Infectious Complications after Haploidentical Hematopoietic Stem Cells Transplantation in Patients with High-Risk Tumors of Hematopoietic and Lymphoid Tissues: A Single-Center Experience

YuS Osipov1, SS Bessmeltsev2, GN Salogub1, VV Ivanov1, ES Mikhailov1, NA Zhukova1, AV Chechetkin2

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

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

For correspondence: Yurii Sergeevich Osipov, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel.: +7(812)702-37-65; e-mail: osipov_yus@almazovcentre.ru

For citation: Osipov YuS, Bessmeltsev SS, Salogub GN, et al. Infectious Complications after Haploidentical Hematopoietic Stem Cells Transplantation in Patients with High-Risk Tumors of Hematopoietic and Lymphoid Tissues: A Single-Center Experience. Clinical oncohematology. 2019;12(4):406–15 (In Russ).

DOI: 10.21320/2500-2139-2019-12-4-406-415


ABSTRACT

Aim. To determine the incidence of viral, bacterial, and fungal infections in post-transplant period and to assess the prognostic value of infections and their influence on early and long-term results of haploidentical hematopoietic stem cells transplantation (haplo-HSCT).

Materials & Methods. Retrospective study included 61 patients older than 18 years with high-risk oncohematological diseases. In the period from 2015 to 2018 all patients received haplo-HSCT. Median follow-up after haplo-HSCT was 12.5 months (376 days, range 6–1202). Patients were divided into two groups. The first group (n = 26) received haplo-HSCT as salvage therapy. It included patients with refractory tumors without remission by the start of haplo-HSCT and patients with early relapses after HLA-matched related or unrelated allo-HSCT. The second group (n = 35) received haplo-HSCT on reaching the optimal pretransplant status (“non-salvage”).

Results. The incidence of cytomegalovirus (CMV) reactivation, invasive mycosis, and bacterial infections was 70.4 %, 11.5 %, and 75.4 %, respectively. CMV reactivation and invasive mycosis did not affect either the 35- or the 100-day overall survival (OS). For the first time bacterial infections were stratified based on severity according to Sepsis 3 consensus, which allowed to identify groups of patients with unfavorable prognosis. Severe bacterial infections (sepsis and septic shock) correlated with worse early and long-term results, especially in patients without remission by the start of haplo-HSCT, whereas febrile neutropenia/bloodstream infection did not affect OS. On the whole, mortality associated with bacterial infections was 26.2 %.

Conclusion. The main factor affecting early lethality after haplo-HSCT is a severe bacterial infection. The key risk factor is lack of remission by the start of haplo-HSCT. Sepsis 3 criteria can be applied in the period of postcytostatic cytopenia to identify the group of patients with most unfavorable prognosis (septic shock). The implementation of current infection control methods (genotyping of multiple drug resistant strains and timely determining the strategy of antimicrobial chemotherapy on the basis of the results obtained) into everyday clinical practice can improve the treatment outcomes in this category of patients.

Keywords: haploidentical hematopoietic stem cells transplantation, infectious complications, sepsis, septic shock, cytomegalovirus reactivation, invasive mycosis.

Received: April 11, 2019

Accepted: September 18, 2019

Read in PDF


REFERENCES

  1. Поп В.П., Рукавицын О.А. Аллогенная трансплантация гемопоэтических стволовых клеток: перспективы и альтернативы, собственный опыт. Российский журнал детской онкологии и гематологии. 2017;4(2):46–69. doi: 10.17650/2311-1267-2017-4-2-46-69.

    [Pop VР, Rukavitsyn OА. Allogeneic transplantation of hematopoietic stem cells: Perspectives and alternatives, own experience. Russian Journal of Children Hematology and Oncology. 2017;4(2):46–69. doi: 10.17650/2311-1267-2017-4-2-46-69. (In Russ)]

  2. Luznik L, O’Donnell PV, Ephraim JF. Post-transplantation cyclophosphamide for tolerance induction in HLA-haploidentical Bone Marrow Transplantation. Semin Oncol. 2012;39(6):683–93. doi: 10.1053/j.seminoncol.2012.09.005.

  3. Luznik L, Fuchs EJ. High-dose, post-transplantation cyclophosphamide to promote graft-host tolerance after allogeneic hematopoietic stem cell transplantation. Immunol Res. 2010;47(1–3):65–77. doi: 10.1007/s12026-009-8139-0.

  4. Burroughs LМ, O’Donnell PV, Sandmaier BM, et al. Comparison of outcomes of HLA-matched related, unrelated, or HLA-haploidentical related hematopoietic cell transplantation following non-myeloablative conditioning for relapsed or refractory Hodgkin lymphoma. Biol Blood Marrow Transplant. 2008;14(11):1279–87. doi: 10.1016/j.bbmt.2008.08.014.

  5. Pagliardini T, Harbi S, Furst S, et al. Post-transplantation cyclophosphamide-based haploidentical versus Atg-based unrelated donor allogeneic stem cell transplantation for patients younger than 60 years with hematological malignancies: a single-center experience of 209 patients. Bone Marrow Transplant. 2018;54(7):1067–76. doi: 10.1038/s41409-018-0387-y.

  6. Kasamon Y, Luznik L, Leffell M, et al. Nonmyeloablative HLA-haploidentical Bone Marrow Transplantation with high-dose post-transplantation cyclophosphamide: effect of HLA disparity on outcome. Biol Blood Marrow Transplant. 2010;16(4):482–9. doi: 10.1016/j.bbmt.2009.11.01

  7. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома: руководство для врачей. М.: СИМК, 2016. 512 с.

    [Bessmeltsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma: rukovodstvo dlya vrachei. (Multiple myeloma: manual for physicians.) Moscow: SIMK Publ.; 2016. 512 p. (In Russ)]

  8. Галстян Г.М., Макарова П.М., Кузьмина Л.А. и др. Успешная трансплантация аллогенного костного мозга у больных с тяжелым грамотрицательным сепсисом и септическим шоком. Клиническая онкогематология. 2014;7(2):122–30.

    [Galstyan GM, Makarova PM, Kuzmina LA, et al. Successful allogeneic bone marrow transplantation in patients with severe gram-negative sepsis and septic shock. Klinicheskaya onkogematologiya. 2014;7(2):122–30. (In Russ)]

  9. Fayard A, Daguenet E, Blaise D, et al. Evaluation of infectious complications after haploidentical hematopoietic stem cell transplantation with post-transplant cyclophosphamide following reduced-intensity and myeloablative conditioning: a study on behalf of the Francophone Society of Stem Cell Transplantation and Cellular Therapy (SFGM-TC). Bone Marrow Transplant. 2019. [ahead of print] doi: 10.1038/s41409-019-0475-7.

  10. Kumar G, Ahmad S, Taneja A, et al. Severe sepsis in hematopoietic stem cell transplant recipients. Crit Care Med. 2015;43(2):411–21. doi: 10.1097/ccm.0000000000000714.

  11. Omrani AS, Almaghrabi RS. Complications of hematopoietic stem cell transplantation: Bacterial infections. Hematol Oncol Stem Cell Ther. 2017;10(4):228–32. doi: 10.1016/j.hemonc.2017.05.018.

  12. Alhemmari SH, Refaat SM, Abdullah AA, Abul M. Infectious complications after allogeneic bone marrow transplantation: Sheikha Badryia Center, Kuwait. Gulf J Oncol. 2015;1(18):79–86.

  13. Freifeld AG, Bow EJ, Sepkowitz KA, et al. Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the infectious diseases society of America. Clin Infect Dis. 2011;52(4):е56–е93. doi: 10.1093/cid/cir073.

  14. EORTC International Antimicrobial Therapy Cooperative Group. Gram-positive bacteraemia in granulocytopenic cancer patients. Eur J Cancer Clin Oncol. 1990;26(5):569–74. doi: 10.1016/0277-5379(90)90079-9.

  15. Klastersky J. Science and pragmatism in the treatment and prevention of neutropenic infection. J Antimicrob Chemother. 1998;41(Suppl 4):13–24. doi: 10.1093/jac/41.suppl_4.13.

  16. Mikulska M, Viscoli C, Orasch C, et al. Aetiology and resistance in bacteraemias among adult and paediatric haematology and cancer patients. J Infect. 2014;68(4):321–31. doi: 10.1016/j.jinf.2013.12.006.

  17. Tomblyn M, Chiller T, Einsele H, et al. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant. 2009;15(10):1143–238. doi: 10.1016/j.bbmt.2009.06.019.

  18. De Pauw B, Walsh TJ, Donnelly JP, et al. Revised Definitions of Invasive Fungal Disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin Infect Dis. 2008;46(12):1813–21. doi: 10.1086/588660.

  19. Abraham E. New Definitions for Sepsis and Septic Shock: Continuing Evolution but With Much Still to Be Done. JAMA. 2016;315(8):757–9. doi: 10.1001/jama.2016.0290.

  20. Gustinetti G, Mikulska M. Bloodstream infections in neutropenic cancer patients: a practical update. Virulence. 2016;7(3):280–97. doi: 10.1080/21505594.2016.1156821.

  21. Wisplinghoff H, Seifert H, Wenzel RP, Edmond MB. Current trends in the epidemiology of nosocomial bloodstream infections in patients with hematological malignancies and solid neoplasms in hospitals in the United States. Clin Infect Dis. 2003;36(9):1103–10. doi: 10.1086/374339.

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 Efficacy Analysis of Mobilization and Collection of Autologous Hematopoietic Stem Cells in Patients with Lymphoproliferative Disorders and Multiple Sclerosis

OV Fedyk, VO Sarzhevskii, DA Fedorenko, VYa Mel’nichenko, YuN Dubinina, NE Mochkin, EG Smirnova, DS Kolesnikova, AE Bannikova

NI Pirogov Russian National Medical Center of Surgery, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203

For correspondence: Oksana Vladimirovna Fedyk, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203; Tel.: +7(968)748-93-42; e-mail: ksen1005@inbox.ru.

