Inactivating mutations of the Ten-Eleven Translocation 2 (TET2) gene were first identified in myeloid malignancies and more recently in peripheral T-cell lymphomas (PTCLs). In the present study, we investigated the presence of TET2 coding sequence mutations and their clinical relevance in a large cohort of 190 PTCL patients. TET2 mutations were identified in 40 of 86 (47%) cases of angioimmunoblastic T-cell lymphoma (AITL) and in 22 of 58 (38%) cases of peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), but were absent in all other PTCL entities, with the exception of 2 of 10 cases of enteropathy-associated T-cell lymphoma. Among PTCL-NOS, a heterogeneous group of lymphoma-comprising cases likely to derive from Th follicular (TFH) cells similarly to AITL, TET2 mutations were more frequent when PTCL-NOS expressed TFH markers and/or had features reminiscent of AITL (58% vs 24%, P = .01). In the AITL and PTCL-NOS subgroups, TET2 mutations were associated with advanced-stage disease, thrombocytopenia, high International Prognostic Index scores, and a shorter progression-free survival.

Inactivating mutations of the Ten-Eleven Translocation 2 (TET2) gene were first identified in myeloid malignancies.1–2 TET2 encodes a 2 oxoglutarate/Fe2+–dependent oxygenase that catalyses the oxidation of 5-methylcytosine to 5-hydroxymethylcytosine,3–5  and its inactivation in mice results in expansion of the hematopoietic progenitor cells and in pleiotropic abnormalities affecting both myeloid and lymphoid lineages.6–8  Recently, TET2 mutations have been reported in human lymphomas with a higher frequency in T cell– than B cell–derived neoplasms (12% vs 2%).7,9  Among peripheral T-cell lymphomas (PTCLs), the mutations identified were almost exclusively in the angioimmunoblastic T-cell lymphoma (AITL) and PTCL, not otherwise specified (PTCL-NOS) subtypes, the 2 most common PTCL entities in Western countries.10  These mutations were demonstrated in tumor cells, with evidence that they may be acquired either in early CD34+ progenitors or at later steps of lymphoid development.7 

In the present study, we extended the analysis of TET2 mutations to a larger, independent series of PTCL samples to correlate mutational status with pathologic features and clinical outcome.

Patients and tumor samples

A series of 190 PTCL patients with frozen lymphoma samples available at diagnosis were selected within the framework of a multicentric T-celllymphoma consortium (Tenomic). The study was approved by the local ethic committee (CPP Ile de France IX 08-009). Clinical data were collected retrospectively. All cases were reviewed and a consensus diagnosis was made at a multihead microscope by 3 hematopathologists according to the criteria of the 2008 World Health Organization classification.11 

Within PTCL-NOS, a group without typical morphology of AITL but expressing Th follicular (TFH) cell markers (ie, PD1, BCL6, and/or CXCL13) and/or having some other features reminiscent of AITL (ie, the presence of 2 of the following criteria: CD20+ large B cells, EBV-encoded small RNAs (EBER)+ cells, CD21 and/or CD23 follicular dendritic cell expansion, or CD10 expression), was designated “TFH-like” PTCL-NOS.

TET2 genotyping

DNA was extracted from several frozen tissue sections with morphologic control using QIAamp DNA mini Kit (QIAGEN) and an aliquot amplified by linear amplification. Analysis of the coding sequences of TET2 was performed by direct sequencing of the PCR fragments and candidate mutations were confirmed by sequencing, an independent PCR product was amplified from native genomic DNA as described previously.7  Frameshift, nonsense mutations, mutations in splice site, and missense mutations (only those affecting the evolutionary conserved regions of the protein) were considered. Single nucleotide polymorphisms either published previously or recorded in the National Center for Biotechnology Information Single Nucleotide Polymorphism database were excluded.

Statistical analysis

Continuous and dichotomic variables were compared with the Wilcoxon sum-rank test or Cochrane Armitage and χ2 or Fisher exact tests. Overall and progression-free survival (PFS) were estimated using the Kaplan-Meier method and compared with the log-rank test. All tests were 2-sided and P < .05 was considered significant.

