Skip to main content
American Journal of Blood Research logoLink to American Journal of Blood Research
. 2021 Feb 15;11(1):22–43.

An updated account on molecular heterogeneity of acute leukemia

Ekta Rahul 1, Harsh Goel 1, Anita Chopra 1, Amar Ranjan 1, Aditya Kumar Gupta 2, Jagdish Prasad Meena 2, Sameer Bakhshi 3, Aroonima Misra 4, Showket Hussain 5, Ganesh Kumar Viswanathan 6, Goura Kishor Rath 7, Pranay Tanwar 1
PMCID: PMC8010602  PMID: 33796387

Abstract

The progress in the field of personalized therapy has been the backbone for the improved mortality and morbidity figure in cancer especially with reference to acute leukemia. The same has been supported by evolving research and development in the field of genomics. The newer discoveries of mutations and the account of already discovered mutations have been playing a pivotal role to refine management strategy. Here, in this review, we are giving an account of relevant mutations and their potential role in the pathogenesis of acute leukemia. The article discusses the old and newly discovered mutations in acute myeloid/lymphoblastic leukemia. The various pathways and cross-talks between the mutations have been briefly described to develop insight towards their contributory and consequent role in the neoplastic process. The article is to sensitize the students, clinicians, and researchers towards the recent updates and development in genomics of acute leukemia.

Keywords: AML, ALL, mutations, genomics

Introduction

Acute leukemias (AL) is clinical outcomes of clonal proliferation of myeloid and lymphoid progenitor cells [1]. In present scenario, most of the leukemias respond with the current chemotherapy. However, refractory disease and multiple relapses are also the major cause of poor prognosis [2]. Earlier, the diagnosis of acute leukemias were based mainly on the clinical & morphological characteristics of myeloid and lymphoid cells which has now changed mainly on the basis of molecular genetics [3]. The molecular genetics have become important in predicting clinical course, but a large cohort is necessary for prognostic validation [4]. The prognostic relevance was first attempted by Nowell & Hungerford in the year of 1960. They discovered a recurrent chromosomal translocation t(9;22)(q34;q11.2) in patients with Chronic Myeloid Leukemia (CML) [5]. This was the first paradigm, where molecular knowledge of specific molecular inhibitor, Imatinib was used for treatment [6]. Now, with the use of advanced high-quality genomic material and high throughput molecular techniques like next generation sequencing (NGS) helped us in characterizing acute myeloid and lymphoid malignancies into various genomic subtypes, which were illustrated by WHO and compiled in 2016 [7]. The aim of this study is to discuss how alterations of the genes that are involved in the various normal cell cycle pathways which includes cell addition, cell death, cell cycle differentiation, metabolism, motility signalling, transcription and transporters ultimately lead to leukemogenesis [8].

Molecular genetics of acute myeloid leukaemia

Acute myeloid leukaemia (AML) is neoplastic proliferation of myeloid precursors. The diagnosis has gone beyond the boundaries of morphology. Now, AML is categorise in heterogeneous groups on the basis of cytogenetic and molecular mutations [9,10]. Some of these recurrent genetic mutations are associated with specific prognosis or management strategies [11]. The most common genes involved for the development of AML are those that encode for transcription factors, epigenetic pathways modifiers, Hox pathways modifiers, cell kinases and nuclear pore proteins (Figure 1).

Figure 1.

Figure 1

Genetic mutation landscape in AML; Mutations involved at the level of transcription factors are t(8;21) [RUNX1-RUNX1T1], t(15;17) [PML-RARA] and inv.16 [CBFB-MYH11], CEBPA. Mutations at the level of epigenetic modifiers are t(9;11) [MLLT3-MLL], IDH1/2, TET2 and DNMT3A. Mutations at the level of kinases are FLT3 [ITD/TKD] mutations and NPM1 mutations.

Transcription factors

Promyelocytic leukemia-retinoic acid receptor-alpha

Promyelocytic leukemia Retinoic Acid receptor Alpha fusion (PML-RARA) results from t(15,17). It has specific morphology (Hypergranular blasts with frequent Auer rods or microgranular variants with dumbbell shaped nucleus). It can be treated with Retinoic Acid. PML/RARA gene has 5’ end formed by PML and 3’ end formed by RARA gene. C-terminal of fusion protein has a ligand binding, dimerization and repression functions. Wild type RARA is a steroid receptor present at the nucleus that acts as a transcription factor and binds to the promoter of genes important for myeloid differentiation. RARA acts as a transcription repressor until it binds to its ligand [12]. In resting state, it binds with Retinoid X receptor protein and forms a hetero-dimer & acts as repressor along with other repressors like SMRT, NCOR, mSin3A & HDACs. Its target genes like GCSF, GCSFR, neutrophil protein; cell surface receptor (CD116, CD18) & regulators of BCL2 [13]. Ligand binding causes recruitment of co activator complex (SRC-1) [14]. PML acts as tumour suppressor protein [13]. RARA (wild-type) influences other genes by binding with retinoic acid response elements (RARE). Fusion protein is not able to convert itself into a transcription activator under physiological level (10-8 M) but can function normally at pharmacological level (10-6 M) [14,15]. Another variant is t(11;17)(ZBTB16/RARA) which does not act as transcription activator even at pharmacological levels. But they are treated with inhibitors of co-repressors of RARA (eg. Inhibitor of HDAC i.e. Trichostatin-A (TSA)) [15,16].

RUNX1-RUNX1T1/(RUNX1-ETO)

Chromosome 19 when positive in t(8,21) is associated with good prognosis but may cause bad prognosis when co-expressed with CD34. When AML with maturation (AML-M2) shows t(8,21), they are positive for CD19 & CD56. RUNX-RUNX1T1 fusion gene is present on Chr-8 [17]. Core binding factor translocation seen in 15% of all AML [18]. RUNX-1 is a part of the core binding factor which binds to a core enhancer sequence of moloney murine leukemia virus as described by Golemis et al. [19]. Core Binding Factor (CBF) has a non-DNA binding CBFβ unit associated with inv.16 in AML [20]. Fusion partner of RUNX-1 is RUNX1T1. Another fusion partner is CBFA2T3 {t(16;21)} [21]. N-terminal of RUNX contains the Runt homology domain (RHD) (DNA binding domain). This domain is mutated in AML and familial platelet disorders [22,23]. The non-DNA binding CBFB of CBF interacts with the N terminal of RUNX-1 which will change the conformation of RUNX-1 to increase DNA binding affinity [24]. C-terminal of the RHD are potential MAP kinase phosphorylation sites [25]. C-terminal’s other weak activation domains are, nuclear matrix target signal, dimerization domain and sequences that are recognized by co-repressor proteins.

RUNX-1 is essential in hematopoietic development. It regulates B-cell tyrosine kinase, TCR α & β, CK (1L-3, GMCSF) & granulocyte proteins (e.g. MPO & elastase) [26-29]. RUNX-1T1 acts as a co-repressor associated with NCOR, mSin3A which is Class I HDAC fusion protein has a DNA binding domain but C terminals are absent [25]. Thus, act as a negative regulator of wild type RUNX-1 [30]. The fusion protein acts as a transcriptional repressor. But, its action depends upon its association with HDACs. HDACs inhibitor such as Trichostatin A (TSA) inhibits the effect of RUNX-1 RUNX1T1 [31]. It also inhibits cell cycle progression of myeloid progenitor cells. The homozygous lack of RUNX1 is lethal, it leads to death of embryos due to haemorrhagic necrosis of central nervous system and abnormal haematopoiesis [32]. In a study done by Gaidzik et al. RUNX1 mutations were associated with resistance to routine chemotherapy in about 30% cases with low survival outcomes [33]. In these patients allogeneic SCT was considered as the best option for improving survival rates [33].

Inversion 16 (CBFB-MYH11)

Inversion 16 with co-expressed CD34 is present in 8% of AML cases. It involves CBF-B & responsible for differentiation of AML M4 with Eosinophilia in morphology (myelo-monocytic leukaemia with eosinophils that has dark purple as well as orange granules) [34]. This inversion leads to fusion of CBFB with C terminal of smooth muscle myosin heavy chain protein gene (MYH11) [20]. Fusion protein acts as a co-repressor associated with mSin3A & HDAC8. This fusion protein needs RUNX-1 for its action [35]. CBF translocation in inv.16 & RUNX-1-RUNX1T1 translocation is not sufficient to produce leukaemia. Multiple genetic hits are necessary for development of AML [36]. They are usually associated with good prognosis [37] and have excellent response when Gemtuzumab ozogamicin is added to normal induction chemotherapy used for other AML [38].

CCAAT/Enhancer binding protein α (CEBPA)

CEBPA is a transcription factor that regulates granulocytic differentiation [39]. Cytogenetically silent mutations of CEBPA are seen in 10% of AML [40]. Other genetic mutations in AML often down regulate CEBPA [41]. RUNX1-RUNX1T1 represses CEBPA promoters. FLT3-lTD activation leads to reduced activity of CEBPA [41,42]. Granulocytic maturation is lost in CEBPA knockout mouse [43].

CEBPA produces two proteins, first large protein (42-kD) which has N terminal that has a trans-activating domain and a C terminal that has b-ZIP domain that mediates dimerization. A second smaller protein (30-kD) whose transactivating domain is absent but has b-ZIP domain. Two types of mutations are seen in CEBPA [44-46]. N terminal truncation mutation leads to increased production of 30-kD protein and a C terminal ZIP domain mutation that inhibits dimerization & DNA binding. N terminal mutation leads to dimerization with long form but inhibits trans-activation by the dimer. All these mutations of CEBPA falls in intermediate risk AML [46]. CEBPA mutation at both alleles is associated with better survival than mutation at single allele [47]. Patients with CEBPA biallelic mutation concurrent with epigenetic modifiers such as TET2 or RNA-splicing genes appear to have a significantly worse prognosis, similar to that seen in the mono-allelic group (Konstandin et al., 2018) [48].

GATA-1

A Zinc finger transcription factor-regulates erythrocyte & megakaryocytic differentiation. Mutations of GATA1 are always present in acute megakaryoblastic leukaemia (AML-M7) that occurs in children with Down Syndrome (DS) [49-51]. Missense mutations in GATA-1 results in syndrome of dys-erythropoietic anaemia & thrombocytopenia. Approximately 10% of DS have GATA-1 mutation patient develops transient myeloproliferative disorder (TMD) in neonatal period [49,52]. A one/third of patients with TMD develops AML-M7 within 5 years. AML-M-7 in DS is sensitive to cytosine arabinosite or anthracycline based chemotherapy with evidence free survival (EFS) rate of 80-100% [53].

Epigenetic factors modifying chromatin & DNA

IDH1/2 and TET2 mutations

IDH1/2 and TET2 mutations are found in 16% of AML with normal karyotype [54]. IDH1/2 mutations are mutually exclusive with mutation in TET-2 in de-novo AML [55]. Wild type IDH1/2 catalyses production of αKG but mutant form produces 2-hydroxyglutarate (2HG). αKG dependent enzymes e.g. Histone demethylase & TET-1/Q are inhibited by 2HG [56]. TET protein is responsible for demethylation of cytosine (5 mc to 5 hmc) [57]. Therefore, mutation in both IDH and TET leads to increased DNA methylation [55-57]. Increased DNA methylation leads to inhibition of myeloid differentiation & promotes development of AML. Drugs mimicking αKG analogues can be used for treatment [56]. Mutations in IDH1 and TET2 have not been shown to have a significant impact on survival [47]. In a study by Feng et al. of 8121 patients it was seen that patients with IDH1 mutation had inferior overall survival (OS), and some patients also had resistance to induction chemotherapy resulting in a lower complete response (CR) rate [58]. Many IDH inhibitors are in preclinical trials. According to a study by Stein et al., the effect of Enasidenib, a IDH1 inhibitor induced hematologic response in patients with refractory AML [59]. In August 2017 FDA approved the use of enasidenib for IDH2 mutated relapsed/refractory AMLs [60].

DNMT3A

It is a methyltranferase family member that catalyses addition of methyl group to cytosine in CpG dinucleotide. Increased methylation of CpG Island results in silencing of many genes [61]. Ley TJ et al. have observed occurance of DNMT3A mutations in 12 to 22% of adult AML patients [62] associated with higher WBC count and old age. Higher frequency has been reported in cases with normal cytogenetics. Role of these mutations as an early leukemic event has also been recognized [63,64]. In meta-analysis conducted by Tie R et al. and Yuan XQ et al., DNMT3A mutations are found to be associated with poor prognosis in de novo adult patients [65,66]. But it is observed that cases with DNMT3A mutations show higher responsiveness and overall survival with use of DNA methyltransferase inhibitor decitabine [67,68].

Mixed lineage leukemia (MLL); 11q23 translocation

MLL is the mammalian homolog of trithorax, a drosophila transcriptional regulator that positively regulates homeobox genes [69]. Homeobox genes are a large family of genes which are developmental regulators essential for growth and differentiation of haematopoietic cells [70]. Wild-type MLL regulates HOX gene expression by methylation of histone H3 lysine (H3K4) [71]. MLL rearrangements involve approximately 10% of chromosomal rearrangements overall in patients with ALL, AML, and MDS [72]. In paediatric and adult ALL, the most common translocation partners are the AFF1 gene (previously known as AF4) at 4q21.3 in t(4;11). However, in AML the most common partner is MLLT3 at 9p23 in t(9;11) [73]. It is most commonly seen in infants, associated with therapy related acute leukemia and associated with poor prognosis [74-76].

Kinases

FLT3 mutation

It encodes a type-III receptor tyrosine kinase. Its ligand is a type I transmembrane protein expressed on hematopoietic and other cells of bone marrow. It normally stimulates growth of myeloid & stem cells [77]. When it binds with ligand FLT3 undergoes dimerization and phosphorylation & leads to activation of signal transduction pathways i.e. PI3K/AKT, MAPK/ERK & STAT pathways [78,79]. Two types of FLT3 mutations are seen, most common is Internal Tandem repeats (ITD) mutations which leads to duplication of juxta-membrane (JM) portion of molecule and it has inhibitory function; when receptor binds with its ligand the inhibitory function is lost [80]. Another less common mutation is Asp825Tyr which leads to activation of FLT3 [81]. The prognostic value of FLT3-TKD is still controversial [82]. FLT3-ITD is detected in most patients with APML (also associated with hyperleukocytosis). FLT3 causes proliferation signals in AML by concomitant balanced translocation or other genetic defects which blocks differentiation is necessary for development of leukemia [81]. It is associated with poor prognosis [83]. In a study by Stirewalt et al. with 140 AML patients aged >55 years the incidence of FLT3-ITD was similar to that of younger patients, and was associated to disease resistance but did not have impact on OS [84]. In a study by Schlenk et al. patients with a high allelic ratio (≥ 0.51) were responded better with allogeneic stem cell transplantation [85] and may be treated with specific FLT-Tyrosine kinase inhibitors [86,87]. Sorafenib and Midostaurin are examples of first generation FLT3 inhibitors, among all the first generation Midostaurin is the only FLT3 inhibitor approved in the US for the treatment of adult, FLT3 mutated AMLs in combination with standard chemotherapy [88].

