TO THE EDITOR:

The frequency of germline RUNX1 variants in an unselected acute myeloid leukemia (AML) population is poorly defined and likely underestimated. The recent study by Simon et al1  is particularly important as a first attempt to define this underlying frequency. Because RUNX1 is part of most next-generation sequencing panels performed on leukemic samples, germline variants are invariably found, highlighted by this and other studies.1-4  Human and medical geneticists, genetic counselors, molecular pathologists, hematopathologists, and hematologists are particularly likely to encounter patients with germline RUNX1 variants and may benefit from guidance on how to interpret these variants and their clinical implications.

In the Simon et al1  study, 10.7% (44/430) of AML patients had a somatic or germline RUNX1 variant. Germline variants represented 27.3% (12/44) of RUNX1 variants, suggesting a 2.8% frequency of germline RUNX1 variants in an unselected AML population. However, it was not clearly delineated whether the identified germline variants were all disease causing (ie, pathogenic or likely pathogenic), although the term “mutation” implies pathogenicity. Inconsistent usage of “variant” and “mutation” can lead to miscommunication of scientific findings, as well as clinical testing results: “mutation” refers to pathogenic/likely pathogenic variations that are deleterious and found less frequently in a population or are nongermline changes in a tumor cell (somatic mutations) that are predictive/therapeutic, diagnostic, or prognostic biomarkers (Table 1).5 

Table 1.

Nomenclature of variants and likelihood of being disease causing

NomenclatureDefinitionLikelihood of being disease causing9,19-21 ,*
Mutation “Mutation” is used for germline variations that are pathogenic and found less frequently in a population or are nongermline changes in a tumor cell (somatic mutations).5  Should only be used when there is clear evidence for pathogenicity 
Variant An alteration in the most common DNA nucleotide sequence. The term “variant” can be used to describe an alteration that may be benign, pathogenic, or of unknown significance. The term “variant” is increasingly being used in place of the term “mutation.”22  Variant is further classified in a 5-tier system: benign, likely benign, uncertain significance, likely pathogenic, pathogenic 
Benign variant This variant does not cause disease. <0.1% 
Likely benign variant This variant is not expected to cause disease. Additional evidence may confirm this assertion of benign, but there is a small chance that new evidence may demonstrate that this variant does have clinical significance. Between 0.1% and 10% 
Variant of uncertain significance There is insufficient evidence to put this variant into a benign or pathogenic category. Further evidence, such as population, segregation, or functional data, may up- or downgrade this variant. This variant is not clinically actionable. Between 10% and 90% 
Likely pathogenic variant This variant is expected to cause disease. Additional evidence may confirm this assertion of pathogenicity, but there is a small chance that new evidence may demonstrate that this variant does not have clinical significance. Between 90% and 99% 
Pathogenic variant This variant does cause disease. >99% 
NomenclatureDefinitionLikelihood of being disease causing9,19-21 ,*
Mutation “Mutation” is used for germline variations that are pathogenic and found less frequently in a population or are nongermline changes in a tumor cell (somatic mutations).5  Should only be used when there is clear evidence for pathogenicity 
Variant An alteration in the most common DNA nucleotide sequence. The term “variant” can be used to describe an alteration that may be benign, pathogenic, or of unknown significance. The term “variant” is increasingly being used in place of the term “mutation.”22  Variant is further classified in a 5-tier system: benign, likely benign, uncertain significance, likely pathogenic, pathogenic 
Benign variant This variant does not cause disease. <0.1% 
Likely benign variant This variant is not expected to cause disease. Additional evidence may confirm this assertion of benign, but there is a small chance that new evidence may demonstrate that this variant does have clinical significance. Between 0.1% and 10% 
Variant of uncertain significance There is insufficient evidence to put this variant into a benign or pathogenic category. Further evidence, such as population, segregation, or functional data, may up- or downgrade this variant. This variant is not clinically actionable. Between 10% and 90% 
Likely pathogenic variant This variant is expected to cause disease. Additional evidence may confirm this assertion of pathogenicity, but there is a small chance that new evidence may demonstrate that this variant does not have clinical significance. Between 90% and 99% 
Pathogenic variant This variant does cause disease. >99% 
*

Most variants do not have data to support a quantitative assignment of variant certainty to any of the 5 categories given the heterogeneous nature of most diseases.

