Use of Blinatumomab in Acute Lymphoblastic Leukemia in Municipal Healthcare: A Case Report

VA Shuvaev1,2, OV Ushakova1, EI Mullo1, TV Tolstykh1, NZ Triputen1

1 VV Veresaev Municipal Clinical Hospital, 10 Lobnenskaya str., Moscow, Russian Federation, 127644

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

For correspondence: Vasilii Anatolevich Shuvaev, MD, PhD, 10 Lobnenskaya str., Moscow, Russian Federation, 127644; e-mail: shuvaev77@mail.ru

For citation: Shuvaev VA, Ushakova OV, Mullo EI, et al. Use of Blinatumomab in Acute Lymphoblastic Leukemia in Municipal Healthcare: A Case Report. Clinical oncohematology. 2021;14(2):198–203. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-198-203


ABSTRACT

Acute lymphoblastic leukemia is one of the groups of most challenging malignant neoplasms of hematopoietic tissue. Despite the success in achieving remission induction in primary patients, later, most of them develop disease relapses. Overall and disease-free survivals have to be improved, which cannot be achieved solely with chemotherapy intensification. The new target drugs and cell technologies improve the treatment options for the resistant forms and relapses of acute lymphoblastic leukemia. The effective use of new drugs presupposes their timely assignment which can be ensured by their availability in routine clinical practice. The provided case report describes the successful use of bispecific antibody blinatumomab for treating an early relapse of acute lymphoblastic leukemia in the clinical practice within the municipal healthcare system.

Keywords: acute lymphoblastic leukemia, clinical practice, target therapy, blinatumomab.

