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.

Acute Leukemias: Immunophenotypic Differences between Blast Cells and Their Nonneoplastic Analogues in Bone Marrow

АM Popov1, ТYu Verzhbitskaya2,3, LG Fechina2, AV Shestopalov1,4, SA Plyasunova1

1 Dmitrii Rogachev Federal Scientific Clinical Centre of Pediatric Hematology, Oncology and Immunology, 1 Samory Mashela str., Moscow, Russian Federation, 117997

2 Regional Children’s Hospital No. 1, 32 Serafimy Deryabinoi str., Yekaterinburg, Russian Federation, 620149

3 Institute of Medical Cell Technologies, 22a Karla Marksa str., Yekaterinburg, Russian Federation, 620026

4 N.I. Pirogov Russian National Research Medical University, 1 Samory Mashela str., Moscow, Russian Federation, 117997

For correspondence: Aleksandr Mikhailovich Popov, PhD, 1 Samory Mashela str., Moscow, Russian Federation, 117997; Tel.: +7(495)287-65-70; e-mail: uralcytometry@gmail.com

For citation: Popov AM, Verzhbitskaya TYu, Fechina LG, et al. Acute Leukemias: Immunophenotypic Differences between Blast Cells and Their Nonneoplastic Analogues in Bone Marrow. Clinical oncohematology. 2016;9(3):302-13 (In Russ).

DOI: http://dx.doi.org/10.21320/2500-2139-2016-9-3-302-313


ABSTRACT

Flow cytometry immunophenotyping of bone marrow tumor blasts is one of the principal methods used for acute leukemia (AL) diagnosing. Normal lymphopoietic and myelopoietic progenitors have very similar antigenic profile with leukemic cells, thus, making the AL diagnosing more difficult. Genetic disorders resulting in formation of a tumor clone contribute to development of an immunophenotype that differs from normal cells. Aberrant expression of markers detected in AL blast cells alone forms a so-called leukemia-associated immunophenotype. The leukemia-associated immunophenotype detection by multicolor flow cytometry permits distinguishing between normal and neoplastic cells. This requires simultaneous assessment of many markers on the same cells, which is possible only if multicolor flow cytometry with well-designed and well-established antibodies panels is used. Moreover, correct interpretation of the cell population location on dot plot requires adequate cytometer setup, standardized sample preparation and enough experienced personnel. That is why correct immunophenotyping is often possible only in large laboratories performing reference immunophenotyping within the frames of multicenter trials.

Keywords: acute leukemias, flow cytometry, antigenic expression, immunophenotype.

