Supplementary Materials

Similar documents
mm C3a. 1 mm C3a Time (s) C5a. C3a. Blank. 10 mm Time (s) Time (s)

Supplemental Material

ph Switchable and Fluorescent Ratiometric Squarylium Indocyanine Dyes as Extremely Alkaline Sensors

Supporting Information for. Boronic Acid Functionalized Aza-Bodipy (azabdpba) based Fluorescence Optodes for the. analysis of Glucose in Whole Blood

Supporting Information

Nitro-Grela-type complexes containing iodides. robust and selective catalysts for olefin metathesis

Synthesis and Blastocyst Implantation Inhibition Potential of Lupeol Derivatives in Female Mice

Supporting Information. Copyright Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, 2007

Manganese powder promoted highly efficient and selective synthesis of fullerene mono- and biscycloadducts at room temperature

Supporting Information

SUPPLEMENTAL FIGURE 1 Structures and IC50 values of compounds 13 32

p-toluenesulfonic Acid-Mediated 1,3-Dipolar Cycloaddition of

Analysis of fatty acid metabolism using Click-Chemistry and HPLC-MS

Lewis acid-catalyzed regioselective synthesis of chiral α-fluoroalkyl amines via asymmetric addition of silyl dienolates to fluorinated sulfinylimines

Supporting Information for

Supporting Information. Recyclable hypervalent-iodine-mediated solid-phase peptide

Supporting Information

Masatoshi Shibuya,Takahisa Sato, Masaki Tomizawa, and Yoshiharu Iwabuchi* Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences,

Supporting Information

Supporting Information. for. Access to pyrrolo-pyridines by gold-catalyzed. hydroarylation of pyrroles tethered to terminal alkynes

Orvinols with Mixed Kappa/Mu Opioid Receptor Agonist Activity

Direct Aerobic Carbonylation of C(sp 2 )-H and C(sp 3 )-H Bonds through Ni/Cu Synergistic Catalysis with DMF as the Carbonyl Source

Supporting information

Preparation, isolation and characterization of N α -Fmoc-peptide isocyanates: Solution synthesis of oligo-α-peptidyl ureas

Fluorescent probes for detecting monoamine oxidase activity and cell imaging

Supporting Information. Design and Synthesis of Bicyclic Pyrimidinones as Potent and Orally. Bioavailable HIV-1 Integrase Inhibitors.

Ethyl 2-hydroxy-4-methyl-1-((prop-2-yn-1-yloxy)methyl)cyclohex-3-enecarboxylate (16):

Novel D-erythro N-Octanoyl Sphingosine Analogs As Chemo- and Endocrine. Resistant Breast Cancer Therapeutics

Supporting Information. Radical fluorination powered expedient synthesis of 3 fluorobicyclo[1.1.1]pentan 1 amine

Allenylphosphine oxides as simple scaffolds for. phosphinoylindoles and phosphinoylisocoumarins

Supporting Information. for. Synthesis of 2,1-benzisoxazole-3(1H)-ones by basemediated. photochemical N O bond-forming

Use of degradable cationic surfactants with cleavable linkages for enhancing the. chemiluminescence of acridinium ester labels. Supplementary Material

Facile Cu(II) mediated conjugation of thioesters and thioacids to peptides and proteins under mild conditions

Supporting Information. Nitrodibenzofuran: a One- and Two-Photon Sensitive Protecting Group that is Superior to

Supplementary Material (ESI) for Chemical Communications This journal is (c) The Royal Society of Chemistry 2008

Christophe Lincheneau, Bernard Jean-Denis and Thorfinnur Gunnlaugsson* Electronic Supplementary Information

Supporting Information

SUPPORTING INFORMATION. Transition metal-promoted synthesis of 2-aryl/heteroaryl-thioquinazoline: C-S

Synthesis of cationic porphyrin modified amino. acids

Electronic Supplementary Information

Supplementary Figure 1. Chemical structures of activity-based probes (ABPs) and of click reagents used in this study.

