Disease alleles that activate signal transduction are common in myeloid malignancies; however, there are additional unidentified mutations that contribute to myeloid transformation. Based on the recent identification of TET2 mutations, we evaluated the mutational status of TET1, TET2, and TET3 in myeloproliferative neoplasms (MPNs), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML). Sequencing of TET2 in 408 paired tumor/normal samples distinguished between 68 somatic mutations and 6 novel single nucleotide polymorphisms and identified TET2 mutations in MPN (27 of 354, 7.6%), CMML (29 of 69, 42%), AML (11 of 91, 12%), and M7 AML (1 of 28, 3.6%) samples. We did not identify somatic TET1 or TET3 mutations or TET2 promoter hypermethylation in MPNs. TET2 mutations did not cluster in genetically defined MPN, CMML, or AML subsets but were associated with decreased overall survival in AML (P = .029). These data indicate that TET2 mutations are observed in different myeloid malignancies and may be important in AML prognosis.

Our understanding of the molecular pathogenesis of myeloid malignancies, most notably acute myeloid leukemia (AML) and chronic myeloid leukemia (CML), has largely resulted from the identification and characterization of recurrent chromosomal translocations.1  However, in many patients with myeloproliferative neoplasms (MPNs) and chronic myelomonocytic leukemia (CMML), recurrent clonal cytogenetic abnormalities are not observed. More recently, DNA resequencing studies of candidate genes,2  gene families,3,4  and the cancer genome5  in MPN, CMML, and AML have identified somatic mutations in FLT3,6 JAK2,7-13 MPL,14,15  and the RAS family of oncogenes.16  These discoveries demonstrate activation of signal transduction pathways is a common pathogenic event in myeloid malignancies and have led to the development of molecularly targeted therapies. However, with the exception of CML, these therapies have yet to substantively improve outcomes for patients with myeloid malignancies.17,18  This may reflect insufficient target inhibition, or, alternatively, this may indicate incomplete dependence on these activated pathways resulting from the presence of additional somatic mutations with prognostic, therapeutic, and biologic relevance.

The role of TET (Ten-Eleven Translocation) family gene members in hematopoietic transformation was thought to be restricted to the involvement of TET1 as a translocation partner MLL-translocated AML, until the recent identification of inactivating mutations in TET2 in MPN and MDS patients.19  We therefore sought to evaluate a large set of MPN, CMML, and AML samples for somatic TET2 alterations. We sequenced all coding exons of TET2 in 408 paired tumor/normal samples and then assessed the frequency of somatic TET2 mutations in 606 patients with MPN, CMML, and AML. We also investigated whether deletion or epigenetic inactivation of TET2 are observed in MPN and evaluated MPN patients for somatic mutations in TET1 and TET3.

Patients

DNA was isolated from peripheral blood and/or bone marrow from 606 MPN, CMML, and AML samples. Matched normal DNA was available for 408 samples, including 354 sporadic MPN samples, 26 CMML samples, and 28 affected members of MPN pedigrees. Blood/bone marrow DNA but not matched normal DNA was available for 198 samples, including 3 sporadic MPN samples, 20 affected members of MPN kindreds, 96 AML samples, 45 CMML samples, and 34 M7 AML samples (16 from the Eastern Cooperative Oncology Group). Approval was obtained from the institutional review boards at the Dana-Farber Cancer Institute and at Memorial Sloan-Kettering Cancer Center for these studies, and informed consent was provided according to the Declaration of Helsinki.

Sequence analysis of TET1, TET2, and TET3

DNA resequencing of all coding exons of TET1-3 was performed (primers/conditions are listed in supplemental Table 1, available on the Blood website; see the Supplemental Materials link at the top of the online article). Nonsynonymous alterations not present in single nucleotide polymorphism (database [db]SNP) were annotated as somatic mutations or SNPs based on sequence analysis of matched germ line DNA. Nonsynonymous alterations not in dbSNP nor determined to be somatic in paired samples or in recently reported data19  were censored. All somatic mutations were validated by resequencing nonamplified DNA.

Copy number analysis of TET1, TET2, and TET3

A total of 207 MPN tumor samples were analyzed using Affymetrix 250K StyI Arrays.20  The JAK2V617F-mutant AML cell lines HEL and SET2 were analyzed using Affymetrix 6.0 SNP Arrays.

Methylation-specific polymerase chain reaction

Methylation of 2 CpG islands in the promoter region of TET2 was assessed in 37 MPN patients and 4 JAK2V617F-positive leukemia cell lines (SET2, MBO2, HEL, UKE1). Methylation-specific polymerase chain reaction was performed as previously described (primers are listed in supplemental Table 1).21 

Statistics

Statistical analyses were performed using MedCalc (MedCalc).

