Published Ahead of Print on January 27, 2016, as doi:10.3324/haematol.2015.139253. Copyright 2016 Ferrata Storti Foundation.
Frequent CTLA4-CD28 gene fusion in diverse types of T cell lymphoma by Hae Yong Yoo, Pora Kim, Won Seog Kim, Seung Ho Lee, Sangok Kim, So Young Kang, Hye Yoon Jang, Jong-Eun Lee, Jaesang Kim, Seok Jin Kim, Young Hyeh Ko, and Sanghyuk Lee Haematologica 2016 [Epub ahead of print] Citation: Yoo HY, Kim P, Kim WS, Lee SH, Kim S, Kang SY, Jang HY, Lee JE, Kim J, Kim SJ, Ko YH, and Lee S. Frequent CTLA4-CD28 gene fusion in diverse types of T cell lymphoma. Haematologica. 2016; 101:xxx doi:10.3324/haematol.2015.139253 Publisher's Disclaimer. E-publishing ahead of print is increasingly important for the rapid dissemination of science. Haematologica is, therefore, E-publishing PDF files of an early version of manuscripts that have completed a regular peer review and have been accepted for publication. E-publishing of this PDF file has been approved by the authors. After having E-published Ahead of Print, manuscripts will then undergo technical and English editing, typesetting, proof correction and be presented for the authors' final approval; the final version of the manuscript will then appear in print on a regular issue of the journal. All legal disclaimers that apply to the journal also pertain to this production process.
Frequent CTLA4-CD28 gene fusion in diverse types of T cell lymphoma Hae Yong Yoo1,2*, Pora Kim3*, Won Seog Kim2,4*, Seung Ho Lee1*, Sangok Kim3,5*, So Young Kang6, Hye Yoon Jang7, Jong-Eun Lee7, Jaesang Kim8, Seok Jin Kim2,4, Young Hyeh Ko2,6†, and Sanghyuk Lee3,5,8† 1
Department of Health Sciences and Technology, Samsung Advanced Institute for Health
Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea 2
Samsung Biomedical Research Institute, Research Institute for Future Medicine, Samsung
Medical Center, Seoul 06351, Korea 3
Ewha Research Center for Systems Biology (ERCSB), Ewha Womans University, Seoul
03760, Korea 4
Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center,
Sungkyunkwan University School of Medicine, Seoul 06351, Korea 5
Department of Bio-Information Science, Ewha Womans University, Seoul 03760, Korea
6
Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of
Medicine, Seoul 06351, Korea 7
DNA Link Inc., Seoul 03759, Korea
8
Department of Life Science, Ewha Womans University, Seoul 06351, Korea
*These authors contributed equally to this work. †
These authors are co-corresponding authors of this article.
Running title: CTLA4-CD28 gene fusion in TCL
Corresponding Authors: Young Hyeh Ko, Department of pathology, Samsung medical Center, Sungkyunkwan University School of Medicine, and Samsung Biomedical Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea, Phone: +82-23140-2762 Fax: +82-2-3410-0025 E-mail:
[email protected] Sanghyuk Lee, Ewha Research Center for Systems Biology (ERCSB), Ewha Womans University, Seoul 03760, Korea, Phone: +82-2-3277-2888
[email protected]
Text word count: 2911 Abstract word count: 201 Number of figures: 4 Number of tables: 0 Number of references: 27 Supplementary file: 1
Fax: +82-2-3277-6809
E-mail:
ABSTRACT CTLA4 and CD28 are co-regulatory receptors of opposite roles in T cell signaling. We identified a fusion between the two genes from partial gene duplication in a case of angioimmunoblastic T cell lymphoma by RNA sequencing. The fusion gene consists of the extracellular domain of CTLA4 and the cytoplasmic region of CD28, likely capable of transforming inhibitory signals into stimulatory signals for T cell activation. Ectopic expression of fusion transcript in Jurkat and H9 cells resulted in enhanced proliferation and AKT and ERK phosphorylation, indicating activation of downstream oncogenic pathways. To estimate the frequency of this gene fusion in mature T cell lymphomas, we examined 115 T cell lymphoma samples of diverse subtypes using RT-PCR and Sanger sequencing. We identified the fusion in 26 of 45 angioimmunoblastic T-cell lymphomas (58%), 9 of 39 peripheral T-cell lymphomas, not otherwise specified (23%), and 9 of 31 extranodal NK/T cell lymphomas (29%). We further investigated the mutation status of 70 lymphomaassociated genes using ultra-deep targeted resequencing for 74 mature T cell lymphoma samples. The mutational landscape we obtained suggests that T cell lymphoma results from diverse combinations of multiple-gene mutations. The CTLA4-CD28 gene fusion is likely a major contributor to T cell lymphoma pathogenesis and represents a potential target for antiCTLA4 cancer immunotherapy.
INTRODUCTION Peripheral T cell lymphoma is a malignant neoplasm of mature T cells. Recent genomic studies have identified highly recurrent somatic mutations in TET2, DNMT3A, IDH2, and RHOA in diverse subtypes of mature T cell lymphoma (TCL)
1-5
. However, their roles in the
regulation of T cell signaling and oncogenesis have yet to be elucidated. Furthermore, none of the mutant genes has been clearly demonstrated as a dominant oncogenic driver. Therefore, identifying additional driver mutations and dissecting the interplay with T cell signaling components is still necessary. CTLA4 and CD28, members of the immunoglobulin superfamily that are expressed on the surfaces of T cells, are regulatory co-receptors of T cell signaling. They play critical and opposite roles in maintaining balanced T cell signaling and, thus, the proper level of immune activation
6,7
. Perturbing this balance can result in a number of undesirable consequences
such as autoimmunity, transplant rejection, or even malignant TCL 6. Accordingly, controlling T cell signaling through these two co-receptors has been a key strategy for recent cancer immunotherapies including anti-CTLA4 antibody therapy
8, 9
or chimeric antigen receptor T
cell therapy utilizing the intracellular signaling domain of CD28
10
. In this study, we
identified a fusion between CTLA4 and CD28 in a case of angioimmunoblastic TCL by whole transcriptome sequencing and analyzed the frequency of gene fusion in 117 cases of TCLs. Functional study of fusion gene indicates fusion between CTLA4 and CD28 results in activation of downstream oncogenic pathways. Mutational status of 70 lymphoma-associated genes using ultra-deep targeted resequencing for 74 mature TCL tumor samples were analyzed.
METHODS
Sample description Clinical information for the patients is summarized in Supplementary Table S2. For Sanger sequencing using 115 TCL tumor samples and targeted deep sequencing using 74 TCL tumor samples, formalin-fixed, paraffin-embedded (FFPE) tumor tissues were utilized. QIAamp DNA Mini Kit (Qiagen) and the RNeasy Mini Kit (Qiagen) were respectively used for DNA and RNA extractions. All patient samples were obtained with informed consent in Samsung Medical Center, Seoul, Korea, and the study was approved by the Institutional Review Board in accordance with the Declaration of Helsinki. Detection of CTLA4-CD28 mutation For detection of CTLA4-CD28 fusion transcript, we carried out reverse transcription (RT) with an random hexamers and total RNA isolated from FFPE samples followed by PCR amplification with the following oligonucleotide primers; RT_F1, RT_R1, RT_F2, RT_R2 and RT_F3. For characterization of genomic rearrangement, the CTLA4-CD28 fusion gene was amplified by PCR using genomic DNA of the clinical specimen as the template. Amplification was performed with Herculase 2 Fusion DNA Polymerase (Agilent Technologies) and the primers; Fusion_F1, Fusion_R1, Fusion_F2 and Fusion_R2. The resulting PCR products were analyzed by agarose gel electrophoresis and sequenced using two independent primer pairs. Cell proliferation and cytokine assays Jurkat (human T cell acute lymphoblastic leukemia) and H9 (human cutaneous T lymphocyte lymphoma) cells transfected with a construct expressing the CTLA4-CD28 fusion protein. Cells expressing CTLA4 and CTLA4-CD28 fusion were seeded in 96-well plates in triplicates at a density of 5×103 cells/well in 100
μl
of RPMI-1640 medium
containing 10% FBS and antibiotics. For stimulation, a 96-well plate was coated with 5 μg/ml
goat anti-mouse IgG (Ab frontier) or with 2 μg/ml anti-CD3 (HIT3a, BD pharmingen) or with the combination of 2 μg/ml anti-CD3 (HIT3a, BD pharmingen), 2 μg/ml anti-CD28 (CD28.2 BD pharmingen) or 5 μg/ml anti-CTLA4 (BNI3, BD phamingen) for overnight or 2 hrs in 37°C. After 48 hrs, cell proliferation was evaluated using Cell Counting Kit-8 (Dojindo), according to the manufacturer's instructions, and the absorbance value for each well was measured at 450 nm using a microplate reader (Spectra Max 180, Molecular Devices). Each experiment was repeated three times. For cytokine assay, cells were stimulated with 15 ng/ml PMA and 290 ng/ml Ionomycin (eBioscience) for 3 hrs. Cells were plated on 24-well plates coated with 5 μg/ml goat anti-mouse IgG (Ab frontier) or with 2
μg/ml anti-CD3
μg/ml anti-CD3 (HIT3a, BD pharmingen), 2 μg/ml anti-CD28 (CD28.2 BD pharmingen) or 5 μg/ml anti(HIT3a, BD pharmingen) or with the combination of 2
CTLA4 (BNI3, BD phamingen). After 24 hrs, the supernatants were examined using Human IL-2 ELISA kits (Thermo scientific) according to the manufacturer's instructions. Each experiment was repeated three times. Selection of 70 frequently mutated genes in lymphoma and bioinformatic analysis for targeted sequencing We have identified 62 frequently mutated genes from 10 genomics studies on the B and T cell lymphoma
1, 3, 5, 11-18
. Then we added three p53-related genes and five JAK-STAT
signaling genes for targeted deep sequencing. The list of 70 target genes is provided in Supplementary Table S1. We further compiled somatic mutations in the target genes from the original references which collectively yielded 832 mutations in 62 genes (Supplementary File S1). Targeted sequencing was performed using 74 TCLs. Bioinformatics analysis of targeted deep sequencing data is described in supplementary method.
