Роль биохимических маркеров воспаления у пациентов с нейтропенией после химиотерапии

Ю.Н. Дубинина1, В.О. Саржевский2, В.Я. Мельниченко2

1 Клиника амбулаторной онкологии и гематологии, ул. Молодогвардейская, д. 2, корп. 1, Москва, Российская Федерация, 121467

2 ФГБУ «Национальный медико-хирургический центр им. Н.И. Пирогова», Минздрава России, ул. Нижняя Первомайская, д. 70, Москва, Российская Федерация, 105203

Для переписки: Юлия Николаевна Дубинина, ул. Молодогвардейская, д. 2, корп. 1, Москва, Российская Федерация, 121467; тел.: +7(499)112-25-04; e-mail: medicinemsc@gmail.com

Для цитирования: Дубинина Ю.Н., Саржевский В.О., Мельниченко В.Я. Роль биохимических маркеров воспаления у пациентов с нейтропенией после химиотерапии. Клиническая онкогематология. 2019;12(4):461–7.

DOI: 10.21320/2500-2139-2019-12-4-461-467


РЕФЕРАТ

Увеличение количества аутологичных и аллогенных трансплантаций костного мозга и гемопоэтических стволовых клеток, а также технологичность процесса ведут к появлению более токсичных курсов лекарственной противоопухолевой терапии, а следовательно, к развитию осложнений. Наиболее серьезными среди осложнений данного вида лечения являются инфекционные. Вероятность развития инфекций у пациентов с нейтропенией после химиотерапии достигает 90 %. В связи с этим растет необходимость поиска оптимального маркера инфекционных осложнений. В настоящем обзоре рассматриваются основные биохимические маркеры воспаления, приводится анализ исследований, позволяющих оценить диагностическую и прогностическую значимость С-реактивного белка, прокальцитонина и пресепсина.

Ключевые слова: сепсис, аутологичная трансплантация костного мозга, аллогенная трансплантация костного мозга, химиотерапия, инфекция, прокальцитонин, пресепсин, С-реактивный белок.

Получено: 7 мая 2019 г.

Принято в печать: 11 сентября 2019 г.

Читать статью в PDF


ЛИТЕРАТУРА

  1. Passweg JR, Baldomero H, Bade P, et al. Is the use of unrelated donor transplantation leveling off in Europe? The 2016 European Society for Blood and Marrow Transplant activity survey report. Bone Marrow Transplant. 2018;53(9):1139–48. doi: 10.1038/s41409-018-0153-1.

  2. Ochs L, Shu XO, Miller J, et al. Late infections after allogeneic bone marrow transplantation: comparison of incidence in related and unrelated donor transplant recipients. Blood. 1995;86(10):3979–86.

  3. Sorely JS, Shea TC. Prevention of infections in bone marrow transplant recipients. Infect Dis Clin North Am. 1997;11(2):459–77. doi: 10.1016/s0891-5520(05)70365-2.

  4. Massaro KSR, Costa SF, Leone C, Chamone DAF. Procalcitonin (PCT) and C-reactive Protein (CRP) as severe systemic infection markers in febrile neutropenic adults. BMC Infect Dis. 2007;7(1). doi: 10.1186/1471-2334-7-137.

  5. Саржевский В.О., Дубинина Ю.Н., Мельниченко В.Я. Диагностическое и прогностическое значение биохимических маркеров инфекционных осложнений высокодозной химиотерапии с аутологичной трансплантацией гемопоэтических стволовых клеток при злокачественных лимфопролиферативных заболеваниях. Клиническая онкогематология. 2017;10(1):113–9. doi: 10.21320/2500-2139-2017-10-1-113-119.

    [Sarzhevskii VO, Dubinina YuN, Mel’nichenko VYa. Diagnostic and Prognostic Value of Biochemical Markers of Infectious Complications of High-Dose Therapy with Autologous Hematopoietic Stem Cell Transplantation in Malignant Lymphoproliferative Diseases. Clinical oncohematology. 2017;10(1):113–9. doi: 10.21320/2500-2139-2017-10-1-113-119. (In Russ)]

  6. Zhang W, Zhao Q, Huang H. Febrile neutropenic infection occurred in cancer patients undergoing autologous peripheral blood stem cell transplantation. Transplant Proc. 2015;47(2):523–7. doi: 10.1016/j.transproceed.2015.01.013.

  7. Schmitz N, Pfistner B, Sextro M, et al. Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin’s disease: a randomised trial. Lancet. 2002;359(9323):2065–71. doi: 10.1016/s0140-6736(02)08938-9.

  8. Krishnamani K, Gandhi LV, Sadashivudu G, et al. Epedimiologic, clinical profile and factors affecting the outcome in febrile neutropenia. South Asian J Cancer. 2017;6(1):25–7. doi: 10.4103/2278-330X.202565.

  9. Bates DW, Sands K, Miller E, et al. Predicting bacteremia in patients with sepsis syndrome. Academic Medical Center Consortium Sepsis Project Working Group. J Infect Dis. 1997;176(6):1538–51. doi: 10.1086/514153.

  10. Klastersky J, de Naurois J, Rolston K, et al. Management of Febrile Neutropaenia: ESMO Clinical Practice Guidelines. Ann Oncol. 2016;27(Suppl 5):v111–v118. doi: 10.1093/annonc/mdw325.

  11. Homsi J, Walsh D, Panta R, et al. Infectious complications of advanced cancer. Support Care Cancer. 2000;8(6):487–92.

  12. Zembower TR. Epidemiology of infections in cancer patients. Cancer Treat Res. 2014;161:43–89. doi: 10.1007/978-3-319-04220-6_2.

  13. European Medicines Agency. Guideline on clinical evaluation of diagnostic agents. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003580.pdf. (accessed 30.07.2019).

  14. Павлушкина Л.В., Черневская Е.А., Дмитриева И.Б., Белобородова Н.В. Биомаркеры в клинической практике. Поликлиника. 2013;3:10–4.

    [Pavlushkina LV, Chernevskaya EA, Dmitrieva IB, Beloborodova NV. Biomarkers in clinical practice. Poliklinika. 2013;3:10–4. (In Russ)]

  15. Sbrana A, Torchio M, Comolli G, et al. Use of procalcitonin in clinical oncology: a literature review. New Microbiol. 2016;39(3):174–80.

  16. Pierrakos C, Vincent JV. Sepsis biomarkers: a review. Crit Care. 2010;14(1):R15. doi: 10.1186/cc8872.

  17. Kustan P, Horvath-Szalai Z, Muhl D. Nonconventional Markers of Sepsis. 2017;28(2):122–33.

  18. Colak A, Yilmaz C, Toprak B, Aktogu S. Procalcitonin and Crp as biomarkers in discrimination of community-acquired pneumonia and exacerbation of COPD. J Med Biochem. 2017;36:122–6. doi: 10.1515/jomb-2017-0011.

