Использование достижений современных геномных технологий при лимфомах

М.В. Немцова1, М.В. Майорова2

1 Российская медицинская академия последипломного образования Минздрава России, ул. Баррикадная, д. 2/1, Москва, Российская Федерация, 125993

2 Московский научно-исследовательский онкологический институт им. П.А. Герцена, 2-й Боткинский пр-д, д. 3, Москва, Российская Федерация, 125284

Для переписки: Марина Вячеславовна Немцова, д-р биол. наук, профессор, ул. Баррикадная, д. 2/1, Москва, Российская Федерация, 125993; тел.: +7(499)252-21-04; e-mail: nemtsova_m_v@mail.ru

Для цитирования: Немцова М.В., Майорова М.В. Использование достижений современных геномных технологий при лимфомах. Клиническая онкогематология. 2016;9(3):265-70.

DOI: 10.21320/2500-2139-2016-9-3-265-270


РЕФЕРАТ

Современные достижения в области геномики и биологии рака позволили значительно расширить объем знаний о молекулярном патогенезе лимфом. С использованием полногеномных методов исследования и современных компьютерных технологий удалось доказать, что разнообразные гистологические и иммуноморфологические подтипы лимфом различаются на молекулярном уровне и возникают в результате действия различных онкогенных механизмов. Стало понятно, что в основе вариабельности клинических симптомов, которые наблюдаются у пациентов с лимфомами, лежат как гетерогенность опухолевых клеток, так и особенности молекулярного патогенеза. Основываясь на полученных данных, предложены стратегии для разработки новых препаратов, которые сегодня используются в лечении лимфом. Они включают определение молекулярных этапов патогенеза, оценку значимости каждого этапа для развития опухоли и получение препарата с направленным действием на этот этап. В результате предложено несколько новых классов молекулярных таргетных агентов для лечения лимфом, которые сегодня изучаются в клинических исследованиях. В современную эпоху персонализированной медицины одной из основных задач при лечении пациентов с лимфомами является определение правильной таргетной терапии для каждого типа лимфоидной опухоли, характеризующейся уникальными молекулярными механизмами опухолеобразования.


Ключевые слова: лимфомы, профиль экспрессии генов, микроРНК, сигнальные пути, NF-kB.

Получено: 13 февраля 2016 г.

Принято в печать: 14 марта 2016 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Intlekofer AM, Younes A. Precision therapy for lymphoma—current state and future directions. Nat Rev Clin Oncol. 2014;11(10):585–96. doi: 10.1038/nrclinonc.2014.137.
  2. Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma—treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11(1):12–23. doi: 10.1038/nrclinonc.2013.197.
  3. Borchmann P, Eichenauer DA, Engert A. State of the art in the treatment of Hodgkin lymphoma. Nat Rev Clin Oncol. 2012;9(8):450–9. doi: 10.1038/nrclinonc.2012.91.
  4. Campo E, Swerdlow SH, Harris NL, et al. The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood. 2011;117(19):5019–32. doi: 10.1182/blood-2011-01-293050.
  5. Немцова М.В., Кушлинский Н.Е. Достижения высокотехнологичных геномных методов для практической онкологии. Медицинский алфавит. 2015;1(2):10–3.
    [Nemtsova MV, Kushlinskii NE. The achievement of high-genomic methods for practical oncology. Meditsinskii alfavit. 2015;1(2):10–3. (In Russ)]
  6. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503–11. doi: 10.1038/35000501.
  7. Lenz G, Dave SS, Xiao W, et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med. 2008;359(22):2313–23. doi: 10.1056/NEJMoa0802885.
  8. Ngo VN, Davis RE, Lamy L, et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature. 2006;441(7089):106–10. doi: 10.1038/nature04687.
  9. Compagno M, Lim WK, Grunn A, et al. Mutations of multiple genes cause deregulation of NF-kappa B in diffuse large B-cell lymphoma. Nature. 2009;459(7247):717–21. doi: 10.1038/nature07968.
  10. Lenz G, Davis RE, Ngo VN, et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science. 2008;319(5870):1676–9. doi: 10.1126/science.1153629.
  11. Wilson WH. The Bruton’s tyrosine kinase (BTK) inhibitor, ibrutinib (PCI-32765), has preferential activity in the ABC subtype of relapsed/refractory de novo diffuse large B-cell lymphoma (DLBCL): interim results of a multicentre, open-label, phase 2 study. Blood. 2012;120: Abstract 686.
  12. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507–16. doi: 10.1056/NEJMoa1306220.
  13. Morin RD, Mendez-Lago M, Mungall AJ, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476(7360):298–303. doi: 10.1038/nature10351.
  14. Chi P, Allis CD, Wang GG. Covalent histone modifications—miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 2010;10(7):457–69. doi: 10.1038/nrc2876.
  15. Pasqualucci L, Dominguez-Sola D, Chiarenza A, et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature. 2011;471(7337):189–95. doi: 10.1038/nature09730.
  16. Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. 2009;27(32):5459–68. doi: 10.1200/jco.2009.22.1291.
  17. Yap DB, Chu J, Berg T, et al. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood. 2011;117(8):2451–9. doi: 10.1182/blood-2010-11-321208.
  18. Beguelin W, Popovic R, Teater M, et al. EZH2 is required for germinal centre formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell. 2013;23(5):677–92. doi: 10.1016/j.ccr.2013.04.011.
  19. Cairns RA, Iqbal J, Lemonnier F, et al. IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma. Blood. 2012;119(8):1901–3. doi: 10.1182/blood-2011-11-391748.
  20. Wang F, Travins J, DeLaBarre B, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 2013;340(6132):622–6. doi: 10.1126/science.1234769.
  21. Pasqualucci L, Khiabanian H, Fangazio M, et al. Genetics of follicular lymphoma transformation. Cell Rep., 2014;6(1):130–40. doi: 10.1016/j.celrep.2013.12.027.
  22. Schmitz R, Young RM, Ceribelli M, et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature. 2012;490(7418):116–20. doi: 10.1038/nature11378.
  23. 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.
  24. Rossi D, Trifonov V, Fangazio M, et al. The coding genome of splenic marginal zone lymphoma: activation of NOTCH2 and other pathways regulating marginal zone development. J Exp Med. 2012;209(9):1537–51. doi: 10.1084/jem.20120904.
  25. Новикова М.В., Рыбко В.А., Хромова Н.В. и др. Роль белков Notch в процессах канцерогенеза. Успехи молекулярной онкологии. 2015;2(3):30–42. doi: 10.17650/2313-805X-2015-2-3-30-42.
    [Novikova MV, Rybko VA, Khromova NV, et al. The role of Notch pathway in carcinogenesis. Advances in molecular oncology. 2015;2(3):30–42. doi: 10.17650/2313-805X-2015-2-3-30-42. (In Russ)]
  26. Zhang J, Grubor V, Love CL, et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc Natl Acad Sci USA. 2013;110(4):1398–403. doi: 10.1073/pnas.1205299110.
  27. Rahal R, Frick M, Romero R, et al. Pharmacological and genomic profiling identifies NF-kappaB-targeted treatment strategies for mantle cell lymphoma. Nat Med. 2014;20(1):87–92. doi: 10.1038/nm.3435.
  28. Anderson K, Lutz C, van Delft FW, et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature. 2011;469(7330):356–61. doi: 10.1038/nature09650.
  29. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–10. doi: 10.1038/nature10738.
  30. Arcaini L, Rossi D. Nuclear Factor-kB Dysregulation In Splenic Marginal Zone Lymphoma: New Therapeutic Opportunities. Haematologica. 2012;97(5):638–40. doi: 10.3324/haematol.2011.058362.

 

Гиперэкспрессия гена WT1 при злокачественных опухолях системы крови: теоретические и клинические аспекты (обзор литературы)

Н.Н. Мамаев, Я.В. Гудожникова, А.В. Горбунова

НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой, ГБОУ ВПО «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; тел.: +(7)911-760-50-86; e-mail: nikmamaev524@gmail.com

Для цитирования: Мамаев Н.Н., Гудожникова Я.В., Горбунова А.В. Гиперэкспрессия гена WT1 при злокачественных опухолях системы крови: теоретические и клинические аспекты (обзор литературы). Клиническая онкогематология. 2016;9(3):257-64.

DOI: 10.21320/2500-2139-2016-9-3-257-264


РЕФЕРАТ

В статье обсуждаются полученные в последние годы данные о феномене гиперэкспрессии гена WT1 у пациентов с острыми лейкозами, миелодиспластическими синдромами, хроническим миелолейкозом, неходжкинскими лимфомами и множественной миеломой. Показана большая перспективность мониторинга уровня экспрессии гена WT1 в посттрансплантационный период и после индукционной химиотерапии. Такой подход может использоваться в диагностике минимальной остаточной болезни и раннем выявлении рецидивов лейкозов, а также для их своевременного и контролируемого лечения. Из других исследовательских направлений представляется перспективным тестирование аутотрансплантата на наличие или отсутствие в нем опухолевых элементов, а также оценка эффективности индукционной терапии у больных из группы повышенного прогностического риска.


Ключевые слова: феномен гиперэкспрессии гена WT1, трансплантация гемопоэтических стволовых клеток, химиотерапия, молекулярный мониторинг лечения.

Получено: 8 февраля 2016 г.

