Clinical and Prognostic Value of Molecular Markers of Diffuse Large B-Cell Lymphoma

SM Rastorguev1, DA Koroleva2, ES Boulygina1, SV Tsygankova1, NG Goncharov1, OS Naraikin1, NG Gabeeva2, EE Zvonkov2, AV Nedoluzhko1

1 National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova sq., Moscow, Russian Federation, 123182

2 National Medical Hematology Research Center, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Artem Valer’evich Nedoluzhko, PhD in Biology, 1 Akademika Kurchatova sq., Moscow, Russian Federation, 123182; Tel.: +7(916)670-55-95; e-mail: nedoluzhko@gmail.com

For citation: Rastorguev SM, Koroleva DA, Bulygina ES, et al. Clinical and Prognostic Value of Molecular Markers of Diffuse Large B-Cell Lymphoma. Clinical oncohematology. 2019:12(1):95–100.

DOI: 10.21320/2500-2139-2019-12-1-95-100


ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid tumor in adults which is associated with approximately 30–40 % of all non-Hodgkin’s lymphomas. Diagnostic criteria include diffuse growth of large anaplastic tumor cells, expression of В-cell markers, and a high proliferative index. Due to the development of molecular genetic technologies it became obvious that underlying cause of clinical diversity is a huge amount of genetic failures which determine epigenetic modification of gene expression, activation variability of certain signaling pathways, and immunological properties of tumor cells. The study and systemization of molecular markers present a significant trend in DLBCL diagnosis and treatment. This review discusses most important molecular markers and current view on their clinical value.

Keywords: lymphoma, DLBCL, B-cells, transcriptomics, gene expression, epigenomics, genomics.

Received: July 3, 2018

Accepted: December 10, 2018

Read in PDF 


REFERENCES

  1. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503–11. doi: 10.1038/35000501.

  2. Rosenwald A, Alizadeh AA, Widhopf G, et al. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med. 2001;194(11):1639–48. doi: 1084/jem.194.11.1639.

  3. Staudt LM, Dave S. The biology of human lymphoid malignancies revealed by gene expression profiling. Adv Immunol. 2005;87:163–208. doi: 10.1016/S0065-2776(05)87005-1.

  4. Звонков Е.Е., Морозова А.К., Кравченко С.К. и др. Восьмилетний опыт применения модифицированной программы NHL-BFM-90 в лечении взрослых больных первичной диффузной В-крупноклеточной лимфомой желудка. Гематология и трансфузиология. 2012;57(3):47–8.

    [Zvonkov EE, Morozova AK, Kravchenko SK, et al. Eight-year experience of using the modified NHL-BFM-90 program for treatment of adult patients with primary diffuse large B-cell gastric lymphoma. Gematologiya i transfuziologiya. 2012;57(3):47–8. (In Russ)]

  5. Магомедова А.У., Кравченко С.К., Кременецкая A.M. и др. Девятилетний опыт лечения больных диффузной В-крупноклеточной лимфосаркомой. Терапевтический архив. 2011;83(7):5–10.

    [Magomedova AU, Kravchenko SK, Kremenetskaya AM, et al. Nine-year experience in the treatment of patients with diffuse large B-cell lymphosarcoma. Terapevticheskii arkhiv. 2011;83(7):5–10. (In Russ)]

  6. Гаврилина О.А., Габеева Н.Г., Морозова А.К. и др. Роль высокодозной химиотерапии и трансплантации аутологичных стволовых клеток крови у пациентов с диффузной В-крупноклеточной лимфомой. Терапевтический архив. 2013;85(7):90–7.

    [Gavrilina OA, Gabeeva NG, Morozova AK, et al. Role of high-dose chemotherapy and autologous blood stem cell transplantation in patients with diffuse large B-cell lymphoma. Terapevticheskii arkhiv. 2013;85(7):90–7. (In Russ)]

  7. Габеева Н.Г., Королева Д.А., Беляева А.В. и др. Диффузная В-крупноклеточная лимфома с сочетанной реаранжировкой генов c-MYC и BCL6 с первичным поражением кожи: собственное наблюдение и обзор литературы. Терапевтический архив. 2017;89(7):85–92.

    [Gabeeva NG, Koroleva DA, Belyaeva AV, et al. Diffuse large B-cell lymphoma with concomitant c-MYC and BCL6 gene rearrangements with primary skin involvement: A case report and a review of literature. Terapevticheskii arkhiv. 2017;89(7):85–92. (In Russ)]

  8. Martelli M, Ferreri AJ, Agostinelli C, et al. Diffuse large B-cell lymphoma. Crit Rev Oncol Hematol. 2013;87(2):146–71. doi: 10.1016/j.critrevonc.2012.12.009.

  9. Cohen M, Vistarop AG, Huaman F, et al. Epstein-Barr virus lytic cycle involvement in diffuse large B cell lymphoma. Hematol Oncol. 2017;36(1):98–103. doi: 10.1002/hon.2465.

  10. Lenz G, Wright G, Dave SS, et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med. 2008;359(22):2313–23. doi: 10.1056/NEJMoa0802885.

  11. 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(17): 9991–6. doi: 10.1073/pnas.1732008100.

  12. Skryabin KG, Prokhortchouk EB, Mazur AM, et al. Combining Two Technologies for Full Genome Sequencing of Human. Acta Nat. 2009;1(3):102–7.

  13. Artemov AV, Boulygina ES, Tsygankova SV, et al. Study of Alzheimer Family Case Reveals Hemochromotosis-Associated HFE Mutation. Hum Gen Var. 2014;1(1):14004. doi: 10.1038/hgv.2014.4.

  14. Scelo G, Riazalhosseini Y, Greger L, et al. Variation in genomic landscape of clear cell renal cell carcinoma across Europe. Nat Commun. 2014;5(1):5135. doi: 10.1038/ncomms6135.

  15. Shipp MA, Ross KN, Tamayo P, et al. Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat Med. 2002;8(1):68–74. doi: 10.1038/nm0102-68.

  16. Scherer F, Kurtz DM, Newman AM, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8(364):364ra155. doi: 10.1126/scitranslmed.aai8545.

  17. Lawrie CH, Soneji S, Marafioti T, et al. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int J Cancer. 2007;121(5):1156–61. doi: 10.1002/ijc.22800.

  18. Malumbres R, Sarosiek KA, Cubedo E, et al. Differentiation stage–specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas. Blood. 2009;113(16):3754–64. doi: 10.1182/blood-2008-10-184077.

  19. Zhu D, Fang C, Li X, et al. Predictive analysis of long non-coding RNA expression profiles in diffuse large B-cell lymphoma. Oncotarget. 2017;8(14):23228–36. doi: 10.18632/oncotarget.15571.

  20. Peng W, Fan H, Wu G, et al. Upregulation of long noncoding RNA PEG10 associates with poor prognosis in diffuse large B cell lymphoma with facilitating tumorigenicity. Clin Exp Med. 2016;16(2):177–82. doi: 10.1007/s10238-015-0350-9.

  21. Peng W, Feng J. Long noncoding RNA LUNAR1 associates with cell proliferation and predicts a poor prognosis in diffuse large B-cell lymphoma. Biomed Pharmacother. 2016;77:65–71. doi: 10.1016/j.biopha.2015.12.001.

  22. Peng W, Wu J, Feng J. Long noncoding RNA HULC predicts poor clinical outcome and represents pro-oncogenic activity in diffuse large B-cell lymphoma. Biomed Pharmacother. 2016;79:188–93. doi: 10.1016/j.biopha.2016.02.032.

  23. Yan Y, Han J, Li Z, et al. Elevated RNA expression of long non-coding HOTAIR promotes cell proliferation and predicts a poor prognosis in patients with diffuse large B cell lymphoma. Mol Med Rep. 2016;13(6):5125–31. doi: 10.3892/mmr.2016.5190.