For citation: Fedyk OV, Sarzhevskii VO, Fedorenko DA, et al. Comparative Efficacy Analysis of Mobilization and Collection of Autologous Hematopoietic Stem Cells in Patients with Lymphoproliferative Disorders and Multiple Sclerosis. Clinical oncohematology. 2019;12(1):51–8.

DOI: 10.21320/2500-2139-2019-12-1-51-58


ABSTRACT

Aim. Comparative efficacy analysis of autologous hematopoietic stem cells (HSC) prior to auto-HSCT in patients with lymphoproliferative disorders (LPDs) and multiple sclerosis (MS).

Materials & Methods. The trial included 237 patients: 103 LPD and 134 MS patients. In 225 patients HSC mobilization involved only colony-stimulating factors (CSFs), in 12 patients chemotherapy (cyclophosphamide, etoposide) was combined with CSFs. On the intended date of cytapheresis all the patients were tested for CD34+ marker expression. Сytapheresis followed in the patients with CD34+ count more than 0.01 × 106/mL.

Results. In 23 (22 %) LPD patients CD34+ count was too low for auto-HSCT (‘collection failure group’). Within this group 19 patients received CSF mobilization, and 4 patients received chemotherapy + CSF. Plerixafor was administered in 5 patients, in 4 of them a repeated mobilization also failed to collect enough cells. In 80 LPD patients the number of mobilized and collected CD34+ cells was sufficient for auto-HSCT (‘collection success group’). Within this group 77 patients received auto-HSCT, 74 patients were treated with CSF mobilization, 6 patients received chemotherapy + CSF, and in 11 patients plerixafor was administered. Median total number of CD34+ cells in the ‘collection success group’ was 2.7 × 106/kg. All 134 MS patients had enough CD34+ cells for auto-HSCT. All of them received CSF mobilization. Median total number of CD34+ cells in the MS group was 2.34 × 106/kg. Potential risk factors for HSC mobilization failure in LPDs were evaluated. They included age, gender, prior radiotherapy, number of antitumor treatment lines prior to auto-HSCT, clinical response prior to auto-HSCT (complete/partial remission or stabilization), and HSC mobilization regimen. These factors with the exception of gender were not associated with mobilization failure parameters. The worst mobilization outcomes were reported in male patients.

Conclusion. In 22 % of LPD patients the planned high-dose chemotherapy and auto-HSCT failed due to insufficient counts of autologous CD34+ cells in apheresis product. Male gender can be considered to be a prognostic factor of mobilization failure in LPDs.

Keywords: lymphoproliferative disorders, autologous transplantation, mobilization of peripheral blood stem cells, autoimmune diseases, multiple sclerosis.

Received: June 25, 2018

Accepted: December 8, 2018

Read in PDF 


REFERENCES

  1. Giralt S, Costa L, Schriber J, et al. Optimizing Autologous Stem Cell Mobilization Strategies to Improve Patient Outcomes: Consensus Guidelines and Recommendations. Biol Blood Marrow Transplant. 2014;20(3):295–308. doi: 10.1016/j.bbmt.2013.10.013.

  2. Oliansky DM, Czuczman M, Fisher RI, et al. The role of cytotoxic therapy with hematopoietic stem cell transplantation in the treatment of diffuse large B cell lymphoma: update of the 2001 evidence-based review. Biol Blood Marrow Transplant. 2011;17(1):20–47. doi: 10.1016/j.bbmt.2010.07.008.

  3. Dreyling M, Lenz G, Hoster E, et al. Early consolidation by myeloablative radiochemotherapy followed by autologous stem cell transplantation in first remission significantly prolongs progression-free survival in mantle-cell lymphoma: results of a prospective randomized trial of the European MCL Network. Blood. 2005;105(7):2677–84. doi: 10.1182/blood-2004-10-3883.

  4. Linch DC, Winfield D, Goldstone AH, et al. Dose intensification with autologous bone-marrow transplantation in relapsed and resistant Hodgkin’s disease: results of a BNLI randomised trial. Lancet. 1993;341(8852):1051–4. doi: 10.1016/0140-6736(93)92411-l.

  5. Mounier N, Canals C, Gisselbrecht C, et al. High-dose therapy and autologous stem cell transplantation in first relapse for diffuse large B cell lymphoma in the rituximab era: an analysis based on data from the European Blood and Marrow Transplantation Registry. Biol Blood Marrow Transplant. 2012;18(5):788–93. doi: 10.1016/j.bbmt.2011.10.010.

  6. Hermine О, Hoster E, Walewski J, et al. Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): a randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. Lancet. 2016;388(10044):565–75. doi: 10.1016/S0140-6736(16)00739-X.

  7. Philip T, Guglielmi C, Hagenbeek A, et al. Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non-Hodgkin’s lymphoma. N Engl J Med. 1995;333(23):1540–5. doi: 10.1056/NEJM199512073332305.

  8. Schmitz N, Pfistner B, Sextro M, et al. Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin’s disease: a randomised trial. Lancet. 2002;359(9323):2065–71. doi: 10.1016/S0140-6736(02)08938-9.

  9. Atkins H. Hematopoietic SCT for the treatment of multiple sclerosis. Bone Marrow Transplant. 2010;45(12):1671–81. doi: 10.1038/bmt.2010.168.

  10. Nash RA, Hutton GJ, Racke MK, et al. High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for relapsing-remitting multiple sclerosis (HALT-MS): a 3-year interim report. JAMA Neurol. 2015;72(2):159–69. doi: 10.1001/jamaneurol.2014.3780.

  11. Shevchenko J, Kuznetcov A, Ionova T, et al. Long-term outcomes of autologous hematopoietic stem cell transplantation with reduced-intensity conditioning in multiple sclerosis: physician’s and patient’s perspectives. Ann Hematol. 2015;94(7):1149–57. doi: 10.1007/s00277-015-2337-8.

  12. Федоренко Д.А. Принципы оценки эффективности аутологичной трансплантации гемопоэтических стволовых клеток у больных лимфомами и рассеянным склерозом: Дис. … д-ра мед. наук. М., 2015. 188 с.

    [Fedorenko DA. Printsipy otsenki effektivnosti autologichnoi transplantatsii gemopoeticheskikh stvolovykh kletok u bol’nykh limfomami i rasseyannym sklerozom. (Principles of efficacy estimation of autologous hematopoietic stem cell transplantation in lymphoma and multiple sclerosis patients.) [dissertation] Moscow; 2015. (In Russ)]

  13. Покровская О.С. Механизм действия и клиническая эффективность антагониста хемокинового рецептора CXCR4 плериксафора при мобилизации гемопоэтических стволовых клеток. Клиническая онкогематология. 2012;5(4):371–9.

    [Pokrovskaya OS. Mechanism of action and clinical activity of CXCR4 antagonist Plerixafor in stem cell mobilization. Klinicheskaya onkogematologiya. 2012;5(4):371–9. (In Russ)]

  14. Покровская О.С. Системные эффекты Г-КСФ при мобилизации стволовых клеток крови (СКК) у больных множественной миеломой (ММ). Клиническая онкогематология. 2009;2(1):67–8.

    [Pokrovskaya OS. Systemic effects of G-CSF in peripheral stem cells (PSC) mobilization in multiple myeloma (MM) patients. Klinicheskaya onkogematologiya. 2009;2(1):67–8. (In Russ)]

  15. Lemoli RM. New Strategies for Stem Cell Mobilization. Mediterr J Hematol Infect Dis. 2012;4(1):e2012066. doi: 10.4084/MJHID.2012.066.

  16. Pavone V, Gaudio F, Console G, et al. Poor mobilization is an independent prognostic factor in patients with malignant lymphomas treated by peripheral blood stem cell transplantation. Bone Marrow Transplant. 2006;37(8):719–24. doi: 10.1038/sj.bmt.1705298.

  17. Kim JG, Sohn SK, Chae YS, et al. Multicenter study of intravenous busulfan, cyclophosphamide, and etoposide (i.v. Bu/Cy/E) as conditioning regimen for autologous stem cell transplantation in patients with non-Hodgkin’s lymphoma. Bone Marrow Transplant. 2007;40(10):919–24. doi: 10.1038/sj.bmt.1705841.

  18. Blank N, Lisenko K, Pavel P, et al. Low-dose cyclophosphamide effectively mobilizes peripheral blood stem cells in patients with autoimmune disease. Eur J Haematol. 2016;97(1):78–82. doi: 10.1111/ejh.12686.

  19. Kyrcz-Krzemien S, Helbig G, Torba K, et al. Safety and efficacy of hematopoietic stem cells mobilization in patients with multiple sclerosis. Hematology. 2016;21(1):42–5. doi: 10.1080/10245332.2015.1101973.

  20. Моталкина М.С., Кулева С.А., Алексеев С.М. и др. Пример успешной мобилизации стволовых кроветворных клеток периферической крови с помощью плериксафора и пэгфилграстима у пациентки с неходжкинской лимфомой. Современная онкология. 2015;17(2):54–6.

    [Motalkina MS, Kuleva SA, Alekseev SM, et al. An example of successful mobilization of peripheral blood stem cells from with plerixafor and pegfilgrastim administration in a non-Hodgkin’s lymphoma patient. Sovremennaya onkologiya. 2015;17(2):54–6. (In Russ)]

  21. Wuchter P, Ran D, Bruckner T, et al. Poor Mobilization of Hematopoietic Stem Cells-Definitions, Incidence, Risk Factors, and Impact on Outcome of Autologous Transplantation. Biol Blood Marrow Transplant. 2010;16(4):490–9. doi: 10.1016/j.bbmt.2009.11.012.

  22. Mendrone AJr, Arrais CA, Saboya R, et al. Factors affecting hematopoietic progenitor cell mobilization: an analysis of 307 patients. Transfus Apher Sci. 2008;39(3):187–92. doi: 10.1016/j.transci.2008.09.012.

