• Aucun résultat trouvé

Expression of PAX5 splice variants: a phenomenon of stress- stress-induced, illegitimate splicing?

Extensive alternative splicing of PAX5has been described in both malignant and normal B-cells and the existence of disease-specific isoforms has been suggested (Santoro et al, 2009). Disturbingly, the PAX5 isoform expression patterns observed were inconsistent between studies, even in normal B-cells. Thus, it has been questioned whether these variations result from an irreproducible pattern of PAX5 alternative transcripts or distinct expression patterns that are associated with specific B-cell malignancies (Borson et al, 2002; Robi-chaud et al, 2004; Sadakane et al, 2007; Sekine et al, 2007).

While a recent study did not reveal any significant differences in thePAX5transcript patterns between normal and cancerous B-lymphocytes derived from chronic lymphocytic leukaemia and lymphoma (Arseneauet al, 2009), others claim that either a higher number or even leukaemia-specific isoforms are expressed in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) (Santoroet al, 2009).

Our own observation – that paediatric BCP-ALL bone marrow (BM) samples which had been shipped overnight expressed higher numbers of isoforms – and previous studies suggesting that ‘ageing’ of blood or cold shock conditions may induce illegitimate splicing (Gaytheret al, 1997; Arset al, 2000;

Wimmeret al, 2000; Liuet al, 2010) prompted us to investigate the expression ofPAX5isoforms under such stress conditions.

The analysis ofPAX5isoforms in cells isolated from freshly drawn blood from healthy donors as compared to those of

‘aged’ lymphocytes isolated from blood which was left at room temperature (RT) for 1–4 d (for details see Appendix S1), irrespective of the anticoagulant used, reproducibly revealed a progressive increase of alternative splice variants at the expense of full-length (FL) PAX5transcripts (Fig 1A, B). Both alter-native FL-PAX5transcripts comprising exons 1A-10 or exons 1B-10 (Fig 1A, B) were affected in the same way, and separate amplification of either the 5¢ region (exons 1A-6) or the 3¢

region (exons 5–10) of PAX5 (Fig 1C, D) verified this observation.

Although in fresh samples inter-individual differences in the PAX5expression patterns were found, FL-PAX5was always the predominant isoform. In our hands also the cell lines NALM6 and REH expressed mainly FL-PAX5, no matter which culture conditions, including starvation and suboptimal cell densities (for details see Appendix S1), were used (data not shown).

However, incubation of the B-cell line RAJI at RT for 24 or 48 h induced the expression ofPAX5isoforms and subsequent re-cultivation at 37!C reversed this process (Fig 1E), support-ing the hypothesis of cold shock-stimulated illegitimate splicing. These data corroborate the recent finding that FL-PAX5is always the predominant isoform (Arseneauet al, 2009), but contradict earlier studies in whichPAX5variants, rather than FL-PAX5, were expressed in some B-cell lines (Robichaudet al, 2004; Arseneauet al, 2009).

Cloning and sequencing of a number of splice variants amplified from ‘aged’ normal lymphocytes revealed that all isoforms identified corresponded to those previously described (Borson et al, 2002; Robichaud et al, 2004; Arseneau et al, 2009), including transcripts with various deletions of exons such as D2, D7/8, D2/7/8, D2/7/8/9, D2/4/8, and D2/3/4/8.

Importantly, also the PAX5D2 isoform, which has been recently suggested to play a role in the development of BCP-ALL (Santoro et al, 2009), was readily expressed in ‘aged’

normal lymphocytes.

The basis for age- and temperature-induced missplicing has not been studied in detail yet. Nevertheless, it has been proposed that the fidelity of splicing – at least for some genes – is affected in ‘aged’ lymphocytes (Wimmer et al, 2000; Liu et al, 2010), rather than caused by a general RNA degradation.

Accordingly, the transcript patterns of HPRT1, IKZF1, and RUNX1 were not affected by ‘ageing’ of the samples (Fig 1F–H). Together, these data strongly suggest that in

‘aged’ blood,PAX5is prone to abnormal isoform expression.