  19. Gao LQ, Liu XH, Zhang DH, et al. Early diagnosis of bacterial infection in patients with septicopyemia by laboratory analysis of PCT, CRP and IL-6. Exp Ther Med. 2017;13(6):3479–83. doi: 10.3892/etm.2017.4417.

  20. Povoa P, Coelho L, Almeida, et al. Early identification of intensive care unit-acquired infections with daily monitoring of C-reactive protein: a prospective observational study. Crit Care. 2006;10(2):R63. doi: 10.1186/cc4892.

  21. Morley JJ, Kushner I. Serum C-reactive protein levels in disease. Ann NY Acad Sci. 1982;389:(1):406–18. doi: 10.1111/j.1749-6632.1982.tb22153.x.

  22. Palmiere C, Augsburger M. Markers for sepsis diagnosis in the forensic setting: state of the art. Croat Med J. 2014;55(2):103–14. doi: 10.3325/cmj.2014.55.103.

  23. Meisner M, Tschaikowsky K, Palmers T. Procalcitonin and CRP in septic shock: Inflammatory parameters with different kinetics. Intens Care Med. 1996;22(S1):s13. doi: 10.1007/BF01921187.

  24. Samraj RS, Zingarelli B, Wong HR. Role of biomarkers in sepsis care. Shock. 2013;40(5):358–65. doi: 10.1097/Shk.0b013e3182a66bd6.

  25. Fujita MQ, Zhu B-L, Ishida K, et al. Serum C-reactive protein levels in postmortem blood – an analysis with special reference to the cause of death and survival time. Forensic Sci Int. 2002;130(2–3):160–6. doi: 10.1016/S0379-0738(02)00381-X.

  26. Pepys MB, Hirschfield GM. C-reactive protein: a critical update. J Clin Invest. 2003;111(12):1805–12. doi: 10.1172/jci18921.

  27. Duzenli KD, Ozdemir ZC, Bor O. Evaluation of febrile neutropenic attacks of pediatric hematology-oncology patients. Turk Pediatr Ars. 2017;52(4):213–20. doi: 10.5152/TurkPediatriArs.2017.5312.

  28. Pineda-Roman M, Barlogie B, Tricot G, et al. High-dose melphalan-based autotransplants for multiple myeloma: the Arkansas experience since 1989 in 3077 patients. Cancer. 2008;112(8):1754–64. doi: 10.1002/cncr.23327.

  29. Kollu V, Mott SL, Khan R, et al. C-Reactive Protein Monitoring Predicts Neutropenic Fever Following Autologous Hematopoietic Stem Cell Transplantation for Multiple Myeloma. Cureus. 2018;10(7):e2945. doi: 10.7759/cureus.2945.

  30. Ortega M, Rovira M, Almela M, et al. Measurement of C-reactive protein in adults with febrile neutropenia after hematopoietic cell transplantation. Bone Marrow Transplant. 2004;33(7):741–4. doi: 10.1038/sj.bmt.1704409.

  31. Schots R, Kaufman L, Van Riet I, et al. Monitoring of C-reactive protein after allogeneic bone marrow transplantation identifies patients at risk of severe transplant-related complications and mortality. Bone Marrow Transplant. 1998;22(1):79–85. doi: 10.1038/sj.bmt.1701286.

  32. Sato M, Nakasone H, Wada H, et al. Prediction of infectious events by the high-sensitivity C-reactive protein level before autologous hematopoietic cell transplantation for lymphoma and multiple myeloma. Transplant Infect Dis. 2013;15(4):E169–E171. doi: 10.1111/tid.12102.

  33. Massaro K, Costa SF. Role of Biomarkers as Predictors of Infection and Death in Neutropenic Febrile Patients after Hematopoietic Stem Cell Transplantation. Mediterr J Hematol Infect Dis. 2015;7(1):e2015059. doi: 10.4084/MJHID.2015.059.

  34. Sato M, Nakasone H, Oshima K, et al. Prediction of transplant-related complications by C-reactive protein levels before hematopoietic SCT. Bone Marrow Transplant. 2013;48(5):698–702. doi: 10.1038/bmt.2012.193.

  35. Pavlu J, Kew AK, Taylor-Roberts B, et al. Optimizing patient selection for myeloablative allogeneic hematopoietic cell transplantation in chronic myeloid leukemia in chronic phase. Blood. 2010;115(2):4018–20. doi: 10.1182/blood-2010-01-263624.

  36. Wang XS, Shi Q, Shah ND, et al. Inflammatory markers and development of symptom burden in patients with multiple myeloma during autologous stem cell transplantation. Clin Cancer Res. 2014;20(5):1366–74. doi: 10.1158/1078-0432.ccr-13-2442.

  37. Fassas AB, Miceli MH, Grazzlutti M, et al. Serial measurement of serum C-reactive protein levels can identify patients at risk for severe complications following autologous stem cell transplantation. Leuk Lymphoma. 2005;46(8):1159–61. doi: 10.1080/10428190500086121.

  38. Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 2016;6(6):664–79. doi: 10.1158/2159-8290.cd-16-0040.

  39. Maruna P, Nedelnikova K, Gurlich R. Physiology and genetics of procalcitonin. Physiol Res. 2000;49(Suppl 1):S57–61.

  40. Becker KL, Snider R, Nylen ES. Procalcitonin assay in systemic inflammation, infection, and sepsis: clinical utility and limitations. Crit Care Med. 2008;36(3):941–52. doi: 10.1097/CCM.0B013E318165BABB.

  41. Reinhart K, Meisner M, Brunkhorst FM. Markers for sepsis diagnosis: what is useful? Crit Care Clin. 2006;22(3):503–19. doi: 10.1016/j.ccc.2006.03.003.

  42. Picariello C, Lazzeri C, Valente S, et al. Procalcitonin in acute cardiac patients. Intern Emerg Med. 2011;6(3):245–52. doi: 10.1007/s11739-010-0462-x.

  43. Reinhart K, Bauer M, Riedelmann NC, et al. New approaches to sepsis: molecular diagnostics and biomarkers. Clin Microbiol Rev. 2012;25(4):609–34. doi: 10.1128/cmr.0001612.

  44. Assicot M, Gendrel D, Carsin H, et al. High serum procalcitonin concentrations in patients with sepsis and infection. Lancet. 1993;341(8844):515–8. doi: 10.1016/0140-6736(93)90277-n.

  45. Wu CW, Wu JY, Chen CK, et al. Does procalcitonin, C-reactive protein, or interleukin-6 test have a role in the diagnosis of severe infection in patients with febrile neutropenia? A systematic review and meta-analysis. Support Care Cancer. 2015;23(10):2863–72. doi: 10.1007/s00520-015-2650-8.

  46. Schuttrumpf S, Binder L, Hagemann T, et al. Utility of procalcitonin concentration in the evaluation of patients with malignant diseases and elevated C-reactive protein plasma concentrations. Clin Infect Dis. 2006;43(3):468–73. doi: 10.1086/505394.

  47. Shomali W, Hachem R, Chaftari AM, et al. Can procalcitonin distinguish infectious fever from tumor-related fever in non-neutropenic cancer patients? Cancer. 2012;118(23):5823–9. doi: 10.1002/cncr.27602.

  48. Meidani M, Khorvash F, Abolghasemi H, et al. Procalcitonin and quantitative C-reactive protein role in the early diagnosis of sepsis in patients with febrile neutropenia. South Asian J Cancer. 2013;2(4):216–9. doi: 10.4103/2278-330x.119913.

  49. Ahn S, Lee YS, Lim KS, et al. Adding Procalcitonin to the MASCC risk-index score could improve risk stratification of patients with febrile neutropenia. Support Care Cancer. 2013;21(8):2303–8. doi: 10.1007/s00520-013-1787-6.

  50. Chaftari AM, Hachem R, Reitzel R, et al. Role of Procalcitonin and Interleukin-6 in Predicting Cancer, and Its Progression Independent of Infection. PLoS One. 2015;10(7):e0130999. doi: 10.1371/journal.pone.0130999.

  51. Jimeno A, Garcia-Velasco A, Val del O, et al. Assessment of Procalcitonin as a Diagnostic and Prognostic Marker in Patients with Solid Tumors and Febrile Neutropenia. Cancer. 2004;100(11):2462–9. doi: 10.1002/cncr.20275.

  52. Carnino L, Betteto S, Loiacono M, et al. Procalcitonin as a predictive marker of infections in chemoinduced neutropenia. J Cancer Res Clin Oncol. 2010;136(4):611–5. doi: 10.1007/s00432-009-0699-9.

  53. Diness LV, Maraldo MV, Mortensen CE, et al. Procalcitonin and C-reactive protein as markers of bacterial infection in patients with solid tumors. Dan Med J. 2014;61(12):A4984.

  54. Giamarellou H, Giamarellos-Bourboulis EJ, Repoussis P, et al. Potential use of procalcitonin as a diagnostic criterion in febrile neutropenia: experience from a multicentre study. Clin Microbiol Infect. 2004;10(7):628–33. doi: 10.1111/j.1469-0691.2004.00883.x.

  55. Persson L, Engervall P, Magnuson A, et al. Use of inflammatory markers for early detection of bacteraemia in patients with febrile neutropenia. Scand J Infect Dis. 2004;36(5):365–71. doi: 10.1080/00365540410020217.

  56. Ruokonen E, Nousiainen T, Pulkki K, et al. Procalcitonin concentrations in patients with neutropenic fever. Eur J Clin Microbiol Infect Dis. 1999;18(4):283–5. doi: 10.1007/s100960050277.

  57. Robinson JO, Lamoth F, Bally F, et al. Monitoring procalcitonin in febrile neutropenia: what is its utility for initial diagnosis of infection and reassessment in persistent fever? PLoS One. 2011;6(4):e18886. doi: 10.1371/journal.pone.0018886.

  58. Patout M, Salaun M, Brunel V, et al. Diagnostic and prognostic value of serum procalcitonin concentrations in primary lung cancers. Clin Biochem. 2014;47(18):263–7. doi: 10.1016/j.clinbiochem.2014.09.002.

  59. Scheinpflug K, Schalk E, Grabert E, et al. Procalcitonin is not useful to discriminate between infectious and noninfectious CrP elevations in patients with non-small cell lung cancer. Infect Control Hosp Epidemiol. 2015;36(9):1117–8. doi: 10.1017/ice.2015.134.

  60. Yaegashi Y, Sato N, Suzuki Y, et al. Evaluation of a newly identified soluble CD14 subtype as a marker for sepsis. J Infect Chemother. 2005;11(5):234–8. doi: 10.1007/s10156-005-0400-4.

  61. Shirakawa K, Naitou K, Hirose J, et al. The new sepsis marker, sCD14-ST, induction mechanism in the rabbit sepsis models. Crit Care. 2010;14(Suppl 2):P19. doi: 10.1186/cc9122.

  62. Shozushima T, Takahashi G, Matsumoto N, et al. Usefulness of presepsin (sCD14-ST) measurements as a marker for the diagnosis and severity of sepsis that satisfied diagnostic criteria of systemic inflammatory response syndrome. J Infect Chemother. 2011;17(6):764–9. doi: 10.1007/s10156-011-0254-x.

  63. Endo S, Suzuki Y, Takahashi G, et al. Usefulness of presepsin in the diagnosis of sepsis in a multicenter prospective study. J Infect Chemother. 2012;18(6):891–7. doi: 10.1007/s10156-012-0435-2.

  64. Urbonas V, Eidukaite A, Tamuliene I. The predictive value of soluble biomarkers (CD14 subtype, interleukin-2 receptor, human leucocyte antigen-G) and procalcitonin in the detection of bacteremia and sepsis in pediatric oncology patients with chemotherapy-induced febrile neutropenia. Cytokine. 2013;62(1):34–7. doi: 10.1016/j.cyto.2013.02.030.

  65. Olad E, Sedighi I, Mehrvar A, et al. Presepsin (scd14) as a marker of serious bacterial infections in chemotherapy induced severe neutropenia. Iran J Pediatr. 2014;24(6):715–22.

  66. Korpelainen S, Intke C, Hamalainen S, et al. Soluble CD14 as a Diagnostic and Prognostic Biomarker in Hematological Patients with Febrile Neutropenia. Dis Mark. 2017;2017:1–8. doi: 10.1155/2017/9805609.

  67. Koh H, Aimoto M, Katayama T, et al. Diagnostic value of levels of presepsin (soluble CD14-subtype) in febrile neutropenia in patients with hematological disorders. J Infect Chemother. 2016;22(7):466–71. doi: 10.1016/j.jiac.2016.04.002.

  68. Stoma I, Karpov I, Uss A, et al. Diagnostic value of sepsis biomarkers in hematopoietic stem cell transplant recipients in a condition of high prevalence of gram-negative pathogens. Hematol Oncol Stem Cell Ther. 2017;10(1):15–21. doi: 10.1016/j.hemonc.2016.09.002.