Принято в печать: 30 марта 2016 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Call KM, Glaser T, Ito CI, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor gene locus. Cell. 1990;60(3):509–20. doi: 10.1016/0092-8674(90)90601-a.
  2. Rose EA, Glaser T, Jones C, et al. Complete physical map of the WAGR region of 11p13 localizes a candidate Wilms’ tumor gene. Cell. 1990;60(3):495–508. doi: 10.1016/0092-8674(90)90600-j.
  3. Miwa H, Beran M, Saunders GF. Expression of the Wilms’ tumor gene (WT1) in human leukemias. Leukemia. 1992;6(5):405–9.
  4. Inoue K, Ogawa H, Sonoda Y, et al. Aberrant overexpression of the Wilms’ tumour gene (WT1) in human leukemia. Blood. 1997;88(4):1405–12.
  5. Абдулкадыров К.М., Грицаев С.В., Капустин С.И. и др. Экспрессия гена опухоли Вилмса (WT1) в клетках крови больных миелодиспластическим синдромом. Вопросы онкологии. 2004;50(6):668–71.
    [Abdulkadyrov KM, Gritsaev SV, Kapustin SI, et al. Wilms’ tumor gene (WT1) expression in blood cells of patients with myelodysplastic syndrome. Voprosy oncologii. 2004;50(6):668–71. (In Russ)]
  6. Yang L, Han Y, Suarez Saiz F, et al. A tumor suppressor and oncogene: The WT1 story. Leukemia. 2007;21(5):868–76. doi: 1038/sj.leu.2404624.
  7. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при миелодиспластических синдромах и клиническое значение гиперэкспрессии гена WT1. Клиническая онкогематология. 2014;7(4):551–63.
    [Mamayev NN, Gorbunova AV, Gindina TL, et al. Allogeneic hematopoietic stem cell transplantation in myelodysplastic syndromes and clinical significance of WT1 gene overexpression. Klinicheskaya onkogematologiya. 2014;7(4):551–63. (In Russ)]
  8. Мамаев Н.Н., Горбунова А.В., Бархатов И.М. и др. Молекулярный мониторинг течения острых миелоидных лейкозов по уровню экспрессии гена WT1 после аллогенной трансплантации гемопоэтических столовых клеток. Клиническая онкогематология. 2015;8(3):309–20. doi: 10.21320/2500-2139-2015-8-3-309-320.
    [Mamaev NN, Gorbunova AV, Barkhatov IM, et al. Molecular monitoring of WT1 gene expression level in acute myeloid leukemias after allogeneic hematopoietic stem cell transplantation. Clinical oncohematology. 2015;8(3):309–20. doi: 10.21320/2500-2139-2015-8-3-309-320. (In Russ)]
  9. Israyelyan A, Goldstein L, Tsai W, et al. Real-time assessment of relapse risk based on the WT1 marker in acute leukemia and myelodysplastic syndrome patients after hematopoietic cell transplantation. Bone Marrow Transplant. 2015;50(1):26–33. doi: 10.1038/bmt.2014.209.
  10. Iwasaki T, Sugisaki Ch, Nagata K, et al. Wilms’ tumor 1 message and protein expression in bone marrow failure syndrome and acute leukemia. Pathol Int. 2007;57(10):645–51. doi: 10.1111/j.1440-2007.02153.x.
  11. Tatsumi N, Hojo N, Yamada O, et al. Deficiency in WT1-targeting microRNA-125a leads to myeloid malignancies and urogenital abnormalities. Oncogene. 2015;35(8):1003–14. doi: 10.1038/onc.2015.154.
  12. Inoue K, Sugiyama H, Ogawa H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood. 1994;84(9):3071–9.
  13. Drakos E, Rassidakis GZ, Tsioli F, et al. Differential expression of WT1 gene product in non-Hodgkin lymphomas. Appl Immunohistochem. Mol Morphol. 2005;13(2):132–7. doi: 10.1097/01.pai.0000143786.62974.66.
  14. Hatta Y, Takeuchi J, Saitoh T, et al. WT1 expression level and clinical factors in multiple myeloma. J Exp Clin Cancer Res. 2005;24(4):595–9.
  15. Na I-K, Kreuzer K-A, Lupberger J, et al. Quantitative RT-PCR of Wilms tumor gene transcripts(WT1) for the molecular monitoring of patients with accelerated phase bcr/abl + CML. Leuk Res. 2005;29(3):343–5. doi: 10.1016/j.leukres.2004.08.003.
  16. Chiusa L, Francia di Celle P, Campisi P, et al. Prognostic value of quantitative analysis of WT1 gene transcripts in adult acute lymphoblastic leukemia. Haematologica. 2006;91(2):270–1. doi: 10.0000/www.haematologica.org/content/91/2/270.short.
  17. Radich JP, Dai H, Mao M, et al. Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci USA. 2006;103(8):2794–7. doi: 10.1073/pnas.0510423103.
  18. Cao X, Gu WY, Chen ZX, et al. Bone marrow WT1 gene expression and clinical significance in chronic myelogenous leukemia. Zhonghua Nei Ke Za Zhi. 2007;46(4):277–9.
  19. Otahalova E, Ullmannova-Benson V, Klamova FI, et al. WT1 expression in peripheral leukocytes of patients with chronic myeloid leukemia serves for the prediction of imatinib resistance. Neoplasma. 2009;56(5):393–7. doi: 10.4149/neo_2009_05_393.
  20. Heesch S, Goekbuget N, Stroux A, et al. Prognostic implications and expression of the Wilms tumor 1 (WT1) gene in adult T-lymphoblastic leukemia. 2010;95(6):942–9. doi: 10.3324/haematol.2009.016386.
  21. Аксенова Е.В. Стандартизированное исследование экспрессии генов BCR-ABL, PRAME и WT1 у больных хроническим миелолейкозом: Диc. ¼ канд. мед. наук. М., 2011. 138 с.
    [Aksyenova EV. Standartizirovannoe issledovanie expressii genov BCR-ABL, PRAME I WT1 u bolnykh chronicheskim myeloleukosom. (Standardized evaluation of the BCR-ABL, PRAME and WT1 gene expression in patients with chronic myeloid leukemia.) [dissertation] Moscow; 2011. 138 p. (In Russ)]
  22. Гапонова Т.В. Экспрессия опухолеассоциированных генов PRAME, WT1 и XIAP у больных множественной миеломой в процессе интенсивной терапии и аутотрансплантации: Диc. ¼ канд. мед. наук. М., 2011. 141 с.
    [Gaponova TV. Expressia opucholeassocirovannykh genov PRAME, WT1 i XIAP u bolnykh mnozhestvennoi myelomoi v processe intensivnoi therapii I autotransplantacii. (Tumor-associated PRAME, WT1 and XIAP gene expression in patients with multiple myeloma during intensive therapy and autografting.) [dissertation] Moscow; 2011. 141 p. (In Russ)]
  23. Tyler EM, Jungbluth AA, O’Reilly RJ, Koehne G. WT1-specific responses in high-risk multiple myeloma patients undergoing allogeneic T-cell-depleted hematopoietic stem cell transplantation and donor lymphocyte infusions. 2012;121(2):308–17. doi: 10.1182/blood-2012-06-435040.
  24. Ujj Z, Buglyo G, Udvardy M, et al. WT1 overexpression affecting clinical outcome in non-Hodgkin lymphomas and adult acute lymphoblastic leukemia. Pathol Oncol Res. 2013;20(3):565–70. doi: 10.1007/s12253-013-9729-
  25. Inoue K, Ogawa H, Yamagami T, et al. Long–term follow-up of minimal residual disease in leukemia patients by monitoring WT1 (Wilms tumor gene) expression levels. Blood. 1996;88(6):2267–78.
  26. Kletzel N, Olzewski M, Huang W, et al. Utility of WT1 as a reliable tool for the detection of minimal disease in children with leukemia. Pediatr Dev Pathol. 2002;5(3):269–75. doi: 10.1007/s10024-001-0208-x.
  27. Cilloni D, Gottardi E, De Micheli D, et al. Quantitative assessment of WT1 expression by real time quantitative PCR may be a useful tool for monitoring residual disease in acute leukemia patients. 2002;16(10):2115–21. doi: 10.1038/sj.leu.2402675.
  28. Cilloni D, Giuseppe S, Gottardi E, et al. WT1 as a universal marker for minimal residual disease detection and quantification in myeloid leukemias and in myelodysplastic syndrome. Acta Hematol. 2004;112(1–2):79–84. doi: 10.1159/000077562.
  29. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: A European LeukemiaNet study. J Clin Oncol. 2009;27(31):5195–201. doi: 10.1200/jco.2009.22.4865.
  30. Weisser M, Kern W, Rauhut S, et al. Prognostic impact of RTPCR-based quantification of WT1 gene expression during MRD monitoring of acute myeloid leukemia. 2005;19(8):1416–23. doi: 10.1038/sj.leu.2403809.
  31. Candoni A, Toffoletti E, Gallina R, et al. Monitoring of minimal residual disease by quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem cell transplantation in acute myeloid leukemia. Clin Transplant. 2011;25(2):308–16. doi: 10.1111/j.1399-0012.2010.01251.x.
  32. Gray JX, McMillen L, Mollee P, et al. WT1 expression as a marker of minimal residual disease predicts outcome in acute myeloid leukemia when measured post-transplantation. Leuk Res. 2012;36(4):453–8. doi: 10.1016/j.leukres.2011.09.005.
  33. Kwon M, Martinez-Laperche C, Infante M, et al. Evaluation of minimal residual disease by real-time quantitative PCR of Wilms’ Tumor 1 expression in patients with acute myelogenous leukemia after allogeneic stem cell trans-plantation: Correlation with flow cytometry and chimerism. Biol Blood Marrow Transplant. 2012;18(8):1235–42. doi: 10.1016/j.bbmt.2012.01.012.
  34. Polak J, Hajkova H, Haskovec C, et al. Quantitative monitoring of WT1 expression in peripheral blood before and after allogeneic stem cell transplantation for acute myeloid leukemia – a useful tool for early detection of minimal residual disease. 2013;60(1):74–82. doi: 10.4149/neo_2013_011.
  35. Cilloni D, Messa F, Arruga F, et al. Early prediction of treatment outcome in acute myeloid leukemia by measurement of WT1 transcript levels in peripheral blood samples collected after chemotherapy. Haematologica. 2008;93(6):921–4. doi: 10.3324/haematol.12165.
  36. Andersson C, Li X, Lorenz F, et al. Reduction in WT1 gene expression during early treatment predicts the outcome in patients with acute myeloid leukemia. Diagn Mol Pathol. 2012;21(4):225–33. doi: 10.1097/pdm.0b013e318257ddb9.
  37. Mossallam GI, Hamid TM, Mahmoud HK, et al. Prognostic significance of WT1 expression at diagnosis and end of induction in Egyptian adult acute myeloid leukemia patients. Hematology. 2013;18(2):69–73. doi: 10.1179/1607845412Y.0000000048.
  38. Ujj Z, Buglyo G, Udvardy M, et al. WT1 expression in adult acute myeloid leukemia: Assessing its presence, magnitude and temporal changes as prognostic factors. Pathol Oncol Res. 2015;22(1):217–21. doi: 10.1007/s12253-015-0002-
  39. Rein LAM, Chao NJ. WT1 vaccination in acute myeloid leukemia: new methods of implementing adoptive immunotherapy. Expert Opin Invest Drugs. 2014;23(3):417–26. doi: 10.1517/13543784.2014.889114.
  40. Paschka P, Marcucci G, Ruppert A.S, et al. Wilms’ tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol. 2008;26(28):4595–602. doi: 10.1200/jco.2007.15.2058.
  41. Sugiyama H. WT1 (Wilms’ tumor gene 1): biology and cancer immunotherapy. Jpn J Clin Oncol. 2010;40(5):377–87. doi: 10.1093/jjco/hyp194.
  42. Vidovic K, Svensson T, Nilsson B, et al. Wilms’ tumor gene 1 protein represses the expression of the tumor suppressor interferon regulatory factor 8 in human hematopoietic progenitors and in leukemic cells. Leukemia. 2010;24(5):9982–1000. doi: 10.1038/leu.2010.33.
  43. Essafi A, Webb A, Berry RL, et al. A WT1-controlled chromatin switching mechanism underpins tissue-specific wnt4 activation and repression. Dev Cell. 2011;21(3):559–74. doi: 10.1016/j.devcel.2011.07.014.
  44. Huff V. Wilms’ tumours: about tumour suppressor genes, an oncogene and chameleon gene. Nat Rev Cancer. 2001;11(2):111–21. doi: 10.1038/nrc3002.
  45. Morrison AA, Viney RL, Landomery MR. The post-transcriptional roles of WT1, a multifunctional zinc-finger protein. Biochim Biophys Acta. 2008;1785(1):55–62. doi: 10.1016/j.bbcan.2007.10.002.
  46. Owen C, Fitzgibbon J, Paschka P. The clinical relevance of Wilms Tumour 1 (WT1) gene mutations in acute leukemias. Hematol Onc 2010;28(1):13–9. doi: 10.1002/hon.931.
  47. Haber DA, Sohn RL, Buckler AJ, et al. Alternative splicing and genomic structure of the Wilms tumor gene WT1. Proc Natl Acad Sci USA. 1991;88(21):9618–22. doi: 10.1073/pnas.88.21.9618.
  48. Keilholz U, Menssen HD, Gaiger A, et al. Wilms’ tumor gene 1(WT1) in human neoplasia. 2005;19(8):1318–23. doi: 10.1038/sj.leu.2403817.
  49. Hosen N, Shirakata T, Nishida S, et al. The Wilms’ tumor gene WT1-GFP knock-in mouse reveals the dynamic regulation of WT1 expression in normal and leukemic hematopoiesis. Leukemia. 2007;21(8):1783–91. doi: 10.1038/sj.leu.2404752.
  50. Miller-Hodges E, Hohenstein P. WT1 in disease: shifting the epithelial-mesenchymal balance. J Pathol. 2012;226(2):229–40. doi: 10.1002/path.2977.
  51. Cunningham TJ, Palumbo I, Grosso M, et al. WT1 regulates murine hematopoiesis via maintenance of VEGF isoform ratio. Blood. 2013;122(2):188–92. doi: 10.1182/blood-2012-11-466086.
  52. Patmasirivat P, Fraizer G, Kantarjian H, Saunders GF. WT1 and GATA1 expression in myelodysplastic syndrome and acute leukemia. Leukemia. 1999;13(6):891–900. doi: 10.1038/sj.leu.2401414.
  53. Gaiger A, Linnerth B, Mann G, et al. Wilms’ tumour gene (wt1) expression at diagnosis has no prognostic relevance in childhood acute lymphoblastic leukemia treated by an intensive chemotherapy protocol. Eur J Haematol. 2009;63(2):86–93. doi: 10.1111/j.1600-0609.1999.tb01121.x.
  54. Arlyaratana S, Loeb DM. The role of the Wilms tumour gene (WT1) in normal and malignant hematopoiesis. Expert Rev Mol Med. 2007;9(14):1–17. doi: 10.1017/s1462399407000336.
  55. Ellisen LW, Carlesso N, Cheng T, et al. The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells. EMBO J. 2001;20(8):1897–909. doi: 10.1093/emboj/20.8.1897.
  56. Scharnhorst V, van den Eb AJ, Jochemsen AG. WT1 proteins: functions in growth and differentiation. Gene. 2001;273(2):141–61. doi: 10.1016/s0378-1119(01)00593-5.
  57. Baird PN, Simmons PJ. Expression of the Wllms’ tumor gene (WT1) in normal hematopoiesis. Eur Haematol. 1997;25(4):312–20.
  58. Lange T, Hubmann M, Burkhard R, et al. Monitoring of WT1 expression in PB and CD34+ donor chimerism of BM predicts early relapse in AML and MDS patients after hematopoietic cell transplantation with reduced-intensity conditioning. 2011;25(3):498–505. doi: 10.1038/leu.2010.283.
  59. Schmid D, Heinze G, Linnert B, et al. Prognostic significance of WT1 gene expression at diagnosis in adult de novo acute myeloid leukemia. Leukemia. 1997;11(5):639–43. doi: 10.1038/sj.leu.2400620.
  60. Lyu X, Xin Y, Mi R, et al. Overexpression of Wilms’ Tumor 1 gene as a negative prognostic indicator in acute myeloid leukemia. PLoS One. 2014;9(3):e92470. doi: 10.1371/journal.pone.0092470.
  61. Wochlecke C, Wittig S, Arndt C, Gruhn B. Prognostic impact of WT1 expression prior to hematopoietic stem cell transplantation in children with malignant hematological diseases. J Cancer Res Clin. Oncol. 2014;141(3):523–9. doi: 10.1007/s00432-014-1832-y.
  62. Zhao X-S, Jin S, Zhu H-H, et al. Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2011;47(4):499–507. doi: 10.1038/bmt.2011.121.
  63. Nomdedeu JF, Hoyos M, Carricondo M, et al. Bone marrow WT1 levels at diagnosis, post-induction and post-intensification in adult de novo AML. Leukemia. 2013;27(11):2157–64. doi: 10.1038/leu.2013.111.
  64. Alonso-Domingues JM, Tenorio M, Velasco D, et al. Correlation of WT1 expression with the burden of total and residual leukemic blasts in bone marrow samples of acute myeloid leukemia patients. Cancer Genet. 2012;205(4):190–1. doi: 10.1016/j.cancergen.2012.02.008.
  65. Tamaki H, Ogawa H, Inoue K, et al. Increased expression of the Wilms tumor gene (WT1) at relapse in acute leukemia. Blood. 1996;88(11):4396–8.
  66. Frairia C, Aydin S, Riera L, et al. WT1 expression in аcute myeloid leukaemia: a useful marker for improving therapy response evaluation. 2013;122(21):2588 (abstract).
  67. Willasch AM, Gruhn B, Coliva T, et al. Standartization of WT1 mRNA quantitation for minimal residual disease monitoring in childhood AML and implications of WT1 gene mutations: a European multicenter study. 2009;23(8):1472–9. doi: 10.1038/leu.2009.51.
  68. Lapillonne H, Renneville A, Auvrignon A, et al. High WT1 expression after induction therapy predicts high risk of relapse and death in pediatric acute myeloid leukemia. J Clin Oncol. 2006;24(10):1507–15. doi: 10.1200/jco.2005.03.5303.
  69. Liu J, Wang Yu, Xu L-P, et al. Monitoring mixed lineage leukemia expression may help identify patients with mixed lineage leukemia-rearranged acute leukemia who are at high risk of relapse after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2014;20(7):929–36. doi: 10.1016/j.bbmt.2014.03.008.
  70. Ogawa H, Tamaki H, Ikegame K, et al. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogeneic stem cell transplantation in acute type leukemia. Blood. 2003;101(5):1698–704. doi: 10.1182/blood-2002-06-
  71. Yoon JH, Kim HJ, Shin SH, et al. BAALC and WT1 expression from diagnosis to hematopoietic stem cell transplantation: consecutive monitoring in adult patients with core-binding-factor-positive AML. Eur J Haematol. 2013;91(2):112–21. doi: 10.1111/ejh.12142.
  72. Yoon JH, Kim H-J, Kim J-W, et al. Identification of molecular and cytogenetic risk factors for unfavorable core-binding factor-positive adult AML with post-remission treatment outcome analysis including transplantation. Bone Marrow Transplant. 2014;49(12):1466–74. doi: 10.1038/bmt.2014.180.
  73. Miyagi T, Ahuja H, Kudota T, et al. Expression of the candidate Wilms’ tumor gene, WT1, in human leukemia cells. Leukemia. 1993;7(7):970–7.
  74. Miyawaki S, Hatsumi N, Tamaki T, et al. Prognostic potential of detection of WT1 mRNA level in peripheral blood in adult acute myeloid leukemia. Leuk Lymphoma. 2010;51(10):1855–61. doi: 10.3109/10428194.2010.507829.
  75. Little M, Wells C. A clinical overview of WT1 gene mutations. Hum Mutat. 1997;9(3):209–25. doi: 10.1002/(sici)1098-1004(1997)9:3<209::aid-humu2>3.0.co;2-2.
  76. Mori N, Okada M, Motoji T, et al. Mutation of the WT1 gene in myelodysplastic syndrome and acute myeloid leukemia post myelodysplastic syndrome. Br J Haematol. 1999;105(3):844–5. doi: 10.1046/j.1365-1999.01497.x.
  77. Damm F, Heuser M, Morgan M, et al. Single nucleotide polymorphism in the mutational hotspot of WT1 predicts a favorable outcome in patients with cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2010;28(4):578–85. doi: 10.1200/jco.2009.23.0342.
  78. Hou HA, Huang TC, Lin LI, et al. WT1 mutation in 470 adult patients with acute myeloid leukemia: stability during disease evolution and implication of its incorporation into a survival scoring system. Blood. 2010;115(25):5222–31. doi: 10.1016/s1040-1741(10)79528-
  79. Shen Y, Zhu Y-M, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood. 2011;118(20):5593–603. doi: 10.1182/blood-2011-03-
  80. Luo S, Yu K, Yan QX, et al. Analysis of WT1 mutations, expression levels and single nucleotide polymorphism rs16754 in de novo non-M3 acute myeloid leukemia. Leuk Lymphoma. 2014;56(2):349–57. doi: 10.3109/10428194.2013.791985.
  81. Park SH, Lee HJ, Kim I-S, et al. Incidences and prognostic impact of c-KIT, WT1, CEBPA, and CBL mutations, and mutations associated with epigenetic modification in core binding factor acute myeloid leukemia: a multicenter study in Korean population. Ann Lab Med. 2015;35(3):288–97. doi: 10.3343/alm.2015.35.3.288.
  82. Rampal R, Alkalin A, Madzo J, et al. DNA hydroxymethylation profiling reveals that WT1 mutations result in loss of TET2 function in acute myeloid leukemia. Cell Rep. 2014;9(5):1841–55. doi: 10.1016/j.celrep.2014.11.004.
  83. Zhang Q, Zhang Q, Li Q. Monitoring of WT1 and its target gene IRF8 expression in acute myeloid leukemia and their significance. Int J Lab Hematol. 2015;37(4):e67–71. doi: 10.1111/ijlh.12309.
  84. Brieger J, Weidmann E, Fenchel K, et al. The expression of the Wilms’ tumor gene in acute myelocytic leukemias as a possible marker for leukemic blast cells. Leukemia. 1994;8(12):2138.
  85. Brieger J, Weidmann E, Maurer U, et al. The Wilms’ tumor gene is frequently expressed in acute myeloblastic leukemia and may provide a marker for residual blast cells detectable by PCR. Ann Oncol. 1995;6(8):811–66.
  86. Bergmann L, Miething C, Maurer U, et al. High levels of Wilms’ tumor gene (wt1) mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood. 1997;90(3):1217–25.
  87. Ogawa H, Ikegame K, Kawakami M, Tamaki H. WT1 gene transcript assay for relapse in acute leukemia after transplantation. Leuk Lymphoma. 2004;45(9):1747–53. doi: 10.1080/10428190410001687503.
  88. Rodrigues PC, Oliveira SN, Vaina MB, et al. Prognostic significance of WT1 gene expression in pediatric acute myeloid leukemia. Pediatr Blood Cancer. 2007;49(2):133–8. doi: 10.1002/pbc.20953.
  89. Miglino M, Colombo N, Pica C, et al. Wt1 overexpression at diagnosis may predict favorable outcome in patients with de novo non-M3 acute myeloid leukemia. Leuk Lymphoma. 2011;52(10):1961–9. doi: 10.3109/10428194.2011.585673.
  90. Zhao BR, Tang XW, Cen JN, et al. Correlation between clinical outcome and WT1 detection after hematopoietic stem cell transplantation in acute leukemia. Zhonghua Yi Xue Za Zhi. 2011;91(20):1375–8.
  91. Gaiger A, Schmid D, Heinze G, et al. Detection of the WT1 transcript by RT-PCR in complete remission has no prognostic relevance in de novo acute myeloid leukemia. Leukemia. 1998;12(12):1886–94. doi: 10.1038/sj.leu.2401213.
  92. Barragan E, Cervera J, Bolufer P, et al. Prognostic implications of Wilms’ tumor gene (WT1) expression in patients with de novo acute myeloid leukemia. Haematologica. 2004;89(8):926–33.
  93. Yi-ning Y, Xiao-rui W, Chu-xian Z, et al. Prognostic significance of diagnosed WT1 level in acute myeloid leukemia: a meta-analyse. Ann Hematol. 2015;94(6):929–38. doi: 10.1007/s00277-014-2295-
  94. Nowakowska-Kopera A, Sacha T, Florek I, et al. Wilms’ tumor gene 1 expression analysis by real-time quantitative polymerase chain reaction for monitoring of minimal residual disease in acute leukemia. Leuk Lymphoma. 2009;50(8):1326–32. doi: 10.1080/10428190903050021.
  95. Guillaumet-Adkins A, Richter J, Odera MD, et al. Hypermethylation of the alternative AWT1 promotor in hematological malignancies is a highly specific marker for acute myeloid leukemias despite high expression levels. J Hematol Oncol. 2014;7(1):4. doi: 10.1186/1756-8722-7-4.
  96. Capelli D, Attolico I, Saraceli F, et al Early cumulative incidence of relapse in 80 acute myeloid leukemia patients after chemotherapy and transplant post-consolidation treatment: prognostic role of post-induction WT1. 40th EBMT Meeting; 2014 30 March – 2 April; Milan, Italy; 2014: Abstract P287.
  97. Messina C, Candoni A, Carraba MG, et al. Wilms’ tumor gene 1 transcript levels in leukopheresis on peripheral blood hematopoietic cells predict relapse risk in patients autografted for acute myeloid leukemia. Biol Blood Marrow Transpl. 2014;20(10):1586–91. doi: 10.1016/j.bbmt.2014.06.017.
  98. Messina C, Sala E, Carrabba M, et al. Early post-allogeneic transplantation WT1 transcript positivity predicts AML relapse. 40th EBMT Meeting; 2014 30 March – 2 April; Milan, Italy; 2014: Abstract P239.
  99. Gianfaldoni G, Mannelli F, Ponziani V, et al. Early reduction of WT1 transcripts during induction chemotherapy predicts for longer disease free and overall survival in acute myeloid leukemia. Haematologica. 2010;95(5):833–6. doi: 10.3324/haematol.2009.011908.
  100. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Трансплантация гемопоэтических стволовых клеток при остром миелоидном лейкозе с транслокацией t(8;21)(q22;q22). Клиническая онкогематология. 2013;6(4):439–44.
    [Mamayev NN, Gorbunova AV, Gindina TL, et al. Hemopoietic stem cell transplantation in AML patients with t(8;21)(q22;q22) translocation. Klinicheskaya onkogematologiya. 2013;6(4):439–44. (In Russ)]
  101. 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 post-transplantation 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 onkogematologiya. 2014;7(1):71–5. (In Russ)]
  102. Barragan E, Pajuelo JC, Ballester S, et al. Minimal residual disease detection in acute myeloid leukemia by mutant nucleophosmin (NPM1): comparison with WT1 gene expression. Clin Chim Acta. 2008;395(1–2):120–3. doi: 10.1016/j.cca.2008.05.021.
  103. Ostergaard M, Olesen LH, Hasle H, et al. WT1 gene expression: an excellent tool for monitoring minimal residual disease in 70% of acute myeloid leukemia patients – results from a single-centre study. Br J Haematol. 2004;125(5):590–600. doi: 10.1111/j.1365-2004.04952.x.
  104. Zhao XS, Yan CH, Liu DH, et al. Combined use of WT1 and flow cytometry monitoring can promote sensitivity of predicting relapse after allogeneic HSCT without affecting specificity. Ann Hematol. 2013;92(8):1111–9. doi: 10.1007/s00277-013-1733-
  105. Candoni A, Tiribelli M, Toffoletti E, et al. Quantitative assessment of WT1 gene expression after allogeneic stem cell transplantation is a useful tool for monitoring minimal residual disease in acute myeloid leukemia. Eur J Haematol. 2009;82(1):61–8. doi: 10.1111/j.1600-2008.01158.x.
  106. Ommen HB, Nyvold CG, Braendstrup K, et al. Relapse prediction in acute myeloid leukemia patients in complete remission using WT1 as a molecular marker: development of a mathematical model to predict time from molecular to clinical relapse and define optimal sampling intervals. Br J Haematol. 2008;141(6):782–991. doi: 10.1111/j.1365-2008.07132.x.
  107. Yamauchi T, Negoro E, Lee S, et al. Detectable Wilms’ tumor-1 transcription at treatment completion is associated with poor prognosis of acute myeloid leukemia: a single institution’s experience. Anticancer Res. 2013;33(8):3335–40.
  108. Woehlecke C, Wittig S, Sanft J, et al. Detection of relapse after hematopoietic stem cell transplantation in childhood by monitoring of WT1 expression and chimerism. J Cancer Res Clin Oncol. 2015;141(7):1283–90. doi: 10.1007/s00432-015-1919-
  109. Jin S, Liu DH, Xu LP, et al. The significance of dynamic detection of WT1 expression on patients of hematologic malignancy following allogeneic hematopoietic stem cell transplantation. Zhonghua Nei Ke Za Zhi. 2008;47(7):578–81.
  110. Rossi G, Minervini MM, Carella AM, et al. Comparison between multiparameter flow cytometry and WT1-RNA quantification in monitoring minimal residual disease in acute myeloid leukemia without specific molecular targets. Leuk Res. 2012;36(4):401–6. doi: 10.1016/j.leukres.2011.11.020.
  111. Zhao Q, Zhao Q, Li Q, et al. Monitoring of WT1 and its target gene IRF8 expression in acute myeloid leukemia and their significance. Int J Lab Hematol. 2014;37(4):e67–71. doi: 10.1111/ijlh.12309.
  112. Mear J-B, Salaun V, Dina N, et al. WT1 and flow cytometry minimal residual disease follow-up after allogeneic transplantation in practice. 40th EBMT Meeting; 2014 30 March – 2 April; Milan, Italy; 2014: Abstract P655.
  113. Tamaki H, Ogawa H, Ohyashiki K, et al. The Wilms’ tumor gene WT1 is a good marker for diagnosis of disease progression of myelodysplastic syndromes. 1999;13(3):393–9. doi: 10.1038/sj.leu.2401341.
  114. Cilloni D, Gottardi E, Messa F, et al. Significant correlation between the degree of WT1 expression and the International Scoring System score in patients with myelodysplastic syndromes. J Clin Oncol. 2003;21(10):1988–95. doi: 10.1200/jco.2003.10.503.
  115. Bader P, Niemeyer C, Weber G, et al. WT1 gene expression: useful marker for minimal residual disease in childhood myelodysplastic syndromes and juvenile myelomonocytic leukemia. Eur J Haematol. 2004;73(1):25–8. doi: 10.1111/j.1600-2004.00260.x.
  116. Tamura H, Dan K, Yokose N, et al. Prognostic significance of WT1 mRNA and antiWT1 antibody levels in peripheral blood in patients with myelodysplastic syndromes. Leuk Res. 2010;34(8):986–90. doi: 10.1016/j.leukres.2009.11.029.
  117. Yamauchi T, Matsuda Y, Takai M, et al. Wilms’ tumor-1 transcript in peripheral blood helps diagnose acute myeloid leukemia and myelodysplastic syndrome in patients with pancytopenia. Anticancer Res. 2012;32(10):4479–83.
  118. Qin Y-Z, Zhu H-H, Liu Y-R, et al. PRAME and WT1 transcripts constitute a good molecular marker combination for monitoring minimal residual disease in myelodysplastic syndromes. Leuk Lymphoma. 2013;54(7):1442–9. doi: 10.3109/10428194.2012.743656.
  119. Ueda Y, Mizutani C, Nannya Y, et al. Clinical evaluation of WT1 mRNA expression levels in peripheral blood and bone marrow in patients with myelodysplastic syndromes. Leuk Lymphoma. 2013;54(7):1450–18. doi: 10.3109/10428194.2012.745074.
  120. Minetto P, Guolo F, Clavio M, et al. Combined assessment of WT1 and BAALC gene expression at diagnosis may improve leukemia-free survival prediction in patients with myelodysplastic syndrome. Leuk Res. 2015;39(8):866–73. doi: 10.1016/j.leukres.2015.04.011.
  121. Santamaria C, Ramos F, Puig N, et al. Simultaneous analysis of the expression of 14 genes with individual prognostic value in myelodysplastic syndrome patients at diagnosis: WT1 detection in peripheral blood adversely affects survival. Ann Hematol. 2012;91(12):1887–95. doi: 10.1007/s00277-012-1538-
  122. Menssen HD, Renkl HJ, Rodeck U, et al. Presence of Wilms’ tumor gene wt1 transcripts and the WT1 nuclear protein in the majority of human acute leukemias. Leukemia. 1995;9(6):1060–7.
  123. He YZ, Liang Z, Wu MR, et al. Overexpression of EPS8 is associated with poor prognosis in patients with acute lymphoblastic leukemia. Leuk Res. 2015;39(6):575–81. doi: 10.1016/j.leukres.2015.03.007.
  124. Xu B, Song S, Yip NC, et al. Simultaneous detection of MDR and WT1 gene expression to predict the prognosis of adult acute lymphoblastic leukemia. Hematology. 2010;15(2):74–80 doi: 10.1179/ 102453310X12583347009937.
  125. Azuma T, Otsuki T, Kuzushima K, et al. Myeloma cells are highly sensitive to the granule exocytosis pathway mediated by WT1-specific cytotoxic T lymphocytes. Clin Cancer Res. 2004;10(21):7402–12. doi: 10.1158/1078-ccr-04-0825.
  126. Hamalainen MM, Kairisto V, Junonen V, et al. Wilms tumour gene 1 overexpression in bone marrow as a marker for minimal residual disease in acute myeloid leukemia. Eur J Haematol. 2008;80(3):201–7. doi: 10.1111/j.1600-2007.01009.x.
  127. Wartheim GB, Bagg A. Minimal residual disease testing to predict relapse following transplant for AML and high-grade myelodysplastic syndromes. Expert Rev Mol Drug. 2011;11(4):361–6. doi: 10.1586/erm.11.19.
  128. Lambert J, Lambert J, Niboured O, et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget. 2014;5(15):6280–8. doi: 10.18632/oncotarget.2196.
  129. Steinbach D, Bader P, Willasch A, et al. Prospective validation of a new method of monitoring minimal residual disease in childhood acute myeloid leukemia. Clin Cancer Res. 2014;21(6):1353–9. doi: 10.1158/1078-ccr-14-1999.
  130. Gray JX, McMillen L, Mollee P, et al. WT1 expression as a marker of minimal residual disease predicts outcome in acute myeloid leukemia when measured post-consolidation. Leuk Res. 2012;36(4):453–8. doi: 10.1016/j.leukres.2011.09.005.
  131. Noronha SA, Farrar JE, Alonzo TA, et al. WT1 expression at diagnosis does not predict survival in pediatric AML: a report from the children’s oncology group. Pediatr Blood Cancer. 2009;53(6):1136–9. doi: 10.1002/pbc.22142.
  132. Kim HJ, Choi EJ, Sohn HJ, et al. Combinatorial molecular marker assays of WT1, survivin, and TERT at initial diagnosis of adult acute myeloid leukemia. Eur J Haematol. 2013;91(5):411–22. doi: 10.1111/ejh.12167.
  133. Niavarani A, Currie E, Reyal Y, et al. APOBEC3A is implicated in a novel class of G-to-A mRNA editing in WT1 transcripts. PloS One. 2015;10(3):e0120089. doi: 10.1371/journal.pone.0120089.
  134. Taira C, Matsuda K, Kamijyo Y, et al. Quantitative monitoring of single nucleotide mutations by allele-specific quantitative PCR can be used for the assessment of minimal residual disease in patients with hematological malignancies throughout their clinical course. Clin Chim Acta. 2011;412(1–2):53–8. doi: 10.1016/j.cca.2010.09.011.
  135. Morita Y, Heike1 Y, Kawakami M, et al. Monitoring of WT1-specific cytotoxic T lymphocytes after allogeneic hematopoietic stem cell transplantation. Int J Cancer. 2006;119(6):1360–7. doi: 10.1002/ijc.21960.
  136. Tsuboi A, Oka Y, Nakajima H, et al. Wilms tumor gene WT1 peptide-based immunotherapy induced a minimal response in a patient with advanced therapy-resistant multiple myeloma. Int J Hematol. 2007;86(5):414–7. doi: 10.1007/bf02983998.
  137. Narita M, Masuko M, Kurasaki T, et al. WT1 peptide vaccination in combination with imatinib for a patient with CML in the chronic phase. Int J Med Sci. 2010;7(2):72–81. doi: 10.7150/ijms.7.72.

Мутации генов при острых миелоидных лейкозах

О.В. Блау

Клиника Шарите, Берлинский медицинский университет, ул. Хинденбургдамм, д. 30, Берлин, Германия, 12200

Для переписки: Ольга Владимировна Блау, д-р мед. наук, Department of Hematology, Oncology and Tumorimmunology, Charite University School of Medicine, Hindenburgdamm 30, 12200, Berlin, Germany; e-mail: olga.blau@charite.de.

Для цитирования: Блау О.В. Мутации генов при острых миелоидных лейкозах. Клиническая онкогематология. 2016;9(3):245-56.

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


РЕФЕРАТ

Острый миелобластный лейкоз (ОМЛ) — клональное злокачественное заболевание, характеризующееся неэффективным гемопоэзом. Большинство больных ОМЛ имеют различные цитогенетические и молекулярно-генетические повреждения, которые сочетаются с определенными биологическими и клиническими особенностями заболевания. Примерно у 50–60 % больных de novo и у 80–95 % больных вторичными ОМЛ обнаруживаются хромосомные изменения. Следует отметить, что структурные цитогенетические аберрации являются наиболее частыми маркерами и встречаются примерно в 40 % случаев ОМЛ de novo. Достаточно большая группа больных с нормальным кариотипом (НК-ОМЛ), формально относящаяся к категории промежуточного риска, является крайне гетерогенной в отношении прогноза течения заболевания. В действующие прогностические классификации ОМЛ включены сегодня только некоторые мутации, характеризующиеся известным прогностическим значением, в частности NPM1, FLT3 и C/EBPa. Пациенты с NPM1, но без мутаций FLT3-ITD или с мутациями C/EBPa характеризуются благоприятным прогнозом заболевания, а с мутацией FLT3-ITD — неблагоприятным. Недавно выявлен новый класс мутаций, при которых повреждаются гены, ответственные за эпигенетические процессы регуляции генома, в частности метилирование ДНК или модификацию гистонов. Среди них наиболее изученными к настоящему времени являются мутации в генах DNMT3A, IDH1/2, TET2 и некоторых других. В целом ряде исследований показан неблагоприятный прогностический эффект мутации DNMT3A при ОМЛ. Что касается прогностического значения IDH1/2, то данный вопрос еще не до конца ясен. На прогноз заболевания влияет ряд биологических факторов, в т. ч. сочетание с цитогенетическими аберрациями и другими мутациями, особенно FLT3 и NPM1. Число исследований, посвященных генетическим мутациям при ОМЛ, постоянно растет. Накопленные к настоящему времени знания о генетических изменениях при ОМЛ подтверждают их роль в возникновении и развитии заболевания.


Ключевые слова: острый миелобластный лейкоз, ОМЛ, кариотип, цитогенетические аберрации, мутации генов, прогноз.

Получено: 23 января 2016 г.

