The high remission rates observed in patients with chronic myelogenous leukemia who receive Imatinib mesylate (Gleevec) indicate that targeted therapy for hematological malignancies is achievable. At the same time, progress in cellular biology over the past decade has resulted in a better understanding of the process of malignant transformation, a better classification of subtypes of each disease on the basis of molecular markers, and a better characterization of the molecular targets for drug development. These advances have already spawned the development of such effective agents as monoclonal antibodies and specific enzyme inhibitors. This review attempts to provide a practical introduction to the complex and growing field of targeted therapy in hematological malignancies.

Cellular proliferation, differentiation, and death are regulated by a number of extracellular molecules such as cytokines and hormones, as well as intercellular interactions mediated by neighboring cell surface antigens. These effectors mediate gene transcription either directly or indirectly by activating intracellular signaling pathways, which in turn activate appropriate cellular machinery (1). Cell-surface receptors that convert external stimuli into intracellular signals are pivotal in this signaling process. They activate intracellular pathways either through their inherent enzymatic function or as a result of their association with other catalytic proteins. Indeed, most growth factors and cytokines bind these receptors and exert their function through their activation, commonly by phosphorylation (1).

Normal hematopoiesis is dependent on intricately regulated signaling cascades that are mediated by cytokines and their receptors. Orderly function of these pathways leads to the generation of appropriate constellation of hematopoietic cells, and their abnormal activation results in neoplastic transformation, impaired apoptosis, and uncontrolled proliferation. Cytokines function in a redundant and pleiotropic manner; different cytokines can exert similar effects on the same cell type, and any particular cytokine can have several differing biological functions (2). This complexity of function is a result of shared receptor subunits as well as overlapping downstream pathways culminating in activation of common transcription factors (3).

The signal transduction cascades involve three major classes of proteins: kinases, adaptor or docking proteins, and transcription factors. Early insights into the cellular signaling pathways came from studies of IFN function (4, 5, 6). These experiments led to the identification of a family of nonreceptor TKs2 called Jaks and their target proteins, Stats, which mediate gene transcription (4, 7). The Jak-Stat pathways are commonly activated during cytokine signaling through phosphorylation of specific tyrosine residues (3). The interaction of a cytokine with its receptor induces its tyrosine phosphorylation and leads to activation of downstream protein TKs including Jaks and Stats. Apart from their catalytic domain, protein TKs contain several other characteristic motifs including the SH2, SH3, and pleckstrin homology domain, which enable them to interact with other signaling molecules and propagate the message (8, 9).

The phosphorylation of serine and threonine residues is integral to the activation of numerous other intracellular proteins that mediate a number of other signaling pathways (10). Cytokine receptors without intrinsic kinase activity transmit their signals primarily through activation of Jak kinases. These receptors as well as those with intrinsic kinase activity, the RTKs, were previously thought to transmit their signals independently of the serine/threonine kinase cascades. More recently, it has been established that both of these pathways interact with serine/threonine kinase cascades such as the Ras/Raf/MEK/ERK (MAPK; Ref. 10). For example, after ligand binding, the βsubunit of IL-3 and GM-CSF receptors are phosphorylated and, through recruitment of adaptor proteins such as Shc, Grb2, and Sos, activate the Ras signaling pathway (11). This in turn activates Raf followed by downstream activation of ERK1 and ERK2, and increased expression of transcription factors c-fos and c-jun(12, 13, 14). Other members of the MAPK family such as p38, and JNK/SAPK are also activated after phosphorylation of their serine/threonine residues as a result of cytokine/receptor interaction (15, 16, 17, 18). Similarly, PI3K associates with the β chain of IL-3 receptor, recruits PKB/AKT by phosphorylation of its serine residues, and transmits cellular survival signals (19, 20, 21). Another downstream protein to IL-3 activation is the p70S6 kinase, which also interacts with the β chain and mediates appropriates signals (22, 23). Ultimately, these pathways influence gene transcription through their ability to recruit transcription factors, regulate apoptosis through the phosphorylation of apoptotic proteins, and cause the cell to progress through cell-cycle checkpoints by activation of specific kinases.

Of considerable interest is the description of a number of oncogenes with constitutive kinase activity. These molecules are derived from genes including c-ABL, c-FMS, FLT3, c-KIT, and PDGFRβ, which are normally involved in the regulation of hematopoiesis (24). The kinase activity of the oncogene is constitutively activated by mutations that remove inhibitory domains of the molecule or induce the kinase domain to adopt an activated configuration (24). As a result of such constitutive activation a number of signaling cascades such as the Jak-Stat pathway, the Ras/Raf/MAPK pathway, and the PI3K pathway are activated.

With better characterization of aberrant signaling through the cell surface receptors and their downstream pathways in neoplastic cells, current research is exploring ways to reverse such dysregulated stimuli (25). Here, we will briefly review the role of cellular signaling pathways in normal cellular processes, neoplastic transformation, and development of hematological malignancies. We then explore the possible ways that their modulation can lead to clinically meaningful benefits.

Hematopoietic cell proliferation and differentiation is regulated by a number of soluble polypeptides such as IFNs, interleukins, and colony-stimulatory factors known collectively as cytokines (26). Cytokines bind to their cognate receptors and mediate downstream effects. A cytokine receptor consists of a unique ligand binding subunit as well as a signal transducing subunit, which may be structurally similar to the other cytokine receptors (27, 28, 29). On the basis of their characteristic structural motifs in their extracellular domains a number of subfamilies of cytokine receptors have been identified (27, 29, 30). These include the gp130 family, the IL-2 receptor family, the growth hormone receptor family, the IFN family, and the gp140 family of receptors (3). A detailed description of the structure of these receptors is beyond the scope of this review, but in general they consist of two or more subunits including the α, β, and γ chains (3, 27). For example, the IL-2 receptor family includes the receptors for IL-2, IL-4, IL-7, IL-9, and IL-15 each consisting of a ligand-binding α-subunit, and signal transducing β and γ subunits. Alternatively, the gp140 family including the receptors for IL-3, IL-5, and GM-CSF have a unique ligand-binding α-subunit and a common β (gp140) signal-transducing unit (3).

Unlike growth factor receptors (RTKs), cytokine receptors do not possess a cytoplasmic kinase domain, and most cytokines transmit their signal by recruiting other TKs (3). Dimerization of the cytoplasmic component of the cytokine receptor as a result of ligand binding is the initial step in the initiation of cellular signaling (31). The dimerized subunits then associate with intracellular TKs such as members of the Src or Jak families of kinases, and the signal is propagated (Fig. 1). Different Src family members are associated with different receptors and phosphorylate distinct but overlapping sets of downstream target molecules. For example, Lck, Lyn, and Fyn can be activated by IL-2 (29, 32).

The Jak family of kinases comprises four known relatively large proteins (Jak1, Jak2, Jak3, and Tyk2) that can bind cytokine receptor subunits, phosphorylate them, and in doing so creat docking sites on the receptors for binding of SH2-containing proteins (33, 34). In general, Jaks consist of several domains (JH1-JH7) of which the functional significance has been characterized by mutational analysis and include a TK domain (JH1; Refs. 3, 33, 35, 36). The precise functions of JH2-JH7 domains are under current investigation (3). Jaks are able to associate with the cytokine receptors as well as with each other (37, 38). Dimerization/oligomerization of cytokine receptor subunits as a result of ligand binding leads to juxtaposition of Jaks (3). This results in transphosphorylation and activation of their kinase activity and the phosphorylation of downstream signaling proteins such as Stats, Src-kinases, and adaptors such as Shc, Grb2, and Cbl (Fig. 1; Refs. 39, 40).

Abnormalities of Jak function have been associated with a number of disorders (34, 41). For example, chromosomal translocations resulting in TEL-JAK2 constructs lead to the constitutive activation of Stat5, IL-3-independent cellular proliferation, and leukemogenesis (42, 43). The translocation t(9;12)(p24;p13) results in the fusion of the kinase catalytic region of JAK2 with the transcription factor TEL generating the constitutively active TEL-JAK2. Similarly, infection with oncogenic viruses such as human T-cell lymphotrophic virus, type I, and Abelson murine leukemia viruses results in enhanced TK activity of Jaks, possibly accounting for their leukemogenic potential (44, 45).

The Stat transcription factors are coded by six known mammalian genes and include 10 different Stat proteins including different isomers of Stats 1, 3, 4, and 5 (3, 46). Like other transcription factors Stats have a well-defined structure including a DNA-binding domain, a conserved NH2-terminal domain, a COOH-terminal transactivation domain, and SH2 and SH3 domains (3). Their activation through tyrosine phosphorylation results in their dimerization and translocation into the nucleus where they activate specific genes (6, 47, 48, 49).

Jak proteins activate a number of intracellular signaling proteins, among which Stats are the best defined (46, 50). Binding of a cytokine to its receptor rapidly induces tyrosine phosphorylation of the cytoplasmic domains of the receptor by activated Jak kinases, thus providing a docking site for Stat proteins, which are then phosphorylated. This phosphorylation of Stats leads to their homo- or heterodimerization and translocation to the nucleus, followed by DNA binding and gene activation (Fig. 1; Refs. 51, 52). The specificity for Stat phosphorylation is determined by the receptor docking sites and not the Jak kinases (53, 54). Also, different Stat proteins have different DNA-binding affinities, resulting in activation of specific genes. Stats also interact with other transcription factors such as the p300/cyclic AMP-responsive element binding protein family of coactivators to activate genes (55, 56). The transcriptional activity of Stats may also be regulated by the phosphorylation of their serine and threonine residues, although the implications of such regulation are not known (7, 57).

Stats mediate diverse and sometimes opposite cellular events affecting growth, differentiation, and apoptosis (58, 59). For example, Stats can mediate both growth arrest and cellular proliferation. Specifically, Stat1 mediates the growth-inhibitory effects of IFN-γ, through the induction of the CDKI p21waf1, whereas Stat5 mediates proliferative effects of IL-3 and GM-CSF (60, 61). Similarly, phosphorylation of Stat3 can result both in IL-6- and IL-10-induced growth arrest, and in GM-CSF- and IL-3-induced proliferation (61, 62, 63). Stats also modulate cellular differentiation and apoptosis. Reconstitution of Stat1 in Stat1-null U3A cells (which do not respond to TNF-α) restores basal caspase expression and renders them sensitive to TNF-induced apoptosis (64). Conversely, Stat3 and Stat5 mediate the antiapoptotic effects of IL-6 and IL-2, respectively (65, 66). Stat1 activates the caspase cascade through up-regulation of Fas and FasL expression in response to IFN-γ (67). The exact mechanisms underlying these diverse effects are being elucidated.

An important property of cellular signaling pathways is that their activation is both rapid and transient. This is because of effective mechanisms of inactivation. In the Jak-Stat system, proteasome-mediated degradation, tyrosine dephosphorylation, and inhibition by various inhibitory proteins are responsible for this process (4). The ubiquitin-proteasome pathway governs the degradation of many intracellular proteins including activated Stats, and effective inhibitors of this system have shown promising early results in clinical trials (68, 69). The cytokine-activated Jak-Stat pathways are also inhibited by tyrosine dephosphorylation mediated by cytoplasmic phosphatases such as SHP-1 (70, 71). SHP-1-deficient mice demonstrate multiple hematopoietic abnormalities, including hyperproliferation and abnormal activation of granulocytes and macrophages in the lungs and in the skin (72). SOCS and PIAS are other important inhibitors of the activated Jaks and Stats (70, 73). Recent studies have established that SOCS are negative regulators of cytokines, and there is ample evidence suggesting the importance of Stats in the induction of SOCS expression, thereby constituting a negative feedback mechanism (74, 75, 76). The role of these inhibitory proteins in the pathogenesis of neoplastic transformation is also becoming clearer (77).

RTKs are membrane-bound enzymes with an extracellular ligand-binding domain, a transmembrane domain, and a highly conserved intracellular domain that mediates the activation, through tyrosine phosphorylation, of a number of downstream signaling proteins (78, 79, 80). These enzymes are activated by ligand binding, by cell-cell interactions via cell adhesion molecules, and by stimulation of G-protein coupled receptors (81). Phosphorylated tyrosine residues in specific domains of these receptors serve as high-affinity docking sites for SH2-containing adaptor and effector proteins (82). RTKs include diverse molecules, which are considered as members of several distinct classes: class I including epidermal growth factor receptor; class II including insulin-like growth factor-1 receptor; class III including PDGFR, macrophage colony-stimulating factor (FMS-R or CSF-1R), stem cell factor receptor (KIT), and FLT3R; and class IV including FGFR (79, 83, 84). The importance of these receptors in malignant transformation and the possibility of modulating them as therapeutic targets are subjects of intense research. The recent reports of constitutive activation of FLT3R resulting in stimulation of multiple signaling pathways and leading to malignant transformation has been of significant interest in leukemia research (85, 86). Such constitutive activation of this receptor has been reported in >30% of patients with AML and results from two well-described molecular events. Internal tandem duplication mutations of FLT3R gene occur at exons 11 and 12 of the gene that code for the juxtamembrane domain of receptor (87, 88, 89, 90). More recently, point mutations of codon 835 of FLT3R receptor gene, located in the activation loop of its TK domain, have been reported in 7% of patients with AML (87, 91). Inhibitors of such aberrant activation are undergoing clinical evaluation (92, 93).

Many intracellular signaling proteins bind the phosphotyrosine on the activated RTKs. These proteins include GTPase activating protein, PI3K, Grb2, and Src-like tyrosine-kinases (1, 10, 78). The activation of these proteins by serine/threonine phosphorylation in turn activates a number of downstream signaling cascades that lead to gene transcription (10).

Although knowledge of the Jak-Stat pathway has been instrumental in understanding cytokine signaling (94), the importance of signaling cascades that involve the activation of serine/threonine kinases is increasingly apparent (10). The serine/threonine MAPKs, which include the Ras-Raf-MEK-ERK pathway, the p38 family of kinases, and the JNK (SAPK) family, are activated by upstream signals and mediate effects on inflammation, cell growth, cell cycle progression, cell differentiation, and apoptosis (95). The Ras family of proteins belongs to the large superfamily of GTPases that localize to the inner surface of the plasma membrane (1, 96). Ras proteins play a pivotal role in a number of signaling pathways mediated by RTKs and other receptors. Ligand binding to these receptors initiates the autophosphorylation of specific tyrosine residues in their cytoplasmic domain and creates phosphotyrosyl-binding sites for adapter proteins such as Shc and Grb2, which in turn recruit guanine nucleotide exchange factors and thereby initiate Ras activation (97, 98).

