TO THE EDITOR:

Indolent T-cell lymphoproliferative disorder of the gastrointestinal tract (GI TLPD) is a newly recognized entity in the World Health Organization (WHO) classification of lymphoid neoplasms.1  GI TLPD is defined as a clonal T-cell proliferation occurring in the GI tract, most commonly in the colon and small bowel, with bland cytologic appearance, a generally noninvasive architectural pattern, and indolent clinical behavior. Patients typically are adults (with a predominance of males) who present with GI symptoms, including abdominal pain, vomiting, diarrhea, and weight loss. Unlike enteropathy-associated T-cell lymphoma (EATL), from which it must be distinguished, GI TLPD is not associated with underlying celiac disease, and its etiology and molecular pathogenesis remain unknown. GI TLPD tends not to respond to chemotherapy and, despite its indolent nature, symptoms are often debilitating and challenging to treat.2 

We examined the pathological and genetic features of GI TLPD to better understand this new entity (see supplemental Data, available on the Blood Web site). By reviewing clinical records and pathological material, we identified and confirmed 10 patients with GI TLPD and 1 patient with natural killer cell enteropathy, which is considered a separate entity by WHO criteria.1-3  There were 6 males and 5 females (mean age, 56 years; range, 39-74 years; Table 1). Histologic features of GI TLPD were similar to those previously reported (Figure 1A-B).1,2  Of the patients with GI TLPD, 5 had a CD4+ T-cell phenotype, 4 had a CD8+ phenotype, and 1 had a double-positive phenotype (Table 1). Clonal T-cell receptor gene rearrangements were identified in all 10 patients with GI TLPD but not in the patient with natural killer cell enteropathy. Samples from four patients underwent testing by FoundationOne Heme (Foundation Medicine, Cambridge, MA; supplemental Table 1). Notably, 1 CD4+ GI TLPD sample was found to have a STAT3-JAK2 fusion. JAK2 fusions have been identified in a variety of myeloid and precursor lymphoid neoplasms4,5  but are rare in mature T-cell lymphomas6-9 ; among 200 Mayo Clinic patients with a variety of T-cell lymphoma subtypes previously studied by fluorescence in situ hybridization (FISH) using a JAK2 break-apart probe, all were negative for rearrangement.7 

Table 1.

Characteristics of 11 patients with indolent GI TLPD or NK-cell enteropathy

PatientAgeSexGI site*CD3CD4CD5CD7CD8CD56TIA1GrzBTCR βF1TCR γδSTAT3-JAK2 fusionpSTAT1Y701pSTAT3Y705pSTAT5Y694
CD4+GI TLPD                  
 1 54 D,J − − − − − − Yes − − 
 2 48 S,D,J,I − − − − − Yes − − 
 3 51 D,J − − − − − − Yes − 
 4 70 S,D,J − − − − − − Yes − − − 
 5 51 D,J,I,C − − − − − No − − 
CD8+GI TLPD                  
 6 47 S,D,J,I − − − − − No − − 
 7 39 S,D,J,I,C − − − − No − − − 
 8 74 D,J − − − − No − − 
 9 57 − − − − No − − − 
CD4+/CD8+GI TLPD                  
 10 60 D,J − − − − − No   
NK-cell enteropathy                  
 11 68 S,D,J − − − − − − No  − − 
PatientAgeSexGI site*CD3CD4CD5CD7CD8CD56TIA1GrzBTCR βF1TCR γδSTAT3-JAK2 fusionpSTAT1Y701pSTAT3Y705pSTAT5Y694
CD4+GI TLPD                  
 1 54 D,J − − − − − − Yes − − 
 2 48 S,D,J,I − − − − − Yes − − 
 3 51 D,J − − − − − − Yes − 
 4 70 S,D,J − − − − − − Yes − − − 
 5 51 D,J,I,C − − − − − No − − 
CD8+GI TLPD                  
 6 47 S,D,J,I − − − − − No − − 
 7 39 S,D,J,I,C − − − − No − − − 
 8 74 D,J − − − − No − − 
 9 57 − − − − No − − − 
CD4+/CD8+GI TLPD                  
 10 60 D,J − − − − − No   
NK-cell enteropathy                  
 11 68 S,D,J − − − − − − No  − − 

F, female; M, male.

*

GI sites of involvement: C, colon; D, duodenum; I, ileum ; J, jejunum; S, stomach.

Insufficient material or technically uninterpretable.

No fusion reported by FoundationOne Heme testing; FISH studies technically uninterpretable.

Figure 1.

Indolent T-cell lymphoproliferative disorder of the gastrointestinal tract with STAT3-JAK2 fusion. (A-B) Duodenal biopsy shows a monotonous population of small lymphocytes in the lamina propria with preservation of the glands (hematoxylin and eosin stain; patient 2; original magnification ×40 [A] and ×400 [B]). By immunohistochemistry, the cells are positive for (C) CD3 and (D) CD4 and negative for (E) CD8 (original magnification ×400 [C-E]). (F) FISH using a break-apart probe to the JAK2 gene region shows separations of red and green signals, indicating the presence of a rearrangement (original magnification ×600). (G) FISH using a dual-fusion probe for the STAT3 (red) and JAK2 (green) gene regions shows red-green fusion signals (f), indicating the presence of fusion of these 2 regions (original magnification ×600). (H) RNA sequencing identifies chimeric reads supporting the presence of STAT3-JAK2 fusion joining the end of exon 23 of STAT3 with the beginning of exon 17 of JAK2. The identical breakpoint was found in all 3 patients who had GI TLPD with STAT3-JAK2 fusion whose samples were successfully sequenced. (I) Sanger sequencing confirms the STAT3-JAK2 breakpoint identified by RNA sequencing. (J) Protein domain structure of STAT3, JAK2, and predicted STAT3-JAK2 fusion protein. The amino acid numbers at the breakpoints are shown as well as the location of STAT3Y705. Using immunohistochemistry showed that the cells lack staining for (K) pSTAT3Y705 but are positive for (L) pSTAT5Y694 (patient 2; original magnification ×1000). CCD, coiled coil domain; DBD, DNA binding domain; FERM, four-point-one, ezrin, radixin, moesin homology domain; JH1, JAK homology 1 (tyrosine kinase) domain; JH2, JAK homology 2 (pseudokinase) domain; LD, linker domain; NTD, N-terminal domain; SH2, Src homology 2 domain; TAD, transcription activation domain.

Figure 1.

Indolent T-cell lymphoproliferative disorder of the gastrointestinal tract with STAT3-JAK2 fusion. (A-B) Duodenal biopsy shows a monotonous population of small lymphocytes in the lamina propria with preservation of the glands (hematoxylin and eosin stain; patient 2; original magnification ×40 [A] and ×400 [B]). By immunohistochemistry, the cells are positive for (C) CD3 and (D) CD4 and negative for (E) CD8 (original magnification ×400 [C-E]). (F) FISH using a break-apart probe to the JAK2 gene region shows separations of red and green signals, indicating the presence of a rearrangement (original magnification ×600). (G) FISH using a dual-fusion probe for the STAT3 (red) and JAK2 (green) gene regions shows red-green fusion signals (f), indicating the presence of fusion of these 2 regions (original magnification ×600). (H) RNA sequencing identifies chimeric reads supporting the presence of STAT3-JAK2 fusion joining the end of exon 23 of STAT3 with the beginning of exon 17 of JAK2. The identical breakpoint was found in all 3 patients who had GI TLPD with STAT3-JAK2 fusion whose samples were successfully sequenced. (I) Sanger sequencing confirms the STAT3-JAK2 breakpoint identified by RNA sequencing. (J) Protein domain structure of STAT3, JAK2, and predicted STAT3-JAK2 fusion protein. The amino acid numbers at the breakpoints are shown as well as the location of STAT3Y705. Using immunohistochemistry showed that the cells lack staining for (K) pSTAT3Y705 but are positive for (L) pSTAT5Y694 (patient 2; original magnification ×1000). CCD, coiled coil domain; DBD, DNA binding domain; FERM, four-point-one, ezrin, radixin, moesin homology domain; JH1, JAK homology 1 (tyrosine kinase) domain; JH2, JAK homology 2 (pseudokinase) domain; LD, linker domain; NTD, N-terminal domain; SH2, Src homology 2 domain; TAD, transcription activation domain.

Close modal

To confirm the JAK2 rearrangement and determine whether it was recurrent in GI TLPD, we performed FISH on formalin-fixed, paraffin-embedded tissue sections using a previously published break-apart probe.7,10  The initial sample identified by FoundationOne Heme and samples from 3 additional patients showed rearrangements of the JAK2 gene region on 9p24.1; they represented 4 of 5 patients with a CD4+ phenotype, whereas patients with other phenotypes lacked rearrangements (Table 1; Figure 1C-F). We tested 23 EATLs (in addition to 4 EATLs previously studied7 ) using a JAK2 break-apart FISH probe, including 9 type II EATLs, now designated as monomorphic epitheliotropic intestinal T-cell lymphomas in the WHO classification11 ; all 27 samples were negative. We then designed a STAT3-JAK2 dual-fusion FISH probe and demonstrated that all 4 GI TLPDs demonstrated fusion signals (Figure 1G), confirming a recurrent t(9;17)(p24.1;q21.2) rearrangement.

To evaluate the precise breakpoints of the fusion transcripts, we performed RNA sequencing from formalin-fixed, paraffin-embedded tissue and obtained interpretable results in 3 of the 4 patients with t(9;17)(p24.1;q21.2) rearrangements. All 3 showed STAT3-JAK2 fusion with the identical messenger RNA breakpoint (Figure 1J), confirmed by Sanger sequencing (Figure 1I). The predicted fusion protein retained the C-terminal JH1 kinase domain of JAK2 and a portion of the JH2 pseudokinase domain but lacked the upstream SH2 and FERM domains (Figure 1J), similar to other JAK2 fusions.4,5,7  Reciprocal JAK2-STAT3 fusion transcripts were also detected in all 3 sequenced samples; however, the breakpoint was different in each sample and likely nonfunctional (1 sample out of frame and 2 with predicted proteins lacking the JAK2 tyrosine kinase domain and virtually all of the STAT3 coding region). These data demonstrate recurrent STAT3-JAK2 fusions in GI TLPDs. The exclusivity for CD4+ samples and identical breakpoints strongly suggest that STAT3-JAK2 fusions contribute to the pathogenesis of this disorder.

STAT3 activation is critical in many T-cell neoplasms, and activating mutations of the STAT3 gene are relatively common.12-16  However, Perry et al2  found minimal phosphorylated STAT3 (pSTAT3) in GI TLPDs and did not observe any STAT3 SH2 domain hotspot mutations. To determine whether pSTAT3 might be present in GI TLPDs with STAT3-JAK2 fusions, we performed immunohistochemistry for pSTAT3Y705 and samples from all 4 patients were negative (Figure 1K), as were samples from 5 of 7 patients who did not have STAT3-JAK2 fusion (Table 1). Although the functionally critical Y705 amino acid of STAT3 was present in the predicted STAT3-JAK2 fusion protein, the loss of the C-terminal portion of the STAT3 transcription activation domain or conformational alterations might inhibit Y705 phosphorylation. Notably, however, the N-terminal domain essential for formation of unphosphorylated STAT3 dimers17  was also retained in the predicted STAT3-JAK2 fusion protein. Interestingly, STAT3-RARA fusions recently were described in acute promyelocytic leukemia, and coimmunoprecipitation studies confirmed the ability of the resultant fusion proteins to homodimerize.18  JAK2 fusion partners such as TEL and PCM1 have been shown to contribute functionally to oncogenic signaling by facilitating fusion protein dimerization, leading to STAT5 activation.4,5,7  Therefore, we examined pSTAT5 by immunohistochemistry and indeed 3 of 4 GI TLPD samples with STAT3-JAK2 fusions were positive (Figure 1L) compared with only 1 of 6 evaluable samples without fusions. The patient sample that was negative for pSTAT5 was also negative for pSTAT1, as were samples from 6 of 8 additional patients tested.