Nuclear pore protein-nucleophosmin (NPM)

It is a molecular chaperone that shuttles between cytoplasm and nucleus with particular nucleolar concentration [89]. There are several functions of Nucleophosmin [90] like, transport of Pre-ribosomal particles from nucleus to cytoplasm, regulation of centrosome duplication, regulation of P53 and stabilization of P19ARF. NPM1 mutation is expressed in leukemic blasts at relapse [91]. Mutation leads to unfolding of C terminal domain and disruption of binding to nucleolus [91,92]. It leads to abnormal sub-cellular localization of NPM which is normally nucleolar in location, comes to cytoplasm. Homozygous mutation is lethal at embryonic stage [93]. Mutant NPM-1 destabilises tumour suppressor gene protein P14ARF which regulates P53 response. P14ARF is normally localized to nucleolus and escapes cytoplasmic degradation by interaction with NPM [91]. When associated with poor prognostic mutation like FLT3 then it improves the response [83]. According to Angenendt et al. abnormalities in karyotype may nullify the effect of NPM1 mutation on patient outcome [94]. Many drugs have been tried which have their effect inhibiting the action of mutant NPM1 for example NSC34884 inhibits the action of hydrophobic region of NPM1 required for oligomerization resulting in apoptosis of leukemic cells [95].

Novel genetic mutations in AML

KIT

The stem cell factor receptor (c-kit, CD117) is part of type III receptor TK family. CD-117 is expressed in 70% of CD34 positive cells and also on megakaryocytes. KIT mutations are found in 5% of adult AML cases and 16-46% in CBF subgroup [96,97]. Point mutation at exon 17 with a gain of function is most common [98]. Prognostic impact of KIT mutations has been studied by Chen et al. in a meta-analysis of 2933 patients which found that KIT mutations adversely affects relapse risk but not CR rate and OS [99]. Screening for KIT mutations might also be useful for targeted therapy using TKIs.

RAS

NRAS mutations are observed in 8-13% of AML patients while KRAS mutations are observed in 2% of such patients. Common sites of mutations are codons 12, 13 or 61. Large scale studies did not find any negative impact of RAS mutations on outcome [100,101].

Tp53

Tumour suppressor gene Tp53 promotes cell cycle arrest, apoptosis and DNA repair [102] and is mutated in 8-14% of all AML cases. Alterations are more frequent (69-73%) in patients with complex karyotypes [103]. Tp53 mutations independently indicate poorer prognosis, higher relapse rate and inferior event free survival (EFS) and OS [104-106].

WT1

WT1 gene encodes for a transcription factor involved in cell growth and metabolism [107]. WT1 expression in AML patients is commonly detected and is associated with poor prognosis, resistance to therapy, higher relapse and poor survival rates [108]. Recurrent somatic loss of function mutations is also described in 6-13% AML patients. They include point mutations at exon 1, 7, and 9, leading to formation of protein lacking zinc finger domain. It’s role in AML pathogenesis is not completely clear, but is hypothesized to act as epigenetic modifier [109] and epigenetic targeted therapies may be employed.

ASXL1

It is mammalian homolog of additional sex comb like gene, a gene family with dual functions in both epigenetic activation and repression of gene transcription [110].

Mutations in ASXL1 gene are observed in 3-5% of AML cases and 11-17% in patients with intermediate risk (age ≥ 60 years) and secondary AML [111,112]. In elderly patients, ASXL1 mutations are associated with reduced CR and shorter OS [113].

EZH2

EZH2 catalyses progressive tri-methylation of H3K27 to induce the repression of target genes. Although, it is rarely seen in AML [114], EZH2 is frequently found genes mutation in childhood AML-M7; seen in 33% DS-AML-M7 & and in 16% of non-DS-AML-M7 cases [115].

Molecular genetics of acute lymphoblastic leukaemia

Acute lymphoblastic leukemia (ALL) is monoclonal neoplastic proliferation of lymphoid progenitor cells. These genetic mutations discussed below may be used for diagnostic prognostic purposes [116]. They can be divided into two main categories either B-ALL or T-ALL depending on the basis of antigen receptor rearrangements (Figure 2).

Figure 2.

Figure 2

Genetic mutation landscape of ALL; Mutations involved at the level of transcription factor activators are mutations involving TLX1/3, TAL1/2, LMO1/2 and PAX5. Mutations at the level of transcription suppressors are t(12;21) [RUNX1-ETV6] t(1;19) [TCF3-PBX1] and WT1 mutation. Mutations at the level of epigenetic modifiers is t(4;11) [AF4-MLL]. Mutations at the level of kinases are t(8;21) [BCR-ABL1]. Mutations at the level cell cycle regulators are p53 mutation and CDKN2A/B mutations.

B-cell-acute lymphoblastic leukaemia

They are derived from B lymphocytes and show B cell receptor rearrangement. As seen in AML, the ALLs also show mutations in genes that are involved in various cell cycle pathways and have their prognostic values.

Transcription factor

PAX-5

This is most common mutation seen in 31.7% cases of B-ALL [117]. It is a paired box domain (PRD) transcription factor. In all mutations, DNA binding region and nuclear localization region are retained but C-terminal trans-activation domain is deleted [118]. Therefore, these mutations facilitates C-terminal to act as transcription repressor. PAX-5 translocation correlates with normal karyotype. PAX-5 deletion is associated with complex karyotype [119].

Core binding factors (CBF) t(12;21) ETV6/RUNX1 fusion

RUNX-1 translocation present in 25% of paediatrics ALL {t(12;21)} [120]. In this translocation N-terminal of ETV6 (also called TEL) fuses with the coding region of RUNX-1 [121]. It is associated with good prognosis [122]. ETV6 is a transcription repressor & interacts with mSin3A, NCOR & HDAC-3 [123]. HDAC-3 inhibitor TSA inhibits two properties of fibroblasts cells [123], expression of Stromelysin 1 gene, and aggregation. It is the most common mutation in paediatric B-ALL [122]. Another mutation involving the RUNX1 gene leading to ALL is intra-chromosomal amplification of Chromosome 21 (iAMP21). There is a common region of amplification (CRA) on chromosome 21. CRA is associated with complex karyotype and multiple mutations associated with poor prognosis [124]. FISH has revealed ≥ 5 or ≥ 3 extra copies of genes on a single anomalous chromosome 21. In such cases there is shorter EFS and OS when treatment is based on the standard-risk protocols [125]. More intensified chemotherapy may lead to improvement in its poor prognosis [126].

TCF3 (E2A) translocation

They are associated with Pre-B-ALL type phenotype. t(1;19) TCF3/PBX-1 gene fusion on chromosome 19 [127,128]. TCF3 locus encodes protein which belongs to class I of βHelix-loop helix family (β-HLH) of transcription factor [129]. TCF3 (E2A) on heterodimerization interacts through the HLH domain with the member of class II β-HLH protein which is tissue specific. These transcriptional regulators are crucial for control of tissue specific genes during development. E2A protein is preferentially present in B lymphocytes [130]. In absence of E2A protein, there is complete block at the pro B cell stage prior to Ig gene rearrangement. Also, there is defective thymocyte differentiation with increased chances of development of B-ALL [131,132].

PBX-1 (Pre-B cell leukemia homeobox-1)

It forms hetero-dimer with other homeobox proteins via homeodomain and C-terminal HOX cooperative motif (HCM) [133]. PBX-1 is not normally present in lymphocytes. PBX-1 knockout mice develop multi-organ abnormalities [134]. PBX-1 is not a strong transactivator [135]. TCF3/PBX-1 fusion protein, developed by translocation between chromosomes 1 and 19 {t(1;19)(q23;p13)} has N-terminal 2/3 of TCF-3 protein that contain Activation Domain I & II and DNA binding domain of PBX-1. This leads to strong transactivation of target genes recognized by PBX-1 homeodomain [136]. In Pre-B cells, this transactivation of BMI-1 (its expression normally decreases during hematopoietic development) [137,138]. BMI-1 inhibits cell cycle inhibitors; CDKN2A [137]. CDKN2A via P14ARF & P16INK4A prevent phosphorylation (inactivation) of Rb gene [139]. It decreases DNA synthesis in S phase [140,141]. When there is repression of CDKN2A, there is increased DNA proliferation leading to leukemia. Another partner of TCF is HLF leading to t(17;19). TCF3/HLF fusion leads to acute leukemia which is seen in adolescent patients; presenting with coagulopathy & hypercalcemia and usually associated with poor prognosis [142,143]. Hepatic leukemia Factor (HLF) encodes DNA binding basic Zipper region can homo or heterodimerize with other b-ZIP protein. TCF3/HLF (transactivator of LMO2 & BCL2) is homologous to TCF3/PBX-1 protein. Unlike PBX1, HLF is strong transactivation [144,145]. The remaining intracellular portion is translocated to the nucleus as a transcription regulator with DNA binding portion of CSL.

IKAROS

IKZF-1 gene encode for IKAROS transcription factor mutated in 76.2% cases of paediatric Ph+ B-ALL [146]. IKAROS is expressed in multi-potent HSCs and promotes lymphoid differentiation & also for repression of genes responsible for self-renewal & multipotency in the differentiating progeny of HSCs [147]. IKZF-1 mutation is associated with down regulation & up regulation of genes involved in cell cycle regulation, apoptosis regulation & DNA damage & JAK-STAT signalling pathway [148]. In B-ALL, there is downregulation of genes involved in B cell differentiation (RAG & EBF-1) which may cause arrest of B cell maturation [148]. Gene expression pattern is the same as BCR-ABL1 positive ALL therefore also known as BCR-ABL-1 like ALL or Ph like ALL [149] associated with mutation of CK receptor & signalling molecules. Therefore, kinase inhibitors can be used for treatment [150].

Kinases

BCR-ABL-1

It results from translocation between chromosome 9 and 22; t(9;22). This is most common ALL in adults [151]. 5’ domain of BCR on chromosome 22 fuses with 3’ domain of ABL1 gene on chromosome 9 [152,153].Mainly two most common types of break point region are there [151], major BCR which are also seen CML and gives rise to 210 KD protein [154], minor BCR which gives rise to 190 KD protein [155] and accounts for 80% of paediatric ALL and 50% of adults ALL [151]. These cases are associated with an aggressive outcome [156]. Up-regulation of BCR-ABL1 fusion gene leads to activation of multiple signalling pathways such as MAPK, Ras, NF-kB, c-Myc, PI-3 kinase, and JAK-STAT [157].

Treatment of Ph+ ALL remains manage-intensive. Initial response to chemotherapy is similar in both Ph+ ALL as well as Ph- ALL, but remissions tend to be short lived in Ph+ cases. However, trials using imatinib in conjunction with standard chemotherapy followed by bone marrow transplant during CR phase have been proven more successful [158,159].

Novel mutations in B-ALL

RAS signalling

Mutations in various RAS Signalling genes (NRAS, KRAS, FLT3, PTPN11, NF1) are present in 48% of cases [160]. These mutations cause deregulation of the RAS-RAF-mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK signalling cascade [161]. Up regulation of RAS signalling, caused by mutations in RAS genes or in genes coding for proteins controlling RAS functions, represent major pathway for aberrant growth of malignant B-cell precursors [162].

NRAS & KRAS

Out of 3 RAS genes, mutations in HRAS are rare in hematologic tumors [163]. Various studies have identified presence of NRAS & KRAS mutations as recurrent genetic mutations in around 15 to 30% of childhood ALLs [162-164]. Presence of RAS mutations is not a significant risk factor [161,165] and associated neither with adverse clinical outcome [166] nor with relapse-free survival [164].

PTPN11

PTPN11 encodes for SHP2 protein which is an activator of RAS pathway. Mutations of PTPN11 causes enhanced signalling of RAS through MAP kinase pathway [167]. About 6 to 7.3% children with B-cell precursor ALL have PTPN11 mutations [162,167]. PTPN11 mutations occur with higher frequency in children and adolescents with hyper diploid DNA content [162].

BRAF

BRAF gene is part of RAF family and acts downstream in the RAS/RAF/MAP kinase pathway. BRAF mutations have been reported in 20% of B-cell ALLs cases [168,169]. BRAF acts by inhibiting need of cytokine for proliferation of hematopoietic cells leading to their uncontrolled proliferation [168]. According to J.W. Lee et al. BRAF inhibitors can be utilised for treatment of acute leukemias with mutant BRAF [168].

JAK/STAT signalling

JAK mutations in pseudokinase and kinase domains have been observed in 10% of BCR-ABL negative high risk paediatric ALL cases [164,170,171] and these mutations are associated with poor outcome [164]. According to one study, mutations in JAK alleles lead to activation of JAK stat pathway in mouse hematopoietic cells expressing erythropoietic receptor [172]. And also, JAK1/2 inhibitor can be used for treating patients harbouring these mutations [171,172].

JAK mutations are present simultaneously with mutations in IKZF1 and CDKN2A/B indicating activation of multiple pathways leading to induction of aggressive lymphoid leukemia in high risk BCR-ABL1-ALL [171]. Gain function mutation of JAK2 is seen in about 18% of ALL Down’s syndrome (DS-ALL) cases [173] and inhibition of JAK2 may be therapeutically useful in treating such cases [174].

TP53/RB1 pathway

P53 plays a crucial role in cell cycle regulation and apoptosis after DNA damage. TP53 alterations result in either a loss of protein expression or the generation of protein variants with (partly) impaired function. Expression of the p53 protein is strictly controlled by MDM2 and its homolog MDMX, which together form an E3 ubiquitin ligase complex that inhibits p53, resulting in nuclear export and subsequent degradation. Mutations in TP53 suppressor gene are rare events in BCP-ALL, occurring in only 3% of primary cases especially with low hypo-diploid type [175,176]. In relapsed ALL, the frequency of TP53 mutations increases and represents a strong and independent predictor of treatment failure [177]. There is strong correlation between platelet count and MRD positivity following induction chemotherapy. Therefore, it can be used as prognostic factor for risk stratification trials [178]. “p53 reactivation and induction of massive apoptosis” (PRIMA-1) and its methylated derivative PRIMA-1, also known as APR-246 leads to reactivation of mutant p53 [179].

T cell-acute lymphoblastic leukaemia

They originate from T-lymphocytes. As seen in AMLs and B-ALLs not much is known about the genetics of T-ALLs. However, some of the genes that have some prognostic significance are described in brief here.

NOTCH1

NOTCH1 is a regulatory protein that is important in many cell fate decisions, including commitment to T-cell lineage and choice of αβ lineage [180-182]. It was first cloned from a t(7;9)(q34;q34) translocation occurring in a T-ALL patient [183]. The t(7;9) translocation turned out to be rare in T-ALL but targeted sequencing revealed that over 60% of T-ALL patients have activating mutations in NOTCH1 [184]. NOTCH1 is synthesized as a single polypeptide protein that is cleaved in the golgi body at site S1 into two sub-units, the ligand-binding NEC (extracellular) and NTM (transmembrane), both bind non-covalently at the hetero-dimerization domain. Upon binding of ligand to NEC, the NTM is cleaved at site S2 by a metalloproteases, and cleaved at S3 by gamma secretase [185]. The remaining intracellular portion, ICN1, goes to the nucleus, where it acts as a transcriptional regulator with DNA-binding protein CSL [186]. The majority of the activating mutations in NOTCH1 found in T-ALL occurs in the heterodimerization domain or in PEST domain. The PEST domain regulates the turnover of NOTCH1. Therefore, the hetero-dimerization domain mutants leads to activation of NOTCH1 even in absence of ligand and the PEST domain mutants increases NOTCH1 protein stability [184]. The NOTCH1 has many targets affecting various pathways required for cell maturation. Among these, MYC and HES1 appear to be important for T-cell leukemogenesis [187-189]. The unique proteolytic pathway leading to activated NOTCH1 can be targeted by small molecule inhibitors of the gamma secretase enzyme that is required for S3 cleavage [190,191].

Transcription factor

TAL1 gene mutations

There are 3 TAL1 related genes; TAL1, TAL2 and LYL1. These genes are not normally present in normal T lymphocytes but various mutations involving these genes leads to aberrant expression of these genes in T-ALLs [192]. TAL1 gene mutation related translocations accounts for 1-3% of all childhood ALLs. They include t(1;14)(p32;q11), t(1;14)(p34;q11) and t(1;7)(p32;q34) with other partner genes such as TR-A and TR-D [193]. Also, there are some point mutations seen in TAL1 gene. All these mutations lead to over-expression of these gene [146]. All these mutations causes silencing of target genes encoding for E47 and E12 variants of E2A transcription factors [194,195]. TAL-1 overexpression is associated with poor prognosis except TAL-1/TCRα/β gene t(1;14) is having good prognosis [196]. TAL-1 is expressed in erythroid, megakaryocytes & mast cell lineage but not in T cells. In non-erythroid cells, TAL-1 is expressed in stem cells but not in differentiated cells [197]. Complete absence of TAL-1 is embryonically lethal [198,199]. Reactivation of silenced genes by administering histone deacetylase (HDAC) inhibitors may prove efficacious in T-ALL patients expressing TAL1 [193].

TAL2 [t(7;9)(q34;q32)]

TAL2 related translocation juxtaposes with TRB gene, t(7;9)(q34;q32), leading to its activation. Both TAL1 and TAL2 gene promotes proliferation of T-ALL blast by a common pathway [200].

LYL1 [t(7;19)(q34;p13)]

In t(7;19)(q34;p13) LYL1 gene juxtaposes with TRB leading to its activation. In normal T lymphocytes this gene is not expressed but seen in T-ALLs. LYL1-transgenic mice developed CD4+, CD8+ precursor T-cell ALL (pre-T-LBL), by its effect on E2A transcription factor, leading to down-regulation of E2A; as result of block in normal development [200].

LMO gene abnormalities

LMO gene encodes for two types of proteins LMO1 and LMO2. This gene is very frequently rearranged with TCR, resulting in over-expression of LMO1 or LMO2 protein in T-ALLs [201]. De Keersmaecker et al. study on transgenic mice showed that TAL1 expression alone is not sufficient for development of T-ALL and develops only when it is coexpressed with either LMO1 or LMO2 [191].

Homeodomain protein

HOX is major gene family being dysregulated in T-ALL & mutually exclusive to b-HLH gene & LMO gene [202]. This family of transcription factors is divided into two classes. Class I HOX gene includes 4 gene clusters; HOXA, HOXB, HOXC and HOXD present at chromosomes 7p15, 17q21, 12q13 and 2q31 respectively. Class II HOX genes are present throughout the genome. In class I HOX genes HOXA is involved in the development of T-ALL and in class II HOX genes TLX1 (HOX11) and TLX3 (HOX11L2) are the most common genes involved [192].

TLX1 (HOX11) gene translocation

TLX-1 is overexpressed in 7% TALL and associated with good prognosis [203,204]. Two types of translocations are seen involving TLX1 gene t(10;14)leading to fusion between TLX1 gene and TRA1 and t(7;10) leading to fusion of genes between TLX1 gene and TRB. TLX-1 (associated with t(10;14) TLX-1/TRD) mutation leads block at cortical stage [205,206]. Loss of TCR rearrangement leads to aberrant expression of TLX1 gene in T lymphocytes [191].

TLX3 (HOX11L2) gene translocation

TLX3 shows fusion of t(5;14)(q35;q32) genes and BCL11B resulting into ectopic expression of TLX3 gene in T lymphocytes [208]. TLX-3 is deregulated by t(5;14) in 20% of adult T-ALL [208,209]. This ectopic expression of TLX3 gene in children with T-ALL is associated with poor prognosis [191]. Other variants of this translocation is t(5;7)(q35;q21) in which CDK6 gene is fused with TLX3 gene.

Novel gene mutations

Inv(7)(p15q34)

The inversion leads to juxtaposition of part of TRB locus 7(q34-35) with the HOX-A cluster 7(p15). This leads to over expression of HOXA10 and HOXA1. HOXA is dysregulated by inv7 in 5% of T-ALL [210-212]. These genes are normally expressed in maturing thymocytes, but, there down regulation is required for normal maturation of both CD4 and CD8 positive T lymphocytes [191]. MLL & its partners PICALM & MLLT10 deregulate expressions of HOX-A. MYB duplication is seen in 8.4% of T-ALL [213,214]. Deletion or missense mutation of BCL11b gene leads to defect in T cell maturation [215,216]. PHF6 mutation is 10-fold more prevalent in males and it is uncommon in TLX-negative cases [217].

Conclusion

Molecular studies for genetic mutations that are involved in acute leukaemia’s have improved our knowledge about synchrony of various pathobiological events in the form of mutations, expression and changed downstream signalling pathways. This knowledge can be utilized to sub-classify these with the target of prognostic risk stratification and clinical management. The compiled reviews were aimed to give a brief insight of the already discovered and upcoming newer mutation and their contributory role in leukemogenesis. The article also emphasizes the impact of such research being translated in to the treatment protocols with overall arching goal of personalized therapy.

Disclosure of conflict of interest

None.

References

  • 1.Saki N, Abroun S, Farshdousti Hagh M, Asgharei F. Neoplastic bone marrow niche: hematopoietic and mesenchymal stem cells. Cell J. 2011;13:131–136. [PMC free article] [PubMed] [Google Scholar]
  • 2.Dohner H, Weisdorf D, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373:1136–1152. doi: 10.1056/NEJMra1406184. [DOI] [PubMed] [Google Scholar]
  • 3.Dohner H, Estey EH, Amadori S, Appelbaum FR, Büchner T, Burnett AK, Dombret H, Fenaux P, Grimwade D, Larson RA, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz MA, Sierra J, Tallman MS, Löwenberg B, Bloomfield CD. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European Leukemia Net. Blood. 2010;115:453–474. doi: 10.1182/blood-2009-07-235358. [DOI] [PubMed] [Google Scholar]
  • 4.Gerstung M, Papaemmanuil E, Martincorena I, Bullinger L, Gaidzik VI, Paschka P, Heuser M, Thol F, Bolli N, Ganly P, Ganser A, McDermott U, Dohner K, Schlenk RF, Döhner H, Campbell PJ. Precision oncology for acute myeloid leukemia using a knowledge bank approach. Nat Genet. 2017;49:332–340. doi: 10.1038/ng.3756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Nowell PC. The minute chromosome (Phl) in chronic granulocytic leukemia. Blut. 1962;8:65–66. doi: 10.1007/BF01630378. [DOI] [PubMed] [Google Scholar]
  • 6.Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S, Sawyers CL. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–1037. doi: 10.1056/NEJM200104053441401. [DOI] [PubMed] [Google Scholar]
  • 7.Cazzola M. Introduction to a review series: the 2016 revision of the WHO classification of tumors of hematopoietic and lymphoid tissues. Blood. 2016;127:2361–2364. doi: 10.1182/blood-2016-03-657379. [DOI] [PubMed] [Google Scholar]
  • 8.Walter MJ, Shen D, Ding L, Shao J, Koboldt DC, Chen K, Larson DE, McLellan MD, Dooling D, Abbott R, Fulton R, Magrini V, Schmidt H, Kalicki-Veizer J, O’Laughlin M, Fan X, Grillot M, Witowski S, Heath S, Frater JL, Eades W, Tomasson M, Westervelt P, DiPersio JF, Link DC, Mardis ER, Ley TJ, Wilson RK, Graubert TA. Clonal architecture of secondary acute myeloid leukemia. N Engl J Med. 2012;366:1090–1098. doi: 10.1056/NEJMoa1106968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S, O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell PJ. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374:2209–2221. doi: 10.1056/NEJMoa1516192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cancer Genome Atlas Research Network. Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson A, Hoadley K, Triche TJ Jr, Laird PW, Baty JD, Fulton LL, Fulton R, Heath SE, Kalicki-Veizer J, Kandoth C, Klco JM, Koboldt DC, Kanchi KL, Kulkarni S, Lamprecht TL, Larson DE, Lin L, Lu C, McLellan MD, McMichael JF, Payton J, Schmidt H, Spencer DH, Tomasson MH, Wallis JW, Wartman LD, Watson MA, Welch J, Wendl MC, Ally A, Balasundaram M, Birol I, Butterfield Y, Chiu R, Chu A, Chuah E, Chun HJ, Corbett R, Dhalla N, Guin R, He A, Hirst C, Hirst M, Holt RA, Jones S, Karsan A, Lee D, Li HI, Marra MA, Mayo M, Moore RA, Mungall K, Parker J, Pleasance E, Plettner P, Schein J, Stoll D, Swanson L, Tam A, Thiessen N, Varhol R, Wye N, Zhao Y, Gabriel S, Getz G, Sougnez C, Zou L, Leiserson MD, Vandin F, Wu HT, Applebaum F, Baylin SB, Akbani R, Broom BM, Chen K, Motter TC, Nguyen K, Weinstein JN, Zhang N, Ferguson ML, Adams C, Black A, Bowen J, Gastier-Foster J, Grossman T, Lichtenberg T, Wise L, Davidsen T, Demchok JA, Shaw KR, Sheth M, Sofia HJ, Yang L, Downing JR, Eley G. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;22:2059–2074. doi: 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Harris NL, Arber DA, Campo E, Jaffe ES, Orazi A, Pileri SA, Stien H, Swerdlow SH, Thiele J, Vardiman JW. Introduction to the WHO classification of tumours of haematopoietic and lymphoid tissue. WHO Classification of Tumours of the Haematopoietic and Lymphoid Tissues. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stien H, Thiele JJ, editors. Geneva: WHO Press; 2017. pp. 13–27. [Google Scholar]
  • 12.Schulman IG, Juguilon H, Evans RM. Activation and repression by nuclear hormone receptors: hormone modulates an equilibrium between active and repressive states. Mol Cell Biol. 1996;16:3807–3813. doi: 10.1128/mcb.16.7.3807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Melnick A, Licht JD. Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia. Blood. 1999;93:3167–3215. [PubMed] [Google Scholar]
  • 14.Guidez F, Ivins S, Zhu J, Soderström M, Waxman S, Zelent A. Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARalpha underlie molecular pathogenesis and treatment of acute promyelocytic leukemia. Blood. 1998;91:2634–2642. [PubMed] [Google Scholar]
  • 15.Lin RJ, Nagy L, Inoue S, Shao W, Miller WH Jr, Evans RM. Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature. 1998;391:811–814. doi: 10.1038/35895. [DOI] [PubMed] [Google Scholar]
  • 16.Grignani F, De Matteis S, Nervi C, Tomassoni L, Gelmetti V, Cioce M, Fanelli M, Ruthardt M, Ferrara FF, Zamir I, Seiser C, Grignani F, Lazar MA, Minucci S, Pelicci PG. Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature. 1998;391:815–818. doi: 10.1038/35901. [DOI] [PubMed] [Google Scholar]
  • 17.Melnick AM, Westendorf JJ, Polinger A, Carlile GW, Arai S, Ball HJ, Lutterbach B, Hiebert SW, Licht JD. The ETO protein disrupted in t8;21-associated acute myeloid leukemia is a corepressor for the promyelocytic leukemia zinc finger protein. Mol Cell Biol. 2000;20:2075–2086. doi: 10.1128/mcb.20.6.2075-2086.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Look AT. Oncogenic transcription factors in the human acute leukemias. Science. 1997;278:1059–1064. doi: 10.1126/science.278.5340.1059. [DOI] [PubMed] [Google Scholar]
  • 19.Golemis EA, Speck NA, Hopkins N. Alignment of U3 region sequences of mammalian type C viruses: identification of highly conserved motifs and implications for enhancer design. J Virol. 1990;64:534–542. doi: 10.1128/jvi.64.2.534-542.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Liu P, Tarle SA, Hajra A, Claxton DF, Marlton P, Freedman M, Siciliano MJ, Collins FS. Fusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemia. Science. 1993;261:1041–1044. doi: 10.1126/science.8351518. [DOI] [PubMed] [Google Scholar]
  • 21.Gamou T, Kitamura E, Hosoda F, Shimizu K, Shinohara K, Hayashi Y, Nagase T, Yokoyama Y, Ohki M. The partner gene of AML1 in t16;21 myeloid malignancies is a novel member of the MTG8ETO family. Blood. 1998;91:4028–4037. [PubMed] [Google Scholar]
  • 22.Michaud J, Wu F, Osato M, Cottles GM, Yanagida M, Asou N, Shigesada K, Ito Y, Benson KF, Raskind WH, Rossier C, Antonarakis SE, Israels S, McNicol A, Weiss H, Horwitz M, Scott HS. In vitro analyses of known and novel RUNX1/AML1 mutations in dominant familial platelet disorder with predisposition to acute myelogenous leukemia: implications for mechanisms of pathogenesis. Blood. 2002;99:1364–1372. doi: 10.1182/blood.v99.4.1364. [DOI] [PubMed] [Google Scholar]
  • 23.Osato M, Asou N, Abdalla E, Hoshino K, Yamasaki H, Okubo T, Suzushima H, Takatsuki K, Kanno T, Shigesada K, Ito Y. Biallelic and heterozygous point mutations in the runt domain of the AML1/PEBP2alphaB gene associated with myeloblastic leukemias. Blood. 1999;93:1817–1824. [PubMed] [Google Scholar]
  • 24.Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K, Shiina M, Sato K, Kumasaka T, Yamamoto M, Ishii S, Ogata K. Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain and its allosteric control by CBFbeta. Cell. 2001;104:755–767. doi: 10.1016/s0092-8674(01)00271-9. [DOI] [PubMed] [Google Scholar]
  • 25.Licht JD. AML1 and the AML1-ETO fusion protein in the pathogenesis of t8;21 AML. Oncogene. 2001;20:5660–5679. doi: 10.1038/sj.onc.1204593. [DOI] [PubMed] [Google Scholar]
  • 26.Libermann TA, Pan Z, Akbarali Y, Hetherington CJ, Boltax J, Yergeau DA, Zhang DE. AML1 CBFalpha2 cooperates with B cell-specific activating protein BSAP/PAX5 in activation of the B cell-specific BLK gene promoter. J Biol Chem. 1999;274:24671–54676. doi: 10.1074/jbc.274.35.24671. [DOI] [PubMed] [Google Scholar]
  • 27.Uchida H, Zhang J, Nimer SD. AML1A and AML1B can transactivate the human IL-3 promoter. J Immunol. 1997;158:2251–2258. [PubMed] [Google Scholar]
  • 28.Takahashi A, Satake M, Yamaguchi-Iwai Y, Bae SC, Lu J, Maruyama M, Zhang YW, Oka H, Arai N, Arai K. Positive and negative regulation of granulocyte-macrophage colony-stimulating factor promoter activity by AML1-related transcription factor, PEBP2. Blood. 1995;86:607–616. [PubMed] [Google Scholar]
  • 29.Nuchprayoon I, Meyers S, Scott LM, Suzow J, Hiebert S, Friedman AD. 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. 1994;14:5558–5568. doi: 10.1128/mcb.14.8.5558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Meyers S, Downing JR, Hiebert SW. Identification of AML-1 and the 8;21 translocation protein AML-1/ETO as sequence-specific DNA-binding proteins: the runt homology domain is required for DNA binding and protein-protein interactions. Mol Cell Biol. 1993;13:6336–6645. doi: 10.1128/mcb.13.10.6336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Amann JM, Nip J, Strom DK, Lutterbach B, Harada H, Lenny N, Downing JR, Meyers S, Hiebert SW. ETO, a target of t8;21 in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain. Mol Cell Biol. 2001;21:6470–6483. doi: 10.1128/MCB.21.19.6470-6483.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang Q, Stacy T, Binder M, Marin-Padilla M, Sharpe AH, Speck NA. Disruption of the Cbfa2 gene causes necrosis and hemorrhaging in the central nervous system and blocks definitive hematopoiesis. Proc Natl Acad Sci U S A. 1996;21:3444–3449. doi: 10.1073/pnas.93.8.3444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gaidzik VI, Bullinger L, Schlenk RF, Zimmermann AS, Rock J, Paschka P, Corbacioglu A, Krauter J, Schlegelberger B, Ganser A, Spath D, Kündgen A, Schmidt-Wolf IG, Gotze K, Nachbaur D, Pfreundschuh M, Horst HA, Dohner H, Dohner K. RUNX1 mutations in acute myeloid leukemia: results from a comprehensive genetic and clinical analysis from the AML study group. J. Clin. Oncol. 2011;29:1364–1372. doi: 10.1200/JCO.2010.30.7926. [DOI] [PubMed] [Google Scholar]
  • 34.Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J. WHO classification of tumors of haematopoietic and lymphoid tissues. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, editors. Lyon, France: IARC Press; 2008. [Google Scholar]
  • 35.Durst KL, Lutterbach B, Kummalue T, Friedman AD, Hiebert SW. The inv16 fusion protein associates with corepressors via a smooth muscle myosin heavy-chain domain. Mol Cell Biol. 2003;23:607–619. doi: 10.1128/MCB.23.2.607-619.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Song WJ, Sullivan MG, Legare RD, Hutchings S, Tan X, Kufrin D, Ratajczak J, Resende IC, Haworth C, Hock R, Loh M, Felix C, Roy DC, Busque L, Kurnit D, Willman C, Gewirtz AM, Speck NA, Bushweller JH, Li FP, Gardiner K, Poncz M, Maris JM, Gilliland DG. Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nat Genet. 1999;23:166–175. doi: 10.1038/13793. [DOI] [PubMed] [Google Scholar]
  • 37.Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, Levine RL, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz M, Sierra J, Tallman MS, Tien HF, Wei AH, Löwenberg B, Bloomfield CD. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:424–447. doi: 10.1182/blood-2016-08-733196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M, Estey EH, Dombret H, Chevret S, Ifrah N, Cahn JY, Récher C, Chilton L, Moorman AV, Burnett AK. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15:986–996. doi: 10.1016/S1470-2045(14)70281-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tenen DG, Hromas R, Licht JD, Zhang DE. Transcription factors, normal myeloid development, and leukemia. Blood. 1997;90:489–519. [PubMed] [Google Scholar]
  • 40.Paz-Priel I, Friedman A. C/EBPα dysregulation in AML and ALL. Crit Rev Oncog. 2011;16:93–102. doi: 10.1615/critrevoncog.v16.i1-2.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pabst T, Mueller BU, Harakawa N, Schoch C, Haferlach T, Behre G, Hiddemann W, Zhang DE, Tenen DG. AML1-ETO downregulates the granulocytic differentiation factor C/EBPalpha in t8;21 myeloid leukemia. Nat Med. 2001;7:444–451. doi: 10.1038/86515. [DOI] [PubMed] [Google Scholar]
  • 42.Radomska HS, Bassères DS, Zheng R, Zhang P, Dayaram T, Yamamoto Y, Sternberg DW, Lokker N, Giese NA, Bohlander SK, Schnittger S, Delmotte MH, Davis RJ, Small D, Hiddemann W, Gilliland DG, Tenen DG. Block of C/EBP alpha function by phosphorylation in acute myeloid leukemia with FLT3 activating mutations. J Exp Med. 2006;203:371–381. doi: 10.1084/jem.20052242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zhang DE, Zhang P, Wang ND, Hetherington CJ, Darlington GJ, Tenen DG. Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein alpha-deficient mice. Proc Natl Acad Sci U S A. 1997;94:569–574. doi: 10.1073/pnas.94.2.569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gombart AF, Hofmann WK, Kawano S, Takeuchi S, Krug U, Kwok SH, Larsen RJ, Asou H, Miller CW, Hoelzer D, Koeffler HP. Mutations in the gene encoding the transcription factor CCAAT/enhancer binding protein alpha in myelodysplastic syndromes and acute myeloid leukemias. Blood. 2002;99:1332–1340. doi: 10.1182/blood.v99.4.1332. [DOI] [PubMed] [Google Scholar]
