Skip to main content
Indian Journal of Hematology & Blood Transfusion logoLink to Indian Journal of Hematology & Blood Transfusion
. 2015 Oct 20;32(2):141–153. doi: 10.1007/s12288-015-0609-z

Intracellular Signaling Pathways Involved in Childhood Acute Lymphoblastic Leukemia; Molecular Targets

Cristian Fabián Layton Tovar 1,, Hugo Mendieta Zerón 1,2
PMCID: PMC4788999  PMID: 27065575

Abstract

Acute lymphoblastic leukemia (ALL) is a malignant disease characterized by an uncontrolled proliferation of immature lymphoid cells. ALL is the most common hematologic malignancy in early childhood, and it reaches peak incidence between the ages of 2 and 3 years. The prognosis of ALL is associated with aberrant gene expression, in addition to the presence of numerical or structural chromosomal alterations, age, race, and immunophenotype. The Relapse rate with regard to pharmacological treatment rises in childhood; thus, the expression of biomarkers associated with the activation of cell signaling pathways is crucial to establish the disease prognosis. Intracellular pathways involved in ALL are diverse, including Janus kinase/Signal transducers and transcription activators (JAK-STAT), Phosphoinositide-3-kinase–protein kinase B (PI3K-AKT), Ras mitogen-activated protein kinase (Ras-MAPK), Glycogen synthase kinase-3β (GSK-3β), Nuclear factor-kappa beta (NF-κB), and Hypoxia-inducible transcription factor 1α (HIF-1α), among others. In this review, we present several therapeutic targets, intracellular pathways, and molecular markers that are being studied extensively at present.

Keywords: Acute lymphoblastic leukemia, Biological markers, Signaling pathways, Targets, Therapeutic

Introduction

The acute leukemia is the most common cancer in children, adolescents, and young adults. These diseases are characterized by great clinical variability, prompting an on-going search for accurate outcome predictors. Using algorithms based on clinical manifestations at presentation, response to therapy, and several molecular analyses, some patients are diagnosed with the features of high-risk disease and are at comparatively greater risk for relapse [1].

Molecular analyses of patients with high-risk acute leukemia have resulted in an improved understanding of how deregulated cellular signaling can affect resistance to conventional therapy. In this regard, molecular therapies that target genetic abnormalities in leukemic cells and their affected signaling pathways have been emerging in pediatric Acute lymphoblastic leukemia (ALL). Expression profiling, whole-genome sequencing, and other molecular analyses have provided insight into the signaling pathways that are mechanistically related to chemotherapy resistance. For pediatric acute leukemia, the molecular determinants of risk have yet to be fully defined, and target therapies remain in the earliest stages of discovery [1]. Whereas exciting discoveries continue to be made in the identification of relevant molecular biomarkers and targeted therapies, the challenges and opportunities associated with these findings continue to await being clearly defined in future clinical trials [1].

Acute Lymphoblastic Leukemia

The clinical presentation of ALL is variable with an insidious initial manifestation of about 4 weeks. This type of leukemia is more common in early childhood, and reaches its highest incidence between the ages of 2 and 3 years (>80 per million per year, pmpy), with rates decreasing to 20 pmpy in children between 8 and 10 years. In many cases, some leukemic cells have begun to accumulate in different organs such as the liver, lymph nodes, spleen and central nervous system (CNS) when the diagnosis is confirmed [1]. The complete blood count generally reflect marrow failure conditioned by the invasion of leukemic cells such as anemia, thrombocytopenia and neutropenia, therefore, in the study of acute leukemia, morphology and cytochemical stains are essential in the initial characterization of the disease [1, 2].

The genetic basis of this disease has been revolutionized with the study of long cohorts [2]. The identification of new ALL subtypes, structural and numerical genetic alterations, transcriptomic profile, key genes in outcome and resistance to the treatment are critical keys to redefine the ALL classification in the shortcoming time [2, 3].

Classification and Staging

French- American -British (FAB) Classification

Cell morphology criteria classifies ALL as L1, L2 and L3 based on the characteristics of the leukemic cell. Approximately 70–85 % of all enrolled pediatric patients belong to the ALL L1 group. While the morphological distinction between L1 and L2 variety losses prognostic value, the L3 morphology has been associated with the mature B- range [2].

Risk Groups in Childhood ALL

Children with ALL are classified into four risk groups according to the National Cancer Institute (NCI): very high, high, standard and lower risk [4].Participants of a recent workshop sponsored by the NCI defined standard-risk ALL as B-precursor cases with age between one and 10 years and an initial leukocyte count of <50 × 109/L. All other patients are considered high-risk [4].

The type of leukemia determines the initial response to treatment; therefore, the predictive factors have a very important role in childhood acute lymphoblastic leukemia (chALL) (Table 2) [3, 5].

Table 2.

chALL characteristics of risk

Characteristics of risk disease Precursor B-Cell T-linage ALL
Age at presentation <1 year, >10 years >10 years
Initial WBC count >50,000
Extramedullary disease CNS/Testes CNS/Testes
Phenotypic subset Early T-precursor and mixed phenotype acute leukemia. (ETP/MPAL)
Common karyotype abnormalities t(9;22), (q34;q22), rearranged MLL, hypodiploidy (<0.84) Complex anomalies
Common gene mutations FLT3/ITD, BCR/ABL, CRLF2, JAK1, JAK2, IKZF ABD, ME2KC, IL-7R
Expression profiling signatures revealing target-able pathways. Overexpression of PI3K-AKT, RAS-MAPK, BCR-ABL-like Dysregulation TGFβ1, G0/G1 cell, cycle arrest
Clinic presentation Frequent fever
Hepatosplenomegaly and lymphadenopathy as an expression of extramedullary disease
Petechiae
Purpura
Bone pain
Blasts morphology Small, large core, scant cytoplasm homogeneous chromatin
Prognosis Standard risk: 85 %, High risk: 75 %, Infants <50 %
Immunophenotype CD10, CD19, CD22, TdT CD2, CD3, CD5, CD7, TdT

CNS central nervous system

Leukemic Cells Immunophenotype

The immunophenotype is a highly significant prognostics feature. With this diagnostic method, the target is to identify the affected cell line, whether B or T. Children with acute pre-B cell leukemia or early pre-B cell leukemia early respond better than those with T-cell leukemia and mature B-cell leukemia [610].

The prognostic significance of cell surface markers in ALL is an integral and important part of disease diagnosis, classification, and prognosis. These cells have been classified by mature, differentiation, and activation markers, according to the expression of these surface markers: for example, CD45 is a blastic cell marker, CD34, CD117, HLA-DR, and TdT are mature cell markers, CD10, CD19, CD20, CD22, and CD79A are B-lymphoid lineage markers, and CD1a, CD2, CD3, CD5, and CD7T- are lymphoid lineage markers.

The previously mentioned cell surface markers have been correlated with the prognosis in ALL; accordingly, early T-precursor ALL and T-ALL demonstrate poorer prognosis than B-cell lineage leukemia [5, 11, 12]. Similarly, the presence of CD20 has been associated with a high WBC count and a lower platelet count [12].

Cytogenetic Abnormalities

The cytogenetic abnormalities include an important number of structural and numerical alterations present in chALL. According with the World Health Organization (WHO) the classification of acute leukemia includes two lineages; B lymphoblastic leukemia and T lymphoblastic leukemia (Table 1) [13].

Table 1.

WHO classification of acute leukemia

B lymphoblastic leukemia/lymphoma
B lymphoblastic leukemia/lymphoma, NOS
B lymphoblastic leukemia/lymphoma with recurrent genetic abnormalities
B lymphoblastic leukemia/lymphoma with t(9;22)(q34;q11.2);BCR-ABL 1
B lymphoblastic leukemia/lymphoma with t(12;21)(p13;q22) TEL-AML1 (ETV6-RUNX1)
B lymphoblastic leukemia/lymphoma with hyperdiploidy
B lymphoblastic leukemia/lymphoma with hypodiploidy
B lymphoblastic leukemia/lymphoma with t(5;14)(q31;q32) IL3-IGH
B lymphoblastic leukemia/lymphoma with t(1;19)(q23;p13.3);TCF3-PBX1
T lymphoblastic leukemia/lymphoma

Leukemic cells are usually associated with an increased number of chromosomes (hyperdiploid), the most common of this phenomenon affects the 4, 10, 17, and 18 chromosomes; moreover, leukemic cells can also be associated with a decreased number of chromosomes related with a poor prognosis. In fact, a translocation between chromosomes 12 and 21 t(12;21) is more likely to be cured [13]. On the contrary, children with a translocation between chromosomes 9 and 22, or between 1 and 19, have a lower cure rate. Children with a translocation affecting chromosomes 4 and 11 or all (q23) translocations also have a lower cure rate [5, 14, 15]. Chromosomal translocations that possess prognostic significance may be detected in a substantial number of cases of pediatric ALL, and some of these rearrangements are described later.