Sequence analysis of all coding exons of TET2 in 408 paired tumor/normal samples identified 8 frameshift, 12 nonsense, and 37 nonsynonymous alterations not present in dbSNP. Analysis of germ line DNA distinguished between 31 somatic missense mutations and 6 unannotated SNPs (Table 1; supplemental Figure 1); all unannotated SNPs were observed in matched normal tissue in at least 2 samples. All frameshift and nonsense mutations were not present in matched normal tissue. The strategy of paired sequencing of normal and tumor tissue is critical for accurate annotation of candidate mutations as 2 novel SNPs, which were recently reported as TET2 mutations (Q1084P and Y867H)22  were present in the germ line in multiple patient samples consistent with their being unannotated SNPs. After defining the spectrum of somatic TET2 mutations in paired tumor/normal samples, we determined the frequency of TET2 mutations in MPN (7.6%, including 9.8% polycythemia vera, 4.4% essential thrombocythemia, and 7.7% primary myelofibrosis), CMML (42.1%), AML (12.1%), and acute megakaryoblastic leukemia (3.6%). We identified biallelic/homozygous TET2 mutations in 1 of 354 MPN patients and in 7 of 69 CMML patients (P < .001, Fisher exact test). Sequencing of TET2 in 48 affected members from 28 MPN kindreds identified somatic TET2 mutations in 7 affected persons. We also identified 4 germ line nonsynonymous variants in affected members of MPD kindreds present in dbSNP that could represent rare familial MPN alleles. However, 3 of these 4 SNPs were observed in only some affected members of kindred but not others, and the fourth variant (M1701I) is observed in many sporadic MPN, CMML, and AML cases. Somatic TET2 mutations were not noted in the 4 JAK2V617F-positive leukemic cell lines.

Table 1

Novel TET2 somatic missense mutations and unannotated SNPs in 4q24

Alteration Nucleotide change Genomic coordinate Amino acid change
Somatic mutation  434 G→A  106374983  S145N 
Somatic mutation  935 A→G  106375484  N312S 
Somatic mutation  1379 C→T  106375928  S460F 
Somatic mutation  1997 A→G  106376546  D666G 
Somatic mutation  2821 C→T  106377370  P941S 
Somatic mutation  3403 G→A  106377953  C1135Y 
Somatic mutation  3575 T→G  106383519  C1194W 
Somatic mutation  3609 A→T  106384192  S1204C 
Somatic mutation  3639 C→T  106384222  R1214W 
Somatic mutation  3724 A→T  106384307  D1242V* 
Somatic mutation  3733 A→C  106384316  Y1245S 
Somatic mutation  3780 C→T  106384363  R1261C 
Somatic mutation  3781 G→A  106384364  R1261H* 
Somatic mutation  3862 G→T  106400285  G1289V 
Somatic mutation  4074 C→T  106410247  R1358C* 
Somatic mutation  4080 G→C  106410253  G1360R 
Somatic mutation  4248 G→T  106413237  V1417F 
Somatic mutation  5151 G→T  106416269  V1718L* 
Somatic mutation  5268 C→G  106416386  H1757D 
Somatic mutation  5283 A→T  106416401  Q1828L 
Somatic mutation  5430 T→C  106416548  C1811R 
Somatic mutation  5617 T→C  106416735  I1873T* 
Somatic mutation  5641 A→G  106416759  H1881R* 
Somatic mutation  5698 T→C  106416816  V1900A 
Somatic mutation  5754 C→T  106416873  A1919V 
Somatic mutation  5776 G→A  106416894  R1926H 
Somatic mutation  5780 G→A  106416898  R1927K 
Somatic mutation  5820 C→T  106416938  P1941S 
Somatic mutation  5896 G→A  106417014  R1966H 
Somatic mutation  5920 C→T  106417038  R1974M 
Somatic mutation  5998 G→A  106417116  R2000K 
SNP  100 C→T  106374649  L34F 
SNP  520 C→A  106375069  P174H 
SNP  2599 T→C  106377148  Y867H 
SNP  3418 A→T  106377767  E1073V 
SNP  3451 A→C  106377800  Q1084P 
SNP  5166 C→T  106416284  P1723S 
Alteration Nucleotide change Genomic coordinate Amino acid change
Somatic mutation  434 G→A  106374983  S145N 
Somatic mutation  935 A→G  106375484  N312S 
Somatic mutation  1379 C→T  106375928  S460F 
Somatic mutation  1997 A→G  106376546  D666G 
Somatic mutation  2821 C→T  106377370  P941S 
Somatic mutation  3403 G→A  106377953  C1135Y 
Somatic mutation  3575 T→G  106383519  C1194W 
Somatic mutation  3609 A→T  106384192  S1204C 
Somatic mutation  3639 C→T  106384222  R1214W 
Somatic mutation  3724 A→T  106384307  D1242V* 
Somatic mutation  3733 A→C  106384316  Y1245S 
Somatic mutation  3780 C→T  106384363  R1261C 
Somatic mutation  3781 G→A  106384364  R1261H* 
Somatic mutation  3862 G→T  106400285  G1289V 
Somatic mutation  4074 C→T  106410247  R1358C* 
Somatic mutation  4080 G→C  106410253  G1360R 
Somatic mutation  4248 G→T  106413237  V1417F 
Somatic mutation  5151 G→T  106416269  V1718L* 
Somatic mutation  5268 C→G  106416386  H1757D 
Somatic mutation  5283 A→T  106416401  Q1828L 
Somatic mutation  5430 T→C  106416548  C1811R 
Somatic mutation  5617 T→C  106416735  I1873T* 
Somatic mutation  5641 A→G  106416759  H1881R* 
Somatic mutation  5698 T→C  106416816  V1900A 
Somatic mutation  5754 C→T  106416873  A1919V 
Somatic mutation  5776 G→A  106416894  R1926H 
Somatic mutation  5780 G→A  106416898  R1927K 
Somatic mutation  5820 C→T  106416938  P1941S 
Somatic mutation  5896 G→A  106417014  R1966H 
Somatic mutation  5920 C→T  106417038  R1974M 
Somatic mutation  5998 G→A  106417116  R2000K 
SNP  100 C→T  106374649  L34F 
SNP  520 C→A  106375069  P174H 
SNP  2599 T→C  106377148  Y867H 
SNP  3418 A→T  106377767  E1073V 
SNP  3451 A→C  106377800  Q1084P 
SNP  5166 C→T  106416284  P1723S 

