OA17202A - Inhibitors of beta-secretase. - Google Patents

Inhibitors of beta-secretase. Download PDF

Info

Publication number
OA17202A
OA17202A OA1201500052 OA17202A OA 17202 A OA17202 A OA 17202A OA 1201500052 OA1201500052 OA 1201500052 OA 17202 A OA17202 A OA 17202A
Authority
OA
OAPI
Prior art keywords
mixture
mmol
disease
added
stirred
Prior art date
Application number
OA1201500052
Inventor
Ulrike Gross
Niklas Heine
Lanqi Jia
Deepak S. Lala
Angel Morales-Ramos
Suresh B. Singh
Achim Sauer
Shankar Venkatraman
Zhenrong Xu
Jing Yuan
Yi Zhao
Yajun Zheng
Yuri Bukhtiyarov
Salvación CACATIAN
Lawrence Wayne Dillard
Dorner-Ciossek
Klaus Fuchs
Original Assignee
Boehringer Ingelheim Interna Tional Gmbh
Vitae Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim Interna Tional Gmbh, Vitae Pharmaceuticals, Inc. filed Critical Boehringer Ingelheim Interna Tional Gmbh
Publication of OA17202A publication Critical patent/OA17202A/en

Links

Abstract

The present invention relates to spirocyclic acylguanidines and their use as inhibitors of the B-secretase enzyme (BACE1) activity, pharmaceutical compositions containing the same, and methods of using the same as therapeutic agents in the treatment of neurodegenerative disorders, disorders characterized by cognitive decline, cognitive impairment, dementia and diseases characterized by production of B-amyloid aggregates.

Description

DETAILED DESCRIPTION OF THE INVENTION
The compounds of the présent invention exhibit potent activity against the BACEI enzyme and Abeta formation together with high selectivity against the hERG channel, low propensity to cause phospholipidosis, and high metabolic stability. For example, the compounds ofthe présent invention show a BACEI inhibition with an IC» < 15 nM, a hERG inhibition of less than 35% at 10 μΜ, phospholipidosis with a First Effect Concentration (FEC) of at least 100 μΜ, and a metabolic stability of less than 25 percent of hepatic blood flow at I μΜ. These combined properties make the
compounds of the présent invention useful for the treatment of pathological states in humons, in particular, for the treatment of Alzheimer’s disease as well os other disorders and diseases mediated byBACEl.
Inhibition of the hERG (human Ether-à-go-go-Related Gene) channel by xenobiotics and subséquent delayed cardiac repolarization Is associated with an increased risk for a spécifie polymorphie ventricular tachyarrhythmia, torsade de pointes, os established by Sanguinetti et al. (1995, Cell, Apr. 21,81(2):299-307) and a large body of subséquent evidence. To avoid this risk eorly on, screening against hERG interaction in an In vitro system using hcterologous expression of the hERG channel is common practice and an assay of this type is also an important part of later preclinicaI candidate profïling as recommended by the ICH guideline S7B (International Conférence on Harmonization (2005): ICHTopic S 7 B; The nonclinical Evaluation of the Potentiel for delayed Ventricular Repolarization; (QT Interva! Prolongation) by Human Pharmaceuticals (www.ich.org/products/guidelines/safety/article/safety-guideltnes.htm!)). As such, low hERG channel inhibition, such as that shown by the compounds of the présent invention, is highly désirable for therapeutlcs.
Phospholipidosis is a lipid storage disorder in which excess phospholipids accumulate within cells. Drug-induced phospholipidosis is an undesirable drug reaction. Therefore, in orderto avoid detrimenta! side effects, compounds with low phospholipidosis potentïal are preferred for human therapeutic use.
Metabolic stability refers to the susceptibility of compounds to biotransformation In the context of selecting and/or designing dru g s with favorable pharmacokirtetic properties. The primary site of metabolism for many drugs is the liver. Intact hépatocytes contain the cytochrome P450s (CYPs), other non-P450 enzymes, and phase II enzymes such as sulfo- and glucuronosyltransferases, and thus represent a prime model system for studying drug metabolization in vitro. Enhanced metabolic stability is associated with several advantages, including increased bloavailabiIity and longer half-lîfe, which can enable lower and less frequent dosing of patients. Thus, enhanced metabolic stability 1s a favorable characterislic for compounds that are to be used for drugs.
Data provided in Table I below show that compounds of the présent invention hâve the combination of potent BACEI inhibitory activity, selectivity against cardiac hERG, low propensity to 30 cause phospholipidosis, and high metabolic stability. Table 2 provides data showing that certain comparator compounds described în W02010/105179 do not meet one or more of these criteria.
Terms not specifically defined herein should be given the meanings that would be given to them by one of skill In the art in light of the disclosure and the context. As used in the spécification, however, unless specified to the contrary, the following terms hâve the meaning indicated and the 35 following conventions are adhered to.
When a compound of the présent invention is depicted by name or structure without indicating ail tautomeric forms, it is to be understood that the compound and its pharmaceutically acceptable salts shall encompass ail tautomers.
When a compound of the présent invention is depicted by name or structure without indicating the stereochemistry, it is to be understood that the compound and its pharmaceutically acceptable salts shall encompass ail stéréo, optical and geometrical isomers (e.g., enantiomers, diastereomers, E/Z isomers, etc.) and racemates thereof, as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereomers, or mixtures of any of the foregoing forms.
When a stéréo, optical or géométrie Isomer is depicted by name or structure, it is to be understood that the stéréo, optical and/or géométrie isomeric purity of the named or depicted stéréo, optical or géométrie isomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% pure by weight. Stéréo, optical and géométrie isomeric purity is determined by dividïng the weight of the named or depicted stéréo, optical and géométrie isomer in a mixture by the total weight of ail stéréo, optical and géométrie isomers in the mixture.
When a compound of the présent invention or its pharmaceutically acceptable sait is named or depicted by structure, it Is to be understood that solvatés, hydrates and the anhydrous form of the compound and solvatés, hydrates and anhydrous form of its pharmaceutically acceptable sait are included in the invention. “Solvatés refer to crystalline forms wherein solvent molécules are incorporated into the crystal lattice during crystallization. Solvaté may indude water or nonaqueous 20 solvents such as éthanol, tsopropanol, DMSO, acetic acid, ethanolamine, and EtOAc. Solvatés, wherein water is the solvent molécule incorporated into the crystal lattice, arc typically referred to os “hydrates. Hydrates include stoichiometric hydrates us well us compositions containing variable amounts of water. “Anhydrous form refers to compounds with no solvent or water or substantiaily no solvent or water incorporated into the crystal structure (e.g., less than 1:10, 1:20; 1:100; or 1:200 25 molar ratio of solvent or water to compound).
Salts
The phrase pharmaceutically acceptable is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animais without excessive toxicity, irritation, allergie response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio.
As used herein, pharmaceutically acceptable salts refer to dérivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but arc not limited to, minerai or organic acid salts of 35 basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and
lhe like. For example, such salts include salts from ammonia, L-urginine, betaine, benethaminc, benzathinc, calcium hydroxide, choline, deanol, diethanolamine (2.2’-iminobis(cthanol)), diethylamine, 2-(diethylamino)-ethanol. 2-aminoethonol, ethylenediamine, N-ethyl-glucamine, hydrabamine, IH-imidazole, lysine, magnésium hydroxide, 4-(2-hydroxycthyl)-morpholinc, piperazine, potassium hydroxide, 1 -(2-hydroxyethy l)-pyrrolidine, sodium hydroxide, triethanolamine (2,2',2w-nitrïlotris(ethanol)), tromethamine, zinc hydroxide, acetic acid, 2.2-dichloro-acetic acid, adipïc acid, alginîc acid, ascorbic acid, L-aspartic acid, benze ne sulfonic acid, benzoic acid, 2,5dihydroxybenzoic acid, 4-acetamido-bcnzoic acid, (+)-comphoric ocid, (+)-camphor-10-sul fonic acid, carbonic acid, cinnamic acid, citric acid, cydamic acid, decanoic acid, dodecylsulfuric acid, ethane10 1,2-disulfonic acid, ethanesul fonic acid, 2-hydroxy-cthanesul fonic acid, ethylenediaminetetraacetic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, D-glucoheptonic acid, D-gluconic acid, D-glucuronic acid, glutamic acid, glutaric ocid, 2-oxo-glutaric acid, glycerophosphoric acid, glycine, glycolic acid, hexanoic ocid, hippuric ocid, hydrobromic acid, hydrochloric acid, isobutyric acid, DLlactic acid, lactobionic acid, lauric acid, lysine, maleic acid, (-)-L-malic ocid, malonic acid, DL15 mandelic ocid, methanesulfonic acid, galactaric acid, naphthalene-1,5-disulfonic acid, naphthalene-2sulfonic acid, l-hydroxy-2-naphthoicacid, nicotimc acid, nitric acid, octanoic acid,oleic acid, orotic acid, oxalîc acid, palmitic acid, pamoïc acid (embonic acid), phosphoric acid, propionic acid, (-)-Lpyroghitamic acid, salicylic acid, 4-amino-salicylic acid, sebacic acid, stearic ocid, succïnic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-loluenesul fonic acid and undecylenic acid. Prefened salts are L-mandelic acid and maleic acid. Further pharmaceutically acceptable salts can be formed with cations from metals like aluminium, calcium, lithium, magnésium, potassium, sodium, zinc and the like (see also Pharmaceutical salts, Berge, S.M. étal., J. Pharm. Sci., (1977), 66:1-19).
The pharmaceutically acceptable salts of the présent invention can be synthesized from the parent compound which contains a basic or acidic moicty by conventional chemical methods.
Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficient amount of the appropriate base or acid in water or in an organic diluent like ether, ethyl acetate, éthanol, isopropanol, or acetonitrile, or a mixture thereof.
Salts of acids other than those mentioned above which for example are usefui for purifying or isolating the compounds of the présent invention (e.g., trifluoro acetate salts) also comprise u port of the invention.
Biologlcal Data
BACE1 Assay (Assay 1)
The inhibitory activity of compounds was assessed by a fluorescence quench assay of BACE1 activity using commerciaüy available substrate HiLyte F!uor™488-Glu-Val-Asn-Leu-Asp-A!a-G!u5 Phe-Lys-(QXL™ 520)-OH (SEQ ID NO: 1 ) (AnaSpec, San José, CA) and truncated human betasecretase, BACEI (amino acids 1-454) fused to a myc-his tag and secreted from HEK293/BACE«t cells into OptiMEM™ (Invitrogen). The substrate was dissolved at l mg/ml in DMSO.
The assay was performed in the presence of OptiMEM™ (supematant collected over 24 h and deared from cellular débris by centrifugation) contaînîng the ectodomaîn of BACEI, 25 μ| water 10 contaînîng the desired 2-fold concentration of test compound and 2% DMSO, 1 μΜ substrate peptide, 20 mM NaOAc, pH 4.4, and 0.04% Triton-XlOO In a total assay volume of 50 μΐ in a 384 well plate. In general, 25 μΙ of compound dilution were added to the plate followed by the addition of 10 μΐ of BACEI contaînîng OptiMEM™ diluted 1:10 in water with0.2% Triton X-100. The réaction was storted with the addition of 15 μί substrate in NaOAc buffcr. The reaction was incubated at rt (dark) 15 in an Envision* multilabel reader (Perkin Elmer) and the cleavage of the substrate was recorded as kinetic for 60 min at ex: 485 nm, em: 538 nm. Blank wells contaînîng no enzyme were inciuded on each plate.
The intensity of fluorescence was regressed against time in order to dérivé velocities of reaction in ail 384 wells. These velocities were used for calcul ating percent contre! using an 20 uninhibited contreI contaînîng I % DMSO as 100% and a blank control performed in the absence of enzyme as 0%. IC30 values were calculated by fitting percent control vs. test compound concentration using Assay Explorer*.
hERG-Channel Assay (Assay 2)
Cells; HEK (human embryonic kidney) 293 cells were stably transfected with hERG cDNA.
Pipettes and solutions:
Cells were superfused with a bath solution contaînîng (mM): NaCl (137), KCI (4.0), MgClj (1.0), CaCI2 (1.8), Glucose (10), HEPES (10). pH 7.4 with NaOH. Patch pipettes were mode from borosilicate glass tubing using a horizontal puller and fîlied with pipette solution contaînîng (mM): Kaspartate (130), MgClj (5.0), EGTA (5.0), KiATP (4.0), HEPES (10.0), pH 7.2 with KOH. 30 Résistance of the microelectrodes was in the range between 2 and 5 ΜΩ.
Stimulation and recording:
Membrane currents were recorded using an EPC-10 patch ciamp amplifier and PatchMaster software. hERG-mediatcd membrane currents were recorded at 35 ’C, using the whole-cell
ίο configuration of the patch-clamp technique. Transfected HEK293 cells were clamped at a holding potential of -60 mV and hERO-mediated inactivating tail currents were elicited using a puise pattern with fixed amplitudes (uctivalion/inactivation: 40 mV for 2000 ms; recovery: -120 mV for 2 ms; ramp to 40 mV in 2 ms; inactivating tail current: 40 mV for 50 ms) repeated at 15 s intervals. During each Inter-pulse interval, 4 puises scaled down by a factor of 0.2 were recorded for a P/π leak subtraction procedure. Ra compensation was employed up to a level that safely allowed recording devoid of ringing.
Compound préparation and application:
The different concentrations of the test compounds were applied sequentially on each of the different cells investigated. A steady state level of baseline current was measured for at least 6 sweeps prior to the application of the first test compound concentration.
The test compound was dissolved In DMSO to yield a master stock solution which was diluted further In DMSO to stock solutions needed for the lower concentrations. Final dilutions in extracellular buffer were prepared freshly from these stocks by a 1:1000 dilution step each before 15 starting the expérimenta.
Data analysis:
Peak current amplitudes were measured 3 ms after the ramp to +40 mV. For baseline and each concentration, the peak currcnts of the three last sweeps before application of the next concentration were averaged. Residuol cutrcnts (I/L) were calculated for each cell os the fraction of 20 actual average peak current and average baseline peak current.
In vitro Phospholipidosîs Assay (Assay 3)
The phospholipidogenic potential of test compounds was assayed using the human hematopoetlc U937 cell line. The test principle was to analyze the phospholipid content by staining the ceils with the fluorescent dye Nile red.
U937 cells were seeded into cell culture plates at 0.5 x 10e cells/mL in RPMI medium containing 10 % FBS, 1 % DMSO, and 0.005 % gentamicin. The cells were cultivated with or without different concentrations of test compound for 48 h under standard culture conditions.
For horvesting, the cells were centrifuged ut 130x g for 4 min and washed once with PBS. Then, 2x 05 mL cell suspensions were prepared for non-fixed cell measurement (05 mL for propidium iodide (PI) viability measurement and 0.5 mL for Nile red measurement).
The remaining cells were fixed with 3.7 % formai de hyde for 30 min. After a further centrifugation step, cells were resuspended with 13 mL Nile red working soluiion ( I pg/mL) and incubated for 5 min at rt. The cell suspension was washed twice with 3 mL PBS and centrifuged with
130x g for 4 min. The supematant was discarded and the celîs were resuspcndcd with 0.5 mL PBS and kept for flow cytometry measurement.
For Nile red staining of the 0.5 mL non-fixed ce! I samples, 50 pL of a ready to use Nïle red solution (10 pg/mL) were added per sample. Samples were kept on lce for 5 min. Thereafter, they were washed once with 4 mL PBS (4 ’C, 250x g for 8 min) and finally rcsuspcnded in 400 pL PBS and kept for flow cytometry measurement.
For the viability measurement, 12.5 pL of the ready to use PI solution (10 pg/mL) were added to the 05 mL non-fixed cell suspension. After 15 min of incubation on ice, the samples were measured by flow cytometry using a Coulter Epies XL/MCL flow cytometer.
The viability of the celîs of each sample was determined by flow cytometry measurement of the PI content at channel 2 (568-590 nm). Cut-off gates for the fluorescence-depcndent différentiation between live and dead cells were defined based on the analysis of cell culture medium control samples.
Only samples with a cell viability of >= 90 % relative to control samples were nnalyzed for phospholipidosis. Each Nïle red sample (non-fixed and fixed samples) was measured by flow cytometry at channel I (504-541 nm) and channel 4 (660-680 nm).
For each channel, relative Nile red fluorescence întensity of a test sample was calculated compared to control samples and expressed as a pcrcentage of control fluorescence Intensity. The assessment of the phospholipidogenic potentiai and the first effective concentration (FEC) of a test 20 compound was done manuully based on the fluorescence intensifies at both wavelengths for the fixed cells, as well as for the non-fixed cells.
