Oncogene (2008) 27, 5759–5773
& 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $32.00 www.nature.com/onc
REVIEW
Complementing mutations in core binding factor leukemias: from mouse models to clinical applications AMS Mu¨ller1,2, J Duque1, JA Shizuru2 and M Lu¨bbert1 1 Department of Hematology/Oncology, University Medical Center Freiburg, Freiburg, Germany and 2Stanford University Medical Center, Department of Medicine, Division of Blood and Marrow Transplantation, Stanford, CA, USA
A great proportion of acute myeloid leukemias (AMLs) display cytogenetic abnormalities including chromosomal aberrations and/or submicroscopic mutations. These abnormalities significantly influence the prognosis of the disease. Hence, a thorough genetic work-up is an essential constituent of standard diagnostic procedures. Core binding factor (CBF) leukemias denote AMLs with chromosomal aberrations disrupting one of the CBF transcription factor genes; the most common examples are translocation t(8;21) and inversion inv(16), which result in the generation of the AML1-ETO and CBFb-MYH11 fusion proteins, respectively. However, in murine models, these alterations alone do not suffice to generate fullblown leukemia, but rather, complementary events are required. In fact, a substantial proportion of primary CBF leukemias display additional activating mutations, mostly of the receptor tyrosine kinase (RTK) c-KIT. The awareness of the impact and prognostic relevance of these ‘second hits’ is increasing with a wider range of mutations tested in clinical trials. Furthermore, novel agents targeting RTKs are emanating rapidly and entering therapeutic regimens. Here, we present a concise review on complementing mutations in CBF leukemias including pathophysiology, mouse models, and clinical implications. Oncogene (2008) 27, 5759–5773; doi:10.1038/onc.2008.196; published online 7 July 2008 Keywords: core binding factor leukemias; AML1-ETO; CBFb-MYH11; receptor tyrosine kinase; c-KIT mutation
Introduction Acute myeloid leukemias (AMLs) comprise a heterogeneous group of hematopoietic neoplasias. Approximately 55% of adult patients with de novo AML display recurrent chromosomal aberrations and/or gene rearrangements (Byrd et al., 2002; Mrozek et al., 2004; Marcucci et al., 2005), which often can be correlated with the morphology-based AML subgroups of the Correspondence: Professor Dr M Lu¨bbert, Department of Hematology/ Oncology, University Medical Center Freiburg, Hugstetterstr. 55, Baden Wuerttemberg, Freiburg D-79106, Germany. E-mail:
[email protected] Received 23 July 2007; revised 10 March 2008; accepted 15 May 2008; published online 7 July 2008
French–American–British (FAB) Working Group classification (Bennett et al., 1985). The increasing knowledge on cytogenetic characteristics of acute leukemias and their association to distinct biological and clinical features demand the designation of novel prognostic categories. Moreover, cytogenetic changes provide important insights into the pathogenesis of the disease: although transcription factors are common targets of chromosomal translocations, it has been hypothesized that only the collaboration with additional genetic events, such as mutations of receptor tyrosine kinases (RTKs), provides the selective advantage required for full leukemogenesis (Kelly and Gilliland, 2002). Leukemias with mutations affecting the core binding factor (CBF) transcription factor genes present an ostensive example for this molecular interplay. Chromosomal translocations that disrupt CBF frequently coincide with specific RTK mutations. This observation has attracted a great deal of attention, not only because of a prognostic impact of certain mutations, but also because RTKs are appealing targets for novel therapeutic agents such as small-molecule tyrosine kinase inhibitors (TKIs). Here, we give a synopsis of recent studies on complementing mutations in CBF leukemias. Pathophysiology, mouse models, state of clinical data and trials, and novel therapeutic implications are discussed. Core binding factor leukemia: chromosomal translocations generate fusion genes Genes encoding the subunits of CBF are common targets of chromosomal aberrations, and alterations are present in up to 20% of human AMLs (Slovak et al., 2000) and 22% of pediatric acute lymphoid leukemias (Shurtleff et al., 1995) (Table 1). The most common examples are translocation t(8;21) and inversion inv(16), which result in the AML1-ETO and CBFb-MYH11 (myosin heavy chain protein) fusion proteins, respectively. AML with disruptions of the CBF are collectively referred to as ‘CBF leukemias,’ as they share many clinical features, including a generally favorable prognosis (Byrd et al., 2002). The term CBF comprises a small family of heterodimeric transcription factors, composed of an alpha (a) and a common beta (b) subunit. The CBF transcription factor complex transactivates the expression of a broad
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5760 Table 1 Translocations involving the core binding factor and associated phenotypes Target
Translocation
Fusion gene
Associated phenotype
AML1
t(8;21)(q22;q22) t(12;21)(p13;q22) t(12;21)(p12;q22) t(3;21)(q26;q22)
AML1-ETO TEL-AML1
CBFb
t(16;21)(q24;q22) t(12;21)(q12;q22) t(19;21) (q13;q22) t(X;21)(p22.3;q22.1) t(7;21) (p22;q22)
AML1-EVI1 or EAP, MDS1 AML1-MTG16 AML1-copine VIII AML1-AMP19 AML1-FOG2 AML1-USP42
t(2;21)(q11;q22)
AML1-LAF4
AML M2 Pediatric B-cell ALL Pediatric pre-B-cell ALL MDS, therapy-related myeloid leukemias, CML blast crisis Therapy-related myeloid leukemia Relapsed AML M2 Radiation-associated secondary AML MDS AML-M0 with aberrant expression of T-cell-markers (case report) Pediatric T-cell ALL
inv(16)(p13;q22) t(16;16)(p13;q22)
CBFb-MYH11 CBFb-MYH11
del(16)(q22)
CBFb-MYH11
Frequency 4–12% 16–22%
Reported by
Rare Rare Rare Rare Rare
Miyoshi et al. (1991) Romana et al. (1995) Golub et al. (1995) Nucifora et al. (1993, 1994) Gamou et al. (1998) Ramsey et al. (2003) Hromas et al. (2001) Chan et al. (2005) Paulsson et al. (2006)
Rare
Chinen et al. (2008)
AML M4eo; (AML M5) AML M4eo; (AML M7)
5% Rare
AML M4eo
Rare
Liu et al. (1993) Shurtleff et al. (1995); Costello et al. (1997) Marlton et al. (1995); Costello et al. (1997)
Rare
Abbreviations: ALL, acute lymphoid leukemia; AML, acute myeloid leukemia; CBF, core binding factor; CML, chronic myelogenous leukemia; MDS, myelodysplastic syndrome; MYH, myosin heavy chain protein.
spectrum of genes that are critical for the establishment and development of normal hematopoiesis, such as interleukin-3 (Cameron et al., 1994), granulocytic differentiation factor C/EBPa (Pabst et al., 2001), MCSF receptor (Zhang et al., 1996) and myeloperoxidase (Nuchprayoon et al., 1994). CBFa The a subunit AML1 (also known as RUNX1, PEBP2aB or CBFA2) is the target of a number of translocations both in myeloid and lymphatic leukemias (Table 1). With an occurrence of 4–12% in adult (Look, 1997; Grimwade et al., 1998; Kottaridis et al., 2001; Byrd et al., 2002; Thiede et al., 2002; Mrozek et al., 2004; Schnittger et al., 2006) and 12–30% of pediatric patients (Grimwade et al., 1998; Goemans et al., 2005; Shimada et al., 2006), the translocation t(8;21)(q22;q22) (henceforth referred to as t(8;21)) represents one of the most frequent cytogenetic events in de novo AML. Typically, t(8;21) is associated with the French–American–British AML M2 subtype, of which B30–40% demonstrate this chromosomal abnormality (Erickson et al., 1992; Downing, 1999). AML 1 is a member of the RUNX family of transcription factors, which are characterized by a Runt homology domain at the amino terminus. The Runt homology domain is required for heterodimerization with CBFb and for DNA binding. AML1 activates transcription from enhancer core motifs (TGT/cGGT), which are present in a number of genes relevant to myeloid and lymphoid development (Meyers et al., 1993; Peterson and Zhang, 2004). Its key regulatory role in hematopoiesis has been confirmed in knockout studies, where AML1/ mice displayed a complete lack of definitive hematopoiesis (Okuda et al., 1996; Wang et al., 1996a). ETO (for 8;21; also called MTG8 and CDR) encodes a zinc-finger-containing protein that belongs to a protein Oncogene
family characterized by four evolutionarily conserved Nervy homology regions (Peterson and Zhang, 2004). Besides its molecular role as an inhibitor of C/EBPb and a regulator of early adipogenesis (Rochford et al., 2004), ETO appears to be involved in early gastrointestinal development as shown by ETO knockout experiments in mice (Calabi et al., 2001). The function of ETO in normal physiology has not yet been fully clarified. The t(8;21) generates the AML1-ETO fusion gene by juxtaposing coding sequences of the AML1 gene on chromosome 21 with sequences of the ETO gene on chromosome 8. The translated chimeric protein consists of the NH2-terminal portion of wild-type AML1, fused in-frame to the nearly full-length ETO protein (Miyoshi et al., 1991; Erickson et al., 1992). Although the Runt homology domain at the amino terminus of AML1 is included in the fusion protein, the transactivation domain at the COOH terminus is absent and replaced by ETO (Miyoshi et al., 1991; Erickson et al., 1992; Kitabayashi et al., 1998). The AML1-ETO fusion protein exerts a dominant-negative effect on AML1dependent transcriptional activation. This suppression occurs mostly through interaction of the ETO moiety with the nuclear receptor corepressor N-CoR-mSin3HDAC1 complex that normally recruits histone deacetylase activity. This interaction is believed to result in a lower level of histone acetylation, less accessible chromatin, and thereby repression of the transactivation activity of wild-type AML1 (Gelmetti et al., 1998; Lutterbach et al., 1998; Wang et al., 1998). Furthermore, AML1-ETO alters the expression of a number of specific intranuclear target genes that are normally not regulated by AML1 (Shimada et al., 2000). CBFb Inversion inv(16)(p13q22) (hereafter termed inv(16)) constitutes another common chromosomal aberration. Inv(16) is detectable in up to 12% of AML patients and
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5761