TET2 mutations were found in 64 of 190 samples at diagnosis (34%). The frequency of TET2 mutations was extremely variable according to the diagnosis category. TET2 mutations were present in 40 of 86 (47%) cases of AITL, in 22 of 58 (38%) cases of PTCL-NOS, and in 2 of 10 (20%) cases of enteropathy-associated T-cell lymphoma. In contrast, no mutation was observed in any of the 18 anaplastic large-cell lymphomas (12 ALK-positive and 6 ALK-negative), the 12 extranodal NK/T lymphomas, nasal-type, or the 6 hepatosplenic T-cell lymphomas tested (Table 1).

Table 1

TET2 mutations in the different PTCL entities

PTCL entityTET2 mutation
nn%
AITL 86 40 47 
PTCL NOS* 58 22 38 
    TFH-like 24 14 58 
    Others 34 24 
ALCL 18 
EATL* 10 20 
Extranodal NK/T 12 
HSTL 
Total 190 64 34 
PTCL entityTET2 mutation
nn%
AITL 86 40 47 
PTCL NOS* 58 22 38 
    TFH-like 24 14 58 
    Others 34 24 
ALCL 18 
EATL* 10 20 
Extranodal NK/T 12 
HSTL 
Total 190 64 34 

ALCL indicates anaplastic large-cell lymphoma; HSTL, hepatosplenic T-cell lymphoma; EATL, enteropathy-associated T-cell lymphoma; and extranodal NK/T, extranodal NK/T-cell lymphoma, nasal-type.

*

TET2 mutations were found in 3 extranodal tissue samples (1 skin, 1 spleen, and 1 liver) diagnosed as PTCL-NOS and in 2 intestinal tissue samples diagnosed as EATL.

Interestingly, and in accordance with previous results,7  the highest frequency of mutations was found in AITL, which is thought to derive from TFH cells normally present in germinal centers.12–14  Within the large group of PTCL-NOS, it is also now recognized that a subset has “TFH-like” features in the form of a molecular profile bearing imprints of the TFH cell signature, the expression of TFH-associated molecules,15–17  and/or overlapping pathologic features.16  Therefore, we questioned whether TET2 mutations were correlated with TFH-like features in PTCL-NOS. Among the 58 PTCL-NOS in our series, 24 were classified as “TFH-like” PTCL-NOS. TET2 mutations were present in 14 of 24 (58%) of the TFH-like PTCL subgroup compared with 8 of 34 (22%) of those lacking AITL features and TFH-markers. Therefore, TET2 mutations in PTCL appear to be correlated with TFH derivation (ie, they occur at a higher frequency in AITL and TFH-like PTCL-NOS), than in non–TFH-like PTCL-NOS and other PTCL entities (49% vs 12.5%, P < .0001).

Among the 64 PTCL patients with TET2 mutations, 37 had single mutations and 27 had 2 or more mutations. TET2 mutations were mainly insertions/deletions generating frameshift and nonsense mutations similar to those found in myeloid malignancies (supplemental Table 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article). Eighteen of 19 missense mutations were predicted as probably damaging by the Polyphen-2 software (http://genetics.bwh.harvard.edu/pph2/) and one as possibly damaging. For 3 mutated patients with available biopsy at relapse, an identical pattern of TET2 mutations was found in paired samples (supplemental Figure 1), suggesting that TET2 mutations are associated with the main driving clone.

When focusing on AITL and PTCL-NOS patients (n = 144), 62 patients had TET2 mutations and 82 were TET2 wild-type. Clinical factors associated with TET2 mutated status (Table 2 and supplemental Figure 2) were advanced-stage disease (P = .02), increased number of involved extranodal sites (P = .017), presence of B symptoms (P = .02), thrombocytopenia (P = .04), high International Prognosis Index (P = .028) and Prognosis Model for PTCL-NOS18  (P = .025) scores, indicating that TET2 mutations are associated with adverse clinical parameters at presentation. Despite incomplete clinical data and heterogeneous therapies in this multicentric retrospective cohort, anthracycline-based chemotherapy was the most common regimen in both the mutated and nonmutated groups (73% and 70%, respectively). TET2-mutated patients with AITL or PTCL-NOS appeared to have a shorter PFS (P = .04) than TET2 wild-type patients, but no significant difference in overall survival was observed (P = .1; supplemental Figure 2). The association among TET2 mutations, poor prognosis clinical parameters, and outcome was even stronger when focusing on the group of patients with AITL and TFH-like PTCL-NOS. In this group, TET2 mutations were associated with a shorter PFS (P = .022) and a trend to a shorter overall survival (P = .058; supplemental Table 2 and supplemental Figure 3)

