Cardio-oncology and Oncohematology: Examination Algorithms, Prophylactic and Treatment of Cardiotoxicity, Trends in Rehabilitation

EI Emelina, GE Gendlin, IG Nikitin

NI Pirogov Russian National Research Medical University, 1 Ostrovityanova str., Moscow, Russian Federation, 117997

For correspondence: Elena Ivanovna Emelina, MD, PhD, 1 Ostrovityanova str., Moscow, Russian Federation, 117997; Tel.: +7(916)412-59-78; e-mail: eei1210@mail.ru

For citation: Emelina EI, Gendlin GE, Nikitin IG. Cardio-oncology and Oncohematology: Examination Algorithms, Prophylactic and Treatment of Cardiotoxicity, Trends in Rehabilitation. Clinical oncohematology. 2021;14(2):239–61. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-239-261


ABSTRACT

Successful chemotherapy in the treatment of hematological diseases is determined not only by the efficacy of antitumor drugs, but by the timely correction of adverse events, among which especially important are cardiac complications associated with both already existing cardiovascular diseases and cardiotoxicity of cytostatic drugs. Of particular importance is also a frequent lack of systemic cardiological examination of oncohematological patients. The urgency of this issue was the reason for creating cardio-oncological clinics focused on the closest co-operation of cardiologists with drug chemotherapy experts. Hematological patients are a particular group among chemotherapy recipients. Potential curability of an oncohematological disease and achieving durable MRD-negative remission raise the importance of irreversible or long-term cardiac complications directly affecting the quality of life and life expectancy. Besides, in some cases long-term or life-long administration of certain cardiotoxic antitumor drugs requires a particular cardiological follow-up. A broad variety of cardiotoxic effects of antitumor drugs and peculiarities of their clinical manifestations call for the exact algorithms of cardiological examination to be observed for the timely detection and treatment of cardiovascular complications. The now available studies and interdisciplinary work of cardiologists and oncologists (oncohematologists) can yield such algorithms for examination and the approaches to prophylactic and treatment of cardiotoxicity as well as to rehabilitation of patients.

Keywords: oncohematology, cardio-oncology, cardiotoxicity, antitumor drugs, prophylactic of cardiac adverse events, cardio-oncological rehabilitation.

Received: November 19, 2020

Accepted: February 10, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Zamorano JL, Lancellotti P, Munoz DR, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines. Eur Heart J. 2016;37(36):2768–801. doi: 10.1093/eurheartj/ehw211.
  2. Lyon AR, Dent S, Stanway S, et al. Baseline cardiovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer therapies: a position statement and new risk assessment tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in collaboration with the International Cardio-Oncology Society. Eur J Heart Fail. 2020;22(11):1945–60. doi: 10.1002/ejhf.1920.
  3. Curigliano G, Lenihan D, Fradley M, et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. ESMO Guidelines Committee. Ann Oncol. 2020;31(2):171–90. doi: 10.1016/j.annonc.2019.10.023.
  4. S. Department of Health and Human Services, National Institutes of Health, National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE). Version 5.0, 2017. Available from: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5×11.pdf (accessed 10.02.2021).
  5. Кулиева А.А., Емелина Е.И., Гендлин Г.Е. и др. Сердечно-сосудистые осложнения терапии ингибиторами контрольных точек иммунитета. Качественная клиническая практика. 2019;4:55–65. doi: 10.1016/2588-0519-2019-4-55-65.
    [Kulieva AA, Emelina EI, Gendlin GE, et al. Cardiovascular complications of immune checkpoint inhibitors. Kachestvennaya klinicheskaya praktika. 2019;4:55–65. doi: 10.1016/2588-0519-2019-4-55-65. (In Russ)]
  6. Kim P, Zarifa A, Salih M, et al. Cardiotoxicity of FDA-approved immune checkpoint inhibitors: A rare but serious adverse event. J Immunother Precis Oncol. 2018;1(2):68–77. doi: 4103/JIPO.JIPO_15_18.
  7. Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med. 2018;378(2):158–68. doi: 10.1056/NEJMra1703481.
  8. Шубникова Е.В., Букатина Т.М., Вельц Н.Ю. и др. Ингибиторы контрольных точек иммунного ответа: новые риски нового класса противоопухолевых средств. Безопасность и риск фармакотерапии. 2020;8(1):9–22. doi: 10.30895/2312-7821-2020-8-1-9-22.
    [Shubnikova EV, Bukatina TM, Velts NYu, et al. Immune Response Checkpoint Inhibitors: New Risks of a New Class of Antitumor Agents. Safety and Risk of Pharmacotherapy. 2020;8(1):9–22. doi: 10.30895/2312-7821-2020-8-1-9-22. (In Russ)]
  9. Armand P. Immune checkpoint blockade in hematologic malignancies. Blood. 2015;125(22):3393–400. doi: 10.1182/blood-2015-02-567453.
  10. Лепик К.В. Ингибиторы иммунных контрольных точек в терапии лимфом. Клиническая онкогематология. 2018;11(4):303–12. doi: 10.21320/2500-2139-2018-11-4-303-312.
    [Lepik KV. Immune Checkpoint Inhibitors in the Treatment of Lymphomas. Clinical oncohematology. 2018;11(4):303–12. doi: 10.21320/2500-2139-2018-11-4-303-312. (In Russ)]
  11. Cardinale D, Colombo A, Sandri MT, et al. Prevention of High-Dose Chemotherapy-Induced Cardiotoxicity in High-Risk Patients by Angiotensin-Converting Enzyme Inhibition. Circulation. 2006;114(23):2474–81. doi: 10.1161/CIRCULATIONAHA.106.635144.
  12. Gilchrist SC, Barac A, Ades PA, et al. Cardio-oncology rehabilitation to manage cardiovascular outcomes in cancer patients and survivors: a scientific statement from the American Heart Association. 2019;139(21):997–1012. doi: 10.1161/CIR.0000000000000679.
  13. Larsen CM, Mulvagh SL. Cardio-oncology: what you need to know now for clinical practice and echocardiography. Echo Res Pract. 2017;4(1):R33–R41. doi: 10.1530/ERP-17-0013.
  14. Maia TN, Araujo GB, Teixeira JA, et al. Cardiotoxicity of Doxorubicin Treatment and Physical Activity: A Systematic Review. Int J Cardiovasc Sci. 2017;30(1):70–80. doi: 10.5935/2359-4802.20170004.
  15. Fischetti F, Greco G, Cataldi S, et al. Effects of Physical Exercise Intervention on Psychological and Physical Fitness in Lymphoma Patients. 2019;55(7):379. doi: 10.3390/medicina55070379.
  16. Lenneman AJ, Wang L, Wigger M, et al. Heart transplant survival outcomes for adriamycin-dilated cardiomyopathy. Am J Cardiol. 2013;111(4):609–12. doi: 10.1016/j.amjcard.2012.10.048.

