Актуальные вопросы таргетной терапии истинной полицитемии

А.Л. Меликян, И.Н. Суборцева

ФГБУ «НМИЦ гематологии» Минздрава России, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167

Для переписки: Анаит Левоновна Меликян, д-р мед. наук, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167; e-mail: anoblood@ mail.ru

Для цитирования: Меликян А.Л., Суборцева И.Н. Актуальные вопросы таргетной терапии истинной полицитемии. Клиническая онкогематология. 2021;14(3):355–60.

DOI: 10.21320/2500-2139-2021-14-3-355-360


РЕФЕРАТ

Вопросы использования критериев ответа на терапию, непереносимости гидроксикарбамида в первой линии, резистентности к нему и раннего изменения тактики лечения остаются спорными и не до конца решенными у пациентов с истинной полицитемией. В обзоре представлены результаты анализа данных литературы в отношении оценки эффективности первой линии терапии, проанализированы спектр и частота нежелательных явлений при применении гидроксикарбамида, опыт использования ингибитора JAK2 руксолитиниба. Приведены результаты, в т. ч. отдаленные, сравнительного анализа использования руксолитиниба и наилучшей доступной терапии у больных истинной полицитемией с резистентностью к гидроксикарбамиду. В настоящем обзоре использован материал работы экспертного совета с профессором Джузеппе А. Палумбо (университет Катании, Сицилия, Италия), который состоялся 7 июня 2020 г.

Ключевые слова: истинная полицитемия, JAK2V617F, прогноз, гидроксикарбамид, руксолитиниб.

Получено: 22 декабря 2020 г.

Принято в печать: 10 мая 2021 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Меликян А.Л., Туркина А.Г., Абдулкадыров К.М. и др. Клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз). Гематология и трансфузиология. 2014;59(4):31–56.
    [Melikyan AL, Turkina AG, Abdulkadyrov KM, et al. Clinical recommendations for the diagnosis and therapy of Ph-negative myeloproliferative diseases (polycythemia vera, essential thrombocythemia, primary myelofibrosis). Gematologiya i transfuziologiya. 2014;59(4):31–56. (In Russ)]
  2. Меликян А.Л., Туркина А.Г., Ковригина А.М. и др. Клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз) (редакция 2016 г.). Гематология и трансфузиология. 2017;62(1):25–60.
    [Melikyan AL, Turkina AG, Kovrigina AM, et al. Clinical recommendations for the diagnosis and therapy of Ph-negative myeloproliferative diseases (polycythemia vera, essential thrombocythemia, primary myelofibrosis) (edition 2016). Gematologiya i transfuziologiya. 2017;62(1):25–60. (In Russ)]
  3. Меликян А.Л., Ковригина А.М., Суборцева И.Н. и др. Национальные клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз) (редакция 2018 г.) Гематология и трансфузиология. 2018;63(3):275–315. doi: 10.25837/HAT.2019.51.88.001.
    [Melikyan AL, Kovrigina AM, Subortseva IN, et al. National clinical recommendations for diagnosis and therapy of Ph-negative myeloproliferative neoplasms (polycythemia vera, essential thrombocythemia, primary myelofibrosis) (edition 2018). Gematologiya i transfuziologiya. 2018;63(3):275–315. doi: 10.25837/HAT.2019.51.88.001. (In Russ)]
  4. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.
  5. Танашян М.М., Кузнецова П.И., Лагода О.В. и др. Миелопролиферативные заболевания и ишемический инсульт. Анналы клинической и экспериментальной неврологии. 2014;8(2):41–5.
    [Tanashyan MM, Kuznetsova PI, Lagoda OV, et al. Myeloproliferative diseases and ischemic stroke. Annaly klinicheskoi i eksperimental’noi nevrologii. 2014;8(2):41–5. (In Russ)]
  6. Танашян М.М., Кузнецова П.И., Суборцева И.Н. и др. Хроническая и острая цереброваскулярная патология при Ph-негативных миелопролиферативных заболеваниях. Гематология и трансфузиология. 2016;61(3):146–50. doi: 18821/0234-5730-2016-61-3-146-150.
    [Tanashyan MM, Kuznetsova PI, Subortseva IN, et al. Chronic and acute cerebrovascular pathology in patients with Ph-negative myeloproliferative diseases. Gematologiya i transfuziologiya. 2016;61(3):146–50. doi: 10.18821/0234-5730-2016-61-3-146-150. (In Russ)]
  7. Меликян А.Л., Суборцева И.Н., Суханова Г.А. Тромбогеморрагические осложнения у больных Ph-негативными миелопролиферативными заболеваниями. Кровь. 2014;2(18):21–5.
    [Melikyan AL, Subortseva IN, Sukhanova GA. Thrombohemorrhagic complications in patients with Ph-negative myeloproliferative diseases. Krov’. 2014;2(18):21–5. (In Russ)]
  8. Суборцева И.Н., Колошейнова Т.И., Пустовая Е.И. и др. Истинная полицитемия: обзор литературы и собственные данные. Клиническая онкогематология. 2015;8(4):397–412. doi: 10.21320/2500-2139-2015-8-4-397-412.
    [Subortseva IN, Kolosheinova TI, Pustovaya EI, et al. Polycythemia Vera: Literature Review and Own Data. Clinical oncohematology. 2015;8(4):397–412. doi: 10.21320/2500-2139-2015-8-4-397-412. (In Russ)]
  9. Falchi L, Newberry KJ, Verstovsek S. New Therapeutic Approaches in Polycythemia Vera. Clin Lymphoma Myel Leuk. 2015;15:27–33. doi: 10.1016/j.clml.2015.02.013.
  10. Barosi G, Mesa R, Finazzi G, et al. Revised response criteria for polycythemia vera and essential thrombocythemia: an ELN and IWG-MRT consensus project. Blood. 2013;121(23):4778–81. doi: 10.1182/blood-2013-01-478891.
  11. Меликян А.Л., Суборцева И.Н., Шуваев В.А. и др. Современный взгляд на диагностику и лечение классических Ph-негативных миелопролиферативных заболеваний. Клиническая онкогематология. 2021;14(1):129–37. doi: 10.21320/2500-2139-2021-14-1-129-137.
    [Melikyan AL, Subortseva IN, Shuvaev VA, et al. Current View on Diagnosis and Treatment of Classical Ph-Negative Myeloproliferative Neoplasms. Clinical oncohematology. 2021;14(1):129–37. doi: 10.21320/2500-2139-2021-14-1-129-137. (In Russ)]
  12. Barosi G, Birgegard G, Finazzi G, et al. Response criteria for essential thrombocythemia and polycythemia vera: result of a European LeukemiaNet consensus conference. Blood. 2009;113(20):4829–33. doi: 10.1182/blood-2008-09-176818.
  13. Palandri F, Elli ME, Benevolo G, et al. Clinical Outcomes Under Hydroxyurea and Impact of ELN Responses in Patients with Polycythemia Vera: A PV-NET Real World Study. Blood. 2019;134(1):4174. doi: 10.1182/blood-2019-125388.
  14. Barosi G, Birgegard G, Finazzi G, et al. A unified definition of clinical resistance and intolerance to hydroxycarbamide in polycythaemia vera and primary myelofibrosis: results of a European LeukemiaNet (ELN) consensus process. Br J Haematol. 2010;148(6):961–3. doi: 10.1111/j.1365-2141.2009.08019.x.
  15. Alvarez-Larran A, Kerguelen A, Hernandez-Boluda JC, et al.; Grupo Espanol de Enfermedades Mieloproliferativas Filadelfia Negativas (GEMFIN). Frequency and prognostic value of resistance/intolerance to hydroxycarbamide in 890 patients with polycythaemia vera. Br J Haematol. 2016;172(5):786–93. doi: 10.1111/bjh.13886.
  16. Kiladjian J-J, Pierre Z, Masayuki H, et al. Long-term efficacy and safety of ruxolitinib versus best available therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase 3 study. Lancet Haematol. 2020;7(3):е226–е doi: 10.1016/S2352-3026(19)30207-8.
  17. Alvarez-Larran A, Verstovsek S, Perez-Encinas M, et al. Comparison of ruxolitinib and real-world best available therapy in terms of overall survival and thrombosis in patients with polycythemia vera who are resistant or intolerant to hydroxyurea. EHA Library. 2018;215071:PF628.
  18. Curto-Garcia N, Baxter J, Harris E, et al. Molecular analysis in MAJIC PV correlation with clinical endpoints. HemaSphere. 2019;3(S1):740. doi: 10.1097/01.HS9.0000564676.68330.b5.

CAR Т-клетки для лечения хронического лимфоцитарного лейкоза: обзор литературы

И.В. Грибкова, А.А. Завьялов

ГБУ «НИИ организации здравоохранения и медицинского менеджмента ДЗМ», ул. Шарикоподшипниковская, д. 9, Москва, Российская Федерация, 115088

Для переписки: Ирина Владимировна Грибкова, канд. биол. наук, ул. Шарикоподшипниковская, д. 9, Москва, Российская Федерация, 115088; тел.: +7(916)078-73-90; e-mail: igribkova@yandex.ru

Для цитирования: Грибкова И.В., Завьялов А.А. CAR Т-клетки для лечения хронического лимфоцитарного лейкоза: обзор литературы. Клиническая онкогематология. 2021;14(2):225–30.

DOI: 10.21320/2500-2139-2021-14-2-225-230


РЕФЕРАТ

Хронический лимфоцитарный лейкоз (ХЛЛ) является наиболее распространенным злокачественным лимфоидным заболеванием взрослых. Несмотря на появление новых высокоэффективных таргетных препаратов, прогноз у больных с рецидивами и резистентной формой заболевания остается неблагоприятным. CAR Т-клеточная терапия, предполагающая использование Т-лимфоцитов с химерным антигенным рецептором (CAR), продемонстрировала свою эффективность в лечении ряда онкогематологических заболеваний, таких как В-клеточные неходжкинские лимфомы и острый лимфобластный лейкоз. В настоящем обзоре литературы рассматривается опыт применения CAR Т-клеток для лечения ХЛЛ. Представлены преимущества и недостатки данной технологии, а также проблемы, которые еще предстоит решить для внедрения метода в широкую клиническую практику.

Ключевые слова: хронический лимфоцитарный лейкоз, CAR T-клеточная терапия, химерный антигенный рецептор, адоптивная терапия, иммунотерапия.

Получено: 15 декабря 2020 г.

Принято в печать: 10 марта 2021 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Hallek M. Chronic lymphocytic leukemia: 2017 update on diagnosis, risk stratification, and treatment. Am J Hematol. 2017;92(9):946–65. doi: 10.1002/ajh.24826.
  2. Fernandez-Martinez JL, de Andres-Galiana EJ, Sonis ST. Genomic data integration in chronic lymphocytic leukemia. J Gene Med. 2017;19(1–2):e2936. doi: 10.1002/jgm.2936.
  3. Kipps TJ, Stevenson FK, Wu CJ, et al. Chronic lymphocytic leukaemia. Nat Rev Dis Primers. 2017;3(1):16096. doi: 10.1038/nrdp.2016.96.
  4. Byrd JC, Brown JR, O’Brien S, et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med. 2014;371(3):213–23. doi: 10.1056/NEJMoa1400376.
  5. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. N Engl J Med. 2016;374(4):311–22. doi: 10.1056/NEJMoa1513257.
  6. Bottcher S, Ritgen M, Fischer K, et al. Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: a multivariate analysis from the randomized GCLLSG CLL8 trial. J Clin Oncol. 2012;30(9):980–8. doi: 10.1200/JCO.2011.36.9348.
  7. Strati P, Keating MJ, O’Brien SM, et al. Outcomes of first-line treatment for chronic lymphocytic leukemia with 17p deletion. Haematologica. 2014;99(8):1350–5. doi: 10.3324/haematol.2014.104661.
  8. Mato AR, Nabhan C, Barr PM, et al. Outcomes of CLL patients treated with sequential kinase inhibitor therapy: a real world experience. Blood. 2016;128(18):2199–205. doi: 10.1182/blood-2016-05-716977.
  9. Anderson MA, Tam C, Lew TE, et al. Clinicopathological features and outcomes of progression of CLL on the BCL2 inhibitor venetoclax. Blood. 2017;129(25):3362–70. doi: 10.1182/blood-2017-01-763003.
  10. Dreger P, Schetelig J, Andersen N, et al. Managing high-risk CLL during transition to a new treatment era: Stem cell transplantation or novel agents? 2014;124(26):3841–9. doi: 10.1182/blood-2014-07-586826.
  11. June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. 2018;359(6382):1361–5. doi: 10.1126/science.aar6711.
  12. Грибкова И.В., Завьялов А.А. Терапия Т-лимфоцитами с химерным антигенным рецептором (CAR) В-клеточной неходжкинской лимфомы: возможности и проблемы. Вопросы онкологии. 2021. В печати.
    [Gribkova IV, Zav’yalov AA. Chimeric antigen receptor T‑cell therapy of B-cell non-Hodgkin’s lymphoma: opportunities and challenges. Voprosy onkologii. 2021. In print. (In Russ)]
  13. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33. doi: 10.1056/NEJMoa1103849.
  14. Forconi F, Moss P. Perturbation of the normal immune system in patients with CLL. Blood. 2015;126(5):573–81. doi: 10.1182/blood-2015-03-567388.
  15. Pourgheysari B, Bruton R, Parry H, et al. The number of cytomegalovirus-specific CD4+ T cells is markedly expanded in patients with B-cell chronic lymphocytic leukemia and determines the total CD4+ T-cell repertoire. 2010;116(16):2968–74. doi: 10.1182/blood-2009-12-257147.
  16. Palma M, Gentilcore G, Heimersson K, et al. T cells in chronic lymphocytic leukemia display dysregulated expression of immune checkpoints and activation markers. 2017;102(3):562–72. doi: 10.3324/haematol.2016.151100.
  17. Riches JC, Davies JK, McClanahan F, et al. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood. 2013;121(9):1612–21. doi: 10.1182/blood-2012-09-457531.
  18. Ramsay AG, Clear AJ, Fatah R, Gribben JG. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: Establishing a reversible immune evasion mechanism in human cancer. Blood. 2012;120(7):1412–21. doi: 10.1182/blood-2012-02-411678.
  19. D’Arena G, Laurenti L, Minervini MM, et al. Regulatory T-cell number is increased in chronic lymphocytic leukemia patients and correlates with progressive disease. Leuk Res. 2011;35(3):363–8. doi: 10.1016/j.leukres.2010.08.010.
  20. Gorgun G, Holderried TA, Zahrieh D, et al. Chronic lymphocytic leukemia cells induce changes in gene expression of CD4 and CD8 T cells. J Clin Invest. 2005;115(7):1797–805. doi: 10.1172/JCI24176.
  21. Piper KP, Karanth M, McLarnon A, et al. Chronic lymphocytic leukaemia cells drive the global CD4+ T cell repertoire towards a regulatory phenotype and leads to the accumulation of CD4+ forkhead box P3+ T cells. Clin Exp Immunol. 2011;166(2):154–63. doi: 10.1111/j.1365-2249.2011.04466.x.
  22. Brentjens RJ, Riviere I, Park JH, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118(18):4817–28. doi: 10.1182/blood-2011-04-348540.
  23. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  24. Kochenderfer JN, Dudley ME, Feldman SA, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. 2012;119(12):2709–20. doi: 10.1182/blood-2011-10-384388.
  25. Cruz CRY, Micklethwaite KP, Savoldo B, et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood. 2013;122(17):2965–73. doi: 10.1182/blood-2013-06-506741.
  26. Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33(6):540–9. doi: 10.1200/JCO.2014.56.2025.
  27. Porter DL, Hwang W-T, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi: 10.1126/scitranslmed.aac5415.
  28. Fraietta JA, Beckwith KA, Patel PR, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. 2016;127(9):1117–27. doi: 10.1182/blood-2015-11-679134.
  29. Brudno JN, Somerville RPT, Shi V, et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J Clin Oncol. 2016;34(10):1112–21. doi: 10.1200/JCO.2015.64.5929.
  30. Ramos CA, Savoldo B, Torrano V, et al. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains. J Clin Invest. 2016;126(7):2588–96. doi: 10.1172/JCI86000.
  31. Turtle CJ, Hay KA, Hanafi L-A, et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J Clin Oncol. 2017;35(26):3010–20. doi: 10.1200/JCO.2017.72.8519.
  32. Geyer MB, Riviere I, Senechal B, et al. Autologous CD19-targeted CAR T cells in patients with residual CLL following initial purine analog-based therapy. Mol Ther J Am Soc Gene Ther. 2018;26(8):1896–905. doi: 10.1016/j.ymthe.2018.05.018.
  33. Gauthier J, Hirayama AV, Hay KA, et al. Comparison of efficacy and toxicity of CD19-specific chimeric antigen receptor T-cells alone or in combination with ibrutinib for relapsed and/or refractory CLL. Blood. 2018;132(Suppl 1):299. doi: 1182/blood-2018-99-111061.
  34. Gill SI, Vides V, Frey NV, et al. Prospective clinical trial of anti-CD19 CAR T cells in combination with ibrutinib for the treatment of chronic lymphocytic leukemia shows a high response rate. Blood. 2018;132(Suppl 1):298. doi: 10.1182/blood-2018-99-115418.
  35. Siddiqi T, Soumerai JD, Wierda WG, et al. Rapid MRD-negative responses in patients with relapsed/refractory CLL treated with Liso-Cel, a CD19-directed CAR T-cell product: preliminary results from transcend CLL 004, a phase 1/2 study including patients with high-risk disease previously treated with ibrutinib. Blood. 2018;132(Suppl 1):300. doi: 10.1182/blood-2018-99-110462.
  36. Geyer MB, Riviere I, Senechal B, et al. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL. JCI Insight. 2019;4(9):e122627. doi: 10.1172/jci.insight.122627.
  37. Fraietta JA, Lacey SF, Orlando EJ, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018;24(5):563–71. doi: 10.1038/s41591-018-0010-1.
  38. Porter DL, Frey NV, Melenhorst JJ, et al. Randomized, phase II dose optimization study of chimeric antigen receptor modified T cells directed against CD19 (CTL019) in patients with relapsed, refractory CLL. Blood. 2014;124(21):1982. doi: 10.1182/blood.V124.21.1982.1982.
  39. Porter DL, Frey NV, Melenhorst JJ, et al. Randomized, phase II dose optimization study of chimeric antigen receptor (CAR) modified T cells directed against CD19 in patients (pts) with relapsed, refractory (R/R) CLL. J Clin Oncol. 2016;34(15_Suppl):3009. doi: 10.1200/JCO.2016.34.15_suppl.3009.
  40. Hofland T, Eldering E, Kater AP, Tonino SH. Engaging Cytotoxic T and NK Cells for Immunotherapy in Chronic Lymphocytic Leukemia. Int J Mol Sci. 2019;20(17):4315. doi: 10.3390/ijms20174315.
  41. Zou Y, Xu W, Li J. Chimeric antigen receptor-modified T cell therapy in chronic lymphocytic leukemia. J Hematol Oncol. 2018;11(1):130. doi: 10.1186/s13045-018-0676-3.
  42. Bair SM, Porter DL. Accelerating chimeric antigen receptor therapy in chronic lymphocytic leukemia: The development and challenges of chimeric antigen receptor T-cell therapy for chronic lymphocytic leukemia. Am J Hematol. 2019;94(Suppl 1):S10–S17. doi: 10.1002/ajh.25457.
  43. Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005;202(7):907–12. doi: 10.1084/jem.20050732.
  44. Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23(10):2346–57. doi: 10.1200/JCO.2005.00.240.
  45. Yin Q, Sivina M, Robins H, et al. Ibrutinib therapy increases T cell repertoire diversity in patients with chronic lymphocytic leukemia. J Immunol. 2017;198(4):1740–7. doi: 10.4049/jimmunol.1601190.
  46. Geyer MB, Park JH, Riviere I, et al. Implications of concurrent ibrutinib therapy on CAR T cell manufacturing and phenotype and on clinical outcomes following CD19-targeted CAR T cell administration in adults with relapsed/refractory CLL. Blood. 2016;128(22):58. doi: 10.1182/blood.V128.22.58.58.
  47. Golubovskaya V, Wu L. Different subsets of T cells, memory, effector functions, and CAR-T immunotherapy. Cancers (Basel). 2016;8(3):36. doi: 10.3390/cancers8030036.
  48. Hoffmann JM, Schubert ML, Wang L, et al. Differences in expansion potential of naive chimeric antigen receptor T cells from healthy donors and untreated chronic lymphocytic leukemia patients. Front Immunol. 2018;8: doi: 10.3389/fimmu.2017.01956.
  49. Sommermeyer D, Hudecek M, Kosasih PL, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016;30(2):492–500. doi: 10.1038/leu.2015.247.
  50. Hill JA, Li D, Hay KA, et al. Infectious complications of CD19-targeted chimeric antigen receptor-modified T-cell immunotherapy. Blood. 2018;131(1):121–30. doi: 10.1182/blood-2017-07-793760.
  51. Hay KA, Hanafi LA, Li D, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295–306. doi: 10.1182/blood-2017-06-793141.
  52. Gust J, Hay KA, Hanafi LA, et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;7(12):1404–19. doi: 10.1158/2159-8290.CD-17-0698.
  53. Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra25. doi: 10.1126/scitranslmed.3008226.
  54. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222.
  55. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy – assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62. doi: 10.1038/nrclinonc.2017.148.

