210
Views
1
CrossRef citations to date
0
Altmetric
Review

Successful Strategies in the Discovery of Small-Molecule Epigenetic Modulators with Anticancer Potential

, &
Pages 2243-2261 | Published online: 29 Oct 2015

References

  • Kouzarides T . Chromatin modifications and their function. Cell128 (4), 693–705 (2007).
  • Venkatesh S , WorkmanJL. Histone exchange, chromatin structure and the regulation of transcription. Nat. Rev. Mol. Cell Biol.16 (3), 178–189 (2015).
  • Rothbart SB , StrahlBD. Interpreting the language of histone and DNA modifications. Biochim. Biophys. Acta1839 (8), 627–643 (2014).
  • Tan M , LuoH, LeeSet al. Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell146 (6), 1016–1028 (2011).
  • Wang GG , AllisCD, ChiP. Chromatin remodeling and cancer, Part I: covalent histone modifications. Trends Mol. Med.13 (9), 363–372 (2007).
  • Hanahan D , WeinbergRA. Hallmarks of cancer: the next generation. Cell144 (5), 646–674 (2011).
  • Dawson MA , KouzaridesT. Cancer epigenetics: from mechanism to therapy. Cell150 (1), 12–27 (2012).
  • Ellis L , AtadjaPW, JohnstoneRW. Epigenetics in cancer: targeting chromatin modifications. Mol. Cancer Ther.8 (6), 1409–1420 (2009).
  • Zheng YG . Epigenetic Technological Applications. ZhengYG ( Ed.). Academic Press, Boston, MA, USA, 1–516 (2015).
  • Wongtrakoongate P . Epigenetic therapy of cancer stem and progenitor cells by targeting DNA methylation machineries. World J. Stem Cells7 (1), 137–148 (2015).
  • Yang X , LayF, HanH, JonesPA. Targeting DNA methylation for epigenetic therapy. Trends Pharmacol. Sci.31 (11), 536–546 (2010).
  • Khan O , La ThangueNB. HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications. Immunol. Cell Biol.90 (1), 85–94 (2012).
  • Wagner JM , HackansonB, LubbertM, JungM. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin. Epigenetics1 (3–4), 117–136 (2010).
  • Bannister AJ , KouzaridesT. The CBP co-activator is a histone acetyltransferase. Nature384 (6610), 641–643 (1996).
  • Rea S , EisenhaberF, O'carrollDet al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature406 (6796), 593–599 (2000).
  • Song JS , KimYS, KimDK, ParkSI, JangSJ. Global histone modification pattern associated with recurrence and disease-free survival in non-small cell lung cancer patients. Pathol. Int.62 (3), 182–190 (2012).
  • Seligson DB , HorvathS, ShiTet al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature435 (7046), 1262–1266 (2005).
  • Muller-Tidow C , KleinHU, HascherAet al. Profiling of histone H3 lysine 9 trimethylation levels predicts transcription factor activity and survival in acute myeloid leukemia. Blood116 (18), 3564–3571 (2010).
  • Piaz FD , VassalloA, RubioOC, CastellanoS, SbardellaG, De TommasiN. Chemical biology of histone acetyltransferase natural compounds modulators. Mol. Divers.15 (2), 401–416 (2011).
  • Wang F , MarshallCB, IkuraM. Transcriptional/epigenetic regulator CBP/p300 in tumorigenesis: structural and functional versatility in target recognition. Cell. Mol. Life Sci.70 (21), 3989–4008 (2013).
  • Bowers EM , YanG, MukherjeeCet al. Virtual ligand screening of the p300/CBP histone acetyltransferase: identification of a selective small molecule inhibitor. Chem. Biol.17 (5), 471–482 (2010).
  • Gao XN , LinJ, NingQYet al. A histone acetyltransferase p300 inhibitor C646 induces cell cycle arrest and apoptosis selectively in AML1-ETO-positive AML cells. PLoS ONE8 (2), e55481 (2013).
