Journal List > Korean J Hematol > v.42(2) > 1032766

Cheong, Kook, Bang, Lee, Joo, Kim, Kim, Park, Park, Ahn, Yoon, Won, Lee, Jung, Jo, Cho, Han, Min, and Kim: The Clinical Guidelines for Myelodysplastic Syndrome

Abstract

The myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis associated with multilineage cytopenias leading to serious morbidity or mortality, and the additional risk of leukemic transformation. The management of patients with MDS can be very complex and varies according to both the clinical manifestations in individual patients as well as the presence of complicating medical conditions. However, therapeutic dilemmas still exist for MDS due to the multifactorial pathogenetic features of the disease, its heterogeneous stages, and the elderly patient population. For these reasons, proper guidelines for management are necessary. This review describes the proper diagnosis for MDS, decision-making approaches for optimal therapeutic options that are based on a consideration of patient
clinical factors and risk-based prognostic categories, and the use of recently available biospecific drugs such as hypomethylating agents that are potentially capable of abrogating the abnormalities associated with MDS. Proper indications and methods for transplantation, response criteria, management for iron overload for highly transfused patients and specific considerations for MDS in childhood are also described. All of these topics were discussed at the third symposium of AML/MDS working party on 3 March, 2007.

REFERENCES

1). Jaffe ES., Harris NL., Stein H., Vardiman JW. World Health Organization classification of tumours. Pathology and genetics of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC Press. 2001. 61–73.
2). Vardiman JW., Harris NL., Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood. 2002. 100:2292–302.
crossref
3). Greenberg PL., Baer MR., Bennett JM, et al. NCCN practice guideline in oncology: Myelodysplastic syndromes-v.1.2007. (Accessed March 2, 2007, at. http://www.nccn.org/professionals/physician_gls/PDF/mds.pdf.
4). Valent P., Horny HP., Bennett JM, et al. Definitions and standards in the diagnosis and treatment of the myelodysplastic syndromes: consensus statements and report from a working conference. Leuk Res. 2007. 31:727–36.
crossref
5). Vardiman JW. Hematopathological concepts and controversies in the diagnosis and classification of myelodysplastic syndromes. Hematology Am Soc Hematol Educ Program. 2006. 199–204.
crossref
6). Kussick SJ., Fromm JR., Rossini A, et al. Four-color flow cytometry shows strong concordance with bone marrow morphology and cytogenetics in the evaluation for myelodysplasia. Am J Clin Pathol. 2005. 124:170–81.
crossref
7). Kong SY., Nam MH., Woo HY, et al. Assessment of the diagnostic utility of methylmalonic acid in megaloblastic anemia due to vitamin B12 deficiency. Korean J Lab Med. 2002. 22:145–52.
8). Santini V., Kantarjian HM., Issa JP. Changes in DNA methylation in neoplasia: pathophysiology and therapeutic implications. Ann Intern Med. 2001. 134:573–86.
crossref
9). Lübbert M. Gene silencing of the p15/INK4B cell-cycle inhibitor by hypermethylation: an early or later epigenetic alteration in myelodysplastic syndromes? Leukemia. 2003. 17:1762–4.
crossref
10). Silverman LR., Demakos EP., Peterson BL, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002. 20:2429–40.
crossref
11). Kantarjian H., Issa JP., Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodys-plastic syndromes: results of a phase III randomized study. Cancer. 2006. 106:1794–803.
12). Kaminskas E., Farrell A., Abraham S, et al. Approval summary: azacitidine for treatment of myelodys-plastic syndrome subtypes. Clin Cancer Res. 2005. 11:3604–8.
crossref
13). Kantarjian H., Oki Y., Garcia-Manero G, et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 2007. 109:52–7.
crossref
14). Bowen D., Culligan D., Jowitt S, et al. Guidelines for the diagnosis and therapy of adult myelodysplastic syndromes. Br J Haematol. 2003. 120:187–200.
crossref
15). Alessandrino EP., Amadori S., Barosi G, et al. Evidence-and consensus-based practices guidelines for the therapy of primary myelodysplastic syndromes. A statement from the Italian Society of Hematology. Haematologica. 2002. 87:1286–306.
16). Lee JH., Lee JH., Shin YR, et al. Application of different prognostic scoring systems and comparison of the FAB and WHO classifications in Korean patients with myelodysplastic syndrome. Leukemia. 2003. 17:305–13.
crossref
17). Schiffer CA. Clinical issues in the management of patients with myelodysplasia. Hematology Am Soc He-matol Educ Program. 2006. 205–10.
crossref
18). Cutler CS., Lee SJ., Greenberg P, et al. A decision analysis of allogeneic bone marrow transplantation for the myelodysplastic syndromes: delayed transplantation for low-risk myelodysplasia is associated with improved outcome. Blood. 2004. 104:579–85.
crossref
19). Scott BL., Sandmaier BM., Storer B, et al. Myeloablative vs nonmyeloablative allogeneic transplantation for patients with myelodysplastic syndrome or acute myelogenous leukemia with multilineage dysplasia: a retrospective analysis. Leukemia. 2006. 20:128–35.
crossref
20). Martino R., Iacobelli S., Brand R, et al. Retrospective comparison of reduced-intensity conditioning and conventional high-dose conditioning for allogeneic hematopoietic stem cell transplantation using HLA-identical sibling donors in myelodysplastic syndromes. Blood. 2006. 108:836–46.
crossref
21). Castro-Malaspina H., Harris RE., Gajewski J, et al. Unrelated donor marrow transplantation for myelo-dysplastic syndromes: outcome analysis in 510 transplants facilitated by the National Marrow Donor Program. Blood. 2002. 99:1943–51.
crossref
22). Deeg HJ. Optimization of transplant regimens for patients with myelodysplastic syndrome (MDS). Hematology Am Soc Hematol Educ Program. 2005. 167–73.
crossref
23). Deeg HJ., Storer B., Slattery JT, et al. Conditioning with targeted busulfan and cyclophosphamide for hemopoietic stem cell transplantation from related and unrelated donors in patients with myelodys-plastic syndrome. Blood. 2002. 100:1201–7.
crossref
24). Andersson BS., Kashyap A., Gian V, et al. Conditioning therapy with intravenous busulfan and cyclophosphamide (IV BuCy2) for hematologic malignancies prior to allogeneic stem cell transplantation: a phase II study. Biol Blood Marrow Transplant. 2002. 8:145–54.
crossref
25). de Lima M., Couriel D., Thall PF, et al. Once-daily intravenous busulfan and fludarabine: clinical and pharmacokinetic results of a myeloablative, reduced-toxicity conditioning regimen for allogeneic stem cell transplantation in AML and MDS. Blood. 2004. 104:857–64.
crossref
26). Russell JA., Tran HT., Quinlan D, et al. Once-daily intravenous busulfan given with fludarabine as conditioning for allogeneic stem cell transplantation: study of pharmacokinetics and early clinical outcomes. Biol Blood Marrow Transplant. 2002. 8:468–76.
crossref
27). Ho AY., Pagliuca A., Kenyon M, et al. Reduced-intensity allogeneic hematopoietic stem cell transplantation for myelodysplastic syndrome and acute myeloid leukemia with multilineage dysplasia using fludarabine, busulphan, and alemtuzumab (FBC) conditioning. Blood. 2004. 104:1616–23.
crossref
28). Chan GW., Foss FM., Klein AK., Sprague K., Miller KB. Reduced-intensity transplantation for patients with myelodysplastic syndrome achieves durable remission with less graft-versus-host disease. Biol Blood Marrow Transplant. 2003. 9:753–9.
crossref
29). de Lima M., Anagnostopoulos A., Munsell M, et al. Nonablative versus reduced-intensity conditioning regimens in the treatment of acute myeloid leukemia and high-risk myelodysplastic syndrome: dose is relevant for long-term disease control after allogeneic hematopoietic stem cell transplantation. Blood. 2004. 104:865–72.
crossref
30). Schmid C., Schleuning M., Ledderose G., Tischer J., Kolb HJ. Sequential regimen of chemotherapy, reduced-intensity conditioning for allogeneic stem-cell transplantation, and prophylactic donor lymphocyte transfusion in high-risk acute myeloid leukemia and myelodysplastic syndrome. J Clin Oncol. 2005. 23:5675–87.
crossref
31). de Witte T., Pikkemaat F., Hermans J, et al. Genotypically nonidentical related donors for transplantation of patients with myelodysplastic syndromes: comparison with unrelated donor transplantation and autologous stem cell transplantation. Leukemia. 2001. 15:1878–84.
crossref
32). Koh LP., Chao NJ. Umbilical cord blood transplantation in adults using myeloablative and nonmye-loablative preparative regimens. Biol Blood Marrow Transplant. 2004. 10:1–22.
crossref
33). Guardiola P., Runde V., Bacigalupo A, et al. Retrospective comparison of bone marrow and granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells for allogeneic stem cell transplantation using HLA identical sibling donors in myelodysplastic syndromes. Blood. 2002. 99:4370–8.
crossref
34). Anderson JE. Bone marrow transplantation for myelodysplasia. Blood Rev. 2000. 14:63–77.
crossref
35). Fukumoto JS., Greenberg PL. Management of patients with higher risk myelodysplastic syndromes. Crit Rev Oncol Hematol. 2005. 56:179–92.
crossref
36). 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. 108:419–25.
crossref
37). Cheson BD., Bennett JM., Kantarjian H, et al. Report of an international working group to standarize response criteria for myelodysplastic syndromes. Blood. 2000. 96:3671–4.
38). List A., Kurtin S., Roe DJ, et al. Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med. 2005. 352:549–57.
crossref
39). Lübbert M., Wijermans P., Kunzmann R, et al. Cytogenetic responses in high-risk myelodysplastic syndrome following low-dose treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Br J Haematol. 2001. 114:349–57.
crossref
40). Lim Z., Killicks S., Cavenagh JD, et al. European multi-centre study on the use of anti-thymocyte globulin in the treatment of myelodysplastic syndromes[ab-stract 2581]. Blood. 2005. 106:707a.
41). Kornblith AB., Hemdon JE., Silverman LR, et al. Impact of azacytidine on the quality of life of patients with myelodysplastic syndrome treated in a randomized phase III trial: a Cancer and Leukemia Group B study. J Clin Oncol. 2002. 20:2441–52.
crossref
42). Andrews NC. Disorders of iron metabolism. N Engl J Med. 1999. 341:1986–95.
crossref
43). Piperno A. Classification and diagnosis of iron overload. Haematologica. 1998. 83:447–55.
44). Gabuti V., Piga A. Results of long-term iron-chelating therapy. Acta Haematol. 1996. 95:26–36.
crossref
45). Olivieri NF., Brittenham GM. Iron-chelating therapy and the treatment of thalassemia. Blood. 1997. 89:739–61.
crossref
46). Ishizaka N., Saito K., Mitani H, et al. Iron overload augments angiotensin II-induced cardiac fibrosis and promotes neointima formation. Circulation. 2002. 106:1840–6.
crossref
47). Jensen PD. Evaluation of iron overload. Br J Hae-matol. 2004. 124:697–711.
crossref
48). Porter JB. Practical management of iron overload. Br J Haematol. 2001. 115:239–52.
crossref
49). Angelucci E., Brittenham GM., McLaren CE, et al. Hepatic iron concentration and total body iron stores in thalassemia major. N Engl J Med. 2000. 343:327–31.
crossref
50). Liu ZD., Hider RC. Design of clinically useful iron (III)-selective chelators. Med Res Rev. 2002. 22:26–64.
51). Piga A., Galanello R., Forni GL, et al. Randomized phase II trial of deferasirox (Exjade, ICL670), a once-daily, orally-administered iron chelator, in comparison to deferoxamine in thalassemia patients with transfusional iron overload. Haematologica. 2006. 91:873–80.
52). Greenberg PL. Myelodysplastic syndromes: iron overload consequences and current chelating therapies. J Natl Compr Canc Netw. 2006. 4:91–6.
crossref
53). Hasle H., Niemeyer CM., Chessells JM, et al. A pediatric approach to the WHO classification of myelodys-plastic and myeloproliferative diseases. Leukemia. 2003. 17:277–82.
crossref
54). Hasle H. Myelodysplastic and myeloproliferative disorders in children. Curr Opin Pediatr. 2007. 19:1–8.
crossref
55). Mandel K., Dror Y., Poon A., Freedman MH. A practical, comprehensive classification for pediatric mye-lodysplastic syndromes: the CCC system. J Pediatr Hematol Oncol. 2002. 24:343–52.
crossref
56). Occhipinti E., Correa H., Yu L., Craver R. Comparison of two new classifications for pediatric myelodys-plastic and myeloproliferative disorders. Pediatr Blood Cancer. 2005. 44:240–4.
crossref
57). Niemeyer CM., Locatelli F. Chronic myeloproliferative disorders. Pui CH, editor. Childhood leukemias. 2nd ed.Cambridge, England: Cambridge University Press;2006. p. 571–98.
crossref
58). Kook H., Kim MK., Ghim TT, et al. Pediatric myelo-dysplastic syndrome in Korea: Clinical characteristics and comparison of prognostic scoring systems. Korean J Pediatr Hematol-Oncol. 2003. 10:1–13.
59). Woods WG., Barnard DR., Alonzo TA, et al. Prospective study of 90 children requiring treatment for juvenile myelomonocytic leukemia or myelodys-plastic syndrome: a report from the Children's Cancer Group. J Clin Oncol. 2002. 20:434–40.
crossref
60). Webb DK., Passmore SJ., Hann IM., Harrison G., Wheatley K., Chessells JM. Results of treatment of children with refractory anaemia with excess blasts (RAEB) and RAEB in transformation (RAEBt) in Great Britain 1990-99. Br J Haematol. 2002. 117:33–9.
crossref
61). Yusuf U., Frangoul HA., Gooley TA, et al. Allogeneic bone marrow transplantation in children with myelo-dysplastic syndrome or juvenile myelomonocytic leukemia: the Seattle experience. Bone Marrow Transplant. 2004. 33:805–14.
crossref
62). Smith FO., Woods WG. Myeloproliferative and mye-lodysplastic disorders. Pizzo PA, Poplack DG, editors. Principles and practice of pediatric oncology. 5th ed.Philadelphia, USA: Lippincott Williams & Wilkins;2006. p. 673–94.
63). Kang HJ., Shin HY., Choi HS., Ahn HS. Novel regimen for the treatment of juvenile myelomonocytic leukemia (JMML). Leuk Res. 2004. 28:167–70.
crossref
64). Smith FO., King R., Nelson G, et al. Unrelated donor bone marrow transplantation for children with juvenile myelomonocytic leukaemia. Br J Haematol. 2002. 116:716–24.
crossref
65). Locatelli F., Nöllke P., Zecca M, et al. Hematopoietic stem cell transplantation (HSCT) in children with juvenile myelomonocytic leukemia (JMML): results of the EWOG-MDS/EBMT trial. Blood. 2005. 105:410–9.
crossref
66). Vidal DO., Paixão VA., Brait M, et al. Aberrant methylation in pediatric myelodysplastic syndrome. Leuk Res. 2007. 31:175–81.
crossref

Fig. 1
The maintenance of methylation process (top) and reactivation of gene expression (bottom) by hypomethylating agents such as AZA (azacytidine) and DAC (decitabine).
kjh-42-71f1.tif
Fig. 2
Comparison of survival without leukemia between AZA (A) or DAC (B) treatment arm and supportive care arm and supportive care arm in two randomized phase III trials.
kjh-42-71f2.tif
Fig. 3
Dosage modification according to the recovery of blood cell counts.
kjh-42-71f3.tif
Table 1.
The WHO classification of myelodysplastic syndromes
Disease Blood findings Bone marrow findings
Refractory anemia (RA) Anemia Erythroid dysplasia only
No or rare blasts <5% blasts
<1×109/L monocytes <15% ringed sideroblasts
Refractory anemia with ringed Anemia Erythroid dysplasia only
sideroblasts (RARS) No blasts ≥15% ringed sideroblasts
  <5% blasts
Refractory cytopenia with Cytopenias (bicytopenia or Dysplasia in ≥10% of cells in two or
multilineage dysplasia (RCMD) pancytopenia) more myeloid cell lineages
No or rare blasts <5% blasts in marrow
No Auer rods No Auer rods
<1×109/L monocytes <15% ringed sideroblasts
Refractory cytopenia with Cytopenias (bicytopenia Dysplasia in ≥10% of cells in
multilineage dysplasia or pancytopenia) two or more myeloid cell lineages
and ringed sideroblasts No or rare blasts ≥5% ringed sideroblasts
(RCMD-RS) No Auer rods <5% blasts
<1×109/L monocytes No Auer rods
Refractory anemia with excess Cytopenias Unilineage or multilineage dysplasia
blasts-1 (RAEB-1) <5% blasts 5∼9% blasts
No Auer rods No Auer rods
<1×109/L monocytes  
Refractory anemia with excess Cytopenias Unilineage or multilineage dysplasia
blasts-2 (RAEB-2) 5∼19% blasts 10∼19% blasts
Auer rods +/? Auer rods +/?
<1×109/L monocytes  
Myelodysplastic syndrome, Cytopenias Unilineage dysplasia in granulocytes or
unclassified No or rare blasts megakaryocytes
(MDS-U) No Auer rods <5% blasts
  No Auer rods
MDS associated with isolateddel(5q) Anemia Normal to increased megakaryocytes with
<5% blasts hypolobated nuclei
Platelets normal or increased <5% blasts
  No Auer rods Isolated del(5q)
Table 2.
General guidelines of initial evaluation for MDS
Comprehensive history and physical examination
CBC with diff, reticulocyte count
Complete serum chemistry
BM aspiration and biopsy
  Iron stain
  Immunohistochemistry
Chromosome analysis including FISH
Exclusion of viral infections (HCV, HIV, CMV, EBV etc)
Serum B12 and RBC folate/methylmalonic acid and homo-
cysteine
Iron studies: iron/TIBC/ferritin
Table 3.
Current issues for preparing guidelines of stem cell transplantation for MDS
Indication for allogeneic transplantation
Optimal time for transplantation
Conditioning regimens
Donor selectino
Source of stem cells: BM or PB
Previous chemotherapy before transplantation
Indication for autologous stem cell transplantation
Table 4.
Clinical guideline of stem cell transplantation for MDS
Selection of patients Should be individualized
  Criteria IPSS or WHO. Newer classification may be needed.
  Age 65 years (60 years with unrelated donor) or younger
Timing of transplantation Low risk: delayed until progression
  High risk: soon after diagnosis
Conditioning regimen Low risk, old age, comorbid condition: RIC>MC
  High risk: MC is preferred in young patients with good performance
  MC regimen: Busulfan-based>TBI-based
  RIC regimen: various, no one preferred
Donor selection Matched sibling donor is preferred.
  Matched unrelated or mismatched familial donor, or cord blood can be considered.
Hematopoietic cell source High risk, RIC: BM<G-PBMC
Pre-transplant chemotherap High risk: Newer agents can be considered.
Autologous transplantation High risk patients in CR after chemotherapy

Abbreviations: IPSS, International Prognostic Scoring System; low risk, BM blast<5% or IPSS Low/INT-1 risk group; RIC reduced intensity conditioning; MC, myeloablative conditioning; TBI, total body irradiation; BM, bone marrow; G-PBMC G-CSF-mobilized peripheral blood mononuclear cell.

Table 5.
Proposed modified International Working Group response criteria for hematologic improvement
Category Response criteria (responses must last at least 4 wk)
Complete remission Bone marrow: ≤5% myeloblasts with normal maturation of all cell lines
Persistent dysplasia will be noted∗
Peripheral blood
Hgb ≥11g/dL 9
Platelets ≥100×109/L 9
Neutrophils ≥1.0×109/L∗
Blasts 0%
Marrow CR Bone marrow: ≤5% myeloblasts and decrease by ≥50% over pretreatment∗
Peripheral blood: if HI responses, they will be noted in addition to marrow CR∗
Failure Death during treatment or disease progression characterized by worsening of cytopenias,
increase in percentage of bone marrow blasts, or progression to a more advanced MDS
FAB subtype than pretreatment
Relapse after CR or PR At least 1 of the following:
Return to pretreatment bone marrow blast percentage
Decrement of ≥50% from maximum remission/response levels in granulocytes or platelets
Reduction in Hgb concentration by 1.5g/dL or transfusion dependence
Cytogenetic response Complete
Disappearance of the chromosomal abnormality without appearance of new ones
Partial
At least 50% reduction of the chromosomal abnormality
Survival Endpoints:
Overall: death from any cause
Event free: failure or death from any cause
PFS: disease progression or death from MDS
DFS: time to relapse
Cause-specific death: death related to MDS

Deletions to IWG response criteria are not shown. To convert hemoglobin from grams per deciliter to grams per liter, multiply grams per deciliter by 10. Abbreviations: MDS, myelodysplastic syndromes; Hgb, hemoglobin; CR, complete remission; HI, hematologic improvement PR, partial remission; FAB, French-American-British; AML, acute myeloid leukemia; PFS, progression-free survival; DFS, disease-free survival. Dysplastic changes should consider the normal range of dysplastic changes (modification). ∗Modification to IWG response criteria.

Table 6.
Proposed modified International Working Group response criteria for hematologic improvement
Hematologic improvement Response criteria (responses must last at least 8 wk)∗
Erythroid response (pretreatment, <11g/dL) Hgb increase by ≥1.5g/dL
  Relevant reduction of units of RBC transfusions by an absolute number of at least 4 RBC transfusions/8 wk compared with the pretreatment transfusion number in the previous 8 wk. Only RBC transfusions given for a Hgb of ≤9.0g/dL pretreatment will count in the RBC transfusion response evaluation∗
Platelet response (pretreatment, <100×109/L) Absolute increase of ≥30×109/L for patients starting with >20×109/L platelets
  Increase from <20×109/L to >20×109/L and by at least 100%∗
Neutrophil response (pretreatment, <1.0×109/L) At least 100% increase and an absolute increase >0.5×109/L∗
Progression or relapse after HI At least 1 of the following:
  At least 50% decrement from maximum response levels in granulocytes or platelets
  Reduction in Hgb by ≥1.5g/dL
  Transfusion dependence

Deletions to the IWG response criteria are not shown. To convert hemoglobin levels from grams per deciliter to grams per liter, multiply grams per deciliter by 10. Abbreviations: Hgb, hemoglobin; RBC, red blood cell; HI, hematologic improvement. Pretreatment counts averages of at least 2 measurements (not influenced by transfusions) ≥1 week apart (modification). Modification to IWG response criteria. ∗In the absence of another explanation, such as acute infection, repeated courses of chemotherapy (modification), gastrointestinal bleeding, hemolysis, and so forth. It is recommended that the 2 kinds of erythroid and platelet responses be reported overall as well as by the individual response pattern.

Table 7.
Remission rates according to the International Working Group response criteria
  IWG response
Treatement No. patient IPSS (%) % hematologic response Median survival, mo %HI-major
Low Int1 Int2 High Overall CR PR
Azacitidine 99 5 53 23 17 11 10 1 20 36 (+minor
Decitabine 89   31 43 26 17 9 8 22.3/13.3 13
Tipifarnib 82   17 39 44 12 7 5 11.9 22
Arsenic 101 NA 39 NA 61 1 1 0 NA 20

Abbreviations: IWG, International Working Group; IPSS, International Prognostic Scoring System; CR, complete remission PR, partial remission; HI, hematologic improvement; Int1, IPSS intermediate-1; Int2, IPSS intermediate-2; TTL, time to leuke mia; NA, not available.

Table 8.
CCC classification of childhood MDS
Category - etiology of the MDS
  Idiopathic disease
  Treatment related disease
  Syndrome related disease
Cytology - morphologic features of BM and PB
  Refractory cytopenia with ringed sideroblasts
  Refractory cytopenia
  Refractory cytopenia with dysplasia
  Refractory cytopenia with excess blasts (5∼29% blasts
  Refractory cytopenia with dysplasia and excess blasts
Cytogenetics
  Normal cytogenetics
  Abnormal cytogenetics
  Cytogenetics unknown
Table 9.
Pediatric WHO classification
Myelodysplastic/Myeloproliferative disease
  Juvenile myelomonocytic leukemia (JMML)
  Chronic myelomonocytic leukemia (CMML) (secondary only)
  BCR-ABL-negative chronic myeloid leukemia (Ph-CML)
Down syndrome (DS) disease
  Transient abnormal myelopoiesis (TAM)
  Myeloid leukemia of DS
Myelodysplastic syndrome (MDS)
  Refractory cytopenia (RC)
  Refractory anemia with excess blasts (RAEB)
  RAEB in transformation (RAEB-t)
Table 10.
Diagnostic criteria for juvenile myelomonocytic leukemia
I. Clinical and hematological features (all 3 features mandatory)
  Peripheral blood monocyte count >1×109/L
  Blast percentage in PB and BM<20%
  Splenomegaly
II. Oncogenetic studies (1 parameter sufficient)
  Somatic mutation in PTPN11 or RAS
  NF1 mutation or clinical diagnosis of NF1
  Monosomy 7
III. 1. In the absence of 1 parameter listed under II, the following criteria have to be fulfilled
  Absence of Ph' chromosome (bcr/abl rearrangement) (mandatory)
2. And at least two of the following criteria
  Spontaneous growth or GM-CSF hypersensitivity in colony assay
  HgF increased for age
  Myeloid precursors on PB smear
  White blood count >10×109/L
  Clonal abnormality besides monosomy 7
TOOLS
Similar articles