Плазмобластная лимфома у пациентов с ВИЧ-инфекцией: обзор литературы и результаты российского многоцентрового ретроспективного исследования

М.О. Попова1, И.В. Цыганков1, Я.В. Гудожникова1, Ю.А. Рогачева1, Н.П. Волков1, К.В. Лепик1, М.В. Демченкова2, М.В. Григорьева2, А.Ю. Ефиркина2, Т.В. Шнейдер3, Ю.В. Копейкина3, С.А. Степанова3, В.Г. Потапенко4, А.В. Климович4, Н.В. Медведева4, М.А. Колесникова5, Т.И. Поспелова5, Н.Б. Михайлова1, В.В. Байков1, А.Д. Кулагин1

1 НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой, ФГБОУ ВО «Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова» Минздрава России, ул. Льва Толстого, д. 6/8, Санкт-Петербург, Российская Федерация, 197022

2 ГБУЗ «Областной онкологический диспансер», ул. Фрунзе, д. 32, Иркутск, Российская Федерация, 664035

3 ГБУЗ «Ленинградская областная клиническая больница», пр-т Луначарского, д. 45, корп. 2А, Санкт-Петербург, Российская Федерация, 194291

4 ГБУЗ «Городская клиническая больница № 31», пр-т Динамо, д. 3, Санкт-Петербург, Российская Федерация, 197110

5 Городской гематологический центр, ул. Ползунова, д. 21, Новосибирск, Российская Федерация, 630051

Для переписки: Марина Олеговна Попова, канд. мед. наук, ул. Льва Толстого, д. 6/8, Санкт-Петербург, Российская Федерация, 197022; тел.: +7(911)711-39-77; e-mail: marina.popova.spb@gmail.com

Для цитирования: Попова М.О., Цыганков И.В., Гудожникова Я.В. и др. Плазмобластная лимфома у пациентов с ВИЧ-инфекцией: обзор литературы и результаты российского многоцентрового ретроспективного исследования. Клиническая онкогематология. 2022;15(1):28–41.

DOI: 10.21320/2500-2139-2022-15-1-28-41


РЕФЕРАТ

Актуальность. Плазмобластная лимфома (ПБЛ) относится к редким вариантам лимфопролиферативных заболеваний и встречается почти исключительно на фоне иммунодефицитных состояний. Наибольший опыт химиотерапии и трансплантации гемопоэтических стволовых клеток (ТГСК) при данном варианте лимфомы накоплен именно у пациентов с ВИЧ-инфекцией.

Цель. Представить современные подходы к диагностике и терапии ПБЛ у пациентов с ВИЧ-инфекцией, а также результаты первого многоцентрового ретроспективного исследования эпидемиологии и эффективности терапии ПБЛ у ВИЧ-инфицированных пациентов в Российской Федерации.

Материалы и методы. В исследование включено 26 пациентов с ПБЛ и ВИЧ-инфекцией, получавших лечение или наблюдавшихся в 2012–2019 гг. в 5 российских центрах. Настоящее исследование является частью многоцентрового ретроспективного исследования по изучению эпидемиологии лимфом у пациентов с ВИЧ-инфекцией в России.

Результаты. ПБЛ составила 9,5 % всех лимфом у ВИЧ-инфицированных пациентов, включенных в многоцентровое ретроспективное исследование по изучению эпидемиологии лимфом у пациентов с ВИЧ-инфекцией в России. Эпидемиологические характеристики включенных пациентов соответствовали ранее опубликованным работам: диагностика преимущественно на поздних стадиях заболевания (88 %), поражение слизистых оболочек полости рта и носа, часто с вовлечением костей лицевого скелета (65 %), отсутствие оптимального контроля ВИЧ-инфекции (66,7 %). В качестве первой линии пациентам чаще всего проводилась EPOCH-подобная терапия (50 %). Однако эффективность первичной терапии была неудовлетворительная. Общая выживаемость (ОВ) и выживаемость без прогрессирования (ВБП) в течение 1 года после начала первой линии терапии составили 57 и 46 % соответственно. Включение бортезомиба в первую линию терапии связано с тенденцией к более благоприятному прогнозу. У половины пациентов регистрируется рецидив или прогрессирование лимфомы после первичного лечения. Наиболее часто используемым режимом второй линии терапии была схема DHAP. Общий ответ на терапию второй линии 38,5 %. После начала второй линии терапии 1-летние ОВ и ВБП составили 26 и 15 % соответственно.

Заключение. Пациенты с ПБЛ на фоне ВИЧ-инфекции имеют неблагоприятный прогноз. Усилия по улучшению прогноза пациентов с ПБЛ на фоне ВИЧ-инфекции должны быть направлены на повышение эффективности первой линии терапии и предполагают включение интенсивных схем химиотерапии с использованием бортезомиба. Место аутоТГСК и аллоТГСК в терапии ПБЛ четко не определено, однако пациентов с ПБЛ, несмотря на ВИЧ-инфекцию, следует рассматривать как кандидатов на аутоТГСК в первой ремиссии и на аллоТГСК в случае развития рецидива. Необходимы проспективные многоцентровые исследования с целью оптимизировать лечение пациентов с ПБЛ на фоне ВИЧ-инфекции.

Ключевые слова: плазмобластная лимфома, ВИЧ-инфекция, вирус Эпштейна—Барр, MYC, PD-1/PD-L1/2, аутоТГСК, аллоТГСК, бортезомиб, ниволумаб, иммунотерапия.

Получено: 20 июля 2021 г.