  • Santer FR , HoschelePP, OhSJet al. Inhibition of the acetyltransferases p300 and CBP reveals a targetable function for p300 in the survival and invasion pathways of prostate cancer cell lines. Mol. Cancer Ther.10 (9), 1644–1655 (2011).
  • Furdas SD , ShekfehS, BissingerEMet al. Synthesis and biological testing of novel pyridoisothiazolones as histone acetyltransferase inhibitors. Bioorg. Med. Chem.19 (12), 3678–3689 (2011).
  • Coffey K , BlackburnTJ, CookSet al. Characterisation of a Tip60 specific inhibitor, NU9056, in prostate cancer. PLoS ONE7 (10), e45539 (2012).
  • Yang H , PinelloCE, LuoJet al. Small-molecule inhibitors of acetyltransferase p300 identified by high-throughput screening are potent anticancer agents. Mol. Cancer Ther.12 (5), 610–620 (2013).
  • Kubicek S , O'sullivanRJ, AugustEMet al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol. Cell25 (3), 473–481 (2007).
  • Chang Y , ZhangX, HortonJRet al. Structural basis for G9a-like protein lysine methyltransferase inhibition by BIX-01294. Nat. Struct. Mol. Biol.16 (3), 312–317 (2009).
  • Liu F , Barsyte-LovejoyD, LiFet al. Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP. J. Med. Chem.56 (21), 8931–8942 (2013).
  • Daigle SR , OlhavaEJ, TherkelsenCAet al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell20 (1), 53–65 (2011).
  • Sarkaria SM , ChristopherMJ, KlcoJM, LeyTJ. Primary acute myeloid leukemia cells with IDH1 or IDH2 mutations respond to a DOT1L inhibitor in vitro. Leukemia28 (12), 2403–2406 (2014).
  • Daigle SR , OlhavaEJ, TherkelsenCAet al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood122 (6), 1017–1025 (2013).
  • Yu W , ChoryEJ, WernimontAKet al. Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors. Nat. Commun.3, 1288 (2012).
  • Grembecka J , HeS, ShiAet al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat. Chem. Biol.8 (3), 277–284 (2012).
  • Shi A , MuraiMJ, HeSet al. Structural insights into inhibition of the bivalent menin-MLL interaction by small molecules in leukemia. Blood120 (23), 4461–4469 (2012).
  • Karatas H , TownsendEC, CaoFet al. High-affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction. J. Am. Chem. Soc.135 (2), 669–682 (2013).
  • Cao F , TownsendEC, KaratasHet al. Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia. Mol. Cell53 (2), 247–261 (2014).
  • Knutson SK , WigleTJ, WarholicNMet al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat. Chem. Biol.8 (11), 890–896 (2012).
  • Mccabe MT , OttHM, GanjiGet al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature492 (7427), 108–112 (2012).
  • Qi W , ChanH, TengLet al. Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation. Proc. Natl Acad. Sci. USA109 (52), 21360–21365 (2012).
  • Konze KD , MaA, LiFet al. An orally bioavailable chemical probe of the lysine methyltransferases EZH2 and EZH1. ACS Chem. Biol.8 (6), 1324–1334 (2013).
  • Knutson SK , WarholicNM, WigleTJet al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad. Sci. USA110 (19), 7922–7927 (2013).
  • Knutson SK , KawanoS, MinoshimaYet al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol. Cancer Ther.13 (4), 842–854 (2014).
  • Garapaty-Rao S , NasveschukC, GagnonAet al. Identification of EZH2 and EZH1 small molecule inhibitors with selective impact on diffuse large B cell lymphoma cell growth. Chem. Biol.20 (11), 1329–1339 (2013).
  • Bradley WD , AroraS, BusbyJet al. EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation. Chem. Biol.21 (11), 1463–1475 (2014).
  • ClinicalTrials.gov . www.clinicaltrials.gov.
  • Chan-Penebre E , KuplastKG, MajerCRet al. A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. Nat. Chem. Biol.11 (6), 432–437 (2015).
  • Kaniskan HU , SzewczykMM, YuZet al. A potent, selective and cell-active allosteric inhibitor of protein arginine methyltransferase 3 (PRMT3). Angew. Chem. Int. Ed. Engl.54 (17), 5166–5170 (2015).
  • Jenuwein T , AllisCD. Translating the histone code. Science293 (5532), 1074–1080 (2001).
  • Taverna SD , LiH, RuthenburgAJ, AllisCD, PatelDJ. How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers. Nat. Struct. Mol. Biol.14 (11), 1025–1040 (2007).
  • Li Y , LiH. Many keys to push: diversifying the ‘readership’ of plant homeodomain fingers. Acta Biochim. Biophys. Sin.44 (1), 28–39 (2012).
  • Hodawadekar SC , MarmorsteinR. Chemistry of acetyl transfer by histone modifying enzymes: structure, mechanism and implications for effector design. Oncogene26 (37), 5528–5540 (2007).
  • Roth SY , DenuJM, AllisCD. Histone acetyltransferases. Annu. Rev. Biochem.70, 81–120 (2001).
  • Cole PA . Chemical probes for histone-modifying enzymes. Nat. Chem. Biol.4 (10), 590–597 (2008).
  • Furdas SD , KannanS, SipplW, JungM. Small molecule inhibitors of histone acetyltransferases as epigenetic tools and drug candidates. Arch. Pharm. (Weinheim)345 (1), 7–21 (2012).
  • Wang J , IwasakiH, KrivtsovAet al. Conditional MLL-CBP targets GMP and models therapy-related myeloproliferative disease. EMBO J.24 (2), 368–381 (2005).
  • Huntly BJ , ShigematsuH, DeguchiKet al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell6 (6), 587–596 (2004).
  • Iyer NG , OzdagH, CaldasC. p300/CBP and cancer. Oncogene23 (24), 4225–4231 (2004).
  • Pasqualucci L , Dominguez-SolaD, ChiarenzaAet al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature471 (7337), 189–195 (2011).
  • Avvakumov N , CoteJ. The MYST family of histone acetyltransferases and their intimate links to cancer. Oncogene26 (37), 5395–5407 (2007).
  • Lau OD , CourtneyAD, VassilevAet al. p300/CBP-associated factor histone acetyltransferase processing of a peptide substrate. Kinetic analysis of the catalytic mechanism. J. Biol. Chem.275 (29), 21953–21959 (2000).
  • Kwie FH , BrietM, SoupayaDet al. New potent bisubstrate inhibitors of histone acetyltransferase p300: design, synthesis and biological evaluation. Chem. Biol. Drug Des.77 (1), 86–92 (2011).
  • Oike T , KomachiM, OgiwaraHet al. C646, a selective small molecule inhibitor of histone acetyltransferase p300, radiosensitizes lung cancer cells by enhancing mitotic catastrophe. Radiother. Oncol.111 (2), 222–227 (2014).
  • Gajer JM , FurdasSD, GrunderAet al. Histone acetyltransferase inhibitors block neuroblastoma cell growth in vivo. Oncogenesis4, e137 (2015).
  • Murray K . The occurrence of Epsilon-N-methyl lysine in histones. Biochemistry3, 10–15 (1964).
  • Shi Y , LanF, MatsonCet al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell119 (7), 941–953 (2004).
  • Martin C , ZhangY. The diverse functions of histone lysine methylation. Nat. Rev. Mol. Cell Biol.6 (11), 838–849 (2005).
  • Black JC , Van RechemC, WhetstineJR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol. Cell48 (4), 491–507 (2012).
  • Greer EL , ShiY. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet.13 (5), 343–357 (2012).
  • Stimson L , RowlandsMG, NewbattYMet al. Isothiazolones as inhibitors of PCAF and p300 histone acetyltransferase activity. Mol. Cancer Ther.4 (10), 1521–1532 (2005).
  • Bostelman LJ , KellerAM, AlbrechtAM, AratA, ThompsonJS. Methylation of histone H3 lysine-79 by Dot1p plays multiple roles in the response to UV damage in Saccharomyces cerevisiae. DNA Repair (Amst.)6 (3), 383–395 (2007).
  • Zhang Y , ReinbergD. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev.15 (18), 2343–2360 (2001).
  • Krause CD , YangZH, KimYS, LeeJH, CookJR, PestkaS. Protein arginine methyltransferases: evolution and assessment of their pharmacological and therapeutic potential. Pharmacol. Ther.113 (1), 50–87 (2007).
  • Schneider R , BannisterAJ, KouzaridesT. Unsafe SETs: histone lysine methyltransferases and cancer. Trends Biochem. Sci.27 (8), 396–402 (2002).
  • Spannhoff A , HauserAT, HeinkeR, SipplW, JungM. The emerging therapeutic potential of histone methyltransferase and demethylase inhibitors. Chem Med Chem4 (10), 1568–1582 (2009).
  • Tachibana M , UedaJ, FukudaMet al. Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9. Genes Dev.19 (7), 815–826 (2005).
  • Huang J , DorseyJ, ChuikovSet al. G9a and Glp methylate lysine 373 in the tumor suppressor p53. J. Biol. Chem.285 (13), 9636–9641 (2010).
  • Kondo Y , ShenL, AhmedSet al. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS ONE3 (4), e2037 (2008).
  • Kondo Y , ShenL, SuzukiSet al. Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol. Res.37 (11), 974–983 (2007).
  • Watanabe H , SoejimaK, YasudaHet al. Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer Cell Int.8, 15 (2008).
  • Goyama S , NittaE, YoshinoTet al. EVI-1 interacts with histone methyltransferases SUV39H1 and G9a for transcriptional repression and bone marrow immortalization. Leukemia24 (1), 81–88 (2010).
  • Liu F , ChenX, Allali-HassaniAet al. Discovery of a 2,4-diamino-7-aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a. J. Med. Chem.52 (24), 7950–7953 (2009).
  • Chang Y , GaneshT, HortonJRet al. Adding a lysine mimic in the design of potent inhibitors of histone lysine methyltransferases. J. Mol. Biol.400 (1), 1–7 (2010).
  • Liu F , ChenX, Allali-HassaniAet al. Protein lysine methyltransferase G9a inhibitors: design, synthesis, and structure activity relationships of 2,4-diamino-7-aminoalkoxy-quinazolines. J. Med. Chem.53 (15), 5844–5857 (2010).
  • Liu F , Barsyte-LovejoyD, Allali-HassaniAet al. Optimization of cellular activity of G9a inhibitors 7-aminoalkoxy-quinazolines. J. Med. Chem.54 (17), 6139–6150 (2011).
  • Ayton PM , ClearyML. Molecular mechanisms of leukemogenesis mediated by MLL fusion proteins. Oncogene20 (40), 5695–5707 (2001).
  • ClinicalTrials.gov . http://clinicaltrials.gov.
  • Wu M , ShuHB. MLL1/WDR5 complex in leukemogenesis and epigenetic regulation. Chin. J. Cancer30 (4), 240–246 (2011).
  • Yokoyama A , SomervailleTC, SmithKS, Rozenblatt-RosenO, MeyersonM, ClearyML. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell123 (2), 207–218 (2005).
  • Borkin D , HeS, MiaoHet al. Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell27 (4), 589–602 (2015).
  • Volkel P , DupretB, Le BourhisX, AngrandPO. Diverse involvement of EZH2 in cancer epigenetics. Am. J. Transl. Res.7 (2), 175–193 (2015).
  • Diaz E , MachuttaCA, ChenSet al. Development and validation of reagents and assays for EZH2 peptide and nucleosome high-throughput screens. J. Biomol. Screen.17 (10), 1279–1292 (2012).
  • Nasveschuk CG , GagnonA, Garapaty-RaoSet al. Discovery and optimization of tetramethylpiperidinyl benzamides as inhibitors of EZH2. ACS Med. Chem. Lett.5 (4), 378–383 (2014).
  • Cheng D , YadavN, KingRW, SwansonMS, WeinsteinEJ, BedfordMT. Small molecule regulators of protein arginine methyltransferases. J. Biol. Chem.279 (23), 23892–23899 (2004).
  • Feng Y , LiM, WangB, ZhengYG. Discovery and mechanistic study of a class of protein arginine methylation inhibitors. J. Med. Chem.53 (16), 6028–6039 (2010).
  • Dillon MB , BachovchinDA, BrownSJet al. Novel inhibitors for PRMT1 discovered by high-throughput screening using activity-based fluorescence polarization. ACS Chem. Biol.7 (7), 1198–1204 (2012).
  • Wang J , ChenL, SinhaSHet al. Pharmacophore-based virtual screening and biological evaluation of small molecule inhibitors for protein arginine methylation. J. Med. Chem.55 (18), 7978–7987 (2012).
  • Yan L , YanC, QianKet al. Diamidine compounds for selective inhibition of protein arginine methyltransferase 1. J. Med. Chem.57 (6), 2611–2622 (2014).
  • Siarheyeva A , SenisterraG, Allali-HassaniAet al. An allosteric inhibitor of protein arginine methyltransferase 3. Structure20 (8), 1425–1435 (2012).
  • Liu F , LiF, MaAet al. Exploiting an allosteric binding site of PRMT3 yields potent and selective inhibitors. J. Med. Chem.56 (5), 2110–2124 (2013).
  • Huynh T , ChenZ, PangSet al. Optimization of pyrazole inhibitors of coactivator associated arginine methyltransferase 1 (CARM1). Bioorg. Med. Chem. Lett.19 (11), 2924–2927 (2009).
  • Therrien E , LaroucheG, MankuSet al. 1,2-Diamines as inhibitors of co-activator associated arginine methyltransferase 1 (CARM1). Bioorg. Med. Chem. Lett.19 (23), 6725–6732 (2009).
  • Allan M , MankuS, TherrienEet al. N-Benzyl-1-heteroaryl-3-(trifluoromethyl)-1H-pyrazole-5-carboxamides as inhibitors of co-activator associated arginine methyltransferase 1 (CARM1). Bioorg. Med. Chem. Lett.19 (4), 1218–1223 (2009).
  • Selvi BR , BattaK, KishoreAHet al. Identification of a novel inhibitor of coactivator-associated arginine methyltransferase 1 (CARM1)-mediated methylation of histone H3 Arg-17. J. Biol. Chem.285 (10), 7143–7152 (2010).
  • Bolden JE , PeartMJ, JohnstoneRW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov.5 (9), 769–784 (2006).
  • Ueda H , MandaT, MatsumotoSet al. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. III. Antitumor activities on experimental tumors in mice. J. Antibiot. (Tokyo)47 (3), 315–323 (1994).
  • Darkin-Rattray SJ , GurnettAM, MyersRWet al. Apicidin: a novel antiprotozoal agent that inhibits parasite histone deacetylase. Proc. Natl Acad. Sci. USA93 (23), 13143–13147 (1996).
  • Tsuji N , KobayashiM, NagashimaK, WakisakaY, KoizumiK. A new antifungal antibiotic, trichostatin. J. Antibiot. (Tokyo)29 (1), 1–6 (1976).
  • Saito A , YamashitaT, MarikoYet al. A synthetic inhibitor of histone deacetylase, MS-27–275, with marked in vivo antitumor activity against human tumors. Proc. Natl Acad. Sci. USA96 (8), 4592–4597 (1999).
  • Richon VM , WebbY, MergerRet al. Second generation hybrid polar compounds are potent inducers of transformed cell differentiation. Proc. Natl Acad. Sci. USA93 (12), 5705–5708 (1996).
  • Bedalov A , GatbontonT, IrvineWP, GottschlingDE, SimonJA. Identification of a small molecule inhibitor of Sir2p. Proc. Natl Acad. Sci. USA98 (26), 15113–15118 (2001).
  • Heltweg B , GatbontonT, SchulerADet al. Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res.66 (8), 4368–4377 (2006).
  • Napper AD , HixonJ, McdonaghTet al. Discovery of indoles as potent and selective inhibitors of the deacetylase SIRT1. J. Med. Chem.48 (25), 8045–8054 (2005).
  • Grozinger CM , ChaoED, BlackwellHE, MoazedD, SchreiberSL. Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening. J. Biol. Chem.276 (42), 38837–38843 (2001).
  • Lara E , MaiA, CalvaneseVet al. Salermide, a Sirtuin inhibitor with a strong cancer-specific proapoptotic effect. Oncogene28 (6), 781–791 (2009).
  • Lain S , HollickJJ, CampbellJet al. Discovery, in vivo activity, and mechanism of action of a small-molecule p53 activator. Cancer Cell13 (5), 454–463 (2008).
  • Hoffmann G , BreitenbucherF, SchulerM, Ehrenhofer-MurrayAE. A novel sirtuin 2 (SIRT2) inhibitor with p53-dependent pro-apoptotic activity in non-small cell lung cancer. J. Biol. Chem.289 (8), 5208–5216 (2014).
  • Rumpf T , SchiedelM, KaramanBet al. Selective Sirt2 inhibition by ligand-induced rearrangement of the active site. Nat. Commun.6, 6263 (2015).
  • Harris WJ , HuangX, LynchJTet al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell21 (4), 473–487 (2012).
  • Huang Y , GreeneE, Murray StewartTet al. Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes. Proc. Natl Acad. Sci. USA104 (19), 8023–8028 (2007).
  • Huang Y , StewartTM, WuYet al. Novel oligoamine analogues inhibit lysine-specific demethylase 1 and induce reexpression of epigenetically silenced genes. Clin. Cancer Res.15 (23), 7217–7228 (2009).
  • Culhane JC , SzewczukLM, LiuX, DaG, MarmorsteinR, ColePA. A mechanism-based inactivator for histone demethylase LSD1. J. Am. Chem. Soc.128 (14), 4536–4537 (2006).
  • Ueda R , SuzukiT, MinoKet al. Identification of cell-active lysine specific demethylase 1-selective inhibitors. J. Am. Chem. Soc.131 (48), 17536–17537 (2009).
  • Sorna V , TheisenER, StephensBet al. High-throughput virtual screening identifies novel N’-(1-phenylethylidene)-benzohydrazides as potent, specific, and reversible LSD1 inhibitors. J. Med. Chem.56 (23), 9496–9508 (2013).
  • Ma LY , ZhengYC, WangSQet al. Design, synthesis, and structure–activity relationship of novel LSD1 inhibitors based on pyrimidine-thiourea hybrids as potent, orally active antitumor agents. J. Med. Chem.58 (4), 1705–1716 (2015).
  • Tamara Maes , IñigoTirapu, CristinaMascaróet al. Preclinical characterization of a potent and selective inhibitor of the histone demethylase KDM1A for MLL leukemia. J. Clin. Oncol.31 (Suppl.), Abstract e13543 (2013).
  • Thinnes CC , EnglandKS, KawamuraA, ChowdhuryR, SchofieldCJ, HopkinsonRJ. Targeting histone lysine demethylases – progress, challenges, and the future. Biochim. Biophys. Acta1839 (12), 1416–1432 (2014).
  • Luo X , LiuY, KubicekSet al. A selective inhibitor and probe of the cellular functions of Jumonji C domain-containing histone demethylases. J. Am. Chem. Soc.133 (24), 9451–9456 (2011).
  • King ON , LiXS, SakuraiMet al. Quantitative high-throughput screening identifies 8-hydroxyquinolines as cell-active histone demethylase inhibitors. PLoS ONE5 (11), e15535 (2010).
  • Sayegh J , CaoJ, ZouMRet al. Identification of small molecule inhibitors of Jumonji AT-rich interactive domain 1B (JARID1B) histone demethylase by a sensitive high throughput screen. J. Biol. Chem.288 (13), 9408–9417 (2013).
  • Labelle M , BoesenT, MehrotraM, KhanQ, UllahF : WO2014053491 (2014).
  • Staller P . The potential applications of enzymatic inhibitors of KDM5 in oncology. Presented at : 21st International Molecular Medicine Tri-Conference. San Francisco, CA, USA, 9–14 February 2014.
  • Itoh Y , SawadaH, SuzukiMet al. Identification of Jumonji AT-Rich interactive domain 1A inhibitors and their effect on cancer cells. ACS Med. Chem. Lett.6 (6), 665–670 (2015).
  • Upadhyay AK , RotiliD, HanJWet al. An analog of BIX-01294 selectively inhibits a family of histone H3 lysine 9 Jumonji demethylases. J. Mol. Biol.416 (3), 319–327 (2012).
  • Wang L , ChangJ, VargheseDet al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat. Commun.4, 2035 (2013).
  • Kruidenier L , ChungCW, ChengZet al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature488 (7411), 404–408 (2012).
  • Rotili D , TomassiS, ConteMet al. Pan-histone demethylase inhibitors simultaneously targeting Jumonji C and lysine-specific demethylases display high anticancer activities. J. Med. Chem.57 (1), 42–55 (2014).
  • Lin YC , LinYC, ShihJYet al. DUSP1 expression induced by HDAC1 inhibition mediates gefitinib sensitivity in non-small cell lung cancers. Clin. Cancer Res.21 (2), 428–438 (2015).
  • Pazolli E , AlspachE, MilczarekA, PriorJ, Piwnica-WormsD, StewartSA. Chromatin remodeling underlies the senescence-associated secretory phenotype of tumor stromal fibroblasts that supports cancer progression. Cancer Res.72 (9), 2251–2261 (2012).
  • Roy SS , GonuguntaVK, BandyopadhyayAet al. Significance of PELP1/HDAC2/miR-200 regulatory network in EMT and metastasis of breast cancer. Oncogene33 (28), 3707–3716 (2014).
  • Weichert W . HDAC expression and clinical prognosis in human malignancies. Cancer Lett.280 (2), 168–176 (2009).
  • Simo-Riudalbas L , EstellerM. Targeting the histone orthography of cancer: drugs for writers, erasers and readers. Br. J. Pharmacol.172 (11), 2716–2732 (2014).
  • Micelli C , RastelliG. Histone deacetylases: structural determinants of inhibitor selectivity. Drug Discov. Today20 (6), 718–735 (2015).
  • Dobler MR , GrobJE, PatnaikA, RadetichB, ShultzM, ZhuY. WO 2007038459A2 (2007).
  • Ramalingam SS , MaitlandML, FrankelPet al. Carboplatin and Paclitaxel in combination with either vorinostat or placebo for first-line therapy of advanced non-small-cell lung cancer. J. Clin. Oncol.28 (1), 56–62 (2010).
  • Juergens RA , WrangleJ, VendettiFPet al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov.1 (7), 598–607 (2011).
  • Younes A , OkiY, BociekRGet al. Mocetinostat for relapsed classical Hodgkin's lymphoma: an open-label, single-arm, Phase 2 trial. Lancet Oncol.12 (13), 1222–1228 (2011).
  • Sauve AA . Sirtuin chemical mechanisms. Biochim. Biophys. Acta1804 (8), 1591–1603 (2010).
  • Grbesa I , PajaresMJ, Martinez-TerrobaEet al. Expression of sirtuin 1 and 2 is associated with poor prognosis in non-small cell lung cancer patients. PLoS ONE10 (4), e0124670 (2015).
  • Yuan H , SuL, ChenWY. The emerging and diverse roles of sirtuins in cancer: a clinical perspective. Onco Targets Ther.6, 1399–1416 (2013).
  • Chu F , ChouPM, ZhengX, MirkinBL, RebbaaA. Control of multidrug resistance gene mdr1 and cancer resistance to chemotherapy by the longevity gene sirt1. Cancer Res.65 (22), 10183–10187 (2005).
  • Peck B , ChenCY, HoKKet al. SIRT inhibitors induce cell death and p53 acetylation through targeting both SIRT1 and SIRT2. Mol. Cancer Ther.9 (4), 844–855 (2010).
  • Li L , WangL, LiLet al. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell21 (2), 266–281 (2012).
  • Helin K , DhanakD. Chromatin proteins and modifications as drug targets. Nature502 (7472), 480–488 (2013).
  • Ding J , ZhangZM, XiaYet al. LSD1-mediated epigenetic modification contributes to proliferation and metastasis of colon cancer. Br. J. Cancer109 (4), 994–1003 (2013).
  • Sakamoto A , HinoS, NagaokaKet al. Lysine demethylase LSD1 coordinates glycolytic and mitochondrial metabolism in hepatocellular carcinoma cells. Cancer Res.75 (7), 1445–1456 (2015).
  • Lei ZJ , WangJ, XiaoHLet al. Lysine-specific demethylase 1 promotes the stemness and chemoresistance of Lgr5 liver cancer initiating cells by suppressing negative regulators of beta-catenin signaling. Oncogene34 (24), 3188–3198 (2015).
  • Sareddy GR , NairBC, KrishnanSKet al. KDM1 is a novel therapeutic target for the treatment of gliomas. Oncotarget4 (1), 18–28 (2013).
  • Etani T , SuzukiT, NaikiTet al. NCL1, a highly selective lysine-specific demethylase 1 inhibitor, suppresses prostate cancer without adverse effect. Oncotarget6 (5), 2865–2878 (2015).
  • Cho HS , ToyokawaG, DaigoYet al. The JmjC domain-containing histone demethylase KDM3A is a positive regulator of the G1/S transition in cancer cells via transcriptional regulation of the HOXA1 gene. Int. J. Cancer131 (3), E179–E189 (2012).
  • Kim JY , KimKB, EomGHet al. KDM3B is the H3K9 demethylase involved in transcriptional activation of lmo2 in leukemia. Mol. Cell. Biol.32 (14), 2917–2933 (2012).
  • Xiang Y , ZhuZ, HanGet al. JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. Proc. Natl Acad. Sci. USA104 (49), 19226–19231 (2007).
  • Yamane K , TateishiK, KloseRJet al. PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation. Mol. Cell25 (6), 801–812 (2007).
  • Sharma SV , LeeDY, LiBet al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell141 (1), 69–80 (2010).
  • Roesch A , VulturA, BogeskiIet al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell23 (6), 811–825 (2013).
  • Kottakis F , FoltopoulouP, SanidasIet al. NDY1/KDM2B functions as a master regulator of polycomb complexes and controls self-renewal of breast cancer stem cells. Cancer Res.74 (14), 3935–3946 (2014).
  • Schiller R , ScozzafavaG, TumberAet al. A cell-permeable ester derivative of the JmjC histone demethylase inhibitor IOX1. Chem Med Chem9 (3), 566–571 (2014).
  • Rose NR , WoonEC, TumberAet al. Plant growth regulator daminozide is a selective inhibitor of human KDM2/7 histone demethylases. J. Med. Chem.55 (14), 6639–6643 (2012).
  • Suzuki T , OzasaH, ItohYet al. Identification of the KDM2/7 histone lysine demethylase subfamily inhibitor and its antiproliferative activity. J. Med. Chem.56 (18), 7222–7231 (2013).
  • Sekar TV , FoygelK, GelovaniJG, PaulmuruganR. Genetically encoded molecular biosensors to image histone methylation in living animals. Anal. Chem.87 (2), 892–899 (2015).
  • Van Rechem C , BlackJC, BoukhaliMet al. Lysine demethylase KDM4A associates with translation machinery and regulates protein synthesis. Cancer Discov.5 (3), 255–263 (2015).
  • Grasso CS , TangY, TruffauxNet al. Functionally defined therapeutic targets in diffuse intrinsic pontine glioma. Nat. Med.21 (6), 555–559 (2015).
  • Hashizume R , AndorN, IharaYet al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat. Med.20 (12), 1394–1396 (2014).
  • Ntziachristos P , TsirigosA, WelsteadGGet al. Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia. Nature514 (7523), 513–517 (2014).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.