Table 2

Clinical data according to TET2 status in AITL and PTCL-NOS patients

nTET2 mutation
TET2 wild-type
P
6282
Sex 
    Male 38 61% 43 52% .18* 
    Female 24 39% 39 48% 
Median age, y 67  65  .3 
Stage 
    1 0% 5% .028 
    2 0% 4% 
    3 11 21% 17 23% 
    4 42 79% 49 67% 
Median no. of extranodal localizations   .056 
Lactate dehydrogenase 
    Elevated 41 76% 46 65% .24* 
    Normal 13 24% 25 35% 
Hemoglobin 
    ≥ 10 g/dL 35 74% 33 60% .14* 
    < 10 g/dL 12 26% 22 40% 
Platelet count 
    ≥ 150 000/mm3 34 72% 48 89% .04* 
    < 150 000/mm3 13 28% 11% 
Direct Coombs test 
    Positive 14 52% 13 43% .6* 
    Negative 13 48% 17 57% 
Hypergammaglobulinemia 
    Yes 11 32% 12 33% > .999* 
    No 23 68% 24 67% 
B symptoms 
    Yes 40 78% 38 58% .028* 
    No 11 22% 27 42% 
Performance status 
    0-1 22 44% 41 62% .06* 
    2-4 28 56% 25 38% 
International Prognosis Index score 
    0 0% 6% .0062 
    1 4% 6% 
    2 16% 11 16% 
    3 10 20% 22 33% 
    4 19 37% 23 34% 
    5 12 24% 4% 
Prognosis Model for PTCL18  
    0 5% 9% .0045 
    1 7% 11 24% 
    2 21% 13 29% 
    3 18 43% 13 29% 
    4 10 24% 9% 
Treatment 
    Anthracycline-based regimen 37 69% 40 58% .5* 
    Anthracycline-based regimen and frontline transplantation 4% 12% 
    Other combination of chemotherapy 13% 12% 
    Single-agent chemotherapy 9% 10% 
    Palliative treatment 6% 9% 
5-y overall survival 28%  31%  .11§ 
5-y PFS 10%  21%  .047§ 
nTET2 mutation
TET2 wild-type
P
6282
Sex 
    Male 38 61% 43 52% .18* 
    Female 24 39% 39 48% 
Median age, y 67  65  .3 
Stage 
    1 0% 5% .028 
    2 0% 4% 
    3 11 21% 17 23% 
    4 42 79% 49 67% 
Median no. of extranodal localizations   .056 
Lactate dehydrogenase 
    Elevated 41 76% 46 65% .24* 
    Normal 13 24% 25 35% 
Hemoglobin 
    ≥ 10 g/dL 35 74% 33 60% .14* 
    < 10 g/dL 12 26% 22 40% 
Platelet count 
    ≥ 150 000/mm3 34 72% 48 89% .04* 
    < 150 000/mm3 13 28% 11% 
Direct Coombs test 
    Positive 14 52% 13 43% .6* 
    Negative 13 48% 17 57% 
Hypergammaglobulinemia 
    Yes 11 32% 12 33% > .999* 
    No 23 68% 24 67% 
B symptoms 
    Yes 40 78% 38 58% .028* 
    No 11 22% 27 42% 
Performance status 
    0-1 22 44% 41 62% .06* 
    2-4 28 56% 25 38% 
International Prognosis Index score 
    0 0% 6% .0062 
    1 4% 6% 
    2 16% 11 16% 
    3 10 20% 22 33% 
    4 19 37% 23 34% 
    5 12 24% 4% 
Prognosis Model for PTCL18  
    0 5% 9% .0045 
    1 7% 11 24% 
    2 21% 13 29% 
    3 18 43% 13 29% 
    4 10 24% 9% 
Treatment 
    Anthracycline-based regimen 37 69% 40 58% .5* 
    Anthracycline-based regimen and frontline transplantation 4% 12% 
    Other combination of chemotherapy 13% 12% 
    Single-agent chemotherapy 9% 10% 
    Palliative treatment 6% 9% 
5-y overall survival 28%  31%  .11§ 
5-y PFS 10%  21%  .047§ 
*

Fisher exact test.

Wilcoxon sum-rank test.

Cochran Armitage test.