  • 45.Pabst T, Mueller BU, Zhang P, Radomska HS, Narravula S, Schnittger S, Behre G, Hiddemann W, Tenen DG. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha C/EBPalpha, in acute myeloid leukemia. Nat Genet. 2001;27:263–270. doi: 10.1038/85820. [DOI] [PubMed] [Google Scholar]
  • 46.Preudhomme C, Sagot C, Boissel N, Cayuela JM, Tigaud I, de Botton S, Thomas X, Raffoux E, Lamandin C, Castaigne S, Fenaux P, Dombret H ALFA Group. Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association ALFA. Blood. 2002;100:2717–2723. doi: 10.1182/blood-2002-03-0990. [DOI] [PubMed] [Google Scholar]
  • 47.Grossmann V, Schnittger S, Kohlmann A, Eder C, Roller A, Dicker F, Schmid C, Wendtner CM, Staib P, Serve H, Kreuzer KA, Kern W, Haferlach T, Haferlach C. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood. 2012;120:2963–2972. doi: 10.1182/blood-2012-03-419622. [DOI] [PubMed] [Google Scholar]
  • 48.Konstandin NP, Pastore F, Herold T, Dufour A, Rothenberg-Thurley M, Hinrichsen T, Ksienzyk B, Tschuri S, Schneider S, Hoster E, Berdel WE, Woermann BJ, Sauerland MC, Braess J, Bohlander SK, Klein HG, Hiddemann W, Metzeler KH, Spiekermann K. Genetic heterogeneity of cytogenetically normal AML with mutations of CEBPA. Blood Adv. 2018;2:2724–2731. doi: 10.1182/bloodadvances.2018016840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Greene ME, Mundschau G, Wechsler J, McDevitt M, Gamis A, Karp J, Gurbuxani S, Arceci R, Crispino JD. Mutations in GATA1 in both transient myeloproliferative disorder and acute megakaryoblastic leukemia of Down syndrome. Blood Cells Mol Dis. 2003;31:351–356. doi: 10.1016/j.bcmd.2003.08.001. [DOI] [PubMed] [Google Scholar]
  • 50.Gurbuxani S, Vyas P, Crispino JD. Recent insights into the mechanisms of myeloid leukemogenesis in Down syndrome. Blood. 2004;103:399–406. doi: 10.1182/blood-2003-05-1556. [DOI] [PubMed] [Google Scholar]
  • 51.Wechsler J, Greene M, McDevitt MA, Anastasi J, Karp JE, Le Beau MM, Crispino JD. Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet. 2002;32:148–152. doi: 10.1038/ng955. [DOI] [PubMed] [Google Scholar]
  • 52.Roy A, Roberts I, Norton A, Vyas P. Acute megakaryoblastic leukaemia AMKL and transient myeloproliferative disorder TMD in Down syndrome: a multi-step model of myeloid leukaemogenesis. Br J Haematol. 2009;147:3–12. doi: 10.1111/j.1365-2141.2009.07789.x. [DOI] [PubMed] [Google Scholar]
  • 53.Creutzig U, Reinhardt D, Diekamp S, Dworzak M, Stary J, Zimmermann M. AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity. Leukemia. 2005;19:1355–1360. doi: 10.1038/sj.leu.2403814. [DOI] [PubMed] [Google Scholar]
  • 54.Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, Koboldt DC, Fulton RS, Delehaunty KD, McGrath SD, Fulton LA, Locke DP, Magrini VJ, Abbott RM, Vickery TL, Reed JS, Robinson JS, Wylie T, Smith SM, Carmichael L, Eldred JM, Harris CC, Walker J, Peck JB, Du F, Dukes AF, Sanderson GE, Brummett AM, Clark E, McMichael JF, Meyer RJ, Schindler JK, Pohl CS, Wallis JW, Shi X, Lin L, Schmidt H, Tang Y, Haipek C, Wiechert ME, Ivy JV, Kalicki J, Elliott G, Ries RE, Payton JE, Westervelt P, Tomasson MH, Watson MA, Baty J, Heath S, Shannon WD, Nagarajan R, Link DC, Walter MJ, Graubert TA, DiPersio JF, Wilson RK, Ley TJ. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361:1058–1066. doi: 10.1056/NEJMoa0903840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Lowenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, Melnick A. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18:553–567. doi: 10.1016/j.ccr.2010.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, Liu LX, Jiang WQ, Liu J, Zhang JY, Wang B, Frye S, Zhang Y, Xu YH, Lei QY, Guan KL, Zhao SM, Xiong Y. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011;19:17–30. doi: 10.1016/j.ccr.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Williams K, Christensen J, Helin K. DNA methylation: TET proteins-guardians of CpG islands? EMBO Rep. 2011;13:28–35. doi: 10.1038/embor.2011.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Feng JH, Guo XP, Chen YY, Wang ZJ, Cheng YP, Tang YM. Prognostic significance of IDH1 mutations in acute myeloid leukemia: a meta-analysis. Am J Blood Res. 2012;2:254–264. [PMC free article] [PubMed] [Google Scholar]
  • 59.Stein EM, DiNardo CD, Pollyea DA, Fathi AT, Roboz GJ, Altman JK, Stone RM, DeAngelo DJ, Levine RL, Flinn IW, Kantarjian HM, Collins R, Patel MR, Frankel AE, Stein A, Sekeres MA, Swords RT, Medeiros BC, Willekens C, Vyas P, Tosolini A, Xu Q, Knight RD, Yen KE, Agresta S, de Botton S, Tallman MS. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130:722–731. doi: 10.1182/blood-2017-04-779405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kim ES. Enasidenib: first global approval. Drugs. 2017;77:1705–1711. doi: 10.1007/s40265-017-0813-2. [DOI] [PubMed] [Google Scholar]
  • 61.Smith ZD, Meissner A. DNA methylation: roles in mammalian development. Nat Rev Genet. 2013;14:204–220. doi: 10.1038/nrg3354. [DOI] [PubMed] [Google Scholar]
  • 62.Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, Kandoth C, Payton JE, Baty J, Welch J, Harris CC, Lichti CF, Townsend RR, Fulton RS, Dooling DJ, Koboldt DC, Schmidt H, Zhang Q, Osborne JR, Lin L, O’Laughlin M, McMichael JF, Delehaunty KD, McGrath SD, Fulton LA, Magrini VJ, Vickery TL, Hundal J, Cook LL, Conyers JJ, Swift GW, Reed JP, Alldredge PA, Wylie T, Walker J, Kalicki J, Watson MA, Heath S, Shannon WD, Varghese N, Nagarajan R, Westervelt P, Tomasson MH, Link DC, Graubert TA, DiPersio JF, Mardis ER, Wilson RK. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363:2424–2433. doi: 10.1056/NEJMoa1005143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Shlush LI, Zandi S, Mitchell A, Chen WC, Brandwein JM, Gupta V, Kennedy JA, Schimmer AD, Schuh AC, Yee KW, McLeod JL, Doedens M, Medeiros JJ, Marke R, Kim HJ, Lee K, McPherson JD, Hudson TJ HALT Pan-Leukemia Gene Panel Consortium. Brown AM, Yousif F, Trinh QM, Stein LD, Minden MD, Wang JC, Dick JE. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014;506:328–333. doi: 10.1038/nature13038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Corces-Zimmerman MR, Hong WJ, Weissman IL, Medeiros BC, Majeti R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci U S A. 2014;111:2548–2553. doi: 10.1073/pnas.1324297111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tie R, Zhang T, Fu H, Wang L, Wang Y, He Y, Wang B, Zhu N, Fu S, Lai X, Shi J, Huang H. Association between DNMT3A mutations and prognosis of adults with de novo acute myeloid leukemia: a systematic review and meta-analysis. PLoS One. 2014;9:e93353. doi: 10.1371/journal.pone.0093353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yuan XQ, Peng L, Zeng WJ, Jiang BY, Li GC, Chen XP. DNMT3A R882 mutations predict a poor prognosis in AML: a meta-analysis from 4474 patients. Medicine (Baltimore) 2016;95:e3519. doi: 10.1097/MD.0000000000003519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Metzeler KH, Walker A, Geyer S, Garzon R, Klisovic RB, Bloomfield CD, Blum W, Marcucci G. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia. 2012;26:1106–1107. doi: 10.1038/leu.2011.342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Im AP, Sehgal AR, Carroll MP, Smith BD, Tefferi A, Johnson DE, Boyiadzis M. DNMT3A and IDH mutations in acute myeloid leukemia and other myeloid malignancies: associations with prognosis and potential treatment strategies. Leukemia. 2014;28:1774–1783. doi: 10.1038/leu.2014.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ingham PW. Trithorax and the regulation of homeotic gene expression in Drosophila: a historical perspective. Int J Dev Biol. 1998;42:423–429. [PubMed] [Google Scholar]
  • 70.Ernst P, Fisher JK, Avery W, Wade S, Foy D, Korsmeyer SJ. Definitive hematopoiesis requires the mixed-lineage leukemia gene. Dev Cell. 2004;6:437–443. doi: 10.1016/s1534-5807(04)00061-9. [DOI] [PubMed] [Google Scholar]
  • 71.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  • 72.Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7:823–833. doi: 10.1038/nrc2253. [DOI] [PubMed] [Google Scholar]
  • 73.Bernt KM, Armstrong SA. Targeting epigenetic programs in MLL-rearranged leukemias. Hematology Am Soc Hematol Educ Program. 2011;2011:354–60. doi: 10.1182/asheducation-2011.1.354. [DOI] [PubMed] [Google Scholar]
  • 74.Ford AM, Ridge SA, Cabrera ME, Mahmoud H, Steel CM, Chan LC, Greaves M. In utero rearrangements in the trithorax-related oncogene in infant leukaemias. Nature. 1993;363:358–360. doi: 10.1038/363358a0. [DOI] [PubMed] [Google Scholar]
  • 75.Super HJ, McCabe NR, Thirman MJ, Larson RA, Le Beau MM, Pedersen-Bjergaard J, Philip P, Diaz MO, Rowley JD. Rearrangements of the MLL gene in therapy-related acute myeloid leukemia in patients previously treated with agents targeting DNA-topoisomerase II. Blood. 1993;82:3705–3711. [PubMed] [Google Scholar]
  • 76.Rubnitz JE, Link MP, Shuster JJ, Carroll AJ, Hakami N, Frankel LS, Pullen DJ, Cleary ML. Frequency and prognostic significance of HRX rearrangements in infant acute lymphoblastic leukemia: a Pediatric Oncology Group study. Blood. 1994;84:570–573. [PubMed] [Google Scholar]
  • 77.Brasel K, Escobar S, Anderberg R, de Vries P, Gruss HJ, Lyman SD. Expression of the flt3 receptor and its ligand on hematopoietic cells. Leukemia. 1995;9:1212–1218. [PubMed] [Google Scholar]
  • 78.Brandts CH, Sargin B, Rode M, Biermann C, Lindtner B, Schwäble J, Buerger H, Müller-Tidow C, Choudhary C, McMahon M, Berdel WE, Serve H. Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res. 2005;65:9643–9650. doi: 10.1158/0008-5472.CAN-05-0422. [DOI] [PubMed] [Google Scholar]
  • 79.Mizuki M, Fenski R, Halfter H, Matsumura I, Schmidt R, Müller C, Grüning W, Kratz-Albers K, Serve S, Steur C, Büchner T, Kienast J, Kanakura Y, Berdel WE, Serve H. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood. 2000;96:3907–3914. [PubMed] [Google Scholar]
  • 80.Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, Sonoda Y, Fujimoto T, Misawa S. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10:1911–1918. [PubMed] [Google Scholar]
  • 81.Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002;100:1532–1542. doi: 10.1182/blood-2002-02-0492. [DOI] [PubMed] [Google Scholar]
  • 82.Gale RE, Green C, Allen C, Mead AJ, Burnett AK, Hills RK, Linch DC Medical Research Council Adult Leukaemia Working Party. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111:2776–2784. doi: 10.1182/blood-2007-08-109090. [DOI] [PubMed] [Google Scholar]
  • 83.Patel JP, Gönen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J, Van Vlierberghe P, Dolgalev I, Thomas S, Aminova O, Huberman K, Cheng J, Viale A, Socci ND, Heguy A, Cherry A, Vance G, Higgins RR, Ketterling RP, Gallagher RE, Litzow M, van den Brink MR, Lazarus HM, Rowe JM, Luger S, Ferrando A, Paietta E, Tallman MS, Melnick A, Abdel-Wahab O, Levine RL. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366:1079–1089. doi: 10.1056/NEJMoa1112304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Stirewalt DL, Kopecky KJ, Meshinchi S, Appelbaum FR, Slovak ML, Willman CL, Radich JP. FLT3, RAS, and TP53 mutations in elderly patients with acute myeloid leukemia. Blood. 2001;97:3589–3595. doi: 10.1182/blood.v97.11.3589. [DOI] [PubMed] [Google Scholar]
  • 85.Schlenk RF, Kayser S, Bullinger L, Kobbe G, Casper J, Ringhoffer M, Held G, Brossart P, Lübbert M, Salih HR, Kindler T, Horst HA, Wulf G, Nachbaur D, Götze K, Lamparter A, Paschka P, Gaidzik VI, Teleanu V, Späth D, Benner A, Krauter J, Ganser A, Dohner H, Döhner K German-Austrian AML Study Group. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124:3441–3449. doi: 10.1182/blood-2014-05-578070. [DOI] [PubMed] [Google Scholar]
  • 86.Kelly LM, Yu JC, Boulton CL, Apatira M, Li J, Sullivan CM, Williams I, Amaral SM, Curley DP, Duclos N, Neuberg D, Scarborough RM, Pandey A, Hollenbach S, Abe K, Lokker NA, Gilliland DG, Giese NA. CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia AML. Cancer Cell. 2002;1:421–432. doi: 10.1016/s1535-6108(02)00070-3. [DOI] [PubMed] [Google Scholar]
  • 87.Weisberg E, Boulton C, Kelly LM, Manley P, Fabbro D, Meyer T, Gilliland DG, Griffin JD. Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell. 2002;1:433–443. doi: 10.1016/s1535-6108(02)00069-7. [DOI] [PubMed] [Google Scholar]
  • 88.Majothi S, Adams D, Loke J, Stevens SP, Wheatley K, Wilson JS. FLT3 inhibitors in acute myeloid leukaemia: assessment of clinical effectiveness, adverse events and future research-a systematic review and meta-analysis. Syst Rev. 2020;9:285. doi: 10.1186/s13643-020-01540-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ruggero D, Pandolfi PP. Does the ribosome translate cancer? Nat Rev Cancer. 2003;3:179–192. doi: 10.1038/nrc1015. [DOI] [PubMed] [Google Scholar]
  • 90.Grisendi S, Pandolfi PP. NPM mutations in acute myelogenous leukemia. N Engl J Med. 2005;352:291–292. doi: 10.1056/NEJMe048337. [DOI] [PubMed] [Google Scholar]
  • 91.Falini B, Bolli N, Liso A, Martelli MP, Mannucci R, Pileri S, Nicoletti I. Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1: molecular basis and clinical implications. Leukemia. 2009;23:1731–1743. doi: 10.1038/leu.2009.124. [DOI] [PubMed] [Google Scholar]