Cryptic translocation t(12;21) favors the fusion of TEL (ETV6) gene on chromosome 12 to the AML1(CBFA2) gene on chromosome 21, resulting in the TEL-AML1 fusion that can be detected in 20–25 % of cases of B precursor ALL, giving a favorable clinical outcome. Patients with TEL-AML1 fusion, have a favorable clinical outcome, although there is controversy concerning whether the final cure rate is actually higher than that of other patients with B-precursor ALL, or whether the final cure rate is similar, but time-at-relapse occurs significantly later in patients with TEL-AML1 fusion compared with other patients with B-precursor ALL.

Linka et al. [16] recently identified, directly and indirectly, regulated target genes with an inducible design TEL-AML1 system to explain the mechanism associated with cellular proliferation and transport, cellular migration, and stress responses in chALL. Therefore, the Philadelphia chromosome (Ph chromosome) implicates the t(9;22) translocation that is present in approximately 4 % of pediatric ALL which confers a poor prognosis, especially when associated with either a high WBC count or a slow, early response to initial therapy. The Ph chromosome is more common among older patients with B-precursor ALL and high WBC count [13].

Another translocation involved in chALL is the t(11;19) that occurs in about 1 % of cases B or T precursors. The t(11;19) have a worse prognosis but the result appears relatively favorable for children with T-cell ALL and the t(11;19) [14, 15].

Finally, t(1;19) presents in 5–6 % of cases of chALL, and consists of E2A gene to the PBX1 gene fusion located on chromosome 19 and 1 respectively. t(1;19) could present either as an unbalanced translocation or as primarily associated with pre-B ALL (cytoplasmic immunoglobulin-positive). E2A-PBX1 fusion was initially associated with inferior outcome in the context of antimetabolite-based therapy [5]. It has been demonstrated that the poor prognosis associated with t(1;19) can be largely overcome by more intensive therapy. However, improvement in the results appears to be primarily for patients with t(1;19) who, whether unbalanced and balanced; remain at higher risk of treatment non-response [14].

Intracellular Signaling Pathways and Outcome

The outcome for children with ALL is determined by a variety of clinical and laboratory variables. Risk stratification provides a tool to adjust the treatment, avoiding intensive therapies or unnecessary dose escalation for patients with a favorable prognosis. However, there is an important percentage of patients who have been classified with low risk, but they present relapse; currently, despite the use of multiple, independent prognostic features to assess the chance for relapse, approximately one half of patients who relapse are found to have favorable clinical features and an excellent response to induction therapy. Hence, the search for additional variables is necessary to redefine risk stratification [17]. The emergence of resistant clones poses a difficult challenge regardless of the previous treatment intensity [14]. It is possible that the size of a resistant subclone will determine whether a patient fails induction or has an early relapse, and also may govern ease-of-identification of an underlying high-risk molecular feature [18]. At present, a redefinition of risk stratification has been proposed based on the study of altered genes during oncogenesis [17, 18]. Actually, a redefinition of risk stratification has been proposed based on the study of altered genes during oncogenesis [17].

JAK-STAT Pathway

The JAK-STAT pathway consists of Janus family protein tyrosine kinases JAK1, JAK2, JAK3 and and TYK2 and a family of seven cytosolic transcription factors, and made up of STAT (Signal transducers and transcription activators), a complex of six subtypes including 1, 2, 3, 4, 5a, 5b, and 6 with the same activation process mediated by cytokine and other stimuli. Several hematologic malignancies appear to be driven by molecular aberrations that affect the JAK-STAT pathway, including JAK2 and JAK1 mutations [19, 20], leading to the targeted development of small molecules that might control deregulated signaling [21].

JAK mutations have been described in high-risk chALL; in mutated cases a gene expression signature similar to BCR-ABL pediatric ALL had been identified. The results of Mullighan et al. suggest that JAK signaling inhibition is a target for therapeutic intervention in JAK mutated ALL; the authors reported activating mutations in JAK1 [3], JAK2 [16], and JAK3 [1], which represent 10.7 % of high-risk chALL cases negative to BCR-ABL [22].

Patients with JAK mutations have a poor outcome; the primary structure of JAK 1, -2 and -3 exhibit different changes; including missense, insertions and deletion mutation [21]. JAK1 is activated by a wide variety of cytokines; JAK 1, -2 and -3 are ubiquitously expressed and interact with many different cytokine receptors [2326]. Based on this data, JAK inhibitors could be used in these hematological diseases, in that they avoid the proliferation of malignant cells [27, 28].

JAK1 in Cytokine-Induced Biologic Responses Associated with chALL

JAK1 affects a wide variety of interleukins (IL), including IL-2, -4, -6, -7, -9, -10, -15, the Leukocyte inhibitory factor (LIF) and all interferons that participate in the hematopoiesis process; JAK1 mutations impair lymphoid development and defective responses to class 2 cytokines and those using gp130 receptor subunits [21, 23]. Cytokines that bind to class I or class II cytokine receptors employ the JAK-STAT signaling pathway [23].

JAK2 in Cytokine Responses and Erythropoietin Receptor Activity

Janus-kinase 2 (JAK2) is required for the function of a variety of cytokine receptors. In addition, the mutations of JAK2 affects the erythropoiesis process and inhibits the correct evolution [29]. Initial studies that suggested the JAK2-EPO relation reported an important number of kinases that have been implicated in signaling [30]. JAK2 gives rise to a regulator effect in EPO, Thrombopoietin (TPO), IL-3, IL-5, (GM-CSF), and Interferon gamma (IFN-γ) expression [29, 31].

JAK2 is constitutively bound to cell surface erythropoietin receptor (EpoR) and is crucial in signaling that include the growth hormone receptor [32]. JAK2 is crucial for EpoR signaling; JAK2 phosphorylated tyrosine residues in EpoR cause docking sites for SH2 and subsequently the activation of STAT5, Ras, mitogen-activated protein kinase (MAPK), JNK, P38, PI3-kinase–AKT, SHP1, SHP2, SHIP, and BCL-xL [33].

A JAK2 inhibitor such as Ruxolitinib has been reported as an active therapeutic in preclinical models of Polycythemia vera (PV) and other myeloproliferative neoplasms associated with somatic gain-of-function JAK2 mutations [34].

The fusion of JAK2 to PCM1 is a product of the translocation t (8; 9)(p22; p24); an uncommon case classified as myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB or FGFR1, in this case there are an aberrant tyrosine kinase expression [35].

Yu et al. [36, 37] tested JAK2 inhibitors with a purified enzyme assays in a high-throughput using a line of Ba/F3 cells that expressing individually the translocated ETS leukemia (TEL) fusions of each JAK family member (TELJAKBa/F3). This technique should provide a more meaningful understanding of selectivity and facilitate the development of additional JAK inhibitors.

JAK3 in the Lymphoid Development

JAK3 mutations are associated with defective lymphoid development, deregulated myelopoiesis, and the affectation of different cytokines, such as IL-4, -7, -9, and -15 [38, 39]. Signaling regulation of proliferation and apoptosis by JAK3 has been found in the physiopathology of different leukemia types, including acute megakaryoblastic leukemia and T-cell lymphoma, and PEPT1 and -2 (peptide transporters) are regulated by JAK3, a powerful regulator of these peptide transporters [40].

Ross et al. [41] have tested JAK3 inhibitors considering the important role of JAK3 in T-cell malignancies, including T-cell ALL (T-ALL), and the authors used a selective and orally active small molecule with high rates of effectiveness to induce therapeutic response in T-cell malignancies with less toxic therapies.

