Plasmablastic Lymphoma in HIV-Positive Patients: A Literature Review and Results of a Russian Multi-Center Retrospective Study

MO Popova1, IV Tsygankov1, YaV Gudozhnikova1, YuA Rogacheva1, NP Volkov1, KV Lepik1, MV Demchenkova2, MV Grigoreva2, AYu Efirkina2, TV Shneider3, YuV Kopeikina3, SA Stepanova3, VG Potapenko4, AV Klimovich4, NV Medvedeva4, MA Kolesnikova5, TI Pospelova5, NB Mikhailova1, VV Baikov1, AD Kulagin1

1 RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

2 Irkutsk Regional Cancer Center, 32 Frunze str., Irkutsk, Russian Federation, 664035

3 Leningrad Regional Clinical Hospital, 45 bld. 2A Lunacharskogo pr-t, Saint Petersburg, Russian Federation, 194291

4 Municipal Clinical Hospital No. 31, 3 Dinamo pr-t, Saint Petersburg, Russian Federation, 197110

5 Municipal Center for Hematology, 21 Polzunova str., Novosibirsk, Russian Federation, 630051

For correspondence: Marina Olegovna Popova, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(911)711-39-77; e-mail: marina.popova.spb@gmail.com

For citation: Popova MO, Tsygankov IV, Gudozhnikova YaV, et al. Plasmablastic Lymphoma in HIV-Positive Patients: A Literature Review and Results of a Russian Multi-Center Retrospective Study. Clinical oncohematology. 2022;15(1):28–41. (In Russ).

DOI: 10.21320/2500-2139-2022-15-1-28-41


ABSTRACT

Background. Plasmablastic lymphoma (PBL) is a rare lymphoproliferative disease which is almost exclusively associated with immunodeficiency. Most ample experience of chemotherapy and hematopoietic stem cells transplantation (HSCT) in this lymphoma variant has been accumulated in HIV-positive patients.

Aim. To describe the current approaches to PBL diagnosis and treatment in HIV-positive patients as well as to provide the results of the first multi-center retrospective study on PBL epidemiology and therapy efficacy in HIV-positive patients in the Russian Federation.

Materials & Methods. The study included 26 HIV-positive patients with PBL who were treated and followed-up at 5 Russian centers during 2012–2019. The present study is a part of multi-center retrospective study on lymphoma epidemiology in HIV-positive patients in Russia.

Results. PBL accounted for 9.5 % of all lymphomas in HIV-positive patients enrolled in multi-center retrospective study on lymphoma epidemiology in HIV-positive patients in Russia. Epidemiological characteristics of these patients corresponded to those described in previously published literature: the disease being diagnosed mainly at late stages (88 %), oral and nasal mucosa lesions with a common involvement of facial bones (65 %), and lack of optimal HIV-infection control (66.7 %). Most commonly, the patients received EPOCH-like treatment as first-line therapy (50 %). However, the efficacy of primary therapy appeared to be low. Overall survival (OS) and progression-free survival (PFS) during a year after first-line therapy onset was 57 % and 46 %, respectively. Bortezomib included in first-line therapy was associated with a trend to a more favorable prognosis. Half of patients showed a lymphoma relapse or progression after first-line therapy. Most used second-line regimen was DHAP. Overall response to second-line therapy was 38.5 %. After second-line therapy onset, 1-year OS and PFS were 26 % and 15 %, respectively.

Conclusion. HIV-positive patients with PBL have poor prognosis. Efforts to improve the prognosis for HIV-positive patients with PBL should be aimed at increasing the efficacy of first-line therapy and should involve the use of intensive chemotherapy regimens with bortezomib. The role of auto- and allo-HSCTs in the treatment of PBL has not been clearly determined, however, PBL patients, despite their HIV-infection, should be regarded as auto-HSCT-eligible in the first remission and allo-HSCT-eligible in case of relapse. Further prospective multi-center studies are needed to optimize the treatment of HIV-positive patients with PBL.

Keywords: plasmablastic lymphoma, HIV-infection, Epstein-Barr virus, MYC, PD-1/PD-L1/2, auto-HSCT, allo-HSCT, bortezomib, nivolumab, immunotherapy.

Received: July 20, 2021

Accepted: November 27, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. World Health Organization. HIV data and statistics. Available from: https://www.who.int/teams/global-hiv-hepatitis-and-stis-programmes/hiv/strategic-information/hiv-data-and-statistics (accessed 20.07.2021).
  2. Shiels MS, Pfeiffer RM, Gail MH, et al. Cancer burden in the HIV-infected population in the United States. J Natl Cancer Inst. 2011;103(9):753–62. doi: 10.1093/jnci/djr076.
  3. Heron M. Deaths: Leading Causes for 2017. Natl Vital Stat Rep. 2019;68(6):1–77.
  4. Selik RM, Mokotoff ED, Branson B, et al. Revised Surveillance Case Definition for HIV Infection — United States, 2014. Morbid Mortal Weekly Rep. 2014;63(RR-03):1–10.
  5. Robbins HA, Shiels MS, Pfeiffer RM, et al. Epidemiologic contributions to recent cancer trends among HIV-infected people in the United States. AIDS. 2014;28(6):881–90. doi: 10.1097/QAD.0000000000000163.
  6. Goncalves PH, Montezuma-Rusca JM, Yarchoan R, Uldrick TS. Cancer prevention in HIV-infected populations. Semin Oncol. 2016;43(1):173–88. doi: 10.1053/j.seminoncol.2015.09.011.
  7. Delecluse HJ, Anagnostopoulos I, Dallenbach F, et al. Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection. 1997;89(4):1413–20.
  8. Campo E, Swerdlow SH, Harris NL, et al. The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood. 2011;117(19):5019–32. doi: 10.1182/blood-2011-01-293050.
  9. Knowles DM, Chadburn A Lymphadenopathy and the lymphoid neoplasms associated with the acquired immune deficiency syndrome (AIDS). In: Knowles DM, ed. Neoplastic Hematopathology. Baltimore: Williams and Walker; 1992. pp. 773.
  10. Zuze T, Painschab MS, Seguin R, et al. Plasmablastic lymphoma in Malawi. Infect Agent Cancer. 2018;13:22. doi: 10.1186/s13027-018-0195-4.
  11. Liu JJ, Zhang L, Ayala E, et al. Human immunodeficiency virus (HIV)-negative plasmablastic lymphoma: a single institutional experience and literature review. Leuk Res. 2011;35(12):1571–7. doi: 10.1016/j.leukres.2011.06.023.
  12. Tchernonog E, Faurie P, Coppo P, et al. Clinical characteristics and prognostic factors of plasmablastic lymphoma patients: analysis of 135 patients from the LYSA group. Ann Oncol. 2017;28(4):843–8. doi: 10.1093/annonc/mdw684.
  13. Castillo JJ, Winer ES, Stachurski D, et al. Clinical and pathological differences between human immunodeficiency virus-positive and human immunodeficiency virus-negative patients with plasmablastic lymphoma. Leuk Lymphoma. 2010;51(11):2047–53. doi: 10.3109/10428194.2010.516040.
  14. Morscio J, Dierickx D, Nijs J, et al. Clinicopathologic comparison of plasmablastic lymphoma in HIV-positive, immunocompetent, and posttransplant patients: single-center series of 25 cases and meta-analysis of 277 reported cases. Am J Surg Pathol. 2014;38(7):875–86. doi: 10.1097/PAS.0000000000000234.
  15. Loghavi S, Alayed K, Aladily TN, et al. Stage, age, and EBV status impact outcomes of plasmablastic lymphoma patients: a clinicopathologic analysis of 61 patients. J Hematol Oncol. 2015;8:65. doi: 10.1186/s13045-015-0163-z.
  16. Bibas M, Antinori A. EBV and HIV-Related Lymphoma. Mediterr J Hematol Infect Dis. 2009;1(2):e2009032. doi: 10.4084/MJHID.2009.032.
  17. Castillo J, Pantanowitz L, Dezube BJ. HIV-associated plasmablastic lymphoma: lessons learned from 112 published cases. Am J Hematol. 2008;83(10):804–9. doi: 10.1002/ajh.21250.
  18. Dolcetti R, Gloghini A, Caruso A, Carbone A. A lymphomagenic role for HIV beyond immune suppression? Blood. 2016;127(11):1403–9. doi: 10.1182/blood-2015-11-681411.
  19. Valera A, Balague O, Colomo L, et al. IG/MYC rearrangements are the main cytogenetic alteration in plasmablastic lymphomas. Am J Surg Pathol. 2010;34(11):1686–94. doi: 10.1097/PAS.0b013e3181f3e29f.
  20. Montes-Moreno S, Martinez-Magunacelaya N, Zecchini-Barrese T, et al. Plasmablastic lymphoma phenotype is determined by genetic alterations in MYC and PRDM1. Mod Pathol. 2017;30(1):85–94. doi: 10.1038/modpathol.2016.162.
  21. Garcia JF, Roncador G, Garcia JF, et al. PRDM1/BLIMP-1 expression in multiple B and T-cell lymphoma. Haematologica. 2006;91(4):467–74.
  22. Liu F, Asano N, Tatematsu A, et al. Plasmablastic lymphoma of the elderly: a clinicopathological comparison with age-related Epstein-Barr virus-associated B cell lymphoproliferative disorder. Histopathology. 2012;61(6):1183–97. doi: 10.1111/j.1365-2559.2012.04339.x.
  23. Boy SC, van Heerden MB, Babb C, et al. Dominant genetic aberrations and coexistent EBV infection in HIV-related oral plasmablastic lymphomas. Oral Oncol. 2011;47(9):883–7. doi: 10.1016/j.oraloncology.2011.06.506.
  24. Gruhne B, Sompallae R, Masucci MG. Three Epstein-Barr virus latency proteins independently promote genomic instability by inducing DNA damage, inhibiting DNA repair and inactivating cell cycle checkpoints. Oncogene. 2009;28(45):3997–4008. doi: 10.1038/onc.2009.258.
  25. Price AM, Luftig MA. Dynamic Epstein-Barr virus gene expression on the path to B-cell transformation. Adv Virus Res. 2014;88:279–313. doi: 10.1016/B978-0-12-800098-4.00006-4.
  26. Ma SD, Hegde S, Young KH, et al. A new model of Epstein-Barr virus infection reveals an important role for early lytic viral protein expression in the development of lymphomas. J Virol. 2011;85(1):165–77. doi: 10.1128/JVI.01512-10.
  27. Shannon-Lowe C, Adland E, Bell AI, et al. Features distinguishing Epstein-Barr virus infections of epithelial cells and B cells: viral genome expression, genome maintenance, and genome amplification. J Virol. 2009;83(15):7749–60. doi: 10.1128/JVI.00108-09.
  28. Di Pietro A. Epstein-Barr Virus Promotes B Cell Lymphomas by Manipulating the Host Epigenetic Machinery. Cancers (Basel). 2020;12(10):3037. doi: 10.3390/cancers12103037.
  29. Montes-Moreno S, Gonzalez-Medina AR, Rodriguez-Pinilla SM, et al. Aggressive large B-cell lymphoma with plasma cell differentiation: immunohistochemical characterization of plasmablastic lymphoma and diffuse large B-cell lymphoma with partial plasmablastic phenotype. Haematologica. 2010;95(8):1342–9. doi: 10.3324/haematol.2009.016113.
  30. Harmon CM, Smith LB. Plasmablastic Lymphoma: A Review of Clinicopathologic Features and Differential Diagnosis. Arch Pathol Lab Med. 2016;140(10):1074–8. doi: 10.5858/arpa.2016-0232-RA.
  31. Grimm KE, O’Malley DP. Aggressive B cell lymphomas in the 2017 revised WHO classification of tumors of hematopoietic and lymphoid tissues. Ann Diagn Pathol. 2019;38:6–10. doi: 10.1016/j.anndiagpath.2018.09.014.
  32. Ramis-Zaldivar JE, Gonzalez-Farre B, Nicolae A, et al. MAP-kinase and JAK-STAT pathways dysregulation in plasmablastic lymphoma. Haematologica. 2021;106(10):2682–93. doi: 10.3324/haematol.2020.271957.
  33. Nasta SD, Carrum GM, Shahab I, et al. Regression of a plasmablastic lymphoma in a patient with HIV on highly active antiretroviral therapy. Leuk Lymphoma. 2002;43(2):423–6. doi: 10.1080/10428190290006260.
  34. Lester R, Li C, Phillips P, et al. Improved outcome of human immunodeficiency virus-associated plasmablastic lymphoma of the oral cavity in the era of highly active antiretroviral therapy: a report of two cases. Leuk Lymphoma. 2004;45(9):1881–5. doi: 10.1080/10428190410001697395.
  35. Armstrong R, Bradrick J, Liu YC. Spontaneous regression of an HIV-associated plasmablastic lymphoma in the oral cavity: a case report. J Oral Maxillofac Surg. 2007;65(7):1361–4. doi: 10.1016/j.joms.2005.12.039.
  36. Yordanova K, Stilgenbauer S, Bohle RM, et al. Spontaneous regression of a plasmablastic lymphoma with MYC rearrangement. Br J Haematol. 2019;186(6):e203–e207. doi: 10.1111/bjh.16082.
