Evolution of concepts for diagnosis and treatment of Burkitt’s lymphoma

T.T. Valiev1 and E.A. Baryakh2

1 Pediatric Oncology and Hematology Institute, N.N. Blokhin Russian Cancer Research Center, RAMS, Moscow, Russian Federation

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


ABSTRACT

The issues of diagnosis and treatment of the most aggressive lymphoid tumor, namely, Burkitt’s lymphoma (BL), are presented in the historical context. The clinical and laboratory features of endemic and sporadic BL variants are described. Possible mechanisms of Epstein-Barr virus and Plasmodium falciparum involvement in BL pathogenesis are suggested. Also, the morphologic, immunologic and cytogenetic BL diagnostic criteria are described. Based on molecular and genetic features, the issues of differential diagnosis with the heterogeneous group of diffuse large B-cell and highly aggressive mature B-cell lymphomas with additional proto-oncogene aberrations («double» and «triple» hit lymphomas) are presented. BL therapy and the role of rituximab in it is emphasized.


Keywords: Burkitt’s lymphoma, clinical presentation, diagnosis, treatment.

Read in PDF (RUS)pdficon


REFERENCES

  1. Burkitt D. A sarcoma involving the jaws in African children. Br. J. Surg. 1958; 46: 218–23.
  2. O’Conor G.T. Malignant lymphoma in African children. A pathology entity. Cancer 1961; 14: 270–83.
  3. Young L.S., Mussay P.G. Epstein-Barr virus and oncogenesis: form latent genes to tumours. Oncogene 2003; 22: 5108–21.
  4. Gulley M.L. Molecular diagnosis of Epstein-Barr virus-related diseases. J. Mol. Diagn. 2001; 3: 1–10.
  5. Epstein M.A., Achong B.R., Barr Y.M. Virus particles in cultured lymphoblasts from Burkitt’s lymphoma. Lancet 1964; 1: 702–3.
  6. Dorfman R.S. Childhood lymphosarcoma in St. Louis, Missouri, clinically and histologically resembling Burkitt tumor. Cancer 1965; 18: 418–30.
  7. Rappapоrt H., Braylan R. Changing concepts in the classification of malignant neoplasms of the hemopoetic system. Int. Acad. Pathol. 1975; 16: 1–19.
  8. The non-Hodgkin’s lymphoma pathologic classification project. National Cancer Institute sponsored study of classification of non-Hodgkin’s lymphomas: summary and description of a working formulation for clinical usage. Cancer 1982; 49: 2112–35.
  9. Lennert K., Feller A.C. Histopathology of Non-Hodgkin’s lymphomas (Based on the Updated Kiel Classification). Berlin: Springer Verlag, 1992: 312.
  10. Harris N.L., Jaffe E.S., Dejbold J. et al. The World Health Organization Classification of Neoplastic Diseases of the Hematopoietic and Lymphoid Tissues. Ann. Oncol. 1999; 10: 1419–32.
  11. Swerdlow S.H., Campo E., Harris N.L. et al. WHO Classification of Tumours of Haematopoetic and Lymphoid tissues, 4th edn. Lyon: IARC Press, 2008: 439.
  12. Agugua N.E., Okeahialam T. Malignant diseases of childhood seen at the University of Nigeria Teaching Hospital, Enugu, Nigeria. East. Afr. Med. J. 1986; 63: 717–23.
  13. De The G. Epidemiologie of Burkitt’s lymphoma: evidence for a casual association with EBV. Epidemiol. Rev. 1979; 36: 692–8.
  14. Oguonu T., Emodi E., Kaine W. Epidemiologie of Burkitt’s lymphoma in Enugu, Nigeria. Ann. Trop. Paediatr. 2002; 22: 369–74.
  15. Kasili E.G. Paediatric malignancy in tropical Africa — a growing concern. East. Afr. Med. J. 1986; 63: 685–6.
  16. Parkin D.M., Stiller C.A., Draper G.J. et al. The international incidence of childhood cancer. Int. J. Cancer. 1988; 42: 511–20.
  17. Magrath I.T. Malignant Non-Hodgkin’s Lymphomas in Children. Pediatr. Oncol. 2002; 119: 661–705.
  18. Zeigler J. Burkitt’s lymphoma. N. Engl. J. Med. 1981; 305: 735–45.
  19. Shannon-Lowe C., Adland E., Bell A.I. et al. Features distinguishing EpsteinBarr virus infections of epithelial cells and B cells: viral genome expression, genome maintenance and genome amplification. J. Virol. 2009; 83: 7749–60.
  20. Richinson A. Epstein-Barr virus. Virus Res. 2002; 82: 109–13.
  21. Thorley-Lawson D.A. Epstein-Barr virus: exploiting the immune system. Nat. Rev. Immunol. 2001; 1: 75–82.
  22. Kelly G., Bell A., Rickinson A. Epstein-Barr virus-associated Burkitt lymphoma genesis selects for down-regulation of the nuclear antigen EBNA2. Nat Med. 2002; 8(10): 1098–104.
  23. Гурцевич В.Э. Роль вируса Эпштейна—Барр в онкогематологиче- ских заболеваниях человека. Клин. онкогематол. 2010; 3(3): 222–35. [Gurtsevich V.E. Role of Epstein—Barr virus in human hematological malignancies. Klin. onkogematol. 2010; 3(3): 222–35. (In Russ.)].
  24. Bornkamm G.W. Epstein-Barr virus and the pathogenesis of Burkitt’s lymphoma: more questions than answers. Int. J. Cancer. 2009; 124(8): 1745–55.
