Preview

Российский журнал детской гематологии и онкологии (РЖДГиО)

Расширенный поиск

Злокачественные рабдоидные опухоли мягких тканей у детей. Обзор литературы

https://doi.org/10.17650/2311-1267-2017-4-4-56-66

Аннотация

Актуальным вопросом детской онкологии является поиск новых методов терапии пациентов со злокачественными рабдоидными опухолями (ЗРО) – одним из видов редких злокачественных  новообразований детского возраста. Недостаточная осведомленность, сложности в диагностике  приводят к тому, что диагноз подтверждается на распространенных стадиях процесса и  является в большинстве случаев фатальным. В обзоре литературы представлены современные  представления о возникновении ЗРО, подходы к диагностике и терапии пациентов.

Об авторе

М. В. Телешова
ФГБУ «НМИЦ ДГОИ им. Дмитрия Рогачева» Минздрава России
Россия
Россия, 117997, Москва, ул. Саморы Машела, 1


Список литературы

1. Heck J.E., Lombardi C.A., Cockburn M. et al. Epidemiology of rhabdoid tumors of early childhood. Pediatr Blood Cancer 2013;60(1):77–81. doi: 10.1002/pbc.24141.

2. Bourdeaut F., Fréneaux .P, Thuille B. et al. Extra-renal non-cerebral rhabdoid tumours. Pediatr Blood Cancer 2008;51(3):363–8. doi: 10.1002/pbc.21632.

3. Kodet R., Newton W.A., Sachs N. et al. Rhabdoid tumors of soft tissues: a clinicopathologic study of 26 cases enrolled on the Intergroup Rhabdomyosarcoma Study. Hum Pathol 1991;22(7):67–84. PMID: 1712749.

4. Benesch M., Bartelheim K., Fleischhack G. et al. High-dose chemotherapy (HDCT) with auto-SCT in children with atypical teratoid/ rhabdoid tumors (AT/RT): a report from the European Rhabdoid Registry (EU-RHAB). Bone Marrow Transplant 2014;49(3):370–5. doi: 10.1038/bmt.2013.208.

5. Sultan I., Qaddoumi I., Rodriguez-Galindo C. et al. Age, stage, and radiotherapy, but not primary tumor site, affects the outcome of patients with malignant rhabdoid tumors. Pediatr Blood Cancer 2010;54(1):35–40. doi: 10.1002/pbc.22285.

6. Van den Heuvel-Eibrink M.M., van Tinteren H., Rehorst H. et al. Malignant rhabdoid tumours of the kidney (MRTKs), registered on recent SIOP protocols from 1993 to 2005: A report of the SIOP renal tumour study group. Pediatr Blood Cancer 2011;56(5):733–7. doi: 10.1002/pbc.22922.

7. Tomlinson G.E., Breslow N.E., Dome J. et al. Rhabdoid tumor of the kidney in the National Wilms’ Tumor Study: age at diagnosis as a prognostic factor. J Clin Oncol 2005;23(30):7641– 5. DOI: 10.1200/JCO.2004.00.8110.

8. Reinhard H., Reinert J., Beier R. et al. Rhabdoid tumors in children: prognostic factors in 70 patients diagnosed in Germany. Oncol Rep 2008;19(3):819–23. PMID: 18288421.

9. Horazdovsky R., Manivel J.C., Cheng E.Y. Surgery and actinomycin improve survival in malignant rhabdoid tumor. Sarcoma 2013;2013:315170. doi: 10.1155/2013/315170.

10. Kerl K., Holsten T., Frühwald M.C. Rhabdoid tumors: clinical approaches and molecular targets for innovative therapy. Pediatr Hematol Oncol 2013;30(7):587–604. doi: 10.3109/08880018.2013.791737.

11. Beckwith J.B., Palmer N.F. Histopathology and prognosis of Wilms tumors: results from the First National Wilms’ Tumor Study. Cancer 1978;41(5):1937–48. PMID: 206343.

12. Haas J.E., Palmer N.F., Weinberg A.G., Beckwith J.B. Ultrastructure of malignant rhabdoid tumor of the kidney. A distinctive renal tumor of children. Hum Pathol 1981;12(7):646–57. PMID: 7275104.