is usually accompanied by monocytic and eosinophilic differentiation, correlating to the French–American– British AML M4Eo subtype (Look, 1997; Gari et al., 1999; Kottaridis et al., 2001; Thiede et al., 2002; Bacher et al., 2006; Schnittger et al., 2006). Inv(16), and less commonly translocation t(16;16), disrupt CBFb and result in the fusion protein CBFb-MYH11. Both inv(16) and t(16;16) may be cryptic or masked by a concomitant deletion del(16)(q22). Although isolated del(16)(q22) is a recurrent abnormality reported in patients with AML M4Eo, the deletion by itself does not create the fusion protein (Mrozek et al., 2001; Merchant et al., 2004). CBFb-MYH11 contains amino acids 1–165 of the CBFb protein, including the heterodimerization domain for the a-subunit, fused to various lengths of the C-terminal a-helical rod domain of the MYH11 (Liu et al., 1993). Physiologically, the b-component increases the transcriptional activity of CBFa by stabilizing the flexible C-terminal loop of the Runt homology domain (Tahirov et al., 2001) and protects it from proteolytic degradation through ubiquitination (Huang et al., 2001). Apparently, CBFb-MYH11 dominantly inhibits CBF in its transactivating function and its capability to regulate gene expression by sequestering the CBFa subunit into cytoplasm (Kanno et al., 1998) and/or into cytoskeletal filaments and aggregates (Adya et al., 1998). The dominant inhibitory effect of fusion proteins targeting CBF is considered a critical step in the development of hematopoietic malignancies. However, there is clear evidence from in vitro and in vivo assays that chromosomal CBF aberrations alone are not sufficient to induce leukemia. Mouse models of CBF leukemia A comprehensive list of mouse models involving CBF is given in Tables 2a and b. Murine embryos lacking either AML1 (AML1/) (Okuda et al., 1996; Wang et al., 1996a) or CBFb (CBFb/) (Sasaki et al., 1996; Wang et al., 1996b; Niki et al., 1997) showed a complete lack of definitive hematopoiesis and died at mid-gestation due to lethal hemorrhage. Mice carrying a monoallelic inactivation of one AML1 allele (AML1 þ /) were phenotypically healthy. Myeloid cell differentiation and proliferation appeared normal in vitro, except for some minor abnormalities in hematopoietic stem cell proliferation (Sun and Downing, 2004). Mouse models of t(8;21) AML Various approaches have been used to create a chimeric murine/human hybrid AML1-ETO gene mimicking t(8;21) in the mouse germline. For example, the human AML1-ETO sequence has been fused in-frame to the murine AML1 exon 4 (Okuda et al., 1998), or exon 5 of the murine AML1 gene (the t(8;21) break point) has been replaced by cDNA containing exon 5 sequences of the human AML1 gene linked to the entire coding region of ETO (Yergeau et al., 1997). With a phenotype similar to that resulting from homozygous disruption of the AML1 or CFBb gene (AML1/ or CBFb/),
knock-in mice heterozygous for the AML1-ETO alleles succumbed to lethal hemorrhage into central nervous sysytem, pericardial sac and soft tissue at embryonic days 11.5–13.5. Fetal livers lacked definitive hematopoiesis, but displayed dysplastic multilineage hematopoietic progenitors with an increased self-renewal capacity in vitro (Okuda et al., 1998). Primitive hematopoiesis in the yolk sac appeared to be unaffected, although in CFU assays these cells gave rise to macrophages only. This selective pattern suggests that AML1-ETO interferes with the differentiation of certain hematopoietic lineages and presumably promotes development down the monocyte/macrophage pathway (Yergeau et al., 1997). To bypass embryonic lethality and define the contribution of AML1-ETO to leukemogenesis in adults, transgenic mice with conditional expression of the fusion protein in hematopoietic stem/progenitor cells or bone marrow (BM) have been generated. Inducible promoter systems, such as the murine mammary tumor virus-tet-controlled transcriptional activator, contain a tetracycline-responsive element that allows the control of the knocked-in AML1-ETO. This approach resulted in a high expression of the fusion gene in BM and peritoneal macrophages. However, despite this, doublepositive adult mice disclosed no leukemic phenotype (Rhoades et al., 2000). Similar results were obtained with the alternative promoter MRP-8, a small calcium-binding protein specifically expressed in myeloid cells of the neutrophil and monocyte lineage. The hMRP8-AML1-ETO transgenic mice were healthy during their lifespans, unless additional mutations were induced (see below) (Yuan et al., 2001). When a loxP-bracketed transcriptional stop cassette was inserted into the AML1-ETO fusion gene, it was transcriptionally silent in its intact state, but could be activated following birth through Cre9-mediated deletion of this stop cassette (Buchholz et al., 2000). The activation of this allele in vivo was insufficient to induce leukemia despite an enhanced replating efficiency of myeloid progenitors in vitro. Again, only the induction of cooperating mutations resulted in the development of an AML (Higuchi et al., 2002). Several models reconstituted lethally irradiated mice with BM or hematopoietic stem/progenitor cells that were retrovirally transduced to express AML1-ETO (for references see Table 2). Some degree of hematopoietic disturbance, manifesting as long-latency myeloproliferative or myelodysplastic syndromes in vivo (Grisolano et al., 2003; Fenske et al., 2004; Nishida et al., 2006) or an increased replating capacity in vitro, was observed. However, despite a high expression of AML1-ETO, none of these models achieved the spontaneous development of leukemia. The idea that AML1-ETO requires secondary mutagenic events to promote AML development was supported by studies treating AML1-ETO-positive mice with N-ethyl-N-nitrosurea (ENU), an efficient DNAalkylating inducer of single base mutations. In contrast to untreated AML1-ETO mice, 30–55% of those additionally receiving ENU developed AML and Oncogene
5762
Oncogene
Table 2 Selected in vivo mouse studies of core binding factor leukemias
Wang et al. (1996a)
Spontaneous clinical phenotype
AML1+/ (D RHD)
AML1+/ F1: phenotype and PB normal AML/ F2: lethal hemorrhage BE12.5 Complete absence of FL-definitive HP AML1/ ES cells: no contribution to HP tissues in chimeric animals AML1/: lethal hemorrhage BE12 Block of definitive HP
AML1+/: k Erythroid and myeloid colonies
Lethal hemorrhage BE13.5 Absence of FL-derived definitive HP Intact YS-derived primitive HP Lethal hemorrhage BE12.5–13.5 Block of FL HP
Dysplastic multilin. FL progenitors m Self-renewal capacity Immortalization into cell lines Progenitor differentiation into monocyte/macrophage pathway
No leukemia in adult AML1-ETO+ mice
AE switch-off in progenitors+ myeloid cells - maturation
AML1+/ (D exon 4)
AML1/ETO (AE): germline/non-conditional knock-in Okuda et al. (1998) Heterozygous mice with murine/human AML1-ETO (AML1+/AE) Yergeau et al. (1997) AML1+/AE heterozygotes (replacement of ML1 ex5 with ex5 fused to ETO?) AML1/ETO: conditional transgenic models Rhoades et al. (2000) MMTV-tTA promoter control of AML1-ETO tetracycline-off system Yuan et al. (2001) hMRP8-promoter - AML1-ETO expression in myeloid lineage Higuchi et al. (2002) Cre/loxP system for expression of murine/human hybrid AML1-ETO AML1/ETO: gene transfer for AE-expression+HCT de Guzman et al. (2002) AML1-ETO+ transduction of HSC (MSCV vector) - HCT Fenske et al. (2004)
Nishida et al. (2006) Grisolano et al. (2003)
Schessl et al. (2005) Schwieger et al. (2002)
[1] AML1-ETO expression in Sca1 locus (targeting HSC compartment): ‘Sca+/AE’ heterozygotes [2] adoptive transfer of Sca+/AE splenocytes [1] AML1-ETO transduction of BM (MSCV vector) - WT/AE+ versus WT1-tg/AE+ BM - HCT AML1-ETO+/ TEL/PDGFRB (TP) transduction of AML1+/+/AML1+/ BM (MSCV vector) - HCT AML1-ETO and/or FLT3 transduction of BM (MSCV vector) - HCT AML1-ETO transduction of ICSBP+/+ and ICSBP/ BM (FMEV vector) - HCT
No leukemia without ENU No significant abnormalities No block in differentiation of HP cells
Phenotype after manipulation/second hit
AML1/YS: no HP colonies AML1/FL: no HP progenitors No monocytic differentiation in AML1/ embryoid bodies
4–7 months after ENU: 55% AML, 45% T-ALL After ENU: 47% of AE+ mice developed AML/granulocytic sarcoma or TLL/ALL
AE+ BM recipients: no leukemia; late MDS TP+ BM recipients: m mature neutrophils AE+ or FLT3+ BM recipients: no leukemia ICSBP+/+AE+ BM recipients: no leukemia k Maturation of lymphoid+myeloid cells k Erythropoiesis m Granulopoiesis, left-shift, blasts
AE+ BM/FL mixed colonies; myeloid progenitors m replating capacity 2 months pTX undifferentiated colonies 10 months pTX: 50 m myeloid colonies Sca+/AE splenocytes: m survival (8 months), IL3 dependent, slow proliferation
29xm of HSC in BM 10 months post-HCT m myeloid+eosinophil progenitors [1] Sca+/AE mice: normal growth+ fertility delayed nonlethal MPD (6 months); 82% (14 months) [2] no leukemia in Sca+/AE recipients WT/AE+ BM recipients: MDS-like abnormal myelopoiesis, no leukemia
In vitro studies
WT1-tg/AE+ BM recipients: rapid AML - after second TX rapid fatal AML
WT1-tg/AE+ BM cells: k myeloid differentiation, m colony formation
TP+/AE+ BM recipients: rapid lethal leukemia AE+/FLT3+ BM recipients leukemia (AML+ALL) after median 233 days ICSBP/AE+ BM recipients: m BM myeloblasts (20–30%) Granulocytic sarcoma Gradual infiltration of several organs
Spleen colony formation: AE+ BM m 3; AE+/FLT3+ m 6.5 ICSBP+/+AE+ BM: m clonogenic myeloid progenitors, k differentiation
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
(a) AML1-ETO models AML1: germline knockout Okuda et al. (1996)
Method and genotype
Table 2 Method and genotype Peterson et al. (2007)
Yan et al. (2004)
Yan et al. (2006)
(b) CBFb-MYH11 models CBFb+/: germline knockout Sasaki et al. (1996)
AML1-ETO transduction (MigR1 vector) of FL cells from p21waf1/ mice - HCT Transduction of BM or FL with full-length AML1-ETO- or truncated AEtr (MigR1 vector) - HCT Transduction of FL cells with full-length AML1-ETO- or truncated AE9a (MigR1 vector) - HCT
Continued
Spontaneous clinical phenotype
/
Full-length AE+ cell recipients: no leukemia
