Recently, the landscape of single base mutations in diffuse large B-cell lymphoma (DLBCL) was described. Here we report the discovery of a gene fusion between TBL1XR1 and TP63, the only recurrent somatic novel gene fusion identified in our analysis of transcriptome data from 96 DLBCL cases. Based on this cohort and a further 157 DLBCL cases analyzed by FISH, the incidence in de novo germinal center B cell–like (GCB) DLBCL is 5% (6 of 115). The fusion appears exclusive to GCB and was not seen in 138 non-GCB cases examined (P = .008, Fisher exact test) but was present at low incidence in follicular lymphoma (1 of 81). In all 7 cases identified, the 3′ end of the fusion consists of exons 4 and onwards of TP63. The recurrence, subtype enrichment, and the remarkably conserved nature of the TP63 portion of the fusion suggest an important functional role in the lymphomas that harbor this event.

Chromosomal rearrangements are a hallmark of hematologic malignancy. The 3 most recurrent translocations in diffuse large B-cell lymphoma (DLBCL) were discovered by examination of karyotypes, with the partner genes identified in the 1980s and 1990s.1  Translocations involving BCL6, BCL2, and MYC are reported in 30%, 20% to 30%, and up to 10% of de novo DLBCLs, respectively.2 

Recently, we and others have described the mutational landscape of DLBCL, focusing on single nucleotide variations and small insertion/deletions.3-5  Analysis of transcriptomes also offers the opportunity to identify novel fusion gene transcripts resulting from cryptic chromosomal rearrangements hitherto unsuspected from karyotype analysis, which is limited by resolution and the complexity of the chromosomal events.6 

TP63 is a paralog of the tumor-suppressive transcription factor TP53.7  In contrast to TP53, TP63 is rarely mutated in malignancy.8-10  However, overexpression of ΔN-TP63, a set of TP63 isoforms lacking the transactivation (TA) domain, has been associated with malignancies of epithelial origin.11,12 TBL1XR1 encodes a protein that is part of the NCoR/SMRT transcription repressor complex.13  Recently, deletion of the TBL1XR1 gene locus has been described in DLBCL4  and primary CNS lymphoma.14  Here, we describe a novel recurrent gene fusion involving TP63 and TBL1XR1 discovered during analysis of DLBCL transcriptomes.

To identify candidate gene fusion transcripts, Trans-ABySS15  and deFuse16  were applied to the previously described3  transcriptome data generated from 96 DLBCL and 13 follicular lymphoma (FL) cases. Validation of candidate fusion transcripts was achieved using Sanger sequencing of amplicons produced by RT-PCR with primers specific for the predicted gene fusion (supplemental Table 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article). Cell of origin was assigned using gene expression profiling data when available. The tally algorithm17  was applied to IHC data to assign cell of origin for the remaining biopsies (supplemental Table 2).

FISH was performed on tissue microarrays as previously described.18  Separate breakapart assays were performed for TBL1XR1 and TP63 (for probe design, see supplemental Table 3). Images, captured using the Ariol imaging system (Leica Microsystems), were scored independently by 2 persons. Quantitative RT-PCR was performed using SYBR Green and primers for wild-type TBL1XR1 and the TBL1XR1/TP63 fusion (supplemental Table 4). The study was approved by the University of British Columbia–British Columbia Cancer Agency Research Ethics Board.

We identified evidence for fusion transcripts from massively parallel RNA sequencing (RNA-seq) of pretreatment de novo DLBCL biopsies. A novel gene fusion involving TBL1XR1 and TP63 was seen in the tumors from 4 patients (Figure 1). The existence of the TBL1XR1/TP63 fusion was validated in all 4 cases by Sanger sequencing of amplicons generated using RT-PCR. The breakpoints in the genomic DNA were identified in all cases (Figure 1B), and the genetic rearrangement was shown to be somatic by PCR and by whole genome shotgun sequencing of the constitutional DNA of one case. The TBL1XR1/TP63 fusion was the only recurrent novel somatic gene fusion observed.