13. Beckwith J.B. Wilms’ tumor and other renal tumors of childhood: a selective review from the National Wilms’ Tumor Study Pathology Center. Hum Pathol 1983;14(6):481– 92. PMID: 6303938.

14. Briner J., Bannwart F., Kleihues P. et al. Malignant small cell tumor of the brain with intermediate filaments - a case of a primary cerebral rhabdoid tumor (abstract). Pediatr Pathol 1985;3:117–8.

15. Gonzalez-Crussi F., Goldschmidt R.A., Hsueh W., Trujillo Y.P. Infantile sarcoma with intracytoplasmic filamentous inclusions: distinctive tumor of possible histiocytic origin. Cancer 1982;49(11):2365–75. PMID: 7200394.

16. Kent A.L., Mahoney D.H., Gresik M.V., Steuber C.P., Fernbach D.J. Malignant rhabdoid tumor of the extremity. Cancer 1987;60(5):1056–9. PMID: 3607724.

17. Small EJ, Gordon GJ, Dahms BB. Malignant rhabdoid tumor of the heart in an infant. Cancer 1985;55(12):2850–3. PMID: 3995491.

18. Tsokos M., Kouraklis G., Chandra R.S., Bhagavan B.S., Triche T.J. Malignant rhabdoid tumor of the kidney and soft tissues. Evidence for a diverse morphological and immunocytochemical phenotype. Arch Pathol Lab Med 1989;113(2):115–20. PMID: 2916902.

19. Biggs P.J., Garen P.D., Powers J.M., Garvin A.J. Malignant rhabdoid tumor of the central nervous system. Hum Pathol 1987;18(4):332–7. PMID: 3030922.

20. Weeks D.A., Beckwith J.B., Mierau G.W., Luckey D.W. Rhabdoid tumor of kidney. A report of 111 cases from the National Wilms’ Tumor Study Pathology Center. Am J Surg Pathol 1989;13(6):439–58. PMID: 2543225.

21. Fanburg-Smith J.C., Hengge M., Hengge U.R., Smith J.S., Miettinen M. Extrarenal rhabdoid tumors of soft tissue: a clinicopathologic and immunohistochemical study of 18 cases. Ann Diagn Pathol 1998;2(6):351–62. PMID: 9930572.

22. Ota S., Crabbe D.C., Tran T.N., Triche T.J., Shimada H. Malignant rhabdoid tumor. A study with two established cell lines. Cancer 1993;71(9):2862–72. PMID: 8385567.

23. Parham D.M., Peiper S.C., Robicheaux G., Ribeiro R.C., Douglass E.C. Malignant rhabdoid tumor of the liver: evidence for epithelial differentiation. Arch Pathol Lab Med 1988;112(1):61–4. PMID: 2447852.

24. Douglass E.C., Valentine M., Rowe S.T. et al. Malignant rhabdoid tumor: a highly malignant childhood tumor with minimal karyotypic changes. Genes Chromosomes Cancer 1990;2(3):210–6. PMID: 1964081.

25. Karnes P.S., Tran T.N., Cui M.Y. et al. Establishment of a rhabdoid tumor cell line with a specific chromosomal abnormality, 46,XY,t(11;22)(p15.5;q11.23). Cancer Genet Cytogenet 1991;56(1):31–8. PMID: 1747867.

26. Biegel J.A., Rorke L.B., Packer R.J., Emanuel B.S. Monosomy 22 in rhabdoid or atypical tumors of the brain. J Neurosurg 1990;73(5):710–4. doi: 10.3171/jns.1990.73.5.0710.

27. Shashi V., Lovell M.A., von Kap-herr C., Waldron P., Golden W.L. Malignant rhabdoid tumor of the kidney: involvement of chromosome 22. Genes Chromosomes Cancer 1994;10(1):49–54. PMID: 7519873.

28. Schofield D.E., Beckwith J.B., Sklar J. Loss of heterozygosity at chromosome regions 22q11-12 and 11p15.5 in renal rhabdoid tumors. Genes Chromosomes Cancer 1996;15(1):10–7. doi: 10.1002/(SICI)1098-2264(199601)15:1<10::AID-GCC2>3.0.CO;2-7.

29. Rousseau-Merck M.F., Versteege I., Legrand I. et al. hSNF5/INI1 inactivation is mainly associated with homozygous deletions and mitotic recombinations in rhabdoid tumors. Cancer Res 1999;59(13):3152–6. PMID: 10397258.

