Leukemia (2002) 16, 486–507  2002 Nature Publishing Group All rights reserved 0887-6924/02 $25.00 www.nature.com/leu REVIEW

The Raf/MEK/ERK signal transduction cascade as a target for chemotherapeutic intervention in leukemia JT Lee Jr1 and JA McCubrey1,2

1Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA; and 2Leo Jenkins Cancer Center, Brody School of Medicine at East Carolina University, Greenville, NC, USA

The Raf/MEK/ERK (MAPK) signal transduction cascade is a constituents of these pathways are also ideal candidates for vital mediator of a number of cellular fates including growth, novel anti-neoplastic chemotherapies. This review will focus proliferation and survival, among others. The focus of this review centers on the MAPK signal transduction pathway, its on the MAP cascades, the repertoire of proteins which mechanisms of activation, downstream mediators of signaling, comprise these pathways, and possible routes of chemothera- and the transcription factors that ultimately alter peutic intervention for leukemic disorders within these expression. Furthermore, negative regulators of this cascade, signaling cascades. SPOTLIGHT including phosphatases, are discussed with an emphasis placed upon chemotherapeutic intervention at various points along the pathway. In addition, mounting evidence suggests that the PI3K/Akt pathway may play a role in the effects elicited 2 Overviewof the MAP via MAPK signaling; as such, potential interactions and their possible cellular ramifications are discussed. The serine/threonine MAP kinases are activated in response Leukemia (2002) 16, 486–507. DOI: 10.1038/sj/leu/2402460 to upstream receptor tyrosine kinases and/or cytokine recep- Keywords: Ras; Raf; MEK; ERK; phosphatase; transcription factor; tors that associate with heterotrimeric G proteins so as to trig- inhibitors; IL-3; signal transduction; leukemia; oncogenes; PI3K; Akt ger downstream pathways. The most extensively studied MAP kinase pathway is the Raf-MEK-ERK cascade. Just over a dec- ade ago, the extracellular signal-regulated kinases (ERKs) were 1 Introduction identified as a novel protein kinase family that exhibited pro- liferative effects when activated.2–5 Subsequent studies soon Hematopoiesis occurs through a complex series of intricate, identified the upstream activators of ERK, the MEK (MAP/ERK highly regulated intracellular steps, each of which ultimately kinase) family of kinases; the MEKs are dual-specificity kinases contribute to the proper development of the body’s myeloid, that can phosphorylate both serine/threonine and tyrosine lymphoid and erythroid lineages. Unfortunately though, these residues on the MAP kinases.6–8 A kinase responsible for acti- processes sometimes become dysregulated due to over- vation of MEK is Raf and directly upstream of Raf is the expression of certain oncogenes, aberrant behavior of onco- GTPase, Ras.9,10 The Ras oncoprotein has been noted to be proteins, and expression of chromosomal abnormalities, mutated in approximately 20% of all human malignancies.11 among others. Thus, the elaborate process of hematopoiesis Together, these proteins relay various stimuli to the cell can be changed from one that is necessary for life to another nucleus, each of which ultimately contribute to cellular that is able to endanger it. proliferation.12 Our understanding of the cellular changes that occur in Another member of the MAP kinase family of proteins is the order for leukemogenesis to arise is essential for the develop- p38 kinase. The p38 group of MAPKs has been found to be ment of successful therapies to treat these types of disorders. involved in inflammation, cell growth, cell differentiation, cell A prime example of how comprehending a cell’s demise can cycle progression, and cell death.13 p38 exists in four different lead to the development of an effective drug is the Bcr-Abl isoforms: ␣, ␤, ␥ and ␦.14–17 Although each of these isoforms inhibitor STI571 (trademark name, Gleevec); is moderately different in sequence(s) and structure(s), their STI571 functions by competitively inhibiting the activation loops each contain a TGY sequence motif. The p38 of ATP on the kinase thus preventing downstream activation MAP kinases are selectively activated by two MEK isoforms, and amplification of proliferative signals. In its first phase of either MEK3 or MEK6. MEK6 appears to phosphorylate each clinical trials, STI571 proved to have hematologic responses of the p38 isoforms with little to no discrimination between in 53 out of 54 patients with chronic myelogenous leukemia each of them. However, MEK3 is only able to activate the (CML) within a 4-week period.1 Furthermore, extensive testing p38␣ and p38␤ isoforms.18

is now underway to explore these promising results. The third MAPK of interest is the c-JUN NH2-terminal pro- As alluded to above, the exploitation of knowledge gained tein kinase (JNK) which is also commonly referred to as the from studying cell signaling pathways can be extremely stress-activated protein kinase (SAPK). The JNK kinases are attractive when considering chemotherapeutic options for tre- similar to the p38 family of kinases in that although many of ating a variety of neoplasia. The -activated protein the stimuli that activate these pathways are pro-apoptotic, the kinases (MAPKs) are key regulators of embryogenesis, cell dif- biological outcome of the aforementioned stimuli appears to ferentiation, cell proliferation, and even apoptosis; thus, the be largely dependent on cell type. There are three JNK family members (JNK1, JNK2, JNK3) which are activated by two dis- tinct MAPK kinases: MKK4/SEK1 and MKK7.19–21 Each of the Correspondence: JA McCubrey, Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, upstream mediators of JNK phosphorylation are activated in Greenville, NC 27858 USA; Fax: 252 816 3104 response to a variable number of stimuli including inflamma- Received 21 December 2001; accepted 16 January 2002 tory cytokines (ie TNF and IL-1␤), UV light, reactive oxygen Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 487 species (ROS), heat, osmotic shock and withdrawal of growth increased tyrosine kinase activity on the intracellular portion factors.22 A simplistic overview of the MAP kinase cascades of the receptor. Much of the tyrosine kinase activity is due to is presented in Figure 1. autophosphorylation that occurs within the two units of the receptor, where one acts as a kinase and the other as a sub- strate.26 These phosphorylated residues then become docking 3 The Ras-Raf-MEK-ERK signal transduction cascade sites for intracellular proteins that contain Src Homology-2 (SH-2) domains. Through this process, an intracellular con- Target: ligand–receptor interaction glomerate of proteins can initiate immediate signaling, in a variety of cascades, from the binding of one ligand to its As previously mentioned, the Raf-MEK-ERK signal transduc- receptor.27–29 tion cascade is primarily activated in response to various Dysregulated expression of either the ligand or the receptor extracellular growth factors which are able to initiate intra- can contribute to the development of neoplastic growth. Thus, cellular signaling. This mitogenic signal most often occurs at targeting either the ligand or the receptor with chemotherapy the level of a ligand–receptor interaction, followed by down- is an ideal candidate for regulating uncontrolled signaling stream signaling, which ultimately causes altered regulation from within these complexes. This tactic would most likely be of the responsible for oncogenesis. For this reason, com- employed so as to neutralize the receptor with a synthesized prehending the complex, interwoven signaling that occurs ligand (or vice versa) by overexpression of such a molecule. from receptor engagement to the onset of gene transcription This section contains a discussion of several growth factors, can help us unlock the mysteries of leukemogenesis and allow their receptors, and their roles in oncogenesis. Furthermore, for the development of chemotherapeutic drugs. possible sites for chemotherapeutic intervention will also be Cytokine and growth factor receptors are composed of at addressed. least two subunits which, when alike, form homodimers (ie granulocyte colony-stimulating factor (G-CSF) and EPO receptor). These receptor subunits may also be different (ie IL- Insulin-like growth factor-1 (IGF-1) receptor 3, GM-CSF, IL-5 receptors), which are collectively known as heterodimers.23,24 Some receptors appear to be non-discrimi- IGF-1-R has been associated with a variety of types of carci- nate with respect to their dimerization preference, in which nomas including breast cancer,30 medullocarcinomas,31 case they may form either hetero- or homodimers (ie epider- Ewing’s sarcoma,32 pancreatic tumors,33 and a variety of leu- mal growth factor receptor (EGF-R) family members).25 kemias,34 among others. Most recently, researchers have been Finally, some receptors exist as trimer entities, such as the investigating the correlation between IGF-1-R/IGF-1-binding tumor necrosis factor receptor (TNFR). When a growth factor, protein (IGF-1-BP) expression and survival in child patients such as EGF, binds to its respective receptor, it induces both with acute lymphoblastic leukemia (ALL).35,36 The mitogenic the aforementioned dimerization of the receptor and signal of the IGF-1 receptor/protein interaction is most often transduced via the PI3K pathway,37 but sometimes signaling is relayed via either the MAPK pathway or by mitochondrial translocation of Raf-1.38 Furthermore, it has been reported that two of these three pathways must be active for the mitogenic survival signal to proceed.39 Because signaling through this receptor is able to lead to tumor development, it is an ideal candidate for chemotherapeutic intervention. A schematic representation of potential signal transduction pathways activated by IGF-1 is described in Figure 2. The IGF-1-R gene encodes for a polypeptide gene product that is subsequently proteolytically cleaved into ␣- and ␤- chains that ultimately comprise the receptor. Theoretically speaking, protease inhibitors should therefore be able to inhibit formation of the cleaved subunits of the IGF-1 receptor. The IGF-1 receptor, specifically the ␤-subunit, has been SPOTLIGHT found to be in an altered state in human leukemic cells where it exists as a larger (105 kDa), fetal version of the protein, thereby causing malignancy.40 Accordingly, it has been a tar- get of high priority for neutralization in leukemic disorders. As such, it has been shown, that when evaluating the potential role of IGF-1 as a growth-promoting factor in malignant lym- phocyte development, that ALL cell lines will halt proliferation in response to the addition of anti-IGF-1R antibodies.41 These results had been predicted from previous research which stated that both insulin and IGF-1 were able to stimulate growth of cultured, serum-starved human leukemia cells via their respective receptors.42 Also, IGF-1R overexpression has been shown to be sufficient to abrogate the cytokine depen- dence of FDC-P1 hematopoietic precursor cells, an essential Figure 1 Overview of the MAPK cascades. The MAPK signal trans- 43 duction pathways can be induced by either cytokines or stress. Acti- step in leukemogenesis. Most recently, research has vation of the different cascades can have diverse effects on cell revealed that in patients with CML, treatment with interferon- growth, inflammatory responses, and/or apoptosis. ␣ is able to increase expression of IGF-1R on lymphoid cells.

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 488 SPOTLIGHT

Figure 3 Mechanisms of regulation of PDGF receptor (PDGF-R) Figure 2 Mechanism and diversity of anti-apoptotic signaling elic- activity. (a) Normal activation of the PDGF receptor by PDGF; (b–d) ited via IGF-1/IGF-1 receptor interactions. (a) Inactive IGF-1 receptor; Different mechanisms to regulate PDGF-R activity by targeting: (b) (b) Induction of the PI3K cascade via an active IGF-1 receptor; (c) ligand binding; (c) receptor dimerization; and (d) receptor tyrosine Induction of the Ras/Raf/MEK/ERK cascade via an active IGF-1 kinase activity. receptor.

ation.51,52 Furthermore, the development of chronic myelo- Thus, overexpression of the IGF-1R may enable the immune monocytic leukemia and progression to AML is directly system to mount a more efficient attack on the malignant cell related to the constitutive activation of PDGF ␤ receptors as clone by enhancing immunocompetent cell proliferation a result of an aberrant chromosomal translocation.53,54 The and action.44 aforementioned activation of the PDGF receptor is a direct Perhaps the most intriguing aspect of the mitogenic sig- result of its translocation with the ets-like transcription factor, naling that occurs from the IGF-1 receptor is the level of cross- TEL.53,55 This association fuses the amino terminus of TEL, talk that is initiated through the PI3K pathway. As previously which contains the helix–loop–helix domain, to the trans- mentioned, the vast majority of signaling through IGF-1R membrane and cytoplasmic domains of the PDGF ␤ receptor occurs at the level of PI3K. PI3K is then able to activate causing constitutive PDGF-R activity. A similar oncogenic Akt/PKB in an indirect manner so as to elicit anti-apoptotic chromosomal translocation with TEL has been noted with the effects.45,46 It is at this point that synergistic signaling has been Janus associated kinase (JAK), whereby cytokine-independent reported. Recent evidence shows that when both Akt and Raf cellular proliferation resulted.56 Moreover, it has also been are active, the frequency of isolation of cytokine-independent discovered that the PDGF-B (ligand) subunit is associated with hematopoietic cells increases over 100-fold.47 A more in- the pathogenesis of marrow fibrosis associated with CML.57 depth discussion about PI3K/MAPK interactions is discussed Lastly, the PDGF-A growth factor has been isolated from two later in this review. pre-B ALL cell lines, implying a possible autocrine component to the transformation process.58 Taken together, these data suggest that PDGF and its cognate receptor are ideal targets Platelet-derived growth factor (PDGF) receptor for drug therapies. Researchers have developed PDGF antagonists at each level PDGF family members are dimeric molecules with a con- of PDGF-induced activation (ie ligand binding, receptor served sequence of roughly 100 amino acid residues contain- dimerization, and activation of the receptor kinase). An over- ing a motif of eight cysteine residues. There are three known view of these antagonists is depicted in Figure 3. Neutralizing existing isoforms, those being homo- and heterodimers of the antibodies against both the PDGF protein isomers and the A and B polypeptide chains.48 More recently, though, PDGF receptor proteins have been raised and tested in various additional PDGF isoforms have been discovered, known as animal systems.59–64 Nucleic acid ligands, known as aptamers, PDGF-C and D, respectively.49,50 can also be used to neutralize PDGF signaling. Aptamers are PDGF, like IGF-1, has been shown to activate both the Raf- generated by a process that selects for molecules with the MEK-ERK and PI3K pathways to induce cellular transform- highest binding capabilities over several rounds of binding to

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 489 the protein of choice. After numerous selective rounds, a high- of the IL-3R ␣-subunit allows for hematopoietic cellular pro- affinity molecule specific for the target is eventually isolated. liferation in suboptimal concentrations of the cytokine, IL-3.88 This process is commercially known as systematic evolution This observation has recently been supported by researchers of ligands by exponential enrichment (SELEX) and is currently who have found that the IL-3R ␣-subunit is a unique marker under extensive investigation. Recently, a group from Gilead for detection of acute myelogenous leukemia (AML) cells, as Sciences isolated an aptamer specific for PDGF that has an it is overexpressed in primitive leukemia cells.89 Conversely, ␤ astonishingly low equilibrium dissociation constant of less constitutive expression of the c chain of IL-3R or GM-CSFR than 3 × 10−10.65 mutants imparts acute, chronic myeloproliferative disorder(s) Scientists have also found that PDGF-A-mediated DNA syn- in murine hematopoietic cells.90–92 It is also interesting to note ␤ thesis can be halted by overexpressing a segment of the extra- that a truncated isoform of the c chain of the GM-CSF, IL-3, cellular portion of the PDGF receptor.66 These proteins have and IL-5 receptors has been found overexpressed in patients been shown to inhibit PDGF mainly by preventing autophos- with AML and ALL; this isoform appears to block signal trans- phorylation as described earlier. Furthermore, these com- duction in an unexplained manner resulting in malignant 93 pounds are able to block PDGF activity with IC50 values rang- transformation. ing from 100 nM up to 1 ␮M.67–69 The development of monoclonal antibodies specific for dis- As denoted previously, receptor dimerization is a critical tinct domains of the aforementioned receptors has shown step in transducing extracellular stimuli to the cell. This stage promise over the past few years. A monoclonal antibody has of mitogenic signal transduction also provides a potential for been developed that is specific for a region of the IL-3R.94 chemotherapeutic intervention. To illustrate this point, the use Shortly thereafter, monoclonal antibodies were generated to of monoclonal antibodies against specified domains of the target the IL-3R, but these also bound the GM-CSF and IL- PDGF receptor have been shown to significantly reduce and 5 receptors due to their high degree of homology on their ␤ 95 sometimes abolish PDGF-mediated signaling at IC50 values of c chains. under 1 nM.64 Furthermore, it was observed that antibodies The vast majority of the research done to antagonize the IL- directed at specific regions of the PDGF-R could inhibit 3 and GM-CSF receptors over the past few years has been in autophosphorylation without interfering with actual PDGF the area of toxic fusion proteins. DT388-GM-CSF, a fusion binding.64,70 protein derived from a truncated diphtheria toxin and GM- Ultimately, it is the kinase activity of the PDGF receptor CSF, has been shown to cause death in AML cells expressing that transduces the extracellular signal to activate intracellular the GM-CSF receptor.96 This effect was not evident in non- oncogenes. As such, several compounds have been developed transformed hematopoietic progenitor cells, thus providing a that specifically target the PDGF-R kinase. These tyrosine kin- rational chemotherapy for the treatment of AML. These experi- ase inhibitors are normally low molecular weight proteins that ments have been extended to an array of leukemic disorders, function to compete with ATP-binding at the kinase active including juvenile and chronic myelomonocytic leukemias, site. Over the past decade, a number of compounds have each of which is susceptible to DT388-GM-CSF.97,98 Nearly been developed including AG1296,71,72 CGP53716,73 STI- identical studies have been performed using the same trunc- 571 (also known as Gleevec, CGP57148),74,75 SU101,76 ated diphtheria toxin attached to IL-3. These studies have also Ki689677 and RPR101511A,78 each of which binds non-dis- yielded positive results with respect to purging leukemic cells criminately to the kinase(s) of the different PDGF receptor iso- from bone marrow grafts.99,100 forms. Each of the aforementioned inhibitors has been applied Recently, modified and chimeric cytokines have been to animal models with some success; but, by far, STI-571 has developed to aid in the repopulation of hematopoietic cell been pegged as the next miracle drug in leukemic ther- compartments following myelosuppressive chemotherapy. apies.79,80 Lastly, other similar compounds have been synthe- Daniplestim is an engineered IL-3 receptor agonist created by sized and used successfully in in vitro models, but have not truncations and amino acid substitutions to the native IL-3 yet been tested in animal models.81,82 protein. It can bind to the IL-3 receptor with significantly higher affinity than native IL-3.101,102 Myelopoietin-1 belongs to a family of engineered chimeric molecules with dual agon- IL-3 and GM-CSF receptors istic capabilities for the IL-3 and G-CSF receptors.101,102 When fused together to produce a chimeric cytokine, these proteins IL-3 and GM-CSF possess unique receptors, in that they lack have an enhanced ability to suppress apoptosis in hematopo- SPOTLIGHT intrinsic tyrosine kinase activity as compared to the PDGF-R ietic cells.101,102 Furthermore, chimeras between G-CSF and ␤ and IGF-R. To compensate for this shortcoming, the c chain FLT-3 ligand have also been developed and show potential of the receptor is able to recruit specific kinases in order to for preventing apoptosis in certain cells, as well as dendritic transduce extracellularly derived signals. One of the prototypi- cells.103,104 These types of studies should provide much ␤ cal kinases that serves this function is the JAK2 kinase. The c needed insight into post-chemotherapeutic care in leukemia chain is also able to attract the adaptor molecule, Shc, which patients. allows for the association of growth factor receptor-bound (Grb) protein and mammalian son of sevenless (SOS), each of which enables subsequent activation of the Ras oncoprotein. Target: Ras It is thought that an 85-kDa protein, identified as the p85 sub- unit of PI3K (described in more detail later) is also able to bind The 21-kDa Ras gene product is a monomeric membrane- ␤ to the c chain, thus enabling activation of the PI3K signal localized G-protein that initiates MAPK signal transduction. It transduction pathway.83 These biochemical events each lead is approximated that Ras is mutated in nearly 20% of all can- to DNA synthesis and prevention of apoptosis. cers.11 Within hematopoietic cancers, it has been seen that Hematopoietic cells can become transformed in response Ras is mutated in approximately 20% to 60% of AMLs.105–108 to mutation(s) occurring within the IL-3 gene which impart Because Ras is widely mutated in a variety of human malig- constitutive IL-3 expression.84–87 Furthermore, overexpression nancies and is a molecular switch in a network of signaling