Figure 1

TBL1XR1/TP63 gene fusion observed using paired-end massively parallel RNA sequencing and the genomic fusion breakpoints. (A) Top panel: 77 paired read sequences are shown aligning on either side of the fusion point pairing TBL1XR1 and TP63 in one case of DLBCL. Genomic coordinates of exon boundaries are shown. Bottom panel: 17 split-reads are shown that cross the fusion junction, aligned to the merged sequence of the 2 genes. The histogram on the right of this panel shows the frequency of each split-read, a total of 55 split-reads. (B) An ideogram of chromosome 3 is shown, indicating the locations of TBL1XR1 and TP63 at 3q26.32 and 3q28, respectively. The gene fusions result from chromosomal rearrangement(s) that include an inversion event. Arrows above the genes indicate the direction of transcription. At the bottom, the genomic fusion breakpoints are shown for the 5 cases detected by RNA sequencing. Genomic coordinates are given for the fusion breakpoints and the exon boundaries. In DLBCL case 3, a further inversion event is shown within intron 3 of TP63. All genomic coordinates correspond to the GRCh37 (hg19) human genome assembly.

Figure 1

TBL1XR1/TP63 gene fusion observed using paired-end massively parallel RNA sequencing and the genomic fusion breakpoints. (A) Top panel: 77 paired read sequences are shown aligning on either side of the fusion point pairing TBL1XR1 and TP63 in one case of DLBCL. Genomic coordinates of exon boundaries are shown. Bottom panel: 17 split-reads are shown that cross the fusion junction, aligned to the merged sequence of the 2 genes. The histogram on the right of this panel shows the frequency of each split-read, a total of 55 split-reads. (B) An ideogram of chromosome 3 is shown, indicating the locations of TBL1XR1 and TP63 at 3q26.32 and 3q28, respectively. The gene fusions result from chromosomal rearrangement(s) that include an inversion event. Arrows above the genes indicate the direction of transcription. At the bottom, the genomic fusion breakpoints are shown for the 5 cases detected by RNA sequencing. Genomic coordinates are given for the fusion breakpoints and the exon boundaries. In DLBCL case 3, a further inversion event is shown within intron 3 of TP63. All genomic coordinates correspond to the GRCh37 (hg19) human genome assembly.

Close modal

TBL1XR1 and TP63 are located 12 Mb apart on the long arm of chromosome 3, flanking the BCL6 locus. To determine the incidence of this genetic rearrangement, FISH was performed on tissue microarrays (TMAs) comprising cores of pretreatment de novo DLBCL biopsies of 187 patients. The 30-patient overlap between the RNA-seq cohort and the TMA showed that RNA-seq and FISH had 100% concordance in detecting the TBL1XR1/TP63 fusion in these samples (Figure 2A). The TMA analysis also revealed 2 new fusion-containing cases that displayed breakapart of both TBL1XR1 and TP63 loci. Sanger sequencing of amplicons generated using RT-PCR from RNA extracted from the formalin-fixed, paraffin-embedded biopsy (Figure 2A) confirmed the presence of the gene fusion in both cases. In aggregate, the incidence of the TBL1XR1/TP63 fusion was 6 of 115 (5%) cases of germinal center B cell–like (GCB) DLBCL. The fusion appeared to be exclusive to GCB DLBCL with no TBL1XR1/TP63 fusions seen in 138 cases of non-GCB DLBCL (P = .008, Fisher exact; Figure 2B).

Figure 2

Incidence of the TBL1XR1/TP63 fusion in DLBCL. (A) FISH and Sanger sequencing of the fusion in 3 cases of DLBCL. Left hand column: results from a case known to be negative for the fusion on analysis of RNA-seq data. Middle column: results from a case where the fusion was detected in RNA-seq data. Right hand column: results of a patient who was not part of the RNA-seq cohort. Top panels: examples of representative cells from breakapart FISH assays on TMA. These images were produced at room temperature using an Olympus BX61 microscope with a UPlanFL N 40×/0.75 objective. The fluorochromes are Spectrum green and Spectrum orange. The images were acquired using a CV-M4+CL camera (JAI) and ARIOL software (Version 3.4; Genetix). Bottom panel: electropherograms from Sanger sequencing of an amplicon that spans the junction between TBL1XR1 and TP63 in the gene fusion transcript. (B) Incidence of the TBL1XR1/TP63 fusion in the total cohort and the cohorts where the 2 detection techniques for the fusion were applied. The nonadditive nature of the cohorts toward the total is the result of an overlap of 30 DLBCLs between the 2 cohorts, including 2 DLBCLs that harbor the fusion. Cell of origin designations are as follows: total GCB, 56 by gene expression profiling and 59 by the tally algorithm; total non-GCB, 38 ABC and 14 unclassifiable by gene expression profiling and 86 non-GCB by the tally algorithm; RNA-seq, GCB, ABC, and unclassifiable are all by gene expression profiling; TMA GCB, 23 by gene expression profiling and 59 by the tally algorithm; and TMA non-GCB, 12 ABC and 7 unclassifiable by gene expression and 86 non-GCB by the tally algorithm.

Figure 2

Incidence of the TBL1XR1/TP63 fusion in DLBCL. (A) FISH and Sanger sequencing of the fusion in 3 cases of DLBCL. Left hand column: results from a case known to be negative for the fusion on analysis of RNA-seq data. Middle column: results from a case where the fusion was detected in RNA-seq data. Right hand column: results of a patient who was not part of the RNA-seq cohort. Top panels: examples of representative cells from breakapart FISH assays on TMA. These images were produced at room temperature using an Olympus BX61 microscope with a UPlanFL N 40×/0.75 objective. The fluorochromes are Spectrum green and Spectrum orange. The images were acquired using a CV-M4+CL camera (JAI) and ARIOL software (Version 3.4; Genetix). Bottom panel: electropherograms from Sanger sequencing of an amplicon that spans the junction between TBL1XR1 and TP63 in the gene fusion transcript. (B) Incidence of the TBL1XR1/TP63 fusion in the total cohort and the cohorts where the 2 detection techniques for the fusion were applied. The nonadditive nature of the cohorts toward the total is the result of an overlap of 30 DLBCLs between the 2 cohorts, including 2 DLBCLs that harbor the fusion. Cell of origin designations are as follows: total GCB, 56 by gene expression profiling and 59 by the tally algorithm; total non-GCB, 38 ABC and 14 unclassifiable by gene expression profiling and 86 non-GCB by the tally algorithm; RNA-seq, GCB, ABC, and unclassifiable are all by gene expression profiling; TMA GCB, 23 by gene expression profiling and 59 by the tally algorithm; and TMA non-GCB, 12 ABC and 7 unclassifiable by gene expression and 86 non-GCB by the tally algorithm.

Close modal

The shared mutational landscape between GCB DLBCL and FL led us to determine the incidence of the fusion in FL. The TBL1XR1/TP63 fusion was predicted and confirmed using RNA-seq data in one of 13 FLs. FISH was performed on a TMA comprising cores of FL biopsies of 68 patients. The results, with no cases identified where both TBL1XR1 and TP63 loci displayed breakapart, indicate that the fusion is a rare event in FL.

With TBL1XR1 encoded from the negative DNA strand and TP63 from the positive DNA strand, the chromosomal rearrangement event(s) include an inversion that produces gene fusions encoded on the same strand (Figure 1B). In all cases, the identified junctions between the TBL1XR1/TP63 exons preserve the distal reading frame. Suggestive of functional significance, the TP63 portion of the gene fusion is conserved across all 7 cases, encoding exons 4 onwards. In 5 of the DLBCL cases, the 5′ junction point is the 3′ end of exon 7 of TBL1XR1, with exon 14 in the remaining DLBCL case and exon 4 in the FL case. In contrast, the predicted corollary, TP63/TBL1XR1, is disrupted in 3 of the 4 DLBCL cases examined. The mechanisms include a subsequent chromosomal translocation, deletion of the 3′ TBL1XR1 portion, and skipping of exon 8 of TBL1XR1 producing a frameshift resulting in a truncated protein.

The observation that the expression of the TBL1XR1/TP63 fusion was 5.2- ± 0.5-fold (mean ± SEM) higher than the wild-type TBL1XR1 mRNA, using quantitative RT-PCR on 3 of the DLBCL cases, suggests that there is a contextual change in the control of the TBL1XR1 promoter, which is driving the expression of this fusion.

The recurrent nature of the TBL1XR1/TP63 gene fusion, the conserved nature of the TP63 portion of the fusion, and its exclusive detection in the GCB subtype suggest a role in the pathogenesis of lymphomas harboring this event. Concomitant alterations of other genes known to be involved in the pathogenesis of DLBCL are given in supplemental Table 5. TP63 has 2 major sets of isoforms, distinguished at the N-terminus by the presence (TA-TP63) or lack (ΔN-TP63) of the TA domain, encoded by the first 3 exons.7  The TA-TP63 isoform has overlapping function with TP53 regarding induction of apoptosis in response to genotoxic stress.19  The ΔN-TP63 isoforms have a distinct function in the development and maintenance of stratified epithelial structures by contributing to self-renewal of basal epithelial cells.20-22  Lacking the TA domain, the TBL1XR1/TP63 protein may function similarly to ΔN-TP63, antagonizing the action of TP53, TA-TP63, and TA-TP73. We suspect that this may not only provide a proliferative advantage but also resistance to the genotoxic stress induced by chemotherapy.23  This is supported by the observation that 3 of the patients with DLBCL had primary refractory disease when treated with R-CHOP (supplemental Table 6). With the observation of deletion of the TBL1XR1 locus in DLBCL4  and CNS lymphoma,14  the disruption of TBL1XR1 function represents another possible mechanism of action. On activation, the E3 ligase activity of TBL1XR1 polyubiquitinates the NCoR/SMRT complex, thus targeting it for degradation, releasing transcriptional repression.24  Similarly, TBL1XR1 is involved in the polyubiquitination and degradation of the tumor oncoprotein BCL3.25 

The TBL1XR1/TP63 gene fusion is predicted to give rise to a unique chimeric protein, in contrast to the deregulated expression of wild-type BCL6, BCL2, and MYC that result from the other recurrent chromosomal rearrangements in DLBCL. Although characterization of the function of this fusion protein is awaited, it raises the possibility that this protein may be a novel target for therapeutic intervention.

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.

This work was supported in part by the National Cancer Institute Office of Cancer Genomics (contract HHSN261200800001E), the Terry Fox Foundation (grant 019001, Biology of Cancer: Insights from Genomic Analyses of Lymphoid Neoplasms), and Genome Canada/Genome British Columbia Grant Competition III (High Resolution Analysis of Follicular Lymphoma Genomes; J.M.C., R.D.G., and M.A.M.); and National Institutes of Health (grants P50CA130805-01, SPORE in Lymphoma, Tissue Resource Core PI Fisher; and 1U01CA114778 Molecular Signatures to Improve Diagnosis and Outcome in Lymphoma PI Chan). D.W.S. and K.L.T. were supported by the Terry Fox Foundation Strategic Health Research Training Program in Cancer Research at Canadian Institutes of Health Research (grant TGT-53912). R.D.M. is a Vanier Scholar (Canadian Institute of Health Research). A.J.M. is a Career Development Program Fellow of the Leukemia & Lymphoma Society. C.S. was supported by postdoctoral fellowships of the Cancer Research Society (Steven E. Drabin Fellowship) and the Michael Smith Foundation for Health Research. The laboratory work for this study was undertaken at the Genome Sciences Center, British Columbia Cancer Research Center and the Center for Translational and Applied Genomics, a program of the Provincial Health Services Authority Laboratories.

The content of this publication does not necessarily reflect the views of policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government.

National Institutes of Health

Contribution: D.W.S. designed and performed the research, analyzed and interpreted data, and wrote the paper; K.L.M., S.R., R.D.M., F.C.C., and R.S.L. analyzed the RNA-seq data; S.B.-N. performed experiments and analyzed data; G.W.S. and K.L.T. reviewed pathology; J.M.C. curated the lymphoma database, participated in the original design of the project, reviewed the manuscript, and provided editorial input; M.A.M. and A.J.M. participated in the design of the original project and oversaw data collection and analysis; C.S. designed experiments and wrote the paper; and R.D.G. participated in the design of the original project, designed experiments, and wrote the paper.

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

Correspondence: Randy D. Gascoyne, Department of Pathology and Experimental Therapeutics, British Columbia Cancer Agency & British Columbia Cancer Research Centre, 675 W 10th Ave, Rm 5-113, Vancouver, BC, V5Z 1L3, Canada; e-mail: rgascoyn@bccancer.bc.ca.

1
Kuppers
 
R
Klein
 
U
Hansmann
 
M-L
Rajewsky
 
K
Cellular origin of human B-cell lymphomas.
N Engl J Med
1999
, vol. 
341
 
20
(pg. 
1520
-
1529
)
2
Swerdlow
 
SH
Compo
 
E
Harris
 
NL
, et al. 
World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
4th Ed
Lyon, France
IARC Press
3
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
)
4
Pasqualucci
 
L
Trifonov
 
V
Fabbri
 
G
, et al. 
Analysis of the coding genome of diffuse large B-cell lymphoma.
Nat Genet
2011
, vol. 
43
 
9
(pg. 
830
-
837
)
5
Lohr
 
JG
Stojanov
 
P
Lawrence
 
MS
, et al. 
Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing.
Proc Natl Acad Sci U S A
2012
, vol. 
109
 
10
(pg. 
3879
-
3884
)
6
Steidl
 
C
Shah
 
SP
Woolcock
 
BW
, et al. 
MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers.
Nature
2011
, vol. 
471
 
7338
(pg. 
377
-
381
)
7
Yang
 
A
Kaghad
 
M
Wang
 
Y
, et al. 
p63, a p53 homolog at 3q27-29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities.
Mol Cell
1998
, vol. 
2
 
3
(pg. 
305
-
316
)
8
Hagiwara
 
K
McMenamin
 
MG
Miura
 
K
Harris
 
CC
Mutational analysis of the p63/p73L/p51/p40/CUSP/KET gene in human cancer cell lines using intronic primers.
Cancer Res
1999
, vol. 
59
 
17
(pg. 
4165
-
4169
)
9
Stransky
 
N
Egloff
 
AM
Tward
 
AD
, et al. 
The mutational landscape of head and neck squamous cell carcinoma.
Science
2011
, vol. 
333
 
6046
(pg. 
1157
-
1160
)
10
Yamaguchi
 
H
Inokuchi
 
K
Sakuma
 
Y
Dan
 
K
Mutation of the p51/p63 gene is associated with blastic crisis in chonic myelogenous leukemia.
Leukemia
2001
, vol. 
15
 
11
(pg. 
1729
-
1734
)
11
Yamaguchi
 
K
Wu
 
L
Caballero
 
OL
, et al. 
Frequent gain of the p40/p51/p63 gene locus in primary head and neck squamous cell carcinoma.
Int J Cancer
2000
, vol. 
86
 
5
(pg. 
684
-
689
)
12
Crook
 
T
Nicholls
 
JM
Brooks
 
L
O'Nions
 
J
Allday
 
MJ
High level expression of deltaN-p63: a mechanism for the inactivation of p53 in undifferentiated nasopharyngeal carcinoma (NPC)?
Oncogene
2000
, vol. 
19
 