Принято в печать: 4 апреля 2016 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Renneville A, Roumier С., Biggio V, et al. Cooperating gene mutations in acute myeloid leukemia: a review of the literature. Leukemia. 2008;22(5):915–31. doi: 10.1038/leu.2008.19.
  2. Knudson AG. Mutation and Cancer: Statistical Study of Retinoblastoma. Proc Natl Acad Sci USA. 1971;68(4):820–3. doi: 10.1073/pnas.68.4.820.
  3. Tucker T, Friedman JM. Pathogenesis of hereditary tumors: beyond the “two-hit” hypothesis. Clin Genet. 2002;62(5):345–57. doi: 10.1034/j.1399-0004.2002.620501.x.
  4. Park S, Koh Y, Yoon SS. Effects of Somatic Mutations Are Associated with SNP in the Progression of Individual Acute Myeloid Leukemia Patient: The Two-Hit Theory Explains Inherited Predisposition to Pathogenesis. Genom Inform. 2013;11(1):34–7. doi: 10.5808/gi.2013.11.1.34.
  5. Genovese G, Kahler AK, Handsaker RE, et al. Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N Engl J Med. 2014;371(26):2477–87. doi: 10.1056/nejmoa1409405.
  6. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11. doi: 10.1038/35102167.
  7. Grimwade D. The changing paradigm of prognostic factors in acute myeloid leukaemia. Best Pract Res Clin Haematol. 2012;25(4):419–25. 10.1016/j.beha.2012.10.004.
  8. Patel JP, Gonen M, Figueroa ME, et al. Prognostic Relevance of Integrated Genetic Profiling in Acute Myeloid Leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/nejmoa1112304.
  9. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.
  10. Frehlick LJ, Eirin-Lopez JM, Ausio J. New insights into the nucleophosmin/nucleoplasmin family of nuclear chaperones. Bioessays. 2007;29(1):49–59. doi: 10.1002/bies.20512.
  11. Kurki S, Peltonen K, Latonen L, et al. Nucleolar protein NPM interacts with HDM2 and protects tumor suppressor protein p53 from HDM2-mediated degradation. Cell. 2004;5(5):465–75. doi: 10.1016/s1535-6108(04)00110-2.
  12. Lindstrom MS. NPM1/B23: A Multifunctional Chaperone in Ribosome Biogenesis and Chromatin Remodeling. Biochem Res Int. 2011;2011:1–16. doi: 10.1155/2011/195209.
  13. Falini B, Bolli NI, Martelli MP, et al. Translocations and mutations involving the nucleophosmin (NPM1) gene in lymphomas and leukemias. 2007;92(4):519–32. doi: 10.3324/haematol.11007.
  14. Falini B, Bigerna B, Pucciarini A, et al. Aberrant subcellular expression of nucleophosmin and NPM-MLF1 fusion protein in acute myeloid leukaemia carrying t(3;5): a comparison with NPMc+ AML. Leukemia. 2006;20(2):368–71. doi: 10.1038/sj.leu.2404068.
  15. Redner R, Rush EA, Faas S, et al. The t(5;17) variant of acute promyelocytic leukemia expresses a nucleophosmin-retinoic acid receptor fusion. 1996;87(3):882–6.
  16. Sportoletti P, Varasano E, Rossi R, et al. Mouse models of NPM1-mutated acute myeloid leukemia: biological and clinical implications. 2015;29(2):269–78. doi: 10.1038/leu.2014.257.
  17. Grisendi S, Mecucci C, Falini B, Pandolfi PP. Nucleophosmin and cancer. Nat Rev Cancer. 2006;6(7):493–505. doi: 10.1038/nrc1885.
  18. Sportoletti P, Grisendi S, Majid SM, et al. Npm1 is a haploinsufficient suppressor of myeloid and lymphoid malignancies in the mouse. Blood; 2008;111(7):3859–62. doi: 10.1182/blood-2007-06-098251.
  19. Ferrara F, Schiffer CA. Acute myeloid leukaemia in adults. The Lancet. 2013;381(9865):484–95. doi: 10.1016/s0140-6736(12)61727-9.
  20. Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic Nucleophosmin in Acute Myelogenous Leukemia with a Normal Karyotype. N Engl J Med. 2005;352(3):254–66. doi: 10.1056/nejmoa041974.
  21. Falini B, Martelli MP, Pileri SA, Mecucci C. Molecular and alternative methods for diagnosis of acute myeloid leukemia with mutated NPM1: flexibility may help. 2010;95(4):529–34. doi: 10.3324/haematol.2009.017822.
  22. Falini B, Albiero E, Bolli N, et al. Aberrant cytoplasmic expression of C-terminal-truncated NPM leukaemic mutant is dictated by tryptophans loss and a new NES motif. Leukemia. 2007;21(9):2052–4. doi: 10.1038/sj.leu.2404839.
  23. Schlenk RF, Dohner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–18. doi: 10.1056/nejmoa074306.
  24. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 Mutations Are Frequent Genetic Alterations in Acute Myeloid Leukemia and Confer Adverse Prognosis in Cytogenetically Normal Acute Myeloid Leukemia With NPM1 Mutation Without FLT3 Internal Tandem Duplication. J Clin Oncol. 2010. 28(22):3636–43. doi: 10.1200/jco.2010.28.3762.
  25. Dvorakova D, Racil Z, Jeziskova I, et al. Monitoring of minimal residual disease in acute myeloid leukemia with frequent and rare patient-specific NPM1 mutations. Am J Hematol. 2010;85(12):926–9. doi: 10.1002/ajh.21879.
  26. Schnittger S, Kern W, Tschulik C, et al. Minimal residual disease levels assessed by NPM1 mutation–specific RQ-PCR provide important prognostic information in AML. Blood. 2009;114(11):2220–31. doi: 10.1182/blood-2009-03-213389.
  27. Stahl T, Badbaran A, Kroger N, et al. Minimal residual disease diagnostics in patients with acute myeloid leukemia in the post-transplant period: comparison of peripheral blood and bone marrow analysis. Leuk Lymphoma. 2010;51(10):1837–43. doi: 10.3109/10428194.2010.508822.
  28. Kronke J, Schlenk RF, Jensen KO, et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol. 2011;29(19):2709–16. doi: 10.1200/jco.2011.35.0371.
  29. Rosnet O, Schiff C, Pebusque MJ, et al. Human FLT3/FLK2 gene: cDNA cloning and expression in hematopoietic cells. Blood. 1993;82(4):1110–9.
  30. Meshinchi S, Appelbaum FR. Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res. 2009;15(13):4263–9. doi: 10.1158/1078-0432.ccr-08-1123.
  31. Sitnicka E, Buza-Vidas N, Larsson S, et al. Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells. Blood. 2003;102(3):881–6. doi: 10.1182/blood-2002-06-1694.
  32. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002;100(5):1532–42. doi: 10.1182/blood-2002-02-0492.
  33. Adolfsson J, Borge OJ, Bryder D, et al. Upregulation of Flt3 Expression within the Bone Marrow Lin–Sca1+c-kit+ Stem Cell Compartment Is Accompanied by Loss of Self-Renewal Capacity. Immunity. 2001;15(4):659–69. doi: 10.1016/s1074-7613(01)00220-5.
  34. Griffith J, Black J, Faerman C, et al. The Structural Basis for Autoinhibition of FLT3 by the Juxtamembrane Domain. Mol Cell. 2004;13(2):169–78. doi: 10.1016/s1097-2765(03)00505-7.
  35. Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111(5):2776–84. doi: 10.1182/blood-2007-08-109090.
  36. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9. doi: 10.1182/blood.v98.6.1752.
  37. Marcucci G, Haferlach T, Dohner H. Molecular Genetics of Adult Acute Myeloid Leukemia: Prognostic and Therapeutic Implications. J Clin Oncol. 2011;29(5):475–86. doi: 10.1200/jco.2010.30.2554.
  38. Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. 2002;100(1):59–66. doi: 10.1182/blood.v100.1.59.
  39. Breitenbuecher F, Schnittger S, Grundler R, et al. Identification of a novel type of ITD mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor. Blood. 2009;113:4074–7. doi: 10.1182/blood-2007-11-125476.
  40. Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood. 2009;114(12):2386–92. doi: 10.1182/blood-2009-03-209999.
  41. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD–positive AML with respect to allogeneic transplantation. Blood. 2014;124(23):3441–9. doi: 10.1182/blood-2014-05-578070.
  42. Gu TL, Nardone J, Wang Y, et al. Survey of Activated FLT3 Signaling in Leukemia. PLoS One, 2011;6(4):e19169. doi: 10.1371/journal.pone.0019169.
  43. Rocnik JL, Okabe R, Yu JC, et al. Roles of tyrosine 589 and 591 in STAT5 activation and transformation mediated by FLT3-ITD. Blood. 2006;108(4):1339–45. doi: 10.1182/blood-2005-11-011429.
  44. Blau O, Berenstein R, Sindram A, Blau IW. Molecular analysis of different FLT3-ITD mutations in acute myeloid leukemia. Leuk Lymphoma. 2013;54(1):145–52. doi: 10.3109/10428194.2012.704999.
  45. Frohling S, Schlenk RF, Breitruck J, et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm. Blood. 2002;100(13):4372–80. doi: 10.1182/blood-2002-05-1440.
  46. Mrozek K, Marcucci G, Paschka P, et al. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification? Blood. 2007;109(2):431–48. doi: 10.1182/blood-2006-06-001149.
  47. Sengsayadeth SM, Jagasia M, Engelhardt BG, et al. Allo-SCT for high-risk AML-CR1 in the molecular era: impact of FLT3/ITD outweighs the conventional markers. Bone Marrow Transplant. 2012;47(12):1535–7. doi: 10.1038/bmt.2012.88.
  48. Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97(8):2434–9. doi: 10.1182/blood.v97.8.2434.
  49. Mead AJ, Linch DC, Hills RK, et al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood. 2007;110(4):1262–70. doi: 10.1182/blood-2006-04-015826.
  50. Whitman SP, Ruppert AS, Radmacher MD, et al. FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood. 2008;111(3):1552–9. doi: 10.1182/blood-2007-08-107946.
  51. Ozeki K, Kiyoi H, Hirose Y, et al. Biologic and clinical significance of the FLT3 transcript level in acute myeloid leukemia. Blood. 2004;103(5):1901–8. doi: 10.1182/blood-2003-06-1845.
  52. Ley TJ, Miller C, Ding L, et al. Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia. N Engl J Med. 2013;368(22):2059–74. doi: 10.1056/nejmoa1301689.
  53. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood. 2013;121(23):4769–77. doi: 10.1182/blood-2012-10-461624.
  54. Kottaridis PD, Gale RE, Langabeer SE, et al. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood. 2002;100(7):2393–8. doi: 10.1182/blood-2002-02-0420.
  55. Shih LY, Huang CF, Wu JH, et al. Internal tandem duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis of bone marrow samples from 108 adult patients at diagnosis and relapse. Blood. 2002;100(7):2387–92. doi: 10.1182/blood-2002-01-0195.
  56. Chu SH, Small D. Mechanisms of resistance to FLT3 inhibitors. Drug Resist Update. 2009;12(1–2):8–16. doi: 10.1016/j.drup.2008.12.001.
  57. Moore AS, Faisal A, Gonzalez de Castro D, et al. Selective FLT3 inhibition of FLT3-ITD+ acute myeloid leukaemia resulting in secondary D835Y mutation: a model for emerging clinical resistance patterns. Leukemia. 2012;26(7):1462–70. doi: 10.1038/leu.2012.52.
  58. Mead AJ, Gale RE, Kottaridis PD, et al. Acute myeloid leukaemia blast cells with a tyrosine kinase domain mutation of FLT3 are less sensitive to lestaurtinib than those with a FLT3 internal tandem duplication. Br J Haematol. 2008;141(4):454–60. doi: 10.1111/j.1365-2141.2008.07025.x.
  59. Koschmieder S, Halmos B, Levantini E, Tenen DG. Dysregulation of the C/EBPa Differentiation Pathway in Human Cancer. J Clin Oncol. 2009;27(4):619–28. doi: 10.1200/jco.2008.17.9812.
  60. Wang H, Iakova P, Wilde M, et al. C/EBPa Arrests Cell Proliferation through Direct Inhibition of Cdk2 and Cdk4. Mol Cell. 2001;8(4):817–28. doi: 10.1016/s1097-2765(01)00366-5.
  61. Radomska HS, Huettner CS, Zhang P, et al. CCAAT enhancer binding protein alpha is a regulatory switch sufficient for induction of granulocytic development from bipotential myeloid progenitors. Mol Cell Biol. 1998;18(7):4301–14. doi: 10.1128/mcb.18.7.4301.
  62. Zhang DE, Zhang P, Wang ND, et al. Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein a-deficient mice. Proc Natl Acad Sci USA. 1997;94(2):569–74. doi: 10.1073/pnas.94.2.569.
  63. Umek RM, Friedman AD, McKnight SL. CCAAT-enhancer binding protein: a component of a differentiation switch. Science. 1991;251(4991):288–92. doi: 10.1126/science.1987644.
  64. Watkins PJ, Condreay JP, Huber BE, et al. Proliferation and tumorigenicity induced by CCAAT/enhancer-binding protein. Cancer Res. 1996;56(5):1063–7.
  65. Pabst T, Mueller BU, Zhang P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-[alpha] (C/EBP [alpha]), in acute myeloid leukemia. Nat Genet. 2001;27(3):263–70. doi: 10.1038/85820.
  66. Nerlov C. C/EBP [alpha] mutations in acute myeloid leukaemias. Nat Rev Cancer. 2004;4(5):394–400. doi: 10.1038/nrc1363.
  67. Wouters BJ, Jorda MA, Keeshan K, et. al. Distinct gene expression profiles of acute myeloid/T-lymphoid leukemia with silenced CEBPA and mutations in NOTCH1. Blood. 2007;110(10):3706–14. doi: 10.1182/blood-2007-02-073486.
  68. Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood. 2011;117(8):2469–75. doi: 10.1182/blood-2010-09-307280.
  69. Kirstetter P, Schuster MB, Bereshchenko O, et al. Modeling of C/EBPa Mutant Acute Myeloid Leukemia Reveals a Common Expression Signature of Committed Myeloid Leukemia-Initiating Cells. Cancer Cell. 2008;13(4):299–310. doi: 10.1016/j.ccr.2008.02.008.
  70. Shih LY, Liang DC, Huang CF, et al. AML patients with CEBP [alpha] mutations mostly retain identical mutant patterns but frequently change in allelic distribution at relapse: a comparative analysis on paired diagnosis and relapse samples. Leukemia. 2006;20(4):604–9. doi: 10.1038/sj.leu.2404124.
  71. Wouters BJ, Lowenberg B, Erpelinck-Verschueren CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood. 2009;113(13):3088–91. doi: 10.1182/blood-2008-09-179895.
  72. Cagnetta A, Adamia S, Acharya C, et al. Role of genotype-based approach in the clinical management of adult acute myeloid leukemia with normal cytogenetics. Leuk Res. 2014;38(6):649–59. doi: 10.1016/j.leukres.2014.03.006.
  73. Wouters BJ, Sanders MA, Lugthart S, et al. Segmental uniparental disomy as a recurrent mechanism for homozygous CEBPA mutations in acute myeloid leukemia. Leukemia. 2007;21(11):2382–4. doi: 10.1038/sj.leu.2404795.
  74. Valk PJM, Verhaak RG, Beijen MA, et al. Prognostically Useful Gene-Expression Profiles in Acute Myeloid Leukemia. N Engl J Med. 2004;350(16):1617–28. doi: 10.1056/nejmoa040465.
  75. Marceau-Renaut A, Guihard S, Castaigne S, et al. Classification of CEBPA mutated acute myeloid leukemia by GATA2 mutations. Am J Hematol. 2015;90(5):E93–4. doi: 10.1002/ajh.23949.
  76. Pabst T, Mueller BU. Transcriptional dysregulation during myeloid transformation in AML. Oncogene. 2007;26(47):6829–37. doi: 10.1038/sj.onc.1210765.
  77. Frohling S, Schlenk RF, Krauter J, et al. Acute myeloid leukemia with deletion 9q within a noncomplex karyotype is associated with CEBPA loss-of-function mutations. Genes Chromos Cancer. 2005;42(4):427–32. doi: 10.1002/gcc.20152.
  78. Green CL, Koo KK, Hills RK, et al. Prognostic Significance of CEBPA Mutations in a Large Cohort of Younger Adult Patients With Acute Myeloid Leukemia: Impact of Double CEBPA Mutations and the Interaction With FLT3 and NPM1 Mutations. J Clin Oncol. 2010;28(16):2739–47. doi: 10.1200/jco.2009.26.2501.
  79. Behdad A, Weigelin HC, Elenitoba-Johnson KS, Betz BL. A Clinical Grade Sequencing-Based Assay for CEBPA Mutation Testing: Report of a Large Series of Myeloid Neoplasms. J Mol Diagn. 2015;17(1):76–84. doi: 10.1016/j.jmoldx.2014.09.007.
  80. Bienz M, Ludwig M, Leibundgut EO, et al. Risk Assessment in Patients with Acute Myeloid Leukemia and a Normal Karyotype. Clin Cancer Res. 2005;11(4):1416–24. doi: 10.1158/1078-0432.ccr-04-1552.
  81. Frohling S, Schlenk RF, Stolze I, et al. CEBPA Mutations in Younger Adults With Acute Myeloid Leukemia and Normal Cytogenetics: Prognostic Relevance and Analysis of Cooperating Mutations. J Clin Oncol. 2004;22(4):624–33. doi: 10.1200/jco.2004.06.060.
  82. Preudhomme C, Sagot C, Boissel N, et al. Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association (ALFA). Blood. 2002;100(8):2717–23. doi: 10.1182/blood-2002-03-0990.
  83. Pastore F, Kling D, Hoster E, et al. Long-term follow-up of cytogenetically normal CEBPA-mutated AML. J Hematol Oncol. 2014;7(1):55. doi: 10.1186/s13045-014-0055-7.
  84. Park SH, Chi H-S, Cho Y-U, et al. CEBPA single mutation can be a possible favorable prognostic indicator in NPM1 and FLT3-ITD wild-type acute myeloid leukemia patients with intermediate cytogenetic risk. Leuk Res. 2013;37(11):1488–94. doi: 10.1016/j.leukres.2013.08.014.
  85. Renneville A, Boissel N, Gachard N, et al. The favorable impact of CEBPA mutations in patients with acute myeloid leukemia is only observed in the absence of associated cytogenetic abnormalities and FLT3 internal duplication. Blood. 2009;113(21):5090–3. doi: 10.1182/blood-2008-12-194704.
  86. Taniuchi I, Littman DR. Epigenetic gene silencing by Runx proteins. Oncogene. 2004;23(24):4341–5. doi: 10.1038/sj.onc.1207671.
  87. Yoshida H, Kitabayashi I. Chromatin regulation by AML1 complex. Int J Hematol. 2008;87(1):19–24. doi: 10.1007/s12185-007-0004-0.
  88. Tang JL, Hou HA, Chen CY, et al. AML1/RUNX1 mutations in 470 adult patients with de novo acute myeloid leukemia: prognostic implication and interaction with other gene alterations. 2009;114(26):5352–61. doi: 10.1182/blood-2009-05-223784.
  89. Dicker F, Haferlach C, Sundermann J, et al. Mutation analysis for RUNX1, MLL-PTD, FLT3-ITD, NPM1 and NRAS in 269 patients with MDS or secondary AML. Leukemia. 2010;24(8):1528–32. doi: 10.1038/leu.2010.124.
  90. Gaidzik VI, Bullinger L, Schlenk RF, et al. RUNX1 Mutations in Acute Myeloid Leukemia: Results From a Comprehensive Genetic and Clinical Analysis From the AML Study Group. J Clin Oncol. 2011;29(10):1364–72. doi: 10.1200/jco.2010.30.7926.
  91. Dicker F, Haferlach C, Kern W, et al. Trisomy 13 is strongly associated with AML1/RUNX1 mutations and increased FLT3 expression in acute myeloid leukemia. Blood. 2007;110:1308–16. doi: 10.1182/blood-2007-02-072595.
  92. Matsuno N, Osato M, Yamashita N, et al. Dual mutations in the AML1 and FLT3 genes are associated with leukemogenesis in acute myeloblastic leukemia of the M0 subtype. Leukemia. 2003;17(12):2492–9. doi: 10.1038/sj.leu.2403160.
  93. Mendler JH, Maharry K, Becker H, et al. In rare acute myeloid leukemia patients harboring both RUNX1 and NPM1 mutations, RUNX1 mutations are unusual in structure and present in the germline. 2013;98(8):e92–4. doi: 10.3324/haematol.2013.089904.
  94. Fasan A, Haferlach C, Kohlmann A, et al. Rare coincident NPM1 and RUNX1 mutations in intermediate risk acute myeloid leukemia display similar patterns to single mutated cases. Haematologica. 2014;99(2):e20–1. doi: 10.3324/haematol.2013.099754.
  95. Fernandez-Medarde A, Santos E. Ras in Cancer and Developmental Diseases. Genes Cancer. 2011;2(3):344–58. doi: 10.1177/1947601911411084.
  96. Stites EC, Ravichandran KS. A Systems Perspective of Ras Signaling in Cancer. Clin Cancer Res. 2009;15(5):1510–3. doi: 10.1158/1078-0432.ccr-08-2753.
  97. Johnson DB, Smalley KSM, Sosman JA. Molecular Pathways: Targeting NRAS in Melanoma and Acute Myelogenous Leukemia. Clin Cancer Res. 2014;20(16):4186–92. doi: 10.1158/1078-0432.ccr-13-3270.
  98. Fedorenko IV, Gibney GT, Smalley KSM. NRAS mutant melanoma: biological behavior and future strategies for therapeutic management. Oncogene. 2013;32(25):3009–18. doi: 10.1038/onc.2012.
  99. Reuter CM, Krauter J, Onono FO, et al. Lack of noncanonical RAS mutations in cytogenetically normal acute myeloid leukemia. Ann Hematol. 2014;93(6):977–82. doi: 10.1007/s00277-014-2061-9.
  100. Bacher U, Haferlach T, Schoch C, et al. Implications of NRAS mutations in AML: a study of 2502 patients. Blood. 2006;107(10):3847–53. doi: 10.1182/blood-2005-08-3522.
  101. Padua RA, West RR. Oncogene mutation and prognosis in the myelodysplastic syndromes. Br J Haematol. 2000;111(3):873–4. doi: 10.1111/j.1365-2141.2000.02472.x.
  102. Berman JN, Gerbing RB, Alonzo TA, et al. Prevalence and clinical implications of NRAS mutations in childhood AML: a report from the Children’s Oncology Group. 2011;25(6):1039–42. doi: 10.1038/leu.2011.31.
  103. Bowen DT, Frew ME, Hills R, et al. RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood. 2005;106(6):2113–9. doi: 10.1182/blood-2005-03-0867.
  104. Roskoski R Jr. Structure and regulation of Kit protein-tyrosine kinase–The stem cell factor receptor. Biochem Biophys Res Commun. 2005;338(3):1307–15. doi: 10.1016/j.bbrc.2005.09.150.
  105. Yarden Y, Ullrich A. Growth Factor Receptor Tyrosine Kinases. Ann Rev Biochem. 1988:57(1):443–78. doi: 10.1146/annurev.bi.57.070188.002303.
  106. Paschka P, Marcucci G, Ruppert AS, et al. Adverse Prognostic Significance of KIT Mutations in Adult Acute Myeloid Leukemia With inv(16) and t(8;21): A Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24(24):3904–11. doi: 10.1200/jco.2006.06.9500.
  107. Riera L, Marmont F, Toppino D, et al. Core binding factor acute myeloid leukaemia and c-KIT mutations. Oncol Rep. 2013;29(5):1867–72. doi: 10.3892/or.2013.2328.
  108. Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood. 2006;107(9):3463–8. doi: 10.1182/blood-2005-09-3640.
  109. Park SH, Chi HS, Min SK, et al. Prognostic impact of c-KIT mutations in core binding factor acute myeloid leukemia. Leuk Res. 2011;35(10):1376–83. doi: 10.1016/j.leukres.2011.06.003.
  110. Hoyos M, Nomdedeu JF, Esteve J, et al. Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease. Eur J Haematol. 2013;91(3):209–18. doi: 10.1111/ejh.12130.
  111. Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107(5):1791–9. doi: 10.1182/blood-2005-04-1466.
  112. Jiao B, Wu CF, Liang Y, et al. AML1-ETO9a is correlated with C-KIT overexpression/mutations and indicates poor disease outcome in t(8;21) acute myeloid leukemia-M2. Leukemia. 2009;23(9):1598–604. doi: 10.1038/leu.2009.104.
  113. Qin YZ, Zhu HH, Jiang Q, et al. Prevalence and prognostic significance of c-KIT mutations in core binding factor acute myeloid leukemia: A comprehensive large-scale study from a single Chinese center. Leuk Res. 2014;38(12):1435–40. doi: 10.1016/j.leukres.2014.09.017.
  114. O’Donnell MR, Tallman MS, Abboud CN, et al. Acute Myeloid Leukemia, Version 2.2013. J Natl Compr Canc Netw. 2013;11(9):1047–55.
  115. Tokumasu M, Murata C, Shimada A, et al. Adverse prognostic impact of KIT mutations in childhood CBF-AML: the results of the Japanese Pediatric Leukemia/Lymphoma Study Group AML-05 trial. Leukemia. 2015;29(12):2438–41. doi: 10.1038/leu.2015.121.
  116. Ito S, D’Alessio AC, Taranova OV, et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466(7310):1129–33. doi: 10.1038/nature09303.
  117. Chen Q, Chen Y, Bian C, et al. TET2 promotes histone O-GlcNAcylation during gene transcription. 2013;493(7433):561–4. doi: 10.1038/nature11742.
  118. Aslanyan M, Kroeze LI, Langemeijer SM, et al. Clinical and biological impact of TET2 mutations and expression in younger adult AML patients treated within the EORTC/GIMEMA AML-12 clinical trial. Ann Hematol. 2014;93(8):1401–12. doi: 10.1007/s00277-014-2055-7.
  119. Chou WC, Chou SC, Liu CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood. 2011;118(14):3803–10. doi: 10.1182/blood-2011-02-339747.
  120. Metzeler KH, Maharry K, Radmacher MD, et al. TET2 Mutations Improve the New European LeukemiaNet Risk Classification of Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol. 2011;29(10):1373–81. doi: 10.1200/jco.2010.32.7742.
  121. Gaidzik VI, Paschka P, Spath D, et al. TET2 Mutations in Acute Myeloid Leukemia (AML): Results From a Comprehensive Genetic and Clinical Analysis of the AML Study Group. J Clin Oncol. 2012;30(12):1350–7. doi: 10.1200/jco.2011.39.2886.
  122. Ko M, Huang Y, Jankowska AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. 2010;468(7325):839–43. doi: 10.1038/nature09586.
  123. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67. doi: 10.1016/j.ccr.2010.11.015.
  124. Moran-Crusio K, Reavie L, Shih A, et al. Tet2 Loss Leads to Increased Hematopoietic Stem Cell Self-Renewal and Myeloid Transformation. Cancer Cell. 2011;20(1):11–24. doi: 10.1016/j.ccr.2011.06.001.
  125. Quivoron C, Couronne L, Della Valle V, et al. TET2 Inactivation Results in Pleiotropic Hematopoietic Abnormalities in Mouse and Is a Recurrent Event during Human Lymphomagenesis. Cancer Cell. 2011;20(1):25–38. doi: 10.1016/j.ccr.2011.06.003.
  126. Nibourel O, Kosmider O, Cheok M, et al. Incidence and prognostic value of TET2 alterations in de novo acute myeloid leukemia achieving complete remission. Blood. 2010;116(7):1132–5. doi: 10.1182/blood-2009-07-234484.
  127. Weissmann S, Alpermann T, Grossmann V, et al. Landscape of TET2 mutations in acute myeloid leukemia. Leukemia. 2012;26(5):934–42. doi: 10.1038/leu.2011.326.
  128. Reitman ZJ, Yan H. Isocitrate Dehydrogenase 1 and 2 Mutations in Cancer: Alterations at a Crossroads of Cellular Metabolism. J Natl Cancer Inst. 2010;102(13):932–41. doi: 10.1093/jnci/djq187.
  129. Molenaar RJ, Radivoyevitch T, Maciejewski JP, et al. The driver and passenger effects of isocitrate dehydrogenase 1 and 2 mutations in oncogenesis and survival prolongation. Biochim Biophys Acta. 2014;1846(2):326–41. doi: 10.1016/j.bbcan.2014.05.004.
  130. Emadi A, Faramand R, Carter-Cooper B, et al. Presence of isocitrate dehydrogenase (IDH) mutations may predict clinical response to hypomethylating agents in patients with acute myeloid leukemia (AML). Am J Hematol. 2015;90(5):E77–9. doi: 10.1002/ajh.23965.
  131. Abbas S, Lugthart S, Kavelaars FG, et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010;116(12):2122–6. doi: 10.1182/blood-2009-11-250878.
  132. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 Gene Mutations Identify Novel Molecular Subsets Within De Novo Cytogenetically Normal Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/jco.2009.27.3730.
  133. Dang L, Jin S, Su SM. IDH mutations in glioma and acute myeloid leukemia. Trends Mol Med. 2010;16(9):387–97. doi: 10.1016/j.molmed.2010.07.002.
  134. Horbinski C. What do we know about IDH1/2 mutations so far, and how do we use it? Acta Neuropathol. 2013;125(5):621–36. doi: 10.1007/s00401-013-1106-9.
  135. Chotirat S, Thongnoppakhun W, Wanachiwanawin W, Auewarakul CU. Acquired somatic mutations of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) in preleukemic disorders. Blood Cells Mol Dis. 2015;54(3):286–91. doi: 10.1016/j.bcmd.2014.11.017.
  136. Green CL, Evans CM, Zhao L, et al. The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood. 2011;118(2):409–12. doi: 10.1182/blood-2010-12-322479.
  137. Zhou KG, Jiang LJ, Shang Z, et al. Potential application of IDH1 and IDH2 mutations as prognostic indicators in non-promyelocytic acute myeloid leukemia: a meta-analysis. Leuk Lymphoma. 2012;53(12):2423–9. doi: 10.3109/10428194.2012.695359.
  138. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2012;30(7):742–50. doi: 10.1200/jco.2011.39.2092.
  139. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33. doi: 10.1056/nejmoa1005143.
  140. Zhang Y, Chen FQ, Sun YH, et al. Effects of DNMT1 silencing on malignant phenotype and methylated gene expression in cervical cancer cells. J Exp Clin Cancer Res. 2011;30(1):98. doi: 10.1186/1756-9966-30-98.
  141. Jasielec J, Saloura V, Godley LA. The mechanistic role of DNA methylation in myeloid leukemogenesis. Leukemia. 2014;28(9):1765–73. doi: 10.1038/leu.2014.163.
  142. Li KK, Luo LF, Shen Y, et al. DNA methyltransferases in hematologic malignancies. Semin Hematol. 2013;50(1):48–60. doi: 10.1053/j.seminhematol.2013.01.005.
  143. O’Brien EC, Brewin J, Chevassut T. DNMT3A: the DioNysian MonsTer of acute myeloid leukaemia. Ther Adv Hematol. 2014;5(6):187–96. doi: 10.1177/2040620714554538.
  144. Holz-Schietinger C, Matje DM, Reich NO. Mutations in DNA methyltransferase (DNMT3A) observed in acute myeloid leukemia patients disrupt processive methylation. J Biol Chem. 2012;287(37):30941–51. doi: 10.1074/jbc.m112.366625.
  145. Russler-Germain DA, Spencer DH, Young MA, et al. The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell. 2014;25(4):442–54. doi: 10.1016/j.ccr.2014.02.010146.
  146. McDevitt MA. Clinical applications of epigenetic markers and epigenetic profiling in myeloid malignancies. Semin Oncol. 2012;39(1):109–22. doi: 10.1053/j.seminoncol.2011.11.003.
  147. Berenstein R, Blau IW, Suckert N, et al. Quantitative detection of DNMT3A R882H mutation in acute myeloid leukemia. J Exp Clin Cancer Res. 2015;34(1):55. doi: 10.1186/s13046-015-0173-2.
  148. Shlush LI, Zandi S, Mitchell A, et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014;506(7488):328–33. doi: 10.1038/nature13038.
  149. Corces-Zimmerman MR, Hong WJ, Weissman IL, et al. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci USA. 2014;111(7):2548–53. doi: 10.1073/pnas.1324297111.
  150. Thol F, Damm F, Ludeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol. 2011;29(21):2889–96. doi: 10.1200/jco.2011.35.4894.
  151. Ribeiro AF, Pratcorona M, Erpelinck-Verschueren C, et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood. 2012;119(24):5824–31. doi: 10.1182/blood-2011-07-367961.
  152. Ibrahem L, Mahfouz R, Elhelw L, et al. Prognostic significance of DNMT3A mutations in patients with acute myeloid leukemia. Blood Cells Mol Dis. 2014;54(1):84–9. doi: 10.1016/j.bcmd.2014.07.015.
  153. Shivarov V, Gueorguieva R, Stoimenov A, Tiu R. DNMT3A mutation is a poor prognosis biomarker in AML: results of a meta-analysis of 4500 AML patients. Leuk Res. 2013;37(11):1445–50. doi: 10.1016/j.leukres.2013.07.032.
  154. Wakita S, Yamaguchi H, Omori I, et al. Mutations of the epigenetics-modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia. 2013;27(5):1044–52. doi: 10.1038/leu.2012.317.
  155. Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood. 2012;119(2):559–68. doi: 10.1182/blood-2011-07-369934.
  156. Ploen GG, Nederby L, Guldberg P, et al. Persistence of DNMT3A mutations at long-term remission in adult patients with AML. Br J Haematol. 2014;167(4):478–86. doi: 10.1111/bjh.13062.
  157. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–98. doi: 10.1056/nejmoa1408617.