Received: February 19, 2016

Accepted: March 16, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Morike A, Zimmermann M, Reiter A, et al. Long-term results of five consecutive trials in childhood acute lymphoblastic leukemia performed by the ALL-BFM study group from 1981 to 2000. Leukemia. 2010;24(2):265–84. doi: 10.1038/leu.2009.257.
  2. Pui CH, Carroll WL, Meshinchi S, Arceci RJ. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J Clin Oncol. 2011;29(5):551–65. doi: 10.1200/jco.2010.30.7405.
  3. Pui CH, Mullighan CG, Evans WE, Relling MV. Pediatric acute lymphoblastic leukemia: where are we going and how do we get there? Blood. 2012;120(6):1165–74. doi: 10.1182/blood-2012-05-
  4. Bene M, Castoldi G, Knapp W, et al. Proposals for the immunological classification of acute leukemias. European Group for the Immunological Characterization of Leukemias (EGIL). Leukemia. 1995;9(10):1783–6.
  5. van Lochem EG, Wiegers YM, van den Beemd R, et al. Regeneration pattern of precursor-B-cells in bone marrow of acute lymphoblastic leukemia patients depends on the type of preceding chemotherapy. Leukemia. 2000;14(4):688–95. doi: 10.1038/sj.leu.2401749.
  6. McKenna RW, Washington LT, Aquino DA, et al. Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662 consecutive bone marrow specimens by 4-color flow cytometry. Blood. 2001;98(8):2498–507. doi: 10.1182/blood.v98.8.2498.
  7. Campana D, Coustan-Smith E. Advances in the immunological monitoring of childhood acute lymphoblastic leukaemia. Best Pract Res Clin Hematol. 2002;15(1):1–19. doi: 1053/beha.2002.0182.
  8. Dworzak MN, Fritsch G, Fleischer C, et al. Comparative phenotype mapping of normal vs. malignant pediatric B-lymphopoiesis unveils leukemia-associated aberrations. Exp Hematol. 1998;26(4):305–13.
  9. Lucio P, Parreira A, van den Beemd MVM, et al. Flow cytometric analysis of normal B cell differentiation: a frame of reference for the detection of minimal residual disease in precursor-B-ALL. Leukemia. 1999;13(3):419–27. doi: 1038/sj.leu.2401279.
  10. Lucio P, Gaipa G, van Lochem EG, et al. BIOMED-I concerted action report: flow cytometric immunophenotyping of precursor B-ALL with standardized triple-stainings. Leukemia. 2001;15(8):1185–92. doi: 10.1038/sj.leu.2402150.
  11. Dworzak MN, Fritsch G, Fleisher C, et al. Multiparameter phenotype mapping of normal and post-chemotherapy B lymphopoiesis in pediatric bone marrow. Leukemia. 1997;11(8):1266–73. doi: 10.1038/sj.leu.2400732.
  12. Попов А.М., Вержбицкая Т.Ю., Цаур Г.А. и др. Аберрации иммунофенотипа, применимые для мониторинга минимальной остаточной болезни методом проточной цитометрии при CD10-позитивном остром лимфобластном лейкозе из В-линейных предшественников. Иммунология. 2010;31(6):299–304.
    [Popov AM, Verzhbitskaya TYu, Tsaur GA, et al. Immunophenotype aberrations used for monitoring of the minimal residual disease using flow cytometry in CD10-positive acute lymphoblastic leukemia from B-linear precursors. 2010;31(6):299–304. (In Russ)]
  13. Мовчан Л.В. Лейкоз-ассоциированный иммунофенотип опухолевых клеток у детей с острым лимфобластным лейкозом из предшественников В-лимфоцитов. Онкогематология. 2012;1:22–8.
    [Movchan LV. Leukemia-associated immunophenotype of tumor cells in childhood B-precursors acute lymphoblastic leukemia. Onkogematologiya. 2012;1:22–8. (In Russ)]
  14. Попов А.М., Вержбицкая Т.Ю., Цаур Г.А. и др. Алгоритм применения проточной цитометрии для мониторинга минимальной остаточной болезни при CD10-негативном остром лимфобластном лейкозе из B-линейных предшественников. Вопросы диагностики в педиатрии. 2012;4(5):31–5.
    [Popov AM, Verzhbitskaya TYu, Tsaur GA, et al. Methodology of flow cytometry application for minimal residual disease monitoring in childhood CD10-negative B-cell precursor acute lymphoblastic leukemia. Voprosy diagnostiki v pediatrii. 2012;4(5):31–5. (In Russ)]
  15. Ciudad J, Orfao A, Vidriales B, et al. Immunophenotypic analysis of CD19+ precursors in normal human adult bone marrow: implications for minimal residual disease detection. Haematologica. 1998;83(12):1069–75.
  16. Veltroni M, De Zen L, Sanzari MC, et al. Expression of CD58 in normal, regenerating and leukemic bone marrow B cells: implications for the detection of minimal residual disease in acute lymphocytic leukemia. Haematologica. 2003;88(11):1245–52.
  17. van Lochem EG, van der Velden VH, Wind HK, et al. Immunophenotypic differentiation patterns of normal hematopoiesis in human bone marrow: reference patterns for age-related changes and disease-induced shifts. Cytometry B Clin Cytom. 2004;60B(1):1–13. doi: 10.1002/cyto.b.20008.