Germline variant classification is performed using 5 ranks of pathogenicity: pathogenic, likely pathogenic, variant of uncertain significance, likely benign, and benign. Variants of uncertain significance, as well as likely benign and benign variants, should not be attributed to disease causality (Table 1). Accurate variant classification is critically important for attribution of pathogenicity of the identified variants and their actionability, because the identification of a deleterious germline variant has clinical implications that extend far beyond the treatment of the diagnosed individual.

In response to interlaboratory curation differences, the Clinical Genome Resource (ClinGen) has launched Variant Curation Expert Panels (VCEPs) to develop gene- or disease-specific American College of Medical Genetics and Genomics (ACMG)/Association for Molecular Pathology (AMP) criteria.6  The Myeloid Malignancy (MM)-VCEP was formed in 2018 and published RUNX1-specific ACMG/AMP criteria in 2019.7,8  Given our familiarity with the RUNX1 variant curation rules, we have reviewed the variants described in the Simon et al1  study and found that only 7 of the 12 germline variants meet the criteria for pathogenic/likely pathogenic classification (Table 2). Thus, the actual yield of deleterious germline RUNX1 variants is 16% (7/44) of all RUNX1 variants and 1.6% (7/430) of all AML patients. Other than early truncating variants leading to non-sense–mediated decay, most causative RUNX1 variants are dependent on a variety of pathogenic evidence. In the case of RUNX1, this is usually a combination of computational and predictive, functional, population, and segregation data in a Bayesian framework.7,9 

Table 2.

Comparison of RUNX1 variant curation between the study by Simon et al and the MM-VCEP