Mutations in receptor of Interleukin 7 (IL7R) and others altered cytokine IL-7 stimulate the differentiation of multi-potent hematopoietic stem cells (HSC) from lymphoid lineage. The receptor of Interleukin 7 (IL-7R) which is formed by IL-7Rα (encoded by IL-7R) is essential for normal T-cell development and homeostasis [42]. IL-7 plays an important role in the development and homeostasis of cells and is involved in the proliferation, maturation and survival of of B, T and Natural Killer (NK) cells (Table 2).

Ott et al. [43] have investigated the chemical compound JQ1 as possible therapeutic option as it reduces the viability of B-ALL cell line through the BET protein inhibition especially in patients with high-risk cytogenetic. Therefore plasma concentration of IL-2 receptor, IL-8, -12 and -15 or C–X–C motif chemokine 10 (CXCL10) in primary myelofibrosis is important due to their association with prognostic. In addition to these cytokines, IL-10 and CXCL9 have been included in the prognosis of large-cell lymphoma [44, 45].

Signaling of TYK2 in ALL

Tyk2 is the last JAK family member with an effect mediated by. Recently, Sanda et al. [46] utilized RNA interference (RNAi) to identify the TYK2-STAT1 pathway in T-ALL, promoting it as a novel oncogenic pathway that upregulates BCL2 expression.

STATs

STAT is a protein complex with an important role in the regulation of hematopoiesis mediated by cytokine signal transduction [47, 48]. The majority of critical processes are regulated by STAT, including cellular proliferation and survival. To our knowledge, STAT activation is mediated by a receptor complex, and it translocates to modify nuclear gene expression [21].

STAT1

A region required of the EpoR for the STAT1 has been identified in a JAK2 dependent process; this transcription factor is activated after EPO treatment and is mediated by the Tyr 432 residue of the human EpoR gene, suggesting its role in the development of normal erythropoiesis [47]. Thus, Interferon (INF) and innate immune responses are associated with the regulation of STAT1 expression [21].

STAT2

Alterations in STAT2 affect the type 1 INF response [21].

STAT3

STAT3 inhibitors have been employed with high effectiveness levels in cellular lines. STAT3 plays an important role in signaling pathways related with oncogenic processes. Despite that in normal cells STAT inhibitors do not demonstrate significant effects, they continue to be molecular targets in malignant cells [48]. In other hematological disorders that affect myeloid linage, the BCR-ABL activates STAT3 in human cells mediated by the JAK pathway including MEK. Coppo et al. [49, 50] established that the activation mechanism of STAT3 has been mediated by JAK and Erk/MAP-kinase, leading to the increase of STAT3 mRNA, which should contribute to CML progression and to increased risk in patients with the Ph chromosome.

STAT4

This STAT is associated with IL-12 response, IFN γ-Rα production by Th1 cells, lymphocyte proliferation, and other Th2 cell responses [21].

STAT5 a/b

STAT5 a/b affects the proliferation signaling, including CFU-Mix, Eos, G, GM, and the absence of NK cells [21]. The active forms, A and B, are mediators of the oncogenic process that utilize a similar mechanism to that of STAT3. In essays employing Pimozide to evaluate the in vitro colony formation of healthy cells, there have been favorable results without hematopoietic toxicity. STAT5 is associated with a normal hematopoiesis process mediating cytokine responses; this latter notion supports the concept that healthy cells are not affected by STAT5 inhibitors, an interesting target in ALL [48].

STAT6

Mutations of this STAT have been identified in cases of IL-4 response failure; these Th2 cell affectations should be reduced simultaneously the lymphocyte proliferation [21]. In fact, in mice models the induction of STAT6 deficiencies affect the biological responses in B cells associated with IL-4 levels [51].

The PI3K-AKT/Ras-MAPK Pathway

RAS proteins are regulated by guanine-nucleotide-exchange factors prior to stimulation of the nucleotide process. Mitogen-activated protein kinase (MAPK) and Phosphatidylinositol 3-kinase (PI3K) pathways are regulated by the formation of an intracellular docking site able to regulate cell proliferation, organization, and survival [52].

The RAS gene family includes KRAS, HRAS, and NRAS genes with similar effects in the development of cell cycle, although Ras genes have specific differences due to the presence of a posttranslational modification [52].

The PI3K-AKT and Ras-MAPK prosurvival signaling pathways are required for normal homeostasis in nonmalignant cells, but there is a continued reliance or “addiction” by leukemia cells on these pathways that has made them popular for targeted therapies. The PI3K-Akt pathway is often constitutively upregulated in many lymphoid malignancies (reviewed by Lee-Sherick et al.) [53], leading to the development of targeted therapies against the mammalian Target of Rapamycin (mTOR) and the TOR complexes [54, 55]. Inhibition of mTOR has been found to be effective in pediatric ALL [55].

The interaction between the PI3K-Akt pathway and the active forms STAT5a and STAT5b is mediated by Grb2 growth factor receptor bound protein 2 (Grb2). There are two different important roles of Gab2 in hematological disorders; Gab2 is associated with the activation PI3K/Akt and Ras/MAPK pathway mediated by STAT5 protein activation, and with ERK and Akt activation [56].

Inhibition of the PI3K/AKT/mTOR pathway has been a therapeutic tool in ALL; unfortunately, resistance to therapy is emerging. The use of selective inhibitors of this pathway, such as NVP-BKM120, RAD001, BEZ235, and NVP-BGT226, were compared in the cell cultures of ALL cells and probed in an experimental design; in this case, the combined use of dual PI3K/mTOR inhibitors provided an interesting therapeutic approach, especially in patients with the ALL linage of pre-B cells [57].

Glycogen Synthase Kinase-3β (GSK-)—NF-κB Pathway

Glycogen synthase kinase-3β (GSK-3β) has recently been found to positively regulate the activity of Nuclear factor-kappa B (NF-κB). The association between GSK-3β inhibition and NF-κB has been studied in pediatric primary leukemia cells obtained from newly diagnosed children with ALL. Hu et al. [58] isolated Bone marrow mononuclear cells (BMMC) by density gradient centrifugation from the heparinized aspirates of children with ALL. This group utilized immunofluorescence staining to detect nuclear GSK-3β in these cells, as well as western blot and electrophoretic mobility shift assays, testing the GSK-3β inhibitor in vitro treatment. By using this approach, these authors showed that inhibition of GSK-3β downregulates the NF-κB activation pathway, leading to suppression of the expression of an NF-κB-regulated gene and the promotion of apoptosis in ALL cells in vitro.

Therefore, GSK- is a multi-faceted kinase that is regulated by different growth factor signaling pathways and that affects a diverse range of physiological processes. Although crosstalk among signaling pathways is a feature of development, homeostasis, and disease, it is widely accepted that the Wnt pathway is associated with other signaling pathways: in Wnt-stimulated cells, inhibition of GSK-3β exerts a profound influence on β-catenin and Axin phosphorylation, but does not substantially affect the phosphorylation of other GSK-3β target substrates, such as tau and glycogen synthase [59].

The mTOR and GSK- cooperate to control the activity of S6K1, an important regulator of cell proliferation and growth [60]. The 40S ribosomal protein S6 kinase (S6K) is a major substrate of mTOR and is a crucial effector of mTOR signaling [61]. One of the S6 K isoforms, S6K1, plays important roles in cell growth, proliferation, and cell differentiation by regulating ribosome biogenesis, protein synthesis, cell cycle progression, and metabolism [62, 63].

Hypoxia-Inducible Transcription Factor-1α (HIF-)—GSK- Pathway

Hypoxia-inducible transcription factor-1α (HIF-1α) is a major regulator of carcinogenesis and various processes by which cells adapt to hypoxic conditions, being an important target for the understanding of angiogenesis and different cancer phenotypes and for unraveling new therapeutic options [61]. Flüel et al. [64] proposed an interactive mechanism between HIF- and GSK-3 across induced phosphorylation and recruitment of the ubiquitin ligase and tumor suppressor F-box and WD protein Fbw7.