  18. Lee RV, Braylan RC, Rimsza LM. CD58 expression decreases as nonmalignant B cells mature in bone marrow and is frequently overexpressed in adult and pediatric precursor B-cell acute lymphoblastic leukemia. Am J Clin Pathol. 2005;123(1):119–24. doi: 1309/x5vv6fkjq6mublpx.
  19. Robillard N, Cave H, Mechinaud F, et al. Four-color flow cytometry bypasses limitations of IG/TCR polymerase chain reaction for minimal residual disease detection in certain subsets of children with acute lymphoblastic leukemia. Haematologica. 2005;90(11):1516–23.
  20. Seegmiller AC, Kroft SH, Karandikar NJ, McKenna RW. Characterization of immunophenotypic aberrancies in 200 cases of B acute lymphoblastic leukemia. Am J Clin Pathol. 2009;132(6):940–9. doi: 10.1309/AJCP8G5RMTWUEMUU.
  21. Sedek L, Bulsa J, Sonsala A, et al. The immunophenotypes of blast cells in B-cell precursor acute lymphoblastic leukemia: how different are they from their normal counterparts. Cytometry B Clin Cytom. 2014;86(5):329–39. doi: 10.1002/cyto.b.21176.
  22. Hulspas R, O’Gorman MRG, Wood BL, et al. Consideration for the control of background fluorescence in clinical flow cytometry. Cytometry B Clin Cytom. 2009;76В(6):355–64. doi: 10.1002/cyto.b.20485.
  23. Hrusak O, Porwit-MacDonald A. Antigen expression patterns reflecting genotype of acute leukemias. Leukemia. 2002;16(7):1233–58. doi: 10.1038/sj.leu.2402504.
  24. Попов А.М., Цаур Г.А., Вержбицкая Т.Ю. и др. Иммунофенотипическая характеристика острого лимфобластного лейкоза у детей первого года жизни. Онкогематология. 2012;7(2):14–24. doi: 17650/1818-8346-2012-7-2-14-24.
    [Popov AM, Tsaur GA, Verzhbitskaya TY, et al. Immunophenotypic investigation of infant acute lymphoblastic leukemia. Oncohematology. 2012;7(2):14–24. doi: 10.17650/1818-8346-2012-7-2-14-24. (In Russ)]
  25. Fuda FS, Karandikar NJ, Chen W. Significant CD5 expression on normal stage 3 hematogones and mature B-lymphocytes in bone marrow. Am J Clin Pathol. 2009;132(5):733–7. doi: 10.1309/AJCPU5E3NXEKLFIY.
  26. Gaipa G, Basso G, Maglia O, et al. Drug-induced immunophenotypic modulation in childhood ALL: implications for minimal residual disease detection. Leukemia. 2005;19(1):49–56. doi: 10.1038/sj.leu.2403559.
  27. Gaipa G, Basso G, Ratei R, et al. Reply to van der Sluijs-Gelling, et al. Leukemia. 2005;19(12):2351–2. doi: 10.1038/sj.leu.2403912.
  28. van der Sluijs-Gelling AJ, van der Velden VHJ, Roeffen ETJM, et al. Immunophenotypic modulation in childhood precursor-B-ALL can be mimicked in vitro and is related to the induction of cell death. Leukemia. 2005;19(10):1845–7. doi: 10.1038/sj.leu.2403911.
  29. Dworzak MN, Schumich A, Printz D, et al. CD20 up-regulation in pediatric B-cell precursor acute lymphoblastic leukemia during induction treatment: setting the stage for anti-CD20 directed immunotherapy. Blood. 2008;112(10):3982–8. doi: 10.1182/blood-2008-06-
  30. Gaipa G, Basso G, Aliprandi S, et al. Prednisone induces immunophenotypic modulation of CD10 and CD34 in nonapoptotic B-cell precursor acute lymphoblastic leukemia cells. Cytometry B Clin Cytom. 2008;74B(3):150–5. doi: 10.1002/cyto.b.20408.
  31. Попов А.М., Вержбицкая Т.Ю., Цаур Г.А. и др. Изменения иммунофенотипа опухолевых бластов при CD10-позитивном остром лимфобластном лейкозе у детей к 15-му дню индукционной терапии по протоколу ALL-MB-2008. Иммунология. 2010;31(2):60–4.
    [Popov AM, Verzhbitskaya TYu, Tsaur GA, et al. Changes of tumor blast immunophenotype in CD10-positive acute lymphoblastic leukemia in children by the 15th day of induction therapy according to the ALL-MB-2008 protocol. Immunologiya. 2010;31(2):60–4. (In Russ)]
  32. Мовчан Л.В., Шман Т.В., Белевцев М.В. и др. Изменение иммунофенотипа лейкемических клеток на этапах индукционной терапии острого лимфобластного лейкоза из предшественников В-лимфоцитов у детей. Вопросы гематологии/онкологии и иммунопатологии в педиатрии. 2011;10(1):21–6. [Movchan LV, Shman TV, Belevtsev MV, et al. Immunophenotypic modulation of the leukemic cells during induction therapy in children with B-cell precursor acute lymphoblastic leukemia. Voprosy gematologii/onkologii i immunopatologii v pediatrii. 2011;10(1):21–6. (In Russ)]
  33. Dworzak MN, Gaipa G, Schumich A, et al. Modulation of antigen expression in B-cell precursor acute lymphoblastic leukemia during induction therapy is partly transient: evidence for a drug-induced regulatory phenomenon. Results of the AIEOP-BFM-ALL-FLOW-MRD-Study Group. Cytometry B Clin Cytom. 2010;78В(3):147–53. doi: 10.1002/cyto.b.20516.
  34. Borowitz MJ, Pullen DJ, Winick N, et al. Comparison of diagnostic and relapse flow cytometry phenotypes in childhood acute lymphoblastic leukemia: implications for residual disease detection: a report from the Children’s Oncology Group. Cytometry B Clin Cytom. 2005;68В(1):18–24. doi: 1002/cyto.b.20071.
