МикроРНК: малые молекулы с большим значением

В.Н. Аушев

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

Для цитирования: Аушев В.Н. МикроРНК: малые молекулы с большим значением. Клиническая онкогематология. 2015;8(1):1–12.

Для переписки: Василий Николаевич Аушев, канд. биол. наук, Каширское ш., д. 24, Москва, Российская Федерация, 115478; тел.: +7(499)324-17-64; e-mail: vaushev@gmail.com


РЕФЕРАТ

Обоснование. МикроРНК были впервые обнаружены как антисмысловые транскрипты у нематоды Caenorhabditis elegans, где они подавляли экспрессию генов, содержащих в мРНК комплементарные последовательности. Таким образом, данные молекулы наряду с короткими интерферирующими микроРНК являются основными медиаторами РНК-интерференции — универсального механизма регуляции экспрессии.

Результаты. МикроРНК представляют собой небольшие молекулы, транскрибируемые с геномной ДНК и подвергающиеся дальнейшему процессингу и экспорту в цитоплазму. Они могут входить в состав транскриптов, кодирующих белки, либо транскрибироваться с белок-некодирующих участков. Первичный процессинг может осуществляться с участием специализированного ферментного комплекса либо в ходе стандартного сплайсинга мРНК. После экспорта в цитоплазму промежуточный продукт подвергается окончательному процессингу с образованием активного РНК-белкового комплекса, способного связываться с комплементарными участками мРНК-«мишеней». Результатом такого связывания является подавление трансляции с данной мРНК. Сама мРНК во многих случаях может быть расщеплена за счет РНКазной активности комплекса.

Выводы. В геноме человека закодировано несколько тысяч микроРНК, образующих обширную регуляторную сеть, которая задействована в самых разных сигнальных путях и клеточных процессах. Нарушения микроРНК-регуляции вовлечены в развитие широкого спектра заболеваний, включая все типы неоплазий. МикроРНК имеют большое значение в онкологии, в частности в онкогематологии, как перспективные маркеры и потенциальные терапевтические агенты. К настоящему времени показано участие отдельных микроРНК в патогенезе большинства заболеваний системы крови. В ряде случаев предлагается использовать данные молекулы в качестве средств молекулярной диагностики и для определения прогноза заболевания.


Ключевые слова: микроРНК, регуляция экспрессии, онкомаркеры.

Получено: 16 июля 2014 г.

Принято в печать: 7 октября 2014 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 2013;12(11):847–65. doi: 10.1038/nrd4140.
  2. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54. doi: 10.1016/0092-8674(93)90529-y.
  3. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75(5):855–62. doi: 10.1016/0092-8674(93)90530-4.
  4. Lee R, Feinbaum R, Ambros V. A short history of a short RNA. Cell. 2004;116(2 Suppl):S89–S92. doi: 10.1016/s0092-8674(04)00035-2.
  5. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5(7):522–31. doi: 10.1038/nrg1379.
  6. Reinhart BJ, Slack FJ, Basson M, et al. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 2000;403(6772):901–6. doi: 10.1038/35002607.
  7. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of Novel Genes Coding for Small Expressed RNAs. Science. 2001;294(5543):853–8. doi: 10.1126/science.1064921.
  8. Lau NC, Lim LP, Weinstein EG, et al. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001;294(5543):858–62. doi: 10.1126/science.1065062.
  9. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of Novel Genes Coding for Small Expressed RNAs. Science. 2001;294(5543):855–8. doi: 10.1126/science.1064921.
  10. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99(24):15524–9.
  11. Ambros V. A uniform system for microRNA annotation. RNA. 2003;9(3):277–9. doi: 10.1261/rna.2183803.
  12. Griffiths-Jones S, Grocock RJ, van Dongen S, et al. miRBase: microRNA sequences, targets and gene nomenclature. Nucl Acids Res. 2006;34(Database issue):D140–4. doi: 10.1093/nar/gkj112.
  13. Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucl Acids Res. 2011;39(Database issue):D152–7. doi: 10.1093/nar/gkq1027.
  14. Lei EP, Silver PA. Protein and RNA export from the nucleus. Dev Cell. 2002;2(3):261–72. doi: 10.1016/s1534-5807(02)00134-x.
  15. Behm-Ansmant I, Rehwinkel J, Doerks T, et al. mRNA degradation by miRNAs and GW182 requires both CCR4:NOT deadenylase and DCP1:DCP2 decapping complexes. Genes Dev. 2006;20(14):1885–98. doi: 10.1101/gad.1424106.
  16. Nishihara T, Zekri L, Braun JE, et al. miRISC recruits decapping factors to miRNA targets to enhance their degradation. Nucl Acids Res. 2013;41(18):8692–705. doi: 10.1093/nar/gkt619.
  17. Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation. Science. 2007;318(5858):1931–4. doi: 10.1126/science.1149460.
  18. Wilson RC, Doudna JA. Molecular mechanisms of RNA interference. Ann Rev Biophys. 2013;42:217–39. doi: 10.1146/annurev-biophys-083012-130404.
  19. Friedman RC, Farh KK, Burge CB, et al. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92–105. doi: 10.1101/gr.082701.108.
  20. Cobb BS, Hertweck A, Smith J, et al. A role for Dicer in immune regulation. J Exp Med. 2006;203(11):2519–27. doi: 10.1084/jem.20061692.
  21. O’Carroll D, Mecklenbrauker I, Das PP, et al. A Slicer-independent role for Argonaute 2 in hematopoiesis and the microRNA pathway. Genes Dev. 2007;21(16):1999–2004. doi: 10.1101/gad.1565607.
  22. Felli N, Fontana L, Pelosi E, et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proc Natl Acad Sci USA. 2005;102(50):18081–6. doi: 10.1073/pnas.0506216102.
  23. Wang Q, Huang Z, Xue H, et al. MicroRNA miR-24 inhibits erythropoiesis by targeting activin type I receptor ALK4. Blood. 2008;111(2):588–95. doi: 10.1182/blood-2007-05-092718.
  24. Elton TS, Selemon H, Elton SM, et al. Regulation of the MIR155 host gene in physiological and pathological processes. Gene. 2013;532(1):1–12. doi: 10.1016/j.gene.2012.12.009.
  25. Dagan LN, Jiang X, Bhatt S, et al. miR-155 regulates HGAL expression and increases lymphoma cell motility. Blood. 2012;119(2):513–20. doi: 10.1182/blood-2011-08-370536.
  26. Teng G, Hakimpour P, Landgraf P, et al. MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity. 2008;28(5):621–9. doi: 10.1016/j.immuni.2008.03.015.
  27. Bissels U, Bosio A, Wagner W. MicroRNAs are shaping the hematopoietic landscape. Haematologica. 2012;97(2):160–7. doi: 10.3324/haematol.2011.051730.
  28. Listowski MA, Heger E, Boguslawska DM, et al. microRNAs: fine tuning of erythropoiesis. Cell Mol Biol Lett. 2013;18(1):34–46. doi: 10.2478/s11658-012-0038-z.
  29. Lawrie CH. MicroRNAs in hematological malignancies. Blood Rev. 2013;27(3):143–54. doi: 10.1016/j.blre.2013.04.002.
  30. Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 2005;102(39):13944–9. doi: 10.1073/pnas.0506654102.
  31. Fabbri M, Bottoni A, Shimizu M, et al. Association of a microRNA/TP53 feedback circuitry with pathogenesis and outcome of B-cell chronic lymphocytic leukemia. JAMA. 2011;305(1):59–67. doi: 10.1001/jama.2010.1919.
  32. Pekarsky Y, Santanam U, Cimmino A, et al. Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res. 2006;66(24):11590–3. doi: 10.1158/0008-5472.can-06-3613.
  33. Starczynowski DT, Morin R, McPherson A, et al. Genome-wide identification of human microRNAs located in leukemia-associated genomic alterations. Blood. 2011;117(2):595–607. doi: 10.1182/blood-2010-03-277012.
  34. Starczynowski DT, Kuchenbauer F, Argiropoulos B, et al. Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype. Nat Med. 2010;16(1):49–58. doi: 10.1038/nm.2054.
  35. Bousquet M, Quelen C, Rosati R, et al. Myeloid cell differentiation arrest by miR-125b-1 in myelodysplastic syndrome and acute myeloid leukemia with the t(2;11)(p21;q23) translocation. J Exp Med. 2008;205(11):2499–506. doi: 10.1084/jem.20080285.
  36. Chapiro E, Russell LJ, Struski S, et al. A new recurrent translocation t(11;14)(q24;q32) involving IGH@ and miR-125b-1 in B-cell progenitor acute lymphoblastic leukemia. Leukemia. 2010;24(7):1362–4. doi: 10.1038/leu.2010.93.
  37. Bousquet M, Harris MH, Zhou B, et al. MicroRNA miR-125b causes leukemia. Proc Natl Acad Sci USA. 2010;107(50):21558–63. doi: 10.1073/pnas.1016611107.
  38. Agirre X, Jimenez-Velasco A, San Jose-Eneriz E, et al. Down-regulation of hsa-miR-10a in chronic myeloid leukemia CD34+ cells increases USF2-mediated cell growth. Mol Cancer Res. 2008;6(12):1830–40. doi: 10.1158/1541-7786.mcr-08-0167.
  39. Bueno MJ, Perez de Castro I, Gomez de Cedron M, et al. Genetic and epigenetic silencing of microRNA-203 enhances ABL1 and BCR-ABL1 oncogene expression. Cancer Cell. 2008;13(6):496–506. doi: 10.1016/j.ccr.2008.04.018.
  40. Venturini L, Battmer K, Castoldi M, et al. Expression of the miR-17-92 polycistron in chronic myeloid leukemia (CML) CD34+ cells. Blood. 2007;109(10):4399–405. doi: 10.1182/blood-2006-09-045104.
  41. Xu C, Fu H, Gao L, et al. BCR-ABL/GATA1/miR-138 mini circuitry contributes to the leukemogenesis of chronic myeloid leukemia. Oncogene. 2014:33(1):44–54. doi: 10.1038/onc.2012.557.
  42. Mi S, Lu J, Sun M, et al. MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc Natl Acad Sci USA. 2007;104(50):19971–6. doi: 10.1073/pnas.0709313104.
  43. Schotte D, De Menezes RX, Akbari Moqadam F, et al. MicroRNA characterize genetic diversity and drug resistance in pediatric acute lymphoblastic leukemia. Haematologica. 2011;96(5):703–11. doi: 10.3324/haematol.2010.026138.
  44. Magrath I. Epidemiology: clues to the pathogenesis of Burkitt lymphoma. Br J Haematol. 2012;156(6):744–56. doi: 10.1111/j.1365-2141.2011.09013.x.
  45. Dorsett Y, McBride KM, Jankovic M, et al. MicroRNA-155 suppresses activation-induced cytidine deaminase-mediated Myc-Igh translocation. Immunity. 2008;28(5):630–8. doi: 10.1016/j.immuni.2008.04.002.
  46. Costinean S, Zanesi N, Pekarsky Y, et al. Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc Natl Acad Sci USA. 2006;103(18):7024–9. doi: 10.1073/pnas.0602266103.
  47. Kluiver J, Poppema S, de Jong D, et al. BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J Pathol. 2005;207(2):243–9. doi: 10.1002/path.1825.
  48. Eis PS, Tam W, Sun L, et al. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc Natl Acad Sci USA. 2005;102(10):3627–32. doi: 10.1073/pnas.0500613102.
  49. Lawrie CH, Soneji S, Marafioti T, et al. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int J Cancer. 2007;121(5):1156–61. doi: 10.1002/ijc.22800.
  50. O’Connell RM, Chaudhuri AA, Rao DS, et al. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc Natl Acad Sci USA. 2009;106(17):7113–8. doi: 10.1073/pnas.0902636106.
  51. Yamanaka Y, Tagawa H, Takahashi N, et al. Aberrant overexpression of microRNAs activate AKT signaling via down-regulation of tumor suppressors in natural killer-cell lymphoma/leukemia. Blood. 2009;114(15):3265–75. doi: 10.1182/blood-2009-06-222794.
  52. Pedersen IM, Otero D, Kao E, et al. Onco-miR-155 targets SHIP1 to promote TNFalpha-dependent growth of B cell lymphomas. EMBO Mol Med. 2009;1(5):288–95. doi: 10.1002/emmm.200900028.
  53. O’Connell RM, Rao DS, Chaudhuri AA, et al. Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder. J Exp Med. 2008;205(3):585–94. doi: 10.1084/jem.20072108.
  54. Roehle A, Hoefig KP, Repsilber D, et al. MicroRNA signatures characterize diffuse large B-cell lymphomas and follicular lymphomas. Br J Haematol. 2008;142(5):732–44. doi: 10.1111/j.1365-2141.2008.07237.x.
  55. Lawrie CH, Chi J, Taylor S, et al. Expression of microRNAs in diffuse large B cell lymphoma is associated with immunophenotype, survival and transformation from follicular lymphoma. J Cell Mol Med. 2009;13(7):1248–60. doi: 10.1111/j.1582-4934.2008.00628.x.
  56. Pichiorri F, Suh SS, Ladetto M, et al. MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc Natl Acad Sci USA. 2008;105(35):12885–90. doi: 10.1073/pnas.0806202105.
  57. Loffler D, Brocke-Heidrich K, Pfeifer G, et al. Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood. 2007;110(4):1330–3.
  58. Wang X, Li C, Ju S, et al. Myeloma cell adhesion to bone marrow stromal cells confers drug resistance by microRNA-21 up-regulation. Leuk Lymphoma. 2011;52(10):1991–8. doi: 10.3109/10428194.2011.591004.
  59. Dimopoulos K, Gimsing P, Gronbaek K. Aberrant microRNA expression in multiple myeloma. Eur J Haematol. 2013;91(2):95–105. doi: 10.1111/ejh.12124.
  60. Chen RW, Bemis LT, Amato CM, et al. Truncation in CCND1 mRNA alters miR-16-1 regulation in mantle cell lymphoma. Blood. 2008;112(3):822–9. doi: 10.1182/blood-2008-03-142182.
  61. Deshpande A, Pastore A, Deshpande AJ, et al. 3¢UTR mediated regulation of the cyclin D1 proto-oncogene. Cell Cycle. 2009;8(21):3592–600. doi: 10.4161/cc.8.21.9993.
  62. Rao E, Jiang C, Ji M, et al. The miRNA-17 approximately 92 cluster mediates chemoresistance and enhances tumor growth in mantle cell lymphoma via PI3K/AKT pathway activation. Leukemia. 2012;26(5):1064–72. doi: 10.1038/leu.2011.305.
  63. Van Vlierberghe P, De Weer A, Mestdagh P, et al. Comparison of miRNA profiles of microdissected Hodgkin/Reed-Sternberg cells and Hodgkin cell lines versus CD77+ B-cells reveals a distinct subset of differentially expressed miRNAs. Br J Haematol. 2009;147(5):686–90. doi: 10.1111/j.1365-2141.2009.07909.x.
  64. Calin GA, Liu CG, Sevignani C, et al. MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA. 2004;101(32):11755–60. doi: 10.1073/pnas.0404432101.
  65. Calin GA, Ferracin M, Cimmino A, et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med. 2005;353(17):1793–801. doi: 10.1056/nejmoa050995.
  66. Moussay E, Palissot V, Vallar L, et al. Determination of genes and microRNAs involved in the resistance to fludarabine in vivo in chronic lymphocytic leukemia. Mol Cancer. 2010;9(1):115. doi: 10.1186/1476-4598-9-115.
  67. Li Z, Lu J, Sun M, et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci USA. 2008;105(40):15535–40. doi: 10.1073/pnas.0808266105.
  68. Jongen-Lavrencic M, Sun SM, Dijkstra MK, et al. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood. 2008;111(10):5078–85. doi: 10.1182/blood-2008-01-133355.
  69. Maki K, Yamagata T, Sugita F, et al. Aberrant expression of MIR9 indicates poor prognosis in acute myeloid leukaemia. Br J Haematol. 2012;158(2):283–5. doi: 10.1111/j.1365-2141.2012.09118.x.
  70. Ishida M, Selaru FM. miRNA-Based Therapeutic Strategies. Curr Anesth Rep. 2013;1(1):63–70. doi: 10.1007/s40139-012-0004-5.
  71. Czauderna F, Fechtner M, Dames S, et al. Structural variations and stabilising modifications of synthetic siRNAs in mammalian cells. Nucl Acids Res. 2003;31(11):2705–16. doi: 10.1093/nar/gkg393.
  72. Davis S, Propp S, Freier SM, et al. Potent inhibition of microRNA in vivo without degradation. Nucl Acids Res. 2009;37(1):70–7. doi: 10.1093/nar/gkn904.
  73. Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368(18):1685–94. doi: 10.1056/nejmoa1209026.
  74. Qiu Z, Dai Y. Roadmap of miR-122-related clinical application from bench to bedside. Expert Opin Invest Drugs. 2014;23(3):347–55. doi: 10.1517/13543784.2014.867327.
  75. Di Martino MT, Campani V, Misso G, et al. In Vivo Activity of MiR-34a Mimics Delivered by Stable Nucleic Acid Lipid Particles (SNALPs) against Multiple Myeloma. PloS One. 2014;9(2):e90005. doi: 10.1371/journal.pone.0090005.
  76. Velu CS, Chaubey A, Phelan JD, et al. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J Clin Invest. 2014;124(1):222–36. doi: 10.1172/jci66005.
  77. Huang X, Schwind S, Yu B, et al. Targeted delivery of microRNA-29b by transferrin-conjugated anionic lipopolyplex nanoparticles: a novel therapeutic strategy in acute myeloid leukemia. Clin Cancer Res. 2013;19(9):2355–67. doi: 10.1158/1078-0432.CCR-12-3191.
  78. Gong JN, Yu J, Lin HS, et al. The role, mechanism and potentially therapeutic application of microRNA-29 family in acute myeloid leukemia. Cell Death Differ. 2014;21(1):100–12. doi: 10.1038/cdd.2013.133.
  79. Ito M, Teshima K, Ikeda S, et al. MicroRNA-150 inhibits tumor invasion and metastasis by targeting the chemokine receptor CCR6 in advanced cutaneous T-cell lymphoma. Blood. 2014;123:1499–511. doi: 10.1182/blood-2013-09-527739.