Received: September 22, 2020

Accepted: February 3, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Pui CH, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet. 2008;371(9617):1030–43. doi: 10.1016/s0140-6736(08)60457-2.
  2. Vos T, Allen C, Arora M, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388(10053):1545–602. doi: 10.1016/S0140-6736(16)31678-6.
  3. Савченко В.Г., Паровичникова Е.Н., Афанасьев Б.В. и др. Клинические рекомендации по диагностике и лечению острых лимфобластных лейкозов взрослых 2018. (электронный ресурс) Доступно по: https://npngo.ru/uploads/media_document/293/556718e9–0ff5–46f3-bff8-bd592c83bpdf. Ссылка активна на 22.09.2020 г.
    [Savchenko VG, Parovichnikova EN, Afanasyev BV, et al. Clinical guidelines on diagnosis and treatment of adult acute lymphoblastic leukemia 2018. [Internet] Available from: https://npngo.ru/uploads/media_document/293/556718e9–0ff5–46f3-bff8-bd592c83b992.pdf. (accessed 22.09.2020) (In Russ)]
  4. Kantarjian HM, Walters RS, Keating MJ, et al. Results of the vincristine, doxorubicin, and dexamethasone regimen in adults with standard- and high-risk acute lymphocytic leukemia. J Clin Oncol. 1990;8(6):994–1004. doi: 10.1200/jco.1990.8.6.994.
  5. Паровичникова Е.Н., Соколов А.Н., Троицкая В.В. и др. Острые Ph-негативные лимфобластные лейкозы взрослых: факторы риска при использовании протокола ОЛЛ-2009. Терапевтический архив. 2016;88(7):15–24. doi: 10.17116/terarkh201688715-24.
    [Parovichnikova EN, Sokolov AN, Troitskaya VV, et al. Acute Ph-negative lymphoblastic leukemias in adults: risk factors in the use of the ALL-2009 protocol. Terapevticheskii arkhiv. 2016;88(7):15–24. doi: 10.17116/terarkh201688715-24. (In Russ)]
  6. Ciftciler R, Sevindik OG, Tekgunduz AIE, et al. Acute Lymphoblastic Leukemia in Routine Practice: A Turkish Multicenter Study. Turk J Haematol. 2019;36(3):169–77. doi: 10.4274/tjh.galenos.2019.2019.0008.
  7. Pehlivan KC, Duncan BB, Lee DW. CAR-T Cell Therapy for Acute Lymphoblastic Leukemia: Transforming the Treatment of Relapsed and Refractory Disease. Curr Hematol Malig Rep. 2018;13(5):396–406. doi: 10.1007/s11899-018-0470-x.
  8. Kantarjian H, Thomas D, Jorgensen J, et al. Results of inotuzumab ozogamicin, a CD22 monoclonal antibody, in refractory and relapsed acute lymphocytic leukemia. Cancer. 2013;119(15):2728–36. doi: 10.1002/cncr.28136.
  9. BLINCYTO® (blinatumomab) for injection, for intravenous use. Initial U.S. approval: 2014. Available from: https://www.pi.amgen.com/~/media/amgen/repositorysites/pi-amgen-com/blincyto/blincyto_pi_hcp_english.pdf. (accessed 21.09.2020).
  10. Kantarjian H, Stein A, Gokbuget N. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. New Engl J Med. 2017;376(9):836–47. doi: 10.1056/NEJMoa1609783.
  11. Martinelli G, Boissel N, Chevallier P, et al. Complete Hematologic and Molecular Response in Adult Patients With Relapsed/Refractory Philadelphia Chromosome-Positive B-Precursor Acute Lymphoblastic Leukemia Following Treatment With Blinatumomab: Results From a Phase II, Single-Arm, Multicenter Study. J Clin Oncol. 2017;35(16):1795–802. doi: 10.1200/jco.2016.69.3531.
  12. von Stackelberg A, Locatelli F, Zugmaier G, et al. Phase 1/2 Study in Pediatric Patients with Relapsed/Refractory B-Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) Receiving Blinatumomab Treatment. Blood. 2014;124(21):2292. doi: 10.1182/blood.V124.21.2292.2292.
  13. von Stackelberg A, Locatelli F, Zugmaier G. Phase I/Phase II Study of Blinatumomab in Pediatric Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia. J Clin Oncol. 2016;34(36):4381–9. doi: 10.1200/jco.2016.67.3301.
  14. Topp MS, Gokbuget N, Zugmaier G, et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J Clin Oncol. 2014;32(36):4134–40. doi: 10.1200/jco.2014.56.3247.
  15. Topp MS, Gokbuget N, Stein AS, et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16(1):57–66. doi: 10.1016/s1470-2045(14)71170-2.
  16. Kantarjian H, Thomas D, O’Brien S, et al. Long-term follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemia. Cancer. 2004;101(12):2788–801. doi: 10.1002/cncr.20668.
  17. Specchia G, Pastore D, Carluccio P, et al. FLAG-IDA in the treatment of refractory/relapsed adult acute lymphoblastic leukemia. Ann Hematol. 2005;84(12):792–5. doi: 10.1007/s00277-005-1090-9.
  18. Hunger SP, Mullighan CG. Acute Lymphoblastic Leukemia in Children. N Engl J Med. 2015;373(16):1541–52. doi: 10.1056/NEJMra1400972.
  19. Pulte D, Gondos A, Brenner H. Improvement in survival in younger patients with acute lymphoblastic leukemia from the 1980s to the early 21st century. Blood. 2009;113(7):1408–11. doi: 10.1182/blood-2008-06-164863.
  20. Geyer MB, Hsu M, Devlin SM, et al. Overall survival among older US adults with ALL remains low despite modest improvement since 1980: SEER analysis. Blood. 2017;129(13):1878–81. doi: 10.1182/blood-2016-11-749507.
  21. Бондаренко С.Н., Паровичникова Е.Н., Масчан А.А. и др. Блинатумомаб в терапии острого лимфобластного лейкоза: Российское многоцентровое исследование. Клиническая онкогематология. 2019;12(2):145–53. doi: 10.21320/2500-2139-2019-12-2-145-153.
    [Bondarenko SN, Parovichnikova EN, Maschan AA, et al. Blinatumomab in the Treatment of Acute Lymphoblastic Leukemia: Russian Multicenter Clinical Trial. Clinical oncohematology. 2019;12(2):145–53. doi: 10.21320/2500-2139-2019-12-2-145-153. (In Russ)]
  22. Markova IV, Bondarenko SN, Paina OV, et al. Features of response to blinatumomab and inotuzumab ozogamicin therapy in patients with relapse/refractory B-cells acute lymphoblastic leukemia in real clinical practice. Cell Ther Transplant. 2020;9(1):47–52. doi: 10.18620/ctt-1866-8836-2020-9-1-47-52.