Once induced, Ras activates Raf serine/threonine kinase, which then phosphorylates MAPK kinases (otherwise known as MEKs; Refs. 10, 97, 99). These in turn activate MAPKs (or ERKs; Refs. 100, 101), which in turn move to the nucleus where they phosphorylate and activate nuclear transcription factors such as Elk-1 (102). ERKs were initially described as a novel family of protein kinases that, when activated, produced proliferative stimuli (103). ERKs can also activate other kinases such as RSKs (also known as MAPK-activated protein kinases), which are involved in cell-cycle regulation and apoptosis (104). ERK-activated RSK kinase catalyzes the proapoptotic protein Bad and suppresses Bad-mediated apoptosis (105). Similarly, the Ras-Raf-MEK-ERK cascade modulates cellular proliferation by regulating the activity of several proteins, including cell-cycle regulators (e.g., cyclin D1, p21waf1/cip1, p27kip1, and cdc25A) and transcription factors (e.g., c-Myc; Ref. 106).

The G1/S cell cycle checkpoint is a critical point determining the commitment of cells to growth arrest or proliferation. During this stage cells are responsive to cytokines (107). Regulatory proteins p21waf1/cip1 and p27kip1 are of particular importance in this transition, which is controlled by both positive and negative regulators. Distinct Rb-E2F repressor complexes suppress the transcription of genes required for progression of various phases of cell cycle. For example, Rb-E2F1 complex suppresses the progression through G1(108). During this progression from G1 to S-phase cyclin/CDKs are sequentially activated, which then inactivate suppressor complexes such as Rb-E2F1 (109). Cyclin/CDK activity results in Rb phosphorylation and its dissociation from E2F1 leading to activation of genes necessary for S phase (110). Activity of a number of these cyclin/CDKs as well their inhibitors such as p21waf1/cip1 is modulated by cytokine-mediated signals through their phosphorylation.

The p38 family of MAPKs is involved in various cellular processes such as inflammation, cell cycle progression, and cell death (111, 112). The four different p38 isoforms (α, β, γ, and δ) are activated by two MEK isoforms (113). Originally, the p38 kinase pathway was reported to have a critical role in the generation of signals in response to stress stimuli. However, its role in cytokine signaling and regulation of the Jak-Stat pathway has been elucidated recently (114). In particular, from the standpoint of leukemogenesis, it modulates the growth-inhibitory effect of type I IFNs in BCR-ABL-expressing cells as well as normal hematopoietic progenitors (115, 116).

The third group of MAPKs includes the JNK (otherwise known as SAPK; Ref. 95). The four different JNK kinases have a similar role to p38 kinases in cellular function and are activated by specific MAPK kinases (MEKKs) in response to inflammatory cytokines such as TNF-α, and other stress stimuli such as reactive oxygen species, heat, and withdrawal of growth factors (117). The MEKK1/JNK signaling increases p53 stability and transcriptional activation, and MEKK1/JNK potentiates the ability of p53 to initiate apoptosis (118).

Normal functioning of MAPK-mediated signaling necessitates its efficient inactivation (95). A number of dual-specificity MAPK phosphatases serve to dephosphorylate and, hence, inactivate MAPKs (119, 120, 121). Similarly, protein phosphatases PP1 and PP2 dephosphorylate and inactivate a number of phosphoproteins including components of the MAPK pathway (122, 123).

Other signaling pathways such as those mediated by PI3K, AKT (also known as PKB), and protein kinase C are also controlled by serine/threonine phosphorylation (Fig. 2; Ref. 10). PI3K consists of two subunits, the p85 regulatory subunit and the p110 catalytic subunit (124, 125). The p85 subunit binds to the cytokine receptor as a consequence of ligand-receptor interaction and receptor autophosphorylation (126). As a result, phosphatidylinositol-dependent kinases and their downstream substrate AKT/PKB are recruited to the membrane (127). PI3K-AKT pathway activates several downstream targets including p70 RSK, forkhead transcription factors, and NFκB (128, 129, 130). The serine/threonine kinase AKT is an important component of the cell survival machinery (10, 131, 132, 133). Its activation via the PI3K pathway leads to a number of events (10, 131, 134, 135). For example, the phosphorylation of the cytosolic protein IκB by AKT releases NFκB from its association with IκB. NFκB then moves into the nucleus, where it induces a number of genes involved in cell survival (131). Meanwhile, the inhibitory protein IκB is degraded by the proteasome (136). AKT also phosphorylates the proapoptotic protein Bad, which leads to higher levels of free antiapoptotic Bcl-xL and thereby inhibits the cell-death protease caspase-9 (134). The tumor suppressor gene PTEN codes for a phosphatase that acts by removing a phosphate group from the 3 position of the inositol ring of the PIP3,4,5 phospholipids located at the cellular membrane. This prevents the proximation of AKT and phosphatidylinositol-dependent kinases, and prevents AKT activation (137, 138, 139, 140). Several lines of evidence including studies of PTEN knockout mice support the role of PTEN as a tumor suppressor gene (141). Serine/threonine kinases, in general, also influence the activity of other antiapoptotic proteins of the Bcl-2 family (10, 135, 142). In the normal cell cycle, Bcl-2 is phosphorylated on its serine/threonine residues at several points during the G2 to M phase transition (10, 143).

PKC, another important signaling enzyme, phosphorylates specific serine or threonine residues on target proteins in different ways (Fig. 2). For example, PKC is a potent activator of Raf-1, which activates the MAPK cascade. This leads to phosphorylation of IκB, release of NFκB, translocation of NFκB into the nucleus, and gene transcription as described above (144, 145, 146). PKC also regulates cytokine signals through its effects on the Jak-Stat pathway in some myeloid progenitor cell lines (147). The significant role of PKC in phosphorylation and activation of Raf has led to its targeting for inhibition of the Raf-Ras-MEK-ERK pathway (95, 148). For example, staurosporine analogs UCN-O1 and CGP 41251 are being examined as inhibitors of PKC and MAPK signaling (149, 150).

Various signaling pathways interact resulting in their modulation at several levels. For example, PI3K interacts with and enhances Raf-Ras-MEK-ERK pathway (151, 152, 153). Other serine/threonine phosphorylation pathways modulate cytokine signals through the Jak-Stat pathway (10, 154). In addition to being tyrosine phosphorylated, several Stats (Stat1α, Stat3, and Stat4) are serine phosphorylated by ERKs at conserved serine residues (155). In fact, during cytokine signaling, Jak and Raf kinases carry on an intricate cross-talk (10).

Signaling pathways may be particularly attractive targets in cancer therapy because they may be inappropriately activated in malignant cells. However, several factors must be carefully considered while developing agents that modulate these pathways. One is the possible toxicity of such therapy. Because these pathways are activated to a significantly greater degree in malignant cells than normal cells, their partial inhibition may be sufficient to interfere with malignant cell growth without causing significant toxicity (25). Therefore, despite the pivotal role of these pathways in normal cellular function, their inhibition may not be toxic to normal cells.

Another point of consideration in designing inhibitors is the specificity of the target pathway and the selectivity of its inhibitors. Adding to this challenge is the fact that these pathways are part of a complex network of interconnecting cascades resulting in a certain degree of redundancy and overlap.

Diseases induced by specific oncogenic alterations leading to constitutive activation of pivotal molecules in the pathways may provide us with such specificity and selectivity. A number of translocations occurring in hematological malignancies are known to result in fusion genes with enhanced kinase activity (24). The oncoprotein Bcr-Abl results from a translocation between chromosomes 9 and 22, and occurs in CML and ALL. Its transforming activity is the product of activation of a number of signaling molecules such as Ras, Raf, PI3K, JNK/SAPK, Crkl, and Stat5 (156, 157, 158, 159, 160). As a result of their activation, a number of pathways, which inhibit apoptosis and promote cell survival, are induced (24). Bcr-Abl is constitutively phosphorylated on a number of tyrosine residues, allowing the docking of adaptor proteins such as Cbl, Crkl, Shc, and Grb2, which recruit the Ras signaling pathway, the p85 regulatory subunit of PI3K, the focal adhesion proteins paxillin and talin, and a number of other signaling pathways (159, 161, 162, 163). The specific Bcr-Abl inhibitor Imatinib mesylate has proven to be very effective in suppressing these pathways in vitro and in patients with CML and ALL (164, 165, 166, 167, 168, 169, 170).

Although the platelet-derived growth factors, PDGFRα and PDGFRβ have significant homology, only the PDGFRβ has been implicated in hematological cancers where a significant number of patients with chronic myelomonocytic leukemia have t(5;12)(q33;p13) generating the fusion protein TEL-PDGFRβ (171, 172). As a result of ligand-independent dimerization and autophosphorylation of the PDGF β-subunit, the TK is constitutively active. PDGF is able to stimulate the growth of primitive hematopoietic, erythroid, and megakaryocytic precursors, and TEL-PDGFRβ can confer cytokine-independent growth to Ba/F3cells (173). Imatinib mesylate inhibits the kinase PDGFRβ and has been shown anecdotally to be effective in patients with this translocation (174).

Similarly, the chimeric gene NPM-ALK is produced as a result of translocation t(2;5)(p23;q35; Refs. 175, 176). Fusion of NH2-terminal domain of NPM to the cytoplasmic region of the ALK TK receptor results in constitutive activation of its catalytic domain (177, 178). NPM-ALK associates with a number of adaptor proteins such as Grb2 and Crkl, and results in the activation of the downstream signaling proteins such as PI3K, AKT, and Stat5 (179, 180, 181, 182). Therefore, design of specific inhibitors of the NPM-ALK may be of considerable interest in treating patients with anaplastic large cell lymphoma where up to 50% of patients express the chromosomal translocation (183).

The TK modulated pathways such as the Jak-stat cascade can be targeted at several steps along the way (Fig. 3). Because a number of cytokines and growth hormones play an important role in the suppression of apoptosis in the malignant clone in hematological cancers (e.g., IL-6 in myeloma; IL-2 in some T-cell lymphomas; and IL-1, IL-3, and GM-CSF in AML; Refs. 25, 184), inhibition of the cytokines and their receptors is a plausible therapeutic strategy (185, 186). Furthermore, where there is constitutive activation of the receptor leading to neoplastic change, selective inhibition of kinase activity of the receptor is likely to be of benefit. For example, inhibition of FLT3-R TK activity is selectively cytotoxic to AML blasts harboring the appropriate activating mutations (92, 187). Activating mutations of FLT3-R including the internal tandem duplication mutation and mutation of the TK domain occur in >30% of patients with AML, confer adverse prognosis, and can be targeted by selective inhibitors (92, 188, 189). Clinical trials examining the efficacy and safety of such inhibitors are currently under way. Intracellular activators of Stats such as Jaks and Src are also likely targets (164, 165, 166). For example, the Jak2 inhibitor AG490 inhibited the growth of ALL cells in a mouse model without affecting normal hematopoiesis (190). AG490 also acts by inhibiting Stat function, and induces apoptosis in Sezary cells and myeloma cells (191, 192). Furthermore, numerous other cellular TKs have been identified that likely have roles in neoplastic cell survival and proliferation, and a number of TK inhibitors are already being explored for their effects on signaling cascades. In general, two classes of TK inhibitors are being developed: inhibitors of the ATP binding site and inhibitors of the substrate binding sites (164, 193, 194).

There are other ways to interrupt Stat signaling. One is the specific inhibition of the Stat SH2 domain, because it is essential for the recruitment of Stats to the receptor subunit and for Stat dimerization (25). Antisense oligonucleotides directed at Stats (195), modulation of the activity of natural Stat inhibitors such as SOCS and PIAS, and inhibition of the binding of activated Stat dimers to their DNA targets (25, 196, 197) are other mechanisms currently under investigation.

A better understanding of the effects of current antineoplastic agents on these pathways may also elucidate their mechanism of action and lead to the development of more effective and less toxic agents. For example, fludarabine causes a profound and specific loss of Stat1 (198), which may be at least partly responsible for its antineoplastic properties as well as its immunosuppressive properties, which is similar to Stat1 knockout mice.

Multiprotein complexes, which can modify the structure of chromatin and influence gene regulation, are often aberrant in hematological malignancies. Although their interactions with various signaling pathways have not been clearly defined, they merit specific mention as they are subject of intense research. Such complexes include proteins such as HDAC, histone acetyltransferase, and DNA methyltransferases. In a significant proportion of patients with AML-M2, the translocation t(8;21)(q22;q22) results in the fusion of the DNA-binding domain of AML1 with ETO, which interacts with corepressor complexes such HDAC and repress cellular differentiation (199, 200). Similarly, t(12;21)(p13;q22) in childhood ALL results in recruitment of the repressor N-CoR by the fusion protein TEL-AML1 (201, 202). Inhibitors of HDACs and DNA methyltransferase such as butyrates, trichostatin A, 5-azacytidine, and 2′-deoxy-azacytidine are currently under investigation in clinical trials (203, 204).

The most common translocation in APL, t(15;17)(q22;q11.2), results in the fusion of RARα with the PML genes, with the resulting fusion protein retaining the functional domains of both (205, 206). The fusion protein recruits nuclear receptor corepressors SMRT and N-CoR, which together with HDACs keep the chromatin in the closed configuration and suppress/inactivate retinoid receptor target genes in myeloid development (207, 208). In patients with PML-RARα, pharmacological doses of ATRA can convert the fusion protein from a dominant-negative inhibitor of retinoid-regulated transcription to an activator (209). Signaling pathways that interact with retinoid-mediated transcription may be modulated to potentiate ATRA-induced differentiation (210). ATRA may also exert effects on upstream signaling pathways. For example, ATRA up-regulates Stat1 and Stat2 activity in the IFN-α signaling pathway, thereby potentiating the growth-inhibitory effects of IFN-α (211, 212, 213). Another novel therapeutic agent in APL, arsenic trioxide, affects a number of cellular signaling pathways, leading to induction of apoptosis and differentiation (214, 215, 216, 217). Therapeutic effects of arsenic trioxide in APL are partly related to its degradation of PML-RARα fusion protein through targeting PML to proteasomal degradation by Sumolation (218). However, arsenic trioxide has a number of other effects such as activation of JNK kinases; translocation of PKC from cytosol to plasma membrane to mediate downstream signaling; enhancement of CDKIs, p21waf1/cip1, and p27kip1; and inhibition of IκB kinase and NFκB (214).

Monoclonal antibodies such as rituximab (anti-CD20) and alemtuzumab (anti-CD52) are increasingly used in treating hematological malignancies (219, 220, 221). Their exact mechanism of action is largely unknown. However, understanding their effects on the signaling pathways downstream of the target receptor is likely to point the way to more effective agents. The ligation of CD20 by rituximab in the presence of FcR-expressing cells may initiate signal transduction events that induce an elevation of intracellular Ca2+ and lead to apoptosis of the target malignant B cells (222). Rituximab has also been shown in vivo to activate caspases in blood leukemia cells immediately after its infusion suggesting a mechanism of action independent of complement-mediated cell lysis or antibody-dependent cellular cytotoxicity (223, 224).

Inhibition of serine/threonine kinases involved in RTK signaling is another major area of research and development. As noted previously, Ras proteins play a significant role in human carcinogenesis. Therefore, inhibition of Ras signal transduction has been the subject of intense research. Ras-mediated signaling can be inhibited by prevention of its membrane localization, by inhibition of Ras protein expression using antisense nucleotides, and by inhibition of its downstream targets (95, 225). Pharmacological inhibitors of farnesyltransferase, the enzyme responsible for the COOH-terminal prenylation of Ras and, hence, its membrane association and transforming activity, have already been developed, and clinical trials of their efficacy in leukemia are under way (96, 226, 227, 228, 229). Four such farnesyl transferase inhibitors are available at various stages of testing: R115777, SCH66336, L778123, and BMS214662 (95, 230, 231). FTIs may also have Ras-independent effects on other cellular signaling components that contribute to their antineoplastic action (232). Other steps in the serine/threonine cascades, from the cell surface receptor to gene transcription, are also being examined as possible targets for therapy (Ref. 95; Fig. 4). Compounds such as geldanamycin and derivatives of radicicol are known to destabilize Raf protein and interfere with Raf signaling (233, 234, 235). Several staurosporine derivatives including UCN-O1, CGP41251, and PKC412 are able to inhibit PKC signaling, and have been examined in cell lines and clinical studies (149, 150, 236). Aberrant MEK and ERK activity has been demonstrated in AML and CML (237, 238, 239, 240), and MEK inhibitors such as PD098059, PD184352, and UO126 are able to modulate cellular proliferation, differentiation, and apoptosis (241, 242, 243). PD184352 has been examined in Phase I trials in patients (244). Pharmacological inhibitors of PI3K, wortmannin, and LY294002 have shown significant potency in preclinical studies (245, 246).

Downstream targets of these signaling pathways involved in cell-cycle regulation, cell survival, and apoptosis can also be modulated (Table 1). For examples, agents capable of promoting apoptosis are being developed (247). These may function by activating the caspase cascade through blockade of the IAPs, inhibition of the NFκB pathway (using IκB kinase inhibitors or proteasome inhibitors), and inhibition of the PI3K pathway (using kinase inhibitors; Refs. 69, 134, 248). IAPs including the XIAP can block the process of programmed cell death through their interaction with members of the caspase family of cysteine proteases (249). The human family of caspases includes 11 members involved in processing of inflammatory cytokines as well as execution of apoptosis (250). Dysregulation of caspase cascade has been implicated in human neoplasia, and IAPs, the natural inhibitors of caspases, are pivotal in this regulatory process (250, 251). XIAP, the most potent member of the IAP family blocks both the initiator caspase-9, and the effector caspases-3 and -7 (252). An association between XIAP levels and survival has been reported in AML (253). Down-regulation of XIAP protein by adenoviral antisense vector results in sensitization to radiation-induced cell death (254).

The ubiquitin-proteasome pathway is the central pathway for degradation of intracellular proteins (255, 256). Targeting of proteins to proteasome is through covalent attachment of a poly-ubiquitin chain. This system is specific allowing up-regulation of degradation of certain proteins without affecting the proteolysis of other substrates; this enables the proteasome to function as a regulator of metabolic pathways (256). Therefore, different classes of proteasome inhibitors can be used to differentially affect cellular levels of oncogenic proteins (256). Important substrates for proteasome degradation include cyclins and CDKIs; transcription factors such as p53, NFκB, c-Myc, c-fos, and c-Jun; a number of apoptosis family of proteins; IAPs; and some caspases (256, 257, 258, 259, 260, 261, 262). PS-341 is a specific and potent inhibitor of proteasome (263). It induces apoptosis and overcomes resistance in a number of cell lines as well as primary cells from patients with chronic lymphocytic leukemia (264, 265, 266, 267, 268). Its in vitro activity in myeloma has led to its ongoing examination in clinical trials (269, 270, 271).

Cell-cycle proteins such as the CDKs and their inhibitors (CDKIs), which modulate cell-cycle progression in response to appropriate signals, are also possible targets. Orderly cell division requires a tight control of cyclins and CDKs, particularly at the G1/S checkpoint (272). Complexes of Rb family of tumor suppressors with E2F transcription factors inhibit the transcription of several genes involved in the S phase of cell cycle both by active repression as well as through the recruitment of HDACs (273, 274). Phosphorylation of Rb by cyclin/CDK complexes leads to release of E2Fs and transactivation of target genes by binding of E2Fs to E2F response elements (275). The activity of cyclin D:CDK4/6 and cyclin E:CDK2 complexes is regulated by CDKIs of the INK4 and KIP families such as p16ink4a, p15ink4b, p18ink4c, p19ink4d, p21waf1/cip1, p27kip1, and p57kip2, which act as inhibitors of G1/S transition. Disrupted activity of a number of G1 checkpoint genes/proteins has been implicated in carcinogenesis (275). Decreased Rb expression, and a negative correlation between Rb expression and survival have been reported in ALL (276). Deletions and mutations of Rb have been reported in a number of hematological malignancies (276). Genes for p16ink4a and p15ink4b are suppressed in various hematological cancers mainly by deletion and by hypermethyl-ation of their promoters (277, 278). The role of inactivation of these genes as a prognostic indicator has been controversial (275). Alternatively, oncogenic amplification, and overexpression of cyclins and CDKs have been reported in a number of hematological cancers (279). Of note are overexpression of cyclin D1 in mantle cell lymphoma and hyperactivation of cdk4/cdk6 in human T-cell leukemia virus type 1 leukemias (279). Inhibitors of cyclins and CDKs such as flavopiridol, and the staurosporine derivative UCN-01 are under active development (248, 280). Early phase clinical trials of these agents have been reported recently (279, 281). However, it is likely that the majority these drugs will be used in combination with standard cytotoxic chemotherapy agents or for therapy of minimal residual disease.

Over the past decade extensive research has elucidated the role of cellular signaling pathways in cellular growth, differentiation, and apoptosis. The disruption of these pathways, on the other hand, has been shown to promote the neoplastic transformation of cells. This knowledge has led to the development of a number of molecules that in preclinical and clinical studies have been shown to be capable of reversing the neoplastic process. Our current challenge is to further understand the complexity of these processes and to devise specific agents that target the neoplastic cells whereas sparing normal tissues. This would truly realize the concept of targeted therapy in treating hematological malignancies and solid tumors alike.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

2

The abbreviations used are: TK, tyrosine kinase; Jak, janus kinase; Stat, signal transducer and activator of transcription; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; IL, interleukin; GM-CSF, granulocyte macrophage colony-stimulating factor; JNK, c-Jun NH2-terminal kinase; SAPK, stress-activated protein kinase; PKB, protein kinase B; PI3K, phosphatidylinositol 3′-kinase; PDGFR, platelet-derived growth factor receptor; TNF, tumor necrosis factor; SHP-1, SH-2 domain containing protein tyrosine phosphatase-1; SOCS, suppressor of cytokine signaling; PIAS, protein inhibitor of activated stat; RTK, receptor tyrosine kinase; FLT3R, FMS-like tyrosine kinase 3 receptor; Grb2, growth factor receptor binding protein 2; RSK, ribosomal S6 kinase; AML, acute myeloid leukemia; CML, chronic myelogenous leukemia; ALL, acute lymphoblastic leukemia; MEK, mitogen-activated protein/extracellular signal-regulated kinase kinase; ALK, anaplastic lymphoma kinase; NPM, nucleophosmin; ATRA, all-trans-retinoic acid; APL, acute promyelocytic leukemia; NFκB, nuclear factor κB; IκB, inhibitor of nuclear factor-κB; HDAC, histone deacetylase; IAP, inhibitor of apoptotic pathway; CDK, cyclin-dependent kinase; CDKI, cyclin-dependent kinase inhibitor; Rb, retinoblastoma; RAR, retinoic acid receptor; XIAP, X-linked inhibitor of apoptosis.

Fig. 1.

The Jak-Stat pathway and its regulation.

Fig. 1.

The Jak-Stat pathway and its regulation.

Close modal
Fig. 2.

Interactions of diverse signaling pathways.

Fig. 2.

Interactions of diverse signaling pathways.

Close modal
Fig. 3.

Targets for inhibition in Jak-Stat signaling pathway.

Fig. 3.

Targets for inhibition in Jak-Stat signaling pathway.

Close modal
Fig. 4.

Targets for inhibition in RTK signaling pathways.

Fig. 4.

Targets for inhibition in RTK signaling pathways.

Close modal
Table 1

Targets for drug development

TargetPrototype drugMode of action
Tyrosine and serine/threonine kinases Rituxan, Campath-1H Ligand-binding inhibition 
 CEP-701, AG1295 FLT3R kinase inhibition 
 Genistein Non-specific kinase 
 Limonene, perillyl alcohol, R115777, SCH66336, L778123, BMS214662 Ras FT inhibitiona 
 Geldanamycin, radicicol Raf inhibition 
 Wortmannin, LY294002 PI3K inhibition 
 Rapamycin P70S6K inhibition 
 PD098059, PD184352, UO126 MEK inhibition 
 AG490 JAK inhibition 
 Imatinib mesylate ABL, PDGFR, KIT 
Cell-cycle proteins Flavopiridol, Olomoucine, UCN-01 CDK inhibition 
 Purvalanol Cdc2 inhibition 
Apoptosis-related proteins Staurosporine, UCN-O1, CGP41251, PKC412 Bcl-2 inhibition (PKC) 
 Bryostatin PKC activation 
 — P53-MDM2 inhibition 
 PS341 Proteasome inhibition 
Angiogenesis-related proteins Suramin, SU5416 VEGFR and bFGFR 
Cell life span targets Anthraquinones Telomerase inhibition 
Transcription factors (Myc, Ets, Fos, Jun, Rel, Myb)   
TargetPrototype drugMode of action
Tyrosine and serine/threonine kinases Rituxan, Campath-1H Ligand-binding inhibition 
 CEP-701, AG1295 FLT3R kinase inhibition 
 Genistein Non-specific kinase 
 Limonene, perillyl alcohol, R115777, SCH66336, L778123, BMS214662 Ras FT inhibitiona 
 Geldanamycin, radicicol Raf inhibition 
 Wortmannin, LY294002 PI3K inhibition 
 Rapamycin P70S6K inhibition 
 PD098059, PD184352, UO126 MEK inhibition 
 AG490 JAK inhibition 
 Imatinib mesylate ABL, PDGFR, KIT 
Cell-cycle proteins Flavopiridol, Olomoucine, UCN-01 CDK inhibition 
 Purvalanol Cdc2 inhibition 
Apoptosis-related proteins Staurosporine, UCN-O1, CGP41251, PKC412 Bcl-2 inhibition (PKC) 
 Bryostatin PKC activation 
 — P53-MDM2 inhibition 
 PS341 Proteasome inhibition 
Angiogenesis-related proteins Suramin, SU5416 VEGFR and bFGFR 
Cell life span targets Anthraquinones Telomerase inhibition 
Transcription factors (Myc, Ets, Fos, Jun, Rel, Myb)   
a

Ras FT, Ras farnesyl transferase; MDM2, mouse double minute-2; PKC, protein kinase C; VEGFR, vascular endothelial growth factor receptor; bFGFR, basic fibroblast growth factor receptor.

