PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 KIProj/Acc8-PreslclRC/DateDue.indd NECESSITY AND SUFFICIENCY OF MITOGEN-ACTIVATED PROTEIN KINASE KINASE SIGNALING PATHWAYS FOR MELANOMA CELL PROLIFERATION By CHIH-SHIA LEE A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILSOPHY Biochemistry and Molecular Biology 2010 ABSTRACT NECESSITY AND SUFFICIENCY OF MITOGEN-ACTIVATED PROTEIN KINASE KINASE SIGNALING PATHWAYS FOR MELANOMA CELL PROLIFERATION By CHIH—SHIA LEE The mitogen activated protein kinase kinases (MKK or MEK) signaling pathways are critical for melanoma survival. Although MEK] and MEK2 generally are considered functionally redundant, a few studies have indicated that they may have distinct biological functions. However, in assessing the relative contribution of MEK] and MEK2 towards extracellular signal-regulated kinase (BRIO-mediated biologic response, investigators have relied on tests of necessity, not sufficiency. Dissecting both necessity and sufficiency of MEK Signaling pathways is important to understand the interplay of the signaling network. To do so, I used two complementary approaches to determine the necessity and sufficiency of MEKI and MEK2 signaling pathways for melanoma cell proliferation. To determine the necessity, I used isoform-specific siRNA to knock down either MEK] or MEK2 in human melanoma SK-MEL—28 cells. An effect on proliferation was Observed only when both MEK] and MEK2 were knocked down, indicating that neither of the individual isoforms is necessary for SK-MEL—ZS cell proliferation. To determine the sufficiency, I have developed a novel experimental system that allows only one MEK/MKK signaling pathway to be present in cells. In this system, anthrax lethal toxin (LeTx), a pan-MEK/MKK protease, is used to inhibit multiple endogenous MEK/MKKS in cells, and either MEKl or MEK2 signaling pathway is simultaneously rescued by the cleavage-resistant form of MEK (MEKcr). Surprisingly, ERK activity persisted in LeTx-treated cells expressing MEK2cr but not MEK] cr. Microarray analysis revealed groups Of non-overlapping downstream transcriptional targets of MEK] and MEK2, and indicated a substantial rescue effect of MEK2cr on proliferation pathways. Furthermore, LeTx efficiently inhibited the cell proliferation and anchorage-independent growth of SK-MEL—28 cells expressing MEKl cr but not MEK2cr. These results indicate MEK2 signaling is sufficient for ERK activation, melanoma cell proliferation, and anchorage-independent growth, and demonstrate that in SK-MEL-28 cells MEK] and MEK2 signaling pathways are not redundant and interchangeable for cell proliferation. I conclude that in the absence of other MKK, MEK2 is sufficient for SK-MEL-28 cell proliferation. MEK] can conditionally compensate for MEK2 only in the presence of other MKK. © Copyright by Chih-Shia Lee 2010 Acknowledgments Nothing is sufficient to express how grateful I am for the experience of being a graduate student and to be trained in the Duesbery Lab. 1 am everlastingly indebted to Dr. Duesbery for his guidance and inexhaustible patience over the past several years. The debt of gratitude that I owe Dr. Duesbery is so deep that I can only repay it by keeping Dr. Duesbery’s words in mind. In an E-mail Dr. Duesbery wrote to me: “Yoshio Masui, my PhD supervisor, told me that my training was conditional on my agreement to do the same with my students. In turn, this obligation will pass to you.” I would to thank Drs. Justin McCormick, Lee Kroos, Gregg Howe, and Kathy Gallo for agreeing to serve as my Guidance Committee members, and for their invaluable advice throughout my graduate school career. Many thanks are also necessary to Drs. Art Alberts, Jeff MacKeigan, Cindy Miranti, and Kyle Furge (Investigators at the Van Andel Research Institute) for their helpful advice and suggestions. I would like to thank Ms. Elissa Boguslawski for her professional assistance with xenografi experiments, as well as Mr. Karl Dykema for his help with microarray data analysis. I would also like to thank the current and former members in the Duesbery Lab for their help, especially to Dr. Jenn Bromberg-White for her invaluable advice, discussion, and encouragement. Finally, I would like to thank the Core Services at the Van Andel Research Institute for their technical help, Mr. David Nadziejka for his professional comments on scientific writing, and Ms. Laura Holman and Laura Round for their administration support during the past five years. This research would not be possible without their supportive help. This dissertation is dedicated to my best-loved family members: my grandmother, parents, aunt, brother and sister-in-law as well as the expected nephew. Table of Contents List of Tables ........................................................................................................... x List of Figures ........................................................................................................ xi List of Abbreviations ........................................................................................... xiii Chapter I. General introduction: MEK signaling pathway and cancer ............... 1 1.1. Introduction to MEK signaling pathway ................................................ 1 1.2. Involvement of MEK Signaling pathway in human cancers .................. 4 1.3. Targeting the Raf-MEK—ERK pathway as a strategy for treating cancers .................................................................................................... 4 1.3.1 . Small-molecule inhibitors .................................................................. 6 1.3.1.1. PD 098059 .................................................................................... 6 1.3.1.2. U0126 ........................................................................................... 7 1.3.1.3. PD 184352 (CI-1040) ................................................................... 8 1.3.1.4. Derivatives of PD 184352 .......................................................... 11 1.3.1.5. ARRY-142886 (AZD6244) ....................................................... 13 1.3.2. Mechanism of allosteric inhibition .................................................. 15 1.3.3. Biological inhibitors ......................................................................... 18 1.3.3.1. Anthrax lethal toxin ................................................................... 18 1.3.3.2. YopJ ........................................................................................... 22 1.3.4. MEK inhibitors fail tO generate clinical responses .......................... 24 1.4. Introduction to cutaneous melanoma ................................................... 26 1.4.1. Melanoma statistics .......................................................................... 27 1.4.2. Risk factors ...................................................................................... 28 1.4.3. Melanoma progression, the Clark model. ........................................ 28 1.4.4. Melanoma staging ............................................................................ 29 1.4.5. Treatment of melanoma ................................................................... 30 1.4.6. Common genetic abnormalities of melanoma ................................. 31 1.4.6.1. BRAF and NRAS ......................................................................... 31 1.4.6.2. CDKNZA (p161NK4 and p194" ) ............................................... 33 1.4.6.3. PTEN .......................................................................................... 35 1.4.7. Disease models ................................................................................. 37 1 .5. Discussion ............................................................................................ 41 1.6. Tables and figures ................................................................................ 43 Chapter II. Cleavage-resistant MEK proteins; a novel experimental model to establish MEK sufficiency .............................................................. 52 1 .1 . Introduction .......................................................................................... 52 1.2. Results .................................................................................................. 53 1.2.1. Point mutations at the Pl' site render MEK resistant to LP- mediated cleavage ............................................................................ 53 1.2.2. The cleavage-resistant mutation does not impair MEK activity. ..... 55 vi 1.2.3. Point mutations at the P1 ’ Site render other MKK members resistant to LF-mediated cleavage .................................................... 57 1.2.4. MKK4 cleavage in mammalian cells ............................................... 57 1.2.5. MKK7 cleavage in mammalian cells ............................................... 60 1 .3. Discussion ............................................................................................ 61 1.4. Figures .................................................................................................. 66 1.5. Materials and methods ......................................................................... 77 1.5.1. Cell lines and stable cell line establishment ..................................... 77 1.5.2. Chemicals and LeTx ........................................................................ 77 1.5.3. VS-MEKcr construction. .................................................................. 77 1.5.4. Construction of MKK4-V5-His6 and deletion mutants. .................. 78 1.5.5. In-cell MEK cleavage assay. ............................................................ 79 1 .5.6. Irnmunoblotting. ............................................................................... 80 1.5.7. Immunoprecipitation ........................................................................ 81 1.5.8. In vitro kinase assay. ........................................................................ 82 Chapter III. Sufficiency and necessity of MEK signaling pathways for melanoma cell proliferation ....................................................... 83 1 .1 . Introduction .......................................................................................... 83 1.2. Results .................................................................................................. 85 1.2.1. Necessity of MEK] and MEK2 signaling pathways for melanoma cell proliferation ............................................................. 85 1.2.1.1. Necessity of MEK] and MEK2 signaling pathways for ERK activation in melanoma cells ...................................................... 85 1.2.1.2. Necessity of MEK] and MEK2 signaling pathways for melanoma cell cycle progression ............................................... 86 1.2.2. Sufficiency Of MEKI and MEK2 signaling pathways for melanoma cell proliferation ............................................................. 87 1.2.2.1. MEK2, but not MEKI, is sufficient to maintain ERK2 activity ........................................................................................ 88 1.2.2.2. Genome-wide cDNA microarray reveals non-overlapping transcriptional patterns downstream of MEK] and MEK2 ....... 88 1.2.2.3. MEK2cr, but not MEchr, rescued proliferation-related pathways in LeTx-treated cells .................................................. 90 1.2.2.4. MEK2, but not MEK] , is sufficient for melanoma cell proliferation in vitro. .................................................................. 90 1.2.2.5. Neither MKK3 nor MKK6 is sufficient for SK-MEL-28 cell proliferation in vitro. .................................................................. 92 1.2.2.6. MEK2 signaling is sufficient for anchorage-independent growth ........................................................................................ 92 1.2.3. A xenograft model to test the sufficiency Of MEK] and MEK2 signaling pathways for melanoma tumor growth in vivo ................. 93 1.2.3.1. SK-MEL-28 xenografi tumor growth ........................................ 93 1.2.3.2. LeTx systemic treatment of SK-MEL-28 xenograft tumors ...... 94 1.2.3.3. Sensitivity of MEKcr-expressing xenograft tumors to LeTx systemic treatment ...................................................................... 95 vii 1.2.3.4. Dose-dependent effects of LeTx on SK-MEL-28 xenograft tumor growth .............................................................................. 96 1.2.3.5. Loss of V5-MEKcr expression in SK-MEL-28 xenograft tumors ......................................................................................... 97 1.2.3.6. LeTx systemic treatment does not cause MEK cleavage in tumor cells. ................................................................................. 97 1.2.3.7. Sufficiency of MEK signaling pathways for SK—MEL-28 tumor growth in vivo was not testable due to technical difficulties. ................................................................................. 98 1 .3. Discussion ............................................................................................ 99 1.4. Tables and figures .............................................................................. 110 1.5. Materials and methods ....................................................................... 143 1.5.1. Cell lines and stable cell line establishment ................................... 143 1.5.2. Chemicals and LeTx ...................................................................... 143 1.5.3. siRNA-mediated MEK knock down. ............................................. 143 1.5.4. In-cell MEK cleavage assay in SK—MEL-28 cells. ........................ 144 1 .5.5. Irnmunoblotting. ............................................................................. 145 1.5.6. Toxicity assay. ............................................................................... 146 1.5.7. SK-MEL-28 tumor xenografi and LeTx systemic treatment ......... 146 1.5.8. Anchorage-independent growth assay. .......................................... 148 1.5.9. Human cDNA microarray and transcriptional signature analysis. 148 1.5.10. Examination of p38 MAPK and JNK activations in SK-MEL-28 cells. ............................................................................................... 149 Chapter IV. General discussion ................................................................... 151 1.1. Experimental tools to study MEK and MK fimctions ..................... 152 1.2. Non-redundant roles of MEK] and MEK2 ........................................ 154 1.3. Complexity of the MEK/MKK signaling network ............................ 156 1.4. Therapeutic implications .................................................................... 159 Appendix I. Transcriptional signatures that are down-regulated by LeTx treatment and significantly rescued by MEK2cr ............................ 163 Appendix II. Transcriptional signatures that are affected by LeTx treatment and significantly rescued by both MEK] er and MEK2cr ............. 169 Appendix III. Transcriptional signatures that are not affected by LF treatment but cannot be rescued by either MEK] or or MEK2cr ................. 182 Appendix IV. Transcriptional signatures that are not affected by LF treatment but affected by VS-MEKI or expression ...................................... 190 Appendix V. Transcriptional signatures that are not affected by LF treatment but affected by V5-MBK2cr expression ....................................... 193 viii Appendix VI. Transcriptional signatures that are not affected by LF treatment but affected by both V5-MEK1cr and V5—MEK2cr expressions.196 Literature Cited .................................................................................................... 201 ix List of Tables Table I. Small-molecule ICso values .................................................................... 44 Table II. Alignment of MEK/MKK amino acid sequences flanking the LF cleavage sites. .................................................................................... 45 Table III. The Clark model of melanoma progression ......................................... 46 Table IV. Percentage of G1 population of MEK siRNA-treated SK-MEL-28 cells. ................................................................................................. 110 Table V. MEK2cr-rescued transcriptional Signatures that are significantly affected by LeTx treatment .............................................................. 1 11 List of Figures Images in this thesis/dissertation are presented in color. Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Figure 8. Figure 9. Figure 10. Figure 11. Figure 12. Figure 13. Figure 14. Figure 15. Figure 16. Figure 17. Figure 18. Figure 19. Figure 20. Schematic illustration of the Raf-MEK-ERK signaling pathway. ....... 47 Structures of MEK inhibitors. .............................................................. 49 Effect of PD 1843 52 on MEK activation in cells. ................................ 51 Resistance of VS-MEKcr to LF-mediated proteolysis. ........................ 66 Dual translation of VS-MEK expression vectors. ................................ 68 Activity of V5-MEKcr. ........................................................................ 69 Resistance of MKK3cr and MKK6cr to LF-mediated proteolysis ....... 71 Resistance of MKK4cr and MKK7cr to LF-mediated proteolysis. ...... 73 In-cell cleavage of MKK4 by LF. ........................................................ 74 Wild-type MKK7 is not cleaved by LP in cells. ................................ 75 LF does not cleave endogenous MKK7 in mammalian cells. ............ 76 Necessity of MEK] and MEK2 signaling pathways for ERK activation in SK-MEL-28 cells. ....................................................... 112 Individual MEK signaling in LeTx-treated SK-MEL-28 cells ......... 114 Overlapping and non-overlapping transcriptional targets downstream Of MEK] and MEK2. .................................................. 116 Expression levels of V5 fusion proteins in SK—MEL-28 cells. ........ 118 Inhibitory effect of LeTx on proliferation of SK-MEL-28 stable clones. .............................................................................................. 119 Sensitivities of SK-MEL-28 cells to LeTx and MEK inhibitors. ..... 121 LeTx treatment inhibits p38 and JNK MAP kinase activation in SK-MEL-28 cells. ............................................................................ 123 Sensitivity of V5-MKK3cr- and V5-MKK6cr-expressing cells to LeTx ................................................................................................. 124 Sufficiency of MEK2 signaling pathway for anchorage- independent growth of SK-MEL-28 cells ........................................ 126 xi Figure 21. Figure 22. Figure 23. Figure 24. Figure 25. Figure 26. Figure 27. Figure 28. SK-MEL-28 xenograft tumor growth and systemic treatment with LeTx ......................................................................................... 128 Reproducibility of SK—MEL-28 xenograft tumor growth in athyrnic nude mice. .......................................................................... 130 Sensitivity of VS-MEchr-expressing xenografi tumors to LeTx. ......................................................................................................... 131 Sensitivity Of V5-MEK2cr-expressing xenograft tumors to LeTx. ......................................................................................................... 133 Does-dependent effect of LeTx on SK-MEL-28 xenograft tumor growth. ............................................................................................. 135 Loss of V5-MEKcr expression in SK-MEL-28 xenograft tumors. ......................................................................................................... 137 Systemic treatment of LeTx does not cause MEK cleavage in SK-MEL-28 xenografl tumor cells. ................................................. 139 Unsupervised clustering of gene expression changes in LeTx- treated cells. ..................................................................................... 141 xii List of Abbreviations B-Raf, v—rafmurine sarcoma viral oncogene homolog Bl CDKN2A, cyclin-dependent kinase inhibitor 2A cDNA, complementary DNA CHO K1, Chinese hamster ovary K1 DMSO, dimethyl sulfoxide DNA, deoxyribonucleic acid EDTA, ethylenediaminetetraacetic acid EGF, epidermal growth factor EGTA, ethylene g1ycol-bis(2-aminoethylether)-N.N,N'. '-tetraacetic acid ERK, extracellular signal-regulated kinase GST, glutathione S-transferase JN K, c-Jun N—terminal kinase LeTx, lethal toxin (anthrax lethal toxin) LF, lethal factor (anthrax lethal factor MAP, mitogen-activated protein MAPK, mitogen-activated protein kinase MAPKK, mitogen-activated protein kinase kinase MEK, mitogen-activated protein kinase/extracellular signal-regulated kinase kinase MEKcr, cleavage-resistant form of MEK MKK, mitogen—activated protein kinase kinase mRN A, messenger RNA PA, protective antigen PMA, phorbol 12-myristate 13-acetate PTEN, phosphatase and Lensin homolog xiii Ras, rat sarcoma viral oncogene homolog RNA, ribonucleic acid SDS, sodium dodecyl sulfate shRNA, small hairpin RNA siRNA, small interfering RNA SV40, simian virus 40 TPA, 12-O-tetradecanoylphorbol-1 3-acetate UV, ultraviolet YopJ, Yersz'nia outer protein J xiv Chapter I. General introduction: MEK signaling pathway and cancer 1.1. Introduction to MEK signaling pathway 1.1.1. Exploring the Raf-MEK-ERK signaling pathway In late 19803, Ray and Sturgill (1987; 1988a) observed that soluble fi'actions of cell extracts prepared from insulin-treated 3T3 —L1 adipocytes were capable of phosphorylating the microtubule—associated protein 2 (MAP-2) protein. Initially this protein kinase was termed microtubule-associated protein (MAP) kinase, as it catalyzed the phosphorylation of MAP-2 on serine and threonine residues (Sturgill & Ray, 1986). Later, MAP kinase activity was shown to be stimulated not only by insulin but also by other mitogenic factors such as serum, insulin-like growth factors, epidermal growth factor (EGF), and phorbol 12—myristate 13-acetate (PMA; also called TPA) (Rossomando et al. , 1989; Erickson et al., 1990). Therefore, this enzyme was also named mitogen- activated protein (MAP) kinase (Rossomando et al., 1989) or extracellular signal- regulated kinase (ERK) (Boulton et al., 1990). Since growth factor—stimulated MAP-2 phosphorylation was accompanied by DNA synthesis and cell cycle progression (Sato et al., 1985; Sato et al., 1986), Ray and Sturgill’s seminal discovery Opened the way to exploring the MAP kinase signal transduction pathway through which these cellular events were regulated. Ray and Sturgill (1988b) also showed that ERK was phosphorylated at threonine and tyrosine residues upon stimulation, and serine/threonine phosphatase as well as tyrosine phosphatase could completely deactivate ERK (Anderson et al., 1990). These findings indicated that ERK itself was a phosphoprotein and that activation of ERK involved phosphorylation by another protein kinase having threonine and tyrosine specificity. Soon after, the activator for ERK was identified and purified from EGF- stimulated cells by Ahn (1991) and colleagues (Seger et al., 1992). This dual-specificity protein kinase (both serine/threonine and tyrosine specificity) (Gomez & Cohen, 1991; Nakielny et al., 1992; Seger et al., 1992) was called MAP kinase kinase (MAPKK, MAP2K or MKK) (Gomez & Cohen, 1991) or MAPK/ERK kinase (MEK) (Crews & Erikson, 1992). Similarly, MEK was characterized as a phosphoprotein that required serine/threonine phosphorylation for its own kinase activity (Gomez & Cohen, 1991). Serine/threonine protein phosphatase 2 (PP2A) —- but not tyrosine phosphatases -— was shown to inactivate MEK activity (Gomez & Cohen, 1991; Nakielny et al., 1992). This indicated that downstream of the growth factor receptors (which were tyrosine kinases) there must be at least one serine/threonine protein kinase that functioned as the MEK activator. Subsequent to this, the Raf family of protein kinases, cellular homologues of the acutely transforming viral oncogene v-raf(Rapp et al., 1983), were identified as direct activators of MEK (Dent et al., 1992; Kyriakis et al., 1992). c-Mos has also been identified as a kinase that activates MEK, though its normal expression is limited to oocytes (Sagata et al., 1988). 1.1.2. Activation mechanism of the MAP kinase cascade MAP kinase pathways are evolutionarily conserved signaling pathways that transduce signals from the cell membrane to the nucleus in eukaryotic cells (reviewed by English et al., 1999; Chen et al., 2001; Pearson et al., 2001; Johnson & Lapadat, 2002; Roux & Blenis, 2004). Signal transduction through the Raf-MEK-ERK pathway (Figure 1) begins with the binding of extracellular growth factors to specific transmernbrane receptors. This results in dimerization and/or conformational changes in the receptors, leading to a series of cytoplasmic protein recruitment/activation steps that include the Raf family of proteins. Raf then activates MEK] and MEK2, the only known MEK isoforms, by phosphorylating them on the conserved serine residues in the activation loop (Ser218/Ser222 of human MEKl and Ser222/Ser226 of human MEK2) (Alessi et al., 1994; Zheng & Guan, 1994). Active MEK] and MEK2 then in turn activate ERK1 and ERK2, the only known substrates of MEK, by phosphorylating the threonine and tyrosine residues in the conserved T-E-Y motif (Thr202/Tyr204 of human ERK] and Thr185/Tyr187 of human ERK2) (Payne et al., 1991). After activation, ERK phosphorylates and activates downstream effectors regulating a variety of cellular events at the transcriptional and posttranslational levels (reviewed by English et al., 1999; Chen et al., 2001; Pearson et al., 2001; Johnson & Lapadat, 2002; Roux & Blenis, 2004). Why does the MAP kinase pathway employ three tiers of protein kinases to transduce signals instead of one? To simulate MAPK phosphorylation and activation, Huang and Ferrell (1996) constructed an elegant mathematical model and then tested predictions Of this model in cytoplasmic extracts oernopus oocytes. They found that whereas Raf behaved as a typical Michaelis-Menten enzyme, MEK and ERK behaved as “ultrasensitive” enzymes. In other words, MEK and ERK are relatively less sensitive to low concentrations of stimuli but above a threshold level stimuli evoke a strikingly rapid response. This makes MEK and ERK respond to stimuli in an all-or-none fashiOn, like a switch. Moreover, according tO their model, the ultrasensitivity of this reaction is directly linked to the dual phosphorylation that is required to activate MEK and then ERK. This observation has profound functional implications and Should be an important consideration in designing strategies to inhibit this pathway. 1.2. Involvement of MEK signaling pathway in human cancers The Raf-MEK-ERK pathway is an important signaling pathway that, in response to growth factor signals, promotes cell cycle progression and cell proliferation (reviewed by English et al., 1999; Chen et al., 2001; Pearson et al., 2001; Johnson & Lapadat, 2002; Roux & Blenis, 2004). Therefore, it is not surprising that deregulated activities of the kinases in this pathway disrupt growth control, which can lead to cancers. The hypothesis that elevated activity of the Raf-MEK-ERK signaling pathway may lead to cancer was tested more than ten years ago when constitutively active MEK-ERK signaling was shown to be sufficient to transform mammalian cells (Mansour et al., 1994a). In support of this, screening of somatic mutations in a panel of human cancer cell lines identified the presence of the constitutively active B-Raf mutant V600E, which results in a persistent activation of the downstream pathway, in 66% of human melanomas and in a lower percentage of other types of cancers (Davies et al., 2002). Based on these findings, targeting the Raf-MEK-ERK signaling pathway became a prospective strategy for treating human cancers. 1.3. Targeting the Raf-MEK-ERK pathway as a strategy for treating cancers In principle inhibitors can be designed that will block any step in this pathway (reviewed by Kohno & Pouyssegur, 2003; Sebolt-Leopold & Herrera, 2004; Kohno & Pouyssegur, 2006). However, because this is an amplifying signal cascade, it makes most sense to block that signal as close to the stimulus (receptor tyrosine kinase) as possible. This is particularly true in this case as MEK and ERK respond to stimuli in an all-or-none fashion. Therefore, it might be argued that Raf kinases are the preferred point of intervention. However, in an unexpected twist several investigators now report that B- Raf inhibitors paradoxically can activate MEK-ERK signaling in cells that express wild- type B-Raf (Hatzivassiliou et al., 2010; Heidorn et al., 2010; Poulikakos et al., 2010). It appears that the binding of ATP-competitive B-Raf inhibitors Causes conformational changes in the kinase that promotes dirnerization and transactivation of non—drug-bound C-Raf in an active Ras-dependent fashion. So, while it makes sense to target tumors harboring B-Raf mutations with B-Raf inhibitors, it may actually be disadvantageous to use these drugs to treat tumors with wild-type B-Raf Since they may enhance proliferation of tumor cells and may have adverse effects on normal cells with active Ras. In this case MEK inhibitors may be the most appropriate therapeutic option. A great deal of effort has gone into looking for highly selective MEK inhibitors. Many Of these are commonly used in laboratories as powerful tools in the study of the MEK-ERK signaling pathway, and select inhibitors are currently in cancer clinical trials, having shown promising anti-cancer activity in preclinical studies. In the following sections, 1 will review the discoveries, properties, and clinical applications of two categories of highly selective MEK inhibitors: non—ATP-competitive small-molecule inhibitors and biological inhibitors. By focusing on their MEK selectivity and inhibition mechanisms, the following review will provide insights into the potential of these inhibitors as tools for studying the Raf-MEK-ERK pathway and as anti-tumor agents. 1.3.1. Small-molecule inhibitors In this section, the best known and commonly used non-ATP-competitive MEK inhibitors will be reviewed. These inhibitors result in MEK-specific inhibition by an allosteric mechanism which contributes to highly selective inhibition of MEK without affecting other protein kinases that have structurally similar ATP binding pockets. 1.3.1.1. PD 098059 By screening a compound library using an in vitro kinase cascade phosphorylation assay, Dudley et al. (1995) identified PD 098059, 2-(2'-aminO-3'- methoxyphenyl)-oxanaphthalen-4-one (Figure 2a), as a synthetic inhibitor of the MAPK pathway. PD 098059 inhibits MEK (IC50 = 10 uM, see Table I) but not other components of the MAPK pathway. In later studies, PD 098059 showed no significant effect on a spectrum of other protein kinases, including protein kinase A (PKA), protein kinase C (PKC), and other kinase members in the MAPK family pathways such as p38 MAP kinase, c-Jun NHz-terminal kinase (JN K), p38 MAP kinase kinase, and MKK4 (Alessi et al., 1995; Davies et al., 2000). Kamakura et al. (1999) showed that PD 098059 had no effect on MEKS (a MEK/MKK family member which is independent from the MEKl/Z-ERKl/Z pathway) in vitro, but did have an inhibition effect on intracellular MEKS activity. This was later confirmed by Mody et al. (2001 ), although a high concentration (100 uM) of PD 098059 was required. PD 098059 inhibits growth factor— stimulated MAPK activation and DNA synthesis in Swiss 3T3 cells (Dudley et al., 1995), suggesting the membrane permeability and in-cell inhibitory activity of PD 098059. Initial observation showed that PD 098059 did not inhibit MEK] that had been maximally phosphorylated by c-Raf (Alessi et al., 1995; Ahn et al., 2001), raising a possibility that PD 098059 did not bind to the active site of MEK but instead bound to another allosteric pocket. The allosteric inhibition mechanism was later confirmed by using classical Michaelis-Menten methods (Favata etal., 1998). 1.3.1.2. U0126 U0126, 1 ,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene (Figure 2b), was the second compound identified as an allosteric inhibitor of MEK. Favata et al. (1998) screened a total of 40,000 compounds for their ability to inhibit activator protein 1 (AP-l)—driven transcription using a luciferase reporter assay. They reported that U0126 inhibited both AP-l—driven gene expression and TPA-induced up-regulation of c-Jun and c-Fos at the protein and the mRNA levels, suggesting that U0126 targeted the upstream activators of c—Jun and c-Fos. Indeed, U0126 inhibits MEK (see Table I), but not other MKK members and MAP kinases (Davies et al., 2000) except for MEKS (Kamakura et al., 1999; Mody et al., 2001). In addition, U0126 showed a 100-fold greater potency than PD 098059 (Favata et al., 1998). This was later confirmed by Davies et al. (2000). Consistent with PD 098059 studies, U0126 has a much lower effect on activated wild- type MEK than constitutively active MEK (Alessi et al., 1995; Ahn et al., 2001), suggesting that the conformational change in MEK upon activation interferes with the binding of these inhibitors. In addition, this implied that these two MEK inhibitors might not compete with the ATP binding pocket of MEK. Using classical Michaelis-Menten enzyme modeling, Favata et al. (1998), confirmed the non—ATP-competitive mechanism of these MEK inhibitors the structural basis of which will be discussed in detail later in this section. The results of in vitro kinase assays using purified proteins suggested that PD 098059 and U0126 inhibited the phosphorylation and activation of MEK but not the ability Of MEK to phosphorylate ERK (Alessi et al., 1995; Ahn et al. , 2001). However, in later studies, the inhibitory effect of PD 098059 and U0126 on MEK phosphorylation was not Observed in intact cells when using antibodies specifically against phosphO-MEK (Ahn et al., 2001). A possible explanation for this controversy is that the action of these two inhibitors in intact cells is mediated by inhibition Of MEK phosphoryl-transferase activity, and that both inhibitors may interfere with Raf-dependent MEK phosphorylation in vitro, only when other intracellular components are absent. This controversy was also pointed out and discussed by Kohno and Pouyssegur (2003). PD 098059 and U0126 are widely used in laboratories as powerful tools for studying the MEK signaling pathway. Both molecules are commercially available and have thousands of entries in the PubMed database. However, neither of them has entered clinical trials due to their limited in viva bioactivity. 1.3.1.3. PD 184352 (CI-1040) PD 1843 52 (Figure 2c), also known as CI—1040, was identified by Sebolt—Leopold et al. (1999) as a potent MEK inhibitor (IC50 = 17 nM; see Table I) in an in vitro kinase cascade assay using GST-fusion proteins of MEKl and MAPK. PD 184352 has a higher potency than PD 098059 and U0126 (Table I), and has no effect on other kinases tested (Sebolt-Leopold et al., 1999; Davies et al., 2000). Similar to PD 098059 and U0126, PD 1843 52 inhibits intracellular MEKS activity at a much higher concentration than that required to inhibit the activation of ERK (Mody et al., 2001). PD 1843 52 is not competitive with ATP or ERK (Sebolt-Leopold et al., 1999), suggesting a similar allosteric inhibition mechanism for PD 184352 as for PD 098059 and U0126. The cellular consequences of MEK inhibition by PD 1843 52 include a complete suppression of platelet-derived growth factor (PDGF)-induced ERK activation, induction of G1 cell cycle arrest, reversion to untransformed morphology, and inhibition of anchorage- independent growth in colon cancer cells as well as in other cancer cell lines (Table I) (Sebolt-Leopold et al., 1999). Interestingly, PD 1843 52 showed a stronger effect on three colon cancer cell lines that had high ERK activity relative to cancer cell lines with low ERK activity, suggesting that the inhibitor is more potent for cancer cells that are addicted to MEK signaling. This was further confirmed by Solit et al. (2006), who showed that the B-Raf V600E oncogenic mutation in cancer cell lines was associated with enhanced and selective sensitivity to PD 184352 when compared with B-Raf wild- type cell lines. In addition to the in vitro effects, Sebolt-Leopold et al. (1999) successfully showed that PD 1843 52 actively suppressed ERK activation in colon cancer cell xenografi tumors in mice within one hour after oral administration. More importantly, this in vivo efficacy was achieved over a wide range of doses (48—200 mg/kg, two treatments per day for 14 days) without signs of toxicity. This was the first study demonstrating the in vivo efficacy of MEK inhibition by a small-molecule inhibitor. A phase I study of PD 184352 was conducted to test the toxicity, pharmacokinetics, pharmacodynamics, maximum tolerated dose, food effect, and clinical activity in patients with advanced cancers, including colorectal, non-small cell lung, pancreatic, and kidney cancers, as well as melanoma (Allen et al., 2003; LoRusso et al., 2005b). PD 1843 52 was administrated orally in single to multiple daily doses from 100 mg to 1,600 mg for 21 days, followed by 7 days off, for a total of three cycles. After the maximum tolerated dose profile was determined, the “holiday” week between treatment cycles was removed to test the safety profile of continuous dosing. Blood and urine samples were collected throughout the treatment for pharrnacokinetics and biomarker assessments. Phospho-ERK levels were assessed in blood samples and tumor'biopsies to determine the clinical targeting activity of PD 1843 52. The study results showed that PD 184352 was well tolerated at a treatment of 800 mg twice daily, with common grade 1 or 2 drug-related adverse events including diarrhea, asthenia, rash, nausea and vomiting. Although this was a phase I study initially designed only to assess toxicity, some patients showed promising signs of efficacy: one out of 66 patients had partial response lasting 355 days, and 19 patients (29%) achieved stable disease for 4-17 months with a median of 5.5 months. Based on these results, a phase II trial was initiated to assess the antiturnor activity and safety of PD 1843 52 in patients with advanced breast, colon, non-small-cell lung, and pancreatic cancers (Rinehart et al., 2004). In this study, PD 1843 52 was given to patients at 800 mg twice daily and continuously, and was reduced later in ZOO-mg decrements for patients with grade 2 or greater adverse event severity. Continuous PD 184352 treatment exceeding one year was well tolerated without cumulative toxicities, and 81% of the patients experienced only grade 1 or 2 adverse events. However, PD 1843 52 was terminated in the phase II trial because no complete or partial responses were 10 observed, although eight patients (12%) achieved stable diseases lasting 4-18 months with a median of 4.4 months. The change in ERK activity in the tumors after PD 184352 treatment was not available from the phase II study as the study was not designed to assess it in tumor biopsies. However, in the earlier phase I study, ERK activation was inhibited by 46% to 100%, with an average of 73% in tumor biopsies from ten out of ten (100%) patients (LoRusso et al., 2005b). This suggests that the lack of response to PD 184352 in the phase II study might not be solely due to incomplete inhibition of ERK activity in the tumors. In addition, the authors of this study also attributed the lack of response to the poor metabolic stability of PD 184352. PD 0325901, a PD 184352 analog, was then developed as a second-generation MEK non—ATP-competitive inhibitor with improved potency and pharmaceutical properties, and it has Since entered clinical trials (discussed immediately below). 1.3.1.4. Derivatives of PD 184352 PD 0325901 (Figure 2d), an analog of PD 184352, is also a non—ATP-competitive inhibitor and possesses high selectivity for MEK (Sebolt-Leopold et al., 2004). PD 0325901 has a 500-fold higher cellular effect on ERK activation than does PD 1843 52 (see Table I). It is believed that this improvement is the result of several factors, including better solubility and higher metabolic stability. Similar to PD 1843 52, PD 0325901 has higher potency for xenograft tumors harboring the B-Raf V6OOE mutation than for tumors with wild-type B-Raf (Solit et al., 2006). In recent studies, PD 0325901 showed in vivo potency for oncogenic B-Raf-induced lung adenocarcinoma and 11 melanoma (Dankort et al., 2007; Dankort et al., 2009a). More importantly, in preclinical studies, PD 0325901 suppressed ERK phosphorylation by more than 50% at a single oral dose of 25 mg/kg at 24 hours after treatment. In comparison, PD 1843 52 suppressed ERK phosphorylation at a much higher dose of 150 mg/kg for only eight hours, and the phosphO-ERK level returned to the control level within 24 hours after treatment (Sebolt- Leopold et al., 2004). The improved biological profile and promising preclinical activity of PD 03 25901 moved it into a phase 1-2 clinical study Of patients with advanced cancers, including breast, colon, and non-small-cell lung cancers as well as melanoma (LoRusso et al., 2005a; Menon et al., 2005). Based on preclinical studies, cycles of PD 0325901 were administrated orally from 1 mg once daily to 1, 2, 4, 8, 15, 20 or 30 mg twice daily for 21 days and then repeated every four weeks. The study was designed to focus on pharmacokinetics, pharmacodynamics, and effectiveness. For this purpose, before and during treatment, tumors were biopsied to assess ERK activation and proliferation index, and blood samples were collected to measure the amount of circulating drug and its metabolite. The results showed that PD 0325901 was well tolerated. In addition to common toxicities (including rash, diarrhea, nausea, and vomiting), some patients had visual effects including blurred vision and halos. Strong inhibitory effects of PD 0325901 treatment on phospho-ERK and proliferation marker Ki67 were observed at a minimum dose of 2 mg twice daily: ERK phosphorylation was consistently suppressed by an average of 84%, while cell proliferation (measured by Ki67 irnmunostaining) declined by an average of 60%. Although one partial response (melanoma) and five stable disease cases (four melanoma and one non-small-cell lung carcinoma) were Observed, this study 12 was terminated prematurely in 2007 mainly due to the safety concerns about the ocular and neurological toxicity that presented at 10 mg twice daily and higher (ClinicalTrials. gov Identifier: NCT00147550). As both PD 1843 52 and PD 0325901 have failed in clinical trials, Pfizer recently has developed and investigated a series of PD 1843 52 derivatives, among which one compound has showed relatively superior ERK inhibition and better solubility then PD 1843 52 [compound 12a reported by Warmus et al. (2008)]. More importantly, the metabolic stability of this compound is much better than that of PD 184352. 1.3.1.5. ARRY-142886 (AZD6244) ARRY-142886, or 6-(4-bromO-2-chloro-phenylamino)-7-fluorO-3 -methyl-3H- benzoimidazole-S-carboxylic acid (2-hydroxy-ethoxy)-amide (Figure 2g), also called AZD6244, was disclosed in 2004 as another second-generation MEK small molecular inhibitor (Wallace et al., 2004). Like PD 0325901, ARRY-142886 was developed based on the PD 1843 52 structure, and therefore was highly selective for MEK and noncompetitive with ATP (Lyssikatos et al., 2004; Yeh et al., 2007). In an in vitro assay, ARRY-l42886 inhibited the activity of purified MEK with an IC50 of 14.1 nM (Table I), but had no effect on more than 40 other kinases and a minimal inhibitory effect on MEKS (Y eh et al., 2007). Similar to other MEK noncompetitive inhibitors, ARRY-l42886 inhibits MEK activity but not the phosphorylation and activation of MEK in intact cells (Y eh et al., 2004; Yeh et al., 2007). ARRY-142886 has been shown to inhibit intracellular phosphorylation of ERKl/2 in several cancer cell lines including melanoma, colon, and pancreatic cancer lines, with an IC50 below 40 nM (Table I), but has no effect 13 on MEKS (Y eh et al., 2007; Haass et al., 2008). Cellular responses to ARRY-142886 include an inhibition in proliferation and cell cycle arrest at the G1/ S transition (Y eh et al., 2004; Yeh et al., 2007; Haass etal., 2008), whereas prolonged treatment (72 h) results in only limited apoptosis in some cell lines (Haass et al., 2008). The cytostatic inhibitory effect of ARRY-l42886 was also observed in a three-dimensional culture model of melanoma (Haass et al., 2008). Like an earlier study of PD 184352 (Solit et al., 2006), ARRY-142886 had a much stronger inhibitory effect on cancer cells harboring Ras or Raf oncogenic mutations than on those without the mutations or on normal fibroblast cells (Y eh et al., 2004; Yeh et al., 2007; Haass et al., 2008). Moreover, ARRY-142886 is active when given orally and shows in vivo efficacy. In mouse xenografi models, ARRY-l42886 not only inhibits the growth of colon cancer and melanoma in a dose-dependent manner, but also induces pancreatic xenografi tumor regression (Lee et al., 2004; Yeh et al., 2007; Haass et al., 2008). Although ARRY- 1 42886 effectively inhibits melanoma xenograft tumor growth, it does not induce apoptosis in the tumor cells unless combined with microtubule- stabilizing agents (Haass et al., 2008). Interestingly, xenograft tumor growth recovers after cessation Of ARRY-l42 886 administration, and the tumors regress again upon re- administration (Y eh et al., 2007). This supports the idea that the effect of ARRY-142886 is cytostatic as Opposed to cytotoxic and that continuous treatment may be necessary to maintain the inhibitory effect. With its promising in viva antiturnor activity, ARRY-142886 entered phase I clinical trial in 2004, which was initiated to assess the maximum-tolerated dose, pharmacokinetics, and pharmacodynamics in patients with advanced cancers, including 14 breast cancer, colorectal cancer, and melanoma (Adjei et al., 2008). This study had two parts. Part A was to determine the maximum-tolerated dose. Patients were orally given ARRY-142886 at doses Of 50, 100, 200, or 300 mg twice daily for 28-day cycles. Although the maximum-tolerated dose Obtained from part A was 200 mg twice daily, this dose was discontinued in the second part of the study (Part B) because a substantial number of patients in Part A required dose reductions, treatment holidays, or even termination due to drug-related toxicities including rash, diarrhea, nausea, fatigue, and blurred vision. Therefore, a dose of 100 mg twice daily was recommended for the Part B study. The results showed that ARRY-l428 86 was well tolerated at 100 mg twice daily with grade 1 or 2 adverse events. Promising targeting effects of ARRY-142886 were observed in post-treatment tumor samples: ERK phosphorylation was strongly inhibited by an average of 79%, while greater than 50% inhibition of cell proliferation (measured by Ki67 irnmunostaining) was seen in 25% of the patients. One patient enrolled in this study with malignant melanoma had a 70% reduction of tumor size after three cycles of treatment but eventually developed brain metastases. Although no obj ective response was achieved in the phase I study, nine patients (16%) had stable disease for more than five months. Based on the promising targeting effect and recommended dosage Obtained from the phase I study, ARRY-l42886 is now in several phase II studies, including studies involving combination therapies. 1.3.2. Mechanism of allosteric inhibition Initially both PD 098059 and U0126 were found to have a stronger effect on less active MEK than highly active MEK (Alessi et al., 1995 ; Favata et al., 1998), suggesting 15 that these kinase inhibitors do not bind to the enzyme active site of MEK. In addition, Favata et al. (1998) also confirmed the non—ATP-competitive mechanism of these MEK inhibitors by using classical Michaelis-Menten methods. While these inhibitors do not compete with ATP or ERK, they do compete with each other, indicating that the inhibitors share a common binding site independent of the ATP or ERK binding sites (Favata et al., 1998). Ohren et al. (2004) identified the binding pocket of non—ATP- competitive MEK inhibitors by co-crystallizing MEK with MgATP and two PD 184352 derivatives [PD 318088 (Figure 2e) for MEKl and PD 334581 (Figure 2f) for MEK2] as ternary complexes with satisfactory resolutions. The crystal structures indicate that the MEK inhibitors bind to MEK in a pocket adjacent to the MgATP binding Site. The inhibitor binding pocket and the MgATP binding pocket are physically separated by the side chains Of Lys97 and Met143, which are conserved in the MEK active site. This was the first structural study directly revealing the allosteric binding pocket of these MEK inhibitors. Ohren et al. (2004) also showed that the diaryl amine (A ring and B ring, Figure 2c-g) structure of PD 184352 and its derivatives — including PD 0325901, PD 318088, PD 334581, and ARRY-142886 —— was the key to stabilizing the inhibitors in the allosteric binding site. The A ring and the B ring form a crucial hydrogen-bond interaction and numerous non—covalent interactions, respectively, with the hydrophobic pocket formed by several residues. The binding of the inhibitors results in a series of conformational changes in MEK, leading to interference with the catalytic residue Lys97 (MEKl) and thus inactivation of MEK activity. In other words, upon inhibitor binding, MEK adopts a “closed” conformation of an active protein kinase, and the phosphoryl- 16 transferase activity of MEK is inhibited. This is supported by a fact that PD 1843 52 treatment inhibits ERK activation in human melanoma SK-MEL-28 cells without affecting the activation of MEKl (Figure 3). The structures of MEKl and MEK2, co- crystallized with MgATP plus the inhibitor, share high homology (Ohren et al., 2004), indicating that the same mechanism of inhibition is involved in both. Most recently, it was shown that U0126 binds to MEK in the same pocket as PD 0325901 (Fischmann et al., 2009). Since the two earliest non—ATP-competitive MEK inhibitors, PD 098059 and U0126, compete with each other (Favata et al., 1998), it is very likely that all of the MEK inhibitors discussed above bind to the same pocket and use the same mechanism to inhibit MEK. The high selectivity of these allosteric inhibitors for MEK can be explained on the basis of structure. First, these inhibitors do not compete with ATP, the major intracellular energy source for all protein kinases; this decreases the binding affinity of the inhibitors to other kinases. Second, these inhibitors bind to MEK in a unique binding pocket, which is formed by several residues of MEK and stabilizes the inhibitor binding. This pocket increases the MEK:inhibitor binding affinity as well as the complexity of the binding, resulting in a high selectivity for MEK. Since the diaryl amines of these MEK inhibitors are such a crucial structure for this MEK-specific inhibition mechanism, all the second-generation MEK allosteric inhibitors have been developed based on this structure in order to maintain the mechanism (W armus et al., 2008). 17 1.3.3. Biological inhibitors 1.3.3.1. Anthrax lethal toxin The MEK-specific inhibitory activity of anthrax lethal toxin (LeTx) was revealed in the National Cancer Institute Antineoplastic Drug Screen project (Duesbery et al., 1998). In this study, sixty human cancer cell lines (termed NCI-60 cell lines) were tested for their sensitivity to a panel of anti-neoplastic molecules. It was shown that the sensitivity profile of the MEK inhibitor PD 098059 was very similar to that of LeTx, raising the possibility that they sensitized the cells by a similar mechanism. Based on this, Duesbery et al. (1998) characterized the MEK-specific protease activity of LeTx. About the same time, Vitale et al. independently demonstrated the same proteolysis of MEK by LeTx (V itale et al., 1998). Anthrax lethal toxin is a binary toxin secreted by the bacterium Bacillus anthracis. It is composed of a binding moiety called protective antigen (PA) and an enzymatic moiety, lethal factor (LF). During cellular intoxication, PA binds to the anthrax receptors expressed on the host cell surface and then is cleaved by a furin-like protease. This cleavage removes a 20-kDa N-terminal fragment and leaves a 63-kDa C-terminal truncation, PA63, on the cell surface. PA63 then forms an Oligomerized channel to which the LF binds. After the binding of LF, the complex is internalized into cells through the endosomal internalization pathway. The acidic environment of the endosome causes a conformational change in PA63 that results in the formation of pores through which LP is released into cytosol [reviewed by Singh et al. (2005)]. Since its initial identification as a MEK1/2-specific protease (Duesbery et al., 1998), additional members of the MAP kinase kinase family, including MKK3, 4, 6 and 7 l8 but not MEKS, have also been found to be substrates of LF [(Vitale et al., 1998; Pellizzari et al., 1999; Vitale et al., 2000) and discussed by Duesbery et al. (2001)]. However, unexpected results from this dissertation research suggested that MKK7 might not be a preferred LF substrate. This will be discussed in Chapter 11. LF specifically cleaves the N-terminus Of MEK and other MKK proteins at the consensus cleavage site: three to four basic or proline residues followed by three variable residues followed by an aliphatic residue: (B/P)3_4-(X)3-Al (T able II). The N-termini of this family Of proteins harbor MAP kinase docking sites that are required for the interaction between MEK/MKK and MAPK (Tanoue et al., 2000). Not surprisingly, after LF cleavage, the C-terminal part of MEK/MKK, which contains the kinase domain, loses its affinity for the downstream MAP kinases (Chopra et al., 2003; Bardwell et al., 2004). In addition, biochemical evidence has Shown that loss of the amino terminus may destabilize MEK, leading to the decreased intrinsic kinase activity that is Observed following LF-mediated proteolysis (Chopra et al., 2003). Thus, cleavage Of MEK/MKK by LF results in a blockade of not only the ERK pathway but also of the p38 MAPK and JNK pathways. Because increased MAP kinase activity has been detected in many types of human cancers, LeTx has potential as a cancer therapeutic. The first evidence for the anti-tumor activity of LeTx was provided by Friedlander et al. (1998). Duesbery et al. (2001) then assessed its effects on cancer cells and found that LeTx not only efficiently inhibited the proliferation and the soft agar colony formation of oncogenic Ras- transformed NIH-3T 3 cells, but it also caused the cells to revert to a nontransformed morphology. LeTx treatment also inhibited the growth of Ras-transformed cells implanted in nude mice, with no overt toxicity. Interestingly, LeTx-treated xenografr l9 tumors had a pale appearance whereas control tumors were dark red-purple. This study demonstrated the in viva antitumor activity of LeTx and also suggested that LeTx could inhibit tumor growth by targeting cell proliferation pathways as well as angiogenesis pathways. This finding has been confirmed in several tumor models including melanoma (KOO et al., 2002; Abi-Habib et al., 2005), Kaposi's sarcoma (Depeille et al., 2007), fibrosarcoma (Ding et al., 2008), kidney cancer (Huang et al., 2008), neuroblastoma, and colorectal adenocarcinoma(Rou1eau et al., 2008) cells in vitro, as well as in xenograft tumor growth in viva. In the earliest xenografi studies, LeTx was not administrated systemically, but intratumorally. In these studies it was noted that intraturnoral injection of LeTx had a growth inhibitory effect on distal, uninjected tumors (Duesbery et al., 2001; Koo et al., 2002). These observations revealed the systemic anticancer potential of LeTx. Abi-Habib et al.’ (2006a) then determined the in viva potency and safety of systemically (intraperitoneally) administrated LeTx in athymic nude mice bearing human melanoma xenografl tumors. In this study, the ratios of PA to LP giving the highest potency were determined to be 3:1 and 5: 1, and LeTx was well tolerated at a cumulative dosage of 24 pg total LF at a 5:1 ratio of PA to LP. An LDlo for a cumulative dose was estimated at 30—36 pg total LF. Since this preclinical study, a standard LeTx systemic treatment has been established for human cancer xenograft tumors in nude mice: 10 pg of PA and 2 pg of LF are pre—mixed and injected intravenously through tail vein every other day, for a total of six injections in two weeks (a cumulative does of 12 pg total LF, which is less than half the LD10 dose). At 12 pg, LeTx effectively inhibits the growth of xenografi tumors without apparent animal toxicity (Depeille et al., 2007; Ding et al., 2008; Huang 20 et al., 2008). In addition LeTx has been modified to selectively target cancer cells expressing high levels of urokinase plasminogen activators or matrix metalloproteases, features of metastatic cancer cells (Abi-Habib et al., 2006b; Liu et al., 2008). Several lines of evidence support the conclusion that LeTx acts in an anti- angiogenic fashion. First, as noted above, gross inspection reveals that LeTx-treated tumors have a pale appearance when compared to untreated tumors. Second, the mean vascular density for LeTx-treated tumors is dramatically reduced (Depeille et al., 2007; Ding et al., 2008; Huang et al., 2008). Finally, a recent study using high-resolution ultrasound enhanced with contrast microbubbles revealed the rapid (within 24 hours) inhibitory effect of LeTx on the perfusion of fibrosarcoma xenografi tumors, but interestingly not in non-tumor host tissues (Ding et al. , 2008). This suggests that the inhibitory effect of LeT x on endothelial function is highly selective for tumor-associated blood vessels. Although it is clear that LeTx reduces tumor vascularity, the mechanism by which it does this is uncertain. The initial explanation that LeTx acted directly on tumor cells to suppress the release of pro-angiogenic factors was supported by in vitro studies demonstrating that LeTx suppressed release of angiogenic cytokines such as vascular endothelial growth factor (VEGF), interleukin-6 (IL-6) and basic fibroblast growth factor (bFGF). However, this hypothesis was questioned after LeTx was demonstrated in subsequent studies to inhibit the growth of tumors that do not express toxin receptors (Liu et al., 2008). Moreover, results from this dissertation research show that systemic LeTx administration can inhibit growth of human melanoma SK-MEL—28 xenograft tumors without directly targeting the MEK-ERK pathway in tumor cells (presented in Chapter 21 III). These observations indicate that LeTx targets a non-tumor compartment, perhaps endothelial cells. In support of this, endothelial cells have been reported to be potential targets of LeTx (Kirby, 2004; Warfel et al., 2005; Alfano et al., 2009), and the MEK signaling pathway is required for tumor-associated endothelial function (Mavria et al., 2006). Regardless of the cellular target, these Observations establish LeTx as an anti- angiogenic agent. However, LeTx appears to be unique in this aspect since an anti- angiogenic effect of other MEK inhibitors has not been reported. Thus this activity may be related to its effects on other MKK signaling pathways In summary, LeTx is a highly selective biological inhibitor of MEK and most other members in the MK protein family, and it shows very good potential to be a potent cancer-targeting therapeutic. Although LeTx is unlikely to enter clinical trials owing to its toxicity, it has become a powerful tool for studying the involvement of MEK/MKK signaling pathways, not only in cancers but also in other diseases such as inflammatory diseases (Pellizzari et al., 1999; Park et al., 2002; Agrawal et al., 2003) and eye diseases (Bromberg-White et al., 2009). 1.3.3.2. YopJ YopJ is a member of the Yersinia outer protein (Y op) family of effectors proteins. The Yops are virulence factors produced by Yersinia species including Yersinia pestis, the bacterial pathogen that caused the bubonic plague or Black Death in the Middle Ages. Upon infection, Yops are translocated into host cells through a contact-dependent type III secretion system. Initial studies showed that YopJ was necessary and sufficient for the pathogen to inhibit multiple eukaryotic signaling pathways, including all the parallel 22 MAPK pathways (Palmer et al. , 1998; Palmer et al., 1999) and the nuclear factor kappa B (N F -KB) pathway (Ruckdeschel et al., 1998; Schesser et al., 1998). Inhibition of these multiple signaling pathways by YopJ results in disruption of cytokine secretion (which in turn disrupts the innate immune response) and an induction of apoptosis in infected cells. Later, Orth et al. (1999) confirmed the direct and physical interactions of YopJ with multiple signaling molecules (including IKKB, an NF-ch activator, and MKKl-S), but not with the members of MKKK or MAPK. This finding suggested that YopJ inhibited these signaling pathways by a mechanism that required direct binding to these signaling molecules. Sequence comparisons showed that YopJ has a secondary structure similar to that of the adenoviral protease (AVP) family. From this, Orth et al. (2000) demonstrated the protease activity of YopJ and suggested that YopJ proteolytically inhibited the MAPK and NF-IcB pathways. However, whether proteolysis was the mechanism for YopJ inhibition of the MAPK and NF -KB pathways was questionable, because the total amount and molecular weight of MKK or IKKB were not affected by YopJ treatment (Orth et al., 1999) The biochemical mechanism for YopJ-mediated inhibition of eukaryotic signaling pathways was not elucidated until recently, when studies identified YopJ as an O-acetyl transferase that acetylates the serine/threonine residues in the activation loop Of MEK/MKK and IKK (Mittal et al., 2006; Mukherjee et al., 2006). Once acetylated, those residues can no longer be phosphorylated and activated, resulting in inhibition of the downstream MAPK and NF-ch pathways. A recent mutagenesis study has identified the conserved G a-helix in the kinase domain of MEK as a YopJ-binding motif (Hao et al., 2008). In the study, a PCR random mutagenesis library was screened for mutant 23 Psz (MKK yeast homologue) proteins that were able to suppress YopJ-induced growth inhibition in yeast. The conserved Ile579 residue of Pbs2 (corresponding to Ile317 of human MEKl) was shown to be required for YopJ to bind and acetylate Psz. This residue is in the conserved G a-helix in the kinase domain of MEK, and mutation Of several residues in this region has been shown to prevent interaction with YopJ (Hao et al., 2008). Interestingly, the same region has been implicated in binding anthrax LF (Chopra et al., 2003), and the G helix is also a part of the MEK homodimer interface (Ohren et al., 2004). Although the link between MEK dirnerization and LF/YopJ binding has not been established, it is tempting to speculate that dimer disruption may be a factor in MEK inhibition. Based on the specific inhibitory effects Of YopJ on the MAPK and NF-ICB signaling pathways, YopJ also has potential as a therapeutic for cancers and inflammatory diseases. However, unlike LeTx, which can be internalized by host cells, the delivery of YopJ into host cells is dependent on a type III secretion system that requires a direct interaction between the pathogen and host cells (Ghosh, 2004). Although YopJ can be expressed following transfection into mammalian cells, efficient delivery of YopJ in a clinical setting must be realized in order to develop YopJ as a therapeutic agent. 1.3.4. MEK inhibitors fail to generate clinical responses Two categories of MEK inhibitors that inhibit the pathway activity were reviewed in this chapter: small-molecule inhibitors that bind and inactivate MEK by a non—ATP- competitive mechanism, and biological inhibitors that are produced by bacteria and 24 inactivate MEK by posttranslational modifications. The MEK inhibitors in each of these categories have advantages and disadvantages as therapeutic agents. Small-molecule inhibitors are generally orally active and non-immunogenic, whereas the biological inhibitors need to be administrated by injection and may cause an undesirable immune response. On the other hand, small-molecule inhibitors are “washable” and they do not physically destroy MEK, whereas the posttranslational modifications of MEK by biological inhibitors are enzyme-mediated and irreversible in cells. Despite their pre- clinical successes, the MEK small-molecule inhibitors have not performed well in cancer clinical trials because of issues such as a lack of clinical response, insufficient efficacy, and safety concerns. Several facts may explain why these MEK inhibitors work well on xenograft tumors in nude mice but fail to generate clinical responses in human patients. First, individuals enrolled in cancer clinical trials are generally those patients who do not respond to conventional treatments and whose diseases have progressed to late stages with metastasis. The tumor responses in these patients to MEK inhibitors may not be well represented by subcutaneous xenografis in nude mice. Second, it has been shown that cancer cells having constitutively active mutations in MEK activators are relatively sensitive to MEK inhibition, whereas cells without these mutations are resistant (Y eh et al., 2004; Solit et al., 2006; Yeh et al., 2007; Haass et al., 2008). However, patients enrolled in most of the cancer clinical trials for MEK inhibitors were not initially selected for the preferred mutations. Therefore, patients’ responses may not correlate well with preclinical predictions. In the Phase I study of ARRY-142886, Ras and Raf mutations were determined in biopsies, and 38% (10 of 26 assessable biopsies) had Ras (n = 9) or Raf (n = 1) mutations (Adjei et al. , 2008). Although the three tumors showing the 25 strongest reduction in Ki-67 staining were positive for Ras or Raf mutations, no statistically significant difference in response was observed, possibly due to the small sample number in this study. Third, along with cancer progression, tumor cells in late- stage patients acquire more and more genetic alterations, which may activate MEK- independent survival pathways, leading to a resistance to MEK-dependent growth inhibition. This could explain why in the clinical studies of PD 184352 and its derivatives, MEK inhibitors did show desirable ERK inhibition but failed to generate clinical responses. 1.4. Introduction to cutaneous melanoma Cutaneous melanoma is a malignancy of melanocytes, which develops in the skin. Rarely melanomas can also develop in other parts of the body, such as brain, uvea, and the interior of the body. Cutaneous melanomas count only 4% of all the skin malignancies. However, 80% Of deaths from skin cancers are melanoma. Two facts result in the challenging management of melanoma. First, melanomas usually progress rapidly without Obvious changes on skin surface. Second, metastatic melanomas are very resistant to radiation and currently available systemic therapies. Therefore, once melanomas progress to metastatic stages, most patients are refractory to treatments. In this section, an overview of melanoma will be presented with a focus on molecular mechanisms of melanoma formation and currently available animal models for melanoma research. 26 1.4.1. Melanoma statistics The American Cancer Society (2010) estimates that 68,130 new cutaneous melanoma cases will be diagnosed in the United States this year. This ranks melanoma th th . . the 5 and the 7 most common cancer type in men and women respectively. The incidence rate Of melanoma has a race-dependent trend. It is more than 20 times higher in whites than in Afiican Americans (Altekruse et al., 2010). Among whites, the rate is about 50% higher in men than in women. In the past 35 years, melanoma incidence rates have been increasing in the population of white adults of 65 years and older (5.1% per year in men since 1985 and 4.1% per year in women since 1975). Interestingly, a rapid increasing of the incidence rate has also been observed among young white women aged 15 to 39 years (3.0% per year since 1992). The American Cancer Society (2010) also estimates that 8,700 deaths from melanoma will occur in the United States this year. If detected in the earliest stage, melanoma is also highly curable. The 5-year survival rate for localized melanoma (confined to primary site) is 98%. Unlike other types of skin cancers, however, melanoma is more likely to spread to other parts of the body, possibly due to its highly motile nature inherited from neural-crest progenitors. If melanoma has spread to regional lymph nodes, the 5-year survival rates range from 78% to 39%, depending on the numbers of metastatic lymph nodes (Balch et al., 2009). If systemic metastasis has occurred, the 5-year survival rate drops to 15% (Altekruse et al., 2010) or even lower than 10% depending on the substages (Balch et al., 2009). Therefore, early detection of melanoma is necessary for better prognosis. 27 1.4.2. Risk factors The two major risk factors of melanoma are ultraviolet (UV) exposure (environmental risk factor) and genetic abnormality (genetic risk factor). Higher sensitivity to sunburn and/or a history of excessive sun exposure (including both sunbums and use of tanning booths) increase the risk. Like many other types of cancers, family history of melanoma is a strong risk factor. Many melanoma patients have affected family members (Lynch et al., 1983). This led to cytogenetic studies and mutation screenings in familial melanoma tumor samples. Genetic abnormalities of several oncogenes and tumor suppressor genes are believed to be risk factors of melanoma. These will be discussed later in the following sections. 1.4.3. Melanoma progression, the Clark model. In 1984, Clark et al. reported a study of melanoma tumor progression. This flow of melanoma progression, known as the Clark model, describes the six steps of melanoma progression from normal melanocytes to metastatic melanoma. Each of these steps is accompanied by histological changes. Table III summarizes these six steps. At the first step, melanocytes may focally proliferate and the lesions are considered benign. The proliferating melanocytes may follow a programmed differentiation pathway which leads to disappearance Of a nevus. If the differentiation pathway is not followed the melanocytes become hyperplastic (the second step). At step 3, random dysplastic melanocytes with atypical nuclei form. According to Clark’s observations, the vast majority of dysplastic precursors are terminal lesions that do not progress to the next step and form melanomas. If the dysplastic precursors do progress, they follow a two-phase 28 growth pattern before they metastasize. The first phase is the radial growth phase (step 4 of the Clark model), which is characterized by an enlargement of the tumor lesion at its periphery. Tumors at this step grow in the epidermis, and are not metastatic. The second phase is the vertical growth phase (step 5 of the Clark model). In this step tumors acquire the competence for metastasis. Tmnors at this step grow vertically into the epidermis in an expansive manner forming metastases, the last step of the Clark model. 1.4.4. Melanoma staging The American Joint Committee on Cancer (AJ CC) staging system is most Often used to describe the stages of melanoma. This system, also referred to as the TNM staging system, is based on the size of the tumor (T), the extent of spread to the regional lymph nodes (N), and the presence of distal metastasis (M). Combining these three categories (T, N, and M), a grouped staging system is described using 0 and Roman numerals from I to IV for melanoma (Balch et al., 2009; Gershenwald et al., 2010). In melanoma staging system, subgroups are defined in each of Stages I to 111 depending of the TNM status. Stage 0. At this stage, the melanoma is in situ, and stays in the epidermis but has not spread to the dermis. This stage is equivalent to the first step of the Clark model. Stage I and Stage II. Melanomas at these two stages are still localized in the skin and have not spread to regional lymph nodes. Therefore, melanomas at these two stages may be between the second step and the filth step of the Clark model. The major criterion distinguishing the two stages is the tumor size. The melanoma with its thickness smaller than 1 mm is defined as Stage I, while the melanoma with its thickness between 1 29 and 2 mm is defined as Stage II. Depending on the tumor Size and ulceration, each of these two stages is subclassified to substages (IA, IB, IIA, IIB, and 11C). Stage III. At this stage, the melanoma has spread to regional lymph nodes, but distal metastasis has not occurred. Depending on the tumor size, ulceration, and the numbers of regional lymph nodes with evidence Of melanoma spread, this stage is subclassified to substages of IIIA, IIIB, and 111C. Stage IV. If the melanoma has spread beyond the regional lymph nodes and has metastasized to distal lymph nodes and/or other organs (mostly the lung, liver, or brain), it is defined as a Stage IV melanoma. 1.4.5. Treatment of melanoma Surgical removal of the primary growth and surrounding normal tissues is essential for melanomas at early stages. Sentinel lymph nodes are sometimes biopsied for stage determination. 1f lymph node metastases are present, more extensive lymph node surgery may be needed. For melanomas with deep invasions or metastasis to regional lymph nodes, immunotherapy, chemotherapy, and/or radiation therapy may be necessary. Unfortunately, none of these therapeutics prolongs survival in patients with metastatic melanoma. For metastatic melanoma, currently dacarbazine (DTIC) is the only US Food and Drug Administration (F DA)-approved chemotherapy agent, and high- dose interleukin—2 (IL-2) is the only F DA-approved irnmunotherapy (Tarhini & Agarwala, 2006). However, DTIC is effective in less than 10% of patients, and IL-2 has a high toxicity and gives a low response rate. As discussed earlier in this Chapter (section 1.3.1), several MEK inhibitors have failed in melanoma clinical trials. A number of clinical 30 trials are ongoing to evaluate the clinical efficacy of some other novel agents for melanomas. 1.4.6. Common genetic abnormalities of melanoma- The most common genetic abnormalities found in melanomas are those of BRAF, NRAS, CDKN2A, and PTEN (Miller & Mihm, 2006; Gray-Schopfer et al., 2007). The involvement of the products of these genes in melanoma formation and progression will be the focus in the following sections. 1.4.6.1. BRAF and NRAS As discussed in the earlier sections in this chapter, oncogenic mutations of BRAF and NRAS lead to a constitutive activation of the MEK-ERK survival pathway, which may cause cancer. In fact, before the members in the Raf-MEK-ERK pathway were identified, mutations in NRAS had been linked to cancers. van 't Veer et al. (1989) screened a panel of melanoma tumor samples and cell lines for NRAS mutation and found that NRAS mutations were detected only in melanomas from sun-exposed skin. Since NRAS mutations were also found in four of the ten primary melanomas examined, van ’t Veer hypothesized that UV exposure causes NRAS mutation in melanocytes which occurs at a premetastatic stage. This hypothesis was supported by Ball et al. (1994) who examined 100 melanoma tumor samples and found NRAS mutations in 36 samples (36%). In addition, they found that NRAS mutations occurred in 19% of the melanoma tumors that were in the second step of the Clark’s model, and at a higher frequency (56%) in primary tumors from continuously sun-exposed Skin. In 2000, Tsao et a1. (2000) 31 evaluated 53 cutaneous melanoma cell lines for NRAS mutation, and found activating mutations in 21% (11 of 53) of the cell lines. Downstream, BRAF mutations were found in 66% Of melanomas examined and at lower frequency in other types of human cancers (Davies et al., 2002). Interestingly, Davies found that all of the identified BRAF mutations were within the kinase domain and the V600E oncogenic mutation accounted for 80%. Collectively these studies Show oncogenic mutations in NRAS and BRAF play a critical role in melanoma development. Based on Davies’ finding, Pollock et al. (2003) examined BRAF status in nevi, primary melanoma tumors, and melanoma metastases to evaluate the timing of oncogenic BRAF mutation. Unexpectedly they detected BRAF mutations resulting in V6OOE substitution in 82% (63 of 77) of nevi. Since most nevi do not progress to malignant melanoma, this finding suggests that oncogenic BRAF alone is insufficient for melanoma turnorigenesis. To support this, Michaloglou et al. (2005) expressed B-Raf_V600E in normal human melanocytes and found that sustained expression of B-Raf_V600E resulted in cell cycle arrest, which was accompanied with inductions of tumor suppressor p16 and other senescence markers. This led them conclude that oncogenic B-Raf induced cell senescence in normal melanocytes. Moreover, Wajapeyee et al. (2008) performed an shRNA screening and identified the insulin grth factor binding protein 7 (IGFBP7) as a candidate required for B-Raf_V6OOE-induced melanocyte senescence, and proposed the therapeutic potential of IGFBP7 for melanoma treatment. Collectively, data from these studies indicate (1) the RaS-Raf-MEK-ERK signaling pathway is essential for melanoma formation, (2) elevated activity of this 32 pathway due to mutations is not sufficient for melanoma tumorigenesis, and (3) other molecular events are required for melanoma development. 1.4.6.2. CDKNZA 0916’MM and 1219‘”) In 1986, Pedersen et al. reported abnormalities of chromosome 9 in malignant melanoma tumors (10/10 melanomas derived from 8 patients). This was the first report of chromosome 9 abnormality in non-cell line melanoma samples. A similar study was also presented by Limon et al. (1988) showing that chromosome 9 was one of the chromosomes that were most fi'equently found to be structurally aberrant in melanoma cells from patients with metastasis. Later studies of dysplastic nevi and malignant melanomas showed this structural aberrancy of chromosome 9 was a deletion of chromosome 9p21 in (Cannon-Albright et al., 1992; Fountain et al., 1992; Petty et al., 1993). A melanoma susceptibility gene was therefore believed to reside in this region. Using yeast two-hybrid screening, Serran et al. (1993) cloned p16 from a HeLa cell cDNA library, and characterized p16 as an inhibitor of cyclin-dependent kinase 4 (CDK4). Therefore p16 was also named p1 61NK4 or cyclin-dependent kinase inhibitor 2A (CDKNZA). Within half a year, p16 was mapped to a locus at chromosome 9p21, and the high frequency of homozygous deletions or mutations ofp16 was confirmed in melanoma as well as other types of human cancer cell lines (Karnb et al., 1994; Nobori et al., 1994). Germline mutations Ofp16 were also detected at a high frequency (92%) in familial melanoma cases and at a lower frequency (30%) in dysplastic nevus cases (Hussussian et al., 1994). The p16 gene was therefore proposed as a candidate of tumor suppressor gene for melanoma. 33 Interestingly, when mapping the CDKNZA locus to chromosome 9p21, Stone and Kamb (Stone et al., 1995) identified a novel form of CDKNZA transcript which contained a different first exon resulting in an entirely different Open reading frame fi'om the original p16 transcript that was initially reported in 1994. This alternative first exon of p16 was later cloned and mapped to chromosome 9p at about 20-kb upstream to the original p16 exon 1 (Mao et al., 1995). In the same year, Quelle et al. (1995) cloned the full-length coding sequence of this novel gene and named it p19 (ARF stands for alternative reading frame) as it encoded a protein with a predicted molecular mass of 19.3 kDa. What was surprising was not only the fact that this dual-utilized DNA sequence gave rise to two different open reading frames for distinct gene products, but that . . 6INK4 . . expressron ofp19 1n p1 -deleted cells could Induce cell cycle arrest, lrke p16 (Quelle et al., 1995). The mechanism by which p19 induced cell cycle arrest was not clear at that time. Since pl 61 NK4 and p19 were cloned and mapped, groups of studies have reported mutations in the CDKNZA locus which harbors both p1 OJNK4 and p19 , in familial melanomas in Australia, Europe and the United States (Walker et al., 1995; Borg et al., 1996; FitzGerald et al., 1996; Flores et al., 1997; Harland et al., 1997; Platz et al., 1997; Soufir et al., 1998; Aitken et al., 1999). These findings support the link between loss of p16/p19 tumor suppressor function and melanoma formation. After years of studies Of these tumor suppressors, the mechanisms by which p16 and p19 suppress tumor formation are better understood. Both p16 and p19 inhibit G1/S cell cycle transition through different but related pathways [reviewed by (Lowe & Sherr, 2003; 34 Polager & Ginsberg, 2009)]. The tumor suppressor pl 6 binds to and inhibits cyclin- dependent kinase 4 (CDK4), the activity of which is required to release the transcription factor E2F fi'om Rb, and in turn, promote cell cycle progression through the G1/ S check point. On the other hand, p19 stabilizes p53 (which is also a tumor suppressor inducing Gl/ S cell cycle transition and/or apoptosis) by binding and inhibiting Mdm2, a p53 negative regulator. Before p16 and p19 were cloned and mapped, Cowan et al. (1988) reported an interesting correlation. Loss of one copy of chromosome 9 was detected at a high frequency in melanomas but at a much lower frequency in nevi. Similarly, germline mutations Of p16 were detected at a high frequency (92%) in familial melanoma cases, but at a lower frequency (30%) in dysplastic nevus cases (Hussussian et al., 1994). These finding suggested that loss Of pl 6/p19 tumor suppressor function is not likely to be responsible for nevi dysplasia. Instead, it may be a late-stage event during melanoma development. 1.4.6.3. PTEN Since Parmiter et al. (1988) presented the first report Of chromosome 10q abnormality (including break, translocation, and chromosome loss) in melanoma cell lines and tumor samples, the involvement of a potential tumor suppressor gene residing on chromosome 10 in melanoma formation had been hypothesized. Following Parmiter’s initial report, LOH of chromosome 10q22-10qter was found in non-familial melanoma tumor samples (Herbst et al., 1994). In 1997, the putative tumor suppressor gene sequence on human chromosome 10q23 was identified by two independent groups almost 35 at the same time (Li et al., 1997; Steck et al., 1997). The gene product was predicted to contain a protein tyrosine phosphatase domain and a large region with homology to chicken tensin and bovine auxilin. The gene was therefore named PT EN (phosphatase and @sin homolog). After PTEN was mapped and cloned, LOH of the PT EN allele was found in 40- 60% of primary or metastatic melanoma tumor samples (Teng et al. , 1997; Birck et al. , 2000), and PT EN mutations were found in about 30-50% of examined melanoma cell lines (Guldberg et al., 1997; Teng et al., 1997; Tsao et al., 1998) as well as 10% of primary melanomas (Tsao et al., 1998). The tumor suppression role of PTEN in melanoma formation was then tested and supported by several studies. First, ectopic expression of PTEN suppressed melanoma tumor cell growth (Robertson et al., 1998). Second, over-expression of PTEN inhibited melanoma cell colony formation (Tsao et al. , 2000). Third, Hwang et al. (2001) Showed that PTEN rescue reduced melanoma tumorigenecity and metastasis. Finally, a similar study demonstrated that PTEN re- expression in melanoma cells which lacked PTEN protein expression retarded tumor development in mice (Stahl et al., 2003). The tumor suppression function of PTEN was also supported by an epigenetic study. Using immunohistochemistry staining, Zhou et al. (2000) found no to low PTEN protein expression in 15% and 50% melanoma samples, respectively. Surprisingly, they found that among the examined melanoma samples which had no PTEN protein expression, 80% (4/5) of them showed neither mutation of the PTEN gene nor deletion of the PTEN allele. Their findings not only demonstrated an epigenetic mechanism by which loss of PTEN function is involved in melanoma formation, but also suggested that 36 the involvement of loss-Of-function of PTEN in melanoma might be underestimated. This provides an explanation for the fact that in some studies the PTEN mutation rate in melanomas was very low and no deletion or LOH of PTEN was detected (Wu et al., 2003) 1.4.7. Disease models Oncogenic mutations and loss of tumor suppressors have been identified as critical drivers of melanoma formation and the disease progression. Based on these findings, great efforts have been made to develop animal models for melanoma research. These melanoma animal models are developed mainly for two purposes. First, these models allow testing the requirements of those genetic alterations and involvements in melanoma tumorigenesis and the disease progression. Second, these melanoma animal models facilitate the development of melanoma therapeutics as the efficacy of novel therapeutic agents and treatment strategies can be tested in these models. This section will discuss these melanoma animal models. In 1997, Chin et al. (1997) reported a cooperation of oncogenic Ras and p] 61 NK4a-deficiency for melanoma susceptibility in viva. In this mouse model, the . G12V . . oncogemc Ras (H—ras ) was expressed specrfically 1n melanocytes (under the control of tyrosinase promoter) on a p] 6INK4a-deficient background. These mice (with double deficiencies) developed spontaneous melanoma after a short latency, indicating the oncogenic Ras and loss ofp16mK4a allele cooperatively promote melanoma formation. In addition, two interesting observations were obtained from this work. First, the 37 fi'equency of H—rasGIZ V-transgenic founder production was unusually low (only three founders from a total of 420 pronuclear injections). This raised the possibility of a . . . GI 2 V . selective pressure against oncogemc Ras. Second, before the H-ras -transgen1c . NK4 . . founders were crossed wrth the p161 a-deficrent mice, no melanoma or other malignancy occurred in the three founders (hyper-pigmentation and a proliferating melanocytic mass were the most relevant phenotypes observed). This indicates that activation of Ras is not sufficient for melanoma tumorigenesis, and that a second genetic alternation is required for melanoma tumorigenesis. This was supported by Powell et al. (1999) using another similar melanoma mouse model in which a mutant H-Ras was expressed specifically in melanocytes. Powell et al. also found that UV radiation or chemical carcinogens such as DMBA and TPA were necessary for melanoma formation in these mice. Since constitutive activation of Ras may result in low fiequency of transgenic founder production, in 1999 Chin et al. (1999) developed an inducible melanoma mouse model, in which the melanocyte-specific H-rasGlzV expression was induced by doxycycline on a p] 6m lad-null background. Not surprisingly, melanoma developed in doxycycline-treated mice within 60 days. More interestingly, withdrawal of doxycycline resulted in a clinical and histological regression of developed tumors, indicating that a persistent Ras activation is required to maintain melanoma tumor growth. In 1998, a melanoma mouse model was established by Kelsall et al. to test the effect of UV exposure on melanoma formation. In this model, the viral oncoprotein small DNA tumor virus (SV40) early region was engineered to be expressed specifically 38 in melanocytes under the control Of mouse tyrosinase promoter. After UV exposure, primary melanoma and melanoma metastases occurred without the use of other chemical carcinogens. This mouse model not only tested the requirement of oncogenic proteins for melanoma tumorigenesis, but also demonstrated the critical involvement of UV exposure in melanoma initiation. Since SV40 oncoprotein inactivates both Rb and p53 (Levine, 2009) pathways, this work also demonstrated the involvement Of loss of the tumor suppressors in UV-induced melanoma formation. More relevant to melanoma-associated genetic abnormalities, Kannan et al. (2003) evaluated the effect of UV exposure on active Ras and pl 6- or pl 9-deficient mice. The results fiom this study showed UV treatment accelerated melanoma formation in active Ras and p19-deficient mice, indicating the cooperation between UV exposure and genetic alteration in melanoma formation. In contrast, this UV-induced acceleration of melanoma formation was not Observed in the active Ras and p16-deficient mice. This finding indicates that the p53 pathway plays a more critical role in suppressing UV-induced melanoma formation in viva than the Rb pathway. To evaluate the functional cooperation between PTEN and p16/p19, a dual inactivation of which was found in several types of human cancers, You et al. (2002) established a PTEN/INK4a double deficient mouse model. They found that deficiency of either PTEN or INK4a was not sufficient to induce melanoma in mice, and double deficiency of both tumor suppressor genes caused only low frequency of melanoma (7- 10%). Although the contribution Of an oncogene to melanoma tumorigenesis in viva was not included in this study, they found that an introduction of oncogenic Ras (H-rasGlzv) increased the anchorage-independent colony formation of the MEFs isolated fi'om the 39 PTEN/INK4a double deficient mice. This also supports that melanoma formation requires both oncogene activation and loss of tumor suppressers. In the past decade, Cre recombinase-mediated conditional gene recombination has been developed as a powerful tool allowing the introduction of desired genetic alternations in a conditional manner. Combining the tissue-specific transgenic approach, Bosenberg et. al (2006) recently established a mouse strain in which the expression of Cre recombinase was inducible by 4-hydroxytamoxifen (OHT) specifically in . T2 . . . . melanocytes(desrgnated TyrssCreER ). Usrng thrs transgemc mouse strain, researchers can conveniently manipulate the genetic context specifically in melanocytes to evaluate the contribution of a molecular event in melanoma tumorigenesis. Most recently, Dankort et al. (2009b) demonstrated a great example of this approach by crossing the TyrirCreERTz mice with another strain of mice in which the mutant B-RafVGOOE . . . . . V600E expressron was inducible to create an mducrble B-Raf melanoma mouse model. Using this Dankort et al. found that, again, constitutive activation of B-Raf was sufficient to develop benign melanocytic hyperplasia, but was not sufficient for melanoma formation. Similar to the finding by Chin et al. (1997) discussed earlier in this section, Dankort et al. concluded that oncogenic B-Raf must cooperate with a loss of tumor suppressor, PTEN in this example, to induce melanoma formation and lung metastasis. In addition, using this melanoma mouse model they demonstrated the efficacy Of kinase pathway inhibitors for melanoma prevention and treatment. This work not only demonstrated the efficacy of kinase pathway inhibitors in a non-xenograft melanoma 40 animal model, but also provided an example of how a melanoma mouse model may be used to test therapeutic agents. Collectively, these studies using melanoma mouse models indicate that both activation of oncogenes and loss of tumor suppressors are required for melanoma formation. More importantly, these studies provide a molecular mechanism of melanoma progression. Acquiring oncogene activation seems to be the earliest event. This is believed to be the cause of hyperplastic lesions as most benign nevi have Ras or B-Raf oncogenic mutations (Pollock et al., 2003), and loss of tumor suppressors is not frequently found in dysplastic nevi (Cowan et al., 1988; Hussussian et al., 1994). According to the Clark model, most benign nevi at step 1 follow a programmed differentiation pathway and disappear. Supporting this, previous studies have suggested that oncogene acquisition is not sufficient for tumor formation, but instead, induces cell senescence (Michaloglou et al., 2005; Braig & Schmitt, 2006). This also explains why most benign nevi have Ras/Raf mutations but do not progress to malignant melanoma. As shown by several melanoma mouse models discussed above, oncogene activations need to cooperate with loss of tumor suppressors to initiate melanoma. This molecular mechanism of melanoma progression is in consistent with the Knudson two-hit theory (Knudson, 1971 ). 1.5. Discussion The Ras-Raf-MEK—ERK signaling pathway plays a critical role in regulating cell cycle progression, cell proliferation, and survival. Deregulation of the pathway activity may cause diseases, including cancers. Somatic mutations of Ras and B-Raf are found in 41 several types of human cancers at a high frequency particularly in melanomas. Using cell-based in vitro studies as well as in viva mouse models, numerous research groups have demonstrated the critical role of this oncogenic signaling as well as other tumor suppressing pathways in melanoma tumorigenesis and survival. Considerable effort has been made to develop therapeutic agents for treating cancers. Among these potential therapeutic agents, small-molecule inhibitors targeting MEKl and MEK2 have shown promising anti-cancer effects in laboratories and pre- clinical studies. However, none Of them has demonstrated clinical efficacy in cancer patients and proved as anti-cancer drugs. This indicates that our understating of the roles of MEK signaling pathways in cancers is incomplete. MEKl and MEK2 are generally thought to be functionally redundant for the following reasons. First, MEKl and MEK2 share greater than 85% homology in amino acid sequences, and almost identical crystal structures (Ohren et al., 2004). Second, both MEK] and MEK2 phosphorylate ERK1 and ERK2, and ERK] and ERK2 are the only known substrates of MEK] and MEK2. Regardless the high structural and biochemical similarities of MEKl and MEK2, however, a handful of studies have suggested that these two protein isoforms may have non-overlapping biological functions (Giroux et al. , 1999; Belanger et al., 2003). A better understanding of the difference between MEK] and MEK2 as well as their distinct biological functions may help developing more efficient anti-cancer drugs. For this purpose, using two complementary approaches I evaluated the necessity and sufficiency of MEKl and MEK2 signaling pathways for melanoma cell 42 proliferation. This research provides insights into the complexity of the MEK signaling pathway. The results are presented in the following chapters. 1.6. Tables and figures 43 .933 33 to... 3 3530.5 5.58» u§u§v50w80303 93 6803 5383005 0528.33 .3 3530.5 £3588 <75 A9923 .8380;on :00 .3 3530.5 28380305 :00 v2.22: 030.6 33:00 a .vEm—énmmosm .233 363:3 wfims 83 80203 was Mam 9 53.3%-: 89a 00253033032” 00—0205 .3360 ME). 8m 95805302 .952 no ~Mm2 ©0502. mam: .3 “00:20.20.“ 0.03 930.3 0350— 9...... :N e 38 r... a 3.3 .88 s... a not 0.... .83 r... a not H.3. 2: mg... I 8.: 2.. v3... 333%? Goon r3 .0 flow 68m 5.33.3.5 a. 9236 ”m3. Goon .Bw0mm30m a. 0.335 «0% 3028 Om 2.. 32...... n :5... 2.. :5... .. .. 38 c... a 8:53 30.0” .20 on - 0 .0 . r . .. . Goom N I m 32 N 2 cog—hr?” WWW...” .88 Na .0 3.39 .33 Na .0 2303-22.05 $0.”. mmmvw. Om 2.. m... .. 5.... : A.oo~..e e 0 «>3 H 0 :33 N a 8 “W 1.22”." .0 Ed... mooom :3 .0 355 ”M33 :3 .0 333.5 «0% 8.53 2.. n. u an... . zoom 4.. a 5.... 608 ..... s 853 33. s... . e .0 20 n 0 . .. . .. . . Ago— N fine—ham .0 303m .30— 3 .0 >085 .33 3 .0 307.5 mom omowoo Om 2.. cm 1 n AN3.303 333.30 .3 3530.5 3:03 33:00 03.233 ch...» 2.. .3 60530.5 .3380 M92 952550 .2... 80. 0.05:3 ch— ciao—eaéafim A 0335 2500. .wam a. 80:00:09 :0me :3 8.30035 .3 .000: 80.020 05 :0 B33 3:303 03 33.. E 03 mm- 35 0:03:00: 035350 0.3. a 003200: 050.35 .33 603:0 0.8 00:0 03.320 05 8 8:: 00:23: 0:28: :0 RU 0233 BE. 00:0 0330—0 "3 05 :0 :_ 60300900 .3.: 03 835 L 5360: 0330.0 0308:: 05 3 2.3 00.6.00: 0.33%? .335 0030—0 ”5 0:03.: :oummo: 05 8 “003:0 03 00:00 53070.»:2 :m :3030 000:0:30 200 05:3. 0 H4 \ x M N 012w... \ m. muamgmm 0:950:00 n... .H. 0 a 0. 0 m a \ .050 a z 0 m m .0. 4 CHISEEM: w w a z m A m .H \ 3.30 q 0. m m a m. 0 $0330.53 0 m m m m M m H \ .0H.M q o A z m M M 0MM2. e m z m z a B h \ .002: .4 e 0 M, m 3 m .mmmummm.0MMz m m m z 0 m z A \ GEM q 4 .M m M o o .00...-00M3MVE m m M 0 z 0 0 H \ .0m.m : a M M M w 0 anM2 H a m 2 H a q d \ .OH.m : > m M m m : mMmz : m m m z a o H \ .0.0 e m m m M A 2 HMmz .0 .0 .0 .0 .0 .m .N .H H m m 0 m 0 h 0 "0.0. 00000000: 000000350 n. ammunmsvmm UHUM ocHad 0000000350 :4 .0030 03.5.0.0 ha 05 was: 00055—000 30: 05:3 MESS—HE no 0:08:33. .= 030% 45 :00:«0 0:26.202 6626208 :0: 00:80:58 5:? :062 «30:00::«0 9.2:: 66220:: :0: 00:20:88 3056» :062 30:00:.«0 586: A2923 820:: 5:3 6:00 :0 00:«:«0::« Ea :0::«u:0:0.m€ ::«::0n«v 2&3 0E0_0$0 E205: w:« «6290:»: 06:00:22: 30:23::2 E0668: 5:3 30: 02308202 .A:0_:«t:0:0.m€ ::«::0n«v «6290:»: 05:00:20.: 6:022:0— 00:6:: :5 ::066:0: 5:3 30: 0230:2022 6:060: :wE0£ 603005—08 _«:::0: b23036 0:8 30: 0:50:22: 00:33:: 3:05:00 «80:20:: 0320202 .0 «60:20:: E86: :0 062:: 539% _«0_::0> .m «80:20:: S86: :0 08:: 538w 203: .v «62:93 2308202 .m «629093 0230:2032 .N 250: 0:50:23): 2 80::6060Q 6:06 200:: 2:20 .:06»0:w0:: «30:20:: «0 360:: 2:20 0:: a: 030:. 46 Figure 1. Schematic illustration of the Raf-MEK—ERK signaling pathway. Ras-mediated Raf activation involves a complex series of events, including protein recruitment to the cell membrane and phosphorylation at numerous residues [reviewed by Chong et al. (2003)]. Active Raf then activates MEK by phosphorylating the two conserved serine residues in the activation loop. Active MEK then in turn activates ERK by phosphorylating the threonine and tyrosine residues in the conserved T- E-Y motif. After its activation, ERK phosphorylates and activates downstream effectors that regulate a variety of cellular events, including cell cycle progression and cell proliferation at the transcriptional and posttranslational levels. The circled P symbols denote phosphate groups added on the critical serine residues and threonine/tyrosine residues of MEK and ERK, respectively. 47 Figure 1. Schematic illustration of the Raf-MEK-ERK signaling pathway. _ lRasI H 6'6 @ @3 Cell cycle progression Cell proliferation 48 Figure 2. Structures of MEK inhibitors. (a). PD 098015. (b). U0126. (c). PD 184352. ((1). PD 0325901. (3). PD 318088. (f). PD 334581. (g). ARRY-142886. (e-g). The A and B rings of the diaryl amines of PD 1843 52 and its derivatives are denoted. 49 Figure 2. Structures of MEK inhibitors. NH2 CN NH2 0 NHZCN NH2 PD 098059 U0126 O,NH o dl-IO’\/\O NH 0 NH ‘\ NH ‘\ PD 184352 PDF 0325901 e f. QCNXNHN O,NH 0 El he PD 318088 PDF 334531 3””fi Br ARRY-142886 50 Figure 3. Effect of PD 184352 on MEK activation in cells. Human melanoma SK—MEL-28 cells were cultured as described in the Materials and methods section of Chapter 11 (page 7 7). Cells were treated with PD 184352 at the concentrations indicated for 24h. Total cell lysates [prepared as described in the Materials and methods section of Chapter 11 (page 79)] were subjected to immunoblotting probed with antibodies against phospho-MEKl/Z, phospho-ERKl/Z, total ERKl/Z, and GAPDH. [PD184352](nM): o 10 1001000 E - Q q MEK1I2-P04 ...—._.' ___ ERK1I2-PO4 1m... up ‘ nun-l ERK1I2 GAPDH 51 Chapter II. Cleavage-resistant MEK proteins; a novel experimental model to establish MEK sufficiency 1.1. Introduction In order to determine the sufficiency of a MEK/MKK signaling pathway for a cellular function, the experimental system used will have to inhibit the pathways of multiple MEK/MKK members, but leave only one active. It is not expected to be efficiently achievable by transfecting several interference RNA molecules into cultured cells to target multiple protein molecules. In this chapter, the development of a novel experimental model achieving this challenging task will be presented. In this system, multiple endogenous MEK/MKK proteins and the downstream signaling pathways are inhibited by treating cells with LeTx, a bacterial toxin with MEK/MKK-specific proteolytic activity (see Chapter I section 1.3.3.1 for the introduction). Simultaneously, a mutant MEK/MKK protein that is resistant to LF-mediated cleavage is expressed in cells to preserve the specific MEK/MKK signaling pathway. This achieves a special cellular context in which only one MEK/MKK signaling pathway is present. If a MEK/MKK pathway is sufficient for a cellular function, the cleavage-resistant form of the corresponding MEK/MKK should rescue this cellular function when the cells are treated with LeTx. 52 1.2. Results 1.2.1. Point mutations at the Pl' site render MEK resistant to LF—mediated cleavage An alignment of all the MEK and MK amino acid sequences (Table II) revealed a consensus sequence for LF cleavage: (B/P)3.4X3/Al, where B represents basic residues, P represents proline, X represents variable residues, and Al represents aliphatic residues. The aliphatic residue at the Pl' position is fully conserved in all the MEK/MKK proteins, and has been shown to be critical for the cleavage by LF (Park et al., 2002; Chopra et al., 2003). Therefore, to make MEK resistant to LF-mediated cleavage, I introduced an aliphatic-to-aspartic acid mutation in this position of human MEK and MK proteins. These cleavage-resistant (or) mutant proteins are named MEKcr or MKKcr in this dissertation. For the subsequent study, a V5 tag was fused to the amino termini of wild- type and the cleavage resistant mutants of MEK/MKK proteins. This NHz-terminal V5 fusion was included to examine the cleavage of the proteins by LF at the NHZ—termini (explained below). To confirm the cleavage resistance of MEKcr, an in-cell LF cleavage assay was developed. In this assay, wild-type VS-MEK or VS-MEKcr proteins were transiently expressed in CHO K1 cells which were then treated with PA alone (1 uyml PA as a control) or LeTx (1 ug/ml PA and 0.1 ug/ml LF) for 12h. As transient transfection of plasmid DNA can efficiently over-express exogenous proteins in CKO Kl cells, MEK cleavage by LF may not be clearly observed if an excessive amount of VS-MEK is produced during LF cleavage. To overcome this, cells were co-treated with 10 ug/ml 53 cycloheximide to block de novo protein synthesis during PA or LeTx treatment. To ensure successful internalization of LF and in-cell proteolytic activity of LeTx, cleavage of endogenous MEKI was first examined by immunoblotting probed with antibodies that specifically recognize the amino terminus of MEKl (LeTx causes loss of epitope) or the carboxyl-terminus of MEKl (LeTx causes mobility shift). As shown in Figure 4A, LeTx treatment resulted in a loss of the NHz-terminal epitope (Figure 4A, second panel) and an increase in the electrophoretic mobility of endogenous MEKl (Figure 4A, third panel) in control cells as well as in cells transfected with VS-lacZ control plasmid. This result indicates LF-mediated MEK cleavage occurs in CHO Kl cells. Under the same conditions, LF-mediated proteolysis resulted in a loss of NHz-terminal V5 epitope only in CHO K1 cells expressing wild-type VS-MEKI or V5-MEK2 but not in cells expressing VS-MEKI or or V5-MEK2cr (Figure 4B and C, top panels). To exclude the possibility that the loss of V5 epitope of wild-type VS-MEKI and VS-MEKZ was a result of failure in expressing the exogenous proteins, and to further confirm the cleavage of wilt-type VS-MEK proteins, antibodies recognizing carboxyl termini of MEK] or MEK2 were used to detect the electrophoretic mobility shift of cleaved VS-MEKI and V5- MEKZ. As shown in the second panels of Figure 4B and C, bands with slightly higher electrophoretic mobility were detected by immunoblotting with antibodies recognizing carboxyl-terminus of MEK] or MEK2 in cells transfected with wild-type VS-MEKI or VS-MEKZ, but not in cells transfected with VS-MEchr or V5-MEK20r. These results demonstrate that VS-MEchr and V5-MEK20r were resistant to LF-mediated proteolysis. In this experiment, a substantial increase of non-tagged MEK expression along with VS-MEK transfection was noticed (Figure 4B middle panel, compare none- 54 transfected cells with V5-MEK1 transfected cells). This was caused by internal translation fiom the original translation initiation codon of the MEK sequence cloned in the V5-expression vector. This was evidenced by the following observation. The original translation initiation codons that were inserted together with MEK sequences into the V5 expression vector were removed from the plasmid by doing a site-directed mutagenesis. The modified expression vectors were then transfected into CHO K1 cells. After this manipulation, the increase of non-tagged MEK expression was no longer observed (Figure 5). These experiments demonstrated that the increased non-tag MEK level after VS-MEK plasmid transfection was due to exogenous expression and not an up-regulation of the endogenous MEK. This dual translation pattern was also observed in VS-MEK-transfected SK-MEK-28 stable cell lines (will be presented in Chapter 111). 1.2.2. The cleavage-resistant mutation does not impair MEK activity. Cleavage by LF removes an amino terminal docking domain that is required for interaction with MAPK (Tanoue et al., 2000; Chopra et al., 2003; Bardwell et al., 2004). To confirm that neither the addition of the V5 tag nor the introduction of the LF cleavage-resistant mutation interferes with MEK’s ability to interact with and phosphorylate ERK, the activity of VS-MEchr and V5-MEK20r was examined. To do this, clonal lines of human melanoma SK-MEL-28 cells stably expressing VS-MEKI or or V5-MEK2cr, or their wild-type counterparts were established. Similar to most melanomas, SK-MEL-28 cells harbor the B-Raf__V600E mutation (Davies et al., 2002), which constitutively phosphorylates and activates MEKl and MEK2. Therefore, this cell line allows a convenient examination of the status of cellular MEK] and MEK2. When 55 an antibody that only recognized activated MEKl and MEK2 was used in the immunoblotting, the activation status of both VS-MEchr and V5-MEK2cr were found to be comparable to that of their wild-type counterparts (Figure 6A). This indicates that (1) neither the NHz-terminal V5 fusion nor the CR mutation impairs MEK activation and (2) VS-MEchr and VS-MEKZCr are activated in SK-MEL-28 cells. To further examine the ability of VS-MEKI or and V5-MEK2cr to phosphorylate their only known substrate, ERK, immunoprecipitation-kinase (IP-kinase) assays were performed. To do this, VS-MEchr and V5-MEK20r were immunoprecipitated from SK-MEL-28 cells and used as the kinase source for ERK in the kinase assay. Since saturating the kinase in a kinase assay may result in a false positivity, the optimum amount of wild-type VS-MEK immunoprecipitates in this IP-kinase assay was determined first. A fixed amount of recombinant ERK2 (400 ng) was used as the substrate and mixed with a decreasing amount of the VS-immunoprecipitates in the presence of active B-Raf to determine the minimum amount of the V5- irnmunoprecipitaes required for the kinase reaction. As shown in Figure 6B, a minimum of 5 ul of VS-MEKI or V5-MEK2 immunoprecipitates was required to fully phosphorylate 400 ng of ERK2 in the kinase reaction. Based on this optimization experiment, a full set of the IP-kinase assays, in which the VS-MEK immunoprecipitates V were mixed with or without active B-Raf and recombinant ERK2, was performed based on the optimized condition to test the kinase activity of VS-MEchr and V5-MEK2cr. As shown in Figure 6C, after considering the amount of the V5 immunoprecipitates used for each assay (Figure 6C, lower panel), VS-MEchr and V5-MEK20r were as capable as their wild-type counterparts in phosphorylating ERK2. These results indicate that 56 neither the addition of the V5 tag nor the introduction of the cleavage-resistant mutation alters the ability of MEK to interact with and phosphorylate ERK. 1.2.3. Point mutations at the Pl’ site render other MKK members resistant to LF- mediated cleavage A similar strategy was used to generate cleavage-resistant forms of MKK3, MKK4, MKK6 and MKK7. As expected, the aliphatic-to-aspartic mutation at the P1 ’ position rendered MKK3 and MKK6 resistant to LP cleavage (Figure 7). As MKK4 and MKK7 were previously reported to harbor two LF cleavage sites (V itale et al., 2000), two cleavage-resistant mutants with the aliphatic-to-aspartic mutation introduced to one of the cleavage sites were generated for each of MKK4 and MKK7: V5-MKK4cr_46 and V5—MKK4cr_59 for MKK4, and V5-MKK7cr_45 and V5-MKK7cr_77 for MKK7. LF completely cleaved wild-type V5-MKK4 in CHO K1 cells (Figure 8A). Unexpectedly, the aliphatic-to-aspartic mutation introduced to Leu46 of MKK4 was sufficient to make . . . 59 MKK4 re51stant to the cleavage, but the same mutation introduced to Phe was not. On the other hand, V5-MKK7cr_45 and V5-MKK7cr_77 were both resistant to the cleavage in CHO Kl cells (Figure SB). However, under the same condition, a convincing cleavage of wild-type MKK7 was not detected (Figure 8B). 