  37. Natkunam Y, Gratzinger D, Chadburn A, et al. Immunodeficiency-associated lymphoproliferative disorders: time for reappraisal? Blood. 2018;132(18):1871–8. doi: 10.1182/blood-2018-04-842559.
  38. Vaubell JI, Sing Y, Ramburan A, et al. Pediatric plasmablastic lymphoma: a clinicopathologic study. Int J Surg Pathol. 2014;22(7):607–16. doi: 10.1177/1066896914531815.
  39. Jariwal R, Raza N, Bhandohal J, Cobos E. Non-Hodgkin’s Plasmablastic Lymphoma as Initial Presentation of Human Immunodeficiency Virus. J Investig Med High Impact Case Rep. 2021;9:23247096211014689. doi: 10.1177/23247096211014689.
  40. Little RF, Dunleavy K. Update on the treatment of HIV-associated hematologic malignancies. Hematology Am Soc Hematol Educ Program. 2013;2013(1):382–8. doi: 10.1182/asheducation-2013.1.382.
  41. Schommers P, Wyen C, Hentrich M, et al. Poor outcome of HIV-infected patients with plasmablastic lymphoma: results from the German AIDS-related lymphoma cohort study. AIDS. 2013;27(5):842–5. doi: 10.1097/QAD.0b013e32835e069d.
  42. Dittus C, Grover N, Ellsworth S, et al. Bortezomib in combination with dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin) induces long-term survival in patients with plasmablastic lymphoma: a retrospective analysis. Leuk Lymphoma. 2018;59(9):2121–7. doi: 10.1080/10428194.2017.1416365.
  43. Barta SK, Xue X, Wang D, et al. Treatment factors affecting outcomes in HIV-associated non-Hodgkin lymphomas: a pooled analysis of 1546 patients. Blood. 2013;122(19):3251–62. doi: 10.1182/blood-2013-04-498964.
  44. Castillo JJ, Furman M, Beltran BE, et al. Human immunodeficiency virus-associated plasmablastic lymphoma: poor prognosis in the era of highly active antiretroviral therapy. Cancer. 2012;118(21):5270–7. doi: 10.1002/cncr.27551.
  45. Ibrahim IF, Shapiro GA, Naina HVK. Treatment of HIV-associated plasmablastic lymphoma: A single-center experience with 25 patients. J Clin Oncol. 2014;32(5 Suppl):8583. doi: 10.1200/jco.2014.32.15_suppl.8583.
  46. NCCN Guidelines Version: 4.2021. B-Cell Lymphomas. Available from: https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf. (accessed 20.07.2021).
  47. Antinori A, Cingolani A, Alba L, et al. Better response to chemotherapy and prolonged survival in AIDS-related lymphomas responding to highly active antiretroviral therapy. AIDS. 2001;15(12):1483–91. doi: 10.1097/00002030-200108170-00005.
  48. Guan B, Zhang X, Ma H, et al. Meta-analysis of highly active anti-retroviral therapy for treatment of plasmablastic lymphoma. Hematol Oncol Stem Cell Ther. 2010;3(1):7–12. doi: 10.1016/s1658-3876(10)50050-5.
  49. Francischini E, Martins FM, Braz-Silva PH, et al. HIV-associated oral plasmablastic lymphoma and role of adherence to highly active antiretroviral therapy. Int J STD AIDS. 2010;21(1):68–70. doi: 10.1258/ijsa.2008.008476.
  50. Re A, Cattaneo C, Michieli M, et al. High-dose therapy and autologous peripheral-blood stem-cell transplantation as salvage treatment for HIV-associated lymphoma in patients receiving highly active antiretroviral therapy. J Clin Oncol. 2003;21(23):4423–7. doi: 10.1200/JCO.2003.06.039.
  51. Re A, Michieli M, Casari S, et al. High-dose therapy and autologous peripheral blood stem cell transplantation as salvage treatment for AIDS-related lymphoma: long-term results of the Italian Cooperative Group on AIDS and Tumors (GICAT) study with analysis of prognostic factors. Blood. 2009;114(7):1306–13. doi: 10.1182/blood-2009-02-202762.
  52. Popova MO, Rogacheva YA, Nekrasova AV, et al. Autologous hematopoietic cell transplantation for HIV-related lymphoma: results of a single center (CIC725) matched case-control study. Cell Ther Transplant. 2017;6(4):42–51. doi: 10.18620/ctt-1866-8836-2017-6-4-42-51.
  53. Popova M, Tsygankov I, Rogacheva Y, et al. High-dose chemotherapy with autologous hematopoietic stem cell transplantation in patients with HIV-related lymphoma. Annals Oncol. 2020;31(Suppl 4):S651. doi: 10.1016/j.annonc.2020.08.010.
  54. Younan P, Kowalski J, Kiem HP. Genetic modification of hematopoietic stem cells as a therapy for HIV/AIDS. Viruses. 2013;5(12):2946–62. doi: 10.3390/v5122946.
  55. Mock U, Machowicz R, Hauber I, et al. mRNA transfection of a novel TAL effector nuclease (TALEN) facilitates efficient knockout of HIV co-receptor CCR5. Nucl Acids Res. 2015;43(11):5560–71. doi: 10.1093/nar/gkv469.
  56. Wang CX, Cannon PM. The clinical applications of genome editing in HIV. 2016;127(21):2546–52. doi: 10.1182/blood-2016-01-678144.
  57. Попова М.О., Сергеев В.С., Лепик К.В. и др. Генная клеточная терапия ВИЧ и злокачественных опухолей кроветворной и лимфатической ткани на основе трансплантации гемопоэтических стволовых клеток с использованием сайт-специфического редактирования генома. Журнал инфектологии. 2017;9(1):31–9. doi: 10.22625/2072-6732-2017-9-1-31-39.
    [Popova MО, Sergeev VS, Lepik KV, et al. Gene-Cell Therapy of HIV and Hematological Malignances Based on Hematopoietic Stem Cell Transplantation and Site-Specific Genome Editing. Journal Infectology. 2017;9(1):31–9. doi: 10.22625/2072-6732-2017-9-1-31-39. (In Russ)]
  58. Morgan RA, Gray D, Lomova A, et al. Hematopoietic Stem Cell Gene Therapy: Progress and Lessons Learned. Cell Stem Cell. 2017;21(5):574–90. doi: 10.1016/j.stem.2017.10.010.
  59. Popova MO, Lepik KV, Sergeev VS, et al. Clinical implementation of genome editing for correction of human diseases. Cell Ther Transplant. 2017;6(1):37–43. doi: 10.18620/ctt-1866-8836-2017-6-1-37-43.
  60. Serrano D, Carrion R, Balsalobre P, et al. Spanish Cooperative Groups GELTAMO and GESIDA. HIV-associated lymphoma successfully treated with peripheral blood stem cell transplantation. Exp Hematol. 2005;33(4):487–94. doi: 10.1016/j.exphem.2004.12.008.
  61. Bowden RA, Coombs RW, Nikora BH, et al. Progression of human immunodeficiency virus type-1 infection after allogeneic marrow transplantation. Am J Med. 1990;88(5N):49N–52N.
  62. Al-Malki MM, Castillo JJ, Sloan JM, et al. Hematopoietic cell transplantation for plasmablastic lymphoma: a review. Biol Blood Marrow Transplant. 2014;20(12):1877–84. doi: 10.1016/j.bbmt.2014.06.009.
  63. Gupta V, Tomblyn M, Pedersen TL, et al. Allogeneic hematopoietic cell transplantation in human immunodeficiency virus-positive patients with hematologic disorders: a report from the center for international blood and marrow transplant research. Biol Blood Marrow Transplant. 2009;15(7):864–71. doi: 10.1016/j.bbmt.2009.03.023.
  64. Hamadani M, Devine SM. Reduced-intensity conditioning allogeneic stem cell transplantation in HIV patients with hematologic malignancies: yes, we can. Blood. 2009;114(12):2564–6. doi: 10.1182/blood-2009-06-229666.
  65. Afanasyev BV, Popova MO, Bondarenko SN, et al. St. Petersburg experience of allogeneic hematopoietic stem cell transplantation in patients with acute leukemia and human immunodeficiency virus. Cell Ther Transplant. 2015;4(1–2):24–30. doi: 10.18620/1866-8836-2015-4-1-2-24-30.
  66. Yoon JH, Jeon YW, Lee SE, et al. Allogeneic stem cell transplantation using lymphoablative rather than myeloablative conditioning regimen for relapsed or refractory lymphomas. Hematol Oncol. 2017;35(1):17–24. doi: 10.1002/hon.2201.
  67. Rong C, Sheng L, Wu A, et al. Allogeneic hematopoietic stem cell transplantation in a patient with HIV-negative recurrent plasmablastic lymphoma: A case report. Medicine (Baltimore). 2021;100(7):e24498. doi: 10.1097/MD.0000000000024498.
  68. Lepik KV, Mikhailova NB, Moiseev IS, et al. Nivolumab for the treatment of relapsed and refractory classical Hodgkin lymphoma after ASCT and in ASCT-naive patients. Leuk Lymphoma. 2019;60(9):2316–9. doi: 10.1080/10428194.2019.1573368.
  69. Ambinder RF, Wu J, Logan B, et al. Allogeneic Hematopoietic Cell Transplant for HIV Patients with Hematologic Malignancies: The BMT CTN-0903/AMC-080 Trial. Biol Blood Marrow Transplant. 2019;25(11):2160–6. doi: 10.1016/j.bbmt.2019.06.033.
  70. Umeanaeto O, Gamboa J, Diaz J, et al. Incorporating Bortezomib in the Management of Plasmablastic Lymphoma. Anticancer Res. 2019;39(9):5003–7. doi: 10.21873/anticanres.13690.
  71. Castillo JJ, Reagan JL, Sikov W, et al. Bortezomib in combination with infusional dose-adjusted EPOCH for the treatment of plasmablastic lymphoma. Br J Haematol. 2015;169(3):352–5. doi: 10.1111/bjh.1330.
  72. Cao C, Liu T, Zhu H, et al. Bortezomib-contained chemotherapy and thalidomide combined with CHOP (Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone) play promising roles in plasmablastic lymphoma: a case report and literature review. Clin Lymphoma Myeloma Leuk. 2014;14(5):e145–е150. doi: 10.1016/j.clml.2014.03.002.
  73. Cencini E, Fabbri A, Guerrini S, et al. Long-term remission in a case of plasmablastic lymphoma treated with COMP (cyclophosphamide, liposomal doxorubicin, vincristine, prednisone) and bortezomib. Eur J Haematol. 2016;96(6):650–4. doi: 10.1111/ejh.12732.
  74. Colomo L, Loong F, Rives S, et al. Diffuse large B-cell lymphomas with plasmablastic differentiation represent a heterogeneous group of disease entities. Am J Surg Pathol. 2004;28(6):736–47. doi: 10.1097/01.pas.0000126781.87158.e3.
  75. Holderness BM, Malhotra S, Levy NB, Danilov AV. Brentuximab vedotin demonstrates activity in a patient with plasmablastic lymphoma arising from a background of chronic lymphocytic leukemia. J Clin Oncol. 2013;31(12):e197–е199. doi: 10.1200/JCO.2012.46.9593.
  76. Pretscher D, Kalisch A, Wilhelm M, et al. Refractory plasmablastic lymphoma-a review of treatment options beyond standard therapy. Ann Hematol. 2017;96(6):967–70. doi: 10.1007/s00277-016-2904-7.
  77. Laurent C, Fabiani B, Do C, et al. Immune-checkpoint expression in Epstein-Barr virus positive and negative plasmablastic lymphoma: a clinical and pathological study in 82 patients. Haematologica. 2016;101(8):976–84. doi: 10.3324/haematol.2016.141978.
  78. Damlaj M, Alzayed M, Alahmari B, et al. Therapeutic Potential of Checkpoint Inhibitors in Refractory Plasmablastic Lymphoma. Clin Lymphoma Myeloma Leuk. 2019;19(10):e559–e563. doi: 10.1016/j.clml.2019.06.008.
  79. Popova M, Rogacheva Y, Tsygankov I, et al. Immunotherapy in patients with relapsed/refractory HIV-related lymphomas. Annals Oncol. 2019;30(11):XI22. doi: 10.1093/annonc/mdz449.015.
  80. Lepik KV, Mikhailova NB, Kondakova EV, et al. A Study of Safety and Efficacy of Nivolumab and Bendamustine (NB) in Patients With Relapsed/Refractory Hodgkin Lymphoma After Nivolumab Monotherapy Failure. Hemasphere. 2020;4(3):e401. doi: 10.1097/HS9.0000000000000401.
  81. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059–68. doi: 10.1200/JCO.2013.54.8800.
  82. Каприн А.Д., Старинский В.В., Шахзадова А.О. Состояние онкологической помощи населению России в 2019 году. М.: МНИОИ им. П.А. Герцена — филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2020. [Kaprin AD, Starinskii VV, Shakhzadova AO. Sostoyanie onkologicheskoi pomoshchi naseleniyu Rossii v 2019 godu. (The state of cancer care in Russia in 2019.) Moscow: MNIOI im. P.A. Gertsena — filial FGBU “NMITs radiologii” Publ.; (In Russ)]
  83. Информационные бюллетени Федерального научно-методического центра по профилактике и борьбе со СПИДом: Бюллетень № 40 (электронный документ). Доступно по: http://www.hivrussia.ru/files/bulpdf. Ссылка активна на 20.07.2021.
    [Information Bulletins of Federal Scientific and Methodological Center for the Prevention and Control of AIDS: Bulletin No. 40 (Internet). Available from: http://www.hivrussia.ru/files/bul_40.pdf. (accessed 20.07.2021) (In Russ)]