Принято в печать: 27 ноября 2021 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. World Health Organization. HIV data and statistics. Available from: https://www.who.int/teams/global-hiv-hepatitis-and-stis-programmes/hiv/strategic-information/hiv-data-and-statistics (accessed 20.07.2021).
  2. Shiels MS, Pfeiffer RM, Gail MH, et al. Cancer burden in the HIV-infected population in the United States. J Natl Cancer Inst. 2011;103(9):753–62. doi: 10.1093/jnci/djr076.
  3. Heron M. Deaths: Leading Causes for 2017. Natl Vital Stat Rep. 2019;68(6):1–77.
  4. Selik RM, Mokotoff ED, Branson B, et al. Revised Surveillance Case Definition for HIV Infection — United States, 2014. Morbid Mortal Weekly Rep. 2014;63(RR-03):1–10.
  5. Robbins HA, Shiels MS, Pfeiffer RM, et al. Epidemiologic contributions to recent cancer trends among HIV-infected people in the United States. AIDS. 2014;28(6):881–90. doi: 10.1097/QAD.0000000000000163.
  6. Goncalves PH, Montezuma-Rusca JM, Yarchoan R, Uldrick TS. Cancer prevention in HIV-infected populations. Semin Oncol. 2016;43(1):173–88. doi: 10.1053/j.seminoncol.2015.09.011.
  7. Delecluse HJ, Anagnostopoulos I, Dallenbach F, et al. Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection. 1997;89(4):1413–20.
  8. 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.
  9. Knowles DM, Chadburn A Lymphadenopathy and the lymphoid neoplasms associated with the acquired immune deficiency syndrome (AIDS). In: Knowles DM, ed. Neoplastic Hematopathology. Baltimore: Williams and Walker; 1992. pp. 773.
  10. Zuze T, Painschab MS, Seguin R, et al. Plasmablastic lymphoma in Malawi. Infect Agent Cancer. 2018;13:22. doi: 10.1186/s13027-018-0195-4.
  11. Liu JJ, Zhang L, Ayala E, et al. Human immunodeficiency virus (HIV)-negative plasmablastic lymphoma: a single institutional experience and literature review. Leuk Res. 2011;35(12):1571–7. doi: 10.1016/j.leukres.2011.06.023.
  12. Tchernonog E, Faurie P, Coppo P, et al. Clinical characteristics and prognostic factors of plasmablastic lymphoma patients: analysis of 135 patients from the LYSA group. Ann Oncol. 2017;28(4):843–8. doi: 10.1093/annonc/mdw684.
  13. Castillo JJ, Winer ES, Stachurski D, et al. Clinical and pathological differences between human immunodeficiency virus-positive and human immunodeficiency virus-negative patients with plasmablastic lymphoma. Leuk Lymphoma. 2010;51(11):2047–53. doi: 10.3109/10428194.2010.516040.
  14. Morscio J, Dierickx D, Nijs J, et al. Clinicopathologic comparison of plasmablastic lymphoma in HIV-positive, immunocompetent, and posttransplant patients: single-center series of 25 cases and meta-analysis of 277 reported cases. Am J Surg Pathol. 2014;38(7):875–86. doi: 10.1097/PAS.0000000000000234.
  15. Loghavi S, Alayed K, Aladily TN, et al. Stage, age, and EBV status impact outcomes of plasmablastic lymphoma patients: a clinicopathologic analysis of 61 patients. J Hematol Oncol. 2015;8:65. doi: 10.1186/s13045-015-0163-z.
  16. Bibas M, Antinori A. EBV and HIV-Related Lymphoma. Mediterr J Hematol Infect Dis. 2009;1(2):e2009032. doi: 10.4084/MJHID.2009.032.
  17. Castillo J, Pantanowitz L, Dezube BJ. HIV-associated plasmablastic lymphoma: lessons learned from 112 published cases. Am J Hematol. 2008;83(10):804–9. doi: 10.1002/ajh.21250.
  18. Dolcetti R, Gloghini A, Caruso A, Carbone A. A lymphomagenic role for HIV beyond immune suppression? Blood. 2016;127(11):1403–9. doi: 10.1182/blood-2015-11-681411.
  19. Valera A, Balague O, Colomo L, et al. IG/MYC rearrangements are the main cytogenetic alteration in plasmablastic lymphomas. Am J Surg Pathol. 2010;34(11):1686–94. doi: 10.1097/PAS.0b013e3181f3e29f.
  20. Montes-Moreno S, Martinez-Magunacelaya N, Zecchini-Barrese T, et al. Plasmablastic lymphoma phenotype is determined by genetic alterations in MYC and PRDM1. Mod Pathol. 2017;30(1):85–94. doi: 10.1038/modpathol.2016.162.
  21. Garcia JF, Roncador G, Garcia JF, et al. PRDM1/BLIMP-1 expression in multiple B and T-cell lymphoma. Haematologica. 2006;91(4):467–74.
  22. Liu F, Asano N, Tatematsu A, et al. Plasmablastic lymphoma of the elderly: a clinicopathological comparison with age-related Epstein-Barr virus-associated B cell lymphoproliferative disorder. Histopathology. 2012;61(6):1183–97. doi: 10.1111/j.1365-2559.2012.04339.x.
  23. Boy SC, van Heerden MB, Babb C, et al. Dominant genetic aberrations and coexistent EBV infection in HIV-related oral plasmablastic lymphomas. Oral Oncol. 2011;47(9):883–7. doi: 10.1016/j.oraloncology.2011.06.506.
  24. Gruhne B, Sompallae R, Masucci MG. Three Epstein-Barr virus latency proteins independently promote genomic instability by inducing DNA damage, inhibiting DNA repair and inactivating cell cycle checkpoints. Oncogene. 2009;28(45):3997–4008. doi: 10.1038/onc.2009.258.
  25. Price AM, Luftig MA. Dynamic Epstein-Barr virus gene expression on the path to B-cell transformation. Adv Virus Res. 2014;88:279–313. doi: 10.1016/B978-0-12-800098-4.00006-4.
  26. Ma SD, Hegde S, Young KH, et al. A new model of Epstein-Barr virus infection reveals an important role for early lytic viral protein expression in the development of lymphomas. J Virol. 2011;85(1):165–77. doi: 10.1128/JVI.01512-10.
  27. Shannon-Lowe C, Adland E, Bell AI, et al. Features distinguishing Epstein-Barr virus infections of epithelial cells and B cells: viral genome expression, genome maintenance, and genome amplification. J Virol. 2009;83(15):7749–60. doi: 10.1128/JVI.00108-09.
  28. Di Pietro A. Epstein-Barr Virus Promotes B Cell Lymphomas by Manipulating the Host Epigenetic Machinery. Cancers (Basel). 2020;12(10):3037. doi: 10.3390/cancers12103037.
  29. Montes-Moreno S, Gonzalez-Medina AR, Rodriguez-Pinilla SM, et al. Aggressive large B-cell lymphoma with plasma cell differentiation: immunohistochemical characterization of plasmablastic lymphoma and diffuse large B-cell lymphoma with partial plasmablastic phenotype. Haematologica. 2010;95(8):1342–9. doi: 10.3324/haematol.2009.016113.
  30. Harmon CM, Smith LB. Plasmablastic Lymphoma: A Review of Clinicopathologic Features and Differential Diagnosis. Arch Pathol Lab Med. 2016;140(10):1074–8. doi: 10.5858/arpa.2016-0232-RA.
  31. Grimm KE, O’Malley DP. Aggressive B cell lymphomas in the 2017 revised WHO classification of tumors of hematopoietic and lymphoid tissues. Ann Diagn Pathol. 2019;38:6–10. doi: 10.1016/j.anndiagpath.2018.09.014.
  32. Ramis-Zaldivar JE, Gonzalez-Farre B, Nicolae A, et al. MAP-kinase and JAK-STAT pathways dysregulation in plasmablastic lymphoma. Haematologica. 2021;106(10):2682–93. doi: 10.3324/haematol.2020.271957.
  33. Nasta SD, Carrum GM, Shahab I, et al. Regression of a plasmablastic lymphoma in a patient with HIV on highly active antiretroviral therapy. Leuk Lymphoma. 2002;43(2):423–6. doi: 10.1080/10428190290006260.
  34. Lester R, Li C, Phillips P, et al. Improved outcome of human immunodeficiency virus-associated plasmablastic lymphoma of the oral cavity in the era of highly active antiretroviral therapy: a report of two cases. Leuk Lymphoma. 2004;45(9):1881–5. doi: 10.1080/10428190410001697395.
  35. Armstrong R, Bradrick J, Liu YC. Spontaneous regression of an HIV-associated plasmablastic lymphoma in the oral cavity: a case report. J Oral Maxillofac Surg. 2007;65(7):1361–4. doi: 10.1016/j.joms.2005.12.039.
  36. Yordanova K, Stilgenbauer S, Bohle RM, et al. Spontaneous regression of a plasmablastic lymphoma with MYC rearrangement. Br J Haematol. 2019;186(6):e203–e207. doi: 10.1111/bjh.16082.
  37. Natkunam Y, Gratzinger D, Chadburn A, et al. Immunodeficiency-associated lymphoproliferative disorders: time for reappraisal? Blood. 2018;132(18):1871–8. doi: 10.1182/blood-2018-04-842559.
  38. Vaubell JI, Sing Y, Ramburan A, et al. Pediatric plasmablastic lymphoma: a clinicopathologic study. Int J Surg Pathol. 2014;22(7):607–16. doi: 10.1177/1066896914531815.
  39. Jariwal R, Raza N, Bhandohal J, Cobos E. Non-Hodgkin’s Plasmablastic Lymphoma as Initial Presentation of Human Immunodeficiency Virus. J Investig Med High Impact Case Rep. 2021;9:23247096211014689. doi: 10.1177/23247096211014689.
  40. Little RF, Dunleavy K. Update on the treatment of HIV-associated hematologic malignancies. Hematology Am Soc Hematol Educ Program. 2013;2013(1):382–8. doi: 10.1182/asheducation-2013.1.382.
  41. Schommers P, Wyen C, Hentrich M, et al. Poor outcome of HIV-infected patients with plasmablastic lymphoma: results from the German AIDS-related lymphoma cohort study. AIDS. 2013;27(5):842–5. doi: 10.1097/QAD.0b013e32835e069d.
  42. Dittus C, Grover N, Ellsworth S, et al. Bortezomib in combination with dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin) induces long-term survival in patients with plasmablastic lymphoma: a retrospective analysis. Leuk Lymphoma. 2018;59(9):2121–7. doi: 10.1080/10428194.2017.1416365.
  43. Barta SK, Xue X, Wang D, et al. Treatment factors affecting outcomes in HIV-associated non-Hodgkin lymphomas: a pooled analysis of 1546 patients. Blood. 2013;122(19):3251–62. doi: 10.1182/blood-2013-04-498964.
  44. Castillo JJ, Furman M, Beltran BE, et al. Human immunodeficiency virus-associated plasmablastic lymphoma: poor prognosis in the era of highly active antiretroviral therapy. Cancer. 2012;118(21):5270–7. doi: 10.1002/cncr.27551.
  45. Ibrahim IF, Shapiro GA, Naina HVK. Treatment of HIV-associated plasmablastic lymphoma: A single-center experience with 25 patients. J Clin Oncol. 2014;32(5 Suppl):8583. doi: 10.1200/jco.2014.32.15_suppl.8583.
  46. NCCN Guidelines Version: 4.2021. B-Cell Lymphomas. Available from: https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf. (accessed 20.07.2021).
  47. Antinori A, Cingolani A, Alba L, et al. Better response to chemotherapy and prolonged survival in AIDS-related lymphomas responding to highly active antiretroviral therapy. AIDS. 2001;15(12):1483–91. doi: 10.1097/00002030-200108170-00005.
  48. Guan B, Zhang X, Ma H, et al. Meta-analysis of highly active anti-retroviral therapy for treatment of plasmablastic lymphoma. Hematol Oncol Stem Cell Ther. 2010;3(1):7–12. doi: 10.1016/s1658-3876(10)50050-5.
  49. Francischini E, Martins FM, Braz-Silva PH, et al. HIV-associated oral plasmablastic lymphoma and role of adherence to highly active antiretroviral therapy. Int J STD AIDS. 2010;21(1):68–70. doi: 10.1258/ijsa.2008.008476.
  50. Re A, Cattaneo C, Michieli M, et al. High-dose therapy and autologous peripheral-blood stem-cell transplantation as salvage treatment for HIV-associated lymphoma in patients receiving highly active antiretroviral therapy. J Clin Oncol. 2003;21(23):4423–7. doi: 10.1200/JCO.2003.06.039.
  51. Re A, Michieli M, Casari S, et al. High-dose therapy and autologous peripheral blood stem cell transplantation as salvage treatment for AIDS-related lymphoma: long-term results of the Italian Cooperative Group on AIDS and Tumors (GICAT) study with analysis of prognostic factors. Blood. 2009;114(7):1306–13. doi: 10.1182/blood-2009-02-202762.
  52. Popova MO, Rogacheva YA, Nekrasova AV, et al. Autologous hematopoietic cell transplantation for HIV-related lymphoma: results of a single center (CIC725) matched case-control study. Cell Ther Transplant. 2017;6(4):42–51. doi: 10.18620/ctt-1866-8836-2017-6-4-42-51.
  53. Popova M, Tsygankov I, Rogacheva Y, et al. High-dose chemotherapy with autologous hematopoietic stem cell transplantation in patients with HIV-related lymphoma. Annals Oncol. 2020;31(Suppl 4):S651. doi: 10.1016/j.annonc.2020.08.010.
  54. Younan P, Kowalski J, Kiem HP. Genetic modification of hematopoietic stem cells as a therapy for HIV/AIDS. Viruses. 2013;5(12):2946–62. doi: 10.3390/v5122946.
  55. Mock U, Machowicz R, Hauber I, et al. mRNA transfection of a novel TAL effector nuclease (TALEN) facilitates efficient knockout of HIV co-receptor CCR5. Nucl Acids Res. 2015;43(11):5560–71. doi: 10.1093/nar/gkv469.
  56. Wang CX, Cannon PM. The clinical applications of genome editing in HIV. 2016;127(21):2546–52. doi: 10.1182/blood-2016-01-678144.
  57. Попова М.О., Сергеев В.С., Лепик К.В. и др. Генная клеточная терапия ВИЧ и злокачественных опухолей кроветворной и лимфатической ткани на основе трансплантации гемопоэтических стволовых клеток с использованием сайт-специфического редактирования генома. Журнал инфектологии. 2017;9(1):31–9. doi: 10.22625/2072-6732-2017-9-1-31-39.
    [Popova MО, Sergeev VS, Lepik KV, et al. Gene-Cell Therapy of HIV and Hematological Malignances Based on Hematopoietic Stem Cell Transplantation and Site-Specific Genome Editing. Journal Infectology. 2017;9(1):31–9. doi: 10.22625/2072-6732-2017-9-1-31-39. (In Russ)]
  58. Morgan RA, Gray D, Lomova A, et al. Hematopoietic Stem Cell Gene Therapy: Progress and Lessons Learned. Cell Stem Cell. 2017;21(5):574–90. doi: 10.1016/j.stem.2017.10.010.
  59. Popova MO, Lepik KV, Sergeev VS, et al. Clinical implementation of genome editing for correction of human diseases. Cell Ther Transplant. 2017;6(1):37–43. doi: 10.18620/ctt-1866-8836-2017-6-1-37-43.
  60. Serrano D, Carrion R, Balsalobre P, et al. Spanish Cooperative Groups GELTAMO and GESIDA. HIV-associated lymphoma successfully treated with peripheral blood stem cell transplantation. Exp Hematol. 2005;33(4):487–94. doi: 10.1016/j.exphem.2004.12.008.
  61. Bowden RA, Coombs RW, Nikora BH, et al. Progression of human immunodeficiency virus type-1 infection after allogeneic marrow transplantation. Am J Med. 1990;88(5N):49N–52N.
  62. Al-Malki MM, Castillo JJ, Sloan JM, et al. Hematopoietic cell transplantation for plasmablastic lymphoma: a review. Biol Blood Marrow Transplant. 2014;20(12):1877–84. doi: 10.1016/j.bbmt.2014.06.009.
  63. Gupta V, Tomblyn M, Pedersen TL, et al. Allogeneic hematopoietic cell transplantation in human immunodeficiency virus-positive patients with hematologic disorders: a report from the center for international blood and marrow transplant research. Biol Blood Marrow Transplant. 2009;15(7):864–71. doi: 10.1016/j.bbmt.2009.03.023.
  64. Hamadani M, Devine SM. Reduced-intensity conditioning allogeneic stem cell transplantation in HIV patients with hematologic malignancies: yes, we can. Blood. 2009;114(12):2564–6. doi: 10.1182/blood-2009-06-229666.
  65. Afanasyev BV, Popova MO, Bondarenko SN, et al. St. Petersburg experience of allogeneic hematopoietic stem cell transplantation in patients with acute leukemia and human immunodeficiency virus. Cell Ther Transplant. 2015;4(1–2):24–30. doi: 10.18620/1866-8836-2015-4-1-2-24-30.
  66. Yoon JH, Jeon YW, Lee SE, et al. Allogeneic stem cell transplantation using lymphoablative rather than myeloablative conditioning regimen for relapsed or refractory lymphomas. Hematol Oncol. 2017;35(1):17–24. doi: 10.1002/hon.2201.
  67. Rong C, Sheng L, Wu A, et al. Allogeneic hematopoietic stem cell transplantation in a patient with HIV-negative recurrent plasmablastic lymphoma: A case report. Medicine (Baltimore). 2021;100(7):e24498. doi: 10.1097/MD.0000000000024498.
  68. Lepik KV, Mikhailova NB, Moiseev IS, et al. Nivolumab for the treatment of relapsed and refractory classical Hodgkin lymphoma after ASCT and in ASCT-naive patients. Leuk Lymphoma. 2019;60(9):2316–9. doi: 10.1080/10428194.2019.1573368.
  69. Ambinder RF, Wu J, Logan B, et al. Allogeneic Hematopoietic Cell Transplant for HIV Patients with Hematologic Malignancies: The BMT CTN-0903/AMC-080 Trial. Biol Blood Marrow Transplant. 2019;25(11):2160–6. doi: 10.1016/j.bbmt.2019.06.033.
  70. Umeanaeto O, Gamboa J, Diaz J, et al. Incorporating Bortezomib in the Management of Plasmablastic Lymphoma. Anticancer Res. 2019;39(9):5003–7. doi: 10.21873/anticanres.13690.
  71. Castillo JJ, Reagan JL, Sikov W, et al. Bortezomib in combination with infusional dose-adjusted EPOCH for the treatment of plasmablastic lymphoma. Br J Haematol. 2015;169(3):352–5. doi: 10.1111/bjh.1330.
  72. Cao C, Liu T, Zhu H, et al. Bortezomib-contained chemotherapy and thalidomide combined with CHOP (Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone) play promising roles in plasmablastic lymphoma: a case report and literature review. Clin Lymphoma Myeloma Leuk. 2014;14(5):e145–е150. doi: 10.1016/j.clml.2014.03.002.
  73. Cencini E, Fabbri A, Guerrini S, et al. Long-term remission in a case of plasmablastic lymphoma treated with COMP (cyclophosphamide, liposomal doxorubicin, vincristine, prednisone) and bortezomib. Eur J Haematol. 2016;96(6):650–4. doi: 10.1111/ejh.12732.
  74. Colomo L, Loong F, Rives S, et al. Diffuse large B-cell lymphomas with plasmablastic differentiation represent a heterogeneous group of disease entities. Am J Surg Pathol. 2004;28(6):736–47. doi: 10.1097/01.pas.0000126781.87158.e3.
  75. Holderness BM, Malhotra S, Levy NB, Danilov AV. Brentuximab vedotin demonstrates activity in a patient with plasmablastic lymphoma arising from a background of chronic lymphocytic leukemia. J Clin Oncol. 2013;31(12):e197–е199. doi: 10.1200/JCO.2012.46.9593.
  76. Pretscher D, Kalisch A, Wilhelm M, et al. Refractory plasmablastic lymphoma-a review of treatment options beyond standard therapy. Ann Hematol. 2017;96(6):967–70. doi: 10.1007/s00277-016-2904-7.
  77. Laurent C, Fabiani B, Do C, et al. Immune-checkpoint expression in Epstein-Barr virus positive and negative plasmablastic lymphoma: a clinical and pathological study in 82 patients. Haematologica. 2016;101(8):976–84. doi: 10.3324/haematol.2016.141978.
  78. Damlaj M, Alzayed M, Alahmari B, et al. Therapeutic Potential of Checkpoint Inhibitors in Refractory Plasmablastic Lymphoma. Clin Lymphoma Myeloma Leuk. 2019;19(10):e559–e563. doi: 10.1016/j.clml.2019.06.008.
  79. Popova M, Rogacheva Y, Tsygankov I, et al. Immunotherapy in patients with relapsed/refractory HIV-related lymphomas. Annals Oncol. 2019;30(11):XI22. doi: 10.1093/annonc/mdz449.015.
  80. Lepik KV, Mikhailova NB, Kondakova EV, et al. A Study of Safety and Efficacy of Nivolumab and Bendamustine (NB) in Patients With Relapsed/Refractory Hodgkin Lymphoma After Nivolumab Monotherapy Failure. Hemasphere. 2020;4(3):e401. doi: 10.1097/HS9.0000000000000401.
  81. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059–68. doi: 10.1200/JCO.2013.54.8800.
  82. Каприн А.Д., Старинский В.В., Шахзадова А.О. Состояние онкологической помощи населению России в 2019 году. М.: МНИОИ им. П.А. Герцена — филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2020. [Kaprin AD, Starinskii VV, Shakhzadova AO. Sostoyanie onkologicheskoi pomoshchi naseleniyu Rossii v 2019 godu. (The state of cancer care in Russia in 2019.) Moscow: MNIOI im. P.A. Gertsena — filial FGBU “NMITs radiologii” Publ.; (In Russ)]
  83. Информационные бюллетени Федерального научно-методического центра по профилактике и борьбе со СПИДом: Бюллетень № 40 (электронный документ). Доступно по: http://www.hivrussia.ru/files/bulpdf. Ссылка активна на 20.07.2021.
    [Information Bulletins of Federal Scientific and Methodological Center for the Prevention and Control of AIDS: Bulletin No. 40 (Internet). Available from: http://www.hivrussia.ru/files/bul_40.pdf. (accessed 20.07.2021) (In Russ)]

Системное Т-клеточное лимфопролиферативное заболевание, ассоциированное с вирусом Эпштейна—Барр: обзор литературы и собственное клиническое наблюдение

Е.А. Шаламова, А.М. Ковригина, И.А. Шуплецова, Е.Е. Никулина, В.Д. Латышев, Н.В. Цветаева

ФГБУ «НМИЦ гематологии» Минздрава России, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167

Для переписки: Алла Михайловна Ковригина, д-р биол. наук, Новый Зыковский пр-д, д. 4, Москва, Российская Федерация, 125167; e-mail: kovrigina.alla@gmail.com

Для цитирования: Шаламова Е.А., Ковригина А.М., Шуплецова И.А. и др. Системное Т-клеточное лимфопролиферативное заболевание, ассоциированное с вирусом Эпштейна—Барр: обзор литературы и собственное клиническое наблюдение. Клиническая онкогематология. 2021;14(4):477–87.