1
Alberts B., Bray D., Lewis J., Raff M., Roberts K., Watson J. D. Molecular Biology of the Cell Ed. 3
721
-785, Garland Publishing Inc. New York, London  
1994
.
2
Kishimoto T. The biology of interleukin-6.
Blood
,
74
:
1
-10,  
1989
.
3
Rane S. G., Reddy E. P. JAKs. STATs and Src kinases in hematopoiesis.
Oncogene
,
21
:
3334
-3358,  
2002
.
4
Shuai K. The STAT family of proteins in cytokine signaling.
Prog. Biophys. Mol. Biol.
,
71
:
405
-422,  
1999
.
5
Ihle J. N., Thierfelder W., Teglund S., Stravapodis D., Wang D., Feng J., Parganas E. Signaling by the cytokine receptor superfamily.
Ann. N. Y. Acad. Sci.
,
865
:
1
-9,  
1998
.
6
Darnell J. E., Jr., Kerr I. M., Stark G. R. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins.
Science (Wash. DC)
,
264
:
1415
-1421,  
1994
.
7
Decker T., Kovarik P. Transcription factor activity of STAT proteins: structural requirements and regulation by phosphorylation and interacting proteins.
Cell Mol. Life Sci.
,
55
:
1535
-1546,  
1999
.
8
Cooper J. A., Howell B. The when and how of Src regulation.
Cell
,
73
:
1051
-1054,  
1993
.
9
Pawson T. Protein modules and signalling networks.
Nature (Lond.)
,
373
:
573
-580,  
1995
.
10
McCubrey J. A., May W. S., Duronio V., Mufson A. Serine/threonine phosphorylation in cytokine signal transduction.
Leukemia (Baltimore)
,
14
:
9
-21,  
2000
.
11
Pratt J. C., Weiss M., Sieff C. A., Shoelson S. E., Burakoff S. J., Ravichandran K. S. Evidence for a physical association between the Shc-PTB domain and the β c chain of the granulocyte-macrophage colony-stimulating factor receptor.
J. Biol. Chem.
,
271
:
12137
-12140,  
1996
.
12
Okuda K., Sanghera J. S., Pelech S. L., Kanakura Y., Hallek M., Griffin J. D., Druker B. J. Granulocyte-macrophage colony-stimulating factor, interleukin-3, and steel factor induce rapid tyrosine phosphorylation of p42 and p44 MAP kinase.
Blood
,
79
:
2880
-2887,  
1992
.
13
Coffer P. J., Geijsen N., M’Rabet L., Schweizer R. C., Maikoe T., Raaijmakers J. A., Lammers J. W., Koenderman L. Comparison of the roles of mitogen-activated protein kinase kinase and phosphatidylinositol 3-kinase signal transduction in neutrophil effector function.
Biochem. J.
,
329
:
121
-130,  
1998
.
14
Itoh T., Muto A., Watanabe S., Miyajima A., Yokota T., Arai K. Granulocyte-macrophage colony-stimulating factor provokes RAS activation and transcription of c-fos through different modes of signaling.
J. Biol. Chem.
,
271
:
7587
-7592,  
1996
.
15
Nagata Y., Moriguchi T., Nishida E., Todokoro K. Activation of p38 MAP kinase pathway by erythropoietin and interleukin- 3.
Blood
,
90
:
929
-934,  
1997
.
16
Nagata Y., Nishida E., Todokoro K. Activation of JNK signaling pathway by erythropoietin, thrombopoietin, and interleukin-3.
Blood
,
89
:
2664
-2669,  
1997
.
17
Foltz I. N., Schrader J. W. Activation of the stress-activated protein kinases by multiple hematopoietic growth factors with the exception of interleukin-4.
Blood
,
89
:
3092
-3096,  
1997
.
18
Terada K., Kaziro Y., Satoh T. Ras-dependent activation of c-Jun N-terminal kinase/stress-activated protein kinase in response to interleukin-3 stimulation in hematopoietic BaF3 cells.
J. Biol. Chem.
,
272
:
4544
-4548,  
1997
.
19
Coffer P. J., Schweizer R. C., Dubois G. R., Maikoe T., Lammers J. W., Koenderman L. Analysis of signal transduction pathways in human eosinophils activated by chemoattractants and the T-helper 2-derived cytokines interleukin-4 and interleukin-5.
Blood
,
91
:
2547
-2557,  
1998
.
20
Tilton B., Andjelkovic M., Didichenko S. A., Hemmings B. A., Thelen M. G-Protein-coupled receptors and Fcγ-receptors mediate activation of Akt/protein kinase B in human phagocytes.
J. Biol. Chem.
,
272
:
28096
-28101,  
1997
.
21
Songyang Z., Baltimore D., Cantley L. C., Kaplan D. R., Franke T. F. Interleukin 3-dependent survival by the Akt protein kinase.
Proc. Natl. Acad. Sci. USA
,
94
:
11345
-11350,  
1997
.
22
Sato N., Sakamaki K., Terada N., Arai K., Miyajima A. Signal transduction by the high-affinity GM-CSF receptor: two distinct cytoplasmic regions of the common β subunit responsible for different signaling.
EMBO J.
,
12
:
4181
-4189,  
1993
.
23
Calvo V., Wood M., Gjertson C., Vik T., Bierer B. E. Activation of 70-kDa S6 kinase, induced by the cytokines interleukin-3 and erythropoietin and inhibited by rapamycin, is not an absolute requirement for cell proliferation.
Eur. J. Immunol.
,
24
:
2664
-2671,  
1994
.
24
Scheijen B., Griffin J. D. Tyrosine kinase oncogenes in normal hematopoiesis and hematological disease.
Oncogene
,
21
:
3314
-3333,  
2002
.
25
Frank D. A. STAT signaling in the pathogenesis and treatment of cancer.
Mol. Med.
,
5
:
432
-456,  
1999
.
26
Metcalf D. The molecular control of cell division, differentiation commitment and maturation in haemopoietic cells.
Nature (Lond.)
,
339
:
27
-30,  
1989
.
27
Taniguchi T. Cytokine signaling through non-receptor protein tyrosine kinase.
Science (Wash. DC)
,
268
:
251
-255,  
1995
.
28
Hunter T. Signal transduction. Cytokine connections.
Nature (Lond.)
,
366
:
114
-116,  
1993
.
29
Stahl N., Yancopoulos G. D. The αs, βs, and kinases of cytokine receptor complexes.
Cell
,
74
:
587
-590,  
1993
.
30
Kishimoto T., Taga T., Akira S. Cytokine signal transduction.
Cell
,
76
:
253
-262,  
1994
.
31
Carpenter L., Yancopoulos G., Stahl N. General mechanisms of cytokine receptor signaling.
Adv. Protein Chem.
,
52
:
109
-140,  
1999
.
32
Kobayashi N., Kono T., Hatakeyama M., Minami Y., Miyazaki T., Perlmutter R. M., Taniguchi T. Functional coupling of the src-family of kinases p59fyn and p53/56lyn with the interleukin 2 receptor: Implications for redundancy and pleiotropism in cytokine signal transduction.
Proc. Natl. Acad. Sci. USA
,
90
:
4201
-4205,  
1993
.
33
Yeh T. C., Pellegrini S. The Janus kinase family of protein tyrosine kinases and their role in signaling.
Cell. Mol. Life Sci.
,
55
:
1523
-1534,  
1999
.
34
Ward A. C., Touw I., Yoshimura A. The Jak-Stat pathway in normal and perturbed hematopoieis.
Blood
,
95
:
19
-29,  
2000
.
35
Zhao Y., Wagner F., Frank S. J., Kraft A. S. The amino-terminal portion of the JAK2 protein kinase is necessary for binding and phosphorylation of the granulocyte-macrophage colony- stimulating factor receptor β c chain.
J. Biol. Chem.
,
270
:
13814
-13818,  
1995
.
36
Saharinen P., Takaluoma K., Silvennoinen O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain.
Mol. Cell. Biol.
,
20
:
3387
-3395,  
2000
.
37
Livnah O., Stura E. A., Middleton S. A., Johnson D. L., Jolliffe L. K., Wilson I. A. Crystallographic evidence for preformed dimers of erythropoietin receptor before ligand activation.
Science (Wash. DC)
,
283
:
987
-990,  
1999
.
38
Remy I., Wilson I. A., Michnick S. W. Erythropoietin receptor activation by a ligand-induced conformation change.
Science (Wash. DC)
,
283
:
990
-993,  
1999
.
39
Duhe R. J., Farrar W. L. Characterization of active and inactive forms of the JAK2 protein- tyrosine kinase produced via the baculovirus expression vector system.
J. Biol. Chem.
,
270
:
23084
-23089,  
1995
.
40
Zhou Y. J., Hanson E. P., Chen Y. Q., Magnuson K., Chen M., Swann P. G., Wange R. L., Changelian P. S., O’Shea J. J. Distinct tyrosine phosphorylation sites in JAK3 kinase domain positively and negatively regulate its enzymatic activity.
Proc. Natl. Acad. Sci. USA
,
94
:
13850
-13855,  
1997
.
41
Notarangelo L. D. Immunodeficiencies caused by genetic defects in protein kinases.
Curr. Opin. Immunol.
,
8
:
448
-453,  
1996
.
42
Lacronique V., Boureux A., Valle V. D., Poirel H., Quang C. T., Mauchauffe M., Berthou C., Lessard M., Berger R., Ghysdael J., Bernard O. A. A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia.
Science (Wash. DC)
,
278
:
1309
-1312,  
1997
.
43
Peeters P., Raynaud S. D., Cools J., Wlodarska I., Grosgeorge J., Philip P., Monpoux F., Van Rompaey L., Baens M., Van den Berghe H., Marynen P. Fusion of TEL, the ETS-variant gene 6 (ETV6), to the receptor- associated kinase JAK2 as a result of t(9;12) in a lymphoid and t(9;15;12) in a myeloid leukemia.
Blood
,
90
:
2535
-2540,  
1997
.
44
Migone T. S., Lin J. X., Cereseto A., Mulloy J. C., O’Shea J. J., Franchini G., Leonard W. J. Constitutively activated Jak-STAT pathway in T cells transformed with HTLV-I.
Science (Wash. DC)
,
269
:
79
-81,  
1995
.
45
Danial N. N., Pernis A., Rothman P. B. Jak-STAT signaling induced by the v-abl oncogene.
Science (Wash. DC)
,
269
:
1875
-1877,  
1995
.
46
Darnell J. E., Jr. STATs and gene regulation.
Science (Wash. DC)
,
277
:
1630
-1635,  
1997
.
47
Fu X. Y., Schindler C., Improta T., Aebersold R., Darnell J. E., Jr. The proteins of ISGF-3, the interferon α-induced transcriptional activator, define a gene family involved in signal transduction, Proc.
Natl. Acad. Sci. USA
,
89
:
7840
-7843,  
1992
.
48
Khan K. D., Shuai K., Lindwall G., Maher S. E., Darnell J. E., Jr., Bothwell A. L. Induction of the Ly-6A/E gene by interferon α/β and γ requires a DNA element to which a tyrosine-phosphorylated 91-kDa protein binds.
Proc. Natl. Acad. Sci. USA
,
90
:
6806
-6810,  
1993
.
49
Shuai K., Schindler C., Prezioso V. R., Darnell J. E., Jr. Activation of transcription by IFN-γ: tyrosine phosphorylation of a 91-kD DNA binding protein.
Science (Wash. DC)
,
258
:
1808
-1812,  
1992
.
50
Shuai K., Stark G. R., Kerr I. M., Darnell J. E., Jr. A single phosphotyrosine residue of Stat91 required for gene activation by interferon-γ.
Science (Wash. DC)
,
261
:
1744
-1746,  
1993
.
51
Zhong Z., Wen Z., Darnell J. E., Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6.
Science (Wash. DC)
,
264
:
95
-98,  
1994
.
52
Shuai K., Horvath C. M., Huang L. H., Qureshi S. A., Cowburn D., Darnell J. E., Jr. Interferon activation of the transcription factor Stat91 involves dimerization through SH2-phosphotyrosyl peptide interactions.
Cell
,
76
:
821
-828,  
1994
.
53
Kohlhuber F., Rogers N. C., Watling D., Feng J., Guschin D., Briscoe J., Witthuhn B. A., Kotenko S. V., Pestka S., Stark G. R., Ihle J. N., Kerr I. M. A JAK1/JAK2 chimera can sustain α and γ interferon responses.
Mol. Cell. Biol.
,
17
:
695
-706,  
1997
.
54
Kotenko S. V., Izotova L. S., Pollack B. P., Muthukumaran G., Paukku K., Silvennoinen O., Ihle J. N., Pestka S. Other kinases can substitute for Jak2 in signal transduction by interferon-γ.
J. Biol. Chem.
,
271
:
17174
-17182,  
1996
.
55
Bhattacharya S., Eckner R., Grossman S., Oldread E., Arany Z., D’Andrea A., Livingston D. M. Cooperation of Stat2 and p300/CBP in signalling induced by interferon- α.
Nature (Lond.)
,
383
:
344
-347,  
1996
.
56
Horvai A. E., Xu L., Korzus E., Brard G., Kalafus D., Mullen T. M., Rose D. W., Rosenfeld M. G., Glass C. K. Nuclear integration of JAK/STAT and Ras/AP-1 signaling by CBP and p300.
Proc. Natl. Acad. Sci. USA
,
94
:
1074
-1079,  
1997
.
57
Eilers A., Georgellis D., Klose B., Schindler C., Ziemiecki A., Harpur A. G. Differentiation-regulated serine phosphorylation of STAT1 promotes GAF activation in macrophages.
Mol. Cell. Biol.
,
15
:
3579
-3586,  
1995
.
58
Schindler C. STATs as activators of apoptosis.
Trends Cell Biol.
,
8
:
97
-98,  
1998
.
59
Mui A. L. The role of STATs in proliferation, differentiation, and apoptosis.
Cell Mol. Life Sci.
,
55
:
1547
-1458,  
1999
.
60
Chin Y. E., Kitagawa M., Su W. C., You Z. H., Iwamoto Y., Fu X. Y. Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF1/CIP1 mediated by STAT1.
Science (Wash. DC)
,
272
:
719
-722,  
1996
.
61
Mui A. L., Wakao H., Kinoshita T., Kitamura T., Miyajima A. Suppression of interleukin-3-induced gene expression by a C-terminal truncated Stat5: role of Stat5 in proliferation.
EMBO J.
,
15
:
2425
-2433,  
1996
.
62
O’Farrell A. M., Liu Y., Moore K. W., Mui A. L. IL-10 inhibits macrophage activation and proliferation by distinct signaling mechanisms: evidence for Stat3-dependent and -independent pathways.
EMBO J.
,
17
:
1006
-1018,  
1998
.
63
Chaturvedi P., Reddy M. V., Reddy E. P. Src kinases and not JAKs activate STATs during IL-3 induced myeloid cell proliferation.
Oncogene
,
16
:
1749
-1758,  
1998
.
64
Kumar A., Commane M., Flickinger T. W., Horvath C. M., Stark G. R. Defective TNF-α-induced apoptosis in STAT1-null cells due to low constitutive levels of caspases.
Science (Wash. DC)
,
278
:
1630
-1632,  
1997
.
65
Zamorano J., Keegan A. D. Regulation of apoptosis by tyrosine-containing domains of IL-4R α: Y497 and Y713, but not the STAT6-docking tyrosines, signal protection from apoptosis.
J. Immunol.
,
161
:
859
-867,  
1998
.
66
Fukada T., Hibi M., Yamanaka Y., Takahashi-Tezuka M., Fujitani Y., Yamaguchi T., Nakajima K., Hirano T. Two signals are necessary for cell proliferation induced by a cytokine receptor gp130: involvement of STAT3 in anti-apoptosis.
Immunity
,
5
:
449
-460,  
1996
.
67
Xu X., Fu X. Y., Plate J., Chong A. S. IFN-γ induces cell growth inhibition by Fas-mediated apoptosis: requirement of STAT1 protein for up-regulation of Fas and FasL expression.
Cancer Res.
,
58
:
2832
-2837,  
1998
.
68
Kim T. K., Maniatis T. Regulation of interferon-γ-activated STAT1 by the ubiquitin- proteasome pathway.
Science (Wash. DC)
,
273
:
1717
-1719,  
1996
.
69
An W. G., Hwang S. G., Trepel J. B., Blagosklonny M. V. Protease inhibitor-induced apoptosis: accumulation of wt p53, p21WAF1/CIP1, and induction of apoptosis are independent markers of proteasome inhibition.
Leukemia (Baltimore)
,
14
:
1276
-1283,  
2000
.
70
Starr R., Hilton D. J. Negative regulation of the Jak/Stat pathway.
Bioassays
,
21
:
47
-52,  
1999
.
71
Hilton D. J. Negative regulators of cytokine signal transduction.
Cell Mol. Life Sci.
,
55
:
1568
-1577,  
1999
.
72
Jiao H., Yang W., Berrada K., Tabrizi M., Shultz L., Yi T. Macrophages from motheaten and viable motheaten mutant mice show increased proliferative responses to GM-CSF: detection of potential HCP substrates in GM-CSF signal transduction.
Exp. Hematol.
,
25
:
592
-600,  
1997
.
73
Alexander W. S., Starr R., Metcalf D., Nicholson S. E., Farley A., Elefanty A. G., Brysha M., Kile B. T., Richardson R., Baca M., Zhang J. G., Willson T. A., Viney E. M., Sprigg N. S., Rakar S., Corbin J., Mifsud S., DiRago L., Cary D., Nicola N. A., Hilton D. J. Suppressors of cytokine signaling (SOCS): negative regulators of signal transduction.
J. Leukoc. Biol.
,
66
:
588
-592,  
1999
.
74
Nicholson S. E., Willson T. A., Farley A., Starr R., Zhang J. G., Baca M., Alexander W. S., Metcalf D., Hilton D. J., Nicola N. A. Mutational analyses of the SOCS proteins suggest a dual domain requirement but distinct mechanisms for inhibition of LIF and IL-6 signal transduction.
EMBO J.
,
18
:
375
-385,  
1999
.
75
Adams T. E., Hansen J. A., Starr R., Nicola N. A., Hilton D. J., Billestrup N. Growth hormone preferentially induces the rapid, transient expression of SOCS-3, a novel inhibitor of cytokine receptor signaling.
J. Biol. Chem.
,
273
:
1285
-1287,  
1998
.
76
Song M. M., Shuai K. The suppressor of cytokine signaling (SOCS) 1 and SOCS3 but not SOCS2 proteins inhibit interferon-mediated antiviral and antiproliferative activities.
J. Biol. Chem.
,
273
:
35056
-35062,  
1998
.
77
Schultheis B., Carapeti-Marootian M., Hochhaus A., Weisser A., Goldman J. M., Melo J. V. Overexpression of SOCS-2 in advanced stages of chronic myeloid leukemia: possible inadequacy of a negative feedback mechanism.
Blood
,
99
:
1766
-1775,  
2002
.
78
Porter A. C., Vaillancourt R. R. Tyrosine kinase receptor-activated signal transduction pathways which lead to oncogenesis.
Oncogene
,
17
:
1343
-1352,  
1998
.
79
Drexler H. G. Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells.
Leukemia (Baltimore)
,
10
:
588
-599,  
1996
.
80
Blume-Jensen P., Hunter T. Oncogenic kinase signalling.
Nature (Lond.)
,
411
:
355
-365,  
2001
.
81
Weiss F. U., Daub H., Ullrich A. Novel mechanisms of RTK signal generation.
Curr. Opin. Genet. Dev.
,
7
:
80
-86,  
1997
.
82
Weiss A., Schlessinger J. Switching signals on or off by receptor dimerization.
Cell
,
94
:
277
-280,  
1998
.
83
Sherr C. J. Colony-stimulating factor-1 receptor.
Blood
,
75
:
1
-12,  
1990
.
84
Matthews W., Jordan C. T., Wiegand G. W., Pardoll D., Lemischka I. R. A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell-enriched populations.
Cell
,
65
:
1143
-1152,  
1991
.
85
Tse K. F., Mukherjee G., Small D. Constitutive activation of FLT3 stimulates multiple intracellular signal transducers and results in transformation.
Leukemia (Baltimore)
,
14
:
1766
-1776,  
2000
.
86
Gilliland D. G., Griffin J. D. The roles of FLT3 in hematopoiesis and leukemia.
Blood
,
100
:
1532
-1542,  
2002
.
87
Thiede C., Steudel C., Mohr B., Schaich M., Schakel U., Platzbecker U., Wermke M., Bornhauser M., Ritter M., Neubauer A., Ehninger G., Illmer T. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis.
Blood
,
99
:
4326
-4335,  
2002
.
88
Gilliland D. G., Griffin J. D. Role of FLT3 in leukemia.
Curr. Opin. Hematol.
,
9
:
274
-281,  
2002
.
89
Kiyoi H., Ohno R., Ueda R., Saito H., Naoe T. Mechanism of constitutive activation of FLT3 with internal tandem duplication in the juxtamembrane domain.
Oncogene
,
21
:
2555
-2563,  
2002
.
90
Kelly L. M., Liu Q., Kutok J. L., Williams I. R., Boulton C. L., Gilliland D. G. FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model.
Blood
,
99
:
310
-318,  
2002
.
91
Yamamoto Y., Kiyoi H., Nakano Y., Suzuki R., Kodera Y., Miyawaki S., Asou N., Kuriyama K., Yagasaki F., Shimazaki C., Akiyama H., Saito K., Nishimura M., Motoji T., Shinagawa K., Takeshita A., Saito H., Ueda R., Ohno R., Naoe T. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies.
Blood
,
97
:
2434
-2439,  
2001
.
92
Levis M., Tse K. F., Smith B. D., Garrett E., Small D. A FLT3 tyrosine kinase inhibitor is selectively cytotoxic to acute myeloid leukemia blasts harboring FLT3 internal tandem duplication mutations.
Blood
,
98
:
885
-887,  
2001
.
93
Levis M., Allebach J., Tse K. F., Zheng R., Baldwin B. R., Smith B. D., Jones-Bolin S., Ruggeri B., Dionne C., Small D. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo.
Blood
,
99
:
3885
-3891,  
2002
.
94
Schwartzberg P. L. The many faces of Src: multiple functions of a prototypical tyrosine kinase.
Oncogene
,
17
:
1463
-1468,  
1998
.
95
Lee J. T., Jr., McCubrey J. A. The Raf/MEK/ERK signal transduction cascade as a target for chemotherapeutic intervention in leukemia.
Leukemia (Baltimore)
,
16
:
486
-507,  
2002
.
96
Beaupre D. M., Kurzrock R. RAS and leukemia: from basic mechanisms to gene-directed therapy.
J. Clin. Oncol.
,
17
:
1071
-1079,  
1999
.
97
Campbell S. L., Khosravi-Far R., Rossman K. L., Clark G. J., Der C. J. Increasing complexity of Ras signaling.
Oncogene
,
17
:
1395
-1413,  
1998
.
98
Prendergast G. C., Gibbs J. B. Ras regulatory interactions: novel targets for anti-cancer intervention?.
Bioessays
,
16
:
187
-191,  
1994
.
99
Crews C. M., Erikson R. L. Extracellular signals and reversible protein phosphorylation: what to Mek of it all.
Cell
,
74
:
215
-217,  
1993
.
100
Crews C. M., Alessandrini A., Erikson R. L. Erks: their fifteen minutes has arrived.
Cell Growth Differ.
,
3
:
135
-142,  
1992
.
101
Dhanasekaran N., Premkumar Reddy E. Signaling by dual specificity kinases.
Oncogene
,
17
:
1447
-1455,  
1998
.
102
Marais R., Wynne J., Treisman R. The SRF accessory protein Elk-1 contains a growth factor-regulated transcriptional activation domain.
Cell
,
73
:
381
-393,  
1993
.
103
Boulton T. G., Nye S. H., Robbins D. J., Ip N. Y., Radziejewska E., Morgenbesser S. D., DePinho R. A., Panayotatos N., Cobb M. H., Yancopoulos G. D. ERKs: a family of protein-serine/threonine kinases that are activated and tyrosine phosphorylated in response to insulin and NGF.
Cell
,
65
:
663
-675,  
1991
.
104
Nebreda A. R., Gavin A. C. Perspectives: signal transduction. Cell survival demands some Rsk.
Science (Wash. DC)
,
286
:
1309
-1310,  
1999
.
105
Bonni A., Brunet A., West A. E., Datta S. R., Takasu M. A., Greenberg M. E. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms.
Science (Wash. DC)
,
286
:
1358
-1362,  
1999
.
106
Kerkhoff E., Rapp U. R. Cell cycle targets of Ras/Raf signalling.
Oncogene
,
17
:
1457
-1462,  
1998
.
107
Sherr C. J. Growth factor-regulated G1 cyclins.
Stem Cells
,
12
:
47
-57,  
1994
.
108
Mayol X., Grana X. pRB, p107 and p130 as transcriptional regulators: role in cell growth and differentiation.
Prog. Cell Cycle Res.
,
3
:
157
-169,  
1997
.
109
Dimri G. P., Nakanishi M., Desprez P. Y., Smith J. R., Campisi J. Inhibition of E2F activity by the cyclin-dependent protein kinase inhibitor p21 in cells expressing or lacking a functional retinoblastoma protein.
Mol. Cell. Biol.
,
16
:
2987
-2997,  
1996
.
110
Steinman R. A. Cell cycle regulators and hematopoiesis.
Oncogene
,
21
:
3403
-3413,  
2002
.
111
Jiang Y., Gram H., Zhao M., New L., Gu J., Feng L., Di Padova F., Ulevitch R. J., Han J. Characterization of the structure and function of the fourth member of p38 group mitogen-activated protein kinases, p38δ.
J. Biol. Chem.
,
272
:
30122
-30128,  
1997
.
112
Lechner C., Zahalka M. A., Giot J. F., Moller N. P., Ullrich A. ERK6, a mitogen-activated protein kinase involved in C2C12 myoblast differentiation.
Proc. Natl. Acad. Sci. USA
,
93
:
4355
-4359,  
1996
.
113
Enslen H., Brancho D. M., Davis R. J. Molecular determinants that mediate selective activation of p38 MAP kinase isoforms.
EMBO J.
,
19
:
1301
-1311,  
2000
.
114
Uddin S., Majchrzak B., Woodson J., Arunkumar P., Alsayed Y., Pine R., Young P. R., Fish E. N., Platanias L. C. Activation of the p38 mitogen-activated protein kinase by type I interferons.
J. Biol. Chem.
,
274
:
30127
-30131,  
1999
.
115
Mayer I. A., Verma A., Grumbach I. M., Uddin S., Lekmine F., Ravandi F., Majchrzak B., Fujita S., Fish E. N., Platanias L. C. The p38 MAPK pathway mediates the growth inhibitory effects of interferon-α in BCR-ABL-expressing cells.
J. Biol. Chem.
,
276
:
28570
-28577,  
2001
.
116
Verma A., Deb D. K., Sassano A., Uddin S., Varga J., Wickrema A., Platanias L. C. Activation of the p38 mitogen-activated protein kinase mediates the suppressive effects of type I interferons and transforming growth factor-β on normal eematopoiesis.
J. Biol. Chem.
,
277
:
7726
-7735,  
2002
.
117
Ichijo H. From receptors to stress-activated MAP kinases.
Oncogene
,
18
:
6087
-6093,  
1999
.
118
Fuchs S. Y., Adler V., Pincus M. R., Ronai Z. MEKK1/JNK signaling stabilizes and activates p53.
Proc. Natl. Acad. Sci. USA
,
95
:
10541
-10546,  
1998
.
119
Keyse S. M. Protein phosphatases and the regulation of mitogen-activated protein kinase signalling.
Curr. Opin. Cell Biol.
,
12
:
186
-192,  
2000
.
120
Camps M., Nichols A., Gillieron C., Antonsson B., Muda M., Chabert C., Boschert U., Arkinstall S. Catalytic activation of the phosphatase MKP-3 by ERK2 mitogen-activated protein kinase.
Science (Wash. DC)
,
280
:
1262
-1265,  
1998
.
121
Camps M., Nichols A., Arkinstall S. Dual specificity phosphatases: a gene family for control of MAP kinase function.
FASEB J.
,
14
:
6
-16,  
2000
.
122
Chung H., Brautigan D. L. Protein phosphatase 2A suppresses MAP kinase signalling and ectopic protein expression.
Cell Signalling
,
11
:
575
-580,  
1999
.
123
Wassarman D. A., Solomon N. M., Chang H. C., Karim F. D., Therrien M., Rubin G. M. Protein phosphatase 2A positively and negatively regulates Ras1- mediated photoreceptor development in Drosophila.
Genes Dev.
,
10
:
272
-278,  
1996
.
124
Klippel A., Escobedo J. A., Fantl W. J., Williams L. T. The C-terminal SH2 domain of p85 accounts for the high affinity and specificity of the binding of phosphatidylinositol 3-kinase to phosphorylated platelet-derived growth factor β receptor.
Mol. Cell. Biol.
,
12
:
1451
-1459,  
1992
.
125