30. Bruch L.A., Hill D.A., Cai D.X. et al. A role for fluorescence in situ hybridization detection of chromosome 22q dosage in distinguishing atypical teratoid/rhabdoid tumors from medulloblastoma/central primitive neuroectodermal tumors. Hum Pathol 2001;32(2):156– 62. doi: 10.1053/hupa.2001.21572.

31. Versteege I., Sévenet N., Lange J. et al. Truncating mutation of hSNF5/INI1 in aggressive pediatric cancer. Nature 1998;394(6689):203–6. doi: 10.1038/28212.

32. Peterson CL. Multiple SWItches to turn on chromatin? Curr Opin Genet Dev 1996;6(2):171–5. PMID: 8722173.

33. Biegel J.A., Kalpana G., Knudsen E.S. et al. The role of INI1 and the SWI/SNF complex in the development of rhabdoid tumors: meeting summary from the workshop on childhood atypical teratoid/rhabdoid tumors. Cancer Res 2002;62(1):323–8. PMID: 11782395.

34. Biegel J.A., Tan L., Zhang F. et al. Alterations of the hSNF5/INI1 gene in central nervous system atypical teratoid/rhabdoid tumors and renal and extrarenal rhabdoid tumors. Clin Cancer Res 2002;8(11):3461–7. PMID: 12429635.

35. Jackson E.M., Sievert A.J., Gai X. et al. Genomic analysis using high-density single nucleotide polymorphism-based oligonucleotide arrays and multiplex ligation-dependent probe amplification provides a comprehensive analysis of INI1/SMARCB1 in malignant rhabdoid tumors. Clin Cancer Res 2009;15(6):1923–30. doi: 10.1158/1078-0432.CCR-08-2091.

36. Frühwald M.C., Hasselblatt M., Wirth S. et al. Non-linkage of familial rhabdoid tumors to SMARCB1 implies a second locus for the rhabdoid tumor predisposition syndrome. Pediatr Blood Cancer 2006;47(3):273–8. doi: 10.1002/pbc.20526.

37. Schneppenheim R., Frühwald M.C., Gesk S. et al. Germline nonsense mutation and somatic inactivation of SMARCA4/ BRG1 in a family with rhabdoid tumor predisposition syndrome. Am J Hum Genet 2010;86(2):279–84. doi: 10.1016/j.ajhg.2010.01.013.

38. Eaton K.W., Tooke L.S., Wainwright L.M., Judkins A.R., Biegel J.A. Spectrum of SMARCB1/INI1 mutations in familial and sporadic rhabdoid tumors. Pediatr Blood Cancer 2011;56(1):7–15. doi: 10.1002/pbc.22831.

39. Witkowski L., Lalonde E., Zhang J. et al. Familial rhabdoid tumour 'avant la lettre' - from pathology review to exome sequencing and back again. J Pathol 2013;231(1):35-43. doi: 10.1002/path.4225.

40. Bourdeaut F., Lequin D., Brugières L. et al. Frequenth SNF5/ INI1 germline mutations in patients with rhabdoid tumor. Clin Cancer Res 2011;17(1):31–8. doi: 10.1158/1078-0432.CCR-10-1795.

41. Teplick A., Kowalski M., Biegel J.A., Nichols K.E. Screening in cancer predisposition syndromes: guidelines for the general pediatrician. Eur J Pediatr 2011;170(3):285–94. doi: 10.1007/s00431-010-1377-2.

42. Gigante L., Paganini I., Frontali M. et al. Rhabdoid tumor predisposition syndrome caused by SMARCB1 constitutional deletion: prenatal detection of new case of recurrence in siblings due to gonadal mosaicism. Fam Cancer 2016;15(1):123–6. doi: 10.1007/s10689-015-9836-6.

43. Bruggers C.S., Bleyl S.B., Pysher T. et al. Clinicopathologic comparison of familial versus sporadic atypical teratoid/rhabdoid tumors (AT/RT) of the central nervous system. Pediatr Blood Cancer 2011;56(7):1026–31. doi: 10.1002/pbc.22757.

44. Bonnin J.M., Rubinstein L.J., Palmer N.F., Beckwith J.B. The association of embryonal tumors originating in the kidney and in the brain. A report of seven cases. Cancer 1984;54(10):2137–46. PMID: 6091860.