EVI1-Positive Leukemias and Myelodysplastic Syndromes: Theoretical and Clinical Aspects (Literature Review)

NN Mamaev, AI Shakirova, EV Morozova, TL Gindina

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Prof. Nikolai Nikolaevich Mamaev, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; e-mail: nikmamaev524@gmail.com

For citation: Mamaev NN, Shakirova AI, Morozova EV, Gindina TL. EVI1-Positive Leukemias and Myelodysplastic Syndromes: Theoretical and Clinical Aspects (Literature Review). Clinical oncohematology. 2021;14(1):103–17. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-103-117


ABSTRACT

The present review provides the analysis of theoretical background and therapy of prognostically poorest EVI1-positive myeloid leukemias and myelodysplastic syndromes which is performed at the RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation. The focus is on the evidence of the dominating role of EVI1 gene in impaired epigenetic regulation of hematopoiesis and, thus, on the feasibility of allogeneic hematopoietic stem cell transplantation with hypomethylating agents and/or trans-retinoic acid used for these diseases treatment.

Keywords: EVI1, acute myeloid leukemia, chronic myeloid leukemia, myelodysplastic syndrome, allo-HSCT, hypomethylating agents, trans-retinoic acid.

Received: September 12, 2020

Accepted: December 6, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Barjesteh van Waalwijk van Doorn-Khosrovani S. High EVI1 expression predicts poor survival in acute myeloid leukemia: a study of 319 de novo AML patients. Blood. 2002;101(3):837–45. doi: 10.1182/blood-2002-05-1459.
  2. Lugthart S, van Drunen E, van Norden Y, et al. High EVI1 levels predict adverse outcome in acute myeloid leukemia: prevalence of EVI1 overexpression and chromosome 3q26 abnormalities underestimated. Blood. 2008;111(8):4329–37. doi: 10.1182/blood-2007-10-119230.
  3. Groschel S, Lugthart S, Schlenk RF, et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. J Clin Oncol. 2010;28(12):2101–7. doi: 10.1200/JCO.2009.26.0646.
  4. Paquette RL, Nicoll J, Chalukya M, et al. Frequent EVI1 translocations in myeloid blast crisis CML that evolves through tyrosine kinase inhibitors. Cancer Genet. 2011;204(7):392–7. doi: 10.1016/j.cancergen.2011.06.002.
  5. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миелодиспластические синдромы с высокой экспрессией гена EVI1: теоретические и клинические аспекты. Клиническая онкогематология. 2012;5(4):361–4.
    [Mamaev NN, Gorbunova AV, Gindina TL, et al. Leukemias and myelodysplastic syndromes with high expression of EVI1 gene: theoretical and clinical aspects. Klinicheskaya onkogematologiya. 2012;5(4):361–4. (In Russ)]
  6. Rogers HJ, Vardiman JW, Anastasi J, et al. Complex or monosomal karyotype and not blast percentage is associated with poor survival in acute myeloid leukemia and myelodysplastic syndrome patients with inv(3)(q21q26.2)/t(3;3)(q21;q26.2): a Bone Marrow Pathology Group study. Haematologica. 2014;99(5):821–9. doi: 10.3324/haematol.2013.096420.
  7. Reiter E, Greinix H, Rabitsch W, et al. Low curative potential of bone marrow transplantation for highly aggressive acute myelogenous leukemia with inversion inv(3)(q21q26) or homologous translocation t(3;3)(q21;q26). Ann Hematol. 2000;79(7):374–7. doi: 10.1007/s002770000158.
  8. He X, Wang Q, Cen J, et al. Predictive value of high EVI1 expression in AML patients undergoing myeloablative allogeneic hematopoietic stem cell transplantation in first CR. Bone Marrow Transplant. 2016;51(7):921–7. doi: 10.1038/bmt.2016.71.
  9. Gindina TL, Mamaev NN, Afanasyev BV. Chromosome abnormalities and hematopoietic stem cell transplantation in acute leukemias. In: ML Larramendy, S Soloneski (eds). Chromosomal abnormalities – A hallmark manifestation of genomic instability. IntechOpen; 2017. рр. 71– doi: 10.5772/67802.
  10. Halaburda K, Labopin M, Houhou M, et al. AlloHSCT for inv(3)(q21;q26)/t(3;3)(q21;q26) AML: a report from the acute leukemia working party of the European society for blood and marrow transplantation. Bone Marrow Transplant. 2018;53(6):683–91. doi: 10.1038/s41409-018-0165-x.
  11. Martinelli G, Ottaviani E, Buonamici S, et al. Association of 3q21q26 syndrome with different RPN1/EVI1 fusion transcripts. Haematologica. 2003;88(11):1221–8.
  12. Poppe B, Dastugue N, Vandesompele J, et al. EVI1 is consistently expressed as principal transcript in common and rare recurrent 3q26 rearrangements. Genes Chromos Cancer. 2006;45(4):349–56. doi: 10.1002/gcc.20295.
  13. De Braekeleer M, Le Bris MJ, De Braekeleer E, et al. 3q26/EVI1 rearrangements in myeloid hemopathies: a cytogenetic review. Fut Oncol. 2015;11(11):1675–86. doi: 10.2217/fon.15.64.
  14. Mamaev NN, Gindina TL, Morozova EV, et al. Primary myelodysplastic syndrome with two rare recurrent chromosome abnormalities [t(3q26/2;q22 and trisomy 13] associated with resistance to chemotherapy and hematopoietic stem cell transplantation. Cell Ther Transplant. 2018;7(2):64–9. doi: 10/18620/ctt-1866-8836-2018-7-2-64-69.
  15. Hodge JC, Bosler D, Rubinstein L, et al. Molecular and pathologic characterization of AML with double inv(3)(q21q26.2). Cancer Genet. 2019;230:28–36. doi: 10.1016/j.cancergen.2018.08.007.
  16. Testoni N, Borsaru G, Martinelli G, et al. 3q21 and 3q26 cytogenetic abnormalities in acute myeloblastic leukemia: biological and clinical features. Haematologica. 1999;84(8):690–4.
  17. Russell M, List A, Greenberg P, et al. Expression of EVI1 in myelodysplastic syndromes and other hematologic malignancies without 3q26 translocations. Blood. 1994;84(4):1243–8. doi: 10.1182/blood.V84.4.1243.1243.
  18. Groschel S, Schlenk RF, Engelmann J, et al. Deregulated expression of EVI1 defines a poor prognostic subset of MLL-rearranged acute myeloid leukemias: a study of the German-Austrian Acute Myeloid Leukemia Study Group and the Dutch-Belgian-Swiss HOVON/SAKK Cooperative Group. J Clin Oncol. 2013;31(1):95–103. doi: 10.1200/JCO.2011.41.5505.
  19. Ho PA, Alonzo TA, Gerbing RB, et al. High EVI1 expression is associated with MLL rearrangements and predicts decreased survival in paediatric acute myeloid leukaemia: a report from the children’s oncology group. Br J Haematol. 2013;162(5):670–7. doi: 10.1111/bjh.12444.
  20. Zhang Y, Owens K, Hatem L, et al. Essential role of PR-domain protein MDS1-EVI1 in MLL-AF9 leukemia. Blood. 2013;122(16):2888–92. doi: 10.1182/blood-2012-08-453662.
  21. Mucenski ML, Taylor BA, Ihle JN, et al. Identification of a common ecotropic viral integration site, Evi-1, in the DNA of AKXD murine myeloid tumors. Mol Cell Biol. 1988;8(1):301–8. doi: 10.1128/mcb.8.1.301.
  22. Goyama S, Kurokawa M. Pathogenetic significance of ecotropic viral integration site-1 in hematological malignancies. Cancer Sci. 2009;100(6):990–5. doi: 10.1111/j.1349-7006.2009.01152.x.
  23. Hinai AA, Valk PJ. Review: Aberrant EVI1 expression in acute myeloid leukaemia. Br J Haematol. 2016;172(6):870–8. doi: 10.1111/bjh.13898.
  24. Yuan X, Wang X, Bi K, Jiang G. The role of EVI-1 in normal hematopoiesis and myeloid malignancies (Review). Int J Oncol. 2015;47(6):2028–36. doi: 10.3892/ijo.2015.3207.
  25. Delwel R, Funabiki T, Kreider BL, et al. Four of the seven zinc fingers of the Evi-1 myeloid-transforming gene are required for sequence-specific binding to GA(C/T)AAGA(T/C)AAGATAA. Mol Cell Biol. 1993;13(7):4291–300. doi: 10.1128/mcb.13.7.4291.
  26. Funabiki T, Kreider BL, Ihle JN. The carboxyl domain of zinc fingers of the Evi-1 myeloid transforming gene binds a consensus sequence of GAAGATGAG. Oncogene. 1994;9(6):1575–81.
  27. Morishita K, Suzukawa K, Taki T, et al. EVI-1 zinc finger protein works as a transcriptional activator via binding to a consensus sequence of GACAAGATAAGATAAN1-28 CTCATCTTC. Oncogene. 1995;10(10):1961–7.
  28. Perkins AS, Kim JH. Zinc fingers 1–7 of EVI1 fail to bind to the GATA motif by itself but require the core site GACAAGATA for binding. J Biol Chem. 1996;271(2):1104–10. doi: 10.1074/jbc.271.2.1104.
  29. Bartholomew C, Kilbey A, Clark AM, Walker M. The Evi-1 proto-oncogene encodes a transcriptional repressor activity associated with transformation. Oncogene. 1997;14(5):569–77. doi: 10.1038/sj.onc.1200864.
  30. Kilbey A, Bartholomew C. Evi-1 ZF1 DNA binding activity and a second distinct transcriptional repressor region are both required for optimal transformation of Rat1 fibroblasts. Oncogene. 1998;16(17):2287–91. doi: 10.1038/sj.onc.1201732.
  31. Bordereaux D, Fichelson S, Tambourin P, Gisselbrecht S. Alternative splicing of the Evi-1 zinc finger gene generates mRNAs which differ by the number of zinc finger motifs. Oncogene. 1990;5(6):925–7.
  32. Alzuherri H, McGilvray R, Kilbey A, Bartholomew C. Conservation and expression of a novel alternatively spliced Evi1 exon. Gene. 2006;384:154–62. doi: 10.1016/j.gene.2006.07.027.
  33. Fears S, Mathieu C, Zeleznik-Le N, et al. Intergenic splicing of MDS1 and EVI1 occurs in normal tissues as well as in myeloid leukemia and produces a new member of the PR domain family. Proc Natl Acad Sci USA. 1996;93(4):1642–7. doi: 10.1073/pnas.93.4.1642.
  34. Huang S, Shao G, Liu L. The PR domain of the Rb-binding zinc finger protein RIZ1 is a protein binding interface and is related to the SET domain functioning in chromatin-mediated gene expression. J Biol Chem. 1998;273(26):15933–9. doi: 10.1074/jbc.273.26.15933.
  35. Goyama S, Yamamoto G, Shimabe M, et al. Evi-1 is a critical regulator for hematopoietic stem cells and transformed leukemic cells. Cell Stem Cell. 2008;3(2):207–20. doi: 10.1016/j.stem.2008.06.002.
  36. Laricchia-Robbio L, Nucifora G. Significant increase of self-renewal in hematopoietic cells after forced expression of EVI1. Blood Cells Mol Dis. 2008;40(2):141–7. doi: 10.1016/j.bcmd.2007.07.012.
  37. Yoshimi A, Kurokawa M. Evi1 forms a bridge between the epigenetic machinery and signaling pathways. Oncotarget. 2011;2(7):575–86. doi: 10.18632/oncotarget.304.
  38. Buonamici S, Li D, Chi Y, et al. EVI1 induces myelodysplastic syndrome in mice. J Clin Invest. 2005;115(8):2296. doi: 1172/jci21716c1.
  39. Cuenco GM, Ren R. Both AML1 and EVI1 oncogenic components are required for the cooperation of AML1/MDS1/EVI1 with BCR/ABL in the induction of acute myelogenous leukemia in mice. Oncogene. 2004;23(2):569–79. doi: 10.1038/sj.onc.1207143.
  40. Glass C, Wilson M, Gonzalez R, et al. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis. 2014;53(1–2):67–76. doi: 10.1016/j.bcmd.2014.01.002.
  41. Jin G, Yamazaki Y, Takuwa M, et al. Trib1 and Evi1 cooperate with Hoxa and Meis1 in myeloid leukemogenesis. Blood. 2007;109(9):3998–4005. doi: 10.1182/blood-2006-08-041202.
  42. Krivtsov AV, Twomey D, Feng Z, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442(7104):818–22. doi: 10.1038/nature04980.
  43. Bindels EM, Havermans M, Lugthart S, et al. EVI1 is critical for the pathogenesis of a subset of MLL-AF9-rearranged AMLs. Blood. 2012;119(24):5838–49. doi: 10.1182/blood-2011-11-393827.
  44. Glass C, Wuertzer C, Cui X, et al. Global Identification of EVI1 Target Genes in Acute Myeloid Leukemia. PLoS One. 2013;8(6):e67134. doi: 10.1371/journal.pone.0067134.
  45. Hoyt PR, Bartholomew C, Davis AJ, et al. The Evi1 proto-oncogene is required at midgestation for neural, heart, and paraxial mesenchyme development. Mech Dev. 1997;65(1–2):55–70. doi: 10.1016/s0925-4773(97)00057-9.
  46. Nucifora G. The EVI1 gene in myeloid leukemia. Leukemia. 1997;11(12):2022–31. doi: 10.1038/sj.leu.2400880.
  47. Kataoka K, Sato T, Yoshimi A, et al. Evi1 is essential for hematopoietic stem cell self-renewal, and its expression marks hematopoietic cells with long-term multilineage repopulating activity. J Exp Med. 2011;208(12):2403–16. doi: 10.1084/jem.20110447.
  48. Zhang Y, Stehling-Sun S, Lezon-Geyda K, et al. PR-domain-containing Mds1-Evi1 is critical for long-term hematopoietic stem cell function. Blood. 2011;118(14):3853–61. doi: 10.1182/blood-2011-02-334680.
  49. Steinleitner K, Rampetsreiter P, Koffel R, et al. EVI1 and MDS1/EVI1 expression during primary human hematopoietic progenitor cell differentiation into various myeloid lineages. Anticancer Res. 2012;32(11):4883–9.
  50. Wieser R. The oncogene and developmental regulator EVI1: expression, biochemical properties, and biological functions. Gene. 2007;396(2):346–57. doi: 10.1016/j.gene.2007.04.012.
  51. Xi ZF, Russell M, Woodward S, et al. Expression of the Zn finger gene, EVI-1, in acute promyelocytic leukemia. Leukemia. 1997;11(2):212–20. doi: 10.1038/sj.leu.2400547.
  52. Aytekin M, Vinatzer U, Musteanu M, et al. Regulation of the expression of the oncogene EVI1 through the use of alternative mRNA 5’-ends. Gene. 2005;356:160–8. doi: 10.1016/j.gene.2005.04.032.
  53. Niederreither K, Subbarayan Y, Dolle P, et al. Embryonic retinoic acid synthesis is essential for early mouse post-implantation development. Nat Genet. 1999;21(4):444–8. doi: 1038/7788.
  54. Valk PJ, Verhaak RG, Beijen MA, et al. Prognostically useful gene-expression profiles in acute myeloid leukemia. N Engl J Med. 2004;350(16):1617–28. doi: 10.1056/NEJMoa040465.
  55. Morishita K, Parganas E, William CL, et al. Activation of EVI1 gene expression in human acute myelogenous leukemias by translocations spanning 300–400 kilobases on chromosome band 3q26. Proc Natl Acad Sci USA. 1992;89(9):3937–41. doi: 10.1073/pnas.89.9.3937.
  56. Ogawa S, Mitani K, Kurokawa M, et al. Abnormal expression of Evi-1 gene in human leukemias. Hum Cell. 1996;9(4):323–32.
  57. Lugthart S, Groschel S, Beverloo HB, et al. Clinical, molecular, and prognostic significance of WHO type inv(3)(q21q26.2)/t(3;3)(q21;q26.2) and various other 3q abnormalities in acute myeloid leukemia. J Clin Oncol. 2010;28(24):3890–8. doi: 10.1200/JCO.2010.29.2771.
  58. Groschel S, Sanders MA, Hoogenboezem R, et al. Mutational spectrum of myeloid malignancies with inv(3)/t(3;3) reveals a predominant involvement of RAS/RTK signaling pathways. Blood. 2015;125(1):133–9. doi: 10.1182/blood-2014-07-591461.
  59. Langabeer SE, Rogers JR, Harrison G, et al. EVI1 expression in acute myeloid leukaemia. Br J Haematol. 2001;112(1):208–11. doi: 10.1046/j.1365-2141.2001.02569.x.
  60. Balgobind BV, Lugthart S, Hollink IH, et al. EVI1 overexpression in distinct subtypes of pediatric acute myeloid leukemia. Leukemia. 2010;24(5):942–9. doi: 10.1038/leu.2010.47.
  61. Matsuo H, Kajihara M, Tomizawa D, et al. EVI1 overexpression is a poor prognostic factor in pediatric patients with mixed lineage leukemia-AF9 rearranged acute myeloid leukemia. Haematologica. 2014;99(11):e225–е227. doi: 10.3324/haematol.2014.107128.
  62. Testa U, Lo-Coco F. Targeting of leukemia-initiating cells in acute promyelocytic leukemia. Stem Cell Invest. 2015;2:8. doi: 10.3978/j.issn.2306-9759.2015.04.03.
  63. Jo A, Mitani S, Shiba N, et al. High expression of EVI1 and MEL1 is a compelling poor prognostic marker of pediatric AML. Leukemia. 2015;29(5):1076–83. doi: 10.1038/leu.2015.5.
  64. Sadeghian MH, Rezaei Dezaki Z. Prognostic Value of EVI1 Expression in Pediatric Acute Myeloid Leukemia: A Systematic Review. Iran J Pathol. 2018;13(3):294–300.
  65. Arai S, Yoshimi A, Shimabe M, et al. Evi-1 is a transcriptional target of mixed-lineage leukemia oncoproteins in hematopoietic stem cells. Blood. 2011;117(23):6304–14. doi: 10.1182/blood-2009-07-234310.
  66. De Weer A, Van der Meulen J, Rondou P, et al. EVI1-mediated down regulation of MIR449A is essential for the survival of EVI1 positive leukaemic cells. Br J Haematol. 2011;154(3):337–48. doi: 10.1111/j.1365-2141.2011.08737.x.
  67. Yamazaki H, Suzuki M, Otsuki A, et al. A remote GATA2 hematopoietic enhancer drives leukemogenesis in inv(3)(q21;q26) by activating EVI1 expression. Cancer Cell. 2014;25(4):415–27. doi: 10.1016/j.ccr.2014.02.008.
  68. Groschel S, Sanders MA, Hoogenboezem R, et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell. 2014;157(2):369–81. doi: 10.1016/j.cell.2014.02.019.
  69. Lugthart S, Figueroa ME, Bindels E, et al. Aberrant DNA hypermethylation signature in acute myeloid leukemia directed by EVI1. Blood. 2011;117(1):234–41. doi: 10.1182/blood-2010-04-281337.
  70. Bartholomew C, Morishita K, Askew D, et al. Retroviral insertions in the CB-1/Fim-3 common site of integration activate expression of the Evi-1 gene. Oncogene. 1989;4(5):529–34.
  71. Kreider BL, Orkin SH, Ihle JN. Loss of erythropoietin responsiveness in erythroid progenitors due to expression of the Evi-1 myeloid-transforming gene. Proc Natl Acad Sci USA. 1993;90(14):6454–8. doi: 10.1073/pnas.90.14.6454.
  72. Kataoka K, Kurokawa M. Ecotropic viral integration site 1, stem cell self-renewal and leukemogenesis. Cancer Sci. 2012;103(8):1371–7. doi: 10.1111/j.1349-7006.2012.02303.x.
  73. Soderholm J, Kobayashi H, Mathieu C, et al. The leukemia-associated gene MDS1/EVI1 is a new type of GATA-binding transactivator. Leukemia. 1997;11(3):352–8. doi: 10.1038/sj.leu.2400584.
  74. Laricchia-Robbio L, Fazzina R, Li D, et al. Point mutations in two EVI1 Zn fingers abolish EVI1-GATA1 interaction and allow erythroid differentiation of murine bone marrow cells. Mol Cell Biol. 2006;26(20):7658–66. doi: 10.1128/MCB.00363-06.
  75. Senyuk V, Sinha KK, Li D, et al. Repression of RUNX1 activity by EVI1: a new role of EVI1 in leukemogenesis. Cancer Res. 2007;67(12):5658–66. doi: 10.1158/0008-5472.CAN-06-3962.