COVID-19 у пациентов с онкогематологическими заболеваниями

А.А. Даниленко, С.В. Шахтарина, Н.А. Фалалеева

Медицинский радиологический научный центр им. А.Ф. Цыба — филиал ФГБУ «НМИЦ радиологии» Минздрава России, ул. Королева, д. 4, Обнинск, Калужская область, Российская Федерация, 249036

Для переписки: Анатолий Александрович Даниленко, д-р мед. наук, ул. Королева, д. 4, Обнинск, Калужская область, Российская Федерация, 249036; тел.: +7(909)250-18-10; e-mail: danilenkoanatol@mail.ru

Для цитирования: Даниленко А.А., Шахтарина С.В., Фалалеева Н.А. COVID-19 у пациентов с онкогематологическими заболеваниями. Клиническая онкогематология. 2021;14(2):220–4.

DOI: 10.21320/2500-2139-2021-14-2-220-224


РЕФЕРАТ

Эпидемия COVID-19, разразившаяся в Ухане (Китай), быстро переросла в пандемию. Высокая смертность от этого заболевания ставит его в ряд наиболее опасных вирусных инфекционных болезней настоящего времени. В то время как наибольшему риску летального исхода подвержены лица пожилого возраста, некоторые сопутствующие заболевания, среди которых вполне могут быть и злокачественные опухоли, также существенно ухудшают течение COVID-19. В силу присущего иммунодефицита, усугубляемого иммуносупрессивной противоопухолевой терапией, ведущее место по влиянию на течение COVID-19 занимают онкогематологические заболевания. В обзоре представлены немногочисленные опубликованные сведения о влиянии коронавирусной инфекции на прогноз у пациентов с опухолями кроветворной и лимфоидной тканей. Кроме того, обсуждаются возможные меры по снижению у этих больных риска летального исхода.

Ключевые слова: SARS-CoV-2, COVID-19, онкогематологические заболевания, смертность.

Получено: 13 октября 2020 г.

Принято в печать: 15 февраля 2021 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Johns Hopkins University of Medicine. COVID-19 Dashboard by the Center for Systems Science and Engineering (CSSE) at Johns Hopkins University (JHU). Available from: https://coronavirus.jhu.edu/map.html. Accessed 02.2021.
  2. Rassy E, Khoury-Abboud R-M, Ibrahim N, et al. What the oncologist needs to know about COVID-19 infection in cancer patients. Fut Oncol. 2020;16(17):1153–6. doi: 10.2217/fon-2020-0312.
  3. Mina A, van Besien K, Platanias LC, et al. Hematological manifestations of COVID-19. Leuk Lymphoma. 2020;61(12):2790–8. doi: 10.1080/10428194.2020.1788017.
  4. Bhatraju PK, Ghassemieh BJ, Nichols M, et al. Covid-19 in critically ill patients in the Seattle Region – case series. N Engl J Med. 2020;382(21):2012–22. doi: 10.1056/nejmoa2004500.
  5. Wang F, Nie J, Wang H, et al. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J Infect Dis. 2020;221(11):1762–69. doi: 10.1093/infdis/jiaa150.
  6. Guan W-J, Ni Z-Y, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708–20. doi: 10.1056/NEJMoa2002032.
  7. Maquet J, Lafaurie M, Sommet A, et al. Thrombocytopenia is independently associated with poor outcome in patients hospitalized for COVID-19. Br J Haematol. 2020;190(5):e276–е279. doi: 10.1111/bjh.16950.
  8. Lippi G, Plebani M, Henry BM. Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: a meta-analysis. Clin Chim Acta. 2020;506:145–8. doi: 10.1016/j.cca.2020.03.022.
  9. Cui S, Chen S, Li X, et al. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J Thromb Haemost. 2020;18(6):1421–4. doi: 10.1111/jth.14830.
  10. Klok FA, Kruip MJ, van der Meer NJ, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020;191:145–7. doi: 10.1016/j.thromres.2020.04.013.
  11. Friman V, Winqvist O, Blimark C, et al. Secondary immunodeficiency in lymphoproliferative malignancies. Hematol Oncol. 2016;34(3):121–32. doi: 10.1002/hon.2323.
  12. Yri OE, Torfoss D, Hungnes O, et al. Rituximab blocks protective serologic response to influenza a (H1N1) 2009 vaccination in lymphoma patients during or within 6 months after treatment. Blood. 2011;118(26):6769–71. doi: 10/1182/blood-2011-08-372649.
  13. Tepasse P-R, Hafezi W, Lutz M, et al. Persisting SARS-CoV-2 viraemia after rituximab therapy: two cases with fatal outcome and a review of the literature. Br J Haematol. 2020;190(2):185–8. doi: 10.1111/bjh.16896.
  14. Davids MS, Hallek M, Wierda W, et al. Comprehensive safety analysis of Venetoclax monotherapy for patients with relapsed/refractory chronic lymphocytic leukemia. Clin Cancer Res. 2018;24(18):4371–9. doi: 10.1158/1078-0432.CCR-17-3761.
  15. Van de Haar J, Hoes LR, Coles CE, et al. Caring for patients with cancer in the COVID-19 era. Nat Med. 2020;26(5):665–71. doi: 10.1038/s41591-020-0874-8.
  16. Lee LYW, Cazier J-B, Starkey T, et al. COVID-19 prevalence and mortality in patients with cancer and the effect of primary tumour subtype and patient demographics: a prospective cohort study. Lancet Oncol. 2020;21(10):1309–16. doi: 10.1016/S1470-2045(20)30442-3.
  17. Yang K, Sheng Y, Huang C, et al. Clinical characteristics, outcomes, and risk factors for mortality in patients with cancer and COVID-19 in Hubei, China: a multicentre, retrospective, cohort study. Lancet Oncol. 2020;21(7):904–13. doi: 10.1016/S1470-2045(20)30310-7.
  18. Booth S, Willan J, Wong H, et al. Regional outcomes of severe acute respiratory syndrome coronavirus 2 infection in hospitalised patients with haematological malignancy. Eur J Haematol. 2020;105(4):476–83. doi: 10.1111/ejh.13469.
  19. Wu Y, Chen W, Li W, et al. Clinical characteristics, therapeutic management, and prognostic factors of adult COVID-19 inpatients with hematological malignancies. Leuk Lymphoma. 2020;61(14):3440–50. doi: 10.1080/10428194.2020.1808204.
  20. Sanchez-Pina JM, Rodriguez RM, Castro Quismondo N, et al. Clinical course and risk factors for mortality from COVID-19 in patients with haematological malignancies. Eur J Haematol. 2020;105(5):597–607. doi: 10.1111/ejh.13493.
  21. Yigenoglu TN, Ata N, Altuntas F, et al. The Outcome of COVID-19 in Patients with Hematological Malignancy. J Med Virol. 2021;93(2):1099–104. doi: 10.1002/jmv.26404.
  22. He W, Chen L, Chen L, et al. COVID-19 in persons with haematological cancers. Leukemia. 2020;34(6):1637–45. doi: 10.1038/s41375-020-0836-7.
  23. Aries JA, Davies JK, Auer LR, et al. Clinical outcome of coronavirus disease 2019 in haemato-oncology patients. Br J Haematol. 2020;190(2):64–7. doi: 10.1111/bjh.16852.
  24. Passamonti F, Cattaneo C, Arcaini L, et al. Clinical characteristics and risk factors associated with COVID-19 severity in patients with haematological malignancies in Italy: a retrospective, multicentre, cohort study. Lancet Haematol. 2020;7(10):e737–e745. doi: 10.1016/S2352-3026(20)30251-9.
  25. Mato AR, Roeker LE, Lamanna N, et al. Outcomes of COVID-19 in patients with CLL: a multicenter international experience. Blood. 2020;136(10):1134–43. doi: 10.1182/blood.2020006965.
  26. Haroon A, Alassani M, Aljurf M, et al. COVID-19 post Hematopoietic Cell Transplant, a Report of 11 Cases from a Single Center. Mediterr J Hematol Infect Dis. 2020;12(1):e2020070. doi: 10.4084/MJHID.2020.070.
  27. Shah GL, De Wolf S, Lee YJ, et al. Favorable outcomes of COVID-19 in recipients of hematopoietic cell transplantation. J Clin Invest. 2020;130(12):6656–67. doi: 10.1172/jci141777.
  28. Perini GF, Fischer T, Gaiolla RD, et al. How to manage lymphoid malignancies during novel 2019 coronavirus (CoVid-19) outbreak: a Brazilian task force recommendation. Hematol Transfus Cell Ther. 2020;42(2):103–10. doi: 10.1016/j.htct.2020.04.002.
  29. Di Ciaccio P, McCaughan G, Trotman J, et al. Australian and New Zealand consensus statement on the management of lymphoma, chronic lymphocytic leukaemia and myeloma during the COVID-19 pandemic. Intern Med J. 2020;50(6):667–79. doi: 10.1111/imj.14859.
  30. De la Cruz-Benito B, Lazaro-Del Campo P, Ramirez-Lopez A, et al. Managing the front-line treatment for diffuse large B-cell lymphoma and high-grade B-cell lymphoma during the COVID-19 outbreak. Br J Haematol. 2020;191(3):386–9. doi: 10.1111/bjh.17066.
  31. Yahalom J, Dabaja BS, Ricardi U, et al. ILROG emergency guidelines for radiation therapy of hematological malignancies during the COVID-19 pandemic. Blood. 2020;135(21):1829–32. doi: 10.1182/blood.2020006028.
  32. Vordermark D. Shift in indications for radiotherapy during the COVID-19 pandemic? A review of organ-specific cancer management recommendations from multidisciplinary and surgical expert groups. Radiat Oncol. 2020;15(1):140. doi: 10.1186/s13014-020-01579-3.

Биотехнология CAR-T и новые возможности лечения опухолевых заболеваний

В.Ю. Павлова1, Е.С. Ливадный2

1 ГАУЗ «Кемеровская областная клиническая больница им. С.В. Беляева», Октябрьский пр-т, д. 22, корп. 2, Кемерово, Российская Федерация, 650066

2 ФГБОУ ВО «Кемеровский государственный медицинский университет», ул. Ворошилова, д. 22а, Кемерово, Российская Федерация, 650056

Для переписки: Вера Юрьевна Павлова, канд. мед. наук, Октябрьский пр-т, д. 22, корп. 2, Кемерово, Российская Федерация, 650066; тел.: +7(951)570-57-86; e-mail: vera.4447.kem@mail.ru

Для цитирования: Павлова В.Ю., Ливадный Е.С. Биотехнология CAR-T и новые возможности лечения опухолевых заболеваний. Клиническая онкогематология. 2021;14(1):149–56.