  69. Ebisawa K, Koya J, Nakazaki K, et al. Usefulness of presepsin for early detection of infections in patients with hematologic disorders. Clin Chim Acta. 2018;486:374–80. doi: 10.1016/j.cca.2018.08.032.

Пневмоцистная пневмония после высокодозной химиотерапии с аутологичной трансплантацией периферических гемопоэтических стволовых клеток

В.О. Саржевский1, В.Я. Мельниченко1, Д.С. Колесникова1, О.Ю. Бронов1, В.М. Сотников2, Н.Н. Смольцова2

1 ФГБУ «Национальный медико-хирургический центр им. Н.И. Пирогова» МЗ РФ, ул. Нижняя Первомайская, д. 70, Москва, Российская Федерация, 105203

2 ФГБУ «Российский научный центр рентгенорадиологии» МЗ РФ, ул. Профсоюзная, д. 86, стр. 5, Москва, Российская Федерация, 117997

Для переписки: В.О. Саржевский, канд. мед. наук, доцент, ул. Нижняя Первомайская, д. 70, Москва, Российская Федерация, 105203; тел.: +7(495)603-72-18; e-mail: vladsar@pochta.ru

Для цитирования: Саржевский В.О., Мельниченко В.Я., Колесникова Д.С., Бронов О.Ю., Сотников В.М., Смольцова Н.Н. Пневмоцистная пневмония после высокодозной химиотерапии с аутологичной трансплантацией периферических гемопоэтических стволовых клеток. Клин. онкогематол. 2014; 7(4): 583–586.


РЕФЕРАТ

Пневмоцистная пневмония — редкое осложнение высокодозной полихимиотерапии и аутологичной трансплантации костного мозга/периферических гемопоэтических стволовых клеток у больных со злокачественными новообразованиями кроветворной и лимфоидной тканей. Отсутствие типичных клинических проявлений, высокий риск летального исхода требуют особого подхода к этой проблеме. Краткий обзор литературы и клиническое наблюдение дают представление о клинических симптомах, особенностях течения, диагностике, профилактике и лечении пневмоцистной пневмонии у данной категории больных.


Ключевые слова: пневмоцистная пневмония, высокодозная химиотерапия, аутологичная трансплантация костного мозга.

Принято в печать: 19 сентября 2014 г.

Читать статью в PDF pdficon


ЛИТЕРАТУРА

  1. Walzer P.D., Perl D.P., Krogstad D.J. et al. Pneumocystis carinii pneumonia in the United States: epidemiologic, diagnostic, and clinical features. Natl. Cancer Inst. Monogr. 1976; 43: 55–63.
  2. Green H., Paul M., Vidal L. et al. Prophylaxis of Pneumocystis pneumonia in immunocompromised non-HIV-infected patients: systematic review and metaanalysis of randomized controlled trials. Mayo Clin. Proc. 2007; 82(9): 1052–9.
  3. Colby C., McAfee S., Sackstein R. et al. A prospective randomized trial comparing the toxicity and safety of atovaquone with trimethoprim/ sulfamethoxazole as Pneumocystis carinii pneumonia prophylaxis following autologous peripheral blood stem cell transplantation. Bone Marrow Transplant. 1999; 24(8): 897–902.
  4. Sattler F.R., Cowan R., Nielsen D.M. et al. Trimethoprim-sulfamethoxazole compared with pentamidine for treatment of Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. A prospective, noncrossover study. Ann. Intern. Med. 1988; 109(4): 280–7.
  5. Hughes W., Leoung G., Kramer F. et al. Comparison of atovaquone (566C80) with trimethoprim-sulfamethoxazole to treat Pneumocystis carinii pneumonia in patients with AIDS. N. Engl. J. Med. 1993; 328(21): 1521–7.
  6. Metzner B., Gruneisl R., Gebauer W. et al. Late infectious complications after high-dose therapy and autologous blood stem cell transplantation. Med. Klin. (Munich) 2002; 97(11): 650–8.
  7. Decaudin D., Brousse N., Brice P. et al. Efficacy of autologous stem cell transplantation in mantle cell lymphoma: a 3-year follow-up study. Bone Marrow Transplant. 2000; 25(3): 251–6.
  8. Chen C.S., Boeckh M., Seidel K. Incidence, risk factors, and mortality from pneumonia developing late after hematopoietic stem cell transplantation. Bone Marrow Transplant. 2003; 32(5): 515–22.
  9. Chuu W.M., Catlett J.P., Perry D.J. Concurrent Pneumocystis carinii and cytomegalovirus pneumonia after autologous peripheral blood stem cell transplantation. Bone Marrow Transplant. 1999; 23(10): 1087–9.
  10. Kato H., Yamamoto K., Taji H. et al. Interstitial pneumonia after autologous hematopoietic stem cell transplantation in B-cell non-Hodgkin lymphoma. Clin. Lymphoma Myeloma Leuk. 2011; 11(6): 483–9.
  11. Munker R., Lazarus H.M., Atkinson K. The BMT Data Book, 2nd ed. Cambridge University Press, 2009: 245.

Кардиотоксичность высокодозной химиотерапии и аутологичной трансплантации костного мозга при онкогематологических заболеваниях

В.О. Саржевский, Д.С. Колесникова, В.Я. Мельниченко, В.П. Тюрин

ФГБУ «Национальный медико-хирургический центр им. Н.И. Пирогова» МЗ РФ, Москва, Российская Федерация


РЕФЕРАТ

В обзоре представлены современные данные, касающиеся кардиотоксичности высокодозной химиотерапии и аутологичной трансплантации костного мозга (периферических кроветворных стволовых клеток) при онкогематологических заболеваниях. Дана подробная информация о методах исследования, используемых для оценки кардиотоксичности, представлены клинические особенности кардиальных осложнений на всех этапах проведения трансплантации (мобилизация, кондиционирование, ранний посттрансплантационный период) и в более поздние сроки после окончания лечения. Приведены подходы к профилактике и лечению кардиотоксичности у данной категории больных.


Ключевые слова: кардиотоксичность, высокодозная химиотерапия, аутологичная трансплантация костного мозга.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Phillips G.L., Reece D.E. Clinical studies of autologous bone marrow transplantation in Hodgkin’s disease. Clin. Haematol. 1986; 15(1): 151–66.
  2. Appelbaum F.R., Sullivan K.M., Buckner C.D. et al. Treatment of malignant lymphoma in 100 patients with chemotherapy, total body irradiation, and marrow transplantation. J. Clin. Oncol. 1987; 5(9): 1340–7.
  3. Cazin B., Gorin N.C., Laporte J.P. et al. Cardiac complications after bone marrow transplantation. A report on a series of 63 consecutive transplantations. Cancer 1986; 57(10): 2061–9.
  4. Goldberg M.A., Antin J.H., Guinan E.C. et al. Cyclophosphamide cardiotoxicity: an analysis of dosing as a risk factor. Blood 1986; 68(5): 1114–8.
  5. Murdych T., Weisdorf D.J. Serious cardiac complications during bone marrow transplantation at the University of Minnesota, 1977–1997. Bone Marrow Transplant. 2001; 28(3): 283–7.
  6. Dolci A., Dominici R., Cardinale D. et al. Biochemical markers for prediction of chemotherapy-induced cardiotoxicity: systematic review of the literature and recommendations for use. Am. J. Clin. Pathol. 2008; 130(5): 688–95.
  7. Cardinale D., Sandri M.T., Martinoni A. et al. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J. Am. Coll. Cardiol. 2000; 36(2): 517–22.
  8. Cardinale D., Sandri M.T., Colombo A. et al. Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation 2004; 109(22): 2749–54.
  9. Chung T., Lim W.C., Sy R. et al. Subacute cardiac toxicity following autologous haematopoietic stem cell transplantation in patients with normal cardiac function. Heart 2008; 94(7): 911–8.
  10. Horacek J.M., Pudil R., Tichy M. et al. Biochemical markers and assessment of cardiotoxicity during preparative regimen and hematopoietic cell transplantation in acute leukemia. Exp. Oncol. 2007; 29(3): 243–7.
  11. Auner H.W., Tinchon C., Linkesch W. et al. Prolonged monitoring of troponin T for the detection of anthracycline cardiotoxicity in adults with hematological malignancies. Ann. Hematol. 2003; 82(4): 218–22.
  12. Cardinale D., Colombo A., Sandri M.T. et al. Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensinconverting enzyme inhibition. Circulation 2006; 114(23): 2474–81.
  13. Кардиология. Национальное руководство: краткое издание. Под ред. Ю.Н. Беленкова, Р.Г. Оганова. М.: ГЭОТАР-Медиа, 2012. [Kardiologiya. Natsionalnoye rukovodstvo: kratkoye izdaniye. Pod red. Yu.N. Belenkova, R.G. Oganova (Cardiology. National manual: brief edition. Ed. by Yu.N. Belenkova and R.G. Oganova). M.: GEOTAR-Media, 2012.]
  14. Suzuki T., Hayashi D., Yamazaki T. et al. Elevated B-type natriuretic peptide levels after anthracycline administration. Am. Heart J. 1998; 136(2): 362–3.
  15. Nousiainen T., Jantunen E., Vanninen E. et al. Acute neurohumoral and cardiovascular effects of idarubicin in leukemia patients. Eur. J. Haematol. 1998; 61(5): 347–53.
  16. Snowden J.A., Hill G.R., Hunt P. et al. Assessment of cardiotoxicity during haemopoietic stem cell transplantation with plasma brain natriuretic peptide. Bone Marrow Transplant. 2000; 26(3): 309–13.
  17. Masuko M., Ito M., Kurasaki T. et al. Plasma brain natriuretic peptide during myeloablative stem cell transplantation. Intern. Med. 2007; 46(9): 551–5.
  18. Zver S., Zadnik V., Bunc M. et al. Cardiac toxicity of high-dose cyclophosphamide in patients with multiple myeloma undergoing autologous hematopoietic stem cell transplantation. Int. J. Hematol. 2007; 85(5): 408–14.
  19. Kupari M., Volin L., Suokas A. et al. Cardiac involvement in bone marrow transplantation: electrocardiographic changes, arrhythmias, heart failure and autopsy findings. Bone Marrow Transplant. 1990; 5(2): 91–8.
  20. Styler M.J., Topolsky D.L., Crilley P.A. et al. Transient high grade heart block following autologous bone marrow infusion. Bone Marrow Transplant. 1992; 10(5): 435–8.
  21. Munker R., Lazarus H. M., Atkinson K. The BMT Data Book, 2nd ed. Cambridge: Cambridge University Press, 2009: 237.
  22. Pihkala J., Saarinen U.M., Lundstrom U. et al. Effects of bone marrow transplantation on myocardial function in children. Bone Marrow Transplant. 1994; 13(2): 149–55.
  23. Zver S., Zadnik V., Cernelc P. et al. Cardiac toxicity of high-dose cyclophosphamide and melphalan in patients with multiple myeloma treated with tandem autologous hematopoietic stem cell transplantation. Int. J. Hematol. 2008; 88(2): 227–36.
  24. Минимальные клинические рекомендации Европейского общества медицинской онкологии (ESMO). М., 2010: 428. [Minimalnye klinicheskiye rekomendatsii Yevropeyskogo obshchestva meditsinskoy onkologii (ESMO) (Minimal clinical recommendations of European Society for Medical Oncology (ESMO)). M., 2010: 428.]
  25. Nagueh S.F., Sua H., Kopelen H.A. et al. Hemodynamic determinants of the mitral annulus diastolic velocities by tissue Doppler. J. Am. Coll. Cardiol. 2001; 37: 278–85.
  26. Шкляева А.В. Ультразвуковая оценка функционального состояния левого желудочка сердца при высокодозной немиелоаблативной химиотерапии у больных злокачественными лимфомами: Автореф. дис. ¼ канд. мед. наук. Обнинск, 2011.
  27. [Shklyayeva A.V. Ultrazvukovaya otsenka funktsionalnogo sostoyaniya levogo zheludochka serdtsa pri vysokodoznoy nemiyeloablativnoy khimioterapii u bolnykh zlokachestvennymi limfomami: Avtoref. dis. ¼ kand. med. nauk (Ultrasound evaluation of left ventricle function during high-dose nonmyeloablative chemotherapy in patients with malignant lymphomas. Author’s summary of dissertation for the degree of PHD). Obninsk, 2011.]
  28. Kuittinen T., Husso-Saastamoinen M., Sipola P. et al. Very acute cardiac toxicity during BEAC chemotherapy in non-Hodgkin’s lymphoma patients undergoing autologous stem cell transplantation. Bone Marrow Transplant. 2005; 36(12): 1077–82.
  29. Bosch X., Esteve J., Sitges M. et al. Prevention of chemotherapy-induced left ventricular dysfunction with enalapril and carvedilol: rationale and design of the OVERCOME trial. J. Card. Fail. 2011; 17(8): 643–8.
  30. Gustavsson A., Eskilsson J., Landberg T. et al. Late cardiac effects after mantle radiotherapy in patients with Hodgkin’s disease. Ann. Oncol. 1990; 1: 355–63.
  31. Girinsky T., Cordova A., Rey A. et al. Thallium-201 scintigraphy is not predictive of late cardiac complications in patients with Hodgkin’s disease treated with mediastinal radiation. Int. J. Radiat. Oncol. Biol. Phys. 2000; 48: 1503–6.
  32. Glanzmann C., Kaufmann P., Jenni R. et al. Cardiac risk after mediastinal irradiation for Hodgkin’s disease. Radiother. Oncol. 1998; 46: 51–62.
  33. Salloum E., Jillella A.P., Nadkarni R. et al. Assessment of pulmonary and cardiac function after high dose chemotherapy with BEAM and peripheral blood progenitor cell transplantation. Cancer 1998; 82(8): 1506–12.
  34. Hertenstein B., Stefanic M., Schmeiser T. et al. Cardiac toxicity of bone marrow transplantation: predictive value of cardiologic evaluation before transplant. J. Clin. Oncol. 1994; 12(5): 998–1004.
  35. Herbay A., Dorken B., Mall G. et al. Cardiac damage in autologous bone marrow transplant patients: an autopsy study. Cardiotoxic pretreatment as a major risk factor. Klin. Wochenschr. 1988; 66(23): 1175–81.
  36. Bearman S.I., Petersen F.B., Schor R.A. et al. Radionuclide ejection fractions in the evaluation of patients being considered for bone marrow transplantation: risk for cardiac toxicity. Bone Marrow Transplant. 1990; 5(3): 173–7.
  37. Lehmann S., Isberg B., Ljungman P. et al. Cardiac systolic function before and after hematopoietic stem cell transplantation. Bone Marrow Transplant. 2000; 26(2): 187–92.
  38. Armenian S.H., Sun C.L., Shannon T. et al. Incidence and predictors of congestive heart failure after autologous hematopoietic cell transplantation. Blood 2011; 118(23): 6023–9.
  39. Schouten H.C., Maragos D., Vose J. et al. Diabetes mellitus or an impaired glucose tolerance as a potential complicating factor in patients treated with high-dose therapy and autologous bone marrow transplantation. Bone Marrow Transplant. 1990; 6(5): 333–5.
  40. Atkinson K. (ed.) Clinical Bone Marrow and Blood Stem Cell Transplantation, 3rd ed. Cambridge: Cambridge University Press, 2000: 1111.
  41. Stillwell E.E., Wessler J.D., Rebolledo B.J. et al. Retrospective outcome data for hematopoietic stem cell transplantation in patients with concurrent coronary artery disease. Biol. Blood Marrow Transplant. 2011; 17(8): 1182–6.
  42. Tyndall A., Passweg J., Gratwohl A. Haemopoietic stem cell transplantation in the treatment of severe autoimmune diseases 2000. Ann. Rheum. Dis. 2001; 60(7): 702–7.
  43. Eckman P.M., Bertog S.C., Wilson R.F. et al. Ischemic cardiac complications following G-CSF. Catheter. Cardiovasc. Interv. 2010; 76(1): 98–101.
  44. Nakane T., Nakamae H., Koh H. et al. Heart rate variability during and after peripheral blood stem cell leukapheresis in autologous transplant patients and allogeneic transplant donors. Int. J. Hematol. 2010; 91(3): 478–84.
  45. Ulrickson M., Aldridge J., Kim H.T. et al. Busulfan and cyclophosphamide (Bu/Cy) as a preparative regimen for autologous stem cell transplantation in patients with non-Hodgkin lymphoma: a single-institution experience. Biol. Blood Marrow Transplant. 2009; 15(11): 1447–54.
  46. Reece D.E., Nevill T.J., Sayegh A. et al. Regimen-related toxicity and non-relapse mortality with high-dose cyclophosphamide, carmustine (BCNU) and etoposide (VP16–213) (CBV) and CBV plus cisplatin (CBVP) followed by autologous stem cell transplantation in patients with Hodgkin’s disease. Bone Marrow Transplant. 1999; 23(11): 1131–8.
  47. Rosenthal M.A., Grigg A.P., Sheridan W.P. High dose busulphan/cyclophosphamide for autologous bone marrow transplantation is associated with minimal non-hemopoietic toxicity. Leuk. Lymphoma 1994; 14(3–4): 279–83.
  48. Kim J.W., Lee H.J., Yi H.G. et al. Mitoxantrone, etoposide, cytarabine, and melphalan (NEAM) followed by autologous stem cell transplantation for patients with chemosensitive aggressive non-Hodgkin lymphoma. Am. J. Hematol. 2012; 87(5): 479–83.
  49. Ferrara F., Palmieri S., Pedata M. et al. Autologous stem cell transplantation for elderly patients with acute myeloid leukaemia conditioned with continuous infusion idarubicin and busulphan. Hematol. Oncol. 2009; 27(1): 40–5.
  50. Krishnan G.S., Chaudhary V., Al-Janadi A. et al. BCNU toxicity presenting with a large pericardial and pleural effusion. Ann. Transplant. 2008; 13(1): 44–7.
  51. Mileshkin L.R., Seymour J.F., Wolf M.M. et al. Cardiovascular toxicity is increased, but manageable, during high-dose chemotherapy and autologous peripheral blood stem cell transplantation for patients aged 60 years and older. Leuk. Lymphoma 2005; 46(11): 1575–9.
  52. Sirohi B., Powles R., Treleaven J. et al. The role of autologous transplantation in patients with multiple myeloma aged 65 years and over. Bone Marrow Transplant. 2000; 25(5): 533–9.
  53. Davis J., Rowley S.D., Santos G.W. Toxicity of autologous bone marrow graft infusion. Prog. Clin. Biol. Res. 1990; 333: 531–40.
  54. Galmes A., Gutierrez A., Sampol A. et al. Long-term hematological reconstitution and clinical evaluation of autologous peripheral blood stem cell transplantation after cryopreservation of cells with 5% and 10% dimethylsulfoxide at 80 degrees C in a mechanical freezer. Haematologica 2007; 92(7): 986–9.
  55. Rapoport A.P., Rowe J.M., Packman C.H. et al. Cardiac arrest after autologous marrow infusion. Bone Marrow Transplant. 1991; 7(5): 401–3.
  56. Martino M., Morabito F., Messina G. et al. Fractionated infusions of cryopreserved stem cells may prevent DMSO-induced major cardiac complications in graft recipients. Haematologica 1996; 81(1): 59–61.
  57. Donmez A., Zoghi M., Cagirgan S. et al. The effect of hematopoietic progenitor cells’ temperature on cardiac arrhythmias in patients given peripheral blood progenitor cells. Transfus. Apher. Sci. 2006; 34(3): 245–51.
  58. Quinn J.P., Counts G.W., Meyers J.D. Intracardiac infections due to coagulase-negative Staphylococcus associated with Hickman catheters. Cancer 1986; 57(5): 1079–82.
  59. Martino P., Micozzi A., Venditti M. et al. Catheter-related right-sided endocarditis in bone marrow transplant recipients. Rev. Infect. Dis. 1990; 12(2): 250–7.
  60. Philip T., Biron P., Herve P. et al. Massive BACT chemotherapy with autologous bone marrow transplantation in 17 cases of non-Hodgkin’s malignant lymphoma with a very bad prognosis. Eur. J. Cancer Clin. Oncol. 1983; 19(10): 1371–9.
  61. Guerin C., Billard J.L., Jaubert J. et al. Pericardial aspergillosis in a bone marrow transplant recipient. Intens. Care Med. 1989; 15(5): 330.
  62. Ellerbroek P., Kuipers S., Rozenberg-Arska M. et al. Oerskovia xanthineolytica: a new pathogen in bone marrow transplantation. Bone Marrow Transplant. 1998; 22(5): 503–5.
  63. Bhatia S., Robison L.L., Francisco L. et al. Late mortality in survivors of autologous hematopoietic-cell transplantation: report from the Bone Marrow Transplant Survivor Study. Blood 2005; 105(11): 4215–22.
  64. Lavoie J.C., Connors J.M., Phillips G.L. et al. High-dose chemotherapy and autologous stem cell transplantation for primary refractory or relapsed Hodgkin lymphoma: long-term outcome in the first 100 patients treated in Vancouver. Blood 2005; 106(4): 1473–8.
  65. Ruiz-Soto R., Sergent G., Gisselbrecht C. et al. Estimating late adverse events using competing risks after autologous stem-cell transplantation in aggressive non-Hodgkin lymphoma patients. Cancer 2005; 104(12): 2735–42.
  66. Silber J.H., Cnaan A., Clark B.J. et al. Enalapril to prevent cardiac function decline in long-term survivors of pediatric cancer exposed to anthracyclines. J. Clin. Oncol. 2004; 22(5): 820–8.
  67. Lipshultz S.E., Lipsitz S.R., Sallan S.E. et al. Long-term enalapril therapy for left ventricular dysfunction in doxorubicin-treated survivors of childhood cancer. J. Clin. Oncol. 2002; 20(23): 4517–22.
  68. Kakavas P.W., Ghalie R., Parrillo J.E. et al. Angiotensin converting enzyme inhibitors in bone marrow transplant recipients with depressed left ventricular function. Bone Marrow Transplant. 1995; 15(6): 859–61

Легочные осложнения высокодозной химиотерапии и аутологичной трансплантации костного мозга при онкогематологических заболеваниях

В.О. Саржевский, В.Я. Мельниченко, В.П. Тюрин

ФГБУ «Национальный медико-хирургический центр им. Н.И. Пирогова» МЗ РФ, Москва, Российская Федерация


РЕФЕРАТ

В обзоре представлены современные данные о легочных осложнениях высокодозной химиотерапии и аутологичной трансплантации костного мозга (периферических гемопоэтических стволовых клеток) у пациентов с онкогематологическими заболеваниями. Представлена подробная информация о методах исследования для оценки легочной токсичности, патогенетических и клинических особенностях легочных осложнений на всех этапах проведения трансплантации (ранняя легочная токсичность) и в более поздние сроки после окончания лечения (поздняя легочная токсичность). Приведены подходы к профилактике и лечению легочных осложнений у данной категории больных.


Ключевые слова: высокодозная химиотерапия, аутологичная трансплантация костного мозга, легочная токсичность.

Читать статью в PDFpdficon


ЛИТЕРАТУРА

  1. Prince D.S., Wingard J.R., Saral R. et al. Longitudinal changes in pulmonary function following bone marrow transplantation. Chest 1989; 96(2): 301–6.
  2. Arvidson J., Bratteby L.E., Carlson K. et al. Pulmonary function after autologous bone marrow transplantation in children. Bone Marrow Transplant. 1994; 14(1): 117–23.
  3. Ghalie R., Szidon J.P., Thompson L. et al. Evaluation of pulmonary complications after bone marrow transplantation: the role of pretransplant pulmonary function tests. Bone Marrow Transplant. 1992; 10(4): 359–65.
  4. Nenadov Beck M., Meresse V., Hartmann O. et al. Long-term pulmonary sequelae after autologous bone marrow transplantation in children without total body irradiation. Bone Marrow Transplant. 1995; 16(6): 771–5.
  5. Badier M., Guillot C., Delpierre S. et al. Pulmonary function changes 100 days and one year after bone marrow transplantation. Bone Marrow Transplant. 1993; 12(5): 457–61.
  6. Ho V.T., Weller E., Lee S.J. et al. Prognostic factors for early severe pulmonary complications after hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2001; 7(4): 223–9.
  7. Shannon V.R., Andersson B.S., Lei X. et al. Utility of early versus late fiberoptic bronchoscopy in the evaluation of new pulmonary infiltrates following hematopoietic stem cell transplantation. Bone Marrow Transplant. 2010; 45(4): 647–55.
  8. Hofmeister C.C., Czerlanis C., Forsythe S. et al. Retrospective utility of bronchoscopy after hematopoietic stem cell transplantation. Bone Marrow Transplant. 2006; 38(10): 693–8.
  9. Azoulay E., Mokart D., Rabbat A. et al. Diagnostic bronchoscopy in hematology and oncology patients with acute respiratory failure: prospective multicenter data. Crit. Care Med. 2008; 36(1): 100–7.
  10. NCCN Guidelines Version 1.2012. Prevention and Treatment of CancerRelated Infection. 2012: 15.
  11. Sharma S., Nadrous H.F., Peters S.G. et al. Pulmonary complications in adult blood and marrow transplant recipients: autopsy findings. Chest. 2005; 128(3): 1385–92.
  12. Roychowdhury M., Pambuccian S.E., Aslan D.L. et al. Pulmonary complications after bone marrow transplantation: an autopsy study from a large transplantation center. Arch. Pathol. Lab. Med. 2005; 129(3): 366–71.
  13. Munker R., Lazarus H.M., Atkinson K. The BMT Data Book, 2nd edn. Cambridge University Press, 2009: 237.
  14. Frankovich J., Donaldson S.S., Lee Y. et al. High-dose therapy and autologous hematopoietic cell transplantation in children with primary refractory and relapsed Hodgkin’s disease: atopy predicts idiopathic diffuse lung injury syndromes. Biol. Blood Marrow Transplant. 2001; 7(1): 49–57.
  15. Avivi I., Hardak E., Shaham B. et al. Low incidence of long-term respiratory impairment in Hodgkin lymphoma survivors. Ann. Hematol. 2012; 91(2): 215–21.
  16. Arimura K., Inoue H., Kukita T. et al. Acute lung Injury in a healthy donor during mobilization of peripheral blood stem cells using granulocyte-colony stimulating factor alone. Haematologica 2005; 90(3): ECR10.
  17. Krishnan G.S., Chaudhary V., Al-Janadi A. et al. BCNU toxicity presenting with a large pericardial and pleural effusion. Ann. Transplant. 2008; 13(1): 44–7.
  18. Jones R.B., Matthes S., Shpall E.J. et al. Acute lung injury following treatment with high-dose cyclophosphamide, cisplatin, and carmustine: pharmacodynamic evaluation of carmustine. J. Natl. Cancer Inst. 1993; 85(8): 640–7.
  19. Puig N., de la Rubia J., Remigia M.J. et al. Morbidity and transplantrelated mortality of CBV and BEAM preparative regimens for patients with lymphoid malignancies undergoing autologous stem-cell transplantation. Leuk. Lymphoma 2006; 47(8): 1488–94.