Cardiovascular Toxicity of Tyrosine Kinase Inhibitors in Patients with Chronic Myeloid Leukemia

IL Davydkin1,2, KV Naumova1, AM Osadchuk1, IA Zolotovskaya1, OE Danilova1, TYu Stepanova1, OV Tereshina1, LV Limareva3, AS Shpigel’1, TP Kuz’mina1

1 Samara State Medical University, 89 Chapaevskaya str., Samara, Russian Federation, 443099

2 SamGMU Research Institute of Hematology, Transfusiology and Intensive Care, 89 Chapaevskaya str., Samara, Russian Federation, 443099

3 SamGMU Institute of Experimental Medicine and Biotechnology, 89 Chapaevskaya str., Samara, Russian Federation, 443099

For correspondence: Kseniya Viktorovna Naumova, 89 Chapaevskaya str., Samara, Russian Federation, 443099; Tel.: +7(905)303-12-08; e-mail: senechka.naumova@rambler.ru

For citation: Davydkin IL, Naumova KV, Osadchuk AM, et al. Cardiovascular Toxicity of Tyrosine Kinase Inhibitors in Patients with Chronic Myeloid Leukemia. Clinical oncohematology. 2018;11(4):378–87.

DOI: 10.21320/2500-2139-2018-11-4-378-387


ABSTRACT

In the present review the cardiovascular complications in patients with chronic myeloid leukemia (CML) receiving tyrosine kinase inhibitors (TKI) are discussed. It covers current views on pathogenesis of TKI cardiovascular toxicity. The pathophysiology of cardiovascular diseases (CVD) is considered as a part of the so-called pathophysiological continuum, i.e. a complex of processes developing at the molecular and cellular levels before clinical symptoms of the above diseases occur. Cardiovascular toxicity of certain TKIs can contribute to progression of pathophysiological processes in CML patients. The study of mechanisms underlying cardiovascular complications of TKI-based therapy is essential for evaluating the risks of their development in each patient. Identification of CVD predictors during TKI-based therapy can allow to elaborate a scheme for cardiovascular monitoring and safe patient management under consideration of individual risks and to avoid severe life-threatening complications.

Keywords: chronic myeloid leukemia, tyrosine kinase inhibitors, adverse effects, cardiotoxicity, cardiovascular events.

Received: May 14, 2018

Accepted: August 29, 2018

Read in PDF 


REFERENCES

  1. Elliott P. Pathogenesis of cardiotoxicity induced by anthracyclines. Sem Oncol. 2006;33:2–7. doi: 10.1053/j.seminoncol.2006.04.020.

  2. Tokarska-Schlattner M, Zaugg M, Zuppinger C, et al. New insights into doxorubicin-induced cardiotoxicity: the critical role of cellular energetics. J Mol Cell Cardiol. 2006;41(3):389–405. doi: 10.1016/j.yjmcc.2006.06.009.

  3. Mellor HR, Bell AR, Valentin JP, Roberts RRA. Cardiotoxicity associated with targeting kinase pathways in cancer. Toxicol Sci. 2011;120(1):14–32. doi: 10.1093/toxsci/kfq378.

  4. Orphanos GS, Ioannidis GN, Ardavanis AG. Cardiotoxicity induced by tyrosine kinase inhibitors. Acta 2009;48(7):964–70. doi: 10.1080/02841860903229124.

  5. Baccarani M, Deininger MW, Rosti G, et European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–84. doi: 10.1182/blood-2013-05-501569.

  6. Moslehi JJ, Deininger M. Tyrosine kinase inhibitor-associated cardiovascular toxicity in chronic myeloid leukemia. J Clin Oncol. 2015;33(35):4210–8. doi: 10.1200/jco.2015.62.4718.

  7. Anhel N, Delgado DH, Lipton JH. Cardiovascular toxicities of BCR-ABL tyrosine kinase inhibitors in chronic myeloid leukemia: preventive strategies and cardiovascular surveillance. Vasc Health Risk Manage. 2017;13:293–303. doi: 10.2147/vhrm.s108874.

  8. Rix U, Hantschel O, Durnberger G, et al. Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood. 2007;110(12):4055–63. doi: 10.1182/blood-2007-07-102061.

  9. Туркина А.Г., Челышева Е.Ю. Стратегия терапии хронического миелолейкоза: возможности и перспективы. Терапевтический архив. 2013;85(7):4–9.

    [Turkina AG, Chelysheva EYu. Therapeutic strategy for chronic myeloid leukemia: possibilities and prospects. Terapevticheskii arkhiv. 2013;85(7):4–9. (In Russ)]

  10. Laneuville P. When to Stop Tyrosine Kinase Inhibitors for the Treatment of Chronic Myeloid Leukemia. Curr Treat Opt 2018;19(3):15. doi: 10.1007/s11864-018-0532-2.

  11. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. Blood. 2016;128(1):17–23. doi: 10.1182/blood-2016-01-694265.

  12. Etienne G, Guilhot J, Rea D, et al. Long-Term Follow-Up of the French Stop Imatinib (STIM1) Study in Patients With Chronic Myeloid Leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/JCO.2016.68.2914.

  13. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2016;129(7):846–54. doi: 10.1182/blood-2016-09-742205.

  14. Hehlmann R. Research in the heart of hematology: chronic myeloid leukemia 2017. Haematologica. 2017;102(3):418–21. doi: 10.3324/haematol.2016.159848.

  15. Куликов С.М., Виноградова О.Ю., Челышева Е.Ю. и др. Заболеваемость хроническим миелолейкозом в 6регионах России по данным популяционного исследования 2009–2012 гг. Терапевтический архив. 2014;86(7):24–30.

    [Kulikov SM, Vinogradova OYu, Chelysheva EYu, et al. Incidence of chronic myeloid leukemia in 6 regions of Russia according to the data of the 2009–2012 population-based study. Terapevticheskii arkhiv. 2014;86(7):24–30. (In Russ)]

  16. Рабочая группа по онкологическим заболеваниям и сердечно-сосудистой токсичности Европейского общества кардиологов (ЕОК). Меморандум ESC по лечению онкологических заболеваний и сердечно-сосудистой токсичности, разработанный под эгидой комитета по практике ESC Российский кардиологический журнал. 2017;3(143):105–39. doi: 10.15829/1560-4071-2017-3-105-139.

    [The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). 2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC committee for practice guidelines. Russian Journal of Cardiology. 2017;3(143):105–39. doi: 10.15829/1560-4071-2017-3-105-139. (In Russ)]

  17. Steegmann JL, Baccarani M, Breccia M, et al. European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia. 2016;30(8):1648–71. doi: 10.1038/leu.2016.104.

  18. Kerkela R, Grazette L, Yacobi R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med. 2006;12(8):908–16. doi: 10.1038/nm1446.

  19. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/nejmoa1609324.

  20. Gambacorti-Passerini С, Cortes JE, Lipton JH, et al. Safety of bosutinib versus imatinib in the phase 3 BELA trial in newly diagnosed chronic phase chronic myeloid leukemia. Am J Hematol. 2014;89(10):947–53. doi: 1002/ajh.23788.

  21. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Bosutinib Versus Imatinib for Newly Diagnosed Chronic Myeloid Leukemia: Results From the Randomized BFORE Trial. J Clin Oncol. 2018;36(3):231–9. doi: 10.1200/jco.2017.74.7162.

  22. Cortes JE, Khoury HJ, Kantarjian H, et al. Long-term evaluation of cardiac and vascular toxicity in patients with Philadelphia chromosome-positive leukemias treated with bosutinib. Am J Hematol. 2016;91(6): 606–16. doi: 10.1002/ajh.24360.

  23. Montani D, Bergot E, Gunther S, et al. Pulmonary arterial hypertension in patients treated by dasatinib. Circulation. 2012;125(17):2128–37. doi: 10.1161/circulationaha.111.079921.

  24. Cortes JE, Saglio G, Kantarjian HM, et al. Final 5-year study results of DASISION: the dasatinib versus imatinib study in treatment-naive chronic myeloid leukemia patients trial. J Clin Oncol. 2016;34(20):2333–40. doi: 10.1200/jco.2015.64.8899.

  25. Shah NP, Rousselot P, Schiffer C, et al. Dasatinib in imatinib-resistant or -intolerant chronic-phase, chronic myeloid leukemia patients: 7-year follow-up of study CA180-034. Am J Hematol. 2016;91(9):869–74. doi: 10.1002/ajh.24423.

  26. Jeon Y-W, Lee S-E, Kim S-H, et al. Six-Year Follow-Up Of Dasatinib-Related Pulmonary Arterial Hypertension (PAH) For Chronic Myeloid Leukemia In Single Center. Blood. 2013;122(21):4017.

  27. Kong JH, Jeon Y-W, Lee S-E, et al. Long-Term Assessment of Dasatinib-Induced Pulmonary Arterial Hypertension in Chronic Myeloid Leukemia. Blood. 2014;124(21):5535.

  28. Рабочая группа по диагностике и лечению легочной гипертензии Европейского общества кардиологов (ESC) и Европейского общества пульмонологов (ERS). Рекомендации ESC/ERS по диагностике и лечению легочной гипертензии 2015. Российский кардиологический журнал. 2016;5(133):5–64. doi: 10.15829/1560-4071-2016-5-5-64.

    [The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS). 2015 ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension. Russian Journal of Cardiology. 2016;5(133):5–64. doi: 10.15829/1560-4071-2016-5-5-64. (In Russ)]

  29. Aichberger KJ, Herndlhofer S, Schernthaner G-H, et al. Progressive peripheral arterial occlusive disease and other vascular events during nilotinib therapy in CML. Am J Hematol. 2011;86(7):533–9. doi: 10.1002/ajh.22037.

  30. Le Coutre P, Rea D, Abruzzese E, et al. Severe peripheral arterial disease during nilotinib therapy. J Nat Cancer Inst. 2011;103(17):1347–8. doi: 10.1093/jnci/djr292.

  31. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.

  32. Cortes JE, Kantarjian H, Shah NP, et al. Ponatinib in refractory Philadelphia chromosome-positive leukemias. N Engl J Med. 2012;367(22):2075–88. doi: 10.1056/nejmoa1205127.

  33. Cortes JE, Kim D-W, Pinilla-Ibarz J. Ponatinib efficacy and safety in Philadelphia chromosome–positive leukemia: Final 5-year results of the phase 2 PACE trial. Blood. 2018:blood-2016-09-739086. doi: 10.1182/blood-2016-09-739086.

  34. Lipton JH, Chuah C, Guerci-Bresler A, et al. Ponatinib versus imatinib for newly diagnosed chronic myeloid leukaemia: an international, randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17(5):612–21. doi: 10.1016/s1470-2045(16)00080-2.

  35. Иклусиг® (инструкция по медицинскому применению). Австралия: Ariad Pharmaceuticals. Доступно по: http://lechimvizraile.ru/articles/iklusig_instruktsiya_po_primeneniyu/ Ссылка активна на 7.06.2018.

    [Iclusig® (package insert). Australia: Ariad Pharmaceuticals. Available from: http://lechimvizraile.ru/articles/iklusig_instruktsiya_po_primeneniyu/ (accessed 7.06.2018) (In Russ)]

  36. Dorer DJ, Knickerbocker RK, Baccarani M, et al. Impact of dose intensity of ponatinib on selected adverse events: multivariate analyses from a pooled population of clinical trial patients. Leuk Res. 2016;48:84–91. doi: 10.1016/j.leukres.2016.07.007.

  37. Dahlen T, Edgren G, Lambe M, et al. Cardiovascular events associated with use of tyrosine kinase inhibitors in chronic myeloid leukemia: a population-based cohort study. Ann Int Med. 2016;165(3):161–6. doi: 10.7326/m15-2306.

  38. Ross DM, Arthur C, Burbury K, et al. Chronic myeloid leukaemia and tyrosine kinase inhibitor therapy: assessment and management of cardiovascular risk factors. Int Med J. 2018;48(Suppl 2):5–13. doi: 10.1111/imj.13716.

  39. Douxfils J, Haguet H, Mullier F, et al. Association Between BCR-ABL Tyrosine Kinase Inhibitors for Chronic Myeloid Leukemia and Cardiovascular Events, Major Molecular Response, and Overall Survival A Systematic Review and Meta-analysis. JAMA Oncol. 2016;2(5):625–32. doi: 10.1001/jamaoncol.2015.5932.

  40. Haguet H, Douxfils J, Mullier F, et al. Risk of arterial and venous occlusive events in chronic myeloid leukemia patients treated with new generation BCR-ABL tyrosine kinase inhibitors: a systematic review and meta-analysis. Exp Opin Drug Safety. 2017;16(1):5–12. doi: 10.1080/14740338.2017.1261824.

  41. Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer. 2009;9(1):28–39. doi: 10.1038/nrc2559.

  42. Gschwind A, Fischer OM, Ullrich A. The discovery of receptor tyrosine kinases: targets for cancer therapy. Nat Rev Cancer. 2004;4(5):361–70. doi: 10.1038/nrc1360.

  43. Zuppinger C, Suter TM. Cancer therapy-associated cardiotoxicity and signaling in the myocardium. J Cardiovasc Pharmacol. 2010;56(2):141–6. doi: 10.1097/fjc.0b013e3181e0f89a.

  44. Зейфман А.А., Челышева Е.Ю., Туркина А.Г., Чилов Г.Г. Роль селективности ингибиторов тирозинкиназ в развитии побочных эффектов при терапии хронического миелолейкоза. Клиническая онкогематология. 2014;7(1):16–27.

    [Zeyfman AA, Chelysheva YeYu, Turkina AG, Chilov GG. Role of tyrosine-kinase inhibitor selectivity in development of adverse effects during treatment of chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2014;7(1):16–27 (In Russ)]

  45. Bellinger AM, Arteaga CL, Force T, et al. Cardio-oncology. 2015;132(23):2248–58. doi: 10.1161/circulationaha.115.010484.

  46. Han MS, Chung KW, Cheon HG, et al. Imatinib mesylate reduces endoplasmic reticulum stress and induces remission of diabetes in db/db mice. 2009;58(2):329–36. doi: 10.2337/db08-0080.

  47. Iurlo A, Orsi E, Cattaneo D, et al. Effects of first- and second-generation tyrosine kinase inhibitor therapy on glucose and lipid metabolism in chronic myeloid leukemia patients: a real clinical problem? Oncotarget. 2015;6(32):33944–51. doi: 10.18632/oncotarget.5580.

  48. Alhawiti N, Burbury KL, Kwa FA, et al. The tyrosine kinase inhibitor, nilotinib potentiates a prothrombotic state. Thromb Res. 2016;145:54–64. doi: 10.1016/j.thromres.2016.07.019.

  49. Albrecht-Schgoer K, Huber K, Grebien F, et al. Nilotinib exerts direct proatherogenic and anti-angiogenic effects on vascular endothelial cells: a potential explanation for drug-induced vasculopathy in CML. Blood. 2013;122(21):257.

  50. Bair SM, Choueiri TK, Moslehi J. Cardiovascular complications associated with novel angiogenesis inhibitors: emerging evidence and evolving perspectives. Trends Cardiovasc Med. 2013;23(4):104–13. doi: 1016/j.tcm.2012.09.008.

  51. Садыкова Д.И. Современные подходы к диагностике и лечению легочной гипертензии. Практическая медицина. 2012;7(62):21–6.

    [Sadykova DI. Modern approaches to diagnostic and treatment of pulmonary hypertension. Prakticheskaya meditsina. 2012;7(62):21–6. (In Russ)]

  52. Godinas L, Guingabert C, Seferian A, et al. Tyrosine kinase inhibitors in pulmonary arterial hypertension: a double-edge sword? Semin Respir Crit Care Med. 2013;34(5):714–24. doi: 10.1055/s-0033-1356494.

  53. Morello F, Perino A, Hirsch E. Phosphoinositide 3-kinase signaling in the vascular system. Cardiovasc Res. 2009;82(2):261–71. doi: 10.1093/cvr/cvn325.

  54. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med. 2003;349(5):427–34. doi: 10.1056/nejmoa021491.

  55. Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer. 2007;7(6):475–85. doi: 10.1038/nrc2152.

  56. Dhaun N, Goddard J, Webb DJ. The endothelin system and its antagonism in chronic kidney disease. J Am Soc Nephrol. 2006;17(4):943–55. doi: 10.1681/asn.2005121256.

  57. Amiri F, Virdis A, Neves MF, et al. Endothelium-restricted overexpression of human endothelin-1 causes vascular remodeling and endothelial dysfunction. Circulation. 2004;110(15):2233–40. doi: 10.1161/01.cir.0000144462.08345.b9.

  58. Lankhorst S, Baelde HJ, Kappers MH, et al. Greater sensitivity of blood pressure than renal toxicity to tyrosine kinase receptor inhibition with sunitinib. Hypertension. 2015;66(3):543–9. doi: 10.1161/hypertensionaha.115.05435.

  59. Kostos L, Burbury K, Srivastava G, Prince HM. Gastrointestinal bleeding in a chronic myeloid leukaemia patient precipitated by dasatinib-induced platelet dysfunction: case report. Platelets. 2015;26(8):809–11. doi: 10.3109/09537104.2015.1049138.

  60. Quintas-Cardama A, Kantarjian H, Ravandi F, et al. Bleeding diasthesis in patients with chronic myelogenous leukaemia receiving dasatinib therapy. Cancer. 2009;115(11):2482–90. doi: 10.1002/cncr.24257.

  61. Quintas-Cardama A, Kantarjian H, O’Brien S, et al. Pleural effusion in patients with chronic myelogenous leukemia treated with dasatinib after imatinib failure. J Clin Oncol. 2007;25(25):3908–14. doi: 10.1200/jco.2007.12.0329.

  62. Poredos P, Jug B. The prevalence of peripheral arterial disease in high risk subject sand coronary or cerebrovascular patients. Angiology. 2007;58(3):309–15. doi: 10.1177/0003319707302494.

  63. Steg Ph, Bhatt DL, Wilson PWF, et al. One-year cardiovascular event rates in outpatients with atherothrombosis. JAMA. 2007;297(11):1197. doi: 10.1001/jama.297.11.1197.

  64. Giles FJ, le Coutre PD, Pinilla-Ibarz J, et al. Nilotinib in imatinib-resistant or imatinib-intolerant patients with chronic myeloid leukemia in chronic phase: 48-month follow-up results of a phase II study. Leukemia. 2013;27(1):107–12. doi: 10.1038/leu.2012.181.

  65. Agostino NM, Chinchilli VM, Lynch CJ, et al. Effect of the tyrosine kinase inhibitors (sunitinib, sorafenib, dasatinib, and imatinib) on blood glucose levels in diabetic and nondiabeticpatients in general clinical practice. J Oncol Pharm Pract. 2010;17(3):197–202. doi: 10.1177/1078155210378913.

  66. Lassila M, Allen TJ, Cao Z, et al. Imatinib attenuates diabetes-associated atherosclerosis. Arterioscler Thromb Vasc Biol. 2004;24(5):935–42. doi: 10.1161/01.atv.0000124105.39900.db.

  67. Hoffmann VS, Baccarani M, Hasford J, et al. The EUTOS population based registry: incidence and clinical characteristics of 2904 CML patients in 20 European Countries. 2015;29(6):1336–43. doi: 10.1038/leu.2015.73.

  68. Dzau VJ, Antman EM, Black HR, et al. The cardiovascular disease continuum: validated clinical evidence of improved patient outcomes. Part I: pathophysiology and clinical trial evidence risk factors through stable coronary artery disease. Circulation. 2006;114(25):2850–70. doi: 10.1161/circulationaha.106.655688.

  69. Daher IN, Daigle TR, Bhatia N, Durand J-B. The Prevention of Cardiovascular Disease In Cancer Survivors. Tex Heart Inst J. 2012;39(2):190–8.

  70. Brown S-A, Nhola L, Herrmann J. Cardiovascular toxicities of small molecule tyrosine kinase inhibitors: an opportunity for systems-based approaches. Clin Pharmacol Ther. 2016;101(1):65–80. doi: 10.1002/cpt.552.

  71. Kim TD, Rea D, Schwarz M, et al. Peripheral artery occlusive disease in chronic phase chronic myeloid leukemia patients treated with nilotinib or imatinib. 2013;27(6):1316–21. doi: 10.1038/leu.2013.70.

  72. Cortes JE, Kim D-W, Pinilla-Ibarz J, et al. A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N Engl J Med. 2013;369(19):1783–96. doi: 10.1056/nejmoa1306494.

  73. Гусарова Г.А., Туркина А.Г. Артериальные события у больных хроническим миелолейкозом, получающих терапию ингибиторами тирозинкиназ 2-го поколения. Клиническая онкогематология. 2016;9(4):474–84. doi: 10.21320/2500-2139-2016-9-4-474-484.

    [Gusarova GA, Turkina AG. Arterial Events in Patients with Chronic Myeloid Leukemia Receiving Treatment with Second Generation Tyrosine Kinase Inhibitors. Clinical oncohematology. 2016;9(4):474–84. doi: 10.21320/2500-2139-2016-9-4-474-484. (In Russ)]

  74. Li W, Croce K, Steensma DP, et al. Vascular and metabolic implications of novel targeted cancer therapies. J Am Coll Cardiol. 2015;66(10):1160–78. doi: 10.1016/j.jacc.2015.07.025.

  75. Conroy RM, Pyorala K, Fitzgerald AP, et al. Estimation of ten-year risk of fatal cardiovascular disease in Europe: the SCORE project. Eur Heart J. 2003;24(11):987–1003. doi: 10.1016/s0195-668x(03)00114-3.

  76. Gerhard-Herman MD, Gornik HL, Barrett C, et al. 2016 AHA/ACC guideline on the management of patients with lower extremity peripheral artery disease: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines. J Am Coll Cardiol. 2017;69(11):e71–e126. doi: 10.1016/j.jacc.2016.11.007.

  77. Овсянникова Е.Г., Попов Е.А., Давыдкин И.Л. и др. Современные аспекты диагностики, прогнозирования и лечения хронического миелолейкоза. Астраханский медицинский журнал. 2015;10(3):27–44.

    [Ovsyannikova EG, Popov EA, Davydkin IL, et al. Modern aspects of diagnosis, prognosis and treatment of chronic myeloid leukemia. Astrakhanskii meditsinskii zhurnal. 2015;10(3):27–44. (In Russ)]

  78. Jessup M, Abraham WT, Casey DE, et al. 2009 ACCF/AHA Guidelines for the Diagnosis and Management of heart failure in adults: a report of the American College of cardiology foundation/American heart association task force on practice guidelines: developed in collaboration with the International Society for heart and lung transplantation. Circulation. 2009;119(14):1977–2016. doi: 10.1161/circulationaha.109.192064.

  79. Granados-Principal S, Quiles JL, Ramirez-Tortosa CL, et al. New advances in molecular mechanisms and the prevention of adriamycin toxicity by antioxidant nutrients. Food Chem Toxicol. 2010;48(6):1425–38. doi: 10.1016/j.fct.2010.04.007.

  80. Wouters KA, Kremer LC, Miller TL, et al. Protecting against anthracycline-induced myocardial damage: a review of the most promising strategies. Br J Haematol. 2005;131(5):561–78. doi: 10.1111/j.1365-2141.2005.05759.x.

  81. Mohamed HE, El-Swefy SE, Hagar HH. The protective effect of glutathione administration on Adriamycin-induced acute cardiac toxicity in rats. Pharmacol Res. 2000;42(2):115–21. doi: 10.1006/phrs.1999.0630.

  82. van Leeuwen RWF, van Gelder T, Mathijssen RHJ, Jansman FGA. Drug–drug interactions with tyrosine-kinase inhibitors: a clinical perspective. Lancet Oncol. 2014;15(8):e315–e26. doi: 10.1016/s1470-2045(13)70579-5.

  83. Haouala A, Widmer N, Duchosal MA, et al. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood. 2011;117(8):e75–e87. doi: 10.1182/blood-2010-07-294330.

  84. Туркина А.Г., Зарицкий А.Ю., Шуваев В.А. и др. Клинические рекомендации по диагностике и лечению хронического миелолейкоза. Клиническая онкогематология. 2017;10(3):294–316. doi: 10.21320/2500-2139-2017-10-3-294-316.

    [Turkina AG, Zaritskii AYu, Shuvaev VA, et al. Clinical Recommendations for the Diagnosis and Treatment of Chronic Myeloid Leukemia. Clinical oncohematology. 2017;10(3):294–316. doi: 10.21320/2500-2139-2017-10-3-294-316. (In Russ)]

  85. Poch MM, Sibai H, Deotare U, Lipton JH. Ponatinib in the therapy of chronic myeloid leukemia. Exp Rev Hematol. 2016;9(10):923–32. doi: 10.1080/17474086.2016.1232163.

  86. Breccia M, Pregno P, Spallarossa P, et al. Identification, prevention and management of cardiovascular risk in chronic myeloid leukaemia patients candidate to ponatinib: an expert opinion. Ann Hematol. 2016;96(4):549–58. doi: 10.1007/s00277-016-2820-x.

Biochemical Markers of Cardiotoxicity of High-Dose Chemotherapy and Autologous Hematopoietic Stem Cell Transplantation in Patients with Malignant Lymphoproliferative Disorders

VO Sarzhevskii, DS Kolesnikova, VYa Mel’nichenko

NI Pirogov National Medical and Surgical Center, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203

For correspondence: Vladislav Olegovich Sarzhevskii, PhD, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203; Tel: +7(495)603-72-18; e-mail: vladsar@pochta.ru

For citation: Sarzhevskii VO, Kolesnikova DS, Mel’nichenko VYa. Biochemical Markers of Cardiotoxicity of High-Dose Chemotherapy and Autologous Hematopoietic Stem Cell Transplantation in Patients with Malignant Lymphoproliferative Disorders. Clinical oncohematology. 2016;9(4):465–73 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-465-473


ABSTRACT

Background. High-dose chemotherapy (HDCT) with autologous hematopoietic stem cells transplantation (auto-HSCT) is an effective therapeutic option for patients with Hodgkin’s lymphoma and aggressive non-Hodgkin’s lymphomas in those cases, when the standard chemotherapy combined with the radiation therapy proves to be ineffective. The HDCT and auto-HSCT are also basic treatment options for multiple myeloma. However, toxic effects of the transplantation, including cardiotoxicity, may significantly worsen the prognosis of patients who receive this treatment.

Aim. To evaluate changes in biochemical markers of cardiotoxicity (troponin and N-terminal prohormone of brain natriuretic peptide (NT-proBNP)) in patients with malignant lymphomas (receiving HDCT and auto-HSCT).

Materials & Methods. 157 patients were enrolled in the study. The sensitivity threshold of the troponin T test was 0.1 ng/mL and troponin I 0.001 ng/mL (highly sensitive troponin). Troponin T (conventional troponin) was measured in 56 patients, troponin I was assessed in 101 patients. Serum troponin levels were evaluated before the conditioning, on D0, D+7, and D+12. The level of NT-proBNP was assessed before the conditioning, on D0 and D+12.

Results. Increased troponin T level was observed in 2 of 56 patients (3.6 %), increased troponin I level — in 27 of 101 patients (26.7 %) (< 0.01). Troponin levels were within normal limits in all patients at admission. Troponin T levels increased only on D+7. Troponin I level increased in 4 patients (4 %) on D0, in 17 patients (16.8 %) on D+7 and in 11 patients (10.9 %) on D+12. The median concentration of troponin I was 0.215 ng/mL after HDCT completion, 0.74 ng/mL on D+7 and 0.21 ng/mL on D+12. No cases of myocardial infarction were observed. NT-proBNP levels in most patients were within normal limits at admission (median level 79.2 pg/mL). The situation changed significantly after conditioning: in most patients the level was almost twice as high as the upper normal limit (medial 240.6 pg/mL). Significant differences in levels of NT-proBNP (< 0.05) were observed at comparison of data before conditioning and D0, and before conditioning and D+12.

Conclusion. The data obtained confirm a significant impact of HDCT and auto-HSCT on the cardiovascular system of patients with malignant lymphomas. Further studies and observation of the patients are needed to clarify the prognostic significance of the findings related to cardiotoxicity (in particular, congestive heart failure).


Keywords: high-dose chemotherapy, autologous hematopoietic stem cells transplantation, cardiotoxicity, troponin, NT-proBNP.

Received: June 13, 2016

Accepted: June 14, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Blay J, Gomez F, Sebban C, et al. The International Prognostic Index correlates to survival in patients with aggressive lymphoma in relapse: analysis of the PARMA trial. Parma Group. Blood. 1998;92(10):3562–8.
  2. 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.
  3. NCCN Guidelines Version 3.2016, Non-Hodgkin’s lymphomas. pр. 56, 65. [Internet] Available from: https://www.nccn.org/professionals/physician_gls/pdf/nhl.pdf (accessed 14.06.2016).
  4. NCCN Guidelines Version 2.2016, Hodgkin lymphoma. pp. 20 [Internet]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/hodgkins.pdf (accessed 14.06.2016).
  5. Cazin B, Gorin NC, Laporte JP, et al. Cardiac complications after bone marrow transplantation. A report on a series of 63 consecutive transplantations. Cancer. 1986;57(10):2061–9. doi: 10.1002/1097-0142(19860515)57:10<2061::aid-cncr2820571031>3.0.co;2-h.
  6. Murdych T, Weisdorf DJ. Serious cardiac complications during bone marrow transplantation at the University of Minnesota, 1977–1997. Bone Marrow Transplant. 2001;28(3):283–7. doi: 10.1038/sj.bmt.1703133.
  7. Majhail NS, Rizzo JD, Lee SJ, et al. Recommended screening and preventive practices for long-term survivors after hematopoietic cell transplantation. Bone Marrow Transplant. 2012;47(3):337–41. doi: 10.1038/bmt.2012.5.
  8. Chi AK, Soubani AO, White AC, et al. An update on pulmonary complications of hematopoietic stem cell transplantation. Chest. 2013;144(6):1913–22. doi: 10.1378/chest.12-1708.
  9. Chow EJ, Wong K, Lee SJ, et al. Late cardiovascular complications after hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2014;20(6):794–800. doi: 10.1016/j.bbmt.2014.02.012.
  10. Cardinale D, Sandri MT, 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. doi: 10.1016/S1062-1458(00)00186-0.
  11. Cardinale D, Sandri MT, Martinoni A, et al. Myocardial injury revealed by plasma troponin I in breast cancer treated with high-dose chemotherapy. Ann Oncol. 2002;13(5):710–5. doi: 10.1093/annonc/mdf170.
  12. Sandri MT, Cardinale D, Zorzino L, et al. Minor increases in plasma troponin I predict decreased left ventricular ejection fraction after high-dose chemotherapy. Clin Chem. 2003;49(2):248–52. doi: 10.1373/49.2.248.
  13. Auner HW, 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.
  14. Cardinale D, Sandri MT, 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. doi: 10.1161/01.cir.0000130926.51766.cc.
  15. Lipshultz SE, Rifai N, Dalton VM, et al. The effect of dexrazoxane on myocardial injury in doxorubicin-treated children with acute lymphoblastic leukemia. N Engl J Med. 2004;351(2):145–53. doi: 10.1056/nejmoa035153.
  16. Kilickap S, Barista I, Akgul E, et al. cTnT can be a useful marker for early detection of anthracycline cardiotoxicity. Ann Oncol. 2005;16(5):798–804. doi: 10.1093/annonc/mdi152.
  17. Lipshultz SE, Rifai N, Sallan SE, et al. Predictive value of cardiac troponin T in pediatric patients at risk for myocardial injury. Circulation. 1997;96(8):2641–8. doi: 10.1161/01.cir.96.8.2641.
  18. 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. doi: 10.1053/hj.1998.v136.89908.
  19. 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. doi: 10.1111/j.1600-0609.1998.tb01099.x.
  20. Snowden JA, Hill GR, 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. doi: 10.1038/sj.bmt.1702507.
  21. 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. doi: 10.1136/hrt.2007.123299.
  22. Horacek JM, 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.
  23. Masuko M, Ito M, Kurasaki T, et al. Plasma brain natriuretic peptide during myeloablative stem cell transplantation. Intern Med. 2007;46(9):551–5. doi: 10.2169/internalmedicine.46.6188.
  24. 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. doi: 10.1532/ijh97.e0620.

 

Single-Photon Emission Computed Tomography Synchronized with ECG as Method for Evaluation of Cardiotoxicity of High-Dose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation for Malignant Lymphoproliferative Disorders

VO Sarzhevskii, DS Kolesnikova, MN Vakhromeeva, VYa Melnichenko

N.I. Pirogov National Medical and Surgical Center under the Ministry of Health of the Russian Federation, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203

For correspondence: Vladislav Olegovich Sarzhevskii, PhD, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203; Tel.: +7(495)603-72-18; e-mail: vladsar@pochta.ru

For citation: Sarzhevskii VO, Kolesnikova DS, Vakhromeeva MN, Mel’nichenko VYa. Single-Photon Emission Computed Tomography Synchronized with ECG as Method for Evaluation of Cardiotoxicity of High-Dose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation for Malignant Lymphoproliferative Disorders. Clinical oncohematology. 2015;8(1):84–90 (In Russ).


ABSTRACT

Background. High-dose chemotherapy (HDC) with autologous hematopoietic stem cells transplantation (auto-HSCT) is currently widely used for the treatment of relapsed and refractory to standard chemotherapy cases of malignant lymphoproliferative disorders. Cardiac monitoring of patients treated with HDC with subsequent auto-HSCT is performed by means of ECG and Echo-CG in most cases. The method of single-photon emission computed tomography of the left ventricle (LV) synchronized with ECG (gated-SPECT) is rarely used to assess cardiotoxic effect of HDC and auto-HSCT.

Objective. To evaluate perfusion and regional myocardial function of the left ventricle (LV) in patients with malignant lymphomas receiving HDC and auto-HSCT.

Methods. The study included 69 patients (37 with Hodgkin’s lymphoma, 19 with non-Hodgkin’s lymphoma, and 13 with multiple myeloma). The median age was 36 year (range from 19 to 66 years); 40 females, 29 males. Perfusion and regional LV function at rest before the HDC and auto-HSCT (point 1) and at discharge (point 2) were assessed. Each study was performed on a double-headed rotating gamma camera Forte (Philips, USA). 740 MBq of technetium-99m-methoxyisobutylisonitrile (99mTc-MIBI) was used as a radiopharmaceutical. Semiquantitative assessment of tomoscintigrams was performed using polar diagrams (20-segment model); they were used for complex analysis of perfusion and LF myocardium function parameters.

Results. The total area of hypoperfusion, expressed as a percentage of the area of the LV myocardium, did not change significantly during treatment (> 0.05). However, the segmental analysis demonstrated a statistically significant decrease in the median uptake level of the radiopharmaceutical in 1, 2, 4, 7, 8, 10, 13, 16, 17, and 19 segments (< 0.05). The total ejection fraction (TEF) did not change (median TEF was 59.5 % at point 1 and 58 % at point 2). But it showed a statistically significant decrease in median of local systolic thickening in 2, 3, 5, 7, 8, 9, 10, 11, 12, 15, 17, 18, 19, and 20 segments of the left ventricle (< 0.05).

Conclusions. HDC and auto-HSCT significantly change perfusion and regional LV myocardial function in patients with malignant lymphomas. The changes demonstrate diffuse myocardial damage. Gated-SPECT can be considered a promising method for assessing cardiotoxicity of HDC and auto-HSCT.


Keywords: high-dose chemotherapy, autologous hematopoietic stem cells transplantation, cardiotoxicity, gated-SPECT.

Received: July 23, 2014

Accepted: November 5, 2014

Read in PDF (RUS)pdficon


REFERENCES

  1. Passweg JR, Baldomero H, Peters C, et al. Hematopoietic SCT in Europe: data and trends in 2012 with special consideration of pediatric transplantation. Bone Marrow Transplant. 2014;49(6):744–50. doi: 10.1038/bmt.2014.55.
  2. Popplewell LL, Forman SJ. Is there an upper age limit for bone marrow transplantation? Bone Marrow Transplant. 2002;29(4):277–84. doi: 10.1038/sj.bmt.1703382.
  3. Badros A, Barlogie B, Siegel E, et al. Autologous stem cell transplantation in elderly multiple myeloma patients over the age of 70 years. Br J Haematol. 2001;114(3):600–7. doi: 10.1046/j.1365-2141.2001.02976.x.
  4. Germano G, Kiat H, Kavanagh P, et al. Automatic quantification and review of ejection fraction from gated myocardial perfusion SPECT. J Nucl Med. 1995;36:2138–47.
  5. Germano G, Kavanagh P, Berman D, et al. An automatic approach to the analysis quantification and review of perfusion and function from myocardial perfusion SPECT imaging. Intern J Card Im. 1997;13(4):337–46.
  6. 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(5):1503–6. doi: 10.1016/s0360-3016(00)00807-5.
  7. Glanzmann C, Kaufmann P, Jenni R, et al. Cardiac risk after mediastinal irradiation for Hodgkin’s disease. Radiother Oncol. 1998;46(1):51–62. doi: 10.1016/s0167-8140(97)00125-4.
  8. 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.
  9. Salloum E, Jillella AP, 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. doi: 10.1002/(sici)1097-0142(19980415)82:8<1506::aid-cncr12>3.0.co;2-8.
  10. 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.
  11. Cardinale D, Sandri MT, 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. doi: 10.1161/01.cir.0000130926.51766.cc.
  12. Auner HW, 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.
  13. Pihkala J, Saarinen UM, Lundstrоm U, et al. Effects of bone marrow transplantation on myocardial function in children. Bone Marrow Transplant. 1994;13(2):149–55.
  14. 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. doi: 10.1007/s12185-008-0112-5.

Cardiotoxicity of high-dose chemotherapy and autologous hematopoietic cell transplantation in patients with hematological malignancies

V.O. Sarzhevsky, D.S. Kolesnikova, V.Ya. Mel’nichenko, and V.P. Tyurin

N.I. Pirogov National Medico-surgical Centre, RF Ministry of Health, Moscow, Russian Federation


ABSTRACT

This review presents the recent data on cardiotoxicity of high-dose chemotherapy and autologous hematopoietic cell transplantation in patients with hematological malignancies. The article includes detailed description of the methods used for cardiotoxicity assessment and clinical features of early and late cardiac complications. Also, the approaches to prevention and treatment of cardiotoxicity in this group of patients are suggested.


Keywords: cardiotoxicity, high-dose chemotherapy, autologous hematopoietic cell transplantation.

Read in PDF (RUS)pdficon


REFERENCES

  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