  76. Laricchia-Robbio L, Premanand K, Rinaldi CR, Nucifora G. EVI1 Impairs myelopoiesis by deregulation of PU.1 function. Cancer Res. 2009;69(4):1633–42. doi: 10.1158/0008-5472.CAN-08-2562.
  77. Steinmetz B, Hackl H, Slabakova E, et al. The oncogene EVI1 enhances transcriptional and biological responses of human myeloid cells to all-trans retinoic acid. Cell Cycle. 2014;13(18):2931–43. doi: 10.4161/15384101.2014.946869.
  78. Yuasa H, Oike Y, Iwama A, et al. Oncogenic transcription factor Evi1 regulates hematopoietic stem cell proliferation through GATA-2 expression. EMBO J. 2005;24(11):1976–87. doi: 10.1038/sj.emboj.7600679.
  79. Shimabe M, Goyama S, Watanabe-Okochi N, et al. Pbx1 is a downstream target of Evi-1 in hematopoietic stem/progenitors and leukemic cells. Oncogene. 2009;28(49):4364–74. doi: 10.1038/onc.2009.288.
  80. Kurokawa M, Mitani K, Irie K, et al. The oncoprotein Evi-1 represses TGF-beta signalling by inhibiting Smad3. Nature. 1998;394(6688):92–6. doi: 10.1038/27945.
  81. Izutsu K, Kurokawa M, Imai Y, et al. The corepressor CtBP interacts with Evi-1 to repress transforming growth factor beta signaling. Blood. 2001;97(9):2815–22. doi: 10.1182/blood.v97.9.2815.
  82. Kurokawa M, Mitani K, Yamagata T, et al. The evi-1 oncoprotein inhibits c-Jun N-terminal kinase and prevents stress-induced cell death. EMBO J. 2000;19(12):2958–68. doi: 10.1093/emboj/19.12.2958.
  83. Buonamici S, Li D, Mikhail FM, et al. EVI1 abrogates interferon-alpha response by selectively blocking PML induction. J Biol Chem. 2004;280(1):428–36. doi: 10.1074/jbc.M410836200.
  84. Pradhan AK, Mohapatra AD, Nayak KB, Chakraborty S. Acetylation of the proto-oncogene EVI1 abrogates Bcl-xL promoter binding and induces apoptosis. PLoS One. 2011;6(9):e25370. doi: 10.1371/journal.pone.0025370.
  85. Yatsula B, Lin S, Read AJ, et al. Identification of binding sites of EVI1 in mammalian cells. J Biol Chem. 2005;280(35):30712–22. doi: 10.1074/jbc.M504293200.
  86. Ernst T, Chase AJ, Score J, et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet. 2010;42(8):722–6. doi: 10.1038/ng.621.
  87. Figueroa ME, Lugthart S, Li Y, et al. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell. 2010;17(1):13–27. doi: 10.1016/j.ccr.2009.11.020.
  88. Wagner JM, Hackanson B, Lubbert M, Jung M. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenet. 2010;1(3–4):117–36. doi: 10.1007/s13148-010-0012-4.
  89. Senyuk V, Zhang Y, Liu Y, et al. Critical role of miR-9 in myelopoiesis and EVI1-induced leukemogenesis. Proc Natl Acad Sci USA. 2013;110(14):5594–9. doi: 10.1073/pnas.1302645110.
  90. Nikoloski G, Langemeijer SM, Kuiper RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet. 2010;42(8):665–7. doi: 10.1038/ng.620.
  91. Makishima H, Jankowska AM, Tiu RV, et al. Novel homo- and hemizygous mutations in EZH2 in myeloid malignancies. Leukemia. 2010;24(10):1799–804. doi: 10.1038/leu.2010.167.
  92. 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.
  93. Walter MJ, Ding L, Shen D, et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia. 2011;25(7):1153–8. doi: 10.1038/leu.2011.44.
  94. 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.
  95. 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.
  96. 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.
  97. van Haaften G, Dalgliesh GL, Davies H, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41(5):521–3. doi: 10.1038/ng.349.
  98. Liu Y, Chen L, Ko TC, et al. Evi1 is a survival factor which conveys resistance to both TGFbeta- and taxol-mediated cell death via PI3K/AKT. Oncogene. 2006;25(25):3565–75. doi: 10.1038/sj.onc.1209403.
  99. Yoshimi A, Goyama S, Watanabe-Okochi N, et al. Evi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteins. Blood. 2011;117(13):3617–28. doi: 10.1182/blood-2009-12-261602.
  100. Bingemann SC, Konrad TA, Wieser R. Zinc finger transcription factor ecotropic viral integration site 1 is induced by all-trans retinoic acid (ATRA) and acts as a dual modulator of the ATRA response. FEBS J. 2009;276(22):6810–22. doi: 10.1111/j.1742-4658.2009.07398.x.
  101. Pauebelle E, Plesa A, Hayette S, et al. Efficacy of All-Trans-Retinoic Acid in high-risk acute myeloid leukemia with overexpression of EVI1. Oncol Ther. 2019;7(2):121–30. doi: 10.1007/s40487-019-0095-9.
  102. Vazquez I, Maicas M, Cervera J, et al. Down-regulation of EVI1 is associated with epigenetic alterations and good prognosis in patients with acute myeloid leukemia. Haematologica. 2011;96(10):1448–56. doi: 10.3324/haematol.2011. 040535.
  103. Daghistani M, Marin D, Khorashad JS, et al. EVI-1 oncogene expression predicts survival in chronic-phase CML patients resistant to imatinib treated with second-generation tyrosine kinase inhibitors. Blood. 2010;116(26):6014–7. doi: 10.1182/blood-2010-01-264234.
  104. Мамаев Н.Н., Шакирова А.И., Бархатов И.М. идр. Ведущая роль BAALC-экспрессирующих клеток-предшественниц в возникновении и развитии посттрансплантационных рецидивов у больных острыми миелоидными лейкозами. Клиническая онкогематология. 2020;13(1):75–88. doi: 10.21320/2500-2139-2020-13-1-75-88.
    [Mamaev NN, Shakirova AI, Barkhatov IM, et al. Crucial Role of BAALCExpressing Progenitor Cells in Emergence and Development of Post-Transplantation Relapses in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2020;13(1):75–88. doi: 10.21320/2500-2139-2020-13-1-75-88. (In Russ)]
  105. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367(6464):645–8. doi: 10.1038/367645a0.
  106. Matsushita H, Yahata T, Sheng Y, et al. Establishment of a humanized APL model via the transplantation of PML-RARA-transduced human common myeloid progenitors into immunodeficient mice. PLoS One. 2014;9(11):e111082. doi: 10.1371/journal.pone.0111082.
  107. Cole CB, Verdoni AM, Ketkar S, et al. PML-RARA requires DNA methyltransferase 3A to initiate acute promyelocytic leukemia. J Clin Invest. 2016;126(1):85–98. doi: 10.1172/JCI82897.
  108. Гудожникова Я.В., Мамаев Н.Н., Бархатов И.М. и др. Результаты молекулярного мониторинга в посттрансплантационный период с помощью серийного исследования уровня экспрессии гена WT1 у больных острыми миелоидными лейкозами. Клиническая онкогематология. 2018;11(3):241–51. doi: 10.21320/2500-2139-2018-11-3-241-251.
    [Gudozhnikova YaV, Mamaev NN, Barkhatov IM, et al. Results of Molecular Monitoring in Posttransplant Period by Means of Series Investigation of WT1 Gene Expression in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2018;11(3):241–51. doi: 10.21320/2500-2139-2018-11-3-241-251. (In Russ)]
  109. Dreyfus F, Bouscary D, Melle J, et al. Expression of the Evi-1 gene in myelodysplastic syndromes. Leukemia. 1995;9(1):203–5. doi: 10.1016/0145-2126(94)90237-2.
  110. Thol F, Yun H, Sonntag AK, et al. Prognostic significance of combined MN1, ERG, BAALC, and EVI1 (MEBE) expression in patients with myelodysplastic syndromes. Ann Hematol. 2012;91(8):1221–33. doi: 10.1007/s00277-012-1457-7.
  111. Russell M, Thompson F, Spier C, Taetle R. Expression of the EVI1 gene in chronic myelogenous leukemia in blast crisis. Leukemia. 1993;7(10):1654–7.
  112. Ogawa S, Kurokawa M, Tanaka T, et al. Increased Evi-1 expression is frequently observed in blastic crisis of chronic myelocytic leukemia. Leukemia. 1996;10(5):788–94.
  113. Kuila N, Sahoo DP, Kumari M, et al. EVI1, BAALC and AME: prevalence of the secondary mutations in chronic and accelerated phases of chronic myeloid leukemia patients from eastern India. Leuk Res. 2009;33(4):594–6. doi: 10.1016/j.leukres.2008.07.018.
  114. Горбунова А.В., Гиндина Т.Л., Морозова Е.В. и др. Влияние молекулярно-генетических и цитогенетических факторов на эффективность аллогенной трансплантации костного мозга у больных хроническим миелолейкозом. Клиническая онкогематология. 2013;6(4):445–50.
    [Gorbunova AV, Gindina TL, Morozova EV, et al. Impact of molecular genetic and cytogenetic characteristics on outcomes of allogeneic hematopoietic stem cell transplantation in chronic myeloid leukemia. Klinicheskaya oncogematologiya. 2013;6(4):445–50. (In Russ)]
  115. Sato T, Goyama S, Kataoka K, et al. Evi1 defines leukemia-initiating capacity and tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Oncogene. 2014;33(42):5028–38. doi: 10.1038/onc.2014.108.
  116. Konantz M, Andre MC, Ebinger M, et al. EVI-1 modulates leukemogenic potential and apoptosis sensitivity in human acute lymphoblastic leukemia. Leukemia. 2013;27(1):56–65. doi: 10.1038/leu.2012.211.
  117. Mittal N, Li L, Sheng Y, et al. A critical role of epigenetic inactivation of miR-9 in EVI1high pediatric AML. Mol Cancer. 2019;18(1):30. doi: 10.1186/s12943-019-0952-z.
  118. Verhagen HJ, Smit MA, Rutten A, et al. Primary acute myeloid leukemia cells with overexpression of EVI-1 are sensitive to all-trans retinoic acid. Blood. 2016;127(4):458–63. doi: 10.1182/blood-2015-07-653840.
  119. Мамаев Н.Н, Горбунова А.В, Гиндина Т.Л. и др. Стойкое восстановление донорского гемопоэза у больной с посттрансплантационным рецидивом острого миеломонобластного лейкоза с inv(3)(q21q26), моносомией 7 и экспрессией онкогена EVI1 после трансфузий донорских лимфоцитов и использования гипометилирующих агентов. Клиническая онкогематология. 2014;7(1):71–5.
    [Mamayev NN, Gorbunova AV, Gindina TL, et al. Stable donor hematopoiesis reconstitution after post­transplantation relapse of acute myeloid leukemia in patient with inv(3)(q21q26), –7 and EVI1 oncogene overexpression treated by donor lymphocyte infusions and hypomethylating agents. Klinicheskaya oncogematologiya. 2014;7(1):71–5. (In Russ)]
  120. He X, Wang Q, Cen J, et al. Predictive value of high EVI1 expression in AML patients undergoing myeloablative allogeneic hematopoietic stem cell transplantation in first CR. Bone Marrow Transplant. 2016;51(7):921–7. doi: 10.1038/bmt.2016.71.
  121. Мамаев Н.Н., Морозова Е.В., Горбунова А.В. Теоретические и клинические аспекты эпигенетических изменений при миелодиспластических синдромах и острых нелимфобластных лейкозах (обзор литературы). Вестник гематологии. 2011;7(3):12–21.
    [Mamaev NN, Morozova EV, Gorbunova AV. Theoretical and practical aspects of epigenetic changes in myelodysplastic syndromes and acute non-lymphoblastic leukemias (literature review). Vestnik gematologii. 2011;7(3):12–21. (In Russ)]
  122. Mamaev N, Morozova E, Gindina T, et al. Dacogen and allogeneic bone marrow transplantation in the treatment of high-risk myelodysplastic syndromes with non-random chromosome abnormalities. Leuk Res. 2011;35(Suppl 1):72–3. doi: 10.1016/S0145-2126(11)70186-2.
  123. Mamaev N, Gorbunova A, Barkhatov I, et al. Biology and treatment of leukemia and myelodysplastic syndromes with high EVI-1 gene expression. ELN Frontiers Meeting 2012 “Myeloid neoplasms: approaching cure”. Istanbul, Turkey. Abstract No. 37.
  124. Yang X, Wong MPM, Ng RK. Aberrant DNA Methylation in Acute Myeloid Leukemia and Its Clinical Implications. Int J Mol Sci. 2019;20(18):4576. doi: 10.3390/ijms20184576.
  125. Nowek K, Sun SM, Dijkstra MK, et al. Expression of a passenger miR-9* predicts favorable outcome in adults with acute myeloid leukemia less than 60 years of age. Leukemia. 2016;30(2):303–9. doi: 10.1038/leu.2015.282.
  126. Li F, He W, Geng R, Xie X. Myeloid leukemia with high EVI1 expression is sensitive to 5-aza-2’-deoxycytidine by targeting miR-9. Clin Transl Oncol. 2020;22(1):137–43. doi: 10.1007/s12094-019-02121-y.
  127. Cattaneo F, Nucifora G. EVI1 recruits the histone methyltransferase SUV39H1 for transcription repression. J Cell Biochem. 2008;105(2):344–52. doi: 10.1002/jcb.21869.
  128. Craddock C, Quek L, Goardon N, et al. Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia. Leukemia. 2013;27(5):1028–36. doi: 10.1038/leu.2012.312.
  129. Trino S, Zoppoli P, Carella AM, et al. DNA methylation dynamic of bone marrow hematopoietic stem cells after allogeneic transplantation. Stem Cell Res Ther. 2019;10(1):138. doi: 10.1186/s13287-019-1245-6.
  130. Ahn JS, Kim YK, Min YH, et al. Azacitidine Pre-Treatment Followed by Reduced-Intensity Stem Cell Transplantation in Patients with Higher-Risk Myelodysplastic Syndrome. Acta Haematol. 2015;134(1):40–8. doi: 10.1159/000368711.
  131. Voso MT, Leone G, Piciocchi A, et al. Feasibility of allogeneic stem-cell transplantation after azacitidine bridge in higher-risk myelodysplastic syndromes and low blast count acute myeloid leukemia: results of the BMT-AZA prospective study. Ann Oncol. 2017;28(7):1547–53. doi: 10.1093/annonc/mdx154.
  132. Овечкина В.Н., Бондаренко С.Н., Морозова Е.В. и др. Роль терапии гипометилирующими препаратами перед аллогенной трансплантацией гемопоэтических стволовых клеток при острых миелоидных лейкозах и миелодиспластическом синдроме. Клиническая онкогематология. 2017;10(3):351–7. doi: 10.21320/2500-2139-2017-10-3-351-357.
    [Ovechkina VN, Bondarenko SN, Morozova EV, et al. The Role of Hypomethylating Agents Prior to Allogeneic Hematopoietic Stem Cells Transplantation in Acute Myeloid Leukemia and Myelodysplastic Syndrome. Clinical oncohematology. 2017;10(3):351–7. doi: 10.21320/2500-2139-2017-10-3-351-357. (In Russ)]
  133. Nishihori T, Perkins J, Mishra A, et al. Pretransplantation 5-azacitidine in high-risk myelodysplastic syndrome. Biol Blood Marrow Transplant. 2014;20(6):776–80. doi: 10.1016/j.bbmt.2014.02.008.
  134. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116(23):5420–31. doi: 10.1002/cncr.25500.
  135. Craddock C, Jilani N, Siddique S, et al. Tolerability and Clinical Activity of Post-Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on the RICAZA Trial. Biol Blood Marrow Transplant. 2016;22(2):385–90. doi: 10.1016/j.bbmt.2015.09.004.
  136. Marini C, Brissot E, Bazarbachi A, et al. Tolerability and Efficacy of Treatment With Azacytidine as Prophylactic or Preemptive Therapy for Myeloid Neoplasms After Allogeneic Stem Cell Transplantation. Clin Lymphoma Myel Leuk. 2020;20(6):377–82. doi: 10.1016/j.clml.2019.10.011.
  137. Бадаев Р.Ш., Заммоева Д.Б., Гиршова Л.Л. и др. Профилактическое применение азацитидина у пациентов с острыми миелоидными лейкозами после гаплоидентичной аллоТКМ. Клиническая онкогематология. 2019;12(1):37–42. doi: 10.21320/2500-2139-2019-12-1-37-42.
    [Badaev RSh, Zammoeva DB, Girshova LL, et al. Preventive Use of Azacitidine in Patients with Acute Myeloid Leukemia after Haploidentical Allo-BMT. Clinical oncohematology. 2019;12(1):37–42. doi: 10.21320/2500-2139-2019-12-1-37-42. (In Russ)]
  138. Cattaneo F, Nucifora G. EVI1 recruits the histone methyltransferase SUV39H1 for transcription repression. J Cell Biochem. 2008;105(2):344–52. doi: 10.1002/jcb.21869.
  139. Estey EH, Thall PF, Pierce S, et al. Randomized phase II study of fludarabine + cytosine arabinoside + idarubicin ± all-trans retinoic acid ± granulocyte colony-stimulating factor in poor prognosis newly diagnosed acute myeloid leukemia and myelodysplastic syndrome. Blood. 1999;93(8):2478–84. doi: 10.1182/blood.v93.8.2478.
  140. Schlenk RF, Frohling S, Hartmann F, et al. Phase III study of all-trans retinoic acid in previously untreated patients 61 years or older with acute myeloid leukemia. Leukemia. 2004;18(11):1798–803. doi: 10.1038/sj.leu.2403528.
  141. Raza A, Buonamici S, Lisak L, et al. Arsenic trioxide and thalidomide combination produces multi-lineage hematological responses in myelodysplastic syndromes patients, particularly in those with high pre-therapy EVI1 expression. Leuk Res. 2004;28(8):791–803. doi: 10.1016/j.leukres.2003.11.018.
  142. Burnett AK, Hills RK, Green C, et al. The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA. Blood. 2010;115(5):948–56. doi: 10.1182/blood-2009-08-236588.
  143. van Gils N, Verhagen HJMP, Smit L. Reprogramming acute myeloid leukemia into sensitivity for retinoic-acid-driven differentiation. Exp Hematol. 2017;52:12–23. doi: 10.1016/j.exphem.2017.04.007.
  144. Plesa A, Dumontet C, Mattei E, et al. High frequency of CD34+CD38-/low immature leukemia cells is correlated with unfavorable prognosis in acute myeloid leukemia. World J Stem Cells. 2017;9(12):227–34. doi: 10.4252/wjsc.v9.i12.227.
  145. Nguyen CH, Bauer K, Hackl H, et al. All-trans retinoic acid enhances, and a pan-RAR antagonist counteracts, the stem cell promoting activity of EVI1 in acute myeloid leukemia. Cell Death Dis. 2019;10(12):944. doi: 10.1038/s41419-019-2172-2.
  146. Field T, Perkins J, Huang Y, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  147. Kim DY, Lee JH, Park YH, et al. Feasibility of hypomethylating agents followed by allogeneic hematopoietic cell transplantation in patients with myelodysplastic syndrome. Bone Marrow Transplant. 2012;47(3):374–9. doi: 10.1038/bmt.2011.86.
  148. Jiang YZ, Su GP, Dai Y, et al. Effect of Decitabine Combined with Unrelated Cord Blood Transplantation in an Adult Patient with -7/EVI1+ Acute Myeloid Leukemia: a Case Report and Literature Review. Ann Clin Lab Sci. 2015;45(5):598–601.
  149. Schlenk RF, Lubbert M, Benner A, et al. All-trans retinoic acid as adjunct to intensive treatment in younger adult patients with acute myeloid leukemia: results of the randomized AMLSG 07-04 study. Ann Hematol. 2016;95(12):1931–42. doi: 10.1007/s00277-016-2810-z.
  150. Taussig DC, Vargaftig J, Miraki-Moud F, et al. Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood. 2010;115(10):1976–84. doi: 10.1182/blood-2009-02-206565.
  151. Patel S, Zhang Y, Cassinat B, et al. Successful xenografts of AML3 samples in immunodeficient NOD/shi-SCID IL2Rγ–/– Leukemia. 2012;26(11):2432–5. doi: 10.1038/leu.2012.154.