Моноклональные антитела: от создания до клинического применения

Ю.И. Будчанов

ГБОУ ВПО «Тверской медицинский университет», ул. Советская, д. 4, Тверь, Российская Федерация, 170000

Для переписки: Юрий Иванович Будчанов, 1-й пер. Красной Слободы, д. 3, Тверь, Российская Федерация, 170001; e-mail: budjur@mail.ru

Для цитирования: Будчанов Ю.И. Моноклональные антитела: от создания до клинического применения. Клиническая онкогематология. 2016;9(3):237-44.

DOI: 10.21320/2500-2139-2016-9-3-237-244


РЕФЕРАТ

Создание моноклональных антител (МКА) привело к революционным достижениям в диагностике и лечении онкогематологических заболеваний. В обзоре рассматриваются история создания, новые улучшенные технологии получения моноклональных антител на примере анти-CD20-МКА, распознающих различные эпитопы антигена CD20 и обладающих повышенной противоопухолевой активностью. Инженерные модификации должны помочь понять эффекторные механизмы использования новых анти-CD20-МКА и направлены на дальнейшее улучшение результатов лечения.


Ключевые слова: моноклональные антитела, ритуксимаб, офатумумаб, обинутузумаб, гибридомная технология.

Получено: 13 января 2016 г.

Принято в печать: 17 марта 2016 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256(5517):495–7. doi: 10.1038/256495a0.
  2. Galfre G. Antibodies to major histocompatibility antigens produced by hybrid cell lines. Nature. 1977;266(5602):550–2. doi: 10.1038/266550a0.
  3. Гордеева О.Б., Семикина Е.Л. Современные возможности определения группы крови и резус-принадлежности в педиатрической практике. Вопросы диагностики в педиатрии. 2010;2(4):9–16.
    [Gordeeva OB, Semikina EL. Current capabilities of the blood group and Rhesus factor typing in pediatric practice. Voprosy diagnostiki v pediatrii. 2010;2(4):9–16. (In Russ)]
  4. Рагимов А.А., Дашкова Н.Г. Трансфузионная иммунология. М.: МИА, 2004. С. 270.
    [Ragimov AA, Dashkova NG. Transfuzionnaya immunologiya. (Transfusion immunology.) Moscow: MIA Publ.; 2004. pp. 270. (In Russ)]
  5. Freedman A. Follicular lymphoma: 2014 update on diagnosis and management. Am J Hematol. 2014;89(4):429–36. doi: 10.1002/ajh.23674.
  6. Preijers FW, Huys E, Moshaver B. OMIP-010: a new 10-color monoclonal antibody panel for polychromatic immunophenotyping of small hematopoietic cell samples. Cytometry A. 2012;81A(6):453–5. doi: 10.1002/cyto.a.22056.
  7. Тупицын Н.Н., Гривцова Л.Ю., Купрышина Н.А. Иммунодиагностика опухолей крови на основании многоцветных (8 цветов панелей) европейского консорциума по проточной цитометрии (EURO-FLOW). Иммунология гемопоэза. 2015;13(1):31–62.
    [Tupitsyn NN, Grivtsova LYu, Kupryshina NA. Haematopoietic malignancies immune diagnostics based on Euroflow Consortium proposals: 8-color flow cytometry. Immunologiya gemopoeza. 2015;13(1):31–62. (In Russ)]
  8. Тупицын Н.Н. Иммунология клеток крови. В кн.: Гематология. Национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. С. 69–79.
    [Tupitsyn NN. Blood cell immunology. In: Rukavitsyn OA, ed. Gematologiya. Natsional’noe rukovodstvo. (Hematology. National guidelines.) Moscow: GEOTAR-Media Publ.; 2015. pp. 69–79. (In Russ)]
  9. Carter PJ. Potent antibody therapeutics by design. Nat Rev Immunol. 2006;6:343–57. doi: 10.1038/nri1837.
  10. Riley JK, Sliwkowski MX. CD20: a gene in search of a function. Semin Oncol. 2000;27(12):17–24.
  11. Tedder TF, Engel P. CD20: a regulator of cell-cycle progression of B lymphocytes. Immunol Today. 1994;15(9):450–4. doi: 10.1016/0167-5699(94)90276-3.
  12. Renaudineau Y, Devauchelle-Pensec V, Hanrotel C, et al. Monoclonal anti-CD20 antibodies: mechanisms of action and monitoring of biological effects. Joint Bone Spine. 2009;76(5):458–63. doi: 10.1016/j.jbspin.2009.03.010.
  13. Martin P, Furman RR, Coleman M, Leonard JP. Phase I to III trials of anti-B cell therapy in non-Hodgkin’s lymphoma. Clin Cancer Res. 2007;13(18):5636–42. doi: 10.1158/1078-0432.ccr-07-1085.
  14. St Clair EW. Novel targeted therapies for autoimmunity. Curr Opin Immunol. 2009;21(6):648–57. doi: 10.1016/j.coi.2009.09.008.
  15. Gurcan H, Keskin D, Stern J, et al. A review of the current use of rituximab in autoimmune diseases. Int Immunopharmacol. 2009;9(1):10–25. doi: 10.1016/j.intimp.2008.10.004.
  16. Castillo-Trivino T, Braithwaite D, Bacchetti P, Waubant E. Rituximab in relapsing and progressive forms of multiple sclerosis: a systematic review. PLoS One. 2013;8(7):e66308. doi: 10.1371/journal.pone.0066308.
  17. Otukesh H, Hoseini R, Rahimzadeh N, Fazel M. Rituximab in the treatment of nephrotic syndrome: a systematic review. Iran J Kidney Dis. 2013;7(4):249–56. doi: 10.13172/2053-0293-1-1-480.
  18. Morrison VA. Immunosuppression associated with novel chemotherapy agents and monoclonal antibodies. Clin Infect Dis. 2014;59(5):360–4. doi: 10.1093/cid/ciu592.
  19. Rosman Z, Shoenfeld Y, Zandman-Goddard G. Biologic therapy for autoimmune diseases: an update. BMC Med. 2013;11(1):88. doi: 10.1186/1741-7015-11-88.
  20. Bhandari PR, Pai VV. Novel applications of Rituximab in dermatological disorders. Indian Dermatol Online J. 2014;5(3):250–9. doi: 10.4103/2229-5178.137766.
  21. Cang S, Mukhi N, Wang K, Liu D. Novel CD20 monoclonal antibodies for lymphoma therapy. J Hematol Oncol. 2012;5(1):64. doi: 10.1186/1756-8722-5-64.
  22. Rioufol C, Salles G. Obinutuzumab for chronic lymphocytic leukemia. Expert Rev Hematol. 2014;7(5):533–43. doi: 10.1586/17474086.2014.953478.
  23. Owen CJ, Stewart DA. Obinutuzumab for the treatment of patients with previously untreated chronic lymphocytic leukemia: overview and perspective. Ther Adv Hematol. 2015;6(4):161–70. doi: 10.1177/2040620715586528.
  24. Shah A. Obinutuzumab: A Novel Anti-CD20 Monoclonal Antibody for Previously Untreated Chronic Lymphocytic Leukemia. Ann Pharmacother. 2014;48(10):1356–61. doi: 10.1177/1060028014543271.
  25. Golay J, Da Roit F, Bologna L, et al. Glycoengineered CD20 antibody obinutuzumab activates neutrophils and mediates phagocytosis through CD16B more efficiently than rituximab. Blood. 2013;122(20):3482–91. doi: 10.1182/blood-2013-05-504043.
  26. Shah A. New developments in the treatment of chronic lymphocytic leukemia: role of obinutuzumab. Ther Clin Risk Manage. 2015;11:1113–22. doi: 10.2147/TCRM.S71839.
  27. Cerquozzi S, Owen C. Clinical role of obinutuzumab in the treatment of naive patients with chronic lymphocytic leukemia. Biol Targ Ther. 2015;9:13–22. doi: 10.2147/BTT.S61600.
  28. Seiter K, Mamorska-Dyga A. Obinutuzumab treatment in the elderly patient with chronic lymphocytic leukemia. Clin Interv Aging. 2015;12(10):951–61. doi: 10.2147/cia.s69278.
  29. Алексеев С.М., Капланов К.Д., Иванов Р.А., Черняева Е.В. Современный подход к разработке и исследованию биоаналогов на примере первого российского препарата моноклональных антител — Ацеллбия® (ритуксимаб). Исследования и практика в медицине. 2015;2(1):8–12. doi: 10.17709/2409-2231-2015-2-1-8-12.
    [Alekseev SM, Kaplanov KD, Ivanov RA, Chernyaeva EV. Current approach to development of biosimilar products containing monoclonal antibodies as an active substance – non-clinical studies of the first Russian rituximab biosimilar, Acellbia®. Research’n Practical Medicine Journal. 2015;2(1):8–12. doi: 10.17709/2409-2231-2015-2-1-8-12. (In Russ)]
  30. Tada M, Tatematsu K-I, Ishii-Watabe A, et al. Characterization of anti-CD20 monoclonal antibody produced by transgenic silkworms (Bombyx mori). mAbs. 2015;7(6):1138–50. doi: 10.1080/19420862.2015.1078054.
  31. Gonzalez-Gonzalez E, Alvarez MM, Marquez-Ipina AR, et al. Anti-Ebola therapies based on monoclonal antibodies: current state and challenges ahead. Crit Rev Biotechnol. 2015;26:1–16. doi: 10.3109/07388551.2015.1114465.

 

Истинная полицитемия: новая концепция диагностики и клинические формы

А.М. Ковригина1, В.В. Байков2

1 ФГБУ «Гематологический научный центр» Минздрава России, Новый Зыковский пр-д, д. 4а, Москва, Российская Федерация, 125167

2 НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой, ГБОУ ВПО «Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова» Минздрава России, ул. Рентгена, д. 12, Санкт-Петербург, Российская Федерация, 197022

Для переписки: Алла Михайловна Ковригина, д-р биол. наук, профессор, Новый Зыковский пр-д., д. 4а, Москва, Российская Федерация, 125167; тел.: +7(495)612-61-12; e-mail: kovrigina.alla@gmail.com

Для цитирования: Ковригина А.М., Байков В.В. Истинная полицитемия: новая концепция диагностики и клинические формы. Клиническая онкогематология. 2016;9(2):115–22.

DOI: 10.21320/2500-2139-2016-9-2-115-122


РЕФЕРАТ

Истинная полицитемия (ИП) — клональное Ph-негативное миелопролиферативное заболевание с избыточной пролиферацией 3 ростков кроветворения и исходом в стадию неэффективного миелопоэза. В классификации ВОЗ (2008) показатели гемоглобина и гематокрита включены в главные критерии диагностики заболевания. Однако у многих пациентов с ИП уровень этих показателей может оказаться ниже диагностического, что приводит к гиподиагностике данного заболевания. В настоящее время выделены три клинических формы болезни: 1) маскированная (латентная/продромальная), 2) классическая (развернутая), 3) ИП с прогрессированием/трансформацией в миелофиброз. Маскированная ИП наиболее сложна для диагностики. Она гетерогенна по клинико-лабораторным данным, анамнезу, течению заболевания; включает в себя ранние стадии, в т. ч. с высоким тромбоцитозом, имитирующие эссенциальную тромбоцитемию, случаи с абдоминальными тромбозами, а также длительно латентно протекающую ИП. Наиболее информативным способом диагностики маскированной формы ИП является исследование трепанобиоптата костного мозга. Обычно выявляется классическая картина ИП: гиперклеточный костный мозг с трехростковой пролиферацией миелопоэза, пролиферация мегакариоцитов со слабой или умеренно выраженной атипией и полиморфизмом клеток. Определение степени ретикулинового фиброза имеет важное прогностическое значение и отражает риск прогрессии/трансформации в миелофиброз. В новой редакции классификации ВОЗ (2016) морфологическое исследование трепанобиоптата костного мозга будет введено в большие критерии диагностики ИП в качестве обязательного при пограничных значениях уровня гемоглобина и гематокрита.


Ключевые слова: истинная полицитемия, миелопролиферативное заболевание, диагностика истинной полицитемии, формы истинной полицитемии.

Получено: 19 января 2016 г.

Принято в печать: 1 февраля 2016 г.

Читать статью в  PDF pdficon


ЛИТЕРАТУРА

  1. Cerquozzi S, Tefferi A. Blast transformation and fibrotic progression in polycythemia vera and essential thrombocythemia: a literature review of incidence and risk factors. Blood Cancer J. 2015;5(11):e366. doi: 10.1038/bcj.2015.95.
  2. Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352(17):1779–90. doi 10.1056/nejmoa051113.
  3. Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97. doi: 10.1016/j.ccr.2005.03.023.
  4. Chloe J, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434(7037):1144–8. doi: 10.1038/nature03546.
  5. Baxter EJ, Scott LM, Campbell PJ, et al. Cancer Genome Project. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. The Lancet. 2005;365(9464):1054–61. doi: 10.1016/s0140-6736(05)71142-9.
  6. Wang YL, Vandris K, Jones A, et al. JAK2 Mutations are present in all cases of polycythemia vera. Leukemia. 2008;22(6):1289. doi: 10.1038/sj.leu.2405047.
  7. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90. doi: 10.1056/NEJMoa1311347.
  8. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405. doi:  10.1056/NEJMoa1312542.
  9. Broseus J, Park JH, Carillo S, et al. Presence of calreticulin mutations in JAK2-negative polycythemia vera. Blood. 2014;124(26):3964–6. doi: 10.1182/blood-2014-06-583161.
  10. Fairbanks VF, Klee GG, Wiseman GA, et al. Measurement of Blood volume and Red Cell Mass: re-examination of 51Cr and 125I methods. Blood Cells Mol Dis. 1996;22(15):169–86. doi: 10.1006/bcmd.1996.0024.
  11. Lorberboym M, Rahimi-Levene N, Lipszyc H, et al. Analysis of Red Cell Mass and Plasma Volume in Patients With Polycythemia. Arch Pathol Lab Med. 2005;129:89–91.
  12. Murphy S. Diagnostic criteria and prognosis in polycythemia vera and essential thrombocythemia. Semin Hematol. 1999;36(1 Suppl 2):9–13.
  13. Alvarez-Larran A, Ancochea A, Angona A, et al. Red cell mass measurement in patients with clinically suspected diagnosis of polycythemia vera or essential thrombocythemia. Haematologica. 2012;97(11):1704–7. doi: 10.3324/haematol.2012.067348.
  14. Tefferi A. Polycythemia vera and essential thrombocythemia: 2012 update on diagnosis, risk stratification, and management. Am J Hematol. 2012;87(3):285–93. doi: 10.1002/ajh.23135.
  15. Johansson PL, Safai-Kutti S, Kutti J. An elevated venous haemoglobin concentration cannot be used as a surrogate marker for absolute erythrocytosis: a study of patients with polycythaemia vera and apparent polycythaemia. Br J Haematol. 2005;129(5):701–5. doi: — 10.1111/j.1365-2141.2005.05517.x.
  16. Thiele J, Kvasnicka HM, Orazi A, et al. Polycythaemia vera. In: Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008. pp. 40–43.
  17. Silver RT, Chow W, Orazi A, et al. Evaluation of WHO criteria for diagnosis of polycythemia vera: a prospective analysis. Blood. 2013;122(11):1881–6. doi: 10.1182/blood-2013-06-508416.
  18. Berk PD, Wasserman LR, Fruchtman SM, et al. Treatment of polycythemia vera: a summary of clinical trials conducted by the polycythemia vera study group. In: Wasserman LR, Berk PD, Berlin NI, eds. Polycythemia Vera and the Myeloproliferative Disorders. Philadelphia: WB Saunders; 1995. pp. 166–94.
  19. McMullin MF, Bareford D, Campbell P, et al. Guidelines for the diagnosis, investigation and management of polycythaemia/erythrocytosis. Br J Haematol. 2005;130(2):174–95. doi: 10.1111/j.1365-2141.2005.05535.x.
  20. McMullin MF, Reilly JT, Campbell P, et al. Amendment to the guideline for diagnosis and investigation of polycythaemia/erythrocytosis. Br J Haematol. 2007;138(6):821–2. doi: 10.1111/j.1365-2141.2007.06741.x.
  21. McMullin MF. The classification and diagnosis of erythrocytosis. Int J Lab Hematol. 2008;30(6):447–59. doi: 10.1111/j.1751-553X.2008.01102.x.
  22. Shih LY, Lee CT. Identification of masked polycythemia vera from patients with idiopathic marked thrombocytosis by endogenous erythroid colony assay. Blood. 1994;83(3):744–8.
  23. Thiele J, Kvasnicka HM, Diehl V. Initial (latent) polycythaemia vera with thrombocytosis mimicking essential thrombocythaemia. Acta Haematol. 2005;113(4):213–9. doi: 10.1159/000084673.
  24. Thiele J, Kvasnicka HM. Diagnostic impact of bone marrow histopathology in polycythemia vera (PV). Histol Histopathol. 2005;20:317–28.
  25. Kvasnicka HM, Thiele J. Prodromal myeloproliferative neoplasms: The 2008 WHO classification. Am J Hematol. 2010;85(1):62–9. doi: 10.1002/ajh.21543.
  26. Barbui T, Thiele J, Gisslinger H, et al. Masked polycythemia vera (mPV): results of an international study. Am J Hematol. 2014;89(1):52–4. doi: 10.1002/ajh.23585.
  27. Barbui T, Thiele J, Carobbio A, et al. Discriminating between essential thrombocythemia and masked polycythemia vera in JAK2 mutated patients. Am J Hematol. 2014;89(6):588–90. doi: 10.1002/ajh.23694.
  28. Barbui T, Thiele J, Vannucchi AM, et al. Rethinking the diagnostic criteria of polycythemia vera. Leukemia. 2014;28(6):1191–5. doi: 10.1038/leu.2013.380.
  29. Barbui T, Thiele J, Carobbio A, et al. Masked polycythemia vera diagnosed according to WHO and BCSH classification. Am J Hematol. 2014;89(2):199–202. doi: 10.1002/ajh.23617.
  30. Chu D, Cho Y-U, Jang S, et al. Straightforward Identification of Masked Polycythemia Vera Based on Proposed Revision of World Health Organization Diagnostic Criteria for BCR-ABL1-Negative Myeloproliferative Neoplasms. Ann Lab Med. 2015;35(6):651–3. doi: 10.3343/alm.2015.35.6.651.
  31. Barbui T, Thiele J, Vannucchi AM, Tefferi A. Rationale for revision and proposed changes of the WHO diagnostic criteria for polycythemia vera, essential thrombocythemia and primary myelofibrosis. Blood Cancer J. 2015;5(8):e337. doi: 10.1038/bcj.2015.64.
  32. Harrison CN, Campbell PJ, Buck G, et al. United Kingdom Medical Research Council Primary Thrombocythemia 1 Study. Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N Engl J Med. 2005;353(1):33–45.
  33. Gisslinger H, Gotic M, Holowiecki J, et al. Anagrelide compared with hydroxyurea in WHO-classified essential thrombocythemia: the ANAHYDRET Study, a randomized controlled trial. Blood. 2013;121(10):1720–8. doi: 10.1182/blood-2012-07-443770.
  34. Thiele J, Kvasnicka HM. Chronic myeloproliferative disorders with thrombocythemia: a comparative study of two classification systems (PVSG, WHO) on 839 patients. Ann Hematol. 2003;82(3):148–52.
  35. Ковригина А.М., Байков В.В. Патоморфологическая дифференциальная диагностика первичного миелофиброза. Учебное пособие для врачей-патологоанатомов. М., 2014. 64 с.
    [Kovrigina AM, Baikov VV. Patomorfologicheskaya differentsial’naya diagnostika pervichnogo mielofibroza. Uchebnoe posobie dlya vrachei-patologoanatomov. (Pathomorphological differential diagnosis of primary myelofibrosis. Manual for pathologists.) Moscow; 2014. 64 p. (In Russ)]
  36. Thiele J, Kvasnicka HM, Zankovich R, Diehl V. The value of bone marrow histology in differentiating between early stage polycythemia vera and secondary (reactive) polycythemias. Haematologica. 2001;86(4):368–74.
  37. Gianelli U, Iurlo A, Vener C. et al. The Significance of Bone Marrow Biopsy and JAK2V617F Mutation in the Differential Diagnosis Between the “Early” Prepolycythemic Phase of Polycythemia Vera and Essential Thrombocythemia. Am J Clin Pathol. 2008;130(3):336–42. doi: 10.1309/6BQ5K8LHVYAKUAF4.