  35. Liu YR, Chang Y, Fu JY, et al. Comparison of the immunophenotype of patients with B lineage acute lymphoblastic leukemia at diagnosis and relapse. Zhonghua Xue Ye Xue Za Zhi. 2006;27(5):335–8.
  36. Dworzak MN, Froschl G, Printz D, et al. Prognostic significance and modalities of flow cytometric minimal residual disease detection in childhood acute lymphoblastic leukemia. Blood. 2002;99(6):1952–8. doi: 10.1182/blood.v99.6.1952.
  37. Coustan-Smith E, Ribeiro RC, Stow P, et al. A simplified flow cytometric assay identifies children with acute lymphoblastic leukemia who have a superior clinical outcome. Blood. 2006;108(1):97–102. doi: 1182/blood-2006-01-0066.
  38. Попов А.М., Вержбицкая Т.Ю., Цаур Г.А. и др. Ограниченная возможность применения упрощенного подхода для определения минимальной остаточной болезни методом проточной цитометрии у детей с острым лимфобластным лейкозом из B-линейных предшественников. Клиническая лабораторная диагностика. 2011;3:25–9.
    [Popov AM, Verzhbitskaya TYu, Tsaur GA, et al. Limited potential for use of simplified approach for determining minimal residual disease by means of flow cytometry in children with acute lymphoblastic leukemia from B-linear precursors. Klinicheskaya laboratornaya diagnostika. 2011;3:25–9. (In Russ)]
  39. Porwit-MacDonald A, Bjorklund E, Lucio P, et al. BIOMED-1 concerted action report: flow cytometric characterization of CD7+ cell subsets in normal bone marrow as a basis for the diagnosis and follow-up of T cell acute lymphoblastic leukemia (T-ALL). Leukemia. 2000;14(5):816–25. doi: 1038/sj.leu.2401741.
  40. Dworzak MN, Fritsch G, Buchinger P, et al. Flow cytometric assessment of human MIC2 expression in bone marrow, thymus, and peripheral blood. Blood. 1994;83(2):415–25.
  41. Dworzak MN, Fritsch G, Fleischer C, et al. CD99 (MIC2) expression in paediatric B-lineage leukaemia/lymphoma reflects maturation-associated patterns of normal B-lymphopoiesis. Br J Haematol. 1999;105(3):690–5. doi:1046/j.1365-2141.1999.01426.x.
  42. Dworzak MN, Froschl G, Printz D, et al. CD99 expression in T-lineage ALL: implications for flow cytometric detection of minimal residual disease. Leukemia. 2004;18(4):703–8. doi:1038/sj.leu.2403303.
  43. Roshal M, Fromm JR, Winter S, et al. Immaturity associated antigens are lost during induction for T cell lymphoblastic leukemia: implications for minimal residual disease detection. Cytometry B Clin Cytom. 2010;78В(3):139–46. doi: 10.1002/cyto.b.20511.
  44. Lund-Johansen F, Terstappen LW. Differential surface expression of cell adhesion molecules during granulocyte maturation. J Leuk Biol. 1993;54(1):47–55.
  45. Terstappen LW, Huang S, Picker LJ. Flow cytometric assessment of human T-cell differentiation in thymus and bone marrow. B 1992;79(3):666–77.
  46. Aalbers AM, van den Heuvel-Eibrink MM, Baumann I, et al. Bone marrow immunophenotyping by flow cytometry in refractory cytopenia of childhood. Haematologica. 2015;100(3):315–23. doi: 10.3324/haematol.2014.107706.
  47. Feng B, Verstovsek S, Jorgensen JL, Lin P. Aberrant myeloid maturation identified by flow cytometry in primary myelofibrosis. Am J Clin Pathol. 2010;133(2):314–20. doi: 10.1309/AJCPNC99DHXIOOTD.
  48. Loken MR, Chu S-Ch, Fritschle W, et al. Normalization of bone marrow aspirates for hemodilution in flow cytometric analyses. Cytometry B Clin Cytom. 2009;76В(1):27–36. doi: 10.1002/cyto.b.20429.
  49. Kussick SJ, Wood BL. Using 4-color flow cytometry to identify abnormal myeloid populations. Arch Pathol Lab Med. 2003;127(9):1140–7.
  50. Leandro MJ, Cooper N, Cambridge G, et al. Bone marrow B-lineage cells in patients with rheumatoid arthritis following rituximab therapy. Rheumatology (Oxford). 2007;46(1):29–36. doi: 10.1093/rheumatology/kel148.
  51. Rehnberg M, Amu S, Tarkowski A, et al. Short- and long-term effects of anti-CD20 treatment on B cell ontogeny in bone marrow of patients with rheumatoid arthritis. Arthritis Res Ther. 2009;11(4):R123. doi: 10.1186/ar2789.
  52. Nakou M, Katsikas G, Sidiropoulos P, et al. Rituximab therapy reduces activated B cells in both the peripheral blood and bone marrow of patients with rheumatoid arthritis: depletion of memory B cells correlates with clinical response. Arthritis Res Ther. 2009;11(4):R131. doi: 10.1186/ar2798.
  53. Borowitz MJ. Minimal residual disease detection in childhood ALL. Haematopoiesis Immunology. 2010;7(1):24–35.
  54. Вержбицкая Т.Ю., Попов А.М., Томилов А.Ф. и др. Определение опухолевых клеток в спинномозговой жидкости у детей с острыми лейкозами методом проточной цитометрии. Вопросы диагностики в педиатрии. 2012;5:31–5.
    [Verzhbitskaya TYu, Popov AM, Tomilov AF, et al. Detection of tumor cells in cerebrospinal fluid in children with acute leukemias using flow cytometry. Voprosy diagnostiki v pediatrii. 2012;5:31–5. (In Russ)]