DOI: 10.21320/2500-2139-2021-14-1-149-156


РЕФЕРАТ

Злокачественные новообразования как причина смерти занимают 2-е место в мире после сердечно-сосудистых заболеваний. CAR T-терапия (chimeric antigen receptor of T-cells) — прогрессивный метод лечения злокачественных опухолей. Использование CAR Т-лимфоцитов относится к адоптивной иммунотерапии. Технология CAR-T основана на «извлечении» клеток иммунной системы (Т-лимфоцитов) из организма, их генетической модификации в целях приобретения ими противоопухолевых свойств с последующей реинфузией пациенту. Преимущество CAR T-терапии в сравнении с другими методами лечения заключается в том, что для распознавания клеток-мишеней Т-лимфоциты не нуждаются в присутствии молекул главного комплекса гистосовместимости 1-го класса (MHC-I). Собранные и проанализированные нами литературные данные свидетельствуют о появлении принципиально нового эффективного метода лечения онкогематологических заболеваний, включая острый лимфобластный лейкоз, хронический лимфолейкоз и неходжкинские лимфомы. На основании клинических исследований доказано преимущество CAR T-терапии в сравнении с другими методами лечения, применяющимися в данной области. Анализ литературы позволил сделать вывод об обоснованности рассмотрения CAR T-терапии как одной из перспективных возможностей воздействия на злокачественную опухоль.

Ключевые слова: адоптивная иммунотерапия, CAR T-лимфоциты, химерный антигенный рецептор.

Получено: 20 сентября 2020 г.

Принято в печать: 1 декабря 2020 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–Е386. doi: 10.1002/ijc.29210.
  2. Stewart BW, Wilde CP (eds). World cancer report 2014. Lyon: IARC Press, 2014. 619 p.
  3. Plummer M, de Martel C, Vignat J, et al. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob 2016;4(9):e609–e616. doi: 10.1016/S2214-109X(16)30143-7.
  4. Состояние онкологической помощи населению России в 2018 году. Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Петровой. М.: МНИОИ им. П.А. Герцена — филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2019. 236 с.
    [Caprin AD, Starinskii VV, Petrova GV, eds. Sostoyanie onkologicheskoi pomoshchi naseleniyu Rossii v 2018 godu. (The state of cancer care in Russia in 2018.) Moscow: MNIOI im. P.A. Gertsena — a branch of FGBU “NMITs radiologii” Minzdrava Rossii Publ.; 2019. 236 p. (In Russ)]
  5. Здравоохранение в России. Статистический сборник. М.: Росстат, 2011. 326 с.
    [Zdravookhranenie v Rossii. Statisticheskii sbornik. (Health care in Russia. Statistics digest.) Мoscow: Rosstat Publ.; 2011. 326 p. (In Russ)]
  6. Halaleh K, Gale RP. Cancer care in the Palestinian territoried. Lancet Oncol. 2018;19(7):e359–е364. doi: 10.1016/S1470-2045(18)30323-1.
  7. Чикилева И.О., Шубина И.Ж., Киселевский М.В. Влияние регуляторных Т-клеток на функциональную активность натуральных киллеров при иммунотерапии злокачественных опухолей. Вестник РАМН. 2012;67(4):60–4.
    [Chikileva IO, Shubina IZh, Kiselevskii MV. Influence of regulatory T-cells on the functioning of natural killer cells during cancer immunotherapy. Vestnik RAMN. 2012;67(4):60–4. (In Russ)]
  8. Титов К.С., Демидов Л.В., Шубина И.Ж. и др. Технологии клеточной иммунотерапии в лечении больных со злокачественными новообразованиями. Вестник Российского государственного медицинского университета. 2014;1:42–7.
    [Titov KS, Demidov LV, Shubina IZh, et al. Technologies of cell immunotherapy in treatment of cancer patients. Vestnik Rossiiskogo gosudarstvennogo meditsinskogo universiteta; 2014;1:42–7. (In Russ)]
  9. Wahlang B, Falkner KC, Cave MC, et al. Role of Cytochrome P450 Monooxygenase in Carcinogen and Chemotherapeutic Drug Metabolism. Adv Pharmacol. 2015;74:1–33. doi: 10.1016/bs.apha.2015.04.004.
  10. Janeway CA Jr, Travers P, Walport M, et al. Immunobiology: The Immune System in Health and Disease. 5th edition. New York: Garland Science, 2001. 884 p.
  11. Croce CM. Oncogenes and cancer. N Engl J Med. 2008;358(5):502–11. doi: 10.1056/NEJMra072367.
  12. Vicente-Duenas C, Romero-Camarero I, Cobaleda C, et al. Function of oncogenes in cancer development: a changing paradigm. EMBO J. 2013;32(11):1502–13. doi: 10.1038/emboj.2013.97.
  13. Den Haan JM, Arens R, Van Zelm MC. The activation of the adaptive immune system: cross-talk between antigen-presenting cells, T cells and B cells. Immunol Lett. 2014;162(2 Pt B):103–12. doi: 10.1016/j.imlet.2014.10.011.
  14. Emtage PC, Lo AS, Gomes EM, et al. Second-generation anti-carcinoembryonic antigen designer T cells resist activation-induced cell death, proliferate on tumor contact, secrete cytokines, and exhibit superior antitumor activity in vivo: a preclinical evaluation. Clin Cancer Res. 2008;14(24):8112–22. doi: 10.1158/1078-0432.
  15. Maher Immunotherapy of Malignant Disease Using Chimeric Antigen Receptor Engrafted T Cells. ISRN Oncol. 2012;2012:278093. doi: 10.5402/2012/278093.
  16. BonifantL, Jackson HJ, Brentjens RJ, et al. Toxicity and management in CAR T-cell therapy. Mol Ther. 2016;3:16011. doi: 10.1038/mto.2016.11.
  17. Xu J, Wang Q, Xu H, et al. Anti-BCMA CAR-T cells for treatment of plasma cell dyscrasia: case report on POEMS syndrome and multiple myeloma. J Hematol Oncol. 2018;11(1):128. doi: 10.1186/s13045-018-0672-7.
  18. Wang J, Chen S, Xiao W, et al. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol. 2018;11(1):7. doi: 10.1186/s13045-017-0553-5.
  19. Wei J, Han X, Bo J, et al. Target selection for CAR-T therapy. J Hematol Oncol. 2019;12(1):62. doi: 10.1186/s13045-019-0758-x.
  20. Si W, Li C, Wei P. Synthetic immunology: T-cell engineering and adoptive immunotherapy. Synth Syst Biotechnol. 2018;3(3):179–85. doi: 10.1016/j.synbio.2018.08.001.
  21. Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. J Cell Immunother. 2016;2(2):59–68. doi: 10.1016/j.jocit.2016.08.001.
  22. Chmielewski M, Abken H. Trucks: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):1145–54. doi: 10.1517/14712598.2015.1046430.
  23. Zhao Z, Chen Y, Francisco NM, et al. The application of CAR-T cell therapy in hematological malignancies: advantages and challenges. Acta Pharm Sin B. 2018;8(4):539–51. doi: 10.1016/j.apsb.2018.03.001.
  24. Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev. 2019;34:45–55. doi: 10.1016/j.blre.2018.11.002.
  25. Brudno JN, Maric I, Hartman SD, et al. T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor prognosis relapsed multiple myeloma. J Clin Oncol. 2018;36(22):2267–80. doi: 10.1200/JCO.2018.77.8084.
  26. Maude SL, Frey N, Shaw PA, et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222.
  27. Fernandez A Cure for Cancer? How CAR T-Cell Therapy is Revolutionizing Oncology. Available from: https://www.labiotech.eu/features/car-t-therapy-cancer-review/ (accessed 25.11.2020).
  28. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi: 10.1126/scitranslmed.aac5415.
  29. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377(26):2545–54. doi: 10.1056/NEJMoa1708566.
  30. Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14(11):1264–70. doi: 10.1038/nm.1882.
  31. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  32. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33. doi: 10.1056/NEJMoa1103849.
  33. Riches JC, Gribben JG. Understanding the immunodeficiency in chronic lymphocytic leukemia: potential clinical implications. Hematol Oncol Clin North Am. 2013;27(2):207–35. doi: 10.1016/j.hoc.2013.01.003.
  34. Ellard R, Stewart O. The EBMT Guidelines for practice. A framework for managing Patient Care, CRS and Neurotoxicity. 1st European CAR T cell meeting, 14–16 February 2019, Paris, France.

Клеточный препарат с химерным антигенным рецептором NKG2D в CAR T-терапии рецидивов/рефрактерных острых миелоидных лейкозов и миелодиспластического синдрома

К.A. Левчук1, Е.В. Белоцерковская1,2, Д.Ю. Поздняков1, Л.Л. Гиршова1, А.Ю. Зарицкий1, А.В. Петухов1,2,3

1 ФГБУ «НМИЦ им. В.А. Алмазова» Минздрава России, ул. Аккуратова, д. 2, Санкт-Петербург, Российская Федерация, 197341

2 ФГБУН «Институт цитологии РАН», Тихорецкий пр-т, д. 4, Санкт-Петербург, Российская Федерация, 194064

3 НТУ «Сириус», Олимпийский пр-т, д. 1, Сочи, Российская Федерация, 354340

Для переписки: Ксения Александровна Левчук, ул. Аккуратова, д. 2, Санкт-Петербург, Российская Федерация, 197341; e-mail: levchuk_ka@almazovcentre.ru

Для цитирования: Левчук К.A., Белоцерковская Е.В., Поздняков Д.Ю. и др. Клеточный препарат с химерным антигенным рецептором NKG2D в CAR T-терапии рецидивов/рефрактерных острых миелоидных лейкозов и миелодиспластического синдрома. Клиническая онкогематология. 2021;14(1):138–48.

DOI: 10.21320/2500-2139-2021-14-1-138-148 


РЕФЕРАТ

NK-клетки как элементы врожденного иммунитета реализуют ключевые реакции противоопухолевого иммунного ответа. NKG2D — активационный трансмембранный рецептор NK-клеток, ответственный за инициацию цитотоксичности в ответ на связывание специфичных лигандов генетически модифицированных клеток. Селективная экспрессия лигандов NKG2D открывает уникальные перспективы для терапии широкого спектра опухолей. Острые миелоидные лейкозы (ОМЛ) — это злокачественные опухоли системы крови, характеризующиеся высоким риском развития рецидивов. Сложность терапевтической стратегии при ОМЛ создает необходимость поиска новых подходов к элиминации опухоли с применением инновационных генетических конструкций. Имеющиеся к настоящему времени CAR T-клеточные препараты, несущие рецептор NKG2D, успешно изучаются в клинических исследованиях у пациентов с ОМЛ, доказывая свой высокий терапевтический потенциал.

Ключевые слова: острые миелоидные лейкозы, химерный антигенный рецептор, адоптивная терапия, NKG2D, NK-клетки.

Получено: 22 августа 2020 г.

