Abstract
Chronic myeloid leukemia and systemic mastocytosis are myeloid neoplasms sharing a number of pathogenetic and clinical features. In both conditions, an aberrantly activated oncoprotein with tyrosine kinase activity, namely BCR-ABL1 in chronic myeloid leukemia, and mutant KIT, mostly KIT D816V, in systemic mastocytosis, is key to disease evolution. The appreciation of the role of such tyrosine kinases in these diseases has led to the development of improved therapies with tyrosine kinase-targeted inhibitors. However, most drugs, including new KIT D816V-blocking agents, have failed to achieve long-lasting remissions in advanced systemic mastocytosis, and there is a similar problem in chronic myeloid leukemia, where imatinib-resistant patients sometimes fail to achieve remission, even with second- or third-line BCR-ABL1 specific tyrosine kinase inhibitors. During disease progression, additional signaling pathways become activated in neoplastic cells, but most converge into major downstream networks. Among these, the AKT and STAT5 pathways appear most critical and may result in drug-resistant chronic myeloid leukemia and systemic mastocytosis. Inhibition of phosphorylation of these targets has proven their crucial role in disease-evolution in both malignancies. Together, these observations suggest that STAT5 and AKT are key drivers of oncogenesis in drug-resistant forms of the diseases, and that targeting STAT5 and AKT might be an interesting approach in these malignancies. The present article provides an overview of our current knowledge about the critical role of AKT and STAT5 in the pathophysiology of chronic myeloid leukemia and systemic mastocytosis and on their potential value as therapeutic targets in these neoplasms.Introduction
Several new targets have recently been identified in neoplastic mast cells (MCs), and various targeted drugs have been examined for their effectiveness in malignant MC disorders. However, most drugs, including new KIT D816V-blocking agents, such as midostaurin (PKC412),1 and various BCR-ABL1 inhibitors known to block KIT-activity, such as imatinib or dasatinib,2 have failed to achieve long-lasting remissions in aggressive systemic mastocytosis (ASM). There is a similar problem in Ph CML, where imatinib-resistant patients do not reach molecular remission even when second-or third-line BCR-ABL1 tyrosine kinase inhibitors (TKIs) are applied, particularly in patients exhibiting the BCR-ABL1 T315I mutant.3 During disease progression, additional signal transduction pathways become activated in neoplastic cells. Among these, AKT and STAT5 are critical downstream signaling molecules constitutively phosphorylated imatinib-resistant chronic myeloid leukemia (CML) and KIT D816V SM.4,5 This has been demonstrated in vitro using KIT D816V and BCR-ABL1+ imatinib-resistant cell lines, where inhibition of phosphorylation of these targets has shown their crucial role in cell proliferation.6,7 It has also been reported that STAT5 and AKT remained activated in neoplastic myeloid cells, even after inhibition of BCR-ABL1 by TKIs.8 Moreover, during disease progression, the levels of STAT5 mRNA and protein increase, and STAT5 production and activation is triggered not only by BCR-ABL1 or mutant KIT, but also by other pro-oncogenic pathways relevant to disease progression and resistance.9 Taken together, these observations strongly suggest that STAT5 and AKT are key drivers in drug-resistant CML and SM, and thus represent potential therapeutic targets. Small molecules targeting STAT5 or AKT may indeed be effective in these malignancies, especially in TKIs-resistant patients. However, inhibitors available today, such as pimozide or BP-1-108 for STAT5,10,11 or perifosine for AKT,12 are neither specific nor potent enough to be applicable in clinical practice. Therefore, it seems important to develop compounds that specifically and potently target STAT5 and AKT.
Mast cells and mastocytosis
KIT and cytokine signaling in normal mast cells
MCs originate from bone marrow (BM)-derived hematopoietic stem cells (HSCs) that enter the peripheral tissues via the bloodstream and undergo maturation under the influence of stem cell factor (SCF), a major cytokine-ligand of KIT (CD117).13 KIT is a transmembrane receptor with intrinsic tyrosine kinase (TK) activity.14 SCF-binding to KIT leads to dimerization and activation of the receptor.15 During KIT phosphorylation on specific tyrosines, the resulting phosphotyrosine (PT) residues become docking sites for signal transduction molecules. Activated KIT also catalyzes the phosphorylation of substrate proteins and triggers multiple signal transduction pathways.14 Once activated, KIT recruits the phosphatidylinositol 3-kinase (PI3K), which in turn activates AKT.16 Activated KIT also recruits the RAS/RAF and JAK/STAT signaling pathways.17,18 These pathways are involved in survival, proliferation, migration and differentiation of MCs.19
Pathogenesis of mastocytosis
Mastocytosis is a term collectively used for a heterogeneous group of disorders defined by expansion of MCs in one or more organs.20 Clinical symptoms result from MC-derived mediators, and from the destructive infiltration of neoplastic MCs in various organs.21 Mastocytosis can affect children and adults.21 However, mastocytosis developing in childhood is usually restricted to the skin and may resolve during or shortly before puberty, whereas in adult-onset mastocytosis, the disease is usually chronic, and defined by systemic involvement, with or without skin lesions.22
The World Health Organization (WHO) classification describes several different categories of mastocytosis, including cutaneous and systemic variants (SM) (Table 1).23 SM can further be divided into indolent SM (ISM), aggressive SM (ASM), MC leukemia (MCL), and SM with associated hematologic non-MC-lineage disease (SM-AHNMD).21 In the latter case, the AHNMD is frequently a myeloid neoplasm, such as an acute myeloid leukemia (AML) or a CML, a myelodysplastic syndrome (MDS) or a chronic myelomonocytic leukemia (CMML), whereas association of SM with a myeloma or a non-Hodgkin’s lymphoma is a rare event (Table 1).21 The KIT D816V mutation is found in up to 90% of all patients with SM (Figure 1).24 By contrast, pediatric patients usually harbor KIT mutations in other exons (Figure 1).25 KIT with the D816V mutation is constitutively activated, leading to the recruitment of major pro-oncogenic signaling cascades, such as the RAS/RAF-, STAT-, or PI3K-AKT-signaling pathways (Figure 2).19
Treatment of systemic mastocytosis
Treatment of non-advanced mastocytosis is based on pharmacological agents targeting symptoms caused by MC mediators.22 The most frequently used drugs are H1 and H2 antihistamines and glucocorticoids.22 Epinephrine is reserved for life-threatening episodes of anaphylaxis. In advanced SM, additional drugs are required to control MC expansion. However, so far, no standard anti-neoplastic therapies for patients with ASM, SM-AHNMD or MCL have been developed. Interferon alpha (IFN-α) showed variable efficacy, but also side effects, limiting its use.26 Cladribine (2-CdA) induces major clinical responses in a smaller group of patients with ASM.27 Both IFN-α and 2CdA can also induce cytopenia and immunosuppression.26 The same also holds true for other conventional anti-neoplastic drugs, like hydroxyurea, or chemotherapy, used to treat patients with ASM. All these agents also have a certain mutagenic potential. More recently, approaches to treat ASM and MCL have focused on KIT and KIT TKIs, because of the ubiquity of KIT mutations detected in these patients.28 Imatinib is usually not indicated, because the KIT D816V mutation confers resistance.2 Nevertheless, imatinib can reduce the MC burden and symptoms in SM patients exhibiting KIT mutations in other regions of the receptor.21 Other TKIs, such as dasatinib or PKC412 (midostaurin), are capable of inhibiting the KIT D816V activity in vitro.29 However, with the exception of PKC412, these drugs have very low efficacy in ASM.26 Therefore, alternative drugs and approaches using combinations of targeted drugs have been proposed for the treatment of patients with ASM and MCL.30
Chronic myelogenous leukemia
Chronic myelogenous leukemia (CML) is a myeloid neoplasm characterized by the presence of the BCR-ABL1 oncoprotein and the expansion and prominent granulocytic differentiation of neoplastic myeloid cells.31 A low rate of apoptosis is considered to lead to accumulation of neoplastic myeloid cells over time in these patients. As a result, CML patients usually present with marked leukocytosis, thrombocytosis, and often also anemia.32 The natural course of CML can be divided into 3 distinct phases: 1) a chronic phase (CP); followed by 2) an accelerated phase (AP); and eventually 3) the blast phase (BP) of CML. If untreated, CP inevitably evolves to AP and finally to BP, which resembles an acute leukemia.
