Outcomes of Allogeneic Hematopoietic Stem Cell Transplantation in Myelodysplastic Syndromes with Trisomy 8 and/or Monosomy 7

MV Latypova, NN Mamaev, TL Gindina, AI Shakirova, OV Paina, AA Osipova, TV Rudakova, EV Morozova, SN Bondarenko, LS Zubarovskaya

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

For correspondence: Prof. Nikolai Nikolaevich Mamaev, MD, PhD, 6/8 L’va Tolstogo ul., Saint Petersburg, Russian Federation, 197022; e-mail: nikmamaev524@gmail.com

For citation: Latypova MV, Mamaev NN, Gindina TL, et al. Outcomes of Allogeneic Hematopoietic Stem Cell Transplantation in Myelodysplastic Syndromes with Trisomy 8 and/or Monosomy 7. Clinical oncohematology. 2022;15(2):198–204. (In Russ).

DOI: 10.21320/2500-2139-2022-15-2-198-204


ABSTRACT

The study assessed the outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in 34 patients with cytogenetically verified variants of myelodysplastic syndrome (MDS) with trisomy 8 and/or monosomy 7, who were treated at the RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation from 2013 to 2020. Both adult and pediatric MDS were analyzed without excluding the variants with two additional chromosomal abnormalities or complex karyotype. The study revealed that а) allo-HSCT should be performed in the treatment of both MDS variants; b) the outcomes of trisomy 8 treatment appeared to be better; c) children with monosomy 7 showed a higher rate of toxic complications in allo-HSCT.

Keywords: myelodysplastic syndromes, cytogenetic variants, trisomy 8, monosomy 7, allo-HSCT.

Received: October 2, 2021

Accepted: March 6, 2022

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Ades L, Itzykson R, Fenaux P. Myelodysplastic syndromes. Lancet. 2014;383(9939):2239–52. doi: 10.1016/S0140-6736(13)61901-7.
  2. Robin M, Porcher R, Zinke-Cerwenka W, et al. Allogeneic hematopoietic stem cell transplant in patients with lower risk myelodysplastic syndrome: a retrospective analysis on behalf of the Chronic Malignancy Working Party of the EBMT. Bone Marrow Transplant. 2017;52(2):209–15. doi: 10.1038/bmt.2016.266.
  3. Deeg HJ, Scott BL, Fang M, et al. Five-group cytogenetic risk classification, monosomal karyotype, and outcome after hematopoietic cell transplantation for MDS or acute leukemia evolving from MDS. Blood. 2012;120(7):1395–409. doi: 10.1182/blood-2012-04-423046.
  4. Malcovati L, Hellstrom-Lindberg E, Bowen D, et al. Diagnosis and treatment of primary myelodysplastic syndromes in adults: recommendations from the European LeukemiaNet. Blood. 2013;122(17):1943–64. doi: 1182/blood-2013-03-492884.
  5. Schanz J, Tuchler H, Sole F, et al. New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from international database merge. J Clin Oncol. 2012;50(8):820–9. doi: 10.1200/JCO.2011.35.6394.
  6. Nevill TJ, Shepherd JD, Sutherland HJ, et al. IPSS poor-risk karyotype as a predictor of outcome for patients with myelodysplastic syndrome following myeloablative stem cell transplantation. Biol Blood Marrow Transplant. 2009;15(2):205–13. doi: 10.1016/j.bbmt.2008.11.015.
  7. Koenecke C, Gohring G, de Wreede LC, et al. Impact of the revised International Prognostic Scoring System, cytogenetics and monosomal karyotype on outcome after allogeneic stem cell transplantation for myelodysplastic syndromes and secondary acute myeloid leukemia evolving from myelodysplastic syndromes: a retrospective multicenter study of the European Society of Blood and Marrow Transplantation. Haematologica. 2015;100(3):400–8. doi: 10.3324/haematol.2014.116715.
  8. Armand P, Kim HT, DeAngelo DJ, et al. Impact of cytogenetics on outcome of de novo and therapy- related AML and MDS after allogeneic transplantation. Biol Blood Marrow Transplant. 2007;13(6):655–64. doi: 10.1016/j.bbmt.2007.01.079
  9. Konuma T, Miyazaki Y, Ohashi K, et al. Outcomes of allogeneic hematopoietic stem cell transplantation in adults patients with myelodysplastic syndrome harboring trisomy 8. Biol Blood Marrow Transplant. 2017;23(1):75–80. doi: 10.1016/j.bbmt.2016.10.015.
  10. Trobaugh-Lotrario AD, Kletzel M, Quinones RR, et al. Monosomy 7 associated with pediatric acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS): Successful management by allogeneic hematopoietic stem cell transplant (HSCT). Bone Marrow Transplant. 2005;25(2):143–9. doi: 10.1038/sj.bmt.1704753.
  11. Al-Anazi KA. Myelodysplastic disorders, monosomy 7. In: Fuchs O, ed. Myelodysplastic syndromes. IntechOpen; 2016. рр. 131–61. doi: 10.5772/64549.
  12. Itonaga Y, Ishiyama K, Aoki K, et al. Clinical Impact of the loss of chromosome 7q on outcomes of patients with myelodysplastic syndromes treated with allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2019;54(9):1471–81. doi: 10.1038/s41409-019-0469-5.
  13. Komrokji RS, Padron E, Ebert BL, et al. Deletion 5q MDS: Molecular and therapeutic implications. Best Pract Res Clin Haematol. 2013;26(4):365–75. doi: 1016/j.beha.2013.10.013.
  14. Gindina TL, Mamaev NN, Afanasyev BV. Chromosome Abnormalities and Hematopoietic Stem Cell Transplantation in Acute Leukemias. In: Larramendy ML, Soloneski S, eds. Chromosomal Abnormalities – A hallmark manifestation of genomic instability. IntechOpen; 2017. рр. 71–86. doi: 10.5772/67802.
  15. Schaffer L, McGovan-Jordan L, Schmid M. An International System for Human Cytogenetic Nomenclature. Basel: S. Karger; 2013. 140 p.
  16. Гиндина Т.Л. Характеристика основных цитогенетических изменений у больных острыми лейкозами и их связь с результатами аллогенной трансплантации стволовых клеток: Дис.… д-ра мед. наук. СПб., 2019. 373 с.
    [Gindina TL. Kharakteristika osnovnykh tsitogeneticheskikh izmenenii u bol’nykh ostrymi leikozami i ikh svyaz’ s rezul’tatami allogennoi transplantatsii stvolovykh kletok. (The characteristics of major cytogenetic changes in acute leukemia patients and their association with the outcomes of allogeneic stem cell transplantation.) [dissertation] Saint Petersburg; 2019. 373 p. (In Russ)]
  17. Bersanelli M, Travaglino E, Meggendorfer M, et al. Classification and Personalized Prognostic Assessment on the Basis of Clinical and Genomic Features in Myelodysplastic Syndromes. J Clin Oncol. 2021;39(11):1223–33. doi: 10.1200/JCO.20.01659.
  18. Мамаев Н.Н., Латыпова М.В., Шакирова А.И. и др. Роль BAALC-экспрессирующих лейкозных клеток-предшественниц в патогенезе миелодиспластических синдромов. Клиническая онкогематология. 2022;15(1):62–8. doi: 10.21320/2500-2139-2022-15-1-62-68.
    [Mamaev NN, Latypova MV, Shakirova AI, et al. The Role of BAALC-Expressing Leukemia Precursor Cells in the Pathogenesis of Myelodysplastic Syndromes. Clinical oncohematology. 2022;15(1):62–8. doi: 10.21320/2500-2139-2022-15-1-62-68. (In Russ)]