To further substantiate that PAX5 isoform expression patterns in BCP-ALL may be altered dependent on the elapsed

ª2011 Blackwell Publishing Ltd,British Journal of Haematology,155,268–280 277

138

Bibliography

139

1. Rodriguez-Abreu, D., A. Bordoni, and E. Zucca, Epidemiology of hematological malignancies. Ann Oncol, 2007. 18 Suppl 1: p. i3-i8.

2. Armitage et al., in Non-Hodgkin Lymphomas, 2nd edition, J. O. Armitage, B. Coiffier, R.

Dalla Favera, N. L. Harris, P. M. Mauch, Eds. (Lippincott Williams & Wilkins, Philadelphia, PA, USA, 2010), pp. 304-326.

3. Martelli, M., et al., Diffuse large B-cell lymphoma. Crit Rev Oncol Hematol, 2013. 87(2):

p. 146-71.

4. Gucalp, A. and A. Noy, Spectrum of HIV lymphoma 2009. Curr Opin Hematol, 2010.

17(4): p. 362-7.

5. Carbone, A. and G. Gaidano, Acquired immunodeficiency syndrome-related cancer. A study model for the mechanisms contributing to the genesis of cancer. Eur J Cancer, 2001.

37(10): p. 1184-7.

6. Martinez, A., J. Carreras, and E. Campo, The follicular lymphoma microenvironment:

From tumor cell to host immunity. Curr Hematol Malig Rep, 2008. 3(4): p. 179-86.

7. de Jong, D., et al., Immunohistochemical prognostic markers in diffuse large B-cell lymphoma: validation of tissue microarray as a prerequisite for broad clinical applications (a study from the Lunenburg Lymphoma Biomarker Consortium). J Clin Pathol, 2009.

62(2): p. 128-38.

8. Rossi, D. and G. Gaidano, Richter syndrome: molecular insights and clinical perspectives.

Hematol Oncol, 2009. 27(1): p. 1-10.

9. Fanale, M.A. and A. Younes, Nodular lymphocyte predominant Hodgkin's lymphoma.

Cancer Treat Res, 2008. 142: p. 367-81.

10. Armitage, J.O., How I treat patients with diffuse large B-cell lymphoma. Blood, 2007.

110(1): p. 29-36.

11. Lenz, G. and L.M. Staudt, Aggressive lymphomas. N Engl J Med, 2010. 362(15): p. 1417-29.

12. A predictive model for aggressive non-Hodgkin's lymphoma. The International Non-Hodgkin's Lymphoma Prognostic Factors Project. N Engl J Med, 1993. 329(14): p. 987-94.

13. Moskowitz, C., Diffuse large B cell lymphoma: how can we cure more patients in 2012?

Best Pract Res Clin Haematol, 2012. 25(1): p. 41-7.

14. Alizadeh, A.A., et al., Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature, 2000. 403(6769): p. 503-11.

15. Rosenwald, A. and L.M. Staudt, Gene expression profiling of diffuse large B-cell lymphoma. Leuk Lymphoma, 2003. 44 Suppl 3: p. S41-7.

16. Savage, K.J., 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): p. 3871-9.

17. Rosenwald, A., 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): p. 1937-47.

18. Visco, C., et al., Patients with diffuse large B-cell lymphoma of germinal center origin with BCL2 translocations have poor outcome, irrespective of MYC status: a report from an International DLBCL rituximab-CHOP Consortium Program Study. Haematologica, 2013.

98(2): p. 255-63.

19. Frick, M., B. Dorken, and G. Lenz, 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): p. 3-12.

20. Monti, S., et al., Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood, 2005. 105(5):

p. 1851-61.

21. Caro, P., et al., Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma. Cancer Cell, 2012. 22(4): p. 547-60.

22. Salles, G., et al., Prognostic significance of immunohistochemical biomarkers in diffuse large B-cell lymphoma: a study from the Lunenburg Lymphoma Biomarker Consortium.

Blood, 2011. 117(26): p. 7070-8.

23. Nyman, H., et al., Prognostic impact of immunohistochemically defined germinal center phenotype in diffuse large B-cell lymphoma patients treated with immunochemotherapy.

Blood, 2007. 109(11): p. 4930-5.

140

24. Natkunam, Y., et al., LMO2 protein expression predicts survival in patients with diffuse large B-cell lymphoma treated with anthracycline-based chemotherapy with and without rituximab. J Clin Oncol, 2008. 26(3): p. 447-54.

25. Meyer, P.N., et al., Immunohistochemical methods for predicting cell of origin and survival in patients with diffuse large B-cell lymphoma treated with rituximab. J Clin Oncol, 2011.

29(2): p. 200-7.

26. Gutierrez-Garcia, G., et al., Gene-expression profiling and not immunophenotypic algorithms predicts prognosis in patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Blood, 2011. 117(18): p. 4836-43.

27. Visco, C., et al., Comprehensive gene expression profiling and immunohistochemical studies support application of immunophenotypic algorithm for molecular subtype classification in diffuse large B-cell lymphoma: a report from the International DLBCL Rituximab-CHOP Consortium Program Study. Leukemia, 2012. 26(9): p. 2103-13.

28. Nyman, H., et al., Prognostic impact of activated B-cell focused classification in diffuse large B-cell lymphoma patients treated with R-CHOP. Mod Pathol, 2009. 22(8): p. 1094-101.

29. Muris, J.J., et al., Immunohistochemical profiling based on Bcl-2, CD10 and MUM1 expression improves risk stratification in patients with primary nodal diffuse large B cell lymphoma. J Pathol, 2006. 208(5): p. 714-23.

30. Barrans, S.L., 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(4): p. 1136-43.

31. Colomo, L., et al., Clinical impact of the differentiation profile assessed by immunophenotyping in patients with diffuse large B-cell lymphoma. Blood, 2003. 101(1):

p. 78-84.

32. Choi, W.W., et al., A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy. Clin Cancer Res, 2009. 15(17): p. 5494-502.

33. Hans, C.P., et al., Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood, 2004. 103(1): p.

275-82.

34. Cedar, H. and Y. Bergman, Epigenetics of haematopoietic cell development. Nat Rev Immunol, 2011. 11(7): p. 478-88.

35. Wilson, A. and A. Trumpp, Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol, 2006. 6(2): p. 93-106.

36. Kuppers, R., Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer, 2005. 5(4): p.

251-62.

37. Janeway C.A.Jr, T.P., Walport M, Shlomchik M.J., immonobiology: the immune system in health and disease. Garland Science, 2005. 6th edition.

38. Klaus, G.G. and J.H. Humphrey, The generation of memory cells. I. The role of C3 in the generation of B memory cells. Immunology, 1977. 33(1): p. 31-40.

39. de Villartay, J.P., A. Fischer, and A. Durandy, The mechanisms of immune diversification and their disorders. Nat Rev Immunol, 2003. 3(12): p. 962-72.

40. Liu, Y.J., et al., Sites of specific B cell activation in primary and secondary responses to T cell-dependent and T cell-independent antigens. Eur J Immunol, 1991. 21(12): p. 2951-62.

41. MacLennan, I.C., Germinal centers. Annu Rev Immunol, 1994. 12: p. 117-39.

42. Klein, U., et al., Transcriptional analysis of the B cell germinal center reaction. Proc Natl Acad Sci U S A, 2003. 100(5): p. 2639-44.

43. Phan, R.T. and R. Dalla-Favera, The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature, 2004. 432(7017): p. 635-9.

44. Hu, B.T., et al., Telomerase is up-regulated in human germinal center B cells in vivo and can be re-expressed in memory B cells activated in vitro. J Immunol, 1997. 159(3): p.

1068-71.

45. Brink, R., et al., Visualizing the effects of antigen affinity on T-dependent B-cell differentiation. Immunol Cell Biol, 2008. 86(1): p. 31-9.

46. Manser, T., Textbook germinal centers? J Immunol, 2004. 172(6): p. 3369-75.

47. Paus, D., et al., Antigen recognition strength regulates the choice between extrafollicular plasma cell and germinal center B cell differentiation. J Exp Med, 2006. 203(4): p. 1081-91.

141

48. Zotos, D., et al., IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism. J Exp Med, 2010. 207(2): p. 365-78.

49. Linterman, M.A., et al., IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses. J Exp Med, 2010. 207(2): p. 353-63.

50. Schwickert, T.A., et al., In vivo imaging of germinal centres reveals a dynamic open structure. Nature, 2007. 446(7131): p. 83-7.

51. Victora, G.D., et al., Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell, 2010. 143(4): p. 592-605.

52. Allen, C.D., et al., Imaging of germinal center selection events during affinity maturation.

Science, 2007. 315(5811): p. 528-31.

53. Victora, G.D., et al., Identification of human germinal center light and dark zone cells and their relationship to human B-cell lymphomas. Blood, 2012. 120(11): p. 2240-8.

54. Kepler, T.B. and A.S. Perelson, Cyclic re-entry of germinal center B cells and the efficiency of affinity maturation. Immunol Today, 1993. 14(8): p. 412-5.

55. Beltman, J.B., et al., B cells within germinal centers migrate preferentially from dark to light zone. Proc Natl Acad Sci U S A, 2011. 108(21): p. 8755-60.

56. Phan, T.G., et al., High affinity germinal center B cells are actively selected into the plasma cell compartment. J Exp Med, 2006. 203(11): p. 2419-24.

57. Blink, E.J., et al., Early appearance of germinal center-derived memory B cells and plasma cells in blood after primary immunization. J Exp Med, 2005. 201(4): p. 545-54.

58. Revy, P., et al., Activation-induced cytidine deaminase (AID) deficiency causes the autosomal recessive form of the Hyper-IgM syndrome (HIGM2). Cell, 2000. 102(5): p.

565-75.

59. Muramatsu, M., et al., Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell, 2000. 102(5): p.

553-63.

60. Odegard, V.H. and D.G. Schatz, Targeting of somatic hypermutation. Nat Rev Immunol, 2006. 6(8): p. 573-83.

61. McKean, D., et al., Generation of antibody diversity in the immune response of BALB/c mice to influenza virus hemagglutinin. Proc Natl Acad Sci U S A, 1984. 81(10): p. 3180-4.

62. Berek, C. and C. Milstein, Mutation drift and repertoire shift in the maturation of the immune response. Immunol Rev, 1987. 96: p. 23-41.

63. Pasqualucci, L., et al., BCL-6 mutations in normal germinal center B cells: evidence of somatic hypermutation acting outside Ig loci. Proc Natl Acad Sci U S A, 1998. 95(20): p.

11816-21.

64. Capello, D., et al., Distribution and pattern of BCL-6 mutations throughout the spectrum of B-cell neoplasia. Blood, 2000. 95(2): p. 651-9.

65. Pasqualucci, L., et al., Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature, 2001. 412(6844): p. 341-6.

66. Gaidano, G., et al., Aberrant somatic hypermutation in multiple subtypes of AIDS-associated non-Hodgkin lymphoma. Blood, 2003. 102(5): p. 1833-41.

67. Rossi, D., et al., Aberrant somatic hypermutation in primary mediastinal large B-cell lymphoma. Leukemia, 2005. 19(12): p. 2363-6.

68. Bhatia, K., et al., Mutations in the coding region of c-myc occur frequently in acquired immunodeficiency syndrome-associated lymphomas. Blood, 1994. 84(3): p. 883-8.

69. Bhatia, K., et al., Point mutations in the c-Myc transactivation domain are common in Burkitt's lymphoma and mouse plasmacytomas. Nat Genet, 1993. 5(1): p. 56-61.

70. Pasqualucci, L., et al., Mutations of the BCL6 proto-oncogene disrupt its negative autoregulation in diffuse large B-cell lymphoma. Blood, 2003. 101(8): p. 2914-23.

71. Klein, U. and R. Dalla-Favera, Germinal centres: role in B-cell physiology and malignancy. Nat Rev Immunol, 2008. 8(1): p. 22-33.

72. Pasqualucci, L., et al., AID is required for germinal center-derived lymphomagenesis. Nat Genet, 2008. 40(1): p. 108-12.

73. Okada, T., et al., Chemokine requirements for B cell entry to lymph nodes and Peyer's patches. J Exp Med, 2002. 196(1): p. 65-75.

74. Gunn, M.D., et al., Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J Exp Med, 1999. 189(3): p.

451-60.

142

75. Vassileva, G., et al., The reduced expression of 6Ckine in the plt mouse results from the deletion of one of two 6Ckine genes. J Exp Med, 1999. 190(8): p. 1183-8.

76. Forster, R., et al., CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell, 1999. 99(1): p. 23-33.

77. Allen, C.D., et al., Germinal center dark and light zone organization is mediated by CXCR4 and CXCR5. Nat Immunol, 2004. 5(9): p. 943-52.

78. Caron, G., et al., CXCR4 expression functionally discriminates centroblasts versus centrocytes within human germinal center B cells. J Immunol, 2009. 182(12): p. 7595-602.

79. Green, J.A., et al., The sphingosine 1-phosphate receptor S1P(2) maintains the homeostasis of germinal center B cells and promotes niche confinement. Nat Immunol, 2011. 12(7): p.

672-80.

80. Voigt, I., et al., CXCR5-deficient mice develop functional germinal centers in the splenic T cell zone. Eur J Immunol, 2000. 30(2): p. 560-7.

81. Balabanian, K., et al., The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem, 2005. 280(42): p. 35760-6.

82. Kunkel, E.J. and E.C. Butcher, Plasma-cell homing. Nat Rev Immunol, 2003. 3(10): p.

822-9.

83. Hargreaves, D.C., et al., A coordinated change in chemokine responsiveness guides plasma cell movements. J Exp Med, 2001. 194(1): p. 45-56.

84. Trentin, L., et al., Homeostatic chemokines drive migration of malignant B cells in patients with non-Hodgkin lymphomas. Blood, 2004. 104(2): p. 502-8.

85. Schmidlin, H., S.A. Diehl, and B. Blom, New insights into the regulation of human B-cell differentiation. Trends Immunol, 2009. 30(6): p. 277-85.

86. Cobaleda, C., et al., Pax5: the guardian of B cell identity and function. Nat Immunol, 2007.

8(5): p. 463-70.

87. Schebesta, A., et al., Transcription factor Pax5 activates the chromatin of key genes involved in B cell signaling, adhesion, migration, and immune function. Immunity, 2007.

27(1): p. 49-63.

88. Horcher, M., A. Souabni, and M. Busslinger, Pax5/BSAP maintains the identity of B cells in late B lymphopoiesis. Immunity, 2001. 14(6): p. 779-90.

89. Urbanek, P., et al., Complete block of early B cell differentiation and altered patterning of the posterior midbrain in mice lacking Pax5/BSAP. Cell, 1994. 79(5): p. 901-12.

90. Lin, L., A.J. Gerth, and S.L. Peng, Active inhibition of plasma cell development in resting B cells by microphthalmia-associated transcription factor. J Exp Med, 2004. 200(1): p. 115-22.

91. Shaffer, A.L., et al., BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control. Immunity, 2000. 13(2): p. 199-212.

92. Fearon, D.T., P.M. Manders, and S.D. Wagner, Bcl-6 uncouples B lymphocyte proliferation from differentiation. Adv Exp Med Biol, 2002. 512: p. 21-8.

93. Tunyaplin, C., et al., Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation. J Immunol, 2004. 173(2): p. 1158-65.

94. Fujita, N., et al., MTA3 and the Mi-2/NuRD complex regulate cell fate during B lymphocyte differentiation. Cell, 2004. 119(1): p. 75-86.

95. Ranuncolo, S.M., et al., Bcl-6 mediates the germinal center B cell phenotype and lymphomagenesis through transcriptional repression of the DNA-damage sensor ATR. Nat Immunol, 2007. 8(7): p. 705-14.

96. Rui, L., et al., Malignant pirates of the immune system. Nat Immunol, 2011. 12(10): p. 933-40.

97. Phan, R.T., et al., Genotoxic stress regulates expression of the proto-oncogene Bcl6 in germinal center B cells. Nat Immunol, 2007. 8(10): p. 1132-9.

98. Basso, K. and R. Dalla-Favera, BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv Immunol, 2010. 105: p. 193-210.

99. Cattoretti, G., et al., Deregulated BCL6 expression recapitulates the pathogenesis of human diffuse large B cell lymphomas in mice. Cancer Cell, 2005. 7(5): p. 445-55.

100. Sciammas, R., et al., Graded expression of interferon regulatory factor-4 coordinates isotype switching with plasma cell differentiation. Immunity, 2006. 25(2): p. 225-36.

101. Klein, U., et al., Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol, 2006. 7(7): p. 773-82.

143

102. Lee, C.H., et al., Regulation of the germinal center gene program by interferon (IFN) regulatory factor 8/IFN consensus sequence-binding protein. J Exp Med, 2006. 203(1): p.

63-72.

103. Arguni, E., et al., JunD/AP-1 and STAT3 are the major enhancer molecules for high Bcl6 expression in germinal center B cells. Int Immunol, 2006. 18(7): p. 1079-89.

104. Muto, A., et al., The transcriptional programme of antibody class switching involves the repressor Bach2. Nature, 2004. 429(6991): p. 566-71.

105. Ochiai, K., et al., Regulation of the plasma cell transcription factor Blimp-1 gene by Bach2 and Bcl6. Int Immunol, 2008. 20(3): p. 453-60.

106. Muto, A., et al., Bach2 represses plasma cell gene regulatory network in B cells to promote antibody class switch. EMBO J, 2010. 29(23): p. 4048-61.

107. Corcoran, L.M., et al., Differential requirement for OBF-1 during antibody-secreting cell differentiation. J Exp Med, 2005. 201(9): p. 1385-96.

108. Emslie, D., et al., Oct2 enhances antibody-secreting cell differentiation through regulation of IL-5 receptor alpha chain expression on activated B cells. J Exp Med, 2008. 205(2): p.

409-21.

109. Sharrocks, A.D., The ETS-domain transcription factor family. Nat Rev Mol Cell Biol, 2001. 2(11): p. 827-37.

110. Su, G.H., et al., Defective B cell receptor-mediated responses in mice lacking the Ets protein, Spi-B. EMBO J, 1997. 16(23): p. 7118-29.

111. Shaffer, A.L., et al., Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program. Immunity, 2002. 17(1): p. 51-62.

112. Chang, T.C., et al., Widespread microRNA repression by Myc contributes to tumorigenesis.

Nat Genet, 2008. 40(1): p. 43-50.

113. Dittmer, J., The biology of the Ets1 proto-oncogene. Mol Cancer, 2003. 2: p. 29.

114. Truong, A.H. and Y. Ben-David, The role of Fli-1 in normal cell function and malignant transformation. Oncogene, 2000. 19(55): p. 6482-9.

115. Barrans, S., 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): p. 3360-5.

116. Savage, K.J., 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): p. 3533-7.

117. Dang, C.V., MYC on the path to cancer. Cell, 2012. 149(1): p. 22-35.

118. Delmore, J.E., et al., BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell, 2011. 146(6): p. 904-17.

119. Mertz, J.A., et al., Targeting MYC dependence in cancer by inhibiting BET bromodomains.

Proc Natl Acad Sci U S A, 2011. 108(40): p. 16669-74.

120. Valera, A., et al., MYC protein expression and genetic alterations have prognostic impact in diffuse large B-cell lymphoma treated with immunochemotherapy. Haematologica, 2013.

121. Cuccuini, W., et al., MYC+ diffuse large B-cell lymphoma is not salvaged by classical R-ICE or R-DHAP followed by BEAM plus autologous stem cell transplantation. Blood, interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A, 2005. 102(43): p.

15545-50.

126. Iqbal, J., et al., BCL2 predicts survival in germinal center B-cell-like diffuse large B-cell lymphoma treated with CHOP-like therapy and rituximab. Clin Cancer Res, 2011. 17(24):

p. 7785-95.

144

127. Boerma, E.G., et al., Translocations involving 8q24 in Burkitt lymphoma and other malignant lymphomas: a historical review of cytogenetics in the light of todays knowledge.

Leukemia, 2009. 23(2): p. 225-34.

128. Davar, D., A.A. Tarhini, and J.M. Kirkwood, Adjuvant immunotherapy of melanoma and development of new approaches using the neoadjuvant approach. Clin Dermatol, 2013.

31(3): p. 237-50.

129. Drake, C.G., Combination immunotherapy approaches. Ann Oncol, 2012. 23 Suppl 8: p.

viii41-6.

130. Ascierto, P.A., et al., Biomarkers for immunostimulatory monoclonal antibodies in combination strategies for melanoma and other tumor types. Clin Cancer Res, 2013. 19(5):

p. 1009-20.

131. Fitzsimmons, D., et al., Pax-5 (BSAP) recruits Ets proto-oncogene family proteins to form functional ternary complexes on a B-cell-specific promoter. Genes Dev, 1996. 10(17): p.

2198-211.

132. John, S.A., et al., Ets-1 regulates plasma cell differentiation by interfering with the activity of the transcription factor Blimp-1. J Biol Chem, 2008. 283(2): p. 951-62.

133. Wang, D., et al., Ets-1 deficiency leads to altered B cell differentiation, hyperresponsiveness to TLR9 and autoimmune disease. Int Immunol, 2005. 17(9): p. 1179-91.

134. Sarrazin, S., et al., Negative and translation termination-dependent positive control of FLI-1 protein synthesis by conserved overlapping 5' upstream open reading frames in Fli-FLI-1 mRNA. Mol Cell Biol, 2000. 20(9): p. 2959-69.

135. Ben-David, Y. and A. Bernstein, Friend virus-induced erythroleukemia and the multistage nature of cancer. Cell, 1991. 66(5): p. 831-4.

136. Kerosuo, L., et al., Myc increases self-renewal in neural progenitor cells through Miz-1. J Cell Sci, 2008. 121(Pt 23): p. 3941-50.

137. Pereira, R., et al., FLI-1 inhibits differentiation and induces proliferation of primary erythroblasts. Oncogene, 1999. 18(8): p. 1597-608.

138. Green, M.R., et al., Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood, 2010. 116(17): p. 3268-77. differences with de novo diffuse large B-cell lymphomas. Hematol Oncol, 2010. 28(2): p.

62-7.

142. Nogai, H., et al., IkappaB-zeta controls the constitutive NF-kappaB target gene network and survival of ABC DLBCL. Blood, 2013. 122(13): p. 2242-50.

143. Fitzgibbon, J., et al., Genome-wide detection of recurring sites of uniparental disomy in follicular and transformed follicular lymphoma. Leukemia, 2007. 21(7): p. 1514-1520.

144. Zhang, L., et al., An immunological renal disease in transgenic mice that overexpress Fli-1,

144. Zhang, L., et al., An immunological renal disease in transgenic mice that overexpress Fli-1,