  23. To LB, Levesque J-P, Herbert KE. How I treat patients who mobilize hematopoietic stem cells poorly. Blood. 2011;118(17):4530–40. doi: 10.1182/blood-2011-06-318220.

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.

Preventive Use of Azacitidine in Patients with Acute Myeloid Leukemia after Haploidentical Allo-BMT

RSh Badaev, DB Zammoeva, LL Girshova, DV Babenetskaya, NA Il’ina, YuA Alekseeva, AYu Zaritskey, DV Motorin

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

For correspondence: Dmitrii Vasil’evich Motorin, MD, PhD, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; e-mail: almazov-bmt@mail.ru

For citation: Badaev RSh, Zammoeva DB, Girshova LL, et al. Preventive Use of Azacitidine in Patients with Acute Myeloid Leukemia after Haploidentical Allo-BMT. Clinical oncohematology. 2019;12(1):37–42.

DOI: 10.21320/2500-2139-2019-12-1-37-42


ABSTRACT

Background. Haploidentical bone marrow transplantation (BMT) can be a reliable alternative if a fully matched donor is not available. The main challenges after BMT are a relapse of major disease, graft-versus-host disease (GVHD), and infections. Azacitidine possesses antileukemic effect together with immunomodulating properties and being administered soon after BMT can significantly improve the outcome.

Aim. To study azacitidine effect on the outcome of haploidentical BMT in patients with acute myeloid leukemia (AML) in the early post-transplantation period.

Materials & Methods. The trial included 18 AML patients who received haploidentical BMT at VA Almazov National Medical Research Center. In all patients MRD-negative remission was achieved on the 30th day after BMT. Azacitidine therapy was initiated not earlier than 2 months after BMT with a complete engraftment of transplant and no GVHD. Azacitidine 100 mg/day was administered on D1–D5 every 28 days within a year after BMT. When a molecular relapse was detected, donor lymphocytes were additionally infused during every other cycle of therapy.

Results. Eleven patients received preventive azacitidine treatment, 7 patients were included in control group. Median onset of azacitidine treatment after haploidentical BMT was 4 months (range 2–10 months), median number of azacitidine courses was 3.5 (range 1–9). During azacitidine treatment acute GVHD was identified in 5 (45.4 %) patients. In 4 of them an exacerbation of earlier GVHD was detected (3 with cutaneous form and 1 with intestinal form), and only in 1 patient de novo acute intestinal GVHD was discovered.

Conclusion. Azacitidine treatment of AML patients after haploidentical allo-BMT is safe and well tolerated. Preventive azacitidine treatment after haploidentical BMT improves overall survival of AML patients.

Keywords: haploidentical allogeneic bone marrow transplantation, azacitidine, acute myeloid leukemia.

Received: June 22, 2018

Accepted: December 11, 2018

Read in PDF 


REFERENCES

  1. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2016;129(4):424–47. doi: 10.1182/blood-2016-08-733196.

  2. McCurdy SR, Kanakry JA, Showel MM, et al. Risk-stratified outcomes of nonmyeloablative HLA-haploidentical BMT with high-dose posttransplantation cyclophosphamide. Blood. 2015;125(19):3024–31. doi: 10.1182/blood-2015-01-623991.

  3. Ciurea SO, Zhang M-J, Bacigalupo AA, et al. Haploidentical transplant with posttransplant cyclophosphamide vs matched unrelated donor transplant for acute myeloid leukemia. Blood. 2015;126(8):1033–40. doi: 10.1182/blood-2015-04-639831.

  4. Bashey A, Zhang X, Jackson K, et al. Comparison of Outcomes of Hematopoietic Cell Transplants from T-Replete Haploidentical Donors Using Post-Transplantation Cyclophosphamide with 10 of 10 HLA-A, -B, -C, -DRB1, and -DQB1 Allele-Matched Unrelated Donors and HLA-Identical Sibling Donors: A Multivariable Analysis Including Disease Risk Index. Biol Blood Marrow Transplant. 2016;22(1):125–33. doi: 10.1016/j.bbmt.2015.09.002.

  5. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126(3):291–9. doi: 10.1182/blood-2015-01-621664.

  6. Pozzi S, Geroldi S, Tedone E, et al. Leukaemia relapse after allogeneic transplants for acute myeloid leukaemia: predictive role of WT1 expression. Br J Haematol. 2013;160(4):503–9. doi: 10.1111/bjh.12181.

  7. Glucksberg H, Storb R, Fefer A, et al. Clinical manifestations of graft-versus-host disease in human recipients of marrow from HL-A-matched sibling donors. Transplantation. 1974;18(4):295–304. doi: 10.1097/00007890-197410000-00001.

  8. 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.

  9. Chang Y-J, Wang Y, Liu Y-R, et al. Haploidentical allograft is superior to matched sibling donor allograft in eradicating pre-transplantation minimal residual disease of AML patients as determined by multiparameter flow cytometry: a retrospective and prospective analysis. J Hematol Oncol. 2017;10(1):134. doi: 10.1186/s13045-017-0502-3.

  10. Frassoni F, Barrett AJ, Granena A, et al. Relapse after allogeneic bone marrow transplantation for acute leukaemia: a survey by the E.B.M.T. of 117 cases. Br J Haematol. 1988;70(3):317–20. doi: 10.1111/j.1365-2141.1988.tb02488.x.

  11. Bosi A, Laszlo D, Labopin M, et al. Second Allogeneic Bone Marrow Transplantation in Acute Leukemia: Results of a Survey by the European Cooperative Group for Blood and Marrow Transplantation. J Clin Oncol. 2001;19(16):3675–84. doi: 10.1200/jco.2001.19.16.3675.

  12. Verdonck L, Petersen E, Lokhorst H, et al. Donor leukocyte infusions for recurrent hematologic malignancies after allogeneic bone marrow transplantation: impact of infused and residual donor T cells. Bone Marrow Transplant. 1998;22(11):1057–63. doi: 10.1038/sj.bmt.1701496.

  13. Collins RH, Shpilberg O, Drobyski WR, et al. Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation. J Clin Oncol. 1997;15(2):433–44. doi: 10.1200/jco.1997.15.2.433.

  14. 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.

  15. Maurillo L, Venditti A, Spagnoli A, et al. Azacitidine for the treatment of patients with acute myeloid leukemia. Cancer. 2011;118(4):1014–22. doi: 10.1002/cncr.26354.

  16. Schroeder T, Czibere A, Platzbecker U, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013;27(6):1229–35. doi: 10.1038/leu.2013.7.

  17. 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.

  18. Craddock C, Labopin M, Robin M, et al. Clinical activity of azacitidine in patients who relapse after allogeneic stem cell transplantation for acute myeloid leukemia. Haematologica. 2016;101(7):879–83. doi: 10.3324/haematol.2015.140996.

  19. 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.

  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. 2011;26(3):381–9. doi: 10.1038/leu.2011.234.

  21. Schroeder T, Frobel J, Cadeddu 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.

  22. Goodyear OC, Dennis M, Jilani NY, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood. 2012;119(14):3361–9. doi: 10.1182/blood-2011-09-377044.

  23. Choi J, Ritchey J, Prior JL, 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.

  24. Cooper ML, Choi J, Karpova D, et al. Azacitidine Mitigates Graft-versus-Host Disease via Differential Effects on the Proliferation of T Effectors and Natural Regulatory T Cells In Vivo. J Immunol. 2017;198(9):3746–54. doi: 10.4049/jimmunol.1502399.

  25. Garcia-Delgado R, de Miguel D, Bailen A, et al. Effectiveness and safety of different azacitidine dosage regimens in patients with myelodysplastic syndromes or acute myeloid leukemia. Leuk Res. 2014;38(7):744–50. doi: 10.1016/j.leukres.2014.03.004.

  26. Lyons RM, Cosgriff TM, Modi SS, et al. Hematologic Response to Three Alternative Dosing Schedules of Azacitidine in Patients With Myelodysplastic Syndromes. J Clin Oncol. 2009;27(11):1850–6. doi: 10.1200/jco.2008.17.1058.

  27. Jabbour E, Giralt S, Kantarjian H, et al. Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia. Cancer. 2009;115(9):1899–905. doi: 10.1002/cncr.24198.

  28. Jabbour E, Short NJ, Montalban-Bravo G, et al. Randomized phase 2 study of low-dose decitabine vs low-dose azacitidine in lower-risk MDS and MDS/MPN. Blood. 2017;130(13):1514–22. doi: 10.1182/blood-2017-06-788497.

Factors Associated with Efficient Harvesting and Engraftment of Auto-Transplants in Multiple Myeloma Patients

II Kostroma, AA Zhernyakova, ZhV Chubukina, NYu Semenova, IM Zapreeva, SA Tiranova, SS Bessmeltsev, AV Chechetkin, SV Gritsaev

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

For correspondence: Ivan Ivanovich Kostroma, MD, PhD, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(921)784-82-82; e-mail: obex@rambler.ru

For citation: Kostroma II, Zhernyakova AA, Chubukina ZhV, et al. Factors Associated with Efficient Harvesting and Engraftment of Auto-Transplants in Multiple Myeloma Patients. Clinical oncohematology. 2019;12(1):32–6.

DOI: 10.21320/2500-2139-2019-12-1-32-36


ABSTRACT

Background. The success of autologous hematopoietic stem cell transplantation (auto-HSCT) depends on the speed of transplant engraftment which in turn is affected by the count of harvested and infused hematopoietic stem cells (HSC).

Aim. To identify predictors of auto-HSCT efficacy in multiple myeloma (MM) patients under introduction of new drugs at the phase of HSC induction and mobilization.

Materials & Methods. The results of auto-transplant harvesting and engraftment were retrospectively analyzed in 75 MM patients during 112 auto-HSCTs. Auto-transplants were harvested using cyclophosphamide and vinorelbine combined with granulocyte colony-stimulating factor (G-CSF) without plerixafor. Conditioning regimen included melphalan 200 mg/m2 or 140 mg/m2, and combination of tiothepa with melphalan. All patients received subcutaneous injections of G-CSF in post-transplantation period. Transplant engraftment was assessed according to absolute neutrophil count of ≥ 0.5 × 109/L, and thrombocyte count of ≥ 20 × 109/L.