In vitro Hépatocyte Stablllty Assay (Assay 4)
The metabolic dégradation of test compounds was assayed in a hépatocyte suspension.
Cryopreserved hépatocytes were incubated in an appropriate buffer system (e.g., Dulbecco's modified 25 eugle medium plus 3.5pg glucagon/SOOmL, 2.5mg insulin/500mL and 3.75mg/500mL hydrocortison) containîng 5% species sérum. Following n 30 min preincubation in an incubator (37 ’C, 10% CO2), 5 μ! of test compound solution (80 μΜ; from 2mM in DMSO stock solution diluted 1:25 with medium) were added into 395 μΙ hépatocyte suspension (ce!! density in the range 025-5 x 10e cells/mL, typically I x IOecelis/mL; final concentration of test compound ΙμΜ, final DMSOconcentration 30 0.05%),
The cells were incubated for six h (incubator, orbital shaker) and samples (25p!) were taken at
0,0.5,1,2,4 and 6 h. Samples were transferred into acetonitrileand pclletedby centrifugation (5 min). The supematant was transferred to a new 9fi DeepWell™ plate, evaporated under nitrogen and
resuspended. Décliné of compound was analyzed by HPLC-MS/MS. CLfM (in vitro hepalic intrinsic clearance) was calculated os follows:
CU = Dose / AUC = (Q/CD) / (AUD + Cbu/k) x 1000/60
Co: initial concentration In the incubation [μΜ];
CD: cell denstty of vital ceils [cells/mLJ;
AUD: area under the data [μΜ x h];
Cu,,: concentration of iast data point [μΜ];
k: slope of the régression line for compound décliné [h*1].
The calculated in vitro hepatic intrinsic clearance was scaled up to the Intrinsic in vivo hepalic 10 clearance (CU in ^«,) and used to predict hepatic in vivo blood clearance (CL) by the use of a liver mode! (well-stirred mode!), ns follows:
CLtmjn «ri«o [mL/min/kg] s (CU [pL/min/IO’cells] x hepatocellularity [10e ccils/g liver] x liver factor [g/kgbody weight])/ 1000
CL [mL/min/kg] = CLlntinV|ÏO [ml/min/kg] x hepalic blood flow [ml/min/kg] / (CU™ [mi/min/kg] 15 + hepatic blood flow [mL/min/kg])
The in vivo blood clearance was transformed into percent of the hepatic blood flow (% Qh):
% Qh = CL [mL/min/kg] / hepatic blood flow [mL/min/kg]) x 100
Hepatocellularity, human: 1.2 xl 0T cells/g liver,
Liver factor, human: 25.7 g/kg body weight;
Hepatic blood flow, human: 21 mL/(min x kg).
Rat Brain A fl Lowering Assay (Assay 5)
The in vivo efïîcacy of compounds of the invention was demonstrated in a rat brain Αβ lowering (réduction) assay, and the data are presented in Table 3. Male Sprague-Dawley rats, 5 to 6 weeks of âge, were used to démon strate the abiiity of compounds of the invention to reduce brain 25 amyloid peptides Αβί-χ. Compounds were administered via oral gavage in 1 % Polysorbate-80 and 0.5% Natrosol*, at the single dosages indicated in Table 3. The animais were sacrificed 3 hrs after dosing. and brnins were excised, dissected into cercbellum and left and right cerebra and flash-frozcn In liquid nitrogen.
The cerebrum was homogenized (5 volumes per weight) in 20 mM Tris-HCL, pH 8.5,0.2% 30 Triton-XI00 supplementcd with protease inhibitors (cOmplete, Roche Applicd Science) at 4*C using a glass Dounce homogenizer. The homogenate was ccntrifuged at 120,000xg for 60 min at 4*C, and
the supematant was collected and analyzed For Abl-X using immunoassay with chemiluminescent détection (Meso-Scale Discovery, Rockville, MD (MSD)).
Streptavidin 96-well plates (MSD) were pre-blocked with 5% Blocker A solution (MSD) for 1 hr at rt on an orbital shaker and washed 4 times with phosphate buffered saline (PBS). The wells were pre-coated with 20 ng/well ofbiotinylated antibody SIG-39155 (Clone M3.2, spécifie For amino acids 10-15 of the rodent Αβ) For I hr at rt and washed 4 times with PBS. For ΑβΙ-χ analysis, 25 μ! of either the cleared braîn lysâtes or ΑβΙ-40 standards (8-500 pg/ml, with 2x incrément) were incubated For I hr at rt with constant shaking. The wells were washed 4 times with PBS, and 25 μΙ of the détection antibody (Sulfo-TAG labeled αη(ί-Αβ40 antibody supplied by MSD) was added and incubated for I hr at rt. After 4 woshes with PBS, 150 μΙ of the chemiluminescence détection reagent (Rend Buffer T, MSD) was added, and the plate was read on an MSD Sector lmager 6000 instrument. The calibration curve was fit into a non-iinear four-parameter régression model, and the ΑβΙ-χ concentrations were calculated for each well containing the cleared brain lysâtes. The percent of Αβ lowering was calculated based on the différence with the average Αβ concentration obtained for the IS brains from the animais treated with vehicle alone.
Table I shows the following properties of the compounds of the présent invention: BACE1 inhibitory potency as measured in assay I, hERG inhibition as measured in assay 2, first effect concentration (FEC) of phospholipidosis as measured in assay 3, and metabolic stability as measured în assay 4.
Table 1.
Example H BACE1 IC»nM (assay 1) % Inhibition hERG @ 10 μΜ (assay 2) Phospholipidosis FEC ICjopM (assay 3) In vitro Human Hépatocytes %Qh@lpM (assay 4)
1 14.6 13 400 0
2 10.3 4.5 400 1.6
3 3.0 20 200 3.1
4 2.7 13 800 6.1
5 2.6 12 400 6.1
6 6.3 1.8 400 11
7 3.4 15 400 13
8 1.9 6 800 19.1
9 10.7 2.5 400 12.4
10 10.6 33 >100 0
11 14.6 19 200 0
Example U BACEI ICm nM (assay 1) % Inhibition hERG @ 10 μΜ (assay 2) Phospholipidosis FECICmMM (assay 3) In vitro Human Hépatocytes %Qh@lpM (assay 4)
12 6.8 15 100 0
13 8.7 12 200 4.2
14 4.S 27 200 5.2
IS 9.7 15.2 800 22.9
16 9.4 1.4 200 19
Table 2 provides data showing that compounds of the présent invention hâve at least one of the following properties relative to certain comparator compounds described in WO20I0/105I79:1) significantly lower ICjo Inhîbitory values in a BACEi enzymatic assay, slgnificanlly lower percent inhibition of hERG. significantly lower propensity to cause phospholipidosis, and significantly greater metabolic stability relative.
Table 2.
Example U BACEI IC50 nM (Assay 1) % inhibition hERG© 10 μΜ (Assay 2) Phospholipidosis FECICSOpM (Assay 3) In vitro Human Hépatocytes % Qh@ 1 μΜ (Assay 4)
Comparlson 1
1 14.6 13 400 0
8.3 87 - -
428 in W02010/105179
512 In W02010/105179 3.5 89 88
Comparlson 2
10 10.6 33 >100 0
o x & 107 - -
121 In W02010/105179
H1W 16 90 400 14
174 In W02010/105179
Comparlson 3
3 3.0 20 200 3.1
251 In W02010/105179 256 - - -
Comparlson 4
12 6.8 15 100 0
1.1 60 25 36
255 In W02010/105179
H2N n 5.1 60 8.5
249 In W02010/105179
Comparlson 5
2 10.3 4.5 400 1.6
F H,N 602 în W02010/105179 73 36 50 43
174 In W02010/105179 16 90 400 14
Comparlson 6
11 14.6 19 200 0
it 172 In W02010/105179 19 58 100 -
HjN o 249 In WO2010/105179 5.1 60 8.5
Comparlson 7
8 1.9 6 800 19.1
174 In W02010/105179 16 90 400 14
251 In W02010/105179 256 - - -
The ability of compounds of the invention to reduce brain Αβ was demonstrated in rats, us described in Assay 5, and the in vivo effîcacy data are presented in Table 3.
Table 3.
Example Dose (mg/kg) % Αβ Réduction
1 25 25
2 12.5 40
4 12.5 21
7 25 58
9 25 42
Method of Treatment
The présent invention is directed to compounds which are useful in the treatment of disorders or diseases characterized by elevated β-amyloid deposits or β-amyloid levels ïn a subject wherein the 10 inhibition of the activity of the β-secretase enzyme (BACEl) is of therapeutic benefit, including but not limited to the treatment, amelioration or prévention of neurodegenerative disorders, disorders characterized by cognitive décliné, cognitive impairment, dementia and diseases characterized by production of β-amyloid deposits and/or neurofibrillary (angles.
Compounds of lhe présent invention are useful for treatment of Aizheimer’s disease, Trisomy 21 (Down Syndrome), Hereditary Cérébral Hemonhage with Amyloidosis of the Dutch-type (HCHWA-D), senile dementia, cérébral amyloid angiopathy, dégénérait ve dementla, dementias of mixed vascular and degenerative engin, dementia associated with Parkinson's disease, dementia associated with progressive supronuclearpalsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Aizheimer's disease, dry âge related macular degeneration (AMD), and giaucoma. The “dry form of AMD, also known os central géographie atrophy, results from atrophy to the retinal pigment épithelial layer below the neurosensory retina, which causes vision loss through loss of photoreceptors (rods and cônes) in the centrai part of the eye. No medical or surgical 10 treatment is currently available for this condition. Treatments available so for (e.g., suggested by the National Eye Institute) include the use of vitamin suppléments with high doses of ontioxidonts, lutein and zeaxanthin, which may slow the progression of dry macular degeneration. Giaucoma is a disease where fluid pressure inside the eye increases, causing irréversible damage to the optic nerve and loss of vision. Abeta coiocalizes with apoptotic retirai ganglion cells in experimental giaucoma and induces significant retina) ganglion cell apoptosis in a dose- and time-dependent manner.
Accordingly, the présent invention relates to a compound or a pharmaceutically acceptable sait thereof os a médicament
Furthermore, the présent invention relates to the use of a compound in the treatment of a disease and/or condition wherein the inhibition of the activity of the β-secretase enzyme (BACE1) is 20 of therapeutic benefit.
Furthermore, the présent invention relates lo the use of a compound in the treatment of ncurodegenerative disorders, disorders characterized by cognitive décliné, cognitive impairment, dementia and diseases characterized by production of β-amyloid deposits or neurofibriliary langles.
Therefore, the présent invention relates to the use of a compound of the présent invention in the treatment of Aizheimer’s disease, Trisomy 21 (Down Syndrome), Hereditary Cérébral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), senile dementia, cérébral amyioid angiopathy, degenerative dementia, dementias of mixed vascular and degenerative origin, dementia associated with Parkinson's disease, dementia associated with progressive supnmuclear paisy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Aizheimer's disease, 30 dry AMD, and giaucoma.
The présent invention also provides a method for the treatment of a disorder related to or associated with excessive BACEI activity in a patient in need thereof which comprises administering to said patient on effective amount of a disclosed compound or a pharmaceutically acceptable sait thereof. The présent invention also provides methods for inhibiting the activity of BACEI in a 35 subject in need thereof, comprising administering to a subject and/or contacting a receptor thereof
with an effective amount ofat least one disclosed compound or a pharmaceutically acceptable sait thereof. The présent Invention also provides methods of ameliorating β-amyloid deposits in a subject in need thereof, comprising administering to said subject an effective amount of at least one disclosed compound or a pharmaceutically acceptable sait thereof.
S The invention includes a therapeutic method for treating or amelioratingan BACEl mediated disorder in a subject tn need thereof comprising administering to a subject in need thereof an effective amount of a compound ofthe invention described herein, or pharmaceutically acceptable salts thereof or composition thereof.
As used herein, the term “subject” and “patient may be used interchangeably, and means a 10 mammal in need of treatment, e.g., companion animais (e.g., dogs, cats, and the iike), farm animais (e.g., cows, pigs, horses, sheep, goats and the like) and laboratory animais (e.g., rats, mice, guînea pigs and the like). Typically, the subject is a human in need of treatment.
As used herein, the term “treating or ’trcatmenf refera to obtaining desired pharmacological and/or physiological effect. The effect can be prophylactic (Le., reducing the likelihood of developing 15 the disorder or disease) or therapeutic, which includes achieving, partially or substantîally, one or more of the following results: partially or totally reducing the extent of the disease, disorder or syndrome; ameliorating or improving a clinical symptom or Indicator ossociated with the disorder; or delaying, inhîbiting or decreasing the likeiihood ofthe progression ofthedisease,disorder or syndrome.
The dose range of the compounds according to the présent invention applicable per day is usually from 0.1 to 3000 mg, preferably from I to 2000 mg, more preferably from 10 to 1000 mg, most preferably, 50 or 500 mg. Each dosage unit may convenientiy contain from 0.1 to 1000 mg, preferably 25 to 250 mg.
The actual pharmaceutically effective amount or therapeutic dosage will of course dépend on 25 factors known by those skilled in the art such os âge and weight of the patient, route of administration and severity of disease. In any case the combination will be administered at dosages and in a mnnner which allows a pharmaceutically effective amount to be delivered based upon patient's unique condition.
Pharmaceutical Compositions
Suitable préparations for administering the compounds of the présent invention will be apparent to those with ordinary skill in the art and include for example tablcts, pills, capsules, suppositories, iozenges, troches, solutions, syrups, élixirs, sachets, injectables, inhatatives and powders, etc. The content of the pharmaceutically active compound(s) should be in the range from 0.i to 95 wt.-%, preferably 5 to 90 wl.-% of the composition as a whole.
Suitable tablets may be obtained, for exampie, by mixing one or more compounds of the invention with known excipients, for example Inert diluents, carriers, disintegrants, adjuvants, surfactants, binders and/or fabricants, The tablets may also consist of scveral layers.
Combination Therapy (n one embodiment, the présent invention includes combination therapy for treating or ameliorating a disease or a disorder described herein. The combination therapy comprises admînistering a combination of at least one compound of the présent invention with one or more agent selected from the group of, for example, gamma-secretase inhibitors or modulators; amyloid aggregation inhibitors blocking the formation of Abeta oligomers or Abeta fibrils (e.g., ELND-005);
directly or indirectly acting neuroprotective and/or disease-modifying substances; anti-oxidants (e.g., vitamin E or ginkolide); anti-inflammatory substances (e.g., Cox inhibitors, NSAIDs additionally or exclusively having Abeta lowering properties); HMG-CoA reductase inhibitors (statins); ncetylcholinesterase inhibitors (e.g., donepezil, rivastigmine, tacrine, and galantamine); NMDA receptor antagonists (e.g., memantine); AMPA receptor agonists; AMPA receptor positive modulators, AMPAkines, monoamine receptor reuptake inhibitors, substances modulating the concentration or release of neurotransmitters; substances inducing the sécrétion of growth hormone (e.g., ibutamoren mesylate and capromorelin); CB-1 receptor antagonists or inverse agonists; antibiotics (e.g., niinocyclin or rifampicin); PDE2, PDE4, PDE5, PDE9, PDE10 inhibitors, GABAA receptor inverse agonists, GABAA receptor antagonists, nicotinic receptor agonists or partial agonists or positive modulators, alpha4beta2 nicotinic receptor agonists or partial agonists or positive modulators, alpha7 nicotinic receptor agonists or partial agonists or positive modulators; histamine H3 antagonists, 5 HT-4 agonists or partial agonists, 5HT-6 antagonists, a!pha2-adrenoreceptor antagonists, calcium antagonists, tnuscarinic receptor ΜI agonists or partial agonists or positive modulators, muscarinic receptor M2 antagonists, muscarinic receptor M4 antagonists, metabotropic gfatamate-receptorS positive modulators, antidepressants, such as citatopram, fluoxetine, paroxetine, sertraline and trazodone; anxiolytics, such as lorazépam and oxazepam; antiphychotics, such as aripiprazole, clozapine, haloperidol, olanzapine, quetiapine, rispéridone and ziprasidone, and other substances that modulate receptors or enzymes in a manner such that the efficacy and/or safety of the compounds according to the invention is increased and/or unwanted side effects are reduced. The compounds according to the invention may also be used in combination with immunothérapies (e.g„ active immunization with Abeta or parts thereof or passive immunization with humanized antî-Abeta antibodies or nanobodies) for the treatment of the above-mentioned diseases and conditions.
