Role of c-MYC, BCL2, and BCL6 Expression in Pathogenesis of Diffuse Large B-Cell Lymphoma

A.E. Misyurina1, V.A. Misyurin2, E.A. Baryakh1, A.M. Kovrigina1, S.K. Kravchenko1

1 Hematology Research Center under the Ministry of Health of the Russian Federation, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

2 N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: A.E. Misyurina, Graduate student 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel: +7(909)637-32-49; e-mail: anna.lukina1@gmail.com

For citation: Misyurina A.E., Misyurin V.A., Baryakh E.A., Kovrigina A.M., Kravchenko S.K. Role of c-MYC, BCL2, and BCL6 Expression in Pathogenesis of Diffuse Large B-Cell Lymphoma. Klin. Onkogematol. 2014; 7(4): 512–521 (In Russ.).


ABSTRACT

According to modern concepts based on results of examination of the gene expression profile, there are several subtypes of diffuse large B cell lymphoma (DLBCL): germinal center B cell-like (GCB) and activated B cell-like (ABC) lymphomas. Genes c-MYC, BCL6, and BCL2 are key regulators of B-cell germinal (follicular) differentiation. Genetic abnormalities with their participation are most common in molecular pathogenesis of DLBCL. A total level of activity as well as mechanisms that lead to overexpression each of these genes and production of corresponding proteins have an impact on a disease prognosis. We assume that quantitative assay of c-MYC, BCL6, and BCL2 gene expression, as well as proteins encoded by these genes, can allow to determine high risk DLBCL patients with great accuracy.


Keywords: diffuse large B cell lymphoma, molecular subtypes, risk groups, c-MYC, BCL6, BCL2.

Accepted: September 8, 2014

Read in PDF (RUS)pdficon


REFERENCES

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

Biology of Hematopoietic Stem Cell Niche

N.Yu. Semenova, S.S. Bessmel’tsev, V.I. Rugal’

Russian Scientific Research Institute of Hematology and Transfusiology under the Federal Medico-Biological Agency, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

For correspondence: S.S. Bessmel’tsev, DSci, Professor, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel: +7(812)717-67-80; e-mail: bsshem@hotmail.com

For citation: Semenova N.Yu., Bessmel’tsev S.S., Rugal’ V.I. Biology of Hematopoietic Stem Cell Niche. Klin. Onkogematol. 2014; 7(4): 501–510 (In Russ.).


ABSTRACT

The article presents up-to-date data on the role of bone marrow stromal niche in hematopoietic stem cells regulation (HSC). It describes stages of development of the hematopoietic niche concept. Characteristics of stromal cellular elements which form the niche are presented. Mechanisms of HSC regulation by the stromal niche are reported. The role of the niche in HSC leukemic transformation is discussed. It also presents data on structural changes in the niche in case of HSC development disorder.


Keywords: hematopoietic stem cell, bone marrow, hematopoietic stem cell niche, microenvironment.

Accepted: September 1, 2014

Read in PDF (RUS)pdficon


REFERENCES

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

Supplemental Blood Circulation System in Hematologic Malignancies

A.A. Vartanyan

N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: A.A. Vartanyan, DSci, Senior scientific worker, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel: +7(499)324-10-65; e-mail: zhivotov57@mail.ru

For citation: Vartanyan A.A. Supplemental Blood Circulation System in Hematologic Malignancies. Klin. Onkogematol. 2014; 7(4): 491–500 (In Russ.).


ABSTRACT

Neoangiogenesis, i.e. formation of new blood microvessels on the basis of already existing vascular network, is a prerequisite for tumor growth. For years, neoangiogenesis has been considered the only way for delivering oxygen and nutrients to the tumor. However, over the last years, other mechanism of tumor vascularization has been studied. Formation of highly structured vascular channels from tumor cells in the absence of endothelial cells and fibroblasts surrounded by basal membrane, or vasculogenic mimicry (VM) is considered a supplemental blood supply system. VM was found in almost all types of tumors and its occurrence is strongly associated with poor prognosis. This review summarizes basic VM characteristics in solid tumors and hematological malignancies. It also discusses the clinical significance of the above mentioned phenomenon in diagnosing tumors and predicting their course.


Keywords: neoangiogenesis, vasculogenic mimicry, hematological malignancies.

Accepted: September 1, 2014

Read in PDF (RUS)pdficon


REFERENCES 

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

Lymphomas in HIV-Infected Patients: Literature Review

A.V. Pivnik1, N.V. Seregin2, Yu.G. Parkhomenko3,4, O.A. Tishkevich4, A.M. Kovrigina5, Е.B. Likunov6

1 N.N. Pletnev Moscow Clinical Research Center, Department of Health, Moscow, Russian Federation

2 Moscow Municipal Cancer Dispensary No. 3, Moscow, Russian Federation

3 Research Institute of Human Morphology, RAMS, Moscow, Russian Federation

4 Clinical Infectious Diseases Hospital No. 2, Department of Health, Moscow, Russian Federation

5 Hematology Research Center, RF MH, Moscow, Russian Federation

6 American Medical Center, Moscow, Russian Federation

Address correspondence to: pivnikav@gmail.com

For citation: Pivnik A.V., Seregin N.V., Parkhomenko Yu.G., Tishkevich O.A., Kovrigina A.M., Likunov E.B. Lymphomas in HIV-Infected Patients: Literature Review. Klin. onkogematol. 2014; 7(3): 264–77 (In Russ.).


ABSTRACT

This review presents data on incidence, pathogenesis, diagnosis, and treatment of lymphoid malignancies in HIV-infected patients. Articles published by Russian and foreign authors are being reviewed. The principle role of decreased CD4+ lymphocyte count in development of secondary diseases in HIV patients is emphasized. Data on the structure of death causes in HIV-infected patients published by Russian authors are presented. They demonstrate that lymphoma is the 5th most common death cause (of 6 leading death causes). Hodgkin’s lymphoma in HIV-infected patients is not discussed in the review, because its pathogenesis differs from that of aggressive lymphomas and requires a separate discussion.


Keywords: lymphomas in HIV-infected patients, HIV, AIDS, secondary disorders, death causes, pathogenesis, diagnosis, treatment.