30
(pg. 
3439
-
3444
)
13
Yoon
 
HG
Chan
 
DW
Huang
 
Z-Q
, et al. 
Purification and functional characterization of the human N-CoR complex: the roles of HDAC3, TBL1 and TBLR1.
EMBO J
2003
, vol. 
22
 
6
(pg. 
1336
-
1346
)
14
Braggio
 
E
McPhail
 
ER
Macon
 
W
, et al. 
Primary central nervous system lymphomas: a validation study of array-based comparative genomic hybridization in formalin-fixed paraffin-embedded tumor specimens.
Clin Cancer Res
2011
, vol. 
17
 
13
(pg. 
4245
-
4253
)
15
Robertson
 
G
Schein
 
J
Chiu
 
R
, et al. 
De novo assembly and analysis of RNA-seq data.
Nat Methods
2010
, vol. 
7
 
11
(pg. 
909
-
912
)
16
McPherson
 
A
Hormozdiari
 
F
Zayed
 
A
, et al. 
deFuse: an algorithm for gene fusion discovery in tumor RNA-Seq data.
PLoS Comput Biol
2011
, vol. 
7
 
5
pg. 
e1001138
 
17
Meyer
 
PN
Fu
 
K
Greiner
 
TC
, et al. 
Immunohistochemical methods for predicting cell of origin and survival in patients with diffuse large B-cell lymphoma treated with rituximab.
J Clin Oncol
2011
, vol. 
29
 
2
(pg. 
200
-
207
)
18
Chin
 
SF
A simple and reliable pretreatment protocol facilitates fluorescent in situ hybridisation on tissue microarrays of paraffin wax embedded tumour samples.
Mol Pathol
2003
, vol. 
56
 
5
(pg. 
275
-
279
)
19
Dohn
 
M
Zhang
 
S
Chen
 
X
p63alpha and ΔNp63alpha can induce cell cycle arrest and apoptosis and differentially regulate p53 target genes.
Oncogene
2001
, vol. 
20
 
25
(pg. 
3193
-
3205
)
20
Yang
 
A
Schweitzer
 
R
Sun
 
D
, et al. 
p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development.
Nature
1999
, vol. 
398
 
6729
(pg. 
714
-
718
)
21
Mills
 
AA
Zheng
 
B
Wang
 
XJ
Vogel
 
H
Roop
 
DR
Bradley
 
A
p63 is a p53 homologue required for limb and epidermal morphogenesis.
Nature
1999
, vol. 
398
 
6729
(pg. 
708
-
713
)
22
Senoo
 
M
Pinto
 
F
Crum
 
CP
McKeon
 
F
p63 Is essential for the proliferative potential of stem cells in stratified epithelia.
Cell
2007
, vol. 
129
 
3
(pg. 
523
-
536
)
23
Mundt
 
HM
Stremmel
 
W
Melino
 
G
Krammer
 
PH
Schilling
 
T
Müller
 
M
Dominant negative (DeltaN) p63alpha induces drug resistance in hepatocellular carcinoma by interference with apoptosis signaling pathways.
Biochem Biophys Res Commun
2010
, vol. 
396
 
2
(pg. 
335
-
341
)
24
Perissi
 
V
Scafoglio
 
C
Zhang
 
J
, et al. 
TBL1 and TBLR1 phosphorylation on regulated gene promoters overcomes dual CtBP and NCoR/SMRT transcriptional repression checkpoints.
Mol Cell
2008
, vol. 
29
 
6
(pg. 
755
-
766
)
25
Keutgens
 
A
Shostak
 
K
Close
 
P
, et al. 
The repressing function of the oncoprotein BCL-3 requires CtBP, while its polyubiquitination and degradation involve the E3 ligase TBLR1.
Mol Cell Biol
2010
, vol. 
30
 
16
(pg. 
4006
-
4021
)

Supplemental data

Sign in via your Institution