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 490 pathways, it has become an important target for the develop- mediates proliferative signaling (Figure 4). Therefore, the ment of anticancer drugs. development of FT inhibitors is a viable strategy used to Ras is a membrane-bound guanosine triphosphate inhibit the oncogenic effects of Ras. At the time of publication, (GTP)/guanosine diphosphate (GDP)-binding (G) protein that there are currently four FTIs in clinical trials worldwide: acts to relay signals from the lipid-rich cellular membrane to R115777, SCH66336, L-778123 and BMS-214662.115 the nucleus. These signals lead to protein synthesis, regulation R115777 has undergone phase I clinical trials and was found of cell survival, cellular proliferation, and differentiation. After to significantly reduce Ras-mediated signaling.116,117 Similar binding of a ligand to its cognate receptor, dimerization of results were observed for the compound SCH66336.118 that receptor occurs, as described previously. Dimerization There are some data that argue that the effects of FTIs are causes the intracellular tyrosine kinase(s) to become phos- not only due to the inhibition of Ras farnesylation, but also phorylated, which then becomes bound by the SH2 domains due to the inhibition of farnesylation of other proteins essential 109 of the adaptor proteins, such as Grb2. Not only does Grb2 for cellular proliferation. RhoB is a 21-kDa G-protein that possess an SH2 domain, but it also has an SH3 domain that helps regulate receptor signaling and, in addition, is a target binds to proline-rich motifs of other peptides (ie son of seven- of FT inhibitors. Du and Prendergast119 showed that in the less (SOS)). SOS is a guanine dissociation stimulator of Ras presence of FTIs, RhoB became geranylgeranylated (RhoB- which binds to Ras after receptor activation. Ras’ native con- GG) in high quantities. At these concentrations, it was found formation is then compromised and GDP is dissociated from that RhoB-GG became toxic to malignant cells, but not nor- Ras, allowing for the subsequent association of GTP. In the mal cells. A second hypothesis dealing with the mechanism GTP-bound form, Ras is in its active state and able to recruit SPOTLIGHT of anti-tumor activity elicited by FTIs has also recently been Raf to the cell membrane, thus enabling activation of developed. Jiang et al120 have demonstrated that FT inhibitors Raf.110–113 However, there exist several GTPase-activating are able to inhibit PI3K-mediated signaling and induce proteins (GAPs) that are able to stimulate the GTPase activity apoptosis in cancer cells that overexpress the AKT2 isoform. of Ras, which convert it back to its original inactive, GDP- bound state.114 Supporting this hypothesis are data that indicate that FTIs are able to block a downstream kinase of PI3K, specifically There have been three main approaches taken to inhibit S6K 121 Ras-mediated signaling: (1) prevention of membrane localiz- p70 . ation of Ras; (2) inhibition of Ras protein expression via anti- Research has also been done to examine the effects of tar- sense nucleotides or RNAs; and (3) inhibition of kinases geting specific RNA sequences to inhibit translation of the Ras downstream of Ras (to be covered at each individual kinase RNA message into a functional protein. This antisense level). Each of these approaches has been successful, to vary- approach uses oligonucleotides, which are complementary to ing degrees, in blocking the proliferative effects of Ras mRNA transcripts of the Ras oncogene. These oligonucleo- activation. tides hybridize to the complementary mRNAs and inhibit their In order for Ras to become active, it must undergo post- subsequent translation into protein(s). Malignant phenotypes translational modifications that add farnesyl or geranylgeranyl have been observed to be suppressed in response to a retrovi- isoprenoid moieties to its carboxy-terminus. These modifi- ral vector-induced transduction of K-Ras antisense RNA of cations are generated by three different , namely far- human cancer cells.122,123 A second, similar approach has nesyltransferase (FT), geranyltransferase type I (GGTI), and also been used to inhibit abnormal Ras expression. This GGTII. By blocking the enzymatic action of these enzymes, method employs the use of plasmid vectors encoding anti- Ras is unable to localize to the cellular membrane where it sense RNAs that are able to inhibit tumorigenic growth.124

Figure 4 Activation of the Ras/Raf/MEK/ERK pathway by IL-3 and inhibition by farnesyltransferase (FT) inhibitors. (a) Activation of Ras/Raf/MEK/ERK by binding of IL-3 to its cognate receptor and subsequent signaling through coupling to Ras. (b) Uncoupling of the IL-3- mediated response by FT inhibitors which prevent the farnesylation of Ras and resultant targeting of Ras to the lipid-rich cell membrane.

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 491 Target: Raf Alternative strategies exist to inhibit Raf-mediated signaling. RNA aptamers have been developed that bind to the Ras-bind- Raf-1 was the first identified downstream effector kinase of ing domain of the Raf-1 isoform. These RNA molecules bind Ras11–113 and is currently the most characterized substrate of with fairly high dissociation constants (300 nM) and may be Ras. However, two other Raf isoforms are known to exist, able to inhibit Ras-Raf signaling in mammalian cells.154 those being A-Raf and B-Raf. This family of kinases has been Another approach has been to synthesize proteins that are suggested to have both overlapping and unique regulatory able to antagonize the binding between Ras and Raf.155 An functions.125–127 All three of the Raf isomers have been found additional method devised to inhibit Raf signaling is the devel- to be expressed in hematopoietic cells of either lymphoid or opment of antisense oligodeoxynucleotides that are specific myeloid lineages.23,128,129 The level of Raf expression in trans- for each isomer of Raf.156 Further research into these types of formed hematopoietic cells has been noted to be elevated.130–133 compounds has yielded marginal results, though.157 However, it is not known whether the heightened level of Raf It is of critical importance to mention that Ras also signals expression actually caused the cells to become transformed through parallel pathways in addition to the Raf-MEK-ERK because the cell lines of interest were established tumors. pathway. As such, chemotherapeutic intervention at any point Conversely, it is known that several mechanisms exist to elicit along the pathway may affect the signaling capacities of these dysregulated Raf expression. They include point mutations, alternative pathways. However, discussing the cellular rami- deletions, gene rearrangements and gene amplifications.134–138 fications of these types of interactions is outside the realm of Thus, the collective evidence suggests that Raf is a viable this review. anticancer target for chemotherapeutic intervention. Raf activation occurs on a dual level. In the first step, active GTP-bound Ras recruits Raf to the plasma membrane, which Target: alternative Raf kinases allows subsequent interaction(s) with additional activators that are not well-described.139–141 Though this step is necessary for Raf is activated by more than just Ras; the kinase suppressor activation of Raf, it is not sufficient to cause Raf activation. of Ras (KSR), PKC and the Src and JAK families of kinases Therefore, a secondary stage of activation occurs when Raf is have all been implicated in the Ras-independent activation phosphorylated on tyrosine and/or serine/threonine residue(s). of Raf.158–163 Each of these kinases activates Raf in a unique, This is normally accomplished on tyrosine residues by a mem- deliberate manner. Targeting their inactivation, collectively or ber of the Src family of kinases and on serine/threonine resi- individually, would result in decreased levels of Raf activity dues by (PKC) isoforms.142–144 It is worth- and the subsequent abrogation of unharnessed proliferation. while to note that the S43, S259 and S621 residues found on mutant, non-active Raf are still able to be phosphorylated, suggesting that these residues are not distinct autophosphoryl- ation sites.145 Lastly, dimerization of Raf is also known to JAK kinases induce Raf activity. Therefore, drugs which are able to prevent potential dimerization(s) may be effective anti-signaling The Janus family of kinases (JAKs) is a receptor-associated compounds.146 group of kinases that become phosphorylated in response to Recently, it was reported that Ras is able to induce the het- ligand-receptor binding. After dimerization of the receptor erodimerization of the Raf-1 and B-Raf isoforms.139 The role occurs, the JAKs become phosphorylated on tyrosine residues, that Raf heterodimers play in signal transduction is largely most likely a result of cross-phosphorylation. After they are unknown. As such, elucidation of the role that each of the Raf activated, the JAKs then are able to phosphorylate other tyro- oncogenes plays in tumor development is imperative for the sine residues on the receptor. The phosphorylated residues development of drugs that can effectively treat leukemic dis- then attract a variety of signaling molecules that each contain orders. This is an increasingly crucial issue because the differ- an SH2 domain which enables them to dock to the phos- ent Raf oncogenes have been shown to possess diverse sig- phorylation site. The signal transducers and activators of tran- naling properties in various cell types.147 For example, A-Raf scription (STATs) are a family of transcription factors that fall may promote cellular proliferation, whereas a different iso- into the aforementioned group of SH2-containing signaling form such as B-Raf induces apoptosis and arrests cell cycle molecules.164 When a STAT molecule binds to a phosphoryl- progression.23,148 Rationale mandates, then, that inhibitors to ated residue, it then becomes phosphorylated by a JAK due SPOTLIGHT each kinase must be developed in order to properly regulate to their close proximity. Phosphorylation allows the STATs to these kinases, as they may have opposing roles in either dimerize and enter the nucleus to act as transcription factors preventing or stimulating cellular proliferation. for genes that lead to altered phenotypes.165,166 Geldanamycin has been the mainstay with respect to Raf WHI-P131 is a compound that has been observed to target inhibition over the previous decade. In effect, though, geldan- JAK3 activity.167 JAK3 has been noted to have a role in lymph- amycin does not inhibit Raf activity, rather it destabilizes a oid development and function and thus is a viable candidate protein–protein interaction between Raf and heat shock pro- for chemotherapeutic intervention. WHI-P131 was able to tein (hsp) 90.149 By binding to hsp90, geldanamycin precludes induce apoptosis in the NALM-6 and LC1;19 human leukemia binding between Raf and hsp90 which ultimately prevents cell lines, suggesting that this compound may be an attractive proper Raf signaling functions.150 However, since many pro- candidate for future pharmaceutical endeavors.168 teins interact with hsp90, geldanamycin is not a specific Raf A tryphostin derivative that acts as a kinase inhibitor, AG- inhibitor.151 More recently, a second type of compound has 490, has also been used to inhibit JAK activity. It was first been shown to destabilize the Raf protein: KF25706 and observed in 1996 that this novel compound had apoptotic- KF58333 are oxime derivatives of radicol that have been inducing properties in ALL cells.169 Since then, AG-490 has shown to exhibit anti-tumor activities by acting in a manner been widely noted to act by inhibiting STAT3 activation and similar to geldanamycin.152,153 The chemical structures of inducing apoptosis in leukemic cells.170,171 As such, this com- these and other kinase inhibitors are presented in Figure 5. pound may be ideal for pharmacologically induced inhibition

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 492 SPOTLIGHT

Figure 5 Chemical structures of various signal transduction inhibitor molecules outlined in this review. (a) PDGF-R and BCR-Abl inhibitors; (b) Raf inhibitors; (c) JAK inhibitors; (d) PKC inhibitors; (e) MEK inhibitors.

of the JAK-STAT pathway and consequent inhibition of the folding of the MAPK proteins of the Raf-MEK-ERK pathway. It Raf-MEK-ERK cascade. has been noted that the kinase domain (CA5) of KSR is able to mediate direct interactions between both MEK1/2 and KSR.183 A similar type of binding interaction is seen with the Protein kinase C (PKC) ERK1/2 kinases; KSR binds to these proteins via a serine-thre- onine rich domain (CA4) located adjacent to the kinase PKC has been shown to be a potent activator of Raf-1.172 As domain.184 Interestingly, the interaction with ERK is depen- a result, blocking PKC has become another approach to halt dent on Ras activation, whereas the binding to MEK seems to Raf-MEK-ERK signaling in leukemic disorders. Staurosporine be constitutive.185,186 Raf-1/KSR interactions appear to be both is a non-specific protein kinase inhibitor that has been shown Ras-dependent and located at the cytoplasmic membrane.187 to halt cell cycle progression at concentrations ranging from These data, taken together, suggest a unique signaling mech- 1 to 100 nM.173 Staurosporine analogues exist, namely UCN- anism where KSR is able to promote the assembly on a multi- 101, which have a higher affinity for PKC isoforms.174,175 Fur- protein complex that brings MEK into close proximity with thermore, UCN-101 elicits both cell cycle inhibitory and anti- both its upstream activator, Raf, and its downstream substrate, proliferative effects in several tumor cell lines.176,177 Another ERK. Although much of the research done on KSR is relatively staurosporine derivative, CGP 41251, has been developed novel, one might hypothesize that inhibition of one or more and has also shown promise as a potential anti-tumor of these interactions may have anti-neoplastic ramifications. agent.178–180

Target: mitogen-activated protein kinase kinase-1/2 Kinase suppressor of Ras (KSR) (MEK-1/2)

The kinase suppressor of Ras (KSR, also known as CAPK) has Targeting the molecule directly downstream of Raf is another recently been identified to be a possible scaffolding protein possibility for the rational design of anticancer drugs. The for the entire Raf-MEK-ERK transduction cascade.181 It was MEKs belong to a multi-gene family that includes MEK-1 to originally reported that KSR was a positive effector of Ras sig- MEK-5. Of these isoforms, MEKs-3, 4 and 5 are relatively less naling that appeared to act either upstream of Raf or in a paral- characterized than either MEK-1 or MEK-2. The dearth of lel pathway, but the exact nature of these interactions was not research associated with the former proteins may be directly known.182 However, more recent studies have indicated that correlated to the lack of detectable kinase activity observed KSR may play an integral role in membrane-associated scaf- with the initial research that has been completed.188,189 Con-

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 493 versely, much research has been done on the MEK-1 and directly blame constitutive ERK activation for the onset of AML MEK-2 isoforms, some of which has shown that they exhibit and/or CML.224,225 Others have reported conflicting results, similar homologies to each other including a proline-rich suggesting that CML cell growth and differentiation are inde- region, which appears to be essential for signal transduction pendent of ERK activation.226 Another group has recently capabilities.190 Furthermore, MEK-1 and MEK-2 (hereafter shown that Ras-mediated ERK activation is absent in primary MEK), are phosphorylated on distinct serine residues, S218 AMLs, indicating that ERK is not an essential mediator of the and S222 by the upstream Raf molecules.191,192 Phosphoryl- development of leukemia(s).227 Crosstalk between parallel ation of MEK on both of these residues has been reported to pathways and/or the use of a variety of cell lines may help to increase its activity by more than 7000-fold.193 Moreover, explain the apparent contradiction in these reports. mammalian cells have been observed to be transformed as a One approach taken to inhibit ERK phosphorylation is result of constitutively activated MEK.194 Increased MEK through the generation of a competitive inhibitor. A 15-amino activity has also been seen in a variety of leukemias including acid peptide was synthesized resembling the ␣-C helix of ERK- AML and CML.195,196 Additionally, aberrant MEK-1 activity 1 and then kinetic analyses were performed to determine the has also been shown to have anti-apoptotic effects in hemato- efficacy of ERK inhibition mediated by this molecule. The poietic cells.197–200 In all, these data suggest that targeting authors found that this peptide was able to inhibit ERK from MEK with specific inhibitors may be sufficient to abrogate binding to MEK via a competitive inhibition mechanism.228 aberrant MAPK cell signaling. More recently, it was reported that ERK activation is inhibited PD098059 is a commercially available MEK inhibitor that by parallel activation of the stress-activated p38MAPK. Upon functions by binding to, and thus preventing activation of, the phosphorylation, p38MAPK binds to ERK, sequestering it away dephosphorylated form of MEK-1.201,202 Studies have shown from MEK-mediated activation, effectively inhibiting the Raf- that PD098059 is not only able to impair cellular proliferation, MEK-ERK signaling pathway at the level of the ERK protein.229 but is also able to impact other cellular events including angiogenesis, differentiation, and apoptosis.203–207 A second MEK inhibitor, U0126, was identified using a cell- Target: 90 kDa (p90RSK) based screen seeking inhibitors of phorbol ester-stimulated AP-1 transactivation.208 This inhibitor, like PD098059, does The p90RSK kinase is a serine-threonine kinase that contains not compete with ATP suggesting that its mechanism of action two functional kinase domains: an amino terminal kinase that is not by binding the ATP site of the . The basis for phosphorylates substrates of RSK and a carboxy terminal kin- the interactions that occur between these molecules is ase that enables the activation of RSK by ERK.230 Three iso- unknown due to the fact that there is a lack of any known forms of p90RSK are known to exist including RSK-1, RSK-2 crystal structure for either MEK-1 or MEK-2.193 and RSK-3, each of which exhibit approximately 75–80% Recently, it was reported that MEK inhibition via the use of to one another.231–233 p90RSK is fully acti- a novel inhibitor, PD184352, is able to suppress growth of vated after phosphorylation on three distinct residues which tumors in mice.209 Accordingly, this compound is presently requires three concerted steps. First, ERK phosphorylates undergoing phase I clinical trials in cancer patients.210 Inter- p90RSK on S369, which triggers phosphorylation of S386 by estingly, it has also been recently seen that the PKC inhibitor, the carboxy terminal kinase. Lastly, the amino terminal kinase UCN-01, when combined with PD184352 (or alternate MEK is phosphorylated on S227 by 3-phosphoinositide-dependent inhibitors, PD098059 and U0126) is able to cause mitochon- protein kinase-1 (PDK-1), which enhances RSK activity 100- drial dysfunction and subsequent induction of apoptosis.211 fold.234 It was later found that the S386 residue is located in Therefore, evidence suggests that targeting the MEK compo- a hydrophobic motif that acts as a docking site and is essential nent of the Raf-MEK-ERK signal transduction cascade may be for PDK-1-mediated phosphorylation of S227.235 This is an a viable option for chemotherapeutic intervention in apparent linker that bridges both the PI3K and Raf-MEK-ERK leukemic disorders. pathways, as PDK-1 also serves to phosphorylate and activate the downstream mediator of PI3K, Akt, on T308. This is an area of increasing interest and thus, this type of crosstalk Target: extracellular signal-regulated kinase (ERK) research merits further, more detailed investigations. The substrates of p90RSK include an array of transcription ERK-1 and ERK-2 are 43/44 and 41/42 kDa kinases that are factors including cAMP-response element-binding protein SPOTLIGHT directly downstream of the MEKs. Phosphorylation of the ERKs (CREB), estrogen receptor-␣ (ER-␣), I␬-B/NF-␬B, and c-Fos.236–240 mainly occurs as a result of growth factor/cytokine stimu- The roles that each of these transcription factors (and others) lation, but has also been shown to be activated by hydrogen play in the onset of leukemogenesis and cell cycle progression peroxide, UV light, and ionizing radiation.212–216 ERK-1 and will be briefly discussed in the following section. ERK-2 phosphorylation occurs on residues T183 and Y185 for maximal activity.6,7,188 When activated, ERK-2 is able to dimerize with other ERK-2 proteins (regardless of their acti- Target: nuclear transcription factors vation state) and induce nuclear translocation.217,218 Once inside the nucleus, ERK-2 is thought to phosphorylate and sub- Aberrant gene expression and the consequential development sequently activate a variety of nuclear targets including topiso- of neoplasia can be targeted at the transcriptional level by merase II-␣, a cell cycle progression mediator.219,220 The pri- examining the mechanisms by which these dysregulated mary cytoplasmic target of the ERK kinases is p90RSK, also genes become activated. Transcription factors (TFs) are the known as the ribosomal protein S6 kinase, but a wide array nuclear proteins that directly influence specific gene of other targets are known to exist.221–223 expression by stabilizing the interactions between the RNA Despite the abundance of information that directly corre- polymerase and the DNA template. In the unphosphorylated lates Ras-Raf-MEK activation with leukemogenesis, ERK acti- state, most TFs neither promote nor antagonize transcription vation is not as readily implicated in the process. Some reports of their respective genes. However, upon phosphorylation,