Although limited follow-up data were available, 2 patients developed overt T-cell lymphomas. Patient 4 had a CD4+ GI TLPD with STAT3-JAK2 fusion and subsequently developed a large-cell transformation that shared the same fusion (supplemental Figure 1). The WHO classification notes that GI TLPDs expressing CD4 seem to have a higher risk of progression than those expressing CD8.1,19  Interestingly, FISH in the large-cell component from patient 4 also showed copy number gains of STAT3, present in a previously reported CD4+ GI TLPD with large-cell transformation.19  Patient 9 had a CD8+ GI TLPD that lacked JAK2 rearrangement and subsequently developed systemic anaplastic lymphoma kinase–negative anaplastic large-cell lymphoma. Polymerase chain reaction studies identified different clonal T-cell receptor gene rearrangements in the 2 specimens (not shown), suggesting that the anaplastic large-cell lymphoma arose as a second, distinct process.

Taken together, our findings conclusively demonstrate STAT3-JAK2 fusions in GI TLPD, representing the first recurrent genetic abnormality reported in this disease. Fusions were seen in 4 of 5 patients with CD4+ phenotypes but not in other phenotypes, suggesting that the molecular pathogenesis of GI TLPD may vary on the basis of cell of origin and that distinct molecular subentities may exist. FISH for JAK2 rearrangements might help distinguish GI TLPD from reactive processes because GI TLPDs are cytologically bland and may lack phenotypic aberrancy. Furthermore, STAT3-JAK2 fusions represent a candidate therapeutic target in GI TLPD. A variety of strategies to inhibit JAK-STAT signaling are clinically available or in preclinical development for lymphoma, other malignancies, and nonneoplastic conditions.20,21  T-cell lymphoma cell lines with PCM1-JAK2 fusion are sensitive to the selective JAK2 inhibitor TG101348,7  whereas clinical responses to the JAK1/2 inhibitor ruxolitinib have been reported in myeloid neoplasms with PCM1-JAK2.22-24  Thus, the activity of JAK2 inhibitors in GI TLPD with STAT3-JAK2 fusion merits further study.

The online version of this article contains a data supplement.

The authors thank Jin S. Jang in the Mayo Clinic Genome Analysis Core, Medical Genome Facility, for his contributions to developing the RNA sequencing pipeline.

This work was supported by National Institutes of Health awards R01 CA177734 (A.L.F.), P30 CA15083 (Mayo Clinic Cancer Center), and P50 CA97274 (University of Iowa/Mayo Clinic Lymphoma Specialized Program of Research Excellence) from the National Cancer Institute, Clinical and Translational Science Award UL1 TR002377 from the National Center for Advancing Translational Science, and the Department of Laboratory Medicine and Pathology and the Clinomics Program of the Center for Individualized Medicine, both at Mayo Clinic.

Contribution: A.S., J.A.M., and A.L.F. designed the study; N.O., G.H., H.K.B., D.L.K., and S.M.K.-N. performed experiments; A.S., K.N.L., N.N.B., G.S.N., and J.A.M. interpreted clinical data; N.O., T.-T.W., and A.L.F. interpreted pathology data; R.L.B., A.A.N., S.D., and A.L.F. interpreted genetic findings; R.P.K. and P.T.G. interpreted FISH results; R.H. interpreted T-cell receptor gene rearrangement studies; B.W.E., J.J., J.I.D., and S.D. interpreted RNA quality control and sequencing data; A.L.F. drafted the manuscript; and all authors approved the final manuscript.

