Efficacy Predictors of the Third-Line Tyrosine Kinase Inhibitor Therapy in Patients with Chronic Phase of Chronic Myeloid Leukemia: Results of a Multi-Center Study

EG Lomaia1, VA Shuvaev2,3, TV Chitanava1, YuD Matvienko1, IS Martynkevich2, SV Voloshin2, EV Efremova2, ES Mileeva2, MS Fominykh4, AE Kersilova3, EV Karyagina5, NV Il’ina5, NV Dorofeeva6, NV Medvedeva6, AV Klimovich6, TV Shneider7, SA Stepanova7, NF Polezhaikovskaya7, NT Siordiya1, EI Sbityakova1, NS Lazorko1, EN Tochenaya1, DV Motorin1, NA Shnalieva1, YuA Alekseeva1, DB Zammoeva1, AYu Zaritskey1

1 VA Almazov National Medical Research Center, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341

2 Russian Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya ul., Saint Petersburg, Russian Federation, 191024

3 VV Veresaev Municipal Clinical Hospital, 10 Lobnenskaya ul., Moscow, Russian Federation, 127644

4 AVA-PETER, Multispecialty Clinic “Skandinaviya”, 55A Liteinyi pr-t, Saint Petersburg, Russian Federation, 191014

5 Municipal Hospital No. 15, 4 Avangardnaya ul., Saint Petersburg, Russian Federation, 198205

6 Municipal Clinical Hospital No. 31, 3 Dinamo pr-t, Saint Petersburg, Russian Federation, 197110

7 Leningrad Regional Clinical Hospital, 45 korp. 2A Lunacharskogo pr-t, Saint Petersburg, Russian Federation, 194291

For correspondence: Tamara Vangelevna Chitanava, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341; e-mail: chitanava.tamara@yandex.ru

For citation: Lomaia EG, Shuvaev VA, Chitanava TV, et al. Efficacy Predictors of the Third-Line Tyrosine Kinase Inhibitor Therapy in Patients with Chronic Phase of Chronic Myeloid Leukemia: Results of a Multi-Center Study. Clinical oncohematology. 2022;15(3):271–81. (In Russ).

DOI: 10.21320/2500-2139-2022-15-3-271-281


ABSTRACT

Background. The introduction of tyrosine kinase inhibitors (TKIs) into real-world clinical practice considerably improved the prognosis for patients with chronic myeloid leukemia (CML). However, during long-term follow-up, almost 1/2 and 2/3 of patients in the chronic phase (CP) discontinue TKI therapy of the first or second line, respectively. According to the Russian and International clinical guidelines, the third-line therapy should include allogeneic hematopoietic stem cell transplantation (allo-HSCT). And yet, some patients on the third-line therapy achieve and sustain optimal response on long-term TKI administration. Up to now, no clear-cut prognostic factors of TKI efficacy in the third-line therapy have been identified. This creates a challenge for treatment decision making after the failures of two lines of TKI therapy.

Aim. To assess the efficacy of the third-line TKI therapy in real-world clinical practice and to identify the factors affecting the long-term therapy outcomes in CML-CP.

Materials & Methods. The retrospective study enrolled 73 CML-CP patients aged ≥ 18 years, treated with TKIs in the third-line at 5 specialized institutions in Saint Petersburg and Leningrad Region. Among the patients there were 26 men (35 %). The median age of the patients was 51 years (range 25–88 years).

Results. With the median (range) third-line TKI therapy duration of 14 (1–120) months, the rate of complete cytogenetic response (CCR) was 30 % (n = 22) in the total cohort. The median time before achieving CCR was 9 (4–25) months. With the median follow-up time from the beginning of third-line TKI therapy till the last visit of 25 (3–136) months, progression to accelerated phase or blast crisis was observed only in 13 (17 %) out of 73 patients. Death was reported in 26 % (n = 19) of cases, among them 5 patients whose death was not CML-associated. At the last visit, 13/73 (18 %) patients were still on third-line TKI therapy. Direct and long-term therapy outcomes, including achievement of CCR and assessment of overall and progression-free survivals, were significantly better in patients with any cytogenetic response (CR) than in those without it or without complete hematologic response.

Conclusion. The implementation of TKIs in the third-line CML-CP therapy seems to be suitable for patients with at least some CR, especially if an optimal donor of hematopoietic stem cells is unavailable or if the risk of severe allo-HSCT complications is too high.

Keywords: chronic myeloid leukemia, chronic phase, complete cytogenetic response, tyrosine kinase inhibitors, third-line therapy, allogeneic hematopoietic stem cell transplantation, minimal residual disease.

Received: April 7, 2022

Accepted: June 20, 2022

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Hochhaus A, Baccarani M, Silver RT, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 2020;34(4):966–84. doi: 10.1038/s41375-020-0776-2.
  2. Baccarani M, Deininger MW, Rosti G, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–84. doi: 10.1182/blood-2013-05-501569.
  3. Клинические рекомендации [Хронический миелолейкоз]: стандарты ведения больных для врачей [электронный документ]. Под ред. А.Г. Туркиной и др. М., 2020. Доступно по: https://npngo.ru/biblioteka/klinicheskie_rekomendatsii__2019_god_ Ссылка активна на 7.04.2022.
    [Clinical guidelines [Chronic myeloid leukemia]: patient management standards for physicians. (Internet) Available from: https://npngo.ru/biblioteka/klinicheskie_rekomendatsii__2019_god_ (accessed 04.2022). (In Russ)]
  4. 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)]
  5. 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.
  6. Stone RM, Kim DW, Kantarjian HM, et al. Dasatinib dose-optimization study in chronic phase chronic myeloid leukemia (CML-CP): three-year follow-up with dasatinib 100 mg once daily and landmark analysis of cytogenetic response and progression-free survival (PFS). J Clin Oncol. 2009;27(15_suppl):7007. doi: 10.1200/jco.2009.27.15_suppl.7007.
  7. Shah NP, Cortes JE, Schiffer CA, et al. Five-year follow-up of patients with imatinib-resistant or -intolerant chronic-phase chronic myeloid leukemia (CML-CP) receiving dasatinib. J Clin Oncol. 2011;29(15_suppl):6512. doi: 10.1200/jco.2011.29.15_suppl.6512.
  8. Kantarjian HM, Giles FJ, Bhalla KN, et al. Nilotinib is effective in patients with chronic myeloid leukemia in chronic phase after imatinib resistance or intolerance: 24-month follow-up results. Blood. 2011;117(4):1141–5. doi: 10.1182/blood-2010-03-277152.
  9. Cortes JE, Kantarjian HM, Brummendorf TH, et al. Safety and efficacy of bosutinib (SKI-606) in chronic phase Philadelphia chromosome-positive chronic myeloid leukemia patients with resistance or intolerance to imatinib. Blood. 2011;118(17):4567–76. doi: 10.1182/blood-2011-05-355594.
  10. Gambacorti-Passerini C, Cortes JE, Lipton JH, et al. Safety and efficacy of second-line bosutinib for chronic phase chronic myeloid leukemia over a five-year period: final results of a phase I/II study. Haematologica. 2018;103(8):1298–307. doi: 10.3324/haematol.2017.171249.
  11. Шуваев В.А., Виноградова О.Ю., Мартынкевич И.С. и др. Опыт и перспективы клинического применения бозутиниба у пациентов с хроническим миелолейкозом. Клиническая онкогематология. 2018;11(4):288–94. doi: 10.21320/2500-2139-2018-11-4-288-294.
    [Shuvaev VA, Vinogradova OYu, Martynkevich IS, et al. Clinical Experience and Perspectives of Bosutinib Use in Patients with Chronic Myeloid Leukemia. Clinical oncohematology. 2018;11(4):288–94. doi: 10.21320/2500-2139-2018-11-4-288-294. (In Russ)]
  12. Лазорко Н.С., Ломаиа Е.Г., Романова Е.Г. и др. Ингибиторы тирозинкиназ второго поколения и их токсичность у больных в хронической фазе хронического миелолейкоза. Клиническая онкогематология. 2015;8(3):302–8. doi: 10.21320/2500-2139-2015-8-3-302-308.
    [Lazorko NS, Lomaia EG, Romanova EG, et al. Second Generation Tyrosine Kinase Inhibitors and Their Toxicity in Treatment of Patients in Chronic Phase of Chronic Myeloid Leukemia. Clinical oncohematology. 2015;8(3):302–8. doi: 21320/2500-2139-2015-8-3-302-308. (In Russ)]
  13. Giralt SA, Arora M, Goldman JM, et al. Impact of imatinib therapy on the use of allogeneic haematopoietic progenitor cell transplantation for the treatment of chronic myeloid leukaemia. Br J Haematol. 2007;137(5):461–7. doi: 10.1111/j.1365-2141.2007.06582.x.
  14. Лазорко Н.С., Ломаиа Е.Г., Зарицкий А.Ю. и др. Результаты третьей линии таргетной терапии у больных хроническим миелолейкозом в хронической фазе при непереносимости или неэффективности двух ингибиторов тирозинкиназ. Клиническая онкогематология. 2016;9(3):352–3.
    [Lazorko NS, Lomaia EG, Zaritskey AYu, et al. The results of the third-line targeted therapy in chronic myeloid leukemia patients in chronic phase with intolerability or inefficacy of two tyrosine kinase inhibitors. Clinical oncohematology. 2016;9(3):352–3. (In Russ)]
  15. Russo Rossi A, Breccia M, Abruzzese E, et al. Outcome of 82 chronic myeloid leukemia patients treated with nilotinib or dasatinib after failure of two prior tyrosine kinase inhibitors. Haematologica. 2013;98(3):399–403. doi: 10.3324/haematol.2012.064337.
  16. Lauseker M, Hanfstein B, Haferlach C, et al. Equivalence of BCR-ABL transcript levels with complete cytogenetic remission in patients with chronic myeloid leukemia in chronic phase. J Cancer Res Clin Oncol. 2014;140(11):1965–9. doi: 10.1007/s00432-014-1746-8.
  17. Pfirrmann M, Hochhaus A, Lauseker M, et al. Recommendations to meet statistical challenges arising from endpoints beyond overall survival in clinical trials on chronic myeloid leukemia. Leukemia. 2011;25(9):1433–8. doi: 10.1038/leu.2011.116.
  18. Kantarjian H, O’Brien S, Jabbour E, et al. Impact of treatment end point definitions on perceived differences in long-term outcome with tyrosine kinase inhibitor therapy in chronic myeloid leukemia. J Clin Oncol. 2011;29(23):3173–8. doi: 10.1200/JCO.2010.33.4169.
  19. Ongoren S, Eskazan AE, Suzan V, et al. Third-line treatment with second-generation tyrosine kinase inhibitors (dasatinib or nilotinib) in patients with chronic myeloid leukemia after two prior TKIs: real-life data on a single center experience along with the review of the literature. Hematology. 2018;23(4):212–20. doi: 10.1080/10245332.2017.1385193.
  20. Ibrahim AR, Paliompeis C, Bua M, et al. Efficacy of tyrosine kinase inhibitors (TKIs) as third-line therapy in patients with chronic myeloid leukemia in chronic phase who have failed 2 prior lines of TKI therapy. Blood. 2010;116(25):5497–500. doi: 10.1182/blood-2010-06-291922.
  21. Bosi GR, Fogliatto LM, Costa TEV, et al. What happens to intolerant, relapsed or refractory chronic myeloid leukemia patients without access to clinical trials? Hematol Transfus Cell Ther. 2019;41(3):222–8. doi: 10.1016/j.htct.2018.11.005.
  22. Garg RJ, Kantarjian H, O’Brien S, et al. The use of nilotinib or dasatinib after failure to 2 prior tyrosine kinase inhibitors: long-term follow-up. Blood. 2009;114(20):4361–8. doi: 10.1182/blood-2009-05-221531.
  23. Ribeiro BF, Miranda EC, Albuquerque DM, et al. Treatment with dasatinib or nilotinib in chronic myeloid leukemia patients who failed to respond to two previously administered tyrosine kinase inhibitors—a single center experience. Clinics (Sao Paulo). 2015;70(8):550–5. doi: 10.6061/clinics/2015(08)04.
  24. Cortes JE, Khoury HJ, Kantarjian HM, et al. Long-term bosutinib for chronic phase chronic myeloid leukemia after failure of imatinib plus dasatinib and/or nilotinib. Am J Hematol. 2016;91(12):1206–14. doi: 10.1002/ajh.24536.
  25. Garcia-Gutierrez V, Milojkovic D, Hernandez-Boluda JC, et al. Safety and efficacy of bosutinib in fourth-line therapy of chronic myeloid leukemia patients. Ann Hematol. 2019;98(2):321–30. doi: 10.1007/s00277-018-3507-2.
  26. 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.
  27. Shah NP, Guilhot F, Cortes JE, et al. Long-term outcome with dasatinib after imatinib failure in chronic-phase chronic myeloid leukemia: follow-up of a phase 3 study. Blood. 2014;123(15):2317–24. doi: 10.1182/blood-2013-10-532341.
  28. Milojkovic D, Nicholson E, Apperley JF, et al. Early prediction of success or failure of treatment with second-generation tyrosine kinase inhibitors in patients with chronic myeloid leukemia. Haematologica. 2010;95(2):224–31. doi: 10.3324/haematol.2009.012781.
  29. Tan J, Xue M, Pan J, et al. Responses to Dasatinib as a Second- and Third-Line Tyrosine Kinase Inhibitor in Chronic Phase Chronic Myeloid Leukaemia Patients. Acta Haematol. 2019;142(2):79–86. doi: 10.1159/000495335.
  30. Jabbour E, Bahceci E, Zhu C, et al. Predictors of Long-Term Cytogenetic Response Following Dasatinib Therapy of Patients with Chronic-Phase Chronic Myeloid Leukemia (CML-CP). Blood. 2009;114(22):3296. doi: 10.1182/blood.V114.22.3296.3296.
  31. Soverini S, Hochhaus A, Nicolini FE, et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood. 2011;118(5):1208–15. doi: 10.1182/blood-2010-12-326405.