  • 92.Falini B, Nicoletti I, Martelli MF, Mecucci C. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin NPMc+ AML: biologic and clinical features. Blood. 2007;109:874–885. doi: 10.1182/blood-2006-07-012252. [DOI] [PubMed] [Google Scholar]
  • 93.Bolli N, De Marco MF, Martelli MP, Bigerna B, Pucciarini A, Rossi R, Mannucci R, Manes N, Pettirossi V, Pileri SA, Nicoletti I, Falini B. A dose-dependent tug of war involving the NPM1 leukaemic mutant, nucleophosmin, and ARF. Leukemia. 2009;23:501–509. doi: 10.1038/leu.2008.326. [DOI] [PubMed] [Google Scholar]
  • 94.Angenendt L, Röllig C, Montesinos P, Martínez-Cuadrón D, Barragan E, García R, Botella C, Martínez P, Ravandi F, Kadia T, Kantarjian HM, Cortes J, Juliusson G, Lazarevic V, Höglund M, Lehmann S, Recher C, Pigneux A, Bertoli S, Dumas PY, Dombret H, Preudhomme C, Micol JB, Terré C, Ráčil Z, Novák J, Žák P, Wei AH, Tiong IS, Wall M, Estey E, Shaw C, Exeler R, Wagenführ L, Stölzel F, Thiede C, Stelljes M, Lenz G, Mikesch JH, Serve H, Ehninger G, Berdel WE, Kramer M, Krug U, Schliemann C. Chromosomal abnormalities and prognosis in NPM1-mutated acute myeloid leukemia: a pooled analysis of individual patient data from nine international cohorts. J. Clin. Oncol. 2019;37:2632–2642. doi: 10.1200/JCO.19.00416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Qi W, Shakalya K, Stejskal A, Goldman A, Beeck S, Cooke L, Mahadevan D. NSC348884, a nucleophosmin inhibitor disrupts oligomer formation and induces apoptosis in human cancer cells. Oncogene. 2008;27:4210–4220. doi: 10.1038/onc.2008.54. [DOI] [PubMed] [Google Scholar]
  • 96.Jones D, Yao H, Romans A, Dando C, Pierce S, Borthakur G, Hamilton A, Bueso-Ramos C, Ravandi F, Garcia-Manero G, Kantarjian H. Modeling interactions between leukemia-specific chromosomal changes, somatic mutations, and gene expression patterns during progression of core-binding factor leukemias. Genes Chromosomes Cancer. 2010;49:182–191. doi: 10.1002/gcc.20732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Beghini A, Ripamonti CB, Cairoli R, Cazzaniga G, Colapietro P, Elice F, Nadali G, Grillo G, Haas OA, Biondi A, Morra E, Larizza L. KIT activating mutations: incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication. Haematologica. 2004;89:920–925. [PubMed] [Google Scholar]
  • 98.Kohl TM, Schnittger S, Ellwart JW, Hiddemann W, Spiekermann K. 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;105:3319–3321. doi: 10.1182/blood-2004-06-2068. [DOI] [PubMed] [Google Scholar]
  • 99.Chen W, Xie H, Wang H, Chen L, Sun Y, Chen Z, Li Q. Prognostic significance of KIT mutations in core-binding factor acute myeloid leukemia: a systematic review and meta-analysis. PLoS One. 2016;11:e0146614. doi: 10.1371/journal.pone.0146614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Bacher U, Haferlach T, Schoch C, Kern W, Schnittger S. Implications of NRAS mutations in AML: a study of 2502 patients. Blood. 2006;107:3847–3853. doi: 10.1182/blood-2005-08-3522. [DOI] [PubMed] [Google Scholar]
  • 101.Bowen DT, Frew ME, Hills R, Gale RE, Wheatley K, Groves MJ, Langabeer SE, Kottaridis PD, Moorman AV, Burnett AK, Linch DC. RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood. 2005;106:2113–2119. doi: 10.1182/blood-2005-03-0867. [DOI] [PubMed] [Google Scholar]
  • 102.Tominaga O, Hamelin R, Remvikos Y, Salmon RJ, Thomas G. p53 from basic research to clinical applications. Crit Rev Oncog. 1992;3:257–282. [PubMed] [Google Scholar]
  • 103.Haferlach C, Dicker F, Herholz H, Schnittger S, Kern W, Haferlach T. Mutations of the TP53 gene in acute myeloid leukemia are strongly associated with a complex aberrant karyotype. Leukemia. 2008;22:1539–1541. doi: 10.1038/leu.2008.143. [DOI] [PubMed] [Google Scholar]
  • 104.Ok CY, Patel KP, Garcia-Manero G, Routbort MJ, Peng J, Tang G, Goswami M, Young KH, Singh R, Medeiros LJ, Kantarjian HM, Luthra R, Wang SA. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J Hematol Oncol. 2015;8:45. doi: 10.1186/s13045-015-0139-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hou HA, Chou WC, Kuo YY, Liu CY, Lin LI, Tseng MH, Chiang YC, Liu MC, Liu CW, Tang JL, Yao M, Li CC, Huang SY, Ko BS, Hsu SC, Chen CY, Lin CT, Wu SJ, Tsay W, Chen YC, Tien HF. TP53 mutations in de novo acute myeloid leukemia patients: longitudinal follow-ups show the mutation is stable during disease evolution. Blood Cancer J. 2015;5:e331. doi: 10.1038/bcj.2015.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Grossmann V, Schnittger S, Kohlmann A, Eder C, Roller A, Dicker F, Schmid C, Wendtner CM, Staib P, Serve H, Kreuzer KA, Kern W, Haferlach T, Haferlach C. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood. 2012;120:2963–2972. doi: 10.1182/blood-2012-03-419622. [DOI] [PubMed] [Google Scholar]
  • 107.Yang L, Han Y, Suarez Saiz F, Minden MD. A tumor suppressor and oncogene: the WT1 story. Leukemia. 2007;21:868–876. doi: 10.1038/sj.leu.2404624. [DOI] [PubMed] [Google Scholar]
  • 108.Barragan E, Cervera J, Bolufer P, Ballester S, Martín G, Fernández P, Collado R, Sayas MJ, Sanz MA. Prognostic implications of Wilms’ tumor gene WT1 expression in patients with de novo acute myeloid leukemia. Haematologica. 2004;89:926–933. [PubMed] [Google Scholar]
  • 109.Rampal R, Figueroa ME. Wilms tumor 1 mutations in the pathogenesis of acute myeloid leukemia. Haematologica. 2016;101:672–679. doi: 10.3324/haematol.2015.141796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fisher CL, Randazzo F, Humphries RK, Brock HW. Characterization of Asxl1, a murine homolog of Additional sex combs, and analysis of the Asx-like gene family. Gene. 2006;369:109–118. doi: 10.1016/j.gene.2005.10.033. [DOI] [PubMed] [Google Scholar]
  • 111.Schnittger S, Eder C, Jeromin S, Alpermann T, Fasan A, Grossmann V, Kohlmann A, Illig T, Klopp N, Wichmann HE, Kreuzer KA, Schmid C, Staib P, Peceny R, Schmitz N, Kern W, Haferlach C, Haferlach T. ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome. Leukemia. 2013;27:82–91. doi: 10.1038/leu.2012.262. [DOI] [PubMed] [Google Scholar]
  • 112.Metzeler KH, Becker H, Maharry K, Radmacher MD, Kohlschmidt J, Mrózek K, Nicolet D, Whitman SP, Wu YZ, Schwind S, Powell BL, Carter TH, Wetzler M, Moore JO, Kolitz JE, Baer MR, Carroll AJ, Larson RA, Caligiuri MA, Marcucci G, Bloomfield CD. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category. Blood. 2011;118:6920–6929. doi: 10.1182/blood-2011-08-368225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Alpermann T, Haferlach C, Eder C, Nadarajah N, Meggendorfer M, Kern W, Haferlach T, Schnittger S. AML with gain of chromosome 8 as the sole chromosomal abnormality +8sole is associated with a specific molecular mutation pattern including ASXL1 mutations in 46.8% of the patients. Leuk Res. 2015;39:265–72. doi: 10.1016/j.leukres.2014.11.026. [DOI] [PubMed] [Google Scholar]
  • 114.Larsson CA, Cote G, Quintás-Cardama A. The changing mutational landscape of acute myeloid leukemia and myelodysplastic syndrome. Mol Cancer Res. 2013;11:815–827. doi: 10.1158/1541-7786.MCR-12-0695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Yoshida K, Toki T, Okuno Y, Kanezaki R, Shiraishi Y, Sato-Otsubo A, Sanada M, Park MJ, Terui K, Suzuki H, Kon A, Nagata Y, Sato Y, Wang R, Shiba N, Chiba K, Tanaka H, Hama A, Muramatsu H, Hasegawa D, Nakamura K, Kanegane H, Tsukamoto K, Adachi S, Kawakami K, Kato K, Nishimura R, Izraeli S, Hayashi Y, Miyano S, Kojima S, Ito E, Ogawa S. The landscape of somatic mutations in Down syndrome-related myeloid disorders. Nat Genet. 2013;45:1293–1299. doi: 10.1038/ng.2759. [DOI] [PubMed] [Google Scholar]
  • 116.Roberts KG, Mullighan CG. Genomics in acute lymphoblastic leukaemia: insights and treatment implications. Nat Rev Clin Oncol. 2015;12:344–357. doi: 10.1038/nrclinonc.2015.38. [DOI] [PubMed] [Google Scholar]
  • 117.Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, Girtman K, Mathew S, Ma J, Pounds SB, Su X, Pui CH, Relling MV, Evans WE, Shurtleff SA, Downing JR. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature. 2007;446:758–764. doi: 10.1038/nature05690. [DOI] [PubMed] [Google Scholar]
  • 118.Medvedovic J, Ebert A, Tagoh H, Busslinger M. Pax5: a master regulator of B cell development and leukemogenesis. Adv Immunol. 2011;111:179–206. doi: 10.1016/B978-0-12-385991-4.00005-2. [DOI] [PubMed] [Google Scholar]
  • 119.Coyaud E, Struski S, Prade N, Familiades J, Eichner R, Quelen C, Bousquet M, Mugneret F, Talmant P, Pages MP, Lefebvre C, Penther D, Lippert E, Nadal N, Taviaux S, Poppe B, Luquet I, Baranger L, Eclache V, Radford I, Barin C, Mozziconacci MJ, Lafage-Pochitaloff M, Antoine-Poirel H, Charrin C, Perot C, Terre C, Brousset P, Dastugue N, Broccardo C. Wide diversity of PAX5 alterations in B-ALL: a Group Francophone de Cytogenetique Hematologique study. Blood. 2010;115:3089–3097. doi: 10.1182/blood-2009-07-234229. [DOI] [PubMed] [Google Scholar]
  • 120.Romana SP, Mauchauffé M, Le Coniat M, Chumakov I, Le Paslier D, Berger R, Bernard OA. The t12;21 of acute lymphoblastic leukemia results in a tel-AML1 gene fusion. Blood. 1995;85:3662–3670. [PubMed] [Google Scholar]
  • 121.Golub TR, Barker GF, Bohlander SK, Hiebert SW, Ward DC, Bray-Ward P, Morgan E, Raimondi SC, Rowley JD, Gilliland DG. Fusion of the TEL gene on 12p13 to the AML1 gene on 21q22 in acute lymphoblastic leukemia. Proc Natl Acad Sci U S A. 1995;92:4917–4921. doi: 10.1073/pnas.92.11.4917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Becker M, Liu K, Tirado CA. The t12;21p13;q22 in pediatric B-acute lymphoblastic leukemia: an update. J Assoc Genet Technol. 2017;43:198. [PubMed] [Google Scholar]
  • 123.Wang L, Hiebert SW. TEL contacts multiple co-repressors and specifically associates with histone deacetylase-3. Oncogene. 2001;20:3716–3625. doi: 10.1038/sj.onc.1204479. [DOI] [PubMed] [Google Scholar]
  • 124.Rand V, Parker H, Russell LJ, Schwab C, Ensor H, Irving J, Jones L, Masic D, Minto L, Morrison H, Ryan S, Robinson H, Sinclair P, Moorman AV, Strefford JC, Harrison CJ. Genomic characterization implicates iAMP21 as a likely primary genetic event in childhood B-cell precursor acute lymphoblastic leukemia. Blood. 2011;117:6848–6855. doi: 10.1182/blood-2011-01-329961. [DOI] [PubMed] [Google Scholar]
  • 125.Harrison CJ, Moorman AV, Schwab C, Carroll AJ, Raetz EA, Devidas M, Strehl S, Nebral K, Harbott J, Teigler-Schlegel A, Zimmerman M, Dastuge N, Baruchel A, Soulier J, Auclerc MF, Attarbaschi A, Mann G, Stark B, Cazzaniga G, Chilton L, Vandenberghe P, Forestier E, Haltrich I, Raimondi SC, Parihar M, Bourquin JP, Tchinda J, Haferlach C, Vora A, Hunger SP, Heerema NA, Haas OA Ponte di Legno International Workshop in Childhood Acute Lymphoblastic Leukemia. An international study of intrachromosomal amplification of chromosome 21 iAMP21: cytogenetic characterization and outcome. Leukemia. 2014;28:1015–1021. doi: 10.1038/leu.2013.317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Liu YF, Wang BY, Zhang WN, Huang JY, Li BS, Zhang M, Jiang L, Li JF, Wang MJ, Dai YJ, Zhang ZG, Wang Q, Kong J, Chen B, Zhu YM, Weng XQ, Shen ZX, Li JM, Wang J, Yan XJ, Li Y, Liang YM, Liu L, Chen XQ, Zhang WG, Yan JS, Hu JD, Shen SH, Chen J, Gu LJ, Pei D, Li Y, Wu G, Zhou X, Ren RB, Cheng C, Yang JJ, Wang KK, Wang SY, Zhang J, Mi JQ, Pui CH, Tang JY, Chen Z, Chen SJ. Genomic profiling of adult and pediatric B-cell acute lymphoblastic leukemia. EBioMedicine. 2016;8:173–183. doi: 10.1016/j.ebiom.2016.04.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hunger SP, Galili N, Carroll AJ, Crist WM, Link MP, Cleary ML. The t1;19q23;p13 results in consistent fusion of E2A and PBX1 coding sequences in acute lymphoblastic leukemias. Blood. 1991;77:687–693. [PubMed] [Google Scholar]
  • 128.Kamps MP, Look AT, Baltimore D. The human t1;19 translocation in pre-B ALL produces multiple nuclear E2A-Pbx1 fusion proteins with differing transforming potentials. Genes Dev. 1991;5:358–368. doi: 10.1101/gad.5.3.358. [DOI] [PubMed] [Google Scholar]
  • 129.Murre C, McCaw PS, Baltimore D. A new DNA binding and dimerization motif in immunoglobulin enhancer binding, daughterless, MyoD, and myc proteins. Cell. 1989;56:777–783. doi: 10.1016/0092-8674(89)90682-x. [DOI] [PubMed] [Google Scholar]
  • 130.Rutherford MN, LeBrun DP. Restricted expression of E2A protein in primary human tissues correlates with proliferation and differentiation. Am J Pathol. 1998;153:165–173. doi: 10.1016/S0002-9440(10)65557-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Bain G, Engel I, Robanus Maandag EC, te Riele HP, Voland JR, Sharp LL, Chun J, Huey B, Pinkel D, Murre C. E2A deficiency leads to abnormalities in alphabeta T-cell development and to rapid development of T-cell lymphomas. Mol Cell Biol. 1997;17:4782–4791. doi: 10.1128/mcb.17.8.4782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zhuang Y, Soriano P, Weintraub H. The helix-loop-helix gene E2A is required for B cell formation. Cell. 1994;79:875–884. doi: 10.1016/0092-8674(94)90076-0. [DOI] [PubMed] [Google Scholar]
  • 133.Chang CP, de Vivo I, Cleary ML. The Hox cooperativity motif of the chimeric oncoprotein E2a-Pbx1 is necessary and sufficient for oncogenesis. Mol Cell Biol. 1997;17:81–88. doi: 10.1128/mcb.17.1.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Selleri L, Depew MJ, Jacobs Y, Chanda SK, Tsang KY, Cheah KS, Rubenstein JL, O’Gorman S, Cleary ML. Requirement for Pbx1 in skeletal patterning and programming chondrocyte proliferation and differentiation. Development. 2001;128:3543–3557. doi: 10.1242/dev.128.18.3543. [DOI] [PubMed] [Google Scholar]
  • 135.Van Dijk MA, Voorhoeve PM, Murre C. Pbx1 is converted into a transcriptional activator upon acquiring the N-terminal region of E2A in pre-B-cell acute lymphoblastoid leukemia. Proc Natl Acad Sci U S A. 1993;90:6061–6065. doi: 10.1073/pnas.90.13.6061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.LeBrun DP. E2A basic helix-loop-helix transcription factors in human leukemia. Front Biosci. 2003;8:206–22. doi: 10.2741/1030. [DOI] [PubMed] [Google Scholar]
  • 137.Smith KS, Chanda SK, Lingbeek M, Ross DT, Botstein D, van Lohuizen M, Cleary ML. Bmi-1 regulation of INK4A-ARF is a downstream requirement for transformation of hematopoietic progenitors by E2a-Pbx1. Mol Cell. 2003;12:393–400. doi: 10.1016/s1097-2765(03)00277-6. [DOI] [PubMed] [Google Scholar]
  • 138.Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, Morrison SJ, Clarke MF. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003;423:302–305. doi: 10.1038/nature01587. [DOI] [PubMed] [Google Scholar]
  • 139.Chin L, Pomerantz J, DePinho RA. The INK4a/ARF tumor suppressor: one gene--two products--two pathways. Trends Biochem Sci. 1998;23:291–296. doi: 10.1016/s0968-0004(98)01236-5. [DOI] [PubMed] [Google Scholar]
  • 140.Dyson N. The regulation of E2F by pRB-family proteins. Genes Dev. 1998;12:2245–2262. doi: 10.1101/gad.12.15.2245. [DOI] [PubMed] [Google Scholar]
  • 141.Talluri S, Dick FA. Regulation of transcription and chromatin structure by pRB: here, there and everywhere. Cell Cycle. 2012;11:3189–3198. doi: 10.4161/cc.21263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Raimondi SC, Privitera E, Williams DL, Look AT, Behm F, Rivera GK, Crist WM, Pui CH. New recurring chromosomal translocations in childhood acute lymphoblastic leukemia. Blood. 1991;77:2016–2022. [PubMed] [Google Scholar]
  • 143.Inaba T, Roberts WM, Shapiro LH, Jolly KW, Raimondi SC, Smith SD, Look AT. Fusion of the leucine zipper gene HLF to the E2A gene in human acute B-lineage leukemia. Science. 1992;257:531–534. doi: 10.1126/science.1386162. [DOI] [PubMed] [Google Scholar]
  • 144.Yoshihara T, Inaba T, Shapiro LH, Kato JY, Look AT. E2A-HLF-mediated cell transformation requires both the trans-activation domains of E2A and the leucine zipper dimerization domain of HLF. Mol Cell Biol. 1995;15:3247–3255. doi: 10.1128/mcb.15.6.3247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Inaba T, Shapiro LH, Funabiki T, Sinclair AE, Jones BG, Ashmun RA, Look AT. DNA-binding specificity and trans-activating potential of the leukemia-associated E2A-hepatic leukemia factor fusion protein. Mol Cell Biol. 1994;14:3403–3413. doi: 10.1128/mcb.14.5.3403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J, White D, Hughes TP, Le Beau MM, Pui CH, Relling MV, Shurtleff SA, Downing JR. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453:110–114. doi: 10.1038/nature06866. [DOI] [PubMed] [Google Scholar]
  • 147.Ng SY, Yoshida T, Zhang J, Georgopoulos K. Genome-wide lineage-specific transcriptional networks underscore Ikaros-dependent lymphoid priming in hematopoietic stem cells. Immunity. 2009;30:493–507. doi: 10.1016/j.immuni.2009.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Iacobucci I, Iraci N, Messina M, Lonetti A, Chiaretti S, Valli E, Ferrari A, Papayannidis C, Paoloni F, Vitale A, Storlazzi CT, Ottaviani E, Guadagnuolo V, Durante S, Vignetti M, Soverini S, Pane F, Foa R, Baccarani M, Müschen M, Perini G, Martinelli G. IKAROS deletions dictate a unique gene expression signature in patients with adult B-cell acute lymphoblastic leukemia. PLoS One. 2012;7:e40934. doi: 10.1371/journal.pone.0040934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Mullighan CG, Su X, Zhang J, Radtke I, Phillips LA, Miller CB, Ma J, Liu W, Cheng C, Schulman BA, Harvey RC, Chen IM, Clifford RJ, Carroll WL, Reaman G, Bowman WP, Devidas M, Gerhard DS, Yang W, Relling MV, Shurtleff SA, Campana D, Borowitz MJ, Pui CH, Smith M, Hunger SP, Willman CL, Downing JR Children’s Oncology Group. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360:470–480. doi: 10.1056/NEJMoa0808253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Roberts KG, Morin RD, Zhang J, Hirst M, Zhao Y, Su X, Chen SC, Payne-Turner D, Churchman ML, Harvey RC, Chen X, Kasap C, Yan C, Becksfort J, Finney RP, Teachey DT, Maude SL, Tse K, Moore R, Jones S, Mungall K, Birol I, Edmonson MN, Hu Y, Buetow KE, Chen IM, Carroll WL, Wei L, Ma J, Kleppe M, Levine RL, Garcia-Manero G, Larsen E, Shah NP, Devidas M, Reaman G, Smith M, Paugh SW, Evans WE, Grupp SA, Jeha S, Pui CH, Gerhard DS, Downing JR, Willman CL, Loh M, Hunger SP, Marra MA, Mullighan CG. Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia. Cancer Cell. 2012;22:153–166. doi: 10.1016/j.ccr.2012.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Secker-Walker LM, Craig JM, Hawkins JM, Hoffbrand AV. Philadelphia positive acute lymphoblastic leukemia in adults: age distribution, BCR breakpoint and prognostic significance. Leukemia. 1991;5:196–199. [PubMed] [Google Scholar]
  • 152.Bartram CR, de Klein A, Hagemeijer A, van Agthoven T, Geurts van Kessel A, Bootsma D, Grosveld G, Ferguson-Smith MA, Davies T, Stone M. Translocation of c-ab1 oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukaemia. Nature. 1983;306:277–280. doi: 10.1038/306277a0. [DOI] [PubMed] [Google Scholar]
  • 153.Groffen J, Stephenson JR, Heisterkamp N, de Klein A, Bartram CR, Grosveld G. Philadelphia chromosomal breakpoints are clustered within a limited region, bcr, on chromosome 22. Cell. 1984;36:93–99. doi: 10.1016/0092-8674(84)90077-1. [DOI] [PubMed] [Google Scholar]
  • 154.Kurzrock R, Gutterman JU, Talpaz M. The molecular genetics of Philadelphia chromosome-positive leukemias. N Engl J Med. 1988;319:990–998. doi: 10.1056/NEJM198810133191506. [DOI] [PubMed] [Google Scholar]
  • 155.Kurzrock R, Shtalrid M, Romero P, Kloetzer WS, Talpas M, Trujillo JM, Blick M, Beran M, Gutterman JU. A novel c-abl protein product in Philadelphia-positive acute lymphoblastic leukaemia. Nature. 1987;325:631–635. doi: 10.1038/325631a0. [DOI] [PubMed] [Google Scholar]
  • 156.Voncken JW, Kaartinen V, Pattengale PK, Germeraad WT, Groffen J, Heisterkamp N. BCR/ABL P210 and P190 cause distinct leukemia in transgenic mice. Blood. 1995;86:4603–4611. [PubMed] [Google Scholar]
  • 157.Sattler M, Griffin JD. Mechanisms of transformation by the BCR/ABL oncogene. Int J Hematol. 2001;73:278–291. doi: 10.1007/BF02981952. [DOI] [PubMed] [Google Scholar]
  • 158.de Labarthe A, Rousselot P, Huguet-Rigal F, Delabesse E, Witz F, Maury S, Réa D, Cayuela JM, Vekemans MC, Reman O, Buzyn A, Pigneux A, Escoffre M, Chalandon Y, MacIntyre E, Lhéritier V, Vernant JP, Thomas X, Ifrah N, Dombret H Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) Imatinib combined with induction or consolidation chemotherapy in patients with de novo Philadelphia chromosome-positive acute lymphoblastic leukemia: results of the GRAAPH-2003 study. Blood. 2007;109:1408–1413. doi: 10.1182/blood-2006-03-011908. [DOI] [PubMed] [Google Scholar]
  • 159.Yanada M, Naoe T. Imatinib combined chemotherapy for Philadelphia chromosome-positive acute lymphoblastic leukemia: major challenges in current practice. Leuk Lymphoma. 2006;47:1747–1753. doi: 10.1080/10428190600634085. [DOI] [PubMed] [Google Scholar]
  • 160.Harrison CJ. Key pathways as therapeutic targets. Blood. 2011;118:2935–2936. doi: 10.1182/blood-2011-07-362723. [DOI] [PubMed] [Google Scholar]
  • 161.Case M, Matheson E, Minto L, Hassan R, Harrison CJ, Bown N, Bailey S, Vormoor J, Hall AG, Irving JA. Mutation of genes affecting the RAS pathway is common in childhood acute lymphoblastic leukemia. Cancer Res. 2008;68:6803–6809. doi: 10.1158/0008-5472.CAN-08-0101. [DOI] [PubMed] [Google Scholar]
  • 162.Tartaglia M, Martinelli S, Cazzaniga G, Cordeddu V, Iavarone I, Spinelli M, Palmi C, Carta C, Pession A, Aricò M, Masera G, Basso G, Sorcini M, Gelb BD, Biondi A. Genetic evidence for lineage-related and differentiation stage-related contribution of somatic PTPN11 mutations to leukemogenesis in childhood acute leukemia. Blood. 2004;104:307–313. doi: 10.1182/blood-2003-11-3876. [DOI] [PubMed] [Google Scholar]
  • 163.Wiemels JL, Zhang Y, Chang J, Zheng S, Metayer C, Zhang L, Smith MT, Ma X, Selvin S, Buffler PA, Wiencke JK. RAS mutation is associated with hyperdiploidy and parental characteristics in pediatric acute lymphoblastic leukemia. Leukemia. 2005;19:415–419. doi: 10.1038/sj.leu.2403641. [DOI] [PubMed] [Google Scholar]
  • 164.Zhang J, Mullighan CG, Harvey RC, Wu G, Chen X, Edmonson M, Buetow KH, Carroll WL, Chen IM, Devidas M, Gerhard DS, Loh ML, Reaman GH, Relling MV, Camitta BM, Bowman WP, Smith MA, Willman CL, Downing JR, Hunger SP. Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: a report from the Children’s Oncology Group. Blood. 2011;118:3080–3087. doi: 10.1182/blood-2011-03-341412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Paulsson K, Horvat A, Strömbeck B, Nilsson F, Heldrup J, Behrendtz M, Forestier E, Andersson A, Fioretos T, Johansson B. Mutations of FLT3, NRAS, KRAS, and PTPN11 are frequent and possibly mutually exclusive in high hyperdiploid childhood acute lymphoblastic leukemia. Genes Chromosomes Cancer. 2008;47:26–33. doi: 10.1002/gcc.20502. [DOI] [PubMed] [Google Scholar]
  • 166.Perentesis JP, Bhatia S, Boyle E, Shao Y, Shu XO, Steinbuch M, Sather HN, Gaynon P, Kiffmeyer W, Envall-Fox J, Robison LL. RAS oncogene mutations and outcome of therapy for childhood acute lymphoblastic leukemia. Leukemia. 2004;18:685–692. doi: 10.1038/sj.leu.2403272. [DOI] [PubMed] [Google Scholar]
  • 167.Pui CH, Relling MV, Downing JR. Acute lymphoblastic leukemia. N Engl J Med. 2004;15:1535–1548. doi: 10.1056/NEJMra023001. [DOI] [PubMed] [Google Scholar]
  • 168.Lee JW, Soung YH, Park WS, Kim SY, Nam SW, Min WS, Lee JY, Yoo NJ, Lee SH. BRAF mutations in acute leukemias. Leukemia. 2004;18:170–172. doi: 10.1038/sj.leu.2403201. [DOI] [PubMed] [Google Scholar]
  • 169.Gustafsson B, Angelini S, Sander B, Christensson B, Hemminki K, Kumar R. Mutations in the BRAF and N-ras genes in childhood acute lymphoblastic leukaemia. Leukemia. 2005;19:310–312. doi: 10.1038/sj.leu.2403589. [DOI] [PubMed] [Google Scholar]
  • 170.Mullighan CG. Genomic profiling of B-progenitor acute lymphoblastic leukemia. Best Pract Res Clin Haematol. 2011;24:489–503. doi: 10.1016/j.beha.2011.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Mullighan CG, Zhang J, Harvey RC, Collins-Underwood JR, Schulman BA, Phillips LA, Tasian SK, Loh ML, Su X, Liu W, Devidas M, Atlas SR, Chen IM, Clifford RJ, Gerhard DS, Carroll WL, Reaman GH, Smith M, Downing JR, Hunger SP, Willman CL. JAK mutations in high-risk childhood acute lymphoblastic leukemia. Proc Natl Acad Sci U S A. 2009;106:9414–9418. doi: 10.1073/pnas.0811761106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Roberts KG, Mullighan CG. How new advances in genetic analysis are influencing the understanding and treatment of childhood acute leukemia. Curr Opin Pediatr. 2011;23:34–40. doi: 10.1097/MOP.0b013e3283426260. [DOI] [PubMed] [Google Scholar]
  • 173.Bercovich D, Ganmore I, Scott LM, Wainreb G, Birger Y, Elimelech A, Shochat C, Cazzaniga G, Biondi A, Basso G, Cario G, Schrappe M, Stanulla M, Strehl S, Haas OA, Mann G, Binder V, Borkhardt A, Kempski H, Trka J, Bielorei B, Avigad S, Stark B, Smith O, Dastugue N, Bourquin JP, Tal NB, Green AR, Izraeli S. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down’s syndrome. Lancet. 2008;372:1484–1492. doi: 10.1016/S0140-6736(08)61341-0. [DOI] [PubMed] [Google Scholar]
  • 174.Mullighan CG. JAK2--a new player in acute lymphoblastic leukaemia. Lancet. 2008;372:1448–1450. doi: 10.1016/S0140-6736(08)61342-2. [DOI] [PubMed] [Google Scholar]
  • 175.Ding LW, Sun QY, Tan KT, Chien W, Mayakonda A, Yeoh AEJ, Kawamata N, Nagata Y, Xiao JF, Loh XY, Lin DC, Garg M, Jiang YY, Xu L, Lim SL, Liu LZ, Madan V, Sanada M, Fernández LT, Hema Preethi SS, Lill M, Kantarjian HM, Kornblau SM, Miyano S, Liang DC, Ogawa S, Shih LY, Yang H, Koeffler HP. Mutational landscape of pediatric acute lymphoblastic leukemia. Cancer Res. 2017;77:390–400. doi: 10.1158/0008-5472.CAN-16-1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Qian M, Cao X, Devidas M, Yang W, Cheng C, Dai Y, Carroll A, Heerema NA, Zhang H, Moriyama T, Gastier-Foster JM, Xu H, Raetz E, Larsen E, Winick N, Bowman WP, Martin PL, Mardis ER, Fulton R, Zambetti G, Borowitz M, Wood B, Nichols KE, Carroll WL, Pui CH, Mullighan CG, Evans WE, Hunger SP, Relling MV, Loh ML, Yang JJ. TP53 germline variations influence the predisposition and prognosis of B-cell acute lymphoblastic leukemia in Children. J. Clin. Oncol. 2018;36:591–599. doi: 10.1200/JCO.2017.75.5215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hof J, Krentz S, van Schewick C, Körner G, Shalapour S, Rhein P, Karawajew L, Ludwig WD, Seeger K, Henze G, von Stackelberg A, Hagemeier C, Eckert C, Kirschner-Schwabe R. Mutations and deletions of the TP53 gene predict nonresponse to treatment and poor outcome in first relapse of childhood acute lymphoblastic leukemia. J. Clin. Oncol. 2011;29:3185–3193. doi: 10.1200/JCO.2011.34.8144. [DOI] [PubMed] [Google Scholar]
  • 178.Zeidler L, Zimmermann M, Möricke A, Meissner B, Bartels D, Tschan C, Schrauder A, Cario G, Goudeva L, Jäger S, Ratei R, Ludwig WD, Teigler-Schlegel A, Skokowa J, Koehler R, Bartram CR, Riehm H, Schrappe M, Welte K, Stanulla M. Low platelet counts after induction therapy for childhood acute lymphoblastic leukemia are strongly associated with poor early response to treatment as measured by minimal residual disease and are prognostic for treatment outcome. Haematologica. 2012;97:402–409. doi: 10.3324/haematol.2011.045229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Lambert JM, Gorzov P, Veprintsev DB, Söderqvist M, Segerbäck D, Bergman J, Fersht AR, Hainaut P, Wiman KG, Bykov VJ. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009;15:376–388. doi: 10.1016/j.ccr.2009.03.003. [DOI] [PubMed] [Google Scholar]
  • 180.Washburn T, Schweighoffer E, Gridley T, Chang D, Fowlkes BJ, Cado D, Robey E. Notch activity influences the alpha beta versus gamma delta T cell lineage decision. Cell. 1997;88:833–843. doi: 10.1016/s0092-8674(00)81929-7. [DOI] [PubMed] [Google Scholar]
  • 181.Sambandam A, Maillard I, Zediak VP, Xu L, Gerstein RM, Aster JC, Pear WS, Bhandoola A. Notch signaling controls the generation and differentiation of early T lineage progenitors. Nat Immunol. 2005;6:663–670. doi: 10.1038/ni1216. [DOI] [PubMed] [Google Scholar]
  • 182.Pui JC, Allman D, Xu L, DeRocco S, Karnell FG, Bakkour S, Lee JY, Kadesch T, Hardy RR, Aster JC, Pear WS. Notch1 expression in early lymphopoiesis influences B versus T lineage determination. Immunity. 1999;11:299–308. doi: 10.1016/s1074-7613(00)80105-3. [DOI] [PubMed] [Google Scholar]
  • 183.Ellisen LW, Bird J, West DC, Soreng AL, Reynolds TC, Smith SD, Sklar J. TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell. 1991;66:649–661. doi: 10.1016/0092-8674(91)90111-b. [DOI] [PubMed] [Google Scholar]
  • 184.Weng AP, Ferrando AA, Lee W, Morris JP 4th, Silverman LB, Sanchez-Irizarry C, Blacklow SC, Look AT, Aster JC. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004;306:269–271. doi: 10.1126/science.1102160. [DOI] [PubMed] [Google Scholar]
  • 185.Aster JC, Pear WS, Blacklow SC. Notch signaling in leukemia. Annu Rev Pathol. 2008;3:587–613. doi: 10.1146/annurev.pathmechdis.3.121806.154300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Roy M, Pear WS, Aster JC. The multifaceted role of Notch in cancer. Curr Opin Genet Dev. 2007;17:52–59. doi: 10.1016/j.gde.2006.12.001. [DOI] [PubMed] [Google Scholar]
  • 187.Palomero T, Lim WK, Odom DT, Sulis ML, Real PJ, Margolin A, Barnes KC, O’Neil J, Neuberg D, Weng AP, Aster JC, Sigaux F, Soulier J, Look AT, Young RA, Califano A, Ferrando AA. NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth. Proc Natl Acad Sci U S A. 2006;103:18261–18266. doi: 10.1073/pnas.0606108103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Sharma VM, Calvo JA, Draheim KM, Cunningham LA, Hermance N, Beverly L, Krishnamoorthy V, Bhasin M, Capobianco AJ, Kelliher MA. Notch1 contributes to mouse T-cell leukemia by directly inducing the expression of c-myc. Mol Cell Biol. 2006;26:8022–8031. doi: 10.1128/MCB.01091-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Weng AP, Millholland JM, Yashiro-Ohtani Y, Arcangeli ML, Lau A, Wai C, Del Bianco C, Rodriguez CG, Sai H, Tobias J, Li Y, Wolfe MS, Shachaf C, Felsher D, Blacklow SC, Pear WS, Aster JC. c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. Genes Dev. 2006;20:2096–2109. doi: 10.1101/gad.1450406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Real PJ, Tosello V, Palomero T, Castillo M, Hernando E, de Stanchina E, Sulis ML, Barnes K, Sawai C, Homminga I, Meijerink J, Aifantis I, Basso G, Cordon-Cardo C, Ai W, Ferrando A. Gamma-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia. Nat Med. 2009;15:50–58. doi: 10.1038/nm.1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.De Keersmaecker K, Marynen P, Cools J. Genetic insights in the pathogenesis of T-cell acute lymphoblastic leukemia. Haematologica. 2005;90:1116–1127. [PubMed] [Google Scholar]
  • 192.Wadman I, Li J, Bash RO, Forster A, Osada H, Rabbitts TH, Baer R. Specific in vivo association between the bHLH and LIM proteins implicated in human T cell leukemia. EMBO J. 1994;13:4831–4839. doi: 10.1002/j.1460-2075.1994.tb06809.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Cavé H, Suciu S, Preudhomme C, Poppe B, Robert A, Uyttebroeck A, Malet M, Boutard P, Benoit Y, Mauvieux L, Lutz P, Méchinaud F, Grardel N, Mazingue F, Dupont M, Margueritte G, Pages MP, Bertrand Y, Plouvier E, Brunie G, Bastard C, Plantaz D, Vande Velde I, Hagemeijer A, Speleman F, Lessard M, Otten J, Vilmer E, Dastugue N EORTC-CLG. Clinical significance of HOX11L2 expression linked to t5;14q35;q32, of HOX11 expression, and of SIL-TAL fusion in childhood T-cell malignancies: results of EORTC studies 58881 and 58951. Blood. 2004;103:442–450. doi: 10.1182/blood-2003-05-1495. [DOI] [PubMed] [Google Scholar]
  • 194.Park ST, Sun XH. The Tal1 oncoprotein inhibits E47-mediated transcription. Mechanism of inhibition. J Biol Chem. 1998;273:7030–7037. doi: 10.1074/jbc.273.12.7030. [DOI] [PubMed] [Google Scholar]
  • 195.O’Neil J, Shank J, Cusson N, Murre C, Kelliher M. TAL1/SCL induces leukemia by inhibiting the transcriptional activity of E47/HEB. Cancer Cell. 2004;5:587–596. doi: 10.1016/j.ccr.2004.05.023. [DOI] [PubMed] [Google Scholar]
  • 196.Carroll AJ, Crist WM, Link MP, Amylon MD, Pullen DJ, Ragab AH, Buchanan GR, Wimmer RS, Vietti TJ. The t1;14p34;q11 is nonrandom and restricted to T-cell acute lymphoblastic leukemia: a Pediatric Oncology Group study. Blood. 1990;76:1220–1224. [PubMed] [Google Scholar]
  • 197.Cheng T, Shen H, Giokas D, Gere J, Tenen DG, Scadden DT. Temporal mapping of gene expression levels during the differentiation of individual primary hematopoietic cells. Proc Natl Acad Sci U S A. 1996;93:13158–13163. doi: 10.1073/pnas.93.23.13158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Robb L, Lyons I, Li R, Hartley L, Köntgen F, Harvey RP, Metcalf D, Begley CG. Absence of yolk sac hematopoiesis from mice with a targeted disruption of the scl gene. Proc Natl Acad Sci U S A. 1995;92:7075–7079. doi: 10.1073/pnas.92.15.7075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Shivdasani RA, Mayer EL, Orkin SH. Absence of blood formation in mice lacking the T-cell leukaemia oncoprotein tal-1/SCL. Nature. 1995;373:432–434. doi: 10.1038/373432a0. [DOI] [PubMed] [Google Scholar]
  • 200.Xia Y, Hwang LY, Cobb MH, Baer R. Products of the TAL2 oncogene in leukemic T cells: bHLH phosphoproteins with DNA-binding activity. Oncogene. 1994;9:1437–1446. [PubMed] [Google Scholar]
  • 201.Harrison CJ. Cytogenetics of paediatric and adolescent acute lymphoblastic leukaemia. Br J Haematol. 2009;144:147–156. doi: 10.1111/j.1365-2141.2008.07417.x. [DOI] [PubMed] [Google Scholar]
  • 202.Ferrando AA, Neuberg DS, Staunton J, Loh ML, Huard C, Raimondi SC, Behm FG, Pui CH, Downing JR, Gilliland DG, Lander ES, Golub TR, Look AT. Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer Cell. 2002;1:75–87. doi: 10.1016/s1535-6108(02)00018-1. [DOI] [PubMed] [Google Scholar]
  • 203.Ferrando AA, Herblot S, Palomero T, Hansen M, Hoang T, Fox EA, Look AT. Biallelic transcriptional activation of oncogenic transcription factors in T-cell acute lymphoblastic leukemia. Blood. 2004;103:1909–1911. doi: 10.1182/blood-2003-07-2577. [DOI] [PubMed] [Google Scholar]
  • 204.Ferrando AA, Neuberg DS, Dodge RK, Paietta E, Larson RA, Wiernik PH, Rowe JM, Caligiuri MA, Bloomfield CD, Look AT. Prognostic importance of TLX1 HOX11 oncogene expression in adults with T-cell acute lymphoblastic leukaemia. Lancet. 2004;363:535–536. doi: 10.1016/S0140-6736(04)15542-6. [DOI] [PubMed] [Google Scholar]
  • 205.Dadi S, Le Noir S, Payet-Bornet D, Lhermitte L, Zacarias-Cabeza J, Bergeron J, Villarèse P, Vachez E, Dik WA, Millien C, Radford I, Verhoeyen E, Cosset FL, Petit A, Ifrah N, Dombret H, Hermine O, Spicuglia S, Langerak AW, Macintyre EA, Nadel B, Ferrier P, Asnafi V. TLX homeodomain oncogenes mediate T cell maturation arrest in T-ALL via interaction with ETS1 and suppression of TCRα gene expression. Cancer Cell. 2012;21:563–576. doi: 10.1016/j.ccr.2012.02.013. [DOI] [PubMed] [Google Scholar]
  • 206.De Keersmaecker K, Ferrando AA. TLX1-induced T-cell acute lymphoblastic leukemia. Clin Cancer Res. 2011;17:6381–6386. doi: 10.1158/1078-0432.CCR-10-3037. [DOI] [PubMed] [Google Scholar]
  • 207.Cauwelier B, Dastugue N, Cools J, Poppe B, Herens C, De Paepe A, Hagemeijer A, Speleman F. Molecular cytogenetic study of 126 unselected T-ALL cases reveals high incidence of TCRbeta locus rearrangements and putative new T-cell oncogenes. Leukemia. 2006;20:1238–1244. doi: 10.1038/sj.leu.2404243. [DOI] [PubMed] [Google Scholar]
  • 208.Su XY, Della-Valle V, Andre-Schmutz I, Lemercier C, Radford-Weiss I, Ballerini P, Lessard M, Lafage-Pochitaloff M, Mugneret F, Berger R, Romana SP, Bernard OA, Penard-Lacronique V. HOX11L2/TLX3 is transcriptionally activated through T-cell regulatory elements downstream of BCL11B as a result of the t5;14q35;q32. Blood. 2006;108:4198–4201. doi: 10.1182/blood-2006-07-032953. [DOI] [PubMed] [Google Scholar]
  • 209.Bernard OA, Busson-LeConiat M, Ballerini P, Mauchauffé M, Della Valle V, Monni R, Nguyen Khac F, Mercher T, Penard-Lacronique V, Pasturaud P, Gressin L, Heilig R, Daniel MT, Lessard M, Berger R. A new recurrent and specific cryptic translocation, t5;14q35;q32, is associated with expression of the Hox11L2 gene in T acute lymphoblastic leukemia. Leukemia. 2001;15:1495–1504. doi: 10.1038/sj.leu.2402249. [DOI] [PubMed] [Google Scholar]
  • 210.Cauwelier B, Cavé H, Gervais C, Lessard M, Barin C, Perot C, Van den Akker J, Mugneret F, Charrin C, Pagès MP, Grégoire MJ, Jonveaux P, Lafage-Pochitaloff M, Mozzicconacci MJ, Terré C, Luquet I, Cornillet-Lefebvre P, Laurence B, Plessis G, Lefebvre C, Leroux D, Antoine-Poirel H, Graux C, Mauvieux L, Heimann P, Chalas C, Clappier E, Verhasselt B, Benoit Y, Moerloose BD, Poppe B, Van Roy N, Keersmaecker KD, Cools J, Sigaux F, Soulier J, Hagemeijer A, Paepe AD, Dastugue N, Berger R, Speleman F. Clinical, cytogenetic and molecular characteristics of 14 T-ALL patients carrying the TCRbeta-HOXA rearrangement: a study of the Groupe Francophone de Cytogénétique Hématologique. Leukemia. 2007;21:121–128. doi: 10.1038/sj.leu.2404410. [DOI] [PubMed] [Google Scholar]
  • 211.Speleman F, Cauwelier B, Dastugue N, Cools J, Verhasselt B, Poppe B, Van Roy N, Vandesompele J, Graux C, Uyttebroeck A, Boogaerts M, De Moerloose B, Benoit Y, Selleslag D, Billiet J, Robert A, Huguet F, Vandenberghe P, De Paepe A, Marynen P, Hagemeijer A. A new recurrent inversion, inv7p15q34, leads to transcriptional activation of HOXA10 and HOXA11 in a subset of T-cell acute lymphoblastic leukemias. Leukemia. 2005;19:358–366. doi: 10.1038/sj.leu.2403657. [DOI] [PubMed] [Google Scholar]
  • 212.Bergeron J, Clappier E, Cauwelier B, Dastugue N, Millien C, Delabesse E, Beldjord K, Speleman F, Soulier J, Macintyre E, Asnafi V. HOXA cluster deregulation in T-ALL associated with both a TCRD-HOXA and a CALM-AF10 chromosomal translocation. Leukemia. 2006;20:1184–1187. doi: 10.1038/sj.leu.2404187. [DOI] [PubMed] [Google Scholar]
  • 213.O’Neil J, Tchinda J, Gutierrez A, Moreau L, Maser RS, Wong KK, Li W, McKenna K, Liu XS, Feng B, Neuberg D, Silverman L, DeAngelo DJ, Kutok JL, Rothstein R, DePinho RA, Chin L, Lee C, Look AT. Alu elements mediate MYB gene tandem duplication in human T-ALL. J Exp Med. 2007;204:3059–3066. doi: 10.1084/jem.20071637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Lahortiga I, De Keersmaecker K, Van Vlierberghe P, Graux C, Cauwelier B, Lambert F, Mentens N, Beverloo HB, Pieters R, Speleman F, Odero MD, Bauters M, Froyen G, Marynen P, Vandenberghe P, Wlodarska I, Meijerink JP, Cools J. Duplication of the MYB oncogene in T cell acute lymphoblastic leukemia. Nat Genet. 2007;39:593–595. doi: 10.1038/ng2025. [DOI] [PubMed] [Google Scholar]
  • 215.Gutierrez A, Kentsis A, Sanda T, Holmfeldt L, Chen SC, Zhang J, Protopopov A, Chin L, Dahlberg SE, Neuberg DS, Silverman LB, Winter SS, Hunger SP, Sallan SE, Zha S, Alt FW, Downing JR, Mullighan CG, Look AT. The BCL11B tumor suppressor is mutated across the major molecular subtypes of T-cell acute lymphoblastic leukemia. Blood. 2011;118:4169–4173. doi: 10.1182/blood-2010-11-318873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Li L, Leid M, Rothenberg EV. An early T cell lineage commitment checkpoint dependent on the transcription factor Bcl11b. Science. 2010;329:89–93. doi: 10.1126/science.1188989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Van Vlierberghe P, Palomero T, Khiabanian H, Van der Meulen J, Castillo M, Van Roy N, De Moerloose B, Philippé J, González-García S, Toribio ML, Taghon T, Zuurbier L, Cauwelier B, Harrison CJ, Schwab C, Pisecker M, Strehl S, Langerak AW, Gecz J, Sonneveld E, Pieters R, Paietta E, Rowe JM, Wiernik PH, Benoit Y, Soulier J, Poppe B, Yao X, Cordon-Cardo C, Meijerink J, Rabadan R, Speleman F, Ferrando A. PHF6 mutations in T-cell acute lymphoblastic leukemia. Nat Genet. 2010;42:338–342. doi: 10.1038/ng.542. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Blood Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES