throbber
This material may be protected by Copyright law (Title 17 U.S. Code)
`
`1
`
`

`

`Clinical Cancer Research 535
`Vol. 9. 535-550, February 2003
`
`
`
`This material may be protected by Copyright law (Title 17 U5. Code)
`
`Minireview
`
`Modulation of Cellular Signaling Pathways: Prospects for Targeted
`Therapy in Hematological Malignancies
`
`Farhad Ravandi,1 Moshe Talpaz, and Zeev Estrov
`Department of Hematology/Oncology, The University of Illinois at
`Chicago, Chicago, Illinois 60612 [F. R.], and Department of
`Bioimmunotherapy, The University of Texas M. D. Anderson Cancer
`Center, Houston, Texas 77030 [M. T., Z. E.]
`
`Abstract
`
`The high remission rates observed in patients with
`chronic myelogenous leukemia who receive Imatinib mesy-
`late (Gleevec) indicate that targeted therapy for hematolog-
`ical 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 inhib-
`itors. This review attempts to provide a practical introduc-
`tion to the complex and growing field of targeted therapy in
`hematological malignancies.
`
`Introduction
`
`Cellular proliferation, differentiation, and death are regu-
`lated 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 intracel—
`lular signaling pathways, which in turn activate appropriate
`cellular machinery (1). Cell-surface receptors that convert ex—
`ternal stimuli into intracellular signals are pivotal in this signal—
`ing process. They activate intracellular pathways either through
`their inherent enzymatic function or as a result of their associ-
`ation 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 gen—
`eration of appropriate constellation of hematopoietic cells, and
`their abnormal activation results in neoplastic transformation,
`impaired apoptosis, and uncontrolled proliferation. Cytokines
`
`Received 6/25/02; revised 9/5/02; accepted 9/6/02.
`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.
`1 To whom requests for reprints should be addressed, at University of
`Illinois—Chicago, 840 South Wood Street, MC 787 Chicago, IL 60612—
`7323. Phone: (312) 996-5982; Fax: (312) 413-4131; E-mail: ravandi@
`, uic.edu.
`
`function in a redundant and pleiotropic manner; different cyto—
`kines can exert similar effects on the same cell type, and any
`particular cytokine can have several differing biological func—
`tions (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—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 pleck-
`strin 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). Cyto-
`kine receptors without intrinsic kinase activity transmit their
`signals primarily through activation of Jak kinases. These re-
`ceptors 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 ser-
`ine/threonine kinase cascades such as the Ras/Raf/MEK/ERK
`(MAPK; Ref. 10). For example, after ligand binding, the [3—
`subunit of IL-3 and GM-CSF receptors are phosphorylated and,
`through recruitment of adaptor proteins such as She, Grb2, and
`
`2 The abbreviations used are: TK, tyrosine kinase; Jak, janus kinase;
`Stat, signal transducer and activator of transcription; ERK, extracellular
`signal—regulated kinase; MAPK, mitugcndiclivated protein kinase; IL,
`interleukin: GM—CSF, grunultacytc macrophage colony—stimulating fac—
`tor; JNK, c—Jun NHZ—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—l, SH—2 domain containing protein tyrosine phosphatase-l;
`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
`myulugenous leukemia; ALL, acute lymphoblastic leukemia; MEK,
`milngcwactivatcd protein/extracellular signal-regulated kinase kinase;
`ALK, anaplastic lymphoma kinase: NPM, nuclcophusmin; A'I‘RA, all-
`lrmis-reiinoic acid; AP}... acute promyclocytic leukemia; NFKB. nuclear
`factor KB; IKB, inhibitor of nuclear factor—KB; HDAC, histone deacety-
`lase; IAP, inhibitor of apoptotic pathway; CDK, cyclin—dependent ki-
`nase; CDKI, cyclin-dependent kinase inhibitor; Rb, retinoblastoma;
`RAR, retinoic acid receptor; XIAP, X—linked inhibitor of apoptosis.
`
`2
`
`

