Pathological Metabolism of Methionine in Malignant Cells Is a Potential Target for the Antitumor Therapy

VS Pokrovskii1, DZh Davydov1, NV Anufrieva2, DD Zhdanov3, EM Treshchalina1, TV Demidkina2, EA Morozova2

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

2 VA Engelhardt Institute of Molecular Biology, 32 Vavilova str., Moscow, Russian Federation, 119991

3 VN Orekhovich Institute of Biomedical Chemistry, 10 bld. 8 Pogodinskaya str., Moscow, Russian Federation, 119121

For correspondence: Vadim Sergeevich Pokrovskii, DSci, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: 8(499)324-14-09; e-mail: vadimpokrovsky@yandex.ru

For citation: Pokrovskii VS, Davydov DZh, Anufrieva NV, et al. Pathological Metabolism of Methionine in Malignant Cells Is a Potential Target for the Antitumor Therapy. Clinical oncohematology. 2017;10(3):324–32 (In Russ).

DOI: 10.21320/2500-2139-2017-10-3-324-332


ABSTRACT

This review presents the characteristics of the cellular metabolism of methionine, as well as known data on the mechanisms of the development of methionine dependence in malignant cells. The possibilities of using a non-methionine diet for the control of the tumor growth in patients with various forms of cancer are considered. The newest information about methionine-γ-lyase, an enzyme providing elimination of methionine from plasma, is grouped and summarised. Its role as a potential antitumor enzyme is disclosed. Data on methionine-γ-lyase producers, activity of this enzyme, obtained from various sources, and information on tumor models and cell cultures, showing methionine dependence are summarised.

Keywords: methionine-γ-liase, methionine, methionine dependency, cancer cells, cancer, anticancer enzymes, antitumor therapy.

Received: December 16, 2016

Accepted: March 6, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Thomas D, Surdin-Kerjan Y. Metabolism of sulfur amino acids in Saccharomyces cerevisiae. Microbiol Mol Biol Rev. 1997;61(4):503–32.
  2. Ravanel S, Gaki B, Job D, Douce R. The specific features of methionine biosynthesis and metabolism in plants. Proc Natl Acad Sci USA. 1998;95(13):7805–12. doi: 10.1073/pnas.95.13.7805.
  3. Sekowska A, Kung H, Danchin A, et al. Sulfur metabolism in Escherichia coli and related bacteria: facts and fiction. J Mol Microbiol Biotechnol. 2000;2(2):145–77.
  4. Guedes RL, Prosdocimi F, Fernandes GR, et al. Amino acids biosynthesis and nitrogen assimilation pathways: A great genomic deletion during eukaryotes. BMC Genom. 2011;12(Suppl 4):S2. doi: 10.1186/1471-2164-12-S4-S2.
  5. Satishchandran C, Taylor JC, Markham GD, et al. Novel Escherichia coli K-12 mutants impaired in S-adenosylmethionine synthesis. J Bacteriol. 1990;172(8):4489–96. doi: 10.1128/jb.172.8.4489-4496.1990.
  6. Zingg JM. Genetic and epigenetic aspects of DNA methylation on genome expression, evolution, mutation and carcinogenesis. Carcinogenesis. 1997;18(5):869–82. doi: 10.1093/carcin/18.5.869.
  7. Krasinskas A, Bartlett DL, Cieply K, et al. CDKN2A and MTAP deletions in peritoneal mesotheliomas are correlated with loss of p16 protein expression and poor survival. Mod Pathol. 2010;23(4):531–8. doi: 10.1038/modpathol.2009.186.
  8. Roje S. S-Adenosyl-L-methionine: Beyond the universal methyl group donor. Phytochemistry 2006;67(15):1686-1698. doi: 10.1016/j.phytochem.2006.04.019.
  9. Anderson ME. Glutatione: an overview of biosynthesis and modulation. Chem Biol Interact. 1998;111(112):1–14. doi: 10.1016/s0009-2797(97)00146-4.
  10. Thomas T, Tomas TJ. Polyamines in cell growth and cell death: molecular mechanisms and therapeutic applications. Cell Mol Life Sci. 2001;58(2):244–58. doi: 10.1007/PL00000852.
  11. Pirkov I, Norbeck J, Gustafsson L, et al. A complete inventory of all enzymes in the eukaryotic methionine salvage pathway. FEBS J. 2008;275(16):4111–20. doi: 10.1111/j.1742-4658.2008.06552.x.
  12. Quash G, Roch AM, Chantepie J, et al. Methional derived from 4-methylthio-2-oxobutanoate is a cellular mediator of apoptosis in BAF3 lymphoid cells. Biochem J. 1995;305(3):1017–25. doi: 10.1042/bj3051017.
  13. Bassila C, Ghemrawi R, Flayac J, et al. Methionine synthase and methionine synthase reductase interact with MMACHC and with MMADHC. Biochim Biophys Acta. 2017;1863(1):103–12. doi: 10.1016/j.bbadis.2016.10.016.
  14. Морозова Е.А., Куликова В.В., Яшин Д.В. и др. Кинетические характеристики и цитотоксическая активность рекомбинантных препаратов метионин–гамма-лиазы Clostridium tetani, Clostridium sporogenes, Porphyromonas gingivalis и Citrobacter freundii. Acta Naturae. 2013;5:54–60.
    [Morozova EA, Kulikova VV, Yashin DV, et al. Kinetic parameters and cytotoxic activity of recombinant methionine γ-lyase from Clostridium tetani, Clostridium sporogenes, Porphyromonas gingivalis and Citrobacter freundii. Acta Naturae. 2013;5:54–60. (In Russ)]
  15. Cavuoto P, Fenech MF. A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension. Cancer Treat Rev. 2012;38(6):726–36. doi: 10.1016/j.ctrv.2012.01.004.
  16. Sugimura T, Birnbaum SM, Winitz M, et al. Quantitative nutritional studies with water-soluble, chemically defined diets. VIII. The forced feeding of diets each lacking in one essential amino acid. Arch Biochem Bioophys. 1959;81(2):448–55. doi: 10.1016/0003-9861(59)90225-5.
  17. Buch L, Streeter D, Halpern RM, et al. Inhibition of transfer ribonucleic acid methylase activity from several human tumors by nicotinamide and nicotinamide analogs. Biochemistry. 1972;11(3):393–7. doi: 10.1021/bi00753a015.
  18. Halpern BC, Clark BR, Hardy DN, et al. The effect of replacement of methionine by homocystine on survival of malignant and normal adult mammalian cells in culture. Proc Natl Acad Sci USA. 1974;71(4):1133–6. doi: 10.1073/pnas.71.4.1133.
  19. Judde JG, Ellis M, Frost P, et al. Biochemical analysis of the role of transmethylation in the methionine dependence of tumor cells. Cancer Res. 1989;49(17):4859–65.
  20. Hoffman RM, Jacobsen J. Reversible growth arrest in simian virus 40-transformed human fibroblasts. Proc Natl Acad Sci USA. 1980;77(12):7306–10. doi: 10.1073/pnas.77.12.7306.
  21. Guo H, Lishko VK, Herrera H, et al. Therapeutic tumor-specific cell cycle block induced by methionine starvation in vivo. Cancer Res. 1993;53(23):5676–9.
  22. Breillout F, Antoine E, Poupon MF. Methionine dependency of malignant tumors: a possible approach for therapy. J Natl Cancer Inst. 1990;82(20):1628–32. doi: 10.1093/jnci/82.20.1628.
  23. Lu S, Epner DE. Molecular mechanisms of cell cycle block by methionine restriction in human prostate cancer cells. Nutr Cancer. 2000;38(1):123–30. doi: 10.1207/S15327914NC381_17.
  24. Poirson-Bichat F, Goncalves RA, Miccoli L, et al. Methionine depletion enhances the antitumoral efficacy of cytotoxic agents in drug-resistant human tumor xenografts. Cancer Res. 2000;6(2):643–53.
  25. Guo H, Herrera H, Groce A, et al. Expression of the biochemical defect of methionine dependence in fresh patient tumors in primary histoculture. Cancer Res. 1993;53(11):2479–83.
  26. Kim DH, Muto M, Kuwahara Y, et al. Array-based comparative genomic hybridization of circulating esophageal tumor cells. Oncol Rep. 2006;16(5):1053–9. doi: 10.3892/or.16.5.1053.
  27. Poirson-Bichat F, Gonfalone G, Bras-Gone RA, et al. Growth of methionine dependent human prostate cancer (PC-3) is inhibited by ethionine combined with methionine starvation. Br J Cancer. 1997;75(11):1605–12. doi: 10.1038/bjc.1997.274.
  28. Jo YK, Park MH, Choi H, et al. Enhancement of the Antitumor Effect of Methotrexate on Colorectal Cancer Cells via Lactate Calcium Salt Targeting Methionine Metabolism / Nutr Cancer. 2017;69(4):663–73. doi: 10.1080/01635581.2017.1299879.
  29. Kreis W, Goodenow M. Methionine requirement and replacement by homocysteine in tissue cultures of selected rodent and human malignant and normal cells. Cancer Res. 1978;38(8):2259–62.
  30. Kennelly JC, Blair JA, Pheasant AE. Metabolism of 5-methyltetrahydrofolate by rats bearing the Walker 256 carcinosarcoma. Br J Cancer. 1982;46(3):440–3. doi: 10.1038/bjc.1982.222.
  31. Watkins D. Cobalamin metabolism in methionine-dependent human tumour and leukemia cell lines. Clin Investig Med. 1998;21(3):151–8.
  32. Bergstrom M, Ericson K, Hagenfeldt L, et al. PET study of methionine accumulation in glioma and normal brain tissue: competition with branched chain amino acids. J Comput Assist Tomogr. 1987;11(2):208–13. doi: 10.1097/00004728-198703000-00002.
  33. Stern PH, Hoffman RM. Elevated overall rates of transmethylation in cell lines from diverse human tumors. In Vitro. 1984;20(8):663–73. doi: 10.1007/bf02619617.
  34. Hoffman RM. Altered methionine metabolism and transmethylation in cancer. Anticancer Res. 1985;5(1):1–30.
  35. Давыдов Д.Ж., Морозова Е.А., Ануфриева Н.В. и др. Динамика содержания метионина в плазме крови мышей после введения метионин-гамма-лиазы. Российский биотерапевтический журнал. 2017;16(Suppl 1):28–9.
    [Davydov DZh, Morozova EA, Anufrieva NV, et al. The changes in plasma methionin concentrations in mice after methionine-gamma-lyase injection. Rossiiskii bioterapevticheskii zhurnal. 2017;16(Suppl 1):28–9. (In Russ)]
  36. Hoffman RM. Altered methionine metabolism, DNA methylation and oncogene expression in carcinogenesis: a review and synthesis. Biochim Biophys Acta. 1983;738(1–2):49–87. doi: 10.1016/0304-419x(84)90019-2.
  37. de Oliveira SF, Ganzinelli M, Chila R, et al. Characterization of MTAP Gene Expression in Breast Cancer Patients and Cell Lines. PLoS One. 2016;11(1):e0145647. doi: 10.1371/journal.pone.0145647.
  38. Nobori T, Karras JG, Della Ragione F, et al. Absence of methilthioadenosine phosphorylase in human gliomas. Cancer Res. 1991;51(12):3193–7.
  39. Schmid M, Malicki D, Nobori T, et al. Homozygous deletions of methilthioadenosine phosphorylase (MTAP) are more frequent then p16INK4A (CDKN2) homozygous deletions in primary non-small cell lung cancer (NSCLC). Oncogene. 1998;17(20):2669–75. doi: 10.1038/sj.onc.1202205.
  40. M’soka TJ, Nishioka J, Taga A, et al. Detection of methylthioadenosine phosphorylase (MTAP) and p16 gene deletion in T cell acute lymphoblastic leukemia by real-time quantitative PCR assay. Leukemia. 2000;14(5):935–40. doi: 10.1038/sj.leu.2401771.
  41. Garcia-Castellano JM, Villanueva A, Healey JH, et al. Methylthioadenosine phosphorylase gene deletions are common in osteosarcoma. Clin Cancer Res. 2002;8(3):782–7.
  42. Behrmann I, Wallner S, Komyod W, et al. Characterization of methylthioadenosin phosphorylase (MTAP) expression in malignant melanoma. Am J Pathol. 2003;162(2):683–90. doi: 10.1016/S0002-9440(10)63695-4.
  43. Komatsu A, Nagasaki K, Fujimori M, et al. Identification of novel deletion polymorphisms in breast cancer. Int J Oncol. 2008;33(2):261–70.
  44. Nobori T, Miura K, Wu DJ, et al. Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature. 1994;368(6473):753–6. doi: 10.1038/368753a0.
  45. Nobori T, Takabayashi K, Tran P, et al. Genomic cloning of methylthioadenosine phosphorylase: a purine metabolic enzyme deficient in multiple different cancers. Proc Natl Acad Sci USA. 2000;93(12):6203–8. doi: 10.1073/pnas.93.12.6203.
  46. Brat DJ, James CD, Jedlicka AE, et al. Molecular genetic alterations in radiation-induced astrocytomas. Am J Pathol. 1999;154(5):1431–8. doi: 10.1016/S0002-9440(10)65397-7.
  47. Christopher SA, Diegelman P, Porter CW, et al. Methylthioadenosine phosphorylase, a gene frequently codeleted with p16(cdkN2a/ARF), acts as a tumor suppressor in a breast cancer cell line. Cancer Res. 2002;62(22):6639–44.
  48. Jagasia AA, Block JA, Diaz MO, et al. Partial deletions of the CDKN2A and MTS2 putative tumor suppressor genes in a myxoid chondrosarcoma. Cancer Lett. 1996:105(1):77–90. doi: 10.1016/0304-3835(96)04273-5.
  49. Jagasia AA, Block JA, Qureshi A, et al. Chromosome 9 related aberration and deletions of the CDKN2 and MTS2 putative tumor suppressor genes in human chondrosarcomas. Cancer Lett. 1996;105(1):91–103. doi: 10.1016/0304-3835(96)04274-7.
  50. Powel EL, Leoni LM, Canto MI, et al. Concordant loss of MTAP and p16/CDKN2A expression in gastroesophageal carcinogenesis: evidence of homozygous deletion in esophageal noninvasive precursor lesions and therapeutic implications. Am J Surg Phatol. 2005;29(11):1497–504. doi: 10.1097/01.pas.0000170349.47680.e8.
  51. Kim J, Kim MA, Min SY, et al. Downregulation of methylthioadenosin phosphorylase by homozygous deletion in gastric carcinoma. Genes Chromos Cancer. 2011;50(6):421–33. doi: 10.1002/gcc.20867.
  52. Huang H-Y, Li S-H, Yu S-C, et al. Homozygous deletion of MTAP gene as a poor prognosticator in gastrointestinal stromal tumors. Clin Cancer Res. 2009;15(22):6963–72. doi: 10.1158/1078-0432.CCR-09-1511.
  53. Suzuki T, Maruno M, Wada K, et al. Genetic analysis of human glioblastomas using a genomic microarray system. Brain Tumor Pathol. 2004;21(1):27–34. doi: 10.1007/bf02482174.
  54. Zhang H, Chen ZH, Savarese TM, et al. Codeletion of the genes for p16INK4 methihthioadenosine phosphorylase, interferon-alpha1, interferon-beta1, and other 9p21 markers in human malignant cell lines. Cancer Genet Cytogenet. 1996;86(1):22–8. doi: 10.1016/0165-4608(95)00157-3.
  55. Perry A, Nobory T, Ru N, et al. Detection of p16 gene deletions in gliomas: comparison of fluorescence in situ hybridization (FISH) versus quantitative PCR. J Neuropathol Exp Neurol. 1997;56(9):999–1008. doi: 10.1097/00005072-199709000-00005.
  56. Orentreich N, Matias JR, DeFelice A, Zimmerman JA. Low methionine ingestion by rats extends life span . J Nutr. 1993;123(2):269–74.
  57. Efferth DE, Miyachi H, Drexler HG, Gebhart E. Methionine phosphorylase as target for chemoselective treatment of T-cell acute lymphoblastic leukemic cells. Blood Cells Mol Dis. 2002;28(1):47–56. doi: 10.1006/bcmd.2002.0483.
  58. Bertin R, Acquaviva C, Mirebeau D, et al. CDKN2A, CDKN2B and MTAP gene dosage permits precise characterization of mono- and bi-allelic 9p21 deletions in childhood acute lymphoblastic leukemia. Genes Chromos Cancer. 2003;37(1):44–57. doi: 10.1002/gcc.10188.
  59. Usvasalo A, Ninomiya S, Raty R, et al. Focal 9p instability in hematologic neoplasias revealed by comparative genomic hybridization and single-nucleotide polymorphism microarray analyses. Genes Chromos Cancer. 2010;49(4):309–18. doi: 10.1002/gcc.20741.
  60. Kamath A, Tara H, Xiang B, et al. Double-minute MYC amplification and deletion of MTAP, CDKN2A, CDKN2B and ELAVL2 in an acute myeloid leukemia characterized by oligonucleotide-array comparative genomic hybridization. J Cancer Genet Cytogenet. 2008;183(2):117–20. doi: 10.1016/j.cancergencyto.2008.02.011.
  61. Marce S, Balague O, Colomo L, et al. Lack of methylthioadenosine phosphorylase expression in mantle cell lymphoma is associated with shorter survival: implications for a potential targeted therapy. Clin Cancer Res. 2006;12(12):3754–61. doi: 10.1158/1078-0432.CCR-05-2780.
  62. Dreyling MH, Roulston D, Bohlander SK, et al. Codelition of CDKN2 and MTAP genes in a subset of non-Hodgkin’s lymphoma may be associated with histologic transformation from low-grade to diffuse large-cell lymphoma. Genes Chromos Cancer. 1998;22(1):72–8. doi: 10.1002/(sici)1098-2264(199805)22:1<72::aid-gcc10>3.3.co;2-g.
  63. Illei PB, Busch VW, Zakowski MF, Ladanyi M. Homozygous deletion of CDKN2A and codeletion of the methylthioadenosine phosphorylase gene in the majority of pleural mesotheliomas. Cancer Res. 2003;9(6):2108–13.
  64. Mora J, Alaminos M, de Torres C, et al. Comprehensive analysis of the 9p21 region in neuroblastoma suggests a role for genes mapping to 9p21–23 in the biology of favorable stage 4 tumours. Br J Cancer. 2004;91(6):1112–8. doi: 10.1038/sj.bjc.6602094.
  65. Hustinx SR, Hruban RH, Leoni LM, et al. Homozygous deletion of the MTAP gene in invasive adenocarcinoma of the pancreas and in periampullary cancer: a potential new target for therapy. Cancer Biol Ther. 2005;4(1):83–6. doi: 10.4161/cbt.4.1.1380.
  66. Hustinx SR, Leoni ML, Yeo CJ, et al. Concordant loss of MTAP and p16/CDRN2A expressions in pancreatic intraepithelial neoplasia: evidence of homozygous deletion in a noninvasive precursor lesion. Mod Pathol. 2005;18(7):959–63. doi: 10.1038/modpathol.3800377.
  67. Chen ZH, Zhang H, Savarese TM. Gene deletion chemoselectivity: codeletion of the genes for p16 (INK4), methylthioadenosine phosphorylase, and the alpha- and beta-interferons in human pancreatic cell carcinoma lines and its implications for. Cancer Res. 1996;56(5):1083–90.
  68. Brownhill SC, Taylor C, Burchill SA. Chromosome 9p21 gene copy number and prognostic significance of p16 in ESFT. Br J Cancer. 2007;96(12):1914–23. doi: 10.1038/sj.bjc.6603819.
  69. Conway C, Beswick S, Elliott F. Deletion at chromosome arm 9p in relation to BRAF and NRAS mutation and prognostic significance for primary melanoma. Genes Chromos Cancer. 2010;49(5):425–38. doi: 10.1002/gcc.20753.
  70. Worsham MJ, Chem KM, Tiwari N, et al. Fine-mapping loss of gene architecture at the CDKN2B (p15INK4b), CDKN2A (p14ARF, p16INK4a) and MTAP genes in head and neck squamous cell carcinoma. Arch Otol Head Neck Surg. 2006;132(4):409–15. doi: 10.1001/archotol.132.4.409.
  71. Mirebeau D, Acquaviva C, Suciu S, et al. The prognostic significance of CDKN2A, CDKN2B and EORTC studies 58881 and 58951. Haematologica. 2006;91(7):881–5.
  72. Tang B, Li YN, Kruger WD. Defects in methylthioadenosine phosphorylase is associated with but not responsible for methionine-dependent tumor cell growth. Cancer Res. 2000;60(19):5.
  73. Basu I, Locker J, Cassera MB, et al. Growth and metastases of human lung cancer are inhibited in mouse xenografts by a transition state analogue of 5ʹ-methilthioadenosine. J Biol Chem. 2010;286(6):4902–11. doi: 10.1074/jbc.M110.198374.
  74. Subhi AL, Diegelman P, Porter CW, et al. Methylthioadenosine phosphorylase regulates ornithine decarboxylase by production of downstream metabolites. J Biol Chem. 2003;278(50):49868–73. doi: 10.1074/jbc.M308451200.
  75. Kenyon SH, Waterfield CJ, Timbrell JA, et al. Methionine synthase activity and sulphur amino acid levels in the rat liver tumor cells HTS and Phi-1. J. Biochem Pharmacol. 2002;63(3):381–91. doi: 10.1016/s0006-2952(01)00874-7.
  76. Ma E, Iwasaki M, Junko I, et al. Dietary intake of folate, vitamin B6, and vitamin B12, genetic polymorphism of related enzymes, and risk of breast cancer: a case-control study in Brazilian women. BMC Cancer. 2009;24(9):122. doi: 10.1186/1471-2407-9-122.
  77. Stern PH, Wallace CD, Hoffman RM. Altered methionine metabolism occurs in all members of a set of diverse human tumor cell lines. J Cell physiol. 1984;119(1):29–34. doi: 10.1002/jcp.1041190106.
  78. Lu M, Wang F, Qiu J. Methionine synthase A2756G polymorphism and breast cancer risk: a meta-analysis involving 18,953 subjects. Breast Cancer Res Treat. 2010;123(1):213–7. doi: 10.1007/s10549-010-0755-9.
  79. Linnebank M, Fliessbach K, Kolsch H, et al. The methionine synthase polymorphism c.2756Aright curved arrow G (D919G) is relevant for disease-free longevity. Int J Mol Med. 2005;16(4):759–61.
  80. Dhillon V, Thomas P, Fenech M. Effect of common polymorphisms in folate uptake and metabolism genes on frequency of micronucleated lymphocytes in a South Australian cohort. Mutat Res. 2009;665(1–2):1–6. doi: 10.1016/j.mrfmmm.2009.02.007.
  81. Beetstra S, Suthers G, Dhillon V, et al. Methionine-dependence phenotype in the de novo pathway in BRCA1 and BRCA2 mutation carriers with and without breast cancer. Cancer Epidemiol Biomark Prev. 2008;17(10):2565–71. doi: 10.1158/1055-9965.EPI-08-0140.
  82. Drennan CL, Huang S, Drummond J, et al. How a protein binds B12: A 3.0 A X-ray structure of B12-binding domains of methionine synthase. Science. 1994;266(5191):1669–74. doi: 10.1126/science.7992050.
  83. Tisdale MJ. Methionine metabolism in Walker carcinosarcoma in vitro. Eur J Cancer. 1980;16(3):407–14. doi: 10.1016/0014-2964(80)90360-6.
  84. Liteplo RG, Hipwell SE, Rosenblatt DS, et al. Changes in cobalamin metabolism are associated with the altered methionine auxotrophy of highly growth autonomous human melanoma. J Cell Physiol. 1991;149(2):332–8. doi: 10.1002/jcp.1041490222.
  85. Fiskerstrand T, Christensen B, Tysnes OB, et al. Development and reversion of methionine dependence in a human glioma cell line: relation to homocysteine remethylation and cobalamin status. Cancer Res. 1994;54(18):4899–906.
  86. Watkins D. Cobalamin metabolism in methionine-dependent human tumour and leukemia cell lines. Clin Invest Med. 1998;21(3):151–8.
  87. Tang B, Mustafa A, Gupta S, et al. Methionine-deficient diet induces post-transcriptional down-regulation of cystathionine beta-synthase. Nutrition. 2009;26(11–12):170–5. doi: 10.1016/j.nut.2009.10.006.
  88. Breillout F, Hadida F, Echinard-Garin P, et al. Decreased rat rhabdomyosarcoma pulmonary metastases in response to low methionine diet. Anticancer Res. 1987;7(4b):861–7.
  89. Komninou D, Leutzinger Y, Reddy BS, et al. Methionine restriction inhibits colon carcinogenesis. Nutr Cancer. 2006;54(2):202–8. doi: 10.1207/s15327914nc5402_6.
  90. Graziosi L, Mencarelli A, Renga B, et al. Epigenetic modulation by methionine deficiency attenuates the potential for gastric cancer cell dissemination. J Gastrointest Surg. 2013;17(1):39–49. doi: 10.1007/s11605-012-1996-1.
  91. Theuer RC. Effect of essential amino acid restriction on the growth of female C57BL mice and their implanted BW10232 adenocarcinomas. J Nutr. 1971;101(2):223–32.
  92. Caro P, Gomez J, Sanchez I, et al. Forty percent methionine restriction decreases mitochondrial oxygen radical production and leak at complex I during forward electron flow and lowers oxidative damage to proteins and mitochondrial DNA in rat kidney and brain mitochondria. Rejuven Res. 2009;12(6):421–34. doi: 10.1089/rej.2009.0902.
  93. Ryu CS, Kwak HC, Lee KS, et al. Sulfur amino acid metabolism in doxorubicin-resistant breast cancer cells. Toxicol Appl Pharmacol. 2011;15;255(1):94–102. doi: 10.1016/j.taap.2011.06.004.
  94. Goseki N, Endo M. Thiol depletion and chemosensitization on nimustine hydrochloride by methionine-depleting total parenteral nutrition. Tohoku J Exp Med. 1990;161(3):227–39. doi: 10.1620/tjem.161.227.
  95. Hoshiya Y, Guo H, Kubota T, et al. Human tumors are methionine dependent in vivo. Anticancer Res. 1995;15(3):717–8.
  96. Epne DE, Morrow S, Wilcox M, Houghton JL. Nutrient intake and nutritional indexes in adults with metastatic cancer on a phase l clinical trial of dietary methionine restriction. Nutr Cancer. 2002;42(2):158–66. doi: 10.1207/S15327914NC422_2.
  97. Goseki N, Yamazaki S, Shimojyu K, et al. Synergistic effect of methionine-depleting total parenteral nutrition with 5-fluorouracil on human gastric cancer: a randomized, prospective clinical trial. Jpn J Cancer Res. 1995;86(5):484–9. doi: 10.1111/j.1349-7006.1995.tb03082.x.
  98. Durando X, Farges MC, Buc E, et al. Dietary methionine restriction with FOLFOX regimen as first line therapy of metastatic colorectal cancer: a feasibility study. Oncology. 2008;78(3–4):205–9. doi: 10.1159/000313700.
  99. Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol. 2005;174(3):1065–70. doi: 10.1097/01.ju.0000169487.49018.73.
  100. McCarty M, Barroso-Aranda J, Contreras F, et al. The low-methionine content of vegan diets may make methionine restriction feasible as a life extension strategy. Med Hypotheses. 2009;72(2):125–8. doi: 10.1016/j.mehy.2008.07.044.
  101. Kack H, Sandmark J, Gibson K, et al. Crystal structure of diaminopelargonic acid synthase: evolutionary relationships between pyridoxal-5ʹ-phosphate-dependent enzymes. J Mol Biol. 1999;291:857–76. doi: 10.1006/jmbi.1999.2997.
  102. Fernandes HS, Silva Teixeira CS, Fernandes PA, et al. Amino acid deprivation using enzymes as a targeted therapy for cancer and viral infections. Expert Opin Ther Pat. 2017;27(3):283–97. doi: 10.1080/13543776.2017.1254194.
  103. Gay F, Aguera K, Senechal K, et al. Methionine tumor starvation by erythrocyte-encapsulated methionine gamma-lyase activity controlled with per os vitamin B6. Cancer Med. 2017. doi: 10.1002/cam4.1086.
  104. Покровский В.С., Трещалина Е.М. Ферментные препараты в онкогематологии: актуальные направления экспериментальных исследований и перспективы клинического применения. Клиническая онкогематология. 2014;7(1):28–38.
    [Pokrovskiy VS, Treshchalina YeM. Enzymes in oncohematology: relevant directions of experimental studies and prospects of clinical use. Klinicheskaya onkogematologiya. 2014;7(1):28–38. (In Russ)]
  105. Манухов И.В., Мамаева Д.В., Морозова Е.А. и др. L-метионин–гамма-лиаза Citrobacter freundii: клонирование гена и кинетические параметры фермента. Биохимия. 2006;71(4):454–63.
    [Manukhov IV, Mamaeva DV, Morozova EA, et al. L-methionine γ-lyase from Citrobacter freundii: cloning of the gene and kinetic parameters of the enzyme. Biokhimiya. 2006;71(4):454–63. (In Russ)]
  106. Cellarier E, Durando X, Vasson MP, et al. Methionine dependency and cancer treatment. Cancer Treat Rev. 2003;29(6):489–99. doi: 10.1016/s0305-7372(03)00118-x.
  107. Tan Y, Xu M, Hoffman RM. Broad selective efficacy of recombinant methioninase and polyethylene glycol-modified recombinant methioninase on cancer cells in vitro. Anticancer Res. 2010;30:1041–6.
  108. Kreis W, Hession C. Isolation and purification of L-methionine-alpha-deamino-gamma-mercaptomethane-lyase (L-methioninase) from Clostridium sporogenes. Cancer Res. 1973;33:1862–5.
  109. 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.
  110. El-Sayed SA, Shouman HM, Nassrat HM. Pharmacokinetics, immunogenicity and anticancer efficiency of Aspergillus flavipes L-methioninase. Enzyme Microb Technol. 2012;51(4):200–10. doi: 10.1016/j.enzmictec.2012.06.004.
  111. Huang K-Y, Hu H-Y, Tang Y-L, et al. High-level expression, purification and large-scale production of L-methionine γ-Lyase from Ideomarina as a novel anti-leucemic drug. Mar Drugs. 2015;13(8):5492–507. doi: 10.3390/md13085492.
  112. Yano S, Li S, Han Q, et al. Selective methioninase-inducted trap of cancer cells in S/G2 phase visualized by FUCCI imaging confers chemosensitivity. Oncotarget. 2014;5(18):8729–36. doi: 10.18632/oncotarget.2369.
  113. Nagahama T, Goseki N, Endo M. Doxorubicin and vincristine with methionine depletion contributed to survival in the Yoshida sarcoma bearing rats. Anticancer Res. 1998;18(1):25–31.
  114. Machrover D, Zittoun J, Broet Ph, et al. Cytotoxic synergism of methioninase in combination with 5-fluorouracil and folinic acid. Biochem Pharmacol. 2001;61(7):867–76. doi: 10.1016/s0006-2952(01)00560-3.
  115. Smiraglia DJ. Excessive CpG island hypermethylation in cancer cell lines versus primary human malignancies. Hum Mol Genet. 2001;10(13):1413–9. doi: 10.1093/hmg/10.13.1413.
  116. Jeanblanc M, Mousli M, Hopfner R, et al. The retinoblastoma gene and its product are targeted by ICBP90: a key mechanism in the G1/S transition during the cell cycle. Oncogene. 2005;24(49):7337–45. doi: 10.1038/sj.onc.1208878.
  117. Hu J, Cheung NK. Methionine depletion with recombinant methioninase: In vitro and in vivo efficacy against neuroblastoma and its synergism with chemotherapeutic drug. Int J Cancer. 2009;124(7):1700–6. doi: 10.1002/ijc.24104.
  118. Kokkinakis DM, Schold H, Hori H, et al. 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. doi: 10.1080/01635589709514624.
  119. Tan Y, Xu M, Guo H, et al. Anticancer efficacy of methioninase in vivo. Anticancer Res. 1996;16(6С):3931–6.
  120. 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.
  121. 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.
  122. Hoshiya Y, Kubota T, Matsuzaki SW, et al. Methionine starvation modulates the efficacy of cisplatin on human breast cancer in nude mice. Anticancer Res. 1996;16(6B):3515–7.
  123. Kokkinakis DM, Hoffman RM, Frenkel EP, et al. Synergy between methionine stress and chemotherapy in the treatment of brain tumor xenografts in athymic mice. Cancer Res. 2001;61(10):4017–23.
  124. Tan Y, Zavala JSr, Xu M, et al. Serum methionine depletion without side effects by methioninase in metastatic breast cancer patients. Anticancer Res. 1996;16(6):3937–42.
  125. Morozova EA, Anufrieva NV, Davydov DZ, et al. Plasma methionine depletion and pharmacokinetic properties in mice of methionine γ-lyase from Citrobacter freundii, Clostridium tetani and Clostridium sporogenes. Biomed Pharmacother. 2017;88:978–84. doi: 10.1016/j.biopha.2017.01.127.
  126. Покровский В.С., Лесная Н.А., Трещалина Е.М. и др. Перспективы разработки новых ферментных противоопухолевых препаратов. Вопросы онкологии. 2011;57(2):155–64.
    [Pokrovskii VS, Lesnaya NA, Treshchalina EM, et al. Perspectives in the development of new enzyme anticancer treatments. Voprosy onkologii. 2011;57(2):155–64. (In Russ)]
  127. Покровская М.В., Покровский В.С., Соколов Н.Н. Дифференциальная среда для выявления штаммов бактерий-продуцентов L-аспарагиназ. Прикладная биохимия и микробиология. 2011;47(2):183–6.
    [Pokrovskaya MV, Pokrovskii VS, Sokolov NN, et al. Differential medium for revealing bacterial producer strains of L-asparaginases.) Prikladnaya biokhimiya i mikrobiologiya. 2011;47(2):183–6. (In Russ)]
  128. Pokrovskii VS, Pokrovskaya MV, Aleksandrova SS, et al. Physicochemical properties and antiproliferative activity of recombinant Yersinia pseudotuberculosis L-asparaginase. Appl Biochem Microbiol. 2013;49(1):18–22. doi: 10.1134/s000368381301016x.
  129. Pokrovskaya MV, Pokrovskiy VS, Aleksandrova SS, et al. Recombinant intracellular Rhodospirillum rubrum L-asparaginase with low L-glutaminase activity and antiproliferative effect. Biochem (Moscow) Suppl. Series B: Biomed Chem. 2012;6(2):123–31. doi: 10.1134/s1990750812020096.
  130. Sidoruk KV, Bogush VG, Pokrovsky VS, et al. Creation of a producent, optimization of expression, and purification of recombinant Yersinia pseudotuberculosis L-asparaginase. Bull Exp Biol Med. 2011;152(2):219–23. doi: 10.1007/s10517-011-1493-7.
  131. Pokrovsky VS, Pokrovskaya MV, Aleksandrova SS, et al. Comparative immunogenicity and structural analysis of epitopes of different bacterial L-asparaginases. BMC Cancer. 2016;16(1):89. doi: 10.1186/s12885-016-2125-4.
  132. Sannikova EP, Bulushova NV, Cheperegin SE, et al. The modified heparin-binding L-asparaginase of Wolinella succinogenes. Mol Biotechnol. 2016;58(8–9):528–39. doi: 10.1007/s12033-016-9950-1.
  133. Pokrovskaya MV, Aleksandrova SS, Pokrovsky VS, et al. Identification of functional regions in the Rhodospirillum rubrum L-asparaginase by site-directed mutagenesis. Mol Biotechnol. 2015;57(3):251–64. doi: 10.1007/s12033-014-9819-0.
  134. Покровский В.С., Лукашева Е.В., Трещалина Е.М. и др. Экспериментальная оценка синергизма цисплатина с L-лизин-α-оксидазой. Вопросы онкологии. 2014;60(1):90–3.
    [Pokrovskii VS, Lukasheva EV, Treshchalina EM, et al. Experimental evaluation of synergism of cisplatin with L-lysine-α-oxidase.) Voprosy onkologii. 2014;60(1):90–3. (In Russ)]
  135. Покровский В.С., Трещалина Е.М., Трещалин И.Д. и др. Оценка противоопухолевой эффективности комбинации L-лизин-α-оксидазы и иринотекана в эксперименте. Онкология. 2012;2:58–61.
    [Pokrovskii VS, Treshchalina EM, Treshchalin ID, et al. Evaluation of the antitumor efficacy of a combination of L-lysine α-oxidase and irinotecan in the experiment. Onkologiya. 2012;2:58–61. (In Russ)]