  25. Raab-Traub N., Flynn K. The structure of the termini of the Epstein-Barr virus as a marker of clonal cellular proliferation. Cell 1986; 47: 883–9.
  26. Klein U., Klein G., Ehlin-Henriksson B. et al. Burkitt’s lymphoma is a malignancy of mature B-cells expressing somatically mutated V region genes. Mol. Med. 1995; 1: 495–506.
  27. Henderson S., Rowe M., Gregory C. et al. Induction of bcl-2 expression by Epstein-Barr virus latent membrane protein 1 protects infected B cells from programmed cell death. Cell 1991; 65(7): 1107–15.
  28. Preudhomme C., Dervite I., Wattel E. et al. Clinical significance of p53 mutations in newly diagnosed Burkitt’s lymphoma and acute lymphoblastic leukemia: a report of 48 cases. J. Clin. Oncol. 1995; 13: 812–20.
  29. Schoch C., Reider H., Stollman-Gibbels B. et al. 17p anomalies in lymphoid malignancies: Diagnostic and prognostic implications. Leuk. Lymphoma 1995; 17: 271–9.
  30. Mawanda O.W. Aspects of epidemiological and clinical features of patients with central nervous system Burkitt’s lymphoma in Kenya. East. Afr. Med. J. 2004; 8: 97–103.
  31. Mawanda O.W. Clinical characteristics of Burkitt’s lymphoma seen in Kenyan patients. East. Afr. Med. J. 2004; 8: 78–89.
  32. Donati D., Zhang L.P., Chene A. et al. Identification of a polyclonal B-cell activator in Plasmodium falciparum. Infect. Immun. 2004; 72: 5412–8.
  33. Chene A., Donati D., Guerreiro-Cacais A.O. et al. A molecular link between malaria and Epstein-Barr virus reactivation. PLoS Pathog. 2007; 3: 80.
  34. Magrath I. Epidemiology: clues to the pathogenesis of Burkitt lymphoma. Br. J. Haematol. 2012; 156(6): 744–56.
  35. Morrow R.H., Gutensohn N., Smith P.G. Epstein-Barr virus-malaria interaction models for Burkitt’s lymphoma: implications for preventive trials. Cancer Res. 1976; 36: 667–9.
  36. Amusa Y.B., Adediran I.A., Akinpelu V.O. et al. Burkitt’s lymphoma of the head and neck region in a Nigeria tertiary hospital. East. Afr. J. Med. 2005; 24(3): 139–42.
  37. Blum K.A., Lozansky G., Byrd J. Adult Burkitt’s leukemia and lymphoma. Blood 2004; 20: 32–7.
  38. Warnke R., Weiss L., Chan J. et al. Atlas of Tumor Pathologie. Tumors of the Lymph Nodes and Spleen. Washington: Armed Forces Institute of Pathology, 1999: 221–32.
  39. Kittivorapart J., Chinthammitr Y. Incidence and risk factors of bone marrow involvement by non-Hodgkin lymphoma. J. Med. Assoc. Thai. 2011; 94(Suppl. 1): S239–45.
  40. Ziegler J.L., Morrow R.H., Fass L. et al. Treatment of Burkitt’s lymphoma tumor with cyclophosphamide. Cancer 1970; 26(2): 474–84.
  41. Janota I. Involvement of the nervous system in malignant lymphoma in Nigeria. Br. J. Cancer 1966; 20: 47.
  42. Booth K., Burkitt D.P., Basset D.J. et al. Burkitt lymphoma in Papua, New Guinea. Br. J. Cancer 1967; 21: 657–64.
  43. Sariban E., Edwards S., Janus C. et al. Central nervous system involvement in American Burkitt’s lymphoma. J. Clin. Oncol. 1983; 1: 677–80.
  44. Wright D.H. Burkitt’s tumor. A post modern study of 50 cases. Br. J. Surg. 1964; 51: 245.
  45. Thomas D., Cortes J., O’Brien S. et al. Hyper-CVAD program in Burkitt’s type adult acute lymphoblastic leukemia. J. Clin. Oncol. 1999; 17: 2461–70.
  46. Murphy S., Fairclough D., Hutchison R. et al. Non-Hodgkin’s lymphoma in childhood: An analysis of the histology, staging and response to the treatment of 338 cases at a single institution. J. Clin. Oncol. 1989; 7: 186–93.
  47. Klumb C.E., Resende L.M., Stefanoff C.G. et al. Burkitt-like lymphoma in an infant: a case report. Rev. Hosp. Clin. Fac. Med. S. Paulo. 2003; 58(1): 33–6.
  48. Hutchinson R.E., Murphy S.B., Fairclough D.L. et al. Diffuse small noncleaved cell lymphoma in children, Burritt’s versus non-Burritt’s types. Results from the Реdiatric Oncology Group and St. Jude Children’s Research Hospital. Cancer 1989; 64: 23–8.
  49. Ferry J.A. Burkitt’s Lymphoma: Clinicopathologic Features and Differential Diagnosis. Oncologist 2006; 11: 375–83.
  50. Shad A., Magrath I. Non-Hodgkin’s lymphoma. Pediatr. Clin. N. Am. 1997; 44: 863–90.
  51. Boerma E.G., van Imhoff G.W., Appel I.M. et al. Gender and age-related differences in Burkitt lymphoma—epidemiological and clinical data from The Netherlands. Eur. J. Cancer 2004; 40: 2781–7.
  52. Huisman T., Tschirch F., Schneider J.F. et al. Burkitt’s lymphoma with bilateral cavernous sinus and mediastinal involvement in a child. Pediatr. Radiol. 2003; 33: 719–21.
  53. Braziel R.M., Arber D.A., Slovac M.L. et al. The Burkitt-like lymphoma: a Southwest Oncologie Group study delineating phenotypic, genotypic and clinical features. Blood 2001; 97(12): 3713–20.
  54. Cheung C.W., Burton C., Smith P. Central nervous system chemoprophylaxis in non-Hodgkin’s lymphoma: current practice in the UK. Br. J. Haematol. 2005; 13(2): 193–200.