RESULTS Identification and validation of the CTLA4-CD28 fusion gene The fusion transcript was initially predicted from analyses of RNA-Seq data from previously described TCL patients
5
using the FusionScan program. Multiple reads that
mapped to exon 3 of the CTLA4 gene in tandem with exon 4 of the CD28 gene were identified (Figure 1). The depth profile of the RNA-Seq data, reflecting the expression level of each exon, shows abrupt changes at the break points in both genes, which is consistent with the presence of fusion transcripts (Figure 1B). We verified the fusion via RT-PCR using two different primer sets in fusion-positive patient samples and seven TCL cell lines (Figure 2A ; Supplementary Figure S1). CTLA4-CD28 fusion cDNA was successfully amplified from the patient samples and two T lymphoblastoid cell lines, CEMC1-15 and CEMC7-14. Subsequent Sanger sequencing confirmed that the PCR products contained the fusion transcripts between exon 3 of the CTLA4 and exon 4 of the CD28 genes. To estimate the frequency of this gene fusion in TCL patients, we examined 115 TCL patient samples of diverse subtypes using RT-PCR and Sanger sequencing (Supplementary Table S2). The results indicated that CTLA4-CD28 fusion occurs in the all tested subtypes of TCL with an overall frequency of 38%. The fusion was observed more frequently in TCL of follicular helper T cell phenotype (TFH) than non-TFH TCL. The fusion was identified in 26 of 45 angioimmunoblastic TCL (AITL) patients (58%), 9 of 39 peripheral TCL not otherwise specified (PTCL-NOS) patients (23%), and 9 of 31 NK/T cell lymphoma patients (29%) (Supplementary Table S2; Supplementary Figure S2). Among PTCL-NOS, 5 of 9 PTCL-NOS with TFH phenotype (56%) and 5 of 16 non-TFH PTCL-NOS (31%) showed the CTLA4CD28 fusion. The CTLA4-CD28 fusion was not observed in blood samples of 50 healthy individuals.
Functional analyses of the CTLA4-CD28 fusion gene Notably, the predicted protein generated from this fusion gene features the extracellular and transmembrane domains of CTLA4 and the cytosolic signaling domain of CD28 (Figure 1A). A possible outcome is inappropriate activation of T cell signaling. Therefore, we proceeded to analyze the effect of the CTLA4-CD28 fusion on cell proliferation and cytokine production in T cells. Jurkat (human T cell acute lymphoblastic leukemia) and H9 (human cutaneous T lymphocyte lymphoma) cells transfected with a construct expressing the CTLA4-CD28 fusion protein showed a proliferation rate approximately 30% higher than cells transfected with only the vector or the wild-type CTLA4 expression construct after stimulation with anti-CTLA4 antibody (Figure 2B; Supplementary Figure S3A). The surface expression levels of CTLA4 and the CTLA4-CD28 fusion were comparable in Jurkat cell line (Supplementary Figure S4). Also, interleukin 2 (IL-2), the definitive marker of T cell activation, was produced at a six fold higher level (Figure 2C; Supplementary Figure S3B). These results indicate that the CTLA4-CD28 fusion protein likely mediates activating signals upon T cell stimulation. Next, we examined the phosphorylation of AKT and ERK1/2, which represents the activation of two critical pathways downstream of T cell receptor signaling. As expected, CD28-mediated co-stimulation of T cells led to increased AKT and ERK phosphorylation (Figure 2D). Importantly, cells expressing the CTLA4-CD28 fusion also showed increased AKT and ERK phosphorylation relative to cells expressing wild-type CTLA4 upon stimulation with an anti-CTLA4 antibody. These results strongly suggest that the CTLA4CD28 fusion could lead to constitutive T cell activation by converting inhibitory signals into activating signals. Of note, Shin et al. developed the CTLA4-CD28 chimera for adoptive T
cell therapy of cancer, and studied its biological roles and therapeutic efficacy using mouse T cells
19
. Importantly, they also demonstrated that the fusion gene delivered activating rather
than inhibitory T cell signal.
Genomic structure of the CTLA4-CD28 fusion gene Two genes are located in tandem 129 kbp apart on chromosome 2, with CD28 preceding CTLA4 on the same strand. The gene order is reversed in the CTLA4-CD28 fusion case, raising a strong possibility that partial duplications have occurred (Figure 3A). Extrachromosomal amplification by episome formation, which was observed for the NUP214-ABL1 fusion in T-cell acute lymphoblastic leukemia20, could not be ruled out in all cases, but was shown not to have occurred in multiple cases examined by fluorescence in situ hybridization (FISH) experiment (Supplementary Figure S5). Consistent with our hypothesis, quantitative PCR analyses of genomic DNA demonstrated that the copy number gains for portions of the CD28 and CTLA4 genes represented in the fusion were significantly higher in the fusion-positive patients than those in the fusion-negative lymphoma patients (Figure 3B; Supplementary Figure S6). Next, we mapped the exact positions of the break points in the genomic DNA of fusionpositive patients. For the fusion-positive patient shown in Figs. 1 and 2, we amplified a 2.5kb genomic DNA fragment (Figure 3D). Subsequent Sanger sequencing revealed that the fragment contained 696 bp of CTLA4 intron 3, 1457 bp of CD28 intron 3, and 47 bp of intervening sequence from a LINE retrotransposon element that is normally located 104 kb downstream of the CTLA4 gene on chromosome 2 (Figure 3E; Supplementary Figure S7). We characterized another patient in whose genome 426 bp of CTLA4 intron 3 was joined to 3185 bp of CD28 intron 3. Surprisingly, the most frequent arrangement was direct fusion of two
exons with no intron sequences included. We observed six other patients and two cell lines with such fusions. Despite the diversity in their genomic structures, all the genome arrangements we observed in patient samples would generate identical fusion transcripts and proteins. We also analyzed the mRNA expression levels of the CD28, CTLA4, and CTLA4-CD28 fusion genes in AITL patient samples using quantitative RT-PCR (Figure 3C). Fusion mRNA was expressed only in fusion-positive patients, and the expression level was comparable to that of CTLA4. CD28 and CTLA4 expression levels were approximately 3 and 7 times higher in fusion-positive patients, respectively, which suggests the presence of a feed-forward circuit that amplifies the signaling from the CTLA4-CD28 fusion receptor. Stronger binding affinity to CD80 and CD86 ligands of CTLA4 compared to CD28 may have roles in amplifying signals in the CTLA4-CD28 fusion receptor 6. In sum, patients with the CTLA4-CD28 fusion have not only copy number gain at the genomic level but also show elevated CD28 and CTLA4 expression that is consistent with abnormal activation of the T cell population.
Mutational landscape of lymphoma-related genes from targeted deep sequencing Several somatic mutations have already been implicated as driver mutations in TCL, including the point mutations IDH2 R172K, DNMT3A R882H, RHOA G17V, and CD28 T195P and loss-of-function (stop-gain or frameshift) mutations in the TET2 gene
3-5
.