 

 

Cytogenetic and Molecular Genetic Prognostic Factors of Acute Lymphoblastic Leukemias

AV Misyurin

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

For correspondence: Andrei Vital’evich Misyurin, PhD, 24 Kashirskoe sh., Moscow, Russian Federation, 115478; e-mail: and@genetechnology.ru

For citation: Misyurin AV. Cytogenetic and Molecular Genetic Prognostic Factors of Acute Lymphoblastic Leukemias. Clinical oncohematology. 2017;10(3):317–23 (In Russ).

DOI: 10.21320/2500-2139-2017-10-3-317-323


ABSTRACT

This review presents characteristic and reproducible chromosome rearrangements in acute lymphoblastic leukemia (ALL), which can be detected with a standard cytogenetic research (G-bands staining) or by FISH. More subtle genetic changes, inaccessible to the observation of cytogeneticists, are detected with the help of modern methods of molecular biological diagnosis. The prognostic value of cytogenetic and molecular genetic markers of ALL is shown in this article. A minimal set of clinically relevant molecular markers is presented, which it is advisable to investigate with ALL.

Keywords: acute lymphoblastic leukemia, chromosomal aberration, chimeric oncogene, gene expression, gene mutation.

Received: December 3, 2016

Accepted: March 8, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. 776 с.
    [Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. 776 p. (In Russ)]
  2. Inaba H, Greaves M, Mullighan CG. Acute lymphoblastic leukaemia. Lancet. 2013;381(9881):1943–55. doi: 10.1016/S0140-6736(12)62187-4.
  3. Shago M. Recurrent Cytogenetic Abnormalities in Acute Lymphoblastic Leukemia. Meth Mol Biol. 2017;1541:257–78. doi: 10.1007/978-1-4939-6703-2_21.
  4. Deshpande PA, Srivastava VM, Mani S, et al. Atypical BCR-ABL11 fusion transcripts in adult B-acute lymphoblastic leukemia, including a novel fusion transcript-e8a1. Leuk Lymphoma. 2016;57(10):2481–4. doi: 10.3109/10428194.2016.1151512.
  5. McGregor S, McNeer J, Gurbuxani S. Beyond the 2008 World Health Organization classification: the role of the hematopathology laboratory in the diagnosis and management of acute lymphoblastic leukemia. Semin Diagn Pathol. 2012;29(1):2–11.
  6. Zerbini MCN, Soares FA, Velloso EDRP, et al. World Health Organization classification of tumors of hematopoietic and lymphoid tissues, 2008: major changes from the 3rd edition. Revista da Associacao Medica Brasileira. 2011;57(1):6–73. doi: 10.1590/S0104-42302011000100019.
  7. Paulsson K, Johansson B. High hyperdiploid childhood acute lymphoblastic leukemia. Genes Chromos Cancer. 2009;48(8):637–60. doi: 10.1002/gcc.20671.
  8. Mrоzek K, Harper DP, Aplan PD. Cytogenetics and Molecular Genetics of Acute Lymphoblastic Leukemia. Hematol Oncol Clin North Am. 2009;23(5):1–20. doi: 10.1016/j.hoc.2009.07.001.
  9. Faderl S, Estrov Z. Residual disease in acute lymphoblastic leukemia of childhood: methods of detection and clinical relevance. Cyt Cell Mol Ther. 1998;4(2):73–85.
  10. Heerema NA, Raimondi SC, Anderson JR, et al. Specific extra chromosomes occur in a modal number dependent pattern in pediatric acute lymphoblastic leukemia. Genes Chromos Cancer. 2007;46(7):684–93. doi: 10.1002/gcc.20451.
  11. Woo JS, Alberti MO, Tirado CA. Childhood B-acute lymphoblastic leukemia: a genetic update. Exper Hematol Oncol. 2014;3(1):16. doi: 10.1186/2162-3619-3-16.
  12. Sutcliffe MJ, Shuster JJ, Sather HN, et al. High concordance from independent studies by the Children’s Cancer Group (CCG) and Pediatric Oncology Group (POG) associating favorABL1e prognosis with combined trisomies 4, 10, and 17 in children with NCI Standard-Risk B-precursor Acute Lymphoblastic Leukemia: a Children’s Oncology Group (COG) initiative. Leukemia. 2005;19(5):734–40. doi: 10.1038/sj.leu.2403673.
  13. Moorman AV, Richards SM, Martineau M, et al. Outcome heterogeneity in childhood high-hyperdiploid acute lymphoblastic leukemia. Blood. 2003;102(8):2756–62. doi: 10.1182/blood-2003-04-1128.
  14. Forestier E, Johansson B, Gustafsson G, et al. Prognostic impact of karyotypic findings in childhood acute lymphoblastic leukaemia: a Nordic series comparing two treatment periods. Br J Haematol. 2000;110(1):147–53.
  15. Raimondi SC, Pui CH, Hancock ML, et al. Heterogeneity of hyperdiploid (51-67) childhood acute lymphoblastic leukemia. Leukemia. 1996;10(2):213–24.
  16. Pullarkat V, Slovak ML, Kopecky KJ, et al. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood. 2008;111(5):2563–72. doi: 10.1182/blood-2007-10-116186.
  17. Oostlander AE, Meijer GA, Ylstra B. Microarray-based comparative genomic hybridization and its applications in human genetics. Clin Genet. 2004;66(6):488–95. doi: 10.1111/j.1399-0004.2004.00322.x.
  18. Rubnitz JE, Wichlan D, Devidas M, et al. Prospective analysis of TEL gene rearrangements in childhood acute lymphoblastic leukemia: a Children’s Oncology Group study. J Clin Oncol. 2008;26(13):2186–91. doi: 10.1200/JCO.2007.14.3552.
  19. Attarbaschi A, Mann G, Konig M, et al. Incidence and relevance of secondary chromosome abnormalities in childhood TEL/AML1+ acute lymphoblastic leukemia: an interphase FISH analysis. Leukemia. 2004;18(10):1611–6. doi: 10.1038/sj.leu.2403471.
  20. Pullarkat V, Slovak ML, Kopecky KJ, et al. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood. 2008;111(5):2563–72. doi: 10.1182/blood-2007-10-116186.
  21. Stock W. Advances in the treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia. Clin Adv Hematol Oncol. 2008;6(7):487–8.
  22. Fielding AK, Rowe JM, Richards SM, et al. Prospective outcome data on 267 unselected adult patients with Philadelphia-chromosome positive acute lymphoblastic leukemia confirms superiority of allogeneic transplantation over chemotherapy in the pre-imatinib era: results from the international ALL trial MRC KALLXII/ECOG2993. Blood. 2009;113(19):4489–96. doi: 10.1182/blood-2009-01-199380.
  23. Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a metaanalysis. Leukemia. 2005;19(8):1345–9. doi: 10.1038/sj.leu.2403838.
  24. Moorman AV, Richards SM, Robinson HM, et al. Prognosis of children with acute lymphoblastic leukemia (ALL) and intrachromosomal amplification of chromosome 21 (iAMP21). Blood. 2007;109(6):2327–30. doi: 10.1182/blood-2006-08-040436.
  25. Heerema NA, Harbott J, Galimberti S, et al. Secondary cytogenetic aberrations in childhood Philadelphia chromosome positive acute lymphoblastic leukemia are nonrandom and may be associated with outcome. Leukemia. 2004;18(4):693–702. doi: 10.1038/sj.leu.2403324.
  26. Wetzler M, Talpaz M, Estrov Z, Kurzrock R. CML: mechanisms of disease initiation and progression. Leuk Lymphoma. 1993;11(Suppl 1):47–50. doi: 10.3109/10428199309047863.
  27. Chessells JM, Swansbury GJ, Reeves B, et al. Cytogenetics and prognosis in childhood lymphoblastic leukaemia: results of MRC UKALL X. Br J Haematol. 1997;99(1):93–100.
  28. Chessells JM, Harrison CJ, Kempski H, et al. Clinical features, cytogenetics and outcome in acute lymphoblastic and myeloid leukaemia of infancy: report from the MRC Childhood Leukaemia working party. Leukemia. 2002;16(5):776–84. doi: 10.1038/sj.leu.2402468.
  29. Moorman AV, Raimondi SC, Pui CH, et al. No prognostic effect of additional chromosomal abnormalities in children with acute lymphoblastic leukemia and 11q23 abnormalities. Leukemia. 2005;19(4):557–63. doi: 10.1038/sj.leu.2403695.
  30. Pui CH, Sandlund JT, Pei D, et al. Results of therapy for acute lymphoblastic leukemia in black and white children. JAMA. 2003;290(15):2001–7. doi: 10.1001/jama.290.15.2001.
  31. Pui C-H, Chessells JM, Camitta B, et al. Clinical heterogeneity in childhood acute lymphoblastic leukemia with 11q23 rearrangements. Leukemia. 2003;17(4):700–6. doi: 10.1038/sj.leu.2402883.
  32. Jeha S, Pei D, Raimondi SC, et al. Increased risk for CNS relapse in pre-B cell leukemia with the t(1;19)/TCF3-PBX1. Leukemia. 2009;23(8):1406–9. doi: 10.1038/leu.2009.42.
  33. Mrozek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;35:365–77. doi: 10.1053/j.seminoncol.2008.04.007.
  34. Wetzler M, Dodge RK, Mrozek K, et al. Prospective karyotype analysis in adult acute lymphoblastic leukemia: the cancer and leukemia Group B experience. Blood. 1999;93(11):3983–93.
  35. Bernard OA, Busson-LeConiat M, Ballerini P, et al. A new recurrent and specific cryptic translocation, t(5;14)(q35;q32), is associated with expression of the Hox11L2 gene in T acute lymphoblastic leukemia. Leukemia. 2001;15(10):1495–504. doi: 10.1038/sj.leu.2402249.
  36. Graux C, Stevens-Kroef M, Lafage M, et al. Heterogeneous patterns of amplification of the NUP214-ABL11 fusion gene in T-cell acute lymphoblastic leukemia. Leukemia. 2009;23(1):125–33. doi: 10.1038/leu.2008.278.
  37. Quintas-Cardama A, Tong W, Manshouri T, et al. Activity of tyrosine kinase inhibitors against human NUP214-ABL11-positive T cell malignancies. Leukemia. 2008;22(6):1117–24. doi: 10.1038/leu.2008.80.
  38. Krawczyk J, Haslam K, Lynam P, et al. No prognostic impact of P2RY8-CRLF2 fusion in intermediate cytogenetic risk childhood B-cell acute lymphoblastic leukaemia. Br J Haematol. 2013;160(4):555–6. doi: 10.1111/bjh.12130.
  39. Hoelzer D. Personalized medicine in adult acute lymphoblastic leukemia. Haematologica. 2015;100(7):855–8. doi: 10.3324/haematol.2015.127837.
  40. Tsuzuki S, Taguchi O, Seto M. Promotion and maintenance of leukemia by ERG. Blood. 2011;117(14):3858–68. doi: 10.1182/blood-2010-11-320515.
  41. Mullighan CG, Su X, Zhang J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360(5):470–80. doi: 10.1056/NEJMoa0808253.
  42. Mullighan CG, Miller CB, Radtke I, et al. BCR-ABL11 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453(7191):110–4. doi: 10.1038/nature06866.
  43. Yoda A, Yoda Y, Chiaretti S, et al. Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA. 2010;107(1):252–7. doi: 10.1073/pnas.0911726107.
  44. Mullighan CG, Zhang J, Kasper LH, et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature. 2011;471(7337):235–9. doi: 10.1038/nature09727.
  45. Van Vlierberghe P, Palomero T, Khiabanian H, et al. PHF6 mutations in T-cell acute lymphoblastic leukemia. Nat Genet. 2010;42(4):338–42. doi: 10.1038/ng.542.
  46. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009;10(2):147–56. doi: 10.1016/S1470-2045(08)70314-0.
  47. Weng AP, Ferrando AA, Lee W, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004;306(5694):269–71. doi: 10.1126/science.1102160.
  48. Гапонова Т.В., Менделеева Л.П., Мисюрин А.В. и др. Экспрессия опухолеассоциированных генов PRAME, WT1 и XIAP у больных множественной миеломой. Онкогематология. 2009;2:52–5.
    [Gaponova TV, Mendeleeva LP, Misyurin AV, et al. PRAME, WT1 and XIAP tumor-associated genes expression in multiple myeloma patients. Onkogematologiya. 2009;2:52–5. (In Russ)]
  49. Абраменко И.В., Белоус Н.И., Крячок И.А. и др. Экспрессия гена PRAME при множественной миеломе. Терапевтический архив. 2004;76(7):77–81.
    [Abramenko IV, Belous NI, Kryachok IA, et al. PRAME gene expression in multiple myeloma. Terapevticheskii arkhiv. 2004;76(7):77–81. (In Russ)]
  50. Мисюрин В.А. Аутосомные раково-тестикулярные гены. Российский биотерапевтический журнал. 2014;13(3):77–82.
    [Misyurin VA. Autosomal cancer-testis genes. Rossiiskii bioterapevticheskii zhurnal. 2014;13(3):77–82. (In Russ)]
  51. Мисюрин А.В. Основы молекулярной диагностики онкогематологических заболеваний. Российский биотерапевтический журнал. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24.
    [Misyurin AV. Essentials of the molecular diagnosis of oncohematological diseases. Rossiysky bioterapevtichesky zhurnal. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24. (In Russ)]

