Myelofibrosis Models: Literature Review and Own Data

AA Silyutina, II Gin, NM Matyukhina, EN Balayan, PA Butylin

VA Almazov Federal North-West Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

For correspondence: Pavel Andreevich Butylin, PhD, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; e-mail: butylinp@gmail.com

For citation: Silyutina AA, Gin II, Matyukhina NM, et al. Myelofibrosis Models: Literature Review and Own Data. Clinical oncohematology. 2017;10(1):75–84 (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-75-84


ABSTRACT

Background & Aims. Chronic myeloproliferative disorders typically develop during a long latent period, and it complicates the study of the mechanism of its pathogenesis. Observations from the clinical practice should be confirmed by experiments. The mechanisms of oncological transformation related to mutations associated with chronic myeloproliferative diseases were confirmed in transgene animal models. Biological models permitted to determine a complex nature of myelofibrosis. However, studies of the cellular mechanisms of myelofibrosis require new models. This paper presents a review of published models of myeloproliferative disorders, mainly, primary myelofibrosis, and results of studies of a new cell line with expression of JAK2 V617F. The aim of this study is to create a new cell line with expression of transforming JAK2 V617F mutation in acute monocytic leukemia THP-1 cells.

Methods. Transgenic cell lines were created on the basis of monocytic leukemia THP-1 cell line that can differentiate into macrophages. Direct mutagenesis was used to cause V617F mutation. Two cell lines were created: one with JAK2 expression with V617F mutation, the other with wild type JAK2.

Results. Both transgenic lines were characterized by increased JAK2 expression as compared to non-modified cells. In routine cultivation, transgenic THP-1 cells retained the morphology of monocytes. After treatment with phorbol ester, THP-1 differentiated into macrophages and become adherent to culture plastic. Adherent cells demonstrated the variety of shapes: some of them were spherical, the other ones had pseudopodia. No significant differences in viability of cells were observed. However, macrophages expressing mutant JAK2 and overexpressing the wild type JAK2 demonstrated a tendency to decreased amount unlivable cells during cultivation.

Conclusion. The obtained cell model can be used for estimating the influence of JAK2 V617F mutation on pro- and antifibrotic potential of macrophages that can help to investigate the pathogenetic role of macrophages in myelofibrosis development. In addition, this model can help to develop novel methods of therapy and diagnostics of primary and secondary myelofibrosis.

Keywords: Ph-negative chronic myeloproliferative disorders, primary myelofibrosis, JAK2 V617F, transgenic animals.

Received: September 15, 2016

Accepted: December 13, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002;100(7):2292–302. doi: 10.1182/blood-2002-04-1199.
  2. Hoffman R, Rondelli D. Biology and treatment of primary myelofibrosis. Hematol Am Soc Hematol Educ Program. 2007;1:346–54. doi: 10.1182/asheducation-2007.1.346.
  3. Mesa RA, Niblack J, Wadleigh M, et al. The burden of fatigue and quality of life in myeloproliferative disorders (MPDs): an international internet-based survey of 1179 MPD patients. Cancer. 2007;109(1):68–76. doi: 10.1002/cncr.22365.
  4. 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.
  5. Scherber R, Dueck AC, Johansson P, et al. The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): international prospective validation and reliability trial in 402 patients. Blood. 2011;118(2):401–8. doi: 10.1182/blood-2011-01-328955.
  6. Barosi G, Mesa RA, Thiele J, et al. Proposed criteria for the diagnosis of post-polycythemia vera and post-essential thrombocythemia myelofibrosis: a consensus statement from the International Working Group for Myelofibrosis Research and Treatment. Leukemia. 2008;22(2):437–8. doi: 10.1038/sj.leu.2404914.
  7. Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med. 2000;342(17):1255–65. doi: 10.1056/nejm200004273421706.
  8. Jacobson RJ, Salo A, Fialkow PJ. Agnogenic myeloid metaplasia: a clonal proliferation of hematopoietic stem cells with secondary myelofibrosis. Blood. 1978;51:189–94.
  9. Reeder TL, Bailey RJ, Dewald GW, Tefferi A. Both B and T lymphocytes may be clonally involved in myelofibrosis with myeloid metaplasia. Blood. 2003;101(5):1981–3. doi: 10.1182/blood-2002-07-2341.
  10. Reilly JT. Idiopathic myelofibrosis: pathogenesis, natural history and management. Blood Rev. 1997;11(4):233–42. doi: 10.1016/s0268-960x(97)90022-9.
  11. Mehta J, Wang H, Iqbal SU, Mesa R. Epidemiology of myeloproliferative neoplasms (MPN) in the United States. Leuk Lymphoma. 2014;55(3):595–600. doi: 10.3109/10428194.2013.813500.
  12. Kaushansky K. Thrombopoietin: The primary regulator of platelet production. Blood. 1995;86(2):419–31.
  13. de Sauvage FJ, Carver-Moore K, Luoh SM, et al. Physiological regulation of early and late stages of megakaryocytopoiesis by thrombopoietin. J Exp Med. 1996;183(2):651–6.
  14. Vannucchi AM, Villeval J-L, Wagner-Ballon O, et al. Animal Models of Myelofibrosis. In: Conn PM, ed. Sourcebook of Models for Biomedical Research. Totowa: Humana Press Inc.; 2008. pр. 713–23. doi: 10.1007/s12223-008-0071-5.
  15. Ulich TR, del Castillo J, Senaldi G, et al. Systemic hematologic effects of PEG-rHuMGDF-induced megakaryocyte hyperplasia in mice. Blood. 1996;87(12):5006–15.
  16. Villeval JL, Cohen-Solal K, Tulliez M, et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood. 1997;90(11):4369–83.
  17. Yan XQ, Lacey D, Fletcher F, et al. Chronic exposure to retroviral vector encoded MGDF (MPL-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood. 1995;86(11):4025–33.
  18. Ohwada A, Rafii S, Moore MA, Crystal RG. In vivo adenovirus vector-mediated transfer of the human thrombopoietin cDNA maintains platelet levels during radiation-and chemotherapy-induced bone marrow suppression. Blood. 1996;88(3):778–84.
  19. Cannizzo SJ, Frey BM, Raffi S, et al. Augmentation of blood platelet levels by intratracheal administration of an adenovirus vector encoding human thrombopoietin cDNA. Nat Biotechnol. 1997;15(6):570–3. doi: 10.1038/nbt0697-570.
  20. Abina MA, Tulliez M, Duffour MT, et al. Thrombopoietin (TPO) knockout phenotype induced by cross-reactive antibodies against TPO following injection of mice with recombinant adenovirus encoding human TPO. J Immunol. 1998;160(9):4481–9.
  21. Frey BM, Rafii S, Teterson M, et al. Adenovector-mediated expression of human thrombopoietin cDNA in immune-compromised mice: Insights into the pathophysiology of osteomyelofibrosis. J Immunol. 1998;160(2):691–9.
  22. Zhou W, Toombs CF, Zou T, et al. Transgenic mice overexpressing human c-MPL ligand exhibit chronic thrombocytosis and display enhanced recovery from 5-fluorouracil or antiplatelet serum treatment. Blood. 1997;89(5):1551–9.
  23. Kakumitsu H, Kamezaki K, Shimoda K, et al. Transgenic mice overexpressing murine thrombopoietin develop myelofibrosis and osteosclerosis. Leuk Res. 2005;29(7):761–9. doi: 10.1016/j.leukres.2004.12.009.
  24. Yanagida M, Ide Y, Imai A, et al. The role of transforming growth factor-beta in PEG-rHuMGDF-induced reversible myelofibrosis in rats. Br J Haematol. 1997;99(4):739–45.
  25. Abina MA, Tulliez M, Lacout C, et al. Major effects of TPO delivered by a single injection of a recombinant adenovirus on prevention of septicemia and anemia associated with myelosuppression in mice: Risk of sustained expression inducing myelofibrosis due to immunosuppression. Gene Ther. 1998;5(4):497–506. doi: 10.1038/sj.gt.3300638.
  26. Shultz LD, SchweitzerPA, Christianson SW, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-SCID mice. J. Immunol. 1995;154:180–91.
  27. Serreze DV, Gaedeke JW, Leiter EH. Hematopoietic stem-cell defects underlying abnormal macrophage development and maturation in NOD/Lt mice: defective regulation of cytokine receptors and protein kinase C. Proc Natl Acad Sci USA. 1993;90(20):9625–9. doi: 10.1073/pnas.90.20.9625.
  28. Wagner-Ballon O, Hedia Chagraoui H, Eric Prina E, et al. Monocyte/Macrophage Dysfunctions Do Not Impair the Promotion of Myelofibrosis by High Levels of Thrombopoietin. J Immunol. 2006;176(11):6425–33. doi: 10.4049/jimmunol.176.11.6425.
  29. Tefferi A. Experimental myelofibrosis in mice and the implications to human disease. Leuk Res. 2005;29(7):723–6. doi: 10.1016/j.leukres.2004.12.006.
  30. Tsai SF, Martin DI, Zon LI, et al. Cloning of cDNA for the major DNA-binding protein of the erythroid lineage through expression in mammalian cells. Nature. 1989;339(6224):446–51. doi: 10.1038/339446a0.
  31. Romeo PH, Prandini MH, Joulin V, et al. Megakaryocytic and erythrocytic lineages share specific transcription factors. Nature. 1990;344(6265):447–9. doi: 10.1038/344447a0.
  32. Yu C, Cantor AB, Yang H, et al. Targeted deletion of a high-affinity GATA-binding site in the GATA-1 promoter leads to selective loss of the eosinophil lineage in vivo. J Exp Med. 2002;195(11):1387–95. doi: 10.1084/jem.20020656.
  33. Migliaccio AR, Rana RA, Sanchez M, et al. GATA-1 as a regulator of mast cell differentiation revealed by the phenotype of the GATA-1low mouse mutant. J Exp Med. 2003;197(3):281–96. doi: 10.1084/jem.20021149.
  34. McDevitt MA, Shivdasani RA, Fujiwara Y, et al. A “knockdown” mutation created by cis-element gene targeting reveals the dependence of erythroid cell maturation on the level of transcription factor GATA-1. Proc Natl Acad Sci USA. 1997;94(13): 6781–5. doi: 10.1073/pnas.94.13.6781.
  35. Shivdasani RA, Fujiwara Y, McDevitt MA, et al. A line-age-selective knockout establishes the critical role of transcription factor GATA-1 in megakaryocyte growth and platelet development. EMBO J. 1997;16(13):3965–73. doi: 10.1093/emboj/16.13.3965.
  36. Vyas P, Ault K, Jackson CW, et al. Consequences of GATA-1 deficiency in megakaryocytes and platelets. Blood. 1999;93(9):2867–75.
  37. Centurione L, Di Baldassarre A, Zingariello M, et al. Increased and pathologic emperipolesis of neutrophils within megakaryocytes associated with marrow fibrosis in GATA-1(low) mice. Blood. 2004;104(12):3573–80. doi: 10.1182/blood-2004-01-0193.
  38. Vannucchi AM, Bianchi L, Cellai C, et al. Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). Blood. 2002;100(4):1123–32. doi: 10.1182/blood-2002-06-1913.
  39. Vannucchi AM, Migliaccio AR, Paoletti F, et al. Pathogenesis of myelofibrosis with myeloid metaplasia: Lessons from mouse models of the disease. Semin Oncol. 2005;32(4):365–72. doi: 10.1053/j.seminoncol.2005.04.008.
  40. Radich J. The Molecular Biology of Myeloproliferative Disorders. Cancer Cell. 2010;18(1):7–8. doi: 10.1016/j.ccr.2010.06.006.
  41. Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365(9464):1054–61. doi: 10.1016/s0140-6736(05)74230-6.
  42. Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779–90. doi: 10.1056/nejmoa051113.
  43. Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97. doi: 10.1016/j.ccr.2005.03.023.
  44. Kiladjian JJ. The spectrum of JAK2-positive myeloproliferative neoplasms. Hematol Am Soc Hematol Educ Program. 2012;2012:561–6. doi: 10.1182/asheducation-2012.1.561.
  45. Levine RL, Gilliland DG. Myeloproliferative disorders. Blood. 2008;112(6):2190–8. doi: 10.1182/blood-2008-03-077966.
  46. Chen M, Cheng A, Chen YQ, et al. The amino terminus of JAK3 is necessary and sufficient for binding to the common gamma chain and confers the ability to transmit interleukin 2-mediated signals. Proc Natl Acad Sci USA. 1997;94(13):6910–5. doi: 10.1073/pnas.94.13.6910.
  47. Saharinen P, Silvennoinen O. The pseudokinase domain is required for suppression of basal activity of Jak2 and Jak3 tyrosine kinases and for cytokine-inducible activation of signal transduction. J Biol Chem. 2002;277(49):47954–63. doi: 10.1074/jbc.m205156200.
  48. Griffith J, Black J, Faerman C, et al. The structural basis for autoinhibition of FLT3 by the juxtamembrane domain. Mol Cell. 2004;13(2):169–78. doi: 10.1016/s1097-2765(03)00505-7.
  49. Lindauer K, Loerting T, Liedl KR, et al. Prediction of the structure of human Janus kinase 2 (JAK2) comprising the two carboxy-terminal domains reveals a mechanism for autoregulation. Protein Engin. Design Select. 2001;14(1):27–37. doi: 10.1093/protein/14.1.27.
  50. Parganas E, Wang D, Stravopodis D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93(3):385–95. doi: 10.1016/s0092-8674(00)81167-8.
  51. Levine RL, Loriaux M, Huntly BJ, et al. The JAK2V617F activating mutation occurs in chronic myelomonocytic leukemia and acute myeloid leukemia, but not in acute lymphoblastic leukemia or chronic lymphocytic leukemia. Blood. 2005;106(10):3377–9. doi: 10.1182/blood-2005-05-1898.
  52. Jelinek J, Oki Y, Gharibyan V, et al. JAK2 mutation 1849G>T is rare in acute leukemias but can be found in CMML, Philadelphia-chromosome negative CML and megakaryocytic leukemia. Blood. 2005;106(10):3370–3. doi: 10.1182/blood-2005-05-1800.
  53. Steensma DP, Dewald GW, Lasho TL, et al. The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both “atypical” myeloproliferative disorders and the myelodysplastic syndrome. Blood. 2005;106(4):1207–9. doi: 10.1182/blood-2005-03-1183.
  54. James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signaling causes polycythaemia vera. Nature. 2005;434(7037):1144–8. doi: 10.1038/nature03546.
  55. Scott LM, Scott MA, Campbell PJ, Green AR. Progenitors homozygous for the V617F mutation occur in most patients with polycythemia vera, but not essential thrombocythemia. Blood. 2006;108(7):2435–7. doi: 10.1182/blood-2006-04-018259.
  56. Wernig G, Mercher T, Okabe R, et al. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107(11):4274–81. doi: 10.1182/blood-2005-12-4824.
  57. Lacout C, Pisani DF, Tulliez M, et al. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108(5):1652–60. doi: 10.1182/blood-2006-02-002030.
  58. Tiedt R, Hao-Shen H, Sobas MA, et al. Ratio of mutant JAK2-V617F to wild type JAK2 determines the MPD phenotypes in transgenic mice. Blood. 2007;111(8):3931–40. doi: 10.1182/blood-2007-08-107748.
  59. Xing S, Ho WT, Zhao W, et al. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood. 2008;111(10):5109–17. doi: 10.1182/blood-2007-05-091579.
  60. Marty C, Lacout C, Martin A, et al. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Blood. 2010;116(5):783–7. doi: 10.1182/blood-2009-12-257063.
  61. Li J, Kent DG, Chen E, Green AR. Mouse models of myeloproliferative neoplasms: JAK of all grades. Dis Model Mech. 2011;4(3):311–7. doi: 10.1242/dmm.006817.
  62. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270. doi: 10.1371/journal.pmed.0030270.
  63. Pardanani AD, Levine RL, Lasho T, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: A study of 1182 patients. Blood. 2006;108(10):3472–6. doi: 10.1182/blood-2006-04-018879.
  64. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405. doi: 10.1056/NEJMoa1312542.
  65. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90. doi: 10.1056/NEJMoa1311347.
  66. Wang WA, Groenendyk J, Michalak M. Calreticulin signaling in health and disease. Int J Biochem Cell Biol. 2012;44(6):842–6. doi: 10.1016/j.biocel.2012.02.009.
  67. Michalak M, Groenendyk J, Szabo E, et al. Calreticulin, a multi-process calcium-buffering chaperone of the endoplasmic reticulum. Biochem J. 2009;417(3):651–66. doi: 10.1042/BJ20081847.
  68. Gold LI, Eggleton P, Sweetwyne MT, et al. Calreticulin: non-endoplasmic reticulum functions in physiology and disease. FASEB J. 2010;24(3):665–83. doi: 10.1096/fj.09-145482.
  69. Luo B, Lee AS. The critical roles of endoplasmic reticulum chaperones and unfolded protein response in tumorigenesis and anticancer therapies. Oncogene. 2012;32(7):805–18. doi: 10.1038/onc.2012.130.
  70. Rumi E, Pietra D, Ferretti V, et al. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood. 2014;123(10):1544–51. doi: 10.1182/blood-2013-11-539098.
  71. Cabagnols X, Defour JP, Ugo V, et al. Differential association of calreticulin type 1 and type 2 mutations with myelofibrosis and essential thrombocytemia: relevance for disease evolution. Leukemia. 2014;29(1):249–52. doi: 10.1038/leu.2014.270.
  72. Marty C, Pecquet C, Nivarthi H, et al. Calreticulin mutants in mice induce an MPL-dependent thrombocytosis with frequent progression to myelofibrosis. Blood. 2016;127(10):1317–24. doi: 10.1182/blood-2015-11-679571.
  73. Chachoua I, Pecquet C, El-Khoury M, et al. Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants. Blood. 2016;127(10):1325–35. doi: 10.1182/blood-2015-11-681932.
  74. Thiele J, Kvasnicka HM, Boeltken B. Resident bone marrow macrophages in idiopathic (primary) myelofibrosis (IMF): a histochemical and morphometric study on sequential trephine biopsies. Leuk Res. 1999;23(11):983–5. doi: 10.1016/s0145-2126(99)00120-4.
  75. Tsuchiya S, Kobayashi Y, Goto Y, et al. Induction of maturation in cultured human monocytic leukemia cells by a phorbol diester. Cancer Res. 1982;42(4):1530–6.

