Роль экспрессии c-MYC, BCL2 и BCL6 в патогенезе диффузной В-крупноклеточной лимфомы

А.Е. Мисюрина1, В.А. Мисюрин2, Е.А. Барях1, А.М. Ковригина1, С.К. Кравченко1

1 ФГБУ «Гематологический научный центр» МЗ РФ, Новый Зыковский пр-д, д. 4а, Москва, Российская Федерация, 125167

2 ФГБУ «Российский онкологический научный центр им. Н.Н. Блохина», Каширское ш., д. 24, Москва, Российская Федерация, 115478

Для переписки: А.Е. Мисюрина, аспирант, Новый Зыковский проезд, д. 4а, Москва, Российская Федерация, 125167; тел.: +7(909)637-32-49; e-mail: anna.lukina1@gmail.com

Для цитирования: Мисюрина А.Е., Мисюрин В.А., Барях Е.А., Ковригина А.М., Кравченко С.К. Роль экспрессии генов c-MYC, BCL2 и BCL6 в патогенезе диффузной В-крупноклеточной лимфомы. Клин. онкогематол. 2014; 7(4): 512–521.


РЕФЕРАТ

Согласно современным представлениям, опирающимся на результаты исследования профиля экспрессии генов, существует несколько подтипов диффузной B-крупноклеточной лимфомы (ДBКЛ): из В-клеток герминативного центра и из активированных В-клеток. Гены c-MYC, BCL6 и BCL2 являются ключевыми регуляторами развития В-лимфоцитов на уровне герминальной (фолликулярной) дифференцировки. В патогенезе ДВКЛ наиболее часто играют роль генетические аномалии с их участием. От общего уровня активности и механизмов, приводящих к гиперэкспрессии каждого из этих генов и продукции соответствующих белков, зависит прогноз заболевания. Мы предполагаем, что определение количественных параметров экспрессии генов c-MYC, BCL6 и BCL2, а также кодируемых ими белков позволит с высокой точностью выделять группы риска среди больных ДBКЛ.


Ключевые слова: диффузная В-крупноклеточная лимфома, молекулярные подтипы, группы риска, c-MYC, BCL6, BCL2.

Принято в печать: 8 сентября 2014 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Swerdlow S.H., Campo E., Harris N.L. et al (eds.). WHO Classification of Tumors of Haematopoetic and Lymphoid Tissues. Lyon: IARC, 2008: 233–4.
  2. Frick M., Dorken B., Lenz G. New insights into the biology of molecular subtypes of diffuse large B-cell lymphoma and Burkitt lymphoma. Best Pract. Res. Clin. Haematol. 2012; 25(1): 3–12.
  3. Alizadeh A.A., Eisen M.B., Davis R.E. et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000; 403: 503–11.
  4. Rosenwald A., Wright G., Chan W.C. et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N. Engl. J. Med. 2002; 346(25): 1937–47.
  5. Alizadeh A.A., Eisen M.B., Davis R.E. et al. The lymphochip: a specialized cDNA microarray for the genomic-scale analysis of gene expression in normal and malignant lymphocytes. Cold Spring Harbor Symp. Quant. Biol. 1999; 62: 71–8.
  6. Lenz G., Wright G., Dave S.S. et al. Stromal gene signatures in large-B-cell lymphomas. N. Engl. J. Med. 2008; 359(22): 2313–23.
  7. Rosenwald A., Wright G., Leroy K. et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J. Exp. Med. 2003; 198(6): 851–62.
  8. Savage K.J., Monti S., Kutok J.L. 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): 3871–9.
  9. Wright G., Tan B., Rosenwald A. et al. A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc. Natl. Acad. Sci. USA. 2003; 100: 9991–6.
  10. Muller A.M., Medvinsky A., Strouboulis J., Grosveld F., Dzierzak E. Development of hematopoietic stem cell activity in the mouse embryo. Immunity. 1994; 1: 291–301.
  11. Melchers F. The pre-B-cell receptor: selector of fitting immunoglobulin heavy chains for the B-cell repertoire. Nat. Rev. Immunol. 2005; 5: 578–84.
  12. van Zelm M.C., Szczepanski T., van der Burg M., van Dongen J.J. Replication history of B lymphocytes reveals homeostatic proliferation and extensive antigen-induced B cell expansion. J. Exp. Med. 2007; 204: 645–55.
  13. Martin F., Oliver A.M., Kearney J.F. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity. 2001; 14: 617–29.
  14. Chen J., Trounstine M., Alt F.W. et al. Immunoglobulin gene rearrangement in B cell deficient mice generated by targeted deletion of the JH locus. Int. Immunol. 1993; 5: 647–56.
  15. Teng G., Papavasiliou F.N. Immunoglobulin somatic hypermutation. Annu. Rev. Genet. 2007; 41: 107–20.
  16. Liu Y.J., Arpin C. Germinal center development. Immunol. Rev. 1997; 156: 111–26.
  17. Yuan D. Regulation of IgM and IgD synthesis in B lymphocytes. II. Translational and post-translational events. J. Immunol. 1984; 132: 1566–70.
  18. Yasodha N. The Biology of the Germinal Center. ASH Education Book. 2007; 1: 210–5.
  19. Komori T., Okada A., Stewart V., Alt F.W. Lack of N regions in antigen receptor variable region genes of TdT-deficient lymphocytes. Science. 1993; 261: 1171–5.
  20. Willenbrock K., Jungnickel B., Hansmann M.L., Kuppers R. Human splenic marginal zone B cells lack expression of activation-induced cytidine deaminase. Eur. J. Immunol. 2005; 35: 3002–7.
  21. Raghavan S.C., Hsieh C.L., Lieber M.R. Both V(D)J coding ends but neither signal end can recombine at the bcl-2 major breakpoint region, and the rejoining is ligase IV dependent. Mol. Cell. Biol. 2005; 15: 6475–84.
  22. Luscher B. MAD1 and its life as a MYC antagonist: an update. Eur. J. Cell. Biol. 2012; 91(6–7): 506–14.
  23. McDuff F.O., Naud J.F., Montagne M., Sauve S., Lavigne P. The Max homodimeric b-HLH-LZ significantly interferes with the specific heterodimerization between the c-Myc and Max b-HLH-LZ in absence of DNA: a quantitative analysis. J. Mol. Recognit. 2009; 22(4): 261–9.
  24. Dang C.V. MYC on the path to cancer. Cell. 2012; 149(1): 22–35.
  25. Luscher B., Vervoorts J. Regulation of gene transcription by the oncoprotein MYC. Gene. 2012; 494(2): 145–60.
  26. Meyer N., Penn L.Z. Reflecting on 25 years with MYC. Nat. Rev. Cancer. 2008; 8(12): 976–90.
  27. Keller U.B., Old J.B., Dorsey F.C. et al. Myc targets Cks1 to provoke the suppression of p27Kip1, proliferation and lymphoma agenesis. EMBO. J. 2007; 26(10): 2562–74.
  28. Bueno M.J., Malumbres M. MicroRNAs and the cell cycle. Biochim. Biophys. Acta. 2011; 1812(5): 592–601.
  29. Nie Z., Hu G., Wei G. et al. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell. 2012; 151(1): 68–79.
  30. Lin C.Y., Loven J., Rahl P.B. et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell. 2012; 151(1): 56–67.
  31. Lin Y., Wong K., Calame K. Repression of c-myc transcription by Blimp-1, an inducer of terminal B cell differentiation. Science. 1997; 276(5312): 596–9.
  32. Basso K., Dalla-Favera R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv. Immunol. 2010; 105: 193–210.
  33. Phan R.T., Saito M., Basso K., Niu H., Dalla-Favera R. BCL6 interacts with the transcription factor Miz-1 to suppress the cyclin-dependent kinase inhibitor p21 and cell cycle arrest in germinal center B cells. Nat. Immunol. 2005; 6(10): 1054–60.
  34. Niu H., Ye B.H., Dalla-Favera R. Antigen receptor signaling induces MAP kinase-mediated phosphorylation and degradation of the BCL-6 transcription factor. Genes Dev. 1998; 12(13): 1953–61.
  35. Phan R.T., Saito M., Kitagawa Y., Means A.R., Dalla-Favera R. Genotoxic stress regulates expression of the proto-oncogene Bcl6 in germinal center B cells. Nat. Immunol. 2007; 8(10): 1132–9.
  36. Phan R.T., Dalla-Favera R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature. 2004; 432(7017): 635–9.
  37. Basso K., Saito M., Sumazin P. et al. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood. 2010; 115(5): 975–84.
  38. Wagner S.D., Ahearne M., Ko Ferrigno P. The role of BCL6 in lymphomas and routes to therapy. Br. J. Haematol. 2011; 152(1): 3–12.
  39. Basso K., Dalla-Favera R. Roles of BCL6 in normal and transformed germinal center B cells. Immunol. Rev. 2012; 247(1): 172–83.
  40. Merino R., Ding L., Veis D.J. et al. Developmental regulation of the Bcl-2 protein and susceptibility to cell death in B lymphocytes. EMBO. J. 1994; 13: 683–91.
  41. McDonnell T.J., Nunez G., Platt F.M. et al. Deregulated Bcl-2-immunoglobulin transgene expands a resting but responsive immunoglobulin M and D-expressing B-cell population. Mol. Cell. Biol. 1990; 10: 1901–7.
  42. McDonnell T.J., Deane N., Platt F.M. et al. Bcl-2-immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell. 1989; 57: 79–88.
  43. Veis D.J., Sorenson C.M., Shutter J.R. et al. Bcl-2-deficient mice demonstrate fulminant lymphoid apoptosis, polycystic kidneys, and hypopigmented hair. Cell. 1993; 75: 229–40.
  44. Wilson W.H., Teruya-Feldstein J., Fest T. et al. Relationship of p53, bcl-2, and tumor proliferation to clinical drug resistance in non-Hodgkin’s lymphomas. Blood. 1997; 89: 601–9.
  45. Monti S., Savage K.J., Kutok J.L. et al. Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood. 2005; 105(5): 1851–61.
  46. Dent A.L., Shaffer A.L., Yu X. et al. Control of inflammation, cytokine expression, and germinal center formation by BCL-6. Science. 1997; 276(5312): 589–92.
  47. Никитин Е.А. Патогенез зрелоклеточных лимфатических опухолей. Материалы конгрессов и конференций. VIII Российский онкологический конгресс [Электронный документ] (http://www.rosoncoweb.ru/library/ congress/ru/08/19.php). [Nikitin E.A. Pathogenesis of mature cell lymphomas. Materialy kongressov i konferentsii. VIII Rossiiskii onkologicheskii kongress (Materials of congresses and conferences. VIII Russian oncological congress). Available at: http://www. rosoncoweb.ru/library/congress/ru/08/19.php (In Russ.)]
  48. Davis R.E., Brown K.D., Siebenlist U. et al. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J. Exp. Med. 2001; 194(12): 1861–74.
  49. Jost P.J., Ruland J. Aberrant NF-kappaB signaling in lymphoma: mechanisms, consequences, and therapeutic implications. Blood. 2007; 109(7): 2700–7.
  50. Ngo V.N., Davis R.E., Lamy L. et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature. 2006; 441(7089): 106–10.
  51. Rawlings D.J., Sommer K., Moreno-Garcia M.E. The CARMA1 signalosome links the signalling machinery of adaptive and innate immunity in lymphocytes. Nat. Rev. Immunol. 2006; 6(11): 799–812.
  52. Lenz G., Davis R.E., Ngo V.N. et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science. 2008; 319(5870): 1676–9.
  53. Davis R.E., Ngo V.N., Lenz G. et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010; 463(7277): 88–92.
  54. Compagno M., Lim W.K., Grunn A. et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. 2009; 459(7247): 717–21.
  55. Kato M., Sanada M., Kato I. et al. Frequent inactivation of A20 in B-cell lymphomas. Nature. 2009; 459(7247): 712–6.
  56. Ding B.B., Yu J.J., Yu R.Y. et al. Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large Bcell lymphomas. Blood. 2008; 111(3): 1515–23.
  57. Lam L.T., Wright G., Davis R.E. et al. Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-{kappa}B pathways in subtypes of diffuse large B-cell lymphoma. Blood. 2008; 111(7): 3701–13.