Complex Chromosomal Aberrations in Patients with Post-Transplantation Relapses of Acute Leukemias: Clinical and Theoretical Aspects

TL Gindina, NN Mamaev, SN Bondarenko, NV Semenova, EN Nikolaeva, ME Vlasova, NV Stancheva, OA Slesarchuk, VN Vavilov, EV Morozova, AL Alyanskii, BV Afanasev

R.M. Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician I.P. Pavlov First St. Petersburg State Medical University, 12 Rentgena str., Saint Petersburg, Russian Federation, 197022

For correspondence: Tat’yana Leonidovna Gindina, PhD, 12 Rentgena str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)233-12-43; e-mail: tatgindina@gmail.com

For citation: 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 (In Russ).


ABSTRACT

Objective. To analyze the incidence of a complex karyotype in patients with post-transplantation relapses of acute myeloid leukemias and to evaluate preliminary treatment results before and after bone marrow transplantation in order to elaborate optimal approaches to the treatment of this disease.

Methods. Cytogenetic investigations (including fluorescent in situ hybridization [FISH]) were performed in 100 patients (53 males, 47 females aged from 1 to 60; median — 23 years) with post-transplantation relapses of acute myeloid leukemias (AML) (n = 61) and acute lymphoblastic leukemia (ALL) (n = 39).

Results. Aberrant karyotypes were found in 90 % of AML and 97 % of ALL patients. The incidence of acute leukemias (AL) with complex karyotypes (CK) was significantly higher in ALL patients than that in the AML group (67 % vs 36 %; = 0.002). At that, the percentage of CK with 4 and more chromosome abnormalities per cell in ALL patients aged 1–18 years was also significantly higher than that in AML patients (60 % vs 30 %; = 0.03). Besides, this difference was observed in the CK+ proportion between ALL and AML groups. Transplantation was performed during the active phase of the disease (i.e. after remission) in 75 % vs 55 %, respectively (= 0.003).

Conclusions. Serial cytogenetic investigations showed that CKs before transplantation and in PTR are closely related, thus confirming their clonal nature. Therefore, it may be assumed that karyotype complication achieved by the PTR can be caused by both chemotherapy performed at early stages of acute leukemia and pre-transplant conditioning regimes. In this case, further increase of the chemotherapeutic intensity in order to prevent and treat expected PTRs in patients with CK+ acute leukemias seems to be unreasonable. In this connection, infusion of donor lymphocytes, administration of hypomethylating agents or medicines with target mechanism of action should be used for management of AML patients during the post-transplant period.


Keywords: acute leukemias, post-transplantation relapses, complex karyotype.

