Efficacy of Chemotherapy in Acute Leukemia Patients Resistant to Previous Standard Treatment According to the Series Measurement of WT1 Gene Expression

NN Mamaev, YaV Gudozhnikova, TL Gindina, IM Barkhatov, AI Shakirova, VA Katerina, MV Gubina, ES Nikolaeva, EV Semenova, OV Paina, EI Darskaya, OV Pirogova, VV Porunova, IS Moiseev, IA Mikhailova, BI Ayubova, VM Kravtsova, SN Bondarenko, LS Zubarovskaya, BV Afanas’ev

IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022

For correspondence: Nikolai Nikolaevich Mamaev, PhD, Professor, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; Tel.: +7(812)233-12-43; e-mail: nikmamaev524@gmail.com

For citation: Mamaev NN, Gudozhnikova YaV, Gindina TL, et al. Efficacy of Chemotherapy in Acute Leukemia Patients Resistant to Previous Standard Treatment According to the Series Measurement of WT1 Gene Expression. Clinical oncohematology. 2018;11(1):78-88.

DOI: 10.21320/2500-2139-2018-11-1-78-88


ABSTRACT

Aim. To estimate the efficacy of chemotherapy in acute leukemia patients resistant to previous standard treatment according to the series measurement of WT1 expression.

Materials & Methods. The series measurement of WT1 expression formed the basis of the efficacy estimation of induction chemotherapy in 31 patients (15 men and 16 women aged from 3 months to 68 years; the median age was 28 years) with prognostically unfavourable variants of acute myeloid (AML) and lymphoblastic leukemia (ALL) (23 AML and 8 ALL patients). The WT1 gene expression was measured at baseline and 2–3 weeks after the treatment by the quantitative real-time PCR. The threshold level for detection was 250 copies of WT1/104 copies of ABL. The cytogenetic profile of leukemia cells was assessed by standard cytogenetics and FISH.

Results. The baseline expression level of WT1 varied from 305 to 58,569 copies/104 copies of ABL. The expected reduction of WT1 expression after the first induction chemotherapy treatment was reported in 22/23 (96 %) AML patients and in 6/8 (75 %) ALL patients. According to our results WT1 expression reached the threshold in 13/31 (42 %) patients, including 9 AML patients and 4 ALL patients. After 11/31 (35 %) patients received the second course of treatment, WT1 expression level became normal in 8 cases (5 ALL and 3 AML patients). Despite high dose chemotherapy, HSCT and such agents as blinatumomab and gemtuzumab, an unfavourable outcome was observed in 18/31 (58 %) patients including 6 patients with complex karyotype (CK+) and 2 patients with monosomal karyotype (MK+). Once the MK+ and CK+ combination was observed, in another case the MK+ was combined with the prognostically unfavourable inv(3)(q21q26) inversion.

Conclusion. Our results show that the molecular monitoring should be included as part of treatment of the prognostically unfavourable acute leukemia. The WT1 gene was shown to be the most appropriate marker. WT1 expression was shown to correlate with the common fusion genes allowing to estimate the blast cell count at the molecular level.

Keywords: acute leukemia, induction chemotherapy, molecular monitoring, WT1.