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

Correspondence: Joseph A. Murray, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St SW, Rochester, MN 55905; e-mail: murray.joseph@mayo.edu; and Andrew L. Feldman, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First St SW, Hilton Building, Room 800F, Rochester, MN 55905; e-mail: feldman.andrew@mayo.edu.

1.
Jaffe
ES
,
Chott
A
,
Ott
G
, et al
.
Indolent T-cell lymphoproliferative disorder of the gastrointestinal tract
. In:
Swerdlow
SH
,
Camp
E
,
Harris
NL
, et al
, eds.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
.
Lyon, France
:
International Agency for Research on Cancer
;
2017
:
379
-
380
.
2.
Perry
AM
,
Warnke
RA
,
Hu
Q
, et al
.
Indolent T-cell lymphoproliferative disease of the gastrointestinal tract
.
Blood
.
2013
;
122
(
22
):
3599
-
3606
.
3.
Mansoor
A
,
Pittaluga
S
,
Beck
PL
,
Wilson
WH
,
Ferry
JA
,
Jaffe
ES
.
NK-cell enteropathy: a benign NK-cell lymphoproliferative disease mimicking intestinal lymphoma: clinicopathologic features and follow-up in a unique case series
.
Blood
.
2011
;
117
(
5
):
1447
-
1452
.
4.
Smith
CA
,
Fan
G
.
The saga of JAK2 mutations and translocations in hematologic disorders: pathogenesis, diagnostic and therapeutic prospects, and revised World Health Organization diagnostic criteria for myeloproliferative neoplasms
.
Hum Pathol
.
2008
;
39
(
6
):
795
-
810
.
5.
Lacronique
V
,
Boureux
A
,
Valle
VD
, et al
.
A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia
.
Science
.
1997
;
278
(
5341
):
1309
-
1312
.
6.
Davis
TH
,
Morton
CC
,
Miller-Cassman
R
,
Balk
SP
,
Kadin
ME
.
Hodgkin’s disease, lymphomatoid papulosis, and cutaneous T-cell lymphoma derived from a common T-cell clone
.
N Engl J Med
.
1992
;
326
(
17
):
1115
-
1122
.
7.
Ehrentraut
S
,
Nagel
S
,
Scherr
ME
, et al
.
t(8;9)(p22;p24)/PCM1-JAK2 activates SOCS2 and SOCS3 via STAT5
.
PLoS One
.
2013
;
8
(
1
):
e53767
.
8.
Adélaïde
J
,
Pérot
C
,
Gelsi-Boyer
V
, et al
.
A t(8;9) translocation with PCM1-JAK2 fusion in a patient with T-cell lymphoma
.
Leukemia
.
2006
;
20
(
3
):
536
-
537
.
9.
Panagopoulos
I
,
Gorunova
L
,
Spetalen
S
, et al
.
Fusion of the genes ataxin 2 like,ATXN2L, and Janus kinase 2,JAK2, in cutaneous CD4 positive T-cell lymphoma
.
Oncotarget
.
2017
;
8
(
61
):
103775
-
103784
.
10.
Patnaik
MM
,
Knudson
RA
,
Gangat
N
, et al
.
Chromosome 9p24 abnormalities: prevalence, description of novel JAK2 translocations, JAK2V617F mutation analysis and clinicopathologic correlates
.
Eur J Haematol
.
2010
;
84
(
6
):
518
-
524
.
11.
Jaffe
ES
,
Chott
A
,
Ott
G
, et al
.
Monomorphic epitheliotropic intestinal T-cell lymphoma
. In:
Swerdlow
SH
,
Camp
E
,
Harris
NL
, et al
, eds.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
.
Lyon
:
International Agency for Research on Cancer
;