 

 

Clinical and Hematological Characteristics of Patients with Chronic Myeloid Leukemia under Present-Day Conditions: Results of the Russian Part of International Multi-Center Prospective EUTOS Population-Based CML Study

OV Lazareva1, AG Turkina1, EYu Chelysheva1, IA Tishchenko1, MA Galaiko2, OM Senderova3, VM Pepelyaeva4, SV Meresii5, AS Luchinin6, GI Milyutina7, LV Gavrilova8, LB Avdeeva9, OYu Vinogradova10, SM Kulikov1

1 Hematology Research Center under the Ministry of Health of the Russian Federation, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

2 NA Semahko Central Clinical Hospital No. 2 under the JSC RZD, 43 Losinoostrovskaya str., Moscow, Russian Federation, 107150

3 Irkutsk Order of the Badge of Honor District Clinical Hospital, 100 Yubileinyi microdistrict, Irkutsk, Russian Federation, 664049

4 Perm Order of the Badge of Honor District Clinical Hospital, 85 Pushkina str., Perm, Russian Federation, 614990

5 Clinical Medical Unit No. 1, 68 Gagarina b-r, Perm, Russian Federation, 614077

6 Kirov District Clinical Hospital, 42 Vorovskogo str., Kirov, Russian Federation, 610027

7 District Clinical Hospital No. 1, 86 Stanke Dmitrova pr-t, Bryansk, Russian Federation, 241033

8 Republican Clinical Hospital No. 4 under the Ministry of Health of Russia, 32 Yl’yanova str., Saransk, Russian Federation, 430032

9 District Clinical Hospital, 7 Kokhanskogo str., Chita, Russian Federation, 672038

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

For correspondence: Ol’ga Veniaminovna Lazareva, PhD, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel: +7(495)612-48-60; e-mail: stakhino@gmail.com

For citation: Lazareva OV, Turkina AG, Chelysheva EYu, et al. Clinical and Hematological Characteristics of Patients with Chronic Myeloid Leukemia under Present-Day Conditions: Results of the Russian Part of International Multi-Center Prospective EUTOS Population-Based CML Study. Clinical oncohematology. 2017;10(1):65–74(In Russ).

DOI: 10.21320/2500-2139-2017-10-1-65-74


ABSTRACT

Background. Much attention has been paid to molecule-genetic diagnostics of chronic myeloid leukemia (CML) and its treatment using new highly effective methods of therapy. The baseline characteristics of patients at primary CML diagnosis are hardly discussed in literature.

Aim. To provide clinical, hematological, molecular genetic and demographic characteristics of patients obtained at primary diagnosis of CML.

Patients & Methods. Characteristics of CML patients are based on data gathered by the Russian Investigational Group for CML within the international project European Treatment and Outcome Study of CML in Europe (EUTOS, the European Treatment and Outcomes Study). The study included 197 patients with newly diagnosed CML in 6 regions of the Russian Federation (Mordovia, Kirov, Perm (2 sites), Bryansk, Irkutsk, and Chita) over the period from 2009 till 2012.

Results. The study demonstrated that 94 % of CML cases were diagnosed in the chronic phase (CP) and 6 % of cases in the acceleration phase (AP) and the blast crisis phase (BC). In 40 % of patients there were no clinical symptoms, and CML was suspected only due to changes in the CBC test. Fatigue was the main subjective complaint presented by 77 % of patients in the CP and 100 % of patients with the AP and BC. Peripheral blood leukocytosis, left shift to immature myeloid cells and increased granulocytic lineage in bone marrow were typical for the patients. In all patients, the CML diagnosis was confirmed by cytogenetic or molecular tests. The social and demographic characteristics of CML patients and comorbidities at diagnosis were analyzed.

Conclusion. Based on the results of the study, a modern «portrait of a CML patient» was obtained. The study demonstrated that cytogenetic and molecular methods allow to diagnose CML in most patients at early stages of the disease in the absence of clinical signs of progression. The data on comorbidities require a special attention while choosing a therapy considering its duration. Demographic and social characteristics of CML patients demonstrate that they are socially active, particularly interested in retaining the working capacity and quality of life.

Keywords: chronic myeloid leukemia, clinical and hematological characteristics, population-based study.

Received: August 2, 2016

Accepted: December 7, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Hehlmann R, Hochhaus A, Baccarani M, on behalf of the European Leukemia Net. Chronic myeloid leukemia. Lancet. 2007;370(9584):342–50. doi: 10.1016/s0140-6736(07)61165-9.
  2. Волкова М.А. Амбулаторное лечение и диспансеризация больных хроническими лейкозами. М.: Медицина, 1979. 216 с.
    [Volkova MA. Ambulatornoe lechenie i dispanserizatsiya bol’nykh khronicheskimi leikozami. (Outpatient treatment and mass health examination of patients with chronic leukemias.) Moscow: Meditsina Publ.; 1979. 216 p. (In Russ)]
  3. Туркина А.Г., Челышева Е.Ю. Стратегия терапии хронического миелолейкоза: возможности и перспективы. Терапевтический архив. 2013;7:4–9.
    [Turkina AG, Chelysheva EYu. Therapeutic strategies for chronic myeloid leukemia: possibilities and perspectives. Terapevticheskii arkhiv. 2013;7:4–9. (In Russ)]
  4. Bjorkholm M, Ohm L, Eloranta S, et al. Success story of targeted therapy in chronic myeloid leukemia: a population-based study of patients diagnosed in Sweden from 1973 to 2008. J Clin Oncol. 2011;29(18):2514–20. doi: 10.1200/jco.2011.34.7146.
  5. Kantarjian H, O’Brien S, Jabbour E, et al. Improved survival in chronic myeloid leukemia since the introduction of imatinib therapy: a single-institution historical experience. Blood. 2012;119(9):1981–7. doi: 10.1182/blood-2011-08-358135.
  6. 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.
  7. Зарицкий А.Ю., Ломаиа Э.Г., Виноградова О.Ю. и др. Результаты многоцентрового исследования терапии гливеком больных хроническим миелолейкозом в хронической фазе. Гематология и трансфузиология. 2007;2:13–7.
    [Zaritskii AYu, Lomaia EG, Vinogradova OYu, et al. Results of a multi-center study of treatment of patients with chronic phase chronic myeloid leukemia with glivec. Gematologiya i transfuziologiya. 2007;2:13–7. (In Russ)]
  8. Лазарева О.В., Туркина А.Г., Гусарова Г.А. и др. Итоги 12-летней терапии ингибиторами тирозинкиназ больных в поздней хронической фазе хронического миелолейкоза после неудач лечения ИФН-a. Сибирский научный медицинский журнал. 2015;35(1):90–7.
    [Lazareva OV, Turkina AG, Gusarova GA, et al. The results of 12 years treatment of patients in late chronic phase of chronic myeloid leukemia by tyrosine kinase inhibitors after failure INF-a. Sibirskii nauchnyi meditsinskii zhurnal. 2015;35(1):90–7. (In Russ)]
  9. Шухов О.А. Молекулярная и цитогенетическая характеристика Ph-позитивного клона у больных хроническим миелолейкозом при длительном воздействии ингибиторов тирозинкиназ: Автореф. дис. ¼ канд. мед. наук. М., 2015. С. 10–11.
    [Shukhov OA. Molekulyarnaya i tsitogeneticheskaya kharakteristika Ph-pozitivnogo klona u bol’nykh khronicheskim mieloleikozom pri dlitel’nom vozdeistvii ingibitorov tirozinkinaz. (Molecular and cytogenetic characteristics of Ph-positive clone in patients with chronic myeloid leukemia after a long-term exposure to tyrokinase inhibitors.) [dissertation] Moscow; 2015. pр. 10–11. (In Russ)]
  10. O’Brien S, Abboud CN, Akhtari M, et al. Clinical Practice Guidelines in Oncology. Chronic Myelogenous Leukemia, Version 1.2014. J Natl Compr Canc Netw. 2013;11:1327–40.
  11. Воробьев А.И. (ред.). Руководство по гематологии. М.: Ньюдиамед, 2005. Т. 1. С. 251–3.
    [Vorob’ev AI, ed. Rukovodstvo po gematologii. (Guidelines on hematology.) Moscow: Newdiamed Publ.; 2005. Vol. 1. pp. 251–3. (In Russ)]
  12. Rowe JM. Clinical and laboratory features of the myeloid and lymphocytic leukemias. Am J Med Technol. 1983;49(2):103–9.
  13. Hughes TP, Goldman JM. Chronic myeloid leukemia. In: Hematology, Basic Principles and Practice, 2nd edition. Ed. by R Hoffman. New York: Churchill Livingstone; 1995. pp. 854–69.
  14. Lichtman MA. Chronic myelogenous leukemia and related disorders. In: Williams Hematology, 5th edition. Ed. by E Beutler, MA Lichtman, et al. New York: McGraw-Hill; 1995. pp. 298–324.
  15. Athens JW. Chronic Myeloid Leukemia. In: Wintrobe’s Clinical Hematology, 9th edition. Ed. by GR Lee, TC Bithell, et al. Philadelphia: Lea & Febiger; 1993. pp. 1969–98.
  16. Savage DG, Szydlo RM, Goldman JM. Clinical features at diagnosis in 430 patients with chronic myeloid leukaemia seen at a referral center over a 16-year period. Br J Haematol. 1997;96(1):111–6. doi: 10.1046/j.1365-2141.1997.d01-1982.x.
  17. O’Brien SG, Guilhot F, Larson R, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2003;348(11):994–1004. doi: 10.1056/nejmoa022457.
  18. Cortes JE, Baccarani M, Guilhot F, et al. Phase III, randomized, open-label study of daily imatinib mesylate 400 mg versus 800 mg in patients with newly diagnosed, previously untreated chronic myeloid leukemia in chronic phase using molecular endpoints: tyrosine kinase inhibitor optimization and selectivity study. J Clin Oncol. 2009;28(3):424–30. doi: 10.1200/jco.2009.25.3724.
  19. Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010;362(24):2251–9. doi: 10.1056/nejmoa0912614.
  20. Kantarjian H, Shah NP, Hochhaus A, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2010;362(24):2260–70. doi: 10.1056/nejmoa1002315.
  21. Cortes JE, Kim DW, Kantarjian HM, et al. Bosutinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: results from the BELA trial. J Clin Oncol. 2012;30(28):3486–92. doi: 10.1200/jco.2011.38.7522.
  22. Castagnetti F, Testoni N, Luatti S, et al. Deletions of the derivative chromosome 9 do not influence the response and the outcome of chronic myeloid leukemia in early chronic phase treated with imatinib mesylate. GIMEMA CML Working Party analysis. J Clin Oncol. 2010;28(16):2748–54. doi: 10.1200/jco.2009.26.7963.
  23. Gugliotta G, Castagnetti F, Palandri F, et al. Frontline imatinib treatment of chronic myeloid leukemia: no impact of age on outcome, a survey by the GIMEMA CML working party. Blood. 2011;117(21):5591–9. doi: 10.1182/blood-2010-12-324228.
  24. Preudhomme C, Guilhot J, Nicolini FE, et al. Imatinib plus Peginterferon alfa-2a in chronic myeloid leukemia. N Engl J Med. 2010;363(26):2511–21. doi: 10.1056/nejmoa1004095.
  25. Hehlmann R, Lauseker M, Jung-Munkwitz S, et al. Tolerability-adapted imatinib 800 mg/d versus 400 mg/d versus 400 mg/d plus interferon-ɑ in newly diagnosed chronic myeloid leukemia. J Clin Oncol. 2011;29(12):1634–42. doi: 10.1200/jco.2010.32.0598.
  26. De Lavallade H, Apperley JF, Khorashad J, et al. Imatinib for newly diagnosed patients with chronic myeloid leukemia: incidence of sustained responses in an intention-to-treat analysis. J Clin Oncol. 2008;26(20):3358–63. doi: 10.1200/jco.2007.15.8154.
  27. Jain P, Kantarjian H, Nazha A, et al. Early responses predict better outcomes in patients with newly diagnosed chronic myeloid leukemia: results with four tyrosine kinase inhibitor modalities. Blood. 2013;121(24):4867–74. doi: 10.1182/blood-2013-03-490128.
  28. Абдулкадыров, Туркина А.Г. и др. Рекомендации по диагностике и терапии хронического миелолейкоза. СПб., М., 2013. С. 67.
    [Abdulkadyrov KM, Turkina AG, et al. Rekomendatsii po diagnostike i terapii khronicheskogo mieloleikoza. (Guidelines for diagnosis and treatment of chronic myeloid leukemia.) Saint Petersburg, Moscow; 2013. pр. 67. (In Russ)]
  29. European Leukemia Net. [Internet] Available from: http://www.leukemia-net.org/content/international_trials/structure/index_eng.html. (accessed 05.01.2017).
  30. Hasford J, Baccarani M, Hoffmann V, et al. Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: the EUTOS score. Blood. 2011;118(3):686–92. doi: 10.1182/blood-2010-12-319038.
  31. Sokal JE, Cox EB, Baccarani M, et al. Prognostic discrimination in «good-risk» chronic granulocytic leukemia. Blood. 1984;63(4):789–99.
  32. Hasford J, Pfirmann M, Hehlmann R, et al. A new prognostic score for survival of patients with chronic myeloid leukemia treated with interferon alfa. J Natl Cancer Inst. 1998;90(11):850–8. doi: 10.1093/jnci/90.11.850.
  33. Куликов С.М., Виноградова О.Ю., Челышева Е.Ю. и др. Заболеваемость хроническим миелолейкозом в 6 регионах России по данным популяционного исследования 2009–2012 гг. Терапевтический архив. 2014;86(7):24–30.
    [Kulikov SM, Vinogradova OYu, Chelysheva EYu, et al. Incidence of chronic myeloid leukemia in 6 regions of Russia according to the data of the 2009–2012 population-based study. Terapevticheskii arkhiv. 2014;86(7):24–30. (In Russ)]
  34. Lazareva OV, Chelysheva EYu, Tischenko IA, et al. Incidence of second neoplasm in patients with chronic myeloid leukemia analyzed in the frame of international research project EUTOS population based study in Russian Federation. EHA-20. EHA Learning Center. 2015: Abstract 103689.
  35. Deininger MWN, Druker BJ. Specific Targeted Therapy of Chronic Myelogenous Leukemia with Imatinib. Pharmacol Rev. 2003;55(3):401–23. doi: 10.1124/pr.55.3.4.
  36. Talpaz M, Silver RT, Druke BJ, et al. Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study. Blood. 2002;99(6):1928–37. doi: 10.1182/blood.v99.6.1928.
  37. Lee SJ. Chronic myeloid leukemia. Br J Haematol. 2000;111(4):993–1009. doi: 10.1111/j.1365-2141.2000.02216.x.
  38. Туркина А.Г., Голенков А.К., Напсо Л.И. и др. Российский регистр по лечению хронического миелоидного лейкоза в рутинной клинической практике: итоги многолетней работы. Эффективная фармакотерапия. 2015;10:10–5.
    [Turkina AG, Golenkov AK, Napso LI, et al. Russian Registry for treatment of chronic myeloid leukemia in routine clinical practice: results of many years of work. Effektivnaya farmakoterapiya. 2015;10:10–5. (In Russ)]
  39. Hoffmann VS, Baccarani M, Hasford J, et al. The EUTOS population-based registry: incidence and clinical characteristics of 2904 CML patients in 20 European countries. Leukemia. 2015;29(6):1336–43. doi: 10.1038/leu.2015.73.
  40. Hehlman R. How I treat CML blast crisis. Blood. 2012;120(4):737–47. doi: 10.1182/blood-2012-03-380147.
  41. Body weight index. [Internet] Available from: http://www.calculator-imt.com. (accessed 05.01.2017).

 

 

Molecular Monitoring of RUNX1-RUNX1T1 Transcript Level in Acute Myeloblastic Leukemias on Treatment

LL Girshova, EG Ovsyannikova, SO Kuzin, EN Goryunova, RI Vabishchevich, AV Petrov, DV Motorin, DV Babenetskaya, VV Ivanov, KV Bogdanov, IV Kholopova, TS Nikulina, YuV Mirolyubova, YuA Alekseeva, AYu Zaritskii

VA Almazov Federal North-West Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

For correspondence: Ekaterina Gennad’evna Ovsyannikova, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel: +7(921)313-68-35; e-mail: katrin51297@mail.ru

For citation: Girshova LL, Ovsyannikova EG, Kuzin SO, et al. Molecular Monitoring of RUNX1-RUNX1T1 Transcript Level in Acute Myeloblastic Leukemias on Treatment. Clinical oncohematology. 2016;9(4):456–64 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-456-464


ABSTRACT

Background. The current approach to treatment of acute myeloblastic leukemia (AML) includes the achievement of maximum tumor reduction and, therefore, eradication of a leukemic clone. The goal of the therapy is to achieve undetectable levels of the target gene, except an isolated molecular rearrangement of RUNX1-RUNX1T1.

Aim. To estimate the dynamics of the RUNX1-RUNX1T1 level and relevant clinical manifestations during the monitoring of various stages of the program therapy and after its completion.

Methods. The article presents a description of 10 cases of AML with isolated RUNX1-RUNX1T1 expression (n = 4) and the expression in combination with different molecular and cytogenetic abnormalities (= 6). In addition, a long-term monitoring of the gene expression by quantitative determination of RUNX1-RUNX1T1 using a real-time PCR was presented.

Results. The incidence of relapses in a group with a decreased RUNX1-RUNX1T1 expression level of >2 log is 75 % as compared to patients with a less significant reduction of the transcript level (with the relapse incidence equal to 0 %) (= 0.05). The increase of the RUNX1-RUNX1T1 level against the background of bone marrow remission by more than 1 log coincided with a bone marrow relapse within 5–18 weeks. In addition, long-term persistence of a certain transcript level after the completion of a program therapy without relapse is possible.

Conclusion. The study analyzed possible molecular background of different clinical outcomes of long-term persistence of the RUNX1-RUNX1T1 transcript that might lead to an individualized approach to AML patients.


Keywords: acute myeloblastic leukemia, AML, RUNX1-RUNX1T1, molecular monitoring.

