Page 124 - Read Online
P. 124

Jiang et al. J Transl Genet Genom 2021;5:323-40  https://dx.doi.org/10.20517/jtgg.2021.21  Page 337

               11.       Kwong YL. Natural killer-cell malignancies: diagnosis and treatment. Leukemia 2005;19:2186-94.  DOI  PubMed
               12.       Fox CP, Civallero M, Ko Y, et al. Survival outcomes of patients with extranodal natural-killer T-cell lymphoma: a prospective cohort
                    study from the international T-cell Project. Lancet Haematol 2020;7:e284-94.  DOI  PubMed
               13.       Huang Y, de Reyniès A, de Leval L, et al. Gene expression profiling identifies emerging oncogenic pathways operating in extranodal
                    NK/T-cell lymphoma, nasal type. Blood 2010;115:1226-37.  DOI  PubMed  PMC
               14.       Montes-Mojarro IA, Chen BJ, Ramirez-Ibarguen AF, et al. Mutational profile and EBV strains of extranodal NK/T-cell lymphoma,
                    nasal type in Latin America. Mod Pathol 2020;33:781-91.  DOI  PubMed
               15.       Li Z, Xia Y, Feng L, et al. Genetic risk of extranodal natural killer T-cell lymphoma: a genome-wide association study. Lancet Oncol
                    2016;17:1240-7.  DOI  PubMed  PMC
               16.       Lin G, Xu C, Chen K, et al. Genetic risk of extranodal natural killer T-cell lymphoma: a genome-wide association study in multiple
                    populations. Lancet Oncol 2020;21:306-16.  DOI  PubMed
               17.       Peng RJ, Han BW, Cai QQ, et al. Genomic and transcriptomic landscapes of Epstein-Barr virus in extranodal natural killer T-cell
                    lymphoma. Leukemia 2019;33:1451-62.  DOI  PubMed  PMC
               18.       Xiong J, Cui BW, Wang N, et al. Genomic and transcriptomic characterization of natural killer T cell lymphoma. Cancer Cell
                    2020;37:403-419.e6.  DOI  PubMed
               19.       Lee SH, Singh I, Tisdale S, Abdel-Wahab O, Leslie CS, Mayr C. Widespread intronic polyadenylation inactivates tumour suppressor
                    genes in leukaemia. Nature 2018;561:127-31.  DOI  PubMed  PMC
               20.       Weinhold N, Ashby C, Rasche L, et al. Clonal selection and double-hit events involving tumor suppressor genes underlie relapse in
                    myeloma. Blood 2016;128:1735-44.  DOI  PubMed  PMC
               21.       Dong G, Li Y, Lee L, et al. Genetic manipulation of primary human natural killer cells to investigate the functional and oncogenic
                    roles of PRDM1. Haematologica 2021;106:2427-38.  DOI  PubMed  PMC
               22.       Kallies A, Carotta S, Huntington ND, et al. A role for Blimp1 in the transcriptional network controlling natural killer cell maturation.
                    Blood 2011;117:1869-79.  DOI  PubMed
               23.       Karube K, Nakagawa M, Tsuzuki S, et al. Identification of FOXO3 and PRDM1 as tumor-suppressor gene candidates in NK-cell
                    neoplasms by genomic and functional analyses. Blood 2011;118:3195-204.  DOI  PubMed
               24.       Küçük C, Iqbal J, Hu X, et al. PRDM1 is a tumor suppressor gene in natural killer cell malignancies. Proc Natl Acad Sci U S A
                    2011;108:20119-24.  DOI  PubMed  PMC
               25.       Desmots F, Roussel M, Pangault C, et al. Pan-HDAC inhibitors restore PRDM1 response to IL21 in CREBBP-mutated follicular
                    lymphoma. Clin Cancer Res 2019;25:735-46.  DOI  PubMed
               26.       Kim BR, Park SH, Jeong YA, et al. RUNX3 enhances TRAIL-induced apoptosis by upregulating DR5 in colorectal cancer.