Risk Factors for Rehospitalizations after Allogeneic Hematopoietic Stem Cell Transplantation

MYu Drokov, AA Dmitrova, LA Kuzmina, VA Vasil’eva, ED Mikhaltsova, OM Koroleva, EV Usikova, EN Parovichnikova, VG Savchenko

National Research Center for Hematology, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Mikhail Yur’evich Drokov, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)614-90-42; e-mail: mdrokov@gmail.com

For citation: Drokov MYu, Dmitrova AA, Kuzmina LA, et al. Risk Factors for Rehospitalization after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2020;13(1):89–94 (In Russ).

DOI: 10.21320/2500-2139-2020-13-1-89-94


ABSTRACT

Aim. To assess the rehospitalization data of patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT), to determine possible risk factors for rehospitalization, and to work out a strategy of post-transplantation follow-up for this category of patients.

Materials & Methods. From 2009 to 2019 at the National Research Center for Hematology 418 patients received allo-HSCT. The final analysis included 374 patients who were discharged from hospital after allo-HSCT. The reasons for rehospitalizations of patients with allo-HSCT within 30 days after their hospital discharge were subjected to analysis. Independent risk factors for rehospitalizations were identified by the Cox model. Risk density was visually estimated within 365 days after hospital discharge with the purpose of working out the optimal strategy of post-transplantation follow-up for this category of patients.

Results. The probability of rehospitalization within 30 days after hospital discharge was 30.7 % for all patients with allo-HSCT. The data assessment showed that the majority of rehospitalizations (55.7 %) were associated with infectious complications. Acute graft-versus-host disease (GVHD) during the first hospitalization, i.e. immediately after allo-HSCT during the hospital stay, proved to enhance the probability of rehospitalizations within 30 days after hospital discharge by 1.7 times compared with the patients without acute GVHD.

Conclusion. The leading cause of rehospitalizations of patients with allo-HSCT within 30 days after hospital discharge was acute GVHD which occurred before, i.e. during the first hospital stay. The data obtained demonstrate the necessity of close monitoring of a patient’s status within the first 120 days after discharge from the hospital where allo-HSCT was performed.

Keywords: allogeneic hematopoietic stem cell transplantation, rehospitalizations, graft-versus-host disease.

Received: July 16, 2019

Accepted: December 17, 2019

Read in PDF


REFERENCES

  1. Morello E, Malagola M, Bernardi S, et al. The role of allogeneic hematopoietic stem cell transplantation in the four P medicine era. Blood Res. 2018;53(1):3–6. doi: 10.5045/br.2018.53.1.3.

  2. Sureda A, Bader P, Cesaro S, et al. Indications for allo- and auto-SCT for haematological diseases, solid tumours and immune disorders: current practice in Europe, 2015. Bone Marrow Transplant. 2015;50(8):1037–56. doi: 10.1038/bmt.2015.6.

  3. Majhail NS, Farnia SH, Carpenter PA, et al. Indications for Autologous and Allogeneic Hematopoietic Cell Transplantation: Guidelines from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2015;21(11):1863–9. doi: 10.1016/j.bbmt.2015.07.032.

  4. Passweg JR, Baldomero H, Basak GW, et al. The EBMT activity survey report 2017: a focus on allogeneic HCT for nonmalignant indications and on the use of non-HCT cell therapies. Bone Marrow Transplant. 2019;54(10):1575–85. doi: 10.1038/s41409-019-0465-9.

  5. Broder MS, Quock TP, Chang E, et al. The Cost of Hematopoietic Stem-Cell Transplantation in the United States. Am Heal Drug Benef. 2017;10(7):366–74.

  6. Моисеев И.С., Галанкин Т.Л., Доценко А.А. и др. Фармакоэкономика различных методов лечения стероид-рефрактерной реакции «трансплантат против хозяина»: анализ результатов лечения в одноцентровом исследовании. Ученые записки Санкт-Петербургского государственного медицинского университета им. И.П. Павлова. 2018;25(1):35–44. doi: 10.24884/1607-4181-2018-25-1-35-44.

    [Moiseev IS, Galankin TL, Dotsenko AA, et al. Pharmacoeconomic analysis of different methods for the treatment of steroid-refractory graft-verus-host disease: single-center study. The Scientific Notes of the I.P. Pavlov St. Petersburg State Medical University. 2018;25(1):35–44. doi: 10.24884/1607-4181-2018-25-1-35-44. (In Russ)]

  7. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among Patients in the Medicare Fee-for-Service Program. N Engl J Med. 2009;360(14):1418–28. doi: 10.1056/NEJMsa0803563.

  8. McIlvennan CK, Eapen ZJ, Allen LA. Hospital readmissions reduction program. Circulation. 2015;131(20):1796–803. doi: 10.1161/CIRCULATIONAHA.114.010270.

  9. Bejanyan N, Bolwell BJ, Lazaryan A, et al. Risk Factors for 30-Day Hospital Readmission following Myeloablative Allogeneic Hematopoietic Cell Transplantation (allo-HCT). Biol Blood Marrow Transplant. 2012;18(6):874–80. doi: 10.1016/J.BBMT.2011.10.032.

  10. Kerbauy MN, Kerbauy LN, Esteves I, et al. Hospital Length of Stay and Impact of Readmission in the First 100 Days of Allogeneic Stem Cell Transplantation: Comparison among Alternative Donor in Pediatric and Adult Population. Biol Blood Marrow Transplant. 2018;24(3):S337–8. doi: 10.1016/j.bbmt.2017.12.401.

  11. Hess KR, Serachitopol DM, Brown BW. Hazard function estimators: a simulation study. Stat Med. 1999;18(22):3075–88. doi: 10.1002/(SICI)1097-0258(19991130)18:22<3075::AID-SIM244>3.0.CO;2–6.

  12. Brissot E, Rialland F, Cahu X, et al. Improvement of overall survival after allogeneic hematopoietic stem cell transplantation for children and adolescents: a three-decade experience of a single institution. Bone Marrow Transplant. 2016;51(2):267–72. doi: 10.1038/bmt.2015.250.

  13. Richardson PG, Grupp SA, Pagliuca A, et al. Defibrotide for the treatment of hepatic veno-occlusive disease/sinusoidal obstruction syndrome with multiorgan failure. Int J Hematol Oncol. 2017;6(3):75–93. doi: 10.2217/ijh-2017-0015.

  14. Richardson PG, Antin JH, Giralt SA, et al. Adults Receiving Defibrotide for the Treatment of Hepatic Veno-Occlusive Disease/Sinusoidal Obstruction Syndrome (VOD/SOS) after Hematopoietic Stem Cell Transplantation (HSCT): Final Results from the Expanded-Access Program (T-IND). Biol Blood Marrow Transplant. 2018;24(3):S300–1. doi: 10.1016/j.bbmt.2017.12.344.

  15. Richardson PG, Triplett BM, Ho VT, et al. Defibrotide sodium for the treatment of hepatic veno-occlusive disease/sinusoidal obstruction syndrome. Expert Rev Clin Pharmacol. 2018;11(2):113–24. doi: 10.1080/17512433.2018.1421943.

  16. Richardson P, Aggarwal S, Topaloglu O, et al. Systematic review of defibrotide studies in the treatment of veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS). Bone Marrow Transplant. 2019;1. doi: 10.1038/s41409-019-0474-8.

  17. McKenna D, Sullivan M, Hill J, et al. Hospital readmission following transplantation: identifying risk factors and designing preventive measures. J Commun Supp Oncol. 2015;13(9):316–22. doi: 10.12788/jcso.0168.

  18. Seto A, Atsuta Y, Kawashima N, et al. Impact of hospital length of stay on the risk of readmission and overall survival after allogeneic stem cell transplantation. Int J Hematol. 2018;108(3):290–7. doi: 10.1007/s12185-018-2477-4.

  19. Spring L, Li S, Soiffer RJ, et al. Risk Factors for Readmission after Allogeneic Hematopoietic Stem Cell Transplantation and Impact on Overall Survival. Biol Blood Marrow Transplant. 2015;21:509–16. doi: 10.1016/j.bbmt.2014.11.682.

  20. Crombie J, Spring L, Li S, et al. Readmissions after Umbilical Cord Blood Transplantation and Impact on Overall Survival. Biol Blood Marrow Transplant. 2017;23(1):113–8. doi: 10.1016/J.BBMT.2016.10.012.

Severe Hypofunction of Allogeneic Hematopoietic Stem Cell Transplant in Patients with Oncohematological Diseases: Incidence, Risk Factors, and Outcomes

TA Rudakova, AD Kulagin, OU Klimova, IK Golubovskaya, EI Darskaya, TA Bykova, AG Smirnova, EV Morozova, SN Bondarenko, IS Moiseev, AV Beinarovich, DE Pevtsov, AL Alyanskii, EV Babenko, IM Barkhatov, BV Afanas’ev

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Tat’yana Aleksandrovna Rudakova, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; e-mail: t_a_rudakova@mail.ru

For citation: Rudakova TA, Kulagin AD, Klimova OU, et al. Severe Hypofunction of Allogeneic Hematopoietic Stem Cell Transplant in Patients with Oncohematological Diseases: Incidence, Risk Factors, and Outcomes. Clinical oncohematology. 2019;12(3):309–18 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-309-318


ABSTRACT

Aim. Based on strict criteria, to assess incidence, pretransplantation risk factors, and outcomes of severe hypofunction of graft, i.e. poor graft function (sPGF), following allogeneic hematopoietic stem cell transplantation (allo-HSCT) in adults.

Materials & Methods. The trial included 710 adult patients (median age was 31 years, range 18–70 years; 55 % male and 45 % female patients) with different hematological diseases and documented transplant engraftment after allo-HSCT from matched sibling (20 %), unrelated (67 %) and haploidentical (13 %) donors in the period from 2008 to 2016. Myeloablative and reduced-intensity conditioning regimens were administered in 30 % and 70 % of patients, respectively. The analysis was based on the following sPGF criteria: 2 or more lines of cytopenia (thrombocytes < 20 × 109/L, absolute neutrophil count < 0.5 × 109/L, and hemoglobin < 70 g/L at any time after documented engraftment), complete or stable mixed donor chimerism > 90 %, and absence of relapse signs, rejection, and severe acute graft-versus-host reaction. The following factors were analyzed: age, sex, diagnosis, presence/absence of remission in acute leukemias, ferritin level, type of donor, HLA-match, blood group and sex match, transplant origin, number of transplanted CD34+ cells, and conditioning regimen. Multivariate analysis included parameters of univariate analysis with < 0.05.

Results. After allo-HSCT sPGF was identified in 103 patients with 2-year cumulative incidence of 15 % (95% confidence interval [95% CI] 12–18 %). In most cases sPGF developed during the 1st year after allo-HSCT (median 50 days). Bi- and trilineage cytopenia was found in 59 % and 41 % of cases, respectively. In multivariate analysis sPGF risk was associated with myelodysplastic syndrome, myeloproliferative disorders (hazard ratio [HR] 3.403; 95% CI 1.972–5.606; < 0.0001), and haploidentical donors (HR 3.830; 95% CI 1.545–8.828; = 0.001). The absence of remission at the time of allo-HSCT in acute leukemias and blood group incompatibility were of borderline significance. In 50 % of cases sPGF determined poor outcome, including death from cytopenia-related complications, further relapses, and graft rejection. Prognosis of bilineage sPGF was slightly more favorable than that of trilineage sPGF.

Conclusion. The present large cohort trial yielded the incidence and analyzed the structure of sPGF in adult patients with oncohematological diseases. In addition, the key pretransplantation sPGF risk factors were identified. The results of the trial can serve to optimize the choice of therapy after allo-HSCT.

Keywords: allogeneic hematopoietic stem cell transplantation, poor graft function.

Received: March 6, 2018

Accepted: June 20, 2019

Read in PDF 


REFERENCES

  1. Sureda A, Bader P, Cesaro S, et al. Indications for allo- and auto-SCT for haematological diseases, solid tumours and immune disorders: current practice in Europe, 2015. Bone Marrow Transplant. 2015;50(8):1037–56. doi: 10.1038/bmt.2015.6.