 

Molecular Genetic Abnormalities in the Pathogenesis of Hematologic Malignancies and Corresponding Changes in Cell Signaling Systems

LR Tilova1, AV Savinkova1, EM Zhidkova1,2, OI Borisova1,3, TI Fetisov1,4, KA Kuzin1, OA Vlasova1, AS Antipova3, OYu Baranova3, KI Kirsanov1, GA Belitskii1, MG Yakubovskaya1, EA Lesovaya1,5

1 Institute of Carcinogenesis, NN Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russia, 115478

2 Moscow Technological University, 78 Vernadskogo pr-t, Moscow, Russia, 119454

3 Institute of Clinical Oncology, NN Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russia, 115478

4 IM Sechenov 1st Moscow Medical State University, 8 bld. 2 Trubetskaya str., Moscow, Russia, 119991

5 IP Pavlov Ryazan Medical State University, 9 Vysokovol’tnaya str., Ryazan, Russia, 390026

For correspondence: Ekaterina Andreevna Lesovaya, PhD, 24 bld. 15 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel: 8(910)471-41-28; e-mail: lesovenok@yandex.ru

For citation: Tilova LR, Savinkova AV, Zhidkova EM, et al. Molecular Genetic Abnormalities in the Pathogenesis of Hematologic Malignancies and Corresponding Changes in Cell Signaling Systems. Clinical oncohematology. 2017;10(2):235–49 (In Russ).

DOI: 10.21320/2500-2139-2017-10-2-235-249


ABSTRACT

Hematological disorders include a wide spectrum of malignancies of hematopoietic and lymphoid tissues. The genetic changes underlying the pathogenesis of the diseases are specific for each disease. High incidence of chromosomal aberrations (deletion, translocation, insertion) is one of the principal characteristics of oncohematological diseases. In addition, mutations in individual genes or blocking of normal regulation of gene functioning in relation to epigenetic events can occur. Progression of oncohematological diseases could be a result of accumulation of different genetic abnormalities. Modern classification of malignancies of hematopoietic and lymphoid tissues is based on the analysis of clinical data, morphological and functional characteristics of tumor cells and identification of specific cytogenetic and molecular-genetic changes. A large number of genetic abnormalities specific for certain types of hematological malignancies has been discovered to date. It allows to optimize the treatment strategy, as well as to design, test and introduce to the clinical practice a number of targeted drugs (inhibitors of chimeric proteins formed as a result of translocations and triggering the malignant cell transformation). Drugs based on monoclonal antibodies (Rituximab, Alemtuzumab, etc.) or low molecular weight compounds (Imatinib, Bortezomib, Carfilzomib) form this group of medications. The knowledge about not only specific gene abnormalities but also about the corresponding changes in cell efferent signaling pathways could be of great interest for the development of new targeted molecules or the repurposing of known chemotherapeutic agents. The present review compares genetic aberrations in diseases listed in the 2008 WHO classification (amended in 2016) of hematopoietic and lymphoid tissue malignancies and main changes in cell signaling pathways associated with malignant transformation of hematopoietic cells.

Keywords: tumors of hematopoietic and lymphoid tissues, chromosomal abnormalities, cell signaling disruption, WHO classification.

Received: September 29, 2016

Accepted: January 16, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Van Etten RA. Aberrant cytokine signaling in leukemia. Oncogene. 2007;26(47):6738–49. doi: 10.1038/sj.onc.1210758.
  2. Tefferi A, Thiele J, Orazi A, et al. Proposals and rationale for revision of the World Health Organization diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary myelofibrosis: recommendations from an ad hoc international expert panel. Blood. 2007;110(4):1092–7. doi: 10.1182/blood-2007-04-083501.
  3. Jabbour EJ, Hughes TP, Cortes JE, et al. Potential mechanisms of disease progression and management of advanced-phase chronic myeloid leukemia. Leuk Lymphoma. 2014;55(7):1451–62. doi: 10.3109/10428194.2013.845883.
  4. Kota J, Caceres N, Constantinescu SN. Aberrant signal transduction pathways in myeloproliferative neoplasms. Leukemia. 2008;22(10):1828–40. doi: 10.1038/leu.2008.236.
  5. Tefferi A, Sirhan S, Lasho TL, et al. Concomitant neutrophil JAK2 mutation screening and PRV-1 expression analysis in myeloproliferative disorders and secondary polycythaemia. Br J Haematol. 2005;131(2):166–71. doi: 10.1111/j.1365–2141.2005.05743.x.
  6. Smalley KS, Sondak VK, Weber JS. c-KIT signaling as the driving oncogenic event in sub-groups of melanomas. Histol Histopathol. 2009;24(5):643–50. doi: 10.14670/HH-24.643.
  7. Campo E, Swerdlow SH, Harris NL, et al. The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood. 2011;117(19):5019–32. doi: 10.1182/blood-2011-01-293050.
  8. Копнин БП. Неопластическая клетка: основные свойства и механизмы их возникновения. Практическая онкология. 2002;3(4):229–35.
    [Kopnin BP. Neoplastic cell: principal characteristics and mechanisms of their development. Prakticheskaya onkologiya. 2002;3(4):229–35. (In Russ)]
  9. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.
  10. Bain BJ, Ahmad S. Should myeloid and lymphoid neoplasms with PCM1-JAK2 and other rearrangements of JAK2 be recognized as specific entities? Br J Haematol. 2014;166(6):809–17. doi: 10.1111/bjh.1296.
  11. Surani MA, Hajkova P. Epigenetic reprogramming of mouse germ cells toward totipotency. Cold Spring Harb Symp Quant Biol. 2010;75:211–8. doi: 10.1101/sqb.2010.75.010.
  12. Carbuccia N, Murati A, Trouplin V, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009;23(11):2183–6. doi: 10.1038/leu.2009.141.
  13. Переводчикова Н.И., Горбунова, В.А. Руководство по химиотерапии опухолевых заболеваний, 4-е издание. Москва: Практическая медицина, 2015.
    [Perevodchikova NI, Gorbunova VA. Rukovodstvo po khimioterapii opukholevykh zabolevanii. (Guidelines for chemotherapy of tumors.) 4th edition. Moscow: Prakticheskaya meditsina Publ.; 2015. (In Russ)]
  14. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937–51. doi: 10.1182/blood-2009-03-209262.
  15. Riveiro-Falkenbach E, Soengas MS. Control of tumorigenesis and chemoresistance by the DEK oncogene. Clin Cancer Res. 2010;16(11):2932–8. doi: 10.1158/1078-0432.CCR-09-2330.
  16. Naoe T. Developing target therapy against oncogenic tyrosine kinase in myeloid maliganacies. Curr Pharm Biotechnol. 2006;7(5):331–7. doi: 10.2174/138920106778521514.
  17. Buonamici S, Chakraborty S, Senyuk V, et al. The role of EVI1 in normal and leukemic cells. Blood Cells Mol Dis. 2003;31(2):206–12. doi: 10.1016/S1079-9796(03)00159-1.
  18. O’Neil J, Calvo J, McKenna K, et al. Activating Notch1 mutations in mouse models of T-ALL. Blood. 2006;107(2):781–5. doi: 10.1182/blood-2005-06-2553.
  19. Williams JH, Daly LN, Ingley E, et al. HLS7, a hemopoietic lineage switch gene homologous to the leukemia-inducing gene MLF1. EMBO J. 1999;18(20):5559–66. doi: 10.1093/emboj/18.20.5559.
  20. Rau R, Brown P. Nucleophosmin (NPM1) mutations in adult and childhood acute myeloid leukaemia: towards definition of a new leukaemia entity. Hematol Oncol. 2009;27(4):171–81. doi: 10.1002/hon.904.
  21. Simon MC. Transcription factor GATA-1 and erythroid development. Proc Soc Exp Biol Med. 1993;202(2):115–21.
  22. Orkin SH, Shivdasani RA, Fujiwara Y, et al. Transcription factor GATA-1 in megakaryocyte development. Stem Cells. 1998;16(Suppl 2):79–83. doi: 10.1002/stem.5530160710.
  23. Shimizu R, Engel JD, Yamamoto M. GATA1-related leukaemias. Nat Rev Cancer. 2008;8(4):279–87. doi: 10.1038/nrc2348.
  24. Yoshida K, Toki T, Okuno Y, et al. The landscape of somatic mutations in Down syndrome-related myeloid disorders. Nat Genet. 2013;45(11):1293–9. doi: 10.1038/ng.2759.
  25. Coluccia AM, Vacca A, Dunach M, et al. Bcr-Abl stabilizes beta-catenin in chronic myeloid leukemia through its tyrosine phosphorylation. EMBO J. 2007;26(5):1456–66. doi: 10.1038/sj.emboj.7601485.
  26. Sengupta A, Banerjee D, Chandra S, et al. Deregulation and cross talk among Sonic hedgehog, Wnt, Hox and Notch signaling in chronic myeloid leukemia progression. Leukemia. 2007;21(5):949–55. doi: 10.1038/sj.leu.240465.
  27. Sano H, Ohki K, Park MJ, et al. CSF3R and CALR mutations in paediatric myeloid disorders and the association of CSF3R mutations with translocations, including t(8;21). Br J Haematol. 2015;170(3):391–7. doi: 10.1111/bjh.13439.
  28. Maxson JE, Gotlib J, Pollyea DA, et al. Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML. N Engl J Med. 2013;368(19):1781–90. doi: 10.1056/NEJMoa1214514.
  29. Tefferi A, Lasho TL, Abdel-Wahab O, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24(7):1302–9. doi: 10.1038/leu.2010.113.
  30. Barbui T, Thiele J, Vannucchi AM, et al. Rationale for revision and proposed changes of the WHO diagnostic criteria for polycythemia vera, essential thrombocythemia and primary myelofibrosis. Blood Cancer J. 2015;5(8):e337. doi: 10.1038/bcj.2015.64.
  31. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67. doi: 10.1016/j.ccr.2010.11.015.
  32. Patnaik MM, Tefferi A. Cytogenetic and molecular abnormalities in chronic myelomonocytic leukemia. Blood Cancer J. 2016;6(2):e393. doi: 10.1038/bcj.2016.5.
  33. Greenberger JS. Ras mutations in human leukemia and related disorders. Int J Cell Cloning. 1989;7(6):343–59. doi: 10.1002/stem.5530070603.
  34. Matynia AP, Szankasi P, Shen W, et al. Molecular genetic biomarkers in myeloid malignancies. Arch Pathol Lab Med. 2015;139(5):594–601. doi: 10.5858/arpa.2014-0096-RA.
  35. Fenaux P. Chromosome and molecular abnormalities in myelodysplastic syndromes. Int J Hematol. 2001;73(4):429–37. doi: 10.1007/bf02994004.
  36. Vallespi T, Imbert M, Mecucci C, et al. Diagnosis, classification, and cytogenetics of myelodysplastic syndromes. Haematologica. 1998;83(3):258–75.
  37. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241–7. doi: 10.1038/leu.2013.336.
  38. Zahid MF, Patnaik MM, Gangat N, et al. Insight into the molecular pathophysiology of myelodysplastic syndromes: targets for novel therapy. Eur J Haematol. 2016;97(4):313–20. doi: 10.1111/ejh.12771.
  39. Griffiths EA, Gore SD, Hooker C, et al. Acute myeloid leukemia is characterized by Wnt pathway inhibitor promoter hypermethylation. Leuk Lymphoma. 2010;51(9):1711–9. doi: 10.3109/10428194.2010.496505.
  40. Niebuhr B, Fischer M, Tager M, et al. Gatekeeper function of the RUNX1 transcription factor in acute leukemia. Blood Cells Mol Dis. 2008;40(2):211–8. doi: 10.1016/j.bcmd.2007.07.018.
  41. Elagib KE, Goldfarb AN. Oncogenic pathways of AML1-ETO in acute myeloid leukemia: multifaceted manipulation of marrow maturation. Cancer Lett. 2007;251(2):179–86. doi: 10.1016/j.canlet.2006.10.010.
  42. Peterson LF, Zhang DE. The 8;21 translocation in leukemogenesis. Oncogene. 2004;23(24):4255–62. doi: 10.1038/sj.onc.1207727.
  43. Slattery ML, Lundgreen A, Herrick JS, et al. Associations between genetic variation in RUNX1, RUNX2, RUNX3, MAPK1 and eIF4E and risk of colon and rectal cancer: additional support for a TGF-beta-signaling pathway. Carcinogenesis. 2011;32(3):318–26. doi: 10.1093/carcin/bgq245.
  44. Ma X, Renda MJ, Wang L, et al. Rbm15 modulates Notch-induced transcriptional activation and affects myeloid differentiation. Mol Cell Biol. 2007;27(8):3056–64. doi: 10.1128/MCB.01339-06.
  45. Feng Y, Bommer GT, Zhai Y, et al. Drosophila split ends homologue SHARP functions as a positive regulator of Wnt/beta-catenin/T-cell factor signaling in neoplastic transformation. Cancer Res. 2007;67(2):482–91. doi: 10.1158/0008-5472.CAN-06-2314.
  46. Cornet E, Mossafa H, Courel K, et al. Persistent polyclonal binucleated B-cell lymphocytosis and MECOM gene amplification. BMC Res Notes. 2016;9(1):138. doi: 10.1186/s13104-015-1742-3.
  47. Yamazaki H, Suzuki M, Otsuki A, et al. A remote GATA2 hematopoietic enhancer drives leukemogenesis in inv(3)(q21;q26) by activating EVI1 expression. Cancer Cell. 2014;25(4):415–27. doi: 10.1016/j.ccr.2014.02.008.
  48. Sato T, Goyama S, Nitta E, et al. Evi-1 promotes para-aortic splanchnopleural hematopoiesis through up-regulation of GATA-2 and repression of TGF-b signaling. Cancer Sci. 2008;99(7):1407–13. doi: 10.1111/j.1349-7006.2008.00842.x.
  49. Tokita K, Maki K, Mitani K. RUNX1/EVI1, which blocks myeloid differentiation, inhibits CCAAT-enhancer binding protein alpha function. Cancer Sci. 2007;98(11):1752–7. doi: 10.1111/j.1349-7006.2007.00597.x.
  50. Chandra P, Luthra R, Zuo Z, et al. Acute myeloid leukemia with t(9;11)(p21–22;q23): common properties of dysregulated ras pathway signaling and genomic progression characterize de novo and therapy-related cases. Am J Clin Pathol. 2010;133(5):686–93. doi: 10.1309/ajcpgii1tt4nyogi.
  51. Grimwade D, Gorman P, Duprez E, et al. Characterization of cryptic rearrangements and variant translocations in acute promyelocytic leukemia. Blood. 1997;90(12):4876–85.
  52. Morgan RG, Pearn L, Liddiard K, et al. Gamma-Catenin is overexpressed in acute myeloid leukemia and promotes the stabilization and nuclear localization of beta-catenin. Leukemia. 2013;27(2):336–43. doi: 10.1038/leu.2012.221.
  53. Koschmieder S, Halmos B, Levantini E, et al. Dysregulation of the C/EBPalpha differentiation pathway in human cancer. J Clin Oncol. 2009;27(4):619–28. doi: 10.1200/JCO.2008.17.9812.
  54. Campidelli C, Agostinelli C, Stitson R, et al. Myeloid sarcoma: extramedullary manifestation of myeloid disorders. Am J Clin Pathol. 2009;132(3):426–37. doi: 10.1309/ajcp1za7hyzkazhs.
  55. Korsmeyer SJ. Bcl-2 initiates a new category of oncogenes: regulators of cell death. Blood. 1992;80(4):879–86.
  56. Showe LC, Croce CM. The role of chromosomal translocations in B- and T-cell neoplasia. Annu Rev Immunol. 1987;5(1):253–77. doi: 10.1146/annurev.iy.05.040187.001345.
  57. Look AT. Oncogenic role of “master” transcription factors in human leukemias and sarcomas: a developmental model. Adv Cancer Res. 1995;67:25–57. doi: 10.1016/s0065-230x(08)60709-5.
  58. Sasaki K, Iwai K. Roles of linear ubiquitinylation, a crucial regulator of NF-kappaB and cell death, in the immune system. Immunol Rev. 2015;266(1):175–89. doi: 10.1111/imr.12308.
  59. Chiaretti S, Foa R. T-cell acute lymphoblastic leukemia. Haematologica. 2009;94(2):160–2. doi: 10.3324/haematol.2008.004150.
  60. Mullighan CG. The genomic landscape of acute lymphoblastic leukemia in children and young adults. Hematol Am Soc Hematol Educ Program. 2014;2014(1):174–80. doi: 10.1182/asheducation-2014.1.174.
  61. Pasqualucci L, Dalla-Favera R. The genetic landscape of diffuse large B-cell lymphoma. Semin Hematol. 2015;52(2):67–76. doi: 10.1053/j.seminhematol.2015.01.005.
  62. Noguchi M, Ropars V, Roumestand C, et al. Proto-oncogene TCL1: more than just a coactivator for Akt. FASEB J. 2007;21(10):2273–84. doi: 10.1096/fj.06-7684com.
  63. Liebisch P, Dohner H. Cytogenetics and molecular cytogenetics in multiple myeloma. Eur J Cancer. 2006;42(11):1520–9. doi: 10.1016/j.ejca.2005.12.028.
  64. Yanai S, Nakamura S, Takeshita M, et al. Translocation t(14;18)/IGH-BCL2 in gastrointestinal follicular lymphoma: correlation with clinicopathologic features in 48 patients. Cancer. 2011;117(11):2467–77. doi: 10.1002/cncr.25811.
  65. Flossbach L, Antoneag E, Buck M, et al. BCL6 gene rearrangement and protein expression are associated with large cell presentation of extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue. Int J Cancer. 2011;129(1):70–7. doi: 10.1002/ijc.25663.
  66. Janz S. Myc translocations in B cell and plasma cell neoplasms. DNA Repair (Amst). 2006;5(9–10):1213–24. doi: 10.1016/j.dnarep.2006.05.017.
  67. Aqeilan RI, Calin GA, Croce CM. miR-15a and miR-16-1 in cancer: discovery, function and future perspectives. Cell Death Differ. 2010;17(2):215–20. doi: 10.1038/cdd.2009.69.
  68. Vermeer MH, van Doorn R, Dijkman R, et al. Novel and highly recurrent chromosomal alterations in Sezary syndrome. Cancer Res. 2008;68(8):2689–98. doi: 10.1158/0008-5472.CAN-07-6398.
  69. Herling M, Patel KA, Teitell MA, et al. High TCL1 expression and intact T-cell receptor signaling define a hyperproliferative subset of T-cell prolymphocytic leukemia. Blood. 2008;111(1):328–37. doi: 10.1182/blood-2007-07-101519.
  70. Joiner M, Le Toriellec E, Despouy G, et al. The MTCP1 oncogene modifies T-cell homeostasis before leukemogenesis in transgenic mice. Leukemia. 2007;21(2):362–6. doi: 10.1038/sj.leu.2404476.
  71. Laine J, Kunstle G, Obata T, et al. The protooncogene TCL1 is an Akt kinase coactivator. Mol Cell. 2000;6(2):395–407. doi: 10.1016/S1097-2765(00)00039-3.
  72. Mosse CA, Stumph JR, Best DH, et al. A B-cell lymphoma diagnosed in “floater” tissue: implications of the diagnosis and resolution of a laboratory error. Am J Med Sci. 2009;338(3):248–51. doi: 10.1097/MAJ.0b013e3181a88dc.
  73. Roukos V, Mathas S. The origins of ALK translocations. Front Biosci. 2015;7(2):260–8. doi: 10.2741/s439.
  74. Re D, Zander T, Diehl V, Wolf J. Genetic instability in Hodgkin’s lymphoma. Ann Oncol. 2002;13(Suppl 1):19–22. doi: 10.1093/annonc/13.s1.19.
  75. Suvajdzic N, Djurdjevic P, Todorovic M, et al. Clinical characteristics of patients with lymphoproliferative neoplasms in the setting of systemic autoimmune diseases. Med Oncol. 2012;29(3):2207–11. doi: 10.1007/s12032-011-0022-x.
  76. Roberts KG, Pei D, Campana D, et al. Outcomes of children with BCR-ABL1-like acute lymphoblastic leukemia treated with risk-directed therapy based on the levels of minimal residual disease. J Clin Oncol. 2014;32(27):3012–20. doi: 10.1200/JCO.2014.55.4105.
  77. Zweidler-McKay PA, Pear WS. Notch and T cell malignancy. Semin Cancer Biol. 2004;14(5):329–40. doi: 10.1016/j.semcancer.2004.04.012.
  78. Atlas of Genetics and Cytogenetics in Oncology and Haematology. [Internet] Available from: http://www.atlasgeneticsoncology.org/ (accessed 13.03.2017).
  79. Lu D, Zhao Y, Tawatao R, et al. Activation of the Wnt signaling pathway in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2004;101(9):3118–23. doi: 10.1073/pnas.0308648100.
  80. Rahmatpanah FB, Carstens S, Hooshmand SI, et al. Large-scale analysis of DNA methylation in chronic lymphocytic leukemia. Epigenomics. 2009;1(1):39–61. doi: 10.2217/epi.09.10.
  81. Dungarwalla M, Appiah-Cubi S, Kulkarni S, et al. High-grade transformation in splenic marginal zone lymphoma with circulating villous lymphocytes: the site of transformation influences response to therapy and prognosis. Br J Haematol. 2008;143(1):71–4. doi: 10.1111/j.1365-2141.2008.07301.x.
  82. Neurath MF, Stuber ER, Strober W. BSAP: a key regulator of B-cell development and differentiation. Immunol Today. 1995;16(12):564–9. doi: 10.1016/0167-5699(95)80078-6.
  83. Bench AJ, Erber WN, Follows GA, et al. Molecular genetic analysis of haematological malignancies II: Mature lymphoid neoplasms. Int J Lab Hematol. 2007;29(4):229–60. doi: 10.1111/j.1751-553X.2007.00876.x.
  84. Brito JL, Walker B, Jenner M, et al. MMSET deregulation affects cell cycle progression and adhesion regulons in t(4;14) myeloma plasma cells. Haematologica. 2009;94(1):78–86. doi: 10.3324/haematol.13426.
  85. Aamot HV, Bjornslett M, Delabie J, et al. t(14;22)(q32;q11) in non-Hodgkin lymphoma and myeloid leukaemia: molecular cytogenetic investigations. Br J Haematol. 2005;130(6):845–51. doi: 10.1111/j.1365-2141.2005.05688.x.
  86. Arcaini L, Lucioni M, Boveri E, et al. Nodal marginal zone lymphoma: current knowledge and future directions of an heterogeneous disease. Eur J Haematol. 2009;83(3):165–74. doi: 10.1111/j.1600-0609.2009.01301.x.
  87. Du MQ. MALT lymphoma: recent advances in aetiology and molecular genetics. J Clin Exp Hematop. 2007;47(2):31–42. doi: 10.3960/jslrt.47.31.
  88. Mateo M, Mollejo M, Villuendas R, et al. 7q31–32 allelic loss is a frequent finding in splenic marginal zone lymphoma. Am J Pathol. 1999;154(5):1583–9. doi: 10.1016/S0002-9440(10)65411-9.
  89. Shimada K, Kinoshita T, Naoe T, et al. Presentation and management of intravascular large B-cell lymphoma. Lancet Oncol. 2009;10(9):895–902. doi: 10.1016/S1470-2045(09)70140-8.
  90. Bogusz AM, Seegmiller AC, Garcia R, et al. Plasmablastic lymphomas with MYC/IgH rearrangement: report of three cases and review of the literature. Am J Clin Pathol. 2009;132(4):597–605. doi: 10.1309/ajcpfur1bk0uodts.
  91. Weerkamp F, van Dongen JJ, Staal FJ. Notch and Wnt signaling in T-lymphocyte development and acute lymphoblastic leukemia. Leukemia. 2006;20(7):1197–205. doi: 10.1038/sj.leu.2404255.
  92. Zhang D, Loughran TP, Jr. Large granular lymphocytic leukemia: molecular pathogenesis, clinical manifestations, and treatment. Hematol Am Soc Hematol Educ Program. 2012;2012:652–9. doi: 10.1182/asheducation-2012.1.652.
  93. Lima M. Aggressive mature natural killer cell neoplasms: from epidemiology to diagnosis. Orphanet J Rare Dis. 2013;8(1):95. doi: 10.1186/1750-1172-8-95.
  94. Ohshima K. Molecular Pathology of Adult T-Cell Leukemia/Lymphoma. Oncology. 2015;89(Suppl 1):7–15. doi: 10.1159/000431058.
  95. Finalet Ferreiro J, Rouhigharabaei L, Urbankova H, et al. Integrative genomic and transcriptomic analysis identified candidate genes implicated in the pathogenesis of hepatosplenic T-cell lymphoma. PLoS One. 2014;9(7):e102977. doi: 10.1371/journal.pone.0102977.
  96. Ferreri AJ, Govi S, Pileri SA. Hepatosplenic gamma-delta T-cell lymphoma. Crit Rev Oncol Hematol. 2012;83(2):283–92. doi: 10.1016/j.critrevonc.2011.10.001.
  97. Devata S, Wilcox RA. Cutaneous T-Cell Lymphoma: A Review with a Focus on Targeted Agents. Am J Clin Dermatol. 2016;17(3):225–37. doi: 10.1007/s40257-016-0177-5.
  98. da Silva Almeida AC, Abate F, Khiabanian H, et al. The mutational landscape of cutaneous T cell lymphoma and Sezary syndrome. Nat Genet. 2015;47(12):1465–70. doi: 10.1038/ng.3442.
  99. Izykowska K, Przybylski GK. Genetic alterations in Sezary syndrome. Leuk Lymphoma. 2011;52(5):745–53. doi: 10.3109/10428194.2010.551159.
  100. Wang SA, Hasserjian RP. Acute Erythroleukemias, Acute Megakaryoblastic Leukemias, and Reactive Mimics: A Guide to a Number of Perplexing Entities. Am J Clin Pathol. 2015;144(1):44–60. doi: 10.1309/ajcprkyat6ezqhc7.
  101. Nicolay JP, Felcht M, Schledzewski K, et al. Sezary syndrome: old enigmas, new targets. J Dtsch Dermatol Ges. 2016;14(3):256–64. doi: 10.1111/ddg.12900.
  102. Pletneva MA, Smith LB. Anaplastic large cell lymphoma: features presenting diagnostic challenges. Arch Pathol Lab Med. 2014;138(10):1290–4. doi: 10.5858/arpa.2014-0295-CC.
  103. Ondrejka SL, Hsi ED. T-cell Lymphomas: Updates in Biology and Diagnosis. Surg Pathol Clin. 2016;9(1):131–41. doi: 10.1016/j.path.2015.11.002.
  104. Churchill H, Naina H, Boriack R, et al. Discordant intracellular and plasma D-2-hydroxyglutarate levels in a patient with IDH2 mutated angioimmunoblastic T-cell lymphoma. Int J Clin Exp Pathol. 2015;8(9):11753–9.
  105. Sakata-Yanagimoto M, Enami T, Yoshida K, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46(2):171–5. doi: 10.1038/ng.2872.
  106. Feldman AL, Dogan A, Smith DI, et al. Discovery of recurrent t(6;7)(p25.3;q32.3) translocations in ALK-negative anaplastic large cell lymphomas by massively parallel genomic sequencing. Blood. 2011;117(3):915–9. doi: 10.1182/blood-2010-08-303305.
  107. Persad P, Pang CS. Composite ALK-negative anaplastic large cell lymphoma and small lymphocytic lymphoma involving the right inguinal lymph node. Pathol Res Pract. 2014;210(2):127–9. doi: 10.1016/j.prp.2013.09.006.
  108. Kikuma K, Yamada K, Nakamura S, et al. Detailed clinicopathological characteristics and possible lymphomagenesis of type II intestinal enteropathy-associated T-cell lymphoma in Japan. Hum Pathol. 2014;45(6):1276–84. doi: 10.1016/j.humpath.2013.10.038.
  109. Djeu JY, Wei S. Clusterin and chemoresistance. Adv Cancer Res. 2009;105:77–92. doi: 10.1016/S0065-230X(09)05005-2.
  110. Sun W, Nordberg ML, Fowler MR. Histiocytic sarcoma involving the central nervous system: clinical, immunohistochemical, and molecular genetic studies of a case with review of the literature. Am J Surg Pathol. 2003;27(2):258–65. doi: 10.1097/00000478-200302000-00017.
  111. Scappaticci S, Danesino C, Rossi E, et al. Cytogenetic abnormalities in PHA-stimulated lymphocytes from patients with Langerhans cell histocytosis. AIEOP-Istiocitosi Group. Br J Haematol. 2000;111(1):258–62. doi: 10.1111/j.1365-2141.2000.02313.x.
  112. Arico M, Danesino C. Langerhans’ cell histiocytosis: is there a role for genetics? Haematologica. 2001;86(10):1009–14.
  113. Nakayama M, Takahashi K, Hori M, et al. Langerhans cell sarcoma of the cervical lymph node: a case report and literature review. Auris Nasus Larynx. 2010;37(6):750–3. doi: 10.1016/j.anl.2010.04.007.
  114. Takahashi E, Nakamura S. Histiocytic sarcoma: an updated literature review based on the 2008 WHO classification. J Clin Exp Hematopathol. 2013;53(1):1–8. doi: 10.3960/jslrt.53.1.
  115. Pettigrew HD, Teuber SS, Kong JS, et al. Contemporary challenges in mastocytosis. Clin Rev Allergy Immunol. 2010;38(2–3):125–34. doi: 10.1007/s12016-009-8164-8.
  116. Chatterjee A, Ghosh J, Kapur R. Mastocytosis: a mutated KIT receptor induced myeloproliferative disorder. Oncotarget. 2015;6(21):18250–64. doi: 10.18632/oncotarget.4213.
  117. Kairouz S, Hashash J, Kabbara W, et al. Dendritic cell neoplasms: an overview. Am J Hematol. 2007;82(10):924–8. doi: 10.1002/ajh.20857.

