• Exome sequencing of adult ETP-ALL reveals new recurrent mutations; in particular, DNMT3A is frequently mutated in adult ETP-ALL.

  • More than 60% of all adult patients with ETP-ALL harbor a mutation that could potentially be targeted by a specific therapy.

Early T-cell precursor (ETP) acute lymphoblastic leukemia (ALL) is a high-risk subgroup of T-lineage ALL characterized by specific stem cell and myeloid features. In adult ETP-ALL, no comprehensive studies on the genetic background have been performed to elucidate molecular lesions of this distinct subgroup. We performed whole-exome sequencing of 5 paired ETP-ALL samples. In addition to mutations in genes known to be involved in leukemogenesis (ETV6, NOTCH1, JAK1, and NF1), we identified novel recurrent mutations in FAT1 (25%), FAT3 (20%), DNM2 (35%), and genes associated with epigenetic regulation (MLL2, BMI1, and DNMT3A). Importantly, we verified the high rate of DNMT3A mutations (16%) in a larger cohort of adult patients with ETP-ALL (10/68). Mutations in epigenetic regulators support clinical trials, including epigenetic-orientated therapies, for this high-risk subgroup. Interestingly, more than 60% of adult patients with ETP-ALL harbor at least a single genetic lesion in DNMT3A, FLT3, or NOTCH1 that may allow use of targeted therapies.

T-lineage acute lymphoblastic leukemia (T-ALL) accounts for approximately 25% of all adult ALL. The novel high-risk subgroup of early T-cell precursor (ETP) represents 10% of all adult T-ALL. This subgroup is characterized by a specific immunophenotype (CD1-, CD5weak, CD8-, or coexpression of stem cell and/or myeloid markers) and distinct molecular features (FLT3 mutations and an absence of NOTCH1 mutations).1,2  ETP-ALL, regarded as stem cell–like leukemia, is associated with a high risk for treatment failure for both pediatric and adult patients. Because of the myeloid features of ETP-ALL, it was proposed that these patients might benefit from therapies used in myeloid malignancies, including high-dose cytarabine or epigenetic modulators.3 

Although in pediatric ETP-ALL whole-genome sequencing has revealed a highly heterogeneous picture of genetic alterations,4  no comprehensive molecular analyses have been performed in adult ETP-ALL. Here we systematically screened for somatic mutations in adult ETP-ALL. We have analyzed exomes of 5 patients and subsequently verified mutations in DNMT3A and members of the polycomb repressor complex 2 (PRC2) in a larger cohort of adult patients with ETP-ALL.

Patients

We investigated paired bone marrow samples of 5 adult patients with ETP-ALL (supplementary Methods and supplementary Table 1, available on the Blood Web site) with sufficient genomic DNA at diagnosis and in remission (minimal residual disease5  level < 4 × 10−3) that had been sent to the reference laboratory within the German Acute Lymphoblastic Leukemia Multicenter (GMALL) study group. In addition, 68 adult patients with ETP-ALL (55 men, 13 women; median age, 38 years) were analyzed for the mutation status of selected genes (DNMT3A, EZH2, EP300, SH2B3, and SUZ12). For 52 (76%) of the 68 patients, clinical follow-up data were available (supplementary Tables 2 and 3). All patients gave written informed consent to participate in the study according to the Declaration of Helsinki. The studies were approved by the ethics board of the Johann Wolfgang Goethe-Universität of Frankfurt/Main in Germany.

Exome sequencing and mutation analysis

The methods are described in the supplemental Data. In brief, exome sequencing was performed for 5 adult patients with ETP-ALL, as previously reported,6,7  using exon capturing from genomic DNA (Sure Select target enrichment; Agilent, Santa Clara, CA), followed by 76-bp paired-end sequencing on an Illumina Genome Analyzer IIx platform (Illumina, San Diego, CA). Candidate variants were confirmed by capillary sequencing of genomic DNA.

Mutation analyses for EZH2 (exons 14-20), SH2B3 (exons 2-8), DNMT3A (exons 13-23), EP300 (exon 30), and SUZ12 (exons 14-15) were performed by Sanger sequencing in 68 adult patients with ETP-ALL. Primers are listed in the supplement (supplementary Table 4). All samples had been previously analyzed for FLT3 and NOTCH1 mutations.2 

Statistical analyses were performed using SPSS software version 17 (SPSS Inc, Chicago, IL) and GraphPad Prism software version 5 (GraphPad Software Inc, La Jolla, CA).