Pathogenesis of CML
Chronic myelogenous leukemia is defined by the presence of a fusion gene acquired in an early hematopoietic progenitor, the BCR-ABL1 oncogene, which leads to the synthesis of the fusion protein BCR/ABL1 in leukemic cells (Figure 3).
The c-ABL gene has 11 exons located on chromosome 9q34, and it encodes a weak TK of 140 kDa.33 It displays an alternative exon I (a or b), with the exon Ib located upstream of the other exons (Ia and 2-11).33 The translocation breakpoint occurs between exon Ib and Ia in approximately 90% of the cases, resulting in a fully functional CABL coding sequence (exons Ia and 2-11) to be recombined to the BCR gene.34 The leukemogenic effect of BCR-ABL1 is mediated through activation of several downstream signaling pathways, including the RAS/RAF-, PI3K/AKT- and STAT-signaling pathway (Figure 3).4,35,36
Treatment of chronic myelogenous leukemia
The first clinically used drugs in CML that showed a survival benefit were busulfan in 1953,37 and hydroxyurea from 1972.38 Cytogenetic responses were first seen in patients treated with IFN-α.39 Despite these advances, however, many patients progressed to AP and BP. A milestone in the history of the treatment of CML remains allogenic hematopoietic stem cell transplantation (SCT), which was first introduced in the mid-1970s.40
In the early 2000s, imatinib, which binds the ATP-binding site of the chimeric BCR-ABL1 TK in a competitive manner, was introduced in clinical trials. It was soon proven to improve the survival rates in patients with CML. Since then, the drug is considered first-line standard therapy in CP CML.41 During the last ten years, two others TKIs have been approved for the treatment of CML. Dasatinib binds BCR-ABL1 as well as other major oncogenic kinases, such as SFK (Src-Family Kinases), in both their active and inactive states.42 This rather specific drug effect leads to broader inhibition of ABL independent of the protein conformation, making dasatinib more potent in advanced CML. Nilotinib (AMN107) exhibits a higher binding affinity and selectivity than imatinib. Both nilotinib and dasatinib have been approved for the treatment of imatinib resistant and newly diagnosed patients.43 More recently, bosutinib (SKI-606), which is more specific for BCR/ABL1 than imatinib or nilotinib, received approval as second-line drug for the treatment of imatinib-resistant Ph CML.44 Finally, the third-generation TKI ponatinib (AP24534) has shown strong anti-leukemia activity in Ph CML patients, including those with the highly resistant BCR-ABL1 T315I mutation.45 It has entered clinical trials for the treatment of T315I patients and other drug-resistant patients, but early analysis of interim data evidenced a high occurrence of arterial thrombotic events, which led to the recent discontinuation of these trials.
Tyrosine kinase inhibitors have greatly improved survival rates and remission in CML, allowing higher major molecular response rates at five years, as reported, for instance, in the International Randomized Study of Interferon and STI571 (IRIS) trial.46 However, approximately 20–30% of patients with CML develop either primary or secondary resistance to imatinib.3 Several different mechanisms underlie TKI resistance in CML, such as increased BCR-ABL1 expression, overexpression of drug-efflux proteins, secondary mutations in BCR-ABL1 that reduce drug affinity or drug effects, or upregulation and activation of downstream signaling molecules, including PI3K/AKT and STAT5 (Figure 3).9,48 However, mutations in the kinase domain of BCR-ABL1 are thought to be a primary cause of resistance in patients with CML, seen in up to 40–60% of cases of secondary resistance.3 These mutations can be detected usually in separate subclones, or rarely as compound mutations in the same clones. The most commonly detected mutation is T315I (20% of the retrieved mutations), which is resistant to almost all currently approved TKIs.49
Another challenging point in CML is the eradication of leukemic stem cells (LSC) which is a prerequisite for the development of curative therapies.50,51 There is growing evidence that TKIs fail to eliminate all primitive CML LSC in most patients.51 A better understanding of the biology and target expression profiles, as well as of the relationship between BCR-ABL1+ LSC and their specific microenvironment in the bone marrow (niche), has paved the way for the development of new treatment approaches.51,52 These strategies involve drug combinations, such as the pharmacological silencing of BCR-ABL1 with simultaneous inhibition of crucial signal transduction pathways,53 which may lead to the elimination, or at least suppression, of all CML LSC subsets.
The phosphoinositide 3-kinase (PI3K)-AKT pathway
Phosphoinositide 3-kinase (PI3Ks) represent a family of cytosolic, intracellular signaling proteins involved in the regulation of several cellular functions including proliferation and differentiation, survival, and malignant transformation.54 The primary enzymatic activity of these kinases is the phosphorylation of the 3-OH of inositol head groups of phosphoinositide (PI) lipids.55 PI3Ks can be divided into three main classes (I–III). These classes are based on their in vitro substrate specificity, structure, and probable mode of regulation. There are four isoforms of the catalytic subunit of class I PI3Ks: p110α, p110β, p110γ and p110δ.56 Whereas α and β isoforms are expressed ubiquitously, γ and δ isoforms are expressed mainly in lymphocytes. Interestingly, increased expression of p110γ is seen in CML and might account for resistance to treatment.57
AKT, also known as protein kinase B (PKB), is the major signaling arm of PI3K. In mammals, AKT is present in three different isoforms, AKT1 (or PKBα), AKT2 (PKBβ), and AKT3 (PKBγ). These isoforms are products of distinct genes, but are highly related and exhibit greater than 80% homology. Each isoform possesses an N-terminal pleckstrin homology (PH) domain, followed by the kinase domain (KD), which shows a high degree of similarity to those found in PKA and PKC (Figure 4A).58 Also present in this region is a threonine residue (T308 in PKBα/AKT1) whose phosphorylation is necessary for activation of AKT. Following the KD is a hydrophobic C-terminal tail containing a second regulatory phosphorylation site (S473 in PKBα/AKT1).