The Role of BAALC-Expressing Leukemia Precursor Cells in the Pathogenesis of Myelodysplastic Syndromes

NN Mamaev, MV Latypova, AI Shakirova, TL Gindina, MM Kanunnikov, NYu Tsvetkov, IM Barkhatov, SN Bondarenko, MD Vladovskaya, EV Morozova

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; e-mail: nikmamaev524@gmail.com

For citation: Mamaev NN, Latypova MV, Shakirova AI, et al. The Role of BAALC-Expressing Leukemia Precursor Cells in the Pathogenesis of Myelodysplastic Syndromes. Clinical oncohematology. 2022;15(1):62–8. (In Russ).

DOI: 10.21320/2500-2139-2022-15-1-62-68


ABSTRACT

The present paper provides evidence for a high detection rate of BAALC gene overexpression, also combined with WT1 gene overexpression, in patients with myelodysplastic syndromes (MDS) and FISH-verified chromosome defects. The BAALC and WT1 gene expression profiling in 16 MDS patients (6 out of them received allogeneic hematopoietic stem cell transplantation) showed an increased BAALC expression in 14 patients. The expression level in 2 patients was near the cut-off. Low expression levels were identified in a female patient with isolated 5q deletion in karyotype and also with its combination with complex karyotype. On the other hand, the highest expression levels were reported in patients with normal karyotype and 3q26 locus rearrangement, which was associated with EVI1 gene overexpression. Since the BAALC expression level, at least in patients with the major (except for М3 and М7) FAB-variants of acute myeloid leukemias (AML), was closely associated with BAALC-producing precursor cells of leukemia clone, a profound study of this phenomenon in MDS patients seems to be important for understanding the finest mechanisms underlying the pathogenesis of AML and AML relapses on the level of precursor cells.

Keywords: myelodysplastic syndromes, BAALC and WT1 genes, overexpression, post-transplantation relapses, BAALC-producing precursor cells, pathogenesis, prognosis.

Received: July 7, 2021

Accepted: November 4, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Gadji M, Pozzo AR. From cellular morphology to molecular and epigenetic anomalies of myelodysplastic syndromes. Genes Chromos Cancer. 2019;58(7):474–83. doi: 10.1002/gcc.22689.
  2. Schanz J, Cevik N, Fonatsch C, et al. Detailed analysis of clonal evolution and cytogenetic evolution patterns in patients with myelodysplastic syndromes (MDS) and related myeloid disorders. Blood Cancer J. 2018;8(3):28. doi: 10.1038/s41408-018-0061-z.
  3. Bersanelli M, Travaglino E, Meggendorfer M, et al. Classification and Personalized Prognostic Assessment on the Basis of Clinical and Genomic Features in Myelodysplastic Syndromes. J Clin Oncol. 2021;39(11):1223–33. doi: 10.1200/JCO.20.01659.
  4. Papaemmanuil E, Gerstung M, Malcovati L, et al. Сlinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122(22):3616–27. doi: 10.1182/blood-2013-08-518886.
  5. Minetto P, Guolo F, Cavio M, et al. Combined assessment of WT1 and BAALC gene expression at diagnosis may improve leukemia-free survival prediction in patients with myelodysplastic syndromes. Leuk Res. 2015;39(8):866–73. doi: 10.1016/leukres.2015/04/011.
  6. Мамаев Н.Н., Шакирова А.И., Бархатов И.М. и др. Ведущая роль BAALC-экспрессирующих клеток-предшественниц в возникновении и развитии посттрансплантационных рецидивов у больных острыми миелоидными лейкозами. Клиническая онкогематология. 2020;13(1):75–88. doi: 10.21320/2500-2139-2020-13-1-75-88.
    [Mamaev NN, Shakirova AI, Barkhatov IM, et al. Crucial Role of BAALCExpressing Progenitor Cells in Emergence and Development of Post-Transplantation Relapses in Patients with Acute Myeloid Leukemia. Clinical oncohematology. 2020;13(1):75–88. doi: 10.21320/2500-2139-2020-13-1-75-88. (In Russ)]
  7. Mamaev NN, Shakirova AI, Barkhatov IM, et al. Crucial role of BAALC-expressing leukemic precursors in origin and development of posttransplant relapses in patients with acute myeloid leukemias. Int J Hematol. 2020;8(6):127–31. doi: 10.15406/htij.2020.08.00240.
  8. Mamaev NN, Shakirova AI, Barkhatov IM, et al. New opportunities for assay of leukemia initiating cells (LICs) participating in post-transplant relapse development in the patients with acute myeloid leukemia. 3rd Annual IACH Meeting, 1–3 October, 2020, Paris. Report #12.
  9. Mamaev NN, Shakirova AI, Gindina TL, et al. Quantitative study of BAALC- and WT1-expressing cell precursors in the patients with different cytogenetic and molecular AML variants treated with Gemtuzumab ozogamicin and hematopoietic stem cell transplantation. Cell Ther Transplant. 2021;10(1):55–62. doi: 10.18620/ctt-1866-8836-2021-10-1-55-62.
  10. Thol F, Kade S, Schlarmann C, et al. Frequency and prognostic impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with myelodysplastic syndromes. Blood. 2012;119(15):3578–84. doi: 10.1182/blood-2011-12-399337.
  11. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodyspl astic sendromes. Leukemia. 2014;18(2):241–7. doi: 10.1038/leu.2013.336.
  12. Pauebelle E, Piesa A, Hayette S, et al. Efficacy of all-trans-retinoic acid in high risk acute myeloid leukemia with overexpression of EVI1. Oncol Ther. 2019;7(2):121–30. doi: 10.1007/s40487-019-0095-9.
  13. Field T, Perkins KJ, Huang Y, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  14. Мамаев Н.Н., Шакирова А.И., Морозова Е.В., Гиндина Т.Л. EVI1-позитивные лейкозы и миелодиспластические синдромы: теоретические и клинические аспекты (обзор литературы). Клиническая онкогематология. 2021;14(1):103–17. doi: 10.21320/2500-2139-2021-14-1-103-117.
    [Mamaev NN, Shakirova AI, Morozova EV, Gindina TL. EVI1-Positive Leukemias and Myelodysplastic Syndromes: Theoretical and Clinical Aspects (Literature Review). Clinical oncohematology. 2021;14(1):103–17. doi: 10.21320/2500-2139-2021-14-1-103-117. (In Russ)]
  15. Geoffroy М-С, Esnault C, de The H. Retinoids in hematology: a timely revival? Blood. 2021;137(18):2429–37. doi: 10.1182/blood.2020010100.

Hypomethylating Agents in Oncohematology

AD Shirin, OYu Baranova

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

For correspondence: Anton Dmitrievich Shirin, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-28-24; e-mail: shirin-anton@mail.ru

For citation: Shirin AD, Baranova OYu. Hypomethylating Agents in Oncohematology. Clinical oncohematology. 2016;9(4):369–82 (In Russ).

DOI: 10.21320/2500-2139-2016-9-4-369-382


ABSTRACT

The review describes epigenetic processes, including methylation of nuclear and mitochondrial DNA, as well as RNA. It dwells on mechanisms of demethylation and corresponding medicinal products. It presents detailed information on results of numerous large randomized studies intended to evaluate hypomethylating agents (azanucleosides). Special attention is paid to outcomes of azanucleoside therapy in patients with acute myeloid leukemias. The article describes several prognostic systems and treatment algorithms for myelodysplastic syndromes. Two azanucleosides have been approved in Russia to date: azacitidine (for SQ administration) and decitabine (for IV administration). International authors analyze the experience in oral and subcutaneous administration of decitabine. However, the problem of off-label use of hypomethylating agents is still open. The review gives a brief description of ongoing clinical trials with azanucleosides.


Keywords: epigenetics, acute myeloid leukemias, myelodysplastic syndromes, azacitidine, decitabine, hypomethylating agents, azanucleosides.

Received: May 10, 2016

Accepted: May 20, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Уоддингтон К.Х. Основные биологические концепции. В кн.: На пути к теоретической биологии. Часть I. Пролегомены. М.: Мир, 1970. С. 11–38.
    [Waddington CH. Basic Ideas of Biology. In: Waddington CH, ed. Towards a Theoretical Biology. Vol. 1. Edinburgh: Edinburgh University Press. 1968–72. (Russ. ed.: Waddington CH. Osnovnye biologicheskie kontseptsii. In: Waddington CH, ed. Na puti k teoreticheskoi biologii. Chast’ I. Prolegomeny. Moscow: Mir Publ.; 1970. pp. 11–38.)]
  2. Huntly BJP, Johnson PWM. Targeting Epigenetic Readers in Hematologic Malignancies: A Good BET? The Hematologist. 2012;9(2):5–7.
  3. Daser A, Rabbitts TH. Extending the repertoire of the mixed-lineage leukemia gene MLL in leukemogenesis. Genes & Dev. 2004;18:965–74. doi: 10.1101/gad.1195504.
  4. Ansorge WJ. Next-generation DNA sequencing techniques. New Biotechnol. 2009;25(4):195–203. doi: 10.1016/j.nbt.2008.12.009.
  5. Foley SB, Rios JJ, Mgbemena V. Use of Whole Genome Sequencing for Diagnosis and Discovery in the Cancer Genetics Clinic. EBioMedicine. 2014;2(1):74–81. doi: 10.1016/j.ebiom.2014.12.003.
  6. Wojdacz TK, Moller TH, Thestrup BB, et al. Limitations and advantages of MS-HRM and bisulfite sequencing for single locus methylation studies. Exp Rev Mol Diagn. 2010;10(5):575–80. doi: 10.1586/erm.10.46.
  7. Reinders J, Paszkowski J. Bisulfite methylation profiling of large genomes. Epigenomics. 2010;2(2):209–20. doi: 10.2217/epi.10.6.
  8. Thompson CB. Targeting Metabolic Inputs into Epigenetic Regulations of Acute Leukemia. Blood. 2013;122(21):SCI-26.
  9. Зиновкина Л.А., Зиновкин Р.А. Метилирование ДНК, митохондрии и программируемое старение. Биохимия. 2015;80(12):1830–7.
    [Zinovkina LA, Zinovkin RA. DNA methylation, mitochondria, and programmed aging. Biokhimiya. 2015;80(12):1830–7. (In Russ)]
  10. Vanyushin BF, Kiryanov GI, Kudryashova IB, Belozersky AN. DNA & methylase in loach embryos (Misgurnus fossilis). FEBS Lett. 1971;15(4):313–6. doi: 10.1016/0014-5793(71)80646-4.
  11. Vanyushin BF, Kirnos MD. The nucleotide composition and pyrimidine clusters in DNA from beef heart mitochondria. FEBS Lett. 1974;39(2):195–9. doi: 10.1016/0014-5793(74)80049-99.
  12. Vanyushin BF, Kirnos MD. The structure of animal mitochondrial DNA (base composition, pyrimidine clusters, character of methylation). Mol Cell Biochem. 1977;14(1–3):31–6. doi: 10.1007/bf01734162.
  13. Byun HM, Panni T, Motta V, et al. Effects of airborne pollutants on mitochondrial DNA methylation. Part Fibre Toxicol. 2013;10(1):18. doi: 10.1186/1743-8977-10-18.
  14. Sun C, Reimers LL, Burk RD. Methylation of HPV16 genome CpG sites is associated with cervix precancer and cancer. Gynecol Oncol. 2011;121(1):59–63. doi: 10.1016/j.ygyno.2011.01.013.
  15. Vanyushin BF, Nemirovsky LE, Klimenko VV, et al. The 5-methylcytosine in DNA of rats. Gerontologia. 1973;19(3):138–52. doi: 10.1159/000211967.
  16. Биология и медицина. Метилирование РНК. [Электронный документ] Доступно по: http://medbiol.ru/medbiol/epigenetica/001a1613.htm. Ссылка активна на 14.05.2013.
    [Biologiya i meditsina. Metilirovanie RNK. (Biology and Medicine. RNA Methylation) [Internet]. Available from: http://medbiol.ru/medbiol/epigenetica/001a1613.htm. (accessed 14.05.2013) (In Russ)]
  17. Yu B, Yang Z, Li J, et al. Methylation as a crucial step in plant microRNA biogenesis. Science. 2005;307(5711):932–5. doi: 10.1126/science.1107130.
  18. Goll MG, Kirpekar E, Maggert KA, et al. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science. 2006;311(5759):395–8. doi: 10.1126/science.1120976.
  19. Dominissini D, Nachtergaele S, Moshitch-Moshkovitz S, et al. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature. 2016;530(7591):441–6. doi: 10.1038/nature16998.
  20. Christman J. 5-Azacytidine and 5-aza-2¢-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene. 2002;21(35):5483–95. doi: 10.1038/sj.onc.1205699.
  21. Kumar A, List A. F, Hozo I, et al. Decitabine versus 5-azacitidine for the treatment of myelodysplastic syndrome: adjusted indirect meta-analysis. Haematologica. 2010;95(2):340–2. doi: 10.3324/haematol.2009.017764.
  22. Phase II Decitabine (DAC) Versus Azacitidine (AZA) in Myelodysplastic Syndrome (MDS). [Internet] Available from: http://www.druglib.com/trial/80/NCT02269280.html. (accessed 15.05.2016).
  23. Fenaux P, Gattermann N, Seymour JF, et al. Prolonged survival with improved tolerability in higher-risk myelodysplastic syndromes: azacitidine compared with low dose ara-C. Br J Haematol. 2010;149(2):244–9. doi: 10.1111/j.1365-2141.2010.08082.x.
  24. Al-Ali HK, Jaekel N, Niederwieser D. The role of hypomethylating agents in the treatment of elderly patients with AML. J Geriatr Oncol. 2014;5(1):89–105. doi: 10.1016/j.jgo.2013.08.004.
  25. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109(6):1114–24. doi: 10.1002/cncr.22496.
  26. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30(21):2670–7. doi: 10.1200/jco.2011.38.9429.
  27. European Medicines Agency: assessment report on Dacogen 19 July 2012. [Internet] Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/002221/WC500133571.pdf2012. (accessed 17.05.2016).
  28. Minutes for the February 9 2012 meeting of the FDA Oncologic Drugs Advisory Committee. [Internet] Available from: http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/OncologicDrugsAdvisoryCommittee/UCM293710.pdf2012. (accessed 19.05.2016).
  29. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454–65. doi: 10.1182/blood-2012-03-420489.
  30. Schanz J, Tuchler H, Sole F, et al. New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge. J Clin Oncol. 2012;30(8):820–9. doi: 10.1200/jco.2011.35.6394.
  31. Kantarjian H, O’Brien S, Ravandi F, et al. Proposal for a new risk model in myelodysplastic syndrome that accounts for events not considered in the original International Prognostic Scoring System. Cancer. 2008;113(6):1351–61. doi: 10.1002/cncr.23697.
  32. Garcia-Manero G. Myelodysplastic syndromes: 2015 Update on diagnosis, risk-stratification and management. Am J Hematol. 2015;90(9):831–41. doi: 10.1002/ajh.24102.
  33. Garcia-Manero G, Fenaux P. Hypomethylating agents and other novel strategies in myelodysplastic syndromes. J Clin Oncol. 2011;29(10):516–23. doi: 10.1200/jco.2010.31.0854.
  34. 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.
  35. Garcia-Manero G, Gore SD, Cogle C, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol. 2011;29(18):2521–7. doi: 10.1200/jco.2010.34.4226.
  36. Garcia-Manero G, Jabbour E, Borthakur G, et al. Randomized open-label phase II study of decitabine in patients with low- or intermediate-risk myelodysplastic syndromes. J Clin Oncol. 2013;31(20):2548–53. doi: 10.1200/jco.2012.44.6823.
  37. Wei Y, Dimicoli S, Bueso-Ramos C, et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia. 2013;27(9):1832–40. doi: 10.1038/leu.2013.180.
  38. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: A randomised, open-label, phase III study. Lancet Oncol. 2009;10(3):223–32. doi: 10.1016/s1470-2045(09)70003-8.
  39. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci USA. 2010;107(16):7473–8. doi: 10.1073/pnas.1002650107.
  40. Itzykson R, Thepot S, Quesnel B, et al. Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. Blood. 2011;117(2):403–11. doi: 10.1182/blood-2010-06-289280.
  41. Jabbour E, Garcia-Manero G, Batty N, et al. Outcome of patients with myelodysplastic syndrome after failure of decitabine therapy. Cancer. 2010;116(16):3830–4. doi: 10.1002/cncr.25247.
  42. Montalban-Bravo G, Garcia-Manero G. Novel drugs for older patients with acute myeloid leukemia. Leukemia. 2015;29(4):760–9. doi: 10.1038/leu.2014.244.
  43. 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.
  44. Pleyer L, Burgstaller S, Girschikofsky M, et al. Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-Study Group. Ann Hematol. 2014;93(11):1825–38. doi: 10.1007/s00277-014-2126-9.
  45. Radujkovic A, Dietrich S, Bochtler T, et al. Azacitidine and low-dose cytarabine in palliative patients with acute myeloid leukemia and high bone marrow blast counts – a retrospective single-center experience. Eur J Haematol. 2014;93(2):112–7. doi: 10.1111/ejh.12308.
  46. Field T, Perkins J, Huang Y, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  47. Gerds AT, Gooley TA, Estey EH, et al. Pretransplantation Therapy with Azacitidine vs Induction Chemotherapy and Posttransplantation Outcome in Patients with MDS. Biol Blood Marrow Transplant. 2012;18(8):1211–8. doi: 10.1016/j.bbmt.2012.01.009.
  48. Damaj G, Duhamel A, Robin M, et al. Impact of azacitidine before allogeneic stem-cell transplantation for myelodysplastic syndromes: a study by the Societe Francaise de Greffe de Moelle et de Therapie-Cellulaire and the Groupe-Francophone des Myelodysplasies. J Clin Oncol. 2012;30(36):4533–40. doi: 10.1200/jco.2012.44.3499.
  49. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogeneous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116(23):5420–31. doi: 10.1002/cncr.25500.
  50. 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.
  51. 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.
  52. Lubbert M, Bertz H, Wasch R, et al. Efficacy of a 3-day, low-dose treatment with 5-azacytidine followed by donor lymphocyte infusions in older patients with acute myeloid leukemia or chronic myelomonocytic leukemia relapsed after allografting. Bone Marrow Transplant. 2010;45:627–32. doi: 10.1038/bmt.2009.222.
  53. Sanchez-Abarca LI, Gutierrez-Cosio S, Santamaria C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115(1):107–21. doi: 10.1182/blood-2009-03-210393.
  54. Goodyear О, Agathanggelou A, Novitzky-Basso, et al. Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. 2010;116(11):1908–18. doi: 10.1182/blood-2009-11-249474.
  55. Atanackovich D, Luetkens T, Kloth B, et al. Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. Am J Hematol. 2011;86(11):918–22. doi: 10.1002/ajh.22141.
  56. Kroger N, Bacher U, Bader P, et al. NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the committee on disease-specific methods and strategies for monitoring relapse following allogeneic stem cell transplantation: II. Chronic leukemias, myeloproliferative neoplasms, and lymphoid malignancies. Biol Blood Marrow Transplant. 2010;16(10):1325–46. doi: 10.1016/j.bbmt.2010.06.008.
  57. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia. 2012;26(3):381–9. doi: 10.1038/leu.2011.234.
  58. Sockel K, Wermke M, Radke J, et al. Minimal Residual Disease-Directed Preemptive Treatment With Azacitidine In Patients With NPM1-Mutant Acute Myeloid Leukemia And Molecular Relapse. Haematologica. 2011;96(10):1568–70. doi: 10.3324/haematol.2011.044388.
  59. The MDS Foundation. New MDS Clinical Trials. [Internet] Available from: http://www.mds-foundation.org/clinical-trial-announcements/#New-MDS-Clinical-Trials. (accessed 17.05.2016).

 

Hematological Improvement is a Favorable Response to Azacitidine in Patients with Acute Myeloid Leukemias and Myelodysplastic Syndromes

I.I. Kostroma1, S.V. Gritsaev1, E.V. Karyagina2, A.S. Nizamutdinova3, I.S. Martynkevich1, K.M. Abdulkadyrov1

1 Russian Scientific Research Institute of Hematology and Transfusiology under the Federal Medico-Biological Agency, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

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

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

For correspondence: Ivan Ivanovich Kostroma, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)717-58-57; e-mail: obex@rambler.ru

For citation: Kostroma II, Gritsaev SV, Karyagina EV, et al. Hematological Improvement is a Favorable Response to Azacitidine in Patients with Acute Myeloid Leukemias and Myelodysplastic Syndromes. Clinical oncohematology. 2015;8(4):413–419 (In Russ).

DOI: 10.21320/2500-2139-2015-8-4-413-419


ABSTRACT

Aim. To evaluate types of response to azacitidine associated with improvement of overall survival (OS) rates of patients with acute myeloid leukemias (AML) and myelodysplastic syndromes (MDS).

Methods. A retrospective analyses of medical records of 14 AML patients and 13 MDS patients at the age of 39 to 84 treated with azacitidine at a dose of 75 mg/m2 subcutaneously for 7 subsequent days every 28 days was performed. The therapy effectiveness was evaluated according to modified 2006 IWG criteria. The OS was calculated beginning with the date of initiation of the azacitidine therapy.

Results. From 2 to 25 azacitidine cycles was performed. Complete remission (CR) was achieved in 6 patients (22.2 %) including 4 AML and 2 MDS patients. Bone marrow remission (mCR) was diagnosed in 1 MDS patient (3.7 %). Hematological improvement was obtained in 11 patients (40.7 %) including 5 AML and 6 MDS patients. The overall response was 66.7 % (18 to 27 patients). There was no correlation between the therapy effectiveness and patients’ age, disease type, duration of the previous period, baseline hemoglobin, leukocytes, and platelets levels, and dependence on transfusions of erythrocyte suspension and thromboconcentrate. The therapy was considered ineffective in 9 patients (33.3 %). Stabilization with retained requirements of blood component transfusion was observed in 4 AML and 3 MDS patients. 2 patients presented gradual increase of the blast cell count in the bone marrow. The follow-up period was 2–29 months. The median OS of all patients was 11.5 months. The median OS of patients with CR, mCR and hematological improvement was significantly greater than that in the group of patients with stable disease and progression: 15.9 versus 7.4 months, respectively (= 0,010).

Conclusion. Reduction of transfusion requirement and/or stable improvement of peripheral blood levels due to azacitidine administration are associated with improved OS rates of AML and MDS patients.


Keywords: acute myeloid leukemia, myelodysplastic syndromes, azacitidine, hematological improvement, overall survival.

Received: April 6, 2015

Accepted: October 22, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Грицаев С.В. Миелодиспластические синдромы. В кн.: Гематология. Национальное руководство по гематологии. Под ред. О.А. Рукавицина. М: ГЭОТАР-Медиа, 2015. С. 300–33.
    [Gritsaev SV. Myelodysplastic syndromes. In: Rukavitsin OA, ed. Gematologiya. Natsional’noe rukovodstvo po gematologii. (Hematology. National guidelines in hematology.) Moscow: GEOTAR-Media Publ.; 2015. p. 300–33. (In Russ)]
  2. Грицаев С.В., Мартынкевич И.С., Абдулкадыров К.М. и др. Возрастные особенности кариотипа больных острым миелоидным лейкозом. Терапевтический архив. 2011;1:51–5.
    [Gritsaev SV, Martynkevich IS, Abdulkadyrov KM, et al. Age-related features of karyotype of patients with acute myeloid leukemias. Terapevticheskii arkhiv. 2011;1:51–5. (In Russ)]
  3. Грицаев С.В., Мартынкевич И.С., Абдулкадыров К.М. и др. Комплексный кариотип — маркер крайне неблагоприятного прогноза у больных острыми миелоидными лейкозами и развернутыми вариантами миелодиспластического синдрома старше 70 лет с высоким индексом коморбидности. Терапевтический архив. 2012;7:16–21.
    [Gritsaev SV, Martynkevich IS, Abdulkadyrov KM, et al. Complex karyotype is a marker for extremely unfavorable prognosis in patients with acute myeloid leukemia and marked myelodysplastic syndrome in patients over 70 years old with high co-morbidity index. Terapevticheskii arkhiv. 2012;7:16–21. (In Russ)]
  4. Грицаев С.В., Мартынкевич И.С., Запреева И.М. и др. Эффективность первого и повторного курсов индукционной терапии больных de novo острым миелоидным лейкозом. Бюллетень СО АМН 2013;33(1):67–75.
    [Gritsaev SV, Martynkevich IS, Zapreeva IM, et al. Efficacy of the first and repeated courses of induced therapy of patients with de novo acute myeloid leukemia. Byulleten’ SO AMN. 2013;33(1):67–75. (In Russ)]
  5. Goldstone AH, Burnett AK, Wheatley K, et al. Attempts to improve treatment outcomes in acute myeloid leukemia in older patients: the results of the United Kingdom Medical Research Council AML11 trial. Blood 2001;98(5):1302–11. doi: 10.1182/blood.v98.5.1302.
  6. Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 2011;29(5):487–94. doi: 10.1200/jco.2010.30.1820.
  7. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109(6):1114–24. doi: 10.1002/cncr.22496.
  8. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10(3):223–32. doi: 10.1016/s1470-2045(09)70003-8.
  9. 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.
  10. Quintas-Cardama A, Ravandi F, Liu-Dumlao T, et al. Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia. Blood. 2012;120(24):4840–5. doi: 10.1182/blood-2012-06-436055.
  11. De Padua Silva L, de Lima M, Kantarjian H, et al. Feasibility of allo-SCT after hypomethylating therapy with decitabine for myelodysplastic syndrome. Bone Marrow Transplant. 2009;43(11):839–43. doi: 10.1038/bmt.2008.400.
  12. Field T, Perkins J, Huang Y, et al. 5-Azacitidine for myelodysplasia before allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2010;45(2):255–60. doi: 10.1038/bmt.2009.134.
  13. Lubbert M, Bertz H, Muller MJ, Finke J. When azanucleoside treatment can be curative: nonintensive bridging strategy before allografting in older patients with myelodysplastic syndrome/acute myeloid leukemia. J Clin Oncol. 2013;31(6):822–3. doi: 10.1200/jco.2012.46.4222.
  14. Cheson BD, Greenberg PL, Bennett JM, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108(2):419–25. doi: 10.1182/blood-2005-10-4149.
  15. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World health Organisation (WHO) classification of myeloid neoplasms and acute myeloid leukemia: rationale and important changes. Blood. 2008;114(5):937–51. doi: 10.1182/blood-2009-03-209262.
  16. Грицаев С.В., Мартынкевич И.С., Кострома И.И. Азацитидин при остром миелобластном лейкозе и миелодиспластическим синдроме. Гематология и трансфузиология. 2012;1:23–9.
    [Gritsaev SV, Martynkevich IS, Kostroma II. Azacitidine in acute myeloblast leukemia and myelodysplactic syndrome. Gematologiya i transfuziologiya. 2012;1:23–9. (In Russ)]
  17. Smith BD, Beach CL, Mahmoud D, et al. Survival and hospitalization among patients with acute myeloid leukemia treated with azacitidine or decitabine in a large managed care population: a real-world, retrospective, claims-based, comparative analysis. Exp Hematol Oncol. 2014;3(1):1–6. doi: 10.1186/2162-3619-3-10.
  18. Gurion R, Vidal L, Gafter-Gvili A, et al. 5-Azacitidine prolongs overall survival in patients with myelodysplastic syndrome – a systematic review and meta-analysis. Haematologica. 2010;95(2):303–10. doi: 10.3324/haematol.2009.010611.
  19. Saunthararajah Y. Key clinical observations after 5-azacytidine and decitabine treatment of myelodysplastic syndromes suggest practical solutions for better outcomes. Hematol Am Soc Hematol Educ Program. 2013:511–21. doi: 10.1182/asheducation-2013.1.511.
  20. Pleyer L, Burgstaller S, Girschikofsky M, et al. Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-study Group. Ann Hematol. 2014;93(11):1825–38. doi: 10.1007/s00277-014-2126-9.
  21. Ramos F, Thepot S, Pleyer L, et al. Azacitidine frontline therapy for unfit acute myeloid leukemia patients: clinical use and outcome prediction. Leuk Res. 2015;39(3):296–306. doi: 10.1016/j.leukres.2014.12.013.
  22. Abaigar M, Ramos F, Benito R, et al. Prognostic impact of the number of methylated genes in myelodysplastic syndromes and acute myeloid leukemias treated with azacytidine. Ann Hematol. 2013;92(11):1543–52. doi: 10.1007/s00277-013-1799-9.
  23. Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705–12. doi: 10.1182/blood-2014-06-582809.
  24. Hwang KL, Song MK, Shin HJ, et al. Monosomal and complex karyotypes as prognostic parameters in patients with International Prognostic Scoring System higher risk myelodysplastic syndrome treated with azacitidine. Blood Res. 2014;49(4):234–40. doi: 10.5045/br.2014.49.4.234.
  25. Xicoy B, Jimenez MJ, Garcia O, et al. Results of treatment with azacitidine in patients aged ³75 years included in the Spanish Registry of Myelodysplastic Syndromes. Leuk Lymphoma. 2014;55(6):1300–3. doi: 10.3109/10428194.2013.834532.
  26. Bally C, Ades L, Renneville A, et al. Prognostic value of TP53 gene mutations in myelodysplastic syndromes and acute myeloid leukemia treated with azacitidine. Leuk Res. 2014;38(7):751–5. doi: 10.1016/j.leukres.2014.03.012.
  27. Calvo X, Nomdedeu M, Navarro A, et al. High levels of global DNA methylation are an independent adverse prognostic factor in a series of 90 patients with de novo myelodysplastic syndrome. Leuk Res. 2014;38(8):874–81. doi: 10.1016/j.leukres.2014.04.015.
  28. Poloni A, Maurizi G, Mattiucci D, et al. Azacitidine treatment in high risk myelodysplastic patients in complete haematological remission reverts mesenchymal stem cells to a normal phenotype. Blood. 2014;124(21): Abstract 1904.
  29. Hasserjian RP, Campigotto F, Klepeis V, et al. De novo acute myeloid leukemia with 20–29% blasts is less aggressive than acute myeloid leukemia with ³30% blasts in older adults: a Bone Marrow Pathology Group study. Am J Hematol. 2014;89(11):e193–9. doi: 10.1002/ajh.23808.
  30. Voso MT, Breccia M, Lunghi M, et al. Rapid loss of response after withdrawal of treatment with azacitidine: a case series in patients with higher-risk myelodysplastic syndromes or chronic myelomonocytic leukemia. Eur J Haematol. 2013;90(4):345–8. doi: 10.1111/ejh.12079.
  31. Nazha A, Sekeres MA, Garcia-Manero G, et al. Outcomes of patients with myelodysplastic syndromes who achieve stable disease after treatment with hypomethylating agents. Blood. 2014;124(21): Abstract 3273.

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.

Selection of patients for iron-chelating therapy

S.V. Gritsaev, B. Davaasambuu, N.A. Romanenko, and K.M. Abdulkadyrov

Russian Research Institute of Hematology and Transfusiology, FMBA, Saint Petersburg, Russian Federation


ABSTRACT

Emergence of iron aggressive forms in patients with hematological or oncohematological diseases is potentially life-threatening. This can be caused by several situations: multiple allogeneic RBC transfusions, increased intestinal absorption of iron, or damaging effects of chemotherapeutic agents. The objective of the study was to identify candidates for iron-chelating therapy. The data on 727 patients with screening blood tests were analyzed. The highest serum ferritin level was revealed in patients with thalassemia, PMF, or MDS. They received 80, 37, or 35 RBC transfusions, respectively. The lowest serum ferritin levels were found in patients with CLL or hemolytic anemia that received lesser number of RBC transfusions, namely, 18.5 and 16.5, respectively. The correlation between serum ferritin level and the total number of RBC transfusions was revealed: r = 0.462; p = 0.000. We concluded that iron-chelating therapy is primarily indicated to the patients with MDS or PMF in whom the high serum ferritin level is due to excessive post-transfusion iron.


Keywords: myelodysplastic syndromes, primary myelofibrosis, thalassemia, acute leukemia, multiple myeloma, serum ferritin, allogeneic RBC transfusions.

Read in PDF (RUS)pdficon


REFERENCES

  1. Cazzola M., Della Porta M.G., Malcovati L. Clinical relevance of anemia and transfusion iron overload in myelodysplastic syndromes. Hematology (Am. Soc. Hematol. Educ. Program) 2008; 166–75.
  2. Gattermann N. Pathophysiological and clinical aspects of iron chelation therapy in MDS. Curr. Pharmac. Design 2012; 18: 3222–34.
  3. Gattermann N., Rachmilewitz E.A. Iron overload in MDS — pathophysiology, diagnosis, and complications. Ann. Hematol. 2011; 90: 1–10.
  4. Ghoti H., Fibach E., Merkel D. et al. Changes in parameters of oxidative stress and free iron biomarkers during treatment with deferasirox in ironoverloaded patients with myelodysplastic syndromes. Haematologica 2010; 95: 1433–4.
  5. Leitch H.A. Controversies surrounding iron chelation therapy for MDS. Blood Rev. 2011; 25: 17–31.
  6. Origa R., Galanello R., Ganz T. et al. Liver iron concentrations and urinary hepcidin in b-thalassemia. Haematologica 2007; 92: 583–8.
  7. Park S., Sapena R., Keladi C. et al. Ferritin level at diagnosis is not correlated with poorer survival in non RBC transfusion dependent lower risk de novo MDS. Leuk. Res. 2011; 35: 1530–3.
  8. Knovich M.A., Storey J.A., Coffman L.G., Torti S.V. Ferritin for the clinician. Blood Rev. 2009; 23: 95–104.
  9. Armand P., Kim H.T. Rhodes J. et al. Iron overload in patients with acute leukemia or MDS undergoing myeloablative stem cell transplantation. Biol. Blood Marrow Transplant. 2011; 17: 852–60.
  10. Brittenham G.M., Cohen A.R., McLaren C.E. et al. Hepatic iron stores and plasma ferritin concentration in patients with sickle cell anemia and thalassemia major. Am. J. Hematol. 1993; 42: 81–5.
  11. Roghi A., Cappellini M.D., Wood J.C. et al. Absence of cardiac siderosis despite hepatic iron overload in Italian patients with thalassemia intermedia: an MRI T2* study. Ann. Hematol. 2010; 89: 585–9.
  12. Taher A., Rassi F.E., Ismaeel H. et al. Correlation of liver iron concentration determined by R2 magnetic resonance imaging with serum ferritin in patients with thalassemia intermedia. Haematologica 2008; 93: 1584–5.
  13. Piga A., Longo F., Duca L. et al. High nontransferrin bound iron levels and heart disease in thalassemia major. Am. J. Hematol. 2009; 84: 29–33.
  14. Sahlstedt L., von Bonsdorff L., Ebeling F. et al. Non-transferrin-bound iron in haematological patients during chemotherapy and conditioning for autologous stem cell transplantation. Eur. J. Haematol. 2009; 83: 455–9.
  15. Sahlstedt L., Ebeling F. von Bonsdorff L. et al. Non-transferrin-bound iron during allogeneic stem cell transplantation. Br. J. Haematol. 2001; 113: 836–8.
  16. Andrews N.C. Disorders of iron metabolism. New Engl. J. Med. 1999; 341: 1986–95.
  17. Porter J.B. Practical management of iron overload. Br. J. Haematol. 2001; 115: 239–52.
  18. Dreyfus F. The deleterious effects of iron overload in patients with myelodysplastic syndromes. Blood Rev. 2008; 22(Suppl. 2): S29–34.
  19. Malcovati L., Porta M.G., Pascutto C. et al. Prognostic factors and life expectancy in myelodysplastic syndromes classified according to WHO criteria: a basis for clinical decision making. J. Clin. Oncol. 2005; 23: 7594–603.
  20. Pakbaz Z., Fischer R., Fung E. et al. Serum ferritin underestimates liver iron concentration in transfusion independent thalassemia patients as compared to regularly transfused thalassemia and sickle cell patients. Pediatr. Blood Cancer 2007; 49: 329–32.
  21. Karam L.B., Disco D., Jackson S.M. et al. Liver biopsy results in patients with sickle cell disease on chronic transfusions: poor correlation with ferritin levels. Pediatr. Blood Cancer 2008; 50: 62–5.
  22. Nielsen P., Gunther U., Durken M. et al. Serum ferritin iron in iron overload and liver damage: correlation to body iron stores and diagnostic relevance. J. Lab. Clin. Med. 2000; 135: 413–8.
  23. Valent P., Krieger O., Stauder R. et al. Iron overload in myelodysplastic syndromes (MDS) — diagnosis, management, and response criteria: a proposal of the Austrian MDS platform. Eur. J. Clin. Invest. 2008; 38: 143–9.
  24. Alessandrino E.P., Della Porta M.G., Bacigalupo A. et al. Prognostic impact of pre-transplantation transfusion history and secondary iron overload in patients with myelodysplastic syndrome undergoing allogeneic stem cell transplantation: a GITMO study. Haematologica 2010; 95: 476–84.
  25. Pullarkat V. Iron overload in patients undergoing hematopoietic stem cell transplantation. Adv. Hematol. 2010; pii: 345756.
  26. Грицаев С.В., Абдулкадыров К.М., Шихбабаева Д.И. Миелодиспла- стический синдром (МДС) и перегрузка железом (результаты скрининго- вого обследования 289 больных de novo МДС). Фарматека 2010; 10: 60–7. Gritsayev S.V., Abdulkadyrov K.M., [Shikhbabayeva D.I. Miyelodisplasticheskiy sindrom (MDS) i peregruzka zhelezom (rezultaty skriningovogo obsledovaniya 289 bolnykh de novo MDS) (Myelodisplastic syndrome (MDS) and iron overload (screening study results in 289 patients with de novo MDS)). Farmateka 2010; 10: 60–7.]
  27. Pullarkat V., Blanchard S., Tegtmeier B. et al. Iron overload adversely affects outcome of allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2008; 42: 799–805.
  28. Kushner J.P., Porter J.P., Olivieri N.F. Secondary iron overload. Hematology (Am. Soc. Hematol. Educ. Program) 2001: 47–61.
  29. Angelucci E., Barosi G., Camaschella C. et al. Italian Society of Hematology practice guidelines for the management of iron overload in thalassemia major and related disorders. Haematologica 2008; 93: 741–52.
  30. de Swart L., Smith A., Fenaux P. et al. Transfusion-dependency is the most important prognostic factor for survival in 1000 newly diagnosed MDS patients with low- and intermediate-1 risk MDS in the European LeukemiaNet MDS registry. Blood 2011; 118: Abstract 2775.
  31. Armand P., Sainvil M.-M., Kim H.T. et al. Pre-transplantation iron chelation in patients with MDS or acute leukemia and iron overload undergoing myeloablative allo-SCT. Bone Marrow Transplantat. 2012 May 21. doi: 10.1038/ bmt.2012.94.
  32. Cappellini M.D., Porter J., El-Beshlawy A. et al. Tailoring iron chelation by iron intake and serum ferritin: the prospective EPIC study of deferasirox in 1744 patients with transfusion-dependent anemias. Haematologica 2010; 95: 557–66.
  33. Cilloni D., Messa E., Biale L. et al. High rate of erythroid response during iron chelation therapy in a cohort of 105 patients affected by hematologic malignancies with transfusional iron overload: an Italian multicenter retrospective study. Blood 2011; 118: Abstract 611.
  34. Gattermann N., Finelli C., Della Porta M. et al. Hematologic responses to deferasirox therapy in transfusion-dependent patients with myelodysplastic syndromes. Haematologica 2012; 97: 1364–71.
  35. Neukirchen J., Fox F., Kundgen A. et al. Improved survival in MDS patients receiving iron chelation therapy — a matched pair analysis of 188 patients from the Dusseldorf MDS registry. Leuk. Res. 2012; 36: 1067–70.
  36. Pullarkat V. Objectives of iron chelation therapy in myelodysplastic syndromes: more than meets the eye? Blood 2009; 114: 5251–5.
  37. Leitch H.A., Chase J.M., Goodman T.A. et al. Improved survival in red blood cell transfusion dependent patients with primary myelofibrosis (PMF) receiving iron chelation therapy. Hematol. Oncol. 2010; 28: 40–8.
  38. Armand P., Kim H.T., Cutler S.C. et al. Prognostic impact of elevated pretransplantation serum ferritin in patients undergoing myeloablative stem cell transplantation. Blood 2007; 109: 4586–8.
  39. Kanda J., Kawabata H., Chao N.J. Iron overload and allogeneic hematopoietic stem-cell transplantation. Expert Rev. Hematol. 2011; 4: 71–80.
  40. Lim Z.Y., Fiaccadori V., Gandhi S. et al. Impact of pre-transplant serum ferritin on outcomes of patients with myelodysplastic syndromes or secondary acute myeloid leukemia receiving reduced intensity conditioning allogeneic haematopoietic stem cell transplantation. Leuk. Res. 2010; 34: 723–7.
  41. Maradei S.C., Maiolino A., de Azevedo A.M. et al. Serum ferritin as risk factor for sinusoidal obstruction syndrome of the liver in patients undergoing hematopoietic stem cell transplantation. Blood 2009; 114: 1270–5.
  42. Platzbecker U., Bornhauser M., Germing U. et al. Red blood cell transfusion-dependence and outcome after allogeneic peripheral blood stem cell transplantation in patients with de novo myelodysplastic syndromes (MDS). Biol. Blood Marrow Transplant. 2008; 14: 1217–25.
  43. Tanaka V., Tachibana N., Numata A. et al. A prognostic score with pretransplant serum ferritin and disease status predicts outcome following reduced-intensity SCT. Bone Marrow Transplant. 2012; 47: 596–7.
  44. Lee J.W., Kang H.J., Kim E.K. et al. Effect of iron overload and ironchelating therapy on allogeneic hematopoietic SCT in children. Bone Marrow Transplant. 2009; 44: 793–7.
  45. Fritsch A., Langebrake C., Nielsen P. et al. Deferasirox (Exjade®) given during conditioning regimen (FLAMSA/Busulfan/ATG) reduces the appearance of labile plasma iron in patients undergoing allogeneic stem cell transplantation. Blood 2011; 118: Abstract 3023.
  46. Armand P., Sainvil M.-M., Kim H.T. et al. Does iron overload really matter in stem cell transplantation? Am. J. Hematol. 2012; 87: 569–72.
  47. Pardanani A., Tefferi A. Prognostic relevance of anemia and transfusion dependency in myelodysplastic syndromes and primary myelofibrosis. Haematologica 2011; 96: 8–10.
  48. Ali S., Pimentel J.D., Munoz J. et al. Iron overload in allogeneic hematopoietic stem cell transplant recipients. Arch. Pathol. Lab. Med. 2012; 136: 532–8.