Accepted: May 15, 2014

Read in PDF (RUS)pdficon 


REFERENCES

  1. Ship J.A., Wolff A., Selik R.M. Epidemiology of acquired immune deficiency syndrome in persons aged 50 years or older. J. Acquir. Immune Defic. Syndr. 1991; 4(1): 84–8.
  2. Lyles R.H., Munoz A., Yamashita T.E. et al. Natural history of HIV type 1 viremia after seroconversion and proximal to AIDS in a large cohort of homosexual men. J. Infect. Dis. 2000; 181: 872–80.
  3. Бобкова М.Р. Иммунитет и ВИЧ-инфекция (популярные лекции). М.: Олимпия Пресс, 2006. 240 с. [Bobkova M.R. Immunitet i VICh-infektsiya (populyarnye lektsii). (Immunity and HIV infection (popular lectures)). Moscow: Olimpiya Press Publ., 2006. 240 p.].
  4. Mellors J.W., Munoz A.M., Giorgi J.V. et al. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann. Intern. Med. 1997; 126: 946–54.
  5. UNAIDS Technical Update (UNAIDS Best Practice Collection: Technical Update). UNAIDS (1997b): HIV testing methods. Geneva: UNAIDS, November 1997.
  6. Покровский В.В., Ермак Т.Н., Беляева В.В. ВИЧ инфекция. Клиника, диагностика, лечение. М.: ГЭОТАР-Медиа, 2003: 356–60. [Pokrovskii V.V., Ermak T.N., Belyaeva V.V. VICh infektsiya. Klinika, diagnostika, lechenie. (HIV infection. Clinical picture, diagnosis, treatment). Moscow: GEOTAR-Media Publ., 2003. pp. 356–60].
  7. ВИЧ-инфекция и СПИД: национальное руководство. Под. ред. В.В. Пок ровского. М.: ГЭОТАР-Медиа, 2013. 608 с.  [Pokrovskii V.V., ed. VICh-infektsiya i SPID: natsional’noe rukovodstvo. (HIV and AIDS: national guidelines). Moscow: GEOTAR-Media Publ., 2013. 608 p.].
  8. Покровский В.И., Покровский В.В., Потекаев С.Н. и др. Первый случай СПИД у гражданина СССР. Тер. арх. 1988; 7: 10–14. [Pokrovskii V.I., Pokrovskii V.V., Potekaev S.N. et al. The first case of AIDS in a USSR resident. Ter. arkh. 1988; 7: 10–4. (In Russ.)].
  9. Тишкевич О.А., Шахгильдян В.И., Пархоменко Ю.Г. Структура ле- тальных исходов и патологическая анатомия у больных ВИЧ-инфекцией в Москве. Эпидемиол. и инфек. бол. 2004; 4: 42–6. [Tishkevich O.A., Shakhgil’dyan V.I., Parkhomenko Yu.G. The Structure of lethal outcomes and morbid anatomy in HIV infected patients in Moscow. Epidemiol. i infek. bol. 2004; 4: 42–6. (In Russ.)].
  10. Гармаева Т.Ц. Вирусные гепатиты В и С у больных заболеваниями системы крови: Автореф. дис. … д-ра мед. наук. М., 2012. [Garmaeva T.Ts. Virusnye gepatity B i C u bol’nykh zabolevaniyami sistemy krovi: Avtoref. dis. … d-ra med. nauk. (Viral hepatitis B and C in patients with blood diseases: Author’s abstract of doctor’s thesis). Moscow, 2012.].
  11. Gonzalez R., Jacobus J., Martin M. Investigating Neurocognitive Features of Hepatitis C Virus Infection in Drug Users: Potential Challenges and Lessons Learned from the HIV Literature. CID 2005; 41: S45–49.
  12. Hilsabeck R.C., Castellon S.A., Hinkin Ch.H. Neuropsychological Aspects of Coinfection with HIV and Hepatitis C Virus. CID 2005; 41: S38–44.
  13. Desai J., Mitnick R.J., Henry D.H., Llena J., Sparano J.A. Patterns of central nervous system recurrence in patients with systemic human immunodeficiency virus-associated non-Hodgkin lymphoma. Cancer 1999; 86: 1840–7.
  14. Леви Дж.Э. ВИЧ и патогенез СПИДа: Пер. с англ., 3-е изд. М.: На- учный мир, 2010: 16. [Levy Jay A. HIV and the Pathogenesis of AIDS. 3rd edition. ASM Press, 2007, 643 p. (Russ. Ed. Levy J.A. VICh i patogenez SPIDa. Moscow: Nauchnyi Mir Publ., 2010. рр. 16.)].
  15. Allers K., Hutter G., Hofmann J. et al. Evidence for the cure of HIV infection 32 stem cell transplantation. Blood 2011; 117(10): 2791–9.D32/Dby CCR5
  16. Hosseini I., MacGabhann F., Thomas P.G. (eds.) APOBEC3G-Augmented Stem Cell Therapy to Modulate HIV Replication: A Computational Study. PLoS One 2013; 8(5): e63984.
  17. Kenneth L.M., Johnson M., D’Aquila R.T. APOBEC3G Complexes Decrease Human Immunodeficiency Virus Type 1 Production. J. Virol. 2011; 85(18): 9314–26.
  18. Collins K.L., Chen B.K., Walker B.D., Baltimore D. HIV-1 protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature 1998; 391: 397–401.
  19. Hammer S.M. Treatment for adult HIV infection: JAVA 2006; 296(7): 827–43.
  20. Hoffmann C., Chow K.U., Wolf E. et al. Strong impact of highly active antiretroviral therapy on survival in patients with human immunodeficiency virusassociated Hodgkin’s disease. Br. J. Haematol. 2004; 125: 455.
  21. Хоффман К., Кампс Б. Лечение ВИЧ-инфекции. Flying Publisher, 2005. 581 c. [Hoffmann C., Kamps B. HIV Therapy. Flying Publisher, 2005. 581 p. (Russ. Ed. Hoffmann C., Kamps B. Lechenie VICh-infektsii. Flying Publisher, 2005. 581 p.)].
  22. Бартлетт Дж. и др. Клинические аспекты ВИЧ-инфекции: Пер. с англ. М., 2013. 540 с. [Bartlett J. et al. Klinicheskie aspekty VICh-infekcii (Clinical aspects of HIV infection). Moscow, 2013. 540 p.].
  23. Ioachim H.L. Lymphadenopathies of HIV infection and AIDS. In: Benign and malignant lymphadenopathies. Ed. by G.A. Pangalis, A. Polliack. Harwood academic publisher, 1993: 159–70.
  24. Малый В.П. ВИЧ/СПИД (Новейший медицинский справочник). М.: Эксмо, 2009. 672 с. [Malyi V.P. VICh/SPID (Noveishii meditsinskii spravochnik) (HIV/AIDS (Up-todate manual)). Moscow: Eksmo Publ., 2009. 672 p.].
  25. Пивник А.В., Лукашев А.М., Туманова М.В. и др. Диагностика и ле- чение больных СПИД-ассоциированными лимфомами. Вестн. Моск. онкол. общества. 2008; 12: 553. [Pivnik A.V., Lukashev A.M., Tumanova M.V. et al. Diagnosis and treatment of patients with AIDS-associated lymphomas. Vestn. Mosk. onkol. obshchestva. 2008; 12: 553. (In Russ.)].
  26. Schuerman D.A. Clinical concerns: AIDS in the elderly. J. Gerontol. Nurs. 1994; 20(7): 11–7.
  27. Пархоменко Ю.Г., Тишкевич О.А., Шахгильдян В.И. Анализ аутопсий при ВИЧ-инфекции. Арх. пат. 2003; 3: 24–9. [Parkhomenko Yu.G., Tishkevich O.A., Shakhgil’dyan V.I. Analysis of autopsies in HIV infection. Arkh. pat. 2003; 3: 24–9. (In Russ.)].
  28. Mack K.A., Ory M.G. AIDS and older Americans at the end of the twentieth century. J. Acquir. Immune Defic. Syndr. 2003; 33(Suppl. 2): S68–75.
  29. Diamond C., Taylor T.H., Im T., Miradi M., Anton-Culver H. Improved survival and chemotherapy response among patients with AIDS-related Hodgkin’s lymphoma receiving highly active antiretroviral therapy. Hematol. Oncol. 2006; 10: 10.
  30. Powles T., Imami N., Nelson M., Gazzard B.G., Bower M. Effects of combination chemotherapy and HAART on immune parameters in HIV-1 associated lymphoma. AIDS 2002; 16: 531–6.
  31. Пивник А.В., Коровушкин В.Г., Пархоменко Ю.Г. и др. Дифференци- альная диагностика лимфаденопатий при ВИЧ/СПИД. Тер. арх. 2006; 4: 28–32. [Pivnik A.V., Korovushkin V.G., Parkhomenko Yu.G. et al. Differential diagnosis of lymphoadenopathies in HIV/AIDS. Ter. arkh. 2006; 4: 28–32. (In Russ.)].
  32. Пивник А.В., Коровушкин В.Г., Туаева А.О. и др. Тромбоцитопения при ВИЧ-инфекции. Тер. арх. 2008; 80(7): 75–80. [Pivnik A.V., Korovushkin V.G., Tuaeva A.O. et al. Thrombocytopenia in HIV infection. Ter. arkh. 2008; 80(7): 75–80. (In Russ.)].
  33. Малеев В.В., Полякова А.М., Кравченко А.В. Патогенетические механизмы нарушений системы гемостаза у больных ВИЧ-инфекцией. Эпидемиол. и инфек. бол. 2000; 3: 45–9. [Maleev V.V., Polyakova A.M., Kravchenko A.V. Pathogenetic mechanisms of hemostatic disorders in HIV infected patients. Epidemiol. i infek. bol. 2000; 3: 45–9. (In Russ.)].
  34. Stebbing J., Gazzard B., Mandalia S. et al. Antiretroviral treatment regimens and immune parameters in the prevention of systemic AIDS-related non-Hodgkin’s lymphoma. J. Clin. Oncol. 2004; 22(11): 2177–83.
  35. Young N.S., Gerson S.L., High K.A. Clinical Hematology. Philadelphia: Mosby Elsevier, 2006: 505–79.
  36. Февралева И.С., Глинщикова О.А., Макарик Т.В., Судариков А.Б. Мультиплексная диагностика вирусов гепатитов В, С и парвовируса В19 у больных, получающих множественные гемотрансфузии. Гематол. и транс- фузиол. 2008; 53(4): 54–6. [Fevraleva I.S., Glinshchikova O.A., Makarik T.V., Sudarikov A.B. Multiplex diagnosis of hepatitis B and C viruses and parvovirus B19 in patients receiving multiple hemotransfusions. Gematol. i transfuziol. 2008; 53(4): 54–6. (In Russ.)].
  37. Судариков А.Б. Молекулярная диагностика вирусов гепатита С, В, G и парвовируса B19 у гематологических больных: Автореф. дис. д-ра … биол. наук. М., 2012. [Sudarikov A.B. Molekulyarnaya diagnostika virusov gepatita S, V, G i parvovirusa  B19 u gematologicheskikh bol’nykh: Avtoref. dis. … d-ra biol. nauk. (Molecular diagnosis of hepatitis C, B, G viruses and parvovirus B19 in hematological patients). Moscow, 2012.].
  38. Wu H., Kuritzkes D.R., Clemon D.R. et al. Characterization of viral dynamics in HIV type 1-infected patients treated with combination antiretroviral therapy: relationships to host factors, cellular restoration, and virologic end points. J. Infect. Dis. 1999; 179: 799–807.
  39. Ganellos G.P., Lister T.A., Sklar J.L. The Lymphomas. Philadelphia: W.B. Saunders, 1998: 399–413.
  40. Armitage J.O., Cavalli F., Longo D. Annual of Lymphoid Malignancies. Taylor & Francis, 2000: 112–9.
  41. Navarro W.H., Kaplan L.D. AIDS-related lymphoproliferative disease. Blood 2006; 107(1): 13–20. 42. Little R.F. AIDS-related non-Hodgkin’s lymphoma: etiology, epidemiology, and impact of highly active antiretroviral therapy. Leuk. Lymphoma 2003; 44(3): 63–8.