Determining the functional relationship between these mutations and the CTLA4-CD28 fusion would be of the utmost importance to understanding the molecular mechanisms underlying TCL. We selected 70 genes that had been reported to be frequently mutated in various types of T and B cell lymphoma (Supplementary Table S1)
1, 3-5, 11-17
. Targeted deep
sequencing data for all exons of the 70 genes were produced with an average sequencing
depth of 1,204X and 98.9% of target coverage using 2,965 primer pairs. The ultra-high depth of the sequencing is expected to reveal variants of low frequency but with important functional roles. In total, 74 tumor tissues from 29 AITL, 15 PTCL-NOS, 26 extranodal NK/T cell lymphoma (ENKL), 2 enteropathy-associated T cell lymphoma (EATL), and 2 anaplastic large cell lymphoma (ALCL) patients were examined. The sequencing data were analyzed using our own computational pipeline, which was designed to identify somatic mutations in tumor cells without the normal control (Supplementary Figure S8). We identified 11 missense mutations in 10 different genes and 24 TET2 loss-of-function mutations, including 8 novel mutations (Figure 4; Supplementary Figure S8). Loss-of-function mutations in the TET2 gene occurred most frequently in all subtypes (47 patients, 64%). Excluding TET2 mutations with low allele frequency (< 5%), which were observed mostly for R544X and R1404X (Supplementary Figure S9), the TET2 mutation rate was only 30% (22 patients), with significant concentration in the AITL subtype (15 patients, P = 0.008). Apparently, many mutations were observed at low frequencies below 20%, demonstrating the power of ultra-deep sequencing. These low frequency mutations are likely from the heterogeneity of the tumor cells as well as presence of normal or stromal cells. The RHOA mutation was mostly observed in the AITL subtype (21 of 29 AITL patients; 72%), with only two mutations observed in the 15 PTCL-NOS patients (13%). We identified 8 low frequency mutations (allele frequency < 5%) in addition to 12 (41% of AITL patients) high frequency mutations, which agrees well with previous reports on AITL based on Sanger sequencing
3-5
. Among other recurrent mutations, IDH2 R172K and DNMT3A R882H
mutations were found in 7 and 5 patients, respectively. These latter two mutations have been described as ageing-related initiating mutations that are associated with the clonal expansion
of prelymphoma stem cells and can be detected in the blood of elderly individuals without apparent hematologic malignancies 21-23. The frequency of the CTLA4-CD28 fusion was 30% in this targeted deep-sequencing analysis (22 out of 74 patients), appearing in all subtypes tested. Sixteen out of 22 fusionpositive patients contained additional mutations. In 11 AITL patients with the CTLA4-CD28 fusion, 9 (82%) patients and 10 (91%) patients also harbored TET2 mutation and RHOA mutation respectively. CTLA4-CD28 fusion was not found in 4 patients with CD28 T195P mutation which also led to up-regulation of TCR signaling
24
, indicating the relevance of
CD28 in the process of lymphomagenesis. In ENKL, most of the fusion cases were devoid of additional mutation. In fact, a substantially high proportion of cases were devoid of any mutations among tested genes, and even TET2 mutation-positive cases mostly contained no other mutations. It is in this regard that a novel recurrent TCF3 G302S is notable, especially for the ENKL subtype. The transcription factor TCF3 (E2A) is required for B and T lymphocyte development, and the significance of mutations in TCF3 and its negative regulator ID3 has recently been highlighted in Burkitt lymphoma
14, 15, 17
. Genomic examination with additional ENKL
patients should be carried out to substantiate TCF3 G302S mutation as a marker and a potential driver of ENKL subtype.
DISCUSSION Here, we report that the CTLA4-CD28 fusion gene is a novel, high-frequency mutation for diverse types of TCL. Two other groups have recently reported the identification of the CTLA4-CD28 fusion gene in Sézary syndrome, an aggressive rare variant of cutaneous T cell
lymphoma 25, 26. That this mutation is not limited to Sézary syndrome but is found in a broad range of TCL types with the overall frequency of 30% and typically in combination with other mutations should be of significance. We also provide the overall mutational landscape for TCL which indicates that the CTLA4-CD28 fusion represents one of recurrent genetic events for full blown neoplastic transformation. Targeted deep sequencing analyses for 70 genes implicated in TCL showed that a large majority of patients contained more than one mutated gene. Although not all of the mutations have been mechanistically demonstrated to be oncogenic, we suggest that multiple mutational events, including those described in this study, are required for the full development of TCL. It has been proposed and partly demonstrated that TCLs and myeloid leukemia feature the age-related accumulation of premalignant mutations in DNMT3A, TET2, JAK2, and GNAS which are associated with subsequent clonal hematopoietic expansion
21-23
.
Similarly in B cell lymphoma, it has been shown that circulating B-cells bearing Bcl-2 translocations do not cause follicular lymphoma per se but can evolve into overt follicular lymphoma with additional mutations 27. Our data indicate that CTLA4-CD28 fusion disrupts cellular homeostasis via inappropriate activation of T cell signaling. Given that such dysregulation likely lies at the core of oncogenesis, it is possible that the CTLA4-CD28 fusion provides a target for potential immunotherapy. In fact, Sekulic et al. reported an n-of-1 trial for a female patient who had suffered from Sézary syndrome for 8 years26. Administration of the CTLA4-blocking antibody ipilimumab 8 showed dramatic initial responses for first two months, but the disease subsequently progressed rapidly resulting in death 3 months after the last dose. The tragic outcome notwithstanding, the result showed that an immunotherapy targeting the fusion gene is viable in principle, and with an improvement in dosage and timing, the response may
become more durable. Targeted therapy and immunotherapy as parts of combination therapy regimen is an emerging paradigm of cancer treatment. Thus, the identification of frequent CTLA4-CD28 fusion gene will provide a new therapeutic opportunity for relevant TCL patients, and elucidation of the exact mechanism by which the CTLA4-CD28 fusion interacts with other mutations should provide further insights into the molecular nature of TCL development, as well as new strategies for curbing this disease.
Acknowledgments This work was supported by grants from the Technology Innovation Program of the Ministry of Trade, Industry and Energy, Republic of Korea (10050154 to S.L.), the National Research Foundation of Korea (NRF-2014M3C9A3065221, NRF-2012M3A9D1054744 to S.L., NRF2015K1A4A3047851 to S.L. and J.K.), the Samsung grant (SM01132671 and SMO1150911 to Y.H.K.), and a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI14C3414 to Y.H.K. and H.Y.Y., HI14C2331 to H.Y.Y.). This study was also supported by the Ministry of Health & Welfare, Republic of Korea (A120262 to JK).
Authorship Contributions Conception and design: S. Lee, Y.H. Ko, Development of methodology: H.Y. Yoo, S. Lee
Acquisition of data (performed experiment, acquired samples and clinical data, provided facilities, etc.): W.S. Kim, Y.H. Ko, S.H. Lee, S.Y. Kang, H.Y. Jang, J.E. Lee Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): P. Kim, S. Kim Writing, review, and/or revision of the manuscript: H.Y. Yoo, J. Kim, S. Lee, Y.H. Ko Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): W.S. Kim, S.J. Kim Study supervision: S. Lee, Y.H. Ko
Disclosure of Potential Conflicts of Interest None.
REFERENCES 1.
Cairns RA, Iqbal J, Lemonnier F, et al. IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma. Blood. 2012;119(8):1901-1903.
2.
Lemonnier F, Couronné L, Parrens M, et al. Recurrent TET2 mutations in peripheral Tcell lymphomas correlate with TFH-like features and adverse clinical parameters. Blood. 2012;120(7):1466-1469.
3.
Palomero T, Couronné L, Khiabanian H, et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nat Genet.
2014;46(2):166-170. 4.
Sakata-Yanagimoto M, Enami T, Yoshida K, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46(2):171-175.
5.
Yoo HY, Sung MK, Lee SH, et al. A recurrent inactivating mutation in RHOA GTPase in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46(4):371-375.
6.
Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev. 2009;229(1):12-26.
7.
Kuehn HS, Ouyang W, Lo B, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science. 2014;345(6204):162316-162327.
8
Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734-1736.
9.
Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723.
10. Johnson LA, Morgan RA, Dudley ME, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114(3):535-546. 11. Gunawardana J, Gunawardana J, Chan FC, et al. Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma. Nat Genet. 2014;46(4):329-335. 12. 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. USA 2012;109(10):3879-3884. 13. Okosun J, Bödör C, Wang J, et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular
lymphoma. Nat Genet. 2014;46(2):176-181. 14. Richter J, Schlesner M, Hoffmann S, et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nat Genet. 2012;44(12):1316-1320. 15. Schmitz R, Young RM, Ceribelli M, et al. Burkitt lymphoma pathogenesis and therapeutic
targets
from
structural
and
functional
genomics.
Nature.
2012;490(7418):116-120. 16. Zhang J, Grubor V, Love CL, et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc Natl Acad Sci USA. 2013;110(4):1398-1403. 17. Love C, Sun Z, Jima D, et al. The genetic landscape of mutations in Burkitt lymphoma. Nat Genet. 2012;44(12):1321-1325. 18. Couronné L, Bastard C, Bernard OA. TET2 and DNMT3A mutations in human T-cell lymphoma. N Engl J Med. 2012;366(1):95-96. 19 Shin JH, Park HB, Oh YM, et al. Positive conversion of negative signaling of CTLA4 potentiates antitumor efficacy of adoptive T-cell therapy in murine tumor models. Blood. 2012;119(24):5678-5687. 20 Graux C, Cools J, Melotte C, et al. Fusion of NUP214 to ABL1 on amplified episomes in T-cell acute lymphoblastic leukemia. Nat Genet. 2004;36(10):1084-1089. 21. Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477-2487. 22. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488-2498. 23. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic
expansion and malignancies. Nat Med. 2014;20(12):1472-1478. 24. Lee SH, Kim JS, Kim J, et al. A highly recurrent novel missense mutation in CD28 among angioimmunoblastic T-cell lymphoma patients. Haematologica. 2015;100(12):e505-507. 25. Sekulic A, Liang WS, Tembe W, et al. Personalized treatment of Sezary syndrome by targeting a novel CTLA4:CD28 fusion. Mol Genet Genomic Med. 2015;3(2):130-136. 26. Ungewickell A, Bhaduri A, Rios E, et al. Genomic analysis of mycosis fungoides and Sezary syndrome identifies recurrent alterations in TNFR2. Nat Genet. 2015;47(9):10561060. 27. Correia C, Schneider PA, Dai H, et al. BCL2 mutations are associated with increased risk of
transformation
2015;125(4):658-667.
and
shortened
survival
in
follicular
lymphoma.
Blood.
FIGURE LEGENDS
Figure 1. Identification of the CTLA4-CD28 gene fusion. A, top, schematic diagram of the gene fusion; bottom, sequencing chromatogram. Numbers on the transcript indicate the nucleotide position of exons. B, alignment of RNA-Seq data from a fusion-positive patient. Read-depth plots indicate the depth coverage of aligned RNA-Seq reads. SP = signal peptide, TM = transmembrane region.
Figure 2. Validation and functional analyses of the CTLA4-CD28 fusion gene. A, validation of the CTLA4-CD28 fusion transcript by RT-PCR using samples
from patients and cell lines.
Arrows indicate the approximate positions of oligonucleotide primers on the CTLA4-CD28 fusion transcript. PCR products were amplified from patient 1, CEMC1-15 cells and CEMC7-14 cells and validated by Sanger sequencing. -actin was used as an internal control, and NTC indicates the no template control. Normal tonsil tissue and 293T cells were used as negative controls. B, Jurkat cells expressing the CTLA4-CD28 fusion show enhanced cell proliferation (*P < 0.05 and **P < 0.01 compared with cells expressing wild-type CTLA4) after co-stimulation with anti-CD3/CTLA4 antibodies. Each experiment was repeated three times with five replicates, and the data are expressed as the mean ± standard deviation. C, expression of the CTLA4-CD28 fusion enhanced interleukin 2 (IL-2) production after CTLA4 activation (**P < 0.01 compared with cells expressing wild-type CTLA4). Jurkat cells were stimulated with PMA/Ionomycin (P/I) without or with anti-CD3/CTLA4 antibodies to activate CTLA4. IL-2 measurement was carried out three independent times, and the data are expressed as the mean ± standard deviation. D, expression of the CTLA4-CD28 fusion enhanced phosphorylation of AKT and ERK1/2 after CTLA4 activation with an anti-CTLA4 antibody.
Figure 3. Genomic structure of the CTLA4-CD28 fusion gene. A, schematic diagram of the gene duplication producing the fusion gene. B, copy-number analysis of CD28 and CTLA4 genes in AITL patient samples using Q-PCR (*P = 0.009, **P = 0.001). Copy number changes, estimated relative to that in the peripheral blood cells from a normal individual, were shown in the box plot. The values are from two independent experiments. C, expression levels of CD28, CTLA4, and CTLA4-CD28 fusion transcripts in AITL patient samples using Q-PCR (*P = 0.001, **P = 0.0001, ***P = 0.002). The values are from three independent experiments. D, structural analysis of genomic loci for CTLA4-CD28 fusion patients. The PCR (2.5 kb) product amplified from genomic DNA of patient 1 was subsequently validated by Sanger sequencing. No product was amplified from the control normal tonsil cells and the no template control (NTC). E, schematic diagrams of the genomic structure of the CTLA4CD28 fusion from 8 patients and two cell lines. The arrows indicate the position of primers (Fusion F1 and Fusion R1).
Figure 4. Mutational landscape of driver genes from targeted resequencing. Each column represents an independent patient. Novel mutations are indicated with asterisks. TCL subtypes are color-coded above the main window, and bars on the right side show the relative proportions of the indicated mutation among TCL subtypes. The CTLA4-CD28 row indicates the presence of fusion gene based on RT-PCR and Sanger sequencing. TET2 mutations of all locations are shown in top rows, with the color of the highest allele frequency for patients with multiple TET2 mutations (position-wise mutation plot provided in Supplementary Fig. S9). The cumulative numbers of patients positive for each mutation are indicated on the right.
Supplementary Information for Frequent CTLA4-CD28 gene fusion in diverse types of T cell lymphoma Hae Yong Yoo1,2*, Pora Kim3*, Won Seog Kim2,4*, Seung Ho Lee1*, Sangok Kim3,5*, So Young Kang6, Hye Yoon Jang7, Jong-Eun Lee7, Jaesang Kim8, Seok Jin Kim2,4, Young Hyeh Ko2,6†, and Sanghyuk Lee3,5,8† 1
Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea 2
Samsung Biomedical Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea 3
Ewha Research Center for Systems Biology (ERCSB), Ewha Womans University, Seoul 03760, Korea 4
Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea 5
Department of Bio-Information Science, Ewha Womans University, Seoul 03760, Korea
6
Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea 7
DNA Link Inc., Seoul 03759, Korea
8
Department of Life Science, Ewha Womans University, Seoul 06351, Korea
*These authors contributed equally to this work. †
These authors are co-corresponding authors of this article.
Corresponding Authors: Young Hyeh Ko, Department of pathology, Samsung medical Center, Sungkyunkwan University School of Medicine, and Samsung Biomedical Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea, Phone: +82-2-3140-2762 Fax: +82-2-3410-0025 E-mail:
[email protected] Sanghyuk Lee, Ewha Research Center for Systems Biology (ERCSB), Ewha Womans 1
University, Seoul 03760, Korea, Phone: +82-2-3277-2888 Fax: +82-2-3277-6809 E-mail:
[email protected]
2
Table of Contents Supplementary Methods Quantitative real time PCR ............................................................................................... 4 Cell culture and transfection
........................................................................................... 4
Antibodies and immunoblotting
..................................................................................... 4
Library preparation and targeted deep sequencing
......................................................... 5
Bioinformatics analysis of targeted deep sequencing data. .............................................. 6
Supplementary Tables Supplementary Table S1. List of genes for targeted deep sequencing .............................. 8 Supplementary Table S2. Study subjects for sequencing and their clinical information.. 10 Supplementary Table S3. List of gene-specific PCR primers........................................... 13 Supplementary Table S4. Summary statistics of targeted deep sequencing ..................... 14
Supplementary Figures Supplementary Fig. S1. Validation of the CTLA4-CD28 fusion transcript ...................... 16 Supplementary Fig. S2. Validation of the CTLA4-CD28 fusion gene in patients ............ 17 Supplementary Fig. S3. Functional analyses of the CTLA4-CD28 fusion gene in H9 cell line ............................................................................................................................ 18 Supplementary Fig. S4. Surface expression levels of CTLA4 and the CTLA4-CD28 fusion ........................................................................................................................ 19 Supplementary Fig. S5. Fluorescence in situ hybridization analysis for the fusion ......... 20 Supplementary Fig. S6. Copy number analysis of CD28 and CTLA4 genes ................... 21 Supplementary Fig. S7. Sanger sequencing of genomic DNA for mapping exact fusion points ........................................................................................................................ 22 Supplementary Fig. S8. Computational pipeline for analyzing targeted deep sequencing data ........................................................................................................................... 23 Supplementary Fig. S9. Mutation profile of TET2 gene from targeted sequencing ......... 24
Supplementary Files Supplementary File S1. List of known mutations in lymphoma
3
Supplementary Methods Quantitative real-time PCR. For quantitative analysis of genomic DNA and mRNA in patient samples, 30 ng genomic DNA and 2 g mRNA were used, respectively. mRNA was reversetranscribed, using Super Script 2 (Invitrogen) with random primer. qRT-PCR was carried out with SYBR Green PCR master mix (Applied Biosystems) and gene specific primers listed (Supplementary Table 3) on ABI PRISM 7900HT. We compared the normalized the CT values using the housekeeping gene -actin.
Cell culture and transfection. The Jurkat E6.1 (human T cell acute lymphoblastic leukemia), H9 (human cutaneous T lymphocyte lymphoma) and HUT78 (human cutaneous T lymphocyte lymphoma) cell lines obtained from the American Type Culture Collection (ATCC) were maintained in RPMI-1640 medium containing 10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin and 250 ng/ml amphotericin B at 37 °C and 5% CO2. Cultured cells were regularly tested for mycoplasma infection using the MycoAlert mycoplasma detection kit (Lonza). To generate expression vector plasmids for CTLA4 and CTLA4-CD28 fusion, cDNA encoding CTLA4 and CD28 were amplified by PCR from appropriate sources and inserted into LZRSpMBN linker IRES EGFP vector. Plasmids expressing CTLA4 and CTLA4-CD28 fusion were transfected into cells using the Nucleofector I device (Amaxa) with Nucleofector solution V and program X-001 (Jurkat) and G-014 (H9). Typically, one million cells were transfected with 2 μg of plasmid. A GFP-expressing plasmid was used to measure transfection efficiency. Expression of CTLA4 and CTLA4-CD28 fusion proteins was analyzed by flow cytometry. Transfected cells were stained with PE-Cy7 CD28 (CD28.2, BD pharmingen) and PE-Cy5 CTLA4 (BNI3, BD pharmingen) for 30 min and analyzed using FACS Verse (BD bioscience).
Antibodies and immunoblotting. Antibodies used for protein blot were phospho-AKT(S473) (Cell Signaling Technology), AKT antibody (Cell Signaling Technology), phosphoERK(T202/Y204) antibody (Cell Signaling Technology), ERK antibody (Cell Signaling Technology), α-tubulin antibody (Santa Cruz Biotechnology). Horseradish peroxidase (HRP) conjugated secondary antibodies (Bio-Rad) were used to detect primary antibodies. Equal protein loading was assessed by immunoblotting for α-tubulin. For immunoblotting analysis of CTLA4 and CTLA4-CD28 fusion signaling, cells transfected with indicated plasmids were treated with 2 μg/ml mouse anti-human CD28 (BD Pharmingen), 2 μg/ml mouse anti-human CTLA4 (BD 4
Pharmingen) or normal 2 μg/ml rabbit IgG for 10 min on ice, followed by crosslinking with 5 μg/ml goat anti-mouse IgG or goat anti-rabbit IgG (Ab frontier) for 10 min on ice. Then cells were incubated at a 37°C for 1 hour and lysed. Equal amounts of cell lysates were subjected to SDS-PAGE, transferred and probed with antibody. Equal protein loading was assessed by immunoblotting for α-tubulin.
Library preparation and targeted deep sequencing. We produced the deep sequencing data for 70 target genes on Ion PI v2 chip following the Ion AmpliSeq protocol of Life Technologies. A set of primer pairs were designed by the Ion AmpliSeq Designer in order to cover the coding DNA sequences and adjacent intronic regions (5 bp) of target genes. We achieved the design coverage of 98.9% for the target region of 227 kbp with 2,965 amplicons. Genomic DNA was pre-treated with uracil-DNA glycosylase (UDG) that had been reported to reduce high number of artifactual single nucleotide changes in FFPE samples (Do H et al. Clinical Chemistry 2013). Sequencing libraries were prepared according to the manufacturer’s protocol. Briefly, 20 ng of UDG-treated FFPE DNAs were amplified with the Ion AmpliSeq Library kit 2.0 for each of 74 lymphoma patient cases. Primers were partially digested, and each sample was barcoded with the Ion Xpress Barcode Adapters kit. Adapter-ligated and barcoded libraries were purified with AMPure XP reagent (Beckman Coulter) and PCR-amplified for 5 cycles. Resulting products were quantified by Agilent 2100 BioAnalyzer and Agilent Bioanalyzer DNA High-Sensitivity LabChip (Agilent Technologies). We pooled 20 uniquely barcoded libraries at the equimolar concentration for multiplexed sequencing on a single Ion PI v2 chip. Emulsion PCR was performed for the pooled sample on Ion OneTouch system using Ion PI Template OT2 200 Kit v3. Template-positive ISPs were sequenced on Ion PI v2 chip using Ion PI Sequencing 200 Kit v3 in the Ion Proton System. Mapping of sequencing data to the human genome (see below) indicated that the depths of reading ranged from 728X to 2578X with the average depth of 1204X. Summary of amplicon sequencing is available in Supplementary Table 4. Library preparation and sequencing procedures were performed by DNA Link Inc. in Korea.
5
Bioinformatics analysis of targeted deep sequencing data. The overview of the computational pipeline for analyzing deep sequencing data is shown in Supplementary Fig. S8. Human genome assembly (GRCh37, hg19) of repeat-masked version was downloaded from the UCSC genome browser. The sequencing data were mapped to the human genome using the Torrent Mapping Alignment Program (TMAP ver. 3.4.1) with the option of ‘mapall -g 2 –a 0 –y stage1 map1’. Genome Analysis Toolkit (GATK ver. 1.6.7)19 was used for local realignment and base quality score recalibration. Next, we called SNPs and indels using the GATK HaplotypeCaller with the option of ‘-dontUseSoftClippedBases -stand_emit_cof 20 -stand_call_cof 20’. Finally, ANNOVAR (ver. 2013-05-20) was used for functional annotation of genetic variants with ljb23_all for hg19_refGene release 66 transcriptome model20. Mutation analysis by HaplotypeCaller yielded 64,062 variations including 9,183 exonic ones. Filtering out nonfunctional candidates (synonymous mutations and in-frame indels), we obtained 9,104 mutations of functional significance comprising 745 nonsynonymous point mutations, 155 stop gain mutations, 2 stop loss mutations, 8,202 frame-shift indels. The large number of frame-shift indels is presumably due to the well-known sequencing error for homopolymer repeats in Ion Proton sequencing. For known point mutations from the literature survey (Supplementary File S1), we simply checked their presence in the list of point mutations, and confirmed the presence of 8 missense mutations (RHOA_G17V, IDH2_R172K, DNMT3A_R882H, CD28_T195P, FYN_R176C, VAV1_E524D, RHOA_T19I, PLCG1_S345F) and 2 nonsense mutations in TET2 gene (TET2_R544X, TET2_R1404X). For novel mutations, we applied several stringent filters to reduce false positives. Firstly, SNPs with population diversity in the dbSNP database (ver. 143) were removed. We further filtered out mutations of germline origin using exome sequencing data for paired samples of tumor and matched normal samples from 5 AITL patients, 100 lung cancer patients, 50 gastric cancer patients in Korean population (in-house data). Finally, we confirmed the mutation by Sanger sequencing for cases when patient samples were available. We have thus identified 3 novel cases of point mutations (CD28_F51I, TCF3_G302S, and KMT2D_S4251F). Loss-of-function mutations in TET2 gene are prevalent in myeloid cancers and lymphoma. Thus, a careful analysis of indels leading to loss-of-function is essential, especially for the sequencing data with homopolymer problem. Through a careful evaluation, we determined reliable indels as (i) no.
with patient recurrency over 10% turned out to be false positives in most cases. Among 182 indels from the GATK HaplotypeCaller predictions on TET2 gene, we have obtained 7 indels satisfying 6
both conditions. In addition, 24 missense mutations were predicted in TET2 gene, 6 of which were filtered out as SNPs with population diversity and additional 11 candidates were nondamaging according to ANNOVAR annotation, leaving 7 missense mutations of functional consequences. Including 10 nonsense mutations, the number of loss-of-function mutations in TET2 gene was 24 (7 misssense mutations, 10 nonsense mutations, 7 indels), 19 of which had been previously reported in hematological tumors (Supplementary Fig. S9). Five mutations (E1151X, 1715-1717del, 1747-1751del, Y1148D, G1861V) were novel from our study, and Y1148D mutation was confirmed by Sanger sequencing. Deep sequencing data have been deposited in the NCBI Sequence Read Archive (SRA) database under accession ID SRP056397 and BioProject ID PRJNA278986.
7
Supplementary Table S1. List of genes for targeted deep sequencing Gene Symbol
Lymphoma Type*
Publication
First Author
Ref. No.
ACTB
DLBCL
PNAS 2012
Lohr JG
14
ANKRD11
TP53-related
ARID1A
DLBCL
PNAS 2013
Zhang J
18
ATM
PTCL
Nat. Genet. 2014
Palomero T
3
B2M
PTCL
Nat. Genet. 2014
Palomero T
3
BCL10
FL
Nat. Genet. 2014
Okosun J
15
BCL2
BL
Nature 2012
Schmitz R
17
BRAF
DLBCL
PNAS 2012
Lohr JG
14
BTG1
DLBCL
PNAS 2012
Lohr JG
14
CARD11
FL
Nat. Genet. 2014
Okosun J
15
CCND3
BL
Nature 2012
Schmitz R
17
CD28
AITL
Nat. Genet. 2014
Yoo HY
5
CD58
PTCL
Nat. Genet. 2014
Palomero T
3
CD79B
FL
Nat. Genet. 2014
Okosun J
15
CDKN2A
PTCL
Nat. Genet. 2014
Palomero T
3
CREBBP
FL
Nat. Genet. 2014
Okosun J
15
DNMT3A
TCL
NEJM 2012
Couronné L
26
EBF1
FL
Nat. Genet. 2014
Okosun J
15
EZH2
AITL
Nat. Genet. 2014
Yoo HY
5
FYN
PTCL
Nat. Genet. 2014
Palomero T
3
GNA13
BL
Nature 2012
Schmitz R
17
HIST1H1C
FL
Nat. Genet. 2014
Okosun J
15
HIST1H1E
FL
Nat. Genet. 2014
Okosun J
15
ID3
BL
Nature 2012
Schmitz R
17
IDH1
DLBCL
PNAS 2013
Zhang J
18
IDH2
AITL
BLOOD 2012
Cairns RA
1
IRF8
DLBCL
PNAS 2013
Zhang J
18
JAK2
JAK-STAT pathway
JAK3
JAK-STAT pathway
KLHL6
FL
Nat. Genet. 2014
Okosun J
15
KMT2C
AITL
Nat. Genet. 2014
Yoo HY
5
KMT2D
AITL
Nat. Genet. 2014
Yoo HY
5
KRAS
DLBCL
PNAS 2012
Lohr JG
14
LILRB1
AITL
Nat. Genet. 2014
Yoo HY
5
MEF2B
FL
Nat. Genet. 2014
Okosun J
15
MKI67
BL
Nature 2012
Schmitz R
17
8
MTOR
DLBCL
PNAS 2013
Zhang J
18
MUC2
AITL
Nat. Genet. 2014
Yoo HY
5
MYC
BL
Nature 2012
Schmitz R
17
MYD88
FL
Nat. Genet. 2014
Okosun J
15
NOTCH1
DLBCL
PNAS 2012
Lohr JG
14
P2RY8
DLBCL
PNAS 2012
Lohr JG
14
PCLO
DLBCL
PNAS 2012
Lohr JG
14
PIK3CD
DLBCL
PNAS 2013
Zhang J
18
PIK3R1
DLBCL
PNAS 2013
Zhang J
18
PIM1
DLBCL
PNAS 2012
Lohr JG
14
PLCG1
AITL
Nat. Genet. 2014
Yoo HY
5
POU2F2
DLBCL
PNAS 2013
Zhang J
18
PRKD2
PTCL
Nat. Genet. 2014
Palomero T
3
PTPN1
PMBCL
Nat. Genet. 2014
Gunawardana J
13
RHOA
AITL
Nat. Genet. 2014
Yoo HY
5
RHOT2
PTCL
Nat. Genet. 2014
Palomero T
3
SGK1
BL
Nature 2012
Schmitz R
17
SMARCAL1
PTCL
Nat. Genet. 2014
Palomero T
3
SMARCD1
AITL
Nat. Genet. 2014
Yoo HY
5
SOCS1
FL
Nat. Genet. 2014
Okosun J
15
STAT1
JAK-STAT pathway
STAT2
JAK-STAT pathway
STAT3
JAK-STAT pathway
STAT6
FL
Nat. Genet. 2014
Okosun J
15
TCF3
BL
Nature 2012
Schmitz R
17
TET2
TCL
NEJM 2012
Couronné L
26
TET3
PTCL
Nat. Genet. 2014
Palomero T
3
TNFAIP3
FL
Nat. Genet. 2014
Okosun J
15
TNFRSF14
FL
Nat. Genet. 2014
Okosun J
15
TP53
TP53-related
TP63
TP53-related
VAV1
AITL
Nat. Genet. 2014
Yoo HY
5
WIF1
DLBCL
PNAS 2013
Zhang J
18
WWOX
TP53-related
*AITL = angioimmunoblastic T cell lymphoma, BL = Burkitt lymphoma, DLBCL = diffuse large B cell lymphoma, FL = Follicular lymphoma, PMBCL = primary mediastinal large B cell lymphoma, PTCL = peripheral T cell lymphoma, TCL = T cell lymphoma
9
Supplementary Table S2. Study subjects for sequencing and their clinical information case no.
Diagnosis
Age
Gender
Tissue of origin
CD28-CTLA4 fusion
Targeted resequencing
1
AITL
66
M
Lymph node
N
no
2
AITL
64
M
Lymph node
N
yes
3
AITL
28
F
Lymph node
P
yes
4
AITL
59
M
Lymph node
P
yes
5
AITL
76
F
Lymph node
P
yes
6
AITL
72
M
Lymph node
P
yes
7
AITL
67
M
Lymph node
P
no
8
AITL
59
M
Lymph node
P
yes
9
AITL
52
M
Lymph node
P
yes
10
AITL
42
M
Lymph node
P
yes
11
AITL
60
F
Lymph node
N
yes
12
AITL
53
F
Lymph node
P
yes
13
AITL
67
M
Lymph node
N
yes
14
AITL
57
F
Lymph node
P
yes
15
AITL
58
M
Lymph node
N
no
16
AITL
62
M
Lymph node
P
no
17
AITL
51
M
Lymph node
P
yes
18
AITL
60
F
Lymph node
P
yes
19
AITL
57
F
Lymph node
P
yes
20
AITL
47
M
Lymph node
P
yes
21
AITL
50
F
Lymph node
N
yes
22
AITL
58
M
Lymph node
N
no
23
AITL
67
M
Lymph node
N
no
24
AITL
75
M
Lymph node
P
no
25
AITL
64
M
Lymph node
N
yes
26
AITL
74
M
Lymph node
P
no
27
AITL
72
M
Lymph node
N
no
28
AITL
75
M
Lymph node
N
yes
29
AITL
62
M
Lymph node
P
yes
30
AITL
38
M
Lymph node
N
yes
31
AITL
54
M
Lymph node
P
yes
32
AITL
57
M
Lymph node
P
yes
33
AITL
65
M
Lymph node
P
yes
34
AITL
41
F
Lymph node
N
no
35
AITL
77
M
Lymph node
N
no
36
AITL
78
M
Lymph node
N
yes
37
AITL
79
M
Lymph node
N
no
38
AITL
57
M
Lymph node
P
no
39
AITL
73
F
Lymph node
N
yes
10
40
AITL
70
M
Lymph node
P
yes
41
AITL
63
M
Lymph node
P
no
42
AITL
60
F
Lymph node
P
yes
43
AITL
77
M
Lymph node
N
no
44
AITL
56
M
Lymph node
N
no
45
AITL
48
M
Lymph node
N
yes
46
ALCL. ALK-
62
M
Lymph node
N
yes
47
ALCL. ALK+
9
M
Mediastinum
N
yes
48
EATL
52
F
Small intestine
N
no
49
EATL
53
F
Jejunum
N
yes
50
EATL
50
M
Ileum
N
yes
51
ENKL
45
M
Nasal cavity
P
yes
52
ENKL
58
F
Lymph node
P
yes
53
ENKL
31
M
Cecum
N
yes
54
ENKL
64
M
Nasal cavity
P
yes
55
ENKL
54
M
Soft palate
N
yes
56
ENKL
50
M
Nasal cavity
N
yes
57
ENKL
46
M
Nasal cavity
N
yes
58
ENKL
66
F
Nasal cavity
N
yes
59
ENKL
57
M
Testis
N
yes
60
ENKL
34
M
Nasal cavity
N
yes
61
ENKL
51
F
Nasal cavity
N
no
62
ENKL
41
F
Lymph node
N
no
63
ENKL
48
M
Nasal cavity
P
yes
64
ENKL
48
M
Paranasal sinus
N
no
65
ENKL
47
M
Testis
N
yes
66
ENKL
43
M
Ileum
P
yes
67
ENKL
25
M
Jejunum
N
yes
68
ENKL
45
M
Testis
N
yes
69
ENKL
83
F
Nasal cavity
N
yes
70
ENKL
54
F
Nasal cavity
N
yes
71
ENKL
40
M
Colon
P
yes
72
ENKL
75
M
Nasal cavity
N
yes
73
ENKL
55
F
Nasal cavity
N
no
74
ENKL
60
M
Nasal cavity
P
yes
75
ENKL
44
F
Nasal cavity
N
yes
76
ENKL
32
F
Nasal cavity
N
yes
77
ENKL
73
M
Nasal cavity
ND
yes
78
ENKL
34
M
Nasal cavity
ND
yes
79
ENKL
76
F
Soft tissue
N
no
80
ENKL
43
F
Nasal cavity
N
no
81
ENKL
62
M
Nasal cavity
P
no
82
ENKL
44
M
Nasopharynx
P
yes
11
83
ENKL
24
M
Skin
N
no
84
ENKL*
55
M
Lymph node
ND
yes
85
PTCL
74
M
Lymph node
P
no
86
PTCL
29
F
Mediastinum
N
yes
87
PTCL
51
M
Lymph node
N
yes
88
PTCL
62
M
Stomach
N
no
89
PTCL?
64
F
Tonsil
P
no
90
PTCL
54
F
Lymph node
N
no
91
PTCL
67
M
Lymph node
N
no
92
PTCL
46
F
Lymph node
P
yes
93
PTCL
48
F
Lymph node
P
yes
94
PTCL
15
M
Lymph node
N
no
95
PTCL
70
M
Lymph node
N
no
96
PTCL
78
M
Larynx
N
no
97
PTCL
69
F
Jejunum
N
no
98
PTCL
49
M
Soft tissue
N
yes
99
PTCL
57
M
Lymph node
N
yes
100
PTCL
66
M
Lymph node
P
yes
101
PTCL
60
F
Lymph node
N
no
102
PTCL
73
F
Lymph node
P
yes
103
PTCL
60
F
Lymph node
N
yes
104
PTCL
49
F
Lymph node
N
no
105
PTCL
47
F
Lymph node
N
no
106
PTCL
51
F
Lymph node
P
no
107
PTCL
77
M
Lymph node
N
no
108
PTCL
24
M
Lymph node
N
no
109
PTCL
38
M
Skin
N
yes
110
PTCL
42
F
Colon
N
no
111
PTCL
71
F
Lymph node
N
no
112
PTCL
55
F
Colon
P
no
113
PTCL
75
F
Nasopharynx
ND
yes
114
PTCL
65
F
Tonsil
P
no
115
PTCL
68
M
Lymph node
N
yes
116
PTCL
53
M
Lymph node
N
yes
117
PTCL
69
M
Lymph node
N
no
118
PTCL
76
M
Lymph node
P
yes
119
PTCL
39
F
Lymph node
N
yes
AITL: Angioimmunoblastric T cell lymphoma, ENKL: Extranodal NK/T cell lymphoma, * ENKL, nodal, ALCL; Anaplastic large cell lymphoma, EATL: Enteropathy-associated T cell lymphoma, PTCL: Peripheral T cell lymphoma, NOS (not otherwise specified) N: negative, P:positive, ND: not done
12
Supplementary Table S3. List of gene-specific PCR primers PCR Type
cDNA RT-PCR primer
cDNA qPCR primer
gDNA PCR primer
gDNA qPCR primer
Figure Reference Fig. 2A, Fig. S2
Primer Name
Sequence
Fusion_cRT1 F Fusion_cRT1 R
5’- GATCCTTGCAGCAGTTAGTTCGGGG-3’ 5’- GGGCTGGTAATGCTTGCGGGTGGGC-3’
Fig. S1A
Fusion_cRT2 F Fusion_cRT2 R
5’- GGACTGAGGGCCATGGACACGGGAC-3’ 5’- GGAGCGATAGGCTGCGAAGTCGC-3’
Fig. S1B
Fusion_cRT3 F Fusion_cRT2 R
5’-AGCTGAACCTGGCTACCAGG-3’ 5’-GGAGCGATAGGCTGCGAAGTCGC-3’
Fig. 2A, Figs. S1,S2
-actin_cRT F -actin_cRT R
5’-CCAACCGCGAGAAGATGACC-3’ 5’-GGTCCAGACGCAGGATGGC-3’
Fig. 3C
CD28_cQ F CD28_cQ R
5’-ACAATGCGGTCAACCTTAGC-3’ 5’-ACCTGAAGCTGCTGGGAGTA-3’
Fig. 3C
CTLA4_cQ F CTLA4_cQ R
5’-GTGCCCAGATTCTGACTTCC-3’ 5’-CTGGCTCTGTTGGGGGCATTTTC-3’
Fig. 3C
Fusion_cQ F Fusion_cQ R
5’-CAGCAGTTAGTTCGGGGTTG-3’ 5’-GCGGGGAGTCATGTTCATGT-3’
Fig. 3C
-actin_cQ F -actin_cQ R
5’-CCAACCGCGAGAAGATGACC-3’ 5’-GGTCCAGACGCAGGATGGC-3’
Fig. 3E
Fusion_G1 F Fusion_G1 R
5’- CTGTCTCAGGGAGGCTCTGC-3’ 5’- CGGCTGGCTTCTGGATAGG-3’
Fig. S8A
Fusion_G1 F Fusion_G2 R
5’-CTGTCTCAGGGAGGCTCTGC-3’ 5’-GGCGGTCATTTCCTATCCAG-3’
Fig. S6
CD28_gQ1 F CD28_gQ1 R
5’-AGGCATTGATGAGGATACGC-3’ 5’-TTCTATCCCTTGCCATGACC-3’
Fig. S6
CD28_gQ2 F CD28_gQ2 R
5’-AGGGATGGGTTACAGCACAG-3’ 5’-GAGTTCGAGGAAGCCAGTTG-3’
Fig. S6
CD28_gQ3 F CD28_gQ3 R
5’-CGGTGAGCAAGCAGAATACA-3’ 5’-GGAAGAGCAACCAACTCCAG-3’
Fig. S6
CD28_gQ4 F CD28_gQ4 R
5’-GGCCCACATTCCAACTTACC-3’ 5’-GGGAAGAGGCTCCCAGAATC-3’
Fig. S6
CD28_gQ5 F CD28_gQ5 R
5’-TCCAATCAGACCAGGTAGGAGC-3’ 5’-CCACAACCCACTTTGGATCTCC-3’
Fig. 3B, Fig. S6
CD28_gQ6 F CD28_gQ6 R
5’-CACAGGCATGTTCCTACCTCAGG-3’ 5’-GGACCTGAAGGGTGACGAGG-3’
Fig. 3B, Fig. S6
CTLA4_gQ1 F CTLA4_gQ1 R
5’-CTCACTATCCAAGGACTGAGGGC-3’ 5’-CTGGGTTCCGTTGCCTATGC-3’
Fig. S6
CTLA4_gQ2 F CTLA4_gQ2 R
5’-TGCAATTTAGGGGTGGACCT-3’ 5’-AGAATCTGGGCACGGTTCTGGAT-3’
Fig. S6
CTLA4_gQ3 F CTLA4_gQ3 R
5’-CCATCACCTGGAAGTCACCT-3’ 5’-CCCAGTCAAGCAAACTGGAT-3’
Fig. S6
CTLA4_gQ4 F CTLA4_gQ4 R
5’-CAGGGAAGTTTTGTGGAGGA-3’ 5’-CACAATTCCACGCAATCAAG-3’
Fig. 3B, Fig. S6
-actin_gQ F -actin_gQ R
5’-TGAGCCTCATCTCCCACGTA-3’ 5’-TCTCAGCCAGCACCATGACT-3’
13
Supplementary Table S4. Summary statistics of targeted deep sequencing No. Exons
No. Amplicons
CDS (bp)
ACTB
5
14
1128
ANKRD11
11
80
ARID1A
20
ATM
Gene
Target (bp)
design coverage
experimental coverage
bp
%
bp
%
1178
1178
100.0
1178
100.0
7992
8102
7650
94.4
7679
94.8
82
6858
7058
6987
99.0
6898
97.7
62
152
9171
9791
9747
99.6
9771
99.8
B2M
3
7
360
390
390
100.0
390
100.0
BCL10
3
10
702
732
732
100.0
732
100.0
BCL2
2
8
753
773
773
100.0
715
92.5
BRAF
18
39
2301
2481
2446
98.6
2449
98.7
BTG1
2
8
516
536
536
100.0
533
99.4
CARD11
24
54
3465
3705
3705
100.0
3705
100.0
CCND3
6
13
927
987
987
100.0
959
97.2
CD28
4
11
663
703
703
100.0
703
100.0
CD58
6
13
757
817
817
100.0
817
100.0
CD79B
6
11
693
753
753
100.0
753
100.0
CDKN2A
5
11
912
962
962
100.0
962
100.0
CREBBP
31
96
7329
7639
7638
100.0
7497
98.1
DNMT3A
24
46
2864
3104
3104
100.0
3104
100.0
EBF1
16
31
1776
1936
1936
100.0
1936
100.0
EZH2
19
38
2256
2446
2446
100.0
2421
99.0
FYN
12
27
1770
1890
1890
100.0
1890
100.0
GNA13
4
15
1134
1174
1174
100.0
1166
99.3
HIST1H1C
1
7
642
652
652
100.0
652
100.0
HIST1H1E
1
7
660
670
640
95.5
670
100.0
ID3
2
5
360
380
380
100.0
380
100.0
IDH1
8
19
1245
1325
1325
100.0
1325
100.0
IDH2
11
20
1359
1469
1464
99.7
1469
100.0
IRF8
8
18
1281
1361
1353
99.4
1361
100.0
JAK2
23
56
3399
3629
3629
100.0
3629
100.0
JAK3
23
51
3375
3605
3601
99.9
3605
100.0
KLHL6
7
24
1866
1936
1936
100.0
1841
95.1
KMT2C
59
199
14736
15326
15103
98.5
9903
99.9
KMT2D
54
199
16614
17154
17148
100.0
1154
97.2
KRAS
5
11
687
737
737
100.0
15212
99.3
LILRB1
14
22
1967
2107
1713
81.3
16726
97.5
MEF2B
8
16
1107
1187
1171
98.7
737
100.0
14
MKI67
14
103
9771
9911
9819
99.1
1770
84.0
MTOR
57
116
7650
8220
8215
99.9
8159
99.3
MUC2
50
108
8442
8942
8191
91.6
7769
86.9
MYC
3
16
1365
1395
1395
100.0
1395
100.0
MYD88
5
14
954
1004
1004
100.0
1004
100.0
NOTCH1
34
99
7668
8008
7901
98.7
7930
99.0
P2RY8
1
10
1080
1090
1090
100.0
0
0
PCLO
25
181
15446
15696
15571
99.2
15601
99.4
PIK3CD
22
44
3135
3355
3355
100.0
3355
100.0
PIK3R1
17
33
2297
2467
2462
99.8
2467
100.0
PIM1
6
16
1215
1275
1275
100.0
1219
95.6
PLCG1
32
64
3876
4196
4196
100.0
4196
100.0
POU2F2
14
22
1444
1584
1584
100.0
1558
98.4
PRKD2
18
39
2637
2817
2817
100.0
2794
99.2
PTPN1
10
20
1308
1408
1408
100.0
1308
92.9
RHOA
4
9
582
622
622
100.0
622
100.0
RHOT2
19
32
1857
2047
1986
97.0
2028
99.1
SGK1
17
32
1935
2105
2105
100.0
2105
100.0
SMARCAL1
16
42
2865
3025
3025
100.0
3025
100.0
SMARCD1
13
27
1548
1678
1678
100.0
1678
100.0
SOCS1
1
6
636
646
646
100.0
588
91.0
STAT1
23
44
2257
2487
2487
100.0
2487
100.0
STAT2
23
40
2556
2786
2782
99.9
2657
95.4
STAT3
23
40
2313
2543
2521
99.1
2543
100.0
STAT6
21
42
2544
2754
2714
98.6
2754
100.0
TCF3
19
34
2192
2382
2382
100.0
2382
100.0
TET2
9
70
6098
6188
6188
100.0
6111
98.8
TET3
11
61
5388
5498
5498
100.0
5498
100.0
TNFAIP3
8
28
2373
2453
2453
100.0
2453
100.0
TNFRSF14
8
15
852
932
932
100.0
932
100.0
TP53
12
21
1263
1383
1383
100.0
1328
96.0
TP63
16
36
2200
2360
2360
100.0
2360
100.0
VAV1
27
44
2538
2808
2808
100.0
2808
100.0
WIF1
10
17
1140
1240
1240
100.0
1240
100.0
WWOX
10
20
1303
1403
1403
100.0
1403
100.0
Total
1105
2965
216,353
227,403
224,902
98.9
222,449
97.82
15
Supplementary Fig. S1. Validation of the CTLA4-CD28 fusion transcript. (a) Validation of the CTLA4-CD28 fusion transcript by RT-PCR using samples from patients and cell lines. Arrows indicate the approximate positions of oligonucleotide primers on the CTLA4-CD28 fusion transcript. PCR products were amplified from patient 1, CEMC1-15 cells and CEMC7-14 cells and validated by Sanger sequencing. -actin was used as an internal control, and NTC indicates the no template control. Normal tonsil tissue and 293T cells were used as negative controls. (b) The fusion products from CEMC1-15 and CEMC7-14 cells were shorter due to the partial deletion in the exon 2 of CTLA4 gene (391-457 bp region). This is a frame-shift deletion that would lead to loss-of-function of the fusion transcript. Thus, the fusion transcript is expected to play no active functional role in these cell lines.
16
Supplementary Fig. S2. Validation of the CTLA4-CD28 fusion gene in patients. Fusion transcripts were validated by RT-PCR using FFPE samples from TCL patients. Arrows indicate the approximate positions of oligonucleotide primers on the CTLA4-CD28 fusion transcript. PCR products were validated by Sanger sequencing. -actin was used as an internal control, and NTC indicates the no template control.
17
Supplementary Fig. S3. Functional analyses of the CTLA4-CD28 fusion gene in H9 cell line. (a) In vitro proliferation assay in H9 cells. H9 cells expressing the CTLA4-CD28 fusion show enhanced cell proliferation (*P < 0.05 compared with cells expressing wild-type CTLA4) after co-stimulation with anti-CD3/CTLA4 antibodies. Each experiment was repeated three times with five replicates, and the data are expressed as the mean ± standard deviation. (b) Expression of the CTLA4-CD28 fusion enhanced interleukin 2 (IL-2) production after CTLA4 activation (**P < 0.01 compared with cells expressing wild-type CTLA4). H9 cells were stimulated with PMA/Ionomycin (P/I) without or with anti-CD3/CTLA4 antibodies to activate CTLA4. IL-2 measurement was carried out three independent times, and the data are expressed as the mean ± standard deviation.
18
Supplementary Fig. S4. Surface expression levels of CTLA4 and the CTLA4-CD28 fusion. Transfected Jurkat cells were stained with PE-Cy5 labeled anti-CTLA4 and PE-Cy7 labeled antiCD28. GFP-positive cells were gated and analyzed. Only the cells expressing CTLA4 and CTLA4-CD28 fusion transcripts have positive signals on PE-Cy5 labeled anti-CTLA4 staining.
19
Supplementary Fig. S5. Fluorescence in situ hybridization analysis for the fusion. Metaphase chromosome of normal cells without fusion showed two orange signals which indicated merged green and red signal derived from closely located CD28 and CTLA genes, respectively. The cells with fusion showed two orange signals as well. There was no separate signal indicating the presence of episomal fusion gene. This does not rule out the possibility of episome formation in all fusion-positive cases.
20
Supplementary Fig. S6. Copy number analysis of CD28 and CTLA4 genes in fusion negative and positive TCL patients. Copy number change was estimated relative to that in the peripheral blood cells from a normal individual. Quantitative real time PCR was carried out using multiple primer pairs covering genomic loci of CD28 and CTLA4 genes. -actin was used as a normalizing control. Four out of five primer pairs within the fusion region showed significant copy number gains in fusion-positive patients. Results from two representative primer pairs (CD28_gQ5 and CTLA4_gQ1) were shown in Fig. 3b. The fusion regions are indicated by the bracket at the top, and the primer positions are indicated by the bars at the bottom. Cases of statistically significant copy number gain are indicated by the asterisks (P-value < 0.05).
21
Supplementary Fig. S7. Sanger sequencing of genomic DNA for mapping exact fusion points. In Case 1, a LINE element of 47 bp long was identified between the fusion introns. Cases 3-6 and two cell lines are examples of direct joining of two exons. Vertical bars indicate the break points.
22
Supplementary Fig. S8. Computational pipeline for analyzing targeted deep sequencing data
23
Supplementary Fig. S9. Mutation profile of TET2 gene from targeted sequencing
24