Лечение распространенных стадий лимфомы Ходжкина: обзор литературы

А.А. Леонтьева, Е.А. Демина

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

Для переписки: Анна Александровна Леонтьева, аспирант, Каширское ш., д. 24, Москва, Российская Федерация, 115478; тел.: +7(499)324-90-89; e-mail: aurevoir-nut@yandex.ru

Для цитирования: Леонтьева А.А., Демина Е.А. Лечение распространенных стадий лимфомы Ходжкина: обзор литературы. Клиническая онкогематология. 2015;8(3):255–66.


РЕФЕРАТ

В последнее десятилетие крупные исследовательские центры в Европе и США, имеющие большие базы данных, провели комплексный анализ эффективности лечебных программ, поздних осложнений терапии и показателей длительной выживаемости больных с распространенными стадиями лимфомы Ходжкина. Этот анализ позволил разработать и предложить практической медицине новые программы, отличающиеся большей эффективностью, и начать поиск менее токсичных вариантов лечения. Однако в отечественной литературе такой комплексный анализ не представлен. Имеющиеся публикации и научные исследования освещают лишь отдельные аспекты диагностики и лечения лимфомы Ходжкина, или в них выборочно обсуждаются проблемы осложнений. Предлагаемый обзор позволяет читателю проследить путь, пройденный в лечении распространенных стадий лимфомы Ходжкина за 75 лет — от абсолютно пессимистического прогноза до современных высоких результатов с дальнейшим совершенствованием терапии этой злокачественной опухоли.


Ключевые слова: лимфома Ходжкина, распространенные стадии, лечение, эффективность, токсичность.

Получено: 20 февраля 2015 г.