Вирус Эпштейна—Барр и классическая лимфома Ходжкина

В.Э. Гурцевич

ФГБУ «Российский онкологический научный центр им. Н.Н. Блохина» Минздрава России, Каширское ш., д. 24, Москва, Российская Федерация, 115478

Для переписки: Владимир Эдуардович Гурцевич, д-р мед. наук, профессор, Каширское ш., д. 24, Москва, Российская Федерация, 115478; тел.: +7(499)324-25-64; e-mail: gurvlad532@yahoo.com

Для цитирования: Гурцевич В.Э. Вирус Эпштейна—Барр и классическая лимфома Ходжкина. Клиническая онкогематология. 2016;9(2):101–14.

DOI: 10.21320/2500-2139-2016-9-2-101-114


РЕФЕРАТ

Среди онкогенных вирусов человека вирус Эпштейна—Барр (ВЭБ) обращает на себя внимание уникальными свойствами. Этот широко распространенный среди населения планеты вирус одновременно является лидером по числу ассоциированных с ним различных доброкачественных и злокачественных новообразований лимфоидного и эпителиального происхождения. Онкогенный потенциал ВЭБ связан с его способностью инфицировать и трансформировать лимфоциты человека. В тех случаях, когда взаимодействие между размножением ВЭБ, его латентным состоянием и иммунным контролем со стороны организма нарушается, создаются условия для длительной пролиферации инфицированных ВЭБ лимфоцитов и их злокачественной трансформации. По мнению ряда исследователей, молекулярные механизмы связанного с ВЭБ канцерогенеза обусловлены способностью вирусного генома стимулировать экспрессию серии продуктов, имитирующих ряд факторов роста, транскрипции и оказывающих антиапоптотическое действие. Эти кодируемые ВЭБ продукты нарушают сигнальные пути, которые регулируют различные клеточные функции гомеостаза, наделяя клетку способностью к неограниченной пролиферации. Тем не менее точный механизм, с помощью которого ВЭБ инициирует онкогенез, остается не до конца выясненным. В обзоре приводится обобщающая информация о структуре и онкогенном потенциале ВЭБ, морфологических и клинических вариантах лимфомы Ходжкина (ЛХ), а также роли ВЭБ в патогенезе связанных с этим вирусом вариантов ЛХ. Кроме того, в обзоре освещены последние данные об использовании уровня вирусной ДНК ВЭБ в плазме больных ЛХ в качестве биомаркера, отражающего эффективность проведенного лечения и прогноз болезни.


Ключевые слова: вирус Эпштейна—Барр, ВЭБ, латентный мембранный белок 1, LMP1, лимфома Ходжкина, копии ДНК ВЭБ.

Получено: 5 февраля 2016 г.

Принято в печать: 8 февраля 2016 г.

Читать статью в PDF pdficon


ЛИТЕРАТУРА

  1. Zur Hausen H, de Villiers EM. Reprint of: cancer “causation” by infections—individual contributions and synergistic networks. Semin Oncol. 2015;42(2):207–22. doi: 10.1053/j.seminoncol.2015.02.019.
  2. Santos-Juanes J, Fernandez-Vega I, Fuentes N, et al. Merkel cell carcinoma and Merkel cell polyomavirus: a systematic review and meta-analysis. Br J Dermatol. 2015;173(1):42–9. doi: 10.1111/bjd.13870.
  3. Rickinson AB, Young LS, Rowe M. Influence of the Epstein-Barr virus nuclear antigen EBNA 2 on the growth phenotype of virus-transformed B cells. J Virol. 1987;61(5):1310–7.
  4. Rickinson AB, Long HM, Palendira U, et al. Cellular immune controls over Epstein-Barr virus infection: new lessons from the clinic and the laboratory. Trends Immunol. 2014;35(4):159–69. doi: 10.1016/j.it.2014.01.003.
  5. Woodman CB, Collins SI, Vavrusova N, et al. Role of sexual behavior in the acquisition of asymptomatic Epstein-Barr virus infection: a longitudinal study. Pediatr Infect Dis J. 2005;24(6):498–502. doi: 10.1097/01.inf.0000164709.40358.b6.
  6. Henle G, Henle W, Diehl V. Relation of Burkitt’s tumor-associated herpes-type virus to infectious mononucleosis. Proc Natl Acad Sci USA. 1968;59(1):94–101. doi: 10.1073/pnas.59.1.94.
  7. Tanner J, Weis J, Fearon D, et al. Epstein-Barr virus gp350/220 binding to the B lymphocyte C3d receptor mediates adsorption, capping, and endocytosis. Cell. 1987;50(2):203–13. doi:10.1016/0092-8674(87)90216-9.
  8. Connolly SA, Jackson JO, Jardetzky TS, et al. Fusing structure and function: a structural view of the herpesvirus entry machinery. Nat Rev Microbiol. 2011;9(5):369–81. doi: 10.1038/nrmicro2548.
  9. Janz A, Oezel M, Kurzeder C, et al. Infectious Epstein-Barr virus lacking major glycoprotein BLLF1 (gp350/220) demonstrates the existence of additional viral ligands. J Virol. 2000;74(21):10142–52. doi: 10.1128/jvi.74.21.10142-10152.2000.
  10. Ogembo JG, Kannan L, Ghiran I, et al. Human complement receptor type 1/CD35 is an Epstein-Barr Virus receptor. Cell Rep. 2013;3(2):371–85. doi:10.1016/j.celrep.2013.01.023.
  11. Kempkes B, Robertson ES. Epstein-Barr virus latency: current and future perspectives. Curr Opin Virol. 2015;14:138–44. doi: 10.1016/j.coviro.2015.09.007.
  12. Sample J, Kieff E. Transcription of the Epstein-Barr virus genome during latency in growth-transformed lymphocytes. J Virol. 1990;64(4):1667–74.
  13. Babcock GJ, Decker LL, Volk M, et al. EBV persistence in memory B cells in vivo. Immunity. 1998;9(3):395–404. doi: 10.1016/S1074-7613(00)80622-6.
  14. Shannon-Lowe C, Adland E, Bell AI, et al. Features distinguishing Epstein-Barr virus infections of epithelial cells and B cells: viral genome expression, genome maintenance, and genome amplification. J Virol. 2009;83(15):7749–60. doi: 10.1128/JVI.00108-09.
  15. Rickinson A. Epstein-Barr virus. Virus Res. 2002;82(1–2):109–13. doi: 10.1016/s0168-1702(01)00436-1.
  16. Rowe M, Lear AL, Croom-Carter D, et al. Three pathways of Epstein-Barr virus gene activation from EBNA1-positive latency in B lymphocytes. J Virol. 1992;66(1):122–31.
  17. Portis T, Dyck P, Longnecker R. Epstein-Barr Virus (EBV) LMP2A induces alterations in gene transcription similar to those observed in Reed-Sternberg cells of Hodgkin lymphoma. Blood. 2003;102(12):4166–78. doi: 10.1182/blood-2003-04-1018.
  18. Sample J, Young L, Martin B, et al. Epstein-Barr virus types 1 and 2 differ in their EBNA-3A, EBNA-3B, and EBNA-3C genes. J Virol. 1990;64(9):4084–92.
  19. Sixbey JW, Shirley P, Chesney PJ, et al. Detection of a second widespread strain of Epstein-Barr virus. The Lancet. 1989;2(8666):761–5. doi: 10.1016/s0140-6736(89)90829-5.
  20. Gratama JW, Ernberg I. Molecular epidemiology of Epstein-Barr virus infection. Adv Cancer Res. 1995;67:197–255. doi: 10.1016/s0065-230x(08)60714-9.
  21. Young LS, Dawson CW, Eliopoulos AG. The expression and function of Epstein-Barr virus encoded latent genes. Mol Pathol. 2000;53(5):238–47. doi: 10.1136/mp.53.5.238.
  22. Mosialos G, Birkenbach M, Yalamanchili R, et al. The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell. 1995;80(3):389–99. doi: 10.1016/0092-8674(95)90489-1.
  23. Nitta T, Chiba A, Yamashita A, et al. NF-kappaB is required for cell death induction by latent membrane protein 1 of Epstein-Barr virus. Cell Signal. 2003;15(4):423–33. doi: 10.1016/S0898-6568(02)00141-9.
  24. Aviel S, Winberg G, Massucci M, Ciechanover A. Degradation of the Epstein-Barr virus latent membrane protein 1 (LMP1) by the ubiquitin-proteasome pathway. Targeting via ubiquitination of the N-terminal residue. J Biol Chem. 2000;275(31):23491–9. doi: 10.1074/jbc.M002052200.
  25. Gires O, Kohlhuber F, Kilger E, et al. Latent membrane protein 1 of Epstein-Barr virus interacts with JAK3 and activates STAT proteins. EMBO J. 1999;18(11):3064–73. doi: 10.1093/emboj/18.11.3064.
  26. Bentz GL, Whitehurst CB, Pagano JS. Epstein-Barr virus latent membrane protein 1 (LMP1) C-terminal-activating region 3 contributes to LMP1-mediated cellular migration via its interaction with Ubc9. J Virol. 2011;85(19):10144–53. doi: 10.1128/JVI.05035-11.
  27. Wang D, Liebowitz D, Kieff E. An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells. Cell. 1985;43(3):831–40. doi: 10.1016/0092-8674(85)90256-9.
  28. Dawson CW, Port RJ, Young LS. The role of the EBV-encoded latent membrane proteins LMP1 and LMP2 in the pathogenesis of nasopharyngeal carcinoma (NPC). Semin Cancer Biol. 2012;22(2):144–53. doi: 10.1016/j.semcancer.2012.01.004.
  29. Смирнова К.В., Дидук С.В., Сенюта Н.Б., Гурцевич В.Э. Молекулярно-биологические свойства гена LMP1 вируса Эпштейна—Барр: структура, функции и полиморфизм. Вопросы вирусологии. 2015;60(3):5–13.
    [Smirnova KV, Diduk SV, Senyuta NB, Gurtsevich VE. Molecular biological properties of the Epstein-Barr virus LMP1 gene: structure, function, and polymorphism. Voprosy virusologii. 2015;60(3):5–13. (In Russ)]
  30. Vockerodt M, Morgan SL, Kuo M, et al. The Epstein-Barr virus oncoprotein, latent membrane protein-1, reprograms germinal centre B cells towards a Hodgkin’s Reed-Sternberg-like phenotype. J Pathol. 2008;216(1):83–92. doi: 10.1002/path.2384.
  31. Raab-Traub N. Epstein-Barr virus in the pathogenesis of NPC. Semin Cancer Biol. 2002;12:431–41. doi: 10.1016/s1044579x0200086x.
  32. Raab-Traub N. Novel mechanisms of EBV-induced oncogenesis. Curr Opin Virol. 2012;2(4):453–8. doi: 10.1016/j.coviro.2012.07.001.
  33. Soni V, Cahir-McFarland E, Kieff E. LMP1 TRAFficking activates growth and survival pathways. Adv Exp Med Biol. 2007;597:173–87. doi: 10.3390/v5041131.
  34. Man C, Rosa J, Lee LT, et al. Latent membrane protein 1 suppresses RASSF1A expression, disrupts microtubule structures and induces chromosomal aberrations in human epithelial cells. Oncogene. 2007;26(21):3069–80. doi: 10.1038/sj.onc.1210106.
  35. Guo L, Tang M, Yang L, et al. Epstein-Barr virus oncoprotein LMP1 mediates surviving upregulation by p53 contributing to G1/S cell cycle progression in nasopharyngeal carcinoma. Int J Mol Med. 2012;29(4):574–80. doi: 10.3892/ijmm.2012.889.
  36. Horikawa T, Yoshizaki T, Kondo S, et al. Epstein-Barr Virus latent membrane protein 1 induces Snail and epithelial-mesenchymal transition in metastatic nasopharyngeal carcinoma. Br J Cancer. 2011;104(7):1160–7. doi: 10.1038/bjc.2011.38.
  37. Xiao L, Hu ZY, Dong X, et al. Targeting Epstein-Barr virus oncoprotein LMP1-mediated glycolysis sensitizes nasopharyngeal carcinoma to radiation therapy. Oncogene. 2014;33(37):4568–78. doi: 10.1038/onc.2014.32.
  38. Sun W, Liu DB, Li WW, et al. Interleukin-6 promotes the migration and invasion of nasopharyngeal carcinoma cell lines and upregulates the expression of MMP-2 and MMP-9. Int J Oncol. 2014;44(5):1551–60. doi: 10.3892/ijo.2014.2323.
  39. Tzellos S, Farrell PJ. Epstein-Barr virus sequence variation-biology and disease. Pathogens. 2012;1(2):156–74. doi: 10.3390/pathogens1020156.
  40. Walling DM, Shebib N, Weaver SC, et al. The molecular epidemiology and evolution of Epstein-Barr virus: sequence variation and genetic recombination in the latent membrane protein-1 gene. J Infect Dis. 1999;179(4):763–74. doi: 10.1086/314672.
  41. Hu LF, Zabarovsky ER, Chen F, et al. Isolation and sequencing of the Epstein-Barr virus BNLF-1 gene (LMP1) from a Chinese nasopharyngeal carcinoma. J Gen Virol. 1991;72(Pt 10):2399–409. doi: 10.1099/0022-1317-72-10-2399.
  42. Nitta T, Chiba A, Yamamoto N, et al. Lack of cytotoxic property in a variant of Epstein-Barr virus latent membrane protein-1 isolated from nasopharyngeal carcinoma. Cell Signal. 2004;16(9):1071–81. doi: 10.1016/s0898-6568(04)00032-4.
  43. da Costa VG, Marques-Silva AC, Moreli ML. The Epstein-Barr virus latent membrane protein-1 (LMP1) 30-bp deletion and XhoI-polymorphism in nasopharyngeal carcinoma: a meta-analysis of observational studies. Syst Rev. 2015;4(1):46. doi: 10.1186/s13643-015-0037-z.
  44. Rowe M, Peng-Pilon M, Huen DS, et al. Upregulation of bcl-2 by the Epstein-Barr virus latent membrane protein LMP1: a B-cell-specific response that is delayed relative to NF-kappaB activation and to induction of cell surface markers. J Virol. 1994;68(9):5602–12.
  45. Trivedi P, Hu LF, Chen F, et al. Epstein-Barr virus (EBV)-encoded membrane protein LMP1 from a nasopharyngeal carcinoma is non-immunogenic in a murine model system, in contrast to a B cell-derived homologue. Eur J Cancer. 1994;30(1):84–8. doi: 10.1016/s0959-8049(05)80024-3.
  46. Knecht H, Bachmann E, Brousset P, et al. Deletions within the LMP1 oncogene of Epstein-Barr virus are clustered in Hodgkin’s disease and identical to those observed in nasopharyngeal carcinoma. Blood. 1993;82(10):2937–42.
  47. Miller WE, Edwards RH, Walling DM, et al. Sequence variation in the Epstein-Barr virus latent membrane protein 1. J Gen Virol. 1994;75(Pt 10):2729–40. doi: 10.1099/0022-1317-75-10-2729.
  48. Weiss LM. Epstein-Barr virus and Hodgkin’s disease. Curr Oncol Rep. 2000;2(2):199–204. doi: 10.1007/s11912-000-0094-9.
  49. Ковригина А.М., Пробатова Н.А. Лимфома Ходжкина и крупноклеточные лимфомы. М.: МИА, 2007.
    [Kovrigina AM, Probatova NA. Limfoma Khodzhkina i krupnokletochnye limfomy. (Hodgkin’s lymphomas and large cell lymphomas.) Moscow: MIA Publ.; 2007. (In Russ)]
  50. Клиническая онкогематология: Руководство для врачей, 2-е изд. Под ред. М.А. Волковой. М.: Медицина, 2007.
    [Volkova MA, ed. Klinicheskaya  onkogematologiya: Rukovodstvo dlya vrachei. (Clinical oncohematology: manual for physicians.) 2nd edition. Moscow: Meditsina Publ.; 2007. (In Russ)]
  51. Dorsett Y, Robbiani DF, Jankovic M, et al. A role for AID in chromosome translocations between c-myc and the IgH variable region. J Exp Med. 2007;204(9):2225–32. doi: 10.1084/jem.20070884.
  52. Stein H. Hodgkin lymphoma – introduction. In: Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008. pp. 321–34.
  53. Diehl V, Stein H, Hummel M, et al. Hodgkin’s lymphoma: biology and treatment strategies for primary, refractory, and relapsed disease. Hematology Am Soc Hematol Educ Program. 2003;1:225–47. doi: 10.1182/asheducation-2003.1.225.
  54. Chapman AL, Rickinson AB. Epstein-Barr virus in Hodgkin’s disease. Ann Oncol. 1998;9(Suppl 5):S5–16. doi: 10.1093/annonc/9.suppl_5.s5.
  55. Deacon EM, Pallesen G, Niedobitek G, et al. Epstein-Barr virus and Hodgkin’s disease: transcriptional analysis of virus latency in the malignant cells. J Exp Med. 1993;177(2):339–49. doi: 10.1084/jem.177.2.339.
  56. Kuppers R, Rajewsky K, Zhao M, et al. Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proc Natl Acad Sci USA. 1994;91(23):10962–6. doi: 10.1073/pnas.91.23.10962.
  57. Thomas RK, Re D, Wolf J, et al. Part I: Hodgkin’s lymphoma—molecular biology of Hodgkin and Reed-Sternberg cells. Lancet Oncol. 2004;5(1):11–8. doi: 10.1016/S1470-2045(03)01319-6.
  58. Cartwright RA, Watkins G. Epidemiology of Hodgkin’s disease: a review. Hematol Oncol. 2004;22(1):11–26. doi: 10.1002/hon.723.
  59. Jarrett AF, Armstrong AA, Alexander E. Epidemiology of EBV and Hodgkin’s lymphoma. Ann Oncol. 1996;7(Suppl 4):5–10. doi: 10.1093/annonc/7.suppl_4.s5.
  60. Glaser SL, Lin RJ, Stewart SL, et al. Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70(4):375–82. doi: 10.1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.0.co;2-t.
  61. Cader FZ, Kearns P, Young L, et al. The contribution of the Epstein-Barr virus to the pathogenesis of childhood lymphomas. Cancer Treat Rev. 2010;36(4):348–53. doi: 10.1016/j.ctrv.2010.02.011.
  62. Jarrett RF, Gallagher A, Jones DB, et al. Detection of Epstein-Barr virus genomes in Hodgkin’s disease: relation to age. J Clin Pathol. 1991;44(10):844–8. doi: 10.1136/jcp.44.10.844.
  63. Armstrong AA, Alexander FE, Cartwright R, et al. Epstein-Barr virus and Hodgkin’s disease: further evidence for the three disease hypothesis. Leukemia. 1998;12(8):1272–6. doi: 10.1038/sj.leu.2401097.
  64. Oyama T, Ichimura K, Suzuki R, et al. Senile EBV+ B-cell lymphoproliferative disorders: a clinicopathologic study of 22 patients. Am J Surg Pathol. 2003;27(1):16–26. doi: 10.1097/00000478-200301000-00003.
  65. Oyama T, Yamamoto K, Asano N, et al. Age-related EBV-associated B-cell lymphoproliferative disorders constitute a distinct clinicopathologic group: a study of 96 patients. Clin Cancer Res. 2007;13(17):5124–32. doi: 10.1158/1078-0432.ccr-06-2823.
  66. Thorley-Lawson DA, Gross A. Persistence of the Epstein-Barr virus and the origins of associated lymphomas. N Engl J Med. 2004;350(13):1328–37. doi: 10.1056/NEJMra032015.
  67. Kuppers R. Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer. 2005;5(4):251–62. doi: 10.1038/nrc1589.
  68. Klein U, Dalla-Favera R. Germinal centres: role in B-cell physiology and malignancy. Nat Rev Immunol. 2008;8(1):22–33. doi: 10.1038/nri2217.
  69. Caldwell RG, Wilson JB, Anderson SJ, et al. Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals. Immunity. 1998;9(3):405–11. doi: 10.1016/s1074-7613(00)80623-8.
  70. Gires O, Zimber-Strobl U, Gonnella R, et al. Latent membrane protein 1 of Epstein-Barr virus mimics a constitutively active receptor molecule. EMBO J. 1997;16(20):6131–40. doi: 10.1093/emboj/18.11.3064.
  71. Laichalk LL, Thorley-Lawson DA. Terminal differentiation into plasma cells initiates the replicative cycle of Epstein-Barr virus in vivo. J Virol. 2005;79(2):1296–307. doi: 10.1128/JVI.79.2.1296-1307.2005.
  72. Alexander FE, Jarrett RF, Lawrence D, et al. Risk factors for Hodgkin’s disease by Epstein-Barr virus (EBV) status: prior infection by EBV and other agents. Br J Cancer. 2000;82(5):1117–21. doi: 10.1054/bjoc.1999.1049.
  73. Mueller N, Evans A, Harris NL, et al. Hodgkin’s disease and Epstein-Barr virus. Altered antibody pattern before diagnosis. N Engl J Med. 1989;320(11):689–95. doi: 10.1056/nejm198903163201103.
  74. Weiss LM, Strickler JG, Warnke RA, et al. Epstein-Barr viral DNA in tissues of Hodgkin’s disease. Am J Pathol. 1987;129(1):86–91.
  75. Anagnostopoulos I, Herbst H, Niedobitek G, et al. Demonstration of monoclonal EBV genomes in Hodgkin’s disease and Ki-1-positive anaplastic large cell lymphoma by combined Southern blot and in situ hybridization. Blood. 1989;74(2):810–6.
  76. Re D, Kuppers R, Diehl V. Molecular pathogenesis of Hodgkin’s lymphoma. J Clin Oncol. 2005;23(26):6379–86. doi: 10.1200/JCO.2005.55.013.
  77. Mancao C, Altmann M, Jungnickel B, et al. Rescue of “crippled” germinal center B cells from apoptosis by Epstein-Barr virus. Blood. 2005;106(13):4339–44. doi: 10.1182/blood-2005-06-2341.
  78. Chaganti S, Bell AI, Pastor NB, et al. Epstein-Barr virus infection in vitro can rescue germinal center B cells with inactivated immunoglobulin genes. Blood. 2005;106(13):4249–52. doi: 10.1182/blood-2005-06-2327.
  79. Kapatai G, Murray P. Contribution of the Epstein Barr virus to the molecular pathogenesis of Hodgkin lymphoma. J Clin Pathol. 2007;60(12):1342–9. doi: 10.1136/jcp.2007.050146.
  80. Kuppers R. B cells under influence: transformation of B cells by Epstein-Barr virus. Nat Rev Immunol. 2003;3(10):801–12. doi: 10.1038/nri1201.
  81. Huen DS, Henderson SA, Croom-Carter D, et al. The Epstein-Barr virus latent membrane protein-1 (LMP1) mediates activation of NF-kappa B and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain. Oncogene. 1995;10:549–60.
  82. Kieser A, Kilger E, Gires O, et al. Epstein-Barr virus latent membrane protein-1 triggers AP-1 activity via the c-Jun N-terminal kinase cascade. EMBO J. 1997;16(21):6478–85. doi: 10.1093/emboj/16.21.6478.
  83. Kube D, Holtick U, Vockerodt M, et al. STAT3 is constitutively activated in Hodgkin cell lines. Blood. 2001;98(3):762–70. doi: 10.1182/blood.V98.3.762.
  84. Dutton A, Reynolds GM, Dawson CW, et al Constitutive activation of phosphatidyl-inositide 3 kinase contributes to the survival of Hodgkin’s lymphoma cells through a mechanism involving Akt kinase and mTOR. J Pathol. 2005;205(4):498–506. doi: 10.1002/path.1725.
  85. Brielmeier M, Mautner J, Laux G, et al. The latent membrane protein 2 gene of Epstein-Barr virus is important for efficient B cell immortalization. J Gen Virol. 1996;77(Pt 11):2807–18. doi: 10.1099/0022-1317-77-11-2807.
  86. Casola S, Otipoby KL, Alimzhanov M, et al. B cell receptor signal strength determines B cell fate. Nat Immunol. 2004;5(3):317–27. doi: 10.1038/ni1036.
  87. Engels N, Yigit G, Emmerich CH, et al. Epstein-Barr virus LMP2A signaling in statu nascendi mimics a B cell antigen receptor-like activation signal. Cell Commun Signal. 2012;10(1):9. doi: 10.1186/1478-811X-10-9.
  88. Portis T, Dyck P, Longnecker R. Epstein-Barr Virus (EBV) LMP2A induces alterations in gene transcription similar to those observed in Reed-Sternberg cells of Hodgkin lymphoma. Blood. 2003;102(12):4166–78. doi: 10.1182/blood-2003-04-1018.
  89. Portis T, Longnecker R. Epstein-Barr virus (EBV) LMP2A mediates B-lymphocyte survival through constitutive activation of the Ras/PI3K/Akt pathway. Oncogene. 2004;23(53):8619–28. doi: 10.1038/sj.onc.1207905.
  90. Farrell K, Jarrett RF. The molecular pathogenesis of Hodgkin lymphoma. Histopathology. 2011;58(1):15–25. doi: 10.1111/j.1365-2559.2010.03705.x.
  91. Herbst H, Foss HD, Samol J, et al. Frequent expression of interleukin-10 by Epstein-Barr virus-harboring tumor cells of Hodgkin’s disease. Blood. 1996;87:2918–29.
  92. Hsu SM, Lin J, Xie SS, et al. Abundant expression of transforming growth factor-beta 1 and -beta 2 by Hodgkin’s Reed-Sternberg cells and by reactive T lymphocytes in Hodgkin’s disease. Hum Pathol. 1993;24(3):249–55. doi: 10.1016/0046-8177(93)90034-e.
  93. Kapp U, Yeh WC, Patterson B, et al. Interleukin 13 is secreted by and stimulates the growth of Hodgkin and Reed-Sternberg cells. J Exp Med. 1999;189(12):1939–46. doi: 10.1084/jem.189.12.1939.
  94. Munz C, Moormann A. Immune escape by Epstein-Barr virus associated malignancies. Semin Cancer Biol. 2008;18(6):381–7. doi: 10.1016/j.semcancer.2008.10.002.
  95. Lichtenstein AV, Melkonyan HS, Tomei LD, et al. Circulating nucleic acids and apoptosis. Ann NY Acad Sci. 2001;945(1):239–49. doi: 10.1111/j.1749-6632.2001.tb03892.x.
  96. Sidransky D. Emerging molecular markers of cancer. Nat Rev Cancer. 2002;2(3):210–9. doi: 10.1038/nrc755.
  97. Skvortsova TE, Rykova EY, Tamkovich SN, et al. Cell-free and cell-bound circulating DNA in breast tumours: DNA quantification and analysis of tumour-related gene methylation. Br J Cancer. 2006;94(10):1492–5. doi: 10.1038/sj.bjc.6603117.
  98. Lo YM, Chan LY, Lo KW, et al. Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res. 1999;59(6):1188–91.
  99. Hou X, Zhao C, Guo Y, et al. Different Clinical Significance of Pre- and Post-treatment Plasma Epstein-Barr Virus DNA Load in Nasopharyngeal Carcinoma Treated with Radiotherapy. Clin Oncol. (R Coll Radiol) 2011;23(2):128–33. doi: 10.1016/j.clon.2010.09.001.
  100. Wang WY, Twu CW, Chen HH, et al. Plasma EBV DNA clearance rate as a novel prognostic marker for metastatic/recurrent nasopharyngeal carcinoma. Clin Cancer Res. 2010;16(3):1016–24. doi: 10.1158/1078-0432.ccr-09-2796.
  101. Lo YM, Chan AT, Chan LY, et al. Molecular prognostication of nasopharyngeal carcinoma by quantitative analysis of circulating Epstein-Barr virus DNA. Cancer Res. 2000;60:6878–81.
  102. Lo YM, Chan LY, Chan AT, et al. Quantitative and temporal correlation between circulating cell-free Epstein-Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res. 1999;59:5452–5.
  103. Au WY, Pang A, Choy C, et al. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104(1):243–9. doi: 10.1182/blood-2003-12-4197.
  104. Wang ZY, Liu QF, Wang H, et al. Clinical implications of plasma Epstein-Barr virus DNA in early-stage extranodal nasal-type NK/T-cell lymphoma patients receiving primary radiotherapy. Blood. 2012;120(10):2003–10. doi: 10.1182/blood-2012-06-435024.
  105. Kasamon YL, Jacene HA, Gocke CD, et al. Phase 2 study of rituximab-ABVD in classical Hodgkin lymphoma. Blood. 2012;119(18):4129–32. doi: 10.1182/blood-2012-01-402792.
  106. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121(18):3547–53. doi: 10.1182/blood-2012-09-454694.
  107. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17(9):2885–92. doi: 10.1158/1078-0432.ccr-10-3327.
  108. Dinand V, Sachdeva A, Datta S, et al. Plasma Epstein Barr Virus (EBV) DNA as a Biomarker for EBV associated Hodgkin lymphoma. Indian Pediatr. 2015;52(8):681–5. doi: 10.1007/s13312-015-0696-9.
  109. Vockerodt М, Yap L-F, Shannon-Lowe C, et al. The Epstein-Barr virus and the pathogenesis of lymphoma. J Pathol. 2015;235(2):312–22. doi: 10.1002/path.4459.
  110. Grywalska E, Markowicz J, Grabarczyk P, et al. Epstein-Barr virus-associated lymphoproliferative disorders. Postepy Hig Med Dosw (Online). 2013;67:481–90. doi 10.5604/17322693.1050999.

