Frequency of numerical and structural changes among B-ALL patients of different cohorts
1. Introduction
Acute leukemia is a broad term used to identify several malignancies of immature hematopoietic cells. Although, variable incidences have been reported between countries, ranging from 46 to 57 cases by million children, it is considered the most common childhood cancer worldwide [1]. Acute lymphoblastic leukemia (ALL) is the most frequent subtype (75%-80% of cases; with the remaining 20-25% being of myeloid origin, AML). In ALL, B cell origin is the most frequently diagnosed (B cell ALL) representing 83%, and T cell ALL comprises 15% [2]. The total of ALL cases represents 30-40% of all types of pediatric cancer[3].
One of the major achievements in cancer therapy has been the increased cure rates for ALL, from 10% in the 60s to 76-86% today, although these favorable numbers are mainly valid for developed countries[4,5]. The improvement in ALL cure rates can be in part attributed to the assessment of conventional prognostic factors and identification of molecular markers associated with a better response to therapy. Suitable risk stratification has permitted a more personalized treatment, selecting patients for receiving standard or intensified therapy, alone or in combination with drugs against ALL specific targets, and together with an enhanced supportive care have contributed to the increase in the event-free survival (EFS) rates[4]. Conventional childhood ALL stratification is based on prognostic factors related to characteristics of the patient (age at diagnosis) and the disease itself white blood cell (WBC) count at diagnosis, immunophenotype of the leukemic cells, presence of known genetic fusions, numerical abnormalities or abnormal gene expression, and early response to therapy (evaluated by morphological methods or using a more accurate measurement such as minimal residual disease (MRD) analysis) [4–6].
From a genetic point of view, ALL is one of the best characterized malignancies. Numerical and structural chromosomal abnormalities have been described by cytogenetic methods, fluorescence
2. Signaling and transcription factors important in lymphopoiesis and leukemogenesis
Generation of lymphoid cells is a highly ordered multi-step process that in adult mammals starts in bone marrow with the differentiation of multipotent hematopoietic stem cells (HSC) (Figure 1A). HSCs start a differentiation pathway in which the capacity to form multiple lineages is gradually lost coinciding with a gain of lineage specific functions. Thus, HSCs yield multipotent progenitor cells (MPPs) still with myeloid and lymphoid potential, which eventually give rise to lymphoid-primed multipotent progenitor (LMPP) and early lymphoid progenitor (ELP) populations, with a progressive more restrictive lymphoid program. Similarly, ELPs generate early T lineage progenitors (ETP) and common lymphoid progenitors (CLP), and these populations, although still exhibit high plasticity, preferentially give rise
B and T cells are characterized by their potential to express receptors with a highly diverse repertoire of specificities: the B and T cell receptors (BCR and TCR). This diverse specificity is given by a recombination process termed VDJ recombination and it is the sequential assembly and testing of the BCR and TCR what defines the B/T development pathway. The first stages (pro and pre) are characterized by recombination of the antigen binding variable sequences (heavy and light chains for the BCR, and the β, α, γ or δ chains for the TCR) (Figure 1B). The subsequent stages require elimination of auto-reactive clones, and only clones selected against self-recognition become functional mature cells. Genetic and biochemical studies have shown that all forms of the BCR and TCR are required for progression through several defined developmental checkpoints [13,14]. This is an important concept, since it illustrates that different signaling and transcription programs are operating through all developmental stages, and therefore, if an aberrant program is established, development is unable to proceed. As we will see in the following sections, the leukemic gene fusions and other genetic abnormalities produce aberrant signaling pathways or abnormal transcriptional activities, leading to a developmental arrest in specific stages, events that seem to be required and characterize B and T cell ALL.

Figure 1.
Early developmental stages are the ones generally found compromised in human pediatric B and T cell ALL. These stages in B cell ALL are early proB or pre-proB (before heavy-chain recombination), preB-I (after heavy-chain recombination), and preB-II (before light-chain rearrangement) (Figure 1B). These stages are also recognized by the expression of stage specific markers, a characteristic that has helped to classify the different types of pediatric ALL. B cells are recognized by the expression of CD19 and CD10, common B cell ALL by the expression of the BCR (IgM) either in cytoplasm (preB-I) or membrane (preB-II), and preB-I can also be differentiated from preB-II cells by expression of the enzyme terminal deoxynucleotidyl transferase (TdT) [10].
T cells are recognized by the expression of CD3, CD5 and CD7. Early T cells lack expression of CD4 and CD8 (double negative or DN stages). Contrary to B cell ALL, T cell ALL clones often express markers of more advanced stages of development (for instance double positive stages). However, these clones also show a lack of expression or cytoplasmic TCRβ, indicating that transformation happened before rearrangement of this TCR component or just after, and thus arguing that transformation targeted ETP/DN1 or DN3´cells [15]. The acquisition of markers of more mature cells is probably due to marker aberrant expression or leukemia-induced developmental progression in absence of the TCR signal. Although, postnatal B cell early maturation only happens in bone marrow, T cells mature in thymus. LMPP, ELP, CLP and ETP cells are all able to leave bone marrow in response to environmental signals and complete the T cell maturation program in thymus. Therefore, ETP/DN1 cells are normal residents of bone marrow, while double positive T cells are only found in thymus. T cell transformation of very early populations also agrees with the predominant presence of the T cell leukemic clone in bone marrow [15].
Limitation of lineage choice during development is regulated by a combination of signaling pathways and transcription factors. The main receptor controlling the proB stage is the IL-7R, which is composed of an α chain (IL-7Rα) and the common cytokine receptor G chain (GC) [16,17]. Deletion of IL-7Rα or GC leads to developmental arrest at the early proB stage [18–21]. IL-7 activates three major signaling pathways: 1) JAK–STAT, 2) phosphatidylinositol 3-kinase (PI3K)–Akt and 3) Ras-Raf-Erk [22]. STAT5 (signal transducer and activator of transcription 5) is the predominant STAT protein activated by IL-7 [22,23] and STAT5 loss also arrest B cells at the early proB stage. Once the preBCR is expressed, it can take over many of the functions performed by the IL-7 receptor, since the preBCR also activates the PI3K-Akt and Ras-Raf-Erk pathways [24,25].
Downstream of IL-7 two transcription factors have been documented as the most important for cell entry into the B cell lineage: E2A/TCF3 (immunoglobulin enhancer binding factors E12/E47/transcription factor 3) and EBF1 (Early B cell Factor 1) [26–28]. On the other hand, PAX5 (Paired box 5) is the more important transcription factor for B cell commitment. Loss of E2A and EBF1 blocks entry into the B cell lineage, and loss of PAX5 redirects B cells into other lineages [28–30]. One of the main molecular functions of PAX5 (acting together with E2A, EBF1 and STAT5) is to allow VDJ recombination [31,32]. Ectopic expression of PAX5 and E2A allows VDJ recombination in non-B cells [45, 46]. Also, E2A, PAX5, IKZF1 and RUNX1, among other transcription factors, are responsible for expression of the VDJ recombinase (RAG) [33,34].
The most important cells that give rise to T cells are ELPs and CLPs. Although, both B and T cells are mainly originated from them, an important genetic difference between cells prone to the B lineage is the expression of EBF1 and PAX5, while for T cells is NOTCH1 signaling. NOTCH1 directs progenitors into the thymus and it is the master orchestrator of T cell lineage entry and development [35,36]. NOTCH contains multiple epidermal growth factor (EGF)-like repeats through which it binds its ligands DLL-1, -2, -4 (Delta-like ligand), and Jagged-1 and -2 expressed by bone marrow and thymus stromal cells. Upon ligand binding NOTCH1 initiates a series of proteolytic cleavage events, the first one catalyzed by the ADAM family of metalloproteinases and the second by the γ−secretase complex. This cleavage activates NOTCH1 removing the extracellular portion and translocating to the nucleus its intracellular region (ICN), where it becomes part of a large transcriptional activation complex together with CSL and histone acetylase p300. Also, ICN has a C-terminal PEST domain involved in regulation of NOTCH1 ubiquitylation and proteasome-mediated degradation, therefore controlling protein turnover [35–38].

Figure 2.
NOTCH1 expression is importantly regulated by E2A [39], and is essential for activation of genes necessary for T cell entry and early development. Indeed, NOTCH1 expression is turned off in late stages of T cell development, forced expression of NOTCH1 in multipotent progenitor cells direct them to the T cell lineage and controls the expression of several transcription factors important for T cell early development, e.g.
Some of the transcription factors drivers of T cell ALL are normally expressed in non-malignant thymocytes since they are essential regulators of T-cell ontogeny, while others are not expressed in normal ones, but they are rather ectopically expressed by transformed cells (Figure 2). This is contrary to B cells, in which most of the transcription factors associated with transformation fulfill an important regulatory function (Figure 2). This observation supports different mechanisms for the origin of B and T cell ALL. In agreement,
3. Criteria for ALL risk stratification
The clinical and laboratory criteria supporting risk stratification vary among institutions, with most groups considering as high risk the following characteristics: age ≥ 10 or <1 years at presentation, WBC ≥ 50,000/µl, presence of extramedullary disease, T cell immunophenotype, presence of adverse genetic abnormalities such as t(9;22) (
All the above-mentioned prognostic factors are used to classify patients into two risk groups, high and standard risk. For instance, it is known that increased WBC count confers poor prognosis for B cell ALL patients and in T cell ALL, a leukocyte count greater than 100,000/µl is associated with high risk of relapse in the central nervous system. Also, patients with hyperleukocytosis, greater than 400,000/µl, are at high risk of central nervous system hemorrhage and pulmonary and neurological events due to leukostasis. However, most of these risk criteria are better understood for B cell and they are not as clear for T cell ALL patients [3]. Recently, evaluation of early response to therapy has been demonstrated being an important parameter for treatment efficacy and disease prognosis. Based on the latter criteria, it is possible to identify the group of patients that require augmented therapy to improve their outcome.