Received: September 2, 2014

Accepted: November 13, 2014

Read in PDF (RUS)pdficon


REFERENCES

  1. Dobbelstein C, Dammann E, Weissinger E, et al. Prognostic impact of a newly defined structurally complex karyotype in patients with AML and MDS after allogeneic stem cell transplantation. Blood (ASH Annual Meeting Abstracts). 2013;122(21):3362–3.
  2. Mohr B, Stolzel F, Kramer M, et al. Karyotypic complexity in acute myeloid leukemia in the context of adverse prognosis. Blood (ASH Annual Meeting Abstracts). 2013;122(21):489.
  3. 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.
  4. Mrozek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;358(4):365–77. doi: 10.1053/j.seminoncol.2008.04.007.
  5. Gohring G, Michalova K, Beverloo HB, et al. Complex karyotype newly defined: the strongest prognostic factor in advanced childhood myelodysplastic syndrome. Blood. 2010;116(19):3766–9. doi: 10.1182/blood-2010-04-280313.
  6. Schoch C, Haferlach T, Haase D, et al. Patients with de novo acute myeloid leukemia and complex karyotype aberrations show a pore 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.
  7. 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, Barkhatov 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)]
  8. Schmid C, Schleuning M, Tischer J, et al. Early allo-SCT for AML with a complex aberrant karyotype – results from a prospective pilot study. Bone Marrow Transplant. 2012;47(1):46–53. doi: 10.1038/bmt.2011.15.
  9. Zaccaria A, Rosti G, Testoni N, et al. Chromosome studies in patients with nonlymphoсytic or acute lymphocytic leukemia submitted to bone marrow transplantation – results of European cooperative study. Cancer Genet Cytogenet. 1987;26(1):51–8.
  10. Schmidt-Hieber M, Blau IW, Richter G, et al. Cytogenetic studies in acute leukemia patients relapsing after allogeneic stem cell transplantation. Cancer Genet Cytogenet. 2010;198(2):135–43. doi: 10.1016/j.cancergencyto.2010.01.005.
  11. Chi HS, Cho YU, Park SH, et al. Comparative analysis of cytogenetic evolution patterns during relapse in the hematopoietic stem cell transplantation and chemotherapy settings of patients with acute leukemia. Blood (ASH Annual Meeting Abstracts). 2013;122(21):1320.
  12. Yuasa M, Uchida M, Kaji D, et al. Prognostic significance of the cytogenetic evolution after the hematopoietic stem cell transplantation in adult acute myeloid leukemia. Blood (ASH Annual Meeting Abstracts). 2013;122(21):1391–2.
  13. Гиндина Т.Л., Мамаев Н.Н., Кондакова Е.В. и др. Острые лимфобластные лейкозы с высокогиперплоидными кариотипами. Вестник гематологии. 2007;4:18–23.
    [Gindina TL, Mamaev NN, Kondakova EV, et al. Acute lymphoblastic leukemias with highly hyperploid karyotypes. Vestnik gematologii. 2007;4:18–23. (In Russ)]
  14. Schaffer LG, McGowan-Jordan J, Schmid M. ISCN. An International System for Human Cytogenetic Nomenclature. Basel: Karger; 2013.
  15. Schmid C, Labopin M, Nagler A, et al. Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT acute leukemia working party. J Clin Oncol. 2007;25(31):4938–45. doi: 10.1200/jco.2007.11.6053.
  16. Schroeder T, Czibere A, Platzbecker U, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013;27(6):1229–35. doi: 10.1038/leu.2013.7.
  17. Porter DL, Alyea EP, Antin JH, et al. NCI First International Workshop on the biology, prevention and treatment of relapse after allogeneic hematopoietic stem cell transplantation: Report from the Committee on treatment of relapse after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2010;16(11):1467–503. doi: 10.1016/j.bbmt.2010.08.001.
  18. Alyea EP, DeAngelo DJ, Moldrem J, et al. NCI First International Workshop on the Biology, Prevention and Treatment of Relapse after Allogeneic Hematopoietic Cell Transplantation: Report from the Committee on Prevention of Relapse Following Allogeneic Cell Transplantation for Hematologic Malignancies. Biol Blood Marrow Transplant. 2010;16(8):1037–69. doi: 10.1016/j.bbmt.2010.05.005.
  19. de Lima M, Giralt S, Thall PF. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome. Cancer. 2010;116(23):5420–31. doi: 10.1002/cncr.25500.
  20. 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 Transplant. 2014;20(1):4–13. doi: 10.1016/j.bbmt.2013.08.012.
  21. Duque-Afonso J, Lubbert M, Cleary ML. Epigenetic modifications mediated by the AML1/ETO and MLL leukemia fusion proteins. In: Lubbert M, Jones PA, eds. Epigenetic Therapy of Cancer. Berlin Heidelberg: Springer-Verlag; 2014. pp. 121–44. doi: 10.1007/978-3-642-38404-2_6.
  22. Buron F, Malvezzi P, Villar E. Profiling sirolimus-induced inflammatory syndrome a prospective tricentric observational study. PloS One. 2013;8(1):e53078. doi: 10.1371/journal.pone.0053078.
  23. Kondo T, Tasaka T, Matsumoto K, et al. Philadelphia chromosome-positive acute lymphoblastic leukemia with extramedullary and meningeal relapse after allogeneic hematopoietic stem cell transplantation that was successfully treated with dasatinib. Springerplus. 2014;3:177. doi: 10.1186/2193-1801-3-177.
  24. Maziarz RT, Slater S. Post-transplant relapse. In: Maziarz RT, Slater S, eds. Blood and Marrow Transplant Handbook. Springer Science+Business Media, LLC; 2011. pp. 271–6. doi: 10.1007/978-1-4419-7506-5_24.

Apoptotic markers in CD34-positive cells in acute leukemias

Ye.N. Parovichnikova1, Ye.Ye. Khodunova1, I.V. Galtseva1, S.М. Kulikov1, V.V. Troitskaya1, L.А. Kuzmina1, D.V. Shcheblyakov2, and V.G. Savchenko1

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

2 N.F. Gamaleya Research Institution of Epidemiology and Microbiology, RF Ministry of Health, Moscow, Russian Federation


ABSTRACT

Objective. To evaluate expression of Bcl-2, Bax, p53, CD95, and ACE on CD34+ cells of peripheral blood and bone marrow during induction chemotherapy in the patients with newly diagnosed acute leukemia.

