A Rationale for a New Operational Integrated Quality and Efficiency Index for Assessing the Performance of Hematological Services in Constituent Entities of the Russian Federation

EN Parovichnikova1, TTs Garmaeva1, OV Lazareva1, KA Lukina1, YuA Chabaeva1, SM Kulikov1, VV Troitskaya1, TV Gaponova1, LI Menshikova2, VG Savchenko1

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

2 Federal Research Institute for Health Organization and Informatics, 11 Dobrolyubova str., Moscow, Russian Federation, 127254

For correspondence: Elena Nikolaevna Parovichnikova, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: parovichnikova@gmail.com

For citation: Parovichnikova EN, Garmaeva TTs, Lazareva OV, et al. A Rationale for a New Operational Integrated Quality and Efficiency Index for Assessing the Performance of Hematological Services in Constituent Entities of the Russian Federation. Clinical oncohematology. 2022;15(1):1–15. (In Russ).

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


ABSTRACT

Background. Since 2018 a widespread national project “Healthcare” has been implemented in the Russian Federation (RF) to improve the quality, efficiency, availability, and affordability of medical care in the profiles of specialties in constituent entities of the RF. Modern hematology as a medical field of high technology and crucial solutions is notable for its multi- and interdisciplinarity of most nosological forms, complexity of diagnostic process, multi-structuredness and diversity of related physician teams in different structural units and subdivisions. One of the key issues in federal projects is to determine the indicators for assessing the efficiency of regional hematological services in constituent entities of the RF.

Aim. To elaborate and substantiate a new integrated operational efficiency index for hematological services in constituent entities of the RF.

Materials & Methods. The analysis of data and assessment of feasibility of a new integrated operational index “early mortality in acute leukemia” (AL) were based on the results of 5 multi-center trials, including an epidemiological one.

Results. Multi-center clinical studies on AL are the only objective tools for assessing the treatment efficacy, its improvement, and further training of hematologists taking part in the trials. AL treatment requires well-developed infrastructure of hematological services involving not only staff matters and organization of hematologists’ activities, but also management of many highly important related subdivisions and laboratories, logistics of their interaction, time specifications, meeting clinical guidelines, and lastly, and most importantly, financial support.

Conclusion. The Unified State Information System “Hematology” is the only platform providing the objective information on patients’ vital status and enabling the use of the suggested integrated index for assessing the quality and efficiency of hematological services in the regions of the RF. This indicator of early mortality in AL patients less than 60 years of age is 15 % for acute myeloid leukemias and 10 % for acute lymphoblastic leukemias. Its low values would demonstrate that this or that constituent entity of the RF is provided with sufficient infrastructure, technologies, and a professional team to keep those patients alive who have severe but curable hematological diseases. The indicator of long-term survival or “life years gained” should become the main strategic criterion for the therapy efficacy in hematological diseases.

Keywords: hematological services, assessment of the medical care quality, early mortality, overall survival, acute leukemia, acute myeloid leukemia, acute lymphoblastic leukemia.

Received: September 27, 2021

Accepted: December 15, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Приказ Минздрава России от 29.12.2012г. № 1706 «Об утверждении методических рекомендаций по разработке органами исполнительной власти субъектов Российской Федерации планов мероприятий («дорожных карт») «Изменения в отраслях социальной сферы, направленные на повышение эффективности здравоохранения в субъекте Российской Федерации».
    [Decree No. 1706 of the Ministry of Health of the Russian Federation dated December 29, 2012. On the approval of methodological guidelines for the development of action plans (roadmaps) by the executive authorities of constituent entities of the Russian Federation. Changes in the social sectors aimed at improving the efficiency of healthcare in constituent entities of the Russian Federation. (In Russ)]
  2. Валиев Т.Т., Шервашидзе М.А., Осипова И.В. и др. Лечение острого лимфобластного лейкоза у детей по протоколу ALL IC-BFM 2002: результаты мультицентрового ретроспективного исследования. Российский журнал детской гематологии и онкологии. 2021;8(3):59–70. doi: 10.21682/2311-1267-2021-8-3-59-70.
    [Valiev TT, Shervashidze MA, Osipova IV, et al. Treatment of acute lymphoblastic leukemia in children by ALL IC-BFM 2002 protocol: results of multicenter retrospective study. Russian Journal of Pediatric Hematology and Oncology. 2021;8(3):59–70. doi: 10.21682/2311-1267-2021-8-3-59-70. (In Russ)]
  3. Шервашидзе М.А., Валиев Т.Т. Совершенствование программ терапии острого лимфобластного лейкоза у детей: акцент на минимальную остаточную болезнь. Онкогематология. 2020;15(3):12–26. doi: 10.17650/1818­8346­2020­15­3­12­26.
    [Shervashidze MA, Valiev TT. Pediatric acute lymphoblastic leukemia treatment protocols improvement: emphasis on minimal residual disease. Oncohematology. 2020;15(3):12–26. doi: 10.17650/1818­8346­2020­15­3­12­26. (In Russ)]
  4. Berwick D, Fox D. Evaluating the Quality of Medical Care: Donabedian’s Classic Article 50 Years Later. Milbank Q. 2016;94(2):237–41. doi: 10.1111/1468-0009.12189.
  5. Donabedian A. Evaluating the quality of medical care. Milbank Q. 2005;83(4):691–729. doi: 10.1111/j.1468-0009.2005.00397.x.
  6. Mengis C, Aebi S, Tobler A, et al. Assessment of differences in patient populations selected for excluded from participation in clinical phase III acute myelogenous leukemia trials. J Clin Oncol. 2003;21(21):3933–9. doi: 10.1200/JCO.2003.03.186.
  7. Гармаева Т.Ц., Савченко В.Г., Лазарева О.В. и др. Организация выездных мероприятий с посещением профильных медицинских организаций (МО) субъектов РФ. Гематология и трансфузиология. 2020;65(S1):65.
    [Garmaeva TTs, Savchenko VG, Lazareva OV, et al. Organization of offsite events with visiting specialized healthcare organizations (HO) of constituent entities of the Russian Federation. Gematologiya i transfuziologiya. 2020;65(S1):65. (In Russ)]
  8. Федеральный закон от 29.07.2017 № 242-ФЗ «О внесении изменений в отдельные законодательные акты Российской Федерации по вопросам применения информационных технологий в сфере охраны здоровья».
    [Federal Law No. 242-FZ dated July 29, 2017. On amendments to certain legislative acts of the Russian Federation on information technology for health protection. (In Russ)]
  9. Злокачественные новообразования в России в 2019году (заболеваемость и смертность). Под ред. А.Д. Каприна, В.В. Старинского, А.О. Шахзадовой. М.: МНИОИ им. П.А. Герцена — филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2020. 252 с.
    [Kaprin AD, Starinskii VV, Shakhzadova AO, eds. Zlokachestvennye novoobrazovaniya v Rossii v 2019 godu (zabolevaemost’ i smertnost’). (Malignant neoplasms in Russia in 2019 (incidence and mortality.) Moscow: MNIOI im. P.A. Gertsena — filial FGBU “NMITs radiologii” Publ.; 252 p. (In Russ)]
  10. Савченко В.Г., Паровичникова Е.Н., Клясова Г.А. и др. Итоги многоцентрового кооперированного исследования по лечению острых миелоидных лейкозов взрослых. Терапевтический архив. 1994;66(7):11–7.
    [Savchenko VG, Parovichnikova EN, Klyasova GA, et al. Results of a multicenter collaborative study on the treatment of acute myeloid leukemia in adults. Terapevticheskii arkhiv. 1994;66(7):11–7. (In Russ)]
  11. Савченко В.Г., Паровичникова Е.Н., Клясова Г.А., Исаев В.Г. Итоги двух с половиной лет работы российского многоцентрового исследования по лечению острых лейкозов взрослых. Терапевтический архив. 1995;67(7):8–12.
    [Savchenko VG, Parovichnikova EN, Klyasova GA, Isaev VG. Results of a two-and-a-half-year Russian multicenter study on the treatment of acute leukemia in adults. Terapevticheskii arkhiv. 1995;67(7):8–12. (In Russ)]
  12. Савченко В.Г., Паровичникова Е.Н., Клясова Г.А. и др. Результаты проводимых в течение 7 лет клинических исследований по лечению острых миелоидных лейкозов взрослых. Терапевтический архив. 1999;71(7):13–20.
    [Savchenko VG, Parovichnikova EN, Klyasova GA, et al. Results of 7-year clinical studies on the treatment of acute myeloid leukemia in adults. Terapevticheskii arkhiv. 1999;71(7):13–20. (In Russ)]
  13. Савченко В.Г., Паровичникова Е.Н., Исаев В.Г. и др. Лечение острых лимфобластных лейкозов взрослых как нерешенная проблема. Терапевтический архив. 2001;73(7):6–15.
    [Savchenko VG, Parovichnikova EN, Isaev VG, et al. Treatment of acute lymphoblastic leukemia in adults as an unsolved problem. Terapevticheskii arkhiv. 2001;73(7):6–15. (In Russ)]
  14. Савченко В.Г., Паровичникова Е.Н., Менделеева Л.П. и др. Многоцентровая кооперация — основа прогресса в лечении лейкозов. Терапевтический архив. 2005;77(7):5–11.
    [Savchenko VG, Parovichnikova EN, Mendeleeva LP, et al. Multicenter cooperation is a basis of progress in the treatment of leukemia. Terapevticheskii arkhiv. 2005;77(7):5–11. (In Russ)]
  15. Паровичникова Е.Н., Савченко В.Г., Исаев В.Г. и др. Итоги многоцентрового рандомизированного исследования по лечению острых миелоидных лейкозов взрослых. Терапевтический архив. 2007;79(7):14–9.
    [Parovichnikova EN, Savchenko VG, Isaev VG, et al. Results of a multicenter randomized study on the treatment of acute myeloid leukemia in adults. Terapevticheskii arkhiv. 2007;79(7):14–9. (In Russ)]
  16. Паровичникова Е.Н., Савченко В.Г., Клясова Г.А. и др. Токсичность различных протоколов лечения острых миелоидных лейкозов взрослых: результат четырех российских многоцентровых исследований. Терапевтический архив. 2010;82(7):5–11.
    [Parovichnikova EN, Savchenko VG, Klyasova GA, et al. Toxicity of different treatment protocols for acute myeloid leukemias in adults: the results of four Russian multicenter studies. Terapevticheskii arkhiv. 2010;82(7):5–11. (In Russ)]
  17. Соколов А.Н., Паровичникова Е.Н., Куликов С.М. и др. Долгосрочные результаты лечения острых миелоидных лейкозов у взрослых в многоцентровом клиническом исследовании ОМЛ 06.06. Клиническая онкогематология. 2012;5(1):30–8.
    [Sokolov AN, Parovichnikova EN, Kulikov SM, et al. Long-term results of acute myeloid leukemia treatment in adults in a multicenter clinical study of AML 06.06. Klinicheskaya onkogematologiya. 2012;5(1):30–8. (In Russ)]
  18. Паровичникова Е.Н., Клясова Г.А., Соколов А.Н. и др. Первые результаты лечения острых миелоидных лейкозов взрослых по протоколу ОМЛ-01.10 Научно-исследовательской группы гематологических центров России. Терапевтический архив. 2012;84(7):10–5.
    [Parovichnikova EN, Klyasova GA, Sokolov AN, et al. The first results of acute myeloid leukemia treatment in adults according to the AML-01.10 protocol of the Research Group of Hematological Centers in Russia. Terapevticheskii arkhiv. 2012;84(7):10–5. (In Russ)]
  19. Паровичникова Е.Н., Троицкая В.В., Клясова Г.А. и др. Лечение больных острыми миелоидными лейкозами по протоколу российского многоцентрового рандомизированного исследования ОМЛ-01.10: результаты координационного центра. Терапевтический архив. 2014;86(7):14–23.
    [Parovichnikova EN, Troitskaya VV, Klyasova GA, et al. Treating patients with acute myeloid leukemias (AML) according to the protocol of the AML-01.10 Russian multicenter randomized trial: the coordinating center’s results. Terapevticheskii arkhiv. 2014;86(7):14–23. (In Russ)]
  20. Паровичникова Е.Н., Лукьянова И.А., Троицкая В.В. и др. Результаты программной терапии острых миелоидных лейкозов в ФГБУ «НМИЦ гематологии» Минздрава России. Терапевтический архив. 2018;90(7):14–22.
    [Parovichnikova EN, Lukyanova IA, Troitskaya VV, et al. Results of the program therapy of acute myeloid leukemia in the FGBU “NMITS Gematologii” of the Ministry of Health of Russia. Terapevticheskii arkhiv. 2018;90(7):14–22. (In Russ)]
  21. Buchner T, Schlenk RF, Schaich M, et al. Acute Myeloid Leukemia (AML): Different Treatment Strategies Versus a Common Standard Arm—Combined Prospective Analysis by the German AML Intergroup. J Clin Oncol. 2012;30(29):3604–10. doi: 10.1200/JCO.2012.42.2907.
  22. Othus M, Kantarjian H, Petersdorf S, et al Declining rates of treatment-related mortality in patients with newly diagnosed AML given ‘intense’ induction regimens: a report from SWOG and MD Anderson. 2013;28(2):289–92. doi: 10.1038/leu.2013.176.
  23. Паровичникова Е.Н., Давидян Ю.Р., Исаев В.Г. и др. Итоги лечения острых лимфобластных лейкозов взрослых по протоколу ОЛЛ-2005 как основа для новых исследований. Терапевтический архив. 2009;81(7):8–15.
    [Parovichnikova EN, Davidyan YuR, Isaev VG, et al. Results of acute lymphoblastic leukemia treatment in adults according to the ALL-2005 protocol as a basis for new research. Terapevticheskii arkhiv. 2009;81(7):8–15. (In Russ)]
  24. Паровичникова Е.Н., Клясова Г.А., Троицкая В.В. и др. Эффективность лечения взрослых больных острым Т-лимфобластным лейкозом по протоколу ОЛЛ-2009 российской научно-исследовательской группы по изучению острых лейкозов. Терапевтический архив. 2013;85(8):29–34.
    [Parovichnikova EN, Klyasova GA, Troitskaya VV, et al. The efficacy of acute T-lymphoblastic leukemia treatment in adults according to the ALL-2009 protocol of the Russian research group for the study of acute leukemia. Terapevticheskii arkhiv. 2013;85(8):29–34. (In Russ)]
  25. Паровичникова Е.Н., Троицкая В.В., Соколов А.Н. и др. Промежуточные результаты по лечению острых Ph-негативных лимфобластных лейкозов у взрослых больных (итоги российской исследовательской группы по лечению острых лимфобластных лейкозов (RALL)). Онкогематология. 2014;9(3):6–15.
    [Parovichnikova EN, Troitskaya VV, Sokolov AN, et al. Interim outcomes of acute Ph-negative lymphoblastic leukemia treatment in adults (results of the Russian research group on the treatment of acute lymphoblastic leukemias (RALL)). Onkogematologiya. 2014;9(3):6–15. (In Russ)]
  26. Паровичникова Е.Н., Соколов А.Н., Троицкая В.В. и др. Острые Ph-негативные лимфобластные лейкозы взрослых: факторы риска при использовании протокола ОЛЛ-2009. Терапевтический архив. 2016;88(7):15–24.
    [Parovichnikova EN, Sokolov AN, Troitskaya VV, et al. Acute Ph-negative lymphoblastic leukemias in adults: ALL-2009 protocol risk factors. Terapevticheskii arkhiv. 2016;88(7):15–24. (In Russ)]
  27. Паровичникова Е.Н., Троицкая В.В., Соколов А.Н. и др. Острые В-лимфобластные лейкозы взрослых: выводы из российского проспективного многоцентрового исследования ОЛЛ-2009. Терапевтический архив. 2017;89(7):10–7.
    [Parovichnikova EN, Troitskaya VV, Sokolov AN, et al. Acute B-lymphoblastic leukemia in adults: findings of the Russian prospective multicenter study ALL-2009. Terapevticheskii arkhiv. 2017;89(7):10–7. (In Russ)]
  28. Gokbuget N, Beck J, Brandt K. Significant Improvement Of Outcome In Adolescents and Young adults (AYAs) Aged 15–35 Years With Acute Lymphoblastic Leukemia (ALL) With a Pediatric Derived Adult ALL Protocol; Results Of 1529 AYAs In 2 Consecutive Trials Of The German Multicenter Study Group For Adult ALL (GMALL). 2013;22(21):839. doi: 10.1182/blood.V122.21.839.839.
  29. Rowe JM, Buck G, Burnett AK, et al. Induction therapy for adults with acute lymphoblastic leukemia: results of more than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993. 2005;106(12):3760–7. doi: 10.1182/blood-2005-04-1623.
  30. Francoise H, Thibaut L, Emmanuel R, et al. Pediatric-Inspired Therapy in Adults With Philadelphia Chromosome–Negative Acute Lymphoblastic Leukemia: The GRAALL-2003 Study. J Clin Oncol. 2009;27(6):911–8. doi:1200/JCO.2008.18.6916.
  31. Гайдамака Н.В., Паровичникова Е.Н., Гармаева Т.Ц. и др. Длительные аплазии костного мозга после химиотерапии у больных острыми лейкозами. Терапевтический архив. 2010;82(7):29–34.
    [Gaidamaka NV, Parovichnikova EN, Garmaeva TTs, et al. Long-term bone marrow aplasias after chemotherapy in acute leukemia patients. Terapevticheskii arkhiv. 2010;82(7):29–34. (In Russ)]
  32. Алгоритмы диагностики и программная терапия заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2018.
    [Savchenko VG, ed. Algoritmy diagnostiki i programmnaya terapiya zabolevanii sistemy krovi. (Diagnostic algorithms and program therapy for hematological diseases.) Moscow: Praktika Publ.; 2018. (In Russ)]
  33. Ахмерзаева З.Х., Паровичникова Е.Н., Русинов М.А. и др. Эпидемиологическое исследование острых лейкозов в пяти регионах Российской Федерации. Гематология и трансфузиология. 2017;62(1):46–51. doi: 10.18821/0234-5730-2017-62-1-46-51.
    [Akhmerzaeva ZKh, Parovichnikova EN, Rusinov MA, et al. The epidemiological study of acute leukemia in five regions of the Russian Federation. Gematologiya i transfuziologiya. 2017;62(1):46–51. doi: 10.18821/0234-5730-2017-62-1-46-51. (In Russ)]
  34. Лукина К.А., Зайцев Д.А., Гармаева Т.Ц., Менделеева Л.П. Телемедицина как инструмент межрегионального дистанционного взаимодействия с профильными медицинскими организациями субъектов Российской Федерации: 5-летний опыт ФГБУ «НМИЦ гематологии» Минздрава России. Врач и информационные технологии. 2020;4:68–77. doi: 10.37690/1811-0193-2020-4-68-77.
    [Lukina KA, Zaytsev DA, Garmaeva TT, Mendeleeva LP. Telemedicine as a tool for remote interaction with regional hospitals: 5-year experience of the National Research Center for Hematology. Vrach i informatsionnye tekhnologii, 2020;4:68–77. doi: 10.37690/1811-0193-2020-4-68-77. (In Russ)]
  35. Железнякова И.А., Серяпина Ю.В., Михайлов И.А. и др. Методологические подходы к внедрению системы контроля качества медицинской помощи в медицинских организациях. Медицинские технологии. Оценка и выбор. 2020;42(4):13–20. doi: 10.17116/medtech
    [Zheleznyakova IA, Seryapina YuV, Mikhailov IA, et al. Methodological approaches to the development of medical care quality control at medical organizations. Medical Technologies. Assessment and Choice. 2020;42(4):13–20. doi: 10.17116/medtech20204204113. (In Russ)]
  36. Брескина Т.Н. Карта экспертизы качества медицинской помощи как основа организации контроля качества медицинской помощи в многопрофильном стационаре. Вестник Росздравнадзора. 2016;1:21–31.
    [Breskina TN. Medical quality evaluation card as a basis for medical care quality control at a multidisciplinary hospital. Vestnik Roszdravnadzora. 2016;1:21–31. (In Russ)]
  37. Сухоруких О.А., Лукьянцева Д.В., Омельяновский В.В. Критерии оценки качества медицинской помощи. Менеджмент качества в медицине. 2018;2:15–21.
    [Sukhorukikh OA, Lukyantseva DV, Omelyanovskii VV. Criteria for quality assessment of medical care. Menedzhment kachestva v meditsine. 2018;2:15–21. (In Russ)]
  38. Семочкин С.В., Толстых Т.Н., Архипова Н.В. и др. Клинико-эпидемиологическая характеристика острых миелоидных лейкозов у взрослых по данным муниципальных отделений гематологии Москвы. Терапевтический архив. 2015;87(7):26–32. doi: 10.17116/terarkh201587726-32.
    [Semochkin SV, Tolstykh TN, Arkhipova NV, et al. Clinical and epidemiological characteristics of acute myeloid leukemias in adults according to the data of the municipal hematology departments of Moscow. Terapevticheskii arkhiv. 2015;87(7):26–32. doi: 10.17116/terarkh201587726-32. (In Russ)]
  39. Лазарева О.В., Куликов С.М., Чабаева Ю.А. и др. Единая информационная система (ЕИС) «Гематология» — учет, регистрация и мониторинг пациентов с заболеваниями системы крови в РФ. Гематология и трансфузиология. 2020;65(S1):33.
    [Lazareva OV, Kulikov SM, Chabaeva YuA, et al. Unified information system (UIE) “Hematology”: administration, registration, and monitoring of patients with hematological diseases in the Russian Federation. Gematologiya i transfuziologiya, 2020;65(S1):33. (In Russ)]
  40. Куликов С.М., ГармаеваТ.Ц., Русинов М.А., Паровичникова Е.Н. Понятия, принципы и задачи популяционной гематологии. Клиническая онкогематология. 2017;10(2):250–7. doi: 10.21320/2500-2139-2017-10-2-250-257.
    [Kulikov SM, Garmaeva TTs, Rusinov MA, Parovichnikova EN. Concept, Principles, and Objectives of Population Hematology. Clinical oncohematology. 2017;10(2):250–7. doi: 10.21320/2500-2139-2017-10-2-250-257. (In Russ)]
  41. Куликов С.М., Чабаева Ю.А., Лазарева О.В. Цель создания новой национальной системы кодирования заболеваний системы крови. Гематология и трансфузиология. 2020;65(S1):32–3.
    [Kulikov SM, Chabaeva YuA, Lazareva OV. The purpose of creating a new national coding system for hematological diseases. Gematologiya i transfuziologiya. 2020;65(S1):32–3. (In Russ)]
  42. Баженов А.В., Галстян Г.М., Паровичникова Е.Н. и др. Роль интенсивной терапии в лечении больных острыми миелоидными лейкозами. Терапевтический архив. 2019;91(7):14–24. doi: 10.26442/00403660.2019.07.000321.
    [Bazhenov AV, Galstyan GM, Parovichnikova EN, et al. Role of the intensive care in treatment of patients with acute myeloid leukemia. Terapevticheskii arkhiv. 2019;91(7):14–24. doi: 10.26442/00403660.2019.07.000321. (In Russ)]
  43. Савченко В.Г., Паровичникова Е.Н., Афанасьев Б.В. Национальные клинические рекомендации по диагностике и лечению острых миелоидных лейкозов взрослых. Гематология и трансфузиология. 2014;59(1-S2):2–29.
    [Savchenko VG, Parovichnikova EN, Afanasyev BV. National clinical guidelines for the diagnosis and treatment of acute myeloid leukemia in adults. Gematologiya i transfuziologiya. 2014;59(1-S2):2–29. (In Russ)]
  44. Савченко В.Г., Паровичникова Е.Н., Афанасьев Б.В. Клинические рекомендации российских экспертов по лечению больных острыми миелоидными лейкозами в возрасте моложе 60 лет. Терапевтический архив. 2014;86(7):4–13.
    [Savchenko VG, Parovichnikova EN, Afanasyev BV. Clinical guidelines of Russian experts for the treatment of acute myeloid leukemia patients less than 60 years of age. Terapevticheskii arkhiv. 2014;86(7):4–13. (In Russ)]
  45. Савченко В.Г., Паровичникова Е.Н., Афанасьев Б.В. Клинические рекомендации по диагностике и лечению острых лимфобластных лейкозов взрослых (редакция 2018 г.) Гематология и трансфузиология. 2018;63(1-S2):5–52.
    [Savchenko VG, Parovichnikova EN, Afanasyev BV. Clinical guidelines for the diagnosis and treatment of acute lymphoblastic leukemia in adults (edition 2018). Gematologiya i transfuziologiya. 2018;63(1-S2):5–52. (In Russ)]
  46. Приказ Минздрава России от 15 ноября 2012 г. № 930н «Об утверждении Порядка оказания медицинской помощи населению по профилю «гематология»». [Decree No. 930n of the Ministry of Health of the Russian Federation dated November 15, 2012. On the approval of medical care provision in the field of hematology. (In Russ)]