Принято в печать: 28 мая 2015 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Hodgkin T. On some morbid appearances of the absorbent glands and spleen. Med Chir Trans. 1832;17:68–114. doi: 10.1177/095952873201700106.
  2. Bonadonna G. Historical review of Hodgkin’s disease. Br J Haematol. 2000;110(3):504–11. doi: 10.1046/j.1365-2141.2000.02197.x.
  3. Diehl V, guest ed. Bailliere’s Clinical Haematology. International Practice and Research. Hodgkin’s Disease. London, Philadelphia, Sydney: Bailliere Tindall; 1996.
  4. Переслегин И.А., Филькова Е.М. Лимфогранулематоз. М.: Медицина, 1975.
    [Pereslegin IA, Fil‘kova EM. Limfogranulematoz. (Lymphogranulomatosis.) Moscow: Meditsina Publ.; 1975. (In Russ)]
  5. Sternberg C. Uber eine Eigenartige, unter dem Bilde der Pseudoleukemie verlaufende Tuberkulose des lymphatische Apparates. Zschr F Heilkunde. 1898;19:21–90.
  6. Reed D. On the pathological changes in Hodgkin’s disease, with especial reference to its relation to tuberculosis. Johns Hopkins Hosp Bull. 1902;10:133–96.
  7. Diehl V, ed. 25 Years German Hodgkin Study Group. Medizin & Wissen; 2004.
  8. Hjalgrim H, Askling J, Sorensen P, et al. Risk of Hodgkin’s disease and other cancer after infectious mononucleosis. J Natl Cancer Inst. 2000;92(18):1522–8. doi: 10.1093/jnci/92.18.1522.
  9. Демина Е.А. Современная терапия первичных больных лимфомой Ходжкина: Автореф. дис. ¼ д-ра мед. наук. М., 2006.
    [Demina EA. Sovremennaya terapiya pervichnykh bol’nykh limfomoi Khodzhkina. (Modern management of primary Hodgkin’s lymphoma patients.) [dissertation] Moscow; 2006. (In Russ)]
  10. Lukes RJ, Butler JJ, Hicks ED. Natural history of Hodgkin’s disease as related to its pathologic picture. Cancer. 1966;19(3):317–44. doi: 10.1002/1097-0142(196603)19:3<317::aid-cncr2820190304>3.0.co;2-o.
  11. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  12. Engert A, Horning SJ, eds. Hematologic malignancies: Hodgkin lymphoma. A Comprehensive Update on Diagnostics and Clinics. Berlin, Heidelberg: Springer; 2011. pp. 65–76.
  13. Давыдов М.И., Аксель Е.М. Статистика злокачественных новообразований в России и странах СНГ в 2009 г. Вестник РОНЦ им. Н.Н. Блохина РАМН. 2011;22(3, прил. 1).
    [Davydov MI, Aksel’ EM. Cancer statistica in Russia and CIS in 2009. Vestnik RONTs im. N.N. Blokhina RAMN. 2011;22(3 Suppl 1). (In Russ)]
  14. Granger W, Whitaker R. Hodgkin’s disease in bone with special reference to periosteal reaction. Br J Radiol. 1967;40(480):939–48. doi: 10.1259/0007-1285-40-480-939.
  15. Bichel J. The alcohol-intolerance syndrome in Hodgkin’s disease. Acta Med Scand. 1959;164(2):105–12. doi: 10.1111/j.0954-6820.1959.tb00168.x.
  16. James AH. Hodgkin’s disease with and without alcohol-induced pain. A clinical and histological comparison. Q J Med. 1960;29:47–66.
  17. Winiwarter A. Du lymphome malin et du lymphosarcome et de leur traitement. Arch F Arch Klin Chir. 1875;18:98–102.
  18. Pussey WA. Cases of sarcoma and of Hodgkin’s disease treated by exposures to X-rays: preliminary report. JAMA. 1902;98:166–9. doi: 10.1001/jama.1902.62480030024001h.
  19. Gilbert R. La roengentherapie de la granulematise maligne. J Radiol Electrol. 1925;9:509–14.
  20. Демина Е.А. Лимфома Ходжкина: от Томаса Ходжкина до наших дней. Клиническая онкогематология. 2008;1(2):114–8.
    [Demina EA. Hodgkin’s lymphoma: from Thomas Hodgkin till present days. Klinicheskaya onkogematologiya. 2008;1(2):114–8. (In Russ)]
  21. Hoppe RT, Hanlon A, Hanks G, et al. Progress in treatment of Hodgkin’s disease in the United States, 1973 versus 1983: the patterns of care study. Cancer. 1994;74(12):3198–203. doi: 10.1002/1097-0142(19941215)74:12<3198::aid-cncr2820741219>3.0.co;2-9.
  22. Hoppe RT. Radiation therapy in the management of Hodgkin’s disease. Semin Oncol. 1990;17(6):704–15.
  23. Peters MV. A study of survivals in Hodgkin’s disease treated radiologically. Am J Roent. 1950;63:299–311.
  24. Kaplan H. The radical radiotherapy of Hodgkin’s disease. Radiology. 1962;78(4):553–61. doi: 10.1148/78.4.553.
  25. Самочатова Е.В., Владимирская Е.Б., Жесткова Н.М. и др. Болезнь Ходжкина у детей. М.: Алтус, 1997.
    [Samochatova EV, Vladimirskaya EB, Zhestkova NM, et al. Bolezn’ Khodzhkina u detei. (Hodgkin’s disease in children.) Moscow: Altus Publ.; 1997. (In Russ)]
  26. Hoppe RT, Mauch PT, Armitage JO, et al. Hodgkin Lymphoma. 2nd edition. Philadelphia: Lippincott Williams & Wilkins; 2007.
  27. Prosnitz LR, Farber LR, Fisher JJ, et al. Long term remissions with combined modality therapy for advanced Hodgkin’s disease. Cancer. 1976;37(6):2826–33. doi: 10.1002/1097-0142(197606)37:6<2826::aid-cncr2820370638>3.0.co;2-f.
  28. Goodman LS, Wintrobe MM, Dameshek W, et al. Nitrogen mustard therapy; use of methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-chloroethyl) amine hydrochloride for Hodgkin’s disease, lymphosarcoma, leukemia and certain allied and miscellaneous disorders. J Am Med Assoc. 1946;132:126–32.
  29. DeVita VT Jr, Carbone PP. Treatment of Hodgkin’s disease. Med Ann Dist Columbia. 1967;36(4):232–4.
  30. DeVita VT, Serpick AA, et al. Combination chemotherapy in the treatment of advanced Hodgkin’s disease. Ann Intern Med. 1970;73(6):881–95. doi: 10.7326/0003-4819-73-6-881.
  31. Longo DL, Young RC, Wesley M, et al. Twenty years of MOPP therapy for Hodgkin’s disease. J Clin Oncol. 1986;4:1295–306.
  32. Bonadonna G, Valagussa P, Santoro A. Alternating non-cross-resistant combination chemotherapy or MOPP in stage IV Hodgkin’s disease. A report of 8-year results. Ann Intern Med. 1986;104(6):739–46. doi: 10.7326/0003-4819-104-6-739.
  33. Даценко П.В., Паньшин Г.А., Сотников В.М. и др. Новые программы комбинированного лечения лимфомы Ходжкина. Онкогематология. 2007;4:27–35.
    [Datsenko PV, Pan’shin GA, Sotnikov VM, et al. New programs of combined treatment of Hodgkin’s lymphoma. Onkogematologiya. 2007;4:27–35. (In Russ)]
  34. Goldman AJ, Goldie JH. A mathematic model for relating the drug sensitivity of tumors to their spontaneous mutation rate. Cancer Treat Rep. 1979;63(11–12):1727–33.
  35. Santoro A, Bonadonna G, Valagussa P, et al. Long-term results of combined chemotherapy-radiotherapy approach in Hodgkin’s disease: superiority of ABVD plus radiotherapy versus MOPP plus radiotherapy. J Clin Oncol. 1987;5(1):27–37.
  36. Canellos GP, Anderson JR, Propert KJ, et al. Chemotherapy of advanced Hodgkin’s disease with MOPP, ABVD, or MOPP alternating with ABVD. N Engl J Med. 1992;327(21):1478–84. doi: 10.1056/nejm199211193272102.
  37. Stefan DC, Stones D. How much does it cost to treat children with Hodgkin lymphoma in Africa? Leuk Lymphoma. 2009;50(2):196–9. doi: 10.1080/10428190802663205.
  38. Canellos GP, Niedzwiecki D. Long-term follow-up of Hodgkin’s disease trial. N Engl J Med. 2002;346(18):1417–8. doi: 10.1056/nejm200205023461821.
  39. Mauch PV, Armitage JO, Diehl V, et al. Hodgkin’s disease. Philadelphia: Lippincott Williams & Wilkins; 1999.
  40. Specht L. Prognostic factors in Hodgkin’s disease. Cancer Treat Rev. 1991;18(1):21–53. doi: 10.1016/0305-7372(91)90003-i.
  41. DeVita VT, Hellman S, Rosenberg SA. Cancer. Principles & Practice of Oncology. 4th edition. Philadelphia; 1993;1819–58.
  42. Richardson SE, McNamara C. The management of classical Hodgkin’s lymphoma: past, present, and future. Adv Hematol. 2011;2011:865870. doi: 10.1155/2011/865870.
  43. Horning SJ, Hoppe RT, Breslin S, et al. Stanford V and radiotherapy for locally extensive and advanced Hodgkin’s disease: mature results of a prospective clinical trial. J Clin Oncol. 2002;20(3):630–7. doi: 10.1200/jco.20.3.630.
  44. Hoskin PJ, Lowry L, Horwich A, et al. Randomized comparison of the Stanford V regimen and ABVD in the treatment of advanced Hodgkin’s Lymphoma: United Kingdom National Cancer Research Institute Lymphoma Group Study ISRCTN 64141244. J Clin Oncol. 2009;27(32):5390–6. doi: 10.1200/jco.2009.23.3239.
  45. Diehl V, Franklin J, Pfreundschuh M, et al. Standard and increased-dose BEACOPP chemotherapy compared with COPP-ABVD for advanced Hodgkin’s disease. N Engl J Med. 2003;348(24):2386–95. doi: 10.1056/nejmoa022473.
  46. Engert A, Diehl V, Franklin J, et al. Escalated-dose BEACOPP in the treatment of patients with advanced-stage Hodgkin’s lymphoma: 10 years of follow-up of the GHSG HD9 study. J Clin Oncol. 2009;27(27):4548–54. doi: 10.1200/jco.2008.19.8820.
  47. Ларина Ю.В., Миненко С.В., Биячуев Э.Р. и др. Лечение распространенных форм лимфомы Ходжкина у подростков и молодых взрослых. Проблема эффективности и токсичности. Онкогематология. 2014;1:11–8.
    [Larina YuV, Minenko SV, Biyachuev ER, et al. Treatment of advance stage Hodgkin’s lymphomas in adolescents and young adults. Efficacy and toxicity problem. Onkogematologiya. 2014;1:11–8. (In Russ)]
  48. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N Engl J Med. 1998;339(21):1506–14.
  49. Diehl V. German Hodgkin Study Group. Haematologica. 2007;92(s5):21, abstract I071.
  50. Богатырева Т.И., Столбовой А.В., Копп М.Ю. и др. Лимфома Ходжкина: трудности на пути реализации стандартов лечения и их преодоление. Врач. 2011;12:34–40.
    [Bogatyreva TI, Stolbovoi AV, Kopp MYu, et al. Hodgkin’s lymphoma: difficulties in implementing treatment standards and ways to overcome them. Vrach. 2011;12:34–40. (In Russ)]
  51. Капланов К.Д., Шипаева А.Л., Васильева В.А. и др. Эффективность программ химиотерапии первой линии при различных стадиях лимфомы Ходжкина. Клиническая онкогематология. 2012;5(1):22–9.
    [Kaplanov KD, Shipaeva AL, Vasil’eva VA, et al. Efficacy of first line chemotherapy programs for different stages of Hodgkin’s lymphomas. Klinicheskaya onkogematologiya. 2012;5(1):22–9. (In Russ)]
  52. Borchmann P, Diehl V, Goergen H, et al. Combined modality treatment with intensified chemotherapy and dose-reduced involved field radiotherapy in patients with early unfavourable Hodgkin Lymphoma: final analysis of the German Hodgkin Study Group HD 11 trial. Blood. 2009;114:299–300.
  53. Thomas J, Ferm C, Noordijk E, et al. Results of the EORTC-GELA H9 randomized trials: the H9-F trials (comparing 3 radiation dose levels) and H9-U trials (comparing 3 chemotherapy schemes) in patients with favorable or unfavorable early stage Hodgkin’s lymphoma (HL). Haematologica. 2007;92(s5):27.
  54. Skoetz N, Trelle S, Rancea M, et al. Effect of initial treatment strategy on survival of patients with advanced-stage Hodgkin’s lymphoma: a systematic review and network meta-analysis. Lancet Oncol. 2013;14(10):943–52. doi: 10.1016/s1470-2045(13)70341-3.
  55. Kobe C, Dietlein M, Franklin J, et al. Positron emission tomography has a high negative predictive value for progression or early relapse for patients with residual disease after first-line chemotherapy in advanced-stage Hodgkin lymphoma. Blood. 2008;112(10):3989–94. doi: 10.1182/blood-2008-06-155820.
