Efficacy Predictors of the Third-Line Tyrosine Kinase Inhibitor Therapy in Patients with Chronic Phase of Chronic Myeloid Leukemia: Results of a Multi-Center Study

EG Lomaia1, VA Shuvaev2,3, TV Chitanava1, YuD Matvienko1, IS Martynkevich2, SV Voloshin2, EV Efremova2, ES Mileeva2, MS Fominykh4, AE Kersilova3, EV Karyagina5, NV Il’ina5, NV Dorofeeva6, NV Medvedeva6, AV Klimovich6, TV Shneider7, SA Stepanova7, NF Polezhaikovskaya7, NT Siordiya1, EI Sbityakova1, NS Lazorko1, EN Tochenaya1, DV Motorin1, NA Shnalieva1, YuA Alekseeva1, DB Zammoeva1, AYu Zaritskey1

1 VA Almazov National Medical Research Center, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341

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

3 VV Veresaev Municipal Clinical Hospital, 10 Lobnenskaya ul., Moscow, Russian Federation, 127644

4 AVA-PETER, Multispecialty Clinic “Skandinaviya”, 55A Liteinyi pr-t, Saint Petersburg, Russian Federation, 191014

5 Municipal Hospital No. 15, 4 Avangardnaya ul., Saint Petersburg, Russian Federation, 198205

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

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

For correspondence: Tamara Vangelevna Chitanava, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341; e-mail: chitanava.tamara@yandex.ru

For citation: Lomaia EG, Shuvaev VA, Chitanava TV, et al. Efficacy Predictors of the Third-Line Tyrosine Kinase Inhibitor Therapy in Patients with Chronic Phase of Chronic Myeloid Leukemia: Results of a Multi-Center Study. Clinical oncohematology. 2022;15(3):271–81. (In Russ).

DOI: 10.21320/2500-2139-2022-15-3-271-281


ABSTRACT

Background. The introduction of tyrosine kinase inhibitors (TKIs) into real-world clinical practice considerably improved the prognosis for patients with chronic myeloid leukemia (CML). However, during long-term follow-up, almost 1/2 and 2/3 of patients in the chronic phase (CP) discontinue TKI therapy of the first or second line, respectively. According to the Russian and International clinical guidelines, the third-line therapy should include allogeneic hematopoietic stem cell transplantation (allo-HSCT). And yet, some patients on the third-line therapy achieve and sustain optimal response on long-term TKI administration. Up to now, no clear-cut prognostic factors of TKI efficacy in the third-line therapy have been identified. This creates a challenge for treatment decision making after the failures of two lines of TKI therapy.

Aim. To assess the efficacy of the third-line TKI therapy in real-world clinical practice and to identify the factors affecting the long-term therapy outcomes in CML-CP.

Materials & Methods. The retrospective study enrolled 73 CML-CP patients aged ≥ 18 years, treated with TKIs in the third-line at 5 specialized institutions in Saint Petersburg and Leningrad Region. Among the patients there were 26 men (35 %). The median age of the patients was 51 years (range 25–88 years).

Results. With the median (range) third-line TKI therapy duration of 14 (1–120) months, the rate of complete cytogenetic response (CCR) was 30 % (n = 22) in the total cohort. The median time before achieving CCR was 9 (4–25) months. With the median follow-up time from the beginning of third-line TKI therapy till the last visit of 25 (3–136) months, progression to accelerated phase or blast crisis was observed only in 13 (17 %) out of 73 patients. Death was reported in 26 % (n = 19) of cases, among them 5 patients whose death was not CML-associated. At the last visit, 13/73 (18 %) patients were still on third-line TKI therapy. Direct and long-term therapy outcomes, including achievement of CCR and assessment of overall and progression-free survivals, were significantly better in patients with any cytogenetic response (CR) than in those without it or without complete hematologic response.

Conclusion. The implementation of TKIs in the third-line CML-CP therapy seems to be suitable for patients with at least some CR, especially if an optimal donor of hematopoietic stem cells is unavailable or if the risk of severe allo-HSCT complications is too high.

Keywords: chronic myeloid leukemia, chronic phase, complete cytogenetic response, tyrosine kinase inhibitors, third-line therapy, allogeneic hematopoietic stem cell transplantation, minimal residual disease.

Received: April 7, 2022

Accepted: June 20, 2022

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Hochhaus A, Baccarani M, Silver RT, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 2020;34(4):966–84. doi: 10.1038/s41375-020-0776-2.
  2. 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.
  3. Клинические рекомендации [Хронический миелолейкоз]: стандарты ведения больных для врачей [электронный документ]. Под ред. А.Г. Туркиной и др. М., 2020. Доступно по: https://npngo.ru/biblioteka/klinicheskie_rekomendatsii__2019_god_ Ссылка активна на 7.04.2022.
    [Clinical guidelines [Chronic myeloid leukemia]: patient management standards for physicians. (Internet) Available from: https://npngo.ru/biblioteka/klinicheskie_rekomendatsii__2019_god_ (accessed 04.2022). (In Russ)]
  4. 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)]
  5. Hochhaus A, Larson RA, Guilhot F, et al. Long-Term Outcomes of Imatinib Treatment for Chronic Myeloid Leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.
  6. Stone RM, Kim DW, Kantarjian HM, et al. Dasatinib dose-optimization study in chronic phase chronic myeloid leukemia (CML-CP): three-year follow-up with dasatinib 100 mg once daily and landmark analysis of cytogenetic response and progression-free survival (PFS). J Clin Oncol. 2009;27(15_suppl):7007. doi: 10.1200/jco.2009.27.15_suppl.7007.
  7. Shah NP, Cortes JE, Schiffer CA, et al. Five-year follow-up of patients with imatinib-resistant or -intolerant chronic-phase chronic myeloid leukemia (CML-CP) receiving dasatinib. J Clin Oncol. 2011;29(15_suppl):6512. doi: 10.1200/jco.2011.29.15_suppl.6512.
  8. Kantarjian HM, Giles FJ, Bhalla KN, et al. Nilotinib is effective in patients with chronic myeloid leukemia in chronic phase after imatinib resistance or intolerance: 24-month follow-up results. Blood. 2011;117(4):1141–5. doi: 10.1182/blood-2010-03-277152.
  9. 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.
  10. Gambacorti-Passerini C, Cortes JE, Lipton JH, et al. Safety and efficacy of second-line bosutinib for chronic phase chronic myeloid leukemia over a five-year period: final results of a phase I/II study. Haematologica. 2018;103(8):1298–307. doi: 10.3324/haematol.2017.171249.
  11. Шуваев В.А., Виноградова О.Ю., Мартынкевич И.С. и др. Опыт и перспективы клинического применения бозутиниба у пациентов с хроническим миелолейкозом. Клиническая онкогематология. 2018;11(4):288–94. doi: 10.21320/2500-2139-2018-11-4-288-294.
    [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. (In Russ)]
  12. Лазорко Н.С., Ломаиа Е.Г., Романова Е.Г. и др. Ингибиторы тирозинкиназ второго поколения и их токсичность у больных в хронической фазе хронического миелолейкоза. Клиническая онкогематология. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308.
    [Lazorko NS, Lomaia EG, Romanova EG, et al. Second Generation Tyrosine Kinase Inhibitors and Their Toxicity in Treatment of Patients in Chronic Phase of Chronic Myeloid Leukemia. Clinical oncohematology. 2015;8(3):302–8. doi: 21320/2500-2139-2015-8-3-302-308. (In Russ)]
  13. Giralt SA, Arora M, Goldman JM, et al. Impact of imatinib therapy on the use of allogeneic haematopoietic progenitor cell transplantation for the treatment of chronic myeloid leukaemia. Br J Haematol. 2007;137(5):461–7. doi: 10.1111/j.1365-2141.2007.06582.x.
  14. Лазорко Н.С., Ломаиа Е.Г., Зарицкий А.Ю. и др. Результаты третьей линии таргетной терапии у больных хроническим миелолейкозом в хронической фазе при непереносимости или неэффективности двух ингибиторов тирозинкиназ. Клиническая онкогематология. 2016;9(3):352–3.
    [Lazorko NS, Lomaia EG, Zaritskey AYu, et al. The results of the third-line targeted therapy in chronic myeloid leukemia patients in chronic phase with intolerability or inefficacy of two tyrosine kinase inhibitors. Clinical oncohematology. 2016;9(3):352–3. (In Russ)]
  15. Russo Rossi A, Breccia M, Abruzzese E, et al. Outcome of 82 chronic myeloid leukemia patients treated with nilotinib or dasatinib after failure of two prior tyrosine kinase inhibitors. Haematologica. 2013;98(3):399–403. doi: 10.3324/haematol.2012.064337.
  16. Lauseker M, Hanfstein B, Haferlach C, et al. Equivalence of BCR-ABL transcript levels with complete cytogenetic remission in patients with chronic myeloid leukemia in chronic phase. J Cancer Res Clin Oncol. 2014;140(11):1965–9. doi: 10.1007/s00432-014-1746-8.
  17. Pfirrmann M, Hochhaus A, Lauseker M, et al. Recommendations to meet statistical challenges arising from endpoints beyond overall survival in clinical trials on chronic myeloid leukemia. Leukemia. 2011;25(9):1433–8. doi: 10.1038/leu.2011.116.
  18. Kantarjian H, O’Brien S, Jabbour E, et al. Impact of treatment end point definitions on perceived differences in long-term outcome with tyrosine kinase inhibitor therapy in chronic myeloid leukemia. J Clin Oncol. 2011;29(23):3173–8. doi: 10.1200/JCO.2010.33.4169.
  19. Ongoren S, Eskazan AE, Suzan V, et al. Third-line treatment with second-generation tyrosine kinase inhibitors (dasatinib or nilotinib) in patients with chronic myeloid leukemia after two prior TKIs: real-life data on a single center experience along with the review of the literature. Hematology. 2018;23(4):212–20. doi: 10.1080/10245332.2017.1385193.
  20. Ibrahim AR, Paliompeis C, Bua M, et al. Efficacy of tyrosine kinase inhibitors (TKIs) as third-line therapy in patients with chronic myeloid leukemia in chronic phase who have failed 2 prior lines of TKI therapy. Blood. 2010;116(25):5497–500. doi: 10.1182/blood-2010-06-291922.
  21. Bosi GR, Fogliatto LM, Costa TEV, et al. What happens to intolerant, relapsed or refractory chronic myeloid leukemia patients without access to clinical trials? Hematol Transfus Cell Ther. 2019;41(3):222–8. doi: 10.1016/j.htct.2018.11.005.
  22. Garg RJ, Kantarjian H, O’Brien S, et al. The use of nilotinib or dasatinib after failure to 2 prior tyrosine kinase inhibitors: long-term follow-up. Blood. 2009;114(20):4361–8. doi: 10.1182/blood-2009-05-221531.
  23. Ribeiro BF, Miranda EC, Albuquerque DM, et al. Treatment with dasatinib or nilotinib in chronic myeloid leukemia patients who failed to respond to two previously administered tyrosine kinase inhibitors—a single center experience. Clinics (Sao Paulo). 2015;70(8):550–5. doi: 10.6061/clinics/2015(08)04.
  24. Cortes JE, Khoury HJ, Kantarjian HM, et al. Long-term bosutinib for chronic phase chronic myeloid leukemia after failure of imatinib plus dasatinib and/or nilotinib. Am J Hematol. 2016;91(12):1206–14. doi: 10.1002/ajh.24536.
  25. Garcia-Gutierrez V, Milojkovic D, Hernandez-Boluda JC, et al. Safety and efficacy of bosutinib in fourth-line therapy of chronic myeloid leukemia patients. Ann Hematol. 2019;98(2):321–30. doi: 10.1007/s00277-018-3507-2.
  26. Giles FJ, le Coutre PD, Pinilla-Ibarz J, et al. Nilotinib in imatinib-resistant or imatinib-intolerant patients with chronic myeloid leukemia in chronic phase: 48-month follow-up results of a phase II study. Leukemia. 2013;27(1):107–12. doi: 10.1038/leu.2012.181.
  27. Shah NP, Guilhot F, Cortes JE, et al. Long-term outcome with dasatinib after imatinib failure in chronic-phase chronic myeloid leukemia: follow-up of a phase 3 study. Blood. 2014;123(15):2317–24. doi: 10.1182/blood-2013-10-532341.
  28. Milojkovic D, Nicholson E, Apperley JF, et al. Early prediction of success or failure of treatment with second-generation tyrosine kinase inhibitors in patients with chronic myeloid leukemia. Haematologica. 2010;95(2):224–31. doi: 10.3324/haematol.2009.012781.
  29. Tan J, Xue M, Pan J, et al. Responses to Dasatinib as a Second- and Third-Line Tyrosine Kinase Inhibitor in Chronic Phase Chronic Myeloid Leukaemia Patients. Acta Haematol. 2019;142(2):79–86. doi: 10.1159/000495335.
  30. Jabbour E, Bahceci E, Zhu C, et al. Predictors of Long-Term Cytogenetic Response Following Dasatinib Therapy of Patients with Chronic-Phase Chronic Myeloid Leukemia (CML-CP). Blood. 2009;114(22):3296. doi: 10.1182/blood.V114.22.3296.3296.
  31. Soverini S, Hochhaus A, Nicolini FE, et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood. 2011;118(5):1208–15. doi: 10.1182/blood-2010-12-326405.

Russian Prospective Non-Randomized Clinical Study on Dose Reduction of Tyrosine Kinase Inhibitors with Subsequent Complete Therapy Discontinuation in Chronic Myeloid Leukemia Patients with Stable Deep Molecular Response (READIT-2020): Background, Aim, Main Objectives, Design, and Expected Results

AG Turkina, MA Gurianova, EYu Chelysheva, OA Shukhov

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

For correspondence: Margarita Anatolevna Gurianova, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(985)201-70-40; e-mail: margarita.samtcova@yandex.ru

For citation: Turkina AG, Gurianova MA, Chelysheva EYu, Shukhov OA. Russian Prospective Non-Randomized Clinical Study on Dose Reduction of Tyrosine Kinase Inhibitors with Subsequent Complete Therapy Discontinuation in Chronic Myeloid Leukemia Patients with Stable Deep Molecular Response (READIT-2020): Background, Aim, Main Objectives, Design, and Expected Results. Clinical oncohematology. 2022;15(1):54–61. (In Russ).

DOI: 10.21320/2500-2139-2022-15-1-54-61


ABSTRACT

Background. A withdrawal of tyrosine kinase inhibitor (TKI) therapy in chronic myeloid leukemia (CML) patients with optimal response, especially in patients with drug toxicity, is a matter of current interest. According to the results of numerous clinical studies, the probability of sustaining treatment-free remission (TFR) in CML patients with deep molecular response (DMR) is about 40–60 %. Great attention has recently been paid to personalized therapy consisting in TKI dose modification aimed at reducing or preventing therapy adverse events. Many large retrospective studies showed that reduced TKI doses in CML patients with major molecular response (MMR) and DMR is a safe therapy option. The follow-up of patients receiving reduced TKI doses is also carried out under prospective clinical studies as a stage prior to therapy discontinuation. This approach shows that the probability of sustaining TFR after the stage of TKI dose reduction is about 70 % which is higher than that after the withdrawal of standard TKI doses.

Aim. To present the background, aim, and main objectives of the study as well as the design and expected results.

Materials & Methods. READIT-2020 (Russian prospective study of REduction And DIscontinuation Treatment of TKI) is a Russian prospective non-randomized clinical study with the main aim of developing a safe management regimen for CML patients with MMR and DMR, who receive reduced TKI doses, with subsequent follow-up in the period of TFR under the control of minimal residual disease. The study is going to enroll 100 patients. Each stage of the clinical study will include a regular molecular genetic monitoring at the central laboratory (National Research Center for Hematology, Moscow). The primary objective is to assess survival without MMR loss (BCR-ABL > 0.1 %) both on reduced TKI doses and during TFR. The primary endpoint is the follow-up period of 12 months after TKI discontinuation.

Trial Registration No.: NCT04578847 (Clinicaltrial.gov).

Keywords: chronic myeloid leukemia, tyrosine kinase inhibitors, major molecular response, deep molecular response, adverse events.