CBFA2/RUNX1

RUNX1 is a runt-related transcription factor 1 also known as CBFA2 (corebinding factor α2) or AML1 (acute myelogenous leukemia 1). RUNX1 is a heterodimeric transcription factor with multiple splice variants that plays a role in normal hematopoiesis. Defects in RUNX1 are associated with several types of leukemia.

At present, detection of human RUNX1/CBFA2 by Western blot comprises a necessary tool to determine this frequent translocation variant between intron 5 of TEL and intron 2 of AML [65].

Mutations in RUNX1 have been identified in T-ALL. Dowdy et al. [66] induced triple-point mutations in mouse model to characterize the effects in different hematopoietic lineages. In this case, induction with a RUNX1 HTY350-352AAA causes several alterations in embryonic development mediated by a mechanism related with transactivation and co-factor interaction TGF (Tumor growth factor)-b/BMP pathway. In specific domains of RUNX1, mutations have been reported that provide abnormal subnuclear targeting and interaction with co-factor processes, inducing alterations in early stages of hematopoiesis and leading to a pre-leukemic phenotype; this punctual mutation is induced by RUNX1 HTY350-352AAA, and the results indicate the crucial role of this transcription factor in the control and development of definitive hematopoiesis at different stages and its importance as a therapeutic target.

Moreover, RUNX1 is not exclusively associated with ALL, as it is also important in myeloid neoplasm suppression. In mouse model, overexpression of RUNX1 inhibited the growth of normal umbilical cord-blood cells and promoted the growth of AML1-ETO. These results are promising for AML treatment [67].

Lymphoid Transcription Factor IKZF1 (IKAROS)

The lymphoid transcription factor IKZF1 (IKAROS) has been associated with poor outcome in patients with ALL. Currently, determination of abnormalities in IKZF1 and other genes by multiplex ligation-dependent probe amplification contributes to promoting the physiopathology and prognosis of the disease [68].

HOXA Cluster

HOX genes have importance in normal development of lymphopoiesis and leukemogenesis processes and there are two classes of HOX genes class I and class II. The first group includes four clusters (HOXA, HOXB,HOXC and HOXD) and the second group TLX1 and TLX3. HOXA, TLX1 and TLX3 have been strongly associated with T- ALL development [15, 69].

Starkova et al. [69] analyzed 61 pediatric samples with ALL diagnosis and quantified RNA expression of HOX genes associated with ALL development: HOXA, HOXB, and CDX1/2 using qRT-PCR. Considering the diagnosis, the patients were analyzed in karyotype subgroups; in BCP-ALL and T-ALL patients expressed HOXA 34, HOXA 7, and HOXB 34 genes including abnormal karyotype as MLL/AF4, TEL/AML1 and BCR/ABL. In immature cells and progenitors were identified HOXA 710, 13 and HOXB 24, moreover HOXB 6 and CDX 2 were detected only in leukemic cells. Therefore, they found a particular expression in each karyotype anomaly. Information related to the most common caryotype associated to chALL is shown in Table 3.

Table 3.

Karyotype abnormalities associated with chALL development

Karyotype abnormalities Affected genes Phenotype abnormalities Detection method Frequency Ref.
Ch. A.
Hyperdiploid 4, 6, 10, 14, 17, 18, 21 and X More than 50 chromosomes. Common. Patients present mutations in the receptor of tyrosine kinase FLT3. Cytogenetic techniques; karyotype analysis, FISH, PCR 15 % 6 % [15, 81, 82]
Hypodiploidy Less than 45 chromosomes. Uncommon. [15]
t(12;21)(p13;q22) TEL-AML1 (ETV6-RUNX1) Good prognosis. Marked sensitivity asparaginase. 22 % 2 % [6, 15, 16, 65]
t(9;22)(q34;q11.2); Ph chromosome. BCR-ABL1 Poor prognosis. Hyperleukocytosis. 3 % 30 % [6, 15]
t(1;19)(q23;p13.3) E2A- PBX1 (TCF3-PBX1) Pre-B phenotype hyperleukocytosis. Need intensive treatment 5 % 3 % [6, 15]
t(4; 11) MLL-AF4 Associated with infant ALL. Hyperleukocytosis. Poor prognosis 8 % 10 % 6,
t(11; 19) Poor prognosis. Relatively favorable for children with T- cell ALL. 8 % 10 % [15]
t(5;14)(q31; q32) IL3- IGH
TLX3 (HOX11L2)
Poor outcome in T-chALL 20 %
T-ALL
13 %
T-ALL
[15]
1p32, t(1;14)(p32;q11) and t(1;7)(p32;q34) TAL1 Is not expressed in normal T- cell development.
Nonrandom genetic defect in T-chALL
Cytogenetic techniques; karyotype analysis, FISH, multiplex FISH, SKY, CGH, Microarray analysis 40 %
T-ALL
[15, 76]
1p34, t(1;7)(p34;q34) Lymphocyte-specific protein tyrosine kinase; LCK LCK is highly expressed in T-cells; it has interactions with TCR signaling [15]
8q24 MYC Juxtaposing of C-MYC [15]
9q34, t(7;9)(q34;q32) TAL2
TAN1/NOTCH1
TAL2 Is not expressed in normal T- cell development. Both proteins promote T-ALL <2 %
T-ALL
[15, 76]
10q24 HOX11 Overexpression of HOX11 in 10q24 absence. Alterations in normal leukemogenesis. [15]
t(11;14)(p15;q11) LMO1 (RBTN1 or TTG1, 11p15) LMO1 or LMO2 is strictly required to induce T cells malignances in co-expression with TAL1 45 %
T-ALL
[15]
t(11;14)(p13;q11) LMO2 ((RBTN2 or TTG2, 11p13) [15]
14q32 TCL1 Control of immunoglobulin (Ig) gene enhancers of the heavy chain (IGH- 14q32) [15, 76, 8388]
19p13, t(7;19)(q35;p13) LYL1 Is not expressed in normal T- cell development.
LYL1 is expressed in some types of leukemia is possible that it participates in T-cell leukemogenesis
[15]
t(8;14)(q24;q32) MYC/IG Burkitt lymphoma mature B-ALL (BL) 5 % 10 % [15]
t(10;14)(q24;q11), t(7;10)(q35;q24) TLX1 (HOX11) Nonrandom alteration identified in T-ALL 5 %
T-ALL
30 %
T-ALL
[15, 89, 90]
inv(7)(p15q34), t(7;7)(p15;q34), t(7;14)(p15;q11) HOXA@ cluster Elevated HOXA10 and HOXA11 expression in developing thymocytes. Participate in positive development of CD4 and CD8 5 %
T-ALL
[15, 91]
Duplication and t(6;7)(q23;q34) MYB T-cell differentiation. Microarray analysis 8–15 %
T-ALL
[15]

Ref. reference, A. adult’s prevalence, Ch. Children’s prevalence, FISH fluorescence in situ hybridization, SKY spectral karyotypes analysis, CGH comparative genomic hybridization

Abnormal expression of HOXA9 has been correlated with the development of T-cell and myeloid leukemia. However, the study of overexpression in hematopoietic tissues demonstrated the HOXA9 role in accelerated lymphoid, but not in myeloid, leukemia. Transformation utilizing Vav regulatory elements in mouse model has provided valuable information about the overexpression of other HOXA cluster genes; however, lower levels of HOXA9 stimulate the development of a T-cell precursor to lymphoblastic leukemia [70]. Finally, HOX11 activation has been associated with a favorable prognosis in T-ALL [71].

LMO1 and LMO2

The presence of chromosomal rearrangements in ALL is common: LMO1 and LMO2 overexpression in the loci of the T-cell receptor has been found at around 45 % of total cases. Therefore, LMO1 and -2 encode in the LIM-domain: 11p15 and 11p13, respectively. LMO proteins are associated with LYL1 or TAL1 dysregulation and the latter is associated with the induction of T-cell malignancies in co- expression with TAL1 [15, 72]. LMO2 has been associated with other chromosomal aberrations, specifically with TAL1 in a same cluster, suggesting that LMO2 and TAL1 rearrangements affect similar pathways. In a cohort of 117 samples of patients with an ALL diagnosis, Homminga et al., by Fluorescent in situ hybridization (FISH) and real-time quantitative PCR (RQ-PCR), the authors found, in their results, that LYL1 translocation and LMO2 rearrangement conform a TAL-LMO subgroup present in immature cells [73]. Moreover, LMO2 gene expression in mouse model has been associated with the development of T-ALL [74].

TAL1 and TAL2

TAL1 (also known as SCL) is not expressed in normal T-cell development, but is rather a non-random genetic defect in T-chALL; moreover, TAL1 gene expression in mouse model has been associated with T-ALL development [74]. TAL1/SCL activation in patients with ALL has been identified in 50–60 % [71]. In leukemogenesis, the TAL1/SCL product could interfere in E2A and HEB transcriptional activity. Thus, Kusy et al. [75] characterized the oncogenic pathways associated with TAL1 activation, corroborating that in T-ALL cells, NKX3.1 is activated by TAL1 in fusion with THE LMO-Ldb1 complex, causing a suppression of heterochromatin protein 1 (HP1) and subsequently the NKX3.1 gene promoter, the latter required for T-ALL proliferation. The authors conclude that TAL1 or NKX3.1 knockdown inhibit the capacity of T-ALL cells to induce leukemia in mouse model. TAL2 is not expressed in normal T-cell development and promotes T-ALL [15, 76]. TAL2 is a helix-loop helix protein, such as TAL1 and LYL1, and is a mediator of T-cell leukemogenesis that is identified in T-ALL [77].

Lymphocyte-Specific Protein Tyrosine Kinase (LCK)

Lymphocyte-specific protein tyrosine kinase (LCK) is predominantly expressed in T cells. LCK gene location is susceptible to chromosomal aberrations and, as a Src family kinase member, it activity is regulated by tyrosine phosphorylation at Tyr 394 and Tyr 505 [78]. LCK has been associated with a deregulation of the tyrosine kinases family and the regulation of the LIM domain. The constitutive activation of LCK has been related with ALL and other lymphoid malignancies. LCK regulates LMO2 expression and mediates a similar mechanism in the nuclear Janus kinase 2 (JAK2). Overexpression of LCK is implicated in hematological malignances [79].

TAN1/NOTCH1 and LYL1

NOTCH1 is another oncogene, like TAL1 and LMO2. In mouse model, the relation between NOTCH1 and LIC was established through treatment with a vehicle of the γ-secretase inhibitor [74]. The role of NOTCH1 in the development of T-ALL is mediated by its influence in signaling pathways, encoding a transmembrane receptor on HSC that induces proteolysis cleavage, allowing the intracellular-to- nucleus translocation of NOTCH1 followed by the activation of HES1 (Hairy/enhancer of split) and Deltex (DTX1), both present in about 50 % of T-ALL cases [52]. Furthermore, LYL1 is a basic Helix-loop-helix protein (bHLH) that is non-expressed in normal T-cell lineages. LYL1 ectopic expression is caused by juxtaposition on chromosome 19 and chromosome 7 T-cell receptor t(7; 19). The LYL1 structure contains a bHLH domain and a conserved region with binding sites [80]. LYL1 is a transcription factor that exhibits protein homology with TAL1, which is also overexpressed in hematopoietic malignancies [73].

In conclusion, there are under development at present an extensive number of inhibitors of molecular targets associated with intracellular signaling pathways in chALL (Table 4). Throughout this review, we compiled information on the main signaling pathways involved in ALL, one of the most diverse and complex diseases that greatly affect the child population in Mexico and worldwide.

Table 4.

Trials of therapeutic targets in ALL

Trial Mechanism/target Phase of trial/disadvantages Ref.
Recombinant human CD19L-sTRAIL fusion protein Heterogeneous expression of surface TRAIL receptors in patients with B cell precursor ALL. Preclinical studies of CD19L-sTRAIL in rodents/toxicity [92]
mTOR inhibitors Decreased leukemia proliferation and growth in pre B-ALL Preclinical studies in mice models [54, 55, 57]
Preclinical setting treating individual patient-derived ALL in vivo [93]
Synergize with histone deacetylase inhibitors to kill B-ALL. Preclinical studies in Balb/c and C57Bl/6 mice [94]
GSK- inhibitors Regulate the NF-κB transcriptional activity. Tests in cell culture obtained from bone marrow mononuclear cells and mice models/Drug toxicity [58, 59]
Sphingosine kinase inhibitors Regulate the autophagy and apostosis in T-cell ALL Cell culture and primary samples [95]
Epigenetic drugs: histone 3 lysine 27 demethylases, JMJD3 and UTX The JMJD3 modulation affects the initiation and maintenance of ALL, modulate H3K27 methylation Tests in a knockout mouse models of JMJD3 and UTX [96]
VLA-5 blocking VLA-5 block may increase susceptibility to therapy in BCL/ABL for Ph+ ALL treatment Tests in human bone marrow stromal cell line HS-5 [97]

Ref. reference

The presence of fusion translocation products and other cytogenetic abnormalities and their role as protein markers, such as LMO1, LMO2, LCK, TAL1, TAL2, TAN1/NOTCH1, LYL1, TCL1, and MYB, have already been extensively described and are crucial to the understanding and development of new therapies given its frequency in the appearance of chALL.

Concerning the widely described and unconventional intracellular signaling pathways, extensive evidence supporting the key role of JAK-STAT pathway point mutations in chALL pathophysiology provides the key to some family members as oncogenic mediators against which are directed the majority of therapeutic targets. However, although the treatment response rate in pediatric population is favorable at present, it is important to continue the characterization of these, especially STAT3 and STAT5 transcriptional activity as JAK proteins effectors, including the recently identified regulators of action.

Another important topic of lymphoid malignancy comprises the determination of molecular prognostic factors that could establish the likelihood of relapse more accurately, including the HOXA cluster. LYL1 and other proteins implicated in cell maturation might be useful as molecular risk factors, at least in T-cell malignancies. NKX3.1 and its relationship with the TAL1 protein as related with the proliferation of T-cells in ALL have gained prominence for its role in leukemogenesis.

Finally, we must take GSK-3β inhibitors into account as potential therapeutic targets, not only in hematologic malignancies but also in metabolic processes, including alterations in the signaling pathways of insulin and Alzheimer. Much research remains to investigate the involvement of cellular pathways in leukemia, and probably a single molecule will not be the gold standard for treatment or prognosis, but rather a mixed expression of many of these.

Compliance with Ethical Standards

Conflict of interest

The authors declare that they have no conflict of interest.

Research involving human participants and/or animals

This article does not contain any studies with human participants or animals performed by any of the authors.

Informed consent

For this type of study formal consent is not required.

References

  • 1.Silverman LB. Balancing cure and long-term risks in acute lymphoblastic leukemia. Hematology. 2014;25(1):190–197. doi: 10.1182/asheducation-2014.1.190. [DOI] [PubMed] [Google Scholar]
  • 2.Campbell K, Gerscher S, Siclair L. Childhood acute lymphoblastic leukaemia. Leukaemia Research Fund. 2001;8(1):1–10. [Google Scholar]
  • 3.Pacheco C, Lucchini G, Valsecchi MG, Malta A, Conter V, Flores A, et al. Childhood acute lymphoblastic leukemia in Nicaragua: long-term results in the context of an international cooperative program. Pediatr Blood Cancer. 2014;61(5):827–832. doi: 10.1002/pbc.24871. [DOI] [PubMed] [Google Scholar]
  • 4.Smith M, Arthur D, Camitta B, Carroll AJ, Crist W, Gaynon P, et al. Uniform approach to risk classification and treatment assignment for children with acute lymphoblastic leukemia. J Clin Oncol. 1996;14(1):18–24. doi: 10.1200/JCO.1996.14.1.18. [DOI] [PubMed] [Google Scholar]
  • 5.Harrison CJ. Targeting signaling pathways in acute lymphoblastic leukemia: new insights. ASH Educ Book. 2013;1:118–125. doi: 10.1182/asheducation-2013.1.118. [DOI] [PubMed] [Google Scholar]
  • 6.Friedmann AM, Weinstein HJ. The role of prognostic features in the treatment of childhood acute lymphoblastic leukemia. Oncologist. 2000;5:321–328. doi: 10.1634/theoncologist.5-4-321. [DOI] [PubMed] [Google Scholar]
  • 7.Winter SS (2011) Pediatric acute leukemia therapies informed by molecular analysis of high-risk disease. Hematology/the Education Program of the American Society of Hematology. American Society of Hematology Education Program, pp 366–73 [DOI] [PubMed]
  • 8.Protocolo de la atención para leucemia linfoblástica. guía clínica y esquema de tratamiento. Instituto nacional de salud pública, validado por el consejo de salubridad general, los institutos nacionales de salud y la comisión nacional de protección social en salud. Seguro Popular 1(1):1–20
  • 9.Wheeler KA, Richards SM, Bailey CC, Gibson B, Hann IM, Hill FG, et al. Bone marrow transplantation versus chemotherapy in the treatment of very high-risk childhood acute lymphoblastic leukemia in first remission: results from Medical Research Council UK ALL X and XI. Blood. 2000;96:2412–2451. [PubMed] [Google Scholar]
  • 10.Waber DP, Carpentieri SC, Klar N, Silverman LB, Schwenn M, Hurwitz CA, et al. Cognitive sequelae in children treated for acute lymphoblastic leukemia with dexamethasone or prednisone. J Pediatr Hematol Oncol. 2000;22:206–213. doi: 10.1097/00043426-200005000-00004. [DOI] [PubMed] [Google Scholar]
  • 11.Cao Y, Lupo PJ, Swartz MD, Nousome D, Scheurer ME. Using a bayesian hierarchical model for identifying single nucleotide polymorphisms associated with childhood acute lymphoblastic leukemia risk in case-parent triads. PLoS One. 2013;8(12):1–5. doi: 10.1371/journal.pone.0084658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Aziz SA, Sharma SK, Sabah I, Jan MA. Prognostic significance of cell surface phenotype in acute lymphoblastic leukemia. South Asian J Cancer. 2015;4(2):91–94. doi: 10.4103/2278-330X.155696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ross ME, Mahfouz R, Onciu M, Liu HC, Zhou X, Song G, et al. Gene expression profiling of pediatric acute myelogenous leukemia. Blood. 2004;104:3679–3687. doi: 10.1182/blood-2004-03-1154. [DOI] [PubMed] [Google Scholar]
  • 14.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;111:5477–5485. doi: 10.1182/blood-2008-01-132837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Roganovic J, Guenova M, Fuchs O, Abdul G, Schuurhuis GJ, Zeijlemaker W et.al. Leukemia, 1ra Ed. ISBN 978-953-51-1127-6
  • 16.Linka Y, Ginzel S, Krüger M, Novosel A, Gombert M, Kremmer E, et al. The impact of TEL-AML1 (ETV6-RUNX1) expression in precursor B cells and implications for leukaemia using three different genome-wide screening methods. Blood Cancer J. 2013;11(3):151. doi: 10.1038/bcj.2013.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang J, Mi JQ, Debernardi A, Emadali AL, Emadali A, Meyer JA, et al. A six gene expression signature defines aggressive subtypes and predicts outcome in childhood and adult acute lymphoblastic leukemia. Oncotarget. 2015;6(18):16527–16542. doi: 10.18632/oncotarget.4113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mullighan CG, Phillips LA, Su X, Ma J, Miller CB, Shurtleff SA, et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science. 2008;322:1377–1380. doi: 10.1126/science.1164266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.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;115:5393–5397. doi: 10.1182/blood-2009-11-256131. [DOI] [PubMed] [Google Scholar]
  • 20.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;372:1484–1492. doi: 10.1016/S0140-6736(08)61341-0. [DOI] [PubMed] [Google Scholar]
  • 21.Ward A, Touw I, Yoshimura A. The Jak-Stat pathway in normal and perturbed hematopoiesis. Blood. 2000;95:19–29. [PubMed] [Google Scholar]
  • 22.Mullighan C, Zhang J, Harvey R, Collins-Underwood J, Schulman B, Phillips L, et al. JAK mutations in highrisk childhood acute lymphoblastic leukemia. Proc Natl Acad Sci USA. 2009;9(106):9414–9418. doi: 10.1073/pnas.0811761106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rodig SJ, Meraz MA, White JM, Lampe PA, Riley JK, Arthur CD, et al. Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-in- duced biologic responses. Cell. 1998;93:373–383. doi: 10.1016/S0092-8674(00)81166-6. [DOI] [PubMed] [Google Scholar]
  • 24.Darnell JE, Kerr IM, Stark GR. Jak-STAT path- ways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–1421. doi: 10.1126/science.8197455. [DOI] [PubMed] [Google Scholar]
  • 25.Ihle JN, Witthuhn BA, Quelle FW, Yamamoto K, Silvennoinen O. Signaling through the hematopoietic cytokine receptors. Annu Rev Immunol. 1995;13:369–398. doi: 10.1146/annurev.iy.13.040195.002101. [DOI] [PubMed] [Google Scholar]
  • 26.O’Shea JJ, Leonard WJ. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science. 1995;270:797–800. doi: 10.1126/science.270.5237.797. [DOI] [PubMed] [Google Scholar]
  • 27.Lo MC, Peterson LF, Yan M, Cong X, Hickman JH, Dekelver RC, et al. JAK inhibitors suppress t(8;21) fusion protein-induced leukemia. Leukemia. 2013;27:2272–2279. doi: 10.1038/leu.2013.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mullighan C (2012) The molecular genetic makeup of acute lymphoblastic leukemia, 1st edn. ASH Educ Book 1:389–396 [DOI] [PubMed]
  • 29.Parganas E, Wang D, Stravopodis D, Topham DJ, Marine JC, Teglund S, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385–395. doi: 10.1016/S0092-8674(00)81167-8. [DOI] [PubMed] [Google Scholar]
  • 30.Neubauer H, Cumano A, Muller M, Wu H, Huffstadt U, Pfeffer K. Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell. 1998;93:397–409. doi: 10.1016/S0092-8674(00)81168-X. [DOI] [PubMed] [Google Scholar]
  • 31.Silvennoinen O, Witthuhn B, Quelle FW, Cleveland JL, Yi T, Ihle JN. Structure of the JAK2 protein tyrosine kinase and its role in IL-3 signal transduction. Proc Natl Acad Sci. 1993;90:8429–8433. doi: 10.1073/pnas.90.18.8429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yeh TC, Pellegrini S. The Janus kinase family of protein tyrosine kinases and their role in signaling. Cell Mol Life Sci. 1999;55:1523–1534. doi: 10.1007/s000180050392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Constantinescu SN, Ghaffari S, Lodish HF. The erythropoietin receptor: structure, activation, and intracellular signal transduction. Trends Endocrinol. 1999;10:18–23. doi: 10.1016/S1043-2760(98)00101-5. [DOI] [PubMed] [Google Scholar]
  • 34.Verstovsek S, Passamonti F, Rambaldi A, Barosi G, Rosen PJ, Rumi E, et al. A phase 2 study of ruxolitinib, an oral JAK1 and JAK2 inhibitor, in patients with advanced polycythemia vera who are refractory or intolerant to hydroxyurea. Cancer. 2013;120:513–520. doi: 10.1002/cncr.28441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Patterer V, Schnittger S, Kern W, Haferlach T, Haferlach C. Hematologic malignancies with PCM1-JAK2 gene fusion share characteristics with myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, and FGFR1. Ann Hematol. 2013;92:759–769. doi: 10.1007/s00277-013-1695-3. [DOI] [PubMed] [Google Scholar]
