A chemical screen identifies the chemotherapeutic drug topotecan as a specific inhibitor of the B-MYB/MYCN axis in neuroblastoma

Francesco Sottile, Ilaria Gnemmi, Sandra Cantilena, Walter C D'Acunto, Arturo Sala, Francesco Sottile, Ilaria Gnemmi, Sandra Cantilena, Walter C D'Acunto, Arturo Sala

Abstract

The transcription factor MycN is the prototypical neuroblastoma oncogene and a potential therapeutic target. However, its strong expression caused by gene amplification in about 30% of neuroblastoma patients is a considerable obstacle to the development of therapeutic approaches aiming at eliminating its tumourigenic activity. We have previously reported that B-Myb is essentially required for transcription of the MYCN amplicon and have also shown that B-MYB and MYCN are engaged in a feed forward loop promoting the survival/proliferation of neuroblastoma cells. We postulated that pharmacological strategies breaking the B-MYB/MYCN axis should result in clinically desirable effects. Thus, we implemented a high throughput chemical screen, using a curated library of ~1500 compounds from the National Cancer Institute, whose endpoint was the identification of small molecules that inhibited B-Myb. At the end of the screening, we found that the compounds pinafide, ellipticine and camptothecin inhibited B-Myb transcriptional activity in luciferase assays. One of the compounds, the topoisomerase-1 inhibitor camptothecin, is of considerable clinical interest since its derivatives topotecan and irinotecan are currently used as first and second line treatment agents for various types of cancer, including neuroblastoma. We found that neuroblastoma cells with amplification of MYCN are more sensitive than MYCN negative cells to camptothecin and topotecan killing. Campothecin and topotecan caused selective down-regulation of B-Myb and MycN expression in neuroblastoma cells. Notably, forced overexpression of B-Myb could antagonize the killing effect of topotecan and camptothecin, demonstrating that the transcription factor is a key target of the drugs. These results suggest that camptothecin and its analogues should be more effective in patients whose tumours feature amplification of MYCN and/or overexpression of B-MYB.

Figures

Figure 1. Chemical screen
Figure 1. Chemical screen
Each dot represents relative luciferase activity (RLU) expressed as percentages relative to the value obtained in cells treated with the vehicle DMSO (indicated by the black diamond and ctrl), which was set as 100%. The highlighted compound codes 50N E8, 50N G9, and 63 F6 indicate ellipticine, camptothecin and pinafide, respectively.
Figure 2. Validation of compounds identified in…
Figure 2. Validation of compounds identified in the primary screen
GIMEN (MYCN non-amplified neuroblastoma cells) were transiently co-transfected with the MYB-responsive promoter pGL2-mim1 and the pcDna-B-MYB/empty vectors or with the p53 responsive p21luc promoter and pCMVp53/empty vectors. A renilla luciferase plasmid was added to the transfection mix for normalisation. Cells were treated with the compounds ellipticine, pinafide and camptothecin at the indicated concentrations for 24h. Error bars indicate standard deviations and the asterisk indicates statistically significant differences (Student's T test p ≤ 0,05) between the activities of cells treated with the compound relative to untreated cells (indicated by ctrl).
Figure 3. MTS proliferation assay in neuroblastoma…
Figure 3. MTS proliferation assay in neuroblastoma cell lines exposed to ellipticine and pinafide
Proliferation of MYCN amplified (panel A) or MYCN non amplified (panel B) cells in the presence of increasing concentrations of Ellipticine and Pinafide, as indicated. Note that cell viability is only reduced by high concentrations (250-500nM) of pinafide, in a MYCN-independent manner. Neuroblastoma cell lines were generally resistant to the killing effect of ellipticine. Error bars indicate standard deviations.
Figure 4. MTS proliferation assay in neuroblastoma…
Figure 4. MTS proliferation assay in neuroblastoma cell lines exposed to camptothecin (CPT) or topotecan (TPT)
A) MYCN-amplified NB cells; B) MYCN non-amplified NB cells. Error bars indicate standard deviations and statistical significance was verified by Student's T test (*= p<0,05 **=p<0,01 ***=p<0,001 of 48hrs 10v20v250nM vs. CTR; 72h 10v20v250nM vs. CTR )
Figure 5. Effects of camptothecin (CPT) and…
Figure 5. Effects of camptothecin (CPT) and topotecan (TPT) on the B-MYB/MYCN axis
Western blot analysis showing the expression of B-Myb and MycN in the indicated neuroblastoma cell lines treated with CPT or TPT (20nM) for 8-16-24-32hrs. Expression of the housekeeping gene GAPDH was used as loading control.
Figure 6. Effects of camptothecin (CPT) and…
Figure 6. Effects of camptothecin (CPT) and topotecan (TPT) on cell cycle proteins and PARP cleavage
Western blot analysis showing the expression of Cyclin A and cyclin B in the indicated neuroblastoma cell lines treated with CPT or TPT (20nM) for 8-16-24-32hrs. Cleavage of PARP is indicated by the appearance of a 89KDa fragment. The housekeeping gene GAPDH was used as loading control.
Figure 7. B-MYB rescues the killing effect…
Figure 7. B-MYB rescues the killing effect of camptothecin (CPT) and topotecan (TPT) in MYCN amplified cells
MTS proliferation assay showing the effect of CPT and TPT in LAN-5 cell clones transfected with pCMV-empty or pCMV-B-MYB plasmids. Bars indicate densitometric values relative to vehicle treated controls. Standard deviations is indicated by the error bars, statistical significance was assessed using the Student's T-test (*= p<0,05; **=p<0,01; ***=p<0,001; o= p<0,05 oo=p<0,01; ooo=p<0,001;)

References

    1. Schwab M, Westermann F, Hero B, Berthold F. Neuroblastoma: biology and molecular and chromosomal pathology. Lancet Oncol. 2003;4:472–480.
    1. Maris JM, Hogarty MD, Bagatell R, Cohn SL. Neuroblastoma. Lancet. 2007;369:2106–2120.
    1. Brodeur GM, Hayes FA, Green AA, Casper JT, Wasson J, Wallach S, Seeger RC. Consistent N-myc copy number in simultaneous or consecutive neuroblastoma samples from sixty individual patients. Cancer Res. 1987;47:4248–4253.
    1. Brodeur GM, Seeger RC. Gene amplification in human neuroblastomas: basic mechanisms and clinical implications. Cancer Genet Cytogenet. 1986;19:101–111.
    1. Brodeur GM, Seeger RC, Schwab M, Varmus HE, Bishop JM. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science. 1984;224:1121–1124.
    1. Seeger RC, Brodeur GM, Sather H, Dalton A, Siegel SE, Wong KY, Hammond D. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N Engl J Med. 1985;313:1111–1116.
    1. Perez CA, Matthay KK, Atkinson JB, Seeger RC, Shimada H, Haase GM, Stram DO, Gerbing RB, Lukens JN. Biologic variables in the outcome of stages I and II neuroblastoma treated with surgery as primary therapy: a children's cancer group study. J Clin Oncol. 2000;18:18–26.
    1. Cohn SL, Look AT, Joshi VV, Holbrook T, Salwen H, Chagnovich D, Chesler L, Rowe ST, Valentine MB, Komuro H, et al. Lack of correlation of N-myc gene amplification with prognosis in localized neuroblastoma: a Pediatric Oncology Group study. Cancer Res. 1995;55:721–726.
    1. Katzenstein HM, Bowman LC, Brodeur GM, Thorner PS, Joshi VV, Smith EI, Look AT, Rowe ST, Nash MB, Holbrook T, Alvarado C, Rao PV, Castleberry RP, Cohn SL. Prognostic significance of age, MYCN oncogene amplification, tumor cell ploidy, and histology in 110 infants with stage D(S) neuroblastoma: the pediatric oncology group experience--a pediatric oncology group study. J Clin Oncol. 1998;16:2007–2017.
    1. Galderisi U, Di Bernardo G, Cipollaro M, Peluso G, Cascino A, Cotrufo R, Melone MA. Differentiation and apoptosis of neuroblastoma cells: role of N-myc gene product. J Cell Biochem. 1999;73:97–105.
    1. Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer. 2008;8:976–990.
    1. Schwab M, Varmus HE, Bishop JM, Grzeschik KH, Naylor SL, Sakaguchi AY, Brodeur G, Trent J. Chromosome localization in normal human cells and neuroblastomas of a gene related to c-myc. Nature. 1984;308:288–291.
    1. Brodeur GM, Fong CT. Molecular biology and genetics of human neuroblastoma. Cancer Genet Cytogenet. 1989;41:153–174.
    1. Corvi R, Amler LC, Savelyeva L, Gehring M, Schwab M. MYCN is retained in single copy at chromosome 2 band p23-24 during amplification in human neuroblastoma cells. Proc Natl Acad Sci U S A. 1994;91:5523–5527.
    1. Schneider SS, Hiemstra JL, Zehnbauer BA, Taillon-Miller P, Le Paslier DL, Vogelstein B, Brodeur GM. Isolation and structural analysis of a 1.2-megabase N-myc amplicon from a human neuroblastoma. Mol Cell Biol. 1992;12:5563–5570.
    1. Graf T. Myb: a transcriptional activator linking proliferation and differentiation in hematopoietic cells. Curr Opin Genet Dev. 1992;2:249–255.
    1. Nomura N, Takahashi M, Matsui M, Ishii S, Date T, Sasamoto S, Ishizaki R. Isolation of human cDNA clones of myb-related genes, A-myb and B-myb. Nucleic Acids Res. 1988;16:11075–11089.
    1. Sakura H, Kanei-Ishii C, Nagase T, Nakagoshi H, Gonda TJ, Ishii S. Delineation of three functional domains of the transcriptional activator encoded by the c-myb protooncogene. Proc Natl Acad Sci U S A. 1989;86:5758–5762.
    1. Nakagoshi H, Takemoto Y, Ishii S. Functional domains of the human B-myb gene product. J Biol Chem. 1993;268:14161–14167.
    1. Golay J, Capucci A, Arsura M, Castellano M, Rizzo V, Introna M. Expression of c-myb and B-myb, but not A-myb, correlates with proliferation in human hematopoietic cells. Blood. 1991;77:149–158.
    1. Reiss K, Travali S, Calabretta B, Baserga R. Growth regulated expression of B-myb in fibroblasts and hematopoietic cells. J Cell Physiol. 1991;148:338–343.
    1. Lam EW, Robinson C, Watson RJ. Characterization and cell cycle-regulated expression of mouse B-myb. Oncogene. 1992;7:1885–1890.
    1. Sitzmann J, Noben-Trauth K, Kamano H, Klempnauer KH. Expression of B-Myb during mouse embryogenesis. Oncogene. 1996;12:1889–1894.
    1. Tanaka Y, Patestos NP, Maekawa T, Ishii S. B-myb is required for inner cell mass formation at an early stage of development. J Biol Chem. 1999;274:28067–28070.
    1. Arsura M, Introna M, Passerini F, Mantovani A, Golay J. B-myb antisense oligonucleotides inhibit proliferation of human hematopoietic cell lines. Blood. 1992;79:2708–2716.
    1. Sala A, Calabretta B. Regulation of BALB/c 3T3 fibroblast proliferation by B-myb is accompanied by selective activation of cdc2 and cyclin D1 expression. Proc Natl Acad Sci U S A. 1992;89:10415–10419.
    1. Bies J, Hoffman B, Amanullah A, Giese T, Wolff L. B-Myb prevents growth arrest associated with terminal differentiation of monocytic cells. Oncogene. 1996;12:355–363.
    1. Gualdrini F, Corvetta D, Cantilena S, Chayka O, Tanno B, Raschella G, Sala A. Addiction of MYCN amplified tumours to B-MYB underscores a reciprocal regulatory loop. Oncotarget. 2010;1:278–288.
    1. Fosse P, Rene B, Charra M, Paoletti C, Saucier JM. Stimulation of topoisomerase II-mediated DNA cleavage by ellipticine derivatives: structure-activity relationship. Mol Pharmacol. 1992;42:590–595.
    1. Monnot M, Mauffret O, Simon V, Lescot E, Psaume B, Saucier JM, Charra M, Belehradek J, Jr., Fermandjian S. DNA-drug recognition and effects on topoisomerase II-mediated cytotoxicity. A three-mode binding model for ellipticine derivatives. J Biol Chem. 1991;266:1820–1829.
    1. Froelich-Ammon SJ, Patchan MW, Osheroff N, Thompson RB. Topoisomerase II binds to ellipticine in the absence or presence of DNA. Characterization of enzyme-drug interactions by fluorescence spectroscopy. J Biol Chem. 1995;270:14998–15004.
    1. Pommier Y, Pourquier P, Fan Y, Strumberg D. Mechanism of action of eukaryotic DNA topoisomerase I and drugs targeted to the enzyme. Biochim Biophys Acta. 1998;1400:83–105.
    1. Li TK, Liu LF. Tumor cell death induced by topoisomerase-targeting drugs. Annu Rev Pharmacol Toxicol. 2001;41:53–77.
    1. Staker BL, Hjerrild K, Feese MD, Behnke CA, Burgin AB, Jr., Stewart L. The mechanism of topoisomerase I poisoning by a camptothecin analog. Proc Natl Acad Sci U S A. 2002;99:15387–15392.
    1. Ioanoviciu A, Antony S, Pommier Y, Staker BL, Stewart L, Cushman M. Synthesis and mechanism of action studies of a series of norindenoisoquinoline topoisomerase I poisons reveal an inhibitor with a flipped orientation in the ternary DNA-enzyme-inhibitor complex as determined by X-ray crystallographic analysis. J Med Chem. 2005;48:4803–4814.
    1. Capranico G, Binaschi M, Borgnetto ME, Zunino F, Palumbo M. A protein-mediated mechanism for the DNA sequence-specific action of topoisomerase II poisons. Trends Pharmacol Sci. 1997;18:323–329.
    1. Nitschke R, Parkhurst J, Sullivan J, Harris MB, Bernstein M, Pratt C. Topotecan in pediatric patients with recurrent and progressive solid tumors: a Pediatric Oncology Group phase II study. J Pediatr Hematol Oncol. 1998;20:315–318.
    1. De Falco G, Bagella L, Claudio PP, De Luca A, Fu Y, Calabretta B, Sala A, Giordano A. Physical interaction between CDK9 and B-Myb results in suppression of B-Myb gene autoregulation. Oncogene. 2000;19:373–379.
    1. Gilbert F, Feder M, Balaban G, Brangman D, Lurie DK, Podolsky R, Rinaldt V, Vinikoor N, Weisband J. Human neuroblastomas and abnormalities of chromosomes 1 and 17. Cancer Res. 1984;44:5444–5449.
    1. Mannefeld M, Klassen E, Gaubatz S. B-MYB is required for recovery from the DNA damage-induced G2 checkpoint in p53 mutant cells. Cancer Res. 2009;69:4073–4080.
    1. Down CF, Millour J, Lam EW, Watson RJ. Binding of FoxM1 to G2/M gene promoters is dependent upon B-Myb. Biochim Biophys Acta. 2012
    1. Vassal G, Pondarre C, Boland I, Cappelli C, Santos A, Thomas C, Lucchi E, Imadalou K, Pein F, Morizet J, Gouyette A. Preclinical development of camptothecin derivatives and clinical trials in pediatric oncology. Biochimie. 1998;80:271–280.
    1. Estlin EJ, Veal GJ. Clinical and cellular pharmacology in relation to solid tumours of childhood. Cancer Treat Rev. 2003;29:253–273.
    1. Mairs RJ, Boyd M. Preclinical assessment of strategies for enhancement of metaiodobenzylguanidine therapy of neuroendocrine tumors. Semin Nucl Med. 2011;41:334–344.
    1. Tavner F, Frampton J, Watson RJ. Targeting an E2F site in the mouse genome prevents promoter silencing in quiescent and post-mitotic cells. Oncogene. 2007;26:2727–2735.

Source: PubMed

3
Subscribe