Elsevier

Seminars in Oncology

Volume 35, Issue 4, August 2008, Pages 410-417
Seminars in Oncology

Acute myeloid leukemia
Core Binding Factor Acute Myeloid Leukemia

https://doi.org/10.1053/j.seminoncol.2008.04.011Get rights and content

Core binding factor (CBF) acute myeloid leukemia (AML) is cytogenetically defined by the presence of t(8;21)(q22;q22) or inv(16)(p13q22)/t(16;16)(p13;q22), which are found in approximately 15% of all adult de novo AML cases. At the molecular level, both cytogenetic abnormalities result in disruption of CBF, a transcription factor that functions as an essential regulator of normal hematopoiesis. Despite this molecular commonality, recent studies have demonstrated differences in genetic, clinical, and prognostic features between t(8;21) and inv(16)/t(16;16) AML, thereby supporting the notion that they represent two distinct biologic and clinical entities. Furthermore, despite being considered as a more favorable AML risk group, only approximately half of the CBF AML patients are cured with current therapy, indicating the need for improved therapeutic approaches. This review summarizes the most recent laboratory and clinical discoveries relevant to this subset of AML and how they are being applied for in an effort to improve the cure rate in patients with the disease.

Section snippets

Molecular Basis of CBF AML

CBF is a heterodimeric transcription factor complex composed of alpha and beta subunits that plays a pivotal role in normal hematopoiesis.9 The CBF alpha subunit is encoded by one of three homologous genes of the RUNX (Runt-related transcription factor) family, the RUNX1 (AML1; CBFA2; PEBP2aB) gene, whereas the CBF beta subunit is encoded by the CBFB (PEBP2B) gene.9 The CBF alpha subunit binds directly to the DNA promoter sequences of target genes involved in hematopoiesis, whereas the beta

Demographic, Clinical, and Secondary Cytogenetic Characteristics

Although commonly reported together as CBF AML, patients with t(8;21) differ from those with inv(16) AML with regard to demographic and clinical characteristics. The t(8;21) AML patients were found to be less frequently white and more frequently African American than those with inv(16).29, 30 Diagnostic white blood cell counts29, 30, 31 and the percentage of blasts in bone marrow and blood29 are higher in inv(16) compared with t(8;21) AML. The typical French-American-British (FAB) blast

Treatment and Outcomes

The CBF AMLs are commonly classified within the favorable AML risk group. The complete remission (CR) rates in both t(8,21) and inv(16) AML are high and reach approximately 90% after standard induction therapy containing anthracyclines and cytarabine.29, 31 One study reported that higher bone marrow blasts, lower platelets, and non-white race in t(8;21) AML and lower platelets and hepatomegaly in inv(16) AML adversely affected the probability to achieve CR.29 A different study showed that the

Prognostic Genetic Factors

Although in comparison to other cytogenetic AML groups, CBF AML patients are considered as a more favorable subset, improved therapy is needed as 50% of CBF AML patients still are not cured. Genetic markers may be useful to predict poor outcome in CBF AML patients, thereby providing a means for risk-adapted stratification to more aggressive and/or novel targeted therapies for those patients that are predicted not to be cured by chemotherapy alone.

In t(8;21) AML, an earlier German study

Monitoring of Minimal Residual Disease in CBF AML

The term “minimal residual disease” (MRD) refers to the persistence of leukemic cells not detectable by microscopic examination of bone marrow. The ultimate goal of MRD analysis is to anticipate an impending hematological relapse, which would allow a timely therapeutic intervention. The combination of reverse transcription of RNA with subsequent polymerase chain reaction (RT-PCR) enables the amplification of individual RNA sequences such as RUNX1/RUNX1T1 and CBFB/MYH11. However, the

References (76)

  • C.J. Harrison et al.

    Fluorescence in situ hybridization analysis of masked (8;21)(q22;q22) translocations

    Cancer Genet Cytogenet

    (1999)
  • R. Cairoli et al.

    Prognostic impact of c-KIT mutations in core binding factor leukemiasan Italian retrospective study

    Blood

    (2006)
  • S. Schnittger et al.

    KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival

    Blood

    (2006)
  • A. Shimada et al.

    KIT mutations, and not FLT3 internal tandem duplication, are strongly associated with a poor prognosis in pediatric acute myeloid leukemia with t(8;21): a study of the Japanese Childhood AML Cooperative Study Group

    Blood

    (2006)
  • R. Roskoski

    Signaling by Kit protein-tyrosine kinase—the stem cell factor receptor

    Biochem Biophys Res Commun

    (2005)
  • J. Cammenga et al.

    Extracellular KIT receptor mutants, commonly found in core binding factor AML, are constitutively active and respond to imatinib mesylate

    Blood

    (2005)
  • T.M. Kohl et al.

    KIT exon 8 mutations associated with core-binding factor (CBF)-acute myeloid leukemia (AML) cause hyperactivation of the receptor in response to stem cell factor

    Blood

    (2005)
  • A. Beghini et al.

    STI 571 inhibition effect on KITAsn822Lys-mediated signal transduction cascade

    Exp Hematol

    (2005)
  • J.D. Growney et al.

    Activation mutations of human c-KIT resistant to imatinib mesylate are sensitive to the tyrosine kinase inhibitor PKC412

    Blood

    (2005)
  • K.V. Gleixner et al.

    PKC412 inhibits in vitro growth of neoplastic human mast cells expressing the D816V-mutated variant of KIT: comparison with AMN107, imatinib, and cladribine (2CdA) and evaluation of cooperative drug effects

    Blood

    (2006)
  • T. Haferlach et al.

    Global approach to the diagnosis of leukemia using gene expression profiling

    Blood

    (2005)
  • M.E. Ross et al.

    Gene expression profiling of pediatric acute myelogenous leukemia

    Blood

    (2004)
  • L. Bullinger et al.

    Gene-expression profiling identifies distinct subclasses of core binding factor acute myeloid leukemia

    Blood

    (2007)
  • S. Nishida et al.

    AML1-ETO rapidly induces acute myeloblastic leukemia in cooperation with the Wilms tumor gene, WT1

    Blood

    (2006)
  • T. Yamagami et al.

    Growth inhibition of human leukemic cells by WT1 (Wilms tumor gene) antisense oligodeoxynucleotides: implications for the involvement of WT1 in leukemogenesis

    Blood

    (1996)
  • J.D. Rowley

    Identificaton of a translocation with quinacrine fluorescence in a patient with acute leukemia

    Ann Genet

    (1973)
  • C.D. Bloomfield et al.

    Evaluation of leukemic cell chromosomes as a guide to therapy

    Blood Cells

    (1982)
  • M.M. Le Beau et al.

    Association of an inversion of chromosome 16 with abnormal marrow eosinophils in acute myelomonocytic leukemiaA unique cytogenetic-clinicopathological association

    N Engl J Med

    (1983)
  • P. Liu et al.

    Fusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemia

    Science

    (1993)
  • N.A. Speck et al.

    Core-binding factors in haematopoiesis and leukaemia

    Nat Rev Cancer

    (2002)
  • J.J. van Dongen et al.

    Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual diseaseReport of the BIOMED-1 Concerted Action: investigation of minimal residual disease in acute leukemia

    Leukemia

    (1999)
  • L.H. Castilla et al.

    The fusion gene Cbfb-MYH11 blocks myeloid differentiation and predisposes mice to acute myelomonocytic leukaemia

    Nat Genet

    (1999)
  • Y. Yuan et al.

    AML1-ETO expression is directly involved in the development of acute myeloid leukemia in the presence of additional mutations

    Proc Natl Acad Sci U S A

    (2001)
  • Gilliland DG, Jordan CT, Felix CA. The molecular basis of leukemia. Hematology Am Soc Hematol Educ Program....
  • L.F. Peterson et al.

    The 8;21 translocation in leukemogenesis

    Oncogene

    (2004)
  • T. Yamagata et al.

    Runx1/AML1 in normal and abnormal hematopoiesis

    Int J Hematol

    (2005)
  • V. Gelmetti et al.

    Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO

    Mol Cell Biol

    (1998)
  • B. Lutterbach et al.

    ETO, a target of t(8;21) in acute leukemia, interacts with the N-CoR and mSin3 corepressors

    Mol Cell Biol

    (1998)
  • Cited by (0)

    View full text