1.2.4. MKK4 cleavage in mammalian cells Further experiments were performed to determine why V5-MKK4cr_46 was not cleaved by LP in CHO Kl cells. As shown in Figure 8A, LeTx treatment of CHO-K1 cells expressing wild-type V5-MKK4 resulted in a complete loss of the V5 epitope, 57 demonstrating the cleavage of MKK4 by LF. Since two LF cleavage sites were identified on MKK4, a mutant MKK4 with the cleavage-resistant mutation introduced at only one of the cleavage sites would be expected to be still cleaved by LF at the other cleavage site which remained unchanged. As expected, V5-MKK4cr_59 was still sensitive to LF- mediated proteolysis (Figure 8A). Presumably LF cleaved this mutant MKK4 at the Lys45-Leu46 position. Unexpectedly, however, the aliphatic-to-aspartic mutation introduced to Leu46 of human MKK4 was sufficient to render MKK4 resistant to cleavage, even if the Phe59 residue was remained unchanged (Figure 8A). Two possibilities can explain this observation. First, LF cleaves MKK4 only at the Lys45- Leu46 position but not at the Argsg-Phe59 position (i. e., Argsg-Phe59 of MKK4 is not a real LF cleavage site). Alternatively, LF cleaves MKK4 at both positions in a processive manner (1'. e. , cleavage at ArgSB-Phe59 requires a prior cleavage at Lys45-Leu46). To test these two possibilities in an unambiguous way, wild-type human MKK4 fused with a V5 tag followed by a 6xHis tag at the carboxyl terminus was constructed (denoted as MKK4-V5-His6 in this dissertation). The carboxy-terminal fusion allowed more readily determining the molecular weight of the carboxyl terminus of MKK4 after LF cleavage. In addition, two MKK4-V5-His6 deletion mutants were constructed as molecular weight indicators: MKK4 with a deletion of amino acid residues 1-45 (denoted as MKK4_d45-V5-His6), and MKK4 with a deletion of amino acid residues 1-58 (denoted as MKK4_d58-V5-His6). The MKK4_d45-V5-His6 deletion mutant also allowed testing the second possibility that LF cleaves MKK4 at both sites in a processive 58 manner. The set of these proteins was then expressed in CHO Kl cells and in-cell cleavage assays were performed. As shown in Figure 9, LeTx treatment on CHO Kl cells expressing wild-type MKK4—V5-His6 resulted in a complete loss of the V5 epitope, indicating MKK4 cleavage by LF. However, under the same conditions I could not detect the LF-cleaved carboxyl terminal fragment of MKK4 using an anti-V5 antibody (Figure 9, lane 2). An explanation for this is that after LF cleavage, the carboxyl terminus of MKK4 is degaded, as previously observed for MEKl (Duesbery et al., 1998) and MEK2. To test this, MG- 132, a proteosome inhibitor, was included in the in-cell cleavage assay. As shown in Figure 9 (lane 4), MG-132 treatment rendered the LF-cleaved carboxyl terminus of MKK4 recognizable by V5 antibody in the immunoblotting, indicating that after LF cleavage MKK4 is degraded through a proteosome-dependent pathway. The two MKK4 deletion mutants, MKK4_d45-V5-His6 and MKK4_d58-V5-His6, were distinguishable by the differential electrophoretic mobility in an SDS-PAGE gel (Figure 9, land 5-8 and lane 9—12). After LF cleavage, the carboxyl terminus of MKK4- VS-His6 (Figure 9, lane 4) had the same electrophoretic mobility as that of MKK4_d45- V5-His6 (Figure 9, land 5-8) but not MKK4_d58-V5-His6 (Figure 9, lane 9-12). This result demonstrates that wild-type MKK4 is cleaved by LF only at the Lys45-Leu46 position but not the Arg58-Phe59 position in cells. In addition, co-treatment of LeTx and MG-132 did not result in an electrophoretic mobility shift of the MKK4_d45-VS-His6 deletion mutant. This indicates that MKK4_d45-V5-His6 was not cleaved by LF and that the Argsg-Phe59 position of MKK4 was not an LP cleavage site. Taken together, these 59 data show that MKK4 has only one LF cleavage site, the Lys45-Leu46 position, as opposed to two LF cleavage sites as previously observed and reported by Vitale et al. (2000) in an in vitro assay. 1.2.5. MKK7 cleavage in mammalian cells As shown in Figure 8B, LeTx treatment resulted in a subtle decrease of the NH2- terminal V5 epitope of wild-type MKK7 (the second lane of the top panel in Figure 8B). One possible explanation for this is that a limited amount of LF could not cleave the excess of V5-MKK7 that was expressed in the transfected cells. To test this, a decreasing amount of the plasmid DNA encoding for wild-type V5-MKK7 was transfected into CHO K1 cells and the cleavability of MKK7 was tested in cells in the presence of cyclohexirnide. Under this condition, the expression level of wild-type V5-MKK7 was decreased proportionate with the decreasing amount of the plasmid DNA transfected (Figure 10, upper panel). However, LF was still not able to cleave wild-type MKK7 even when the expression of V5-MKK7 was decreased to a barely-detectable level. The uncleavability of wild-type V5-MKK7 in this experiment was not due to experimental failure as a complete cleavage of endogenous MEKl by LF was readily detected in the same cell lysates (Figure 10, middle panel). This result indicates that MKK7 might not be an enzymatic substrate of LP in mammalian cells. To further test this, 293FT cells (a human embryonic kidney 293-derived cell line) were treated with LeTx for 24, 48, and 72 hours, and whole cell extracts were prepared for immunoblotting to examine MKK7 cleavage by LF. If LF cleaves MKK7 at Gln44-Leu45 and Gln76-Leu77 positions as 60 previously reported by Vitale et al. (2000), then a decrease of NHz-terminal signal and an electrophoretic mobility shift of MKK7 should be detected following cleavage. As shown in Figure 11A, LeTx treatment did not result in a loss or a decrease of the immunoblotting signal of endogenous MKK7 in 293FT cells, even if the cells were treated with LeTx for 72h. In addition, an antibody recognizing the last 20 amino acid of MKK7 failed to detect an electrophoretic mobility shift of MKK7. This result clearly shows that MKK7 is not a preferred LF substrate in mammalian cells, and argues the MKK7 cleavage by LF observed by Vitale et al. (2000) may be an in vitro artifact. 1.3. Discussion In this chapter, the design and the engineering of cleavage-resistant forms of MEK and MK (MEKcr and MKKcr) were presented. This set of MEKcr were designed to be used as a tool to study the sufficiency of MEKl and MEK2 signaling pathways for melanoma cell proliferation, which will be presented in Chapter III. Cleavage resistant forms of MEK/MKK proteins have been previously reported for MEKl (Duesbery et al., 1998), MKK3 (Park et al., 2002), and MKK6 (Park et al., 2002; Chopra et al., 2003). Although the mutations were introduced at slightly different positions on each of MEKI (P1), MKK3 (P1 and P1 '), and MKK6 (P1 and P1 ', or Pl' alone), all of these studies suggest that hydrophobicity and surface charge surrounding the LF cleavage position are critical for the cleavage. In this dissertation project, aliphatic-to-aspartic acid mutations were introduced only to the Pl' positions of MKK1-4, 6 and 7, and the produced MEKcr and MKKcr were all resistant to LF-mediated proteolysis. These results extend previous observation (Chopra et al. , 2003) showing that the aliphatic residue adjacent to the LF 6] cleavage site are critical for LF-mediated proteolysis. When VS-MEchr and V5-MEK20r were stably expressed in human melanoma SK-MEL-28 cells, which harbor B-Raf_V600E constitutive activation mutation, these cleavage-resistant MEK mutants were phosphorylated and activated in the cells (examined by immunoblotting with an antibody that specifically recognizes MEK] and MEK2 with phosphate groups at the activation loops). These MEKcr proteins were phosphorylated to a comparable level as their wild-type counterparts, indicating that introducing the aliphatic-to-aspartic acid mutation to MEKl and MEK2 does not alter their ability to be activated. Moreover, these VS-MEKcr proteins could be immunoprecipitated from SK-MBL-28 cells and used as kinase source in an in vitro kinase assay. These V5-MEKcr proteins were as capable as their wild-type counterparts in phosphorylating their substrate, ERK2, in the assay. Interestingly, immunoprecipitated VS-MEK and VS-MEKcr proteins were not able to phosphorylate ERK2 in the absence of active B-Raf, although they were activated in SK-MEL—28 cells. A few possibilities can explain this. First, during cell lysis and immunoprecipitation, these proteins may have been de-phosphorylated by phosphatases, the activity of which was not fully inhibited by phosphatase inhibitors added in the lysis buffer. Second, in SK-MEL—28 cells, the B-Raf_V600E may have only activated a small portion of VS-MEK, yet this was sufficient to be detected by the phospho-MEK1/2 antibody by immunoblotting. After immunoprecipitation, the vast majority of the immunoprecipitated VS-MEK may not have been activated by B-Raf_V600E and was not able to phosphorylate ERK2 in the kinase assay until after the addition of active B-Raf. Finally, it is also possible that active 62 VS-MEK may be sequestered in a protein complex that may mask the epitope for antibody recognition. As a result of this, only less active VS-MEK could be immunoprecipitated. Vitale et al. (2000) reported that in addition to MEK] and MEK2, most members in the MK family were enzymatic substrates of LF with an exception of MEKS. For the most part the results repeated here support this observation. However, my data do not wholly support their findings. Both MKK4 and MKK7 were previously found to harbor two LF cleavage sites. The data presented above in this chapter show that in mammalian cells LF cleaves MKK4 only at the Lys45-Leu46 position but not the Argsg-Phe59 position, and that LF does not cleave MKK7 in cells. A comparison of the LF cleavage site consensus sequence [(B/P)3_4X3/Al] and the amino acid sequences of the two reported MKK4 cleavage sites shows that the amino acid sequence of the first LF cleavage site of MKK4, QGKRKALK45L46, matches the consensus LF cleavage site . 58 59 . whereas the second srte, PPFKSTAR F , does not (Table II in Chapter I, see page 45). . 58 59 . . . . This supports that Arg -Phe of MKK4 is not a real LF cleavage srte in mammalian cells, and that the cleavage at the Argsg-Phe59 of MKK4 observed by Vitale et al. was possibly an artifact resulted from the in vitro experimental system. Since Vitale’s initial report ten years ago, there have been no further reports of LF-mediated MKK7 cleavage published in the literature. The in-cell cleavage assays of 63 endogenous MKK7 by LF presented in this chapter were carried out in two different mammalian cell lines, Chinese hamster ovary cells (CHO K1) and human embryonic kidney 293 -derived cells (293 FT). The cleavability of either a low expression level of exogenous MKK7 or of endogenous MKK7 by LF was examined in these two mammalian cell lines, and no MKK7 cleavage was observed. The data strongly suggest that MKK7 is not a preferred LF substrate in mammalian cells. Moreover, on careful analysis of Vitale’s initial report, it is not clear whether MKK7 is cleaved by LP in vitro [the MKK7 panel of Figure 4 in the report by Vitale et al. (2000)]. In their in vitro cleavage experiments, LF resulted in a noticeable decrease of GST::MKK3, 4, and 6 Coomassie Blue staining signals. However, LF did not result in any decrease of the GST::MKK7 signal. Both reported MKK7 cleavage sites were identified by isolating two very weak Coomassie Blue bands with higher electrophoretic mobility in the gel. These two truncated MKK7 may alternatively be explained by protein degradation, which is commonly observed in GST-fusion protein purification. Collectively, the results presented in this chapter argue against the cleavability of MKK7 by LF. Unlike MKK4, the amino acid sequences of both cleavage sites of MKK7 match the consensus sequence of LF cleavage site perfectly (Table II in Chapter I, see page 45). If the amino acid sequences match the LF cleavage site consensus sequence and MKK7 also contains the conserved the LF-interacting region (Chopra et al., 2003), then why is MKK7 not cleaved by LF in cells? Perhaps the sequence of the LF cleavage site gives an answer to this question. The consensus sequence of LF cleavage site is generalized based on an alignment of all the reported MEK/MKK cleavage sites, including the two of 64 MKK7. If MKK7 is not an LP substrate, the two reported cleavage sites of MKK7 should not be included in the alignment, and the consensus of LF cleavage site may be different. In other words, if the two reported LF cleavage sites of MKK7 are excluded from Table 11, then a better consensus sequence of LF cleavage site would be (B)3X2_4(B/P)1/Al, where B represents basic residues, X represents variable residues, P represents proline, and Al represents aliphatic residues. In this new consensus sequence, a proline residue or a basic residue at the Pl position is conserved in MEK] , MEK2, MK3, 4, and 6, but not in MKK7. Therefore, it is very possible that the amino acid residue at the Pl position of MEK/MKK is also critical for LP cleavage. Two previous studies support this possibility. First, Duesbery et al. (1998) have demonstrated a mutant MEK] with an alanine substitution for this critical proline residue was resistant to LP cleavage. Second, the critical role of the amino acid at P1 position of MEK/MKK for LF cleavage is supported by a crystal structure study of LF. Pannifer et al. (2001) have shown that the side chain of proline or arginine at the P1 position of MEK/MKK sits in the SI substrate-binding pocket of LP to generate a productive cleavage complex. This chapter describes the development of a novel experimental system that allows evaluating sufficiency of an individual MEK/MKK signaling pathway when multiple other parallel family pathways are inhibited by LeTx. This system was then utilized and coupled with isoform-specific MEK knockdown to determine both sufficiency and necessity of MEK] and MEK2 signaling pathways for melanoma cell proliferation. This will be presented in Chapter III. 65 1.4. Figures Figure 4. Resistance of VS-MEKcr to LF-mediated proteolysis. (A) Non-transfected (None), mock-transfected (Mock), and VS-lacZ-transfected CHO K1 cells, as well as cells transfected with (B) wild-type VS-MEKI or VS-MEchr, or (C) wild-type V5-MEK2 or V5-MEK2cr, were treated with PA alone control (PA) or LeTx (LT) in the presence of cycloheximide for 12 h as described in Materials and methods. Total cell lysates were then harvested and subjected to immunoblotting probed with the antibodies indicated on the right of each panel. V5 antibody (V 5) and an antibody against NHz-terminus of MEK] (MEKl (NH2)) were used to detect loss of NHz-terminal epitopes following LF-mediated proteolysis. Antibodies against the carboxyl terminus of MEK] (MEKI (COOH)) and MEK2 (MEK2 (COOH)) were used to detect cleaved MEK] and MEK2, respectively, with increasing electrophoretic mobility. Antibodies against a-tubulin and B-tubulin were used as loading controls. 66 Figure 4. Resistance of VS-MEKcr to LF-mediated proteolysis. A None Mock V5-IacZ PA LT PA LT PA LT [ - -] vs E -- --- J MEK1(NH2) F- : :- ...;. A; q MEK1 (coou) M a-tubulin None V5-MEK1 V5-MEK1cr V5-MEK2 V5-MEK2cr PA LT PA LT PA LT PA LT PA 1 f" -iV5 l- .2...ng .-- M B-tubulin MEK1 (coon) 67 Figure 5. Dual translation of VS-MEK expression vectors. CHO K1 cells were transfected with plasmids encoding for VS-MEKl or V5- MEchr, or the modified plasmids in which the original translation initiation codons of MEK sequences were removed (V 5-MEK1_dATG and V5-MEK1cr_dATG). Non- transfected cells (None) or cells transfected with VS-IacZ were used as controls. After transfection, cells were treated with PA alone (PA) or LeTx (LT) in the presence of cycloheximide for 12 h as described in Materials and methods. Total cell lysates were then harvested and subjected to immunoblotting probed with antibodies against NH2- terminus of MEK] (top panel) and GAPDH (bottom panel). VS-MEKI and non-tagged MEK] are indicated. Lane numbers are labeled at the bottom of the blots. VS-MEK‘I VS-IIEKt None V5-lacZ V5-MEK1 V5-MEK1cr dATG an: PA LT PA LT PA LT PA LT PA LT PA LT V5-MEK1 : MEK1 GAPDH --~“~~“~~- H” 123456789101112 68 Figure 6. Activity of VS-MEKcr. (A) Phosphorylation and activation of VS-MEKcr in cells. SK-MEL-28 cells stably expressing VS-MEK or VS-MEKcr were treated with PA alone (PA) or LeTx (LT) for 24 h. Whole cell lysates were prepared and immunoblotted with anti- phosphoMEKl/Z antibody to examined the activation status of cellular V5-MEchr and V5-MEK2cr (upper panel), or with anti-GAPDH antibody as a loading control (lower panel). (B) Optimization of V5-MEK IP-kinase assay. VS-lacZ, VS-MEKI, V5- MEchr, VS-MEKZ, and V5-MEK2cr were immunoprecipitated from SK-MEL—28 cells as described in Material and methods. A decreasing amount (5, 0.5, and 0.05 ul) of the VS-MEK immunoprecipitates was added into in vitro kinase assay reactions which were then carried out as described in Material and methods. (C) Kinase activity of VS-MEKcr. Anti-V5 immunoprecipitates were prepared from SK-MEL—28 cells as described in Material and methods, and then used for in vitro kinase assays (top panel) in the presence or absence of active B-Raf and/or recombinant ERK2. Equal amounts of V5 immunoprecipitates were subjected to V5 immunoblotting (bottom panel) as the V5- MEK input control. 69 Figure 6. Activity of VS-MEKcr. A ca‘ vs“ 0" 0 “94°“ «6"? «6’6 46"“, sf)" LT PA LT PA LT PA LT PA LT 1!!! Mafia- O ...... O in MEKW-Pqi E-cfiu-—--—.--—-“]GAPDH B ‘3' A“ V5-MEK1 V5-MEK1cr V5-MEK2 V5-MEK2cr 5 5 0.5 0.05 5 0.5 0.05 5 0.5 0.05 5 0.5 0.05 I . . . . 14- ERK2 Q9 31" V5-MEK1 V5-MEK1cr V5-Iacz V5-MEK2 V5-MEK2cr B-RafT-+.+.+.+ .+.+-+-+.+-+ ERK2+--++.-++ .-++..++..++ .. 'TTW‘Trf . . . ,4. l” n. 4ERK2 r..- 2 75 m 7 L I“, __-:_, -E 3 xxgg Emmmm w¥¥¥¥ mmmmm >>>>> up..- 70 Figure 7. Resistance of MKK3cr and MKK6cr to LF-mediated proteolysis. (A) Human melanoma SK-MEL-28 cells stably expressing V5-MKK3 or V5- MKKBcr were treated with LeTx in an LP concentration-dependent manner (indicated) for 24h. Total cell lysates were harvested and subjected to immunoblotting probed with antibodies against V5 epitope (top panel), carboxyl terminal of MKK3 (middle panel), and a-tubulin (bottom panel). (B) CHO Kl cells were transfected with V5-MKK6, or V5-MKK6cr. In-cell cleavage assays were performed as described in Materials and methods. Total cell lysates were then collected and irnmunoblotted with antibodies against V5 epitope (top panel), carboxyl terminus of M6 (middle panel) and a-tubulin (bottom panel). 71 Figure 7. Resistance of MKK3cr and MKK6cr to LF-mediated proteolysis. A V5-MKK3 V5-MKK3cr [LF](ngImI): o 1 10 100 o 1 10 100 E 1,, ~‘flfl]<106 cells were washed with ice-code PBS (Invitro gen) three times. Whole cell extracts were prepared on ice in 1 ml of lysis buffer (20 mM Tris-HCl pH 7.5, 150 mM NaCl, 1.5 mM MgC12, 2 mM EGTA, 1% Triton X-100, 2 mM DTT, 10 mM NaF, 1 mM Na3VO4, and 12 mM [3- glycerophosphate), and then homogenized by sonication in ice bath. Soluble fraction of cell extracts was isolated by centiifugation at 12,000g at 4°C for 10 min. Protein TM concentrations were determined by BCA Protein Assay Kit (Pierce) according to the manufacturer’s instructions. Two hundred mg of clear extracts were incubated with 25 ul of agarose-immobilized V5 antibody (Bethyl Laboratories) in a total volume of 500 ill of lysis buffer on a rotator at 4°C for 12b. The precipitates were then washed twice with the 81 lysis buffer and twice with kinase assay buffer (25 mM Tris-HCl pH 7.5, 5 mM [3- glycerophosphate, 2 mM DTT, 0.1 mM Na3VO4, and 10 mM MgClz), and resuspended in 50 ul of kinase assay buffer as the kinase source for the following kinase assay. 1.5.8. In vitro kinase assay. Five rnicrolitters of prepared V5 immunoprecipitates were incubated on ice with or without 0.05 ug of active B-Raf (Upstate Biotechnology) and 400 ng of recombinant ERK2 in a total volume of 2 ul of assay dilution buffer (20 mM MOPS pH 7.2, 25 mM [3- glycerophosphate, 5 mM EGTA, 1 mM Na3VO4 and 1 mM DTT), and then 3 ul of ATP mixture [0.5 ul of [y-3ZP]ATP (Amersham; 10 mCi/ml, 3000 mCi/mmole) in 250 uM ATP and 37.5 mM MgC12] was added. In vitro phosphorylation reaction was carried out by incubating the reactions in 30°C water bath for 30 min, and then stopped by addition of 10 ul of 2>50 uni diameter) within 21 days (Figure 20A, top panels). However, in the presence of LeTx, colony formation of all cell types tested was inhibited, except for V5-MEK2cr-expressing cells (Figure 20A, lower panels). When these results were quantified, it was noted that expression of V5-MKK2cr in SK—MEL—28 cells resulted in a significant rescue of 70% of the colony formation in the presence of LeTx (Figure 20B). This result, in agreement with the proliferation results presented earlier, demonstrates that the MEK2 signaling pathway alone is sufficient for anchorage-independent growth of SK-MEL-28 cells. 1.2.3. A xenograft model to test the sufficiency of MEK] and MEK2 signaling pathways for melanoma tumor growth in viva As discussed in the General Introduction (Chapter I), LeTx possesses a potent inhibitory activity on tumor growth in vivo in different cancer xenograft models (Duesbery et al., 2001; Koo et al., 2002; Depeille et al., 2007; Ding et al., 2008). A similar approach was used in this dissertation project to determine the sufficiency of MEK signaling pathways for melanoma tumor growth in vivo. 1.2.3.1. SK-MEL—28 xenograft tumor growth To grow SK-MEL-28 xenograft tumors in nude mice, cells were subcutaneously injected into the right side of the dorsalateral area of a group of athymic nude mice. 93 Tumor volumes were measured every two days by using a caliper. Figure 21 shows a representative SK—MEL—28 xenograft tumor growth curve. Shortly after subcutaneous inoculation, lesions were measurable due to swelling. The swellings generally disappeared within two weeks and the volume of the lesions remained not measurable for the next five weeks. Seven weeks after cell inoculation, xenograft tumors started growmg in an exponential manner (Figure 21). Although considerable variation of tumor size between mice was normally observed in this xenograft model, the experimental reproducibility was revealed when tumor growth curves from multiple independent experiments were overlaid on the same plot (Figure 22). 1.2.3.2. LeTx systemic treatment of SK—MEL—28 xenograft tumors In this dissertation project, an established approach for LeTx systemic treatment of cancer xenograft tumors in nude mice was used. Xenogratt tumors were allowed to 3 . . . . . . grow to an average volume of 50 mm , at which time mice were divrded into two groups. Mice in each group were then intravenously injected with LeTx (PA plus LP) or control (PA plus LF_E687C) at a dosage of one standard dose (1 XSD equals 10 ug PA plus 2 ug LP or LF_E687C) every two days for a total of six injections (6 standard doses). Previous studies have shown that this treatment has no obvious adverse effects on the health of athymic nude mice while xenograft tumor growth is inhibited (Abi-Habib et al., 2006a; Depeille et al., 2007; Ding et al., 2008; Huang et al., 2008). As shown in Figure 21, SK-MEL-28 xenograft tumor growth was also inhibited by the same systemic LeTx treatment. 94 1.2.3.3. Sensitivity of MEKcr-expressing xenograft tumors to LeTx systemic treatment The inhibitory effect of LeTx on SK-MEL-28 xenograft tumor growth suggests that MEK signaling pathways are required for melanoma tumor growth in viva. Based on this, sufficiency of MEK signaling pathways for SK-MEL-28 xenograft tumor growth should be testable by expressing MEKcr proteins in xenograft tumors. If the MEK] or MEK2 signaling pathway individually is sufficient for SK-MEL-28 xenograft tumor growth in viva, tumors expressing VS-MEchr or V5-MEK2cr are expected be resistant to the growth inhibition effect of LeTx systemic treatment. To test this, SK-MEL-28 parental cells or cells expressing high levels of V5-MEK1 , VS-MEchr, V5-MEK2, or V5-MEK20r were subcutaneously injected into groups of athymic nude mice to establish SK-MEL-28 xenograft tumors expressing each of these VS-fiision proteins. After these tumors reached an average volume of 50 m3, mice received LeTx or control (PA plus LF_E687C) systemic treatment as described. As shown in Figure 23, VS-MEchr- exressing tumors were still sensitive to LeTx although a slight decrease in sensitivity was observed when compared with parental and wild-type V5-MEK1-expressing tumors. Similarly, tumors expressing V5-MEK2cr were also sensitive to LeTx systemic treatment of LeTx (Figure 24). These results suggest that although the MEK2 signaling pathway alone was sufficient to drive SK-MEL-28 cell proliferation in vitro, neither MEKl nor MEK2 was sufficient for SK-MEL—28 xenograft tumor growth in viva. 95 1.2.3.4. Dose-dependent effects of LeTx on SK-MEL-28 xenograft tumor growth When the MEKcr proteins were tested for their resistance to LF-mediated proteolysis in SK-MEL—28 cells, restored cleavage of MEchr and MEK2cr by LF was observed at the highest concentration of LF (Figure 13A, top panels). Therefore, it was possible that elevated LeTx levels in SK-MEL-28 xenograft tumors were sufficient to overcome the cleavage resistance of MEKcr and masked their sufficiency for tumor growth. To test this, a LeTx dose-dependency xenograft study was conducted. In this experiment, cells were inoculated into groups of 20 mice. After tumors were established, mice were divided into four groups, and mice in each group were intravenously injected with either l>wo_ofioacow.333\\fitn ”Sac—3:0 080V 00 am.n.nohambcwmmahomwhco.Bfiumfipmofi.333\\uat: ”flow 050 memmE c .256 55 532w 839 a. 96: £8 mammoaxo.8_v~m2-m> B 888 05 was $585on 3508:: “cacaomowfi 8:: Bow 3:350 083 85:83 a H 595%? E 338on mm madamcwmm anzntomcab mo am: 03388 23. a $3556 8m 250 mean: Rs 9: 5m- 8228 296 =8 £6 bfiuoalomfiommgbgvtoflo: Z ”am 280 mama: x88 :3 a being «am new on. mam- Sage scagaéaoaeaz o L 308 N00 32m mm; cod- coumfinaooou <75 $248 moo ow. m Se 8%. 288on neaoem 3808 N00 3am mmd vim- wEmmooofi SAM“ SIEQENmézsgmmooom ease com 0:00 mama—z amd 84 £6. 53:8qu EM .3an .Emm GSA :3 3 EB «am as N? E H- as: 9 855.8% Sam: .8552 Ncu~-m> a meow oaagmxaspfimfimobdgwmm -m> -m> a 85:82 4:25.35 what— »: wauooba banana—Mme. 93 «a5 non—5.3»? Buewntcmgh cognates—HE .> 93.; 111 Figure 12. Necessity of MEK] and MEK2 signaling pathways for ERK activation in SK—MEL—28 cells. SK—MEL—28 cells were transfected with nothing (N); lipid only (L); non-silencing control siRNA (NS); MEK-specific siRNA (MEKl siRNA 7, 8, or 11, and MEK2 siRNA 9 or 14), or pools of MEK-specific siRNA (MEKI siRNA 7+8+11 or MEK2 siRNA 9+14) as described in Materials and methods. As controls, cells were transfected with combinations of both MEKl- and MEK2-specific siRNA (P1, MEKl siRNA 8+11 plus MEK2 siRNA 9+14; P2, MEK] siRNA 7+8+11 plus MEK2 siRNA 9+14). After transfection, cells were trypsinized and split to two dishes for cell lysate collection and for cell cycle analysis (results shown in Table IV). Seventy-two hours later, whole cell lysates were harvested and immunoblotted. The efficiency of siRNA-mediated MEK knock-down was examined by immunoblottings with antibodies against MEK] or MEK2. ERK activation was detected by antibodies specifically against phospho-ERK. Total ERK expression was detected by ERK antibody as a control. Antibody against GAPDH was used as an equal loading control. ' 112 Figure 12. Necessity of MEK] and MEK2 signaling pathways for ERK activation in SK—MEL-28 cells. MEK1 siRNA + MEK1 siRNA MEK2 siRNA MEK2 siRNA L NS 7 8 1178119 14 914 P1 P2 [11" .1... 1...... --..:“’"‘ .. JMEKZ 3'” , .. I--..----Q- - ~ mu] ERK1I2-PO4 z .'3;= 3 33.] ERK1I2 113 Figure 13. Individual MEK signaling in LeTx-treated SK-MEL-28 cells. SK-MEL—28 parental cells and cells stably expressing (A) VS-MEK or V5- MEKcr, or (B) V5-lacZ were treated with LeTx (1 ug/ml PA plus 0, 1, 10, or 100 11ng LF) for 24 h. Total cell lysates were then harvested and immunoblotted with antibodies against the V5 epitope to confirm the cleavage resistance of VS-MEKcr (top panels). Antibodies against the carboxyl terminus of MEK] (second panel) and the carboxyl terminus of MEK2 (third panel) were used to demonstrate individual MEK expression in LeTx-treated cells. Antibodies against phospho-ERKl/Z (fourth panel) and total ERKl/Z (fifih panel) were used to examine ERK activation, and an antibody against B-actin and B- tubulin were used as a loading control (bottom panels). 114 Figure 13. Individual MEK signaling in LeTx-treated SK-MEL-28 cells. A V5-MEK1 V5-MEK1cr Parental V5-MEK2 V5-MEK2cr 0 1 10100 0 1 10100 0 1 10100 0 1 10100 O 1 10 100 2 8 fl 0 .05 50,6 0 e U Eat div 2 25% O (I: 0% _ T I t 1 Parental V5-lacZ V5-MEK1 V5-MEK1cr V5-MEK2 V5-MEK26r 127 Figure 21. SK-MEL—28 xenograft tumor growth and systemic treatment with LeTx. SK-MEL-28 xenograft tumors were established as described in Material and methods. Briefly, cells were subcutaneously injected at a number of 107 cells in 100 pl of Hanks' balanced salt solution (HBSS) into the right side of the dorsalateral area of groups of ten athymic nude mice. Tumor volmnes were measured every two days by using a caliper. After the average volume of tumors in the group reached to 50 m3, mice were divided to two groups. Five mice in each group were intravenously injected with either LeTx (PA plus LF) or control (PA plus LF_E687C, an inactive form of LF) prepared in 50 pl of HBSS at a dosage of one standard dose (1 XSD) every two days for a total of six injections (6XSD total). One standard dose equals 10 pg of PA plus 2 pg of LF or LF_E687C. Xenografi tumor growth is presented by average tumor volume (y axis) as a function of time (x axis). The thick solid line represents the tumor growth before treatment. The dashed line represents the growth of control-treated tumors. The thin solid line represents the growth of LeTx-treated tumors. Error bars represent standard deviations of tumor volumes. 128 Figure 21. SK—MEL-28 xenograft tumor growth and systemic treatment with LeTx. 100 asp-Before treatment ‘ - oA- . Control 3 +Le'rx ' 75 A} 5. .11., A .I 25 FT ,. .‘ 111.. Mb- 0 71421 28 35 424956 63 70 778491 98105 Time (Days) Average Tumor Volume (mm‘3) 129 Figure 22. Reproducibility of SK-MEL-28 xenograft tumor growth in athymic nude mice. SK-MEL-28 xenograft tumors were established as described in Material and methods and the legend of Figure 21. Xenograft tumor growth is presented by average tumor volume (y axis) as a function of time (x axis). Tumor growth curves from five independent experiments are presented as lines with different colors and indicated by ND xenograft project numbers. Total numbers (n) of mice used in each project are indicated. Image in this figure is presented in color. 100 + N086, n=14 + N088, n=14 a i- ND89, n=11 < ~9- N091, n=14 E + N093, n=20 E 75 I, 9 a / 3 x B .-/ i, ' > W h 50 all i g '9 O (I? ' E ['1 3. ' / a l— 3;", 0 .9/ /. g 25 > . ’ < , /‘ /J BIC-I... 2835424956637077849198 Days after cell line injection 130 Figure 23. Sensitivity of VS-MEKI cr-expressing xenograft tumors to LeTx. SK-MEL-28 parental xenograft tumors (black lines) and tumors expressing V5- MEKl (green lines) or VS-MEchr (red lines) were established as described in Material and methods and the legend of Figure 21. After the average volume of tumors in each group reached to 75 m3, mice were divided to two groups. Five mice in each group were intravenously injected with either LeTx (PA plus LF) or control G’A plus LF_E687C) at a dosage of one standard dose (1 XSD) every two days for a total of six injections (6XSD total). One standard dose equals 10 pg of PA plus 2 pg of LP or LF_E687C. Xenografi tumor growth is presented by average tumor volume (y axis) as a fimction of time (x axis). The thick solid lines represent the tumor growth before treatment. The dashed lines represent the growth of control-treated tumors. The thin solid lines represent the growth of LeTx-treated tumors. Error bars represent standard deviations of tumor volumes. Image in this figure is presented in color. 131 Figure 23. Sensitivity of VS-MEchr-expressing xenograft tumors to LeTx. 15° -a—Parenta, before treatment 3 . oe- - Parental, control V5-MEK1cr Parental < 125 -_ —0—Parental, LeTx 9 E -|s-V5-MEK1, before treatment A V5-MEK1 .. Q .E, o o» - V5—MEK1, control ' : 0 +V5—MEK1, LeTx 0 § 100 " -a—V5-MEK1cr, before treatment f 3 . 0A0 - V5-MEK1cr, control ' 2 -0—V5-MEK1cr, LeTx ‘ x? o 75 E '3 . é so 5 3 25 0 ..‘—“-Ii;393...ss.‘..u..mm... 0 7 14 21 28 35 424956 63 70 778491 98105 Time (Days) 132 Figure 24. Sensitivity of V5-MEK2cr-expressing xenograft tumors to LeTx. SK-MEL-28 parental xenograft tumors (black lines) and tumors expressing V5- MEK2 (green lines) or V5-MEK2cr (red lines) were established as described in Material and methods and the legend of Figure 21. After the average volume of tumors in each group reached to 50-70 mm3, mice were divided to two groups. Five mice in each group were intravenously injected with LeTx (PA plus LF) or control (PA plus LF_E687C) at a dosage of one standard dose (1 XSD) every two days for a total of six injections (6XSD total). One standard dose equals 10 pg of PA plus 2 pg of LP or LF_E687C. Xenografi tumor growth is presented by average tumor volume (y axis) as a function of time (x axis). The thick solid lines represent the tumor growth before treatment. The dashed lines represent the growth of control-treated tumors. The thin solid lines represent the growth of LeTx-treated tumors. Error bars represent standard deviations of tumor volumes. Image in this figure is presented in color. 133 Figure 24. Sensitivity of VS-MEKZcr-expressing xenograft tumors to LeTx. Average Tumor Volume (mm‘3) 175 -A-Parental, before treatment . on - Parental, control V5-MEK2 150 a -e—Parental, LeTx t -a-V5-MEK2, before treatment .' Parental 125 “ ovo-VS-MEKZ, control 4 v5.MEKgcr A -e—V5-MEK2, LeTx ' .K -a-V5-MEK2cr, before treatment ‘ f .- 100 ll °-A--V5-MEK2cr, control 1 ,5, ' -e—V5-MEK2cr, LeTx . i A A ,0 " 75 ’ ,- A so " 25 0 .2 "‘ seams-.ta'u'd‘l'a's'a‘fi... as...“ 0 714 21 28 35 42 49 56 63 70 77 84 91 98105 Time (Days) 134 Figure 25. Does-dependent effect of LeTx on SK-MEL-28 xenograft tumor growth. SK-MEL—28 parental xenograft tumors (black lines),and tumors expressing V5- MEKZ (green lines) or V5-MEK2cr (red lines) were established in 20 athymic nude mice as described in Material and methods and the legend of Figure 21. After the average volume of tumors in each group reached to 50-70 mm3, mice were divided to four groups. Five mice in each group were intravenously injected with PA plus LF_E687C control (solid triangles) at a dosage of one standard dose, or LeTx (PA plus LF) at a dosage of 1 (open diamonds), 0.5 (open squares), or 0.25 (closed circles) standard dose every two days for a total of six injections. One standard dose equals 10 pg of PA plus 2 pg of LF or LF_E687C. Xenograft tumor growth is presented by average tumor volume (y axis) as a function of time (x axis). The thickest solid lines with open triangle data points represent the tumor growth before treatment. Error bars represent standard deviations of tumor volumes. Image in this figure is presented in color. 135 Figure 25. Does-dependent effect of LeTx on SK—MEL—28 xenograft tumor growth. Average Tumor Volume (mm‘3) 175 A 0| 0 1 .3 M (II 100 N 0| 01 O N 0| 0 -a—Parenta|, before treatment 0 0A0 0 Parental, control 1 SD l-o—Parental, LeTx 1 $0 -—a— Parental, LeTx 0.5 SD . ------ .,....- Parental, LeTx 0.25 SD -a—V5-MEK2, before treatment . 0A0 - V5-MEK2, control 1 SD --+—V5-MEK2, LeTx 1 $0 —a— V5-MEK2, LeTx 0.5 SD ~----------~eV5-MEK2, LeTx 0.25 SD h-Is—VS-MEKZcr, before treatment . vo - V5-MEK2cr, control 1 SD .—o—V5-MEK20r, LeTx 1 $0 —a— V5-MEK20r, LeTx 0.5 SD wow-- V5-MEK20r, LeTx 0.25 SD 0 71421283542 "-"_"___-_-v~ - 7.7.1.4673. . . . u uuuvuuuruuuvuuuruu your“ 49 56 63 70 77 84 91 98105 V5-MEK2 A : Parental 5 V5-MEK2cr A .x . f 1‘ A Time (Days) 136 .353 Sauce mange es. . .983 28:5 $92923on .288 no: 82.8 m> is“... 863:5 5E 8:28.556. 85 so; 835:: 8:83.02 SEE .3 3836:: mpg—9:3 83%— .883 382% was Banana ob? 8:33 .583. .mEoEtomxo 05 we can 05 Ban 8:: Bot Buses 203 22:5 8858.. a A“: Ba <3 £3 .5 650ml": 33 «6 89:8 55 38.3. 3.8835 203 8952 -m> 8 $52-3 .8Qm2-m> .Em2-m> message £25: 8 £85 canoes. wNJmEQm were... 8:: 82. case... .9353 «.3.—ween autumz¢nm E agencies gov—qumNr me 364 6N 0.5»; 137 SWLOON 966 'I-CSZIV 99,BI~ZSI-clvg 0|»? 5.0.. 3.5000 5.0.. 3.5000 5.0.— 3.5000 5.04 38:00 02.0.. 3.8000 5.0.. 33:00 35.5265 35565 32.25.. 5035265 53265 .0395“. .9535 ”.3.-wen: aim—Z05 5 5:29.93 nova—2.3» be $94 6N 9:.me 138 Figure 27. Systemic treatment of LeTx does not cause MEK cleavage in SK—MEL- 28 xenograft tumor cells. SK-MEL-28 parental xenograft tumors were established in athymic nude mice and LeTx was administrated intravenously as described with the following minor . . th . . . . th modification. Two days after the 5 intravenous injection, the 6 LeTx or control treatment (1 SD) was switched to intratumoral injection (it) on selected mice. Other mice still received the 6th intravenous injection (iv). Twenty-four hours after the last administration, tumors (indicated by tumor identification numbers) were dissected for tumor lysates preparation. Immunoblotting was performed using antibodies against NHZ- terminus of MEKl (top panel), carboxyl terminus of MEK] (the second panel), phospho- ERKl/Z (the third panel), total ERK1/2 (the fourth panel), and GAPDH (bottom panel). 139 Figure 27. Systemic treatment of LeTx does not cause MEK cleavage in SK—MEL— 28 xenograft tumor cells. N093 Parental Control 1 SD LeTx 1 80 iv it iv it N h P n §§ss§§ss§ 8 g A_nt__lbody [§--fl¥ 9°? 4”:] MKK1 (NT) [M “.M.. J p-ERK‘I’Z ;- .. . 5:- 7“: '"" I; I: w_ serum total GAPDH 140 Figure 28. Unsupervised clustering of gene expression changes in LeTx-treated cells. SK-MEL—28 parental cells and the cells stably expressing V5-lacZ, VS-MEKl , VS-MEchr, V5-MEK2, or V5-MEK2cr were treated with PA plus LF_E687C control (BC) or LeTx (LF) for 24h as described in Material and methods. Total RNA samples were collected and subjected to cDNA microarray hybridization and data processing as described in Material and methods. Three independent experiments (indicated by _A, _B, and _C) were performed to generate statistical significance. Total RNA isolated from control V5-IacZ-expressing cells (treated with PA plus LF_E687C) were used as a gene expression reference control to normalize the changes in LeTx-treated V5-lacZ-, V5- MEKl cr-, and V5-MEK2cr-expressing cells. Normalized gene expression changes were organized by hierarchical clustering. 141 Figure 28. Unsupervised clustering of gene expression changes in LeTx-treated cells. _l oufifl 55.92-? _l .I mrfifltflxwze> l. mrfifi town—26> .| 3%.. \ 82%-? I 093459ng m 03 \ N87? 1H 3.8.. . New»? r . 5.0.. \ Nom.-m> 254 . _ q . q q 5 0 5 0 5 O 5 5 4 4 3 3 142 1.5. Materials and methods 1.5.1. Cell lines and stable cell line establishment. SK-MEL-28 cells were obtained from CeeTox Inc. and grown in RPMI 1640 medium supplemented with 5% FBS and 50 units/m1 penicillin/streptomycin. Cells were cultured at 37°C in a humidified 5% C02 incubator. To establish stable cell lines, SK- MEL-28 cells were transfected with V5-MEK, VS-MEKcr or V5-IacZ control expression TM vectors by using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s . . . . . ® instructions. Clonal stable transfectants were selected against 1 mg/ml Genetlcm . Expression levels of V5-fusion proteins in each stable clone were determined by immunoblotting. After stable cell lines were isolated, cells were maintained in medium containing 0.5 mg/ml Geneticin®. One day before subsequent assays, stable cells were . . . . . ® subcultured once, and cultured 1n culture medium w1thout Genetlcm . 1.5.2. Chemicals and LeTx U0126 (Calbiochem) and PD 1843 52 (U SBiological) were dissolved in dimethyl sulfoxide (DMSO) (Sigma) at a stock concentration of 100 myml. PA and LF were expressed in an attenuated strain of Bacillus anthracis (BH445) and purified by fast pressure liquid chromatography as described (Bromberg-White & Duesbery, 2008). 1.5.3. siRNA-mediated MEK knock down. 143 Multiple siRNAs specifically against human MEK] or MEK2 and AllStars non- silencing control siRNA were purchased from Qiagen. To deliver siRN A into SK-MEL— TM 28 cells, the siLentFect Lipid (Bio-Rad) was used as the transfection reagent. To do this, SK-MEL-28 cells (3 X 105 cells) were seeded and cultured in 60-mm dishes. The next day, cells were washed three times with PBS and covered with 1.6 ml of OPTI- MEM®I (Invitrogen), to which siRNAzLipid complexes (prepared as described below) were then added. To prepare siRNAzLipid complexes, 10 ul of lipid was added into 30 p1 of OPTI-MEM®I and incubate at room temperature for five minutes. The prepared lipid was then added into 360 pl of OPTI—MEM®I containing 100 pmole of each control or MEK siRNAs plus 100 pmole of AlexaFluor488-conjugated non-silencing siRN A, and incubated at room temperature for 20 minutes to allow siRNAzLipid complexes form. Eight hours after addition of siRNAzLipid complexes, cells were then trypsinized and split to two 60-mm dishes and culture in DMEM containing 10 % FBS and 50 units/ml penicillin/ streptomycin for 72 hours. At this time point, AlexaFluor488 fluorescence could be observed in greater than 95% of cells under a fluorescence microscope (data not shown), indicating a high efficiency of siRNA transfection. Cells were then harvested for immunoblotting and cell cycle analysis. 1.5.4. In-cell MEK cleavage assay in SK—MEL-28 cells. SK-MEL—28 parental cells and cells stably expressing VS-fiision proteins were treated with LeTx in a LF concentration-dependent manner (1 ug/ml PA plus 0, 1, 10, 100 11ng LF) for 24h. Total cell lysates were collected on ice in RIPA lysis buffer [50 144 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 2 mM Na3VO4, 20 mM sodium pyrophosphate, 1% sodium deoxycholate, 1% Triton X-100, 0.1% SDS, and l X EDTA-free protease inhibitor cocktail (Roche)] and homogenized by sonication in ice TM bath. Protein concentrations were determined by BCA Protein Assay Kit (Pierce) according to the manufacturer’s instructions. Lysates were then prepared in SDS sample buffer [47.5% Laemmli Sample Buffer (Bio-Rad) and 2.5% B-mercaptoethanol (Sigma)]. Five micrograms of total cell lysates were subjected to immunoblotting to detect LF- mediated cleavage and ERK activation as described in the results. 1.5.5. Immunoblotting. Five micrograms of total cell lysates were separated in 10% Novex® Pre-Cast Tris-Glycine Gels (Invitrogen) and then electro-transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore) according to the manufacturers’ instructions. Membranes were then soaked in 5% non-fat milk for 1 hour and hybridized with primary antibodies against V5 epitope (Bethyl Laboratories), NHz-terminus of MEK] (Upstate, #07-641), COOH-terminus of MEK] (Santa Cruz Biotechnology, SC-219), NHZ- terminus of MEK2 (Santa Cruz Biotechnology, SC-524), COOH-terminus of MEK2 Santa Cruz Biotechnology, SC-525), phospho-ERK (Cell Signaling, #9106), ERK (Cell Signaling, #9102), phospho-p38 MAPK (Cell Signaling #4631), phospho-JNK (Cell Signaling #9255), a-tubulin (Sigma, T9026), B-tubulin (Sigma, T5201), B-actin (Sigma, A1978), or GAPDH (Cell Signaling, #2118) at 4°C for overnight. Conditions for primary antibody hybridizations were followed according to the antibody datasheets. 145 After primary antibody hybridization, membranes were washed three times in TBST buffer (50 mM Tris, 150 mM NaCl, and 0.1% Tween-20), hybridized with HRP- conjugated secondary antibodies according to the instructions of the antibodies, and then washed three times in TBST buffer. Immunoblotting signals were then detected by TM LumiGLO Reagent and Peroxide (Cell Signaling). 1.5.6. Toxicity assay. For LeTx toxicity assay, SK-MEL-28 cells (1,500 cells) were cultured in 96—well plates for 24 hours, and then treated with control (1 ug/ml PA) or LeTx (l ug/ml PA plus 0.01-10,000 ng/ml LF) for 72h. For U0126 toxicity assay, cells were treated with DMSO control or 1-100,000 nM U0126 for 72h. For PD 1843 52 toxicity assay, cells were treated with DMSO control or 0.01-10,000 nM PD 1843 52 for 72h. Cell viability was determined by using CellTiter 96® Aqueous Non-Radioactive Cell Proliferation Assay (Promega) according to the manufacturer’s instructions. LeTx toxicity was presented by plotting the relative viability (normalized by no LF control) against LF concentrations. IC50 values were determined by SigmaPlot software. 1.5.7. SK—MEL—28 tumor xenograft and LeTx systemic treatment SK-MEL-28 parental cells or cells stably expressing VS-MEK or VS-MEKcr were cultured in exponential growth phase and then collected in Hanks' balanced salt solution (HBSS) (Invitrogen) at a density of 108 cells/ml. Cells were then subcutaneously injected at a number of 107 cells (in 100 pl of HBSS) into the right side 146 of the dorsalateral area of groups of ten athymic nude mice (Charles River Laboratories). Tumor volumes were measured every two days by using a caliper. Animal health and tumor volume were monitored till the end of the experiment. After the average volume of tumors in the group reached to 50 m3, mice were divided to two groups. Five mice in each group were intravenously injected with either LeTx (PA plus LF) or control (PA plus LF_E687C, an inactive form of LF) prepared in 50 pl of HBSS at a dosage of one standard dose (1 >ueo<<233883 98 souaificfi mo eoumfinaoo S|o|mem§o§muo ”am use mama: 3m :2 as. See coinage Emm 815.20g? new use mama: 8a o3 :é. x shoe 83683-0»: use a: Goom :3 a. 2m: use m3. «2 is- a»: a 88$; .5sz a Em: NSE> a3 afieiaafiaeafiaaa -m> -m> e 85:83 ~0ng .3 6253.. 52:85:»? 6:: 2.25.3.5 who..— .3 wouiwouugee 0.3 :2: 3.53:»? _anetatomaauh A €=25< 164 83898 83. an m _ .m- 33:3 <75 82.3335 EZec flow 0503>a35afimo§m-wmm a 8:333 165 2.8 552 a .8833 32833 A88 3334 38m .3 3 333d .33 33 one 3.3.- 8% 33332.33 33 35 380 £3 3388 233 3230 x38. :3 3 333$ .33 3.». 32 8.3. 33 33338 23» 3230 m2 3332: 23» 3230 x38 :3 3 3338 .33 33.3 NS 8.3. m2 3332: 353 3230 8 2:32: 23» 3230 x38 .3 3 3339 .33 23 _E 33.3- S 33336 233 3230 manuzhflmae? 3m 230 mama: x88 ..3 3 38$ .33 mom one 2.3- 38» 3333832333 zauqq flow ocoOR>33€afimo§3—&mm -m> -m> 3 85383 166 98% b.2558 0308 3.595 $159932; 3m 250 mama: SN. 23 an». «a 5 8332.9. as as 880 awmxézfio’xvofi 8m 250 mama: 8a. 8.? N3 “3-23:8 8m 080 mafia: 35. 33. SN Ba 3585 3. x8880: 58898 EN. woo- ova mews; ooamoaofimoé zalmmooupmfimomemlzammm 8:535.“ spam pom 250 mama: x38 :3 3 328 «um 8+. 8. T 9% E 833?-..38 8“ as 850 high ogwoono mg zgnzuw ”am 080 832 NE- 3o- o3 space .732 mo coofionfisz gab: O 093 SE88 63332:” 0521220 new 080 mama: 3a. 93. as 2:85 53% .6 Bofiofiwsz manézomuéfiéa 8m 050 mawaz G3 33 SN- 8:% Baawoasaiaize m=oo FADE S #58895 Eon 2888 soon £83 68m :3 3 send mom SN 84 SN- 8% 8332.9. 2“ 35 880 £8 E02 5 Beaumon— 3353333 829% ‘ EON £63 ”88. :3 3 Quad gem RN w _ A man. 8% 8838-598 03 as 880 a 885$ 63m: aim: N335 38 Sooigagmofigwa -m> -m> 85:33 167 ._Efido3wofil>nlom38£Bo§oB§zBB.Eomfiamdutch—chug: ”33on 09% 5050 ”\wl—oxwo—ofioocomékrzrkugn ”came—8:0 33$ 00 Em_..aouwom\nnw_m8\somw\wuo335530~D§BB§at: ”flow 050 memmE a .256 55 83on mos—g . Q 96: £3 wfimmofixo.8~v~m2-m> 5 880m 05 was figs—tong 3580:: E09589: 08% Sch @0538 0.53 moumuflmé a Room :3 3 56 «oz cod- wed- mwé 85% NEE Cbom r3 8 Emfiév mom OWN- 3;. 9mm méz a 858mom ~0ng .5552 Noc~-m> flow oaowhxmafimfimohsmzwmm -m> -m> a 8:383 168 Appendix II. Transcriptional signatures that are affected by LeTx treatment and significantly rescued by both MEchr and MEK2cr 169 znuavfimISHZm 8m 25a mag: m; «3 3m- znuovfimlfinzm m DEUCE 08w .5280 ~N.O~ V0.0 . mwdu m 24608 2.5m .5280 mm 07608 08w .5050 ©w.© 056 mod- mm DEUCE 05% .5050 Svozmufizo com 250 8%: 8.0 ”3 mod- Seozmufizo <20m|~mzo com 250 3%: a; man 2d- 90m 0G00\m%m>>5mm\mo§m&mw a 83333 .8ng was hos—HE 52— .3 1283.. .5.—35:»: E... 2.25.3.5 who:— .3 683:: 0.3 2:: 3:52—me iueflntomnauh .= anomad‘ 170 ”2 0258 25» beau 3m 5m 8s. m: 0388 08» beau 339; ”B 8295 w? «2. N3- .5 52%? 2:33: ”a 3295 3m 9% 8.»- n5 ”Bade: Saddam? 8m 080 8me 26 gm :3. Snoodmmm> m2 2:88 8% 5050 an own 3.”- g 0388 25w 588 w 2:on 25w 5280 a; v3“ vow- w 2358 25m .50ch S 2358 08» beau one 0? 3w- 3 0388 25» 825 53.220 3m 080 mama: 8.0 «.3 v3- voéummzo «Eozlfizo 8m 089 mama: 86 8+ m3- $621820 mm 0258 25m .850 R6 25 9;- Ma 0:62: 08w 5050 m§ 30m 0§U\m%m35wm\wo§mflmmm a 89353 171 513%? ”am 080 mama: :3 Rd mi. 54293 2.228.320 8m 250 mama: :6 a? 8.». 2.2281320 SN 0358 25m 505.0 mg 8.9 NE. SN 2:88 08» beau 5. 2:85 8% 525 3.0 8.0 N2- 5 2258 8% .5030 $289.20 ”5m 250 8%: 03 a3 3. ~ 30m ocochxmafiafimogacwmm a 8:383 172 220:220 80 080 0000: 00+ 00.0 2.0- 220:220 800:220 “00 080 000202 3.0 8+ +00- 800:220 20200310220022 0.00 080 00062 :0 .2 +00- Su+0000|02<|2002mzezugum>uq<2215 000 0000 0005: 5% 00.0 2.0- 20u>mzo§|EIm>J<02|E +2 03020 080 0850 5+ 00.0 5.0. +2 0302: 080 0880 000>0J020n2 _3 300 0000\020350Qmoggwmm 0 0020280-“ 173 000001220 000 080 000002 :0 0++ ++.0- 000001220 002001220 000 0000 0000: 00+ 00+ 00.0- 002001220 02-01220 000 080 050002 00+ 00.0 00.0- 00.01220 0000000 ”00 00.0 00.0 00.0- 080 0008002 23022201330000.0000; 000 080 050002 00.0 0+.0 00.0- 2302220133555; 00+ 20008 0000 .8000 00+ 00.0 00.0- 00+ 03008 0000 0850 000000: ”B 000,300 00+ 00.0 00.0- .5 20 00. .22: 0580080000 2010023000102 000 080 050002 00.0 00+ 00.0- 2012<10001200 0000000 ”00 00+ 00 00.0- 0308 020005.00 0020000210020me 000 050 000002 +0+ 00+ 00.0- 00200021002200 00201001002202 000 080 000002 00.0 00+ 00.0- 00200100100280: 0012300002100 000 0:00 05000.2 _00 00.0 +0.0- 001250002100 000000: “B 002000 00.0 00.0 00.0- .5 2: 0 200228 00100005010210.05020000 000 080 05000.2 30 00.0 00.0- 001020.001021005020000 22221220 000 0000 050002 00.0 00+ 000. 02221220 0 000000-002 Hum-WW: BmW—ME N00~10> 0000 ogwhmaafimfimoganwmm 0 002000000 174 EL. r70.- 00100002012-01002-00 000 080 000002 0+.0 2+ 051 001000020600100200 00 0 03008 050 .8000 00+ 00.0 00+- 000 20008 080 0050 000000: ”9 002000 00.0 00.0 00+. 00 28 0 00020232050800 020001220 000 080 0500: 00+ 00.0 00+. 020201220 0.0000002012055000 000 080 0500: 00+ ++.0 00+- 00000000000120.2300 0200201221202 000 0000 000002 +0.0 00.0 00+: 0200201221202 00:00: ”B 000,200 _00 00.0 00+- 00 00.055020 22001220 000 080 0000: 00+ 00.0 00+. 0002001220 0010001000 000 080 050002 00.0 0+0 00+. 0010001000 0200020020 000 080 050002 00.0 00.0 20. 0200020220 03020001222002010-0100200002 00002 000 0000 0000: 00+ 00.0 00.0- 00012200000210.0100200002 00221220 000 050 0500: 00+ 00.0 00.0- . 02221220 03000010010000001020 000 080 0000: 00+ 0+0 00.0- 0+000001o01m-00001dm0 1 1 <2 0000000_ ”B 0020.5 00+ 00.0 00.0- 0+E.E§000§.0000 0000 080. 00150210530222":02012025 0015.021 000 0000 050002 00.0 00.0 0+0- 00.03020020020120050 0 000000.002 bmWME 8 ”WE N00~1m> $00 ocowkxaBfimfimoggwmm 0 002000000 175 +00 03008 080 08000 00.0 00.0 00+. +00 03008 080 08000 50001220 000 080 000002 0++ 00.0 00+. 02001220 00000001000510.0000 000 080 000002 +0+ 00.0 00.? 00.200201000061020 0020010005 0002100021000 000 0:00 000002 00.0 00.0 +++- 00.20010030000010-002120 20001220 000 080 000002 00.0 00.0 0++- 20001220 00100<10025<0 000 080 0000: 00+ 00.0 0+.+- 00100<1o0200<0 201>z<102 000 080 000002 00.0 00.0 0+.+- 201>2<102 000000: ”00 002000 00.0 00.0 0+? 00 2: 0 222: 20000800 1 1 200005000201 1 1 00.20000 02200100 0220.50 000+0<02 2000500020 000.2002 02 000 0000 000002 +_+ 00.0 00+- 20.501022030100035: 02212200020020 000 080 000002 00.0 0.0 00+- . 221-2200020020 000000010012000.20000210000 0000 000 080 000002 :0 00.0 00+. 00010012022052.3000 300201220 000 050 000002 00.0 _00 00.01 500201220 0200200312100200000 000 080 0000: 0+ _0.0 00+- 02520031810020.0002 3030013000000: 000 080 0000: 00+ 00.0 20+- 30500130000002 0 Dog-5.00% 8w”: .5 WWW: NUQN1W> flow 0G00\m%035«m\m0§wcwmm 0 000000000 176 H... LJ 000000: ”00 002000 00+ 00.0 00+. 2300 2: 00 00000 2.020.31002.2002201201200050002 1 15000-0300 000 0000 000002 00.0 00.0 00+- 502025:- 20 20250000 0030000 ”0. 002000 00+ 00.0 00+- 00 0.002 0 _ 0+000 U00 00+ 00.0 +0 .01 00,08 008302 001E1§1S§1§02102<12<<220> 1 1 1 1 “510.310 000 080 000002 +2.0 00.0 0_.+- > 52 :202 00,2 2<<220> 0000000 ”00 2+ 00.0 023 00202 000000 ”00 3+ 0.0 0 _ .+. 2000 =8 00220 00 00000 2 0000000 “00 00.0 00.0 0:1 00,000 0820 5380 002021220 000 080 00002 2+ 0+.0 00+. 002021220 E0200<100<22100120E<00002 1 1 1 E20. 000 0000 000002 00.0 00.0 00+- 0< 00302 .00 29.050000 '- I I I t I 0>F0< 00<2§ 220.02 00 200050002 1 1 21020-3 00<2 000 0:00 00002 00.0 0.0 00+- 2 200002 .00 29.2.5002 00120210000100.0020 000 080 000002 00.0 0+.0 00+. 00120210000100.0020 00<>1xom00<102<020 000 080 000002 00.0 00.0 00+- 03001202030252: 0000000 ”00 00+ 0.0 00+. 2000 08 000002 0 8020000 50002 6202 020-00, 003. 00000003500850.0000 -0> -0> 0 00200800 177 00000000 ”00 002000 00.0 00.0 00.0- 2300 2: 00 000008580 0010.2020010001005020020 000 080 000002 00.0 00+ 00.0- 001<2000010001005020000 00213001+0001002122<2002 200 000 050 0000: 00+ 000 00.0- 013001+0001002122<2002 0000000 ”00 _+.+ 00.0 00.0- 000008 =8 00 00003000 201-20.02 000 080 0000: 00+ 0++ 00.0- 201000.02 0000000 ”00 00+ 00.0 00. 0008000058 0000. 002012 000 080 000002 000 00.0 00.0- 003212 20350120231250: 0.00 0:00 000002 00.0 00.0 00.0- 20020510000000.1253 0000000 ”00 00.0 00.0 00.0- 0509.800 2000 :00 02025120022012? 000 080 000002 00.0 00.0 000- 00000201200322.1050 00000010000102.0002 000 080 00002 +0 00.0 00.0- 00051000010202: 0010-2100070 0.00 080 00002 00.0 00.0 00.0- 0010-21007: 00000301000103 000 050 0000: 00+ 000 0+. 02000301200103 00201000061020 000 080 000002 00.0 00.0 00+- 00201000061020 2010262002000 000 080 0000: 00+ 20 00+- 2010215002000 0 oocobmom pom-WM} bmwwmrzz N0001m> 0000 0000\000350Q00030020m a momumflwwmuu 178 ringing-1.53.! 0.1... . .. .y 29002310201022.0000 000 0000 000002 00.0 00+ 00.0- 20000231020103.2000 E>F0<100<00002 000 050 000002 +0.0 00.0 00.0- 3030310020002 0.000020100001200 000 0000 000002 00.0 000 00.0- 02005100001200 20000001200103 000 080 0000: 00+ 000 00.0- 0.000000010000103 ++0 0000 ”00 00+ 0++ +0 .0- 08802300 232803, 000 0000 ”00 0+ 00+ 000- 05802200 0002, 0020 00000002 ”00 002000 00+ 00.0 _00- 2300 20 000 0000008030 0000000 ”00 0+ 00.0 00.0- 08802300 000.0 \>om.:E.000.&0\\”&E 000w0 $000002 00000020000 - 0020... 00.0 002 600 020800 23800 00+ 00.0 00.0- ”02530002 0. 0+000 0010000021200 000 2.00 0000: 00+ 00.0 0+0- 001000202120. 20102080210212?me 000 050 000002 00.0 00.0 0+0- 201-02000021021000.5000 00010220<000<>1<0000<2 000 0000 000002 00.0 2+ 0+0- 00010020<200<>1<0000<2 2010+00210+0<1><0<00< 000 050 000002 00.0 00+ 00.0- 2010202105955? , 20003010062102 000 800 0000: 00+ 00 00.0- 00000000010015.0109 0 000000002 BN-WME Hog-WM} N00~-m> 0000 0000\000350Q000300wmm U momumflaumna 179 3:080:0>:0000:0<>_350:00: 00:00 000 0000 00002 00.0- 00.0. 00.0 00:0>:0000:0<>_>000:00m 020000 ”00 000. 00.0. 00.0. 2303 09.08-00.020 000005 000 _ 000 ”00 00.0 00.0 00.0- 000802008 00.80 00 00020000 0 03008 050 0800 00.0 ~00 00.0- 0 03008 0000 00000 0 330 000 200 00302 00.0 00.0 00.0- 0:: 000 000080 3:5ng 002000 ”00 00.0 00.0 00.0- 0308208 00 0002002 020000 0002050000020 000 200 00002 02.0 00.0 00.0- 006205000002": 00 20008 0000 50000 00.0 0 2 .0 00.0- 00 20008 080 00000 _00 20008 080 0200 00.0 2.0 00.0- am 03008 200 00000 20:x0000<:02<:a 000 200 0000: $0 00.0 00.0- 20:200034503 00:0000:>20::<2,0 000 200 000002 02.0 00.0 20.0- Edy—$920002”: 002000 ”00 00.0 00.0 :2. 0000005030 00080 0:850:20: 000 050 0000: 00:0. 3.0 00.0- 0:050:20 0000000 ”00 00.0 00.0 00.0- 0008000 =8 00 00003032 5:025:03 000 0000 000202 00.0 00.0 00.0- 5:025:03 a 0000.600M BWWME BMW—2 N007m> 0000 0000\020350E00500wmm 0 00005000..“ 180 .Eb:.00_038I00I0038£000§0\0§002\500.0.8.053000:30:}9E ”001608 000w 000000 {waxwofiaoocowéghdta ”Ago—8:0 050V 00 E00.500000\€w_08\000w\m800300050000930.0030BE ”000m 0000 mama—2 .086 :05 088% 0030M & 0%: 0:00 3800000095 ~vm=20> E 000000 05 0:0 000055020 00.58028 0000:0009: 00.05 80¢ 0000050 0.003 000000000-“ 0 181 Appendix III. Transcriptional signatures that are not affected by LF treatment but cannot be rescued by either MEchr or MEK2cr 182 183 0000000 ”00 00.0 00.0 00.0- 000,000 008000 <20 20.003000200000000 000 200 00002 00.0 00.0 20. 20:0.0<0:0002000000::0 002000 “00 00.0 00.0 000- 0000000 <20E 0000000 ”00 00.0 000 00.0- 0000282 <20 002000 ”00 00.0 000 00.0- 00000080. <20 00 08600000000 0000000 ”00 00.0 000 00.0- 0000000 <20 0000000 ”00 00.0 00.0 00.0- 0000000 000 00002 00000.& 800000 0000000 ”00 00.0 00.2 00.0- 000800 00 0002002 30000 202552525 000 200 000002 00.0 00.0- 00.0- 2920902525 0_ _0000 ”00 00.0- 00.0 00.0- 0000>000 :8 ,0 0.80055 200 320:: 0:0 .000 _00 “00 0: 02 00.0- 0008000 0208000 00008002 00 00000 H00 02 00.0 00.0- 000,08 008000 0000002 US 002000 00.2 00.0 00.0- <2 000500.080. 0 000000030 BMWME 5 ”Wm/dz N00~:m> 0000 0000\000350E00030&_m 0 0030390-» you—HE .3 005—82 00500 00. 60:000.. 0.. 00:58 :5 «no—500.5 rad 03 68000.3 00: 9:0 :5» 00032.30 13030—20050..." 4: 0:50.30. 0500:000220000003000: : :05 000 0000 0:00:02 :00 0:0- 00.0- 00 00000200000>00000 :00 00 0000:0000 30000 00000600 0000000 ”00 00.0- 00.0- 00.0- 0000000200 000000000 030000 0003:000-002000000030d0: : :50 000 0000 000002 00.0 :0- 00.0-- 0< 00022000005020 00: 00: 03000. 0000 000000 00.0 :0- 00.0- 00: 0.0008 0000 00000 : : : <2: amt-000 0000000: ”a 002000 00.0- 00.0 00.0- 000:0 :0 00000000 00 00500.0 000000 0000000 ”00 00.0- 00.0- 00.0- 00000050 000008080000 00:2 20:00<-002:02200 000 0:00 00002 00.0 _0. _- 00.0- 20300502020000 0000000 ”00 00.: :0 00.0- 0000>000 :00 00 00003000 0000000 ”00 00.: 00.0 00.0- 00002000 000000320 00000000 00:03.00 0000000 ”00 00.0 :0- 00.0- =00 :0 0000:0000 020000 a 00:000-00M .8sz B~Mm2 N00~:m> 0000 0:00\0>03£0m\00030:w_m -0> -0> 0 0000:0000 184 00Zm0unm0<000m0120000oz300 I 00:000-005- 000 0000 0:00:02 00.: 00.0 00.0- 00000 20000020. 2:500:00 201000120000400000 000 0000 000002 :00 00.0- 00.0- 20100023004300 050800 30800-00030 5300032305: Bob 0:05 0.800008 05:55 .00 0030:5889: 0:088 0000000 ”00 00.0 :00 00.0- 00 00000 00000000 000.000 300000- 00000500000002 000 0000 0:00:02 00.0 00.0 00.0- 0.0000002010005007: 000.5808 0 0000000 ”00 00.: :00 00.0- 000000 0000000 0:380 003000000: 2300 0000000: ”B 00200.: 0:: 00.0 00.0- :20 00: :22: 0000000000000 :000000 H00 00.: 00.0 00.0- 000000008 <20 0020010000000200001300000100200,: I 000000100 000 0000 0:00:02 00.: 00.0 00.0- 000002-000 $0000: 0020:): £000.00: 000 0000 0:00:02 00.: 00.0 00.0- :0000u0002 303000 0080-0055—0508 0000000 “00 00.0 :00 00.0- 00000000000200.0000000000 2000010000,: 000 0000 00000:): :00 :00 00.0- 20000000: 0000000 Ho0 00.0 :00- 00.0- 000,000 0.000000 00000003000 0 8000.000 00000: 00 :00: 000:-0> 0:00 0000000300000000000 -0> -0> 0 00000000-“ 185 $I2:<00:I0z:00< ”:00 0000 0:00:02 0:0- 0:0 0:.0 0::I2:<00I02:m0< 0000000: ”0: 002000 00.0- 00.0 0:.0 0300 000300500 >000I0<00002 000 0000 05000,: 00.0 0:.0- 00.0- >000I0 000 0000 0:00:02 00.0 00.0 00.0.. 00I:-0<000> E<00000I0EO0000< 000 0000 0:00:02 00.: . 0:: :00- 2<00000I00000000< 0000m>20Im00mz<000 000 0000 0:00:02 0:0- 00.0- 00.0- 00000020000020.0000 00000520 000 0000 0:00:02 0:0- 00.0- 00.0- 00000520 000:000 “00 000 00.0- 00.0- 0000>000 000000;:00 0000:0000 0000000 ”00 :0: 00.0 00.0- 00000 00000000002002 m 00 : 0000 ”00 0:0- 00.0- 00.0- 00,000 0.0000000000000002 I I >002000 300 0500000350385035 0 00000800 0000000: ”0: 0002000 00.:- 0:.0 :00 00 :.00m> 000-0020002030 000 0000 00000:): 00.0- 00.: 00.0 000-002.0353 :0I00I:000> 000 0000 0:00:02 0:.0 00.0- 00.0 :0I00I:000> :00:000 ”00 00.0- 00.0 00.0 =00 0: 00000200 00000000002 0000000 ”00 00.0 00.0 00.0 00000000000 000< 0000000 H00 00.0 00.0 00.0 000000 00 00000000000 000002<00I000<0002Im020m> 0.00 000 0000 0:00:02 00. :- 00.0- 00.0 002<00I000S002Im00000> 0000000: :0: 002000 00. :- :00- 00.0 0300 00.0>.0<00 :00 0:000:0 0000 000000 00. :- 00. :- 00.0 :00 030000 0000 000000 003800 0: :0000 ”00 :00- 00.0 00.0 00:00:00 :08 0005:0000 000:-0< 20:000I002I0000 ”:00 0000 000:0: 00. :- 00.0 00.0 20:0:0I002I0000 20:0000000I2000I0200I00000<0= 20:00 000 0000 0:00:02 00.0- :0 00.0 00000I2000I0200I00000<00 00I00> 000 0000 0000000,: 00. :- 00.0- 00.0 00I00>0> 0 000000-030 ENHWME BMW/=2 N373, 0000 00000000350000.0300me 0 00000500 187 00::0oo0> 000 0000 05000,: 00.0- 0:.0- 00.0 004,000? 0000000 000 00.0- 00.0- 00.0 00000000: 000000 0: :0000 do 00.0- 00.0- 00.0 0000 00000800,: 5.00000, 000 0000 05000,: 00.:- 00.0- 00.0 8100:0000 0000000: ”0:: 002000 00.:- 0:.0- 00.0 0300 0o.0>.0<0:: 00'00m0> 000 0000 050:0: 00.0- 00.0- 00.0 00:00m0> 0000000 How 00.0- 00.0- 00.0 000:: 20000000: 0000000: 00:: 002000 :0.:- 0:.0 :0.0 7:300: 00: 0000000: ”0:: 00:200.: :0. :- 0:.0 :00 0000:0 50020 0: :0000 000 00.0- 00.0- 00.0 :08 00006 000:000 ”00 :0. :- 0 : .0 00.0 00:00:00 00 00000000: 0000000 How 00.0 00.0- 0: .0 :00 000:: 0000000000 8505024000590 000 0000 0:90:02 00. :- 00.0 :00 51000505005520; \>ow.£::._o:.&_0\\x§: Emma warm—5100 - 000000 0:00: :07: 0000 0000600 0000000 00.0- 0:.0 00.0 0:20 2:000:00 0.00:000 €6.52.“ 0000 :00 How 00.0 00.0- 00.0 0: 00000000 00000 00 0000:0000: a 3:85.03: ENHWME BMW/:2 Nuu~-m> 080 ocoO\0>0350m\0o§00:wmm 0 00:00:30-: 188 ._EE.0o3:02:50plo0300£Bo§o\0§Bz\=wo68.090000033:5090: :02:on 008w 08:80 a: 00.800 2:: 98 000580598 000.5098? 0:353:05 025 89a 35030 0.83 00:00:80-: 0 189 Appendix IV. Transcriptional signatures that are not affected by LF treatment but affected by VS-MEchr expression 190 20:00:00.: 000 0000 0:00:02 0:. :- 00.0- 00.0 20100105 00|0MB0IE<0010020 000 0000 0:00:02 00.: 00.0 00.0 00:00:30u2:<00:|0:0:00 0000000 000 0000 0:00:02 00.: 00.0 :00 00000 0:0 0030000500: 0000000 ”00 00.0 :00 :00 00000 00 0000:0000 0000007: 00:00.5; 0.00 0000 0:00:02 00.: 00.0 00.0 0010:0000 00:00.00?an: 00 0000 0:00:02 00.: 00.0 00.0 0010.230120: 00 0:0000: 0000 000000 00.0- 00.0- 00.0- 00 20:00.0 0000 08000 00105005002062.0300 000 0000 00002 00.: 00.0 00.0- 00.030050020620100 292003.300 000 0000 0:00:02 00.: 00.0 00.0- 20:05:21,500 05:00:me 000 0000 0:00:02 :0.: 0:.0 00.0- 0510:1000 001020.000 0.00 0000 0:00:02 00.: 00.0 00.0- 00:020..me I I 000000003000 u 20200000000 0:0 20:05:00.2 05:20.02 0000000 0002020000050 000 0000 0:00:02 00.: 00.0 00.:- 100:7:0:0<,:000:0um>:,0<0mz 0 00008003: BMW”: 00 ”Wm/=2 N007m> 0000 0000000350m00§0§mm 0050580-: 0 . . 1]! 003000500 355—20> 03 6390b: :5 0:0—500.5 ha— »: cowuoba «0: 0.3 :05 09:50:90 _auoflntoauuh .>— 0:051:30 191 u . ‘- F .04: 01... o .00.... 0'— ._EE.003008..»nl00300é00000800802300000.00.000.00000Q0t0g: ”00:60:: 000m 000000 ”\w00xwo—0000000wégtfit: A3285 0:09 00 a030000033008000m\w00.0085050005.333:”BE ”000m 0000 mawmmz . wood :05 000.00% 0020m 00 020:: 0:00 wE000ax0¢3MmE$> E 000000 05 0:0 0000800000 0000:00me 0000000005 00:5 80¢ 005030 0.53 00:00:30-0 0 192 Appendix V. Transcriptional signatures that are not affected by LF treatment but affected by V5-MEK2cr expression 193 $088 ”00 $0. 3.0 8.0- 00.68 0:55 805.. 00:E§> 00m 080 0006: SN 5.0 08- 00:E§> 2 0388 250 0850 3.0 ~00 8.0- a 2:88 25» 08:00 £088 ”00 RN 8.0 as- 8080 0328 £305 0%. 250 0005: 6+ 35 So- 3.: .50 8058 ”00 2.0 8.0 3.0.. 0505.0 50800 gingham? 00m 080 0006: 00m 2.0 3.0- 0013:0030, £305 00m 080 0005: $0 03 8.0- $3 .0000 300000 0200000000 0888 ”00 02 NM: NM. 0- 800% 80?. 26080 08 8005 23:20 0% 250 000%: 0.3 3.0- 2.0- SE: .20 @388 ”00 8.0 2.0 mm. 0- as: 2,0080% 3 38080 $00000 00000050000 8: So ”00 02 3: mm. 0- 529a 00 800302 96080 5608 ”00 85 02 am. 0- 00.58 030108000 zaummmamélmmmfimxo 00m 250 00052 03 3.0 8.0- 20000050001300.0me 0 00000003— BmWME BMW—2 N00~-m> 30m 0000\30350Qm00300w0m 0 85:80-0 000800900 0085200» .3 000250 :5 000050000 ma .3 000000.00 00: 000 0000 0000000000 3000000000000. .> 00000007.. 194 .0000:.00300000I>0100300£000§00§001300000-000000.000000%9E ”00300000 000w 000000 mh0oxw0000000000w§3§>900 ”ammo—00:0 00000 00 am20000033008000w\w00.003000500000.333\\uat: “000m 0000 meEE .300 005 000.00% 830m A 023 0:00 w00mm00qx0.000v~m2-m> 00 000000 08 05 5000000000900 0000000000000 0000000000000 0005 800-0 000000000 0003 0000000300 0 00.0 03008 080 08000 00.0 00.0 <2 0% 03008 080 08000 50000 ”00 00 .0- 00. 0- 00.0 E088 8000 020050 0.0 00.008 050 0850 00.0- 00. 0- 00.0 90 03008 250 08:00 02.0000 H00 00.0- 00.0- 00.0 0000088000 200:6 000 250 000002 00.0. 00.0 00.0 2000 .000 02-002.000-0020-00005 000 250 000002 00.0- 00.0- 00.0 02-002.000-002.0-0000<0 001000.000? 000 250 000002 00.0 00.0 00.0 00180-00000» 00000000008 0>00300£00000 .0000 .00 0000005000 00000000000000.0000 0000 _00 ”00 00.0- 00.0- 2.0 e 020:8 .0308 800-5. 0000 _00 ”00 00.0 00.0 00.0 05005 5835 Q oofiobmom BMW”: .5 “WM—2 NUQNum> meow oaomv\m%m>>5am\moéafiwmm 0 0000000800 195 Appendix VI. Transcriptional signatures that are not affected by LF treatment but affected by both V5-MEchr and VS-MEKZcr expressions 196 00I00I0§00I0002I<050<0 000 080 000002 00.0 00.0 00. _- 00I00I0§00I0002I<0mm<0 00902900620000. 0%. 080 000002 00.0 :0 00. 0- 00902000062000.6- 00000000I20I<00Ixe 000 2000 0000: 0.0.0 00.0 2.0- 00000000I20I<00Ix00 zgozmzaIB0FzzzZzzm<<<>m I 2302 000 0000 000002 00.0 0.0.0 2.0- 0075 30007022223: 00I00<00000I0xmmm>04<0mm I I I 00 000 0000 000002 00.0 00.0 00.0- 0050000 000000-50 2000 0000000 ”00 00.0 00.0 mm. 0- 0500085 055800 0DImmmvamoiIEEBIEEDmIqum I I I SHEEN 000 050 000002 00.0 00.0 00.0- 00000 52.3 20.200 0000 20I0002020>Im§d<0§2Immq 000 080 000002 00.0. 02 00. 0- 00I0>2Immq 0 000000.030 003502 00 _anz N00~Im> 000m 0000\003350Q0003000w0m .9 .3 0 00000000000 000000000900 00052 Im> 0:0 00 “Mm—20> 500 .3 00000000 0:: 0000050000 0: .3 0200b: 0000 000 005 00030500 .00050—0000000H .—> 05:00.50- 197 20I0§00I000000020I>200I>20 I I anImm: 000 250 000002 00.0 0.3. 00.0- 00 0000000200 >20: >20 00000000 ”B 82000 00.0 00.0 00.0- 2300 :00 0 03008 800 08000 00.0 00.0 00.0- 0 20008 080 08000 00 03008 050 08000 00.0 00.0 :0- 00 20008 080 000000 00600000000600.2002 000 080 000002 00.0 00.0 00.0- 00ISx00>mI0000I002mmaam 000 080 000002 00.0. 00.0 00.0- 02020000>00Im02mmemm 00I00202000Imsmm-000 000 080 000002 00.0 00.0 00.0- 00Impz020~mIm<000000 000:5 000 080 000002 :01 00.0- 00.0- 0.00000 20I220mI>20 I I IzmI-0 000 00an0 000002 00.0 :0 00.0- 0< 0000000200 >200 >20 0000.2? 000 080 00052 00.0 00.0. 00.0- 00020? 000d 9500me 00260505 I I I I 000 080 00002 00.0 03 00.0- 00 00 0500: 0.52 0.00005- 0 8800000 003002 030002 Nos-g 03 080\m00350&mo§0&_m -m> -0> 0 00000000000 198 ".0 r4hll 5.4: ‘3I.EIJ.-.1 00m 20008 250 0850 00.0 00.0 00.0 000 03008 250 08:00 0:0-056000000 000 080 000002 00.0 00.0 00.0 000206000000- 0: 03008 080 0850 00.0 00.0 00.0 E 03005 250 08000 0085m0£ 000 00 00000 ”00 00.0 00.0 00.0 80305000 0.508 0000:8300 0.00 20020 080 08000 0.0.0 00.0. 00.0 «mm 20008 250 03000 N00 03008 250 08000 00.0 ~00 00.0 000 03008 080 000000 mad-20 000 080 000002 03.. 00.0 :0- _ mad-20 00ImMBNIz0§mI00000 000 080 000002 00.0 00.0 00.0- 00ImMB~Iz0§mI00000 2000§Im<00200§§m 000 050 000002 3.0 00.... 00.0- 200000Im<03000§0xm0 000000002Izo_mm:00w.£0.000.&0\\x€0_ wofig 0000800 ”020000.253 - 888 00.0 002 as... 0000600 8800000 00.0 00.0 00.0- 8050000.: 0.80: 00I0E0m0mI>szMI0z000< 000 250 000002 00.0 :0 00.0- §I0E0000I>szMI0Em0< 00I020Immoz0-E0m0 000 250 000002 00.0 00.0 00.0. 00I02mI-00020-E000 mm 03005 080 08000 00.0 00.0 00.0- 00 03005 080 0850 0 000000-030 BmWME bmW—ME N00~Im> 0000 0000320350Em0§00w0m a moflmmumumnu 199 .Eng.mo_svofilxnlomzrthSogohgszB.3853.mouonetuntn ”mo—=88 28m .5230 ”\muoxwo—ofioonowighfitn ”came—9:0 0:09 00 EmEouwomivwfifihommEho.Baumfiuaofi.333\\uat: ”30m 250 mama—2 .mood 55 083% $3.9m m 03E mzoo wimmoaxo.8_v_m2-m> E 8.58 05 was .358598 08086:: 259532: 8:: 800m @0390 0003 85:83 a 200 SM 0386 25» 8080 3mm 30 N3 3 0388 8% 6050 $330 How :3. 8.0.- 3o :8 382on 3335 do :2. 8.0.- 3o Ba @3803 maua¢m0mu02 30m oaogmxfisfiafimoggwmm a moumtfimé Literature Cited 201 'C Literature Cited Abi-Habib RJ, Singh R, Leppla SH, Greene JJ, Ding Y, Berghuis B, Duesbery NS , Frankel AE (2006a). Systemic Anthrax Lethal Toxin Therapy Produces Regressions of Subcutaneous Human Melanoma Tumors in Athymic Nude Mice. Clin Cancer Res. 12, 7437-7443. Abi-Habib RJ, Singh R, Liu S, Bugge TH, Leppla SH , Frankel AE (2006b). A urokinase-activated recombinant anthrax toxin is selectively cytotoxic to many human tumor cell types. Mol Cancer Ther. 5, 2556-2562. Abi-Habib RJ, Urieto J 0, Liu S, Leppla SH, Duesbery NS , Frankel AE (2005). BRAF status and mitogen-activated protein/extracellular signal-regulated kinase kinase 1/2 activity indicate sensitivity of melanoma cells to anthrax lethal toxin. Mol Cancer Ther. 4, 1303-1310. Adjei AA, Cohen RB, Franklin W, Morris C, Wilson D, Molina JR, Hanson LJ, Gore L, Chow L, Leong S, Maloney L, Gordon G, Simmons H, Marlow A, Litwiler K, ,. Brown S, Poch G, Kane K, Haney J , Eckhardt SG (2008). Phase I H Pharmacokinetic and Pharrnacodynamic Study of the Oral, Small-Molecule Mitogen-Activated Protein Kinase Kinase 1/2 Inhibitor AZD6244 (ARRY- 142886) in Patients With Advanced Cancers. J Clin Oncol. 26, 2139-2146. Agrawal A, Lingappa J, Leppla SH, Agrawal S, Jabbar A, Quinn C , Pulendran B (2003). Impairment of dendritic cells and adaptive immunity by anthrax lethal toxin. Nature. 424, 329-334. Ahn NG, Seger R, Bratlien RL, Diltz CD, Tonks NK , Krebs EG (1991). Multiple components in an epidermal growth factor-stimulated protein kinase cascade. In vitro activation of a myelin basic protein/microtubule-associated protein 2 kinase. Journal of Biological Chemishy. 266, 4220—4227. Ahn NG, Stines Nahreini T, Tolwinski NS, Resing KA , W.E. Balch CJDaAH (2001). Pharmacologic Inhibitors of MK] and MKKZ. In Methods in Enzymology): Academic Press, pp. 417-431. Aitken J, Welch J, Duffy D, Milligan A, Green A, Martin N , Hayward N (1999). CDKNZA variants in a population-based sample of Queensland families with melanoma. J Natl Cancer Inst. 91, 446-452. Alessi DR, Cuenda A, Cohen P, Dudley DT , Saltiel AR (1995). PD 098059 Is a Specific Inhibitor of the Activation of Mitogen-activated Protein Kinase Kinase in Vitro and in Vivo. J. Biol. Chem. 270, 27489-27494. Alessi DR, Saito Y, Campbell DG, Cohen P, Sithanandam G, Rapp U, Ashworth A, Marshall CJ , Cowley S (1994). Identification of the sites in MAP kinase kinase- ] phosphorylated by p74raf-l. EMBO J. 13, 1610-1619. 202 Alfano RW, Leppla SH, Liu S, Bugge TH, Meininger CJ, Lairmore TC, Mulne AF, Davis SH, Duesbery NS , Frankel AE (2009). Matrix metalloproteinase—activated anthrax lethal toxin inhibits endothelial invasion and neovasculature formation during in vitro morphogenesis. Mol Cancer Res. 7, 452-461. Allen LF, Sebolt-Leopold J , Meyer MB (2003). CI-1040 (PD184352), a targeted signal transduction inhibitor of MEK (MAPKK). Seminars in Oncology. 30, 105-116. Altekruse SF, Kosary CL, Krapcho M, Neyman N, Aminou R, Waldron W, Ruhl J, Howlader N, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Cronin K, Chen HS, Feuer EJ, Stinchcomb DG , Edwards BK: SEER Cancer Statistics Review, 1975-2007, based on November 2009 SEER data submission, posted to the SEER web site, 2010. (Accessed June 19, 2010) American Cancer Society (2010). Cancer Facts & Figures 2010edAeds). Atlanta: American Cancer Society. Anderson NG, Maller J L, Tonks NK , Sturgill TW (1990). Requirement for integration of signals from two distinct phosphorylation pathways for activation of MAP kinase. Nature. 343, 651-653. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese J C, Richardson J E, Ringwald M, Rubin GM , Sherlock G (2000). Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25, 25-29. Astier AL, Xu R, Svoboda M, Hinds E, Munoz 0, de Beaumont R, Crean CD, Gabig T , Freedman AS (2003). Temporal gene expression profile of human precursor B leukemia cells induced by adhesion receptor: identification of pathways regulating B-cell survival. Blood. 101, 1118-1127. Balch CM, Gershenwald J E, Soong SJ, Thompson JF, Atkins MB, Byrd DR, Buzaid AC, Cochran AJ, Coit DG, Ding S, Eggermont AM, F laherty KT, Gimotty PA, Kirkwood JM, McMasters KM, Mihm MC, Jr., Morton DL, Ross MI, Sober AJ , Sondak VK (2009). Final version of 2009 AJ CC melanoma staging and classification. J Clin Oncol. 27, 6199-6206. Ball NJ, Yohn JJ, Morelli JG, Norris DA, Golitz LE , Hoefiler JP (1994). Ras mutations in human melanoma: a marker of malignant progression. J Invest Dermatol. 102, 285-290. Bardwell AJ, Abdollahi M , Bardwell L (2004). Anthrax lethal factor-cleavage products of MAPK (mitogen—activated protein kinase) kinases exhibit reduced binding to their cognate MAPKs. Biochem. J. 378, 569-577. 203 Belanger L-F, Roy S, Tremblay M, Brott B, Steff A-M, Mourad W, Hugo P, Erikson R , Charron J (2003). Mek2 Is Dispensable for Mouse Growth and Development. Mol. Cell. Biol. 23, 4778-4787. Bild AH, Yao G, Chang JT, Wang Q, Potti A, Chasse D, Joshi MB, Harpole D, Lancaster JM, Berchuck A, Olson JA, Jr., Marks JR, Dressman HK, West M , Nevins JR (2006). Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature. 439, 353-357. Birck A, Ahrenkiel V, Zeuthen J, Hon-Jensen K , Guldberg P (2000). Mutation and allelic loss of the PTEN/MMACI gene in primary and metastatic melanoma biopsies. J Invest Dermatol. 114, 277-280. Borg A, Johannsson U, Johannsson O, Hakansson S, Westerdahl J, Masback A, Olsson H , Ingvar C (1996). Novel gerrnline p16 mutation in familial malignant melanoma in southern Sweden. Cancer Res. 56, 2497-2500. Bosenberg M, Muthusamy V, Curley DP, Wang Z, Hobbs C, Nelson B, Nogueira C, Homer J W, 2nd, Depinho R , Chin L (2006). Characterization of melanocyte- specific inducible Cre recombinase transgenic mice. genesis. 44, 262-267. Boulton TG, Yancopoulos GD, Gregory J S, Slaughter C, Moomaw C, Hsu J , Cobb MH (1990). An insulin-stimulated protein kinase similar to yeast kinases involved in cell cycle control. Science. 249, 64-67. Bradley KA, Mogridge J, Mourez M, Collier RJ , Young JA (2001). Identification of the cellular receptor for anthrax toxin. Nature. 414, 225-229. Brai g M , Schmitt CA (2006). Oncogene-induced senescence: putting the brakes on tumor development. Cancer Res. 66, 2881 -2884. Bromberg-White J L, Boguslawski E , Duesbery NS (2009). Perturbation of Mouse Retinal Vascular Morphogenesis by Anthrax Lethal Toxin. PLoS ONE. 4, e6956. Bromberg-White J L , Duesbery NS (2008). Biological and biochemical characterization of anthrax lethal factor, a proteolytic inhibitor of MEK signaling pathways. Methods Enzymol. 438, 355-365. Cannon-Albright LA, Goldgar DE, Meyer LJ, Lewis CM, Anderson DE, Fountain JW, Hegi ME, Wiseman RW, Petty EM, Bale AE , et a1. (1992). Assignment of a locus for familial melanoma, MLM, to chromosome 9p13-p22. Science. 258, 1 148-1 152. 204 Catalanotti F, Reyes G, J esenberger V, Galabova-Kovacs G, de Matos Simoes R, Carugo O , Baccarini M (2009). A Mekl-Mek2 heterodimer determines the strength and duration of the Erk signal. Nat Struct Mol Biol. 16, 294-303. Chang HY, Sneddon J B, Alizadeh AA, Sood R, West RB, Montgomery K, Chi JT, van de Rijn M, Botstein D , Brown PO (2004). Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2, E7. Chen Z, Gibson TB, Robinson F, Silvestro L, Pearson G, Xu B-e, Wright A, Vanderbilt C , Cobb MH (2001). MAP Kinases. Chemical Reviews. 101, 2449-2476. Chin L, Pomerantz J, Polsky D, Jacobson M, Cohen C, Cordon-Cardo C, Homer JW, 2nd , DePinho RA (1997). Cooperative effects of IN K4a and ras in melanoma susceptibility in vivo. Genes Dev. 11, 2822-2834. Chin L, Tam A, Pomerantz J, Wong M, Holash J, Bardeesy N, Shen Q, O'Hagan R, Pantginis J, Zhou H, Homer JW, 2nd, Cordon-Cardo C, Yancopoulos GD , DePinho RA (1999). Essential role for oncogenic Ras in tumour maintenance. Nature. 400, 468-472. Chong H, Vikis HG , Guan KL (2003). Mechanisms of regulating the Raf kinase family. Cell Signal. 15, 463-469. Chopra AP, Boone SA, Liang X , Duesbery NS (2003). Anthrax Lethal Factor Proteolysis and Inactivation of MAPK Kinase. J. Biol. Chem. 278, 9402-9406. Clark WH, Jr., Elder DE, Guerry Dt, Epstein MN, Greene MH , Van Horn M (1984). A study of tumor progression: the precursor lesions of superficial spreading and nodular melanoma. Hum Pathol. 15, 1147-1165. ClinicalTrials.gov Identifier: NCT00147550 , US. National Institutes of Health. A service of the US. National Institutes of Health: ClinicalTrials. gov Identifier: NCT00147 550. MEK Inhibitor PD-325901 To Treat Advanced Breast Cancer, Colon Cancer, And Melanoma. http://clinicaltrials. gov/ct2/show/N CT00147 550 (Accessed August 12, 2009) Cohen C, Zavala-Pompa A, Sequeira JH, Shoji M, Sexton DG, Cotsonis G, Cerimele F, Govindarajan B, Macaron N , Arbiser J L (2002). Mitogen-actived Protein Kinase Activation Is an Early Event in Melanoma Progression. Clin Cancer Res. 8, 3728- 3733. Cohen P (1999). The development and therapeutic potential of protein kinase inhibitors. Current Opinion in Chemical Biology. 3, 459-465. 205 Collisson EA, De A, Suzuki H, Gambhir SS , Kolodney MS (2003). Treatment of Metastatic Melanoma with an Orally Available Inhibitor of the Ras-Raf-MAPK Cascade. Cancer Res. 63, 5669-5673. Cowan JM, Halaban R , Francke U (1988). Cytogenetic analysis of melanocytes from premalignant nevi and melanomas. J Natl Cancer Inst. 80, 1159-1164. Crews CM , Erikson RL (1992). Purification of a murine protein-tyrosine/threonine kinase that phosphorylates and activates the Erk-1 gene product: relationship to the fission yeast byrl gene product. Proc Natl Acad Sci U S A. 89, 8205-8209. Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky Jr WE, You MJ, DePinho RA, McMahon M , Bosenberg M (20093). BratV600E cooperates with Pten loss to induce metastatic melanoma. Nat Genet. 41, 544-552. Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE, Jr., You MJ, DePinho RA, McMahon M , Bosenberg M (2009b). Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat Genet. 41, 544-552. Dankort D, Filenova E, Collado M, Serrano M, Jones K , McMahon M (2007). A new mouse model to explore the initiation, progression, and therapy of BRAFV600E- induced lung tumors. Genes & Development. 21, 379-384. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosrnidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, J ayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-J ones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JWC, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR , Futreal PA (2002). Mutations of the BRAF gene in human cancer. Nature. 417, 949-954. Davies SP, Reddy H, Caivano M , Cohen P (2000). Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem. J. 351, 95-105. Dent P, Haser W, Haystead TA, Vincent LA, Roberts TM , Sturgill TW (1992). Activation of mitogen-activated protein kinase kinase by v-Raf in NIH 3T3 cells . and in vitro. Science. 257, 1404-1407. Depeille P, Young JJ, Boguslawski EA, Berghuis BD, Kort EJ, Resau J H, Frankel AE , Duesbery NS (2007). Anthrax Lethal Toxin Inhibits Growth of and Vascular Endothelial Growth Factor Release fi'om Endothelial Cells Expressing the Human Herpes Virus 8 Viral G Protein Coupled Receptor. Clin Cancer Res. 13, 5926- 5934. 206 Ding Y, Boguslawski EA, Berghuis BD, Young JJ, Zhang Z, Hardy K, Furge K, Kort E, Frankel AE, Hay RV, Resau J H , Duesbery NS (2008). Mitogen-activated protein kinase kinase signaling promotes growth and vascularization of fibrosarcoma. Mol Cancer Ther. 7, 648-658. Dudley DT, Pang L, Decker SJ, Bridges AJ , Saltiel AR (1995). A synthetic inhibitor of the mitogen-activated protein kinase cascade. Proceedings of the National Academy of Sciences of the United States of America. 92, 7686-7689. Duesbery NS, Resau J, Webb CP, Koochekpour S, Koo HM, Leppla SH , Vande Woude GF (2001). Suppression of ras-mediated transformation and inhibition of tumor growth and angiogenesis by anthrax lethal factor, a proteolytic inhibitor of multiple MEK pathways. Proceedings of the National Academy of Sciences of the United States of America. 98, 4089-4094. Duesbery NS, Webb CP, Leppla SH, Gordon VM, Klirnpel KR, Copeland TD, Ahn NG, Oskarsson MK, Fukasawa K, Paull KD , Vande Woude GF (1998). Proteolytic Inactivation of MAP-Kinase-Kinase by Anthrax Lethal Factor. Science. 280, 734- 737. Eisen T, Ahmad T, Flaherty KT, Gore M, Kaye S, Marais R, Gibbens I, Hackett S, James M, Schuchter LM, Nathanson KL, Xia C, Simantov R, Schwartz B, Poulin- Costello M, O'Dwyer PJ , Ratain MJ (2006). Sorafenib in advanced melanoma: a Phase II randomised discontinuation trial analysis. Br J Cancer. 95, 581-586. English J, Pearson G, Wilsbacher J, Swantek J, Karandikar M, Xu S , Cobb MH (1999). New Insights into the Control of MAP Kinase Pathways. Experimental Cell Research. 253, 255-270. Erickson AK, Ray LB , Sturgill TW (1990). Microtubule-associated protein 1A is the fibroblast HMW MAP undergoing mitogen-stimulated serine phosphorylation. Biochem Biophys Res Commun. 166, 827-832. Estrada Y, Dong J , Ossowski L (2009). Positive crosstalk between ERK and p38 in melanoma stimulates migration and in vivo proliferation. Pigment Cell Melanoma Res. 22, 66-76. Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, Van Dyk DE, Pitts WJ, Earl RA, Hobbs F, Copeland RA, Magolda RL, Scherle PA , Trzaskos J M (1998). Identification of a Novel Inhibitor of Mitogen-activated Protein Kinase Kinase. .1. Biol. Chem. 273, 18623-18632. Ferrando AA, Armstrong SA, Neuberg DS, Sallan SE, Silverrnan LB, Korsmeyer SJ , Look AT (2003). Gene expression signatures in MLL-rearranged T-lineage and B-precursor acute leukemias: dominance of HOX dysregulation. Blood. 102, 262- 268. 207 Fischrnann TO, Smith CK, Mayhood TW, Myers J E, Reichert P, Mannarino A, Carr D, Zhu H, Wong J, Yang R-S, Le HV , Madison VS (2009). Crystal Structures of MEK] Binary and Ternary Complexes with Nucleotides and Inhibitors. Biochemistry. 48, 2661-2674. FitzGerald MG, Harkin DP, Silva-Arrieta S, MacDonald DJ, Lucchina LC, Unsal H, O'Neill E, Koh J, Finkelstein DM, Isselbacher KJ, Sober AJ , Haber DA (1996). Prevalence of gerrn-line mutations in p16, p19ARF, and CDK4 in familial melanoma: analysis of a clinic-based population. Proc Natl Acad Sci U S A. 93, 8541-8545. Flores JF, Pollock PM, Walker GJ, Glendening JM, Lin AH, Palmer J M, Walters MK, Hayward NK , Fountain JW (1997). Analysis of the CDKN 2A, CDKN2B and CDK4 genes in 48 Australian melanoma kindreds. Oncogene. 15, 2999-3005. Fountain JW, Karayiorgou M, Emstoff MS, Kirkwood J M, Vlock DR, Titus-Ernstoff L, Bouchard B, Vijayasaradhi S, Houghton AN, Lahti J , et al. (1992). Homozygous deletions within human chromosome band 9p21 in melanoma. Proc Natl Acad Sci USA. 89, 10557-10561. Friday BB, Yu C, Dy GK, Smith PD, Wang L, Thibodeau SN , Adjei AA (2008). BRAF V600E Disrupts AZD6244-Induced Abrogation of Negative Feedback Pathways between Extracellular Signal-Regulated Kinase and Raf Proteins. Cancer Res. 68, 6145-6153. Friedlander AM (1998). Antitumor effects of anthrax lethal toxin. In Third International Conference on AnthraxedAeds). University of Plymouth, pp. 104. Furge KA, Chen J, Koeman J, Swiatek P, Dykema K, Lucin K, Kahnoski R, Yang XJ , Teh BT (2007a). Detection of DNA copy number changes and oncogenic signaling abnormalities from gene expression data reveals MYC activation in hi gh- grade papillary renal cell carcinoma. Cancer Res. 67, 3171-3176. Furge KA, Tan MH, Dykema K, Kort E, Stadler W, Yao X, Zhou M , Teh BT (2007b). Identification of deregulated oncogenic pathways in renal cell carcinoma: an integrated onco genomic approach based on gene expression profiling. Oncogene. 26, 1346-1350. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, Homik K, Hothorn T, Huber W, Iacus S, Irizarry R, Leisch F, Li C, Maechler M, Rossini AJ, Sawitzki G, Smith C, Smyth G, Tierney L, Yang JY , Zhang J (2004). Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80. 208 Gershenwald JE, Soong SJ , Balch CM (2010). 2010 TNM staging system for cutaneous melanoma...and beyond. Ann Surg Oncol. 17, 1475-1477. Ghosh P (2004). Process of Protein Transport by the Type III Secretion System. Microbial. Mal. Biol. Rev. 68, 771-795. Giroux S, Tremblay M, Bernard D, Cardin-Girard J F, Aubry S, Larouche L, Rousseau S, Huot J, Landry J, J eannotte L , Charron J (1999). Embryonic death of Mekl- deficient mice reveals a role for this kinase in angiogenesis in the labyrinthine region of the placenta. Current Biology. 9, 369. Gomez N , Cohen P (1991). Dissection of the protein kinase cascade by which nerve growth factor activates MAP kinases. Nature. 353, 170-173. Govindarajan B, Bai X, Cohen C, Zhong H, Kilroy S, Louis G, Moses M , Arbiser JL (2003). Malignant Transformation of Melanocytes to Melanoma by Constitutive Activation of Mitogen-activated Protein Kinase Kinase (MAPKK) Signaling. J. Biol. Chem. 278, 9790-9795. Gray-Schopfer V, Wellbrock C , Marais R (2007). Melanoma biology and new targeted therapy. Nature. 445, 851-857. Guldberg P, thor Straten P, Birck A, Ahrenkiel V, Kirkin AF , Zeuthen J (1997). Disruption of the MMACl/PTEN gene by deletion or mutation is a frequent event in malignant melanoma. Cancer Res. 57, 3660-3663. Haass NK, Sproesser K, Nguyen TK, Contractor R, Medina CA, Nathanson KL, Herlyn M , Smalley KSM (2008). The Mitogen-Activated Protein/Extracellular Signal- Regulated Kinase Kinase Inhibitor AZD6244 (ARRY-142886) Induces Growth Arrest in Melanoma Cells and Tumor Regression When Combined with Docetaxel. Clin Cancer Res. 14, 230-239. Hao Y-H, Wang Y, Burdette D, Mukherjee S, Keitany G, Goldsmith E , Orth K (2008). Structural requirements for Yersinia YopJ inhibition of MAP kinase pathways. PLoS ONE. 3, el375. Harland M, Meloni R, Gruis N, Pinney E, Brookes S, Spurr NK, Frischauf AM, Bataille V, Peters G, Cuzick J, Selby P, Bishop DT , Bishop JN (1997). Germline mutations of the CDKN2 gene in UK melanoma families. Hum Mol Genet. 6, 2061-2067. Hatzivassiliou G, Song K, Yen I, Brandhuber B], Anderson DJ, Alvarado R, Ludlam MJ, Stokoe D, Gloor SL, Vigers G, Morales T, Aliagas 1, Lin B, Sideris S, Hoeflich KP, Jaiswal BS, Seshagiri S, Koeppen H, Belvin M, Friedman LS , Malek S (2010). RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 464, 431-435. 209 Heidom SJ, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, Hussain J, Reis-Filho J S, Springer CJ, Pritchard C , Marais R (2010). Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell. 140, 209-221. Herbst RA, Weiss J, Ehnis A, Cavenee WK , Arden KC (1994). Loss of heterozygosity for 10q22-10qter in malignant melanoma progression. Cancer Res. 54, 31 11-3114. Huang CY , Ferrell J E, Jr. (1996). Ultrasensitivity in the mitogen-activated protein kinase cascade. Proc Natl Acad Sci U S A. 93, 10078-10083. Huang D, Ding Y, Luo W-M, Bender S, Qian C-N, Kort B, Zhang Z-F, VandenBeldt K, Duesbery NS, Resau JH , Teh BT (2008). Inhibition of MAPK Kinase Signaling Pathways Suppressed Renal Cell Carcinoma Growth and Angiogenesis In vivo. Cancer Res. 68, 81 -88. Hussussian CJ, Struewing JP, Goldstein AM, Higgins PA, Ally DS, Sheahan MI), Clark WH, Jr., Tucker MA , Dracopoli NC (1994). Germline p16 mutations in familial melanoma. Nat Genet. 8, 15-21. Hwang PH, Yi HK, Kim DS, Nam SY, Kim JS , Lee DY (2001). Suppression of tumorigenicity and metastasis in B16F 10 cells by PTEN/MMACl/TEPI gene. Cancer Lett. 172, 83-91. Johnson GL , Lapadat R (2002). Mitogen-Activated Protein Kinase Pathways Mediated by ERK, JNK, and p38 Protein Kinases. Science. 298, 1911-1912. Kamakura S, Moriguchi T , Nishida E (1999). Activation of the Protein Kinase ERKS/BMKI by Receptor Tyrosine Kinases. IDENTIFICATION AND CHARACTERIZATION OF A SIGNALING PATHWAY TO THE NUCLEUS. J. Biol. Chem. 274, 26563-26571. Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavtigian SV, Stockert E, Day RS, 3rd, Johnson BE , Skolnick MH (1994). A cell cycle regulator potentially involved in genesis of many tumor types. Science. 264, 436-440. Kanehisa M, Araki M, Goto S, Hattori M, Hirakawa M, Itoh M, Katayama T, Kawashima S, Okuda S, Tokimatsu T , Yamanishi Y (2008). KEGG for linking genomes to life and the environment. Nucleic Acids Res. 36, D480-484. Kannan K, Sharpless NE, Xu J, O'Hagan RC, Bosenberg M , Chin L (2003). Components of the Rb pathway are critical targets of UV mutagenesis in a murine melanoma model. Proc Natl Acad Sci U S A. 100, 1221-1225. 210 Kelsall SR , Mintz B (1998). Metastatic cutaneous melanoma promoted by ultraviolet radiation in mice with transgene-initiated low melanoma susceptibility. Cancer Res. 58, 4061-4065. Kirby J E (2004). Anthrax Lethal Toxin Induces Human Endothelial Cell Apoptosis. Infect. Immun. 72, 430-439. Knudson AG, Jr. (1971). Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A. 68, 820-823. Kohno M , Pouyssegur J (2003). Pharmacological inhibitors of the ERK signaling pathway: application as anticancer drugs. Prog Cell Cycle Res. 5, 219-224. Kohno M , Pouyssegur J (2006). Targeting the ERK signaling pathway in cancer therapy. Ann Med. 38, 200-211. Koo H-M, VanBrocklin M, McWilliams MJ, Leppla SH, Duesbery NS , Woude GFV (2002). Apoptosis and melanogenesis in human melanoma cells induced by anthrax lethal factor inactivation of mitogen-activated protein kinase kinase. Proceedings of the National Academy of Sciences of the United States of America. 99, 3052-3057. Kyriakis JM, App H, Zhang XF, Banerjee P, Brautigan DL, Rapp UR , Avruch J (1992). Raf-1 activates MAP kinase-kinase. Nature. 358, 417-421. Lamb J (2007). The Connectivity Map: a new tool for biomedical research. Nat Rev Cancer. 7, 54-60. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M, Hieronymus H, Wei G, Armstrong SA, Haggarty SJ, Clemons PA, Wei R, Carr SA, Lander ES , Golub TR (2006). The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 313, 1929-1935. Lee CS , Duesbery NS (2010). Highly Selective MEK inhibitors. Current Enzyme Inhibition (in press). Lee P, Wallace E, Yeh T, Poch G, Litwiler K, Pheneger T, Lyssikatos J , Winkler J (2004). ARRY-142886, a potent and selective MEK inhibitor: III) Efficacy in murine xenograft models correlates with decreased ERK phosphorylation. In Annual Meeting of the American Association for Cancer ResearchedAeds), pp. 897. Levine AJ (2009). The common mechanisms of transformation by the small DNA tumor viruses: The inactivation of tumor suppressor gene products: p53. Virology. 384, 285-293. 211 P Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittrnann M, Tycko B, Hibshoosh H, Wigler MH , Parsons R (1997). PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 275, 1943- 1947. Limon J, Dal Cin P, Sait SN, Karakousis C , Sandberg AA (1988). Chromosome changes in metastatic human melanoma. Cancer Genet Cytogenet. 30, 201-211. Liu S, Wang H, Currie BM, Molinolo A, Leung HJ, Moayeri M, Basile JR, Alfano RW, Gutkind J S, Frankel AE, Bugge TH , Leppla SH (2008). Matrix Metalloproteinase—activated Anthrax Lethal Toxin Demonstrates High Potency in Targeting Tumor Vasculature. J. Biol. Chem. 283, 529-540. LoRusso P, Krishnarnurthi S, Rinehart JR, Nabell L, Croghan G, Varterasian M, Sadis SS, Menon SS, Leopold J , Meyer MB (2005a). A phase 1-2 clinical study of a second generation oral MEK inhibitor, PD 0325901 in patients with advanced cancer. In American Society of Clinical Oncology Annual MeetingedAeds), pp. 301 1. LoRusso PM, Adjei AA, Varterasian M, Gadgeel S, Reid J, Mitchell DY, Hanson L, DeLuca P, Bruzek L, Piens J, Asbury P, Van Becelaere K, Herrera R, Sebolt- Leopold J , Meyer MB (2005b). Phase I and Pharmacodynamic Study of the Oral MEK Inhibitor CI-1040 in Patients With Advanced Malignancies. J Clin Oncol. 23, 5281-5293. Lowe SW , Sherr CJ (2003). Tumor suppression by Ink4a-Arf: progress and puzzles. Curr Opin Genet Dev. 13, 77-83. Lynch HT, Fusaro RM, Danes BS, Kimberling WJ , Lynch JF (1983). A review of hereditary malignant melanoma including biomarkers in familial atypical multiple mole melanoma syndrome. Cancer Genet Cytogenet. 8, 325-358. Lyssikatos J, Yeh T, Wallace E, Marsh V, Bemat B, Gross S, Evans R, Colwell H, Parry J, Baker S, Ballard J, Morales T, Smith D, Brandhuber B , Winkler] (2004). ARRY-l42886, a potent and selective MEK inhibitor: I) ATP-independent inhibition results in high enzymatic and cellular selectivity. In Annual Meeting of the American Association for Cancer ResearchedAeds), pp. 896-b. MacKeigan JP, Collins TS , Ting JP (2000). MEK inhibition enhances paclitaxel- induced tumor apoptosis. J Biol Chem. 275, 38953-38956. Mansour SJ, Matten WT, Hermann AS, Candia J M, Rong S, Fukasawa K, Vande Woude GF , Ahn NG (1994a). Transformation of mammalian cells by constitutively active MAP kinase kinase. Science. 265, 966-970. 212 Mansour SJ, Resing KA, Candi J M, Hermann AS, Gloor JW, Herskind KR, Wartmann M, Davis RJ , Ahn NG (1994b). Mitogen-activated protein (MAP) kinase phosphorylation of MAP kinase kinase: determination of phosphorylation sites by mass spectrometry and site-directed mutagenesis. J Biochem. 116, 304-314. Mao L, Merlo A, Bedi G, Shapiro GI, Edwards CD, Rollins BJ , Sidransky D (1995). A novel p16INK4A transcript. Cancer Res. 55, 2995-2997. Mavria G, Vercoulen Y, Yeo M, Paterson H, Karasarides M, Marais R, Bird D , Marshall CJ (2006). ERK-MAPK signaling opposes Rho-kinase to promote endothelial cell survival and sprouting during angiogenesis. Cancer Cell. 9, 33-44. Menon SS, Whitfield LR, Sadis S, Meyer MB, Leopold J, Lorusso PM, Krishnamurthi S, Rinehart JR, Nabell L , Croghan G (2005). Pharmacokinetics (PK) and pharmacodynamics (PD) of PD 0325901, a second generation MEK inhibitor after multiple oral doses of PD 0325901 to advanced cancer patients. In American Society of Clinical Oncology Annual MeetingedAeds), pp. 3066. Michaloglou C, Vredeveld LC, Soengas MS, Denoyelle C, Kuilrnan T, van der Horst CM, Majoor DM, Shay JW, Mooi WJ , Peeper DS (2005). BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature. 436, 720-724. Miller AJ , Mihm MC, Jr. (2006). Melanoma. N Engl J Med. 355, 51-65. Mittal R, Peak-Chew S-Y , McMahon HT (2006). Acetylation of MEK2 and IicB kinase (IKK) activation loop residues by YopJ inhibits signaling. Proceedings of the National Academy of Sciences. 103, 18574-18579. Mody N, Leitch J, Armstrong C, Dixon J , Cohen P (2001). Effects of MAP kinase cascade inhibitors on the MKKS/ERKS pathway. FEBS letters. 502, 21-24. Mukherjee S, Keitany G, Li Y, Wang Y, Ball HL, Goldsmith EJ , Orth K (2006). Yersinia YopJ Acetylates and Inhibits Kinase Activation by Blocking Phosphorylation. Science. 312, 1211-1214. Nakielny S, Cohen P, Wu J , Sturgill T (1992). MAP kinase activator from insulin- stimulated skeletal muscle is a protein threonine/tyrosine kinase. EMBO J. 11, 2123-2129. Naumov GN, Akslen LA , Folkrnan J (2006). Role of angiogenesis in human tumor dormancy: animal models of the angiogenic switch. Cell Cycle. 5, 1779-1787. Nobori T, Miura K, Wu DJ, Lois A, Takabayashi K , Carson DA (1994). Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature. 368, 753-756. 213 Ohren ] F, Chen H, Pavlovsky A, Whitehead C, Zhang E, Kuffa P, Yan C, McConnell P, Spessard C, Banotai C, Mueller WT, Delaney A, Omer C, Sebolt-Leopold J, Dudley DT, Leung IK, Flamme C, Warmus J, Kaufman M, Barrett S, Tecle H , Hasemann CA (2004). Structures of human MAP kinase kinase 1 (MEKl) and MEK2 describe novel noncompetitive kinase inhibition. Nat Struct Mol Biol. 11, 1192-1197. Orth K, Palmer LE, Bao ZQ, Stewart S, Rudolph AE, Bliska JB , Dixon JE (1999). Inhibition of the Mitogen-Activated Protein Kinase Kinase Superfamily by a Yersinia Effector. Science. 285, 1920-1923. Orth K, Xu Z, Mudgett MB, Bao ZQ, Palmer LE, Bliska JB, Mange] WF, Staskawicz B , Dixon J E (2000). Disruption of Signaling by Yersinia Effector YopJ, a Ubiquitin- Like Protein Protease. Science. 290, 1594-1597. Palmer LE, Hobbie S, Galan J E , Bliska JB (1998). YopJ of Yersinia pseudotuberculosis is required for the inhibition of macrophage TNF-alpha production and downregulation of the MAP kinases p38 and JNK. Mal Microbial. 27, 953-965. Palmer LE, Pancetti AR, Greenberg S , Bliska J B (1999). YopJ of Yersinia spp. Is Sufficient To Cause Downregulation of Multiple Mitogen-Activated Protein Kinases in Eukaryotic Cells. Infect. Immun. 67, 708-716. Pannifer AD, Wong TY, Schwarzenbacher R, Renatus M, Petosa C, Bienkowska J, Lacy DB, Collier RJ, Park S, Leppla SH, Hanna P , Liddington RC (2001). Crystal structure of the anthrax lethal factor. Nature. 414, 229-233. Park J M, Greten FR, Li Z-W , Karin M (2002). Macrophage Apoptosis by Anthrax Lethal Factor Through p38 MAP Kinase Inhibition. Science. 297, 2048-2051. Parmiter AH, Balaban G, Clark WH, Jr. , Nowell PC (1988). Possible involvement of the chromosome region 10q24----q26 in early stages of melanocytic neoplasia. Cancer Genet Cytogenet. 30, 313-317. Patterson TA, Lobenhofer EK, Fulmer-Smentek SB, Collins PJ, Chu TM, Bao W, Fang H, Kawasaki ES, Hager J, Tikhonova IR, Walker SJ, Zhang L, Hurban P, de Longueville F, Fuscoe J C, Tong W, Shi L , Wolfinger RD (2006). Performance comparison of one-color and two-color platforms within the MicroArray Quality Control (MAQC) project. Nat Biotechnol. 24, 1 140-1150. Payne DM, Rossomando AJ, Martino P, Erickson AK, Her J H, Shabanowitz J, Hunt DF, Weber M] , Sturgill TW (1991). Identification of the regulatory phosphorylation sites in pp42/mitogen-activated protein kinase (MAP kinase). EMBO J. 10, 885- 892. 214 Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berrnan K , Cobb MH (2001). Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 22, 153-183. Pedersen MI, Bennett JW , Wang N (1986). Nonrandom chromosome structural aberrations and oncogene loci in human malignant melanoma. Cancer Genet Cytogenet. 20, 11-27. Pellizzari R, Guidi-Rontani C, Vitale G, Mock M , Montecucco C (1999). Anthrax lethal factor cleaves MKK3 in macmphages and inhibits the LPS/IFNgamma-induced release of NO and TNFalpha. FEBS Lett. 462, 199-204. Peng T, Golub TR , Sabatini DM (2002). The immunosuppressant rapamycin mimics a starvation-like signal distinct from amino acid and glucose deprivation. Mol Cell Biol. 22, 5575-5584. Petty EM, Gibson LH, Fountain JW, Bolognia J L, Yang-Peng TL, Housman DE , Bale AE (1993). Molecular definition of a chromosome 9p21 germ-line deletion in a woman with multiple melanomas and a plexiform neurofibroma: implications for 9p turnor-suppressor gene(s). Am J Hum Genet. 53, 96-104. Platz A, Hansson J, Mansson-Brahme E, Lagerlof B, Linder S, Lundqvist E, Sevigny P, Inganas M , Ringborg U (1997). Screening of gerrnline mutations in the CDKN2A and CDKN2B genes in Swedish families with hereditary cutaneous melanoma. J Natl Cancer Inst. 89, 697-702. Polager S , Ginsberg D (2009). p53 and E2f: partners in life and death. Nat Rev Cancer. 9, 73 8-748. Pollock PM, Harper UL, Hansen KS, Yudt LM, Stark M, Robbins CM, Moses TY, Hostetter G, Wagner U, Kakareka J, Salem G, Pohida T, Heenan P, Duray P, Kallioniemi O, Hayward NK, Trent J M , Meltzer PS (2003). High frequency of BRAF mutations in nevi. Nat Genet. 33, 19-20. Poulikakos PI, Zhang C, Bollag G, Shokat KM , Rosen N (2010). RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 464, 427-430. Powell MB, Gause PR, Hyman P, Gregus J, Lluria-Prevatt M, Nagle R , Bowden GT (1999). Induction of melanoma in TPras transgenic mice. Carcinogenesis. 20, 1 7 47 -1 753. Quelle DE, Zindy F, Ashmun RA , Sherr CJ (1995). Alternative reading frames of the INK4a tumor suppressor gene encode two unrelated proteins capable of inducing cell cycle arrest. Cell. 83, 993-1000. 215 Rapp UR, Goldsborough MD, Mark GE, Bonner TI, Groffen J, Reynolds FH, Jr. , Stephenson JR (1983). Structure and biological activity of v-raf, a unique ' oncogene transduced by a retrovirus. Proc Natl Acad Sci U S A. 80, 4218-4222. Ray LB , Sturgill TW (1987). Rapid stimulation by insulin of a serine/threonine kinase in 3T3-L1 adipocytes that phosphorylates microtubule-associated protein 2 in vitro. Proc Natl Acad Sci U S A. 84, 1502-1506. Ray LB , Sturgill TW (1988a). Characterization of insulin-stimulated microtubule- associated protein kinase. Rapid isolation and stabilization of a novel serine/threonine kinase from 3T3-L1 cells. J Biol Chem. 263, 12721-12727. Ray LB , Sturgill TW (1988b). Insulin-stimulated microtubule-associated protein kinase is phosphorylated on tyrosine and threonine in vivo. Proc Natl Acad Sci U S A. 85, 3753-3757. Rinehart J, Adjei AA, LoRusso PM, Waterhouse D, Hecht JR, Natale RB, Hamid O, Varterasian M, Asbury P, Kaldjian EP, Gulyas S, Mitchell DY, Herrera R, Sebolt- Leopold J S , Meyer MB (2004). Multicenter Phase II Study of the Oral MEK Inhibitor, CI-1040, in Patients With Advanced Non-Small-Cell Lung, Breast, Colon, and Pancreatic Cancer. J Clin Oncol. 22, 4456-4462. Roberts PJ , Der CJ (2007). Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26, 3291-3310. Robertson GP, Furnari FB, Miele ME, Glendening MJ, Welch DR, Fountain JW, Lugo TG, Huang HJ , Cavenee WK (1998). In vitro loss of heterozygosity targets the PTEN/MMACl gene in melanoma. Proc Natl Acad Sci U S A. 95, 9418-9423. Rossomando AJ, Payne DM, Weber MJ , Sturgill TW (1989). Evidence that pp42, a major tyrosine kinase target protein, is a mitogen—activated serine/threonine protein kinase. Proc Natl Acad Sci U S A. 86, 6940-6943. Rouleau C, Menon K, Boutin P, Guyre C, Yoshida H, Kataoka S, Perricone M, Shankara S, Frankel AE, Duesbery NS, Vande Woude G, Biemann HP , Teicher BA (2008). The systemic administration of lethal toxin achieves a growth delay of human melanoma and neuroblastoma xenografts: assessment of receptor contribution. IntJ Oncol. 32, 739-748. Roux PP , Blenis J (2004). ERK and p38 MAPK-Activated Protein Kinases: a Family of Protein Kinases with Diverse Biological Functions. Microbial. Mal. Biol. Rev. 68, 320-344. Ruckdeschel K, Harb S, Roggenkarnp A, Hornef M, Zumbihl R, Kohler S, Heesemann J , Rouot B (1998). Yersinia enterocolitica Irnpairs Activation of Transcription Factor NF-kappa B: Involvement in the Induction of Programmed Cell Death and 216 in the Suppression of the Macrophage Tumor Necrosis Factor alpha roduction. J. Exp. Med. 187, 1069-1079. Sagata N, Oskarsson M, Copeland T, Brumbaugh J , Vande Woude GF (1988). Function of c-mos proto-onco gene product in meiotic maturation in Xenopus oocytes. Nature. 335, 519-525. Sato C, Nishizawa K, Nakayama T , Kobayashi T (1985). Effect upon mitogenic stimulation of calcium-dependent phosphorylation of cytoskeleton—associated 350,000- and 80,000-mol-wt polypeptides in quiescent 3Y1 cells. J. Cell Biol. 100, 748-753. Sato C, Nishizawa K, Nakayama T, Nose K, Takasaki Y, Hirose S , Nakamura H (1986). Intranuclear appearance of the phosphorylated form of cytoskeleton-associated 350-kDa proteins in Ul-ribonucleoprotein regions after growth stimulation of fibroblasts. Proc Natl Acad Sci U S A. 83, 7287-7291. Schechter I , Berger A (1967). On the size of the active site in proteases. I. Papain. Biochem Biophys Res Commun. 27, 157-162. _ Schesser K, Spiik A-K, Dulcuzumuremyi J -M, Neurath MF, Pettersson S , Wolf-Watz H (1998). The yopJ locus is required for Yersinia-mediated inhibition of NF-kappaB activation and cytokine expression: YopJ contains a eukaryotic SH2-like domain that is essential for its repressive activity. Molecular Microbiology. 28, 1067-1079. Scholl FA, Dumesic PA, Barragan DI, Charron J , Khavari PA (2009a). Mekl/2 gene dosage determines tissue response to oncogenic Ras signaling in the skin. Oncogene. 28, 1485-1495. Scholl FA, Dumesic PA, Barragan DI, Harada K, Charron J , Khavari PA (2009b). Selective Role for Mekl but not Mek2 in the Induction of Epidermal Neoplasia. Cancer Res. 69, 3772-3778. Scobie HM, Rainey GJ, Bradley KA , Young JA (2003). Human capillary morphogenesis protein 2 functions as an anthrax toxin receptor. Proc Natl Acad Sci USA. 100, 5170-5174. Sebolt-Leopold J S, Dudley DT, Herrera R, Becelaere KV, Wiland A, Gowan RC, Tecle H, Barrett SD, Bridges A, Przybranowski S, Leopold WR , Saltiel AR (1999). Blockade of the MAP kinase pathway suppresses growth of colon tumors in vivo. Nat Med. 5, 810-816. Sebolt-Leopold J S , Herrera R (2004). Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 4, 937-947. 217 Sebolt-Leopold J S, Merriman R, Omer C, Tecle H, Bridges A, Klohs W, Loi C-M, Valik H, Przybranowski S, Meyer M , Leopold WR (2004). The biological profile of PD 0325901: A second generation analog of CI-1040 with improved pharmaceutical potential. In Annual Meeting of the American Association for Cancer ResearchedAeds), pp. 925. Segal E, Friedman N, Koller D , Regev A (2004). A module map showing conditional activity of expression modules in cancer. Nat Genet. 36, 1090-1098. Seger R, Ahn NG, Posada J, Munar ES, Jensen AM, Cooper JA, Cobb MH , Krebs EG (1992). Purification and characterization of mitogen-activated protein kinase activator(s) from epidermal growth factor-stimulated A431 cells. Journal of Biological Chemistry. 267, 14373-14381. Serrano M, Hannon GJ , Beach D (1993). A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature. 366, 704-707. Singh Y, Liang X , Duesbery NS (2005). Pathogenesis of Bacillus anthracis: the role of Anthrax toxins. In Microbial Toxins. Molecular and Cellular Biology. (T Profi, ed). Norfolk, England: Horizon Bioscience, pp. 285-312. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A, Ye Q, Lobo JM, She Y, Osman I, Golub TR, Sebolt-Leopold J, Sellers WR , Rosen N (2006). BRAF mutation predicts sensitivity to MEK inhibition. Nature. 439, 358. Soufir N, Avril MF, Chompret A, Demenais F, Bombled J, Spatz A, Stoppa-Lyonnet D, Benard J , Bressac-de Paillerets B (1998). Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in France. The French Familial Melanoma Study Group. Hum Mol Genet. 7, 209-216. St Croix B, Rago C, Velculescu V, Traverso G, Romans KE, Montgomery E, Lal A, Riggins GJ, Lengauer C, Vogelstein B , Kinzler KW (2000). Genes expressed in human tumor endothelium. Science. 289, 1197-1202. Stahl J M, Cheung M, Sharma A, Trivedi NR, Shanmugam S , Robertson GP (2003). Loss of PTEN promotes tumor development in malignant melanoma. Cancer Res. 63, 2881-2890. Steck PA, Pershouse MA, J asser SA, Yung WK, Lin H, Ligon AH, Langford LA, Baumgard ML, Hattier T, Davis T, Frye C, Hu R, Swedlund B, Teng DH , Tavtigian SV (1997). Identification of a candidate tumour suppressor gene, MMACI , at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 15, 356-362. 218 Stone S, Jiang P, Dayananth P, Tavtigian SV, Katcher H, Parry D, Peters G , Kamb A (1995). Complex structure and regulation of the P16 (MTSI) locus. Cancer Res. 55, 2988-2994. Sturgill TW , Ray LB (1986). Muscle proteins related to microtubule associated protein- 2 are substrates for an insulin-stimulatable kinase. Biochem Biophys Res Commun. 134, 565-571. Tanami H, Irnoto I, Hirasawa A, Yuki Y, Sonoda I, Inoue J, Yasui K, Misawa-Furihata A, Kawakami Y , Inazawa J (2004). Involvement of overexpressed wild-type BRAF in the growth of malignant melanoma cell lines. Oncogene. 23, 8796. Tanoue T, Adachi M, Moriguchi T , Nishida E (2000). A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat Cell Biol. 2, 110-116. Tarhini AA , Agarwala SS (2006). Cutaneous melanoma: available therapy for metastatic disease. Dermatol Ther. 19, 19-25. Teng DH, Hu R, Lin H, Davis T, Iliev D, Frye C, Swedlund B, Hansen KL, Vinson VL, Gumpper KL, Ellis L, El-Naggar A, Frazier M, J asser S, Langford LA, Lee J, Mills GB, Pershouse MA, Pollack RE, Tomos C, Troncoso P, Yung WK, Fujii G, Berson A, Steck PA , et al. (1997). MMACl/PTEN mutations in primary tumor specimens and tumor cell lines. Cancer Res. 57, 5221-5225. Tian B, Nowak DE, J amaluddin M, Wang S , Brasier AR (2005). Identification of direct genomic targets downstream of the nuclear factor-kappaB transcription factor mediating tumor necrosis factor signaling. J Biol Chem. 280, 17435-17448. Trisciuoglio D, Iervolino A, Zupi G , Del Bufalo D (2005). Involvement of PI3K and MAPK Signaling in bcl-2-induced Vascular Endothelial Growth Factor Expression in Melanoma Cells. Mol. Biol. Cell. 16, 4153-4162. Tsao H, Zhang X, Benoit E , Haluska PG (1998). Identification of PTEN/MMACI alterations in uncultured melanomas and melanoma cell lines. Oncogene. 16, 3397-3402. Tsao H, Zhang X, Fowlkes K , Haluska F G (2000). Relative Reciprocity of NRAS and PTEN/MMACI Alterations in Cutaneous Melanoma Cell Lines. Cancer Res. 60, 1 800-1 804. van 't Veer LJ, Burgering BM, Versteeg R, Boot AJ, Ruiter DJ, Osanto S, Schrier PI , Bos J L (1989). N-ras mutations in human cutaneous melanoma from sun-exposed body sites. Mol Cell Biol. 9, 3114-3116. 219 Vitale G, Bemardi L, Napolitani G, Mock M , Montecucco C (2000). Susceptibility of mitogen-activated protein kinase kinase family members to proteolysis by anthrax lethal factor. Biochem. J. 352, 739-745. Vitale G, Pellizzari R, Recchi C, Napolitani G, Mock M , Montecucco C (1998). Anthrax Lethal Factor Cleaves the N-Terrninus of MAPKKs and Induces Tyrosine/Threonine Phosphorylation of MAPKs in Cultured Macrophages. Biochemical and Biophysical Research Communications. 248, 706-711. Voisin L, Julien C, Duharnel S, Gopalbhai K, Claveau I, Saba-El-Leil M, Rodrigue- Gervais I, Gaboury L, Lamarre D, Basik M , Meloche S (2008). Activation of MEKI or MEK2 isoform is sufficient to fully transform intestinal epithelial cells and induce the formation of metastatic tumors. BMC Cancer. 8, 337. Wajapeyee N, Serra RW, Zhu X, Mahalingam M , Green MR (2008). Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell. 132, 363-374. Walker GJ, Hussussian CJ, Flores JF, Glendening JM, Haluska FG, Dracopoli NC, Hayward NK , Fountain JW (1995). Mutations of the CDKN2/p16INK4 gene in Australian melanoma kindreds. Hum Mol Genet. 4, 1845-1852. Wallace E, Yeh T, Lyssikatos J, Winkler J, Lee P, Marlow A, Hurley B, Marsh V, Bemat B, Evans R, Colwell H, Ballard J, Morales T, Smith D, Brandhuber B, Gross S, Poch G, Litwiler K, Hingorani G, Otten J, Sullivan F, Blake J, Pheneger T, Goyette M , Koch K (2004). Preclinical development of ARRY-142886, a potent and selective MEK inhibitor. In Annual Meeting of the American Association for Cancer ResearchedAeds), pp. 897-a. Warfel JM, Steele AD , D'Agnillo F (2005). Anthrax Lethal Toxin Induces Endothelial Barrier Dysfimction. Am J Pathol. 166, 1871-1881. Warmus J S, Flamme C, Zhang LY, Barrett S, Bridges A, Chen H, Gowan R, Kaufman M, Sebolt-Leopold J, Leopold W, Merriman R, Ohren J, Pavlovsky A, Przybranowski S, Tecle H, Valik H, Whitehead C , Zhang B (2008). 2- Alkylamino- and alkoxy-substituted 2-amino-1,3,4-oxadiazoles--O-Alkyl benzohydroxamate esters replacements retain the desired inhibition and selectivity against MEK (MAP ERK kinase). Bioarganic & Medicinal Chemistry Letters. 18, 6171-6174. Wellbrock C, Ogilvie L, Hedley D, Karasarides M, Martin J, Niculescu-Duvaz D, Springer CJ , Marais R (2004). V599EB-RAF is an Oncogene in Melanocytes. Cancer Res. 64, 2338-2342. 220 Whitwarn T, Vanbrocklin MW, Russo ME, Haak PT, Bilgili D, Resau JH, Koo HM , Holmen SL (2007). Differential oncogenic potential of activated RAS isoforms in melanocytes. Oncogene. 26, 4563-4570. ' Wu H, Goel V , Haluska FG (2003). PTEN signaling pathways in melanoma. Oncogene. 22, 3113-3122. Xia Z, Dickens M, Raingeaud J, Davis RJ , Greenberg ME (1995). Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science. 270, 1326-1331. Yeh T, Wallace E, Lyssikatos J , Winkler J (2004). ARRY-142886, a potent and selective MEK inhibitor: II) Potency against cellular MEK leads to inhibition of cellular proliferation and induction of apoptosis in cell lines with mutant Ras or B-Raf. In Annual Meeting of the American Association for Cancer ResearchedAeds), pp. 896-c-897. Yeh TC, Marsh V, Bemat BA, Ballard J, Colwell H, Evans RJ, Parry J, Smith D, Brandhuber BJ, Gross S, Marlow A, Hurley B, Lyssikatos J, Lee PA, Winkler JD, Koch K , Wallace E (2007). Biological Characterization of ARRY-142886 (AZD6244), a Potent, Highly Selective Mitogen-Activated Protein Kinase Kinase 1/2 Inhibitor. Clin Cancer Res. 13, 1576-1583. You MJ, Castrillon DH, Bastian BC, O'Hagan RC, Bosenberg MW, Parsons R, Chin L , DePinho RA (2002). Genetic analysis of Pten and Ink4a/Arf interactions in the suppression of tumorigenesis in mice. Proc Natl Acad Sci U S A. 99, 1455-1460. Zheng CF , Guan KL (1994). Activation of MEK family kinases requires phosphorylation of two conserved Ser/Thr residues. EMBO J. 13, 1 123-1131. Zhou XP, Girnm O, Hampel H, Niemann T, Walker MJ , Eng C (2000). Epigenetic PTEN silencing in malignant melanomas without PTEN mutation. Am J Pathol. 157, 1123-1128. 221 irritant 030 3 1293