  2. Афанасьев Б.В., Зубаровская Л.С., Семенова Е.В. и др. Опыт применения неродственной аллогенной трансплантации стволовых гемопоэтических клеток в клинике трансплантации костного мозга СПБГМУ им. акад. И.П. Павлова. Терапевтический архив. 2007;79(7):36–43.

    [Afanas’ev BV, Zubarovskaya LS, Semenova EV, et al. Experience of non-related allogeneic transplantation of stem hematopoietic cells in the Clinic of Bone Marrow Transplantation at I.P. Pavlov St. Petersburg Medical University. Terapevticheskii arkhiv. 2007;79(7):36–43. (In Russ)]

  3. Афанасьев Б.В., Зубаровская Л.С., Моисеев И.С. Аллогенная трансплантация гемопоэтических стволовых клеток у детей: настоящее, проблемы, перспективы. Российский журнал детской гематологии и онкологии. 2015;2(2):28–42. doi: 10.17650/2311-1267-2015-2-2-28-42.

    [Afanasiev BV, Zubarovskaya LS, Moiseev IS. Allogeneic hematopoietic stem cell transplantation in children: now, problems and prospects. Russian Journal of Children Hematology and Oncology. 2015;2(2):28–42. doi: 10.17650/2311-1267-2015-2-2-28-42. (In Russ)]

  4. Румянцев А.Г., Масчан А.А. Трансплантация гемопоэтических стволовых клеток у детей. М.: МИА, 2003. 912 с.

    [Rumyantsev AG, Maschan AA. Transplantatsiya gemopoeticheskikh stvolovykh kletok u detei. (Hematopoietic stem cell transplantation in children.) Moscow: MIA Publ.; 2003. 912 p. (In Russ)]

  5. Савченко В.Г., Любимова Л.С., Паровичникова Е.Н. и др. Трансплантация аллогенных и аутологичных гемопоэтических стволовых клеток при острых лейкозах (итоги 20-летнего опыта). Терапевтический архив. 2007;79(7):30–5.

    [Savchenko VG, Lyubimova LS, Parovichnikova EN, et al. Transplantation of allogeneic and autologous hematopoietic stem cells in acute leukemias (summary of 20-year experience). Terapevticheskii arkhiv. 2007;79(7):30–5. (In Russ)]

  6. Olsson R, Remberger M, Schaffer M, et al. Graft failure in the modern era of allogeneic hematopoietic SCT. Bone Marrow Transplant. 2013;48(4):537–43. doi: 10.1038/bmt.2012.239.

  7. Locatelli F, Lucarelli B, Merli P. Current and future approaches to treat graft failure after allogeneic hematopoietic stem cell transplantation. Expert Opin Pharmacother. 2014;15(1):23–36. doi: 10.1517/14656566.2014.852537.

  8. Kong Y, Chang Y-J, Wang Y-Z, et al. Association of an impaired bone marrow microenviroment with secondary poor graft function after allogenic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19(10):1465–73. doi: 10.1016/j.bbmt.2013.07.014.

  9. Stasia A, Ghiso A, Galaverna F, et al. CD34 selected cells for the treatment of poor graft function following allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2014;20(9):1440–3. doi: 10.1016/j.bbmt.2014.05.016.

  10. Лисуков И.А., Успенская О.С., Кулагин А.Д. и др. Использование ромиплостима в терапии тромбоцитопений после аллогенной трансплантации костного мозга. Онкогематология. 2012;7(1):29–34. doi: 17650/1818-8346-2012-7-1-29-34.

    [Lisukov IA, Uspenskaya OS, Kulagin AD, et al. Romiplostim in thrombocytopenia treatment after allogeneic bone marrow transplantation. Oncohematology. 2012;7(1):29–34. doi: 10.17650/1818-8346-2012-7-1-29-34. (In Russ)]

  11. Алянский А.Л., Макаренко О.А., Иванова Н.Е. и др. Развитие регистра неродственных доноров костного мозга в Российской Федерации: опыт НИИ детской онкологии, гематологии и трансплантологии им. Р.М. Горбачeвой. Российский журнал детской гематологии и онкологии. 2016;3(2):68–74. doi: 10.17650/2311-1267-2016-3-2-68-74.

    [Alyanskiy AL, Makarenko OA, Ivanova NE, et al. Development of donor bone marrow registry in Russian Federation: experience of Raisa Gorbacheva Memorial Research Institute of Children Oncology, Hematology and Transplantation. Russian Journal of Children Hematology and Oncology. 2016;3(2):68–74. doi: 10.17650/2311-1267-2016-3-2-68-74. (In Russ)]

  12. Serov YA, Barkhatov IM, Klimov AS, Berkos AS. Current methods and opportunities of next-generation sequencing (NGS) for HLA-typing. Cellular Therapy and Transplantation. 2016;5(4):63–70. doi: 10.18620/ctt-1866-8836-2016-5-4-63-70.

  13. Бархатов И.М., Шакирова А.И., Евдокимов А.В. и др. InDel-полиморфизм в количественной оценке посттрансплантационного химеризма. Ученые записки СПбГМУ им. акад. И.П. Павлова. 2016;23(4):40–5. doi: 10.24884/1607-4181-2016-23-4-40-45.

    [Barkhatov IM, Shakirova AI, Evdokimov AV, et al. InDel polymorphisms in quantitative posttransplant chimerism evaluation. The Scientific Notes of the I.P. Pavlov St. Petersburg State Medical University. 2016;23(4):40–5. doi: 10.24884/1607-4181-2016-23-4-40-45. (In Russ)]

  14. Valcarcel D, Sureda A. Graft Failure. In: E Carreras, C Dufour, M Mohty, N Kroger, eds. The EBMT Handbook. Hematopoietic Stem Cell Transplantation and Cellular Therapies. Springer, Cham; 2019. pp. 314. doi: 10.1007/978-3-030-02278-5.

  15. Davies SM, Weisdorf DJ, Haake RJ, et al. Second infusion of bone marrow for treatment of graft failure after allogeneic bone marrow transplantation. Bone Marrow Transplant. 1994;14:73–7.

  16. Dominietto A, Raiola AM, van Lint MT, et al. Factors influencing haematological recovery after allogeneic haemopoietic stem cell transplants: graft-versus-host disease, donor type, cytomegalovirus infections and cell dose. Br J Haematol. 2001;112(1):219–27. doi: 10.1046/j.1365-2141.2001.02468.x.

  17. Rondon G, Saliba RM, Khouri I, et al. Long Term Follow Up Of Patients Who Experienced Graft failure Post Allogeneic Progenitor Cell Transplantation. Results of a Single Institution Analysis. Biol Blood Marrow Transplant. 2008;14(8):859–66. doi: 10.1016/j.bbmt.2008.05.005.

  18. Tamari R, Ramnath Sh, Kuk D, et al. Poor graft function in recipients of T-cell depleted (TCD) allogeneic hematopoietic cell transplants (HSCT) is mostly related to viral infections and anti-viral therapy. Blood. 2012;120:3147.

  19. Xiao Y, Song J, Jiang Z, et al. Risk-Factor Analysis of Poor Graft Function after Allogeneic Hematopoietic Stem Cell Transplantation. Int J Med Sci. 2014;11(6):652–7. doi: 10.7150/ijms.6337.

  20. Askaa B, Fischer-Nielsen A, Vindelov L, et al. Treatment of poor graft function after allogeneic hematopoietic cell transplantation with a booster of CD34-selected cells infused without conditioning. Bone Marrow Transplant. 2014;49(5):720–1. doi: 10.1038/bmt.2014.5.

  21. Tang C, Chen F, Rong D, et al. Successful treatment of secondary poor graft function post allogeneic hematopoietic stem cell transplantation with eltrombopag. J Hematol Oncol. 2018;11(1):103. doi: 10.1186/s13045-018-0649-6.

  22. Rudakova TA, Eismont YuA, Moiseev IS, et al. Role of polyomavirus in emerging secondary hypofunction of marrow graft following allogeneic bone marrow transplantation in adults. Cellular Therapy and Transplantation. 2016;5(3):79–82. doi: 10.18620/ctt-1866-8836-2016-5-3-79-82.

  23. Alchalby H, Yunus D-R, Zabelina T, et al. Incidence and risk factors of poor graft function after allogeneic stem cell transplantation for myelofibrosis. Bone Marrow Transplant. 2016;51(9):1223–7. doi: 10.1038/bmt.2016.98.

  24. Klyuchnikov E, El-Cheikh J,Sputtek A, et al. CD34(+)-selected stem cell boost without further conditioning for poor graft function after allogeneic stem cell transplantation in patients with hematological malignancies. Biol Blood Marrow Transplant. 2014;20(3):382–6. doi: 10.1016/j.bbmt.2013.11.034.

  25. Kroger N, Holler E, Kobbe G, et al. Allogeneic stem cell transplantation after reduced-intensity conditioning in patients with myelofibrosis: a prospective, multicenter study of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Blood. 2009;114(26):5264–70. doi: 10.1182/blood-2009-07-234880.

  26. Bacigalupo A, Soraru M, Dominietto A, et al. Allogeneic hemopoietic SCT for patients with primary myelofibrosis: a predictive transplant score based on transfusion requirement, spleen size and donor type. Bone Marrow Transplant. 2010;45(3):458–63. doi: 10.1038/bmt.2009.188.

  27. Akpek G, Pasquini MC, Logan B, et al. Effects of spleen status on early outcomes after hematopoietic cell transplantation. Bone Marrow Transplant. 2013;48(6):825–31. doi: 10.1038/bmt.2012.249.

  28. Champlin RE, Horowitz MM, van Bekkum DW, et al. Graft failure following bone marrow transplantation for severe aplastic anemia: risk factors and treatment results. Blood. 1989;73:606–13.

  29. Shi M-M, Kong Y, Song Y, et al. Atorvastatin enhances endothelial cell function in posttransplant poor graft function. Blood. 2016;128(25):2988–99. doi: 10.1182/blood-2016-03-702803.

  30. Armand P, Kim HT, Cutler CS, et al. Prognostic impact of elevated pretransplantation serum ferritin in patients undergoing myeloablative stem-cell transplantation. Blood. 2007;109(10):4586–8. doi: 1182/blood-2006-10-054924.

  31. Shaheen M, Ivanova MO, Moiseev IS, et al. Impact of initial serum ferritin on early post-HSCT complications: a single-center study. Cellular Therapy and Transplantation. 2016;5(2):40–9. doi: 10.18620/1866-8836-2016-5-2-40-49.

  32. Chang Y-J, Zhao X-Y, Xu L-P, et al. Donor-specific anti-human leukocyte antigen antibodies were associated with primary graft failure after unmanipulated haploidentical blood and marrow transplantation: a prospective study with randomly assigned training and validation sets. J Hematol Oncol. 2015;8(1):84. doi: 10.1186/s13045-015-0182-9.

  33. Lee K-H, Lee J-H, Choi S-J et al. Failure of trilineage blood cell reconstitution after initial neutrophil engraftment in patients undergoing allogeneic hematopoietic cell transplantation – frequency and outcomes. Bone Marrow Transplant. 2004;33(7):729–34. doi: 10.1038/sj.bmt.1704428.

  34. Larocca A, Piaggio G, Podesta M, et al. Boost of CD34+-selected peripheral blood cells without further conditioning in patients with poor graft function following allogeneic stem cell transplantation. Haematologica. 2006;91:935–40.

Clinical Value of miR-3151 Overexpression in Synergistic Interaction with BAALC Host Gene in Patients with Acute Myeloid Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation

AI Shakirova, IM Barkhatov, AI Churkina, NN Mamaev, LS Zubarovskaya, BV Afanas’ev

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Alena Igorevna Shakirova, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)338-62-72; e-mail: alyona.i.shakirova@gmail.com

For citation: Shakirova AI, Barkhatov IM, Churkina AI, et al. Clinical Value of miR-3151 Overexpression in Synergistic Interaction with BAALC Host Gene in Patients with Acute Myeloid Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2019;12(3):303–8 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-303-308


ABSTRACT

Background. Among a multitude of molecular genetic changes underlying acute myeloid leukemia (AML) disordered epigenetic regulation is of special importance. It includes expression change in miR-3151 gene forming a part of BAALC gene on chromosome 8 in q22.3 locus. At present BAALC gene overexpression is observed in a half of AML patients. A considerable part of them shows a combination of it with an increased transcriptional activity of miR-3151 gene, which is associated with the poorest AML prognosis.

Aim. To assess the prognostic value of miR-3151 overexpression in synergistic interaction with BAALC host gene in AML patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Materials & Methods. The trial included bone marrow samples taken from 10 healthy SCT donors and 29 AML patients after receiving allo-HSCT. Relative miR-3151 expression level and relative BAALC copy number were measured by quantitative real-time polymerase chain reaction.

Results. The analysis yielded a poor correlation between miR-3151 expression level and blast cell count in bone marrow (r = 0.330; = 0.005) as well as between the expression levels of miR-3151 and BAALC (r = 0.273; = 0.020). In addition, a great prognostic value of miR-315 overexpression in post-transplantation period was confirmed (= 0.005). Patients with miR-315 and BAALC co-expression in post-transplantation period have also the poorest prognosis than the control group with regard to both disease-free survival and relapse risks within 2 years after allo-HSCT.