Results. It is established that the predictors of a high CD34+ cell count in auto-transplant are a single previous induction regimen (p = 0.0315) and administration of cyclophosphamide in mobilization regimen (р = 0.0001). Transplant engraftment period is determined by auto-HSCT serial number and amount of infused CD34+ cells. Hematopoiesis regeneration after the second auto-HSCT was accelerated by more frequent use of Mel140 (р = 0.001).

Conclusion. Auto-transplant quality and engraftment period in MM patients primarily depend on the efficacy of induction therapy and the intensity of HSC mobilization regimen. Therefore, induction therapy and mobilization regimen need to be tailored to an individual patient, MM prognostic variant, probability of response to standard induction regimens, and the number of planned auto-HSCTs.

Keywords: multiple myeloma, autologous hematopoietic stem cell transplantation, auto-HSCT efficacy predictors, transplant, engraftment.

Received: May 14, 2018

Accepted: December 2, 2018

Read in PDF 


REFERENCES

  1. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома: руководство для врачей. М.: СИМК, 2016. 512 с.

    [Bessmeltsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma: rukovodstvo dlya vrachei. (Multiple myeloma: manual for physicians.) Moscow: SIMK Publ.; 2016. 512 p. (In Russ)]

  2. Менделеева Л.П., Вотякова О.М., Покровская О.С. и др. Национальные клинические рекомендации по диагностике и лечению множественной миеломы. Гематология и трансфузиология. 2016;61(1, прил. 2):1–24. doi: 10.18821/0234-5730-2016-61-1(Прил.2).

    [Mendeleeva LP, Votyakova OM, Pokrovskaya OS, et al. National clinical guidelines on diagnosis and treatment of multiple myeloma. Gematologiya i transfuziologiya. 2016;61(1, Suppl 2):1–24. doi: 10.18821/0234-5730-2016-61-1(Прил.2). (In Russ)

  3. Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med. 2003;348(19):1875–83. doi: 10.1056/NEJMoa022340.

  4. Palumbo A, Gay F, Spencer A, et al. A phase III study of ASCT vs cyclophosphamide lenalidomide dexamethasone and lenalidomide prednisone maintenance vs lenalidomide alone in newly diagnosed myeloma patients. Blood. 2013;122(21):763, abstract.

  5. Dhakal B, Szabo A, Chhabra S, et al. Autologous transplantation for newly diagnosed multiple myeloma in the era of novel agent induction: a systematic review and meta-analysis. JAMA Oncol. 2018;4(3):343–50. doi: 10.1001/jamaoncol.2017.4600.

  6. Sola C, Maroto P, Salazar R, et al. Bone Marrow Transplantation: prognostic factors of peripheral blood stem cell mobilization with cyclophosphamide and filgrastim (r-metHuG-CSF): the CD34+ cell dose positively affects the time to hematopoietic recovery and supportive requirements after high-dose chemotherapy. Hematology. 1999;4(3):195–209. doi: 10.1080/10245332.1999.11746443.

  7. Duggan PR, Guo D, Luider J, et al. Predictive factors for long term engraftment of autologous blood stem cells. Bone Marrow Transplant. 2000;26(12):1299–304. doi: 10.1038/sj.bmt.1702708.

  8. Wallington-Beddoe CT, Gottlieb DJ, Garvin F, et al. Failure to achieve a threshold dose of CD34+CD110+ progenitor cells in the graft predicts delayed platelet engraftment after autologous stem cell transplantation for multiple myeloma. Biol Blood Marrow Transplant. 2009;15(11):1386–93. doi: 10.1016/j.bbmt.2009.06.018.

  9. Stiff PJ, Micallef I, Nademanee AP, et al. Transplanted CD34+ cell dose is associated with long-term platelet count recovery following autologous peripheral blood stem cell transplant in patients with non-Hodgkin lymphoma or multiple myeloma. Biol Blood Marrow Transplant. 2011;17(8):1146–53. doi: 10.1016/j.bbmt.2010.11.021.

  10. Asfour I, Afify H, Elkourashy S, et al. CXCR4 (CD184) expression on stem cell harvest and CD34+ cells post-transplant. Hematol Oncol Stem Cell Ther. 2017;10(2):63–9. doi: 10.1016/j.hemonc.2017.01.002.

  11. Кострома И.И., Жернякова А.А., Чубукина Ж.В. и др. Заготовка гемопоэтических стволовых клеток у больных множественной миеломой: влияние предшествующей аутоТГСК терапии леналидомидом и режима мобилизации. Клиническая онкогематология. 2018;11(2):192–7. doi: 10.21320/2500-2139-2018-11-2-192-197.

    [Kostroma II, Zhernyakova AA, Chubukina ZhV, et al. Hematopoietic Stem Cell Collection in Multiple Myeloma Patients: Influence of the Lenalidomide-Based Therapy and Mobilization Regimen Prior to Auto-HSCT. Clinical oncohematology. 2018;11(2);192–7. doi: 10.21320/2500-2139-2018-11-2-192-197. (In Russ)]

  12. Stewart DA, Guo D, Morris D, et al. Superior autologous blood stem cell mobilization from dose-intensive cyclophosphamide, etoposide, cisplatin plus G-CSF than from less intensive chemotherapy regimens. Bone Marrow Transplant. 1999;23(2):111–7. doi: 10.1038/sj.bmt.1701536.

  13. Corso A, Caberlon S, Pagnucco G, et al. Blood stem cell collections in multiple myeloma: definition of a scoring system. Bone Marrow Transplant. 2000;26(3):283–6. doi: 10.1038/sj.bmt.1702514.

  14. Morris CL, Siegel E, Barlogie B, et al. Mobilization of CD34+ cells in elderly patients (≥70 years) with multiple myeloma: influence of age, prior therapy, platelet count and mobilization regimen. Br J Haematol. 2003;120(3):413–23. doi: 10.1046/j.1365-2141.2003.04107.x.

  15. Pusic I, Jiang SY, Landua S, et al. Impact of mobilization and remobilization strategies on achieving sufficient stem cell yields for autologous transplantation. Biol Blood Marrow Transplant. 2008;14(9):1045–56. doi: 10.1016/j.bbmt.2008.07.004.

  16. Lee JL, Kim SB, Lee GW, et al. Collection of peripheral blood progenitor cells: analysis of factors predicting the yields. Transfus Apher Sci. 2003;29(1):29–37. doi: 10.1016/S1473-0502(03)00097-1.

  17. Kumar S, Dispenzieri A, Lacy MQ, et al. Impact of lenalidomide therapy on stem cell mobilization and engraftment post-peripheral blood stem cell transplantation in patients with newly diagnosed myeloma. Leukemia. 2007;21(9):2035–42. doi: 10.1038/sj.leu.2404801.

  18. Mark T, Stern J, Furst JR, et al. Stem cell mobilization with cyclophosphamide overcomes the suppressive effect of lenalidomide therapy on stem cell collection in multiple myeloma. Biol Blood Marrow Transplant. 2008;14(7):795–8. doi: 10.1016/j.bbmt.2008.04.008.

  19. DiPersio JF, Stadtmauer EA, Nademanee A, et al. Plerixafor and G-CSF versus placebo and G-CSF to mobilize hematopoietic stem cells for autologous stem cell transplantation in patients with multiple myeloma. Blood. 2009;113(23):5720–6. doi: 10.1182/blood-2008-08-174946.

  20. Popat U, Saliba R, Thandi R, et al. Impairment of filgrastim-induced stem cell mobilization after prior lenalidomide in patients with multiple myeloma. Biol Blood Marrow Transplant. 2009;15(6):718–23. doi: 10.1016/j.bbmt.2009.02.011.

  21. Ruiz-Delgado GJ, Lopez-Otero A, Hernandez-Arizpe A, et al. Poor hematopoietic stem cell mobilizers in multiple myeloma: a single institution experience. Medit J Hemat Infect Dis. 2010;2(2):e2010016. doi: 10.4084/MJHID.2010.016.

  22. Mohty M, Hubel K, Kroger N, et al. Autologous haematopoietic stem cell mobilization in multiple myeloma and lymphoma patients: a position statement from the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2014;49(7):865–72. doi: 10.1038/bmt.2014.39.

  23. Silvennoinen R, Anttila P, Saily M, et al. A randomized phase II study of stem cell mobilization with cyclophosphamide + G-CSF or G-CSF alone after lenalidomide-based induction in multiple myeloma. Bone Marrow Transplant. 2016:51(3):372–6. doi: 10.1038/bmt.2015.236.

  24. Sarmiento M, Ramirez P, Parody R, et al. Advantages of non-cryopreserved autologous hematopoietic stem cell transplantation against a cryopreserved strategy. Bone Marrow Transplant. 2018;53(8):960–6. doi: 10.1038/s41409-018-0117-5.

  25. Кучер M.A., Моталкина М.С., Климова О.У. и др. Плериксафор у пациентов со сниженной мобилизационной способностью аутологичных гемопоэтических стволовых клеток. Клиническая онкогематология. 2016;9(2):155–61. doi: 10.21320/2500-2139-2016-9-2-155-161.

    [Kucher MA, Motalkina MS, Klimova OU, et al. Plerixafor in Patients with Decreased Mobilizing Ability of Autologous Hematopoietic Stem Cells. Clinical oncohematology. 2016;9(2):155–61. doi: 10.21320/2500-2139-2016-9-2-155-161. (In Russ)]

  26. Грицаев C.В., Кострома И.И., Чубукина Ж.В. и др. Сравнительная эффективность винорелбина и циклофосфана в режиме мобилизации для заготовки аутотрансплантата. Medline.ru. 2017;18:409–24.