ELECTRONIC SUPPLEMENTARY INFORMATION

Supporting Information

Thermal shift binding experiments were carried out using Thermofluor 384 ELS system. Protein

Title Revision n date

Preparation of Fluorinated Tetrahydropyrans and Piperidines using a New Nucleophilic Fluorination Reagent DMPU/HF

Microwave heating in peptide side chain modification via sulfhydryl reaction

All chemicals were obtained from Aldrich, Acros, Fisher, or Fluka and were used without

Supporting Information

Thiol-Activated gem-dithiols: A New Class of Controllable. Hydrogen Sulfide (H 2 S) Donors

Analytical Method for 2, 4, 5-T (Targeted to Agricultural, Animal and Fishery Products)

Supporting Information Synthesis of 2-Aminobenzonitriles through Nitrosation Reaction and Sequential Iron(III)-Catalyzed C C Bond Cleavage of 2-Arylin

3016 Oxidation of ricinoleic acid (from castor oil) with KMnO 4 to azelaic acid

Regioective Halogenation of 2-Substituted-1,2,3-Triazole via sp 2 C-H Activation

Supporting Information. Copyright Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, 2006

Evaluation of Chemical Labeling Strategies for Monitoring HCV RNA using Vibrational Microscopy

Supporting Information. for. Synthesis of dye/fluorescent functionalized. dendrons based on cyclotriphosphazene

Supporting Information for. Use of the Curtius Rearrangement of Acryloyl Azides in the Synthesis of. 3,5-Disubstituted Pyridines: Mechanistic Studies

Supporting Information. First synthetic entry to the trimer stage of 5,6-dihydroxyindole polymerization: orthoalkynylaniline-based

Inhibition of Cancer-Associated Mutant Isocitrate. Dehydrogenases: Synthesis, SAR and Selective Antitumor. Activity

Supporting Information

Naoya Takahashi, Keiya Hirota and Yoshitaka Saga* Supplementary material

Supplementary Figure 1, LOX controls surface EGFR retention and AKT activation. (a, b) Western blots of py1068 EGFR, surface EGFR, total EGFR and

Supporting information

Supporting Information. Efficient copper-catalyzed Michael addition of acrylic derivatives with primary alcohols in the presence of base

Supporting Information: Cis-to-Trans Isomerization of Azobenzene Investigated by Using Thin Films of Metal-Organic Frameworks

Enantioselective synthesis of anti- and syn-β-hydroxy-α-phenyl carboxylates via boron-mediated asymmetric aldol reaction

Electronic Supplementary Information

Supporting Information

in palmitoylation of numerous proteins. P10 mitochondria enriched fractions from primary rat

Easily removable olefin metathesis catalysts

Supporting Information

Supporting Information

SUPPLEMENTARY INFORMATION

Preparation of Stable Aziridinium Ions and Their Ring Openings

Heparin Sodium ヘパリンナトリウム

Supplemental Information. Reactivity of Monovinyl (Meth)Acrylates Containing Cyclic Carbonates

by Donor-Acceptor Complex

Supplementary Materials Contents

Supporting Information

Supporting Information

A biocatalytic hydrogenation of carboxylic acids

Design and Synthesis of Orally Bioavailable. Aminopyrrolidinone Histone Deacetylase-6. Inhibitors

Supplementary Figures

Highly enantioselective tandem enzyme-organocatalyst crossed aldol reactions. with acetaldehyde in deep-eutectic-solvents.

Novel Aldosterone Synthase Inhibitors with Extended Carbocyclic Skeleton by a Combined Ligand-Based and Structure-Based Drug Design Approach

CDI Mediated Monoacylation of Symmetrical Diamines and Selective Acylation of Primary Amines of Unsymmetrical Diamines

Supporting Information

Schwartz s reagent-mediated regiospecific synthesis of 2,3-disubstituted indoles from isatins

Organic and biochemical synthesis of monolignol biosynthetic pathway intermediates

Supporting Information

Pyridazine N-Oxides as Precursors of Metallocarbenes: Rhodium-Catalyzed Transannulation with Pyrroles. Supporting Information

Supporting Information

Solid Phase Peptide Synthesis (SPPS) and Solid Phase. Fragment Coupling (SPFC) Mediated by Isonitriles

Supporting Information

Triptycene-Based Small Molecules Modulate (CAG) (CTG) Repeat Junctions

Student Handout. This experiment allows you to explore the properties of chiral molecules. You have

Cu-Catalyzed Direct C6-Arylation of Indoles

Anticancer pyrroloquinazoline LBL1 targets nuclear lamins

Supporting Information

Transcription:

Supplementary Materials Supplementary Materials and Methods Biochemical Methods Methods to assay HMT activities have been previously described (1). In vitro cell assays Proliferation and LCC calculations were performed as previously described (2). ELISA Histones were isolated from tumors and analyzed as previously described (3) Xenograft study All the procedures related to animal handling, care and the treatment in this study were performed according to the guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of Shanghai Chempartner following the guidance of the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC). For the in vivo study, mice were inoculated subcutaneously at the right flank with KARPAS-422 tumor cells (1x10 7 cells/mouse, 50% Matrigel) in 0.2 ml mixture of base media and Matrigel (McCoy s 5A : Matrigel=1:1) for tumor development. The treatments were started when the tumor size reached approximately 107 mm 3 for the anti-tumor activity study (n=10 mice per group) or 410 mm 3 for the PK/PD study (n=12 mice per group). EPZ011989 or vehicle (0.5% NaCMC+0.1% Tween-80 in water) was administered orally BID at a dose volume of 10 µl/g for 7 or 21 days. Animal body weights were measured twice weekly. Tumor size was measured twice weekly in two dimensions using a caliper, and the volume was expressed in mm 3. For the day 7 PK/PD timecourse, tumors were harvested and blood was collected (by mandibular bleeds) from n=2 mice of the 30 minutes, 1, 6, and 12 hour groups, and n=4 mice from the 3 hours post-last dose group. Bone marrow was collected from n=4 mice at the 3 hour post-last dose timepoint. Pharmacokinetic analyses Dexamethasone was used as internal standard. An aliquot of 30 µl plasma sample was added with 30 µl IS (Dexamethasone, 1000 ng/ml) and 150 µl ACN. The mixture was vortexed for 5 min and centrifuged at 14000 rpm for 5 min. An aliquot of 2 µl supernatant was injected for LC- MS/MS analysis (Q-trap 3200). For 10-fold diluted plasma samples an aliquot of 3 µl plasma sample was added with 27 µl blank plasma, the dilution factor was 10, then added with 30 µl IS (Dexamethasone, 1000 ng/ml) and 150 µl ACN. The mixture was vortexed for 5 min and centrifuged at 14000 rpm for 5 min. An aliquot of 2 µl supernatant was injected for LC-MS/MS analysis.

REFERENCES 1. S. K. Knutson et al., Nat. Chem. Biol. 2012, 8, 890-896. 2. S. R. Daigle et al., Cancer Cell 2011, 20, 53-65. 3. S. K. Knutson et al., Proc. Natl. Acad. Sci. 2013, 7922-7927. Figure F1: Reported EZH2 inhibitors

Table S1: Histone Methyltransferase Inhibition by EPZ011989 Enzyme Assay IC 50 (nm) % Inhibition at 1 µm EPZ011989 a CARM1 ND -3 ± 14 DOT1L ND 1 ± 4 EHMT1 ND 9 ± 3 EHMT2 ND 7 ± 1 EZH1 b,c 103 ± 15 98 ± 3 EZH2 Peptide Assay b,c 6 ± 8 100 ± 16 EZH2 Nucleosome Assay b ND 93 ± 11 Y641F EZH2 b,c 7 ± 5 ND PRMT1 ND 10 ± 6 PRMT3 ND 10 ± 12 PRMT5/MEP50 ND -4 ± 2 PRMT6 ND 6 ± 4 PRMT8 ND 9 ± 5 SETD2 ND 20 ± 1 SETD7 ND 6 ± 10 SUV39H1 ND 11 ± 5 SMYD2 ND 8 ± 1 SMYD3 ND 13 ± 6 WHSC1 ND 26 ± 4 WHSC1L1 ND 16 ± 10 a: Values represent the mean and standard deviation of duplicate experiments determined at 10 µmol/l EPZ011989. b: All EZH1 and EZH2 proteins were assayed in the context of 4 PRC2 components (EZH1/2, SUZ12, RBAP48, EED). c: Assayed with H3K27 peptides as substrates. ND = Not Determined

Table S2: Histone Methyltransferase Inhibition by EPZ-6438 (Previously published in the Supporting Info for S. K. Knutson et al., Proc. Natl. Acad. Sci. 2013, 7922-7927) Enzyme Assay IC 50 (nm) % Inhibition at 1 µm EPZ-6438 a CARM1 >50,000 b 5 ± 3 DOT1L >50,000 c 2 ± 8 EHMT1 >50,000 c 6 ± 6 EHMT2 >50,000 c 7 ± 3 EZH1 d,e 392 ± 72 f 98 ± 1 EZH2 Peptide Assay d,e 11 ± 5 f ND EZH2 Nucleosome Assay d 16 ± 12 f 100 ± 1 A677G EZH2 d,e 2 b ND A687V EZH2 d,e 2 b ND Y641F EZH2 d,e 14 ± 5 f ND Y641C EZH2 d,e 16 c ND Y641H EZH2 d,e 6 c ND Y641N EZH2 d,e 38 b ND Y641S EZH2 d,e 6 c ND rat EZH2 d,e 4 c ND PRMT1 >50,000 c 5 ± 4 PRMT3 ND 2 ± 2 PRMT5/MEP50 >50,000 c 2 ± 6 PRMT6 ND 3 ± 3 PRMT8 >50,000 c 7 ± 3 SETD7 ND 4 ± 3 SMYD2 >50,000 c 1 ± 2 SMYD3 ND 0 ± 5 WHSC1 >100,000 c 8 ± 3 WHSC1L1 >100,000 c 9 ± 8 a: Values represent the mean and standard deviation of duplicate experiments determined at 10 µmol/l EPZ-6438. b: Values represent the mean of duplicate experiments with two replicates per experiment. c: Values represent one experiment with two replicates per experiment. d: All EZH1 and EZH2 proteins were assayed in the context of 4 PRC2 components (EZH1/2, SUZ12, RBAP48, EED). e: Assayed with H3K27 peptides as substrates. f: Values represent mean and standard deviation of replicates (EZH1 n=4, EZH2 Peptide Assay n=4, EZH2 Nucleosome Assay n=6, Y461F EZH2 n=3). ND = Not Determined

Synthetic scheme EPZ011989: Synthesis of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)amino)cyclohexyl)- amino)-2-methylbenzoate (2): To a stirred solution of methyl 3-amino-5-bromo-2-methylbenzoate (140 g, 578 mmol) and tertbutyl (4-oxocyclohexyl)carbamate (147.8 g, 694.2 mmol) in dichloroethane (1L), acetic acid (208.2 g, 3471 mmol) was added and reaction was stirred at room temperature for 20 minutes. Then sodium triacetoxyborohydride (367.9 g, 1735 mmol) was added at 0 o C and reaction was stirred at room temperature for 16 h. On completion (monitored by TLC), the reaction was quenched with aqueous sodium bicarbonate, the organic layer was separated and the aqueous layer was extracted with dichloromethane (1.5L x 3). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography (100-200 mesh size) eluting with 2, 4, 6 & 8% ethyl acetate in hexane to remove maximum cis isomer. This afforded 180 g of mixture of cis and trans isomers (40:60 by HPLC). The trans isomer was purified by repetitive recrystallization with ethyl acetate: hexane (1:2) to afford 80 g of pure trans isomer with 99% purity. 1 H NMR (DMSO-d 6, 400 MHz) δ 6.94 (s, 1H), 6.84 (s, 1H), 6.79-6.77 (m, 1H), 4.90-4.88 (m,1h), 3.79 (s, 3H), 3.22 (m, 2H), 2.11 (s, 3H), 1.91 (m, 2H), 1.80 (m, 2H), 1.38 (s, 9H), 1.31 (m, 4H).

Synthesis of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)amino)cyclohexyl)- (ethyl)amino)-2-methylbenzoate (3): To a stirred solution of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)amino)cyclohexyl)- amino)-2-methylbenzoate (70 g, 159 mmol), acetaldehyde (17.49 g, 397.7 mmol) in dichloroethane (700 ml) and acetic acid (57.27 g, 954.5 mmol) was added and reaction was stirred at room temperature for 20 minutes. Then sodium triacetoxyborohydride (101.18 g, 477.2 mmol) was added at 0 o C and reaction was stirred at room temperature for 16 h. On completion, the reaction was quenched with aqueous sodium bicarbonate, the organic layer was separated and the aqueous layer was extracted with dichloromethane (1L x 3). The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford the title compound (58 g, 77.9%). 1 H NMR (DMSO-d 6, 400 MHz) δ 7.56 (s, 1H), 7.47 (s, 1H), 6.65-6.64 (m,1h), 3.82 (s, 3H), 3.14 (m, 1H), 3.05 (m, 2H), 2.61-2.57 (m, 1H), 2.32 (s, 3H), 1.77-1.69 (m, 4H), 1.43-1.40 (m, 2H), 1.36 (s, 9H), 1.15-1.06 (m, 2H). 0.78 (t, J=6.8 Hz, 3H). Synthesis of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)-(methyl)-amino)- cyclohexyl)(ethyl)amino)-2-methylbenzoate (4): To a stirred solution of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)amino)cyclohexyl)- (ethyl)amino)-2-methylbenzoate (30 g, 63.96 mmol) in dry DMF (300 ml), methyl iodide (40 ml, 639.6 mmol) was added at 0 o C and stirred it at same temperature for 20 min. Then NaH (60% 6.39 g, 159.9 mmol) was added at 0 o C and reaction was stirred for overnight at room temperature. On completion, the reaction was quenched with ice water and extracted with ethyl acetate (500 ml x 3). The combined organic layers were washed with water, dried, concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford the title compound (30 g, 97.3%).

1 H NMR (DMSO-d 6, 400 MHz) δ 7.55 (s, 1H), 7.46 (s, 1H), 3.82 (s, 3H), 3.05 (m, 2H), 2.65 (m, 1H), 2.61 (s, 3H), 2.33 (s, 3H), 1.77 (m, 2H), 1.52-1.47 (m, 6H), 1.38 (s, 9H). 0.79 (t, J=6.8 Hz, 3H), 1H merged in solvent peak. Synthesis of methyl 5-bromo-3-(ethyl((trans)-4-(methylamino)cyclohexyl)amino)-2- methylbenzoate (5): To a stirred solution of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)-(methyl)-amino)- cyclohexyl)(ethyl)amino)-2-methylbenzoate (30 g, 62.24 mmol) in methanol (100 ml) at 0 o C, methanolic HCl (500 ml) was added and reaction was stirred for 2 h at room temperature. After completion, the reaction was concentrated to dryness. The residue was basified with aqueous sat. bicarbonate solution till ph 8 and aqueous layer was extracted with 10% methanol in DCM (200 ml X 3). The combined organic layers were dried over Na 2 SO 4 and solvent removed under reduced pressure to afford the title compound (25g, crude). The isolated compound was used in the next step without further purification. Synthesis of methyl 5-bromo-3-(ethyl((trans)-4-((2-methoxyethyl)-(methyl)- amino)cyclohexyl)amino)-2-methylbenzoate (6): To a stirred solution of crude methyl 5-bromo-3-(ethyl((trans)-4- (methylamino)cyclohexyl)amino)-2-methylbenzoate (25 g, 65.44 mmol), 1-bromo-2- methoxyethane (18.19 g, 130.8 mmol) in acetonitrile (250 ml), K 2 CO 3 (18.06 g, 130.8 mmol) and KI (6.51 g, 39.21 mmol) were added. The resulting reaction mass was stirred at 65 o C for 16 h. On completion, the reaction mixture was diluted with water (300 ml) and extracted with DCM (500 ml x 3). The combined organic layers were washed with water, dried over sodium sulfate and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford the title compound (20 g, 69.3%). 1 H NMR (DMSO-d 6, 400 MHz) δ 7.55 (s, 1H), 7.45 (s, 1H), 3.82 (s, 3H), 3.32 (m, 4H), 3.20 (s, 3H), 3.05 (m, 2H), 2.61 (m, 1H), 2.32 (s, 3H), 2.30 (m, 1H), 2.15 (s, 3H), 1.77-1.67 (m, 4H), 1.37-1.31(m, 2H), 1.24-1.18 (m, 2H), 0.78 (t, J=6.8 Hz, 3H).

Synthesis of methyl 3-(ethyl((trans)-4-((2-methoxyethyl)-(methyl)-amino)-cyclohexyl)- amino)-2-methyl-5-(3-morpholinoprop-1-yn-1-yl)benzoate (7): The solution of methyl 5-bromo-3-(ethyl((trans)-4-((2-methoxyethyl)-(methyl)- amino)cyclohexyl)amino)-2-methylbenzoate (30 g, 68.02 mmol), crude 4-(prop-2-yn-1- yl)morpholine (25.51 g, 204 mmol) and triethylamine (20.61 g, 204 mmol) in DMF (300 ml) was bubbled through Argon for 20 min. Then CuI (3.87 g, 20.36 mmol) and Pd(PPh 3 ) 4 (7.85 g, 6.79 mmol) were added and Argon was bubbled through for further 20 min. The reaction mixture was heated at 105 o C for 4 h and then cooled to room temperature. The reaction was quenched with water (100 ml) and the separated aq. phase was extracted with 10 % MeOH/DCM (400 ml x 3). The combined organic extracts were dried over Na 2 SO 4, filtered and concentrated. The residue was purified by silica gel column chromatography to give the title compound (21 g, 63.7%). 1 H NMR (DMSO-d 6, 400 MHz) δ 7.46 (s, 1H), 7.32 (s, 1H), 3.82 (s, 3H), 3.62-3.57 (m, 6H), 3.50 (s, 2H), 3.35-3.32 (m, 2H), 3.21 (s, 3H), 3.17 (m, 1H), 3.05 (m, 2H), 2.61-2.58 (m, 2H), 2.38 (s, 3H), 2.33 (m, 1H), 2.18 (m, 2H), 1.77-1.70 (m, 4H), 1.36-1.20 (m, 4H), 0.77 (t, J=6.8 Hz, 3H), 3H merged in solvent peak. Synthesis of 4-(prop-2-yn-1-yl) morpholine (B): To a stirred solution of propargyl bromide (50 g, 420 mmol) in acetone (300 ml), CS 2 CO 3 (136.5 g, 420 mmol) was added at 0 o C. Then morpholine (36.60 g, 420 mmol) in acetone (200 ml) was added dropwise and reaction was stirred at room temperature for 16h. On completion, reaction mass was filtered and the filtrate was concentrated under reduced pressure to afford the title compound (50 g, crude). The isolated compound was taken directly to next step without further purification. Synthesis of N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-3-(ethyl((trans)-4-((2- methoxyethyl)(methyl)amino)cyclohexyl)amino)-2-methyl-5-(3-morpholinoprop-1-yn-1- yl)benzamide (EPZ011989): Aqueous NaOH (2.59 g, 64.91 mmol in 10 ml H 2 O) was added to the solution of 3- (ethyl((trans)-4-((2-methoxyethyl)-(methyl)-amino)-cyclohexyl)-amino)-2-methyl-5-(3-

morpholinoprop-1-yn-1-yl)benzoate (21 g, 43.29 mmol) in EtOH (100 ml) and stirred at 60 o C for 1 h. After completion of the reaction, ethanol was removed under reduced pressure and acidified using dilute HCl up to ph 6 and ph 4 was adjusted using citric acid. Extraction was carried out using 10 % MeOH/DCM (200 ml x 3). Combined organic layers were dried and concentrated to afford the desired hydrolyzed acid (15.5 g, 76.0%). To the solution of acid (15.5 g, 32.90 mmol) in DMSO (50 ml), 3-(amino methyl)-4, 6- dimethylpyridin-2(1h)-one (10 g, 65.80 mmol) and triethylamine (23 ml, 164.5 mmol) were added. The reaction mixture was stirred at room temperature for 15 min before PYBOP (25.66 g, 49.34 mmol) was added to it at 0 o C and further stirred for overnight at room temperature. After completion, the reaction mass was poured into ice water (100 ml) and extraction was carried out using 10 % MeOH/DCM (200 ml x 3). Combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography over basic alumina eluting with MeOH: DCM to afford the desired compound EPZ011989 as light brown solid (11g, 55.3%). Analytical data for EPZ011989 free base: LCMS: 606.50 [M+1]; HPLC: 99.07% (@ 210 nm-400 nm) (Rt; 3.791; Method: Column: YMC ODS-A 150 mm x 4.6 mm x 5 μ; Mobile Phase: A; 0.05% TFA in water/ B; 0.05% TFA in acetonitrile; Inj. Vol: 10 μl, Col. Temp.: 30 o C; Flow rate: 1.4 ml/min.; Gradient: 5% B to 95% B in 8 min, Hold for 1.5 min, 9.51-12 min 5% B); 1 H NMR (MeOD, 400 MHz) δ 7.23 (s, 1H), 7.09 (s, 1H), 6.11 (s, 1H), 4.46 (s, 2H), 3.74-3.72 (m, 4H), 3.51 (s, 2H), 3.47 (t, J=5.6 Hz, 2H), 3.32 (s, 3H), 3.07 (q, J=7.2 Hz, 2H), 2.64-2.63 (m, 7H), 2.38 (m,1h), 2.37 (s, 3H), 2.27 (s, 3H), 2.26 (s, 3H), 2.25 (s, 3H), 1.89-1.86 (m, 4H), 1.50-1.30 (m, 4H), 0.83 (t, J=7.2 Hz, 3H).