Clinical Experience and Perspectives of Bosutinib Use in Patients with Chronic Myeloid Leukemia

VA Shuvaev1, OYu Vinogradova2,3,4, IS Martynkevich1, NV Novitskaya2, MS Fominykh1, SN Tsareva2, DI Shikhbabaeva2, MM Pankrashkina2,3, MV Chernikov2, NN Sharkunov2, II Zotova1, VYu Udal’eva1, EV Motyko1, SV Voloshin1,5,6

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

2 SP Botkin Municipal Clinical Hospital, 5 2-i Botkinskii pr-d, Moscow, Russian Federation, 125284

3 Dmitrii Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 1 Samory Mashela str., Moscow, Russian Federation, 117198

4 NI Pirogov Russian National Research Medical University, 1 Ostrovityanova str., Moscow, Russian Federation, 117997

5 SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

6 II Mechnikov North-Western State Medical University, 41 Kirochnaya str., Saint Petersburg, Russian Federation, 191015

For correspondence: Ol’ga Yur’evna Vinogradova, MD, PhD, 5 2-i Botkinskii pr-d, Moscow, Russian Federation, 125284; Tel.: +7(495)945-97-61; e-mail: olgavinz@mail.ru.

For citation: Shuvaev VA, Vinogradova OYu, Martynkevich IS, et al. Clinical Experience and Perspectives of Bosutinib Use in Patients with Chronic Myeloid Leukemia. Clinical oncohematology. 2018;11(4):288–94.

DOI: 10.21320/2500-2139-2018-11-4-288-294


ABSTRACT

Aim. To evaluate the clinical experience of bosutinib use for treatment of chronic myeloid leukemia (CML) patients with intolerance and resistance to other tyrosine kinase inhibitors (TKI), as well as to compare the obtained results with the data of clinical trials.

Materials & Methods. The analysis was conducted on case history records of 51 CML patients (25 men and 26 women; median age was 56 years, range 28–86). By the beginning of bosutinib therapy 37 chronic phase, 8 acceleration phase, and 6 blast crisis patients were included in the study. Bosutinib was administered as secondline TKI treatment in 10 patients, as thirdline treatment in 18 patients, and as fourthline treatment in 23 patients. The causes for switching to bosutinib were poor tolerance of previous TKI therapy in 21 patients and resistance to previous TKI therapy in 30 patients.

Results. The median duration of bosutinib treatment was 6 months (range 1–50). Bosutinib toxicity profile and its tolerance in common clinical practice corresponded to the data of clinical trials. Because of adverse events the therapy was discontinued only in 5 (10 %) patients. Complete hematological response was 88 % (persistent response was maintained in 76 % of patients); complete cytogenetic response (CCyR) was 39 %, (persistent response in 37 % of cases); major molecular response (MMR) was 31 % (it was confirmed in 25 % of patients during the last follow-up visit). The efficacy of bosutinib in the real clinical setting was slightly higher compared to the results of clinical trials. This difference was associated with a disease phase, a reason for withdrawal of the previous TKI, line of treatment, BCRABL mutations, and the form of them. The therapy was continued in 22 (43 %) patients, most of them reached stable optimal response, both CCyR and MMR.

Conclusion. Bosutinib appears to be an acceptable alternative to other TKIs having its specific mechanisms of action and adverse events. The efficacy and safety of bosutinib proved in routine clinical practice are sufficient to recommend it for use in national hematology.

Keywords: chronic myeloid leukemia, bosutinib, target therapy, tyrosine kinase inhibitors, clinical practice.

Received: May 9, 2018

Accepted: August 10, 2018

Read in PDF 


REFERENCES

  1. Hehlmann R, Lauseker M, Saussele S, et al. Assessment of imatinib as first-line treatment of chronic myeloid leukemia: 10-year survival results of the randomized CML study IV and impact of non-CML determinants. Leukemia. 2017;31(11):2398–406. doi: 10.1038/leu.2017.253.

  2. 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. 2009;114(22):1126.

  3. Абдулкадыров К.М., Шуваев В.А., Мартынкевич И.С. и др. Хронический миелолейкоз: многолетний опыт таргетной терапии. Клиническая онкогематология. 2016;9(1):54–60. doi: 10.21320/2500-2139-2016-9-1-54-60.