`

`536 Cellular Signaling Pathways as Therapeutic Targets
`
`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—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—18). Similarly, PI3K associates with the B chain
`of IL-3 receptor, recruits PKB/AKT by phosphorylation of its
`serine residues, and transmits cellular survival signals (19—21).
`Another downstream protein to IL-3 activation is the p7OS6
`kinase, which also interacts with the B chain and mediates
`appropriates signals (22, 23). Ultimately, these pathways influ—
`ence gene transcription through their ability to recruit transcrip—
`tion 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
`PDGFRB, 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.
`
`Jak-Stat Signaling Pathways
`Hematopoietic cell proliferation and differentiation is reg-
`ulated 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—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 gpl30 family, the IL—2 receptor family, the growth
`hormone receptor family, the IFN family, and the gpl40 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 OL, B, and y 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 ot—subunit, and signal
`transducing B and y
`subunits. Alternatively, the gp140 family including the receptors
`for IL—3,
`IL—5, and GM—CSF have a unique ligand—binding
`(it—subunit and a common B (gpl40) signal—transducing unit (3).
`
`3
`
`Cytokine
`receptor
`
`
`
`Eli!
`Phosphate groups
`Nuciaar
`membrane
`
`Phonphnnrlallon
`by Jaks of Stats
`
`/
`
`
`
`(—)\g
` fl
`
`[+) Slat dimer
`
`
`
`
`
`
`
`Cell surface
`
`Fig. 1 The Jak-Stat pathway and its regulation.
`
`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 in-
`tracellular 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 tar—
`get 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 (Jakl, Jak2, Jak3, and Tyk2) that can bind cyto-
`kine receptor subunits, phosphorylate them, and in doing so
`creat docking sites on the receptors for binding of SH2-contain-
`ing proteins (33, 34). In general, Jaks consist of several domains
`(JHl-JH7) of which the functional significance has been char-
`acterized 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 'pro-
`teins 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 trans-
`locations resulting in TEL—JAK2 constructs lead to the con-
`stitutive activation of Stat5, IL-3-independent cellular proli-
`feration,
`and leukemogenesis
`(42, 43). The translocation
`t(9;12)(p24;p13) results in the fusion of the kinase catalytic
`
`3
`
`