Received: June 2, 2021

Accepted: November 1, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Bower H, Bjorkholm M, Dickman PV, et al. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34(24):2851–7. doi: 10.1200/JCO.2015.66.2866.
  2. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.
  3. Шухов О.А. Молекулярно-цитогенетическая характеристика Ph-позитивного клона у больных хроническим миелолейкозом при длительном воздействии ингибиторов тирозинкиназ: Дис. … канд. мед. наук. М., 2015. 114 с.
    [Shukhov OA. Molekulyarno-tsitogeneticheskaya kharakteristika Ph-pozitivnogo klona u bol’nykh khronicheskim mieloleikozom pri dlitel’nom vozdeistvii ingibitorov tirozinkinaz. (Molecular and cytogenetic characteristics of the Ph-positive clone in patients with chronic myeloid leukemia with prolonged exposure to tyrosine kinase inhibitors.) [dissertation] Moscow; 2015. 114 p. (In Russ)]
  4. Туркина А.Г., Челышева Е.Ю. Стратегия терапии хронического миелоидного лейкоза: 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)]
  5. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.
  6. Etienne G, Guilhot J, Rea D, et al. Long-term follow-up of the French Stop Imatinib (STIM1) study in patients with chronic myeloid leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/jco.2016.68.2914.
  7. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.
  8. Hughes T, Boquimpani C, Takahashi N, et al. Durable treatment free remission after stopping second-line nilotinib in patients with chronic myeloid leukemia in chronic phase: ENESTOP 96-wk update. Haematologica. 2017;102(S2):75.
  9. Туркина А.Г., Петрова А.Н., Челышева Е.Ю. и др. Результаты проспективного исследования по наблюдению больных хроническим миелолейкозом после прекращения терапии ингибиторами тирозинкиназ. Гематология и трансфузиология. 2020;65(4):370–85. doi: 10.35754/0234-5730-2020-65-4-370-385.
    [Turkina AG, Petrova AN, Chelysheva EYu, et al. A prospective study of the monitoring of patients with chronic myeloid leukemia upon withdrawal of tyrosine kinase inhibitor therapy. Russian journal of hematology and transfusiology. 2020;65(4):370–85. doi: 10.35754/0234-5730-2020-65-4-370-385. (In Russ)]
  10. Ломаиа Е.Г., Романова Е.Г., Сбитякова Е.И., Зарицкий А.Ю. Эффективность и безопасность ингибиторов тирозинкиназ 2-го поколения (дазатиниб, нилотиниб) в терапии хронической фазы хронического миелолейкоза. Онкогематология. 2013;2:22–33.
    [Lomaia EG, Romanova EG, Sbityakova EI, Zaritskiy AYu. Efficacy and safety of tyrosine kinase inhibitors (dasatinib, nilotinib) in the treatment of chronic phase chronic myeloid leukemia. 2013;2:22–33. (In Russ)]
  11. Лазорко Н.С., Ломаиа Е.Г., Романова Е.Г. и др. Ингибиторы тирозинкиназ второго поколения и их токсичность у больных в хронической фазе хронического миелолейкоза. Клиническая онкогематология. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308.
    [Lazorko NS, Lomaia EG, Romanova EG, et al. Second Generation Tyrosine Kinase Inhibitors and Their Toxicity in Treatment of Patients in Chronic Phase of Chronic Myeloid Leukemia. Clinical oncohematology. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308. (In Russ)]
  12. Гурьянова М.А., Челышева Е.Ю., Туркина А.Г. Возможности терапии ингибиторами тирозинкиназ в сниженных дозах у пациентов с хроническим миелолейкозом. Клиническая онкогематология. 2021;14(1):118–28. doi: 10.21320/2500-2139-2021-14-1-118-128.
    [Gurianova MA, Chelysheva EYu, Turkina AG. Opportunities of Chronic Myeloid Leukemia Treatment with Reduced Doses of Tyrosine Kinase Inhibitors. Clinical oncohematology. 2021;14(1):118–28. doi: 10.21320/2500-2139-2021-14-1-118-128. (In Russ)]
  13. Talpaz M, Shah NP, Kantarjian H, et al. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med. 2006;354(24):2531–41. doi: 10.1056/NEJMoa055229.
  14. Santana-Hernandez P, Pedraza RCP, Duque SG, et al. Low-Dose Dasatinib as First-Line Treatment for Chronic Myeloid Leukemia: Preliminary Blood. 2017;130(Suppl 1):5254. doi: 10.1182/blood.V130.Suppl_1.5254.5254.
  15. Naqvi K, Jabbour E, Skinner J, et al. Long-term follow-up of lower dose dasatinib (50 mg daily) as frontline therapy in newly diagnosed chronic-phase chronic myeloid leukemia. Cancer. 2020;126(1):67–75. doi: 10.1002/cncr.32504.
  16. Cervantes F, Correa J-G, Perez I, et al. Imatinib dose reduction in patients with chronic myeloid leukemia in sustained deep molecular response. Ann Hematol. 2017;96(1):81–5. doi: 10.1007/s00277-016-2839-z.
  17. Iriyama N, Ohashi K, Hashino S, et al. The efficacy of reduced-dose dasatinib as a subsequent therapy in patients with chronic myeloid leukemia in the chronic phase: the LD-CML study of the Kanto CML Study Group. Intern Med. 2018;57(1):17–23. doi: 10.2169/internalmedicine.9035-17.
  18. Faber E, Divoka M, Skoumalova I, et al. A lower dosage of imatinib is sufficient to maintain undetectable disease in patients with chronic myeloid leukemia with long-term low-grade toxicity of the treatment. Leuk Lymphoma. 2016;57(2):370–5. doi: 3109/10428194.2015.1056184.
  19. Шухов О.А., Гурьянова М.А., Челышева Е.Ю. и др. Оценка стабильности молекулярного ответа у больных хроническим миелоидным лейкозом на сниженных дозах ингибиторов тирозинкиназ второго поколения. Гематология и трансфузиология. 2020;65(1, прил. 1):111–2.
    [Shukhov OA, Gurianova MA, Chelysheva EYu, et al. Assessment of the molecular response stability in patients with chronic myeloid leukemia on reduced doses of second-generation tyrosine kinase inhibitors. Gematologiya i transfuziologiya. 2020;65(1 Suppl 1):111–2. (In Russ)]
  20. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor dose in patients with chronic myeloid leukaemia with stable major molecular response (DESTINY): an interim analysis of a non-randomised, phase 2 trial. Lancet Haematol. 2017;4(7):e310–e316. doi: 10.1016/s2352-3026(17)30066-
  21. Rea D, Cayuela J-M, Dulucq S, et al. Molecular responses after switching from a standard-dose twice-daily nilotinib regimen to a reduced-dose once-daily schedule in patients with chronic myeloid leukemia: a real life observational study (NILO-RED). 2017;130(Suppl 1):318. doi: 10.1182/blood.V130.Suppl_1.318.318.
  22. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor therapy before complete treatment discontinuation in patients with chronic myeloid leukaemia (DESTINY): a non-randomised, phase 2 trial. Lancet Haematol. 2019;6(7):e375–e383. doi: 10.1016/S2352-3026(19)30094-8.
  23. Claudiani S, Apperley J, Khan A, et al. Dose reduction of first and second generation TKIs is effective in the maintenance of major molecular response and may predict successful Tfr in CML patients. 2018;132(Suppl 1):3007. doi: 10.1182/blood-2018-99-119602.
  24. Cayssials E, Torregrosa‐Diaz J, Gallego‐Hernanz P, et al. Low-dose tyrosine kinase inhibitors before treatment discontinuation do not impair treatment-free remission in chronic myeloid leukemia patients: results of a retrospective study. Cancer. 2020;126(15):3438–47. doi: 10.1002/cncr.32940.
  25. Kim DDH, Busque L, Forrest DL, et al. Second attempt of TKI discontinuation with dasatinib for treatment-free remission after failing first attempt with imatinib: treatment-free remission accomplished by dasatinib (TRAD) trial. 2018;132(Suppl 1):787. doi: 10.1182/blood-2018-99-114656.
  26. Legros L, Nicolini F-Е, Etienne G, et al. The TKI-Free Duration after a First Discontinuation Attempt That Failed in CP CML Patients Is a Predictive Factor of TKI-Free Remission after a Second Attempt. 2019;134(Suppl 1):28. doi: 10.1182/blood-2019-123719.
  27. Шуваев В.А., Абдулкадыров К.М., Мартынкевич И.С., Фоминых М.С. Фармакоэкономическое моделирование таргетной терапии у больных хроническим миелолейкозом в ремиссии. Онкогематология. 2014;9(3):16–24. doi: 17650/1818-8346-2014-9-3-16-24.
    [Shuvaev VA, Abdulkadyrov KM, Martynkevich IS, Fominykh MS. Pharmacoeconomic modeling of target therapy of chronic myeloid leukemia in remission. Oncohematology. 2014;9(3):16–24. doi: 10.17650/1818-8346-2014-9-3-16-24. (In Russ)]

Biological Mechanisms of Sustaining Deep Molecular Response in Chronic Myeloid Leukemia Upon Withdrawal of Tyrosine Kinase Inhibitors

EYu Chelysheva, MA Guryanova, AG Turkina

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

For correspondence: Ekaterina Yurevna Chelysheva, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: denve@bk.ru

For citation: Chelysheva EYu, Guryanova MA, Turkina AG. Biological Mechanisms of Sustaining Deep Molecular Response in Chronic Myeloid Leukemia Upon Withdrawal of Tyrosine Kinase Inhibitors. Clinical oncohematology. 2021;14(4):427–35. (In Russ).

DOI: 10.21320/2500-2139-2021-14-4-427-435


ABSTRACT

The feasibility of treatment-free follow-up in chronic myeloid leukemia (CML) patients is an important issue in the era of tyrosine kinase inhibitors (TKI). The clinical trials of TKI withdrawal in case of a stable deep molecular response prove the probability of sustaining molecular remission in 40–60 % of patients. Treatment-free remission (TFR), even under persistence of residual leukemia cells, suggests that there are special biologically determined mechanisms of tumor cell proliferation control, which are independent of BCR-ABL kinase activity. The search for factors determining differences in residual leukemia clone kinetics upon TKI withdrawal is an objective which is crucial for understanding TFR as a new biological phenomenon. The review provides worldwide evidence dealing with the study of immunological, genetic, and other biological mechanisms underlying the control of minimal residual disease upon TKI discontinuation in CML patients.

Keywords: chronic myeloid leukemia, tyrosine kinase inhibitors, treatment-free remission, deep molecular response, minimal residual disease.

Received: May 10, 2021

Accepted: August 23, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Bower H, Bjorkholm M, Dickman PW, et al. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34(24):2851–7. doi: 10.1200/JCO.2015.66.2866.
  2. Cross NCP, White HE, Colomer D, et al. Laboratory recommendations for scoring deep molecular responses following treatment for chronic myeloid leukemia. Leukemia. 2015;29(5):999–1003. doi: 10.1038/leu.2015.29.
  3. Hochhaus A, Baccarani M, Silver RT, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 2020;34(4):966–84. doi: 10.1038/s41375-020-0776-2.
  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. Castagnetti F, Gugliotta G, Breccia M, et al. Long-term outcome of chronic myeloid leukemia patients treated frontline with imatinib. Leukemia. 2015;29(9):1823–31. doi: 10.1038/leu.2015.152.
  6. Claudiani S, Gatenby A, Szydlo R, et al. MR4 sustained for 12 months is associated with stable deep molecular responses in chronic myeloid leukemia. Haematologica. 2019;104(11):2206–14. doi: 10.3324/haematol.2018.214809.
  7. Hehlmann R, Muller MC, Lauseker M, et al. Deep molecular response is reached by the majority of patients treated with imatinib, predicts survival, and is achieved more quickly by optimized high-dose imatinib: results from the randomized CML-study IV. J Clin Oncol. 2014;32(5):415–23. doi: 10.1200/JCO.2013.49.9020.
  8. Туркина А.Г., Новицкая Н.В., Голенков А.К. и др. Регистр больных хроническим миелолейкозом в Российской Федерации: от наблюдательного исследования к оценке эффективности терапии в клинической практике. Клиническая онкогематология. 2017;10(3):390–401. doi: 10.21320/2500-2139-2017-10-3-390-401.
    [Turkina AG, Novitskaya NV, Golenkov AK, et al. Chronic Myeloid Leukemia Patient Registry in the Russian Federation: From Observational Studies to the Efficacy Evaluation in Clinical Practice. Clinical oncohematology. 2017;10(3):390–401. doi: 10.21320/2500-2139-2017-10-3-390-401. (In Russ)]
  9. Graham SM, Jorgensen HG, Allan E, et al. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood. 2002;99(1):319–25. doi: 10.1182/blood.v99.1.319.
  10. Copland M, Hamilton A, Elrick LJ, et al. Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction. Blood. 2006;107(11):4532–9. doi: 10.1182/blood-2005-07-2947.
  11. Goldman J, Gordon M. Why do chronic myelogenous leukemia stem cells survive allogeneic stem cell transplantation or imatinib: does it really matter? Leuk Lymphoma. 2006;47(1):1–7. doi: 10.1080/10428190500407996.
  12. Goldman JM. Chronic myeloid leukemia: molecular targeting as a basis for therapy. Rev Clin Exp Hematol. 2004;7(1):64–72.
  13. Holyoake TL, Vetrie D. The chronic myeloid leukemia stem cell: stemming the tide of persistence. Blood. 2017;129(12):1595–606. doi: 10.1182/blood-2016-09-696013.
  14. Zhou H, Xu R. Leukemia stem cells: the root of chronic myeloid leukemia. Protein Cell. 2015;6(6):403–12. doi: 10.1007/s13238-015-0143-7.
  15. Melo JV, Ross DM. Minimal residual disease and discontinuation of therapy in chronic myeloid leukemia: can we aim at a cure? Hematology Am Soc Hematol Educ Program. 2011;2011(1):136–42. doi: 10.1182/asheducation-2011.1.136.
  16. Tang M, Gonen M, Quintas-Cardama A, et al. Dynamics of chronic myeloid leukemia response to long-term targeted therapy reveal treatment effects on leukemic stem cells. Blood. 2011;118(6):1622–31. doi: 10.1182/blood-2011-02-339267.
  17. Roeder I, Horn M, Glauche I, et al. Dynamic modeling of imatinib-treated chronic myeloid leukemia: functional insights and clinical implications. Nat Med. 2006;12(10):1181–4. doi: 10.1038/nm1487.
  18. Branford S, Seymour JF, Grigg A, et al. BCR-ABL messenger RNA levels continue to decline in patients with chronic phase chronic myeloid leukemia treated with imatinib for more than 5 years and approximately half of all first-line treated patients have stable undetectable BCR-ABL using strict sensitivity criteria. Clin Cancer Res. 2007;13(23):7080–5. doi: 10.1158/1078-0432.CCR-07-0844.
  19. Mughal T, Goldman J. Chronic myeloid leukemia: current status and controversies. Oncology. 2004;18(7):837–44.
  20. Etienne G, Guilhot J, Rea D, et al. Long-term follow-up of the French Stop Imatinib (STIM1) study in patients with chronic myeloid leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/JCO.2016.68.2914.
  21. Pagani IS, Shanmuganathan N, Kok CH, et al. Long-term treatment-free remission of chronic myeloid leukemia with falling levels of residual leukemic cells. Leukemia. 2018;32(12):2572–9. doi: 10.1038/s41375-018-0264-0.
  22. Петрова А.Н., Челышева Е.Ю., Туркина А.Г. Ремиссия без лечения у больных хроническим миелолейкозом: обзор литературы. Онкогематология. 2019;14(3):12–22. doi: 10.17650/1818-8346-2019-14-3-12-22.
    [Petrova AN, Chelysheva EYu, Turkina AG. Treatment-free remission in patients with chronic myeloid leukemia: literature review. Onkogematologiya. 2019;14(3):12–22. doi: 10.17650/1818-8346-2019-14-3-12-22. (In Russ)]
  23. Туркина А.Г., Петрова А.Н., Челышева Е.Ю. и др. Результаты проспективного исследования по наблюдению больных хроническим миелолейкозом после прекращения терапии ингибиторами тирозинкиназ. Гематология и трансфузиология. 2020;65(4):370–85. doi: 10.35754/0234-5730-2020-65-4-370-385.
    [Turkina AG, Petrova AN, Chelysheva EYu, et al. A prospective study of the monitoring of patients with chronic myeloid leukemia upon withdrawal of tyrosine kinase inhibitor therapy. Gematologiya i transfuziologiya. 2020;65(4):370–85. doi: 10.35754/0234-5730-2020-65-4-370-385. (In Russ)]
  24. Шухов О.А., Петрова А.Н., Челышева Е.Ю. и др. Факторы сохранения молекулярной ремиссии после прекращения терапии ингибиторами тирозинкиназ у пациентов с хроническим миелолейкозом: результаты нерандомизированного проспективного клинического исследования. Клиническая онкогематология. 2021;14(1):1–12. doi: 10.21320/2500-2139-2021-14-1-1-12.
    [Shukhov OA, Petrova AN, Chelysheva EYu, et al. Factors for Sustaining Molecular Remission after Discontinuation of Tyrosine Kinase Inhibitors Therapy in Chronic Myeloid Leukemia: Results of Non-Randomized Prospective Clinical Trial. Clinical oncohematology. 2021;14(1):1–12. doi: 10.21320/2500-2139-2021-14-1-1-12. (In Russ)]
  25. Rousselot P, Charbonnier A, Cony-Makhoul P, et al. Loss of major molecular response as a trigger for restarting tyrosine kinase inhibitor therapy in patients with chronic-phase chronic myelogenous leukemia who have stopped imatinib after durable undetectable disease. J Clin Oncol. 2014;32(5):424–30. doi: 10.1200/JCO.2012.48.5797.
  26. Ross DM, Branford S, Seymour JF, et al. Patients with chronic myeloid leukaemia who maintain a complete molecular response after stopping imatinib treatment have evidence of persistent leukaemia by DNA PCR. Leukemia. 2010;24(10):1719–24. doi: 10.1038/leu.2010.185.
  27. Rousselot P, Loiseau C, Delord M, et al. A report on 114 patients who experienced treatment free remission in a single institution during a 15 years period: long term follow-up, late molecular relapses and second attempts. Blood. 2019;134(1):27. doi: 10.1182/blood-2019-129919.
  28. Imagawa J, Tanaka H, Okada M, et al. DADI Trial Group. Discontinuation of dasatinib in patients with chronic myeloid leukaemia who have maintained deep molecular response for longer than 1 year (DADI trial): a multicentre phase 2 trial. Lancet Haematol. 2015;2(12):528–35. doi: 10.1016/S2352-3026(15)00196-9.
  29. Takahashi N, Nishiwaki K, Nakaseko Ch, et al. Treatment-free remission after two-year consolidation therapy with nilotinib in patients with chronic myeloid leukemia: STAT2 trial in Japan. 2018;103(11):1835–42. doi: 10.3324/haematol.2018.194894.
  30. Ilander M, Olsson-Stromberg U, Schlums H, et al. Increased proportion of mature NK cells is associated with successful imatinib discontinuation in chronic myeloid leukemia. Leukemia. 2017;31(5):1108–16. doi: 10.1038/leu.2016.360.
  31. Rea D, Henry G, Khaznadar Z, et al. Natural killer-cell counts are associated with molecular relapse-free survival after imatinib discontinuation in chronic myeloid leukemia: the IMMUNOSTIM study. Haematologica. 2017;102(8):1368–77. doi: 10.3324/haematol.2017.165001.
  32. Borg C, Terme M, Taieb J, et al. Novel mode of action of c-kit tyrosine kinase inhibitors leading to NK cell-dependent antitumor effects. J Clin Invest. 2004;114(4):379–88. doi: 10.1172/JCI21102.
  33. Yoshimoto T, Mizoguchi I, Katagiri S, et al. Immunosurveillance markers may predict patients who can discontinue imatinib therapy without relapse. OncoImmunology. 2014;3(5):28861. doi: 10.4161/onci.28861.
  34. Mizoguchi I, Yoshimoto T, Katagiri S, et al. Sustained upregulation of effector natural killer cells in chronic myeloid leukemia after discontinuation of imatinib. Cancer Sci. 2013;201(104):1146–53. doi: 10.1111/cas.12216.
  35. Ohyashiki K, Katagiri S, Tauchi T, et al. Increased natural killer cells and decreased CD3+CD8 +CD62L+ T cells in CML patients who sustained complete molecular remission after discontinuation of imatinib. Br J Haematol. 2012;157(2):254–6. doi: 10.1111/j.1365-2141.2011.08939.x.
  36. Blake SJ, Lyons AB, Hughes TP. Nilotinib inhibits the Src-family kinase LCK and T-cell function in vitro. J Cell Mol Med. 2009;13(3):599–601. doi: 10.1111/j.1582-4934.2009.00500_1.x.
  37. Schade AE, Schieven GL, Townsend R, et al. Dasatinib, a small-molecule protein tyrosine kinase inhibitor, inhibits T-cell activation and proliferation. Blood. 2008;111(3):1366–77. doi: 10.1182/blood-2007-04-084814.
  38. Mustjoki S, Ekblom M, Arstila TP, et al. Clonal expansion of T/NK-cells during tyrosine kinase inhibitor dasatinib therapy. Leukemia. 2009;23(8):1398–405. doi: 10.1038/leu.2009.46.
  39. Molldrem JJ, Lee PP, Wang C, et al. Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nat Med. 2000;6(9):1018–23. doi: 10.1038/79526.
  40. Ross DM, Branford S, Seymour JF, et al. Safety and efficacy of imatinib cessation for CML patients with stable undetectable minimal residual disease: results from the TWISTER study. Blood. 2013;122(4):515–22. doi: 10.1182/blood-2013-02-483750.
  41. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.
  42. Burchert A, Wolfl S, Schmidt M, et al. Interferon-alpha, but not the ABL-kinase inhibitor imatinib (STI571), induces expression of myeloblastin and a specific T-cell response in chronic myeloid leukemia. Blood. 2003;101(1):259–64. PMID: 12393722.
  43. Essers MA, Offner S, Blanco-Bose WE, et al. IFN alpha activates dormant haematopoietic stem cells in vivo. Nature. 2009;458(7240):904–8. doi: 10.1038/nature07815.
  44. Burchert A, Muller MC, Kostrewa P, et al. Sustained molecular response with interferon alfa maintenance after induction therapy with imatinib plus interferon alfa in patients with chronic myeloid leukemia. J Clin Oncol. 2010;28(8):1429–35. doi: 10.1200/JCO.2009.25.5075.
  45. Hochhaus A, Burchert A, Saussele S, et al. Nilotinib vs nilotinib plus pegylated interferon α (Peg-IFN) induction and nilotinib or Peg-IFN maintenance therapy for newly diagnosed BCR-ABL1 positive chronic myeloid leukemia patients in chronic phase (TIGER study): the addition of Peg-IFN is associated with higher rates of deep molecular response. Blood. 2019;134(1):495. doi: 10.1182/blood-2019-130043.
  46. Nicolini FE, Etienne G, Huguet F, et al. The combination of nilotinib + pegylated IFN alpha 2a provides somewhat higher cumulative incidence rates of MR4.5 at M36 versus nilotinib alone in newly diagnosed CP CML patients. Updated results of the Petals phase III national study. Blood. 2019;134(1):494. doi: 10.1182/blood-2019-123674.
  47. Schutz C, Inselmann S, Sausslele S, et al. Expression of the CTLA-4 ligand CD86 on plasmacytoid dendritic cells (pDC) predicts risk of disease recurrence after treatment discontinuation in CML. Leukemia. 2017;31(4):829–36. doi: 10.1038/leu.2017.9.
  48. Ross DM, Hughes TP, Melo JV. Do we have to kill the last CML cell? Leukemia. 2011;25(2):193–200. doi: 10.1038/leu.2010.197.
  49. Ilaria S, Pagani IS, Dang P, et al. Lineage of measurable residual disease in patients with chronic myeloid leukemia in treatment-free remission. Leukemia. 2020;34(4):1052–61. doi: 10.1038/s41375-019-0647-x.
  50. Herrmann H, Sadovnik I, Cerny-Reiterer S, et al. Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia. Blood. 2014;123(25):3951–62. doi: 10.1182/blood-2013-10-536078.
  51. Raspadori D, Pacelli P, Sicuranza A, et al. Flow cytometry assessment of CD26+ leukemic stem cells in peripheral blood: a simple and rapid new diagnostic tool for chronic myeloid leukemia. Cytometry B Clin Cytom. 2019;96(4):294–9. doi: 10.1002/cyto.b.21764.
  52. Valent P, Sadovnik I, Racil Z, et al. DPPIV (CD26) as a novel stem cell marker in Ph+ chronic myeloid leukaemia. Eur J Clin Invest. 2014;44(12):1239–45. doi: 10.1111/eci.12368.
  53. Blatt K, Menzl I, Eisenwort G, et al. Phenotyping and target expression profiling of CD34+/CD38– and CD34+/CD38+ stem- and progenitor cells in acute lymphoblastic leukemia. Neoplazia. 2018;20(6):632–42. doi: 10.1016/j.neo.2018.04.004.
  54. Cui J, Zhu Z, Liu S, et al. Monitoring of leukemia stem cells in chronic myeloid leukemia patients. Leuk Lymphoma. 2018;59(9):2264–6. doi: 10.1080/10428194.2017.1421755.
  55. Bocchia M, Sicuranza A, Abruzzese E, et al. Residual peripheral blood CD26+ leukemic stem cells in chronic myeloid leukemia patients during TKI therapy and during treatment-free remission. Front Oncol. 2018;8:194. doi: 10.3389/fonc.2018.00194.
  56. Iwasaki M, Liedtke M, Gentles A, et al. Cleary. CD93 marks a non-quiescent human Leukemia Stem Cell population and is required for development of MLL-rearranged acute myeloid leukemia. Cell Stem Cell. 2015;17(4):412–21. doi: 10.1016/j.stem.2015.08.008.
  57. Kinstrie R, Horne GA, Morrison H, et al. CD93 is expressed on chronic myeloid leukemia stem cells and identifies a quiescent population, which persists after tyrosine kinase inhibitor therapy. Leukemia. 2020;34(6):1613–25. doi: 10.1038/s41375-019-0684-5.
  58. Agarwal P, Bhatia R. Influence of bone marrow microenvironment on leukemic stem cells: breaking up an intimate relationship. Adv Cancer Res. 2015;127:227–52. doi: 10.1016/bs.acr.2015.04.007.
  59. Park M, Park C.J, Cho YW, et al. Alterations in the bone marrow microenvironment may elicit defective hematopoiesis: a comparison of aplastic anemia, chronic myeloid leukemia, and normal bone marrow. Exp Hematol. 2017;45:56–63. doi: 10.1016/j.exphem.2016.09.009.
  60. Schepers K, Pietras EM, Reynaud D, et al. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self reinforcing leukemic niche. Cell Stem Cell. 2013;13(3):285–99. doi: 10.1016/j.stem.2013.06.009.
  61. Schepers K, Campbell TB, Passegue E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell. 2015;16(3):254–67. doi: 10.1016/j.stem.2015.02.014.
  62. Петинати Н.А., Шипунова И.Н., Бигильдеев А.Е. и др. Изменения в клетках-предшественницах стромального микроокружения костного мозга больных хроническим миелолейкозом в дебюте заболевания и в ходе лечения. Гематология и трансфузиология. 2019;64(4):424–35. doi: 10.35754/0234-5730-2019-64-4-424-435.
    [Petinati NA, Shipunova IN, Bigildeev AE, et al. Changes in stromal progenitor cells derived from bone marrow in patients with chronic myelogenous leukaemia at the onset of the disease and during treatment. Gematologiya i transfuziologiya. 2019;64(4):424–35. doi: 10.35754/0234-5730-2019-64-4-424-435. (In Russ)]
  63. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor therapy before complete treatment discontinuation in patients with chronic myeloid leukaemia (DESTINY): a non-randomised, phase 2 trial. Lancet Haematol. 2019;6(7):e375–e383. doi: 10.1016/S2352-3026(19)30094-8.
  64. Claudiani S, Apperley JF, Gale RP, et al. E14a2 BCR-ABL1 transcript is associated with a higher rate of treatment-free remission in individuals with chronic myeloid leukemia after stopping tyrosine kinase inhibitor therapy. Haematologica. 2017;102(8):e297–e299. doi: 10.3324/haematol.2017.168740.
  65. D’Adda M, Farina M, Schieppati F, et al. The e13a2 BCR-ABL transcript negatively affects sustained deep molecular response and the achievement of treatment-free remission in patients with chronic myeloid leukemia who receive tyrosine kinase inhibitors. Cancer. 2019;125(10):1674–82. doi: 10.1002/cncr.31977.
  66. Lucas CM, Harris RJ, Giannoudis A, et al. Chronic myeloid leukemia patients with the e13a2 BCR-ABL fusion transcript have inferior responses to imatinib compared to patients with the e14a2 transcript. Haematologica. 2009;94(10):1362–7. doi: 10.3324/haematol.2009.009134.
  67. Lee SE, Choi SY, Song HY, et al. Imatinib withdrawal syndrome and longer duration of imatinib have a close association with a lower molecular relapse after treatment discontinuation: the KID study. Haematologica. 2016;101(6):717–23. doi: 10.3324/haematol.2015.139899.
  68. Schmidt M, Hochhaus A, Konig-Merediz SA, et al. Expression of interferon regulatory factor 4 in chronic myeloid leukemia: correlation with response to interferon alfa therapy. J Clin Oncol. 2000;18(19):3331–8. doi: 10.1200/JCO.2000.18.19.3331.
  69. Schmidt M, Hochhaus A, Nitsche A, et al. Expression of nuclear transcription factor interferon consensus sequence binding protein in chronic myeloid leukemia correlates with pretreatment risk features and cytogenetic response to interferon-alpha. Blood. 2001;97(11):3648–50. doi: 10.1182/blood.v97.11.3648.
  70. La Nasa G, Caocci G, Littera R, et al. Homozygosity for killer immunoglobin-like receptor haplotype A predicts complete molecular response to treatment with tyrosine kinase inhibitors in chronic myeloid leukemia patients. Exp Hematol. 2013;41(5):424–31. doi: 10.1016/j.exphem.2013.01.008.
  71. Caocci G, Martino B, Greco M, et al. Killer immunoglobulin-like receptors can predict TKI treatment-free remission in chronic myeloid leukemia patients. Exp Hematol. 2015;43(12):1015–8. doi: 10.1016/j.exphem.2015.08.004.
  72. Caocci G, Greco M, Delogu G, et al. Telomere length shortening is associated with treatment-free remission in chronic myeloid leukemia patients. J Hematol Oncol. 2016;9(1):63. doi: 10.1186/s13045-016-0293-y.
  73. Смирнихина С.А., Лавров А.В., Адильгереева Э.П. и др. Клиническое значение полноэкзомных исследований миелоидных опухолей методом секвенирования следующего поколения. Клиническая онкогематология. 2013;6(1):11–9.
    [Smirnikhina SA, Lavrov AV, Adilgereeva EP, et al. Clinical significance of the whole-exome studies in myeloid neoplasms using next­generation sequencing. Klinicheskaya onkogematologiya. 2013;6(1):11–9. (In Russ)]
  74. Smirnikhina S, Chelysheva E, Lavrov A, et al. Genetic markers of stable molecular remission in chronic myeloid leukemia after targeted therapy discontinuation. Leuk Lymphoma. 2018;59(10):2512–5. doi: 10.1080/10428194.2018.1434880.
  75. Mori S, Vagge E, le Coutre P, et al. Age and dPCR can predict relapse in CML patients who discontinued imatinib: the ISAV study. Am J Hematol. 2015;90(10):910–4. doi: 10.1002/ajh.24120.
  76. Nicolini FE, Dulucq S, Boureau P, et al. Evaluation of residual disease and TKI duration are critical predictive factors for molecular recurrence after stopping imatinib first-line in chronic phase CML patients. Clin Cancer Res. 2019;25(22):6606–13. doi: 10.1158/1078-0432.CCR-18-3373.
  77. Mahon FX, Boquimpani C, Kim DW, et al. Treatment-free remission after second-line nilotinib treatment in patients with chronic myeloid leukemia in chronic phase: results from a single-group, phase 2, open-label study. Ann Intern Med. 2018;168(7):461–70. doi: 10.7326/M17-1094.
  78. Ross DM, Masszi T, Gomez Casares TM, et al. Durable treatment-free remission in patients with chronic myeloid leukemia in chronic phase following frontline nilotinib: 96-week update of the ENESTfreedom study. J Cancer Res Clin Oncol. 2018;144(5):945–54. doi: 10.1007/s00432-018-2604-x.
  79. Kimura S, Imagawa J, Kazunori M, et al. Treatment-free remission after first-line dasatinib discontinuation in patients with chronic myeloid leukaemia (first-line DADI trial): a single-arm, multicentre, phase 2 trial. Lancet Haematol. 2020;7(3):e218–e225. doi: 10.1016/S2352-3026(19)30235-2.
  80. Kumagai T, Nakaseko C, Nishiwaki K, et al. Dasatinib cessation after deep molecular response exceeding 2 years and natural killer cell transition during dasatinib consolidation. Cancer Sci. 2018;109(1):182–92. doi: 10.1111/cas.13430.
  81. Shah NP, Garcia-Gutierrez V, Jimenez-Velasco A, et al. Dasatinib discontinuation in patients with chronic-phase chronic myeloid leukemia and stable deep molecular response: the DASFREE study. Leuk Lymphoma. 2020;61(3):650–9. doi: 10.1080/10428194.2019.1675879.
  82. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.
  83. Nagafuji K, Matsumura I, Shimose T, et al. Cessation of nilotinib in patients with chronic myelogenous leukemia who have maintained deep molecular responses for 2 years: a multicenter phase 2 trial, stop nilotinib (NILSt). Int J Hematol. 2019;110(6):675–82. doi: 10.1007/s12185-019-02736-5.

Opportunities of Chronic Myeloid Leukemia Treatment with Reduced Doses of Tyrosine Kinase Inhibitors

MA Guryanova, EYu Chelysheva, AG Turkina

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

For correspondence: Margarita Anatolevna Guryanova, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(985)201-70-40; e-mail: margarita.samtcova@yandex.ru

For citation: Guryanova MA, Chelysheva EYu, Turkina AG. Opportunities of Chronic Myeloid Leukemia Treatment with Reduced Doses of Tyrosine Kinase Inhibitors. Clinical oncohematology. 2021;14(1):118–28. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-118-128


ABSTRACT

Tyrosine kinase inhibitor (TKI) therapy results in deep molecular response (MR) in 60–70 % of chronic myeloid leukemia (CML) patients. However, despite high efficacy of TKIs, many patients experience drug toxicity during the treatment. According to clinical studies, the probability of sustaining off-treatment remission in CML patients with deep MR is about 40–60 %. Great attention has recently been paid to personalized therapy of chronic phase CML. It consists in TKI dose modification to reduce or prevent adverse events. Major retrospective studies proved that in patients with optimal response TKI reduced doses can be considered safe from the point of view of sustaining major and deep MRs achieved with standard TKI doses. Also, prospective clinical trials deal with the follow-up using TKI reduced doses as pre-withdrawal period. But up to now, the results of only 4 of such studies have been available. To take a closer look at long-term follow-up of CML patients receiving reduced doses of TKIs, prospective clinical trials need to be carried out. The present article reviews the results of main studies dealing with management of CML patients treated with TKI reduced doses.

Keywords: chronic myeloid leukemia, tyrosine kinase inhibitors, major molecular response, deep molecular response, adverse events, pharmacokinetics of tyrosine kinase inhibitors.

Received: August 3, 2020

Accepted: November 20, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Туркина А.Г., Новицкая Н.В., Голенков А.К. идр. Регистр больных хроническим миелолейкозом в Российской Федерации: от наблюдательного исследования к оценке эффективности терапии в клинической практике. Клиническая онкогематология. 2017;10(3):390–401. doi: 10.21320/2500-2139-2017-10-3-390-401.
    [Turkina AG, Novitskaya NV, Golenkov AK, et al. Chronic Myeloid Leukemia Patient Registry in the Russian Federation: From Observational Studies to the Efficacy Evaluation in Clinical Practice. Clinical oncohematology. 2017;10(3):390–401. doi: 10.21320/2500-2139-2017-10-3-390-401. (In Russ)]
  2. Sasaki K, Strom SS, O’Brien S, et al. Relative survival in patients with chronic-phase chronic myeloid leukaemia in the tyrosine-kinase inhibitor era: analysis of patient data from six prospective clinical trials. Lancet Haematol. 2015;2(5):e186–е193. doi: 10.1016/S2352-3026(15)00048-4.
  3. 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.
  4. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.
  5. Etienne G, Guilhot J, Rea D, et al. Long-term follow-up of the French Stop Imatinib (STIM1) study in patients with chronic myeloid leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/jco.2016.68.2914.
  6. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.
  7. Chelysheva EYu, Petrova AN, Shukhov OA, et al. First interim analysis of the Russian multicenter prospective study RU-SKI: discontinuation of tyrosine kinase inhibitors in patients with chronic myeloid leukemia and deep molecular response. Hemasphere. 2018;2(S1):141.
  8. Туркина А.Г., Челышева Е.Ю., Шуваев В.А. и др. Результаты наблюдения больных хроническим миелолейкозом с глубоким молекулярным ответом без терапии ингибиторами тирозинкиназ. Терапевтический архив. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96.
    [Turkina AG, Chelysheva EYu, Shuvaev VA, et al. Results of following up patients with chronic myeloid leukemia and a deep molecular response without tyrosine kinase inhibitor therapy. Terapevticheskii arkhiv. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96. (In Russ)]
  9. Зейфман А.А., Челышева Е.Ю., Туркина А.Г. и др. Роль селективности ингибиторов тирозинкиназ в развитии побочных эффектов при терапии хронического миелолейкоза. Клиническая онкогематология. 2014;7(1):16–27.
    [Zeifman AA, Chelysheva EYu, Turkina AG, et al. Role of tyrosine­kinase inhibitor selectivity in development of adverse effects during treatment of chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2014;7(1):16–27. (In Russ)]
  10. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.
  11. Cortes JE, Saglio G, Kantarjian H, et al. Final 5-Year Study Results of DASISION: The Dasatinib Versus Imatinib Study in Treatment-Naive Chronic Myeloid Leukemia Patients Trial. J Clin Oncol. 2016;34(20):2333–40. doi: 10.1200/jco.2015.64.8899.
  12. Ломаиа Е.Г., Романова Е.Г., Сбитякова Е.И. и др. Эффективность и безопасность ингибиторов тирозинкиназ 2-го поколения (дазатиниб, нилотиниб) в терапии хронической фазы хронического миелолейкоза. Онкогематология. 2013;2:22–33.
    [Lomaia EG, Romanova EG, Sbityakova EI, et al. Efficacy and safety of 2nd generation tyrosine kinase inhibitors (dasatinib, nilotinib) in the treatment of chronic phase of chronic myeloid leukemia. Onkogematologiya. 2013;2:22–33. (In Russ)]
  13. Лазорко Н.С., Ломаиа Е.Г., Романова Е.Г. и др. Ингибиторы тирозинкиназ второго поколения и их токсичность у больных в хронической фазе хронического миелолейкоза. Клиническая онкогематология. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308.
    [Lazorko NS, Lomaia EG, Romanova EG, et al. Second Generation Tyrosine Kinase Inhibitors and Their Toxicity in Treatment of Patients in Chronic Phase of Chronic Myeloid Leukemia. Clinical oncohematology. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308. (In Russ)]
  14. Kantarjian H, Pasquini R, Levy V, et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia resistant to imatinib at a dose of 400 to 600 milligrams daily: two-year follow-up of a randomized phase 2 study (START-R). Cancer. 2009;115(18):4136–47. doi: 10.1002/cncr.24504.
  15. 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.
  16. de Lavallade H, Punnialingam S, Milojkovic D, et al. Pleural effusions in patients with chronic myeloid leukaemia treated with dasatinib may have an immune-mediated pathogenesis. Br J Haematol. 2008;141(5):745–7. doi: 10.1111/j.1365-2141.2008.07108.
  17. 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.
  18. Brummendorf HT, Cortes JE, de Souza CA, et al. Bosutinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukaemia: results from the 24-month follow-up of the BELA trial. Br J Haematol. 2015;168(1):69–81. doi: 10.1111/bjh.13108.
  19. Kerkela R, Grazette I, Yacolti R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med. 2006;12(8):908–16. doi: 10.1038/nm1446.
  20. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/nejmoa1609324.
  21. Hadzijusufovic E, Albrecht-Schgoer K, Huber K, et al. Nilotinib-induced vasculopathy: identifi cation of vascular endothelial cells as a primary target site. Leukemia. 2017;31(11):2388–97. doi: 10.1038/leu.2017.245.
  22. Троицкая Е.А., Вельмакин С.В., Кобалава Ж.Д. Концепция сосудистого возраста: новый инструмент оценки сердечно-сосудистого риска. Артериальная гипертензия. 2017;23(2):160–71. doi: 10.18705/1607-419X-2017-23-2-160-171.
    [Troitskaya EA, Velmakin SV, Kobalava ZD. Concept of vascular age: new tool in cardiovascular risk assessment. Arterial’naya gipertenziya. 2017;23(2):160–71. doi: 10.18705/1607-419X-2017-23-2-160-171. (In Russ)]
  23. Туркина А.Г., Лазарева О.В., Челышева Е.Ю. и др. Результаты терапии больных хроническим миелолейкозом по данным российской части международного многоцентрового популяционного исследования Eutos Population-Based Study (EUTOS-PBS). Гематология и трансфузиология. 2019;64(2):106–21. doi: 10.35754/0234-5730-2019-64-2-106-121.
    [Turkina AG, Lazareva OV, Chelysheva EYu, et al. Treatment outcomes in patients with chronic myeloid leukemia according to the Russian part of the EUTOS Population-Based Study. Russian journal of hematology and transfusiology. 2019;64(2):106–21. doi: 10.35754/0234-5730-2019-64-2-106-121. (In Russ)]
  24. 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.
  25. Rousselot P, Johnson-Ansah H, Huguet F, et al. Personalized daily doses of imatinib by therapeutic drug monitoring increase the rates of molecular responses in patients with chronic myeloid leukemia. Final results of the randomized OPTIM imatinib study. Blood. 2015;126(23):133. doi: 10.1182/blood.v126.23.133.133.
  26. Shah NP, Rousselot P, Schiffer C, et al. Dasatinib in imatinib-resistant or -intolerant chronic-phase, chronic myeloid leukemia patients: 7-year follow-up of study CA180-034. Am J Hematol. 2016;91(9):869–74. doi: 10.1002/ajh.24423.
  27. Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010;28(14):2381–8. doi: 10.1200/JCO.2009.26.3087.
  28. Ibrahim AR, Milojkovic D, Bua M, et al. Poor adherence is the main reason for loss of CCyR and imatinib failure for CML patients on long term imatinib therapy. Blood. 2010;116(21):3414. doi: 10.1182/blood.v116.21.3414.3414.
  29. Куцев С.И., Шатохин Ю.В. Влияние перерывов терапии иматинибом на достижение цитогенетического и молекулярного ответов у больных хроническим миелолейкозом. Казанский медицинский журнал. 2009;90(6):827–31.
    [Kutsev SI, Shatokhin YuV. Effect of interruptions in imatinib therapy on achievement of cytogenetic and molecular responses in patients with chronic myeloid leukemia. Kazanskii meditsinskii zhurnal. 2009;90(6):827–31. (In Russ)]
  30. Talpaz M, Shah NP, Kantarjian H, et al. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med. 2006;354(24):2531–41. doi: 10.1056/NEJMoa055229.
  31. Santana-Hernandez P, Pedraza RCP, Duque SG, et al. Low-Dose Dasatinib as First-Line Treatment for Chronic Myeloid Leukemia: Preliminary Report. Blood. 2017;130(Suppl 1):5254.
  32. Naqvi K, Jabbour E, Skinner J, et al. Long-term follow-up of lower dose dasatinib (50 mg daily) as frontline therapy in newly diagnosed chronic-phase chronic myeloid leukemia. Cancer. 2020;126(1):67–75. doi: 10.1002/cncr.32504.
  33. Carella AM, Lerma E. Durable responses in chronic myeloid leukemia patients maintained with lower doses of imatinib mesylate after achieving molecular remission. Ann Hematol. 2007;86(10):749–52. doi: 10.1007/s00277-007-0326-2.
  34. Cervantes F, Correa JG, Perez I, et al. Imatinib dose reduction in patients with chronic myeloid leukemia in sustained deep molecular response. Ann Hematol. 2017;96(1):81–5. doi: 10.1007/s00277-016-2839-z.
  35. Iriyama N, Ohashi K, Hashino S, et al. The efficacy of reduced-dose dasatinib as a subsequent therapy in patients with chronic myeloid leukemia in the chronic phase: the LD-CML study of the Kanto CML Study Group. Intern Med. 2018;57(1):17–23. doi: 10.2169/internalmedicine.9035-17.
  36. Hjorth-Hansen H, Stenke L, Soderlund S, et al. Dasatinib induces fast and deep responses in newly diagnosed chronic myeloid leukaemia patients in chronic phase: clinical results from a randomised phase-2 study (NordCML006). Eur J Haematol. 2015;94(3):243–50. doi: 10.1111/ejh.12423.
  37. Santos FP, Kantarjian H, Fava C, et al. Clinical impact of dose reductions and interruptions of second-generation tyrosine kinase inhibitors in patients with chronic myeloid leukaemia. Br J Haematol. 2010;150(3):303–12. doi: 10.1111/j.1365-2141.2010.08245.x
  38. Russo D, Martinelli G, Malagola M, et al. Effects and outcome of a policy of intermittent imatinib treatment in elderly patients with chronic myeloid leukemia. Blood. 2013;121(26):5138–44. doi: 10.1182/blood-2013-01-480194.
  39. La Rosee P, Martiat P, Leitner A, et al. Improved tolerability by a modified intermittent treatment schedule of dasatinib for patients with chronic myeloid leukemia resistant or intolerant to imatinib. Ann Hematol. 2013;92(10):1345–50. doi: 10.1007/s00277-013-1769-2.
  40. Faber E, Divoka M, Skoumalova I, et al. A lower dosage of imatinib is sufficient to maintain undetectable disease in patients with chronic myeloid leukemia with long-term low-grade toxicity of the treatment. Leuk Lymphoma. 2016;57(2):370–5. doi: 10.3109/10428194.2015.1056184.
  41. Шухов О.А., Гурьянова М.А., Челышева Е.Ю. и др. Оценка стабильности молекулярного ответа у больных хроническим миелоидным лейкозом на сниженных дозах ингибиторов тирозинкиназ второго поколения. Гематология и трансфузиология. 2020;65(1, приложение 1):111–2.
    [Shukhov OA, Gur’yanova MA, Chelysheva EYu, et al. Assessment of molecular response stability in chronic myeloid leukemia patients treated with second generation tyrosine kinase inhibitors. Gematologiya i transfuziologiya. 2020;65(1, Suppl 1):111–2. (In Russ)]
  42. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor dose in patients with chronic myeloid leukaemia with stable major molecular response (DESTINY): an interim analysis of a non-randomised, phase 2 trial. Lancet Haematol. 2017;4(7):e310–е316. doi: 10.1016/s2352-3026(17)30066-2.
  43. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor therapy before complete treatment discontinuation in patients with chronic myeloid leukaemia (DESTINY): a non-randomised, phase 2 trial. Lancet Haematol. 2019;6(7):e375–е383. doi: 10.1016/S2352-3026(19)30094-8.
  44. Rea D, Cayuela J, Dulucq S, et al. Molecular responses after switching from a standard-dose twice-daily nilotinib regimen to a reduced-dose once-daily schedule in patients with chronic myeloid leukemia: a real life observational study (NILO-RED). Blood 2017;130(1): Abstract 590.
  45. Claudiani S, Apperley J, Khan A, et al. Dose reduction of first and second generation TKI is effective in the maintenance of major molecular response and may predict successful TFR in CML patients. Blood. 2018;132(1): Abstract 3007.
  46. Cayssials E, Torregrosa-Diaz J, Gallego-Hernanz P, et al. Low-dose tyrosine kinase inhibitors before treatment discontinuation do not impair treatment-free remission in chronic myeloid leukemia patients: results of a retrospective study. Cancer. 2020;126(15):3438–47. doi: 10.1002/cncr.32940.
  47. Singh N, Kumar L, Meena R, et al. Drug monitoring of imatinib levels in patients undergoing therapy for chronic myeloid leukaemia: comparing plasma levels of responders and non-responders. Eur J Clin Pharmacol. 2009;65(6):545–9. doi: 10.1007/s00228-009-0621-z.
  48. Larson RA, Druker BJ, Guilhot F, et al. Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study. Blood. 2008;111(8):4022–8. doi: 10.1182/blood-2007-10-116475.
  49. Picard S, Titier K, Etienne G, et al. Trough imatinib plasma levels are associated with both cytogenetic and molecular responses to standard dose imatinib in chronic myeloid leukemia. Blood. 2007;109(8):3496–9. doi: 10.1182/blood-2006-07-036012.
  50. Takahashi N, Wakita H, Miura M, et al. Correlation between imatinib pharmacokinetics and clinical response in Japanese patients with chronic-phase chronic myeloid leukemia. Clin Pharmacol Ther. 2010;88(6):809–13. doi: 10.1038/clpt.2010.186.
  51. Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010;28(14):2381–8. doi: 10.1200/JCO.2009.26.3087.
  52. Куцев С.И., Оксенюк О.С. Мониторинг в терапии хронического миелолейкоза иматинибом. Клиническая онкогематология. 2009;2(3):225–31.
    [Kutsev SI, Oksenyuk OS. Monitoring in imatinib treatment of chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2009;2(3):225–31. (In Russ)]
  53. Larson RA, Yin OQ, Hochhaus A, et al. Population pharmacokinetic and exposure-response analysis of nilotinib in patients with newly diagnosed Ph+ chronic myeloid leukemia in chronic phase. Eur J Clin Pharmacol. 2012;68(5):723–33. doi: 10.1007/s00228-011-1200-7.
  54. Takahashi N, Miura M, Kuroki J, et al. Multicenter phase II clinical trial of nilotinib for patients with imatinib-resistant or -intolerant chronic myeloid leukemia from the East Japan CML study group evaluation of molecular response and he efficacy and safety of nilotinib. Biomark Res. 2014;2(1):6. doi: 10.1186/2050-7771-2-6.
  55. Tanaka C, Yin OQP, Sethuraman V, et al. Clinical pharmacokinetics of the BCR–ABL tyrosine kinase inhibitor nilotinib. Clin Pharmacol Ther. 2010;87(2):197–203. doi: 10.1038/clpt.2009.208.
  56. Miura M. Therapeutic drug monitoring of imatinib, nilotinib, and dasatinib for patients with chronic myeloid leukemia. Biol Pharm Bull. 2015;38(5):645–54. doi: 10.1248/bpb.b15-00103.
  57. Wang X, Roy A, Hochhaus A, et al. Differential effects of dosing regimen on the safety and efficacy of dasatinib: retrospective exposure–response analysis of a phase III study. Clin Pharmacol. 2013;10(5):85–97. doi: 10.2147/CPAA.S42796.
  58. Mita A, Abumiya M, Miura M, et al. Correlation of plasma concentration and adverse effects of bosutinib: standard dose or dose-escalation regimens of bosutinib treatment for patients with chronic myeloid leukemia. Exp Hematol Oncol. 2018;7(1):9. doi: 10.1186/s40164-018-0101-1.

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

NN Mamaev, AI Shakirova, EV Morozova, TL Gindina

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

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

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

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


ABSTRACT

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

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

Received: September 12, 2020

Accepted: December 6, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Barjesteh van Waalwijk van Doorn-Khosrovani S. High EVI1 expression predicts poor survival in acute myeloid leukemia: a study of 319 de novo AML patients. Blood. 2002;101(3):837–45. doi: 10.1182/blood-2002-05-1459.
  2. Lugthart S, van Drunen E, van Norden Y, et al. High EVI1 levels predict adverse outcome in acute myeloid leukemia: prevalence of EVI1 overexpression and chromosome 3q26 abnormalities underestimated. Blood. 2008;111(8):4329–37. doi: 10.1182/blood-2007-10-119230.
  3. Groschel S, Lugthart S, Schlenk RF, et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. J Clin Oncol. 2010;28(12):2101–7. doi: 10.1200/JCO.2009.26.0646.
  4. Paquette RL, Nicoll J, Chalukya M, et al. Frequent EVI1 translocations in myeloid blast crisis CML that evolves through tyrosine kinase inhibitors. Cancer Genet. 2011;204(7):392–7. doi: 10.1016/j.cancergen.2011.06.002.
  5. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миелодиспластические синдромы с высокой экспрессией гена EVI1: теоретические и клинические аспекты. Клиническая онкогематология. 2012;5(4):361–4.
    [Mamaev NN, Gorbunova AV, Gindina TL, et al. Leukemias and myelodysplastic syndromes with high expression of EVI1 gene: theoretical and clinical aspects. Klinicheskaya onkogematologiya. 2012;5(4):361–4. (In Russ)]
  6. Rogers HJ, Vardiman JW, Anastasi J, et al. Complex or monosomal karyotype and not blast percentage is associated with poor survival in acute myeloid leukemia and myelodysplastic syndrome patients with inv(3)(q21q26.2)/t(3;3)(q21;q26.2): a Bone Marrow Pathology Group study. Haematologica. 2014;99(5):821–9. doi: 10.3324/haematol.2013.096420.
  7. Reiter E, Greinix H, Rabitsch W, et al. Low curative potential of bone marrow transplantation for highly aggressive acute myelogenous leukemia with inversion inv(3)(q21q26) or homologous translocation t(3;3)(q21;q26). Ann Hematol. 2000;79(7):374–7. doi: 10.1007/s002770000158.
  8. He X, Wang Q, Cen J, et al. Predictive value of high EVI1 expression in AML patients undergoing myeloablative allogeneic hematopoietic stem cell transplantation in first CR. Bone Marrow Transplant. 2016;51(7):921–7. doi: 10.1038/bmt.2016.71.
  9. Gindina TL, Mamaev NN, Afanasyev BV. Chromosome abnormalities and hematopoietic stem cell transplantation in acute leukemias. In: ML Larramendy, S Soloneski (eds). Chromosomal abnormalities – A hallmark manifestation of genomic instability. IntechOpen; 2017. рр. 71– doi: 10.5772/67802.
  10. Halaburda K, Labopin M, Houhou M, et al. AlloHSCT for inv(3)(q21;q26)/t(3;3)(q21;q26) AML: a report from the acute leukemia working party of the European society for blood and marrow transplantation. Bone Marrow Transplant. 2018;53(6):683–91. doi: 10.1038/s41409-018-0165-x.
  11. Martinelli G, Ottaviani E, Buonamici S, et al. Association of 3q21q26 syndrome with different RPN1/EVI1 fusion transcripts. Haematologica. 2003;88(11):1221–8.
  12. Poppe B, Dastugue N, Vandesompele J, et al. EVI1 is consistently expressed as principal transcript in common and rare recurrent 3q26 rearrangements. Genes Chromos Cancer. 2006;45(4):349–56. doi: 10.1002/gcc.20295.
  13. De Braekeleer M, Le Bris MJ, De Braekeleer E, et al. 3q26/EVI1 rearrangements in myeloid hemopathies: a cytogenetic review. Fut Oncol. 2015;11(11):1675–86. doi: 10.2217/fon.15.64.
  14. Mamaev NN, Gindina TL, Morozova EV, et al. Primary myelodysplastic syndrome with two rare recurrent chromosome abnormalities [t(3q26/2;q22 and trisomy 13] associated with resistance to chemotherapy and hematopoietic stem cell transplantation. Cell Ther Transplant. 2018;7(2):64–9. doi: 10/18620/ctt-1866-8836-2018-7-2-64-69.
  15. Hodge JC, Bosler D, Rubinstein L, et al. Molecular and pathologic characterization of AML with double inv(3)(q21q26.2). Cancer Genet. 2019;230:28–36. doi: 10.1016/j.cancergen.2018.08.007.
  16. Testoni N, Borsaru G, Martinelli G, et al. 3q21 and 3q26 cytogenetic abnormalities in acute myeloblastic leukemia: biological and clinical features. Haematologica. 1999;84(8):690–4.
  17. Russell M, List A, Greenberg P, et al. Expression of EVI1 in myelodysplastic syndromes and other hematologic malignancies without 3q26 translocations. Blood. 1994;84(4):1243–8. doi: 10.1182/blood.V84.4.1243.1243.
  18. Groschel S, Schlenk RF, Engelmann J, et al. Deregulated expression of EVI1 defines a poor prognostic subset of MLL-rearranged acute myeloid leukemias: a study of the German-Austrian Acute Myeloid Leukemia Study Group and the Dutch-Belgian-Swiss HOVON/SAKK Cooperative Group. J Clin Oncol. 2013;31(1):95–103. doi: 10.1200/JCO.2011.41.5505.
  19. Ho PA, Alonzo TA, Gerbing RB, et al. High EVI1 expression is associated with MLL rearrangements and predicts decreased survival in paediatric acute myeloid leukaemia: a report from the children’s oncology group. Br J Haematol. 2013;162(5):670–7. doi: 10.1111/bjh.12444.
  20. Zhang Y, Owens K, Hatem L, et al. Essential role of PR-domain protein MDS1-EVI1 in MLL-AF9 leukemia. Blood. 2013;122(16):2888–92. doi: 10.1182/blood-2012-08-453662.
  21. Mucenski ML, Taylor BA, Ihle JN, et al. Identification of a common ecotropic viral integration site, Evi-1, in the DNA of AKXD murine myeloid tumors. Mol Cell Biol. 1988;8(1):301–8. doi: 10.1128/mcb.8.1.301.
  22. Goyama S, Kurokawa M. Pathogenetic significance of ecotropic viral integration site-1 in hematological malignancies. Cancer Sci. 2009;100(6):990–5. doi: 10.1111/j.1349-7006.2009.01152.x.
  23. Hinai AA, Valk PJ. Review: Aberrant EVI1 expression in acute myeloid leukaemia. Br J Haematol. 2016;172(6):870–8. doi: 10.1111/bjh.13898.
  24. Yuan X, Wang X, Bi K, Jiang G. The role of EVI-1 in normal hematopoiesis and myeloid malignancies (Review). Int J Oncol. 2015;47(6):2028–36. doi: 10.3892/ijo.2015.3207.
  25. Delwel R, Funabiki T, Kreider BL, et al. Four of the seven zinc fingers of the Evi-1 myeloid-transforming gene are required for sequence-specific binding to GA(C/T)AAGA(T/C)AAGATAA. Mol Cell Biol. 1993;13(7):4291–300. doi: 10.1128/mcb.13.7.4291.
  26. Funabiki T, Kreider BL, Ihle JN. The carboxyl domain of zinc fingers of the Evi-1 myeloid transforming gene binds a consensus sequence of GAAGATGAG. Oncogene. 1994;9(6):1575–81.
  27. Morishita K, Suzukawa K, Taki T, et al. EVI-1 zinc finger protein works as a transcriptional activator via binding to a consensus sequence of GACAAGATAAGATAAN1-28 CTCATCTTC. Oncogene. 1995;10(10):1961–7.
  28. Perkins AS, Kim JH. Zinc fingers 1–7 of EVI1 fail to bind to the GATA motif by itself but require the core site GACAAGATA for binding. J Biol Chem. 1996;271(2):1104–10. doi: 10.1074/jbc.271.2.1104.
  29. Bartholomew C, Kilbey A, Clark AM, Walker M. The Evi-1 proto-oncogene encodes a transcriptional repressor activity associated with transformation. Oncogene. 1997;14(5):569–77. doi: 10.1038/sj.onc.1200864.
  30. Kilbey A, Bartholomew C. Evi-1 ZF1 DNA binding activity and a second distinct transcriptional repressor region are both required for optimal transformation of Rat1 fibroblasts. Oncogene. 1998;16(17):2287–91. doi: 10.1038/sj.onc.1201732.
  31. Bordereaux D, Fichelson S, Tambourin P, Gisselbrecht S. Alternative splicing of the Evi-1 zinc finger gene generates mRNAs which differ by the number of zinc finger motifs. Oncogene. 1990;5(6):925–7.
  32. Alzuherri H, McGilvray R, Kilbey A, Bartholomew C. Conservation and expression of a novel alternatively spliced Evi1 exon. Gene. 2006;384:154–62. doi: 10.1016/j.gene.2006.07.027.
  33. Fears S, Mathieu C, Zeleznik-Le N, et al. Intergenic splicing of MDS1 and EVI1 occurs in normal tissues as well as in myeloid leukemia and produces a new member of the PR domain family. Proc Natl Acad Sci USA. 1996;93(4):1642–7. doi: 10.1073/pnas.93.4.1642.
  34. Huang S, Shao G, Liu L. The PR domain of the Rb-binding zinc finger protein RIZ1 is a protein binding interface and is related to the SET domain functioning in chromatin-mediated gene expression. J Biol Chem. 1998;273(26):15933–9. doi: 10.1074/jbc.273.26.15933.
  35. Goyama S, Yamamoto G, Shimabe M, et al. Evi-1 is a critical regulator for hematopoietic stem cells and transformed leukemic cells. Cell Stem Cell. 2008;3(2):207–20. doi: 10.1016/j.stem.2008.06.002.
  36. Laricchia-Robbio L, Nucifora G. Significant increase of self-renewal in hematopoietic cells after forced expression of EVI1. Blood Cells Mol Dis. 2008;40(2):141–7. doi: 10.1016/j.bcmd.2007.07.012.
  37. Yoshimi A, Kurokawa M. Evi1 forms a bridge between the epigenetic machinery and signaling pathways. Oncotarget. 2011;2(7):575–86. doi: 10.18632/oncotarget.304.
  38. Buonamici S, Li D, Chi Y, et al. EVI1 induces myelodysplastic syndrome in mice. J Clin Invest. 2005;115(8):2296. doi: 1172/jci21716c1.
  39. Cuenco GM, Ren R. Both AML1 and EVI1 oncogenic components are required for the cooperation of AML1/MDS1/EVI1 with BCR/ABL in the induction of acute myelogenous leukemia in mice. Oncogene. 2004;23(2):569–79. doi: 10.1038/sj.onc.1207143.
  40. Glass C, Wilson M, Gonzalez R, et al. The role of EVI1 in myeloid malignancies. Blood Cells Mol Dis. 2014;53(1–2):67–76. doi: 10.1016/j.bcmd.2014.01.002.
  41. Jin G, Yamazaki Y, Takuwa M, et al. Trib1 and Evi1 cooperate with Hoxa and Meis1 in myeloid leukemogenesis. Blood. 2007;109(9):3998–4005. doi: 10.1182/blood-2006-08-041202.
  42. Krivtsov AV, Twomey D, Feng Z, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442(7104):818–22. doi: 10.1038/nature04980.
  43. Bindels EM, Havermans M, Lugthart S, et al. EVI1 is critical for the pathogenesis of a subset of MLL-AF9-rearranged AMLs. Blood. 2012;119(24):5838–49. doi: 10.1182/blood-2011-11-393827.
  44. Glass C, Wuertzer C, Cui X, et al. Global Identification of EVI1 Target Genes in Acute Myeloid Leukemia. PLoS One. 2013;8(6):e67134. doi: 10.1371/journal.pone.0067134.
  45. Hoyt PR, Bartholomew C, Davis AJ, et al. The Evi1 proto-oncogene is required at midgestation for neural, heart, and paraxial mesenchyme development. Mech Dev. 1997;65(1–2):55–70. doi: 10.1016/s0925-4773(97)00057-9.
  46. Nucifora G. The EVI1 gene in myeloid leukemia. Leukemia. 1997;11(12):2022–31. doi: 10.1038/sj.leu.2400880.
  47. Kataoka K, Sato T, Yoshimi A, et al. Evi1 is essential for hematopoietic stem cell self-renewal, and its expression marks hematopoietic cells with long-term multilineage repopulating activity. J Exp Med. 2011;208(12):2403–16. doi: 10.1084/jem.20110447.
  48. Zhang Y, Stehling-Sun S, Lezon-Geyda K, et al. PR-domain-containing Mds1-Evi1 is critical for long-term hematopoietic stem cell function. Blood. 2011;118(14):3853–61. doi: 10.1182/blood-2011-02-334680.
  49. Steinleitner K, Rampetsreiter P, Koffel R, et al. EVI1 and MDS1/EVI1 expression during primary human hematopoietic progenitor cell differentiation into various myeloid lineages. Anticancer Res. 2012;32(11):4883–9.
  50. Wieser R. The oncogene and developmental regulator EVI1: expression, biochemical properties, and biological functions. Gene. 2007;396(2):346–57. doi: 10.1016/j.gene.2007.04.012.
  51. Xi ZF, Russell M, Woodward S, et al. Expression of the Zn finger gene, EVI-1, in acute promyelocytic leukemia. Leukemia. 1997;11(2):212–20. doi: 10.1038/sj.leu.2400547.
  52. Aytekin M, Vinatzer U, Musteanu M, et al. Regulation of the expression of the oncogene EVI1 through the use of alternative mRNA 5’-ends. Gene. 2005;356:160–8. doi: 10.1016/j.gene.2005.04.032.
  53. Niederreither K, Subbarayan Y, Dolle P, et al. Embryonic retinoic acid synthesis is essential for early mouse post-implantation development. Nat Genet. 1999;21(4):444–8. doi: 1038/7788.
  54. Valk PJ, Verhaak RG, Beijen MA, et al. Prognostically useful gene-expression profiles in acute myeloid leukemia. N Engl J Med. 2004;350(16):1617–28. doi: 10.1056/NEJMoa040465.
  55. Morishita K, Parganas E, William CL, et al. Activation of EVI1 gene expression in human acute myelogenous leukemias by translocations spanning 300–400 kilobases on chromosome band 3q26. Proc Natl Acad Sci USA. 1992;89(9):3937–41. doi: 10.1073/pnas.89.9.3937.
  56. Ogawa S, Mitani K, Kurokawa M, et al. Abnormal expression of Evi-1 gene in human leukemias. Hum Cell. 1996;9(4):323–32.
  57. Lugthart S, Groschel S, Beverloo HB, et al. Clinical, molecular, and prognostic significance of WHO type inv(3)(q21q26.2)/t(3;3)(q21;q26.2) and various other 3q abnormalities in acute myeloid leukemia. J Clin Oncol. 2010;28(24):3890–8. doi: 10.1200/JCO.2010.29.2771.
  58. Groschel S, Sanders MA, Hoogenboezem R, et al. Mutational spectrum of myeloid malignancies with inv(3)/t(3;3) reveals a predominant involvement of RAS/RTK signaling pathways. Blood. 2015;125(1):133–9. doi: 10.1182/blood-2014-07-591461.
  59. Langabeer SE, Rogers JR, Harrison G, et al. EVI1 expression in acute myeloid leukaemia. Br J Haematol. 2001;112(1):208–11. doi: 10.1046/j.1365-2141.2001.02569.x.
  60. Balgobind BV, Lugthart S, Hollink IH, et al. EVI1 overexpression in distinct subtypes of pediatric acute myeloid leukemia. Leukemia. 2010;24(5):942–9. doi: 10.1038/leu.2010.47.
  61. Matsuo H, Kajihara M, Tomizawa D, et al. EVI1 overexpression is a poor prognostic factor in pediatric patients with mixed lineage leukemia-AF9 rearranged acute myeloid leukemia. Haematologica. 2014;99(11):e225–е227. doi: 10.3324/haematol.2014.107128.
  62. Testa U, Lo-Coco F. Targeting of leukemia-initiating cells in acute promyelocytic leukemia. Stem Cell Invest. 2015;2:8. doi: 10.3978/j.issn.2306-9759.2015.04.03.
  63. Jo A, Mitani S, Shiba N, et al. High expression of EVI1 and MEL1 is a compelling poor prognostic marker of pediatric AML. Leukemia. 2015;29(5):1076–83. doi: 10.1038/leu.2015.5.
  64. Sadeghian MH, Rezaei Dezaki Z. Prognostic Value of EVI1 Expression in Pediatric Acute Myeloid Leukemia: A Systematic Review. Iran J Pathol. 2018;13(3):294–300.
  65. Arai S, Yoshimi A, Shimabe M, et al. Evi-1 is a transcriptional target of mixed-lineage leukemia oncoproteins in hematopoietic stem cells. Blood. 2011;117(23):6304–14. doi: 10.1182/blood-2009-07-234310.
  66. De Weer A, Van der Meulen J, Rondou P, et al. EVI1-mediated down regulation of MIR449A is essential for the survival of EVI1 positive leukaemic cells. Br J Haematol. 2011;154(3):337–48. doi: 10.1111/j.1365-2141.2011.08737.x.
  67. Yamazaki H, Suzuki M, Otsuki A, et al. A remote GATA2 hematopoietic enhancer drives leukemogenesis in inv(3)(q21;q26) by activating EVI1 expression. Cancer Cell. 2014;25(4):415–27. doi: 10.1016/j.ccr.2014.02.008.
  68. Groschel S, Sanders MA, Hoogenboezem R, et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell. 2014;157(2):369–81. doi: 10.1016/j.cell.2014.02.019.
  69. Lugthart S, Figueroa ME, Bindels E, et al. Aberrant DNA hypermethylation signature in acute myeloid leukemia directed by EVI1. Blood. 2011;117(1):234–41. doi: 10.1182/blood-2010-04-281337.
  70. Bartholomew C, Morishita K, Askew D, et al. Retroviral insertions in the CB-1/Fim-3 common site of integration activate expression of the Evi-1 gene. Oncogene. 1989;4(5):529–34.
  71. Kreider BL, Orkin SH, Ihle JN. Loss of erythropoietin responsiveness in erythroid progenitors due to expression of the Evi-1 myeloid-transforming gene. Proc Natl Acad Sci USA. 1993;90(14):6454–8. doi: 10.1073/pnas.90.14.6454.
  72. Kataoka K, Kurokawa M. Ecotropic viral integration site 1, stem cell self-renewal and leukemogenesis. Cancer Sci. 2012;103(8):1371–7. doi: 10.1111/j.1349-7006.2012.02303.x.
  73. Soderholm J, Kobayashi H, Mathieu C, et al. The leukemia-associated gene MDS1/EVI1 is a new type of GATA-binding transactivator. Leukemia. 1997;11(3):352–8. doi: 10.1038/sj.leu.2400584.
  74. Laricchia-Robbio L, Fazzina R, Li D, et al. Point mutations in two EVI1 Zn fingers abolish EVI1-GATA1 interaction and allow erythroid differentiation of murine bone marrow cells. Mol Cell Biol. 2006;26(20):7658–66. doi: 10.1128/MCB.00363-06.
  75. Senyuk V, Sinha KK, Li D, et al. Repression of RUNX1 activity by EVI1: a new role of EVI1 in leukemogenesis. Cancer Res. 2007;67(12):5658–66. doi: 10.1158/0008-5472.CAN-06-3962.
  76. Laricchia-Robbio L, Premanand K, Rinaldi CR, Nucifora G. EVI1 Impairs myelopoiesis by deregulation of PU.1 function. Cancer Res. 2009;69(4):1633–42. doi: 10.1158/0008-5472.CAN-08-2562.
  77. Steinmetz B, Hackl H, Slabakova E, et al. The oncogene EVI1 enhances transcriptional and biological responses of human myeloid cells to all-trans retinoic acid. Cell Cycle. 2014;13(18):2931–43. doi: 10.4161/15384101.2014.946869.
  78. Yuasa H, Oike Y, Iwama A, et al. Oncogenic transcription factor Evi1 regulates hematopoietic stem cell proliferation through GATA-2 expression. EMBO J. 2005;24(11):1976–87. doi: 10.1038/sj.emboj.7600679.
  79. Shimabe M, Goyama S, Watanabe-Okochi N, et al. Pbx1 is a downstream target of Evi-1 in hematopoietic stem/progenitors and leukemic cells. Oncogene. 2009;28(49):4364–74. doi: 10.1038/onc.2009.288.
  80. Kurokawa M, Mitani K, Irie K, et al. The oncoprotein Evi-1 represses TGF-beta signalling by inhibiting Smad3. Nature. 1998;394(6688):92–6. doi: 10.1038/27945.
  81. Izutsu K, Kurokawa M, Imai Y, et al. The corepressor CtBP interacts with Evi-1 to repress transforming growth factor beta signaling. Blood. 2001;97(9):2815–22. doi: 10.1182/blood.v97.9.2815.
  82. Kurokawa M, Mitani K, Yamagata T, et al. The evi-1 oncoprotein inhibits c-Jun N-terminal kinase and prevents stress-induced cell death. EMBO J. 2000;19(12):2958–68. doi: 10.1093/emboj/19.12.2958.
  83. Buonamici S, Li D, Mikhail FM, et al. EVI1 abrogates interferon-alpha response by selectively blocking PML induction. J Biol Chem. 2004;280(1):428–36. doi: 10.1074/jbc.M410836200.
  84. Pradhan AK, Mohapatra AD, Nayak KB, Chakraborty S. Acetylation of the proto-oncogene EVI1 abrogates Bcl-xL promoter binding and induces apoptosis. PLoS One. 2011;6(9):e25370. doi: 10.1371/journal.pone.0025370.
  85. Yatsula B, Lin S, Read AJ, et al. Identification of binding sites of EVI1 in mammalian cells. J Biol Chem. 2005;280(35):30712–22. doi: 10.1074/jbc.M504293200.
  86. Ernst T, Chase AJ, Score J, et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet. 2010;42(8):722–6. doi: 10.1038/ng.621.
  87. Figueroa ME, Lugthart S, Li Y, et al. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell. 2010;17(1):13–27. doi: 10.1016/j.ccr.2009.11.020.
  88. Wagner JM, Hackanson B, Lubbert M, Jung M. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenet. 2010;1(3–4):117–36. doi: 10.1007/s13148-010-0012-4.
  89. Senyuk V, Zhang Y, Liu Y, et al. Critical role of miR-9 in myelopoiesis and EVI1-induced leukemogenesis. Proc Natl Acad Sci USA. 2013;110(14):5594–9. doi: 10.1073/pnas.1302645110.
  90. Nikoloski G, Langemeijer SM, Kuiper RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet. 2010;42(8):665–7. doi: 10.1038/ng.620.
  91. Makishima H, Jankowska AM, Tiu RV, et al. Novel homo- and hemizygous mutations in EZH2 in myeloid malignancies. Leukemia. 2010;24(10):1799–804. doi: 10.1038/leu.2010.167.
  92. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33. doi: 10.1056/NEJMoa1005143.
  93. Walter MJ, Ding L, Shen D, et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia. 2011;25(7):1153–8. doi: 10.1038/leu.2011.44.
  94. Delhommeau F, Dupont S, Della Valle V, et al. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360(22):2289–301. doi: 10.1056/NEJMoa0810069.
  95. Langemeijer SM, Kuiper RP, Berends M, et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet. 2009;41(7):838–42. doi: 10.1038/ng.391.
  96. Gelsi-Boyer V, Trouplin V, Adelaide J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800. doi: 10.1111/j.1365-2141.2009.07697.x.
  97. van Haaften G, Dalgliesh GL, Davies H, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41(5):521–3. doi: 10.1038/ng.349.
  98. Liu Y, Chen L, Ko TC, et al. Evi1 is a survival factor which conveys resistance to both TGFbeta- and taxol-mediated cell death via PI3K/AKT. Oncogene. 2006;25(25):3565–75. doi: 10.1038/sj.onc.1209403.
  99. Yoshimi A, Goyama S, Watanabe-Okochi N, et al. Evi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteins. Blood. 2011;117(13):3617–28. doi: 10.1182/blood-2009-12-261602.
  100. Bingemann SC, Konrad TA, Wieser R. Zinc finger transcription factor ecotropic viral integration site 1 is induced by all-trans retinoic acid (ATRA) and acts as a dual modulator of the ATRA response. FEBS J. 2009;276(22):6810–22. doi: 10.1111/j.1742-4658.2009.07398.x.
  101. Pauebelle E, Plesa A, Hayette S, et al. Efficacy of All-Trans-Retinoic Acid in high-risk acute myeloid leukemia with overexpression of EVI1. Oncol Ther. 2019;7(2):121–30. doi: 10.1007/s40487-019-0095-9.
  102. Vazquez I, Maicas M, Cervera J, et al. Down-regulation of EVI1 is associated with epigenetic alterations and good prognosis in patients with acute myeloid leukemia. Haematologica. 2011;96(10):1448–56. doi: 10.3324/haematol.2011. 040535.
  103. Daghistani M, Marin D, Khorashad JS, et al. EVI-1 oncogene expression predicts survival in chronic-phase CML patients resistant to imatinib treated with second-generation tyrosine kinase inhibitors. Blood. 2010;116(26):6014–7. doi: 10.1182/blood-2010-01-264234.
  104. Мамаев Н.Н., Шакирова А.И., Бархатов И.М. идр. Ведущая роль BAALC-экспрессирующих клеток-предшественниц в возникновении и развитии посттрансплантационных рецидивов у больных острыми миелоидными лейкозами. Клиническая онкогематология. 2020;13(1):75–88. doi: 10.21320/2500-2139-2020-13-1-75-88.
    [Mamaev NN, Shakirova AI, Barkhatov IM, et al. Crucial Role of BAALCExpressing Progenitor Cells in Emergence and Development of Post-Transplantation Relapses in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2020;13(1):75–88. doi: 10.21320/2500-2139-2020-13-1-75-88. (In Russ)]
  105. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367(6464):645–8. doi: 10.1038/367645a0.
  106. Matsushita H, Yahata T, Sheng Y, et al. Establishment of a humanized APL model via the transplantation of PML-RARA-transduced human common myeloid progenitors into immunodeficient mice. PLoS One. 2014;9(11):e111082. doi: 10.1371/journal.pone.0111082.
  107. Cole CB, Verdoni AM, Ketkar S, et al. PML-RARA requires DNA methyltransferase 3A to initiate acute promyelocytic leukemia. J Clin Invest. 2016;126(1):85–98. doi: 10.1172/JCI82897.
  108. Гудожникова Я.В., Мамаев Н.Н., Бархатов И.М. и др. Результаты молекулярного мониторинга в посттрансплантационный период с помощью серийного исследования уровня экспрессии гена WT1 у больных острыми миелоидными лейкозами. Клиническая онкогематология. 2018;11(3):241–51. doi: 10.21320/2500-2139-2018-11-3-241-251.
    [Gudozhnikova YaV, Mamaev NN, Barkhatov IM, et al. Results of Molecular Monitoring in Posttransplant Period by Means of Series Investigation of WT1 Gene Expression in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2018;11(3):241–51. doi: 10.21320/2500-2139-2018-11-3-241-251. (In Russ)]
  109. Dreyfus F, Bouscary D, Melle J, et al. Expression of the Evi-1 gene in myelodysplastic syndromes. Leukemia. 1995;9(1):203–5. doi: 10.1016/0145-2126(94)90237-2.
  110. Thol F, Yun H, Sonntag AK, et al. Prognostic significance of combined MN1, ERG, BAALC, and EVI1 (MEBE) expression in patients with myelodysplastic syndromes. Ann Hematol. 2012;91(8):1221–33. doi: 10.1007/s00277-012-1457-7.
  111. Russell M, Thompson F, Spier C, Taetle R. Expression of the EVI1 gene in chronic myelogenous leukemia in blast crisis. Leukemia. 1993;7(10):1654–7.
  112. Ogawa S, Kurokawa M, Tanaka T, et al. Increased Evi-1 expression is frequently observed in blastic crisis of chronic myelocytic leukemia. Leukemia. 1996;10(5):788–94.
  113. Kuila N, Sahoo DP, Kumari M, et al. EVI1, BAALC and AME: prevalence of the secondary mutations in chronic and accelerated phases of chronic myeloid leukemia patients from eastern India. Leuk Res. 2009;33(4):594–6. doi: 10.1016/j.leukres.2008.07.018.
  114. Горбунова А.В., Гиндина Т.Л., Морозова Е.В. и др. Влияние молекулярно-генетических и цитогенетических факторов на эффективность аллогенной трансплантации костного мозга у больных хроническим миелолейкозом. Клиническая онкогематология. 2013;6(4):445–50.
    [Gorbunova AV, Gindina TL, Morozova EV, et al. Impact of molecular genetic and cytogenetic characteristics on outcomes of allogeneic hematopoietic stem cell transplantation in chronic myeloid leukemia. Klinicheskaya oncogematologiya. 2013;6(4):445–50. (In Russ)]
  115. Sato T, Goyama S, Kataoka K, et al. Evi1 defines leukemia-initiating capacity and tyrosine kinase inhibitor resistance in chronic myeloid leukemia. Oncogene. 2014;33(42):5028–38. doi: 10.1038/onc.2014.108.
  116. Konantz M, Andre MC, Ebinger M, et al. EVI-1 modulates leukemogenic potential and apoptosis sensitivity in human acute lymphoblastic leukemia. Leukemia. 2013;27(1):56–65. doi: 10.1038/leu.2012.211.
  117. Mittal N, Li L, Sheng Y, et al. A critical role of epigenetic inactivation of miR-9 in EVI1high pediatric AML. Mol Cancer. 2019;18(1):30. doi: 10.1186/s12943-019-0952-z.
  118. Verhagen HJ, Smit MA, Rutten A, et al. Primary acute myeloid leukemia cells with overexpression of EVI-1 are sensitive to all-trans retinoic acid. Blood. 2016;127(4):458–63. doi: 10.1182/blood-2015-07-653840.
  119. Мамаев Н.Н, Горбунова А.В, Гиндина Т.Л. и др. Стойкое восстановление донорского гемопоэза у больной с посттрансплантационным рецидивом острого миеломонобластного лейкоза с inv(3)(q21q26), моносомией 7 и экспрессией онкогена EVI1 после трансфузий донорских лимфоцитов и использования гипометилирующих агентов. Клиническая онкогематология. 2014;7(1):71–5.
    [Mamayev NN, Gorbunova AV, Gindina TL, et al. Stable donor hematopoiesis reconstitution after post­transplantation relapse of acute myeloid leukemia in patient with inv(3)(q21q26), –7 and EVI1 oncogene overexpression treated by donor lymphocyte infusions and hypomethylating agents. Klinicheskaya oncogematologiya. 2014;7(1):71–5. (In Russ)]
  120. He X, Wang Q, Cen J, et al. Predictive value of high EVI1 expression in AML patients undergoing myeloablative allogeneic hematopoietic stem cell transplantation in first CR. Bone Marrow Transplant. 2016;51(7):921–7. doi: 10.1038/bmt.2016.71.
  121. Мамаев Н.Н., Морозова Е.В., Горбунова А.В. Теоретические и клинические аспекты эпигенетических изменений при миелодиспластических синдромах и острых нелимфобластных лейкозах (обзор литературы). Вестник гематологии. 2011;7(3):12–21.
    [Mamaev NN, Morozova EV, Gorbunova AV. Theoretical and practical aspects of epigenetic changes in myelodysplastic syndromes and acute non-lymphoblastic leukemias (literature review). Vestnik gematologii. 2011;7(3):12–21. (In Russ)]
  122. Mamaev N, Morozova E, Gindina T, et al. Dacogen and allogeneic bone marrow transplantation in the treatment of high-risk myelodysplastic syndromes with non-random chromosome abnormalities. Leuk Res. 2011;35(Suppl 1):72–3. doi: 10.1016/S0145-2126(11)70186-2.
  123. Mamaev N, Gorbunova A, Barkhatov I, et al. Biology and treatment of leukemia and myelodysplastic syndromes with high EVI-1 gene expression. ELN Frontiers Meeting 2012 “Myeloid neoplasms: approaching cure”. Istanbul, Turkey. Abstract No. 37.
  124. Yang X, Wong MPM, Ng RK. Aberrant DNA Methylation in Acute Myeloid Leukemia and Its Clinical Implications. Int J Mol Sci. 2019;20(18):4576. doi: 10.3390/ijms20184576.
  125. Nowek K, Sun SM, Dijkstra MK, et al. Expression of a passenger miR-9* predicts favorable outcome in adults with acute myeloid leukemia less than 60 years of age. Leukemia. 2016;30(2):303–9. doi: 10.1038/leu.2015.282.
  126. Li F, He W, Geng R, Xie X. Myeloid leukemia with high EVI1 expression is sensitive to 5-aza-2’-deoxycytidine by targeting miR-9. Clin Transl Oncol. 2020;22(1):137–43. doi: 10.1007/s12094-019-02121-y.
  127. Cattaneo F, Nucifora G. EVI1 recruits the histone methyltransferase SUV39H1 for transcription repression. J Cell Biochem. 2008;105(2):344–52. doi: 10.1002/jcb.21869.
  128. Craddock C, Quek L, Goardon N, et al. Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia. Leukemia. 2013;27(5):1028–36. doi: 10.1038/leu.2012.312.
  129. Trino S, Zoppoli P, Carella AM, et al. DNA methylation dynamic of bone marrow hematopoietic stem cells after allogeneic transplantation. Stem Cell Res Ther. 2019;10(1):138. doi: 10.1186/s13287-019-1245-6.
  130. Ahn JS, Kim YK, Min YH, et al. Azacitidine Pre-Treatment Followed by Reduced-Intensity Stem Cell Transplantation in Patients with Higher-Risk Myelodysplastic Syndrome. Acta Haematol. 2015;134(1):40–8. doi: 10.1159/000368711.
  131. Voso MT, Leone G, Piciocchi A, et al. Feasibility of allogeneic stem-cell transplantation after azacitidine bridge in higher-risk myelodysplastic syndromes and low blast count acute myeloid leukemia: results of the BMT-AZA prospective study. Ann Oncol. 2017;28(7):1547–53. doi: 10.1093/annonc/mdx154.
  132. Овечкина В.Н., Бондаренко С.Н., Морозова Е.В. и др. Роль терапии гипометилирующими препаратами перед аллогенной трансплантацией гемопоэтических стволовых клеток при острых миелоидных лейкозах и миелодиспластическом синдроме. Клиническая онкогематология. 2017;10(3):351–7. doi: 10.21320/2500-2139-2017-10-3-351-357.
    [Ovechkina VN, Bondarenko SN, Morozova EV, et al. The Role of Hypomethylating Agents Prior to Allogeneic Hematopoietic Stem Cells Transplantation in Acute Myeloid Leukemia and Myelodysplastic Syndrome. Clinical oncohematology. 2017;10(3):351–7. doi: 10.21320/2500-2139-2017-10-3-351-357. (In Russ)]
  133. Nishihori T, Perkins J, Mishra A, et al. Pretransplantation 5-azacitidine in high-risk myelodysplastic syndrome. Biol Blood Marrow Transplant. 2014;20(6):776–80. doi: 10.1016/j.bbmt.2014.02.008.
  134. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116(23):5420–31. doi: 10.1002/cncr.25500.
  135. Craddock C, Jilani N, Siddique S, et al. Tolerability and Clinical Activity of Post-Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on the RICAZA Trial. Biol Blood Marrow Transplant. 2016;22(2):385–90. doi: 10.1016/j.bbmt.2015.09.004.
  136. Marini C, Brissot E, Bazarbachi A, et al. Tolerability and Efficacy of Treatment With Azacytidine as Prophylactic or Preemptive Therapy for Myeloid Neoplasms After Allogeneic Stem Cell Transplantation. Clin Lymphoma Myel Leuk. 2020;20(6):377–82. doi: 10.1016/j.clml.2019.10.011.
  137. Бадаев Р.Ш., Заммоева Д.Б., Гиршова Л.Л. и др. Профилактическое применение азацитидина у пациентов с острыми миелоидными лейкозами после гаплоидентичной аллоТКМ. Клиническая онкогематология. 2019;12(1):37–42. doi: 10.21320/2500-2139-2019-12-1-37-42.
    [Badaev RSh, Zammoeva DB, Girshova LL, et al. Preventive Use of Azacitidine in Patients with Acute Myeloid Leukemia after Haploidentical Allo-BMT. Clinical oncohematology. 2019;12(1):37–42. doi: 10.21320/2500-2139-2019-12-1-37-42. (In Russ)]
  138. Cattaneo F, Nucifora G. EVI1 recruits the histone methyltransferase SUV39H1 for transcription repression. J Cell Biochem. 2008;105(2):344–52. doi: 10.1002/jcb.21869.
  139. Estey EH, Thall PF, Pierce S, et al. Randomized phase II study of fludarabine + cytosine arabinoside + idarubicin ± all-trans retinoic acid ± granulocyte colony-stimulating factor in poor prognosis newly diagnosed acute myeloid leukemia and myelodysplastic syndrome. Blood. 1999;93(8):2478–84. doi: 10.1182/blood.v93.8.2478.
  140. Schlenk RF, Frohling S, Hartmann F, et al. Phase III study of all-trans retinoic acid in previously untreated patients 61 years or older with acute myeloid leukemia. Leukemia. 2004;18(11):1798–803. doi: 10.1038/sj.leu.2403528.
  141. Raza A, Buonamici S, Lisak L, et al. Arsenic trioxide and thalidomide combination produces multi-lineage hematological responses in myelodysplastic syndromes patients, particularly in those with high pre-therapy EVI1 expression. Leuk Res. 2004;28(8):791–803. doi: 10.1016/j.leukres.2003.11.018.
  142. Burnett AK, Hills RK, Green C, et al. The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA. Blood. 2010;115(5):948–56. doi: 10.1182/blood-2009-08-236588.
  143. van Gils N, Verhagen HJMP, Smit L. Reprogramming acute myeloid leukemia into sensitivity for retinoic-acid-driven differentiation. Exp Hematol. 2017;52:12–23. doi: 10.1016/j.exphem.2017.04.007.
  144. Plesa A, Dumontet C, Mattei E, et al. High frequency of CD34+CD38-/low immature leukemia cells is correlated with unfavorable prognosis in acute myeloid leukemia. World J Stem Cells. 2017;9(12):227–34. doi: 10.4252/wjsc.v9.i12.227.
  145. Nguyen CH, Bauer K, Hackl H, et al. All-trans retinoic acid enhances, and a pan-RAR antagonist counteracts, the stem cell promoting activity of EVI1 in acute myeloid leukemia. Cell Death Dis. 2019;10(12):944. doi: 10.1038/s41419-019-2172-2.
  146. Field T, Perkins J, Huang Y, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  147. Kim DY, Lee JH, Park YH, et al. Feasibility of hypomethylating agents followed by allogeneic hematopoietic cell transplantation in patients with myelodysplastic syndrome. Bone Marrow Transplant. 2012;47(3):374–9. doi: 10.1038/bmt.2011.86.
  148. Jiang YZ, Su GP, Dai Y, et al. Effect of Decitabine Combined with Unrelated Cord Blood Transplantation in an Adult Patient with -7/EVI1+ Acute Myeloid Leukemia: a Case Report and Literature Review. Ann Clin Lab Sci. 2015;45(5):598–601.
  149. Schlenk RF, Lubbert M, Benner A, et al. All-trans retinoic acid as adjunct to intensive treatment in younger adult patients with acute myeloid leukemia: results of the randomized AMLSG 07-04 study. Ann Hematol. 2016;95(12):1931–42. doi: 10.1007/s00277-016-2810-z.
  150. Taussig DC, Vargaftig J, Miraki-Moud F, et al. Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood. 2010;115(10):1976–84. doi: 10.1182/blood-2009-02-206565.
  151. Patel S, Zhang Y, Cassinat B, et al. Successful xenografts of AML3 samples in immunodeficient NOD/shi-SCID IL2Rγ–/– Leukemia. 2012;26(11):2432–5. doi: 10.1038/leu.2012.154.

Factors for Sustaining Molecular Remission after Discontinuation of Tyrosine Kinase Inhibitors Therapy in Chronic Myeloid Leukemia: Results of Non-Randomized Prospective Clinical Trial

OA Shukhov, AN Petrova, EYu Chelysheva, AV Bykova, IS Nemchenko, AG Turkina

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

For correspondence: Oleg Aleksandrovich Shukhov, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-16-36, +7(985)287-12-69; e-mail: shuhov@list.ru

For citation: Shukhov OA, Petrova AN, Chelysheva EYu, et al. Factors for Sustaining Molecular Remission after Discontinuation of Tyrosine Kinase Inhibitors Therapy in Chronic Myeloid Leukemia: Results of Non-Randomized Prospective Clinical Trial. Clinical oncohematology. 2021;14(1):1–12. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-1-12


ABSTRACT

Aim. To study the impact of different clinical and biological factors on sustaining molecular remission after discontinuation of tyrosine kinase inhibitors (TKI) therapy in chronic myeloid leukemia (CML) patients with a stable deep molecular response (MR).

Materials & Methods. The prospective multi-center trial on molecular remission sustainability after TKIs withdrawal, held from 2015 to 2019, enrolled 98 CML patients. The trial included patients with chronic phase CML treated with TKIs at least during 3 years and having a stable deep MR (≤ МО4; BCR-ABL < 0.01 %) during at least 2 years. Molecular monitoring was carried out every month during first 6 months after TKIs withdrawal, every 2 months during 0.5–1 year, and every 3 months after 1-year follow-up. In case of the loss of major MR (BCR-ABL > 0.1 %) therapy was reinitiated.

Results. Three-year molecular relapse-free survival was 51 % (95% confidence interval 41–61 %) in all patients, 25 % in patients with the failure of prior treatment discontinuation, and 53 % in patients who discontinued TKI therapy for the first time. According to univariate analysis, the following factors proved to be significant: persistance of deep MR, duration of therapy, and depth of MR. It was shown that TKI therapy duration, but not deep MR persistance, has independent prognostic value for the Russian population of CML patients. No significant differences were identified in 3-year molecular relapse-free survival in the groups of patients treated only with imatinib (55 %) compared with patients who received 2nd generation TKI (TKI2) as first-line (70 %; = 0.26) and second-line (39 %; = 0.09) therapy. However, duration of therapy in patients treated with TKI2 as first-line therapy was more than twice as short as in patients treated with imatinib as first-line therapy (median 41.5 vs. 96.4 months, respectively; < 0.0001).

Conclusion. Longer therapy duration and MR depth (≤ MО4.5) before TKI withdrawal raise the probability of sustaining off-treatment remission. The study showed that molecular relapse-free survival does not significantly increase with the use of TKI2 as first-line treatment compared to imatinib. Nevertheless, TKI2 as first-line treatment enables to halve the duration of therapy needed to achieve comparable molecular relapse-free survival, as compared with imatinib.

Keywords: chronic myeloid leukemia, tyrosine kinase inhibitors, deep molecular response, off-treatment remission.

Received: July 30, 2020

Accepted: December 1, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Deininger MW, Goldman JM, Melo JV. The molecular biology of chronic myeloid leukemia. Blood. 2000;96(10):3343–56.
  2. Branford S, Kim DDH, Apperley JF, et al. Laying the foundation for genomically-based risk assessment in chronic myeloid leukemia. Leukemia. 2019;33(8):1835–50. doi: 10.1038/s41375-019-0512-y.
  3. Swerdlow SH. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: IARC Press; 2017.
  4. Druker BJ, Guilhot F, O’Brien SG, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355(23):2408–17. doi: 10.1056/NEJMoa062867.
  5. Saussele S, Krauss MP, Hehlmann R, et al. Impact of comorbidities on overall survival in patients with chronic myeloid leukemia: results of the randomized CML study IV. Blood. 2015;126(1):42–9. doi: 10.1182/blood-2015-01-617993.
  6. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.
  7. Cortes JE, Saglio G, Kantarjian HM, et al. Final 5-Year Study Results of DASISION: The Dasatinib Versus Imatinib Study in Treatment-Naive Chronic Myeloid Leukemia Patients Trial. J Clin Oncol. 2016;34(20):2333–40. doi: 10.1200/JCO.2015.64.8899.
  8. Lipton JH, Chuah C, Guerci-Bresler A, et al. Ponatinib versus imatinib for newly diagnosed chronic myeloid leukaemia: an international, randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17(5):612–21. doi: 10.1016/S1470-2045(16)00080-2.
  9. Hochhaus A, Larson RA, Guilhot F, et al. Long-Term Outcomes of Imatinib Treatment for Chronic Myeloid Leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.
  10. Goldman J, Gordon M. Why do chronic myelogenous leukemia stem cells survive allogeneic stem cell transplantation or imatinib: does it really matter?. Leuk Lymphoma. 2006;47(1):1–7. doi: 10.1080/10428190500407996.
  11. Baccarani M, Castagnetti F, Gugliotta G, et al. The proportion of different BCR-ABL1 transcript types in chronic myeloid leukemia. An international overview. Leukemia. 2019;33(5):1173–83. doi: 10.1038/s41375-018-0341-4.
  12. Hughes T, Deininger M, Hochhaus A, et al. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood. 2006;108(1):28–37. doi: 10.1182/blood-2006-01-0092.
  13. Branford S, Seymour JF, Grigg A, et al. BCR-ABL messenger RNA levels continue to decline in patients with chronic phase chronic myeloid leukemia treated with imatinib for more than 5 years and approximately half of all first-line treated patients have stable undetectable BCR-ABL using strict sensitivity criteria. Clin Cancer Res. 2007;13(23):7080–5. doi: 10.1158/1078-0432.CCR-07-0844.
  14. Branford S, Cross NC, Hochhaus A, et al. Rationale for the recommendations for harmonizing current methodology for detecting BCR-ABL transcripts in patients with chronic myeloid leukaemia. Leukemia. 2006;20(11):1925–30. doi: 10.1038/sj.leu.2404388.
  15. Cross NC, White HE, Colomer D, et al. Laboratory recommendations for scoring deep molecular responses following treatment for chronic myeloid leukemia. Leukemia. 2015;29(5):999–1003. doi: 10.1038/leu.2015.29.
  16. Hehlmann R, Muller MC, Lauseker M, et al. Deep molecular response is reached by the majority of patients treated with imatinib, predicts survival, and is achieved more quickly by optimized high-dose imatinib: results from the randomized CML-study IV. J Clin Oncol. 2014;32(5):415–23. doi: 10.1200/JCO.2013.49.9020.
  17. Rousselot P, Huguet F, Rea D, et al. Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years. Blood. 2007;109(1):58–60. doi: 10.1182/blood-2006-03-011239.
  18. NCCN Clinical Practice Guidelines in Oncology. Chronic Myeloid Leukemia. Version 3.2020. Available from: https://www.nccn.org/professionals/physician_gls/pdf/aml_blocks.pdf. (accessed 23.10.2020).
  19. Hochhaus A, Saussele S, Rosti G, et al. Chronic myeloid leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(Suppl 4):iv41–iv51. doi: 10.1093/annonc/mdx219.
  20. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. Blood. 2016;128(1):17–23. doi: 10.1182/blood-2016-01-694265.
  21. Mahon FX, Rea D, Guilhot J, et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 2010;11(11):1029–35. doi: 10.1016/S1470-2045(10)70233-3.
  22. Etienne G, Guilhot J, Rea D, et al. Long-Term Follow-Up of the French Stop Imatinib (STIM1) Study in Patients With Chronic Myeloid Leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/JCO.2016.68.2914.
  23. Ross DM, Branford S, Seymour JF, et al. Safety and efficacy of imatinib cessation for CML patients with stable undetectable minimal residual disease: results from the TWISTER study. Blood. 2013;122(4):515–22. doi: 10.1182/blood-2013-02-483750.
  24. Ross DM, Pagani IS, Shanmuganathan N, et al. Long-term treatment-free remission of chronic myeloid leukemia with falling levels of residual leukemic cells. Leukemia. 2018;32(12):2572–9. doi: 10.1038/s41375-018-0264-0.
  25. Rousselot P, Charbonnier A, Cony-Makhoul P, et al. Loss of major molecular response as a trigger for restarting tyrosine kinase inhibitor therapy in patients with chronic-phase chronic myelogenous leukemia who have stopped imatinib after durable undetectable disease. J Clin Oncol. 2014;32(5):424–30. doi: 10.1200/JCO.2012.48.5797.
  26. Mori S, Vagge E, le Coutre P, et al. Age and dPCR can predict relapse in CML patients who discontinued imatinib: the ISAV study. Am J Hematol. 2015;90(10):910–4. doi: 10.1002/ajh.24120.
  27. Takahashi N, Tauchi T, Kitamura K, et al. Deeper molecular response is a predictive factor for treatment-free remission after imatinib discontinuation in patients with chronic phase chronic myeloid leukemia: the JALSG-STIM213 study. Int J Hematol. 2018;107(2):185–93. doi: 10.1007/s12185-017-2334-x.
  28. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor dose in patients with chronic myeloid leukaemia with stable major molecular response (DESTINY): an interim analysis of a non-randomised, phase 2 trial. Lancet Haematol. 2017;4(7):e310–e316. doi: 10.1016/S2352-3026(17)30066-
  29. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor therapy before complete treatment discontinuation in patients with chronic myeloid leukaemia (DESTINY): a non-randomised, phase 2 trial. Lancet Haematol. 2019;6(7):e375–e383. doi: 10.1016/S2352-3026(19)30094-8.
  30. Lee SE, Choi SY, Song HY, et al. Imatinib withdrawal syndrome and longer duration of imatinib have a close association with a lower molecular relapse after treatment discontinuation: the KID study. Haematologica. 2016;101(6):717–23. doi: 10.3324/haematol.2015.139899.
  31. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.
  32. Claudiani S, Apperley JF, Gale RP, et al. E14a2 BCR-ABL1 transcript is associated with a higher rate of treatment-free remission in individuals with chronic myeloid leukemia after stopping tyrosine kinase inhibitor therapy. Haematologica. 2017;102(8):e297–e299. doi: 10.3324/haematol.2017.168740.
  33. Sokal JE, Cox EB, Baccarani M, et al. Prognostic discrimination in “good-risk” chronic granulocytic leukemia. Blood. 1984;63(4):789–99. doi: 10.1182/blood.V63.4.789.789.
  34. Nicolini FE, Dulucq S, Boureau L, et al. Evaluation of Residual Disease and TKI Duration Are Critical Predictive Factors for Molecular Recurrence after Stopping Imatinib First-line in Chronic Phase CML Patients. Clin Cancer Res. 2019;25(22):6606–13. doi: 10.1158/1078-0432.CCR-18-3373.
  35. Kumagai T, Nakaseko C, Nishiwaki K, et al. Dasatinib cessation after deep molecular response exceeding 2 years and natural killer cell transition during dasatinib consolidation. Cancer Sci. 2018;109(1):182–92. doi: 10.1111/cas.13430.
  36. Takahashi N, Nishiwaki K, Nakaseko C, et al. Treatment-free remission after two-year consolidation therapy with nilotinib in patients with chronic myeloid leukemia: STAT2 trial in Japan. Haematologica. 2018;103(11):1835–42. doi: 10.3324/haematol.2018.194894.
  37. Benjamini O, Kantarjian H, Rios MB, et al. Patient-driven discontinuation of tyrosine kinase inhibitors: single institution experience. Leuk Lymphoma. 2014;55(12):2879–86. doi: 10.3109/10428194.2013.831092.
  38. Hochhaus A, Masszi T, Giles FJ, et al. Treatment-free remission following frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the ENESTfreedom study. Leukemia. 2017;31(7):1525–31. doi: 10.1038/leu.2017.63.
  39. Ross DM, Masszi T, Casares GMT, et al. Durable treatment-free remission in patients with chronic myeloid leukemia in chronic phase following frontline nilotinib: 96-week update of the ENESTfreedom study. J Cancer Res Clin Oncol. 2018;144(5):945–54. doi: 10.1007/s00432-018-2604-x.
  40. Mahon FX, Boquimpani C, Kim DW, et al. Treatment-Free Remission After Second-Line Nilotinib Treatment in Patients With Chronic Myeloid Leukemia in Chronic Phase: Results From a Single-Group, Phase 2, Open-Label Study. Ann Intern Med. 2018;168(7):461–70. doi: 10.7326/M17-1094.
  41. Kimura S, Imagawa J, Murai K, et al. Treatment-free remission after first-line dasatinib discontinuation in patients with chronic myeloid leukaemia (first-line DADI trial): a single-arm, multicentre, phase 2 trial. Lancet Haematol. 2020;7(3):e218–e225. doi: 10.1016/S2352-3026(19)30235-2.
  42. Imagawa J, Tanaka H, Okada M, et al. Discontinuation of dasatinib in patients with chronic myeloid leukaemia who have maintained deep molecular response for longer than 1 year (DADI trial): a multicentre phase 2 trial. Lancet Haematol. 2015;2(12):e528–e535. doi: 10.1016/s2352-3026(15)00196-9.
  43. Okada M, Imagawa J, Tanaka H, et al. Final 3-year Results of the Dasatinib Discontinuation Trial in Patients With Chronic Myeloid Leukemia Who Received Dasatinib as a Second-line Treatment. Clin Lymphoma Myel Leuk. 2018;18(5):353–60.e1. doi: 10.1016/j.clml.2018.03.004.
  44. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.
  45. Baccarani M, Saglio G, Goldman J, et al. Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2006;108(6):1809–20. doi: 10.1182/blood-2006-02-005686.
  46. Baccarani M, Cortes J, Pane F, et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. J Clin Oncol. 2009;27(35):6041–51. doi: 10.1200/JCO.2009.25.0779.
  47. 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.
  48. Radich JP, Hochhaus A, Giles FJ, et al. Analyses of Predictors of Durable Treatment-Free Remission in Patients with Chronic Myeloid Leukemia in Chronic Phase Following Frontline or Second-Line Nilotinib. 2019;134(Suppl_1):2932. doi: 10.1182/blood-2019-129393.
  49. D’Adda M, Farina M, Schieppati F, et al. The e13a2 BCR-ABL transcript negatively affects sustained deep molecular response and the achievement of treatment-free remission in patients with chronic myeloid leukemia who receive tyrosine kinase inhibitors. Cancer. 2019;125(10):1674–82. doi: 10.1002/cncr.31977.
  50. Legros L, Nicolini FE, Etienne G, et al. Second tyrosine kinase inhibitor discontinuation attempt in patients with chronic myeloid leukemia. Cancer. 2017;123(22):4403–10. doi: 10.1002/cncr.30885.
  51. Shih YT, Cortes JE, Kantarjian HM. Treatment value of second-generation BCR-ABL1 tyrosine kinase inhibitors compared with imatinib to achieve treatment-free remission in patients with chronic myeloid leukaemia: a modelling study. Lancet Haematol. 2019;6(8):e398–e408. doi: 10.1016/S2352-3026(19)30087-0.
  52. Шуваев В.А., Абдулкадыров К.М., МартынкевичИ.С., Фоминых М.С. Выбор терапии первой линии хронического миелолейкоза: моделирование клинико-экономических факторов. Клиническая онкогематология. 2015;8(1):78–83. doi: 10.21320/2500-2139-2015-8-1-78-83.
    [Shuvaev VA, Abdulkadyrov KM, Martynkevich IS, Fominykh MS. First Line Treatment Choice for Chronic Myelogenous Leukemia: Modeling of Clinical and Economic Factors. Clinical oncohematology. 2015;8(1):78–83. doi: 10.21320/2500-2139-2015-8-1-78-83. (In Russ)]
  53. Рубрикатор клинических рекомендаций. Хронический миелолейкоз [электронный документ]. Доступно по: http://cr.rosminzdrav.ru/#!/recomend/120. Ссылка активна на 23.10.2020.
    [List of clinical guidelines. Chronic myeloid leukemia. [Internet] Available from: http://cr.rosminzdrav.ru/#!/recomend/120. (accessed 10.2020) (In Russ)]
  54. Hochhaus A, Baccarani M, Silver RT, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 2020;34(4):966–84. doi: 10.1038/s41375-020-0776-

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

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

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

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

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

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


ABSTRACT

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

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

Received: November 20, 2019

Accepted: February 28, 2020

Read in PDF


REFERENCES

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

Comparative Analysis of Cardiovascular Disorders in Patients with Chronic Myeloid Leukemia on Tyrosine Kinase Inhibitor Therapy

LM Makeeva1, EI Emelina1, AV Bykova2, GE Gendlin1, GA Gusarova2, IG Nikitin1, EYu Chelysheva2, OYu Vinogradova1,3,4, IE Lazarev3, EG Arshanskaya3, AG Turkina2

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

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

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

4 Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, 1 Samory Mashela str., Moscow, Russian Federation, 117997

For correspondence: Prof. Gennadii Efimovich Gendlin, MD, PhD, 1 Ostrovityanova str., Moscow, Russian Federation, 117997; e-mail: rgmugt2@mail.ru

For citation: Makeeva LM, Emelina EI, Bykova AV, et al. Comparative Analysis of Cardiovascular Disorders in Patients with Chronic Myeloid Leukemia on Tyrosine Kinase Inhibitor Therapy. Clinical oncohematology. 2020;13(1):104–111 (In Russ).

DOI: 10.21320/2500-2139-2020-13-1-104-111


ABSTRACT

Aim. To analyze adverse cardiovascular events in chronic myeloid leukemia (CML) patients who received various tyrosine kinase inhibitors (TKI).

Materials & Methods. The trial included 97 CML patients with nilotinib, dasatinib or imatinib indications. By the time of examination the patients had undergone TKI therapy for 1–138 months. The three of them were sequentially treated with 2 drugs over the monitoring period. All CML patients were aged 22–79 years (median 53.5 years): 55 women were aged 22–71 years (median 53.5 years) and 42 men were aged 24–79 years (median 53 years).

Results. The comparative analysis demonstrated significantly higher impact of nilotinib on QTc duration compared with other TKIs. The patients who received nilotinib (n = 15) throughout 38 months had QTc of 0.47 s (interquartile range [IQR] 0.46–0.47 s), in imatinib group (n = 17) QTc was 0.43 s (IQR 0.43–0.44 s), and in dasatinib group (n = 4) QTc was 0.43 s (IQR 0.42–0.44 s) (= 0.0008). Among all patients treated with nilotinib there were 62 % (31/50) with QTc > 0.46 s, in imatinib (6/41) and dasatinib (2/18) groups it was detected in 14.6 % and 11.1 % of patients, respectively (= 0.0008). Five patients had QTc > 0.48 s, which is the criterion for discontinuation of treatment or dose reduction. In two patients the identified changes of QTc duration required TKI temporary suspension. After nilotinib dose reduction or discontinuation QTc duration normalized in all cases within 2 weeks. Decreased ankle-brachial index (ABI) < 0.9 without pronounced clinical symptoms was identified in two patients who received nilotinib. Afterwards they showed peripheral occlusive disease of lower extremities, and nilotinib treatment was discontinued. In patients treated with other TKIs no occlusive vascular lesions were observed. A case of chronic heart failure with reduced left ventricular ejection fraction developing on nilotinib therapy was revealed and described.

Conclusion. Despite high specificity for BCR-ABL tyrosine kinase, new TKIs can, although rarely, induce cardiovascular adverse events. Prior to TKI treatment assignment CML patients should be examined with ECG and EchoCG with systolic function evaluation, and the measurement of pulmonary artery pressure as well as ABI. The examination should be repeated in the end of the 1st year TKI treatment if there is no reason for extra examinations. It is recommended to hold 24-hour ECG monitoring with QTc max measurement prior to nilotinib assignment, then once a year within 2 years of nilotinib treatment, and once in 6 months after 3 years of therapy.

Keywords: imatinib, dasatinib, nilotinib, chronic myeloid leukemia, QTc prolongation, sudden cardiac death, peripheral occlusive disease of lower extremities, chronic heart failure, cardiomyopathy, pulmonary arterial hypertension.

Received: September 8, 2019

Accepted: December 21, 2019

Read in PDF


REFERENCES

  1. O’Brien SG, Guilhot F, Larson RA, 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.

  2. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.

  3. Aghel N, Delgado DH, Lipton JH. Cardiovascular toxicities of BCR-ABL tyrosine kinase inhibitors in chronic myeloid leukemia: preventive strategies and cardiovascular surveillance. Vasc Health Risk Manage. 2017;13:293–303. doi: 10.2147/VHRM.S108874.

  4. Shah AM, Campbell P, Rocha GQ, et al. Effect of imatinib as add-on therapy on echocardiographic measures of right ventricular function in patients with significant pulmonary arterial hypertension. Eur Heart J. 2015;36(10):623–32. doi: 10.1093/eurheartj/ehu035.

  5. Galie N, Humbert M, Vachiery JL, et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Heart J. 2016;37(1):67–119. doi: 10.1093/eurheartj/ehv317.

  6. Valent P, Hadzijusufovic E, Hoermann G, et al. Risk factors and mechanisms contributing to TKI-induced vascular events in patients with CML. Leuk Res. 2017;59:47–54. doi: 10.1016/j.leukres.2017.05.008.

  7. Barber MC, Mauro MJ, Moslehi J. Cardiovascular care of patients with chronic myeloid leukemia (CML) on tyrosine kinase inhibitor (TKI) therapy. Hematology Am Soc Hematol Educ Program. 2017;2017(1):110–4. doi: 10.1182/asheducation-2017.1.110.

  8. Ross DM, Arthur C, Burbury K, et al. Chronic myeloid leukaemia and tyrosine kinase inhibitor therapy: assessment and management of cardiovascular risk factors. Intern Med J. 2018;48(Suppl 2):5–13. doi: 10.1111/imj.13716.

  9. Dahlen T, Edgren G, Lambe M, et al. Cardiovascular events associated with use of tyrosine kinase inhibitors in chronic myeloid leukemia: A population-based cohort study. Ann Intern Med. 2016;165(3):161–6. doi: 10.7326/M15-2306.

  10. Chai-Adisaksopha C, Lam W, Hillis C. Major arterial events in patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors: a meta-analysis. Leuk Lymphoma. 2016;57(6):1300–10. doi: 10.3109/10428194.2015.1091929.

  11. Aghel N, Lipton JH, Atenafu EG, et al. Cardiovascular Events After Exposure to Nilotinib in Chronic Myeloid Leukemia: Long-term Follow-up. Clin Lymph Myel Leuk. 2017;17(12):870–8. doi: 10.1016/j.clml.2017.07.006.

  12. Pasvolsky O, Leader A, Iakobishvili Z, et al. Tyrosine kinase inhibitor associated vascular toxicity in chronic myeloid leukemia. Cardio-Oncol. 2015;1(1):5. doi: 10.1186/s40959-015-0008-5.

  13. Steegmann JL, Baccarani M, Breccia M, et al. Recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia. 2016;30(8):1648–71. doi: 10.1038/leu.2016.104.

  14. 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)]

  15. Porta-Sanchez A, Gilbert C, Spears D, et al. Incidence, Diagnosis, and Management of QT Prolongation Induced by Cancer Therapies: A Systematic Review. J Am Heart Assoc. 2017;6(12):e007724. doi: 10.1161/JAHA.117.007724.

  16. Cheng Y-J, Nie X-Y, Chen X-M, et al. The Role of Macrolide Antibiotics in Increasing Cardiovascular Risk. J Am College Cardiol. 2015;66(20):2173–84. doi: 10.1016/j.jacc.2015.09.029.

  17. Объединенная рабочая группа. Национальные российские рекомендации по применению методики холтеровского мониторирования в клинической практике. Российский кардиологический журнал. 2014;2(106):6–71.

    [The Joint Task Force. National Russian guidelines on application of the methods of Holter monitoring in clinical practice. Rossiiskii kardiologicheskii zhurnal. 2014;2(106):6–71. (In Russ)]

  18. Piepoli MF, Hoes AW, Agewall S, et al. 2016 European Guidelines on cardiovascular disease prevention in clinical practice. Eur Heart J. 2016;37(29):2315–81. doi: 10.1093/eurheartj/ehw106.

  19. Guirguis-Blake JM, Evans CV, Redmond N, et al. Screening for Peripheral Artery Disease Using the Ankle-Brachial Index: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2018;320(2):184–96. doi: 10.1001/jama.2018.4250.

  20. O’Neal WT, Singleton MJ, Roberts JD, et al. Association Between QT-Interval Components and Sudden Cardiac Death. Circ Arrhythm Electrophysiol. 2017;10(10):e005485. doi: 10.1161/CIRCEP.117.005485.

  21. PrTASIGNA®. Product monograph. Available from: https://www.novartis.ca/sites/www.novartis.ca/files/tasigna_scrip_e.pdf (accessed 10.12.2019).

  22. Hadzijusufovic E, Albrecht-Schgoer K, Hoermann G, et al. Nilotinib-induced vasculopathy: identification of vascular endothelial cells as a primary target site. Leukemia. 2017;31(11):2388–97. doi: 10.1038/leu.2017.245.

  23. Gora-Tybor J, Medras E, Calbecka M, et al. Real-life comparison of severe vascular events and other non-hematological complications in patients with chronic myeloid leukemia undergoing second-line nilotinib or dasatinib treatment. Leuk Lymphoma. 2015;56(8):2309–14. doi: 10.3109/10428194.2014.994205.

  24. Kim TD, Rea D, Schwarz M, et al. Peripheral artery occlusive disease in chronic phase chronic myeloid leukemia patients treated with nilotinib or imatinib. Leukemia. 2013;27(6):1316–21. doi: 10.1038/leu.2013.70.

  25. Catapano AL, Graham I, De Backer G, et al. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Eur Heart J. 2016;37(39):2999–3058. doi: 10.1093/eurheartj/ehw272.

  26. Kim TD, le Coutre P, Schwarz M, et al. Clinical cardiac safety profile of nilotinib. Haematologica. 2012;97(6):883–9. doi: 13324/haematol.2011.058776.

  27. Xu Z, Cang S, Yang N, Liu D. Cardiotoxicity of tyrosine kinase inhibitors in chronic myelogenous leukemia therapy. Hematol Rev. 2009;1(1):e4. doi: 10.4081/hr.2009.e4.

  28. Gurguis C, de Armas RL, Kantarjian HM. Echocardiographic Findings in Patients (pts) Receiving Tyrosine Kinase Inhibitors (TKIs) for the Treatment of Chronic Myeloid Leukemia (CML). Blood. 2017;130(Suppl 1):2893.

  29. Larsen C.M, Mulvagh S.L. Cardio-oncology: what you need to know now for clinical practice and echocardiography. Echo Res Pract. 2017;4(1):R33–R41. doi: 10.1530/ERP-17-0013.

Management of Chronic Myeloid Leukemia Patients During Pregnancy (Analysis of Literature and Practical Recommendations)

EYu Chelysheva1, AG Turkina1, ES Polushkina2, MA Vinogradova2, RG Shmakov2

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

2 VI Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 4 Akademika Oparina str., Moscow, Russian Federation, 117997

For correspondence: Ekaterina Yur’evna Chelysheva, MD, PhD, 4a Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-48-60; e-mail: denve@bk.ru

For citation: Chelysheva EYu, Turkina AG, Polushkina ES, et al. Management of Chronic Myeloid Leukemia Patients During Pregnancy (Analysis of Literature and Clinical Experience). Clinical oncohematology. 2019;12(2):202–10.

DOI: 10.21320/2500-2139-2019-12-2-202-210


ABSTRACT

Background. The tyrosine kinase inhibitors (TKI) era is marked by a long-term favorable prognosis of chronic myeloid leukemia (CML). In this context CML patients of reproductive age are faced with major issues of family planning with due regard to the risk of TKI treatment interruption during pregnancy. Additionally, TKI impact is another potential risk to the fetus.

Aim. To develop differentiated approach to CML treatment during pregnancy.

Materials & Methods. Analysis includes literature data and clinical experience based on 166 pregnancies of 120 CML patients from CML Pregnancy Registry.

Results. Pregnancy planning is recommended after achieving stable and deep molecular response (with BCR-ABL > 0.01 %, IS) within the period of at least 2 years. At conception TKI therapy does not have to be interrupted. However, early pregnancy detection and TKI treatment interruption after pregnancy confirmation are of vital importance due to teratogenic risks. Furthermore, no TKI may be administered during organogenetic period, i.e. up to the 15th week of gestation. In the absence or loss of complete hematologic response and growth of BCR-ABL > 1 % after the 15th week of gestation imatinib or nilotinib administration is justified in the interest of pregnant patients taking into account limited transfer of these drugs through placenta. In the absence of complete hematologic response before the 15th week of gestation interferon-α can be administered. With BCR-ABL < 1 % patients can be either followed-up without therapy or they can receive interferon-α throughout pregnancy. Dasatinib, bosutinib, and other TKI are contraindicated at any stage of pregnancy. There are no special recommendations for childbirth, delivery is to be adapted to obstetric conditions. Breast feeding is not recommended because of the lack of practical evidence for its safety.

Conclusion. A regular molecular monitoring of BCR-ABL and hematologic status is indispensable, health condition of fetus should be continuously monitored as well. CML patient management should be conducted by cooperating hematologists and gynecologists.

Keywords: chronic myeloid leukemia, pregnancy, tyrosine kinase inhibitors, imatinib, nilotinib, dasatinib, bosutinib.

Received: January 9, 2019

Accepted: March 20, 2019

Read in PDF 


REFERENCES

  1. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.

  2. Shukhov O, Chelysheva E, Gusarova G, et al. Imatinib treatment in chronic myeloid leukemia patients in early and late chronic phase: current incidence of cytogenetic remission and a very long-term an intention-to-treat analysis. Haematologica. 2015;100(Suppl 1):437.

  3. 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)]

  4. 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.

  5. Carlier P, Bernard N, Lagarce L, et al. Pregnancy outcome among partners of male patients receiving imatinib, dasatinib or nilotinib in chronic myeloid leukemia: reports collected by the French network pharmacovigilance centers. Arch Gynecol Obstet. 2017;295(2):269–71. doi: 10.1007/s00404-016-4262-z.

  6. Abruzzese E, Trawinska MM, de Fabritiis P, et al. Tyrosine kinase inhibitors and pregnancy. Mediterr J Hematol Infect Dis. 2014;6(1):2014028. doi: 10.4084/MJHID.2014.028.

  7. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Pregnancy outcomes in patients treated with bosutinib. Blood. 2018;132:1729, abstract.

  8. Palani R, Milojkovic D, Apperley JF. Managing pregnancy in chronic myeloid leukemia. Ann Hematol. 2015;94(Suppl 2):S167–76. doi: 10.1007/s00277-015-2317-z.

  9. Bhandari A, Rolen K, Shah BK. Management of chronic myelogenous leukemia in pregnancy. Anticancer Res. 2015;35(1):1–11.

  10. Abruzzese E, Trawinska MM, de Fabritiis P, et al. Management of pregnant chronic myeloid leukemia patients. Expert Rev Hematol. 2016;9(8):781–91. doi: 10.1080/17474086.2016.1205479.

  11. Челышева Е.Ю., Туркина А.Г. Протокол лечения хронического миелолейкоза во время беременности. В кн.: Алгоритмы диагностики и протоколы лечения заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2018. Т. 2. С. 927–49.

    [Chelysheva EYu, Turkina AG. Protocol of chronic myeloid leukemia treatment during pregnancy. In: Savchenko VG, ed. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. (Diagnostic algorithms and treatment protocols for blood system diseases.) Moscow: Praktika Publ.; 2018. Vol. 2. pp. 927–49. (In Russ)]

  12. Челышева Е.Ю., Туркина А.Г., Чабаева Ю.А. и др. Регистр случаев беременности при хроническом миелолейкозе: клинико-демографическая характеристика пациентов. Гематология и трансфузиология. 2016;61(1-S1):79.

    [Chelysheva EYu, Turkina AG, Chabaeva YuA, et al. Registry of pregnancy cases in chronic myeloid leukemia: clinical and demographic characteristics of patients. Gematologiya i transfuziologiya, 2016;61(1-S1):79. (In Russ)]

  13. 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.

  14. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. 2016;128(1):17–23. doi: 10.1182/blood-2016-01-694265.

  15. Hochhaus A, Saussele S, Rosti G, et al. Chronic myeloid leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29(Suppl 4):iv261. doi: 10.1093/annonc/mdy159.

  16. National Comprehensive Cancer Network. Chronic myeloid leukemia (Version 4.2018). Available from: https://www.nccn.org/professionals/physician_gls/pdf/cml.pdf. (accessed 8.01.2019).

  17. Hensley ML, Ford JM. Imatinib treatment: specific issues related to safety, fertility, and pregnancy. Semin Hematol. 2003;40(2 Suppl 2):21–5. doi: 10.1053/shem.2003.50038.

  18. Cortes JE, Abruzzese E, Chelysheva E, et al. The impact of dasatinib on pregnancy outcomes. Am J Hematol. 2015;90(12):1111–5. doi: 10.1002/ajh.24186.

  19. Pye SM, Cortes J, Ault P, et al. The effects of imatinib on pregnancy outcome. Blood. 2008;111(12):5505–8. doi: 10.1182/blood-2007-10-114900.

  20. Berveiller P, Andreoli A, Mir O, et al. A dramatic fetal outcome following transplacental transfer of dasatinib. Anti-Cancer Drugs. 2012;23(7):754–7. doi: 10.1097/CAD.0b013e328352a8fe.

  21. Polin RA, Fox WW, Abman SH. Fetal and Neonatal Physiology. 4th edition; 2011. 2208 p. [Internet] Available from: http://readli.net/fetal-and-neonatal-physiology. (accessed 8.01.2019).

  22. Abruzzese E, Scortechini AR, Gugliotta G, et al. Gimema Registry of conception/pregnancy in adult patients diagnosed with chronic myeloid leukemia (CML) treated with tyrosine kinase inhibitors (TKIs). Blood. 2014;124:1806, abstract.

  23. Chelysheva E, Galaiko MV, Kolosheinova TI, et al. Outcomes of pregnancy and therapeutic approaches in chronic myeloid leukemia during pregnancy. Hematologica. 2014;99(Suppl 1):336–7.

  24. Jovelet C, Seck A, Mir O, et al. Variation in transplacental transfer of tyrosine kinase inhibitors in the human perfused cotyledon model. Ann Oncol. 2015;26(7):1500–4. doi: 10.1093/annonc/mdv172.

  25. Chelysheva E, Turkina A, Polushkina E, et al. Placental transfer of tyrosine kinase inhibitors used for chronic myeloid leukemia treatment. Leuk Lymphoma. 2018;59(3):733–8. doi: 10.1080/10428194.2017.1347929.

  26. Russel MA, Carpenter MW, Akhtar MS, et al. Imatinib mesylate and metabolite concentration in maternal blood, umbilical cord blood, placenta and breast milk. J Perinatol. 2007;27(4):241–3. doi: 10.1038/sj.jp.7211665.

  27. Cole S, Kantarjian H, Ault P, et al. Successful completion of pregnancy in a patient with chronic myeloid leukemia without active intervention: a case report and review of the literature. Clin Lymph Myel. 2009;9(4):324–7. doi: 10.3816/CLM.2009.n.064.

  28. Iqbal J, Ali Z, Khan AU, et al. Pregnancy outcomes in patients with chronic myeloid leukemia treated with imatinib mesylate: short report from a developing country. Leuk Lymphoma. 2014;55(9):2109–13. doi: 10.3109/10428194.2013.866662.

  29. Alizadeh H, Jaafar H, Rajnics P, et al. Outcome of pregnancy in chronic myeloid leukaemia patients treated with tyrosine kinase inhibitors: short report from a single centre. Leuk Res. 2015;39(1):47–51. doi: 10.1016/j.leukres.2014.10.002.

  30. Saussele S, Richter J, Hochhaus A, et al. The concept of treatment-free remission in chronic myeloid leukemia. Leukemia. 2016;30(8):1638–47. doi: 10.1038/leu.2016.115.

  31. Mahon FX, Rea D, Guilhot J, et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 2010;11(11):1029–35. doi: 10.1016/S1470-2045(10)70233-3.

  32. Rea D, Nicolini F, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI Study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.

  33. Saussele S, Richter J, Guilhot J, et al Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–7. doi: 10.1016/S1470-2045(18)30192-X.

  34. Rousselot P, Charbonnier A, Cony-Makhoul P, et al. Loss of major molecular response as a trigger for restarting tyrosine kinase inhibitor therapy in patients with chronic-phase chronic myelogenous leukemia who have stopped imatinib after durable undetectable disease. J Clin Oncol. 2014;32(5):424–30. doi: 10.1200/JCO.2012.48.5797.

  35. Chelysheva E, Apperley J, Abruzzese E, et al. Kinetics of the leukemic clone in patients with chronic myeloid leukemia during pregnancy. 2018;132(Suppl 1):4254, abstract.

  36. Burchert A, Muller MC, Kostrewa P, et al. Sustained molecular response with interferon alfa maintenance after induction therapy with imatinib plus interferon alfa in patients with chronic myeloid leukemia. J Clin Oncol. 2010;28(8):1429–35. doi: 10.1200/JCO.2009.25.5075.

  37. Law AD, Kim DHD, Lipton JH. Pregnancy: part of life in chronic myelogenous leukemia. Leuk Lymphoma. 2017;58(2):280–7. doi: 10.1080/10428194.2016.1201571.

  38. Patel M, Dukes IA, Hull JC. Use of hydroxyurea in chronic myeloid leukemia during pregnancy; a case report. Am J Obstet Gynecol. 1991;165(3):565–6. doi: 10.1016/0002-9378(91)90285-y.

  39. Tretian G, Tchernia G, Papiernik E, et al. Hydroxyurea and pregnancy. Am J Obstet Gynecol. 1992;166(6):1868. doi: 10.1016/0002-9378(92)91590-7.

  40. Assi R, Kantarjian HM, Keating MJ, et al. Management of chronic myeloid leukemia (CML) during pregnancy among patients (pts) treated with a tyrosine kinase inhibitor (TKI): a single-center experience. Blood. 2017;130:2881, abstract.

  41. Ali R, Ozkalemkas F, Kimya Y, et al. Imatinib use during pregnancy and breast feeding: a case report and review of the literature. Arch Gynecol Obstet. 2009;280(2):169–75. doi: 10.1007/s00404-008-0861-7.

  42. Chelysheva E, Aleshin S, Polushkina E, et al. Breastfeeding in patients with chronic myeloid leukaemia: case series with measurements of drug concentrations in maternal milk and literature review. Mediterr J Hematol Infect Dis. 2018;10(1):2018027. doi: 10.4084/MJHID.2018.027.

  43. Chelysheva E, Turkina A, Polushkina E, et al. Results of treatment of patients with chronic myeloid leukemia and pregnancy in accordance with the leukemic burden and term of pregnancy (the LET scheme). EHA Learning Center. 2018. Abstract PF375. Available from: https://learningcenter.ehaweb.org/eha/2018/stockholm/214848/ekaterina.chelysheva.results.of.treatment.of.patients.with.chronic.myeloid.html?f=menu=6*ce_id=1346*ot_id=19052*media=3*marker=167 (accessed 8.01.2019).

  44. Chelysheva E, Turkina A, Polushkina E, et al. Treatment of patients with chronic myeloid leukemia during pregnancy according to scheme considering the leukemic burden and term of pregnancy (the LRT scheme). Clin Lymph Myel Leuk. 2018;18:S227–8. doi: 10.1016/j.clml.2018.07.102.

  45. Chelysheva E, Abruzzese E, Rea D, et al. Chronic myeloid leukemia diagnosed during pregnancy: therapy, outcomes and follow-up. 2018;132(Suppl 1):4255, abstract.

  46. Abruzzese E, de Fabritiis P, Trawinska MM, et al. Back to the future: Treatment-free remission and pregnancy in chronic myeloid leukemia. Eur J Haematol. 2018;102(2):197–9. doi: 10.1111/ejh.13192.

Treatment of Chronic Myeloid Leukemia According to Current Guidelines: The Results of the Pilot Prospective Study “Early Induction Therapy and Monitoring” (РИТМ)

OA Shukhov, AG Turkina, EYu Chelysheva, AV Bykova, AN Petrova, GA Gusarova, IS Nemchenko, AO Abdullaev, TN Obukhova, AB Sudarikov

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

For correspondence: Oleg Aleksandrovich Shukhov, MD, PhD, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: shuhov@list.ru

For citation: Shukhov OA, Turkina AG, Chelysheva EYu, et al. Treatment of Chronic Myeloid Leukemia According to Current Guidelines: The Results of the Pilot Prospective Study “Early Induction Therapy and Monitoring” (РИТМ). Clinical oncohematology 2019;12(2):194–201.

DOI: 10.21320/2500-2139-2019-12-2-194-201


ABSTRACT

Background. Current clinical guidelines on diagnosis and treatment of chronic myeloid leukemia (CML) define indications for substitution of first-line tyrosine kinase inhibitor (TKI) at therapy failure during different phases of disease progression.

Aim. To assess the efficacy of CML treatment with implementing the protocol of timely monitoring and switching to another TKI.

Materials & Methods. Patients were included into pilot prospective study РИТМ during 5 years. Data on 100 CML patients were analyzed. Therapy and monitoring were conducted according to the Federal clinical guidelines on CML diagnosis and therapy, 2013.

Results. Median follow-up after initiation of treatment was 46 months (range 12–74). Imatinib mesylate was administered as first-line therapy to 91 (91 %) patients, 9 (9 %) patients received 2nd generation TKI (TKI2). Therapy failure was registered in 31 (31 %) patients; 26 (84 %) of them were switched to TKI2. At the time of analysis 95 (95 %) patients were followed-up. Cumulative incidence of CML-associated mortality was 2 %. By the fifth year of follow-up cumulative probability of complete cytogenetic, major and deep molecular responses was 93 %, 88 % and 66 %, respectively.

Conclusion. CML treatment according to current guidelines yields the results comparable with those achieved by first-line TKI2 therapy. This approach reduces CML treatment costs and lowers the risk of TKI2-associated adverse events. Due to a high rate of deep molecular response the proportion of CML patients in remission without treatment can be increased in the future.

Keywords: chronic myeloid leukemia, monitoring, tyrosine kinase inhibitors, TKI switch.

Received: October 21, 2018

Accepted: February 4, 2019

Read in PDF 


REFERENCES

  1. O’Brien SG, Guilhot F, Larson RA, 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.

  2. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.

  3. Bower H, Bjorkholm M, Dickman PW, et al. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34(24):2851–7. doi: 10.1200/JCO.2015.66.2866.

  4. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.

  5. Cortes JE, Saglio G, Kantarjian HM, et al. Final 5-year study results of DASISION: the dasatinib versus imatinib study in treatment-naive chronic myeloid leukemia patients trial. J Clin Oncol. 2016;34(20):2333–40. doi: 10.1200/JCO.2015.64.8899.

  6. 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.

  7. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. Ponatinib efficacy and safety in Philadelphia chromosome-positive leukemia: final 5-year results of the phase 2 PACE trial. Blood. 2018;132(4):393–404. doi: 10.1182/blood-2016-09-739086.

  8. Marin D, Ibrahim AR, Lucas C, et al. Assessment of BCR-ABL1 transcript levels at 3 months is the only requirement for predicting outcome for patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. J Clin Oncol. 2012;30(3):232–8. doi: 10.1200/JCO.2011.38.6565.

  9. Jabbour E, Kantarjian HM, Saglio G, et al. Early response with dasatinib or imatinib in chronic myeloid leukemia: 3-year follow-up from a randomized phase 3 trial (DASISION). Blood. 2014;123(4):494–500. doi: 10.1182/blood-2013-06-511592.

  10. Saglio G, Snedecor SJ, Xiang J, et al. Early molecular response (EMR) with frontline treatment is a significant predictor of long-term BCR-ABL transcript levels in patients with Philadelphia chromosome-positive chronic myeloid leukemia (CML) in chronic phase. Blood. 2015;126(23):1579,

  11. Dulucq S, Mahon FX. Deep molecular responses for treatment-free remission in chronic myeloid leukemia. Cancer Med. 2016;5(9):2398–411. doi: 10.1002/cam4.801.

  12. 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.

  13. National Comprehensive Cancer Network. Chronic myeloid leukemia. Version 1.2019. Available from: https://www.nccn.org/professionals/physician_gls/pdf/cml.pdf. (accessed 3.12.2018).

  14. Абдулкадыров К.М., Абдуллаев А.О., Авдеева Л.Б. и др. Федеральные клинические рекомендации по диагностике и терапии хронического миелолейкоза. Вестник гематологии. 2013;9(3):4–41.

    [Abdulkadyrov KM, Abdullaev AO, Avdeeva LB, et al. Federal clinical guidelines for diagnosis and treatment of chronic myeloid leukemia. Vestnik gematologii. 2013;9(3):4–41. (In Russ)]

  15. 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: 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)]

  16. Klil-Drori AJ, Yin H, Azoulay L, et al. Early switch to second-line tyrosine kinase inhibitor in chronic myeloid leukemia patients failing to achieve early molecular response. Am J Hematol. 2017;92(10):E602–4. doi: 10.1002/ajh.24838.

  17. Milojkovic D, Clarck RE, Byrne JL, et al. The Target UK study: Real-world evidence of molecular response to tyrosine kinase inhibitors supports European LeukemiaNet 2013 recommendations for the management of chronic myeloid leukaemia. Blood. 2017;130(1):2892, abstract.

  18. Yeung DT, Osborn MP, White DL, et al. TIDEL-II: first-use of imatinib in CML with early switch to nilotinib for failure to achieve time-dependent molecular targets. Blood. 2015;125(6):915–23. doi: 10.1182/blood-2014-07-590315.

  19. Mahon FX. Discontinuation of tyrosine kinase therapy in CML. Ann Hematol. 2015;94(Suppl 2):187–93. doi: 10.1007/s00277-015-2320-4.

  20. Hochhaus A, Masszi T, Giles FJ, et al. Treatment-free remission following frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the ENESTfreedom study. Leukemia. 2017;31(7):1525–31. doi: 10.1038/leu.2017.63.

  21. Mahon FX, Boquimpani C, Kim DW, et al. Treatment-free remission after second-line nilotinib treatment in patients with chronic myeloid leukemia in chronic phase: results from a single-group, phase 2, open-label study. Ann Intern Med. 2018;168(7):461–70. doi: 10.7326/M17-1094.

  22. Etienne G, Guilhot J, Rea D, et al. Long-Term Follow-Up of the French Stop Imatinib (STIM1) Study in Patients With Chronic Myeloid Leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/JCO.2016.68.2914.

  23. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.

  24. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. Blood. 2016;128(1):17–23. doi: 10.1182/blood-2016-01-694265.

  25. Тасигна® (инструкция по медицинскому применению). Швейцария: Novartis pharma, AG. Доступно по: https://www.vidal.ru/drugs/tasigna__42603. Ссылка активна на12.2018.

    [Tasigna® (package insert). Switzerland: Novartis pharma, AG. Available from: https://www.vidal.ru/drugs/tasigna__42603. (accessed 3.12.2018) (In Russ)]

  26. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.

  27. ТуркинаА.Г., Челышева Е.Ю., Шуваев В.А. и др. Результаты наблюдения больных хроническим миелолейкозом с глубоким молекулярным ответом без терапии ингибиторами тирозинкиназ. Терапевтический архив. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96.

    [Turkina AG, Chelysheva EYu, Shuvaev VA, et al. Results of following up patients with chronic myeloid leukemia and a deep molecular response without tyrosine kinase inhibitor therapy. Terapevticheskii arkhiv. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96. (In Russ)]

  28. 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)]

  29. Шуваев В.А., Абдулкадыров К.М., Туркина А.Г. и др. Фармакоэкономический анализ ремиссии хронического миелолейкоза без лечения. Гематология и трансфузиология. 2015;60(4):14–20.

    [Shuvaev VA, Abdulkadyrov KM, Turkina AG, et al. Pharmacoeconomic analysis of chronic myeloid leukemia remission without treatment. Gematologiya i transfuziologiya. 2015;60(4):14–20. (In Russ)]