  24. Li L-J, Chai Y, Guo X-J, et al. The effects of the long non-coding RNA MALAT-1 regulated autophagy-related signaling pathway on chemotherapy resistance in diffuse large B-cell lymphoma. Biomed Pharmacother. 2017;89:939–48. doi: 10.1016/j.biopha.2017.02.011.

  25. Sun J, Cheng L, Shi H, et al. A potential panel of six-long non-coding RNA signature to improve survival prediction of diffuse large-B-cell lymphoma. Sci Rep. 2016;6(1):27842. doi: 10.1038/srep27842.

  26. Verma A, Jiang Y, Du W, et al. Transcriptome sequencing reveals thousands of novel long non-coding RNAs in B cell lymphoma. Gen Med. 2015;7(1):110. doi: 10.1186/s13073-015-0230-7.

  27. Gutierrez-Garcia G, Cardesa-Salzmann T, Climent F, 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):4836–43. doi: 10.1182/blood-2010-12-322362.

  28. Schuetz JM, Johnson NA, Morin RD, et al. BCL2 mutations in diffuse large B-cell lymphoma. Leukemia. 2012;26(6):1383–90. doi: 10.1038/leu.2011.378.

  29. Greenough A, Moffitt A, Jima D, et al. Strand-Specific Total RNA Sequencing Establishes the Complete Transcriptome and Alternative Splicing Repertoire in Diffuse Large B Cell Lymphoma. Blood. 2014;124(21):864.

  30. Park HY, Lee SB, Yoo HY, et al. Whole-Exome and Transcriptome Sequencing of Refractory Diffuse Large B-Cell Lymphoma. Oncotarget. 2016;7(52): 86433–45. doi: 10.18632/oncotarget.13239.

  31. Dekker JD, Park D, Shaffer AL, et al. Subtype-Specific Addiction of the Activated B-Cell Subset of Diffuse Large B-Cell Lymphoma to FOXP1. Proc Natl Acad Sci USA. 2016;113(5):E577–86. doi: 10.1073/pnas.1524677113.

  32. Reddy A, Zhang J, Davis NS, et al. Genetic and Functional Drivers of Diffuse Large B Cell Lymphoma. Cell. 2017;171(2):481–94.e15. doi: 10.1016/j.cell.2017.09.027.

  33. Saez AI, Saez AJ, Artiga MJ, et al. Building an outcome predictor model for diffuse large B-cell lymphoma. Am J Pathol. 2004;164(2):613–22. doi: 10.1016/S0002-9440(10)63150-1.

  34. Campo E. MYC in DLBCL: partners matter. Blood. 2015;126(22):2439–40. doi: 10.1182/blood-2015-10-671362.

  35. Schmitz R, Wright GW, Huang DW, et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N Engl J Med. 2018;378(15):1396–407. doi: 10.1056/NEJMoa1801445.

  36. 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):2919–28. doi: 10.1158/1078-0432.CCR-15-2305.

  37. Lohr JG, Stojanov P, Lawrence MS, et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc Natl Acad Sci USA. 2012;109(10):3879–84. doi: 10.1073/pnas.1121343109.

  38. 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):1256–65. doi: 10.1182/blood-2013-02-483727.

  39. Pasqualucci L, Trifonov V, Fabbri G, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet. 2011;43(9):830–7. doi: 10.1038/ng.892.

  40. Roschewski M, Dunleavy K, Pittaluga S, et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16(5):541–9. doi: 10.1016/S1470-2045(15)70106-3.

  41. Yeh P, Hunter T, Sinha D, Ftouni S, et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nat Commun. 2017;8:14756. doi: 10.1038/ncomms14756.

  42. Khare D, Goldschmidt N, Bardugo A, et al. Plasma microRNA profiling: Exploring better biomarkers for lymphoma surveillance. PLoS One. 2017;12(11):e0187722. doi: 10.1371/journal.pone.0187722.

  43. Meng Y, Quan L, Liu A. Identification of key microRNAs associated with diffuse large B-cell lymphoma by analyzing serum microRNA expressions. Gene. 2018;642:205–11. doi: 10.1016/j.gene.2017.11.022.

  44. Kurtz DM, Green MR, Bratman SV, et al. Noninvasive Monitoring of Diffuse Large B-Cell Lymphoma by Immunoglobulin High-Throughput Sequencing. Blood. 2015;125(24):3679–87. doi: 10.1182/blood-2015-03-635169.

  45. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359(6378):926–30. doi: 10.1126/science.aar3247.

  46. Shaknovich R, Melnick A. Epigenetics and B-Cell Lymphoma. Curr Opin Hematol. 2011;18(4):293–9. doi: 10.1097/MOH.0b013e32834788cf.

  47. Shaknovich R, Geng H, Johnson NA, et al. DNA methylation signatures define molecular subtypes of diffuse large B-cell lymphoma. Blood. 2010;116(20):e81–9. doi: 10.1182/blood-2010-05-285320.

  48. Lai AY, Fatemi M, Dhasarathy A, et al. DNA methylation prevents CTCF-mediated silencing of the oncogene BCL6 in B cell lymphomas. J Exp Med. 2010;207(9):1939–50. doi: 10.1084/jem.20100204.

  49. Kristensen LS, Hansen JW, Kristensen SS, et al. Aberrant Methylation of Cell-Free Circulating DNA in Plasma Predicts Poor Outcome in Diffuse Large B Cell Lymphoma. Clin Epigen. 2016;8(1):5. doi: 10.1186/s13148-016-0261-y.

  50. Wedge E, Hansen JW, Garde C, et al. Global hypomethylation is an independent prognostic factor in diffuse large B cell lymphoma. Am J Hematol. 2017;92(7):689–94. doi: 10.1002/ajh.24751.

  51. Krajnovic M, Jovanovic MP, Mihaljevic B, et al. Hypermethylation of p15 Gene in Diffuse – Large B‐Cell Lymphoma: Association with Less Aggressiveness of the Disease. Clin Transl Sci. 2014;7(5):384–90. doi: 10.1111/cts.12162.

  52. Chambwe N, Kormaksson M, Geng H, et al. Variability in DNA methylation defines novel epigenetic subgroups of DLBCL associated with different clinical outcomes. Blood. 2014;123(11):1699–708. doi: 10.1182/blood-2013-07-509885.

  53. Clozel T, Yang S, Elstrom RL, et al. Mechanism-based epigenetic chemosensitization therapy of diffuse large B-cell lymphoma. Cancer Discov. 2013;3(9):1002–19. doi: 10.1158/2159-8290.CD-13-0117.

  54. Pan H, Jiang Y, Boi M, et al. Epigenomic evolution in diffuse large B-cell lymphomas. Nat Commun. 2015;6(1):6921. doi: 10.1038/ncomms7921.

  55. Jing L, Su L, Ring BZ. Ethnic Background and Genetic Variation in the Evaluation of Cancer Risk: A Systematic Review. PLoS ONE. 2014;9(6):e97522. doi: 10.1371/journal.pone.0097522.

  56. Li Y, Wang Y, Wang Z, et al. Racial Differences in Three Major NHL Subtypes: Descriptive Epidemiology. Cancer Epidemiol. 2015;39(1):8–13. doi: 10.1016/j.canep.2014.12.001.

 

Cytogenetic and Molecular Genetic Prognostic Factors of Acute Lymphoblastic Leukemias

AV Misyurin

NN Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Andrei Vital’evich Misyurin, PhD, 24 Kashirskoe sh., Moscow, Russian Federation, 115478; e-mail: and@genetechnology.ru

For citation: Misyurin AV. Cytogenetic and Molecular Genetic Prognostic Factors of Acute Lymphoblastic Leukemias. Clinical oncohematology. 2017;10(3):317–23 (In Russ).

DOI: 10.21320/2500-2139-2017-10-3-317-323


ABSTRACT

This review presents characteristic and reproducible chromosome rearrangements in acute lymphoblastic leukemia (ALL), which can be detected with a standard cytogenetic research (G-bands staining) or by FISH. More subtle genetic changes, inaccessible to the observation of cytogeneticists, are detected with the help of modern methods of molecular biological diagnosis. The prognostic value of cytogenetic and molecular genetic markers of ALL is shown in this article. A minimal set of clinically relevant molecular markers is presented, which it is advisable to investigate with ALL.

Keywords: acute lymphoblastic leukemia, chromosomal aberration, chimeric oncogene, gene expression, gene mutation.

Received: December 3, 2016

Accepted: March 8, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. 776 с.
    [Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. 776 p. (In Russ)]
  2. Inaba H, Greaves M, Mullighan CG. Acute lymphoblastic leukaemia. Lancet. 2013;381(9881):1943–55. doi: 10.1016/S0140-6736(12)62187-4.
  3. Shago M. Recurrent Cytogenetic Abnormalities in Acute Lymphoblastic Leukemia. Meth Mol Biol. 2017;1541:257–78. doi: 10.1007/978-1-4939-6703-2_21.
  4. Deshpande PA, Srivastava VM, Mani S, et al. Atypical BCR-ABL11 fusion transcripts in adult B-acute lymphoblastic leukemia, including a novel fusion transcript-e8a1. Leuk Lymphoma. 2016;57(10):2481–4. doi: 10.3109/10428194.2016.1151512.
  5. McGregor S, McNeer J, Gurbuxani S. Beyond the 2008 World Health Organization classification: the role of the hematopathology laboratory in the diagnosis and management of acute lymphoblastic leukemia. Semin Diagn Pathol. 2012;29(1):2–11.
  6. Zerbini MCN, Soares FA, Velloso EDRP, et al. World Health Organization classification of tumors of hematopoietic and lymphoid tissues, 2008: major changes from the 3rd edition. Revista da Associacao Medica Brasileira. 2011;57(1):6–73. doi: 10.1590/S0104-42302011000100019.
  7. Paulsson K, Johansson B. High hyperdiploid childhood acute lymphoblastic leukemia. Genes Chromos Cancer. 2009;48(8):637–60. doi: 10.1002/gcc.20671.
  8. Mrоzek K, Harper DP, Aplan PD. Cytogenetics and Molecular Genetics of Acute Lymphoblastic Leukemia. Hematol Oncol Clin North Am. 2009;23(5):1–20. doi: 10.1016/j.hoc.2009.07.001.
  9. Faderl S, Estrov Z. Residual disease in acute lymphoblastic leukemia of childhood: methods of detection and clinical relevance. Cyt Cell Mol Ther. 1998;4(2):73–85.
  10. Heerema NA, Raimondi SC, Anderson JR, et al. Specific extra chromosomes occur in a modal number dependent pattern in pediatric acute lymphoblastic leukemia. Genes Chromos Cancer. 2007;46(7):684–93. doi: 10.1002/gcc.20451.
  11. Woo JS, Alberti MO, Tirado CA. Childhood B-acute lymphoblastic leukemia: a genetic update. Exper Hematol Oncol. 2014;3(1):16. doi: 10.1186/2162-3619-3-16.
  12. Sutcliffe MJ, Shuster JJ, Sather HN, et al. High concordance from independent studies by the Children’s Cancer Group (CCG) and Pediatric Oncology Group (POG) associating favorABL1e prognosis with combined trisomies 4, 10, and 17 in children with NCI Standard-Risk B-precursor Acute Lymphoblastic Leukemia: a Children’s Oncology Group (COG) initiative. Leukemia. 2005;19(5):734–40. doi: 10.1038/sj.leu.2403673.
  13. Moorman AV, Richards SM, Martineau M, et al. Outcome heterogeneity in childhood high-hyperdiploid acute lymphoblastic leukemia. Blood. 2003;102(8):2756–62. doi: 10.1182/blood-2003-04-1128.
  14. Forestier E, Johansson B, Gustafsson G, et al. Prognostic impact of karyotypic findings in childhood acute lymphoblastic leukaemia: a Nordic series comparing two treatment periods. Br J Haematol. 2000;110(1):147–53.
  15. Raimondi SC, Pui CH, Hancock ML, et al. Heterogeneity of hyperdiploid (51-67) childhood acute lymphoblastic leukemia. Leukemia. 1996;10(2):213–24.
  16. Pullarkat V, Slovak ML, Kopecky KJ, et al. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood. 2008;111(5):2563–72. doi: 10.1182/blood-2007-10-116186.
  17. Oostlander AE, Meijer GA, Ylstra B. Microarray-based comparative genomic hybridization and its applications in human genetics. Clin Genet. 2004;66(6):488–95. doi: 10.1111/j.1399-0004.2004.00322.x.
  18. Rubnitz JE, Wichlan D, Devidas M, et al. Prospective analysis of TEL gene rearrangements in childhood acute lymphoblastic leukemia: a Children’s Oncology Group study. J Clin Oncol. 2008;26(13):2186–91. doi: 10.1200/JCO.2007.14.3552.
  19. Attarbaschi A, Mann G, Konig M, et al. Incidence and relevance of secondary chromosome abnormalities in childhood TEL/AML1+ acute lymphoblastic leukemia: an interphase FISH analysis. Leukemia. 2004;18(10):1611–6. doi: 10.1038/sj.leu.2403471.
  20. Pullarkat V, Slovak ML, Kopecky KJ, et al. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood. 2008;111(5):2563–72. doi: 10.1182/blood-2007-10-116186.
  21. Stock W. Advances in the treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia. Clin Adv Hematol Oncol. 2008;6(7):487–8.
  22. Fielding AK, Rowe JM, Richards SM, et al. Prospective outcome data on 267 unselected adult patients with Philadelphia-chromosome positive acute lymphoblastic leukemia confirms superiority of allogeneic transplantation over chemotherapy in the pre-imatinib era: results from the international ALL trial MRC KALLXII/ECOG2993. Blood. 2009;113(19):4489–96. doi: 10.1182/blood-2009-01-199380.
  23. Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a metaanalysis. Leukemia. 2005;19(8):1345–9. doi: 10.1038/sj.leu.2403838.
  24. Moorman AV, Richards SM, Robinson HM, et al. Prognosis of children with acute lymphoblastic leukemia (ALL) and intrachromosomal amplification of chromosome 21 (iAMP21). Blood. 2007;109(6):2327–30. doi: 10.1182/blood-2006-08-040436.
  25. Heerema NA, Harbott J, Galimberti S, et al. Secondary cytogenetic aberrations in childhood Philadelphia chromosome positive acute lymphoblastic leukemia are nonrandom and may be associated with outcome. Leukemia. 2004;18(4):693–702. doi: 10.1038/sj.leu.2403324.
  26. Wetzler M, Talpaz M, Estrov Z, Kurzrock R. CML: mechanisms of disease initiation and progression. Leuk Lymphoma. 1993;11(Suppl 1):47–50. doi: 10.3109/10428199309047863.
  27. Chessells JM, Swansbury GJ, Reeves B, et al. Cytogenetics and prognosis in childhood lymphoblastic leukaemia: results of MRC UKALL X. Br J Haematol. 1997;99(1):93–100.
  28. Chessells JM, Harrison CJ, Kempski H, et al. Clinical features, cytogenetics and outcome in acute lymphoblastic and myeloid leukaemia of infancy: report from the MRC Childhood Leukaemia working party. Leukemia. 2002;16(5):776–84. doi: 10.1038/sj.leu.2402468.
  29. Moorman AV, Raimondi SC, Pui CH, et al. No prognostic effect of additional chromosomal abnormalities in children with acute lymphoblastic leukemia and 11q23 abnormalities. Leukemia. 2005;19(4):557–63. doi: 10.1038/sj.leu.2403695.
  30. Pui CH, Sandlund JT, Pei D, et al. Results of therapy for acute lymphoblastic leukemia in black and white children. JAMA. 2003;290(15):2001–7. doi: 10.1001/jama.290.15.2001.
  31. Pui C-H, Chessells JM, Camitta B, et al. Clinical heterogeneity in childhood acute lymphoblastic leukemia with 11q23 rearrangements. Leukemia. 2003;17(4):700–6. doi: 10.1038/sj.leu.2402883.
  32. Jeha S, Pei D, Raimondi SC, et al. Increased risk for CNS relapse in pre-B cell leukemia with the t(1;19)/TCF3-PBX1. Leukemia. 2009;23(8):1406–9. doi: 10.1038/leu.2009.42.
  33. Mrozek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;35:365–77. doi: 10.1053/j.seminoncol.2008.04.007.
  34. Wetzler M, Dodge RK, Mrozek K, et al. Prospective karyotype analysis in adult acute lymphoblastic leukemia: the cancer and leukemia Group B experience. Blood. 1999;93(11):3983–93.
  35. Bernard OA, Busson-LeConiat M, Ballerini P, et al. A new recurrent and specific cryptic translocation, t(5;14)(q35;q32), is associated with expression of the Hox11L2 gene in T acute lymphoblastic leukemia. Leukemia. 2001;15(10):1495–504. doi: 10.1038/sj.leu.2402249.
  36. Graux C, Stevens-Kroef M, Lafage M, et al. Heterogeneous patterns of amplification of the NUP214-ABL11 fusion gene in T-cell acute lymphoblastic leukemia. Leukemia. 2009;23(1):125–33. doi: 10.1038/leu.2008.278.
  37. Quintas-Cardama A, Tong W, Manshouri T, et al. Activity of tyrosine kinase inhibitors against human NUP214-ABL11-positive T cell malignancies. Leukemia. 2008;22(6):1117–24. doi: 10.1038/leu.2008.80.
  38. Krawczyk J, Haslam K, Lynam P, et al. No prognostic impact of P2RY8-CRLF2 fusion in intermediate cytogenetic risk childhood B-cell acute lymphoblastic leukaemia. Br J Haematol. 2013;160(4):555–6. doi: 10.1111/bjh.12130.
  39. Hoelzer D. Personalized medicine in adult acute lymphoblastic leukemia. Haematologica. 2015;100(7):855–8. doi: 10.3324/haematol.2015.127837.
  40. Tsuzuki S, Taguchi O, Seto M. Promotion and maintenance of leukemia by ERG. Blood. 2011;117(14):3858–68. doi: 10.1182/blood-2010-11-320515.
  41. Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360(5):470–80. doi: 10.1056/NEJMoa0808253.
  42. Mullighan CG, Miller CB, Radtke I, et al. BCR-ABL11 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453(7191):110–4. doi: 10.1038/nature06866.
  43. Yoda A, Yoda Y, Chiaretti S, et al. Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA. 2010;107(1):252–7. doi: 10.1073/pnas.0911726107.
  44. Mullighan CG, Zhang J, Kasper LH, et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature. 2011;471(7337):235–9. doi: 10.1038/nature09727.
  45. Van Vlierberghe P, Palomero T, Khiabanian H, et al. PHF6 mutations in T-cell acute lymphoblastic leukemia. Nat Genet. 2010;42(4):338–42. doi: 10.1038/ng.542.
  46. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009;10(2):147–56. doi: 10.1016/S1470-2045(08)70314-0.
  47. Weng AP, Ferrando AA, Lee W, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004;306(5694):269–71. doi: 10.1126/science.1102160.
  48. Гапонова Т.В., Менделеева Л.П., Мисюрин А.В. и др. Экспрессия опухолеассоциированных генов PRAME, WT1 и XIAP у больных множественной миеломой. Онкогематология. 2009;2:52–5.
    [Gaponova TV, Mendeleeva LP, Misyurin AV, et al. PRAME, WT1 and XIAP tumor-associated genes expression in multiple myeloma patients. Onkogematologiya. 2009;2:52–5. (In Russ)]
  49. Абраменко И.В., Белоус Н.И., Крячок И.А. и др. Экспрессия гена PRAME при множественной миеломе. Терапевтический архив. 2004;76(7):77–81.
    [Abramenko IV, Belous NI, Kryachok IA, et al. PRAME gene expression in multiple myeloma. Terapevticheskii arkhiv. 2004;76(7):77–81. (In Russ)]
  50. Мисюрин В.А. Аутосомные раково-тестикулярные гены. Российский биотерапевтический журнал. 2014;13(3):77–82.
    [Misyurin VA. Autosomal cancer-testis genes. Rossiiskii bioterapevticheskii zhurnal. 2014;13(3):77–82. (In Russ)]
  51. Мисюрин А.В. Основы молекулярной диагностики онкогематологических заболеваний. Российский биотерапевтический журнал. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24.
    [Misyurin AV. Essentials of the molecular diagnosis of oncohematological diseases. Rossiysky bioterapevtichesky zhurnal. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24. (In Russ)]

 

Cytogenetic and Molecular Genetic Prognostic Factors of Acute Myeloid Leukemia

AV Misyurin

NN Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Andrei Vital’evich Misyurin, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: and@genetechnology.ru

For citation: Misyurin AV. Cytogenetic and Molecular Genetic Prognostic Factors of Acute Myeloid Leukemia. Clinical oncohematology. 2017;10(2):227–34 (In Russ).

DOI: 10.21320/2500-2139-2017-10-2-227-234


ABSTRACT

The review presents data on the diagnostic and prognostic value of cytogenetic and molecular genetic markers of acute myeloid leukemia (AML). It demonstrates that some cases, different types of AML subdivided on the basis of clinical and morphological characteristics earlier may be distinguished based on identification of specific genetic and chromosomal defects. However, some repeated chromosomal abnormalities may be detected in AML patients that may be assigned to different variants based in clinical and morphocytochemical signs. At present, it is widely accepted that changes in the karyotype are the key prognostic factors which are more important than criteria based on morphological and cytochemical signs. Therefore, the risk-adaptive therapy of AML should be chosen based on the cytogenetic test findings. The review contains a section discussing gene mutations known to date that may affect the AML treatment outcome.

Keywords: AML, chromosomal aberration, chimeric oncogene, gene expression, gene mutation.

Received: September 16, 2016

Accepted: January 3, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. 776 с.
    [Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. 776 p. (In Russ)]
  2. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  3. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937–51. doi: 10.1182/blood-2009-03-209262.
  4. Zerbini MCN, Soares FA, Velloso EDRP, et al. World Health Organization classification of tumors of hematopoietic and lymphoid tissues, 2008: major changes from the 3rd edition. Revista da Associacao Medica Brasileira. 2011;57(1): 6–73. doi: 10.1590/S0104-42302011000100019.
  5. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br J Haematol. 1976;33(4):451–8. doi: 10.1111/j.1365-2141.1976.tb03563.
  6. Kuhnl A, Grimwade D. Molecular markers in acute myeloid leukaemia. Int J Hematol. 2012;96(2):153–63. doi: 10.1007/s12185-012-1123-9.
  7. Burnett AK, Wheatley K, Goldstone AH, et al. The value of allogeneic bone marrow transplant in patients with acute myeloid leukaemia at differing risk of relapse: results of the UK MRC AML 10 trial. Br J Haematol. 2002;118(2):385–400. doi: 10.1046/j.1365-2141.2002.03724.x.
  8. Cornelissen JJ, van Putten WL, Verdonck LF, et al. Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom? Blood. 2007;109(9):3658–66. doi: 10.1182/blood-2006-06-025627.
  9. Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood. 2000;96(13):4075–83.
  10. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116(3):354–65. doi: 10.1182/blood-2009-11-254441.
  11. Blum W, Mrozek K, Ruppert AS, et al. Adult de novo acute myeloid leukemia with t(6;11)(q27;q23): results from Cancer and Leukemia Group B Study 8461 and review of the literature. Cancer. 2005;103(6):1316. doi: 10.1002/cncr.20931
  12. Krauter J, Wagner K, Schafer I, et al. Prognostic factors in adult patients up to 60 years old with acute myeloid leukemia and translocations of chromosome band 11q23: individual patient data-based meta-analysis of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol. 2009;27(18):3000–6. doi: 10.1200/jco.2008.16.7981.
  13. von Neuhoff C, Reinhardt D, Sander A, et al. Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol. 2010;28(16):2682–9. doi: 10.1200/JCO.2009.25.6321.
  14. Rucker FG, Bullinger L, Schwaenen C, et al. Disclosure of candidate genes in acute myeloid leukemia with complex karyotypes using microarray-based molecular characterization. J Clin Oncol. 2006;24(24):3887–94. doi: 10.1200/jco.2005.04.5450.
  15. Mrozek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;35(4):365–77. doi: 10.1053/j.seminoncol.2008.04.007.
  16. Breems DA, Van Putten WL, De Greef GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol. 2008;26(29):4791–7. doi: 10.1200/JCO.2008.16.0259.
  17. Smith ML, Hills RK, Grimwade D. Independent prognostic variables in acute myeloid leukaemia. Blood Rev. 2011;25(1):39–51. doi: 10.1016/j.blre.2010.10.002.
  18. Delhommeau F, Dupont S, Della Valle V, et al. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360(22):2289–301. doi: 10.1056/NEJMoa0810069.
  19. Metzeler KH, Maharry K, Radmacher MD, et al. TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2011;29(10):1373–81. doi: 10.1200/JCO.2010.32.7742.
  20. Chou WC, Chou SC, Liu CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood. 2011;118(14):3803–10. doi: 10.1182/blood-2011-02-339747.
  21. Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361(11):1058–66.
  22. Thol F, Damm F, Wagner K, et al. Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia. Blood. 2010(4);116:614–6. doi: 10.1182/blood-2010-03-272146.
  23. Chou WC, Hou HA, Chen CY, et al. Distinct clinical and biologic characteristics in adult acute myeloid leukemia bearing the isocitrate dehydrogenase 1 mutation. Blood. 2010;115(14):2749–54. doi: 10.1182/blood-2009-11-253070.
  24. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/jco.2009.27.3730.
  25. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol. 2010;28(22):3636–43. doi: 10.1200/jco.2010.28.3762.
  26. Schnittger S, Haferlach C, Ulke M, et al. IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status. Blood. 2010;116(25):5486–96. doi: 10.1182/blood-2010-02-267955.
  27. Ravandi F, Patel K, Luthra R, et al. Prognostic significance of alterations in IDH enzyme isoforms in patients with AML treated with high-dose cytarabine and idarubicin. Cancer. 2012;118(10):2665–73. doi: 10.1002/cncr.26580.
  28. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33. doi: 10.1056/NEJMoa1005143.
  29. Thol F, Damm F, Ludeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol. 2011;29:2889–96. doi: 10.1200/JCO.2011.35.4894.
  30. Shen Y, Zhu YM, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood. 2011;118(20):5593–603. doi: 10.1182/blood-2011-03-343988.
  31. Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood. 2011(2);119:559–68. doi: 10.1182/blood-2011-07-369934.
  32. Renneville A, Boissel N, Nibourel O, et al. Prognostic significance of DNA methyltransferase 3A mutations in cytogenetically normal acute myeloid leukemia: a study by the Acute Leukemia French Association. Leukemia. 2012;26(6):1247–54. doi: 10.1038/leu.2011.382.
  33. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2012;30(7):742–50. doi: 10.1200/jco.2011.39.2092.
  34. Markova J, Michkova P, Burckova K, et al. Prognostic impact of DNMT3A mutations in patients with intermediate cytogenetic risk profile acute myeloid leukemia. Eur J Haematol. 2012;88(2):128–35. doi: 10.1111/j.1600-0609.2011.01716.x.
  35. King-Underwood L, Renshaw J, Pritchard-Jones K. Mutations in the Wilms’ tumor gene WT1 in leukemias. Blood. 1996;87(6):2171–9.
  36. Virappane P, Gale R, Hills R, et al. Mutation of the Wilms’ tumor 1 gene is a poor prognostic factor associated with chemotherapy resistance in normal karyotype acute myeloid leukemia: the United Kingdom Medical Research Council Adult Leukaemia Working Party. J Clin Oncol. 2008;26(33):5429–35. doi: 10.1200/jco.2008.16.0333.
  37. Paschka P, Marcucci G, Ruppert AS, et al. Wilms’ tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study. J Clin Oncol. 2008;26(28):4595–602. doi: 10.1200/JCO.2007.15.2058.
  38. Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia. 2006;20(6):965–70. doi: 10.1038/sj.leu.2404188.
  39. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24(24):3904–11. doi: 10.1200/JCO.2006.06.9500.
  40. Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood. 2006;107(9):3463–8. doi: 10.1182/blood-2005-09-3640.
  41. Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107(5):1791–9. doi: 10.1182/blood-2005-04-1466.
  42. Gelsi-Boyer V, Trouplin V, Adelaide J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800. doi: 10.1111/j.1365-2141.2009.07697.x.
  43. Chou WC, Huang HH, Hou HA, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood. 2010;116(20):4086–94. doi: 10.1182/blood-2010-05-283291.
  44. Metzeler KH, Becker H, Maharry K, et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category. Blood. 2011;118(26):6920–9. doi: 10.1182/blood-2011-08-368225.
  45. Pratcorona M, Abbas S, Sanders MA, et al. Acquired mutations in ASXL1 in acute myeloid leukemia: prevalence and prognostic value. Haematologica. 2012;97(3):388–92. doi: 10.3324/haematol.2011.051532.
  46. Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/NEJMoa1112304.
  47. Grossmann V, Tiacci E, Holmes AB, et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood. 2011;118(23):6153–63. doi: 10.1182/blood-2011-07-365320.
  48. Li M, Collins R, Jiao Y, et al. Somatic mutations in the transcriptional corepressor gene BCORL1 in adult acute myelogenous leukemia. Blood. 2011;118(22):5914–7. doi: 10.1182/blood-2011-05-356204.
  49. Van Vlierberghe P, Patel J, Abdel-Wahab O, et al. PHF6 mutations in adult acute myeloid leukemia. Leukemia. 2011;25(1):130–4. doi: 10.1038/leu.2010.247.
  50. Mano H. Stratification of acute myeloid leukemia based on gene expression profiles. Int J Hematol. 2004;80(5):389–94. doi: 10.1532/ijh97.04111.
  51. Marcucci G, Mrozek K, Radmacher MD, et al. The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood. 2011;117(4):1121–9. doi: 10.1182/blood-2010-09-191312.
  52. Smith ML, Hills RK, Grimwade D. Independent prognostic variables in acute myeloid leukaemia. Blood Rev. 2011;25(1):39–51. doi: 10.1016/j.blre.2010.10.002.
  53. Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352(3):254–66. doi: 10.1056/NEJMoa041974.
  54. Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106(12):3740–6. doi: 10.1182/blood-2005-05-2164.
  55. Thiede C, Creutzig E, Illmer T, et al. Rapid and sensitive typing of NPM1 mutations using LNA-mediated PCR clamping. Leukemia. 2006;20(10):1897–9. doi: 10.1038/sj.leu.2404367.
  56. Schlenk RF, Dohner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–18. doi: 10.1056/NEJMoa074306.
  57. Green CL, Koo KK, Hills RK, et al. Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations. J Clin Oncol. 2010;28(16):2739–47. doi: 10.1200/JCO.2009.26.2501.
  58. Grisendi S, Mecucci C, Falini B, et al. Nucleophosmin and cancer. Nat Rev Cancer. 2006;6(7):493–505. doi: 10.1038/nrc1885.
  59. Freeman SD, Jovanovic JV, Grimwade D. Development of minimal residual disease-directed therapy in acute myeloid leukemia. Semin Oncol. 2008;35(4):388–400. doi: 10.1053/j.seminoncol.2008.04.009.
  60. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10(12):1911–8.
  61. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9. doi: 10.1182/blood.V98.6.1752.
  62. Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99(12):4326–35. doi: 10.1182/blood.V99.12.4326.
  63. Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis. Leukemia. 2005;19(8):1345–9. doi: 10.1038/sj.leu.2403838.
  64. Mead AJ, Linch DC, Hills RK, et al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients. Blood. 2007;110(4):1262–70. doi: 10.1182/blood-2006-04-015826.
  65. Bacher U, Haferlach C, Kern W, et al. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters—an analysis of 3082 patients. Blood. 2008;111(5):2527–37. doi: 10.1182/blood-2007-05-091215.
  66. Whitman SP, Ruppert AS, Radmacher MD, et al. FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood. 2008;111(3):1552–9. doi: 10.1182/blood-2007-08-107946.
  67. Gale RE, Hills R, Pizzey AR, et al. Relationship between FLT3 mutation status, biologic characteristics, and response to targeted therapy in acute promyelocytic leukemia. Blood. 2005;106(12):3768–76. doi: 10.1182/blood-2005-04-1746.
  68. Souza Melo CP, Campos CB, Dutra AP, et al. Correlation between FLT3-ITD status and clinical, cellular and molecular profiles in promyelocytic acute leukemias. Leuk Res. 2015;39(2):131–7. doi: 10.1016/j.leukres.2014.11.010.
  69. Cicconi L, Divona M, Ciardi C, et al. PML-RARα kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy. Leukemia. 2016;30(10):1987–92. doi: 10.1038/leu.2016.122.
  70. Radomska HS, Huettner CS, Zhang P, et al. CCAAT/enhancer binding protein alpha is a regulatory switch sufficient for induction of granulocytic development from bipotential myeloid progenitors. Mol Cell Biol. 1998;18(7):4301–14. doi: 10.1128/mcb.18.7.4301.
  71. Pabst T, Mueller BU, Zhang P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia. Nat Genet. 2001;27(3):263–70. doi: 10.1038/85820.
  72. Smith ML, Cavenagh JD, Lister TA, et al. Mutation of CEBPA in familial acute myeloid leukemia. N Engl J Med. 2004;351(23):2403–7. doi: 10.1056/NEJMoa041331.
  73. Pabst T, Eyholzer M, Fos J, et al. Heterogeneity within AML with CEBPA mutations; only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis. Br J Cancer. 2009;100(8):1343–6. doi: 10.1038/sj.bjc.6604977.
  74. Kirstetter P, Schuster MB, Bereshchenko O, et al. Modeling of C/EBPalpha mutant acute myeloid leukemia reveals a common expression signature of committed myeloid leukemia-initiating cells. Cancer Cell. 2008;13(4):299–310. doi: 10.1016/j.ccr.2008.02.008.
  75. Wouters BJ, Lowenberg B, Erpelinck-Verschueren CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood. 2009;113(13):3088–91. doi: 10.1182/blood-2008-09-179895.
  76. Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood. 2011;117(8):2469–75. doi: 10.1182/blood-2010-09-307280.
  77. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.
  78. Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 2011;333(6047):1300–3. doi: 10.1126/science.1210597.
  79. Abdel-Wahab O, Mullally A, Hedvat C, et al. Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood. 2009;114(1):144–7. doi: 10.1182/blood-2009-03-210039.
  80. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67. doi: 10.1016/j.ccr.2010.11.015.
  81. Ko M, Huang Y, Jankowska AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010;468(7325):839–43. doi: 10.1038/nature09586.
  82. Langemeijer SM, Kuiper RP, Berends M, et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet. 2009;41(7):838–42. doi: 10.1038/ng.391.
  83. Jankowska AM, Szpurka H, Tiu RV, et al. Loss of heterozygosity 4q24 and TET2 mutations associated with myelodysplastic/myeloproliferative neoplasms. Blood. 2009;113(25):6403–10. doi: 10.1182/blood-2009-02-205690.
  84. Gaidzik VI, Paschka P, Spath D, et al. TET2 mutations in acute myeloid leukemia (AML): results from a comprehensive genetic and clinical analysis of the AML Study Group. J Clin Oncol. 2012;30(12):1350–7. doi: 10.1200/JCO.2011.39.2886.
  85. Ward PS, Patel J, Wise DR, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010;17(3):225–34. doi: 10.1016/j.ccr.2010.01.020.
  86. Gross S, Cairns RA, Minden MD, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010;207(2):339–44. doi: 10.1084/jem.20092506.
  87. Sanz MA, Grimwade D, Tallman MS, et al. Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2009;113(9):1875–91. doi: 10.1182/blood-2008-04-150250.
  88. Diehl F, Rossig L, Zeiher AM, et al. The histone methyltransferase MLL is an upstream regulator of endothelial-cell sprout formation. Blood. 2007;109(4):1472–8. doi: 10.1182/blood-2006-08-039651.
  89. Li Y, Han J, Zhang Y, et al. Structural basis for activity regulation of MLL family methyltransferases. Nature. 2016;530(7591):447–52. doi: 10.1038/nature16952.
  90. Bower M, Parry P, Carter M, et al. Prevalence and clinical correlations of MLL gene rearrangements in AML-M4/5. Blood. 1994;84(11):3776–80.
  91. Schichman SA, Caligiuri MA, Strout MP, et al. ALL-1 tandem duplication in acute myeloid leukemia with a normal karyotype involves homologous recombination between Alu elements. Cancer Res. 1994;54(16):4277–80.
  92. Caligiuri MA, Schichman SA, Strout MP, et al. Molecular rearrangement of the ALL-1 gene in acute myeloid leukemia without cytogenetic evidence of 11q23 chromosomal translocations. Cancer Res. 1994;54(2):370–3.
  93. Park JP, Ladd SL, Ely P, et al. Amplification of the MLL region in acute myeloid leukemia. Cancer Genet Cytogenet. 2000;121(2):198–205. doi: 10.1016/S0165-4608(00)00256-9.
  94. Schnittger S, Kinkelin U, Schoch C, et al. Screening for MLL tandem duplication in 387 unselected patients with AML identify a prognostically unfavorable subset of AML. Leukemia. 2000;14(5):796–804. doi: 10.1038/sj.leu.2401773.
  95. Slovak ML, Traweek ST, Willman CL, et al. Trisomy 11: an association with stem/progenitor cell immunophenotype. Br J Haematol. 1995;90(2):266–73. doi: 10.1111/j.1365-2141.1995.tb05146.x.
  96. Strout MP, Marcucci G, Bloomfield CD, et al. The partial tandem duplication of ALL1 (MLL) is consistently generated by Alu-mediated homologous recombination in acute myeloid leukemia. Proc Natl Acad Sci USA. 1998;95(5):2390–5. doi: 10.1073/pnas.95.5.2390.
  97. Klymenko S, Bebeshko V, Bazyka D, et al. AML1 gene rearrangements and mutations in radiation-associated acute myeloid leukemia and myelodysplastic syndromes. J Rad Res. 2005;46(2):249–55. doi: 10.1269/jrr.46.249.
  98. Мисюрин В.А., Лукина А.Е., Мисюрин А.В. и др. Особенности соотношения уровней экспрессии генов PRAME и PML/RARA в дебюте острого промиелоцитарного лейкоза. Российский биотерапевтический журнал. 2014;13(1):9–16.
    [Misyurin VA, Lukina AE, Misyurin AV. A ratio between gene expression levels of PRAME and PML/RARA at the onset of acute promyelocytic leukemia and clinical features of the disease. Rossiiskii bioterapevticheskii zhurnal. 2014;13(1):9–16. (In Russ)]
  99. Мисюрин А.В. Основы молекулярной диагностики онкогематологических заболеваний. Российский биотерапевтический журнал. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24.
    [Misyurin AV. Fundamentals of the molecular diagnosis of oncohematological diseases. Rossiiskii bioterapevticheskii zhurnal. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24. (In Russ)]

 

 

Correlation between Expression of RARa Transcription Factor and Genes of VEGFR3-Dependent Signaling System in Multiple Myeloma

NN Kalitin, IV Buravtsova

N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Nikolai Nikolaevich Kalitin, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-17-69; e-mail: f.oskolov@mail.ru

For citation: Kalitin NN, Buravtsova IV. Correlation between Expression of RARa Transcription Factor and Genes of VEGFR3-Dependent Signaling System in Multiple Myeloma. Clinical oncohematology. 2015;8(1):31–5 (In Russ).


ABSTRACT

Background. All-trans retinoic acid (ATRA) is a natural metabolite of vitamin A, which can regulate the gene expression by means of interaction between different types of nuclear retinoic acid receptors (RARs). It has been demonstrated that it may lead to suppression of in vivo and in vitro tumor cell growth and can contribute to its survival. For example, in a number of studies, it has been demonstrated that one of RAR subtypes, RARa, modulates expression of a number of vascular endothelial growth factors (VEGFs), mainly VEGF-A. At the same time, the exact mechanisms regulating the RARa-mediated regulation of VEGF (especially VEGF-C and VEGF-D and their receptor VEGFR3) expression are still unclear.

Methods. Changes in expression of mRNAs of VEGF-C, VEGF-D genes and their receptor VEGFR3 in a group of 17 multiple myeloma patients before and after treatment were analyzed in the article. The obtained data were then compared with changes in gene expression of the RARa receptor.

Results. It has been found that overall levels of VEGF-C, VEGF-D and VEGFR3 gene expression were reduced in response to the therapy. Changes in expression of these genes correlated with the RARa expression.

Conclusions. The correlation between VEGF-C, VEGF-D and VEGFR3 expression and RARa expression could indicate a possible involvement of RARa- protein in regulation of VEGFR3-associated signaling system gene expression.

Scientific value. These results may describe a possible mechanism of VEGF-C, VEGF-D and VEGFR3 expression by the RARa transcription factor.


Keywords: multiple myeloma, gene expression, VEGFR3-dependent system, RARa.

Received: August 27, 2014

Accepted: October 20, 2014

Read in  PDF (RUS)pdficon


REFERENCES

  1. Neufeld G. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13(1):9–22.
  2. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med. 2003;9(6):677–84. doi: 10.1038/nm0603-677.
  3. Ristimaki A, Narko K, Enholm B, et al. Proinflammatory cytokines regulate expression of the lymphatic endothelial mitogen vascular endothelial growth factor-C. J Biol Chem. 1998;273(14):8413–8.
  4. Chambon P. A decade of molecular biology of retinoic acid receptors. FASEB J. 1996;10(9):940–54.
  5. Leid M, Kastner P, Chambon P. Multiplicity generates diversity in the retinoic acid signalling pathways. Trends Biochem Sci. 1992;17(10):427–33. doi: 10.1016/0968-0004(92)90014-z.
  6. Mangelsdorf DJ, Evans RM. The RXR heterodimers and orphan receptors. Cell. 1995;83(6):841–50. doi: 10.1016/0092-8674(95)90200-7.
  7. Delacroix L, Moutier E, Altobelli G, et al. Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Mol Cell Biol. 2010;30(1):231–44. doi: 10.1128/mcb.00756-09.
  8. Eifert C, Sangster-Guity N, Yu LM, et al. Global gene expression profiles associated with retinoic acid-induced differentiation of embryonal carcinoma cells. Mol Reprod Dev. 2006;73(7):796–824. doi: 10.1002/mrd.20444.
  9. Maeno T, Tanaka T, Sando Y, et al. Stimulation of vascular endothelial growth factor gene transcription by all trans retinoic acid through Sp1 and Sp3 sites in human bronchioloalveolar carcinoma cells. Am J Respir Cell Mol Biol. 2002;26(2):246–53. doi: 10.1165/ajrcmb.26.2.4509.
  10. Tsuzuki S, Kitajima K, Nakano T, et al. Cross talk between retinoic acid signaling and transcription factor GATA-2. Mol Cell Biol. 2004;24(15):6824–36. doi: 10.1128/mcb.24.15.6824-6836.2004.
  11. Kappel A, Schlaeger TM, Flamme I, et al. Role of SCL/Tal-1, GATA, and its transcription factor binding sites for the regulation of flk-1 expression during murine vascular development. Blood. 2000;96(9):3078–85.
  12. Durie BGM, Salmon SE. A clinical staging system for multiple myeloma. Cancer. 1975;36(3):842–54. doi: 10.1002/1097-0142(197509)36:3<842::aid-cncr2820360303>3.0.co;2-u.
  13. Rajkumar SV, Leong T, Roche PC, et al. Prognostic value of bone marrow angiogenesis in multiple myeloma. Clin Cancer Res. 2000;6(8):3111–6.
  14. Vacca A, Ribatti D, Presta M, et al. Bone marrow neovascularization, plasma cell angiogenic potential, and matrix metalloproteinase-2 secretion parallel progression of human multiple myeloma. Blood. 1999;93(9):3064–73.
  15. Schafer G, Wissmann C, Hertel J, et al. Regulation of vascular endothelial growth factor D by orphan receptors hepatocyte nuclear factor-4A and chicken ovalbumin upstream promoter transcription factors 1 and 2. Cancer Res. 2008;68(2):457–66. doi: 10.1158/0008-5472.CAN-07-5136.
  16. Калитин Н.Н., Какпакова Е.С., Карамышева А.Ф. Влияние ретиноевой кислоты на экспрессию мРНК генов факторов роста эндотелия сосудов VEGF и рецептора VEGFR1 в культурах клеток множественной миеломы человека. Вопросы биологической, медицинской и фармацевтической химии. 2012;10:64–8.
    [Kalitin NN, Kakpakova ES, Karamysheva AF. Effect of retinoid acid on expression of mRNA in genes of vascular endothelial growth factor VEGF and VEGFR1 receptor in cultures of human multiple myeloma cells. Voprosy biologicheskoi, meditsinskoi i farmatsevticheskoi khimii. 2012;10:64–8. (In Russ)]

New molecular markers of CML progression

V.A. Misyurin1,2, A.V. Misyurin1,2, L.A. Kesayeva1,2, Yu.P. Finashutina1,2, Ye.N. Misyurina2, I.N. Soldatova1,2, A.A. Krutov2, N.A. Lyzhko1,2, T.V. Akhlynina1,2, A.Ye. Lukina3, T.I. Kolosheynova3, N.V. Novitskaya1, Ye.G. Arshanskaya4, Ye.G. Ovsyannikova5, R.A. Golubenko6, V.A. Lapin7, T.I. Pospelova8, V.A. Tumakov9, and A.Yu. Baryshnikov1

1 N.N. Blokhin Russian Cancer Research Center, Moscow, Russian Federation

2 GeneTechnology Medical Center, Moscow, Russian Federation

3 Hematology Research Center, RF Ministry of Health, Moscow, Russian Federation

4 Moscow Hematological City Center, S.P. Botkin City Clinical Hospital, Moscow, Russian Federation

5 Astrakhan State Medical Academy, Astrakhan, Russian Federation

6 Orel Regional Clinical Hospital, Orel, Russian Federation

7 Hematological Center, Yaroslavl City Clinical Hospital #1, Yaroslavl, Russian Federation

8 Novosibirsk State Medical University, Novosibirsk, Russian Federation

9 Ivanovo Regional Clinical Hospital #1, Ivanovo, Russian Federation


ABSTRACT

In the contrast to Ph’-negative chronic myeloproliferative disorders (cMPD), chronic myelogenous leukemia (CML) is prone to rather early transformation into the later stage disease, known as the acceleration phase (AP) and blast crisis (BC). Myeloproliferative disorders are termed myeloproliferative neoplasms in the WHO classification, 2008. Molecular mechanisms underlying CML progression are unclear and still being studied. Recently, it was shown that progression of some malignancies was associated with activation and hyperexpression of some genes from the cancer-testis (CT) family. In this study, we evaluated the gene expression profile of CT genes (GAGE1, NY-ESO-1, MAGEA1, SCP1, SEMG1, SPANXA1, SSX1 and PRAME) in the blood of patients with initially diagnosed cMPD, as well as in the blood and bone marrow of CML patients in CP, AP and BC. It was found that activation of these eight CT genes expression was strongly correlated with CML progression to AP and BP. These data suggest that at least some of CT genes can be involved in CML evolution towards the terminal phases. Expression of these genes can be used as an early molecular predictor of CML progression to AP and BC.