ID*Variant cDNA/protein*Described in MDS/AML*Described in RUNX1 FPD*Functional impact on RUNX1*MM-VCEP ACMG/AMP criteria code*MM-VCEP RUNX1-specific criteriaFurther explanation of criteriaMM-VCEP classification
c.44_45delAG/p.Q15fsX — — Truncating PVS1_moderate, PS4_supporting, PM2 PS4_supporting, PM2 PVS1 cannot be used for early truncating variants only affecting RUNX1 isoform C. VUS 
c.179C>T/p.A60V Carnicer et al23  Lorente24  — BS1 BS1, BS3 This variant meets the calculated BS1 threshold (Latino subpopulation) and BS3 (normal transactivation and normal DNA binding/subcellular localization).25  The presence of the variant in patients with a RUNX1 phenotype is not sufficient to call a variant PATH, in particular not if the variant is present in gnomAD at a MAF incompatible with disease prevalence. BEN 
3+4 c.421T>G/p.S141A — RUNX1db Normal transactivation26  PS4_supporting, PP3, BS3_supporting PM1_supporting, PP3 Variant not present in RUNX1db. Although there is no effect on heterodimerization ability with CBF,26  data from an additional secondary assay or transactivation assay are missing; this does not permit application of any BS3 strength level. PS4 cannot be applied (2 alleles in gnomAD). VUS 
c.427G>T/p.E143X — — Truncating PVS1, PS4_supporting, PM2 PVS1, PS4_supporting, PM2  PATH 
c.454_456insA/p.K152fsX Ernst et al27  — Truncating PVS1, PS4_supporting, PM2 PVS1, PS4_supporting, PM2 Variant nomenclature does not conform with HGVS recommendations for sequence variants. We assume this variant is not present in gnomAD (PM2) and leads to NMD (PVS1). PATH 
c.496C>G/p.R166G Imai et al28  — LOF/dominant negative28  PS4_supporting, PM2, PM5, PP3 PS4_supporting, PM1, PM2, PM5, PP3 R166Q has been curated by the MM-VCEP as PATH. LPATH 
c.496C>T/p.R166X Preudhomme et al29  Bluteau et al30  Truncating PVS1, PS4, PM2, PP1 PVS1, PS4, PM2, PP1_strong  PATH 
9+10 c.610C>T/p.R204X Osato et al31  Song et al32  LOF31  PVS1, PS4, PM2, PP1 PVS1, PS4, PM2, PP1_strong  PATH 
11 c.619C>T/p.R207W You et al33  — — PS4_supporting, PM2, PP3 PS4_moderate, PM2, PP3 In silico prediction alone (ie, in this case pathogenic predictions by using SIFT, Polyphen, VEST, CHASM, and REVEL) is only supporting evidence and insufficient to classify a variant as PATH. VUS 
12 c.1243_1244insC/p.Q415fsX — — Elongated RUNX1 isoform PVS1_strong, PS4_supporting, PM2 PVS1_strong, PS4_supporting, PM2  LPATH 
ID*Variant cDNA/protein*Described in MDS/AML*Described in RUNX1 FPD*Functional impact on RUNX1*MM-VCEP ACMG/AMP criteria code*MM-VCEP RUNX1-specific criteriaFurther explanation of criteriaMM-VCEP classification
c.44_45delAG/p.Q15fsX — — Truncating PVS1_moderate, PS4_supporting, PM2 PS4_supporting, PM2 PVS1 cannot be used for early truncating variants only affecting RUNX1 isoform C. VUS 
c.179C>T/p.A60V Carnicer et al23  Lorente24  — BS1 BS1, BS3 This variant meets the calculated BS1 threshold (Latino subpopulation) and BS3 (normal transactivation and normal DNA binding/subcellular localization).25  The presence of the variant in patients with a RUNX1 phenotype is not sufficient to call a variant PATH, in particular not if the variant is present in gnomAD at a MAF incompatible with disease prevalence. BEN 
3+4 c.421T>G/p.S141A — RUNX1db Normal transactivation26  PS4_supporting, PP3, BS3_supporting PM1_supporting, PP3 Variant not present in RUNX1db. Although there is no effect on heterodimerization ability with CBF,26  data from an additional secondary assay or transactivation assay are missing; this does not permit application of any BS3 strength level. PS4 cannot be applied (2 alleles in gnomAD). VUS 
c.427G>T/p.E143X — — Truncating PVS1, PS4_supporting, PM2 PVS1, PS4_supporting, PM2  PATH 
c.454_456insA/p.K152fsX Ernst et al27  — Truncating PVS1, PS4_supporting, PM2 PVS1, PS4_supporting, PM2 Variant nomenclature does not conform with HGVS recommendations for sequence variants. We assume this variant is not present in gnomAD (PM2) and leads to NMD (PVS1). PATH 
c.496C>G/p.R166G Imai et al28  — LOF/dominant negative28  PS4_supporting, PM2, PM5, PP3 PS4_supporting, PM1, PM2, PM5, PP3 R166Q has been curated by the MM-VCEP as PATH. LPATH 
c.496C>T/p.R166X Preudhomme et al29  Bluteau et al30  Truncating PVS1, PS4, PM2, PP1 PVS1, PS4, PM2, PP1_strong  PATH 
9+10 c.610C>T/p.R204X Osato et al31  Song et al32  LOF31  PVS1, PS4, PM2, PP1 PVS1, PS4, PM2, PP1_strong  PATH 
11 c.619C>T/p.R207W You et al33  — — PS4_supporting, PM2, PP3 PS4_moderate, PM2, PP3 In silico prediction alone (ie, in this case pathogenic predictions by using SIFT, Polyphen, VEST, CHASM, and REVEL) is only supporting evidence and insufficient to classify a variant as PATH. VUS 
12 c.1243_1244insC/p.Q415fsX — — Elongated RUNX1 isoform PVS1_strong, PS4_supporting, PM2 PVS1_strong, PS4_supporting, PM2  LPATH 

All variants are annotated using RefSeq ID NM_001754.4. PS4 is applied assuming that the variants in the Simon et al1  study are germline variants. Germline status should be confirmed in DNA derived from cultured skin fibroblasts, cultured bone marrow mesenchymal stromal cells, or hair roots.

—, no data; BEN, benign; cDNA, complementary DNA; FPD, familial platelet disorder; gnomAD, Genome Aggregation Database; HGVS, Human Genome Variation Society; LOF, loss of function; LPATH, likely pathogenic; MAF, minor allele frequency; NMD, non-sense–mediated decay, PATH, pathogenic, VUS, variant of unknown significance.

*

From the Simon et al1  study.

MM-VCEP assessment.

Patients are related.

With regard to RUNX1 variant curation in the Simon et al1  study as an example, we would like to highlight the following points. (1) Three major RUNX1 isoforms (A, B, and C) are expressed by the use of 2 promoters and alternative splicing. Isoform function, biological relevance, and expression differ in hematopoietic tissue,10,11  which makes PVS1 not applicable for N-terminal truncating variants affecting only isoform C.7,12  (2) Different strength levels of pathogenic functional evidence (PS3) are based on decreased or enhanced transactivation activity, with or without a secondary assay showing decreased DNA binding affinity, diminished heterodimerization ability with CBFb, abnormal cellular localization, reduced colony-forming potential, or abnormal function of mutant RUNX1 in vivo. Of note, other functional assays, such as interaction of RUNX1 with MLL, are not valid secondary assays for RUNX1 function.13,14  (3) The presence of a RUNX1 germline variant in a proband with hereditary myelodysplastic syndrome (MDS)/acute leukemia, even with the typical phenotype including lifelong thrombocytopenia and platelet dysfunction, does not justify a pathogenic classification, but it is always dependent on a combination of additional functional, cosegregation, predictive, or population data. More information regarding the application of RUNX1-specific ACMG/Association for Molecular Pathology criteria in the classification of variants identified in this study is shown in Table 2.

As of 30 June 2020, 591 RUNX1 variants have been reported in the ClinVar database (https://www.ncbi.nlm.nih.gov/clinvar/). Many germline disease-causing RUNX1 variants are unique to individuals or families; thus, detailed annotation is not always available for reference when a new RUNX1 variant is identified.15  Only 21% of RUNX1 variants are clinically significant (pathogenic/likely pathogenic), whereas the majority (79%) are benign/likely benign or variants of uncertain significance, which are not clinically actionable (Figure 1). It is worth mentioning that 50% of RUNX1 variants are variants of uncertain significance that warrant more collaborative efforts for the scientific community to up- or downgrade them based on new evidence, such as observation in multiple probands, segregation with disease, or functional impact of the variant or absence in affected individuals, nonsegregation with disease, or no effects on protein function.

Figure 1.

RUNX1 variants in ClinVar and their clinical significance. All 591 RUNX1 variants deposited in ClinVar as of 30 June 2020 and their clinical significance based on the 5-tier system for germline variants (benign, likely benign, variant of uncertain significance, likely pathogenic, pathogenic) are shown in the pie chart. Only 21% of the RUNX1 variants are clinically actionable (ie, likely pathogenic and pathogenic).

Figure 1.

RUNX1 variants in ClinVar and their clinical significance. All 591 RUNX1 variants deposited in ClinVar as of 30 June 2020 and their clinical significance based on the 5-tier system for germline variants (benign, likely benign, variant of uncertain significance, likely pathogenic, pathogenic) are shown in the pie chart. Only 21% of the RUNX1 variants are clinically actionable (ie, likely pathogenic and pathogenic).

Close modal

Phenotypic criteria have been proposed by the ClinGen MM-VCEP, and they can be helpful in the determination of RUNX1 variant pathogenicity, because a high penetrance, with regard to thrombocytopenia and/or underlying platelet dysfunction, is typically recognized, and patients display ≥1 of the following features7 : mild to moderate thrombocytopenia with normal platelet size and volume in the absence of other causative factors; platelet ultrastructural and/or functional defects; and diagnosis of a hematologic malignancy, most commonly affecting the myeloid lineage (causing AML or MDS) and less frequently involving the lymphoid lineage and manifesting as T-cell acute lymphoblastic leukemia or others.

The following example highlights the importance of variant annotation for management decisions. A 56-year-old female with a diagnosis of MDS and a family history of hematologic malignancies was identified to have a germline RUNX1 c.167T>C (p.Leu56Ser) variant and was counseled that this RUNX1 variant was disease causing. Family members were tested for this variant to determine who lacked the variant and, thus, could be an appropriate stem cell transplant donor for the index patient and who in the family carries the variant and should receive surveillance on research protocols for RUNX1-associated familial platelet disorder with myeloid malignancy. Importantly, upon further review at the time of a second opinion, the RUNX1 c.167T>C (p.Leu56Ser) variant was reclassified to be a benign germline variant and a “red herring” in the evaluation of this family.

Patients with chronic otherwise unexplained thrombocytopenia, platelet ultrastructural and/or functional defects, and/or AML, MDS, or T-cell acute lymphoblastic leukemia should undergo genetic testing whenever there is a positive family history for a RUNX1 phenotype and when the patient has been diagnosed at a young age or a RUNX1 variant has been identified upon molecular testing of the leukemic clone. Germline material for testing should represent tissues that are not contaminated with blood/circulating blasts, such as cultured skin fibroblasts, which are the gold standard. Upon confirmation of a germline disease-causing RUNX1 variant, additional family members can be tested and followed-up long-term, including a baseline bone marrow biopsy with cytogenetic/molecular analysis and additional biopsies at the time of any significant/persistent change in blood counts. Most importantly, a family member with the RUNX1 variant should not be considered as a related stem cell donor, which makes recognition of the underlying germline syndrome paramount.16-18 

Our clinical example and the variant interpretation by Simon et al1  highlight how easily variants can be misclassified when criteria are not applied correctly, too much weight is put on the observation of the variant in affected probands, or the criteria are not combined correctly to reach the level of clinical significance (ie, disease causing). The accuracy of RUNX1 variant classification and interpretation is of great importance for treatment and follow-up of affected patients, related donor selection, and counseling of family members. Therefore, we emphasize that MM-VCEP RUNX1-specific rules, as the most accurate standards of germline RUNX1 variant classification, should be applied in clinical and research settings.7,8 

Data sharing requests should be sent to Simone Feurstein (feurstein@uchicago.edu).

This work was supported by the National Human Genome Research Institute National Institutes of Health (grants U41HG009649 and U41HG009650) and a 2018 National Institutes of Health/National Cancer Institute Leukemia SPORE DRP award (P50CA100632-16, project 00007529) (C.D.D.). The results provided in this publication were generated by the American Society of Hematology in collaboration with Baylor College of Medicine and the University of North Carolina, National Institutes of Health–funded ClinGen grant award recipients.

This commentary was written on behalf of the collaborative group of the American Society of Hematology - Clinical Genome Resource MM-VCEP, which is cochaired by Lucy A. Godley (University of Chicago) and David Wu (University of Washington). A complete list of MM-VCEP members is available at: https://clinicalgenome.org/affiliation/50034/#heading_membership.

Contribution: S.F. reviewed and classified the RUNX1 variants and explained the use of RUNX1-specific criteria; L.Z. worked on the nomenclature of variants; C.D.D. focused on the clinical impact of germline RUNX1 variants; and all authors wrote and edited the manuscript.

Conflict-of-interest disclosure: L.Z. has received honoraria from Future Technology Research LLC, BGI, and Illumina and honoraria and travel and accommodation expenses from Roche Diagnostics Asia Pacific; family members hold leadership positions and ownership interests in the Shanghai Genome Center. The remaining authors declare no competing financial interests.

Correspondence: Simone Feurstein, The University of Chicago, KCBD 7123A, 900 East 57th St, Chicago, IL 60637; e-mail: feurstein@uchicago.edu.

1.
Simon
L
,
Spinella
J-F
,
Yao
C-Y
, et al
.
High frequency of germline RUNX1 mutations in patients with RUNX1-mutated AML
.
Blood
.
2020
;
135
(
21
):
1882
-
1886
.
2.
DiNardo
CD
,
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
.
3.
DiNardo
CD
,
Bannon
SA
,
Routbort
M
, et al
.
Evaluation of patients and families with concern for predispositions to hematologic malignancies within the Hereditary Hematologic Malignancy Clinic (HHMC)
.
Clin Lymphoma Myeloma Leuk
.
2016
;
16
(
7
):
417
-
428.e2
.
4.
Yannakou
CK
,
Jones
K
,
Ryland
GL
, et al
.
Incidental detection of germline variants of potential clinical significance by massively parallel sequencing in haematological malignancies
.
J Clin Pathol
.
2018
;
71
(
1
):
84
-
87
.
5.
Leonard
DGB
, ed.
Molecular Pathology in Clinical Practice
. 2nd ed.
Cham, Switzerland
:
Springer International Publishing
;
2016
.
6.
Rivera-Muñoz
EA
,
Milko
LV
,
Harrison
SM
, et al
.
ClinGen Variant Curation Expert Panel experiences and standardized processes for disease and gene-level specification of the ACMG/AMP guidelines for sequence variant interpretation
.
Hum Mutat
.
2018
;
39
(
11
):
1614
-
1622
.
7.
Luo
X
,
Feurstein
S
,
Mohan
S
, et al
.
ClinGen Myeloid Malignancy Variant Curation Expert Panel recommendations for germline RUNX1 variants
.
Blood Adv
.
2019
;
3
(
20
):
2962
-
2979
.
8.
Wu
D
,
Luo
X
,
Feurstein
S
, et al;
collaborative group of the American Society of Hematology - Clinical Genome Resource Myeloid Malignancy Variant Curation Expert Panel
.
How I curate: applying American Society of Hematology-Clinical Genome Resource Myeloid Malignancy Variant Curation Expert Panel rules for RUNX1 variant curation for germline predisposition to myeloid malignancies
.
Haematologica
.
2020
;
105
(
4
):
870
-
887
.
9.
Tavtigian
SV
,
Greenblatt
MS
,
Harrison
SM
, et al;
ClinGen Sequence Variant Interpretation Working Group (ClinGen SVI)
.
Modeling the ACMG/AMP variant classification guidelines as a Bayesian classification framework
.
Genet Med
.
2018
;
20
(
9
):
1054
-
1060
.
10.
Brady
G
,
Elgueta Karstegl
C
,
Farrell
PJ
.
Novel function of the unique N-terminal region of RUNX1c in B cell growth regulation
.
Nucleic Acids Res
.
2013
;
41
(
3
):
1555
-
1568
.
11.
Tsuzuki
S
,
Seto
M
.
Expansion of functionally defined mouse hematopoietic stem and progenitor cells by a short isoform of RUNX1/AML1
.
Blood
.
2012
;
119
(
3
):
727
-
735
.
12.
Abou Tayoun
AN
,
Pesaran
T
,
DiStefano
MT
, et al;
ClinGen Sequence Variant Interpretation Working Group (ClinGen SVI)
.
Recommendations for interpreting the loss of function PVS1 ACMG/AMP variant criterion
.
Hum Mutat
.
2018
;
39
(
11
):
1517
-
1524
.
13.
Koh
CP
,
Wang
CQ
,
Ng
CEL
, et al
.
RUNX1 meets MLL: epigenetic regulation of hematopoiesis by two leukemia genes
.
Leukemia
.
2013
;
27
(
9
):
1793
-
1802
.
14.
Huang
G
,
Zhao
X
,
Wang
L
, et al
.
The ability of MLL to bind RUNX1 and methylate H3K4 at PU.1 regulatory regions is impaired by MDS/AML-associated RUNX1/AML1 mutations
.
Blood
.
2011
;
118
(
25
):
6544
-
6552
.
15.
Sood
R
,
Kamikubo
Y
,
Liu
P
.
Role of RUNX1 in hematological malignancies
.
Blood
.
2017
;
129
(
15
):
2070
-
2082
.
16.
Feurstein
S
,
Drazer
MW
,
Godley
LA
.
Genetic predisposition to leukemia and other hematologic malignancies
.
Semin Oncol
.
2016
;
43
(
5
):
598
-
608
.
17.
Churpek
JE
,
Godley
LA
;
University of Chicago Hematopoietic Malignancies Cancer Risk Team
.
How I diagnose and manage individuals at risk for inherited myeloid malignancies
.
Blood
.
2016
;
128
(
14
):
1800
-
1813
.
18.
Rafei
H
,
DiNardo
CD
.
Hereditary myeloid malignancies
.
Best Pract Res Clin Haematol
.
2019
;
32
(
2
):
163
-
176
.
19.
Plon
SE
,
Eccles
DM
,
Easton
D
, et al;
IARC Unclassified Genetic VariantsWorking Group
.
Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results
.
Hum Mutat
.
2008
;
29
(
11
):
1282
-
1291
.
20.
Richards
S
,
Aziz
N
,
Bale
S
, et al;
ACMG Laboratory Quality Assurance Committee
.
Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology
.
Genet Med
.
2015
;
17
(
5
):
405
-
424
.
21.
Harrison
SM
,
Rehm
LR
.
Is “likely pathogenic” really 90% likely? Reclassification data in ClinVar
.
Genome Med
.
2019
;
11
(
1
):
72
.
22.
National Institutes of Health
.
NCI dictionary of genetics terms.
https://www.cancer.gov/publications/dictionaries/genetics-dictionary. Accessed 18 October 2020.
23.
Carnicer
,
MJ
,
Nomdedéu
JF
,
Lasa
A
, et al
.
AML-1 mutations outside the RUNT domain: description of two cases in myeloid malignancies
.
Leukemia
.
2002
;
16
(
11
):
2329
-
2332
.
24.
Lorente
NP
. Molecular Characterization of Patients with Hereditary Myeloid Neoplasms by Next-Generation Sequencing [thesis]. Valencia, Spain: Universitat Politècnica de València;
2018
.
25.
Huang
G
,
Zhao
X
,
Wing
L
, et al
.
The ability of MLL to bind RUNX1 and methylate H3K4 at PU.1 regulatory regions is impaired by MDS/AML-associated RUNX1/AML1 mutations
.
Blood
.
2011
;
118
(
25
):
6544
-
6552
.
26.
Zhang
L
,
Li
Z
,
Yan
J
, et al
.
Mutagenesis of the Runt domain defines two energetic hot spots for heterodimerization with the core binding factor beta subunit
.
J Biol Chem
.
2003
;
278
(
35
):
33097
-
33104
.
27.
Ernst
T
,
Chase
A
,
Zoi
K, et al.
Transcription factor mutations in myelodysplastic/myeloproliferative neoplasms
.
Haematologica
.
2010
;
95
(
9
):
1473
-
1480
.
28.
Imai
Y
,
Kurokawa
M
,
Izutsu
K
, et al
.
Mutations of the AML1 gene in myelodysplastic syndrome and their functional implications in leukemogenesis
.
Blood
.
2000
;
96
(
9
):
3154
-
3160
.
29.
Preudhomme
C
,
Warot-Loze
D
,
Roumier
C
, et al
.
High incidence of biallelic point mutations in the Runt domain of the AML1/PEBP2 alpha B gene in Mo acute myeloid leukemia and in myeloid malignancies with acquired trisomy 21
.
Blood
.
2000
;
96
(
8
):
2862
-
2869
.
30.
Bluteau
O
,
Sebert
M
,
Leblanc
T
, et al
.
A landscape of germ line mutations in a cohort of inherited bone marrow failure patients
.
Blood
.
2018
;
96
(
8
):
2862
-
2869
.
31.
Osato
M
,
Asou
N
,
Abdalla
E
, et al
.
Biallelic and heterozygous point mutations in the runt domain of the AML1/PEBP2alphaB gene associated with myeloblastic leukemias
.
Blood
.
1999
;
93
(
6
):
1817
-
1824
.
32.
Song
WJ
,
Sullivan
MG
,
Legare
RD
, et al
.
Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia
.
Nat Genet
.
1999
;
23
(
2
):
166
-
175
.
33.
You
E
,
Cho
Y-U
,
Jang
S
, et al
.
Frequency and clinicopathologic features of RUNX1 mutations in patients with acute myeloid leukemia not otherwise specified
.
Am J Clin Pathol
.
2017
;
148
(
1
):
64
-
72
.

Author notes

*

S.F., L.Z., and C.D.D. contributed equally to this work.