    [Abdulkadyrov KM, Shuvaev VA, Martynkevich IS, et al. Chronic Myeloid Leukemia: Long-Term Experience of Target Therapy. Clinical oncohematology. 2016;9(1):54–60. doi: 10.21320/2500-2139-2016-9-1-54-60. (In Russ)]

  4. Шухов О.А., Туркина А.Г., Челышева Е.Ю. и др. Отдаленные результаты терапии ингибиторами тирозинкиназ у больных хроническим миелолейкозом в ранней и поздней хронической фазе. Клиническая онкогематология. 2016;9(3):368.

    [Shukhov OA, Turkina AG, Chelysheva EYu, et al. Long-Term Results of Tyrosine Kinase Inhibitors Treatment in Chronic Myeloid Leukemia Patients in Early and Late Chronic Phase. Clinical oncohematology. 2016;9(3):368, abstract. (In Russ)]

  5. Steegmann JL, Michallet M, Morra E, et al. Imatinib use in chronic phase CML in clinical practice: the UNIC study. J Clin Oncol. 2008;26(15 Suppl):7077. doi: 10.1200/jco.2008.26.15_suppl.7077.

  6. Michallet M, Tulliez M, Corm S, et al. Management of chronic myeloid leukaemia in clinical practice in France: results of the French subset of patients from the UNIC study. Curr Med Res Opin. 2010;26(2):307–17. doi: 10.1185/03007990903479299.

  7. Goldberg SL, Cortes JE, Gambacorti‐Passerini C, et al. First‐line treatment selection and early monitoring patterns in chronic phase‐chronic myeloid leukemia in routine clinical practice: SIMPLICITY. Am J Hematol. 2017;92(11):1214–23. doi: 10.1002/ajh.24887.

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

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

  9. Стахина О.В., Туркина А.Г., Гусарова Г.А. и др. Отдаленные результаты выживаемости больных в поздней хронической фазе Ph+ хронического 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)]

  10. Туркина А.Г., Челышева Е.Ю. Стратегия терапии хронического миелолейкоза: возможности и перспективы. Терапевтический архив. 2013;85(7):4–9.

    [Turkina AG, Chelysheva EYu. Therapeutic strategy for chronic myeloid leukemia: possibilities and prospects. Terapevticheskii arkhiv. 2013;85(7):4–9. (In Russ)]

  11. Branford S, Lawrence R, Grigg A, et al. Long Term Follow up of Patients with CML in Chronic Phase Treated with First-Line Imatinib Suggests That Earlier Achievement of a Major Molecular Response Leads to Greater Stability of Response. Blood. 2008;112(11):2113.

  12. 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.

  13. Kantarjian H, Cortes JE. Complete Cytogenetic Response, Not Deep Molecular Response, Is Associated With Survival in Chronic Myeloid Leukemia. J Clin Oncol. 2014;32(27):3077. doi: 10.1200/jco.2014.56.0904.

  14. Hughes TP, Hochhaus A, Branford S, et al. Long-term prognostic significance of early molecular response to imatinib in newly diagnosed chronic myeloid leukemia: an analysis from the International Randomized Study of Interferon and STI571 (IRIS). Blood. 2010;116(19):3758–65. doi: 10.1182/blood-2010-03-273979.

  15. Smith AG, Painter D, Howell DA, et al. Determinants of survival in patients with chronic myeloid leukaemia treated in the new era of oral therapy: findings from a UK population-based patient cohort. BMJ Open. 2014;4(1):e004266. doi: 10.1136/bmjopen-2013-004266.

  16. Шуваев В.А., Абдулкадырова А.С., Мартынкевич И.С. и др. Опыт лечения хронического миелолейкоза в Санкт-Петербурге. Вестник гематологии. 2011;7(1):43.

    [Shuvaev VA, Abdulkadyrova AS, Martynkevich IS, et al. Experience of chronic myeloid leukemia treatment in Saint Petersburg. Vestnik gematologii. 2011;7(1):43. (In Russ)]

  17. 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.

  18. NCCN Guidelines. Chronic Myelogenous Leukemia Version 4.2018. Available from: https://www.nccn.org/professionals/physician_gls/pdf/cml.pdf. (accessed 10.05.2018).