As no comprehensive analysis exists on the molecular background of the high-risk subgroup of adult ETP-ALL, we performed whole-exome sequencing in 5 adult patients (supplementary Table 1). At least 3.8 Gbp of exome sequence was generated for each sample (supplementary Table 5). We found a total of 63 nonsynonymous somatic mutations or indels in 5 patients with ETP-ALL (range, 6-16 per patient). Eleven mutations/indels affected cancer genes, according to the census of human cancer genes8  (see also http://www.sanger.ac.uk/genetics/CGP/Census). Recurrent mutations affected DNMT3A and FAT3 (FAT, tumor suppressor homolog 3) in 2 of the 5 patients. The DNA-methyl-transferase DNMT3A is a frequent mutational target in acute myeloid leukemia (AML; 20%),9  whereas FAT3 mutations were reported in ovarian carcinoma.10  New mutational targets identified involved genes in epigenetic regulation (MLL2, BMI1, and FAT1) and genes reported to be mutated in pediatric ETP-ALL and other ALL subgroups (JAK1, ETV6, NOTCH1, DNM2, and NF1)4,11,12  (supplementary Table 6).

Deregulated DNA methylation is a commonly found in many malignancies.13 DNMT3A is mutated in myelodysplastic syndrome (8%) and AML,9,14  where it is associated with a poor prognosis.9,15-18  As the same mutations occur in myelodysplastic syndrome and the corresponding secondary AML,19 DNMT3A mutations likely represent an early event in leukemogenesis.20  Until now, a single study has reported a single DNMT3A mutation in T-ALL, and just recently, a second paper described frequent mutations of DNMT3A in adult T-ALL, although not specifying ETP-ALL.21,22  Interestingly, no DNMT3A mutations were identified by whole-genome sequencing in pediatric ETP-ALL.4 

By Sanger sequencing, we determined the frequency of DNMT3A mutations in a large cohort of adult ETP-ALL. In 16% of the 68 patients with ETP-ALL, DNMT3A was mutated, reflecting a mutation rate similar to that of AML, with a similar mutation pattern and mutational hot spot in exon 23. Amino acid R882 of DNMT3A was mutated in 5 ETP-ALL samples. Four of the 6 remaining mutations were previously described in AML (supplementary Figure 1).9,15,23 

With respect to clinical characteristics, no difference was observed between DNMT3A mutated and wild-type patients regarding sex distribution. In addition, DNMT3A mutated and wild-type patients did not differ in the immunophenotype or the mutational status of NOTCH1 and FLT3 (Table 1 and supplementary Table 3). Thus, in contrast to AML, no association between DNMT3A and FLT3 mutations can be found.9,15,23  Similar to in AML,9,16  patients with ETP-ALL with DNMT3A mutations were significantly older (median, 63 vs 37 years; P = .016) than patients with DNMT3A wild-type (Table 1). The lack of DNMT3A mutations in pediatric ETP-ALL and the low rate of DNMT3A mutations in pediatric AML24,25  point toward age-specific genetic differences. Because of the occurrence of genetic alterations in epigenetic regulators, we verified genetic mutations, including members of PRC2 in a larger ETP-ALL cohort (n = 68). Mutations were found for EZH2 in 4 of 68 (6%) and SUZ12 in 1 of 68 (1%) of adult ETP-ALL (supplementary Figure 2). No mutation was found in EP300. Thus, in contrast to pediatric ETP-ALL, with a high rate of mutations in PRC2,4,21  we found a low rate of PRC mutations in adult ETP-ALL. DNMT3A mutations had no significant prognostic effect in this small cohort of adult ETP-ALL. Patients with at least a single mutation in epigenetic regulators (DNMT3A, SUZ12, EP300, or EZH2) showed a trend toward inferior survival compared with unaffected patients (1-year survival, 56% vs 88%; log rank, P value = .24; Breslow, P value = .16; supplementary Figure 3).

Table 1

Characteristics of adult patients with ETP-ALL and comparison between DNMT3A mutated and DNMT3A wild-type patients

CharacteristicDNMT3mut (n = 11)DNMT3wt (n = 57)P
n%n%
Sex     .35 
 Female 27.3 10 17.6  
 Male 72.7 47 82.4  
Age, y     .02 
 Median 63 37  
 Range 24-73 17-74  
Clinical data available     .27 
 n 64 45 79  
FLT3     .41 
 Mutated 27.3 21 36.8  
 Wild-type 72.7 36 63.2  
NOTCH1     .68 
 Mutated 9.1 15.8  
 Wild-type 10 90.9 48 84.2  
EZH2     .52 
 Mutated 9.1 5.3  
 Wild-type 10 90.9 54 94.7  
SH2B3     .49 
 Mutated  
 Wild-type 11 100 53 93  
SUZ12     .84 
 Mutated 1.8  
 Wild-type 11 100 56 98.2  
CharacteristicDNMT3mut (n = 11)DNMT3wt (n = 57)P
n%n%
Sex     .35 
 Female 27.3 10 17.6  
 Male 72.7 47 82.4  
Age, y     .02 
 Median 63 37  
 Range 24-73 17-74  
Clinical data available     .27 
 n 64 45 79  
FLT3     .41 
 Mutated 27.3 21 36.8  
 Wild-type 72.7 36 63.2  
NOTCH1     .68 
 Mutated 9.1 15.8  
 Wild-type 10 90.9 48 84.2  
EZH2     .52 
 Mutated 9.1 5.3  
 Wild-type 10 90.9 54 94.7  
SH2B3     .49 
 Mutated  
 Wild-type 11 100 53 93  
SUZ12     .84 
 Mutated 1.8  
 Wild-type 11 100 56 98.2  

To further explore these genetic alterations as recurrent events, additional patients with ETP-ALL (n = 20) were analyzed by targeted next-generation sequencing (supplemental Data). In addition to DNMT3A, mutations in MLL2 (10%), previously linked to lymphoma,26  underscore the relevance of alterations in epigenetic regulators. Moreover, mutations in the cadherins FAT1 (25%) and FAT3 (20%) implicate alterations in cell adhesion, interaction mechanisms, and activation of the Wnt pathway.27  In particular, FAT1 may act as a tumor suppressor.27  Finally mutations DNM2 (35%) and JAK1 (15%)4  point to molecular targets for novel therapies (supplementary Figure 4, supplementary Figure 5, and supplementary Table 7).

In conclusion, adult ETP-ALL displays a distinct mutation spectrum, particularly affecting genes involved in epigenetic regulation (in particular, MLL2, DNMT3A, and PRC2). This mutation spectrum is different for pediatric patients, with a lower rate of PRC2 mutations and a higher rate of DNMT3A mutations pointing toward distinct molecular alterations in adult ETP-ALL. This might result from preexisting lesions in the hematopoietic progenitors, as it was shown that TET2 mutations result in an age-related myeloid lineage bias in elderly individuals.28 DNMT3A mutations in adult ETP-ALL show a similar frequency to that seen in AML, are located at the same hot spot, and are in conjunction with mutations in other epigenetic regulators of potential unfavorable prognostic effect, underscoring the stem cell/myeloid origin of ETP-ALL. These data may provide a potential rationale to study epigenetic directed therapies in the high-risk subgroup of ETP-ALL. Importantly, we can now define at least 3 different molecular subgroups of adult ETP-ALL characterized by alterations in epigenetic regulators (25%), mutations in FLT3 (35%) and other kinases (JAK pathway), and the presence of NOTCH1 mutations (15%) (Figure 1).2  In total, more than 60% of all adult patients with ETP-ALL harbor alterations that can potentially be molecularly targeted by new treatment approaches such as demethylating agents, kinase inhibitors, and γ secretase inhibitors.

Figure 1

The spectrum of mutations in DNMT3A, FLT3, NOTCH1, EZH2, SH2B3, and SUZ12 in 68 adult patients with ETP-ALL.

Figure 1

The spectrum of mutations in DNMT3A, FLT3, NOTCH1, EZH2, SH2B3, and SUZ12 in 68 adult patients with ETP-ALL.

Close modal

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 Liliana H. Mochmann for critical reading of the manuscript.

This work was supported by grants from the Deutsche Krebshilfe (Mildred Scheel Professur) and Gutermuth-Stiftung (C.D.B.) and the Deutsche Krebshilfe (70-2657-Ho2 [D.H.], BMBF 01GI 9971 [D.H. and N.G.], and 109031 [P.A.G. and S.K.B.]).

Contribution: M.N. contributed to the research, analyzed data, and wrote the manuscript; S.S. supervised the reference laboratory and reviewed the manuscript; N.G. supervised the GMALL study center and reviewed the manuscript; D.H. supervised the GMALL study group; S.H., C.S., N.P.K., B.K., S.V., A.G., S.K., H.B., T.R., M.B., J.H., and S.K.B. contributed to the research; W.-K.H. contributed to the study analysis and reviewed the manuscript; P.A.G. designed the research, contributed to the data analysis, and edited the manuscript; C.D.B. designed the research, contributed to the data analysis, and edited the manuscript; and P.A.G. and C.D.B. jointly supervised the study.

Conflict-of-interest disclosure: P.A.G. and S.K. received honoraria from Illumina. The remaining authors declare no competing financial interests.

Correspondence: Claudia D. Baldus, Charité, University Hospital Berlin, Campus Benjamin Franklin, Department of Hematology and Oncology, Hindenburgdamm 30, 12203 Berlin, Germany; e-mail: claudia.baldus@charite.de.

1
Coustan-Smith
 
E
Mullighan
 
CG
Onciu
 
M
, et al. 
Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia.
Lancet Oncol
2009
, vol. 
10
 
2
(pg. 
147
-
156
)
2
Neumann
 
M
Heesch
 
S
Gökbuget
 
N
, et al. 
Clinical and molecular characterization of early T-cell precursor leukemia: a high-risk subgroup in adult T-ALL with a high frequency of FLT3 mutations.
Blood Cancer J
2012
, vol. 
2
 
1
pg. 
e55
 
3
Pui
 
CH
Mullighan
 
CG
Evans
 
WE
Relling
 
MV
Pediatric acute lymphoblastic leukemia: where are we going and how do we get there?
Blood
2012
, vol. 
120
 
6
(pg. 
1165
-
1174
)
4
Zhang
 
J
Ding
 
L
Holmfeldt
 
L
, et al. 
The genetic basis of early T-cell precursor acute lymphoblastic leukaemia.
Nature
2012
, vol. 
481
 
7380
(pg. 
157
-
163
)
5
Gökbuget
 
N
Kneba
 
M
Raff
 
T
, et al. 
German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia
Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies.
Blood
2012
, vol. 
120
 
9
(pg. 
1868
-
1876
)
6
Greif
 
PA
Yaghmaie
 
M
Konstandin
 
NP
, et al. 
Somatic mutations in acute promyelocytic leukemia (APL) identified by exome sequencing.
Leukemia
2011
, vol. 
25
 
9
(pg. 
1519
-
1522
)
7
Greif
 
PA
Dufour
 
A
Konstandin
 
NP
, et al. 
GATA2 zinc finger 1 mutations associated with biallelic CEBPA mutations define a unique genetic entity of acute myeloid leukemia.
Blood
2012
, vol. 
120
 
2
(pg. 
395
-
403
)
8
Futreal
 
PA
Coin
 
L
Marshall
 
M
, et al. 
A census of human cancer genes.
Nat Rev Cancer
2004
, vol. 
4
 
3
(pg. 
177
-
183
)
9
Ley
 
TJ
Ding
 
L
Walter
 
MJ
, et al. 
DNMT3A mutations in acute myeloid leukemia.
N Engl J Med
2010
, vol. 
363
 
25
(pg. 
2424
-
2433
)
10
Cancer Genome Atlas Research Network
Integrated genomic analyses of ovarian carcinoma.
Nature
2011
, vol. 
474
 
7353
(pg. 
609
-
615
)
11
Biagi
 
C
Astolfi
 
A
Masetti
 
R
, et al. 
Pediatric early T-cell precursor leukemia with NF1 deletion and high-sensitivity in vitro to tipifarnib.
Leukemia
2010
, vol. 
24
 
6
(pg. 
1230
-
1233
)
12
De Keersmaecker
 
K
Atak
 
ZK
Li
 
N
, et al. 
Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia.
Nat Genet
2013
, vol. 
45
 
2
(pg. 
186
-
190
)
13
Sandoval
 
J
Esteller
 
M
Cancer epigenomics: beyond genomics.
Curr Opin Genet Dev
2012
, vol. 
22
 
1
(pg. 
50
-
55
)
14
Walter
 
MJ
Ding
 
L
Shen
 
D
, et al. 
Recurrent DNMT3A mutations in patients with myelodysplastic syndromes.
Leukemia
2011
, vol. 
25
 
7
(pg. 
1153
-
1158
)
15
Ribeiro
 
AF
Pratcorona
 
M
Erpelinck-Verschueren
 
C
, et al. 
Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia.
Blood
2012
, vol. 
119
 
24
(pg. 
5824
-
5831
)
16
Thol
 
F
Damm
 
F
Lüdeking
 
A
, et al. 
Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
21
(pg. 
2889
-
2896
)
17
Marcucci
 
G
Metzeler
 
KH
Schwind
 
S
, et al. 
Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia.
J Clin Oncol
2012
, vol. 
30
 
7
(pg. 
742
-
750
)
18
Shen
 
Y
Zhu
 
YM
Fan
 
X
, et al. 
Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia.
Blood
2011
, vol. 
118
 
20
(pg. 
5593
-
5603
)
19
Fried
 
I
Bodner
 
C
Pichler
 
MM
, et al. 
Frequency, onset and clinical impact of somatic DNMT3A mutations in therapy-related and secondary acute myeloid leukemia.
Haematologica
2012
, vol. 
97
 
2
(pg. 
246
-
250
)
20
Shih
 
AH
Abdel-Wahab
 
O
Patel
 
JP
Levine
 
RL
The role of mutations in epigenetic regulators in myeloid malignancies.
Nat Rev Cancer
2012
, vol. 
12
 
9
(pg. 
599
-
612
)
21
Simon
 
C
Chagraoui
 
J
Krosl
 
J
, et al. 
A key role for EZH2 and associated genes in mouse and human adult T-cell acute leukemia.
Genes Dev
2012
, vol. 
26
 
7
(pg. 
651
-
656
)
22
Grossmann
 
V
Haferlach
 
C
Weissmann
 
S
, et al. 
The molecular profile of adult T-cell acute lymphoblastic leukemia: mutations in RUNX1 and DNMT3A are associated with poor prognosis in T-ALL.
Genes Chromosomes Cancer
2013
, vol. 
52
 
4
(pg. 
410
-
422
)
23
Renneville
 
A
Boissel
 
N
Nibourel
 
O
, et al. 
Prognostic significance of DNA methyltransferase 3A mutations in cytogenetically normal acute myeloid leukemia: a study by the Acute Leukemia French Association.
Leukemia
2012
, vol. 
26
 
6
(pg. 
1247
-
1254
)
24
Thol
 
F
Heuser
 
M
Damm
 
F
Klusmann
 
JH
Reinhardt
 
K
Reinhardt
 
D
DNMT3A mutations are rare in childhood acute myeloid leukemia.
Haematologica
2011
, vol. 
96
 
8
(pg. 
1238
-
1240
)
25
Liang
 
DC
Liu
 
HC
Yang
 
CP
, et al. 
Cooperating gene mutations in childhood acute myeloid leukemia with special reference on mutations of ASXL1, TET2, IDH1, IDH2, and DNMT3A.
Blood
2013
, vol. 
121
 
15
(pg. 
2988
-
2995
)
26
Morin
 
RD
Mendez-Lago
 
M
Mungall
 
AJ
, et al. 
Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma.
Nature
2011
, vol. 
476
 
7360
(pg. 
298
-
303
)
27
Morris
 
LG
Kaufman
 
AM
Gong
 
Y
, et al. 
Recurrent somatic mutation of FAT1 in multiple human cancers leads to aberrant Wnt activation.
Nat Genet
2013
, vol. 
45
 
3
(pg. 
253
-
261
)
28
Busque
 
L
Patel
 
JP
Figueroa
 
ME
, et al. 
Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis.
Nat Genet
2012
, vol. 
44
 
11
(pg. 
1179
-
1181
)

Author notes

P.A.G. and C.D.B. contributed equally to this study.

Sign in via your Institution