  58. Ngo V.N., Young R.M., Schmitz R. et al. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011; 470(7332): 115–9.
  59. Bea S., Zettl A., Wright G. et al. Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve geneexpression-based survival prediction. Blood. 2005; 106(9): 3183–90.
  60. Boerma E.G., Siebert R., Kluin P.M., Baudis M. Translocations involving 8q24 in Burkitt lymphoma and other malignant lymphomas: a historical review of cytogenetics in the light of today’s knowledge. Leukemia. 2009; 23(2): 225–34.
  61. Salaverria I., Zettl A., Bea S. et al. Chromosomal alterations detected by comparative genomic hybridization in subgroups of gene expression-defined Burkitt’s lymphoma. Haematologica. 2008; 93(9): 1327–34.
  62. Scholtysik R., Kreuz M., Klapper W. et al. Detection of genomic aberrations in molecularly defined Burkitt’s lymphoma by array-based, high resolution, single nucleotide polymorphism analysis. Haematologica. 2010; 95(12): 2047–55.
  63. Pasqualucci L., Neumeister P., Goossens T. et al. Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature. 2001; 412(6844): 341–6.
  64. Hemann M.T., Bric A., Teruya-Feldstein J. et al. Evasion of the p53 tumour surveillance network by tumour-derived MYC mutants. Nature. 2005; 436(7052): 807–11.
  65. Giulino-Roth L., Wang K., MacDonald T.Y. et al. Targeted genomic sequencing of pediatric Burkitt lymphoma identifies recurrent alterations in antiapoptotic and chromatin-remodeling genes. Blood. 2012; 120(26): 5181–4.
  66. Bhatia K., Huppi K., Spangler G. et al. Point mutations in the c-Myc transactivation domain are common in Burkitt’s lymphoma and mouse plasmacytomas. Nat. Genet. 1993; 5(1): 56–61.
  67. Snuderl M., Kolman O.K., Chen Y.B. et al. B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2010; 34(3): 327–40.
  68. Le Gouill S., Talmant P., Touzeau C. et al. The clinical presentation and prognosis of diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC rearrangement. Haematologica. 2007; 92(10): 1335–42.
  69. Li S., Lin P., Fayad L.E. et al. B-cell lymphomas with B-cell lymphomas with MYC/8q24 rearrangements and IGH@BCL2/t(14;18)(q32;q21): an aggressive disease with heterogeneous histology, germinal center B-cell immunophenotype and poor outcome. Mod. Pathol. 2012; 25(1): 145–56.
  70. Klapper W., Stoecklein H., Zeynalova S. et al. Structural aberrations affecting the MYC locus indicate a poor prognosis independent of clinical risk factors in diffuse large B-cell lymphomas treated within randomized trials of the German High-Grade Non-Hodgkin’s Lymphoma Study Group (DSHNHL). Leukemia. 2008; 22(12): 2226–9.
  71. Savage K.J., Johnson N.A., Ben-Neriah S. et al. MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy. Blood. 2009; 114(17): 3533–7.
  72. Horn H., Ziepert M., Becher C. et al. MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma. Blood. 2013; 121(12): 2253–63.
  73. Barrans S., Crouch S., Smith A. et al. Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab. J. Clin. Oncol. 2010; 28(20): 3360–5.
  74. Valera A., Lopez-Guillermo A., Cardesa-Salzman T. et al. MYC protein expression and genetic alterations have prognostic impact in diffuse large B-cell lymphoma treated with immunochemotherapy. Haematologica. 2013; 98(10): 1554–62.
  75. Hummel M., Bentink S., Berger H. et al. A biologic definition of Burkitt’s lymphoma from transcriptional and genomic profiling. N. Engl. J. Med. 2006; 354(23): 2419–30.
  76. Salaverria I., Siebert R. The gray zone between Burkitt’s lymphoma and diffuse large B-cell lymphoma from a genetics perspective. J. Clin. Oncol. 2011; 29(14): 1835–43.
  77. Bertrand P., Bastard C., Maingonnat C. et al. Mapping of MYC breakpoints in 8q24 rearrangements involving non-immunoglobulin partners in B-cell lymphomas. Leukemia. 2007; 21(3): 515–23.