DOI: 10.21320/2500-2139-2021-14-4-477-487


РЕФЕРАТ

Вирус Эпштейна—Барр (ВЭБ) является широко распространенным и выявляется у 90–95 % взрослого населения. Его реактивация в условиях иммунодефицита часто приводит к клональной трансформации В-лимфоцитов, развитию В-клеточных лимфопролиферативных заболеваний (ЛПЗ) и В-клеточных лимфом. В то же время в странах Северо-Восточной и Восточной Азии, Латинской Америки у пациентов без выявленного иммунодефицита описаны случаи развития Т-клеточных лимфопролиферативных заболеваний, связанных с ВЭБ. В настоящей работе представлено редкое наблюдение системного T-ЛПЗ, ассоциированного с ВЭБ, протекающего с лимфаденопатией, спленомегалией в сочетании с острой формой аутоиммунной гемолитической анемии у мужчины европеоидной расы. Комплексный анализ анамнестических, патоморфологических и лабораторных данных позволил дифференцировать данное заболевание с Т-клеточной лимфомой и выбрать обоснованную тактику ведения пациента.

Ключевые слова: лимфопролиферативное заболевание, вирус Эпштейна—Барр, ВЭБ+ T-ЛПЗ, диагностика, патоморфология.

Получено: 30 мая 2021 г.

Принято в печать: 2 сентября 2021 г.

Читать статью в PDF

Статистика Plumx русский

ЛИТЕРАТУРА

  1. Smatti MK, Al-Sadeq DW, Ali NH, et al. Epstein-Barr Virus Epidemiology, Serology, and Genetic Variability of LMP-1 Oncogene Among Healthy Population: An Update. Front Oncol. 2018;8:211. doi: 10.3389/fonc.2018.00211.
  2. Kuri A, Jacobs BM, Vickaryous N, et al. Epidemiology of Epstein-Barr virus infection and infectious mononucleosis in the United Kingdom. BMC Public Health. 2020;20(1):912. doi: 10.1186/s12889-020-09049-x.
  3. Rostgaard K, Balfour HH Jr, Jarrett R, et al. Primary Epstein-Barr virus infection with and without infectious mononucleosis. PLoS One. 2019;14(12):e0226436. doi: 10.1371/journal.pone.0226436.
  4. Montes-Mojarro IA, Kim WY, Fend F, Quintanilla-Martinez L. Epstein-Barr virus positive T and NK-cell lymphoproliferations: Morphological features and differential diagnosis. Semin Diagn Pathol. 2020;37(1):32–46. doi: 10.1053/j.semdp.2019.12.004.
  5. Shannon-Lowe C, Rickinson A. The Global Landscape of EBV-Associated Tumors. Front Oncol. 2019;9:713. doi: 10.3389/fonc.2019.00713.
  6. Pei Y, Lewis AE, Robertson ES. Current Progress in EBV-Associated B-Cell Lymphomas. Adv Exp Med Biol. 2017;1018:57–74. doi: 10.1007/978-981-10-5765-6_5.
  7. Martinez OM, Krams SM. The Immune Response to Epstein Barr Virus and Implications for Posttransplant Lymphoproliferative Disorder. 2017;101(9):2009–16. doi: 10.1097/TP.0000000000001767.
  8. Compagno F, Basso S, Panigari A, et al. Management of PTLD After Hematopoietic Stem Cell Transplantation: Immunological Perspectives. Front Immunol. 2020;11:567020. doi: 10.3389/fimmu.2020.567020.
  9. Ковригина А.М. ВЭБ-позитивные лимфопролиферативные заболевания: новая концепция, дифференциальная диагностика (обзор литературы и собственные наблюдения). Клиническая онкогематология. 2018;11(4):326–37. doi: 10.21320/2500-2139-2018-11-4-326-337.
    [Kovrigina AM. EBV-Positive Lymphoproliferative Diseases: A New Concept and Differential Diagnosis (Literature Review and Case Reports). Clinical oncohematology. 2018;11(4):326–37. doi: 10.21320/2500-2139-2018-11-4-326-337. (In Russ)]
  10. Kimura H, Fujiwara S. Overview of EBV-Associated T/NK-Cell Lymphoproliferative Diseases. Front Pediatr. 2019;6:417. doi: 10.3389/fped.2018.00417.
  11. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Revised 4th edition. Lyon: IARC Press; 2017. pp. 358–60.
  12. Coffey AM, Lewis A, Marcogliese AN, et al. A clinicopathologic study of the spectrum of systemic forms of EBV‐associated T‐cell lymphoproliferative disorders of childhood: A single tertiary care pediatric institution experience in North America. Pediatr Blood Cancer. 2019;66(8):e27798. doi: 10.1002/pbc.27798.
  13. Ohshima K, Kimura H, Yoshino T, et al. Proposed categorization of pathological states of EBV-associated T/natural killer-cell lymphoproliferative disorder (LPD) in children and young adults: overlap with chronic active EBV infection and infantile fulminant EBV T-LPD. Pathol Int. 2008;58(4):209–17. doi: 10.1111/j.1440-1827.2008.02213.x.
  14. Kawamoto K, Miyoshi H, Suzuki T, et al. A distinct subtype of Epstein-Barr virus-positive T/NK-cell lymphoproliferative disorder: adult patients with chronic active Epstein-Barr virus infection-like features. 2018;103(6):1018–28. doi: 10.3324/haematol.2017.174177.
  15. Fujiwara S, Kimura H, Imadome K, et al. Current research on chronic active Epstein-Barr virus infection in Japan. Pediatr Int. 2014;56(2):159–66. doi: 10.1111/ped.12314.
  16. van Dongen JJ, Langerak AW, Bruggemann M, et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia. 2003;17(12):2257–317. doi: 10.1038/sj.leu.2403202.
  17. Чернова Н.Г., Сидорова Ю.В., Смирнова С.Ю. и др. Молекулярная диагностика ангиоиммунобластной Т-клеточной лимфомы. Терапевтический архив. 2019;91(7):63–9. doi: 10.26442/00403660.2019.07.000330.
    [Chernova NG, Sidorova YuV, Smirnova SYu, et al. Molecular diagnosis angioimmunoblastic T-cell lymphoma. Terapevticheskii arkhiv. 2019;91(7):63–9. doi: 10.26442/00403660.2019.07.000330. (In Russ)]
  18. Cohen JI, Jaffe ES, Dale JK, et al. Characterization and treatment of chronic active Epstein-Barr virus disease: a 28-year experience in the United States. 2011;117(22):5835–49. doi: 10.1182/blood-2010-11-316745.
  19. Arai A. Advances in the Study of Chronic Active Epstein-Barr Virus Infection: Clinical Features Under the 2016 WHO Classification and Mechanisms of Development. Front Pediatr. 2019;7:14. doi: 10.3389/fped.2019.00014.
  20. Fournier B, Boutboul D, Bruneau J, et al. Rapid identification and characterization of infected cells in blood during chronic active Epstein-Barr virus infection. J Exp Med. 2020;217(11):e20192262. doi: 10.1084/jem.20192262.
  21. Kawabe S, Ito Y, Gotoh K, et al. Application of flow cytometric in situ hybridization assay to Epstein-Barr virus-associated T/natural killer cell lymphoproliferative diseases. Cancer Sci. 2012;103(8):1481–8. doi: 10.1111/j.1349-7006.2012.02305.x.
  22. Paik JH, Choe JY, Kim H, et al. Clinicopathological categorization of Epstein-Barr virus-positive T/NK-cell lymphoproliferative disease: an analysis of 42 cases with an emphasis on prognostic implications. Leuk Lymphoma. 2017;58(1):53–63. doi: 10.1080/10428194.2016.1179297.
  23. Kimura H. EBV in T-/NK-Cell Tumorigenesis. Adv Exp Med Biol. 2018;1045:459–75. doi: 10.1007/978-981-10-7230-7_21.
  24. Takada H, Imadome KI, Shibayama H, et al. EBV induces persistent NF-κB activation and contributes to survival of EBV-positive neoplastic T- or NK-cells. PLoS One. 2017;12(3):e0174136. doi: 10.1371/journal.pone.0174136.
  25. Okuno Y, Murata T, Sato Y, et al. Defective Epstein-Barr virus in chronic active infection and haematological malignancy. Nat Microbiol. 2019;4(3):404–13. doi: 10.1038/s41564-018-0334-0.
  26. Katano H, Ali MA, Patera AC, et al. Chronic active Epstein-Barr virus infection associated with mutations in perforin that impair its maturation. 2004;103(4):1244–52. doi: 10.1182/blood-2003-06-2171.
  27. Beer T, Dorion P. Angioimmunoblastic T-Cell Lymphoma Presenting with an Acute Serologic Epstein-Barr Virus Profile. Hematol Rep. 2015;7(2):5893. doi: 10.4081/hr.2015.5893.
  28. Steciuk MR, Massengill S, Banks PM. In immunocompromised patients, Epstein-Barr virus lymphadenitis can mimic angioimmunoblastic T-cell lymphoma morphologically, immunophenotypically, and genetically: a case report and review of the literature. Hum Pathol. 2012;43(1):127–33. doi: 10.1016/j.humpath.2011.02.024.
  29. Chiba S, Sakata-Yanagimoto M. Advances in understanding of angioimmunoblastic T-cell lymphoma. 2020;34(10):2592–606. doi: 10.1038/s41375-020-0990-y.
  30. Yabe M, Dogan A, Horwitz SM, Moskowitz AJ. Angioimmunoblastic T-Cell Lymphoma. In: Querfeld C, Zain J, Rosen S, eds. T-Cell and NK-Cell Lymphomas. Cancer Treatment and Research. Springer; Vol. 176. pp. 99–126. doi: 10.1007/978-3-319-99716-2_5.
  31. Kato S, Takahashi E, Asano N, et al. Nodal cytotoxic molecule (CM)-positive Epstein-Barr virus (EBV)-associated peripheral T cell lymphoma (PTCL): a clinicopathological study of 26 cases. 2012;61(2):186–99. doi: 10.1111/j.1365-2559.2012.04199.x.
  32. Jeon YK, Kim J-H, Sung J-Y, et al.; Hematopathology Study Group of the Korean Society of P. Epstein-Barr virus-positive nodal T/NK-cell lymphoma: an analysis of 15 cases with distinct clinicopathological features. Hum Pathol. 2015;46(7):981–90. doi: 10.1016/j.humpath.2015.03.002.
  33. Takahashi E, Asano N, Li C, et al. Nodal T/NK-cell lymphoma of nasal type: a clinicopathological study of six cases. 2008;52(5):585–96. doi: 10.1111/j.1365-2559.2008.02997.x.
  34. Ng SB, Chung TH, Kato S, et al. Epstein-Barr virus-associated primary nodal T/NK-cell lymphoma shows a distinct molecular signature and copy number changes. 2018;103(2):278–87. doi: 10.3324/haematol.2017.180430.
  35. Edwards ESJ, Bier J, Cole TS, et al. Activating PIK3CD mutations impair human cytotoxic lymphocyte differentiation and function and EBV immunity. J Allergy Clin Immunol. 2019;143(1):276–291.e6. doi: 10.1016/j.jaci.2018.04.030.
  36. Latour S, Fischer A. Signaling pathways involved in the T-cell-mediated immunity against Epstein-Barr virus: Lessons from genetic diseases. Immunol Rev. 2019;291(1):174–89. doi: 10.1111/imr.12791.
  37. Files JK, Boppana S, Perez MD, et al. Sustained cellular immune dysregulation in individuals recovering from SARS-CoV-2 infection. J Clin Invest. 2021;131(1):e140491. doi: 10.1172/JCI140491.
  38. Liu J, Yang X, Wang H, et al. The analysis of the long-term impact of SARS-CoV-2 on the cellular immune system in individuals recovering from COVID-19 reveals a profound NK/T cell impairment. mBio. 2021 (Preprint). doi: 10.1101/2020.08.21.20179358.
  39. Kovoor JG, Scott NA, Tivey DR, et al. Proposed delay for safe surgery after COVID-19. ANZ J Surg. 2021;91(4):495–506. doi: 10.1111/ans.16682.
  40. Dematapitiya C, Perera C, Chinthaka W, et al. Cold type autoimmune hemolytic anemia – a rare manifestation of infectious mononucleosis; serum ferritin as an important biomarker. BMC Infect Dis. 2019;19(1):68. doi: 10.1186/s12879-019-3722-z.
  41. Teijido J, Tillotson K, Liu JM. A Rare Presentation of Epstein-Barr Virus Infection. J Emerg Med. 2020;58(2):e71-e73. doi: 10.1016/j.jemermed.2019.11.043.
  42. Whitelaw F, Brook MG, Kennedy N, Weir WR. Haemolytic anaemia complicating Epstein-Barr virus infection. Br J Clin Pract. 1995;49(4):212–3.
  43. Aveiro M, Ferreira G, Matias C, et al. Hard-To-Treat Idiopathic Refractory Autoimmune Haemolytic Anaemia with Reticulocytopenia. Eur J Case Rep Intern Med. 2020;7(12):002112. doi: 10.12890/2020_002112.
  44. Fattizzo B, Giannotta JA, Serpenti F, Barcellini W. Difficult Cases of Autoimmune Hemolytic Anemia: A Challenge for the Internal Medicine Specialist. J Clin Med. 2020;9(12):3858. doi: 10.3390/jcm9123858.
  45. Barcellini W, Fattizzo B, Zaninoni A, et al. Clinical heterogeneity and predictors of outcome in primary autoimmune hemolytic anemia: a GIMEMA study of 308 patients. 2014;124(19):2930–6. doi: 10.1182/blood-2014-06-583021.
  46. Barcellini W, Fattizzo B. Clinical Applications of Hemolytic Markers in the Differential Diagnosis and Management of Hemolytic Anemia. Dis Markers. 2015;2015:635670. doi: 10.1155/2015/635670.
  47. Fink S, Tsai MH, Schnitzler P, et al. The Epstein–Barr virus DNA load in the peripheral blood of transplant recipients does not accurately reflect the burden of infected cells. Transpl Int. 2017;30(1):57–67. doi: 10.1111/tri.12871.
  48. Andrei G, Trompet E, Snoeck R. Novel Therapeutics for Epstein-Barr Virus. 2019;24(5):997. doi: 10.3390/molecules24050997.