Принято в печать: 5 декабря 2020 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Arber D, Orazi A, Hasserjian R. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.
  2. Bullinger L, Dohner K, Dohner H. Genomics of Acute Myeloid Leukemia Diagnosis and Pathways. J Clin Oncol. 2017;35(9):934–46. doi: 10.1200/JCO.2016.71.2208.
  3. The Leukemia & Lymphoma Society Updated data on blood cancers. Facts 2018–2019. Available from: https://www.lls.org/facts-and-statistics/facts-and-statistics-overview/facts-and-statistics (accessed 30.11.2020).
  4. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. 2017;129(4):424–47. doi: 10.1182/blood-2016-08-733196.
  5. Herold T, Rothenberg-Thurley M, Grunwald VV, et al. Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia Leukemia. [published online ahead of print, 2020 Mar 30] doi: 10.1038/s41375-020-0806-0.
  6. Estey EH, Schrier SL. Prognosis of the myelodysplastic syndromes in adults. UpToDate. 2017. Available from: https://www.uptodate.com/contents/prognosis-of-the-myelodysplastic-syndromes-in-adults (accessed 28.11.2020).
  7. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. 2005;106(4):1154–63. doi: 10.1182/blood-2005-01-0178.
  8. Burnett AK, Milligan D, Goldstone A, et al. The impact of dose escalation and resistance modulation in older patients with acute myeloid leukemia and high risk myelodysplastic syndrome: the results of the LRF AML14 trial. Br J Haematol. 2009;145(3):318–32. doi: 10.1111/j.1365-2141.2009.07604.x.
  9. Lowenberg G. Strategies in the treatment of acute myeloid leukemia. Haematologica. 2004;89(9):1029–32.
  10. Burnett AK. Acute myeloid leukemia: Treatment of adults under 60 years. Rev Clin Exp Hematol. 2002;6(1):26–45. doi: 10.1046/j.1468-0734.2002.00058.x.
  11. Estey EH. Treatment of relapsed and refractory acute myelogenous leukemia. 2000;14(3):476–9. doi: 10.1038/sj.leu.2401568.
  12. Giles F, O’Brien S, Cortes J, et al. Outcome of patients with acute myelogenous leukemia after second salvage therapy. 2005;104(3):547–54. doi: 10.1002/cncr.21187.
  13. Leopold LH, Willemze R. The treatment of acute myeloid leukemia in first relapse: A comprehensive review of the literature. Leuk Lymphoma. 2002;43(9):1715–27. doi: 10.1080/1042819021000006529.
  14. Lee S, Tallman MS, Oken MM, et al. Duration of second complete remission compared with first complete remission in patients with acute myeloid leukemia. 2000;14(8):1345–8. doi: 10.1038/sj.leu.2401853.
  15. Patel SA, Gerber JM. A User’s Guide to Novel Therapies for Acute Myeloid Leukemia. Clin Lymphoma Myel Leuk. 2020;20(5):277–88. doi: 10.1016/j.clml.2020.01.011.
  16. Kucukyurt S, Eskazan AE. New drugs approved for acute myeloid leukemia in 2018. Br J Clin Pharmacol. 2018;85(12):2689–93. doi: 10.1111/bcp.14105.
  17. Spear P, Wu MR, Sentman ML, Sentman CL. NKG2D ligands as therapeutic targets. Cancer Immun. 2013;13:8.
  18. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. 2012;120(12):2454–65. doi: 10.1016/s0145-2126(13)70009-2.
  19. Blum WG. Hypomethylating agents in myelodysplastic syndromes. Clin Adv Hematol Oncol. 2011;9(2):123–8.
  20. Семочкин С.В., Толстых Т.Н., Иванова В.Л. и др. Азацитидин в лечении миелодиспластических синдромов: клиническое наблюдение и обзор литературы. Клиническая онкогематология. 2012;5(3):233–8.
    [Semochkin SV, Tolstykh TN, Ivanova VL, et al. Azacitidine in the treatment of myelodysplastic syndromes: case report and literature review. Klinicheskaya onkogematologiya. 2012;5(3):233–8. (In Russ)]
  21. 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)]
  22. Richard-Carpentier G, DeZern AE, Takahashi K, et al. Preliminary Results from the Phase II Study of the IDH2-Inhibitor Enasidenib in Patients with High-Risk IDH2-Mutated Myelodysplastic Syndromes (MDS). 2019;134(1):678. doi: 10.1182/blood-2019-130501.
  23. Foran JM, DiNardo CD, Watts JM, et al. Ivosidenib (AG-120) in Patients with IDH1-Mutant Relapsed/Refractory Myelodysplastic Syndrome: Updated Enrollment of a Phase 1 Dose Escalation and Expansion Study. 2019;134(1):4254. doi: 10.1182/blood-2019-123946.
  24. Garcia JS. Prospects for Venetoclax in Myelodysplastic Syndromes. Hematol Oncol Clin N Am. 2020;34(2):441–8. doi: 10.1016/j.hoc.2019.10.005.
  25. Germing U, Schroeder T, Kaivers J, et al. Novel therapies in low- and high-risk myelodysplastic syndrome. Exp Rev Hematol. 2019;12(10):893–908. doi: 10.1080/17474086.2019.1647778.
  26. Platzbecker U. Treatment of MDS. 2019;133(10):1096–107. doi: 10.1182/blood-2018-10-844696.
  27. Swoboda DM, Sallman DA. Mutation-Driven Therapy in MDS. Curr Hematol Malig Rep. 2019;14(6):550–60. doi: 10.1007/s11899-019-00554-4.
  28. Миелодиспластические синдромы. Интервью с С.В. Грицаевым. Клиническая онкогематология. 2018;11(2):125–37.
    [Myelodysplastic syndromes. Interview with SV Gritsaev. Clinical oncohematology. 2018;11(2):125–37. (In Russ)]
  29. Manley PW, Weisberg E, Sattler M, et al. Midostaurin, a Natural Product-Derived Kinase Inhibitor Recently Approved for the Treatment of Hematological Malignancies. 2018;57(5):477–8. doi: 10.1021/acs.biochem.7b01126.
  30. Liu X, Gong Y. Isocitrate dehydrogenase inhibitors in acute myeloid leukemia. Biomark Res. 2019;7(1):22. doi: 10.1186/s40364-019-0173-z.
  31. Kim ES. Enasidenib: First Global Approval. 2017;77(15):1705–11. doi: 10.1007/s40265-017-0813-2.
  32. Garcia-Aranda M, Perez-Ruiz E, Redondo M. Bcl-2 Inhibition to Overcome Resistance to Chemo- and Immunotherapy. Int J Mol Sci. 2018;19(12):3950. doi: 10.3390/ijms19123950.
  33. Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for patients with relapse after allogeneic transplantation. Leukemia and Lymphoma Society Blood Cancer Research Partnership. N Engl J Med. 2016;375(2):143–53. doi: 10.1056/NEJMoa1601202.
  34. Li F, Sutherland MK, Yu C, et al. Characterization of SGN-CD123A, A Potent CD123-Directed Antibody-Drug Conjugate for Acute Myeloid Leukemia. Mol Cancer Ther. 2018;17(2):554–64. doi: 10.1158/1535-7163.MCT-17-0742.
  35. Mawad R, Gooley TA, Rajendran JG, et al. Radiolabeled AntiCD45 Antibody with Reduced-Intensity Conditioning and Allogeneic Transplantation for Younger Patients with Advanced Acute Myeloid Leukemia or Myelodysplastic Syndrome. Biol Blood Marrow Transplant. 2014;20(9):1363–8. doi: 10.1016/j.bbmt.2014.05.014.
  36. Guy DG, Uy GL. Bispecific Antibodies for the Treatment of Acute Myeloid Leukemia. Curr Hematol Malig Rep. 2018;13(6):417– doi: 10.1007/s11899-018-0472-8.
  37. Di Stasi A, Jimenez AM, Minagawa K, et al. Review of the Results of WT1 Peptide Vaccination Strategies for Myelodysplastic Syndromes and Acute Myeloid Leukemia from Nine Different Studies. Front Immunol. 2015;6:36. doi: 10.3389/fimmu.2015.00036.
  38. Van Acker HH, Versteven M, Lichtenegger FS, et al. Dendritic Cell-Based Immunotherapy of Acute Myeloid Leukemia. J Clin Med. 2019;8(5):579. doi: 10.3390/jcm8050579.
  39. Yabe T, McSherry C, Bach FH, et al. A multigene family on human chromosome 12 encodes natural killer-cell lectins. 1993;37(6):455–60. doi: 10.1007/bf00222470.
  40. Houchins JP, Yabe T, McSherry C, Bach FH. DNA sequence analysis of NKG2, a family of related cDNA clones encoding type II integral membrane proteins on human natural killer cells. J Exp Med. 1991;173(4):1017–20. doi: 10.1084/jem.173.4.1017.
  41. Upshaw JL, Arneson LN, Schoon RA, et al. NKG2D-mediated signaling requires a DAP10-bound Grb2-Vav1 intermediate and phosphatidylinositol-3-kinase in human natural killer cells. Nat Immunol. 2006;7(5):524–32. doi: 10.1038/ni1325.
  42. Diefenbach A, Tomasello E, Lucas M, et al. Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat Immunol. 2002;3(12):1142–9. doi: 10.1038/ni858.
  43. Duan S, Guo W, Xu Z, et al. Natural killer group 2D receptor and its ligands in cancer immune escape. Mol Cancer. 2019;18(1):29. doi: 10.1186/s12943-019-0956-8.
  44. Wu J, Song Y, Bakker AB, et al. An activating immunoreceptor complex formed by NKG2D and DAP10. 1999;285(5428):730–2. doi: 10.1126/science.285.5428.730.
  45. Ogasawara K, Lanier LL. NKG2D in NK and T cell-mediated immunity. J Clin Immunol. 2005;25(6):534–40. doi: 10.1007/s10875-005-8786-4.
  46. Gilfillan S, Ho EL, Cella M, et al. NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol. 2002;3(12):1150–5. doi: 10.1038/ni857.
  47. Groh V, Rhinehart R, Randolph-Habecker J, et al. Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol. 2001;2(3):255–60. doi: 10.1038/85321.
  48. Jamieson AM, Diefenbach A, McMahon CW, et al. The role of the NKG2D immunoreceptor in immune cell activation and natural killing. 2002;17(1):19–29. doi: 10.1016/s1074-7613(02)00333-3.
  49. Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol. 2003;3(10):781–90. doi: 10.1038/nri1199.
  50. Roberts AI, Lee L, Schwarz E, et al. NKG2D receptors induced by IL-15 costimulate CD28-negative effector CTL in the tissue microenvironment. J Immunol. 2001;167(10):5527–30. doi: 10.4049/jimmunol.167.10.5527.
  51. Lanier LL. NK cell recognition. Annu Rev Immunol. 2005;23(1):225–74. doi: 10.1146/annurev.immunol.23.021704.115526.
  52. Raulet DH, Gasser S, Gowen BG, et al. Regulation of ligands for the NKG2D activating receptor. Annu Rev Immunol. 2013;31(1):413–41. doi: 10.1146/annurev-immunol-032712-095951.
  53. Stephens HA. MICA and MICB genes: can the enigma of their polymorphism be resolved?. Trends Immunol. 2001;22(7):378–85. doi: 10.1016/s1471-4906(01)01960-3.
  54. Carapito R, Bahram S. Genetics, genomics, and evolutionary biology of NKG2D ligands. Immunol Rev. 2015;267(1):88–116. doi: 10.1111/imr.12328.
  55. Bartkova J, Horejsi Z, Koed K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. 2005;434(7035):864–70. doi: 10.1038/nature03482.
  56. Gorgoulis VG, Vassiliou LV, Karakaidos P, et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. 2005;434(7035):907–13. doi: 10.1038/nature03485.
  57. Maeda T, Towatari M, Kosugi H, Saito H. Up-regulation of costimulatory/adhesion molecules by histone deacetylase inhibitors in acute myeloid leukemia cells. Blood. 2000;96(12):3847–56. doi: 1182/blood.v96.12.3847.
  58. Diermayr S, Himmelreich H, Durovic B, et al. NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. 2008;111(3):1428–36. doi: 10.1182/blood-2007-07-101311.
  59. Chang YH, Connolly J, Shimasaki N, et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73(6):1777–86. doi: 10.1158/0008-5472.CAN-12-3558.
  60. Hamerman JA, Ogasawara K, Lanier LL. Cutting edge: Toll-like receptor signaling in macrophages induces ligands for the NKG2D receptor. J Immunol. 2004;172(4):2001–5. doi: 10.4049/jimmunol.172.4.2001.
  61. Carlsten M, Bjorkstrom NK, Norell H, et al. DNAX accessory molecule-1 mediated recognition of freshly isolated ovarian carcinoma by resting natural killer cells. Cancer Res. 2007;67(3):1317–25. doi: 10.1158/0008-5472.CAN-06-2264.
  62. McGilvray RW, Eagle RA, Rolland P, et al. ULBP2 and RAET1E NKG2D ligands are independent predictors of poor prognosis in ovarian cancer patients. Int J Cancer. 2010;127(6):1412–20. doi: 10.1002/ijc.25156.
  63. Cathro HP, Smolkin ME, Theodorescu D, et al. Relationship between HLA class I antigen processing machinery component expression and the clinicopathologic characteristics of bladder carcinomas. Cancer Immunol Immunother. 2010;59(3):465–72. doi: 10.1007/s00262-009-0765-9.
  64. Seitz S, Hohla F, Schally AV, et al. Inhibition of estrogen receptor positive and negative breast cancer cell lines with a growth hormone-releasing hormone antagonist. Oncol Rep. 2008;20(5):1289–94.
  65. Mamessier E, Sylvain A, Thibult ML, et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 2011;121(9):3609–22. doi: 10.1172/JCI45816.
  66. Busche A, Goldmann T, Naumann U, et al. Natural killer cell-mediated rejection of experimental human lung cancer by genetic overexpression of major histocompatibility complex class I chain-related gene A. Hum Gene Ther. 2006;17(2):135–46. doi: 10.1089/hum.2006.17.135.
  67. Platonova S, Cherfils-Vicini J, Damotte D, et al. Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 2011;71(16):5412–22. doi: 10.1158/0008-5472.CAN-10-4179.
  68. Jinushi M, Takehara T, Tatsumi T, et al. Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int J Cancer. 2003;104(3):354–61. doi: 10.1002/ijc.10966.
  69. Watson NF, Spendlove I, Madjd Z, et al. Expression of the stress-related MHC class I chain-related protein MICA is an indicator of good prognosis in colorectal cancer patients. Int J Cancer. 2006;118(6):1445–52. doi: 10.1002/ijc.21510.
  70. Sconocchia G, Spagnoli GC, Del Principe D, et al. Defective infiltration of natural killer cells in MICA/B-positive renal cell carcinoma involves beta(2)-integrin-mediated interaction. 2009;11(7):662–71. doi: 10.1593/neo.09296.
  71. Wu JD, Higgins LM, Steinle A, et al. Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest. 2004;114(4):560–8. doi: 10.1172/JCI22206.
  72. Salih HR, Antropius H, Gieseke F, et al. Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. 2003;102(4):1389–96. doi: 10.1182/blood-2003-01-0019.
  73. Diermayr S, Himmelreich H, Durovic B, et al. NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. 2008;111(3):1428–36. doi: 10.1182/blood-2007-07-101311.
  74. Sconocchia G, Lau M, Provenzano M, et al. The antileukemia effect of HLA-matched NK and NK-T cells in chronic myelogenous leukemia involves NKG2D-target-cell interactions. 2005;106(10):3666–72. doi: 10.1182/blood-2005-02-0479.
  75. Nuckel H, Switala M, Sellmann L, et al. The prognostic significance of soluble NKG2D ligands in B-cell chronic lymphocytic leukemia. 2010;24(6):1152–9. doi: 10.1038/leu.2010.74.
  76. Zhang B, Kracker S, Yasuda T, et al. Immune surveillance and therapy of lymphomas driven by Epstein-Barr virus protein LMP1 in a mouse model. 2012;148(4):739–51. doi: 10.1016/j.cell.2011.12.031.
  77. Girlanda S, Fortis C, Belloni D, et al. MICA expressed by multiple myeloma and monoclonal gammopathy of undetermined significance plasma cells costimulates pamidronate-activated gammadelta lymphocytes. Cancer Res. 2005;65(16):7502–8. doi: 10.1158/0008-5472.CAN-05-0731.
  78. Paschen A, Sucker A, Hill B, et al. Differential clinical significance of individual NKG2D ligands in melanoma: soluble ULBP2 as an indicator of poor prognosis superior to S100B. Clin Cancer Res. 2009;15(16):5208–15. doi: 10.1158/1078-0432.CCR-09-0886.
  79. Verhoeven DH, de Hooge AS, Mooiman EC, et al. NK cells recognize and lyse Ewing sarcoma cells through NKG2D and DNAM-1 receptor dependent pathways. Mol Immunol. 2008;45(15):3917–25. doi: 10.1016/j.molimm.2008.06.016.
  80. Friese MA, Platten M, Lutz SZ, et al. MICA/NKG2D-mediated immunogene therapy of experimental gliomas. Cancer Res. 2003;63(24):8996–9006.
  81. Raffaghello L, Prigione I, Airoldi I, et al. Downregulation and/or release of NKG2D ligands as immune evasion strategy of human neuroblastoma. 2004;6(5):558–68. doi: 10.1593/neo.04316.
  82. Chitadze G, Lettau M, Bhat J, et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int J Cancer. 2013;133(7):1557–66. doi: 10.1002/ijc.28174.
  83. Zhang T, Lemoi BA, Sentman CL. Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy. 2005;106(5):1544–51. doi: 10.1182/blood-2004-11-4365.
  84. Zhang T, Barber A, Sentman CL. Generation of antitumor responses by genetic modification of primary human T cells with a chimeric NKG2D receptor. Cancer Res. 2006;66(11):5927–33. doi: 10.1158/0008-5472.CAN-06-0130.
  85. Barber A, Zhang T, DeMars LR, et al. Chimeric NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. Cancer Res. 2007;67(10):5003–8. doi: 10.1158/0008-5472.CAN-06-4047.
  86. Barber A, Zhang T, Megli CJ, et al. Chimeric NKG2D receptor-expressing T cells as an immunotherapy for multiple myeloma. Exp Hematol. 2008;36(10):1318–28. doi: 10.1016/j.exphem.2008.04.010.
  87. Barber A, Meehan KR, Sentman CL. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D receptor-expressing T cells. Gene Ther. 2011;18(5):509–16. doi: 10.1038/gt.2010.174.
  88. Barber A, Rynda A, Sentman CL. Chimeric NKG2D expressing T cells eliminate immunosuppression and activate immunity within the ovarian tumor microenvironment. J Immunol. 2009;183(11):6939–47. doi: 10.4049/jimmunol.0902000.
  89. Zhang T, Sentman CL. Cancer immunotherapy using a bispecific NK receptor fusion protein that engages both T cells and tumor cells. Cancer Res. 2011;71(6):2066–76. doi: 10.1158/0008-5472.CAN-10-3200.
  90. Zhang T, Sentman CL. Mouse tumor vasculature expresses NKG2D ligands and can be targeted by chimeric NKG2D-modified T cells. J Immunol. 2013;190(5):2455–63. doi: 10.4049/jimmunol.1201314.
  91. Lehner M, Gotz G, Proff J, et al. Redirecting T cells to Ewing’s sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS One. 2012;7(2):e31210. doi: 10.1371/journal.pone.0031210.
  92. Song DG, Ye Q, Santoro S, et al. Chimeric NKG2D CAR-expressing T cell-mediated attack of human ovarian cancer is enhanced by histone deacetylase inhibition. Hum Gene Ther. 2013;24(3):295–305. doi: 10.1089/hum.2012.143.
  93. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  94. Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra38. doi: 10.1126/scitranslmed.3005930.
  95. Meehan KR, Talebian L, Tosteson TD, et al. Adoptive cellular therapy using cells enriched for NKG2D+CD3+CD8+ T cells after autologous transplantation for myeloma. Biol Blood Marrow Transplant. 2013;19(1):129–37. doi: 10.1016/j.bbmt.2012.08.018.
  96. Nakajima J, Murakawa T, Fukami T, et al. A phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous gammadelta T cells. Eur J Cardiothorac Surg. 2010;37(5):1191–7. doi: 10.1016/j.ejcts.2009.11.051.
  97. Abe Y, Muto M, Nieda M, et al. Clinical and immunological evaluation of zoledronate-activated Vgamma9gammadelta T-cell-based immunotherapy for patients with multiple myeloma. Exp Hematol. 2009;37(8):956–68. doi: 10.1016/j.exphem.2009.04.008.
  98. Gattinoni L, Powell DJ Jr, Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6(5):383–93. doi: 10.1038/nri1842.
  99. June CH. Principles of adoptive T cell cancer therapy. J Clin Invest. 2007;117(5):1204–12. doi: 10.1172/JCI31446.
  100. Morgan RA, Chinnasamy N, Abate-Daga D, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36(2):133–51. doi: 10.1097/CJI.0b013e3182829903.
  101. Morgan RA, Yang JC, Kitano M, et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–51. doi: 10.1038/mt.2010.24.
  102. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. 2005;105(8):3051–7. doi: 10.1182/blood-2004-07-2974.
  103. Sentman CL, Meehan KR. NKG2D CARs as cell therapy for cancer. Cancer J. 2014;20(2):156–9. doi: 10.1097/PPO.0000000000000029.
  104. Lonez C, Hendlisz A, Shaza L, et al. Celyad’s novel CAR T-cell therapy for solid malignancies. Curr Res Transl Med. 2018;66(2):53–6. doi: 10.1016/j.retram.2018.03.001.
  105. Baumeister SH, Murad J, Werner L, et al. Phase I Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol Res. 2019;7(1):100–12. doi: 10.1158/2326-6066.CIR-18-0307.
  106. Al-Homsi S, Purev E, Lewalle P, et al. Interim Results from the Phase I Deplethink Trial Evaluating the Infusion of a NKG2D CAR T-Cell Therapy Post a Non-Myeloablative Conditioning in Relapse or Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. 2019;134(Suppl_1):3844. doi: 10.1182/blood-2019-128267.
  107. Liu H, Wang S, Xin J, et al. Role of NKG2D and its ligands in cancer immunotherapy. Am J Cancer Res. 2019;9(10):2064–78.