Recipients of AEtr cells - rapid development of leukemia
Full-length AE+ cell recipients: no leukemia
Recipients of—AE9a+ FL: AML AE+/AE9a+ FL: m leukemic potential
CBFb/: lethal hemorrhage E11.5–13.5 Block of definitive HP in FL or YS CBFb+/: normal HP
Niki et al. (1997)
CBFb+/- exon 1 replaced with PGK-neo-cassette
Wang et al. (1996b)
CBFb+/- exon 5 replaced with PGK-neo-cassette
CBFb/: lethal hemorrhage E11.5–13.5 Block of definitive HP in FL; normal YS HP CBFb+/: normal development+HP CBFb/: lethal hemorrhage E11.5–14.5 impaired FL HP; normal YS HP
Generation of CBFb+/CM chimeras from CBFb+/CM heterozygous ES cells
CBFb-MYH11 (CM): conditional transgenic/knock-in models with/without Kogan et al. (1998) [1] hMRP8 promoter-controlled expression of huCM+/ NRAS in myeloid cells [2] HCT of CM+/NRAS+ BM Yan et al. (2002) HCT of WT/CM+ or p16INK4a/ p19ARF(/)/CM+ or E7+/CM+ BM Castilla et al. (2004)
[1] systemic mutagenesis: retrovirus 4070A injection of CBFb+/CM chimeric neonates [2] HCT/second TX of leukemic BM
CBFb+/CM chimeras healthy (by 7 months) CBFb+/CM heterozygotes: Lethal hemorrhage E11.5–13.5 Block of definitive HP CBFb+/CM ES: no contribution to HP tissues CBFb+/CM chimeras: no tumors within 1 year; block of k myeloid+lymphoid lineages HCT [1] CM+ mice: healthy PB normal BM: altered granulopoiesis WT/CM+ BM recipients: low incidence of leukemia; latency >1 year WT control+4070A: no leukemia (1 year)
CBFb/ YS: no definitive HP colonies CBFb+/ YS: abundant colony formation CBFb/ YS/FL: no definitive HP colonies CBFb+/ YS/FL: normal colony formation CBFb/ YS/FL: kk/no colonies CBFb+/ YS/FL: normal colony formation Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
Castilla et al. (1999)
In vitro studies
+
p21waf1 /AE HC recipients: AML (17 weeks) second TX - leukemia within 2 months
CBFb+/- exon 5 replaced with PGK-neo-cassette
CBFb-MYH11 (CM): germline/non-conditional knock-in Castilla et al. (1996) Murine/human hybrid CBFb-MYH11 - CBFb+/CM chimeras and - heterozygous CBFb+/CM mice
Phenotype after manipulation/second hit
CBFb+/CM ES+YS+FL cells: no definitive HP colonies
After ENU: AML in 84% (2–6 months); second TX - acute leukemia (4–12 weeks) [1] CM+/NRAS+ mice: myelodysplasia, dysgranulopoiesis [2] no leukemia Ptx in second recipient
CM+ BM: k neutrophilic differentiation
Recipients of WT/CM+ BM+ENU mALL p16INK4a/p19ARF(/)/CM+ BM: m ALL E7+/CM+ BM: m ALL [1] CBFb+/CM chimeras+4070A - AML 63% [2] second TX - AML (8 weeks)
5763
Oncogene
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
Oncogene
HCT model: Arf+/CM+, Arf+//CM+ or Arf//CM+ BM (MSCV-IRES-GFP retrovirus infected) recipients Moreno-Miralles et al. (2005)
Abbreviations: AE, AML1/ETO; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; AMP, abnormal myeloid progenitors; ARF/Arf, alternative reading frame; tumor suppressor; p19Arf, murine homolog of human p14ARF; BM, bone marrow; CM, CBFb-MYH11; Cre-loxP system, floxed allel is silent in intact state, activation through Cre-mediated deletion of loxP bracketed transcriptional stop cassette; E, embryonic day; ENU, N-ethyl-N-nitrosurea (potent DNA alkylating mutagen); FL, fetal liver; HCT, hematopoietic cell transplantation; HP, hematopoiesis; ICSBP, interferon consensus sequence-binding protein; MPD, myeloproliferative disorder; MSCV, murine stem cell retroviral vector; PGK-neo-cassette, neomycin resistant gene cassette driven by PGK (phosphoglycerol kinase); pIpC, polyinosinic-polycytidylic acid; pTX, post-transplantation; WT, wild type; WT1-tg, Wilms’ tumor gene transgenic (tumor suppressor gene); YS, yolk sac.
[1] Cre/loxP system for CBFb expression [2] repopulation assays of restored CBFb+/CM-BM or -AMP Kuo et al. (2006)
+/CM
Arf//CM+ BM (12 weeks) pTX AML 56% Arf+//CM+ BM (15 weeks) pTX AML 57%
Pagl2-transduced BM/AMP recipients: - AML: penetrance dose-dependent; phenotype similar to spontaneous AML
: PB: k B cells, [1] CBFb k platelets; BM: m AMP; m HSC; AML after repeated induction [2] after second TX AML development dependent on restored CBFb+/CM-BM cell dose Arf+/CM+ BM 6–7 months pTX AML 40%
[1] CBFb+/CM BM: m myeloid+ erythroid colonies; k megakaryocyte maturation; k replating capacity of AMPs
Phenotype after manipulation/second hit Spontaneous clinical phenotype Method and genotype
+/CM
Table 2
Continued
In vitro studies
5764
granulocytic sarcomas mimicking many of the features of human t(8;21) AML (Yuan et al., 2001; Higuchi et al., 2002). These leukemic cells expressed AML1-ETO, grew efficiently in vitro, readily established immortalized immature myeloid cell lines, and were transplantable into secondary recipients (Higuchi et al., 2002). In the same studies, a substantial proportion of animals came down with T-acute lymphoid leukemia/T-cell lymphoblastic lymphomas, but these leukemic cells did not express AML1-ETO (Yuan et al., 2001; Higuchi et al., 2002). Evidently, AML1-ETO transgenic mice develop both myeloid and lymphoid malignancies after treatment with ENU, whereas animals without AML1-ETO expression, including wild-type controls and AML1deficient chimeric mice, only develop lymphoid malignancies (Kundu et al., 2005). These findings imply on the one hand that AML1-ETO, although not solely sufficient, is required for myeloid leukemogenesis, and on the other that development of T-cell neoplasias appears to be independent from the presence of CBF mutations. Advanced CBF leukemia models have combined AML1-ETO expression with defined additional mutational events, such as activating mutations of the RTKs TEL-PDGFRb (Grisolano et al., 2003) or FLT3 (Schessl et al., 2005), overexpression of Wilms’ tumor gene (Nishida et al., 2006), or deficiencies of ICSBP (interferon consensus sequence-binding protein) (Schwieger et al., 2002), or p21/waf1 (Peterson et al., 2007). Coexpression of the TEL-PDGFRbR and AML1-ETO fusion genes in primary hematopoietic cells rapidly induced an acute leukemia resembling AML1ETO-positive AML M2. Blasts transplanted into secondary recipients readily established the same disease (Grisolano et al., 2003). To study the functional correlation between AML1ETO and the FLT3-length mutation, retrovirally transduced hematopoietic cells expressing AML1-ETO and/ or FLT3-length mutation were used for BM transplantation. No disease developed in recipients of BM singly transduced with AML1-ETO or FLT3-length mutation, whereas all recipients of doubly transduced BM succumbed to an aggressive acute leukemia (Schessl et al., 2005). AML1-ETO-transduced BM from transgenic mice that overexpress the Wilms’ tumor gene rapidly induced AML in all recipients. Myeloid differentiation was inhibited at an immature stage, and the in vitro colonyforming ability was higher as compared to AML1-ETOtransduced BM cells from wild-type mice (Nishida et al., 2006). Similarly, collaboration between AML1-ETO and inactivating mutations, such as deficiency of ICSBP, appeared to advance myeloid tumorigenesis. Accumulation of morphologically altered myeloblasts (20–30%) in the BM was observed as well as generation of granulocytic sarcoma-like tumors. Both of these findings resemble the pathognomonic features of t(8;21) AML (Schwieger et al., 2002). Transplanted AML1-ETOtransduced p21/waf1-deficient hematopoietic cells facilitated leukemogenesis in the recipients, which provides another example of the necessity of collaborative
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5765
interplay of AML1-ETO with additional cytogenetic events (Peterson et al., 2007). In contrast to studies of full-length AML1-ETO, no additional cytogenetic events were required for leukemogenesis in recent mouse studies on a truncated AML1-ETO protein: a single-nucleotide insertion mutation in the AML1-ETO DNA sequence was found to result in a truncated form due to a frame shift with introduction of a stop codon downstream of the insertion. The truncated AML1-ETO protein of 575 amino acids lacked a critical domain for NCoR/SMRT and ETO interactions, and, unlike its full-length counterpart of 752 amino acids, resulted in a rapid onset of leukemia in transplant recipients (Yan et al., 2004). More importantly, the coexpression of the fulllength AML1-ETO and its truncated form resulted in a substantially earlier onset of AML and blocked myeloid cell differentiation at a more immature stage (Yan et al., 2006). Mouse models of inv(16) Fewer models disrupting the CBFb subunit exist, but the pathologic findings observed resemble models involving AML1. CBFb-MYH11 germline knock-in strategies with F1 embryos heterozygous for the CBFb-MYH11 allele fail to generate definitive hematopoiesis and die around embryonic day 12.5 owing to central nervous sysytem hemorrhage and a lack of definitive hematopoiesis in the fetal liver. The resemblance to the AML1/- and CBFb/-phenotype suggests that CBFb-MYH11, similar to AML1-ETO, abrogates CBF function (Castilla et al., 1996). Chimeric mice containing only one CBFb-MYH11 allele appeared healthy aside from some alterations in adult multilineage hematopoietic differentiation, and did not develop any spontaneous tumors (Castilla et al., 1999). Similarly, conditional knock-in of CBFb-MYH11 controlled by an hMRP8 promoter element resulted in impaired neutrophil maturation but no overt leukemia (Kogan et al., 1998). In contrast, a mouse model utilizing Cre-mediated expression of CBFb-MYH11 generated an abnormal myeloid progenitor compartment with leukemic predisposition, block of megakaryocytic maturation, and delayed but spontaneous development of AML (after 11–25 weeks). Latency and efficacy correlated with the number of polyinosinicpolycytidylic acid injections given (1–3) as well as the number of transplanted BM cells. This interval implies that the penetrance depends upon the size of the progenitor population at risk to develop full-blown disease after acquiring cooperating random mutations (Kuo et al., 2006). Models combining the expression of the CBFbMYH11 fusion protein with additional genetic events applied ENU mutagenesis (Castilla et al., 1999), retroviral insertional mutagenesis (Castilla et al., 2004), removal of tumor suppressors (Yang et al., 2002) or the coexpression of oncogenes (Kogan et al., 1998; Yang et al., 2002). Only random mutagenesis approaches, in addition to chimeric inv(16), resulted in AML. Mostly,