Combination therapy includes co-administration of the compound of the invention with one or more other agent sequentîal administration of the compound and one or more other agent, administration of a composition containing a compound and one or more other agent, or simultaneous administration of separate compositions containing the compound and one or more other agent.
EXPERIMENTAL SECTION
Methods of Préparation of Compounds
Compounds of the invention can be prepared employing conventional methods that utilize readiiy available reagents and starting materials. The reagents used in the préparation of the intermediates of this invention can be either commerciaily obtained orcan be prepared by standard procedures described in the literature.
Microwave reactions were carricd out in CEM reactor using discovery SP system or in Biotage, Initîator 60 EXP. Where NMR data are presented, spectra were obtained in Vorian -400 (400 MHz). Spectra are reported as ppm downfield from tetramethylsilane with number of proton, multiplicities and, in certain instances, coupling constants indicated parenthetically along with reFerence to deuterated solvent Compounds were purified by basic préparative HPLC method as described below.
Method I:
Mobile phase A: water with 0.05% NH4OH; Mobile phase B: ACN; Flow rate: 25 mL/min; Détection: UV 220 nm / 254 nm; Column: Phenomenex Gemini C18 250*30mm*5um; Column température: 30 ’C.
Time in min %A %B
0.0 68 32
12.00 38 62
12.20 0 100
13.5 0 100
13.7 90 10
Method 2:
Mobile phase A: water with 0.05% NHiOH; Mobile phase B: ACN; Flow rate: 25 mL/min; Détection: UV 220 nm /254 nm; Column: Durashell Cl8 250*30mm*5um; Column température: 30 ’C.
Time in min %A %B
0.0 67 33
12.00 47 53
12.20 0 100
I3J 0 100
13.7 90 10
LC-MS data were obtained by utilizing the following chromatographie conditions:
Method 1:
HPLC System: Waters ACQUITY; Coiumn: Waters ACQUITY CSH™ C18 1.7 pM.
Guard coiumn: Waters Assy. Frit, 0.2 μΜ, 2.1 mm; Coiumn tem: 40 °C.
Mobile Phase: A: TFA: Water (1:1000, v:v) Mobile phase B: TFA: ACN (1:1000, v:v): Flow Rate:
0.65 mL/min; Injection Volume: 2 pL; Acquisition time: approximately 1.5 minute.
Gradient Program:
Time (min) B%
0 10
0.8 90
1.20 90
1.21 10
Mass Spectrometer Parameters
Mass Spectrometer: Waters SQD; Ionization: Positive Electrospray Ionization (ESI); Mode Scan (100-1400 m/z in every 0.2 second); ES Capillary Voltage: 3.5 kV; ES Cône Voltage: 25 v.
Source Température: 120 eC; Disolvation Température: 500 °C; Desolvation Gas Flow: Nitrogen Setting 650 (L/h); Cône Gas Flow: Nitrogen Setting 50 (L/h).
Method 2:
HPLC System: Waters Alliance with DA- and MS-Detector; Coiumn: Waters XBridge CI8 4.6 x 30 mm, 3 J pm; Coiumn temp: 60 “C.
Mobile Phase: A: TFA: Water (1:1000, v:v) Mobile phase B: MeOH; Flow Rate: 4 mL/min.
Gradient Program:
Time (min) B%
0 5
1.6 100
i.85 100
i.9 5
Method 3:
HPLC System: Waters Alliance with DA- and MS-Detector; Column: Waters XBridge CI8 4.6 x 30 mm, 3.5 pm; Column temp: 60 eC.
Mobile Phase: A: TFA: Water (l: 1000, v:v) Mobile phase B: ACN; Flow Rate: 5 mL/min.
Gradient Program:
Time (min) B%
0.23
1.6100
1.7100
Method 4:
HPLC System: Waters Alliance with DA- and MS-Detector; Column: Waters XBridge C18 4.6 x 30 mm. 3.5 pm; Coiumn temp: 60 °C.
Mobile Phase: A: TFA: Water ( l : 1000, v:v) Mobile phase B: MeOH; Flow Rate: 4 mL/min.
Gradient Program:
Time (min) B%
0.25
15100
1.75100
1.855
SFC séparation and characterization of compounds were carried out under the following methods:
Method A:
Instrument: Thar SFC 80; Column: AD 250mm*30mm, 5um; Mobile phase: A: Supercritica! COj. B: 25 IPA (0.05% DEA), A: B =80:20 at 60ml/min; Column Temp: 38 °C; Nozzle Pressure: 100 Bar;
Nozzle Temp: 60 °C; EvaporatorTemp: 20 eC;TrimmerTemp: 25 “C; Wavelength: 220 nm.
Method B:
Instrument: SFC MG2; Column: OJ 250mm*30mm, 5um; Mobile phase: A: Supercritical COj, B: MeOH(0.05% DEA), A:B =90:10 at 70ml/min; Column Temp: 38 ’C: Nozzle Pressure: 100 Bar Nozzle Temp: 60 ’C; Evaporator Temp: 20 ’C; Trimmer Temp: 25 ’C; Wavelength: 220nm.
The invention is illustrated by way of the following examples, in which the following abbreviations may be employed:
Abbreviation Meaning
ACN acetonitrile
Boc tert-butoxy carbonyl or t-butoxy carbonyl
BocîO d i-tert-buty l-d icarbnate
brine saturated aqueous NaC!
DAST (dielhylamino) sulfur trifluoride
DCM methylene chloride
DŒA diisopropyl ethyl amine
DMA dimethyl acetamide
DMF dimethyl formamide
DMSO dimethyl sulfoxide
Et ethyl
dppf t, 1 -bi s(d i phenyl phosphino)fcraocene
EDCI 1 -(3-dimethylaminopropyl)-3-ethy!carbodiÎimidc hydrochloride
EtI ethyl iodide
EtjN triethylamine
EtjO ethyl ether
EtOAc ethyl acetate
EtOH éthanol
h hour(s)
HPLC high performance liquid chromatography
Abbreviation Meaning
LDA lithium diisopropylamide
min minute
MeOH methanol
Mel methyl iodide
Me methyl
Me^ dimethyl sulflde
MsCI methane sulfonyl chloride
mL milliliters
mmol millimoles
mg milligram
NaOMc sodium meth oxide
NCS N-chlorosuccinamide
PdClîdppf [ 1,1 -bis( diphe ny 1 phosphino) ferrocene] dichloropalladi um(IT)
Pd2(dba)j t ri s(d ibenzy 1 ideneacetone)di pal 1 adium(O)
PE pctroleum ether
rt room température
sat. saturated
SFC super critical fluid chromatography
r-BuOK potassium tert butoxide
r-BuLi tert butyl lithium
/•BuNHrBH3 tert butylamin-borane complex
r-BuOOH tert butyl peroxide
TEA triethy lamine
TFA trifluoroacetic acid
TFAA trilluoroacetic acid anhydride
THF tetrahydrofuran
Abbreviation Meaning
Ti(OEt)4 titanium tetra ethoxide
TLC thin layer chromatography
TMSI trimcthylsilyl iodide
v volume
XPhos d icyclohexy 1 phosph i no-2’,4',6'-tri iso-propy 1-1, Γ-bîpheny 1
Zn(CN)î zinc cyanide
To a mixture of 6-bromo-indan-l -one ( 100.00 g, 473.8 mmol) in anhydrous THF ( I L) at 0 °C was added r-BuOK (583 g, 521.2 mmol). After 5 minutes, the mixture was warmed to rt and was stirred for another 10 min before methyl méthacrylate (49.8 g, 53.2 mL, 4973 mmol, 1.05 eq) was added in one portion. After 2 h, methyl acrylate (49.0 g, 51.2 mL, 568.6 mmol, 1.2 eq) was added to the reaction mixture. After 3 h of stirring at rt, Mel (101 g. 44.3 mL, 710.7 mmol, 13 eq) was added to the reaction mixture, and the mixture was further stirred for 16 h. H2O ( I L) was added followed by LiOH*HjO (793 g, 1895 mmol, 4.0 eq). The mixture was stirred for 28 h at rt THF was removed under reduced pressure. The residue was diluted with HjO ( t L), fîltered, and washed with H2O until the filirate was neutral. The product was washed with MeOH to afford 50 g of intermediate 3.
A mixture of FeClj (6.0 g, 37.0 mmol) and toluene (60 mL) was cooled to 0 °C. A mixture of intermediate 3 (l 1.9 g, 37.0 mmol) in THF (48 mL) was added to the mixture. The mixture was stirred for 5 min at 0eC and then cooled to -10 eC. A solution of t-BuNHrBHj (3.5 g, 40.7 mmol) in THF (12 mL) was ndded dropwîse to the réaction mixture at -10 eC. The réaction mixture was stirred at about -10 “C for 30 min. quenched with aqueous HCl solution (6N. 10 mL), stirred at about 0 °C for 30 min, and then allowed to warm to rt. The mixture was concentrated to remove THF, and toluene (60 mL) was added. The aqueous layer was removed, and the organic phase was washed with water (3 x 60 mL). The organic phase was concentrated to half volume, heated to 50 °C to obtain a solution, and then cooled to 0 °C over 1 h and held at 0 °C for 1 h. The solid was fîltered and washed with cold (0 °C) toluene ( 12 mL), and dried under vacuum to give compound 4 (9.93 g).
LC-MS (method 1): tR« 1.24 min, MS (ESI) m/z 323.1 [M+HJ*. ‘H-NMRtCDCDtS: 7.889-7.894(s. 1 H),7.671-7.696(d. IH), 7311-7332 (d, IH), 3.6O5(s, IH), 2.981 (s,2H), 1.769-1.797 (m,4H), 1.072-1.082 (m,2H), 1.019-1.056 (m,6H).
Step 3: Synthesis of intermediate 5,
A mixture of intermediate 4 (6.0 g, 18.6 mmol) and Cul (0.71 g, 3.72 mmol, 0.2 eq) in ACN ( 120 mL) was heated to 60 °C and 2-(nuorosulfonyl)difIuoloacelic acid (13.2 g, 74.4 mmol) wus added. The mixture was stirred at 60 ÙC for 20 min. The mixture was cooled, quenched with H2O and extracted with EtOAc. The combined organic phases were washed with HjO and brine, dried over anhydrous Na2SO4> concentrated to afford 15 g crude product, which was purified by column on slica gel (eluent: petroleum ether: ethyl acetate from 300: I to 50: 1) to afford intermediate 5 (4.6 g).
Step 4: Synthesis of intermediate 6.
The mixture of intermediate 5 (3.4 g, 9.2 mmol) and titanium (IV) ethoxide (21 g, 92 mmol) in dry THF (40 mL) was stirred at rt for I h. (S)-AMert-butylsu]finamide (4.5 g, 36.8 mmol) was added and the resulting mixture was stirred at 80 ’C under N2 atmosphère for 12 h. The reaction mixture was cooled and water (400 mL) was added. The mixture was filtered and the aqueous layer was extracted with ethyl acetate (3 x 400 mL). The separated organic phase was dried over NajSO4 and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether: ethyl acetate = 20:1) to yield intermediate 6 (3.9 g).
Step 5: Synthesis of intermediate 7.
A r-BuLi solution (32 mL, 41.0 mmol, 1.3 M in hexane) was added dropwise to a solution of ethyl vinyl ether (7.05 g, 82 mmol) in anhydrous THF (50 mL) at -78 ’C under N2 atmosphère and the mixture was stirred for 20 min. The resulting mixture was stirred at 0 ’C for another 45 min and then cooled to -78 ’C. A pre-cooled solution at -78 ’C, containing intermediate 6 (3.9 g, 8.2 mmol) in anhydrous THF (80 mL) was added dropwise and the mixture was stirred for 2 h at -78 ’C. The reaction was quenched with sat. NH4CI (50 mL) aqueous solution and extracted with ethyl acetate (3 x 300 mL). The organic phases were combined and concentrated under reduced pressure. The crude product was purified by preparative HPLC (method 2) to afford intermediate 7 (33 g).
Intermediate 7 (10 g, 18.2 mmol) was added to a solution of MeOH in DCM (5:1, 100 mL) and cooled to *78 °C. Ozone was bubbled through the mixture for 20 min. After 10 minutes of 5 additional stirring. the mixture was purged with Nj for 15 minutes and then treated with MejS (20 mL) al -78 °C. It was allowed to warm to rt and stirred for 3 h at rt. The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica gel (petroleum ether ethyl acetate = 20:1 to 5:1) to yield intermediate 8 (6 g).
Step 7: Synthesis of intermediate 9.
Concentrated sulfuric acid(48 pL) was added to DMA (20 mL) and the solvent was purged with Nj for 20 min. A 50 mL round bottom flask was charged with Pd(OAc)i (03 g) and Xphos (1.25 g) under N2, then the above solvent was transferred in. The resulting mixture was heated at 80 °C for 30 min to give mixture A.
Γη nn another flask, DMA (50 mL) was purged with N2 and intermediate 8 (2.2 g, 4.0 mmol),
Zn(CN)i (0.5 g, 4.0 mmol) and zinc dust ( 14.1 mg) were added. The mixture A was added to this solution, and the resulting mixture was heated at 90 °C for I h. The reaction mixture was cooled to rt, diluted with water (100 mL) and ethyl acetate ( 100 mL) and stirred for 10 minutes. The mixture was filtered through celite, and the organic layer was separated. The aqueous layer was extracted with ethyl acetate (2 x 30 mL). The combined organic layer was washed with water, brine and dried and the solvent was removed under reduced pressure. The residue was purified on flash column on silica gel (petroleum e then ethyl acetate; 20:1 to 3:l)to yield intermediate 9 (1.5 g).
Step 8: Synthesis of intermediate 10.
To a mixture of intermediate 9 (0 J g, 1.01 mmol) In MeOH (l I mL) was added HCl In dioxane (4 M, 2.25 mL). The resultîng mixture was stirred for I h. The solvent was removed under 5 reduced pressure to afford crude intermediate 10 (529 mg) which was used for the next step without further purification.
Step 9: Synthesis of intermediate 11.
To a solution of intermediate 10 (529 mg, 135 mmol) in DCM (6 mL), H:O (6 mL) and
NaHCOj ( 1.13 g, 13 J mmol) were added at rt. Thiophosgene (310 mg, 2.7 mmol) was added with vigorous stirring and the mixture was stirred for I h. The organic loyer was separated and the aqueous layer was extracted with DCM (3 x 40 mL). The organic layers were combined and washed with brine (2 x 40 mL), dried and the solvent was removed under reduced pressure to afford crude intermediate 11 (520 mg), which was used for the next step without further purification.
Step 10: Synthesis of intermediate 12.
OEt
Et3N, THF
To a mixture of intermediate 11 (200 mg, 0.52 mmol) in THF (10 mL) was added 3,3,3trifiuoro-propylamine hydrochloride (156 mg, 1.04 mmol) and TEA (526 mg, 52 mmol). The mixture was stirred ovemight at rt. The réaction was diluted with water and extracted with EtOAc (30 mL). The organic layers were combined, washed with brine (10 mL), dried over Na2SO.i, filtered and concentrated under reduced pressure to afford crude product. The residue was purified by préparative TLC (petroleum ether ethyl acetate; 1; I) to afford Intermediate 12 (265 mg).
Step 11: Synthesis of Example 1,
To a mixture of intermediate 12 (265 mg, 059 mmol) in MeOH (10 ml), aqueous ammonium hydroxide (1.5 mL) and r-BuOjH (0.8 mL, 5.0 M solution in nonane) were added. The mixture was stirred at rt for 16 h and then quenched with sat. aqueous NajSîOj solution (05 mL). The residue was partitioned between EtOAc (20 mL) and HjO ( 10 mL). The organic layer was separated and washed with brine (10 mL), dried over NajSOj, filtered and concentrated under reduced pressure. The residue was purified by préparative HPLC (method 2) to give Example 1 (86.9 mg).
LC-MS (method 1): tR = 0.92 min, MS (ESI) m/z 435.2 [M+HJ*.
'H-NMR (CDjOD):57.65(d, J=7.6 Hz, IH),7.49(d,J«7.6Hz, !H),730(s, IH),3.82-3.85 (U = 7.6Hz.2H), 3.24 (s, IH), 3.15 (d, J = 16.0 Hz. IH). 256 (m. 3H). 1.23-1.79 (m,5H), 0.956-1.02(m, 7H).
”F NMR: 3 -66.64.
Synthesis of Intermediate 20 Step 1: Synthesis of intermediate 13.
To a mixture of intermediate 4 (20.0 g, 61,9 mmol) in DMF (200 mL) was added NaH (5.0 g.
123.8 mmol)atO°Cand the mixture was stirred for 15 minât 0eC. Methyl iodide(17.6 g. 123.8 mmol) was added at 0 C and the mixture was warmed to rt and stirred for 13 h at rt The mixture was quenched with HjO and extracted with EtOAc. The combined organic phases were washed with HjO followed by brine, dried and concentrated to afford crude product, which was purified by column on silica gel (petroleum ether ethyl acetate; 30:1 to 5:1) to afford intermediate 13 (20 g).
A mixture of intermediate 13 (20.0 g, 59.3 mmol) and titanium (IV) ethoxide (108.2 g, 474.4 mmol) in dry THF (200 ml) was stirred at rt for I h. (S)-N-tert-butylsulfi numide (29 g, 237.2 mmol) was added and the resulting mixture was stirred at 80 ’C under N2 atmosphère ovemight. The reaction mixture was cooled and water (400 mi) was added. The mixture was filtered and the aqueous layer was extracted with ethyl acetate (3 x 400 mL). The combined organic phase was dried and concentrated under reduced pressure to give crude intermediate. This was purified by column chromatography on silica gel (petroleum ether ethyl acetate; 20:1 ) to yield intermediate 14 (18.4 g).
r-BuLi (131 mL, 170.3 mmol, 1.3 M in hexane) was added dropwise to a solution of ethyl vinyl ether (123 g, 170.3 mmol, 5.0 eq) in anhydrousTHF ( 100 mL) at -78 ’C under N2 and the mixture was stirred for 20 min. The resulting mixture was stirred at 0 ’C for another45 min and recooled to -78 ’C. A pre-cooled solution of intermediate 14 ( 15.0 g, 34.1 mmol) in anhydrous THF (50 mL) at -78 ’C was added dropwise and the mixture was stirred for 2 h at -78 ’C. The reaction mixture was quenched with saL NH4CI aqueous solution (50 mL) and extracted with EtOAc (3 x 300 mL). The organic phases were combined and concentrated under reduced pressure to give the residue, which was purified by column chromatography on silica gel (petroleum ether ethyl acetate; 50:1 to
3:1) to afford intermediates 15 ( 11 g) and 15A ( 1.44 g), respectively.
LC-MS (method 1) tR = 5.67 min; MS (ES1) m/z 514.2 [M+H]*. ' 'H-NMR(CDjOD): 8 7546 (s, IH), 7.454-7.479 (d, IH), 7.208-7.228 (d, IH), 4.620-4.755 (d, IH), 4373-4381 (m, IH),4.048-4j055(m, 1 H), 3.844-3.903 (m,2H), 3.458-3474 (s, 3H), 2.986-3.000 (m, 2H),2326-2377 (m, IH), 1.969-2.001 (m, IH), 1.671 (s, IH), 1457-1520 (t. J = 12 Hz, 3H), 13731.408 (m,2H), 1328 (s,9H), 1.169-1378 (m,5H), I.O73-l.lO6(d, 3H).
Step 4: Synthesis of intermediate 16.
Intermediate 15 (4.8 g, 937 mmol) was added to a mixture of DCM in MeOH (5:1,40 mL), and the mixture was cooled to -78 °C. Ozone was bubbled through the mixture for 20 min. The mixture was purged with N2 for 10 minutes and treated with Me2S (10 mL) at -78 eC. The mixture was allowed to worm up to rt and stirred at rt for 3 h. The solvent was removed under vacuum, the residue was purirled by column chromatography on silica gel (petroleum ether ethyl acetate; 20:1 to 8:1) to give intermediate 16 (35 g).
LC-MS (method I): tR = 130min; MS (ESI) m/z5!6.l [M+H]*.
’H NMR(CDClj): 6 7.84 (s, IH), 7.42-7.44 (d. J = 8.0 Hz, 1 H), 7.09-7.11 (d, J = 8.0 Hz, 1 H), 4.40(s, IH), 436-439 (m, 2H), 3.44 (s, 3H), 2.93-2.97 (d, J = 15.6 Hz, IH), 2.70-2.74 (d, J = 15.2 Hz, IH), 232-230 (t, J a 10.0 Hz, IH). 1.75-1.79 (m, IH), 1.61-1.66 (m, IH), 154-137(m,2H), 132-138 (m,4H), LI4(s,9H), 1.06-1.08 (d, J = 6.0Hz, 3 H), 0.89-0.91 (d, J = 6.0 Hz, 3 H), 0.67-0.74 (m, IH).
Step 5: Synthesis of intermediate 17.
To a mixture of intermediate 16 (5.1 g, 10 mmol) in MeOH (10 mL) was added HCl in dioxane (4.0M, 8.0 mL). The resulting mixture was stirred for I h. Solvent was removed under reduced pressure to afford crude intermediate 17 (6.0 g), which was used for the next step without further purification.
Alternative synthesis of Intermediate 17
Step L Synthesis of intermediate 18.
A mixture of Intermediate 14 (5.00 g, 11.4 mmol), diethoxyacetonitrile (3.5 mL, 24.4 mmol) and THF (50 mL) was cooied to -7 °C and treated dropwise with LDA (25.0 mL, 45.0 mmol, 1.8M in THF/beptane/ethylbenzene). The mixture was stirred at -7 to -2 °C for 2 h, and then quenchcd with water (50 mL) and sat. aqueous NH4CI (25 mL). Hexanes (100 mL) was added, and the layers were separated. The organic layer was washed with water, brine, and was concentrated under reduced pressure to give crude intermediate 18 (9.00 g) which was used directly in the next step.
LC-MS (method I): tR = 3.74 min. MS (EST) m/z 523.2/525.2 [M-OEt+H]*
Step 2. Synthesis of intermediate 17.
A mixture of above intermediate 18 (9.00 g, 11.4 mmol) in EtOH (30 mL) was treated with aqueous HCl (6 N, 20 mL). The reaction mixture was heated at 75 eC for 24 h and cooied to rt The réaction was extracted with toluene (50 mL), and the aqueous phase was basifïed (pH = 8) with
aqueous NaOH (2 N, **60 mL). Toluène ( 100 mL) was added and the mixture was stirred for 10 minutes. The organic layer was separated and washed with aqueous NaHCOj, brine and concentrated under reduced pressure. Hexanes was added and the solution was concentrated under reduced pressure to give crude intermediate 17 (3.47 g) which was used dïrectly in the next step.
LC-MS: tR a 0.86 min, MS (ESD m/z 410.2/412.2 [M+H]*
Step 6: Synthesis of intermediate 19.
A mixture of compound 17 (500 mg, 1.9 mmol) under nitrogen. Zn(CN)j (300 mg. 2.6 mmol), Pdj(dba)j (150 mg, 0.16 mmol),dppf (160 mg, 0.32 mmol) and Zndust (60 mg, 0.9 mmol) in 10 DMF ( 15 mL) was heated to 120 °C for 3 h in CEM micro wave reactor. The mixture was concentrated under vacuum and the residue was purified by column on silica gel (eluent: petroleum ether: ethyl acetate: 20:1 to 8: I ) to afford intermediate 19 (300 mg).
LC-MS: tR a 0.880; MS (ESI) m/z 308.1 [M+H] *.
Το a solution of intermediate 19 (3.1 g. 7.4 mmol) In CHClj (20 m!) was added TMSI (10 mi) and stirred at 65 “C for 2 h. The mixture was cooled to rt and sat NajSjOj (10 mL), sat. NaHCOj aqueous solution ( 10 mL) were added, and the mixture was stined for 10 minutes. The residue was 20 partitioned between DCM (40 mL) and HjO ( 10 mL). The organic layer was separated and washed with brine (10 mL), dried over NûjSOî, filtered and concentrated under vacuum to afford crude intermediate 20 (2.6 g), which was used for the next step without further purification.
Synthesis of Intermediate 26
Step 1; Synthesis intermediate 22.
A mixture of intermediate 21 (2.0 g, 10.6 mmol) in anhydrous THF (20 mL) was added to methyl magnésium bromide (14 mL, 42 mmol, 3.0 M in EtjO) at -30 °C under a N2 atmosphère. The mixture was stirred at -30 “C for 4 h, and then quenched by addition of H2O (40 mL) and aqueous HCl (1 M, 50 mL) with stirring at 0 ’C. The mixture was separated, and the aqueous layer was extracted with EtOAc (2 x 50 mL). The combined organie layers were washed with brine (2 x 50 mL). dried, filtered and concentrated under vacuum to give the crude intermediate 22 (2.1 g), which was used directly in the next step without purification.
Ή NMR: (CDClj):δ4.97 (br, IH),3.10(s,2H),2.17(br, IH), i.44(s,9H). 1.20(s. 6H).
Step 2: Synthesis of intermediate 23.
NHBoc
To a mixture of intermediate 22 (3.0 g, 15.9 mmol) in anhydrous DCM (50 mL) was added DAST (23 mL, 17.4 mmol) at -78 ’C under a N2 atmosphère. The mixture was stirred at -78 ’C for l h, and then allowed to warm to rt ovemight The mixture was cooled to 0 ’C, andquenched by slow addition of sat aqueous layer NaHCOj (30 mL) with stirring at 0 ’C. The mixture was separated, and the aqueous layer was extracted with DCM (2x 20 mL). The combined organic layer was washed with brine (2 x 30 mL), dried, filtered and concentrated under vacuum to yield the crude intermediate 23 (23 g), which was used directly in the next step without purification.
1H NMR: (CDClj): 54.82 (br. IH), 3.30-335 (d, J =6.0 Hz. I H), 3.24-3.26 (d, J = 6.0 Hz, IH), 1.44 (s, 9H), 137 (s, 3H), 135 (s,3H).
*’F NMR: (CDClj): δ -144.93.
Step 3: Synthesis of Intermediate 24.
Το a mixture of intermediate 23 (2.0 g, 103 mmol, crude) in anhydrous DCM (10 mL) was added HCl in dioxane (4M, 10 mL,40 mmol) with stirring. The mixture was stirred atrt for 2 h after which time the solvent was removed under reduced pressure. The residue was treated with a mixture of DCM-petroleum ether (1: 1,3 x 10 mL) and the precipitate was collected and dried under vacuum to yield the crude intermediate24 (I.I g), which was used dîrectly in the next step without purification.
*H NMR: (CDjOD): δ 3.15-325 (d. J = 20.0 Hz, 2H), 131 (s, 3H), 148 (s, 3H). ”F NMR: (CDCIj): <5 -14739.
Intermediate 24 can altematively be obtained from dibenzyl amine according to the following procedure:
Step 1: Synthesis of Intermediate 22a
22a
To a slurry of L!Br( 1.66 g, 19.06 mmol, 0.2equiv) in MeOH (3.8 mL) was added BnzNH (18.80 g, 9530 mmol, I.Oequiv) at about20-25’C. Isobutylene oxide (10.31 g, 142.95 mmol, I3equiv) was added at a rate to maintain the température below 65 °C. After the addition was complété, the batch was stined at about 60 °C for 6 h. The batch was cooled to about 20*C, and toluene (37.6 mL) and water (18.8 mL) were added. After stirring for about 5 min, the layers were separated. The organic phase was concentrated under vacuum to an oil, and toluene was added and the solution was again distilled to an oil. Compound 22a was obtained as a toluene solution (33.88 g, 75.1 wt.%) in 99% yield and used dîrectly in the next step.
'H NMR: (CDCIj):<S 1.11 (s, 6H), 2.42 (s, lH),236(s, 2H), 3.70(s, 4H), 7.23-7.35 (m, 10H).
Step 2: Synthesis of Intermediate 23a
1. Deoxo-Ruor toluene, -20 *C
2. HCl, APrOH
22a
23a
A solution of intermediate 22a (10.32 g, 75.1 wt.%, 28.77 mmol) In anhydrous toluene (40 mL) was cooled to-20 °C. Deoxo-Fluor (7.0 g, 31.64 mmol, 1.10 eq) was added dropwise while keeping the température below -10°C. The mixture was stirred at -20 °C to -10°C for 3 h. The reaction was then quenched by the addition of aqueous KOH solution (6.46 g of 85 wt.% KOH pellets, 96.86 mmol, 3.40 eq In 25.84 g of water) while keeping the température below 10°C. The mixture was warmed to rt and the layers were separated. The organic layer was washed with water (3 x 25 mL). The organic phase was concentrated under vacuum and repeatedly distilled with heptane until the water content was < 200 ppm. The crude product was diluted with heptane (25 mL) and filtered through a silica gel pad (8 g silica gel). The silica gel pad was rinsed with heptane (2 x 20 mL) and the combined heptane filtrâtes were distilled under vacuum to the minimum volume and repeatedly distilled with isopropanol. Isopropanol (40 mL) was added and the solution was cooled to -10 C. Hydrogen chloride solution in isopropanol (83 mL, 5.2 N, 43.16 mmol, 130 eq) was added while keeping the température below 30 °C. After stirrïng at 20-25 °C for l h. the mixture was heated to 75 °C to get a clear solution and held at this température for 15 min. The mixture was cooled to 20-25 °C and stirred at this température for 2-3 h. The solid was filtered, washed with heptane, and dried under vacuum at
20-25 “C to give the product as a white solid (5.74 g, 91 wt.%) in 65% yield.
'H NMR: (CDClj): δ 131 -135 (d, J = 213 Hz, 6H), 335-338 (d. J = 18.8 Hz, 2H). 4394.45 (dd, J - 18.6 Hz, J = 33 Hz, 4H), 730-7.62 (m, Ι0Η).
’F NMR: (CDClj): δ -14338.
Interrnediate 23a can altematively be obtained from dibenzyl amine according to the following procedure:
1. NFSI
2. NaBH4
HCl, 2-PrOH
23a
To a réaction vessel equippcd with a Dean-Stark trap (prefilled with isobutyraldéhyde) was charged dibenzylamine (40.06 g, 203.06 mmol), isobutyraldéhyde (19.04 g, 264.98 mmol, 130 equiv) and toluene (20 mL). The mixture was heated to reflux under nitrogen to remove water (- 3.8 mL) in - 4 h, whiie the température was gradually raised to - 115 ’C). The excess of isobutyraldéhyde and toluene was then distilled under reduced pressure. The crude liquid was cooled to -10 ’C and a solution of N-fluorobenzenesulfonimide (NESI. 76.84 g. 243.67 mmol, 1.20 equiv) in N,N,-dimethyl acetamide (100 mL) was slowly added below 20 ’C. The mixture was stirred at room température until complété conversion (5-20 h). The mixture was cooled to 0 ’C and a solution of NaBHj (4.22 g, 111.68 mmol, 035 equiv) in N, N,-dimethyl acetamide (48 mL) was added below 20 ’C. After addition, the mixture was stirred at room température for 2.5 h. The mixture was cooled to 10 ’C and a solution of NaOH ( 10.56 g, 263.98, 1.50 equiv) in water (40 mL) was slowly added (gas was released), followed by 200 mL of water. The mixture was stirred at room température for 03 h, and extracted with heptane (250 mL). The organic layer was washed with water (2 x 150 mL) and distilled at normal pressure (up to 115 ’C). 2-Propanol ( 150 mL) was added and the mixture was distilled to remove solvents (50 mL). Acetic anhydride (2.07 g, 20.29 mmol, 0.10 equiv) was added at -30 ’C and stirred for 03 h. To the mixture was added 43 M HCl In 2-propanol (54 mL, 243.67 mol, 1.20 equiv) at -30 ’C. The resulting suspension was stirred at 60 ’C for 1 h, and then cooled to 20 ’C in 1 h. The solid was filtered, rinsed with 2-propanol (50 mL), and dried to give a white solid (23a) (47.46 g, 97.7 % purity) in 74 % yield.
Step 3: Synthesis of Intermediate 2-1
F 1.H2. Pd/C Zk/
Jx^NBnjHCI MeOH, 60 ’C _ HCPHjN
2. MeOH/MTBE
23a 24
To a hydrogénation vessel was charged 10% palladium on carbon (50% wet with water, 033 g. 0.25 25 mmol, 0.01 equiv), 23a (8.74 g, 893 wt.%, 25.41 mmol, 1.00 equiv) and methanol (24 mL). The mixture was hydrogenated at 60’C and 400 psi of H2 for 5-8 h. After cooling to 20-25 ’C, the mixture was filtered through a Celite pad, and the pad was rinsed with MeOH. The solvent was distilled under vacuum at 50°C to a volume of 4-5 mL. MTBE (25 mL) was added to the batch dropwise with stirring to form a slurry. After stirring for 30 min at 50°C, the batch was cooled to 20-25’C, held at this température for I h, and filtered. The solid was rinsed with MTBE and then dried at 25”C under vacuum for 4 h. Compound 24 was obtained as a white solid (3.24 g. 96 wt.%) tn 96% yield. ‘H
NMR: (CDjOD): δ 1.44-1.49 (d, J = 213 Hz, 6H), 3.13-3.18 (d. J = 19.7 Hz. 2H). ”F NMR: (CDClj): <5-14735.