Hiles I. D., Otsu M., Volinia S., Fry M. J., Gout I., Dhand R., Panayotou G., Ruiz-Larrea F., Thompson A., Totty N. F., et al Phosphatidylinositol 3-kinase: structure and expression of the 110 kd catalytic subunit.
Cell
,
70
:
419
-429,  
1992
.
126
Shoelson S. E., Sivaraja M., Williams K. P., Hu P., Schlessinger J., Weiss M. A. Specific phosphopeptide binding regulates a conformational change in the PI 3-kinase SH2 domain associated with enzyme activation.
EMBO J.
,
12
:
795
-802,  
1993
.
127
Anderson K. E., Coadwell J., Stephens L. R., Hawkins P. T. Translocation of PDK-1 to the plasma membrane is important in allowing PDK-1 to activate protein kinase B.
Curr. Biol.
,
8
:
684
-691,  
1998
.
128
Scheid M. P., Duronio V. Dissociation of cytokine-induced phosphorylation of Bad and activation of PKB/akt: involvement of MEK upstream of Bad phosphorylation.
Proc. Natl. Acad. Sci. USA
,
95
:
7439
-7444,  
1998
.
129
del Peso L., Gonzalez-Garcia M., Page C., Herrera R., Nunez G. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt.
Science (Wash. DC)
,
278
:
687
-689,  
1997
.
130
Datta S. R., Dudek H., Tao X., Masters S., Fu H., Gotoh Y., Greenberg M. E. Akt phosphorylation of BAD couples survival signals to the cell- intrinsic death machinery.
Cell
,
91
:
231
-241,  
1997
.
131
Khwaja A. Akt is more than just a Bad kinase.
Nature (Lond.)
,
401
:
33
-34,  
1999
.
132
Dong F., Larner A. C. Activation of Akt kinase by granulocyte colony-stimulating factor (G-CSF): evidence for the role of a tyrosine kinase activity distinct from the Janus kinases.
Blood
,
95
:
1656
-1662,  
2000
.
133
Aoki M., Batista O., Bellacosa A., Tsichlis P., Vogt P. K. The akt kinase: molecular determinants of oncogenicity.
Proc. Natl. Acad. Sci. USA
,
95
:
14950
-14955,  
1998
.
134
Sellers W. R., Fisher D. E. Apoptosis and cancer drug targeting.
J. Clin. Investig.
,
104
:
1655
-1661,  
1999
.
135
Blagosklonny M. V., Chuman Y., Bergan R. C., Fojo T. Mitogen-activated protein kinase pathway is dispensable for microtubule-active drug-induced Raf-1/Bcl-2 phosphorylation and apoptosis in leukemia cells.
Leukemia (Baltimore)
,
13
:
1028
-1036,  
1999
.
136
Pahl H. L. Activators and target genes of Rel/NF-κB transcription factors.
Oncogene
,
18
:
6853
-6866,  
1999
.
137
Li J., Yen C., Liaw D., Podsypanina K., Bose S., Wang S. I., Puc J., Miliaresis C., Rodgers L., McCombie R., Bigner S. H., Giovanella B. C., Ittmann M., Tycko B., Hibshoosh H., Wigler M. H., Parsons R. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Science (Wash. DC)
,
275
:
1943
-1947,  
1997
.
138
Stambolic V., Suzuki A., de la Pompa J. L., Brothers G. M., Mirtsos C., Sasaki T., Ruland J., Penninger J. M., Siderovski D. P., Mak T. W. Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN.
Cell
,
95
:
29
-39,  
1998
.
139
Maehama T., Dixon J. E. The tumor suppressor. PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3, 4, 5-trisphosphate.
J. Biol. Chem.
,
273
:
13375
-13378,  
1998
.
140
Myers M. P., Pass I., Batty I. H., Van der Kaay J., Stolarov J. P., Hemmings B. A., Wigler M. H., Downes C. P., Tonks N. K. The lipid phosphatase activity of PTEN is critical for its tumor supressor function.
Proc. Natl. Acad. Sci. USA
,
95
:
13513
-13518,  
1998
.
141
Podsypanina K., Ellenson L. H., Nemes A., Gu J., Tamura M., Yamada K. M., Cordon-Cardo C., Catoretti G., Fisher P. E., Parsons R. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems.
Proc. Natl. Acad. Sci. USA
,
96
:
1563
-1568,  
1999
.
142
Hu Z. B., Minden M. D., McCulloch E. A. Phosphorylation of BCL-2 after exposure of human leukemic cells to retinoic acid.
Blood
,
92
:
1768
-1775,  
1998
.
143
Haldar S., Basu A., Croce C. M. Bcl2 is the guardian of microtubule integrity.
Cancer Res.
,
57
:
229
-233,  
1997
.
144
Asaoka Y., Nakamura S., Yoshida K., Nishizuka Y. Protein kinase C, calcium and phospholipid degradation.
Trends Biochem. Sci.
,
17
:
414
-417,  
1992
.
145
Liou H. C., Baltimore D. Regulation of the NF-κ B/rel transcription factor and I κ B inhibitor system.
Curr. Opin. Cell Biol.
,
5
:
477
-487,  
1993
.
146
Nishizuka Y. Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C.
Science (Wash. DC)
,
258
:
607
-614,  
1992
.
147
Kovanen P. E., Junttila I., Takaluoma K., Saharinen P., Valmu L., Li W., Silvennoinen O. Regulation of Jak2 tyrosine kinase by protein kinase C during macrophage differentiation of IL-3-dependent myeloid progenitor cells.
Blood
,
95
:
1626
-1632,  
2000
.
148
Kolch W., Heidecker G., Kochs G., Hummel R., Vahidi H., Mischak H., Finkenzeller G., Marme D., Rapp U. R. Protein kinase C α activates RAF-1 by direct phosphorylation.
Nature (Lond.)
,
364
:
249
-252,  
1993
.
149
Seynaeve C. M., Kazanietz M. G., Blumberg P. M., Sausville E. A., Worland P. J. Differential inhibition of protein kinase C isozymes by UCN-01, a staurosporine analogue.
Mol. Pharmacol.
,
45
:
1207
-1214,  
1994
.
150
Thavasu P., Propper D., McDonald A., Dobbs N., Ganesan T., Talbot D., Braybrook J., Caponigro F., Hutchison C., Twelves C., Man A., Fabbro D., Harris A., Balkwill F. The protein kinase C inhibitor CGP41251 suppresses cytokine release and extracellular signal-regulated kinase 2 expression in cancer patients.
Cancer Res.
,
59
:
3980
-3984,  
1999
.
151
King W. G., Mattaliano M. D., Chan T. O., Tsichlis P. N., Brugge J. S. Phosphatidylinositol 3-kinase is required for integrin-stimulated AKT and Raf-1/mitogen-activated protein kinase pathway activation.
Mol. Cell. Biol.
,
17
:
4406
-4418,  
1997
.
152
Wennstrom S., Downward J. Role of phosphoinositide 3kinase in activation of ras and mitogen- activated protein kinase by epidermal growth factor.
Mol. Cell. Biol.
,
19
:
4279
-4288,  
1999
.
153
McCubrey J. A., Lee J. T., Steelman L. S., Blalock W. L., Moye P. W., Chang F., Pearce M., Shelton J. G., White M. K., Franklin R. A., Pohnert S. C. Interactions between the PI3K and Raf signaling pathways can result in the transformation of hematopoietic cells.
Cancer Detect. Prev.
,
25
:
375
-393,  
2001
.
154
Platanias L. C., Fish E. N. Signaling pathways activated by interferons.
Exp. Hematol.
,
27
:
1583
-1592,  
1999
.
155
David M., Petricoin E., 3rd, Benjamin C., Pine R., Weber M. J., Larner A. C. Requirement for MAP kinase (ERK2) activity in interferon α- and interferon β-stimulated gene expression through STAT proteins.
Science (Wash. DC)
,
269
:
1721
-1723,  
1995
.
156
Dickens M., Rogers J. S., Cavanagh J., Raitano A., Xia Z., Halpern J. R., Greenberg M. E., Sawyers C. L., Davis R. J. A cytoplasmic inhibitor of the JNK signal transduction pathway.
Science (Wash. DC)
,
277
:
693
-696,  
1997
.
157
Sawyers C. L., McLaughlin J., Witte O. N. Genetic requirement for Ras in the transformation of fibroblasts and hematopoietic cells by the Bcr-Abl oncogene.
J. Exp. Med.
,
181
:
307
-313,  
1995
.
158
Skorski T., Kanakaraj P., Nieborowska-Skorska M., Ratajczak M. Z., Wen S. C., Zon G., Gewirtz A. M., Perussia B., Calabretta B. Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells.
Blood
,
86
:
726
-736,  
1995
.
159
Oda T., Heaney C., Hagopian J. R., Okuda K., Griffin J. D., Druker B. J. Crkl is the major tyrosine-phosphorylated protein in neutrophils from patients with chronic myelogenous leukemia.
J. Biol. Chem.
,
269
:
22925
-22928,  
1994
.
160
Ilaria R. L., Jr., Van Etten R. A. P210 and P190(BCR/ABL) induce the tyrosine phosphorylation and DNA binding activity of multiple specific STAT family members.
J. Biol. Chem.
,
271
:
31704
-31710,  
1996
.
161
Pendergast A. M., Quilliam L. A., Cripe L. D., Bassing C. H., Dai Z., Li N., Batzer A., Rabun K. M., Der C. J., Schlessinger J., et al BCR-ABL-induced oncogenesis is mediated by direct interaction with the SH2 domain of the GRB-2 adaptor protein.
Cell
,
75
:
175
-185,  
1993
.
162
Tauchi T., Miyazawa K., Feng G. S., Broxmeyer H. E., Toyama K. A coiled-coil tetramerization domain of BCR-ABL is essential for the interactions of SH2-containing signal transduction molecules.
J. Biol. Chem.
,
272
:
1389
-1394,  
1997
.
163
Salgia R., Sattler M., Pisick E., Li J. L., Griffin J. D. p210BCR/ABL induces formation of complexes containing focal adhesion proteins and the protooncogene product p120c-Cbl.
Exp. Hematol.
,
24
:
310
-313,  
1996
.
164
Levitzki A., Gazit A. Tyrosine kinase inhibition: an approach to drug development.
Science (Wash. DC)
,
267
:
1782
-1788,  
1995
.
165
Levitzki A. Tyrphostins: tyrosine kinase blockers as novel antiproliferative agents and directors of signal transduction.
FASEB J.
,
6
:
3275
-3282,  
1992
.
166
Sawyers C. L., Druker B. Tyrosine kinase inhibitors in chronic myeloid leukemia.
Cancer J. Sci. Am.
,
5
:
63
-69,  
1998
.
167
Druker B. J., Talpaz M., Resta D. J., Peng B., Buchdunger E., Ford J. M., Lydon N. B., Kantarjian H., Capdeville R., Ohno-Jones S., Sawyers C. L. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N. Engl. J. Med.
,
344
:
1031
-1037,  
2001
.
168
Druker B. J., Sawyers C. L., Kantarjian H., Resta D. J., Reese S. F., Ford J. M., Capdeville R., Talpaz M. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome.
N. Engl. J. Med.
,
344
:
1038
-1042,  
2001
.
169
Kantarjian H., Sawyers C., Hochhaus A., Guilhot F., Schiffer C., Gambacorti-Passerini C., Niederwieser D., Resta D., Capdeville R., Zoellner U., Talpaz M., Druker B. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia.
N. Engl. J. Med.
,
346
:
645
-652,  
2002
.
170
Talpaz M., Silver R. T., Druker B. J., Goldman J. M., Gambacorti-Passerini C., Guilhot F., Schiffer C. A., Fischer T., Deininger M. W., Lennard A. L., Hochhaus A., Ottmann O. G., Gratwohl A., Baccarani M., Stone R., Tura S., Mahon F. X., Fernandes-Reese S., Gathmann I., Capdeville R., Kantarjian H. M., Sawyers C. L. Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study.
Blood
,
99
:
1928
-1937,  
2002
.
171
Dierov J., Xu Q., Dierova R., Carroll M. TEL/platelet-derived growth factor receptor β activates phosphatidylinositol 3 (PI3) kinase and requires PI3 kinase to regulate the cell cycle.
Blood
,
99
:
1758
-1765,  
2002
.
172
Golub T. R., Barker G. F., Lovett M., Gilliland D. G. Fusion of PDGF receptor β to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation.
Cell
,
77
:
307
-316,  
1994
.
173
Magnusson M. K., Meade K. E., Brown K. E., Arthur D. C., Krueger L. A., Barrett A. J., Dunbar C. E. Rabaptin-5 is a novel fusion partner to platelet-derived growth factor β receptor in chronic myelomonocytic leukemia.
Blood
,
98
:
2518
-2525,  
2001
.
174
Magnusson M. K., Meade K. E., Nakamura R., Barrett J., Dunbar C. E. Activity of STI571 in chronic myelomonocytic leukemia with a platelet- derived growth factor β receptor fusion oncogene.
Blood
,
100
:
1088
-1091,  
2002
.
175
Mason D. Y., Bastard C., Rimokh R., Dastugue N., Huret J. L., Kristoffersson U., Magaud J. P., Nezelof C., Tilly H., Vannier J. P., et al CD30-positive large cell lymphomas (’Ki-1 lymphoma’) are associated with a chromosomal translocation involving 5q35, Br.
J. Haematol.
,
74
:
161
-168,  
1990
.
176
Wellmann A., Otsuki T., Vogelbruch M., Clark H. M., Jaffe E. S., Raffeld M. Analysis of the t(2;5)(p23;q35) translocation by reverse transcription- polymerase chain reaction in CD30+ anaplastic large-cell lymphomas, in other non-Hodgkin’s lymphomas of T-cell phenotype, and in Hodgkin’s disease.
Blood
,
86
:
2321
-2328,  
1995
.
177
Bischof D., Pulford K., Mason D. Y., Morris S. W. Role of the nucleophosmin (NPM) portion of the non-Hodgkin’s lymphoma- associated NPM-anaplastic lymphoma kinase fusion protein in oncogenesis.
Mol. Cell. Biol.
,
17
:
2312
-3325,  
1997
.
178
Fujimoto J., Shiota M., Iwahara T., Seki N., Satoh H., Mori S., Yamamoto T. Characterization of the transforming activity of p80, a hyperphosphorylated protein in a Ki-1 lymphoma cell line with chromosomal translocation t(2;5).
Proc. Natl. Acad. Sci. USA
,
93
:
4181
-4186,  
1996
.
179
Bai R. Y., Dieter P., Peschel C., Morris S. W., Duyster J. Nucleophosmin-anaplastic lymphoma kinase of large-cell anaplastic lymphoma is a constitutively active tyrosine kinase that utilizes phospholipase C-γ to mediate its mitogenicity.
Mol. Cell. Biol.
,
18
:
6951
-6961,  
1998
.
180
Bai R. Y., Ouyang T., Miething C., Morris S. W., Peschel C., Duyster J. Nucleophosmin-anaplastic lymphoma kinase associated with anaplastic large-cell lymphoma activates the phosphatidylinositol 3-kinase/Akt antiapoptotic signaling pathway.
Blood
,
96
:
4319
-4327,  
2000
.
181
Nieborowska-Skorska M., Slupianek A., Xue L., Zhang Q., Raghunath P. N., Hoser G., Wasik M. A., Morris S. W., Skorski T. Role of signal transducer and activator of transcription 5 in nucleophosmin/anaplastic lymphoma kinase-mediated malignant transformation of lymphoid cells.
Cancer Res.
,
61
:
6517
-6523,  
2001
.
182
Slupianek A., Nieborowska-Skorska M., Hoser G., Morrione A., Majewski M., Xue L., Morris S. W., Wasik M. A., Skorski T. Role of phosphatidylinositol 3-kinase-Akt pathway in nucleophosmin/anaplastic lymphoma kinase-mediated lymphomagenesis.
Cancer Res.
,
61
:
2194
-2199,  
2001
.
183
Stein H., Foss H. D., Durkop H., Marafioti T., Delsol G., Pulford K., Pileri S., Falini B. CD30(+) anaplastic large cell lymphoma: a review of its histopathologic, genetic, and clinical features.
Blood
,
96
:
3681
-3695,  
2000
.
184
Sachs L. The control of hematopoiesis and leukemia: from basic biology to the clinic.
Proc. Natl. Acad. Sci. USA
,
93
:
4742
-4749,  
1996
.
185
Savino R., Ciapponi L., Lahm A., Demartis A., Cabibbo A., Toniatti C., Delmastro P., Altamura S., Ciliberto G. Rational design of a receptor super-antagonist of human interleukin-6.
EMBO J.
,
13
:
5863
-5870,  
1994
.
186
Klein B., Wijdenes J., Zhang X. G., Jourdan M., Boiron J. M., Brochier J., Liautard J., Merlin M., Clement C., Morel-Fournier B., et al Murine anti-interleukin-6 monoclonal antibody therapy for a patient with plasma cell leukemia.
Blood
,
78
:
1198
-1204,  
1991
.
187
Tse K. F., Novelli E., Civin C. I., Bohmer F. D., Small D. Inhibition of FLT3-mediated transformation by use of a tyrosine kinase inhibitor.
Leukemia (Baltimore)
,
15
:
1001
-1010,  
2001
.
188
Kottaridis P. D., Gale R. E., Frew M. E., Harrison G., Langabeer S. E., Belton A. A., Walker H., Wheatley K., Bowen D. T., Burnett A. K., Goldstone A. H., Linch D. C. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials.
Blood
,
98
:
1752
-1759,  
2001
.
189
Abu-Duhier F. M., Goodeve A. C., Wilson G. A., Gari M. A., Peake I. R., Rees D. C., Vandenberghe E. A., Winship P. R., Reilly J. T. FLT3 internal tandem duplication mutations in adult acute myeloid leukaemia define a high-risk group.
Br. J. Haematol.
,
111
:
190
-195,  
2000
.
190
Meydan N., Grunberger T., Dadi H., Shahar M., Arpaia E., Lapidot Z., Leeder J. S., Freedman M., Cohen A., Gazit A., Levitzki A., Roifman C. M. Inhibition of acute lymphoblastic leukaemia by a Jak-2 inhibitor.
Nature (Lond.)
,
379
:
645
-648,  
1996
.
191
Eriksen K. W., Kaltoft K., Mikkelsen G., Nielsen M., Zhang Q., Geisler C., Nissen M. H., Ropke C., Wasik M. A., Odum N. Constitutive STAT3-activation in Sezary syndrome: tyrphostin AG490 inhibits STAT3-activation, interleukin-2 receptor expression and growth of leukemic Sezary cells.
Leukemia (Baltimore)
,
15
:
787
-793,  
2001
.
192
De Vos J., Jourdan M., Tarte K., Jasmin C., Klein B. JAK2 tyrosine kinase inhibitor tyrphostin AG490 downregulates the mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription (STAT) pathways and induces apoptosis in myeloma cells.
Br. J. Haematol.
,
109
:
823
-828,  
2000
.
193
Levitt M. L., Koty P. P. Tyrosine kinase inhibitors in preclinical development.
Investig. New Drugs
,
17
:
213
-226,  
1999
.
194
Klohs W. D., Fry D. W., Kraker A. J. Inhibitors of tyrosine kinase.
Curr. Opin. Oncol.
,
9
:
562
-568,  
1997
.
195
Marra F., Choudhury G. G., Abboud H. E. Interferon-γ-mediated activation of STAT1α regulates growth factor-induced mitogenesis.
J. Clin. Investig.
,
98
:
1218
-1230,  
1996
.
196
Turkson J., Bowman T., Garcia R., Caldenhoven E., De Groot R. P., Jove R. Stat3 activation by Src induces specific gene regulation and is required for cell transformation.
Mol. Cell. Biol.
,
18
:
2545
-2552,  
1998
.
197
Sillaber C., Gesbert F., Frank D. A., Sattler M., Griffin J. D. STAT5 activation contributes to growth and viability in Bcr/Abl-transformed cells.
Blood
,
95
:
2118
-2125,  
2000
.
198
Frank D. A., Mahajan S., Ritz J. Fludarabine-induced immunosuppression is associated with inhibition of STAT1 signaling.
Nat. Med.
,
5
:
444
-447,  
1999
.
199
Minucci S., Nervi C., Lo Coco F., Pelicci P. G. Histone deacetylases: a common molecular target for differentiation treatment of acute myeloid leukemias?.
Oncogene
,
20
:
3110
-3115,  
2001
.
200
Amann J. M., Nip J., Strom D. K., Lutterbach B., Harada H., Lenny N., Downing J. R., Meyers S., Hiebert S. W. ETO, a target of t(8;21) in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain.
Mol. Cell. Biol.
,
21
:
6470
-6483,  
2001
.
201
Guidez F., Zelent A. Role of nuclear receptor corepressors in leukemogenesis.
Curr. Top. Microbiol. Immunol.
,
254
:
165
-185,  
2001
.
202
Chakrabarti S. R., Sood R., Nandi S., Nucifora G. Posttranslational modification of TEL and TEL/AML1 by SUMO-1 and cell-cycle-dependent assembly into nuclear bodies.
Proc. Natl. Acad. Sci. USA
,
97
:
13281
-13285,  
2000
.
203
Wang J., Saunthararajah Y., Redner R. L., Liu J. M. Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukemia cells.
Cancer Res.
,
59
:
2766
-2769,  
1999
.
204
Silverman L. R., Demakos E. P., Peterson B. L., Kornblith A. B., Holland J. C., Odchimar-Reissig R., Stone R. M., Nelson D., Powell B. L., DeCastro C. M., Ellerton J., Larson R. A., Schiffer C. A., Holland J. F. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B.
J. Clin. Oncol.
,
20
:
2429
-2440,  
2002
.
205
Dyck J. A., Maul G. G., Miller W. H., Jr., Chen J. D., Kakizuka A., Evans R. M. A novel macromolecular structure is a target of the promyelocyte- retinoic acid receptor oncoprotein.
Cell
,
76
:
333
-343,  
1994
.
206
Weis K., Rambaud S., Lavau C., Jansen J., Carvalho T., Carmo-Fonseca M., Lamond A., Dejean A. Retinoic acid regulates aberrant nuclear localization of PML-RAR α in acute promyelocytic leukemia cells.
Cell
,
76
:
345
-356,  
1994
.
207
Lin R. J., Nagy L., Inoue S., Shao W., Miller W. H., Jr., Evans R. M. Role of the histone deacetylase complex in acute promyelocytic leukaemia.
Nature (Lond.)
,
391
:
811
-814,  
1998
.
208
Guidez F., Ivins S., Zhu J., Soderstrom M., Waxman S., Zelent A. Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARα underlie molecular pathogenesis and treatment of acute promyelocytic leukemia.
Blood
,
91
:
2634
-2642,  
1998
.
209
Miller W. H., Jr., Waxman S. Differentiation induction as a treatment for hematologic malignancies.
Oncogene
,
21
:
3496
-3506,  
2002
.
210
Jansen J. H., de Ridder M. C., Geertsma W. M., Erpelinck C. A., van Lom K., Smit E. M., Slater R., vd Reijden B. A., de Greef G. E., Sonneveld P., Lowenberg B. Complete remission of t(11;17) positive acute promyelocytic leukemia induced by all-trans retinoic acid and granulocyte colony-stimulating factor.
Blood
,
94
:
39
-45,  
1999
.
211
Gianni M., Terao M., Fortino I., LiCalzi M., Viggiano V., Barbui T., Rambaldi A., Garattini E. Stat1 is induced and activated by all-trans retinoic acid in acute promyelocytic leukemia cells.
Blood
,
89
:
1001
-1012,  
1997
.
212
Kolla V., Weihua X., Kalvakolanu D. V. Modulation of interferon action by retinoids. Induction of murine STAT1 gene expression by retinoic acid(Published erratum appears in J. Biol. Chem., 272: 16068, 1997).
J. Biol. Chem.
,
272
:
9742
-9748,  
1997
.
213
Matikainen S., Ronni T., Lehtonen A., Sareneva T., Melen K., Nordling S., Levy D. E., Julkunen I. Retinoic acid induces signal transducer and activator of transcription (STAT) 1. STAT2, and p48 expression in myeloid leukemia cells and enhances their responsiveness to interferons.
Cell Growth Differ.
,
8
:
687
-698,  
1997
.
214
Miller W. H., Jr., Schipper H. M., Lee J. S., Singer J., Waxman S. Mechanisms of action of arsenic trioxide.
Cancer Res.
,
62
:
3893
-3903,  
2002
.
215
Chen G. Q., Zhu J., Shi X. G., Ni J. H., Zhong H. J., Si G. Y., Jin X. L., Tang W., Li X. S., Xong S. M., Shen Z. X., Sun G. L., Ma J., Zhang P., Zhang T. D., Gazin C., Naoe T., Chen S. J., Wang Z. Y., Chen Z. In vitro studies on cellular and molecular mechanisms of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia: As2O3 induces NB4 cell apoptosis with downregulation of Bcl-2 expression and modulation of PML-RAR α/PML proteins.
Blood
,
88
:
1052
-1061,  
1996
.
216
Wang Z. G., Rivi R., Delva L., Konig A., Scheinberg D. A., Gambacorti-Passerini C., Gabrilove J. L., Warrell R. P., Jr., Pandolfi P. P. Arsenic trioxide and melarsoprol induce programmed cell death in myeloid leukemia cell lines and function in a PML and PML-RARα independent manner.
Blood
,
92
:
1497
-1504,  
1998
.
217
Shao W., Fanelli M., Ferrara F. F., Riccioni R., Rosenauer A., Davison K., Lamph W. W., Waxman S., Pelicci P. G., Lo Coco F., Avvisati G., Testa U., Peschle C., Gambacorti-Passerini C., Nervi C., Miller W. H., Jr. Arsenic trioxide as an inducer of apoptosis and loss of PML/RAR α protein in acute promyelocytic leukemia cells.
J. Natl. Cancer Inst.
,
90
:
124
-133,  
1998
.
218
Zhu J., Koken M. H., Quignon F., Chelbi-Alix M. K., Degos L., Wang Z. Y., Chen Z., de The H. Arsenic-induced PML targeting onto nuclear bodies: implications for the treatment of acute promyelocytic leukemia.
Proc. Natl. Acad. Sci. USA
,
94
:
3978
-3983,  
1997
.
219
Coiffier B., Lepage E., Briere J., Herbrecht R., Tilly H., Bouabdallah R., Morel P., Van Den Neste E., Salles G., Gaulard P., Reyes F., Lederlin P., Gisselbrecht C. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma.
N. Engl. J. Med.
,
346
:
235
-242,  
2002
.
220
Rai K. R. Future strategies toward the cure of indolent B-cell malignancies. New biologic therapies.
Semin. Hematol.
,
36
:
12
-17,  
1999
.
221
Keating M. J. Chronic lymphocytic leukemia.
Semin. Oncol.
,
26
:
107
-114,  
1999
.
222
Shan D., Ledbetter J. A., Press O. W. Apoptosis of malignant human B cells by ligation of CD20 with monoclonal antibodies.
Blood
,
91
:
1644
-1652,  
1998
.
223
Byrd J. C., Kitada S., Flinn I. W., Aron J. L., Pearson M., Lucas D., Reed J. C. The mechanism of tumor cell clearance by rituximab in vivo in patients with B-cell chronic lymphocytic leukemia: evidence of caspase activation and apoptosis induction.
Blood
,
99
:
1038
-1043,  
2002
.
224
Reed J. C., Kitada S., Kim Y., Byrd J. Modulating apoptosis pathways in low-grade B-cell malignancies using biological response modifiers.
Semin. Oncol.
,
29
:
10
-24,  
2002
.
225
Adjei A. A. Blocking oncogenic Ras signaling for cancer therapy.
J. Natl. Cancer Inst.
,
93
:
1062
-1074,  
2001
.
226
Kohl N. E., Mosser S. D., deSolms S. J., Giuliani E. A., Pompliano D. L., Graham S. L., Smith R. L., Scolnick E. M., Oliff A., Gibbs J. B. Selective inhibition of ras-dependent transformation by a farnesyltransferase inhibitor.
Science (Wash. DC)
,
260
:
1934
-1937,  
1993
.
227
James G. L., Goldstein J. L., Brown M. S., Rawson T. E., Somers T. C., McDowell R. S., Crowley C. W., Lucas B. K., Levinson A. D., Marsters J. C., Jr. Benzodiazepine peptidomimetics: potent inhibitors of Ras farnesylation in animal cells.
Science (Wash. DC)
,
260
:
1937
-1942,  
1993
.
228
Gibbs J. B. Ras C-terminal processing enzymes-new drug targets?.
Cell
,
65
:
1
-4,  
1991
.
229
Karp J. E., Lancet J. E., Kaufmann S. H., End D. W., Wright J. J., Bol K., Horak I., Tidwell M. L., Liesveld J., Kottke T. J., Ange D., Buddharaju L., Gojo I., Highsmith W. E., Belly R. T., Hohl R. J., Rybak M. E., Thibault A., Rosenblatt J. Clinical and biologic activity of the farnesyltransferase inhibitor R115777 in adults with refractory and relapsed acute leukemias: a phase 1 clinical-laboratory correlative trial.
Blood
,
97
:
3361
-3369,  
2001
.
230
Karp J. E. Farnesyl protein transferase inhibitors as targeted therapies for hematologic malignancies.
Semin. Hematol.
,
38
:
16
-23,  
2001
.
231
Adjei A. A., Erlichman C., Davis J. N., Cutler D. L., Sloan J. A., Marks R. S., Hanson L. J., Svingen P. A., Atherton P., Bishop W. R., Kirschmeier P., Kaufmann S. H. A Phase I trial of the farnesyl transferase inhibitor SCH66336: evidence for biological and clinical activity.
Cancer Res.
,
60
:
1871
-1877,  
2000
.
232
Kurzrock R., Sebti S. M., Kantarjian H. M., Wright J., Cortes J. E., Thomas D. A., Wilson E., Beran M., Koller C. A., O’Brien S., Freireich E. J., Talpaz M. Phase I Study of a Farnesyl Transferase Inhibitor. R115777, in Patients with Myelodysplastic Syndrome.
Blood
,
98
:
623a
2001
.
233
Blagosklonny M. V., Fojo T., Bhalla K. N., Kim J. S., Trepel J. B., Figg W. D., Rivera Y., Neckers L. M. The Hsp90 inhibitor geldanamycin selectively sensitizes Bcr-Abl- expressing leukemia cells to cytotoxic chemotherapy.
Leukemia (Baltimore)
,
15
:
1537
-1543,  
2001
.
234
Soga S., Neckers L. M., Schulte T. W., Shiotsu Y., Akasaka K., Narumi H., Agatsuma T., Ikuina Y., Murakata C., Tamaoki T., Akinaga S. KF25706, a novel oxime derivative of radicicol, exhibits in vivo antitumor activity via selective depletion of Hsp90 binding signaling molecules.
Cancer Res.
,
59
:
2931
-2938,  
1999
.
235
Shiotsu Y., Neckers L. M., Wortman I., An W. G., Schulte T. W., Soga S., Murakata C., Tamaoki T., Akinaga S. Novel oxime derivatives of radicicol induce erythroid differentiation associated with preferential G(1) phase accumulation against chronic myelogenous leukemia cells through destabilization of Bcr-Abl with Hsp90 complex.
Blood
,
96
:
2284
-2291,  
2000
.
236
Propper D. J., McDonald A. C., Man A., Thavasu P., Balkwill F., Braybrooke J. P., Caponigro F., Graf P., Dutreix C., Blackie R., Kaye S. B., Ganesan T. S., Talbot D. C., Harris A. L., Twelves C. Phase I and pharmacokinetic study of PKC412, an inhibitor of protein kinase C.
J. Clin. Oncol.
,
19
:
1485
-1492,  
2001
.
237
Okuda K., Matulonis U., Salgia R., Kanakura Y., Druker B., Griffin J. D. Factor independence of human myeloid leukemia cell lines is associated with increased phosphorylation of the proto-oncogene Raf-1.
Exp. Hematol.
,
22
:
1111
-1117,  
1994
.
238
Kang C. D., Yoo S. D., Hwang B. W., Kim K. W., Kim D. W., Kim C. M., Kim S. H., Chung B. S. The inhibition of ERK/MAPK not the activation of JNK/SAPK is primarily required to induce apoptosis in chronic myelogenous leukemic K562 cells.
Leuk. Res.
,
24
:
527
-534,  
2000
.
239
Kim S. C., Hahn J. S., Min Y. H., Yoo N. C., Ko Y. W., Lee W. J. Constitutive activation of extracellular signal-regulated kinase in human acute leukemias: combined role of activation of MEK, hyperexpression of extracellular signal-regulated kinase, and downregulation of a phosphatase. PAC1.
Blood
,
93
:
3893
-3899,  
1999
.
240
Morgan M. A., Dolp O., Reuter C. W. Cell-cycle-dependent activation of mitogen-activated protein kinase kinase (MEK-1/2) in myeloid leukemia cell lines and induction of growth inhibition and apoptosis by inhibitors of RAS signaling.
Blood
,
97
:
1823
-1834,  
2001
.
241
Alessi D. R., Cuenda A., Cohen P., Dudley D. T., Saltiel A. R. PD 098059 is a specific inhibitor of the activation of mitogen- activated protein kinase kinase in vitro and in vivo.
J. Biol. Chem.
,
270
:
27489
-27494,  
1995
.
242
Dudley D. T., Pang L., Decker S. J., Bridges A. J., Saltiel A. R. A synthetic inhibitor of the mitogen-activated protein kinase cascade.
Proc. Natl. Acad. Sci. USA
,
92
:
7686
-7689,  
1995
.
243
Favata M. F., Horiuchi K. Y., Manos E. J., Daulerio A. J., Stradley D. A., Feeser W. S., Van Dyk D. E., Pitts W. J., Earl R. A., Hobbs F., Copeland R. A., Magolda R. L., Scherle P. A., Trzaskos J. M. Identification of a novel inhibitor of mitogen-activated protein kinase kinase.
J. Biol. Chem.
,
273
:
18623
-18632,  
1998
.
244
Sebolt-Leopold J. S. Development of anticancer drugs targeting the MAP kinase pathway.
Oncogene
,
19
:
6594
-6599,  
2000
.
245
Powis G., Bonjouklian R., Berggren M. M., Gallegos A., Abraham R., Ashendel C., Zalkow L., Matter W. F., Dodge J., Grindey G., et al Wortmannin, a potent and selective inhibitor of phosphatidylinositol-3- kinase.
Cancer Res.
,
54
:
2419
-2423,  
1994
.
246
Vlahos C. J., Matter W. F., Hui K. Y., Brown R. F. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4- morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002).
J. Biol. Chem.
,
269
:
5241
-5248,  
1994
.
247
Roth W., Reed J. C. Apoptosis and cancer: when BAX is TRAILing away.
Nat. Med.
,
8
:
216
-218,  
2002
.
248
Buolamwini J. K. Novel anticancer drug discovery.
Curr. Opin. Chem. Biol.
,
3
:
500
-509,  
1999
.
249
Martin S. J. Destabilizing influences in apoptosis: sowing the seeds of IAP destruction.
Cell
,
109
:
793
-796,  
2002
.
250
Stennicke H. R., Ryan C. A., Salvesen G. S. Reprieval from execution: the molecular basis of caspase inhibition.
Trends Biochem. Sci.
,
27
:
94
-101,  
2002
.
251
LaCasse E. C., Baird S., Korneluk R. G., MacKenzie A. E. The inhibitors of apoptosis (IAPs) and their emerging role in cancer.
Oncogene
,
17
:
3247
-3259,  
1998
.
252
Holcik M., Gibson H., Korneluk R. G. XIAP: apoptotic brake and promising therapeutic target.
Apoptosis
,
6
:
253
-261,  
2001
.
253
Tamm I., Kornblau S. M., Segall H., Krajewski S., Welsh K., Kitada S., Scudiero D. A., Tudor G., Qui Y. H., Monks A., Andreeff M., Reed J. C. Expression and prognostic significance of IAP-family genes in human cancers and myeloid leukemias.
Clin. Cancer Res.
,
6
:
1796
-1803,  
2000
.
254
Holcik M., Yeh C., Korneluk R. G., Chow T. Translational upregulation of X-linked inhibitor of apoptosis (XIAP) increases resistance to radiation induced cell death.
Oncogene
,
19
:
4174
-4177,  
2000
.
255
Ciechanover A. The ubiquitin-proteasome proteolytic pathway.
Cell
,
79
:
13
-21,  
1994
.
256
Almond J. B., Cohen G. M. The proteasome: a novel target for cancer chemotherapy.
Leukemia (Baltimore)
,
16
:
433
-443,  
2002
.
257
King R. W., Deshaies R. J., Peters J. M., Kirschner M. W. How proteolysis drives the cell cycle.
Science (Wash. DC)
,
274
:
1652
-1659,  
1996
.
258
Karin M., Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF-[κ]B activity.
Annu. Rev. Immunol.
,
18
:
621
-663,  
2000
.
259
Chang Y. C., Lee Y. S., Tejima T., Tanaka K., Omura S., Heintz N. H., Mitsui Y., Magae J. mdm2 and bax, downstream mediators of the p53 response, are degraded by the ubiquitin-proteasome pathway.
Cell Growth Differ.
,
9
:
79
-84,  
1998
.
260
Breitschopf K., Zeiher A. M., Dimmeler S. Ubiquitin-mediated degradation of the proapoptotic active form of bid. A functional consequence on apoptosis induction.
J. Biol. Chem.
,
275
:
21648
-21652,  
2000
.
261
Li B., Dou Q. P. Bax degradation by the ubiquitin/proteasome-dependent pathway: involvement in tumor survival and progression.
Proc. Natl. Acad. Sci. USA
,
97
:
3850
-3855,  
2000
.
262
Marshansky V., Wang X., Bertrand R., Luo H., Duguid W., Chinnadurai G., Kanaan N., Vu M. D., Wu J. Proteasomes modulate balance among proapoptotic and antiapoptotic Bcl-2 family members and compromise functioning of the electron transport chain in leukemic cells.
J. Immunol.
,
166
:
3130
-3142,  
2001
.
263
Gardner R. C., Assinder S. J., Christie G., Mason G. G., Markwell R., Wadsworth H., McLaughlin M., King R., Chabot-Fletcher M. C., Breton J. J., Allsop D., Rivett A. J. Characterization of peptidyl boronic acid inhibitors of mammalian 20 S and 26 S proteasomes and their inhibition of proteasomes in cultured cells.
Biochem. J.
,
346 Pt 2
:
447
-454,  
2000
.
264
Almond J. B., Snowden R. T., Hunter A., Dinsdale D., Cain K., Cohen G. M. Proteasome inhibitor-induced apoptosis of B-chronic lymphocytic leukaemia cells involves cytochrome c release and caspase activation, accompanied by formation of an approximately 700 kDa Apaf-1 containing apoptosome complex.
Leukemia (Baltimore)
,
15
:
1388
-1397,  
2001
.
265
Masdehors P., Omura S., Merle-Beral H., Mentz F., Cosset J. M., Dumont J., Magdelenat H., Delic J. Increased sensitivity of CLL-derived lymphocytes to apoptotic death activation by the proteasome-specific inhibitor lactacystin.
Br. J. Haematol.
,
105
:
752
-757,  
1999
.
266
Masdehors P., Merle-Beral H., Maloum K., Omura S., Magdelenat H., Delic J. Deregulation of the ubiquitin system and p53 proteolysis modify the apoptotic response in B-CLL lymphocytes.
Blood
,
96
:
269
-274,  
2000
.
267
Chandra J., Niemer I., Gilbreath J., Kliche K. O., Andreeff M., Freireich E. J., Keating M., McConkey D. J. Proteasome inhibitors induce apoptosis in glucocorticoid-resistant chronic lymphocytic leukemic lymphocytes.
Blood
,
92
:
4220
-4229,  
1998
.
268
Adams J., Palombella V. J., Sausville E. A., Johnson J., Destree A., Lazarus D. D., Maas J., Pien C. S., Prakash S., Elliott P. J. Proteasome inhibitors: a novel class of potent and effective antitumor agents.
Cancer Res.
,
59
:
2615
-2622,  
1999
.
269
Hideshima T., Richardson P., Chauhan D., Palombella V. J., Elliott P. J., Adams J., Anderson K. C. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells.
Cancer Res.
,
61
:
3071
-3076,  
2001
.
270
Hideshima T., Chauhan D., Richardson P., Mitsiades C., Mitsiades N., Hayashi T., Munshi N., Dang L., Castro A., Palombella V., Adams J., Anderson K. C. NF-κ B as a therapeutic target in multiple myeloma.
J. Biol. Chem.
,
277
:
16639
-16647,  
2002
.
271
Mitsiades N., Mitsiades C. S., Poulaki V., Chauhan D., Richardson P. G., Hideshima T., Munshi N., Treon S. P., Anderson K. C. Biologic sequelae of nuclear factor-κB blockade in multiple myeloma: therapeutic applications.
Blood
,
99
:
4079
-4086,  
2002
.
272
Sherr C. J. The Pezcoller lecture: cancer cell cycles revisited.
Cancer Res.
,
60
:
3689
-3695,  
2000
.
273
Sellers W. R., Rodgers J. W., Kaelin W. G., Jr. A potent transrepression domain in the retinoblastoma protein induces a cell cycle arrest when bound to E2F sites, Proc.
Natl. Acad. Sci. USA
,
92
:
11544
-11548,  
1995
.
274
Magnaghi-Jaulin L., Groisman R., Naguibneva I., Robin P., Lorain S., Le Villain J. P., Troalen F., Trouche D., Harel-Bellan A. Retinoblastoma protein represses transcription by recruiting a histone deacetylase.
Nature (Lond.)
,
391
:
601
-605,  
1998
.
275
Krug U., Ganser A., Koeffler H. P. Tumor suppressor genes in normal and malignant hematopoiesis.
Oncogene
,
21
:
3475
-3495,  
2002
.
276
Sauerbrey A., Stammler G., Zintl F., Volm M. Expression of the retinoblastoma tumor suppressor gene (RB-1) in acute leukemia.
Leuk. Lymphoma
,
28
:
275
-283,  
1998
.
277
Drexler H. G. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia-lymphoma cells.
Leukemia (Baltimore)
,
12
:
845
-859,  
1998
.
278
Chim C. S., Liang R., Tam C. Y., Kwong Y. L. Methylation of p15 and p16 genes in acute promyelocytic leukemia: potential diagnostic and prognostic significance.
J. Clin. Oncol.
,
19
:
2033
-2040,  
2001
.
279
Senderowicz A. M. Development of cyclin-dependent kinase modulators as novel therapeutic approaches for hematological malignancies.
Leukemia (Baltimore)
,
15
:
1
-9,  
2001
.
280
Shapiro G. I., Harper J. W. Anticancer drug targets: cell cycle and checkpoint control.
J. Clin. Investig.
,
104
:
1645
-1653,  
1999
.
281
Sausville E. A., Arbuck S. G., Messmann R., Headlee D., Bauer K. S., Lush R. M., Murgo A., Figg W. D., Lahusen T., Jaken S., Jing X., Roberge M., Fuse E., Kuwabara T., Senderowicz A. M. Phase I trial of 72-hour continuous infusion UCN-01 in patients with refractory neoplasms.
J. Clin. Oncol.
,
19
:
2319
-2333,  
2001
.