Novel unannotated SNPs in 4q24. SNPs were defined as missense mutations that were seen in more than one tumor and paired buccal sample.

*

Somatic missense mutations that occurred in more than 2 samples.

We did not identify methylation at either of 2 CpG islands of the TET2 promoter in 37 MPN samples or in 4 JAK2V617F-positive leukemic cell lines (supplemental Figure 2). Copy number analysis of 207 MPN patients identified 3 patients with heterozygous deletions of one copy of the region containing TET2 (4q24), suggesting that TET2 mutations are more common than large deletions in MPN patients. Sequencing data from these 3 patients revealed that one patient had a homozygous somatic missense mutation, consistent with heterozygous mutation followed by deletion of the remaining copy of TET2 (supplemental Figure 3). The HEL cell line had a heterozygous deletion of the TET2 locus. One MPN patient had a large deletion on chromosome 10, which included the TET1 locus (10q21.3). Furthermore, although we identified several novel SNPs in TET1 and TET3 (supplemental Table 3), we did not identify somatic TET1 or TET3 mutations in 96 MPN patients. No MPN samples or cell lines had loss of the TET3 locus (2p13.1) or amplifications of TET1, TET2, and TET3.

The frequency of TET2 mutations did not differ between JAK2V617F-positive (16.4%) and JAK2V617F-negative (2.5%) MPN (P = .08, Fisher exact test). Likewise, TET2 mutations were equally frequent in MPN patients with and without the recently described JAK2V617F-positive MPN predisposition haplotype (P = .9).20  We did not note a correlation between TET2 alterations and mutations in FLT3, JAK2, and RAS in CMML, nor did we observe a correlation between TET2 mutations and specific cytogenetic subgroups MLL, FLT3, CEBPA, or NPM1 mutations, or a history of antecedent MPN/MDS in AML (P > .5, Fisher exact test). However, we did note that TET2 mutations are associated with decreased overall survival in AML compared with TET2-wild-type AML patients (P = .03, Figure 1D; supplemental Table 2).

Figure 1

Exons of TET2 with locations of mutations and effect of mutations on overall survival in AML. Locations of mutations in MPN (A), CMML (B), and AML/acute megakaryoblastic leukemia samples (C) as well Kaplan-Meier estimates of overall survival in AML (D) are shown. Shaded regions represent non–protein-coding exons and introns are not shown. Exons are drawn to relative scale. Missense mutations (down arrowheads), nonsense mutations (up arrowheads), and frameshifts (diamonds) are shown at their approximate location along the exon. Mutations occurring within the same patient sample are represented in the same color. Mutations that were homozygous are highlighted in yellow.

Figure 1

Exons of TET2 with locations of mutations and effect of mutations on overall survival in AML. Locations of mutations in MPN (A), CMML (B), and AML/acute megakaryoblastic leukemia samples (C) as well Kaplan-Meier estimates of overall survival in AML (D) are shown. Shaded regions represent non–protein-coding exons and introns are not shown. Exons are drawn to relative scale. Missense mutations (down arrowheads), nonsense mutations (up arrowheads), and frameshifts (diamonds) are shown at their approximate location along the exon. Mutations occurring within the same patient sample are represented in the same color. Mutations that were homozygous are highlighted in yellow.

Close modal

In this report, we sequenced all coding exons of TET2 to define the spectrum of somatic TET2 mutations in myeloid malignancies. The broad range of myeloid disorders linked to mutations in TET2 suggests that mutations in TET2 have a pleiotropic role in myeloid transformation. Although our data suggest that TET2 mutations may hold prognostic significance in AML, larger clinical correlative studies will be needed to more accurately assess the effect of TET2 mutations on prognosis, diagnosis, and therapeutic relevance to myeloid malignancies. Whether TET2 mutations dysregulate pathways already known to contribute to hematopoietic transformation, or represent a novel pathway, remains to be elucidated, and the role of TET family alterations in neoplasms other than myeloid malignancies is not yet known.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the patients who have contributed to our understanding of these disorders.

This work was supported by the National Institutes of Health (grant CA10863101, G.G.-M., H.M.K., and D.G.G.; HL082677, R.L.L.), the Howard Hughes Medical Institute, the Starr Cancer Consortium (New York, NY), the Myeloproliferative Disorders Foundation (Chicago, IL), the Doris Duke Charitable Foundation (New York, NY), and the ECOG Leukemia Tissue Bank (Boston, MA; CA021115 and CA114737). O.A.-W. is supported by a Dana Fellowship from the Memorial Sloan Kettering Clinical Scholars Program. O.K. is supported by a grant from the Academy of Finland (Helsinki, Finland). D.G.G. is an Investigator of the Howard Hughes Medical Institute and is a Doris Duke Charitable Foundation Distinguished Clinical Scientist. R.L.L. is an Early Career Award recipient of the Howard Hughes Medical Institute, a Clinical Scientist Development Award recipient of the Doris Duke Charitable Foundation, and the Geoffrey Beene Junior Chair at Memorial Sloan Kettering Cancer Center. J.D.C. is a Scholar of the Leukemia & Lymphoma Society (White Plains, NY).

National Institutes of Health

Contribution: O.A.-W., A.M., J.P., D.G.G., J.D.C., and R.L.L. designed the study; C.H., G.G.-M., M.W., S.M., J.Y., R.B., E.P., M.M.L.B., M.B., M.S.T., H.M.K., and R.L.L. collected and processed samples and provided genetic and clinical annotation; O.A.-W., J.P., K.H., S.T., I.D., A.H., and R.L.L. performed sequence analysis, analyzed sequence traces, and validated mutations; O.A.-W., B.L.E., R.M.S., and R.L.L. acquired and analyzed SNP array data; O.A.-W., J.P., and O.K. performed methylation-specific polymerase chain reaction; O.A.-W. and R.L.L. wrote the manuscript with assistance from A.M., C.H., and J.D.C.

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

Correspondence: Ross L. Levine, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 20, New York, NY 10065; e-mail: leviner@mskcc.org.

1
Fröhling
 
S
Scholl
 
C
Gilliland
 
DG
Levine
 
RL
Genetics of myeloid malignancies: pathogenetic and clinical implications.
J Clin Oncol
2005
, vol. 
23
 (pg. 
6285
-
6295
)
2
Ley
 
TJ
Minx
 
PJ
Walter
 
MJ
, et al. 
A pilot study of high-throughput, sequence-based mutational profiling of primary human acute myeloid leukemia cell genomes.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
14275
-
14280
)
3
Loriaux
 
MM
Levine
 
RL
Tyner
 
JW
, et al. 
High-throughput sequence analysis of the tyrosine kinome in acute myeloid leukemia.
Blood
2008
, vol. 
111
 (pg. 
4788
-
4796
)
4
Tomasson
 
MH
Xiang
 
Z
Walgren
 
R
, et al. 
Somatic mutations and germline sequence variants in the expressed tyrosine kinase genes of patients with de novo acute myeloid leukemia.
Blood
2008
, vol. 
111
 (pg. 
4797
-
4808
)
5
Ley
 
