Page 148 - 2019_04-Haematologica-web
P. 148

R. Burkhard et al.
DLBCLs.32,47 Furthermore, we linked the enhanced prolif- eration of TIRAP p.R81C B cells with an increased expres- sion of NF-kB target genes and genes involved in cell sur- vival and proliferation in PBMCs. In this context, it is important to stress that we determined the lymphocytes to account for more than 65% of cells in the PMBC sam- ples analyzed. In addition, TIRAP knockdown was paral- leled by a significant decrease in the gene expression sig- nature of the NF-kB pathway, particularly in PBMCs car- rying the p.R81C variant. In contrast, overexpression of TIRAP p.R81C increased NF-kB gene signature in vitro. Moreover, TIRAP p.R81C-expressing cells showed enhanced resistance to stress-induced apoptosis, indicat- ing that TIRAP p.R81C provides a survival advantage under those conditions.
Our data link TIRAP p.R81C variant/expression with increased B-cell proliferation as well as survival, and thus add TIRAP to the existing network of lymphoma risk genes that are associated with deregulated NF-kB signal- ing such as TNFAIP3, CD79A/B, MYD88 and CARD11. Interestingly, all these genes were unmutated in the lym- phomas of both sisters. Similar to patients expressing the oncogenic p.L265P MYD88 variant, patients with aberrant TIRAP signaling might benefit from IRAK4-selective kinase inhibitors.48
Diffuse large B-cell lymphoma is a polygenic disease with a complex pathogenesis. Therefore, additional alter- ations are required for the full malignant transformation of B-cells. Interestingly, all the family members investigated were found to have a homozygous germline loss of GSTT1, a reported risk factor for lymphomas (Online Supplementary Figure S7).18 One can hypothesize that the interplay of the germline TIRAP p.R81C variant and
GSTT1 loss coupled with additional genomic changes cul- minated in B-cell transformation in the investigated fami- ly. The identification of the TIRAP p.R81C variant in a family with mixed ethnic background, along with the demonstration of distinct targets of recurrent mutations in Chinese DLBCLs,49 might be an important additional aspect.
Overall, our findings revealed TIRAP p.R81C to be a potential lymphoma risk variant in a family of mixed eth- nic background. Our analysis complements the existing view on the different players of the NF-kB pathway cru- cially involved in DLBCL.
Acknowledgments
The authors would like to thank Magne Osteras and his team at Fasteris (Geneva, Switzerland) for their excellent technical assistance. The probe for the TNFAIP3 FISH analysis was a gift from Laura Pasqualucci and Riccardo Dalla-Favera (Institute for Cancer Genetics, Columbia University, New York, NY, USA). The determination of CIITA rearrangement by FISH was kindly performed by Laurence de Leval (Institute of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland). We thank Lukas Mager, Deborah Krauer, Mario P. Tschan, Emilly Auma, Adrian F. Ochsenbein, Radek Skoda, Federico Santoni, André Schaller, and Outi Kilpivaara for helpful discussions and critical comments.
Funding
This work was supported by grants from the "Bernische Krebsliga", the "Werner und Hedy Berger-Janser Stiftung", the "Bernische Stiftung für klinische Krebsforschung", the "Stiftung für klinisch-experimentelle Tumorforschung", and a grant from the Inselspital/Bern University Hospital, all to UN.
References
1. Swerdlow SH, World Health Organization, International Agency for Research on Can- cer, editors. WHO classification of tumours of haematopoietic and lymphoid tissues. Revised 4th edition. Lyon: International Agency for Research on Cancer; 2017.
2. Rosenwald A, Wright G, Leroy K, et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favor- able subgroup of diffuse large B cell lym- phoma related to Hodgkin lymphoma. J Exp Med. 2003;198(6):851-862.
3. Savage KJ, Monti S, Kutok JL, et al. The molecular signature of mediastinal large B- cell lymphoma differs from that of other diffuse large B-cell lymphomas and shares features with classical Hodgkin lymphoma. Blood. 2003;102(12):3871-3879.
4. Pasqualucci L, Trifonov V, Fabbri G, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet. 2011; 43(9):830-837.
5. Morin RD, Mendez-Lago M, Mungall AJ, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476(7360):298-303.
6. Morin RD, Mungall K, Pleasance E, et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood. 2013; 122(7):1256-1265.
7. Lohr JG, Stojanov P, Lawrence MS, et al.
Discovery and prioritization of somatic muta-tions in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc Natl Acad Sci U S A. 2012; 109(10):3879- 3884.
8. Zhang J, Grubor V, Love CL, et al. Genetic heterogeneity of diffuse large B-cell lym- phoma. Proc Natl Acad Sci. 2013; 110(4):1398-1403.
9. Dubois S, Viailly P-J, Mareschal S, et al. Next-Generation Sequencing in Diffuse Large B-Cell Lymphoma Highlights Molecular Divergence and Therapeutic Opportunities: a LYSA Study. Clin Cancer Res. 2016;22(12):2919-2928.
10. Gunawardana J, Chan FC, Telenius A, et al. Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma. Nat Genet. 2014;46(4):329-335.
11. Wessendorf S, Barth TFE, Viardot A, et al. Further delineation of chromosomal con- sensus regions in primary mediastinal B-cell lymphomas: an analysis of 37 tumor sam- ples using high-resolution genomic profiling (array-CGH). Leukemia. 2007; 21(12):2463- 2469.
12. Joos S, Otaño-Joos MI, Ziegler S, et al. Primary mediastinal (thymic) B-cell lym- phoma is characterized by gains of chromo- somal material including 9p and amplifica- tion of the REL gene. Blood. 1996; 87(4):1571-1578.
13. Lenz G, Wright GW, Emre NCT, et al. Molecular subtypes of diffuse large B-cell lym-phoma arise by distinct genetic path- ways. Proc Natl Acad Sci U S A. 2008; 105(36):13520-13525.
14. Pasqualucci L, Dalla-Favera R. Genetics of diffuse large B cell lymphoma. Blood. 2018;131(21):2307-2319.
15. Altieri A, Bermejo JL, Hemminki K. Familial risk for non-Hodgkin lymphoma and other lymphoproliferative malignancies by histopathologic subtype: the Swedish Family-Cancer Database. Blood. 2005; 106(2):668-672.
16. Goldin LR, Björkholm M, Kristinsson SY, Turesson I, Landgren O. Highly increased familial risks for specific lymphoma sub- types. Br J Haematol. 2009;146(1):91-94.
17. Cerhan JR, Berndt SI, Vijai J, et al. Genome- wide association study identifies multiple susceptibility loci for diffuse large B cell lymphoma. Nat Genet. 2014;46(11):1233- 1238.
18. Skibola CF, Curry JD, Nieters A. Genetic sus- ceptibility to lymphoma. Haematologica. 2007;92(7):960.
19. Saarinen S, Kaasinen E, Karjalainen- Lindsberg M-L, et al. Primary mediastinal large B-cell lymphoma segregating in a fam- ily: exome sequencing identifies MLL as a candi-date predisposition gene. Blood. 2013;121(17):3428-3430.
20. Horng T, Barton GM, Medzhitov R. TIRAP:
776
haematologica | 2019; 104(4)


































































































   146   147   148   149   150