3.1. Prognostic significance of treatment response
The frequency of bone marrow or circulating lymphoblasts after one week of chemotherapy is associated with risk for relapse [41] and nowadays, this constitutes one of the most useful prognostic factors in childhood ALL. An efficient early response to treatment is determined by evaluating clearance rates of leukemic cells after the induction phase of treatment [42]. This pharmacological response depends on numerous variables, including drug sensitivity /resistance of the leukemic cells, the dosage and the ability of individual patients to metabolize and eliminate anti-leukemic drugs [43,44].
The Berlin-Frankfurt Munster (BFM) group has traditionally employed the response to prednisone for 7 days and one dose of intrathecal methotrexate to stratify patients. Peripheral blood blast count of 1,000/µl after prednisone treatment is used as a threshold to assign patients into two groups, prednisone good responders (GR) and poor responders (PR). The ALL-BMF Group demonstrated in large series of infant patients treated with effective risk-based ALL therapy that prednisone response is a strong prognostic parameter for outcome; 75% of infants were good responders (GR) and achieved an EFS of 53% at 6 years using conventional therapy, whereas poor responder infants had an EFS of 15% [41]. The Tokyo Cancer Children´s Leukemia Group also showed that B and T cell ALL patients with high blast counts at day 8, had a 4 years EFS of 74%; in contrast, patients without blasts presented an EFS of 89% for B and 95% for T cell ALL [43]. Thus, it is well accepted that early response to prednisone treatment is a strong indicator of EFS [41]. However, this assessment is limited by the low sensitivity (5-10% blasts) of microscopy-based methods of blast quantification [45]. The morphological analysis of blasts by conventional methods easily underestimates the presence and frequency of residual cells. PCR or flow cytometry- based methods for detecting MRD are at least 100 times more sensitive.
The common principle for all MRD assessments is that leukemogenic process results in molecular and cellular changes, which distinguish leukemic cells from their normal counterparts [46]. In patients with ALL, MRD can be monitored by flow cytometry, PCR amplification of gene fusion transcripts, and PCR amplification of the B and T cell antigen receptors (BCR/TCR specific VDJ recombinants). Combining information about cell size, granularity and expression of surface and intracellular molecules, it is possible to identify by flow cytometry a phenotypic signature characteristic of leukemic cells. Flow cytometry-based identification of cell immunophenotypes allows the detection of one leukemic cell among 10,000 normal cells (0.01%) [47,48]; however, these assays require high expertise for quality results, previous knowledge of immunophenotypic profiles of normal and leukemic cells and experience to select the best markers useful for each patient [49]. Other option to distinguish leukemic from normal cells is the PCR screening of gene fusion transcripts, produced by specific chromosomal translocations, among the most common of them are:
MRD studies revealed that many patients who achieve remission by traditional methods harbored residual disease predisposing them to relapse [46,48]. The most immediate application of MRD testing is the identification of patients who are candidates for treatment intensification, since levels of MRD are proportional to the risk of relapse [51]. The most appropriate time for evaluation of MRD vary between different groups, for the ALL-BMF 95 protocol in Austria, MRD quantification by flow cytometry of bone marrow samples must be estimated on days 33 and 78 post-treatment. In the experience of St. Jude Children’s Research Hospital, the presence of 0.01% residual cells on days 19, 46, or subsequent time points during treatment, is strongly associated with a high risk of relapse [54,55]. The Children’s Oncology Group quantifies MRD in bone marrow on day 29 post-treatment, and ≥ 0.01% of MRD is associated with poor outcome [56]. The Dana-Farber Cancer Institute ALL Consortium, considers MRD cut-off values of 0.1% for prediction of 5-year relapse hazard [57]. Recently, the Italian cooperative group AIEOP identified 3 risk groups based on MRD values by flow cytometry of bone marrow samples on day 15 of treatment. Those risk groups are: standard (<0.01% MRD) with a 5-years cumulative incidence of relapse (CIR) of 7.5%, intermediate (0.01% - <10% MRD) with CIR of 17.5%, and high (>10% MRD) with CIR of 47.2% [58]. MRD is also useful as an independent predictor of second relapse in patients with ALL who had a previous relapse and achieved a second remission [59,60]. Notably, the time of first relapse and MRD are the only 2 significant predictors of outcome in a multivariate analysis [60].
3.2. Genetic abnormalities in ALL as prognostic factors
From a genetic point of view, ALL is one of the best characterized malignancies. Numerical and structural chromosomal abnormalities have been described by cytogenetic methods, FISH, PCR, and more recently, by next generation sequencing. Chromosomal abnormalities are clonal markers of the ALL blast, since the cytogenetic and molecular analyses have revealed that approximately 75% of ALL-children present these genetic lesions [7]. To date, more than 200 genes have been found participating, downstream of common ALL translocations. Interestingly, a handful of these genes are affected by more than one translocation, thus supporting specific mechanisms of leukemogenesis [9].
The genetic abnormalities found in ALL are basically of two types: 1) gains or losses of one or several chromosomes (numerical abnormalities) and 2) translocations generating gene fusions that encode proteins with novel functions (chimeric proteins), or that re-locate a gene close to a strong transcriptional promoter causing gene overexpression. These translocations are produced by double-strand breaks (DSB) in different chromosomes or different regions of one chromosome, that are then recombined through non-homologous end-joining mechanisms [9,61]. These events of illegitimate recombination result in juxtaposition of normally separated regions, relocating a gene or producing a chimeric fusion gene [3].
Several studies have demonstrated that the first genetic lesion in childhood ALL often occurs in uterus. Screening of many of the genetic lesions that characterize the ALL blast in blood samples from Guthrie cards supports their prenatal origin. These studies have shown the presence of the same gene fusion in blood samples collected at birth and in the leukemic blasts at diagnosis. Thus, an intrauterine origin of
The known ALL genetic abnormalities have been relevant for the identification of genes involved in cancer and therefore for the insights in the biology of the leukemogenic process. Importantly, these genetic abnormalities are a disease signature that has been an invaluable tool for the precise disease diagnosis, prognosis and stratification into risk groups, guiding patient management and treatment choice [63]. The Third International Workshop on Chromosomes in Leukemia was the first major study demonstrating the independent prognostic significance of cytogenetic findings in ALL, providing data on clinical relevance of chromosomal recurrent aberrations, and elucidating its molecular basis and biologic consequences [64]. Given their importance, it is the main goal of this chapter to describe in detail the most important genetic abnormalities in the stratification of ALL patients, highlighting aspects of their oncogenic mechanisms, incidence and prognosis.
4. Molecular and cytogenetic subgroups in pediatric B cell ALL
As it was previously mentioned, several genetic abnormalities are characteristic of ALL and have been relevant for the identification of genes involved in cancer and therefore have given insights into the biology of the leukemogenic process, plus they have been an invaluable tool for the precise disease diagnosis, prognosis and stratification into risk groups. Several of them will be discussed in the coming sections.
4.1. BCR-ABL1 fusion
The
The Ph chromosome detected in CML varies from the one in ALL, with different
Ph positive childhood ALL is associated with older age at presentation, high leukocyte count, French-American-British (FAB) L2 morphology, and high incidence of central nervous system. Age at ALL presentation influences the prognosis of this genetic rearrangement; patients with ages ranging from one to nine years have a better prognosis than adolescents and young adults [70,74]. Thus, Ph positive is associated with a very high risk and poor prognosis. Although more than 95% of patients achieve an adequate response to induction therapy, these remissions are shallow and short-lived [6]; additionally, these patients frequently present high levels of MRD at the end of the induction therapy [75]. Ph positive ALL incidence varies among different cohorts (Table 1), ranging between 2-3% for Western European countries (Germany, Italy, Austria, Britain, Switzerland) [76–78], 1-4% for American countries (USA and Mexico) [4,79] and 7-15% for Eastern countries (China, Taiwan, Malaysia-Singapore) [63].
Intensive research efforts were done to demonstrate the BCR-ABL1 transforming activity
|
|
|
||||||||||||||||||||
|
|
|
||||||||||||||||||||
|
||||||||||||||||||||||
|
38 | - | 31 | 25 | 41 | 31 | - | 24 | - | Excellent prognosis with anti-metabolite treatment | ||||||||||||
|
- | - | - | 1-2 | <1 | - | - | - | - | Poor prognosis | ||||||||||||
|
||||||||||||||||||||||
|
4 | 2 | - | 5 | 5 |
5 | 7 | 5 | 4 | Improved prognosis with high-dose methotrexate treatment | ||||||||||||
|
2 | 3 | 2 | 8 | 2 | 9 | 0 | 3 | 5 | Poor prognosis | ||||||||||||
|
3 | 2 | 2 | 2 | 1 | 4 | 5 | 17 | 7 | Improved early treatment outcome with imatinib | ||||||||||||
|
25 | 22 | 21 | 25 | 13 | 9 | 7 | 19 | 13 | Excellent prognosis with asparaginase |
Table 1.