Keywords: cancer-testis genes, PRAME, gene expression, chronic myelogenous leukemia, chronic myeloproliferative diseases

Read in в PDF (RUS)pdficon


REFERENCES

  1. Dameshek W. Some speculations on the myeloproliferative syndromes. Blood 1951; 6(4): 372–5.
  2. Tefferi A., Vainchenker W. Myeloproliferative neoplasms: molecular pathophysiology, essential clinical understanding, and treatment strategies. J. Clin. Oncol. 2011; 29(5): 573–82.
  3. Rowley J.D. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and giemsa staining. Nature 1973; 243(5405): 290–3.
  4. Scott L.M., Tong W., Levine R.L. et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N. Engl. J. Med. 2007; 356(5): 459–68.
  5. Gabler K., Behrmann I., Haan C. JAK2 mutants (e.g., JAK2V617F) and their importance as drug targets in myeloproliferative neoplasms. JAKSTAT 2013; 2(3): 250–5.
  6. Deininger M.W., Goldman J.M., Melo J.V. The molecular biology of chronic myeloid leukemia. Blood 2000; 96(10): 3343–56.
  7. Мисюрин А.В. Молекулярный патогенез миелопролиферативных за- болеваний. Клин. онкогематол. 2009; 2(3): 201–9. [Misyurin A.V. Molecular pathogenesis of myeloproliferative disorders. Klin. onkogematol. 2009; 2(3): 201–9. (In Russ.)].
  8. Tutaeva V., Misurin A.V., RoZenberg J.M. et al. Application of prv-1 mrna expression level and Jak2v617f mutation for the differentiating between polycytemia vera and secondary erythrocytosis and assessment of treatment by interferon or hydroxyurea. Hematology 2007; 12(6): 473–9.
  9. Heaney M.L., Soriano G. Acute myeloid leukemia following a myeloproliferative neoplasm: clinical characteristics, genetic features and effects of therapy. Curr. Hematol. Malig. Rep. 2013; 8(2): 116–22.
  10. Turkina A.G., Zabotina T.N., Kusnetzov S.V. et al. Studies of some mechanisms of drug resistence in chronic myeloid leukemia (CML). Adv. Exper. Med. Biol. 1999; 457: 477–88.
  11. Kremenetskaya O.S., Logacheva N.P., Baryshnikov A.Y. et al. Distinct effects of various p53 mutants on differentiation and viability of human K562 leukemia cells. Oncol. Res. 1997; 9: 155–66.
  12. Turkina A.G., Baryshnikov A.Y., Sedyakhina N.P. et al. Studies of Pglycoprotein in chronic myelogenous leukaemia patients: Expression, activity and correlations with CD34 antigen. Br. J. Haematol. 1996; 92: 88–96.
  13. Stavrovskaya A.A., Sedyakhina N.P., Stromskaya T. et al. Prognastic value of P-glicoprotein and correlation with CD34 antigen. Br. J. Heamatol. 1998; 28(5–6): 469–82.
  14. Барышников А.Ю. Взаимодействие опухоли и иммунной системы организма. Практ. онкол. 2003; 4(3): 127–30. [Baryshnikov A.Yu. Interaction between tumor and immune system. Prakt. onkol. 2003; 4(3): 127–30. (In Russ.)].
  15. Барышников А.Ю. Принципы и практика вакцинотерапии рака. Бюл. СО РАМН 2004; 2: 59–63. [Baryshnikov A.Yu. Principles and practice of cancer vaccine-prophylaxis. Byul. SO RAMN 2004; 2: 59–63. (In Russ.)].
  16. Барышников А.Ю., Демидов Л.В., Михайлова И.Н., Петенко Н.Н. Современные проблемы биотерапии опухолей. Вестн. Моск. онкол. общ. 2008; 1: 6–10. [Baryshnikov A.Yu., Demidov L.V., Mikhaylova I.N., Petenko N.N. Current issues of biotherapy for tumors. Vestn. Mosk. onkol. obshch. 2008; 1: 6–10. (In Russ.)]. 17. Michailova I.N., Morozova L.Ph., Golubeva V.A. et al. Cancer/testis genes expression in human melanoma cell lines. Melanoma Res. 2008; 18(5): 303–13.
  17. Turkina A.G., Logacheva N.P., Stromskaya T.P. et al. Studies of some mechanisms of drug resistance in chronic myeloid leukemia (CML). 3rd International Symposium on Drug Resistance in Leukemia and Lymphoma. Amsterdam, 1998. Drug resistance in leukemia and lymphoma III Book Series: advances in experimental medicine and biology. Ed. by G.J.L. Kaspers, R. Pieters, A.J.P. Veerman. 1999: 457, 477–88.
  18. Lim S.H., Zhang Y., Zhang J. Cancer-testis antigens: the current status on antigen regulation and potential clinical use. Am. J. Blood Res. 2012; 2(1): 29–35.
  19. Гапонова Т.В., Менделеева Л.П., Мисюрин А.В., Варламова Е.В., Савченко В.Г. Экспрессия опухолеассоциированных генов PRAME, WT1 и XIAP у больных множественной миеломой. Онкогематол. 2009; 2: 52–7. [Gaponova T.V., Mendeleyeva L.P., Misyurin A.V., Varlamova Ye.V., Savchenko V.G. Expression of PRAME, WT1 and XIAP tumor-associated genes in patients with multiple myeloma. Onkogematol. 2009; 2: 52–7. (In Russ.)].
  20. Абраменко И.В., Белоус Н.И., Крячок И.А. и др. Экспрессия гена PRAME при множественной миеломе. Тер. арх. 2004; 7: 77–81. [Abramenko I.V., Belous N.I., Kryachok I.A., et al. Expression of PRAME gene in multiple myeloma. Ter. arkh. 2004; 7: 77–81. (In Russ.)].
  21. Radich J.P., Dai H., Mao M. et al. Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc. Natl. Acad. Sci. U S A 2006; 103(8): 2794–9.
  22. Демидова И.А., Савченко В.Г., Ольшанская Ю.В. и др. Аллогенная трансплантация костного мозга после режимов кондиционирования по- ниженной интенсивности в терапии больных гемобластозами. Тер. арх. 2003; 75(7): 15–21. [Demidova I.A., Savchenko V.G., Olshanskaya Yu.V., et al. Allogeneic bone martrow transplantation after reduced-intensity conditioning regimens in management of patients with hematological malignancies. Ter. arkh. 2003; 75(7): 15–21. (In Russ.)].
  23. Anguille S., Van Tendeloo V.F., Berneman Z.N. Leukemia-associated antigens and their relevance to the immunotherapy of acute myeloid leukemia. Leukemia 2012; 26(10): 2186–96.
  24. Lichtenegger F.S., Schnorfeil F.M., Hiddemann W., Subklewe M. Current strategies in immunotherapy for acute myeloid leukemia. Immunotherapy 2013; 5(1): 63–78.