TJ
Mardis
 
ER
Ding
 
L
, et al. 
DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome.
Nature
2008
, vol. 
456
 (pg. 
66
-
72
)
6
Kiyoi
 
H
Naoe
 
T
Yokota
 
S
, et al. 
Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia: Leukemia Study Group of the Ministry of Health and Welfare (Kohseisho).
Leukemia
1997
, vol. 
11
 (pg. 
1447
-
1452
)
7
James
 
C
Ugo
 
V
Le Couedic
 
JP
, et al. 
A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera.
Nature
2005
, vol. 
434
 (pg. 
1144
-
1148
)
8
Baxter
 
EJ
Scott
 
LM
Campbell
 
PJ
, et al. 
Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders.
Lancet
2005
, vol. 
365
 (pg. 
1054
-
1061
)
9
Kralovics
 
R
Passamonti
 
F
Buser
 
AS
, et al. 
A gain-of-function mutation of JAK2 in myeloproliferative disorders.
N Engl J Med
2005
, vol. 
352
 (pg. 
1779
-
1790
)
10
Zhao
 
R
Xing
 
S
Li
 
Z
, et al. 
Identification of an acquired JAK2 mutation in polycythemia vera.
J Biol Chem
2005
, vol. 
280
 (pg. 
22788
-
22792
)
11
Levine
 
RL
Loriaux
 
M
Huntly
 
BJ
, et al. 
The JAK2V617F activating mutation occurs in chronic myelomonocytic leukemia and acute myeloid leukemia, but not in acute lymphoblastic leukemia or chronic lymphocytic leukemia.
Blood
2005
, vol. 
106
 (pg. 
3377
-
3379
)
12
Levine
 
RL
Wadleigh
 
M
Cools
 
J
, et al. 
Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis.
Cancer Cell
2005
, vol. 
7
 (pg. 
387
-
397
)
13
Scott
 
LM
Scott
 
MA
Campbell
 
PJ
Green
 
AR
Progenitors homozygous for the V617F mutation occur in most patients with polycythemia vera, but not essential thrombocythemia.
Blood
2006
, vol. 
108
 (pg. 
2435
-
2437
)
14
Pardanani
 
AD
Levine
 
RL
Lasho
 
T
, et al. 
MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients.
Blood
2006
, vol. 
108
 (pg. 
3472
-
3476
)
15
Pikman
 
Y
Lee
 
BH
Mercher
 
T
, et al. 
MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia.
PLoS Med
2006
, vol. 
3
 pg. 
e270
 
16
Murray
 
MJ
Cunningham
 
JM
Parada
 
LF
Dautry
 
F
Lebowitz
 
P
Weinberg
 
RA
The HL-60 transforming sequence: a ras oncogene coexisting with altered myc genes in hematopoietic tumors.
Cell
1983
, vol. 
33
 (pg. 
749
-
757
)
17
Verstovek
 
S
Kantarjian
 
H
Pardanani
 
A
, et al. 
A phase I/II study of ICNB018424, an oral, selective JAK inhibitor, in patients with primary myelofibrosis and postPV/ET myelofibrosis.
Am Soc Clin Oncol Annual Meeting
2008
 
abstract 7004
18
Stone
 
RM
DeAngelo
 
DJ
Klimek
 
V
, et al. 
Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412.
Blood
2005
, vol. 
105
 (pg. 
54
-
60
)
19
Delhommeau
 
F
Dupont
 
S
James
 
C
, et al. 
TET2 is a novel tumor suppressor gene inactivated in myeloproliferative neoplasms: identification of a pre-JAK2 V617F event [abstract].
Blood
2008
, vol. 
112
 (pg. 
1
-
3
)
20
Kilpivaara
 
O
Mukherjee
 
S
Schram
 
AM
, et al. 
Genome-wide analysis of myeloproliferative neoplasms (MPN) identifies a germline JAK2 SNP that is associated with predisposition to the development of JAK2V617F-positive MPN and modifier loci that influence MPN phenotype.
Nat Genet
2009
, vol. 
41
 (pg. 
455
-
459
)
21
Herman
 
JG
Graff
 
JR
Myohanen
 
S
Nelkin
 
BD
Baylin
 
SB
Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 (pg. 
9821
-
9826
)
22
Jankowska
 
AM
Szpurka
 
H
Tiu
 
RV
, et al. 
Loss of heterozygosity 4q24 and TET2 mutations associated with myelodysplastic/myeloproliferative neoplasms.
Blood
2009
4
16
 
Epub ahead of print
Sign in via your Institution