Received: August 18, 2017

Accepted: November 12, 2017

Read in PDF 


REFERENCES

  1. Cilloni D, Gottardi A, 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. Leikemia. 2002;16(10):2115–21. doi: 10.1038/sj.leu.2402675.
  2. Hochenstein P, Hastie ND. The many facets of the Wilms’ tumor gene, WT1. Hum Mol Genet. 2006;15(15):R196–R201. doi: 10.1093/hmg/ddl196.
  3. Ujj Z, Buglyo G, Udvardy M, et al. WT1 overexpression affecting clinical outcome in non-Hdgkin lymphomas and adult acute lymphoblastic leukemia. Pathol Oncol Res. 2014;20(3):20565–70. doi: 10.1007/s12253-013-9729-7.
  4. Мамаев Н.Н., Гудожникова Я.В., Горбунова А.В. Гиперэкспрессия гена WT1 при 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)]
  5. Miwa H, Beran M, Anders GF. Expression of the Wilms’ tumor gene (WT1) in human leukemias. Leukemia. 1992;6(5):405–9.
  6. Bergmann L, Miething C, Maurer U, et al. High levels of Wilms’ tumor gene (WT1) mRNA in acute myeloid leukemia are associated with a worse long-turn outcome. Blood. 1997;90(3):1217–25.
  7. Lasa A, Carricondo M, Estevii S, et al. WT1 monitoring in core binding factor AML comparison with specific chimeric products. Leuk Res. 2009;33(12):1643–9. doi: 10.1016/j.leukres.2009.03.046.
  8. Zhao XS, Jin S, Zhu H-Y, et al. Wilms’ tumor 1 gene 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.
  9. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Трансплантация гемопоэтических стволовых клеток при остром миелоидном лейкозе с транслокацией t(8;21)(q22;q22). Клиническая онкогематология. 2013;6(4):439–44.[Mamayev NN, Gorbunova AV, Gindina TL, et al. Hematopoietic Stem Cell Transplantation in AML Patients with t(8;21)(q22;q22) Translocation. Klinicheskaya onkogematologiya. 2013;6(4):439–44. (In Russ)]
  10. Мамаев Н.Н., Семенова Е.В., Станчева Н.В. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при остром лимфобластном лейкозе с транслокацией t(12;21)(p13;q22). Клиническая онкогематология. 2014;7(3):327–34. [Mamaev NN, Semenova EV, Stancheva NV, et al. Allogeneic Hematopoietic Stem Cell Transplantation for ALL Patients with t(12;21)(p13;q22) Translocation. Klinicheskaya onkogematologiya. 2014;7(3):327–34. (In Russ)]
  11. 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)]
  12. Гиршова Л.Л., Будаева И.Г., Овсянникова Е.Г. и др. Прогностическое значение и корреляция динамики гиперэкспрессии гена WT1 и мутации гена NPM1 у пациентов с острым миелобластным лейкозом. Клиническая онкогематология. 2017;10(4):485–93. doi: 10.21320/2500-2139-2017-10-4-485-493. [Girshova LL, Budaeva IG, Ovsyannikova EG, et al. Prognostic Value and Correlation Between WT1 Overexpression and NPM1 Mutation in Patients with Acute Myeloblastic Leukemia. Clinical oncohematology. 2017;10(4):485–93. doi: 10.21320/2500-2139-2017-10-4-485-493. (In Russ)]
  13. Alonso-Dominiquez 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. (Letter to the Editor). doi: 10.1016/j.cancergen.2012.02.008.
  14. Cilloni D, Giuseppe S, Gottardi E, et al. WT1 as a 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.
  15. Weisser M, Kern W, Rauhut S, et al. Prognostic impact of RT-PCR-based quantification of WT1 gene expression during MRD monitoring of acute myeloid leukemia. Leukemia. 2005;19(8):1416–23. doi: 10.1038/sj.leu.2403809.
  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. Ostergaard M, Olesen LH, Hasle H, et al. WT1 gene expression: an excellent tool for monitoring minimal residual disease in 70% of acute myeloid leukemia patients – results from a single-centre study. Br J Haematol. 2004;125(5):590–600. doi: 10.1111/j/1365-2141.2004.04952.x.
  18. Candoni A, Toffoletti E, Gallina 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 Transplant. 2011;25(2):308–16. doi: 10..1111/j.1399-0012.201001251.x.
  19. Rossi G, Minervini MM, Carella AM, et al. Comparison between multiparameter flow cytometry and WT1 RNA quantification in monitoring minimal residual disese in acute myeloid leukemia without specific molecular targets. Leuk Res. 2011;36(4):401–6. doi: 10.1016/j.leukres.2011.11.020.
  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. Nomdedeu JF, Esquirol A, Carricondo M, et al. Bone marrow WT1 levels in allogeneic hematopoietic stem cell transplant (HCT) for acute myeloid leukemia and myelodysplasia: Clinically relevant time-points and 100 copies threshold value. Biol Blood Marrow Transplant. 2018;24(1):55–63. doi: 10.1016/j.bbmt.2017.09.001.
  22. Ogawa H, Tamaki Y, Ikeegame K, et al. The usefulness of monitoring WT1 gene transcripts for the prediction of relapse following allogeneic stem cell transplantation in acute type leukemia. Blood. 2003;101(5):1698–704. doi: 10.1182/blood-2002-06-1831.
  23. Ommen HB, Nyvold CG, Braendstrup K, et al. Relapse prediction in acute myeloid leukemia patients in complete remission using WT1 as a molecular marker: development of a mathematical model to predict time from molecular to clinical relapse and define optimal sampling intervals. Br J Haematol. 2008;141(6):782–91. doi: 10.1111/j.1365-2141.2008.07132.x.
  24. Lange T, Hubmann M, Burkhurdt R, et al. Monitoring of WT1 expression in PB and CD34+ donor chimerism of BM predicts early relapse in AML and MDS patients after hematopoietic cell transplantation with reduced-intensity conditioning. Leukemia. 2011;25(3):498–505. doi: 10.1038/leu.2010.283.
  25. Pozzi S, Geraldi S, Tedfone E, et al. Leukemia relapse after allogeneic transplants for acute myeloid leukemia: predictive role of WT1 expression. Br J Haematol. 2013;160(4):503–9. doi: 10.1111/bjh.12181.
  26. Yoon JH, Kim HJ, Shin SH, et al. BAALC and WT1 expressions from diagnosis to hematopoietic stem cell transplantation: consecutive monitoring in adult patients with core-binding-factor-positive AML. Eur J Haematol. 2013;91(2):112–21. doi: 10.1111/ejh.12142.
  27. Yoon JH, Kim HJ, Kim JW, et al. Identification of molecular and cytogenetic risk factors for unfavorable core-binding factor-positive adult AML with post-remission treatment outcome analysis including transplantation. Bone Marrow Transplant. 2014;49(12):1466–74. doi: 10.1038/bmt.2014.180.
  28. Messina C, Sala E, Carraba M, et al. Early post-allogeneic transplantation WT1 transcript positivity predicts AML relapse. Bone Marrow Transplant. 2015;50(S1):236–7. doi: 10.1038/bmt.2015.29.
  29. Boublikova L, Kalinova M, Ryan J, et al. Wilms’ tumor gene 1 (WT1) expression in childhood acute lymphoblastic leukemia: a wide range of WT1 expression levels, its impact on prognosis and minimal residual disease monitoring. Leukemia. 2006;20(2):254–63. doi: 10.1038/sj.leu.2404047.
  30. Lapillonne H, Renneville A, Auvrignon A, et al. High WT1 expression after induction therapy predicts high risk of relapse and death in pediatric acute myeloid leukemia. J Clin Oncol. 2006;24(10):1507–15. doi: 10.1200/jco.2005.03.5303.
  31. Busse A, Gokbuget N, Sechl JM, et al. Wilms’ tumor gene 1 (WT1) expression in subtypes of acute lymphoblastic leukemia (ALL) of adults and impact on clinical outcome. Ann Hematol. 2009,88(12):1199–205. doi: 10.1007/s00277-009-0746-2.
  32. .Sadek HA, El-Metnawey WH, Shaheen IA, et al. Quantitative assessment of Wilms’ tumor 1 (WT1) gene transcripts in Egyptian acute lymphoblastic leukemia patients. J Invest Med. 2011;59(8):1258–62. doi: 10.2130/JIM.0b013e31822a24f7.33.
  33. Mossalam GI, Abdel Hamid TM, Mahmoud HK. Prognostic significance of WT1 expression at diagnosis and end of induction in Egyptian adult acute myeloid leukemia patients. Hematology. 2013;18(2):69–73. doi: 10.1179/1607845412Y.0000000048.34.
  34. Capelli D, Attolico I, Saraceli F, et al. Early cumulative incidence of relapse in 80 acute leukemia patients after chemotherapy and transplant post-consolidation treatment prognostic role of post-induction WT1. Bone Marrow Transplant. 2015;50(S1):262–3. doi: 10.1038/bmt.2015.29.35.
  35. Ujj Z, Buglyo G, Udvardy M, et al. WT1 expression in adult acute myeloid leukemia: assessing its presence, magnitude and temporal changes as prognostic factors. Pathol Oncol Res. 2016;22(1):217–21. doi: 10.1007/s12253-015-0002-0.
  36.  Гиршова Л.Л., Овсянникова Е.Г., Кузин С.О. и др. Молекулярный мониторинг уровня транскрипта RUNX1-RUNX1T1 при острых миелобластных лейкозах на фоне терапии. Клиническая онкогематология. 2016;9(4):456–64. doi: 10.21320/2500-2139-2016-9-4-456-464. [Girshova LL, Ovsyannikova EG, Kuzin SO, et al. Molecular Monitoring of RUNX1-RUNX1T1 Transcript Level in Acute Myeloblastic Leukemias on Treatment. Clinical oncohematology. 2016;9(4):456–64. doi: 10.21320/2500-2139-2016-9-4-456-464. (In Russ)]
  37. Nomdedeu JF, Hoyos M, Carricondo M, et al. Bone marrow WT1 levels at diagnosis, post-induction and post-intensification in adult de novo AML. Leukemia. 2013;27(11):2157–64. doi: 10.1038/leu.2013.111.38.
  38. Schmid C, Schleuning M, Tischer J, et al. Early allo-SCT for AML with a complex aberrant karyotype – results from a prospective pilot study. Bone Marrow Transplant. 2012;47(1):46–53. doi: 10.1038/bmt.2011.15.39.
  39. Мамаев Н.Н, Горбунова А.В, Гиндина Т.Л. и др. Стойкое восстановление донорского гемопоэза у больной с посттрансплантационным рецидивом острого миеломонобластного лейкоза с inv(3)(q21q26), моносомией 7 и экспрессией онкогена EVI1 после трансфузий донорских лимфоцитов и использования гипометилирующих агентов. Клиническая онкогематология. 2014;7(1):71–5. [Mamayev NN, Gorbunova AV, Gindina TL, et al. Stable donor hematopoiesis reconstitution after post­transplantation relapse of acute myeloid leukemia in patient with inv(3)(q21q26), –7 and EVI1 oncogene overexpression treated by donor lymphocyte infusions and hypomethylating agents. Klinicheskaya onkogematologiya. 2014;7(1):71–5. (In Russ)
  40. Гиндина Т.Л., Мамаев Н.Н., Паина О.В. и др. Острый лимфобластный лейкоз c транслокацией t(4;11)(q21;q23)/KMT2A-AFF1: результаты аллогенной трансплантации гемопоэтических стволовых клеток у детей и взрослых. Клиническая онкогематология. 2017;10(3):342–50. doi: 10.21320/2500-2139-2017-10-3-342-350. [Gindina TL, Mamaev NN, Paina OV, et al. Acute Lymphoblastic Leukemia with t(4;11)(q21;q23)/KMT2A-AFF1 Translocation: The Results of Allogeneic Hematopoietic Stem Cells Transplantation in Children and Adults. Clinical oncohematology. 2017;10(3):342–50. doi: 10.21320/2500-2139-2017-10-3-342-350. (In Russ)]

Prognostic Value and Correlation Between WT1 Overexpression and NPM1 Mutation in Patients with Acute Myeloblastic Leukemia

LL Girshova, IG Budaeva, EG Ovsyannikova, SO Kuzin, DV Motorin, RSh Badaev, DB Zammoeva, VV Ivanov, KV Bogdanov, OS Pisotskaya, YuV Mirolyubova, TS Nikulina, YuA Alekseeva, AYu Zaritskii

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

For correspondence: Irina Garmaevna Budaeva, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel.: +7(931)351-07-06; e-mail: irina2005179@mail.ru

For citation: Girshova LL, Budaeva IG, Ovsyannikova EG, et al. Prognostic Value and Correlation Between WT1 Overexpression and NPM1 Mutation in Patients with Acute Myeloblastic Leukemia. Clinical oncohematology. 2017;10(4):485–93 (In Russ).

DOI: 10.21320/2500-2139-2017-10-4-485-493


ABSTRACT

Background. Acute myeloblastic leukemia (AML) with NPM1 mutation amounts to 30 % of all AML and is characterized by good prognosis with the exception of cases with FLT3-ITD mutation. Despite the good prognosis, the likelihood of relapses in patients with NPM1 mutation may significantly differ. Thus, the estimation of the minimal residual disease (MRD) after chemotherapy and during follow-up is becoming increasingly important. This approach will make it possible to predict the sensitivity of a tumoral clone to chemotherapy.

Aim. To evaluate the prognostic value of highly specific marker (NPM1 mutation) and non-specific marker (WT1 overexpression) of MRD, as well as to identify the correlation between the levels of NPM1 and WT1 at different stages of therapy and in the follow-up period.

Materials & Methods. The research included 14 patients with AML. All patients had the NPM1 mutation and WT1 overexpression: 50 % of patients had additional molecular markers (BAALC overexpression, FLT3-ITD, DNMT3A, and MLL mutations). Real-time PCR was used for long-term monitoring of WT1 expression levels and NPM1 mutation.

Results. The median decrease of NPM1 levels after the induction therapy was 3 log. All patients had relapses, NPM1 mutation, and lower rates of OS/RFS, which significantly correlated with prognostically negative molecular markers. There were no statistically significant differences in RFS in groups with the decrease of WT1 expression level < 2 log and > 2 log on day 28 of treatment. At the same time, the decrease of WT1 expression by > 2 log was associated with significant differences in early relapses, which correlated with the decrease of NPM1 levels (> and < than 3 log) is revealed. RFS rates were higher in patients with WT1 expression level of < 100 per 104 copies ABL on day 28 and WT1 of < 250 per 104 copies ABL on day 14 of treatment. WT1 expression was significantly lower on days 14 and 28 in patients with NPM1 decrease of > 3 log on day 28. The decrease in WT1 expression of < 100 per 104 copies ABL on day 28 was more common in patients with isolated NPM1 mutation, compared to patients with additional negative molecular markers.

Conclusion. The decrease in NPM1 levels after the induction therapy may serve as reliable prognostic marker of RFS and OS rates. New correlation between the degree of NPM1 reduction and the presence of additional molecular markers was established. Highly specific (NPM1 mutation) was shown to be more specific compared to non-specific markers (WT1 overexpression). The research showed the predictive value of a lower limit level of WT1 on day 28 of treatment (100 per 104 copies ABL), and for the first time, the importance of the early assessment WT1 expression reduction on day 14 of induction therapy.

Keywords: acute myeloblastic leukemia, AML, NPM1, WT1, molecular monitoring.

Received: February 22, 2017

Accepted: May 26, 2017

Read in PDF


REFERENCES

  1. Dohner H, Estey E, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.
  2. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–47.  doi: 10.1182/blood-2016-08-733196.
  3. Yin C.C. Detection and molecular monitoring of Minimal residual diseases in acute myeloid leukemias. J Mol Biomark Diagn. 2012;3(2):1000e105–6. doi: 10.4172/2155-9929.1000e106.
  4. Hourigan CS, Karp JE. Minimal residual disease in acute myeloid leukaemia. Nat Rev Clin Oncol. 2013;10(8):460–71. doi: 10.1038/nrclinonc.2013.100.
  5. Gabert J, Beillard E, van der Velden VH, et al. Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia–a Europe Against Cancer program. Leukemia. 2003;17(12):2318–57. doi: 10.1038/sj.leu.2403135.
  6. 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.
  7. Cilloni D, Saglio G. Usefulness of quantitative assessment of Wilms tumor suppressor gene expression in chronic myeloid leukemia patients undergoing imatinib therapy. Semin Hematol. 2003;40:37–41. doi: 10.1053/shem.2003.50040.
  8. Ogawa H, Tamaki H, Ikegame K, et al. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogenic stem cell transplantation in acute type leukemia. Blood. 2003;101(5):1698–702. doi: 10.1182/blood-2002-06-1831.
  9. Cilloni D, Messa F, Rosso V, et al. Increase sensitivity to chemotherapeutical agents and cytoplasmatic interaction between NPM leukemic mutant and NF-kappaB in AML carrying NPM1 mutations. Leukemia. 2008;22(6):1234–40. doi: 10.1038/leu.2008.68.
  10. 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.
  11. Pozzi S, Geroldi S, Tedone E, et al. Leukemia 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.
  12. Grazia CD, Pozzi S, Geroldi S, et al. Wilms Tumor 1 Expression and Pre-Emptive Immunotherapy in Patients with Acute Myeloid Leukemia Undergoing an Allogeneic Hemopoietic Stem Cell Transplant. Biol Blood Marrow Transplant. 2016;22(7):1242–6. doi: 10.1016/j.bbmt.2016.03.005.
  13. Nomdederu J, Hoyos M, Carricondo M, et al. Bone marrow WT1 levels at diagnosis, post-induction and post-intensification in adult de novo AML. Leukemia. 2013;27(11):2157–64. doi: 10.1038/leu.2013.111.
  14. Schnittger S, Kern W, Tschulik C, et al. Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Blood. 2009;114(11):2220–31. doi: 10.1182/blood-2009-03-213389.
  15. Falini B, Nicoletti I, Martelli M, et al. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood. 2007;109(3):874–85. doi: 10.1182/blood-2006-07-012252.
  16. Chang JH, Olson MO. Structure of the gene for rat nucleolar protein B23. J Biol Chem. 1990;265(30):18227–33.
  17. Wang W, Budhu A, Forgues M, et al. Temporal and spatial control of nucleophosmin by the Ran-Crm1 complex in centrosome duplication. Nat Cell Biol. 2005;7(8):823–30. doi: 10.1038/ncb1282.
  18. Ghandforoush NA, Chahardouli B, Rostami S,et. al. Evaluation of Minimal Residual Disease in Acute Myeloid Leukemia with NPM1 Marker. Int J Hematol Oncol Stem Cell Res. 2016;10(3):147–52.
  19. Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352(3):254–66. doi: 10.1056/NEJMoa041974.
  20. Nafea D, Rahman MA, Boris D, et al. Incidence and prognostic value of NPM1 and FLT3 gene mutations in AML with normal karyotype. Open Hematol J. 2011;5(1):14–20. doi: 10.2174/1874276901105010014.
  21. Schneider F, Hoster E, Unterhalt M, et al. NPM1 but not FLT3-ITD mutations predict early blast cell clearance and CR rate in patients with normal karyotype AML (NK-AML) or high-risk myelodysplastic syndrome (MDS). Blood. 2009;113(21):5250–3. doi: 10.1182/blood-2008-09-172668.
  22. Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106(12):3740–6. doi: 10.1182/blood-2005-05-2164.
  23. Schnittger S, Schoch C, Kern Q. Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype. Blood. 2005;106(12):3733–9. doi: 10.1182/blood-2005-06-2248.
  24. Kronke J, Schlenk RF, Jensen KO, et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a Study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol. 2011;29(19):2709–16. doi: 10.1200/JCO.2011.35.0371.
  25. Gorin N-C, Labopin M, Meloni G, et al. Impact of FLT3 ITD/NPM1 mutation status in adult patients with acute myelocytic leukemia autografted in first remission. Haematologica. 2013;98(2):e12–4. doi: 10.3324/haematol.2012.064436.
  26. Patel JL, Schumacher JA, Frizzell K, et al. Coexisting and cooperating mutations in NPM1-mutated acute myeloid leukemia. Leukemia Research 2017;56:7–12. doi: 10.1016/j.leukres.2017.01.027.
  27. Yoon J, Kim H, Shin S, et al. Implication of higher BAALC expression in combination with other gene mutations in adult cytogenetically normal AML. Leuk Lymphoma. 2013;55(1):110–20. doi: 10.3109/10428194.2013.800869.
  28. Tiribellia M, Raspadorib D, Geromin A, et al. High CD200 expression is associated with poor prognosis in cytogenetically normal acute myeloid leukemia, even in FlT3-ITD/NPM1+ patients. Leuk Res. 2017;58:31–8. doi: 10.1016/j.leukres.2017.04.001.
  29. Damiani D, Tiribelli M, Raspadori D, et al. Clinical impact of CD200 expression in patients with acute myeloid leukemia and correlation with other molecular prognostic factors. Oncotarget. 2015;6(30):30212–21. doi: 10.18632/oncotarget.4901.
  30. Hubmann M, Kohnke T, Hoster E, et al. Molecular response assessment by quantitative real-time polymerase chain reaction after induction therapy in NPM1-mutated patients identifies those at high risk of relapse. Haematologica. 2014;99(8):1317–25. doi: 10.3324/haematol.2014.104133.
  31. Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106(12):3740–6. doi: 10.1182/blood-2005-05-2164.
  32. Rollig C, Bornhаuser M, Kramer M, et al. Allogeneic Stem-Cell Transplantation in Patients with NPM1-Mutated Acute Myeloid Leukemia: Results from a Prospective Donor Versus No-Donor Analysis of Patients After Upfront HLA Typing Within the SAL-AML 2003 Trial. J Clin Oncol. 2015;33(5):403–10. doi: 10.1200/JCO.2013.54.4973.
  33. Balsat M, Renneville A, Thomas X, et al. Postinduction Minimal Residual Disease Predicts Outcome and Benefit From Allogeneic Stem Cell Transplantation in Acute Myeloid Leukemia With NPM1 Mutation: A Study by the Acute Leukemia French Association Group. J Clin Oncol. 2016;35(2):185–93. doi: 10.1200/JCO.2016.67.1875.
  34. Kristensen T, Mоller MB, Friis L, et al. NPM1 mutation is a stable marker for minimal residual disease monitoring in acute myeloid leukaemia patients with increased sensitivity compared to WT1 expression. Eur J Haematol. 2011;87(5):400–8. doi: 10.1111/j.1600-0609.2011.01673.х.
  35. Barragan E, Pajuelo JC, Ballester S, et al. Minimal residual disease detection in acute myeloid leukemia by mutant nucleophosmin (NPM1): comparison with WT1 gene expression. Clin Chim Acta. 2008;395(1–2):120–3. doi: 10.1016/j.cca.2008.05.021.
  36. Gorello P, Cazzaniga G, Alberti F, et al. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations. Leukemia. 2006;20(6):1103–8. doi: 10.1038/sj.leu.2404149.
  37. Paietta E. Minimal residual disease in acute myeloid leukemia: coming of age. Hematology Am Soc Hematol Educ Program. 2012;1:35–42. doi: 10.1182/asheducation-2012.1.35.
  38. Verhaak RGW, Goudswaard CS, van Putten W, et al. Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance. Blood. 2005;106(12):3747–54. doi: 10.1182/blood-2005-05-2168.
  39. Dvorakova D, Racil Z, Jeziskova I, et al. Monitoring of minimal residual disease in acute myeloid leukemia with frequent and rare patient-specific NPM1 mutations. Am J Hematol. 2010;85(12):926–9. doi: 10.1002/ajh.21879.
  40. Chou WC, Tang JL, Wu SJ, et al. Clinical implications of minimal residual disease monitoring by quantitative polymerase chain reaction in acute myeloid leukemia patients bearing nucleophosmin (NPM1) mutations. Leukemia. 2007;21(5):998–1004. doi: 10.1038/sj.leu.2404637.
  41. Meloni G, Mancini M, Gianfelici V, et al. Late relapse of acute myeloid leukemia with mutated NPM1 after eight years: evidence of NPM1 mutation stability. Haematologica. 2009;94(2):298–300. doi: 10.3324/haematol.2008.000059.
  42. Papadaki C, Dufour A, Seibl M, et al. Monitoring minimal residual disease in acute myeloid leukaemia with NPM1 mutations by quantitative PCR: clonal evolution is a limiting factor. Br J Haematol. 2009;144(4):517–23. doi: 10.1111/j.1365-2141.2008.07488.х.
  43. Kayser S, Walter RB, Stock W, Schlenk RF. Minimal residual disease in acute myeloid leukemia-current status and future perspectives. Curr Hematol Malig Rep. 2015;10(2):132–44. doi: 10.1007/s11899-015-0260-7.
  44. Terre C, Rousselot P, Dombret H, et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget. 2014;5(15):6280–8. doi: 10.18632/oncotarget.2196.
  45. Ommen HB, Schnittger S, Jovanovic JV, et al. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood. 2010;115(2):198–205. doi: 10.1182/blood-2009-04-212530.
  46. Гиршова Л.Л., Овсянникова Е.Г., Кузин С.О. и др. Молекулярный мониторинг уровня транскрипта RUNX1-RUNX1T1 при острых миелобластных лейкозах на фоне терапии. Клиническая онкогематология. 2016;9(4):456–64. doi: 10.21320/2500-2139-2016-9-4-456-464.[Girshova LL, Ovsyannikova EG, Kuzin SO, et al. Molecular Monitoring of RUNX1-RUNX1T1 Transcript Level in Acute Myeloblastic Leukemias on Treatment. Clinical oncohematology. 2016;9(4):456–64. doi: 10.21320/2500-2139-2016-9-4-456-464. (In Russ)]
  47. Balsat M, Renneville A, Thomas X, et al. Postinduction Minimal Residual Disease Predicts Outcome and Benefit From Allogeneic Stem Cell Transplantation in Acute Myeloid Leukemia With NPM1 Mutation: A Study by the Acute Leukemia French Association Group. J Clin Oncol. 2016;35(2):185–93. doi: 10.1200/JCO.2016.67.1875.
  48. Rossi G, et al. Comparison between multiparameter flow cytometry and WT1-RNA quantification in monitoring minimal residual disease in acute myeloid leukemia without specific molecular targets. Leuk Res. 2012;36(4):401–6. doi: 10.1016/j.leukres.2011.11.020.

Molecular Monitoring of RUNX1-RUNX1T1 Transcript Level in Acute Myeloblastic Leukemias on Treatment

LL Girshova, EG Ovsyannikova, SO Kuzin, EN Goryunova, RI Vabishchevich, AV Petrov, DV Motorin, DV Babenetskaya, VV Ivanov, KV Bogdanov, IV Kholopova, TS Nikulina, YuV Mirolyubova, YuA Alekseeva, AYu Zaritskii

VA Almazov Federal North-West Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

For correspondence: Ekaterina Gennad’evna Ovsyannikova, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel: +7(921)313-68-35; e-mail: katrin51297@mail.ru

For citation: Girshova LL, Ovsyannikova EG, Kuzin SO, et al. Molecular Monitoring of RUNX1-RUNX1T1 Transcript Level in Acute Myeloblastic Leukemias on Treatment. Clinical oncohematology. 2016;9(4):456–64 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-456-464


ABSTRACT

Background. The current approach to treatment of acute myeloblastic leukemia (AML) includes the achievement of maximum tumor reduction and, therefore, eradication of a leukemic clone. The goal of the therapy is to achieve undetectable levels of the target gene, except an isolated molecular rearrangement of RUNX1-RUNX1T1.

Aim. To estimate the dynamics of the RUNX1-RUNX1T1 level and relevant clinical manifestations during the monitoring of various stages of the program therapy and after its completion.

Methods. The article presents a description of 10 cases of AML with isolated RUNX1-RUNX1T1 expression (n = 4) and the expression in combination with different molecular and cytogenetic abnormalities (= 6). In addition, a long-term monitoring of the gene expression by quantitative determination of RUNX1-RUNX1T1 using a real-time PCR was presented.

Results. The incidence of relapses in a group with a decreased RUNX1-RUNX1T1 expression level of >2 log is 75 % as compared to patients with a less significant reduction of the transcript level (with the relapse incidence equal to 0 %) (= 0.05). The increase of the RUNX1-RUNX1T1 level against the background of bone marrow remission by more than 1 log coincided with a bone marrow relapse within 5–18 weeks. In addition, long-term persistence of a certain transcript level after the completion of a program therapy without relapse is possible.

Conclusion. The study analyzed possible molecular background of different clinical outcomes of long-term persistence of the RUNX1-RUNX1T1 transcript that might lead to an individualized approach to AML patients.


Keywords: acute myeloblastic leukemia, AML, RUNX1-RUNX1T1, molecular monitoring.

Received: April 5, 2016

Accepted: April 18, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Bitter MA, Le Beau MM, Rowley JD, et al. Association between morphology, karyotype, and clinical features in myeloid leukemias. Hum Pathol. 1987;18(3):211–25. doi: 10.1016/s0046-8177(87)80002-3.
  2. Mrozek K, Heinonen K, de la Chapelle A, Bloomfield CD. Clinical significance of cytogenetics in acute myeloid leukemia. Semin Oncol. 1997;24(1):17–31.
  3. Rowe D, Cotterill SJ, Ross FM, et al. Cytogenetically cryptic AML1-ETO and CBFbeta-MYH11 gene rearrangement: incidence in 412 cases of acute myeloid leukaemia. Br J Haematol. 2000;111(4):1051–6. doi: 10.1111/j.1365-2141.2000.02474.x.
  4. Downing JR. AML1/CBFbeta transcription complex: its role in normal hematopoiesis and leukemia. Leukemia. 2001;15(4):664–5. doi: 10.1038/sj.leu.2402035.
  5. Рулина А.В., Спирин П.В., Прасолов В.С. Активированные лейкозные онкогены AML1-ETO и C-KIT: роль в развитии острого миелоидного лейкоза и современные подходы к их ингибированию. Успехи биологической химии. 2010;50:349–86.
    [Rulina AV, Spirin PV, Prasolov VS. Activated leukemic AML1-ETO и C-KIT oncogenes: their role in the development of acute myeloid leukemia and modern approaches to their inhibition. Uspekhi biologicheskoi khimii. 2010;50:349–86. (In Russ)]
  6. Grimwade D, Walker H, Oliver F, et al. A on behalf of the Medical Research Council Adult and Children’s Leukaemia Working Parties. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. Blood. 1998;92(7):2322–33.
  7. Lowenberg B. Postremission treatment of acute myelogenous leukemia. N Eng J Med. 1995;332(4):260–2. doi: 10.1056/nejm199501263320411.
  8. Byrd JC, Dodge RK, Carroll A, et al. Patients with t(8;21) (q22;q22) and acute myeloid leukemia have superior failure-free and overall survival when repetitive cycles of high-dose cytarabine are administered. J Clin Oncol. 1999;17(12):3767–75.
  9. Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood. 2002;100(13):4325–36. doi: 10.1182/blood-2002-03-0772.
  10. Byrd JC, Ruppert AS, Mrozek K, et al. Repetitive cycles of high-dose cytarabine benefit patients with acute myeloid leukemia and inv(16) (p13q22) or t(16;16): results from CALGB 8461. J Clin Oncol. 2004;22(6):1087–94. doi: 10.1200/jco.2004.07.012.
  11. Schlenk RF, Benner A, Krauter J, et al. Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol. 2004;22(18):3741–50. doi: 10.1200/jco.2004.03.012.
  12. Marcucci G, Mrozek K, Ruppert AS, et al. Prognostic factors and outcome of core binding factor acute myeloid leukemia patients with t(8;21) differ from those of patients with inv(16): a Cancer and Leukemia Group B Study. J Clin Oncol. 2005;23(24):5705–17. doi: 10.1200/jco.2005.15.610.
  13. Yin JAL, O’Brien MA, Hills RK, et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 Trial. Blood. 2012;120(14):2826–35. doi: 10.1182/blood-2012-06-435669.
  14. Jourdan E, Boissel N, Chevret S, et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood. 2013;121(12):2213–23. doi: 10.1182/blood-2012-10-462879.
  15. Byrd JC, Weiss RB, Arthur DC, et al. Extramedullary leukemia adversely affects hematologic complete remission rate and overall survival in patients with t(8;21)(q22;q22): results from Cancer and Leukemia Group B 8461. J Clin Oncol. 1997;15(2):466–75.
  16. Nguyen S, Leblanc T, Fenaux P, et al. A white blood cell index as the main prognostic factor in t(8;21) acute myeloid leukemia (AML): a survey of 161 cases from the French AML Intergroup. Blood. 2002;99(10):3517–23. doi: 10.1182/blood.V99.10.3517.
  17. Baer MR, Stewart CC, Lawrence D, et al. Expression of the neural cell adhesion molecule CD56 is associated with short remission duration and survival in acute myeloid leukemia with t(8;21)(q22;q22). Blood. 1997;90(4):1643–8.
  18. Schoch C, Haase D, Haferlach T, et al. Fifty-one patients with acute myeloid leukemia and translocation t(8;21)(q22;q22): an additional deletion in 9q is an adverse prognostic factor. Leukemia. 1996;10(8):1288–95.
  19. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24(24):3904–11. doi: 10.1200/jco.2006.06.9500.
  20. Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia. 2006;20(6):965–70. doi: 10.1038/sj.leu.2404188.
  21. Hoyos M, Nomdedeu JF, Esteve J, et al. Core binding factor acute myeloid leukemia: the impact of age, leukocyte count, molecular findings, and minimal residual disease. Eur J Haematol. 2013;91(3):209–18. doi: 10.1111/ejh.12130.
  22. Демидова И.А. Использование молекулярно-биологических методов для определения генетических нарушений при миелоидных лейкозах и мониторирования минимальной остаточной болезни. Онкогематология. 2007;4:17–25.
    [Demidova IA. Application of molecular-biological methods for determining genetic disorders in myeloid leukemias and monitoring of minimal residual diseases. Onkogematologiya. 2007;4:17–25. (In Russ)]
  23. Estey EH. Acute myeloid leukemia: 2013 update on risk-stratification and management. Am J Hematol. 2013;88(4):318–27. doi: 10.1002/ajh.23404.
  24. Buccisano F, Maurillo L, Del Principe MI, et al. Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia. Blood. 2012;119(2):332–41. doi: 10.1182/blood-2011-08-363291.
  25. Tobal K, Newton J, Nige S, et al. Molecular quantitation of minimal residual disease in acute myeloid leukemia with с t(8;21) can identify patients in durable remission and predict clinical relapse. Blood. 2000;95(3):815–9.
  26. Yin JAL, O’Brien MA, Hills RK, et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 Trial. Blood. 2012;120(14):2826–30. doi: 10.1182/blood-2012-06-435669.
  27. Jourdan E, Boissel N, Chevret S, et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood. 2013;121(12):2213–23. doi: 10.1182/blood-2012-10-462879.
  28. Zhu H-H, Zhang X-H, Qin Y-Z, et al. MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial. Blood. 2013;121(2):4056–62. doi: 10.1182/blood-2012-11-468348.
  29. Morschhauser F, Cayuela JM, Martini S, et al. Evaluation of minimal residual disease using reverse-transcription polymerase chain reaction in t(8;21) acute myeloid leukemia: a multicenter study of 51 patients. J Clin Oncol. 2000;18(4):778–94.
  30. Willekens C, Blanchet O, Renneville A, et al. Prospective long-term minimal residual disease monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: results of the French CBF-2006 trial. Haematologica. 2016;101(3):328–35. doi: 10.3324/haematol.2015.131946.
  31. Schnittger S, Weisser M, Schoch C, et al. New score predicting for prognosis in PML-RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid leukemia based on quantification of fusion transcripts. Blood. 2003;102(8):2746–55. doi: 10.1182/blood-2003-03-0880.
  32. Ommen HB, Schnittger S, Jovanovic JV, et al. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood. 2010;115(2):198–205. doi: 10.1182/blood-2009-04-212530.
  33. Krauter J, Gorlich K, Ottmann O, et al. Prognostic value of minimal residual disease quantification by real-time reverse transcriptase polymerase chain reaction in patients with core binding factor leukemias. J Clin Oncol. 2003;21(23):4413–22. doi: 10.1200/jco.2003.03.166.
  34. Marcucci G, Livak KJ, Bi W, et al. Detection of minimal residual disease in patients with AML1/ETO-associated acute myeloid leukemia using a novel quantitative reverse ttranscriptase polymerase chain reaction assay. Leukemia. 1998;12(9):1482–9. doi: 10.1038/sj.leu.2401128.
  35. Lane S, Saal R, Molle P, et al. A ³ 1 log rise in RQ-PCR transcript levels defines molecular relapse in core binding factor acute myeloid leukemia and predicts subsequent morphologic relapse. Leuk Lymphoma. 2008;49(3):517–23. doi: 10.1080/10428190701817266.
  36. Perrea G, Lasa A, Aventi A, et al. Prognostic value of minimal residual disease in acute myeloid leukemia with favorable cytogenetics [t(8.21) and inv(16)]. Leukemia. 2006;20(1):87–94. doi: 10.1038/sj.leu.2404015.
  37. Jaso JM, Wang SA, Jorgensen JL, et al. Multicolor flow cytometric immunophenotyping for detection of minimal residual disease in AML: Past, present and future. Bone Marrow Transplant. 2014;49(9):1129–38. doi: 10.1038/bmt.2014.99.
  38. Bruggermann M, Raff T, Flohr T, et al. Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006;107(3):1116–23. doi: 10.1182/blood-2005-07-2708.
  39. Borowitz MJ, Devidas M, Hunger SP, et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship in other prognostic factors: a Children’s Oncology Group Study. Blood. 2008;111(12):5477–85. doi: 10.1182/blood-2008-01-132837.
  40. Muller MC, Cross NC, Erben P, et al. Harmonisation of molecular monitoring of CML therapy in Europe. Leukemia. 2009;23(11):1957–63. doi: 10.1038/leu.2009.168.
  41. Jurlander J, Caligiuri MA, Ruutu T, et al. Persistence of the AMLI/ETO Fusion Transcript in Patients Treated With Allogeneic Bone Marrow Transplantation for t(8;21) Leukemia. Blood. 1996;88(6):2183–219.
  42. Kayser S, Schlenk RF, Grimwade D, et al. Minimal residual disease–directed therapy in acute myeloid leukemia. Blood. 2015;125(15):2331–5. doi: 10.1182/blood-2014-11-578815.
  43. Evans PA, Short MA, Jack AS, et al. Detection and quantitation of the transcripts associated with the inv(16) in presentation and follow-up samples from patients with AML. Leukemia. 1997;11(3):364–9. doi: 10.1038/sj.leu.2400578.
  44. Laczika K, Novak M, Hilgarth B, et al. Competitive CBFbeta/MYH11 reverse transcriptase polymerase chain reaction for quantitative assessment of minimal residual disease during post remission therapy in acute myeloid leukemia with inversion 16: a pilot study. J Clin Oncol. 1998;16(4):1519–25.
  45. Krauter J, Hoellge W, Wattjies MP, et al. Detection and quantitation of CBFB/MYH11 fusion transcript in patients with inv(16) positive acute myeloblastic leukemia by real-time RT-PCR. Genes Chromos Cancer. 2001;30(4):342–8. doi: 10.1002/gcc.1100.
  46. Marcucci G, Caligiuri MA, Dohner H, et al. Quantification of CBFbeta/MYH11 fusion trancript byreal-time RT-PCR in patients with inv(16) acute myeloid leukemia. Leukemia. 2001;15(7):1072–80. doi: 10.1038/sj.leu.2402159.
  47. Duployez N, Willekens C, Marceau-Renaut A, et al. Prognosis and monitoring of core-binding factor acute myeloid leukemia: current and emerging factors. Exp Rev Hematol. 2014;8(1):43–56. doi: 10.1586/17474086.2014.976551.
  48. Shima T, Miyamoto T, Kikushige Y, et al. The ordered acquisition of Class II and Class I mutations directs formation of human t(8;21) acute myelogenous leukemia stem cell. Exp Hematol. 2014;42(11):955–65. doi: 10.1016/j.exphem.2014.07.267.
  49. Buonamici S, Ottaviani E, Visani G, et al. Patterns of AML-ETO1 transcript expression in patients with acute myeloid leukemia and t(8;21) in complete hematologic remission. Haematologica. 2004;89(1):103–5.
  50. Song J, Mercer D, Hu X, et al. Common Leukemia- and Lymphoma-Associated Genetic Aberrations in Healthy Individuals. J Mol Diagn. 2011;13(2);213–9. doi: 10.1016/j.jmoldx.2010.10.009.
  51. Miyamoto T, Weissman IL, Akashi K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Nat Acad Sci. 2000;97(13):7521–6. doi: 10.1073/pnas.97.13.7521.
  52. Yin JAL, Tobal K. Detection of minimal residual disease in acute myeloid leukemias: methodologies, clinical and biological significance. Br J Haematol. 1999;106(3):578–90. doi: 10.1046/j.1365-2141.1999.01522.x.
  53. Corces-Zimmerman MR, Hong W-J, Weissman IL, et al. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Nat Acad Sci. 2014;111(7):2548–53. doi: 10.1073/pnas.1324297111.
  54. Russler-Germain DA, Spencer DH, Young MA, et al. The R882H DNMT3A Mutation Associated with AML Dominantly Inhibits Wild-Type DNMT3A by Blocking Its Ability to Form Active Tetramers. Cancer Cell. 2014;25(4):442–54. doi: 10.1016/j.ccr.2014.02.010.
  55. Ommen HB, Schnittger S, Jovanovic JV, et al. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood. 2010;115(2):198–205. doi: 10.1182/blood-2009-04-212530.
  56. Wang Y, Wu D-P, Liu Q-F, et al. In adults with t(8;21) AML, posttransplant RUNX1/RUNX1T1-based MRD monitoring, rather than c-KIT mutations, allows further risk stratification. Blood. 2014;124(12):1880–6. doi: 10.1182/blood-2014-03-563403.

 

Allogeneic Hematopoietic Stem Cell Transplantation in Myelodysplastic Syndromes and Clinical Significance of WT1 Gene Overexpression

N.N. Mamaev1, A.V. Gorbunova1, T.L. Gindina1, E.V. Morozova1, Ya.V. Gudozhnikova1, O.A. Slesarchuk1, V.N. Ovechkina1, A.A. Rats1, E.G. Boichenko2, E.A. Ukrainchenko3, V.M. Kravtsova1, A.V. Evdokimov1, I.M. Barkhatov1, S.N. Bondarenko1, B.V. Afanasev1

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

2 Municipal Children’s Hospital No. 1, 14 Avangardnaya str., Saint Petersburg, Russian Federation, 198205

3 Alexandrovskaya Municipal Hospital No. 17, 4 pr-t Solidarnosti, Saint Petersburg, Russian Federation, 193312

For correspondence: N.N. Mamaev, DSci, Professor, 12 Rentgena str., Saint Petersburg, Russian Federation, 197022; Tel: +7(812)233-12-43; e-mail: nikmamaev524@gmail.com

For citation: Mamaev N.N., Gorbunova A.V., Gindina T.L., Morozova E.V., Gudozhnikova Ya.V., Slesarchuk O.A., Ovechkina V.N., Rats A.A., Boichenko E.G., Ukrainchenko E.A., Kravtsova V.M., Evdokimov A.V., Barkhatov I.M., Bondarenko S.N., Afanas’ev B.V. Allogeneic Hematopoietic Stem Cell Transplantation in Myelodysplastic Syndromes and Clinical Significance of WT1 Gene Overexpression. Klin. Onkogematol. 2014; 7(4): 551–563 (In Russ.).


ABSTRACT

The results of allogeneic hematopoietic stem cell transplantation (HSCT) in 17 patients (pts, 11 male, 6 female) with myelodysplastic syndromes (3 RA/RARS/RCMD, 5 RAEB-1, 7 RAEB-2, 2 JMML) are presented. The median age was 26 years with a range between 1 and 55 years. Serial cytogenetic investigations were carried out in all of them. Seven pts demonstrated monosomy 7, which was associated with other chromosome abnormalities in 4 cases. In addition, deletion at 11q23 (n = 3), trisomy 8 (n = 2) and 21 (n = 2), involvement into rearrangement at 3q (n = 2), t(6;9) translocation, and others more rare abnormalities were found. Prior to aHSCT, 11 of 7 received hypomethylating agents (HA) which proved to be effective in a half of them. In order to prepare for aHSCT, ablative (busulfan, cyclophosphamide) or non-ablative (fludarabine, cyclophosphamide) conditioning regimes were applied (4 and 13 respectively). Repeated aHSCT was carried out in 6 pts because of transplant rejection or post-transplant relapses. Molecular monitoring of minimal residual disease as well as early diagnosis of these relapses was performed by means of serial tests of the WT1 gene level expression and donor chimerism. Maximum WT1 values varied between 15 and 43133 copies/104 copies of ABL gene; and molecular relapses were registered in a half of them, including 5 patients with transformation into acute leukemia (AL). HA were used for prevention and treatment of relapses in 4 (24 %) patients; and HA were combined with donor lymphocyte infusions. Standard chemotherapy was applied for these purposes relatively rarely. This study demonstrated WT1 gene overexpression to be not only an important marker for diagnosis of post-transplant MDS/AL relapses, but it also can be used for evaluation of the treatment efficacy.


Keywords: myelodysplastic syndromes, allogeneic HSCT, post-transplant relapses, minimal residual disease, molecular monitoring, serial WT1 gene expression.

Accepted: September 30, 2014

Read in PDF (RUS) pdficon


REFERENCES 

  1. Barrett A.J., Battiwala M. Relapse after allogeneic stem cell transplantation. Exp. Rev. Hematol. 2012; 3(4): 429–41.
  2. Tamura K., Kanazawa T., Suzuki M. et al. Successful rapid discontinuation of immunosuppressive therapy at molecular relapse after allogeneic bone marrow transplantation in a pediatric patient with myelodysplastic syndrome. Am. J. Hematol. 2006; 81: 139–41.
  3. Wertheim G.B., Bagg A. Minimal residual disease testing to predict relapse following transplant for AML and high-grade myelodysplastic syndromes. Exp. Rev. Mol. Diagn. 2011; 11(4): 361–6.
  4. Brieger J., Weidmann E., Fenchel K. et al. The expression of the Wilms’ tumor gene in acute myelocytic leukemias as a possible marker for leukemic blast cells. Leukemia. 1994; 8: 2138–43.
  5. Inoue K., Sugiyama H., Ogawa 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: 3071–9.
  6. Inoue K., Ogawa H., Yamagami T. et al. Long-term follow-up of minimal residual disease in leukemia patients by monitoring WT1 (Wilms tumor gene) expression levels. Blood. 1996; 88: 2267–78.
  7. Tamaki H., Ogawa H., Inoue K. et al. Increased expression of the Wilms tumor gene (WT1) at relapse in acute leukemia. Blood. 1996; 88: 4396–8.
  8. Patmasirivat P., Fraizer G., Kantarjian H. et al. WT1 and GATA1 expression in myelodysplastic syndrome and acute leukemia. Leukemia. 1999; 13: 891–900.
  9. Ogawa H., Ikegame K., Kawakami M., Tamaki H. WT1 gene transcript assay for relapse in acute leukemia after transplantation. Leuk. Lymphoma. 2004; 45: 1747–53.
  10. 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: 2115–21.
  11. 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: 921–4.
  12. Candoni A., Tribelli M., Cilloni D. et al. Quantitative assessment of WT1 gene expression after allogeneic stem cell transplantation is a useful tool for monitoring minimal residual disease in acute myeloid leukemia. Eur. J. Haematol. 2009; 82: 61–8.
  13. Miyawaki S., Hatsumi N., Tamaki T. et al. Prognostic potential of detection of WT1 mRNA level in peripheral blood in adult acute myeloid leukemia. Leuk. Lymphoma 2010; 51: 1855–61.
  14. 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. 2012; 47: 499–507.
  15. Nomdedeu J.F., Hoyos M., Carricondo M. et al. Bone marrow WT1 levels at diagnosis, post-induction and post-intensification in adult de novo AML. Leukemia. 2013; 27: 2157–64.
  16. 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: 503–9.
  17. Frairia Ch., Aydin S., Riera L. et al. WT1 expression in acute myeloid leukemia: a useful marker for improving therapy response evaluation. Blood. 2013; 123(21): 2588.
  18. Tamaki H., Ogawa H., Ohyashiki K. et al. The Wilm’s tumor gene is a good marker for diagnosis of disease progression of myelodysplastic syndromes. Leukemia. 1999; 13: 393–9.
  19. Patmasiriwat P., Fraizer G., Kantarjian H. et al. WT1 and GATA1 expression in myelodysplastic syndrome and acute leukemia. Leukemia. 1999; 13: 891–900.
  20. Cilloni D., Gottardi E., Messa F. et al. Significant correlation between the degree of WT1 expression and the international prognostic scoring system score in patients with myelodysplastic syndromes. J. Clin. Oncol. 2003; 21: 1988–95.
  21. Cilloni D., Saglio G. WT1 as a universal marker for minimal residual disease detection and quantification in myeloid leukemias and in myelodysplastic syndrome. Acta Haematologica. 2004; 112: 79–84.
  22. Абдулкадыров К.М., Грицаев С.В., Капустин С.И. и др. Экспрессия гена опухоли Вилмса (WT1) в клетках крови больных миелодиcпластическим синдромом. Вопросы онкологии 2004; 50(6): 668–71. [Abdulkadyrov K.M., Gritsaev S.V., Kapustin S.I. et al. Wilms tumor gene (WT1) expression in blood cells of patients with myelodysplastic syndrome. Voprosy Onkologii. 2004; 50(6): 668–71. (In Russ.)]
  23. Bader P., Niemeyer C., Weber G. et al. WT1 gene expression: useful marker for minimal residual disease in childhood myelodysplastic syndromes and juvenile myelomonocytic leukemia? Eur. J. Haematol. 2004; 73: 25–8.
  24. Iwasaki T., Sugisaki C., Nagata K. et al. Wilms’ tumor 1 message and protein expression in bone marrow failure syndrome and acute leukemia. Pathol. Int. 2007; 57: 645–51.
  25. Qin Y.-Z., Zhu H.-H., Liu Y.-R. et al. PRAME and WT1 transcripts constitute a good molecular marker combination for monitoring minimal residual disease in myelodysplastic syndromes. Leuk. Lymphoma. 2012; DOI: 10.3109/10428194.2012.743656.
  26. Ueda Y., Mizutani C., Nannya Y. et al. Clinical evaluation of WT1 mRNA expression levels in peripheral blood and bone marrow in patients with myelodysplastic syndromes. Leuk. Lymphoma. 2013; 54(7): 1450–8.
  27. Lange T., Hubmann M., Burkhardt R. et al. Monitoring of WT1 expression in PB and CD34+ donor chimerism of BM predicts early relapse in AML and MDS patients after hematopoietic cell transplantation with reduced-intensity conditioning. Leukemia. 2011; 25: 498–505.
  28. Maurer U., Brieger J., Weidmann E. et al. The Wilms’ tumor gene is expressed in a subset of CD34+ progenitors and downregulated early in the course of differentiation in vitro. Exp. Hematol. 1997; 25: 945–50.
  29. Kwon M., Marti nez-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: 1235–42.
  30. Jacobsohn D.A., Tse W.T., Chaleff S. et al. High WT1 gene expression before haematopoietic stem cell transplant in children with acute myeloid leukaemia predicts poor event-free survival. Br. J. Haematol. 2009; 146: 669–74.
  31. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миело- диспластические синдромы с высокой экспрессией гена EVI1: теоретиче- ские и клинические аспекты. Клин. онкогематол. 2012; 5(4): 361–4. [Mamaev N.N., Gorbunova A.V., Gindina T.L. et al. Leukemias and myelodysplastic syndromes with high EVI1 gene expression: theoretical and clinical aspects. Klin. Onkogematol. 2012; 5(4): 361–4. (In Russ.)]
  32. Alonso-Dominguez J.M., 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 Gen. 2012; 205: 190–1.
  33. Gerds A.T., Deeg H.J. Transplantation for myelodysplastic syndrome in the era of hypomethylating agents. Curr. Opin. Hematol. 2012; 19: 71–5.
  34. Nishihori T., Perkins J., Mishra A. et al. Pretransplantation 5-Azacitidine in high-risk myelodysplastic syndrome. Biol. Blood Marrow Transplant. 2014; 20: 776–80.
  35. Raza A., Gezer S., Mundle S. et al. Apoptosis in bone marrow biopsy samples involving stromal and hematopoietic cells in 50 patients with myelodysplastic syndromes. Blood. 1995; 86(1): 268–76.

Allogeneic Hematopoietic Stem Cell Transplantation for ALL Patients with t(12;21)(p13;q22) Translocation

N.N. Mamaev1, E.V. Semenova1, N.V. Stancheva1, V.A. Katerina1, I.M. Barkhatov1, A.V. Evdokimov1, E.G. Boychenko2, T.L. Gindina1, V.M. Kravtsova1, L.S. Zubarovskaya1, B.V. Afanasyev1

1 R.M. Gorbacheva Memorial Institute of Children Oncology, Hematology and Transplantation under I.P. Pavlov State Medical University, Saint Petersburg, Russian Federation

2 1st Municipal Children’s Hospital, Saint Petersburg, Russian Federation

For citation: Mamaev N.N., Semenova E.V., Stancheva N.V., Katerina V.A., Barkhatov I.M., Evdokimov A.V., Boichenko E.G., Gindina T.L., Kravtsova V.M., Zubarovskaya L.S., Afanas’ev B.V. Allogeneic Hematopoietic Stem Cell Transplantation for ALL Patients with t(12;21)(p13;q22) Translocation. Klin. onkogematol. 2014; 7(3): 327–34 (In Russ.).


ABSTRACT

The results of allogeneic hematopoietic stem cell transplantation (alloHSCT) in 10 pediatric patients (4 boys, 6 girls at the age of 4 to 17 years, mean age is 9.8 years) with relapses of acute lymphoblastic leukemia (ALL) with TEL-AMLI fusion gene are presented. The first remission duration ranged from 20 to 70 months (mean duration 39.9 months). Transplantation was performed in 6 patients during the second (and further) remission, whereas 4 patients underwent transplantation during the relapse. Six patients received a graft from matched related (n = 3) or unrelated (n = 3) donors, haploidentical HSCT was performed in four other patients because there was no donor. Conditioning regimens were myeloablative in 8 cases and RIC (Reduced Intensity Conditioning) in 2 cases. Successful engrafting took place in 9 (90 %) of 10 patients. Additional haploidentical transplantation was performed in 1 case, when the transplant was rejected.

Monitoring of treatment was performed by means of serial testing of TEL-AMLI fusion gene expression level, of serial donor chimerism and the blast cell count in bone marrow and peripheral blood. The study demonstrated that four patients younger than 4 years had TEL-AML1 gene expression at all stages of their disease, including pre- and post-transplantation period. Due to it, even the donor chimerism and blast cell count in bone marrow and/or peripheral blood changed. On the contrary, in 3 more patients, TEL-AML1 gene expression levels were low before alloHSCT, being absent after HSCT.

In general, seven patients have been monitored for 178–2627 days (at the average of 870 days) including two patients with post-transplant relapses. At the same time, 3 patients died on days 20–263 after transplantation.

The observed difference in response to chemotherapy might be supposedly explained by involvement of not only hematopoietic, but also mesenchymal cells into the leukemic process (this fact was demonstrated for this group of patients by S. Shalapour et al., 2010). But this fact should be confirmed in further studies.

Keywords: ALL, t(12;21)(p13;q22) translocation, alloHSCT, molecular monitoring, difference in response to treatment.

Address correspondence to: nikmamaev524@gmail.com

Accepted: May 22, 2014

Read in PDF (RUS) pdficon


REFERENCES

  1. Romana S.P., Mauchauffe M., Le Coniat M. et al. The t(12;21) of acute lymphoblastic leukemia results in a tel-AML1 gene fusion. Blood 1995; 85: 3662–70.
  2. Shurtleff S.A., Buijs A., Behm F.G. et al. TEL/AML1 fusion resulting from a cryptic t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup of patients with an excellent prognosis. Leukemia 1995; 9: 1985–9.
  3. McLeen T.W., Ringold S., Neuberg D. et al. TEL/AML-1 dimerizes and is associated with a favorable outcome in childhood acute lymphoblastic leukemia. Blood 1996; 88: 4252–8.
  4. Loh M.L., Rubnitz J.E. TEL/AML1-positive pediatric leukemia: prognostic significance and therapeutic approaches. Curr. Opin. Hematol. 2002; 9: 345–52.
  5. Pui C.H., Campana D., Evans W.E. Childhood acute lymphoblastic leukemia – current status and future perspectives. Lancet Oncol. 2001; 2: 597–607.
  6. Loh M.L., Goldwasser M.A., Silverman L.B. et al. Prospective analysis of TEL/AML1-positive patients treated on Dana-Faber Cancer Institute Consortium Protocol 95-01. Blood 2006; 107: 4508–13.
  7. Burmeister T., Gokbuget N., Schwartz S. et al. Clinical features and prognostic implications of TCF3-PBX1 and ETV6-RUNX1 in adult acute lymphoblastic leukemia. Haematologica 2010; 95: 241–6.
  8. Pui C.H., Pei D., Campana D. et al. Improved prognosis for older adolescents with acute lymphoblastic leukemia. J. Clin. Oncol. 2011; 29(4): 386–91.
  9. Seeger K., Adams H.P., Buchwald D. et al. TEL-AML 1 fusion transcript in relapsed childhood acute lymphoblastic leukemia: the Berlin-Frankfurt-Munster Study Group. Blood 1998; 91: 1716–22.
  10. Seeger K., Stackelberg A.V., Taube T. et al. Relapse of TEL-AML1- positive acute lymphoblastic leukemia in childhood: A matched-pair analysis. J. Clin. Oncol. 2001; 19: 3188–93.
  11. Shalapour S., Eckert C., Seeger K. et al. Leukemia-associated genetic aberrations in mesenchymal stem cells of children with acute lymphoblastic leukemia. J. Mol. Med. 2010; 88: 249–65.