Received: April 5, 2016

Accepted: April 18, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Bitter MA, Le Beau MM, Rowley JD, et al. Association between morphology, karyotype, and clinical features in myeloid leukemias. Hum Pathol. 1987;18(3):211–25. doi: 10.1016/s0046-8177(87)80002-3.
  2. Mrozek K, Heinonen K, de la Chapelle A, Bloomfield CD. Clinical significance of cytogenetics in acute myeloid leukemia. Semin Oncol. 1997;24(1):17–31.
  3. Rowe D, Cotterill SJ, Ross FM, et al. Cytogenetically cryptic AML1-ETO and CBFbeta-MYH11 gene rearrangement: incidence in 412 cases of acute myeloid leukaemia. Br J Haematol. 2000;111(4):1051–6. doi: 10.1111/j.1365-2141.2000.02474.x.
  4. Downing JR. AML1/CBFbeta transcription complex: its role in normal hematopoiesis and leukemia. Leukemia. 2001;15(4):664–5. doi: 10.1038/sj.leu.2402035.
  5. Рулина А.В., Спирин П.В., Прасолов В.С. Активированные лейкозные онкогены AML1-ETO и C-KIT: роль в развитии острого миелоидного лейкоза и современные подходы к их ингибированию. Успехи биологической химии. 2010;50:349–86.
    [Rulina AV, Spirin PV, Prasolov VS. Activated leukemic AML1-ETO и C-KIT oncogenes: their role in the development of acute myeloid leukemia and modern approaches to their inhibition. Uspekhi biologicheskoi khimii. 2010;50:349–86. (In Russ)]
  6. Grimwade D, Walker H, Oliver F, et al. A on behalf of the Medical Research Council Adult and Children’s Leukaemia Working Parties. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. Blood. 1998;92(7):2322–33.
  7. Lowenberg B. Postremission treatment of acute myelogenous leukemia. N Eng J Med. 1995;332(4):260–2. doi: 10.1056/nejm199501263320411.
  8. 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(12):3767–75.
  9. Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood. 2002;100(13):4325–36. doi: 10.1182/blood-2002-03-0772.
  10. Byrd JC, Ruppert AS, Mrozek K, et al. Repetitive cycles of high-dose cytarabine benefit patients with acute myeloid leukemia and inv(16) (p13q22) or t(16;16): results from CALGB 8461. J Clin Oncol. 2004;22(6):1087–94. doi: 10.1200/jco.2004.07.012.
  11. Schlenk 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.
  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. Yin JAL, O’Brien MA, Hills RK, et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 Trial. Blood. 2012;120(14):2826–35. doi: 10.1182/blood-2012-06-435669.
  14. Jourdan E, Boissel N, Chevret S, et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood. 2013;121(12):2213–23. doi: 10.1182/blood-2012-10-462879.
  15. Byrd JC, Weiss RB, Arthur DC, et al. Extramedullary leukemia adversely affects hematologic complete remission rate and overall survival in patients with t(8;21)(q22;q22): results from Cancer and Leukemia Group B 8461. J Clin Oncol. 1997;15(2):466–75.
  16. Nguyen S, Leblanc T, Fenaux P, et al. A white blood cell index as the main prognostic factor in t(8;21) acute myeloid leukemia (AML): a survey of 161 cases from the French AML Intergroup. Blood. 2002;99(10):3517–23. doi: 10.1182/blood.V99.10.3517.
  17. Baer MR, Stewart CC, Lawrence D, et al. Expression of the neural cell adhesion molecule CD56 is associated with short remission duration and survival in acute myeloid leukemia with t(8;21)(q22;q22). Blood. 1997;90(4):1643–8.
  18. Schoch C, Haase D, Haferlach T, et al. Fifty-one patients with acute myeloid leukemia and translocation t(8;21)(q22;q22): an additional deletion in 9q is an adverse prognostic factor. Leukemia. 1996;10(8):1288–95.
  19. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24(24):3904–11. doi: 10.1200/jco.2006.06.9500.
  20. Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia. 2006;20(6):965–70. doi: 10.1038/sj.leu.2404188.
  21. Hoyos M, Nomdedeu JF, Esteve J, et al. Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease. Eur J Haematol. 2013;91(3):209–18. doi: 10.1111/ejh.12130.
  22. Демидова И.А. Использование молекулярно-биологических методов для определения генетических нарушений при миелоидных лейкозах и мониторирования минимальной остаточной болезни. Онкогематология. 2007;4:17–25.
    [Demidova IA. Application of molecular-biological methods for determining genetic disorders in myeloid leukemias and monitoring of minimal residual diseases. Onkogematologiya. 2007;4:17–25. (In Russ)]
  23. Estey EH. Acute myeloid leukemia: 2013 update on risk-stratification and management. Am J Hematol. 2013;88(4):318–27. doi: 10.1002/ajh.23404.
  24. Buccisano F, Maurillo L, Del Principe MI, et al. Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia. Blood. 2012;119(2):332–41. doi: 10.1182/blood-2011-08-363291.
  25. Tobal K, Newton J, Nige S, et al. Molecular quantitation of minimal residual disease in acute myeloid leukemia with с t(8;21) can identify patients in durable remission and predict clinical relapse. Blood. 2000;95(3):815–9.
  26. Yin JAL, O’Brien MA, Hills RK, et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 Trial. Blood. 2012;120(14):2826–30. doi: 10.1182/blood-2012-06-435669.
  27. Jourdan E, Boissel N, Chevret S, et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood. 2013;121(12):2213–23. doi: 10.1182/blood-2012-10-462879.
  28. Zhu H-H, Zhang X-H, Qin Y-Z, et al. MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial. Blood. 2013;121(2):4056–62. doi: 10.1182/blood-2012-11-468348.
  29. Morschhauser F, Cayuela JM, Martini S, et al. Evaluation of minimal residual disease using reverse-transcription polymerase chain reaction in t(8;21) acute myeloid leukemia: a multicenter study of 51 patients. J Clin Oncol. 2000;18(4):778–94.
  30. Willekens C, Blanchet O, Renneville A, et al. Prospective long-term minimal residual disease monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: results of the French CBF-2006 trial. Haematologica. 2016;101(3):328–35. doi: 10.3324/haematol.2015.131946.
  31. Schnittger S, Weisser M, Schoch C, et al. New score predicting for prognosis in PML-RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid leukemia based on quantification of fusion transcripts. Blood. 2003;102(8):2746–55. doi: 10.1182/blood-2003-03-0880.
  32. Ommen HB, Schnittger S, Jovanovic JV, et al. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood. 2010;115(2):198–205. doi: 10.1182/blood-2009-04-212530.
  33. Krauter J, Gorlich K, Ottmann O, et al. Prognostic value of minimal residual disease quantification by real-time reverse transcriptase polymerase chain reaction in patients with core binding factor leukemias. J Clin Oncol. 2003;21(23):4413–22. doi: 10.1200/jco.2003.03.166.
  34. Marcucci G, Livak KJ, Bi W, et al. Detection of minimal residual disease in patients with AML1/ETO-associated acute myeloid leukemia using a novel quantitative reverse ttranscriptase polymerase chain reaction assay. Leukemia. 1998;12(9):1482–9. doi: 10.1038/sj.leu.2401128.
  35. Lane S, Saal R, Molle P, et al. A ³ 1 log rise in RQ-PCR transcript levels defines molecular relapse in core binding factor acute myeloid leukemia and predicts subsequent morphologic relapse. Leuk Lymphoma. 2008;49(3):517–23. doi: 10.1080/10428190701817266.
  36. Perrea G, Lasa A, Aventi A, et al. Prognostic value of minimal residual disease in acute myeloid leukemia with favorable cytogenetics [t(8.21) and inv(16)]. Leukemia. 2006;20(1):87–94. doi: 10.1038/sj.leu.2404015.
  37. Jaso JM, Wang SA, Jorgensen JL, et al. Multicolor flow cytometric immunophenotyping for detection of minimal residual disease in AML: Past, present and future. Bone Marrow Transplant. 2014;49(9):1129–38. doi: 10.1038/bmt.2014.99.
  38. Bruggermann M, Raff T, Flohr T, et al. Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006;107(3):1116–23. doi: 10.1182/blood-2005-07-2708.
  39. Borowitz MJ, Devidas M, Hunger SP, et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship in other prognostic factors: a Children’s Oncology Group Study. Blood. 2008;111(12):5477–85. doi: 10.1182/blood-2008-01-132837.
  40. Muller MC, Cross NC, Erben P, et al. Harmonisation of molecular monitoring of CML therapy in Europe. Leukemia. 2009;23(11):1957–63. doi: 10.1038/leu.2009.168.
  41. Jurlander J, Caligiuri MA, Ruutu T, et al. Persistence of the AMLI/ETO Fusion Transcript in Patients Treated With Allogeneic Bone Marrow Transplantation for t(8;21) Leukemia. Blood. 1996;88(6):2183–219.
  42. Kayser S, Schlenk RF, Grimwade D, et al. Minimal residual disease–directed therapy in acute myeloid leukemia. Blood. 2015;125(15):2331–5. doi: 10.1182/blood-2014-11-578815.
  43. Evans PA, Short MA, Jack AS, et al. Detection and quantitation of the transcripts associated with the inv(16) in presentation and follow-up samples from patients with AML. Leukemia. 1997;11(3):364–9. doi: 10.1038/sj.leu.2400578.
  44. Laczika K, Novak M, Hilgarth B, et al. Competitive CBFbeta/MYH11 reverse transcriptase polymerase chain reaction for quantitative assessment of minimal residual disease during post remission therapy in acute myeloid leukemia with inversion 16: a pilot study. J Clin Oncol. 1998;16(4):1519–25.
  45. Krauter J, Hoellge W, Wattjies MP, et al. Detection and quantitation of CBFB/MYH11 fusion transcript in patients with inv(16) positive acute myeloblastic leukemia by real-time RT-PCR. Genes Chromos Cancer. 2001;30(4):342–8. doi: 10.1002/gcc.1100.
  46. Marcucci G, Caligiuri MA, Dohner H, et al. Quantification of CBFbeta/MYH11 fusion trancript byreal-time RT-PCR in patients with inv(16) acute myeloid leukemia. Leukemia. 2001;15(7):1072–80. doi: 10.1038/sj.leu.2402159.
  47. Duployez N, Willekens C, Marceau-Renaut A, et al. Prognosis and monitoring of core-binding factor acute myeloid leukemia: current and emerging factors. Exp Rev Hematol. 2014;8(1):43–56. doi: 10.1586/17474086.2014.976551.
  48. Shima T, Miyamoto T, Kikushige Y, et al. The ordered acquisition of Class II and Class I mutations directs formation of human t(8;21) acute myelogenous leukemia stem cell. Exp Hematol. 2014;42(11):955–65. doi: 10.1016/j.exphem.2014.07.267.
  49. Buonamici S, Ottaviani E, Visani G, et al. Patterns of AML-ETO1 transcript expression in patients with acute myeloid leukemia and t(8;21) in complete hematologic remission. Haematologica. 2004;89(1):103–5.
  50. Song J, Mercer D, Hu X, et al. Common Leukemia- and Lymphoma-Associated Genetic Aberrations in Healthy Individuals. J Mol Diagn. 2011;13(2);213–9. doi: 10.1016/j.jmoldx.2010.10.009.
  51. Miyamoto T, Weissman IL, Akashi K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Nat Acad Sci. 2000;97(13):7521–6. doi: 10.1073/pnas.97.13.7521.
  52. Yin JAL, Tobal K. Detection of minimal residual disease in acute myeloid leukemias: methodologies, clinical and biological significance. Br J Haematol. 1999;106(3):578–90. doi: 10.1046/j.1365-2141.1999.01522.x.
  53. Corces-Zimmerman MR, Hong W-J, Weissman IL, et al. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Nat Acad Sci. 2014;111(7):2548–53. doi: 10.1073/pnas.1324297111.
  54. Russler-Germain DA, Spencer DH, Young MA, et al. The R882H DNMT3A Mutation Associated with AML Dominantly Inhibits Wild-Type DNMT3A by Blocking Its Ability to Form Active Tetramers. Cancer Cell. 2014;25(4):442–54. doi: 10.1016/j.ccr.2014.02.010.
  55. Ommen HB, Schnittger S, Jovanovic JV, et al. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood. 2010;115(2):198–205. doi: 10.1182/blood-2009-04-212530.
  56. Wang Y, Wu D-P, Liu Q-F, et al. In adults with t(8;21) AML, posttransplant RUNX1/RUNX1T1-based MRD monitoring, rather than c-KIT mutations, allows further risk stratification. Blood. 2014;124(12):1880–6. doi: 10.1182/blood-2014-03-563403.

 

Rearrangement of 11q23 Chromosome Region in Acute Myeloid Leukemias in Children

EV Fleishman1, OI Sokova1, AV Popa1, GA Tsaur2,3,4, LN Konstantinova1, OM Plekhanova2, MV Strigaleva2, ES Nokhrina2, VS Nemirovchenko1, OR Arakaev2,3

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

2 District Children’s Hospital No. 1, 32 S. Deryabinoy str., Yekaterinburg, Russia, 620149

3 Research Institute of Medical Cell Technologies, 22a Karla Marksa str., Yekaterinburg, Russia, 620026

4 The First President of Russia BN Yeltsin Ural Federal University, 19 Mira str., Yekaterinburg, Russia, 620002

For correspondence: Elena Vol’fovna Fleishman, DSci, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel: +7(499)323-57-22; e-mail: flesok@yandex.ru

For citation: Fleishman EV, Sokova OI, Popa AV, et al. Rearrangement of 11q23 Chromosome Region in Acute Myeloid Leukemias in Children. Clinical oncohematology. 2016;9(4):446–55 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-446-455


ABSTRACT

Aim. To study characteristics of 11q23 involvement, age-specific differences in the incidence of these chromosomal markers in acute myeloid leukemias (AML) in children, and to determine their prognostic significance in patients treated according to the protocols applied in leading Russian pediatric hematological clinics.

Methods. The chromosomal analysis of bone marrow and peripheral blood cells has been performed prior to initiation of treatment in 395 children with primary AML aged from 0 to 16 years. The patients were treated in pediatric hematological clinics of Moscow and Moscow Region and in Yekaterinburg District Children’s Hospital No. 1. Clinical outcomes of 300 followed-up pediatric patients treated with similar modern therapy protocols were analyzed to evaluate the prognostic impact of 11q23/MLL abnormalities. To determine the incidence of 11q23/MLL rearrangements in AML of different age groups, we examined not only children, but also adult patients (= 212).

Results. In AML, the frequency of changes in the 11q23 region exceeded 40 % in children aged from 0 to 2 years. The frequency decrease with age and in patients over 40 years it was only 2 %. Significant heterogeneity of changes in karyotypes with 11q23/MLL rearrangements was observed: both various translocations with different regions of other chromosomes, and 11q23 deletions were detected. In addition, a great variability of numerical and structural additional chromosomal abnormalities was observed. The 10-year relapse-free survival rates (30.4 ± 6.7 %) and overall survival rates (35.1 ± 7.0 %) in AML with changes in the 11q23 region (= 61) were significantly lower than those in patients from the intermediate risk group (n = 103): 48.9 ± 5.8 % and 43.8 ± 7.5 %, respectively (= 0.035). The data are close to those in the high-risk group (n = 44): 35.9 ± 8.1 % and 38.3 ± 7.6 %, respectively. The study failed to confirm the published data that t(9;11) is a more favorable prognostic factor, and that t(6;11) and t(10;11) are less favorable ones than all other 11q23 translocations. Our results did not confirm a negative prognostic effect of additional chromosome abnormalities associated with 11q23 rearrangements.

Conclusion. Pediatric AML patients with 11q23 abnormalities should be included in a high-risk group if therapy is performed according protocols applied in leading hematological centers of Russia.


Keywords: pediatric acute myeloid leukemias, 11q23/MLL rearrangements, risk groups.

Received: May 12, 2016

Accepted: June 15, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Mitelman F. Catalog of chromosome aberrations in cancer. 5th edition. Willey-Liss; 1995.
  2. Heim S, Mitelman F. Cancer Cytogenetics. 2nd edition. Wiley-Liss; 1995.
  3. Heim S, Mitelman F. Cancer Cytogenetics. Chromosomal and Molecular Genetic Aberrations of Tumor Cells. 3rd edition. Wiley-Blackwell; 2009.
  4. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  5. Marshalek R. MLL leukemia and future treatment strategies. Arch Pharm Chem Life Sci. 2015;348(4):1–8. doi: 10.1002/ardp.201400449.
  6. Balgobind BV, Zwaan CM, Pieters R, van den Heuvel-Eibrink MM. The heterogeneity of pediatric MLL-rearranged acute myeloid leukemia. Leukemia. 2011;25(8):1239–48. doi: 10.1038/leu.2011.90.
  7. Meyer C, Hoffmann J, Burmeister T, et al. The MLL recombinome of acute leukemias in 2013. Leukemia. 2013;27(11):2165–76. doi: 10.1038/leu.2013.135.
  8. Balgobind BV, Raimondi SC, Harbott J, et al. Novel prognostic subgroups in childhood 11q23/MLL – rearranged acute myeloid leukemia: results of an International retrospective study. Blood. 2009;114(12):2489–96. doi: 10.1182/blood-2009-04-
  9. Grimwade D, Walker H, Oliver F, et al. The importance of diagnostic cytogenetics on outcome of 1,612 patients entered into the MRC AML 10 trial. Blood. 1998;92(7):2322–33.
  10. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United kingdom Medical Research Council trials. Blood. 2010;116(3):354–65. doi: 10.1182/blood-2009-11-
  11. Mrozek K, Heinonen K, Lawrence D, et al. Adult patients with de novo acute myeloid leukemia and t(9;11)(p22;q23) have a superior outcome to patients with other translocations involving band 11q23: a Cancer and Leukemia Group Study. Blood. 1997;90(11):4532–8.
  12. Rubnitz JE, Raimondi SC, Tong X, et al. Favorable impact of the t(9;11) in childhood acute myeloid leukemia. J Clin Oncol. 2002;20(9):2302–9. doi: 1200/jco.2002.08.400.
  13. von Neuhoff C, Reinhardt D, Sander A, et al. Prognostic impact of specific chromosomal aberrations in large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol. 2010;28(16):2682–8. doi: 1200/jco.2009.25.6321.
  14. Blum W, Mrozek K, Ruppert AS, et al. Adult de novo acute myeloid leukemia with t(6;11)(q27;q23). Cancer. 2004;101(6):1420–7. doi: 10.1002/cncr.20489.
  15. Karol SE, Coustan-Smith E, Cao X, et al. Prognostic factors in children with acute myeloid leukemia and excellent response to remission induction therapy. Br J Haematol. 2015;168(1):94–101. doi: 1111/bjh.13107.
  16. Coenen EA, Raimondi SC, Harbott J, et al. Prognostic significance of additional cytogenetic aberrations in 733 de novo pediatric 11q23/MLL-rearranged AML patients: results of an international study. Blood. 2011;117(26):7102–11. doi: 1182/blood-2010-12-328302.
  17. Schaffer L, McGovan-Jordan J, Schmid M. ISCN. An International System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013. pp. doi: 10.1002/ajmg.a.35995.
  18. Цаур Г.А., Наседкина Т.В., Попов А.М. и др. Время достижения молекулярной ремиссии как фактор прогноза у детей первого года жизни острым лимфобластным лейкозом. Онкогематология. 2010;2:46–54.
    [Tsaur GA, Nasedkina TV, Popov AM, et al. Time required to achieve molecular remission as a prognostic factor in children of the first year of life with acute lymphoblastic leukemia. Onkogematologiya. 2010;2:46–54. (In Russ)]
  19. Meyer C, Schneider B, Reichel M, et al. Diagnostic tool for the identification of MLL rearrangements including unknown partner genes. Proc Natl Acad Sci USA. 2005;102(2):449–54. doi: 10.1073/pnas.0406994102.
  20. Kaplan E, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53(282):457–81. doi: 1080/01621459.1958.10501452.
  21. Meyer С, Kowarz E, Hofmann J, et al. New insights to the MLL recombinome of acute leukemias. Leukemia. 2009;23(8):1490–9. doi: 10.1038/leu.2009.33.
  22. Ross M, Mahfouz R, Onciu M, et al. Gene expression profiling of pediatric acute myelogenous leukemia. Blood. 2004;104(12):3679–87. doi: 10.1182/blood-2004-03-1154.
  23. Shih L, Liang D, Fu J, et al. Characterization of fusion partner genes in 114 patients with de novo acute myeloid leukemia and MLL rearrangement. Leukemia. 2006;20:218–23. doi: 10.1038/sj.leu.2404024.
  24. Balgobind B, Hollink I, Reinhardt D, et al. Low frequency of MLL-partial tandem duplications in paediatric acute myeloid leukaemia using MLPA as a novel DNA screenings technique. Eur J Cancer. 2010;46(10):1892–9. doi: 10.1016/j.ejca.2010.02.019.
  25. Shimada A, Taki T, Tabuchi K, et al. Tandem duplications of MLL and FLT3 are correlated with poor prognoses in pediatric acute myeloid leukemia: a study of the Japanese childhood AML Cooperative Study Group. Pediatr Blood Cancer. 2008;50(2):264–9. doi: 10.1002/pbc.21318.
  26. Steudel C, Wermke M, Schaich M, et al. Comparative analysis of MLL partial tandem duplication and FLT3 internal tandem duplication mutations in 956 adult patients with acute myeloid leukemia. Gene Chromos Cancer. 2003;37(3):237–51. doi: 10.1002/gcc.10219.
  27. Rege-Cambrin G, Giugliano E, Michaux L, et al. Trisomy 11 in myeloid malignancies is associated with internal tandem duplication of both MLL and FLT3 genes. Haematologica. 2005;90(2):262–4.
  28. Swansbury GJ, Slater R, Bain BJ, et al. Hematological malignancies with t(9;11)(p21-22;q23) – laboratory and clinical study of 125 cases. European 11q23 Workshop participants. Leukemia. 1998;12(5):792–800. doi: 10.1038/sj.leu.2401014.
  29. Harrison CJ, Hills RK, Moorman AV, et al. Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML10 and 12. J Clin Oncol. 2010;28(16):2674–81. doi: 10.1200/jco.2009.24.8997.
  30. Pession A, Masetti R, Rizzari C, et al. Results of the ALEOP AML 2002/01 multicenter prospective trial for the treatment of children with acute myeloid leukemia. Blood. 2013;122(2):170–8. doi: 10.1182/blood-2013-03-491621.
  31. Schoch C, Schnittger S, Klaus M, et al. AML with 11q23/MLL abnormalities as defined by the WHO classification: incidence, partner chromosome, FAB subtype, age distribution, and prognostic impact in an unselected series of 1897 cytogenetically analyzed AML cases. Blood. 2003;102(7):2395–402. doi: 10.1182/blood-2003-02-0434.
  32. Tamai H, Yamaguchi H, Hamaguchi H, et al. Clinical features of adult acute leukemia with 11q23 abnormalities in Japan: A co-operative multicenter study. Int J Hematol. 2008;87(2):193–200. doi: 10.1007/s12185-008-0034-
  33. Balgobind BV, Zwaan CM, Reinhardt D, et al. High BRE expression in pediatric MLL-rearranged AML is associated with favorable outcome. Leukemia. 2010;24(12):2048–55. doi: 10.1038/leu.2010.211.
  34. 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.
  35. Ho PA, Alonzo TA, Gerbing RB, et al. High EVI1 expression is associated with MLL rearrangements and predicts decreased survival. Br J Haematol. 2013;62(5):670–7. doi: 1111/bjh.12444.
  36. Chen C, Armstrong S. Targeting DOT1L and HOX gene expression in MLL-rearranged leukemia and beyond. Exp Hematol. 2015;43(8):673–84. doi: 10.1016/j.exphem.2015.05.012.
  37. Nguyen AT, Taranova O, He J, Zhang Y. DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis. Blood. 2011;117(25):6912–22. doi: 10.1182/blood-2011-02-
  38. Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L Inhibitor EPZ-5676: safety and activity in relapsed/refractory patients with MLL-rearranged leukemia. [Internet] Available from: http://www.epizyme.com/wp-content/uploads/2014/12/ASH-EPZ-5676-Presentation-Final.pdf. (accessed 10.05.2016).

Biology of Myeloproliferative Malignancies

AL Melikyan, IN Subortseva

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

For correspondence: Irina Nikolaevna Subortseva, PhD, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-44-71; e-mail: soubortseva@yandex.ru

For citation: Melikyan AL, Subortseva IN. Biology of Myeloproliferative Malignancies. Clinical oncohematology. 2016;9(3):326-35 (In Russ).

DOI: 10.21320/2500-2139-2016-9-3-314-325


ABSTRACT

Chronic myeloproliferative diseases (WHO, 2001), or myeloproliferative neoplasms/malignancies (MPN) (WHO, 2008), are clonal diseases characterized by proliferation of one or more myelopoietic cell line in the bone marrow with signs of unimpaired terminal differentiation and is normally associated with changes in peripheral blood characteristics. The group of classical Ph-negative MPNs consists of polycythemia vera, essential thrombocythemia, primary myelofibrosis and unclassified MPNs. Acquired somatic mutations contributing to the pathogenesis of Ph-negative MPNs include JAK2 (V617F, exon 12), MPL, CALR gene mutations found in about 90 % of patients. However, these molecular events are not unique in the pathogenesis of the diseases. Mutations of other genes (ТЕТ2, ASXL1, CBL, IDH1/IDH2, IKZF1, DNMT3A, SOCS, EZH2, TP53, RUNX1, and HMGA2) are involved in formation of the disease phenotype. This review describes current concepts concerning the molecular biology of MPNs.


Keywords: chronic myeloproliferative diseases, myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia, primary myelofibrosis, genes JAK2, CALR, and MPL.

Received: April 11, 2016

Accepted: April 11, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Barbui T, Barosi G, Birgegard G, et al. Philadelphia-negative classical myeloproliferative neoplasms: critical concepts and management recommendations from European LeukemiaNet. J Clin Oncol. 2011;29(6):761–70. doi: 10.1200/jco.2010.31.8436.
  2. Tefferi A, Thiele J, Vardiman JW. The 2008 World Health Organization classification system for myeloproliferative neoplasms: order out of chaos. Cancer. 2009;115(17):3842–7. doi: 10.1002/cncr.24440.
  3. Barosi G. Essential thrombocythemia vs. early/prefibrotic myelofibrosis: why does it matter. Best Pract Res Clin Haematol. 2014;27(2):129–40. doi: 10.1016/j.beha.2014.07.004.
  4. Vannucchi AM. Management of myelofibrosis. Am Soc Hematol Educ Program. 2011;2011(1):222–30. doi:10.1182/asheducation-2011.1.222.
  5. 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.
  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. 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. 2010;115(9):1703–8. doi: 10.1182/blood-2009-09-245837.
  8. Tefferi A. How I treat myelofibrosis. Blood. 2011;117(13):3494–504. doi: 10.1182/blood-2010-11-315614.
  9. Tefferi A, Guglielmelli P, Lasho TL, et al. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia. 2014;28(7):1494–500. doi: 10.1038/leu.2014.57.
  10. Agarwal MB, Malhotra H, Chakrabarti P, et al. Myeloproliferative neoplasms working group consensus recommendations for diagnosis and management of primary myelofibrosis, polycythemia vera, and essential thrombocythemia. Indian J Med Paediatr Oncol. 2015;36(1):3–16. doi: 10.4103/0971-5851.151770.
  11. Campregher PV, Santos FP, Perini GF, Hamerschlak N. Molecular biology of Philadelphia-negative myeloproliferative neoplasms. Rev Bras Hematol Hemoter. 2012;34(2):150–5. doi: 10.5581/1516-8484.20120035.
  12. Ghoreschi K, Laurence A, O’Shea JJ. Janus kinases in immune cell signaling. Immunol Rev. 2009;228(1):273–87. doi: 10.1111/j.1600-065X.2008.00754.x.
  13. Liu KD, Gaffen SL, Goldsmith MA. JAK/STAT signaling by cytokine receptors. Curr Opin Immunol. 1998;10(3):271–8. doi: 10.1016/s0952-7915(98)80165-9.
  14. Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia. 2010;24(6):1128–38. doi: 10.1038/leu.2010.69.
  15. Riedy MC, Dutra AS, Blake TB, et al. Genomic sequence, organization, and chromosomal localization of human JAK3. Genomics. 1996;37(1):57–61. doi: 10.1006/geno.1996.0520.
  16. Saharinen P, Silvennoinen O. The pseudokinase domain is required for suppression of basal activity of Jak2 and Jak3 tyrosine kinases and for cytokine-inducible activation of signal transduction. J Biol Chem. 2002;277(49):47954–63. doi: 10.1074/jbc.M205156200.
  17. Benekli M, Baer MR, Baumann H, Wetzler M. Signal transducer and activator of transcription proteins in leukemias. Blood. 2003;101(8):2940–54. doi: 10.1182/blood-2002-04-1204.
  18. Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723–35. doi: 10.1182/blood-2011-02-292102.
  19. Lacout C, Pisani DF, Tulliez M, et al. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108(5):1652–660. doi: 10.1182/blood-2006-02-002030.
  20. James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144–8. doi: 10.1038/nature03546.
  21. Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779–90. doi: 10.1056/NEJMoa051113.
  22. Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97. doi: 10.1016/j.ccr.2005.03.023.
  23. Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. The Lancet. 2005;365(9464):1054–61. doi: 10.1016/S0140-6736(05)71142-9.
  24. Butcher CM, Hahn U, To LB, et al. Two novel JAK2 exon 12 mutations in JAK2V617F-negative polycythaemia vera patients. Leukemia. 2008;22(4):870–3. doi: 10.1038/sj.leu.2404971.
  25. Jelinek J, Oki Y, Gharibyan V, et al. JAK2 mutation 1849G>T is rare in acute leukemias but can be found in CMML, Philadelphia chromosome-negative CML, and megakaryocyticleukemia. Blood. 2005;106(10):3370–3. doi: 10.1182/blood-2005-05-1800.
  26. Pich A, Riera L, Sismondi F, et al. JAK2V617F activating mutation is associated with the myeloproliferative type of chronic myelomonocytic leukaemia. J Clin Pathol. 2009;62(9):798–801. doi: 10.1136/jcp.2009.065904.
  27. Johan MF, Goodeve AC, Bowen DT, et al. JAK2 V617F Mutation is uncommon in chronic myelomonocytic leukaemia. Br J Haematol. 2005;130(6):968. doi: 10.1111/j.1365-2141.2005.05719.x.
  28. Renneville A, Quesnel B, Charpentier A, et al. High occurrence of JAK2 V617 mutation in refractory anemia with ringed sideroblasts associated with marked thrombocytosis. Leukemia. 2006;20(11):2067–70. doi: 10.1038/sj.leu.2404405.
  29. Verstovsek S, Silver RT, Cross NC, Tefferi A. JAK2V617F mutational frequency in polycythemia vera: 100%, > 90%, less? Leukemia. 2006;20(11):2067. doi:10.1038/sj.leu.2404379.
  30. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood. 2007;110(3):840–6. doi: 10.1182/blood-2006-12-064287.
  31. Barosi G, Bergamaschi G, Marchetti M, et al. JAK2 V617F mutational status predicts progression to large splenomegaly and leukemic transformation in primary myelofibrosis. Blood. 2007;110(12):4030–6. doi: 10.1182/blood-2007-07-099184.
  32. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27(9):1861–9. doi: 10.1038/leu.2013.119.
  33. Lussana F, Caberlon S, Pagani C, et al. Association of V617F Jak2 mutation with the risk of thrombosis among patients with essential thrombocythaemia or idiopathic myelofibrosis: a systematic review. Thromb Res. 2009;124(4):409–17. doi: 10.1016/j.thromres.2009.02.004.
  34. Wang M, He N, Tian T, et al. Mutation analysis of JAK2V617F, FLT3-ITD, NPM1, and DNMT3A in Chinese patients with myeloproliferative neoplasms. BioMed Res Int. 2014;2014:485645. doi: 10.1155/2014/485645.
  35. Passamonti F, Thiele J, Girodon F, et al. A prognostic model to predict survival in 867 World Health Organization-defined essential thrombocythemia at diagnosis: a study by the International Working Group on Myelofibrosis Research and Treatment. Blood. 2012;120(6):1197–201. doi: 10.1182/blood-2012-01-403279.
  36. Barbui T, Carobbio A, Rambaldi A, Finazzi G. Perspectives on thrombosis in essential thrombocythemia and polycythemia vera: is leukocytosis a causative factor? Blood. 2009;114(4):759–63. doi: 10.1182/blood-2009-02-206797.
  37. Barbui T, Finazzi G, Carobbio A, et al. Development and validation of an International Prognostic Score of thrombosis in World Health Organization-essential thrombocythemia (IPSET-thrombosis). Blood. 2012;120(26):5128–33. doi: 10.1182/blood-2012-07-444067.
  38. Tefferi A, Pardanani A. Myeloproliferative neoplasms – a contemporary review. JAMA Oncol. 2015;1(1):97–105. doi: 10.1001/jamaoncol.2015.89.
  39. Nussenzveig RH, Swierczek SI, Jelinek J, et al. Polycythemia vera is not initiated by JAK2V617F mutation. Exp Hematol. 2007;35(1):32–8. doi: 10.1016/j.exphem.2006.11.012.
  40. Scott LM, Tong W, Levine RL, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356(5):459–68. doi: 10.1056/NEJMoa065202.
  41. Williams DM, Kim AH, Rogers O, et al. Phenotypic variations and new mutations in JAK2 V617F-negative polycythemia vera, erythrocytosis, and idiopathic myelofibrosis. Exp Hematol. 2007;35(11):1641–6. doi: 10.1016/j.exphem.2007.08.010.
  42. Passamonti F, Elena C, Schnittger S, et al. Molecular and clinical features of the myeloproliferative neoplasm associated with JAK2 exon 12 mutations. Blood. 2011;117(10):2813–6. doi: 10.1182/blood-2010-11-316810.
  43. Campbell PJ, Griesshammer M, Dohner K, et al. V617F mutation in JAK2 is associated with poorer survival in idiopathic myelofibrosis. Blood. 2006;107(5):2098–100. doi: 10.1182/blood-2005-08-3395.
  44. Martinez-Aviles L, Besses C, Alvarez-Larran A, et al. JAK2 exon 12 mutations in polycythemia vera or idiopathic erythrocytosis. Haematologica. 2007;92(12):1717–8. doi: 10.3324/haematol.12011.
  45. Sangkhae V, Etheridge SL, Kaushansky K, Hitchcock IS. The thrombopoietin receptor, MPL, is critical for development of a JAK2V617F-induced myeloproliferative neoplasm. Blood. 2014;124(26):3956–63. doi: 10.1182/blood-2014-07-587238.
  46. Chou FS, Mulloy JC. The thrombopoietin/MPL pathway in hematopoiesis and leukemogenesis. J Cell Biochem. 2011;112(6):1491-8. doi: 10.1002/jcb.23089.
  47. Abe M, Suzuki K, Inagaki O, et al. A novel MPL point mutation resulting in thrombopoietin-independent activation. Leukemia. 2002;16(8):1500–6. doi: 10.1038/sj.leu.2402554.
  48. Ding J, Komatsu H, Wakita A, et al. Familial essential thrombocythemia associated with a dominant-positive activating mutation of the c-MPL gene, which encodes for the receptor for thrombopoietin. Blood. 2004;103(11):4198–200. doi: 10.1182/blood-2003-10-3471.
  49. Moliterno AR, Williams DM, Gutierrez-Alamillo LI, et al. Mpl Baltimore: A thrombopoietin receptor polymorphism associated with thrombocytosis. Proc Natl Acad Sci USA. 2004;101(31):11444–7. doi: 10.1073/pnas.0404241101.
  50. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270. doi: 10.1371/journal.pmed.0030270.
  51. Staerk J, Lacout C, Sato T, et al. An amphipathic motif at the transmembrane-cytoplasmic junction prevents autonomous activation of the thrombopoietin receptor. Blood. 2006;107(5):1864–71. doi: 10.1182/blood-2005-06-2600.
  52. Boyd EM, Bench AJ, Goday-Fernandez A, et al. Clinical utility of routine MPL exon 10 analysis in the diagnosis of essential thrombocythaemia and primary myelofibrosis. Br J Haematol. 2010;149(2):250–7. doi: 10.1111/j.1365-2141.2010.08083.x.
  53. Lambert MP, Jiang J, Batra V, et al. A novel mutation in MPL (Y252H) results in increased thrombopoietin sensitivity in essential thrombocythemia. Am J Hematol. 2012;87(5):532–4. doi: 10.1002/ajh.23138.
  54. Hussein K, Bock O, Theophile K, et al. MPLW515L mutation in acute megakaryoblastic leukaemia. Leukemia. 2009;23(5):852–5. doi: 10.1038/leu.2008.371.
  55. Beer PA, Campbell PJ, Scott LM, et al. MPL mutations in myeloproliferative disorders: analysis of the PT-1 cohort. Blood. 2008;112(1):141–9. doi: 10.1182/blood-2008-01-131664.
  56. Akpinar TS, Hancer VS, Nalcaci M, Diz-Kucukkaya R. MPL W515L/K Mutations in chronic myeloproliferative neoplasms. Turk J Haematol. 2013;30(1):8–12. doi: 10.4274/tjh.65807.
  57. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Characteristics and clinical correlates of MPL 515W>L/K mutation in essential thrombocythemia. Blood. 2008;112(3):844–7. doi: 10.1182/blood-2008-01-135897.
  58. Teofili L, Giona F, Torti L, et al. Hereditary thrombocytosis caused by MPLSer505Asn is associated with a high thrombotic risk, splenomegaly and progression to bone marrow fibrosis. Haematologica. 2010;95(1):65–70. doi: 10.3324/haematol.2009.007542.
  59. Sun C, Zhang S, Li J. Calreticulin gene mutations in myeloproliferative neoplasms without Janus kinase 2 mutations. Leuk Lymphoma. 2015;56(6):1593–8. doi: 10.3109/10428194.2014.953153.
  60. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90. doi: 10.1056/NEJMoa1311347.
  61. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405. doi: 10.1056/NEJMoa1312542.
  62. Shirane S, Araki M, Morishita S, et al. JAK2, CALR, and MPL mutation spectrum in Japanese myeloproliferative neoplasms patients. Haematologica. 2015;100(2):46–8. doi: 10.3324/haematol.2014.115113.
  63. Lundberg P, Karow A, Nienhold R, et al. Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood. 2014;123(14):2220–8. doi: 10.1182/blood-2013-11-537167.
  64. Lavi N. Calreticulin mutations in myeloproliferative neoplasms. Rambam Maimonides Med J. 2014;5(4):e0035. doi: 10.5041/RMMJ.10169.
  65. Rumi E, Harutyunyan AS, Pietra D, et al. CALR exon 9 mutations are somatically acquired events in familial cases of essential thrombocythemia or primary myelofibrosis. Blood. 2014;123(15):2416–9. doi: 10.1182/blood-2014-01-550434.
  66. Haslam K, Langabeer SE. Incidence of CALR mutations in patients with splanchnic vein thrombosis. Br J Haematol. 2015;168(3):459–60. doi: 10.1111/bjh.13121.
  67. Turon F, Cervantes F, Colomer D, et al. Role of calreticulin mutations in the aetiological diagnosis of splanchnic vein thrombosis. J Hepatol. 2015;62(1):72–4. doi: 10.1016/j.jhep.2014.08.032.
  68. Tefferi A, Wassie EA, Lasho TL, et al. Calreticulin mutations and long-term survival in essential thrombocythemia. Leukemia. 2014;28(12):2300–3. doi: 10.1038/leu.2014.148.
  69. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia. 2014;28(7):1472–7. doi: 10.1038/leu.2014.3.
  70. Tefferi A, Lasho TL, Finke C, et al. Type 1 vs type 2 calreticulin mutations in primary myelofibrosis: differences in phenotype and prognostic impact. Leukemia. 2014;28(7):1568–70. doi: 10.1038/leu.2014.83.
  71. Shide K, Kameda T, Shimoda H, et al. TET2 is essential for survival and hematopoietic stem cell homeostasis. Leukemia. 2012;26(10):2216–23. doi: 10.1038/leu.2012.94.
  72. Ito S, D’Alessio AC, Taranova OV, et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466(7310):1129–33. doi: 10.1038/nature09303.
  73. Paulsson K, Haferlach C, Fonatsch C, et al. The idic(X)(q13) in myeloid malignancies: breakpoint clustering in segmental duplications and association with TET2 mutations. Hum Mol Genet. 2010;19(8):1507–14. doi: 10.1093/hmg/ddq024.
  74. Tefferi A, Pardanani A, Lim KH, et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia. 2009;23(5):905–11. doi: 10.1038/leu.2009.47.
  75. Martinez-Aviles L, Besses C, Alvarez-Larran A, et al. TET2, ASXL1, IDH1, IDH2, and c-CBL genes in JAK2- and MPL-negative myeloproliferative neoplasms. Ann Hematol. 2012;91(4):533–41. doi: 10.1007/s00277-011-1330-0.
  76. Patriarca A, Colaizzo D, Tiscia G, et al. TET2 mutations in Ph-negative myeloproliferative neoplasms: identification of three novel mutations and relationship with clinical and laboratory findings. BioMed Res Int. 2013;2013:929840. doi: 10.1155/2013/929840.
  77. Schaub FX, Looser R, Li S, et al. Clonal analysis of TET2 and JAK2 mutations suggests that TET2 can be a late event in the progression of myeloproliferative neoplasms. Blood. 2010;115(10):2003–7. doi: 10.1182/blood-2009-09-245381.
  78. 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.
  79. Beer PA, Delhommeau F, LeCouedic JP, et al. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood. 2010;115(14):2891–900. doi: 10.1182/blood-2009-08-236596.
  80. Ortmann CA, Kent DG, Nangalia J, et al. Effect of mutation order on myeloproliferative neoplasms. N Engl J Med. 2015;372(7):601–12. doi: 10.1056/NEJMoa1412098.
  81. 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.
  82. Carbuccia N, Murati A, Trouplin V, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009;23(11):2183–6. doi: 10.1038/leu.2009.141.
  83. Carbuccia N, Trouplin V, Gelsi-Boyer V, et al. Mutual exclusion of ASXL1 and NPM1 mutations in a series of acute myeloid leukemias. Leukemia. 2010;24(2):469–73. doi: 10.1038/leu.2009.218.
  84. Abdel-Wahab O, Adli M, LaFave LM, et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012;22(2):180–93. doi: 10.1016/j.ccr.2012.06.032.
  85. Brecqueville M, Rey J, Bertucci F, et al. Mutation analysis of ASXL1, CBL, DNMT3A, IDH1, IDH2, JAK2, MPL, NF1, SF3B1, SUZ12, and TET2 in myeloproliferative neoplasms. Genes Chromos Cancer. 2012;51(8):743–55. doi: 10.1002/gcc.21960.
  86. Katoh M. Functional and cancer genomics of ASXL family members. Br J Cancer. 2013;109(2):299–306. doi: 10.1038/bjc.2013.281.
  87. Cervantes F. How I treat myelofibrosis. Blood. 2014;124(17):2635–42. doi: 10.1182/blood-2014-07-575373.
  88. 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.
  89. Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 2008;647(1–2):21–9. doi: 10.1016/j.mrfmmm.2008.07.010.
  90. Im AP, Sehgal AR, Carroll MP, et al. DNMT3A and IDH mutations in acute myeloid leukemia and other myeloid malignancies: associations with prognosis and potential treatment strategies. Leukemia. 2014;28(9):1774–83. doi: 10.1038/leu.2014.124.
  91. 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.
  92. Yamashita Y, Yuan J, Suetake I, et al. Array-based genomic resequencing of human leukemia. Oncogene. 2010;29(25):3723–31. doi: 10.1038/onc.2010.117.
  93. Abdel-Wahab O, Pardanani A, Rampal R, et al. DNMT3A mutational analysis in primary myelofibrosis, chronic myelomonocytic leukemia and advanced phases of myeloproliferative neoplasms. Leukemia. 2011;25(7):1219–20. doi: 10.1038/leu.2011.82.
  94. Brecqueville M, Cervera N, Gelsi-Boyer V, et al. Rare mutations in DNMT3A in myeloproliferative neoplasms and myelodysplastic syndromes. Blood Cancer J. 2011;1(5):e18. doi: 10.1038/bcj.2011.15.
  95. Rudd CE. Lnk adaptor: novel negative regulator of B cell lymphopoiesis. Sci STKE. 2001;2001(85):pe1. doi: 10.1126/stke.2001.85.pe1.
  96. Gery S, Cao Q, Gueller S, et al. Lnk inhibits myeloproliferative disorder-associated JAK2 mutant, JAK2V617F. J Leuk Biol. 2009;85(6):957–65. doi: 10.1189/jlb.0908575.
  97. Soriano G, Heaney M. Polycythemia vera and essential thrombocythemia: new developments in biology with therapeutic implications. Curr Opin Hematol. 2013;20(2):169–75. doi: 10.1097/MOH.0b013e32835d82fe.
  98. Oh ST, Simonds EF, Jones C, et al. Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. Blood. 2010;116(6):988–92. doi: 10.1182/blood-2010-02-270108.
  99. Lasho TL, Pardanani A, Tefferi A. LNK mutations in JAK2 mutation-negative erythrocytosis. N Engl J Med. 2010;363(12):1189–90. doi: 10.1056/NEJMc1006966.
  100. Rathinam C, Thien CB, Flavell RA, Langdon WY. Myeloid leukemia development in c-Cbl RING finger mutant mice is dependent on FLT3 signaling. Cancer Cell. 2010;18(4):341–52. doi: 10.1016/j.ccr.2010.09.008.
  101. Loh ML, Sakai DS, Flotho C, et al. Mutations in CBL occur frequently in juvenile myelomonocytic leukemia. Blood. 2009;114(9):1859–63. doi: 10.1182/blood-2009-01-198416.
  102. Sanada M, Suzuki T, Shih LY, et al. Gain-of-function of mutated C-CBL tumour suppressor in myeloid neoplasms. Nature. 2009;460(7257):904–8. doi: 10.1038/nature08240.
  103. Zhang MY, Fung TK, Chen FY, Chim CS. Methylation profiling of SOCS1, SOCS2, SOCS3, CISH and SHP1 in Philadelphia-negative myeloproliferative neoplasm. J Cell Mol Med. 2013;17(10):1282–90. doi: 10.1111/jcmm.12103.
  104. Fourouclas N, Li J, Gilby DC, et al. Methylation of the suppressor of cytokine signaling 3 gene (SOCS3) in myeloproliferative disorders. Haematologica. 2008;93(11):1635–44. doi: 10.3324/haematol.13043.
  105. Kastner P, Chan S. Role of Ikaros in T-cell acute lymphoblastic leukemia. World J Biol Chem. 2011;2(6):108–14. doi: 10.4331/wjbc.v2.i6.108.
  106. Jager R, Kralovics R. Molecular pathogenesis of Philadelphia chromosome negative chronic myeloproliferative neoplasms. Curr Cancer Drug Targets. 2011;11(1):20–30. doi: 10.2174/156800911793743628.
  107. Ikeda K, Ogawa K, Takeishi Y. The role of HMGA2 in the proliferation and expansion of a hematopoietic cell in myeloproliferative neoplasms. Fukushima J Med Sci. 2012;58(2):91–100. doi: 10.5387/fms.58.91.
  108. Harada-Shirado K, Ikeda K, Ogawa K, et al. Dysregulation of the MIRLET7/HMGA2 axis with methylation of the CDKN2A promoter in myeloproliferative neoplasms. Br J Haematol. 2015;168(3):338–49. doi: 10.1111/bjh.13129.
  109. Raza S, Viswanatha D, Frederick L, et al. TP53 mutations and polymorphisms in primary myelofibrosis. Am J Hematol. 2012;87(2):204–6. doi: 10.1002/ajh.22216.
  110. Lu M, Hoffman R. p5 as a target in myeloproliferative neoplasms. Oncotarget. 2012;3(10):1052–3. doi: 10.18632/oncotarget.719.
  111. Gurney AL, Wong SC, Henzel WJ, de Sauvage FJ. Distinct regions of c-Mpl cytoplasmic domain are coupled to the JAK-STAT signal transduction pathway and Shc phosphorylation. Proc Natl Acad Sci USA. 1995;92(12):5292–6. doi: 10.1073/pnas.92.12.5292
  112. Tefferi A, Thiele J, Vannucchi AM, Barbui T. An overview on CALR and CSF3R mutations and a proposal for revision of WHO diagnostic criteria for myeloproliferative neoplasms. Leukemia. 2014;28(7):1407–13. doi: 10.1038/leu.2014.35.
  113. Broseus J, Park JH, Carillo S, et al. Presence of calreticulin mutations in JAK2-negative polycythemia vera. Blood. 2014;124(26):3964–6. doi: 10.1182/blood-2014-06-583161.
  114. Hasan S, Lacout C, Marty C, et al. JAK2V617F expression in mice amplifies early hematopoietic cells and gives them a competitive advantage that is hampered by IFNa. Blood. 2013;122(8):1464–77. doi: 10.1182/blood-2013-04-498956.
  115. Pardanani A, Lasho T, Finke C, et al. LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia. 2010;24(10):1713–8. doi: 10.1038/leu.2010.163.

New Сytogenetic Approaches in Patients with Primary Myelofibrosis

TL Gindina, NN Mamaev, VV Baikov, MV Barabanshchikova, EN Nikolaeva, IA Petrova, IS Moiseev, OV Pirogova, YuYu Vlasova, MO Ivanova, EV Morozova, SN Bondarenko, BV Afanasev

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, Baikov VV, et al. New Сytogenetic Approaches in Patients with Primary Myelofibrosis. Clinical oncohematology. 2016;9(1):61–9 (In Russ).

DOI: 10.21320/2500-2139-2016-9-1-61-69


ABSTRACT

Aim. To evaluate the potential of a new cytogenetic technique in patients with primary myelofibrosis (PMF).

Materials and methods. 48-hour blood cell cultures (according to Singh et al., 2013) were used for cytogenetic study in 11 PMF patients (5 female, 6 men, aged 32–60 years; median 48.6 years). GTG-banding and different types of fluorescence in situ hybridization (FISH) techniques were used for identification of chromosomal aberrations.

Results. The incidence of abnormal karyotypes in blood cultures was significantly higher than that in standard bone marrow cultures (82 vs 27 %; < 0.01). The polyploid clones were found in blood cultures of 45 % of patients. Structural chromosomal aberrations were found in chromosomes 6, 1, 3, as well as 16 and 17 (in 2 and 1 patients with each aberration, respectively). In all but one patients these abnormalities in diploid and polyploid metaphases were identical. Partial 1q trisomy resulted from adding of additional (1q21–1q44) material translocated to the short arm of chromosome 5 to the material of 2 normal homologue of chromosome 1. It seems that 1q+, i(17q) and some others chromosomal abnormalities were secondary, whereas 6p21 locus involvement may be a primary defect in PMF. The t(3;6)(q25;p21) translocation described for the first time and confirmed by FISH should be considered a variant of well-known translocation t(1;6). Allo-HSCT in 2 patients with 1q+ was successful, whereas there were problems with engraftment in a female patient with prognostically unfavorable t(3;3)(q21;q26) translocation associated with the EVI1 gene overexpression.

Conclusion. Cytogenetic examinations in blood cultures provide important additional information about PMF patients.


Keywords: primary myelofibrosis, 48-hour peripheral blood cell culture, cytogenetics.

Received: July 14, 2015

Accepted: November 1, 2015

Read in PDF (RUS) pdficon


REFERENCES

  1. Heuck G. Zwei Falle von Leukamie mit eigenthumlichem Blut-resp Knochenmarkbefund. Archiv fur pathologische Anatomie und Physiologie und fur klinische Medicin. 1879;78(3):475–96. doi: 10.1007/bf01878089.
  2. Dameshek W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6(4):372–5.
  3. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369:2379–90. doi: 10.1056/nejmoa1311347.
  4. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia. 2014;28:1472–7. doi: 10.1038/leu.2014.3.
  5. Panagiota V, Thol F, Markus B, et al. Prognostic effect of calreticulin mutations in patients with myelofibrosis after allogeneic hematopoetic stem cell transplantation. Leukemia. 2014;28:1552–5. doi: 10.1038/leu.2014.66.
  6. Komrokji RS, Seymour JF, Roberts AW, et al. Results of a phase 2 study of pacritinib (SB1518), a JAK2/JAK2 (V617F) inhibitor, in patients with myelofibrosis. Blood. 2015;125(17):2649–55. doi: 10.1182/blood-2013-02-484832.
  7. Hoffman R, Rondalli D. Biology and treatment of primary myelofibrosis. Hematology (Am Soc Hematol Educ Program). 2007:346–54. doi: 10.1182/asheducation-2007.1.346.
  8. Geraedts JP, den Ottolander GJ, Ploem JE, Muninghe OG. An identical translocation between chromosome 1 and 7 in three patients with myelofibrosis and myeloid metaplasia. Br J Haematol. 1980;44:569–75. doi: 10.1111/j.1365-2141.1980.tb08711.x.
  9. Bernstein R, Pinto MR, Behr A, Mendelow B. Chromosome 3 abnormalities in acute non-lymphocytic leukemia (ANLL) with abnormal thrombopoiesis: report of three patients with a “new” inversion anomaly and a further case of homologous translocation. Blood. 1982;60:613–7.
  10. Clare N, Elson D, Mannhof L. Cytogenetic studies of peripheral myeloblasts and bone marrow fibroblasts in acute myelofibrosis. Am J Clin Pathol. 1982;77:762–6. doi: 10.1093/ajcp/77.6.762.
  11. Borgstrom GM, Knutila S, Ruutu T, et al. Abnormalities of chromosome 13 in myelofibrosis. Scand J Haematol. 1984;33:15–21.
  12. Miller JB, Testa JR, Lindgren V, Rowley JD. The pattern and clinical significance of karyotypic abnormalities in patients with idiopathic and postpolycythemic myelofibrosis. Cancer. 1985;55:582–91. doi: 10.1002/1097-0142(19850201)55:3<582::aid-cncr2820550318>3.0.co;2-o.
  13. Castoldi G, Cuneo A, Tomasi P, Ferrari L. Chromosome abnormalities in myelofibrosis. Acta Haematol. 1987;78(Suppl. 1):104–8. doi: 10.1159/000205913.
  14. Smadja N, Krulik M, de Gramount A, et al. Cytogenetic studies in twelve patients with primary myelofibrosis and myeloid metaplasia. Cancer Genet Cytogenet. 1987;24:151–8. doi: 10.1016/0165-4608(87)90092-6.
  15. Demory JL, Dupriez B, Fenaux P, et al. Cytogenetic studies and their prognostic significance in agnogenic myeloid metaplasia: a report of 47 cases. Blood. 1988;72:855–9.
  16. Djordjevic V, Dencic-Fekete M, Jovanovic J, et al. Cytogenetics of agnogenic myeloid metaplasia: a study of 61 patients. Cancer Genet Cytogenet. 2007;173:57–62. doi: 10.1016/j.cancergencyto.2006.09.02.
  17. Hidaka K, Shide K, Shimoda H, et al. The impact of cytogenetic abnormalities on the prognosis of primary myelofibrosis: a prospective survey of 202 cases in Japan. Eur J Haematol. 2009;83:328–33. doi: 10.1111/j.1600-0609.2009.01298.x.
  18. Hussein K, Van Dyke DL, Tefferi A. Conventional cytogenetics in myelofibrosis: literature review and discussion. Am J Hematol. 2009;82:329–38. doi: 10.1111/j.1600-0609.2009.01224.x.
  19. Caramazza D, Begna KH, Gangar N, et al. Refined cytogenetic-risk categorization for overall and leukemia-free survival in primary myelofibrosis: a single center study of 433 patients. Leukemia. 2011;25:82–8. doi: 10.1038/leu.2010.234.
  20. Tefferi A, Lasho TL, Jimma Th, et al. One thousand patients with primary myelofibrosis: The Mayo clinic experience. Mayo Clin Proc. 2012;87(1):25–33. doi: 10.1016/j.mayocp.2011.11.001.
  21. Wassie E, Finke Ch., Gangat N, et al. A compendium of cytogenetic abnormalities in myelofibrosis: molecular and phenotypic correlates in 826 patients. Br J Haematol. 2014; doi: 10.1111/bjh.132. doi: 10.1111/bjh.13260.
  22. Li B, Xu J, Li Ch, et al. Cytogenetic studies and their prognostic contribution in 565 Chinese patients with primary fibrosis. Am J Hematol. 2014;89(11):1043–7. doi: 10.1002/ajh.23824.
  23. Абдулкадыров К.М., Шуваев В.А., Мартынкевич И.С. Первичный миелофиброз: собственный опыт и новое в диагностике и лечении. Онкогематология. 2015;10(2):26–36.
    [Abdulkadyrov KM, Shuvaev VA, Martynkevich IS. Primary myelofibrosis: own experience and news from diagnostic and treatment. Onkogematologiya. 2015;10(2):26–36. (In Russ)]
  24. Singh NR, Morris ChM, Koleth M, et al. Polyploidy in myelofibrosis: analysis by cytogenetic and SNP indicates association with advancing disease. Mol Cytogenet. 2013;6:59–71. doi: 10.1186/1755-8166-6-59.
  25. Devine SM, Hoffman R, Verma A, et al. Allogeneic blood cell transplantation following reduced-intensity conditioning is effective therapy for older patients with myelofibrosis with myeloid metaplasia. Blood. 2002;99:2255–8. doi: 10.1182/blood.v99.6.2255.
  26. Kroger N, Zabelina T, Schieder H, et al. Pilot study of reduced-intensity conditioning followed by allogeneic stem cell transplantation from related and unrelated donors in patients with myelofibrosis. Br J Haematol. 2005;128:690–7. doi: 10.1111/j.1365-2141.2005.05373.x.
  27. Merup M, Lazarevic V, Nahi H, et al. Different outcome of allogeneic transplantation in myelofibrosis using conventional or reduced-intensity conditioning regimens. Br J Haematol. 2006;135:367–73. doi: 10.1111/j.1365-2141.2006.06302.x.
  28. 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;114:5264–70. doi: 10.1182/blood-2009-07-234880.
  29. Schaffer LG, McGovan-Jordan J, Schmid M. ISCN. An international System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013.
  30. Tefferi A, Dingli D, Li C, Dewald GW. Prognostic diversity among cytogenetic abnormalities in myelofibrosis with myeloid metaplasia. Cancer. 2005;104:1656–60. doi: 10.1002/cncr.21358.
  31. Strasser-Weiple K, Steurer M, Kees M, et al. Chromosome 7 deletions are associated with unfavorable prognosis in myelofibrosis and myeloid metaplasia. Blood. 2005;105(10):4146. doi: 10.1182/blood-2004-11-4319.
  32. Hussein K, Huang J, Lasho T, et al. Karyotype complements the International Prognostic Scoring System for primary myelofibrosis. Eur J Haematol. 2009;82:256–9. doi: 10.1111/j.1600-0609.2009.01216.x.
  33. Hussein K, Pardanani AD, Van Dyke DL, et al. International Prognostic Scoring System-independent cytogenetic risk categorization in primary myelofibrosis. Blood. 2010;115:496–9. doi: 10.1182/blood-2009-08-240135.
  34. 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.
  35. Vaidya R, Caramazza D, Begna KH, et al. Monosomal karyotype in primary myelofibrosis is detrimental to both overall and leukemia-free survival. Blood. 2011;117(21):5612–5. doi: 10.1182/blood-2010-11-320002.
  36. Huret JL. Atlas of Genetics and Cytogenetics in Oncology and Haematology. Poitiers: University Hospital; 2011. p. 5. doi: 10.4267/2042/44590.

Chronic Myeloid Leukemia: Long-Term Experience of Target Therapy

KM Abdulkadyrov, VA Shuvaev, IS Martynkevich, MS Fominykh, NA Potikhonova, II Zotova, VYu Udal’eva, RA Golovchenko, NV Shakhvorostova, DI Shikhbabaeva, MN Zenina, SA Tiranova, SA Kudryashova, LS Martynenko, MP Ivanova, NYu Tsybakova, EV Petrova, LB Polushkina, EV Kleina

Russian Scientific Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

For correspondence: Vasilii Anatol’evich Shuvaev, PhD, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(921)636-54-72; e-mail: shuvaev77@mail.ru

For citation: Abdulkadyrov KM, Shuvaev VA, Martynkevich IS, et al. Chronic Myeloid Leukemia: Long-Term Experience of Target Therapy. Clinical oncohematology. 2016;9(1):54–60 (In Russ).

DOI: 10.21320/2500-2139-2016-9-1-54-60


ABSTRACT

Background & Aims. Interpretation of key aspects of pathogenesis of chronic myeloid leukemia (CML) and development and introduction of target therapy have changed the prognosis of this once fatal disease dramatically. Results of numerous clinical trials demonstrated substantial superiority of tyrosine kinase inhibitors over previous therapy techniques. At the same time, clinical trials had limitations in patient enrollment, as well as treatment conditions and duration. The analysis of our clinical experience in CML target therapy (over the period from 2003 till 2015) is an important argument for introduction of novel drugs into routine clinical practice. The aim of the study is to analyze our own experience in CML target therapy and to compare our results with clinical trials data.

Methods. Outpatient’s cards and case histories of CML patients treated in the Russian Scientific Research Institute of Hematology and Transfusiology over last 12 years were analyzed in this work. Published results of multi-center clinical trials evaluating the use of tyrosine kinase inhibitors in CML were used for a comparative analysis. The primary morbidity rate and the prevalence of CML, results of first and subsequent treatment lines were studied with assessment of survival rates, adverse events, and the nature of the response (hematologic, cytogenetic and molecular).

Results. The experience in treatment of 208 CML patients was analyzed. The use of imatinib led to clinical and hematological remission (complete hematologic response) was achieved in 95 % of patients. The frequency of complete cytogenetic responses (CCyR) was 69 %, and that of major molecular responses (MMR) was 58 %. The overall 5-year survival (OS) was 86.4 %, the 10-years OS was 67.5 %. The use of nilotinib during the second line permitted to achieve CCyR in 61 % of patients, and the MMR in 55 % of cases. The two-year OS was 96 % and the 5-year OS was 68 %. CCyR and MMR were achieved in 50 % patients treated with dasatinib during the second line. As for the third line, CCyR was achieved in 50 % of patients and MMR in 25 %. In case of previous imatinib and nilotinib resistance, CCyR was observed only in 36 % of patients and MMR in 18 % of cases. During second-line dasatinib treatment, the 2-year OS was 85 %, and the 5-year OS was 51 %; as for the third line, the results were 75 % and 50 %, respectively. The range and rates of adverse events of the therapy, in general, corresponded to results of clinical trials.

Conclusion. The use of tyrosine kinase inhibitors in treatment of CML permits to prolong patient’s life span and quality of life significantly. The use of nilotinib and dazatinib (in case of nilotinib intolerance and/or resistance) could be effective in most patients.


Keywords: chronic myeloid leukemia, target therapy, tyrosine kinase inhibitors, imatinib, nilotinib, dasatinib, clinical practice.

Received: September 10, 2015

Accepted: October 20, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Salesse S, Verfaillie CM. BCR/ABL: from molecular mechanisms of leukemia induction to treatment of chronic myelogenous leukemia. Oncogene. 2002;21:8547–59. doi: 10.1038/sj.onc.1206082.
  2. Goldman JM, Melo JV. Targeting the BCR-ABL Tyrosine Kinase in Chronic Myeloid Leukemia. N Engl J Med. 2001;344(14):1084–6. doi: 10.1056/ nejm200104053441409.
  3. Абдулкадыров К.М., Абдуллаев А.О., Авдеева Л.Б. и др. Федеральные клинические рекомендации по диагностике и терапии хронического мие- лолейкоза. Вестник гематологии. 2013;9(3):4–40. [Abdulkadyrov KM, Abdullaev AO, Avdeeva LB, et al. Federal clinical recommendations for diagnosis and treatment of chronic myeloid leukemia. Vestnik gematologii. 2013;9(3):4–40. (In Russ)]
  4. 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.
  5. NCCN Guidelines Chronic Myelogenous Leukemia. Version 1.2015. Available from: www.nccn.org/cml.pdf. (accessed 14.05.2015).
  6. Шуваев В.А., Абдулкадырова А.С., Мартынкевич И.С. и др. Опыт лечения хронического миелолейкоза в Санкт-Петербурге. Вестник гема- тологии. 2011;7(1):43. [Shuvaev VA, Abdulkadyrova AS, Martynkevich IS, et al. Experience in treatment of chronic myeloid leukemia in Saint-Petersburg. Vestnik gematologii. 2011;7(1):43. (In Russ)]
  7. Стахина О.В., Туркина А.Г., Гусарова Г.А. и др. Отдаленные резуль- таты выживаемости больных в поздней хронической фазе Ph+ хрониче- ского миелолейкоза при лечении иматиниб мезилатом (Гливек®). Вестник гематологии. 2009;5(2):42. [Stakhina OV, Turkina AG, Gusarova GA, et al. Long-term results of survival rates of patients in late chronic phase of Ph+ chronic myeloid leukemia treated with imatinib mesylate (Glivec®). Vestnik gematologii. 2009;5(2):42. (In Russ)]
  8. Cortes J, Rousselot P, Kim D-W, et al. Dasatinib induces complete hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in blast crisis. Blood. 2007;109(8):3207–13. doi: 10.1182/blood-2006-09-046888.
  9. Cortes J, Saglio G, Baccarani M, et al. Final Study Results of the Phase 3 Dasatinib Versus Imatinib in Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase (CML-CP) Trial (DASISION, CA180-056). 56th Annual Meeting and Exposition, San Francisco, CA December 6–9, 2014. Blood. 2014;21: Abstract 152.
  10. Deininger M, O’Brien SG, Guilhot F, et al. International Randomized Study of Interferon Vs STI571 (IRIS) 8-Year Follow up: Sustained Survival and Low Risk for Progression or Events in Patients with Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase (CML-CP) Treated with Imatinib. Blood (ASH Annual Meeting Abstracts). 2009;114(22): Abstract 1126.
  11. Giles FJ, Rosti G, Beris P, et al. Nilotinib is superior to imatinib as first-line therapy of chronic myeloid leukemia: the ENESTnd study. Expert Rev Hematol. 2010;3(6):665–73. doi: 10.1586/ehm.10.61.
  12. Hochhaus A, Shah NP, Cortes JE. Dasatinib versus imatinib (IM) in newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP): DASISION 3-year follow-up. Program and abstracts of the 2012 Annual Meeting of the American Society of Clinical Oncology, Chicago, Illinois, June 1–5, 2012. Abstract 6504.
  13. Hoglund M, Sandin F, Simonsson B. Epidemiology of chronic myeloid leukaemia: an update. Ann Hematol. 2015;94(2):241–7. doi: 10.1007/s00277- 015-2314-2.
  14. Kantarjian HM, Shah NP, Cortes JE, et al. Dasatinib or imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: 2-year follow-up from a randomized phase 3 trial (DASISION). Blood. 2012;119(5):1123–9. doi: 10.1182/ blood-2011-08-376087.
  15. Powell BL, Khoury HJ, Lipton JH, et al. Nilotinib Responses and Tolerability Confirmed in North American Patients with Chronic Myeloid Leukemia (CML) From ENACT (Expanding Nilotinib Access in Clinical Trials). Blood. 2009;114(22):3295.
  16. Saglio G, LeCoutre PD, Pasquini R, et al. Nilotinib Versus Imatinib in Patients (pts) with Newly Diagnosed Philadelphia Chromosome-Positive (Ph+) Chronic Myeloid Leukemia in Chronic Phase (CML-CP): ENESTnd 36-Month (mo) Follow-up. Blood. 2011;118(21):452.
  17. Куликов С.М., Виноградова О.Ю., Челышева Е.Ю. и др. Заболе- ваемость хроническим миелолейкозом в 6 регионах России по данным популяционного исследования 2009–2012 гг. Терапевтический архив. 2014;7:24–30. [Kulikov SM, Vinogradova OYu, Tchelysheva EYu, et al. Incidence of chronic myeloid leukemia in 6 regions of Russia, according to a population-based study over the period from 2009 to 2012. Terapevticheskii arkhiv. 2014;7:24–30. (In Russ)]
  18. Лазарева О.В., Туркина А.Г., Гусарова Г.А. и др. Итоги 12-летней те- рапии ингибиторами тирозинкиназ больных в поздней хронической фазе хронического миелолейкоза после неудачи лечения ИФН-α. Сибирский научный медицинский журнал. Бюллетень СО РАМН. 2015;35(1):90–7. [Lazareva OV, Turkina AG, Gusarova GA, et al. Results of 12-year therapy with tyrosine kinase inhibitors in patients with late chronic phase of chronic myeloid leukemia after IFN-a treatment failure. Sibirkii meditcinskiy jurnal. Bulleten’ SO RAMN. 2015;35(1):90–7. (In Russ)]
  19. Zdenek R, Belohlavkova P, Cetkovsky P, et al. Comparison of Glucose and Lipid Metabolism Abnormality during Nilotinib, Imatinib and Dasatinib Therapy – Results of Enigma 2 Study. Blood. 2014;124(21):1813.
  20. Nicolini FE, Turkina A, Shen Z-X, et al. Expanding Nilotinib Access in Clinical Trials (ENACT). Cancer. 2012;118(1):118–26. doi: 10.1002/cncr.26249.
  21. Hughes TP, le Coutre PD, Jootar S, et al. ENESTnd 5-year follow-up: continued benefit of frontline nilotinib (NIL) compared with imatinib (IM) in patients (pts) with chronic myeloid leukemia in chronic phase (CML-CP). Haematologica. 2014;99(Suppl 1):236–7.
  22. Shah NP, Kantarjian HM, Kim D-W, et al. Intermittent Target Inhibition With Dasatinib 100 mg Once Daily Preserves Efficacy and Improves Tolerability in Imatinib-Resistant and -Intolerant Chronic-Phase Chronic Myeloid Leukemia. J Clin Oncol. 2008;26(19):3204–12. doi: 10.1200/jco.2007.14.9260.
  23. Jabbour E, Makenbaeva D, Lingohr-Smith M, et al. Evaluation of Comorbidities Relevant to Tyrosine Kinase Inhibitor Treatment Among Patients with Chronic Myelogenous Leukemia in the US. Managed Care Setting. Blood. 2014;124(21):4550.

Low Dose Cytarabine and Cladribine for Treatment of Relapsed or Refractory Acute Myeloid Leukemia: Clinical Experience

SV Gritsaev, II Kostroma, AA Kuzyaeva, IM Zapreeva, EV Litvinskaya, LV Stelmashenko, SA Tiranova, IS Martynkevich, NA Potikhonova, KM Abdulkadyrov

Russian Scientific Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

For correspondence: Sergei Vasil’evich Gritsaev, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)717-54-68; e-mail: gritsaevsv@mail.ru

For citation: Gritsaev SV, Kostroma II, Kuzyaeva AA, et al. Low Dose Cytarabine and Cladribine for Treatment of Relapsed or Refractory Acute Myeloid Leukemia: Clinical Experience. Clinical oncohematology. 2016;9(1):48–53 (In Russ).

DOI: 10.21320/2500-2139-2016-9-1-48-53


ABSTRACT                                      

Aim. The aim of this paper is to evaluate the effectiveness of low dose cytarabine (Ara-C) combined with cladribine for the treatment of relapsed or refractory acute myeloid leukemia (AML) and to determine clinical and lab factors associated with response to the therapy.

Methods. Data of 10 patients aged 26–58 years (median 48 years) were analyzed. The diagnoses were de novo AML (7 patients), secondary AML (sAML) (2 patients) and refractory anemia with excess of blasts (RAEB-2) (1 patient). Four patients had primary refractory AML. Relapse was diagnosed in 3 patients. The induction scheme 7+3 was ineffective in patient with RAEB-2. There was no response to any kind of therapy in sAML patients. The treatment scheme under trial consisted of Ara-C 10–15 mg/m2 subcutaneously twice a day for 1–14 days and cladribine 5 mg/m2 intravenously once a day for 1–5 days. The course was repeated in case of at least two-fold decrease in bone marrow blasts level in a punctate versus baseline. Medical examination and maintenance therapy were performed in accordance with protocols approved by the clinic.

Results. According to the protocol, the patients received 1–2 courses. Response was achieved in 5 patients: 2 patients achieved complete response (CR) and 3 achieved partial response (PR). The most common complication was hematologic toxicity. All patients received transfusions of blood components. No lethal outcomes were observed within 8 weeks. The duration of the response was 2 to 3 months. During this period of time, allogeneic stem cell transplantation was performed in 2 patients with CR; however, in one patient, the conditioning regimen began at the same time with the increase in blast cell count in the bone marrow. The search for unrelated donors of hematopoietic stem cells for 2 patients with CR was begun. The distinct features of all patients with CR and PR were the following factors: de novo AML, absence of FLT3 or c-KIT mutations and the course duration was not less than 10 days.

Conclusion. Low dose Ara-C in combination with cladribine may be considered a treatment option for some patients with relapsed or refractory de novo AML.


Keywords: acute myeloid leukemia, relapse, refractory, chemotherapy, low dose cytarabine, cladribine.

Received: June 4, 2015

Accepted: October 8, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Weick JK, Kopecky KJ, Appelbaum FR, et al. A randomized investigation of high dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: a Southwest Oncology Group study. Blood. 1996;88(8):2841–51.
  2. Tallman MS, Gilliland G, Rowe JM. Drug therapy for acute myeloid leukemia. Blood. 2005;106(4):1154–63. doi: 10.1182/blood-2005-01-0178.
  3. Estey EH. Acute myeloid leukemia: 2014 update on risk-stratification and management. Am J Hematol. 2014;89(11):1063–81. doi: 10.1002/ajh.23834.
  4. Yanada M, Garcia-Manero G, Borthakur G, et al. Potential cure of acute myeloid leukemia. Cancer. 2007;110(12):2756–60. doi: 10.1002/cncr.23112.
  5. Mangan JK, Luger SM. Salvage therapy for relapsed or refractory acute myeloid leukemia. Ther Adv Hematol. 2011;2(2):73–82. doi: 10.1177/2040620711402533.
  6. Савченко В.Г., Паровичникова Е.Н., Афанасьев Б.В. и др. Национальные клинические рекомендации по диагностике лечению острых миелоидных лейкозов взрослых. Гематология и трансфузиология 2014;59(1):3–29.
    [Savchenko VG, Parovichnikova EN, Afanas’ev BV, et al. National clinical recommendations for diagnosis and treatment of acute myeloid leukemias in adults. Gematologiya i transfuziologiya. 2014;59(1):3–29. (In Russ)]
  7. Грицаев С.В., Мартынкевич И.С., Зюзгин И.С. и др. Гетерогенность острого миелоидного лейкоза с транслокацией t(8;21)(q22;q22). Терапевтический архив. 2014;86(7):45–52.
    [Gritsaev SV, Martynkevich IS, Zyuzgin IS, et al. Heterogenicity of acute myeloid leukemia with t(8;21)(q22;q22) translocation. Terapevticheskii arkhiv. 2014;86(7):45–52. (In Russ)]
  8. Milligan DW, Grimwade D, Cullis JO, et al. Guidelines on the management of acute myeloid leukemia in adults. Br J Haematol 2006; 135(4): 450–74. doi: 10.1111/j.1365-2141.2006.06314.x.
  9. Fey MF, Buske C. Acute myeloblastic leukemias in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013;24(Suppl. 6):138–43. doi: 10.1093/annonc/mdt320.
  10. Parker WB, Bapat AR, Shen JX, et al. Interaction of 2-halogenated dATP analogs (F, Cl, and Br) with human DNA polymerases, DNA primase, and ribonucleotide reductase. Mol Pharmacol. 1988;34(4):485–91.
  11. Hirota Y, Yoshioka A, Tanaka S, et al. Imbalance of deoxyribonucleoside triphosphates, DNA double-strand breaks, and cell death caused by 2-chlorodeoxyadenosine in mouse FM3A cells. Cancer Res. 1989;49(4):915–9.
  12. Wrzesien-Kus A, Robak T, Lech-Maranda E, et al. A multicenter, open, non-comparative, phase II study of the combination of cladribine, cytarabine, and G-CSF and induction therapy in refractory acute myeloid leukemia – a report of the Polish Adult Leukemia Group (PALG). Eur J Hematol. 2003;71(3):155–62. doi: 10.1034/j.1600-0609.2003.00122.x.
  13. Price SL, Lancet JE, George TJ, et al. Salvage chemotherapy regimens for acute myeloid leukemia: Is one better? Efficacy comparison between CLAG and MEC regimens. Leuk Res. 2011;35(3):301–4. doi: 10.1016/j.leukres.2010.09.002.
  14. Kern W, Schleyer E, Braess J, et al. Efficacy of fludarabine, intermittent sequential high-dose cytosine arabinoside, and mitoxantrone (FIS-HAM) salvage therapy in highly resistant acute leukemias. Ann Hematol. 2001;80(6):334–9. doi: 10.1007/s002770100293.
  15. Hashmi KU, Khan B, Ahmed P, et al. FLAG-IDA in the treatment of refractory/relapsed acute leukaemias: single centre study. J Pak Med Assoc. 2005;55(6):234–8.
  16. Pastore D, Specchia G, Carluccio P, et al. FLAG-IDA in the treatment of refractory/relapsed acute myeloid leukemia: single-center experience. Ann Hematol. 2003;82(4):231–5.
  17. Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol. 2003;21(24):4642–9. doi: 10.1200/jco.2003.04.036.
  18. Breems DA, van Putten WLJ, Huijgens PC, et al. Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol. 2005;23(9):1969–78. doi: 10.1200/jco.2005.06.027.
  19. Breems DA, van Putten WL, de Greef GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol. 2008;26(29):4791–7. doi: 10.1200/jco.2008.16.0259.
  20. Armistead PM, de Lima M, Pierce S, et al. Quantifying the survival benefit for allogeneic stem cell transplantation in relapsed acute myeloid leukemia. Biol Blood Marrow Transplant. 2009;15(11):1431–8. doi: 10.1016/j.bbmt.2009.07.008.
  21. Kurosawa S, Yamaguchi T, Miyawaki S, et al. Prognostic factors and outcomes of adult patients with acute myeloid leukemia after first relapse. Haematologica. 2010;95(11):1857–64. doi: 10.3324/haematol.2010.027516.
  22. Mangan JK, Luger SM. Salvage therapy for relapsed or refractory acute myeloid leukemia. Ther Adv Hematol. 2011;2(2):73–82. doi: 10.1177/2040620711402533.
  23. Freyer CW, Gupta N, Wetzler M, Wang ES. Revisiting the role of cladribine in acute myeloid leukemia: an improvement on past accomplishments or more old news? Am J Hematol. 2015;90(1):62–72. doi: 10.1002/ajh.23862.
  24. Zhang WG, Wang FX, Chen YX, et al. Combination chemotherapy with low-dose cytarabine, homoharringtonine, and granulocyte colony-stimulating factor priming in patients with relapsed or refractory acute myeloid leukemia. Am J Hematol. 2008;83(3):185–8. doi: 10.1002/ajh.20903.
  25. Liu L, Zhang Y, Jin Z, et al. Increasing the dose of aclarubicin in low-dose cytarabine and aclarubicin in combination with granulocyte colony-stimulating factor (CAG regimen) can safely and effectively treat relapsed or refractory acute myeloid leukemia. Int J Hematol. 2014;99(5):603–8. doi: 10.1007/s12185-014-1528-8.
  26. Assouline S, Culjkovic-Kraljacic B, Bergeron J, et al. A phase I trial of ribavirin and low-dose cytarabine for the treatment of relapsed and refractory acute myeloid leukemia with elevated eIF4E. Haematologica. 2015;100(1):e7–9. doi: 10.3324/haematol.2014.111245.

Hematological Improvement is a Favorable Response to Azacitidine in Patients with Acute Myeloid Leukemias and Myelodysplastic Syndromes

I.I. Kostroma1, S.V. Gritsaev1, E.V. Karyagina2, A.S. Nizamutdinova3, I.S. Martynkevich1, K.M. Abdulkadyrov1

1 Russian Scientific Research Institute of Hematology and Transfusiology under the Federal Medico-Biological Agency, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

2 Municipal Hospital No. 15, 4 Avangardnaya str., Saint Petersburg, Russian Federation, 198205

3 Alexandrovskaya Municipal Hospital No. 17, 4 pr-t Solidarnosti, Saint Petersburg, Russian Federation, 193312

For correspondence: Ivan Ivanovich Kostroma, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)717-58-57; e-mail: obex@rambler.ru

For citation: Kostroma II, Gritsaev SV, Karyagina EV, et al. Hematological Improvement is a Favorable Response to Azacitidine in Patients with Acute Myeloid Leukemias and Myelodysplastic Syndromes. Clinical oncohematology. 2015;8(4):413–419 (In Russ).

DOI: 10.21320/2500-2139-2015-8-4-413-419


ABSTRACT

Aim. To evaluate types of response to azacitidine associated with improvement of overall survival (OS) rates of patients with acute myeloid leukemias (AML) and myelodysplastic syndromes (MDS).

Methods. A retrospective analyses of medical records of 14 AML patients and 13 MDS patients at the age of 39 to 84 treated with azacitidine at a dose of 75 mg/m2 subcutaneously for 7 subsequent days every 28 days was performed. The therapy effectiveness was evaluated according to modified 2006 IWG criteria. The OS was calculated beginning with the date of initiation of the azacitidine therapy.

Results. From 2 to 25 azacitidine cycles was performed. Complete remission (CR) was achieved in 6 patients (22.2 %) including 4 AML and 2 MDS patients. Bone marrow remission (mCR) was diagnosed in 1 MDS patient (3.7 %). Hematological improvement was obtained in 11 patients (40.7 %) including 5 AML and 6 MDS patients. The overall response was 66.7 % (18 to 27 patients). There was no correlation between the therapy effectiveness and patients’ age, disease type, duration of the previous period, baseline hemoglobin, leukocytes, and platelets levels, and dependence on transfusions of erythrocyte suspension and thromboconcentrate. The therapy was considered ineffective in 9 patients (33.3 %). Stabilization with retained requirements of blood component transfusion was observed in 4 AML and 3 MDS patients. 2 patients presented gradual increase of the blast cell count in the bone marrow. The follow-up period was 2–29 months. The median OS of all patients was 11.5 months. The median OS of patients with CR, mCR and hematological improvement was significantly greater than that in the group of patients with stable disease and progression: 15.9 versus 7.4 months, respectively (= 0,010).

Conclusion. Reduction of transfusion requirement and/or stable improvement of peripheral blood levels due to azacitidine administration are associated with improved OS rates of AML and MDS patients.


Keywords: acute myeloid leukemia, myelodysplastic syndromes, azacitidine, hematological improvement, overall survival.

Received: April 6, 2015

Accepted: October 22, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Грицаев С.В. Миелодиспластические синдромы. В кн.: Гематология. Национальное руководство по гематологии. Под ред. О.А. Рукавицина. М: ГЭОТАР-Медиа, 2015. С. 300–33.
    [Gritsaev SV. Myelodysplastic syndromes. In: Rukavitsin OA, ed. Gematologiya. Natsional’noe rukovodstvo po gematologii. (Hematology. National guidelines in hematology.) Moscow: GEOTAR-Media Publ.; 2015. p. 300–33. (In Russ)]
  2. Грицаев С.В., Мартынкевич И.С., Абдулкадыров К.М. и др. Возрастные особенности кариотипа больных острым миелоидным лейкозом. Терапевтический архив. 2011;1:51–5.
    [Gritsaev SV, Martynkevich IS, Abdulkadyrov KM, et al. Age-related features of karyotype of patients with acute myeloid leukemias. Terapevticheskii arkhiv. 2011;1:51–5. (In Russ)]
  3. Грицаев С.В., Мартынкевич И.С., Абдулкадыров К.М. и др. Комплексный кариотип — маркер крайне неблагоприятного прогноза у больных острыми миелоидными лейкозами и развернутыми вариантами миелодиспластического синдрома старше 70 лет с высоким индексом коморбидности. Терапевтический архив. 2012;7:16–21.
    [Gritsaev SV, Martynkevich IS, Abdulkadyrov KM, et al. Complex karyotype is a marker for extremely unfavorable prognosis in patients with acute myeloid leukemia and marked myelodysplastic syndrome in patients over 70 years old with high co-morbidity index. Terapevticheskii arkhiv. 2012;7:16–21. (In Russ)]
  4. Грицаев С.В., Мартынкевич И.С., Запреева И.М. и др. Эффективность первого и повторного курсов индукционной терапии больных de novo острым миелоидным лейкозом. Бюллетень СО АМН 2013;33(1):67–75.
    [Gritsaev SV, Martynkevich IS, Zapreeva IM, et al. Efficacy of the first and repeated courses of induced therapy of patients with de novo acute myeloid leukemia. Byulleten’ SO AMN. 2013;33(1):67–75. (In Russ)]
  5. Goldstone AH, Burnett AK, Wheatley K, et al. Attempts to improve treatment outcomes in acute myeloid leukemia in older patients: the results of the United Kingdom Medical Research Council AML11 trial. Blood 2001;98(5):1302–11. doi: 10.1182/blood.v98.5.1302.
  6. Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 2011;29(5):487–94. doi: 10.1200/jco.2010.30.1820.
  7. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109(6):1114–24. doi: 10.1002/cncr.22496.
  8. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10(3):223–32. doi: 10.1016/s1470-2045(09)70003-8.
  9. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28(4):562–9. doi: 10.1200/jco.2009.23.8329.
  10. Quintas-Cardama A, Ravandi F, Liu-Dumlao T, et al. Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia. Blood. 2012;120(24):4840–5. doi: 10.1182/blood-2012-06-436055.
  11. De Padua Silva L, de Lima M, Kantarjian H, et al. Feasibility of allo-SCT after hypomethylating therapy with decitabine for myelodysplastic syndrome. Bone Marrow Transplant. 2009;43(11):839–43. doi: 10.1038/bmt.2008.400.
  12. 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.
  13. Lubbert M, Bertz H, Muller MJ, Finke J. When azanucleoside treatment can be curative: nonintensive bridging strategy before allografting in older patients with myelodysplastic syndrome/acute myeloid leukemia. J Clin Oncol. 2013;31(6):822–3. doi: 10.1200/jco.2012.46.4222.
  14. Cheson BD, Greenberg PL, Bennett JM, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108(2):419–25. doi: 10.1182/blood-2005-10-4149.
  15. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World health Organisation (WHO) classification of myeloid neoplasms and acute myeloid leukemia: rationale and important changes. Blood. 2008;114(5):937–51. doi: 10.1182/blood-2009-03-209262.
  16. Грицаев С.В., Мартынкевич И.С., Кострома И.И. Азацитидин при остром миелобластном лейкозе и миелодиспластическим синдроме. Гематология и трансфузиология. 2012;1:23–9.
    [Gritsaev SV, Martynkevich IS, Kostroma II. Azacitidine in acute myeloblast leukemia and myelodysplactic syndrome. Gematologiya i transfuziologiya. 2012;1:23–9. (In Russ)]
  17. Smith BD, Beach CL, Mahmoud D, et al. Survival and hospitalization among patients with acute myeloid leukemia treated with azacitidine or decitabine in a large managed care population: a real-world, retrospective, claims-based, comparative analysis. Exp Hematol Oncol. 2014;3(1):1–6. doi: 10.1186/2162-3619-3-10.
  18. Gurion R, Vidal L, Gafter-Gvili A, et al. 5-Azacitidine prolongs overall survival in patients with myelodysplastic syndrome – a systematic review and meta-analysis. Haematologica. 2010;95(2):303–10. doi: 10.3324/haematol.2009.010611.
  19. Saunthararajah Y. Key clinical observations after 5-azacytidine and decitabine treatment of myelodysplastic syndromes suggest practical solutions for better outcomes. Hematol Am Soc Hematol Educ Program. 2013:511–21. doi: 10.1182/asheducation-2013.1.511.
  20. Pleyer L, Burgstaller S, Girschikofsky M, et al. Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-study Group. Ann Hematol. 2014;93(11):1825–38. doi: 10.1007/s00277-014-2126-9.
  21. Ramos F, Thepot S, Pleyer L, et al. Azacitidine frontline therapy for unfit acute myeloid leukemia patients: clinical use and outcome prediction. Leuk Res. 2015;39(3):296–306. doi: 10.1016/j.leukres.2014.12.013.
  22. Abaigar M, Ramos F, Benito R, et al. Prognostic impact of the number of methylated genes in myelodysplastic syndromes and acute myeloid leukemias treated with azacytidine. Ann Hematol. 2013;92(11):1543–52. doi: 10.1007/s00277-013-1799-9.
  23. Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705–12. doi: 10.1182/blood-2014-06-582809.
  24. Hwang KL, Song MK, Shin HJ, et al. Monosomal and complex karyotypes as prognostic parameters in patients with International Prognostic Scoring System higher risk myelodysplastic syndrome treated with azacitidine. Blood Res. 2014;49(4):234–40. doi: 10.5045/br.2014.49.4.234.
  25. Xicoy B, Jimenez MJ, Garcia O, et al. Results of treatment with azacitidine in patients aged ³75 years included in the Spanish Registry of Myelodysplastic Syndromes. Leuk Lymphoma. 2014;55(6):1300–3. doi: 10.3109/10428194.2013.834532.
  26. Bally C, Ades L, Renneville A, et al. Prognostic value of TP53 gene mutations in myelodysplastic syndromes and acute myeloid leukemia treated with azacitidine. Leuk Res. 2014;38(7):751–5. doi: 10.1016/j.leukres.2014.03.012.
  27. Calvo X, Nomdedeu M, Navarro A, et al. High levels of global DNA methylation are an independent adverse prognostic factor in a series of 90 patients with de novo myelodysplastic syndrome. Leuk Res. 2014;38(8):874–81. doi: 10.1016/j.leukres.2014.04.015.
  28. Poloni A, Maurizi G, Mattiucci D, et al. Azacitidine treatment in high risk myelodysplastic patients in complete haematological remission reverts mesenchymal stem cells to a normal phenotype. Blood. 2014;124(21): Abstract 1904.
  29. Hasserjian RP, Campigotto F, Klepeis V, et al. De novo acute myeloid leukemia with 20–29% blasts is less aggressive than acute myeloid leukemia with ³30% blasts in older adults: a Bone Marrow Pathology Group study. Am J Hematol. 2014;89(11):e193–9. doi: 10.1002/ajh.23808.
  30. Voso MT, Breccia M, Lunghi M, et al. Rapid loss of response after withdrawal of treatment with azacitidine: a case series in patients with higher-risk myelodysplastic syndromes or chronic myelomonocytic leukemia. Eur J Haematol. 2013;90(4):345–8. doi: 10.1111/ejh.12079.
  31. Nazha A, Sekeres MA, Garcia-Manero G, et al. Outcomes of patients with myelodysplastic syndromes who achieve stable disease after treatment with hypomethylating agents. Blood. 2014;124(21): Abstract 3273.

Polycythemia Vera: Literature Review and Own Data

I.N. Subortseva, T.I. Kolosheinova, E.I. Pustovaya, E.K. Egorova, A.M. Kovrigina, Yu.V. Pliskunova, T.V. Makarik, A.O. Abdullaev, A.L. Melikyan

Hematology Research Center under the Ministry of Health of the Russian Federation, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Irina Nikolaevna Subortseva, PhD, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-44-72; e-mail: soubortseva@yandex.ru

For citation: Subortseva IN, Kolosheinova TI, Pustovaya EI, et al. Polycythemia Vera: Literature Review and Own Data. Clinical oncohematology. 2015;8(4):397–412 (In Russ).

DOI: 10.21320/2500-2139-2015-8-4-397-412


ABSTRACT

Background & Aims. Polycythemia vera (PV) refers to a group of classic Ph-negative myeloproliferative neoplasms characterized by panmyelosis, pancytosis, high risk of thrombotic and hemorrhagic complications, poor quality of life due to symptoms of tumor proliferation. Low-dose acetylsalicylic acid and phlebotomy/erythrocytapheresis are recommended for patients at low risk of complications, while the cytoreductive therapy (hydroxyurea or interferon alpha) is recommended for patients at high risk of thrombotic and hemorrhagic complications. At present, much attention is paid to the problems of diagnosis and treatment PV.

Methods. The article presents a brief description of the condition, a review of modern methods of treatment, results of observation over 100 PV patients who have been treated in the outpatient department of the Hematology Research Center. The observation period ranged from 6 to 262 months (median follow-up is 14 months).

Results. Patients’ age ranged from 23 to 80 years (median 56 years); 67 % of them were women, and 33 % were men. PV was diagnosed according to 2008 WHO guidelines. JAK2V617F mutation was detected in 100 % of cases. Splenomegaly was found in 70 % of patients. Plethoric symptoms (such as facial and hand hyperemia, and scleral injection) were found in 65 % of patients. All patients complained of headaches and dizziness, and 25 % of patients complained of itching. All patients received symptomatic therapy, antiaggregants, medications improving microcirculation, or antihypoxants. Patients were treated according to clinical guidelines: 49 % of them received hydroxyurea, 14 % IFN a-2b, 14 % a combination therapy (hydroxyurea and phlebotomy or IFN a-2b and phlebotomy), and 23 % of patients phlebotomy alone. Response to treatment was evaluated according to 2009 European LeukemiaNet criteria. Among all patients, the frequency of complete remission was 48 %, the partial response rate was 41 %, and no effect was achieved in 11 % of patients regardless they received therapy or not. The therapy was changed, if the treatment had proved to be ineffective, or in case of intolerance or complications. Complete remission was not achieved after switching treatment from one method to another.

Conclusion. Treatment of PV is mainly symptomatic. The effectiveness of first line therapy (complete remission) ranged from 14.5 to 71 %. It is necessary to conduct clinical trials designed to evaluate the effectiveness and safety of new targeted therapies.


Keywords: myeloproliferative neoplasms, polycythemia vera, JAK2V617F, target therapy.

Received: June 30, 2015

Accepted: November 5, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Stuart BJ, Viera AJ. Polycythemia vera. Am Fam Physician. 2004;69:2139–44.
  2. Hensley B, Geyer H, Mesa R. Polycythemia vera: current pharmacotherapy and future directions. Expert Opin. Pharmacother. 2013;14:609–17. doi: 10.1517/14656566.2013.779671.
  3. Vannucchi AM. Insights into the pathogenesis and management of thrombosis in polycythemia vera and essential thrombocythemia. Intern Emerg Med. 2010;5:177–84. doi: 10.1007/s11739-009-0319-3.
  4. Mehta J, Wang H, Iqbal SU, et al. Epidemiology of myeloproliferative neoplasms in the United States. Leuk Lymphoma. 2014;55:595–600. doi: 10.3109/10428194.2013.813500.
  5. Moulard O, Mehta J, Fryzek J, et al. Epidemiology of myelofibrosis, essential thrombocythemia, and polycythemia vera in the European Union. Eur J Haematol. 2014;92:289–97. doi: 10.1111/ejh.12256.
  6. Passamonti F, Rumi E, Pungolino E, et al. Life expectancy and prognostic factors for survival in patients with polycythemia vera and essential thrombocythemia. Am J Med. 2004;117:755–61. doi: 10.1016/j.amjmed.2004.06.032.
  7. Tibes R, Mesa RA. Emerging drugs for polycythemia vera. Expert Opin Emerg Drugs. 2013;18:393–404. doi: 10.1517/14728214.2013.832754.
  8. Tefferi A, Rumi E, Finazzi G, et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27:1874–81. doi: 10.1038/leu.2013.163.
  9. Bandaranayake RM, Ungureanu D, Shan Y, et al. Crystal structures of the JAK2 pseudo kinase domain and the pathogenic mutant V617F. Nat Struct Mol Biol. 2012;19(8):754–9. doi: 10.1038/nsmb.2348.
  10. Brooks AJ, Dai W, O’Mara ML, et al. Mechanism of activation of protein kinase JAK2 by the growth hormone receptor. Science. 2014;344(6185). doi:10.1126/science.1249783.
  11. Pradhan A, Lambert QT, Griner LN, et al. Activation of American Society of Hematology JAK2-V617F by components of heterodimeric cytokine receptors. J Biol Chem. 2010;285(22):16651–63. doi: 10.1074/jbc.m109.071191.
  12. Chen E, Mullally A. How does JAK2V617F contribute to the pathogenesis of myeloproliferative neoplasms? Hematol Am Soc Hematol Educ Program. 2015;2014:268–76. doi: 10.1182/asheducation-2014.1.268.
  13. James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144–8. doi: 10.1038/nature03546.
  14. Scott LM, Tong W, Levine RL, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356(5):459–68. doi: 10.1056/nejmoa065202.
  15. Ma W, Kantarjian H, Zhang X, et al. Mutation profile of JAK2 transcripts in patients with chronic myeloproliferative neoplasias. J. Mol. Diagn. 2009;11(1):49–53. doi: 10.2353/jmoldx.2009.080114.
  16. Milosevic JD, Kralovics R. Genetic and epigenetic alterations of myeloproliferative disorders. Int J Hematol. 2013;97(2):183–97. doi:10.1007/s12185-012-1235-2.
  17. Tong W, Zhang J, Lodish HF. LNK inhibits erythropoiesis and Epo-dependent JAK2 activation and downstream signaling pathways. Blood. 2005;105(12):4604–12. doi: 10.1182/blood-2004-10-4093.
  18. Gery S, Cao Q, Gueller S, et al. LNK inhibits myeloproliferative disorder-associated JAK2 mutant, JAK2V617F. J. Leukoc. Biol. 2009;85(6):957–65. doi: 10.1189/jlb.0908575.
  19. Gery S, Gueller S, Chumakova K, et al. Adaptor protein LNK negatively regulates the mutant MPL, MPLW515L associated with myeloproliferative disorders. Blood. 2007;110(9):3360–4. doi: 10.1182/blood-2007-05-089326.
  20. Pardanani A, Lasho T, Finke C, et al. LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia. 2010;24(10):1713–8. doi: 10.1038/leu.2010.163.
  21. 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.
  22. Moran-Crusio K, Reavie L, Shih A, et al. TET2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell. 2011;20(1):11–24. doi: 10.1016/j.ccr.2011.06.001.
  23. Swierczek SI, Yoon D, Bellanne-Chantelot C, et al. Extent of hematopoietic involvement by TET2 mutations in JAK2V617F polycythemia vera. Haematologica. 2011;96(5):775–8. doi: 10.3324/haematol.2010.029678.
  24. Li Z, Cai X, Cai CL, et al. Deletion of TET2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 2011;118(17):4509–18. doi: 10.1182/blood-2010-12-325241.
  25. Busque L, Patel JP, Figueroa ME, et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat. Genet. 2012;44(11):1179–81. doi: 10.1038/ng.2413.
  26. Pardanani A, Lasho TL, Finke CM, et al. IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010;24(6):1146–51. doi: 10.1038/leu.2010.77.
  27. Tefferi A, Lasho TL, Abdel-Wahab O, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24(7):1302–9. doi: 10.1038/leu.2010.113.
  28. Tefferi A, Jimma T, Sulai NH, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia. 2012;26(3):475–80. doi: 10.1038/leu.2011.253.
  29. 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.
  30. Stegelmann F, Bullinger L, Schlenk RF, et al. DNMT3A mutations in myeloproliferative neoplasms. Leukemia. 2011;25(7):1217–9. doi: 10.1038/leu.2011.77.
  31. Abdel-Wahab O, Pardanani A, Rampal R, et al. DNMT3A mutational analysis in primary myelofibrosis, chronic myelomonocytic leukemia and advanced phases of myeloproliferative neoplasms. Leukemia. 2011;25(7):1219–20. doi: 10.1038/leu.2011.82.
  32. Abdel-Wahab O, Adli M, Lafave LM, et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012;22(2):180–93. doi: 10.1016/j.ccr.2012.06.032.
  33. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369:2379–90. doi: 10.1056/nejmoa1311347.
  34. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369:2391–405. doi: 10.1056/nejmoa1312542.
  35. Girodon F, Broseus J, Park-Alexandre JH, et al. Presence of Calreticulin Mutations in JAK2-Negative Polycythemia Vera. Blood. 2014;124: Abstract 1819.
  36. Spivak JL, Considine M, Williams DM, et al. Two Clinical Phenotypes in Polycythemia Vera. N Engl J Med. 2014;371:808–17. doi: 10.1056/NEJMoa1403141.
  37. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114:937–51. doi: 10.1182/blood-2009-03-209262.
  38. McMullin MF, Reilly JT, Campbell P, et al. Amendment to the guideline for diagnosis and investigation of polycythaemia/erythrocytosis. Br J Haematol. 2007;138:821–2. doi: 10.1111/j.1365-2141.2007.06741.x.
  39. Berk PD, Goldberg JD, Donovan PB, et al. Therapeutic recommendations in polycythemia vera based on polycythemia vera study group protocols. Semin. Hematol. 1986;23:132–43.
  40. Murphy S. Diagnostic criteria and prognosis in polycythemia vera and essential thrombocythemia. Semin Hematol. 1999;36:9–13.
  41. Barbui T, Thiele J, Vannucchi AM, et al. Rethinking the diagnostic criteria of polycythemia vera. Leukemia. 2014;28:1191–5. doi: 10.1038/leu.2013.380.
  42. Barbui T, Thiele J, Carobbio A, et al. Masked polycythemia vera diagnosed according to WHO and BCSH classification. Am J Hematol. 2014;89:199–202. doi: 10.1002/ajh.23617.
  43. Scherber R, Dueck AC, Johansson P, et al. The myeloproliferative neoplasm symptom assessment form (MPN-SAF): international prospective validation and reliability trial in 402 patients. Blood. 2011;118:401–8. doi: 10.1182/blood-2011-01-328955.
  44. Emanuel RM, Dueck AC, Geyer HL, et al. Myeloproliferative neoplasm (MPN) symptom assessment form total symptom score: prospective international assessment of an abbreviated symptom burden scoring system among patients with MPNs. J Clin Oncol. 2012;30:4098–103. doi: 10.1200/jco.2012.42.3863.
  45. Abelsson J, Andreasson B, Samuelsson J, et al. Patients with polycythemia vera have worst impairment of quality of life among patients with newly diagnosed myeloproliferative neoplasms. Leuk Lymphoma. 2013;54:2226–30. doi: 10.3109/10428194.2013.766732.
  46. Geyer HL, Emanuel RM, Dueck AC, et al. Distinct clustering of symptomatic burden amongst myeloproliferative neoplasm patients: retrospective assessment in 1470 patients. Blood. 2014;123(24):3803–10. doi: 10.1182/blood-2013-09-527903.
  47. Johansson P, Mesa R, Scherber R, et al. Association between quality of life and clinical parameters in patients with myeloproliferative neoplasms. Leuk Lymphoma. 2012;53:441–4. doi: 10.3109/10428194.2011.619608.
  48. Scherber R, Dueck AC, Kiladjian JJ, et al. (HU/IFNa/Aspirin) Conventional therapeutic options have limited impact on MPN symptoms: insights from a prospective analysis of the MPN-SAF. In: European Hematology Association, Amsterdam, Netherlands, June 14–17 2012. Abstract 366.
  49. Samuelsson J, Hasselbalch H, Bruserud O, et al. A phase II trial of pegylated interferon alpha-2b therapy for polycythemia vera and essential thrombocythemia: feasibility, clinical and biologic effects, and impact on quality of life. Cancer. 2006;106:2397–405. doi: 10.1002/cncr.21900.
  50. Tefferi A, Elliott M. Thrombosis in myeloproliferative disorders: prevalence, prognostic factors, and the role of leukocytes and JAK2V617F. Semin Thromb Hemost. 2007;33:313–20. doi: 10.1055/s-2007-976165.
  51. Chou YS, Gau JP, Yu YB, et al. Leukocytosis in polycythemia vera and splenomegaly in essential thrombocythemia are independent risk factors for hemorrhage. Eur J Haematol. 2013;90:228–36. doi: 10.1111/ejh.12064.
  52. Меликян А.Л., Суборцева И.Н., Суханова Г.А. Тромбогеморрагические осложнения у больных Ph-негативными миелопролиферативными заболеваниями. Кровь. 2014;2(18):21–5.
    [Melikyan AL, Subortseva IN, Sukhanova GA. Thrombohemorrhagic complications in patients with Ph-negative myeloproliferative diseases. Krov’. 2014;2(18):21–5. (In Russ)]
  53. Finazzi G, Caruso V, Marchioli R, et al. Acute leukemia in polycythemia vera: an analysis of 1638 patients enrolled in a prospective observational study. Blood. 2005;105:2664–70. doi: 10.1182/blood-2004-09-3426.
  54. Passamonti F, Brusamolino E, Lazzarino M, et al. Efficacy of pipobroman in the treatment of polycythemia vera: long-term results in 163 patients. Haematologica 2000;85:1011–8.
  55. Kanitakis J, Arbona-Vidal E, Faure M. Porokeratosis in patients with polycythemia rubra vera: a new side effect of hydroxyurea? J Eur Acad Dermatol Venereol. 2012;26:1040–1. doi: 10.1111/j.1468-3083.2011.04202.x.
  56. Marchioli R, Finazzi G, Landolfi R, et al. Vascular and neoplastic risk in a large cohort of patients with polycythemia vera. J Clin Oncol. 2005;23:2224–32. doi: 10.1200/jco.2005.07.062.
  57. Kiladjian JJ, Chevret S, Dosquet C, et al. Treatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980. J Clin Oncol. 2011;29:3907–13. doi: 10.1200/jco.2011.36.0792.
  58. Tefferi A, Rumi E, Finazzi G, et al. Chronic myeloproliferative neoplasias survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27:1874–81. doi: 10.1038/leu.2013.163.
  59. Barbui T, Finazzi MC, Finazzi G. Front-line therapy in polycythemia vera and essential thrombocythemia. Blood. 2012;26:205–11. doi: 10.1016/j.blre.2012.06.002.
  60. Tefferi A. Polycythemia vera and essential thrombocythemia: 2013 update on diagnosis, risk-stratification, and management. Am J Hematol. 2013;88:507–16. doi: 10.1002/ajh.23417.
  61. Finazzi G, Barbui T. Evidence and expertise in the management of polycythemia vera and essential thrombocythemia. Leukemia. 2008;22:1494–502. doi: 10.1038/leu.2008.177.
  62. Landolfi R, Di Gennaro L, Barbui T, et al. Leukocytosis as a major thrombotic risk factor in patients with polycythemia vera. Blood. 2007;109:2446–52. doi: 10.1182/blood-2006-08-042515.
  63. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Prospective identification of high-risk polycythemia vera patients based on JAK2(V617F) allele burden. Leukemia. 2007;21:1952–9. doi: 10.1038/sj.leu.2404854.
  64. Tefferi A, Strand JJ, Lasho TL, et al. Bone marrow JAK2V617F allele burden and clinical correlates in polycythemia vera. Leukemia. 2007;21:2074–5. doi: 10.1038/sj.leu.2404724.
  65. Landolfi R, Marchioli R, Kutti J, et al. Efficacy and safety of low-dose aspirin in polycythemia vera. N Engl J Med. 2004;350:114–24. doi: 10.1056/nejmoa035572.
  66. Mughal TI, Vaddi K, Sarlis NJ, et al. Myelofibrosis-associated complications: pathogenesis, clinical manifestations, and effects on outcomes. Int J Gen Med. 2014;7:89–101. doi: 10.2147/ijgm.s51800.
  67. Marchioli R, Finazzi G, Specchia G, et al. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med. 2013;368:22–33. doi: 10.1056/nejmoa1208500.
  68. Braekkan SK, Mathiesen EB, Njolstad I, et al. Hematocrit and risk of venous thromboembolism in a general population. The Tromso study. Haematologica. 2010;95:270–5. doi: 10.3324/haematol.2009.008417.
  69. Gori T. Viscosity, platelet activation, and hematocrit: progress in understanding their relationship with clinical and subclinical vascular disease. Clin Hemorheol Microcirc. 2011;49:37–42. doi: 10.3233/CH-2011-1455.
  70. Marchioli R, Finazzi G, Specchia G, et al. The CYTO-PV: a large-scale trial testing the intensity of CYTO-reductive therapy to prevent cardiovascular events in patients with Polycythemia Vera. Thrombosis. 2011;2011:794240. doi: 10.1155/2011/794240.
  71. Sever M, Newberry KJ, Verstovsek S. Therapeutic options for patients with polycythemia vera and essential thrombocythemia refractory/resistant to hydroxyurea. Leuk Lymphoma. 2014;55:2685–90. doi: 10.3109/10428194.2014.893310.
  72. Alvarez-Larran A, Pereira A, Cervantes F, et al. Assessment and prognostic value of the European LeukemiaNet criteria for clinicohematologic response, resistance, and intolerance to hydroxyurea in polycythemia vera. Blood. 2012;119:1363–9. doi: 10.1182/blood-2011-10-387787.
  73. Kiladjian JJ, Cassinat B, Chevret S, et al. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 2008;112:3065–72. doi: 10.1182/blood-2008-03-143537.
  74. Kiladjian JJ. Chomienne C, Fenaux P. Interferon-alpha therapy in bcr-abl-negative myeloproliferative neoplasms. Leukemia. 2008;22:1990–8. doi: 10.1038/leu.2008.280.
  75. Bjorkholm M, Derolf AR, Hultcrantz M, et al. Treatment-related risk factors for transformation to acute myeloid leukemia and myelodysplastic syndromes in myeloproliferative neoplasms. J Clin Oncol. 2011;29:2410–5. doi: 10.1200/jco.2011.34.7542.
  76. Barosi G, Vannucchi AM, De Stefano V, et al. Identifying and addressing unmet clinical needs in Ph-neg classical myeloproliferative neoplasms: a consensus-based SIE, SIES, GITMO position paper. Leuk Res. 2014;38:155–60. doi: 10.1016/j.leukres.2013.09.008.
  77. Bleeker JS, Hogan WJ. Thrombocytosis: diagnostic evaluation, thrombotic risk stratification, and risk-based management strategies. Thrombosis. 2011;2011:536062. doi: 10.1155/2011/536062.
  78. Barosi G, Birgegard G, Finazzi G, et al. A unified definition of clinical resistance and intolerance to hydroxycarbamide in polycythaemia vera and primary myelofibrosis: results of a European LeukemiaNet (ELN) consensus process. Br J Haematol. 2010;148:961–3. doi: 10.1111/j.1365-2141.2009.08019.x.
  79. Barosi G, Mesa R, Finazzi G, et al. Revised response criteria for polycythemia vera and essential thrombocythemia: a ELN and IWG-MRT consensus project. Blood. 2013;121:4778–81. doi: 10.1182/blood-2013-01-478891.
  80. Hexner EO, Serdikoff C, Jan M, et al. Lestaurtinib (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the proliferation of primary erythroid cells from patients with myeloproliferative disorders. Blood. 2008;111:5663–71. doi: 10.1182/blood-2007-04-083402.
  81. Hexner E, Roboz G, Hoffman R, et al. Open-label study of oral CEP-701 (lestaurtinib) in patients with polycythaemia vera or essential thrombocythaemia with JAK2-V617F mutation. Br J Haematol. 2014;164:83–93. doi: 10.1111/bjh.12607.
  82. Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799–807. doi: 10.1056/nejmoa1110557.
  83. Cervantes F, Vannucchi AM, Kiladjian JJ, et al. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood. 2013;122:4047–53. doi: 10.1182/blood-2013-02-485888.
  84. Quintas-Cardama A, Kantarjian H, Cortes J, et al. Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond. Nat Rev Drug Discov. 2011;10:127–40. doi: 10.1038/nrd3264.
  85. Verstovsek S, Passamonti F, Rambaldi A, et al. A phase 2 study of ruxolitinib, an oral JAK1 and JAK2 inhibitor, in patients with advanced polycythemia vera who are refractory or intolerant to hydroxyurea. Cancer. 2014;120:513–20. doi: 10.1002/cncr.28441.
  86. Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372:426–35. doi: 10.1056/nejmoa1409002.
  87. Passamonti F, Saydam G, Lim L, et al. RESPONSE 2: a phase 3b study evaluating the efficacy and safety of ruxolitinib in patients with hydroxyurea-resistant/intolerant polycythemia vera vs best available therapy. J Clin Oncol. 2014;32: Abstract TPS7128.
  88. Mesa R, Vannucchi AM, Yacoub A, et al. The Efficacy and Safety of Continued Hydroxyurea Therapy Versus Switching to Ruxolitinib in Patients with Polycythemia Vera: A Randomized, Double-Blind, Double-Dummy, Symptom Study (RELIEF). ASH Education Book. 2014;124(21):3168.
  89. Pardanani A, Gotlib J, Gupta V, et al. Phase I/II study of CYT387, a JAK1/JAK2 inhibitor for the treatment of myelofibrosis. Blood. 2012;120: Abstract 178.
  90. Verstovsek S, Mesa RA, Salama ME, et al. Phase I study of LY2784544, a JAK2 selective inhibitor, in patients with myelofibrosis (MF), polycythemia vera (PV), and essential thrombocythemia (ET). Blood. 2013;122: Abstract 665.
  91. Vigushin DM, Coombes RC. Targeted histone deacetylase inhibition for cancer therapy. Curr Cancer Drug Targets. 2004;4:205–18. doi: 10.2174/1568009043481560.
  92. Rambaldi A, Dellacasa CM, Finazzi G, et al. A pilot study of the histone-deacetylase inhibitor givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. Br J Haematol. 2010;150:446–55. doi: 10.1111/j.1365-2141.2010.08266.x.
  93. Andersen C, Mortensen N, Vestergaard H, et al. A phase II study of vorinostat (MK-0683) in patients with primary myelofibrosis (PMF) and post-polycythemia vera myelofibrosis (PPV-MF). Haematologica. 2013;98: Abstract P279.
  94. Quintas-Cardama A, Kantarjian H, Manshouri T, et al. Pegylated interferon alfa-2a yields high rates of hematologic and molecular response in patients with advanced essential thrombocythemia and polycythemia vera. J Clin Oncol. 2009;27:5418–24. doi: 10.1200/jco.2009.23.6075.
  95. Mascarenhas J, Mesa R, Prchal J, Hoffman R. Optimal therapy for polycythemia vera and essential thrombocythemia can only be determined by the completion of randomized clinical trials. Haematologica. 2014;99(6):945–9. doi: 10.3324/haematol.2014.106013.
  96. Harrison CN, Garcia NC. Management of MPN beyond JAK2. ASH Education Book. 2014;2014(1):348–54. doi:10.1182/asheducation-2014.1.348.