  19. Hochhaus A, Kantarjian HM, Baccarani M, et al. Dasatinib induces notable hematologic and cytogenetic responses in chronic-phase chronic myeloid leukemia after failure of imatinib therapy. Blood. 2007;109(6):2303–9. doi: 10.1182/blood-2006-09-047266.

  20. Mauro MJ, Baccarani M, Cervantes F, et al. Dasatinib 2-year efficacy in patients with chronic-phase chronic myelogenous leukemia (CML-CP) with resistance or intolerance to imatinib (START-C). J Clin Oncol. 2008;26(15 Suppl):1. doi: 10.1200/jco.2008.26.15_suppl.7009.

  21. 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.

  22. Tefferi A, Letendre L. Nilotinib treatment-associated peripheral artery disease and sudden death: Yet another reason to stick to imatinib as front-line therapy for chronic myelogenous leukemia. Am J Hematol. 2011;86(7):610–1. doi: 10.1002/ajh.22051.

  23. Valent P, Hadzijusufovic E, Schernthaner G-H, et al. Vascular safety issues in CML patients treated with BCR/ABL1 kinase inhibitors. Blood. 2014;125(6):901–6. doi: 10.1182/blood-2014-09-594432.

  24. Quintas-Cardama A, Han X, Kantarjian H, et al. Tyrosine kinase inhibitor–induced platelet dysfunction in patients with chronic myeloid leukemia. Blood. 2009;114(2):261–3. doi: 10.1182/blood-2008-09-180604.

  25. Quintas-Cardama A, Kantarjian H, O’Brien S, et al. Pleural Effusion in Patients With Chronic Myelogenous Leukemia Treated With Dasatinib After Imatinib Failure. J Clin Oncol. 2007;25(25):3908–14. doi: 10.1200/jco.2007.12.0329.

  26. Krauth M-T, Herndlhofer S, Schmook M-T, et al. Extensive pleural and pericardial effusion in chronic myeloid leukemia during treatment with dasatinib at 100 mg or 50 mg daily. Haematologica. 2011;96(1):163–6. doi: 10.3324/haematol.2010.030494.

  27. Cortes JE, Kantarjian HM, Brummendorf TH, et al. Safety and efficacy of bosutinib (SKI-606) in chronic phase Philadelphia chromosome–positive chronic myeloid leukemia patients with resistance or intolerance to imatinib. Blood. 2011;118(17):4567–76. doi: 10.1182/blood-2011-05-355594.

  28. Puttini M, Coluccia AML, Boschelli F, et al. In vitro and In vivo Activity of SKI-606, a Novel Src-Abl Inhibitor, against Imatinib-Resistant Bcr-Abl+ Neoplastic Cells. Cancer Res. 2006;66(23):11314–22. doi: 10.1158/0008-5472.CAN-06-1199.

  29. Redaelli S, Piazza R, Rostagno R, et al. Activity of Bosutinib, Dasatinib, and Nilotinib Against 18 Imatinib-Resistant BCR/ABL Mutants. J Clin Oncol. 2009;27(3):469–71. doi: 10.1200/jco.2008.19.8853.

  30. Khoury HJ, Cortes JE, Kantarjian HM, et al. Bosutinib is active in chronic phase chronic myeloid leukemia after imatinib and dasatinib and/or nilotinib therapy failure. Blood. 2012;119(15):3403–12. doi: 10.1182/blood-2011-11-390120.

  31. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Bosutinib Versus Imatinib for Newly Diagnosed Chronic Myeloid Leukemia: Results From the Randomized BFORE Trial. J Clin Oncol. 2017;36(3):231–7. doi: 10.1200/jco.2017.74.7162.

  32. Бозулиф® (инструкция по медицинскому применению). Pfizer, США. Доступно по: https://www.24farm.ru/preparats/bozulif_pfizer/. Ссылка активна на 30.05.2018.

    [Bosulif® (package insert). Pfizer, USA. Available from: https://www.24farm.ru/preparats/bozulif_pfizer/. (accessed 30.05.2018) (In Russ)]

  33. National Cancer Institute. Common Terminology Criteria for Adverse Events v4.03 (CTCAE). Available from: https://www.eortc.be/services/doc/ctc/CTCAE_4.03_2010-06-14_QuickReference_5x7.pdf. (accessed 29.04.2018).

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.

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.