§

Log-rank test.

We recently reported recurrent IDH2 mutations in AITL.19  These mutations alter IDH enzymatic function and may result in the functional inactivation of TET protein activity.20  In acute myeloid leukemia, IDH and TET2 mutations are reported to be mutually exclusive.20  Of the 31 AITL samples in the present study also analyzed for IDH2 mutations,19  6 had both IDH2 and TET2 mutations, 2 were IDH2 mutated/TET2 wild-type, and 10 were TET2 mutated/IDH2 wild-type, indicating that both mutations can accumulate in AITL. Ultimately, both IDH2 and TET2 mutations, which are recurrent in AITL and in TFH-derived PTCL, may deregulate the control of chromatin structure. This situation may be reminiscent of that observed in B-cell lymphomas, in which mutations of several genes involving epigenetic changes and chromatin remodeling, such as CREEBP, EP300,21 EZH2,22 MLL2, or MEF2B,23  have been described recently.

In addition to similarities in their ontogeny, AITLs and a subset of TFH-like PTCLs share common oncogenic alterations, including TET2 abnormalities, further supporting their close relationship. The strong association among TET2 mutations, AITL, and TFH-like PTCL may provide molecular rationale to merge together AITL and PTCL-NOS–expressing TFH markers, as has been suggested previously,15–16  and to refine the spectrum of AITL. Overall, the identification of frequent TET2 mutations in AITL and other TFH-related PTCLs extends the importance of epigenetic alterations in lymphomagenesis. The results of the present study suggest that in these diseases, in which conventional chemotherapies are ineffective24  in most patients, the use of new agents such as demethylating agents should be considered.

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 Virginie Fataccioli for the management of the TENOMIC program and Magalie Abrantes, Prof K Leroy (Tumor Tissue Bank), Jérome Cuvillier, and Nadine Vailhen for their assistance.

This study was supported in part by grants from the Institut National du Cancer, Programme Hospitalier de Recherche Clinique, Fondation pour la recherche Médicale (FRM; équipe labellisée), and the Cancer Plan Research Program (Belgium; L.d.L.). F.L. was an M2 student receiving grants from Association pour la Recherche contre le Cancer, and M.T. received grants from FRM.

Contribution: O.B., C.B., and P.G. designed the research: F.L., P.G., L.d.L., and M.P. analyzed and interpreted the results; L.C., T.M., and R.A.C. performed the study; L.L., O.T., T.R., B.F., and M.T. collected the data and/or the tumor samples; J.P.J. supervised statistical analysis; and F.L., L.d.L., and P.G. wrote the paper.

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

Correspondence: Pr Philippe Gaulard, Département de Pathologie, Hôpital Henri Mondor, Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; e-mail: philippe.gaulard@hmn.aphp.fr.

1
Delhommeau
 
F
Dupont
 
S
Della Valle
 
V
, et al. 
Mutation in TET2 in myeloid cancers.
N Engl J Med
2009
, vol. 
360
 
22
(pg. 
2289
-
2301
)
2
Abdel-Wahab
 
O
Mullally
 
A
Hedvat
 
C
, et al. 
Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies.
Blood
2009
, vol. 
114
 
1
(pg. 
144
-
147
)
3
Tahiliani
 
M
Koh
 
KP
Shen
 
Y
, et al. 
Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1.
Science
2009
, vol. 
324
 
5929
(pg. 
930
-
935
)
4
Ito
 
S
Shen
 
L
Dai
 
Q
, et al. 
Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine.
Science
2011
, vol. 
333
 
6047
(pg. 
1300
-
1303
)
5
Cimmino
 
L
Abdel-Wahab
 
O
Levine
 
RL
Aifantis
 
I
TET family proteins and their role in stem cell differentiation and transformation.
Cell Stem Cell
2011
, vol. 
9
 
3
(pg. 
193
-
204
)
6
Moran-Crusio
 
K
Reavie
 
L
Shih
 
A
, et al. 
Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation.
Cancer Cell
2011
, vol. 
20
 
1
(pg. 
11
-
24
)
7
Quivoron
 
C
Couronne
 
L
Della Valle
 
V
, et al. 
TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis.
Cancer Cell
2011
, vol. 
20
 
1
(pg. 
25
-
38
)
8
Li
 
Z
Cai
 
X
Cai
 
CL
, et al. 
Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies.
Blood
2011
, vol. 
118
 