Materials and methods. Expression of Bcl-2, Bax, p53, CD95, and ACE on CD34+ cells of the peripheral blood and bone marrow in 23 patients with AL (14 AML and 9 ALL) was measured using flow cytometry analysis. Peripheral blood and bone marrow samples were analyzed before chemotherapy and during the induction course: on Days +8, +21 (blood only), and +36–38. The control group consisted of 8 healthy donors.

Results. Bcl-2 expression on CD34+ sells in BM was 34.8 ± 6 % and significantly higher compared to the donors (11.5 ± 1.8 %) at the time of diagnosis. On Days +36–38 after the onset of chemotherapy, no significant difference between the patients and control groups were found. CD34/Bax coexpression in BM cells of ALL patients was significantly higher than in AML patients and donors. ACE and p53 expression on CD34+ cells in AL patients before and during chemotherapy was significantly lower than in the donors. CD34/ACE coexpression in PB and BM cells of AL patients and donors showed no significant differences at any time-points of evaluation.

Conclusion. The above changes suggest the imbalance between the pro- and anti-apoptotic proteins in AL patients. After chemotherapy, the expression profile of these proteins considerably changed, but did not reach the healthy donor values.


Keywords: acute leukemias, apoptosis, expression of Bcl-2, Bax, р53, CD95, and ACE.