Conclusion. Monitoring expression level of miR-3151 and its host gene BAALC in AML patients after receiving allo-HSCT seems to be important not only in AML prognosis but also in therapy efficacy evaluation.

Keywords: acute myeloid leukemia, miR-3151, BAALC, prognosis, allogeneic hematopoietic stem cell transplantation.

Received: October 22, 2018

Accepted: June 7, 2019

Read in PDF 


REFERENCES

  1. Testa U, Pelosi E. MicroRNAs expressed in hematopoietic stem/progenitor cells are deregulated in acute myeloid leukemias. Leuk Lymphoma. 2015;56(5):1466–74. doi: 3109/10428194.2014.955019.

  2. Liao Q, Wang B, Li X, Jiang G. miRNAs in acute myeloid leukemia. Oncotarget. 2017;8(2):3666–82. doi: 10.18632/oncotarget.12343.

  3. Ambros V. MicroRNAs: tiny regulators with great potential. Cell. 2001;107(7):823–6. doi: 1016/S0092-8674(01)00616-X.

  4. Marcucci G, Haferlach T, Dohner H. Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. J Clin Oncol. 2011;29(5):475–86. doi: 10.1200/JCO.2010.30.2554.

  5. Ehtesham N, Sharifi M. From conventional therapy toward microRNA-based therapy in acute promyelocytic leukemia. Adv Biomed Res. 2016;5:187. doi: 10.4103/2277-9175.190996.

  6. Li Z, Lu J, Sun M, et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci. 2008;105:15535–40. doi: 10.1073/pnas.0808266105.

  7. Dixon-McIver A, East P, Mein CA, et al. Distinctive patterns of microRNA expression associated with karyotype in acute myeloid leukaemia. PLoS One. 2008;3(5):е2141. doi: 10.1371/journal.pone.0002141.

  8. Jongen-Lavrencic M, Sun SM, Dijkstra MK, et al. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood. 2008;111(10):5078–85. doi: 10.1182/blood-2008-01-133355.

  9. Stark M, Tyagi S, Nancarrow D, et al. Characterization of the Melanoma miRNAome by Deep Sequencing. PLoS One. 2010;5(3):e9685. doi: 10.1371/journal.pone.0009685.

  10. Eisfeld A-K, Schwind S, Patel R, et al. Intronic miR-3151 within BAALC drives leukemogenesis by deregulating the TP53 Pathway. Sci Signal. 2014;7(321):ra36. doi: 10.1126/scisignal.2004762.

  11. Eisfeld A-K, Marcucci G, Maharry K, et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood. 2012;120(2):249–58. doi: 10.1182/blood-2012-02-408492.

  12. Diaz-Beya M, Brunet S, Nomdedeu J, et al. The expression level of BAALC-associated microRNA miR-3151 is an independent prognostic factor in younger patients with cytogenetic intermediate-risk acute myeloid leukemia. Blood Cancer J. 2015;5(10):e352. doi: 10.1038/bcj.2015.76.

  13. Weber S, Haferlach T, Alpermann T, et al. Feasibility of BAALC gene expression for detection of minimal residual disease and risk stratification in normal karyotype acute myeloid leukaemia. Br J Haematol. 2016;175(5):904–16. doi: 10.1111/bjh.14343.

  14. Shakirova A, Barkhatov I, Churkina A, et al. Prognostic significance of BAALC overexpression in patients with AML during the posttransplant period. Cellular Therapy and Transplantation. 2018;7(2):54–63. doi: 10.18620/ctt-1866-8836-2018-7-2–54-63.

  15. Schnerch D, Yalcintepe J, Schmidts A, et al. Cell cycle control in acute myeloid leukemia. Am J Cancer Res. 2012;2(5):508–28.

  16. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol. 2009;27(31):5195–201. doi: 10.1200/JCO.2009.22.4865.

  17. Мамаев Н.Н., Горбунова А.В., Бархатов И.М. и др. Молекулярный мониторинг течения острых миелоидных лейкозов по уровню экспрессии гена WT1 после аллогенной трансплантации гемопоэтических стволовых клеток. Клиническая онкогематология. 2015;8(3):309–20. doi: 10.21320/2500-2139-2015-8-3-309-320.

    [Mamaev NN, Gorbunova AV, Barkhatov IM, et al. Molecular Monitoring of WT1 Gene Expression Level in Acute Myeloid Leukemias after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2015;8(3):309–20. doi: 21320/2500-2139-2015-8-3-309-320. (In Russ)]

  18. Hosen N, Sonoda Y, Oji Y, et al. Very low frequencies of human normal CD34+ haematopoietic progenitor cells express the Wilms’ tumour gene WT1 at levels similar to those in leukaemia cells. Br J Haematol. 2002;116(2):409–20. doi: 10.1046/j.1365-2141.2002.03261.x.

  19. Ellisen LW, Carlesso N, Cheng T, et al. The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells. EMBO J. 2001;20(8):1897–909. doi: 10.1093/emboj/20.8.1897.

  20. Panyajai P, Amnajphook N, Keawsangthongcharoen S, et al. Study of Leukemic Stem Cell Population (CD34+/CD38-) and WT1 Protein Expression in Human Leukemic Cell Lines. J Assoc Med Sci. 2018;51(1):38–44. doi: 10.14456/jams.2018.5.

  21. Baldus C, Tanner S, Kusewitt D, et al. BAALC, a novel marker of human hematopoietic progenitor cells. Exp Hematol. 2003;31(11):1051–6. doi: 10.1016/j.exphem.2003.08.004.

  22. Najima Y, Ohashi K, Kawamura M, et al. Molecular monitoring of BAALC expression in patients with CD34-positive acute leukemia. Int J Hematol. 2010;91(4):636–45. doi: 10.1007/s12185-010-0550-8.

  23. Xiao S, Shen JZ, Huang JL, et al. Prognostic significance of the BAALC gene expression in adult patients with acute myeloid leukemia: A meta-analysis. Mol Clin Oncol. 2015;3(4):880–8. doi: 10.3892/mco.2015.562.

  24. Lucena-Araujo A, Pereira-Martins D, Koury L, et al. Clinical impact of BAALC expression in high-risk acute promyelocytic leukemia. Blood Adv. 2017;1(21):1807–14. doi: 10.1182/bloodadvances.2017005926.

Comparative Study of Mycophenolate Mofetil and Methotrexate in Graft-Versus-Host Disease Prophylaxis in Adult Recipients of Related and Unrelated Allo-HSCT

IS Moiseev, YuA Tarakanova, AL Alyanskii, EV Babenko, MM Kanunnikov, VA Dubkova, EV Morozova, EI Darskaya, OA Slesarchuk, AD Kulagin, SN Bondarenko, BV Afanas’ev

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Ivan Sergeevich Moiseev, MD, PHD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)338-55-03; e-mail: moisiv@mail.ru

For citation: Moiseev IS, Tarakanova YuA, Alyanskii AL, et al. Comparative Study of Mycophenolate Mofetil and Methotrexate in Graft-Versus-Host Disease Prophylaxis in Adult Recipients of Related and Unrelated Allo-HSCT. Clinical oncohematology. 2019;12(1):43–50.

DOI: 10.21320/2500-2139-2019-12-1-43-50


ABSTRACT

Background. Although the use of methotrexate (MTX) and mycophenolate mofetil (MMF) for prophylaxis of graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT) was compared in a large number of studies, the published results are contradictory. This fact provides ground for the present retrospective single-center trial comparing these two approaches in GVHD prophylaxis.

Materials & Methods. The present study included 294 allo-HSC recipients with MTX prophylaxis and 172 allo-HSC recipients with MMF prophylaxis. 36 % of patients underwent matched related donor transplantation, and 64 % of patients received matched unrelated donor transplantation.

Results. Univariate and multivariate analyses showed that probability of acute grade 2–4 GVHD is 36 % vs. 39 % (hazard ratio [HR] 1.297; 95% confidence interval [95% CI] 0.931–1.795;= 0.122), grade 3–4 GVHD was 21 % vs. 25 % (HR 1.472; 95% CI 0.951–2.256;= 0.05), and probability of chronic GVHD was 52 % vs. 55 % (HR 0.978; 95% CI 0.951–1.406;= 0.91). In the MTX and MMF groups there were no significant differences in transplantation mortality (HR 1.173; 95% CI 0.797–1.708;= 0.43), relapse incidence (HR 1.034; 95% CI 0.743–1.428;= 0.84), overall survival (HR 1.087; 95% CI 0.825–1.433;= 0.55), event-free survival (HR 1.108; 95% CI 0.854–1.437;= 0.43), disease and GVHD free survival (HR 1.065; 95% CI 0.845–1.343;= 0.59). Engraftment occurred earlier when MMF was used (= 0.035). Administration of MMF instead of MTX was associated with lower probability of toxic grade 3–4 hepatitis (7 % vs. 31 %; p < 0.0001) and grade 3–4 mucositis (23 % vs. 45 %;= 0.0002).

Conclusion. The efficacy of GVHD prophylaxis using MMF is comparable with that of MTX, but MMF is associated with a better safety profile due to reduced incidence of severe liver toxicity and mucositis.

Keywords: allogeneic hematopoietic stem cell transplantation, graft-versus-host disease, prophylaxis, methotrexate, mycophenolate mofetil.

Received: May 23, 2018

Accepted: December 4, 2018

Read in PDF 


REFERENCES

  1. Савченко В.Г., Любимова Л.С., Паровичникова Е.Н. и др. Трансплантация аллогенных и аутологичных гемопоэтических стволовых клеток при острых лейкозах (итоги 20-летнего опыта). Терапевтический архив. 2007;79(7):30–5.

    [Savchenko VG, Lyubimova LS, Parovichnikova EN, et al. Transplantation of allogeneic and autologous hematopoietic stem cells in acute leukemias (summary of 20-year experience). Terapevticheskii arkhiv. 2007;79(7):30–5. (In Russ)]

  2. Афанасьев Б.В., Зубаровская Л.С., Цисская К.О. и др. Результаты трансплантации гемопоэтических предшественников у детей в России и Белоруссии по данным отчета рабочей группы по трансплантации у детей. Педиатрия. 1997;76(4):3.

    [Afanas’ev BV, Zubarovskaya LS, Tsisskaya KO, et al. Results of hematopoietic progenitor cell transplantation in children in Russia and Belorussia according to the report of the working group on transplantation in children. 1997;76(4):3. (In Russ)]

  3. Saliba RM, Couriel DR, Giralt S, et al. Prognostic value of response after upfront therapy for acute GVHD. Bone Marrow Transplant. 2012;47(1):125–31. doi: 10.1038/bmt.2011.41.

  4. Perez-Simon JA, Encinas C, Silva F, et al. Prognostic factors of chronic graft-versus-host disease following allogeneic peripheral blood stem cell transplantation: the National Institutes Health scale plus the type of onset can predict survival rates and the duration of immunosuppressive therapy. Biol Blood Marrow Transplant. 2008;14(10):1163–71. doi: 10.1016/j.bbmt.2008.07.015.

  5. Storb R, Deeg HJ, Pepe M, et al. Methotrexate and cyclosporine versus cyclosporine alone for prophylaxis of graft-versus-host disease in patients given HLA-identical marrow grafts for leukemia: long-term follow-up of a controlled trial. Blood. 1989;73(6):1729–34.

  6. Bacigalupo A, Lamparelli T, Bruzzi P, et al. Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO). Blood. 2001;98(10):2942–7. doi: 10.1182/blood.v98.10.2942.

  7. Ruutu T, van Biezen A, Hertenstein B, et al. Prophylaxis and treatment of GVHD after allogeneic haematopoietic SCT: a survey of centre strategies by the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2012;47(11):1459–64. doi: 10.1038/bmt.2012.45.

  8. Storb R, Leisenring W, Anasetti C, et al. Methotrexate and cyclosporine for graft-vs.-host disease prevention: what length of therapy with cyclosporine? Biol Blood Marrow Transplant. 1997;3(4):194–201.

  9. Niederwieser D, Maris M, Shizuru JA, et al. Low-dose total body irradiation (TBI) and fludarabine followed by hematopoietic cell transplantation (HCT) from HLA-matched or mismatched unrelated donors and postgrafting immunosuppression with cyclosporine and mycophenolate mofetil (MMF) can induce durable complete chimerism and sustained remissions in patients with hematological diseases. Blood. 2003;101(4):1620–9. doi: 10.1182/blood-2002-05-1340.

  10. Osunkwo I, Bessmertny O, Harrison L, et al. A pilot study of tacrolimus and mycophenolate mofetil graft-versus-host disease prophylaxis in childhood and adolescent allogeneic stem cell transplant recipients. Biol Blood Marrow Transplant. 2004;10(4):246–58. doi: 10.1016/j.bbmt.2003.11.005.