  20. Stuart M.J., Chao N.S., Horning S.J. et al. Efficacy and toxicity of a CCNU-containing high-dose chemotherapy regimen followed by autologous hematopoietic cell transplantation in relapsed or refractory Hodgkin’s disease. Biol. Blood Marrow Transplant. 2001; 7(10): 552–60.
  21. Munker R., Hilderbrandt G.C., Lazarus H.M., Atkinson K. The BMT Data Book: including cellular therapy, 3nd edn. Cambridge University Press, 2013: 282.
  22. Malik S.W., Myers J.L., DeRemee R.A. et al. Lung toxicity associated with cyclophosphamide use. Two distinct patterns. Am. J. Respir. Crit. Care Med. 1996; 154(6 Pt. 1): 1851–6.
  23. Chen C.I., Abraham R., Tsang R. et al. Radiation-associated pneumonitis following autologous stem cell transplantation: predictive factors, disease characteristics and treatment outcomes. Bone Marrow Transplant. 2001; 27(2): 177–82.
  24. Afessa B., Abdulai R.M., Kremers W.K. et al. Risk factors and outcome of pulmonary complications after autologous hematopoietic stem cell transplant. Chest 2012; 141(2): 442–50.
  25. Halaburda K., Nasilowska-Adamska B., Grabarczyk P. et al. Limited predictive value of real-time quantitative PCR cytomegalovirus monitoring in the blood. Fatal CMV pneumonia in an autologous stem cell transplant recipient previously treated with alemtuzumab. Ann. Transplant. 2007; 12(2): 37–40.
  26. Mariotte E., Schnell D., Scieux C. et al. Significance of herpes-virus 6 in BAL fluid of hematology patients with acute respiratory failure. Infection 2011; 39(3): 225–30.
  27. Robbins R.A., Linder J., Stahl M.G. et al. Diffuse alveolar hemorrhage in autologous bone marrow transplant recipients. Am. J. Med. 1989; 87(5): 511–8.
  28. Afessa B., Tefferi A., Litzow M.R. et al. Diffuse alveolar hemorrhage in hematopoietic stem cell transplant recipients. Am. J. Respir. Crit. Care Med. 2002; 166(5): 641–5.
  29. Metcalf J.P., Rennard S.I., Reed E.C. et al. Corticosteroids as adjunctive therapy for diffuse alveolar hemorrhage associated with bone marrow transplantation. University of Nebraska Medical Center Bone Marrow Transplant Group. Am. J. Med. 1994; 96(4): 327–34.
  30. Lee C.K., Gingrich R.D., Hohl R.J. et al. Engraftment syndrome in autologous bone marrow and peripheral stem cell transplantation. Bone Marrow Transplant. 1995; 16(1): 175–82.
  31. Spitzer T.R. Engraftment syndrome following hematopoietic stem cell transplantation. Bone Marrow Transplant. 2001; 27(9): 893–8.
  32. Edenfield W.J., Moores L.K., Goodwin G. et al. An engraftment syndrome in autologous stem cell transplantation related to mononuclear cell dose. Bone Marrow Transplant. 2000; 25(4): 405–9.
  33. Nurnberger W., Willers R., Burdach S. et al. Risk factors for capillary leakage syndrome after bone marrow transplantation. Ann. Hematol. 1997; 74(5): 221–4.
  34. Kantrow S.P., Hackman R.C., Boeckh M. et al. Idiopathic pneumonia syndrome: changing spectrum of lung injury after marrow transplantation. Transplantation 1997; 63(8): 1079–86.
  35. Clark J.G., Hansen J.A., Hertz M.I. et al. NHLBI workshop summary. Idiopathic pneumonia syndrome after bone marrow transplantation. Am. Rev. Respir. Dis. 1993; 147(6 Pt. 1): 1601–6.
  36. Kato H., Yamamoto K., Taji H. et al. Interstitial pneumonia after autologous hematopoietic stem cell transplantation in B-cell non-Hodgkin lymphoma. Clin. Lymph. Myel. Leuk. 2011; 11(6): 483–9.
  37. Metzner B., Gruneisl R., Gebauer W. et al. Late infectious complications after high-dose therapy and autologous blood stem cell transplantation. Med. Klin. (Munich) 2002; 97(11): 650–8.
  38. Roy V., Weisdorf D. Mycobacterial infections following bone marrow transplantation: a 20 year retrospective review. Bone Marrow Transplant. 1997; 19(5): 467–70.
  39. Akan H., Arslan O., Akan O.A. et al. Tuberculosis in stem cell transplant patients. J. Hosp. Infect. 2006; 62(4): 421–6.
  40. Marras T.K., Chan C.K. Obliterative bronchiolitis complicating bone marrow transplantation. Semin. Respir. Crit. Care Med. 2003; 24(5): 531–42.
  41. Paz H.L., Crilley P., Patchefsky A., Schiffman R.L., Brodsky I. Bronchiolitis obliterans after autologous bone marrow transplantation. Chest 1992; 101(3): 775–8.
  42. Gulbahce H.E., Pambuccian S.E., Jessurun J. et al. Pulmonary nodular lesions in bone marrow transplant recipients: impact of histologic diagnosis on patient management and prognosis. Am J. Clin. Pathol. 2004; 121(2): 205–10.
  43. Witherspoon R.P., Fisher L.D., Schoch G. et al. Secondary cancers after bone marrow transplantation for leukemia or aplastic anemia. N. Engl. J. Med. 1989; 321(12): 784–9.
  44. Danner-Koptik K.E., Majhail N.S., Brazauskas R. et al. Second malignancies after autologous hematopoietic cell transplantation in children. Bone Marrow Transplant. 2013; 48(3): 363–8.