-, non described
4.2. E2A-PBX1 fusion
The
According to studies in different populations (Table 1),
Clinical features of pre-B ALL positive for
Another translocation involving the
4.3. MLL translocations
Myeloid/lymphoid or Mixed lineage leukemia gene (
MLL in its mature form consists of two non-covalently associated subunits, an N-terminal 320 kDa fragment (MLLN) and a C-terminal 180 kDa moiety (MLLC), which are both core components of the MLL complex and result from the cleavage of nascent MLL by an aspartic protease named taspase 1. The MLLN fragment is thought to bind DNA regulatory regions of clustered
Most
All
As mentioned before, the frequency of
4.4. ETV6-RUNX1 fusion
Several abnormalities secondary to
Several studies have supported that
The clonal origin of relapse has been investigated comparing CNA profiles from matched
More recently, it has been shown that genes associated with glucocorticoid mediated apoptosis could be deleted in
4.5. Hyperdiploidy
Hyperdiploidy with 51-65 chromosomes is also a frequent abnormality, 25-41% of ALL patients present this numerical aberration [10,83,85] and are generally associated with a favorable outcome (Table 1). This includes age 3-5 years and relative low WBC count at presentation, B cell precursor immunophenotype [127] and a 5-year EFS estimate of 85-95% when patients are treated with anti-metabolite based therapy [4,127]. Leukemic lymphoblasts in this subgroup have a high propensity to undergo apoptosis
High hyperdiploidy can be detected by cytogenetic analysis or flow cytometry. This latter technique measures the DNA content of the leukemic blasts in comparison to the normal cell pool and DNA content of 1.16 is considered as a prognostic indicator of favorable outcome. However, it is recommended to perform additional cytogenetic studies to detect specific chromosome gains, and discard the presence of additional genetic rearrangements, which could also influence disease outcome. About 50% of hyperdiploid cases present additional abnormalities as duplications of 1q or isochromosome 17q, this last abnormality confers adverse prognosis [128]. High hyperdiploidy is often characterized cytogenetically by massive aneuploidy, originating a non-random gain of specific chromosomes, including some or all of +X, +4, +6, +10, +14, +17, +18, and +21; trisomies and tetrasomies of other chromosomes are also present in this group of patients [127].
In spite of the excellent prognosis associated to this genetic subtype, about 25% of the patients develop adverse events, indicating outcome differences and genetic subgroups between high hyperdiploid patients. For this reason, diverse studies have been performed trying to identify prognostic characteristics in these ALL patients. Based on cytogenetic studies and survival analyses, specific trisomies have been found associated to prognosis. Results from univariate analyses informed that gain of individual chromosomes 6, 4, 10 and 18 improves prognosis, in contrast, trisomy 5 confers worse prognosis [129–131]. Currently, the Children's Cancer Group (CCG) and the Pediatric Oncology Group (POG) consider the presence of simultaneous trisomies of chromosomes 4, 10, and 17 as a favorable prognostic factor [132].
Analysis by SNP array of high hyperdiploid patients have been performed and revealed that 80% presented CNAs, which are not detected by traditional cytogenetic methods. An association between duplication of 1q and +5 has often been observed, and also uniparental isodisomies of chromosomes 9 and 11, gains of chromosomes 17q and 21q, deletions and microdeletions of
ALL cases with 47-50 chromosomes have an intermediate prognosis [71], near-triploidy (69 to 81 chromosomes) [134] have a response to therapy similar to that of non-hyperdiploid, and ALL cases with near tetraploidy (82 to 94 chromosomes) have a high frequency of T cell immunophenotype (see T cell ALL section) and frequently harbors a cryptic ETV6-RUNX1 fusion [135]. These tetraploid leukemias, although significantly less common, have a worse prognosis than the ones with 51-65 chromosomes. The genetic reason for this differential prognosis is presently unclear.
4.6. Hypodiploidy
The hypodiploid ALL is defined as leukemic blasts with less that 46 chromosomes and it is present in 6-7% of patients with childhood ALL. Three different subgroups have been defined according to the number of chromosomes, which are also important for disease outcome: near-haploid ALL (less than 30 chromosomes), low hypodiploid ALL (33-39 chromosomes) and high hypodiploid ALL (42-45 chromosomes). Near-haploidy is observed approximately in 0.5% of ALL cases and it is most frequently associated with females, and together with low hypodiploidy is related with the worst prognosis. Also, children with near-haploidy tend to be younger than those with low hypodiploidy [134,136]. Most of the hypodiploid ALL patients belong to the high hypodiploid group.
The pattern of chromosome loss in near-haploidy is not random as there is preferential retention of two copies of chromosomes 6, 8, 10, 14, 18, 21, and the sex chromosomes. In rare cases, an apparent hyperdiploid genome is observed but the number of chromosomes results from doubling haploid or near-haploid chromosome content. In these cases, although there is an increased in the total number of chromosomes, this ALL is still characterized by losses of specific chromosomes. This ALL is frequently wrongly diagnosed without a careful cytogenetic and DNA content analysis [136], and an appropriate diagnosis is important as near-haploidy defines a rare type of ALL associated with short remission duration and poor prognosis. Therefore, a clear diagnosis of the total chromosome number is essential to stratify patients into the appropriate risk group.
5. Molecular and cytogenetic subgroups in pediatric T-cell ALL
T cell ALL is a neoplastic disorder characterized by malignant transformation of early thymocytes [37]. It accounts for approximately 10-15% of pediatric ALL cases [2,37,137–139] and tends to present clinically with high circulating blast cell counts, mediastinal masses, and often central nervous system involvement [37,140]. Therefore, it is a high risk ALL with a relapse rate of about 30% within the first 2 years following diagnosis [15,139]. T cell ALL is caused by genetic alterations leading to a variety of changes that can affect cell cycle control, unlimited self-renewal capacity, impaired differentiation and loss of sensitivity to death signals [37]. As previously described, T cell ALL shares some chromosome rearrangements with B cell ALL; however, about 50% of T-ALL patients have recurrent chromosomal translocations specific of this subtype. The most common chromosome abnormalities include rearrangements affecting the
|
||||
|
|
|
|
|
|
4-10 | Homeodomain transcription factor Spleen development |
Good | [37,38,143] |
|
3 | bHLH transcription factor HSC survival |
Undefined | [15,37] |
|
<1 | Transmembrane receptor T-cell development |
Poor | [35–38] |
|
||||
|
20 | Homeodomain transcription factor Neural development |
Poor | [37,38,137] |
|
17 | bHLH transcription factor | Undefined | [15,37] |
|
>50 | Transmembrane receptor T-cell development |
Poor | [35–38] |
Table 2.
Translocations and mutations in T-ALL
HSC, Hematopoietic Stem Cell
5.1. Impaired differentiation caused by defects in transcription factors expression/function
5.1.1. Deregulation of TLX1 and TLX3 Homeobox genes
Homeobox genes (
The cryptic chromosomal translocation t(5;14)(q35;q32) juxtaposes
5.1.2. Deregulation of TAL1 , a basic Helix-Loop-Helix (bHLH) gene
Two different models have explained the oncogenic potential and transformation mechanism of
It is documented that
A novel subgroup of early T cell precursor leukemia has been reported, characterized by simultaneous expression of T cell/ stem-cell/myeloid markers and very poor prognosis when treated with standard intensive chemotherapy. Interestingly, this subgroup includes a part of those patients with
5.2. Activation of the NOTCH1 signaling pathway
The first alteration described affecting
Glucocorticoids are normally used to treat T cell ALL patients and glucocorticoid resistance have been mapped to NOTCH1 aberrant expression. Recently, a combination therapy with glucocorticoids and GSIs in a mouse model of resistant to treatment T cell ALL show promising results, arguing that NOTCH1 inhibitors in combination with traditional anti-leukemic drugs might improve disease prognosis in patients with NOTCH1 mutations [149].
6. New prognostic markers detected by genomic variation assays and gene expression evaluation in childhood ALL
The previously described genetic abnormalities in ALL influence the aggressive behavior of leukemic cells and the response to treatment in an important manner. Unfortunately, those abnormalities are not 100% predictive of disease outcome. More recently, genome wide analysis has identified genes associated with risk to relapse in patients with primary gene fusions and hyperdiploidy. These studies have also found novel gene abnormalities probably leading to altered signaling pathways and gene expression patterns in the leukemic blast. Nowadays, many novel cryptic translocations, mutations, deletions, and abnormal expression profiles are considered useful outcome markers in children with ALL and several of these more common markers will be further detailed in this section.
6.1. CASP8AP2
The Caspase-8-Associated Protein 2 gene, also known as FLICE associated Huge Protein (
The clinical significance of
Analyses of
Biologic basis of the variation of
The usefulness of
6.2. IKZF1
The
Ikaros plays an essential role in development and differentiation of lymphoid and myeloid lineages. It acts as a tumor suppressor and as a regulator of gene expression through a chromatin remodeling function. In normal cells, long Ik-1 and Ik-2 isoforms are more expressed than the predominantly dominant-negative isoforms, Ik-3, Ik-4, Ik-5 and Ik-6 [162,163]. During alternative splicing Ikaros is susceptible to loss the amino-terminal DNA-binding domain, leading to increased expression of specific isoforms, in particular Ik-6, which is strongly associated with B and T cell ALL [164–166].
On the other hand, SNP array analysis of B cell ALL children has revealed deletions of complete
“Short” and “long” isoforms can be expressed in leukemic cells from both B and T cell ALL patients, however, the frequency and expression levels seem to vary between specific immunophenotype and genetic subgroups [169,170]. For instance, Ph positive B cell ALL patients tend to have higher levels of Ik-6 in contrast to Ik-1 and Ik-2 [170]. Interestingly, one study found that
Regarding prognosis, there is a strong correlation between mutations, deletions in
6.3. JAK2
The
Recently, it has been shown that the mutation R683, within the JAK2 pseudokinase domain, is present in approximately 3-4% of childhood ALL patients [178]. About 10% of high risk B cell ALL patients are R683+, however, the incidence is increased in patients with Down syndrome (18-28%) [179–181]. The incidence of
6.4. CRLF2
The
Approximately 40% of children with B cell ALL have
About 5-7% of Caucasian non-selected B cell ALL patients present
Rearrangements and overexpression of
A strong interaction among
According to these observations, it has been speculated that
7. Conclusions
Progress in risk adapted treatment of childhood ALL can currently cure up to 80% of patients. Prognostic factors including patient and disease characteristics as well as response to treatment, play a key role in stratification. Through exhaustive genetic characterization of ALL, gene fusions, point mutations, deletions and gross losses or gains of genetic material have been associated to prognosis. Recently, gene expression and comparative genomic hybridization microarrays have identified new potential genetic markers for predicting outcome. These markers have been evaluated in order to recognize patients prone to relapse, even when they present low risk characteristics by conventional parameters of risk stratification. Based on those studies, gene signatures, mutations and signaling pathways no previously associated to ALL have been identified. Detected abnormalities are involved in diverse cellular processes, as cell cycle progression, cell death, and regulation of gene expression. These activities directly influence how the leukemic blast responds to treatment, and have an important role in the relapse process. Novel genetic alterations that have been associated with poor outcome in ALL patients are rearrangements/mutations that trigger
Acknowledgements
To CONACyT FONSEC SSA/IMSS/ISSSTE//44402. (PPV) and CONACyT PhD grant 165427 (RJV).