2017
:
377
-
378
.
12.
Koskela
HL
,
Eldfors
S
,
Ellonen
P
, et al
.
Somatic STAT3 mutations in large granular lymphocytic leukemia
.
N Engl J Med
.
2012
;
366
(
20
):
1905
-
1913
.
13.
Ohgami
RS
,
Ma
L
,
Merker
JD
,
Martinez
B
,
Zehnder
JL
,
Arber
DA
.
STAT3 mutations are frequent in CD30+ T-cell lymphomas and T-cell large granular lymphocytic leukemia
.
Leukemia
.
2013
;
27
(
11
):
2244
-
2247
.
14.
Küçük
C
,
Jiang
B
,
Hu
X
, et al
.
Activating mutations of STAT5B and STAT3 in lymphomas derived from γδ-T or NK cells
.
Nat Commun
.
2015
;
6
(
1
):
6025
.
15.
Crescenzo
R
,
Abate
F
,
Lasorsa
E
, et al
;
European T-Cell Lymphoma Study Group, T-Cell Project: Prospective Collection of Data in Patients with Peripheral T-Cell Lymphoma and the AIRC 5xMille Consortium “Genetics-Driven Targeted Management of Lymphoid Malignancies”
.
Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma
.
Cancer Cell
.
2015
;
27
(
4
):
516
-
532
.
16.
Shi
M
,
He
R
,
Feldman
AL
, et al
.
STAT3 mutation and its clinical and histopathologic correlation in T-cell large granular lymphocytic leukemia
.
Hum Pathol
.
2018
;
73
:
74
-
81
.
17.
Vogt
M
,
Domoszlai
T
,
Kleshchanok
D
, et al
.
The role of the N-terminal domain in dimerization and nucleocytoplasmic shuttling of latent STAT3
.
J Cell Sci
.
2011
;
124
(
Pt 6
):
900
-
909
.
18.
Yao
L
,
Wen
L
,
Wang
N
, et al
.
Identification of novel recurrentSTAT3-RARAfusions in acute promyelocytic leukemia lacking t(15;17)(q22;q12)/PML-RARA
.
Blood
.
2018
;
131
(
8
):
935
-
939
.
19.
Margolskee
E
,
Jobanputra
V
,
Lewis
SK
,
Alobeid
B
,
Green
PH
,
Bhagat
G
.
Indolent small intestinal CD4+ T-cell lymphoma is a distinct entity with unique biologic and clinical features
.
PLoS One
.
2013
;
8
(
7
):
e68343
.
20.
Scott
LM
,
Gandhi
MK
.
Deregulated JAK/STAT signalling in lymphomagenesis, and its implications for the development of new targeted therapies
.
Blood Rev
.
2015
;
29
(
6
):
405
-
415
.
21.
Roskoski
R
Jr
.
Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases
.
Pharmacol Res
.
2016
;
111
:
784
-
803
.
22.
Lierman
E
,
Selleslag
D
,
Smits
S
,
Billiet
J
,
Vandenberghe
P
.
Ruxolitinib inhibits transforming JAK2 fusion proteins in vitro and induces complete cytogenetic remission in t(8;9)(p22;p24)/PCM1-JAK2-positive chronic eosinophilic leukemia
.
Blood
.
2012
;
120
(
7
):
1529
-
1531
.
23.
Rumi
E
,
Milosevic
JD
,
Casetti
I
, et al
.
Efficacy of ruxolitinib in chronic eosinophilic leukemia associated with a PCM1-JAK2 fusion gene
.
J Clin Oncol
.
2013
;
31
(
17
):
e269
-
e271
.
24.
Rumi
E
,
Milosevic
JD
,
Selleslag
D
, et al
.
Efficacy of ruxolitinib in myeloid neoplasms with PCM1-JAK2 fusion gene
.
Ann Hematol
.
2015
;
94
(
11
):
1927
-
1928
.

Author notes

*

J.A.M. and A.L.F. contributed equally to this work.

Sign in via your Institution