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

NN Mamaev, AI Shakirova, EV Morozova, TL Gindina

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

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

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

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


ABSTRACT

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

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

Received: September 12, 2020

Accepted: December 6, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

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

Molecular Diagnosis of FLT3 Mutations in Acute Myeloid Leukemia Patients

EK Zaikova1,2, EV Belotserkovskaya1,2, DV Zaytsev1, AV Petukhov1,2, OA Fedorova2, DV Motorin1, VV Ivanov1, AYu Zaritskey1, LL Girshova1

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

2 Institute of Citology, 4 Tikhoretskii pr-t, Saint Petersburg, Russian Federation, 194064

For correspondence: Ekaterina Vasil’evna Belotserkovskaya, PhD in Biology, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; e-mail: belotserkovskaya.ev@gmail.com

For citation: Zaikova EK, Belotserkovskaya EV, Zaytsev DV, et al. Molecular Diagnosis of FLT3 Mutations in Acute Myeloid Leukemia Patients. Clinical oncohematology. 2020;13(2):150–60 (In Russ).

DOI: 10.21320/2500-2139-2020-13-2-150-160


ABSTRACT

Background. FLT3 gene is an important prognostic molecular marker in acute myeloid leukemia (AML). However, the detection of FLT3 mutations presents a challenge.

Aim. To compare techniques used for the detection of FLT3 mutations, and to develop a test-system based on polymerase chain reaction (PCR) for quick and reliable determination of FLT3 mutation status.

Materials & Methods. Bone marrow samples obtained from AML patients were subjected to examination. To detect FLT3-ITD и FLT3-TKD mutations PCR was performed with subsequent agarose gel electrophoresis visualization. The results were verified by Sanger sequencing. The data obtained using our test-system were compared with widely applied commercial kit ‘FLT3 Mutation Assay for Gel Detection’ by Invivoscribe.

Results. To determine the FLT3 mutation status a PCR test was developed. This technique was validated on 22 bone marrow samples obtained from AML patients. FLT3-ITD mutation was detected in 4 patients, 3 patients showed FLT3-TKD mutation. In 1 patient both mutations were identified. These results fully corresponded to the molecular genetic analysis of FLT3, performed by ‘FLT3 Mutation Assay for Gel Detection’. The chosen technique was validated using Sanger sequencing data analysis.

Conclusion. The article offers the review of all existing FLT3 mutation screening techniques and describes the experience of developing the PCR test for FLT3-ITD and FLT3-TKD mutation detection. The chosen technique is affordable and easy to use compared with the others. The present study with its applied nature can provide guidance for both doctors and researchers.

Keywords: acute myeloid leukemia, FLT3-ITD and FLT3-TKD mutations.

Received: January 10, 2020

Accepted: March 27, 2020

Read in PDF


REFERENCES

  1. Kiyoi H, Naoe T, Yokota S, et al. Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia. 1997;11(9):1447–52. doi: 10.1038/sj.leu.2400756.

  2. Блау О.В. Мутации генов при острых миелоидных лейкозах. Клиническая онкогематология. 2016;9(3):245–56. doi: 10.21320/2500-2139-2016-9-3-245-256.

    [Blau OV. Genetic Mutations in Acute Myeloid Leukemia. Clinical oncohematology. 2016;9(3):245–56. doi: 10.21320/2500-2139-2016-9-3-245-256. (In Russ)]

  3. Gu T, Nardone J, Wang Y, et al. Survey of Activated FLT3 Signaling in Leukemia. PLoS ONE. 2011;6(4):e19169. doi: 10.1371/journal.pone.0019169.

  4. Deeb KK, Smonskey MT, Defedericis H-C, et al. Deletion and deletion/insertion mutations in the juxtamembrane domain of the FLT-3 gene in adult acute myeloid leukemia. Leuk Res Rep. 2014;3(2):86–9. doi: 10.1016/j.lrr.2013.09.003.

  5. Sandhofer N, Bauer J, Reiter K, et al. The new and recurrent FLT3 juxtamembrane deletion mutation shows a dominant negative effect on the wild-type FLT3 receptor. Sci Rep. 2016;6(1):28032. doi: 10.1038/srep28032.

  6. Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99(12):4326–35. doi: 10.1182/blood.v99.12.4326.

  7. Metzeler K, Herold T, Rothenberg-Thurley M, et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood. 2016;128(5):686–98. doi: 10.1182/blood-2016-01-693879.

  8. Schnittger S, Bacher U, Haferlach C, et al. Diversity of the juxtamembrane and TKD1 mutations (Exons 13–15) in the FLT3 gene with regards to mutant load, sequence, length, localization, and correlation with biological data. Genes Chromos Cancer. 2012;51(10):910–24. doi: 10.1002/gcc.21975.

  9. Kottaridis P, Gale R, Frew M, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9. doi: 10.1182/blood.v98.6.1752.

  10. Moreno I, Martin G, Bolufer P, et al. Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia. Haematologica. 2003;88(1):19–24.

  11. Bacher U, Haferlach C, Kern W, et al. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters—an analysis of 3082 patients. Blood. 2008;111(5):2527–37. doi: 10.1182/blood-2007-05-091215.

  12. Moore AS, Faisal A, Gonzalez de Castro D, et al. Selective FLT3 inhibition of FLT3-ITD+ acute myeloid leukaemia resulting in secondary D835Y mutation: a model for emerging clinical resistance patterns. Leukemia. 2012;26(7):1462–70. doi: 10.1038/leu.2012.52.

  13. Bagrintseva K, Geisenhof S, Kern R, et al. FLT3-ITD-TKD dual mutants associated with AML confer resistance to FLT3 PTK inhibitors and cytotoxic agents by overexpression of Bcl-x(L). Blood. 2005;105(9):3679–85. doi: 10.1182/blood-2004-06-2459.

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

  15. O’Donnell MR, Tallman MS, Abboud CN, et al. Acute myeloid leukemia, version 3.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2017;15(7):926–57. doi: 10.6004/jnccn.2017.0116.

  16. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10(12):1911–8.

  17. Kim Y, Lee GD, Park J, et al. Quantitative fragment analysis of FLT3-ITD efficiently identifying poor prognostic group with high mutant allele burden or long ITD length. Blood Cancer J. 2015;5(8):e336. doi: 10.1038/bcj.2015.61.

  18. Lin PH, Lin CC, Yang HI, et al. Prognostic impact of allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia patients with internal tandem duplication of FLT3. Leuk Res. 2013;37(3):287–92. doi: 10.1016/j.leukres.2012.10.005.

  19. Grunwald MR, Tseng LH, Lin MT, et al. Improved FLT3 internal tandem duplication PCR assay predicts outcome after allogeneic transplant for acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(12):1989–95. doi: 10.1016/j.bbmt.2014.08.015.

  20. Murphy KM, Levis M, Hafez MJ, et al. Detection of FLT3 internal tandem duplication and D835 mutations by a multiplex polymerase chain reaction and capillary electrophoresis assay. J Mol Diagn. 2003;5(2):96–102. doi: 10.1016/s1525-1578(10)60458-8.

  21. Stirewalt DL, Willman CL, Radich JP. Quantitative, real-time polymerase chain reactions for FLT3 internal tandem duplications are highly sensitive and specific. Leuk Res. 2001;25(12):1085–8. doi: 10.1016/s0145-2126(01)00087-x.

  22. Beretta C, Gaipa G, Rossi V, et al. Development of a quantitative-PCR method for specific FLT3/ITD monitoring in acute myeloid leukemia. Leukemia. 2004;18(8):1441–4. doi: 10.1038/sj.leu.2403409.

  23. Daver N, Schlenk RF, Russell NH, et al. Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia. 2019;33(2):299–312. doi: 10.1038/s41375-018-0357-9.

  24. Patnaik MM. The importance of FLT3 mutational analysis in acute myeloid leukemia. Leuk Lymphoma. 2018;59(10):2273–86. doi: 10.1080/10428194.2017.1399312.

  25. Kamps R, Brandao RD, Bosch BJ, et al. Next-generation sequencing in oncology: genetic diagnosis, risk prediction and cancer classification. Int J Mol Sci. 2017;18(2):308. doi: 10.3390/ijms18020308.

  26. Chin EL, da Silva C, Hegde M. Assessment of clinical analytical sensitivity and specificity of next-generation sequencing for detection of simple and complex mutations. BMC Genet. 2013;14(1):6. doi: 10.1186/1471-2156-14-6.

  27. Bibault JE, Figeac M, Helevaut N, et al. Next-generation sequencing of FLT3 internal tandem duplications for minimal residual disease monitoring in acute myeloid leukemia. Oncotarget. 2015;6(26):22812–21. doi: 10.18632/oncotarget.4333.

  28. Spencer DH, Abel HJ, Lockwood CM, et al. Detection of FLT3 Internal Tandem Duplication in Targeted, Short-Read-Length, Next-Generation Sequencing Data. J Mol Diagn. 2013;15(1):81–93. doi: 10.1016/j.jmoldx.2012.08.001.

  29. Au CH, Wa A, Ho DN, et al. Clinical evaluation of panel testing by next-generation sequencing (NGS) for gene mutations in myeloid neoplasms. Diagn Pathol. 2016;11(1):11. doi: 10.1186/s13000-016-0456-8.

  30. Abdelhamid E, Preudhomme C, Helevaut N, et al. Minimal residual disease monitoring based on FLT3 internal tandem duplication in adult acute myeloid leukemia. Leuk Res. 2012;36(3):316–23. doi: 10.1016/j.leukres.2011.11.002.

  31. Schranz K, Hubmann M, Harin E, et al. Clonal heterogeneity of FLT3-ITD detected by high-throughput amplicon sequencing correlates with adverse prognosis in acute myeloid leukemia. Oncotarget. 2018;9(53):30128–45. doi: 10.18632/oncotarget.25729.

  32. Thiede C, Prior T, Lavorgna S, et al. FLT3 mutation Assay Laboratory Cross Validation: Results from the CALGB 10603/Ratify Trial in Patients with Newly Diagnosed FLT3-Mutated Acute Myeloid Leukemia (AML). Blood. 2018;132(Suppl_1):2800. doi: 10.1182/blood-2018-99-112127.

  33. Сабурова И.Ю., Горбунова А.В., Слободнюк К.Ю. и др. Выявление внутренних тандемных дупликаций и мутации D835Y в гене FLT3 у пациентов с острым миелобластным лейкозом. Ученые записки Санкт-Петербургского государственного медицинского университета имени академика И.П. Павлова. 2010;XVII(3):48–51.

    [Saburova IYu, Gorbunova AV, Slobodnyuk KYu, et al. Detection of FLT3 internal tandem duplications and D835Y mutation in patients with acute myeloid leukemia. Uchenye zapiski Sankt-Peterburgskogo gosudarstvennogo meditsinskogo universiteta imeni akademika I.P. Pavlova. 2010;XVII(3):48–51. (In Russ)]

  34. Петрова Е.В., Мартынкевич И.С., Полушкина Л.Б. и др. Клинические, гематологические и молекулярно-генетические особенности острых миелобластных лейкозов с мутациями в генах FLT3, CKIT, NRAS и NPM1. Гематология и трансфузиология. 2016;61(2):72–80. doi: 10.18821/0234-5730-2016-61-2-72-80.

    [Petrova EV, Martynkevich IS, Polushkina LB, et al. Clinical, hematological and molecular-genetic features of acute myeloid leukemia with mutations in FLT3, CKIT, NRAS and NPM1. Russian journal of hematology and transfusiology. 2016;61(2):72–80. doi: 10.18821/0234-5730-2016-61-2-72-80. (In Russ)]

  35. Гук Л.В., Савицкая Т.В., Домнинский Д.А. и др. Анализ частоты и прогностического значения мутаций генов FLT3, c-KIT и NPM1 у детей с острым миелобластным лейкозом. Онкогематология. 2009;4(4):27–32.

    [Guk LV, Savitskaya TV, Domninskii DA, et al. Analysis of incidence and prognostic value of FLT3, c-KIT and NPM1 genes mutations in children with acute myeloid leukemia. Onkogematologiya. 2009;4(4):27–32. (In Russ)]

  36. Виноградов А.В. Разработка технологии детекции мутаций генов CDKN2A/ARF, FLT3, KIT, NPM1, NRAS, TET2, TP53, WT1 при острых миелоидных лейкозах. Российский онкологический журнал. 2013;4:34–5.

    [Vinogradov AV. Development of the technique for CDKN2A/ARF, FLT3, KIT, NPM1, NRAS, TET2, TP53, WT1 genes mutation detection in acute myeloid leukemia. Rossiiskii onkologicheskii zhurnal. 2013;4:34–5. (In Russ)]

  37. Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood. 1999;93(9):3074–80.

  38. Gari M, Abuzenadah A, Chaudhary A, et al. Detection of FLT3 oncogene mutations in acute myeloid leukemia using conformation sensitive gel electrophoresis. Int J Mol Sci. 2008;9(11):2194–204. doi: 10.3390/ijms9112194.

  39. Sly N, Gaspar K. Midostaurin for the management of FLT3-mutated acute myeloid leukemia and advanced systemic mastocytosis. Am J Health-Syst Pharm. 2019;76(5):268–74. doi: 10.1093/ajhp/zxy050.

  40. Bazarbachi AH, Hamed RA, Malard F, et al. Allogeneic transplant for FLT3-ITD mutated AML: a focus on FLT3 inhibitors before, during, and after transplant. Ther Adv Hematol. 2019;10:1–14. doi: 10.1177/2040620719882666.

  41. Levis M. FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematology Am Soc Hematol Educ Program. 2013;2013:220–6. doi: 10.1182/asheducation-2013.1.220.

Clinical Value of miR-3151 Overexpression in Synergistic Interaction with BAALC Host Gene in Patients with Acute Myeloid Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation

AI Shakirova, IM Barkhatov, AI Churkina, NN Mamaev, LS Zubarovskaya, BV Afanas’ev

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

For correspondence: Alena Igorevna Shakirova, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)338-62-72; e-mail: alyona.i.shakirova@gmail.com

For citation: Shakirova AI, Barkhatov IM, Churkina AI, et al. Clinical Value of miR-3151 Overexpression in Synergistic Interaction with BAALC Host Gene in Patients with Acute Myeloid Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2019;12(3):303–8 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-303-308


ABSTRACT

Background. Among a multitude of molecular genetic changes underlying acute myeloid leukemia (AML) disordered epigenetic regulation is of special importance. It includes expression change in miR-3151 gene forming a part of BAALC gene on chromosome 8 in q22.3 locus. At present BAALC gene overexpression is observed in a half of AML patients. A considerable part of them shows a combination of it with an increased transcriptional activity of miR-3151 gene, which is associated with the poorest AML prognosis.

Aim. To assess the prognostic value of miR-3151 overexpression in synergistic interaction with BAALC host gene in AML patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Materials & Methods. The trial included bone marrow samples taken from 10 healthy SCT donors and 29 AML patients after receiving allo-HSCT. Relative miR-3151 expression level and relative BAALC copy number were measured by quantitative real-time polymerase chain reaction.

Results. The analysis yielded a poor correlation between miR-3151 expression level and blast cell count in bone marrow (r = 0.330; = 0.005) as well as between the expression levels of miR-3151 and BAALC (r = 0.273; = 0.020). In addition, a great prognostic value of miR-315 overexpression in post-transplantation period was confirmed (= 0.005). Patients with miR-315 and BAALC co-expression in post-transplantation period have also the poorest prognosis than the control group with regard to both disease-free survival and relapse risks within 2 years after allo-HSCT.

Conclusion. Monitoring expression level of miR-3151 and its host gene BAALC in AML patients after receiving allo-HSCT seems to be important not only in AML prognosis but also in therapy efficacy evaluation.

Keywords: acute myeloid leukemia, miR-3151, BAALC, prognosis, allogeneic hematopoietic stem cell transplantation.

Received: October 22, 2018

Accepted: June 7, 2019

Read in PDF 


REFERENCES

  1. Testa U, Pelosi E. MicroRNAs expressed in hematopoietic stem/progenitor cells are deregulated in acute myeloid leukemias. Leuk Lymphoma. 2015;56(5):1466–74. doi: 3109/10428194.2014.955019.

  2. Liao Q, Wang B, Li X, Jiang G. miRNAs in acute myeloid leukemia. Oncotarget. 2017;8(2):3666–82. doi: 10.18632/oncotarget.12343.

  3. Ambros V. MicroRNAs: tiny regulators with great potential. Cell. 2001;107(7):823–6. doi: 1016/S0092-8674(01)00616-X.

  4. Marcucci G, Haferlach T, Dohner H. Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. J Clin Oncol. 2011;29(5):475–86. doi: 10.1200/JCO.2010.30.2554.

  5. Ehtesham N, Sharifi M. From conventional therapy toward microRNA-based therapy in acute promyelocytic leukemia. Adv Biomed Res. 2016;5:187. doi: 10.4103/2277-9175.190996.

  6. Li Z, Lu J, Sun M, et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci. 2008;105:15535–40. doi: 10.1073/pnas.0808266105.

  7. Dixon-McIver A, East P, Mein CA, et al. Distinctive patterns of microRNA expression associated with karyotype in acute myeloid leukaemia. PLoS One. 2008;3(5):е2141. doi: 10.1371/journal.pone.0002141.

  8. Jongen-Lavrencic M, Sun SM, Dijkstra MK, et al. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood. 2008;111(10):5078–85. doi: 10.1182/blood-2008-01-133355.

  9. Stark M, Tyagi S, Nancarrow D, et al. Characterization of the Melanoma miRNAome by Deep Sequencing. PLoS One. 2010;5(3):e9685. doi: 10.1371/journal.pone.0009685.

  10. Eisfeld A-K, Schwind S, Patel R, et al. Intronic miR-3151 within BAALC drives leukemogenesis by deregulating the TP53 Pathway. Sci Signal. 2014;7(321):ra36. doi: 10.1126/scisignal.2004762.

  11. Eisfeld A-K, Marcucci G, Maharry K, et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood. 2012;120(2):249–58. doi: 10.1182/blood-2012-02-408492.

  12. Diaz-Beya M, Brunet S, Nomdedeu J, et al. The expression level of BAALC-associated microRNA miR-3151 is an independent prognostic factor in younger patients with cytogenetic intermediate-risk acute myeloid leukemia. Blood Cancer J. 2015;5(10):e352. doi: 10.1038/bcj.2015.76.

  13. Weber S, Haferlach T, Alpermann T, et al. Feasibility of BAALC gene expression for detection of minimal residual disease and risk stratification in normal karyotype acute myeloid leukaemia. Br J Haematol. 2016;175(5):904–16. doi: 10.1111/bjh.14343.

  14. Shakirova A, Barkhatov I, Churkina A, et al. Prognostic significance of BAALC overexpression in patients with AML during the posttransplant period. Cellular Therapy and Transplantation. 2018;7(2):54–63. doi: 10.18620/ctt-1866-8836-2018-7-2–54-63.

  15. Schnerch D, Yalcintepe J, Schmidts A, et al. Cell cycle control in acute myeloid leukemia. Am J Cancer Res. 2012;2(5):508–28.

  16. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol. 2009;27(31):5195–201. doi: 10.1200/JCO.2009.22.4865.

  17. Мамаев Н.Н., Горбунова А.В., Бархатов И.М. и др. Молекулярный мониторинг течения острых миелоидных лейкозов по уровню экспрессии гена WT1 после аллогенной трансплантации гемопоэтических стволовых клеток. Клиническая онкогематология. 2015;8(3):309–20. doi: 10.21320/2500-2139-2015-8-3-309-320.

    [Mamaev NN, Gorbunova AV, Barkhatov IM, et al. Molecular Monitoring of WT1 Gene Expression Level in Acute Myeloid Leukemias after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2015;8(3):309–20. doi: 21320/2500-2139-2015-8-3-309-320. (In Russ)]

  18. Hosen N, Sonoda Y, Oji Y, et al. Very low frequencies of human normal CD34+ haematopoietic progenitor cells express the Wilms’ tumour gene WT1 at levels similar to those in leukaemia cells. Br J Haematol. 2002;116(2):409–20. doi: 10.1046/j.1365-2141.2002.03261.x.

  19. Ellisen LW, Carlesso N, Cheng T, et al. The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells. EMBO J. 2001;20(8):1897–909. doi: 10.1093/emboj/20.8.1897.

  20. Panyajai P, Amnajphook N, Keawsangthongcharoen S, et al. Study of Leukemic Stem Cell Population (CD34+/CD38-) and WT1 Protein Expression in Human Leukemic Cell Lines. J Assoc Med Sci. 2018;51(1):38–44. doi: 10.14456/jams.2018.5.

  21. Baldus C, Tanner S, Kusewitt D, et al. BAALC, a novel marker of human hematopoietic progenitor cells. Exp Hematol. 2003;31(11):1051–6. doi: 10.1016/j.exphem.2003.08.004.

  22. Najima Y, Ohashi K, Kawamura M, et al. Molecular monitoring of BAALC expression in patients with CD34-positive acute leukemia. Int J Hematol. 2010;91(4):636–45. doi: 10.1007/s12185-010-0550-8.

  23. Xiao S, Shen JZ, Huang JL, et al. Prognostic significance of the BAALC gene expression in adult patients with acute myeloid leukemia: A meta-analysis. Mol Clin Oncol. 2015;3(4):880–8. doi: 10.3892/mco.2015.562.

  24. Lucena-Araujo A, Pereira-Martins D, Koury L, et al. Clinical impact of BAALC expression in high-risk acute promyelocytic leukemia. Blood Adv. 2017;1(21):1807–14. doi: 10.1182/bloodadvances.2017005926.

Prediction of FLAG ± Ida Regimen Efficacy in Patients with Relapsed/Refractory Acute Myeloid Leukemia

IG Budaeva, EG Ovsyannikova, EN Goryunova, OV Kulemina, DV Zaitsev, DV Motorin, RSh Badaev, DB Zammoeva, VV Ivanov, KV Bogdanov, OS Pisotskaya, YuV Mirolyubova, TS Nikulina, YuA Alekseeva, AYu Zaritskey, LL Girshova

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

For correspondence: Irina Garmaevna Budaeva, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel.: +7(931)351-07-06; e-mail: irina2005179@mail.ru

For citation: Budaeva IG, Ovsyannikova EG, Goryunova EN, et al. Prediction of FLAG ± Ida Regimen Efficacy in Patients with Relapsed/Refractory Acute Myeloid Leukemia. Clinical oncohematology. 2019;12(3):289-96 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-289-296


ABSTRACT

Aim. To assess the efficacy of FLAG/FLAG-Ida regimen and to identify factors that influence remission, duration of disease-free survival (DFS) and overall survival (OS) of patients with relapsed/refractory acute myeloid leukemia (AML).

Materials & Methods. The trial included 54 patients (28 men and 26 women), median age was 37 years (range 18–70 years). 27 (50 %) out of 54 patients had refractory AML and 27 (50 %) patients had relapsed AML. FLAG and FLAG-Ida regimens were administered as induction therapy. 37 (68.5 %) patients received bone marrow transplantation. Molecular genetic and cytogenetic examinations were performed prior to therapy and on the 28th day after the start of treatment. WT1 gene expression was evaluated on the 14th–16th day of treatment.

Results. Complete remission (CR) was achieved in 42 (77.8 %) out of 54 patients. Refractoriness to therapy was observed in 9 (16.7 %) out of 54 patients, mortality was 5.5 % (3/54). Remission rate was higher in patients with relapsed AML compared with refractory AML: 85.2 % (23/27) and 70.4 % (19/27), respectively. On the 14th–16th day of treatment patients with blast cell count ≥ 10 % in bone marrow (BM) showed significantly lower CR rate (60 %) compared with the group of patients with < 10 % blast cells in BM (89.6 %; = 0.024) and shorter DFS (median 7.6 vs. 17.6 months, respectively; = 0.03). Median DFS in patients with WT1 expression reduction to < 1 log on the 14th–16th day was 5 vs. 18 months in patients without WT1 expression reduction (= 0.01). DFS varied in groups of patients with blast cell count < 10 % in BM on the 14th–16th day of treatment based on the level of WT1 expression reduction (= 0.04). MRD-negative patients (57.1 %) showed significantly longer DFS and OS compared with MRD-positive patients (median DFS was 17.6 vs. 5.2 months, respectively, = 0.02; median OS was 19 vs. 6.9 months, = 0.0002). Median DFS and OS were different only in ELN low- and high-risk groups (median not reached vs. 5.2 months, respectively, = 0.039; median not reached vs. 10.2 months, = 0.039).

Conclusion. FLAG and FLAG-Ida are effective and safe regimens in the treatment of relapsed/refractory AML. Achieving remission depends on neither the risk group nor the time of relapse occurrence. The blast cell count in BM on the 14th–16th day of FLAG/FLAG-Ida treatment is a prognostic factor determining achievement and duration of remission. WT1 expression level in the early post-induction period is a sensitive DFS marker. MRD status and molecular genetic risk (ELN) group affiliation are essential prognostic factors determining DFS and OS.

Keywords: acute myeloid leukemia, relapse, refractoriness, FLAG and FLAG-Ida regimens.

Received: November 2, 2018

Accepted: May 28, 2019

Read in PDF 


REFERENCES

  1. Papaemmanuil E, Gerstung M, Bullinger L. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–21. doi: 10.1056/NEJMoa1516192.

  2. Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol. 2003;21(24):4642–9. doi: 10.1200/JCO.2003.04.036.

  3. Dohner H, Elihu H, Estey EH, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.

  4. Othus M, Appelbaum FR, Petersdorf SH, et al. Fate of patients with newly diagnosed acute myeloid leukemia who fail primary induction therapy. Biol Blood Marrow Transplant. 2015;21(3):559–64. doi: 10.1016/j.bbmt.2014.10.025

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

  6. Elihu H, Estey E. Acute myeloid leukemia: 2016 Update on risk-stratification and management. Am J Hematol. 2016;91(8):824–46. doi: 10.1002/ajh.24439.

  7. Biggs JC, Horowitz MM, Gale RP, et al. Bone marrow transplants may cure patients with acute leukemia never achieving remission with chemotherapy. Blood. 1992;80(4):1090–3.

  8. Duval M, Klein JP, He W, et al. Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure. J Clin Oncol. 2010;28(23):3730–8. doi: 10.1200/JCO.2010.28.8852.

  9. Sureda A. Indications for allo- and auto-SCT for hematological diseases, solid tumours and immune disorders: current practice in Europe. Bone Marrow Transplant. 2015;50(8):1037–56. doi: 10.1038/bmt.2015.6.

  10. Araki D, Othus M, Walter RB, et al. Effect of allogeneic hematopoietic cell transplantation in first complete remission on post-relapse complete remission rate and survival in acute myeloid leukemia. Haematologica. 2015;100(7):254–6. doi: 10.3324/haematol.2014.

  11. Delia M, Pastore D, Carluccio P, et al. FLAG-Ida regimen as bridge therapy to allotransplant in refractory/relapsed AML patients. Clin Lymph Myel Leuk. 2017;17(11):767–773. doi: 10.1016/j.clml.2017.06.002.

  12. Estey E, Kornblau S, Pierce S, et al. A stratification system for evaluating and selecting therapies in patients with relapsed or primary refractory acute myelogenous leukemia. Blood. 1996;88(2):756.

  13. Estey EH. Treatment of relapsed and refractory acute myelogenous leukemia. Leukemia. 2000;14(3):476–9. doi: 10.1038/sj.leu.2401568.

  14. Estey E, Plunkett W, Gandhi V, et al. Fludarabine and arabinosylcytosine therapy for refractory and relapsed acute myelogenous leukemia. Leuk Lymphoma. 1993;9(4–5):343–50. doi: 10.3109/10428199309148532.

  15. Estey E, Thall P, Andreeff M, et al. Use of granulocyte colony-stimulating factor before, during, and after fludarabine plus cytarabine induction therapy of newly diagnosed acute myelogenous leukemia or myelodysplastic syndromes; comparison with fludarabine plus cytarabine without granulocyte colony-stimulating factor. J Clin Oncol. 1994;12(4):671–8. doi: 10.1200/JCO.1994.12.4.671.

  16. Gandhi V, Plunkett W. Modulation of arabinosylnucleoside metabolism by arabinosylnucleotides in human leukemia cells. Cancer Res. 1988;48(2):329–34.

  17. Gandhi V, Estey E, Keating MJ, et al. Fludarabine potentiates metabolism of cytarabine in patients with acute myelogenous leukemia during therapy. J Clin Oncol. 1993;11(1):116–24. doi: 10.1200/JCO.1993.11.1.116.

  18. Anderlini P. Idarubicin cardiotoxicity: A retrospective study in acute myeloid leukemia and myelodysplasia. J Clin Oncol. 1995;13(11):2827–34. doi: 10.1200/JCO.1995.13.11.2827.

  19. Lee SR, Yang DH, Ahn JS, et al. The Clinical outcome of FLAG chemotherapy without idarubicin in patients with relapsed or refractory acute myeloid leukemia. J Korean Med Sci. 2009;24(3):498–503. doi: 10.3346/jkms.2009.24.3.498.

  20. Dohner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med. 2015;373(12):1136–52. doi: 10.1056/NEJMra1406184.

  21. Patel JP, Gonen M, Figueroa ME. Prognostic Relevance of Integrated Genetic Profiling in Acute Myeloid Leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/NEJMoa1112304.

  22. Wang LJ, Ding J, Zhu CY, et al. Clinic outcome of FLAG regimen treating patients with refractory and relapse acute myeloid leukemia. J Exper Hematol. 2016;24(1):19–24.

  23. Jun Xu, Ting-Ting Lv, Xiao-Fen Zhou, et al. Efficacy of common salvage chemotherapy regimens in patients with refractory or relapsed acute myeloid leukemia: A retrospective cohort study. Medicine. 2018;97(39): doi: 10.1097/MD.0000000000012102.

  24. Breems DA, Van Putten WL, Huijgens PC, et al. Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol. 2005;23(9):1969–78. doi: 10.1200/jco.2005.06.027.

  25. Carella AM, Cascavilla N, Greco MM, et al. Treatment of poor risk acute myeloid leukemia with fludarabine, cytarabine and G-CSF (flag regimen): a single center study. Leuk Lymphoma. 2001;40(3–4):295–303. doi: 10.3109/10428190109057928.

  26. Ferrara F, Palmieri S, Pocali B, et al. De novo acute myeloid leukemia with multilineage dysplasia: treatment results and prognostic evaluation from a series of 44 patients treated with fludarabine, cytarabine and G-CSF (FLAG). Eur J Haematol. 2002;68(4):203–9. doi: 10.1034/j.1600-0609.2002.01651.x.

  27. Bao Y, Zhao J, Li Z-Z. Comparison of clinical remission and survival between CLAG and FLAG induction chemotherapy in patients with refractory or relapsed acute myeloid leukemia: a prospective cohort study. Clin Transl Oncol. 2018;20(7):870–80. doi: 10.1007/s12094-017-1798-8.

  28. Ossenkoppele GJ, Graveland WJ, Sonneveld P, et al. The value of fludarabine in addition to ARA-C and G-CSF in the treatment of patients with high-risk myelodysplastic syndromes and AML in elderly patients. Blood. 2004;103(8):2908–13. doi: 10.1182/blood-2003-07-2195.

  29. Jackson G, Taylor P, Smith GM, et al. A multicentre, open, non-comparative phase II study of a combination of fludarabine phosphate, cytarabine and granulocyte colony-stimulating factor in relapsed and refractory acute myeloid leukaemia and de novo refractory anaemia with excess of blasts in transformation. Br J Haematol. 2001;112(1):127–37. doi: 1046/j.1365-2141.2001.02551.x.

  30. Virchis A, Koh M, Rankin P, et al. Fludarabine, cytosine arabinoside, granulocyte-colony stimulating factor with or without idarubicin in the treatment of high risk acute leukaemia or myelodysplastic syndromes. Br J Haematol. 2004;124(1):26–32. doi: 10.1046/j.1365-2141.2003.04728.x.

  31. Farooq MU, Mushtaq F, Farooq A, et al. FLAG vs FLAG-IDA: outcomes in relapsed/refractory acute leukemias. Cancer Chemother Pharmacol. 2019;83(2):1–2. doi: 10.1007/s00280-019-03792-8.

  32. Heinemann V, Murray D, Walters R, et al. Mitoxantrone-induced DNA damage in leukemia cells is enhanced by treatment with high-dose arabinosylcytosine. Cancer Chemother Pharmacol. 1988;22(3):205–10. doi: 10.1007/BF00273412.

  33. Loughlin S, Gandhi V, Plunkett W, et al. The effect of 9-beta-D-arabinofuranosyl-2-fluoroadenine and 1-beta-D-arabinofuranosylcytosine on the cell cycle phase distribution, topoisomerase II level, mitoxantrone cytotoxicity, and DNA strand break production in K562 human leukemia cells. Cancer Chemother Pharmacol. 1996;38(3):261–8. doi: 10.1007/s002800050480.

  34. Gabert J, Beillard E, Velden VH, et al. Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia – a Europe Against Cancer program. Leukemia. 2003;17(12):2318–57. doi: 10.1038/sj.leu.2403135.

  35. Willasch AM, Gruhn B, Coliva T, et al. Combined usage of Wilms’ tumor gene quantitative analysis and multiparameter flow cytometry for minimal residual disease monitoring of acute myeloid leukemia patients after allogeneic hematopoietic stem cells transplantation. Exp Ther Med. 2018;15(2):1403–9. doi: 10.3892/etm.2017.5547.

  36. Богданов К.В., Моторин Д.В., Никулина Т.С. и др. Мониторинг донорского химеризма и минимальной остаточной болезни у онкогематологических больных после аллогенной трансплантации гемопоэтических стволовых клеток. Биомедицинская химия. 2017;63(6):570–81. doi: 10.18097/PBMC

    [Bogdanov KV, Motorin DV, Nikulina TS, et al. Donor chimerism and minimal residual disease monitoring in leukemia patients after allo-HSCT. Biomeditsinskaya khimiya. 2017;63(6):570–81. doi: 10.18097/PBMC20176306570. (In Russ)]

  37. Pastore D, Specchia G, Carluccio P, et al. FLAG-IDA in the treatment of refractory/relapsed acute myeloid leukemia: single-center experience. Ann Hematol. 2003;82(4):231–5. doi: 10.1007/s00277-003-0624-2.

  38. Montillo M, Mirto S, Petti MC, et al. Fludarabine, cytarabine, and G-CSF (FLAG) for the treatment of poor risk acute myeloid leukemia. Am J Hematol. 1998;58(2):105–9. doi: 1002/(sici)1096-8652(199806)58:2<105::aid-ajh3>3.0.co;2-w.

  39. Nokes TJ, Johnson S, Harvey D, et al. FLAG is a useful regimen for poor prognosis adult myeloid leukaemias and myelodysplastic syndromes. Leuk Lymphoma. 1997;27(1–2):93–101. doi: 10.3109/10428199709068275.

Preventive Use of Azacitidine in Patients with Acute Myeloid Leukemia after Haploidentical Allo-BMT

RSh Badaev, DB Zammoeva, LL Girshova, DV Babenetskaya, NA Il’ina, YuA Alekseeva, AYu Zaritskey, DV Motorin

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

For correspondence: Dmitrii Vasil’evich Motorin, MD, PhD, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; e-mail: almazov-bmt@mail.ru

For citation: 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


ABSTRACT

Background. Haploidentical bone marrow transplantation (BMT) can be a reliable alternative if a fully matched donor is not available. The main challenges after BMT are a relapse of major disease, graft-versus-host disease (GVHD), and infections. Azacitidine possesses antileukemic effect together with immunomodulating properties and being administered soon after BMT can significantly improve the outcome.

Aim. To study azacitidine effect on the outcome of haploidentical BMT in patients with acute myeloid leukemia (AML) in the early post-transplantation period.

Materials & Methods. The trial included 18 AML patients who received haploidentical BMT at VA Almazov National Medical Research Center. In all patients MRD-negative remission was achieved on the 30th day after BMT. Azacitidine therapy was initiated not earlier than 2 months after BMT with a complete engraftment of transplant and no GVHD. Azacitidine 100 mg/day was administered on D1–D5 every 28 days within a year after BMT. When a molecular relapse was detected, donor lymphocytes were additionally infused during every other cycle of therapy.

Results. Eleven patients received preventive azacitidine treatment, 7 patients were included in control group. Median onset of azacitidine treatment after haploidentical BMT was 4 months (range 2–10 months), median number of azacitidine courses was 3.5 (range 1–9). During azacitidine treatment acute GVHD was identified in 5 (45.4 %) patients. In 4 of them an exacerbation of earlier GVHD was detected (3 with cutaneous form and 1 with intestinal form), and only in 1 patient de novo acute intestinal GVHD was discovered.

Conclusion. Azacitidine treatment of AML patients after haploidentical allo-BMT is safe and well tolerated. Preventive azacitidine treatment after haploidentical BMT improves overall survival of AML patients.

Keywords: haploidentical allogeneic bone marrow transplantation, azacitidine, acute myeloid leukemia.

Received: June 22, 2018

Accepted: December 11, 2018

Read in PDF 


REFERENCES

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

  2. McCurdy SR, Kanakry JA, Showel MM, et al. Risk-stratified outcomes of nonmyeloablative HLA-haploidentical BMT with high-dose posttransplantation cyclophosphamide. Blood. 2015;125(19):3024–31. doi: 10.1182/blood-2015-01-623991.

  3. Ciurea SO, Zhang M-J, Bacigalupo AA, et al. Haploidentical transplant with posttransplant cyclophosphamide vs matched unrelated donor transplant for acute myeloid leukemia. Blood. 2015;126(8):1033–40. doi: 10.1182/blood-2015-04-639831.

  4. Bashey A, Zhang X, Jackson K, et al. Comparison of Outcomes of Hematopoietic Cell Transplants from T-Replete Haploidentical Donors Using Post-Transplantation Cyclophosphamide with 10 of 10 HLA-A, -B, -C, -DRB1, and -DQB1 Allele-Matched Unrelated Donors and HLA-Identical Sibling Donors: A Multivariable Analysis Including Disease Risk Index. Biol Blood Marrow Transplant. 2016;22(1):125–33. doi: 10.1016/j.bbmt.2015.09.002.

  5. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126(3):291–9. doi: 10.1182/blood-2015-01-621664.

  6. Pozzi S, Geroldi S, Tedone E, et al. Leukaemia relapse after allogeneic transplants for acute myeloid leukaemia: predictive role of WT1 expression. Br J Haematol. 2013;160(4):503–9. doi: 10.1111/bjh.12181.

  7. Glucksberg H, Storb R, Fefer A, et al. Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors. Transplantation. 1974;18(4):295–304. doi: 10.1097/00007890-197410000-00001.

  8. Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. Diagnosis and Staging Working Group Report. Biol Blood Marrow Transplant. 2005;11(12):945–56. doi: 10.1016/j.bbmt.2005.09.004.

  9. Chang Y-J, Wang Y, Liu Y-R, et al. Haploidentical allograft is superior to matched sibling donor allograft in eradicating pre-transplantation minimal residual disease of AML patients as determined by multiparameter flow cytometry: a retrospective and prospective analysis. J Hematol Oncol. 2017;10(1):134. doi: 10.1186/s13045-017-0502-3.

  10. Frassoni F, Barrett AJ, Granena A, et al. Relapse after allogeneic bone marrow transplantation for acute leukaemia: a survey by the E.B.M.T. of 117 cases. Br J Haematol. 1988;70(3):317–20. doi: 10.1111/j.1365-2141.1988.tb02488.x.

  11. Bosi A, Laszlo D, Labopin M, et al. Second Allogeneic Bone Marrow Transplantation in Acute Leukemia: Results of a Survey by the European Cooperative Group for Blood and Marrow Transplantation. J Clin Oncol. 2001;19(16):3675–84. doi: 10.1200/jco.2001.19.16.3675.

  12. Verdonck L, Petersen E, Lokhorst H, et al. Donor leukocyte infusions for recurrent hematologic malignancies after allogeneic bone marrow transplantation: impact of infused and residual donor T cells. Bone Marrow Transplant. 1998;22(11):1057–63. doi: 10.1038/sj.bmt.1701496.

  13. Collins RH, Shpilberg O, Drobyski WR, et al. Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation. J Clin Oncol. 1997;15(2):433–44. doi: 10.1200/jco.1997.15.2.433.

  14. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Azacitidine Prolongs Overall Survival Compared With Conventional Care Regimens in Elderly Patients With Low Bone Marrow Blast Count Acute Myeloid Leukemia. J Clin Oncol. 2010;28(4):562–9. doi: 10.1200/jco.2009.23.8329.

  15. Maurillo L, Venditti A, Spagnoli A, et al. Azacitidine for the treatment of patients with acute myeloid leukemia. Cancer. 2011;118(4):1014–22. doi: 10.1002/cncr.26354.

  16. Schroeder T, Czibere A, Platzbecker U, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013;27(6):1229–35. doi: 10.1038/leu.2013.7.

  17. Tessoulin B, Delaunay J, Chevallier P, et al. Azacitidine salvage therapy for relapse of myeloid malignancies following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2014;49(4):567–71. doi: 10.1038/bmt.2013.233.

  18. Craddock C, Labopin M, Robin M, et al. Clinical activity of azacitidine in patients who relapse after allogeneic stem cell transplantation for acute myeloid leukemia. Haematologica. 2016;101(7):879–83. doi: 10.3324/haematol.2015.140996.

  19. Schroeder T, Rachlis E, Bug G, et al. Treatment of Acute Myeloid Leukemia or Myelodysplastic Syndrome Relapse after Allogeneic Stem Cell Transplantation with Azacitidine and Donor Lymphocyte Infusions—A Retrospective Multicenter Analysis from the German Cooperative Transplant Study Group. Biol Blood Marrow Transplant. 2015;21(4):653–60. doi: 10.1016/j.bbmt.2014.12.016.

  20. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia. 2011;26(3):381–9. doi: 10.1038/leu.2011.234.

  21. Schroeder T, Frobel J, Cadeddu R-P, et al. Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation. Leukemia. 2013;27(9):1910–3. doi: 10.1038/leu.2013.64.

  22. Goodyear OC, Dennis M, Jilani NY, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood. 2012;119(14):3361–9. doi: 10.1182/blood-2011-09-377044.

  23. Choi J, Ritchey J, Prior JL, et al. In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood. 2010;116(1):129–39. doi: 10.1182/blood-2009-12-257253.

  24. Cooper ML, Choi J, Karpova D, et al. Azacitidine Mitigates Graft-versus-Host Disease via Differential Effects on the Proliferation of T Effectors and Natural Regulatory T Cells In Vivo. J Immunol. 2017;198(9):3746–54. doi: 10.4049/jimmunol.1502399.

  25. Garcia-Delgado R, de Miguel D, Bailen A, et al. Effectiveness and safety of different azacitidine dosage regimens in patients with myelodysplastic syndromes or acute myeloid leukemia. Leuk Res. 2014;38(7):744–50. doi: 10.1016/j.leukres.2014.03.004.

  26. Lyons RM, Cosgriff TM, Modi SS, et al. Hematologic Response to Three Alternative Dosing Schedules of Azacitidine in Patients With Myelodysplastic Syndromes. J Clin Oncol. 2009;27(11):1850–6. doi: 10.1200/jco.2008.17.1058.

  27. Jabbour E, Giralt S, Kantarjian H, et al. Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia. Cancer. 2009;115(9):1899–905. doi: 10.1002/cncr.24198.

  28. Jabbour E, Short NJ, Montalban-Bravo G, et al. Randomized phase 2 study of low-dose decitabine vs low-dose azacitidine in lower-risk MDS and MDS/MPN. Blood. 2017;130(13):1514–22. doi: 10.1182/blood-2017-06-788497.

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

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

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

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

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

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


ABSTRACT

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

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

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

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

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

Received: January 20, 2018

Accepted: April 18, 2018

Read in PDF 


REFERENCES

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

The Role of Hypomethylating Agents Prior to Allogeneic Hematopoietic Stem Cells Transplantation in Acute Myeloid Leukemia and Myelodysplastic Syndrome

VN Ovechkina1, SN Bondarenko1, EV Morozova1, IS Moiseev1, AA Osipova1, TL Gindina1, AI Shakirova1, TA Bykova1, AD Kulagin1, IA Samorodova2, EV Karyakina3, EA Ukrainchenko4, LS Zubarovskaya1, BV Afanas’ev1

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

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

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

4 Aleksandrov Hospital, 4 Solidarnosti pr-t, Saint Petersburg, Russian Federation, 193312

For correspondence: Varvara Nikolaevna Ovechkina, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)338-62-72; e-mail ovetchkina@gmail.com

For citation: 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 (In Russ).

DOI: 10.21320/2500-2139-2017-10-3-351-357


ABSTRACT

Background & Aims. The aim of the study was to evaluate the efficacy and safety of azacytidine and decitabine prior to allogeneic hematopoietic stem cell transplantation (allo-HSCT) in acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia.

Materials & Methods. The research included 62 patients who received hypomethylating agents (HMA) prior to allo-HSCT. The median age was 28 years (range from 1 to 68 years), the study population consisted of 27 (43.5 %) women and 35 (56.5 %) men.

Results. The overall response (complete + partial remission) was observed in 42 % (n = 26) of cases. At the time of allo-HSCT no disease progression was observed in 41 (66 %) patients. The multivariant analysis showed the overall survival (OS) statistically significantly increased with the graft retention (hazard ratio [HR] 0.002; 95% confidence interval [95% CI] 0.001–0.74; p = 0.03), and also with the administration of HMA after allo-HSCT (HR 0.24; 95% CI 0.08–0.67; p = 0.007). The response (stabilisation, partial or complete remission) due to HMA administration prior to allo-HSCT (HR 6.4; 95% CI 0.75–54.0; p = 0.08) was associated with improved OS. The event-free survival (EFS) was significantly higher with the response to azacytidine and decitabine at the time of allo-HSCT (HR 38.9; 95% CI 1.3–1198.0; p = 0.03) and with the graft retention (HR 0.02; 95% CI 0.005–0.1; p = 0.001). In patients with MDS compared with AML (HR 2.3; 95% CI 0.9–22.0; p = 0.08), there was a tendency to EFS improvement. Progression-free survival rates were higher in patients with a number of blast cells in the bone marrow less than 31 % at the time of diagnosis (HR 1.1; 95% CI 1.1–9.9; p = 0.01).

Conclusion. The use of azacytidine and decitabine prior to allo-HSCT allows to safely control the tumor mass in patients with MDS and to maintain the achieved remission with AML. In patients with a response to HMA, the best OS and EFS values are seen after allo-HSCT.

Keywords: acute myeloid leukemia, myelodysplastic syndrome, allogeneic hematopoietic stem cell transplantation, hypomethylating agents, azacitidine, decitabine.

Received: December 19, 2016

Accepted: March 9, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Ширин А.Д., Баранова О.Ю. Гипометилирующие препараты в онкогематологии. Клиническая онкогематология. 2016;9(4):369–82. doi: 10.21320/2500-2139-2016-9-4-369–382.
    [Shirin AD, Baranova OYu. Hypomethylating Agents in Oncohematology. Clinical oncohematology. 2016;9(4):369–82. doi: 10.21320/2500-2139-2016-9-4-369–382. (In Russ)]
  2. Бондаренко С.Н., Семенова Е.В., Афанасьев Б.В. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при остром миелобластном лейкозе в первой ремиссии. Терапевтический архив. 2013;84(7):18–25.
    [Bondarenko SN, Semenova EV, Afanas’ev BV, et al. Allogeneic hematopoietic stem cell transplantation for acute myeloblastic leukemia in first remission. Terapevticheskii arkhiv. 2013;84(7):18–25. (In Russ)]
  3. Паровичникова Е.Н., Троицкая В.В., Савченко В.Г. и др. Лечение больных острыми миелоидными лейкозами по протоколу российского многоцентрового рандомизированного исследования ОМЛ-01.10: результаты координационного центра. Терапевтический архив. 2014;86(7):14–23.
    [Parovichnikova EN, Troitskaya VV, Savchenko VG, et al. Treating patients with acute myeloid leukemias according to the protocol of the AML-01.10 Russian multicenter randomized trial: the Coordinating Center’s results. Terapevticheskii arkhiv. 2014;86(7):14–23. (In Russ)]
  4. de Witte T, Bowen D, Robin M, et al. Allogeneic hematopoietic stem cell transplantation for MDS and CMML: recommendations from an international expert panel. Blood. 2017;129(13):1753–62. doi: 10.1182/blood-2016-06-724500.
  5. Sohn SK, Moon JH. Survey of expert opinions and related recommendations regarding bridging therapy using hypomethylating agents followed by allogeneic transplantation for high-risk MDS. Crit Rev Oncol Hematol. 2015;95(2):243–50. doi: 10.1016/j.critrevonc.2015.03.004.
  6. Al-Ali HK, Jaekel N, Niederwieser D, et al. Azacitidine in patients with acute myeloid leukemia medically unfit for or resistant to chemotherapy: a multicenter phase I/II study. Leuk Lymphoma. 2012;53(1):110–7. doi: 10.3109/10428194.2011.606382.
  7. Cruijsen M, Lubbert M, Huls G, et al. Clinical Results of Hypomethylating Agents in AML Treatment. J Clin Med. 2014;4(1):1–17. doi: 10.3390/jcm4010001.
  8. Field T, Perkins J, Anasetti C, 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.
  9. Al-Ali HK, Jaekel N, Niederwieser D. The role of hypomethylating agents in the treatment of elderly patients with AML. J Geriatr Oncol. 2014;5(1):89–105. doi: 10.1016/j.jgo.2013.08.004.
  10. Komrokji RS, DeZern AE, Sekeres MA, et al. Validation of International Working Group (IWG) Response Criteria in Higher-Risk Myelodysplastic Syndromes (MDS): A Report on Behalf of the MDS Clinical Research Consortium (MDS CRC). Blood. 2015;126:909.
  11. Seymour JF, Buckstein R, Santini V, et al. Efficacy and Safety of Azacitidine (AZA) Versus Conventional Care Regimens (CCR) in Patients Aged ≥ 75 Years with Acute Myeloid Leukemia (AML) in the Phase 3 AZA-AML-001 Study. Blood. 2016;128:2818.
  12. Garcia JS, Jain N, Godley LA. An update on the safety and efficacy of decitabine in the treatment of myelodysplastic syndromes. Onco Targets Ther. 2010;3:1–13. doi: 10.2147/ott.s7222.
  13. Кострома И.И., Грицаев С.В., Карягина Е.В. и др. Гематологическое улучшение — вариант благоприятного противоопухолевого ответа на лечение азацитидином при острых миелоидных лейкозах и миелодиспластических синдромах. Клиническая онкогематология. 2015;8(4):413–9. doi: 10.21320/2500-2139-2015-8-4-413-419.
    [Kostroma II, Gritsaev SV, Karyagina EV, et al. Hematological Improvement is a Favorable Response to Azacitidine in Patients with Acute Myeloid Leukemias and Myelodysplastic Syndromes. Clinical oncohematology. 2015;8(4):413–9. doi: 10.21320/2500-2139-2015-8-4-413-419. (In Russ)]
  14. Potter VT, Iacobelli S, Biezen A, et al. Comparison of Intensive Chemotherapy and Hypomethylating Agents before Allogeneic Stem Cell Transplantation for Advanced Myelodysplastic Syndromes: A Study of the Myelodysplastic Syndrome Subcommittee of the Chronic Malignancies Working Party of the European Society for Blood and Marrow Transplant Research. Biol Blood Marrow Transplant. 2016;22(9):1615–20. doi: 10.1016/j.bbmt.2016.05.026.
  15. Jabbour E, Mathisen MS, Garcia-Manero G, et al. Allogeneic hematopoietic stem cell transplantation versus hypomethylating agents in patients with myelodysplastic syndrome: A retrospective case-control study. Am J Hematol. 2013;88(3):198–200. doi: 10.1002/ajh.23371.
  16. 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.
  17. Waespe N, Akker Van Den M, Klaassen RJ, et al. Response to treatment with azacitidine in children with advanced myelodysplastic syndrome prior to hematopoietic stem cell transplantation. Haematologica. 2016;101(12):1508–15. doi: 10.3324/haematol.2016.145821.
  18. Prebet Th, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. J Clin Oncol. 2011;29(24):3322–7. doi: 10.1200/jco.2011.35.8135.
  19. Bally C, Thepot S, Quesnel B, et al. Azacitidine in the treatment of therapy related myelodysplastic syndrome and acute myeloid leukemia (tMDS/AML): A report on 54 patients by the Groupe Francophone Des Myelodysplasies (GFM). Leuk Res. 2013;37(6):637–40. doi: 10.1016/j.leukres.2013.02.014.
  20. Fenaux P, Mufti GJ, Peterson BL, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomized, open-label, phase III study. Lancet Oncol. 2009;10(3):223–32. doi: 10.1016/s1470-2045(09)70003-8.
  21. Quintas-Cardama A, Ravandi F, Liu-Dumlao Th, et al. Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia. Blood. 2012;120(24):4840–5. doi: 10.1182/blood-2012-06-436055.
  22. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28(4):562–9. doi: 10.1200/jco.2009.23.8329.
  23. Pleyer L, Burgstaller B, Greil R, et al. Azacitidine front-line in 339 patients with myelodysplastic syndromes and acute myeloid leukaemia: comparison of French-American-British and World Health Organization classifications. J Hematol Oncol. 2016;9(1):39. doi: 10.1186/s13045-016-0263-4.
  24. Yahng SA, Yooh JH, Shin SH, et al. Response to pretransplant hypomethylating agents influences the outcome of allogeneic hematopoietic stem cell transplantation in adults with myelodysplastic syndromes. Eur J Haematol. 2013;90(2):111–20. doi: 10.1111/ejh.12038.
  25. Овечкина В.Н., Бондаренко С.Н., Морозова Е.В. и др. Острый миелобластный лейкоз и миелодиспластический синдром: применение азацитидина с профилактической и превентивной целью после аллогенной трансплантации гемопоэтических стволовых клеток. Клиническая онкогематология. 2017;10(1):45–51. doi: 10.21320/2500-2139-2017-10-1-45-51.
    [Ovechkina VN, Bondarenko SN, Morozova EV, et al. Acute Myeloblastic Leukemia and Myelodysplastic Syndrome: Azacitidine for Prophylactic and Preventive Purposes after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2017;10(1):45–51. doi: 10.21320/2500-2139-2017-10-1-45-51. (In Russ)]
  26. Craddock Ch, Jilani N, Siddique Sh, 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.

Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias with Hyperdiploid Karyotype

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

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

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

For citation: Gindina TL, Mamaev NN, Nikolaeva ES, et al. Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias with Hyperdiploid Karyotype. Clinical oncohematology. 2016;9(4):383–90 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-383-390


ABSTRACT

Aim. To evaluate the prognostic impact of the different cytogenetic characteristics, including the modal number, the number of chromosomal aberrations in a complex karyotype, and adverse chromosomal abnormalities (ACA) (–7/7q–, –5/5q–, –17/17p–, t(6;9)(p22;q34)) on the results of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with hyperdiploid acute myeloid leukemia (H-AML).

Methods. Forty seven H-AML patients (21 women and 26 men, aged from 1 to 58 years, median — 23.9 years) were examined. The analysis of overall (OS) and event-free survival (EFS) predictors after allo-HSCT in patients with different clinical, transplant and cytogenetic characteristics was performed.

Results. The modal number of chromosomes (MN) of 47–48 was the most common one in the karyotype which was observed in 31 (66 %) patients. High hyperdiploidy with the modal number of 49–65 was identified in 13 (28 %) patients, near-triploid and near-tetraploid karyotypes were found in 3 (6 %) patients. Quantitative chromosomal abnormalities were nonrandom. Chromosome 8 (50 %), 21 (32 %), 13 (16 %) и 22 (16 %) trisomy was the most common one. Structural chromosomal abnormalities were detected in 22 (47 %) patients, at that, ACA were found in 7 (19 %) patients. In univariate analysis, the OS and EFS after allo-HSCT differed in patients with different clinical status (remission vs. active disease; = 0.003 and = 0.002, respectively), different chromosomal abnormalities in hyperdiploid karyotype (ACA– vs. ACA+; = 0.001 and = 0.03, respectively). An additional analysis of selected patients group with a structurally complex karyotype (n = 19) showed, that patients without ACA had a higher OS than patients with ACA (= 0.03). In multivariate analysis, the disease status (relapse) at allo-HSCT was an independent predictor of decreased OS and EFS (= 0.004 и = 0.006, respectively), as well as the presence of the ACA (= 0.002 only for OS).

Conclusion. ACA were high-risk factors in H-AML patients received allo-HSCT. Therefore, the patients with formal criteria of a complex karyotype should not be automatically included in the cytogenetic unfavorable risk group.


Keywords: hyperdiploid and complex karyotypes, acute myeloid leukemia, allogeneic hematopoietic stem cell transplantation, prognosis.

Received: April 17, 2016

Accepted: May 5, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Chilton L, Hills RK, Harrison CJ, et al. Hyperdiploidy with 49-65 chromosomes represents a heterogeneous cytogenetic subgroup of acute myeloid leukemia with differential outcome. Leukemia. 2013;28(2):321–8. doi: 1038/leu.2013.198.
  2. Sandahl JD, Kjeldsen E, Abrahamsson J, et al. Ploidy and clinical characteristics of childhood acute myeloid leukemia: a NOPHO-AML study. Genes Chromos Cancer. 2014;53(8):667–75. doi: 1002/gcc.22177.
  3. Stolzel F, Mohr B, Kramer M, et al. Karyotype complexity and prognosis in acute myeloid leukemia. Blood Cancer J. 2016;6:e386. doi: 101038/bcj.2015.114.
  4. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 1182/blood-2009-07-235358.
  5. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116(3):354–65. doi: 1182/blood-2009-11-254441.
  6. Гиндина Т.Л., Мамаев Н.Н., Бархатов И.М. и др. Сложные повреждения хромосом у больных с рецидивами острых лейкозов после аллогенной трансплантации гемопоэтических стволовых клеток. Терапевтический архив. 2012;8:61–6.
    [Gindina TL, Mamaev NN, Barkhatov IM, et al. Complex chromosome damages in patients with recurrent acute leukemias after allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2012;8:61–6. (In Russ)]
  7. Schaffer L, McGovan-Jordan J, Schmid M. An International System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013. pp. 140. doi: 10.1002/ajmg.a.35995.
  8. Guo RJ, Atenafu EG, Craddock K, et al. Allogeneic hematopoietic cell transplantation may alleviate the negative prognostic impact of monosomal and complex karyotypes on patients with acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(5):690–5. doi: 1016/j.bbmt.2014.01.027.

Genetic Mutations in Acute Myeloid Leukemia

OV Blau

Charite Clinic, Berlin Medical University, 30 Hindenburgdamm, Berlin, Germany, 12200

For correspondence: Ol’ga Vladimirovna Blau, DSci, Department of Hematology, Oncology and Tumorimmunology, Charite University School of Medicine, Hindenburgdamm 30, 12200, Berlin, Germany; e-mail: olga.blau@charite.de.

For citation: Blau OV. Genetic Mutations in Acute Myeloid Leukemia. Clinical oncohematology. 2016;9(3):245-56 (In Russ).

DOI: 10.21320/2500-2139-2016-9-3-245-256


ABSTRACT

Acute myeloid leukemia (AML) is a clonal malignancy characterized by ineffective hematopoiesis. Most AML patients present different cytogenetic and molecular defects associated with certain biologic and clinical features of the disease. Approximately 50–60 % of de novo AML and 80–95 % of secondary AML patients demonstrate chromosomal aberrations. Structural chromosomal aberrations are the most common cytogenetic abnormalities in about of 40 % of de novo AML patients. A relatively large group of intermediate risk patients with cytogenetically normal (CN) AML demonstrates a variety of outcomes. Current AML prognostic classifications include only some mutations with known prognostic value, namely NPM1, FLT3 and C/EBPa. Patients with NPM1 mutation, but without FLT3-ITD or C/EBPa mutations have a favorable prognosis, whereas patients with FLT3-ITD mutation have a poor prognosis. A new class of mutations affecting genes responsible for epigenetic mechanisms of genome regulations, namely for DNA methylation and histone modification, was found recently. Among them, mutations in genes DNMT3A, IDH1/2, TET2 and some others are the most well-studied mutations to date. A number of studies demonstrated an unfavorable prognostic effect of the DNMT3A mutation in AML. The prognostic significance of the IDH1/2 gene is still unclear. The prognosis is affected by a number of biological factors, including those associated with cytogenetic aberrations and other mutations, especially FLT3 and NPM1. The number of studies of genetic mutations in AML keeps growing. The data on genetic aberrations in AML obtained to date confirm their role in the onset and development of the disease.


Keywords: acute myeloid leukemia, AML, karyotype, cytogenetic aberrations, gene mutation, prognosis.

Received: January 23, 2016

Accepted: April 4, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Renneville A, Roumier С., Biggio V, et al. Cooperating gene mutations in acute myeloid leukemia: a review of the literature. Leukemia. 2008;22(5):915–31. doi: 10.1038/leu.2008.19.
  2. Knudson AG. Mutation and Cancer: Statistical Study of Retinoblastoma. Proc Natl Acad Sci USA. 1971;68(4):820–3. doi: 10.1073/pnas.68.4.820.
  3. Tucker T, Friedman JM. Pathogenesis of hereditary tumors: beyond the “two-hit” hypothesis. Clin Genet. 2002;62(5):345–57. doi: 10.1034/j.1399-0004.2002.620501.x.
  4. Park S, Koh Y, Yoon SS. Effects of Somatic Mutations Are Associated with SNP in the Progression of Individual Acute Myeloid Leukemia Patient: The Two-Hit Theory Explains Inherited Predisposition to Pathogenesis. Genom Inform. 2013;11(1):34–7. doi: 10.5808/gi.2013.11.1.34.
  5. Genovese G, Kahler AK, Handsaker RE, et al. Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N Engl J Med. 2014;371(26):2477–87. doi: 10.1056/nejmoa1409405.
  6. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11. doi: 10.1038/35102167.
  7. Grimwade D. The changing paradigm of prognostic factors in acute myeloid leukaemia. Best Pract Res Clin Haematol. 2012;25(4):419–25. 10.1016/j.beha.2012.10.004.
  8. Patel JP, Gonen M, Figueroa ME, et al. Prognostic Relevance of Integrated Genetic Profiling in Acute Myeloid Leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/nejmoa1112304.
  9. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.
  10. Frehlick LJ, Eirin-Lopez JM, Ausio J. New insights into the nucleophosmin/nucleoplasmin family of nuclear chaperones. Bioessays. 2007;29(1):49–59. doi: 10.1002/bies.20512.
  11. Kurki S, Peltonen K, Latonen L, et al. Nucleolar protein NPM interacts with HDM2 and protects tumor suppressor protein p53 from HDM2-mediated degradation. Cell. 2004;5(5):465–75. doi: 10.1016/s1535-6108(04)00110-2.
  12. Lindstrom MS. NPM1/B23: A Multifunctional Chaperone in Ribosome Biogenesis and Chromatin Remodeling. Biochem Res Int. 2011;2011:1–16. doi: 10.1155/2011/195209.
  13. Falini B, Bolli NI, Martelli MP, et al. Translocations and mutations involving the nucleophosmin (NPM1) gene in lymphomas and leukemias. 2007;92(4):519–32. doi: 10.3324/haematol.11007.
  14. Falini B, Bigerna B, Pucciarini A, et al. Aberrant subcellular expression of nucleophosmin and NPM-MLF1 fusion protein in acute myeloid leukaemia carrying t(3;5): a comparison with NPMc+ AML. Leukemia. 2006;20(2):368–71. doi: 10.1038/sj.leu.2404068.
  15. Redner R, Rush EA, Faas S, et al. The t(5;17) variant of acute promyelocytic leukemia expresses a nucleophosmin-retinoic acid receptor fusion. 1996;87(3):882–6.
  16. Sportoletti P, Varasano E, Rossi R, et al. Mouse models of NPM1-mutated acute myeloid leukemia: biological and clinical implications. 2015;29(2):269–78. doi: 10.1038/leu.2014.257.
  17. Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nat Rev Cancer. 2006;6(7):493–505. doi: 10.1038/nrc1885.
  18. Sportoletti P, Grisendi S, Majid SM, et al. Npm1 is a haploinsufficient suppressor of myeloid and lymphoid malignancies in the mouse. Blood; 2008;111(7):3859–62. doi: 10.1182/blood-2007-06-098251.
  19. Ferrara F, Schiffer CA. Acute myeloid leukaemia in adults. The Lancet. 2013;381(9865):484–95. doi: 10.1016/s0140-6736(12)61727-9.
  20. Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic Nucleophosmin in Acute Myelogenous Leukemia with a Normal Karyotype. N Engl J Med. 2005;352(3):254–66. doi: 10.1056/nejmoa041974.
  21. Falini B, Martelli MP, Pileri SA, Mecucci C. Molecular and alternative methods for diagnosis of acute myeloid leukemia with mutated NPM1: flexibility may help. 2010;95(4):529–34. doi: 10.3324/haematol.2009.017822.
  22. Falini B, Albiero E, Bolli N, et al. Aberrant cytoplasmic expression of C-terminal-truncated NPM leukaemic mutant is dictated by tryptophans loss and a new NES motif. Leukemia. 2007;21(9):2052–4. doi: 10.1038/sj.leu.2404839.
  23. Schlenk RF, Dohner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–18. doi: 10.1056/nejmoa074306.
  24. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 Mutations Are Frequent Genetic Alterations in Acute Myeloid Leukemia and Confer Adverse Prognosis in Cytogenetically Normal Acute Myeloid Leukemia With NPM1 Mutation Without FLT3 Internal Tandem Duplication. J Clin Oncol. 2010. 28(22):3636–43. doi: 10.1200/jco.2010.28.3762.
  25. Dvorakova D, Racil Z, Jeziskova I, et al. Monitoring of minimal residual disease in acute myeloid leukemia with frequent and rare patient-specific NPM1 mutations. Am J Hematol. 2010;85(12):926–9. doi: 10.1002/ajh.21879.
  26. Schnittger S, Kern W, Tschulik C, et al. Minimal residual disease levels assessed by NPM1 mutation–specific RQ-PCR provide important prognostic information in AML. Blood. 2009;114(11):2220–31. doi: 10.1182/blood-2009-03-213389.
  27. Stahl T, Badbaran A, Kroger N, et al. Minimal residual disease diagnostics in patients with acute myeloid leukemia in the post-transplant period: comparison of peripheral blood and bone marrow analysis. Leuk Lymphoma. 2010;51(10):1837–43. doi: 10.3109/10428194.2010.508822.
  28. Kronke J, Schlenk RF, Jensen KO, et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol. 2011;29(19):2709–16. doi: 10.1200/jco.2011.35.0371.
  29. Rosnet O, Schiff C, Pebusque MJ, et al. Human FLT3/FLK2 gene: cDNA cloning and expression in hematopoietic cells. Blood. 1993;82(4):1110–9.
  30. Meshinchi S, Appelbaum FR. Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res. 2009;15(13):4263–9. doi: 10.1158/1078-0432.ccr-08-1123.
  31. Sitnicka E, Buza-Vidas N, Larsson S, et al. Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells. Blood. 2003;102(3):881–6. doi: 10.1182/blood-2002-06-1694.
  32. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002;100(5):1532–42. doi: 10.1182/blood-2002-02-0492.
  33. Adolfsson J, Borge OJ, Bryder D, et al. Upregulation of Flt3 Expression within the Bone Marrow Lin–Sca1+c-kit+ Stem Cell Compartment Is Accompanied by Loss of Self-Renewal Capacity. Immunity. 2001;15(4):659–69. doi: 10.1016/s1074-7613(01)00220-5.
  34. Griffith J, Black J, Faerman C, et al. The Structural Basis for Autoinhibition of FLT3 by the Juxtamembrane Domain. Mol Cell. 2004;13(2):169–78. doi: 10.1016/s1097-2765(03)00505-7.
  35. Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111(5):2776–84. doi: 10.1182/blood-2007-08-109090.
  36. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9. doi: 10.1182/blood.v98.6.1752.
  37. Marcucci G, Haferlach T, Dohner H. Molecular Genetics of Adult Acute Myeloid Leukemia: Prognostic and Therapeutic Implications. J Clin Oncol. 2011;29(5):475–86. doi: 10.1200/jco.2010.30.2554.
  38. Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. 2002;100(1):59–66. doi: 10.1182/blood.v100.1.59.
  39. Breitenbuecher F, Schnittger S, Grundler R, et al. Identification of a novel type of ITD mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor. Blood. 2009;113:4074–7. doi: 10.1182/blood-2007-11-125476.
  40. Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood. 2009;114(12):2386–92. doi: 10.1182/blood-2009-03-209999.
  41. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD–positive AML with respect to allogeneic transplantation. Blood. 2014;124(23):3441–9. doi: 10.1182/blood-2014-05-578070.
  42. Gu TL, Nardone J, Wang Y, et al. Survey of Activated FLT3 Signaling in Leukemia. PLoS One, 2011;6(4):e19169. doi: 10.1371/journal.pone.0019169.
  43. Rocnik JL, Okabe R, Yu JC, et al. Roles of tyrosine 589 and 591 in STAT5 activation and transformation mediated by FLT3-ITD. Blood. 2006;108(4):1339–45. doi: 10.1182/blood-2005-11-011429.
  44. Blau O, Berenstein R, Sindram A, Blau IW. Molecular analysis of different FLT3-ITD mutations in acute myeloid leukemia. Leuk Lymphoma. 2013;54(1):145–52. doi: 10.3109/10428194.2012.704999.
  45. Frohling S, Schlenk RF, Breitruck J, et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm. Blood. 2002;100(13):4372–80. doi: 10.1182/blood-2002-05-1440.
  46. Mrozek K, Marcucci G, Paschka P, et al. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification? Blood. 2007;109(2):431–48. doi: 10.1182/blood-2006-06-001149.
  47. Sengsayadeth SM, Jagasia M, Engelhardt BG, et al. Allo-SCT for high-risk AML-CR1 in the molecular era: impact of FLT3/ITD outweighs the conventional markers. Bone Marrow Transplant. 2012;47(12):1535–7. doi: 10.1038/bmt.2012.88.
  48. Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97(8):2434–9. doi: 10.1182/blood.v97.8.2434.
  49. Mead AJ, Linch DC, Hills RK, et al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood. 2007;110(4):1262–70. doi: 10.1182/blood-2006-04-015826.
  50. Whitman SP, Ruppert AS, Radmacher MD, et al. FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood. 2008;111(3):1552–9. doi: 10.1182/blood-2007-08-107946.
  51. Ozeki K, Kiyoi H, Hirose Y, et al. Biologic and clinical significance of the FLT3 transcript level in acute myeloid leukemia. Blood. 2004;103(5):1901–8. doi: 10.1182/blood-2003-06-1845.
  52. Ley TJ, Miller C, Ding L, et al. Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia. N Engl J Med. 2013;368(22):2059–74. doi: 10.1056/nejmoa1301689.
  53. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood. 2013;121(23):4769–77. doi: 10.1182/blood-2012-10-461624.
  54. Kottaridis PD, Gale RE, Langabeer SE, et al. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood. 2002;100(7):2393–8. doi: 10.1182/blood-2002-02-0420.
  55. Shih LY, Huang CF, Wu JH, et al. Internal tandem duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis of bone marrow samples from 108 adult patients at diagnosis and relapse. Blood. 2002;100(7):2387–92. doi: 10.1182/blood-2002-01-0195.
  56. Chu SH, Small D. Mechanisms of resistance to FLT3 inhibitors. Drug Resist Update. 2009;12(1–2):8–16. doi: 10.1016/j.drup.2008.12.001.
  57. Moore AS, Faisal A, Gonzalez de Castro D, et al. Selective FLT3 inhibition of FLT3-ITD+ acute myeloid leukaemia resulting in secondary D835Y mutation: a model for emerging clinical resistance patterns. Leukemia. 2012;26(7):1462–70. doi: 10.1038/leu.2012.52.
  58. Mead AJ, Gale RE, Kottaridis PD, et al. Acute myeloid leukaemia blast cells with a tyrosine kinase domain mutation of FLT3 are less sensitive to lestaurtinib than those with a FLT3 internal tandem duplication. Br J Haematol. 2008;141(4):454–60. doi: 10.1111/j.1365-2141.2008.07025.x.
  59. Koschmieder S, Halmos B, Levantini E, Tenen DG. Dysregulation of the C/EBPa Differentiation Pathway in Human Cancer. J Clin Oncol. 2009;27(4):619–28. doi: 10.1200/jco.2008.17.9812.
  60. Wang H, Iakova P, Wilde M, et al. C/EBPa Arrests Cell Proliferation through Direct Inhibition of Cdk2 and Cdk4. Mol Cell. 2001;8(4):817–28. doi: 10.1016/s1097-2765(01)00366-5.
  61. Radomska HS, Huettner CS, Zhang P, et al. CCAAT enhancer binding protein alpha is a regulatory switch sufficient for induction of granulocytic development from bipotential myeloid progenitors. Mol Cell Biol. 1998;18(7):4301–14. doi: 10.1128/mcb.18.7.4301.
  62. Zhang DE, Zhang P, Wang ND, et al. Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein a-deficient mice. Proc Natl Acad Sci USA. 1997;94(2):569–74. doi: 10.1073/pnas.94.2.569.
  63. Umek RM, Friedman AD, McKnight SL. CCAAT-enhancer binding protein: a component of a differentiation switch. Science. 1991;251(4991):288–92. doi: 10.1126/science.1987644.
  64. Watkins PJ, Condreay JP, Huber BE, et al. Proliferation and tumorigenicity induced by CCAAT/enhancer-binding protein. Cancer Res. 1996;56(5):1063–7.
  65. Pabst T, Mueller BU, Zhang P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-[alpha] (C/EBP [alpha]), in acute myeloid leukemia. Nat Genet. 2001;27(3):263–70. doi: 10.1038/85820.
  66. Nerlov C. C/EBP [alpha] mutations in acute myeloid leukaemias. Nat Rev Cancer. 2004;4(5):394–400. doi: 10.1038/nrc1363.
  67. Wouters BJ, Jorda MA, Keeshan K, et. al. Distinct gene expression profiles of acute myeloid/T-lymphoid leukemia with silenced CEBPA and mutations in NOTCH1. Blood. 2007;110(10):3706–14. doi: 10.1182/blood-2007-02-073486.
  68. Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood. 2011;117(8):2469–75. doi: 10.1182/blood-2010-09-307280.
  69. Kirstetter P, Schuster MB, Bereshchenko O, et al. Modeling of C/EBPa Mutant Acute Myeloid Leukemia Reveals a Common Expression Signature of Committed Myeloid Leukemia-Initiating Cells. Cancer Cell. 2008;13(4):299–310. doi: 10.1016/j.ccr.2008.02.008.
  70. Shih LY, Liang DC, Huang CF, et al. AML patients with CEBP [alpha] mutations mostly retain identical mutant patterns but frequently change in allelic distribution at relapse: a comparative analysis on paired diagnosis and relapse samples. Leukemia. 2006;20(4):604–9. doi: 10.1038/sj.leu.2404124.
  71. Wouters BJ, Lowenberg B, Erpelinck-Verschueren CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood. 2009;113(13):3088–91. doi: 10.1182/blood-2008-09-179895.
  72. Cagnetta A, Adamia S, Acharya C, et al. Role of genotype-based approach in the clinical management of adult acute myeloid leukemia with normal cytogenetics. Leuk Res. 2014;38(6):649–59. doi: 10.1016/j.leukres.2014.03.006.
  73. Wouters BJ, Sanders MA, Lugthart S, et al. Segmental uniparental disomy as a recurrent mechanism for homozygous CEBPA mutations in acute myeloid leukemia. Leukemia. 2007;21(11):2382–4. doi: 10.1038/sj.leu.2404795.
  74. Valk PJM, 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.
  75. Marceau-Renaut A, Guihard S, Castaigne S, et al. Classification of CEBPA mutated acute myeloid leukemia by GATA2 mutations. Am J Hematol. 2015;90(5):E93–4. doi: 10.1002/ajh.23949.
  76. Pabst T, Mueller BU. Transcriptional dysregulation during myeloid transformation in AML. Oncogene. 2007;26(47):6829–37. doi: 10.1038/sj.onc.1210765.
  77. Frohling S, Schlenk RF, Krauter J, et al. Acute myeloid leukemia with deletion 9q within a noncomplex karyotype is associated with CEBPA loss-of-function mutations. Genes Chromos Cancer. 2005;42(4):427–32. doi: 10.1002/gcc.20152.
  78. Green CL, Koo KK, Hills RK, et al. Prognostic Significance of CEBPA Mutations in a Large Cohort of Younger Adult Patients With Acute Myeloid Leukemia: Impact of Double CEBPA Mutations and the Interaction With FLT3 and NPM1 Mutations. J Clin Oncol. 2010;28(16):2739–47. doi: 10.1200/jco.2009.26.2501.
  79. Behdad A, Weigelin HC, Elenitoba-Johnson KS, Betz BL. A Clinical Grade Sequencing-Based Assay for CEBPA Mutation Testing: Report of a Large Series of Myeloid Neoplasms. J Mol Diagn. 2015;17(1):76–84. doi: 10.1016/j.jmoldx.2014.09.007.