Following activation of receptor tyrosine kinases (RTKs), or of other cell surface receptors, the p85 adaptor subunit of PI3K associates with the receptor, leading to the activation of the p110 catalytic subunit. Activated p110 phosphorylates phosphatidylinositol 4-phosphate (PI4P), phosphatidylinositol 5-phosphate (PI5P) or phosphatidylinositol-4,5-bisphosphate (PIP2), generating thus the second messenger, phosphatidylinositol-3,4,5-triphosphate (PIP3) at the inner side of the plasma membrane (Figure 5). The interaction of the AKT PH domain with PIP3 induces conformational changes in AKT, resulting in the exposure of its two main phosphorylation sites at T308 and S473. Phosphorylation of these two sites by the S/TK3′-phosphoinositide-dependent kinases 1 and 2 (PDK1 and PDK2), which are also recruited by PIP3, is required for maximal activation of AKT. Phosphorylated AKT mediates the activation of various targets like Foxo, NF-κB and CREB transcription factors, the proapoptotic protein BAD, and cyclin D regulation through activating transcription factors downstream of mTOR/FRAP signaling. Overall, this results in anti-apoptotic effects, cell growth and proliferation (Figure 5).59 Through these interactions, AKT may contribute to malignant cell growth and disease evolution in BCR-ABL1+ CML and KIT D816V SM.60,61
Signals from PI3K are mainly antagonized by the 3′-inositol phosphatase and tensin homolog (PTEN) (Figure 5). PTEN is a tumor suppressor gene phosphatase that negatively regulates signaling through the PI3K pathway, inhibiting proliferation and survival.62 Interestingly, using an animal model in which PTEN can be deleted in an inducible way, Furumoto et al. have recently shown that this deletion caused MCs hyperplasia in various organs, which was associated with increased phosphorylation of STAT5 and AKT.63 Moreover, in another study, Peng et al. have reported that PTEN is down-regulated by BCR-ABL1 in an in vivo model of BCR-ABL1-induced CML and that overexpression of PTEN delayed the development of CML and prolonged survival of leukemia.64 In the same study, it was shown that PTEN suppressed leukemia stem cells and induced cell-cycle arrest of leukemia cells.64
AKT and mastocytosis
Recently, AKT activation has been identified as a key signaling molecule involved in KIT-dependent growth of neoplastic MCs harboring oncogenic KIT mutants.61 AKT was found constitutively phosphorylated in neoplastic MCs isolated from patients suffering from KIT D816V SM and in the HMC-1 cell line, a human KIT D816V leukemia MC line, raising the hypothesis that AKT activation plays a critical role in the pathogenesis of mastocytosis.61 In line with this hypothesis, abrogation of AKT activity is associated with growth inhibition of neoplastic MCs expressing the oncogenic KIT D816V mutant.61
AKT and CML
Downstream signaling cascades in BCR-ABL1-transformed cells include the STAT-, RAS/RAF- and the PI3K/AKT/mTOR pathways, all of which affect cell viability, cell-cycle progression and leukemogenesis.65 Activation of PI3K has emerged as one essential signaling step in ABL-mediated leukemogenesis. In line with this assumption, PI3K enzyme activity can be detected in BCR-ABL1 immunoprecipitates, which led to the initial assumption that activation of PI3K occurred mainly from a direct association of ABL with PI3K.60
The essential role of AKT in BCR-ABL1-mediated leuke-mogenesis was established by experiments demonstrating that the kinase-deficient dominant-negative Akt K179M mutant inhibits BCR-ABL1 induced transformation of BM cells in vitro and suppressed leukemia development in mice.36 The residual leukemogenic potential of wild-type (wt) BCR-ABL1 in the presence of the dominant-negative Akt mutant is most likely due to Akt-independent mechanisms of transformation, although one cannot exclude incomplete suppression of Akt activation in cells co-expressing wt BCR-ABL1 and the Akt K179M mutant.36 Consistent with the important role of AKT in BCR-ABL1-driven leukemogenesis, the constitutively active Akt E40K mutant rescued the defective transformation mediated by BCR-ABL1 SH2 mutants (delta SH2 and R1053L) in vitro.36 The importance of Akt as a signal transducer from the SH2 domain of BCR-ABL1, established by in vitro experiments, was confirmed in vivo using retrovirally infected BM cells injected into SCID mice.36
Compared with wt BCR-ABL1, cells expressing ΔSH2 BCR-ABL1 have markedly decreased leukemic potential as demonstrated by decreased tumor burden, only occasional involvement of non-hematopoietic organs, and diminished frequency of blastic transformation.36 Co-expression of the constitutively active Akt E40K restored the leuke-mogenic properties of ΔSH2 BCR-ABL1 in vivo.36 In addition, Chu et al. have demonstrated that, in CD34 cells from CML patients and human CD34 cells ectopically expressing the BCR-ABL1 gene, cytoplasmic p27 levels were increased, allowing increased cell cycling and expansion in culture.66 Interestingly, cytoplasmic relocation of p27 in CML progenitors was related to signaling through BCR-ABL1 Y177, activation of AKT kinase and phosphorylation of p27 on Thr-157 (T157).66 These observations underline the importance of AKT-mediated p27 phosphorylation in altered p27 localization and enhanced proliferation and expansion of primary CML progenitors.
On the other hand, all the BCR-ABL1 mutants capable of activating PI3K can also activate AKT, as demonstrated for the T315I mutation in the KBM-5 cell line.67 Thus, AKT appears to be the primary target of PI3K in the signaling pathway activated from the SH2 domain of BCR-ABL1, and is required for the BCR-ABL1-mediated leukemogenic transformation of hematopoietic cells.36
AKT inhibitors
AKT is viewed as an attractive target for cancer therapy and inhibition of AKT by targeted drugs is currently being evaluated in pre-clinical and clinical studies. AKT inhibitors include MK-2206, Tricibine (API-2), GSK690963, GSK2141795, KP372-1, Perifosine, Enzasturin (LY317615), PBI-05204, Erucylphosphocholine (ErPC), Erucylphosphohomocholine (ErPC3) and RX-0201.68
GSK2141795 is an AKT inhibitor with activity against various neoplastic cells, including blood and solid cancers. The drug has also been investigated in clinical trials with some success.68 KP372-1 induces mitochondrial dysfunction and apoptosis in AML cells at concentrations ranging between 0.5 μM and 1.0 μM, but did not induce apoptosis in normal hematopoietic progenitor cells below 1.0 μM.69
Effects of perifosine have been evaluated in different tumor types, resulting in an IC50 between 8 and 20 μM after 24 h on T-acute lymphoblastic leukemia cells,70 and between 1.25 and 6 μM (after 72 h) in human endometrial cancer cell lines.71
MK-2206, an allosteric AKT inhibitor, decreases the auto-phosphorylation of both AKT T308 and AKT S473. In addition, MK-2206 decreases T-acute lymphocytic leukemia cell viability by arresting the cells in the G0/G1 phase of the cell cycle, and by inducing apoptosis with IC50 values ranging between 1.7 and 5.1 μM.72 It has also been shown that inhibition of PI3K/AKT signaling by MK-2206 affects the growth of both JAK2 V617F- or MPLW515L-expressing primary neoplastic cells and cell lines via reduced phosphorylation of AKT and inhibition of its downstream signaling molecules.73 In the same study, MK-2206 alleviated hepato-splenomegaly and reduced the megakaryocyte burden in the BM, liver and spleen of mice with MPL W515L-induced MPN.73 However, most of these effects were only observed at concentrations of MK-2206 above 1.0 μM.73 All in all, the currently available AKT inhibitors exhibit IC50 values at the micromolar range, suggesting that there is a need to develop inhibitors of AKT acting at the nanomolar range. In addition, the use of AKT inhibitors in combination may increase their effects on cell proliferation. For instance, it has been recently demonstrated in the human leukemic MC line (HMC-1.2) expressing mutant KIT that treatment with STAT5-shRNA and LY294002 (PI3K inhibitor) resulted in an 80% inhibition of proliferation, which was superior to that induced by either STAT5-shRNA alone (60%) or Ly294002 treatment alone (55%).74 Moreover, the effects of STAT5 and PI3K/AKT inhibition on cell cycle are additive.74 Thus, the simultaneous targeting of PI3K/AKT and STAT5 signaling pathways may even better inhibit malignant cell proliferation in CML and SM.
