179
Views
4
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

The Mutation of BTG2 Gene Predicts a Poor Outcome in Primary Testicular Diffuse Large B-Cell Lymphoma

, ORCID Icon, , , , & ORCID Icon show all
Pages 1757-1769 | Published online: 10 Mar 2022

References

  • Cheah CY, Wirth A, Seymour JF. Primary testicular lymphoma. Blood. 2014;123(4):486–493. doi:10.1182/blood-2013-10-530659
  • Xu H, Yao F. Primary testicular lymphoma: a SEER analysis of 1169 cases. Oncol Lett. 2019;17(3):3113–3124. doi:10.3892/ol.2019.9953
  • Zelenetz AD, Gordon LI, Chang JE, et al. NCCN guidelines® insights: B-cell lymphomas, version 5.2021. J Natl Compr Canc Netw. 2021;19(11):1218–1230. doi:10.6004/jnccn.2021.0054
  • Coursey Moreno C, Small WC, Camacho JC, et al. Testicular tumors: what radiologists need to know–differential diagnosis, staging, and management. Radiographics. 2015;35(2):400–415. doi:10.1148/rg.352140097
  • Zucca E, Conconi A, Mughal TI, et al. Patterns of outcome and prognostic factors in primary large-cell lymphoma of the testis in a survey by the International Extranodal Lymphoma Study Group. J Clin Oncol. 2003;21(1):20–27. doi:10.1200/JCO.2003.11.141
  • Pollari M, Leivonen SK, Leppä S. Testicular diffuse large B-cell lymphoma-clinical, molecular, and immunological features. Cancers. 2021;13(16):4049.
  • Menter T, Ernst M, Drachneris J, et al. Phenotype profiling of primary testicular diffuse large B-cell lymphomas. Hematol Oncol. 2014;32(2):72–81. doi:10.1002/hon.2090
  • Kridel R, Telio D, Villa D, et al. Diffuse large B-cell lymphoma with testicular involvement: outcome and risk of CNS relapse in the rituximab era. Br J Haematol. 2017;176(2):210–221. doi:10.1111/bjh.14392
  • Tokiya R, Yoden E, Konishi K, et al. Efficacy of prophylactic irradiation to the contralateral testis for patients with advanced-stage primary testicular lymphoma: an analysis of outcomes at a single institution. Int J Hematol. 2017;106(4):533–540. doi:10.1007/s12185-017-2274-5
  • Trama F, Illiano E, Aveta A, Pandolfo SD, Bertuzzi G, Costantini E. Bilateral primary testicular diffuse large B-CELL lymphoma. Urol Case Rep. 2021;38:101733. doi:10.1016/j.eucr.2021.101733
  • Raut TP, Bhatt M, Hastak M, et al. Neurolymphomatosis as a presenting feature of primary testicular lymphoma. Ann Indian Acad Neurol. 2021;24(2):269–272. doi:10.4103/aian.AIAN_304_20
  • Chen B, Cao DH, Lai L, et al. Adult primary testicular lymphoma: clinical features and survival in a series of patients treated at a high-volume institution in China. BMC Cancer. 2020;20(1):220. doi:10.1186/s12885-020-6711-0
  • Shi Y, Han Y, Yang J, et al. Clinical features and outcomes of diffuse large B-cell lymphoma based on nodal or extranodal primary sites of origin: analysis of 1085 WHO classified cases in a single institution in China. Chin J Cancer Res. 2019;31(1):152–161. doi:10.21147/j.issn.1000-9604.2019.01.10
  • Deng L, Xu-Monette ZY, Loghavi S, et al. Primary testicular diffuse large B-cell lymphoma displays distinct clinical and biological features for treatment failure in rituximab era: a report from the International PTL Consortium. Leukemia. 2016;30(2):361–372. doi:10.1038/leu.2015.237
  • Kraan W, van Keimpema M, Horlings HM, et al. High prevalence of oncogenic MYD88 and CD79B mutations in primary testicular diffuse large B-cell lymphoma. Leukemia. 2014;28(3):719–720. doi:10.1038/leu.2013.348
  • Koukourakis G, Kouloulias V. Lymphoma of the testis as primary location: tumour review. Clin Transl Oncol. 2010;12(5):321–325. doi:10.1007/s12094-010-0513-9
  • Chapuy B, Roemer MG, Stewart C, et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood. 2016;127(7):869–881. doi:10.1182/blood-2015-10-673236
  • Chapuy B, Stewart C, Dunford AJ, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24(5):679–690. doi:10.1038/s41591-018-0016-8
  • Roschewski M. Preventing central nervous system spread in diffuse large B-cell lymphoma - novel approaches needed. Haematologica. 2021;106(9):2298–2300. doi:10.3324/haematol.2021.278559
  • Twa DDW, Mottok A, Savage KJ, Steidl C. The pathobiology of primary testicular diffuse large B-cell lymphoma: implications for novel therapies. Blood Rev. 2018;32(3):249–255. doi:10.1016/j.blre.2017.12.001
  • Ma RZ, Tian L, Tao LY, et al. The survival and prognostic factors of primary testicular lymphoma: two-decade single-center experience. Asian J Androl. 2018;20(6):615–620. doi:10.4103/aja.aja_73_18
  • Zhu D, Zhu J, Yu W, et al. Expression of programmed cell death-ligand 1 in primary testicular diffuse large B cell lymphoma: a retrospective study. Oncol Lett. 2019;18(3):2670–2676. doi:10.3892/ol.2019.10595
  • Wang Y, Li J, Fang Y. Primary testicular T-lymphoblastic lymphoma in a child: a case report. Medicine. 2020;99(26):e20861. doi:10.1097/MD.0000000000020861
  • Yhim HY, Yoon DH, Kim SJ, et al. First-line treatment for primary breast diffuse large B-cell lymphoma using immunochemotherapy and central nervous system prophylaxis: a multicenter phase 2 trial. Cancers. 2020;12(8):2192. doi:10.3390/cancers12082192
  • Miyazaki K, Asano N, Yamada T, et al. DA-EPOCH-R combined with high-dose methotrexate in patients with newly diagnosed stage II-IV CD5-positive diffuse large B-cell lymphoma: a single-arm, open-label, Phase II study. Haematologica. 2020;105(9):2308–2315. doi:10.3324/haematol.2019.231076
  • Bobillo S, Joffe E, Sermer D, et al. Prophylaxis with intrathecal or high-dose methotrexate in diffuse large B-cell lymphoma and high risk of CNS relapse. Blood Cancer J. 2021;11(6):113. doi:10.1038/s41408-021-00506-3
  • Oishi N, Kondo T, Nakazawa T, et al. High prevalence of the MYD88 mutation in testicular lymphoma: immunohistochemical and genetic analyses. Pathol Int. 2015;65(10):528–535. doi:10.1111/pin.12336
  • Zhang Q, Hu H, Chen SY, et al. Transcriptome and regulatory network analyses of CD19-CAR-T immunotherapy for B-ALL. Genomics Proteomics Bioinformatics. 2019;17(2):190–200. doi:10.1016/j.gpb.2018.12.008
  • Leeksma OC, de Miranda NF, Veelken H. Germline mutations predisposing to diffuse large B-cell lymphoma. Blood Cancer J. 2017;7(2):e532. doi:10.1038/bcj.2017.15
  • 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. doi:10.1182/blood-2013-02-483727
  • Zhang Q, Liu W, Zhang HM, et al. hTFtarget: a comprehensive database for regulations of human transcription factors and their targets. Genomics Proteomics Bioinformatics. 2020;18(2):120–128. doi:10.1016/j.gpb.2019.09.006
  • Zhou Y, Liu W, Xu Z, et al. Analysis of genomic alteration in primary central nervous system lymphoma and the expression of some related genes. Neoplasia. 2018;20(10):1059–1069. doi:10.1016/j.neo.2018.08.012
  • Dubois S, Viailly PJ, 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. doi:10.1158/1078-0432.CCR-15-2305
  • Szydłowski M, Dębek S, Prochorec-Sobieszek M, et al. PIM kinases promote survival and immune escape in primary mediastinal large B-cell lymphoma through modulation of JAK-STAT and NF-κB activity. Am J Pathol. 2021;191(3):567–574. doi:10.1016/j.ajpath.2020.12.001
  • Schmitz R, Wright GW, Huang DW, et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 2018;378(15):1396–1407. doi:10.1056/NEJMoa1801445
  • Mandelbaum J, Bhagat G, Tang H, et al. BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma. Cancer Cell. 2010;18(6):568–579. doi:10.1016/j.ccr.2010.10.030
  • Jung H, Yoo HY, Lee SH, et al. The mutational landscape of ocular marginal zone lymphoma identifies frequent alterations in TNFAIP3 followed by mutations in TBL1XR1 and CREBBP. Oncotarget. 2017;8(10):17038–17049. doi:10.18632/oncotarget.14928
  • Vela V, Juskevicius D, Dirnhofer S, Menter T, Tzankov A. Mutational landscape of marginal zone B-cell lymphomas of various origin: organotypic alterations and diagnostic potential for assignment of organ origin. Virchows Arch. 2021. doi:10.1007/s00428-021-03186-3
  • Wang X, Xu X, Cai W, et al. TBL1XR1 mutation predicts poor outcome in primary testicular diffuse large B-cell lymphoma patients. Biomark Res. 2020;8:10. doi:10.1186/s40364-020-00189-1
  • Szydłowski M, Garbicz F, Jabłońska E, et al. Inhibition of PIM kinases in DLBCL targets MYC transcriptional program and augments the efficacy of anti-CD20 antibodies. Cancer Res. 2021;81(23):6029–6043. doi:10.1158/0008-5472.CAN-21-1023
  • Kozloski GA, Jiang X, Bhatt S, et al. miR-181a negatively regulates NF-κB signaling and affects activated B-cell-like diffuse large B-cell lymphoma pathogenesis. Blood. 2016;127(23):2856–2866. doi:10.1182/blood-2015-11-680462
  • Boudesco C, Verhoeyen E, Martin L, et al. HSP110 sustains chronic NF-κB signaling in activated B-cell diffuse large B-cell lymphoma through MyD88 stabilization. Blood. 2018;132(5):510–520. doi:10.1182/blood-2017-12-819706
  • Padi SKR, Luevano LA, An N, et al. Targeting the PIM protein kinases for the treatment of a T-cell acute lymphoblastic leukemia subset. Oncotarget. 2017;8(18):30199–30216. doi:10.18632/oncotarget.16320
  • Fukumura K, Kawazu M, Kojima S, et al. Genomic characterization of primary central nervous system lymphoma. Acta Neuropathol. 2016;131(6):865–875. doi:10.1007/s00401-016-1536-2
  • Reddy A, Zhang J, Davis NS, et al. Genetic and functional drivers of diffuse large B cell lymphoma. Cell. 2017;171(2):481–494.e415. doi:10.1016/j.cell.2017.09.027
  • Wang Y, Feng W, Liu P. Genomic pattern of intratumor heterogeneity predicts the risk of progression in early stage diffuse large B-cell lymphoma. Carcinogenesis. 2019;40(11):1427–1434. doi:10.1093/carcin/bgz068
  • Lee B, Lee H, Cho J, et al. Mutational profile and clonal evolution of relapsed/refractory diffuse large B-cell lymphoma. Front Oncol. 2021;11:628807. doi:10.3389/fonc.2021.628807
  • Rouault JP, Falette N, Guéhenneux F, et al. Identification of BTG2, an antiproliferative p53-dependent component of the DNA damage cellular response pathway. Nat Genet. 1996;14(4):482–486. doi:10.1038/ng1296-482
  • Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059–3068. doi:10.1200/JCO.2013.54.8800
  • International Non-Hodgkin’s Lymphoma Prognostic Factors Project. A predictive model for aggressive non-Hodgkin’s lymphoma. N Engl J Med. 1993;329(14):987–994. doi:10.1056/NEJM199309303291402
  • Masliah-Planchon J, Garinet S, Pasmant E. RAS-MAPK pathway epigenetic activation in cancer: miRNAs in action. Oncotarget. 2016;7(25):38892–38907. doi:10.18632/oncotarget.6476
  • Rahrmann EP, Wolf NK, Otto GM, et al. Sleeping beauty screen identifies RREB1 and other genetic drivers in human B-cell lymphoma. Mol Cancer Res. 2019;17(2):567–582. doi:10.1158/1541-7786.MCR-18-0582