  42. Gerard L., Galicier L., Maillard A. et al. Systemic non-Hodgkin lymphoma in HIV-infected patients with effective suppression of HIV replication: persistent occurrence but improved survival. J. Acquir. Immune Defic. Syndr. 2002; 30: 478–84.
  43. Levine A.M., Seneviratne L., Espina B.M. et al. Evolving characteristics of AIDS-related lymphoma. Blood 2000; 96: 4084–90.
  44. Matthews G.V., Bower M., Mandalia S. et al. Changes in AIDS-related lymphoma since the introduction of HAART. Blood 2000; 96: 2730–4.
  45. Besson C., Goubar A., Gabarre J. et al. Changes in AIDS-related lymphoma since the era of HAART. Blood 2001; 98: 2339–44.
  46. Levine A.M. AIDS-related lymphoma. Semin. Oncol. Nurs. 2006; 22(2): 80–9.
  47. Aboulafia D.M., Pantanowitz L., Dezube B.J. AIDS-Related Non-Hodgkin Lymphoma: Still a Problem in the Era of HAART. AIDS 2004; 14(11): 605–17.
  48. Kadan-Lottick N.S., Skluzacek M.C., Gurney J.G. Decreasing incidence rates of primary central nervous system lymphoma. Cancer 2002; 95: 193–202.
  49. Franceschi S., Dal Maso L., La Vecchia C. Advances in the epidemiology of HIV-associated non-Hodgkin’s lymphoma and other lymphoid neoplasms. Intern. J. Cancer 1999; 83: 481–5.
  50. Cinque P., Brytting M., Vago L. et al. Epstein-Barr virus DNA in cerebrospinal fluid from patients with AIDS-related primary lymphoma of the central nervous system. Lancet 1993; 342: 398–401.
  51. Oriol A., Ribera J.M., Esteve J. et al. Lack of influence of human immunodeficiency virus infection status in the response to therapy and survival of adult patients with mature B-cell lymphoma or leukemia. Results of the PETHEMALAL3/97 study. Haematologica 2003; 88(4): 445–53.
  52. Mounier N., Spina M., Gabarre J. et al. AIDS-related non-Hodgkin lymphoma: final analysis of 485 patients treated with risk adapted intensive chemotherapy. Blood 2006; 107(10): 3832–40.
  53. Lim S.T., Karim R., Tulpule A., Nathwani B.N., Levine A.M. Prognostic Factors in HIV-Related Diffuse Large-Cell Lymphoma: Before Versus After Highly Active Antiretroviral Therapy. J. Clin. Oncol. 2005; 23(33): 8477–82.
  54. Carbone A., Cesarman E., Spina M. HIV-associated lymphomas and gamma-herpesviruses. Blood 2009; 113(6): 1213–24.
  55. Simonelli C., Spina M., Cinelli R. et al. Clinical features and outcome of primary effusion lymphoma in HIV-infected patients: a single-institution study. J. Clin. Oncol. 2003; 21: 3948–54.
  56. Menendez J.A., Lilien D.L., Nanda А. et al. AIDS-Related Lymphoma. Abstr. Hematol. Oncol. 2005; 8(1): 20–30, 68–70.
  57. Delecluse H.J., Anagnostopoulos I., Dallenbach F. et al. Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection. Blood 1997; 89: 1413–20.
  58. Gisselbrecht C., Mounier N. Treatment of relapsed and refractory Hodgkin’s Lymphoma. In: Education program of the 10th congress of the European Hematology Association, 2005: 182–5.
  59. Gisselbrecht C., Mounier N. Treatment of poor prognosis non-Hodgkin’s lymphoma. In: Education program of the 10th congress of the European hematology association, 2005: 160–5.
  60. Bower M., Gazzard B., Mandalia S., Newsom-Davis T. et al. A prognostic index for systemic AIDS-related non-Hodgkin lymphoma treated in the era of highly active antiretroviral therapy. Ann. Intern. Med. 2005; 143: 265–73.
  61. Feigal E.G., Levine A.M., Biggar R.J. AIDS-related Cancers and Their Treatment. Marcel Dekker Inc., 2000: 97–124.
  62. Little R.F., Gutierrez M., Jaffe E.S. et al. HIV-Associated non-Hodgkin lymphoma: incidence, presentation, and prognosis. JAMA 2001; 285: 1880–5.
  63. Pauza C.D., Pyzalski R., Perlman S.B. et al. Positron emission tomography images of AIDS pathogenesis. Conf. Adv. AIDS Vaccine, 1997.
  64. Литвинов В.И., Макарова М.В., Краснова М.А. Нетуберкулезные микобактерии и микобактериозы. Эпидемиол. и инфек. бол. 2011; 6: 4–10. [Litvinov V.I., Makarova M.V., Krasnova M.A. Nontuberculous mycobacteria and mycobacteriosis. Epidemiol. i infek. bol. 2011; 6: 4–10. (In Russ.)].
  65. Литвинов В.И., Макарова М.В., Краснова М.А. Нетуберкулезные микобактерии. М.: МНПЦБТ, 2008. 256 с.  [Litvinov V.I., Makarova M.V., Krasnova M.A. Netuberkuleznye mikobakterii (Nontuberculous mycobacteria). Moscow: MNPTsBT Publ., 2008. 256 р.].
  66. Clarke C.A., Glaser S.L. Epidemiologic trends in HIV-associated lymphomas. Curr. Opin. Oncol. 2001; 13: 354–9.
  67. Clarke C.A., Glaser S.L. Epidemiologic trends in HIV-associated lymphomas. Curr. Opin. Oncol. 2003; 15: 267–90.
  68. Патологическая анатомия: национальное руководство. Под ред. М.А. Пальцева, Л.В. Кактурского, О.В. Зайратьянца. М.: ГЭОТАР-Медиа, 2011. 1264 с. [Pal’tsev M.A., Kakturskii L.V., Zairat’yants O.V., eds. Patologicheskaya anatomiya: natsional’noe rukovodstvo. (Morbid anatomy: national guidelines). Moscow: GEOTAR-Media Publ., 2011. 1264 p.].
  69. Stebbing J., Marvin V., Bower M. et al. The evidence-based treatment of AIDS-related non-Hodgkins lymphoma. Cancer Treat. Rev. 2004; 30: 249–53.
  70. Hartmann P., Rehwald U., Salzberger B. et al. BEACOPP therapeutic regimen for patients with Hodgkin’s disease and HIV infection. Ann. Oncol. 2003; 14: 1562–9.
  71. Antinori A., Cingolani A., Alba L. et al. Better response to chemotherapy and prolonged survival in AIDS-related lymphomas responding to highly active antiretroviral therapy. AIDS 2001; 15: 1483–91.
  72. Chadburn A., Hyjek E., Mathew S. et al. KSHV-positive solid lymphomas represent an extra-cavitary variant of primary effusion lymphoma. Am. J. Surg. Pathol. 2004; 28(11): 1401–16.
  73. Пивник А.В. Применение ритуксимаба у больных с ВИЧ-инфекцией. Клин. онкогематол. 2013; 6(1): 84–90.  [Pivnik A.V. The use of rituximab in patients with HIV infection. Klin. onkogematol. 2013; 6(1): 84–90. (In Russ.)].
  74. Lim S.T., Karim R., Nathwani B.N. et al. AIDS-related Burkitt’s lymphoma versus diffuse large cell lymphoma in the pre-highly active antiretroviral therapy (HAART) and HAART eras: significant differences in survival with standard chemotherapy. J. Clin. Oncol. 2005; 23: 4430–8.
  75. Boue F., Gabarre J., Gisselbrecht C. et al. CHOP chemotherapy plus Rituximab in HIV patients with high grade lymphoma – results of an ANRS Trial. Blood 2002; 22: 470a (abstract 1824).
  76. Dunleavy K., Wilson W.H. How I treat HIV-associated lymphoma. Blood 2012; 119(14): 3245–55.
  77. Krishnan A., Molina A., Zaia J. et al. Durable remissions with autologous stem cell transplantation for high-risk HIV-associated lymphomas. Blood 2005; 105(2): 874–8.
  78. Sparano J.A., Wiernik P.H., Strack M. et al. Infusional cyclophosphamide, doxorubicin, and etoposide in human immunodeficiency virus- and human T-cell leukemia virus type I-related non-Hodgkin’s lymphoma: a highly active regimen. Blood 1993; 81: 2810–5.
  79. Cortes J., Thomas D., Rios A. et al. Hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone and highly active antiretroviral therapy for patients with acquired immunodeficiency syndrome-related Burkitt lymphoma/leukemia. Cancer 2002; 94: 1492–9.
  80. Molina A., Zaia J., Krishnan A. Treatment of human immunodeficiency virus-related lymphoma with haematopoietic stem cell transplantation. Blood Rev. 2003; 17: 249–58.
  81. Navarro J.T., Ribera J.M., Oriol A. et al. Influence of HAART on response to treatment and survival in patients with AIDS-related non-Hodgkin’s lymphoma treated with cyclophosphamide, hydroxydoxorubicin, vincristine and prednisone. Br. J. Haematol. 2001; 112: 909–15.
  82. Барях Е.А., Кременецкая А.М., Кравченко С.К. и др. Новый короткий высокоинтенсивный протокол терапии лимфомы Беркитта у взрослых ЛБ- М-04: промежуточные результаты. Гематол. и трансфузиол. 2006; 51(6): 45–9. [Baryakh E.A., Kremenetskaya A.M., Kravchenko S.K. et al. New abridged highly intensive protocol LB-M-04 for treatment of Burkitt’s lymphoma in adults: interim results. Gematol. i transfuziol. 2006; 51(6): 45–9. (In Russ.)].
  83. Барях Е.А., Звонков Е.Е., Кременецкая А.М. и др. Лечение Беркитто- подобной лимфомы у взрослых. Тер. арх. 2005; 7: 53–8. [Baryakh E.A., Zvonkov E.E., Kremenetskaya A.M. et al. Treatment of Burkittlike lymphoma in adults. Ter. arkh. 2005; 7: 53–8. (In Russ.)].
  84. Пивник А.В., Пархоменко Ю.Г., Криволапов Ю.А. и др. Соматические проблемы ВИЧ-медицины: СПИД-ассоциированные лимфомы. Онкогема- тология 2007; 3: 27–37. [Pivnik A.V., Parkhomenko Yu.G., Krivolapov Yu.A. et al. Somatic problems of HIV medicine: AIDS-associated lymphomas. Onkogematologiya 2007; 3: 27–37. (In Russ.)].
  85. Kaplan L.D., Lee J.Y., Ambinder R.F. еt al. Rituximab does not improve clinical outcome in a randomized phase 3 trial of CHOP with or without rituximab in patients with HIV-associated non-Hodgkin’s lymphoma: AIDS-Malignancies Consortium Trial 010. Blood 2005; 106: 1538–43.
  86. Kaplan L.D., Scadden D.T. No benefit from Rituximab in a randomized phase III trial of CHOP with or without rituximiab for patients with HIV-associated non-Hodgkins lymphoma: AIDS-Malignancies Consortium study 010. Proc. Am. Soc. Clin. Oncol. 2003; 22: 564 (abstract 2268).
  87. Krishnan A., Molina A., Zaia J. et al. Autologous stem cell transplantation for HIV-associated lymphoma. Blood 2001; 98: 3857–9.

B-Cell Receptor Signaling Pathway: Mechanisms and Inhibitors

E.A. Nikitin

Hematology Research Center, RF MH, Moscow, Russian Federation

For citation: Nikitin E.A. B-Cell Receptor Signaling Pathway: Mechanisms and Inhibitors. Klin. onkogematol. 2014; 7(3): 251–63 (In Russ.).


ABSTRACT

Differentiation and survival of normal B-lymphocytes critically depends on the B-cell receptor (BCR) signaling pathway. Lymphoid malignancies use different aspects of the BCR-signaling pathway to provide their proliferation and growth. They manifest themselves in different forms, such as the form of BCR particular antigenic specificity, or the form of activating or, to the contrary, inhibiting gene mutations encoding proteins involved in BCR-signaling. A number of small molecules inhibit different proteins of BCR-signaling cascade. In this review, we dwell on normal and defective BCR-signaling pathways, as well as on tyrosine kinase inhibitors that are being widely used in clinical trials and will likely change the management of lymphoid malignancies.


Keywords: В-cell lymphomas, B-cell receptor, kinase inhibitors.

Address correspondence to: еugene_nikitin@mail.ru

Read in PDF(RUS)pdficon


REFERENCES

  1. Dameshek W., Schwartz R.S. Leukemia and auto-immunization-some possible relationships. Blood 1959; 14: 1151–8.
  2. Goodlad J.R. et al. Primary cutaneous B-cell lymphoma and Borrelia burgdorferi infection in patients from the Highlands of Scotland. Am. J. Surg. Pathol. 2000; 24(9): 1279–85.
  3. Vasudevan B., Chatterjee M. Lyme borreliosis and skin. Indian J. Dermatol. 2013; 58(3): 167–74.
  4. Schollkopf C., Melbye M., Munksgaard L. et al. Borrelia infection and risk of non-Hodgkin lymphoma. Blood 2008; 111(12): 5524–9.
  5. Garbe C., Stein H., Dienemann D., Orfanos C.E. Borrelia burgdorferiassociated cutaneous B cell lymphoma: clinical and immunohistologic characterization of four cases. J. Am. Acad. Dermatol. 1991; 24(4): 584–90.
  6. Wotherspoon A.C., Doglioni C., Diss T.C. et al. Regression of primary lowgrade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet 1993; 342(8871): 575–7.
  7. Du M.Q., Isaccson P.G. Gastric MALT lymphoma: from aetiology to treatment. Lancet Oncol. 2002; 3(2): 97–104.
  8. Ferreri A.J., Ponzoni M., Guidoboni M. et al. Regression of ocular adnexal lymphoma after Chlamydia psittaci-eradicating antibiotic therapy. J. Clin. Oncol. 2005; 23(22): 5067–73.
  9. Ferreri A.J., Govi S., Pasini E. et al. Chlamydophila psittaci eradication with doxycycline as first-line targeted therapy for ocular adnexae lymphoma: final results of an international phase II trial. J. Clin. Oncol. 2012; 30(24): 2988–94.
  10. Al-Saleem T., Al-Mondhiry H. Immunoproliferative small intestinal disease (IPSID): a model for mature B-cell neoplasms. Blood 2005; 105(6): 2274–80.
  11. Anttila T.I., Lehtinen T., Leinonen M. et al. Serological evidence of an association between chlamydial infections and malignant lymphomas. Br. J. Haematol. 1998; 103(1): 150–6.
  12. Ishimatsu Y., Mukae H., Matsumoto K. et al. Two cases with pulmonary mucosa-associated lymphoid tissue lymphoma successfully treated with clarithromycin. Chest 2010; 138(3): 730–3.
  13. Fujimura M., Chin K., Sekita N. et al. Regression of mucosa-associated lymphoid tissue lymphoma of the bladder after antibiotic therapy: a case report. Hinyokika Kiyo 2008; 54(12): 783–6.
  14. Van den Bosch J., Kropman R.F., Blok P., Wijermans P.W. Disappearance of a mucosa-associated lymphoid tissue (MALT) lymphoma of the urinary bladder after treatment for Helicobacter pylori. Eur. J. Haematol. 2002; 68(3): 187–8.
  15. Oscier D., Bramble J., Hodges E., Wright D. Regression of mucosaassociated lymphoid tissue lymphoma of the bladder after antibiotic therapy. J. Clin. Oncol. 2002; 20(3): 882.
  16. Quinn E.R., Chan C.H., Hadlock K.G. et al. The B-cell receptor of a hepatitis C virus (HCV)-associated non-Hodgkin lymphoma binds the viral E2 envelope protein, implicating HCV in lymphomagenesis. Blood 2001; 98(13): 3745–9.
  17. Kuppers R. Mechanisms of B-cell lymphoma pathogenesis. Nat. Rev. Cancer 2005; 5(4): 251–62.
  18. Martin S.W., Goodnow C.C. Burst-enhancing role of the IgG membrane tail as a molecular determinant of memory. Nat. Immunol. 2002; 3(2): 182–8.
  19. Dogan I., Bertocci B., Vilmont V. et al. Multiple layers of B cell memory with different effector functions. Nat. Immunol. 2009; 10(12): 1292–9.
  20. Vaandrager J.-W., Schuuring Ed., Kluin-Nelemans H.C. et al. DNA fiber fluorescence in situ hybridization analysis of immunoglobulin class switching in B-cell neoplasia: aberrant CH gene rearrangements in follicle center-cell lymphoma. Blood 1998; 92(8): 2871–8.
  21. Alizadeh A.A., Eisen M.B., Davis R.E. et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000; 403(6769): 503–11.
  22. Davis R.E., Ngo V.N., Lenz G. et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 2010; 463(7277): 88–92.
  23. Lenz G., Nagel I., Siebert R. et al. Aberrant immunoglobulin class switch recombination and switch translocations in activated B cell-like diffuse large B cell lymphoma. J. Exp. Med. 2007; 204(3): 633–43.
  24. Ruminy P., Etancelin P., Couronne L. et al. The isotype of the BCR as a surrogate for the GCB and ABC molecular subtypes in diffuse large B-cell lymphoma. Leukemia 2011; 25(4): 681–8.
  25. Klein U., Klein G., Ehlin-Henriksson B. et al. Burkitt’s lymphoma is a malignancy of mature B cells expressing somatically mutated V region genes. Mol. Med. 1995; 1(5): 495–505.
  26. Damle R.N., Wasil T., Fais F. et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 1999; 94(6): 1840–7.
  27. Hamblin T.J., Davis Z., Gardiner A. et al. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 1999; 94(6): 1848–54.
  28. Nikitin E.A., Pivnik A.V., Sudarikov A.B. et al. A comparison of the forms of chronic lympholeukemia in relation to the mutational status of the genes of the immunoglobulin variable region. Ter. Arkh. 2000; 72(7): 52–6.
  29. Hadzidimitriou A., Agathangelidis A., Darzentas N. et al. Is there a role for antigen selection in mantle cell lymphoma? Immunogenetic support from a series of 807 cases. Blood 2011; 118(11): 3088–95.
  30. Agathangelidis A., Darzentas N., Hadzidimitriou A. et al. Stereotyped B-cell receptors in one-third of chronic lymphocytic leukemia: a molecular classification with implications for targeted therapies. Blood 2012; 119(19): 4467–75.
  31. Herve M., Xu K., Ng Y.S. et al. Unmutated and mutated chronic lymphocytic leukemias derive from self-reactive B cell precursors despite expressing different antibody reactivity. J. Clin. Invest. 2005; 115(6): 1636–43.
  32. Catera R., Silverman G.J., Hatzi K. et al. Chronic lymphocytic leukemia cells recognize conserved epitopes associated with apoptosis and oxidation. Mol. Med. 2008; 14(11–12): 665–74.
  33. Chu C.C., Catera R., Zhang L. et al. Many chronic lymphocytic leukemia antibodies recognize apoptotic cells with exposed nonmuscle myosin heavy chain IIA: implications for patient outcome and cell of origin. Blood 2010; 115(19): 3907–15.
  34. Herishanu Y., Perez-Galan P., Liu D. et al. The lymph node microenvironment promotes B-cell receptor signaling, NF-kappaB activation, and tumor proliferation in chronic lymphocytic leukemia. Blood 2011; 117(2): 563–74.
  35. Duhren-von Minden M., Ubelhart R., Schneider D. et al. Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature 2012; 489(7415): 309–12.
  36. Zhu D., Ottensmeier C.H., Du M.Q. et al. Incidence of potential glycosylation sites in immunoglobulin variable regions distinguishes between subsets of Burkitt’s lymphoma and mucosa-associated lymphoid tissue lymphoma. Br. J. Haematol. 2003; 120(2): 217–22.
  37. Radcliffe C.M., Arnold J.N., Suter D.M. et al. Human follicular lymphoma cells contain oligomannose glycans in the antigen-binding site of the B-cell receptor. J. Biol. Chem. 2007; 282(10): 7405–15.
  38. CoelhoV., Krysov S., Ghaemmaghami A.M. et al. Glycosylation of surface Ig creates a functional bridge between human follicular lymphoma and microenvironmental lectins. PNAS 2010; 107(43): 18587–92.
  39. Sachen K.L., Strohman M.J., Singletary J. et al. Self-antigen recognition by follicular lymphoma B-cell receptors. Blood 2012; 120(20): 4182–90.
  40. Marcucci F., Mele A. Hepatitis viruses and non-Hodgkin lymphoma: epidemiology, mechanisms of tumorigenesis, and therapeutic opportunities. Blood 2011; 117(6): 1792–8.
  41. Gisbert J.P., Garcia-Buey L., Pajares J.M. et al. Systematic review: regression of lymphoproliferative disorders after treatment for hepatitis C infection. Aliment. Pharmacol. Ther. 2005; 21(6): 653–62.
  42. Victora G.D., Nussenzweig M.C. Germinal centers. Annu. Rev. Immunol. 2012; 30: 429–57.
  43. Clark M.R., Tanaka A., Powers S.E., Veselits M. Receptors, subcellular compartments and the regulation of peripheral B cell responses: the illuminating state of anergy. Mol. Immunol. 2011; 48(11): 1281–6.
  44. Yang J., Reth M. Oligomeric organization of the B-cell antigen receptor on resting cells. Nature 2010; 467(7314): 465–9.
  45. Pierce S.K., Liu W. The tipping points in the initiation of B cell signalling: how small changes make big differences. Nat. Rev. Immunol. 2010; 10(11): 767–77.
  46. Reth M. Antigen receptor tail clue. Nature 1989; 338(6214): 383–4.
  47. Saijo K., Schmedt C., Su I.H. et al. Essential role of Src-family protein tyrosine kinases in NF-kappaB activation during B cell development. Nat. Immunol. 2003; 4(3): 274–9.
  48. Rowley R.B., Burkhardt A.L., Chao H.G. et al. Syk protein-tyrosine kinase is regulated by tyrosine-phosphorylated Ig alpha/Ig beta immunoreceptor tyrosine activation motif binding and autophosphorylation. J. Biol. Chem. 1995; 270(19): 11590–4.
  49. Oellerich T., Bremes V., Neumann K. et al. The B-cell antigen receptor signals through a preformed transducer module of SLP65 and CIN85. EMBO J. 2011; 30(17): 3620–34.
  50. Watanabe D., Hashimoto S., Ishiai M. et al. Four tyrosine residues in phospholipase C-gamma 2, identified as BTK-dependent phosphorylation sites, are required for B cell antigen receptor-coupled calcium signaling. J. Biol. Chem. 2001; 276(42): 38595–601.
  51. Ozdener F., Dangelmaier C., Ashby B. et al. Activation of phospholipase Cgamma2 by tyrosine phosphorylation. Mol. Pharmacol. 2002; 62(3): 672–9.
  52. Shinohara H., Yasuda T., Aiba Y. et al. PKC beta regulates BCR-mediated IKK activation by facilitating the interaction between TAK1 and CARMA1. J. Exp. Med. 2005; 202(10): 1423–31.
  53. Coughlin J.J., Stang S.L., Dower N.A., Stone J.C. RasGRP1 and RasGRP3 regulate B cell proliferation by facilitating B cell receptor-Ras signaling. J. Immunol. 2005; 175(11): 7179–84.
  54. Xu Y., Harder K.W., Huntington N.D. et al. Lyn tyrosine kinase: accentuating the positive and the negative. Immunity 2005; 22(1): 9–18.
  55. Deane J.A., Fruman D.A. Phosphoinositide 3-kinase: diverse roles in immune cell activation. Annu. Rev. Immunol. 2004; 22: 563–98.
  56. Yuan T.L., Cantley L.C. PI3K pathway alterations in cancer: variations on a theme. Oncogene 2008; 27(41): 5497–510.
  57. Laplante M., Sabatini D.M. mTOR signaling in growth control and disease. Cell 2012; 149(2): 274–93.
  58. Stone J.C. Regulation and Function of the RasGRP Family of Ras Activators in Blood Cells. Genes Cancer 2011; 2(3): 320–34.
  59. Guo B., Su T.T., Rawlings D.J. Protein kinase C family functions in B-cell activation. Curr. Opin. Immunol. 2004; 16(3): 367–73.
  60. Suzuki A., Kaisho T., Ohishi M. et al. Critical roles of PTEN in B cell homeostasis and immunoglobulin class switch recombination. J. Exp. Med. 2003; 197(5): 657–67.
  61. O’Neill S.K., Getahun A., Gauld S.B. et al. Monophosphorylation of CD79a and CD79b ITAM motifs initiates a SHIP-1 phosphatase-mediated inhibitory signaling cascade required for B cell anergy. Immunity 2011; 35(5): 746–56.
  62. Pao L.I., Lam K.P., Henderson J.M. et al. B cell-specific deletion of protein-tyrosine phosphatase Shp1 promotes B-1a cell development and causes systemic autoimmunity. Immunity 2007; 27(1): 35–48.
  63. Liu C., Bai X., Wuet J. et al. N-wasp is essential for the negative regulation of B cell receptor signaling. PLoS Biol. 2013; 11(11): e1001704.
  64. Ingley E. Src family kinases: regulation of their activities, levels and identification of new pathways. Biochim. Biophys. Acta 2008; 1784(1): 56–65.
  65. Lam K.P., Kuhn R., Rajewsky K. In vivo ablation of surface immunoglobulin on mature B cells by inducible gene targeting results in rapid cell death. Cell 1997; 90(6): 1073–83.
  66. Kraus M., Alimzhanov M.B., Rajewsky N., Rajewsky K. Survival of resting mature B lymphocytes depends on BCR signaling via the Igalpha/beta heterodimer. Cell 2004; 117(6): 787–800.
  67. Srinivasan L., Sasaki Y., Calado D.P. et al. PI3 kinase signals BCRdependent mature B cell survival. Cell 2009; 139(3): 573–86.
  68. Baracho G.V., Miletic A.V., Omori S.A. et al. Emergence of the PI3-kinase pathway as a central modulator of normal and aberrant B cell differentiation. Curr. Opin. Immunol. 2011; 23(2): 178–83.
  69. Ramadani F., Bolland D.J., Garcon F. et al. The PI3K isoforms p110alpha and p110delta are essential for pre-B cell receptor signaling and B cell development. Sci. Signal. 2010; 3(134): ra60.
  70. Ngo V.N., Davis R.E., Lamy L. et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature 2006; 441(7089): 106–10.
  71. Lenz G. et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science 2008; 319(5870): 1676–9.
  72. Davis R.E., Davis E., Ngo V.N. et al. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J. Exp. Med. 2001; 194(12): 1861–74.
  73. Rawlings D.J., Sommer K., Moreno-Garcia M.E. The CARMA1 signalosome links the signalling machinery of adaptive and innate immunity in lymphocytes. Nat. Rev. Immunol. 2006; 6(11): 799–812.
  74. Bajpai U.D., Zhang K., Teutsch M. et al. Bruton’s tyrosine kinase links the B cell receptor to nuclear factor kappaB activation. J. Exp. Med. 2000; 191(10): 1735–44.
  75. Petro J.B., Rahman S.M.J., Ballard D.W. et al. Bruton’s tyrosine kinase is required for activation of IkappaB kinase and nuclear factor kappaB in response to B cell receptor engagement. J. Exp. Med. 2000; 191(10): 1745–54.
  76. Naylor T.L., Tang H., Ratsch B.A. et al. Protein kinase C inhibitor sotrastaurin selectively inhibits the growth of CD79 mutant diffuse large B-cell lymphomas. Cancer Res. 2011; 71(7): 2643–53.
  77. Wardemann H., Yurasov S., Schaefer A. et al. Predominant autoantibody production by early human B cell precursors. Science 2003; 301(5638): 1374–7.
  78. Gauld S.B., Benschop R.J., Merrell K.T., Cambier J.C. Maintenance of B cell anergy requires constant antigen receptor occupancy and signaling. Nat. Immunol. 2005; 6(11): 1160–7.
  79. Yarkoni Y., Getahun A., Cambier J.C. Molecular underpinning of B-cell anergy. Immunol. Rev. 2010; 237(1): 249–63.
  80. Quach T.D., Manjarrez-Orduno N., Adlowitz D.G. et al. Anergic responses characterize a large fraction of human autoreactive naive B cells expressing low levels of surface IgM. J. Immunol. 2011; 186(8): 4640–8.
  81. Smedby K.E., Hjalgrim H., Askling J. et al. Autoimmune and chronic inflammatory disorders and risk of non-Hodgkin lymphoma by subtype. J. Natl. Cancer Inst. 2006; 98(1): 51–60.
  82. Rui L., Schmitz R., Ceribelli M., Staudt L.M. Malignant pirates of the immune system. Nat. Immunol. 2011; 12(10): 933–40.
  83. Alfarano A., Indraccolo S., Circostaet P. et al. An alternatively spliced form of CD79b gene may account for altered B-cell receptor expression in Bchronic lymphocytic leukemia. Blood 1999; 93(7): 2327–35.
  84. Schmitz R., Young R.M., Ceribelliet M. et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 2012; 490(7418): 116–20.
  85. Sander S., Calado D.P., Srinivasan L. et al. Synergy between PI3K signaling and MYC in Burkitt lymphomagenesis. Cancer Cell 2012; 22(2): 167–79.
  86. Evan G.I., Wyllie A.H., Gilbert C.S. et al. Induction of apoptosis in fibroblasts by c-myc protein. Cell 1992; 69(1): 119–28.
  87. Quesada V., Conde L., Villamor N. et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat. Genet. 2012; 44(1): 47–52.
  88. Wang L., Lawrence M.S., Wan Y. et al. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N. Engl. J. Med. 2011; 365(26): 2497–506.
  89. Puente X.S. et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 2011; 475(7354): 101–5.
  90. Amrein P.C., Attar E.C., Takvorian T. et al. Phase II study of dasatinib in relapsed or refractory chronic lymphocytic leukemia. Clin. Cancer Res. 2011; 17(9): 2977–86.
  91. Friedberg J.W., Sharman J., Sweetenham J. et al. Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood 2010; 115(13): 2578–85.
  92. Advani R.H., Buggy J.J., Sharman J.P. et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/ refractory B-cell malignancies. J. Clin. Oncol. 2013; 31(1): 88–94.
  93. Wang M.L., Rule S., Martin P. et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 2013; 369(6): 507–16.
  94. Wyndham W., Gerecitano J.F., Goy A. et al. The Bruton’s tyrosine kinase (BTK) inhibitor, ibrutinib (PCI-32765), has preferential activity in the ABC subtype of relapsed/refractory de novo diffuse large B-cell lymphoma (DLBCL): interim results of a multicenter, open-label, phase 2 study. Blood (ASH Annual Meeting Abstracts) 2012; 120: 686.
  95. Byrd J.C., Furman R.R., Coutre S.E. et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 2013; 369(1): 32–42.
  96. Coutre S., Byrd J.C., Furman R.R. et al. Phase I study of CAL-101, an isoform-selective inhibitor of phosphatidylinositol 3-kinase P110d, in patients with previously treated chronic lymphocytic leukemia. J. Clin. Oncol. (ASCO Annual Meeting Abstracts) 2011; 29: 6631.
  97. Kahl B., Byrd J.C., Flinn I.W. et al. Clinical safety and activity in a phase 1 study of CAL-101, an isoform- selective inhibitor of phosphatidylinositol 3-kinase P110{delta}, in patients with relapsed or refractory non-Hodgkin lymphoma. Blood (ASH Annual Meeting Abstracts) 2010; 116(21): 1777.
  98. Zent C.S., LaPlant B.R., Johnston P.B. et al. The treatment of recurrent/ refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) with everolimus results in clinical responses and mobilization of CLL cells into the circulation. Cancer 2010; 116(9): 2201–7.
  99. Renner C., Zinzani P.L., Gressin R. et al. A multicenter phase II trial (SAKK 36/06) of single-agent everolimus (RAD001) in patients with relapsed or refractory mantle cell lymphoma. Haematologica 2012; 97(7): 1085–91.
  100. Witzig T.E., Reeder C.B., LaPlant B.R. et al. A phase II trial of the oral mTOR inhibitor everolimus in relapsed aggressive lymphoma. Leukemia 2011; 25(2): 341–7.
  101. Witzig T.E., Geyer S.M., Ghobrial I. et al. Phase II trial of single-agent temsirolimus (CCI-779) for relapsed mantle cell lymphoma. J. Clin. Oncol. 2005; 23(23): 5347–56.
  102. Smith S.M., van Besien K., Karrison T. et al. Temsirolimus has activity in non-mantle cell non-Hodgkin’s lymphoma subtypes: The University of Chicago phase II consortium. J. Clin. Oncol. 2010; 28(31): 4740–6.
  103. Bruton O.C. Agammaglobulinemia. Pediatrics 1952; 9(6): 722–8.
  104. Rawlings D.J., Saffran D.C., Tsukada S. et al. Mutation of unique region of Bruton’s tyrosine kinase in immunodeficient XID mice. Science 1993; 261(5119): 358–61.
  105. Quek L.S., Bolen J., Watson S.P. A role for Bruton’s tyrosine kinase (Btk) in platelet activation by collagen. Curr. Biol. 1998; 8(20): 1137–40.
  106. Kurosaki T., Hikida M. Tyrosine kinases and their substrates in B lymphocytes. Immunol. Rev. 2009; 228(1): 132–48.
  107. Rawlings D.J., Lin S., Scharenberg A.M. et al. Activation of BTK by a phosphorylation mechanism initiated by SRC family kinases. Science 1996; 271(5250): 822–5.
  108. Mohamed A.J., Yu L., Backesjo C.M. et al. Bruton’s tyrosine kinase (Btk): function, regulation, and transformation with special emphasis on the PH domain. Immunol. Rev. 2009; 228(1): 58–73.
  109. Hantschel O., Rix U., Schmidtet U. et al. The Btk tyrosine kinase is a major target of the Bcr-Abl inhibitor dasatinib. PNAS 2007; 104(33): 13283–8.
  110. Pan Z., Scheerens H., Li S.J. et al. Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. Chem. Med. Chem. 2007; 2(1): 58–61.
  111. Honigberg L.A., Smith A.M., Sirisawadet M. et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc. Natl. Acad. Sci. U S A 2010; 107(29): 13075–80.
  112. Herman S.E., Gordon A.L., Hertlein E. et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood 2011; 117(23): 6287–96.
  113. Ponader S., Chen Sh.-Sh., Buggy J.J. et al. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood 2012; 119(5): 1182–9.
  114. de Rooij M.F., Kuil A., Geest C.R. et al. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood 2012; 119(11): 2590–4.
  115. Mocsai A., Ruland J., Tybulewicz V.L. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat. Rev. Immunol. 2010; 10(6): 387–402.
  116. Clemens G.R., Schroeder R.E., Magness S.H. et al. Developmental toxicity associated with receptor tyrosine kinase Ret inhibition in reproductive toxicity testing. Birth Defects Res. Clin. Mol. Teratol. 2009; 85(2): 130–6.
  117. Braselmann S., Taylor V., Zhao H. et al. R406, an orally available spleen tyrosine kinase inhibitor blocks Fc receptor signaling and reduces immune complex-mediated inflammation. J. Pharmacol. Exp. Ther. 2006; 319(3): 998–1008.
  118. Gobessi S., Laurenti L., Longo P.G. et al. Inhibition of constitutive and BCR-induced Syk activation downregulates Mcl-1 and induces apoptosis in chronic lymphocytic leukemia B cells. Leukemia 2009; 23(4): 686–97.
  119. Quiroga M.P., Balakrishnan K., Kurtova A.V. et al. B-cell antigen receptor signaling enhances chronic lymphocytic leukemia cell migration and survival: specific targeting with a novel spleen tyrosine kinase inhibitor, R406. Blood 2009; 114(5): 1029–37.
  120. So L., Fruman D.A. PI3K signalling in B- and T-lymphocytes: new developments and therapeutic advances. Biochem. J. 2012; 442(3): 465–81.
  121. Okkenhaug K., Vanhaesebroeck B. PI3K in lymphocyte development, differentiation and activation. Nat. Rev. Immunol. 2003; 3(4): 317–30.
  122. Kloo B., Nagel D., Pfeifer M. et al. Critical role of PI3K signaling for NF-kappaB-dependent survival in a subset of activated B-cell-like diffuse large B-cell lymphoma cells. PNAS 2011; 108(1): 272–7.
  123. Rudelius M., Pittaluga S., Nishizuka S. et al. Constitutive activation of Akt contributes to the pathogenesis and survival of mantle cell lymphoma. Blood 2006; 108(5): 1668–76.
  124. Herman S.E., Gordon A.L., Wagner A.J. et al. Phosphatidylinositol 3-kinase-delta inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals. Blood 2010; 116(12): 2078–88.
  125. Hoellenriegel J., Meadows S.A., Sivina M. et al. The phosphoinositide 3’-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood 2011; 118(13): 3603–12.
  126. Lannutti B.J., Meadows S.A., Herman S.E.M. et al. CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood 2011; 117(2): 591–4.
  127. Mattmann M.E., Stoops S.L., Lindsley C.W. Inhibition of Akt with small molecules and biologics: historical perspective and current status of the patent landscape. Expert Opin. Ther. Pat. 2011; 21(9): 1309–38.

 

Influence of chemotherapy on reproductive system: methods of protection and preservation of ovarian function

M.V. Volochayeva1, R.G. Shmakov1, and Ye.A. Demina2

1 V.I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, RF Ministry of Health, Moscow, Russian Federation

2 N.N. Blokhin Russian Cancer Research Centre, RAMS, Moscow, Russian Federation


ABSTRACT

The recent studies showed the great importance of discussing the issue of protection and preservation of the ovarian function in cancer patients. Nowadays, when new effective methods of treatment are available, many cancer patients wish to realize their reproductive function. In this review, we discuss the methods of ovarian protection such as pharmacological protection and IVF program including a cryopreservation and transplantation of ovarian tissue or cryopreservation of oocytes and embryos.

Keywords: fertility preservation, cancer, cryopreservation tissue, oocytes, embryos

Read in PDF (RUS)pdficon


REFERENCES

  1. Демина Е.А., Махова Е.Е., Сусулева Н.А., Ильященко В.А. Возможности сохранения детородной функции у женщин с лимфомой Ходжкина. РМЖ 2005; 1: 26–8.[Demina Ye.A., Makhova Ye.Ye., Susuleva N.A., and Ilyashchenko V.A. Potentials for preservation of reproductive function in females with Hodgkin’s lymphoma. RMZh 2005: 1:26–8. (In Russ.)].
  2. Шмаков Р.Г. Репродуктивное здоровье женщин с онкогематологическими заболеваниями: Автореф. дис. … д-ра мед. наук. М., 2008.[Shmakov R.G. Reproduktivnoye zdorove zhenshchin s onkogematologicheskimi zabolevaniyami. Dokt. diss. (Reproductive health in women with hematological malignancies : Dr. med. sci. diss.). M., 2008.]
  3. Maltaris T., Seufert R., Fischl F. et al. The effect of cancer treatment on female fertility and strategies for preserving fertility. Eur. J. Obstet. Gynecol. Reprod. Biol. 2007; 130: 148–55.
  4. Blumenfeld Z. Ovarian rescue/protection from chemotherapeutic agents. J. Soc. Gynecol. Investig. 2001; 8: 60–4.
  5. Blumenfeld Z. Reservation of fertility and ovarian function and minimalization of chemotherapy associated gonadotoxicity and premature ovarian failure: the role of inhibin-A and -B as markers. Mol. Cell. Endocrinol. 2002; 187: 93–105.
  6. Minton S.E., Munster P.N. Chemotherapy-induced amenorrhea and fertility in women undergoing adjuvant treatment for breast cancer. Cancer Control 2002; 9: 466–72.
  7. Mrozek E., Shapiro C.L. Survivorship and complications of treatment in breast cancer. Clin. Adv. Hematol. Oncol. 2005; 3: 211–22.
  8. Friedman D.L., Constine L.S. Late effects of treatment for Hodgkin lymphoma. J. Natl. Compr. Cancer Network 2006; 4: 249–57.
  9. Haukvik U.K., Dieset I., Bjoro T. et al. Treatment-related premature ovarian failure as a long-term complication after Hodgkin’s lymphoma. Ann. Oncol. 2006; 17: 1428–33.
  10. Behringer K., Mueller H., Goergen H. et al. Gonadal function and fertility in survivors after Hodgkin lymphoma treatment within the German Hodgkin Study Group HD13 to HD15 trials. J. Clin. Oncol. 2013; 31: 231–9.
  11. De Bruin M.L., Huisbrink J., Hauptmann M. et al. Treatment-related risk factors for premature menopause following Hodgkin lymphoma. Blood 2008; 111: 101–8.
  12. Van der Kaaij M.A., van Echten-Arends J., Simons A.H. et al. Fertility preservation after chemotherapy for Hodgkin lymphoma. Hematol. Oncol. 2010; 28: 168–79.
  13. Fornier M.N., Modi S., Panageas K.S. et al. Incidence of chemotherapy induced, long-term amenorrhea in patients with breast carcinoma age 40 years and younger after adjuvant anthracycline and taxane. Cancer 2005; 104: 1575–9.
  14. Behringer K., Breuer K., Reineke T. et al. Secondary amenorrhea after Hodgkin’s lymphoma is influenced by age at treatment, stage of disease, chemotherapy regimen, and the use of oral contraceptives during therapy: a report from the German Hodgkin’s Lymphoma Study Group. J. Clin. Oncol. 2005; 23: 7555–64.
  15. Gerber B., Dieterich M., Muеller H. et al. Controversies in preservation of ovary function and fertility in patients with breast cancer. Breast Cancer Res. Treat. 2008; 108: 1–7.
  16. Okanami Y., Ito Y., Watanabe C. et al. Incidence of chemotherapy induced amenorrhea in premenopausal patients with breast cancer following adjuvant anthracycline and taxane. Breast Cancer 2011; 18: 182–8.
  17. Lee S.J., Schover L.R., Partridge A.H. et al. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J. Clin. Oncol. 2006; 24: 2917–31.
  18. Tham Y.L., Sexton K., Weiss H. et al. The rates of chemotherapy induced amenorrhea in patients treated with adjuvant doxorubicin and cyclophosphamide followed by a taxane. Am. J. Clin. Oncol. 2007; 30: 126–32.
  19. Sklar C.A., Mertens A.C., Mitby P. et al. Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study. J. Natl. Cancer Inst. 2006; 98: 890–6.
  20. Dillon K.E., Sammel M.D., Prewitt M. et al. Pretreatment antimullerian hormone levels determine rate of post therapy ovarian reserve recovery: acute changes in ovarian reserve during and after chemotherapy. Fertil. Steril. 2013; 99: 477–83.
  21. Wallace W.H., Thomson A.B., Saran F. et al. Predicting age of ovarian failure after radiation to a field that includes the ovaries. Int. J. Radiat. Oncol. Biol. Phys. 2005; 62: 738–44.
  22. Knauff E.A., Eijkemans M.J., Lambalk C.B. et al. Anti-Mullerian hormone, inhibin B, and antral follicle count in young women with ovarian failure. J. Clin. Endocrinol. Metab. 2009; 94: 786–92.
  23. Iwase A., Sugita A., Hirokawa W. et al. Anti-Mullerian hormone as a marker of ovarian reserve following chemotherapy in patients with gestational trophoblastic neoplasia. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013; 167: 194–8.
  24. Munster P.N., Moore A.P., Ismail-Khan R. et al. Randomized trial using gonadotropin-releasing hormone agonist triptorelin for the preservation of ovarian function during (neo)adjuvant chemotherapy for breast cancer. J. Clin. Oncol. 2012; 30: 533–8.
  25. Recchia F., Saggio G., Amiconi G. et al. Gonadotropin-releasing hormone analogues added to adjuvant chemotherapy protect ovarian function and improve clinical outcomes in young women with early breast carcinoma. Cancer 2006; 106: 514–23.
  26. Han S.S., Kim Y.H., Lee S.H. et al. Underuse of ovarian transposition in reproductive-aged cancer patients treated by primary or adjuvant pelvic irradiation. J. Obstet. Gynaecol. Res. 2011; 37: 825–9.
  27. Pentheroudakis G., Pavlidis N., Castiglione M. Cancer, fertility and pregnancy: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann. Oncol. 2009; 20: 178–81 (Suppl. 4).
  28. Donnez J., Dolmans M.M. Cryopreservation and transplantation of ovarian tissue. Clin. Obstet. Gynecol. 2010; 53: 787–96.
  29. Bedaiwy M.A., Abou-Setta A.M., Desai N. et al. Gonadotropin-releasing hormone analog cotreatment for preservation of ovarian function during gonadotoxic chemotherapy: A systematic review and meta-analysis. Fertil. Steril. 2011; 95: 906–14, e1–4.
  30. Blumenfeld Z., von Wolff M. GnRH analogues and oral contraceptives for fertility preservation in women during chemotherapy. Hum. Reprod. Update 2008; 14: 543–52.
  31. Демина Е.А., Перилова Е.Е., Шмаков Р.Г. Использование комбинированных пероральных контрацептивов для профилактики повреждения функции яичников у больных лимфомой Ходжкина. М., 2004: 1352–4. [Demina Ye.A., Perilova Ye.Ye., Shmakov R.G. Ispolzovaniye kombinirovannykh peroralnykh kontratseptivov dlya profilaktiki povrezhdeniya funktsii yaichnikov u bolnykh limfomoy Khodzhkina (Use of oral contraceptives for prevention of ovarian function damage in patients with Hodgkin’s lymphoma). M., 2004: 1352–4.]
  32. Blumenfeld Z., Avivi I., Linn S. et al. Prevention of irreversible chemotherapy-induced ovarian damage in young women with lymphoma by a gonadotrophin-releasing hormone agonist in parallel to chemotherapy. Hum. Reprod. 1996; 11: 1620–6.
  33. Badawy A., Elnashar A., El-Ashry M. et al. Gonadotropin-releasing hormone agonists for prevention of chemotherapy-induced ovarian damage: prospective randomized study. Fertil. Steril. 2009; 91: 694–7.
  34. Sverrisdottir A., Nystedt M., Johansson H. et al. Adjuvant goserelin and ovarian preservation in chemotherapy treated patients with early breast cancer: results from a randomized trial. Breast Cancer Res. Treat. 2009; 117: 561–7.
  35. Clowse M.E., Behera M.A., Anders C.K. et al. Ovarian preservation by GnRH agonists during chemotherapy: a meta-analysis. J. Womens Health 2009; 18: 311–9.
  36. Del Mastro L., Boni L., Michelotti A. et al. Effect of the gonadotropinreleasing hormone analogue triptorelin on the occurrence of chemotherapyinduced early menopause in premenopausal women with breast cancer: a randomized trial. JAMA 2011; 306: 269–76.
  37. Chen H., Li J., Cui T. et al. Adjuvant gonadotropin-releasing hormone analogues for the prevention of chemotherapy induced premature ovarian failure in premenopausal women. Cochrane Database Syst Rev 2011; 9: CD008018.
  38. Wong M., O’Neill S., Walsh G. et al. Goserelin with chemotherapy to preserve ovarian function in pre-menopausal women with early breast cancer: menstruation and pregnancy outcomes. Ann. Oncol. 2013; 24: 133–8.
  39. Yang B., Shi W., Yang J. et al. Concurrent treatment with gonadotropinreleasing hormone agonists for chemotherapy-induced ovarian damage in premenopausal women with breast cancer: A meta-analysis of randomized controlled trials. Breast 2013; 22: 150–7.
  40. Leonard R.C., Adamson D., Anderson R. et al. The OPTION trial of adjuvant ovarian protection by goserelin in adjuvant chemotherapy for early breast cancer. J. Clin. Oncol. 2010; 28: Abstract 590.
  41. Gerber B., von Minckwitz G., Stehle H. et al. Effect of luteinizing hormonereleasing hormone agonist on ovarian function after modern adjuvant breast cancer chemotherapy: the GBG 37 ZORO study. J. Clin. Oncol. 2011; 29: 2334–41.
  42. Elgindy E.A., El-Haieg D.O., Khorshid O.M. et al. Gonadatrophin suppression to prevent chemotherapy-induced ovarian damage: a randomized controlled trial. Obstet. Gynecol. 2013; 121: 78–86.
  43. American Society for Reproductive Medicine. http://www.asrm.org
  44. Azim A.A., Costantini-Ferrando M., Oktay K. Safety of fertility preservation by ovarian stimulation with letrozole and gonadotropins in patients with breast cancer: A prospective controlled study. J. Clin. Oncol. 2008; 26: 2630–5.
  45. Oktay K., Buyuk E., Libertella N. et al. Fertility preservation in breast cancer patients: A prospective controlled comparison of ovarian stimulation with tamoxifen and letrozole for embryo cryopreservation. J. Clin. Oncol. 2005; 23: 4347–53.
  46. Lee S., Oktay K. Does higher starting dose of FSH stimulation with letrozole improve fertility preservation outcomes in women with breast cancer? Fertil. Steril. 2012; 98: 961.e1–4.e1.
  47. Sonmezer M., Turkcuoglu I., Coskun U., Oktay K. Random-start controlled ovarian hyperstimulation for emergency fertility preservation in letrozole cycles. Fertil. Steril. 2011; 95: 2125.e9–11.
  48. Isachenko V., Isachenko E., Keck G. et al. First live birth in Germany after re-transplantation of cryopreserved ovarian tissue: Original device for initiation of ice formation. Clin. Lab. 2012; 58: 933–8.
  49. Bacigalupo A., Ballen K., Rizzo D. et al. Defining the intensity of conditioning regimens: Working definitions. Biol. Blood Marrow Transplant. 2009; 15: 1628–33.
  50. Borini A., Bianchi V. Cryopreservation of mature and immature oocytes. Clin. Obstet. Gynecol. 2010; 53: 763–74.
  51. Huang J.Y., Chian R.C., Gilbert L. et al. Retrieval of immature oocytes from unstimulated ovaries followed by in vitro maturation and vitrification: A novel strategy of fertility preservation for breast cancer patients. Am. J. Surg. 2010; 200: 177–83.
  52. Rudick B., Opper N., Paulson R. et al. The status of oocyte cryopreservation in the United States. Fertil. Steril. 2010; 94: 2642–6.
  53. Demeestere I., Simon P., Emiliani S., Delbaere A., Englert Y. Orthotopic and heterotopic ovarian tissue transplantation. Hum. Reprod. Update 2009; 15: 649–65.
  54. Demeestere I., Moffa F., Peccatori F., Poirot C., Shalom-Paz E. Multiple approaches for individualized fertility protective therapy in cancer patients. Obstet. Gynecol. Int. 2012: 495142 (Medline Abstract).
  55. Oktay K., Cil A.P., Bang H. Efficiency of oocyte cryopreservation: A metaanalysis. Fertil. Steril. 2006; 86: 70–80.
  56. Donnez J., Squifflet J., Jadoul P. et al. Pregnancy and live birth after autotransplantation of frozen-thawed ovarian tissue in a patient with metastatic disease undergoing chemotherapy and hematopoietic stem cell transplantation. Fertil. Steril. 2011; 95: 1787.e1–4.
  57. Kim M.K., Lee D.R., Han J.E. et al. Live birth with vitrified-warmed oocytes of a chronic myeloid leukemia patient nine years after allogenic bone marrow transplantation. J. Assist. Reprod. Genet. 2011; 28: 1167–70.
  58. Dittrich R., Lotz L., Keck G. et al. Live birth after ovarian tissue autotransplantation following overnight transportation before cryopreservation. Fertil. Steril. 2012; 97: 387–90.
  59. Andersen C.Y., Silber S.J., Berghold S.H. et al. Long-term duration of function of ovarian tissue transplants: Case reports. Reprod. Biomed. Online 2012; 25: 128–32.
  60. Oktay K., Rodriguez-Wallberg K.A. Four spontaneous pregnancies and three live births following subcutaneous transplantation of frozen banked ovarian tissue: What is the explanation? Fertil. Steril. 2011; 95: 804.e7–10.
  61. Meirow D., Levron J., Eldar-Geva T. et al. Pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy. N. Engl. J. Med. 2005; 353: 318–21.
  62. Anderson R.A., Wallace W.H., Baird D.T. Ovarian cryopreservation for fertility preservation: Indications and outcomes. Reproduction 2008; 136: 681–9.
  63. Reebals J.F., Brown R., Buckner E.B. Nurse practice issues regarding sperm banking in adolescent male cancer patients. J. Pediatr. Oncol. Nurs. 2006; 23: 182–8.
  64. Keros V., Hultenby K., Borgstrumlom B. et al. Methods of cryopreservation of testicular tissue with viable spermatogonia in pre-pubertal boys undergoing gonadotoxic cancer treatment. Hum. Reprod. 2007; 22: 1384–95.
  65. Jadoul P., Dolmans M.M., Donnez J. Fertility preservation in girls during childhood: Is it feasible, efficient and safe and to whom should it be proposed? Hum. Reprod. Update 2010; 16: 617–30.
  66. Cvancarova M., Samuelsen S.O., Magelssen H. et al. Reproduction rates after cancer treatment: Experience from the Norwegian radium hospital. J. Clin. Oncol. 2009; 27: 334–43.
  67. Nieman C.L., Kinahan K.E., Yount S.E. et al. Fertility preservation and adolescent cancer patients: Lessons from adult survivors of childhood cancer and their parents. Cancer Treat. Res. 2007; 138: 201–17

Autophagy: cell death or survival strategy?

O.V. Kovaleva, M.S. Shitova, and I.B. Zborovskaya

N.N. Blokhin Russian Cancer Research Center, RAMS, Moscow, Russian Federation


ABSTRACT

The interaction between proliferation, differentiation, and programmed cell death is an integral part of the life-sustaining activity of multicellular organisms. The imbalance between these processes underlies the development of many human diseases. Understanding molecular mechanisms of these processes is important for identifying new diagnostic and therapeutic targets. During the last decade, the autophagy processed and its role in the cell life and death became a subject of great scientific interest. Autophagy represents a mechanism of intracellular substance utilization. Autophagy is a process that occurs in all cells under normal conditions. But under certain circumstances, this process can result in the cell death. Impaired autophagy significantly contributes into the development of some diseases (cancer, neurodegenerative and cardiovascular disorders etc.).


Keywords: apoptosis, autophagy, carcinogenesis

Read in PDF (RUS)pdficon


REFERENCES

  1. Galluzzi L., Vitale I., Abrams J.M. et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012; 19(1): 107–20.
  2. Kerr J.F., Wyllie A.H., Currie A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972; 26: 239–57.
  3. Wong R.S. Apoptosis in cancer: from pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011; 26: 30–87.
  4. Hu Y., Benedict M.A., Ding L., Nunez G. Role of cytochrome c and dATP/ ATP hydrolysis in Apaf-1-mediated caspase-9 activation and apoptosis. EMBO J. 1999; 18: 3586–95.
  5. Saelens X., Festjens N., Vande Walle L. et al. Toxic proteins released from mitochondria in cell death. Oncogene 2004; 23(16): 2861–74.
  6. Altieri D.C. Surviving in apoptosis control and cell cycle regulation in cancer. Prog. Cell Cycle Res. 2003; 5: 447–52.
  7. Tamm I., Wang Y., Sausville E. et al. IAP-family protein surviving inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res. 1998; 58(23): 5315–20.
  8. Padanilam B.J. Cell death induced by acute renal injury: a perspective on the contributions of apoptosis and necrosis. Am. J. Physiol. Renal. Physiol. 2003; 284(4): F608–27.
  9. Hoste E., Kemperman P., Devos M. et al. Caspase-14 is required for filaggrin degradation to natural moisturizing factors in the skin. J. Invest. Dermatol. 2011; 131(11): 2233–41.
  10. Levine B., Klionsky D.J. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev. Cell 2004; 6(4): 463–77.
  11. Okada H., Mak T.W. Pathways of apoptotic and non-apoptotic death in tumor cells. Nat. Rev. Cancer 2004; 4(8): 592–603.
  12. Kaminskyy V., Zhivotovsky B. Proteases in autophagy. Biochimica er Biophysica Acta. 2012; 1824: 44–50.
  13. Mijaljica D., Prescott M., Devenish R.J. Microautophagy in mammalian cells: Revisiting a 40-year-old conundrum. Autophagy 2011; 7: 673–82.
  14. Klionsky DJ., Codogno P., Cuervo A.M. et al. A comprehensive glossary of autophagy-related molecules and processes. Autophagy 2010; 6(4): 438–48.
  15. Massey A.C., Zhang C., Cuervo A.M. Chaperone-mediated autophagy in aging and disease. Curr. Top. Dev. Biol. 2006; 73: 205–35.
  16. Kimmelman A.C. The dynamic nature of autophagy in cancer. Genes Dev. 2011; 25(19): 1999–2010.
  17. Reggiori F., Tucker K.A., Stromhaug P.E., Klionsky D.J. The Atg1-Atg13 complex regulates Atg9 and Atg23 retrieval transport from the pre-autophagosomal structure. Dev. Cell 2004; 6(1): 79–90.
  18. Wang C.W., Klionsky D.J. The molecular mechanism of autophagy. Mol. Med. 2003; 9: 65–76.
  19. Teter S.A., Klionsky D.J. Transport of proteins to the yeast vacuole: autophagy, cytoplasm-to-vacuole targeting, and role of the vacuole in degradation. Semin. Cell Dev. Biol. 2000; 11(3): 173–9.
  20. Chen N., Debnath J. Autophagy and Tumorigenesis. FEBS Lett. 2010; 584(7): 1427–35.
  21. Guertin D.A., Sabatini D.M. Defining the role of mTOR in cancer. Cancer Cell 2007; 12: 9–22.
  22. He C., Klionsky D.J. Regulation Mechanisms and Signaling Pathways of Autophagy. Annu. Rev. Genet. 2009; 43: 67–93.
  23. Zhou H., Huang S. The complexes of mammalian target of rapamycin. Curr. Protein Pept. Sci. 2010; 11: 409–24.
  24. Teter T., Hall M.N. TOR, a central controller of cell growth. Cell 2000; 103(2): 253–62.
  25. Tee A.R., Manning B.D., Roux P.P., Cantley L.C., Blenis J. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr. Biol. 2003; 13: 1259–68.
  26. Shackelford D.B., Shaw R.J. The LKB1-AMPK pathway: metabolism and growth control in tumor suppression. Nat. Rev. Cancer 2009; 9: 563–75.
  27. Liang J., Shao SH., Xu Z.X. et al. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat. Cell Biol. 2007; 9: 218–24.
  28. Feng Z., Hu W., Stanchina E. et al. The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res. 2007; 67: 3043–53.
  29. Mortensen M., Watson A.S., Simon A.K. Lack of autophagy in the hematopoietic system leads to loss of hematopoietic stem cell function and dysregulated myeloid proliferation. Autophagy 2011; 7(9): 1069–70.
  30. Mortensen M., Soilleux E.J., Djordjevic G. et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 2011; 208: 455–67.
  31. Pua H.H., Komatsu M., He Y.W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J. Immunol. 2009; 182: 4046–55.
  32. Pua H.H., Dzhagalov I., Chuck M. et al. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 2007; 204: 25–31.
  33. Pua H.H., He Y.W. Maintaining T lymphocyte homeostasis: another duty of autophagy. Autophagy 2007; 3: 266–7.
  34. Miller B.C., Zhao Z., Stephenson L.M. et al. The autophagy gene Atg5 plays an essential role in B lymphocyte development. Autophagy 2008; 4: 309–14.
  35. Novak I., Kirkin V., McEwan D.G. et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010; 11(1): 45–51.
  36. Kundu M., Lindsten T., Yang C.Y. et al. Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood 2008; 112: 1493–502.
  37. Raslova H., Kauffmann A., Sekkai D. et al. Interrelation between polyploidization and megakaryocyte differentiation: a gene profiling approach. Blood. 2007; 109: 3225–34.
  38. Delgado M.A., Elmaoued R.A., Davis A.S., Kyei G., Deretic V. Toll-like receptors control autophagy. EMBO J. 2008; 27: 1110–21.
  39. Shi C.S., Kehrl J.H. MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages. J. Biol. Chem. 2008; 283: 33175–82.
  40. Xu Y., Jagannath C., Liu X.D. et al. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity. 2007; 27: 135–44.
  41. Канцерогенез. Под ред. Д.Г. Заридзе. М.: Медицина, 2004. [Kantserogenez. Pod red. D.G. Zaridze (Carcinogenesis. Ed. by: D.G. Zaridze). M.: Meditsina, 2004.]
  42. Liang X.H., Jackson S., Seaman M. et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999; 402: 672–6.
  43. Yue Z., Jin S., Yang C., Levine A.J., Heintz N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. U S A 2003; 100: 15077–82.
  44. Qu X., Yu J., Bhagat G. et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Invest. 2003; 112: 1809–20.
  45. Karantza-Wadsworth V., Patel S., Kravchuk O. et al. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 2007; 21: 1621–35.
  46. Meek D.W. Tumor suppression by p53: a role for the DNA damage response? Nat. Rev. Cancer 2009; 9: 714–23.
  47. Degenhardt K., Mathew R., Beaudoin B. et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006; 10: 51–64.
  48. DeNardo D., Johansson M., Coussens L. Immune cells as mediators of solid tumor metastasis. Cancer Metast. Rev. 2008; 27: 11–8.
  49. DeNardo D.G., Barreto J.B., Andreu P. et al. CD4+ T Cells Regulate Pulmonary Metastasis of Mammary Carcinomas by Enhancing Protumor Properties of Macrophages. Cancer Cell 2009; 16: 91–102.
  50. Bingle L., Brown N.J., Lewis C.E. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J. Pathol. 2002; 196: 254–65.
  51. Young A.R., Narita M., Ferreira M. et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009; 23: 798–803.
  52. Petiot A., Ogier-Denis E., Blommaart E.F., Meijer AJ., Codogno P. Distinct classes of phosphatidylinositol 3¢-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J. Biol. Chem. 2000; 275: 992–8.
  53. Luiken J.J., Aerts J.M., Meijer A.J. The role of the intralysosomal pH in the control of autophagic proteolytic flux in rat hepatocytes. Eur. J. Biochem. 1996; 235: 564–73.
  54. Yamamoto A., Tagawa Y., Yoshimori T. et al. Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E cells. Cell Struct. Funct. 1998; 23: 33–42.
  55. Ito H., Daido S., Kanzawa T., Kondo S., Kondo Y. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int. J. Oncol. 2005; 26: 1401–10.
  56. Lomonaco S.L., Finniss S., Xiang C. et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int. J. Cancer 2009; 125: 717–22.
  57. Shingu T., Fujiwara K., Bogler O. et al. Stage-specific effect of inhibition of autophagy on chemotherapy-induced cytotoxicity. Autophagy 2009; 5: 537–9.
  58. Vazquez-Martin A., Oliveras-Ferraros C., Menendez J.A. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. PLoS One 2009; 4: e6251.
  59. Abedin M.J., Wang D., McDonnell M.A., Lehmann U., Kelekar A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2007; 14: 500–10.
  60. 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.
  61. Bellodi C., Lidonnici M.R., Hamilton A. et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosomepositive cells, including primary CML stem cells. J. Clin. Invest. 2009; 119: 1109–23.
  62. Carew J.S., Nawrocki S.T., Kahue C.N. et al. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 2007; 10: 313–22.
  63. Ertmer A., Huber V., Gilch S. et al. The anticancer drug imatinib induces cellular autophagy. Leukemia 2007; 21: 936–42.
  64. Kamitsuji Y., Kuroda J., Kimura S. et al. The Bcr-Abl kinase inhibitor INNO-406 induces autophagy and different modes of cell death execution in Bcr-Abl-positive leukemias. Cell Death Differ. 2008; 15: 1712–22.
  65. Goussetis D.J., Altman J.K., Glaser H. et al. Autophagy is a critical mechanism for the induction of the antileukemic effects of arsenic trioxide. J. Biol. Chem. 2010; 285: 29989–97.
  66. Qian W., Liu J., Jin J. et al. Arsenic trioxide induces not only apoptosis but also autophagic cell death in leukemia cell lines via up-regulation of Beclin-1. Leuk. Res. 2007; 31: 329–39.
  67. Charoensuk V., Gati WP., Weinfeld M. et al. Differential cytotoxic effects of arsenic compounds in human acute promyelocytic leukemia cells. Toxicol. Appl. Pharmacol. 2009; 239: 64–70.
  68. Chiarini F., Grimaldi C., Ricci F. et al. Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVPBEZ235 against T-cell acute lymphoblastic leukemia. Cancer Res. 2010; 70: 8097–107.
  69. Crazzolara R., Bradstock K.F., Bendall L.J. RAD001 (everolimus) induces autophagy in acute lymphoblastic leukemia. Autophagy 2009; 5: 727–8.
  70. Crazzolara R., Cisterne A., Thien M. et al. Potentiating effects of RAD001 (everolimus) on vincristine therapy in childhood acute lymphoblastic leukemia. Blood 2009; 113: 3297–306.
  71. Puissant A., Auberger P. AMPK- and p62/SQSTM1-dependent autophagy mediate resveratrol-induced cell death in chronic myelogenous leukemia. Autophagy 2010; 6(5): 655–7.
  72. Puissant A., Robert G., Fenouille N. et al. Resveratrol promotes autophagic cell death in chronic myelogenous leukemia cells via JNK mediated p62/SQSTM1 expression and AMPK activation. Cancer Res. 2010; 70: 1042–52.
  73. Kroemer G., Levine B. Autophagic cell death: the story of a misnomer. Nat. Rev. Mol. Cell Biol. 2008; 9: 1004–10.
  74. Turcotte S., Chan D.A., Sutphin P.D. et al. A molecule targeting VHLdeficient renal cell carcinoma that induces autophagy. Cancer Cell. 2008; 14: 90–102.
  75. Kanzawa T., Germano I.M., Komata T. et al. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ. 2004; 11: 448–57.

Enzymes in oncohematology: relevant directions of experimental studies and prospects of clinical use

V.S. Pokrovskiy and H.M. Treshchalina

N.N. Blokhin Russian Cancer Research Center of RAMS, Moscow, Russian Federation


ABSTRACT

Recently, the field of development of the novel enzyme-based drugs showed remarkable advances. In addition to L-asparaginase which have been already used in oncohematology for more than 30 years, the two new enzymes, L-arginine deiminase and ranpirnase, underwent the several phases of clinical trials. Anticancer activity in vivo at the preclinical stage was shown for a number of enzymes: L-methionine-gamma-lyase, L-lysine-alpha-oxydase, and binase. This review discusses the enzymes which possess anticancer activity and prospects for their use in oncohematology.


Keywords: anticancer enzymes, L-asparaginase, L-methionine gamma-lyase, L-lysine alpha-oxydase, L-arginine deiminase, L-phenylalanine ammonia-lyase, ranpirnase.

Read in PDF (RUS)pdficon


REFERENCES

  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.

Role of tyrosine-kinase inhibitor selectivity in development of adverse effects during treatment of chronic myeloid leukemia

A.A. Zeifman1,2, E.Yu. Chelysheva3, A.G. Tukrina3, and G.G. Chilov1,2

1 N.D. Zelinsky Institute of Organic Chemistry, RAS, Moscow, Russian Federation

2 Fusion Pharma LLC, Moscow, Russian Federation

3 Hematology Research Center, RF MH, Moscow, Russian Federation


ABSTRACT

This review focuses on association between the selectivity of Bcr-Abl kinase inhibitors and the spectrum of their adverse effects during treatment of patients with chronic myeloid leukemia. The data on the structure and natural biochemical functions of the well-known adverse targets for inhibitors of Bcr-Abl kinases, including BRAF, FMS, EGFR, PDGFR, PYK2, TIE2, and VEGFR1/2/3 are summarized, and the potential association between their off-target inhibition and adverse effects of tyrosine-kinase inhibitors is suggested.


Keywords: chronic myeloid leukemia, tyrosine-kinase inhibitors, selectivity, imatinib, nilotinib, dasatinib, ponatinib, PF-114, BRAF, FMS, EGFR, PDGFR, PYK2, TIE2, VEGFR1/2/3.

Read in PDF (RUS)pdficon


REFERENCES

  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.)].

Past and present of radiotherapy in management of malignancies

O.P. Trofimova, S.I. Tkachev, and T.V. Yuryeva

N.N. Blokhin Russian Cancer Research Center, RAMS, Moscow, Russian Federation


ABSTRACT

The article describes the stages of emergence and development of radiotherapy in management of malignancies of various locations, covering the period from the discovery of X-rays to the use of 60Co artificial isotope and development of the linear electron accelerator. Currently, radiotherapy is required for 70 % of cancer patients at some stage of tumor treatment. Improvements in radiotherapy technologies and development of new irradiation techniques made possible to solve the main task of radiotherapy, i.e. tumor destruction with minimal radiation exposure to surrounding healthy tissues, at the time being. This paper describes the radiotherapy techniques and explains in detail the differences between conventional and conformal radiotherapy. Also, preparation of patients for radiotherapy and the features of various conformal radiotherapy technologies are included.


Keywords: irradiation, radiotherapy, preparation for radiotherapy, linear electron accelerator.

Read in PDF (RUS)pdficon


References

  1. Гернек Ф. Пионеры атомного века. Великие исследователи от Максвелла до Гейзенберга. М.: Прогресс, 1974.  [Gernek F. Pionery atomnogo veka. Velikiye issledovateli ot Maksvella do Geyzenberga (Pioneers of atomic age. Great explorers from Maxwell to Heisenberg). M.: Progress, 1974.]
  2. Труфанов Г.Е., Асатурян М.А., Жаринов Г.М. Лучевая терапия (учебник для вузов). М.: ГЭОТАР-Медиа, 2007. Т. 2. [Trufanov G.E., Asaturyan M.A., Zharinov G.M. Luchevaya terapiya (uchebnik dlya vuzov) (Radiotherapy (manual for medical institutes)). M.: GEOTAR-Media, 2007. Vol. 2.]
  3. Grubbe E.H. Priority in the therapeutic use of X-Rays. Radiology 1933: 156–62.
  4. Hoffman R., Furie B., McGlave Ph. et al. Hematology: Basic Principles and Practice, 5th edn. Churchill Livingstone, 2008: 2560.
  5. Козлова А.В. Лучевая терапия злокачественных опухолей. М., 1971. [Kozlova A.V. Luchevaya terapiya zlokachestvennykh opukholey (Radiotherapy for malignant tumors). M., 1971.]
  6. Chao А., Tigner M. Handbook of Accelerator Physics and Engineering, 1999.
  7. Ткачев С.И., Нечушкин М.И., Юрьева Т.В. Современные возможности лучевой терапии злокачественных опухолей. Вестн. РАМН 2011; 12: 34–40.  [Tkachev S.I., Nechushkin M.I., Yuryeva T.V. Sovremennyye vozmozhnosti luchevoy terapii zlokachestvennykh opukholey (Current potentialities of radiotherapy for malignant tumors. In: Bull. of RAMS). Vestn. RAMN 2011; 12: 34–40.]
  8. Юрьева Т.В., Ратнер Т.Г., Сахаровская В.Г. Оценка качества рентге- новского симулятора с функцией компьютерной томографии. Сб. мат-лов III Евразийского конгресса по медицинской физике и инженерии «Меди- цинская физика — 2010». М., 2010; 2: 343–5. [Yuryeva T.V., Ratner T.G., Sakharovskaya V.G. Otsenka kachestva rentgenovskogo simulyatora s funktsiyey kompyuternoy tomografii. Sb. mat-lov III Yevraziyskogo kongressa po meditsinskoy fizike i inzhenerii «Meditsinskaya fizika — 2010» (Assessment of quality of X-ray simulator with computed tomography function. Coll. of materials of 3rd Eurasian congress on medical physics and engineering “Medical physics — 2010”). M., 2010; 2: 343–5.]
  9. Favier O., Heutte N., Stamatoullas-Bastard A. Survival after Hodgkin lymphoma: causes of death and excess mortality in patients treated in 8 consecutive trials. Cancer 2009; 115(8): 1680–91.
  10. Переслегин И.А., Саркисян Ю.Х. Клиническая радиология. М., 1973. [Pereslegin I.A., Sarkisyan Yu.Kh. Klinicheskaya radiologiya (Clinical radiology). M., 1973.]
  11. Артемова Н.А., Минайло И.И., Страх А.Г. Объемное планирование лучевой терапии. Мед. новости 2005; 11: 5–10. [Artemova N.A., Minaylo I.I., Strakh A.G. Obyemnoye planirovaniye luchevoy terapii (Three dimensional planning in radiotherapy. In: Med. news). Med. novosti 2005; 11: 5–10.]
  12. Артемова Н.А., Минайло И.И., Страх А.Г. Предлучевая подготовка с использованием объемного планирования. В сб.: Контроль качества лучевой терапии и лучевой диагностики. Минск, 2009: 261–70. [Artemova N.A., Minaylo I.I., Strakh A.G. Predluchevaya podgotovka s ispolzovaniyem obyemnogo planirovaniya. V sb.: Kontrol kachestva luchevoy terapii i luchevoy diagnostiki (Pre-irradiation preparation using three dimensional planning. : Quality control in radiotherapy and radiologic diagnosis). Minsk, 2009: 261–70.]
  13. ICRU Report 50: Prescribing, Recording and Reporting Photon Beam Therapy. Bethesda: International Commission on Radiation Units and Measurements, 1993.
  14. ICRU Report 62: Prescribing, Recording and Reporting Photon Beam Therapy. Bethesda: International Commission on Radiation Units and Measurements, 1999.
  15. Leeuwen F.E., Klokman W.J., Stovall M. Roles of radiation dose, chemotherapy, and hormonal factors in breast cancer following Hodgkin’s disease. J. Natl. Cancer Inst. 2003; 95(13): 971–80.
  16. Markova J., Kobe С., Skopalova М. FDG–PET for assessment of early treatment response after four cycles of chemotherapy in patients with advancedstage Hodgkin’s lymphoma has a high negative predictive value. Ann. Oncol. 2009; 20(7): 1270–4.