References
- 1.
Pérez-Saldivar ML, Fajardo-Gutiérrez A, Bernáldez-Ríos R, Martínez-Avalos A, Medina-Sanson A, Espinosa-Hernández L, et al. Childhood acute leukemias are frequent in Mexico City: descriptive epidemiology. BMC Cancer. 2011;11:355. - 2.
Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Pei D, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009 Feb;10(2):147–56. - 3.
Pui C-H, Campana D. Childhood Leukemia. In Abeloff’s Clinical Oncology. (ed.) Elsevier Inc: Churchill Livingstone; 2008. p. 2139–69. - 4.
Pui C-H, Carroll WL, Meshinchi S, Arceci RJ. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J. Clin. Oncol. 2011 Feb 10;29(5):551–65. - 5.
Neale GAM, Campana D, Pui C-H. Minimal residual disease detection in acute lymphoblastic leukemia: real improvement with the real-time quantitative PCR method? J. Pediatr. Hematol. Oncol. 2003 Feb;25(2):100–2. - 6.
Bhojwani D, Howard SC, Pui C-H. High-risk childhood acute lymphoblastic leukemia. Clin Lymphoma Myeloma. 2009;9 Suppl 3:S222–230. - 7.
Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development. Nat. Rev. Cancer. 2007 Nov;7(11):823–33. - 8.
Pui CH, Crist WM, Look AT. Biology and clinical significance of cytogenetic abnormalities in childhood acute lymphoblastic leukemia. Blood. 1990 Oct 15;76(8):1449–63. - 9.
Greaves MF, Wiemels J. Origins of chromosome translocations in childhood leukaemia. Nat. Rev. Cancer. 2003 Sep;3(9):639–49. - 10.
Pérez-Vera P, Reyes-León A, Fuentes-Pananá EM. Signaling proteins and transcription factors in normal and malignant early B cell development. Bone Marrow Res. 2011;2011:502751. - 11.
Richie Ehrlich LI, Serwold T, Weissman IL. In vitro assays misrepresent in vivo lineage potentials of murine lymphoid progenitors. Blood. 2011 Mar 3;117(9):2618–24. - 12.
Schlenner SM, Madan V, Busch K, Tietz A, Läufle C, Costa C, et al. Fate mapping reveals separate origins of T cells and myeloid lineages in the thymus. Immunity. 2010 Mar 26;32(3):426–36. - 13.
Fuentes-Pananá EM, Bannish G, Karnell FG, Treml JF, Monroe JG. Analysis of the individual contributions of Igalpha (CD79a)- and Igbeta (CD79b)-mediated tonic signaling for bone marrow B cell development and peripheral B cell maturation. J. Immunol. 2006 Dec 1;177(11):7913–22. - 14.
Fuentes-Pananá EM, Bannish G, Monroe JG. Basal B-cell receptor signaling in B lymphocytes: mechanisms of regulation and role in positive selection, differentiation, and peripheral survival. Immunol. Rev. 2004 Feb;197:26–40. - 15.
van Grotel M, Meijerink JPP, Beverloo HB, Langerak AW, Buys-Gladdines JGCAM, Schneider P, et al. The outcome of molecular-cytogenetic subgroups in pediatric T-cell acute lymphoblastic leukemia: a retrospective study of patients treated according to DCOG or COALL protocols. Haematologica. 2006 Sep;91(9):1212–21. - 16.
Noguchi M, Nakamura Y, Russell SM, Ziegler SF, Tsang M, Cao X, et al. Interleukin-2 receptor gamma chain: a functional component of the interleukin-7 receptor. Science. 1993 Dec 17;262(5141):1877–80. - 17.
Milne CD, Paige CJ. IL-7: a key regulator of B lymphopoiesis. Semin. Immunol. 2006 Feb;18(1):20–30. - 18.
Peschon JJ, Morrissey PJ, Grabstein KH, Ramsdell FJ, Maraskovsky E, Gliniak BC, et al. Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice. J. Exp. Med. 1994 Nov 1;180(5):1955–60. - 19.
von Freeden-Jeffry U, Vieira P, Lucian LA, McNeil T, Burdach SE, Murray R. Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine. J. Exp. Med. 1995 Apr 1;181(4):1519–26. - 20.
Cao X, Shores EW, Hu-Li J, Anver MR, Kelsall BL, Russell SM, et al. Defective lymphoid development in mice lacking expression of the common cytokine receptor gamma chain. Immunity. 1995 Mar;2(3):223–38. - 21.
DiSanto JP, Müller W, Guy-Grand D, Fischer A, Rajewsky K. Lymphoid development in mice with a targeted deletion of the interleukin 2 receptor gamma chain. Proc. Natl. Acad. Sci. U.S.A. 1995 Jan 17;92(2):377–81. - 22.
Kovanen PE, Leonard WJ. Cytokines and immunodeficiency diseases: critical roles of the gamma(c)-dependent cytokines interleukins 2, 4, 7, 9, 15, and 21, and their signaling pathways. Immunol. Rev. 2004 Dec;202:67–83. - 23.
Hennighausen L, Robinson GW. Interpretation of cytokine signaling through the transcription factors STAT5A and STAT5B. Genes Dev. 2008 Mar 15;22(6):711–21. - 24.
Ramadani F, Bolland DJ, Garcon F, Emery JL, Vanhaesebroeck B, Corcoran AE, et al. The PI3K isoforms p110alpha and p110delta are essential for pre-B cell receptor signaling and B cell development. Sci Signal. 2010;3(134):ra60. - 25.
Marshall AJ, Fleming HE, Wu GE, Paige CJ. Modulation of the IL-7 dose-response threshold during pro-B cell differentiation is dependent on pre-B cell receptor expression. J. Immunol. 1998 Dec 1;161(11):6038–45. - 26.
Cobaleda C, Schebesta A, Delogu A, Busslinger M. Pax5: the guardian of B cell identity and function. Nat. Immunol. 2007 May;8(5):463–70. - 27.
Nutt SL, Kee BL. The transcriptional regulation of B cell lineage commitment. Immunity. 2007 Jun;26(6):715–25. - 28.
Nutt SL, Heavey B, Rolink AG, Busslinger M. Commitment to the B-lymphoid lineage depends on the transcription factor Pax5. Nature. 1999 Oct 7;401(6753):556–62. - 29.
Smith E, Sigvardsson M. The roles of transcription factors in B lymphocyte commitment, development, and transformation. J. Leukoc. Biol. 2004 Jun;75(6):973–81. - 30.
O’Riordan M, Grosschedl R. Coordinate regulation of B cell differentiation by the transcription factors EBF and E2A. Immunity. 1999 Jul;11(1):21–31. - 31.
Sato H, Saito-Ohara F, Inazawa J, Kudo A. Pax-5 is essential for kappa sterile transcription during Ig kappa chain gene rearrangement. J. Immunol. 2004 Apr 15;172(8):4858–65. - 32.
Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, Busslinger M. Pax5 induces V-to-DJ rearrangements and locus contraction of the immunoglobulin heavy-chain gene. Genes Dev. 2004 Feb 15;18(4):411–22. - 33.
Kuo TC, Schlissel MS. Mechanisms controlling expression of the RAG locus during lymphocyte development. Curr. Opin. Immunol. 2009 Apr;21(2):173–8. - 34.
Bain G, Maandag EC, Izon DJ, Amsen D, Kruisbeek AM, Weintraub BC, et al. E2A proteins are required for proper B cell development and initiation of immunoglobulin gene rearrangements. Cell. 1994 Dec 2;79(5):885–92. - 35.
Graux C, Cools J, Michaux L, Vandenberghe P, Hagemeijer A. Cytogenetics and molecular genetics of T-cell acute lymphoblastic leukemia: from thymocyte to lymphoblast. Leukemia. 2006 Sep;20(9):1496–510. - 36.
Van Vlierberghe P, Pieters R, Beverloo HB, Meijerink JPP. Molecular-genetic insights in paediatric T-cell acute lymphoblastic leukaemia. Br. J. Haematol. 2008 Oct;143(2):153–68. - 37.
De Keersmaecker K, Marynen P, Cools J. Genetic insights in the pathogenesis of T-cell acute lymphoblastic leukemia. Haematologica. 2005 Aug;90(8):1116–27. - 38.
Koch U, Radtke F. Mechanisms of T cell development and transformation. Annu. Rev. Cell Dev. Biol. 2011 Nov 10;27:539–62. - 39.
Ikawa T, Hirose S, Masuda K, Kakugawa K, Satoh R, Shibano-Satoh A, et al. An essential developmental checkpoint for production of the T cell lineage. Science. 2010 Jul 2;329(5987):93–6. - 40.
Riz I, Hawley TS, Johnston H, Hawley RG. Role of TLX1 in T-cell acute lymphoblastic leukaemia pathogenesis. Br. J. Haematol. 2009 Apr;145(1):140–3. - 41.
Dördelmann M, Reiter A, Borkhardt A, Ludwig WD, Götz N, Viehmann S, et al. Prednisone response is the strongest predictor of treatment outcome in infant acute lymphoblastic leukemia. Blood. 1999 Aug 15;94(4):1209–17. - 42.
Pui C-H. Recent research advances in childhood acute lymphoblastic leukemia. J. Formos. Med. Assoc. 2010 Nov;109(11):777–87. - 43.
Manabe A, Ohara A, Hasegawa D, Koh K, Saito T, Kiyokawa N, et al. Significance of the complete clearance of peripheral blasts after 7 days of prednisolone treatment in children with acute lymphoblastic leukemia: the Tokyo Children’s Cancer Study Group Study L99-15. Haematologica. 2008 Aug;93(8):1155–60. - 44.
Pui CH, Campana D, Evans WE. Childhood acute lymphoblastic leukaemia--current status and future perspectives. Lancet Oncol. 2001 Oct;2(10):597–607. - 45.
Aricò M, Valsecchi MG, Conter V, Rizzari C, Pession A, Messina C, et al. Improved outcome in high-risk childhood acute lymphoblastic leukemia defined by prednisone-poor response treated with double Berlin-Frankfurt-Muenster protocol II. Blood. 2002 Jul 15;100(2):420–6. - 46.
Szczepański T, Orfão A, van der Velden VH, San Miguel JF, van Dongen JJ. Minimal residual disease in leukaemia patients. Lancet Oncol. 2001 Jul;2(7):409–17. - 47.
Campana D. Status of minimal residual disease testing in childhood haematological malignancies. Br. J. Haematol. 2008 Nov;143(4):481–9. - 48.
Campana D. Determination of minimal residual disease in leukaemia patients. Br. J. Haematol. 2003 Jun;121(6):823–38. - 49.
Campana D, Coustan-Smith E. Detection of minimal residual disease in acute leukemia by flow cytometry. Cytometry. 1999 Aug 15;38(4):139–52. - 50.
Gabert J, Beillard E, van der Velden VHJ, Bi W, Grimwade D, Pallisgaard N, et al. Standardization and quality control studies of “real-time” quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia - a Europe Against Cancer program. Leukemia. 2003 Dec;17(12):2318–57. - 51.
Campana D. Minimal residual disease in acute lymphoblastic leukemia. Semin. Hematol. 2009 Jan;46(1):100–6. - 52.
van der Velden VHJ, Hochhaus A, Cazzaniga G, Szczepanski T, Gabert J, van Dongen JJM. Detection of minimal residual disease in hematologic malignancies by real-time quantitative PCR: principles, approaches, and laboratory aspects. Leukemia. 2003 Jun;17(6):1013–34. - 53.
Flohr T, Schrauder A, Cazzaniga G, Panzer-Grümayer R, van der Velden V, Fischer S, et al. Minimal residual disease-directed risk stratification using real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements in the international multicenter trial AIEOP-BFM ALL 2000 for childhood acute lymphoblastic leukemia. Leukemia. 2008 Apr;22(4):771–82. - 54.
Coustan-Smith E, Sancho J, Hancock ML, Boyett JM, Behm FG, Raimondi SC, et al. Clinical importance of minimal residual disease in childhood acute lymphoblastic leukemia. Blood. 2000 Oct 15;96(8):2691–6. - 55.
Coustan-Smith E, Behm FG, Sanchez J, Boyett JM, Hancock ML, Raimondi SC, et al. Immunological detection of minimalresidual disease in children with acute lymphoblastic leukaemia. Lancet. 1998 Feb 21;351(9102):550–4. - 56.
Borowitz MJ, Devidas M, Hunger SP, Bowman WP, Carroll AJ, Carroll WL, et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children’s Oncology Group study. Blood. 2008 Jun 15;111(12):5477–85. - 57.
Zhou J, Goldwasser MA, Li A, Dahlberg SE, Neuberg D, Wang H, et al. Quantitative analysis of minimal residual disease predicts relapse in children with B-lineage acute lymphoblastic leukemia in DFCI ALL Consortium Protocol 95-01. Blood. 2007 Sep 1;110(5):1607–11. - 58.
Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D, et al. Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J. Clin. Oncol. 2009 Nov 1;27(31):5168–74. - 59.
Eckert C, Biondi A, Seeger K, Cazzaniga G, Hartmann R, Beyermann B, et al. Prognostic value of minimal residual disease in relapsed childhood acute lymphoblastic leukaemia. Lancet. 2001 Oct 13;358(9289):1239–41. - 60.
Coustan-Smith E, Gajjar A, Hijiya N, Razzouk BI, Ribeiro RC, Rivera GK, et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia after first relapse. Leukemia. 2004 Mar;18(3):499–504. - 61.
Pfeiffer P, Goedecke W, Obe G. Mechanisms of DNA double-strand break repair and their potential to induce chromosomal aberrations. Mutagenesis. 2000 Jul;15(4):289–302. - 62.
Greaves M. In utero origins of childhood leukaemia. Early Hum. Dev. 2005 Jan;81(1):123–9. - 63.
Chen Z, Sandberg AA. Molecular cytogenetic aspects of hematological malignancies: clinical implications. Am. J. Med. Genet. 2002 Oct 30;115(3):130–41. - 64.
Mrózek K, Harper DP, Aplan PD. Cytogenetics and molecular genetics of acute lymphoblastic leukemia. Hematol. Oncol. Clin. North Am. 2009 Oct;23(5):991–1010. - 65.
Heisterkamp N, Stam K, Groffen J, de Klein A, Grosveld G. Structural organization of the bcr gene and its role in the Ph’ translocation. Nature. 1985 Jul 27;315(6022):758–61. - 66.
Barilá D, Superti-Furga G. An intramolecular SH3-domain interaction regulates c-Abl activity. Nat. Genet. 1998 Mar;18(3):280–2. - 67.
Chissoe SL, Bodenteich A, Wang YF, Wang YP, Burian D, Clifton SW, et al. Sequence and analysis of the human ABL gene, the BCR gene, and regions involved in the Philadelphia chromosomal translocation. Genomics. 1995 May 1;27(1):67–82. - 68.
Fröhling S, Döhner H. Chromosomal abnormalities in cancer. N. Engl. J. Med. 2008 Aug 14;359(7):722–34. - 69.
Chalandon Y, Schwaller J. Targeting mutated protein tyrosine kinases and their signaling pathways in hematologic malignancies. Haematologica. 2005 Jul;90(7):949–68. - 70.
van Dongen JJ, Macintyre EA, Gabert JA, Delabesse E, Rossi V, Saglio G, et al. Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Report of the BIOMED-1 Concerted Action: investigation of minimal residual disease in acute leukemia. Leukemia. 1999 Dec;13(12):1901–28. - 71.
Raimondi SC, Roberson PK, Pui CH, Behm FG, Rivera GK. Hyperdiploid (47-50) acute lymphoblastic leukemia in children. Blood. 1992 Jun 15;79(12):3245–52. - 72.
Nahar R, Ramezani-Rad P, Mossner M, Duy C, Cerchietti L, Geng H, et al. Pre-B cell receptor-mediated activation of BCL6 induces pre-B cell quiescence through transcriptional repression of MYC. Blood. 2011 Oct 13;118(15):4174–8. - 73.
Skorski T. Genomic instability: The cause and effect of BCR/ABL tyrosine kinase. Curr Hematol Malig Rep. 2007 May;2(2):69–74. - 74.
Pui C-H, Evans WE. Treatment of acute lymphoblastic leukemia. N. Engl. J. Med. 2006 Jan 12;354(2):166–78. - 75.
Pieters R, Carroll WL. Biology and treatment of acute lymphoblastic leukemia. Hematol. Oncol. Clin. North Am. 2010 Feb;24(1):1–18. - 76.
Szczepański T, Harrison CJ, van Dongen JJM. Genetic aberrations in paediatric acute leukaemias and implications for management of patients. Lancet Oncol. 2010 Sep;11(9):880–9. - 77.
Moorman AV, Ensor HM, Richards SM, Chilton L, Schwab C, Kinsey SE, et al. Prognostic effect of chromosomal abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia: results from the UK Medical Research Council ALL97/99 randomised trial. Lancet Oncol. 2010 May;11(5):429–38. - 78.
Schrappe M, Reiter A, Ludwig WD, Harbott J, Zimmermann M, Hiddemann W, et al. Improved outcome in childhood acute lymphoblastic leukemia despite reduced use of anthracyclines and cranial radiotherapy: results of trial ALL-BFM 90. German-Austrian-Swiss ALL-BFM Study Group. Blood. 2000 Jun 1;95(11):3310–22. - 79.
Jiménez-Morales S, Miranda-Peralta E, Saldaña-Alvarez Y, Perez-Vera P, Paredes-Aguilera R, Rivera-Luna R, et al. BCR-ABL, ETV6-RUNX1 and E2A-PBX1: prevalence of the most common acute lymphoblastic leukemia fusion genes in Mexican patients. Leuk. Res. 2008 Oct;32(10):1518–22. - 80.
Borkhardt A, Cazzaniga G, Viehmann S, Valsecchi MG, Ludwig WD, Burci L, et al. Incidence and clinical relevance of TEL/AML1 fusion genes in children with acute lymphoblastic leukemia enrolled in the German and Italian multicenter therapy trials. Associazione Italiana Ematologia Oncologia Pediatrica and the Berlin-Frankfurt-Münster Study Group. Blood. 1997 Jul 15;90(2):571–7. - 81.
Harrison CJ, Moorman AV, Barber KE, Broadfield ZJ, Cheung KL, Harris RL, et al. Interphase molecular cytogenetic screening for chromosomal abnormalities of prognostic significance in childhood acute lymphoblastic leukaemia: a UK Cancer Cytogenetics Group Study. Br. J. Haematol. 2005 May;129(4):520–30. - 82.
Pui C-H, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet. 2008 Mar 22;371(9617):1030–43. - 83.
Aldrich MC, Zhang L, Wiemels JL, Ma X, Loh ML, Metayer C, et al. Cytogenetics of Hispanic and White children with acute lymphoblastic leukemia in California. Cancer Epidemiol. Biomarkers Prev. 2006 Mar;15(3):578–81. - 84.
Pérez-Vera P, Salas C, Montero-Ruiz O, Frías S, Dehesa G, Jarquín B, et al. Analysis of gene rearrangements using a fluorescence in situ hybridization method in Mexican patients with acute lymphoblastic leukemia: experience at a single institution. Cancer Genet. Cytogenet. 2008 Jul 15;184(2):94–8. - 85.
Chen B, Wang Y-Y, Shen Y, Zhang W-N, He H-Y, Zhu Y-M, et al. Newly diagnosed acute lymphoblastic leukemia in China (I): abnormal genetic patterns in 1346 childhood and adult cases and their comparison with the reports from Western countries. Leukemia. 2012 Jul;26(7):1608–16. - 86.
Ariffin H, Chen S-P, Kwok CS, Quah T-C, Lin H-P, Yeoh AEJ. Ethnic differences in the frequency of subtypes of childhood acute lymphoblastic leukemia: results of the Malaysia-Singapore Leukemia Study Group. J. Pediatr. Hematol. Oncol. 2007 Jan;29(1):27–31. - 87.
Hunger SP. Tyrosine kinase inhibitor use in pediatric Philadelphia chromosome-positive acute lymphoblastic anemia. Hematology Am Soc Hematol Educ Program. 2011;2011:361–5. - 88.
Nourse J, Mellentin JD, Galili N, Wilkinson J, Stanbridge E, Smith SD, et al. Chromosomal translocation t(1;19) results in synthesis of a homeobox fusion mRNA that codes for a potential chimeric transcription factor. Cell. 1990 Feb 23;60(4):535–45. - 89.
Biondi A, Masera G. Molecular pathogenesis of childhood acute lymphoblastic leukemia. Haematologica. 1998 Jul;83(7):651–9. - 90.
Chan KK-K, Zhang J, Chia N-Y, Chan Y-S, Sim HS, Tan KS, et al. KLF4 and PBX1 directly regulate NANOG expression in human embryonic stem cells. Stem Cells. 2009 Sep;27(9):2114–25. - 91.
LeBrun DP. E2A basic helix-loop-helix transcription factors in human leukemia. Front. Biosci. 2003 May 1;8:s206–222. - 92.
Look AT. Oncogenic transcription factors in the human acute leukemias. Science. 1997 Nov 7;278(5340):1059–64. - 93.
Graham DK, Salzberg DB, Kurtzberg J, Sather S, Matsushima GK, Keating AK, et al. Ectopic expression of the proto-oncogene Mer in pediatric T-cell acute lymphoblastic leukemia. Clin. Cancer Res. 2006 May 1;12(9):2662–9. - 94.
Shiozawa Y, Pedersen EA, Taichman RS. GAS6/Mer axis regulates the homing and survival of the E2A/PBX1-positive B-cell precursor acute lymphoblastic leukemia in the bone marrow niche. Exp. Hematol. 2010 Feb;38(2):132–40. - 95.
Armstrong SA, Look AT. Molecular genetics of acute lymphoblastic leukemia. J. Clin. Oncol. 2005 Sep 10;23(26):6306–15. - 96.
Iacobucci I, Papayannidis C, Lonetti A, Ferrari A, Baccarani M, Martinelli G. Cytogenetic and molecular predictors of outcome in acute lymphocytic leukemia: recent developments. Curr Hematol Malig Rep. 2012 Jun;7(2):133–43. - 97.
Pui C-H, Sandlund JT, Pei D, Campana D, Rivera GK, Ribeiro RC, et al. Improved outcome for children with acute lymphoblastic leukemia: results of Total Therapy Study XIIIB at St Jude Children’s Research Hospital. Blood. 2004 Nov 1;104(9):2690–6. - 98.
Privitera E, Kamps MP, Hayashi Y, Inaba T, Shapiro LH, Raimondi SC, et al. Different molecular consequences of the 1;19 chromosomal translocation in childhood B-cell precursor acute lymphoblastic leukemia. Blood. 1992 Apr 1;79(7):1781–8. - 99.
Crist WM, Carroll AJ, Shuster JJ, Behm FG, Whitehead M, Vietti TJ, et al. Poor prognosis of children with pre-B acute lymphoblastic leukemia is associated with the t(1;19)(q23;p13): a Pediatric Oncology Group study. Blood. 1990 Jul 1;76(1):117–22. - 100.
Inaba T, Inukai T, Yoshihara T, Seyschab H, Ashmun RA, Canman CE, et al. Reversal of apoptosis by the leukaemia-associated E2A-HLF chimaeric transcription factor. Nature. 1996 Aug 8;382(6591):541–4. - 101.
Harper DP, Aplan PD. Chromosomal rearrangements leading to MLL gene fusions: clinical and biological aspects. Cancer Res. 2008 Dec 15;68(24):10024–7. - 102.
Slany RK. The molecular biology of mixed lineage leukemia. Haematologica. 2009 Jul;94(7):984–93. - 103.
Liu H, Takeda S, Kumar R, Westergard TD, Brown EJ, Pandita TK, et al. Phosphorylation of MLL by ATR is required for execution of mammalian S-phase checkpoint. Nature. 2010 Sep 16;467(7313):343–6. - 104.
Sung PA, Libura J, Richardson C. Etoposide and illegitimate DNA double-strand break repair in the generation of MLL translocations: new insights and new questions. DNA Repair (Amst.). 2006 Sep 8;5(9-10):1109–18. - 105.
Chuk MK, McIntyre E, Small D, Brown P. Discordance of MLL-rearranged (MLL-R) infant acute lymphoblastic leukemia in monozygotic twins with spontaneous clearance of preleukemic clone in unaffected twin. Blood. 2009 Jun 25;113(26):6691–4. - 106.
Alexander FE, Patheal SL, Biondi A, Brandalise S, Cabrera ME, Chan LC, et al. Transplacental chemical exposure and risk of infant leukemia with MLL gene fusion. Cancer Res. 2001 Mar 15;61(6):2542–6. - 107.
Bueno C, Catalina P, Melen GJ, Montes R, Sánchez L, Ligero G, et al. Etoposide induces MLL rearrangements and other chromosomal abnormalities in human embryonic stem cells. Carcinogenesis. 2009 Sep;30(9):1628–37. - 108.
Schnyder S, Du NT, Le HB, Singh S, Loredo GA, Vaughan AT. Estrogen treatment induces MLL aberrations in human lymphoblastoid cells. Leuk. Res. 2009 Oct;33(10):1400–4. - 109.
Blanco JG, Edick MJ, Relling MV. Etoposide induces chimeric Mll gene fusions. FASEB J. 2004 Jan;18(1):173–5. - 110.
Barjesteh van Waalwijk van Doorn-Khosrovani S, Janssen J, Maas LM, Godschalk RWL, Nijhuis JG, van Schooten FJ. Dietary flavonoids induce MLL translocations in primary human CD34+ cells. Carcinogenesis. 2007 Aug;28(8):1703–9. - 111.
Rojas E, Mussali P, Tovar E, Valverde M. DNA-AP sites generation by etoposide in whole blood cells. BMC Cancer. 2009;9:398. - 112.
Moneypenny CG, Shao J, Song Y, Gallagher EP. MLL rearrangements are induced by low doses of etoposide in human fetal hematopoietic stem cells. Carcinogenesis. 2006 Apr;27(4):874–81. - 113.
Brassesco MS, Montaldi AP, Gras DE, Camparoto ML, Martinez-Rossi NM, Scrideli CA, et al. Cytogenetic and molecular analysis of MLL rearrangements in acute lymphoblastic leukaemia survivors. Mutagenesis. 2009 Mar;24(2):153–60. - 114.
Montero-Ruíz O, Alcántara-Ortigoza MA, Betancourt M, Juárez-Velázquez R, González-Márquez H, Pérez-Vera P. Expression of RUNX1 isoforms and its target gene BLK in childhood acute lymphoblastic leukemia. Leuk. Res. 2012 Sep;36(9):1105–11. - 115.
Roudaia L, Cheney MD, Manuylova E, Chen W, Morrow M, Park S, et al. CBFbeta is critical for AML1-ETO and TEL-AML1 activity. Blood. 2009 Mar 26;113(13):3070–9. - 116.
De Braekeleer E, Douet-Guilbert N, Morel F, Le Bris M-J, Basinko A, De Braekeleer M. ETV6 fusion genes in hematological malignancies: A review. Leuk. Res. 2012 Aug;36(8):945–61. - 117.
Wiemels JL, Hofmann J, Kang M, Selzer R, Green R, Zhou M, et al. Chromosome 12p deletions in TEL-AML1 childhood acute lymphoblastic leukemia are associated with retrotransposon elements and occur postnatally. Cancer Res. 2008 Dec 1;68(23):9935–44. - 118.
Shurtleff SA, Buijs A, Behm FG, Rubnitz JE, Raimondi SC, Hancock ML, et al. TEL/AML1 fusion resulting from a cryptic t(12;21) is the most common genetic lesion in pediatric ALL and defines a subgroup of patients with an excellent prognosis. Leukemia. 1995 Dec;9(12):1985–9. - 119.
Loh ML, Goldwasser MA, Silverman LB, Poon W-M, Vattikuti S, Cardoso A, et al. Prospective analysis of TEL/AML1-positive patients treated on Dana-Farber Cancer Institute Consortium Protocol 95-01. Blood. 2006 Jun 1;107(11):4508–13. - 120.
Rubnitz JE, Downing JR, Pui CH, Shurtleff SA, Raimondi SC, Evans WE, et al. TEL gene rearrangement in acute lymphoblastic leukemia: a new genetic marker with prognostic significance. J. Clin. Oncol. 1997 Mar;15(3):1150–7. - 121.
Pérez-Vera P, Montero-Ruiz O, Frías S, Ulloa-Avilés V, Cárdenas-Cardós R, Paredes-Aguilera R, et al. Detection of ETV6 and RUNX1 gene rearrangements using fluorescence in situ hybridization in Mexican patients with acute lymphoblastic leukemia: experience at a single institution. Cancer Genet. Cytogenet. 2005 Oct 15;162(2):140–5. - 122.
Siraj AK, Kamat S, Gutiérrez MI, Banavali S, Timpson G, Sazawal S, et al. Frequencies of the major subgroups of precursor B-cell acute lymphoblastic leukemia in Indian children differ from the West. Leukemia. 2003 Jun;17(6):1192–3. - 123.
Liang D-C, Shih L-Y, Yang C-P, Hung I-J, Liu H-C, Jaing T-H, et al. Frequencies of ETV6-RUNX1 fusion and hyperdiploidy in pediatric acute lymphoblastic leukemia are lower in far east than west. Pediatr Blood Cancer. 2010 Sep;55(3):430–3. - 124.
Seeger K, Adams HP, Buchwald D, Beyermann B, Kremens B, Niemeyer C, et al. TEL-AML1 fusion transcript in relapsed childhood acute lymphoblastic leukemia. The Berlin-Frankfurt-Münster Study Group. Blood. 1998 Mar 1;91(5):1716–22. - 125.
van Delft FW, Horsley S, Colman S, Anderson K, Bateman C, Kempski H, et al. Clonal origins of relapse in ETV6-RUNX1 acute lymphoblastic leukemia. Blood. 2011 Jun 9;117(23):6247–54. - 126.
Kuster L, Grausenburger R, Fuka G, Kaindl U, Krapf G, Inthal A, et al. ETV6/RUNX1-positive relapses evolve from an ancestral clone and frequently acquire deletions of genes implicated in glucocorticoid signaling. Blood. 2011 Mar 3;117(9):2658–67. - 127.
Paulsson K, Forestier E, Lilljebjörn H, Heldrup J, Behrendtz M, Young BD, et al. Genetic landscape of high hyperdiploid childhood acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. U.S.A. 2010 Dec 14;107(50):21719–24. - 128.
Pui CH, Raimondi SC, Williams DL. Isochromosome 17q in childhood acute lymphoblastic leukemia: an adverse cytogenetic feature in association with hyperdiploidy? Leukemia. 1988 Apr;2(4):222–5. - 129.
Harris MB, Shuster JJ, Carroll A, Look AT, Borowitz MJ, Crist WM, et al. Trisomy of leukemic cell chromosomes 4 and 10 identifies children with B-progenitor cell acute lymphoblastic leukemia with a very low risk of treatment failure: a Pediatric Oncology Group study. Blood. 1992 Jun 15;79(12):3316–24. - 130.
Heerema NA, Sather HN, Sensel MG, Zhang T, Hutchinson RJ, Nachman JB, et al. Prognostic impact of trisomies of chromosomes 10, 17, and 5 among children with acute lymphoblastic leukemia and high hyperdiploidy (> 50 chromosomes). J. Clin. Oncol. 2000 May;18(9):1876–87. - 131.
Moorman AV, Richards SM, Martineau M, Cheung KL, Robinson HM, Jalali GR, et al. Outcome heterogeneity in childhood high-hyperdiploid acute lymphoblastic leukemia. Blood. 2003 Oct 15;102(8):2756–62. - 132.
Sutcliffe MJ, Shuster JJ, Sather HN, Camitta BM, Pullen J, Schultz KR, et al. High concordance from independent studies by the Children’s Cancer Group (CCG) and Pediatric Oncology Group (POG) associating favorable prognosis with combined trisomies 4, 10, and 17 in children with NCI Standard-Risk B-precursor Acute Lymphoblastic Leukemia: a Children’s Oncology Group (COG) initiative. Leukemia. 2005 May;19(5):734–40. - 133.
Treviño LR, Yang W, French D, Hunger SP, Carroll WL, Devidas M, et al. Germline genomic variants associated with childhood acute lymphoblastic leukemia. Nat. Genet. 2009 Sep;41(9):1001–5. - 134.
Moorman AV, Harrison CJ, Buck GAN, Richards SM, Secker-Walker LM, Martineau M, et al. Karyotype is an independent prognostic factor in adult acute lymphoblastic leukemia (ALL): analysis of cytogenetic data from patients treated on the Medical Research Council (MRC) UKALLXII/Eastern Cooperative Oncology Group (ECOG) 2993 trial. Blood. 2007 Apr 15;109(8):3189–97. - 135.
Raimondi SC, Zhou Y, Shurtleff SA, Rubnitz JE, Pui C-H, Behm FG. Near-triploidy and near-tetraploidy in childhood acute lymphoblastic leukemia: association with B-lineage blast cells carrying the ETV6-RUNX1 fusion, T-lineage immunophenotype, and favorable outcome. Cancer Genet. Cytogenet. 2006 Aug;169(1):50–7. - 136.
Raimondi SC, Zhou Y, Mathew S, Shurtleff SA, Sandlund JT, Rivera GK, et al. Reassessment of the prognostic significance of hypodiploidy in pediatric patients with acute lymphoblastic leukemia. Cancer. 2003 Dec 15;98(12):2715–22. - 137.
Cavé H, Suciu S, Preudhomme C, Poppe B, Robert A, Uyttebroeck A, et al. Clinical significance of HOX11L2 expression linked to t(5;14)(q35;q32), of HOX11 expression, and of SIL-TAL fusion in childhood T-cell malignancies: results of EORTC studies 58881 and 58951. Blood. 2004 Jan 15;103(2):442–50. - 138.
Zhu Y-M, Zhao W-L, Fu J-F, Shi J-Y, Pan Q, Hu J, et al. NOTCH1 mutations in T-cell acute lymphoblastic leukemia: prognostic significance and implication in multifactorial leukemogenesis. Clin. Cancer Res. 2006 May 15;12(10):3043–9. - 139.
van Grotel M, Meijerink JPP, van Wering ER, Langerak AW, Beverloo HB, Buijs-Gladdines JGCAM, et al. Prognostic significance of molecular-cytogenetic abnormalities in pediatric T-ALL is not explained by immunophenotypic differences. Leukemia. 2008 Jan;22(1):124–31. - 140.
Ferrando AA, Neuberg DS, Staunton J, Loh ML, Huard C, Raimondi SC, et al. Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer Cell. 2002 Feb;1(1):75–87. - 141.
Ballerini P, Landman-Parker J, Cayuela JM, Asnafi V, Labopin M, Gandemer V, et al. Impact of genotype on survival of children with T-cell acute lymphoblastic leukemia treated according to the French protocol FRALLE-93: the effect of TLX3/HOX11L2 gene expression on outcome. Haematologica. 2008 Nov;93(11):1658–65. - 142.
Aifantis I, Raetz E, Buonamici S. Molecular pathogenesis of T-cell leukaemia and lymphoma. Nat. Rev. Immunol. 2008 May;8(5):380–90. - 143.
Bergeron J, Clappier E, Radford I, Buzyn A, Millien C, Soler G, et al. Prognostic and oncogenic relevance of TLX1/HOX11 expression level in T-ALLs. Blood. 2007 Oct 1;110(7):2324–30. - 144.
Asnafi V, Beldjord K, Libura M, Villarese P, Millien C, Ballerini P, et al. Age-related phenotypic and oncogenic differences in T-cell acute lymphoblastic leukemias may reflect thymic atrophy. Blood. 2004 Dec 15;104(13):4173–80. - 145.
145. De Keersmaecker K, Real PJ, Gatta GD, Palomero T, Sulis ML, Tosello V, et al. The TLX1 oncogene drives aneuploidy in T cell transformation. Nat. Med. 2010 Nov;16(11):1321–7. - 146.
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 Jun;5(6):587–96. - 147.
Mansur MB, Hassan R, Barbosa TC, Splendore A, Jotta PY, Yunes JA, et al. Impact of complex NOTCH1 mutations on survival in paediatric T-cell leukaemia. BMC Cancer. 2012;12:9. - 148.
Mansur MB, Emerenciano M, Brewer L, Sant’Ana M, Mendonça N, Thuler LCS, et al. SIL-TAL1 fusion gene negative impact in T-cell acute lymphoblastic leukemia outcome. Leuk. Lymphoma. 2009 Aug;50(8):1318–25. - 149.
Real PJ, Tosello V, Palomero T, Castillo M, Hernando E, de Stanchina E, et al. Gamma-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia. Nat. Med. 2009 Jan;15(1):50–8. - 150.
Kittler R, Putz G, Pelletier L, Poser I, Heninger A-K, Drechsel D, et al. An endoribonuclease-prepared siRNA screen in human cells identifies genes essential for cell division. Nature. 2004 Dec 23;432(7020):1036–40. - 151.
Choi YH, Kim KB, Kim HH, Hong GS, Kwon YK, Chung CW, et al. FLASH coordinates NF-kappa B activity via TRAF2. J. Biol. Chem. 2001 Jul 6;276(27):25073–7. - 152.
Jun J-I, Chung C-W, Lee H-J, Pyo J-O, Lee KN, Kim N-S, et al. Role of FLASH in caspase-8-mediated activation of NF-kappaB: dominant-negative function of FLASH mutant in NF-kappaB signaling pathway. Oncogene. 2005 Jan 20;24(4):688–96. - 153.
Alm-Kristiansen AH, Saether T, Matre V, Gilfillan S, Dahle O, Gabrielsen OS. FLASH acts as a co-activator of the transcription factor c-Myb and localizes to active RNA polymerase II foci. Oncogene. 2008 Aug 7;27(34):4644–56. - 154.
Alm-Kristiansen AH, Lorenzo PI, Molværsmyr A-K, Matre V, Ledsaak M, Sæther T, et al. PIAS1 interacts with FLASH and enhances its co-activation of c-Myb. Mol. Cancer. 2011;10:21. - 155.
Barcaroli D, Dinsdale D, Neale MH, Bongiorno-Borbone L, Ranalli M, Munarriz E, et al. FLASH is an essential component of Cajal bodies. Proc. Natl. Acad. Sci. U.S.A. 2006 Oct 3;103(40):14802–7. - 156.
Barcaroli D, Bongiorno-Borbone L, Terrinoni A, Hofmann TG, Rossi M, Knight RA, et al. FLASH is required for histone transcription and S-phase progression. Proc. Natl. Acad. Sci. U.S.A. 2006 Oct 3;103(40):14808–12. - 157.
Yang X, Xu B, Sabath I, Kunduru L, Burch BD, Marzluff WF, et al. FLASH is required for the endonucleolytic cleavage of histone pre-mRNAs but is dispensable for the 5’ exonucleolytic degradation of the downstream cleavage product. Mol. Cell. Biol. 2011 Apr;31(7):1492–502. - 158.
Flotho C, Coustan-Smith E, Pei D, Iwamoto S, Song G, Cheng C, et al. Genes contributing to minimal residual disease in childhood acute lymphoblastic leukemia: prognostic significance of CASP8AP2. Blood. 2006 Aug 1;108(3):1050–7. - 159.
Flotho C, Coustan-Smith E, Pei D, Cheng C, Song G, Pui C-H, et al. A set of genes that regulate cell proliferation predicts treatment outcome in childhood acute lymphoblastic leukemia. Blood. 2007 Aug 15;110(4):1271–7. - 160.
Jiao Y, Cui L, Gao C, Li W, Zhao X, Liu S, et al. CASP8AP2 is a promising prognostic indicator in pediatric acute lymphoblastic leukemia. Leuk. Res. 2012 Jan;36(1):67–71. - 161.
Remke M, Pfister S, Kox C, Toedt G, Becker N, Benner A, et al. High-resolution genomic profiling of childhood T-ALL reveals frequent copy-number alterations affecting the TGF-beta and PI3K-AKT pathways and deletions at 6q15-16.1 as a genomic marker for unfavorable early treatment response. Blood. 2009 Jul 30;114(5):1053–62. - 162.
Molnár A, Georgopoulos K. The Ikaros gene encodes a family of functionally diverse zinc finger DNA-binding proteins. Mol. Cell. Biol. 1994 Dec;14(12):8292–303. - 163.
Sun L, Liu A, Georgopoulos K. Zinc finger-mediated protein interactions modulate Ikaros activity, a molecular control of lymphocyte development. EMBO J. 1996 Oct 1;15(19):5358–69. - 164.
Sun L, Goodman PA, Wood CM, Crotty ML, Sensel M, Sather H, et al. Expression of aberrantly spliced oncogenic ikaros isoforms in childhood acute lymphoblastic leukemia. J. Clin. Oncol. 1999 Dec;17(12):3753–66. - 165.
Sun L, Heerema N, Crotty L, Wu X, Navara C, Vassilev A, et al. Expression of dominant-negative and mutant isoforms of the antileukemic transcription factor Ikaros in infant acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. U.S.A. 1999 Jan 19;96(2):680–5. - 166.
Sun L, Crotty ML, Sensel M, Sather H, Navara C, Nachman J, et al. Expression of dominant-negative Ikaros isoforms in T-cell acute lymphoblastic leukemia. Clin. Cancer Res. 1999 Aug;5(8):2112–20. - 167.
Mullighan CG, Su X, Zhang J, Radtke I, Phillips LAA, Miller CB, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N. Engl. J. Med. 2009 Jan 29;360(5):470–80. - 168.
Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008 May 1;453(7191):110–4. - 169.
Olivero S, Maroc C, Beillard E, Gabert J, Nietfeld W, Chabannon C, et al. Detection of different Ikaros isoforms in human leukaemias using real-time quantitative polymerase chain reaction. Br. J. Haematol. 2000 Sep;110(4):826–30. - 170.
Meleshko AN, Movchan LV, Belevtsev MV, Savitskaja TV. Relative expression of different Ikaros isoforms in childhood acute leukemia. Blood Cells Mol. Dis. 2008 Dec;41(3):278–83. - 171.
Ruiz A, Jiang J, Kempski H, Brady HJM. Overexpression of the Ikaros 6 isoform is restricted to t(4;11) acute lymphoblastic leukaemia in children and infants and has a role in B-cell survival. Br. J. Haematol. 2004 Apr;125(1):31–7. - 172.
Yang JJ, Bhojwani D, Yang W, Cai X, Stocco G, Crews K, et al. Genome-wide copy number profiling reveals molecular evolution from diagnosis to relapse in childhood acute lymphoblastic leukemia. Blood. 2008 Nov 15;112(10):4178–83. - 173.
Kuiper RP, Waanders E, van der Velden VHJ, van Reijmersdal SV, Venkatachalam R, Scheijen B, et al. IKZF1 deletions predict relapse in uniformly treated pediatric precursor B-ALL. Leukemia. 2010 Jul;24(7):1258–64. - 174.
Waanders E, van der Velden VHJ, van der Schoot CE, van Leeuwen FN, van Reijmersdal SV, de Haas V, et al. Integrated use of minimal residual disease classification and IKZF1 alteration status accurately predicts 79% of relapses in pediatric acute lymphoblastic leukemia. Leukemia. 2011 Feb;25(2):254–8. - 175.
Den Boer ML, van Slegtenhorst M, De Menezes RX, Cheok MH, Buijs-Gladdines JGCAM, Peters STCJM, et al. A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study. Lancet Oncol. 2009 Feb;10(2):125–34. - 176.
Pellegrini S, Dusanter-Fourt I. The structure, regulation and function of the Janus kinases (JAKs) and the signal transducers and activators of transcription (STATs). Eur. J. Biochem. 1997 Sep 15;248(3):615–33. - 177.
Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J. Cell. Sci. 2004 Mar 15;117(Pt 8):1281–3. - 178.
Mullighan CG, Collins-Underwood JR, Phillips LAA, Loudin MG, Liu W, Zhang J, et al. Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia. Nat. Genet. 2009 Nov;41(11):1243–6. - 179.
Bercovich D, Ganmore I, Scott LM, Wainreb G, Birger Y, Elimelech A, et al. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down’s syndrome. Lancet. 2008 Oct 25;372(9648):1484–92. - 180.
Gaikwad A, Rye CL, Devidas M, Heerema NA, Carroll AJ, Izraeli S, et al. Prevalence and clinical correlates of JAK2 mutations in Down syndrome acute lymphoblastic leukaemia. Br. J. Haematol. 2009 Mar;144(6):930–2. - 181.
Kearney L, Gonzalez De Castro D, Yeung J, Procter J, Horsley SW, Eguchi-Ishimae M, et al. Specific JAK2 mutation (JAK2R683) and multiple gene deletions in Down syndrome acute lymphoblastic leukemia. Blood. 2009 Jan 15;113(3):646–8. - 182.
Mullighan CG, Zhang J, Harvey RC, Collins-Underwood JR, Schulman BA, Phillips LA, et al. JAK mutations in high-risk childhood acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. U.S.A. 2009 Jun 9;106(23):9414–8. - 183.
Russell LJ, Capasso M, Vater I, Akasaka T, Bernard OA, Calasanz MJ, et al. Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia. Blood. 2009 Sep 24;114(13):2688–98. - 184.
Ziegler SF, Liu Y-J. Thymic stromal lymphopoietin in normal and pathogenic T cell development and function. Nat. Immunol. 2006 Jul;7(7):709–14. - 185.
Akasaka T, Balasas T, Russell LJ, Sugimoto K, Majid A, Walewska R, et al. Five members of the CEBP transcription factor family are targeted by recurrent IGH translocations in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Blood. 2007 Apr 15;109(8):3451–61. - 186.
Hertzberg L, Vendramini E, Ganmore I, Cazzaniga G, Schmitz M, Chalker J, et al. Down syndrome acute lymphoblastic leukemia, a highly heterogeneous disease in which aberrant expression of CRLF2 is associated with mutated JAK2: a report from the International BFM Study Group. Blood. 2010 Feb 4;115(5):1006–17. - 187.
Cario G, Zimmermann M, Romey R, Gesk S, Vater I, Harbott J, et al. Presence of the P2RY8-CRLF2 rearrangement is associated with a poor prognosis in non-high-risk precursor B-cell acute lymphoblastic leukemia in children treated according to the ALL-BFM 2000 protocol. Blood. 2010 Jul 1;115(26):5393–7. - 188.
Harvey RC, Mullighan CG, Chen I-M, Wharton W, Mikhail FM, Carroll AJ, et al. Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood. 2010 Jul 1;115(26):5312–21. - 189.
Ensor HM, Schwab C, Russell LJ, Richards SM, Morrison H, Masic D, et al. Demographic, clinical, and outcome features of children with acute lymphoblastic leukemia and CRLF2 deregulation: results from the MRC ALL97 clinical trial. Blood. 2011 Feb 17;117(7):2129–36. - 190.
Palmi C, Vendramini E, Silvestri D, Longinotti G, Frison D, Cario G, et al. Poor prognosis for P2RY8-CRLF2 fusion but not for CRLF2 over-expression in children with intermediate risk B-cell precursor acute lymphoblastic leukemia. Leukemia: official journal of the Leukemia Society of America, Leukemia Research Fund, U.K [Internet]. 2012 Apr 9 [cited 2012 Jul 30]; Available from: http://www.ncbi.nlm.nih.gov/pubmed/22484421 - 191.
Yoda A, Yoda Y, Chiaretti S, Bar-Natan M, Mani K, Rodig SJ, et al. Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. U.S.A. 2010 Jan 5;107(1):252–7. - 192.
Buitenkamp TD, Pieters R, Gallimore NE, van der Veer A, Meijerink JPP, Beverloo HB, et al. Outcome in children with Down’s syndrome and acute lymphoblastic leukemia: role of IKZF1 deletions and CRLF2 aberrations. Leukemia. 2012. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22441210 ( accessed 22 Mar 2012) - 193.
Carpino N, Thierfelder WE, Chang M, Saris C, Turner SJ, Ziegler SF, et al. Absence of an essential role for thymic stromal lymphopoietin receptor in murine B-cell development. Mol. Cell. Biol. 2004 Mar;24(6):2584–92.