myelomonocytic leukemias developed after ENU treatment (84%) (Castilla et al., 1999) and after neonatal application of the retrovirus 4070A (63%) (Castilla et al., 2004). Regarding the potential collaboration of several mutagenic events, the disturbance of neutrophilic maturation was more severe in transgenic mice coexpressing CBFb-MYH11 and activated RAS as compared to those without activated RAS, although none of them displayed a full AML phenotype (Kogan et al., 1998). When the CBFb-MYH11 vector was inserted into BM cells lacking the genes for the cell cycle inhibitor p16INK4a and the tumor suppressor p19ARF (alternative reading frame), or alternatively BM cells expressing the HPV16 oncogene E7 (which is known to decrease p53 activity), a higher rate of clonal acute leukemia could be observed, presumably due to cooperation between an accelerated G1 cell cycle phase and CBF mutations. However, penetrance was incomplete. Exposure to ENU further increased the rate of disease, and leukemias were lymphoid rather than myeloid, regardless of whether CBFb-MYH11 was coexpressed with E7, lack of p16INK4a and p19ARF or random mutations. Hypothetically, these specific secondary mutations may have selected for lymphoid leukemias (Yang et al., 2002). A different outcome was observed in another BM transplant model transducing CBFb-MYH11 into either wild-type, p19ARF þ / or p19ARF/ BM. Expression of the fusion gene was sufficient to induce myelomonocytic leukemia even when expressed in wild-type BM, yet removal of a single allele of p19ARF þ / accelerated the disease (Moreno-Miralles et al., 2005). Similarly, the coexpression of Plag1 or Plagl2 with CBFb-MYH11 in BM and transplantation efficiently resulted in AML in 100% of recipients after 3–12 weeks. In vitro, Plag1 and Plagl2 expanded hematopoietic progenitors and increased proliferation by increasing the transition of G1 to the S phase of the cell cycle (Landrette et al., 2005). 2-Hit hypothesis The evidence from mouse models suggests that neither AML1-ETO nor CBFb-MYH11 alone is sufficient to induce or sustain leukemia. This conclusion is supported by the observation that patients with t(8;21) AML often retain low numbers of AML1-ETO-positive cells in BM and peripheral blood persisting during many years of clinical remission (Nucifora et al., 1993). Moreover, in children with t(8;21) AML, clonotypic AML1-ETO sequences were identified in DNA retrospectively extracted from stored neonatal blood samples that preceded the development of AML by 5–10 years (Wiemels et al., 2002). As studies have detected AML1-ETO transcripts in fractions of stem cells, monocytes and B cells, but not in T lymphocytes of remission BM, it appears that the acquisition of the t(8;21) occurs at the level of stem cells or progenitors capable of differentiating into B cells as well as all myeloid lineages (Miyamoto et al., 2000; Passegue et al., 2003). Oncogene
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5766
The so-called ‘2-hit-hypothesis’ proposes that translocations involving the CBF genes constitute a required but not solely sufficient precondition for transformation and that only collaboration with additional oncogenic events can ultimately result in transformation into a leukemic (stem) cell (Miyamoto et al., 2000). Class II mutations are usually chromosomal translocations (such as AML1-ETO, HOX fusion genes and PML-RARA) that cause loss of function of hematopoietic transcription factors. Consequently, they impair hematopoietic differentiation, promote growth arrest and provoke apoptosis (Burel et al., 2001; Elsa¨sser et al., 2003; Li et al., 2006; Lu et al., 2006). Class I mutations often affect and activate genes involved in signal-transduction pathways, particularly RTKs, such as FLT3, c-KIT, N-RAS and K-RAS. Thus, they confer a proliferative and/or survival advantage to hematopoietic progenitors (Alcalay et al., 2001; Kelly and Gilliland, 2002; Passegue et al., 2003). With emerging evidence of the widespread existence of alternatively spliced t(8;21) transcripts, this hypothesis needs to be regarded with restrictions. As mentioned above, mouse models transplanting retrovirally transduced BM revealed that in contrast to full-length AML1-ETO, truncated forms such as AML1-ETO9a do not require additional genetic events to induce spontaneous leukemia. Notably, recent studies were able to detect alternatively spliced transcripts in human leukemic cells and cell lines (Yan et al., 2006). Alternatively spliced transcripts such as AML1-ETO9a also lead to the production of a truncated AML-ETO protein, which is very similar to the variant found in mouse models. Receptor tyrosine kinase mutations in core binding factor leukemias In a considerable proportion of acute leukemias, secondary activating class I mutations have been identified in coexistence with (class II) translocations. Overall, FLT3 represents the single most common mutated gene in AML, and it is affected by length mutations (internal tandem duplications) in the juxtamembrane domain in 24% (Kottaridis et al., 2001) and by activating loop mutations in 7% of cases (Thiede et al., 2002). Although normal karyotype AMLs display FLT3 mutations in 26–39%, their frequency is even higher in those with a PML/RARA fusion (30–43%) (Kottaridis et al., 2001; Stirewalt et al., 2001; Schnittger et al., 2002; Thiede et al., 2002; Kuchenbauer et al., 2005). In contrast, t(8;21) and inv(16) AMLs feature FLT3 mutations at a comparatively low rate (o10%) (for references, see Table 3), which led to the expectation that the prevalence of RTK mutations in CBF leukemias is low. It has only recently become clear, that c-KIT instead of FLT3 represents a common target for class I mutations in CBF leukemias. In fact, c-KIT mutations cluster within CBF AMLs; 9–48% of AML M2 patients with t(8;21) and 10–45% with inv(16) carry Oncogene
activating mutations in the c-KIT receptor, whereas the total incidence of c-KIT mutations in AML in general is B5% (references listed in Table 3). The c-KIT belongs to the type III RTK family, which consists of five immunoglobulin-like repeats in the extracellular domain, a single transmembrane a juxtamembrane and a cytoplasmic kinase domain. The latter comprises the first and second catalytic domains (TK1 and TK2), which carry an adenosine triphosphate (ATP)-binding region, and the kinase-activating loop, respectively. TK1 and TK2 are separated by the kinase insert sequence (Figure 1). Autoinhibitory function to maintain the kinase in an inactive conformation is exerted by the juxtamembrane and the activating loop domain (Roskoski, 2005). In CBF leukemias, c-KIT mutations accumulate within exons 8 and 17 (Figure 1). These exons are uncommon targets in other neoplasias, as mutations typically involve exon 11 or 9. Exon 17 translates into the activating loop of the second TK domain and corresponds to the D835 location of FLT3. In particular, the c-KIT D816 residue of exon 17 represents a mutational ‘hot spot’ harboring B30% of c-KIT mutations (for example, Beghini et al., 2004; Nanri et al., 2005a). Remarkably, in inv(16) AML, exon 17 mutations seem to occur exclusively at codon c-KIT D816. In t(8;21) also mutations at c-KIT N822 have been described (Goemans et al., 2005; Wang et al., 2005; Nanri et al., 2005a; Paschka et al., 2006). Exon 8 encodes the fifth immunoglobulin-like unit and comprises an evolutionarily highly conserved region in the extracellular domain. It is believed to play a role in dimerization. Exon 8 mutations usually result in single amino acid substitutions at codon D419 and cause hyperactivation of the receptor in response to the stem cell factor (Kohl et al., 2005). Exon 8 mutations can be detected in B10% of CBF AML patients. According to some investigators, they appear to be more common in patients with inv(16) than in those with t(8;21), with incidences of 20–25 versus B6%, respectively (Care et al., 2003; Boissel et al., 2006). However, others have reported equal distributions (Cairoli et al., 2006). Determination of the frequency at which association between specific c-KIT mutations and distinct cytogenetic AML subtypes occurs remains hampered by the fact that in most studies, only selected known mutations were probed. A multitude of isolated cases of ‘novel’ cKIT mutations have been described as single cases but have not been systematically evaluated (Gari et al., 1999). As more mutations continue to be discovered, it is likely that the overall incidence of c-KIT mutations may have been underestimated. Other common mutations in CBF leukemias, predominantly in those with inv(16), involve the oncogenes N-RAS (26–38%) (Valk et al., 2004; Bacher et al., 2006) and K-RAS (7–17%) (Valk et al., 2004; Bowen et al., 2005). Moreover, the novel mutation V617F of the Janus kinase JAK2 has been found primarily in therapyrelated or secondary but also in some rare cases of de novo t(8;21) positive leukemias (Do¨hner et al., 2006; Lee et al., 2006; Illmer et al., 2007; Schnittger
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5767 Table 3
RTK and RAS mutations in clinical studies of CBF leukemias
Author
Mutations
in t(8;21) (%)
in inv(16) (%)
Gari et al. (1999) Beghini et al. (2000) Kottaridis et al. (2001) Thiede et al. (2002) Schnittger et al. (2002)
c-KIT c-KIT FLT3 FLT3 FLT3
exon 8 D816 LM LM/TKD LM
1/19 4/9 6/67 3/41 6/69
(5) (44) (9) (7) (9)
7/21 2/6 3/42 7/43 0/47
(33) (33) (7) (16) (0)
Care et al. (2003)
c-KIT
exon 8 D816 LM D835
1/47 5/47 2/47 1/47
(2) (11) (4) (2)
15/63 5/63 2/63 3/63
(24) (8) (3) (5)
FLT3 Cairoli et al. (2003) Beghini et al. (2004)
c-KIT c-KIT
Valk et al. (2004)
c-KIT FLT3 RAS N/KRAS
exon 2/8/10/11/17 exon 8 D816 D835
Wang et al. (2005)
c-KIT
exon 17 KIS exon 11 exon 8
Goemans et al. (2005)
c-KIT
exon 8 exon 17
N/KRAS Schessl et al. (2005)
FLT3 c-KIT NRAS
Nanri et al. (2005a)
c-KIT
LM D835 D816 codon 12/13/61 exon 17 JM exon 8
FLT3 KRAS Nanri et al. (2005b)
c-KIT FLT3
8/12 (67) 15/32 (47)
15/58 4/58 3/58 19/58 15/5/58 21/54 1/54 2/54 2/54
(39) (2) (4) (4)
2/16 (13) 3/16 (19) 4/16 (25) 11/135 3/135 11/135 13/135
(8) (2) (8) (10)
10/37 1/37 3/37 3/37 1/37
(27) (3) (8) (8) (3)
D816/ JM exon 8 D835
NRAS KRAS
Paschka et al. (2006)
c-KIT
exon 17 exon 8
Boissel et al. (2006)
c-KIT
exon 8 D816 LM D835
3/50 3/50 1/56 4/56 2/2/50
8/76 (11) 13/132 (10)
N/KRAS Schnittger et al. (2006) Bacher et al. (2006)
c-KIT NRAS
D816 codon 12/13/61
Kuchenbauer et al. (2006)
c-KIT NRAS FLT3
D816 codon 12/13/61 LM D835
AML1 PU1 Cairoli et al. (2006)
c-KIT
Shimada et al. (2006)
c-KIT FLT3 JAK2
Schnittger et al. (2007a, b)
exon 17 exon 8 exon 11 exon 10 exon 17 LM/D835
(26) (7) (5) (33) (26/9)
0/7
3/11 (27) 3/11 (27) 4/11 (36)
1/15 1/15 1/15 2/15
Bowen et al. (2005)
FLT3
2/6 (33) 9/20 (45)
(7) (7) (7) (13)
5/87 (6) 3/59 (5)
11/60 (18) 7/42 (17)
9/49 (18) 2/49 (4)
10/61 (16) 8/61 (13)
3/24 8/87 2/88 1/21 1/26 1/19
(6) (6) (2) (7) (4/4)
9/46 (20) 1/46 (2) 0/47 3/47 (6) 15/3/47 (32/6) 2/97 (2) 50/133 (38)
(13) (9) (2) (5) (4) (5)
12/42 (29) 5/42 (12) 2/42 (5)
8/25 (32) 3/25 (12) 1/25 (4)
8/46 (17) 2/46 (4) 2/24 (8)
Abbreviations: CBF, core binding factor; LM, length mutation; RTK, receptor tyrosine kinase. Oncogene
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5768 Codon D419
V530I M541L L546L L550L 571
GTA-ATA ATC-CTG AAA-AAG AAA-AAG +14 (ESVDPTQLPYDHKW)
572 Q575 Y578-586
1714-1715 +17 (EFCELPYDHKWEFPRNR)
Extracellular domain
5 immunoglobulin-like repeats
exon 10
cytoplasmic kinase domain
Intracellular domain
Juxtamembrane domain (JM) TK1: 1st catalytic domain: ATP-binding region
exon 11
6 bp insertion IFdel+ins IFdel+ins IFdel+ins
polymorphism silent transition polymorphism 6 bp IFins ITD
exon 17
exon 18
L773S T798I L813 D816 D816 D816 D816 D816 S821G N822K N822K Y823K V825I L862L
reference [4] [4] [4] [1] [1] [1] [1] [1] [1] [1] [2] [2] [2] [2] [2] [2] [3, 5, 7] [3, 7] [7] [7] [7] [7] [7] [7] [7] [3] [3] [3] [3] [3] [5] [1, 2] [4] [1] [4] [5] [4] [5] [2] [5]
I748T
KIS: kinase insert sequence TK2: nd 2 catalytic domain: A-loop
comment
502
exon 8
Transmembrane domain (TM)
nucleotide change 1254-1255 insTTCTTC 1249-1265 delACTTACGAinsCT 1250-1260 delCTTACGACAinsGCGTCATTGTGG delCTTACGAinsT delGACAGGCinsT delACTTACGACinsGTG delCTTACGAinsGAGG delCTTACGACAinsTAC delCTTACGACinsAT delGACAinsTCCTCCGACG delACTTACGinsT delCTTACGAinsAATC delTTAinsATTCCG insTTCTTT delGAC delACTTACGAinsTT delD419 delT417 and D419 delT417_D419delinsI delT417_D419delinsW delT417_D419delinsIP delT417_D419delinsNG delT417_D419delinsRG delT419_D421delinsVHV Y418delinsGFF delTY417_418H delYD418_419S delYD418_419G delTYD417_419RA delTY417_418H 502 (AYFNF) repeat
ACT-ATT L813-A814insSLL D816V D816H D816Y D816I D816F GAC-ATC
polymorphism substitution substitution substitution substitution substitution substitution
CTG-CTC
polymorphism
[5] [1] [7] [3, 5, 7] [4, 5, 7] [3, 4, 5, 7] [7] [6] [7] [3; 4, 5, 7] [7] [3] [5] [1]
References: [1] Gari et al., 1999; [2] Beghini et al., 2004; [3] Goemans et al., 2005; [4] Nanri et al., 2005; [5] Wang et al., 2005; [6] Lasa et al., 2006; [7] Paschka et al., 2006 Abbreviations: bp:basepair; IFdel+ins: in-frame deletion+insertion; ITD: internal tandem duplication
Figure 1 The c-KIT receptor—structure and reported mutations. The human c-KIT gene belongs to the type III receptor tyrosine kinase (RTK) family with five immunoglobulin (Ig)-like repeats in the extracellular (EC) domain, a single transmembrane (TM) domain, a juxtamembrane (JM) domain and a cytoplasmic kinase domain that is split by a kinase insert sequence (KIS) into the adenosine triphosphate (ATP)-binding and phosphotransferase regions. In core binding factor (CBF) acute myeloid leukemia (AML), c-KIT mutations cluster most frequently within exons 17 and 8. Exon 8 mutations are usually represented by a single amino acid substitution at locus D419, which presumably alters conformation of the fourth and fifth Ig-like repeats with consecutive receptor activation or altered ligand affinity. The substitution of a variety of amino acids for the normal c-KIT D816 residue results in kinase activation. Conversely, the deletion of codon 816 severely impairs RTK activity in response to ligand binding. c-KIT D816 mutations are predicted to be in the activating loop (between the ATP binding and substrate phosphotransferase loops).
et al., 2007a, 2007b). In contrast, no expression of the gain-of-function fusion gene FIP1L1/PDGFRa was detectable in 22 patients tested (Monma et al., 2006). Prognostic and therapeutic implications of mutated c-KIT Acute myeloid leukemia patients with CBF leukemia present at a lower median age, have a higher rate of Oncogene
complete remissions, prolonged complete remission duration and a better prognosis as compared to those with a normal karyotype or other chromosomal aberrations (Byrd et al., 2002). However, certain patients appear to suffer a worse outcome, and only B50% are alive at 5 years after diagnosis (Marcucci et al., 2005). Studies that have attempted to identify those at risk suggest that a high white blood cell (WBC) index (expressed as WBC (% BM blasts/100)) at diagnosis is the most important adverse clinical
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5769
indicator. Thrombocytopenia, CD56 expression, extramedullary disease and/or the existence of additional cytogenetic abnormalities also appear to be of prognostic relevance (Baer et al., 1997; Nguyen et al., 2002; Schlenk et al., 2004). Given the heterogeneity of mutations and the lack of comprehensive standardized clinical trials, it is not surprising that the predictive value of (activating) c-KIT mutations in CBF leukemias has not yet been fully clarified. Although available data from previous trials are not completely consistent, they provide evidence that there are correlations between outcome and RTK mutations. These studies report a significantly higher cumulative incidence of relapse (80 versus 13.5%) and a lower 6-year relapse-free survival of 18 versus 60% in patients with c-KIT-mutated as compared to unmutated t(8;21) AML (Nanri et al., 2005a). Similarly, patients with t(8;21), but not those with inv(16), appear to have a shorter event-free survival and relapse-free survival when carrying an additional c-KIT mutation (Boissel et al., 2006). Subgroup analyses have revealed that exon 17 activating-loop mutations, which often present with a high WBC count at diagnosis (Beghini et al., 2004), predict an adverse outcome with regard to event-free survival, overall survival, relapse risk and salvage after relapse in t(8;21) AML (Cairoli et al., 2006; Paschka et al., 2006; Schnittger et al., 2006). In inv(16) AML the prognostic impact of c-KIT mutations remains controversial. Although some studies were unable to establish an association between c-KIT mutations and prognosis in inv(16) AML (Boissel et al., 2006; Cairoli et al., 2006), others found that exon 8 mutations increased the relapse rate but did not affect overall survival (Care et al., 2003). Of note, the adverse effect of mutated c-KIT on the cumulative incidence of relapse in inv(16) AML observed by Paschka et al. was due mainly to the presence of exon 17 mutations (Paschka et al., 2006). These discrepancies indicate that larger trials performing multivariable analyses are warranted to evaluate the predictive value of c-KIT mutations in the context of other prognostic factors and different treatments received. The differentiation of specific c-KIT mutations is particularly crucial with the availability of novel TKIs, which are important emerging treatment options for patients with RTK mutations. Overall, 59% of c-KIT exon 8 (Goemans et al., 2005) and exon 17 c-KIT N822, but not D816, mutations were sensitive to the TKI imatinib in vitro. Despite their resistance to imatinib or the TKI SU5614, c-KIT D816 mutations were reported to be successfully targeted by other TKI compounds
such as PKC412 or dasatinib (Schittenhelm et al., 2006; Schnittger et al., 2006). This difference in the sensitivity of a mutated c-KIT D816 to different PTK inhibitors and their dephosphorylation efficiency quite likely relies on the binding mode of these compounds. Exon 17 c-KIT D816 mutations with autophosphorylation of the activating loop confer constitutive activation of the receptor. Their resistance to imatinib may be due to imatinib binding the TK domain in the inactive state. In contrast, PKC412 is thought to bind within the ATP-binding pocket of the active conformation of the RTK. Hence, assessment of the exact c-KIT mutational status in CBF leukemias may have direct therapeutic consequences and will presumably guide treatment strategies in the future, as patients with unfavorable c-KIT mutation might benefit from early hematopoietic stem cell transplantation or they may be considered for regimens containing novel RTK inhibitors. Conclusions and outlook Over the past years, cytogenetic and molecular studies have substantially changed the classification system of acute leukemias. Morphology of the leukemic cell has become less relevant for clinical treatment decisions, which at the present time largely rely on cytogenetic and molecular features. Besides the well-established metaphase cytogenetics, including the assessment of specific translocations, mutational status has become important, and in a great proportion of leukemias, additional genetic events (point mutations, deletions or insertions) can be detected. These cytogenetic findings increase our understanding of the pathogenesis of acute leukemias and appear to be of major clinical and prognostic value. Furthermore, emerging new therapeutic agents targeting specific (mutated) receptors are opening novel treatment strategies. Therefore, clinical trials that include screening methods for assessment of the mutational status of RTKs in specific subgroups of leukemias are warranted and will provide a better understanding of the pathogenesis and treatment of acute leukemias. Acknowledgements We are grateful for outstanding support by the comments and remarks of both reviewers of this paper. This study was supported by the Deutsche Forschungsgemeinschaft (DFG), Germany (AM), the La Caixa Stiftung–Deutscher Akademischer Austauschdienst (DAAD), Germany (JD), and the German Jose-Carreras Leukemia Foundation (ML), respectively.
References Adya N, Stacy T, Speck NA, Liu PP. (1998). The leukemic protein core binding factor beta (CBFbeta)-smooth-muscle myosin heavy chain sequesters CBFalpha2 into cytoskeletal filaments and aggregates. Mol Cell Biol 18: 7432–7443. Alcalay M, Orleth A, Sebastiani C, Meani N, Chiaradonna F, Casciari C et al. (2001). Common themes in the pathogenesis of acute myeloid leukemia. Oncogene 20: 5680–5694.
Bacher U, Haferlach T, Schoch C, Kern W, Schnittger S. (2006). Implications of NRAS mutations in AML: a study of 2502 patients. Blood 107: 3847–3853. Baer MR, Stewart CC, Lawrence D, Arthur DC, Byrd JC, Davey FR et al. (1997). Expression of the neural cell adhesion molecule CD56 is associated with short remission duration and survival in acute myeloid leukemia with t(8;21)(q22;q22). Blood 190: 1643–1648. Oncogene
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5770 Beghini A, Peterlongo P, Ripamonti CB, Larizza L, Cairoli R, Morra E et al. (2000). C-kit mutations in core binding factor leukemias. Blood 95: 726–727. Beghini A, Ripamonti CB, Cairoli R, Cazzaniga G, Colapietro P, Elice F et al. (2004). KIT activating mutations: incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication. Haematologica 89: 920–925. Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR et al. (1985). Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French– American–British Cooperative Group. Ann Intern Med 103: 620–625. Boissel N, Leroy H, Brethon B, Philippe N, de Botton S, Auvrignon A et al. (2006). Acute Leukemia French Association (ALFA); Leucemies Aigues Myeloblastiques de l’Enfant (LAME) Cooperative Groups. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia 20: 965–970. Bowen DT, Frew ME, Hills R, Gale RE, Wheatley K, Groves MJ et al. (2005). RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood 106: 2113–2119. Buchholz F, Refaeli Y, Trumpp A, Bishop JM. (2000). Inducible chromosomal translocation of AML1 and ETO genes through Cre/ loxP-mediated recombination in the mouse. EMBO Rep 1: 133–139. Burel SA, Harakawa N, Zhou L, Pabst T, Tenen DG, Zhang DE. (2001). Dichotomy of AML1-ETO functions: growth arrest versus block of differentiation. Mol Cell Biol 21: 5577–5590. Byrd JC, Mrozek K, Dodge RK, Carroll AJ, Edwards CG, Arthur DC et al. (2002). Cancer and Leukemia Group B (CALGB 8461). Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood 100: 4325–4336. Cairoli R, Beghini A, Grillo G, Nadali G, Elice F, Ripamonti CB et al. (2006). Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood 107: 3463–3468. Cairoli R, Grillo G, Beghini A, Tedeschi A, Ripamonti CB, Larizza L et al. (2003). C-Kit point mutations in core binding factor leukemias: correlation with white blood cell count and the white blood cell index. Leukemia 17: 471–472. Calabi F, Pannell R, Pavloska G. (2001). Gene targeting reveals a crucial role for MTG8 in the gut. Mol Cell Biol 21: 5658–5666. Cameron S, Taylor DS, TePas EC, Speck NA, Mathey-Prevot B. (1994). Identification of a critical regulatory site in the human interleukin-3 promoter by in vivo footprinting. Blood 83: 2851–2859. Care RS, Valk PJ, Goodeve AC, Abu-Duhier FM, GeertsmaKleinekoort WM, Wilson GA et al. (2003). Incidience and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias. Br J Haematol 121: 775–777. Castilla LH, Garrett L, Adya N, Orlic D, Dutra A, Anderson S et al. (1999). The fusion gene Cbfb-MYH11 blocks myeloid differentiation and predisposes mice to acute myelomonocytic leukaemia. Nat Genet 23: 144–146. Castilla LH, Perrat P, Martinez NJ, Landrette SF, Keys R, Oikemus S et al. (2004). Identification of genes that synergize with CbfbMYH11 in the pathogenesis of acute myeloid leukemia. Proc Natl Acad Sci 101: 4924–4929. Castilla LH, Wijmenga C, Wang Q, Stacy T, Speck NA, Eckhaus M et al. (1996). Failure of embryonic hematopoiesis and lethal hemorrhages in mouse embryos heterozygous for a knocked-in leukemia gene CBFB-MYH11. Cell 87: 687–696. Chan EM, Comer EM, Brown FC, Richkind KE, Holmes ML, Chong BH et al. (2005). AML1-FOG2 fusion protein in myelodysplasia. Blood 105: 4523–4526. Chinen Y, Taki T, Nishida K, Shimizu D, Okuda T, Yoshida N et al. (2008). Identification of the novel AML1 fusion partner gene, LAF4, a fusion partner of MLL, in childhood T-cell acute Oncogene
lymphoblastic leukemia with t(2;21)(q11;q22) by bubble PCR method for cDNA. Oncogene 27: 2249–2256. Costello R, Sainty D, Lecine P, Cusenier A, Mozziconacci MJ, Arnoulet C et al. (1997). Detection of CBFbeta/MYH11 fusion transcripts in acute myeloid leukemia: heterogeneity of cytological and molecular characteristics. Leukemia 11: 644–650. de Guzman CG, Warren AJ, Zhang Z, Gartland L, Erickson P, Drabkin H et al. (2002). Hematopoietic stem cell expansion and distinct myeloid developmental abnormalities in a murine model of the AML1-ETO translocation. Mol Cell Biol 22: 5506–5517. Do¨hner K, Du J, Corbacioglu A, Scholl C, Schlenk RF, Do¨hner H. (2006). JAK2V617F mutations as cooperative genetic lesions in t(8;21)-positive acute myeloid leukemia. Haematologica 91: 1569–1570. Downing JR. (1999). The AML1-ETO chimaeric transcription factor in acute myeloid leukaemia: biology and clinical significance. Br J Haematol 106: 296–308. Elsa¨sser A, Franzen M, Kohlmann A, Weisser M, Schnittger S, Schoch C et al. (2003). The fusion protein AML1-ETO in acute myeloid leukemia with translocation t(8;21) induces c-jun protein expression via the proximal AP-1 site of the c-jun promoter in an indirect, JNK-dependent manner. Oncogene 22: 5646–5657. Erickson P, Gao J, Chang KS, Look T, Whisenant E, Raimondi S et al. (1992). Identification of breakpoints in t(8;21) acute myelogenous leukemia and isolation of a fusion transcript, AML1/ETO, with similarity to Drosophila segmentation gene, runt. Blood 80: 1825–1831. Fenske TS, Pengue G, Mathews V, Hanson PT, Hamm SE, Riaz N et al. (2004). Stem cell expression of the AML1/ETO fusion protein induces a myeloproliferative disorder in mice. Proc Natl Acad Sci USA 101: 15184–15189. Gamou T, Kitamura E, Hosoda F, Shimizu K, Shinohara K, Hayashi Y et al. (1998). The partner gene of AML1 in t(16;21) myeloid malignancies is a novel member of the MTG8(ETO) family. Blood 91: 4028–4037. Gari M, Goodeve A, Wilson G, Winship P, Langabeer S, Linch D et al. (1999). c-kit proto-oncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia. Br J Haematol 105: 894–900. Gelmetti V, Zhang J, Fanelli M, Minucci S, Pelicci PG, Lazar MA. (1998). Aberrant recruitment of the nuclear receptor corepressorhistone deacetylase complex by the acute myeloid leukemia fusion partner ETO. Mol Cell Biol 18: 7185–7191. Goemans BF, Zwaan CM, Miller M, Zimmermann M, Harlow A, Meshinchi S et al. (2005). Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute myeloid leukemia. Leukemia 19: 1536–1542. Golub TR, Barker GF, Bohlander SK, Hiebert SW, Ward DC, Bray-Ward P et al. (1995). Fusion of the TEL gene on 12p13 to the AML1 gene on 21q22 in acute lymphoblastic leukemia. Proc Natl Acad Sci USA 92: 4917–4921. Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, Harrison G et al. (1998). The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children’s Leukaemia Working Parties. Blood 92: 2322–2333. Grisolano JL, O’Neal J, Cain J, Tomasson MH. (2003). An activated receptor tyrosine kinase, TEL/PDGFbetaR, cooperates with AML1/ETO to induce acute myeloid leukemia in mice. Proc Natl Acad Sci USA 100: 9506–9511. Higuchi M, O’Brien D, Kumaravelu P, Lenny N, Yeoh EJ, Downing JR. (2002). Expression of a conditional AML1-ETO oncogene bypasses embryonic lethality and establishes a murine model of human t(8;21) acute myeloid leukemia. Cancer Cell 1: 63–74. Hromas R, Busse T, Carroll A, Mack D, Shopnick R, Zhang DE et al. (2001). Fusion AML1 transcript in a radiation-associated leukemia results in a truncated inhibitory AML1 protein. Blood 97: 2168–2170.