  55. McWilliams N., Hatfield W., Jackson R. Epidemiological notes and reports on Burkitt’s lymphoma. Winchester, Virginia. Morbil. Mortal. Wkly. 1997; 46: 4674–8.
  56. Барях Е.А., Кравченко С.К., Обухова Т.Н. и др. Лимфома Беркитта: клиника, диагностика, лечение. Клин. онкогематол. 2009; 2(2): 137–47. [Baryakh Ye.A., Kravchenko S.K., Obukhova T.N et al. Burkitt’s lymphoma: clinical presentation, diagnosis, management. Klin. onkogematol. 2009; 2(2): 137–47. (In Russ.)].
  57. Malani A.K., Gupta C., Weigand R.T. et al. Spinal Burkitt’s lymphoma in adults. Clin. Lymph. Myel. 2006; 6(4): 333–6.
  58. Mizugami T., Mikata A., Hajikano H. et al. Primary spinal epidural Burkitt’s lymphoma. Surg. Neurol. 1987; 28(2): 158–62.
  59. Ses E., N’dri Oka D., Varlet G. et al. Medullary compression by Burkitt lymphoma. Analysis of 7 cases. Neurochirurgie 2001; 47(6): 552–6.
  60. Wilkening A., Brack M., Brandis A. et al. Unusual presentation of a primary spinal Burkitt’s lymphoma. J. Neurol. Neurochirurg. Psychiatry 2001; 70(6): 794–7.
  61. Holland J., Cada M., Ling S. et al. Melena: a rare presentation of childhood Burkitt’s lymphoma. CMAJ 2005; 173(3): 247–8.
  62. Saton S., Saito T., Akiba J. et al. Burkitt lymphoma occurring as a primary lymphomatous effusion. Rinsho Ketsueki 2000; 41(4): 329–33.
  63. Blanc S., Bertrand Y., Lorthois-Ninou S. et al. Burkitt’s lymphoma revealed by a rectal tumor. Arch. Pediatr. 2002; 9(10): 1056–8.
  64. Rakoto-Ratsimba H.N., Razafimahandry H.J.C., Samison L.H. et al. A case of anal Burkitt’s lymphoma. Ann. Chir. 2003; 128(4): 265–7.
  65. Meshref M., Sassolas F., Schell M. et al. Primary cardiac Burkitt lymphoma in a child. Pediatr. Blood Cancer 2004; 42(4): 380–3.
  66. Baloglu H., Turken O., Turuncu L., Kizilkaya E. 24-year-old female with amenorhea: bilateral primary ovarian Burkitt lymphoma. Gynecol. Oncol. 2003; 91(2): 449–51.
  67. Grassi M., Lee A.G. Lymphomatous meningitis of the Burkitt type presenting with multiple cranial neuropathies. Am. J. Ophthalmol. 2002; 133(3): 424–5.
  68. Ardekian L., Rachmiel A., Rosen D. et al. Burkitt’s lymphoma of the oral cavity in Israel. J. Craniomaxillofac. Surg. 1999; 27(5): 294–7.
  69. Барях Е.А., Красильникова Б.Б. Поражение лицевого скелета при спорадическом варианте лимфомы Беркитта. В кн.: Редкие гематологические болезни и синдромы. Под ред. М.А. Волковой. М.: Практическая медицина, 2011: 311–9. [Baryakh Ye.A., Krasilnikova B.B. Porazheniye litsevogo skeleta pri sporadicheskom variante limfomy Berkitta. V kn.: Redkiye gematologicheskiye bolezni i sindromy. Pod red. M.A. Volkovoy (Involvement of facial skeleton in sporadic variant of Burkitt’s lymphoma. In: Rare hematological disorders and syndromes. Ed. by: M.A. Volkova). M.: Prakticheskaya meditsina, 2011: 311–9.]
  70. Banthia V., Jen A., Kacker A. Sporadic Burkitt’s lymphoma of the head and neck in the pediatric population. Int. J. Pediatr. Otorhinolaryngol. 2003; 67(1): 59–65.
  71. Bauer G.P., Volk M.S., Siddiqui S. Burkitt’s lymphoma of the parapharingeal space. Arch. Otolaryngol. Head Neck Surg. 1993; 119(1): 117–20.
  72. Барях Е.А., Валиев Т.Т., Звонков Е.Е. и др. Интенсивная терапия лимфомы Беркитта: описание двух клинических случаев. Гематол. и транс- фузиол. 2007; 1: 41–3. [Baryakh Ye.A., Valiyev T.T., Zvonkov Ye.Ye. et al. Intensive therapy for Burkitt’s lymphoma: presentation of two clinical cases. Gematol. i transfuziol. 2007; 1: 41–3. (In Russ.)].
  73. Shukla N., Trippett T. Non-Hodgkin’s lymphoma in children and adolescents. Curr. Oncol. Rep. 2006; 8(5): 387–94.
  74. Evans J.A., Gribb D.M., Holland F.J. et al. Malignancies in UK in children with HIV infection acquired from mother to child transmission. Arch. Dis. Child. 1997; 76: 330–3.
  75. Subar M., Neri A., Inghirami G. et al. Frequent c-myc oncogene activation and infrequent presence of Epstein-Barr virus genome in AIDS-associated lymphoma. Blood 1988; 72: 667–71.
  76. Beral V., Petrman T., Berkelman R. et al. AIDS-associated non-Hodgkin’s lymphoma. Lancet 1991; 337: 805–9.
  77. Carbone A., Gloghini A., Gaidano G. et al. AIDS-related Burkitt’s lymphoma. Morphologic and immunophenotypic study of biopsy specimens. Am. J. Clin. Pathol. 1995; 103: 561–7.
  78. Kasamon Y.L., Swinnen L.J. Treatment advances in adult Burkitt lymphoma and leukemia. Curr. Opin. Oncol. 2004; 16: 429–35.
  79. Knowles D.M. Etiology and pethogenesis of AIDS-related non-Hodgkin’s lymphoma. Hematol. Oncol. Clin. N. Am. 2003; 17: 785–820.
  80. Martinez-Maza O., Breen E.C. B-cell activation and lymphoma in patients with HIV. Curr. Opin. Oncol. 2002; 14: 528–32.
  81. Mbulaiteye S.M., Clarke C.A., Morton L.M. Burkitt lymphoma risk in U.S. solid organ transplant recipients. Am. J. Hematol. 2013; 88: 245.
  82. Gong J.Z., Stenzel T.T., Bennet E.R. et al. Burkitt’s lymphoma arising in organ transplant recipients: a clinicopathologic study of five cases. Am. J. Surg. Pathol. 2003; 27(6): 818–27.
  83. Davi F., Delecluse H.J., Guiet P. et al. Burkitt-like lymphomas in AIDS patients: characterisation within a series of 103 human immunodeficiency virusassociated non-Hodgkin’s lymphomas. Burkitt’s Lymphoma Study Group. J. Clin. Oncol. 1998; 16: 3788–95.
  84. Барях Е.А., Кравченко С.К., Кременецкая А.М. и др. Лейкоз/лимфома Беркитта: клинические особенности, диагностические критерии, терапевтическая тактика. Клин. онкогематол. 2010; 3(2): 138–43. [Baryakh Ye.A., Kravchenko S.K., Kremenetskaya A.M. et al. Burkitt’s lymphoma: clinical features, diagnostic criteria, therapeutic approach. Klin. onkogematol. 2010; 3(2): 138–43. (In Russ.)].
  85. Валиев Т.Т., Морозова О.В., Попа А.В. и др. Результаты лечения лимфомы Беркитта у детей. Гематол. и трансфузиол. 2012: 57(3): 34–5. [Valiyev T.T., Morozova O.V., Popa A.V. et al. Therapeutic outcomes in childhood Burkitt’s lymphoma. Gematol. i transfuziol. 2012: 57(3): 34–5. (In Russ.)].
  86. Ковригина А.М., Пробатова Н.А. Лимфома Ходжкина и крупноклеточные лимфомы. М.: МИА, 2007: 212. [Kovrigina A.M., Probatova N.A. Limfoma Khodzhkina i krupnokletochnye limfomy (Hodgkin’s lymphoma and large-cell lymphomas). M.: MIA, 2007: 212.]
  87. Тупицын Н.Н., Шолохова Е.Н., Андреева Л.Ю. и др. Иммунодиагностика лимфом. Совр. онкол. Экстравыпуск 2002: 4–12. [Tupitsyn N.N., Sholokhova Ye.N., Andreyeva L.Yu. et al. Immunodiagnosis of lymphomas. Sovr. onkol. Ekstravypusk 2002: 4–12. (In Russ.)].
  88. Луговская С.А., Почтарь М.Е., Тупицын Н.Н. Иммунофенотипиро- вание в диагностике гемобластозов. М., 2005. [Lugovskaya S.A., Pochtar M.Ye., Tupitsyn N.N. Immunofenotipirovaniye v diagnostike gemoblastozov (Immunophenotyping in diagnosis of hematological malignancies). M., 2005.]
  89. Zech L., Haglund U., Nilsson. et al. Characteristic chromosomal abnormalities in biopsies and lymphoid-cell lines from patients with Butkitt and non-Burkitt lymphomas. Int. J. Cancer 1976; 17: 47–56.
  90. Croce C. Role of chromosome translocations in human neoplasia. Cell 1987; 49(2): 155–6.
  91. Showe L.C., Moore R.C., Erikson J. et al. MYC oncogene involved in a t(8;22) chromosome translocation is not altered in it’s putative regulatory regions. Proc. Natl. Acad. Sci. U S A 1987; 84(9): 2824–8.
  92. Tagawa H., Ikeda S., Sawada K. Role of microRNA in the pathogenesis of malignant lymphoma. Cancer Sci. 2013; 10: 121–6.
  93. Sander S., Calado D.P., Srinivasan L. et al. Synergy between PI3K signaling and MYC in Burkitt lymphomagenesis. Cancer Cell 2012; 22(2): 167–79.
  94. Love C., Sun Z., Jima D. The genetic landscape of mutations in Burkitt lymphoma. Nat. Genet. 2012; 44(12): 1321–5.
  95. Green T.M., Nielsen O., de Stricker K. et al. High levels of nuclear MYC protein predict the presence of MYC rearrangement in diffuse large B-cell lymphoma. Am. J. Surg. Pathol. 2012; 36(4): 612–9.
  96. Aukema S.M., Siebert R., Schuuring E. et al. Double-hit B-cell lymphomas. Blood 2011; 117(8): 2319–31.
  97. Ben-Neriah S., Woods R., Steidl C. et al. Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival. Blood 2009; 114: 2273–9.
  98. Johnson N.A., Savage K.J., Ludkovski O. et al. Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival. Blood 2009; 114: 2273–9.
  99. Aukema S.M., Siebert R., Schuuring E. et al. Double-hit B-cell lymphomas. Blood 2011; 117: 2319–31.
  100. Niitsu N., Okamoto M., Miura I. et al. Clinical features and prognosis of de novo diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC translocations. Leukemia 2009; 23: 777–83.
  101. Salaverria I., Siebert R. The gray zone between Burkitt’s lymphoma and diffuse large B-cell lymphoma from a genetics perspective. J. Clin. Oncol. 2011; 29: 1835–43.
  102. Snuderl M., Kolman O., Chen Y. et al. B-cell Lymphomas with Concurrent IGH-BCL2 and MYC Rearrangements Are Aggressive Neoplasms with Clinical and Pathologic Features Distinct from Burkitt Lymphoma and Diffuse Large B-cell Lymphoma. Am. J. Surg. Pathol. 2010; 34(3): 327–40.
  103. Tomita N. BCL2 and MYC Dual-Hit Lymphoma/Leukemia. J. Clin. Exp. Hematopathol. 2011; 51(1): 7–12.
  104. Pillai R.K., Sathanoori M., Van Oss S.B. Double-hit B-cell Lymphomas With BCL6 and MYC Translocations Are Aggressive, Frequently Extranodal Lymphomas Distinct From BCL2 Double-hit B-cell Lymphomas. Am. J. Surg. Pathol. 2013; 37(3): 323–32.
  105. Nakayama S., Yokote T., Iwaki K. Triple-hit B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma associated with a novel complex karyotype including t(2;3)(q21;q27), t(8;14)(q24;q32) and t(14;18)(q32;q21). Br. J. Haematol. 2013; 160(5): 569.
  106. Deibold J. Burkitt lymphoma. In: Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues. Ed. by E. Jaffe, N. Harris et al. Washington: IARC Press, 2001: 181–4.
  107. Baccarani M., Corbelli G., Amadori S. et al. Adolescent and adult lymphoblastic leukemia: prognostic features outcome of therapy — a study of 293 patients. Blood 1982; 60: 677–84.
  108. Gill P.S., Meyer P.R., Pavlova Z. et al. B-cell acute lymphoblastic leukemia in adults: clinical, morphologic and immunologic findings. J. Clin. Oncol. 1986; 4: 737–43.
  109. Bernstein J.I., Coleman C.N., Strickler J.G. et al. Combined modality therapy for adult with small noncleaved cell lymphoma (Burkitt and Burkitt-like type). J. Clin. Oncol. 1986; 4: 847–58.
  110. Reiter A., Schrappe M., Tiemann M. et al. Improved treatment results in childhood B-cell neoplasms with tailored intensification of therapy: a report of the Berlin-Frankfurt-Munster Group Trial NHL-BFM-90. Blood 1999; 94(10): 3294–306.
  111. Schwenn M., Blattner S., Lynch E. et al. HiC-COM: a 2-month intensive chemotherapy regimen for children with stage III and IV Burkitt’s lymphoma and B-cell acute lymphoblastic leukemia. J. Clin. Oncol. 1991; 9: 133–8.
  112. Bowman W.P., Shuster J.J., Cook B. et al. Improved survival for children with B-cell acute lymphoblastic leukemia and stage IV small noncleaved-cell lymphoma: a pediatric oncology group study. J. Clin. Oncol. 1996; 14(4): 1252–61.
  113. Patte C., Philip T., Rodary C. et al. High survival rate in advanced-stage B-cell lymphomas and leukemias without CNS involvement with a short intensive polychemotherapy: results from the French Pediatric Oncology Society of a randomized trial of 216 children. J. Clin. Oncol. 1991; 9: 123–32.
  114. Patte C., Michon J., Frappaz D. et al. Therapy of Burkitt and other Bcell acute lymphoblastic leukaemia and lymphoma: experience with the LMB protocols of the SFOP (French Paediatric Oncology Society) in children and adults. Baillieres Clin. Haematol. 1994; 7(2): 339–48.
  115. Magrath I., Adde M., Shad A. et al. Adults and children with small noncleaved-cell lymphoma have similar excellent outcome when treated with the same chemotherapy regimen. J. Clin. Oncol. 1996; 14: 925–34.
  116. Mead G.M., Sydes M.R., Walewski J. et al. An international evaluation of CODOX-M and CODOX-M alternating with IVAC in adult Burkitt’s lymphoma: results of United Kingdom Lymphoma Group LY06 study. Ann. Oncol. 2002; 13(8): 1264–74.
  117. Барях Е.А., Кравченко С.К. Протокол терапии лимфомы Беркитта взрослых по программе ЛБ-М-04. В кн. Программное лечение заболеваний системы крови. Т. II. Под ред. В.Г. Савченко. М.: Практика, 2012: 720–34. [Baryakh Ye.A., Kravchenko S.K. Protokol terapii limfomy Berkitta vzroslykh po programme LB-M-04. V kn. Programmnoye lecheniye zabolevaniy sistemy krovi. T. II. Pod red. V.G. Savchenko (Protocol of therapy for adult Burkitt’s lymphoma in accordance with LB-M-04 program. In: Program therapy for hematological malignancies. Vol. II. Ed. by: V.G. Savchenko). M.: Praktika, 2012: 720–34.]
  118. Thomas D.A., Faderl S., O’Brien S. et al. Chemoimmunotherapy with hyper-CVAD plus rituximab for the treatment of adult Burkitt and Burkitt-type lymphoma or acute lymphoblastic leukemia. Cancer 2006; 106(7): 1569–80.
  119. Fayad L., Thomas D., Romaguera J. Update of the M.D. Anderson Cancer Center experience with hyper-CVAD and rituximab for the treatment of mantle cell and Burkitt-type lymphomas. Clin. Lymph. Myel. 2007; 8(2): 57–62.
  120. Bence Z., Kovacs G., Jakab Z. et al. Lymphomas in adolescents: are childhood lymphoma therapy protocols suitable for this patient group? Magy Onkol. 2008; 52(4): 357–62.
  121. Cairo M.S., Sposto R., Gerrard M. et al. Advanced Stage, Increased Lactate Dehydrogenase, and Primary Site, but Not Adolescent Age (³ 15 Years), Are Associated With an Increased Risk of Treatment Failure in Children and Adolescents With Mature B-Cell Non-Hodgkin’s Lymphoma: Results of the FAB LMB 96 Study. J. Clin. Oncol. 2012; 30(4): 387–93.
  122. Woessmann W., Reiter A. Re-induction approaches to relapsed/ refractory childhood and adolescent non Hodgkin’s lymphoma: BFM perspective. Br. J. Haematol. 2012; 159(1): 41.
  123. Griffin T.C., Weitzman S., Weinstein H. et al. A study of rituximab and ifosfamide, carboplatin, and etoposide chemotherapy in children with recurrent/ refractory B-cell (CD20+) non-Hodgkin lymphoma and mature B-cell acute lymphoblastic leukemia: A report from the Childrens Oncology Group. Pediatr. Blood Cancer 2009; 53: 177–81.
  124. Harris N., Jaffe E., Stein H. et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood 1994; 84: 1361–92.

Factors determining in vitro survival of human multiple myeloma cells

S.S. Shushanov1, T.A. Kravtsova1, and Yu.B. Cherhykh2

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

2 M.F. Vladimirskiy Moscow Regional Research-and-Clinical Institute, Moscow, Russian Federation


ABSTRACT

In this work, we investigated the survival of the human MM cell lines, i.e. RPMI1640, RPMI8226, and IM9, depending on the degree of their differentiation and impact of exogenous IGF-1. The study showed that the most and least sensitive to growth factors are IM9 cells with CD138+, CD38–, CD45+, CD56–, or CD19+ immunophenotype and RPMI1640 and RPMI8226 cells with (CD138+, CD38+, CD45–, CD56+/–, or CD19– immunophenotype, respectively The gene expression studies revealed that the level of IGF-1R and IRA mRNA expression was significantly lower in the IM9 cells than in the RPMI1640 or RPMI8226 cells. Also, we established that exogenous IGF-1 could variously influence the survival and growth of MM cells. IGF-1 alone exerts no effect on the myeloma cell viability, but, in combination with serum growth factors, it increases the parameter.


Keywords: multiple myeloma (MM), insulin-like growth factor 1 (IGF-1), cell survival, mRNA expression.

Read in PDF (RUS)pdficon


REFERENCES

  1. Вотякова О.М., Демина Е.А. Множественная миелома. В кн.: Клиниче- ская онкогематология. Руководство для врачей, 2-е изд., перераб и доп. Под ред. М.А. Волковой. М.: Медицина, 2007: 847–73. [Votyakova O.M., Demina Ye.A. Mnozhestvennaya miyeloma. V kn.: Klinicheskaya onkogematologiya. Rukovodstvo dlya vrachey, 2-e izd., pererab i dop. Pod red. M.A. Volkovoy. (Multiple myeloma. In: Clinical oncohematology. Manual for medical practitioners. 2nd ed., rev.&upd. Ed by: M.A. Volkova). M.: Meditsina, 2007: 847–73.]
  2. Jaffe E.E., Harris N., Stein H. et al. World Health Organization Classification of Tumors. Pathology and Genetics of Tumors of Hematopoietic and Lymphoid Tissues. Lyon: IARC Press, 2001: 351.
  3. Raab M.S, Podar K., Breitkreutz I. et al. Multiple myeloma. Lancet 2009; 374: 324–39.
  4. Dispenzieri A., Kyle R.A. Multiple myeloma: clinical features and indications for therapy. Best Pract. Res. Clin. Haematol. 2005; 18: 553–68.
  5. Hideshima T., Bergsagel P.L., Kuehl W.M. et al. Advances in Biology of Multiple Myeloma: Clinical Applications. Blood 2004; 104: 607–18.
  6. Sprynski A.C., Hose D., Caillot L. et al. The role of IGF-1 as a major growth factor for myeloma cell lines and the prognostic relevance of the expression of its receptor. Blood 2009; 113: 4614–26.
  7. Mitsiades C.S., Mitsiades N., Kung A.L. et al. The IGF/IGF-1R system is a major therapeutic target for multiple myeloma, other hematologic malignancies and solid tumors. Blood 2002; 100(11): Abstract 637.
  8. Samani A.A., Yakar S., LeRoith D. et al. The role of the IGF system in cancer growth and metastasis: Overview and recent insights. Endocr. Rev. 2007; 28: 20–47.
  9. Hernandez-Sanchez С., Werner H., Roberts C.T. et al. Differential Regulation of Insulin-like Growth Factor-I (IGF-I) Receptor Gene Expression by IGF-I and Basic Fibroblastic Growth Factor: JBC 1997; 272(8): 4663–70.
  10. Kalitin N., Kostjukova M., Kakpakova E. et al. Vascular endothelial growth factor 1 (VEGFR1) gene expression depends on immunophenotype of human multiple myeloma сells. Eur. J. Cancer 2011; 47(1): S644.
  11. Шушанов С.С., Кравцова Т.А. Цитотоксическое действие доксору- бицина in vitro на клетки множественной миеломы человека. Бюл. экспер. биол. и мед. 2013; 155(2): 195–200. [Shushanov S.S., Kravtsova T.A. Doxorubicine cytotoxic in vitro effect on cells of human multiple myeloma. Byul. eksper. biol. i med. 2013; 155(2): 195–200. (In Russ.)].
  12. Шушанов C.C. Роль инсулиноподобного фактора роста 1 типа (IGF-1) и некоторых других членов системы IGF/инсулин в прогрессии множественной миеломы. Рос. биотер. журн. 2012; 11(3):71–80. [Shushanov C.C. Role of insulin-like growth factor 1 and some other members of IGF/insulin system in progression of multiple myeloma. Ros. bioter. zhurn. 2012; 11(3):71–80. (In Russ.)].
  13. Ludwig T., EggenschwillerJ., Fisher P. et al. Mouse mutants lacking the type 2 IGF receptor (IGF2R) are rescued from perinatal lethality in igf2 and igf-1r null backgrounds. Dev. Biol. 1996; 177: 517–35.
  14. Sacco A., Morcavallo A., Pandini G. et al. Differential signaling activation by insulin and insulin-like growth factors I and II upon binding to insulin receptor isoform A. Endocrinology 2009; 150(8): 3594–602.
  15. Moxham C.P., Duronio V., Jacobs S. et al. Insulin-like growth factor 1 receptor beta–subunit geterogeneity. Evidence for hybrid tetramers composed of insulin-like growth factor 1 and insulin receptor heterodimers. J. Biol. Chem. 1989; 264: 13238–44.
  16. Frasca F., Pandini G., Scalia P. et al. Insulin receptor isoform A a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol. Cell Biol. 1999; 19: 3278–88.
  17. Yamaguchi Y., Flier J.S., Yokoto A. et al. Functional properties of two naturally occurring isoforms of the human insulin receptor in Chinese hamster ovary cells. Endocrinology 1991; 129: 2058–66.
  18. Yu H., Rohan T. Role of insulin-like growth factor family in cancer development and Progression. J. Natl. Cancer Inst. 2000; 92: 1472–89.
  19. Weber J.D., Kuo M.L., Bothner B. et al. Cooperative signals governing ARF mdm2 interaction and nucleolar localization of the complex. Mol. Cell Biol. 2000; 20: 2517–28.
  20. Barton E.R. The ABCs of IGF-1 isoforms: impact of muscle hypertrophy and implications for repair. Appl. Physiol. Nutr. Metab. 2006; 31: 791–7.
  21. Parker A., Cheville J.C., Lohse C. et al. Expression of insulin-like growth factor I receptor and survival in patients with clear cell renal cell carcinoma. J. Urol. 2003; 170: 420–4.
  22. Parker A.S., Cheville J.C., Janney C.A. High expression levels of insulinlike growth factor-I receptor predict poor survival among women with clear-cell renal cell carcinomas. Hum. Pathol. 2002; 33(8): 801–5.
  23. Gicquel C., Bertagna X., Gaston V. et al. Molecular Markers and LongTerm Recurrences in a Large Cohort of Patients with Sporadic Adrenocortical Tumors. Cancer Res. 2001; 61: 6762–67.
  24. Sayer R.A., Lancaster J.M., Pittman J. et al. High insulin-like growth factor-2 (IGF-2) gene expression is an independent predictor of poor survival for patients with advanced stage serous epithelial ovarian cancer. Gynecol. Oncol. 2005; 96(2): 355–61.
  25. Shariat S.F., Kim J., Nguyen C. et al. Correlation of preoperative levels of IGF-I and IGFBP-3 with pathologic parameters and clinical outcome in patients with bladder cancer. Urology 2003; 61(2): 359–64.
  26. Hernandez-Sanchez S., Werner H. et al. Differential regulation of insulinlike growth factor-1 (IGF-1) receptor gene expression by IGF-1 and basic fibroblastic growth factor. Biol. Chem. 1997; 272(8): 4663–70.
  27. Rubini M., Werner H. Gandini E. et al. Platelet-derived growth factor increases the activity of the promotor of the insulin-like growth factor-1 (IGF-1) receptor gene. Exp. Cell Res. 1994; 211(2): 374–9.
  28. Van Riet I., Vande Broek I., Asosingh K. et al. Endothelial cell-tumor cell interactions in multiple myeloma. Hematol. J. 2003; 4(Suppl. 1): P6.3 (abstract).
  29. Vanderkerken K., De Greef C., Asosingh K. et al. Selective initial in vivo homing pattern of 5T2 multiple myeloma cells in the C57BL/KalwRij mouse. Br. J. Cancer 2000; 82: 953–9.
  30. Coppola D., Ferber A., Miura M. et al. A functional insulin-like growth factor 1 receptor is required for the mitogenic and transforming activities of the epidermal growth factor receptor. Mol. Cell Biol. 1994; 14(7): 4588–99.
  31. DeAngelis T., Ferber A., Baserga R. A functional insulin-like growth factor 1 receptor is required for the mitogenic and transforming activities of the platelet derived growth factor receptor. J. Cell Physiol. 1995; 164(1): 214–21.
  32. Ferlin M., Noraz N., Hertogh C. Insulin-like growth factor induces the survival and proliferation of myeloma cells through an interleukin-6-independent transduction pathway. Br. J. Haematol. 2000; 111: 626–4.
  33. Georgii-Hemming P., Wiklund H.J., Ljunggren O. et al. Insulin-like growth factor I is a growth and survival factor in human multiple myeloma cell lines. Blood 1996; 88: 2250–8.
  34. Qiang Y.W., Kopantzev E., Rudikoff S. et al. Insulin like growth factor-I signaling in multiple myeloma: downstream elements, functional correlates, and pathway cross-talk. Blood 2002; 99: 4138–46.

In vitro model of myelofibrosis using human platelet lysate

Ye.N. Bulycheva, N.T. Siordiya, E.G. Lomaia, A.Yu. Zaritskiy, and P.A. Butylin

V.A. Almazov Federal Heart, Blood and Endocrinology Center, Saint Petersburg, Russian Federation


ABSTRACT

The development and studies of the myelofibrosis (MF) in vitro model is an important issue, since such model can lead to understanding of pathogenesis and identifying the new targets for therapy.

Objectives. Here, we studied the properties of mesenchymal stromal cells (MSCs) cultured in the medium containing the human platelet lysate (HPL).

Design and methods. Bone marrow MSCs from healthy donors and a patient with primary myelofibrosis (PMF) were cultured in the media containing various HPL concentrations. We measured the proliferative activity, the collagen type I and III expression, and capability to differentiate into the osteogenic or adipogenic lineages. The concentrations of the vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF-b), and hepatocyte growth factor (HGF) were measured in HPL from 17 patients with primary myelofibrosis or post-polycythemia vera myelofibrosis (post-PV MF) using the specific ELISA kits.

Results. The highest MSC proliferative rate was found in the cultures with high HPL concentrations (10–20 %). The ratio of collagen type III/collagen type I expression was the highest in the cultures containing 10 % HPL. The use of HPL for MSCs culturing caused no changes in their osteogenic differentiation capability, but the increase in the HPL concentration resulted in the decreased capability to differentiate into the adipogenic lineage. Further, we observed the significantly increased VEGF and bFGF concentrations in HPL from MF patients, compared to the age-matched healthy controls (2.5- and 2.4-fold, respectively, < 0.01), while the TGF-b and HGF concentrations showed the trend towards an increase, but with no significant difference from the controls. MSCs cultured with HPL from MF patients showed a higher proliferative rate compared to HPL from healthy donors. MSCs from the PMF patient tended to proliferate more actively compared to the cells from healthy donors.

Conclusion. MSCs culturing using varying HPL concentrations can be used as an adequate MF in vitro model, since it leads to pro-fibrotic changes in the bone marrow stromal cells.


Keywords: primary myelofibrosis, platelet lysate, mesenchymal stromal cells, myelofibrosis in vitro model.

Read in  PDF (RUS)pdficon


Refernces

  1. Kutti J., Ridell B. Epidemiology of the myeloproliferative disorders: essential thrombocythaemia, polycythaemia vera and idiopathic myelofibrosis. Biol. 2001; 49: 164–6.
  2. Johansson P., Kutti J., Andreasson B. et al. Trends in the incidence of chronic Philadelphia chromosome negative (Ph-) myeloproliferative disorders in the city of Goteborg, Sweden, during 1983–99. Inter. Med. 2004; 256: 161–5.
  3. Cervantes F., Dupriez B., Passamonti F. et al. Improving survival trends in primary myelofibrosis: an international study. Clin. Oncol. 2012; 30: 2981–7.
  4. Barosi G., Ambrosetti A., Finelli C. et al. The Italian Consensus Conferen ce on Diagnostic Criteria for Myelofibrosis with Myeloid Metaplasia. J. Haematol. 1999; 104(4): 730–7.
  5. Колосков А.В. Мегакариоциты и фиброз костного мозга. Гематол. и трансфузиол. 1997; 42: 29–31. [Koloskov A.V. Megakariotsity i fibroz kostnogo mozga (Megakaryocytes and bone marrow fibrosis. In: Hematol. & transfuziol.). Gematol. i transfuziol. 1997; 42: 29–31.]
  6. Reilly J.T. Idiopathic myelofibrosis: pathogenesis to treatment. Oncol. 2006; 24: 56–63.
  7. Le Bousse-Kerdiles M.-C., Martyre M.-C., Samson M. Cellular and molecular mechanisms underlying bone marrow and liver fibrosis: a review. Cytokine Netw. 2008; 19: 69–80.
  8. Yan X.Q., Lacey D., Fletcher F. et al. Chronic exposure to retroviral encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood 1995; 86; 4025–33.
  9. Vannucchi A.M., Bianchi L., Paoleti F. et al. Impaired GATA-1 expression and myelofibrosis in an animal model. Biol. 2004; 52: 275–9.
  10. Xing S., Wanting T.H., Zhao W. et al. Transgenic expression of Jak2V617F causes myeloproliferative disorders in mice. Blood 2008; 111: 5109–17.
  11. Wernig G., Mercher T., Okabe R. et al. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood 2006; 107: 4274–81.
  12. Kirabo A., Park S.O., Wamsley H.L. et al. The small molecule inhibitor G6 significantly reduces bone marrow fibrosis and the mutant burden in a mouse model of Jak2-mediated myelofibrosis. AJP 2012; 181: 858–65.
  13. Triviai I., Stubing Th., Niebuhr B. et al. A mouse model for human myelofibrosis. 18th Congress of EHA 2013: Abstract P248.
  14. Xia W., Li H., Wang Zh. et al. Human platelet lysate supports ex vivo expansion and enhances osteogenic differentiation of human bone
  15. Sankaranarayanan K., Chandana T., Gency Ponrose G. et al. Humanized substitutes for animal sera in human mesenchymal stem cell culture and differentiation. Cell Biol. Intern. 2011; Manuscript CBI20100649.
  16. Jenhani F., Durand V., Ben Azouna N. et al. Human cytokine expression profile in various conditioned media for in vitro expansion bone marrow and umbilical cord blood immunophenotyped mesenchymal stem cells. Proceed. 2011; 43: 639–43.
  17. Doucet Ch., Ernou I., Zhang Y. et al. Platelet lysates promote mesenchymal stem cell expansion: a safety substitute for animal serum in cell-based therapy applications. Cell. Physiol. 2005; 205: 228–36.
  18. Horn P., Bokermann G., Cholewa D. et al. Impact of individual platelet lysates on isolation and growth of human mesenchymal stromal cells. Cytotherapy 2010; 12: 888–98.
  19. Schmitt A., Jouault H., Guichard J. et al. Pathologic interaction between megacaryocytes and polymorphonuclear leukocytes in myelofibrosis. Blood 2000; 96: 1342–7.