45. Hulsebos T.J., Plomp A.S., Wolterman R.A. et al. Germline mutation of INI1/SMARCB1 in familial schwannomatosis. Am J Hum Genet 2007;80(4):805–10. doi: 10.1086/513207.

46. Hasselblatt M., Oyen F., Gesk S. et al. Cribriform neuroepithelial tumor (CRINET): a non rhabdoid ventricular tumor with INI1 loss and relatively favorable prognosis. J Neuropathol Exp Neurol 2009;68(12):1249–55. doi: 10.1097/NEN.0b013e3181c06a51.

47. Hoot A.C., Russo P., Judkins A.R., Perlman E.J., Biegel J.A. Immunohistochemical analysis of hSNF5/INI1 distinguishes renal and extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Surg Pathol 2004;28(11):1485–91. PMID: 15489652.

48. Cheng J.X., Tretiakova M., Gong C. et al. Renal medullary carcinoma: rhabdoid features and the absence of INI1 expression as markers of aggressive behavior. Mod Pathol 2008;21(6):647–52. doi: 10.1038/modpathol.2008.44.

49. Ogino S, Ro JY, Redline RW. Malignant rhabdoid tumor: a phenotype? An entity? A controversy revisited. Adv Anat Pathol 2000;7(3):181–90. PMID: 10809224.

50. Wick MR, Ritter JH, Dehner LP. Malignant rhabdoid tumors: a clinicopathologic review and conceptual discussion. Semin Diagn Pathol 1995;12(3):233–48. PMID: 8545590.

51. Fuller C.E. All things rhabdoid and SMARC: An enigmatic exploration with Dr. Louis P. Dehner. Semin Diagn Pathol 2016;33(6):427–40. doi: 10.1053/j.semdp.2016.08.003.

52. Buccoliero A.M., Castiglione F., Rossi Degl’Innocenti D. et al. Pediatric rhabdoid meningioma: a morphological, immunohistochemical, ultrastructural and molecular case study. Neuropathology 2011;31(1):59–65. doi: 10.1111/j.1440-1789.2010.01113.x.

53. Ginat D.T., Cipriani N.A., Purakal A. et al. Disseminated Malignant Rhabdoid Tumor of the Head and Neck. Head Neck Pathol 2017;11(2):224–7. doi: 10.1007/s12105-016-0754-1.

54. Chung C.T., Liu Y.L., Cheng C.J. et al. Extrarenal rhabdoid tumor presented with an immobile arm in a one-year-old boy. Brain Dev 2017;39(8):717–21. doi: 10.1016/j.braindev.2017.04.001.

55. Stolten M., Betrand K., Lotterman C., Warrier R. Neonate With Paraplegia. Clin Pediatr (Phila) 2016;55(2):199–201. doi: 10.1177/0009922815595063.

56. Yasui N., Yoshida A., Kobayashi E., Nakatani F., Kawamoto H. Successful Treatment of Extra-Renal Noncerebral Rhabdoid Tumors with VIDE. Pediatr Blood Cancer 2016;63(2):352– 4. doi: 10.1002/pbc.25777.

57. Nguyen H., Stelling A., Kuramoto A., Patel C., Keller J. Malignant rhabdoid tumor of the liver: Findings at US, CT, and MRI, with histopathologic correlation. Radiol Case Rep 2015;9(1):e00031. doi: 10.2484/rcr.v9i1.854.

58. Hernández-Marqués C., Lassaletta A., Cormenzana M., García-Esparza E., Madero- López L. All children with malignant rhabdoid tumors should be treated initially with intensive chemotherapy. Pediatr Hematol Oncol 2015;32(3):193–8. doi: 10.3109/08880018.2014.914112.

59. Oita S., Terui K., Komatsu S. et al. Malignant rhabdoid tumor of the liver: a case report and literature review. Pediatr Rep 2015;7(1):5578. doi: 10.4081/pr.2015.5578.

60. Kerl K., Oyen F., Leuschner I. et al. Detection of SMARCB1 loss in ascites cells in the diagnosis of an abdominal rhabdoid tumor. Pediatr Blood Cancer 2015;62(5):897–900. doi: 10.1002/pbc.25412.