Chronic Myeloid Leukemia: Role of Allogeneic Hematopoietic Stem Cell Transplantation in the Era of Tyrosine Kinase Inhibitors

EV Morozova, YuYu Vlasova, MV Barabanshchikova, NN Mamaev, IM Barkhatov, AL Alyanskii, EI Darskaya, MV Vladovskaya, SN Bondarenko, IS Moiseev, BV Afanasyev

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Elena Vladislavovna Morozova, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; e-mail: dr_morozova@mail.ru

For citation: Morozova EV, Vlasova YuYu, Barabanshchikova MV, et al. Chronic Myeloid Leukemia: Role of Allogeneic Hematopoietic Stem Cell Transplantation in the Era of Tyrosine Kinase Inhibitors. Clinical oncohematology. 2020;13(2):193–8 (In Russ).

DOI: 10.21320/2500-2139-2020-13-2-193-198


ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a radical method of chronic myeloid leukemia (CML) treatment. In the 1990s CML became the most frequent indication for allo-HSCT worldwide. CML treatment drastically changed after tyrosine kinase inhibitors (TKI) had been introduced into clinical practice. However, despite considerable progress achieved in treatment, low survival rate is observed in patients with CML either diagnosed at an advanced stage or characterized with resistance, TKI intolerance, and loss of response. For such patients allo-HSCT remains the only and the best treatment solution. The present article discusses current views on the importance of allo-HSCT for CML treatment in the era of extensive use of TKIs.

Keywords: allo-HSCT, chronic myeloid leukemia, TKI.

Received: November 20, 2019

Accepted: February 28, 2020

Read in PDF


REFERENCES

  1. Афанасьев Б.В. Результаты различных видов аллогенных трансплантаций гемопоэтических стволовых клеток у детей и взрослых. Клеточная трансплантология и тканевая инженерия. 2011;6(1):11.

    [Afanasyev BV. Results of different types of allogeneic hematopoietic stem cell transplantation in children and adults. 2011;6(1):11. Kletochnaya transplantologiya i tkanevaya inzheneriya. (In Russ)]

  2. Buckner CD, Clift RA, Fefer A, et al. Treatment of blastic transformation of chronic granulocytic leukemia by high dose cyclophosphamide, total body irradiation and infusion of cryopreserved autologous marrow. Exp Hematol. 1974;2(3):138–46.

  3. Goldman JM, Catovsky D, Hows J, et al. Cryopreserved peripheral blood cells functioning as autografts in patients with chronic granulocytic leukaemia in transformation. Br Med J. 1979;1(19):1310–3. doi: 10.1136/bmj.1.6174.1310.

  4. Fefer A, Buckner CD, Thomas ED, et al. Cure of hematologic neoplasia with transplantation of marrow from identical twins. N Engl J Med. 1977;297(3):146–8. doi: 10.1056/nejm197707212970307.

  5. Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990;75(3):555–62. doi: 10.1182/blood.v75.3.555.bloodjournal753555.

  6. Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76(12):2462–5. doi: 1182/blood.v76.12.2462.bloodjournal76122462.

  7. Mukherjee S, Kalaycio M. Accelerated Phase CML: Outcomes in Newly Diagnosed vs. Progression From Chronic Phase. Curr Hematol Malig Rep. 2016;11(2):86–93. doi: 10.1007/s11899-016-0304-7.

  8. Hehlmann R, Saussele S, Voskanyan A, et al. Management of CML-blast crisis. Best Pract Res Clin Haematol. 2016;29(3):295–307. doi: 10.1016/j.beha.2016.10.005.

  9. Туркина А.Г., Зарицкий А.Ю., Шуваев В.А. и др. Клинические рекомендации по диагностике и лечению хронического миелолейкоза. Клиническая онкогематология. 2017;10(3):294–316. doi: 10.21320/2500-2139-2017-10-3-294-316.

    [Turkina AG, Zaritskii AYu, Shuvaev VA, et al. Clinical Recommendations for the Diagnosis and Treatment of Chronic Myeloid Leukemia. Clinical oncohematology. 2017;10(3):294–316. doi: 10.21320/2500-2139-2017-10-3-294-316. (In Russ)]

  10. Baccarani M, Deininger MW, Rosti G, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–84. doi: 10.1182/blood-2013-05-501569.

  11. Bacher U, Klyuchnikov E, Zabelina T, et al. The changing scene of allogeneic stem cell transplantation for chronic myeloid leukemia—a report from the German Registry covering the period from 1998 to 2004. Ann Hematol. 2009;88(12):1237–47. doi: 10.1007/s00277-009-0737-3.

  12. Любимова Л.С., Кузьмина Л.А., Урнова Е.С. и др. HLA-идентичная трансплантация костного мозга в первой хронической фазе хронического миелолейкоза в ранние сроки заболевания или длительная терапия ингибиторами тирозинкиназ? Гематология и трансфузиология. 2012;57(3):6–10.

    [Lyubimova LS, Kuz’mina LA, Urnova ES, et al. Early HLA-identical bone marrow transplantation during the chronic phase of chronic myeloid leukemia or long-term tyrosine kinase inhibitor therapy? Gematologiya i transfuziologiya. 2012;57(3):6–10. (In Russ)]

  13. Vlasova YY, Morozova EV, Shukhov OA, et al. Clinical features and outcomes in chronic myeloid leukemia with T315I mutation. Cell Ther Transplant. 2017;6(2):26–35. doi: 18620/ctt-1866-8836-2017-6-2-26-35.

  14. Nicolini FE, Basak GW, Kim D-W, et al. Overall survival with ponatinib versus allogeneic stem cell transplantation in Philadelphia chromosome-positive leukemias with the T315I mutation. Cancer. 2017;123(15):2875–80. doi: 10.1002/cncr.30558.

  15. Ozen M, Ustun C, Ozturk B, et al. Allogeneic transplantation in chronic myeloid leukemia and the effect of tyrosine kinase inhibitors on survival, a quasi-experimental study. Turkish J Hematol. 2017;34(1):16–26. doi: 10.4274/tjh.2015.0346.

  16. Saussele S, Lauseker M, Gratwohl A, et al. Allogeneic hematopoietic stem cell transplantation (allo SCT) for chronic myeloid leukemia in the imatinib era: Evaluation of its impact within a subgroup of the randomized German CML study IV. Blood. 2010;115(10):1880–5. doi: 10.1182/blood-2009-08-237115.

  17. Gratwohl A, Pfirrmann M, Zander A, et al. Long-term outcome of patients with newly diagnosed chronic myeloid leukemia: A randomized comparison of stem cell transplantation with drug treatment. Leukemia. 2016;30(3):562–9. doi: 10.1038/leu.2015.281.

  18. Chhabra S, Ahn KW, Hu ZH, et al. Myeloablative vs reduced-intensity conditioning allogeneic hematopoietic cell transplantation for chronic myeloid leukemia. Blood Adv. 2018;2(21):2922–36. doi: 10.1182/bloodadvances.2018024844.

  19. Kato K, Miyamoto T, Yonemoto K, et al. Second Allogeneic Hematopoietic Stem Cell Transplantation (allo-HSCT) for Relapse of Hematological Malignancies after First Allo-HSCT. Blood. 2014;124(21):3947. doi: 10.1182/blood.v124.21.3947.3947.

  20. Christopeit M, Tischer J, Bornhauser M, et al. Haploidentical Second Allogeneic Hematopoietic Stem Cell Transplantation for the Treatment of Acute Leukemia Relapse after First Allo-HSCT: A Retrospective Registry Analysis of 60 Patients on Behalf of the German Cooperative Transplant Group. 2014;124(21):2590. doi: 10.1182/blood.v124.21.2590.2590.

Achievements and Challenges in Allogeneic Hematopoietic Stem Cell Transplantation in Cytogenetically Unfavorable Acute Leukemias (Literature Review)

NN Mamaev, TL Gindina, BV Afanas’ev

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Nikolai Nikolaevich Mamaev, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; e-mail: nikmamaev524@gmail.com

For citation: Mamaev NN, Gindina TL, Afanas’ev BV. Achievements and Challenges in Allogeneic Hematopoietic Stem Cell Transplantation in Cytogenetically Unfavorable Acute Leukemias (Literature Review). Clinical oncohematology. 2019;12(2):111–9.

DOI: 10.21320/2500-2139-2019-12-2-111-119


ABSTRACT

Literature review provides the analysis of treatment results of implementing allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with cytogenetically unfavorable acute myeloid and lymphoblastic leukemias including monosomal, complex, and hyperdiploid karyotypes, t(3;3)/inv(3), t(v;11)(v;q23), t(4;11)(q21;q23), t(9;22)(q34;q11) translocations, 17p abnormalities, and some other disorders. The major disadvantage of allo-HSCT seems to be linked to a strong chromosome-damaging effect of cytostatic drugs used in conditioning regimens which in turn is associated with additional chromosome abnormalities occurring in tumors, increasing genomic instability, and tumor progression. On the other hand, one of the advantages of allo-HSCT can consist in its specific “graft versus leukemia” (GVL) effect whose degree has not yet been adequately studied. To minimize the risks of allo-HSCT in above mentioned patients it appears appropriate to apply new treatment approaches based on de-escalation of chromosome- and whole-genome-damaging effects and also to introduce recent methods of active stimulation and qualitative assessment of GVL effect into clinical practice.

Keywords: acute leukemias, cytogenetically unfavorable variants, allo-HSCT, outcomes, additional chromosome abnormalities, “graft versus leukemia” effect.

Received: October 22, 2018

Accepted: February 2, 2019

Read in PDF 


REFERENCES

  1. Burnett AK, Hills RK. Who should be transplanted in first remission of acute myeloid leukemia? Curr Treatment Opt Oncol. 2011;12(4):329–40. doi: 10.1007/s11864-011-0169-x.

  2. Stelljes M, Beelen DW, Braess J, et al. Allogeneic transplantation as post-remission therapy for cytogenetically high-risk acute myeloid leukemia: landmark analysis from a single prospective multicenter trial. Haematologica. 2011;96(7):972–9. doi: 3324/haematol.2011.041004.

  3. Bejanyan N, Weisdorf DJ, Logan BR, et al. Survival of patients with acute myeloid leukemia relapsing after allogeneic hematopoietic cell transplantation: A Center for International Blood and Marrow Transplant Research Study. Biol Blood Marrow Transpl. 2015;21(3):454–9. doi: 10.1016/j.bbmt.2014.11.007.

  4. Dohner H, Estey EH, Grimwade D, et al. Diagnosis and management of acute myeloid leukemia in adults: 2017 ELN recommendation from an international expert panel. Blood. 2017;129(4):424–47. doi: 10.1182/blood-2016-08-733196.

  5. Vasu S, Kohlschmidt J, Mrozek K, et al. Ten-year outcome of patients with acute myeloid leukemia not treated with allogeneic transplantation in first complete remission. Blood Adv. 2018;2(13):1645–50. doi: 10.1182/bloodadvances.2017015222.

  6. de Lima M, Porter DL, Battiwalla M, et al. Proceedings from the National Cancer Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: Part III. Prevention and Treatment of Relapse after Allogeneic Transplantation. Biol Blood Marrow Transpl. 2014;20(1):4–13. doi: 1016/j.bbmt.2013.08.012.

  7. Christopeit М, Kroger N, Haferlach T, et al. Relapse assessment following allogeneic SCT in patients with MDS and AML. Ann Hematol. 2014;93(7):1097–110. doi: 1007/s00277-014-2046-8.

  8. Tsirigotis Р, Byrne M, Schmid C, et al. Relapse of AML after hematopoietic stem cell transplantation: methods of monitoring and preventive strategies. A review from the ALWP of the EBMT. Bone Marrow Transplant. 2016;51(11):1431–8. doi: 1038/bmt.2016.167.

  9. Cruz NM, Mencia-Trinchant N, Hassane DC, et al. Minimal residual disease in acute myelogenous leukemia. Int J Lab Hematol. 2017;39(Suppl 1):53–60. doi: 1111/ijlh.12670.

  10. Kroger N, Bishop M, Giralt S, et al. Third International workshop on the biology, prevention, and treatment of relapse after stem cell transplantation. Bone Marrow Transplant. 2018;53(1):1–2. doi: 10.1038/bmt.2017.218.

  11. Nahi H, Remberger M, Machaczka M, et al. Different impact of intermediate and unfavorable cytogenetics at the time of diagnosis of de novo AML after allo-SCT: a long-term retrospective analysis from a single institution. Med Oncol. 2012;29(4):2348–58. doi: 10.1007/s12032-011-0155-y.

  12. Cornelissen JJ, Blaise D. Hemopoietic stem cell transplantation for patients with AML in first complete remission. Blood. 2016;127(1):62–70. doi: 10.1182/blood-2015-07-604546.

  13. Gindina T, Mamaev N, Afanasyev B. Chromosome Abnormalities and Hematopoietic Stem Cell Transplantation. In: ML Larramendy, S Soloneski, eds. Chromosomal abnormalities – a hallmark manifestation of genomic instability. Croatia: INTECH; 2017. рр. 71–86. doi: 5772/67802.

  14. Hemmati PG, Schulze-Luchkov A, Terwey ThN, et al. Cytogenetic risk grouping by the monosomal karyotype classification is superior in predicting the outcome of acute myeloid leukemia undergoing allogeneic stem cell transplantation in complete remission. Eur J Hematol. 2013;92(2):102–10. doi: 10.1111/ejh.12216.

  15. Wang Y, Liu QF, Qin YZ, et al. Improved outcome of hematopoietic stem cell transplantation in a poor prognostic subgroup patients with mixed-lineage-leukemia-rearranged acute leukemia: results from a prospective, multicenter study. Am J Hematol. 2014;89(2):130–6. doi: 10.1002/ajh.23593.

  16. Parma M, Vigano C, Fumagalli M, et al. Good outcome for very high risk adult B cell acute lymphoblastic leukemia carring genetic abnormalities t(4;11)(q21;q23) or t(9;22)(q34;q11), if promtly submitted to allogeneic transplantation after obtaining a good molecular remission. Mediterr J Hematol Infect Dis. 2015;7(1):e2015041. doi: 10.4084/MJHID.2015.041.

  17. Fang M, Storer B, Estey E, et al. Outcome of patients with acute myeloid leukemia with monosomal karyotype who undergo hematopoietic stem cell transplantation. Blood. 2011;118(6):1490–4. doi: 10.1182/blood-2011-02-339721.

  18. Oran B, Dolan M, Cao Q, et al. Monosomal karyotype provides better prognostic prediction after allogeneic stem cell transplantation in patients with acute myelogenous leukemia. Biol Blood Marrow Transplant. 2011;17(3):356–64. doi: 10.1016/j.bbmt.2010.05.012.

  19. Cornelissen JJ, Breems D, van Putten WL, et al. Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories. J Clin Oncol. 2012;30(17):2140–6. doi: 10.1200/jco.2011.39.6499.

  20. Guo RJ, Atenafu FG, Craddock K, et al. Allogeneic hematopoietic cell transplantation may alleviate the negative prognostic impact of monosomal and complex karyotype on patients with acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(5):690–5. doi: 10.1016/j.bbmt.2014.01.027.

  21. Pasquini M, Zhang M-J, Medeiros BC, et al. Hematopoietic cell transplantation outcomes in monosomal karyotype myeloid malignancies. Biol Blood Marrow Transplant. 2016;22(2):248–57. doi: 10.1016/j.bbmt.2015.08.024.

  22. Гиндина Т.Л., Мамаев Н.Н., Бондаренко С.Н. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при острых миелоидных лейкозах: прогностическое значение сложного кариотипа, включающего аномалии del(5q), –7, del(7q). Клиническая онкогематология. 2016;9(3):271–8. doi: 21320/2500-2139-2016-9-3-271-278.