  78. Tomita N. BCL2 and MYC Dual-Hit Lymphoma/Leukemia. J. Clin. Exp. Hematopathol. 2011; 51(1): 7–12.
  79. Johnson N.A., Savage K.J., Ludkovski O. et al. Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival. Blood. 2009; 114(11): 2273–9.
  80. Snuderl M., Kolman O.K., Chen Y.B. et al. B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2010; 34(3): 327–40.
  81. Hoeller S., Copie-Bergman C. Grey Zone Lymphomas: Lymphomas with Intermediate Features. Advances in Hematology 2012. http://dx.doi. org/10.1155/2012/460801.
  82. Tauro S., Cochrane L., Lauritzsen G.F. et al. Dose-intensified treatment of Burkitt lymphoma and B-cell lymphoma unclassifiable, (with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma) in young adults (< 50 years): A comparison of two adapted BFM protocols. Am. J. Hematol. 2010; 85(4): 261–3.
  83. Kobayashi T., Tsutsumi Y., Sakamoto N. et al. Double-hit Lymphomas Constitute a Highly Aggressive Subgroup in Diffuse Large B-cell Lymphomas in the Era of Rituximab. Jpn. J. Clin. Oncol. 2012; 42(11): 1035–42.
  84. Fanidi A., Harrington E.A., Evan G.I. Cooperative in reactions between c-myc and bcl-2 protooncogenes. Nature. 1992; 359: 554–6.
  85. Vaux D.L., Cory S., Adams J.M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988; 335(6189): 440–2.
  86. Zhaohui J., Stratford M.W., Fengqin G., Tammy F., Xingming D. Bcl2 suppresses DNA repair by enhancing c-myc transcriptional activity. J. Biol. Chem. 2005; 281: 14446–56.
  87. Masao N., Shinobu T., Keiichiro H., Osamu T., Masao S. Synergistic effect of Bcl2, Myc and Ccnd1 transforms mouse primary B cells into malignant cells. Haematologica. 2011; 96(9): 1318–26.
  88. DeoCampo N.D., Wilson M.R., Trosko J.E. Cooperation of bcl-2 and myc in the neoplastic transformation of normal rat liver epithelial cells is related to the down-regulation of gap junction-mediated intercellular communication. Carcinogenesis. 2000; 21(8): 1501–6.
  89. Leucci E., Cocco M., Onnis A. et al. MYC translocation-negative classical Burkitt lymphoma cases: an alternative pathogenetic mechanism involving miRNA deregulation. J. Pathol. 2008; 216(4): 440–50.
  90. Onnis A., De Falco G., Antonicelli G. et al. Аlteration of microRNAs regulated by c-MYC in Burkitt lymphoma. PLoS One. 2010; 5(9); e12960.
  91. Stasik C.J., Nitta H., Zhang W. et al. Increased MYC gene copy number correlates with increased mRNA levels in diffuse large B-cell lymphoma. Haematologica. 2010; 95(4): 597–603.
  92. Schrader A., Bentink S., Spang R. et al. High MYC activity is an independent negative prognostic factor for DLBCL. Cancer. 2012; 131(4): 348–61.
  93. Yoon S.O., Jeon Y.K., Paik J.H. et al. MYC translocation and an increased copy number predict poor prognosis in adult DLBCL, especially in GCB-type. Histopathology. 2008; 53(2): 205–17.
  94. Mossafa H., Damotte D., Jenabian A. et al. Non-Hodgkin lymphomas with Burkitt-like cells are associated with c-Myc amplification and poor prognosis. Leuk. Lymphoma. 2006; 47(9): 1885–93.
  95. Martin-Subero J.I., Odero M.D., Hernandez R. et al. Amplification of IGH/ MYC fusion in clinically aggressive IGH/BCL2-positive germinal center B-cell lymphomas. Genes Chromosomes Cancer. 2005; 43(4): 414–23.
  96. Tapia G., Lopez R., Munoz-Marmol A.M. et al. Immunohistochemical detection of MYC protein correlates with MYC gene status in aggressive B-cell lymphoma. Histopathology. 2011; 59(4): 672–8.
  97. Green T.M., Nielsen O., de SK. et al. High levels of nuclear MYC protein predict the presence of MYC rearrangement in diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2012; 36(4): 612–9.
  98. Johnson N.A., Slack G.W., Savage K.J. et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. 2012; 30(28): 3452–9.
  99. Kluk M.J., Chapuy B., Sinha P. et al. Immunohistochemical detection of MYC-driven diffuse large B-cell lymphomas. PLoS One. 2012; 7(4): e33813.
  100. Testoni M., Kwee I., Greiner T.C. et al. Gains of MYC locus and outcome in patients with diffuse large B-cell lymphoma treated with R-CHOP. Br. J. Haematol. 2011; 155(2): 274–7.
  101. Hu S., Xu-Monette Z.Y., Tzankov A. et al. MYC/BCL2 protein co-expression contributes to the inferior survival of activated B-cell subtype of diffuse large B-cell lymphoma and demonstrates high-risk gene expression signatures: a report from The International DLBCL Rituximab-CHOP Consortium Program Study. Blood. 2013; 121(20): 4021–31.
  102. Piris M.A., Pezzella F., Martinez-Montero J.C. et al. p53 and bcl-2 expression in high-grade B-cell lymphomas: Correlation with survival time. Br. J. Cancer. 1994; 69: 337–41.
  103. Tang S.C., Visser L., Hepperle B. et al. Clinical significance of bcl-2-MBR gene rearrangement and protein expression in diffuse large-cell non-Hodgkin’s lymphoma: An analysis of 83 cases. J. Clin. Oncol. 1994; 12: 149–54.
  104. Barrans S.L., Carter I., Owen R.G. et al. Germinal center phenotype and bcl-2 expression combined with the International Prognostic Index improves patient risk stratification in diffuse large B-cell lymphoma. Blood. 2002; 99: 1136–43.
  105. Colomo L., Lopez-Guillermo A., Perales M. et al. Clinical impact of the differentiation profile assessed by immunophenotyping in patients with diffuse large B-cell lymphoma. Blood. 2003; 101: 78–84.
  106. Gascoyne R.D., Adomat S.A., Krajewski S. et al. Prognostic significance of Bcl-2 protein expression and Bcl-2 gene rearrangement in diffuse aggressive non-Hodgkin’s lymphoma. Blood. 1997; 90: 244–51.
  107. Martinka M., Comeau T., Foyle A. et al. Prognostic significance of t(14;18) and bcl-2 gene expression in follicular small cleaved cell lymphoma and diffuse large cell lymphoma. Clin. Invest. Med. 1997; 20: 364–70.
  108. Hill M.E., MacLennan K.A., Cunningham D.C. et al. Prognostic significance of BCL-2 expression and bcl-2 major breakpoint region rearrangement in dif- fuse large cell non-Hodgkin’s lymphoma: A British National Lymphoma Investigation Study. Blood. 1996; 88: 1046–51.
  109. Kramer M.H., Hermans J., Wijburg E. et al. Clinical relevance of BCL2, BCL6, and MYC rearrangements in diffuse large B-cell lymphoma. Blood. 1998; 92: 3152–62.
  110. Hermine O., Haioun C., Lepage E. et al. Prognostic significance of bcl-2 protein expression in aggressive non-Hodgkin’s lymphoma: Groupe d’Etude des Lymphomes de l’Adulte (GELA). Blood. 1996; 87: 265–72.
  111. Iqbal J., Neppalli V.T., Wright G., Dave B.J. BCL2 Expression Is a Prognostic Marker for the Activated B-Cell–Like Type of Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. 2006; 24(6): 961–8.
  112. Green T.M., Young K.H., Visco C. et al. Immunohistochemical DoubleHit Score Is a Strong Predictor of Outcome in Patients With Diffuse Large B-Cell Lymphoma Treated With Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone. J. Clin. Oncol. 2012; 30(28): 3460–7.
  113. Johnson N.A., Slack G.W., Savage K.J. et al. Concurrent Expression of MYC and BCL2 in Diffuse Large B-Cell Lymphoma Treated With Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone. J. Clin. Oncol. 2012; 30(28): 3452–9.
  114. Valera A., Lopez-Guillermo A., Cardesa-Salzmann T. et al. MYC protein expression and genetic alterations have prognostic impact in patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Haematologica. 2013; 98(10): 1554–62.