Step 4: Synthesis of intermediate 25.
Το a stirred mixture of thïourea (23.0 g, 302 mmol) in THF (5.0 L) under argon at 0 °C was added NaH (29.9 g, 755 mmol, 60% in minerai oil). After 5 min, the ice bath was removed. and the reaction mixture was stirred at rt for 10 min. The mixture was cooled to 0 °C and B0C2O (138 g, 635 mmol) was added. The ice bath was removed aller 30 min of stirring at that température. The resultîng sïurry was stirred for another 2 h at rt. The reaction was quenched with an aqueous solution of sat. NaHCOj (500 mL) and poured into water (5.0 L) and extracted with EtOAc (3 x 2.0 L). The combined organic layer was dried over NajSO^ filtered, and concentrated under reduced pressure to afford intermediate 25 (80.0 g), which was used for the next step without further purification. LCMS (method 1): tR = 1.15 min, MS (ESD m/z 575.2 [2M+Na]*.
Step 5: Synthesis of Intermediate 26.
1) NaH. THF. 0C. 1h
XIIJTFAWIhS
Bec -----►
H H > ,ν^ΝΗ,Ηα μ Η Η Γ' '20
Το a mixture of intermediate 25 (3.9 g, 14.2 mmol) and anhydrous THF (285 mL) was added NaH (0.68 g, 17.0 mmol, 60% in minerai oil) at 0 °C and the mixture was stirred for 1 h, then TFAA (2.20 mL, 15.6 mmol) was added and the stirring continued for an additional I h. A pre-mixed mixture of intermediate 24 (2.0 g, 15.6 mmol) and EtjN (3.96 mL, 28.40 mmol) in anhydrous THF (130 mL) was added and the resultîng mixture was stirred at rt ovemight. Water (150 mL) was added to quench the reaction and the mixture was extracted with EtOAc (3 x 200 mL). The combined organic layer was dried, filtered and the solvent was removed under reduced pressure. The residue was purified by flash column chromatography on silica gel (eluent: petroleum ether: ethyl acetate from 50: I to 8: l)to afford intermediate 26 (2.49 g).
LC-MS (method l): tR = 1.08 min, MS (ESD m/z 194.8 [M-55]*.
'H NMR (CDjOD): δ 3.88-3.93 (m, 2H). 133 (s, 9H), 1.43 (s, 3H), 138 (s, 3H) ”F NMR (CDjOD): δ-144.15
Example 2
Step 1: Synthesis of Intermediate 27.
To a solution of intermediate 20 (550 mg. 1.61 mmol) in DMF (5 mL). intermediate 26 (425 mg. 1.69 mmol), EDC1 (614 mg, 3.22 mmol) and DIE A (416 mg. 3.22 mmol) were added. The mixture was stirred at rt for 36 h. EtOAc (200 mL) was added, followed by water (20 mL), and the mixture was stirred for 10 minutes. The organic layer was separated and washed with water (3 X 20 10 mL), brine (3 x 50 mL), dried, and solvent was removed under reduced pressure to afford crude product. The residue was purified by column chromatography (petrolcum ether: ethyl acetate; 5:1) to afford intermediate 27 (547 mg).
LC-MS: tR = 1.14; MS (ESI) m/z 5133 [M+Hf.
Step 2: Synthesis of Example 2.
To a mixture of intermediate 27 (400 mg, 0.56 mmol) in DCM (5 mL) was added TFA (1 mL) and the mixture was stirred m rt for 2 h. To this mixture, a sat. NaHCOj solution (10 mL) was added and stirred for 10 minutes. The mixture was partitioned between DCM (10 mL) and HjO (10 mL). The organic loyer was separated and washed with brine ( 10 mL), dried over Na3SO4 and concentrated under reduced pressure. The residue was purified by préparative HPLC (basic method
I) and SFC method A to give compound Example 2 (303.9 mg).
LC-MS (method I): tR = 0.90 min, MS (ESI) m/z 413.2[M+HJ*.
’H-NMR(CDjOD): 57.63-7.65 (dd, J = 8.0,1.6 Hz, IH),7.49-731 (d,./=8.0 Hz, IH),7.30(s, IH),
3.69-3.76 (m,2H), 326-330 (m, IH),3.15-3.19 (m, 1 H),2.55-239(1, J = 8.0Hz, IH), 1.79-1.84 (m,
IH), 1.27-1.63(m, IIH), 1.03-1.09(t, J = 12.0Hz, IH), 1.00-1.01 (t,J =4.0Hz,3H),0.96-0.97(d, J = 4.0 Hz, 3H). ”F NMR: (CDjOD): S -1393.
Example 3
Step It Synthesis of intermediate 28.
f-BuNH2, CHCIj
NCS, con. HCl
2-methylproponal (93 g. 129 mmol) was added to f-BuNHi (4.75 g, 129 mmol) at 0 °C and stirred at rt for 2 h. To this mixture CHCIj ( 130 mL) was added, and the mixture was dried over
NajSOt and filtered. To the résultant solution. NCS (18.20 g. 136 mmol) was added at 0 °C, followed by stirring at rt for 5 h. Water ( 100 mL) was poured into the reaction mixture and the mixture was extracted with CHCIj(3 x 100 mL). The combined organic layer was washed with water (200 mL), dried,and concentrated under reduced pressure. To the résultant residue, concentrated HO was added. The mixture was stirred at rt for 5 h. and sat. NaHCOj (200 mL) was added. The product was extracted wi th CHCIj and the residue was d isti I lcd at atmospheric pressu re to obtai n intermed iate 28 (2 g).
'H NMR (CDClj): 59.44(s. IH), 1.65 (s, 6H).
Step 2: Synthesis of intermediate 29.
The mixture of intermediate 28 (1.06 g, 10 mmol ) and titaniu m( IV) ethoxide (2.72 mL, 12 mmol) in anhydrous THF (22 mL), (±) N-tert-butylsulfinamide (1.21 g, 9 mmol) was added. The resulting mixture was stirred at reflux under N2 atmosphère for 4 h. The reaction mixture was cooied and water (20 mL) was added. The resulting mixture was filtered and the aqueous layer was extracted with ethyl acetate (3 x 20 mL). The combined organic phase was dried over NaïSCh and concentrated under reduced pressure to give intermediate 29 (I g).
Ή NMR (CDCIj): ί 7.94(s, IH), 1.71 (s, 6H). l.l4(s,9H).
Step 3: Synthesis of intermediate 30.
30
To a solution of intermediate 29 (0.7 g. 333 mmol) in anhydrous THF (5 mL) was added NaBH| (0.25 g. 6.66 mmol) at 0 °C. The reaction mixture was stirred at rt for 16 h, the reaction was quenched with sut. NH4CI solution (5 mL), aqueous KHCOj (20 mL), and EtOAc (20 mL). The aqueous layer was extracted with EtOAc (2 x 20 mL). The combined organic layer was dried, and concentrated under reduced pressure to yield crude intermediate 30 (260 mg), which was used for the next step without further purification.
Ή NMR(CDCh): S3.40-3.45 (m, IH), 3.11-3.17(m, IH), 155-157 (m, 6H), 1.23 (s,9H).
Step 4: Synthesis of intermediate 31.
To the mixture of intermediate 30 (450 mg, 2.12 mmol) in dry MeOH (3 mL) was added HQ in dioxane (4 M, 2 mL). The resulting mixture was stirred at rt for 2 h. The solvent was removed under reduced pressure to afford crude product 31, which was used for the next step without further purification.
Example 3 was synthesized tn a manner similar to Example 1, using intermediate 31 in step 10.
LC-MS (method 1): tR = 0.86 min, MS (ESI) m/z 429.2 [M+H]*.
1H NMR: (CDjOD): 6 7.60-7.62 (d, J = 7.6 Hz, 1 H). 7.45-7.47 (d, J = 7.6 Hz, 1 H), 7.27 (s, I H), 3.763.85 (m, 2H), 3.11-3.25 (m, 2H), 2.51-2.56(m, IH), 157-158 (d, J = 4.0 Hz, IH), 1.42-152 (m, 9H).
1.23-1.26 (m, IH), 1.03-1.09 (m, IH), 0.97-0.99 (m, 3H), 0.93-0.94(m. 3H)ppm.
A mixture of dihydro-2H-pyran-4(3H)-one (50.0 g, 500 mmol) and 2-chloroacetonitrile (35.0 g, 350 mmol) in tert-butano! (50 mL) was stirred for 30 min. To this mixture was added a solution of r-BuOK (60 g, 550 mmol) in tert-butanol (500 mL) over 40 minutes. The reaction mixture was stirred at rt for 16 h. It was diluted with water (100 mL) and quenched with HCl (10% aqueous, 20 mL). The reaction mixture was concentrated to one-third of its original volume, and extracted with Et2O (3 x 200 mL). The combined organic layer was washed with brine, dried, and concentrated to afford crude intermediate 32 (57 g), which was used directly in the next step without purification.
Step 2: Synthesïs of intermediate 33.
CN
HF/Pyridlne J
33
Το a mixture of intermediate 32 (57 g) in DCM (200 mL) in a polypropylene bottle at 0 ”C, 70% hydrogen fluoride-pyridîne (50 mL) was added slowly. The mixture was allowed to warm to rt 5 ovemight. The reaction mixture was diluted with EtOAc (500 mL) and poured into sat. aqueous NaHCOj (200 mL). Additional solid NaHCOj was used to neutraüze the mixture carefiilly until bubbling ceased. The aqueous layer was extracted with EtOAc (3 x 500 mL). The combined organic layer was washed with aqueous HCl (1%, 200 mL) brine, dried and concentrated to give crude întennediate 33 (54 g), which was used directly in the next step without purification.
'H NMR: (CDClj): δ 437 (m, 2H), 3.96-2.70 (m, 4H), 1.97 - 1.81 (m, 4H).
Step 3: Synthesïs of intermediate 34.
CN
NaBH, »
2-Propanol 1
To a mixture of intermediate 33 (54 g; 340 mmol) in 2-propanol (1000 mL) and water (250 mL) at 0 °C, NaBFL (20 g, 509 mmol) was added. The mixture was stirred and allowed to warm to rt over 3 h. The reaction was quenched with acetone (50 mL), and stirred for another l h. The clear liquid was separated from solid by décantation. EtOAc (100 mL) was used to wash the solid, and the 20 filtrâtes were combined. The combined organic solution was concentrated under reduced pressure and purified with flash column chromatography on silica gel (5-20% ethyl acetate in hexanes) to give intermediate 34 (22 g).
'H NMR: (CDClj): 8:3.82-3.77 (m, 4H). 3.72-332(dd, J = 20.8,6.4 Hz, 2H), 2.69(s, IH), 1.82-1.60 (m,4H).
Step 4: Synthesis of intermediate 35.
MsCl (25.8 g. 225 mmol) was added to a mixture of intermediate 34 (20 g, 150 mmol) and TEA (22.7 g, 225 mmol) in DCM (200 mL) at 0 °C. The mixture was stirred at rt for 2 h, and then water( 100 mL) was added. The aqueous layer was extracted with DCM (2 x 200 mL), organic phases were combined, dried and solvent was removed under reduced pressure to afford crude intermediate 35 (30 g), which was used for the next step without further purification.
Ή NMR: (CDCIj): 8:4.22 (d, J = 20.0Hz, 2H), 3.87-3.82 (m,4H), 3.06(s, 3H), 1.88-1.68 (m, 4H).
Step 5: Synthesis of intermediate 36.
Το a mixture of intermediate 35 (10 g, 47 mmol) in DMF(150mL) was added NaNj (16 g, 250 mmol) and NaHCOj (9.3 g, 100 mmol) at rt. The mixture was stirred at 120 ’C for 20 h. The reaction quenched with water at rt, extracted with EtOAc (2 x 200 mL). The organic phases were combined, dried and solvent was removed under vacuum to afford crude intermediate 36 (8 g), which was used for the next step without further purification.
Step 6: Synthesis of intermediate 37.
Το α mixture of intermediate 36 (8 g, 50 mmol) In EtOAc (100 mL) was added 10% Pd/C ( 0.8 g) under a N2 atmosphère, the mixture was degassed and ex ch ange d with hydrogen for 3 times. The final mixture was stirred at rt under I atm. hydrogen atmosphère for 24 h. The catalyst was filtered off through a pad ofcelite and washed with EtOAc (2 x 50 mL). The combined filtrate was concentrated under reduced pressure to yield intermediate 37 (53 g).
Ή NMR: (CDjOD): δ 3.83-3.79 (m, 4H), 2.76-2.71 (d. J = 8.0 Hz, 2H), 1.83-1.65 (m, 4H).
”F NMR: (CDjOD) δ:-169.66.
Example 4 was synthesized In a manner simiiar to Example 1, using intermediate 37 in step 10.
LC-MS (method 1 ): tR « 0.80 min, MS (ESI) m/z 455.2 [M+H]*.
Ή-NMR: (CDjOD): δ7.61-7.63 (d, J = 7.6 Hz, IH), 7.47-7.49 (d, J=8.0 Hz, IH), 730 (s. IH), 3.633.83 (m, 6H), 3.23-3.27 (m, IH), 3.12-3.16 (m, IH), 232-237 (1,7 = |0.0 Hz, 1H), 1.48-1.82 (m, 7H). 138-1.44(1,7 = 12.0Hz. IH), 1.23-1.28 (m, IH), 0.97-1.05 (m, 4H), 0.94-0.95 (d, 7 = 4.0 Hz, 3H).
WF NMR: (CDjOD): δ -160.48
To a solution of intermediate 17 (3.6 g, 7.4 mmol) in CHCI3 (25 ml) was added TMSI ( 10 ml) and the mixture was stirred at 65 ”C for 2 h. The mixture was cooled to rt, sat. Na2S2O3(10 mL), and sat. NaHCOj aqueous solution (10 mL) were added, and the mixture was stirred for 10 minutes. The mixture was partîtioned between DCM (50 mL) and H2O ( 10 mL). The organic layer was separated
tutd washed with brine (10 mL), dried over NajSOj, filtered and concentrated under reduced pressure to afford crude intermediate 38 (Z6 g), which was used for the next step wïthout further purification.
Step 2; Synthesis of Intermediate 39.
Toamixtureof intermediate38(2.5g.6.4 mmol) in DMF(20mL) wasadded intermediate ( 1.8 g, 7.0 mmol ,1.1 eq), EDCI (2 J g, 13 mmol) and DŒA ( 1.7 g, 13 mmol). The mixture was stirred ovemight The reaction mixture was diluted with water and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried, the solvent was removed under reduced pressure to afford crude product, which was purified by column (petroleum ether: ethyl acetate = 20: 1 to 5: l) to give intermediate 39 (2.8 g).
Step 3: Synthesis of intermediate 40.
To a solution of intermediate 39 (350 mg, 0.6 mmo!) in dioxane (5 mL) under N2 atmosphère, 5-pyrimidine boronic acid (90 mg, 0.66 mmol) and aqueous Cs2COj solution (2 mL, 2 M in water) were added. The mixture was purged by bubbling a steam of N2 for 5 min, then Pd(dppf)CI2 (40 mg, 0.06 mmol) was added. The mixture was stirred for 2 h ut 110 °C under a N2 atmosphère. The reaction was cooled to rt, diluted with EtOAc and filtered. The filtrate was washed with aqueous Na2COj (5 mL) und concentrated under reduced pressure to afford crude intermediate 40 (300 mg) which was used for the next step without further purification.
To a solution of intermediate 40 (300 mg, 0.53 mmol) in DCM (5 mL) was added TFA ( I mL) and the mixture was stirred at rt for 2 h, To this mixture, sat. NaHCOj solution ( 10 mL) was added and stirred for 10 minutes. The residue was partitioned between DCM (10 mL) and HjO (10 mL). The organic layer was separated and washed with brine ( 10 mL), dried over NajSCh, filtered and concentrated under reduced pressure. The residue was purified by préparative HPLC (basic, method 1) to give Example 5(141 mg). LC-MS (method 1): MS (ESI) m/z 466.2[M+Hf. *H NMR: (CD5OD): S 9.12 (s, 1 H), 9.02 (s, 2H), 7.64-7.62 (dd, J = 7.6,1.6 Hz, 1 H). 731 (d, J = 8.0 Hz, I H).