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5771 Huang G, Shigesada K, Ito K, Wee HJ, Yokomizo T, Ito Y. (2001). Dimerization with PEBP2beta protects RUNX1/AML1 from ubiquitin-proteasome-mediated degradation. EMBO J 20: 723–733. Illmer T, Schaich M, Ehninger G, Thiede C, DSIL2003 AML study group. (2007). Tyrosine kinase mutations of JAK2 are rare events in AML but influence prognosis of patients with CBF-leukemias. Haematologica 92: 137–138. Kanno Y, Kanno T, Sakakura C, Bae SC, Ito Y. (1998). Cytoplasmic sequestration of the polyomavirus enhancer binding protein 2 (PEBP2)/core binding factor alpha (CBFalpha) subunit by the leukemia-related PEBP2/CBFbeta-SMMHC fusion protein inhibits PEBP2/CBF-mediated transactivation. Mol Cell Biol 18: 4252–4261. Kelly LM, Gilliland DG. (2002). Genetics of myeloid leukemias. Annu Rev Genomics Hum Genet 3: 179–198. Kitabayashi I, Ida K, Morohoshi F, Yokoyama A, Mitsuhashi N, Shimizu K et al. (1998). The AML1-MTG8 leukemic fusion protein forms a complex with a novel member of the MTG8(ETO/CDR) family, MTGR1. Mol Cell Biol 18: 846–858. Kogan SC, Lagasse E, Atwater S, Bae SC, Weissman I, Ito Y et al. (1998). The PEBP2betaMYH11 fusion created by Inv(16)(p13;q22) in myeloid leukemia impairs neutrophil maturation and contributes to granulocytic dysplasia. Proc Natl Acad Sci USA 95: 11863–11868. Kohl TM, Schnittger S, Ellwart JW, Hiddemann W, Spiekermann K. (2005). 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 105: 3319–3321. Kottaridis PD, Gale RE, Frew ME, Harrison G, Langabeer SE, Belton AA et al. (2001). The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood 98: 1752–1759. Kuchenbauer F, Schnittger S, Look T, Gilliland G, Tenen D, Haferlach T et al. (2006). Identification of additional cytogenetic and molecular genetic abnormalities in acute myeloid leukaemia with t(8;21)/AML1-ETO. Br J Haematol 134: 616–619. Kuchenbauer F, Schoch C, Kern W, Hiddemann W, Haferlach T, Schnittger S. (2005). Impact of FLT3 mutations and promyelocytic leukaemia-breakpoint on clinical characteristics and prognosis in acute promyelocytic leukaemia. Br J Haematol 130: 196–202. Kundu M, Compton S, Garrett-Beal L, Stacy T, Starost MF, Eckhaus M et al. (2005). Runx1 deficiency predisposes mice to T-lymphoblastic lymphoma. Blood 106: 3621–3624. Kuo YH, Landrette SF, Heilman SA, Perrat PN, Garrett L, Liu PP et al. (2006). Cbf beta-SMMHC induces distinct abnormal myeloid progenitors able to develop acute myeloid leukemia. Cancer Cell 9: 57–68. Landrette SF, Kuo YH, Hensen K, Barjesteh van Waalwijk van Doorn-Khosrovani S, Perrat PN, Van de Ven WJ et al. (2005). Plag1 and Plagl2 are oncogenes that induce acute myeloid leukemia in cooperation with Cbfb-MYH11. Blood 105: 2900–2907. Lasa A, Carricondo MT, Carnicer MJ, Perea G, Aventin A, Nomdedeu JF. (2006). A new D816 c-KIT gene mutation in refractory AML1-ETO leukemia. Haematologica 91: 1283–1284. Lee JW, Kim YG, Soung YH, Han KJ, Kim SY, Rhim HS et al. (2006). The JAK2 V617F mutation in de novo acute myelogenous leukemias. Oncogene 25: 1434–1436. Li X, Xu YB, Wang Q, Lu Y, Zheng Y, Wang YC et al. (2006). Leukemogenic AML1-ETO fusion protein upregulates expression of connexin 43: the role in AML 1-ETO-induced growth arrest in leukemic cells. J Cell Physiol 208: 594–601. Liu P, Tarle SA, Hajra A, Claxton DF, Marlton P, Freedman M et al. (1993). Fusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemia. Science 261: 1041–1044. Look AT. (1997). Oncogenic transcription factors in the human acute leukemias. Science 278: 1059–1064. Lu Y, Xu YB, Yuan TT, Song MG, Lu¨bbert M, Fliegauf M et al. (2006). Inducible expression of AML1-ETO fusion protein endows
leukemic cells with susceptibility to extrinsic and intrinsic apoptosis. Leukemia 20: 987–993. Lutterbach B, Westendorf JJ, Linggi B, Patten A, Moniwa M, Davie JR et al. (1998). ETO, a target of t(8;21) in acute leukemia, interacts with the N-CoR and mSin3 corepressors. Mol Cell Biol 18: 7176–7184. Marcucci G, Mrozek K, Ruppert AS, Maharry K, Kolitz JE, Moore JO et al. (2005). Prognostic factors and outcome of core binding factor acute myeloid leukemia patients with t(8;21) differ from those of patients with inv(16): a Cancer and Leukemia Group B study. J Clin Oncol 23: 5705–5717. Marlton P, Claxton DF, Liu P, Estey EH, Beran M, LeBeau M et al. (1995). Molecular characterization of 16p deletions associated with inversion 16 defines the critical fusion for leukemogenesis. Blood 85: 772–779. Merchant SH, Haines S, Hall B, Hozier J, Viswanatha DS. (2004). Fluorescence in situ hybridization identifies cryptic t(16;16) (p13;q22) masked by del(16)(q22) in a case of AML-M4 Eo. J Mol Diagn 6: 271–274. Meyers S, Downing JR, Hiebert SW. (1993). Identification of AML-1 and the (8;21) translocation protein (AML-1/ETO) as sequencespecific DNA-binding proteins: the runt homology domain is required for DNA binding and protein-protein interactions. Mol Cell Biol 13: 6336–6345. Miyamoto T, Weissman IL, Akashi K. (2000). AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci USA 97: 7521–7526. Miyoshi H, Shimizu K, Kozu T, Maseki N, Kaneko Y, Ohki M. (1991). t(8;21) breakpoints on chromosome 21 in acute myeloid leukemia are clustered within a limited region of a single gene, AML1. Proc Natl Acad Sci USA 88: 10431–10434. Monma F, Nishii K, Lorenzo V F, Usui E, Ueda Y, Watanabe Y et al. (2006). Molecular analysis of PDGFRalpha/beta genes in core binding factor leukemia with eosinophilia. Eur J Haematol 76: 18–22. Moreno-Miralles I, Pan L, Keates-Baleeiro J, Durst-Goodwin K, Yang C, Kim HG et al. (2005). The inv(16) cooperates with ARF haploinsufficiency to induce acute myeloid leukemia. J Biol Chem 280: 40097–40103. Mrozek K, Heerema NA, Bloomfield CD. (2004). Cytogenetics in acute leukemia. Blood Rev 18: 115–136. Mrozek K, Prior TW, Edwards C, Marcucci G, Carroll AJ, Snyder PJ et al. (2001). Comparison of cytogenetic and molecular genetic detection of t(8;21) and inv(16) in a prospective series of adults with de novo acute myeloid leukemia: a Cancer and Leukemia Group B Study. J Clin Oncol 19: 2482–2492. Nanri T, Matsuno N, Kawakita T, Suzushima H, Kawano F, Mitsuya H et al. (2005a). Mutations in the receptor tyrosine kinase pathway are associated with clinical outcome in patients with acute myeloblastic leukemia harboring t(8;21)(q22;q22). Leukemia 19: 1361–1366. Nanri T, Matsuno N, Kawakita T, Mitsuya H, Asou N. (2005b). Imatinib mesylate for refractory acute myeloblastic leukemia harboring inv(16) and a c-KIT exon 8 mutation. Leukemia 19: 1673–1675. Nguyen S, Leblanc T, Fenaux P, Witz F, Blaise D, Pigneux A et al. (2002). A white blood cell index as the main prognostic factor in t(8;21) acute myeloid leukemia (AML): a survey of 161 cases from the French AML Intergroup. Blood 99: 3517–3523. Niki M, Okada H, Takano H, Kuno J, Tani K, Hibino H et al. (1997). Hematopoiesis in the fetal liver is impaired by targeted mutagenesis of a gene encoding a non-DNA binding subunit of the transcription factor, polyomavirus enhancer binding protein 2/core binding factor. Proc Natl Acad Sci USA 94: 5697–5702. Nishida S, Hosen N, Shirakata T, Kanato K, Yanagihara M, Nakatsuka S et al. (2006). AML1-ETO rapidly induces acute myeloblastic leukemia in cooperation with the Wilms tumor gene, WT1. Blood 107: 3303–3312. Oncogene