  56. Chesson B, Pfistner B, Juweid M, et al. Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579–86. doi: 10.1200/jco.2006.09.2403.
  57. Juweid ME, Stroobants S, Hoekstra OS, et al. Use of positron emission tomography for response assessment of lymphoma: consensus of the Imaging Subcommittee of International Harmonization Project in Lymphoma. J Clin Oncol. 2007;25(5):571–8. doi: 10.1200/jco.2006.08.2305.
  58. Шахтарина С.В., Павлов В.В., Даниленко А.А., Афанасова Н.В. Лечение больных лимфомой Ходжкина с локальными стадиями: опыт медицинского радиологического научного центра. Онкогематология. 2007;4:36–46.
    [Shakhtarina SV, Pavlov VV, Danilenko AA, Afanasova NV. Treatment of patients with local stages Hodgkin’s lymphomas: experience of medical radiological scientific center. Onkogematologiya. 2007;4:36–46. (In Russ)]
  59. Gallamini A, Hutchings M, Rigacci I, et al. Early interim FDG-PET overshadows the prognostic role of IPS in advanced-stage Hodgkin’s lymphoma treated by conventional ABVD therapy. Haematologica. 2007;32(s5): Abstract C022.
  60. Hoppe RT. Hodgkin’s disease: Second cancer after treatment Hodgkin’s disease: Complications of therapy and excess mortality. Ann Oncol. 1997;8(1):115.
  61. Шахтарина С.В., Даниленко А.А., Павлов В.В. Злокачественные новообразования у больных лимфомой Ходжкина после лучевой терапии по радикальной программе и комбинированной химиолучевой терапии. Клиническая онкогематология. 2008;1(3):246–51.
    [Shakhtarina SV, Danilenko AA, Pavlov VV. Malignant neoplasms in Hodgkin’s lymphoma patients after radiation therapy (according to radical program) and combined chemoradiation therapy. Klinicheskaya onkogematologiya. 2008;1(3):246–51. (In Russ)]
  62. Ильин Н.В., Виноградова Ю.Н. Поздние осложнения терапии больных лимфомой Ходжкина. Практическая онкология. 2007;8(2):96–101.
    [Il’in NV, Vinogradova YuN. Delayed treatment complications in Hodgkin’s lymphoma patients. Prakticheskaya onkologiya. 2007;8(2):96–101. (In Russ)]
  63. Поддубная И.В. Неходжкинские лимфомы. В кн.: Клиническая онкогематология. Под ред. М.А. Волковой. М.: Медицина, 2007. C. 724–71.
    [Poddubnaya IV. Non-Hodgkin’s lymphomas. In: Volkova MA, ed. Klinicheskaya onkogematologiya. (Clinical oncohematology.) Moscow: Meditsina Publ.; 2007. pp. 724–71. (In Russ)]
  64. Поддубная И.В. Обоснование лечебной тактики при неходжкинских лимфомах. Современная онкология. 2002;4(1):3–7.
    [Poddubnaya IV. Rationale for therapeutic management of non-Hodgkin’s lymphoma. Sovremennaya onkologiya. 2002;4(1):3–7. (In Russ)]
  65. Federico M, Luminari S, Iannitto E, et al. ABVD compared with BEACOPP compared with CEC for the initial treatment of patients with advanced Hodgkin’s lymphoma: results from the HD2000 Gruppo Italiano per lo Studio dei Limfomi Trial. J Clin Oncol. 2009;27(5):805–11. doi: 10.1200/jco.2008.17.0910.
  66. Engert A, Haverkamp H, Kobe C, et al. Reduced-intensity chemotherapy and PET-guided radiotherapy in patients with advanced stage Hodgkin’s lymphoma (HD15 trial): a randomised, open-label, phase 3 non-inferiority trial. The Lancet. 2012;379(9828):1791–9. doi: 10.1016/S0140-6736(11)61940-5.
  67. Bovelli D, Plataniotis G, Roila F. Кардиологическая токсичность химиотерапевтических препаратов и заболевания сердца, обусловленные проведением лучевой терапии. В кн.: Минимальные клинические рекомендации Европейского общества медицинской онкологии. М., 2010. C. 423–33.
    [Bovelli D, Plataniotis G, Roila F. Cardiac toxicity of chemotherapeutic agents and radiotherapy-associated heart diseases. In: Minimal’nye klinicheskie rekomendatsii Evropeiskogo obshchestva meditsinskoi onkologii. (European Society for Medical Oncology (ESMO) Minimum Clinical Recommendations.) Moscow; 2010. pp. 423–33. (In Russ)]
  68. Поддубная И.В., Орел Н.Ф. Кардиотоксичность. В кн.: Руководство по химиотерапии опухолевых заболеваний. Под ред. Н.И. Переводчиковой. М.: Практическая медицина, 2011. С. 435–6.
    [Poddubnaya IV, Orel NF. Cardiac toxicity. In: Perevodchikova NI, ed. Rukovodstvo po khimioterapii opukholevykh zabolevanii. (Guidelines for chemotherapy of tumors.) Moscow: Prakticheskaya Meditsina Publ.; 2011. pp. 435–6. (In Russ)]
  69. Емелина Е.И. Состояние сердечно-сосудистой системы у больных лимфопролиферативными заболеваниями, получавших антрациклиновые антибиотики: Дис. ¼ канд. мед. наук. М., 2007. С. 10–36.
    [Emelina EI. Sostoyanie serdechno-sosudistoi sistemy u bol’nykh limfoproliferativnymi zabolevaniyami, poluchavshikh antratsiklinovye antibiotiki. (Condition of the cardiovascular system inpatients with lymphoproliferative disorders treated with anthracycline antibiotics.) [dissertation] Moscow; 2007. pp. 10–36. (In Russ)]
  70. Матяш М.Г., Кравчук Т.Л., Высоцкая В.В. и др. Индуцированная антрациклинами кардиотоксичность: механизмы развития и клинические проявления. Сибирский онкологический журнал. 2008;6(30):66–75.
    [Matyash MG, Kravchuk TL, Vysotskaya VV, et al. Anthracycline-induced cardiac toxicity: mechanisms of development and clinical manifestations. Sibirskii onkologicheskii zhurnal. 2008;6(30):66–75. (In Russ)]
  71. Семенова А.Е. Кардио- и нейротоксичность противоопухолевых препаратов (патогенез, клиника, профилактика и лечение). Практическая онкология. 2009;10(3):168–76.
    [Semenova AE. Cardiac and neurotoxicity of anti-tumor agents (pathogenesis, clinical presentation, prevention, and treatment). Prakticheskaya onkologiya. 2009;10(3):168–76. (In Russ)]
  72. Brana I, Tabernero J. Cardiotoxicity. Ann Oncol. 2010;21(Suppl 7):173–9. doi: 10.1093/annonc/mdq295.
  73. Гендлин Г.Е., Сторожаков Г.И., Шуйкова К.В. и др. Острые сердечно-сосудистые события во время применения противоопухолевых химиопрепаратов: клинические наблюдения. Клиническая онкогематология. 2011;4(2):155–64.
    [Gendlin GE, Storozhakov GI, Shuikova KV, et al. Acute cardiovascular events during treatment with anti-tumor chemotherapeutic agents: clinical observations. Klinicheskaya onkogematologiya. 2011;4(2):155–64. (In Russ)]
  74. Allen A. The cardiotoxicity of chemotherapeutic drugs. Semin Oncol. 1992;19(5):529–42.
  75. Gewlling M, Mertens L, Moerman P, et al. Idiopathic restrictive cardiomyopathy in childhood. Eur Heart J. 1996;17(9):1413–20. doi: 10.1093/oxfordjournals.eurheartj.a015076.
  76. Матяш М.Г., Кравчук Т.Л., Высоцкая В.В. и др. Неантрациклиновая кардиотоксичность. Сибирский онкологический журнал. 2009;5(35):73–82.
    [Matyash MG, Kravchuk TL, Vysotskaya VV, et al. Non-anthracycline-related cardiac toxicity. Sibirskii onkologicheskii zhurnal. 2009;5(35):73–82. (In Russ)]
  77. Escoto H, Ringewald J, Kalpatthi R. Etoposide-related cardiotoxicity in a child with haemophagocytic lymphohistiocytosis. J Cardiol Young. 2010;20(1):105–7. doi: 10.1017/s1047951109991272.
  78. Calvo-Romero JM, Fernandez-Soria-Pantoja R, Arrebola-Garcia JD. Ischemic heart disease associated with vincristine and doxorubicin chemotherapy. Ann Pharmacother. 2001;35(11):1403–5. doi: 10.1345/aph.10358.
  79. Bovelli D, Plataniotis G, Roila F. Cardiotoxicity of chemotherapeutic agents and radiotherapy-related heart disease: ESMO Clinical Practice Guidelines. Ann Oncol. 2010;21(Suppl 5):277–82. doi: 10.1093/annonc/mdq200.
  80. Meirow D, Lewis H, Nugent D, Epstein M. Subclinical depletion of primordial follicular reserve in mice treated with cyclophosphamide: clinical importance and proposed accurate investigative tool. Hum Reprod. 1999;14(7):1903–7. doi: 10.1093/humrep/14.7.1903.
  81. Шахтарина С.В., Даниленко А.А., Щелконогова Л.Н., Павлов В.В. Беременность, роды и состояние здоровья детей, родившихся у женщин с лимфомой Ходжкина после лучевого или комбинированного химиолучевого лечения. Клиническая онкогематология. 2012;5(3):218–24.
    [Shakhtarina SV, Danilenko AA, Shchelkonogova LN, Pavlov VV. Pregnancy, delivery, and health state of children born to women with Hodgkin’s lymphoma after radiation or combined chemoradiation therapy. Klinicheskaya onkogematologiya. 2012;5(3):218–24. (In Russ)]
  82. Familiary G, Caggiani A, Nottola SA, et al. Ultrastructure of human ovarian primordial follicles after combination chemotherapy for Hodgkin’s disease. Hum Reprod. 1993;8(12):2080–7.
  83. Zhang Y, Xiao Z, Wang Y, et al. Gonadotropin-releasing hormone for preservation of ovarian function during chemotherapy in lymphoma patients of reproductive age: a summary based on 434 patients. PLoS One. 2013;8(11):e80444. doi: 10.1371/journal.pone.0080444.
  84. Huser M, Crha I, Ventruba P, et al. Prevention of ovarian function damage by a GnRh analogue during chemotherapy in Hodgkin lymphoma patients. Hum Reprod. 2008;23(4):863–8. doi: 10.1093/humrep/den005.
  85. Kulkarni SS, Sastry PS, Saikia TK, et al. Gonadal function following ABVD therapy for Hodgkin’s disease. J Clin Oncol. 1997;20(4):354–7. doi: 10.1097/00000421-199708000-00006.
  86. Пивник А.В., Расстригин Н.А., Моисеева Т.Н. и др. Результаты лечения лимфогранулематоза по протоколу МОРР-ABVD в сочетании с лучевой терапией (десятилетнее наблюдение). Терапевтический архив. 2006;8:57–62.
    [Pivnik AV, Rasstrigin NA, Moiseeva TN, et al. Results of treatment of lymphogranulematosis according to the МОРР-ABVD protocol in combination with radiation therapy (10-year follow-up). Terapevticheskii arkhiv. 2006;8:57–62. (In Russ)]
  87. Redman JR, Bajorunas DR, Goldstein MC, et al. Semen cryopreservation and artificial insemination for Hodgkin’s disease. J Clin Oncol. 1987;5(2):233–8.
  88. Винокуров А.А., Варфоломеева С.Р., Тарусин Д.И. Гонадотоксичность терапии лимфомы Ходжкина у подростков и молодых мужчин: актуальность проблемы и пути решения (обзор литературы). Онкогематология. 2011;2:12–8.
    [Vinokurov AA, Varfolomeeva SR, Tarusin DI. Gonadal toxicity of treatment for Hodgkin’s lymphoma in adolescents and young adults: topicality of the problem and ways of its solution (literature review). Onkogematologiya. 2011;2:12–8. (In Russ)]
  89. Sieniawski M, Reineke T, Nogova L, et al. Fertility in male patients with advanced Hodgkin’s lymphoma treated with BEACOPP: a report of the German Hodgkin Study Group (GHSG). Blood. 2008;111(1):71–6. doi: 10.1182/blood-2007-02-073544.
  90. Винокуров А.А., Варфоломеева С.Р., Тарусин Д.И., Моисеева Т.Н. Оценка гонадотоксичности терапии по схеме ВЕАСОРР-14 у молодых мужчин, излеченных от лимфомы Ходжкина. Клиническая онкогематология. 2011;4(3):235–9.
    [Vinokurov AA, Varfolomeeva SR, Tarusin DI, Moiseeva TN. Evaluation of gonadal toxicity of ВЕАСОРР-14 treatment regimen in young males cured from Hodgkin’s lymphoma. Klinicheskaya onkogematologiya. 2011;4(3):235–9. (In Russ)]
  91. Даниленко А.А., Шахтарина С.В., Афанасова Н.В., Павлов В.В. Изменения в легких у больных лимфомой Ходжкина после химиотерапии по схемам СОРР, ABVD, ВЕАСОРР и облучения средостения в суммарной очаговой дозе 20–30 Грей. Клиническая онкогематология. 2010;3(4):354–8.
    [Danilenko AA, Shakhtarina SV, Afanasova NV, Pavlov VV. Changes in lugs of patients with Hodgkin’s lymphoma after chemotherapy according to СОРР, ABVD, ВЕАСОРР and radiation of mediastinum (total focal dose of 20–30 Gray). Klinicheskaya onkogematologiya. 2010;3(4):354–8. (In Russ)]
  92. Даценко П.В. Сбалансированное сочетание лучевого и лекарственного компонентов при комплексном лечении лимфогранулематоза: Автореф. дис. ¼ д-ра мед. наук. М., 2004.
    [Datsenko PV. Sbalansirovannoe sochetanie luchevogo i lekarstvennogo komponentov pri kompleksnom lechenii limfogranulematoza. (Balanced combination of radiation and chemotherapy in complex treatment of lymphogranulematosis.) [dissertation] Moscow; 2004. (In Russ)]
  93. Duggan DB, Petroni GR, Johnson JL, et al. Randomized comparison of ABVD and MOPP/ABV hybrid for the treatment of advanced Hodgkin’s disease: Report of an intergroup trial. J Clin Oncol. 2003;21(4):607–14. doi: 10.1200/jco.2003.12.086.
  94. Diehl V, Franklin J, Pfreundschuh M, et al. Standard and increased dose BEACOPP chemotherapy compared with COPP-ABVD for advanced Hodgkin’s disease. N Engl J Med. 2003;348(24):2386–95. doi: 10.1056/nejmoa022473.
  95. Onuma T, Holland JF, Hosi S, et al. Microbiological assay of bleomycin: inactivation, tissue distribution, and clearance. Cancer. 1974;33(5):1230–8. doi: 10.1002/1097-0142(197405)33:5<1230::aid-cncr2820330507>3.0.co;2-c.
  96. Santrach PJ, Askin FB, Wells RJ, et al. Nodular form of bleomycin-related pulmonary injury in patients with osteogenic sarcoma. Cancer. 1989;64(4):806–11. doi: 10.1002/1097-0142(19890815)64:4<806::aid-cncr2820640407>3.0.co;2-x.
  97. Holoye PY, Luna MH, Mackay B, et al. Bleomycin hypersensitivity pneumonitis. Ann Intern Med. 1978;88(1):47–9. doi: 10.7326/0003-4819-88-1-47.
  98. Martin WG, Ristow KM, Habermann TM, et al. Bleomycin pulmonary toxicity has a negative impact on the outcome of patients with Hodgkin’s lymphoma. J Clin Oncol. 2005;23(30):7614–20. doi: 10.1200/jco.2005.02.7243.
  99. Carlson RW, Sikic BJ. Continuous infusion or bolus injection in cancer chemotherapy. Ann Intern Med. 1983;99(6):823–33. doi: 10.7326/0003-4819-99-6-823.
  100. Samuals MI, Johnson PE, Holoye PY, et al. Large-dose bleomycin therapy and pulmonary toxicity. JAMA. 1976;235(11):1117–20. doi: 10.1001/jama.1976.03260370025026.
  101. Catravas LD, Laza JS, Dobuker KJ, et al. Pulmonary endothelial dysfunction in the presence or absence of interstitial injury induced by intratracheally injected bleomycin in rabbits. Am Rev Respir Dis. 1983;128(4):740–6.
  102. Simpson AB, Paul J, Graham J, et al. Fatal bleomycin pulmonary toxicity in the west of Scotland 1991–95; a review of patients with germ cells tumors. Br J Cancer. 1998;78(8):1061–6. doi: 10.1038/bjc.1998.628.
  103. Lower EE, Strohofer S, Baughman RP. Bleomycin causes alveolar macrophages from cigarette smokers to release hydrogen peroxide. Am J Med Sci. 1988;295(3):193–7. doi: 10.1097/00000441-198803000-00006.
  104. Boll B, Gorgen H, Fuchs M, et al. Feasibility and efficacy of ABVD in elderly Hodgkin lymphoma patients: analysis of two randomized prospective multicenter trials of the German Hodgkin Study Group (HD10 and HD11). Blood (ASH Annual Meeting Abstracts). 2010;116:418.
  105. Proctor SJ, Wilkinson J, Culligan D, et al. Comparative clinical responses of three chemotherapy schedules (VEPEMB, ABVD, CLVPP) in 175 Hodgkin lymphoma patients over 60 YS evaluated as part of the SHIELD (Hodgkin Elderly) study. Ann Oncol. 2011;22(4):117–8.
  106. Evens AM, Hong F, Gordon LI, et al. Efficacy and tolerability of ABVD and Stanford V for Elderly Advanced-Stage Hodgkin-Lymphoma (HL): analysis from the Phase III Randomized US Intergroup Trial E2496. Ann Oncol. 2011;22(4):118.
  107. Behringer K, Goergen H, Borchmann P, et al. Impact of bleomycin and dacarbazine within the ABVD regimen in the treatment of early-stage favorable Hodgkin lymphoma: final results of the GHSG HD13 trial. EHA. 2014: Abstract S1290.
  108. Hirsch A, Vander EN, Straus DJ, et al. Effect of ABVD chemotherapy with and without mantle or mediastinal irradiation on pulmonary function and symptoms in early-stage Hodgkin’s disease. J Clin Oncol. 1996;14(4):1297–305.
  109. Horning SJ, Adhikary A, Rizk N, et al. Effect of treatment for Hodgkin’s disease on pulmonary function: results of a prospective study. J Clin Oncol. 1994;12(2):297–305.
  110. Kaplan HS. Hodgkin’s Disease. 2nd edition. Cambridge: Harvard University Press; 1980.
  111. Prosnitz LR, Farber LR, Fisher JJ, et al. Long term remissions with combined modality therapy for advanced Hodgkin’s disease. Cancer. 1976;37(6):2826–33. doi: 10.1002/1097-0142(197606)37:6<2826::aid-cncr2820370638>3.0.co;2-f.
  112. Mauch PV, Armitage JO, Diehl V, et al, eds. Hodgkin’s disease. Philadelphia; 1999.
  113. Brincker H, Bentzen SM. A re-analysis of available dose-response and time-dose data in Hodgkin’s disease. J Radiother Oncol. 1994;30(3):227–30. doi: 10.1016/0167-8140(94)90462-6.
  114. Loeffler M, Diehl V, Pfreundschuh M, et al. Dose-response relationship of complementary radiotherapy following four cycles of combination chemotherapy in intermediate-stage Hodgkin’s disease. J Clin Oncol. 1997;15(6):2275–87. doi: 10.1016/s1278-3218(98)89074-4.
  115. Ярмоненко С.П., Вайнсон А.А. Радиобиология человека и животных. М.: Высшая школа, 2004.
    [Yarmonenko SP, Vainson AA. Radiobiologiya cheloveka i zhivotnykh. (Radiobiology of human and animal.) Moscow: Vysshaya shkola Publ.; 2004. (In Russ)]
  116. Jakobsson PA, Littbrand B. Fractionation scheme with low individual tumor doses and high total dose. Actа Radiol Ther Phys Biol. 1973;12(4):337–46. doi: 10.3109/02841867309131099.
  117. Акимов А.А., Ильин Н.В. Некоторые биологические аспекты лимфомы Ходжкина и новые подходы к ее терапии. Вопросы онкологии. 2003;49(1):31–40.
    [Akimov AA, Il’in NV. Some biological aspects of Hodgkin’s lymphoma and new approaches to its treatment. Voprosy onkologii. 2003;49(1):31–40. (In Russ)]
  118. Hall EJ. Clinical response of normal tissues. In: Hall EJ, ed. Radiobiology for the Radiologist. 5th edition. Philadelphia: Lippincott Williams &Wilkins, 2000. pp. 352.
  119. Ильин Н.В., Виноградова Ю.Н., Николаева Е.Н., Смирнова Е.В. Значение мультифракционирования дозы радиации при первичном лучевом лечении больных лимфомой Ходжкина. Онкогематология. 2007;4:47–52.
    [Il’in NV, Vinogradova YuN, Nikolaeva EN, Smirnova EV. Value of multifractionation radiotherapy dose for primary treatment of patients with Hodgkin’s lymphoma. Onkogematologiya. 2007;4:47–52. (In Russ)]
  120. Magagnoli M, Marzo K, Balzarotti M, et al. Dimension of Residual CT Scan Mass in Hodgkin’s Lymphoma (HL) Is a Negative Prognostic Factor in Patients with PET Negative After Chemo +/– Radiotherapy. Blood (ASH Annual Meeting Abstracts). 2011;118:93.
  121. Russo F, Corazzelli G, Frigeri F, et al. A phase II study of dose-dense and dose-intense ABVD (ABVDDD-DI) without consolidation radiotherapy in patients with advanced Hodgkin lymphoma. Br J Haematol. 2014;166(1):118–29. doi: 10.1111/bjh.12862.
  122. Laskar S, Kumar DP, Khanna N, et al. Radiation therapy for early stage unfavorable Hodgkin lymphoma: is dose reduction feasible? Leuk Lymphoma. 2014;55(10):2356–61. doi: 10.3109/10428194.2013.871631.
  123. Boll B, Bredenfeld H, Gorgen H, et al. Phase 2 study of PVAG (prednisone, vinblastine, doxorubicin, gemcitabine) in elderly patients with early unfavorable or advanced stage Hodgkin lymphoma. Blood. 2011;118(24):6292–8. doi: 10.1182/blood-2011-07-368167.
  124. Younes A, Oki Y, McLaughlin P, et al. Phase 2 study of rituximab plus ABVD in patients with newly diagnosed classical Hodgkin lymphoma. Blood. 2012;119(18):4123–8. doi: 10.1182/blood-2012-01-405456.
  125. Engert A, Haverkamp H, Kobe C, et al. Reduced-intensity chemotherapy and PET-guided radiotherapy in patients with advanced stage Hodgkin’s lymphoma (HD15 trial): a randomised, open-label, phase 3 non-inferiority trial. The Lancet. 2012;379(9828):1791–9. doi: 10.1016/s0140-6736(11)61940-5.
  126. Younes A, Connors JM, Park S, et al. Brentuximab vedotin combined with ABVD or AVD for patients with newly diagnosed Hodgkin’s lymphoma: a phase 1, open-label, dose-escalation study. Lancet Oncol. 2013;14(13):1348–56. doi: 10.1016/s1470-2045(13)70501-1.
  127. Demina EA, Tumyan GS, Stroyakovskiy DL. Treatment results of six cycles EACOPP-14 ± RT in advanced stage Hodgkin lymphoma. Multicenters study in Russia. 9th International Symposium on Hodgkin Lymphoma, Cologne, Germany, October 12–15, 2013. Haematologica. 2013;98(2): Abstract P013.
  128. Демина Е.А. Дискуссионные вопросы лечения распространенных стадий лимфомы Ходжкина. Материалы XVII Российского онкологического конгресса, Москва, 12–14 ноября 2013 г. Злокачественные опухоли. 2013;2:19–22.
    [Demina EA. Controversial issues of treatment of advanced stage Hodgkin’s lymphoma. (Materials of XVII Russian oncological congress, Moscow, November 12–14, 2013.) Zlokachestvennye opukholi. 2013;2:19–22. (In Russ)]
  129. Younes A, Gopal AK, Smith SE. еt al. Smith еt al. Results of a Pivotal Phase II Study of Brentuximab Vedotin for Patients With Relapsed or Refractory Hodgkin’s Lymphoma. J Clin Oncol. 2012;30(18):2183–9. doi: 10.1200/jco.2011.38.0410.
  130. LaCasce A, Bociek RG, Matous J, et al. Brentuximab Vedotin in Combination with Bendamustine for Patients with Hodgkin Lymphoma who are Relapsed or Refractory after Frontline Therapy. Blood. 2014;124(21): Abstract 293.
  131. Connors J, Ansell S, Park SI, et al. Brentuximab Vedotin Combined with ABVD or AVD for Patients with Newly Diagnosed Advanced Stage Hodgkin Lymphoma: Long Term Outcomes. Blood. 2014;124(21): Abstract 292.
  132. Borchmann P, Eichenauer D, Pluetschow A, et al. Targeted BEACOPP variants in patients with newly diagnosed advanced stage classical Hodgkin lymphoma: interim results of a randomized phase II study. Blood. 2013;122(21): Abstract 4344.
  133. Armand P, Ansell SM, Lesokhin AM, et al. Nivolumab in Patients with Relapsed or Refractory Hodgkin Lymphoma – Preliminary Safety, Efficacy and Biomarker Results of a Phase I Study. Blood. 2014;124(21): Abstract 289.
  134. Moskowitz CH, Ribrag V, Michot J, et al. PD-1 Blockade with the Monoclonal Antibody Pembrolizumab (MK-3475) in Patients with Classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Preliminary Results from a Phase 1b Study. Blood. 2014;124(21): Abstract 290.
  135. Lesokhin AM, Ansell SM, Armand P, et al. Preliminary Results of a Phase I Study of Nivolumab (BMS-936558) in Patients with Relapsed or Refractory Lymphoid Malignancies. Blood. 2014;124(21): Abstract 291.

Ферментные препараты в онкогематологии: актуальные направления экспериментальных исследований и перспективы клинического применения

В.С. Покровский, Е.М. Трещалина

ФГБУ «Российский онкологический научный центр им. Н.Н. Блохина» РАМН, Москва, Российская Федерация


РЕФЕРАТ

Последние годы в области разработки новых противоопухолевых препаратов на основе ферментов продемонстрированы существенные достижения. Помимо L-аспарагиназы, которая применяется в онкогематологии уже более 30 лет, два фермента — L-аргининдезиминаза и ранпирназа — прошли несколько этапов клинических исследований. Для целого ряда ферментов показана противоопухолевая активность на доклиническом этапе в экспериментах in vivo: L-метионин-гамма-лиаза, L-лизин-альфа-оксидаза, биназа. В настоящем обзоре представлены ферменты, продемонстрировавшие на различных этапах исследований противоопухолевую активность, и перспективы их использования в онкогематологии.


Ключевые слова: противоопухолевые ферменты, L-аспарагиназа, L-метионин-гамма-лиаза, L-лизин-альфа-оксидаза, L-аргининдезиминаза, L-фенилаланин-аммиак-лиаза, ранпирназа.

 Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Kidd J.G. Regression of transplanted lymphomas induced in vivo by means of normal guinea pig serum. J. Exp. Med. 1953; 98: 565–82.
  2. Broome J.D. Evidence that the L-asparaginase activity of guinea pig serum is responsible for its antilymphoma effects. Nature 1961; 191: 1114–5.
  3. Трещалина Е.М. Противоопухолевая активность веществ природного происхождения. М.: Практическая медицина, 2005. [Treshchalina Ye.M. Protivoopukholevaya aktivnost veshchestv prirodnogo proiskhozhdeniya (Anti-tumor activity of substances of natural origin). M.: Prakticheskaya meditsina, 2005.]
  4. Jaccard A., Petit B., Girault S. et al. L-asparaginase-based treatment of 15 western patients with extranodal NK/T-cell lymphoma and leukemia and a review of the literature. Ann Oncol. 2009; 20(1): 110–6.
  5. Obama K., Tara M., Niina K. L-asparaginase induced complete remission in Epstein-Barr virus positive, multidrug resistant, cutaneous T-cell lymphoma. Int. J. Hematol. 1999; 69(4): 260–2.
  6. Yong W., Zheng W., Zhang Y. et al. L-аsparaginase-based regimen in the treatment of refractory midline nasal/nasal-type T/NK-cell lymphoma. Int. J. Hematol. 2003; 78(2): 163–7.
  7. Ollenschlager G., Roth E., Linkesch W. et al. Asparaginase-induced derangements of glutamine metabolism: the pathogenetic basis for some drugrelated side-effects. Eur. J. Clin Invest. 1988; 18(5): 512–6.
  8. Villa P., Corada M., Bartosek I. L-asparaginase effects on inhibition of protein synthesis and lowering of the glutamine content in cultured rat hepatocytes. Toxicol. Lett. 1986; 32(3): 235–41.
  9. Warrell R.P.Jr., Chou T.C., Gordon C. et al. Phase I evaluation of succinylated Acinetobacter glutaminase-asparaginase in adults. Cancer Res. 1980; 40(12): 4546–51.
  10. Reinert R.B., Oberle L.M., Wek A.S. et al. Role of glutamine depletion in directing tissue-specific nutrient stress responses to L-asparagine. J. Biol. Chem. 2006; 281: 31222–33.
  11. Woods J.S., Handschumacher R.E. Hepatic homeostasis of plasma L-asparagine. Am. J. Physiol. 1971; 221: 1785–90.
  12. Bendich A., Kafkewitz D., Abuchowski A., Davis F.F. Immunological effects of native and polyethylene glycol-modified asparaginases from Vibrio succinogenes and Escherichia coli in normal and tumour-bearing mice. Clin. Exp. Immunol. 1982; 48: 273–8.
  13. Distasio J.A., Salazar A.M., Nadji M., Durden D.L. Glutaminase-free asparaginase from vibrio succinogenes: an antilymphoma enzyme lacking hepatotoxicity. Int. J. Cancer. 1982; 30(3): 343–7.
  14. Capizzy R.L., Cheng Y.C. Therapy of neoplasia with asparaginase. In: Enzymes as drug. Ed. by J.S. Holcenberg, J. Roberts. NY: John Wiley and Sons, 1981: 1–24.
  15. Storti E., Quaglino D. Dysmetabolic and neurological complications in leukemic patients treated with L-asparaginase. In: Experimental and clinical effects of L-asparaginase. Ed. by E. Grundmann, H.F. Oettgen. Berlin, Heidelberg, NY: Springer Verlag, 1970: 344–9.
  16. Roberts J., Schmid F.A., Old L.J., Stockert E. A comparative study of the antitumor effectiveness of E. coli and Erwinia asparaginases. Cancer Biochem. Biophys. 1976; 1(4): 175–8.
  17. Steiner M., Attarbaschi A., Kastner U. et al. Distinct fluctuations of ammonia levels during asparaginase therapy for childhood acute leukemia. Pediatr. Blood Cancer 2007; 9(5): 640–2.
  18. Watanabe S., Miyake K., Ogawa C. et al. The ex vivo production of ammonia predicts L-asparaginase biological activity in children with acute lymphoblastic leukemia. Int. J. Hematol. 2009; 90(3): 347–52.
  19. Гладилина Ю.А., Соколов Н.Н., Красоткина Ю.В. Клонирование, экспрессия и выделение L-аспарагиназы Helicobacter pylori. Биомед. хим. 2008; 54(4): С. 482–6. [Gladilina Yu.A., Sokolov N.N., Krasotkina Yu.V. Cloning, expression, and isolation of Helicobacter pylori L-asparaginase. Biomed. khim. 2008; 54(4): S. 482–6. (In Russ.)].
  20. Cappelletty D., Chiarelli L.R., Pasquetto M.V. et al. Helicobacter pylori L-asparaginase: A promising new chemotherapeutic agent. Biochem. Biophys. Res. Commun. 2008; 377: 1222–6.
  21. Derst C., Henseling J., R hm K.H. Engineering the substrate specificity of Escherichia coli asparaginase II. Selective reduction of glutaminase activity by amino acid replacements at position 248. Protein Sci. 2000; 9: 2009–17.
  22. Avramis V.I., Panosyan E.H. Pharmacokinetic/pharmacodynamic relationships of asparaginase formulations: the past, the present and recommendations for the future. Clin Pharmacokinet. 2005; 44: 367–93.
  23. Avramis V.I., Tiwari P.N. Asparaginase (native ASNase or pegylated ASNase) in the treatment of acute lymphoblastic leukemia. Int. J. Nanomed. 2006; 1(3): 241–54.
  24. Panosyan E.H., Grigoryan R.S., Avramis I.A. et al. Deamination of glutamine is a prerequisite for optimal asparagine deamination by asparaginases in vivo (CCG-1961). Anticancer Res. 2004; 24(2C): 1121–5.
  25. Rotoli B.M., Uggeri J., Dall’Asta V. et al. Inhibition of glutamine synthetase triggers apoptosis in asparaginase-resistant cells. Cell Physiol. Biochem. 2005; 15(6): 281–92.
  26. Tardito S., Uggeri J., Bozzetto C. et al. The inhibition of glutamine synthetase sensitizes human sarcoma cells to L-asparaginase. Cancer Chemother. Pharmacol. 2007; 60(5): 751–8.
  27. Abuchowski A., Kazo G.M., Verhoest C.R. Jr. et al. Cancer therapy with chemically modified enzymes. I. Antitumor properties of polyethylene glycolasparaginase conjugates. Cancer Biochem. Biophys. 1984; 7(2): 175–86.
  28. Asselin B.L., Whitin J.C., Coppola D.J. et al. Comparative pharmacokinetic studies of three asparaginase preparations. J. Clin. Oncol. 1993; 11: 1780–6. 29. Khan A., Hill J.M. Atopic hypersensitivity to L-asparaginase: resistance to immunosupression. Int. Arch. Allergy Appl. Immunol. 1971; 40(3): 463–569.
  29. Alvarez O.A., Zimmerman G. Pegaspargase-induced pancreatitis. Med. Pediatr. Oncol. 2000; 34(3): 200–5.
  30. Кучумова А.В., Красоткина Ю.В., Хасигов П.З., Соколов Н.Н. Пегили- рование рекомбинантной аспарагиназы Erwinia carotovora полиэтиленгликолем 5000. Биомед. хим. 2007; 53(1): 107–11. [Kuchumova A.V., Krasotkina Yu.V., Khasigov P.Z., Sokolov N.N. Pegylation of recombinant Erwinia carotovora asparaginase with polyethilenglycol. 5000. Biomed. khim. 2007; 53(1): 107–11. (In Russ.)].
  31. Gaspar M.M., Perez-Soler R., Cruz M.E. Biological characterization of L-asparaginase liposomal formulations. Cancer Chemother. Pharmacol. 1996; 38(4): 373–7.
  32. Jean-Francois J., D’Urso E.M., Fortier G. Immobilization of L-asparaginase into a biocompatible poly(ethylene glycol)-albumin hydrogel: evaluation of performance in vivo. Biotechnol. Appl. Biochem. 1997; 26(Pt. 3): 203–12.
  33. Gaspar M.M., Blanco D., Cruz M.E., Alonso M.J. Formulation of L-asparaginase load poly(lactide-to-glycolide) nanoparticles: influence of polymer properties on enzyme loading, activity and in vitro release. J. Control. Release 1998; 52: 53–62.
  34. Qian G., Zhou J., Wang D., He B. The chemical modification of E. coli L-asparaginase by N, O-carboxymethyl chitosan. Artif. Cell. Blood Substit. Immobil. Biotechnol. 1996; 24: 567–77.
  35. Uren J.R., Hargis B.J., Beardsley P. Immunological and pharmacological characterization of poly-DL-alanyl-modified Erwinia carotovora L-asparaginase. Cancer Res. 1982; 42: 4068–71.
  36. Jorge J.C., Perez-Soler R., Morais J.G., Cruz M.E. Liposomal palmitoylL-asparaginase: characterization and biological activity. Cancer Chemother. Pharmacol. 1994; 34(3): 230–4.
  37. Zhang Y.Q., Zhou W.L., Shen W.D. et al. Synthesis, characterization and immunogenicity of silk fibroin-L-asparaginase bioconjugates. J. Biotechnol. 2005; 120(3): 315–26.
  38. Leal-Egana A., Scheibel T. Silk-based materials for biomedical applications. Biotechnol. Appl. Biochem. 2010; 55(3): 155–67.
  39. Spiess K., Lammel A., Scheibel T. Recombinant spider silk proteins for applications in biomaterials. Macromol. Biosci. 2010; 10(9): 998–1007.
  40. Kwon Y.M., Chung H.S., Moon C. et al. L-Asparaginase encapsulated intact erythrocytes for treatment of acute lymphoblastic leukemia. J. Control. Release 2009; 139(3): 182–9.
  41. Moola Z.B., Scawen M.D., Atkinson T., Nicholls D.J. Erwinia chrysanthemi L-asparaginase: epitope mapping and production of antigenically modified enzymes. Biochem. J. 1994; 302(Pt. 3): 921–7.
  42. Goldberg A.I., Cooney D.A., Glynn J.P. et al. The effects of immunization to L-asparaginase on antitumor and enzymatic activity. Cancer Res. 1973; 33: 256–61.
  43. Vrooman L.M., Supko J.G., Neuberg D.S. et al. Erwinia asparaginase after allergy to E. coli asparaginase in children with acute lymphoblastic leukemia. Pediatr. Blood Cancer 2010; 54(2): 199–205.
  44. Zalewska-Szewczyk B., Gach A., Wyka K. et al. The cross-reactivity of anti-asparaginase antibodies against different L-asparaginase preparations. Clin. Exp. Med. 2009; 2: 113–6.
  45. Distasio J.A., Niederman R.A. Purification and characterization of Lasparaginase with anti-lymphoma activity from Vibrio succinogenes. J. Biol. Chem. 1976; 251(22): 6929–33.
  46. Абакумова О.Ю., Подобед О.В., Борисова А.А. и др. Противоопухолевая активность L-аспарагиназы из Yersinia pseudotuberculosis. Биомед. хим. 2008; 54(6): 712–9. [Abakumova O.Yu., Podobed O.V., Borisova A.A. et al. Anti-tumor activity of Yersinia pseudotuberculosis L-asparaginase. Biomed. khim. 2008; 54(6): 712–9. (In Russ.)].
  47. Carta De-Angeli L., Pocchiari F. et al. Effect of L-asparaginase from Aspergillus terreus on ascites sarcoma in the rat. Nature (London) 1970; 225: 549–50.
  48. Peterson L.E., Ciegler A. L-asparaginase production by Erwinia aroideae. Appl. Microbiol. 1969; 18: 64–7.
  49. Pritsa A.A., Papazisis K.T., Kortsaris A.H. et al. Antitumor activity of Lasparaginase from Thermus thermophilus. Anticancer Drugs 2001; 12: 137–42.
  50. Reddy V.V.S., Jayaram H.N., Sirsi M., Ramakrishnan T. Inhibitory activity of L-asparaginase from Mycobacterium tuberculosis on Yoshida ascites sarcoma in rats. Arch. Biochem. Biophys. 1969; 132: 262–7.
  51. Rowley B., Wriston J.C. Partial purification and antilymphoma activity of Serratia marcescens L-asparaginase. Biochem. Biophys. Res. Commun. 1967; 28: 160–5.
  52. Pokrovskaya M.V., Pokrovsky V.S., Aleksandrova S.S. et al. Recombinant intracellular Rhodospirillum riubrum L-asparaginase with low L-glutaminase activity and antiproliferative effect. Biochem. (Mosc.). Suppl. Series B: Biomed. Chem. 2012; 6: 121–31.
  53. Appel I.M., Hop W.C., Pieters R. Changes in hypercoagulability by asparaginase: a randomized study between two asparaginases. Blood Coagul. Fibrinol. 2006; 17: 139–46.
  54. Duval M., Suciu S., Ferster A. et al. Comparison of Escherichia coliasparaginase with Erwinia-asparaginase in the treatment of childhood lymphoid malignancies: results of a randomized European Organisation for Research and Treatment of Cancer-Children’s Leukemia Group phase 3 trial. Blood 2002; 99(8): 2734–9.
  55. Durden D.L., Salazar A.M., Distasio J.A. Kinetic analisys of hepatotoxicity associated with antineoplastic asparaginases. Cancer Res. 1983; 43: 1602–5.
  56. Eden O.B., Shaw M.P., Lilleyman J.S., Richards S. Non-randomised study comparing toxicity of Escherichia coli and Erwinia asparaginase in children with leukaemia. Med. Pediatr. Oncol. 1990; 18(6): 497–502.
  57. Howard J.B., Carpenter F.H. L-asparaginase from Erwinia carotovora. Substrate specificity and enzymatic properties. J. Biol. Chem. 1972; 247: 1020–30.
  58. Bach S.J., Lasnitzki I. Some aspects of the role of arginine and arginase in mouse carcinoma 63. Enzymologia 1947; 12(3): 198–205.
  59. Bach S.J., Maw G.A. Creatine synthesis by tumor-bearing rats. Biochem. Biophys. Acta 1953; 11(1): 69–78.
  60. Osunkoya B.O., Adler W.H., Smith R.T. Effect of arginine deficiency on synthesis of DNA and immunoglobulin receptor of Burkitt lymphoma cells. Nature 1970; 227: 398–9.
  61. Storr J.M., Burton A.F. The effects of arginine deficiency on lymphoma cells. Br. J. Cancer 1974; 30: 50–9.
  62. Cheng P.N., Lam T.L., Lam W.M. et al. Pegylated recombinant human arginase inhibits the in vitro and in vivo proliferation of human hepatocellular carcinoma through arginine depletion. Cancer Res. 2007; 67(1): 309–17.
  63. Savoca K.V., Davis F.F., van Es T. et al. Cancer therapy with chemically modified enzymes. II. The therapeutic effectiveness of arginase, and arginase modified by the covalent attachment of polyethylene glycol, on the taper liver tumor and the L5178Y murine leukemia. Cancer Biochem. Biophys. 1984; 7(3): 261–8.
  64. Hernandez C.P., Morrow K., Lopez-Barcons L.A. et al. Pegylated arginase I: a potential therapeutic approach in T-ALL. Blood 2010; 115(25): 5214–21.
  65. Hsueh E.C., Knebel S.M., Lo W.H. et al. Deprivation of arginine by recombinant human arginase in prostate cancer cells. J. Hematol. Oncol. 2012; 5: 17. doi: 10.1186/1756-8722-5-17.
  66. Shibatani T., Kakimoto T., Chibata I. Crystallization and properties of L-arginine deiminase of Pseudomonas putida. J. Biol. Chem. 1975; 250(12): 4580–3.
  67. Takaku H., Takase M., Abe S. et al. In vivo anti-tumor activity of arginine deiminase purified from Mycoplasma arginini. Int. J. Cancer. 1992; 51(2): 244–9.
  68. Park I.S., Kang S.W., Shin Y.J. et al. Arginine deiminase: a potential inhibitor of angiogenesis and tumour growth. Br. J. Cancer 2003; 89: 907–14.
  69. Ni Y., Li Z., Sun Z. et al. Expression of arginine deiminase from Pseudomonas plecoglossicida CGMCC2039 in Escherichia coli and its anti-tumor activity. Curr. Microbiol. 2009; 58(6): 593–8.
  70. Ensor C.M., Holtsberg F.W., Bomalaski J.S., Clark M.A. Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 2002; 62: 5443–50.
  71. Gong H., Zolzer F., von Recklinghausen G. et al. Arginine deiminase inhibits proliferation of human leukemia cells more potently than asparaginase by inducing cell cycle arrest and apoptosis. Leukemia 2000; 14(5): 826–9.
  72. Noh E.J., Kang S.W., Shin Y.J. et al. Arginine deiminase enhances dexamethasone-induced cytotoxicity in human T-lymphoblastic leukemia CCRF-CEM cells. Int. J. Cancer 2004: 112: 502–8.
  73. Ascierto P.A., Scala S., Castello G. et al. Pegylated arginine deiminase treatment of patients with metastatic melanoma: results from phase I and II studies. J. Clin. Oncol. 2005; 23: 7660–8.
  74. Curley S.A., Bomalaski J.S., Ensor C.M. et al. Regression of hepatocellular cancer in a patient treated with arginine deiminase. Hepatogastroenterology 2003; 50(53): 1214–6.
  75. Glazer E.S., Piccirillo M., Albino V. et al. Phase II study of pegylated arginine deiminase for nonresectable and metastatic hepatocellular carcinoma. J. Clin. Oncol. 2010; 28(13): 2220–6.
  76. Izzo F., Marra P., Beneduce G. et al. Pegylated arginine deiminase treatment of patients with unresectable hepatocellular carcinoma: results from phase I/II studies. J. Clin. Oncol. 2004; 22: 1815–22.
  77. Glazer E.S., Piccirillo M., Albino V. et al. Phase II study of pegylated arginine deiminase for nonresectable and metastatic hepatocellular carcinoma. J. Clin. Oncol. 2010; 28(13): 2220–6.
  78. Ott P.A., Carvajal R.D., Pandit-Taskar N. et al. Phase I/II study of pegylated arginine deiminase (ADI-PEG20) in patients with advanced melanoma. Invest. New Drugs 2013; 31(2): 425–34.
  79. Delage B., Luong P., Maharaj L. et al. Promoter methylation of argininosuccinate synthetase-1 sensitises lymphomas to arginine deiminase treatment, autophagy and caspase-dependent apoptosis. Cell Death Dis. 2012; 3: e342.
  80. Wu L., Li L., Meng S. et al. Expression of argininosuccinate synthetase in patients with hepatocellular carcinoma. J. Gastroenterol. Hepatol. 2013; 28(2): 365–8.
  81. Szlosarek P.W., Luong P., Phillips M.M. et al. Metabolic response to pegylated arginine deiminase in mesothelioma with promoter methylation of argininosuccinate synthetase. J. Clin. Oncol. 2013; 31(7): e111–3.
  82. Feun L.G., Marini A., Walker G. et al. Negative argininosuccinate synthetase expression in melanoma tumours may predict clinical benefit from arginine-depleting therapy with pegylated arginine deiminase. Br. J. Cancer 2012; 106(9): 1481–5.
  83. Kelly M.P., Jungbluth A.A., Wu B.W. et al. Arginine deiminase PEG20 inhibits growth of small cell lung cancers lacking expression of argininosuccinate synthetase. Br. J. Cancer 2012; 106(2): 324–32.
  84. Manca A., Sini M.C., Izzo F. et al. Induction of arginosuccinate synthetase (ASS) expression affects the antiproliferative activity of arginine deiminase (ADI) in melanoma cells. Oncol. Rep. 2011; 25(6): 1495–502.
  85. Bowles T.L., Kim R., Galante J. et al. Pancreatic cancer cell lines deficient in argininosuccinate synthetase are sensitive to arginine deprivation by arginine deiminase. Int. J. Cancer 2008; 123: 1950–5.
  86. Kim H.J., Kim J.H., Yu Y.S. et al. Anti-tumor activity of arginine deiminase via arginine deprivation in retinoblastoma. Oncol. Rep. 2007; 18: 1373–7.
  87. Kim R.H., Coates J.M., Bowles T.L. et al. Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res. 2009; 69: 700–8.
  88. Sigimura K., Ohno T., Kusuyama T., Azuma I. High sensitivity of human melanoma cell lines to the growth inhibitory activity of mycoplasmal arginine deiminase in vitro. Melanoma Res. 1992; 2: 191–6.
  89. Szlosarek P.W., Klabatsa A., Pallaska A. et al. In vivo loss of expression of argininosuccinate synthetase in malignant pleural mesothelioma is a biomarker for susceptibility to arginine depletion. Clin. Cancer Res. 2006; 12: 7126–31.
  90. Yoon C.Y., Shim Y.J., Kim E.H. et al. Renal cell carcinoma does not express argininosuccinate synthetase and is highly sensitive to arginine deprivation via arginine deiminase. Int. J. Cancer 2008; 120: 897–905.
  91. Tsai W.B., Aiba I., Lee S.Y. et al. Resistance to arginine deiminase treatment in melanoma cells is associated with induced argininosuccinate synthetase expression involving c-Myc/HIF-1alpha/Sp4. Mol. Cancer Ther. 2009; 8(12): 3223–33.
  92. Ni Y., Liu Y., Schwaneberg U. et al. Rapid evolution of arginine deiminase for improved anti-tumor activity. Appl. Microbiol. Biotechnol. 2011; 90(1): 193–201.
  93. Holtsberg F.W., Ensor C.M., Steiner M.R. et al. Poly(ethylene glycol) (PEG) conjugated arginine deiminase: effects of PEG formulations on its pharmacological properties. J. Control. Release 2002; 80: 259–71.
  94. Kreis W., Hession C. Biological effects of enzymatic deprivation of Lmethionine in cell culture and an experimental tumor. Cancer Res. 1973; 33(8): 1866–9.
  95. Занин В.А., Лукина В.И., Березов Т.Т. Выделение и некоторые фи- зико-химические и каталитические свойства L-лизин-альфа-оксидазы из Pseudomonas putida. Вопр. мед. хим. 1989; 4: 84–9. [Zanin V.A., Lukina V.I., Berezov T.T. Isolation and some physicochemical and catalytical properties of Pseudomonas putida L-lysine alpha-oxidase. Vopr. med. khim. 1989; 4: 84–9. (In Russ.)].
  96. Манухов И.В., Мамаева Д.В., Морозова Е.А. и др. L-Метионин-гамма- лиаза Citrobacter freundii: клонирование гена и кинетические параметры фермента. Биохим. 2006; 74(4): 454–63. [Manukhov I.V., Mamayeva D.V., Morozova Ye.A. et al. Citrobacter freundii L-methionine gamma-lyase: gene cloning and clinical parameters of enzyme. Biokhim. 2006; 74(4): 454–63. (In Russ.)].
  97. Ito S., Nakamura T., Eguchi Y. Purification and characterization of methioninase from Pseudomonas putida. J. Biochem. 1976; 79(6): 1263–72.
  98. Lockwood B.C., Coombs G.H. Purification and characterization of methionine gamma-lyase from Trichomonas vaginalis. Biochem. J. 1991; 279: 675–82.
  99. Sato D., Yamagata W., Kamei K. et al. Expression, purification and crystallization of L-methionine gamma-lyase 2 from Entamoeba histolytica. Acta Crystallogr. 2006; 62(10): 1034–6.
  100. Tanaka H., Esaki N., Yamamoto T., Soda K. Purification and properties of methioninase from Pseudomonas ovalis. FEBS Lett. 1976; 66(2): 307–11.
  101. El-Sayed A.S. Purification and characterization of a new L-methioninase from solid cultures of Aspergillus flavipes. J. Microbiol. 2011; 49(1): 130–40.
  102. Пехов А.А., Жукова О.С., Занин В.А., Березов Т.Т. Цитостатический эффект L-метионин-g-лиазы на раковые клетки в культуре. Бюл. эксп. биол. мед. 1983; 5: 87–9. [Pekhov A.A., Zhukova O.S., Zanin V.A., Berezov T.T. Cytostatic effect of L-methionine g-lyase on cultured cancer cells. Byul. eksp. biol. med. 1983; 5: 87–9. (In Russ.)].
  103. Hu J., Cheung N.K. Methionine depletion with recombinant methioninase: in vitro and in vivo efficacy against neuroblastoma and its synergism with chemotherapeutic drugs. Int. J. Cancer 2009; 124(7): 1700–6.
  104. Kokkinakis D.M., Schold S.C.Jr., Hori H., Nobori T. Effect of long-term depletion of plasma methionine on the growth and survival of human brain tumor xenografts in athymic mice. Nutr. Cancer 1997; 29(3): 195–204.
  105. Tan Y., Sun X., Xu M. et al. Efficacy of recombinant methioninase in combination with cisplatin on human colon tumors in nude mice. Clin. Cancer Res. 1999; 5(8): 2157–63.
  106. Tan Y., Xu M., Guo H. et al. Anticancer efficacy of methioninase in vivo. Anticancer Res. 1996; 16(6C): 3931–6.
  107. Yoshioka T., Wada T., Uchida N. et al. Anticancer efficacy in vivo and in vitro, synergy with 5-fluorouracil, and safety of recombinant methioninase. Cancer Res. 1998; 58(12): 2583–7.
  108. Hori H., Takabayashi K., Orvis L. et al Gene cloning and characterization of Pseudomonas putida L-methionine-alpha-deamino-gamma-mercaptomethane-lyase. Cancer Res. 1996; 56(9): 2116–22.
  109. El-Sayed A.S., Shouman S.A., Nassrat H.M. Pharmacokinetics, immunogenicity and anticancer efficiency of Aspergillus flavipes L-methioninase. Enzyme Microb. Technol. 2012; 51(4): 200–10.
  110. Tan Y., Zavala J.Sr., Xu M. et al. Serum methionine depletion without side effects by methioninase in metastatic breast cancer patients. Anticancer Res. 1996; 16(6C): 3937–42.
  111. Sun X., Yang Z., Li S. et al. In vivo efficacy of recombinant methioninase is enhanced by the combination of polyethylene glycol conjugation and pyridoxal 5¢-phosphate supplementation. Cancer Res. 2003; 63(23): 8377–83.
  112. Xin L., Cao J., Cheng H. et al. Stealth cationic liposomes modified with anti-CAGE single-chain fragment variable deliver recombinant methioninase for gastric carcinoma therapy. J. Nanosci. Nanotechnol. 2013; 13(1): 178–83.
  113. Смирнова И.П., Хадуев С.Х. L-лизин-альфа-оксидазная активность некоторых видов Trichoderma. Микробиология 1984; 53: 163–5. [Smirnova I.P., Khaduyev S.Kh. L-lysine alpha-oxidase activity of some Trichoderma spp. Mikrobiologiya 1984; 53: 163–5. (In Russ.)].
  114. Kusakabe H., Kodama K., Kuninaka A. et al. A new antitumor enzyme, L-lysine alpha-oxidase from Trichoderma viride. Purification and enzymological properties. J. Biol. Chem. 1980; 255(3): 976–81.
  115. Жукова О.С., Хадуев С.Х., Добрынин Я.В. и др. Влияние L-лизин-a- оксидазы на кинетику клеточного цикла культивируемых клеток лимфомы Беркитта. Экспер. онкол. 1985; 7(6): 42–4. [Zhukova O.S., Khaduyev S.Kh., Dobrynin Ya.V. et al. Influence of L-lysine a-oxidase on kinetics of cell cycle of Burkitt’s lymphoma cultured cells. Eksper. onkol. 1985; 7(6): 42–4. (In Russ.)].
  116. Гогичаева Н.В., Лукашева Е.В., Гаврилова Е.М. и др. Получение конъюгатов L-лизин-a-оксидазы с антителами. Вопр. мед. хим. 2000; 46(4): 410–8. [Gogichayeva N.V., Lukasheva Ye.V., Gavrilova Ye.M. et al. Synthesis of conjugates of L-lysine a-oxidase with antibodies. Vopr. med. khim. 2000; 46(4): 410–8. (In Russ.)].
  117. Лукашева Е.В., Березов Т.Т. L-лизин-a-оксидаза: физико-химиче- ские и биологические свойства. Биохимия 2002; 67(10): 1394–402. [Lukasheva Ye.V., Berezov T.T. L-lysine a-oxidase: physicochemical and biological properties. Biokhimiya 2002; 67(10): 1394–402. (In Russ.)].
  118. Sarkissian C.N., Shao Z., Blain F. et al. A different approach to treatment of phenylketonuria: phenylalanine degradation with recombinant phenylalanine ammonia lyase. Proc. Natl. Acad. Sci. U S A 1999; 96(5): 2339–44.
  119. Calabrese J.C., Jordan D.B., Boodhoo A. et al. Crystal structure of phenylalanine ammonia-lyase: multiple helix dipoles implicated in catalysis. Biochemistry 2004; 43: 11403–16.
  120. Ritter H., Schulz G.E. Structural basis for the entrance into the phenylpropanoid metabolism catalyzed by phenylalanine ammonia-lyase. Plant Cell 2004; 16: 3426–36.
  121. Bourget L., Chang T.M. Artificial cell-microencapsulated phenylalanine ammonia-lyase. Applied Biochem. Biotechnol. 1984; 10: 57–9.
  122. Sarkissian C.N., Gamez A. Phenylalanine ammonia lyase, enzyme substitution therapy for phenylketonuria, where are we now? Mol. Gen. Metab. 2005; 86(Suppl. 1): S22–6.
  123. Abell C.W., Hodgins D.S., Stith W.J. An in vivo evaluation of the chemotherapeutic potency of phenylalanine ammonia-lyase. Cancer Res. 1973; 33(10): 2529–32.
  124. Stith W.J., Hodgins D.S., Abell C.W. Effects of phenylalanine ammonialyase and phenylalanine deprivation on murine leukemic lymphoblasts in vitro. Cancer Res. 1973; 33(5): 966–71.
  125. Ambrus C.M., Anthone S., Horvath C. et al. Extracorporeal enzyme reactors for depletion of phenylalanine in phenylketonuria. Ann. Intern. Med. 1987; 106: 531–7.
  126. Ledoux L. Action of ribonuclease on two solid tumours in vivo. Nature 1955; 176(4470): 36–7.
  127. Mitkevich V.A., Tchurikov N.A., Zelenikhin P.V. et al. Binase cleaves cellular noncoding RNAs and affects coding mRNAs. FEBS J. 2010; 277(1): 186–96.
  128. Darzynkiewicz Z., Carter S.P., Mikulski S.M. et al. Cytostatic and cytotoxic effects of Pannon (P-30 Protein), a novel anticancer agent. Cell Tissue Kinet. 1988; 21(3): 169–82.
  129. Ardelt W., Mikulski S.M., Shogen K. Amino acid sequence of an antitumor protein from Rana pipiens oocytes and early embryos. Homology to pancreatic ribonucleases. Biol. Chem. 1991; 266(1): 245–51.
  130. Wu Y., Mikulski S.M., Ardelt W. et al. A cytotoxic ribonuclease. Study of the mechanism of onconase cytotoxicity. J. Biol. Chem. 1993; 268(14): 10686–93.
  131. Juan G., Ardelt B., Li X. et al. G1 arrest of U937 cells by onconase is associated with suppression of cyclin D3 expression, induction of p16INK4A, p21WAF1/CIP1 and p27KIP and decreased pRb phosphorylation. Leukemia 1998; 12(8): 1241–8.
  132. Deptala A., Halicka H.D., Ardelt B. et al. Potentiation of tumor necrosis factor induced apoptosis by onconase. Int. J. Oncol. 1998; 13(1): 11–6.
  133. Lee I., Kalota A., Gewirtz A.M., Shogen K. Antitumor efficacy of the cytotoxic RNase, ranpirnase, on A549 human lung cancer xenografts of nude mice. Anticancer Res. 2007; 27(1A): 299–307.
  134. Lee I., Lee Y.H., Mikulski S.M., Shogen K. Effect of onconase +/- tamoxifen on ASPC-1 human pancreatic tumors in nude mice. Adv. Exp. Med. Biol. 2003; 530: 187–96.
  135. Воробьев И.И., Пономаренко Н.А., Дурова О.М. и др. Структурно- функциональное исследование рекомбинантных форм онконазы. Био- орган. хим. 2001; 27(4): 257–64. [Vorobyev I.I., Ponomarenko N.A., Durova O.M. et al. Structural-functional evaluation of Onconase recombinant forms. Bioorgan. khim. 2001; 27(4): 257–64. (In Russ.)].
  136. Notomista E., Cafaro V., Fusiello R. et al. Effective expression and purification of recombinant onconase, an antitumor protein. FEBS Lett. 1999; 463(3): 211–5.
  137. Ita M., Halicka H.D., Tanaka T. et al. Remarkable enhancement of cytotoxicity of onconase and cepharanthine when used in combination on various tumor cell lines. Cancer Biol Ther. 2008; 7(7): 1104–8.
  138. Costanzi J., Sidransky D., Navon A. et al. Ribonucleases as a novel pro-apoptotic anticancer strategy: review of the preclinical and clinical data for ranpirnase. Cancer Invest. 2005; 23(7): 643–50.
  139. Mikulski S.M., Costanzi J.J., Vogelzang N.J. et al. Phase II trial of a single weekly intravenous dose of ranpirnase in patients with unresectable malignant mesothelioma. J. Clin. Oncol. 2002; 20(1): 274–81.
  140. Porta C., Paglino C., Mutti L. Ranpirnase and its potential for the treatment of unresectable malignant mesothelioma. Biologics 2008; 2(4): 601–9.
  141. Chang C.H., Sapra P., Vanama S.S. et al. Effective therapy of human lymphoma xenografts with a novel recombinant ribonuclease/anti-CD74 humanized IgG4 antibody immunotoxin. Blood 2005; 106(13): 4308–14.
  142. Calabrese J.C., Jordan D.B., Boodhoo A. et al. Crystal structure of phenylalanine ammonia-lyase: multiple helix dipoles implicated in catalysis. Biochemistry 2004; 43: 11403–16.
  143. Ardelt B., Ardelt W., Pozarowski P. et al. Cytostatic and cytotoxic properties of Amphinase: a novel cytotoxic ribonuclease from Rana pipiens oocytes. Cell Cycle 2007; 24: 3097–102.
  144. Ильинская О.Н., Зеленихин П.В., Колпаков А.И. и др. Избирательная цитотоксичность биназы в отношении фибробластов, экспрессирующих онкогены ras и AML/ETO. Учен. зап. Казан. ун-та. Серия «Естественные науки» 2008; 150(4): 268–73. [Ilinskaya O.N., Zelenikhin P.V., Kolpakov A.I. et al. Selective binase cytotoxicity against ras- and AML/ETO-oncogene-expressing fibroblasts. Uchen. zap. Kazan. un-ta. Seriya «Estestvennye nauki» 2008; 150(4): 268–73. (In Russ.)].
  145. Mitkevich V.A., Kretova O.V., Petrushanko I.Y. et al. Ribonuclease binase apoptotic signature in leukemic Kasumi-1 cells. Biochemie 2013; 95(6): 1344–9.
  146. Mitkevich V.A., Petrushanko I.Y., Spirin P.V. et al. Sensitivity of acute myeloid leukemia Kasumi-1 cells to binase toxic action depends on the expression of KIT and АML1-ETO oncogenes. Cell Cycle 2011; 10(23): 4090–7.

Роль селективности ингибиторов тирозинкиназ в развитии побочных эффектов при терапии хронического миелолейкоза

А.А. Зейфман1,2, Е.Ю. Челышева3, А.Г. Туркина3, Г.Г. Чилов1,2

1 ФГБУ «Институт органической химии им. Н.Д. Зелинского РАН», Москва, Российская Федерация

2 ООО «Фьюжн Фарма», Москва, Российская Федерация

3 ФГБУ «Гематологический научный центр» МЗ РФ, Москва, Российская Федерация


РЕФЕРАТ

В обзоре рассмотрен вопрос о связи селективности ингибиторов Bcr-Abl-киназ со спектром нежелательных побочных эффектов у больных хроническим миелолейкозом при проведении терапии. Суммированы данные по структуре и естественным биохимическим функциям наиболее хорошо изученных побочных мишеней ингибиторов Bcr-Abl-киназ: BRAF, FMS, EGFR, PDGFR, PYK2, TIE2, VEGFR1/2/3, а также оценена возможная связь их нецелевого ингибирования и нежелательных побочных эффектов ингибиторов тирозинкиназ.


Ключевые слова: хронический миелолейкоз, ингибиторы тирозинкиназ, селективность, иматиниб, нилотиниб, дазатиниб, понатиниб, PF-114, BRAF, FMS, EGFR, PDGFR, PYK2, TIE2, VEGFR1/2/3.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Chartier M., Chenard T., Barker J. et al. Kinome Render: a stand-alone and web-accessible tool to annotate the human protein kinome tree. Peer J. 2013; 1: e126.
  2. Soverini S., Hochhaus A., Nicolini F.E. et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011; 118(5): 1208–15.
  3. Куцев С.И., Вельченко М.В. Значение анализа мутаций гена BCR-ABL в оптимизации таргетной терапии хронического миелолейкоза. Клин. онкогематол. 2008; 1(3): 190–9. [Kutsev S.I., Velchenko M.V. Significance of analysis of BCR-ABL gene mutations in optimization of target therapy for chronic myeloid leukemia. Klin. onkogematol. 2008; 1(3): 190–9. (In Russ.)].
  4. Челышева Е.Ю., Шухов О.А., Лазарева О.В. и др. Мутации гена BCR-ABL при хроническом миелолейкозе. Клин. онкогематол. 2012; 5(1): 13–21. [Chelysheva Ye.Yu., Shukhov O.A., Lazareva O.V. et al. BCR-ABL gene mutations in chronic myeloid leukemia. Klin. onkogematol. 2012; 5(1): 13–21. (In Russ.)].
  5. Lombardo L.J., Lee F.Y., Chen P. et al. Discovery of N-(2-chloro-6-methylphenyl)-2-(6-(4-(2-hydroxyethyl)-piperazin-1-yl)-2-methylpyrimidin-4-ylamino) thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J. Med. Chem 2004; 47(27): 6658–61.
  6. Weisberg E., Manley P.W., Breitenstein W. et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 2005; 7(2): 129–41.
  7. Golas J.M., Arndt K., Etienne C. et al. SKI-606, a 4-anilino-3-quinoline carbonitrile dual inhibitor of Src and Abl kinases, is a potent antiproliferative agent against chronic myelogenous leukemia cells in culture and causes regression of K562 xenografts in nude mice. Cancer Res 2003; 63(2): 375–81.
  8. O’Hare T., Shakespeare W.C., Zhu X. et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 2009; 16(5): 401–12.
  9. Mian A.A., Badura S., Rafiei A. et al. PF-114, a novel selective pan-Bcr/ Abl inhibitor for Philadelphia chromosome positive (Ph+) leukemia, effectively targets T315I and the other resistance mutants. European Hematologic Association, Stockholm, Sweden, June 13–16, 2013: S1177.
  10. Anastassiadis T., Deacon S.W., Devarajan K. et al. Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat. Biotechnol. 2011; 29(11): 1039–45.
  11. Saglio G., Kim D.W., Issaragrisil S. et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N. Engl. J. Med. 2010; 362(24): 2251–9.
  12. Kantarjian H., Shah N.P., Hochhaus A. et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 2010; 362(24): 2260–70.
  13. Davis M.I., Hunt J.P., Herrgard S. et al. Comprehensive analysis of kinase inhibitor selectivity. Nat. Biotechnol. 2011; 29(11): 1046–51.
  14. Martins D.H., Wagner S.C., Dos Santos T.V. et al. Monitoring imatinib plasma concentrations in chronic myeloid leukemia. Rev. Bras. Hematol. Hemother. 2011; 33(4): 302–6.
  15. Demetri G.D., Lo Russo P., MacPherson I.R. et al. Phase I dose-escalation and pharmacokinetic study of dasatinib in patients with advanced solid tumors. Clin. Cancer Res. 2009; 15(19): 6232–40.
  16. Manley P.W., Drueckes P., Fendrich G. et al. Extended kinase profile and properties of the protein kinase inhibitor nilotinib. Biochem. Biophys. Acta 2010; 1804(3): 445–53.
  17. Bradeen H.A., Eide C.A., O’Hare T. et al. Comparison of imatinib mesylate, dasatinib (BMS-354825), and nilotinib (AMN107) in an N-ethyl-N-nitrosourea (ENU)-based mutagenesis screen: high efficacy of drug combinations. Blood 2006; 108(7): 2332–8.
  18. Remsing Rix L.L., Rix U., Colinge J. et al. Global target profile of the kinase inhibitor bosutinib in primary chronic myeloid leukemia cells. Leukemia 2009; 23(3): 477–85.
  19. Cortes J.E., Kantarjian H.M., Brummendorf T.H. et al. Safety and efficacy of bosutinib (SKI-606) in chronic phase Philadelphia chromosome-positive chronic myeloid leukemia patients with resistance or intolerance to imatinib. Blood 2011; 118(17): 4567–76.
  20. Schrock A.B., Gozgit J.M., Rivera V. The pan-BCR-ABL inhibitor ponatinib inhibits viability of gatekeeper mutant BCR-ABLT315I cells with greater potency than a nilotinib/MEK inhibitor combination. Clin. Cancer Res. 2012; 18: Abstract B15.
  21. Sonnichsen D., Dorer D.J., Cortes J. et al. Analysis of the potential effect of ponatinib on the QTc interval in patients with refractory hematological malignancies. Cancer Chemother. Pharmacol. 2013; 71(6): 1599–607.
  22. Chan W.W., Wise S.C., Kaufman M.D. et al. Conformational control inhibition of the BCR-ABL1 tyrosine kinase, including the gatekeeper T315I mutant, by the switch-control inhibitor DCC-2036. Cancer Cell 2011; 19(4): 556–68.
  23. Fiskus W., Smith C.C., Smith J. et al. Activity of Allosteric, Switch-Pocket, ABL/FLT3 Kinase Inhibitor DCC2036 Against Cultured and Primary AML Progenitors with FLT-ITD or FLT3 Kinase Domain Mutations. 53rd ASH Annual Meeting and Exposition, 2011.
  24. Fancelli D., Moll J., Varasi M. et al. 1,4,5,6-tetrahydropyrrolo[3,4-c] pyrazoles: identification of a potent Aurora kinase inhibitor with a favorable antitumor kinase inhibition profile. J. Med. Chem. 2006; 49(24): 7247–51.
  25. Steeghs N., Eskens F.A., Gelderblom H. et al. Phase I pharmacokinetic and pharmacodynamic study of the aurora kinase inhibitor danusertib in patients with advanced or metastatic solid tumors. J. Clin. Oncol. 2009; 27(30): 5094–101.
  26. Ruthardt M. PF-114, a novel selective PAN BCR/ABL inhibitor for Philadelphia chromosome-positive (Ph+) leukemia, effectively targets T315I and other resistance mutant. 15th International Conference on Chronic Myeloid Leukemia: Biology and Therapy, 2013.
  27. Uniprot for BRAF. Available from: http://www.uniprot.org/uniprot/P15056.
  28. Davies H., Bignell G.R., Cox C. et al. Mutations of the BRAF gene in human cancer. Nature 2002; 417(6892): 949–54.
  29. Pratilas C.A., Xing F., Solit D.B. Targeting oncogenic BRAF in human cancer. Curr. Top Microbiol. Immunol. 2012; 355: 83–98.
  30. Roskoski R.Jr. RAF protein-serine/threonine kinases: structure and regulation. Biochem. Biophys. Res. Commun. 2010; 399(3): 313–7.
  31. Chang F., Steelman L.S., Lee J.T. et al. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia 2003; 17(7): 1263–93.
  32. Wellbrock C., Karasarides M., Marais R. The RAF proteins take centre stage. Nat. Rev. Mol. Cell Biol. 2004; 5(11): 875–85.
  33. Freeman A.K., Ritt D.A., Morrison D.K. Effects of Raf dimerization and its inhibition on normal and disease-associated Raf signaling. Mol. Cell 2013; 49(4): 751–8.
  34. Sabbatino F., Wang Y., Wang X. et al. Emerging BRAF inhibitors for melanoma. Exp. Opin. Emerg. Drugs 2013; 18(4): 431–43.
  35. Boussemart L., Routier E., Mateus C. et al. Prospective study of cutaneous side-effects associated with the BRAF inhibitor vemurafenib: a study of 42 patients. Ann. Oncol. 2013; 24(6): 1691–7.
  36. Huang V., Hepper D., Anadkat M. et al. Cutaneous toxic effects associated with vemurafenib and inhibition of the BRAF pathway. Arch. Dermatol. 2012; 148(5): 628–33.
  37. Hey F., Pritchard C. A new mode of RAF autoregulation: a further complication in the inhibitor paradox. Cancer Cell 2013; 23(5): 561–3.
  38. FDA, Risk Assessment And Risk Mitigation Review(S) for Iclusig (ponatinib), 2012.
  39. Drucker A.M., Wu S., Busam K.J. et al. Rash with the multitargeted kinase inhibitors nilotinib and dasatinib: meta-analysis and clinical characterization. Eur. J. Haematol. 2013; 90(2): 142–50.
  40. Uniprot for c-FMS. Available from: http://www.uniprot.org/uniprot/P07333.
  41. Bourette R.P., Rohrschneider L.R. Early events in M-CSF receptor signaling. Growth Factors 2000; 17(3): 155–66.
  42. Zaidi M. Skeletal remodeling in health and disease. Nat. Med. 2007; 13(7): 791–801.
  43. Kimura K., Kitaura H., Fujii T. et al. An anti-c-Fms antibody inhibits osteoclastogenesis in a mouse periodontitis model. Oral Dis. 2013 [Epub ahead of print].
  44. Nurmio M., Joki H., Kallio J. et al. Receptor tyrosine kinase inhibition causes simultaneous bone loss and excess bone formation within growing bone in rats. Toxicol. Appl. Pharmacol. 2011; 254(3): 267–79.
  45. Hamilton J.A. Colony-stimulating factors in inflammation and autoimmunity. Nat. Rev. Immunol. 2008; 8(7): 533–44.
  46. Paniagua R.T., Chang A., Mariano M.M. et al. c-Fms-mediated differentiation and priming of monocyte lineage cells play a central role in autoimmune arthritis. Arthritis Res. Ther. 2010; 12(1): R32.
  47. Lim A.K., Ma F.Y., Nikolic-Paterson D.J. et al. Antibody blockade of c-fms suppresses the progression of inflammation and injury in early diabetic nephropathy in obese db/db mice. Diabetologia 2009; 52(8): 1669–79.
  48. Baay M., Brouwer A., Pauwels P. et al. Tumor Cells and Tumor-Associated Macrophages: Secreted Proteins as Potential Targets for Therapy. Clin. Dev. Immunol. 2011; 2011: 12.
  49. Ovadia S., Insogna K., Yao G.Q. The cell-surface isoform of colony stimulating factor 1 (CSF1) restores but does not completely normalize fecundity in CSF1-deficient mice. Biol. Reprod. 2006; 74(2): 331–6.
  50. Salmassi A., Mettler L., Jonat W. et al. Circulating level of macrophage colony-stimulating factor can be predictive for human in vitro fertilization outcome. F rtil. Steril. 2010; 93(1): 116–23.
  51. Narayanan K.R., Bansal D., Walia R. et al. Growth failure in children with chronic myeloid leukemia receiving imatinib is due to disruption of GH/IGF-1 axis. Pediatr. Blood Cancer 2013; 60(7): 1148–53.
  52. Iclusig (ponatinib) prescribing information. 53. Bosulif (Bosutinib) prescribing information.
  53. Uniprot for EGFR. Available from: http://www.uniprot.org/uniprot/P00533.
  54. Hynes N.E., Lane H.A. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat. Rev. Cancer 2005; 5(5): 341–54.
  55. Reuter C.W., Morgan M.A., Eckardt A. Targeting EGF-receptor-signalling in squamous cell carcinomas of the head and neck. Br. J. Cancer 2007; 96(3): 408–16.
  56. Lenz H.J. Anti-EGFR mechanism of action: antitumor effect and underlying cause of adverse events. Oncology (Williston Park) 2006; 20(5 Suppl. 2): 5–13.
  57. Perez-Soler R. Can rash associated with HER1/EGFR inhibition be used as a marker of treatment outcome? Oncology (Williston Park) 2003; 17(11 Suppl. 12): 23–8.
  58. Murillas R., Larcher F., Conti C.J. et al. Expression of a dominant negative mutant of epidermal growth factor receptor in the epidermis of transgenic mice elicits striking alterations in hair follicle development and skin structure. EMBO J. 1995; 14(21): 5216–23.
  59. Yano S., Kondo K., Yamaguchi M. et al. Distribution and function of EGFR in human tissue and the effect of EGFR tyrosine kinase inhibition. Anticancer Res. 2003; 23(5A): 3639–50.
  60. Lee Y., Shim H.S., Park M.S. et al. High EGFR gene copy number and skin rash as predictive markers for EGFR tyrosine kinase inhibitors in patients with advanced squamous cell lung carcinoma. Clin. Cancer Res. 2012; 18(6): 1760–8.
  61. Perez-Soler R., Delord J.P., Halpern A. et al. HER1/EGFR inhibitorassociated rash: future directions for management and investigation outcomes from the HER1/EGFR inhibitor rash management forum. Oncologist 2005; 10(5): 345–56.
  62. Takeda K., Hida T., Sato T. et al. Randomized phase III trial of platinumdoublet chemotherapy followed by gefitinib compared with continued platinumdoublet chemotherapy in Japanese patients with advanced non-small-cell lung cancer: results of a west Japan thoracic oncology group trial (WJTOG0203). J. Clin. Oncol. 2010; 28(5): 753–60.
  63. Erlotinib(Iressa) prescribing information.
  64. Sprycel (dasatinib) prescribing information.
  65. Uniprot for PDGFRA. Available from: http://www.uniprot.org/uniprot/ P16234.
  66. Uniprot for PDGFRB. Available from: http://www.uniprot.org/uniprot/ P09619.
  67. Hoch R.V., Soriano P. Roles of PDGF in animal development. Development 2003; 130(20): 4769–84.
  68. Shim A.H., Liu H., Focia P.J. et al. Structures of a platelet-derived growth factor/propeptide complex and a platelet-derived growth factor/receptor complex. Proc. Natl. Acad. Sci. U S A 2010; 107(25): 11307–12.
  69. Andrae J., Gallini R., Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008; 22(10): 1276–312.
  70. Eckhardt S.G., Rizzo J., Sweeney K.R. et al. Phase I and pharmacologic study of the tyrosine kinase inhibitor SU101 in patients with advanced solid tumors. J. Clin. Oncol. 1999; 17(4): 1095–104.
  71. Kuenen B.C., Giaccone G., Ruijter R. et al. Dose-finding study of the multitargeted tyrosine kinase inhibitor SU6668 in patients with advanced malignancies. Clin. Cancer Res. 2005; 11(17): 6240–6.
  72. Jayson G.C., Parker G.J., Mullamitha S. et al. Blockade of platelet-derived growth factor receptor-beta by CDP860, a humanized, PEGylated di-Fab’, leads to fluid accumulation and is associated with increased tumor vascularized volume. J. Clin. Oncol. 2005; 23(5): 973–81.
  73. Kelly K., Swords R., Mahalingam D. et al. Serosal inflammation (pleural and pericardial effusions) related to tyrosine kinase inhibitors. Target Oncol. 2009; 4(2): 99–105.
  74. Berman E., Nicolaides M., Maki R.G. et al. Altered bone and mineral metabolism in patients receiving imatinib mesylate. N. Engl. J. Med. 2006; 354(19): 2006–13.
  75. O’Sullivan S., Naot D., Callon K. et al. Imatinib promotes osteoblast differentiation by inhibiting PDGFR signaling and inhibits osteoclastogenesis by both direct and stromal cell-dependent mechanisms. J. Bone Miner. Res. 2007; 22(11): 1679–89.
  76. Tasigna (nilotinib) prescribing information.
  77. Uniprot for PYK2. Available from: http://www.uniprot.org/uniprot/Q14289.
  78. Lipinski C.A., Loftus J.C. Targeting Pyk2 for therapeutic intervention. Exp. Opin. Ther. Targets 2010; 14(1): 95–108.
  79. Raja S., Avraham S., Avraham H. Tyrosine phosphorylation of the novel protein-tyrosine kinase RAFTK during an early phase of platelet activation by an integrin glycoprotein IIb-IIIa-independent mechanism. J. Biol. Chem. 1997; 272(16): 10941–7.
  80. Ohmori T., Yatomi Y., Asazuma N. et al. Involvement of proline-rich tyrosine kinase 2 in platelet activation: tyrosine phosphorylation mostly dependent on alphaIIb beta3 integrin and protein kinase C, translocation to the cytoskeleton and association with Shc through Grb2. Biochem. J. 2000; 347(Pt. 2): 561–9.
  81. Canobbio I., Cipolla L., Consonni A. et al. Impaired thrombin-induced platelet activation and thrombus formation in mice lacking the Ca(2+)-dependent tyrosine kinase Pyk2. Blood 2013; 121(4): 648–57.
  82. Okigaki M., Davis C., Falasca M. et al. Pyk2 regulates multiple signaling events crucial for macrophage morphology and migration. Proc. Natl. Acad. Sci. U S A 2003; 100(19): 10740–5.
  83. Kamen L.A., Schlessinger J., Lowell C.A. Pyk2 is required for neutrophil degranulation and host defense responses to bacterial infection. J. Immunol. 2011; 186(3): 1656–65.
  84. Gil-Henn H., Destaing O., Sims N.A. et al. Defective microtubule-dependent podosome organization in osteoclasts leads to increased bone density in Pyk2(-/-) mice. J. Cell Biol. 2007; 178(6): 1053–64.
  85. Buckbinder L., Crawford D.T., Qi H. et al. Proline-rich tyrosine kinase 2 regulates osteoprogenitor cells and bone formation, and offers an anabolic treatment approach for osteoporosis. Proc. Natl. Acad. Sci. U S A 2007; 104(25): 10619–24.
  86. Eleniste P.P., Bruzzaniti A. Focal adhesion kinases in adhesion structures and disease. J. Signal Transduct. 2012; 2012: 296450.
  87. Uniprot for Angiopoietin-1 receptor. Available from: http://www.uniprot. org/uniprot/Q02763.
  88. Barton W.A., Tzvetkova-Robev D., Miranda E.P. et al. Crystal structures of the Tie2 receptor ectodomain and the angiopoietin-2-Tie2 complex. Nat. Struct. Mol. Biol. 2006; 13(6): 524–32.
  89. Huang H., Bhat A., Woodnutt G. et al. Targeting the ANGPT-TIE2 pathway in malignancy. Nat. Rev. Cancer 2010; 10(8): 575–85.
  90. Sato T.N., Tozawa Y., Deutsch U. et al. Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature 1995; 376(6535): 70–4.
  91. Jones N., Voskas D., Master Z. et al. Rescue of the early vascular defects in Tek/Tie2 null mice reveals an essential survival function. EMBO Rep. 2001; 2(5): 438–45.
  92. Peters K.G., Kontos C.D., Lin P.C. et al. Functional significance of Tie2 signaling in the adult vasculature. Rec. Prog. Horm. Res. 2004; 59: 51–71.
  93. Fukuhara S., Sako K., Noda K. et al. Angiopoietin-1/Tie2 receptor signaling in vascular quiescence and angiogenesis. Histol. Histopathol. 2010; 25(3): 387–96.
  94. Elice F., Rodeghiero F. Side effects of anti-angiogenic drugs. Thromb. Res. 2012; 129(Suppl. 1): S50–3.
  95. Aichberger K.J., Herndlhofer S., Schernthaner G.H. et al. Progressive peripheral arterial occlusive disease and other vascular events during nilotinib therapy in CML. Am. J. Hematol. 2011; 86(7): 533–9.
  96. Uniprot for VEGFR1. Available from: http://www.uniprot.org/uniprot/ P17948.
  97. Uniprot for VEGFR2. Available from: http://www.uniprot.org/uniprot/ P35968.
  98. Uniprot for VEGFR3. Available from: http://www.uniprot.org/uniprot/ P35916.
  99. Leppanen V.M., Tvorogov D., Kisko K. et al. Structural and mechanistic insights into VEGF receptor 3 ligand binding and activation. Proc. Natl. Acad. Sci. U S A 2013; 110(32): 12960–5.
  100. Stuttfeld E., Ballmer-Hofer K. Structure and function of VEGF receptors. IUBMB Life 2009; 61(9): 915–22.
  101. Olsson A.K., Dimberg A., Kreuger J. et al. VEGF receptor signalling — in control of vascular function. Nat. Rev. Mol. Cell Biol. 2006; 7(5): 359–71.
  102. Takahashi H., Shibuya M. The vascular endothelial growth factor (VEGF)/VEGF receptor system and its role under physiological and pathological conditions. Clin. Sci. (London) 2005; 109(3): 227–41.
  103. Kamba T., McDonald D.M. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br. J. Cancer 2007; 96(12): 1788–95.
  104. Dy G.K., Adjei A.A. Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA Cancer J. Clin. 2013; 63(4): 249–79.
  105. Baccarani M., Deininger M.W., Rosti G. et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood 2013; 122(6): 872–84.
  106. Soverini S., Colarossi S., Gnani A. et al. Resistance to dasatinib in Philadelphia-positive leukemia patients and the presence or the selection of mutations at residues 315 and 317 in the BCR-ABL kinase domain. Haematologica 2007; 92(3): 401–4.
  107. Гусарова Г.А., Туркина А.Г., Колошейнова Т.И. и др. Клинические аспекты применения нилотиниба при лечении больных хроническим миелолейкозом в хронической фазе. Гематол. и трансфузиол. 2012; 4: 3–11. [Gusarova G.A., Turkina A.G., Kolosheynova T.I. et al. Clinical aspects of nilotinib administration in management of patients with chronic myeloid leukemia in chronic phase. Gematol. i transfuziol. 2012; 4: 3–11. (In Russ.)].
  108. Лазарева О.В., Костина И.Э., Туркина А.Г. Лекарственно-индуци- рованный пневмонит: редкое осложнение терапии иматиниба мезилатом у больных хроническим миелолейкозом. Клин. онкогематол. 2010; 3(1): 47–52.  [Lazareva O.V., Kostina I.Ye., Turkina A.G. Drug-induced pneumonitis: rare complication of imatinib mesylate therapy in patients with chronic myeloid leukemia. Klin. onkogematol. 2010; 3(1): 47–52. (In Russ.)].
  109. Виноградова О.Ю., Туркина А.Г., Воронцова А.В. и др. Применение дазатиниба у больных в хронической стадии хронического миелолейкоза, резистентных либо не переносящих терапию иматинибом. Тер. арх. 2009; 7: 41–6.  [Vinogradova O.Yu., Turkina A.G., Vorontsova A.V. et al. Dasatinib administration to patients with chronic phase of chronic myeloid leukemia, who are resistant or intolerant to dasatinib. Ter. arkh. 2009; 7: 41–6. (In Russ.)].