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 494 some TFs translocate from the cytosol to the nucleus so as to and ERK are able to phosphorylate c-Fos on a defined region activate transcription of their respective genes (Table 1). of its C-terminus, thereby enhancing the proliferation of Others, originating in the nucleus, merely localize to their fibroblastic cell lines.285,286 Also, activation of both the JNK respective sequence(s) on the DNA template in order to and MAPK pathways are sufficient to raise the protein levels enhance transcriptional efficiency. Research to antagonize and/or activity of c-Jun and c-Fos.287,288 Lastly, both p38MAPK these types of interactions between TFs and DNA is underway and JNK activation are necessary to elicit transformation and preliminary findings are revealing that targeting induced by c-Jun.289 TFs may be a sensible approach for the development of anti-neoplastic drugs. Nuclear factor-␬B

Cyclic AMP (cAMP)-response element binding protein NF-␬B is the generic name for a broad array of heterodimeric (CREB) transcription factors that are key modulators of a number of genes involved in cellular proliferation and apoptosis.290 NF- Activation of p90RSK can lead to phosphorylation of the CREB ␬B is found in the cytosol complexed with the inhibitory pro- transcription factor on S133.241,269,270 CREB, when phos- tein, I␬-B. A number of stimuli are able to induce phosphoryl- phorylated on this residue, is bound by the CREB-binding pro- ation of I-␬B, which allows for the dissociation of the com- tein (CBP) that enhances stabilization of the post-trans- plex, I-␬B degradation by the proteasome, and subsequent lationally altered CREB protein.271,272 Furthermore, this NF-␬B activation. Upon activation, NF-␬B is able to translo- SPOTLIGHT phosphorylation event is correlated with activation of immedi- cate to the nucleus to enhance transcription of a variety of ate–early genes that can cause tumorgenesis, such as c-fos.243 genes. Over 150 potential target genes have been identified for NF-␬B, a large proportion of which are cytokines required for immune responses.291 Activator protein-1 family of transcription factors (AP- The MAPKs can activate NF-␬B through several different 1) mechanisms. MEKK-1, a kinase of the JNK pathway, has been reported to phosphorylate I␬-B both in vitro and in vivo.292 The AP-1 family of transcription factors include c-Jun, c-Fos Raf-1 and p90RSK have also been shown to be I␬-B kinases, and activating transcription factor-2 (ATF-2).273 These compo- thereby inducing NF-␬B activation.293–296 Furthermore, the nents are able to dimerize with each other via leucine zipper constitutively active RelA/p65 subunit of the NF-␬B protein domains in order to activate transcription of specific genes.274 has been isolated from adenocarcinomas. This activated tran- c-Jun is phosphorylated on serines 63 and 73 by JNK and is scriptional activator was found to be phosphorylated by phosphorylated on C-terminal inhibitory sites by ERK1/2.275– MAPKs of the Raf/MEK/ERK pathway.297 279 Phosphorylation of the N-terminal amino acids (S63/S73) leads to enhanced c-Jun activity, which is coordinated with Ha-Ras-mediated oncogenic transformation.280 c-Myc The AP-1 transcription factors are defined targets of the MAPKs. For example, Jun:Fos and Jun:ATF dimers have been The c-Myc oncogene product is a transcription factor that is implicated in Ras-mediated oncogenesis.281 Moreover, both able to regulate gene expression in both normal and neoplas- c-Jun and Fos are necessary for cellular transformation tic cells.298–300 c-Myc is known to heterodimerize with two induced by Ras.282–284 Evidence exists suggesting that p90RSK proteins: Mad and Max.301 These proteins, when complexed,

Table 1 Common transcription factors of the RAf/MEK/ERK pathway

Name Consensus sequence Target gene Gene product

CREB241 TGACGTCA242 c-fos243 Transcription factor egr-1244 Transcription factor bcl-2245 Anti-apoptotic factor c-Myc246 CACGTG247 C/EBP␣248 Transcription factor Cyclin D1249 Cell cycle regulator p53250 Tumor suppressor eIF-4E251 Translation initiation factor ODC251 Cell cycle regulator Ets253 C/AGGAA/T253 u/PA254 Angiogenic factor MMP1255 Angiogenic factor MET256 Oncogene PTHrP257 Tumor regulator AP-1258 TGACTCA259 HB-EGF260 Growth factor SSeCKS261 Cell cycle regulator SPARC262 Oncogenic factor

263 264 265 NF-␬B GGGA/GNT/ATTCC BCL-XL Anti-apoptotic factor GM-CSF266 Growth factor c-Myc267 Transcription factor Cyclin D1268 Cell cycle regulator

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 495 have diverse cellular effects. Myc:Max heterodimers activate activation is dependent on the delicate balance between the transcription, while Mad:Max complexes repress gene activating signal(s) and inactivation mechanism(s). As pre- expression from the same promoter.302,303 Recently, another viously mentioned, ERK is fully activated by phosphorylation c-Myc binding protein was discovered that possessed tumor- on both a tyrosine and a threonine residue. Removing a phos- suppressor characteristics. This peptide, MM-1, complexes phate through the action of phosphatases from either of these with c-Myc’s transactivation domain to suppress its regulat- residues results in decreased ERK activity. The predominant ory capacities.304,305 problem with phosphatase research to date has been the Activated ERK dimers are c-Myc kinases that traverse the determination of their cellular localization. Consequently, nuclear membrane to phosphorylate c-Myc on S62, thus sti- there is little evidence that unequivocably implicates any mulating activity of the transactivation domain.306,307 In con- particular phosphatase with any definite kinase. However, trast, another group reported that although MAPK phosphoryl- research into the actions and modalities of these negative ation of c-Myc occurs in vitro, the actual in vivo kinase(s) of regulators is underway and thus, is presented hereafter. c-Myc are still unknown.308 Upstream of ERK, Ras is a There is an array of dual-specificity phosphatases known as mediator of c-Myc activation as it increases c-Myc transactiv- MAP kinase phosphatases (MKPs) that serve to dephosphoryl- ation over five-fold by interacting with the N-terminal region, ate ERK.320,321 At the time of publication, there are seven perhaps by stabilizing the activated protein.309,310 In addition, known MKPs (MKPs1–7). MKP-1 is the original family mem- a Raf-1 mutant which blocked Ras signaling caused by down- ber and was the first ERK phosphatase described.322 MKP-2 regulation of c-Myc expression directly implicates the MAPK (also known as TYP-1) was later isolated and also found to be cascade in c-Myc regulation.311 Interestingly, the stress-acti- an effective regulator of ERK activity.323,324 MKP-3 and MKP- vated pathway, headed by the JNK kinase(s), has also been 4 were each soon described as ERK phosphatases.325–327 The implicated in c-Myc activation, but in these cases, c-Myc acti- isolation of MKP-5 was the first phosphatase to be isolated vation can lead to apoptosis.312 Together these data suggest that was not effective in inhibiting ERK activity. Rather, MKP- that a degree of interplay can occur, perhaps mediated by dis- 5 was observed to inactivate the stress-activated kinases, JNK tinct extracellular stimuli, that ultimately leads to either cell and p38MAPK.328,329 Most recently, MKP-7 has been isolated death or cellular proliferation by c-Myc. and found to be similar in function to that of MKP-5.330 Lastly, MKP-6 is unique in that it functions as a novel negative-feed- back regulator of CD28 costimulatory signaling that controls Ets family of transcription factors the activation of MAP kinases in peripheral blood T cells.331 As previously alluded to, each of these phosphatases have dis- The Ets family of transcription factors has been implicated as tinct subcellular locales which imply the varying functions of one of the many downstream targets of the MAPK family these relatively homologous proteins. The utilization (ie (p38MAPK, JNK and ERK), as well as the PI3K pathway.313 This overexpression) of one or several of the previously mentioned group of TFs is characteristically defined by a conserved phosphatases in a pharmaco-preventative nature may help winged helix–turn–helix DNA binding motif.314 Current litera- inhibit MAPK signaling and thus, halt uncontrolled cellular ture suggests that the Ets TFs are responsible for targeting over proliferation. 200 different genes.315 The targeted gene is a function of the Of the above MKPs, two have been extensively reported to protein heterodimers that occur as a result of activation. Two be effective in the dephosphorylation and subsequent inacti- major groups of these TFs are currently defined: the Ets group vation of ERK molecules: MKP-1 and MKP-3. Activation of the which includes Ets1, Ets2 and Pointed, and also the TCF Raf/MEK/ERK cascade has been reported to be of instrumental (ternary complex factors) family which include Elk1, Sap1a, importance in the induction of MKP-1 expression to dephos- Sap1b, Fli1 and Net.316 phorylate ERK, which denotes the presence of an inhibitory The Ets subgroup has only one phosphorylation site located feedback loop.332 Further studies have endorsed this theory near the N-terminal domain, which is primarily activated by by showing that overexpression of MKP-1 is sufficient to the Ras/Raf/MEK/ERK family.317 The TCFs, though, are unique increase both Raf and MEK kinase activities.333 MKP-3 is in that they have multiple serines and threonines within a another phosphatase that has been shown to dephosphorylate transactivation domain that can be phosphorylated for acti- ERK. MKP-3 is activated merely by binding to ERK and func- vation.318 This phosphorylation event is sufficient to allow for tions primarily in the cytoplasm, whereas MKP-1 expression

efficient binding to Ras-responsive elements (RREs) and/or SPOTLIGHT is reserved for nuclear activities.334,335 serum response elements (SSEs) that exist in the promoters of Protein phosphatases-1 and 2a (PP1 and PP2a) are a duo of multiple immediate–early genes, including c-fos, egr-1 and dephosphorylating agents with highly conserved amino-acid junB.319 RREs require the binding of both an AP-1 dimer sequences that remove phosphate groups from a wide array (previously discussed) and an Ets component. Therefore, gene of phosphoproteins.336,337 It has been seen that these two expression controlled by these promoter sequences is phosphatases have an affinity for interaction with each of the immeasurably complex, considering that over 25 Ets members components of the MAPK pathway (Raf, MEK and ERK).335–340 exist and that both the AP1 and Ets regulation are controlled The role that these phosphatases play in regulating each of coordinately by Ras.316 The multifaceted and interwoven nat- these proteins may be quite similar. One report showed that ure of the signaling pathways combined with the number of PP2a inactivation and MAPK activation are strongly corre- unique TF heterodimers that can occur help explain why these lated, indicating that PP2a plays a negative modulatory role transcriptional activators and their mode(s) of action are not in the regulation of MAP kinase activity.341 Other reports, well understood. dealing with the Raf kinase, state that PP1 and PP2a dephos- phorylate phospho-Raf, when it is bound to the 14-3-3 pro- Negative regulation of MAPKs: phosphatases teins, in order to allow for subsequent Ras-mediated activation of Raf.342,343 In each case, PP1 and PP2a activity permit the The premise of this review has been based on phosphoregul- initiation of additional MAPK signaling. In addition, okadaic ation of signaling cascades. The duration and degree of MAPK acid has been noted to be a potent inhibitor of both PP1 and

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 496 PP2a phosphatase activities, albeit through a currently unde- PDKs and Akt are not brought into close proximity to each scribed mechanism.336,344 The cellular phosphatases that act other. Thus, activation of Akt is minimal, if at all. The theory upon p90RSK have not been identified as of yet. that presence of a functional PTEN phosphatase is essential for the prevention of tumorigenesis is endorsed by data that showed that PTEN−/− mice either die during embryogenesis or Crosstalk: MAPK interactions with PI3K/Akt spontaneously develop a variety of tumors later in life.363,364 Two pharmacological inhibitors of PI3K have been used As alluded to previously, the PI3K/Akt pathway is one that extensively over the past decade. Wortmannin, originally often transduces signals that are similar in nature to that of known as an inhibitor of respiratory burst activities, has been the Raf/MEK/ERK pathway. It has been shown that a certain shown to be a potent inhibitor of PI3K-mediated sig- degree of crosstalk exists between these pathways, which naling.365,366 LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1- either enhance or sometimes negate oncogenic extracellular benzopyran-4-one] is another inhibitor of PI3K that has been ␮ 367 signals. By understanding the interactions that occur between used with high degrees of efficacy at low IC50s (1.4 M). the components of these pathways, we will be better advised The two aforementioned inhibitors have been useful in block- in the development of treatment strategies that are able to ing PI3K signaling, but have no capacity to directly inhibit Akt more efficiently target leukemic cells within the body. activity. More recently though, CalBioChem (San Diego, CA, Phosphatidylinositol 3-kinase (PI3K) is located in the cyto- USA) has developed a highly potent, quasi-specific inhibitor plasm in resting cells. It exists as a dimer of two subunits: a of Akt known as IL-6-hydroxymethyl-chiro-inositol-2(R)-2-O- regulatory p85 subunit combined with a catalytic p110 methyl-3-O-octadecylcarbonate. This compound has been SPOTLIGHT 345,346 component. Upon activation of a receptor tyrosine kin- seen to selectively inhibit Akt activity with an IC50 of approxi- ase and subsequent autophosphorylation, the SH-2 domain(s) mately 5.0 ␮M, while only faintly affecting PI3K activity.368 of the p85 regulatory subunit bind to the receptor’s phos- Further studies using this inhibitor should provide valuable phorylated Y residues.83 PI3K then generates a series of phos- insight into Akt-mediated signaling, especially that inde- phorylated lipids at the cytoplasmic membrane that act to pendent of PI3K. recruit the phosphatidylinositol-dependent kinases (PDKs) and Crosstalk between the PI3K and Raf/MEK/ERK pathways has their downstream substrate Akt/PKB to the membrane.347,348 been reported on multiple levels, with some research stating PDK1 serves to phosphorylate Akt on T308, whereas PDK2 that PI3K activity is essential for induction of Raf/MEK/ERK phosphorylates S473 of Akt.349–352 Activated Akt is then able activity.369–371 Additional studies suggest that the PI3K path- to phosphorylate a variety of downstream substrates each of way enhances and/or synergizes with Raf/MEK/ERK signaling which enable the cell to proliferate including the pro-apop- to provide a more robust signal through the lower components totic molecule BAD, caspase-9 (human cell lines), the for- of the MAPK cascade (ie ERK).47,371–373 However, there is con- khead family transcription factors, and I-K, a kinase that regu- flicting evidence that states that Akt is able to phosphorylate lates the NF-B transcription factor.352–357 An overview of the Raf, thereby efficiently abrogating Raf activity on downstream potential interactions within this pathway are depicted in substrates.374–376 Furthermore, some data suggest that this Figure 6. effect may be dependent upon the differentiation state of the Abrogation of oncogenic signals from the PI3K pathway cell, which may account for the apparent discrepancies in the occurs via the tumor suppressor gene product, PTEN.358 PTEN current literature.377 Regardless of cell type, differentiation is a phosphatase that acts by removing a phosphate group state, or assay conditions, it is quite apparent that there is a

from the 3 position of the inositol ring of the PIP3,4,5 phosphol- varying degree of crosstalk that occurs between these path- ipids located at the cellular membrane.359–362 In doing so, the ways. Examining the level, nature and circumstances by which these interactions occur will be instrumental in the development of new agents designed to battle leukemia.