    [Gindina TL, Mamaev NN, Bondarenko SN, et al. Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias: Prognostic Significance of Complex Karyotype Including del(5q), –7, del(7q) Abnormalities. Clinical oncohematology. 2016;9(3):271–8. 2016;9(3):271–8. doi: 10.21320/2500-2139-2016-9-3-271-278. (In Russ)]

  23. Koh K, Tomozawa D, Moriya Saito AM, et al. Early use of allogeneic hematopoietic stem cell transplantation for infants with MLL gene rearrangement-positive acute lymphoblastic leukemia. Leukemia. 2015;29(2):290–6. doi: 1038/leu.2014.172.

  24. Гиндина Т.Л., Мамаев Н.Н., Паина О.В.и др. Острый лимфобластный лейкоз c транслокацией t(4;11)(q21;q23)/KMT2A-AFF1: результаты аллогенной трансплантации гемопоэтических стволовых клеток у детей и взрослых. Клиническая онкогематология. 2017;10(3):342–50. doi: 21320/2500-2139-2017-10-3-342-350.

    [Gindina TL, Mamaev NN, Paina OV, et al. Acute Lymphoblastic Leukemia with t(4;11)(q21;q23)/KMT2A-AFF1 Translocation: The Results of Allogeneic Hematopoietic Stem Cells Transplantation in Children and Adults. Clinical oncohematology. 2017;10(3):342–50. doi: 10.21320/2500-2139-2017-10-3-342-350. (In Russ)]

  25. Poire X, Labopin M, Maertens J, et al. Allogeneic stem cell transplantation in adult patients with acute myeloid leukemia and 17p abnormalities in first complete remission: a study from the Acute Leukemia Working Party (ALWP) of the European Society for Blood and Marrow Transplantation (EBMT). J Hematol Oncol. 2017;10(1):20. doi: 10.1186/s13045-017-0393-3.

  26. Strickland SA, Sun Z, Ketterling RP, et al. Independent prognostic significance of monosomy 17 and impact of karyotype complexity in monosomal karyotype/complex karyotype acute myeloid leukemia: Results from FOUR ECOG-AGRIN prospective therapeutic trials. Leuk Res. 2017;59:55–64. doi: 10.1016/j.leukres.2017.05.010.

  27. Halaburda K, Labopin M, Houhou M, et al. AlloHSCT for inv(3)(q21;q26)t(3;3)(q21;q26) AML: a report from the acute leukemia working party of the European society for blood and marrow transplantation. Bone Marrow Transplant. 2018;53(6):683–91. doi: 10.1038/s41409-018-0165-x.

  28. Passerini V, Ozeri-Galai E, de Pagter MS, et al. The presence of extra chromosomes leads to genomic instability. Nat Commun. 2016;7(1):10754. doi: 10.1038/ncomms10754.

  29. Schmidt-Hieber M, Blau IW, Richter G, et al. Cytogenetic studies in acute leukemia patients relapsing after allogeneic stem cell transplantation. Cancer Gen Cytogenet. 2010;198(2):135–43. doi: 1016/j.cancergencyto.2010.01.005.

  30. Гиндина Т.Л., Мамаев Н.Н., БондаренкоС.Н. и др. Сложные хромосомные нарушения у больных с посттрансплантационными рецидивами острых лейкозов: клинические и теоретические аспекты. Клиническая онкогематология. 2015;8(1):69–77. doi: 10.21320/2500-2139-2015-8-1-69-77.

    [Gindina TL, Mamaev NN, Bondarenko SN, et al. Complex Chromosomal Aberrations in Patients with Post-Transplantation Relapses of Acute Leukemias: Clinical and Theoretical Aspects. Clinical oncohematology. 2015;8(1):69–77. doi: 10.21320/2500-2139-2015-8-1-69-77. (In Russ)]

  31. Breems DA, Van Putten WLL, 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.

  32. Kayzer S, Zucknick M, Dohner K, et al. Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies. Blood. 2011;119(2):551–8. doi: 10.1182/blood-2011-07-367508.

  33. Ciurea SM, Labopin G, Socie G, et al. Relapse and survival after transplantation for complex karyotype acute myeloid leukemia: a report from the acute leukemia working party of the European society for Blood and Marrow Transplantation and the University of Texas MD Anderson Cancer Center. 2018;124(10):2134–41. doi: 10.1002/cncr.31311.

  34. Schoch C, Haferlach T, Haase D, et al. Patients with the de novo acute myeloid leukemia and complex karyotype aberrations show a poor prognosis despite intensive treatment: a study of 90 patients. Br J Haematol. 2001;112(1):118–26. doi: 10.1046/j.1365-2141.2001.02511.x.

  35. Mrozek K. Cytogenetic, molecular genetics, 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.

  36. Schoch C, Kern W, Kohlmann A, et al. Acute myeloid leukemia with a complex aberrant karyotype is a distinct biological entity characterized by genomic imbalance and a special gene expression profile. Genes Chromos Cancer. 2005;43(3):227–38. doi: 1002/gcc.20193.

  37. Гиндина Т.Л., Мамаев Н.Н., Бархатов И.М. и др. Сложные повреждения хромосом у больных с рецидивами острых лейкозов после аллогенной трансплантации гемопоэтических стволовых клеток. Терапевтический архив. 2012;84(8):61–6. [Gindina TL, Mamaev NN, Barkhatov IM, et al. Complex chromosome damages in patients with recurrent acute leukemias after allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2012;84(8):61–6. (In Russ)]

  38. Bacher U, Haferlach T, Alpermann T, et al. Comparison of cytogenetic clonal evolution patterns following allogeneic hematopoietic transplantation versus convential treatment in patients at relapse of AML. Biol Blood Marrow Transplant. 2010;16(12):1649–57. doi: 10/1016/j.bbmt.2010.06.007.

  39. Chen Y, Kantarjian H, Pierce S, et al. Prognostic significance of 11q23 aberrations in adult acute myeloid leukemia and the role of allogeneic stem cell transplantation. Leukemia. 2013;27(4):836–42. doi: 1038/leu.2012.319.

  40. Yang H, Huang S, Zhu C-Y, et al. The superiority of allogeneic hematopoietic stem cell transplantation over chemotherapy alone in the treatment of acute myeloid leukemia patients with mixed lineage leukemia (MLL) rearrangements. Med Sci Monitor. 2016;22:2315–23. doi: 12659/MSM.899186.

  41. Pigneux A, Labopin M, Maertens J, et al. Outcome of allogeneic hematopoietic stem-cell transplantation in adult patients with AML and 11q23/MLL rearragement (MLL-r-AML). Leukemia. 2015;29(12):2375–81. doi: 1038/leu.2015.143.

  42. Gindina T, Mamaev N, Alyanskiy A, et al. Outcome of allogeneic hematopoietic stem cell transplantation in patients with KMT2A (MLL)-related leukemia, depending on number of transplanted CD34+ cells. Bone Marrow Transplant. 2015;50(Suppl 1):S481.

  43. Гиндина Т.Л., Мамаев Н.Н., Николаева Е.С. и др. Исход аллогенной трансплантации гемопоэтических стволовых клеток при острых миелоидных лейкозах с гипердиплоидным кариотипом. Клиническая онкогематология. 2016;9(4):383–90. doi: 21320/2500-2139-2016-9-4-383-390.

    [Gindina TL, Mamaev NN, Nikolaeva ES, et al. Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias with Hyperdiploid Karyotype. Clinical oncohematology. 2016;9(4):383–90. doi: 10.21320/2500-2139-2016-9-4-383-390. (In Russ)]

  44. Chevallier P, Labopin M, Nagler A, et al. Outcome after allogeneic transplantation for adult acute myeloid leukemia patients exhibiting isolated trisomy 8 chromosomal abnormality: a survey on behalf of the ALWP of the EBMT. Bone Marrow Transplant. 2009;44(9):589–94. doi: 10.1038/bmt.2009.68.

  45. Konuma T, Kondo T, Yamashita T, et al. Outocome of allogeneic hematopoietic stem cell transplantation in adult patients with acute myeloid leulkemia harboring trisomy 8. Ann Hematol. 2017;96(3):469–78. doi: 1007/s00277-016-2009-2.

  46. Herold T, Metzeler KH, Vosberg S, et al. Isolated trisomy 13 defines a homogenous AML subgroup with high frequency of mutations in splisome genes and poor prognosis. Blood. 2014;124(8):1304–11. doi: 10.1182/blood-2013-12-540716.

  47. Mohr B, Schetelig J, Schafer-Eckart K, et al. Impact of allogeneic haematopoietic stem cell transplantation in patients with abnl(17p) acute myeloid leukemia. Br J Haematol. 2013;161(2):237–44. doi: 10.1111/bjh.12253.

  48. Middeke JM, Fang M, Cornellisen JJ, et al. Outcome of patients with abnl(17p) acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Blood. 2014;123(19):2960–7. doi: 10.1182/blood-201312-544957.

  49. Vey N, Thomas X, Picard C, et al. Allogeneic stem cell transplantation improves the outcome of adults with t(1;19)/E2A-PBX1 and t(4;11)/MLL-AF4 positive B-cell acute lymphoblastic leukemia: results of the prospective multicenter LALA-94 study. Leukemia. 2006;20:2155–61. doi: 10.1038/sj.leu.2404420.

  50. Marks DI, Moorman AV, Chilton L, et al. The clinical characteristics, therapy and outcome of 85 adults with acute lymphoblastic leukemia and t(4;11)(q21q23)/MLL-AFF1 prospectively treated in the UKALLXII/ECOG2993 trial. 2013;98(6):945–52. doi: 10.3324/haematol.2012.081877.

  51. Cimino G, Cenfra N, Elia L, et al. The therapeutic response and clinical outcome of adults with ALL1(MLL)/AF4 fusion positive acute lymphoblastic leukemia according to the GIMEMA experience. Haematologica. 2010;95(5):837–40. doi: 10.3324/haematol.2009.009035.

  52. Kato M, Hasegawa D, Koh K, et al. Allogeneic haematopoietic stem cell transplantation for infant acute lymphoblastic leukemia with KMT2A (MLL) rearrangements: a retrospective study from the paediatric acute lymphoblastic leukemia working group of the Japan Society for Haematopoietic Cell Transplantation. Br J Haematol. 2014;168(4):564–70. doi: 10.1111/bjh.13174.

  53. Ribera JM, Oriol A, Gonzalez M, et al. Concurrent intensive chemotherapy and imatinib before and after stem cell transplantation in newly diagnosed Philadelphia chromosome‐positive acute lymphoblastic leukemia. Final results of the CSTIBES02 trial. 2010;95(1):87–95. doi: 10.3324/haematol.2009.011221.

  54. Ribera JM, Garcia O, Montesinos P, et al. Treatment of young patients with Philadelphia chromosome-positive acute lymphoblastic leukemia using increased dose of imatinib and deintensified chemotherapy before allogeneic stem cell transplantation. Br J Haematol. 2012;159(1):78–81. doi: 10.1111/j.1365-2141.2012.09240.x.

  55. Kebriaie P, Saliba R, Rondon G, et al. Long-term follow-up of allogeneic hematopoietic stem cell transplantation for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: impact of tyrosine kinase inhibitors on treatment outcomes. Biol Blood Marrow Transplant. 2012;18(4):584–92. doi: 10.1016/j.bbmt.2011.08.011.

  56. Aldoss I, Stiller T, Cao TM, et al. Impact of additional cytogenetic abnormalities in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia undergoing allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2015;21(7):1326–9. doi: 10.1016/j.bbmt.2015.03.021.

  57. Chiaretti S, Foa R. Management of adult Ph‐positive acute lymphoblastic leukemia. 2015;2015(1):406–13. doi: 10.1182/asheducation‐2015.1.406.

  58. Giebel S, Labopin M, Gorin NC, et al. Improving results of autologous stem cell transplantation for Philadelphia-positive acute lymphoblastic leukemia in the era of tyrosine kinase inhibitors: A report from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Eur J Cancer. 2014;50(2):411–7. doi: 10.1016/j.ejca.2013.08.027.

  59. Giebel S, Labopin M, Potter M, et al. Comparable results of autologous and allogeneic haematopoietic stem cell transplantation for adults with Philadelphia-positive acute lymphoblastic leukaemia in first complete molecular remission: An analysis by the Acute Leukemia Working Party of the EBMT. Eur J Cancer. 2018;96:73–81. doi: 10.1016/j.ejca.2018.03.018.

  60. Gindina TL, Mamaev NN, Nikolaeva ES, et al. Results of allogeneic hematopoietic stem cell transplantation in a mixed cohort of patients with Ph-positive acute lymphoblastic leukemia. Cellular Therapy and Transplantation. 2017;6(1):10–9. doi: 10.18620/ctt-1866-8836-2017-6-1-10-19.

  61. Maino E, Scattolin AM, Viero P, et al. Modern immunotherapy of adult B-lineage acute lymphoblastic leukemia with monoclonal antibodies and chimeric antigen receptor modified T cells. Mediterr J Hematol Infect Dis. 2015;7(1):e2015001. doi: 10.4884/mjhid.2015001.

  62. Zhang J-P, Zhang R, Tsao Sh-T, et al. Sequential allogeneic and autologous CAR-T–cell therapy to treat an immune-compromised leukemic patient. Blood Adv. 2018;2(14):1691–5. doi: 10.1182/bloodadvances.2018017004.

  63. Ziser R. Introduction to a review series on strategies to improve GVL effects. Blood. 2018;131(10):1039. doi: 10.1182/blood-2017-11-814467.

Results of Molecular Monitoring in Posttransplant Period by Means of Series Investigation of WT1 Gene Expression in Patients with Acute Myeloid Leukemia

YaV Gudozhnikova, NN Mamaev, IM Barkhatov, VA Katerina, TL Gindina, AI Shakirova, SN Bondarenko, OA Slesarchuk, EI Darskaya, OV Paina, LS Zubarovskaya, BV Afanas’ev

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Prof. Nikolai Nikolaevich Mamaev, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)233-12-43; e-mail: nikmamaev524@gmail.com

For citation: Gudozhnikova YaV, Mamaev NN, Barkhatov IM, et al. Results of Molecular Monitoring in Posttransplant Period by Means of Series Investigation of WT1 Gene Expression in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2018;11(3):241–51.

DOI: 10.21320/2500-2139-2018-11-3-241-251


ABSTRACT

Aim. To demonstrate diagnostic and prognostic significance of series measurement of WT1 expression in patients with acute myeloid leukemia (AML) after allogenic hematopoietic stem cell transplantation (allo-HSCT).

Materials & Methods. The clinical trial included 88 AML patients (38 females (43 %) and 50 males (57 %) aged 2–68, median 30 years). All the patients received allo-HSCT. Bone marrow was aspirated before (D0) and after HSCT (D+30, D+60, and D+100).

Results. The univariate analysis showed statistically significant differences in 2-year overall survival with respect to the following factors: with and without remission at the moment of HSCT (< 0.001), with and without chronic graft vs. host disease (cGVHD) (= 0.002), primary or secondary (MDS) AML (= 0,028), WT1 gene expression < and > 250 copies before HSCT (< 0.001) and at time points D+60 (= 0.012), and D+100 (< 0.001). Multivariate analysis revealed similar statistical significance of differences among patients transplanted in remission (= 0.041) and with cGVHD (= 0.03). In univariate analysis statistically significant differences in 2-year event-free survival (EFS) were found: a) in patients with allo-HSCT, either in remission or not (< 0.001); b) using HSC, but not bone marrow, as transplant source (p < 0.026); c) with normal or high WT1 expression at the stage of HSCT (< 0.001) and at time point D+100 (< 0.001); d) using HSC from related or unrelated donor (= 0.006); e) in patients with cGVHD (= 0.05). In multivariate analysis independent positive effect on EFS was observed only in patients with normal WT1 expression at D+100 (= 0.011) and with cGVHD (= 0.038). Cumulative incidence of posttransplant relapse (PTR) in AML patients with normal or high WT1 expression at the stage of HSCT within the 2-year follow-up was significantly different (28.2 vs. 58.9 %; = 0.002), also in measurements of this parameter at D+60 and D+100 (= 0.015 and < 0.001, respectively). In 1/4 of patients cytological relapses (cPTR) appeared considerably later than molecular relapses (mPTR), i.e. 13–489 days later (median 35 days), which is accounted for by early preventive therapy aimed at cPTR prophylaxis against the background of already recorded mPTR. According to our data, GVHD plays a crucial role in cPTR management.

Conclusion. Phenomenon of WT1 expression normalization after allo-HSCT in AML patients proves to have a high diagnostic and prognostic significance. Introduction of this approach into clinical practice seems highly advisable for national oncohematological centers.