Современный взгляд на диагностику и лечение классических Ph-негативных миелопролиферативных заболеваний

А.Л. Меликян1, И.Н. Суборцева1, В.А. Шуваев2,3, Е.Г. Ломаиа4, Е.В. Морозова5, Л.А. Кузьмина1, О.Ю. Виноградова6,7,8, А.Ю. Зарицкий4

1 ФГБУ «НМИЦ гематологии» Минздрава России, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167

2 ФГБУ «Российский НИИ гематологии и трансфузиологии ФМБА России», ул. 2-я Советская, д. 16, Санкт-Петербург, Российская Федерация, 191024

3 ГБУЗ » Городская клиническая больница им. В.В. Вересаева» ДЗМ, ул. Лобненская, д. 10, Москва, Российская Федерация, 127644

4 ФГБУ «НМИЦ им. В.А. Алмазова» Минздрава России, ул. Аккуратова, д. 2, Санкт-Петербург, Российская Федерация, 197341

5 НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой, ФГБОУ ВО «Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова» Минздрава России, ул. Льва Толстого, д. 6/8, Санкт-Петербург, Российская Федерация, 197022

6 ГБУЗ «Городская клиническая больница им. С.П. Боткина» ДЗМ, 2-й Боткинский пр-д, д. 5, Москва, Российская Федерация, 125284

7 ФГБУ «НМИЦ детской гематологии, онкологии и иммунологии им. Дмитрия Рогачева» Минздрава России, ул. Саморы Машела, д. 1, Москва, Российская Федерация, 117997

8 ФГАОУ ВО «РНИМУ им. Н.И. Пирогова» Минздрава России, ул. Островитянова, д. 1, Москва, Российская Федерация, 117997

Для переписки: Анаит Левоновна Меликян, д-р мед. наук, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167; e-mail: anoblood@mail.ru

Для цитирования: Меликян А.Л., Суборцева И.Н., Шуваев В.А. и др. Современный взгляд на диагностику и лечение классических Ph-негативных миелопролиферативных заболеваний. Клиническая онкогематология. 2021;14(1):129–37.

DOI: 10.21320/2500-2139-2021-14-1-129-137


РЕФЕРАТ

Классические Ph-негативные миелопролиферативные заболевания (МПЗ) — группа опухолей, включающая в себя истинную полицитемию, эссенциальную тромбоцитемию и первичный миелофиброз. В последнее десятилетие существенно изменились подходы к пониманию патогенеза и лечению МПЗ. В то же время продолжается тщательное изучение этиологических факторов, патофизиологических механизмов развития заболевания. Совершенствование методов диагностики, новые подходы к лечению способны снизить риски осложнений и летальных исходов. В обзоре представлены современные методы диагностики, в т. ч. молекулярно-генетический, приведены прогностические шкалы. Кроме того, оцениваются различные методы консервативного лечения. Особое внимание уделено оценке качества жизни пациентов и таргетному лечению заболеваний.

Ключевые слова: миелопролиферативные заболевания, истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз, JAK2V617F, CALR, MPL, прогноз, конституциональные симптомы, MPN10, руксолитиниб.

Получено: 1 сентября 2020 г.

Принято в печать: 10 декабря 2020 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Меликян А.Л., Туркина А.Г., Абдулкадыров К.М. и др. Клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз). Гематология и трансфузиология. 2014;59:31–56.
    [Melikyan AL, Turkina AG, Abdulkadyrov KM, et al. Clinical guidelines on diagnosis and therapy of Ph-negative myeloproliferative neoplasms (polycythemia vera, essential thrombocythemia, primary myelofibrosis). Gematologiya i transfuziologiya. 2014;59:31–56. (In Russ)]
  2. Меликян А.Л., Ковригина А.М., Суборцева И.Н. и др. Национальные клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз) (редакция 2018 г.) Гематология и трансфузиология. 2018;63(3):275–315.
    [Melikyan AL, Kovrigina AM, Subortseva IN, et al. National clinical guidelines on diagnosis and therapy of Ph-negative myeloproliferative neoplasms (polycythemia vera, essential thrombocythemia, primary myelofibrosis) (edition 2018). Gematologiya i transfuziologiya. 2018;63(3):275–315. (In Russ)]
  3. Абрамова А.В., Абдуллаев А.О., Азимова М.Х. и др. Алгоритмы диагностики и протоколы лечения заболеваний системы крови. В 2 томах. М.: Практика, 2018. Том 2.
    [Abramova AV, Abdullaev AO, Azimova MKh, et al. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. V 2 tomakh. (Diagnostic algorithms and treatment protocols in hematological diseases. 2 volumes.) Moscow: Praktika Publ.; 2018. 2. (In Russ)]
  4. Меликян А.Л., Суборцева И.Н., Галстян Г.М. Протокол дифференцированного посиндромного лечения больных первичным миелофиброзом. В кн.: Алгоритмы диагностики и протоколы лечения заболеваний системы крови. По ред. А.В. Абрамовой, А.О. Абдуллаева и др. В 2 томах. М.: Практика, 2018. Том 2. С. 777–802.
    [Melikyan AL, Subortseva IN, Galstyan GM. Protocol of differentiated syndromic treatment of patients with primary myelofibrosis. In: Abramova AV, Abdullaev AO, et al., eds. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. V 2 tomakh. (Diagnostic algorithms and treatment protocols in hematological diseases. 2 volumes.) Moscow: Praktika Publ.; 2018. Vol. 2. pр. 777–802. (In Russ)]
  5. Geyer H, Scherber R, Kosiorek H, et al. Symptomatic Profiles of Patients With Polycythemia Vera: Implications of Inadequately Controlled Disease. J Clin Oncol. 2016. 34(2):151–9. doi: 10.1200/JCO.2015.62.9337.
  6. Ионова Т.И., Анчукова Л.В., Виноградова О.Ю. и др. Качество жизни и спектр симптомов у больных миелофиброзом на фоне терапии: данные клинической практики. Гематология и трансфузиология. 2016;61(1):17–25. doi: 10.18821/0234-5730-2016-61-1-17-25.
    [Ionova TI, Anchukova LV, Vinogradova OYu, et al. Quality of life and symptom profile in patients with myelofibrosis undergoing treatment: Data of clinical practice. Gematologiya i transfuziologiya. 2016;61(1):17–25. doi: 10.18821/0234-5730-2016-61-1-17-25. (In Russ)]
  7. Xiao Z, Chang C-S, Morozova E, et al. Impact of myeloproliferative neoplasms (MPNS) and perceptions of treatment goals amongst physicians and patients in 6 countries: an expansion of the MPN landmark survey. 2019;3(s1):294–5. doi: 10.1097/01.hs9.0000561008.75001.e7.
  8. Cervantes F, Dupriez B, Pereira A, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. 2009;113(13):2895–901. doi: 10.1182/blood-2008-07-170449.
  9. Passamonti F, Cervantes F, Vannucchi AM, et al. Dynamic International Prognostic Scoring System (DIPSS) predicts progression to acute myeloid leukemia in primary myelofibrosis. 2010;116(15):2857–8. doi: 10.1182/blood-2010-06-293415.
  10. Gangat N, Caramazza D, Vaidya R, et al. DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol. 2011;29(4):392–7. doi: 10.1200/JCO.2010.32.2446.
  11. Vannucchi AM, Guglielmelli P, Rotunno G, et al. Mutation-Enhanced International Prognostic Scoring System (MIPSS) for primary myelofibrosis: an AGIMM & IWG-MRT project. 2014;124(21):405. doi: 10.1182/blood.v124.21.405.405.
  12. Guglielmelli P, Lasho TL, Rotunno G, et al. MIPSS70: Mutation-Enhanced International Prognostic Score System for transplantation-age patients with primary myelofibrosis. J Clin Oncol. 2018;36(4):310–8. doi: 10.1200/JCO.2017.76.4886.
  13. Passamonti F, Giorgino T, Mora B, et al. A clinical-molecular prognostic model to predict survival in patients with post polycythemia vera and post essential thrombocythemia myelofibrosis. 2017;31(12):2726–31. doi: 10.1038/leu.2017.169.
  14. Robin M, de Wreede LC, Wolschke C, et al. Long-term outcome after allogeneic hematopoietic cell transplantation for myelofibrosis. 2019;104(9):1782–8. doi: 10.3324/haematol.2018.205211.
  15. Барабанщикова М.В., Морозова Е.В., Байков В.В. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при миелофиброзе. Клиническая онкогематология. 2016;9(3):279–86. doi: 10.21320/2500-2139-2016-9-3-279-286.
    [Barabanshchikova MV, Morozova EV, Baykov VV, et al. Allogeneic Hematopoietic Stem Cell Transplantation in Myelofibrosis. Clinical oncohematology. 2016;9(3):279–86. doi: 10.21320/2500-2139-2016-9-3-279-286. (In Russ)]
  16. Виноградова О.Ю., Шуваев В.А., Мартынкевич И.С. и др. Таргетная терапия миелофиброза. Клиническая онкогематология. 2017;10(4):471–8. doi: 10.21320/2500-2139-2017-10-4-471-478.
    [Vinogradova OYu, Shuvaev VA, Martynkevich IS, et al. Targeted Therapy of Myelofibrosis. Clinical oncohematology. 2017;10(4):471–8. doi: 10.21320/2500-2139-2017-10-4-471-478. (In Russ)]
  17. Руксолитиниб (инструкция по медицинскому применению). Доступно по: https://www.vidal.ru/drugs/molecule/2304. Ссылка активна на 22.10.2020.
    [Ruxolitinib (package insert). Available from: https://www.vidal.ru/drugs/molecule/2304. (accessed 22.10.2020) (In Russ)]
  18. Tefferi A, Cervantes F, Mesa R, et al. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European Leukemia Net (ELN) consensus report. 2013;122(8):1395–8. doi: 10.1182/blood-2013-03-488098.
  19. Ломаиа Е.Г., Сиордия Н.Т., Сендерова О.М. и др. Ранний ответ и отдаленные результаты терапии миелофиброза руксолитинибом: многоцентровое ретроспективное исследование в 10 центрах Российской Федерации. Клиническая онкогематология. 2020;13(3):335–45. doi: 10.21320/2500-2139-2020-13-3-335-345.
    [Lomaia EG, Siordiya NT, Senderova OM, et al. Early Response and Long-Term Outcomes of Ruxolitinib Therapy in Myelofibrosis: Multicenter Retrospective Study in 10 Centers of the Russian Federation. Clinical oncohematology. 2020;13(3):335–45. doi: 10.21320/2500-2139-2020-13-3-335-345. (In Russ)]
  20. Lomaia E, Siordiya N, Dimov G, et al. Early spleen response is a good prognostic factor of ruxolinib outcome in patients with myelofibrosis. 2019;3(S1):989. doi: 10.1097/01.hs9.0000567308.09016.52.

Возможности терапии ингибиторами тирозинкиназ в сниженных дозах у пациентов с хроническим миелолейкозом

М.А. Гурьянова, Е.Ю. Челышева, А.Г. Туркина

ФГБУ «НМИЦ гематологии» Минздрава России, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167

Для переписки: Маргарита Анатольевна Гурьянова, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167; тел.: +7(985)201-70-40; e-mail: margarita.samtcova@yandex.ru

Для цитирования: Гурьянова М.А., Челышева Е.Ю., Туркина А.Г. Возможности терапии ингибиторами тирозинкиназ в сниженных дозах у пациентов с хроническим миелолейкозом. Клиническая онкогематология. 2021;14(1):118–28.

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


РЕФЕРАТ

Терапия ингибиторами тирозинкиназ (ИТК) у 60–70 % больных хроническим миелоидным лейкозом (ХМЛ) позволяет получить глубокий молекулярный ответ (МО). Однако, несмотря на высокую эффективность ИТК, у многих пациентов лечение сопровождается проявлениями лекарственной токсичности. По результатам клинических исследований вероятность сохранения ремиссии без лечения у больных ХМЛ с глубоким МО составляет примерно 40–60 %. В последнее время большое внимание уделяется персонализированному подходу к терапии при ХМЛ в хронической фазе. Он заключается в модификации дозы ИТК с целью уменьшить или предотвратить развитие нежелательных явлений. В рамках крупных ретроспективных исследований было доказано, что терапия сниженными дозами ИТК у больных с оптимальным ответом является безопасной с точки зрения сохранения достигнутого большого МО и глубокого МО при стандартных дозах ИТК. Наблюдение за пациентом при применении сниженных доз ИТК также рассматривается в рамках проспективных клинических исследований как этап перед отменой терапии. Тем не менее к настоящему времени опубликованы результаты всего 4 подобных исследований. Для подробного изучения вопросов, касающихся длительного наблюдения за больными ХМЛ при использовании сниженных доз ИТК, необходимо проведение проспективных клинических исследований. В настоящем обзоре представлены результаты основных исследований по ведению больных ХМЛ, получающих ИТК в сниженных дозах.

Ключевые слова: хронический миелоидный лейкоз, ингибиторы тирозинкиназ, большой молекулярный ответ, глубокий молекулярный ответ, нежелательные явления, фармакокинетика ингибиторов тирозинкиназ.

Получено: 3 августа 2020 г.

Принято в печать: 20 ноября 2020 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Туркина А.Г., Новицкая Н.В., Голенков А.К. идр. Регистр больных хроническим миелолейкозом в Российской Федерации: от наблюдательного исследования к оценке эффективности терапии в клинической практике. Клиническая онкогематология. 2017;10(3):390–401. doi: 10.21320/2500-2139-2017-10-3-390-401.
    [Turkina AG, Novitskaya NV, Golenkov AK, et al. Chronic Myeloid Leukemia Patient Registry in the Russian Federation: From Observational Studies to the Efficacy Evaluation in Clinical Practice. Clinical oncohematology. 2017;10(3):390–401. doi: 10.21320/2500-2139-2017-10-3-390-401. (In Russ)]
  2. Sasaki K, Strom SS, O’Brien S, et al. Relative survival in patients with chronic-phase chronic myeloid leukaemia in the tyrosine-kinase inhibitor era: analysis of patient data from six prospective clinical trials. Lancet Haematol. 2015;2(5):e186–е193. doi: 10.1016/S2352-3026(15)00048-4.
  3. Hehlmann R, Lauseker M, Saussele S, et al. Assessment of imatinib as first-line treatment of chronic myeloid leukemia: 10-year survival results of the randomized CML study IV and impact of non-CML determinants. Leukemia. 2017;31(11):2398–406. doi: 10.1038/leu.2017.253.
  4. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.
  5. Etienne G, Guilhot J, Rea D, et al. Long-term follow-up of the French Stop Imatinib (STIM1) study in patients with chronic myeloid leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/jco.2016.68.2914.
  6. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.
  7. Chelysheva EYu, Petrova AN, Shukhov OA, et al. First interim analysis of the Russian multicenter prospective study RU-SKI: discontinuation of tyrosine kinase inhibitors in patients with chronic myeloid leukemia and deep molecular response. Hemasphere. 2018;2(S1):141.
  8. Туркина А.Г., Челышева Е.Ю., Шуваев В.А. и др. Результаты наблюдения больных хроническим миелолейкозом с глубоким молекулярным ответом без терапии ингибиторами тирозинкиназ. Терапевтический архив. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96.
    [Turkina AG, Chelysheva EYu, Shuvaev VA, et al. Results of following up patients with chronic myeloid leukemia and a deep molecular response without tyrosine kinase inhibitor therapy. Terapevticheskii arkhiv. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96. (In Russ)]
  9. Зейфман А.А., Челышева Е.Ю., Туркина А.Г. и др. Роль селективности ингибиторов тирозинкиназ в развитии побочных эффектов при терапии хронического миелолейкоза. Клиническая онкогематология. 2014;7(1):16–27.
    [Zeifman AA, Chelysheva EYu, Turkina AG, et al. Role of tyrosine­kinase inhibitor selectivity in development of adverse effects during treatment of chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2014;7(1):16–27. (In Russ)]
  10. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.
  11. Cortes JE, Saglio G, Kantarjian H, et al. Final 5-Year Study Results of DASISION: The Dasatinib Versus Imatinib Study in Treatment-Naive Chronic Myeloid Leukemia Patients Trial. J Clin Oncol. 2016;34(20):2333–40. doi: 10.1200/jco.2015.64.8899.
  12. Ломаиа Е.Г., Романова Е.Г., Сбитякова Е.И. и др. Эффективность и безопасность ингибиторов тирозинкиназ 2-го поколения (дазатиниб, нилотиниб) в терапии хронической фазы хронического миелолейкоза. Онкогематология. 2013;2:22–33.
    [Lomaia EG, Romanova EG, Sbityakova EI, et al. Efficacy and safety of 2nd generation tyrosine kinase inhibitors (dasatinib, nilotinib) in the treatment of chronic phase of chronic myeloid leukemia. Onkogematologiya. 2013;2:22–33. (In Russ)]
  13. Лазорко Н.С., Ломаиа Е.Г., Романова Е.Г. и др. Ингибиторы тирозинкиназ второго поколения и их токсичность у больных в хронической фазе хронического миелолейкоза. Клиническая онкогематология. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308.
    [Lazorko NS, Lomaia EG, Romanova EG, et al. Second Generation Tyrosine Kinase Inhibitors and Their Toxicity in Treatment of Patients in Chronic Phase of Chronic Myeloid Leukemia. Clinical oncohematology. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308. (In Russ)]
  14. Kantarjian H, Pasquini R, Levy V, et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia resistant to imatinib at a dose of 400 to 600 milligrams daily: two-year follow-up of a randomized phase 2 study (START-R). Cancer. 2009;115(18):4136–47. doi: 10.1002/cncr.24504.
  15. Quintas-Cardama A, Kantarjian H, O’Brien S, et al. Pleural Effusion in Patients With Chronic Myelogenous Leukemia Treated With Dasatinib After Imatinib Failure. J Clin Oncol. 2007;25(25):3908–14. doi: 10.1200/JCO.2007.12.0329.
  16. de Lavallade H, Punnialingam S, Milojkovic D, et al. Pleural effusions in patients with chronic myeloid leukaemia treated with dasatinib may have an immune-mediated pathogenesis. Br J Haematol. 2008;141(5):745–7. doi: 10.1111/j.1365-2141.2008.07108.
  17. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Bosutinib versus imatinib for newly diagnosed chronic myeloid leukemia: results from the randomized BFORE trial. J Clin Oncol. 2017;36(3):231–7. doi: 10.1200/jco.2017.74.7162.
  18. Brummendorf HT, Cortes JE, de Souza CA, et al. Bosutinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukaemia: results from the 24-month follow-up of the BELA trial. Br J Haematol. 2015;168(1):69–81. doi: 10.1111/bjh.13108.
  19. Kerkela R, Grazette I, Yacolti R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med. 2006;12(8):908–16. doi: 10.1038/nm1446.
  20. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/nejmoa1609324.
  21. Hadzijusufovic E, Albrecht-Schgoer K, Huber K, et al. Nilotinib-induced vasculopathy: identifi cation of vascular endothelial cells as a primary target site. Leukemia. 2017;31(11):2388–97. doi: 10.1038/leu.2017.245.
  22. Троицкая Е.А., Вельмакин С.В., Кобалава Ж.Д. Концепция сосудистого возраста: новый инструмент оценки сердечно-сосудистого риска. Артериальная гипертензия. 2017;23(2):160–71. doi: 10.18705/1607-419X-2017-23-2-160-171.
    [Troitskaya EA, Velmakin SV, Kobalava ZD. Concept of vascular age: new tool in cardiovascular risk assessment. Arterial’naya gipertenziya. 2017;23(2):160–71. doi: 10.18705/1607-419X-2017-23-2-160-171. (In Russ)]
  23. Туркина А.Г., Лазарева О.В., Челышева Е.Ю. и др. Результаты терапии больных хроническим миелолейкозом по данным российской части международного многоцентрового популяционного исследования Eutos Population-Based Study (EUTOS-PBS). Гематология и трансфузиология. 2019;64(2):106–21. doi: 10.35754/0234-5730-2019-64-2-106-121.
    [Turkina AG, Lazareva OV, Chelysheva EYu, et al. Treatment outcomes in patients with chronic myeloid leukemia according to the Russian part of the EUTOS Population-Based Study. Russian journal of hematology and transfusiology. 2019;64(2):106–21. doi: 10.35754/0234-5730-2019-64-2-106-121. (In Russ)]
  24. Hehlmann R, Lauseker M, Saussele S, et al. Assessment of imatinib as first-line treatment of chronic myeloid leukemia: 10-year survival results of the randomized CML study IV and impact of non-CML determinants. Leukemia. 2017;31(11):2398–406. doi: 10.1038/leu.2017.253.
  25. Rousselot P, Johnson-Ansah H, Huguet F, et al. Personalized daily doses of imatinib by therapeutic drug monitoring increase the rates of molecular responses in patients with chronic myeloid leukemia. Final results of the randomized OPTIM imatinib study. Blood. 2015;126(23):133. doi: 10.1182/blood.v126.23.133.133.
  26. Shah NP, Rousselot P, Schiffer C, et al. Dasatinib in imatinib-resistant or -intolerant chronic-phase, chronic myeloid leukemia patients: 7-year follow-up of study CA180-034. Am J Hematol. 2016;91(9):869–74. doi: 10.1002/ajh.24423.
  27. Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010;28(14):2381–8. doi: 10.1200/JCO.2009.26.3087.
  28. Ibrahim AR, Milojkovic D, Bua M, et al. Poor adherence is the main reason for loss of CCyR and imatinib failure for CML patients on long term imatinib therapy. Blood. 2010;116(21):3414. doi: 10.1182/blood.v116.21.3414.3414.
  29. Куцев С.И., Шатохин Ю.В. Влияние перерывов терапии иматинибом на достижение цитогенетического и молекулярного ответов у больных хроническим миелолейкозом. Казанский медицинский журнал. 2009;90(6):827–31.
    [Kutsev SI, Shatokhin YuV. Effect of interruptions in imatinib therapy on achievement of cytogenetic and molecular responses in patients with chronic myeloid leukemia. Kazanskii meditsinskii zhurnal. 2009;90(6):827–31. (In Russ)]
  30. Talpaz M, Shah NP, Kantarjian H, et al. Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med. 2006;354(24):2531–41. doi: 10.1056/NEJMoa055229.
  31. Santana-Hernandez P, Pedraza RCP, Duque SG, et al. Low-Dose Dasatinib as First-Line Treatment for Chronic Myeloid Leukemia: Preliminary Report. Blood. 2017;130(Suppl 1):5254.
  32. Naqvi K, Jabbour E, Skinner J, et al. Long-term follow-up of lower dose dasatinib (50 mg daily) as frontline therapy in newly diagnosed chronic-phase chronic myeloid leukemia. Cancer. 2020;126(1):67–75. doi: 10.1002/cncr.32504.
  33. Carella AM, Lerma E. Durable responses in chronic myeloid leukemia patients maintained with lower doses of imatinib mesylate after achieving molecular remission. Ann Hematol. 2007;86(10):749–52. doi: 10.1007/s00277-007-0326-2.
  34. Cervantes F, Correa JG, Perez I, et al. Imatinib dose reduction in patients with chronic myeloid leukemia in sustained deep molecular response. Ann Hematol. 2017;96(1):81–5. doi: 10.1007/s00277-016-2839-z.
  35. Iriyama N, Ohashi K, Hashino S, et al. The efficacy of reduced-dose dasatinib as a subsequent therapy in patients with chronic myeloid leukemia in the chronic phase: the LD-CML study of the Kanto CML Study Group. Intern Med. 2018;57(1):17–23. doi: 10.2169/internalmedicine.9035-17.
  36. Hjorth-Hansen H, Stenke L, Soderlund S, et al. Dasatinib induces fast and deep responses in newly diagnosed chronic myeloid leukaemia patients in chronic phase: clinical results from a randomised phase-2 study (NordCML006). Eur J Haematol. 2015;94(3):243–50. doi: 10.1111/ejh.12423.
  37. Santos FP, Kantarjian H, Fava C, et al. Clinical impact of dose reductions and interruptions of second-generation tyrosine kinase inhibitors in patients with chronic myeloid leukaemia. Br J Haematol. 2010;150(3):303–12. doi: 10.1111/j.1365-2141.2010.08245.x
  38. Russo D, Martinelli G, Malagola M, et al. Effects and outcome of a policy of intermittent imatinib treatment in elderly patients with chronic myeloid leukemia. Blood. 2013;121(26):5138–44. doi: 10.1182/blood-2013-01-480194.
  39. La Rosee P, Martiat P, Leitner A, et al. Improved tolerability by a modified intermittent treatment schedule of dasatinib for patients with chronic myeloid leukemia resistant or intolerant to imatinib. Ann Hematol. 2013;92(10):1345–50. doi: 10.1007/s00277-013-1769-2.
  40. Faber E, Divoka M, Skoumalova I, et al. A lower dosage of imatinib is sufficient to maintain undetectable disease in patients with chronic myeloid leukemia with long-term low-grade toxicity of the treatment. Leuk Lymphoma. 2016;57(2):370–5. doi: 10.3109/10428194.2015.1056184.
  41. Шухов О.А., Гурьянова М.А., Челышева Е.Ю. и др. Оценка стабильности молекулярного ответа у больных хроническим миелоидным лейкозом на сниженных дозах ингибиторов тирозинкиназ второго поколения. Гематология и трансфузиология. 2020;65(1, приложение 1):111–2.
    [Shukhov OA, Gur’yanova MA, Chelysheva EYu, et al. Assessment of molecular response stability in chronic myeloid leukemia patients treated with second generation tyrosine kinase inhibitors. Gematologiya i transfuziologiya. 2020;65(1, Suppl 1):111–2. (In Russ)]
  42. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor dose in patients with chronic myeloid leukaemia with stable major molecular response (DESTINY): an interim analysis of a non-randomised, phase 2 trial. Lancet Haematol. 2017;4(7):e310–е316. doi: 10.1016/s2352-3026(17)30066-2.
  43. Clark RE, Polydoros F, Apperley JF, et al. De-escalation of tyrosine kinase inhibitor therapy before complete treatment discontinuation in patients with chronic myeloid leukaemia (DESTINY): a non-randomised, phase 2 trial. Lancet Haematol. 2019;6(7):e375–е383. doi: 10.1016/S2352-3026(19)30094-8.
  44. Rea D, Cayuela J, Dulucq S, et al. Molecular responses after switching from a standard-dose twice-daily nilotinib regimen to a reduced-dose once-daily schedule in patients with chronic myeloid leukemia: a real life observational study (NILO-RED). Blood 2017;130(1): Abstract 590.
  45. Claudiani S, Apperley J, Khan A, et al. Dose reduction of first and second generation TKI is effective in the maintenance of major molecular response and may predict successful TFR in CML patients. Blood. 2018;132(1): Abstract 3007.
  46. Cayssials E, Torregrosa-Diaz J, Gallego-Hernanz P, et al. Low-dose tyrosine kinase inhibitors before treatment discontinuation do not impair treatment-free remission in chronic myeloid leukemia patients: results of a retrospective study. Cancer. 2020;126(15):3438–47. doi: 10.1002/cncr.32940.
  47. Singh N, Kumar L, Meena R, et al. Drug monitoring of imatinib levels in patients undergoing therapy for chronic myeloid leukaemia: comparing plasma levels of responders and non-responders. Eur J Clin Pharmacol. 2009;65(6):545–9. doi: 10.1007/s00228-009-0621-z.
  48. Larson RA, Druker BJ, Guilhot F, et al. Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study. Blood. 2008;111(8):4022–8. doi: 10.1182/blood-2007-10-116475.
  49. Picard S, Titier K, Etienne G, et al. Trough imatinib plasma levels are associated with both cytogenetic and molecular responses to standard dose imatinib in chronic myeloid leukemia. Blood. 2007;109(8):3496–9. doi: 10.1182/blood-2006-07-036012.
  50. Takahashi N, Wakita H, Miura M, et al. Correlation between imatinib pharmacokinetics and clinical response in Japanese patients with chronic-phase chronic myeloid leukemia. Clin Pharmacol Ther. 2010;88(6):809–13. doi: 10.1038/clpt.2010.186.
  51. Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010;28(14):2381–8. doi: 10.1200/JCO.2009.26.3087.
  52. Куцев С.И., Оксенюк О.С. Мониторинг в терапии хронического миелолейкоза иматинибом. Клиническая онкогематология. 2009;2(3):225–31.
    [Kutsev SI, Oksenyuk OS. Monitoring in imatinib treatment of chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2009;2(3):225–31. (In Russ)]
  53. Larson RA, Yin OQ, Hochhaus A, et al. Population pharmacokinetic and exposure-response analysis of nilotinib in patients with newly diagnosed Ph+ chronic myeloid leukemia in chronic phase. Eur J Clin Pharmacol. 2012;68(5):723–33. doi: 10.1007/s00228-011-1200-7.
  54. Takahashi N, Miura M, Kuroki J, et al. Multicenter phase II clinical trial of nilotinib for patients with imatinib-resistant or -intolerant chronic myeloid leukemia from the East Japan CML study group evaluation of molecular response and he efficacy and safety of nilotinib. Biomark Res. 2014;2(1):6. doi: 10.1186/2050-7771-2-6.
  55. Tanaka C, Yin OQP, Sethuraman V, et al. Clinical pharmacokinetics of the BCR–ABL tyrosine kinase inhibitor nilotinib. Clin Pharmacol Ther. 2010;87(2):197–203. doi: 10.1038/clpt.2009.208.
  56. Miura M. Therapeutic drug monitoring of imatinib, nilotinib, and dasatinib for patients with chronic myeloid leukemia. Biol Pharm Bull. 2015;38(5):645–54. doi: 10.1248/bpb.b15-00103.
  57. Wang X, Roy A, Hochhaus A, et al. Differential effects of dosing regimen on the safety and efficacy of dasatinib: retrospective exposure–response analysis of a phase III study. Clin Pharmacol. 2013;10(5):85–97. doi: 10.2147/CPAA.S42796.
  58. Mita A, Abumiya M, Miura M, et al. Correlation of plasma concentration and adverse effects of bosutinib: standard dose or dose-escalation regimens of bosutinib treatment for patients with chronic myeloid leukemia. Exp Hematol Oncol. 2018;7(1):9. doi: 10.1186/s40164-018-0101-1.

EVI1 -позитивные лейкозы и миелодиспластические синдромы: теоретические и клинические аспекты (обзор литературы)

Н.Н. Мамаев, А.И. Шакирова, Е.В. Морозова, Т.Л. Гиндина

НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой, ФГБОУ ВО «Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова» Минздрава России, ул. Льва Толстого, д. 6/8, Санкт-Петербург, Российская Федерация, 197022

Для переписки: Николай Николаевич Мамаев, д-р мед. наук, профессор, ул. Льва Толстого, д. 6/8, Санкт-Петербург, Российская Федерация, 197022; e-mail: nikmamaev524@gmail.com

Для цитирования: Мамаев Н.Н., Шакирова А.И., Морозова Е.В., Гиндина Т.Л. EVI1-позитивные лейкозы и миелодиспластические синдромы: теоретические и клинические аспекты (обзор литературы). Клиническая онкогематология. 2021;14(1):103–17.

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


РЕФЕРАТ

Настоящий обзор посвящен анализу теоретической базы и проводимой в клинике НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой терапии наиболее неблагоприятных в прогностическом отношении EVI1-позитивных вариантов миелоидных лейкозов и миелодиспластических синдромов. Основной акцент в работе сделан на доказательстве ведущей роли гена EVI1 в нарушении эпигенетической регуляции гемопоэза и, следовательно, целесообразности использования трансплантации аллогенных гемопоэтических стволовых клеток с гипометилирующими агентами и/или транс-ретиноевой кислотой для лечения этих заболеваний.

Ключевые слова: EVI1, острые миелоидные лейкозы, хронический миелоидный лейкоз, миелодиспластический синдром, аллоТГСК, гипометилирующие агенты, транс-ретиноевая кислота.

Получено: 12 сентября 2020 г.

Принято в печать: 6 декабря 2020 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  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.

Мантийноклеточная лимфома: история, современные принципы диагностики, лечение (обзор литературы)

Г.С. Тумян

ФГБУ «НМИЦ онкологии им. Н.Н. Блохина» Минздрава России, Каширское ш., д. 24, Москва, Российская Федерация, 115478

Для переписки: Гаяне Сепуговна Тумян, д-р мед. наук, Каширское ш., д. 24, Москва, Российская Федерация, 115478; e-mail: gaytum@mail.ru

Для цитирования: Тумян Г.С. Мантийноклеточная лимфома: история, современные принципы диагностики, лечение (обзор литературы). Клиническая онкогематология. 2020;13(4):366–81.

DOI: 10.21320/2500-2139-2020-13-4-366-381


РЕФЕРАТ

Мантийноклеточная лимфома (МКЛ) — гетерогенное заболевание с широким спектром клинических проявлений от редких индолентных случаев, не требующих немедленного начала терапии, до агрессивных, быстро пролиферирующих типов опухоли. Разное клиническое поведение имеет молекулярное обоснование, которое позволило в последней редакции классификации опухолей кроветворной и лимфоидной тканей ВОЗ разделить МКЛ на два варианта: классический (в большинстве случаев) и индолентный. Последние десятилетия расширили наши представления о биологии и механизмах развития заболевания. Это делает возможным в дальнейшем стратифицировать больных на разные группы риска не только в зависимости от клинических факторов (MIPI), но и с учетом молекулярных и биологических особенностей опухоли (индекс пролиферации Ki-67, мутации генов ТР53, NOTCH1, NOTCH2, комплексный кариотип, немутантный статус IGHV). Алгоритмы лечения, основанные на интенсивной химиотерапии с включением цитарабина в высоких дозах, трансплантации аутологичных гемопоэтических стволовых клеток с дальнейшей поддерживающей терапией ритуксимабом, позволили длительно контролировать заболевание у значительного числа больных МКЛ. Применение новых «chemo-free» режимов и рациональных комбинаций (бортезомиб, ингибиторы BTK, леналидомид, венетоклакс) вселяет надежду на возможность ухода от традиционной химиотерапии у определенной части пациентов. Появление новых лекарственных средств с уникальными механизмами действия позволили в какой-то степени снять «печать фатальности» с МКЛ.

Ключевые слова: мантийноклеточная лимфома, лечение.

Получено: 17 июня 2020 г.

Принято в печать: 2 сентября 2020 г.

Читать статью в PDF


ЛИТЕРАТУРА

  1. Rappaport H, Winter WJ, Hicks EB. Follicular lymphoma. A re-evaluation of its position in the scheme of malignant lymphoma, based on a survey of 253 cases. 1956;9(4):792–821. doi: 10.1002/1097-0142(195607/08)9:4<792::aid-cncr2820090429>3.0.co;2-b.

  2. Gerard-Marchant R, Hamlin I, Lennert K, et al. Classification of non-Hodgkin’s lymphomas. Lancet. 1974;2:406–8.

  3. Tubbs RR, Fishleder A, Weiss RA, et al. Immunohistologic cellular phenotypes of lymphoproliferative disorders. Comprehensive evaluation of 564 cases including 257 non-Hodgkin’s lymphomas classified by the International Working Formulation. Am J Pathol. 1983;113(2):207–21.

  4. Banks PM, Chan J, Cleary ML, et al. Mantle cell lymphoma: a proposal for unification of morphologic, immunologic, and molecular data. Am J Surg Pathol. 1992;16(7):637–40. doi: 10.1097/00000478-199207000-00001.

  5. Harris NL, Jaffe ES, Stein H, Banks PM. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood. 1994;84(5):1361–92. doi: 10.1182/blood.v84.5.1361.bloodjournal8451361.

  6. Dreyling M, Klapper W, Rule S. Blastoid and pleomorphic mantle cell lymphoma: still a diagnostic and therapeutic challenge! Blood. 2018;132(26):2722–9. doi: 10.1182/blood-2017-08-737502.

  7. Hernandez L, Fest T, Cazorla M, et al. p53 gene mutations and protein overexpression are associated with aggressive variants of mantle cell lymphomas. Blood. 1996;87(8):3351–9. doi: 10.1182/blood.v87.8.3351.bloodjournal8783351.

  8. Stefancikova L, Moulis M, Fabian P, et al. Loss of the p53 tumor suppressor activity is associated with negative prognosis of mantle cell lymphoma. Int J Oncol. 2010;36(3):699–706. doi: 10.3892/ijo_00000545.

  9. Aukema SM, Hoster E, Rosenwald A, et al. Expression of TP53 is associated with the outcome of MCL independent of MIPI and Ki-67 in trials of the European MCL Network. Blood. 2018;131(4):417–20. doi: 10.1182/blood-2017-07-797019.

  10. Hoster E, Rosenwald A, Berger F, et al. Prognostic Value of Ki-67 Index, Cytology, and Growth Pattern in Mantle-Cell Lymphoma: Results from Randomized Trials of the European Mantle Cell Lymphoma Network. J Clin Oncol. 2016;34(12):1386–94. doi: 10.1200/JCO.2015.63.8387.

  11. Jiang W, Kahn SM, Zhou P, et al. Overexpression of Cyclin D1 in Rat Fibroblasts Causes Abnormalities in Growth Control, Cell Cycle Progression and Gene Expression. Oncogene. 1993;8(12):3447–57.

  12. Jaffe ES, Harris NL, Stein H, Isaacson PG. Classification of lymphoid neoplasms: the microscope as a tool for disease discovery. Blood. 2008;112(12):4384–99. doi: 10.1182/blood-2008-07-077982.

  13. Klener P. Advances in Molecular Biology and Targeted Therapy of Mantle Cell Lymphoma. Int J Mol Sci. 2019;20(18):4417. doi: 10.3390/ijms20184417.

  14. Jares P, Colomer D, Campo E. Genetic and molecular pathogenesis of mantle cell lymphoma: Perspectives for new targeted therapeutics. Nat Rev Cancer. 2007;7(10):750–62. doi: 10.1038/nrc2230.

  15. Vincent-Fabert C, Fiancette R, Rouaud P, et al. A defect of the INK4-Cdk4 checkpoint and Myc collaborate in blastoid mantle cell lymphoma-like lymphoma formation in mice. Am J Pathol. 2012;180(4):1688–701. doi: 10.1016/j.ajpath.2012.01.004.

  16. Choe JY, Yun JY, Na HY, et al. MYC overexpression correlates with MYC amplification or translocation, and is associated with poor prognosis in mantle cell lymphoma. Histopathology. 2016;68(3):442–9. doi: 10.1111/his.12760.

  17. Salaverria I, Royo C, Carvajal-Cuenca A, et al. CCND2 rearrangements are the most frequent genetic events in cyclin D1– mantle cell lymphoma. Blood. 2013;121(8):1394–402. doi: 10.1182/blood-2012-08-452284.

  18. Bea S, Valdes-Mas R, Navarro A, et al. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci USA. 2013;110(45):18250–5. doi: 10.1073/pnas.1314608110.

  19. Vegliante MC, Palomero J, Perez-Galan P, et al. SOX11 regulates PAX5 expression and blocks terminal B-cell differentiation in aggressive mantle cell lymphoma. Blood. 2013;121(12):2175–85. doi: 10.1182/blood-2012-06-438937.

  20. Kuo PY, Jatiani SS, Rahman AH, et al. SOX11 augments BCR signaling to drive MCL-like tumor development. Blood. 2018;131(20):2247–55. doi: 10.1182/blood-2018-02-832535.

  21. Balsas P, Palomero J, Eguileor A, et al. SOX11 promotes tumor protective microenvironment interactions through CXCR4 and FAK regulation in mantle cell lymphoma. Blood. 2017;130(4):501–13. doi: 10.1182/blood-2017-04-776740.

  22. Narurkar R, Alkayem M, Liu D. SOX11 is a biomarker for cyclin D1-negative mantle cell lymphoma. Biomark Res. 2016;4(1):6. doi: 10.1186/s40364-016-0060-9.

  23. Zhang J, Jima D, Moffitt AB, et al. The genomic landscape of mantle cell lymphoma is related to the epigenetically determined chromatin state of normal B cells. Blood. 2014;123(19):2988–96. doi: 10.1182/blood-2013-07-517177.

  24. Yang P, Zhang W, Wang J, et al. Genomic landscape and prognostic analysis of mantle cell lymphoma. Cancer Gene Ther. 2018;25(5–6):129–40. doi: 10.1038/s41417-018-0022-5.

  25. Eskelund CW, Dahl C, Hansen JW, et al. TP53 Mutations Identify Younger Mantle Cell Lymphoma Patients Who Do Not Benefit From Intensive Chemoimmunotherapy. Blood. 2017;130(17):1903–10. doi: 10.1182/blood-2017-04-779736.

  26. Morello L, Rattotti S, Giordano L, et al. Mantle Cell Lymphoma of Mucosa-Associated Lymphoid Tissue. A European Mantle Cell Lymphoma Network Study. Hemasphere. 2019;4(1):e302. doi: 10.1097/hs9.0000000000000302.

  27. Hoster E, Dreyling M, Klapper W, et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood. 2008;111(2):558–65. doi: 10.1182/blood-2007-06-095331.

  28. Scott DW, Abrisqueta P, Wright GW, et al. New Molecular Assay for the Proliferation Signature in Mantle Cell Lymphoma Applicable to Formalin-Fixed Paraffin-Embedded Biopsies. J Clin Oncol. 2017;35(15):1668–77. doi: 10.1200/jco.2016.70.7901.

  29. Kridel R, Meissner B, Rogic S, et al. Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma. Blood. 2012;119(9):1963–71. doi: 10.1182/blood-2011-11-391474.

  30. Halldorsdottir AM, Lundin A, Murray F, et al. Impact of TP53 mutation and 17p deletion in mantle cell lymphoma. Leukemia. 2011;25(12):1904–8. doi: 10.1038/leu.2011.162.

  31. Delfau-Larue MH, Klapper W, Berger F, et al. European Mantle Cell Lymphoma Network. High-dose cytarabine does not overcome the adverse prognostic value of CDKN2A and TP53 deletions in mantle cell lymphoma. Blood. 2015;126(5):604–11. doi: 10.1182/blood-2015-02-628792.

  32. Cohen JB. TP53 mutations in MCL: more therapy is not better. Blood. 2017;130(17):1876–7. doi: 10.1182/blood-2017-08-803551.

  33. Jain P, Wang M. Mantle cell lymphoma: 2019 update on the diagnosis, pathogenesis, prognostication, and management. Am J Hematol. 2019;94(6):710–25. doi: 10.1002/ajh.25487.

  34. Chihara D, Cheah CY, Westin JR, et al. Rituximab plus hyper-CVAD alternating with MTX/Ara-C in patients with newly diagnosed mantle cell lymphoma: 15-year follow-up of a phase II study from the MD Anderson Cancer Center. Br J Haematol. 2016;172(1):80–8. doi: 10.1111/bjh.13796.

  35. Romaguera JE, Wang M, Feng L, et al. Phase 2 trial of bortezomib in combination with rituximab plus hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with bortezomib, rituximab, methotrexate, and cytarabine for untreated mantle cell lymphoma. Cancer. 2018;124(12):2561–9. doi: 10.1002/cncr.31361.

  36. Merli F, Luminari S, Ilariucci F, et al. Rituximab plus HyperCVAD alternating with high dose cytarabine and methotrexate for the initial treatment of patients with mantle cell lymphoma, a multicentre trial from Gruppo Italiano studio Linfomi. Br J Haematol. 2012;156(3):346–53. doi: 10.1111/j.1365-2141.2011.08958.x.

  37. Bernstein SH, Epner E, Unger JM, et al. A phase II multicenter trial of hyperCVAD MTX/Ara-C and rituximab in patients with previously untreated mantle cell lymphoma; SWOG 0213. Ann Oncol. 2013;24(6):1587–93. doi: 10.1093/annonc/mdt070.

  38. Eskelund CW, Kolstad A, Jerkeman M, et al. 15-year follow-up of the second Nordic mantle cell lymphoma trial (MCL2): prolonged remissions without survival plateau. Br J Haematol. 2016;175(3):410–8. doi: 10.1111/bjh.14241.

  39. Hermine O, Hoster E, Walewski J, et al. Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL younger): a randomised, open-label, phase 3 trial of the European mantle cell lymphoma network. Lancet. 2016;388(10044):565–75. doi: 10.1016/S0140-6736(16)00739-X.

  40. Armand P, Redd R, Bsat J, et al. A phase 2 study of Rituximab-Bendamustine and Rituximab-Cytarabine for transplant-eligible patients with mantle cell lymphoma. Br J Haematol. 2016;173(1):89–95. doi: 10.1111/bjh.13929.

  41. Le Gouill S, Thieblemont C, Oberic L, et al. Rituximab after autologous stem-cell transplantation in mantle-cell lymphoma. N Engl J Med. 2017;377(13):1250–60. doi: 10.1056/NEJMoa1701769.

  42. Kluin-Nelemans HC, Hoster E, Hermine O, et al. Treatment of Older Patients With Mantle Cell Lymphoma (MCL): Long-Term Follow-Up of the Randomized European MCL Elderly Trial. J Clin Oncol. 2020;38(3):248–56. doi: 10.1200/JCO.19.01294.

  43. Rummel MJ, Niederle N, Maschmeyer G, et al. Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial. Lancet. 2013;381(9873):1203–10. doi: 10.1016/S0140-6736(12)61763-2.

  44. Flinn IW, van der Jagt R, Kahl BS, et al. Randomized trial of bendamustine-rituximab or RCHOP/RVP in first line treatment of indolent NHL or MCL: the BRIGHT study. Blood. 2014;123(19):2944–52. doi: 10.1182/blood-2013-11-531327.

  45. Kluin-Nelemans JC, Hoster E, Hermine O, et al. Treatment of older patients with mantle cell lymphoma. N Engl J Med. 2012;367(6):520–31. doi: 10.1056/NEJMoa1200920.

  46. Robak T, Huang H, Jin J, et al. Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N Engl J Med. 2015;372(10):944–53. doi: 10.1056/NEJMoa1412096.

  47. Robak T, Jin J, Pylypenko H, et al. Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: final overall survival results of a randomised, open-label, phase 3 study. Lancet Oncol. 2018;19(11):1449–58. doi: 10.1016/S1470-2045(18)30685-5.

  48. Ratnasingam S, Casan J, Shortt J, et al. Cytarabine-based induction immunochemotherapy in the front-line treatment of older patients with mantle cell lymphoma. Sci Rep. 2019;9(1):13544. doi: 10.1038/s41598-019-49776-9.

  49. Visco C, Finotto S, Zambello R, et al. Combination of Rituximab, Bendamustine, and Cytarabine for Patients With Mantle-Cell Non-Hodgkin Lymphoma Ineligible for Intensive Regimens or Autologous Transplantation. J Clin Oncol. 2013;31(11):1442–9. doi: 10.1200/JCO.2012.45.9842.

  50. Visco C, Chiappella A, Nassi L, et al. Rituximab, bendamustine, and low-dose cytarabine as induction therapy in elderly patients with mantle cell lymphoma: a multicentre, phase 2 trial from Fondazione Italiana Linfomi. Lancet Haematol. 2017;4(1):e15–e23. doi: 10.1016/S2352-3026(16)30185-5.

  51. Ruan J, Martin P, Christos P, et al. Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma. Blood. 2018;132(19):2016–25. doi: 10.1182/blood-2018-07-859769.

  52. Hoster E, Pott C. Minimal residual disease in mantle cell lymphoma: insights into biology and impact on treatment. Hematology. 2016;2016(1):437–45. doi: 10.1182/asheducation-2016.1.437.

  53. Pott C, Bruggemann M, Ritgen M, et al. MRD detection in B-cell non-Hodgkin lymphomas using Ig gene rearrangements and chromosomal translocations as targets for real-time quantitative PCR. In: R Kuppers (ed). Lymphoma. Methods in Molecular Biology (Methods and Protocols). Vol. 971. Totowa: Humana Press; 2013. рр. 175–200. doi: 10.1007/978-1-62703-269-8_10.

  54. Kumar A, Sha F, Toure A, et al. Patterns of survival in patients with recurrent mantle cell lymphoma in the modern era: Progressive shortening in response duration and survival after each relapse. Blood Cancer J. 2019;9(6):50. doi: 10.1038/s41408-019-0209-5.

  55. Herrmann A, Hoster E, Zwingers T, et al. Improvement of overall survival in advanced stage mantle cell lymphoma. J Clin Oncol. 2009;27(4):511–8. doi: 10.1200/JCO.2008.16.8435.

  56. Fisher RI, Bernstein SH, Kahl BS, et al. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2006;24(30):4867–74. doi: 10.1200/JCO.2006.07.9665.

  57. Chen RW, Palmer JM, Tomassetti S, et al. Multi-center phase II trial of bortezomib and rituximab maintenance combination therapy in patients with mantle cell lymphoma after consolidative autologous stem cell transplantation. J Hematol Oncol. 2018;11(1):87. doi: 10.1186/s13045-018-0631-3.

  58. Albertsson-Lindblad A, Kolstad A, Laurell A, et al. Lenalidomide-bendamustine-rituximab in patients older than 65 years with untreated mantle cell lymphoma. Blood. 2016;128(14):1814–20. doi: 10.1182/blood-2016-03-704023.

  59. Wang ML, Blum KA, Martin P, et al. Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood. 2015;126(6):739–45. doi: 10.1182/blood-2015-03-635326.

  60. Rule S, Jurczak W, Jerkeman M, et al. Ibrutinib vs temsirolimus: 3-year follow-up of patients with previously treated mantle cell lymphoma from the phase 3, international, randomized, open-label RAY study. Leukemia. 2018;32(8):1799–803. doi: 10.1038/s41375-018-0023-2.

  61. Rule S, Dreyling M, Goy A, et al. Ibrutinib for the treatment of relapsed/refractory mantle cell lymphoma: extended 3.5-year follow-up from a pooled analysis. Haematologica. 2019;194(5):е211–е214. doi: 10.3324/haematol.2018.205229.

  62. Wang M, Rule S, Zinzani PL, et al. Long-Term Follow-Up of Acalabrutinib Monotherapy in Patients with Relapsed/Refractory Mantle Cell Lymphoma. Clin Lymphoma Myel Leuk. 2019;19:S316. doi: 10.1016/j.clml.2019.07.291.

  63. Telford C, Kabadi SM, Abhyankar S, et al. Matching-adjusted Indirect Comparisons of the Efficacy and Safety of Acalabrutinib Versus Other Targeted Therapies in Relapsed/Refractory Mantle Cell Lymphoma. Clin Ther. 2019;41(11):2357–79.e1. doi: 10.1016/j.clinthera.2019.09.012.

  64. Davids MS, Roberts AW, Seymour JF, et al. Phase I first-in-human study of Venetoclax in patients with relapsed or refractory non-Hodgkin lymphoma. J Clin Oncol. 2017;35(8):826–33. doi: 10.1200/JCO.2016.70.4320.

  65. Tam CS, Anderson MA, Pott C, et al. Ibrutinib plus Venetoclax for the treatment of mantle-cell lymphoma. N Engl J Med. 2018;378(13):1211–23. doi: 10.1056/NEJMoa1715519.

  66. Hess G, Herbrecht R, Romaguera J, et al. Phase III study to evaluate temsirolimus compared with investigator’s choice therapy for the treatment of relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2009;27(23):3822–9. doi: 10.1200/JCO.2008.20.7977.

  67. Kahl BS, Spurgeon SE, Furman RR, et al. A phase 1 study of the PI3Kδ inhibitor idelalisib in patients with relapsed/refractory mantle cell lymphoma (MCL). Blood. 2014;123(22):3398–405. doi: 10.1182/blood-2013-11-537555.

Пироптоз — воспалительная форма клеточной гибели

А.А. Вартанян, В.С. Косоруков

ФГБУ «НМИЦ онкологии им. Н.Н. Блохина» Минздрава России, Каширское ш., д. 24, Москва, Российская Федерация, 115478

Для переписки: Aмалия Арташевна Вартанян, д-р биол. наук, ул. Каширская, д. 24, Москва, Российская Федерация, 115478; тел.: +7(499)324-10-65; e-mail: zhivotov57@mail.ru

Для цитирования: Вартанян А.А., Косоруков В.С. Пироптоз — воспалительная форма клеточной гибели. Клиническая онкогематология. 2020;13(2):129–35.

DOI: 10.21320/2500-2139-2020-13-2-129-135


РЕФЕРАТ

Пироптоз, каспаза-1-зависимая воспалительная форма гибели клетки, индуцируется внутриклеточными патогенами или повреждением тканей. Активация прокаспазы-1, необходимая для процессинга провоспалительных цитокинов про-IL-1β и про-IL-18, происходит в макромолекулярных белковых комплексах, называемых инфламмасомами. При инфекциях, вызванных грамотрицательными бактериями, в сборке инфламмасомы участвует каспаза-4 и каспаза-5. Первоначально идентифицированный как защитный механизм врожденного иммунитета, пироптоз сегодня не ограничивается ингибированием размножения внутриклеточных патогенов. В данном обзоре обсуждаются молекулярные механизмы гибели клетки по типу пироптоза и возможности вовлечения пироптоза в гибель опухолевых клеток.

Ключевые слова: клеточная гибель, пироптоз, каспазы, воспаление, врожденный иммунитет.

Получено: 24 декабря 2019 г.

Принято в печать: 17 марта 2020 г.

Читать статью в PDF


ЛИТЕРАТУРА

  1. Zychlinsky A, Prevost MC, Sansonetti PJ. Shigella flexneri induces apoptosis in infected macrophages. Nature. 1992;358(6382):167–9. doi: 10.1038/358167a0.

  2. Brennan MA, Cookson BT. Salmonella induces macrophage death by caspase-1-dependent necrosis. Mol Microbiol. 2000;38(1):31–40. doi: 10.1046/j.1365-2958.2000.02103.x.

  3. Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–4. doi: 10.1016/s0966-842x(00)01936-3.

  4. Li P, Allen H, Banerjee S, et al. Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock. Cell. 1995;80(3):401–11. doi: 10.1016/0092-8674(95)90490-5.

  5. Yuan YY, Xie KX, Wang SL, et al. Inflammatory caspase-related pyroptosis: mechanism, regulation and therapeutic potential for inflammatory bowel disease. Gastroenterology Report. 2018;6(3):167–76. doi: 10.1093/gastro/goy011.

  6. Doitsh G, Cavrois M, Lassen KG, et al. Abortive HIV infection mediates CD4 T cell depletion and inflammation in human lymphoid tissue. Cell. 2010;143(5):789–801. doi: 10.1016/j.cell.2010.11.001.

  7. Franchi L, Warner N, Viani K, et al. Function of Nod-like receptors in microbial recognition and host defense. Immunol Rev. 2009;227(1):106–28. doi: 10.1111/j.1600-065x.2008.00734.x.

  8. Bortoluci KR, Medzhitov R. Control of infection by pyroptosis and autophagy: role of TLR and NLR. Cell Mol Life Sci. 2010;67(10):1643–51. doi: 10.1007/s00018-010-0335-5.

  9. Lamkanfi M, Dixit V. Mechanisms and functions of inflammasomes. Cell. 2014;157(5):1013–22. doi: 10.1016/j.cell.2014.04.007.

  10. Man SM, Karki R, Kanneganti TD. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev. 2017;277(1):61–75. doi: 10.1111/imr.12534.

  11. Malireddi RK, Ippagunta S, Lamkanfi M, et al. Cutting edge: proteolytic inactivation of poly(ADP-ribose) polymerase 1 by the Nlrp3 and Nlrc4 inflammasomes. J Immunol. 2010;185(6):3127–30. doi: 10.4049/jimmunol.1001512.

  12. Kovacs SB, Miao EA. Gasdermins: Effectors of Pyroptosis. Trends Cell Biol. 2017;27(9):673–84. doi: 10.1016/j.tcb.2017.05.005.

  13. Feng S, Fox D, Man SM. Mechanisms of Gasdermin Family Members in Inflammasome Signaling and Cell Death. J Mol Biol. 2018;430(18):3068–80. doi: 10.1016/j.jmb.2018.07.002.

  14. Prajwal G, Lukens JR, Thirumala-Devi K. Mitochondria: diversity in the regulation of the NLRP3 inflammasome. Trends Mol Med. 2015;21(3):193–201. doi: 10.1016/j.molmed.2014.11.008.

  15. Yu J, Nagasu H, Murakami T, et al. Inflammasome activation leads to Caspase-1-dependent mitochondrial damage and block of mitophagy. Proc Natl Acad Sci USA. 2014;111(43):15514–9. doi: 10.1073/pnas.1414859111.

  16. Diamond CE, Khameneh HJ, Brough D, et al. Novel perspectives on non-canonical inflammasome activation. Immunotargets Ther. 2015;4:131–41. doi: 10.2147/ITT.S57976.

  17. Lamkanfi M, Dixit VM. Modulation of inflammasome pathways by bacterial and viral pathogens. J Immunol. 2011;187(2):597–602. doi: 10.4049/jimmunol.1100229.

  18. Strowig T, Henao-Mejia J, Elinav E, et al. Inflammasomes in health and disease. Nature. 2012;481(7381):278–86. doi: 10.1038/nature10759.

  19. Rashidi M, Simpson DS, Hempel A, et al. The Pyroptotic Cell Death Effector Gasdermin D Is Activated by Gout-Associated Uric Acid Crystals but Is Dispensable for Cell Death and IL-1β J Immunol. 2019;203(3):736–48. doi: 10.4049/jimmunol.1900228.

  20. Sahoo AK, Dandapat J, Dash UC, et al. Features and outcomes of drugs for combination therapy as multi-targets strategy to combat Alzheimer’s disease. J Ethnopharmacol. 2018;215:42–73. doi: 10.1016/j.jep.2017.12.015.

  21. Freeman LC, Ting JP. The pathogenic role of the inflammasome in neurodegenerative diseases. J Neurochem. 2016;136(Suppl 1):29–38. doi: 10.1111/jnc.13217.

  22. Corrigan KL, Wall KC, Bartlett JA, et al. Cancer disparities in people with HIV: A systematic review of screening for non-AIDS-defining malignancies. 2019;125(6):843–53. doi: 10.1002/cncr.31838.

  23. Doitsh G, Galloway NL, Geng X, et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature. 2014;505(7484):509–14. doi: 10.1038/nature12940.

  24. Monroe KM, Yang Z, Johnson JR, et al. IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science. 2014;343(6169):428–32. doi: 10.1126/science.1243640.

  25. Wannamaker W, Davies R, Namchuk M, et al. VX-765, an orally available selective interleukin (IL)-converting enzyme/caspase-1 inhibitor, exhibits potent anti-inflammatory activities by inhibiting the release of IL-1beta and IL-18. J Pharmacol Exp Ther. 2007;321(2):509–16. doi: 10.1124/jpet.106.111344.

  26. Asahchop EL, Meziane O, Mamik MK, et al. Reduced antiretroviral drug efficacy and concentration in HIV-infected microglia contributes to viral persistence in brain. Retrovirology. 2017;14(1):47. doi: 10.1186/s12977-017-0370-5.

  27. Mamik MK, Hui E, Branton WG, et al. HIV-1 Viral Protein R Activates NLRP3 Inflammasome in Microglia: Implications for HIV-1 Associated Neuroinflammation. J Neuroimmune Pharmacol. 2017;12(2):233–48. doi: 10.1007/s11481-016-9708-3.

  28. Vandanmagsar B, Youm Y, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17(2):179–88. doi: 10.1038/nm.2279.

  29. Nagarajan K, Soundarapandian K, Thorne RF, et al. Activation of Pyroptotic Cell Death Pathways in Cancer: An Alternative Therapeutic Approach. Transl Oncol. 2019;12(7):925–31. doi: 10.1016/j.tranon.2019.04.010.

  30. Luan J, Ju D. Inflammasome: A Double-Edged Sword in Liver Front Immunol. 2018;9:2201–9. doi: 10.3389/fimmu.2018.02201.

  31. Chu Q, Jiang Y, Zhang W, et al. Pyroptosis is involved in the pathogenesis of human hepatocellular carcinoma. Oncotarget. 2016;7(51):84658–65. doi: 10.18632/oncotarget.12384.

  32. Place DE, Kanneganti TD. Recent advances in inflammasome biology. Curr Opin Immunol. 2018;50:32–8. doi: 10.1016/j.coi.2017.10.011.

  33. Wang H, Luo Q, Feng X, et al. NLRP3 promotes tumor growth and metastasis in human oral squamous cell carcinoma. BMC Cancer. 2018;18(1):500–12. doi: 10.1186/s12885-018-4403-9.

  34. Elion DL, Jacobson ME, Hicks DJ, et al. Therapeutically Active RIG-I Agonist Induces Immunogenic Tumor Cell Killing in Breast Cancer Res. 2018;78(21):6183–95. doi: 10.1158/0008-5472.can-18-0730.

  35. Pizato N, Luzete BC, Kiffer LV, et al. Omega-3 docosahexaenoic acid induces pyroptosis cell death in triple-negative breast cancer cells. Sci Rep. 2018;8(1):1952–60. doi: 10.1038/s41598-018-20422-0.

  36. Baranovskiy AG, Babayeva ND, Suwa Y, et al. Structural basis for inhibition of DNA replication by aphidicolin. Nucl Acids Res. 2014;42(22):14013–21. doi: 10.1093/nar/gku1209.

  37. Han T, Goralski M, Capota E, et al. The antitumor toxin CD437 is a direct inhibitor of DNA polymerase α. Nat Chem Biol. 2016;12(7):511–5. doi: 10.1038/nchembio.2082.

  38. Gutteridge RE, Ndiaye MA, Liu X, et al. Plk1 Inhibitors in Cancer Therapy: From Laboratory to Clinics. Mol Cancer 2016;15(7):1427–35. doi: 10.1158/1535-7163.mct-15-0897.

  39. Liu X. Targeting Polo-Like Kinases: A Promising Therapeutic Approach for Cancer Transl Oncol. 2015;8(3):185–95. doi: 10.1016/j.tranon.2015.03.010.

  40. Konjevic GM, Vuletic AM, Mirjacic Martinovic KM, et al. The role of cytokines in the regulation of NK cells in the tumor environment. Cytokine. 2019;117:30–40. doi: 10.1016/j.cyto.2019.02.001.

  41. Nakanishi K. Unique Action of Interleukin-18 on T Cells and Other Immune Cells. Front Immunol. 2018;9:763. doi: 10.3389/fimmu.2018.00763.

  42. Cao R, Farnebo J, Kurimoto M, et al. Interleukin-18 acts as an angiogenesis and tumor suppressor FASEB J. 1999;13(15):2195–202. doi: 10.1096/fasebj.13.15.2195.

  43. Li A, Yi M, Qin S, et al. Prospects for combining immune checkpoint blockade with PARP inhibition. J Hematol Oncol. 2019;12(1):98. doi: 10.1186/s13045-019-0784-8.