Conclusions

The Raf/MEK/ERK signal transduction cascade is responsible for many of the phenotypes observed in a wide array of oncogenic disorders, including leukemia. Drug intervention and/or the use of pharmacological inhibitors will be practical at each level of this cascade, from the cell receptor at the plasma membrane to the transcription factors at the nucleus. This review would be remiss if it did not address the impor- tance of acknowledging the complexities of the signal trans- duction and apoptotic pathways discussed (a more detailed, but not all-encompassing schematic is shown in Figure 7). All conclusions drawn are not necessarily true for all cell types and the degree of crosstalk that occurs between parallel path- ways remains largely unknown.378,379 Furthermore, blocking the activity of a protein far upstream of others (ie Ras) may have other injurious effects that were not intended, as inhibiting its activity may also suppress other vital functions. Conversely, by inhibiting the activity of a downstream compo- RSK Figure 6 Roles of the PI3K/Akt pathway in growth and the preven- nent of the MAPK pathway (ie p90 ), one may overlook tion of apoptosis. alternative mechanisms by which the cell is able to bypass

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 497

Figure 7 Sites of interaction of various inhibitors of cytokine and stress-mediated signal transduction. that signaling component. For that reason, a multi-tiered GD. ERKs: a family of protein-serine/threonine kinases that are approach targeting the inhibition of uncontrolled cell activated and tyrosine phosphorylated in response to insulin and signaling may be merited. NGF. Cell 1991; 65: 663–675. 4 Rossomando AJ, Payne DM, Weber MJ, Sturgill TW. Evidence that The hallmark of every neoplasia is uncontrolled cellular pp42, a major tyrosine kinase target protein, is a mitogen-activated proliferation. This aberrant behavior relies on a series of over- serine/threonine protein kinase. Proc Natl Acad Sci USA 1989; 86: expressed oncogenes and on the accumulation of tumor-sup- 6940–6943. pressor gene mutations. The combinatorial effects of these 5 Payne DM, Rossomando AJ, Martino P, Erickson AK, Her JH, Shab- genotypes are detrimental not only to the cell, but also to the anowitz J, Hunt DF, Weber MJ, Sturgill TW. Identification of the entire body. If the cellular processes by which these anomalies regulatory phosphorylation sites in pp42/mitogen-activated pro- occur are better understood, then scientists can begin to tein kinase (MAP kinase). EMBO J 1991; 10: 885–892. 6 Seger R, Seger D, Lozeman FJ, Ahn NG, Graves LM, Campbell exploit that knowledge so as to develop more effective, JS, Ericsson L, Harrylock M, Jensen AM, Krebs EG. Human T-cell non-invasive chemotherapeutic treatments. mitogen-activated protein kinase kinases are related to yeast signal Transduction kinases. J Biol Chem 1992; 267: 25628–25631. 7 Crews C, Alesandrini A, Erikson R. The primary structure of MEK, a Acknowledgements protein kinase that phosphorylates the ERK gene product. Science 1992; 258: 478–480. SPOTLIGHT This work was supported in part by RO1CA51025 from the 8 Kosako H, Gotoh Y, Matsuda S, Ishikawa M, Nishida E. Xenopus MAP kinase activator is a serine/threonine/tyrosine kinase acti- National Cancer Institute and 2000-ARG-0003 from the North vated by threonine phosphorylation. EMBO J 1992; 11: 2903– Carolina Biotechnology Center. We wish to thank Ms Cather- 2908. ine Spruill for the excellent artwork. 9 Kyriakis JM, App H, Zhang XF, Banerjee P, Brautigan DL, Rapp UR, Avruch J. Raf-1 activates MAP kinase-kinase. Nature 1992; 358: 417–421. References 10 Dent P, Haser W, Haystead TAJ, Vincent LA, Roberts TM, Sturgill TW. Activation of mitogen-activated protein kinase kinase by v- 1 Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, Raf in NIH 3T3 cells and in vitro. Science 1992; 257: 1404–1407. Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S, Sawyers CL. 11 Khleif SN, Abrams SI, Hamilton JM, Bergmann-Leitner E, Chen A, Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine Bastian A, Bernstein S, Chung Y, Allegra CH, Schlom J. A phase kinase in chronic myeloid leukemia. N Engl J Med 2001; 344: I vaccine trial with peptides reflecting ras oncogene mutations of 1031–1037. solid tumors. J Immunother 1999; 22: 155–165. 2 Boulton TG, Yancopoulos GD, Gregory JS, Slaughter C, Moomaw 12 McCubrey JA, May WS, Duronio V, Mufson A. Serine/Threonine C, Hsu J, Cobb MH. An insulin-stimulated protein kinase similar phosphorylation in cytokine signal transduction. Leukemia 2000; to yeast kinases involved in cell cycle control. Science 1990; 249: 14: 9–21. 64–67. 13 New L, Han J. The p38 MAP kinase pathway and its biological 3 Boulton TG, Nye SH, Robbins DJ, Ip NY, Radziejewska E, Morgen- function. Trends Cardiovasc Med 1998; 8: 220–228. besser SD, DePinho RA, Ranayotatos N, Cobb MH, Yancopoulos 14 Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumary S, Green

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 498 D, McNulty D, Blummenthal MJ, Heys JR, Landvatter SW, Strickler lin-like growth factor-I and insulin-like growth factor binding pro- JE, McLaughlin MM, Siemens IR, Fisher SM, Livi GP, White JR, tein-3 during maintenance chemotherapy of acute lymphoblastic Adams JL, Young PR. A protein kinase involved in the regulation of leukemia in children. J Pediatr Hematol Oncol 1999; 21:268– inflammatory cytokine biosynthesis. Nature 1994; 372: 739–746. 273. 15 Jiang Y, Chen C, Li Z, Guo W, Gegner JA, Lin S, Han J. Charac- 36 Arguelles B, Barrios V, Pozo J, Munoz MT, Argente J. Modifi- terization of the structure and function of a new mitogen-activated cations of growth velocity and the insulin-like growth factor sys- protein kinase (p38␤). J Biol Chem 1996; 271: 17920–17926. tem in children with acute lymphoblastic leukemia: a longitudinal 16 Jiang Y, Gram H, Zhao M, New L, Gu J, Feng L, Di Padova F, study. J Clin Endocrinol Metab 2001; 85: 4087–4092. Ulevitch RG, Han J. Characterization of the structure and function 37 Valertinis B, Baserga R. IGF-1 receptor signaling in transformation of the fourth member of p38 group mitogen-activated protein kin- and differentiation. Mol Pathol 2001; 54: 133–137. ases, p38. J Biol Chem 1997; 272: 30122–30128. 38 Peruzzi F, Prisco M, Dews M, Salomoni P, Grassilli E, Romano 17 Lechner C, Zahalka MA, Giot JF, Moller NP, Ullrich A. ERK6, a G, Calabretta B, Baserga R. Multiple signaling pathways of the mitogen-activated protein kinase involved in C2C12 myoblast dif- insulin-like growth factor 1 receptor in protection from apoptosis. ferentiation. Proc Natl Acad Sci USA 1996; 93: 4355–4359. Mol Cell Biol 1999; 19: 7203–7215. 18 Enslen H, Brancho DM, Davis RJ. Molecular determinants that 39 Navarro M, Baserga R. Limited redundancy of survival signals mediate selective activation of p38 MAP kinase isoforms. EMBO from the type I insulin-like growth factor receptor. Endocrinology J 2000; 19: 1301–1311. 2001; 142: 1073–1081. 19 Sanchez I, Hughes RT, Mayer BJ, Yee K, Woodgett JR, Avruch J, 40 Kellerer M, Obermaier-Kusser B, Ermel B, Wallner U, Haring HU, Kyriakis J, Zon LI. Role of SAPK/ERK kinase-1 in the stress-acti- Petrides PE. An altered IGF-1 receptor is present in human leu- vated pathway regulating transcription factor c-Jun. Nature 1994; kemic cells. J Biol Chem 1990; 265: 9340–9345. 372: 794–798. 41 Baier TG, Jenne EW, Blum W, Schonberg D, Hartmann KK. Influ-

SPOTLIGHT 20 Derijard B, Rainegeaud J, Barrett T, Wu IH, Han J, Ulevitch RJ, ence of antibodies against IGF-1, insulin, or their receptors on Davis RJ. Independent human MAP-kinase signal transduction proliferation of human acute lymphoblastic leukemia cell lines. pathways defined by MEK and MKK isoforms. Science 1995; 267: Leuk Res 1992; 16: 807–814. 682–685. 42 Sinclair J, McClain D, Taetle R. Effects of insulin and insulin-like 21 Moriguchi T, Toyoshima F, Masuyama N, Hanafusa H, Gotoh Y, growth factor I on growth of human leukemia cells in serum-free Nishida E. A novel SAPK/JNK kinase, MKK7, stimulated by TNFal- and protein-free medium. Blood 1988; 71: 66–72. pha and cellular stresses. EMBO J 1997; 16: 7045–7053. 43 McCubrey JA, Steelman LS, Mayo MW, Algate PA, Dellow RA, 22 Ichijo H. From receptors to stres-activated MAP kinases. Oncog- Kaleko M. Growth-promoting effects of insulin-like growth factor- ene 1999; 18: 6087–6093. 1 (IGF-1) on hematopoietic cells: overexpression of introduced 23 Blalock WL, Weinstein-Oppenheimer CR, Chang F, Hoyle PE, IGF-1 receptor abrogates interleukin-3 dependency of murine fac- Wang XY, Algate PA, Franklin RA, Oberhaus SM, Steelman LS, tor-dependent cells by a ligand-dependent mechanism. Blood McCubrey JA. Signal transduction, cell cycle regulatory, and anti- 1991; 78: 921–929. apoptotic pathways regulated by IL-3 in hematopoietic cells: poss- 44 Oliveira MR, Ohnuma L, Bendit I, Dorlhiac-Lacer P, Giannella- ible sites for intervention with anti-neoplastic drugs. Leukemia Neto D. Interferon-alpha therapy increases type I insulin-like 1999; 14: 1109–1166. growth factor receptors expression on lymphoid cells from patients 24 Okuda K, Foster R, Griffin J. Signaling domains of the ␤c chain with chronic myelogenous leukemia. Leuk Res 2001; 25:711– of the GM-CSF/IL-3/IL-5 receptor. Ann NY Acad Sci 1999; 872: 717. 305–312. 45 Dudek H, Datta SR, Franke TF, Birnbaum MJ, Yao R, Cooper GM, 25 Hackel PO, Zwick E, Prenzel N, Ullrich A. Epidermal growth fac- Segal RA, Kaplan DR, Greenberg ME. Regulation of neuronal sur- tor receptors: critical mediators of multiple receptor pathways. vival by the serine-threonine protein kinase Akt. Science 1997; Curr Opin Cell Biol 1999; 11: 184–189. 275: 295–299. 26 Emaduddin M, Ekman S, Ronnstrand L, Heldin CH. Functional 46 Kauffmann-Zeh A, Rodriguez-Viciana P, Ulrich E, Gilbert C, Cof- cooperation between the subunits in heterodimeric platelet- fer P, Downward J, Evan G. Suppression of c-Myc-induced derived growth factor receptor complexes. Biochem J 1999; 341: apoptosis by Ras signalling through PI(3)K and PKB. Nature 1997; 523–528. 385: 544–548. 27 Wells A. EGF Receptor. Int J Biochem Cell Biol 1999; 31: 637– 47 McCubrey JA, Steelman LS, Blalock WL, Lee JT, Moye PW, Chang 643. F, Pearce M, Shelton JG, White MK, Franklin RA, Pohnert SC. Syn- 28 Heldin CH, Westermark B. Mechanism of action and in vivo role ergistic effects of PI3K/Akt on abrogation of cytokine-dependency of platelet-derived growth factor. Physiol Rev 1999; 79: 1283– induced by oncogenic Raf. Adv Enz Reg 2001; 41: 289–323. 1316. 48 Heldin CH, Westermark B. Mechanism of action and in vivo role 29 Khazaie K, Schirrmacher V, Lichtner RB. EGF receptor in neo- of platelet-derived growth factor. Physiol Rev 1999; 79: 1283– plasia and metastasis. Cancer Metas Rev 1993; 12: 255–274. 1316. 30 Oesterreich S, Zhang P, Guler RL, Sun X, Curran EM, Welshons 49 Li X, Ponten A, Aase K, Karlsson L, Abramsson A, Uutela M, Back- WV, Osborne CK, Lee AV. Re-expression of estrogen receptor strom G, Hellstrom M, Bostrom H, Li H, Soriano P, Betsholtz C, alpha in estrogen receptor alpha-negative MCF-7 cells restores Heldin CH, Alitalo K, Ostman A, Eriksson U. PDGF-C is a new both estrogen and insulin-like growth factor-mediated signaling protease-activated ligand for the PDGF alpha-receptor. Nat Cell and growth. Cancer Res 2001; 15: 5771–5777. Biol 2000; 2: 302–309. 31 Wang JY, Del Valle L, Gordon J, Rubini M, Romano G, Croul 50 LaRochelle WJ, Jeffers M, McDonald WF, Chillakuru RA, Giese S, Peruzzi F, Khalili K, Reiss K. Activation of the IGF-IR system NA, Lokker NA, Sullivan C, Boldog FL, Yang M, Vernet C, Burgess contributes to malignant growth of human and mouse medullobla- CE, Fernandes E, Deegler LL, Rittman B, Shimkets J, Shimkets RA, stomas. Oncogene 2001; 29: 3857–3868. Rothberg JM, Lichenstein HS. PDGF-D, a new protease-activated 32 Benini S, Manara MC, Baldini N, Cerisano V, Serra M, Mercuri growth factor. Nat Cell Biol 2001; 3: 517–521. M, Lollini PL, Nanni P, Picci P, Scotlandi K. Inhibition of insulin- 51 Jones SM, Klinghoffer R, Prestwich GD, Toker A, Kazlauskas A. like growth factor I receptor increases the antitumor activity of PDGF induces an early and a late wave of PI 3-kinase activity, doxorubicin and vincristine against Ewing’s sarcoma cells. Clin and only the late wave is required for progression through G1. Cancer Res 2001; 6: 1790–1797. Curr Biol 1999; 9: 512–521. 33 Hoog A, Kjellman M, Nordqvist AC, Hoog CM, Juhlin C, Falkmer 52 Bos JL. Ras-like GTPases. Biochim Biophys Acta 1997; 1333: S, Schalling M, Grimelius L. Insulin-like growth factor-II in endo- M19–M31. crine pancreatic tumours. Immunohistochemical, biochemical 53 Carroll M, Tomasson MH, Barker GF, Golub TR, Gilliland DG. and in situ hybridization findings. APMIS 2001; 109: 127–140. The TEL/platelet-derived growth factor beta receptor (PDGF beta 34 MerchavS. The haematopoietic effects of growth hormone and R) fusion in chronic myelomonocytic leukemia is a transforming insulin-like growth factor-I. J Pediatr Endocrinol Metab 1998; 11: protein that self-associates and activates PDGF beta R kinase- 677–685. dependent signaling pathways. Proc Natl Acad Sci USA 1996; 93: 35 Schmiegelow M, Hertz H, Schmiegelow K, Holm K, Muller J. Insu- 14845–14850.

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 499 54 Golub TR, Barker GF, Lovett M, Gilliland DG. Fusion of PDGF broadly active protein tyrosine kinase inhibitor. J Pharmacol Exp receptor beta to a novel ets-like gene, tel, in chronic myelomono- Ther 1997; 283: 1433–1444. cytic leukemia with t(5;12) chromosomal translocation. Cell 1994; 70 Omura T, Miyazawa K, Ostman A, Heldin CH. Identification of a 77: 307–316. 190-kDa vascular endothelial growth factor 165 cell surface bind- 55 Sjoblom T, Boureux A, Ronnstrand L, Heldin CH, Ghysdael J, Ost- ing protein on a human glioma cell line. J Biol Chem 1997; 272: man A. Characterization of the chronic myelomonocytic leukemia 12676–12682. associated TEL-PDGF beta R fusion protein. Oncogene 1999; 18: 71 Kovalenko M, Gazit A, Bohmer A, Rorsman C, Ronnstrand L, Hel- 7055–7062. din CH, Waltenberger J, Bohmer FD, Levitzki A. Selective platelet- 56 Lacronique V, Boureux A, Valle VD, Poirel H, Quang CT, Mau- derived growth factor receptor kinase blockers reverse sis-trans- chauffe M, Berthou C, Lessard M, Berger R, Ghysdael J, Bernard formation. Cancer Res 1994; 54: 6106–6114. OA. A TEL-JAK2 fusion protein with constitutive kinase activity in 72 Kovalenko M, Ronnstrand L, Heldin CH, Loubtchenkov M, Gazit human leukemia. Science 1997; 278: 1309–1312. A, Levitzki A, Bohmer FD. Phosphorylation site-specific inhibition 57 Kimura A, Nakata Y, Hyodo H, Kuramoto A, Satow Y. Platelet- of platelet-derived growth factor beta-receptor autophosphoryl- derived growth factor expression in accelerated and blastic phase ation by the receptor blocking tyrphostin AG1296. Biochemistry of chronic myelogenous leukaemia with myelofibrosis. Br J 1997; 36: 6260–6269. Haematol 1994; 86: 303–307. 73 Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, 58 Tsai LH, White L, Raines E, Ross R, Smith RG, Cushley W, Ozanne Regenass U, Lydon NB. Selective inhibition of the platelet-derived B. Expression of platelet-derived growth factor and its receptors growth factor signal transduction pathway by a protein-tyrosine by two pre-B acute lymphocytic leukemia cell lines. Blood 1994; kinase inhibitor of the 2-phenylaminopyrimidine class. Proc Natl 83: 51–55. Acad Sci USA 1995; 92: 2558–2562. 59 Ferns GA, Raines EW, Sprugel KH, Motani AS, Reidy MA, Ross 74 Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, Druker R. Inhibition of neointimal smooth muscle accumulation after BJ, Lydon NB. Inhibition of the Abl protein-tyrosine kinase in vitro angioplasty by an antibody to PDGF. Science 1991; 253: 1129– and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res 1132. 1996; 56: 100–104. 60 Rutherford C, Martin W, Salame M, Carrier M, Anggard E, Ferns 75 Carroll M, Ohno-Jones S, Tamura S, Buchdunger E, Zimmerman J, G. Substantial inhibition of neo-intimal response to balloon injury Lydon NB, Gilliland DG, Druker BJ. CGP57148, a tyrosine kinase in the rat carotid artery using a combination of antibodies to plate- inhibitor, inhibits the growth of cells expressing BCR-ABL, TEL- let-derived growth factor-BB and basic fibroblast growth factor. ABL, and TEL-PDGFR fusion proteins. Blood 1997; 90: 4947– Atherosclerosis 1997; 130: 45–51. 4952. 61 Vassbotn FS, Langeland N, Hagen I, Holmsen H. A monoclonal 76 Shawver LK, Schwartz DP, Mann E, Chen H, Tsai J, Chu L, Taylor- antibody against PDGF B-chain inhibits PDGF-induced DNA syn- son L, Longhi M, Meredith S, Germain L, Jacobs JS, Tang C, Ullrich thesis in C3H fibroblasts and prevents binding of PDGF to its A, Berens ME, Hersh E, McMahon G, Hirth KP, Powell TJ. Inhi- receptor. Biochim Biophys Acta 1990; 1054: 246–249. bition of platelet-derived growth factor-mediated signal transduc- 62 Koyama H, Nishizawa Y, Hosoi M, Fukumoto S, Kogawa K, Shioi tion and tumor growth by N-[4-(trifluoromethyl)-phenyl]5-methyl- A, Morii H. The fumagillin analogue TNP-470 inhibits DNA syn- isoxazole-4-carboxamide. Clin Cancer Res 1997; 3: 1167–1177. thesis of vascular smooth muscle cells stimulated by platelet- 77 Yagi M, Kato S, Kobayashi Y, Kobayashi N, Iinuma N, Nakamura derived growth factor and insulin-like growth factor-I. Possible K, Kubo K, Ohyama SI, Murooka H, Shimizu T, Nishitoba T, involvement of cyclin-dependent kinase 2. Circ Res 1996; 79: Osawa T, Nagano N. Beneficial effects of a novel inhibitor of plat- 757–764. elet-derived growth factor receptor autophosphorylation in the rat 63 LaRochelle WJ, Jensen RA, Heidaran MA, May-Siroff M, Wang with mesangial proliferative glomerulonephritis. Gen Pharmacol LM, Aaronson SA, Pierce JH. Inhibition of platelet-derived growth 1998; 31: 765–773. factor autocrine growth stimulation by a monoclonal antibody to 78 Bilder G, Wentz T, Leadley R, Amin D, Byan L, O’Conner B, the human alpha platelet-derived growth factor receptor. Cell Needle S, Galczenski H, Bostwick J, Kasiewski C, Myers M, Spada Growth Diff 1993; 4: 547–553. A, Merkel L, Ly C, Persons P, Page K, Perrone M, Dunwiddie C. 64 Lokker NA, O’Hare JP, Barsoumian A, Tomlinson JE, Ramakrish- Restenosis following angioplasty in the swine coronary artery is nan V, Fretto LJ, Giese NA. Functional importance of platelet- inhibited by an orally active PDGF-receptor tyrosine kinase inhibi- derived growth factor (PDGF) receptor extracellular immunoglob- tor, RPR101511A. Circulation 1999; 99: 3292–3299. ulin-like domains. Identification of PDGF binding site and neu- 79 Mauro MJ, Druker BJ. STI571: targeting BCR-ABL as therapy for tralizing monoclonal antibodies. J Biol Chem 1997; 272: CML. Oncologist 2001; 6: 230–232. 33037–33044. 80 Mauro MJ, Druker BJ. STI571: a gene product-targeted therapy for 65 Drolet DW, Jenison RD, Smith DE, Pratt D, Hicke BJ. A high leukemia. Curr Oncol Rep 2001; 3: 223–227. throughput platform for systematic evolution of ligands by 81 Wood JM, Bold G, Buchdunger E, Cozens R, Ferrari S, Frei J, Hof- exponential enrichment (SELEX). Comb Chem High Throughput mann F, Mestan J, Mett H, O’Reilly T, Persohn E, Rosel J, Schnell Screen 1999; 2: 271–278. C, Stover D, Theuer A, Towbin H, Wenger F, Woods-Cook K, 66 Duan DS, Pazin MJ, Fretto LJ, Williams LT. A functional soluble Menrad A, Siemeister G, Schirner M, Thierauch KH, Schneider SPOTLIGHT extracellular region of the platelet-derived growth factor (PDGF) MR, Drevs J, Martiny-Baron G, Totzke F. PTK787/ZK 222584, a beta-receptor antagonizes PDGF-stimulated responses. J Biol novel and potent inhibitor of vascular endothelial growth factor Chem 1991; 266: 413–418. receptor tyrosine kinases, impairs vascular endothelial growth fac- 67 Schroeder MC, Hamby JM, Connolly CJ, Grohar PJ, Winters RT, tor-induced responses and tumor growth after oral administration. Barvian MR, Moore CW, Boushelle SL, Crean SM, Kraker AJ, Dris- Cancer Res 2000; 60: 2178–2189. coll DL, Vincent PW, Elliott WL, Lu GH, Batley BL, Dahring TK, 82 Iwamoto H, Nakamuta M, Tada S, Sugimoto R, Enjoji M, Nawata Major TC, Panek RL, Doherty AM, Showalter HD. Soluble 2-sub- H. Platelet-derived growth factor receptor tyrosine kinase inhibitor stituted aminopyrido[2,3-d]pyrimidin-7-yl ureas. Structure– AG1295 attenuates rat hepatic stellate cell growth. J Lab Clin Med activity relationships against selected tyrosine kinases and explo- 2001; 135: 406–412. ration of in vitro and in vivo anticancer activity. J Med Chem 83 Shoelson SE, Sivaraja M, Williams KP, Hu P, Schlessinger J, Weiss 2001; 44: 1915–1926. MA. Specific phosphopeptide binding regulates a conformational 68 Palmer BD, Kraker AJ, Hartl BG, Panopoulos AD, Panek RL, Batley change in the PI 3-kinase SH2 domain associated with enzyme BL, Lu GH, Trumpp-Kallmeyer S, Hollis Showalter HD, Denny activation. EMBO J 1993; 12: 795–802. WA. Structure–activity relationships for 5-substituted 1-phenyl- 84 Wang XY, McCubrey JA. Malignant transformation induced by benzimidazoles as selective inhibitors of the platelet-derived cytokine genes: a comparison of the abilities of germline and growth factor receptor. J Med Chem 1999; 42: 2373–2382. mutated interleukin 3 genes to transform hematopoietic cells by 69 Panek RL, Lu GH, Klutchko SR, Batley BL, Dahring TK, Hamby transcriptional and posttranscriptional mechanisms. Cell Growth JM, Hallak H, Doherty AM, Keiser JA. In vitro pharmacological Diff 1996; 7: 487–500. characterization of PD 166285, a new nanomolar potent and 85 Wang XY, McCubrey JA. Regulation of interleukin 3 expression