  11. Neumann F, Graef T, Tapprich C, et al. Cyclosporine A and mycophenolate mofetil vs cyclosporine A and methotrexate for graft-versus-host disease prophylaxis after stem cell transplantation from HLA-identical siblings. Bone Marrow Transplant. 2005;35(11):1089–93. doi: 10.1038/sj.bmt.1704956.

  12. Perkins J, Field T, Kim J, et al. A randomized phase II trial comparing tacrolimus and mycophenolate mofetil to tacrolimus and methotrexate for acute graft-versus-host disease prophylaxis. Biol Blood Marrow Transplant. 2010;16(7):937–47. doi: 10.1016/j.bbmt.2010.01.010.

  13. Yerushalmi R, Shem-Tov N, Danylesko I, et al. The combination of cyclosporine and mycophenolate mofetil is less effective than cyclosporine and methotrexate in the prevention of acute graft-versus host disease after stem-cell transplantation from unrelated donors. Am J Hematol. 2017;92(3):259–68. doi: 10.1002/ajh.24631.

  14. Terakura S, Wake A, Inamoto Y, et al. Exploratory research for optimal GvHD prophylaxis after single unit CBT in adults: short-term methotrexate reduced the incidence of severe GvHD more than mycophenolate mofetil. Bone Marrow Transplant. 2017;52(3):423–30. doi: 10.1038/bmt.2016.255.

  15. Przepiorka D, Weisdorf D, Martin P, et al. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995;15(6):825–8.

  16. Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol Blood Marrow Transplant. 2005;11(12):945–56. doi: 10.1016/j.bbmt.2005.09.004.

  17. Armand P, Kim HT, Logan BR, et al. Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation. Blood. 2014;123(23):3664–71. doi: 10.1182/blood-2014-01-552984.

  18. Morishima Y, Kawase T, Malkki M, et al. Significance of ethnicity in the risk of acute graft-versus-host disease and leukemia relapse after unrelated donor hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19(8):1197–203. doi: 10.1016/j.bbmt.2013.05.020.

  19. Kofler S, Deutsch MA, Bigdeli AK, et al. Proton pump inhibitor co-medication reduces mycophenolate acid drug exposure in heart transplant recipients. J Heart Lung Transplant. 2009;28(6):605–11. doi: 10.1016/j.healun.2009.03.006.

  20. van Gelder T, Klupp J, Barten MJ, et al. Comparison of the effects of tacrolimus and cyclosporine on the pharmacokinetics of mycophenolic acid. Ther Drug Monit. 2001;23(2):119–28. doi: 10.1097/00007691-200104000-00005.

  21. Maris MB, Sandmaier BM, Storer BE, et al. Unrelated donor granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cell transplantation after nonmyeloablative conditioning: the effect of postgrafting mycophenolate mofetil dosing. Biol Blood Marrow Transplant. 2006;12(4):454–65. doi: 10.1016/j.bbmt.2005.12.030.

  22. Hamad N, Shanavas M, Michelis FV, et al. Mycophenolate-based graft versus host disease prophylaxis is not inferior to methotrexate in myeloablative-related donor stem cell transplantation. Am J Hematol. 2015;90(5):392–9. doi: 10.1002/ajh.23955.

  23. Kiehl MG, Schafer-Eckart K, Kroger M, et al. Mycophenolate mofetil for the prophylaxis of acute graft-versus-host disease in stem cell transplant recipients. Transplant Proc. 2002;34(7):2922–4. doi: 10.1016/s0041-1345(02)03489-9.

  24. Bolwell B, Sobecks R, Pohlman B, et al. A prospective randomized trial comparing cyclosporine and short course with cyclosporine and mycophenolate mofetil for GVHD prophylaxis in myeloablative allogeneic bone marrow transplantation. Bone Marrow Transplant. 2004;34(7):621–5. doi: 10.1038/sj.bmt.1704647.

  25. Russell JA, Woodman RC, Poon MC. Addition of low-dose folinic acid to a methotrexate/cyclosporin A regimen for prevention of acute graft-versus-host disease. Bone Marrow Transplant. 1994;14(3):397–401.

  26. Моисеев И.С., Галанкин Т.Л., Доценко А.А. и др. Фармакоэкономика различных методов лечения стероид-рефрактерной реакции «трансплантат против хозяина»: анализ результатов лечения в одноцентровом исследовании. Ученые записки Санкт-Петербургского государственного медицинского университета имени академика И.П. Павлова. 2018;25(1):35–44. doi: 10.24884/1607-4181-2018-25-1-35-44.

    [Moiseev IS, Galankin TL, Dotsenko AA, et al. Pharmacoeconomic analysis of different methods for the treatment of steroid-refractory graft-versus-host disease: single-center study. The Scientific Notes of the I.P. Pavlov St. Petersburg State Medical University. 2018;25(1):35–44. doi: 10.24884/1607-4181-2018-25-1-35-44. (In Russ)]

  27. Моисеев И.С., Бурмина Е.А., Тараканова Ю.А. и др. Лечение хронической рефрактерной реакции «трансплантат против хозяина» после трансплантации гемопоэтических стволовых клеток с помощью низких доз интерлейкина-2. Ученые записки Санкт-Петербургского государственного медицинского университета имени академика И.П. Павлова. 2015;22(4):44–8. doi: 10.24884/1607-4181-2015-22-4-44-48.

    [Moiseev IS, Burmina EA, Tarakanova YuA, et al. Treatment of refractory chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation with low-dose interleukin-2. The Scientific Notes of the I.P. Pavlov St. Petersburg State Medical University. 2015;22(4):44–8. doi: 10.24884/1607-4181-2015-22-4-44-48. (In Russ)]

  28. Luznik L, O’Donnell PV, Symons HJ, et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2008;14(6):641–50. doi: 10.1016/j.bbmt.2008.03.005.

  29. Moiseev IS, Pirogova OV, Babenko EV, et al. Single-agent post-transplantation cyclophosphamide versus calcineurin-based graft-versus-host disease prophylaxis in matched related bone marrow transplantation. Cell Ther Transplant. 2017;6(4):52–9. doi: 10.18620/ctt-1866-8836-2017-6-4-52-59.

  30. Balashov D, Shcherbina A, Maschan M, et al. Single-Center Experience of Unrelated and Haploidentical Stem Cell Transplantation with TCRαβ and CD19 Depletion in Children with Primary Immunodeficiency Syndromes. Biol Blood Marrow Transplant. 2015;21(11):1955–62. doi: 10.1016/j.bbmt.2015.07.008.

Classification of Conditioning Regimens for Bone Marrow Transplantation: Historical Background and Current Perspectives

KN Melkova, GD Petrova, NV Gorbunova, TZ Chernyavskaya, OP Trofimova

NN Blokhin National Medical Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Kapitolina Nikolaevna Melkova, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: frolov63@bk.ru

For citation: Melkova KN, Petrova GD, Gorbunova NV, et al. Classification of Conditioning Regimens for Bone Marrow Transplantation: Historical Background and Current Perspectives. Clinical oncohematology. 2017;10(4):494–500 (In Russ).

DOI: 10.21320/2500-2139-2017-10-4-494-500


ABSTRACT

Hematopoietic stem cells transplantation is a current standard treatment for many oncohematological diseases. The milestone of any type of transplantation is the choice of conditioning regimen. This article presents the principles of classification of conditioning regimens in terms of myeloablativity and discusses the concepts of “autologous transplantation”, “high-dose chemotherapy supported by hematopoietic stem cells”, “allogeneic transplantation” and “immunotherapy”. Up-to-date uniform classification of conditioning regimens may serve an important prognostic component in assessing both the risks and efficacy of hematopoietic stem cells transplantation.

Keywords: conditioning regimens, allogeneic hematopoietic stem cell transplantation, autologous hematopoietic stem cell transplantation, total therapeutic exposure, acute leukemia, Hodgkin’s lymphoma, multiple myeloma.

Received: March 29, 2017

Accepted: July 8, 2017

Read in PDF


REFERENCES

  1. Passweg JR, Baldomero H, Bader P, et al. Hematopoietic SCT in Europe 2013: recent trends in the use of alternative donors showing more haploidentical donors but fewer cord blood transplants. Bone Marrow Transplant. 2015;50(4):476–82. doi: 10.1038/bmt.2014.312.
  2. Мелкова К.Н., Абдусаламов С.Н., Горбунова Н.В. и др. Трансплантация костного мозга при острых лейкозах. Клиническая онкогематология. 2010;3(4):395–9.[Melkova KN, Abdusalamov SN, Gorbunova NV, et al. Bone Marrow Transplantation in Case of Acute Leukemia. Klinicheskaya onkogematologiya. 2010;3(4):395–9. (In Russ)]
  3. Петрова Г.Д., Мелкова К.Н., Чернявская Т.З. и др. Петрова Г.Д., Мелкова К.Н., Чернявская Т.З. и др. Аутологичная трансплантация гемопоэтических стволовых клеток при первично-рефрактерном течении лимфомы Ходжкина: мнимый цугцванг или промежуточный ход? Клиническая онкогематология. 2015;8(3):321–30. doi: 10.21320/2500-2139-2015-8-3-321-330.[Petrova GD, Melkova KN, Chernyavskaya TZ, et al. Autologous Stem Cell Transplantation in Primary Refractory Hodgkin’s Lymphoma: Supposed Zugzwang or Zwischenzug? Clinical oncohematology. 2015;8(3):321–30. doi: 10.21320/2500-2139-2015-8-3-321-330. (In Russ)]
  4. Петрова Г.Д., Мелкова К.Н., Чернявская Т.З. и др. Первично-рефрактерное течение лимфомы Ходжкина и аутологичная трансплантация гемопоэтических стволовых клеток. Результаты одноцентрового проспективного исследования. Российский онкологический журнал. 2015;20(3):4–11.[Petrova GD, Melkova KN, Chernyavskaya TZ, et al. Primary-Refractory Course of Hodgkin’s Lymphoma and Autologous Hematopoietic Stem Cell Transplantation. Results of Single Center Prospective Study. Rossiiskii onkologicheskii zhurnal. 2015;20(3):4–11. (In Russ)]
  5. Петрова Г.Д., Мелкова К.Н., Горбунова Н.В. и др. Аутологичная трансплантация гемопоэтических стволовых клеток для консолидации ремиссии острого миелобластного лейкоза с факторами неблагоприятного прогноза в дебюте заболевания. Онкогематология. 2016;11(1):52–61. doi: 10.17650/1818-8346-2016-11-1-52-61.[Petrova GD, Melkova KN, Gorbunova NV, et al. Autologous Hematopoietic Stem Cell Transplantation for Remission Consolidation of Acute Myeloblastic Leukemia with Factors of Poor Prognosis in Disease Onset. Oncohematology. 2016;11(1):52–61. doi: 10.17650/1818-8346-2016-11-1-52-61. (In Russ)]
  6. Мелкова К.Н., Петрова Г.Д., Чернявская Т.З. Заготовка гемопоэтических стволовых клеток для проведения аутологичной трансплантации у пациентов с лимфомой Ходжкина неблагоприятного прогноза. Вестник ФГБНУ «РОНЦ им. Н.Н. Блохина». 2015;26(3):51–6.[Melkova KN, Petrova GD, Chernyavskaya TZ. Hematopoietic Stem Cells Preparation for Autologous Transplantation in Patients with Hodgkin’s Lymphoma of Poor Prognosis. Vestnik FGBNU «RONTs im. N.N. Blokhina». 2015;26(3):51–6. (In Russ)]
  7. Чернявская Т.З., Мелкова К.Н., Горбунова Н.В. и др. Использование стимулированного костного мозга для трансплантации в онкологии. Онкология. 2012;(2, приложение):28–33.[Chernyavskaya TZ, Melkova KN, Gorbunova NV, et al. Usage of Stimulated Bone Marrow for Oncology Transplantation. Onkologiya. 2012;(2 Suppl):28–33. (In Russ)]
  8. Melkova K, Chernyavskaya T, Abdusalamov S, et al. Application of autologous stimulated bone marrow as a source of hematopoietic material for transplantation. Cell Ther Transplant. 2011;3(12):33.
  9. Melkova K, Chernyavskaya T, Abdusalamov S, et al. Using stimulated bone marrow as a source of hematopoietic stem cells for allogenic transplantation. Cell Ther Transplant. 2011;3(12):34.
  10. Мелкова К.Н., Абдусаламов С.Н., Горбунова Н.В. и др. Интенсивная сопроводительная терапия в онкологии и гематологии. Вестник московского онкологического общества. 2011;2:3–4.[Melkova KN, Abdusalamov SN, Gorbunova NV, et al. Intensive Supportive Therapy in Oncology and Hematology. Vestnik moskovskogo onkologicheskogo obshchestva. 2011;2:3–4. (In Russ)]
  11. Мелкова К.Н., Абдусаламов С.Н., Горбунова Н.В. и др. Интенсивная сопроводительная терапия в гематологии. Клиническая онкогематология. 2011;4(1):70–4.[Melkova KN, Abdusalamov SN, Gorbunova NV, et al. Intensive Supportive Therapy in Hematology. Klinicheskaya onkogematologiya. 2011;4(1):70–4. (In Russ)]
  12. Мелкова К.Н. Аллогенная трансплантация костного мозга. Клиническая онкогематология. 2012;5(1):1–12.[Melkova KN. Allogeneic Bone Marrow Transplantation. Klinicheskaya onkogematologiya. 2012;5(1):1–12. (In Russ)]
  13. Vriesendorp HM. Aims of the conditioning. Exp Hematol. 2003;31(10):844–54. doi: 10.1016/s0301-472x(03)00229-7.
  14. Bacigalupo A, Sormani MP, Lamparelli T, et al. Reducing transplant-related mortality after allogeneic hematopoietic stem cell transplantation. Haematologica. 2004;89(10):1238–47.
  15. Мелкова К.Н., Горбунова Н.В., Чернявская Т.З. и др. Тотальное облучение организма человека при трансплантации костного мозга. Клиническая онкогематология. 2012;5(2):96–114.[Melkova KN, Gorbunova NV, Chernyavskaya TZ, et al. Total Human Body Irradiation at Bone Marrow Transplantation. Klinicheskaya onkogematologiya. 2012;5(2):96–114. (In Russ)]
  16.  Gratwohl A, Carreras E. Principles of conditioning. In: Apperley J, Carreras E, Gluckman E, Masszi T, eds. Hematopoietic Stem Cell Transplantation, 6th edition. Genoa: Forum service editore; 2012. pp. 122–37.
  17. Bensinger WI. High-dose preparatory regimens. In: Forman SJ, Negrin RS, Antin JH, eds. Thomas’ Hematopoietic Cell Transplantation, 5th edition. Willey Blackwell; 2016. Vol. 1. pp. 223–31.
  18. Лебеденко И.М., Ратнер Т.Г., Водяник В.В. и др. Проведение тотального облучения пациента перед трансплантацией костного мозга. Радиационная онкология и ядерная медицина. 2012;2:30–6.[Lebedenko IM, Ratner TG, Vodyanik VV, et al. Performance of Total Patient Irradiation before Bone Marrow Transplantation. Radiatsionnaya onkologiya i yadernaya meditsina. 2012;2:30–6. (In Russ)]
  19. Лебеденко И.М., Ратнер Т.Г., Водяник В.В. и др. Техническое и дозиметрическое обеспечение тотального облучения пациента перед трансплантацией костного мозга. Медицинская физика. 2012;3(55):11–9.[Lebedenko IM, Ratner TG, Vodyanik VV, et al. Technical and Radiation-Monitoring Supply of Total Patient Irradiation before Bone Marrow Transplantation. Meditsinskaya fizika. 2012;3(5):11–9. (In Russ)]
  20. Лебеденко И.М., Чернявская Т.З., Ставицкий Р.В. и др. Технический контроль состояния организма и его систем в процессе химио-лучевой терапии и трансплантации костного мозга при острых лейкозах. Медицинская техника. 2014;5:32–7.[Lebedenko IM, Chernyavskaya TZ, Stavitskii RV, et al. Technical Control of Patient’s Body State and Its Systems in Process of Chemoradiotherapy and Bone Marrow Transplantation. Meditsinskaya tekhnika. 2014;5:32–7. (In Russ)]
  21. Storb RF, Champlin R, Riddell SR, et al. Non-myeloablative transplants for malignant disease. Hematology Am Soc Hematol Educ Program. 2001;1:375–91. doi: 10.1182/asheducation-2001.1.375.
  22. Мелкова К.Н., Петрова Г.Д. Материалы 41-го конгресса Европейского общества по трансплантации костного мозга. Клиническая онкогематология. 2015;8(3):343–52.[Melkova KN, Petrova GD. Materials of 41st Congress of European Society of Bone Marrow Transplantation. Clinical oncohematology. 2015;8(3):343–52. (In Russ)]
  23. Sorror ML, Maris MB, Storer B, et al. Comparing morbidity and mortality of HLA-matched unrelated donor hematopoietic cell transplantation after nonmyeloablative and myeloablative conditioning: influence of pretransplantation comorbidities. Blood. 2004;104(4):961–8. doi: 10.1182/blood-2004-02-0545.
  24. Giralt S, Ballen K, Rizzo D, et al. Reduced-intensity conditioning regimen workshop: defining the dose spectrum. Report of a workshop convened by the center for international blood and marrow transplant research. Biol Blood Marrow Transplant. 2009;15(3):367. doi: 10.1016/j.bbmt.2008.12.497.
  25. Khouri I, Giralt S, Champlin R. Non-Myeloablative Allogeneic Hematopoietic Transplantation and Induction of Graft-Versus-Malignancy. In: Bashey A, Ball ED, eds. Cancer Treatment and Research. Boston: Springer; 2002. рр. 137–47. doi: 10.1007/978-1-4615-0919-6_7.

The Role of Hypomethylating Agents Prior to Allogeneic Hematopoietic Stem Cells Transplantation in Acute Myeloid Leukemia and Myelodysplastic Syndrome

VN Ovechkina1, SN Bondarenko1, EV Morozova1, IS Moiseev1, AA Osipova1, TL Gindina1, AI Shakirova1, TA Bykova1, AD Kulagin1, IA Samorodova2, EV Karyakina3, EA Ukrainchenko4, LS Zubarovskaya1, BV Afanas’ev1

1 RM Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

2 Municipal Clinical Hospital No. 31, 3 Dinamo pr-t, Saint Petersburg, Russian Federation, 197110

3 Municipal Hospital No. 15, 4 Avangardnaya str., Saint Petersburg, Russian Federation, 198205

4 Aleksandrov Hospital, 4 Solidarnosti pr-t, Saint Petersburg, Russian Federation, 193312

For correspondence: Varvara Nikolaevna Ovechkina, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)338-62-72; e-mail ovetchkina@gmail.com

For citation: Ovechkina VN, Bondarenko SN, Morozova EV, et al. The Role of Hypomethylating Agents Prior to Allogeneic Hematopoietic Stem Cells Transplantation in Acute Myeloid Leukemia and Myelodysplastic Syndrome. Clinical oncohematology. 2017;10(3):351–7 (In Russ).

DOI: 10.21320/2500-2139-2017-10-3-351-357


ABSTRACT

Background & Aims. The aim of the study was to evaluate the efficacy and safety of azacytidine and decitabine prior to allogeneic hematopoietic stem cell transplantation (allo-HSCT) in acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia.

Materials & Methods. The research included 62 patients who received hypomethylating agents (HMA) prior to allo-HSCT. The median age was 28 years (range from 1 to 68 years), the study population consisted of 27 (43.5 %) women and 35 (56.5 %) men.

Results. The overall response (complete + partial remission) was observed in 42 % (n = 26) of cases. At the time of allo-HSCT no disease progression was observed in 41 (66 %) patients. The multivariant analysis showed the overall survival (OS) statistically significantly increased with the graft retention (hazard ratio [HR] 0.002; 95% confidence interval [95% CI] 0.001–0.74; p = 0.03), and also with the administration of HMA after allo-HSCT (HR 0.24; 95% CI 0.08–0.67; p = 0.007). The response (stabilisation, partial or complete remission) due to HMA administration prior to allo-HSCT (HR 6.4; 95% CI 0.75–54.0; p = 0.08) was associated with improved OS. The event-free survival (EFS) was significantly higher with the response to azacytidine and decitabine at the time of allo-HSCT (HR 38.9; 95% CI 1.3–1198.0; p = 0.03) and with the graft retention (HR 0.02; 95% CI 0.005–0.1; p = 0.001). In patients with MDS compared with AML (HR 2.3; 95% CI 0.9–22.0; p = 0.08), there was a tendency to EFS improvement. Progression-free survival rates were higher in patients with a number of blast cells in the bone marrow less than 31 % at the time of diagnosis (HR 1.1; 95% CI 1.1–9.9; p = 0.01).

Conclusion. The use of azacytidine and decitabine prior to allo-HSCT allows to safely control the tumor mass in patients with MDS and to maintain the achieved remission with AML. In patients with a response to HMA, the best OS and EFS values are seen after allo-HSCT.

Keywords: acute myeloid leukemia, myelodysplastic syndrome, allogeneic hematopoietic stem cell transplantation, hypomethylating agents, azacitidine, decitabine.

Received: December 19, 2016

Accepted: March 9, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Ширин А.Д., Баранова О.Ю. Гипометилирующие препараты в онкогематологии. Клиническая онкогематология. 2016;9(4):369–82. doi: 10.21320/2500-2139-2016-9-4-369–382.
    [Shirin AD, Baranova OYu. Hypomethylating Agents in Oncohematology. Clinical oncohematology. 2016;9(4):369–82. doi: 10.21320/2500-2139-2016-9-4-369–382. (In Russ)]
  2. Бондаренко С.Н., Семенова Е.В., Афанасьев Б.В. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при остром миелобластном лейкозе в первой ремиссии. Терапевтический архив. 2013;84(7):18–25.
    [Bondarenko SN, Semenova EV, Afanas’ev BV, et al. Allogeneic hematopoietic stem cell transplantation for acute myeloblastic leukemia in first remission. Terapevticheskii arkhiv. 2013;84(7):18–25. (In Russ)]
  3. Паровичникова Е.Н., Троицкая В.В., Савченко В.Г. и др. Лечение больных острыми миелоидными лейкозами по протоколу российского многоцентрового рандомизированного исследования ОМЛ-01.10: результаты координационного центра. Терапевтический архив. 2014;86(7):14–23.
    [Parovichnikova EN, Troitskaya VV, Savchenko VG, et al. Treating patients with acute myeloid leukemias according to the protocol of the AML-01.10 Russian multicenter randomized trial: the Coordinating Center’s results. Terapevticheskii arkhiv. 2014;86(7):14–23. (In Russ)]
  4. de Witte T, Bowen D, Robin M, et al. Allogeneic hematopoietic stem cell transplantation for MDS and CMML: recommendations from an international expert panel. Blood. 2017;129(13):1753–62. doi: 10.1182/blood-2016-06-724500.
  5. Sohn SK, Moon JH. Survey of expert opinions and related recommendations regarding bridging therapy using hypomethylating agents followed by allogeneic transplantation for high-risk MDS. Crit Rev Oncol Hematol. 2015;95(2):243–50. doi: 10.1016/j.critrevonc.2015.03.004.
  6. Al-Ali HK, Jaekel N, Niederwieser D, et al. Azacitidine in patients with acute myeloid leukemia medically unfit for or resistant to chemotherapy: a multicenter phase I/II study. Leuk Lymphoma. 2012;53(1):110–7. doi: 10.3109/10428194.2011.606382.
  7. Cruijsen M, Lubbert M, Huls G, et al. Clinical Results of Hypomethylating Agents in AML Treatment. J Clin Med. 2014;4(1):1–17. doi: 10.3390/jcm4010001.
  8. Field T, Perkins J, Anasetti C, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  9. Al-Ali HK, Jaekel N, Niederwieser D. The role of hypomethylating agents in the treatment of elderly patients with AML. J Geriatr Oncol. 2014;5(1):89–105. doi: 10.1016/j.jgo.2013.08.004.
  10. Komrokji RS, DeZern AE, Sekeres MA, et al. Validation of International Working Group (IWG) Response Criteria in Higher-Risk Myelodysplastic Syndromes (MDS): A Report on Behalf of the MDS Clinical Research Consortium (MDS CRC). Blood. 2015;126:909.
  11. Seymour JF, Buckstein R, Santini V, et al. Efficacy and Safety of Azacitidine (AZA) Versus Conventional Care Regimens (CCR) in Patients Aged ≥ 75 Years with Acute Myeloid Leukemia (AML) in the Phase 3 AZA-AML-001 Study. Blood. 2016;128:2818.
  12. Garcia JS, Jain N, Godley LA. An update on the safety and efficacy of decitabine in the treatment of myelodysplastic syndromes. Onco Targets Ther. 2010;3:1–13. doi: 10.2147/ott.s7222.
  13. Кострома И.И., Грицаев С.В., Карягина Е.В. и др. Гематологическое улучшение — вариант благоприятного противоопухолевого ответа на лечение азацитидином при острых миелоидных лейкозах и миелодиспластических синдромах. Клиническая онкогематология. 2015;8(4):413–9. doi: 10.21320/2500-2139-2015-8-4-413-419.
    [Kostroma II, Gritsaev SV, Karyagina EV, et al. Hematological Improvement is a Favorable Response to Azacitidine in Patients with Acute Myeloid Leukemias and Myelodysplastic Syndromes. Clinical oncohematology. 2015;8(4):413–9. doi: 10.21320/2500-2139-2015-8-4-413-419. (In Russ)]
  14. Potter VT, Iacobelli S, Biezen A, et al. Comparison of Intensive Chemotherapy and Hypomethylating Agents before Allogeneic Stem Cell Transplantation for Advanced Myelodysplastic Syndromes: A Study of the Myelodysplastic Syndrome Subcommittee of the Chronic Malignancies Working Party of the European Society for Blood and Marrow Transplant Research. Biol Blood Marrow Transplant. 2016;22(9):1615–20. doi: 10.1016/j.bbmt.2016.05.026.
  15. Jabbour E, Mathisen MS, Garcia-Manero G, et al. Allogeneic hematopoietic stem cell transplantation versus hypomethylating agents in patients with myelodysplastic syndrome: A retrospective case-control study. Am J Hematol. 2013;88(3):198–200. doi: 10.1002/ajh.23371.
  16. Ahn JS, Kim YK, Min YH, et al. Azacitidine Pre-Treatment Followed by Reduced-Intensity Stem Cell Transplantation in Patients with Higher-Risk Myelodysplastic Syndrome. Acta Haematol. 2015;134(1):40–8. doi: 10.1159/000368711.
  17. Waespe N, Akker Van Den M, Klaassen RJ, et al. Response to treatment with azacitidine in children with advanced myelodysplastic syndrome prior to hematopoietic stem cell transplantation. Haematologica. 2016;101(12):1508–15. doi: 10.3324/haematol.2016.145821.
  18. Prebet Th, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. J Clin Oncol. 2011;29(24):3322–7. doi: 10.1200/jco.2011.35.8135.
  19. Bally C, Thepot S, Quesnel B, et al. Azacitidine in the treatment of therapy related myelodysplastic syndrome and acute myeloid leukemia (tMDS/AML): A report on 54 patients by the Groupe Francophone Des Myelodysplasies (GFM). Leuk Res. 2013;37(6):637–40. doi: 10.1016/j.leukres.2013.02.014.
  20. Fenaux P, Mufti GJ, Peterson BL, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomized, open-label, phase III study. Lancet Oncol. 2009;10(3):223–32. doi: 10.1016/s1470-2045(09)70003-8.
  21. Quintas-Cardama A, Ravandi F, Liu-Dumlao Th, et al. Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia. Blood. 2012;120(24):4840–5. doi: 10.1182/blood-2012-06-436055.
  22. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28(4):562–9. doi: 10.1200/jco.2009.23.8329.
  23. Pleyer L, Burgstaller B, Greil R, et al. Azacitidine front-line in 339 patients with myelodysplastic syndromes and acute myeloid leukaemia: comparison of French-American-British and World Health Organization classifications. J Hematol Oncol. 2016;9(1):39. doi: 10.1186/s13045-016-0263-4.
  24. Yahng SA, Yooh JH, Shin SH, et al. Response to pretransplant hypomethylating agents influences the outcome of allogeneic hematopoietic stem cell transplantation in adults with myelodysplastic syndromes. Eur J Haematol. 2013;90(2):111–20. doi: 10.1111/ejh.12038.
  25. Овечкина В.Н., Бондаренко С.Н., Морозова Е.В. и др. Острый миелобластный лейкоз и миелодиспластический синдром: применение азацитидина с профилактической и превентивной целью после аллогенной трансплантации гемопоэтических стволовых клеток. Клиническая онкогематология. 2017;10(1):45–51. doi: 10.21320/2500-2139-2017-10-1-45-51.
    [Ovechkina VN, Bondarenko SN, Morozova EV, et al. Acute Myeloblastic Leukemia and Myelodysplastic Syndrome: Azacitidine for Prophylactic and Preventive Purposes after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2017;10(1):45–51. doi: 10.21320/2500-2139-2017-10-1-45-51. (In Russ)]
  26. Craddock Ch, Jilani N, Siddique Sh, et al. Tolerability and Clinical Activity of Post-Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on the RICAZA Trial. Biol Blood Marrow Transplant. 2016;22(2):385–90. doi: 10.1016/j.bbmt.2015.09.004.

Acute Myeloblastic Leukemia and Myelodysplastic Syndrome: Azacitidine for Prophylactic and Preventive Purposes after Allogeneic Hematopoietic Stem Cell Transplantation

VN Ovechkina1, SN Bondarenko1, EV Morozova1, IS Moiseev1, OA Slesarchuk1, AG Smirnova1, OS Uspenskaya2, YaV Gudozhnikova1, AA Osipova1, VS Sergeev1, NN Mamaev1, LS Zubarovskaya1, BV Afanas’ev1

1 RM Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician IP Pavlov First St. Petersburg State Medical University, 12 Rentgena str., Saint Petersburg, Russian Federation, 197022

2 Leningrad District Clinical Hospital, 45–49 Lunacharskogo pr-t, Saint Petersburg, Russian Federation, 194291

For correspondence: Varvara Nikolaevna Ovechkina, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel. +7(812)338-62-72; e-mail: ovetchkina@gmail.com

For citation: Ovechkina VN, Bondarenko SN, Morozova EV, et al. Acute Myeloblastic Leukemia and Myelodysplastic Syndrome: Azacitidine for Prophylactic and Preventive Purposes after Allogeneic Hematopoietic Stem Cell Transplantation. Clinical oncohematology. 2017;10(1):45-51 (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-45-51


ABSTRACT

Aim. To evaluate the effectiveness of preventive and prophylactic post-transplantation therapy using azacitidine (5-AZA) in patients at high risk of post-transplantation relapse.

Methods. 136 patients were included in the study performed by the pairwise comparison: 68 of them received 5-AZA after allo-HSCT and 68 patients were included in the historical control group. 5-AZA was prescribed for prophylactic or preventive purposes. The results were assessed according to the OS, RR, EFS, DUM, and relapse-free and GVHR-free survival.

Results. 1-year OS was 76 % in the 5-AZA group (95% CI 60–84 %) and 44 % in the reference group (95% CI 33–55 %) (= 0.001); 2-year OS was 63 % (95% CI 39–67 %) and 37 % (95% CI 26–48 %) (= 0.007), respectively. The relapse rate (RR) in the 5-AZA group was 34 % (95% CI 22–46 %) during 1 year and 51 % (95% CI 38–64 %) in the reference group (= 0.02). 1- and 2-year disease unrelated mortality (DUM) was similar: 5 % in the 5-AZA group (95% CI 0.1–14.0 %) and 25 % (95% CI 13–37 %) in the reference group (= 0.005). 1-year EFS was 76 % in the 5-AZA group (95% CI 61–85 %) and 44 % in the reference group (95% CI 33–55 %) (= 0.001); 2-year EFS was 63 % (95% CI 39–67 %) and 37 % (95% CI 26–48 %) (= 0.01), respectively. 1-year relapse-free and GVHR-free survival was 55 % in the 5-AZA group (95% CI 41–69 %) and 28 % in the reference group (95% CI 17–39 %) (= 0.001); 2-year relapse-free and GVHR-free survival was 47 % (95% CI 32–62 %) and 27 % (95% CI 17–37 %) (= 0.002), respectively.

Conclusion. The use of 5-AZA for prophylactic and preventive purposes after allo-HSCT does not increase the risk of GVHR and DUM, does not suppress the GVL effect and can be used in combination with the donor lymphocyte infusion (DLI). The therapy with 5-AZA is safe during the early period after allo-HSCT. The drug does not suppress the GVL effect and can be used in high risk patients to prevent early post-transplantation relapse. The use of 5-AZA in combination with DLI does not increase the incidence of severe GVHR.

Keywords: acute myeloblastic leukemia, myelodysplastic syndrome, allogeneic hematopoietic stem cell transplantation, hypomethylating therapy, azacitidine.

Received: July 18, 2016

Accepted: December 17, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. O’Donnell MR, Tallman MS, Abboud CN, et al. Clinical Practice Guidelines in Oncology. Acute Myeloid Leukemia, Version 2.2013. J Natl Compr Canc Netw. 2013;11:1047–55.
  2. Cornelissen JJ, Gratwohl A, Schlenk RF, et al. The European LeukemiaNet AML Working Party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach. Nat Rev Clin Oncol. 2012;9(10):579–90. doi: 10.1038/nrclinonc.2012.150.
  3. Бондаренко С.Н., Семенова Е.В., Афанасьев Б.В. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при остром миелобластном лейкозе в первой ремиссии. Терапевтический архив. 2013;84(7):18–25.
    [Bondarenko SN, Semenova EV, Afanas’ev BV, et al. Allogeneic hematopoietic stem cell transplantation in acute myeloblastic leukemia at the first remission. Terapevticheskii arkhiv. 2013;84(7):18–25. (In Russ)]
  4. Паровичникова Е.Н., Троицкая В.В., Савченко В.Г. и др. Лечение больных острыми миелоидными лейкозами по протоколу российского многоцентрового рандомизированного исследования ОМЛ-01.10: результаты координационного центра. Терапевтический архив. 2014;86(7):14–23.
    [Parovichnikova EN, Troitskaya VV, Savchenko VG, et al. Treatment of patients with acute myeloid leukemias according to the protocol of the OML-01.10 multi-center randomized trial: coordination center results. Terapevticheskii arkhiv. 2014;86(7):14–23. (In Russ)]
  5. Greenberg PL, Stone RM, Al-Kali A, et al. Clinical Practice Guidelines in Oncology. Myelodysplastic Syndromes, Version 2.2017. J Natl Compr Canc Netw. 2017;15:60–87.
  6. Pavletic SZ, Kumar S, Mohty M, et al. NCA First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogenic Hematopoietic Stem Cell Transplantation: report from the Committee on the Epidemiology and Natural History of Relapse following Allogeneic Cell Transplantation. Biol Blood Marrow Transplant. 2010;16(7):871–90. doi: 10.1016/j.bbmt.2010.04.004.
  7. Mawad R, Lionberger JM, Pagel JM. Strategies to Reduce Relapse after Allogeneic Hematopoietic Cell Transplantation in Acute Myeloid Leukemia. Curr Hematol Malig Rep. 2013;8(2):132–40. doi: 10.1007/s11899-013-0153-6.
  8. de Lima M, Porter DL, Battiwalla M, et al. Proceedings from the National CANCER Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: part III, prevention and treatment of relapse after allogeneic transplantation. Biol Blood Marrow Transplant. 2014;20(1):4–13. doi: 10.1016/j.bbmt.2013.08.012.
  9. Porter DL, Aleya EP, Antin JH, et al. NCI First International Workshop NCA First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogenic Hematopoietic Stem Cell Transplantation: report from the Committee on Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation, Biol Blood Marrow Transplant. 2010;16(11):1467–503. doi: 10.1016/j.bbmt.2010.08.001.
  10. Слесарчук О.А., Бабенко Е.В., Афанасьев Б.В. и др. Эффективность инфузии донорских лимфоцитов у пациентов после различных видов аллогенных трансплантаций гемопоэтических стволовых клеток. Терапевтический архив. 2013;84(7):26–33.
    [Slesarchuk OA, Babenko EV, Afanas’ev BV, et al. Effectiveness of donor lymphocyte infusion of patients after different types of allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2013;84(7):26–33. (In Russ)]
  11. Schmid C, Labopin M, Nagler A, et al. Acute Leukaemia Working Party of the European Group for B. Marrow Transplantation. Treatment, risk factors, and outcome of adult with relapsed AML after reduced intensity conditioning for allogeneic stem cell transplantation. Blood. 2012;119(6):1599–606. doi: 10.1182/blood-2011-08-375840.
  12. Christopeit M, Kuss O, Finke J, et al. Second allograft for hematologic relapse of acute leukemia after first allogeneic stem-cell transplantation from related and unrelated donors: the role of donor change. J Clin Oncol. 2013;31(26):3259–71. doi: 10.1200/jco.2012.44.7961.
  13. Craddock C, Nagra S, Peniket A, et al. Factors predicting long-term survival after T-cell depleted reduced intensity allogeneic stem cell transplantation for acute myeloid leukemia. Haematologica. 2010;95(6):989–95. doi: 10.3324/haematol.2009.013920.
  14. Kroger N, Stubig T, Atanackovic D. Immune-Modulating Drugs and Hypomethylating Agents to Prevent or Treat Relapse after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2014;20(2):168–72. doi: 10.1016/j.bbmt.2013.09.009.
  15. Thomas X. DNA methyltransferase inhibitors in acute myeloid leukemia: discovery, design and first therapeutic experiences. Expert Opin Drug Discov. 2012;7(11):1039–51. doi: 10.1517/17460441.2012.722618.
  16. Choi J, Ritchey J, Prior LJ, et al. In vivo administration of hypomethylating agents mitigate graft-versus-host-disease without sacrificing graft-versus-leukemia. Blood. 2010;116(1):129–39. doi: 10.1182/blood-2009-12-257253.
  17. Goodyear CO, Dennis M, Jilani N, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia. Blood. 2012;119(14):3361–9. doi: 10.1182/blood-2011-09-377044.
  18. Silverman LR, Demakos EP, Peterson BL, et al. Randomized controlled trial of azacitidine in patients with myelodisplastic syndrome: a study of leukemia and cancer group B. J Clin Oncol. 2002;20(10):2429–40. doi: 10.1200/jco.2002.04.117.
  19. de Lima M, Giralt S, Thall PF, et al. Maintenance Therapy With Low-Dose Azacitidine After Allogeneic Hematopoietic Stem Cell Transplantation for Recurrent Acute Myelogenous Leukemia or Myelodysplastic Syndrome. Cancer. 2010;116(23):5420–31. doi: 10.1002/cncr.25500.
  20. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia. 2012;26(3):381–9. doi: 10.1038/leu.2011.234.
  21. Craddock Ch, Jilani N, Siddique Sh, et al. Tolerability and Clinical Activity of Post-Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on the RICAZA Trial. Biol Blood Marrow Transplant. 2016;22(2):385–90. doi: 10.1016/j.bbmt.2015.09.004.
  22. Antar A, Otrock ZK, Kharfan-Dabaja M, et al. Azacitidine in the treatment of extramedullary relapse of AML after allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2013;48(7):994–5. doi: 10.1038/bmt.2012.256.
  23. Schroeder T, Rachlis E, Bug G, et al. Treatment of Acute Myeloid Leukemia or Myelodysplastic Syndrome Relapse after Allogeneic Stem Cell Transplantation with Azacitidine and Donor Lymphocyte Infusions – A Retrospective Multicenter Analysis from the German Cooperative Transplant Study Group. Biol Blood Marrow Transplant. 2015;21(4):653–60. doi: 10.1016/j.bbmt.2014.12.016.
  24. Craddock Ch, Labopin M, Houhou M, et al. Activity and Tolerability of Azacitidine in Patients Who Relapse after Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia and Myelodysplasia: a Survey from the European Society for Blood and Marrow Transplantation. Blood. 2014;124: Poster 2506.
  25. Schroeder T, Czibere A, Platzbecker U, et al. Azacitidine and donor lymphocyte infusions ad first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013;27(6):1–7. doi: 10.1038/leu.2013.7.
  26. Steinmann J, Bertz H, Wasch R, et al. 5-Azacitidine and DLI can induce long-term remissions an AML patients relapsed after allograft. Bone Marrow Transplant. 2015;50(5):690–5. doi: 10.1038/bmt.2015.10.
  27. Schroeder T, Frobel J, Cadedduu R-P, et al. Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation. Leukemia. 2013;27(9):1910–3. doi: 10.1038/leu.2013.64.
  28. Czibere A, Bruns I, Kroger N, et al. 5-Azacytidine for the treatment of patients with acute myeloid leukemia or myelodysplastic syndrome who relapse after allo-SCT: a retrospective analysis. Bone Marrow Transplant. 2010;45(5):872–6. doi: 10.1038/bmt.2009.266.
  29. Tessoulin B, Delaunay J, Chevallier P, et al. Azacitidine salvage therapy for relapse of myeloid malignancies following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2014;49(4):567–71. doi: 10.1038/bmt.2013.233.

 

 

Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias with Hyperdiploid Karyotype

TL Gindina, NN Mamaev, ES Nikolaeva, SN Bondarenko, OA Slesarchuk, AS Borovkova, SV Razumova, OV Pirogova, AL Alyanskii, LS Zubarovskaya, BV Afanas’ev

RM Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician IP Pavlov First St. Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Tat’yana Leonidovna Gindina, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel: + 7(812)233-12-43; e-mail: cytogenetics.bmt.lab@gmail.com

For citation: Gindina TL, Mamaev NN, Nikolaeva ES, et al. Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias with Hyperdiploid Karyotype. Clinical oncohematology. 2016;9(4):383–90 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-383-390


ABSTRACT

Aim. To evaluate the prognostic impact of the different cytogenetic characteristics, including the modal number, the number of chromosomal aberrations in a complex karyotype, and adverse chromosomal abnormalities (ACA) (–7/7q–, –5/5q–, –17/17p–, t(6;9)(p22;q34)) on the results of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with hyperdiploid acute myeloid leukemia (H-AML).

Methods. Forty seven H-AML patients (21 women and 26 men, aged from 1 to 58 years, median — 23.9 years) were examined. The analysis of overall (OS) and event-free survival (EFS) predictors after allo-HSCT in patients with different clinical, transplant and cytogenetic characteristics was performed.

Results. The modal number of chromosomes (MN) of 47–48 was the most common one in the karyotype which was observed in 31 (66 %) patients. High hyperdiploidy with the modal number of 49–65 was identified in 13 (28 %) patients, near-triploid and near-tetraploid karyotypes were found in 3 (6 %) patients. Quantitative chromosomal abnormalities were nonrandom. Chromosome 8 (50 %), 21 (32 %), 13 (16 %) и 22 (16 %) trisomy was the most common one. Structural chromosomal abnormalities were detected in 22 (47 %) patients, at that, ACA were found in 7 (19 %) patients. In univariate analysis, the OS and EFS after allo-HSCT differed in patients with different clinical status (remission vs. active disease; = 0.003 and = 0.002, respectively), different chromosomal abnormalities in hyperdiploid karyotype (ACA– vs. ACA+; = 0.001 and = 0.03, respectively). An additional analysis of selected patients group with a structurally complex karyotype (n = 19) showed, that patients without ACA had a higher OS than patients with ACA (= 0.03). In multivariate analysis, the disease status (relapse) at allo-HSCT was an independent predictor of decreased OS and EFS (= 0.004 и = 0.006, respectively), as well as the presence of the ACA (= 0.002 only for OS).

Conclusion. ACA were high-risk factors in H-AML patients received allo-HSCT. Therefore, the patients with formal criteria of a complex karyotype should not be automatically included in the cytogenetic unfavorable risk group.


Keywords: hyperdiploid and complex karyotypes, acute myeloid leukemia, allogeneic hematopoietic stem cell transplantation, prognosis.

Received: April 17, 2016

Accepted: May 5, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Chilton L, Hills RK, Harrison CJ, et al. Hyperdiploidy with 49-65 chromosomes represents a heterogeneous cytogenetic subgroup of acute myeloid leukemia with differential outcome. Leukemia. 2013;28(2):321–8. doi: 1038/leu.2013.198.
  2. Sandahl JD, Kjeldsen E, Abrahamsson J, et al. Ploidy and clinical characteristics of childhood acute myeloid leukemia: a NOPHO-AML study. Genes Chromos Cancer. 2014;53(8):667–75. doi: 1002/gcc.22177.
  3. Stolzel F, Mohr B, Kramer M, et al. Karyotype complexity and prognosis in acute myeloid leukemia. Blood Cancer J. 2016;6:e386. doi: 101038/bcj.2015.114.
  4. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 1182/blood-2009-07-235358.
  5. Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116(3):354–65. doi: 1182/blood-2009-11-254441.
  6. Гиндина Т.Л., Мамаев Н.Н., Бархатов И.М. и др. Сложные повреждения хромосом у больных с рецидивами острых лейкозов после аллогенной трансплантации гемопоэтических стволовых клеток. Терапевтический архив. 2012;8:61–6.
    [Gindina TL, Mamaev NN, Barkhatov IM, et al. Complex chromosome damages in patients with recurrent acute leukemias after allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2012;8:61–6. (In Russ)]
  7. Schaffer L, McGovan-Jordan J, Schmid M. An International System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013. pp. 140. doi: 10.1002/ajmg.a.35995.
  8. Guo RJ, Atenafu EG, Craddock K, et al. Allogeneic hematopoietic cell transplantation may alleviate the negative prognostic impact of monosomal and complex karyotypes on patients with acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(5):690–5. doi: 1016/j.bbmt.2014.01.027.

Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias: Prognostic Significance of Complex Karyotype Including del(5q), –7, del(7q) Abnormalities

TL Gindina, NN Mamaev, SN Bondarenko, ES Nikolaeva, IA Petrova, OA Slesarchuk, AS Borovkova, SV Razumova, AL Alyanskii, LS Zubarovskaya, BV Afanas’ev

R.M. Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician I.P. Pavlov First St. Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Tat’yana Leonidovna Gindina, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)233-12-43; e-mail: cytogenetics.bmt.lab@gmail.com

For citation: Gindina TL, Mamaev NN, Bondarenko SN, et al. Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias: Prognostic Significance of Complex Karyotype Including del(5q), –7, del(7q) Abnormalities. Clinical oncohematology. 2016;9(3):271-78(In Russ).

DOI: 10.21320/2500-2139-2016-9-3-271-278


ABSTRACT

Aim. To evaluate the prognostic significance of the complex karyotype including del(5q), –7, del(7q) abnormalities in acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Materials & Methods. Forty-four AML patients with chromosome 5 and/or 7 abnormalities (22 women and 22 men, aged from 1.2 to 67 years, median 31.2 years) were examined. Analysis of overall (OS) and event-free survival (EFS) predictors after allo-HSCT in patients with different clinical, transplant and cytogenetic characteristics was performed.

Results. Prior to allo-HSCT, the complex karyotype (with three or more chromosomal abnormalities) was observed in 19 (43 %) patients, the monosomal karyotype was in 8 (18 %) patients. Univariate analysis demonstrated that OS and EFS differed in patients from different age groups (³ 18 vs. < 18 years; = 0.01 and = 0.05, respectively), with different disease status at transplantation (1 remission vs. other clinical status; = 0.1 and = 0.008, respectively), with and without complex karyotype (СK– vs. CK+; = 0.05 and = 0.002, respectively), with and without monosomal karyotype (МK– vs. MK+; = 0.009, only for EFS), and with different stem cells source (bone marrow vs. other source; = 0.03 only for OS). Multivariate analysis confirmed that age of 18 years and more (= 0.02 and = 0.01, respectively), active disease at allo-HSCT (= 0.04 and = 0.005, respectively), complex karyotype (= 0.04 и = 0.0008, respectively) and stem cell source other than bone marrow (= 0.02 only for OS) were independent predictors of OS and EFS deterioration.

Conclusion. The study demonstrates that chromosome 5 and/or 7 abnormalities as a part of the complex karyotype is high-risk factor in AML patients undergoing allo-HSCT (unlike the monosomal karyotype), that requires the special therapeutic approach.


Keywords: acute myeloid leukemias, complex karyotype, chromosome 5 and 7 abnormalities, allogeneic hematopoietic stem cell transplantation, prognosis.

Received: March 5, 2016

Accepted: April 5, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-
  2. Breems DA, Van Putten WL, De Greef GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol. 2008;26(29);4791–7. doi: 10.1200/jco.2008.16.0259.
  3. Medeiros BC, Othus M, Fang M, et al. Prognostic impact of monosomal karyotype in young adult and elderly acute myeloid leukemia: the Southwest Oncology Group (SWOG) experience. Blood. 2012;116(13):2224–8. doi: 10.1182/blood-2010-02-
  4. Fang M, Storer B, Estey E, et al. Outcome of patients with acute myeloid leukemia with monosomal karyotype who undergo hematopoietic cell transplantation. Blood. 2011;118(6):1490–4. doi: 10.1182/blood-2011-02-
  5. Lazarus HM, Litzow MR. AML cytogenetics: the complex just got simpler. Blood. 2012;120(12):2357–8. doi: 10.1182/blood-2012-08-
  6. Kayzer S, Zucknick M, Dohner K, et al. Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies. Blood. 2011;119(2):551–8. doi: 10.1182/blood-2011-07-
  7. Voutiadou G, Papaioannou G, Gaitatzi M, et al. Monosomal karyotype in acute myeloid leukemia defines a distinct subgroup within the adverse cytogenetic risk category. Cancer Genet. 2013;206(1–2):32–6. doi: 10.1016/j.cancergen.2012.10.003.
  8. Guo RJ, Atenafu EG, Craddock K, et al. Allogeneic hematopoietic cell transplantation may alleviate the negative prognostic impact of monosomal and complex karyotypes on patients with acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(5):690–5. doi: 10.1016/j.bbmt.2014.01.027.
  9. Cornelissen JJ, Breems D, Putten WLJ, et al. Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories. J Clin Oncol. 2012;30(17):2140–6. doi: 10.1200/jco.2011.39.6499.
  10. Hemmati P, Schuzle-Luckow A, Terwey T, et al. Cytogenetic risk grouping by the monosomal karyotype classification is superior in predicting the outcome of acute myeloid leukemia undergoing allogeneic stem cell transplantation in complete remission. Eur J Haematol. 2013;92(2):102–10. doi: 10.1111/ejh.12216.
  11. Гиндина Т.Л., Мамаев Н.Н., Бархатов И.М. и др. Сложные повреждения хромосом у больных с рецидивами острых лейкозов после аллогенной трансплантации гемопоэтических стволовых клеток. Терапевтический архив. 2012;84(8):61–6.
    [Gindina TL, Mamaev NN, Barhatov IM, et al. Complex chromosome damages in patients with recurrent acute leukemias after allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2012;84(8):61–6. (In Russ)]
  12. Schaffer L, McGovan-Jordan J, Schmid M. ISCN. An international System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013. pp. 140.
  13. Wawrzyniak E, Wierzbowska A, Kotkowska A, et al. Different prognosis of acute myeloid leukemia harboring monosomal karyotype with total or partial monosomies determined by FISH: Retrospective PALG study. Leuk Res. 2013;37(3):293–9. doi: 10.1016/j.leukres.2012.10.022.
  14. Yoon JH, Kim HJ, Shin SH, et al. Stratification of de novo adult acute myelogenous leukemia with adverse-risk karyotype: can we overcome the worse prognosis of adverse-risk group acute myelogenous leukemia with hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2014;20(1):80–8. doi: 10.1016/j.bbmt.2013.10.015.