Keywords: acute myeloid leukemia, allo-HSCT, posttransplant relapse, diagnostics and treatment with molecular monitoring of WT1 expression, graft vs. host disease.

Received: January 20, 2018

Accepted: April 18, 2018

Read in PDF 


REFERENCES

  1. Cilloni D, Gottardi E, De Micheli D, et al. Quantitative assessment of WT1 expression by real time quantitative PCR may be a useful tool for monitoring minimal residual disease in acute leukemia patients. Leukemia. 2002;16(10):2115–21. doi: 10:1038/sj.leu.2402675.
  2. Cilloni D, Gottardi E, Fava M, et al. Usefulness of quantitative assessment of the WT1 gene transcript as a marker for minimal residual disease detection. Blood. 2003;102(2);773–4. doi: 1182/blood-2003-03-0980.
  3. Ogawa H, Tamaki H, Ikegame K, et al. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogeneic stem cell transplantation in acute type leukemia. Blood. 2003;101(5):1698–704. doi: 1182/blood-2002-06-1831.
  4. Zhao X-S, Jin S, Zhu H-H, et al. Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2011;47(4):499–507. doi: 10.1038/bmt.2011.121.
  5. Мамаев Н.Н., Горбунова А.В., Бархатов И.М. и др. Молекулярный мониторинг течения острых миелоидных лейкозов по уровню экспрессии гена WT1 после аллогенной трансплантации гемопоэтических стволовых клеток. Клиническая онкогематология. 2015;8(3):309–20. doi: 10.21320/2500-2139-2015-8-3-309-320.[Mamaev NN, Gorbunova AV, Barkhatov IM, et al. Molecular Monitoring of WT1 Gene Expression Level in Acute Myeloid Leukemias after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2015;8(3):309–20. doi: 10.21320/2500-2139-2015-8-3-309-320. (In Russ)]
  6. Мамаев Н.Н., Гудожникова Я.В., Горбунова А.В.  Гиперэкспрессия гена WT1при злокачественных опухолях системы крови: теоретические и клинические аспекты (обзор литературы). Клиническая онкогематология. 2016;9(3):257–64. doi: 10.21320/2500-2139-2016-9-3-257-264.[Mamaev NN, Gudozhnikova YaV, Gorbunova AV. WT1 Gene Overexpression in Oncohematological Disorders: Theoretical and Clinical Aspects (Literature Review). Clinical oncohematology. 2016;9(3):257–64. doi: 10.21320/2500-2139-2016-9-3-257-264. (In Russ)]
  7. Call KM, Gieser T, Ito CI, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor gene locus. Cell. 1990;60(3):509–20. doi: 10:1016/0092-8674(90)90601-a.
  8. Rose EA, Glaser T, Jones C, et al. Complete physical map of the WAGR region of 11p13 localizes a candidate Wilms’ tumor gene. 1990;60(3):495–508. doi: 10.1016/0092-8674(90)90600-j.
  9. Miwa H, Beran M, Saunders GF. Expression of the Wilms’ tumor gene (WT1) in human leukemias. Leukemia. 1992;6(5):405–9.
  10. Inoue K, Sugiyama H, Ogava H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood. 1994;84(9):3071–9.
  11. Inoue K, Ogawa H, Sonoda Y, et al. Aberrant overexpression of the Wilms’ tumor gene (WT1) in human leukemia. Blood. 1997;88(4):1405–12.
  12. Cilloni D, Gottardi E, Messa F, et al. Significant correlation between the degree of WT1 expression and the International Scoring System score in patients with myelodysplastic syndromes. J Clin Oncol. 2003;21(10):1988–95. doi: 10.1200/jco.2003.10.503.
  13. Alonso-Domingues JM, Tenorio M, Velasco D, et al. Correlation of WT1 expression with the burden of total and residual leukemic blasts in bone marrow samples of acute myeloid leukemia patients. Cancer Genet. 2012;205(4):190–1. doi: 10.1016/j.cancergen.2012.02.008.
  14. Cilloni D, Messa F, Arruga F, et al. Early prediction of treatment outcome in acute myeloid leukemia by measurement of WT1 transcript levels in peripheral blood samples collected after chemotherapy. Haematologica. 2008;93(6):921–4. doi: 10.3324/haematol.12165.
  15. Ogava H, Ikegame K, Kawakami M, Tamaki H. WT1 gene transcript assay for relapse in acute myeloid leukemia after transplantation. Leuk Lymphoma. 2004;45(9):1747–53. doi: 10.1080/10428190410001687503.
  16. Pozzi S, Geroldi S, Tedone E, et al. Leukemia relapse after allogeneic transplant for acute myeloid leukemia: predictive role of WT1 expression. Br J Haematol. 2013;160(4);503–9. doi: 10.1111/bjh.12181.
  17. Nendedeu J, Esquirol A, Carricondo M, et al. Bone marrow WT1 levels in allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia and myelodysplasia: Clinically relevant time-points and 100 copies threshold value. Biol Blood Marrow Transplant. 2017;24(1):55–63. doi: 10.1016/j.bbmt.2017.09.001.
  18. Cilloni D, Saglio G, Gottardi E, et al. WT1 as universal marker for minimal residual disease detection and quantification in myeloid leukemias and in myelodysplastic syndrome. Acta Hematol. 2004;112(1–2):79–84. doi: 10.1159/000077562.
  19. Candoni A, Toffoletti E, Galina R, et al. Monitoring of minimal residual disease by quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem cell transplantation in acute myeloid leukemia. Clin Transpl. 2011;25(2):308–16. doi: 10.1111/j.1399-0012.2010.01251.x.
  20. Kwon M, Martinez-Laperche C, Infante M, et al. Evaluation of minimal residual disease by real-time quantitative PCR of Wilms’ Tumor 1 expression in patients with acute myelogenous leukemia after allogeneic stem cell transplantation: Correlation with flow cytometry and chimerism. Biol Blood Marrow Transplant. 2012;18(8):1235–42. doi: 10.1016/j.bbmt.2012.01.012.
  21. Polak J, Hajkova H, Haskovec C, et al. Quantitative monitoring of WT1 expression in peripheral blood before and after allogeneic stem cell transplantation for acute myeloid leukemia – a useful tool for early detection of minimal residual disease. Neoplasma. 2013;60(01):74–82. doi: 10.4149/neo_2013_011.
  22. Lapillone H, Renneville A, Auvrignon A, et al. High WT1 expression after induction therapy predicts high risk or relapse and death in pediatric acute myeloid leukemia. J Clin Oncol. 2006;24(10):1507–15. doi: 10.1200/jco.2005.03.5303.
  23. Messina C, Sala E, Carrabba M, et al. Early post-allogeneic transplantation WT1 transcript positivity predicts AML relapse. 40th EBMT Meeting. 30 March – 2 April; Milan, Italy; 2014: Abstract P239.
  24. Mear J-B, Salaun V, Dina N, et al. WT1 and flow cytometry minimal residual disease follow-up after allogeneic transplantation in practice. 40th EBMT Meeting. 30 March – 2 April; Milan, Italy; 2014: Abstract P655.
  25. Capelli D, Attolico I, Saraceli F, et al. Early cumulative incidence of relapse in 80 acute myeloid leukemia patients after chemotherapy and transplant post-consolidation treatment prognostic role of post-induction WT1. 40th EBMT Meeting. 30 March – 2 April; Milan, Italy; 2014: Abstract P287.
  26. Rossi G, Carella AM, Minervini MM, et al. Optimal time-points for minimal residual disease monitoring change on the basis of the method used in patients with acute myeloid leukemia who underwent allogeneic stem cell transplantation: A comparison between multiparameter flow cytometry and Wilms’ tumor 1 expression. Leuk Res. 2015;39(2):138–43. doi: 1016/j.leukres.2014.11.011.

Allogeneic Hematopoietic Stem Cell Transplantation in Myelofibrosis

MV Barabanshchikova, EV Morozova, VV Baikov, IM Barkhatov, NN Mamaev, SN Bondarenko, AL Alyanskii, LS Zubarovskaya, BV Afanas’ev

R.M. Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician I.P. Pavlov First St. Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Lyudmila Stepanovna Zubarovskaya, DSci, Professor, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)338-62-64; e-mail: zubarovskaya_ls@mail.ru

For citation: Barabanshchikova MV, Morozova EV, Baikov VV, et al. Allogeneic Hematopoietic Stem Cell Transplantation in Myelofibrosis. Clinical oncohematology. 2016;9(3):279-86 (In Russ).

DOI: 10.21320/2500-2139-2016-9-3-279-286


ABSTRACT

Background & Aims. At present, the allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only treatment option with curative potential in patients with myelofibrosis (MF), especially in intermediate and high risk categories. The aim of the study is to perform a retrospective analysis of allo-HSCT outcomes in MF patients.

Materials & Methods. Outcomes of allo-HSCT in 11 intermediate-2 (= 3) and high (= 6) risk patients (based on Dynamic International Prognostic Scoring Scale, DIPSSplus) performed in the R.M. Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation over the period from 2005 till 2015 were analyzed in the study. Two more patients underwent allo-HSCT in MF blast phase. Two patients received ruxolitinib before allo-HSCT and 1 patient before and after allo-HSCT. Reduced intensity conditioning regimen was used in all cases.

Results. Primary engraftment was documented in 8 patients. 72 % of patients achieved complete hematological remission. Molecular remission and myelofibrosis regression were confirmed in 5 patients. 5 of 11 patients were still with remission and followed-up by the date of the paper submission. The overall two-year survival was 46 %.

Conclusion. Allo-HSCT is an effective treatment option for MF patients. Further trials are required to evaluate an optimal timing for allo-HSCT in MF patients and efficacy of Janus kinase (JAK) inhibitors as pre- and posttransplant therapy in MF.


Keywords: myelofibrosis, allo-HSCT, reduced intensity conditioning regimen, ruxolitinib.

Received: January 28, 2016

Accepted: March 22, 2016

Read in  PDF (RUS)pdficon


REFERENCES

  1. Cervantes F, Dupriez B, Pereira A, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood. 2009;113(13):2895–901. doi: 10.1182/blood-2008-07-170449.
  2. Passamonti F, Rumi E, Caramella M, et al. A dynamic prognostic model to predict survival in post-polycythemia vera myelofibrosis. Blood. 2008;111(7):3383–7. doi: 10.1182/blood-2007-11-121434.
  3. Passamonti F, Rumi E, Arcaini L, et al. Prognostic factors for thrombosis, myelofibrosis, and leukemia in essential thrombocythemia: a study of 605 patients. Haematologica. 2008;93(11):1645–51. doi: 10.3324/haematol.13346.
  4. Dupriez BB, Morel P, Demory JL, et al. Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system. Blood. 1996;88(3):1013–8.
  5. Passamonti F, Cervantes F, Vannucchi AM, et al. A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood. 2009;115(9):1703–8. doi: 10.1182/blood-2009-09-245837.
  6. Gangat N, Caramazza D, Vaidya R, et al. DIPSS Plus: A Refined Dynamic International Prognostic Scoring System for Primary Myelofibrosis That Incorporates Prognostic Information From Karyotype, Platelet Count, and Transfusion Status. J Clin Oncol. 2011;29(4):392–7. doi: 10.1200/jco.2010.32.2446.
  7. Vannucchi AM, Guglielmelli P, Rotunno G, et al. Mutation-Enhanced International Prognostic Scoring System (MIPSS) for Primary Myelofibrosis: An AGIMM & IWG-MRT Project. ASH; 2014. Abstract 405.
  8. Verstovsek S, Mesa R, Gotlib J, et al. Efficacy, safety, and survival with ruxolitinib in patients with myelofibrosis: results of a median 3-year follow-up of COMFORT-I. Haematologica. 2015;100(4):479–88. doi: 10.3324/haematol.2014.115840.
  9. Kvasnicka HM, Thiele J, Bueso-Ramos CE, et al. Long-Term Effects of Ruxolitinib on Bone Marrow Morphology in Patients With Myelofibrosis and Comparison to Best Available Therapy. Haematologica. 2014;14: Abstract S155. doi:10.1016/j.clml.2014.06.098.
  10. Giorgino T, Scott BL, Ditschkowski M, et al. CME Article Impact of allogeneic stem cell transplantation on survival of patients less than 65 years of age with primary myelofibrosis. Blood. 2015;125(21):3347–51. doi: 10.1182/blood-2014-10-608315.
  11. Kroger N, Holler E, Kobbe G, et al. Allogeneic stem cell transplantation after reduced-intensity conditioning in patients with myelofibrosis: a prospective, multicenter study of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Blood. 2009;114(26):5264–70. doi: 10.1182/blood-2009-07-234880.
  12. Thiele J, Kvasnica HM, Facchetti F, et al. European consensus on grading bone marrow fibrosis and assessment of cellularity. Haematologica. 2005;90(8):1128–32.
  13. Jagasia MH, Greinix HT, Arora M, et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21(3):389–401. doi: 10.1016/j.bbmt.2014.12.001.
  14. Kroger N, Zabelina T, Alchalby H, et al. Dynamic of bone marrow fibrosis regression predicts survival after allogeneic stem cell transplantation for myelofibrosis. Biol Blood Marrow Transplant. 2014;20(6):812–5. doi: 10.1016/j.bbmt.2014.02.019.
  15. Slot S, Smits K, van de Donk NW, et al. Effect of conditioning regimens on graft failure in myelofibrosis: a retrospective analysis. Bone Marrow Transplant. 2015;11;1424–31. doi: 10.1038/bmt.2015.172.
  16. Shanavas M, Popat U, Michaelis LC, et al. Outcomes of Allogeneic Hematopoietic Cell Transplantation in Patients with Myelofibrosis with Prior Exposure to Janus Kinase 1/2 Inhibitors. Biol Blood Marrow Transplant. 2016;22(3):432–40. doi: 10.1016/j.bbmt.2015.10.005.
  17. Verstovsek S, Kantarjian H, Mesa RA, et al. Safety and Efficacy of INCB018424, a JAK1 and JAK2 Inhibitor, in Myelofibrosis. N Engl J Med. 2010;363(12):1117–27. doi: 10.1056/nejmoa1002028.
  18. Stubig T, Alchalby H, Ditschkowski M, et al. JAK inhibition with ruxolitinib as pretreatment for allogeneic stem cell transplantation in primary or post-ET/PV myelofibrosis. Leukemia. 2014;28(8):1736–8. doi: 10.1038/leu.2014.86.
  19. Jaekel N, Behre G, Behning A, et al. Allogeneic hematopoietic cell transplantation for myelofibrosis in patients pretreated with the JAK1 and JAK2 inhibitor ruxolitinib. Bone Marrow Transplant. 2014;49(2):179–84. doi: 10.1038/bmt.2013.173.

Results of Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Acute Myeloid Leukemia with t(8;21)(q22;q22)/RUNX1-RUNX1T1 and Additional Cytogenetic Abnormalities

TL Gindina, NN Mamaev, SN Bondarenko, ES Nikolaeva, OA Slesarchuk, AS Borovkova, OV Paina, SV Razumova, AL Alyanskii, LS Zubarovskaya, BV Afanas’ev

R.M. Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician I.P. Pavlov First St. Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Tat’yana Leonidovna Gindina, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)233-12-43; e-mail: cytogenetics.bmt.lab@gmail.com

For citation: Gindina TL, Mamaev NN, Bondarenko SN, et al. Results of Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Acute Myeloid Leukemia with t(8;21)(q22;q22)/RUNX1-RUNX1T1 and Additional Cytogenetic Abnormalities. Clinical oncohematology. 2016;9(2):148–54 (In Russ).

DOI: 10.21320/2500-2139-2016-9-2-148-154


ABSTRACT

Aim. To evaluate the impact of additional chromosomal aberrations on outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with acute myeloid leukemia (AML) with t(8;21)(q22;q22)/RUNX1-RUNX1T1 translocation.

Methods. Twenty-five AML patients with t(8;21)(q22;q22)/RUNX1-RUNX1T1 translocation (10 women and 15 men, aged from 2 to 58 years; median 20.2) were examined. Analysis of overall (OS) and event-free survival (EFS) predictors after allo-HSCT in patients with different clinical, transplant and cytogenetic characteristics was performed.

Results. The additional cytogenetic abnormalities were found in 13 (52 %) patients before the transplantation, at that, complex karyotype with three or more chromosomal abnormalities were registered in 9 (69 %) patients. The univariate analysis showed that OS and EFS after allo-HSCT differed in patients with different characteristics such as age (= 0.03; = 0.0006), clinical status at transplantation (= 0.0002; = 0,006), donor type (= 0.0003; = 0.002), the interval from diagnosis of leukemia to allo-HSCT (= 0,008, for OS only), additional cytogenetic abnormalities (= 0.03; = 0.009) and complex karyotype (= 0.004; = 0.0003), respectively. In multivariate analysis, independent predictors of OS were donor type (= 0.01), the interval from diagnosis of leukemia to allo-HSCT (= 0.01), and additional cytogenetic abnormalities in karyotype (= 0.04), as well as donor type (= 0.04) and patient’s age (= 0.004) for EFS.

Conclusion. AML with t(8;21)(q22;q22)/RUNX1-RUNX1T1 translocation is a heterogeneous disease. The prognosis in patients with the additional cytogenetic abnormalities, especially in those with the complex karyotype, is worse both after the standard chemotherapy (i.e. before allo-HSCT), and after allo-HSCT.


Keywords: AML with t(8;21) translocation, allo-HSCT, cytogenetic abnormalities.

Received: February 6, 2016

Accepted: February 15, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Mrozek K, Bloomfield CD. Chromosomal abnormalities in acute leukemia and their clinical importance. In: Rowley JD, et al, eds. Chromosomal translocations and genome rearrangements in cancer. Switzerland: Springer International Publishing; 2015. pp. 275–306. doi: 10.1007/978-3-319-19983-2_13.
  2. Klein K, Kaspers G, Harrison CJ, et al. Clinical impact of additional cytogenetic aberrations, cKIT and RAS mutations, and treatment elements in pediatric t(8;21)-AML: results from an international retrospective study by the international Berlin-Frankfurt-Munster study group. J Clin Oncol. 2015;33(36):4247. doi: 10.1200/jco.2015.61.1947.
  3. Krauth MT, Eder C, Alpermann T, et al. High number of additional genetic lesions in acute myeloid leukemia with t(8;21)/RUNX1-RUNX1T1: frequency and impact on clinical outcome. Leukemia. 2014;28(7):1449–58. doi:10.1038/leu.2014.4.
  4. Byrd JC, Dodge RK, Carroll A, et al. Patients with t(8;21)(q22;q22) and acute myeloid leukemia have superior failure-free and overall survival when repetitive cycles of high-dose cytarabine are administered. J Clin Oncol. 1999;17:3767–75.
  5. Numata A, Fujimaki K, Aoshima T, et al. Retrospective analysis of treatment outcomes in 70 patients with t(8;21) acute myeloid leukemia. Jpn J Clin Oncol. 2012;53(7):698–704.
  6. Kuwatsuka Y, Miyamura K, Suzuki R, et al. Hematopoietic cell transplantation for core binding factor acute myeloid leukemia: t(8;21) and inv(16) represent different clinical outcomes. Blood. 2009;113(9):2096–103. doi: 10.1182/blood-2008-03-145862/
  7. Shlenk RF, Benner A, Krauter J, et al. Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol. 2004;22(18):3741–50. doi: 10.1200/JCO.2004.03.012.
  8. Shlenk RF, Pasquini MC, Perez WS, et al. HLA-identical sibling allogeneic transplant versus chemotherapy in acute myelogenous leukemia with t(8;21) in first complete remission: collaborative study between the German AML Intergroup and CIBMTR. Biol Blood Marrow Transplant. 2008;14(2):187–96. doi: 10.1016/j.bbmt.2007.10.006.
  9. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Трансплантация гемопоэтических стволовых клеток при остром миелоидном лейкозе с транслокацией t(8;21)(q22;q22). Клиническая онкогематология. 2013;6(4):439–50.
    [Mamaev NN, Gorbunova AV, Gindina TL, et al. Hematopoietic stem cell transplantation in AML patients with t(8;21) (q22;q22) translocation. Klinicheskaya onkogematologiya. 2013;6(4):439–50. (In Russ)]
  10. Appelbaum FR, Kopecky KJ, Tallman MS, et al. The clinical spectrum of adult acute myeloid leukemia associated with core binding factor translocations. Br J Haematol. 2006;135(2):165–73. doi: 10.1111/j.1365-2141.2006.06276.x.
  11. Yoon JH, Kim HJ, Kim JW, et al. Identification of molecular and cytogenetic risk factors for unfavorable core-binding factor-positive adult AML with post-remission treatment outcome analysis including transplantation. Bone Marrow Transplant. 2014;49(12):1466–74. doi: 10.1038/bmt.2014.180.
  12. Marcucci G, Mrozek K, Ruppert AS, et al. Prognostic factors and outcome of core binding factor acute myeloid leukemia patients with t(8;21) differ from those of patients with inv(16): a Cancer and Leukemia Group B Study. J Clin Oncol. 2005;23(24):5705–17. doi: 10.1200/jco.2005.15.610.
  13. Qin YZ, Zhu HH, Jiang Q, et al. Prevalence and prognostic significance of c-KIT mutations in core binding factor acute myeloid leukemia: a comprehensive large-scale study from a single Chinese center. Leuk Res. 2016;38(12):1435–40. doi: 10.1016/j.leukres.2014.09.017.
  14. Mosna F, Papayannidis C, Martinelli G, et al. Complex karyotype, older age, and reduced first-line dose intensity determine poor survival in core binding factor acute myeloid leukemia patients with long-term follow-up. Am J Hematol. 2015;90(6):515–23. doi: 10.1002/ajh.24000.
  15. Гиндина Т.Л., Мамаев Н.Н., Бархатов И.М. и др. Сложные повреждения хромосом у больных с рецидивами острых лейкозов после аллогенной трансплантации гемопоэтических стволовых клеток. Терапевтический архив. 2012;8:61–6.
    [Gindina TL, Mamaev NN, Barhatov IM, et al. Complex chromosome damages in patients with recurrent acute leukemias after allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2012;8:61–6. (In Russ)]
  16. Schaffer L, McGovan-Jordan J, Schmid M. ISCN. An international System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013.
  17. Gindina T, Mamaev N, Nikolaeva E, et al. Jumping translocations in a 13-year-old child with RUNX1/RUNX1T1-positive acute myeloid leukemia. 10th European Cytogenetics Conference 2015. Chromosome Res. 2015;23(Suppl 1):88. doi: 10.1007/s10577-015-9476-6.
  18. Мамаев Н.Н., Горбунова А.В., Бархатов И.М. и др. Молекулярный мониторинг течения острых миелоидных лейкозов по уровню экспрессии гена WT1 после аллогенной трансплантации гемопоэтических стволовых клеток. Клиническая онкогематология. 2015;8(3):309–20.
    [Mamaev NN, Gorbunova AV, Barkhatov IM, et al. Molecular Monitoring of WT1 Gene Expression Degree in Acute Myeloid Leukemias after Allogeneic Hematopoietic Stem Cell Transplantation. Klinicheskaya onkogematologiya. 2015;8(3):309–20. (In Russ)]
  19. Mamaev N, Mamaeva S. Two cases of acute myeloblastic leukemia (M2-type) with karyotypes 45X,-X,t(6;8)(q27;q22),inv(9) and 46,XY, t(8;21)(q22;q22),del(9)(q22). Cancer Genet Cytogenet. 1985;18(2):105–11. doi: 10.1016/0165-4608(85)90060-3.

 

Impact of molecular genetic and cytogenetic characteristics on outcomes of allogeneic hematopoietic stem cell transplantation in chronic myeloid leukemia

A.V. Gorbunova, T.L. Gindina, E V. Morozova, I.M. Barkhatov, N.N. Mamayev, and B.V. Afanasyev

R.M. Gorbacheva Institute of Pediatric Oncology, Hematology and Transplantology, I.P. Pavlov State Medical University, Saint Petersburg, Russian Federation


ABSTRACT

Point mutations in the BCR-ABL kinase domain, BCR-ABL and EVI1 gene expression alterations, and additional chromosomal aberrations in Philadelphia chromosome-positive chronic myeloid leukemia are strongly associated with resistance to tyrosine kinase inhibitors (TKIs) and disease progression, but their effect on the outcome of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is uncertain. This retrospective study included 35 CML patients with resistance to TKI therapy who received a related or unrelated HSCT. Additional chromosomal aberrations were associated with the decreased rate of the complete molecular response (CMR) after allo-HSCT. EVI1 expression level was associated with a decreased disease-free survival (DFS). BCR-ABL kinase domain mutations showed no influence on CMR, OS, and DFS in this patient cohort. 9 out of 10 patients with T315I mutation achieved CMR. EVI1-directed stratification of patients during the post-transplantation period may improve outcome of HSCT.


Keywords: chronic myeloid leukemia, CML, allogeneic hematopoietic stem cells transplantation, allo-HSCT, BCR-ABL, EVI1.

Read in PDF (RUS)pdficon


Refernces

  1. Oyekunle A., Klyuchnikov E., Ocheni S. et al. Challenges for allogeneic hematopoietic stem cell transplantation in chronic myeloid leukemia in the era of tyrosine kinase inhibitors. Acta Haematol. 2011; 126(1): 30–9.
  2. Baccarani M., Cortes J., Pane F. et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. Clin. Oncol. 2009; 27(35): 6041–51.
  3. Soverini S., Hochhaus A., Nicolini F.E. et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011; 118(5): 1208–15.
  4. Cortes J.E., Kantarjian H., Shah N.P. et al. Ponatinib in refractory Philadelphia chromosome-positive leukemias. Engl. J. Med. 2012; 367(22): 2075–88.
  5. Hochhaus A., Kreil S., Corbin A.S. et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia 2002; 16: 2190–6.
  6. Wang Y., Cai D., Brendel C. et al. Adaptive secretion of granulocytemacrophage colony-stimulating factor (GM-CSF) mediates imatinib and nilotinib resistance in BCR/ABL+ progenitors via JAK-2/STAT-5 pathway activation. Blood 2007; 109: 2147–55.
  7. Chu S., Holtz M., Gupta M. et al. BCR/ABL kinase inhibition by imatinib mesylate enhances MAP kinase activity in chronic myelogenous leukemia CD34+ cells. Blood 2004; 103: 3167–74.
  8. Burchert A., Wang Y., Cai D. et al. Compensatory PI3-kinase/Akt/mTOR activation regulates imatinib resistance development. Leukemia 2005; 19: 1774–82.
  9. Daghistani M., Marin D., Khorashad J.S. et al. EVI-1 oncogene expression predicts survival in chronic-phase CML patients resistant to imatinib treated with second-generation tyrosine kinase inhibitors. Blood 2010; 116(26): 6014–7.
  10. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миело- диспластические синдромы с экспрессией гена EVI1: теоретические и клинические аспекты. Клин. онкогематол. 2012; 5(4): 361–4. [Mamayev N.N., Gorbunova A.V., Gindina T.L. i dr. Leykozy i miyelodis_ plasticheskiye sindromy s vysokoy ekspressiyey gena EVI1: teoreticheskiye i klinicheskiye aspekty (Leukemias and myelodisplastic syndromes with high EVI1 gene expression: theoretical and clinical aspects. In: Clin. oncohematol.). Klin. onkogematol. 2012; 5(4): 361–4.]
  11. Groschel S., Lugthart S., Schlenk R.F. et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. Clin. Oncol. 2010; 28(12): 2101–7.

 

Management of refractory acute lymphoblastic leukemia in children and adolescents: remission re-induction followed by allogeneic hematopoietic stem cell transplantation

E.V. Semenova, N.V. Stancheva, S.N. Bondarenko, V.N. Vavilov, D.A. Bagge, O.V. Paina, S.V. Razumova, A.S. Borovkova, T.A. Bykova, A.A. Raths, L.S. Zuborovskaya, B.V. Afanasyev

R.M. Gorbacheva Memorial Institute of Children Hematology and Transplantation, Pavlov State Medical University, Saint-Petersburg, Russian Federation


ABSTRACT

Objective: to analyze efficacy of fludarabine- and nelarabine-containing chemotherapy  before  allogeneic hematopoietic stem cell transplantation (allo-HSCT) in children and adolescents with relapsed and refractory acute lymphoblastic leukemia (ALL) .

Patients and methods: Thirty three patients (pts) with relapsed and refractory ALL from 1 till 21 y.o. (median age was 11,5 y.o.) were treated by fludarabine- (n=23) and nelarabine- (n=10) containing therapy.  Subsequently 24 patients underwent allo-HSCT.  

Results: CR was achieved in 11(48%) of 23 pts after FLAG±IDA and 7 (70%) of 10 pts after nelarabine-containg regimens. Duration of remissions were 1-18 months (median was 4,9 months).

Overall 3-year survival (OS) after allo-HSCT in remission was 58%, OS  after allo-HSCT in relapse was 8%, OS without allo-HSCT was 0%.

Conclusion: Fludarabine- and nelarabine-containing therapy can be used as bridge to allo-HSCT in children and adolescents with poor-prognosis ALL.


Кeywords: fludarabine, nelarabine, allo-HSCT, relapsed or refractory acute lymphoblastic leukemia, children and adolescents

Read in PDF (RUS)pdficon


REFERENCES

 

  1. Schrappe M., Reiter A., Ludwig W.D. et al. Improved outcome in childhood acute Lymphoblastic leukemia despite reduced use of antracyclines and cranial radiotherapy: results of trial ALL-BFM 90. Blood 2000; 95(11): 3310–22.
  2. Silverman L.B., Gelber R.D., Dalton V.K. et al. Improved outcome for children with acute lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91-01. Blood 2001; 97(5): 1211–8.
  3. Pui C.-H., Schrappe M., Ribeiro R.C., Niemeyer C.M. Childhood and adolescent lymphoid and myeloid leukemia. Hematology 2004; 84: 124–32.
  4. Pui C.H., Evans W.E. Treatment of acute lymphoblastic leukemia. N. Engl. J. Med. 2006; 354(2): 166–78.
  5. Chessells J.M., Veys P., Kempski H. et al. Long-term follow-up of relapsed childhood acute lymphoblastic leukaemia. Br. J. Haematol. 2003; 123: 396–405.
  6. Nguyen K., Devidas M., Cheng S.C. et al. Factors influencing survival after relapse from acute lymphoblastic leukemia: A Children’s Oncology Group study. Leukemia 2008; 22: 2142–50.
  7. Афанасьев Б.В., Зубаровская Л.С. Трансплантация гемопоэтических стволовых клеток крови. Детская онкология: Руководство. СПб., 2002: 90–108. [Afanas’ev B.V., Zubarovskaya L.S. Transplantatsiya gemopoeticheskikh stvolovykh kletok krovi. Detskaya onkologiya: Rukovodstvo (Hematopoietic stem cell transplantation. Pediatric oncology. Manual). , 2002: 90–108.]
  8. Румянцев А.Г., Масчан А.А. Трансплантация гемопоэтических стволовых клеток у детей: Руководство для врачей. М., 2003. [Rumyantsev A.G., Maschan A.A. Transplantatsiya gemopoeticheskikh stvolovykh kletok u detei: Rukovodstvo dlya vrachei (Hematopoietic stem cell transplantation in children. Manual for medical practitioners)., 2003.]
  9. McCarthy A.J., Pitcher L.A., Hann I.M., Oakhill A. FLAG (fludarabine, high-dose cytarabine, and G-CSF) for refractory and high-risk relapsed acute leukemia in children. Med. Pediatr. Oncol. 1999; 32(6): 411–5.
  10. Yang S.W. et al. Dual mode of inhibition of purified DNA ligase from human cells by 9-β-D-arabinosyl-2-fluoroadenine triphosphate. J. Biol. Chem. 1992; 267: 2345–9.
  11. Ross S.P. et al. Fludarabine: a review of its pharmacological properties and therapeutic potential in malignancy. Drug 1993; 45: 737–59.
  12. Gandhi V. et al. Combination of fludarabine and arabinosyl-cytosine for the treatment of chronic lymphocytic leukemia: clinical efficacy and modulation of arabinosyl-cytosine pharmacology. Cancer Chem. Pharmacol. 1994; 34: 30–6.
  13. Tavil B., Aytac S., Balci Y.I. et al. Fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin (FLAG-IDA) for the treatment of children with poor-prognosis acute leukemia: the Hacettepe experience. Hematol. Oncol. 2010; 27(7): 517–28.
  14. Quarello P., Berger M., Rivetti E. et al. FLAG-liposomal doxorubicin (Myocet) regimen for refractory or relapsed acute leukemia pediatric patients. J. Pediatr. Hematol. Oncol. 2012; 34(3): 208–16.
  15. Berg S.L., Blaney S.M., Devidas M. et al. Phase II study of Nelarabine in children and young adults with refractory T-cell malignancies: a report from the Children’s Oncology Group. J. Clin. Oncol. 2005; 23(15): 3376–82.
  16. Commander L.A., Seif A.E., Insogna I.G., Susan R. Rheingold. Salvage therapy with nalarabine, etoposide and cyclophosphamide in relapsed/refractory paediatric T-cell lymphoblastic leukaemia and lymphoma. J. Haem. 2010; 150: 345–51.
  17. Trotti A., Colevas A.D. et al. CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment. Radiat. O 2007; 13: 176–81.