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 500 in normal and autocrine transformed hematopoietic cells. Intl J nelly AM, McKearn JP. Circular permutation of the granulocyte Oncol 1997; 10: 989–1001. colony-stimulating factor receptor agonist domain of myelopoie- 86 Wang XY, McCubrey JA. Differential effects of retroviral, long ter- tin. Biochemistry 1999; 38: 4564–4571. minal repeats on interleukin-3 gene expression and autocrine 102 McCubrey JA, Blalock WL, Saleh O, Pearce M, Burrows C, Steel- transformation. Leukemia 1997; 11: 1711–1725. man LS, Lee JT, Franklin RA, Oberhaus SM, Moye PW, Doshi PD, 87 Wang XY, Hoyle PE, McCubrey JA. Characterization of proteins McKearn JP. Enhanced ability of daniplestim and myelopoietin-1 binding the 3Ј regulatory region of the IL-3 gene in IL-3 dependent to suppress apoptosis in human hematopoietic cells. Leukemia and autocrine-transformed hematopoietic cells. Leukemia 1998; 2001; 15: 1203–1216. 12: 520–531. 103 Abegg AL, Vickery LE, Bremer ME, Donnelly AM, Doshi PD, 88 Steelman LS, Algate PA, Blalock WL, Wang XY, Prevost KD, Hoyle Evans ML, Thurman TL, Braford SR, Caparon MH, Bauer SC, Giri PE, McCubrey JA. Oncogenic effects of overexpression of the JG, Welply JK, McKearn JP, Smith WG. The enhanced in vitro interleukin-3 receptor on hematopoietic cells. Leukemia 1996; 10: hematopoietic activity of leridistim, a chimeric dual G-CSF and 528–542. IL-3 receptor agonist. Leukemia 2001 (in press). 89 Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS, 104 Srinivasa SP, Doshi PD. Extracellular signal-regulated kinase and Pettigrew AL, Meyerrose T, Rossi R, Grimes B, Rizzieri DA, Luger p38 mitogen-activated protein kinase pathways cooperate in SM, Phillips GL. The interleukin-3 receptor alpha chain is a unique mediating cytokine-induced proliferation of a leukemic cell line. marker for human acute myelogenous leukemia stem cells. Leuke- Leukemia 2001; 16: 244–253. mia 2001; 14: 1777–1784. 105 Padua RA, Guinn BA, Al-Sabah AI, Smith M, Taylor C, Pettersson 90 McCormack MP, Gonda TJ. Myeloproliferative disorder and leu- T, Ridge S, Carter G, White D, Oscier D, Chevret S, West R. kaemia in mice induced by different classes of constitutive RAS, FMS and p53 mutations and poor clinical outcome in myel- mutants of the human IL-3/IL-5/GM-CSF receptor common beta odysplasias: a 10-year follow-up. Leukemia 1998; 12: 887–892.

SPOTLIGHT subunit. Oncogene 1999; 18: 7190–7199. 106 MacKenzie KL, DolnikovA, Millington M, Shounan Y, Symonds 91 D’Andrea RJ, Harrison-Findik D, Butcher CM, Finnie J, Blumbergs G. Mutant N-ras induces myeloproliferative disorders and P, Bartley P, McCormack M, Jones K, Rowland R, Gonda TJ, Vadas apoptosis in bone marrow repopulated mice. Blood 1999; 93: MA. Dysregulated hematopoiesis and a progressive neurological 2043–2056. disorder induced by expression of an activated form of the human 107 Schaich M, Ritter M, Illmer T, Lisske P, Thiede C, Schakel U, common beta chain in transgenic mice. J Clin Invest 1998; 102: Mohr B, Ehninger G, Neubauer A. Mutations in ras proto-onco- 1951–1960. genes are associated with lower mdr1 gene expression in adult 92 Hoyle PE, Steelman LS, McCubrey JA. Autocrine transformation acute myeloid leukaemia. Br J Haematol 2001; 112: 300–307. of human hematopoietic cells after transfection with an activated 108 Stirewalt DL, Kopecky KJ, Meshinchi S, Appelbaum FR, Slovak GM-CSF gene. Cytokines Cell Mol Ther 1997; 3: 159–168. ML, Willman CL, Radich JP. FLT3, RAS, and TP53 mutations in 93 Gale RE, Freeburn RW, Khwaja A, Chopra R, Linch DC. A trunc- elderly patients with acute myeloid leukemia. Blood 2001; 97: ated isoform of the human beta chain common to the receptors for 3589–3595. granulocyte–macrophage colony-stimulating factor, interleukin-3 109 Buday L, Downward J. Epidermal growth factor regulates p21ras (IL-3), and IL-5 with increased mRNA expression in some patients through the formation of a complex of receptor, Grb2 adapter with acute leukemia. Blood 1998; 91: 54–63. protein, and Sos nucleotide exchange factor. Cell 1993; 73: 94 Sun Q, Woodcock JM, Rapoport A, Stomski FC, Korpelainen EI, 611–620. Bagley CJ, Goodall GJ, Smith WB, Gamble JR, Vadas MA, Lopez 110 Moodie SA, Willumsen BM, Weber MJ, Wolfman A. Complexes AF. Monoclonal antibody 7G3 recognizes the N-terminal domain of Ras-GTP with Raf-1 and mitogen-activated protein kinase kin- of the human interleukin-3 (IL-3) receptor alpha-chain and func- ase. Science 1993; 260: 1658–1660. tions as a specific IL-3 receptor antagonist. Blood 1996; 87: 83– 111 Vojtek AB, Hollenberg SM, Cooper JA. Mammalian Ras interacts 92. directly with the serine/threonine kinase Raf. Cell 1993; 74: 95 Sun Q, Jones K, McClure B, Cambareri B, Zacharakis B, Iversen 205–214. PO, Stomski F, Woodcock JM, Bagley CJ, D’Andrea R, Lopez AF. 112 Zhang X, Settleman J, Kyriakis JM, Takeuchi-Suzuki E, Elledge SJ, Simultaneous antagonism of interleukin-5, granulocyte–macro- Marshall MS, Bruder JT, Rapp UR, Avruch J. Normal and oncog- phage colony-stimulating factor, and interleukin-3 stimulation of enic p21ras proteins bind to the amino-terminal regulatory human eosinophils by targetting the common cytokine binding domain of c-Raf-1. Nature 1993; 364: 308–314. site of their receptors. Blood 1999; 94: 1943–1951. 113 Warne PH, Viciana RP, Downward J. Direct interaction of Ras 96 Hogge DE, Willman CL, Kreitman RJ, Berger M, Hall PD, Kopecky and the amino-terminal region of Raf-1 in vitro. Nature 1993; KJ, McLain C, Tagge EP, Eaves CJ, Frankel AE. Malignant progeni- 364: 353–355. tors from patients with acute myelogenous leukemia are sensitive 114 Ross EM, Wilkie TM. GTPase-activating proteins for heterotrim- to a diphtheria toxin granulocyte–macrophage colony-stimulating eric G proteins: regulators of G protein signaling (RGS) and RGS- factor fusion protein. Blood 1998; 92: 589–595. like proteins. Annu Rev Biochem 2000; 69: 795–827. 97 Frankel AE, Lilly M, Kreitman R, Hogge D, Beran M, Freedman 115 Adjei AA. Blocking oncogenic Ras signaling for cancer therapy. MH, Emanuel PD, McLain C, Hall P, Tagge E, Berger M, Eaves C. J Natl Cancer Inst 2001; 93: 1062–1074. Diphtheria toxin fused to granulocyte–macrophage colony-stimul- 116 Karp JE, Lancet JE, Kaufmann SH, End DW, Wright JJ, Bol K, ating factor is toxic to blasts from patients with juvenile myelo- Horak I, Tidwell ML, Liesveld J, Kottke TJ, Ange D, Buddharaju monocytic leukemia and chronic myelomonocytic leukemia. L, Gojo I, Highsmith WE, Belly RT, Hohl RJ, Rybak ME, Thibault Blood 1998; 92: 4279–4286. A, Rosenblatt J. Clinical and biologic activity of the farnesyl- 98 Frankel AE, McCubrey JA, Miller MS, Delatte S, Ramage J, Kiser inhibitor R115777 in adults with refractory and M, Kucera GL, Alexander RL, Beran M, Tagge EP, Kreitman RJ, relapsed acute leukemias: a phase 1 clinical–laboratory correla- Hogge DE. Diphtheria toxin fused to human interleukin-3 is toxic tive trial. Blood 2001; 97: 3361–3369. to blasts from patients with myeloid leukemias. Leukemia 2000; 117 Karp JE. Farnesyl protein transferase inhibitors as targeted ther- 14: 576–585. apies for hematologic malignancies. Semin Hematol 2001; 38: 99 Kiser M, McCubrey JA, Steelman LS, Shelton JG, Miller MS, Ram- 16–23. age J, Alexander RL, Kucera GL, Pettenati M, Willingham MC, 118 Adjei AA, Erlichman C, Davis JN, Cutler DL, Sloan JA, Marks RS, Frankel AE. Oncogene-dependent engraftment of human myeloid Hanson LJ, Svingen PA, Atherton P, Bishop WR, Kirschmeier P, leukemia cells in immunosuppressed mice. Leukemia 2001; 15: Kaufmann SH. A phase I trial of the farnesyl transferase inhibitor 814–818. SCH66336: evidence for biological and clinical activity. Cancer 100 Vallera DA, Seo SY, Panoskaltsis-Mortari A, Griffin JD, Blazar Res 2000; 60: 1871–1877. BR. Targeting myeloid leukemia with a DT(390)-mIL-3 fusion 119 Du W, Prendergast GC. Geranylgeranylated RhoB mediates sup- immunotoxin: ex vivo and in vivo studies in mice. Protein Eng pression of human tumor cell growth by farnesyltransferase 1999; 12: 779–785. inhibitors. Cancer Res 1999; 59: 5492–5496. 101 McWherter CA, Feng Y, Zurfluh LL, Klein BK, Baganoff MP, Pol- 120 Jiang K, Coppola D, Crespo NC, Nicosia SV, Hamilton AD, Sebti azzi JO, Hood WF, Paik K, Abegg AL, Grabbe ES, Shieh JJ, Don- SM, Cheng JQ. The phosphoinositide 3-OH kinase/AKT2 path-

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 501 way as a critical target for farnesyltransferase inhibitor-induced directly with the serine/threonine kinase Raf. Cell 1993; 74: apoptosis. Mol Cell Bio 2000; 20: 139–148. 205–214. 121 Law BK, Norgaard P, Gnudi L, Kahn BB, Poulson HS, Moses 142 Erpel T, Courtneidge SA. Src family protein tyrosine kinases and HL. Inhibition of DNA synthesis by a farnesyltransferase inhibitor cellular signal transduction pathways. Curr Opin Cell Biol 1995; involves inhibition of the p70(s6k) pathway. J Biol Chem 1999; 7: 176–182. 274: 4743–4748. 143 Pathan NI, Ashendel CL, Geahlen RL, Harrison ML. Activation 122 Zhang Y, Mukhopadhyay T, Donehower LA, Georges RN, Roth of T cell Raf-1 at mitosis requires the protein-tyrosine kinase Lck. JA. Retroviral vector-mediated transduction of K-ras antisense J Biol Chem 1996; 271: 30315–30317. RNA into human lung cancer cells inhibits expression of the 144 Sozeri O, Vollmer K, Liyanage M, Frith D, Kour G, Mark GE 3rd, malignant phenotype. Hum Gene Ther 1993; 4: 451–460. Stabel S. Activation of the c-Raf protein kinase by protein kinase 123 Song JJ, Lee H, Kim E, Kim YS, Yoo NC, Roh JK, Kim BS, Kim J. C phosphorylation. Oncogene 1992; 7: 2259–2262. Transduction effect of antisense K-ras on malignant phenotypes 145 Morrison DK, Heidecker G, Rapp UR, Copeland TD. Identifi- in gastric cancer cells. Cancer Lett 2000; 157: 1–7. cation of the major phosphorylation sites of the Raf-1 kinase. J 124 Mukhopadhyay T, Tainsky M, Cavender AC, Roth JA. Specific Biol Chem 1993; 268: 17309–17316. inhibition of K-ras expression and tumorgenicity of lung cancer 146 Farrar MA, Alberol-Ila J, Perlmutter RM. Activation of the Raf-1 by antisense RNA. Cancer Res 1991; 51: 1744–1748. kinase cascade by coumermycin-induced dimerization. Nature 125 Hagemann C, Rapp UR. Isotype-specific functions of Raf kinases. 1996; 383: 178–181. Exp Cell Res 1999; 253: 34–46. 147 Weber CK, Slupsky JR, Herrmann C, Schuler M, Rapp UR, Block 126 Franklin RA, McCubrey JA. Kinases: positive and negative regu- C. Mitogenic signaling of Ras is regulated by differential interac- lators of apoptosis. Leukemia 2000; 14: 2019–2034. tion with Raf isozymes. Oncogene 2000; 19: 169–176. 127 White MK, McCubrey JA. Suppression of apoptosis: role in cell 148 Chang F, McCubrey JA. p21Cip1 induced by Raf is associated with growth and neoplasia. Leukemia 2001; 15: 1011–1021. increased Cdk4 activity in hematopoietic cells. Oncogene 2001; 128 Eychene A, Dusanter-Fourt I, Barnier JV, Papin C, Charon M, 20: 4353–4364. Gisselbrecht S, Calothy G. Expression and activation of B-Raf 149 Schulte TW, Blagosklonny MV, Ingui C, Neckers L. Disruption kinase isoforms in human and murine leukemia cell lines. Onco- of the Raf-1-Hsp90 molecular complex results in destabilization gene 1995; 10: 1159–1165. of Raf-1 and loss of Raf-1-Ras association. J Biol Chem 1995; 129 McCubrey JA, Steelman LS, Hoyle PE, Blalock WL, Weinstein- 270: 24585–24588. Oppenheimer C, Franklin RA, Cherwinski H, Bosch E, McMahon 150 Schulte TW, Blagosklonny MV, Romanova L, Mushinski JF, M. Differential abilities of activated Raf oncoproteins to abrogate Monia BP, Johnston JF, Nguyen P, Trepel J, Neckers LM. Destabi- cytokine dependency, prevent apoptosis and induce autocrine lization of Raf-1 by geldanamycin leads to disruption of the Raf- growth factor synthesis in human hematopoietic cells. Leukemia 1-MEK-mitogen-activated protein kinase signalling pathway. Mol 1998; 12: 1903–1929. Cell Biol 1996; 16: 5839–5845. 130 McCubrey JA, Steelman LS, Moye PW, Hoyle PE, Weinstein- 151 Blagosklonny MV, Fojo T, Bhalla KN, Kim JS, Trepel JB, Figg Oppenheimer C, Chang F, Pearce M, White MK, Franklin RA, WD, Rivera Y, Neckers LM. The Hsp90 inhibitor geldanamycin Blalock WL. Effects of deregulated Raf and MEK1 expression on selectively sensitizes Bcr-Abl-expressing leukemia cells to cyto- the cytokine-dependency of hematopoietic cells. Adv Enzyme toxic chemotherapy. Leukemia 2001; 15: 1537–1543. Regl 2000; 40: 305–337. 152 Soga S, Neckers LM, Schulte TW, Shiotsu Y, Akasaka K, Narumi 131 Weinstein-Oppenheimer C, Steelman LS, Algate PA, Blalock WL, H, Agatsuma T, Ikuina Y, Murakata C, Tamaoki T, Akinaga S. Burrows C, Hoyle PE, Lee JT, Moye PW, Shelton JG, Franklin RA, KF25706, a novel oxime derivative of radicicol, exhibits in vivo McCubrey JA. Effects of deregulated Raf activation on integrin, antitumor activity via selective depletion of Hsp90 binding sig- cytokine-receptor expression and the induction of apoptosis in naling molecules. Cancer Res 1999; 59: 2931–2938. hematopoietic cells. Leukemia 2000; 14: 1921–1938. 153 Shiotsu Y, Neckers LM, Wortman I, An WG, Schulte TW, Soga 132 Moye PW, Blalock WL, Hoyle PE, Chang F, Franklin RA, S, Murakata C, Tamaoki T, Akinaga S. Novel oxime derivatives Weinstein-Oppenheimer C, Pearce M, Steelman LS, McMahon of radicicol induce erythroid differentiation associated with pref- M, McCubrey JA. Synergy between Raf and BCL2 in abrogating erential G(1) phase accumulation against chronic myelogenous the cytokine-dependency of hematopoietic cells. Leukemia leukemia cells through destabilization of Bcr-Abl with Hsp90 2000; 14: 1060–1079. complex. Blood 2000; 96: 2284–2291. 133 Weinstein-Oppenheimer CR, Blalock WL, Steelman LS, Chang 154 Kimoto M, Sakamoto K, Shirouzu M, Hirao I, Yokoyama S. RNA F, McCubrey JA. Raf signal transduction cascade as a target for aptamers that specifically bind to the Ras-binding domain of Raf- chemotherapeutic intervention in growth factor-dependent 1. FEBS Lett 1998; 441: 322–326. tumors. Pharm Ther 2000; 88: 229–279. 155 Barnard D, Sun H, Baker L, Marshall MS. In vitro inhibition of 134 Chung GT, Huang DP, Lo KW, Chan MK, Wong FW. Genetic Ras-Raf association by short peptides. Biochem Biophys Res lesion in the carcinogenesis of cervical cancer. Anticancer Res Commun 1998; 247: 176–180. 1992; 12: 1485–1490. 156 Cioffi CL, Garay M, Johnston JF, McGraw K, Boggs RT, Hreniuk