61. Gupta R.K., Batra V.V., Das M.C., Sharma A., Narang P. Malignant extra-renal rhabdoid tumor with unusual presentation: A report of two cases. J Cancer Res Ther 2015;11(4):963– 6. doi: 10.4103/0973-1482.163668.

62. Sterling M.E., Long C.J., Bosse K.R., Bagatell R., Shukla A.R. A rapid progression of disease after surgical excision of a malignant rhabdoid tumor of the bladder. Urology 2015;85(3):664–6. doi: 10.1016/j.urology.2014.11.011.

63. Asada N., Kato I., Daifu T. et al. Good response to chemotherapy spares irradiation for extrarenal rhabdoid tumor conferring better activities of daily living. J Pediatr Hematol Oncol 2015;37(1):e57–9. doi: 10.1097/MPH.0000000000000150.

64. Chakrapani A.L., White C.R., Korcheva V. et al. Congenital extrarenal malignant rhabdoid tumor in an infant with distal 22q11.2 deletion syndrome: the importance of SMARCB1. Am J Dermatopathol 2012;34(6):e77–80. doi: 10.1097/DAD.0b013e31825793c3.

65. Mulay K., Honavar S.G. Primary, orbital, malignant extra-renal, non-cerebral rhabdoid tumour. Orbit 2014;33(4):292–4. doi: 10.3109/01676830.2014.900088.

66. Kachanov D., Teleshova M., Kim E. et al. Malignant rhabdoid tumor of the liver presented with initial tumor rupture. Cancer Genet 2014;207(9):412–4. doi: 10.1016/j.cancergen.2014.04.006.

67. Bartelheim K., Sumerauer D., Behrends U. et al. Clinical and genetic features of rhabdoid tumors of the heart registered with the European Rhabdoid Registry (EU-RHAB). Cancer Genet 2014;207(9):379–83. doi: 10.1016/j.cancergen.2014.04.005.

68. Tam H.K.Y., Li A., Lee P.S.F. et al. Infantile Intravesical Malignant Rhabdoid Tumour. Hong Kong J Radiol 2013;16:e21–5.

69. Hong C.R., Kang H.J., Ju H.Y. et al. Extra-cranial Malignant Rhabdoid Tumor in Children: A Single Institute Experience. Cancer Res Treat 2015;47(4):889–96. doi: 10.4143/crt.2013.176.

70. Uwineza A., Gill H., Buckley P. et al. Rhabdoid tumor: the Irish experience 1986– 2013. Cancer Genet 2014;207(9):398–402. doi: 10.1016/j.cancergen.2014.05.015.

71. Parham D.M., Weeks D.A., Beckwith J.B. The clinicopathologic spectrum of putative extrarenal rhabdoid tumors. An analysis of 42 cases studied with immunohistochemistry or electron microscopy. Am J Surg Pathol 1994;18(10):1010–29. PMID: 8092393.

72. Gururangan S., Bowman L.C., Parham D.M. et al. Primary extracranial rhabdoid tumors. Clinicopathologic features and response to ifosfamide. Cancer 1993;71(8):2653–9. PMID: 8453588.

73. Gündüz K., Shields J.A., Eagle R.C. et al. Malignant rhabdoid tumor of the orbit. Arch Ophthalmol 1998;116(2):243–6. PMID: 9488282.

74. Trobaugh-Lotrario A.D., Finegold M.J., Feusner J.H. Rhabdoid Tumors of the Liver: Rare, Aggressive, and Poorly Responsive to Standard Cytotoxic Chemotherapy. Pediatr Blood Cancer 2011;57(3):423–8. doi: 10.1002/pbc.22857.

75. Kwon J.Y., Park K.I., Lee K.S., Yang W.I., Kim Y.H. Prenatal detection of congenital malignant extrarenal rhabdoid tumor primarily involving the right upper extremity that ruptured in utero. Prenat Diagn 2009;29(8):819–21. doi: 10.1002/pd.2286.

76. Yurdakul Z., Berrak S.G., Bilgen H. et al. Congenital disseminated malignant rhabdoid tumor of the soft tissue. Pediatr Blood Cancer 2007;49(3):364–5. doi: 10.1002/pbc.20960.

77. de Tar M., Sanford Biggerstaff J. Congenital renal rhabdoid tumor with placental metastases: immunohistochemistry, cytogenetic, and ultrastructural findings. Pediatr Dev Pathol 2006;9(2):161–7. doi: 10.2350/08-05-0090.1.

78. Hsueh C., Kuo T.T. Congenital malignant rhabdoid tumor presenting as a cutaneous nodule: report of 2 cases with review of the literature. Arch Pathol Lab Med 1998;122(12):1099–102. PMID: 9870859.

79. Issacs H. Fetal and neonatal rhabdoid tumor. J Pediatr Surg 2010;45(3):619–26. doi: 10.1016/j.jpedsurg.2009.12.011.

80. Kato M., Koh K., Oshima K. et al. Longterm survivor of relapsed stage IV malignant rhabdoid tumor of the kidney. Pediatr Int 2013;55(2):245–8. doi: 10.1111/j.1442-200X.2012.03663.x.

81. Wagner L., Hill D.A., Fuller C. et al. Treatment of metastatic rhabdoid tumor of the kidney. J Pediatr Hematol Oncol 2002;24(5):385–8. PMID: 12142788.

82. Yamamoto M., Suzuki N., Hatakeyama N. et al. Treatment of stage IV malignant rhabdoid tumor of the kidney (MRTK) with ICE and VDCy: a case report. J Pediatr Hematol Oncol 2006;28(5):286–9. doi: 10.1097/01.mph.0000212901.84146.5a.

83. Dallorso S., Dini G., Ladenstein R. et al. Evolving role of myeloablative chemotherapy in the treatment of childhood brain tumours. Bone Marrow Transplant 2005;35 Suppl 1:S31–4. doi: 10.1038/sj.bmt.1704841.

84. Garrè M.L., Dallorso S., Massimino M. et al. Atypical teratoid/rhabdoid tumour of the central nervous system (CNS): Final results of the first Italian Cooperative Study for the very young children. Haematol Rep 2006;2:6.

85. Madigan C.E., Armenian S.H., Malogolowkin M.H., Mascarenhas L. Extracranial malignant rhabdoid tumors in childhood: the Childrens Hospital Los Angeles experience. Cancer 2007;110(9):2061–6. doi: 10.1002/cncr.23020.

86. Watanabe H., Watanabe T., Kaneko M. et al. Treatment of unresectable malignant rhabdoid tumor of the orbit with tandem high-dose chemotherapy and gamma-knife radiosurgery. Pediatr Blood Cancer 2006;47(6):846–50. doi: 10.1002/pbc.20699.

87. Gardner S.L., Asgharzadeh S., Green A. et al. Intensive induction chemotherapy followed by high dose chemotherapy with autologous hematopoietic progenitor cell rescue in young children newly diagnosed with central nervous system atypical teratoid rhabdoid tumors. Pediatr Blood Cancer 2008;51(2):235–40. doi: 10.1002/pbc.21578.

88. Squire S.E., Chan M.D., Marcus K.J. et al. Atypical teratoid/rhabdoid tumor: the controversy behind radiation therapy. J Neurooncol 2007;81(1):97–111. doi: 10.1007/s11060-006-9196-z.

89. Seeringer A., Bartelheim K., Kerl K. et al. Feasibility of intensive multimodal therapy in infants affected by rhabdoid tumors - experience of the EU-RHAB registry. Klin Padiatr 2014;226(3):143–8. doi: 10.1055/s-0034-1368719.

90. Lee R.S., Stewart C., Carter S.L. et al. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J Clin Invest 2012;122(8):2983–8. doi: 10.1172/JCI64400.

91. Jones S., Wang T.L., Shih Ie.M. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 2010;330(6001):228–31. doi: 10.1126/science.1196333.

92. Varela I., Tarpey P., Raine K. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 2011;469(7331):539–42. doi: 10.1038/nature09639.

93. Zhang Z.K., Davies K.P., Allen J. et al. Cell cycle arrest and repression of cyclin D1 transcription by INI1/hSNF5. Mol Cell Biol 2002;22(16):5975–88. PMCID: PMC133966.

94. Fujisawa H., Misaki K., Takabatake Y., Hasegawa M., Yamashita J. Cyclin D1 is overexpressed in atypical teratoid/rhabdoid tumor with hSNF5/INI1 gene inactivation. J Neurooncol 2005;73(2):117–24. doi: 10.1007/s11060-004-4276-4.

95. Das B.C., Smith M.E., Kalpana G.V. Design, synthesis of novel peptidomimetic derivatives of 4-HPR for rhabdoid tumors. Bioorg Med Chem Lett 2008;18(14):4177–80. doi: 10.1016/j.bmcl.2008.05.097.

96. Moreno N., Kerl K. Preclinical Evaluation of Combined Targeted Approaches in Malignant Rhabdoid Tumors. Anticancer Res 2016;36(8):3883–7. PMID: 27466490.

97. Flaherty K.T., Lorusso P.M., Demichele A. et al. Phase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor PD 0332991, administered using a 21-day schedule in patients with advanced cancer. Clin Cancer Res 2012;18(2):568–76. doi: 10.1158/1078-0432.CCR-11-0509.

98. Jiang J., Hui C.C. Hedgehog signaling in development and cancer. Dev Cell 2008;15(6):801–12. doi: 10.1016/j.devcel.2008.11.010.

99. Jagani Z., Mora-Blanco E.L., Sansam C.G. et al. Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway. Nat Med 2010;16(12):1429–33. doi: 10.1038/nm.2251.

100. Kinzler K.W., Bigner S.H., Bigner D.D. et al. Identification of an amplified, highly expressed gene in a human glioma. Science 1987;236(4797):70–3. PMID: 3563490.

101. Northcott P.A., Nakahara Y., Wu X. et al. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet 2009;41(4):465–72. doi: 10.1038/ng.336.

102. LoRusso P.M., Rudin C.M., Reddy J.C. et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res 2011;17(8):2502–11. doi: 10.1158/1078-0432.CCR-10-2745.

103. Rodon J., Tawbi H.A., Thomas A.L. et al. A phase I, multicenter, open-label, firstin- human, dose-escalation study of the oral smoothened inhibitor sonidegib (LDE225) in patients with advanced solid tumors. Clin Cancer Res 2014;20(7):1900–9. doi: 10.1158/1078-0432.CCR-13-1710.

104. Ghavamzadeh A., Alimoghaddam K., Rostami S. et al. Phase II study of single- agent arsenic trioxide for the front-line therapy of acute promyelocytic leukemia. J Clin Oncol 2011;29(20):2753–7. doi: 10.1200/JCO.2010.32.2107.

105. Huang C., Xu M., Zhu B. Epigenetic inheritance mediated by histone lysine methylation: maintaining transcriptional states without the precise restoration of marks? Philos Trans R Soc Lond B Biol Sci 2013;368(1609):20110332. doi: 10.1098/rstb.2011.0332.

106. Yoo K.H., Hennighausen L. EZH2 methyltransferase and H3K27 methylation in breast cancer. Int J Biol Sci 2012;8(1):59–65. PMCID: PMC3226033.

107. Kikuchi J., Takashina T., Kinoshita I. et al. Epigenetic therapywith 3-deazaneplanocin A, an inhibitor of the histone methyltransferase EZH2, inhibits growth of nonsmall cell lung cancer cells. Lung Cancer 2012;78(2):138–43. doi: 10.1016/j.lungcan.2012.08.003.

108. Knutson S.K., Warholic N.M., Wigle T.J. et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A 2013;110(19):7922–7. doi: 10.1073/pnas.1303800110.

109. Watanabe M., Adachi S., Matsubara H. et al. Induction of autophagy in malignant rhabdoid tumor cells by the histone deacetylase inhibitor FK228 through AIF translocation. Int J Cancer 2009;124(1):55– 67. doi: 10.1002/ijc.23897.

110. Mizushima N., Ohsumi Y., Yoshimori T. Autophagosome formation in mammalian cells. Cell Struct Funct 2002;27(6):421–9. PMID: 12576635.

111. Kanzawa T., Germano I.M., Komata T. et al. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ 2004;11(4):448–57. doi: 10.1038/sj.cdd.4401359.

112. Takeuchi H., Kondo Y., Fujiwara K. et al. Synergistic augmentation of rapamycin- induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors. Cancer Res 2005;65(8):3336–46. DOI: 10.1158/0008-5472.CAN-04-3640.

113. Garcia-Manero G., Tambaro F.P., Bekele N.B. et al. Phase II trial of vorinostat with idarubicin and cytarabine for patients with newly diagnosed acutemyelogenous leukemia ormyelodysplastic syndrome. J Clin Oncol 2012;30(18):2204–10. doi: 10.1200/JCO.2011.38.3265.

114. Lee E.Q., Puduvalli V.K., Reid J.M. et al. Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04–03. Clin Cancer Res 2012;18(21):6032–9. doi: 10.1158/1078-0432.CCR-12-1841.

115. Muscal J.A., Thompson P.A., Horton T.M. et al. A phase I trial of vorinostat and bortezomib in children with refractory or recurrent solid tumors: a Children’s Oncology Group phase I consortium study (ADVL0916). Pediatr Blood Cancer 2013;60(3):390–5. doi: 10.1002/pbc.24271.

116. Kerl K., Ries D., Unland R. et al. The histone deacetylase inhibitor SAHA acts in synergism with fenretinide and doxorubicin to control growth of rhabdoid tumor cells. BMC Cancer 2013;13:286. doi: 10.1186/1471-2407-13-286.

117. Hannak E., Kirkham M., Hyman A.A., Oegema K. Aurora-A kinase is required for centrosome maturation in Caenorhabditis elegans. J Cell Biol 2001;155(7):1109–16. doi: 10.1083/jcb.200108051.

118. Lee S., Cimica V., Ramachandra N., Zagzag D., Kalpana G.V. Aurora A is a repressed effector target of the chromatin remodeling protein INI1/hSNF5 required for rhabdoid tumor cell survival. Cancer Res 2011;71(9):3225–35. doi: 10.1158/0008-5472.CAN-10-2167.

119. Venkataraman S., Alimova I., Tello T. et al. Targeting Aurora kinase A enhances radiation sensitivity of atypical teratoid rhabdoid tumor cells. J Neurooncol 2012;107(3):517– 26. doi: 10.1007/s11060-011-0795-y.

120. Melichar B., Adenis A., Lockhart A.C. et al. Safety and activity of alisertib, an investigational aurora kinase A inhibitor, in patients with breast cancer, small-cell lung cancer, non-small-cell lung cancer, head and neck squamous-cell carcinoma, and gastro- oesophageal adenocarcinoma: a five-arm phase 2 study. Lancet Oncol 2015;16(4):395– 405. doi: 10.1016/S1470-2045(15)70051-3.

121. Barr P.M., Li H., Spier C. et al. Phase II Intergroup Trial of Alisertib in Relapsed and Refractory Peripheral T-Cell Lymphoma and Transformed Mycosis Fungoides: SWOG 1108. J Clin Oncol 2015;33(21):2399–404. doi: 10.1200/JCO.2014.60.6327.

122. DuBois S.G., Marachelian A., Fox E. et al. Phase I Study of the Aurora A Kinase Inhibitor Alisertib in Combination With Irinotecan and Temozolomide for Patients With Relapsed or Refractory Neuroblastoma: A NANT (New Approaches to Neuroblastoma Therapy) Trial. J Clin Oncol 2016;34(12):1368–75. doi: 10.1200/JCO.2015.65.4889.

123. Johann P.D., Erkek S., Zapatka M. et al. Atypical Teratoid/Rhabdoid Tumors Are Comprised of Three Epigenetic Subgroups with Distinct Enhancer Landscapes. Cancer Cell 2016;29(3):379–93. doi: 10.1016/j.ccell.2016.02.001.

124. Birks D.K., Donson A.M., Patel P.R. et al. High expression of BMP pathway genes distinguishes a subset of atypical teratoid/ rhabdoid tumors associated with shorter survival. Neuro Oncol 2011;13(12):1296–307. doi: 10.1093/neuonc/nor140.


Рецензия

Для цитирования:


Телешова М.В. Злокачественные рабдоидные опухоли мягких тканей у детей. Обзор литературы. Российский журнал детской гематологии и онкологии (РЖДГиО). 2017;4(4):56-66. https://doi.org/10.17650/2311-1267-2017-4-4-56-66

For citation:


Teleshova M.V. Malignant rhabdoid tumors of soft tissues in children. Literature review. Russian Journal of Pediatric Hematology and Oncology. 2017;4(4):56-66. (In Russ.) https://doi.org/10.17650/2311-1267-2017-4-4-56-66

Просмотров: 1050


Creative Commons License
Контент доступен под лицензией Creative Commons Attribution 4.0 License.


ISSN 2311-1267 (Print)
ISSN 2413-5496 (Online)