                    Oncogene 2019;38:3903-18.  DOI  PubMed
               27.       Haider A, Steininger A, Ullmann R, et al. Inactivation of RUNX3/p46 promotes cutaneous T-Cell lymphoma. J Invest Dermatol
                    2016;136:2287-96.  DOI  PubMed
               28.       Chisholm KM, Bangs CD, Bacchi CE, Molina-Kirsch H, Cherry A, Natkunam Y. Expression profiles of MYC protein and MYC
                    gene rearrangement in lymphomas. Am J Surg Pathol 2015;39:294-303.  DOI  PubMed
               29.       Ng SB, Selvarajan V, Huang G, et al. Activated oncogenic pathways and therapeutic targets in extranodal nasal-type NK/T cell
                    lymphoma revealed by gene expression profiling. J Pathol 2011;223:496-510.  DOI  PubMed
               30.       Selvarajan V, Osato M, Nah GSS, et al. RUNX3 is oncogenic in natural killer/T-cell lymphoma and is transcriptionally regulated by
                    MYC. Leukemia 2017;31:2219-27.  DOI  PubMed  PMC
               31.       Chen XJ, Zhang WN, Chen B, et al. Homoharringtonine deregulates. MYC ;116:2220-5.  DOI  PubMed  PMC
               32.       Jin J, Wang J, Chen F, et al. Homoharringtonine-based induction regimens for patients with de-novo acute myeloid leukaemia: a
                    multicentre, open-label, randomised, controlled phase 3 trial. Lancet Oncol 2013;14:599-608.  DOI  PubMed
               33.       Yan J, Ng SB, Tay JL, et al. EZH2 overexpression in natural killer/T-cell lymphoma confers growth advantage independently of
                    histone methyltransferase activity. Blood 2013;121:4512-20.  DOI  PubMed
               34.       McCabe MT, Ott HM, Ganji G, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations.
                    Nature 2012;492:108-12.  DOI  PubMed
               35.       Morin RD, Johnson NA, Severson TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell
                    lymphomas of germinal-center origin. Nat Genet 2010;42:181-5.  DOI  PubMed  PMC
               36.       Yan J, Li B, Lin B, et al. EZH2 phosphorylation by JAK3 mediates a switch to noncanonical function in natural killer/T-cell
                    lymphoma. Blood 2016;128:948-58.  DOI  PubMed
               37.       Li B, Yan J, Phyu T, et al. MELK mediates the stability of EZH2 through site-specific phosphorylation in extranodal natural killer/T-
                    cell lymphoma. Blood 2019;134:2046-58.  DOI  PubMed
               38.       Italiano A, Soria J, Toulmonde M, et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma
                    and advanced solid tumours: a first-in-human, open-label, phase 1 study. Lancet Oncol 2018;19:649-59.  DOI  PubMed
               39.       Patmore DM, Jassim A, Nathan E, et al. DDX3X suppresses the susceptibility of hindbrain lineages to medulloblastoma. Dev Cell
                    2020;54:455-470.e5.  DOI  PubMed  PMC
               40.       Phung B, Cieśla M, Sanna A, et al. The X-Linked DDX3X RNA helicase dictates translation reprogramming and metastasis in
                    melanoma. Cell Rep 2019;27:3573-3586.e7.  DOI  PubMed
               41.       Dobashi A, Tsuyama N, Asaka R, et al. Frequent BCOR aberrations in extranodal NK/T-Cell lymphoma, nasal type. Genes
                    Chromosomes Cancer 2016;55:460-71.  DOI  PubMed
               42.       Jiang L, Gu ZH, Yan ZX, et al. Exome sequencing identifies somatic mutations of DDX3X in natural killer/T-cell lymphoma. Nat
   119   120   121   122   123   124   125   126   127   128   129