 

Cytogenetic and Molecular Genetic Prognostic Factors of Acute Myeloid Leukemia

AV Misyurin

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

For correspondence: Andrei Vital’evich Misyurin, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: and@genetechnology.ru

For citation: Misyurin AV. Cytogenetic and Molecular Genetic Prognostic Factors of Acute Myeloid Leukemia. Clinical oncohematology. 2017;10(2):227–34 (In Russ).

DOI: 10.21320/2500-2139-2017-10-2-227-234


ABSTRACT

The review presents data on the diagnostic and prognostic value of cytogenetic and molecular genetic markers of acute myeloid leukemia (AML). It demonstrates that some cases, different types of AML subdivided on the basis of clinical and morphological characteristics earlier may be distinguished based on identification of specific genetic and chromosomal defects. However, some repeated chromosomal abnormalities may be detected in AML patients that may be assigned to different variants based in clinical and morphocytochemical signs. At present, it is widely accepted that changes in the karyotype are the key prognostic factors which are more important than criteria based on morphological and cytochemical signs. Therefore, the risk-adaptive therapy of AML should be chosen based on the cytogenetic test findings. The review contains a section discussing gene mutations known to date that may affect the AML treatment outcome.

Keywords: AML, chromosomal aberration, chimeric oncogene, gene expression, gene mutation.

Received: September 16, 2016

Accepted: January 3, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. 776 с.
    [Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. 776 p. (In Russ)]
  2. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  3. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937–51. doi: 10.1182/blood-2009-03-209262.
  4. Zerbini MCN, Soares FA, Velloso EDRP, et al. World Health Organization classification of tumors of hematopoietic and lymphoid tissues, 2008: major changes from the 3rd edition. Revista da Associacao Medica Brasileira. 2011;57(1): 6–73. doi: 10.1590/S0104-42302011000100019.
  5. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br J Haematol. 1976;33(4):451–8. doi: 10.1111/j.1365-2141.1976.tb03563.
  6. Kuhnl A, Grimwade D. Molecular markers in acute myeloid leukaemia. Int J Hematol. 2012;96(2):153–63. doi: 10.1007/s12185-012-1123-9.
  7. Burnett AK, Wheatley K, Goldstone AH, et al. The value of allogeneic bone marrow transplant in patients with acute myeloid leukaemia at differing risk of relapse: results of the UK MRC AML 10 trial. Br J Haematol. 2002;118(2):385–400. doi: 10.1046/j.1365-2141.2002.03724.x.
  8. Cornelissen JJ, van Putten WL, Verdonck LF, et al. Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom? Blood. 2007;109(9):3658–66. doi: 10.1182/blood-2006-06-025627.
  9. Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood. 2000;96(13):4075–83.
  10. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116(3):354–65. doi: 10.1182/blood-2009-11-254441.
  11. Blum W, Mrozek K, Ruppert AS, et al. Adult de novo acute myeloid leukemia with t(6;11)(q27;q23): results from Cancer and Leukemia Group B Study 8461 and review of the literature. Cancer. 2005;103(6):1316. doi: 10.1002/cncr.20931
  12. Krauter J, Wagner K, Schafer I, et al. Prognostic factors in adult patients up to 60 years old with acute myeloid leukemia and translocations of chromosome band 11q23: individual patient data-based meta-analysis of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol. 2009;27(18):3000–6. doi: 10.1200/jco.2008.16.7981.
  13. von Neuhoff C, Reinhardt D, Sander A, et al. Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol. 2010;28(16):2682–9. doi: 10.1200/JCO.2009.25.6321.
  14. Rucker FG, Bullinger L, Schwaenen C, et al. Disclosure of candidate genes in acute myeloid leukemia with complex karyotypes using microarray-based molecular characterization. J Clin Oncol. 2006;24(24):3887–94. doi: 10.1200/jco.2005.04.5450.
  15. Mrozek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;35(4):365–77. doi: 10.1053/j.seminoncol.2008.04.007.
  16. Breems DA, Van Putten WL, De Greef GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol. 2008;26(29):4791–7. doi: 10.1200/JCO.2008.16.0259.
  17. Smith ML, Hills RK, Grimwade D. Independent prognostic variables in acute myeloid leukaemia. Blood Rev. 2011;25(1):39–51. doi: 10.1016/j.blre.2010.10.002.
  18. Delhommeau F, Dupont S, Della Valle V, et al. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360(22):2289–301. doi: 10.1056/NEJMoa0810069.
  19. Metzeler KH, Maharry K, Radmacher MD, et al. TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2011;29(10):1373–81. doi: 10.1200/JCO.2010.32.7742.
  20. Chou WC, Chou SC, Liu CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood. 2011;118(14):3803–10. doi: 10.1182/blood-2011-02-339747.
  21. Mardis ER, Ding L, Dooling DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361(11):1058–66.
  22. Thol F, Damm F, Wagner K, et al. Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia. Blood. 2010(4);116:614–6. doi: 10.1182/blood-2010-03-272146.
  23. Chou WC, Hou HA, Chen CY, et al. Distinct clinical and biologic characteristics in adult acute myeloid leukemia bearing the isocitrate dehydrogenase 1 mutation. Blood. 2010;115(14):2749–54. doi: 10.1182/blood-2009-11-253070.
  24. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/jco.2009.27.3730.
  25. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol. 2010;28(22):3636–43. doi: 10.1200/jco.2010.28.3762.
  26. Schnittger S, Haferlach C, Ulke M, et al. IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status. Blood. 2010;116(25):5486–96. doi: 10.1182/blood-2010-02-267955.
  27. Ravandi F, Patel K, Luthra R, et al. Prognostic significance of alterations in IDH enzyme isoforms in patients with AML treated with high-dose cytarabine and idarubicin. Cancer. 2012;118(10):2665–73. doi: 10.1002/cncr.26580.
  28. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33. doi: 10.1056/NEJMoa1005143.
  29. Thol F, Damm F, Ludeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol. 2011;29:2889–96. doi: 10.1200/JCO.2011.35.4894.
  30. Shen Y, Zhu YM, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood. 2011;118(20):5593–603. doi: 10.1182/blood-2011-03-343988.
  31. Hou HA, Kuo YY, Liu CY, et al. DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood. 2011(2);119:559–68. doi: 10.1182/blood-2011-07-369934.
  32. Renneville A, Boissel N, Nibourel O, et al. Prognostic significance of DNA methyltransferase 3A mutations in cytogenetically normal acute myeloid leukemia: a study by the Acute Leukemia French Association. Leukemia. 2012;26(6):1247–54. doi: 10.1038/leu.2011.382.
  33. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2012;30(7):742–50. doi: 10.1200/jco.2011.39.2092.
  34. Markova J, Michkova P, Burckova K, et al. Prognostic impact of DNMT3A mutations in patients with intermediate cytogenetic risk profile acute myeloid leukemia. Eur J Haematol. 2012;88(2):128–35. doi: 10.1111/j.1600-0609.2011.01716.x.
  35. King-Underwood L, Renshaw J, Pritchard-Jones K. Mutations in the Wilms’ tumor gene WT1 in leukemias. Blood. 1996;87(6):2171–9.
  36. Virappane P, Gale R, Hills R, et al. Mutation of the Wilms’ tumor 1 gene is a poor prognostic factor associated with chemotherapy resistance in normal karyotype acute myeloid leukemia: the United Kingdom Medical Research Council Adult Leukaemia Working Party. J Clin Oncol. 2008;26(33):5429–35. doi: 10.1200/jco.2008.16.0333.
  37. Paschka P, Marcucci G, Ruppert AS, et al. Wilms’ tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study. J Clin Oncol. 2008;26(28):4595–602. doi: 10.1200/JCO.2007.15.2058.
  38. Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia. 2006;20(6):965–70. doi: 10.1038/sj.leu.2404188.
  39. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24(24):3904–11. doi: 10.1200/JCO.2006.06.9500.
  40. Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood. 2006;107(9):3463–8. doi: 10.1182/blood-2005-09-3640.
  41. Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107(5):1791–9. doi: 10.1182/blood-2005-04-1466.
  42. Gelsi-Boyer V, Trouplin V, Adelaide J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800. doi: 10.1111/j.1365-2141.2009.07697.x.
  43. Chou WC, Huang HH, Hou HA, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood. 2010;116(20):4086–94. doi: 10.1182/blood-2010-05-283291.
  44. Metzeler KH, Becker H, Maharry K, et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category. Blood. 2011;118(26):6920–9. doi: 10.1182/blood-2011-08-368225.
  45. Pratcorona M, Abbas S, Sanders MA, et al. Acquired mutations in ASXL1 in acute myeloid leukemia: prevalence and prognostic value. Haematologica. 2012;97(3):388–92. doi: 10.3324/haematol.2011.051532.
  46. Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/NEJMoa1112304.
  47. Grossmann V, Tiacci E, Holmes AB, et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood. 2011;118(23):6153–63. doi: 10.1182/blood-2011-07-365320.
  48. Li M, Collins R, Jiao Y, et al. Somatic mutations in the transcriptional corepressor gene BCORL1 in adult acute myelogenous leukemia. Blood. 2011;118(22):5914–7. doi: 10.1182/blood-2011-05-356204.
  49. Van Vlierberghe P, Patel J, Abdel-Wahab O, et al. PHF6 mutations in adult acute myeloid leukemia. Leukemia. 2011;25(1):130–4. doi: 10.1038/leu.2010.247.
  50. Mano H. Stratification of acute myeloid leukemia based on gene expression profiles. Int J Hematol. 2004;80(5):389–94. doi: 10.1532/ijh97.04111.
  51. Marcucci G, Mrozek K, Radmacher MD, et al. The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood. 2011;117(4):1121–9. doi: 10.1182/blood-2010-09-191312.
  52. Smith ML, Hills RK, Grimwade D. Independent prognostic variables in acute myeloid leukaemia. Blood Rev. 2011;25(1):39–51. doi: 10.1016/j.blre.2010.10.002.
  53. Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352(3):254–66. doi: 10.1056/NEJMoa041974.
  54. Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106(12):3740–6. doi: 10.1182/blood-2005-05-2164.
  55. Thiede C, Creutzig E, Illmer T, et al. Rapid and sensitive typing of NPM1 mutations using LNA-mediated PCR clamping. Leukemia. 2006;20(10):1897–9. doi: 10.1038/sj.leu.2404367.
  56. Schlenk RF, Dohner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–18. doi: 10.1056/NEJMoa074306.
  57. Green CL, Koo KK, Hills RK, et al. Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations. J Clin Oncol. 2010;28(16):2739–47. doi: 10.1200/JCO.2009.26.2501.
  58. Grisendi S, Mecucci C, Falini B, et al. Nucleophosmin and cancer. Nat Rev Cancer. 2006;6(7):493–505. doi: 10.1038/nrc1885.
  59. Freeman SD, Jovanovic JV, Grimwade D. Development of minimal residual disease-directed therapy in acute myeloid leukemia. Semin Oncol. 2008;35(4):388–400. doi: 10.1053/j.seminoncol.2008.04.009.
  60. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10(12):1911–8.
  61. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9. doi: 10.1182/blood.V98.6.1752.
  62. Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99(12):4326–35. doi: 10.1182/blood.V99.12.4326.
  63. Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis. Leukemia. 2005;19(8):1345–9. doi: 10.1038/sj.leu.2403838.
  64. Mead AJ, Linch DC, Hills RK, et al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients. Blood. 2007;110(4):1262–70. doi: 10.1182/blood-2006-04-015826.
  65. Bacher U, Haferlach C, Kern W, et al. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters—an analysis of 3082 patients. Blood. 2008;111(5):2527–37. doi: 10.1182/blood-2007-05-091215.
  66. Whitman SP, Ruppert AS, Radmacher MD, et al. FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood. 2008;111(3):1552–9. doi: 10.1182/blood-2007-08-107946.
  67. Gale RE, Hills R, Pizzey AR, et al. Relationship between FLT3 mutation status, biologic characteristics, and response to targeted therapy in acute promyelocytic leukemia. Blood. 2005;106(12):3768–76. doi: 10.1182/blood-2005-04-1746.
  68. Souza Melo CP, Campos CB, Dutra AP, et al. Correlation between FLT3-ITD status and clinical, cellular and molecular profiles in promyelocytic acute leukemias. Leuk Res. 2015;39(2):131–7. doi: 10.1016/j.leukres.2014.11.010.
  69. Cicconi L, Divona M, Ciardi C, et al. PML-RARα kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy. Leukemia. 2016;30(10):1987–92. doi: 10.1038/leu.2016.122.
  70. Radomska HS, Huettner CS, Zhang P, et al. CCAAT/enhancer binding protein alpha is a regulatory switch sufficient for induction of granulocytic development from bipotential myeloid progenitors. Mol Cell Biol. 1998;18(7):4301–14. doi: 10.1128/mcb.18.7.4301.
  71. Pabst T, Mueller BU, Zhang P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia. Nat Genet. 2001;27(3):263–70. doi: 10.1038/85820.
  72. Smith ML, Cavenagh JD, Lister TA, et al. Mutation of CEBPA in familial acute myeloid leukemia. N Engl J Med. 2004;351(23):2403–7. doi: 10.1056/NEJMoa041331.
  73. Pabst T, Eyholzer M, Fos J, et al. Heterogeneity within AML with CEBPA mutations; only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis. Br J Cancer. 2009;100(8):1343–6. doi: 10.1038/sj.bjc.6604977.
  74. Kirstetter P, Schuster MB, Bereshchenko O, et al. Modeling of C/EBPalpha mutant acute myeloid leukemia reveals a common expression signature of committed myeloid leukemia-initiating cells. Cancer Cell. 2008;13(4):299–310. doi: 10.1016/j.ccr.2008.02.008.
  75. Wouters BJ, Lowenberg B, Erpelinck-Verschueren CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood. 2009;113(13):3088–91. doi: 10.1182/blood-2008-09-179895.
  76. Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact, concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood. 2011;117(8):2469–75. doi: 10.1182/blood-2010-09-307280.
  77. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.
  78. Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 2011;333(6047):1300–3. doi: 10.1126/science.1210597.
  79. Abdel-Wahab O, Mullally A, Hedvat C, et al. Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood. 2009;114(1):144–7. doi: 10.1182/blood-2009-03-210039.
  80. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–67. doi: 10.1016/j.ccr.2010.11.015.
  81. Ko M, Huang Y, Jankowska AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010;468(7325):839–43. doi: 10.1038/nature09586.
  82. Langemeijer SM, Kuiper RP, Berends M, et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet. 2009;41(7):838–42. doi: 10.1038/ng.391.
  83. Jankowska AM, Szpurka H, Tiu RV, et al. Loss of heterozygosity 4q24 and TET2 mutations associated with myelodysplastic/myeloproliferative neoplasms. Blood. 2009;113(25):6403–10. doi: 10.1182/blood-2009-02-205690.
  84. Gaidzik VI, Paschka P, Spath D, et al. TET2 mutations in acute myeloid leukemia (AML): results from a comprehensive genetic and clinical analysis of the AML Study Group. J Clin Oncol. 2012;30(12):1350–7. doi: 10.1200/JCO.2011.39.2886.
  85. Ward PS, Patel J, Wise DR, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010;17(3):225–34. doi: 10.1016/j.ccr.2010.01.020.
  86. Gross S, Cairns RA, Minden MD, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010;207(2):339–44. doi: 10.1084/jem.20092506.
  87. Sanz MA, Grimwade D, Tallman MS, et al. Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2009;113(9):1875–91. doi: 10.1182/blood-2008-04-150250.
  88. Diehl F, Rossig L, Zeiher AM, et al. The histone methyltransferase MLL is an upstream regulator of endothelial-cell sprout formation. Blood. 2007;109(4):1472–8. doi: 10.1182/blood-2006-08-039651.
  89. Li Y, Han J, Zhang Y, et al. Structural basis for activity regulation of MLL family methyltransferases. Nature. 2016;530(7591):447–52. doi: 10.1038/nature16952.
  90. Bower M, Parry P, Carter M, et al. Prevalence and clinical correlations of MLL gene rearrangements in AML-M4/5. Blood. 1994;84(11):3776–80.
  91. Schichman SA, Caligiuri MA, Strout MP, et al. ALL-1 tandem duplication in acute myeloid leukemia with a normal karyotype involves homologous recombination between Alu elements. Cancer Res. 1994;54(16):4277–80.
  92. Caligiuri MA, Schichman SA, Strout MP, et al. Molecular rearrangement of the ALL-1 gene in acute myeloid leukemia without cytogenetic evidence of 11q23 chromosomal translocations. Cancer Res. 1994;54(2):370–3.
  93. Park JP, Ladd SL, Ely P, et al. Amplification of the MLL region in acute myeloid leukemia. Cancer Genet Cytogenet. 2000;121(2):198–205. doi: 10.1016/S0165-4608(00)00256-9.
  94. Schnittger S, Kinkelin U, Schoch C, et al. Screening for MLL tandem duplication in 387 unselected patients with AML identify a prognostically unfavorable subset of AML. Leukemia. 2000;14(5):796–804. doi: 10.1038/sj.leu.2401773.
  95. Slovak ML, Traweek ST, Willman CL, et al. Trisomy 11: an association with stem/progenitor cell immunophenotype. Br J Haematol. 1995;90(2):266–73. doi: 10.1111/j.1365-2141.1995.tb05146.x.
  96. Strout MP, Marcucci G, Bloomfield CD, et al. The partial tandem duplication of ALL1 (MLL) is consistently generated by Alu-mediated homologous recombination in acute myeloid leukemia. Proc Natl Acad Sci USA. 1998;95(5):2390–5. doi: 10.1073/pnas.95.5.2390.
  97. Klymenko S, Bebeshko V, Bazyka D, et al. AML1 gene rearrangements and mutations in radiation-associated acute myeloid leukemia and myelodysplastic syndromes. J Rad Res. 2005;46(2):249–55. doi: 10.1269/jrr.46.249.
  98. Мисюрин В.А., Лукина А.Е., Мисюрин А.В. и др. Особенности соотношения уровней экспрессии генов PRAME и PML/RARA в дебюте острого промиелоцитарного лейкоза. Российский биотерапевтический журнал. 2014;13(1):9–16.
    [Misyurin VA, Lukina AE, Misyurin AV. A ratio between gene expression levels of PRAME and PML/RARA at the onset of acute promyelocytic leukemia and clinical features of the disease. Rossiiskii bioterapevticheskii zhurnal. 2014;13(1):9–16. (In Russ)]
  99. Мисюрин А.В. Основы молекулярной диагностики онкогематологических заболеваний. Российский биотерапевтический журнал. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24.
    [Misyurin AV. Fundamentals of the molecular diagnosis of oncohematological diseases. Rossiiskii bioterapevticheskii zhurnal. 2016;15(4):18–24. doi: 10.17650/1726-9784-2016-15-4-18-24. (In Russ)]

 

 

Myeloid-Derived Suppressor Cells in Some Oncohematological Diseases

AV Ponomarev

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

For correspondence: Aleksandr Vasil’evich Ponomarev, graduate student, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: kl8546@yandex.ru

For citation: Ponomarev AV. Myeloid-Derived Suppressor Cells in Some Oncohematological Diseases. Clinical oncohematology. 2017;10(1):29-38–хх (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-29-38


ABSTRACT

Myeloid-derived suppressor cells are immature myeloid cells with immunosuppressive properties. The review presents characteristics of myeloid-derived suppressor cells. It includes phenotype variants, mechanisms of the suppressive effect on the immune system, and tumor recruitment mechanisms of myeloid suppressors. It provides a brief description of works which studied myeloid suppressor in oncohematological diseases including multiple myeloma, lymphomas, and leukemias.

Keywords: myeloid suppressors, myeloid-derived suppressor cells, multiple myeloma, lymphomas, leukemias.

Received: September 8, 2016

Accepted: December 3, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Тупицына Д.Н., Ковригина А.М., Тумян Г.С. и др. Клиническое значение внутриопухолевых FOXP3+ Т-регуляторных клеток при солидных опухолях и фолликулярных лимфомах: обзор литературы и собственные данные. Клиническая онкогематология. 2012;(5)3:193–203.
    [Tupitsyna DN, Kovrigina AM, Tumian GS, et al. Different clinical meaning of intratumoral FOXP3+ T-regulatory cells in solid tumors and follicular lymphomas: literature review and own data. Klinicheskaya onkogematologiya. 2012;(5)3:193–203. (In Russ)]
  2. Кадагидзе З.Г., Черткова А.И., Славина Е.Г. NKT-клетки и противоопухолевый иммунитет. Российский биотерапевтический журнал. 2011;10(3):9–16.
    [Kadagidze ZG, Chertkova AI, Slavina EG. NKT-cells and antitumor immunity. Rossiiskii bioterapevticheskii zhurnal. 2011;10(3):9–16. (In Russ)]
  3. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev 2012;12(4):253–68. doi: 10.1038/nri3175.
  4. Gabrilovich DI, Bronte V, Chen S-H, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007;67(1):425– doi: 10.1158/0008-5472.CAN-06-3037.
  5. Bowen JL, Olson JK. Innate immune CD11b+Gr-1+ cells, suppressor cells, affect the immune response during Theiler’s virus-induced demyelinating disease. J Immunol. 2009;183(11):6971–80. doi: 10.4049/jimmunol.0902193.
  6. Tsiganov EN, Verbina EM, Radaeva TV, et al. Gr-1dim CD11b+ immature myeloid-derived suppressor cells but not neutrophils are markers of lethal tuberculosis infection in mice. J Immunol. 2014;192(10):4718–27. doi: 10.4049/jimmunol.1301365.
  7. Delano MJ, Scumpia PO, Weinstein JS, et al. MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J Exp Med. 2007;204(6):1463–74.
  8. Гапонов М.А., Хайдуков С.В., Писарев В.М. и др. Субпопуляционная гетерогенность миелоидных иммуносупрессорных клеток у пациентов с септическими состояниями. Российский иммунологический журнал. 2015;9(18):11–14.
    [Gaponov MA, Khaidukov SV, Pisarev VM, et al. Subpopulation heterogeneity of immunosuppressive myeloid cells in patients with sepsis. Rossiiskii immunologicheskii zhurnal. 2015;9(18):11–14. (In Russ)]
  9. Makarenkova VP, Bansal V, Matta BM, et al. CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic stress. J Immunol. 2006;176(4):2085–94. doi: 10.4049/jimmunol.176.4.2085.
  10. Greten TF, Manns MP, Korangy F. Myeloid derived suppressor cells in human diseases. Int. 2011;11(7):802–7. doi: 10.1016/j.intimp.2011.01.003.
  11. Diaz-Montero CM, Salem ML, Nishimura MI, et al. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin–cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58(1):49–59. doi: 10.1007/s00262-008-0523-
  12. Yazdani Y, Mohammadnia-Afrouzi M, Yousefi M, et al. Myeloid-derived suppressor cells in B cell malignancies. Tumour Biol. 2015;36(10):7339–53. doi: 10.1007/s13277-015-4004-z.
  13. Пономарев А.В. Миелоидные супрессорные клетки: общая характеристика. Иммунология. 2016;37(1):47–50. doi: 10.18821/0206-4952-2016-37-1-47-50.
    [Ponomarev AV. Myeloid suppressor cells: general characteristics. Immunologiya. 2016;37(1):47–50. doi: 10.18821/0206-4952-2016-37-1-47- (In Russ)]
  14. Gabrilovich DI, Nagaraj S. Myeloid-derived-suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–74. doi: 10.1038/nri2506.
  15. Lechner MG, Megiel C, Russell SM, et al. Functional characterization of human Cd33+ And Cd11b+ myeloid-derived suppressor cell subsets induced from peripheral blood mononuclear cells co-cultured with a diverse set of human tumor cell lines. J Transl 2011;9(1):90. doi: 10.1186/1479-5876-9-90.
  16. Rodriguez PC, Ernstoff MS, Hernandez C, et al. Arginase I–Producing Myeloid-Derived Suppressor Cells in Renal Cell Carcinoma Are a Subpopulation of Activated Granulocytes. Cancer Res. 2009;69(4):1553–60.
  17. Schmielau J, Finn OJ. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of T-cell function in advanced cancer patients. Cancer Res. 2001;61(12):4756–60.
  18. Youn J-I, Collazo M, Shalova I, et al. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J Leuk 2012;91(1):167–81. doi: 10.1189/jlb.0311177.
  19. Youn J-I, Nagaraj S, Collazo M, et al. Subsets of Myeloid-Derived Suppressor Cells in Tumor Bearing Mice. J Immunol. 2008;181(8):5791–802. doi: 10.4049/jimmunol.181.8.5791.
  20. Corzo CA, Condamine T, Lu L, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439–53. doi: 10.1084/jem.20100587.
  21. Yang L, DeBusk LM, Fukuda K, et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6(4):409–21. doi: 10.1016/j.ccr.2004.08.031.
  22. Zhuang J, Zhang J, Lwin ST, et al. Osteoclasts in multiple myeloma are derived from Gr-1+CD11b+ myeloid-derived suppressor cells. PLoS One. 2012;7(11):e48871. doi: 1371/journal.pone.0048871.
  23. Choi J, Suh B, Ahn YO, et al. CD15+/CD16low human granulocytes from terminal cancer patients: granulocytic myeloid-derived suppressor cells that have suppressive function. Tumour Biol. 2012;33(1):121–9. doi: 10.1007/s13277-011-0254-
  24. Stanojevic I, Miller K, Kandolf-Sekulovic L, et al. A subpopulation that may correspond to granulocytic myeloid-derived suppressor cells reflects the clinical stage and progression of cutaneous melanoma. Int Immunol. 2016;28(2):87–97. doi: 10.1093/intimm/dxv053.
  25. Saiwai H, Kumamaru H, Ohkawa Y, et al. Ly6C+Ly6G– Myeloid-derived suppressor cells play a critical role in the resolution of acute inflammation and the subsequent tissue repair process after spinal cord injury. J Neurochem. 2013;125(1):74–88. doi: 10.1111/jnc.12135.
  26. Rodriguez PC, Augusto CO. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol 2008;222(1):180–91. doi: 10.1111/j.1600-065X.2008.00608.x.
  27. Srivastava MK, Sinha P, Clements VK, et al. Myeloid-derived suppressor cells inhibit T cell activation by depleting cystine and cysteine. Cancer Res. 2010;70(1):68–77. doi: 10.1158/0008-CAN-09-2587.
  28. Chevolet I, Speeckaert R, Schreuer M, et al. Characterization of the in vivo immune network of IDO, tryptophan metabolism, PD-L1, and CTLA-4 in circulating immune cells in melanoma. Oncoimmunology. 2015;4(3):e982382. doi: 10.4161/2162402X.2014.982382.
  29. Jitschin R, Braun M, Buttner M, et al. CLL-cells induce IDOhiCD14+HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote Tregs. Blood. 2014;124(5):750–60. doi: 10.1182/blood-2013-12-
  30. Nagaraj S, Gupta K, Pisarev V, et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13(7):828–35. doi: 10.1038/nm1609.
  31. Lu T, Ramakrishnan R, Altiok S, et al. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J Clin 2011;121(10):4015–4029. doi: 10.1172/JCI45862.
  32. Hanson EM, Clements VK, Sinha P, et al. Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J. Immunol. 2009;183(2):937–44. doi: 10.4049/jimmunol.0804253.
  33. Noman MZ, Desantis G, Janji B, et al. PD-L1 is a novel direct target of HIF-1a, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90. doi: 10.1084/jem.20131916.
  34. Filipazzi P, Valenti R, Huber V, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol. 2007;25(18):2546–53. doi: 10.1200/JCO.2006.08.5829.
  35. Sinha P, Clements VK, Bunt SK, et al. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol. 2007;179(2):977–83. doi: 10.4049/jimmunol.179.2.977.
  36. Li H, Han Y, Guo Q, et al. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol. 2009;182(1):240–9. doi: 10.4049/jimmunol.182.1.240.
  37. Liu C, Yu S, Kappes J, et al. Expansion of spleen myeloid suppressor cells represses NK cell cytotoxicity in tumor-bearing host. Blood. 2007;109(10):4336–42. doi: 10.1182/blood-2006-09-
  38. Elkabets M, Ribeiro VSG, Dinarello CA, et al. IL-1b regulates a novel myeloid-derived suppressor cell subset that impairs NK cell development and function. Eur J Immunol. 2010;40(12):3347–57. doi: 10.1002/eji.201041037.
  39. Hoechst B, Voigtlaender T, Ormandy L, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50(3):799–807. doi: 10.1002/hep.23054.
  40. Pan PY, Ma G, Weber KJ, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70(1):99–108. doi: 10.1158/0008-CAN-09-1882.
  41. Hoechst B, Gamrekelashvili J, Manns MP, et al. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood. 2011;117(24):6532–41. doi: 10.1182/blood-2010-11-
  42. Shojaei F, Wu X, Malik AK, et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25(8):911–20. doi: 10.1038/nbt1323.
  43. Connolly MK, Mallen-St Clair J, Bedrosian AS, et al. Distinct populations of metastases-enabling myeloid cells expand in the liver of mice harboring invasive and preinvasive intra-abdominal tumor. J Leuk Biol. 2010;87(4):713–25. doi: 10.1189/jlb.0909607.
  44. Yang L, Huang J, Ren X, et al. Abrogation of TGFb signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13(1):23–35. doi: 10.1016/j.ccr.2007.12.004.
  45. Giles A, Vicioso Y, Kasai M, et al. Bone marrow-derived progenitor cells develop into myeloid-derived suppressor cells at metastatic sites. J Immunother Cancer. 2013;1(Suppl 1):188. doi: 10.1186/2051-1426-1-S1-P188.
  46. Solito S, Falisi E, Diaz-Montero CM, et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood. 2011;118(8):2254–65. doi: 10.1182/blood-2010-12-
  47. Marigo I, Bosio E, Solito S, et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity. 2010;32(6):790–802. doi: 10.1016/j.immuni.2010.05.010.
  48. Highfill SL, Rodriguez PC, Zhou Q, et al. Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13. Blood. 2010;116(25):5738–47. doi: 10.1182/blood-2010-06-
  49. Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol. 2010;185(4):2273–84. doi: 10.4049/jimmunol.1000901.
  50. Atretkhany KS, Nosenko MA, Gogoleva VS, et al. TNF Neutralization Results in the Delay of Transplantable Tumor Growth and Reduced MDSC Accumulation. Front Immunol. 2016;7:147. doi: 10.3389/fimmu.2016.00147.
  51. De Veirman K, Van Valckenborgh E, Lahmar Q, et al. Myeloid-derived suppressor cells as therapeutic target in hematological malignancies. Front Oncol. 2014;4:349. doi: 10.3389/fonc.2014.00349.
  52. Ramachandran I, Martner A, Pisklakova A, et al. Myeloid-derived suppressor cells regulate growth of multiple myeloma by inhibiting T cells in bone marrow. J Immunol. 2013;190(7):3815–23. doi: 10.4049/jimmunol.1203373.
  53. De Veirman K, Van Ginderachter JA, Lub S, et al. Multiple myeloma induces Mcl-1 expression and survival of myeloid-derived suppressor cells. Oncotarget. 2015;6(12):10532–47. doi: 10.18632/oncotarget.3300.
  54. Brimnes MK, Vangsted AJ, Knudsen LM, et al. Increased level of both CD4+FOXP3+ regulatory T cells and CD14+HLA-DR/low myeloid-derived suppressor cells and decreased level of dendritic cells in patients with multiple myeloma. Scand J Immunol. 2010;72(6):540–7. doi: 10.1111/j.1365-2010.02463.x.
  55. Gorgun GT, Whitehill G, Anderson JL, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121(15):2975–87. doi: 10.1182/blood-2012-08-
  56. Gorgun GТ, Samur MK, Cowens KB, et al. Lenalidomide Enhances Immune Checkpoint Blockade-Induced Immune Response in Multiple Myeloma. Clin Cancer Res. 2015;21(20):4607–18. doi: 10.1158/1078-CCR-15-0200.
  57. Busch A, Zeh D, Janzen V, et al. Treatment with lenalidomide induces immuno-activating and counter-regulatory immunosuppressive changes in myeloma patients. Clin Exp Immunol. 2014;177(2):439–53. doi: 10.1111/cei.12343.
  58. Wang Z, Zhang L, Wang H, et al. Tumor-induced CD14+HLA-DR (-/low) myeloid-derived suppressor cells correlate with tumor progression and outcome of therapy in multiple myeloma patients. Cancer Immunol Immunother. 2015;64(3):389–99. doi: 10.1007/s00262-014-1646-
  59. De Keersmaecker B, Fostier K, Corthals J, et al. Immunomodulatory drugs improve the immune environment for dendritic cell-based immunotherapy in multiple myeloma patients after autologous stem cell transplantation. Cancer Immunol Immunother. 2014;63(10):1023–36. doi: 10.1007/s00262-014-1571-
  60. Castella B, Foglietta M, Sciancalepore P, et al. Anergic bone marrow Vg9Vd2 T cells as early and long-lasting markers of PD-1-targetable microenvironment-induced immune suppression in human myeloma. Oncoimmunology. 2015;4(11):e1047580. doi: 10.1080/2162402X.2015.1047580.
  61. Giallongo C, Tibullo D, Parrinello NL, et al. Granulocyte-like myeloid derived suppressor cells (G-MDSC) are increased in multiple myeloma and are driven by dysfunctional mesenchymal stem cells (MSC). Oncotarget. 2016;7(52):85764– doi: 10.18632/oncotarget.7969.
  62. Lee SE, Lim JY, Ryu DB, et al. Circulating immune cell phenotype can predict the outcome of lenalidomide plus low-dose dexamethasone treatment in patients with refractory/relapsed multiple myeloma. Cancer Immunol Immunother. 2016;65(8):983–94. doi: 10.1007/s00262-016-1861-
  63. Favaloro J, Liyadipitiya T, Brown R, et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk Lymphoma. 2014;55(12):2893–900. doi: 10.3109/10428194.2014.904511.
  64. Franssen LE, van de Donk NW, Emmelot ME, et al. The impact of circulating suppressor cells in multiple myeloma patients on clinical outcome of DLIs. Bone Marrow Transplant. 2015;50(6):822–8. doi: 10.1038/bmt.2015.48.
  65. Lin Y, Gustafson MP, Bulur PA, et al. Immunosuppressive CD14+HLA-DRlow/– monocytes in B-cell non-Hodgkin lymphoma. Blood. 2011;117(3):872–81. doi: 10.1182/blood-2010-05-
  66. Tadmor T, Fell R, Polliack A, et al. Absolute monocytosis at diagnosis correlates with survival in diffuse large B-cell lymphoma—possible link with monocytic myeloid-derived suppressor cells. Hematol 2013;31(2):65–71. doi: 10.1002/hon.2019.
  67. Gustafson MP, Lin Y, LaPlant B, et al. Immune monitoring using the predictive power of immune profiles. J Immunother Cancer. 2013;1(1):7. doi: 10.1186/2051-1426-1-7.
  68. Wu C, Wu X, Zhang X, et al. Prognostic significance of peripheral monocytic myeloid-derived suppressor cells and monocytes in patients newly diagnosed with diffuse large B-cell lymphoma. Int J Clin Exp Med. 2015;8(9):15173–81.
  69. Sato Y, Shimizu K, Shinga J, et al. Characterization of the myeloid-derived suppressor cell subset regulated by NK cells in malignant lymphoma. Oncoimmunology. 2015;4(3):e995541. doi: 10.1080/2162402X.2014.995541.
  70. Romano A, Parrinello NL, Vetro C, et al. Circulating myeloid-derived suppressor cells correlate with clinical outcome in Hodgkin Lymphoma patients treated up-front with a risk-adapted strategy. Br J Haematol. 2015;168(5):689–700. doi: 10.1111/bjh.13198.
  71. Marini O, Spina C, Mimiola E, et al. Identification of granulocytic myeloid-derived suppressor cells (G-MDSCs) in the peripheral blood of Hodgkin and non-Hodgkin lymphoma patients. Oncotarget. 2016;19(7):27677–88. doi: 10.18632/oncotarget.8507.
  72. Azzaoui I, Uhel F, Rossille D, et al. T-cell defect in diffuse large B-cell lymphomas involves expansion of myeloid derived suppressor cells expressing IL-10, PD-L1 and S100A12. Blood. 2016;128(8):1081–92. doi: 10.1182/blood-2015-08-
  73. Zhang H, Li ZL, Ye SB, et al. Myeloid-derived suppressor cells inhibit T cell proliferation in human extranodal NK/T cell lymphoma: a novel prognostic indicator. Cancer Immunol Immunother. 2015;64(12):1587- doi: 10.1007/s00262-015-1765-6.
  74. Christiansson L, Sоderlund S, Svensson E, et al. Increased Level of Myeloid-Derived Suppressor Cells, Programmed Death Receptor Ligand 1/Programmed Death Receptor 1, and Soluble CD25 in Sokal High Risk Chronic Myeloid Leukemia. PLoS One. 2013;8(1):e55818. doi: 10.1371/journal.pone.0055818.
  75. Giallongo C, Romano A, Parrinello NL, et al. Mesenchymal Stem Cells (MSC) Regulate Activation of Granulocyte-Like Myeloid Derived Suppressor Cells (G-MDSC) in Chronic Myeloid Leukemia Patients. PLoS One. 2016;11(7):e0158392. doi: 10.1371/journal.pone.0158392.
  76. Gustafson МP, Abraham RS, Lin Y, et al. Association of an increased frequency of CD14+HLA-DRlo/neg monocytes with decreased time to progression in chronic lymphocytic leukaemia (CLL). Br J Haematol. 2012;156(5):674–6. doi: 10.1111/j.1365-2011.08902.x.
  77. Liu J, Zhou Y, Huang Q, et al. CD14+HLA-DRlow/– expression: a novel prognostic factor in chronic lymphocytic leukemia. Oncol 2015;9(3):1167–72. doi: 10.3892/ol.2014.2808.
  78. Sun H, Li Y, Zhang ZF, et al. Increase in myeloid-derived suppressor cells (MDSCs) associated with minimal residual disease (MRD) detection in adult acute myeloid leukemia. Int J Hematol. 2015;102(5):579–86. doi: 10.1007/s12185-015-1865-
  79. Gleason MK, Ross JA, Warlick ED, et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood. 2014;123(19):3016–26. doi: 10.1182/blood-2013-10-
  80. Chen X, Eksioglu EA, Zhou J, et al. Induction of myelodysplasia by myeloid-derived suppressor cells. J Clin Invest. 2013;123(11):4595–611. doi: 10.1172/JCI67580.
  81. Kittang AO, Kordasti S, Sand KE, et al. Expansion of myeloid derived suppressor cells correlates with number of T regulatory cells and disease progression in myelodysplastic syndrome. Oncoimmunology. 2015;5(2):e1062208. doi: 10.1080/2162402X.2015.1062208.
  82. Noonan KA, Ghosh N, Rudraraju L, et al. Targeting immune suppression with PDE5 inhibition in end-stage multiple myeloma. Cancer Immunol Res. 2014;2(8):725–31. doi: 10.1158/2326-CIR-13-0213.

 

 

Erythroferron: Modern Concepts of Its Role in Iron Metabolism Regulation

VT Sakhin1, NV Kremneva1, AV Gordienko2, OA Rukavitsyn3

1 Military Clinical Hospital No. 1586 under the Ministry of Defense of Russia, 4 Mashtakova str., Podol’sk, Moscow Oblast, Russian Federation, 142110

2 SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

3 NN Burdenko Principal Military Clinical Hospital under the Ministry of Defense of Russia, 3 Gospital’naya pl., Moscow, Russian Federation, 105229

For correspondence: Valerii Timofeevich Sakhin, PhD, 4 Mashtakova str., Podol’sk, Moscow Oblast, Russian Federation, 142110; Tel: +7(916)314-31-11; e-mail: SahinVT@yandex.ru

For citation: Sakhin VT, Kremneva NV, Gordienko AV, Rukavitsyn OA. Erythroferron: Modern Concepts of Its Role in Iron Metabolism Regulation. Clinical oncohematology. 2017;10(1):25–8 (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-25-28


ABSTRACT

The article presents the results of experimental and clinical studies evaluating the importance of supposed erythroid regulators of hepcidin levels and mechanism of their action. It demonstrates that the role of growth differentiation factor 15 and twisted gastrulation protein homolog 1 in regulation of hepcidin levels in humans has not been confirmed yet. The data confirming the importance of erythroferron in the pathogenesis of anemia related to blood loss, hemolysis, and hereditary anemias with ineffective erythropoiesis are presented. The studies demonstrated that erythroferron plays the greatest role in the regulation of hepcidin levels in pathological conditions and at stress and does not play a leading role in erythropoiesis under normal conditions. Erythroferron suppresses the hepcidin synthesis by affecting the liver cells directly through an unknown receptor cellular pathway.

Keywords: anemia of chronic disease, hepcidin, erythroferron.

Received: September 14, 2016

Accepted: November 13, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Гематология: национальное руководство. Под ред. О.А. Рукавицына. М.: ГЭОТАР-Медиа, 2015. С. 143–9.
    [Rukavitsyn OA, ed. Gematologiya: natsional’noe rukovodstvo. (Hematology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2015. pp. 143–9. (In Russ)]
  2. Ganz T, Nemeth E. Hepcidin and iron homeostasis. Biochim Biophys Acta. 2012;1823(9):1434–43. doi: 10.1016/j.bbamcr.2012.01.014.
  3. Nicolas G, Chauvet C, Viatte L, et al. The gene encoding the iron regulatory peptide hepcidinis regulated by anemia, hypoxia, and inflammation. J Clin Invest. 2002;110(7):1037–44. doi: 10.1172/jci0215686.
  4. Peyssonnaux C, Zinkernagel AS, Schuepbach RA, et al. Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs). J Clin Invest. 2007;117(7):1926–32. doi: 10.1172/jci31370.
  5. Pinto JP, Ribeiro S, Pontes H, et al. Erythropoietin mediates hepcidin expression in hepatocytes through EPOR signaling and regulation of C/EBP alpha. Blood. 2008;111(12):5727–33. doi: 10.1182/blood-2007-08-106195.
  6. Liu Q, Davidoff O, Niss K, et al. Hypoxia-inducible factor regulates hepcidin via erythropoietin-induced erythropoiesis. J Clin Invest. 2012;122(12):4635–44. doi: 10.1172/jci63924.
  7. Pak M, Lopez MA, Gabayan V, et al. Suppression of hepcidin during anemia requires erythropoietic activity. Blood. 2006;108(12):3730–5. doi: 10.1182/blood-2006-06-028787.
  8. Vokurka M, Krijt J, Sulc K, Necas E. Hepcidin mRNA levels in mouse liver respond to inhibition of erythropoiesis. Phys Res. 2006;55(6):667–74.
  9. Tanno T, Bhanu NV, Oneal PA, et al. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat Med. 2007;13(9):1096–101. doi: 10.1038/nm1629.
  10. Tanno T, Porayette P, Sripichai O, et al. Identification of TWSG1 as a second novel erythroid regulator of hepcidin expression in murine and human cells. Blood. 2009;114(1):181–6. doi: 10.1182/blood-2008-12-195503.
  11. Unsicker K, Spittau B, Krieglstein K. The multiple facets of the TGF-b family cytokine growth/differentiation factor-15/macrophage inhibitory cytokine-1. Cyt Growth Factor Rev. 2013;24(4):373–84. doi: 10.1016/j.cytogfr.2013.05.003.
  12. Forsman CL, Ng BC, Heinze RK, et al. BMP-binding protein twisted gastrulation is required in mammary gland epithelium for normal ductal elongation and myoepithelial compartmentalization. Devel Biol. 2013;373(1):95–106. doi: 10.1016/j.ydbio.2012.10.007.
  13. Frazer DM, Wilkins SJ, Darshan D, et al. Stimulated erythropoiesis with secondary iron loading leads to a decrease in hepcidin despite an increase in bone morphogenetic protein 6 expression. Br J Haematol. 2012;157(5):615–26. doi: 10.1111/j.1365-2141.2012.09104.x.
  14. Casanovas G, Spasic MV, Casu C, et al. The murine growth differentiation factor 15 is not essential for systemic iron homeostasis in phlebotomized mice. Haematologica. 2013;98(3):444–7. doi: 10.3324/haematol.2012.069807.
  15. Tamary H, Shalev H, Perez-Avraham G, et al. Elevated growth differentiation factor 15 expression in patients with congenital dyserythropoietic anemia type I. Blood. 2008;112(13):5241–4. doi: 10.1182/blood-2008-06-165738.
  16. An X, Schulz VP, Li J, et al. Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood. 2014;123(22):3466–77. doi: 10.1182/blood-2014-01-548305.
  17. Tanno T, Rabel A, Lee YT, et al. Expression of growth differentiation factor 15 is not elevated in individuals with iron deficiency secondary to volunteer blood donation. Transfusion. 2010;50(7):1532–5. doi: 10.1111/j.1537-2995.2010.02601.x.
  18. Theurl I, Finkenstedt A, Schroll A, et al. Growth differentiation factor 15 in anaemia of chronic disease, iron deficiency anaemia and mixed type anaemia. Br J Haematol. 2010;148(3):449–55. doi: 10.1111/j.1365-2141.2009.07961.x.
  19. Waalen J, von Lohneysen K, Lee P, et al. Erythropoietin, GDF15, IL6, hepcidin and testosterone levels in a large cohort of elderly individuals with anaemia of known and unknown cause. Eur J Haematol. 2011;87(2):107–16. doi: 10.1111/j.1600-0609.2011.01631.x.
  20. Kautz L, Jung G, Valore EV, et al. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet. 2014;46(7):678–84. doi: 10.1038/ng.2996.
  21. Merryweather-Clarke AT, Atzberger A, Soneji S, et al. Global gene expression analysis of human erythroid progenitors. Blood. 2011;118(26):e96–108. doi: 10.1182/blood-2010-07-290825.
  22. Diaz V, Gammella E, Recalcati S, et al. Liver iron modulates hepcidin expression during chronically elevated erythropoiesis in mice. Hepatology. 2013;58(6):2122–32. doi: 10.1002/hep.26550.
  23. Pippard MJ, Warner GT, Callender ST, Weatherall DJ. Iron absorption and loading in b-thalassaemia intermedia. Lancet. 1979;314(8147):819–21. doi: 10.1016/s0140-6736(79)92175-5.
  24. Papanikolaou G, Tzilianos M, Christakis JI, et al. Hepcidin in iron overload disorders. Blood. 2005;105(10):4103–5. doi: 10.1182/blood-2004-12-4844.
  25. Centis F, Tabellini L, Lucarelli G, et al. The importance of erythroid expansion in determining the extent of apoptosis in erythroid precursors in patients with b-thalassemia major. Blood. 2000;96(10):3624–9.
  26. Kattamis A, Papassotiriou I, Palaiologou D, et al. The effects of erythropoetic activity and iron burden on hepcidin expression in patients with thalassemia major. Haematologica. 2006;91(6):809–12.
  27. Origa R, Galanello R, Ganz T, et al. Liver iron concentrations and urinary hepcidin in beta-thalassemia. Haematologica. 2007;92(5):583–8. doi: 10.3324/haematol.10842.
  28. Nai A, Pagani A, Mandelli G, et al. Deletion of TMPRSS6 attenuates the phenotype in a mouse model of beta-thalassemia. Blood. 2012;119(21):5021–9. doi: 10.1182/blood-2012-01-401885.
  29. Guo S, Casu C, Gardenghi S, et al. Reducing TMPRSS6 ameliorates hemochromatosis and beta-thalassemia in mice. J Clin Invest. 2013;123(4):1531–41. doi: 10.1172/JCI66969.
  30. Schmidt PJ, Toudjarska I, Sendamarai AK, et al. An RNAi therapeutic targeting Tmprss6 decreases iron overload in Hfe–/– mice and ameliorates anemia and iron overload in murine beta-thalassemia intermedia. Blood. 2013;121(7):1200–8. doi: 10.1182/blood-2012-09-453977.
  31. Goodnough LT, Nemeth E, Ganz T. Detection, evaluation, and management of iron-restricted erythropoiesis. Blood. 2010;116(23):4754–61. doi: 10.1182/blood-2010-05-286260.

 

Primary Mediastinal (Thymic) Large B-Cell Lymphoma

GS Tumyan, IZ Zavodnova, MYu Kichigina, EG Medvedovskaya

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

For correspondence: Gayane Sergeevna Tumyan, DSci, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel: +7(499)324-98-29; e-mail: gaytum@mail.ru

For citation: Tumyan GS, Zavodnova IZ, Kichigina MYu, Medvedovskaya EG. Primary Mediastinal (Thymic) Large B-Cell Lymphoma. Clinical oncohematology. 2017;10(1):13–24 (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-13-24


ABSTRACT

Primary mediastinal (thymic) large B-cell lymphoma (PMBCL) is one of the primary extranodal tumors and originates from thymic medulla B cells. The disease is more common in young women and declares itself by mainly locally advanced growth within the anterior upper mediastinum with frequent involvement of chest organs. PMBCL has specific morphological, immunological, and genetic characteristics that permit to differentiate it from other similar diseases: diffuse large В-cell lymphoma, nodular sclerosis Hodgkin’s lymphoma, and mediastinal gray zone lymphoma. Immunochemotherapy with subsequent irradiation of the residual mediastinal tumor is the standard treatment of PMBCL. No benefits of one drug therapy over another have been demonstrated to date in controlled studies. Application of new imaging techniques (PET/CT) may result in withdrawal of the radiotherapy in some PMBCL patients without impairment of delayed survival rates.

Keywords: primary mediastinal (thymic) large B-cell lymphoma, primary extranodal lymphomas, diagnosis, pathogenesis, morphological, immunological/genetic characteristics, treatment.

Received: August 22, 2016

Accepted: December 17, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Benjamin SP, McCormack LJ, Effler DB, et al. Primary lymphatic tumors of the mediastinum. Cancer. 1972;30(3):708–12. doi: 10.1002/1097-0142(197209)30:3<708::AID-CNCR2820300318>3.0.CO;2–5.
  2. Lichtenstein AK, Levine A, Taylor CR, et al. Primary mediastinal lymphoma in adults. Am J Med. 1980;68(4):509–14. doi: 10.1016/0002-343(80)90294-6.
  3. National Cancer Institute sponsored study of classifications of non-Hodgkin’s lymphomas: summary and description of a working formulation for clinical usage. The Non-Hodgkin’s Lymphoma Pathologic Classification Project. Cancer. 1982;49(10):2112–35. doi: 10.1002/1097-0142(19820515)49:10<2112::AID-CNCR2820491024>3.0.CO;2–2.
  4. Stansfeld AG, Diebold J, Noel H, et al. Updated Kiel classification for lymphomas. Lancet. 1988;1(8580):292–3. doi: 10.1016/S0140-6736(88)90367-4.
  5. Harris NL, Jaffe ES, Stein H, et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood. 1994;84(5):1361–92. doi: 10.1016/S0968-6053(00)80051-4.
  6. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008. doi: 10.1002/9781118853771.ch51.
  7. Cazals-Hatem D, Lepage E, Brice P, et al. Primary mediastinal large B-cell lymphoma. A clinicopathologic study of 141 cases compared with 916 nonmediastinal large B-cell lymphomas, a GELA (“Groupe d’Etude des Lymphomes de l’Adulte”) study. Am J Surg Pathol. 1996;20(7):877–88. doi: 10.1097/00000478-199607000-00012.
  8. Harris NL. Shades of gray between large B-cell lymphomas and Hodgkin lymphomas: differential diagnosis and biological implications. Mod Pathol. 2013;26(Suppl 1):S57–70. doi: 10.1038/modpathol.2012.182.
  9. Kanavaros P, Gaulard P, Charlotte F, et al. Discordant expression of immunoglobulin and its associated molecule mb-1/CD79a is frequently found in mediastinal large B cell lymphomas. Am J Pathol. 1995;146(3):735–41.
  10. Pileri SA, Zinzani PL, Gaidano G, et al. Pathobiology of primary mediastinal B-cell lymphoma. Leuk Lymphoma. 2003;44(Suppl 3):S21–6. doi: 10.1080/10428190310001623810.
  11. Loddenkemper C, Anagnostopoulos I, Hummel M, et al. Differential Emu enhancer activity and expression of BOB.1/OBF.1, Oct2, PU.1, and immunoglobulin in reactive B-cell populations, B-cell non-Hodgkin lymphomas, and Hodgkin lymphomas. J Pathol. 2004;202(1):60–9. doi: 10.1002/path.1485.
  12. De Leval L, Ferry JA, Falini B, et al. Expression of bcl-6 and CD10 in primary Mediastinal large B-cell lymphoma: evidence for derivation from germinal center B cells? Am J Surg Pathol. 2001;25(10):1277–82. doi: 10.1097/00000478-200110000-00008.
  13. 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. doi: 10.1084/jem.20031074.
  14. CopieBergman C, Plonquet A, Alonso MA, et al. MAL expression in lymphoid cells: further evidence for MAL as a distinct molecular marker of primary mediastinal large B-cell lymphomas. Mod Pathol. 2002;15:1172–80. doi: 10.1097/01.MP.0000032534.81894.B3.
  15. Joos S, Otano-Joos MI, Ziegler S, et al. Primary mediastinal (thymic) B-cell lymphoma is characterized by gains of chromosomal material including 9p and amplification of the REL gene. Blood. 1996;87(4):1571–8.
  16. Feuerhake F, Kutok JL, Monti S, et al. NFkappaB activity, function, and target-gene signatures in primary mediastinal large B-cell lymphoma and diffuse large B-cell lymphoma subtypes. Blood. 2005;106(4):1392–9. doi: 10.1182/blood-2004-12-4901.
  17. Zhang B, Wang Z, Li T, et al. NF-kappaB2 mutation targets TRAF1 to induce lymphomagenesis. Blood. 2007;110(2):743–51. doi: 10.1182/blood-2006-11-058446.
  18. Meier C, Hoeller S, Bourgau C, et al. Recurrent numerical aberrations of JAK2 and deregulation of the JAK2-STAT cascade in lymphomas. Mod Pathol. 2009;22(3):476–87. doi: 10.1038/modpathol.2008.207.
  19. Rossi D, Cerri M, Capello D, et al. Aberrant somatic hypermutation in primary mediastinal large B-cell lymphoma. Leukemia. 2005;19(12):2363–6. doi: 10.1038/sj.leu.2403982.
  20. Steidl C, Gascoyne RD. The molecular pathogenesis of primary mediastial large B-cell lymphoma. Blood. 2011;118(10):2659–69. doi: 10.1182/blood-2011-05-326538.
  21. Martelli M, Di Rocco A, Russo E, et al. Primary mediastinal lymphoma: diagnosis and treatment options. Expert Rev Hematol. 2014;8(2):173–86. doi: 10.1586/17474086.2015.994604.
  22. Eberle FC, Salaverria I, Steidl C, et al. Gray zone lymphoma: chromosomal aberrations with immunophenotypic and clinical correlations. Mod Pathol. 2011;24(12):1586–97. doi: 10.1038/modpathol.2011.116.
  23. Eberle FC, Rodriguez-Canales J, Wei L, et al. Methylation profiling of mediastinal gray zone lymphoma reveals a distinctive signature with elements shared by classical Hodgkin’s lymphoma and primary mediastinal large B-cell lymphoma. Haematologica. 2011;96(4):558–66. doi: 10.3324/haematol.2010.033167.
  24. Moller P, Lammler B, Herrmann B, et al. The primary mediastinal clear cell lymphoma of B-cell type has variable defects in MHC antigen expression. Immunology. 1986;59(3):411–7. doi: 10.1007/bf00705408.
  25. Hamlin PA, Portlock CS, Straus DJ, et al. Primary mediastinal large B-cell lymphoma: optimal therapy and prognostic factor analysis in 141 consecutive patients treated at Memorial Sloan Kettering from 1980 to 1999. Br J Haematol. 2005;130(5):691–9. doi: 10.1111/j.1365-2141.2005.05661.x.
  26. Jacobson JO, Aisenberg AC, Lamarre L, et al. Mediastinal large cell lymphoma. An uncommon subset of adult lymphoma curable with combined modality therapy. Cancer. 1988;62(9):1893–8. doi: 10.1002/1097-0142(19881101)62:9<1893::AID-CNCR2820620904>3.0.CO;2-X.
  27. Zinzani PL, Martelli M, Magagnoli M, et al. Treatment and clinical management of primary mediastinal large B-cell lymphoma with sclerosis: MACOP-B regimen and mediastinal radiotherapy monitored by (67)Gallium scan in 50 patients. Blood. 1999;94(10):3289–93.
  28. Bishop PC, Wilson WH, Pearson D, et al. CNS involvement in primary mediastinal large B-cell lymphoma. J Clin Oncol. 1999;17(8):2479–85.
  29. Savage K, Al-Rajhi N, Voss N, et al. Favorable outcome of primary mediastinal large B-cell lymphoma in a single institution: the British Columbia experience. Ann Oncol. 2006;17:123–30. doi: 10.1016/s0360-3016(00)80463-0.
  30. Zinzani PL, Martelli M, Bertini M, et al. Induction chemotherapy strategies for primary mediastinal large B-cell lymphoma with sclerosis: a retrospective multinational study on 426 previously untreated patients. Haematologica. 2002;87(12):1258–6. doi: 10.3816/clm.2009.n.074.
  31. Fisher RI, Gaynor ER, Dahlberg S, et al. Comparison of a standard regimen (CHOP) with three intensive chemotherapy regimens for advanced non-Hodgkin’s lymphoma. N Engl J Med. 1993;328(14):1002–6. doi: 10.1056/NEJM199304083281404.
  32. Levitt LJ, Aisenberg AC, Harris NL, et al. Primary non-Hodgkin’s lymphoma of the mediastinum. Cancer. 1982;50(11):2486–92. doi: 10.1002/1097-0142(19821201)50:11<2486::AID-CNCR2820501138>3.0.CO;2-G.
  33. Todeschini G, Ambrosetti A, Meneghini V, et al. Mediastinal large-B-cell lymphoma with sclerosis: a clinical study of 21 patients. J Clin Oncol. 1990;8(5):804–8.
  34. Bertini M, Orsucci L, Vitolo U, et al. Stage II large B-cell lymphoma with sclerosis treated with MACOP-B. Ann Oncol. 1991;2(10):733–7.
  35. Falini B, Venturi S, Martelli M, et al. Mediastinal large B-cell lymphoma: clinical and immunohistological findings in 18 patients treated with different third-generation regimens. Br J Haematol. 1995;89(4):780–9. doi: 10.1111/j.1365-2141.1995.tb08415.x.
  36. van Besien K, Kelta M, Bahaguna P. Primary mediastinal B-cell lymphoma: a review of pathology and management. J Clin Oncol. 2001;19(6):1855–64.
  37. Zinzani PL, Martelli M, Bendandi M, et al. Primary mediastinal large B-cell lymphoma with sclerosis: a clinical study of 89 patients treated with MACOP-B chemotherapy and radiation therapy. Haematologica. 2001;86(2):187–91.
  38. Zinzani PL, Stefoni V, Finolezzi E, et al. Rituximab combined with MACOP-B or VACOP-B and radiation therapy in primary mediastinal large B-cell lymphoma: a retrospective study. Clin Lymph Myel. 2009;9(5):381–5. doi: 10.3816/CLM.2009.n.074.
  39. Dunleavy K, Pittaluga S, Maeda LS, et al. Dose-adjusted EPOCH-rituximab therapy in primary mediastinal B-cell lymphoma. N Engl J Med. 2013;368(15):1408–16. doi: 10.1056/NEJMoa1214561.
  40. Moskowitz CH, Schoder H, Teruya-Feldstein J, et al. Risk-adapted dose-dense immunochemotherapy determined by interim FDG-PET in Advanced-stage diffuse large B-Cell lymphoma. J Clin Oncol. 2010;28(11):1896–903. doi: 10.1200/JCO.2009.26.5942.
  41. Savage KJ, Yenson PR, Shenkier T, et al. The outcome of primary mediastinal large B-cell lymphoma (PMBCL) in the R-CHOP treatment era. Blood. 2012;120(Suppl 1–2): Abstract 303.
  42. Martelli M, Ceriani L, Zucca E, et al. [18F]fluorodeoxyglucose positron emission tomography predicts survival after chemoimmunotherapy for primary mediastinal large B-cell lymphoma: results of the International Extranodal Lymphoma Study Group IELSG-26 Study. J Clin Oncol. 2014;32(17):1769–75. doi: 10.1200/JCO.2013.51.7524.
  43. Pinnix CC, Dabaja B, Ahmed MAet al. Single-institution experience in the treatment of primary mediastinal B cell lymphoma treated with immunochemotherapy in the setting of response assessment by 18fluorodeoxyglucose positron emission tomography. Int J Radiat Oncol Biol Phys. 2015;92(1):113–21. doi: 10.1016/j.ijrobp.2015.02.006.
  44. Sehn LH, Antin JH, Shulman LN, et al. Primary diffuse large B-cell lymphoma of the mediastinum: outcome following high-dose chemotherapy and autologous hematopoietic cell transplantation. Blood. 1998;91(2):717–23.
  45. Kuruvilla J, Pintilie M, Tsang R, et al. Salvage chemotherapy and autologous stem cell transplantation are inferior for relapsed or refractory primary mediastinal large B-cell lymphoma compared with diffuse large B-cell lymphoma. Leuk Lymphoma. 2008;49(7):1329–36. doi: 10.1080/10428190802108870.
  46. Hao Y, Chapuy B, Monti S, Sun HH. Selective JAK2 inhibition specifically decreases Hodgkin lymphoma and mediastinal large B-cell lymphoma growth in vitro and in vivo. Clin Cancer Res. 2014;20(10):2674–83. doi: 10.1158/1078-0432.CCR-13-3007.
  47. Dunleavy K, Wilson W. Primary mediastinal B-cell lymphoma and mediastinal gray zone lymphoma: do they require a unique therapeutic approach? Blood. 2015;125(1):33–9. doi: 10.1182/blood-2014-05-575092.
  48. Berger R, Rotem-Yehudar R, Slama G, et al. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res. 2008;14(10):3044–51. doi: 10.1158/1078-0432.CCR-07-4079.

Certain Aspects of Autologous Hematopoietic Stem Cell Transplantation in Patients with Multiple Myeloma

SV Gritsaev, AA Kuzyaeva, SS Bessmel’tsev

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: Sergei Vasil’evich Gritsaev, DSci, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel: +7(812)717-58-57; e-mail: gritsaevsv@mail.ru

For citation: Gritsaev SV, Kuzyaeva AA, Bessmel’tsev SS. Certain Aspects of Autologous Hematopoietic Stem Cell Transplantation in Patients with Multiple Myeloma. Clinical oncohematology. 2017;10(1):7–12 (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-7-12


ABSTRACT

The review dwells on certain problems of mobilization and conditioning regimens, as well as autologous hematopoietic stem cell transplantation (auto-HSCT) in patients with multiple myeloma. The aim of the review is to determine new approaches to improve the effectiveness of the auto-HSCT.

Keywords: multiple myeloma, autologous hematopoietic stem cell transplantation, mobilization regimen, conditioning regimen.

Received: July 13, 2016

Accepted: November 12, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома: руководство для врачей. М.: МК, 2016. 504 с.
    [Bessmel’tsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma: rukovodstvo dlya vrachei. (Multiple myeloma: manual for physicians.) Moscow: MK Publ.; 2016. 504 p. (In Russ)]
  2. Менделеева Л.П., Вотякова О.М., Покровская О.С. и др. Национальные рекомендации по диагностике и лечению множественной миеломы. Гематология и трансфузиология. 2014;1(Приложение № 3):1–24.
    [Mendeleeva LP, Votyakova OM, Pokrovskaya OS, et al. National guidelines for diagnosis and treatment of multiple myeloma. Gematologiya i transfuziologiya. 2014;1(Suppl 3):1–24. (In Russ)]
  3. Reece DE. Management of multiple myeloma: The changing landscape. Blood Rev. 2007;21(6):301–14. doi: 10.1016/j.blre.2007.07.001.
  4. Cavo M, Tosi P, Zamagni E, et al. Prospective, randomized study of single compared with double autologous stem-cell transplantation for multiple myeloma: Bologna 96 clinical study. J Clin Oncol. 2007;25(17):2434–41. doi: 10.1200/jco.2006.10.2509.
  5. Attal M, Harousseau JL, Facon T, et al. Single versus double autologous stem-cell transplantation for multiple myeloma. N Engl J Med. 2003;349(26):2495–502. doi: 10.1056/nejmoa032290.
  6. Allan DS, Keeney M, Howson-Jan K, et al. Number of viable CD34(+) cells reinfused predicts engraftment in autologous hematopoietic stem cell transplantation. Bone Marrow Transplant. 2002;29(12):967–72. doi: 10.1038/sj.bmt.1703575.
  7. Michaelis LC, Saad A, Zhong X, et al. Salvage second hematopoietic cell transplantation in myeloma. Biol Blood Marrow Transplant. 2013;19(5):760–6. doi: 10.1016/j.bbmt.2013.01.004.
  8. Cook G, Williams C, Brown JM, et al. High dose chemotherapy plus autologous stem-cell transplantation as consolidation therapy in patients with relapsed multiple myeloma after previous autologous stem-cell transplantation (NCRI Myeloma X Relapse [Intensive trial]): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(14):874–85. doi: 10.1016/s1470-2045(14)70245-1.
  9. Musto P, Simeon V, Grossi A, et al. Predicting poor peripheral blood stem cell collection in patients with multiple myeloma receiving pre-transplant induction therapy with novel agents and mobilized with cyclophosphamide plus granulocyte-colony stimulating factor: results from a Gruppo Italiano Malattie Ematologiche dell’Adulto Multiple Myeloma Working Party study. Stem Cell Res Ther. 2015;6:64. doi: 10.1186/s13287-015-0033-1.
  10. Olivieri A, Marchetti M, Lemoli R, et al. Proposed definition of “poor mobilizer” in lymphoma and multiple myeloma: an analytic hierarchy process by ad hoc working group Gruppo ItalianoTrapianto di Midollo Osseo. Bone Marrow Transplant. 2012;47(3):342–51. doi: 10.1038/bmt.2011.82.
  11. To LB, Levesque JP, Herbert KE. How I treat patients who mobilize hematopoietic stem cells poorly. Blood. 2011;118(17):4530–40. doi: 10.1182/blood-2011-06-318220.
  12. Gertz MA. Current status of stem cell mobilization. Br J Haematol. 2010;150(6):647–62. doi: 10.1111/j.1365-2141.2010.08313.x.
  13. Popat U, Saliba R, Thandi R, et al. Impairment of filgrastim induced stem cell mobilization after prior lenalidomide in patients with multiple myeloma. Biol Blood Marrow Transplant. 2009;15(6):718–23. doi: 10.1016/j.bbmt.2009.02.011.
  14. Mazumder A, Kaufman J., Niesvizky R, et al. Effect of lenalidomide therapy on mobilization of peripheral blood stem cells in previously untreated multiple myeloma patients (letter). Leukemia. 2008;22(60):1280–1. doi: 10.1038/sj.leu.2405035.
  15. Giralt S, Costa L, Schriber J, et al. Optimizing autologous stem cell mobilization strategies to improve patient outcomes: consensus guidelines and recommendations. Biol Blood Marrow Transplant. 2014;20(3):295–308. doi: 10.1016/j.bbmt.2013.10.013.
  16. Duong HK, Savani BN, Copelan E, et al. Peripheral blood progenitor cell mobilization for autologous and allogeneic hematopoietic cell transplantation: guidelines from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2014;20(9):1262–73. doi: 10.1016/j.bbmt.2014.05.003.
  17. Sung AD, Grima DT, Bernard LM, et al. Outcomes and costs of autologous stem cell mobilization with chemotherapy plus G-CSF vs G-CSF alone. Bone Marrow Transplant. 2013;48(11):1444–9. doi: 10.1038/bmt.2013.80.
  18. Gertz MA, Kumar SK, Lacy MQ, et al. Comparison of high-dose CY and growth factor with growth factor alone for mobilization of stem cells for transplantation in patients with multiple myeloma. Bone Marrow Transplant. 2009;43(8):619–25. doi: 10.1038/bmt.2008.369.
  19. Arora M, Burns LJ, Barker JN, et al. Randomized comparison of granulocyte colony-stimulating factor versus granulocyte-macrophage colony-stimulating factor plus intensive chemotherapy for peripheral blood stem cell mobilization and autologous transplantation in multiple myeloma. Biol Blood Marrow Transplant. 2004;10(6):395–404. doi: 10.1016/s1083-8791(04)00068-0.
  20. Nakasone H, Kanda Y, Ueda T, et al. Retrospective comparison of mobilization methods for autologous stem cell transplantation in multiple myeloma. Am J Hematol. 2009;84(12):809–14. doi: 10.1002/ajh.21552.
  21. Mark T, Stern J, Furst JR, et al. Stem cell mobilization with cyclophosphamide overcomes the suppressive effect of lenalidomide therapy on stem cell collection in multiple myeloma. Biol Blood Marrow Transplant. 2008;14(7):795–8. doi: 10.1016/j.bbmt.2008.04.008.
  22. Costa LJ, Miller AN, Alexander ET, et al. Growth factor and patient-adapted use of plerixafor is superior to CY and growth factor for autologous hematopoietic stem cells mobilization. Bone Marrow Transplant. 2011;46(4):523–8. doi: 10.1038/bmt.2010.170.
  23. DiPersio J., Stadtmauer EA, Nademanee A, et al. Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma. Blood. 2009;113(23):5720–6. doi: 10.1182/blood-2008-08-174946.
  24. Покровская О.С. Механизм действия и клиническая эффективность антагониста хемокинового рецептора CXCR4 плериксафора при мобилизации гемопоэтических стволовых клеток. Клиническая онкогематология. 2012;5(4):371–9.
    [Pokrovskaya OS. Mechanism of action and clinical activity of CXCR4 antagonist Plerixafor in stem cell mobilization. Klinicheskaya onkogematologiya. 2012;5(4):371–9. (In Russ)]
  25. Кучер М.А., Моталкина М.С., Климова О.У. и др. Плериксафор у пациентов со сниженной мобилизационной способностью аутологичных гемопоэтических стволовых клеток. Клиническая онкогематология. 2016;9(2):155–61. doi: 10.21320/2500-2139-2016-9-2-155-61.
    [Kucher MA, Motalkina MS, Klimova OU, et al. Plerixafor in Patients with Decreased Mobilizing Ability of Autologous Hematopoietic Stem Cells. Clinical oncohematology. 2016;9(2):155–61. doi: 10.21320/2500-2139-2016-9-2-155-61. (In Russ)]
  26. Levesque JP, Takamatsu Y, Nilsson SK, et al. Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases in the bone marrow following hematopoietic progenitor cell mobilization by granulocyte colony-stimulating factor. Blood. 2001;98(5):1289–97. doi: 10.1182/blood.V98.5.1289.
  27. Levesque JP, Hendy J, Takamatsu Y, et al. Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by GCSF or cyclophosphamide. J Clin Invest. 2003;111(2):187–96. doi: 10.1172/jci15994.
  28. Petit I, Szyper-Kravitz M, Nagler A, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and upregulating CXCR4. Nat Immunol. 2002;3(7):687–94. doi: 10.1038/ni813.
  29. Cesana C, Carlo-Stella C, Regazzi E, et al. CD34+ cells mobilized by cyclophosphamide and granulocyte colonystimulating factor (G-CSF) are functionally different from CD34+ cells mobilized by G-CSF. Bone Marrow Transplant. 1998;21(6):561–8. doi: 10.1038/sj.bmt.1701133.
  30. Bruns I, Steidl U, Fischer JC, et al. Pegylated granulocyte colony-stimulating factor mobilizes CD34+ cells with different stem and progenitor subsets and distinct functional properties in comparison with unconjugated granulocyte colony-stimulating factor. Haematologica. 2008;93(3):347–55. doi: 10.3324/haematol.12081.
  31. Kim MG, Han N, Lee EK, Kim T. Pegfilgrastim vs filgrastim in PBSC mobilization for autologous hematopoietic SCT: a systematic review and meta-analysis. Bone Marrow Transplant. 2015;50(4):523–30. doi: 10.1038/bmt.2014.297.
  32. Tuchman SA, Bacon WA, Huang LW, et al. Cyclophosphamide-based hematopoietic stem cell mobilization before autologous stem cell transplantation in newly diagnosed multiple myeloma. J Clin Apher. 2015;30(3):176–82. doi: 10.1002/jca.21360.
  33. Dingli D, Nowakowski GS, Dispenzieri A, et al. Cyclophosphamide mobilization does not improve outcome in patients receiving stem cell transplantation for multiple myeloma. Clin Lymphoma Myeloma. 2006;6(5):384–8. doi: 10.3816/clm.2006.n.014.
  34. Hamadani M, Kochuparambil ST, Osman S, et al. Intermediate-dose versus low-dose cyclophosphamide and granulocyte colony-stimulating factor for peripheral blood stem cell mobilization in patients with multiple myeloma treated with novel induction therapies. Biol Blood Marrow Transplant. 2012;18(7):1128–35. doi: 10.1016/j.bbmt.2012.01.005.
  35. Hiwase DK, Bollard G, Hiwase S. Intermediate-dose CY and G-CSF more efficiently mobilize adequate numbers of PBSC for tandem autologous PBSC transplantation compared with low-dose CY in patients with multiple myeloma. Cytotherapy. 2007;9(6):539–47. doi: 10.1080/14653240701452800.
  36. Jantunen E, Putkonen M, Nousiainen T, Low-dose or intermediate-dose cyclophosphamide plus granulocyte colonystimulating factor for progenitor cell mobilisation in patients with multiple myeloma. Bone Marrow Transplant. 2003; 31(5):347–51. doi: 10.1038/sj.bmt.1703840.
  37. Nazha A, Cook R, Vogl DT, et al. Stem cell collection in patients with multiple myeloma: impact of induction therapy and mobilization regimen. Bone Marrow Transplant. 2011;46(1):59–63. doi: 10.1038/bmt.2010.63.
  38. Brioli A, Perrone G, Patriarca F, et al. Successful mobilization of PBSCs predicts favorable outcomes in multiple myeloma patients treated with novel agents and autologous transplantation. Bone Marrow Transplant. 2015;50(5):673–8. doi: 10.1038/bmt.2014.322.
  39. Samaras P, Pfrommer S, Seifert B, et al. Efficacy of vinorelbine plus granulocyte colonye-stimulation factor for CD34+ hematopoietic progenitor cell mobilization in patients with multiple myeloma. Biol Blood Marrow Transplant. 2015;21(1):74–80. doi: 10.1016/j.bbmt.2014.09.020.
  40. Heizmann M, O’Meara AC, Moosmann PR, et al. Efficient mobilization of PBSC with vinorelbine/G-CSF in patients with malignant lymphoma. Bone Marrow Transplant. 2009;44(2):75–9. doi: 10.1038/bmt.2008.434.
  41. Annunziata M, Celentano M, Pocali B, et al. Vinorelbine plus intermediate dose cyclophosphamide is an effective and safe regimen for the mobilization of peripheral blood stem cells in patients with multiple myeloma. Ann Hematol. 2006;85(6):394–9. doi: 10.1007/s00277-005-0058-0.
  42. Bargetzi MJ, Passweg J, Baertschi E, et al. Mobilization of peripheral blood progenitor cells with vinorelbine and granulocyte colony-stimulating factor in multiple myeloma patients is reliable and cost effective. Bone Marrow Transplant. 2003;31(2):99–103. doi: 10.1038/sj.bmt.1703787.
  43. Schmid A, Friess D, Taleghani BM, et al. Role of plerixafor in autologous stem cell mobilization with vinorelbine chemotherapy and granulocyte-colony stimulating factor in patients with myeloma: a phase II study (PAV-trial). Leuk Lymphoma. 2015;56(3):608–14. doi: 10.3109/10428194.2014.927454.
  44. Moreau P, Facon T, Attal M, et al. Comparison of 200 mg/m2 melphalan and 8 Gy total body irradiation plus 140 mg/m2 melphalan as conditioning regimens for peripheral blood stem cell transplantation in patients with newly diagnosed multiple myeloma: final analysis of the Intergroupe Francophone du Myelome 9502 randomized trial. Blood. 2002;99(3):731–5. doi: 10.1182/blood.v99.3.731.
  45. Palumbo A, Bringhen S, Bruno B, et al. Melphalan 200 mg/m(2) versus melphalan 100 mg/m(2) in newly diagnosed myeloma patients: a prospective, multicenter phase 3 study. Blood. 2010;115(10):1873–9. doi: 10.1182/blood-2010-08-301085.
  46. Giralt S. 200mg/m2 melphalan – the gold standard for multiple myeloma. Nat Rev. 2010;7(9):490–1. doi: 10.1038/nrclinonc.2010.104.
  47. Philips GL, Meisenberg BR, Reece DE, et al. Activity of single-agent melphalan 220 to 300 mg/m2 with amifostine cytoprotection and autologous hematopoietic stem cell support in non-Hodgkin and Hodgkin lymphoma. Bone Marrow Transplant. 2004;33(8):781–7. doi: 10.1038/sj.bmt.1704424.
  48. Moreau P, Milpied N, Mahe B. Melphalan 220 mg/m2 followed by peripheral blood stem cell transplantation in 27 patients with advanced multiple myeloma. Bone Marrow Transplant. 1999;23(10):1003–6. doi: 10.1038/sj.bmt.1701763.
  49. Reece D., Song K., Leblanc R., et al. Efficacy and safety of busulfan-based conditioning regimens for multiple myeloma. Oncologist. 2013;18:611–8. doi: 10.1634/theoncologist.2012-0384.
  50. Roussel M, Moreau P, Huynh A, et al. Bortezomib ad high-dose melphalan as conditioning regimen before autologous stem cell transplantation in patients with de novo multiple myeloma: a phase 2 study of the Intergroupe Francophone du Myelome (IFM). Blood. 2010;115(1):32–7. doi: 10.1182/blood-2009-06-229658.
  51. Nishihori T, Alekshun TJ, Shain K, et al. Bortezomib salvage followed by a phase I/II study of bortezomib plus high-dose melphalan and tandem autologous transplantation for patients with primary resistant myeloma. Br J Haematol. 2012;157(5):553–63. doi: 10.1111/j.1365-2141.2012.09099.x.
  52. Huang W, Li J, Li H, et al. High-dose melphalan with bortezomib as conditioning regimen for autologous stem cell transplant in patients with newly diagnosed multiple myeloma who exhibited at least very good partial response to bortezomib-based induction therapy. Leuk Lymphoma. 2012;53(12):2507–10. doi: 10.3109/10428194.2012.685735.
  53. Mark TM, Reid W, Niesvizky R, et al. A phase 1 study of bendamustine and melphalan conditioning for autologous stem cell transplant in multiple myeloma. Biol Blood Marrow Transplant. 2013;19(5):831–7. doi: 10.3109/10428194.2012.685735.
  54. Martino M, Tripepi G, Messina G, et al. A phase II, single-arm, prospective study of bendamustine plus melphalan conditioning for second autologous stem cell transplantation in de novo multiple myeloma patients through a tandem transplant strategy. Bone Marrow Transplant. 2016;51(9):1197–203. doi: 10.1038/bmt.2016.94.
  55. Visani G, Malerba L, Stefani PM, et al. BeEAM (bendamustine, etoposide, cytarabine, melphalan) before autologous stem cell transplantation is safe and effective for resistant/relapsed lymphoma patients. Blood. 2011;118(12):3419–25. doi: 10.1182/blood-2011-04-351924.
  56. Veeraputhiran M, Jain T, Deol A, et al. BEAM conditioning regimen has higher toxicity compared with high-dose melphalan for salvage autologous hematopoietic stem cell transplantation in multiple myeloma. Clin Lymph Myeloma Leuk. 2015;15(9):531–5. doi: 10.1016/j.clml.2015.05.008.
  57. Abu Zaid B, Abdul-Hai A, Grotto I, et al. Autologous transplant in multiple myeloma with an augmented conditioning protocol. Leuk Lymphoma. 2013;54(11):2480–4. doi: 10.3109/10428194.2013.782608.
  58. Musso M, Messina G, Marcacci G, et al. High-dose melphalan plus thiotepa as conditioning regimen before second autologous stem cell transplantation for “de novo” multiple myeloma patients: a phase II study. Biol Blood Marrow Transplant. 2015;21(11):1932–8. doi: 10.1016/j.bbmt.2015.06.011.

 

Hypomethylating Agents in Oncohematology

AD Shirin, OYu Baranova

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

For correspondence: Anton Dmitrievich Shirin, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-28-24; e-mail: shirin-anton@mail.ru

For citation: Shirin AD, Baranova OYu. Hypomethylating Agents in Oncohematology. Clinical oncohematology. 2016;9(4):369–82 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-369-382


ABSTRACT

The review describes epigenetic processes, including methylation of nuclear and mitochondrial DNA, as well as RNA. It dwells on mechanisms of demethylation and corresponding medicinal products. It presents detailed information on results of numerous large randomized studies intended to evaluate hypomethylating agents (azanucleosides). Special attention is paid to outcomes of azanucleoside therapy in patients with acute myeloid leukemias. The article describes several prognostic systems and treatment algorithms for myelodysplastic syndromes. Two azanucleosides have been approved in Russia to date: azacitidine (for SQ administration) and decitabine (for IV administration). International authors analyze the experience in oral and subcutaneous administration of decitabine. However, the problem of off-label use of hypomethylating agents is still open. The review gives a brief description of ongoing clinical trials with azanucleosides.


Keywords: epigenetics, acute myeloid leukemias, myelodysplastic syndromes, azacitidine, decitabine, hypomethylating agents, azanucleosides.

Received: May 10, 2016

Accepted: May 20, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Уоддингтон К.Х. Основные биологические концепции. В кн.: На пути к теоретической биологии. Часть I. Пролегомены. М.: Мир, 1970. С. 11–38.
    [Waddington CH. Basic Ideas of Biology. In: Waddington CH, ed. Towards a Theoretical Biology. Vol. 1. Edinburgh: Edinburgh University Press. 1968–72. (Russ. ed.: Waddington CH. Osnovnye biologicheskie kontseptsii. In: Waddington CH, ed. Na puti k teoreticheskoi biologii. Chast’ I. Prolegomeny. Moscow: Mir Publ.; 1970. pp. 11–38.)]
  2. Huntly BJP, Johnson PWM. Targeting Epigenetic Readers in Hematologic Malignancies: A Good BET? The Hematologist. 2012;9(2):5–7.
  3. Daser A, Rabbitts TH. Extending the repertoire of the mixed-lineage leukemia gene MLL in leukemogenesis. Genes & Dev. 2004;18:965–74. doi: 10.1101/gad.1195504.
  4. Ansorge WJ. Next-generation DNA sequencing techniques. New Biotechnol. 2009;25(4):195–203. doi: 10.1016/j.nbt.2008.12.009.
  5. Foley SB, Rios JJ, Mgbemena V. Use of Whole Genome Sequencing for Diagnosis and Discovery in the Cancer Genetics Clinic. EBioMedicine. 2014;2(1):74–81. doi: 10.1016/j.ebiom.2014.12.003.
  6. Wojdacz TK, Moller TH, Thestrup BB, et al. Limitations and advantages of MS-HRM and bisulfite sequencing for single locus methylation studies. Exp Rev Mol Diagn. 2010;10(5):575–80. doi: 10.1586/erm.10.46.
  7. Reinders J, Paszkowski J. Bisulfite methylation profiling of large genomes. Epigenomics. 2010;2(2):209–20. doi: 10.2217/epi.10.6.
  8. Thompson CB. Targeting Metabolic Inputs into Epigenetic Regulations of Acute Leukemia. Blood. 2013;122(21):SCI-26.
  9. Зиновкина Л.А., Зиновкин Р.А. Метилирование ДНК, митохондрии и программируемое старение. Биохимия. 2015;80(12):1830–7.
    [Zinovkina LA, Zinovkin RA. DNA methylation, mitochondria, and programmed aging. Biokhimiya. 2015;80(12):1830–7. (In Russ)]
  10. Vanyushin BF, Kiryanov GI, Kudryashova IB, Belozersky AN. DNA & methylase in loach embryos (Misgurnus fossilis). FEBS Lett. 1971;15(4):313–6. doi: 10.1016/0014-5793(71)80646-4.
  11. Vanyushin BF, Kirnos MD. The nucleotide composition and pyrimidine clusters in DNA from beef heart mitochondria. FEBS Lett. 1974;39(2):195–9. doi: 10.1016/0014-5793(74)80049-99.
  12. Vanyushin BF, Kirnos MD. The structure of animal mitochondrial DNA (base composition, pyrimidine clusters, character of methylation). Mol Cell Biochem. 1977;14(1–3):31–6. doi: 10.1007/bf01734162.
  13. Byun HM, Panni T, Motta V, et al. Effects of airborne pollutants on mitochondrial DNA methylation. Part Fibre Toxicol. 2013;10(1):18. doi: 10.1186/1743-8977-10-18.
  14. Sun C, Reimers LL, Burk RD. Methylation of HPV16 genome CpG sites is associated with cervix precancer and cancer. Gynecol Oncol. 2011;121(1):59–63. doi: 10.1016/j.ygyno.2011.01.013.
  15. Vanyushin BF, Nemirovsky LE, Klimenko VV, et al. The 5-methylcytosine in DNA of rats. Gerontologia. 1973;19(3):138–52. doi: 10.1159/000211967.
  16. Биология и медицина. Метилирование РНК. [Электронный документ] Доступно по: http://medbiol.ru/medbiol/epigenetica/001a1613.htm. Ссылка активна на 14.05.2013.
    [Biologiya i meditsina. Metilirovanie RNK. (Biology and Medicine. RNA Methylation) [Internet]. Available from: http://medbiol.ru/medbiol/epigenetica/001a1613.htm. (accessed 14.05.2013) (In Russ)]
  17. Yu B, Yang Z, Li J, et al. Methylation as a crucial step in plant microRNA biogenesis. Science. 2005;307(5711):932–5. doi: 10.1126/science.1107130.
  18. Goll MG, Kirpekar E, Maggert KA, et al. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science. 2006;311(5759):395–8. doi: 10.1126/science.1120976.
  19. Dominissini D, Nachtergaele S, Moshitch-Moshkovitz S, et al. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature. 2016;530(7591):441–6. doi: 10.1038/nature16998.
  20. Christman J. 5-Azacytidine and 5-aza-2¢-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene. 2002;21(35):5483–95. doi: 10.1038/sj.onc.1205699.
  21. Kumar A, List A. F, Hozo I, et al. Decitabine versus 5-azacitidine for the treatment of myelodysplastic syndrome: adjusted indirect meta-analysis. Haematologica. 2010;95(2):340–2. doi: 10.3324/haematol.2009.017764.
  22. Phase II Decitabine (DAC) Versus Azacitidine (AZA) in Myelodysplastic Syndrome (MDS). [Internet] Available from: http://www.druglib.com/trial/80/NCT02269280.html. (accessed 15.05.2016).
  23. Fenaux P, Gattermann N, Seymour JF, et al. Prolonged survival with improved tolerability in higher-risk myelodysplastic syndromes: azacitidine compared with low dose ara-C. Br J Haematol. 2010;149(2):244–9. doi: 10.1111/j.1365-2141.2010.08082.x.
  24. Al-Ali HK, Jaekel N, Niederwieser D. The role of hypomethylating agents in the treatment of elderly patients with AML. J Geriatr Oncol. 2014;5(1):89–105. doi: 10.1016/j.jgo.2013.08.004.
  25. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109(6):1114–24. doi: 10.1002/cncr.22496.
  26. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30(21):2670–7. doi: 10.1200/jco.2011.38.9429.
  27. European Medicines Agency: assessment report on Dacogen 19 July 2012. [Internet] Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/002221/WC500133571.pdf2012. (accessed 17.05.2016).
  28. Minutes for the February 9 2012 meeting of the FDA Oncologic Drugs Advisory Committee. [Internet] Available from: http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/OncologicDrugsAdvisoryCommittee/UCM293710.pdf2012. (accessed 19.05.2016).
  29. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454–65. doi: 10.1182/blood-2012-03-420489.
  30. Schanz J, Tuchler H, Sole F, et al. New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge. J Clin Oncol. 2012;30(8):820–9. doi: 10.1200/jco.2011.35.6394.
  31. Kantarjian H, O’Brien S, Ravandi F, et al. Proposal for a new risk model in myelodysplastic syndrome that accounts for events not considered in the original International Prognostic Scoring System. Cancer. 2008;113(6):1351–61. doi: 10.1002/cncr.23697.
  32. Garcia-Manero G. Myelodysplastic syndromes: 2015 Update on diagnosis, risk-stratification and management. Am J Hematol. 2015;90(9):831–41. doi: 10.1002/ajh.24102.
  33. Garcia-Manero G, Fenaux P. Hypomethylating agents and other novel strategies in myelodysplastic syndromes. J Clin Oncol. 2011;29(10):516–23. doi: 10.1200/jco.2010.31.0854.
  34. Lyons RM, Cosgriff TM, Modi SS, et al. Hematologic response to three alternative dosing schedules of azacitidine in patients with myelodysplastic syndromes. J Clin Oncol. 2009;27(11):1850–6. doi: 10.1200/jco.2008.17.1058.
  35. Garcia-Manero G, Gore SD, Cogle C, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol. 2011;29(18):2521–7. doi: 10.1200/jco.2010.34.4226.
  36. Garcia-Manero G, Jabbour E, Borthakur G, et al. Randomized open-label phase II study of decitabine in patients with low- or intermediate-risk myelodysplastic syndromes. J Clin Oncol. 2013;31(20):2548–53. doi: 10.1200/jco.2012.44.6823.
  37. Wei Y, Dimicoli S, Bueso-Ramos C, et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia. 2013;27(9):1832–40. doi: 10.1038/leu.2013.180.
  38. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: A randomised, open-label, phase III study. Lancet Oncol. 2009;10(3):223–32. doi: 10.1016/s1470-2045(09)70003-8.
  39. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci USA. 2010;107(16):7473–8. doi: 10.1073/pnas.1002650107.
  40. Itzykson R, Thepot S, Quesnel B, et al. Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. Blood. 2011;117(2):403–11. doi: 10.1182/blood-2010-06-289280.
  41. Jabbour E, Garcia-Manero G, Batty N, et al. Outcome of patients with myelodysplastic syndrome after failure of decitabine therapy. Cancer. 2010;116(16):3830–4. doi: 10.1002/cncr.25247.
  42. Montalban-Bravo G, Garcia-Manero G. Novel drugs for older patients with acute myeloid leukemia. Leukemia. 2015;29(4):760–9. doi: 10.1038/leu.2014.244.
  43. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with > 30% blasts. Blood. 2015;126(3):291–9. doi: 10.1182/blood-2015-01-621664.
  44. Pleyer L, Burgstaller S, Girschikofsky M, et al. Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-Study Group. Ann Hematol. 2014;93(11):1825–38. doi: 10.1007/s00277-014-2126-9.
  45. Radujkovic A, Dietrich S, Bochtler T, et al. Azacitidine and low-dose cytarabine in palliative patients with acute myeloid leukemia and high bone marrow blast counts – a retrospective single-center experience. Eur J Haematol. 2014;93(2):112–7. doi: 10.1111/ejh.12308.
  46. Field T, Perkins J, Huang Y, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  47. Gerds AT, Gooley TA, Estey EH, et al. Pretransplantation Therapy with Azacitidine vs Induction Chemotherapy and Posttransplantation Outcome in Patients with MDS. Biol Blood Marrow Transplant. 2012;18(8):1211–8. doi: 10.1016/j.bbmt.2012.01.009.
  48. Damaj G, Duhamel A, Robin M, et al. Impact of azacitidine before allogeneic stem-cell transplantation for myelodysplastic syndromes: a study by the Societe Francaise de Greffe de Moelle et de Therapie-Cellulaire and the Groupe-Francophone des Myelodysplasies. J Clin Oncol. 2012;30(36):4533–40. doi: 10.1200/jco.2012.44.3499.
  49. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogeneous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116(23):5420–31. doi: 10.1002/cncr.25500.
  50. Jabbour E, Giralt S, Kantarjian H, et al. Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia. Cancer. 2009;115(9):1899–905. doi: 10.1002/cncr.24198.
  51. Schroeder T, Czibere A, Platzbecker U, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013 27(6), 1229–35. doi: 10.1038/leu.2013.7.
  52. Lubbert M, Bertz H, Wasch R, et al. Efficacy of a 3-day, low-dose treatment with 5-azacytidine followed by donor lymphocyte infusions in older patients with acute myeloid leukemia or chronic myelomonocytic leukemia relapsed after allografting. Bone Marrow Transplant. 2010;45:627–32. doi: 10.1038/bmt.2009.222.
  53. Sanchez-Abarca LI, Gutierrez-Cosio S, Santamaria C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115(1):107–21. doi: 10.1182/blood-2009-03-210393.
  54. Goodyear О, Agathanggelou A, Novitzky-Basso, et al. Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. 2010;116(11):1908–18. doi: 10.1182/blood-2009-11-249474.
  55. Atanackovich D, Luetkens T, Kloth B, et al. Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. Am J Hematol. 2011;86(11):918–22. doi: 10.1002/ajh.22141.
  56. Kroger N, Bacher U, Bader P, et al. NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the committee on disease-specific methods and strategies for monitoring relapse following allogeneic stem cell transplantation: II. Chronic leukemias, myeloproliferative neoplasms, and lymphoid malignancies. Biol Blood Marrow Transplant. 2010;16(10):1325–46. doi: 10.1016/j.bbmt.2010.06.008.
  57. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia. 2012;26(3):381–9. doi: 10.1038/leu.2011.234.
  58. Sockel K, Wermke M, Radke J, et al. Minimal Residual Disease-Directed Preemptive Treatment With Azacitidine In Patients With NPM1-Mutant Acute Myeloid Leukemia And Molecular Relapse. Haematologica. 2011;96(10):1568–70. doi: 10.3324/haematol.2011.044388.
  59. The MDS Foundation. New MDS Clinical Trials. [Internet] Available from: http://www.mds-foundation.org/clinical-trial-announcements/#New-MDS-Clinical-Trials. (accessed 17.05.2016).

 

Application of Modern Genome Technologies in Treatment of Lymphomas

MV Nemtsova1, MV Maiorova2

1 Russian Medical Academy of Postgraduate Education, 2/1 Barrikadnaya str., Moscow, Russian Federation, 125993

2 PA Hertzen Moscow Oncology Research Institute, 3 2-y Botkinskii pr-d, Moscow, Russian Federation, 125284

For correspondence: Marina Vyacheslavovna Nemtsova, DSci, Professor, 2/1 Barrikadnaya str., Moscow, Russian Federation, 125993; Tel: +7(499)252-21-04; e-mail: nemtsova_m_v@mail.ru

For citation: Nemtsova MV, Maiorova MV. Application of Modern Genome Technologies in Treatment of Lymphomas. Clinical oncohematology. 2016;9(3):265-70 (In Russ).

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


ABSTRACT

Modern achievements in genomics and cancer biology have provided an unprecedented body of knowledge regarding the molecular pathogenesis of lymphoma. Genome-wide association studies and modern computer technologies demonstrated that various histological and immunomorphological subtypes of lymphomas differ at the molecular level, and result from various oncogenic mechanisms. It is clear that the variability of clinical symptoms presented by patients with lymphomas is based on the heterogeneity of tumor cells and features of the molecular pathogenesis. Based on data obtained, strategies for the development of new drugs for treatment of lymphoma have been proposed, including identification of the molecular pathogenesis, assessment of the significance of each stage for the development of tumors and synthesis of a drug with a targeted effect. As a result, several new classes of molecular targeted agents for treatment of lymphomas have been proposed and are being tested in clinical trials. In the modern era of personalized medicine, correct targeted therapy for each type of lymphoma characterized by a unique molecular mechanism of tumor formation is a major challenge in lymphoma treatment.


Keywords: lymphoma, genes expression profile, microRNA, signaling pathways, NF-kB.

Received: February 13, 2016

Accepted: March 14, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Intlekofer AM, Younes A. Precision therapy for lymphoma—current state and future directions. Nat Rev Clin Oncol. 2014;11(10):585–96. doi: 10.1038/nrclinonc.2014.137.
  2. Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma—treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11(1):12–23. doi: 10.1038/nrclinonc.2013.197.
  3. Borchmann P, Eichenauer DA, Engert A. State of the art in the treatment of Hodgkin lymphoma. Nat Rev Clin Oncol. 2012;9(8):450–9. doi: 10.1038/nrclinonc.2012.91.
  4. Campo E, Swerdlow SH, Harris NL, et al. The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood. 2011;117(19):5019–32. doi: 10.1182/blood-2011-01-293050.
  5. Немцова М.В., Кушлинский Н.Е. Достижения высокотехнологичных геномных методов для практической онкологии. Медицинский алфавит. 2015;1(2):10–3.
    [Nemtsova MV, Kushlinskii NE. The achievement of high-genomic methods for practical oncology. Meditsinskii alfavit. 2015;1(2):10–3. (In Russ)]
  6. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503–11. doi: 10.1038/35000501.
  7. Lenz G, Dave SS, Xiao W, et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med. 2008;359(22):2313–23. doi: 10.1056/NEJMoa0802885.
  8. Ngo VN, Davis RE, Lamy L, et al. A loss-of-function RNA interference screen for molecular targets in cancer. Nature. 2006;441(7089):106–10. doi: 10.1038/nature04687.
  9. Compagno M, Lim WK, Grunn A, et al. Mutations of multiple genes cause deregulation of NF-kappa B in diffuse large B-cell lymphoma. Nature. 2009;459(7247):717–21. doi: 10.1038/nature07968.
  10. Lenz G, Davis RE, Ngo VN, et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science. 2008;319(5870):1676–9. doi: 10.1126/science.1153629.
  11. Wilson WH. The Bruton’s tyrosine kinase (BTK) inhibitor, ibrutinib (PCI-32765), has preferential activity in the ABC subtype of relapsed/refractory de novo diffuse large B-cell lymphoma (DLBCL): interim results of a multicentre, open-label, phase 2 study. Blood. 2012;120: Abstract 686.
  12. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507–16. doi: 10.1056/NEJMoa1306220.
  13. Morin RD, Mendez-Lago M, Mungall AJ, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476(7360):298–303. doi: 10.1038/nature10351.
  14. Chi P, Allis CD, Wang GG. Covalent histone modifications—miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 2010;10(7):457–69. doi: 10.1038/nrc2876.
  15. Pasqualucci L, Dominguez-Sola D, Chiarenza A, et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature. 2011;471(7337):189–95. doi: 10.1038/nature09730.
  16. Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. 2009;27(32):5459–68. doi: 10.1200/jco.2009.22.1291.
  17. Yap DB, Chu J, Berg T, et al. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood. 2011;117(8):2451–9. doi: 10.1182/blood-2010-11-321208.
  18. Beguelin W, Popovic R, Teater M, et al. EZH2 is required for germinal centre formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell. 2013;23(5):677–92. doi: 10.1016/j.ccr.2013.04.011.
  19. Cairns RA, Iqbal J, Lemonnier F, et al. IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma. Blood. 2012;119(8):1901–3. doi: 10.1182/blood-2011-11-391748.
  20. Wang F, Travins J, DeLaBarre B, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 2013;340(6132):622–6. doi: 10.1126/science.1234769.
  21. Pasqualucci L, Khiabanian H, Fangazio M, et al. Genetics of follicular lymphoma transformation. Cell Rep., 2014;6(1):130–40. doi: 10.1016/j.celrep.2013.12.027.
  22. Schmitz R, Young RM, Ceribelli M, et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature. 2012;490(7418):116–20. doi: 10.1038/nature11378.
  23. Bea S, Valdes-Mas R, Navarro A, et al. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci USA. 2013;110(45):18250–5. doi: 10.1073/pnas.1314608110.
  24. Rossi D, Trifonov V, Fangazio M, et al. The coding genome of splenic marginal zone lymphoma: activation of NOTCH2 and other pathways regulating marginal zone development. J Exp Med. 2012;209(9):1537–51. doi: 10.1084/jem.20120904.
  25. Новикова М.В., Рыбко В.А., Хромова Н.В. и др. Роль белков Notch в процессах канцерогенеза. Успехи молекулярной онкологии. 2015;2(3):30–42. doi: 10.17650/2313-805X-2015-2-3-30-42.
    [Novikova MV, Rybko VA, Khromova NV, et al. The role of Notch pathway in carcinogenesis. Advances in molecular oncology. 2015;2(3):30–42. doi: 10.17650/2313-805X-2015-2-3-30-42. (In Russ)]
  26. Zhang J, Grubor V, Love CL, et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc Natl Acad Sci USA. 2013;110(4):1398–403. doi: 10.1073/pnas.1205299110.
  27. Rahal R, Frick M, Romero R, et al. Pharmacological and genomic profiling identifies NF-kappaB-targeted treatment strategies for mantle cell lymphoma. Nat Med. 2014;20(1):87–92. doi: 10.1038/nm.3435.
  28. Anderson K, Lutz C, van Delft FW, et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature. 2011;469(7330):356–61. doi: 10.1038/nature09650.
  29. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–10. doi: 10.1038/nature10738.
  30. Arcaini L, Rossi D. Nuclear Factor-kB Dysregulation In Splenic Marginal Zone Lymphoma: New Therapeutic Opportunities. Haematologica. 2012;97(5):638–40. doi: 10.3324/haematol.2011.058362.