17
(pg. 
4509
-
4518
)
9
Couronné
 
L
Bastard
 
C
Bernard
 
OA
TET2 and DNMT3A mutations in human T-cell lymphoma.
N Engl J Med
2012
, vol. 
366
 
1
(pg. 
95
-
96
)
10
Vose
 
J
Armitage
 
J
Weisenburger
 
D
International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes.
J Clin Oncol
2008
, vol. 
26
 
25
(pg. 
4124
-
4130
)
11
Swerdlow
 
S
Campo
 
E
Harris
 
NL
International Agency for Research on Cancer.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissue
2008
Geneva, Switzerland
World Health Organization
12
Dupuis
 
J
Boye
 
K
Martin
 
N
, et al. 
Expression of CXCL13 by neoplastic cells in angioimmunoblastic T-cell lymphoma (AITL): a new diagnostic marker providing evidence that AITL derives from follicular helper T cells.
Am J Surg Pathol
2006
, vol. 
30
 
4
(pg. 
490
-
494
)
13
de Leval
 
L
Rickman
 
DS
Thielen
 
C
, et al. 
The gene expression profile of nodal peripheral T-cell lymphoma demonstrates a molecular link between angioimmunoblastic T-cell lymphoma (AITL) and follicular helper T (TFH) cells.
Blood
2007
, vol. 
109
 
11
(pg. 
4952
-
4963
)
14
Piccaluga
 
PP
Agostinelli
 
C
Califano
 
A
, et al. 
Gene expression analysis of angioimmunoblastic lymphoma indicates derivation from T follicular helper cells and vascular endothelial growth factor deregulation.
Cancer Res
2007
, vol. 
67
 
22
(pg. 
10703
-
10710
)
15
de Leval
 
L
Gaulard
 
P
Pathobiology and molecular profiling of peripheral T-cell lymphomas.
Hematology Am Soc Hematol Educ Program
2008
, vol. 
2008
 (pg. 
272
-
279
)
16
Rodríguez-Pinilla
 
SM
Atienza
 
L
Murillo
 
C
, et al. 
Peripheral T-cell lymphoma with follicular T-cell markers.
Am J Surg Pathol
2008
, vol. 
32
 
12
(pg. 
1787
-
1799
)
17
Iqbal
 
J
Weisenburger
 
DD
Greiner
 
TC
, et al. 
Molecular signatures to improve diagnosis in peripheral T-cell lymphoma and prognostication in angioimmunoblastic T-cell lymphoma.
Blood
2010
, vol. 
115
 
5
(pg. 
1026
-
1036
)
18
Gallamini
 
A
Stelitano
 
C
Calvi
 
R
, et al. 
Peripheral T-cell lymphoma unspecified (PTCL-U): a new prognostic model from a retrospective multicentric clinical study.
Blood
2004
, vol. 
103
 
7
(pg. 
2474
-
2479
)
19
Cairns
 
RA
Iqbal
 
J
Lemonnier
 
F
, et al. 
IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma.
Blood
2012
, vol. 
119
 
8
(pg. 
1901
-
1903
)
20
Figueroa
 
ME
Abdel-Wahab
 
O
Lu
 
C
, et al. 
Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation.
Cancer Cell
2010
, vol. 
18
 
6
(pg. 
553
-
567
)
21
Pasqualucci
 
L
Dominguez-Sola
 
D
Chiarenza
 
A
, et al. 
Inactivating mutations of acetyltransferase genes in B-cell lymphoma.
Nature
2011
, vol. 
471
 
7337
(pg. 
189
-
195
)
22
Morin
 
RD
Johnson
 
NA
Severson
 
TM
, et al. 
Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin.
Nat Genet
2010
, vol. 
42
 
2
(pg. 
181
-
185
)
23
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
)
24
Mourad
 
N
Mounier
 
N
Briere
 
J
, et al. 
Clinical, biologic, and pathologic features in 157 patients with angioimmunoblastic T-cell lymphoma treated within the Groupe d'Etude des Lymphomes de l'Adulte (GELA) trials.
Blood
2008
, vol. 
111
 
9
(pg. 
4463
-
4470
)

Author notes

*

F.L. and L.C. contributed equally to this work.

L.d.L. and P.G. contributed equally to this work.

Sign in via your Institution