    [Gritsaev SV, Kostroma II, Chubukina ZhV, et al. Comparative efficacy of vinorelbine and cyclophosphamide in mobilization regimen for auto-transplant harvesting. Medline.ru. 2017;18:409–24. (In Russ)]

  27. Грицаев С.В., Кузяева А.А., Кострома И.И. и др. Первый опыт мобилизации гемопоэтических стволовых клеток в периферическую кровь винорелбином у больных лимфопролиферативными заболеваниями. Медицина экстремальных ситуаций. 2017;62(4):30–5.

    [Gritsaev SV, Kuzyaeva AA, Kostroma II, et al. The first attempt to use vinorelbine for mobilization of hematopoietic stem cells into the peripheral blood in patients with lymphoproliferative disoders. Meditsina ekstremal’nykh situatsii. 2017;62(4):30–5. (In Russ)]

Correlation of CD34+ Hematopoietic Stem Cells and CFU in Peripheral Blood Apheresis Products in Patients with Malignant Lymphoproliferative Diseases Before and After Cryopreservation Prior to auto-HSCT

VA Balashova, VI Rugal’, SS Bessmel’tsev, SV Gritsaev, NYu Semenova, SV Voloshin, ZhV Chubukina, AV Shmidt, AD Garifullin, IM Zapreeva, AA Kuzyaeva, II Kostroma, AYu Kuvshinov, AV Chechetkin

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

For correspondence: Valentina Andreevna Balashova, MD, PhD, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)717-19-37; e-mail: vbspb37@mail.ru

For citation: Balashova VA, Rugal’ VI, Bessmel’tsev SS, et al. Correlation of CD34+ Hematopoietic Stem Cells and CFU in Peripheral Blood Apheresis Products in Patients with Malignant Lymphoproliferative Diseases Before and After Cryopreservation Prior to auto-HSCT. Clinical oncohematology. 2018;11(4):368–77.

DOI: 10.21320/2500-2139-2018-11-4-368-377


ABSTRACT

Aim. To establish correlation between CD34+ autologous hematopoietic stem cell (HSC) count and colony-forming units (CFU) in the same peripheral blood apheresis product samples before and after cryopreservation in multiple myeloma and lymphoma patients, and to assess clinical value of these parameters.

Materials & Methods. Cell samples of peripheral blood cytapheresis product and cell cultures were studied before and after cryopreservation in 32 multiple myeloma and 25 lymphoma patients who underwent autologous HSC transplantation. The material was analyzed using culture technique and flow cytometry.

Results. The paper provides information on the relationship between CD34+ HSC count obtained by flow cytometry, and CFU in cell culture obtained by cytapheresis of the same peripheral blood samples. A direct correlation was confirmed between CD34+ count and all the CFUs before and after cryopreservation in lymphoma patients. Correlation between CD34+ count and granulocyte-macrophage CFUs was revealed in multiple myeloma and lymphoma patients before cryopreservation.

Conclusion. The parameter of colony-forming capacity used for the assessment of the functional HSC was shown to be equally reliable criterion for condition evaluation of autotransplant proliferative pool than CD34+ cells. Both methods should be applied for qualitative and quantitative evaluation of an autotransplant for multiple myeloma and lymphoma patients.

Keywords: CD34+ cells, CFU, CFU-GM, correlation, lymphoma, multiple myeloma, apheresis, auto-HSCT.

Received: April 11, 2018

Accepted: July 28, 2018

Read in PDF 


REFERENCES

  1. Lansdorp PM. Self-renewal of stem cells. Biol Blood Marrow Transplant. 1997;3(4):171–8.

  2. Bryder D, Rossi DJ, Weissman IL. Hematopoietic stem cells: the paradigmatic tissue specific stem cell. Am J Pathol. 2006;169(2):338–46. doi: 10.2353/ajpath.2006.060312.

  3. Wodnar-Filipowicz A. Biological properties of haematopoietic stem cells. The EBMT Handbook, 6th edition; 2012. pp. 61–72.

  4. Moreb JS, Salmosinia D, Hsu J, et al. Long-term outcome after autologous stem cell transplantation with adequate peripheral blood stem cell mobilization using plerixafor and G-CSF in poor mobilizer lymphoma and myeloma patients. Adv Hematol. 2011;2011:1–8. doi: 10.1155/2011/517561.

  5. Птушкин В.В., Жуков Н.В., Миненко С.В. и др. Роль высокодозной химиотерапии с трансплантацией стволовых кроветворных клеток у больных с неходжкинскими лимфомами. Онкогематология. 2006;1–2:86–96.

    [Ptushkin VV, Zhukov NV, Minenko SV, et al. Role of high-dose chemotherapy with hematopoietic stem cell transplantation in patients with non-Hodgkin’s lymphomas. Onkogematologiya. 2006;1–2:86–96. (In Russ)]

  6. Avet-Loiseau H, Attal M, Moreau P, et al. Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroup Francophone du Myeloma. Blood. 2007;109(8):3489–95. doi: 10.1182/blood-2006-08-040410.

  7. Avet-Loiseau H, Soulier J, Fermand JP, et al. Impact of high-risk cytogenetics and prior therapy on outcomes in patients with advanced relapsed or refractory multiple myeloma treated with lenalidomide plus dexamethasone. 2010;24(3):623–8. doi: 10.1038/leu.2009.273.

  8. Dabusti M, Lanza F, Campioni D, et al. CXCR4 expression on bone marrow CD34+ cells prior to mobilization can predict mobilization adequacy in patients with hematological malignancy. J Hematother Stem Cell Res. 2003;12(4):425–34. doi: 10.1089/152581603322286051.

  9. Ratip S. Mobilization failure in hematopoietic stem cell transplantation. XXXIX Ulusal Hematoloji Kongresi. Antalya, Turkey; 2013. рр. 106–10.

  10. Артюхина З.Е., Семенова Н.Ю., Балашова В.А. и др. Кроветворная ткань и стромальное микроокружение больных множественной миеломой. Вестник гематологии. 2017;13(1):15–8.

    [Artyukhina ZE, Semenova NYu, Balashova VA, et al. Hematopoietic tissue and stromal microenvironment in patients with multiple myeloma. Vestnik gematologii. 2017;13(1):15–8. (In Russ)]

  11. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома: руководство для врачей. М.: МК, 2016. 504 с.

    [Bessmel’tsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma: rukovodstvo dlya vrachei. (Multiple myeloma: manual for doctors.) Moscow: MK Publ.; 2016. 504 p. (In Russ)]

  12. Покровская О.С., Менделеева Л.П., Гальцева И.В. и др. Мобилизация гемопоэтических клеток крови у больных миеломной болезнью. Проблемы гематологии и переливания крови. 2003;2:55–65.

    [Pokrovskaya OS, Mendeleeva LP, Gal’tseva IV, et al. Mobilization of hematopoietic cells in myeloma patients. Problemy gematologii i perelivaniya krovi. 2003;2:55–65. (In Russ)]

  13. Покровская О.С. Кроветворная ткань и стромальное микроокружение в процессе интенсивной терапии и мобилизации гемопоэтических стволовых клеток у больных множественной миеломой: Автореф. дис.… канд. мед. наук. М., 2011.

    [Pokrovskaya OS. Krovetvornaya tkan’ i stromal’noe mikrookruzhenie v protsesse intensivnoi terapii i mobilizatsii gemopoeticheskikh stvolovykh kletok u bol’nykh mnozhestvennoi mielomoi. (Hematopoietic tissue and stromal microenvironment in intensive treatment and mobilization of hematopoietic stem cells in multiple myeloma ) [dissertation] Moscow; 2011. (In Russ)]

  14. Haizmann M, O’Meara AC, Moosmann PR, et al. Efficient mobilization of PBSC with vinorelbine/G-CSF in patients with malignant lymphoma. Bone Marrow Transplant. 2009;44(2):75–9. doi: 10.1038/bmt.2008.434.

  15. Haverkos BM, McBride A, O’Donnell L, et al. An effective mobilization strategy for lymphoma patients after failed upfront mobilization with plerixafor. Bone Marrow Transplant. 2014;49(8):1052–5. doi: 10.1038/bmt.2014.90.

  16. Lansdorp PM, Sutherland HJ, Eaves CJ. Selective expression of CD45 isoforms on functional subpopulations of CD34+ hemopoietic cells from human bone marrow. J Exp Med. 1990;172(1):363–6. doi: 10.1084/jem.172.1.363.

  17. Fritsch G, Buchinger P, Printz D, et al. Rapid discrimination of early CD34+ myeloid progenitors using CD45-RA analysis. Blood. 1993;1(9):2301–9.

  18. Fritsch G, Buchinger P, Printz D. Use of flow cytometric CD34 analysis to quantify hematopoietic progenitor cells. Leuk Lymphoma. 1993;10(6):443–51. doi: 10.3109/10428199309148201.

  19. Nissen-Druey C, Tichelli A, Mayer-Monard S. Human hematopoietic colonies in health and disease. Acta Haematol. 2005;113(1):5–10. doi: 10.1159/000081987.

  20. Takano H, Ema H, Sudo K, et al. Asymmetric division and lineage commitment at the level of hematopoietic stem cells: Inference from differentiation in daughter cell and granddaughter cell pairs. J Exp Med. 2004;199(3):295–302. doi: 10.1084/jem.20030929.

  21. Sieburg HB, Cho RH, Dykstra B, et al. The hematopoietic stem compartment consists of a limited number of discrete stem cell subsets. Blood. 2006;107(6):2311–6. doi: 10.1182/blood-2005-07-2970.

  22. Guo Y, Lubbert M, Engelhard M. CD34-hematopoietic stem cells: current concepts and controversies. Stem Cell. 2003;21(1):15–20. doi: 10.1634/stemcells.21-1-15.

  23. Donahue RE, Yang YC, Clark SC. Human P40 T-cell growth factor (interleukin-9) supports erythroid colony formation. Blood. 1990;75(12):2271–5.

  24. Ema H, Suda T, Miura Y, Nakauchi H. Colony formation of clone-sorted human haematopoietic progenitors. Blood. 1990;75(10):1941–6.

  25. Serke S, Sauberlich S, Huhn D. Multiparameter flow-cytometrical quantitation of circulating CD34+ cells: correlation to the quantitation of circulating haemopoietic progenitor cells by in vitro colony-assay. Br J Haematol. 2008;77(4):453–9. doi: 10.1111/j.1365-2141.1991.tb08609.x.

  26. Bensinger WI, Longin K, Appelbaum F, et al. Peripheral blood stem cells (PBSCs) collected after recombinant granulocyte colony stimulating factor (rhG-CSF): An analysis of factors correlating with the tempo of engraftment after transplantation. Br J Haematol. 1994;87(4):825–31. doi: 10.1111/j.1365-2141.1994.tb06744.x.

  27. Bensinger WI, Appelbaum F, Rowley S, et al. Factors that influence collection and engraftment of autologous peripheral blood stem cells. J Clin Oncol. 1995;13(10):2547–55. doi: 10.1200/jco.1995.13.10.2547.

  28. Weaver CH, Haselton B, Birch R, et al. An analysis of engrafment kinetics as a function of the CD34 content of peripheral blood progenitor cell collections in 692 patients after administration of myeloablative chemotherapy. Blood. 1995;86(10):3961–9.

  29. Weaver CH, Potz J, Redmond J, et al. Engraftment and outcomes of patients receiving myeloablative therapy followed by autologous peripheral blood cells with a low CD34+ cell content. Bone Marrow Transplant. 1997;19(11):1103–10. doi: 10.1038/sj.bmt.1700808.

  30. Watts MJ, Sullivan AM, Jamieson E, et al. Progenitor-cell mobilization after low-dose cyclophosphamide and granulocyte colony-stimulating factor, an analysis of progenitor-cell quantity and quality and factors predicting for these parameters in 101 pretreated patients with malignant lymphoma. J Clin Oncol. 1997;15(2):535–46. doi: 10.1200/jco.1997.15.2.535.

  31. Serke S, Watts M, Knudsen LM, et al. In-vitro clonogenity of mobilized peripheral blood CD34 expressing cells: inverse correlation to both relative and absolute number of CD34-expressing cells. Br J Haematol. 1996;95(2):234–40. doi: 10.1046/j.1365-2141.1996.d01-1918.x.

  32. Fritsch G, Emminger W, Buchinger P, et al. CD34-positive cell proportions in peripheral blood correlate with colony-forming capacity. Exp Hematol. 1991;19(11):1079–83.

  33. Fritsch G, Emminger W, Buchinger P, et al. CD34 analysis in peripheral blood correlates with colony-forming capacity. Progr Clin Biol Res. 1992;377:531–6.

  34. Scott MA, Ager S, Apperley JF, et al. Peripheral blood progenitor cell harvesting in multiple myeloma and malignant lymphoma. Leuk Lymphoma. 1995;19(5–6):479–84. doi: 10.3109/10428199509112208.

  35. Buzzi M, Granchi D, Bacci G, et al. CD34+ cells and clonogenicity of peripheral blood stem cells during chemotherapy treatment in association with granulocyte colony stimulating factor in osteosarcoma. J Chemother. 1999;11(4):293–300. doi: 10.1179/joc.1999.11.4.293.

  36. Андреева Л.Ю., Тупицын Н.Н., Овумян Г.Ш. и др. Гемопоэтические предшественники в крови онкологических больных: взаимосвязь колониеобразования и экспрессии CD Вестник РОНЦ им. Н.Н. Блохина РАМН. 2000;11(1):5–10.

    [Andreeva LYu, Tupitsyn NN, Ovumyan GSh, et al. Hematopoietic progenitors in blood of cancer patients: relationship between colony formation and CD34 expression. Vestnik RONTs im NN Blokhina RAMN. 2000;11(1):5–10. (In Russ)]

  37. Healy LE, Nirsimloo N, Scott M, et al. In vitro proliferation by cells mobilized into the peripheral blood for collection and autologous transplantation. Exp Hematol. 1994;22(13):1278–82.

  38. Magagnoli M, Spina M, Balzarotti M, et al. IGEV regimen and a fixed dose of lenograstim: an effective mobilization regimen in pretreated Hodgkin’s lymphoma patients. Bone Marrow Transplant. 2007;40(11):1019–25. doi: 10.1038/sj.bmt.1705862.

  39. Koutna I, Peterkova M, Simara P, et al. Proliferation and differentiation potential CD133+ and CD34+ populations from the bone marrow and mobilized peripheral blood. Ann Hematol. 2011;90(2):127–37. doi: 10.1007/s00277-010-1058-2.

  40. Балашова В.А., Ругаль В.И., Грицаев С.В. и др. Колониеобразующая способность гемопоэтических стволовых клеток мобилизованной периферической крови больных множественной миеломой до и после криоконсервирования. Трансфузиология. 2016;17(4):63–70.

    [Balashova VA, Rugal’ VI, Gritsaev SV, et al. Colony-forming capacity of hematopoietic stem cells of mobilized peripheral blood in multiple myeloma patients before and after cryopreservation. Transfuziologiya. 2016;17(4):63–70. (In Russ)]

  41. Балашова В.А., Ругаль В.И., Бессмельцев С.С. и др. Колониеобразующая способность гемопоэтических стволовых клеток мобилизованной периферической крови больных злокачественными лимфомами до и после криоконсервирования. Medline. 2018;19(3):45–54.

    [Balashova VA., Rugal VI., Bessmeltsev SS. et al. Colonyforming capacity of hematopoietic stem cells of mobilized peripheral blood in patients with malignant lymphomas before and after cryopreservation. Medline. 2018;19(3):45–54. (In Russ)]

Magnetic Resonance Imaging of Bone Marrow and its Results as a Criterion for Administration of Maintenance Therapy After Auto-HSCT in Multiple Myeloma Patients

MV Solov’ev, LP Mendeleeva, GA Yatsyk, NS Lutsik, MV Firsova, EG Gemdzhian, VG Savchenko

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

For correspondence: Maksim Valer’evich Solov’ev, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-31-92; е-mail: maxsolovej@mail.ru

For citation: Solov’ev MV, Mendeleeva LP, Yatsyk GA, et al. Magnetic Resonance Imaging of Bone Marrow and its Results as a Criterion for Administration of Maintenance Therapy After Auto-HSCT in Multiple Myeloma Patients. Clinical oncohematology. 2018;11(4):360–7.

DOI: 10.21320/2500-2139-2018-11-4-360-367


ABSTRACT

Aim. To evaluate the efficacy of maintenance therapy in multiple myeloma (MM) patients after autologous hematopoietic stem cell transplantation (auto-HSCT) based on the results of MRI of bone marrow.

Materials & Methods. The study included 32 MM patients aged 36 to 66 years (median 57 years) with complete remission after a single auto-HSCT. MRI of spine and pelvic bones was performed to identify the nature of bone marrow lesions and to determine the volume of tumor tissue on the day 100 after auto-HSCT. As maintenance therapy after auto-HSCT 14 patients received daily 15 mg lenalidomide in the period from day 1 to day 21 of the 28-day treatment course within 1 year. Monitoring of 18 patients was conducted without maintenance therapy. Statistical analysis included the assessment of progression-free survival (PFS) and relapse risk relationship to clinical and laboratory parameters.

Results. Twenty patients had a positive MRI (tumor volume > 1 cm3). Zero variation of MR signal in bone marrow and detection of a < 1 cm3 tumor were regarded as a negative MRI, which was the case in 12 patients. After reaching the negative MRI the best rates of 2-year PFS were registered: 100 % with maintenance therapy and 84 % without maintenance therapy. In patients with tumor load on MR scans the 2-year PFS significantly (= 0.03) varied and accounted for 80 % in patients who received maintenance therapy vs. 33 % in patients without maintenance therapy. Administration of maintenance therapy after detecting residual tumor on MR scans on day 100 after auto-HSCT has a positive effect on PFS rates. Multivariate analysis confirmed the residual tumor on MR scans of bone marrow to be the most important parameter PFS depends on.

Conclusion. A negative MRI after auto-HSCT is a favourable prognostic factor determining a long-lasting (> 2 years) MM free period, despite the lack of maintenance therapy.

Keywords: multiple myeloma, magnetic resonance imaging (MRI), autologous hematopoietic stem cell transplantation (auto-HSCT), maintenance therapy, minimal residual disease.

Received: May 11, 2018

Accepted: August 29, 2018

Read in PDF 


REFERENCES

  1. Менделеева Л.П., Вотякова О.М., Покровская О.С. и др. Национальные клинические рекомендации по диагностике и лечению множественной миеломы. Гематология и трансфузиология. 2016;61(1, прил. 2):1–24. doi: 10.18821/0234-5730-2016-61-1(Прил.2). [Mendeleeva LP, Votyakova OM, Pokrovskaya OS, et al. National clinical guidelines on diagnosis and treatment of multiple myeloma. Gematologiya i transfuziologiya. 2016;61(1, Suppl. 2):1–24. doi: 10.18821/0234-5730-2016-61-1(Прил.2). (In Russ)]

  2. Kumar SK, Rajkumar SV, Dispenzieri A, et Improved survival in multiple myeloma and the impact of novel therapies. Blood. 2008;111(5):2516–20. doi: 10.1182/blood-2007-10-116129.

  3. Mendeleeva LP, Solovev MV, Alexeeva A, at al. Multiple Myeloma in Russia (First Results of the Registration Trial). Blood. 2017;130(Suppl 1):5408.

  4. Passweg JR, Baldomero H, Bader Р, et al. Is the use of unrelated donor transplantation leveling off in Europe? The 2016 European Society for Blood and Marrow Transplant activity survey report. Bone Marrow Transplant. 2018. doi: 10.1038/s41409-018-0153-1. [Epub ahead of print]

  5. Passweg JR, Baldomero H, Bader P, et al. Hematopoietic stem cell transplantation in Europe 2014: more than 40 000 transplants annually. Bone Marrow Transplant. 2016;51(6):786–92. doi: 10.1038/bmt.2016.20.

  6. Gay F, Oliva S, Petrucci MT, et al. Autologous transplant vs oral chemotherapy and lenalidomide in newly diagnosed young myeloma patients: a pooled analysis. Leukemia. 2017;31(8):1727–34. doi: 10.1038/leu.2016.381.

  7. Roussel M, Lauwers-Cances V, Robillard N, et al. Front-Line Transplantation Program With Lenalidomide, Bortezomib, and Dexamethasone Combination As Induction and Consolidation Followed by Lenalidomide Maintenance in Patients With Multiple Myeloma: A Phase II Study by the Intergroupe Francophone du Myelome. J Clin Oncol. 2014;32(25):2712–7. doi: 10.1200/JCO.2013.54.8164.

  8. Moreau P, San Miguel J, Sonneveld P, et al. Multiple myeloma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(Suppl 4):iv52–61. doi: 10.1093/annonc/mdx096.

  9. Syed YY. Lenalidomide: A Review in Newly Diagnosed Multiple Myeloma as Maintenance Therapy After ASCT. Drugs. 2017;77(13):1473–80. doi: 10.1007/s40265-017-0795-0.

  10. Goldschmidt H, Lokhorst HM, Mai EK, et al. Bortezomib before and after high-dose therapy in myeloma: long-term results from the phase III HOVON-65/GMMG-HD4 trial. Leukemia. 2018;32(2):383–90. doi: 10.1038/leu.2017.211.

  11. Rosinol L, Oriol A, Teruel AI, et al. Bortezomib and thalidomide maintenance after stem cell transplantation for multiple myeloma: a PETHEMA/GEM trial. Leukemia. 2017;31(9):1922–7. doi: 10.1038/leu.2017.35.

  12. Mellqvist UH, Gimsing P, Hjertner O, et al. Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial. Blood. 2013;121(23):4647–54. doi: 10.1182/blood-2012-11-464503.

  13. Sonneveld P, Schmidt-Wolf IG, van der Holt B, et al. Bortezomib induction and maintenance treatment in patients with newly diagnosed multiple myeloma: results of the randomized phase III HOVON-65/GMMG-HD4 trial. J Clin Oncol. 2012;30(24):2946–55. doi: 10.1200/JCO.2011.39.6820.

  14. McCarthy PL, Owzar K, Hofmeister C, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med. 2012;366(19):1770–81. doi: 10.1056/NEJMoa1114083.

  15. Attal M, Lauwers-Cances V, Marit G, et al. Lenalidomide Maintenance after Stem-Cell Transplantation for Multiple Myeloma. N Engl J Med. 2012;366(19):1782–91. doi: 10.1056/NEJMoa1114138.

  16. Palumbo A, Cavallo F, Gay F, et al. Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med. 2014;371(10):895–905. doi: 10.1056/NEJMoa1402888.

  17. Solovev MV, Mendeleeva LP, Pokrovskaya OS, et al. Maintenance Therapy after Autologous Haematopoietic Stem Cell Transplantation (auto-HSCT) in Multiple Myeloma Patients with and without Minimal Residual Disease (MRD). Blood. 2016;128(22):2260.

  18. Solovev MV, Mendeleeva LP, Pokrovskaya OS, et al. The Duration of MRD-Negative Status in Multiple Myeloma (MM) Patients after Auto-HSCT Is a Criterion for Prolonged Remission without Maintenance Therapy. Blood. 2017;130(Suppl 1):3294.

  19. Dutoit JC, Verstraete KL. Whole-body MRI, dynamic contrast-enhanced MRI, and diffusion-weighted imaging for the staging of multiple myeloma. Skelet Radiol. 2017;46(6):733–50. doi: 10.1007/s00256-017-2609-6.

  20. Latifoltojar A, Hall‐Craggs M, Rabin N, et al. Whole body magnetic resonance imaging in newly diagnosed multiple myeloma: early changes in lesional signal fat fraction predict disease response. Br J Haematol. 2017;176(2):222–33. doi: 10.1111/bjh.14401.

  21. Lasocki A, Gaillard F, Harrison SJ. Multiple myeloma of the spine. Neuroradiol J. 2017;30(3):259–68. doi: 10.1177/1971400917699426.

  22. Bray TJ, Singh S, Latifoltojar A, et al. Diagnostic utility of whole body Dixon MRI in multiple myeloma: A multi-reader study. PLoS One. 2017;12(7):e0180562. doi: 10.1371/journal.pone.0180562.

  23. Sabour S. Whole-body ultra-low dose computed tomography in comparison with spinal magnetic resonance imaging in the assessment of disease in multiple myeloma; Methodological issues on Diagnostic value. Br J Haematol. 2017. doi: 10.1111/bjh.14849. [Epub ahead of print]

  24. Chantry A, Kazmi M, Barrington S, et al. Guidelines for the use of imaging in the management of patients with myeloma. Br J Haematol. 2017;178(3):380–93. doi: 10.1111/bjh.14827.

  25. Moulopoulos LA, Gika D, Anagnostopoulos A, et al. Prognostic significance of magnetic resonance imaging of bone marrow in previously untreated patients with multiple myeloma. Ann Oncol. 2005;16(11):1824–8. doi: 10.1093/annonc/mdi362.

  26. Mai EK, Hielscher T, Kloth JK, et al. Association between magnetic resonance imaging patterns and baseline disease features in multiple myeloma: analyzing surrogates of tumour mass and biology. Eur Radiol. 2016;26(11):3939–48. doi: 10.1007/s00330-015-4195-0.

  27. Walker R, Barlogie B, Haessler J, et al. Magnetic resonance imaging in multiple myeloma: diagnostic and clinical implications. J Clin Oncol. 2007;25(9):1121–8. doi: 10.1200/JCO.2006.08.5803.

  28. Richardson PG, Holstein SA, Schlossman RL, et al. Lenalidomide in combination or alone as maintenance therapy following autologous stem cell transplant in patients with multiple myeloma: a review of options for and against. Expert Opin Pharmacother. 2017;18(18):1975–85. doi: 10.1080/14656566.2017.1409207.

  29. Pulte ED, Dmytrijuk A, Nie L, et al. FDA Approval Summary: Lenalidomide as Maintenance Therapy After Autologous Stem Cell Transplant in Newly Diagnosed Multiple Myeloma. Oncologist. 2018;23(6):734–9. doi: 10.1634/theoncologist.2017-0440.

  30. Sengsayadeth S, Malard F, Savani BN, et al. Posttransplant maintenance therapy in multiple myeloma: the changing landscape. Blood Cancer J. 2017;7(3):e545. doi: 10.1038/bcj.2017.23.

  31. McCarthy PL, Holstein SA, Petrucci MT, et al. Lenalidomide Maintenance After Autologous Stem-Cell Transplantation in Newly Diagnosed Multiple Myeloma: A Meta-Analysis. J Clin Oncol. 2017;35(29):3279–89. doi: 10.1200/JCO.2017.72.6679.

  32. Sonneveld P, Avet-Loiseau H, Lonial S, et al. Treatment of multiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group. Blood. 2016;127(24):2955–62. doi: 10.1182/blood-2016-01-631200.

  33. Sivaraj D, Green MM, Li Z, et al. Outcomes of Maintenance Therapy with Bortezomib after Autologous Stem Cell Transplantation for Patients with Multiple Myeloma. Biol Blood Marrow Transplant. 2017;23(2):262–8. doi: 10.1016/j.bbmt.2016.11.010.

  34. Jackson GH, Davies FE, Pawlyn C, et al. Lenalidomide Is a Highly Effective Maintenance Therapy in Myeloma Patients of All Ages; Results of the Phase III Myeloma XI Study. Blood. 2016;128(22):1143.

  35. Neben K, Lokhorst HM, Jauch A, et al. Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood. 2012;119(4):940–8. doi: 10.1182/blood-2011-09-379164.

  36. Jackson GH, Davies FE, Pawlyn C, et al. Lenalidomide Maintenance Significantly Improves Outcomes Compared to Observation Irrespective of Cytogenetic Risk: Results of the Myeloma XI Trial. Blood. 2017;130(Suppl 1):436.

  37. Mellqvist UH, Gimsing P, Hjertner O, et al. Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial. Blood. 2013;121(23):4647–54. doi: 10.1182/blood-2012-11-464503.

  38. Phase III Studies Present Additional Evidence for REVLIMID® (lenalidomide) as Maintenance Therapy in Multiple Myeloma. Available from: http://ir.celgene.com/releasedetail.cfm?releaseid=1003026 (accessed 2.05.2018).

  39. Соловьев М.В., Менделеева Л.П., Покровская О.С. и др. Множественная миелома: поддерживающая терапия после трансплантации аутологичных гемопоэтических стволовых клеток в зависимости от минимальной остаточной болезни. Терапевтический архив. 2017;89(7):25–31. doi: 10.17116/terarkh201789725-31.

    [Solovyev MV, Mendeleeva LP, Pokrovskaya OS, et al. Multiple myeloma: Maintenance therapy after autologous hematopoietic stem cell transplantation, depending on minimal residual disease. Terapevticheskii arkhiv. 2017;89(7):25–31. doi: 10.17116/terarkh201789725-31. (In Russ)]

 

Results of Molecular Monitoring in Posttransplant Period by Means of Series Investigation of WT1 Gene Expression in Patients with Acute Myeloid Leukemia

YaV Gudozhnikova, NN Mamaev, IM Barkhatov, VA Katerina, TL Gindina, AI Shakirova, SN Bondarenko, OA Slesarchuk, EI Darskaya, OV Paina, 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: Prof. Nikolai Nikolaevich Mamaev, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)233-12-43; e-mail: nikmamaev524@gmail.com

For citation: Gudozhnikova YaV, Mamaev NN, Barkhatov IM, et al. Results of Molecular Monitoring in Posttransplant Period by Means of Series Investigation of WT1 Gene Expression in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2018;11(3):241–51.