`

`Clinical Cancer Research 537
`
`
`region of JAK2 with the transcription factor TEL generating the
`constitutively active TEL-IAKZ. Similarly, infection with on—
`cogenic 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 includ—
`ing different isomers of Stats 1, 3, 4, and 5 (3, 46). Like other
`transcription factors Stats have a well-defined structure includ—
`ing 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—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 translo-
`cation to the nucleus, followed by DNA binding and gene
`activation (Fig. l; Refs. 51, 52). The specificity for Stat phos-
`phorylation 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 spe—
`cific genes. Stats also interact with other transcription factors
`such as the p300/cyclic AMP-responsive element binding pro-
`tein farme of coactivators to activate genes (55, 56). The
`transcriptional activity of Stats may also be regulated by the
`phosphorylation of their scrine 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, Statl mediates the gro\\-'tli—inhibitory
`effects of lFN—y. through the induction of the CDKI p'_’l“““t
`whereas Stat5 mediates proliferative effects of IL-3 and GM-
`CSF (60, 61). Similarly, phosphorylation of Stat3 can result both
`in IL—6- and lL—lO—induced growth arrest, and in GM-CSF— and
`IL-3—induced proliferation (61—63). Stats also modulate cellular
`differentiation and apoptosis. Reconstitution of Statl in Statl—
`null U3A cells (which do not respond to TNF-OL) restores basal
`caspase expression and renders them sensitive to TNF—induced
`apoptosis (64). Conversely, Stat3 and Stat5 mediate the anti—
`apoptotic effects of IL-6 and IL-2, respectively (65, 66). Statl
`activates the caspase cascade through up—regulation of Fas and
`FasL expression in response to IFN-y (67). The exact mecha-
`nisms 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 prom—
`ising early results in clinical
`trials (68, 69). The cytokine-
`
`activated Jak—Stat pathways are also inhibited by tyrosine de-
`phosphorylation mediated by cytoplasmic phosphatases such as
`SHP—l
`(70, 71). SHP-l-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—76). The role of these inhib-
`itory proteins in the pathogenesis of neoplastic transformation is
`also becoming clearer (77).
`
`RTK and Serine/Threonine Signaling Pathways
`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—80). These enzymes are activated by
`ligand binding, by cell-cell interactions via cell adhesion mol-
`ecules, 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-contain-
`ing 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
`11 including insulin-like growth factor-1 receptor; class 111
`including PDGFR, macrophage
`colony-stimulating factor
`(FMS-R or CSF—lR), stem cell factor receptor (KIT), and
`FLT3R; and class IV including FGFR (79, 83, 84). The impor-
`tance of these receptors in malignant transformation and the
`possibility of modulating them as therapeutic targets are sub—
`jects of intense research. The recent reports of constitutive
`activation of FLT3R resulting in stimulation of multiple signal—
`ing pathways and leading to malignant transformation has been
`of significant interest in leukemia research (85, 86). Such con—
`stitutive 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 juxtarnembrane domain of receptor (87—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 phosphoty-
`rosine on the activated RTKs. These proteins include GTPasc
`activating protein, PI3K, Grb2, and Src—like tyrosine-kinases (1,
`10, 78). The activation of these proteins by scrine/threonine
`phosphorylation in turn activates a number of downstream sig—
`naling cascades that lead to gene transcription (10).
`Although knowledge of the Jak-Stat pathway has been
`instrumental in understanding cytokine signaling (94), the im—
`portance of signaling cascades that involve the activation of
`serine/threonine kinases is increasingly apparent (10). The ser—
`ine/threonine MAPKs, which include the Ras—Raf-MEK-ERK
`pathway,
`the p38 family of kinases, and the JNK (SAPK)
`
`4
`
`

`

`538 Cellular Signaling Pathways as Therapeutic Targets
`
`family, are activated by upstream signals and mediate effects on
`inflammation, cell growth, cell cycle progression, cell differen-
`tiation, 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 prolif—
`erative 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 prolifera—
`tion by regulating the activity of several proteins, including
`cell-cycle regulators (e.g., cyclin D1, p21wnfmip], p27kip], and
`cchSA) and transcription factors (e.g., c-Myc; Ref. 106).
`The Gl/S cell cycle checkpoint is a critical point determin-
`ing the commitment of cells to growth arrest or proliferation.
`During this stage cells are responsive to cytokines (107). Reg-
`ulatory proteins p21"’“f“Cipl and p27kipl are of particular impor-
`tance in this transition, which is controlled by both positive and
`negative regulators. Distinct Rb—E2F repressor complexes sup—
`press the transcription of genes required for progression of
`various phases of cell cycle. For example, Rb—E2Fl complex
`suppresses the progression through G1 (108). During this pro—
`gression from G1 to S—phase cyclin/CDKs are sequentially ac—
`tivated, which then inactivate suppressor complexes such as
`Rb—E2F1 (109). Cyclin/CDK activity results in Rb phosphoryl—
`ation and its dissociation from E2Fl leading to activation of
`genes necessary for S phase (110). Activity of a number of these
`cyclin/CDKs as well their inhibitors such as p21‘w‘fllCipl is mod-
`ulated by cytokine-mediated signals through their phosphory-
`lation.
`
`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 (or, B, y, and
`8) 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 stand—
`point 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 aretacti—
`
`vated by specific MAPK kinases (MEKKs) in response to
`inflammatory cytokines such as TNF—a, and other stress stimuli
`such as reactive oxygen species, heat, and withdrawal of growth
`factors (117). The MEKKl/JNK signaling increases p53 stabil—
`ity and transcriptional activation, and MEKKl/JNK potentiates
`the ability of p53 to initiate apoptosis (118).
`Normal functioning of MAPK-mediated signaling necessi—
`tates its efficient inactivation (95). A number of dual—specificity
`MAPK phosphatases serve to dephosphorylate and, hence, in-
`activate MAPKs (119—121). Similarly, protein phosphatases
`PM and PP2 dephosphorylate and inactivate a number of phos—
`phoproteins 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 inter-
`action 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 NFKB (128—130).
`The serine/threonine kinase AKT is an important component of
`the cell survival machinery (10, 131—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 IKB
`by AKT releases NFKB from its association with IKB. N_FKB
`then moves into the nucleus, where it induces a number of genes
`involved in cell survival (131). Meanwhile, the inhibitory pro—
`tein IKB is degraded by the proteasome (136). AKT also phos-
`phorylates 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 phos—
`phate 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—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, Bel-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 differ—
`ent ways (Fig. 2). For example, PKC is a potent activator of
`Raf—1, which activates the MAPK cascade. This leads to phos—
`phorylation of IKB, release of NFKB, translocation of NFKB into
`the nucleus, and gene transcription as described above (144—
`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, stauros—
`
`5
`
`