Read in  PDF (RUS)pdficon


Refernces

  1. Hengartner M.O. The biochemistry of apoptosis. Nature 2000; 407(6805): 770–6.
  2. Lodish H., Berk A., Matsudaira P. et al. Molecular cell biology, 5th edn. New York: W.H. Freeman and Company, 2003: 961.
  3. Salminen A., Ojala J., Kaarniranta K. Apoptosis and aging: increased resistance to apoptosis enhances the aging process. Cell. Mol. Life Sci. 2011; 68(6): 1021–31.
  4. Spencer S.L., Sorger P.K. Measuring and modeling apoptosis in single cells. Cell 2011; 144(6): 926–39.
  5. Herr I., Debatin K.-M. Cellular stress response and apoptosis in cancer therapy. Blood 2001; 98(9): 2603–14.
  6. Chao D.T., Korsmeyer S. J. BCL-2 family: regulators of cell death. Ann. Rev. Immunol. 1998; 16: 395–419.
  7. Green D.R. Apoptotic pathways: The roads to ruin. Cell 1998; 94: 695–8.
  8. Chauncey T.R. Drug resistance mechanisms in acute leukemia. Curr. Opin. Oncol. 2001; 13(1): 21–6.
  9. Del Poeta G., Bruno A., Del M.I. Principe et al. Deregulation of the mitochondrial apoptotic machinery and development of molecular targeted drugs in acute myeloid leukemia. Curr. Cancer Drug Targets 2008; 8(3): 202–7.
  10. Testa U., Riccioni R. Deregulation of apoptosis in acute myeloid leukemia. Haematologica 2007; 92(1): 81–94.
  11. Banker D.E., Groudine M., Norwood T., Appelbaum F.R. Measurement of spontaneous and therapeutic agent-induced apoptosis with BCL-2 protein expression in acute myeloid leukemia. Blood 1997; 89(1): 243–55.
  12. Stijn A., Pol M.A., Kok A. et al. Differences between the CD34+ and CD34– blast compartments in apoptosis resistance in acute myeloid leukemia. Haematologica 2003; 88(5): 497–508.
  13. Fulda S., Los M., Friesen C., Debatin K.M. Chemosensitivity of solid tumor cells in vitro is related to activation of the CD95 system. Int. J. Cancer 1998; 76(1): 105–14.
  14. Fulda S., Sieverts H., Friesen C. et al. The CD95 (APO-1/Fas) system mediates drug-induced apoptosis in neuroblastoma cells. Cancer Res. 1997; 57(17): 3823–9.
  15. Барышников А.Ю., Полосухина Е.Р., Шишкин Ю.В. и др. Новый прогностический маркер острого лимфобластного лейкоза — антиген CD95 (FAS/APO-1). Гематол. и трансфузиол. 1998; 2: 8–11. [Baryshnikov A.Yu., Polosukhina Ye.R., Shishkin Yu.V. i dr. Novyy prognosticheskiy marker ostrogo limfoblastnogo leykoza — antigen CD95 (FAS/APO-1) (New prognostic marker of acute lymphoblastic leukemia — CD95 antigen (FAS/ APO-1). In: Hematol. & transfusiol.) Gematol. i transfuziol. 1998; 2: 8–11.]
  16. Molica S., Mannella A., Dattilo A. et al. Differential expression of BCL-2 oncoprotein and Fas antigen on normal peripheral blood and leukemic bone marrow cells. A flow cytometric analysis. Haematologica 1996; 81(4): 302–9.
  17. Greenblatt M.S., Bennett W.P., Hollstein M., Harris C.C. Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 1994; 54(18): 4855–78.
  18. Ko L.J., Prives C. p53: puzzle and paradigm. Genes & Dev. 1996; 10(9): 1054–72.
  19. Levine A.J. p53, the cellular gatekeeper for growth and division. Cell 1997; 88(3): 323–31.
  20. Resnick M.A., Tomso D., Inga A. et al. Functional diversity in the gene network controlled by the master regulator p53 in humans. Cell. Cycle 2005; 4(8): 1026–9.
  21. Prokocimer M., Rotter V. Structure and function of p53 in normal cells and their aberrations in cancer cells: projection on the hematologic cell lineages. Blood 1994; 84(8): 2391–411.
  22. Wada M., Bartram C.R., Nakamura H. et al. Analysis of p53 mutations in a large series of lymphoid hematologic malignancies of childhood. Blood 1993; 82(10): 3163–9.
  23. Cavalcanti G.B., Scheiner M.A., Simoes Magluta E.P. et al. p53 flow cytometry evaluation in leukemias: correlation to factors affecting clinical outcome. Cytometry B. Clin. Cytom. 2010; 78(4): 253–9.
  24. Beyazit Y., Aksu S., Haznedaroglu I.C. et al. Overexpression of the local bone marrow renin-angiotensin system in acute myeloid leukemia. J. Natl. Med. Assoc. 2007; 99: 57–63.
  25. Goker H., Haznedaroglu I.C., Beyazit Y. et al. Local umbilical cord blood renin-angiotensin system. Ann. Hematol. 2005; 84: 277–81.
  26. Jokubaitis V.J., Sinka L., Driessen R. et al. Angiotensin-converting enzyme (CD143) marks hematopoietic stem cells in human embryonic, fetal, and adult hematopoietic tissues. Blood 2008; 111: 4055–63.
  27. Li J., Volkov L., Comte L. et al. Production and consumption of the tetrapeptide AcSDKP, a negative regulator of hematopoietic stem cells, by hematopoietic microenvironmental cells. Exp. Hematol. 1997; 25: 140–6.
  28. Гальцева И.В., Пашин Л.Е., Савченко В.Г. Лейкемические дендритные клетки. Тер. арх. 2008; 80(7): 84–8. [Galtseva I.V., Pashin L.Ye., Savchenko V.G. Leykemicheskiye dendritnyye kletki (Leukemic dendritic cells. In: Ther. archive). Ter. arkh. 2008; 80(7): 84–8.]
  29. Danilov S.M., Sadovnikova E., Scharenborg N. et al. Angiotensinconverting enzyme (CD 143) is abundantly expressed by dendritic cells and discriminates human monocyte-derived dendritic cells from acute myeloid leukemia-derived dendritic cells. Exp. Hematol. 2003; 31(12): 1301–9.
  30. Del Poeta G., Venditti A., Del Principe M.I. et al. Amount of spontaneous apoptosis detected by Bax/Bcl-2 ratio predicts outcome in acute myeloid leukemia (AML). Blood 2003; 101(6): 2125–31.
  31. El-Manallawy H.A., El-Shkankiry N.H., El-Guindy S. et al. The Expression of Bcl-2 and Bax Proteins and Their Clinical Relevance in ALL and CLL Patients. J. Egypt. Nat. Cancer Inst. 2001; 13(1): 35–42.
  32. Hogarth L.A., Hall A.G. Increased BAX expression is associated with an increased risk of relapse in childhood acute lymphocytic leukemia. Blood 1999; 93(8): 2671–8.
  33. Aksu S., Beyazit Y., Haznedaroglu I.C. et al. Enhanced expression of the local haematopoietic bone marrow renin-angiotensin system in polycythemia rubra vera. J. Int. Med. Res. 2005; 33(6): 661–7.
  34. Aksu S., Beyazit Y., Haznedaroqlu I.C. et al. Over-expression of angiotensin-converting enzyme (CD 143) on leukemic blasts as a clue for the activated local bone marrow RAS in AML. Leuk. Lymphoma 2006; 47(5): 891–6.
  35. Барышников А.Ю., Шишкин Ю.В. Иммунологические проблемы апоптоза. М., 2002.  [Baryshnikov A.Yu., Shishkin Yu.V. Immunologicheskiye problemy apoptoza (Immunological problems of apoptosis). , 2002.]
  36. Miyawaki T., Uehara T., Nibu R. et al. Differential expression of apoptosisrelated Fas antigen on lymphocyte subpopulations in human peripheral blood. J. Immunol. 1992; 149(11): 3753–8.
  37. Lechner H., Amort M., Steger M.M. et al. Regulation of CD95 (APO-1) expression and the induction of apoptosis in human T cells: changes in old age. Int. Arch. Allergy Immunol. 1996; 110(3): 238–43.
  38. Kotani T., Aratake Y., Kondo S. et al. Expression of functional Fas antigen on adult T-cell leukemia. Leuk. Res. 1994; 18(4): 305–10.