Метаболизм железа в норме и при патологии

Е.А. Лукина, А.В. Деженкова

ФГБУ «Гематологический научный центр» Минздрава России, Новый Зыковский пр-д, д. 4а, Москва, Российская Федерация, 125167

Для переписки: Елена Алексеевна Лукина, д-р мед. наук, профессор, Новый Зыковский пр-д, д. 4а, Москва, Российская Федерация, 125167; тел.: +7(495)612-09-23; e-mail: elenalukina02@gmail.com

Для цитирования: Лукина Е.А., Деженкова А.В. Метаболизм  железа в норме и при патологии. Клиническая онкогематология. 2015;8(4):355–361.

DOI: 10.21320/2500-2139-2015-8-4-355-361


РЕФЕРАТ

В обзоре изложены современные представления о физиологической и патологической роли железа, а также основных механизмах регуляции метаболизма железа в организме человека. В последние годы показано, что не только дефицит, но и избыток данного микроэлемента имеют катастрофические последствия для организма, а содержание данного микроэлемента жестко регулируется, что позволяет говорить о гомеостазе железа. Из общего количества железа (3–5 г), содержащегося в организме здорового человека, основная часть входит в состав клеток крови, костного мозга и печени и находится в связанном с белками состоянии, что необходимо для предотвращения цитотоксических эффектов свободных ионов железа. В обзоре приводится краткая информация об основных белках, участвующих в метаболизме железа, и их роли в поддержании гомеостаза данного микроэлемента. Особое внимание уделяется функциональному значению гепсидина, играющего ключевую роль в регуляции внеклеточного содержания железа, и процессам рециркуляции железа. Приводится краткая информация о механизмах развития функционального дефицита железа и его роли в патогенезе анемии хронических заболеваний. Существенное внимание уделяется характеристике состояния перегрузки железом, способам диагностики и средствам коррекции вторичного гемохроматоза.


Ключевые слова: метаболизм железа, ферритин, гепсидин, рециркуляция железа, анемия хронических заболеваний, перегрузка железом.

Получено: 1 июля 2015 г.

Принято в печать: 9 ноября 2015 г.

Читать статью в PDF pdficon


ЛИТЕРАТУРА

  1. Петров В.Н. Физиология и патология обмена железа. Львов: Наука, 1982. 224 c.
    [Petrov VN. Fiziologiya i patologiya obmena zheleza. (Physiology and pathology of iron metabolism.) L’vov: Nauka Publ.; 1982. 224 p. (In Russ)]
  2. Finch CA, Huebers HA. Iron metabolism. Clin Physiol Biochem. 1986;4:5–15.
  3. Richardson DR. Role of iron in cell cycle progression and cellular proliferation. Book of Abstracts. BioIron; 2005:7.
  4. Roetto A, Camaschella C. New insights into iron homeostasis through the study of non-HFE hereditary haemochromatosis. Best Pract Res Clin Haematol. 2005;18:235–50. doi: 10.1016/j.beha.2004.09.004.
  5. Sussman HH. Iron in cancer. Pathobiology. 1992;60:2–9. doi: 10.1159/000163690.
  6. Идельсон Л.И., Воробьев А.И. Железодефицитная анемия. Руководство по гематологии. Под ред. А.И. Воробьева. В 3 томах. М.: Ньюдиамед, 2005. Т. 2. С. 171–90.
    [Idel’son LI, Vorob’ev AI. Iron-deficiency anemia. In: Vorob’ev AI, ed. Rukovodstvo po gematologii. (Manual of Hematology.) In 3 vol. Moscow: Newdiamed Publ.; 2005. Vol. 2. p. 171–90. (In Russ)]
  7. Porter JB. Monitoring and treatment of iron overload: state of the art and new approaches. Sem Hematol. 2005;42(2 Suppl. 1):14–8. doi: 10.1053/j.seminhematol.2005.01.004.
  8. Kuntz E, Kuntz H-D. Haemochromatosis. In: Hepatology – Principles and Practice. Berlin: Springer-Verlag; 2002. p. 556–65.
  9. Ilickstein H, El RB, Shvartsman M, Cabantchik ZY. Intracellular labile iron pools as direct targets of iron chelators: a fluorescence study of chelator action in living cells. Blood. 2005;106:3242–50. doi: 10.1182/blood-2005-02-0460.
  10. Corce V, Renaud St, Cannie I, et al. Tumoral vectorization of new iron chelators for antiproliferative activity: biological properties of polyaminoquinolines. The abstract book of 5th Congress of the International Bioiron Society; 2013. Poster #230.
  11. Guyader CD, Thirouard A-S, Erdtmann L, et al. Liver iron is surrogate marker of severe fibrosis in chronic hepatitis. J Hepatol. 2007;46:587–96. doi: 10.1016/j.jhep.2006.09.021.
  12. Camaschella C. Iron and hepcidin: a story of recycling and balance. Hematol Am Soc Hematol Educ Program. 2013;2013:1–8. doi: 10.1182/asheducation-2013.1.1.
  13. Sikorska K, Romanowski T, Stalke P, et al. Association of Hepcidin mRNA Expression With Hepatocyte Iron Accumulation and Effects of Antiviral Therapy in Chronic Hepatitis C Infection. Hepat Mon. 2014;14(11):e21184. doi:10.5812/hepatmon.21184.
  14. Raha AA, Vaishnav RA, Friedland RP, et al. The systemic iron-regulatory proteins hepcidin and ferroportin are reduced in the brain in Alzheimer’s disease. BMC Neuroscience. 2015;16(1):24. doi: 10.1186/2051-5960-1-55.
  15. Xu X, Pin S, Gathinji M, et al. Aceruloplasminemia: an inherited neurodegenerative disease with impairment of iron homeostais. Ann N Y Acad Sci. 2004;1012:299–305. doi: 10.1196/annals.1306.024.
  16. Moreau C, Devedjian J, Kluza J, et al. Targeting brain chelatable iron as therapeutic strategy for parkinson’s disease. Translational and clinical studies. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #52.
  17. Collingwood J, Finnegan M, Visanji N, et al. Brain iron and MRI in Alzheimer’s disease, Parkinson’s disease, and Multiple System Atrophy. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #67.
  18. Долгов В.В., Луговская С.А., Почтарь М.Е. Лабораторная диагностика нарушений метаболизма железа. СПб.: Vital Diagnostics, 2002. 51 c.
    [Dolgov VV, Lugovskaya SA, Pochtar’ ME. Laboratornaya diagnostika narushenii metabolizma zheleza. (Laboratory diagnosis of impaired iron metabolism.) Saint Petersburg: Vital Diagnostics Publ.; 2002. 51 p. (In Russ)]
  19. Cabantchik ZY, Brener W, Zanninelili G. LPI-labile plasma iron in iron overload. Best Pract Res Clin Haematol. 2005;18:277–87. doi: 10.1016/j.beha.2004.10.003.
  20. Cheng Y, Zak O, Aisen P, et al. Structure of the human transferring receptor-transferrin complex. Cell. 2004;116:483–5. doi: 10.1016/s0092-8674(04)00130-8.
  21. Левина А.А., Андреева А.П., Замчий А.А. Определение концентрации ферритина в сыворотке крови радиоиммунным методом. Гематология и трансфузиология. 1984;5:57–60.
    [Levina AA, Andreeva AP, Zamchii AA. Evaluation of serum ferritin levels using radioimmunoassay technique. Gematologiya i transfuziologiya. 1984;5:57–60. (In Russ)]
  22. Lukina EA, Levina AA, Mokeeva NA. The diagnostic significance of serum ferritin indices in patients with malignant and reactive histiocytoses. Br J Haematol. 1993;83:326–9. doi: 10.1111/j.1365-2141.1993.tb08289.x.
  23. Denz H, Orth B, Huber P, et al. Immune activation and anemia of chronic disorders. Blood. 1993;81:1404–9.
  24. Gunshin H, Mackenzie B, Berger UV, et al. Cloning and characterization of mammalian proton-coupled metal-ion transporter gene. Nature. 1997;388:482–8. doi: 10.1038/41343.
  25. Mims MP, Guan Y, Pospisilova D, et al. Identification of a human mutation of DMT1 in a patient with microcytic anemia and iron overload. Blood. 2005;105(3):1337–42. doi: 10.1182/blood-2004-07-2966.
  26. Napier I, Ponka P, Richardson DR. Iron trafficking in the mitochondrion: novel pathways revealed by disease. Blood. 2005;105:1867–974. doi: 10.1182/blood-2004-10-3856.
  27. D’Angelo G. Role of hepcidin in the pathophysiology and diagnosis of anemia. Blood Res. 2013;48(1):10–5. doi: 10.5045/br.2013.48.1.10.
  28. Hellman N, Gitlin JD. Ceruloplasmin metabolism and function. Ann Rev Nutr. 2002;22:439–58. doi: 10.1146/annurev.nutr.22.012502.114457.
  29. Fuqua B, Darshan D, Frazer D, et al. Severe defects in iron metabolism in mice with double knockout of the multicopper ferroxidases hephaestin and ceruloplasmin. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #24.
  30. Krause A, Neitz S, Magert HJ, et al. LEAP-1, a novel highly disulfide-bonded human peptide, exhibit antimicrobial activity. FEBS Lett. 2000;480(2):147–50. doi: 10.1016/s0014-5793(00)01920-7.
  31. Park CH, Valore EV, Waring AJ, et al. Hepcidin: a urinary antibacterial peptide synthesized in the liver. J Biol Chem. 2001;276:7806–10. doi: 10.1074/jbc.m008922200.
  32. Ganz T. Hepcidin – a regulator of intestinal iron absorption and iron recycling by macrophages. Best Pract Res Clin Haematol. 2005;18:171–82. doi: 10.1016/j.beha.2004.08.020.
  33. Pigeon C, Ilyin G, Courselaud B, et al. A new mouseт liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem. 2001;276(4):7811–9. doi: 10.1074/jbc.m008923200.
  34. Hunter HN, Fulton DB, Vogel HJ. The solution structure of human hepcidin, a antibicrobial activity that is involved in iron uptake and hereditary hemochromatosis. J Biol Chem. 2002;277:37597–603. doi: 10.1074/jbc.m205305200.
  35. Harrison-Findik D, Lu S, Zmijewski E. Regulation of hepcidin transcription by reactive oxygen species and hypoxia. The abstract book of 5th Congress of the International Bioiron Society; 2013. Poster #6.
  36. Luo Q, Cheng Ch, Wang D, et al. Regulation of intracellular iron homeostasis under hypoxia. The abstract book of 5th Congress of the International Bioiron Society; 2013. Poster #166.
  37. Means RT, Krantz SB. Progress in understanding the pathogenesis of anemia of chronic disease. Blood. 1992;80:1639–44.
  38. Gardenghi S, Casu C, Renaud T, et al. Investigating the role of cytokines and hepcidin in anemia of inflammation. The abstract book of 5th Congress of the International Bioiron Society; 2013. Poster #138.
  39. Nairz M, Ferring-Appel D, Schroll A, et al. Iron regulatory proteins mediate macrophage innate immunity against salmonella. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #34.
  40. Kautz L, Nemeth E, Ganz T. The erythroid factor erythroferrone and its role in iron homeostasis. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #30.
  41. Frazer D, Wilkins S, Whitelaw N, et al. Hepcidin-independent iron recycling in a mouse model of haemolytic anaemia. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #32.
  42. Gerhard G, Still Ch, Wood C, et al. Primary hepatic iron overload in extreme obesity is common and not associated with metabolic abnormalities. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #58.
  43. Miyanishi K, Tanaka Sh, Kobune M, et al. Increased hepatic oxidative DNA damage in patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. The abstract book of 5th Congress of the International Bioiron Society; 2013. Poster #227.
  44. Kobune M, Kikuchi S, Iyama S, et al. Iron chelation therapy improves oxidative DNA damage in hematopoietic cells derived from transfusion-dependent myelodysplastic syndrome. The abstract book of 5th Congress of the International Bioiron Society; 2013. Poster #93.
  45. Jones E, Allen A, Evans P, et al. Differences in hepcidin regulation distinguish mild and severe phenotypes of e-beta thalassaemia. The abstract book of 5th Congress of the International Bioiron Society; 2013. Podium #27.

Роль экспрессии c-MYC, BCL2 и BCL6 в патогенезе диффузной В-крупноклеточной лимфомы

А.Е. Мисюрина1, В.А. Мисюрин2, Е.А. Барях1, А.М. Ковригина1, С.К. Кравченко1

1 ФГБУ «Гематологический научный центр» МЗ РФ, Новый Зыковский пр-д, д. 4а, Москва, Российская Федерация, 125167

2 ФГБУ «Российский онкологический научный центр им. Н.Н. Блохина», Каширское ш., д. 24, Москва, Российская Федерация, 115478

Для переписки: А.Е. Мисюрина, аспирант, Новый Зыковский проезд, д. 4а, Москва, Российская Федерация, 125167; тел.: +7(909)637-32-49; e-mail: anna.lukina1@gmail.com

Для цитирования: Мисюрина А.Е., Мисюрин В.А., Барях Е.А., Ковригина А.М., Кравченко С.К. Роль экспрессии генов c-MYC, BCL2 и BCL6 в патогенезе диффузной В-крупноклеточной лимфомы. Клин. онкогематол. 2014; 7(4): 512–521.


РЕФЕРАТ

Согласно современным представлениям, опирающимся на результаты исследования профиля экспрессии генов, существует несколько подтипов диффузной B-крупноклеточной лимфомы (ДBКЛ): из В-клеток герминативного центра и из активированных В-клеток. Гены c-MYC, BCL6 и BCL2 являются ключевыми регуляторами развития В-лимфоцитов на уровне герминальной (фолликулярной) дифференцировки. В патогенезе ДВКЛ наиболее часто играют роль генетические аномалии с их участием. От общего уровня активности и механизмов, приводящих к гиперэкспрессии каждого из этих генов и продукции соответствующих белков, зависит прогноз заболевания. Мы предполагаем, что определение количественных параметров экспрессии генов c-MYC, BCL6 и BCL2, а также кодируемых ими белков позволит с высокой точностью выделять группы риска среди больных ДBКЛ.


Ключевые слова: диффузная В-крупноклеточная лимфома, молекулярные подтипы, группы риска, c-MYC, BCL6, BCL2.

