Combined HAT/EZH2 modulation leads to cancer-selective cell death(284 views) Petraglia F, Singh AA, Carafa V, Nebbioso A, Conte M, Scisciola L, Valente S, Baldi A, Mandoli A, Petrizzi VB, Ingenito C, De Falco S, Cicatiello V, Apicella I, Janssen-megens EM, Kim B, Yi G, Logie C, Heath S, Ruvo M, Wierenga ATJ, Flicek P, Yaspo ML, Della Valle V, Bernard O, Tomassi S, Novellino E, Feoli A, Sbardella G, Gut I, Vellenga E, Stunnenberg HG, Mai A, Martens JHA, Altucci L
Oncotarget (ISSN: 1949-2553electronic, 1949-2553linking), 2018 May 22; 9(39): 25630-25646.
Keywords: Acetylation, Apoptosis, Cancer, Epigenetics, Methylation, Antineoplastic Agent, Caspase, Mc 2884, Transcription Factor Ezh2, Unclassified Drug, Acute Myeloid Leukemia, Animal Experiment, Animal Model, Antineoplastic Activity, Antiproliferative Activity, Article, Cancer Cell, Cell Cycle, Cell Death, Cell Growth, Cell Migration, Cell Proliferation, Colon Carcinoma, Controlled Study, Cytotoxicity, Drug Effect, Drug Efficacy, Drug Mechanism, Drug Response, Drug Structure, Human, Human Cell, Mouse, Nonhuman, Tumor Volume, Tumor Xenograft,
Affiliations: *** IBB - CNR ***
Dipartimento di Medicina di Precisione, Universita degli Studi della Campania Luigi Vanvitelli, Napoli 80138, Italy., Department of Molecular Biology, Radboud University, HB Nijmegen 6500, The Netherlands., IRCCS SDN, Napoli 80143, Italy., Dipartimento di Chimica e Tecnologie del Farmaco 'Sapienza' Universita, Roma 00185, Italy., Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Universita della Campania 'Luigi Vanvitelli', Caserta 81100, Italy., Ospedale Umberto I, Nocera Inferiore 84014, Italy., Istituto di Genetica e Biofisica, Napoli 80131, Italy., Centro Nacional de Analisis Genomico, Barcelona, Spain., Istituto di Biostrutture e Bioimmagini, Napoli, Italy., Department of Hematology, University of Groningen and University Medical Center Groningen, RB Groningen 9700, The Netherlands., European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom., Max Planck Institute for Molecular Genetics, Berlin, Germany., Institute Gustave Roussy, Equipe labellisee Ligue Nationale contre le Cancer (LNCC), Universtite Paris-Saclay, INSERM U1170, Paris, France., Dipartimento di Farmacia, Universita di Napoli 'Federico II', Napoli 80131, Italy., Dipartimento di Farmacia, Universita degli Studi di Salerno, Fisciano I-84084, Italy., Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, Roma 00185, Italy.,
IRCCS SDN, Napoli, 80143, Italy
References: Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia (2013) N Engl J Med, 368, pp. 2059-2074. , https://doi.org/10.1056/NEJMoa130168
Weinstein, J.N., Collisson, E.A., Mills, G.B., Shaw, K.R., Ozenberger, B.A., Ellrott, K., Shmulevich, I., Stuart, J.M., The Cancer Genome Atlas Pan-Cancer analysis project (2013) Nat Genet, 45, pp. 1113-1120. , https://doi.org/10.1038/ng.2764
Sun, X.J., Man, N., Tan, Y., Nimer, S.D., Wang, L., The Role of Histone Acetyltransferases in Normal and Malignant Hematopoiesis (2015) Front Oncol, 5, p. 108. , https://doi.org/10.3389/fonc.2015.00108
Avvakumov, N., Cote, J., The MYST family of histone acetyltransferases and their intimate links to cancer (2007) Oncogene, 26, pp. 5395-5407. , https://doi.org/10.1038/sj.onc.1210608
Greer, E.L., Shi, Y., Histone methylation: a dynamic mark in health, disease and inheritance (2012) Nat Rev Genet, 13, pp. 343-357. , https://doi.org/10.1038/nrg3173
Falkenberg, K.J., Johnstone, R.W., Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders (2014) Nat Rev Drug Discov, 13, pp. 673-691. , https://doi.org/10.1038/nrd4360
Parry, L., Clarke, A.R., The Roles of the Methyl-CpG Binding Proteins in Cancer (2011) Genes Cancer, 2, pp. 618-630. , https://doi.org/10.1177/1947601911418499
Kolla, V., Zhuang, T., Higashi, M., Naraparaju, K., Brodeur, G.M., Role of CHD5 in human cancers: 10 years later (2014) Cancer Res, 74, pp. 652-658. , https://doi.org/10.1158/0008-5472.CAN-13-3056
Jariwala, N., Rajasekaran, D., Srivastava, J., Gredler, R., Akiel, M.A., Robertson, C.L., Emdad, L., Sarkar, D., Role of the staphylococcal nuclease and tudor domain containing 1 in oncogenesis (review) (2015) Int J Oncol, 46, pp. 465-473. , https://doi.org/10.3892/ijo.2014.2766
Musselman, C.A., Kutateladze, T.G., PHD fingers: epigenetic effectors and potential drug targets (2009) Mol Interv, 9, pp. 314-323. , https://doi.org/10.1124/mi.9.6.7
Conte, M., Altucci, L., Molecular pathways: the complexity of the epigenome in cancer and recent clinical advances (2012) Clin Cancer Res, 18, pp. 5526-5534. , https://doi.org/10.1158/1078-0432.CCR-12-2037
You, J.S., Jones, P.A., Cancer genetics and epigenetics: two sides of the same coin? (2012) Cancer Cell, 22, pp. 9-20. , https://doi.org/10.1016/j.ccr.2012.06.008
Shen, H., Laird, P.W., Interplay between the cancer genome and epigenome (2013) Cell, 153, pp. 38-55. , https://doi.org/10.1016/j.cell.2013.03.008
Mandoli, A., Singh, A.A., Prange, K.H., Tijchon, E., Oerlemans, M., Dirks, R., Ter Huurne, M., Matarese, F., The Hematopoietic Transcription Factors RUNX1 and ERG Prevent AML1-ETO Oncogene Overexpression and Onset of the Apoptosis Program in t(8
21) AMLs (2016) Cell Rep, 17, pp. 2087-2100. , https://doi.org/10.1016/j.celrep.2016.08.082
Papaemmanuil, E., Gerstung, M., Bullinger, L., Gaidzik, V.I., Paschka, P., Roberts, N.D., Potter, N.E., Martincorena, I., Genomic Classification and Prognosis in Acute Myeloid Leukemia (2016) N Engl J Med, 374, pp. 2209-2221. , https://doi.org/10.1056/NEJMoa1516192
Queiros, A.C., Beekman, R., Vilarrasa-Blasi, R., Duran-Ferrer, M., Clot, G., Merkel, A., Raineri, E., Verdaguer-Dot, N., Decoding the DNA Methylome of Mantle Cell Lymphoma in the Light of the Entire B Cell Lineage (2016) Cancer Cell, 30, pp. 806-821. , https://doi.org/10.1016/j.ccell.2016.09.014
Nebbioso, A., Benedetti, R., Conte, M., Iside, C., Altucci, L., Genetic mutations in epigenetic modifiers as therapeutic targets in acute myeloid leukemia (2015) Expert Opin Ther Targets, 19, pp. 1187-1202. , https://doi.org/10.1517/14728222.2015.1051728
Nebbioso, A., Carafa, V., Benedetti, R., Altucci, L., Trials with 'epigenetic' drugs: an update (2012) Mol Oncol, 6, pp. 657-682. , https://doi.org/10.1016/j.molonc.2012.09.004
Baylin, S.B., Jones, P.A., A decade of exploring the cancer epigenome - biological and translational implications (2011) Nat Rev Cancer, 11, pp. 726-734. , https://doi.org/10.1038/nrc3130
Laubach, J.P., Moreau, P., San-Miguel, J.F., Richardson, P.G., Panobinostat for the Treatment of Multiple Myeloma (2015) Clin Cancer Res, 21, pp. 4767-4773. , https://doi.org/10.1158/1078-0432.CCR-15-0530
Wu, H., Zeng, H., Dong, A., Li, F., He, H., Senisterra, G., Seitova, A., Schapira, M., Structure of the catalytic domain of EZH2 reveals conformational plasticity in cofactor and substrate binding sites and explains oncogenic mutations (2013) PLoS One, 8. , https://doi.org/10.1371/journal.pone.0083737
Wagener, N., Macher-Goeppinger, S., Pritsch, M., Husing, J., Hoppe-Seyler, K., Schirmacher, P., Pfitzenmaier, J., Hohenfellner, M., Enhancer of zeste homolog 2 (EZH2) expression is an independent prognostic factor in renal cell carcinoma (2010) BMC Cancer, 10, p. 524. , https://doi.org/10.1186/1471-2407-10-524
Antonysamy, S., Condon, B., Druzina, Z., Bonanno, J.B., Gheyi, T., Zhang, F., MacEwan, I., Gately Luz, J., Structural context of disease-associated mutations and putative mechanism of autoinhibition revealed by X-ray crystallographic analysis of the EZH2-SET domain (2013) PLoS One, 8. , https://doi.org/10.1371/journal.pone.0084147
Dillon, S.C., Zhang, X., Trievel, R.C., Cheng, X., The SET-domain protein superfamily: protein lysine methyltransferases (2005) Genome Biol, 6, p. 227. , https://doi.org/10.1186/gb-2005-6-8-227
Franci, G., Sarno, F., Nebbioso, A., Altucci, L., Identification and characterization of PKF118-310 as a KDM4A inhibitor (2016) Epigenetics, , https://doi.org/10.1080/15592294.2016.1249089
Morera, L., Lubbert, M., Jung, M., Targeting histone methyltransferases and demethylases in clinical trials for cancer therapy (2016) Clin Epigenetics, 8, p. 57. , https://doi.org/10.1186/s13148-016-0223-4
Valente, S., Lepore, I., Dell'Aversana, C., Tardugno, M., Castellano, S., Sbardella, G., Tomassi, S., Mai, A., Identification of PR-SET7 and EZH2 selective inhibitors inducing cell death in human leukemia U937 cells (2012) Biochimie, 94, pp. 2308-2313. , https://doi.org/10.1016/j.biochi.2012.06.003
Boccellino, M., Quagliuolo, L., Verde, A., La Porta, R., Crispi, S., Piccolo, M.T., Vitiello, A., Signorile, P.G., In vitro model of stromal and epithelial immortalized endometriotic cells (2012) J Cell Biochem, 113, pp. 1292-1301. , https://doi.org/10.1002/jcb.24000
Miceli, M., Franci, G., Dell'Aversana, C., Ricciardiello, F., Petraglia, F., Carissimo, A., Perone, L., Altucci, L., MePR: a novel human mesenchymal progenitor model with characteristics of pluripotency (2013) Stem Cells Dev, 22, pp. 2368-2383. , https://doi.org/10.1089/scd.2012.0498
Miceli, M., Aversana, C.D., Russo, R., Rega, C., Cupelli, L., Ruvo, M., Altucci, L., Chambery, A., (2015) Secretome profiling of cytokines and growth factors reveals that neuro-glial differentiation is associated with the down-regulation of Chemokine Ligand 2 (MCP-1/CCL2) in amniotic fluid derived-mesenchymal progenitor cells, , https://doi.org/10.1002/pmic.201500223, Proteomics
Rotili, D., Tomassi, S., Conte, M., Benedetti, R., Tortorici, M., Ciossani, G., Valente, S., Tumber, A., Pan-histone demethylase inhibitors simultaneously targeting Jumonji C and lysine-specific demethylases display high anticancer activities (2014) J Med Chem, 57, pp. 42-55. , https://doi.org/10.1021/jm4012802
Maurano, M.T., Wang, H., John, S., Shafer, A., Canfield, T., Lee, K., Stamatoyannopoulos, J.A., Role of DNA Methylation in Modulating Transcription Factor Occupancy (2015) Cell Rep, 12, pp. 1184-1195. , https://doi.org/10.1016/j.celrep.2015.07.024
Rosenauer, A., Raelson, J.V., Nervi, C., Eydoux, P., DeBlasio, A., Miller, W.H., Jr., Alterations in expression, binding to ligand and DNA, and transcriptional activity of rearranged and wild-type retinoid receptors in retinoid-resistant acute promyelocytic leukemia cell lines (1996) Blood, 88, pp. 2671-2682
Dell'Aversana, C., Giorgio, C., D'Amato, L., Lania, G., Matarese, F., Saeed, S., Di Costanzo, A., Carissimo, A., miR-194-5p/BCLAF1 deregulation in AML tumorigenesis (2017) Leukemia, , https://doi.org/10.1038/leu.2017.64
Delhommeau, F., Dupont, S., Della Valle, V., James, C., Trannoy, S., Masse, A., Kosmider, O., Dreyfus, F.J., Mutation in TET2 in myeloid cancers (2009) N Engl J Med, 360, pp. 2289-2301. , https://doi.org/10.1056/NEJMoa0810069
Quivoron, C., Couronne, L., Della Valle, V., Lopez, C.K., Plo, I., Wagner-Ballon, O., Do Cruzeiro, M., Tilly, H., TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis (2011) Cancer Cell, 20, pp. 25-38. , https://doi.org/10.1016/j.ccr.2011.06.003
Malinge, S., Ragu, C., Della-Valle, V., Pisani, D., Constantinescu, S.N., Perez, C., Villeval, J.L., Vainchenker, W., Activating mutations in human acute megakaryoblastic leukemia (2008) Blood, 112, pp. 4220-4226. , https://doi.org/10.1182/blood-2008-01-136366
Rooswinkel, R.W., van de Kooij, B., Verheij, M., Borst, J., Bcl-2 is a better ABT-737 target than Bcl-xL or Bcl-w and only Noxa overcomes resistance mediated by Mcl-1, Bfl-1, or Bcl-B (2012) Cell Death Dis, 3. , https://doi.org/10.1038/cddis.2012.109
Nebbioso, A., Clarke, N., Voltz, E., Germain, E., Ambrosino, C., Bontempo, P., Alvarez, R., Gronemeyer, H., Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells (2005) Nat Med, 11, pp. 77-84. , https://doi.org/10.1038/nm1161
Nebbioso, A., Manzo, F., Miceli, M., Conte, M., Manente, L., Baldi, A., De Luca, A., Altucci, L., Selective class II HDAC inhibitors impair myogenesis by modulating the stability and activity of HDAC-MEF2 complexes (2009) EMBO Rep, 10, pp. 776-782. , https://doi.org/10.1038/embor.2009.88
Valente, S., Mellini, P., Spallotta, F., Carafa, V., Nebbioso, A., Polletta, L., Carnevale, I., Cencioni, C., 1, 4-Dihydropyridines Active on the SIRT1/AMPK Pathway Ameliorate Skin Repair and Mitochondrial Function and Exhibit Inhibition of Proliferation in Cancer Cells (2016) J Med Chem, , https://doi.org/10.1021/acs.jmedchem.5b01117
De Bellis, F., Carafa, V., Conte, M., Rotili, D., Petraglia, F., Matarese, F., Francoijs, K.J., Mandoli, A., Context-selective death of acute myeloid leukemia cells triggered by the novel hybrid retinoid-HDAC inhibitor MC2392 (2014) Cancer Res, 74, pp. 2328-2339. , https://doi.org/10.1158/0008-5472.CAN-13-2568
Conte, M., Dell'Aversana, C., Benedetti, R., Petraglia, F., Carissimo, A., Petrizzi, V.B., D'Arco, A.M., Altucci, L., HDAC2 deregulation in tumorigenesis is causally connected to repression of immune modulation and defense escape (2015) Oncotarget, 6, pp. 886-901. , https://doi.org/10.18632/oncotarget.2816
Chaib, H., Nebbioso, A., Prebet, T., Castellano, R., Garbit, S., Restouin, A., Vey, N., Collette, Y., Anti-leukemia activity of chaetocin via death receptor-dependent apoptosis and dual modulation of the histone methyl-transferase SUV39H1 (2012) Leukemia, 26, pp. 662-674. , https://doi.org/10.1038/leu.2011.271
Weisberg, E., Halilovic, E., Cooke, V.G., Nonami, A., Ren, T., Sanda, T., Simkin, I., Galinsky, I., Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097 (2015) Mol Cancer Ther, 14, pp. 2249-2259. , https://doi.org/10.1158/1535-7163.MCT-15-0429
Rhee, I., Bachman, K.E., Park, B.H., Jair, K.W., Yen, R.W., Schuebel, K.E., Cui, H., Vogelstein, B., DNMT1 and DNMT3b cooperate to silence genes in human cancer cells (2002) Nature, 416, pp. 552-556. , https://doi.org/10.1038/416552a
Martens, J.H., Rao, N.A., Stunnenberg, H.G., Genome-wide interplay of nuclear receptors with the epigenome (2011) Biochim Biophys Acta, 1812, pp. 818-823. , https://doi.org/10.1016/j.bbadis.2010.10.005
Saeed, S., Logie, C., Francoijs, K.J., Frige, G., Romanenghi, M., Nielsen, F.G., Raats, L., Stunnenberg, H.G., Chromatin accessibility, p300, and histone acetylation define PML-RARalpha and AML1-ETO binding sites in acute myeloid leukemia (2012) Blood, 120, pp. 3058-3068. , https://doi.org/10.1182/blood-2011-10-386086
Martens, J.H., Stunnenberg, H.G., The molecular signature of oncofusion proteins in acute myeloid leukemia (2010) FEBS Lett, 584, pp. 2662-2669. , https://doi.org/10.1016/j.febslet.2010.04.002
Combined HAT/EZH2 modulation leads to cancer-selective cell death