135 Habuchi T, Kinoshita H, Yamada H, Kakehi Y, Ogawa O, Wu D, Monia BP. Selective inhibition of A-Raf and C-Raf mRNA SPOTLIGHT WJ, Takahashi R, Sugiyama T, Yoshida O. Oncogene amplifi- expression by antisense oligodeoxynucleotides in rat vascular cation in urothelial cancers with p53 gene mutation or MDM2 smooth muscle cells: role of A-Raf and C-Raf in serum-induced amplification. J Natl Cancer Inst 1994; 86: 1331–1335. proliferation. Mol Pharmacol 1997; 51: 383–389. 136 Ishikawa F, Takaku F, Nagao M, Sugimura T. Rat c-raf oncogene 157 Schumacher C, Cioffi CL, Sharif H, Haston W, Monia BP, Wen- activation by a rearrangement that produces a fused protein. Mol nogle L. Exposure of human vascular smooth muscle cells to Raf- Cell Biol 1987; 7: 1226–1232. 1 antisense oligodeoxynucleotides: cellular responses and phar- 137 Sithanandam G, Dean M, Brennscheidt U, Beck T, Gazdar A, macodynamic implications. Mol Pharmacol 1998; 53: 97–104. Minna JD, Brauch H, Zbar B, Rapp UR. Loss of heterozygosity 158 Kolch W, Heidecker G, Kochs G, Hummel R, Vahidi H, Mischak at the c-raf locus in small cell lung carcinoma. Oncogene 1989; H, Finkenzeller G, Marme D, Rapp UR. Protein kinase C alpha 4: 451–455. activates RAF-1 by direct phosphorylation. Nature 1993; 364: 138 Storm SM, Rapp UR. Oncogene activation: c-raf-1 gene 249–252. mutations in experimental and naturally occurring tumors. Tox- 159 Cacace AM, Ueffing M, Philipp A, Han EK, Kolch W, Weinstein icol Lett 1993; 67: 201–210. IB. PKC epsilon functions as an oncogene by enhancing acti- 139 Weber CK, Slupsky JR, Kalmes HA, Rapp UR. Active Ras induces vation of the Raf kinase. Oncogene 1996; 13: 2517–2526. heterodimerization of cRaf and BRaf. Cancer Res 2001; 61: 160 Zhang Y, Yao B, Delikat S, Bayoumy S, Lin XH, Basu S, McGin- 3595–3598. ley M, Chan-Hui PY, Lichenstein H, Kolesnick R. Kinase sup- 140 Van Aelst L, Barr M, Marcus S, Polverino A, Wigler M. Complex pressor of Ras is ceramide-activated protein kinase. Cell 1997; formation between RAS and RAF and other protein kinases. Proc 89: 63–72. Natl Acad Sci USA 1993; 90: 6213–6217. 161 Xia K, Mukhopadhyay NK, Inhorn RC, Barber DL, Rose PE, Lee 141 Vojtek AB, Hollenberg SM, Cooper JA. Mammalian Ras interacts RS, Narsimhan RP, D’Andrea AD, Griffin JD, Roberts TM. The

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 502 cytokine-activated tyrosine kinase JAK2 activates Raf-1 in a S, Mett H, O’Reilly T, Meyer T. Inhibitors of protein kinases: CGP p21ras-dependent manner. Proc Natl Acad Sci USA 1996; 93: 41251, a protein kinase inhibitor with potential as an anticancer 11681–11686. agent. Pharmacol Ther 1999; 82: 293–301. 162 Marais R, Light Y, Paterson HF, Mason CS, Marshall CJ. Differen- 181 Morrison DK. KSR: a MAPK scaffold of the Ras pathway? J Cell tial regulation of Raf-1, A-Raf, and B-Raf by oncogenic ras and Sci 2001; 114: 1609–1612. tyrosine kinases. J Biol Chem 1997; 272: 4378–4383. 182 Zhang Y, Yao B, Delikat S, Bayoumy S, Lin XH, Basu S, McGin- 163 Mason CS, Springer CJ, Cooper RG, Superti-Furga G, Marshall ley M, Chan-Hui PY, Lichenstein H, Kolesnick R. Kinase sup- CJ, Marais R. Serine and tyrosine phosphorylations cooperate in pressor of Ras is ceramide-activated protein kinase. Cell 1997; Raf-1, but not B-Raf activation. EMBO J 1999; 18: 2137–2148. 89: 63–72. 164 Shuai K, Horvath CM, Huang LH, Qureshi SA, Cowburn D, Dar- 183 Denouel-Galy A, Douville EM, Warne PH, Papin C, Laugier D, nell JE Jr. Interferon activation of the transcription factor Stat91 Calothy G, Downward J, Eychene A. Murine Ksr interacts with involves dimerization through SH2-phosphotyrosyl peptide inter- MEK and inhibits Ras-induced transformation. Cell Biol 1998; 8: actions. Cell 1994; 76: 821–828. 46–55. 165 Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways and tran- 184 Jacobs D, Glossip D, Xing H, Muslin AJ, Kornfeld K. Multiple scriptional activation in response to IFNs and other extracellular docking sites on substrate proteins form a modular system that signaling proteins. Science 1994; 264: 1415–1421. mediates recognition by ERK MAP kinase. Genes Dev 2001; 13: 166 Ihle JN, Witthuhn BA, Quelle FW, Yamamoto K, Thierfelder WE, 163–175. Kreider B, Silvennoinen O. Signaling by the cytokine receptor 185 Muller J, Cacace AM, Lyons WE, McGill CB, Morrison DK. superfamily: JAKs and STATs. Trends Biochem Sci 1994; 19: Identification of B-KSR1, a novel brain-specific isoform of KSR1 222–227. that functions in neuronal signaling. Mol Cell Biol 2000; 20: 167 Goodman PA, Niehoff LB, Uckun FM. Role of tyrosine kinases 5529–5539.

SPOTLIGHT in induction of the c-jun proto-oncogene in irradiated B-lineage 186 Cacace AM, Michaud NR, Therrien M, Mathes K, Copeland T, lymphoid cells. J Biol Chem 1998; 273: 17742–17748. Rubin GM, Morrison DK. Identification of constitutive and ras- 168 Sudbeck EA, Liu XP, Narla RK, Mahajan S, Ghosh S, Mao C, inducible phosphorylation sites of KSR: implications for 14–3-3 Uckun FM. Structure-based design of specific inhibitors of Janus binding, mitogen-activated protein kinase binding, and KSR kinase 3 as apoptosis-inducing antileukemic agents. Clin Cancer overexpression. Mol Cell Biol 1999; 19: 229–240. Res 1999; 5: 1569–1582. 187 Therrien M, Michaud NR, Rubin GM, Morrison DK. KSR modu- 169 Meydan N, Grunberger T, Dadi H, Shahar M, Arpaia E, Lapidot lates signal propagation within the MAPK cascade. Genes Dev Z, Leeder JS, Freedman M, Cohen A, Gazit A, Levitzki A, Roif- 1996; 10: 2684–2695. man CM. Inhibition of acute lymphoblastic leukaemia by a Jak- 188 Zheng CF, Guan KL. Properties of MEKs, the kinases that phos- 2 inhibitor. Nature 1996; 379: 645–648. phorylate and activate the extracellular signal-regulated kinases. 170 Eriksen KW, Kaltoft K, Mikkelsen G, Nielsen M, Zhang Q, Geisler J Biol Chem 1993; 268: 23933–23939. C, Nissen MH, Ropke C, Wasik MA, Odum N. Constitutive 189 English JM, Vanderbilt CA, Xu S, Marcus S, Cobb MH. Isolation STAT3-activation in Sezary syndrome: tyrphostin AG490 inhibits of MEK5 and differential expression of alternatively spliced STAT3-activation, interleukin-2 receptor expression and growth forms. J Biol Chem 1995; 270: 28897–28902. of leukemic Sezary cells. Leukemia 2001; 15: 787–793. 190 Catling AD, Schaeffer HJ, Reuter CW, Reddy GR, Weber MJ. A 171 De Vos J, Jourdan M, Tarte K, Jasmin C, Klein B. JAK2 tyrosine proline-rich sequence unique to MEK1 and MEK2 is required for kinase inhibitor tyrphostin AG490 downregulates the mitogen- raf binding and regulates MEK function. Mol Cell Biol 1995; 15: activated protein kinase (MAPK) and signal transducer and acti- 5214–5225. vator of transcription (STAT) pathways and induces apoptosis in 191 Zheng CF, Guan KL. Activation of MEK family kinases requires myeloma cells. Br J Haematol 2000; 109: 823–828. phosphorylation of two conserved Ser/Thr residues. EMBO J 172 Kolch W, Heidecker G, Kochs G, Hummel R, Vahidi H, Mischak 1994; 13: 1123–1131. H, Finkenzeller G, Marme D, Rapp UR. Protein kinase C alpha 192 Papin C, Eychene A, Brunet A, Pages G, Pouyssegur J, Calothy activates RAF-1 by direct phosphorylation. Nature 1993; 364: G, Barnier JV. B-Raf protein isoforms interact with and phos- 249–252. phorylate Mek-1 on serine residues 218 and 222. Oncogene 173 Tamaoki T. Use and specificity of staurosporine, UCN-01, and 1995; 10: 1647–1651. calphostin C as protein kinase inhibitors. Meth Enzymol 1991; 193 Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, 201: 340–347. Berman K, Cobb MH. Mitogen-activated protein (MAP) kinase 174 Takahashi I, Kobayashi E, Asano K, Yoshida M, Nakano H. UCN- pathways: regulation and physiological functions. Endocr Rev 01, a selective inhibitor of protein kinase C from Streptomyces. 2001; 22: 153–183. J Antibiot 1987; 40: 1782–1784. 194 Mansour SJ, Matten WT, Hermann AS, Candia JM, Rong S, Fukas- 175 Seynaeve CM, Kazanietz MG, Blumberg PM, Sausville EA, Wor- awa K, Vande Woude GF, Ahn NG. Transformation of mam- land PJ. Differential inhibition of protein kinase C isozymes by malian cells by constitutively active MAP kinase kinase. Science UCN-01, a staurosporine analogue. Mol Pharmacol 1994; 45: 1994; 265: 966–970. 1207–1214. 195 Okuda K, Matulonis U, Salgia R, Kanakura Y, Druker B, Griffin 176 Seynaeve CM, Stetler-Stevenson M, Sebers S, Kaur G, Sausville JD. Factor independence of human myeloid leukemia cell lines EA, Worland PJ. Cell cycle arrest and growth inhibition by the is associated with increased phosphorylation of the proto-oncog- protein kinase antagonist UCN-01 in human breast carcinoma ene Raf-1. Exp Hematol 1994; 11: 1111–1117. cells. Cancer Res 1993; 53: 2081–2086. 196 Kang CD, Yoo SD, Hwang BW, Kim KW, Kim DW, Kim CM, Kim 177 Wang Q, Worland PJ, Clark JL, Carlson BA, Sausville EA. SH, Chung BS. The inhibition of ERK/MAPK not the activation of Apoptosis in 7-hydroxystaurosporine-treated T lymphoblasts cor- JNK/SAPK is primarily required to induce apoptosis in chronic relates with activation of cyclin-dependent kinases 1 and 2. Cell myelogenous leukemic K562 cells. Leuk Res 2000; 24: 527–534. Growth Differ 1995; 6: 927–936. 197 Blalock WL, Pearce M, Steelman LS, Franklin RA, McCarthy SA, 178 Meyer T, Regenass U, Fabbro D, Alteri E, Rosel J, Muller M, Cherwinski H, McMahon M, McCubrey JA. A conditionally Caravatti G, Matter A. A derivative of staurosporine (CGP 41 251) active form of MEK1 results in autocrine transformation of human shows selectivity for protein kinase C inhibition and in vitro anti- and mouse hematopoietic cells. Oncogene 2000; 19: 526–536. proliferative, as well as in vivo anti-tumor activity. Int J Cancer 198 Blalock WL, Moye PW, Chang F, Pearch M, Steelman LS, McMa- 1989; 43: 851–856. hon M, McCubrey JA. Combined effects of aberrant MEK1 179 Thavasu P, Propper D, McDonald A, Dobbs N, Ganesan T, Tal- activity and BCL2 overexpression on relieving the cytokine- bot D, Braybrook J, Caponigro F, Hutchison C, Twelves C, Man dependency of hematopoietic cells. Adv Enzyme Regl 2000; 40: A, Fabbro D, Harris A, Balkwill F. The protein kinase C inhibitor 305–337. CGP41251 suppresses cytokine release and extracellular signal- 199 Blalock WL, Pearce M, Chang F, Lee JT, Pohnert S, Burrows C, regulated kinase 2 expression in cancer patients. Cancer Res Steelman LS, Franklin RA, McMahon M, McCubrey JA. Effects of 1999; 59: 3980–3984. inducible MEK1 activation on the cytokine-dependency of 180 Fabbro D, Buchdunger E, Wood J, Mestan J, Hofmann F, Ferrari lymphoid cells. Leukemia 2001; 15: 794–807.

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 503 200 Hoyle PE, Moye PW, Steelman LS, Blalock WL, Franklin RA, 219 Cobb MH, Goldsmith EJ. Dimerization in MAP-kinase signaling. Pearce M, Cherwinski H, Bosch E, McMahon M, McCubrey JA. Trends Biochem Sci 2000; 25: 7–9. Differential abilities of the Raf family of protein kinases to abro- 220 Shapiro PS, Whalen AM, Tolwinski NS, Wilsbacher J, Froelich- gate cytokine-dependency and prevent apoptosis in murine hem- Ammon SJ, Garcia M, Osheroff N, Ahn NG. Extracellular signal- atopoietic cells by a MEK1-dependent mechanism. Leukemia regulated kinase activates topoisomerase IIalpha through a mech- 2000; 14: 642–656. anism independent of phosphorylation. Mol Cell Biol 1999; 19: 201 Alessi DR, Cuenda A, Cohen P, Dudley DT, Saltiel AR. PD 3551–3560. 098059 is a specific inhibitor of the activation of mitogen-acti- 221 Sturgill TW, Ray LB, Erikson E, Maller JL. Insulin-stimulated MAP- vated protein kinase kinase in vitro and in vivo. J Biol Chem 2 kinase phosphorylates and activates ribosomal protein S6 kin- 1995; 270: 27489–27494. ase II. Nature 1988; 334: 715–718. 202 Dudley DT, Pang L, Decker SJ, Bridges AJ, Saltiel AR. A synthetic 222 Seger R, Krebs EG. The MAPK signaling cascade. FASEB J 1995; inhibitor of the mitogen-activated protein kinase cascade. Proc 9: 726–735. Natl Acad Sci USA 1995; 92: 7686–7689. 223 Poteet-Smith CE, Smith JA, Lannigan DA, Freed TA, Sturgill TW. 203 Pages G, Lenormand P, L’Allemain G, Chambard JC, Meloche Generation of constitutively active p90 ribosomal S6 kinase in S, Pouyssegur J. Mitogen-activated protein kinases p42mapk and vivo. Implications for the mitogen-activated protein kinase-acti- p44mapk are required for fibroblast proliferation. Proc Natl Acad vated protein kinase family. J Biol Chem 1999; 274: 22135– Sci USA 1993; 90: 8319–8323. 22138. 204 Eliceiri BP, Klemke R, Stromblad S, Cheresh DA. Integrin 224 Kim SC, Hahn JS, Min YH, Yoo NC, Ko YW, Lee WJ. Constitutive alphavbeta3 requirement for sustained mitogen-activated protein activation of extracellular signal-regulated kinase in human acute kinase activity during angiogenesis. J Cell Biol 1998; 140: leukemias: combined role of activation of MEK, hyperexpression 1255–1263. of extracellular signal-regulated kinase, and downregulation of a 205 Pang L, Sawada T, Decker SJ, Saltiel AR. Inhibition of MAP kin- phosphatase, PAC1. Blood 1999; 93: 3893–3899. ase kinase blocks the differentiation of PC-12 cells induced by 225 Morgan MA, Dolp O, Reuter CW. Cell-cycle-dependent acti- nerve growth factor. J Biol Chem 1995; 270: 13585–13588. vation of mitogen-activated protein kinase kinase (MEK-1/2) in 206 Finlay D, Healy V, Furlong F, O’Connell FC, Keon NK, Martin myeloid leukemia cell lines and induction of growth inhibition F. MAP kinase pathway signalling is essential for extracellular and apoptosis by inhibitors of RAS signaling. Blood 2001; 97: matrix determined mammary epithelial cell survival. Cell Death 1823–1834. Differ 2000; 7: 302–313. 226 Ajenjo N, Aaronson DS, Ceballos E, Richard C, Leon J, Crespo 207 Holmstrom TH, Tran SE, Johnson VL, Ahn NG, Chow SC, Eriks- P. Myeloid leukemia cell growth and differentiation are inde- son JE. Inhibition of mitogen-activated kinase signaling sensitizes pendent of mitogen-activated protein kinase ERK1/2 activation. HeLa cells to Fas receptor-mediated apoptosis. Mol Cell Biol J Biol Chem 2000; 275: 7189–7197. 1999; 19: 5991–6002. 227 Iida M, Towatari M, Nakao A, Iida H, Kiyoi H, Nakano Y, Tanim- 208 Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, oto M, Saito H, Naoe T. Lack of constitutive activation of MAP Feeser WS, Van Dyk DE, Pitts WJ, Earl RA, Hobbs F, Copeland kinase pathway in human acute myeloid leukemia cells with N- RA, Magolda RL, Scherle PA, Trzaskos JM. Identification of a Ras mutation. Leukemia 1999; 13: 585–589. novel inhibitor of mitogen-activated protein kinase kinase. J Biol 228 Horiuchi KY, Scherle PA, Trzaskos JM, Copeland RA. Competi- Chem 1998; 273: 18623–18632. tive inhibition of MAP kinase activation by a peptide representing 209 Sebolt-Leopold JS, Dudley DT, Herrera R, Van Becelaere K, the alpha C helix of ERK. Biochemistry 1998; 37: 8879–8885. Wiland A, Gowan RC, Tecle H, Barrett SD, Bridges A, Przyb- 229 Zhang H, Shi X, Hampong M, Blanis L, Pelech S. Stress-induced ranowski S, Leopold WR, Saltiel AR. Blockade of the MAP kinase inhibition of ERK1 and ERK2 by direct interaction with p38 MAP pathway suppresses growth of colon tumors in vivo. Nat Med kinase. J Biol Chem 2001; 276: 6905–6908. 1999; 5: 810–816. 230 Fisher TL, Blenis J. Evidence for two catalytically active kinase 210 Sebolt-Leopold JS. Development of anticancer drugs targeting the domains in pp90rsk. Mol Cell Biol 1996; 16: 1212–1219. MAP kinase pathway. Oncogene 2000; 19: 6594–6599. 231 Alcorta DA, Crews CM, Sweet LJ, Bankston L, Jones SW, Erikson 211 Dai Y, Yu C, Singh V, Tang L, Wang Z, McInistry R, Dent P, RL. Sequence and expression of chicken and mouse rsk: homo- Grant S. Pharmacological inhibitors of the mitogen-activated pro- logs of Xenopus laevis ribosomal S6 kinase. Mol Cell Biol 1989; tein kinase (MAPK) kinase/MAPK cascade interact synergistically 9: 3850–3859. with UCN-01 to induce mitochondrial dysfunction and apoptosis 232 Moller DE, Xia CH, Tang W, Zhu AX, Jakubowski M. Human rsk in human leukemia cells. Cancer Res 2001; 61: 5106–5115. isoforms: cloning and characterization of tissue-specific 212 Deb A, Zamanian-Daryoush M, Xu Z, Kadereit S, Williams RG. expression. Am J Physiol 1994; 266: C351–C359. Protein kinase PKR is required for platelet-derived growth factor 233 Zhao Y, Bjorbaek C, Weremowicz S, Morton CC, Moller DE. signaling of c-fos gene expression via Erks and Stat3. EMBO J 2001; 20: 2487–2496. RSK3 encodes a novel pp90rsk isoform with a unique N-terminal 213 Peus D, Vasa RA, Beyerle A, Meves A, Krautmacher C, Pittelkow sequence: growth factor-stimulated kinase function and nuclear translocation. Mol Cell Biol 1995; 15: 4353–4363.

MR. UVB activates ERK1/2 and p38 signaling pathways via SPOTLIGHT reactive oxygen species in cultured keratinocytes. J Invest Derm 234 Jensen CJ, Buch MB, Krag TO, Hemmings BA, Gammeltoft S, 1999; 112: 751–756. Frodin M. 90-kDa ribosomal S6 kinase is phosphorylated and 214 Bhat NR, Zhang P. Hydrogen peroxide activation of multiple activated by 3-phosphoinositide-dependent protein kinase-1. J mitogen-activated protein kinases in an oligodendrocyte cell Biol Chem 1999; 274: 27168–27176. line: role of extracellular signal-regulated kinase in hydrogen per- 235 Frodin M, Jensen CJ, Merienne K, Gammeltoft S. A phosphoser- oxide-induced cell death. J Neurochem 1999; 72: 112–119. ine-regulated docking site in the protein kinase RSK2 that recruits 215 Stevenson MA, Pollock SS, Coleman CN, Calderwood SK. X- and activates PDK1. EMBO J 2000; 19: 2924–2934. irradiation, phorbol esters, and H2O2 stimulate mitogen-acti- 236 Xing J, Ginty DD, Greenberg ME. Coupling of the RAS-MAPK vated protein kinase activity in NIH-3T3 cells through the forma- pathway to gene activation by RSK2, a growth factor-regulated tion of reactive oxygen intermediates. Cancer Res 1994; 54: CREB kinase. Science 1996; 273: 959–963. 12–15. 237 Joel PB, Smith J, Sturgill TW, Fisher TL, Blenis J, Lannigan DA. 216 Ferrell JE Jr. MAP kinases in mitogenesis and development. Curr pp90rsk1 regulates estrogen receptor-mediated transcription Top Dev Biol 1996; 33: 1–60. through phosphorylation of Ser-167. Mol Cell Biol 1998; 18: 217 KhokhlatchevAV, Canagarajah B, Wilsbacher J, Robinson M, 1978–1984. Atkinson M, Goldsmith E, Cobb MH. Phosphorylation of the 238 Schouten GJ, Vertegaal AC, Whiteside ST, Israel A, Toebes M, MAP kinase ERK2 promotes its homodimerization and nuclear Dorsman JC, van der Eb AJ, Zantema A. IkappaB alpha is a target translocation. Cell 1998; 93: 605–615. for the mitogen-activated 90 kDa ribosomal S6 kinase. EMBO J 218 Canagarajah BJ, KhokhlatchevA, Cobb MH, Goldsmith EJ. Acti- 1997; 16: 3133–3144. vation mechanism of the MAP kinase ERK2 by dual phosphoryl- 239 Ghoda L, Lin X, Greene WC. The 90-kDa ribosomal S6 kinase ation. Cell 1997; 90: 859–869. (pp90rsk) phosphorylates the N-terminal regulatory domain of

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 504 IkappaBalpha and stimulates its degradation in vitro. J Biol Chem regulated target, SSeCKS, reverses v-Jun-induced transformation 1997; 272: 21281–21288. of 10T1/2 murine fibroblasts. Oncogene 2001; 20: 141–146. 240 Chen RH, Abate C, Blenis J. Phosphorylation of the c-Fos transre- 262 Vial E, Perez S, Castellazzi M. Transcriptional control of SPARC pression domain by mitogen-activated protein kinase and 90- by v-Jun and other members of the AP1 family of transcription kDa ribosomal S6 kinase. Proc Natl Acad Sci USA 1993; 90: factors. Oncogene 2000; 19: 5020–5029. 10952–10956. 263 Epinat JC, Gilmore TD. Diverse agents act at multiple levels to 241 Crans HN. Transcription factors and translocations in lymphoid inhibit the Rel/NF-kappaB signal transduction pathway. Oncog- and myeloid leukemia. Leukemia 2001; 15: 313–331. ene 1999; 18: 6896–6909. 242 Jean D, Harbison M, McConkey DJ, Ronai Z, Bar-Eli M. CREB 264 Chen FE, Ghosh G. Regulation of DNA binding by Rel/NF-kap- and its associated proteins act as survival factors for human mela- paB transcription factors: structural views. Oncogene 1999; 18: noma cells. J Biol Chem 1998; 273: 24884–24890. 6845–6852. 243 Ginty DD, Bonni A, Greenberg ME. Nerve growth factor acti- 265 Chen C, Edelstein LC, Gelinas C. The Rel/NF-kappaB family vates a Ras-dependent protein kinase that stimulates c-fos tran- directly activates expression of the apoptosis inhibitor Bcl-x(L). scription via phosphorylation of CREB. Cell 1994; 77: 713–725. Mol Cell Biol 2000; 20: 2687–2695. 244 Sakamoto KM, Fraser JK, Lee HJ, Lehman E, Gasson JC. Granulo- 266 Schreck R, Baeuerle PA. NF-kappa B as inducible transcriptional cyte–macrophage colony-stimulating factor and interleukin-3 activator of the granulocyte–macrophage colony-stimulating fac- signaling pathways converge on the CREB-binding site in the tor gene. Mol Cell Biol 1990; 10: 1281–1286. human egr-1 promoter. Mol Cell Biol 1994; 14: 5975–5985. 267 Duyao MP, Kessler DJ, Spicer DB, Sonenshein GE. Transactiv- 245 Pugazhenthi S, Miller E, Sable C, Young P, Heidenreich KA, ation of the c-myc gene by HTLV-1 tax is mediated by NFkB. Boxer LM, Reusch JE. Insulin-like growth factor-I induces bcl-2 Curr Top Microbiol Immunol 1992; 182: 421–424. promoter through the transcription factor cAMP-response 268 Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS

SPOTLIGHT element-binding protein. J Biol Chem 1999; 274: 27529–27535. Jr. NF-kappaB controls cell growth and differentiation through 246 Facchini LM, Penn LZ. The molecular role of Myc in growth and transcriptional regulation of cyclin D1. Mol Cell Biol 1999; 19: transformation: recent discoveries lead to new insights. FASEB 5785–5799. J 1998; 12: 633–651. 269 Xing J, Ginty DD, Greenberg ME. Coupling of the RAS-MAPK 247 Boyd KE, Farnham PJ. Identification of target genes of oncogenic pathway to gene activation by RSK2, a growth factor-regulated transcription factors. Proc Soc Exp Biol Med 1999; 222: 9–28. CREB kinase. Science 1996; 273: 959–963. 248 Li LH, NerlovC, Prendergast G, MacGregor D, Ziff EB. c-Myc 270 Xing J, Kornhauser JM, Xia Z, Thiele EA, Greenberg ME. Nerve represses transcription in vivo by a novel mechanism dependent growth factor activates extracellular signal-regulated kinase and on the initiator element and Myc box II. EMBO J 1994; 13: p38 mitogen-activated protein kinase pathways to stimulate 4070–4079. CREB serine 133 phosphorylation. Mol Cell Biol 1998; 18: 249 Philipp A, Schneider A, Vasrik I, Finke K, Xiong Y, Beach D, 1946–1955. Alitalo K, Eilers M. Repression of cyclin D1: a novel function of 271 Parker D, Ferreri K, Nakajima T, LaMorte VJ, Evans R, Koerber MYC. Mol Cell Biol 1994; 14: 4032–4043. SC, Hoeger C, Montminy MR. Phosphorylation of CREB at Ser- 250 Reisman D, Elkind NB, Roy B, Beamon J, Rotter V. c-Myc trans- 133 induces complex formation with CREB-binding protein via activates the p53 promoter through a required downstream a direct mechanism. Mol Cell Biol 1996; 16: 694–703. CACGTG motif. Cell Growth Differ 1993; 4: 57–65. 272 Chrivia JC, Kwok RP, Lamb N, Hagiwara M, Montminy MR, 251 Jones RM, Branda J, Johnston KA, Polymenis M, Gadd M, Rustgi Goodman RH. Phosphorylated CREB binds specifically to the A, Callanan L, Schmidt EV. An essential E box in the promoter nuclear protein CBP. Nature 1993; 365: 855–859. of the gene encoding the mRNA cap-binding protein (eukaryotic 273 Wagner EF. AP-1: Introductory remarks. Oncogene 2001; 20: initiation factor 4E) is a target for activation by c-myc. Mol Cell 2334–2335. Biol 1996; 16: 4754–4764. 274 Yu W, Fantl WJ, Harrowe G, Williams LT. Regulation of the MAP 252 Bello-Fernandez C, Packham G, Cleveland JL. The ornithine kinase pathway by mammalian Ksr through direct interaction decarboxylase gene is a transcriptional target of c-Myc. Proc Natl with MEK and ERK. Curr Biol 1998; 8: 56–64. Acad Sci USA 1993; 90: 7804–7808. 275 Smeal T, Binetruy B, Mercola DA, Birrer M, Karin M. Oncogenic 253 Dittmer J, Nordheim A. Ets transcription factors and human dis- and transcriptional cooperation with Ha-Ras requires phos- ease. Biochim Biophys Acta 1998; 1377: F1–F11. phorylation of c-Jun on serines 63 and 73. Nature 1991; 354: 254 Iwasaka C, Tanaka K, Abe M, Sato Y. Ets-1 regulates angiogenesis 494–496. by inducing the expression of urokinase-type plasminogen acti- 276 Hibi M, Lin A, Smeal T, Minden A, Karin M. Identification of an vator and matrix metalloproteinase-1 and the migration of vascu- oncoprotein- and UV-responsive protein kinase that binds and lar endothelial cells. J Cell Physiol 1996; 169: 522–531. potentiates the c-Jun activation domain. Genes Dev 1993; 7: 255 Schneikert J, Peterziel H, Defossez PA, Klocker H, Launoit Y, 2135–2148. Cato AC. Androgen receptor-Ets protein interaction is a novel 277 Derijard B, Hibi M, Wu IH, Barrett T, Su B, Deng T, Karin M, mechanism for steroid hormone-mediated down-modulation of Davis RJ. JNK1: a protein kinase stimulated by UV light and Ha- matrix metalloproteinase expression. J Biol Chem 1996; 271: Ras that binds and phosphorylates the c-Jun activation domain. 23907–23913. Cell 1994; 76: 1025–1037. 256 Gambarotta G, Boccaccio C, Giordano S, Ando M, Stella MC, 278 Minden A, Lin A, Smeal T, Derijard B, Cobb M, Davis R, Karin Comoglio PM. Ets up-regulates MET transcription. Oncogene M. c-Jun N-terminal phosphorylation correlates with activation 1996; 13: 1911–1917. of the JNK subgroup but not the ERK subgroup of mitogen-acti- 257 Dittmer J, Gitlin SD, Reid RL, Brady JN. Transactivation of the vated protein kinases. Mol Cell Biol 1994; 14: 6683–6688. P2 promoter of parathyroid hormone-related protein by human 279 Chou SY, Baichwal V, Ferrell JE Jr. Inhibition of c-Jun DNA bind- T-cell lymphotropic virus type I Tax1: evidence for the involve- ing by mitogen-activated protein kinase. Mol Biol Cell 1992; 3: ment of transcription factor Ets1. J Virol 1993; 67: 6087–6095. 1117–1130. 258 Shaulian E, Karin M. AP-1 in cell proliferation and survival. 280 Smeal T, Binetruy B, Mercola DA, Birrer M, Karin M. Oncogenic Oncogene 2001; 20: 2390–2400. and transcriptional cooperation with Ha-Ras requires phos- 259 Lee W, Haslinger A, Karin M, Tjian R. Activation of transcription phorylation of c-Jun on serines 63 and 73. Nature 1991; 354: by two factors that bind promoter and enhancer sequences of 494–496. the human metallothionein gene and SV40. Nature 1987; 325: 281 Binetruy B, Smeal T, Karin M. Ha-Ras augments c-Jun activity 368–372. and stimulates phosphorylation of its activation domain. Nature 260 Fu Sl, Bottoli I, Goller M, Vogt PK. Heparin-binding epidermal 1991; 351: 122–127. growth factor-like growth factor, a v-Jun target gene, induces 282 Lloyd A, Yancheva N, Wasylyk B. Transformation suppressor oncogenic transformation. Proc Natl Acad Sci USA 1999; 96: activity of a Jun transcription factor lacking its activation domain. 5716–5721. Nature 1991; 352: 635–638. 261 Cohen SB, Waha A, Gelman IH, Vogt PK. Expression of a down- 283 Johnson R, Spiegelman B, Hanahan D, Wisdom R. Cellular trans-

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 505 formation and malignancy induced by ras require c-jun. Mol Cell activation of gene expression. J Biol Chem 1991; 266: 23521– Biol 1996; 16: 4504–4511. 23524. 284 Suzuki T, Murakami M, Onai N, Fukuda E, Hashimoto Y, Sonobe 307 Seth A, Gonzalez FA, Gupta S, Raden DL, Davis RJ. Signal trans- MH, Kameda T, Ichinose M, Miki K, Iba H. Analysis of AP-1 duction within the nucleus by mitogen-activated protein kinase. function in cellular transformation pathways. J Virol 1994; 68: J Biol Chem 1992; 267: 24796–24804. 3527–3535. 308 Lutterbach B, Hann SR. c-Myc transactivation domain-associated 285 Chen RH, Abate C, Blenis J. Phosphorylation of the c-Fos transre- kinases: questionable role for map kinases in c-Myc phosphoryl- pression domain by mitogen-activated protein kinase and 90- ation. J Cell Biochem 1999; 72: 483–491. kDa ribosomal S6 kinase. Proc Natl Acad Sci USA 1993; 90: 309 Colman MS, Ostrowski MC. The transactivation potential of a c- 10952–10956. Myc N-terminal region (residues 92–143) is regulated by growth 286 Chen RH, Juo PC, Curran T, Blenis J. Phosphorylation of c-Fos factor/Ras signaling. Nucleic Acids Res 1996; 24: 1971–1978. at the C-terminus enhances its transforming activity. Oncogene 310 Sears R, Leone G, DeGregori J, Nevins JR. Ras enhances Myc 1996; 12: 1493–1502. protein stability. Mol Cell 1999; 3: 169–179. 287 Davis RJ. Signal transduction by the c-Jun N-terminal kinase. 311 Kerkhoff E, Houben R, Loffler S, Troppmair J, Lee JE, Rapp UR. Biochem Soc Symp 1999; 64: 1–12. Regulation of c-myc expression by Ras/Raf signalling. Oncogene 288 van Dam H, Castellazzi M. Distinct roles of Jun : Fos and 1998; 16: 211–216. Jun : ATF dimers in oncogenesis. Oncogene 2001; 20: 2453– 312 Noguchi K, Kitanaka C, Yamana H, Kokubu A, Mochizuki T, 2464. Kuchino Y. Regulation of c-Myc through phosphorylation at Ser- 289 Chiariello M, Marinissen MJ, Gutkind JS. Multiple mitogen-acti- 62 and Ser-71 by c-Jun N-terminal kinase. J Biol Chem 1999; vated protein kinase signaling pathways connect the cot oncop- 274: 32580–32587. rotein to the c-jun promoter and to cellular transformation. Mol 313 Wasylyk B, Hagman J, Gutierrez-Hartmann A. Ets transcription Cell Biol 2000; 20: 1747–1758. factors: nuclear effectors of the Ras-MAP-kinase signaling path- 290 Verma IM, Stevenson JK, Schwarz EM, Van Antwerp D, Miyam- way. Trends Biochem Sci 1998; 23: 213–216. oto S. Rel/NF-kappa B/I kappa B family: intimate tales of associ- 314 Wasylyk B, Hahn SL, Giovane A. The Ets family of transcription ation and dissociation. Genes Dev 1995; 9: 2723–2735. factors. Eur J Biochem 1993; 211: 7–18. 291 Pahl HL. Activators and target genes of Rel/NF-kappaB transcrip- 315 Sementchenko VI, Watson DK. Ets target genes: past, present and tion factors. Oncogene 1999; 18: 6853–6866. future. Oncogene 2000; 19: 6533–6548. 292 Lee FS, Hagler J, Chen ZJ, Maniatis T. Activation of the IkappaB 316 Yordy JS, Muise-Helmericks RC. Signal transduction and the Ets alpha kinase complex by MEKK1, a kinase of the JNK pathway. family of transcription factors. Oncogene 2000; 19: 6503–6513. Cell 1997; 88: 213–222. 317 Brunner D, Ducker K, Oellers N, Hafen E, Scholz H, Klambt C. 293 Baeuerle PA, Baltimore D. NF-kappa B: ten years after. Cell The ETS domain protein pointed-P2 is a target of MAP kinase in 1996; 87: 13–20. the sevenless signal transduction pathway. Nature 1994; 370: 294 Li S, Sedivy JM. Raf-1 protein kinase activates the NF-kappa B 386–389. transcription factor by dissociating the cytoplasmic NF-kappa B- 318 Hipskind RA, Buscher D, Nordheim A, Baccarini M. Ras/MAP I kappa B complex. Proc Natl Acad Sci USA 1993; 90: 9247– kinase-dependent and -independent signaling pathways target 9251. distinct ternary complex factors. Genes Dev 1994; 8: 1803– 295 Ghoda L, Lin X, Greene WC. The 90-kDa ribosomal S6 kinase 1816. (pp90rsk) phosphorylates the N-terminal regulatory domain of 319 Hipskind RA, Baccarini M, Nordheim A. Transient activation of IkappaBalpha and stimulates its degradation in vitro. J Biol Chem RAF-1, MEK, and ERK2 coincides kinetically with ternary com- 1997; 272: 21281–21288. plex factor phosphorylation and immediate-early gene promoter activity in vivo. Mol Cell Biol 1994; 14: 6219–6231. 296 Schouten GJ, Vertegaal AC, Whiteside ST, Israel A, Toebes M, 320 Keyse SM. Protein phosphatases and the regulation of mitogen- Dorsman JC, van der Eb AJ, Zantema A. IkappaB alpha is a target activated protein kinase signalling. Curr Opin Cell Biol 2000; 12: for the mitogen-activated 90 kDa ribosomal S6 kinase. EMBO J 186–192. 1997; 16: 3133–3144. 321 Camps M, Nichols A, Arkinstall S. Dual specificity phosphatases: 297 Wang W, Abbruzzese JL, Evans DB, Larry L, Cleary KR, Chiao a gene family for control of MAP kinase function. FASEB J 2000; PJ. The nuclear factor-kappa B RelA transcription factor is consti- 14: 6–16. tutively activated in human pancreatic adenocarcinoma cells. 322 Sun H, Charles CH, Lau LF, Tonks NK. MKP-1 (3CH134), an Clin Cancer Res 1999; 5: 119–127. immediate early gene product, is a dual specificity phosphatase 298 Giallongo A, Appella E, Ricciardi R, Rovera G, Croce CM. Identi- that dephosphorylates MAP kinase in vivo. Cell 1993; 75: fication of the c-myc oncogene product in normal and malignant 487–493. B cells. Science 1983; 222: 430–432. 323 King AG, Ozanne BW, Smythe C, Ashworth A. Isolation and 299 Ramsay G, Evan GI, Bishop JM. The protein encoded by the characterisation of a uniquely regulated threonine, tyrosine phos- human proto-oncogene c-myc. Proc Natl Acad Sci USA 1984; phatase (TYP 1) which inactivates ERK2 and p54jnk. Oncogene 81: 7742–7746.

1995; 11: 2553–2563. SPOTLIGHT 300 Schmidt EV. The role of c-myc in cellular growth control. Oncog- 324 Guan KL, Butch E. Isolation and characterization of a novel dual ene 1999; 18: 2988–2996. specific phosphatase, HVH2, which selectively dephosphoryl- 301 Grandori C, Cowley SM, James LP, Eisenman RN. The ates the mitogen-activated protein kinase. J Biol Chem 1995; Myc/Max/Mad network and the transcriptional control of cell 270: 7197–7203. behavior. Annu Rev Cell Dev Biol 2000; 16: 653–699. 325 Muda M, Theodosiou A, Rodrigues N, Boschert U, Camps M, 302 Roussel MF, Ashmun RA, Sherr CJ, Eisenman RN, Ayer DE. Inhi- Gillieron C, Davies K, Ashworth A, Arkinstall S. The dual speci- bition of cell proliferation by the Mad1 transcriptional repressor. ficity phosphatases M3/6 and MKP-3 are highly selective for Mol Cell Biol 1996; 16: 2796–2801. inactivation of distinct mitogen-activated protein kinases. J Biol 303 Baudino TA, Cleveland JL. The Max network gone mad. Mol Cell Chem 1996; 271: 27205–27208. Biol 2001; 21: 691–702. 326 Muda M, Boschert U, Dickinson R, Martinou JC, Martinou I, 304 Fujioka Y, Taira T, Maeda Y, Tanaka S, Nishihara H, Iguchi-Ariga Camps M, Schlegel W, Arkinstall S. MKP-3, a novel cytosolic SM, Nagashima K, Ariga H. MM-1, a c-Myc-binding protein, is protein-tyrosine phosphatase that exemplifies a new class of a candidate for a tumor suppressor in leukemia/lymphoma and mitogen-activated protein kinase phosphatase. J Biol Chem tongue cancer. J Biol Chem 2001; 276: 45137–45144. 1996; 271: 4319–4326. 305 Mori K, Maeda Y, Kitaura H, Taira T, Iguchi-Ariga SM, Ariga H. 327 Muda M, Boschert U, Smith A, Antonsson B, Gillieron C, Chabert MM-1, a novel c-Myc-associating protein that represses tran- C, Camps M, Martinou I, Ashworth A, Arkinstall S. Molecular scriptional activity of c-Myc. J Biol Chem 1998; 273: 29794– cloning and functional characterization of a novel mitogen-acti- 29800. vated protein kinase phosphatase, MKP-4. J Biol Chem 1997; 306 Seth A, Alvarez E, Gupta S, Davis RJ. A phosphorylation site 272: 5141–5151. located in the NH2-terminal domain of c-Myc increases trans- 328 Tanoue T, Moriguchi T, Nishida E. Molecular cloning and

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 506 characterization of a novel dual specificity phosphatase, MKP- 347 Whitman M, Downes CP, Keeler M, Keller T, Cantley L. Type I 5. J Biol Chem 1999; 274: 19949–19956. phosphatidylinositol kinase makes a novel inositol phospholipid, 329 Theodosiou A, Smith A, Gillieron C, Arkinstall S, Ashworth A. phosphatidylinositol-3-phosphate. Nature 1988; 332: 644–646. MKP5, a new member of the MAP kinase phosphatase family, 348 Anderson KE, Coadwell J, Stephens LR, Hawkins PT. Translo- which selectively dephosphorylates stress-activated kinases. cation of PDK-1 to the plasma membrane is important in Oncogene 1999; 18: 6981–6988. allowing PDK-1 to activate . Curr Biol 1998; 8: 330 Tanoue T, Yamamoto T, Maeda R, Nishida E. A novel MAPK 684–691. phosphatase MKP-7 acts preferentially on JNK/SAPK and p38 349 Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N, alpha and beta MAPKs. J Biol Chem 2001; 276: 26629–26639. Cohen P, Hemmings BA. Mechanism of activation of protein kin- 331 Marti F, Krause A, Post NH, Lyddane C, Dupont B, Sadelain M, ase B by insulin and IGF-1. EMBO J 1996; 15: 6541–6551. King PD. Negative-feedback regulation of CD28 costimulation 350 Alessi DR, Deak M, Casamayor A, Caudwell FB, Morrice N, Nor- by a novel mitogen-activated protein kinase phosphatase, MKP6. man DG, Gaffney P, Reese CB, MacDougall CN, Harbison D, J Immunol 2001; 166: 197–206. Ashworth A, Bownes M. 3-Phosphoinositide-dependent protein 332 Brondello JM, Brunet A, Pouyssegur J, McKenzie FR. The dual kinase-1 (PDK1): structural and functional homology with the specificity mitogen-activated protein kinase phosphatase-1 and - Drosophila DSTPK61 kinase. Curr Biol 1997; 7: 776–789. 2 are induced by the p42/p44MAPK cascade. J Biol Chem 1997; 351 Aoki M, Batista O, Bellacosa A, Tsichlis P, Vogt PK. The akt kin- 272: 1368–1376. ase: molecular determinants of oncogenicity. Proc Natl Acad Sci 333 Shapiro PS, Ahn NG. Feedback regulation of Raf-1 and mitogen- USA 1998; 95: 14950–14955. activated protein kinase (MAP) kinase kinases 1 and 2 by MAP 352 Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese kinase phosphatase-1 (MKP-1). J Biol Chem 1998; 273: 1788– CB, Cohen P. Characterization of a 3-phosphoinositide-depen- 1793. dent protein kinase which phosphorylates and activates protein

SPOTLIGHT 334 Camps M, Nichols A, Gillieron C, Antonsson B, Muda M, Chab- kinase Balpha. Curr Biol 1997; 7: 261–269. ert C, Boschert U, Arkinstall S. Catalytic activation of the phos- 353 Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg phatase MKP-3 by ERK2 mitogen-activated protein kinase. ME. Akt phosphorylation of BAD couples survival signals to the Science 1998; 280: 1262–1265. cell-intrinsic death machinery. Cell 1997; 91: 231–241. 335 Reffas S, Schlegel W. Compartment-specific regulation of extra- 354 del Peso L, Gonzalez-Garcia M, Page C, Herrera R, Nunez G. cellular signal-regulated kinase (ERK) and c-Jun N-terminal kin- Interleukin-3-induced phosphorylation of BAD through the pro- ase (JNK) mitogen-activated protein kinases (MAPKs) by ERK- tein kinase Akt. Science 1997; 278: 687–689. dependent and non-ERK-dependent inductions of MAPK phos- 355 Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franke TF, phatase (MKP)-3 and MKP-1 in differentiating P19 cells. Biochem Stanbridge E, Frisch S, Reed JC. Regulation of cell death protease J 2000; 352: 701–708. caspase-9 by phosphorylation. Science 1998; 282: 1318–1321. 336 Haystead TA, Sim AT, Carling D, Honnor RC, Tsukitani Y, Cohen 356 Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson P, Hardie DG. Effects of the tumour promoter okadaic acid on MJ, Arden KC, Blenis J, Greenberg ME. Akt promotes cell survival intracellular protein phosphorylation and metabolism. Nature by phosphorylating and inhibiting a Forkhead transcription fac- 1989; 337: 78–81. tor. Cell 1999; 96: 857–868. 337 Orgad S, Brewis ND, Alphey L, Axton JM, Dudai Y, Cohen PT. 357 Kane LP, Shapiro VS, Stokoe D, Weiss A. Induction of NF-kappaB The structure of protein phosphatase 2A is as highly conserved by the Akt/PKB kinase. Curr Biol 1999; 9: 601–604. as that of protein phosphatase 1. FEBS Lett 1990; 275: 44–48. 358 Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, 338 Wassarman DA, Solomon NM, Chang HC, Karim FD, Therrien Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, M, Rubin GM. Protein phosphatase 2A positively and negatively Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R. PTEN, regulates Ras1-mediated photoreceptor development in Droso- a putative protein tyrosine phosphatase gene mutated in human phila. Genes Dev 1996; 10: 272–278. brain, breast, and prostate cancer. Science 1997; 275: 1943– 339 Manfroid I, Martial JA, Muller M. Inhibition of protein phospha- 1947. tase PP1 in GH3B6, but not in GH3 cells, activates the 359 Stambolic V, Suzuki A, de la Pompa JL, Brothers GM, Mirtsos C, MEK/ERK/c-fos pathway and the human prolactin promoter, Sasaki T, Ruland J, Penninger JM, Siderovski DP, Mak TW. Nega- involving the coactivator CPB/p300. Mol Endocrinol 2001; 15: tive regulation of PKB/Akt-dependent cell survival by the tumor 625–637. suppressor PTEN. Cell 1998; 95: 29–39. 340 Chung H, Brautigan DL. Protein phosphatase 2A suppresses MAP 360 Gu J, Tamura M, Yamada KM. Tumor suppressor PTEN inhibits kinase signalling and ectopic protein expression. Cell Signal integrin- and growth factor-mediated mitogen-activated protein 1999; 11: 575–580. (MAP) kinase signaling pathways. J Cell Biol 1998; 143: 1375– 341 Braconi Quintaje SB, Church DJ, Rebsamen M, Valloton MB, 1383. Hemmings BA, Lang U. Role of protein phosphatase 2A in the 361 Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, regulation of mitogen-activated protein kinase activity in ven- dephosphorylates the lipid second messenger, phosphatidylinosi- tricular cardiomyocytes. Biochem Biophys Res Commun 1996; tol 3,4,5-trisphosphate. J Biol Chem 1998; 273: 13375–13378. 221: 539–547. 362 Myers MP, Pass I, Batty IH, Van der Kaay J, StolarovJP, Hem- 342 Jaumot M, Hancock JF. Protein phosphatases 1 and 2A promote mings BA, Wigler MH, Downes CP, Tonks NK. The lipid phos- Raf-1 activation by regulating 14–3-3 interactions. Oncogene phatase activity of PTEN is critical for its tumor supressor func- 2000; 20: 3949–3958. tion. Proc Natl Acad Sci USA 1998; 95: 13513–13518. 343 Abraham D, Podar K, Pacher M, Kubicek M, Welzel N, Hem- 363 Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is mings BA, Dilworth SM, Mischak H, Kolch W, Baccarini M. Raf- essential for embryonic development and tumour suppression. 1-associated protein phosphatase 2A as a positive regulator of Nat Genet 1998; 19: 348–355. kinase activation. J Biol Chem 2000; 275: 22300–22304. 364 Podsypanina K, Ellenson LH, Nemes A, Gu J, Tamura M, Yamada 344 Kiguchi K, Glesne D, Chubb CH, Fujiki H, Huberman E. Differ- KM, Cordon-Cardo C, Catoretti G, Fisher PE, Parsons R. Mutation ential induction of apoptosis in human breast tumor cells by oka- of Pten/Mmac1 in mice causes neoplasia in multiple organ sys- daic acid and related inhibitors of protein phosphatases 1 and tems. Proc Natl Acad Sci USA 1999; 96: 1563–1568. 2A. Cell Growth Differ 1994; 5: 995–1004. 365 Arcaro A, Wymann MP. Wortmannin is a potent phosphatidyli- 345 Hiles ID, Otsu M, Volinia S, Fry MJ, Gout I, Dhand R, Panayotou nositol 3-kinase inhibitor: the role of phosphatidylinositol 3,4,5- G, Ruiz-Larrea F, Thompson A, Totty NF et al. Phosphatidylinosi- trisphosphate in neutrophil responses. Biochem J 1993; 296: tol 3-kinase: structure and expression of the 110 kd catalytic sub- 297–301. unit. Cell 1992; 70: 419–429. 366 Powis G, Bonjouklian R, Berggren MM, Gallegos A, Abraham R, 346 Klippel A, Escobedo JA, Fantl WJ, Williams LT. The C-terminal Ashendel C, Zalkow L, Matter WF, Dodge J, Grindey G, Vlahos SH2 domain of p85 accounts for the high affinity and specificity CJ. Wortmannin, a potent and selective inhibitor of phosphatidyl- of the binding of phosphatidylinositol 3-kinase to phosphorylated inositol-3-kinase. Cancer Res 1994; 54: 2419–2423. platelet-derived growth factor beta receptor. Mol Cell Biol 1992; 367 Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of 12: 1451–1459. phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-

Leukemia Therapeutic intervention in leukemia via Raf/MEK/ERK pathway JT Lee and JA McCubrey 507 benzopyran-4-one (LY294002). J Biol Chem 1994; 269: 5241– Chang F, Pearce M, Shelton JG, White MK, Franklin RA, Pohnert 5248. SC. Interactions between the PI3K and Raf signaling pathways 368 Hu Y, Qiao L, Wang S, Rong SB, Meuillet EJ, Berggren M, Gal- can result in the transformation of hematopoietic cells. Cancer legos A, Powis G, Kozikowski AP. 3-(Hydroxymethyl)-bearing Detect Prev 2001; 25: 375–393. phosphatidylinositol ether lipid analogues and carbonate surro- 374 Reusch HP, Zimmermann S, Schaefer M, Paul M, Moelling K. gates block PI3-K, Akt, and cancer cell growth. J Med Chem Regulation of Raf by Akt controls growth and differentiation in 2000; 43: 3045–3051. vascular smooth muscle cells. J Biol Chem 2001; 276: 33630– 369 King WG, Mattaliano MD, Chan TO, Tsichlis PN, Brugge JS. Pho- 33637. sphatidylinositol 3-kinase is required for integrin-stimulated AKT 375 Guan KL, Figueroa C, Brtva TR, Zhu T, Taylor J, Barber TD, and Raf-1/mitogen-activated protein kinase pathway activation. Vojtek AB. Negative regulation of the serine/threonine kinase B- Mol Cell Biol 1997; 17: 4406–4418. Raf by Akt. J Biol Chem 2000; 275: 27354–27359. 370 Wennstrom S, Downward J. Role of phosphoinositide 3-kinase 376 Zimmermann S, Moelling K. Phosphorylation and regulation of in activation of ras and mitogen-activated protein kinase by epi- Raf by Akt (protein kinase B). Science 1999; 286: 1741–1744. dermal growth factor. Mol Cell Biol 1999; 19: 4279–4288. 377 Rommel C, Clarke BA, Zimmermann S, Nunez L, Rossman R, 371 Sheng H, Shao J, DuBois RN. Akt/PKB activity is required for Ha- Reid K, Moelling K, Yancopoulos GD, Glass DJ. Differentiation Ras-mediated transformation of intestinal epithelial cells. J Biol stage-specific inhibition of the Raf-MEK-ERK pathway by Akt. Chem 2001; 276: 14498–14504. Science 1999; 286: 1738–1741. 372 von Gise A, Lorenz P, Wellbrock C, Hemmings B, Berberich- 378 Ruvolo PP, Deng X, May WS. Phosphorylation of Bcl2 and regu- Siebelt F, Rapp UR, Troppmair J. Apoptosis suppression by Raf- lation of apoptosis. Leukemia 2001; 15: 515–522. 1 and MEK1 requires MEK- and phosphatidylinositol 3-kinase- 379 Ruvolo PP. Ceramide regulates cellular homeostasis via diverse dependent signals. Mol Cell Biol 2001; 21: 2324–2236. stress signaling pathways. Leukemia 2001; 15: 1153–1160. 373 McCubrey JA, Lee JT, Steelman LS, Blalock WL, Moye PW, SPOTLIGHT

Leukemia