Принято в печать: 8 сентября 2014 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Swerdlow S.H., Campo E., Harris N.L. et al (eds.). WHO Classification of Tumors of Haematopoetic and Lymphoid Tissues. Lyon: IARC, 2008: 233–4.
  2. Frick M., Dorken B., Lenz G. New insights into the biology of molecular subtypes of diffuse large B-cell lymphoma and Burkitt lymphoma. Best Pract. Res. Clin. Haematol. 2012; 25(1): 3–12.
  3. Alizadeh A.A., Eisen M.B., Davis R.E. et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000; 403: 503–11.
  4. Rosenwald A., Wright G., Chan W.C. et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N. Engl. J. Med. 2002; 346(25): 1937–47.
  5. Alizadeh A.A., Eisen M.B., Davis R.E. et al. The lymphochip: a specialized cDNA microarray for the genomic-scale analysis of gene expression in normal and malignant lymphocytes. Cold Spring Harbor Symp. Quant. Biol. 1999; 62: 71–8.
  6. Lenz G., Wright G., Dave S.S. et al. Stromal gene signatures in large-B-cell lymphomas. N. Engl. J. Med. 2008; 359(22): 2313–23.
  7. Rosenwald A., Wright G., Leroy K. et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J. Exp. Med. 2003; 198(6): 851–62.
  8. Savage K.J., Monti S., Kutok J.L. et al. The molecular signature of mediastinal large B-cell lymphoma differs from that of other diffuse large B-cell lymphomas and shares features with classical Hodgkin lymphoma. Blood. 2003; 102(12): 3871–9.
  9. Wright G., Tan B., Rosenwald A. et al. A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc. Natl. Acad. Sci. USA. 2003; 100: 9991–6.
  10. Muller A.M., Medvinsky A., Strouboulis J., Grosveld F., Dzierzak E. Development of hematopoietic stem cell activity in the mouse embryo. Immunity. 1994; 1: 291–301.
  11. Melchers F. The pre-B-cell receptor: selector of fitting immunoglobulin heavy chains for the B-cell repertoire. Nat. Rev. Immunol. 2005; 5: 578–84.
  12. van Zelm M.C., Szczepanski T., van der Burg M., van Dongen J.J. Replication history of B lymphocytes reveals homeostatic proliferation and extensive antigen-induced B cell expansion. J. Exp. Med. 2007; 204: 645–55.
  13. Martin F., Oliver A.M., Kearney J.F. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity. 2001; 14: 617–29.
  14. Chen J., Trounstine M., Alt F.W. et al. Immunoglobulin gene rearrangement in B cell deficient mice generated by targeted deletion of the JH locus. Int. Immunol. 1993; 5: 647–56.
  15. Teng G., Papavasiliou F.N. Immunoglobulin somatic hypermutation. Annu. Rev. Genet. 2007; 41: 107–20.
  16. Liu Y.J., Arpin C. Germinal center development. Immunol. Rev. 1997; 156: 111–26.
  17. Yuan D. Regulation of IgM and IgD synthesis in B lymphocytes. II. Translational and post-translational events. J. Immunol. 1984; 132: 1566–70.
  18. Yasodha N. The Biology of the Germinal Center. ASH Education Book. 2007; 1: 210–5.
  19. Komori T., Okada A., Stewart V., Alt F.W. Lack of N regions in antigen receptor variable region genes of TdT-deficient lymphocytes. Science. 1993; 261: 1171–5.
  20. Willenbrock K., Jungnickel B., Hansmann M.L., Kuppers R. Human splenic marginal zone B cells lack expression of activation-induced cytidine deaminase. Eur. J. Immunol. 2005; 35: 3002–7.
  21. Raghavan S.C., Hsieh C.L., Lieber M.R. Both V(D)J coding ends but neither signal end can recombine at the bcl-2 major breakpoint region, and the rejoining is ligase IV dependent. Mol. Cell. Biol. 2005; 15: 6475–84.
  22. Luscher B. MAD1 and its life as a MYC antagonist: an update. Eur. J. Cell. Biol. 2012; 91(6–7): 506–14.
  23. McDuff F.O., Naud J.F., Montagne M., Sauve S., Lavigne P. The Max homodimeric b-HLH-LZ significantly interferes with the specific heterodimerization between the c-Myc and Max b-HLH-LZ in absence of DNA: a quantitative analysis. J. Mol. Recognit. 2009; 22(4): 261–9.
  24. Dang C.V. MYC on the path to cancer. Cell. 2012; 149(1): 22–35.
  25. Luscher B., Vervoorts J. Regulation of gene transcription by the oncoprotein MYC. Gene. 2012; 494(2): 145–60.
  26. Meyer N., Penn L.Z. Reflecting on 25 years with MYC. Nat. Rev. Cancer. 2008; 8(12): 976–90.
  27. Keller U.B., Old J.B., Dorsey F.C. et al. Myc targets Cks1 to provoke the suppression of p27Kip1, proliferation and lymphoma agenesis. EMBO. J. 2007; 26(10): 2562–74.
  28. Bueno M.J., Malumbres M. MicroRNAs and the cell cycle. Biochim. Biophys. Acta. 2011; 1812(5): 592–601.
  29. Nie Z., Hu G., Wei G. et al. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell. 2012; 151(1): 68–79.
  30. Lin C.Y., Loven J., Rahl P.B. et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell. 2012; 151(1): 56–67.
  31. Lin Y., Wong K., Calame K. Repression of c-myc transcription by Blimp-1, an inducer of terminal B cell differentiation. Science. 1997; 276(5312): 596–9.
  32. Basso K., Dalla-Favera R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv. Immunol. 2010; 105: 193–210.
  33. Phan R.T., Saito M., Basso K., Niu H., Dalla-Favera R. BCL6 interacts with the transcription factor Miz-1 to suppress the cyclin-dependent kinase inhibitor p21 and cell cycle arrest in germinal center B cells. Nat. Immunol. 2005; 6(10): 1054–60.
  34. Niu H., Ye B.H., Dalla-Favera R. Antigen receptor signaling induces MAP kinase-mediated phosphorylation and degradation of the BCL-6 transcription factor. Genes Dev. 1998; 12(13): 1953–61.
  35. Phan R.T., Saito M., Kitagawa Y., Means A.R., Dalla-Favera R. Genotoxic stress regulates expression of the proto-oncogene Bcl6 in germinal center B cells. Nat. Immunol. 2007; 8(10): 1132–9.
  36. Phan R.T., Dalla-Favera R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature. 2004; 432(7017): 635–9.
  37. Basso K., Saito M., Sumazin P. et al. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood. 2010; 115(5): 975–84.
  38. Wagner S.D., Ahearne M., Ko Ferrigno P. The role of BCL6 in lymphomas and routes to therapy. Br. J. Haematol. 2011; 152(1): 3–12.
  39. Basso K., Dalla-Favera R. Roles of BCL6 in normal and transformed germinal center B cells. Immunol. Rev. 2012; 247(1): 172–83.
  40. Merino R., Ding L., Veis D.J. et al. Developmental regulation of the Bcl-2 protein and susceptibility to cell death in B lymphocytes. EMBO. J. 1994; 13: 683–91.
  41. McDonnell T.J., Nunez G., Platt F.M. et al. Deregulated Bcl-2-immunoglobulin transgene expands a resting but responsive immunoglobulin M and D-expressing B-cell population. Mol. Cell. Biol. 1990; 10: 1901–7.
  42. McDonnell T.J., Deane N., Platt F.M. et al. Bcl-2-immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell. 1989; 57: 79–88.
  43. Veis D.J., Sorenson C.M., Shutter J.R. et al. Bcl-2-deficient mice demonstrate fulminant lymphoid apoptosis, polycystic kidneys, and hypopigmented hair. Cell. 1993; 75: 229–40.
  44. Wilson W.H., Teruya-Feldstein J., Fest T. et al. Relationship of p53, bcl-2, and tumor proliferation to clinical drug resistance in non-Hodgkin’s lymphomas. Blood. 1997; 89: 601–9.
  45. Monti S., Savage K.J., Kutok J.L. et al. Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood. 2005; 105(5): 1851–61.
  46. Dent A.L., Shaffer A.L., Yu X. et al. Control of inflammation, cytokine expression, and germinal center formation by BCL-6. Science. 1997; 276(5312): 589–92.
  47. Никитин Е.А. Патогенез зрелоклеточных лимфатических опухолей. Материалы конгрессов и конференций. VIII Российский онкологический конгресс [Электронный документ] (http://www.rosoncoweb.ru/library/ congress/ru/08/19.php). [Nikitin E.A. Pathogenesis of mature cell lymphomas. Materialy kongressov i konferentsii. VIII Rossiiskii onkologicheskii kongress (Materials of congresses and conferences. VIII Russian oncological congress). Available at: http://www. rosoncoweb.ru/library/congress/ru/08/19.php (In Russ.)]
  48. Davis R.E., Brown K.D., Siebenlist U. et al. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J. Exp. Med. 2001; 194(12): 1861–74.
  49. Jost P.J., Ruland J. Aberrant NF-kappaB signaling in lymphoma: mechanisms, consequences, and therapeutic implications. Blood. 2007; 109(7): 2700–7.
  50. Ngo V.N., Davis R.E., Lamy L. et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature. 2006; 441(7089): 106–10.
  51. Rawlings D.J., Sommer K., Moreno-Garcia M.E. The CARMA1 signalosome links the signalling machinery of adaptive and innate immunity in lymphocytes. Nat. Rev. Immunol. 2006; 6(11): 799–812.
  52. Lenz G., Davis R.E., Ngo V.N. et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science. 2008; 319(5870): 1676–9.
  53. Davis R.E., Ngo V.N., Lenz G. et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010; 463(7277): 88–92.
  54. Compagno M., Lim W.K., Grunn A. et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. 2009; 459(7247): 717–21.
  55. Kato M., Sanada M., Kato I. et al. Frequent inactivation of A20 in B-cell lymphomas. Nature. 2009; 459(7247): 712–6.
  56. Ding B.B., Yu J.J., Yu R.Y. et al. Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large Bcell lymphomas. Blood. 2008; 111(3): 1515–23.
  57. Lam L.T., Wright G., Davis R.E. et al. Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-{kappa}B pathways in subtypes of diffuse large B-cell lymphoma. Blood. 2008; 111(7): 3701–13.
  58. Ngo V.N., Young R.M., Schmitz R. et al. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011; 470(7332): 115–9.
  59. Bea S., Zettl A., Wright G. et al. Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve geneexpression-based survival prediction. Blood. 2005; 106(9): 3183–90.
  60. Boerma E.G., Siebert R., Kluin P.M., Baudis M. Translocations involving 8q24 in Burkitt lymphoma and other malignant lymphomas: a historical review of cytogenetics in the light of today’s knowledge. Leukemia. 2009; 23(2): 225–34.
  61. Salaverria I., Zettl A., Bea S. et al. Chromosomal alterations detected by comparative genomic hybridization in subgroups of gene expression-defined Burkitt’s lymphoma. Haematologica. 2008; 93(9): 1327–34.
  62. Scholtysik R., Kreuz M., Klapper W. et al. Detection of genomic aberrations in molecularly defined Burkitt’s lymphoma by array-based, high resolution, single nucleotide polymorphism analysis. Haematologica. 2010; 95(12): 2047–55.
  63. Pasqualucci L., Neumeister P., Goossens T. et al. Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature. 2001; 412(6844): 341–6.
  64. Hemann M.T., Bric A., Teruya-Feldstein J. et al. Evasion of the p53 tumour surveillance network by tumour-derived MYC mutants. Nature. 2005; 436(7052): 807–11.
  65. Giulino-Roth L., Wang K., MacDonald T.Y. et al. Targeted genomic sequencing of pediatric Burkitt lymphoma identifies recurrent alterations in antiapoptotic and chromatin-remodeling genes. Blood. 2012; 120(26): 5181–4.
  66. Bhatia K., Huppi K., Spangler G. et al. Point mutations in the c-Myc transactivation domain are common in Burkitt’s lymphoma and mouse plasmacytomas. Nat. Genet. 1993; 5(1): 56–61.
  67. Snuderl M., Kolman O.K., Chen Y.B. et al. B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2010; 34(3): 327–40.
  68. Le Gouill S., Talmant P., Touzeau C. et al. The clinical presentation and prognosis of diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC rearrangement. Haematologica. 2007; 92(10): 1335–42.
  69. Li S., Lin P., Fayad L.E. et al. B-cell lymphomas with B-cell lymphomas with MYC/8q24 rearrangements and IGH@BCL2/t(14;18)(q32;q21): an aggressive disease with heterogeneous histology, germinal center B-cell immunophenotype and poor outcome. Mod. Pathol. 2012; 25(1): 145–56.
  70. Klapper W., Stoecklein H., Zeynalova S. et al. Structural aberrations affecting the MYC locus indicate a poor prognosis independent of clinical risk factors in diffuse large B-cell lymphomas treated within randomized trials of the German High-Grade Non-Hodgkin’s Lymphoma Study Group (DSHNHL). Leukemia. 2008; 22(12): 2226–9.
  71. Savage K.J., Johnson N.A., Ben-Neriah S. et al. MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy. Blood. 2009; 114(17): 3533–7.
  72. Horn H., Ziepert M., Becher C. et al. MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma. Blood. 2013; 121(12): 2253–63.
  73. Barrans S., Crouch S., Smith A. et al. Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab. J. Clin. Oncol. 2010; 28(20): 3360–5.
  74. Valera A., Lopez-Guillermo A., Cardesa-Salzman T. et al. MYC protein expression and genetic alterations have prognostic impact in diffuse large B-cell lymphoma treated with immunochemotherapy. Haematologica. 2013; 98(10): 1554–62.
  75. Hummel M., Bentink S., Berger H. et al. A biologic definition of Burkitt’s lymphoma from transcriptional and genomic profiling. N. Engl. J. Med. 2006; 354(23): 2419–30.
  76. Salaverria I., Siebert R. The gray zone between Burkitt’s lymphoma and diffuse large B-cell lymphoma from a genetics perspective. J. Clin. Oncol. 2011; 29(14): 1835–43.
  77. Bertrand P., Bastard C., Maingonnat C. et al. Mapping of MYC breakpoints in 8q24 rearrangements involving non-immunoglobulin partners in B-cell lymphomas. Leukemia. 2007; 21(3): 515–23.
  78. Tomita N. BCL2 and MYC Dual-Hit Lymphoma/Leukemia. J. Clin. Exp. Hematopathol. 2011; 51(1): 7–12.
  79. Johnson N.A., Savage K.J., Ludkovski O. et al. Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival. Blood. 2009; 114(11): 2273–9.
  80. Snuderl M., Kolman O.K., Chen Y.B. et al. B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2010; 34(3): 327–40.
  81. Hoeller S., Copie-Bergman C. Grey Zone Lymphomas: Lymphomas with Intermediate Features. Advances in Hematology 2012. http://dx.doi. org/10.1155/2012/460801.
  82. Tauro S., Cochrane L., Lauritzsen G.F. et al. Dose-intensified treatment of Burkitt lymphoma and B-cell lymphoma unclassifiable, (with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma) in young adults (< 50 years): A comparison of two adapted BFM protocols. Am. J. Hematol. 2010; 85(4): 261–3.
  83. Kobayashi T., Tsutsumi Y., Sakamoto N. et al. Double-hit Lymphomas Constitute a Highly Aggressive Subgroup in Diffuse Large B-cell Lymphomas in the Era of Rituximab. Jpn. J. Clin. Oncol. 2012; 42(11): 1035–42.
  84. Fanidi A., Harrington E.A., Evan G.I. Cooperative in reactions between c-myc and bcl-2 protooncogenes. Nature. 1992; 359: 554–6.
  85. Vaux D.L., Cory S., Adams J.M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988; 335(6189): 440–2.
  86. Zhaohui J., Stratford M.W., Fengqin G., Tammy F., Xingming D. Bcl2 suppresses DNA repair by enhancing c-myc transcriptional activity. J. Biol. Chem. 2005; 281: 14446–56.
  87. Masao N., Shinobu T., Keiichiro H., Osamu T., Masao S. Synergistic effect of Bcl2, Myc and Ccnd1 transforms mouse primary B cells into malignant cells. Haematologica. 2011; 96(9): 1318–26.
  88. DeoCampo N.D., Wilson M.R., Trosko J.E. Cooperation of bcl-2 and myc in the neoplastic transformation of normal rat liver epithelial cells is related to the down-regulation of gap junction-mediated intercellular communication. Carcinogenesis. 2000; 21(8): 1501–6.
  89. Leucci E., Cocco M., Onnis A. et al. MYC translocation-negative classical Burkitt lymphoma cases: an alternative pathogenetic mechanism involving miRNA deregulation. J. Pathol. 2008; 216(4): 440–50.
  90. Onnis A., De Falco G., Antonicelli G. et al. Аlteration of microRNAs regulated by c-MYC in Burkitt lymphoma. PLoS One. 2010; 5(9); e12960.
  91. Stasik C.J., Nitta H., Zhang W. et al. Increased MYC gene copy number correlates with increased mRNA levels in diffuse large B-cell lymphoma. Haematologica. 2010; 95(4): 597–603.
  92. Schrader A., Bentink S., Spang R. et al. High MYC activity is an independent negative prognostic factor for DLBCL. Cancer. 2012; 131(4): 348–61.
  93. Yoon S.O., Jeon Y.K., Paik J.H. et al. MYC translocation and an increased copy number predict poor prognosis in adult DLBCL, especially in GCB-type. Histopathology. 2008; 53(2): 205–17.
  94. Mossafa H., Damotte D., Jenabian A. et al. Non-Hodgkin lymphomas with Burkitt-like cells are associated with c-Myc amplification and poor prognosis. Leuk. Lymphoma. 2006; 47(9): 1885–93.
  95. Martin-Subero J.I., Odero M.D., Hernandez R. et al. Amplification of IGH/ MYC fusion in clinically aggressive IGH/BCL2-positive germinal center B-cell lymphomas. Genes Chromosomes Cancer. 2005; 43(4): 414–23.
  96. Tapia G., Lopez R., Munoz-Marmol A.M. et al. Immunohistochemical detection of MYC protein correlates with MYC gene status in aggressive B-cell lymphoma. Histopathology. 2011; 59(4): 672–8.
  97. Green T.M., Nielsen O., de SK. et al. High levels of nuclear MYC protein predict the presence of MYC rearrangement in diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2012; 36(4): 612–9.
  98. Johnson N.A., Slack G.W., Savage K.J. et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. 2012; 30(28): 3452–9.
  99. Kluk M.J., Chapuy B., Sinha P. et al. Immunohistochemical detection of MYC-driven diffuse large B-cell lymphomas. PLoS One. 2012; 7(4): e33813.
  100. Testoni M., Kwee I., Greiner T.C. et al. Gains of MYC locus and outcome in patients with diffuse large B-cell lymphoma treated with R-CHOP. Br. J. Haematol. 2011; 155(2): 274–7.
  101. Hu S., Xu-Monette Z.Y., Tzankov A. et al. MYC/BCL2 protein co-expression contributes to the inferior survival of activated B-cell subtype of diffuse large B-cell lymphoma and demonstrates high-risk gene expression signatures: a report from The International DLBCL Rituximab-CHOP Consortium Program Study. Blood. 2013; 121(20): 4021–31.
  102. Piris M.A., Pezzella F., Martinez-Montero J.C. et al. p53 and bcl-2 expression in high-grade B-cell lymphomas: Correlation with survival time. Br. J. Cancer. 1994; 69: 337–41.
  103. Tang S.C., Visser L., Hepperle B. et al. Clinical significance of bcl-2-MBR gene rearrangement and protein expression in diffuse large-cell non-Hodgkin’s lymphoma: An analysis of 83 cases. J. Clin. Oncol. 1994; 12: 149–54.
  104. Barrans S.L., Carter I., Owen R.G. et al. Germinal center phenotype and bcl-2 expression combined with the International Prognostic Index improves patient risk stratification in diffuse large B-cell lymphoma. Blood. 2002; 99: 1136–43.
  105. Colomo L., Lopez-Guillermo A., Perales M. et al. Clinical impact of the differentiation profile assessed by immunophenotyping in patients with diffuse large B-cell lymphoma. Blood. 2003; 101: 78–84.
  106. Gascoyne R.D., Adomat S.A., Krajewski S. et al. Prognostic significance of Bcl-2 protein expression and Bcl-2 gene rearrangement in diffuse aggressive non-Hodgkin’s lymphoma. Blood. 1997; 90: 244–51.
  107. Martinka M., Comeau T., Foyle A. et al. Prognostic significance of t(14;18) and bcl-2 gene expression in follicular small cleaved cell lymphoma and diffuse large cell lymphoma. Clin. Invest. Med. 1997; 20: 364–70.
  108. Hill M.E., MacLennan K.A., Cunningham D.C. et al. Prognostic significance of BCL-2 expression and bcl-2 major breakpoint region rearrangement in dif- fuse large cell non-Hodgkin’s lymphoma: A British National Lymphoma Investigation Study. Blood. 1996; 88: 1046–51.
  109. Kramer M.H., Hermans J., Wijburg E. et al. Clinical relevance of BCL2, BCL6, and MYC rearrangements in diffuse large B-cell lymphoma. Blood. 1998; 92: 3152–62.
  110. Hermine O., Haioun C., Lepage E. et al. Prognostic significance of bcl-2 protein expression in aggressive non-Hodgkin’s lymphoma: Groupe d’Etude des Lymphomes de l’Adulte (GELA). Blood. 1996; 87: 265–72.
  111. Iqbal J., Neppalli V.T., Wright G., Dave B.J. BCL2 Expression Is a Prognostic Marker for the Activated B-Cell–Like Type of Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. 2006; 24(6): 961–8.
  112. Green T.M., Young K.H., Visco C. et al. Immunohistochemical DoubleHit Score Is a Strong Predictor of Outcome in Patients With Diffuse Large B-Cell Lymphoma Treated With Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone. J. Clin. Oncol. 2012; 30(28): 3460–7.
  113. Johnson N.A., Slack G.W., Savage K.J. et al. Concurrent Expression of MYC and BCL2 in Diffuse Large B-Cell Lymphoma Treated With Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone. J. Clin. Oncol. 2012; 30(28): 3452–9.
  114. Valera A., Lopez-Guillermo A., Cardesa-Salzmann T. et al. MYC protein expression and genetic alterations have prognostic impact in patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Haematologica. 2013; 98(10): 1554–62.

Биология ниши гемопоэтических стволовых клеток

Н.Ю. Семенова, С.С. Бессмельцев, В.И. Ругаль

ФГБУ «Российский научно-исследовательский институт гематологии и трансфузиологии» ФМБА РФ, 2-я Советская ул., д. 16, Санкт-Петербург, Российская Федерация, 191024

Для переписки: С.С. Бессмельцев, д-р мед. наук, профессор, 2-я Советская ул., д. 16, Санкт-Петербург, Российская Федерация, 191024; тел.: +7(812)717-67-80; e-mail: bsshem@hotmail.com

Для цитирования: Семенова Н.Ю., Бессмельцев С.С., Ругаль В.И. Биология ниши гемопоэтических стволовых клеток. Клин. онкогематол. 2014; 7(4): 501–510.


РЕФЕРАТ

В статье представлены современные данные о роли стромальной ниши костного мозга в регуляции гемопоэтических стволовых клеток (ГСК). Отражены этапы формирования концепции гемопоэтической ниши. Дана характеристика стромальных клеточных элементов, образующих нишу, и освещены механизмы регуляции ГСК. Обсуждаются вопросы роли ниши в лейкозной трансформации ГСК. Представлены сведения о структурных изменениях ниши при нарушении развития ГСК.


Ключевые слова: гемопоэтические стволовые клетки, костный мозг, ниша гемопоэтических стволовых клеток, микроокружение.

Принято в печать: 1 сентября 2014 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978; 4: 7–25.
  2. Пальцев М.А., Терских В.В., Васильев А.В. Что есть стволовая клетка. В кн.: Биология стволовых клеток и клеточные технологии. Под ред. М.А. Паль цева. Т. 1. М.: Медицина, Шико, 2009: 13–31. [Pal’tsev M.A., Terskikh V.V., Vasil’ev A.V. What is stem cell? In: Pal’tsev M.A., ed. Biologiya stvolovykh kletok i kletochnye tekhnologii. (Biology of stem cells and cell technologies.) Vol. 1. Moscow: Meditsina Publ., Shiko Publ.; 2009. pp. 13–31. (In Russ.)]
  3. O’Malley D.P., Kim Y.S., Perkins S.L. et al. Morphologic and immunohistochemical evaluation of splenic hematopoietic proliferations in neoplastic and benign disorders. Mod. Pathol. 2005; 18: 1550–61.
  4. Weiss L. A. Scanning electron microscopic study of the spleen. Blood. 1974; 43: 665–91.
  5. Kricun M.E. Red-yellow marrow conversion: its effect on the location of some solitary bone lesions. Skeletal Radiol. 1985; 14: 10–9.
  6. Williams W., Nelson D.A. Examination of the marrow. In: Hematology Williams. Ed. by E. Beulter, M.A. Lichtman et al. New York: McGraw-Hill, 1995: 15–22.
  7. Bradford G.B., Williams B., Rossi R., Bertoncello I. Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp. Hematol. 1997; 25: 445–53.
  8. Lichtman M.A. The ultrastructure of the hemopoietic environment of the marrow: a review. Exp. Hematol. 1981; 9: 391–410.
  9. Trentin J.J. Determination of bone marrow stem cell differentiation by stromal hemopoietic inductive microenvironments (HIM). Am. J. Pathol. 1971; 65: 621–8.
  10. Wolf N.S., Trentin J.J. Hemopoietic colony studies: V. Effect of hemopoietic organ stroma on differentiation of pluripotent stem cells. J. Exp. Med. 1968; 127: 205–14.
  11. Avecilla S.T., Hattori K., Heissig B. et al. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat. Med. 2004; 10: 64–71.
  12. Tokoyoda K., Egawa T., Sugiyama T. et al. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity. 2004; 20: 707–18.
  13. Dexter T.M., Allen T.D., Lajtha et al. Stimulation of differentiation and proliferation of haemopoietic cells in vitro. J. Cell Physiol. 1973; 82: 461–73.
  14. Dexter T.M., Allen T.D., Lajtha L.G. Conditions controlling the proliferation of haemopoietic stem cells in vitro. J. Cell Physiol. 1977; 91: 335–44.
  15. Cheshier S.H., Morrison S.J., Liao X., Weissman I.L. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc. Natl. Acad. Sci. USA. 1999; 96: 3120–5.
  16. Calvi L.M., Adams G.B., Weibrecht K.W. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003; 425: 841–46.
  17. Zhang J., Niu C., Ye L. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003; 425: 836–41.
  18. Kiel M.J., Yilmaz O.H., Iwashita T. et al. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005; 121: 1109–21.
  19. Nagasawa T., Omatsu Y., Sugiyama T. Control of hematopoietic stem cells by the bone marrow stromal niche: the role of reticular cells. Trends Immunol. 2011; 32(7): 315–20.
  20. Martin T.J., Sims N.A. Osteoclast-derived activity in the coupling of bone formation to resorption. Trends Mol. Med. 2005; 11: 76–81.
  21. Lian J.B., Stein G.S., Aubin J.E. Bone formation: maturation and functional activities of osteoblast lineage cells. In: Primer on the metabolic bone diseases and disorders of mineral metabolism. Ed. by M.J. Favus. Washington, DC: American Society for Bone and Mineral Research, 2003: 13–28.
  22. Adams G.B., Martin R.P., Alley I.R. et al. Therapeutic targeting of a stem cell niche. Nat. Biotechnol. 2007; 25: 238–43.
  23. Taichman R.S., Emerson S.G. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J. Exp. Med. 1994; 179: 1677–82.
  24. Taichman R.S., Reilly M.J., Emerson S.G. Human osteoblasts support human hematopoietic progenitor cells in vitro bone marrow cultures. Blood. 1996; 87: 518–24.
  25. Taichman R.S., Emerson S.G. The role of osteoblasts in the hematopoietic microenvironment. Stem Cells. 1998; 16: 7–15.
  26. Taichman R.S., Reilly M.J., Emerson S.G. The hematopoietic microenvironment: osteoblasts and the hematopoietic microenvironment. Hematology. 2000; 4: 421–6.
  27. Visnjic D., Kalajzic Z., Rowe D.W. et al. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004; 103: 3258–64.
  28. Kiel M.J., Radice G.L., Morrison S.J. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Stem Cell. 2007; 1: 204–17.
  29. Arai F., Hirao A., Ohmura M. et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004; 118: 149–61.
  30. Wilson A., Murphy M.J., Oskarsson T. et al. C-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Genes Dev. 2004; 18: 2747–63.
  31. Yoshihara H., Arai F., Hosokawa K. et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 2007; 1: 685–97.
  32. Fleming H.E., Janzen V., Lo Celso C. et al. Wnt-signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell. 2008; 2: 274–83.
  33. Nilsson S.K., Johnston H.M., Whitty G.A. et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005; 106: 1232–9.
  34. Stier S., Ko Y., Forkert R. et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J. Exp. Med. 2005; 201: 1781–91.
  35. Adams G.B., Chabner K.T., Alley I.R. et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006; 439: 599–603.
  36. Yin T., Li L. The stem cell niches in bone. J. Clin. Invest. 2006; 116: 1195–201.
  37. Broxmeyer H.E., Orschell C.M., Clapp D.W. et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J. Exp. Med. 2005; 201: 1307–18.
  38. Papayannopoulou T., Scadden D.T. Stem-cell ecology and stem cells in motion. Blood. 2008; 111: 3923–30.
  39. Sugiyama T., Kohara H., Noda M., Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006; 25: 977–88.
  40. Sipkins D.A., Wei X., Wu J.W. et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 2005; 435: 969–73.
  41. Ругаль В.И., Семенова Н.Ю. Морфология синусоидальных сосудов гемопоэтической ниши костного мозга. В кн.: Актуальные вопросы меди- цинских морфологических дисциплин. Коллективная монография под ред. В.П. Волкова. Новосибирск: СибАК, 2014: 62–80. [Rugal’ V.I., Semenova N.Yu. Morphology of sinousoid vessels of the bonemarrow hematopoietic-stem-cell niche. In: Volkov V.P., ed. Aktual’nye voprosy meditsinskikh morfologicheskikh distsiplin. (Urgent problems of medical morphological disciplines.) Novosibirsk: SibAK Publ.; 2014. pp. 62–80. (In Russ.)]
  42. Rafii S., Shapiro F., Pettengell R. et al. Human bone marrow microvascular endothelial cells support long-term proliferation and differentiation of myeloid and megakaryocytic progenitors. Blood. 1995; 86: 3353–63.
  43. Li W., Johnson S.A., Shelley W.C., Yoder M.C. Hematopoietic stem cell repopulating ability can be maintained in vitro by some primary endothelial cells. Exp. Hematol. 2004; 32: 1226–37.
  44. Cumano A., Godin I. Ontogeny of the hematopoietic system. Ann. Rev. Immunol. 2007; 25: 745–85.
  45. Orkin S.H., Zon L.I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell. 2008; 132: 631–44.
  46. Orkin S.H., Zon L.I. SnapShot: hematopoiesis. Cell. 2008; 132: 712.
  47. de Saint-Georges L., Miller S.C. The microcirculation of bone and marrow in the diaphysis of the rat hemopoietic long bones. Anat. Rec. 1992; 233: 169–77.
  48. Narayan K., Juneja S., Garcia C. Effects of 5-fluorouracil or total-body irradiation on murine bone marrow microvasculature. Exp. Hematol. 1994; 22: 142–8.
  49. Brandi M.L., Collin-Osdoby P. Vascular biology and the skeleton. J. Bone Miner. Res. 2006; 21: 183–92.
  50. Maes C., Carmeliet P., Moermans K. et al. Impaired angiogenesis and endochondral bone formation in mice lacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188. Mech. Dev. 2002; 111: 61–73.
  51. Maes C., Kobayashi T., Kronenberg H.M. A novel transgenic mouse model to study the osteoblast lineage in vivo. Ann. N.Y. Acad. Sci. 2007; 1116: 149–64.
  52. Haug J.S., He X.C., Grindley J.C. et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell. 2008; 2: 367–79.
  53. Wilson A., Oser G.M., Jaworski M. et al. Dormant and self-renewing hematopoietic stem cells and their niches. Ann. N.Y. Acad. Sci. 2007; 1106: 64–75.
  54. Morrison S.J., Wright D.E., Weissman I.L. Cyclophosphamide/granulocyte colony-stimulating factor induces hematopoietic stem cells to proliferate prior to mobilization. Proc. Natl. Acad. Sci. USA. 1997; 94: 1908–13.
  55. Randall T.D., Weissman I.L. Phenotypic and functional changes induced at the clonal level in hematopoietic stem cells after 5-fluorouracil treatment. Blood. 1997; 89: 3596–606.
  56. Zhang J., Li L. Stem cell niche: microenvironment and beyond. J. Biol. Chem. 2008; 283: 9499–503.
  57. Baron R. General Principles of Bone Biology. In: Primer on the metabolic bone diseases and disorders of mineral metabolism. Ed. by M.J. Favus. Washington, DC: American Society for Bone and Mineral Research, 2003: 1–8.
  58. Belloni P.N., Tressler R.J. Microvascular endothelial cell heterogeneity: interactions with leukocytes and tumor cells. Cancer Metastas. Rev. 1990; 8: 353–89.
  59. Afan A.M., Broome C.S., Nicholls S.E. et al. Bone marrow innervation regulates cellular retention in the murine haemopoietic system. Br. J. Haematol. 1997; 98: 569–77.
  60. Katayama Y., Battista M., Kao W.M. et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006; 124: 407–21.
  61. Mendez-Ferrer S., Lucas D., Battista M., Frenette P.S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008; 452: 442–7.
  62. Calvo W., Forteza-Vila J. On the development of bone marrow innervation in new-born rats as studied with silver impregnation and electron microscopy. Am. J. Anat. 1969; 126: 355–71.
  63. Calvo W., Forteza-Vila J. Schwann cells of the bone marrow. Blood. 1970; 36: 180–8.
  64. Yamazaki K., Allen T.D. Ultrastructural morphometric study of efferent nerve terminals on murine bone marrow stromal cells, and the recognition of a novel anatomical unit: the «neuro-reticular complex». Am. J. Anat. 1990; 187: 261–76.
  65. Spiegel A., Shivtiel S., Kalinkovich A. et al. Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling. Nat. Immunol. 2007; 8: 1123–31.
  66. Jacenko O., Roberts D.W., Campbell M.R. et al. Linking hematopoiesis to endochondral skeletogenesis through analysis of mice transgenic for collagen X. Am. J. Pathol. 2002; 160: 2019–34.
  67. Walkley C.R., Olsen G.H., Dworkin S. et al. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell. 2007; 129: 1097–110.
  68. Walkley C.R., Shea J.M., Sims N.A. et al. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell. 2007; 129: 1081–95.
  69. Iwata M., Awaya N., Graf L. et al. Human marrow stromal cells activate monocytes to secrete osteopontin, which down-regulates Notch1 gene expression in CD34+ cells. Blood. 2004; 103: 4496–502.
  70. Li L., Milner L.A., Deng Y. et al. The human homolog of rat Jagged1 expressed by marrow stroma inhibits differentiation of 32D cells through interaction with Notch1. Immunity. 1998; 8: 43–55.
  71. Kollet O., Dar A., Shivtiel S. et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat. Med. 2006; 12: 657–64.
  72. Fukuhara S., Sako K., Minami T. et al. Differential function of Tie2 at cellcell contacts and cell-substratum contacts regulated by angiopoietin-1. Nat. Cell Biol. 2008; 10: 513–26.
  73. Saharinen P., Eklund L., Miettinen J. et al. Angiopoietins assemble distinct Tie2 signalling complexes in endothelial cell-cell and cell-matrix contacts. Nat. Cell Biol. 2008; 10: 527–37.
  74. Ferrara N., Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr. Rev. 1997; 18: 4–25.
  75. Zelzer E., Olsen B.R. Multiple roles of vascular endothelial growth factor (VEGF) in skeletal development, growth, and repair. Curr. Top. Dev. Biol. 2005; 65: 169–87.
  76. Sacchetti B., Funari A., Michienzi S. et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell. 2007; 131: 324–36.
  77. Shi S., Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J. Bone Miner Res. 2003; 18: 696–704.
  78. Duhrsen U., Hossfeld D.K. Stromal abnormalities in neoplastic bone marrow diseases. Ann. Hematol. 1996; 73: 53–70.
  79. Бессмельцев С.С. Множественная миелома (патогенез, клиника, диагностика, дифференциальный диагноз). Часть 1. Клин. онкогематол. 2013; 6(3): 237–58. [Bessmel’tsev S.S. Multiple myeloma (pathogenesis, clinical features, diagnosis, differential diagnosis). Part 1. Klin. Onkogematol. 2013; 6(3): 237–58. (In Russ.)]
  80. Semenova N., Bessmeltsev S., Rugal V. Nicheforming stromal elements of bone marrow and lymph nodes in CLL. Haematologica. 2014; 99(s1): 743.
  81. Ругаль В.И., Бессмельцев С.С., Семенова Н.Ю. и др. Структурные особенности паренхимы и стромы костного мозга больных множественной миеломой. Биомедицинский журнал Medline.ru. 2012; 13: 515–23. [Rugal’ V.I., Bessmel’tsev S.S., Semenova N.Yu. et al. Structural features of bone marrow parenchyma and stroma in patients with multiple myeloma. Biomeditsinskii zhurnal Medline.ru. 2012; 13: 515–23. (In Russ.)]
  82. Bessmeltsev S., Rugal V. Stromal microenvironment and stem cells niche in multiple myeloma. Haematologica. 2010; 95(25): 569–570.
  83. Kim Y.W., Koo B.K., Jeong H.W. et al. Defective Notch activation in microenvironment leads to myeloproliferative disease. Blood. 2008; 112: 4628–38.
  84. Raajimakers M.H., Mukherjee S., Guo S. et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010; 464: 852–7.
  85. Blau O., Hofmann W.K., Baldus C.D. et al. Chromosomal aberrations in bone marrow mesenchymal stroma cells from patients with myelodysplastic syndrome and acute myeloblastic leukemia. Exp. Hematol. 2007; 35: 221–9.
  86. Sala-Torra O., Hanna C., Loken M.R. et al. Evidence of donor-derived hematologic malignancies after hematopoietic stem cell transplantation. Biol. Blood Marrow Transpl. 2006; 12: 511–7.
  87. Colmone A., Amorim M., Pontier A.L. et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008; 322: 1861–5.
  88. Jin L., Hope K.J., Zhai Q. et al. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat. Med. 2006; 12: 1167.
  89. Krause D.S., Lazarides K., von Andrian U.H., van Etten R.A. Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells. Nat. Med. 2006; 12: 1175–80.
  90. Miyake K., Underhill C.B., Lesley J., Kincade P.W. Hyaluronate can function as a cell adhesion molecule and CD44 participates in hyaluronate recognition. J. Exp. Med. 1990; 172: 69–75.
  91. Katayama Y., Hidalgo A., Chang J. et al. CD44 is a physiological Eselectin ligand on neutrophils. J. Exp. Med. 2005; 201: 1183–9.
  92. Dimitroff C.J., Lee J.Y., Rafii S. et al. CD44 is a major E-selectin ligand on human hematopoietic progenitor cells. J. Cell Biol. 2001; 153: 1277–86.
  93. Krause D.S., von Andrian U.H., van Etten R.A. Selectins and their ligands are required for for homing and engraftment of BCR-ABL leukemia-initiating cells. Blood. 2005; 106: 106a.
  94. Jin L., Lee E.M., Ramshaw H.S. et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell. 2009; 5: 31–42.
  95. Garg M., Moore H., Tobal K., Liu Yin J.A. Prognostic significance of quantitative analysis of WT1 gene transcripts by competitive reverse transcription polymerase chain reaction in acute leukaemia. Br. J. Haematol. 2003; 123: 49–59.
  96. Ishikawa F., Yoshida S., Saito Y. et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat. Biotechnol. 2007; 25: 1315–21.
  97. Saito Y., Uchida N., Tanaka S. et al. Induction of cell cycle entry eliminates human leukemia stem cells in s a mouse model of AML. Nat. Biotechnol. 2010; 28: 275–80.
  98. Klyuchnikov E., Kroger N. Sensitising leukemic cells by targeting microenvironment. Leuk. Lymphoma. 2009; 50: 319–20.
  99. Matsunaga T., Takemoto N., Sato T. et al. Interaction between leukemiccell VLA-4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia. Nat. Med. 2003; 9: 1158–65.
  100. Mraz M., Zent C.S., Church A.K. et al. Bone marrow stromal cells protect lymphoma B-cells from rituximab-induced apoptosis and targeting integrin alpha-4-beta-1 (VLA-4) with natalizumab can overcome this resistance. Br. J. Haematol. 2011; 155: 53–64.
  101. Vianello F., Villanova F., Tisato V. et al. Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis. Haematologica. 2010; 95: 1081–9.
  102. Weisberg E., Azab A.K., Manley P.W. et al. Inhibition of CXCR4 in CML cells disrupts their interaction with the bone marrow microenvironment and sensitizes them to nilotinib. Leukemia. 2012; 26: 985–90.
  103. Bhatia R., McGlave P.B., Dewald G.W. et al. Abnormal function of the bone marrow microenvironment in chronic myelogenous leukemia: role of malignant stromal macrophages. Blood. 1995; 85: 3636–45.
  104. Bewry N.N., Nair R.R., Emmons M.F. et al. Stat3 contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment model of drug resistance. Mol. Cancer Ther. 2008; 7: 3169–75.
  105. Scupoli M.T., Perbellini O., Krampera M. et al. Interleukin 7 requirement for survival of T-cell acute lymphoblastic leukemia and human thymocytes on bone marrow stroma. Haematologica. 2007; 92: 264–6.
  106. Yamamoto-Sugitani M., Kuroda J., Ashihara E. et al. Galectin-3 (Gal-3) induced by leukemia microenvironment promotes drug resistance and bone marrow lodgment in chronic myelogenous leukemia. Proc. Natl. Acad. Sci. USA. 2011; 108: 17468–73.
  107. Lane S.W., Wang Y.J., Lo Celso C. et al. Differential niche and Wnt requirements during acute myeloid leukemia progression. Blood. 2011; 118: 2849–56.
  108. Wei J., Wunderlich M., Fox C. et al. Microenvironment determines lineage fate in a human model of MLL-AF9 leukemia. Cancer Cell. 2008; 13: 483–95.
  109. Spitzer T.R., Dey B.R., Chen Y.B. et al. The expanding frontier of hematopoietic cell transplantation. Cytometr. B. Clin. Cytom. 2012; 82(5): 271–9.
  110. Jordan C.T., Upchurch D., Szilvassy S.J. et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000; 14: 1777–84.
  111. Kugler M., Stein C., Kellner C. et al. A recombinant trispecific singlechain Fv derivative directed against CD123 and CD33 mediates effective elimination of acute myeloid leukaemia cells by dual targeting. Br. J. Haematol. 2010; 150: 574–86.
  112. Krause D.S., Fulzele K., Catic A. et al. Parathyroid hormone-induced modulation of the bone marrow microenvironment reduces leukemic stem cells in murine chronic myelogenous-leukemia-like disease via a TGFbeta-dependent pathway. Blood. 2011; 118: 1670.

Альтернативное кровоснабжение в костном мозге при онкогематологических заболеваниях

А.А. Вартанян

ФГБУ «Российский онкологический научный центр им. Н.Н. Блохина», Каширское ш., д. 24, Москва, Российская Федерация, 115478

Для переписки: А.А. Вартанян, д-р биол. наук, ст. науч. сотрудник, Каширское ш., д. 24, Москва, Российская Федерация, 115478; тел.: +7(499)324-10-65; e-mail: zhivotov57@mail.ru

Для цитирования: Вартанян А.А. Альтернативное кровоснабжение в костном мозге при онкогематологических заболеваниях. Клин. онкогематол. 2014; 7(4): 491–500.


РЕФЕРАТ

Неоангиогенез, или формирование новых микрососудов на основе уже существующей в ткани сети сосудов, является необходимым условием для роста опухоли. Долгое время неоангиогенез считали единственной возможностью доставки в опухоль питательных веществ и кислорода. В последние годы рассматриваются также альтернативные механизмы васкуляризации опухоли. Формирование высокоструктурированных васкулярных каналов из опухолевых клеток в отсутствие эндотелиальных клеток и фибробластов, ограниченных базальной мембраной, или васкулогенная мимикрия (ВМ), сегодня рассматривается как дополнительная система кровоснабжения опухоли. ВМ обнаружена практически во всех опухолях, и ее появление ассоциируется с плохим прогнозом. В настоящем обзоре суммированы основные характеристики ВМ в солидных опухолях и при онкогематологических заболеваниях. Обсуждается также значение указанного феномена в диагностике опухолей и в прогнозировании их течения.


Ключевые слова: неоангиогенез, васкулогенная мимикрия, онкогематологические заболевания.

Принято в печать: 1 сентября 2014 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Persson A., Buschmann I. Vascular growth in health and disease. Front. Mol. Neurosci. 2011; 24: 14–8.
  2. Balaji S., King A., Crombleholme T. et al. The Role of Endothelial Progenitor Cells in Postnatal Vasculogenesis: Implications for Therapeutic Neovascularization and Wound Healing. Adv. Wound Care (New Rochelle). 2013; 2(6): 283–95.
  3. LeBlanc A.J., Krishnan L., Sullivan C.J. et al. Microvascular repair: postangiogenesis vascular dynamics. Microcirculation. 2012; 19(8): 676–95.
  4. Folkman J. New perspectives in clinical oncology from angiogenesis research. Eur. J. Cancer. 1996; 32A(14): 2534–9.
  5. Shibuya M. VEGF-VEGFR Signals in Health and Disease. Biomol. Ther. 2014; 22(1): 1–9.
  6. Vempati P., Popel A.S., MacGabhann S. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev. 2014; 25(1): 1–19.
  7. De Falco S. The discovery of placenta growth factor and its biological activity. Exp. Mol. Med. 2012; 44(1): 1–9.
  8. Lieu C., Heymach J., Overman M. et al. Beyond VEGF: inhibition of the fibroblast growth factor pathway and antiangiogenesis. Clin. Cancer Res. 2011; 17(19): 6130–9.
  9. Hellberg C., Ostman A., Heldin C.H. PDGF and vessel maturation. Recent Results Cancer Res. 2010; 180: 103–14.
  10. Fagiani E., Christofori G. Angiopoietins in angiogenesis. Cancer Lett. 2013; 328(1): 18–26.
  11. Moschetta M., Mishima Y., Sahin I. et al. Role of endothelial progenitor cells in cancer progression. Biochim. Biophys. Acta. 2014; 1846(1): 26–39.
  12. Donnem T., Hu J., Ferguson M. et al. Vessel co-option in primary human tumors and metastases: an obstacle to effective anti-angiogenic treatment? Cancer Med. 2013; 2(4): 427–36.
  13. Maniotis A.J., Folberg R., Hess A. et al. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am. J. Pathol. 1999; 155(3): 739–52.
  14. Hendrix M.J., Seftor E.A., Hess A.R. et al. Molecular plasticity of human melanoma cells. Oncogene. 2003; 22(20): 3070–5.
  15. Welti J., Loges S., Dimmeler S., Carmeliet P. Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J. Clin. Invest. 2013; 123(8): 3190–200.
  16. Cao Z., Bao M., Miele L., Sarkar F.H., Wang Z., Zhou Q. Tumour vasculogenic mimicry is associated with poor prognosis of human cancer patients: a systemic review and meta-analysis. Eur. J. Cancer. 2013; 49(18): 3914–23.
  17. Seftor R.E., Hess A.R., Seftor E.A. et al. Tumor cell vasculogenic mimicry: from controversy to therapeutic promise. Am. J. Pathol. 2012; 181(4): 1115–25.
  18. Fan Y.Z., Sun W. Molecular regulation of vasculogenic mimicry in tumors and potential tumor-target therapy. World J. Gastrointest. Surg. 2010; 2(4): 117–27.
  19. Hess A.R., Seftor E.A., Gruman L.M. et al. VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway: implications for vasculogenic mimicry. Cancer Biol. Ther. 2006; 5(2): 228–33.
  20. Mourad-Zeidan A.A., Melnikova V.O., Wang H. Expression profiling of Galectin-3-depleted melanoma cells reveals its major role in melanoma cell plasticity and vasculogenic mimicry. Am. J. Pathol. 2008; 173(6): 1839–52.
  21. Basu G.D., Pathangey L.B., Tinder T.L. Mechanisms underlying the growth inhibitory effects of the cyclo-oxygenase-2 inhibitor celecoxib in human breast cancer cells. Breast Cancer Res. 2005; 7(4): R422–35.
  22. Vartanian A., Gatsina G., Grigorieva I. et al. The involvement of Notch signaling in melanoma vasculogenic mimicry. Clin. Exp. Med. 2013; 13(3): 201–9.
  23. Vartanian A., Stepanova E., Grigorieva I. et al. Melanoma vasculogenic mimicry capillary-like structure formation depends on integrin and calcium signaling. Microcirculation. 2011; 18(5): 390–9.
  24. Vartanian A., Stepanova E., Grigorieva I. VEGFR1 and PKC control melanoma vasculogenic mimicry in a VEGFR2 kinase-independent manner. Melanoma Res. 2011; 21(2): 91–8.
  25. Lissitzky J.C., Parriaux D., Ristorcelli E. Cyclic AMP signaling as a mediator of vasculogenic mimicry in aggressive human melanoma cells in vitro. Cancer Res. 2009; 69(3): 802–9.
  26. Xi Y., Nakajima G., Hamil T. Association of insulin-like growth factor binding protein-3 expression with melanoma progression. Mol. Cancer Ther. 2006; 5(12): 3078–84.
  27. Hess A.R., Hendrix M.J. Focal adhesion kinase signaling and the aggressive melanoma phenotype. Cell Cycle. 2006; 5(5): 478–80.
  28. Ruf W., Seftor E.A., Petrovan R.J. et al. Differential role of tissue factor pathway inhibitors 1 and 2 in melanoma vasculogenic mimicry. Cancer Res. 2003; 63(17): 5381–9.
  29. Ciurea M.E., Georgescu A.M., Purcaru S.O. Cancer stem cells: biological functions and therapeutically targeting. Int. J. Mol. Sci. 2014; 15(5): 8169–85.
  30. Friedmann-Morvinski D., Verma I.M. Dedifferentiation and reprogramming: origins of cancer stem cells. EMBO Rep. 2014; 15(3): 244–53.
  31. Stewart J.M., Shaw P.A., Geyde C. et al. Phenotypic heterogenity and instability of human ovarian tumor-initiating cells. Proc. Natl. Acad. Sci. USA. 2011; 108(16): 6468–73.
  32. Meier P., Finch A., Evan G. Apoptosis in development. Nature. 2000; 407(6805): 796–801.
  33. Tait S.W., Ichim G., Green D.R. Die another way — non-apoptotic mechanisms of cell death. J. Cell Sci. 2014; 127(Pt. 10): 2135–44.
  34. Vartanian A., Burova O., Stepanova E. et al. The involvement of apoptosis in melanoma vasculogenic mimicry. Mel Res. 2007; 1: 1–8.
  35. Vartanian A., Burova O., Stepanova E. et al. Melanoma vasculogenic mimicry is strongly related to reactice oxygen species level. Mel. Res. 2007; 17(6): 370–9.
  36. Narendhirakannan R.T., Hannah M.A. Oxidative Stress and Skin Cancer: An Overview. Indian J. Clin. Biochem. 2013; 28(2): 110–5.
  37. Holmstrom K.M., Finkel T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 2014; 15(6): 411–21.
  38. Brakenhielm E., Cao R., Cao Y. et al. Suppression of angiogenesis, tumor growth, and wound healing by resveratrol, a natural compound from red wine and grapes. FASEB J. 2001; 15: 1798–800.
  39. Vartanian A., Stepanova E., Grigorieva I. et al. Melanoma vasculogenic mimicry capillary-like structure formation depends on integrin and calcium signaling. Microcirculation. 2011; 18(5): 390–9.
  40. Shirakawa K., Kobayashi H., Heike Y. et al. Hemodynamics in vasculogenic mimicry and angiogenesis of inflammatory breast cancer xenograft. Cancer Res. 2002; 62(2): 560–6.
  41. Folberg R., Rummel V., Ginderdeuren R. et al. The prognostic value of tumor blood vessel morphology in primary uveal melanoma. Ophthalmology. 1993; 100: 1389–98.
  42. Vartanian A., Stepanova E., Baryshnikov A. et al. Prognostic significance of Periodic Acid-Shiff-positive patterns in clear cell renal cell carcinoma. Canad. J. Urol. 2009; 16(4): 4726–31.
  43. Григорьева И.Н., Вишневская Я.В., Абрамов М.Е. и др. Особенности васкуляризации меланомы кожи человека. Забайкальский медицинский вестник. 2011; 2: 12–8.  [Grigor’eva I.N., Vishnevskaya Ya.V., Abramov M.E. et al. Peculiarities of vacularization of human skin melanoma. Zabaikal’skii meditsinskii vestnik. 2011; 2: 12–8. (In Russ.)]
  44. Wang S.Y., Ke Y.Q., Lu G.H. et al. Vasculogenic mimicry is a prognostic factor for postoperative survival in patients with glioblastoma. J. Neurooncol. 2013; 112(3): 339–45.
  45. Lin P., Wang W., Sun B.C. et al. Vasculogenic mimicry is a key prognostic factor for laryngeal squamous cell carcinoma: a new pattern of blood supply. Chin. Med. J. (Engl.) 2012; 125(19): 3445–9.
  46. Liu R., Yang K., Meng C. Vasculogenic mimicry is a marker of poor prognosis in prostate cancer. Cancer Biol. Ther. 2012; 13(7): 527–33.
  47. Wang S.Y., Yu L., Ling G.Q. et al. Vasculogenic mimicry and its clinical significance in medulloblastoma. Cancer Biol. Ther. 2012; 13(5): 341–8.
  48. Liu X.M., Zhang Q.P., Mu Y.G. et al. Clinical significance of vasculogenic mimicry in human gliomas. J. Neurooncol. 2011; 105(2): 173–9.
  49. Liu W.B., Xu G.L., Jia W.D. et al. Prognostic significance and mechanisms of patterned matrix vasculogenic mimicry in hepatocellular carcinoma. Med. Oncol. 2011; 28: S228–38.
  50. Li M., Gu Y., Zhang Z. et al. Vasculogenic mimicry: a new prognostic sign of gastric adenocarcinoma. Pathol. Oncol. Res. 2010; 16(2): 259–66.
  51. Baeten C.I., Hillen F., Pauwels P. et al. Prognostic role of vasculogenic mimicry in colorectal cancer. Dis. Colon Rectum. 2009; 52(12): 2028–35.
  52. Sood A.K., Fletcher M.S., Zahn C.M. et al. The clinical significance of tumor cell-lined vasculature in ovarian carcinoma: implications for anti-vasculogenic therapy. Cancer Biol. Ther. 2002; 1(6): 661–4.
  53. Sun B., Zhang S., Zhao X. et al. Vasculogenic mimicry is associated with poor survival in patients with mesothelial sarcomas and alveolar rhabdomyosarcomas. Int. J. Oncol. 2004; 25(6): 1609–14.
  54. Wu S., Yu L., Wang D. et al. Aberrant expression of CD133 in non-small cell lung cancer and its relationship to vasculogenic mimicry. BMC Cancer. 2012; 12: 535–8.
  55. Cameron D., Brown J., Dent R. et al. Adjuvant bevacizumab-containing therapy in triple-negative breast cancer (BEATRICE): primary results of a randomised, phase 3 trial. Lancet Oncol. 2013; 14(10): 933–42.
  56. Corrie P.G., Marshall A., Dunn J.A. et al. Adjuvant bevacizumab in patients with melanoma at high risk of recurrence (AVAST-M): preplanned interim results from a multicentre, open-label, randomised controlled phase 3 study. Lancet Oncol. 2014; 15(6): 620–30.
  57. Dias S., Hattori K., Zhu Z. et al. Autocrine stimulation of VEGFR-2 activates human leukemic cell growth and migration. J. Clin. Invest. 2000; 106: 511–21.
  58. Li W.W., Hutnik M., Gehr G. Antiangiogenesis in haematological malignancies. Br. J. Haematol. 2008; 143(5): 622–31.
  59. Grosicki S., Grosicka A., Holowiecki J. Clinical importance of angiogenesis and angiogenic factors in oncohematology. Wiad. Lek. 2007; 60(1–2): 39–46.
  60. Dimopoulos M.A., Delimpasi S., Katodritou E. et al. Significant improvement in the survival of patients with multiple myeloma presenting with severe renal impairment after the introduction of novel agents. Ann. Oncol. 2014; 25(1): 195–200.
  61. Song G., Li Y., Jiang G. Role of VEGF/VEGFR in the pathogenesis of leukemias and as treatment targets. Oncol. Rep. 2012; 28(6): 1935–44.
  62. Ruan J. Antiangiogenic therapies in non-Hodgkin’s lymphoma. Curr. Cancer Drug Targets. 2011; 11(9): 1030–43.
  63. Gong J.K. Endosteal marrow: a rich source of hematopoietic stem cells. Science. 1978; 199: 1443–45.
  64. Yin T., Li L. The stem cell niches in bone. J. Clin. Invest. 2006; 116(5): 1195–201.
  65. Bradford G.B., Williams B., Rossi R. et al. Quiescence, cycling and turnover in the hematopoietic stem cell compartment. Exp. Hematol. 1997; 25(5): 445–53.
  66. Вартанян А. Основные закономерности ангиогенеза при онкогематологических заболеваниях. Клин. онкогематол. 2013; 6(4): 343–54. [Vartanyan A. Basic principles of angiogenesis in hematological malignancies. Klin. Onkogematol. 2013; 6(4): 343–54. (In Russ.)]
  67. Nico B., Margieri D., Crivellato E. et al. Mast cells contribute to vasculogenic mimicry in multiple myeloma. Stem Cell Dev. 2008; 17(1): 19–22.
  68. Scavelli C., Nico B., Cirulli T. et al. Vasculogenic mimicry by bone marrow macrophages in patients with multiple myeloma. Oncogene. 2008; 27(5): 663–74.
  69. Mirshahi P., Raffi A., Vincent I. et al. Vasculogenic mimicry of acute leukemic bone marrow stromal cells. Leukemia. 2009; 23: 1039–48.
  70. Ding Y.P., Yang X.D., Wu Y. et al. Autophagy promotes the survival and development of tumors by participating in the formation of vasculogenic mimicry. Oncol. Rep. 2014; 31(5): 2321–7.
  71. Mizushima N., Levine B., Cuervo A.M. et al. Autophagy fights disease through cellular selfdigestion. Nature. 2008; 451: 1069–75.
  72. Shimizu S., Yoshida T., Tsujioka M. et al. Autophagic cell death and cancer. Int. J. Mol. Sci. 2014; 15(2): 3145–53.