  • 36.Yu V, Pistillo J, Archibeque I, Han Lee J, Sun BC, Schenkel LB, et al. Differential selectivity of JAK2 inhibitors in enzymatic and cellular settings. Exp Hematol. 2013;41:491–500. doi: 10.1016/j.exphem.2013.01.005. [DOI] [PubMed] [Google Scholar]
  • 37.Ghaffari S, Kitidis C, Fleming M, Neubauer H, Pfeffer K, Lodish H. Erythropoiesis in the absence of januskinase 2: BCR-ABL induces red cell formation in JAK2−/− hematopoietic progenitors. Blood. 2001;98:2948–2957. doi: 10.1182/blood.V98.10.2948. [DOI] [PubMed] [Google Scholar]
  • 38.Nosaka T, van Deursen JM, Tripp RA, Thierfelder WE, Witthuhn BA, McMickle AP, et al. Defectivelymphoid development in mice lacking Jak3. Science. 1995;270:800–802. doi: 10.1126/science.270.5237.800. [DOI] [PubMed] [Google Scholar]
  • 39.Grossman WJ, Verbsky JW, Yang L, Berg LJ, Fields LE, Chaplin DD, et al. Dysregulated myelopoiesis in mice lacking Jak3. Blood. 1999;94:932–939. [PubMed] [Google Scholar]
  • 40.Warsi J, Hosseinzadeh Z, Dong L, Pakladok T, Umbach A, Bhavsar S, et al. Effect of Janus kinase 3 on the peptide transporters PEPT1 and PEPT2. J Membr Biol. 2013;246:885–892. doi: 10.1007/s00232-013-9582-3. [DOI] [PubMed] [Google Scholar]
  • 41.Ross JA, Spadaro M, Rosado DC, Cavallo F, Kirken RA, Pericle F. Inhibition of JAK3 with a novel, selective and orally active small molecule induces therapeutic response in T-cell malignancies. Leukemia. 2013;28(4):941–944. doi: 10.1038/leu.2013.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zenatti PP, Ribeiro D, Li W, Zuurbier L, Silva MC, Paganin M, et al. Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia. Nat Genet. 2011;43:932–939. doi: 10.1038/ng.924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ott CJ, Kopp N, Bird L, Paranal RM, Qi J, Bowman T. BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood. 2012;120:2843–2852. doi: 10.1182/blood-2012-02-413021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Pardanani A, Finke C, Lasho TL, Al-Kali A, Begna KH, Hanson C, et al. IPSS-independent prognostic value of plasma CXCL10, IL-7 and IL-6 levels in myelodysplastic síndromes. Leukemia. 2012;26:693–699. doi: 10.1038/leu.2011.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kornblau SM, McCue D, Singh N, Chen W, Estrov Z, Coombes KR. Recurrent expression signatures of cytokines and chemokines are present and are independently prognostic in acute myelogenous leukemia and myelodysplasia. Blood. 2010;116:4251–4261. doi: 10.1182/blood-2010-01-262071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sanda T, Tyner JW, Gutierrez A, Ngo VN, Glover J, Chang BH, et al. TYK2-STAT1-BCL2 pathway dependence in T-cell acute lymphoblastic leukemia. Cancer Discov. 2013;3:564–577. doi: 10.1158/2159-8290.CD-12-0504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kirito K, Nakajima K, Watanabe T, Uchida M, Tanaka M, Ozawa K, et al. Identification of the human erythropoietin receptor region required for Stat1 and Stat3 activation. Blood. 2002;99:102–110. doi: 10.1182/blood.V99.1.102. [DOI] [PubMed] [Google Scholar]
  • 48.Walker S, Frank DA. Screening approaches to generating STAT inhibitors: allowing the hits to identify the targets. JAK-STAT. 2012;1(4):292–299. doi: 10.4161/jkst.22662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nelson EA, Walker SR, Weisberg E, Bar-Natan M, Barrett R, Gashin LB, et al. The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood. 2011;117:3421–3429. doi: 10.1182/blood-2009-11-255232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Coppo P, Flamant S, De Mas V, Jarrier P, Guillier M, Bonnet ML, et al. BCR–ABL activates STAT3 via JAK and MEK pathways in human cells. Br J Haematol. 2006;134:171–179. doi: 10.1111/j.1365-2141.2006.06161.x. [DOI] [PubMed] [Google Scholar]
  • 51.Takeda K, Tanaka T, Shi W, Matsumoto M, Minami M, Kashiwamura S. Essential role of Stat6 in IL-4 signalling. Nature. 1996;380:627–630. doi: 10.1038/380627a0. [DOI] [PubMed] [Google Scholar]
  • 52.Kindler T, Cornejo M, Scholl C, Liu J, Leeman D, Haydu J, et al. K-RasG12D–induced T-cell lymphoblastic lymphoma/leukemias harbor Notch1 mutations and are sensitive to γ-secretase inhibitors. Blood. 2008;112:3373–3382. doi: 10.1182/blood-2008-03-147587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lee-Sherick AB, Linger RM, Gore L, Keating AK, Graham DK. Targeting paediatric acute lymphoblastic leukaemia: novel therapies currently in development. Br J Haematol. 2010;151:295–311. doi: 10.1111/j.1365-2141.2010.08282.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hidalgo M, Rowinsky EK. The rapamycin-sensitive signal transduction pathway as a target for cancer therapy. Oncogene. 2000;19:6680–6686. doi: 10.1038/sj.onc.1204091. [DOI] [PubMed] [Google Scholar]
  • 55.Avellino R, Romano S, Parasole R, Bisogni R, Lamberti A, Poggi V, et al. Rapamycin stimulates apoptosis of childhood acute lymphoblastic leukemia cells. Blood. 2005;106:1400–1406. doi: 10.1182/blood-2005-03-0929. [DOI] [PubMed] [Google Scholar]
  • 56.Nyga R, Pecquet C, Harir N, Gu H, Dhennin-Duthille I, Regnier A, et al. Activated STAT5 proteins induce activation of the PI 3-kinase/Akt and Ras/MAPK pathways via the Gab2 scaffolding adapter. Biochem J. 2005;390:359–366. doi: 10.1042/BJ20041523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Badura S, Tesanovic T, Pfeifer H, Wystub S, Nijmeijer B, Liebermann M, et al. Differential effects of selective inhibitors targeting the PI3K/AKT/mTOR pathway in acute lymphoblastic leukemia. PLoS One. 2013;8(11):e80070. doi: 10.1371/journal.pone.0080070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hu Y, Gu X, Li R, Luo Q, Xu Y. Glycogen synthase kinase-3 beta inhibition induces nuclear factor-kappaBmediated apoptosis in pediatric acute lymphocyte leukemia cells. J Exp Clin Cancer Res. 2010;29:154. doi: 10.1186/1756-9966-29-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ding VW, Chen RH, McCormick F. Differential regulation of glycogen synthase kinase 3beta by insulin and Wnt signaling. J Biol Chem. 2000;275:32475–32481. doi: 10.1074/jbc.M005342200. [DOI] [PubMed] [Google Scholar]
  • 60.Shin S, Wolgamott L, Yu Y, Blenis J, Yoon SO. Glycogen synthase kinase (GSK)-3 promotes p70 ribosomal protein S6 kinase (p70S6K) activity and cell proliferation. Proc Natl Acad Sci USA. 2011;108:1204–1213. doi: 10.1073/pnas.1110195108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Dann SG, Selvaraj A, Thomas G. mTOR Complex1-S6K1 signaling: at the crossroads of obesity, diabetes and cancer. Trends Mol Med. 2007;13:252–259. doi: 10.1016/j.molmed.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 62.Carnevalli LS, Masuda K, Frigerio F, Le Bacquer O, Um SH, Gandin V, et al. S6K1 plays a critical role in early adipocyte differentiation. Dev Cell. 2010;18:763–774. doi: 10.1016/j.devcel.2010.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, et al. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell. 2010;39:171–183. doi: 10.1016/j.molcel.2010.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Flügel D. Görlach, Kietzmann T. GSK-3β regulates cell growth, migration, and angiogenesis via Fbw7 and USP28-dependent degradation of HIF-1. Blood. 2012;119:1292–1301. doi: 10.1182/blood-2011-08-375014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Torrano V, Procter J, Cardus P, Greaves M, Ford A. ETV6-RUNX1 promotes survival of early B lineage progenitor cells via a dysregulated erythropoietin receptor. Blood. 2011;118:4910–4918. doi: 10.1182/blood-2011-05-354266. [DOI] [PubMed] [Google Scholar]
  • 66.Dowdyb C, Frederickb D, Zaidia S, Colbyb J, Liana J, Van Wijnenc J, et al. A germline point mutation in Runx1 uncouples its role in definitive hematopoiesis from differentiation. Exp Hematol. 2013;41:980–991. doi: 10.1016/j.exphem.2013.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Goyama S, Schibler J, Cunningham L, Zhang Y, Rao Y, Nishimoto N, et al. Transcription factor RUNX1 promotes survival of acute myeloid leukemia cells. J Clin Invest. 2013;123(9):3876–3888. doi: 10.1172/JCI68557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mullighan CG, Su X, Zhang J, Radtke I, Phillips LA, Christopher B, et al. 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]
  • 69.Starkova J, Zamostna B, Mejstrikova E, Krejci R, Drabkin HA, Trka J. HOX gene expression in phenotypic and genotypic subgroups and low HOXA gene expression as an adverse prognostic factor in pediatric ALL. Pediatr Blood Cancer. 2010;55:1072–1082. doi: 10.1002/pbc.22749. [DOI] [PubMed] [Google Scholar]
  • 70.Beachy SH, Onozawa M, Silverman D, Chung YJ, Rivera MM, Aplan PD. Isolated Hoxa9 overexpression predisposes to the development of lymphoid but not myeloid leukemia. Exp Hematol. 2013;41:518–529. doi: 10.1016/j.exphem.2013.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ferrando AA, Neuberg DS, Staunton J, Loh ML, Huard C, Raimondi SC, et al. Gene expression signatures define novel onco- genic 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]
  • 72.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]
  • 73.Homminga I, Vuerhard MJ, Langerak AW, Buijs-Gladdines J, Pieters R, Meijerink JP. Characterization of a pediatric T-cell acute lymphoblastic leukemia patient with simultaneous LYL1 and LMO2 rearrangements. Haematologica. 2012;97:258–261. doi: 10.3324/haematol.2011.051722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Tatarek J, Cullion K, Ashworth T, Gerstein R, Aster JC, Kelliher MA. Notch1 inhibition targets the leukemia initiating cells in a Tal1/Lmo2 mouse model of T-ALL. Blood. 2011;118(6):1579–1590. doi: 10.1182/blood-2010-08-300343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kusy S, Gerby B, Goardon N, Gault N, Ferri F, Gérard D, et al. NKX3.1 is a direct TAL1 target gene that mediates proliferation of TAL1-expressing human T cell acute lymphoblastic leukemia. J Exp Med. 2010;207:2141–2156. doi: 10.1084/jem.20100745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wadman I, Li J, Bash RO, Forster A, Osada H, Rabbitts TH, et al. 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]
  • 77.Baer R. TAL1, TAL2 and LYL1: a family of basic helix-loop-helix proteins implicated in T cell acute leukaemia. Semin Cancer Biol. 1993;4:341–347. [PubMed] [Google Scholar]
  • 78.Martin GS. The hunting of the Src. Nat Rev Mol Cell Biol. 2001;2:467–475. doi: 10.1038/35073094. [DOI] [PubMed] [Google Scholar]
  • 79.Venkitachalam S, Chueh FY, Yu CL. Nuclear localization of lymphocyte-specific protein tyrosine kinase (Lck) and its role in regulating LIM domain only 2 (Lmo2) gene. Biochem Biophys Res Commun. 2012;417:1058–1062. doi: 10.1016/j.bbrc.2011.12.095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Miyamoto A, Cui X, Naumovski L, Cleary ML. Helix-loop-helix proteins LYL1 and E2a form heterodimeric complexes with distinctive DNA-binding properties in hematolymphoid cells. Mol Cell Biol. 1996;16(5):2394–2401. doi: 10.1128/MCB.16.5.2394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pui CH, Carroll WL, Meshinchi S, Arceci RJ. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J Clin Oncol. 2011;29:551–565. doi: 10.1200/JCO.2010.30.7405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Armstrong SA, Look AT. Molecular genetics of acute lymphoblastic leukemia. J Clin Oncol. 2005;23:6306–6315. doi: 10.1200/JCO.2005.05.047. [DOI] [PubMed] [Google Scholar]
  • 83.García JL, Hernández JM, Gutiérrez NC, Flores T, González D, Calasanz MJ, et al. Abnormalities on 1q and 7q are associated with poor outcome in sporadic Burkitt’s lymphoma. A cytogenetic and comparative genomic hybridization study. Leukemia. 2003;17:2016–2024. doi: 10.1038/sj.leu.2403080. [DOI] [PubMed] [Google Scholar]
  • 84.Zunino A, Viaggi S, Ottaggio L, Fronza G, Schenone A, Roncella S, et al. Chromosomal aberrations evaluated by CGH, FISH and GTG-banding in a case of AIDS-related Burkitt’s lymphoma. Haematologica. 2000;85:250–255. [PubMed] [Google Scholar]
  • 85.Zimonjic DB, Keck-Waggoner C, Popescu NC. Novel genomic imbalances and chromosome translocations involving c-myc gene in Burkitt’s lymphoma. Leukemia. 2001;15:1582–1588. doi: 10.1038/sj.leu.2402281. [DOI] [PubMed] [Google Scholar]
  • 86.Salaverria I, Zettl A, Beà S, Hartmann EM, Dave SS, Wright GW, et al. Chromosomal alterations detected by comparative genomic hybridization in subgroups of gene expression-defined Burkitt’s lymphoma. Haematologica. 2008;93:1327–1334. doi: 10.3324/haematol.13071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Barth TF, Müller S, Pawlita M, Siebert R, Rother JU, Mechtersheimer G, et al. Homogeneous immunophenotype and paucity of secondary genomic aberrations are distinctive features of endemic but not of sporadic Burkitt’s lymphoma and diffuse large B-cell lymphoma with MYC rearrangement. J Pathol. 2004;203:940–945. doi: 10.1002/path.1596. [DOI] [PubMed] [Google Scholar]
  • 88.Toujani S, Dessen P, Ithzar N, Danglot G, Richon C, Vassetzky Y, et al. High resolution genome-wide analysis of chromosomal alterations in Burkitt’s lymphoma. PLoS One. 2009;4:7089. doi: 10.1371/journal.pone.0007089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hawley RG, Fong AZ, Reis MD, Zhang N, Lu M, Hawley TS. Transforming function of the HOX11/TCL3 homeobox gene. Cancer Res. 1997;57:337–345. [PubMed] [Google Scholar]
  • 90.Keller G, Wall C, Fong AZ, Hawley TS, Hawley RG. Overexpression of HOX11 leads to the immortalization of embryonic precursors with both primitive and definitive hematopoietic potential. Blood. 1998;92:877–887. [PubMed] [Google Scholar]
  • 91.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]
  • 92.Uckun FM, Myers DE, Qazi S, Ozer Z, Rose R, D’Cruz OJ, et al. Recombinant human CD19L-sTRAIL effectively targets B cell precursor acute lymphoblastic leukemia. J Clin Investig. 2015;125(3):1006–1018. doi: 10.1172/JCI76610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hasan M, Queudeville M, Trentin L, Mirjam S, Bronzini I, Palmi C, et al. Targeting of hyperactivated mTOR signaling in high-risk acute lymphoblastic leukemia in a pre-clinical model. Oncotarget. 2014;6(3):1382–1395. doi: 10.18632/oncotarget.2842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Beagle BR, Nguyen DM, Mallya S, Tang SS, Lu M, Zeng Z, et al. mTOR kinase inhibitors synergize with histone deacetylase inhibitors to kill B-cell acute lymphoblastic leukemia cells. Oncotarget. 2015;6(4):2088–2100. doi: 10.18632/oncotarget.2992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Evangelisti C, Evangelisti C, Teti G, Chiarini F, Falconi M, Melchionda F, et al. Assessment of the effect of sphingosine kinase inhibitors on apoptosis, unfolded protein response and autophagy of T-cell acute lymphoblastic leukemia cells; indications for novel therapeutics. Oncotarget. 2014;5(17):7886–7901. doi: 10.18632/oncotarget.2318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ntziachristos P, Tsirigos A, Welstead G, Trimarchi T, Bakogianni S, Xu L, et al. Contrasting roles for histone 3 lysine 27 demethylases in acute lymphoblastic leukemia. Nature. 2014;514(7523):513–517. doi: 10.1038/nature13605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hu Z, Slayton WB. Integrin VLA-5 and FAK are good targets to improve treatment response in the philadelphia chromosome positive acute lymphoblastic leukemia. Front Oncol. 2014;4:112. doi: 10.3389/fonc.2014.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Indian Journal of Hematology & Blood Transfusion are provided here courtesy of Springer

RESOURCES