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5772 Nuchprayoon I, Meyers S, Scott LM, Suzow J, Hiebert S, Friedman AD. (1994). PEBP2/CBF, the murine homolog of the human myeloid AML1 and PEBP2 beta/CBF beta proto-oncoproteins, regulates the murine myeloperoxidase and neutrophil elastase genes in immature myeloid cells. Mol Cell Biol 14: 5558–5568. Nucifora G, Begy CR, Kobayashi H, Roulston D, Claxton D, Pedersen-Bjergaard J et al. (1994). Consistent intergenic splicing and production of multiple transcripts between AML1 at 21q22 and unrelated genes at 3q26 in (3;21)(q26;q22) translocations. Proc Natl Acad Sci USA 91: 4004–4008. Nucifora G, Larson RA, Rowley JD. (1993). Persistence of the 8;21 translocation in patients with acute myeloid leukemia type M2 in long-term remission. Blood 82: 712–715. Okuda T, Cai Z, Yang S, Lenny N, Lyu CJ, van Deursen JM et al. (1998). Expression of a knocked-in AML1-ETO leukemia gene inhibits the establishment of normal definitive hematopoiesis and directly generates dysplastic hematopoietic progenitors. Blood 91: 3134–3143. Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. (1996). AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis. Cell 84: 321–330. Pabst T, Mueller BU, Harakawa N, Schoch C, Haferlach T, Behre G et al. (2001). AML1-ETO downregulates the granulocytic differentiation factor C/EBPalpha in t(8;21) myeloid leukemia. Nat Med 7: 444–451. Paschka P, Marcucci G, Ruppert AS, Mrozek K, Chen H, Kittles RA et al. (2006). Cancer and Leukemia Group B. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol 24: 3904–3911. Passegue E, Jamieson CH, Ailles LE, Weissman IL. (2003). Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics? Proc Natl Acad Sci USA 100(Suppl 1): 11842–11849. Paulsson K, Bekassy AN, Olofsson T, Mitelman F, Johansson B, Panagopoulos I. (2006). A novel and cytogenetically cryptic t(7;21)(p22;q22) in acute myeloid leukemia results in fusion of RUNX1 with the ubiquitin-specific protease gene USP42. Leukemia 20: 224–229. Peterson LF, Yan M, Zhang DE. (2007). The p21waf1 pathway is involved in blocking leukemogenesis by the t(8;21) fusion protein AML1-ETO. Blood 109: 4392–4398. Peterson LF, Zhang DE. (2004). The 8;21 translocation in leukemogenesis. Oncogene 23: 4255–4262. Ramsey H, Zhang DE, Richkind K, Burcoglu-O’Ral A, Hromas R. (2003). Fusion of AML1/Runx1 to copine VIII, a novel member of the copine family, in an aggressive acute myelogenous leukemia with t(12;21) translocation. Leukemia 17: 1665–1666. Rhoades KL, Hetherington CJ, Harakawa N, Yergeau DA, Zhou L, Liu LQ et al. (2000). Analysis of the role of AML1-ETO in leukemogenesis, using an inducible transgenic mouse model. Blood 96: 2108–2115. Rochford JJ, Semple RK, Laudes M, Boyle KB, Christodoulides C, Mulligan C et al. (2004). ETO/MTG8 is an inhibitor of C/EBPbeta activity and a regulator of early adipogenesis. Mol Cell Biol 24: 9863–9872. Romana SP, Poirel H, Leconiat M, Flexor MA, Mauchauffe M, Jonveaux P et al. (1995). High frequency of t(12;21) in childhood B-lineage acute lymphoblastic leukemia. Blood 86: 4263–4269. Roskoski Jr R. (2005). Structure and regulation of Kit protein-tyrosine kinase—the stem cell factor receptor. Biochem Biophys Res Commun 338: 1307–1315. Sasaki K, Yagi H, Bronson RT, Tominaga K, Matsunashi T, Deguchi K et al. (1996). Absence of fetal liver hematopoiesis in mice deficient in transcriptional coactivator core binding factor beta. Proc Natl Acad Sci USA 93: 12359–12363. Schessl C, Rawat VP, Cusan M, Deshpande A, Kohl TM, Rosten PM et al. (2005). The AML1-ETO fusion gene and the FLT3 length Oncogene
mutation collaborate in inducing acute leukemia in mice. J Clin Invest 115: 2159–2168. Schittenhelm MM, Shiraga S, Schroeder A, Corbin AS, Griffith D, Lee FY et al. (2006). Dasatinib (BMS-354825), a dual SRC/ABL kinase inhibitor, inhibits the kinase activity of wild-type, juxtamembrane, and activation loop mutant KIT isoforms associated with human malignancies. Cancer Res 66: 473–481. Schlenk RF, Benner A, Krauter J, Buchner T, Sauerland C, Ehninger G et al. (2004). Individual patient data-based meta-analysis of patients aged 16 to 60 years with core binding factor acute myeloid leukemia: a survey of the German Acute Myeloid Leukemia Intergroup. J Clin Oncol 22: 3741–3750. Schnittger S, Bacher U, Kern W, Haferlach C, Haferlach T. (2007a). JAK2 seems to be a typical cooperating mutation in therapy-related t(8;21)/AML1-ETO-positive AML. Leukemia 21: 183–184. Schnittger S, Bacher U, Kern W, Haferlach T, Haferlach C. (2007b). JAK2V617F as progression marker in CMPD and as cooperative mutation in AML with trisomy 8 and t(8;21): a comparative study on 1103 CMPD and 269 AML cases. Leukemia 21: 1843–1845. Schnittger S, Kohl TM, Haferlach T, Kern W, Hiddemann W, Spiekermann K et al. (2006). KIT-D816 mutations in AML1-ETOpositive AML are associated with impaired event-free and overall survival. Blood 107: 1791–1799. Schnittger S, Schoch C, Dugas M, Kern W, Staib P, Wuchter C et al. (2002). Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood 100: 59–66. Schwieger M, Lohler J, Friel J, Scheller M, Horak I, Stocking C. (2002). AML1-ETO inhibits maturation of multiple lymphohematopoietic lineages and induces myeloblast transformation in synergy with ICSBP deficiency. J Exp Med 196: 1227–1240. Shimada A, Taki T, Tabuchi K, Tawa A, Horibe K, Tsuchida M et al. (2006). 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 107: 1806–1809. Shimada H, Ichikawa H, Nakamura S, Katsu R, Iwasa M, Kitabayashi I et al. (2000). Analysis of genes under the downstream control of the t(8;21) fusion protein AML1-MTG8: overexpression of the TIS11b (ERF-1, cMG1) gene induces myeloid cell proliferation in response to G-CSF. Blood 96: 655–663. Shurtleff SA, Buijs A, Behm FG, Rubnitz JE, Raimondi SC, Hancock ML et al. (1995). TEL/AML1 fusion resulting from a cryptic t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup of patients with an excellent prognosis. Leukemia 9: 1985–1989. Shurtleff SA, Meyers S, Hiebert SW, Raimondi SC, Head DR, Willman CL et al. (1995). Heterogeneity in CBF beta/MYH11 fusion messages encoded by the inv(16)(p13q22) and the t(16;16) (p13;q22) in acute myelogenous leukemia. Blood 85: 3695–3703. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A et al. (2000). Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood 96: 4075–4083. Stirewalt DL, Kopecky KJ, Meshinchi S, Appelbaum FR, Slovak ML, Willman CL et al. (2001). FLT3, RAS, and TP53 mutations in elderly patients with acute myeloid leukemia. Blood 97: 3589–3595. Sun W, Downing JR. (2004). Haploinsufficiency of AML1 results in a decrease in the number of LTR-HSCs while simultaneously inducing an increase in more mature progenitors. Blood 104: 3565–3572. Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K et al. (2001). Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain and its allosteric control by CBFbeta. Cell 104: 755–767. Thiede C, Steudel C, Mohr B, Schaich M, Schakel U, Platzbecker U et al. (2002). Analysis of FLT3-activating mutations in 979 patients
Complementing mutations in core binding factor leukemias AMS Mu¨ller et al
5773 with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 99: 4326–4335. Valk PJ, Bowen DT, Frew ME, Goodeve AC, Lowenberg B, Reilly JT. (2004). Second hit mutations in the RTK/RAS signaling pathway in acute myeloid leukemia with inv(16). Haematologica 89: 106. Wang J, Hoshino T, Redner RL, Kajigaya S, Liu JM. (1998). ETO, fusion partner in t(8;21) acute myeloid leukemia, represses transcription by interaction with the human N-CoR/mSin3/ HDAC1 complex. Proc Natl Acad Sci USA 95: 10860–10865. Wang Q, Stacy T, Binder M, Marin-Padilla M, Sharpe AH, Speck NA. (1996a). Disruption of the Cbfa2 gene causes necrosis and hemorrhaging in the central nervous system and blocks definitive hematopoiesis. Proc Natl Acad Sci USA 93: 3444–3449. Wang Q, Stacy T, Miller JD, Lewis AF, Gu TL, Huang X et al. (1996b). The CBFbeta subunit is essential for CBFalpha2 (AML1) function in vivo. Cell 87: 697–708. Wang YY, Zhou GB, Yin T, Chen B, Shi JY, Liang WX et al. (2005). AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia: implication in stepwise leukemogenesis and response to Gleevec. Proc Natl Acad Sci USA 102: 1104–1109. Wiemels JL, Xiao Z, Buffler PA, Maia AT, Ma X, Dicks BM et al. (2002). In utero origin of t(8;21) AML1-ETO translocations in childhood acute myeloid leukemia. Blood 99: 3801–3805.
Yan M, Burel SA, Peterson LF, Kanbe E, Iwasaki H, Boyapati A et al. (2004). Deletion of an AML1-ETO C-terminal NcoR/SMRTinteracting region strongly induces leukemia development. Proc Natl Acad Sci USA 101: 17186–17191. Yan M, Kanbe E, Peterson LF, Boyapati A, Miao Y, Wang Y et al. (2006). A previously unidentified alternatively spliced isoform of t(8;21) transcript promotes leukemogenesis. Nat Med 12: 945–949. Yang Y, Wang W, Cleaves R, Zahurak M, Cheng L, Civin CI et al. (2002). Acceleration of G(1) cooperates with core binding factor beta-smooth muscle myosin heavy chain to induce acute leukemia in mice. Cancer Res 62: 2232–2235. Yergeau DA, Hetherington CJ, Wang Q, Zhang P, Sharpe AH, Binder M et al. (1997). Embryonic lethality and impairment of haematopoiesis in mice heterozygous for an AML1-ETO fusion gene. Nat Genet 15: 303–306. Yuan Y, Zhou L, Miyamoto T, Iwasaki H, Harakawa N, Hetherington CJ et al. (2001). AML1-ETO expression is directly involved in the development of acute myeloid leukemia in the presence of additional mutations. Proc Natl Acad Sci USA 98: 10398–10403. Zhang DE, Hetherington CJ, Meyers S, Rhoades KL, Larson CJ, Chen HM et al. (1996). CCAAT enhancer-binding protein (C/EBP) and AML1 (CBF alpha2) synergistically activate the macrophage colony-stimulating factor receptor promoter. Mol Cell Biol 16: 1231–1240.
Oncogene