Oncogene (2010) 29, 1249–1259 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc REVIEW as therapeutic targets

WC Gustafson1,2 and WA Weiss1,3,4

1Department of Pediatrics, University of California, San Francisco, CA, USA; 2Division of Pediatric Hematology/Oncology, University of California, San Francisco, CA, USA; 3Department of Neurology, University of California, San Francisco, CA, USA and 4Department of Neurosurgery, University of California, San Francisco, CA, USA

Myc proteins (c-myc, Mycn and Mycl) target prolifera- tumors and is the best characterized genetic-risk factor tive and apoptotic pathways vital for progression in for high-risk chemotherapy-refractory disease (Brodeur cancer. Amplification of the MYCN has emerged et al., 1984; Seeger et al., 1985; Riley et al., 2004). as one of the clearest indicators of aggressive and Deletion or suppression of caspase 8, loss of chromo- chemotherapy-refractory disease in children with neuro- somes 1p and 11q, and gain of 17q also correlate with blastoma, the most common extracranial solid tumor of aggressive disease (Bown et al., 1999; Guo et al., 1999; childhood. Phosphorylation and ubiquitin-mediated mod- Riley et al., 2004; Attiyeh et al., 2005; Stupack et al., ulation of Myc influence stability and represent 2006; Maris et al., 2008b). potential targets for therapeutic intervention. Phosphor- In contrast with most other treatment-refractory ylation of Myc proteins is controlled in-part by the cancers, neuroblastomas, irrespective of risk group, tyrosine kinase/phosphatidylinositol 3-kinase/ generally respond to initial therapy, which typically Akt/mTOR signaling, with additional contributions from includes high doses of chemotherapy (Park et al., 2008). Aurora A kinase. Myc proteins regulate apoptosis in part Low- and intermediate-risk patients are subsequently through interactions with the /Mdm2/Arf signaling cured and rarely progress. Patients with high-risk pathway. Mutation in p53 is commonly observed in disease typically relapse with treatment-refractory tu- patients with relapsed neuroblastoma, contributing to mors. It is conceivable that low- and high-risk neuro- both biology and therapeutic resistance. This review blastoma are entirely separate disease entities. Overall, examines Myc function and regulation in neuroblastoma, prognosis in the high-risk group is quite poor, with long- and discusses emerging therapies that target Mycn. term survival of 30–40%. Oncogene (2010) 29, 1249–1259; doi:10.1038/onc.2009.512; published online 25 January 2010 Familial neuroblastoma Keywords: myc; mycn; neuroblastoma; N-myc; mTor; PI3K Neuroblastoma may be associated with Hirschsprung disease and neurofibromatosis type 1 (Clausen et al., 1989; Rohrer et al., 2002). Mutations in PHOX2B,a homeodomain-containing important Introduction for the development in the autonomic nervous system, underlie the congenital central hypoventilation syndrome Neuroblastoma, a neoplasm of peripheral neural crest and confer a heritable predisposition to neuroblastoma; origin, is the most common malignant extracranial solid however, these mutations are not found in spontaneous tumor of childhood and accounts for 15% of cancer tumors (Mosse et al.,2004;Trochetet al.,2005;Raabe deaths in children (Park et al., 2008). Approximately 650 et al., 2008). In fact, familial neuroblastoma is quite rare, new cases are diagnosed in the United States annually and is most often because of dominant gain-of-function with peak incidence in early childhood (ages 0–4 years). mutations in the orphan receptor tyrosine kinase (RTK) The most common site of origin is in the adrenal anaplastic lymphoma kinase (ALK). ALK, which is linked medulla; however, tumors can occur anywhere along the to MYCN on 2p23, also shows sporadic gain- sympathetic chain. of-function mutation in 8% of spontaneous tumors (Chen Patients with new diagnoses are typically stratified et al., 2008; George et al., 2008; Janoueix-Lerosey et al., into risk groups based on age, stage, histopathology, 2008; Mosse´et al., 2008). Although direct connections DNA index and genetic/genomic factors. Amplification between MYCN and ALK have yet to be elucidated, of the proto-oncogene MYCN occurs in B25% of activation of Alk and other RTKs may contribute to stabilization of Mycn protein (detailed below).

Correspondence: Dr WC Gustafson, Pediatric Hematology/Oncology, University of California, Helen Diller Family Cancer Research Myc family of proto-oncogenes Building, 1450 3rd Street, San Francisco, CA 94158-9001, USA. E-mail: [email protected] Received 22 September 2009; revised 9 December 2009; accepted 15 Broadly implicated in oncogenesis, the human MYC December 2009; published online 25 January 2010 family of proto-oncogenes is among the most studied Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1250 in cancer (Meyer and Penn, 2008). Early inser- Chromatin immunoprecipitation experiments show tional mutagenesis studies in mouse identified c-MYC that c-Myc and Mycn proteins bind to promoters with (homologous to the v-myc gene that drives avian variable specificity determined by DNA ultrastructure myelocytosis) as capable of transformation by retroviral and cellular context (Fernandez et al., 2003; Li et al., promoter insertion (Payne et al., 1982). MYCN was 2003; Mao et al., 2003; Chen et al., 2004; Guccione subsequently identified as an MYC homolog amplified et al., 2006; Zeller et al., 2006; Kim et al., 2008; in neuroblastoma tumors (reviewed in Meyer and Penn, Martinato et al., 2008; Westermann et al., 2008). Myc/ 2008). Amplification of MYCN has emerged as among Max heterodimers bind to E-boxes and interact with a the clearest genetic indicators of high-risk, aggressive variety of histone modifiers, increasing histone acetyla- disease (Brodeur et al., 1984; Seeger et al., 1985). MYC tion (Bouchard et al., 2001). These alterations modify family members (c-MYC, MYC and MYCL) show chromatin at promoters, affecting differential expression in normal tissues. Expression of (McMahon et al., 2000; Frank et al., 2001; Vervoorts murine N-myc in particular is elevated in normal et al., 2003; Guccione et al., 2006; Martinato et al., 2008; mammalian developing retina, forebrain, hindbrain, Liu et al., 2009). In fact, histone modification by Mycn intestine, kidney and has functions in neuronal progeni- effectors is being exploited pharmacologically using tor cells, developing lung tissues, hematopoetic stem histone deacetylase inhibitors. These drugs may alter cells and programmed cell death in the developing limb acetylation in neuroblastomas and other MYC depen- (Zimmerman et al., 1986; Mugrauer et al., 1988; Downs dent processes thereby modulating transcription (re- et al., 1989; Hirvonen et al., 1990; Hirning et al., 1991; viewed in Witt et al., 2009). Knoepfler et al., 2002; Bettess et al., 2005; Okubo et al., Several studies have attempted to elucidate specific 2005; Ota et al., 2007; Martins et al., 2008; Xu et al., transcriptional targets for Mycn in neuroblastoma 2009). (Alaminos et al., 2003). A large number of targets Myc proteins are basic helix-loop-helix important for cell cycle control and differentiation have transcription factors. Mycn and c-Myc proteins share been characterized, including: downregulation of SKP2 several regions of homology and share similar cellular and TP53INP1 with resultant decrease in p21WAF1 (Bell functions. Myc proteins localize to the nucleus and form et al., 2007), downregulation of DKK1 upstream of the heterodimers with the basic helix-loop-helix molecule, wnt/b-catenin pathway (Koppen et al., 2007), upregula- Max (Blackwood and Eisenman, 1991; Prendergast tion of NLRR1 both important in neural cell prolifera- et al., 1991; Berberich and Cole, 1992; Blackwood tion (Hossain et al., 2008), downregulation of Fyn et al., 1992; Kato et al., 1992). Myc/Max heterodimers kinase important in differentiation (Berwanger et al., bind to DNA at specific CAC(G/A)TG ‘E-box’ se- 2002), regulation of multiple genes responsible for quences to drive transcription of targets important for pluripotency (Cotterman and Knoepfler, 2009) and proliferation, apoptosis and differentiation (Blackwell modulation of apoptosis by upregulation of p53 and et al., 1990, 1993; Amati et al., 1992; Kretzner et al., Mdm2 (Slack et al., 2005b). The multidrug resistance 1992). Max also heterodimerizes with Mxd or Mnt gene MRP1 is regulated by Mycn, driving chemotherapy proteins to influence the transcription of other down- resistance (Manohar et al., 2004). Importantly, Mycn stream genes and often to antagonize the proliferative upregulates oncogenic microRNAs, which have wide effects of Myc proteins (Ayer et al., 1993; Zervos et al., ranging effects on cancer (reviewed in Schulte et al., 1993; Hurlin et al., 1995, 1997; Walkley et al., 2005). In 2009). Mycn also controls several proteins important in many systems, Mxd/Max or Mnt/Max heterodimers ribosome biogenesis (Boon et al., 2001) affecting protein oppose the actions of Myc/Max to repress transcription; synthesis (reviewed in Ruggero, 2009). however, very little is known about these interactions in Different groups seeking to stratify neuroblastoma neuroblastoma (Grandori et al., 2000; Patel et al., 2004). risk using gene expression micoarrays have generated The complex competition among Myc, Mxd and Mnt gene lists that are largely non-overlapping (Berwanger proteins for binding to Max further modulates the effects et al., 2002; Ohira et al., 2005; Schramm et al., 2005, of both c-Myc, and probably Mycn, on gene expression. 2009; Oberthuer et al., 2006). Similar strategies using real-time PCR transcript analysis, are also being performed (Vermeulen et al., 2009). Direct comparison Downstream of Mycn of targets from chromatin immunoprecipitation shows that Mycn and c-Myc have many overlapping targets E-boxes are common (B25% of known promoters) with (Westermann et al., 2008). Further refinement of 410 000 sites per cell (Fernandez et al., 2003; Li et al., microarray and chromatin immunoprecipitation techni- 2003; Zeller et al., 2006). That there are more E-box ques and identification of critical transcriptional targets sequences than Myc molecules in cells represents a specific to Mycn may provide insights into both biology conundrum apparently common to many transcription and therapy for neuroblastoma. factors (Farnham, 2009). Adding further complexity to this system, Myc proteins also regulate downstream targets through cap-dependent methylation, altering Mycn in cell cycle control both global translation as well as the translation of specific proteins (Barna et al., 2008; Cole and Cowling, In normal cells, levels of Myc proteins are tightly 2008). regulated, with increased levels driven in part through

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1251 activation of phosphatidylinositol 3-kinase (PI3K), natural antisense transcripts are frequently co-amplified which stabilizes Mycn and c-Myc proteins (Figure 1), with MYCN in neuroblastoma, the importance of these enabling entry into the cell cycle (Marque´s et al., 2008). co-amplified sequences remains unclear (Krystal et al., In addition to the clearly defined function of Myc family 1990; Armstrong and Krystal, 1992; Jacobs et al., 2009). members in control of the cell cycle, c-Myc contributes non-transcriptionally to the initation of DNA replica- tion (Dominguez-Sola et al., 2007). Myc/Max dimers also bind and inhibit Miz-1, a helix-loop-helix transcrip- Mycn and apoptosis tion factor (reviewed in Herold et al., 2009). Free Miz-1 promotes transcription of p15INK4b and p21Cip1 In addition to promoting proliferation, the expression of proteins associated with cell cycle arrest. Inhibition of c-Myc and Mycn actually drives apoptosis (Askew et al., Miz-1 in response to Myc/Max binding contributes to 1991; Evan et al., 1992; Shi et al., 1992; Fulda et al., immortalization, transformation and oncogenesis 1999; Paffhausen et al., 2007; Ushmorov et al., 2008). (Seoane et al., 2001; Staller et al., 2001; Herold et al., Transformation by Myc proteins, therefore, requires 2008). Interactions between Mycn and Miz-1 are concomitant inhibition of apoptosis. Tissue-specific incompletely characterized. Expression of Miz-1 has expression of a switchable allele of c-Myc also induced been associated with favorable outcome in neuroblas- apoptosis in vivo. In contrast, proliferation and tumori- toma, however, consistent with an interaction among genesis required lower level, continuous and deregulated Miz-1, Mycn and Max (Ikegaki et al., 2007). expression of c-Myc (Murphy et al., 2008). Transcription of MYCN is downregulated by the neuroblastoma differentiating agent retinoic acid and upregulated by several known transcription factors Figure 1 Schematic model of regulatory pathways involved in including and Sp1/Sp3 (Thiele et al., 1985; Inge Mycn stabilization. In proliferating cells, Mycn is initially et al., 2002; Kramps et al., 2004; Kanemaru et al., 2008). phosphorylated at serine 62 by cyclin-dependent kinase 1/CyclinB. Sonic hedgehog indirectly regulates transcription of The S62-phosphorylated protein is stabilized and competent to MYCN in developing neurons (Kenney et al., 2003, enter the nucleus. Nuclear Mycn binds to its partner, the helix- 2004; Oliver et al., 2003; Mill et al., 2005). Levels of Myc loop-helix transcription factor Max and stimulates the transcrip- tion of a target genes important in cell cycle, proliferation, mRNA are also by alternate internal ribosomal entry differentiation and apoptosis. This priming phosphorylation at sites (Barna et al., 2008; Cobbold et al., 2008). Although S62 allows the binding of Gsk3b, as well as Pin1 and PP2A in a complex also containing Axin. Active Gsk3b phosphorylates Mycn at threonine 58, producing doubly phosphorylated, stabilized and transcriptionally active Mycn. In a Pin1-mediated process, PP2A dephosphorylates the S62 phosphate, enabling binding of ubiquitin ligase Fbw7. Fbw7 subsequently drives poly-ubiquitination and proteasomal degradation of Mycn. Aurora A kinase can bind to and stabilize polyubiquitinated/phosphorylated Mycn, potentially leaving it competent to bind to Max and activate transcription. In this model, upstream signaling starts at the membrane with receptor tyrosine kinases (RTKs), which are either activated by ligand or by constitutively activating mutations (in Alk, for example). RTKs activate phosphatidylinositol 3-kinase (PI3K), which catalyzes the conversion of phosphatidylinositol-3,4- bisphosphate (PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3). PIP3 then binds to Akt, localizing it to the membrane and allowing the phosphorylation and activation of Akt by membrane bound Pdk1 at threonine 308. Active Akt then phosphorylates Glycogen synthase kinase 3b (Gsk3b) and inacti- vates it, blocking Gsk3b-mediated phosphorylation of Mycn T58 and thereby stabilizing Mycn. Akt also activates mammalian target of rapamycin (mTOR) through several indirect signaling mechan- isms including through Tsc2/Tsc1 and Rheb. mTOR exists in two distinct complexes. mTORC1 complex, the rapamycin sensitive form, consists of mTOR bound to multiple effector proteins, including Raptor and Lst8, and is important for the promotion of global translation through S6K and 4EBP. mTORC1 also directly phosphorylates and inhibits PP2A, enabling the accumulation of doubly phosphorylated, active Mycn and contributing to the general proliferation-promoting downstream effects of the PI3K/ Akt/Mycn pathway (Cheng et al., 2007). The mTORC2 complex, also known as Pdk2, contains mTOR, rictor, Lst8 and mSin1. mTORC2 phosphorylates Akt at serine 473 further activating this kinase. Inhibitors of this pathway could potentially destabilize Mycn. These inhibitors are currently in clinical trials or in clinical development and are denoted by red octagons. The Aurora A Kinase inhibitor is denoted by an orange octagon, which represents targeted kinase inhibition, but no anticipated activity against Mycn itself. Relevant references are contained within the corre- sponding text.

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1252 Although the association between MYCN amplifica- apoptotic stimuli (reviewed in Li and Hann, 2009; Van tion and poor outcome has been reproduced in Maerken et al., 2009b). numerous studies over decades, a similar association Neuroblastoma tumors at diagnosis are generally wild between expression of Mycn and outcome remains type for p53 and respond to chemotherapy. Children controversial (Chan et al., 1997; Cohn et al., 2000; with high-risk tumors generally relapse, whereas those Tang et al., 2006). Data from preclinical models of with low and intermediate disease are typically cured. As switchable myc above suggest that low levels of myc stated above, Mycn-induced downregulation of the p53 proteins may drive proliferation, with higher levels axis could potentially underlie the minimal residual required to induce apoptosis. If it is true in the context disease that drives subsequent relapse in newly diag- of Mycn and neuroblastoma, then MYCN amplification nosed MYCN-amplified neuroblastomas. This hypo- may serve primarily to dysregulate Mycn during the cell thesis is supported by the identification of Mdm2 as an cycle, rather than simply driving high-level expression. Mycn target, and in studies showing that Mdm2 This hypothesis is consistent with the claims that haploinsufficiency inhibits tumorigenesis in MYCN- amplification and not overexpression is predictive of driven models for neuroblastoma (Slack et al., 2005a; aggressive disease, although inconsistent with observa- Chen et al., 2009). In fact, small molecule inhibitors of tions that MYCN is often amplified and overexpressed the p53/Mdm2 interaction (‘Nutlins’) are currently in to extreme levels in neuroblastoma. development and have shown some promise for the pre- Mechanisms through which apoptosis is inhibited as a clinical treatment of cancers including neuroblastoma contributer to Mycn-driven transformation are complex (Barbieri et al., 2006; Chen et al., 2009; Van Maerken and not yet fully elucidated. Silencing of the apoptotic et al., 2009a). initiator Casp8 is observed frequently in neuroblastoma (Stupack et al., 2006). Crosstalk with the p53 pathway has been implicated in apoptosis mediated by both Modeling MYCN-amplified neuroblastoma Mycn and c-Myc (reviewed in Hoffman and Lieber- mann, 2008; Van Maerken et al., 2009b). Mutations in Mice carrying an MYCN transgene under control of the p53 are rare in primary neuroblastoma (o2%) irrespec- rat tyrosine hydroxylase promoter develop neuroblas- tive of MYCN amplification (Vogan et al., 1993). toma tumors several months after birth (Norris et al., However, inactivating mutations in p53 and in p53 2000; Weiss et al., 2000). Tumors from mice transgenic pathway members are common at relapse (Keshelava for TH-MYCN develop in adrenal and mesenteric et al., 1997, 2000; Tweddle et al., 2001; Carr et al., 2006). ganglia and in paraspinous locations. Histolology and Myc proteins indirectly regulate the p53 pathway and genetics show similarities with high-risk neuroblastoma p53-dependent apoptosis through the p14ARF-MDM2- (Weiss et al., 2000; Hackett et al., 2003; Moore et al., p53 axis (reviewed in Van Maerken et al., 2009b). 2008). In relatively resistant strains or subspecies (for At baseline, p53 is tightly regulated by Mdm2, which example C57BL/6, BALBc or Mus musculus castaneus), binds to and inhibits the transactivation domain of p53 a range of differentiation was observed in murine (Oliner et al., 1993; Thut et al., 1997). Mdm2 also tumors. However, when crossed into highly penetrant ubiquitinates and targets p53 protein for degradation strains, such as 129 SvJ, the histology was uniformly (reviewed in Coutts et al., 2009). Further regulation is undifferentiated small round blue cells. conferred by the tumor suppressor protein p14ARF, Cell lines derived from TH-MYCN tumors retain the which binds to and inhibits Mdm2, allowing activation ability to form tumors in nude mice (Cheng et al., 2007). and stabilization of p53 (Kamijo et al., 1998; Zindy Unlike xenograft models of neuroblastoma, TH-MYCN et al., 1998; Weber et al., 1999; Midgley et al., 2000; Lin tumors show native host interactions with the tumor and Lowe, 2001). The balance between apoptosis and microenvironment including vascular cells (Chesler survival remains in equilibrium through multiple feed- et al., 2007). When treated with chemotherapy, tumors back and feed-forward loops affected by other signaling in this model, similar to their human p53 wild-type pathways including external apoptotic stimuli. counterparts, undergo apoptosis in a p53-dependent In neuroblastoma cells, Mycn directly stimulates manner. Furthermore, tumors from TH-MYCN;p53À/ þ transcription of MDM2 (Slack et al., 2005a; Barbieri mice were refractory to cytoxic chemotherapy (Chesler et al., 2006; Chen et al., 2009). The resulting inhibition et al., 2008). These data and corresponding data from of p53 may in part allow cells to escape Mycn-primed human tumors (Keshelava et al., 1998, 2000; Xue et al., apoptosis. Myc and Mycn also indirectly inhibit p14ARF, 2007) collectively suggest a model in which newly through directly driving transcription factors including diagnosed neuroblastoma tumors impair the p14ARF- TWIST1, resulting in activation of MDM2 and escape MDM2-p53 axis in the absence of p53 mutation, from apoptosis (Maestro et al., 1999; Valsesia-Witt- enabling tumors to arise. Subsequent chemotherapy mann et al., 2004). The p14ARF protein can in-turn feed subsequently provides strong selective pressure for back and bind to Myc and Mycn proteins, abrogating inactivating mutations in p53 or in components of the their ability to activate downstream targets (Qi et al., p53 pathway, resulting in chemotherapy-refractory 2004; Amente et al., 2007). The complex interactions tumors seen clinically in relapsed patients. Thus, among Myc/Mycn, Mdm2 and p14ARF provide mechan- although mutation at p53 rarely contributes to the isms through which Mycn both indirectly activates or biology of primary neuroblastoma, therapy-selected inactivates p53, altering the sensitivity of cells to mutations in p53 drive a genetically distinct tumor at

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1253 relapse. This therapy-associated alteration in the biology network involving both feedback and feed-forward of neuroblastoma presents a challenge in identifying loops (Figure 1) (Kenney et al., 2004; Sjostrom et al., effective treatments for relapsed tumors. 2005; Chesler et al., 2006; Kang et al., 2008). Although both c-Myc and Mycn are widely phosphorylated, sites critical to stabilization of Myc proteins are located in the Mycn as a target for therapy N-terminal Myc Box I, which is commonly mutated in c-Myc in Burkitt lymphoma (Bhatia et al., 1993; Smith- In light of both the frequency and importance of MYCN Srensen et al., 1996; Bahram et al., 2000). Comparable amplification in pathogenesis of high-risk neuroblastoma, mutations in c-Myc and Mycn are rare in solid tumors blockade of Mycn signaling represents an important including neuroblastoma. The important residues in the approach for the developmental therapeutics. The Myc Box I region of c-Myc and Mycn are threonine 58 resistance of high-risk relapsed neuroblastoma to con- and serine 62 (Henriksson et al., 1993; Pulverer et al., ventional chemotherapy and the high morbidity and 1994; Lutterbach and Hann, 1999; Sjostrom et al., mortality in these patients present a formidable chal- 2005). lenge for clinicians. The prominence of MYCN ampli- Phosphorylation at S62 stabilizes Mycn protein fication in the pathogenesis of this disease points to and primes it for phosphorylation at T58. Candidate Mycn as a potential therapeutic target. kinases proposed to phosphorylate c-Myc at S62 Neuroblastoma presents both common and unique include extracellular signal-regulated kinase, the challenges for therapy. The initial response to chemother- canonical downstream effector of the Ras/Raf/ apy even in high-risk disease is somewhat unusual. MAPK; c-Jun N-terminal kinase and cyclin-depen- However, the acquisition of p53 mutant, therapy- dent kinase 1 pathway (Henriksson et al., 1993; refractory disease is common to many cancers (McDer- Lutterbach and Hann, 1999; Sears et al., 2000; Benassi mott et al., 2008). Deregulated expression of Mycn may et al.,2006).Asshownincerebellarneuralprogenitor contribute to genomic instability and, in combination cells and synchronized mitotic SJ8 neuroblastoma with the strong selective pressures of chemotherapy and cells, cyclin-dependent kinase 1 is thus far the only radiation, select for mutation at p53 through interactions candidate priming kinase for Mycn at S62 (Sjostrom with Mdm2 and p14Arf detailed above, and by driving et al., 2005). the cell cycle and activating p53-dependent check points. Myc and Mycn proteins monophosphorylated at S62 Alleviation of check-point activation by blocking Mycn are substrates for a second phosphorylation at T58, itself could conceivably impair the acquisition of p53 controlled by glycogen synthase kinase 3b (Gsk3b). mutant, refractory disease. Gsk3b signals in the Wnt pathway (reviewed in Transcription factors have long been considered as MacDonald et al., 2009), and also downstream of the targets for cancer therapy; however, clinical approaches PI3K/Akt/mTOR pathway (reviewed in Engelman, to block this class of molecules remain elusive. Clearly, 2009; Ma and Blenis, 2009; Memmott and Dennis, the most direct means of silencing these molecules would 2009). The S62 priming phosphorylation site in Myc/ be by disrupting Myc synthesis directly through RNAi Mycn promotes interaction with a complex containing methods and there are numerous examples of siRNA Gsk3b, Pin1, PP2A and Axin. This complex induces the used successfully on cultured cells. However, although phosphorylation at T58 by Gsk3b in both Myc and these approaches are extremely useful tools in the Mycn. For c-Myc, Pin1 has been shown to regulate laboratory, they have yet to achieve regular use in the dephosphorylation of S62 by PP2A, a phosphatase and clinic largely because of difficulty with delivery (White- tumor suppressor that itself is regulated by mTOR head et al., 2009). (Peterson et al., 1999; Gingras et al., 2001; Hartley and It is difficult to develop drugs with activities sufficient Cooper, 2002; Yeh et al., 2004; Arnold and Sears, to block protein–protein interactions or binding of such 2006; Arnold et al., 2009). Regulation of Mycn dephos- factors to target sequences. Molecules developed against phorylation by Pin1 and PP2A has not been fully c-Myc have been primarily directed against the Myc/Max characterized. interaction domain and so far have fairly low potency. Myc proteins monophosphorylated at T58, then bind Progress is being made in this area, however, and the to the E3 ligase Fbxw7 and are targeted for ubiquitina- development of inhibitors, which dissociate Myc/Max tion and degradation (Saksela et al., 1992; Lutterbach heterodimers is important (Yin et al., 2003; Wang et al., and Hann, 1994; Sears et al., 1999; Gregory and Hann, 2006; Berg, 2008; Brooks and Hurley, 2009). Chemists 2000; Oliver et al., 2003; Herbst et al., 2004; Kenney are good at developing kinase inhibitors, however, and et al., 2004; Welcker et al., 2004; Yada et al., 2004; Otto the stability of Myc molecules is regulated at a number of et al., 2009). Aurora kinase A, which has critical levels by kinases and critical phospho-residues. functions in cell cycle regulation and spindle assembly, contributes at this step to the stabilization of phos- phorylated and ubiquitinated Mycn, but not of equiva- Post-transcriptional modification and stabilization of Myc lently modified c-myc (Otto et al., 2009). Consistent with proteins these findings, expression of Aurora A kinase is a negative prognostic factor in neuroblastoma (Shang In neuroblastoma and in neural progenitor cells, Mycn et al., 2009). Although the emerging function of protein stability is regulated by a complex signaling inhibitors of Aurora kinase A in cancer therapy

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1254 suggested that inhibitors of Aurora kinase A might have 2005; Chesler et al., 2006). These data show that unique and multifaceted functions in the therapy of degradation of Mycn is a critical downstream factor in neuroblastoma, stabilization of Mycn apparently re- the efficacy of PI3K/mTOR pathway and suggest that quires a scaffold function of Aurora kinase and was clinical inhibitors of PI3K should show activity in independent of Aurora A kinase activity (Maris, 2009; Mycn-driven neuroblastoma (detailed review in Fulda, Otto et al., 2009). 2009).

Myc proteins as downstream targets of PI3K Mycn stability as a therapeutic target in neuroblastoma

The phosphorylation of c-Myc and Mycn is thus The post-translational modification and stabilization regulated directly by Gsk3b, and indirectly by upstream of Myc and Mycn proteins represents an area with signaling through RTKs, PI3K, Akt and mTOR. RTKs promise for currently available and emerging-tar- are important to PI3K signaling in neuroblastoma. As geted therapies. Inhibitors of RTKs (Alk and Trk), mentioned above, activating mutations in the orphan PI3K and Akt should activate Gsk3b (which is RTK ALK are found in B9% of neuroblastoma. Other negatively regulated by phosphorylation), whereas RTKs that could potentially regulate stabilization of inhibitors of mTOR kinase activity should inhibit Mycn include the insulin-like growth factor receptor and the de-activation of PP2A by mTORC1, enabling the Trk family of neurotropin receptors (TrkA, TrkB PP2A to dephosphorylate S62, collectively driving and TrkC). Although mutation in Trk genes is not degradation of Mycn in neuroblastoma (Figure 1). typically observed in neuroblastoma, expression of Specific Alk and Trk inhibitors are currently in TrkA correlates with favorable prognosis, whereas development and have shown promise preclinically expression of TrkB correlates with amplification of and in phase I trials (reviewed in Chiarle et al., 2008; MYCN and poor outcome (reviewed in Brodeur et al., Li and Morris, 2008; Brodeur et al., 2009; Mosse´ 2009). Although neither Trk nor Alk proteins have been et al., 2009). directly linked to stabilization of Mycn, these kinases Downstream of RTKs, inhibitors of PI3K, mTOR, are all upstream activators of PI3K, implying a possible dual inhibitors of PI3K/mTOR and inhibitors of Akt connection (Bai et al., 2000; Norris et al., 2000; Ho are all in clinical trials (reviewed in Garcia-Echeverria et al., 2002; Marzec et al., 2007). and Sellers, 2008; Engelman, 2009). Allosteric inhibitors RTKs activate PI3K, which catalyzes the conversion of mTORC1 (so-called rapalogs) block mTOR indepen- of phosphatidylinositol-3,4-bisphosphate (PIP2) to dently of ATP binding, are currently being tested in phosphatidylinositol-3,4,5-triphosphate (PIP3) (re- neuroblastoma and have shown mixed results preclin- viewed in Ma and Blenis, 2009; Memmott and Dennis, cally (Houghton et al., 2008; Johnsen et al., 2008; Maris 2009). PIP3 binds to Akt and localizes it to the et al., 2008a; Wagner and Danks, 2009). These agents membrane, enabling phosphorylation at T308 by the only affect some outputs of mTORC1. In comparison, kinase PDK1. Activation of Akt leads to phosphoryla- ATP-competitive inhibitors of mTOR more completely tion and inactivation of Gsk3b, stabilizing Mycn by block mTORC1 and also block mTORC2 (Feldman blocking phosphorylation at T58. Activated Akt also et al., 2009; Thoreen et al., 2009; Yu et al., 2009; Zask phosphorylates and inhibits the tuberous sclerosis 2 et al., 2009). The availability of inhibitors targeting (Tsc2) tumor suppressor protein. Tsc2 binds to Tsc1, RTKs, PI3K, Akt and mTOR, and the use of these to enabling the Ras-related GTPase Rheb to stimulate destabilize Mycn protein, represent important areas of mTOR bound in the mTORC1 complex of proteins. As investigation. Further, because of the complex interrela- mentioned above, mTORC1 downregulates PP2A, tion of pathway members, inhibition at one point often which normally dephosphorylates Myc/Mycn at S62, induces feedback activation in other signaling pathways, targeting it for ubiquitination, and promoting stabiliza- justifying the need to test these agents in combination tion of Myc and Mycn. Thus, activation of RTKs and using preclinical models. PI3K converge on Akt to phosphorylate and inhibit The regulation of Mycn phosphorylation also Gsk3b, blocking phosphorylation at T58 and promot- involves a priming phosphorylation at S62. As efficient ing stabilization. In addition, Akt activates mTORC1, destabilization of Mycn would require activators of inhibiting dephosphorylation of T58/S62-phosphory- kinases responsible for these phosphorylation steps, lated Myc/Mycn at S62, leading to further stabilization the ability to finesse phosphorylation at S62 presents a of Myc/Mycn. therapeutic challenge. Aurora kinase A represents an The critical importance of Mycn phosphorylation and additional therapeutic target, as Aurora kinase A stability as a downstream target of PI3K/Akt/mTOR in stabilizes Mycn at later steps. Inhibitors of Aurora A neuroblastoma cells is apparent when neuroblastoma kinase are currently in clinical trials in neuroblastoma cells are treated with a broad spectrum PI3K inhibitor. (Shang et al., 2009). However, as Mycn is stabilized by Activation of Akt predicts poor outcome in neuroblas- a kinase-independent activity of Aurora A, these toma patients (Opel et al., 2007). Treated cells show a inhibitors are unlikelytoaffectMycnprotein decreased proliferation, which is largely rescued (Gautschi et al., 2008; Maris, 2009). An allosteric when they are engineered to express T58/S62 phospho- and ATP-competitive small molecule inhibitor of rylation site-deficient Mycn mutants (Sjostrom et al., Aurora A–Mycn interactions, if it could be developed,

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1255 should retain the ability to block kinase-dependent Conflict of interest functions, whereas also twisting Aurora A kinase, therebydisruptingascaffolding function and degrad- The authors declare no conflict of interest. ing Mycn protein. The interplay among RTKs, PI3K, Akt, mTORC1/2, Aurora A and Mycn is quite complex. However, the broad functions for these Acknowledgements kinases in cancer biology, and the specific function in regulating the stabilization of Mycn proteins suggests We thank Chris Hackett and Theo Nicolaides for critical functions in both Mycn-driven and Mycn-independent review. We acknowledge support from NIH grants CA133091, NS055750, CA102321, CA097257, CA128583; Burroughs cancers including neuroblastoma. The current avail- Wellcome Fund, American Brain Tumor Association, The ability of clinical Alk, Trk, PI3K, mTOR and Aurora Brain Tumor Society, Accelerate Brain Cancer Cure; Alex’s inhibitors presents an important translational oppor- Lemonade Stand, Children’s National Brain Tumor, Wallace tunity to test these agents inchildrenwithhigh-risk H. Coulter, Katie Dougherty, Pediatric Brain Tumor, Samuel neuroblastoma. G Waxman and V Foundations.

References

Alaminos M, Mora J, Cheung N-KV, Smith A, Qin J, Chen L et al. Berberich SJ, Cole MD. (1992). Casein kinase II inhibits the DNA- (2003). Genome-wide analysis of gene expression associated with binding activity of Max homodimers but not Myc/Max hetero- MYCN in human neuroblastoma. Cancer Res 63: 4538–4546. dimers. Genes Dev 6: 166–176. Amati B, Dalton S, Brooks MW, Littlewood TD, Evan GI, Land H. Berg T. (2008). Inhibition of transcription factors with small organic (1992). Transcriptional activation by the human c-Myc oncoprotein molecules. Curr Opin Chem Biol 12: 464–471. in yeast requires interaction with Max. Nature 359: 423–426. Berwanger B, Hartmann O, Bergmann E, Bernard S, Nielsen D, Amente S, Gargano B, Diolaiti D, Della Valle G, Lania L, Majello B. Krause M et al. (2002). Loss of a FYN-regulated differentiation and (2007). p14ARF interacts with N-Myc and inhibits its transcrip- growth arrest pathway in advanced stage neuroblastoma. Cancer tional activity. FEBS Lett 581: 821–825. Cell 2: 377–386. Armstrong BC, Krystal GW. (1992). Isolation and characterization of Bettess MD, Dubois N, Murphy MJ, Dubey C, Roger C, Robine S complementary DNA for N-cym, a gene encoded by the DNA et al. (2005). c-Myc is required for the formation of intestinal crypts strand opposite to N-myc. Cell Growth Differ 3: 385–390. but dispensable for homeostasis of the adult intestinal epithelium. Arnold HK, Sears RC. (2006). Protein phosphatase 2A regulatory Mol Cell Biol 25: 7868–7878. subunit B56alpha associates with c-myc and negatively regulates c- Bhatia K, Huppi K, Spangler G, Siwarski D, Iyer R, Magrath I. myc accumulation. Mol Cell Biol 26: 2832–2844. (1993). Point mutations in the c-Myc transactivation domain are Arnold HK, Zhang X, Daniel CJ, Tibbitts D, Escamilla-Powers J, common in Burkitt0s lymphoma and mouse plasmacytomas. Nat Farrell A et al. (2009). The Axin1 scaffold protein promotes Genet 5: 56–61. formation of a degradation complex for c-Myc. EMBO J 28: Blackwell TK, Kretzner L, Blackwood EM, Eisenman RN, Weintraub 500–512. H. (1990). Sequence-specific DNA binding by the c-Myc protein. Askew DS, Ashmun RA, Simmons BC, Cleveland JL. (1991). Science 250: 1149–1151. Constitutive c-myc expression in an IL-3-dependent myeloid cell Blackwell TK, Huang J, Ma A, Kretzner L, Alt FW, Eisenman RN line suppresses cell cycle arrest and accelerates apoptosis. Oncogene et al. (1993). Binding of myc proteins to canonical and noncanonical 6: 1915–1922. DNA sequences. Mol Cell Biol 13: 5216–5224. Attiyeh EF, London WB, Mosse´YP, Wang Q, Winter C, Khazi D Blackwood EM, Eisenman RN. (1991). Max: a helix-loop-helix zipper et al. (2005). Chromosome 1p and 11q deletions and outcome in protein that forms a sequence-specific DNA-binding complex with neuroblastoma. N Engl J Med 353: 2243–2253. Myc. Science 251: 1211–1217. Ayer DE, Kretzner L, Eisenman RN. (1993). Mad: a heterodimeric Blackwood EM, Lu¨scher B, Eisenman RN. (1992). Myc and Max partner for Max that antagonizes Myc transcriptional activity. Cell associate in vivo. Genes Dev 6: 71–80. 72: 211–222. Boon K, Caron HN, van Asperen R, Valentijn L, Hermus MC, van Bahram F, von der Lehr N, Cetinkaya C, Larsson LG. (2000). c-Myc Sluis P et al. (2001). N-myc enhances the expression of a large set of hot spot mutations in lymphomas result in inefficient ubiquitination genes functioning in ribosome biogenesis and protein synthesis. and decreased proteasome-mediated turnover. Blood 95: 2104–2110. EMBO J 20: 1383–1393. Bai RY, Ouyang T, Miething C, Morris SW, Peschel C, Duyster J. Bouchard C, Dittrich O, Kiermaier A, Dohmann K, Menkel A, Eilers (2000). Nucleophosmin-anaplastic lymphoma kinase associated M et al. (2001). Regulation of cyclin D2 gene expression by the with anaplastic large-cell lymphoma activates the phosphatidylino- Myc/Max/Mad network: Myc-dependent TRRAP recruitment sitol 3-kinase/Akt antiapoptotic signaling pathway. Blood 96: and histone acetylation at the cyclin D2 promoter. Genes Dev 15: 4319–4327. 2042–2047. Barbieri E, Mehta P, Chen Z, Zhang L, Slack A, Berg S et al. (2006). Bown N, Cotterill S, Lastowska M, O0Neill S, Pearson AD, Plantaz D MDM2 inhibition sensitizes neuroblastoma to chemotherapy- et al. (1999). Gain of chromosome arm 17q and adverse outcome in induced apoptotic cell death. Mol Cancer Ther 5: 2358–2365. patients with neuroblastoma. N Engl J Med 340: 1954–1961. Barna M, Pusic A, Zollo O, Costa M, Kondrashov N, Rego E et al. Brodeur GM, Seeger RC, Schwab M, Varmus HE, Bishop JM. (1984). (2008). Suppression of Myc oncogenic activity by ribosomal protein Amplification of N-myc in untreated human neuroblastomas haploinsufficiency. Nature 456: 971–975. correlates with advanced disease stage. Science 224: 1121–1124. Bell E, Lunec J, Tweddle DA. (2007). Cell cycle regulation targets of Brodeur GM, Minturn JE, Ho R, Simpson AM, Iyer R, Varela CR MYCN identified by gene expression microarrays. Cell Cycle 6: et al. (2009). Trk receptor expression and inhibition in neuroblas- 1249–1256. tomas. Clin Cancer Res 15: 3244–3250. Benassi B, Fanciulli M, Fiorentino F, Porrello A, Chiorino G, Loda M Brooks T, Hurley L. (2009). The role of supercoiling in transcriptional et al. (2006). c-Myc phosphorylation is required for cellular response control of MYC and its importance in molecular therapeutics. Nat to oxidative stress. Mol Cell 21: 509–519. Rev Cancer 9: 849–861.

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1256 Carr J, Bell E, Pearson ADJ, Kees UR, Beris H, Lunec J et al. (2006). Feldman ME, Apsel B, Uotila A, Loewith R, Knight ZA, Increased frequency of aberrations in the p53/MDM2/p14(ARF) Ruggero D et al. (2009). Active-site inhibitors of mTOR target pathway in neuroblastoma cell lines established at relapse. Cancer rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol Res 66: 2138–2145. 7: e38. Chan HS, Gallie BL, DeBoer G, Haddad G, Ikegaki N, Fernandez PC, Frank SR, Wang L, Schroeder M, Liu S, Greene J et al. Dimitroulakos J et al. (1997). MYCN protein expression as a (2003). Genomic targets of the human c-Myc protein. Genes Dev 17: predictor of neuroblastoma prognosis. Clin Cancer Res 3: 1115–1129. 1699–1706. Frank SR, Schroeder M, Fernandez P, Taubert S, Amati B. (2001). Chen Q-R, Bilke S, Wei JS, Whiteford CC, Cenacchi N, Krasnoselsky Binding of c-Myc to chromatin mediates mitogen-induced acetyla- AL et al. (2004). cDNA array-CGH profiling identifies genomic tion of histone H4 and gene activation. Genes Dev 15: 2069–2082. alterations specific to stage and MYCN-amplification in neuro- Fulda S, Lutz W, Schwab M, Debatin KM. (1999). MycN sensitizes blastoma. BMC Genomics 5: 70. neuroblastoma cells for drug-induced apoptosis. Oncogene 18: Chen Y, Takita J, Choi YL, Kato M, Ohira M, Sanada M et al. (2008). 1479–1486. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 455: Fulda S. (2009). The PI3K/Akt/mTOR pathway as therapeutic target 971–974. in neuroblastoma. Curr Cancer Drug Targets 9: 729–737. Chen Z, Lin Y, Barbieri E, Burlingame S, Hicks J, Ludwig A et al. Garcia-Echeverria C, Sellers WR. (2008). Drug discovery approaches (2009). Mdm2 deficiency suppresses MYCN-driven neuroblastoma targeting the PI3K/Akt pathway in cancer. Oncogene 27: 5511–5526. tumorigenesis in vivo. Neoplasia 11: 753–762. Gautschi O, Heighway J, Mack PC, Purnell PR, Lara PN, Gandara Cheng AJ, Cheng NC, Ford J, Smith J, Murray JE, Flemming C et al. DR. (2008). Aurora kinases as anticancer drug targets. Clin Cancer (2007). Cell lines from MYCN transgenic murine tumours reflect the Res 14: 1639–1648. molecular and biological characteristics of human neuroblastoma. George RE, Sanda T, Hanna M, Fro¨hling S, Luther W, Zhang J et al. Eur J Cancer 43: 1467–1475. (2008). Activating mutations in ALK provide a therapeutic target in Chesler L, Schlieve C, Goldenberg DD, Kenney A, Kim G, McMillan neuroblastoma. Nature 455: 975–978. A et al. (2006). Inhibition of phosphatidylinositol 3-kinase Gingras AC, Raught B, Gygi SP, Niedzwiecka A, Miron M, Burley destabilizes Mycn protein and blocks malignant progression in SK et al. (2001). Hierarchical phosphorylation of the translation neuroblastoma. Cancer Res 66: 8139–8146. inhibitor 4E-BP1. Genes Dev 15: 2852–2864. Chesler L, Goldenberg DD, Seales IT, Satchi-Fainaro R, Grimmer M, Grandori C, Cowley SM, James LP, Eisenman RN. (2000). The Myc/ Collins R et al. (2007). Malignant progression and blockade of Max/Mad network and the transcriptional control of cell behavior. angiogenesis in a murine transgenic model of neuroblastoma. Annu Rev Cell Dev Biol 16: 653–699. Cancer Res 67: 9435–9442. Gregory MA, Hann SR. (2000). c-Myc proteolysis by the ubiquitin- Chesler L, Goldenberg DD, Collins R, Grimmer M, Kim GE, Tihan T proteasome pathway: stabilization of c-Myc in Burkitt’s lymphoma et al. (2008). Chemotherapy-induced apoptosis in a transgenic cells. Mol Cell Biol 20: 2423–2435. model of neuroblastoma proceeds through p53 induction. Neoplasia Guccione E, Martinato F, Finocchiaro G, Luzi L, Tizzoni L, Dall0 10: 1268–1274. Olio V et al. (2006). Myc-binding-site recognition in the Chiarle R, Voena C, Ambrogio C, Piva R, Inghirami G. (2008). The is determined by chromatin context. Nat Cell Biol anaplastic lymphoma kinase in the pathogenesis of cancer. Nat Rev 8: 764–770. Cancer 8: 11–23. Guo C, White PS, Weiss MJ, Hogarty MD, Thompson PM, Stram DO Clausen N, Andersson P, Tommerup N. (1989). Familial occurrence of et al. (1999). Allelic deletion at 11q23 is common in MYCN single neuroblastoma, von Recklinghausen0s neurofibromatosis, Hirsch- copy neuroblastomas. Oncogene 18: 4948–4957. sprung’s agangliosis and jaw-winking syndrome. Acta Paediatr Hackett CS, Hodgson JG, Law ME, Fridlyand J, Osoegawa K, de Scand 78: 736–741. Jong PJ et al. (2003). Genome-wide array CGH analysis of murine Cobbold LC, Spriggs KA, Haines SJ, Dobbyn HC, Hayes C, de Moor neuroblastoma reveals distinct genomic aberrations which parallel CH et al. (2008). Identification of internal ribosome entry segment those in human tumors. Cancer Res 63: 5266–5273. (IRES)-trans-acting factors for the Myc family of IRESs. Mol Cell Hartley D, Cooper GM. (2002). Role of mTOR in the degradation of Biol 28: 40–49. IRS-1: regulation of PP2A activity. J Cell Biochem 85: 304–314. Cohn SL, London WB, Huang D, Katzenstein HM, Salwen HR, Henriksson M, Bakardjiev A, Klein G, Lu¨scher B. (1993). Phosphor- Reinhart T et al. (2000). MYCN expression is not prognostic of ylation sites mapping in the N-terminal domain of c-myc modulate adverse outcome in advanced-stage neuroblastoma with nonampli- its transforming potential. Oncogene 8: 3199–3209. fied MYCN. J Clin Oncol 18: 3604–3613. Herbst A, Salghetti SE, Kim SY, Tansey WP. (2004). Multiple cell- Cole MD, Cowling VH. (2008). Transcription-independent functions type-specific elements regulate Myc protein stability. Oncogene 23: of MYC: regulation of translation and DNA replication. Nat Rev 3863–3871. Mol Cell Biol 9: 810–815. Herold S, Hock A, Herkert B, Berns K, Mullenders J, Beijersbergen R Cotterman R, Knoepfler PS. (2009). N-Myc regulates expression of et al. (2008). Miz1 and HectH9 regulate the stability of the pluripotency genes in neuroblastoma including lif, , , and checkpoint protein, TopBP1. EMBO J 27: 2851–2861. lin28b. PLoS One 4: e5799. Herold S, Herkert B, Eilers M. (2009). Facilitating replication under Coutts AS, Adams CJ, La Thangue NB. (2009). p53 ubiquitination by stress: an oncogenic function of MYC? Nat Rev Cancer 9: 441–444. Mdm2: a never ending tail? DNA Repair (Amst) 8: 483–490. Hirning U, Schmid P, Schulz WA, Rettenberger G, Hameister H. Dominguez-Sola D, Ying CY, Grandori C, Ruggiero L, Chen B, Li M (1991). A comparative analysis of N-myc and c-myc expression et al. (2007). Non-transcriptional control of DNA replication by c- and cellular proliferation in mouse organogenesis. Mech Dev 33: Myc. Nature 448: 445–451. 119–125. Downs KM, Martin GR, Bishop JM. (1989). Contrasting patterns of Hirvonen H, Ma¨kela¨TP, Sandberg M, Kalimo H, Vuorio E, Alitalo myc and N-myc expression during gastrulation of the mouse K. (1990). Expression of the myc proto-oncogenes in developing embryo. Genes Dev 3: 860–869. human fetal brain. Oncogene 5: 1787–1797. Engelman JA. (2009). Targeting PI3K signalling in cancer: opportu- Ho R, Eggert A, Hishiki T, Minturn JE, Ikegaki N, Foster P et al. nities, challenges and limitations. Nat Rev Cancer 9: 550–562. (2002). Resistance to chemotherapy mediated by TrkB in neuro- Evan GI, Wyllie AH, Gilbert CS, Littlewood TD, Land H, Brooks M blastomas. Cancer Res 62: 6462–6466. et al. (1992). Induction of apoptosis in fibroblasts by c-myc protein. Hoffman B, Liebermann DA. (2008). Apoptotic signaling by c-MYC. Cell 69: 119–128. Oncogene 27: 6462–6472. Farnham PJ. (2009). Insights from genomic profiling of transcription Hossain MS, Ozaki T, Wang H, Nakagawa A, Takenobu H, Ohira M factors. Nat Rev Genet 10: 605–616. et al. (2008). N-MYC promotes cell proliferation through a direct

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1257 transactivation of neuronal leucine-rich repeat protein-1 (NLRR1) Koppen A, Ait-Aissa R, Hopman S, Koster J, Haneveld F, gene in neuroblastoma. Oncogene 27: 6075–6082. Versteeg R et al. (2007). Dickkopf-1 is down-regulated by Houghton PJ, Morton CL, Kolb EA, Gorlick R, Lock R, Carol H MYCN and inhibits neuroblastoma cell proliferation. Cancer Lett et al. (2008). Initial testing (stage 1) of the mTOR inhibitor 256: 218–228. rapamycin by the pediatric preclinical testing program. Pediatr Kramps C, Strieder V, Sapetschnig A, Suske G, Lutz W. (2004). E2F Blood Cancer 50: 799–805. and Sp1/Sp3 Synergize but are not sufficient to activate the MYCN Hurlin PJ, Que´va C, Koskinen PJ, Steingrı´msson E, Ayer DE, gene in neuroblastomas. J Biol Chem 279: 5110–5117. Copeland NG et al. (1995). Mad3 and Mad4: novel Max-interacting Kretzner L, Blackwood EM, Eisenman RN. (1992). Myc and transcriptional repressors that suppress c-myc dependent transfor- Max proteins possess distinct transcriptional activities. Nature mation and are expressed during neural and epidermal differentia- 359: 426–429. tion. EMBO J 14: 5646–5659. Krystal GW, Armstrong BC, Battey JF. (1990). N-myc mRNA forms Hurlin PJ, Que´va C, Eisenman RN. (1997). Mnt, a novel Max- an RNA-RNA duplex with endogenous antisense transcripts. Mol interacting protein is coexpressed with Myc in proliferating cells and Cell Biol 10: 4180–4191. mediates repression at Myc binding sites. Genes Dev 11: 44–58. Li R, Morris SW. (2008). Development of anaplastic lymphoma kinase Ikegaki N, Gotoh T, Kung B, Riceberg JS, Kim DY, Zhao H et al. (ALK) small-molecule inhibitors for cancer therapy. Med Res Rev (2007). De novo identification of MIZ-1 (ZBTB17) encoding a 28: 372–412. MYC-interacting zinc-finger protein as a new favorable neuroblas- Li Z, Van Calcar S, Qu C, Cavenee WK, Zhang MQ, Ren B. (2003). A toma gene. Clin Cancer Res 13: 6001–6009. global transcriptional regulatory role for c-Myc in Burkitt’s Inge TH, Casson LK, Priebe W, Trent JO, Georgeson KE, Miller DM lymphoma cells. Proc Natl Acad Sci USA 100: 8164–8169. et al. (2002). Importance of Sp1 consensus motifs in the MYCN Li Z, Hann S. (2009). The Myc-nucleophosmin-ARF network: a promoter. Surgery 132: 232–238. complex web unveiled. Cell Cycle 8: 2703–2707. Jacobs JFM, van Bokhoven H, van Leeuwen FN, Hulsbergen-van de Lin AW, Lowe SW. (2001). Oncogenic ras activates the ARF-p53 Kaa CA, de Vries IJM, Adema GJ et al. (2009). Regulation pathway to suppress epithelial cell transformation. Proc Natl Acad of MYCN expression in human neuroblastoma cells. BMC Cancer Sci USA 98: 5025–5030. 9: 239. Liu T, Liu PY, Tee AEL, Haber M, Norris MD, Gleave ME et al. Janoueix-Lerosey I, Lequin D, Brugie` res L, Ribeiro A, de Pontual L, (2009). Over-expression of clusterin is a resistance factor to the anti- Combaret V et al. (2008). Somatic and germline activating cancer effect of histone deacetylase inhibitors. Eur J Cancer 45: mutations of the ALK kinase receptor in neuroblastoma. Nature 1846–1854. 455: 967–970. Lutterbach B, Hann SR. (1994). Hierarchical phosphorylation at N- Johnsen JI, Segerstro¨m L, Orrego A, Elfman L, Henriksson M, terminal transformation-sensitive sites in c-Myc protein is regulated Ka˚gedal B et al. (2008). Inhibitors of mammalian target of by mitogens and in mitosis. Mol Cell Biol 14: 5510–5522. rapamycin downregulate MYCN protein expression and inhibit Lutterbach B, Hann SR. (1999). c-Myc transactivation domain- neuroblastoma growth in vitro and in vivo. Oncogene 27: 2910–2922. associated kinases: questionable role for map kinases in c-Myc Kamijo T, Weber JD, Zambetti G, Zindy F, Roussel MF, Sherr CJ. phosphorylation. J Cell Biochem 72: 483–491. (1998). Functional and physical interactions of the ARF Ma XM, Blenis J. (2009). Molecular mechanisms of mTOR-mediated tumor suppressor with p53 and Mdm2. Proc Natl Acad Sci USA translational control. Nat Rev Mol Cell Biol 10: 307–318. 95: 8292–8297. MacDonald BT, Tamai K, He X. (2009). Wnt/beta-catenin signaling: Kanemaru KK, Tuthill MC, Takeuchi KK, Sidell N, Wada RK. components, mechanisms, and diseases. Dev Cell 17: 9–26. (2008). Retinoic acid induced downregulation of MYCN is not Maestro R, Dei Tos AP, Hamamori Y, Krasnokutsky S, Sartorelli V, mediated through changes in Sp1/Sp3. Pediatr Blood Cancer 50: Kedes L et al. (1999). Twist is a potential oncogene that inhibits 806–811. apoptosis. Genes Dev 13: 2207–2217. Kang J, Rychahou PG, Ishola TA, Mourot JM, Evers BM, Chung Manohar CF, Bray JA, Salwen HR, Madafiglio J, Cheng A, Flemming DH. (2008). N-myc is a novel regulator of PI3K-mediated VEGF C et al. (2004). MYCN-mediated regulation of the MRP1 promoter expression in neuroblastoma. Oncogene 27: 3999–4007. in human neuroblastoma. Oncogene 23: 753–762. Kato GJ, Lee WM, Chen LL, Dang CV. (1992). Max: functional Mao DYL, Watson JD, Yan PS, Barsyte-Lovejoy D, Khosravi F, domains and interaction with c-Myc. Genes Dev 6: 81–92. Wong WW-L et al. (2003). Analysis of Myc bound loci identified by Kenney AM, Cole MD, Rowitch DH. (2003). Nmyc upregulation by CpG island arrays shows that Max is essential for Myc-dependent sonic hedgehog signaling promotes proliferation in developing repression. Curr Biol 13: 882–886. cerebellar granule neuron precursors. Development 130: 15–28. Maris JM, Courtright J, Houghton PJ, Morton CL, Kolb EA, Lock R Kenney AM, Widlund HR, Rowitch DH. (2004). Hedgehog and PI-3 et al. (2008a). Initial testing (stage 1) of sunitinib by the pediatric kinase signaling converge on Nmyc1 to promote cell cycle preclinical testing program. Pediatr Blood Cancer 51: 42–48. progression in cerebellar neuronal precursors. Development 131: Maris JM, Mosse YP, Bradfield JP, Hou C, Monni S, Scott RH et al. 217–228. (2008b). Chromosome 6p22 associated with clinically aggres- Keshelava N, Seeger RC, Reynolds CP. (1997). Drug resistance in sive neuroblastoma. N Engl J Med 358: 2585–2593. human neuroblastoma cell lines correlates with clinical therapy. Eur Maris JM. (2009). Unholy matrimony: Aurora A and N-Myc as J Cancer 33: 2002–2006. malignant partners in neuroblastoma. Cancer Cell 15: 5–6. Keshelava N, Seeger RC, Groshen S, Reynolds CP. (1998). Marque´s M, Kumar A, Corte´s I, Gonzalez-Garcı´a A, Herna´ndez C, Drug resistance patterns of human neuroblastoma cell lines Moreno-Ortiz MC et al. (2008). Phosphoinositide 3-kinases derived from patients at different phases of therapy. Cancer Res p110alpha and p110beta regulate cell cycle entry, exhibiting distinct 58: 5396–5405. activation kinetics in G1 phase. Mol Cell Biol 28: 2803–2814. Keshelava N, Zuo JJ, Waidyaratne NS, Triche TJ, Reynolds CP. Martinato F, Cesaroni M, Amati B, Guccione E. (2008). Analysis of (2000). p53 mutations and loss of p53 function confer multidrug Myc-induced histone modifications on target chromatin. PLoS One resistance in neuroblastoma. Med Pediatr Oncol 35: 563–568. 3: e3650. Kim J, Lee J-h, Iyer VR. (2008). Global identification of Myc target Martins RAP, Zindy F, Donovan S, Zhang J, Pounds S, Wey A et al. genes reveals its direct role in mitochondrial biogenesis and its E- (2008). N-myc coordinates retinal growth with eye size during box usage in vivo. PLoS One 3: e1798. mouse development. Genes Dev 22: 179–193. Knoepfler PS, Cheng PF, Eisenman RN. (2002). N-myc is essential Marzec M, Kasprzycka M, Liu X, El-Salem M, Halasa K, Raghunath during neurogenesis for the rapid expansion of progenitor cell PN et al. (2007). Oncogenic tyrosine kinase NPM/ALK induces populations and the inhibition of neuronal differentiation. Genes activation of the rapamycin-sensitive mTOR signaling pathway. Dev 16: 2699–2712. Oncogene 26: 5606–5614.

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1258 McDermott U, Iafrate AJ, Gray NS, Shioda T, Classon M, Park JR, Eggert A, Caron H. (2008). Neuroblastoma: biology, Maheswaran S et al. (2008). Genomic alterations of anaplastic prognosis, and treatment. Pediatr Clin North Am 55: 97–120. lymphoma kinase may sensitize tumors to anaplastic lymphoma Patel JH, Loboda AP, Showe MK, Showe LC, McMahon SB. (2004). kinase inhibitors. Cancer Res 68: 3389–3395. Analysis of genomic targets reveals complex functions of MYC. Nat McMahon SB, Wood MA, Cole MD. (2000). The essential cofactor Rev Cancer 4: 562–568. TRRAP recruits the histone acetyltransferase hGCN5 to c-Myc. Payne GS, Bishop JM, Varmus HE. (1982). Multiple arrangements of Mol Cell Biol 20: 556–562. viral DNA and an activated host oncogene in bursal lymphomas. Memmott RM, Dennis PA. (2009). Akt-dependent and -independent Nature 295: 209–214. mechanisms of mTOR regulation in cancer. Cell Signal 21: 656–664. Peterson RT, Desai BN, Hardwick JS, Schreiber SL. (1999). Protein Meyer N, Penn LZ. (2008). Reflecting on 25 years with MYC. Nat Rev phosphatase 2A interacts with the 70-kDa S6 kinase and is activated Cancer 8: 976–990. by inhibition of FKBP12-rapamycinassociated protein. Proc Natl Midgley CA, Desterro JM, Saville MK, Howard S, Sparks A, Hay RT Acad Sci USA 96: 4438–4442. et al. (2000). An N-terminal p14ARF peptide blocks Mdm2- Prendergast GC, Lawe D, Ziff EB. (1991). Association of dependent ubiquitination in vitro and can activate p53 in vivo. Myn, the murine homolog of , with c-Myc stimulates Oncogene 19: 2312–2323. methylation-sensitive DNA binding and ras cotransformation. Cell Mill P, Mo R, Hu MC, Dagnino L, Rosenblum ND, Hui C-C. (2005). 65: 395–407. Shh controls epithelial proliferation via independent pathways that Pulverer BJ, Fisher C, Vousden K, Littlewood T, Evan G, Woodgett converge on N-Myc. Dev Cell 9: 293–303. JR. (1994). Site-specific modulation of c-Myc cotransformation by Moore HC, Wood KM, Jackson MS, Lastowska MA, Hall D, Imrie H residues phosphorylated in vivo. Oncogene 9: 59–70. et al. (2008). Histological profile of tumours from MYCN Qi Y, Gregory MA, Li Z, Brousal JP, West K, Hann SR. (2004). transgenic mice. J Clin Pathol 61: 1098–1103. p19ARF directly and differentially controls the functions of c-Myc Mosse´Y, Wood A, Maris J. (2009). Inhibition of ALK signaling for independently of p53. Nature 431: 712–717. cancer therapy. Clin Cancer Res 15: 5609–5614. Raabe EH, Laudenslager M, Winter C, Wasserman N, Cole K, Mosse YP, Laudenslager M, Khazi D, Carlisle AJ, Winter CL, Laquaglia M et al. (2008). Prevalence and functional consequence of Rappaport E et al. (2004). Germline PHOX2B mutation in PHOX2B mutations in neuroblastoma. Oncogene 27: 469–476. hereditary neuroblastoma. Am J Hum Genet 75: 727–730. Riley RD, Heney D, Jones DR, Sutton AJ, Lambert PC, Abrams KR Mosse´YP, Laudenslager M, Longo L, Cole KA, Wood A, Attiyeh EF et al. (2004). A systematic review of molecular and biological tumor et al. (2008). Identification of ALK as a major familial neuroblas- markers in neuroblastoma. Clin Cancer Res 10: 4–12. toma predisposition gene. Nature 455: 930–935. Rohrer T, Trachsel D, Engelcke G, Hammer J. (2002). Congenital Mugrauer G, Alt FW, Ekblom P. (1988). N-myc proto-oncogene central hypoventilation syndrome associated with Hirschsprung’s expression during organogenesis in the developing mouse as disease and neuroblastoma: case of multiple neurocristopathies. revealed by in situ hybridization. J Cell Biol 107: 1325–1335. Pediatr Pulmonol 33: 71–76. Murphy DJ, Junttila MR, Pouyet L, Karnezis A, Shchors K, Bui DA Ruggero D. (2009). The Role of Myc-Induced Protein Synthesis in et al. (2008). Distinct thresholds govern Myc’s biological output Cancer. Cancer Res 69: 8839–8843. in vivo. Cancer Cell 14: 447–457. Saksela K, Ma¨kela¨TP, Hughes K, Woodgett JR, Alitalo K. (1992). Norris MD, Burkhart CA, Marshall GM, Weiss WA, Haber M. Activation of protein kinase C increases phosphorylation of the L- (2000). Expression of N-myc and MRP genes and their relationship myc trans-activator domain at a GSK-3 target site. Oncogene 7: to N-myc gene dosage and tumor formation in a murine 347–353. neuroblastoma model. Med Pediatr Oncol 35: 585–589. Schramm A, Schulte JH, Klein-Hitpass L, Havers W, Sieverts H, Oberthuer A, Berthold F, Warnat P, Hero B, Kahlert Y, Spitz R et al. Berwanger B et al. (2005). Prediction of clinical outcome and (2006). Customized oligonucleotide microarray gene expression- biological characterization of neuroblastoma by expression profil- based classification of neuroblastoma patients outperforms current ing. Oncogene 24: 7902–7912. clinical risk stratification. J Clin Oncol 24: 5070–5078. Schramm A, Mierswa I, Kaderali L, Morik K, Eggert A, Schulte JH. Ohira M, Oba S, Nakamura Y, Isogai E, Kaneko S, Nakagawa A et al. (2009). Reanalysis of neuroblastoma expression profiling data using (2005). Expression profiling using a tumor-specific cDNA micro- improved methodology and extended follow-up increases validity of array predicts the prognosis of intermediate risk neuroblastomas. outcome prediction. Cancer Lett 282: 55–62. Cancer Cell 7: 337–350. Schulte JH, Horn S, Schlierf S, Schramm A, Heukamp LC, Okubo T, Knoepfler PS, Eisenman RN, Hogan BLM. (2005). Nmyc Christiansen H et al. (2009). MicroRNAs in the pathogenesis of plays an essential role during lung development as a dosage-sensitive neuroblastoma. Cancer Lett 274: 10–15. regulator of progenitor cell proliferation and differentiation. Sears R, Leone G, DeGregori J, Nevins JR. (1999). Ras enhances Myc Development 132: 1363–1374. protein stability. Mol Cell 3: 169–179. Oliner JD, Pietenpol JA, Thiagalingam S, Gyuris J, Kinzler KW, Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. (2000). Vogelstein B. (1993). Oncoprotein MDM2 conceals the activation Multiple Ras-dependent phosphorylation pathways regulate Myc domain of tumour suppressor p53. Nature 362: 857–860. protein stability. Genes Dev 14: 2501–2514. Oliver TG, Grasfeder LL, Carroll AL, Kaiser C, Gillingham CL, Lin Seeger RC, Brodeur GM, Sather H, Dalton A, Siegel SE, Wong KY SM et al. (2003). Transcriptional profiling of the Sonic hedgehog et al. (1985). Association of multiple copies of the N-myc oncogene response: a critical role for N-myc in proliferation of neuronal with rapid progression of neuroblastomas. N Engl J Med 313: precursors. Proc Natl Acad Sci USA 100: 7331–7336. 1111–1116. Opel D, Poremba C, Simon T, Debatin K-M, Fulda S. (2007). Seoane J, Pouponnot C, Staller P, Schader M, Eilers M, Massague´J. Activation of Akt predicts poor outcome in neuroblastoma. Cancer (2001). TGFbeta influences Myc, Miz-1 and Smad to control the Res 67: 735–745. CDK inhibitor p15INK4b. Nat Cell Biol 3: 400–408. Ota S, Zhou Z-Q, Keene DR, Knoepfler P, Hurlin PJ. (2007). Shang X, Burlingame S, Okcu M, Ge N, Russell H, Egler R et al. Activities of N-Myc in the developing limb link control of skeletal (2009). Aurora A is a negative prognostic factor and a new size with digit separation. Development 134: 1583–1592. therapeutic target in human neuroblastoma. Mol Cancer Ther 8: Otto T, Horn S, Brockmann M, Eilers U, Schu¨ttrumpf L, Popov N 2461–2469. et al. (2009). Stabilization of N-Myc is a critical function of Aurora Shi Y, Glynn JM, Guilbert LJ, Cotter TG, Bissonnette RP, Green DR. A in human neuroblastoma. Cancer Cell 15: 67–78. (1992). Role for c-myc in activation-induced apoptotic cell death in Paffhausen T, Schwab M, Westermann F. (2007). Targeted MYCN T cell hybridomas. Science 257: 212–214. expression affects cytotoxic potential of chemotherapeutic drugs in Sjostrom SK, Finn G, Hahn WC, Rowitch DH, Kenney AM. (2005). neuroblastoma cells. Cancer Lett 250: 17–24. The Cdk1 complex plays a prime role in regulating N-myc

Oncogene Mycn signaling in neuroblastoma WC Gustafson and WA Weiss 1259 phosphorylation and turnover in neural precursors. Dev Cell 9: Walkley CR, Fero ML, Chien W-M, Purton LE, McArthur GA. 327–338. (2005). Negative cell-cycle regulators cooperatively control self- Slack A, Chen Z, Tonelli R, Pule M, Hunt L, Pession A et al. (2005a). renewal and differentiation of haematopoietic stem cells. Nat Cell The p53 regulatory gene MDM2 is a direct transcriptional target of Biol 7: 172–178. MYCN in neuroblastoma. Proc Natl Acad Sci USA 102: 731–736. Wang W, Kim S-H, El-Deiry WS. (2006). Small-molecule modulators Slack A, Lozano G, Shohet JM. (2005b). MDM2 as MYCN of p53 family signaling and antitumor effects in p53-deficient human transcriptional target: implications for neuroblastoma pathogenesis. colon tumor xenografts. Proc Natl Acad Sci USA 103: 11003–11008. Cancer Lett 228: 21–27. Weber JD, Taylor LJ, Roussel MF, Sherr CJ, Bar-Sagi D. (1999). Smith-Srensen B, Hijmans EM, Beijersbergen RL, Bernards R. Nucleolar Arf sequesters Mdm2 and activates p53. Nat Cell Biol 1: (1996). Functional analysis of Burkitt’s lymphoma mutant c-Myc 20–26. proteins. J Biol Chem 271: 5513–5518. Weiss WA, Godfrey T, Francisco C, Bishop JM. (2000). Genome-wide Staller P, Peukert K, Kiermaier A, Seoane J, Lukas J, Karsunky H screen for allelic imbalance in a mouse model for neuroblastoma. et al. (2001). Repression of p15INK4b expression by Myc through Cancer Res 60: 2483–2487. association with Miz-1. Nat Cell Biol 3: 392–399. Welcker M, Orian A, Jin J, Grim JE, Grim JA, Harper JW et al. Stupack DG, Teitz T, Potter MD, Mikolon D, Houghton PJ, Kidd VJ (2004). The Fbw7 tumor suppressor regulates glycogen synthase et al. (2006). Potentiation of neuroblastoma metastasis by loss of kinase 3 phosphorylation-dependent c-Myc protein degradation. caspase-8. Nature 439: 95–99. Proc Natl Acad Sci USA 101: 9085–9090. Tang XX, Zhao H, Kung B, Kim DY, Hicks SL, Cohn SL et al. Westermann F, Muth D, Benner A, Bauer T, Henrich K, Oberthuer A (2006). The MYCN enigma: significance of MYCN expression in et al (2008). Distinct transcriptional MYCN/c-MYC activities are neuroblastoma. Cancer Res 66: 2826–2833. associated with spontaneous regression or malignant progression in Thiele CJ, Reynolds CP, Israel MA. (1985). Decreased expression of neuroblastomas. Genome Biol 9: R150. N-myc precedes retinoic acid-induced morphological differentiation Whitehead KA, Langer R, Anderson DG. (2009). Knocking of human neuroblastoma. Nature 313: 404–406. down barriers: advances in siRNA delivery. Nat Rev Drug Discov Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y et al. 8: 129–138. (2009). An ATP-competitive mammalian target of rapamycin Witt O, Deubzer HE, Lodrini M, Milde T, Oehme I. (2009). inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Targeting histone deacetylases in neuroblastoma. Curr Pharm Des Chem 284: 8023–8032. 15: 436–447. Thut CJ, Goodrich JA, Tjian R. (1997). Repression of p53-mediated Xu XL, Fang Y, Lee TC, Forrest D, Gregory-Evans C, Almeida D transcription by MDM2: a dual mechanism. Genes Dev 11: et al (2009). Retinoblastoma has properties of a cone precursor 1974–1986. tumor and depends upon cone-specific MDM2 signaling. Cell 137: Trochet D, O’Brien LM, Gozal D, Trang H, Nordenskjo¨ld A, Laudier 1018–1031. B et al. (2005). PHOX2B genotype allows for prediction of tumor Xue C, Haber M, Flemming C, Marshall GM, Lock RB, MacKenzie risk in congenital central hypoventilation syndrome. Am J Hum KL et al (2007). p53 determines multidrug sensitivity of childhood Genet 76: 421–426. neuroblastoma. Cancer Res 67: 10351–10360. Tweddle DA, Malcolm AJ, Bown N, Pearson AD, Lunec J. (2001). Yada M, Hatakeyama S, Kamura T, Nishiyama M, Tsunematsu R, Evidence for the development of p53 mutations after cytotoxic Imaki H et al (2004). Phosphorylation-dependent degradation therapy in a neuroblastoma cell line. Cancer Res 61: 8–13. of c-Myc is mediated by the F-box protein Fbw7. EMBO J 23: Ushmorov A, Hogarty MD, Liu X, Knauss H, Debatin KM, Beltinger 2116–2125. C. (2008). N-myc augments death and attenuates protective effects Yeh E, Cunningham M, Arnold H, Chasse D, Monteith T, Ivaldi G of Bcl-2 in trophically stressed neuroblastoma cells. Oncogene 27: et al (2004). A signalling pathway controlling c-Myc degradation 3424–3434. that impacts oncogenic transformation of human cells. Nat Cell Biol Valsesia-Wittmann S, Magdeleine M, Dupasquier S, Garin E, Jallas A- 6: 308–318. C, Combaret V et al. (2004). Oncogenic cooperation between H- Yin X, Giap C, Lazo JS, Prochownik EV. (2003). Low molecular Twist and N-Myc overrides failsafe programs in cancer cells. Cancer weight inhibitors of Myc-Max interaction and function. Oncogene Cell 6: 625–630. 22: 6151–6159. Van Maerken T, Ferdinande L, Taildeman J, Lambertz I, Yigit N, Yu K, Toral-Barza L, Shi C, Zhang W-G, Lucas J, Shor B et al (2009). Vercruysse L et al. (2009a). Antitumor activity of the selective Biochemical, cellular, and in vivo activity of novel ATP-competitive MDM2 antagonist Nutlin-3 against chemoresistant neuroblastoma and selective inhibitors of the mammalian target of rapamycin. with wild-type p53. J Natl Cancer Inst 101: 1562–1574. Cancer Res 69: 6232–6240. Van Maerken T, Vandesompele J, Rihani A, De Paepe A, Speleman F. Zask A, Verheijen J, Curran K, Kaplan J, Richard D, Nowak P et al (2009b). Escape from p53-mediated tumor surveillance in neuro- (2009). ATP-competitive inhibitors of the mammalian target of blastoma: switching off the p14(ARF)-MDM2-p53 axis. Cell Death rapamycin: design and synthesis of highly potent and selective Differ 16: 1563–1572. pyrazolopyrimidines. J Med Chem 52: 5013–5016. Vermeulen J, De Preter K, Naranjo A, Vercruysse L, Van Roy N, Zeller KI, Zhao X, Lee CWH, Chiu KP, Yao F, Yustein JT et al Hellemans J et al. (2009). Predicting outcomes for children (2006). Global mapping of c-Myc binding sites and target with neuroblastoma using a multigene-expression signature: a gene networks in human B cells. Proc Natl Acad Sci USA 103: retrospective SIOPEN/COG/GPOH study. Lancet Oncol 10: 17834–17839. 663–671. Zervos AS, Gyuris J, Brent R. (1993). Mxi1, a protein that specifically Vervoorts J, Lu¨scher-Firzlaff JM, Rottmann S, Lilischkis R, interacts with Max to bind Myc-Max recognition sites. Cell 72: Walsemann G, Dohmann K et al. (2003). Stimulation of c-MYC 223–232. transcriptional activity and acetylation by recruitment of the Zimmerman KA, Yancopoulos GD, Collum RG, Smith RK, Kohl cofactor CBP. EMBO Rep 4: 484–490. NE, Denis KA et al (1986). Differential expression of myc family Vogan K, Bernstein M, Leclerc JM, Brisson L, Brossard J, Brodeur genes during murine development. Nature 319: 780–783. GM et al. (1993). Absence of p53 gene mutations in primary Zindy F, Eischen CM, Randle DH, Kamijo T, Cleveland JL, Sherr CJ neuroblastomas. Cancer Res 53: 5269–5273. et al (1998). Myc signaling via the ARF tumor suppressor Wagner LM, Danks MK. (2009). New therapeutic targets for the regulates p53-dependent apoptosis and immortalization. Genes treatment of high-risk neuroblastoma. J Cell Biochem 107: 46–57. Dev 12: 2424–2433.

Oncogene