JAK-STAT signaling
Seven mammalian STAT proteins have been identified, namely STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6.75 All STAT proteins share the ability to transmit a cellular signal from the cell membrane to the DNA, by steering transcriptional regulation of important genes relevant for normal or neoplastic cell growth or cell survival. STAT transcription factors are activated by various ligands and act together with cell type-specific co-factors or co-repressors which may explain in part their cell-specificity.76 Likewise, essential tyrosine and serine/threonine phosphorylation sites related to the transforming function of STATs have been mapped (Figure 4B).77
Although non-phosphorylated STAT5 (non-pSTAT5) may epigenetically suppress tumor growth by promoting heterochromatin formation, acting thus as a “tumor-suppressor”,78 the major role of STAT5 (like other STATs) is to promote the transcription of different genes. To fulfill this role, STAT5: i) undergoes an activation consisting in a tyrosine-phosphorylation step; ii) dimerizes through reciprocal interaction mediated by the phosphor-tyrosyl residue and the SH2 domain of the STAT monomers; iii) is internalized into the cell nucleus via associating with importins; iv) binds a specific DNA sequence; and v) activates the transcription through recruitment of protein partners. In addition, heterochromatin protein 1 (HP1), a conserved chromatin binding protein involved in heterochromatin assembly and gene silencing, and acting as a tumor suppressor in leukemogenesis, is inhibited by pSTAT5 protein, leading to enhanced cell proliferation activity (Figure 2).79
Several STAT activators have been described of which the canonical JAK-STAT pathway is best known (Figure 6).80 Thus, activating mutations in JAK2 may lead to STAT5 phosphorylation. Notably, the JAK2 V617F mutation is present in over 95% of patients with polycythemia vera (PV), and in approximately 50% of patients with primary myelofibrosis (PMF) or essential thrombocythemia (ET).81 Interestingly, this mutation induces increased expression of Stat5 in mice.82 There are differences in the recruitment of different STATs between JAK2 V617F MPNs, but both mice and humans with PV have a very significant activation of STAT5.83 Furthermore, in mice, it has been shown that activation of STAT5 was essential to the initiation and maintenance of PV after introduction of the JAK2 V617F mutant.82
Besides, the implications of the TK receptors and transforming intracellular TKs as STATs activators have been extensively reported.84,85 Among the STAT proteins, particularly activated STAT5a/b proteins (Figure 6) are thought to be of importance in MPNs, including SM and CML.5,7 However, SM or CML are often slowly progressing diseases, and it is thus not surprising that there are cytoplasmic pathways that maintain high pSTAT5 levels in the cytoplasm. Direct docking of pSTAT5 via the scaffold molecule GAB2 to the regulatory p85 subunit of PI3K is a prominent mechanism of cytoplasmic retention (Figures 2 and 6).86
In myeloproliferative neoplasms (MPN), important target genes of activated STAT5 are survival genes such as Mcl-1, Bcl-2, Bcl-XL, proliferation genes such as Cyclin D1 to D3, C-Myc, or cytokine receptor chains such as IL-4Ralpha chain, CD25 and CD123, both of which are expressed on CML LSC, the lymphocyte antigen Ly-6E,87 or negative regulators such as Cis or Socs1-3 (Figure 2).88
Thus, STAT5 is a potential major target in MPNs for which so far there are no specific and potent pharmacological inhibitors. However, Page et al. could show that the SF-1-088 salicylic acid-containing inhibitor binds the SH2 domain of STAT5, decreasing the binding of STAT5 to its phosphorylating partner, thus inducing a lower phosphorylation level and diminished transcription through it.11
STAT5 and mastocytosis
The involvement of STAT5 in growth and survival of normal and neoplastic MCs is well known.89 Consequently, as the uncontrolled cell growth is one feature of tumors, several teams studied the implication of STAT5 in neoplastic MC growth, survival and transformation, and some light was shed on its implication in tumor growth downstream of KIT D816V.
A first study published by Gouilleux and colleagues showed that pSTAT5 is found in the cytoplasm of MCs from patients with SM.90 The study also further emphasized the molecular interactions between STAT5 and PI3K via the GAB2 scaffold protein interaction bridging p85 and pSTAT5 interaction (Figures 2 and 6). Moreover, knockdown of STAT5 (or AKT) led to cell growth inhibition.61 Thus, STAT5 function in MC neoplasms is linked to PI3K-AKT signaling and intrinsic cytokine signaling by IL-3/-4 will further boost their synergism.61,91
A second study has further explored this non-canonical STAT pathway, and has shown that neoplastic MCs express cytoplasmic and nuclear pSTAT5.91 Furthermore, the same team showed that KIT D816V promotes direct STAT5-activation, and that it contributes to growth of neoplastic MCs.91 Finally, despite STAT1/3 activation, the expression levels of STAT5 seem to be critical for transcriptional regulation in HMC-1 and P815 MC lines, and for neoplastic cell growth and survival.7
Altogether, these results strongly suggest that STAT5 is one major cellular effector in mastocytosis by controlling the mutant KIT-mediated aberrant growth signaling. However, the pharmacological inhibition of STAT5 remains challenging, and new STAT5 inhibitors active at pharmacological doses on both indolent and aggressive forms of SM are still needed.
STAT5 and CML
In CML, BCR-ABL1 was shown to directly phosphorylate STAT5 (Y694/Y699; Figure 6) that then dimerizes in a parallel fashion to allow rapid nuclear translocation and oligomerization on chromatin to regulate gene transcription, which subsequently promotes myeloid cell survival and growth.92 However, pSTAT5 appears mostly retained in the cytoplasm in BCR–ABL1-positive cells, this retention being linked to binding to GAB2 or to Src family kinases (Figure 6).90,93 Whatever mechanism underlies cytoplasmic retention of pSTAT5, more recent studies on STAT5 in CML cells have proven that this molecule is necessary for both transformation and cell cycle progression.4 In addition, STAT5a and STAT5b suppression by siRNA transfection mediated CML cell apoptosis,94 and STAT5a suppression induced a higher sensitivity of imatinib-sensitive K562 cells to imatinib, and sensitized imatinib-resistant K562 cells to imatinib.95 Interestingly, high levels of pSTAT5 are correlated to TKI resistance in vitro and in vivo, and to CML progression.9 Furthermore, a recent publication described a highly significant correlation between the level of STAT5a mRNA and the occurrence of BCR-ABL1 mutations in a cohort of 50 CML patients, possibly mediated by the enforced production of reactive oxygen intermediates.96 Moreover, using a mouse model with a conditional null mutation in the Stat5a/b gene locus, Waltz et al. have determined the requirement for STAT5 in MPNs induced by BCR-ABL1 in a retroviral transplantation model of CML.82 They provided evidence that the loss of one Stat5a/b allele results in a decrease in BCR-ABL1-induced CML-like MPN and the appearance of B-cell acute lymphoblastic leukemia, whereas complete deletion of Stat5a/b prevented the development of leukemia in primary recipients.82 However, the specific contributions of the two related genes, Stat5a and Stat5b, to growth and survival of CML cells were not clarified in this report. In a recent study using an RNAi-based strategy, Casetti et al. showed that STAT5a/STAT5b double-knockdown triggers CML cell apoptosis and suppresses the long-term clonogenic potential of normal and CML progenitor cells.97 In addition, the same authors reported that STAT5a attenuation alone was ineffective at impairing growth of normal and CML CD34 cells isolated at diagnosis. In contrast, STAT5a attenuation was reported to be sufficient to enhance basal oxidative stress and DNA damage of normal CD34 and CML cells and to inhibit growth of CML CD34 cells from patients with acquired resistance to imatinib.97 These data are in line with those reported by Rousselot et al. who have demonstrated that targeting expression of STAT5a and b using pioglitazone, a peroxi-some proliferator-activated receptor (PPARs)-agonist, resulted in an improvement in molecular response in patients with CP CML treated with imatinib.98
All in all, the above-mentioned reports provide solid evidence that targeting STAT5 may be an attractive therapeutic approach in CML. A complete loss of STAT5 might not be beneficial because of the important biological roles this molecule plays in physiological tissues. Rather, interfering with the extra production or activation of (too much) pSTAT5 in neoplastic cells might be the right way to go. This should be done by direct targeting of the molecule or by targeting distinct STAT5-controlled survival proteins such as BCL-2/BCL-XL or to interfere with cytoplasmic control via STAT5 on AKT/mTOR signaling.99 Finally, targeting STAT5 or related signals activated by this molecule could not only overcome drug resistance as well as disease progression, but also might open opportunities to eradicate the most primitive and TKI-resistant CML LSC populations.
STAT5 inhibitors
Cell lines or CD34 cells from CML patients treated with pimozide revealed decreased pSTAT5 levels.10 Furthermore, pimozide showed major effects on cell survival and induced cell cycle arrest and apoptosis in CML cells. In addition, pimozide showed synergistic anti-leukemic effects together with imatinib, presumably through decreased STAT5 phosphorylation.10 Finally, pimozide also exhibits inhibitory effect on CD34 CML cell growth, whereas non-CML cells are only slightly affected,10 suggesting that a STAT5-targeted therapy may act rather specifically on leukemic cells over-expressing activated STAT5. However, the concentrations of pimozide required to elicit anti-leukemic effects were rather high.10
Two other classes of STAT5 inhibitors have also been reported recently.11,100 These drugs share common features in their inhibition-profiles: suppression of STAT5 activation and induction of apoptosis. Although no results on imatinib-resistant or primary CML cells are available for acid-salicylic-containing molecules,11 indirubin derivatives have shown anti-neoplastic activity in imatinib-sensitive, T315I-positive-imatinib-resistant and CD34 primary CML cells.100 However, these effects were obtained at an IC50 of around 5 μM, which might be difficult to achieve during administration to humans. Further side group modifications or screening of new small molecular weight compound libraries could improve selectivity and efficacy in a next generation of STAT5 isoform inhibitors.
Conclusions and future perspectives
Multiple lines of evidence suggest that the STAT and the PI3-K/AKT pathways are crucial for disease evolution and progression in CML and advanced SM. Indeed, in both types of neoplasm, these two effector molecules are activated and may act together to trigger proliferation and survival as well as drug resistance in neoplastic (stem) cells. Moreover, chemical inhibition or gene silencing experiments have shown that both AKT and STAT5 are required for oncogenesis and disease evolution, and that co-inhibition of both STAT5 and AKT cells may elicit synergistic effects on leukemic cell growth and proliferation in these two malignancies. As a result, STAT5 and AKT are currently regarded to be the most attractive potential targets of therapy in advanced CML and SM, and there is hope that their simultaneous pharmacological inhibition could lead to improved anti-neoplastic effects. Whether such an approach will indeed overcome drug resistance in neoplastic (stem) cells in these malignancies remains to be determined in forthcoming pre-clinical and clinical trials. In addition, besides CML and SM, there is substantial interest in targeting PI3-K/AKT and STAT5 molecules in other MPNs such as PV, ET or PMF whereas JAK2 inhibitors have failed to provide substantial therapeutic effects, being unable to lead to complete remission of these diseases.
Footnotes
- Funding This work has been supported in part by a grant from Fondation de France as well as by the Austrian Science Fund (FWF), grants SFB-F2807, SFB-F4704 and SFB-F4707.
- Authorship and Disclosures Information on authorship, contributions, and financial & other disclosures was provided by the authors and is available with the online version of this article at www.haematologica.org.
- Received October 8, 2013.
- Accepted December 20, 2013.
References
- Ashman LK, Griffith R. Therapeutic targeting of c-KIT in cancer. Expert Opin Investig Drugs. 2013; 22(1):103-15. PubMedhttps://doi.org/10.1517/13543784.2013.740010Google Scholar
- Akin C, Brockow K, D’Ambrosio C, Kirshenbaum AS, Ma Y, Longley BJ. Effects of tyrosine kinase inhibitor STI571 on human mast cells bearing wild-type or mutated c-kit. Exp Hematol. 2003; 31(8):686-92. PubMedhttps://doi.org/10.1016/S0301-472X(03)00112-7Google Scholar
- Cortes J, Goldman JM, Hughes T. Current issues in chronic myeloid leukemia: monitoring, resistance, and functional cure. J Natl Compr Canc Netw. 2012; 10(Suppl 3):S1-S13. Google Scholar
- Hoelbl A, Schuster C, Kovacic B, Zhu B, Wickre M, Hoelzl MA. Stat5 is indispensable for the maintenance of bcr/abl-positive leukaemia. EMBO Mol Med. 2010; 2(3):98-110. PubMedhttps://doi.org/10.1002/emmm.201000062Google Scholar
- Hantschel O, Warsch W, Eckelhart E, Kaupe I, Grebien F, Wagner KU. BCR-ABL uncouples canonical JAK2-STAT5 signaling in chronic myeloid leukemia. Nat Chem Biol. 2012; 8(3):285-93. PubMedhttps://doi.org/10.1038/nchembio.775Google Scholar
- Sillaber C, Gesbert F, Frank DA, Sattler M, Griffin JD. STAT5 activation contributes to growth and viability in Bcr/Abl-transformed cells. Blood. 2000; 95(6):2118-25. PubMedGoogle Scholar
- Chaix A, Lopez S, Voisset E, Gros L, Dubreuil P, De Sepulveda P. Mechanisms of STAT protein activation by oncogenic KIT mutants in neoplastic mast cells. J Biol Chem. 2011; 286(8):5956-66. PubMedhttps://doi.org/10.1074/jbc.M110.182642Google Scholar
- Konig H, Holtz M, Modi H, Manley P, Holyoake TL, Forman SJ. Enhanced BCR-ABL kinase inhibition does not result in increased inhibition of downstream signaling pathways or increased growth suppression in CML progenitors. Leukemia. 2008; 22(4):748-55. PubMedhttps://doi.org/10.1038/sj.leu.2405086Google Scholar
- Warsch W, Kollmann K, Eckelhart E, Fajmann S, Cerny-Reiterer S, Holbl A. High STAT5 levels mediate imatinib resistance and indicate disease progression in chronic myeloid leukemia. Blood. 2011; 117(12):3409-20. PubMedhttps://doi.org/10.1182/blood-2009-10-248211Google Scholar
- Nelson EA, Walker SR, Weisberg E, Bar-Natan M, Barrett R, Gashin LB. The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood. 2011; 117(12):3421-9. PubMedhttps://doi.org/10.1182/blood-2009-11-255232Google Scholar
- Page BD, Khoury H, Laister RC, Fletcher S, Vellozo M, Manzoli A. Small molecule STAT5-SH2 domain inhibitors exhibit potent antileukemia activity. J Med Chem. 2012; 55(3):1047-55. PubMedhttps://doi.org/10.1021/jm200720nGoogle Scholar
- Tong Y, Liu YY, You LS, Qian WB. Perifosine induces protective autophagy and upregulation of ATG5 in human chronic myelogenous leukemia cells in vitro. Acta Pharmacol Sin. 2012; 33(4):542-50. PubMedhttps://doi.org/10.1038/aps.2011.192Google Scholar
- Galli SJ, Tsai M, Wershil BK, Tam SY, Costa JJ. Regulation of mouse and human mast cell development, survival and function by stem cell factor, the ligand for the c-kit receptor. Int Arch Allergy Immunol. 1995; 107(1–3):51-3. PubMedhttps://doi.org/10.1159/000236928Google Scholar
- Roskoski R. Signaling by Kit protein-tyro-sine kinase--the stem cell factor receptor. Biochem Biophys Res Commun. 2005; 337(1):1-13. PubMedhttps://doi.org/10.1016/j.bbrc.2005.08.055Google Scholar
- Blume-Jensen P, Claesson-Welsh L, Siegbahn A, Zsebo KM, Westermark B, Heldin CH. Activation of the human c-kit product by lig-and-induced dimerization mediates circular actin reorganization and chemotaxis. EMBO J. 1991; 10(13):4121-8. PubMedGoogle Scholar
- Blume-Jensen P, Janknecht R, Hunter T. The kit receptor promotes cell survival via activation of PI 3-kinase and subsequent Akt-mediated phosphorylation of Bad on Ser136. Curr Biol. 1998; 8(13):779-82. PubMedhttps://doi.org/10.1016/S0960-9822(98)70302-1Google Scholar
- Weiler SR, Mou S, DeBerry CS, Keller JR, Ruscetti FW, Ferris DK. JAK2 is associated with the c-kit proto-oncogene product and is phosphorylated in response to stem cell factor. Blood. 1996; 87(9):3688-93. PubMedGoogle Scholar
- Kitamura Y, Hirotab S. Kit as a human oncogenic tyrosine kinase. Cell Mol Life Sci. 2004; 61(23):2924-31. PubMedhttps://doi.org/10.1007/s00018-004-4273-yGoogle Scholar
- Roskoski R. Structure and regulation of Kit protein-tyrosine kinase--the stem cell factor receptor. Biochem Biophys Res Commun. 2005; 338(3):1307-15. PubMedhttps://doi.org/10.1016/j.bbrc.2005.09.150Google Scholar
- Valent P, Sperr WR, Schwartz LB, Horny HP. Diagnosis and classification of mast cell proliferative disorders: delineation from immunologic diseases and non-mast cell hematopoietic neoplasms. J Allergy Clin Immunol. 2004; 114(1):3-11. PubMedhttps://doi.org/10.1016/j.jaci.2004.02.045Google Scholar
- Arock M, Valent P. Pathogenesis, classification and treatment of mastocytosis: state of the art in 2010 and future perspectives. Expert Rev Hematol. 2010; 3(4):497-516. PubMedhttps://doi.org/10.1586/ehm.10.42Google Scholar
- Brockow K, Metcalfe DD. Mastocytosis. Chem Immunol Allergy. 2010; 95:110-24. PubMedhttps://doi.org/10.1159/000315946Google Scholar
- Sanchez-Munoz L, Alvarez-Twose I, Garcia-Montero AC, Teodosio C, Jara-Acevedo M, Pedreira CE. Evaluation of the WHO criteria for the classification of patients with mastocytosis. Mod Pathol. 2011; 24(9):1157-68. PubMedhttps://doi.org/10.1038/modpathol.2011.84Google Scholar
- Valent P. Systemic mastocytosis. Cancer Treat Res. 2008; 142:399-419. PubMedGoogle Scholar
- Bodemer C, Hermine O, Palmerini F, Yang Y, Grandpeix-Guyodo C, Leventhal PS. Pediatric mastocytosis is a clonal disease associated with D816V and other activating c-KIT mutations. J Invest Dermatol. 2010; 130(3):804-15. PubMedhttps://doi.org/10.1038/jid.2009.281Google Scholar
- Metcalfe DD. Mast cells and mastocytosis. Blood. 2008; 112(4):946-56. PubMedhttps://doi.org/10.1182/blood-2007-11-078097Google Scholar
- Tefferi A, Li CY, Butterfield JH, Hoagland HC. Treatment of systemic mast-cell disease with cladribine. N Engl J Med. 2001; 344(4):307-9. PubMedhttps://doi.org/10.1056/NEJM200101253440415Google Scholar
- Verstovsek S. Advanced systemic mastocytosis: the impact of KIT mutations in diagnosis, treatment, and progression. Eur J Haematol. 2013; 90(2):89-98. PubMedhttps://doi.org/10.1111/ejh.12043Google Scholar
- Gleixner KV, Mayerhofer M, Sonneck K, Gruze A, Samorapoompichit P, Baumgartner C. Synergistic growth-inhibitory effects of two tyrosine kinase inhibitors, dasatinib and PKC412, on neoplastic mast cells expressing the D816V-mutated oncogenic variant of KIT. Haematologica. 2007; 92(11):1451-9. PubMedhttps://doi.org/10.3324/haematol.11339Google Scholar
- Gleixner KV, Mayerhofer M, Cerny-Reiterer S, Hormann G, Rix U, Bennett KL. KIT-D816V-independent oncogenic signaling in neoplastic cells in systemic mastocytosis: role of Lyn and Btk activation and disruption by dasatinib and bosutinib. Blood. 2011; 118(7):1885-98. PubMedhttps://doi.org/10.1182/blood-2010-06-289959Google Scholar
- Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H. WHO Classification of tumours of Haematopoietic and Lymphoid Tissues. Lyon, France; 2008. Google Scholar
- Melo JV, Deininger MW. Biology of chronic myelogenous leukemia--signaling pathways of initiation and transformation. Hematol Oncol Clin North Am. 2004; 18(3):545-68. PubMedhttps://doi.org/10.1016/j.hoc.2004.03.008Google Scholar
- Hantschel O. Structure, regulation, signaling, and targeting of abl kinases in cancer. Genes Cancer. 2012; 3(5–6):436-46. PubMedhttps://doi.org/10.1177/1947601912458584Google Scholar
- Zion M, Ben-Yehuda D, Avraham A, Cohen O, Wetzler M, Melloul D. Progressive de novo DNA methylation at the bcr-abl locus in the course of chronic myelogenous leukemia. Proc Natl Acad Sci USA. 1994; 91(22):10722-6. PubMedhttps://doi.org/10.1073/pnas.91.22.10722Google Scholar
- Marais R, Light Y, Paterson HF, Marshall CJ. Ras recruits Raf-1 to the plasma membrane for activation by tyrosine phosphorylation. EMBO J. 1995; 14(13):3136-45. PubMedGoogle Scholar
- Skorski T, Bellacosa A, Nieborowska-Skorska M, Majewski M, Martinez R, Choi JK. Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway. EMBO J. 1997; 16(20):6151-61. PubMedhttps://doi.org/10.1093/emboj/16.20.6151Google Scholar
- Buggia I, Locatelli F, Regazzi MB, Zecca M. Busulfan. Ann Pharmacother. 1994; 28(9):1055-62. PubMedhttps://doi.org/10.1177/106002809402800911Google Scholar
- Rushing D, Goldman A, Gibbs G, Howe R, Kennedy BJ. Hydroxyurea versus busulfan in the treatment of chronic myelogenous leukemia. Am J Clin Oncol. 1982; 5(3):307-13. PubMedhttps://doi.org/10.1097/00000421-198206000-00013Google Scholar
- Talpaz M, Hehlmann R, Quintas-Cardama A, Mercer J, Cortes J. Re-emergence of inter-feron-alpha in the treatment of chronic myeloid leukemia. Leukemia. 2013; 27(4):803-12. PubMedhttps://doi.org/10.1038/leu.2012.313Google Scholar
- Andersson BS. Advances in CML: The role of allogeneic hematopoietic stem cell transplantation. Clin Adv Hematol Oncol. 2010; 8(11):746-8. PubMedGoogle Scholar
- Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2012 update on diagnosis, monitoring, and management. Am J Hematol. 2012; 87(11):1037-45. PubMedhttps://doi.org/10.1002/ajh.23282Google Scholar
- Lindauer M, Hochhaus A. Dasatinib. Recent Results Cancer Res. 2010; 184:83-102. PubMedhttps://doi.org/10.1007/978-3-642-01222-8_7Google Scholar
- Kantarjian H, Giles F, Wunderle L, Bhalla K, O’Brien S, Wassmann B. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med. 2006; 354(24):2542-51. PubMedhttps://doi.org/10.1056/NEJMoa055104Google Scholar
- Puttini M, Coluccia AM, Boschelli F, Cleris L, Marchesi E, Donella-Deana A. In vitro and in vivo activity of SKI-606, a novel Src-Abl inhibitor, against imatinib-resistant Bcr-Abl+ neoplastic cells. Cancer Res. 2006; 66(23):11314-22. PubMedhttps://doi.org/10.1158/0008-5472.CAN-06-1199Google Scholar
- Frankfurt O, Licht JD. Ponatinib--a step forward in overcoming resistance in chronic myeloid leukemia. Clin Cancer Res. 2013; 19(21):5828-34. PubMedhttps://doi.org/10.1158/1078-0432.CCR-13-0258Google Scholar
- Hughes TP, Hochhaus A, Branford S, Müller MC, Kaeda JS, Foroni L. Long-term prognostic significance of early molecular response to imatinib in newly diagnosed chronic myeloid leukemia: an analysis from the International Randomized Study of Interferon and STI571 (IRIS). Blood. 2010; 116(19):3758-65. PubMedhttps://doi.org/10.1182/blood-2010-03-273979Google Scholar
- Rajala HL, Eldfors S, Kuusanmaki H, van Adrichem AJ, Olson T, Lagstrom S. Discovery of somatic STAT5b mutations in large granular lymphocytic leukemia. Blood. 2013; 121(22):4541-50. PubMedhttps://doi.org/10.1182/blood-2012-12-474577Google Scholar
- Bixby D, Talpaz M. Mechanisms of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia and recent therapeutic strategies to overcome resistance. Hematology Am Soc Hematol Educ Program. 2009;461-76. Google Scholar
- Burke AC, Swords RT, Kelly K, Giles FJ. Current status of agents active against the T315I chronic myeloid leukemia phenotype. Expert Opin Emerg Drugs. 2011; 16(1):85-103. PubMedhttps://doi.org/10.1517/14728214.2011.531698Google Scholar
- Valent P. Emerging stem cell concepts for imatinib-resistant chronic myeloid leukaemia: implications for the biology, management, and therapy of the disease. Br J Haematol. 2008; 142(3):361-78. PubMedhttps://doi.org/10.1111/j.1365-2141.2008.07197.xGoogle Scholar
- Valent P, Bonnet D, De Maria R, Lapidot T, Copland M, Melo JV. Cancer stem cell definitions and terminology: the devil is in the details. Nat Rev Cancer. 2012; 12(11):767-75. PubMedhttps://doi.org/10.1038/nrc3368Google Scholar
- Gallipoli P, Abraham SA, Holyoake TL. Hurdles toward a cure for CML: the CML stem cell. Hematol Oncol Clin North Am. 2011; 25(5):951-66. PubMedhttps://doi.org/10.1016/j.hoc.2011.09.001Google Scholar
- O’Hare T, Zabriskie MS, Eiring AM, Deininger MW. Pushing the limits of targeted therapy in chronic myeloid leukaemia. Nat Rev Cancer. 2012; 12(8):513-26. PubMedhttps://doi.org/10.1038/nrc3317Google Scholar
- Martini M, Ciraolo E, Gulluni F, Hirsch E. Targeting PI3K in Cancer: Any Good News?. Front Oncol. 2013; 3:108. PubMedhttps://doi.org/10.3389/fonc.2013.00108Google Scholar
- Toker A, Cantley LC. Signalling through the lipid products of phosphoinositide-3-OH kinase. Nature. 1997; 387(6634):673-6. PubMedhttps://doi.org/10.1038/42648Google Scholar
- Vogt PK, Hart JR, Gymnopoulos M, Jiang H, Kang S, Bader AG. Phosphatidylinositol 3-kinase: the oncoprotein. Curr Top Microbiol Immunol. 2010; 347:79-104. PubMedhttps://doi.org/10.1007/82_2010_80Google Scholar
- Hickey FB, Cotter TG. BCR-ABL regulates phosphatidylinositol 3-kinase-p110gamma transcription and activation and is required for proliferation and drug resistance. J Biol Chem. 2006; 281(5):2441-50. PubMedhttps://doi.org/10.1074/jbc.M511173200Google Scholar
- Andjelkovic M, Jones PF, Grossniklaus U, Cron P, Schier AF, Dick M. Developmental regulation of expression and activity of multiple forms of the Drosophila RAC protein kinase. J Biol Chem. 1995; 270(8):4066-75. PubMedhttps://doi.org/10.1074/jbc.270.8.4066Google Scholar
- Nicholson KM, Anderson NG. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002; 14(5):381-95. PubMedhttps://doi.org/10.1016/S0898-6568(01)00271-6Google Scholar
- Kharas MG, Fruman DA. ABL oncogenes and phosphoinositide 3-kinase: mechanism of activation and downstream effectors. Cancer Res. 2005; 65(6):2047-53. PubMedhttps://doi.org/10.1158/0008-5472.CAN-04-3888Google Scholar
- Harir N, Boudot C, Friedbichler K, Sonneck K, Kondo R, Martin-Lanneree S. Oncogenic Kit controls neoplastic mast cell growth through a Stat5/PI3-kinase signaling cascade. Blood. 2008; 112(6):2463-73. PubMedhttps://doi.org/10.1182/blood-2007-09-115477Google Scholar
- Di Cristofano A, Pandolfi PP. The multiple roles of PTEN in tumor suppression. Cell. 2000; 100(4):387-90. PubMedhttps://doi.org/10.1016/S0092-8674(00)80674-1Google Scholar
- Furumoto Y, Charles N, Olivera A, Leung WH, Dillahunt S, Sargent JL. PTEN deficiency in mast cells causes a mastocyto-sis-like proliferative disease that heightens allergic responses and vascular permeability. Blood. 2011; 118(20):5466-75. PubMedhttps://doi.org/10.1182/blood-2010-09-309955Google Scholar
- Peng C, Chen Y, Yang Z, Zhang H, Osterby L, Rosmarin AG. PTEN is a tumor suppressor in CML stem cells and BCR-ABL-induced leukemias in mice. Blood. 2010; 115(3):626-35. PubMedhttps://doi.org/10.1182/blood-2009-06-228130Google Scholar
- Steelman LS, Abrams SL, Whelan J, Bertrand FE, Ludwig DE, Basecke J. Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to leukemia. Leukemia. 2008; 22(4):686-707. PubMedhttps://doi.org/10.1038/leu.2008.26Google Scholar
- Chu S, McDonald T, Bhatia R. Role of BCR-ABL-Y177-mediated p27kip1 phosphoryla-tion and cytoplasmic localization in enhanced proliferation of chronic myeloid leukemia progenitors. Leukemia. 2010; 24(4):779-87. PubMedhttps://doi.org/10.1038/leu.2010.24Google Scholar
- Lu Z, Jin Y, Chen C, Li J, Cao Q, Pan J. Pristimerin induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation by blocking NF-kappaB signaling and depleting Bcr-Abl. Mol Cancer. 2010; 9:112. PubMedhttps://doi.org/10.1186/1476-4598-9-112Google Scholar
- McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Franklin RA, Montalto G. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/ mTOR Cascade Inhibitors: How Mutations Can Result in Therapy Resistance and How to Overcome Resistance. Oncotarget. 2012; 3(10):1068-111. PubMedGoogle Scholar
- Zeng Z, Samudio IJ, Zhang W, Estrov Z, Pelicano H, Harris D. Simultaneous inhibition of PDK1/AKT and Fms-like tyro-sine kinase 3 signaling by a small-molecule KP372-1 induces mitochondrial dysfunction and apoptosis in acute myelogenous leukemia. Cancer Res. 2006; 66(7):3737-46. PubMedhttps://doi.org/10.1158/0008-5472.CAN-05-1278Google Scholar
- Chiarini F, Del Sole M, Mongiorgi S, Gaboardi GC, Cappellini A, Mantovani I. The novel Akt inhibitor, perifosine, induces caspase-dependent apoptosis and downregulates P-glycoprotein expression in multidrug-resistant human T-acute leukemia cells by a JNK-dependent mechanism. Leukemia. 2008; 22(6):1106-16. PubMedhttps://doi.org/10.1038/leu.2008.79Google Scholar
- Engel JB, Honig A, Schonhals T, Weidler C, Hausler S, Krockenberger M. Perifosine inhibits growth of human experimental endometrial cancers by blockade of AKT phosphorylation. Eur J Obstet Gynecol Reprod Biol. 2008; 141(1):64-9. PubMedhttps://doi.org/10.1016/j.ejogrb.2008.06.007Google Scholar
- Simioni C, Neri LM, Tabellini G, Ricci F, Bressanin D, Chiarini F. Cytotoxic activity of the novel Akt inhibitor, MK-2206, in T-cell acute lymphoblastic leukemia. Leukemia. 2012; 26(11):2336-42. PubMedhttps://doi.org/10.1038/leu.2012.136Google Scholar
- Khan I, Huang Z, Wen Q, Stankiewicz MJ, Gilles L, Goldenson B. AKT is a therapeutic target in myeloproliferative neoplasms. Leukemia. 2013; 27(9):1882-90. PubMedhttps://doi.org/10.1038/leu.2013.167Google Scholar
- Buet D, Gallais I, Lauret E, Denis N, Lombard B, Guillonneau F. Cotargeting signaling pathways driving survival and cell cycle circumvents resistance to Kit inhibitors in leukemia. Blood. 2012; 119(18):4228-41. PubMedhttps://doi.org/10.1182/blood-2011-07-368316Google Scholar
- Hirano T, Matsuda T, Nakajima K. Signal transduction through gp130 that is shared among the receptors for the interleukin 6 related cytokine subfamily. Stem Cells. 1994; 12(3):262-77. PubMedhttps://doi.org/10.1002/stem.5530120303Google Scholar
- Bromberg JF. Activation of STAT proteins and growth control. Bioessays. 2001; 23(2):161-9. PubMedhttps://doi.org/10.1002/1521-1878(200102)23:2<161::AID-BIES1023>3.0.CO;2-0Google Scholar
- Lim CP, Cao X. Structure, function, and regulation of STAT proteins. Mol Biosyst. 2006; 2(11):536-50. PubMedhttps://doi.org/10.1039/b606246fGoogle Scholar
- Hu X, Dutta P, Tsurumi A, Li J, Wang J, Land H. Unphosphorylated STAT5A stabilizes heterochromatin and suppresses tumor growth. Proc Natl Acad Sci USA. 2013; 110(25):10213-8. PubMedhttps://doi.org/10.1073/pnas.1221243110Google Scholar
- Shi S, Larson K, Guo D, Lim SJ, Dutta P, Yan SJ. Drosophila STAT is required for directly maintaining HP1 localization and heterochromatin stability. Nat Cell Biol. 2008; 10(4):489-96. PubMedhttps://doi.org/10.1038/ncb1713Google Scholar
- Lamb P, Tapley P, Rosen J. Biochemical approaches to discovering modulators of the JAK-STAT pathway. Drug Discovery Today. 2007; 3(3):122-30. Google Scholar
- Staerk J, Constantinescu SN. The JAK-STAT pathway and hematopoietic stem cells from the JAK2 V617F perspective. JAKSTAT. 2012; 1(3):184-90. PubMedGoogle Scholar
- Walz C, Ahmed W, Lazarides K, Betancur M, Patel N, Hennighausen L. Essential role for Stat5a/b in myeloproliferative neoplasms induced by BCR-ABL1 and JAK2(V617F) in mice. Blood. 2012; 119(15):3550-60. PubMedhttps://doi.org/10.1182/blood-2011-12-397554Google Scholar
- Risum M, Madelung A, Bondo H, Bzorek M, Kristensen MH, Stamp IM. The JAK2V617F allele burden and STAT3- and STAT5 phosphorylation in myeloproliferative neoplasms: early prefibrotic myelofibrosis compared with essential thrombocythemia, polycythemia vera and myelofibrosis. APMIS. 2011; 119(8):498-504. PubMedhttps://doi.org/10.1111/j.1600-0463.2011.02754.xGoogle Scholar
- Quesnelle KM, Boehm AL, Grandis JR. STAT-mediated EGFR signaling in cancer. J Cell Biochem. 2007; 102(2):311-9. PubMedhttps://doi.org/10.1002/jcb.21475Google Scholar
- Kim YR, Byun HS, Won M, Park KA, Kim JM, Choi BL. Modulatory role of phospholipase D in the activation of signal transducer and activator of transcription (STAT)-3 by thyroid oncogenic kinase RET/PTC. BMC Cancer. 2008; 8:144. PubMedhttps://doi.org/10.1186/1471-2407-8-144Google Scholar
- Nyga R, Pecquet C, Harir N, Gu H, Dhennin-Duthille I, Regnier A. Activated STAT5 proteins induce activation of the PI 3-kinase/Akt and Ras/MAPK pathways via the Gab2 scaffolding adapter. Biochem J. 2005; 390(Pt 1):359-66. PubMedhttps://doi.org/10.1042/BJ20041523Google Scholar
- Khan KD, Shuai K, Lindwall G, Maher SE, Darnell JE, Bothwell AL. Induction of the Ly-6A/E gene by interferon alpha/beta and gamma requires a DNA element to which a tyrosine-phosphorylated 91-kDa protein binds. Proc Natl Acad Sci USA. 1993; 90(14):6806-10. PubMedhttps://doi.org/10.1073/pnas.90.14.6806Google Scholar
- Ferbeyre G, Moriggl R. The role of Stat5 transcription factors as tumor suppressors or oncogenes. Biochim Biophys Acta. 2011; 1815(1):104-14. PubMedGoogle Scholar
- Shelburne CP, McCoy ME, Piekorz R, Sexl V, Roh KH, Jacobs-Helber SM. Stat5 expression is critical for mast cell development and survival. Blood. 2003; 102(4):1290-7. PubMedhttps://doi.org/10.1182/blood-2002-11-3490Google Scholar
- Harir N, Pecquet C, Kerenyi M, Sonneck K, Kovacic B, Nyga R. Constitutive activation of Stat5 promotes its cytoplasmic localization and association with PI3-kinase in myeloid leukemias. Blood. 2007; 109(4):1678-86. PubMedhttps://doi.org/10.1182/blood-2006-01-029918Google Scholar
- Baumgartner C, Cerny-Reiterer S, Sonneck K, Mayerhofer M, Gleixner KV, Fritz R. Expression of activated STAT5 in neoplastic mast cells in systemic mastocytosis: subcellular distribution and role of the transforming oncoprotein KIT D816V. Am J Pathol. 2009; 175(6):2416-29. PubMedhttps://doi.org/10.2353/ajpath.2009.080953Google Scholar
- Warsch W, Grundschober E, Sexl V. Adding a new facet to STAT5 in CML: multitasking for leukemic cells. Cell Cycle. 2013; 12(12):1813-4. PubMedhttps://doi.org/10.4161/cc.25116Google Scholar
- Chatain N, Ziegler P, Fahrenkamp D, Jost E, Moriggl R, Schmitz-Van de Leur H. Src family kinases mediate cytoplasmic retention of activated STAT5 in BCR-ABL-positive cells. Oncogene. 2013; 32(31):3587-97. PubMedhttps://doi.org/10.1038/onc.2012.369Google Scholar
- Kaymaz BT, Selvi N, Gokbulut AA, Aktan C, Gunduz C, Saydam G. Suppression of STAT5A and STAT5B chronic myeloid leukemia cells via siRNA and antisense-oligonucleotide applications with the induction of apoptosis. Am J Blood Res. 2013; 3(1):58-70. PubMedGoogle Scholar
- Kosova B, Tezcanli B, Ekiz HA, Cakir Z, Selvi N, Dalmizrak A. Suppression of STAT5A increases chemotherapeutic sensitivity in imatinib-resistant and imatinib-sensitive K562 cells. Leuk Lymphoma. 2010; 51(10):1895-901. PubMedhttps://doi.org/10.3109/10428194.2010.507830Google Scholar
- Warsch W, Grundschober E, Berger A, Gille L, Cerny-Reiterer S, Tigan AS. STAT5 triggers BCR-ABL1 mutation by mediating ROS production in chronic myeloid leukaemia. Oncotarget. 2012; 3(12):1669-87. PubMedGoogle Scholar
- Casetti L, Martin-Lanneree S, Najjar I, Plo I, Auge S, Roy L. Differential contributions of STAT5A and STAT5B to stress protection and tyrosine kinase inhibitor resistance of chronic myeloid leukemia stem/progenitor cells. Cancer Res. 2013; 73(7):2052-8. PubMedhttps://doi.org/10.1158/0008-5472.CAN-12-3955Google Scholar
- Rousselot P, Roy L, Etienne G, Legros L, Charbonnier A, Coiteux V. Targeting STAT5 Expression Resulted in Molecular Response Improvement in Patients with Chronic Phase CML Treated with Imatinib. ASH Annual Meeting Abstracts. 2012; 120(21):696. Google Scholar
- Li G, Miskimen KL, Wang Z, Xie XY, Tse W, Gouilleux F. Effective targeting of STAT5-mediated survival in myeloproliferative neoplasms using ABT-737 combined with rapamycin. Leukemia. 2010; 24(8):1397-405. PubMedhttps://doi.org/10.1038/leu.2010.131Google Scholar
- Kim WS, Lee MJ, Kim DH, Lee JE, Kim JI, Kim YC. 5′-OH-5-nitro-Indirubin oxime (AGM130), an Indirubin derivative, induces apoptosis of Imatinib-resistant chronic myeloid leukemia cells. Leuk Res. 2013; 37(4):427-33. PubMedhttps://doi.org/10.1016/j.leukres.2012.12.017Google Scholar