DOI: 10.21320/2500-2139-2018-11-3-241-251


ABSTRACT

Aim. To demonstrate diagnostic and prognostic significance of series measurement of WT1 expression in patients with acute myeloid leukemia (AML) after allogenic hematopoietic stem cell transplantation (allo-HSCT).

Materials & Methods. The clinical trial included 88 AML patients (38 females (43 %) and 50 males (57 %) aged 2–68, median 30 years). All the patients received allo-HSCT. Bone marrow was aspirated before (D0) and after HSCT (D+30, D+60, and D+100).

Results. The univariate analysis showed statistically significant differences in 2-year overall survival with respect to the following factors: with and without remission at the moment of HSCT (< 0.001), with and without chronic graft vs. host disease (cGVHD) (= 0.002), primary or secondary (MDS) AML (= 0,028), WT1 gene expression < and > 250 copies before HSCT (< 0.001) and at time points D+60 (= 0.012), and D+100 (< 0.001). Multivariate analysis revealed similar statistical significance of differences among patients transplanted in remission (= 0.041) and with cGVHD (= 0.03). In univariate analysis statistically significant differences in 2-year event-free survival (EFS) were found: a) in patients with allo-HSCT, either in remission or not (< 0.001); b) using HSC, but not bone marrow, as transplant source (p < 0.026); c) with normal or high WT1 expression at the stage of HSCT (< 0.001) and at time point D+100 (< 0.001); d) using HSC from related or unrelated donor (= 0.006); e) in patients with cGVHD (= 0.05). In multivariate analysis independent positive effect on EFS was observed only in patients with normal WT1 expression at D+100 (= 0.011) and with cGVHD (= 0.038). Cumulative incidence of posttransplant relapse (PTR) in AML patients with normal or high WT1 expression at the stage of HSCT within the 2-year follow-up was significantly different (28.2 vs. 58.9 %; = 0.002), also in measurements of this parameter at D+60 and D+100 (= 0.015 and < 0.001, respectively). In 1/4 of patients cytological relapses (cPTR) appeared considerably later than molecular relapses (mPTR), i.e. 13–489 days later (median 35 days), which is accounted for by early preventive therapy aimed at cPTR prophylaxis against the background of already recorded mPTR. According to our data, GVHD plays a crucial role in cPTR management.

Conclusion. Phenomenon of WT1 expression normalization after allo-HSCT in AML patients proves to have a high diagnostic and prognostic significance. Introduction of this approach into clinical practice seems highly advisable for national oncohematological centers.

Keywords: acute myeloid leukemia, allo-HSCT, posttransplant relapse, diagnostics and treatment with molecular monitoring of WT1 expression, graft vs. host disease.

Received: January 20, 2018

Accepted: April 18, 2018

Read in PDF 


REFERENCES

  1. Cilloni D, Gottardi E, De Micheli D, et al. Quantitative assessment of WT1 expression by real time quantitative PCR may be a useful tool for monitoring minimal residual disease in acute leukemia patients. Leukemia. 2002;16(10):2115–21. doi: 10:1038/sj.leu.2402675.
  2. Cilloni D, Gottardi E, Fava M, et al. Usefulness of quantitative assessment of the WT1 gene transcript as a marker for minimal residual disease detection. Blood. 2003;102(2);773–4. doi: 1182/blood-2003-03-0980.
  3. Ogawa H, Tamaki H, Ikegame K, et al. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogeneic stem cell transplantation in acute type leukemia. Blood. 2003;101(5):1698–704. doi: 1182/blood-2002-06-1831.
  4. Zhao X-S, Jin S, Zhu H-H, et al. Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2011;47(4):499–507. doi: 10.1038/bmt.2011.121.
  5. 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: 10.21320/2500-2139-2015-8-3-309-320. (In Russ)]
  6. Мамаев Н.Н., Гудожникова Я.В., Горбунова А.В.  Гиперэкспрессия гена WT1при злокачественных опухолях системы крови: теоретические и клинические аспекты (обзор литературы). Клиническая онкогематология. 2016;9(3):257–64. doi: 10.21320/2500-2139-2016-9-3-257-264.[Mamaev NN, Gudozhnikova YaV, Gorbunova AV. WT1 Gene Overexpression in Oncohematological Disorders: Theoretical and Clinical Aspects (Literature Review). Clinical oncohematology. 2016;9(3):257–64. doi: 10.21320/2500-2139-2016-9-3-257-264. (In Russ)]
  7. Call KM, Gieser T, Ito CI, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor gene locus. Cell. 1990;60(3):509–20. doi: 10:1016/0092-8674(90)90601-a.
  8. Rose EA, Glaser T, Jones C, et al. Complete physical map of the WAGR region of 11p13 localizes a candidate Wilms’ tumor gene. 1990;60(3):495–508. doi: 10.1016/0092-8674(90)90600-j.
  9. Miwa H, Beran M, Saunders GF. Expression of the Wilms’ tumor gene (WT1) in human leukemias. Leukemia. 1992;6(5):405–9.
  10. Inoue K, Sugiyama H, Ogava H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood. 1994;84(9):3071–9.
  11. Inoue K, Ogawa H, Sonoda Y, et al. Aberrant overexpression of the Wilms’ tumor gene (WT1) in human leukemia. Blood. 1997;88(4):1405–12.
  12. Cilloni D, Gottardi E, Messa F, et al. Significant correlation between the degree of WT1 expression and the International Scoring System score in patients with myelodysplastic syndromes. J Clin Oncol. 2003;21(10):1988–95. doi: 10.1200/jco.2003.10.503.
  13. Alonso-Domingues JM, Tenorio M, Velasco D, et al. Correlation of WT1 expression with the burden of total and residual leukemic blasts in bone marrow samples of acute myeloid leukemia patients. Cancer Genet. 2012;205(4):190–1. doi: 10.1016/j.cancergen.2012.02.008.
  14. Cilloni D, Messa F, Arruga F, et al. Early prediction of treatment outcome in acute myeloid leukemia by measurement of WT1 transcript levels in peripheral blood samples collected after chemotherapy. Haematologica. 2008;93(6):921–4. doi: 10.3324/haematol.12165.
  15. Ogava H, Ikegame K, Kawakami M, Tamaki H. WT1 gene transcript assay for relapse in acute myeloid leukemia after transplantation. Leuk Lymphoma. 2004;45(9):1747–53. doi: 10.1080/10428190410001687503.
  16. Pozzi S, Geroldi S, Tedone E, et al. Leukemia relapse after allogeneic transplant for acute myeloid leukemia: predictive role of WT1 expression. Br J Haematol. 2013;160(4);503–9. doi: 10.1111/bjh.12181.
  17. Nendedeu J, Esquirol A, Carricondo M, et al. Bone marrow WT1 levels in allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia and myelodysplasia: Clinically relevant time-points and 100 copies threshold value. Biol Blood Marrow Transplant. 2017;24(1):55–63. doi: 10.1016/j.bbmt.2017.09.001.
  18. Cilloni D, Saglio G, Gottardi E, et al. WT1 as universal marker for minimal residual disease detection and quantification in myeloid leukemias and in myelodysplastic syndrome. Acta Hematol. 2004;112(1–2):79–84. doi: 10.1159/000077562.
  19. Candoni A, Toffoletti E, Galina R, et al. Monitoring of minimal residual disease by quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem cell transplantation in acute myeloid leukemia. Clin Transpl. 2011;25(2):308–16. doi: 10.1111/j.1399-0012.2010.01251.x.
  20. Kwon M, Martinez-Laperche C, Infante M, et al. Evaluation of minimal residual disease by real-time quantitative PCR of Wilms’ Tumor 1 expression in patients with acute myelogenous leukemia after allogeneic stem cell transplantation: Correlation with flow cytometry and chimerism. Biol Blood Marrow Transplant. 2012;18(8):1235–42. doi: 10.1016/j.bbmt.2012.01.012.
  21. Polak J, Hajkova H, Haskovec C, et al. Quantitative monitoring of WT1 expression in peripheral blood before and after allogeneic stem cell transplantation for acute myeloid leukemia – a useful tool for early detection of minimal residual disease. Neoplasma. 2013;60(01):74–82. doi: 10.4149/neo_2013_011.
  22. Lapillone H, Renneville A, Auvrignon A, et al. High WT1 expression after induction therapy predicts high risk or relapse and death in pediatric acute myeloid leukemia. J Clin Oncol. 2006;24(10):1507–15. doi: 10.1200/jco.2005.03.5303.
  23. Messina C, Sala E, Carrabba M, et al. Early post-allogeneic transplantation WT1 transcript positivity predicts AML relapse. 40th EBMT Meeting. 30 March – 2 April; Milan, Italy; 2014: Abstract P239.
  24. Mear J-B, Salaun V, Dina N, et al. WT1 and flow cytometry minimal residual disease follow-up after allogeneic transplantation in practice. 40th EBMT Meeting. 30 March – 2 April; Milan, Italy; 2014: Abstract P655.
  25. Capelli D, Attolico I, Saraceli F, et al. Early cumulative incidence of relapse in 80 acute myeloid leukemia patients after chemotherapy and transplant post-consolidation treatment prognostic role of post-induction WT1. 40th EBMT Meeting. 30 March – 2 April; Milan, Italy; 2014: Abstract P287.
  26. Rossi G, Carella AM, Minervini MM, et al. Optimal time-points for minimal residual disease monitoring change on the basis of the method used in patients with acute myeloid leukemia who underwent allogeneic stem cell transplantation: A comparison between multiparameter flow cytometry and Wilms’ tumor 1 expression. Leuk Res. 2015;39(2):138–43. doi: 1016/j.leukres.2014.11.011.