  80. Bienz M, Ludwig M, Leibundgut EO, et al. Risk Assessment in Patients with Acute Myeloid Leukemia and a Normal Karyotype. Clin Cancer Res. 2005;11(4):1416–24. doi: 10.1158/1078-0432.ccr-04-1552.
  81. Frohling S, Schlenk RF, Stolze I, et al. CEBPA Mutations in Younger Adults With Acute Myeloid Leukemia and Normal Cytogenetics: Prognostic Relevance and Analysis of Cooperating Mutations. J Clin Oncol. 2004;22(4):624–33. doi: 10.1200/jco.2004.06.060.
  82. Preudhomme C, Sagot C, Boissel N, et al. Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association (ALFA). Blood. 2002;100(8):2717–23. doi: 10.1182/blood-2002-03-0990.
  83. Pastore F, Kling D, Hoster E, et al. Long-term follow-up of cytogenetically normal CEBPA-mutated AML. J Hematol Oncol. 2014;7(1):55. doi: 10.1186/s13045-014-0055-7.
  84. Park SH, Chi H-S, Cho Y-U, et al. CEBPA single mutation can be a possible favorable prognostic indicator in NPM1 and FLT3-ITD wild-type acute myeloid leukemia patients with intermediate cytogenetic risk. Leuk Res. 2013;37(11):1488–94. doi: 10.1016/j.leukres.2013.08.014.
  85. Renneville A, Boissel N, Gachard N, et al. The favorable impact of CEBPA mutations in patients with acute myeloid leukemia is only observed in the absence of associated cytogenetic abnormalities and FLT3 internal duplication. Blood. 2009;113(21):5090–3. doi: 10.1182/blood-2008-12-194704.
  86. Taniuchi I, Littman DR. Epigenetic gene silencing by Runx proteins. Oncogene. 2004;23(24):4341–5. doi: 10.1038/sj.onc.1207671.
  87. Yoshida H, Kitabayashi I. Chromatin regulation by AML1 complex. Int J Hematol. 2008;87(1):19–24. doi: 10.1007/s12185-007-0004-0.
  88. Tang JL, Hou HA, Chen CY, et al. AML1/RUNX1 mutations in 470 adult patients with de novo acute myeloid leukemia: prognostic implication and interaction with other gene alterations. 2009;114(26):5352–61. doi: 10.1182/blood-2009-05-223784.
  89. Dicker F, Haferlach C, Sundermann J, et al. Mutation analysis for RUNX1, MLL-PTD, FLT3-ITD, NPM1 and NRAS in 269 patients with MDS or secondary AML. Leukemia. 2010;24(8):1528–32. doi: 10.1038/leu.2010.124.
  90. Gaidzik VI, Bullinger L, Schlenk RF, et al. RUNX1 Mutations in Acute Myeloid Leukemia: Results From a Comprehensive Genetic and Clinical Analysis From the AML Study Group. J Clin Oncol. 2011;29(10):1364–72. doi: 10.1200/jco.2010.30.7926.
  91. Dicker F, Haferlach C, Kern W, et al. Trisomy 13 is strongly associated with AML1/RUNX1 mutations and increased FLT3 expression in acute myeloid leukemia. Blood. 2007;110:1308–16. doi: 10.1182/blood-2007-02-072595.
  92. Matsuno N, Osato M, Yamashita N, et al. Dual mutations in the AML1 and FLT3 genes are associated with leukemogenesis in acute myeloblastic leukemia of the M0 subtype. Leukemia. 2003;17(12):2492–9. doi: 10.1038/sj.leu.2403160.
  93. Mendler JH, Maharry K, Becker H, et al. In rare acute myeloid leukemia patients harboring both RUNX1 and NPM1 mutations, RUNX1 mutations are unusual in structure and present in the germline. 2013;98(8):e92–4. doi: 10.3324/haematol.2013.089904.
  94. Fasan A, Haferlach C, Kohlmann A, et al. Rare coincident NPM1 and RUNX1 mutations in intermediate risk acute myeloid leukemia display similar patterns to single mutated cases. Haematologica. 2014;99(2):e20–1. doi: 10.3324/haematol.2013.099754.
  95. Fernandez-Medarde A, Santos E. Ras in Cancer and Developmental Diseases. Genes Cancer. 2011;2(3):344–58. doi: 10.1177/1947601911411084.
  96. Stites EC, Ravichandran KS. A Systems Perspective of Ras Signaling in Cancer. Clin Cancer Res. 2009;15(5):1510–3. doi: 10.1158/1078-0432.ccr-08-2753.
  97. Johnson DB, Smalley KSM, Sosman JA. Molecular Pathways: Targeting NRAS in Melanoma and Acute Myelogenous Leukemia. Clin Cancer Res. 2014;20(16):4186–92. doi: 10.1158/1078-0432.ccr-13-3270.
  98. Fedorenko IV, Gibney GT, Smalley KSM. NRAS mutant melanoma: biological behavior and future strategies for therapeutic management. Oncogene. 2013;32(25):3009–18. doi: 10.1038/onc.2012.
  99. Reuter CM, Krauter J, Onono FO, et al. Lack of noncanonical RAS mutations in cytogenetically normal acute myeloid leukemia. Ann Hematol. 2014;93(6):977–82. doi: 10.1007/s00277-014-2061-9.
  100. Bacher U, Haferlach T, Schoch C, et al. Implications of NRAS mutations in AML: a study of 2502 patients. Blood. 2006;107(10):3847–53. doi: 10.1182/blood-2005-08-3522.
  101. Padua RA, West RR. Oncogene mutation and prognosis in the myelodysplastic syndromes. Br J Haematol. 2000;111(3):873–4. doi: 10.1111/j.1365-2141.2000.02472.x.
  102. Berman JN, Gerbing RB, Alonzo TA, et al. Prevalence and clinical implications of NRAS mutations in childhood AML: a report from the Children’s Oncology Group. 2011;25(6):1039–42. doi: 10.1038/leu.2011.31.
  103. Bowen DT, Frew ME, Hills R, et al. RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood. 2005;106(6):2113–9. doi: 10.1182/blood-2005-03-0867.
  104. Roskoski R Jr. Structure and regulation of Kit protein-tyrosine kinase–The stem cell factor receptor. Biochem Biophys Res Commun. 2005;338(3):1307–15. doi: 10.1016/j.bbrc.2005.09.150.
  105. Yarden Y, Ullrich A. Growth Factor Receptor Tyrosine Kinases. Ann Rev Biochem. 1988:57(1):443–78. doi: 10.1146/annurev.bi.57.070188.002303.
  106. Paschka P, Marcucci G, Ruppert AS, et al. Adverse Prognostic Significance of KIT Mutations in Adult Acute Myeloid Leukemia With inv(16) and t(8;21): A Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24(24):3904–11. doi: 10.1200/jco.2006.06.9500.
  107. Riera L, Marmont F, Toppino D, et al. Core binding factor acute myeloid leukaemia and c-KIT mutations. Oncol Rep. 2013;29(5):1867–72. doi: 10.3892/or.2013.2328.
  108. Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood. 2006;107(9):3463–8. doi: 10.1182/blood-2005-09-3640.
  109. Park SH, Chi HS, Min SK, et al. Prognostic impact of c-KIT mutations in core binding factor acute myeloid leukemia. Leuk Res. 2011;35(10):1376–83. doi: 10.1016/j.leukres.2011.06.003.
  110. Hoyos M, Nomdedeu JF, Esteve J, et al. Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease. Eur J Haematol. 2013;91(3):209–18. doi: 10.1111/ejh.12130.
  111. Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107(5):1791–9. doi: 10.1182/blood-2005-04-1466.
  112. Jiao B, Wu CF, Liang Y, et al. AML1-ETO9a is correlated with C-KIT overexpression/mutations and indicates poor disease outcome in t(8;21) acute myeloid leukemia-M2. Leukemia. 2009;23(9):1598–604. doi: 10.1038/leu.2009.104.
  113. Qin YZ, Zhu HH, Jiang Q, et al. Prevalence and prognostic significance of c-KIT mutations in core binding factor acute myeloid leukemia: A comprehensive large-scale study from a single Chinese center. Leuk Res. 2014;38(12):1435–40. doi: 10.1016/j.leukres.2014.09.017.
  114. O’Donnell MR, Tallman MS, Abboud CN, et al. Acute Myeloid Leukemia, Version 2.2013. J Natl Compr Canc Netw. 2013;11(9):1047–55.
  115. Tokumasu M, Murata C, Shimada A, et al. Adverse prognostic impact of KIT mutations in childhood CBF-AML: the results of the Japanese Pediatric Leukemia/Lymphoma Study Group AML-05 trial. Leukemia. 2015;29(12):2438–41. doi: 10.1038/leu.2015.121.
  116. Ito S, D’Alessio AC, Taranova OV, et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466(7310):1129–33. doi: 10.1038/nature09303.
  117. Chen Q, Chen Y, Bian C, et al. TET2 promotes histone O-GlcNAcylation during gene transcription. 2013;493(7433):561–4. doi: 10.1038/nature11742.
  118. Aslanyan M, Kroeze LI, Langemeijer SM, et al. Clinical and biological impact of TET2 mutations and expression in younger adult AML patients treated within the EORTC/GIMEMA AML-12 clinical trial. Ann Hematol. 2014;93(8):1401–12. doi: 10.1007/s00277-014-2055-7.
  119. Chou WC, Chou SC, Liu CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood. 2011;118(14):3803–10. doi: 10.1182/blood-2011-02-339747.
  120. Metzeler KH, Maharry K, Radmacher MD, et al. TET2 Mutations Improve the New European LeukemiaNet Risk Classification of Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol. 2011;29(10):1373–81. doi: 10.1200/jco.2010.32.7742.
  121. Gaidzik VI, Paschka P, Spath D, et al. TET2 Mutations in Acute Myeloid Leukemia (AML): Results From a Comprehensive Genetic and Clinical Analysis of the AML Study Group. J Clin Oncol. 2012;30(12):1350–7. doi: 10.1200/jco.2011.39.2886.
  122. Ko M, Huang Y, Jankowska AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. 2010;468(7325):839–43. doi: 10.1038/nature09586.
  123. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67. doi: 10.1016/j.ccr.2010.11.015.
  124. Moran-Crusio K, Reavie L, Shih A, et al. Tet2 Loss Leads to Increased Hematopoietic Stem Cell Self-Renewal and Myeloid Transformation. Cancer Cell. 2011;20(1):11–24. doi: 10.1016/j.ccr.2011.06.001.
  125. Quivoron C, Couronne L, Della Valle V, et al. TET2 Inactivation Results in Pleiotropic Hematopoietic Abnormalities in Mouse and Is a Recurrent Event during Human Lymphomagenesis. Cancer Cell. 2011;20(1):25–38. doi: 10.1016/j.ccr.2011.06.003.
  126. Nibourel O, Kosmider O, Cheok M, et al. Incidence and prognostic value of TET2 alterations in de novo acute myeloid leukemia achieving complete remission. Blood. 2010;116(7):1132–5. doi: 10.1182/blood-2009-07-234484.
  127. Weissmann S, Alpermann T, Grossmann V, et al. Landscape of TET2 mutations in acute myeloid leukemia. Leukemia. 2012;26(5):934–42. doi: 10.1038/leu.2011.326.
  128. Reitman ZJ, Yan H. Isocitrate Dehydrogenase 1 and 2 Mutations in Cancer: Alterations at a Crossroads of Cellular Metabolism. J Natl Cancer Inst. 2010;102(13):932–41. doi: 10.1093/jnci/djq187.
  129. Molenaar RJ, Radivoyevitch T, Maciejewski JP, et al. The driver and passenger effects of isocitrate dehydrogenase 1 and 2 mutations in oncogenesis and survival prolongation. Biochim Biophys Acta. 2014;1846(2):326–41. doi: 10.1016/j.bbcan.2014.05.004.
  130. Emadi A, Faramand R, Carter-Cooper B, et al. Presence of isocitrate dehydrogenase (IDH) mutations may predict clinical response to hypomethylating agents in patients with acute myeloid leukemia (AML). Am J Hematol. 2015;90(5):E77–9. doi: 10.1002/ajh.23965.
  131. Abbas S, Lugthart S, Kavelaars FG, et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010;116(12):2122–6. doi: 10.1182/blood-2009-11-250878.
  132. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 Gene Mutations Identify Novel Molecular Subsets Within De Novo Cytogenetically Normal Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/jco.2009.27.3730.
  133. Dang L, Jin S, Su SM. IDH mutations in glioma and acute myeloid leukemia. Trends Mol Med. 2010;16(9):387–97. doi: 10.1016/j.molmed.2010.07.002.
  134. Horbinski C. What do we know about IDH1/2 mutations so far, and how do we use it? Acta Neuropathol. 2013;125(5):621–36. doi: 10.1007/s00401-013-1106-9.
  135. Chotirat S, Thongnoppakhun W, Wanachiwanawin W, Auewarakul CU. Acquired somatic mutations of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) in preleukemic disorders. Blood Cells Mol Dis. 2015;54(3):286–91. doi: 10.1016/j.bcmd.2014.11.017.
  136. Green CL, Evans CM, Zhao L, et al. The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood. 2011;118(2):409–12. doi: 10.1182/blood-2010-12-322479.
  137. Zhou KG, Jiang LJ, Shang Z, et al. Potential application of IDH1 and IDH2 mutations as prognostic indicators in non-promyelocytic acute myeloid leukemia: a meta-analysis. Leuk Lymphoma. 2012;53(12):2423–9. doi: 10.3109/10428194.2012.695359.
  138. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2012;30(7):742–50. doi: 10.1200/jco.2011.39.2092.
  139. 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.
  140. Zhang Y, Chen FQ, Sun YH, et al. Effects of DNMT1 silencing on malignant phenotype and methylated gene expression in cervical cancer cells. J Exp Clin Cancer Res. 2011;30(1):98. doi: 10.1186/1756-9966-30-98.
  141. Jasielec J, Saloura V, Godley LA. The mechanistic role of DNA methylation in myeloid leukemogenesis. Leukemia. 2014;28(9):1765–73. doi: 10.1038/leu.2014.163.
  142. Li KK, Luo LF, Shen Y, et al. DNA methyltransferases in hematologic malignancies. Semin Hematol. 2013;50(1):48–60. doi: 10.1053/j.seminhematol.2013.01.005.
  143. O’Brien EC, Brewin J, Chevassut T. DNMT3A: the DioNysian MonsTer of acute myeloid leukaemia. Ther Adv Hematol. 2014;5(6):187–96. doi: 10.1177/2040620714554538.
  144. Holz-Schietinger C, Matje DM, Reich NO. Mutations in DNA methyltransferase (DNMT3A) observed in acute myeloid leukemia patients disrupt processive methylation. J Biol Chem. 2012;287(37):30941–51. doi: 10.1074/jbc.m112.366625.
  145. Russler-Germain DA, Spencer DH, Young MA, et al. The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell. 2014;25(4):442–54. doi: 10.1016/j.ccr.2014.02.010146.
  146. McDevitt MA. Clinical applications of epigenetic markers and epigenetic profiling in myeloid malignancies. Semin Oncol. 2012;39(1):109–22. doi: 10.1053/j.seminoncol.2011.11.003.
  147. Berenstein R, Blau IW, Suckert N, et al. Quantitative detection of DNMT3A R882H mutation in acute myeloid leukemia. J Exp Clin Cancer Res. 2015;34(1):55. doi: 10.1186/s13046-015-0173-2.
  148. Shlush LI, Zandi S, Mitchell A, et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014;506(7488):328–33. doi: 10.1038/nature13038.
  149. Corces-Zimmerman MR, Hong WJ, Weissman IL, et al. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci USA. 2014;111(7):2548–53. doi: 10.1073/pnas.1324297111.
  150. Thol F, Damm F, Ludeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol. 2011;29(21):2889–96. doi: 10.1200/jco.2011.35.4894.
  151. Ribeiro AF, Pratcorona M, Erpelinck-Verschueren C, et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood. 2012;119(24):5824–31. doi: 10.1182/blood-2011-07-367961.
  152. Ibrahem L, Mahfouz R, Elhelw L, et al. Prognostic significance of DNMT3A mutations in patients with acute myeloid leukemia. Blood Cells Mol Dis. 2014;54(1):84–9. doi: 10.1016/j.bcmd.2014.07.015.
  153. Shivarov V, Gueorguieva R, Stoimenov A, Tiu R. DNMT3A mutation is a poor prognosis biomarker in AML: results of a meta-analysis of 4500 AML patients. Leuk Res. 2013;37(11):1445–50. doi: 10.1016/j.leukres.2013.07.032.
  154. Wakita S, Yamaguchi H, Omori I, et al. Mutations of the epigenetics-modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia. 2013;27(5):1044–52. doi: 10.1038/leu.2012.317.
  155. Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood. 2012;119(2):559–68. doi: 10.1182/blood-2011-07-369934.
  156. Ploen GG, Nederby L, Guldberg P, et al. Persistence of DNMT3A mutations at long-term remission in adult patients with AML. Br J Haematol. 2014;167(4):478–86. doi: 10.1111/bjh.13062.
  157. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–98. doi: 10.1056/nejmoa1408617.