7.29 (s, IH), 3.80-3.66 (m. 2H). 3.30-3.13 (m,2H), 2.59 (t, J = 10.0Hz, IH), 1.85 (d, 12.4 Hz.
IH). 1.65-1.32 (m, 10H), 1.11-1.05 (m, IH), 1.02(d, J = 6.4 Hz,3H), l.02(d, J=6.4 Hz, 3H). ”F NMR: (CDjOD): <5-139.27.
Example 6
To a mixture of intermediate 39 ( 1.0 g, 1.8 mmol) in éthanol (23 mL) and water (10 mL), NaNj (300 mg, 3.6 mmol), Cul (40 mg, 10%) sodium ascorbate (40 mg, 0.20 mmol, 5%) and N,N’· 20 dimethyl-cyclohexane-1,2-diamine (40 mg, 0.28 mmol. 15%) were added under Nj atmosphère. The mixture was stirred for 3h at 90 °C undera N: atmosphère. The mixture was cooled to rt, diluted with
EtOAc and filtered. The filtrate was concentrated ta afford crude Intermediate 41 (S30 mg) which was used for the next step without ftirther purification.
Step 2: Synthesis of intermediate 42.
42
To a mixture of intermediate 41 (830 mg, 1.6 mmol) tn MeOH (10 mL) was added 10% Pd/C (0.1 g) under n nitrogen atmosphère, the mixture was degassed and exchanged with hydrogen for 3 times. The mixture was stirred at rt under 1 atm hydrogen atmosphère for 4 h. The mixture was filtered through a pad of celitc and washed with EtOAc (2 x 10 mL). The combined filtrate and washing were concentrated under reduced pressure to give Intermediate 42 (0.7 g), which was used for the next step without further purification. LC-MS (method I): t« = 0.99 min, MS (ESI) m/z 503.2 [M+H]*.
To a mixture of intermediate 42 (350 mg, 0.7 mmol) in DCM (5 mL) was added EtjN (0.2 mL, 13 mmol, 2.0 eq) and 2-fluoro-2-methylpropanoyl chloride (150 mg. 2 mmol). The mixture was stinred at rt for 3 h. The reaction was quenched with water and extracted with DCM (2x10 mL).
The combined organic layers were washed with brine (3 x 15 mL), dried over NaîSC^ and the solvent was removed under reduced pressure to afford crude intermediate 43 (320 mg) which was used for the 20 next step without further purification.
Step 4: Synthesis of Example 6.
To a mixture of intermediate 43 (320 mg, 0.54 mmol) in DCM (5 mL) TFA (I mL) was added and the mixture was stirred at rt for 2 h. The réaction was quenchcd with sat. NaHCOj aqueous solution (10 mL). The mixture was partitioned between DCM (10 mL) and H2O (10 mL). The organic layer was separated and washed with brine (10 mL), dried overNajSO4, filtered and concentrated under reduced pressure. The residue was purified by preparative HPLC (basic method I) to give Example 6 (179.9 mg).
LC-MS (method 1): LC-MS tH » 0.86 min, MS (ESI) m/z 49 U(M+H]*.
'H NMR: (CDjOD): J7.42 (m IH), 7.30 (m, 2H), 3.78-3.71 (m, 2H), 3.17-3.05 (m, 2H), 2.59 (t, 10.0 Hz, IH), 1.80(d. J = 12.0 Hz, IH), 1.64-133 (m, 8H), 1.42-134(m. 8H), 1.08-0.96 (m,7H). ’FNMR: (CDjOD): <5-138.95,-147.61.
Example 7
To a solution of 6-bromo-indan-l-one (100.0 g. 0.48 mol) dissolved in THF (2.0 L, 0.24 M) was added r-BuOK (64.0 g. 037 mol) in one portion at 0 °C. The reaction was stirred for 5 min at 0 °C and stirred for on additional 10 min at rt Methyl méthacrylate (56.0 mL, 0.53 mol) was added in one portion. After 30 min. methyl acrylate (52.0 mL, 0.57 mol) was added in one portion and the mixture stirred ovemight. To this réaction mixture, DMF (260 mL, 1.8 M) and EtI (76.0 mL, 0.96 mal) were added and the mixture was stirred ovemight. The reaction was quenched with sat aqueous citric acid solution (200 mL) and the organic layer was separated. The EtOAc was removed under reduced pressure and the crude material was diluted with H^O (3 L) and extracted with EtOAc (3 x 3L). The combined organic layers were washed with brine, dried over Na^SO^ filtered, and the solvent removed under reduced pressure to afford crude intermediate 44 (200 g), which was used for the next step without further purification.
Step 2: Synthesis of intermediate 45.
Br.
Intermediate 44 (200.0 g, 0.51 mol) was mixed with DMSO ( 1.0 L, 03 M) and LiCI (215.0 g, 5.1 moi) was added. The mixture was heated to 120 °C for 4 days. The mixture was concentrated in vacuo. The residue was dissolved in EtOAc. filtered, and the filtrate was concentrated. The residue wns purified by column chromatographyon silica gel (petroleumether: EtOAc=3Ch I) to afiord the crude intermediate. The intermediate was dissolved in minimal amount of MeOH, and NaOMe In MeOH (30%, 20 mL) was added, After 20 min. the mixture was filtered to give intermediate 45 (40 g)· 'H-NMR;(CDCi3):i7.93(cW« 1.6Hz, !H),7.77(dd.J=> 10.8,2.4Hz. IH),7.42-7.45(d, J» 10.8 Hz. IH).334(s,2H),2.60-2.70(m, IH),236-247(m, IH). 1.76-1.99 (m.5H), 121-|30(m, IH), 1.07 (d, J « 8.8 Hz. 3H), 0.90 (t, J = 9.6 Hz, 3H) ppm.
Step 3: Synthesis of intermediate 46.
Br<
OH
4S
Intermediate 46 was synthesized from intermediate 45 by a method similar to that described in step 2 of Example 1.
‘H-NMR: (CDCb): δ 7.81 (d. J =< 1.8 Hz, IH). 7.63 (dd, J = 8.1,2.1 Hz, IH), 7.42-7.45 (d, 8.4 Hz,
IH), 334 (s, 2H), 2.60-2.70 (m, IH), 2.36-2.47 (m, 1H), 1.76-1.99 (m, 5H). 1.21-130 (m, IH), 1.07 (d, J = 7.2 Hz, 3H), 0.90 (t, J = 75 Hz, 3H) ppm.
Step 4: Synthesis of intermediate 47.
To a solution of intermediate 46 (30.0 g, 0.08 mol) in DMF (500 mL) was added NaH (8.0 g, 0.16 mol) ai 0 °C. The mixture was stirred at 0 °C 30 min, and then Met (25.0 mL, 0.4 mol) was added and the mixture was stirred at rt ovemight. The mixture was quenched with H2O (100 mL), extracted with EtOAc (3 x 300 mL). The organic layer was concentrated and purified by column chromatography (petroleum ether/EtOAc = 20/ 1) to afford intermediate 47 (22.0 g).
1 H-NMR: (CDCb): <57.81 (d,J = 1.8 Hz, 1 H), 7.63 (m, J 8.1.1.8 Hz, IH), 7.27 (d, J« 8.1 Hz, IH), 3.41 (s, 3H), 2.91 (s, 2H), 2.46-252 (m, IH), 1.75-1.79 (m, IH), 159-1.62 (m, IH), 1.29-1.47(m, 5H), 1.08-1.15 (m, l H), 0.98 (d, J =63 Hz, 3H), 0.78 (t, J = 75 Hz,3H).
Step 5: Synthesis of intermediate 48.
A mixture of intermediate 47 (22.0 g, 0.06 mol) and titanium (IV) ethoxide ( 130 mL, 0,62 mol) in dry THF (400 mL) was stirred at rt for I h. (S)-A/-tert-butylsulfinamide (30.0 g, 0.25 mol) was added and the resulting mixture was stirred at 80 °C under Nj ovemight. The reaction mixture was cooled and water (200 ml) was added. The mixture was filtered and the aqueous layer was extracted with EtOAc (3 x 400 mL). The combined organic layer was dried over NajSOt and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether EtOAc = 20: I) to give intermediate 48A (7.0 g) and 48 (10.0 g) respectively.
Step 6: Synthesis of intermediate 49.
Ton mixture ofethoxy-ethcne (4.7 mL,493 mmol) in anhydrousTHF(50 mL)at-78 ’C was added r-BuLi (38.0 mL, 493 mmol, 1.3 M in hexane) dropwise over 20 minutes and the mixture was stirred for 20 min. The resulting mixture was stirred at 0 ’C foranother45 min and then cooled back to -78 ’C.
A pre-cooled solution of intermediate 48 (43 g, 9.9 mmol) in anhydrous THF (60 mL) at -78 ’C was added to the above solution, dropwise over 30 minutes and the mixture was stirred for 23 h at -78 ’C. The reaction was quenched with sat NHiCI (50 mL) and extracted with EtOAc (3 x 100 mL). The combined organic phase was concentrated under reduced pressure to give the residue, which was purified by column chromatography (petroleum ether EtOAc = 20; I) to afford intermediate 49 (33 g)·
LC-MS (method 1): tR = 5.94 min, MS (ESI) m/z 528.1 [M+H]*.
Step 7: Synthesis of intermediate S0.
Intermediate 49 (33 g, 6.60 mmol) was dissolved in a mixture of DCM and MeOH (5: I; 40 mL), and cooled to -78 ’C. Ozone was bubbled through the mixture for 20 min. The réaction was stirred for an additional 10 minutes, after which the mixture was purged with N2 and treated with Me2S at -78 ’C. The reaction was allowed to warm to rt and stirred for an additional 3 h. The solvent was removed under reduced pressure and the residue was purified by column chromatography on silica gel (petroleum/ EtOAc « 15/1) to give intermediate 50 (2.3 g).
’H-NMR: (CDCIj): δ 7.83 (s, IH), 7.43 (dd, J = 8.0, 2.0 Hz, 1 H). 7.10 (d, J = 8.0 Hz, 2H), 4.26-4.43 (m, 3H), 3.42 (s, 3H), 2.95 (d, J = 16.0 Hz, IH), 2.68 (d, J - 16.0 Hz, IH), 232-237 (m, 1 H), 2.17 (s, 5 IH), 1.91-1.97 (m, IH), 1.82-1.89 (m, IH), 1.63-1.68 (m, IH), 131-1.40(m,6H), 1.13 (s, 9H), 0.900.95 (m, 6H), 0.68 (t. 12.0 Hz, IH).
Step 8: Synthesis of intermediate 51.
Zn(CN)a,Pd(OAc)2.X-phos
Zn. cal H2SO4, DMA
Concentrated sulfuric acid (49 pL), was added to DMA (20 mL) and the solvent was purged with Nj for 20 min. A 50 mL round bottom flask was charged with Pd (OAc) 2 (135 g) and Xphos (3.15 g) under N2, and transferred to the above solution. The resulting mixture was heated at 80 °C for 30 mîn to give mixture A.
In an another flask, DMA (30 mL) was purged under N2 for 20 min and intermediate 50 (23 15 g, 430 mmol), Zn (CN)j (527 mg, 4.50 mmol) and Zn dust ( 15 mg) were added followed by mixture A. The resulting mixture was heated al 90 °C for 40 min. The reaction mixture was cooled to rt, diluted with water (80 mL) and EtOAc (100 mL). After stirring for 10 minutes, the mixture was filtered through celite, and the organic layer was separated. The aqueous layer was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with water, brine, dried over
NajSOj, and the solvent was removed under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum.* EtOAc» 10:1) to afford intermediate 51 (1.8 g).
LC-MS (method 1): tR« 1.19 min, MS (ESI) m/z 475.2 [M+Hf.
Step 9; Synthesis of intermediate 52.
To a mixture of intermediate 51 (590 mg, 1.24 mmol) In MeOH ( 10 mL) was added a 4 M HCI solution in dioxane (2 mL). The resulting mixture was stirred for 30 min. Solvent was removed under reduced pressure to afford crude intermediate 52 (550 mg), which was used for the next step without further purification.
LC-MS (method 1 ): tR = 0.88 min, MS (ESI) m/z 322.1 [M-48]*.
Step 10: Synthesis of intermediate 53.
To a mixture of intermediate 52 (550 mg, 1.59 mmol) in CHClj (10 mL) was added TMSI (2.5 mL, 15.9 mmol) slowly et rt. The mixture was stirred at 60 °C for 2 h and then allowed to cool to rt. MeOH (5 mL) and sat. Na2S;Oj(5 mL) solution was added over 10 minutes. The layers were separated and the aqueous layer was extracted with CH2CI1 (3 x 40 mL). The organic layers were combined, washed with water (2 x 40 mL), dried and solvent was removed under reduced pressure to yieid crude intermediate 53 (400 mg).
Step 11: Synthesis of intermediate 54.
To a mixture of intermediate 53 (1.2 g, 3.30 mmol) in DMF (15 mL), intermediate 26(850 mg, 3.30 mmol), EDCI (1.28 g. 6.60 mmol) and DIEA (1.2 mL, 6.60 mmol) were added. The mixture was stirred at 30 °C ovemight The solution was cooled to rt and EtOAc (20 mL) and water (20 mL) were added. The organic layer was separated and the aqueous loyer was extracted with EtOAc (3 x 60 mL). The combined organic layers were washed with brine (3 x 50 mL), dried and the solvent was removed under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ EtOAc « 5/1 ) to afford 54 (760 mg).
Το α mixture of intermediate 54 (750 mg, 1.43 mmol) in DCM (8 mL) was added TFA (2 mL) and stirred at rt for t h. The pH of the reaction mixture was adjusted to 85 by addition of sat.
NaHCOj solution. The organic layer was separated and concentrated under reduced pressure to yield crude product. The residue was purified by préparative HPLC (basic, method 2) to give Example 7 (465 mg).
LC-MS (method I): tR = 0.85 min. MS (ESI) m/z 427.2 (M+H]*.
'H-NMR: (CD3OD): 7.65 (dcW« 8.0,1.6 Hz, IH), 7.49 (d, J = 8.0 Hz, ! H), 730 (s. IH), 3.65-3.80 10 (m. 2H), 3.12-3.29 (m, 2H), 2.64-2.69 (m. I H). 1.79-1.84 (m, 2H), 151-135 (m, I H), 1.32-130 (m,
9H), 132-1.42 (m, IH). 1.00-1.20(m,4H), 0.78 (t. J = 7.6 Hz, 3H).
'’F-NMR: (CDjOD): -139.444.
cj^^oh bf3ei2o
S5
NaOH, HjO r,t,2h
SSB
NaOH.HjO
S0*C,2h
ΌΗ se
A mixture of (R)-2-(ch!oromethy!)oxirane (55,109 mL, 1.62 mol) and BFj.OEtj (3.4 mL,
0.027 mol) in toluene (200 mL) was heated to an internai température of 30 °C and 2-cho!oroethanol (49 g, 0.53 mol) was added dropwise at a rate sufficient to main the reaction température at 36-38 °C. The resulting mixture was aged at 36 °C for 20 min. The mixture was cooled to 16 °C and aqueous
NaOH (250 mL, 23%) was added with vigorous stirring over Ih, maintaîning the reaction température below 20 “C. The mixture was aged for 1 h at rt. The two layers were separated, and the aqueous phase was extracted with toluene (130 mL). The combined organic layers were washed with water ( 100 mL), the resulting organic layer was distilled to low volume, monitoring the distillate for loss of product to give the final intermediate 56 (23 g).
’H-NMR: (CDCI3): δ 3.33-3.84 (m, 9H).
Step 2: Synthesis of intermediate 57,
To a mixture of intermediate 56 (81.0 g, 0.686 mol) in DCM (500 mL) was added TEA (196 mL, 1.37 mol) and MsCI (80 mL. 1.029 mol) at 0 °C. The mixture was stirred for 5 h at rt, and quenched with water (200 mL), extracted with dichioromethane (2 x 200 mL). The combined organic layer was washed with brine, dried overNa2SO4 and concentrated to afford crude intermediate 57 ( 132.8 g), which was used for the next step directly without purification.
Step 3: Synthesis of intermediate 58.
To a mixture of intermediate 57 (132.8 g, 0.677 mol) in DMF (640 mL), NaNj (88.0 g, 135 mol), NaHCOj ( 170.6 g, 2.03 mol) and Nal (20.3 g, 0.135 mol) were added. The mixture was stirred at rt ovemight. The réaction mixture was quenched with water (300 mL), and then extracted with ethyl acetate (2 x 300 mL). The combined organic layer was washed with water and then brine, dried over NajSOj and concentrated under reduced pressure to afford crude intermediate 58 (96 g), which was used for the next step directly without purification.
Step 4: Synthesis of intermediate 59.
Pd/C, Hj
MeOH
59
Το a mixture of intermediate 58 (3.6 g, 25.48 mmol) in MeOH (100 mL) was added Pd/C (0.4 g. 10% content) under a nitrogen atmosphère, the mixture was degassed and exchanged with hydrogen for 3 times. The final mixture was stirred at rt under hydrogen balloon for 24 h. The catalyst was filtered off through a pad of celite and washed with MeOH (2 x 50 mL). The combined filtrate and washing were concentrated under reduced pressure to give crude intermediate 59 (2.93 g).
Step 5: Synthesis of intermediate 60.
NH; (Boe);Q _ EtjN.THF
To a solution of intermediate 59 (1.1 g. 10 mmol) in THF (50 mL) was added EtjN (3.0 g, 30 mmol) and (Boc)jO (2.6 g. 12 mmol). The mixture was stirred at rt ovemight The réaction was quenched with water (20 mL) and extracted with EtOAc (2 x 30 mL). The combined organic layer was washed with brine (20 mL). dried over Na^SOj. filtered and concentrated under vacuum to give crude product, which was purified by column chromatography on silica gel (petroleum ethen ethyl acetate; 100:1 to 20: I) to afford pure intermediate 60 (500 mg).
Step 6: Synthesis of intermediate 61.
HCI-dtoxane MeOH
NHjHCI
Intermediate 60 (20 g. 92 mmol) was dissolved in MeOH ( 150 mL), and then a solution of HCl in dioxane (4 M, 30 mL, 120 mmol,) was added. The reaction mixture was stirred at rt for 18 h. MeOH was removed under vacuum to yield pure intermediate 61 (14 g), which was used for the next step without further purification.
‘H NMR (CDjOD): â 3.62-3.90 (m, 6H), 3.32-3.35 (m. IH), 3.01-3.04 (m, IH), 2.85-2.90 (m, IH).
Step 7; Synthesis of intermediate 62,
Το a mixture of Intermediate 25 (12 J g, 4435 mmol) in anhydrous THF (600 mL) was added NaH (2.1 g, 53.46 mmol, 60% in minerai oil) atO “C. The reaction mixture was stirred for 1 h, followed by addition of TFAA (6.9 mL, 49.0 mmol) and the stirring was continucd for additional 1 h. Intermediate 61 (73 g, 49.0 mmol) und EtjN (12.4 mL, 89.1 mmoi) In anhydrous THF (300 mL) were added and the resulting reaction mixture was stirred at rt o verni g ht. H2O (300 mL) was added to quench the reaction and the mixture was extracted with EtOAc (3 x 350 mL). The combined organic layers were dried, filtered and the solvent was removed under reduced pressure. The residue was purified by coiumn chromatography on silica gei (5-50% ethyl acetate in hexane) to afford intermédiare 62 (7.95 g).
LCMS (method 1): tR « 0.90 min. MS (ESI) m/z 221.1 [M-55]*.
'H NMR (CDjOD): <5 3.80-3.90 (m,4H), 3.70-3.80(m, 2H), 335-3.65 (m 2H), 3.35-3.40 (m, IH). 137 (s, 9H).
Example 8 was synthesized from intermediate 53 and intermediate 62 in accordance with the method described in steps 11 and 12 for Example 7.
LC-MS (method I): tu = 0.87 min, MS (ESI) m/z 453.2[M+HJ*.
’H-NMR (CDjOD): 7.64 (dd, J = 8.0, 1.2 Hz. IH). 7.49 (d, 7.6 Hz, I H), 730 (s, IH), 331-3.86 (m, 8H),3.11-335 (m, 3H), 2.64-2.69 (m. IH), 1.78-1.84 (m, 2H), 1.49-135 (m, IH), 139 (s, 3H). 1.13-1.20 (m, 1 H), 0.99-1.06 (m, 4H), 0.78 (t. J = 7.6 Hz, 3H).
Example 9
Step 1: Synthesis of Intermediate 62,
P IBuOKTHF methyl acryiate Br
DMF, CHjCHjl
Lia, DMSO
140*0
A mixture of 6-bromo-indan-l-one 1 (50.0 g, 236 mmol) and methyl acryiate (42.0 g, 472 mmol) in anhydrous THF (900 mL) was pre-cooled to 0 °C and t-BuOK (31.8 g, 284 mmol, 1.1 eq) was added portion wise over 30 min. The mixture was warmed to rt over I h and stirred for an additional 40 mjn at rt. DMF (200mL) and EtI (74 g, 472 mmol) were added to this reaction mixture, and the mixture was stîned at rt ovemight. THF was removed under reduced pressure. The residue was diluted with HjO (300 mL) and extracted with EtOAc (300 mL). The organic layer was concentrated under reduced pressure to afford the crude intermediate 63 (120.0 g). This product was used as is for the next step.
A mixture of intermediate 63 (120.0 g, 310 mmol) and LICI (130.0 g, 3100 mmol) in DMSO (900 mL) was refluxed ovemight. The mixture was quenched with water (3 L) and extracted with EtOAc (3 x 400 mL). The combined organic phase was dried and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether: EtOAc; 20:1 ) to give intermediate 64 ( 15 g).
Ή NMR: (CDClj): δ 7.91 (s, IH), 7.74 (dd, J= 8.0 Hz, IH), 7.41 (d, J = 8.0 Hz, IH), 3.80 (s, 2H), 248-233 (m,2H), 233-249 (m, IH), 2.15-233 (m, IH), 1.75-1.95 (m,4H), 1.21-1.40 (m, 1H).O.88 (t, ./ = 8.0 Hz, 3H).
Step 2: Synthesis of intermediate 66.
O O
p \ NaBH,. _ -, NaH.OMF .
XXX- /3° MeOH -/ Mel, \—f
64 \__ 65 \— ββ
To a mixture of THF (20 mL) and MeOH (5 mL) at -78 eC was added intermediate 64 (6.0 g, 18.7 mmol), NaBHj (355 mg, 93 mmol) and CeCljjHjO (70 mg, 0.19 mmol). The mixture was stirred at -78 °C for 20 min, quenched with satd. NH4CI solution (30 mL), and extracted with EtOAc (400 mL X 4). The organic layers were combined and concentrated under reduced pressure to afford a crude intermediate 65 (63 g).
To a mixture of intermediate 65 (6.5 g, 20.0 mmol) and NaH (3.2 g, 80.0 mmol) in DMF (100 mL) was added Mel (11.4 g, 80.0 mmol) at 0 °C. The mixture was stirred at rt ovemight.The mixture was quenched with HjO, extracted with EtOAc, and concentrated under reduced pressure to afford the crude product, which was purified by column chromatography on silica gel (eluent: petroleum ether ethyl acetate; 20: 1 to 15:1) to afford intermediate 66 (3.5 g).
LC-MS (method 1): tR = 1.32 min, MS (ESI) m/z 339.1 [M+H]*.
Ή NMR: (CDCIj): 5 7.88 (s, IH). 7.69 (dd, J= 8.4,2.0 Hz. 1 H), 7.31 (d. J = 8.4 Hz, IH). 3.39 (s, 3H), 2.97 (s, 2H), 2.88-2.94 (m. IH). 2.21-2.26 (m, 1H), 1.81-1.87 (m. IH), 1.70-1.78 (m. IH), 1.40139 (m, 4H). 1.12-139 (m. 2H). 0.88 (t, J = 8.0 Hz. 3H).
Step 3: Synthesis of intermediate 67 & 67A.
The mixture of intermediate 66 (33 g, 10.4 mmol) and titanium (IV) ethoxïde (23.7 g, 104 mmol) în dry THF (40 mL) was stirred ut rt for 1 hour. (S)-N-tert-butylsulfinamide (1.6 g, 11.6 mmol) was added and the resulting mixture was stirred at 80 °C under Ni atmosphère ovemight. The reaction mixture was cooled and water (400 mL) was added and filtered. The aqueous layer was extracted with EtOAc (3 x 200 mL). The separated organic phases were combined and dried and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether: EtOAc; 20:1) and compounds eluted in the following order to give Intermediate 67 A (13 g) and 67 (13 g) respectively.
Step 4: Synthesis of intermediate 68.
To a mixture of ethoxy-ethene (13 g, 17.0 mmol) in anhydrous THF (20 mL) al -78 *C under a Ni atmosphère, f-BuLÎ (13.0 mL, 17.0 mmol, 13 M in hexane) was added dropwise and stirred for 20 min. The resulting mixture was stirred at 0 °C for an additional 45 min and then cooled back to 17202 °C. To this mixture, a pre-cooled solution of intermediate 67 (1.5 g, 3.4 mmol) in anhydrous THF (20 mL) at -78 °C was added dropwise and stirred for 23 h. The reaction was quenched with sat. NH4CI (50 mL) and then extracted with EtOAc (3 x 100 mL). The organic phases were combined and concentrated under reduced pressure to afford the crude product, which was purified by column on silica gel (petroleum ether; ethyl acetate; 20:1 ) to afford Intermediate 68 ( 1.2 g).
Step S: Synthesis of intermediate 69.
The intermediate 68 ( 13 g, 2.4 mmol) was added to a mixture of methanol in DCM (5:1,20 mL), and cooled to -78 °C. Ozone was bubbled through the mixture for 20 min. The mixture was purged with N2 and treated with Me:S (5 mL) at -78 “C. The réaction was allowed to warm to rt and stirred for an additional 3 h. The solvent was removed under vacuum, the residue was purified by préparative TLC (petroleum ether. ethyl acetate; 3*. 1) to afford intermediate 69 (860 mg).
LC-MS (method l): tR = 135 min, MS (ESI) m/z 516.1 [M+H]*.
Step 6: Synthesis of intermediate 70.
To intermediate 69 (860 mg, 1.7 mmol) in MeOH ( 10 mL) was added HCl in dioxane (4 M, 2 mL). The resulting mixture was stirred for 30 min at rt The solvent was removed under reduced pressure to afford crude intermediate 70 (800 mg) which was used for the next step without further purification.
Example 9 was synthesized from Intermediate 70 by the method described in Exampie 7, from step 10 through step 12.
LC-MS (method I ): tR = 0.79 min, MS (ESI) m/z 413.2 [M+H]*.
Ή-NMR(CDjOD):7.66(dd, J = 7.6,1.6Hz, IH),731 (d,7=8.0Hz, IH),7.32(s, IH),3.69-3.76 (m, 2 H), 3.12-3.27 (m,3H), 1.78-1.95 (m,3H), 1.32-1.42 (m, IIH), 1.11-1.18 (m, IH),O.78(t,7=7.6 Hz,3H).
i9F-NMR (CDjOD): -139.768.
Example 10
Example 10 was prepared by the method described In Example 1, using cyclopropyl methyl amine in step 10.
LC-MS (method 1 ): tR = 1.02 min; [M+Hf = 393.
'HNMR (CDjOD) δ (ppm); 7.62 (dd, 7 = 8.0,2.0 Hz, 1 H), 7.47 (d. 7 = 8.0 Hz, I H), 7.26 (d, 7 =2.0 Hz, 1 H),3.47(dd,7= 14.8,6.4Hz. 1 H).334(dd,7 = 14.8,6.4Hz, 1 H).3.24(d,7 =16.0Hz. I H). 3.12 (d,7= 16.0 Hz, 1 H),234(t,7= 10.0 Hz, I H), 1.79, (d,7= 12.4 Hz, I H), 1.60-1.40 (m, 3 H), 1.22 (m, 2 H), 1.03 (m. I H). 0.99 (d. 7 = 6.4 Hz, 3 H), 0.94 (d, 7 = 6.0 Hz. 3 H), 0.49 (m, 2H), 032 (m, 2 H).
IS Example 11
Example 11 was prepared by the method described in Example 1, using 6-chloro indan-1-one in the first step and 2-aminomethyl pyrimidine in step 10.
LC-MS (method I ) rR = 0.90 min. m/z 440,442 (MH*) 'H NMR (CDjOD) δ 8.73 (d, 7 = 4.7 Hz. 2H), 738 (t, 7=4.7 Hz. I H), 7.27-7.24 (m, 2H), 5.01 (s,
2H), 3.14-3.05 (m,2H),2.60(1,7=9.8 Hz, 1H), 1.77-1.74 (m, IH). 1.67-138 (m, IH), 135-1.45 (m, IH), 1.40-1.24 (m»3H), 1.01-0.97 (m,6H).
Example 12
Example 12
Step 1: Synthesis of intermediate 71.
LtBuOK, THF,
6-bromo-indan- 1-one 1 ( 100 g, 474 mmol) and methyl acrylate (86.4 g, 995 mmol) were mixed with 800 mL THF and t-BuOK (1.0 g) was added in two portions under ice cooiing. The cooiing bath was removed and the remaining t-BuOK (63.0 g) was added in even portions over 20 min (total of 64.0 g, 569 mmol). The mixture was stirred for 2 h at rt. DMF (240 mL) was added to the reaction mixture, followed by Mel (135 g, 948 mmol) and the mixture was stirred for 2 h. The reaction was quenched with 10% citric acid solution. The reaction mixture was concentrated under reduced pressure and filtered. The cake was washed with water, followed by MeOH to give a crude intermediate which was mixed withTHF/HiO(1.8 L/l.8 L). LiOH*HiO (92.0 g, 2.19 mol) was added. The mixture was stirred for 16 h at rt and then 12 h at 70 °C. The reaction mixture was concentrated under reduced procedure and filtered. The cake was washed with HîO, and then it was stirred with MeOH (50 mL) for 5 min, filtered again, and washed with additional amount of MeOH (50 mL). The solid was collected to give 75 g intermediate 71 which was used as such in the next step.
Step 2: Synthesis of intemediate 72.
10.0 g (32.5 mmol) of intermediate 71 and 530 mg (3.27 mmol) ferrie chloride were mixed with of 200 ml THF. To the stirred mixture 14.0 mL (102 mmol) methoxy trimcthylsilnne and 16.0 mL ( 100 mmol) triethylsilane were added and the mixture was stirred for 35 min at ambient température. The mixture was added to phosphate buffer (pH 7) and stirred for 14 h. The mixture was extracted with ethyl acetate. the organic phase dried and evaporated. The residue was purified by MPLC (340 g silica, cyclohexanes/cthyl acetate (100/0 to 85/15 In 60 min). Fractions containing the product were combined and the solvent was evaporated to yield 369 g of intermediate 72.
LC-MS (method 2): rR » 153 min. m/z 323/5 Br (M+H*) ’HNMR (DMSO-dfi) corresponds with the desired product.
Step 3: Synthesis of intemediate 73.
g
Ti(0Et)4
THF
16.0 g (495 mmol) of intermediate 72 were mixed with 100 mLTHF, 57.0 g (249 mmol) titanium (IV)-cthoxide were added and the mixture was stirred for 1 h at ambient température. After this, 12.0 g (99.0 mmol) of (S)-2-methyl-2-propanesulfinamide were added nnd mixture was refluxed under nitrogen for 3 days. 200 mL of water and 200 mL of DCM were added and the mixture filtered through celite. The orgnnic layer was separated, and the solvent removed under vacuum. The residue was purified by MPLC (600 g silica, cyclohexanes/ethyl acetate ( 100/0 to 75/25 in 3 h, 95/5 to 85/15 in 15 min,0/100 for 10 min). Fractions containing the product were combined. Mixed fractions were again chromatographed by MPLC. The desired product eluted first. The solvent was evaporated to yield 3.69 g of intermediate 73.
LC-MS (method 2): rK = 1.61 min. mh. 426/8 Br (M+H*) 'H NM R (DMSO-d6) corresponds with the desired product
Step 4; Synthesis of intemediate 74,
tBuU
THF
Under nitrogen 4.14 mL g (433 mmol) of ethyIvlnyl ether mixed with 70 mLTHF were cooled to-78 °Cand 25 mLtert-butyllithium(l.7 M in pentane,43.4 mmol) were added. The mixture was warmed to 0 °C and stirred for 30 min. The mixture was transfened by a cannula to a mixture of 3.69 g (8.65 mmol) intermediate 73 in 130 mLTHFat -78 °C. The mixture was stirred for 30 min at this température 100 mLsat. aqueous solution of ammonium chloride were added and the mixture extracted with ethyl acetate. The solvent was removed under vacuum and the residue purified by MPLC (340 g silica, cyclohexanes/ethyl acetate (90/10 to 60/40 in 70 min). Fractions containing the product were combined and the solvent was evaporated to yield 3.62 g of intermediate 74.
LC-MS (method 2): fa = 130 min. m/z. 598/500 Br (M+H*) ’HNMR (DMSO-d6) corresponds with the desired product.
Step 5: Synthesis of intemediate 75.
o3
3.62 g (95%. 6.89 mmol) of intermediate 74 were mixed with 60 mL DCM and 15 mL methanol and cooled to -78 °C. Ozone was bubbled through the mixture for 20 min. The mixture was purged with Nj and treated with 5 ml (68.4 mmol) Me2S at -78 °C. The reaction was allowed to warm
to rt. The solvent was removed under vacuum, the residue was purified by MPLC (340 g silica, cyclohexanes/ethyl acetate (95/25 to 65/35 In 35 min). Fractions containing the product were combined and the solvent was evaporated to yield 230 g of intermediate 75.
LC-MS (method 2): rR = 1.20 min. m/z 500/2 Br (M+H*) ’HNMR (DMSO-d6) corresponds with the desired product.
Step 6: Synthesis of intemediate 76,
650 mg (0.98 mmol) of intermediate 75 were mixed with 8 ml methanol and I ml of a 4 M 10 solution of HCl in 1,4-dioxan was added at 0 ’C. The mixture was stirred for 2 h al the same température. The mixture was evaporated and the remaining crude product 76 used as such forthe next step.
LC-MS (method 2): rR = 1.20 min. m/z 396/8 Br (M+H*) ’HNMR (DMSO-d6) corresponds with the desired product.
Step 7: Synthesis of intemediate 77.
Cl
DCM / water
530 mg (1.23 mmol) of Intermediate 76 and 675 mg (8.04 mmol) NaHCOj were mixed with 8 mL water and 4 ml DCM. 188 pL (234 mmol) thiophosgene were added at 0 ’C while stirring. The mixture was stirred at 0 ’C for I h. The mixture was extracted with DCM, the solvent evaporated and the remaining crude product 77 used as such for the next step.
LC-MS (method 2) tK = 1.74 min. mZi 347/9 Br (M+H4)
Step 8: Synthesis of intemediate 78.
S 510 mg (80 %, 0.93 mmol) of 2-aminomethylpyrimidine hydrochloride was mixed with 3 mL
THF and 290 pL (2.07 mmol) triethylamine were added. After 5 min, intermediate 77 mixed with 7 mL THF was added, and the mixture was stirred at ambient température for 2 h. 290 pL (2.07 mmol) triethylamine were added and the mixture was stirred for an additional 2 h. The mixture was evaporated and the residue purified by MPLC (25 g silica, cyclohexanes/ethyl acetate (110/0 to 70/30 in 50 min). Fractions containing the product were combined and the solvent was evaporated to yield 305 mg of intermediate 78.
LC-MS (method 2):rR= 1.00 min. mh. 501/3 Br (M+H4) 'HNMR (DMSO-d6) corresponds with the desired product.
303 mg (0.60 mmol) of intermediate 78.2.65 mL (14.6 mmol, 53 M in decane) tertbutyhydroperoxide, 10 mL (70.0 mmol, 7 M in methanol) ammonia were mixed and stirred for 14 h at rt. The mixture was evaporated and the residue purified by HPLC (column: Waters Sunfire: eluent A:
water + 0.1% TFA; eluent B: MeOH). Fractions containing the product were combined. the methanol
was evaporated and the residue iyophil ized to yield 155 mg of the intermediate 79 as trifluoro acetîc add sait.
LC-MS (method 2): iR = 1.00 min. m/z 484/6 Br (M+H*) lHNMR (DMS0-d6) corresponds with the desired product.
mg (90 %, 0.11 mmol) of intermediate 79.525 mg (0.26 mmol) 2-(3-pyridy1)-4,455tetramethyi-l,3,2-dioxaborolanc, 16.1 mg 0.022 mmol) ch|oro(2-dicyclohexylphosphino-2’,4'5'-tri-l10 propyl-l,r-biphenyl)[2-(2-aminoethy1)phenyi]palladium(II), 210 pL 2 M aqueous NajCOj solution.
1.4 mL dioxane and 0.75 mL méthanol were mixed in a microwave via! which was charged with argon. The mixture was stirred 30 min at 140 “C in a microwave oven (Biotage). The mixture was fîltered over a thioi cartridge (Agilent Technologies. 500 mg, PL-Thiol MP SPE), the méthanol evaporated and the residue purified by HPLC (column: Waters Sunfire; eluent A: waier + 0.1% TFA;
eluent B: MeOH). Fractions containîng the product were combined, the méthanol was evaporated and the residue Iyophilized to yieid 565 mg of the intermediate 80 as trifluoro acetie acid sait.
LC-MS (method 2): = 1.00 min. mZz 483 (M+H*) ’HNMR (DMSO-d6) corresponds with the desired product.
Step 11: Synthesis of Example 12.
Το a suspension of 25 mg (0.042 mmol) of intermediate 80 in 1 mL chtoroform were added 30 pL (97 %, 0.21 mmot) iodotrimethylsilane and the mixture was stirred for 60 min. The reaction was quenched with 05 mL methanol and 5 mL sat. NaHCOj aqueous solution and 5 mL 10 % NajSOj aqueous solution were added. The mixture was extracted with ethyl acetate and the combined organic layers dried, evaporated and the residue purified by HPLC (column: Waters Sunfire; eluent A: water + 0.1% TFA; eluent B: MeOH). Fractions contaînîng the product were combined, the methanol wus evaporated and the remoining residue lyophilized to yield 15.9 mg of Example 12 as trifluoro acetic acid sait
LC-MS (method 2): r* = 0.84 min. m/z 469 (M+H*) 'HNMR (DMSO-d6): 5 10.96 (br s, 1 H), 958 (br s, 2H), 8.93 (d, 1 H), 8.79 (d, 2H). 8.63 (dd, 1 H), 8.14 (brd, lH),7.76(dd, IH),7.70(brs, lH,759(dd, 1H),75I (m,2H),5.18(d, lH),5.08(d, IH), 4.30(brs. OH)3.16(d, lH),3.02(d, 1H), 2.94(m, IH), 1.78 (m, IH). 158- 1. 24(m,6H),0.92 (d, 15 3H).
Example 13
Example 13
Step 1: Synthesis of intermediate 81.
Zn(CN)a Pd-catalyst
DMA
138 g (90 %, 2.84 mmol) of intermediate 75 were mixed with 60 mL DMA and argon was bubblcd through the mixture. Zinc cyartide (556 mg, 4.74 mmol) and chloro(2dicyciohexylphosphino-2',4’,6,’tri-i-propyl· I, r-biphenyl)[2-(2-aminoethyl)phenyl]palladium(n) (690 mg. 0.93 mmoi) were added nt rt. The mixture was stirred for 20 min at 120 ’C. After this, the mixture was evaporated at 3 mbar at 70 ’C, and the residue was mixed with waterand ethyl acetate, filtered over celite, and the phases separated. The aqueous phase was extracted with ethyi acetate, and the organic phases were combined, dried and evaporated. The residue was purified by MPLC ( 100 g silica, CH/EE 80/20 to 55/45 in 70 min). Fractions containing the product were combined to give 1,17 g of intermediate 81.
LC-MS (method 2): rR = l .40 min. m/z 447 (M+H*) *HNMR (DMSO-d6) corresponds with the desired product.
Step 2: Synthesis of intermediate 82.
Intermediate 82 was synthesized by n method described in Example 12 step 6 from 4.00 g (80%, 7.17 mmoi) intermediate 81. 4.13 g of the crude product were obtained and used as such in the next step.
LC-MS (method 2): fR = 1.11 min. m/z 343 (M+H*) *HNMR (DMSO-d6) corresponds with the desired product.
Intermediate 83 was synthesized by a method described in Example 12 step 7 from 4.13 g (70%, 7.63 mmol) intermediate 82. 4.6 g of the crude product were obtained and used as such in the 5 next step.
LC-MS (method 2): fR « 138 min. mh. 294 (M+H*)
Step 4: Synthesis of intermediate 84.
Intermediate 84 was synthesized in accordance with the method described in Example 12, step 8, from 200 mg (0.33 mmol) intermediate 83. Instead of 2-aminomethylpyrimidine hydrochloride, 63 mg (0.49 mmol) 3,3-dlfluorocyclobutyl)methanamine and 3 équivalents of trieethylamine were used. The crude product was purified by MPLC (25 g silica, CH/EE 65/35 in 45 min) to yield 141 mg of intermediate 84.
IS LC-MS (method 2): rB 133 min. mSi 460 (M+H*)
Intermediate 85 was synthesized by in accordance with the method described în Example 12, step 9 using 139 mg (030 mmol) of intermediate 84. The crude product was purified by HPLC 5 (column: Waters Sunfire; eluent A: water + 0.1% TFA; eluent B: MeOH) to yield 96.8 mg of intermediate 85 as trifiuoroacetic acid sait.
LC-MS (method 2): îr = 1.19 min. m/z 443 (M+H*) ’HNMR (DMSO-d6) corresponds with the desired product.
Step 6; Synthesis of Example 13.
Exampie 13 was synthesized in accordance with the method described in Example 12, step 11 using 40 mg (0.072 mmol) of intermediate 85. The crude product was purified by HPLC (column:
Waters Sunfire; eluent A; water + 0.1% TFA; eluent B: MeOH) to yield 20 mg of Example 13.
LC-MS (method 2): tK = 0.88 min. m/z 429 (M+H*) 'HNMR(DMSO-d6): δ 10.83 (brs, lH),9.65(brs,2H),7.90(d. IH),7.84(dd, lH),739(d, IH), 435 (brs, OH),3.78(m,2H), 3.20(d, lH),3.03(d, lH),2.88(m, IH), 2.68-2.26 (m,5H), 1.72 (m, IH), 1.54(m, IH), U5-1.28(m, 3H), 1.16(m, IH), 1.05 (t, IH),0.91 (d,3H).
Example 14
Example 14
Example 14 was synthesized by the method described in Example 13 using intermediate 17 instead of intermediate 75 in step I.
LC-MS (method 3): « 0.96 min. m/z 443 (M+H*) ’HNMR(DMSO-dâ): δ 10.85 (br s, IH), 9.65 (br s. 2H), 7.88 (d, IH), 7.84 (dd, IH). 7.59 (d, 1 H), 4.40(br s, OH), 3.78 (m, 2H), 3.20(d, IH),3.06(d, IH), 2.68 -2.26(m,6H), 1.60- 1.04(m,6H), 0.92 (d, 3 H), 0.88 (d, 3H).
Example 15 was synthesized by the method described in Example 13 using intermediate 17 instead of intermediate 75 in step l und 2-pyridin-4-yl-ethylaminc Instead of3,3dinuorocyclobutyl)methanaminc in step 4.
LC-MS (method 3): 0.96 min. m/z 4445 (M+H*) 'HNMR(DMSO-dâ): δ 10.81 (br s, IH), 9.70 (brs, 2H), 8.55 (d. 2H), 7.84(brs, IH),7.83(dd, IH), 755 (m, 3H),430 (brs,OH), 4.06-3.90(m,2H), 3.16-2.96 (m,4H), 2.40 (t, IH), l.49(m, IH), 135 (m,2H), 1.18(m, IH), 1.02 - 0.90(m,2H),0.88(d,3H),0.86(d,3H).
Example 16 was synlhesized by the method described in Example 13 using intermediate 17
Instead of intermediate 75 in step I and (2-methyl-pyridln-4-yl)-methylamine Instead of 335 diriuorocyclobutyl)melhanamine in step 4.
LC-MS (method 4): rR = 0.82 min. m/z 444 (M+H*) 'HNMR (DMSOdfi): δ 10.96 (br s, I H), 9.70 (br s, 2H), 8.52 (d, I H), 7.98 (d, lH). 7.85 (dd, 1 H), 7.60 (d, IH). 7.32 (d, 1 H), 7.28 (brs, IH),4.93(s,2H),430(brs,OH),3.20(d, lH),3.03 (d, IH), 232 (s, 3H), 2.47 (m, 1 H), 139 - I. 10 (m, 6H), 0.90 (d, 3H), 0.88 (d, 3H).

Claims (11)

1. A compound represented by a structural formula selected from:
^CCaZ/'0H * t h2n rN x L JOCz’’OH » w VN\ «L JL/C /l0H t h2n /^C3° w IJC/d/,0H * W /-Ζρ y if jzxy °h i R2N / F 1 X o LJZ/Cv0H H2N /—\~p n \- L ZC\/'0H vO l^C/CZ/,,oH R2N ^f H 2 l jCX/‘',0H î HA y* jC/C~/oh * î
N=\ X JlX)oh ch3 . N=\ CCA t ^y-A^F iQOO”,oh » Ι0ΟΟ'“ΟΗ XXX_/,,,oh » ; and H.N /“Λ L xXZX/ 0H t
or a pharmaceutically acceptable sait thereof.
2. A compound according to claim 1 or a pharmaceutically acceptable sait thereof for use as a médicament.
5
3. A pharmaceutical composition comprising at least one compound according to claim 1 or a pharmaceutically acceptable sait thereof In admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier.
4. A compound according to daim 1 or a pharmaceutically acceptable sait thereof for use in the 10 treatment of a B ACE 1 mediated disorder or disease.
5. A compound or a pharmaceutically acceptable sait thereof for use according to daim 4. wherein the BACE1 mediated disorder or disease is selected from the group consisting of a neurodegencrative disorder, cognitive décliné, cognitive impairment, dementia and disease
15 characterized by the production of β-amyloid deposits or neurofibrillary (angles.
C
6. A compound or a pharmaceutically acceptable sait thereof for use according to claim 5, wherein the disorder or disease 1s selected from the group consisting of Alzheimer's disease, Tri s omy 21 (Down Syndrome), Hereditary Cérébral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), senile dementia, cérébral amyloid angiopathy, degenerative dementia, dementias of
5 mixed vascular and degenerative origin, dementia associated with Parkinson's disease, dementia associated with progressive supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, dry âge related maculer degeneration (AMD), and glaucome.
10 7. A compound or a pharmaceutically acceptable sait thereof for use according to daim 6, wherein the disease or disorder is Alzheimer’s disease.
S. A compound or a pharmaceutically acceptable sait thereof for use according to claim 6, wherein the disease or disorder is glaucoma.
9. Use of a compound according to daim I or a pharmaceutically acceptable sait thereof for the manufacture of a médicament for the treatment of a BACE1 mediated disorder in a subject.
10. Use of a compound according to claim 9 or a pharmaceutically acceptable sait thereof,
20 wherein the BACE1 mediated disease or disorder is selected from the group consisting of a neurodegenerative disorder, cognitive décliné, cognitive impairment, dementia and disease characterized by the production of β-amyloid deposits or neurofibrillary tangles.
11. Use of a compound according to daim 10 or a pharmaceutically acceptable sait thereof,
25 wherein the disease or disorder is selected from the group consisting of Alzheimer’s disease, Trisomy
21 (Down Syndrome), Hereditary Cérébral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), senile dementia, cérébral amyloid angiopathy, degenerative dementia, dementias of mixed vascular and degenerative origin, dementia associated with Parkinson's disease, dementia associated with progressive supranuclear palsy, dementia associated with cortical basal degeneration, 30 diffuse Lewy body type of Alzheimer’s disease, dry âge related macular degeneration (AMD), and glaucoma.
12. Use of a compound according to claim 11 or a pharmaceutically acceptable sait thereof, wherein the disease or disorder is Alzheimer's disease.
13. Use of a compound according to claim I lor a pharmaceutically acceptable sait thereof, wherein the disease or disorder is glaucoma.
OA1201500052 2012-08-27 2013-08-26 Inhibitors of beta-secretase. OA17202A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US61/693,512 2012-08-27
US61/788,839 2013-03-15
US61/816,458 2013-04-26

Publications (1)

Publication Number Publication Date
OA17202A true OA17202A (en) 2016-04-05

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017236042B2 (en) Inhibitors of beta-secretase
AU2018391675B2 (en) Sulphonyl urea derivatives as NLRP3 inflammasome modulators
US9018391B2 (en) Inhibitors of beta-secretase
AU2017223416A1 (en) Heteroaryl inhibitors of PAD4
CA2957898C (en) Pyrrolopyrimidine derivatives as nr2b nmda receptor antagonists
AU2016366405A1 (en) Aza-benzimidazole inhibitors of PAD4
SG190246A1 (en) Kat ii inhibitors
OA17202A (en) Inhibitors of beta-secretase.
US9290477B2 (en) Inhibitors of β-secretase
OA17087A (en) Inhibitors of beta-secretase.
JP2023527792A (en) TLR2 modulator compounds, pharmaceutical compositions and uses thereof