Полимеразная цепная реакция в оценке прогноза и мониторинге вирус Эпштейна—Барр-ассоциированной лимфомы Ходжкина

Н.А. Катин1, И.В. Жильцов1, В.М. Семенов1, Д.К. Новик2

1 УО «Витебский государственный медицинский университет», пр-т Фрунзе, д. 27, Витебск, Республика Беларусь, 210023

2 ГУ «Республиканский научно-практический центр радиационной медицины и экологии человека», ул. Ильича, д. 290, Гомель, Республика Беларусь, 246040

Для переписки: Иван Викторович Жильцов, д-р мед. наук, профессор, пр-т Фрунзе, д. 27, Витебск, Республика Беларусь, 210023; тел.: +375(29)7104368-93-29; e-mail: zhyltsou@tut.by

Для цитирования: Катин Н.А., Жильцов И.В., Семенов В.М., Новик Д.К. Полимеразная цепная реакция в оценке прогноза и мониторинге вирус Эпштейна—Барр-ассоциированной лимфомы Ходжкина. Клиническая онкогематология. 2018;11(2):182–6.

DOI: 10.21320/2500-2139-2018-11-2-182-186


РЕФЕРАТ

В обзоре представлен анализ 34 статей, посвященных изучению полимеразной цепной реакции (ПЦР) в качестве метода определения ДНК вируса Эпштейна—Барр (ВЭБ) в биологическом материале у пациентов с ВЭБ-ассоциированными онкологическими заболеваниями, включая лимфому Ходжкина (ЛХ). Приводится сравнительный анализ различных методик определения ДНК ВЭБ в биологическом материале. ВЭБ связан с ЛХ в 20–100 % случаев в зависимости от географического региона и ВИЧ-статуса пациента. Для ВЭБ-ассоциированных ЛХ характерен тип латенции II. Установлено, что ВЭБ находится во всех атипичных клетках и может определяться в крови у пациентов с ВЭБ-ассоциированной ЛХ методом ПЦР. Представлены результаты различных исследований, в которых выявление ВЭБ методом ПЦР сравнивается с методами in situ гибридизации и иммуногистохимии при различных ВЭБ-ассоциированных онкологических заболеваниях, включая ЛХ. Полученные данные указывают на возможность использовать ПЦР для количественного определения ДНК ВЭБ в плазме у больных ЛХ при мониторинге эффективности лечения, оценке прогноза течения и развития рецидивов заболевания. Количественное определение ДНК ВЭБ с помощью метода ПЦР в режиме реального времени в плазме у пациентов с ВЭБ-ассоциированной ЛХ является перспективным методом. Для дальнейшего внедрения в практику необходимы стандартизация методики, проведение более крупных исследований, а также сопоставление с позитронно-эмиссионной томографией.

Ключевые слова: вирус Эпштейна—Барр, лимфома Ходжкина, полимеразная цепная реакция.

Получено: 20 декабря 2017 г.

Принято в печать: 28 февраля 2018 г.

Читать статью в PDF 


ЛИТЕРАТУРА

  1. Thorley-Lawson DA. Epstein-Barr virus: exploiting the immune system. Nat Rev Immunol. 2001;1(1):75–82. doi: 10.1038/35095584.
  2. Murata T. Regulation of Epstein-Barr virus reactivation from latency. Microbiol Immunol. 2014;58(6):307–17. doi: 10.1111/1348-0421.12155.
  3. Shannon-Lowe C, Rickinson AB, Bell AI. Epstein–Barr virus-associated lymphomas. Philos Trans R Soc B Biol Sci. 2017;372(1732):20160271.
  4. Young LS, Murray PG. Epstein-Barr virus and oncogenesis: from latent genes to tumours. Oncogene. 2003;22(33):5108–21. doi: 10.1038/sj.onc.1206556.
  5. Rickinson AB, Rowe M, Hart IJ, et al. T-cell-mediated regression of ‘spontaneous’ and of Epstein-Barr virus-induced B-cell transformation in vitro: studies with cyclosporin A. Cell Immunol. 1984;87(2):646–58. doi: 1016/0008-8749(84)90032-7.
  6. Mancao C, Hammerschmidt W. Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood. 2007;110(10):3715–21. doi: 1182/blood-2007-05-090142.
  7. Massini G, Siemer D, Hohaus S. EBV in Hodgkin Lymphoma. Mediterr J Hematol Infect Dis. 2009;1(2):e2009013. doi: 10.4084/MJHID.2009.013.
  8. Flavell KJ, Murray PG. Hodgkin’s disease and the Epstein-Barr virus. Mol Pathol. 2000;53(5):262–9. doi:1136/mp.53.5.262.
  9. Weinreb M, Day PJ, Niggli F, et al. The role of Epstein-Barr virus in Hodgkin’s disease from different geographical areas. Arch Dis Child. 1996;74(1):27–31. doi: 10.1136/adc.74.1.27.
  10. Gandhi MK, Tellam JT, Khanna R. Epstein-Barr virus-associated Hodgkin’s lymphoma. Br J Haematol. 2004;125(3):267–81. doi: 10.1111/j.1365-2141.2004.04902.x.
  11. Glaser SL, Lin RJ, Stewart SL, et al. Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70(4):375–82. doi: 10.1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.0.co;2-t.
  12. Dolcetti R, Boiocchi M, Gloghini A, et al. Pathogenetic and histogenetic features of HIV-associated Hodgkin’s disease. Eur J Cancer. 2001;37(10):1276–87. doi: 10.1016/S0959-8049(01)00105-8.
  13. Gulley ML, Tang W. Laboratory Assays for Epstein-Barr Virus-Related Disease. J Mol Diagn. 2008;10(4):279–92. doi: 2353/jmoldx.2008.080023.
  14. Maurmann S, Fricke L, Wagner H-J, et al. Molecular parameters for precise diagnosis of asymptomatic Epstein-Barr virus reactivation in healthy carriers. J Clin Microbiol. 2003;41(12):5419–28. doi: 10.1128/jcm.41.12.5419-5428.2003.
  15. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17(9):2885–92. doi: 10.1158/1078-0432.CCR-10-3327.
  16. Qi Z-L, Han X-Q, Hu J, et al. Comparison of three methods for the detection of Epstein-Barr virus in Hodgkin’s lymphoma in paraffin-embedded tissues. Mol Med Rep. 2013;7(1):89–92. doi: 10.3892/mmr.2012.1163.
  17. Ryan JL, Fan H, Glaser SL, et al. Epstein-Barr virus quantitation by real-time PCR targeting multiple gene segments: a novel approach to screen for the virus in paraffin-embedded tissue and plasma. J Mol Diagn. 2004;6(4):378–85. doi: 1016/S1525-1578(10)60535-1.
  18. Lo YM, Leung SF, Chan LY, et al. Kinetics of plasma Epstein-Barr virus DNA during radiation therapy for nasopharyngeal carcinoma. Cancer Res. 2000;60(9):2351-5.
  19. To EWH, Chan KCA, Leung S-F, et al. Rapid clearance of plasma Epstein-Barr virus DNA after surgical treatment of nasopharyngeal carcinoma. Clin Cancer Res. 2003;9(9):3254–9.
  20. Zhao F, Liu X, Chen X, et al. Levels of plasma Epstein-Barr virus DNA prior and subsequent to treatment predicts the prognosis of nasopharyngeal carcinoma. Oncol Lett. 2015;10(5):2888–94. doi: 3892/ol.2015.3628.
  21. Au W-Y, Pang A, Choy C, et al. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104(1):243–9. doi: 1182/blood-2003-12-4197.
  22. Lei KIK, Chan LYS, Chan W-Y, et al. Diagnostic and prognostic implications of circulating cell-free Epstein-Barr virus DNA in natural killer/T-cell lymphoma. Clin Cancer Res. 2002;8(1):29–34.
  23. Hohaus S, Giachelia M, Massini G, et al. Cell-free circulating DNA in Hodgkin’s and non-Hodgkin’s lymphomas. Ann Oncol. 2009;20(8):1408–13. doi: 1093/annonc/mdp006.
  24. Gallagher A, Armstrong AA, MacKenzie J, et al. Detection of Epstein-Barr virus (EBV) genomes in the serum of patients with EBV-associated Hodgkin’s disease. Int J Cancer. 1999;84(4):442–8. doi:1002/(sici)1097-0215(19990820)84:4<442::aid-ijc20>3.0.co;2-j.
  25. Chan KCA, Zhang J, Chan ATC, et al. Molecular characterization of circulating EBV DNA in the plasma of nasopharyngeal carcinoma and lymphoma patients. Cancer Res. 2003;63(9):2028–32.
  26. Lei KIK, Chan LYS, Chan WY, et al. Quantitative analysis of circulating cell-free Epstein-Barr virus (EBV) DNA levels in patients with EBV-associated lymphoid malignancies. Br J Haematol. 2000;111(1):239–46. doi:1111/j.1365-2141.2000.02344.x.
  27. Gandhi MK, Lambley E, Burrows J, et al. Plasma Epstein-Barr virus (EBV) DNA is a biomarker for EBV-positive Hodgkin’s lymphoma. Clin Cancer Res. 2006;12(2):460–4. doi: 1158/1078-0432.CCR-05-2008.
  28. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121(18):3547–53. doi: 1182/blood-2012-09-454694.
  29. Sinha M, Rao CR, Shafiulla M, et al. Plasma Epstein Barr viral load in adult-onset Hodgkin lymphoma in South India. Hematol Oncol Stem Cell Ther. 2016;9(1):8–13. doi: 10.1016/j.hemonc.2015.11.004.
  30. Spacek M, Hubacek P, Markova J, et al. Plasma EBV-DNA monitoring in Epstein-Barr virus-positive Hodgkin lymphoma patients. APMIS. 2011;119(1):10–6. doi: 1111/j.1600-0463.2010.02685.x.
  31. Welch JJG, Schwartz CL, Higman M, et al. Epstein-Barr virus DNA in serum as an early prognostic marker in children and adolescents with Hodgkin lymphoma. Blood Adv. 2017;1(11):681–4. doi: 1182/bloodadvances.2016002618.
  32. De Paoli P, Pratesi C, Bortolin MT. The Epstein Barr virus DNA levels as a tumor marker in EBV-associated cancers. J Cancer Res Clin Oncol. 2007;133(11):809–15. doi: 1007/s00432-007-0281-2.
  33. Le Q-T, Jones CD, Yau T-K, et al. A Comparison Study of Different PCR Assays in Measuring Circulating Plasma Epstein-Barr Virus DNA Levels in Patients with Nasopharyngeal Carcinoma. Clin Cancer Res. 2005;11(16):5700–7. doi: 1158/1078-0432.CCR-05-0648.
  34. Bortolin MT, Pratesi C, Dolcetti R, et al. Clinical value of Epstein–Barr virus DNA levels in peripheral blood samples of Italian patients with undifferentiated carcinoma of nasopharyngeal type. Cancer Lett. 2006;233(2):247–54. doi: 10.1016/j.canlet.2005.03.015.

Вирус Эпштейна—Барр и классическая лимфома Ходжкина

В.Э. Гурцевич

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

Для переписки: Владимир Эдуардович Гурцевич, д-р мед. наук, профессор, Каширское ш., д. 24, Москва, Российская Федерация, 115478; тел.: +7(499)324-25-64; e-mail: gurvlad532@yahoo.com

Для цитирования: Гурцевич В.Э. Вирус Эпштейна—Барр и классическая лимфома Ходжкина. Клиническая онкогематология. 2016;9(2):101–14.

DOI: 10.21320/2500-2139-2016-9-2-101-114


РЕФЕРАТ

Среди онкогенных вирусов человека вирус Эпштейна—Барр (ВЭБ) обращает на себя внимание уникальными свойствами. Этот широко распространенный среди населения планеты вирус одновременно является лидером по числу ассоциированных с ним различных доброкачественных и злокачественных новообразований лимфоидного и эпителиального происхождения. Онкогенный потенциал ВЭБ связан с его способностью инфицировать и трансформировать лимфоциты человека. В тех случаях, когда взаимодействие между размножением ВЭБ, его латентным состоянием и иммунным контролем со стороны организма нарушается, создаются условия для длительной пролиферации инфицированных ВЭБ лимфоцитов и их злокачественной трансформации. По мнению ряда исследователей, молекулярные механизмы связанного с ВЭБ канцерогенеза обусловлены способностью вирусного генома стимулировать экспрессию серии продуктов, имитирующих ряд факторов роста, транскрипции и оказывающих антиапоптотическое действие. Эти кодируемые ВЭБ продукты нарушают сигнальные пути, которые регулируют различные клеточные функции гомеостаза, наделяя клетку способностью к неограниченной пролиферации. Тем не менее точный механизм, с помощью которого ВЭБ инициирует онкогенез, остается не до конца выясненным. В обзоре приводится обобщающая информация о структуре и онкогенном потенциале ВЭБ, морфологических и клинических вариантах лимфомы Ходжкина (ЛХ), а также роли ВЭБ в патогенезе связанных с этим вирусом вариантов ЛХ. Кроме того, в обзоре освещены последние данные об использовании уровня вирусной ДНК ВЭБ в плазме больных ЛХ в качестве биомаркера, отражающего эффективность проведенного лечения и прогноз болезни.


Ключевые слова: вирус Эпштейна—Барр, ВЭБ, латентный мембранный белок 1, LMP1, лимфома Ходжкина, копии ДНК ВЭБ.

Получено: 5 февраля 2016 г.

Принято в печать: 8 февраля 2016 г.

Читать статью в PDF pdficon


ЛИТЕРАТУРА

  1. Zur Hausen H, de Villiers EM. Reprint of: cancer “causation” by infections—individual contributions and synergistic networks. Semin Oncol. 2015;42(2):207–22. doi: 10.1053/j.seminoncol.2015.02.019.
  2. Santos-Juanes J, Fernandez-Vega I, Fuentes N, et al. Merkel cell carcinoma and Merkel cell polyomavirus: a systematic review and meta-analysis. Br J Dermatol. 2015;173(1):42–9. doi: 10.1111/bjd.13870.
  3. Rickinson AB, Young LS, Rowe M. Influence of the Epstein-Barr virus nuclear antigen EBNA 2 on the growth phenotype of virus-transformed B cells. J Virol. 1987;61(5):1310–7.
  4. Rickinson AB, Long HM, Palendira U, et al. Cellular immune controls over Epstein-Barr virus infection: new lessons from the clinic and the laboratory. Trends Immunol. 2014;35(4):159–69. doi: 10.1016/j.it.2014.01.003.
  5. Woodman CB, Collins SI, Vavrusova N, et al. Role of sexual behavior in the acquisition of asymptomatic Epstein-Barr virus infection: a longitudinal study. Pediatr Infect Dis J. 2005;24(6):498–502. doi: 10.1097/01.inf.0000164709.40358.b6.
  6. Henle G, Henle W, Diehl V. Relation of Burkitt’s tumor-associated herpes-type virus to infectious mononucleosis. Proc Natl Acad Sci USA. 1968;59(1):94–101. doi: 10.1073/pnas.59.1.94.
  7. Tanner J, Weis J, Fearon D, et al. Epstein-Barr virus gp350/220 binding to the B lymphocyte C3d receptor mediates adsorption, capping, and endocytosis. Cell. 1987;50(2):203–13. doi:10.1016/0092-8674(87)90216-9.
  8. Connolly SA, Jackson JO, Jardetzky TS, et al. Fusing structure and function: a structural view of the herpesvirus entry machinery. Nat Rev Microbiol. 2011;9(5):369–81. doi: 10.1038/nrmicro2548.
  9. Janz A, Oezel M, Kurzeder C, et al. Infectious Epstein-Barr virus lacking major glycoprotein BLLF1 (gp350/220) demonstrates the existence of additional viral ligands. J Virol. 2000;74(21):10142–52. doi: 10.1128/jvi.74.21.10142-10152.2000.
  10. Ogembo JG, Kannan L, Ghiran I, et al. Human complement receptor type 1/CD35 is an Epstein-Barr Virus receptor. Cell Rep. 2013;3(2):371–85. doi:10.1016/j.celrep.2013.01.023.
  11. Kempkes B, Robertson ES. Epstein-Barr virus latency: current and future perspectives. Curr Opin Virol. 2015;14:138–44. doi: 10.1016/j.coviro.2015.09.007.
  12. Sample J, Kieff E. Transcription of the Epstein-Barr virus genome during latency in growth-transformed lymphocytes. J Virol. 1990;64(4):1667–74.
  13. Babcock GJ, Decker LL, Volk M, et al. EBV persistence in memory B cells in vivo. Immunity. 1998;9(3):395–404. doi: 10.1016/S1074-7613(00)80622-6.
  14. Shannon-Lowe C, Adland E, Bell AI, et al. Features distinguishing Epstein-Barr virus infections of epithelial cells and B cells: viral genome expression, genome maintenance, and genome amplification. J Virol. 2009;83(15):7749–60. doi: 10.1128/JVI.00108-09.
  15. Rickinson A. Epstein-Barr virus. Virus Res. 2002;82(1–2):109–13. doi: 10.1016/s0168-1702(01)00436-1.
  16. Rowe M, Lear AL, Croom-Carter D, et al. Three pathways of Epstein-Barr virus gene activation from EBNA1-positive latency in B lymphocytes. J Virol. 1992;66(1):122–31.
  17. Portis T, Dyck P, Longnecker R. Epstein-Barr Virus (EBV) LMP2A induces alterations in gene transcription similar to those observed in Reed-Sternberg cells of Hodgkin lymphoma. Blood. 2003;102(12):4166–78. doi: 10.1182/blood-2003-04-1018.
  18. Sample J, Young L, Martin B, et al. Epstein-Barr virus types 1 and 2 differ in their EBNA-3A, EBNA-3B, and EBNA-3C genes. J Virol. 1990;64(9):4084–92.
  19. Sixbey JW, Shirley P, Chesney PJ, et al. Detection of a second widespread strain of Epstein-Barr virus. The Lancet. 1989;2(8666):761–5. doi: 10.1016/s0140-6736(89)90829-5.
  20. Gratama JW, Ernberg I. Molecular epidemiology of Epstein-Barr virus infection. Adv Cancer Res. 1995;67:197–255. doi: 10.1016/s0065-230x(08)60714-9.
  21. Young LS, Dawson CW, Eliopoulos AG. The expression and function of Epstein-Barr virus encoded latent genes. Mol Pathol. 2000;53(5):238–47. doi: 10.1136/mp.53.5.238.
  22. Mosialos G, Birkenbach M, Yalamanchili R, et al. The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell. 1995;80(3):389–99. doi: 10.1016/0092-8674(95)90489-1.
  23. Nitta T, Chiba A, Yamashita A, et al. NF-kappaB is required for cell death induction by latent membrane protein 1 of Epstein-Barr virus. Cell Signal. 2003;15(4):423–33. doi: 10.1016/S0898-6568(02)00141-9.
  24. Aviel S, Winberg G, Massucci M, Ciechanover A. Degradation of the Epstein-Barr virus latent membrane protein 1 (LMP1) by the ubiquitin-proteasome pathway. Targeting via ubiquitination of the N-terminal residue. J Biol Chem. 2000;275(31):23491–9. doi: 10.1074/jbc.M002052200.
  25. Gires O, Kohlhuber F, Kilger E, et al. Latent membrane protein 1 of Epstein-Barr virus interacts with JAK3 and activates STAT proteins. EMBO J. 1999;18(11):3064–73. doi: 10.1093/emboj/18.11.3064.
  26. Bentz GL, Whitehurst CB, Pagano JS. Epstein-Barr virus latent membrane protein 1 (LMP1) C-terminal-activating region 3 contributes to LMP1-mediated cellular migration via its interaction with Ubc9. J Virol. 2011;85(19):10144–53. doi: 10.1128/JVI.05035-11.
  27. Wang D, Liebowitz D, Kieff E. An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells. Cell. 1985;43(3):831–40. doi: 10.1016/0092-8674(85)90256-9.
  28. Dawson CW, Port RJ, Young LS. The role of the EBV-encoded latent membrane proteins LMP1 and LMP2 in the pathogenesis of nasopharyngeal carcinoma (NPC). Semin Cancer Biol. 2012;22(2):144–53. doi: 10.1016/j.semcancer.2012.01.004.
  29. Смирнова К.В., Дидук С.В., Сенюта Н.Б., Гурцевич В.Э. Молекулярно-биологические свойства гена LMP1 вируса Эпштейна—Барр: структура, функции и полиморфизм. Вопросы вирусологии. 2015;60(3):5–13.
    [Smirnova KV, Diduk SV, Senyuta NB, Gurtsevich VE. Molecular biological properties of the Epstein-Barr virus LMP1 gene: structure, function, and polymorphism. Voprosy virusologii. 2015;60(3):5–13. (In Russ)]
  30. Vockerodt M, Morgan SL, Kuo M, et al. The Epstein-Barr virus oncoprotein, latent membrane protein-1, reprograms germinal centre B cells towards a Hodgkin’s Reed-Sternberg-like phenotype. J Pathol. 2008;216(1):83–92. doi: 10.1002/path.2384.
  31. Raab-Traub N. Epstein-Barr virus in the pathogenesis of NPC. Semin Cancer Biol. 2002;12:431–41. doi: 10.1016/s1044579x0200086x.
  32. Raab-Traub N. Novel mechanisms of EBV-induced oncogenesis. Curr Opin Virol. 2012;2(4):453–8. doi: 10.1016/j.coviro.2012.07.001.
  33. Soni V, Cahir-McFarland E, Kieff E. LMP1 TRAFficking activates growth and survival pathways. Adv Exp Med Biol. 2007;597:173–87. doi: 10.3390/v5041131.
  34. Man C, Rosa J, Lee LT, et al. Latent membrane protein 1 suppresses RASSF1A expression, disrupts microtubule structures and induces chromosomal aberrations in human epithelial cells. Oncogene. 2007;26(21):3069–80. doi: 10.1038/sj.onc.1210106.
  35. Guo L, Tang M, Yang L, et al. Epstein-Barr virus oncoprotein LMP1 mediates surviving upregulation by p53 contributing to G1/S cell cycle progression in nasopharyngeal carcinoma. Int J Mol Med. 2012;29(4):574–80. doi: 10.3892/ijmm.2012.889.
  36. Horikawa T, Yoshizaki T, Kondo S, et al. Epstein-Barr Virus latent membrane protein 1 induces Snail and epithelial-mesenchymal transition in metastatic nasopharyngeal carcinoma. Br J Cancer. 2011;104(7):1160–7. doi: 10.1038/bjc.2011.38.
  37. Xiao L, Hu ZY, Dong X, et al. Targeting Epstein-Barr virus oncoprotein LMP1-mediated glycolysis sensitizes nasopharyngeal carcinoma to radiation therapy. Oncogene. 2014;33(37):4568–78. doi: 10.1038/onc.2014.32.
  38. Sun W, Liu DB, Li WW, et al. Interleukin-6 promotes the migration and invasion of nasopharyngeal carcinoma cell lines and upregulates the expression of MMP-2 and MMP-9. Int J Oncol. 2014;44(5):1551–60. doi: 10.3892/ijo.2014.2323.
  39. Tzellos S, Farrell PJ. Epstein-Barr virus sequence variation-biology and disease. Pathogens. 2012;1(2):156–74. doi: 10.3390/pathogens1020156.
  40. Walling DM, Shebib N, Weaver SC, et al. The molecular epidemiology and evolution of Epstein-Barr virus: sequence variation and genetic recombination in the latent membrane protein-1 gene. J Infect Dis. 1999;179(4):763–74. doi: 10.1086/314672.
  41. Hu LF, Zabarovsky ER, Chen F, et al. Isolation and sequencing of the Epstein-Barr virus BNLF-1 gene (LMP1) from a Chinese nasopharyngeal carcinoma. J Gen Virol. 1991;72(Pt 10):2399–409. doi: 10.1099/0022-1317-72-10-2399.
  42. Nitta T, Chiba A, Yamamoto N, et al. Lack of cytotoxic property in a variant of Epstein-Barr virus latent membrane protein-1 isolated from nasopharyngeal carcinoma. Cell Signal. 2004;16(9):1071–81. doi: 10.1016/s0898-6568(04)00032-4.
  43. da Costa VG, Marques-Silva AC, Moreli ML. The Epstein-Barr virus latent membrane protein-1 (LMP1) 30-bp deletion and XhoI-polymorphism in nasopharyngeal carcinoma: a meta-analysis of observational studies. Syst Rev. 2015;4(1):46. doi: 10.1186/s13643-015-0037-z.
  44. Rowe M, Peng-Pilon M, Huen DS, et al. Upregulation of bcl-2 by the Epstein-Barr virus latent membrane protein LMP1: a B-cell-specific response that is delayed relative to NF-kappaB activation and to induction of cell surface markers. J Virol. 1994;68(9):5602–12.
  45. Trivedi P, Hu LF, Chen F, et al. Epstein-Barr virus (EBV)-encoded membrane protein LMP1 from a nasopharyngeal carcinoma is non-immunogenic in a murine model system, in contrast to a B cell-derived homologue. Eur J Cancer. 1994;30(1):84–8. doi: 10.1016/s0959-8049(05)80024-3.
  46. Knecht H, Bachmann E, Brousset P, et al. Deletions within the LMP1 oncogene of Epstein-Barr virus are clustered in Hodgkin’s disease and identical to those observed in nasopharyngeal carcinoma. Blood. 1993;82(10):2937–42.
  47. Miller WE, Edwards RH, Walling DM, et al. Sequence variation in the Epstein-Barr virus latent membrane protein 1. J Gen Virol. 1994;75(Pt 10):2729–40. doi: 10.1099/0022-1317-75-10-2729.
  48. Weiss LM. Epstein-Barr virus and Hodgkin’s disease. Curr Oncol Rep. 2000;2(2):199–204. doi: 10.1007/s11912-000-0094-9.
  49. Ковригина А.М., Пробатова Н.А. Лимфома Ходжкина и крупноклеточные лимфомы. М.: МИА, 2007.
    [Kovrigina AM, Probatova NA. Limfoma Khodzhkina i krupnokletochnye limfomy. (Hodgkin’s lymphomas and large cell lymphomas.) Moscow: MIA Publ.; 2007. (In Russ)]
  50. Клиническая онкогематология: Руководство для врачей, 2-е изд. Под ред. М.А. Волковой. М.: Медицина, 2007.
    [Volkova MA, ed. Klinicheskaya  onkogematologiya: Rukovodstvo dlya vrachei. (Clinical oncohematology: manual for physicians.) 2nd edition. Moscow: Meditsina Publ.; 2007. (In Russ)]
  51. Dorsett Y, Robbiani DF, Jankovic M, et al. A role for AID in chromosome translocations between c-myc and the IgH variable region. J Exp Med. 2007;204(9):2225–32. doi: 10.1084/jem.20070884.
  52. Stein H. Hodgkin lymphoma – introduction. In: Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008. pp. 321–34.
  53. Diehl V, Stein H, Hummel M, et al. Hodgkin’s lymphoma: biology and treatment strategies for primary, refractory, and relapsed disease. Hematology Am Soc Hematol Educ Program. 2003;1:225–47. doi: 10.1182/asheducation-2003.1.225.
  54. Chapman AL, Rickinson AB. Epstein-Barr virus in Hodgkin’s disease. Ann Oncol. 1998;9(Suppl 5):S5–16. doi: 10.1093/annonc/9.suppl_5.s5.
  55. Deacon EM, Pallesen G, Niedobitek G, et al. Epstein-Barr virus and Hodgkin’s disease: transcriptional analysis of virus latency in the malignant cells. J Exp Med. 1993;177(2):339–49. doi: 10.1084/jem.177.2.339.
  56. Kuppers R, Rajewsky K, Zhao M, et al. Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proc Natl Acad Sci USA. 1994;91(23):10962–6. doi: 10.1073/pnas.91.23.10962.
  57. Thomas RK, Re D, Wolf J, et al. Part I: Hodgkin’s lymphoma—molecular biology of Hodgkin and Reed-Sternberg cells. Lancet Oncol. 2004;5(1):11–8. doi: 10.1016/S1470-2045(03)01319-6.
  58. Cartwright RA, Watkins G. Epidemiology of Hodgkin’s disease: a review. Hematol Oncol. 2004;22(1):11–26. doi: 10.1002/hon.723.
  59. Jarrett AF, Armstrong AA, Alexander E. Epidemiology of EBV and Hodgkin’s lymphoma. Ann Oncol. 1996;7(Suppl 4):5–10. doi: 10.1093/annonc/7.suppl_4.s5.
  60. Glaser SL, Lin RJ, Stewart SL, et al. Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70(4):375–82. doi: 10.1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.0.co;2-t.
  61. Cader FZ, Kearns P, Young L, et al. The contribution of the Epstein-Barr virus to the pathogenesis of childhood lymphomas. Cancer Treat Rev. 2010;36(4):348–53. doi: 10.1016/j.ctrv.2010.02.011.
  62. Jarrett RF, Gallagher A, Jones DB, et al. Detection of Epstein-Barr virus genomes in Hodgkin’s disease: relation to age. J Clin Pathol. 1991;44(10):844–8. doi: 10.1136/jcp.44.10.844.
  63. Armstrong AA, Alexander FE, Cartwright R, et al. Epstein-Barr virus and Hodgkin’s disease: further evidence for the three disease hypothesis. Leukemia. 1998;12(8):1272–6. doi: 10.1038/sj.leu.2401097.
  64. Oyama T, Ichimura K, Suzuki R, et al. Senile EBV+ B-cell lymphoproliferative disorders: a clinicopathologic study of 22 patients. Am J Surg Pathol. 2003;27(1):16–26. doi: 10.1097/00000478-200301000-00003.
  65. Oyama T, Yamamoto K, Asano N, et al. Age-related EBV-associated B-cell lymphoproliferative disorders constitute a distinct clinicopathologic group: a study of 96 patients. Clin Cancer Res. 2007;13(17):5124–32. doi: 10.1158/1078-0432.ccr-06-2823.
  66. Thorley-Lawson DA, Gross A. Persistence of the Epstein-Barr virus and the origins of associated lymphomas. N Engl J Med. 2004;350(13):1328–37. doi: 10.1056/NEJMra032015.
  67. Kuppers R. Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer. 2005;5(4):251–62. doi: 10.1038/nrc1589.
  68. Klein U, Dalla-Favera R. Germinal centres: role in B-cell physiology and malignancy. Nat Rev Immunol. 2008;8(1):22–33. doi: 10.1038/nri2217.
  69. Caldwell RG, Wilson JB, Anderson SJ, et al. Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals. Immunity. 1998;9(3):405–11. doi: 10.1016/s1074-7613(00)80623-8.
  70. Gires O, Zimber-Strobl U, Gonnella R, et al. Latent membrane protein 1 of Epstein-Barr virus mimics a constitutively active receptor molecule. EMBO J. 1997;16(20):6131–40. doi: 10.1093/emboj/18.11.3064.
  71. Laichalk LL, Thorley-Lawson DA. Terminal differentiation into plasma cells initiates the replicative cycle of Epstein-Barr virus in vivo. J Virol. 2005;79(2):1296–307. doi: 10.1128/JVI.79.2.1296-1307.2005.
  72. Alexander FE, Jarrett RF, Lawrence D, et al. Risk factors for Hodgkin’s disease by Epstein-Barr virus (EBV) status: prior infection by EBV and other agents. Br J Cancer. 2000;82(5):1117–21. doi: 10.1054/bjoc.1999.1049.
  73. Mueller N, Evans A, Harris NL, et al. Hodgkin’s disease and Epstein-Barr virus. Altered antibody pattern before diagnosis. N Engl J Med. 1989;320(11):689–95. doi: 10.1056/nejm198903163201103.
  74. Weiss LM, Strickler JG, Warnke RA, et al. Epstein-Barr viral DNA in tissues of Hodgkin’s disease. Am J Pathol. 1987;129(1):86–91.
  75. Anagnostopoulos I, Herbst H, Niedobitek G, et al. Demonstration of monoclonal EBV genomes in Hodgkin’s disease and Ki-1-positive anaplastic large cell lymphoma by combined Southern blot and in situ hybridization. Blood. 1989;74(2):810–6.
  76. Re D, Kuppers R, Diehl V. Molecular pathogenesis of Hodgkin’s lymphoma. J Clin Oncol. 2005;23(26):6379–86. doi: 10.1200/JCO.2005.55.013.
  77. Mancao C, Altmann M, Jungnickel B, et al. Rescue of “crippled” germinal center B cells from apoptosis by Epstein-Barr virus. Blood. 2005;106(13):4339–44. doi: 10.1182/blood-2005-06-2341.
  78. Chaganti S, Bell AI, Pastor NB, et al. Epstein-Barr virus infection in vitro can rescue germinal center B cells with inactivated immunoglobulin genes. Blood. 2005;106(13):4249–52. doi: 10.1182/blood-2005-06-2327.
  79. Kapatai G, Murray P. Contribution of the Epstein Barr virus to the molecular pathogenesis of Hodgkin lymphoma. J Clin Pathol. 2007;60(12):1342–9. doi: 10.1136/jcp.2007.050146.
  80. Kuppers R. B cells under influence: transformation of B cells by Epstein-Barr virus. Nat Rev Immunol. 2003;3(10):801–12. doi: 10.1038/nri1201.
  81. Huen DS, Henderson SA, Croom-Carter D, et al. The Epstein-Barr virus latent membrane protein-1 (LMP1) mediates activation of NF-kappa B and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain. Oncogene. 1995;10:549–60.
  82. Kieser A, Kilger E, Gires O, et al. Epstein-Barr virus latent membrane protein-1 triggers AP-1 activity via the c-Jun N-terminal kinase cascade. EMBO J. 1997;16(21):6478–85. doi: 10.1093/emboj/16.21.6478.
  83. Kube D, Holtick U, Vockerodt M, et al. STAT3 is constitutively activated in Hodgkin cell lines. Blood. 2001;98(3):762–70. doi: 10.1182/blood.V98.3.762.
  84. Dutton A, Reynolds GM, Dawson CW, et al Constitutive activation of phosphatidyl-inositide 3 kinase contributes to the survival of Hodgkin’s lymphoma cells through a mechanism involving Akt kinase and mTOR. J Pathol. 2005;205(4):498–506. doi: 10.1002/path.1725.
  85. Brielmeier M, Mautner J, Laux G, et al. The latent membrane protein 2 gene of Epstein-Barr virus is important for efficient B cell immortalization. J Gen Virol. 1996;77(Pt 11):2807–18. doi: 10.1099/0022-1317-77-11-2807.
  86. Casola S, Otipoby KL, Alimzhanov M, et al. B cell receptor signal strength determines B cell fate. Nat Immunol. 2004;5(3):317–27. doi: 10.1038/ni1036.
  87. Engels N, Yigit G, Emmerich CH, et al. Epstein-Barr virus LMP2A signaling in statu nascendi mimics a B cell antigen receptor-like activation signal. Cell Commun Signal. 2012;10(1):9. doi: 10.1186/1478-811X-10-9.
  88. Portis T, Dyck P, Longnecker R. Epstein-Barr Virus (EBV) LMP2A induces alterations in gene transcription similar to those observed in Reed-Sternberg cells of Hodgkin lymphoma. Blood. 2003;102(12):4166–78. doi: 10.1182/blood-2003-04-1018.
  89. Portis T, Longnecker R. Epstein-Barr virus (EBV) LMP2A mediates B-lymphocyte survival through constitutive activation of the Ras/PI3K/Akt pathway. Oncogene. 2004;23(53):8619–28. doi: 10.1038/sj.onc.1207905.
  90. Farrell K, Jarrett RF. The molecular pathogenesis of Hodgkin lymphoma. Histopathology. 2011;58(1):15–25. doi: 10.1111/j.1365-2559.2010.03705.x.
  91. Herbst H, Foss HD, Samol J, et al. Frequent expression of interleukin-10 by Epstein-Barr virus-harboring tumor cells of Hodgkin’s disease. Blood. 1996;87:2918–29.
  92. Hsu SM, Lin J, Xie SS, et al. Abundant expression of transforming growth factor-beta 1 and -beta 2 by Hodgkin’s Reed-Sternberg cells and by reactive T lymphocytes in Hodgkin’s disease. Hum Pathol. 1993;24(3):249–55. doi: 10.1016/0046-8177(93)90034-e.
  93. Kapp U, Yeh WC, Patterson B, et al. Interleukin 13 is secreted by and stimulates the growth of Hodgkin and Reed-Sternberg cells. J Exp Med. 1999;189(12):1939–46. doi: 10.1084/jem.189.12.1939.
  94. Munz C, Moormann A. Immune escape by Epstein-Barr virus associated malignancies. Semin Cancer Biol. 2008;18(6):381–7. doi: 10.1016/j.semcancer.2008.10.002.
  95. Lichtenstein AV, Melkonyan HS, Tomei LD, et al. Circulating nucleic acids and apoptosis. Ann NY Acad Sci. 2001;945(1):239–49. doi: 10.1111/j.1749-6632.2001.tb03892.x.
  96. Sidransky D. Emerging molecular markers of cancer. Nat Rev Cancer. 2002;2(3):210–9. doi: 10.1038/nrc755.
  97. Skvortsova TE, Rykova EY, Tamkovich SN, et al. Cell-free and cell-bound circulating DNA in breast tumours: DNA quantification and analysis of tumour-related gene methylation. Br J Cancer. 2006;94(10):1492–5. doi: 10.1038/sj.bjc.6603117.
  98. Lo YM, Chan LY, Lo KW, et al. Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. Cancer Res. 1999;59(6):1188–91.
  99. Hou X, Zhao C, Guo Y, et al. Different Clinical Significance of Pre- and Post-treatment Plasma Epstein-Barr Virus DNA Load in Nasopharyngeal Carcinoma Treated with Radiotherapy. Clin Oncol. (R Coll Radiol) 2011;23(2):128–33. doi: 10.1016/j.clon.2010.09.001.
  100. Wang WY, Twu CW, Chen HH, et al. Plasma EBV DNA clearance rate as a novel prognostic marker for metastatic/recurrent nasopharyngeal carcinoma. Clin Cancer Res. 2010;16(3):1016–24. doi: 10.1158/1078-0432.ccr-09-2796.
  101. Lo YM, Chan AT, Chan LY, et al. Molecular prognostication of nasopharyngeal carcinoma by quantitative analysis of circulating Epstein-Barr virus DNA. Cancer Res. 2000;60:6878–81.
  102. Lo YM, Chan LY, Chan AT, et al. Quantitative and temporal correlation between circulating cell-free Epstein-Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res. 1999;59:5452–5.
  103. Au WY, Pang A, Choy C, et al. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104(1):243–9. doi: 10.1182/blood-2003-12-4197.
  104. Wang ZY, Liu QF, Wang H, et al. Clinical implications of plasma Epstein-Barr virus DNA in early-stage extranodal nasal-type NK/T-cell lymphoma patients receiving primary radiotherapy. Blood. 2012;120(10):2003–10. doi: 10.1182/blood-2012-06-435024.
  105. Kasamon YL, Jacene HA, Gocke CD, et al. Phase 2 study of rituximab-ABVD in classical Hodgkin lymphoma. Blood. 2012;119(18):4129–32. doi: 10.1182/blood-2012-01-402792.
  106. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121(18):3547–53. doi: 10.1182/blood-2012-09-454694.
  107. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17(9):2885–92. doi: 10.1158/1078-0432.ccr-10-3327.
  108. Dinand V, Sachdeva A, Datta S, et al. Plasma Epstein Barr Virus (EBV) DNA as a Biomarker for EBV associated Hodgkin lymphoma. Indian Pediatr. 2015;52(8):681–5. doi: 10.1007/s13312-015-0696-9.
  109. Vockerodt М, Yap L-F, Shannon-Lowe C, et al. The Epstein-Barr virus and the pathogenesis of lymphoma. J Pathol. 2015;235(2):312–22. doi: 10.1002/path.4459.
  110. Grywalska E, Markowicz J, Grabarczyk P, et al. Epstein-Barr virus-associated lymphoproliferative disorders. Postepy Hig Med Dosw (Online). 2013;67:481–90. doi 10.5604/17322693.1050999.

Вторичный гемофагоцитарный синдром в клинической практике гематолога: обзор литературы и собственные данные

Потапенко В.Г.1,2,  Потихонова Н.А.4, Байков В.В. 2, Белогурова М.Б. 1, Лисуков И.А.3, Климович А.В. 1,Лапин  С.В. 2, Иванова М.О. 2,  Кравцова В.М.2,  Подольцева Э.И.1, Медведева Н.В. 1,  Афанасьев Б.В.2

1 СПб ГБУЗ «Городская клиническая больница № 31», пр-т Динамо, д. 3, Санкт-Петербург, Российская Федерация, 197110

2 НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачевой, Первый Санкт-Петербургский государственный медицинский университет им. акад. И.П. Павлова, ул. Рентгена, д. 12, Санкт-Петербург, Российская Федерация, 197022

3 Северо-Западный государственный медицинский университет им. И.И. Мечникова, ул. Кирочная, д. 41, Санкт-Петербург, Российская Федерация, 191015

4 ФГБУ «Российский научно-исследовательский институт гематологии и трансфузиологии Федерального медико-биологического агентства», ул. 2-я Советская, д. 16, Санкт-Петербург, Российская Федерация, 191024

Для переписки: Всеволод Геннадьевич Потапенко, СПб ГБУЗ «Городская клиническая больница № 31», пр. Динамо, д. 3, Санкт-Петербург, Российская Федерация, 197110; тел.: +7(812)230-19-33; e-mail: potapenko.vsevolod@mail.ru

Для цитирования: Потапенко В.Г., Потихонова Н.А., Байков В.В. и др. Вторичный гемофагоцитарный синдром у взрослых в клинической практике гематолога: обзор литературы и собственные данные. Клиническая онкогематология. 2015;8(2):169–84.


РЕФЕРАТ

Актуальность и цели. Гемофагоцитарный синдром представляет собой опасный гипервоспалительный синдром, причиной которого наиболее часто служит инфекция. Он является следствием избыточной активации клеток системы фагоцитирующих мононуклеаров, что проявляется цитопенией, системной воспалительной реакцией, повреждением печени, селезенки. В связи с редкостью заболевания, сложностью диагностики этот синдром является малоизученным и часто остается нераспознанным. Цель работы — описательный анализ собственных наблюдений вторичного гемофагоцитарного синдрома (ГФС) и представление литературного обзора.

Методы. Анализу подвергнуты клинические и лабораторные данные 15 пациентов в возрасте 16–64 года (медиана 48 лет) со вторичным ГФС, которые наблюдались с 2009 по 2013 г. Вторичные ГФС были диагностированы у больных злокачественными лимфопролиферативными и инфекционными заболеваниями. Признаки ГФС выявлены при злокачественных лимфопролиферативных заболеваниях (n = 5), хронической активной инфекции, вызванной вирусом Эпштейна—Барр (ВЭБ) (n = 3), аллогенной трансплантации стволовых клеток крови (n = 3), остром лейкозе (n = 1), множественной миеломе (n = 1), пневмонии (n = 1), гломерулонефрите (n = 1). Терапия ГФС проводилась 8 пациентам: этопозид (n = 1), глюкокортикоиды (n = 1), внутривенный иммуноглобулин (n = 2), комбинация ритуксимаба и глюкокортикоидов (n = 2), этопозида и циклоспорина А (n = 1), а также комбинированная химиотерапия по программе HLH-2004 (n = 1). Медиана наблюдения за больными составила 42 мес.

Результаты. У включенных в ретроспективный анализ 15 взрослых пациентов самыми частыми фоновыми заболеваниями при вторичном ГФС были злокачественные лимфопролиферативные заболевания и хроническая ВЭБ-инфекция. Ранняя диагностика сложна, т. к. принятые в настоящее время критерии болезни характерны для ГФС поздней стадии. Все это диктует необходимость разработки более чувствительных и универсальных диагностических критериев.

Заключение. В онкогематологической клинике вторичный ГФС является тяжелым осложнением, требующим проведения дифференциальной диагностики с другими критическими состояниями и интенсивной терапии. При ГФС, связанным с онкогематологическими заболеваниями, пациенты нуждаются в тщательном наблюдении в процессе и после окончании противоопухолевого лечения.


Ключевые слова: вторичный гемофагоцитарный синдром, лимфома, вирус Эпштейна—Барр, этопозид, трансплантация стволовых клеток крови.

Получено: 9 декабря 2014 г.

Принято в печать: 7 февраля 2015 г.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Carroll WL, Finlay JL, Sudbury MA. Cancer in children and adolescents. Jones & Bartlett; 2010. pp. 254–6.
  2. Chu T, D’Angio GJ, Favara B, et al. Histiocytosis syndromes in children. The Lancet. 1987;329(8526):208–9. doi: 10.1016/s0140-6736(87)90016-x.
  3. Favara BE, Feller AC, Pauli M, et al. Contemporary classification of histiocytic disorders. The WHO Committee On Histiocytic/Reticulum Cell Proliferations. Reclassification Working Group of the Histiocyte Society. Med Pediatr Oncol. 1997;29(3):157–66. doi: 10.1002/(sici)1096-911x(199709)29:3<157::aid-mpo1>3.0.co;2-c.
  4. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  5. Jordan MB, Allen CE, Weitzman S, et al. How I treat hemophagocytic lymphohistiocytosis. Blood. 2011;118(15):4041–52. doi: 10.1182/blood-2011-03-278127.
  6. Janka GE. Hemophagocytic syndromes. Blood Rev. 2007;21(5):245–53. doi: 10.1016/j.blre.2007.05.001.
  7. Gotze KS, Hoffmann D, Schatzl HM, et al. Fatal Epstein-Barr virus-associated lymphoproliferative disorder following treatment with a novel mTOR inhibitor for relapsed chronic lymphocytic leukemia leukemia cells. Haematologica. 2007;92(9):1282–3. doi: 10.3324/haematol.11155.
  8. Emmenegger U, Schaer DJ, Larroche C, et al. Haemophagocytic syndromes in adults: current concepts and challenges ahead. Swiss Med Wkly. 2005;135(21-22):299–314.
  9. Arico M, Danesino C, Pende D, Moretta L. Pathogenesis of hemophagocytic lymphohistiocytosis. Br J Haematol. 2001;114(4):761–9. doi: 10.1046/j.1365-2141.2001.02936.x.
  10. Henter JI, Horne A, Arico M, et al. HLH-2004: diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2007;48(2):124–31. doi: 10.1002/pbc.21039.
  11. Filipovich AH. Hemophagocytic lymphohistiocytosis (HLH) and related disorders. Hematology. 2009;2009(1):127–31. doi: 10.1182/asheducation-2009.1.127.
  12. Trottestam H, Horne A, Arico M, et al. Chemoimmunotherapy for hemophagocytic lymphohistiocytosis: long-term results of the HLH-94 treatment protocol. Blood. 2011;118(17):4577–84. doi: 10.1182/blood-2011-06-356261.
  13. Охотникова Е.Н., Меллина К.В., Усова Е.И. и др. Гемофагоцитарный синдром в педиатрической практике. Клиническая иммунология, аллергология, инфектология. 2008;2(13):61–70.
    [Okhotnikova EN, Mellina KV, Usova EI, et al. Hematophagocytic syndrome in pediatric practice. Klinicheskaya immunologiya, allergologiya, infektologiya. 2008;2(13):61–70. (In Russ)]
  14. Diaz-Guzman E, Dong B, Hobbs SB, et al. Hemophagocytic lymphohistiocytosis after lung transplant: report of 2 cases and a literature review. Exp Clin Transplant. 2011;9(3):217–22.
  15. Охотникова Е.Н., Меллина К.В., Усова Е.И. и др. Гемофагоцитарный синдром в педиатрической практике (Обзор литературы). Здоровье ребенка. 2008;4(13):131–8.
    [Okhotnikova EN, Mellina KV, Usova EI, et al. Hematophagocytic syndrome in pediatric practice (literature review). Zdorov’e rebenka. 2008;4(13):131–8. (In Russ)]
  16. Karapinar B, Yilmaz D, Balkan C, et al. An unusual cause of multiple organ dysfunction syndrome in the pediatric intensive care unit: hemophagocytic lymphohistiocytosis. Pediatr Crit Care Med. 2009;10(3):285–90. doi: 10.1097/pcc.0b013e318198868b.
  17. Schaer DJ, Schaer CA, Schoedon G, et al. Hemophagocytic macrophages constitute a major compartment of heme oxygenase expression in sepsis. Eur J Haematol. 2006;77(5):432–6. doi: 10.1111/j.1600-0609.2006.00730.x.
  18. Besset S, Schnell D, Azoulay E. Hemophagocytic lymphohistiocytosis mimicking septic shock. Chest. 2012;141(3):835; author reply 836. doi: 10.1378/chest.11-2717.
  19. Raschke RA, Garcia-Orr R. Hemophagocytic lymphohistiocytosis: a potentially underrecognized association with systemic inflammatory response syndrome, severe sepsis, and septic shock in adults. Chest. 2011;140(4):933–8. doi: 10.1378/chest.11-0619.
  20. Gupta A, Tyrrell P, Valani R, et al. The role of the initial bone marrow aspirate in the diagnosis of hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2008;51(3):402–4. doi: 10.1002/pbc.21564.
  21. Wang Z, Chen X, Wu L, et al. Significance of hemophagocytosis in diagnosis of hemophagocytic lymphohistiocytosis. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2009;17(4):1064–6.
  22. Favara BE. Histopathology of the liver in histiocytosis syndromes. Pediatr Pathol Lab Med. 1996;16(3):413–33. doi: 10.3109/15513819609168681.
  23. Wang Z, Wang YN, Feng CC, et al. Diagnostic significance of NK cell activity and soluble CD25 level in serum from patients with secondary hemophagocytic lymphohistiocytosis. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2008;16(5):1154–7.
  24. Wang LL, Hu YX, Chen WF, et al. Significance of soluble interleukin-2 receptor and NK cell activity in patients with hemophagocytic lymphohistiocytosis. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2012;20(2):401–4.
  25. Wang Z, Wang YN, Feng CC, et al. The early diagnosis and clinical analysis of 57 cases of acquired hemophagocytic lymphohistiocytosis. Zhonghua Nei Ke Za Zhi. 2009;48(4):312–5.
  26. Gotoh Y, Okamoto Y, Uemura O, et al. Determination of age-related changes in human soluble interleukin 2 receptor in body fluids of normal subjects as a control value against disease states. Clin Chim Acta. 1999;289(1–2):89–97. doi: 10.1016/s0009-8981(99)00161-8.
  27. Rothkrantz-Kos S, Drent M, Schmitz MP, et al. Biochemical parameters in monitoring severity of sarcoidosis. Chapter 4: Analytical evaluation and determination of reference values of soluble interleukin-2-receptor and serum amyloid-A. 2004.
  28. Janka G. Hemophagocytic lymphohistiocytosis: when the immune system runs amok. Klin Padiatr. 2009;221(5):278–85. doi: 10.1055/s-0029-1237386.
  29. Crook MA. Hyperferritinaemia; laboratory implications. Ann Clin Biochem. 2012;49(Pt 3):211–3. doi: 10.1258/acb.2012.012059.
  30. Park HS, Kim DY, Lee JH, et al. Clinical features of adult patients with secondary hemophagocytic lymphohistiocytosis from causes other than lymphoma: an analysis of treatment outcome and prognostic factors. Ann Hematol. 2012;91(6):897–904. doi: 10.1007/s00277-011-1380-3.
  31. Dhote R, Simon J, Papo T, et al. Reactive hemophagocytic syndrome in adult systemic disease: report of twenty-six cases and literature review. Arthritis Rheum. 2003;49(5):633–9. doi: 10.1002/art.11368.
  32. Mayordomo-Colunga J, Rey C, Gonzalez S, Concha A. Multiorgan failure due to hemophagocytic syndrome: A case report. Cases J. 2008;1(1):209. doi: 10.1186/1757-1626-1-209.
  33. Karras A, Thervet E, Legendre C. Hemophagocytic syndrome in renal transplant recipients: report of 17 cases and review of literature. Transplantation. 2004;77(2):238–43. doi: 10.1097/01.tp.0000107285.86939.37.
  34. Han AR, Lee HR, Park BB, et al. Lymphoma-associated hemophagocytic syndrome: clinical features and treatment outcome. Ann Hematol. 2007;86(7):493–8. doi: 10.1007/s00277-007-0278-6.
  35. Wijsman CA, Roeters van Lennep JE, von dem Borne PA, Fogteloo AJ. A diagnostic difficulty: two cases of haemophagocytic syndrome in adults. Neth J Med. 2009;67(1):29–31.
  36. Machaczka M. Hemophagocytic lymphohistiocytosis–a contemporary medical problem. Pol Merkur Lekarski. 2012;32(187):59–63.
  37. Allen CE, Yu X, Kozinetz CA, McClain KL. Highly elevated ferritin levels and the diagnosis of hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2008;50(6):1227–35. doi: 10.1002/pbc.21423.
  38. Henter JI, Samuelsson-Horne A, Arico M, et al. Treatment of hemophagocytic lymphohistiocytosis with HLH-94 immunochemotherapy and bone marrow transplantation. Blood. 2002;100(7):2367–73. doi: 10.1182/blood-2002-01-0172.
  39. Shin HJ, Chung JS, Lee JJ, et al. Treatment Outcomes with CHOP Chemotherapy in Adult Patients with Hemophagocytic Lymphohistiocytosis. J Korean Med Sci. 2008;23(3):439–44. doi: 10.3346/jkms.2008.23.3.439.
  40. Goede JS, Peghini PE, Fehr J. Oral Low Dose Etoposide in the Treatment of Macrophage Activation Syndrome. Blood (ASH Annual Meeting Abstracts). 2004;104:3817.
  41. Bhattacharyya M, Ghosh MK. Hemophagoctic lymphohistiocytosis–recent concept. J Assoc Physicians India. 2008;56:453–7.
  42. Imashuku S, Hibi S, Kuriyama K, et al. Management of severe neutropenia with cyclosporin during initial treatment of Epstein-Barr virus-related hemophagocytic lymphohistiocytosis. Leuk Lymphoma. 2000;36(3–4):339–46. doi: 10.3109/10428190009148855.
  43. Ishii E, Ohga S, Imashuku S, et al. Review of hemophagocytic lymphohistiocytosis (HLH) in children with focus on Japanese experiences. Crit Rev Oncol Hematol. 2005;53(3):209–23. doi: 10.1016/j.critrevonc.2004.11.002.
  44. Emmenegger U, Reimers A, Frey U, et al. Reactive macrophage activation syndrome: a simple screening strategy and its potential in early treatment initiation. Swiss Med Wkly. 2002;132(17–18):230–6.
  45. Imashuku S, Kuriyama K, Teramura T, et al. Requirement for etoposide in the treatment of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. J Clin Oncol. 2001;19(10):2665–73.
  46. Imashuku S, Kuriyama K, Sakai R, et al. Treatment of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis (EBV-HLH) in young adults: A report from the HLH study center. Med Pediatr Oncol. 2003;41(2):103–9. doi: 10.1002/mpo.10314.
  47. Bosman G, Langemeijer SM, Hebeda KM, et al. The role of rituximab in a case of EBV–related lymphoproliferative disease presenting with haemophagocytosis. Neth J Med. 2009;67(8):364–5.
  48. Kimura H. Pathogenesis of chronic active Epstein-Barr virus infection: is this an infectious disease, lymphoproliferative disorder, or immunodeficiency? Rev Med Virol. 2006;16(4):251–61. doi: 10.1002/rmv.505.
  49. Imashuku S, Teramura T, Tauchi H, et al. Longitudinal follow-up of patients with Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. Haematologica. 2004;89(2):183–8.
  50. Balamuth NJ, Nichols KE, Paessler M, Teachey DT. Use of rituximab in conjunction with immunosuppressive chemotherapy as a novel therapy for Epstein Barr virus-associated hemophagocytic lymphohistiocytosis. J Pediatr Hematol Oncol. 2007;29(8):569–73. doi: 10.1097/mph.0b013e3180f61be3.
  51. Bosman G, Langemeijer SM, Hebeda KM, et al. The role of rituximab in a case of EBV-related lymphoproliferative disease presenting with haemophagocytosis. Neth J Med. 2009;67(8):364–5.
  52. So MW, Koo BS, Kim YJ, et al. Successful rituximab treatment of refractory hemophagocytic lymphohistiocytosis and autoimmune hemolytic anemia associated with systemic lupus erythematosus. Mod Rheumatol. 2013 Feb 7 (abstract). doi: 10.1007/s10165-013-0838-7.
  53. Stebbing J, Ngan S, Ibrahim H, et al. The successful treatment of haemophagocytic syndrome in patients with humanimmunodeficiency virus-associated multi-centric Castleman’s disease. Clin Exp Immunol. 2008;154(3):399–405. doi: 10.1111/j.1365-2249.2008.03786.x.
  54. Масчан М. Молекулярно-генетическая диагностика и дифференциальная терапия гистиоцитарных пролиферативных заболеваний у детей: Автореф. ¼ д-ра мед. наук. М., 2011.
    [Maschan M. Molekulyarno-geneticheskaya diagnostika i differentsial’naya terapiya gistiotsitarnykh proliferativnykh zabolevanii u detei. (Molecular genetic diagnosis and differentiated therapy of histiocytic proliferative diseases in children.) [dissertation] Moscow; 2011. (In Russ)]
  55. Filipovich A, McClain K, Grom A. Histiocytic disorders: recent insights into pathophysiology and practical guidelines. Biol Blood Marrow Transplant. 2010;16(1 Suppl):S82–9. doi: 10.1016/j.bbmt.2009.11.014.
  56. Shabbir M, Lucas J, Lazarchick J, Shirai K. Secondary hemophagocytic syndrome in adults: a case series of 18 patients in a single institution and a review of literature. Hematol Oncol. 2011;29(2):100–6. doi: 10.1002/hon.960.
  57. Ramanan AV, Schneider R. Macrophage activation syndrome–what’s in a name! Rheumatol. 2003;30(12):2513–6.
  58. Weitzman S. Approach to hemophagocytic syndromes. Hematology Am Soc Hematol Educ Program. 2011;2011(1):178–83.
  59. Sada E, Shiratsuchi M, Kiyasu J, et al. Primary mediastinal non-seminomatous germ cell tumor associated with hemophagocytic syndrome. J Clin Exp Hematol. 2009;49(2):117–20. doi: 10.3960/jslrt.49.117.
  60. Chaudary IU, Bojal SA, Attia A, et al. Mediastinal endodermal sinus tumor associated with fatal hemophagocytic syndrome. Hematol Oncol Stem Cell Ther. 2011;4(3):138–41. doi: 10.5144/1658-3876.2011.138.
  61. Kounami S, Nakayama K, Yoshiyama M, et al. Early-onset hemophagocytic lymphohistiocytosis after the start of chemotherapy for advanced neuroblastoma. Pediatr Hematol Oncol. 2012;29(1):99–103. doi: 10.3109/08880018.2011.643529.
  62. Karapinar B, Yilmaz D, Balkan C, et al. An unusual cause of multiple organ dysfunction syndrome in the pediatric intensive care unit: hemophagocytic lymphohistiocytosis. Pediatr Crit Care Med. 2009;10(3):285–90. doi: 10.1097/pcc.0b013e318198868b.
  63. Takahashi N. Lymphoma-associated hemophagocytic syndrome (LAHS). Nihon Rinsho. 2000;58(3):665–8 (abstract).
  64. Chang CS, Wang CH, Su IJ, et al. Hematophagic histiocytosis: a clinicopathologic analysis of 23 cases with special reference to the association with peripheral T-cell lymphoma. J Formos Med Assoc. 1994;93:421–8.
  65. Miyahara M, Sano M, Shibata K, et al. B-cell lymphoma-associated hemophagocytic syndrome: clinicopathological characteristics. Ann Hematol. 2000;79(7):378–88. doi: 10.1007/s002770000155.
  66. Takahashi N, Miura I, Chubachi A, et al. A clinicopathological study of 20 patients with T/natural killer (NK)-cell lymphoma-associated hemophagocytic syndrome with special reference to nasal and nasal-type NK/T-cell lymphoma. Int J Hematol. 2001;74(3):303–8. doi: 10.1007/bf02982065.
  67. Abe Y, Hara K, Shiratsuchi M, et al. Two cases of B cell lymphoma associated with hemophagocytic syndrome. Rinsho Ketsueki. 2001;42(1):35–40.
  68. Shimazaki C, Inaba T, Okano A, et al. Clinical characteristics of B-cell lymphoma-associated hemophagocytic syndrome (B-LAHS): comparison of CD5+ with CD5- B-LAHS. Intern Med. 2001;40(9):878–82. doi: 10.2169/internalmedicine.40.878.
  69. Janka G, Imashuku S, Elinder G, et al. Infection- and malignancy-associated hemophagocytic syndromes. Secondary hemophagocytic lymphohistiocytosis. Hematol Oncol Clin North Am. 1998;12(2):435–44. doi: 10.1016/s0889-8588(05)70521-9.
  70. Matzner Y, Behar A, Beeri E, et al. Systemic leishmaniasis mimicking malignant histiocytosis. Cancer. 1979;43(1):398–402. doi: 10.1002/1097-0142(197901)43:1<398::aid-cncr2820430156>3.0.co;2-3.
  71. Castillo L, Carcillo J. Secondary hemophagocytic lymphohistiocytosis and severe sepsis/systemic inflammatory response syndrome/multiorgan dysfunction syndrome/macrophage activation syndrome share common intermediate phenotypes on a spectrum of inflammation. Pediatr Crit Care Med. 2009;10(3):387–92. doi: 10.1097/pcc.0b013e3181a1ae08.
  72. Karapinar B, Yilmaz D, Balkan C, et al. An unusual cause of multiple organ dysfunction syndrome in the pediatric intensive care unit: hemophagocytic lymphohistiocytosis. Pediatr Crit Care Med. 2009;10(3):285–90. doi: 10.1097/pcc.0b013e318198868b.
  73. Buyse S, Teixeira L, Galicier L, et al. Critical care management of patients with hemophagocytic lymphohistiocytosis. Intens Care Med. 2010;36(10):1695–702. doi: 10.1007/s00134-010-1936-z.
  74. Takahashi N, Chubachi A, Kume M, et al. A clinical analysis of 52 adult patients with hemophagocytic syndrome: the prognostic significance of the underlying diseases. Int J Hematol. 2001;74(2):209–13. doi: 10.1007/bf02982007.
  75. Cohen JI, Jaffe ES, Dale JK, et al. Characterization and treatment of chronic active Epstein-Barr virus disease: a 28-year experience in the United States. Blood. 2011;117(22):5835–49. doi: 10.1182/blood-2010-11-316745.
  76. Ishii E, Ohga S, Imashuku S, et al. Nationwide survey of hemophagocytic lymphohistiocytosis in Japan. Int J Hematol. 2007;86(1):58–65. doi: 10.1532/ijh97.07012.
  77. Xiao L, Xian Y, Dai BT, et al. Clinical features and outcome analysis of 83 childhood Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis with HLH-2004 protocol. Zhonghua Xue Ye Xue Za Zhi. 2011;32(10):668–72.
  78. Maia DM, Peace-Brewer AL. Chronic, active Epstein-Barr virus infection. Curr Opin Hematol. 2000;7(1):59–63. doi: 10.1097/00062752-200001000-00011.
  79. Kunitomi A, Kimura H, Ito Y, et al. Unrelated bone marrow transplantation induced long-term remission in a patient with life-threatening Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. J Clin Exp Hematol. 2011;51(1):57–61. doi: 10.3960/jslrt.51.57.
  80. Ohshima K, Suzumiya J, Sugihara M, et al. Clinicopathological study of severe chronic active Epstein-Barr virus infection that developed in association with lymphoproliferative disorder and/or hemophagocytic syndrome. Pathol Int. 1998;48(12):934–43. doi: 10.1111/j.1440-1827.1998.tb03864.x.
  81. Katano H, Ali MA, Patera AC, et al. Chronic active Epstein-Barr virus infection associated with mutations in perforin that impair its maturation. Blood. 2004;103(4):1244–52. doi: 10.1182/blood-2003-06-2171.
  82. Kasahara Y, Yachie A, Takei K, et al. Differential cellular targets of Epstein-Barr virus (EBV) infection between acute EBV-associated hemophagocytic lymphohistiocytosis and chronic active EBV infection. Blood. 2001;98(6):1882–8. doi: 10.1182/blood.v98.6.1882.
  83. Taniai N, Akimaru K, Kawano Y, et al. Hemophagocytic syndrome after living-donor liver transplantation for fulminant liver failure: a case report. Hepatogastroenterology. 2005;52(63):923–6.
  84. Yoshizumi T, Taketomi A, Kayashima H, et al. Successful treatment for a patient with hemophagocytic syndrome after a small-for-size graft liver transplantation. Hepatogastroenterology. 2008;55(82–83):359–62.
  85. Soyama A, Eguchi S, Takatsuki M, et al. Hemophagocytic syndrome after liver transplantation: report of two cases. Surg Today. 2011;41(11):1524–30. doi: 10.1007/s00595-010-4512-9.
  86. Fukunaga A, Nakamura F, Yoshinaga N, et al. Successful treatment with combined chemotherapy of two adult cases of hemophagocytic lymphohistiocytosis in recipients of umbilical cord blood cell transplantation. Int J Hematol. 2011;93(4):551–4. doi: 10.1007/s12185-011-0792-0.
  87. Asano T, Kogawa K, Morimoto A, et al. Hemophagocytic lymphohistiocytosis after hematopoietic stem cell transplantation in children: a nationwide survey in Japan. Pediatr Blood Cancer. 2012;59(1):110–4. doi: 10.1002/pbc.23384.
  88. Abdelkefi A, Ben Jamil W, Torjman L, et al. Hemophagocytic syndrome after hematopoietic stem cell transplantation: a prospective observational study. Int J Hematol. 2009;89(3):368–73. doi: 10.1007/s12185-009-0261-1.
  89. Okano M, Kawa K, Kimura H, et al. Proposed Guidelines for Diagnosing Chronic Active Epstein-Barr Virus Infection. Am J Hematol. 2005;80(1):64–9. doi: 10.1002/ajh.20398.
  90. Horne A, Trottestam H, Arico M, et al. Frequency and spectrum of central nervous system involvement in 193 children with haemophagocytic lymphohistiocytosis. Br J Haematol. 2008;140(3):327–35. doi: 10.1111/j.1365-2141.2007.06922.x.
  91. Gupta S, Weitzman S. Primary and secondary hemophagocytic lymphohistiocytosis: clinical features, pathogenesis and therapy. Exp Rev Clin Immunol. 2010;6(1):137–54. doi: 10.1586/eci.09.58.