline mutations develop MDS/AML/T-cell acute lymphoblastic leukemia at a median age of 33 years. Thus, monitoring patients found to harbor these mutations is important to identify those at risk.

DDX41 mutations

Autosomal dominant mutations in DDX41 (5q35) have been recently identified to confer a predisposition to developing MDS/AML at a median age of 65 years. DDX41 is mutated in 1% of all patients with MDS/AML, and retrospective studies have shown that these patients are particularly sensitive to lenalidomide. The majority of DDX41 mutations leading to MDS/AML are germline mutations, and therefore inherited. The estimated risk of developing MDS/AML in family members of an individual with a DDX41 mutation is   ̴20─30%.

Genetic characterization of disorders

An 81-gene panel, called EndLeukemia panel, was developed in 2017 to allow comprehensive molecular profiling of a variety of hematologic malignancies.2 Genes included in the panel, reported in Table 2, cover genetic alterations in AML, MDS, MPN, MDS/MPN, aplastic anemia, acute lymphoblastic leukemia, hairy cell leukemia, T-large granular lymphocytic leukemia, and T-prolymphocytic leukemia.2

When suspected germline variants are detected, a multidisciplinary review is required to identify patients that should be further evaluated; the workflow is reported in Figure 1.1,3

Table 2. Genes included in the EndLeukemia panel1

Chromosome

Gene

1

BRINP3, CSF3R, GFI1, JAK1, MPL, NRAS,

2

ASXL2, DNMT3A, IDH1, SF3B1

3

CBLB, GATA2, STAG1, TERC

4

FBXW7, KIT, TET2

5

DDX41, IL7R, NPM1, TERT

7

BRAF, CUX1, EZH2, IKZF1

8

RAD21

9

HNRNPK, JAK2, NOTCH1, PAX5

10

ANKRD26, PTEN, SMC3

11

CBL, EED, HRAS, MLL, SF1, WT1

12

ETNK1, ETV6, KRAS, PTPN11, PRPF40B, SH2B3

13

FLT3

15

IDH2, MAP2K1, PML

16

CREBBP

17

NF1, RARA, SRSF2, STAT3, STAT5A, STAT5B, SUZ12, TP53

18

SETBP1

19

CALR, CBLC, CEBPA, ELANE, JAK3, U2AF2

20

ASXL1, GNAS

21

RUNX1, U2AF1

22

SF3A1

X

BCOR, BCORL1, CRLF2, GATA1, IL2RG, KDM6A, PHF6, PIGA, SMC1A, STAG2, ZRSR2

Figure 1. Workflow for the multidisciplinary review of suspected germline variants3

HHMC, hereditary hematologic malignancies clinic; WHO, World Health Organization. 

Current consensus statement1

The recommendations for clinicians managing patients with familial hematologic malignancies syndrome are the following:

  • Clinical exam with complete blood count one or two times a year;
  • Bone marrow biopsy at the time of diagnosis, to identify any clonal changes or secondary mutations that may lead to the development of hematologic malignancies, and then every year for research surveillance;
  • Consider allogeneic stem cell transplant at time of development of dysplasia, or clonal somatic cytogenetic or molecular abnormality; and

For younger and interested patients, suggest reproductive counseling and preimplantation genetic testing.

  1. DiNardo C. Familial AML. Oral presentation #MTP11. SOHO Eighth Annual Meeting; Sep 9, 2020; Virtual.
  2. Ok CY, Singh R, Luthra R, et al. Endleukemia Assay v1: Enabling NGS-based comprehensive routine molecular profiling of leukemias in routine clinical care. 2017;130(S1):2679. DOI: 10.1182/blood.V130.Suppl_1.2679.2679
  3. DiNardo C, Routbort MJ, Bannon SA, et al. Improving the detection of patients with inherited predispositions to hematologic malignancies using next-generation sequencing-based leukemia prognostication panels. Cancer. 2018;124(13):2704-2713. DOI:1002/cncr.31331

More about...

Your opinion matters

Do you intend to implement next-generation sequencing for measurable residual disease monitoring in MDS patients?
1 vote - 4 days left ...

Newsletter

Subscribe to get the best content related to AML delivered to your inbox