`

`Clinical Cancer Research 539
`
`
`Neighboring cell
`
` Cytokine
`receptor
`
`
`
`Fig. 2
`ways.
`
`Interactions of diverse signaling path—
`
`
`30172 4'" """“' " "
`
`
`
`
`
`Cailular membrane
`
`porine analogs UCN—Ol and CGP 41251 are being examined as
`inhibitors of PKC and MAPK signaling (149, 150).
`Various signaling pathways interact resulting in their mod-
`ulation at several levels. For example, PI3K interacts with and
`enhances Raf-Ras-MEK—ERK pathway (151—153). Other ser-
`ine/threonine phosphorylation pathways modulate cytokine sig-
`nals through the Jak—Stat pathway (10, 154). In addition to being
`tyrosine phosphorylated, several Stats (StatIoL, 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).
`
`Therapeutic Implications
`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 path-
`ways. 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 caus—
`ing 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 path—
`ways 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 be-
`
`tween 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, INK/SAPK, Crkl,
`and Stat5 (156—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—163). The specific Bcr-Abl inhib-
`itor Imatinib mesylate has proven to be very effective in sup-
`pressing these pathways in vitro and in patients with CML and
`ALL (164—170).
`Although the platelet—derived growth factors, PDGFROI.
`and PDGFRB have significant homology, only the PDGFRB 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—PDGFRB
`(171, 172). As a result of ligand-independent dimerization and
`autophosphorylation of the PDGF B—subunit, the TK is consti-
`tutively active. PDGF is able to stimulate the growth of primi-
`tive hematopoietic, erythroid, and megakaryocytic precursors,
`and TEL-PDGFRB can confer cytokine-independent growth to
`Ba/F3cells
`(173).
`Imatinib mesylate
`inhibits
`the kinase
`PDGFRB 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
`NHz—terminal domain of NPM to the cytoplasmic region of the
`ALK TK receptor results in constitutive activation of its cata-
`lytic 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—182). Therefore, design of specific inhib-
`
`6
`
`

`

`540 Cellular Signaling Pathways as Therapeutic Targets
`
` "scri t'on
`
`pl
`
`Fig. 3 Targets for inhibition in Jak-Stat signaling pathway.
`
`Gene 1:33
`
`Cell Membrane
`
`in
`itors of the NPM-ALK may be of considerable interest
`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; lL—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 neo—
`plastic 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). Acti—
`vating mutations of FLT3—R including the internal tandem du—
`plication 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 cur-
`rently under way. Intracellular activators of Stats such as Jaks
`and Src are also likely targets (164—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, nu-
`merous other cellular TKs have been ident

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket