OA17087A - Inhibitors of beta-secretase. - Google Patents

Inhibitors of beta-secretase. Download PDF

Info

Publication number
OA17087A
OA17087A OA1201400390 OA17087A OA 17087 A OA17087 A OA 17087A OA 1201400390 OA1201400390 OA 1201400390 OA 17087 A OA17087 A OA 17087A
Authority
OA
OAPI
Prior art keywords
disease
compound
mmol
pharmaceutically acceptable
mixture
Prior art date
Application number
OA1201400390
Inventor
Yuri Bukhtiyarov
Salvacion Cacatian
Lawrence Wayne Dillard
Cornelia Dorner-Ciossek
Klaus Fuchs
Lanqi Jia
Deepak S. Lala
Angel Morales-Ramos
Georg Rast
Jonathan Reeves
Suresh B. Singh
Shankar Venkatraman
Zhenrong Xu
Jing Yuan
Yi Zhao
Yajun Zheng
Original Assignee
Boehringer Ingelheim International Gmbh
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 International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Publication of OA17087A publication Critical patent/OA17087A/en

Links

Abstract

The present invention relates to spirocyclic acylguanidines and their use as inhibitors of the ßsecretase enzyme (BACEl) 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 ß-amyloid aggregates.

Description

INHIBITORS OF BETA-SECRETASE
RELATED APPLICATIONS
This application daims the benefit of U.S. Provision^ Application Serial No. 61/606786, fîled Marc h 5, 20(2, the entire teachings of which are incorponited herein by référencé.
FIELD OFTHE INVENTION
The présent invention relates to spirocyclic acylguanîdincs and their use as inhibitors of the β-secretase enzyme (BACE I) 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 décliné, cognitive impairment, dementia and diseases characterized by production of β-amyloid deposits or neurofibrillary tangles.
BACKGROUNDOFTHE INVENTION β-Amylotd (also rcferred te herein as “Abeta or “Αβ”) deposits and neurofibrillary tangles are two major pathologie characterizations associated with Alzheimer's disease (AD), including the genetically linked early onset familial forms due to mutations in amyloid precursor protein (APP), presenilin 1 and 2, as well as late onset sporadic AD. Clinically, AD ïs characterized by the loss of memory, cognition, reasoning, judgment, and orientation. Also afïected, as the disease progresses, are motor, sensory and Iinguistic abilities until global impairment of multiple cognitive functions occurs. Thèse cognitive losses take place gradualiy, but typically lead to severe impairment and eventual death in 4-12 years. β-Amyloid deposits are predominantly on aggregate of Abeta peptide, which in tum is a product of the proteolysis of APP. More specifically, Αβ peptide results from the cleavage of APP at the C-terminals by one or morey-secrelases, and at the N-terminus by β-secretase enzyme (BACEI), also known ns asparty) protease and memapsin2, as part of the β-amyloidogenic pathway.
B ACE activity is conelated directly to the génération of Αβ peptide from APP, and studies Increasingly indicate that the inhibition of BACE inhibits the production of Αβ peptide.
Amyloidogenic plaques and vascular amyloid angiopathy also characterize the bratns of patients with Trisomy 21 (Down Syndrome), Hereditory Cérébral Hemonhage with Amyloidosis of the Dutch-type (HCHWA-D), and other neurodegenerative disorders. Neurofibrillary tangles also occur in other neurodegenerative disorders induding dcmentia-inducing disorders.
Recently, Abeta has been reported to be implicated in the development of rctinal ganglion cell (RGC) apoptosis in glaucoma, with evidence of caspase-3-mediated abnormal amyloid precursor protein processing, increased expression of Abeta in RGCs în experimental glaucoma and decreased vitreous Αβ levels (consistent with retinal Αβ déposition) in patients with glaucoma. Amyloid deposits hâve also been associated with macular degeneration in patients sufTcring from dry age-related macular degeneration (AMD) and in animal models of A MD.
WO 2010/02(680, WO201 l/l 06414 and WO 2010/105179 disclose spirocyclic acylguanidines with a spirocyclic scaffold as inhibitors of beta-secretase.
SUMMARY OF THE INVENTION
The present invention provides compounds that are BACE1 inhibitors and are useful os therapeutic agents in the treatment of a disease or disorder characterized by elevated β-amyloid deposits or β-amyloid levels in a patient The disclosed BACE1 Inhibitors are not only highly potent inhibitors of the BACE1 enzyme (assay 1) but also show:
(1) highly potent inhibitory activity in the cellular Abeta assay (assay 2), (2) selectivity against the cardiac hERQ channel in a cellular assay (assay 3), and (3) a low propensity to cause phospholipidosis in a cellular phospholipidosis assay (assay 4), as well. Thus, the present invention provides compounds which show a combination of high potency as BACE1 inhibitors, high selectivity against the cordtac hERQ channel, and low phospholipidosis activity.
One embodiment of the invention is a compound represented by a structural formula selected from:
or a pharmaceutically acceptable sait of any of the foregoing compounds. The immediately foregoing compounds are referred to herein as “compounds of the présent invention.
Another embodiment of the invention is a compound of the présent invention or a pharmaceutically 10 acceptable sait thereof for use as a médicament.
Another embodiment of the invention is a pharmaceutical composition comprising a compound of the présent invention or a pharmaceutically acceptable sait thereof in ad mixture with a pharmaceutically acceptable adjuvant, diluent or carrier.
Another embodiment of the invention is a compound of the présent invention or a pharmaceutically xceptable sait thereof for use in the treatment of a BACE1 mediated disorder or discase in a subject. Another embodiment of the invention is the use of a compound of the présent invention or a pharmaceutically acceptable sait thereof for the manufacture of a médicament for the treatment of a B A CEI mediated disorder in a subject.
Anotherembodiment ofthe invention is a pharmaceutical composition fortreatment ofa BACE1 mediated disorder or disease in a subject comprising a compound of the présent invention.
Another embodiment of the invention is a method of treating a subject with a BACEl mediated disease or disorder, comprisingadminîstering to the subject an effective amount of a compound of the présent Invention or a pharmaceutically acceptable sait thereof.
Yet another embodiment of the invention is an intermediate used in the préparation of a compound of the présent invention. These intermediates are represented by a structural formula selected from:
OEt
DETAILED DESCRIPTION OF THE INVENTION
The compounds of the présent invention exhibit potent activity against the BACEI enzyme and Abeta formation together with the selectivity against the hERG channel and the low propensity to cause 10 phospholipidosis. For example, the compounds of the présent invention show a BACE! inhibition with an IC» < 15 πΜ, a cellular Abeta production inhibition with an ICM of < 2 nM. a hERG inhibition of <50% at !0 μΜ, and phospholipidosis with a First EfTect Concentration (FEC) of > 150 μΜ. These combined properties make the compounds of the présent invention useful for the treatment of pathoiogical states in humans, in particular, for the treatment of Alzheïmer's disease as wel] as other disorders and 15 diseases mediated by B ACE !.
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 fora spécifie polymorphie ventricular tachyarrhythmia, torsade de pointes, as established by Sanguinetti et al. (1995, Ceil, Apr. 21,81 (2):299307) and a large body of subséquent evidence. To avoid this risk early on, screening against hERG interaction in an in vitro System using heterologous expression of the hERG channel is common practice and on assay of this type is also an important part of later preclinical candidate profil i ng as recommended by the 1CH guideiine S7B (International Conférence on Harmonization (2005): ICH Topic S 7 B The
nonclinical Evaluation of the Potential for delayed Ventricular Repolarization; (QT Interval Prolongation) by Human Pharmaceuticals (www.lch.org/products/guidelines/safety/article/safety-guidelines.html)). As such, low hERG channel inhibition, such as that shown by the compounds of the présent Invention, is highly désirable for therapeutics.
Phospholipidosis is a lipid storage disorder in which excess phospholipids nccumulate within cells. Druginduced phospholipidosis is an undesïrable dnig réaction. Therefore, in order to avoid detrimental sida effects, compounds with low phospholipidosis potential are preferred for human therapeutic use.
Data provided in Table 1 below show that compounds of the présent Invention hâve this combination of potent BACE1 cellular activity, selectivîty against cardiac hERG and low propensity to cause phospholipidosis. It is believed that the compounds exemplified in WO 2010/021680 and
WO 2010/105179 do not hâve this combination of désirable propertîes. In addition. Table 2 provides data showing that certain compounds of the présent invention hâve significantly lower ICM irrhibitory values in a BACEI enzymatic assay and also in a cellular Abeta assay relative to certain comparut or compounds described in WO 2010/105179.
Terms not specifically defined herein should be given the meanîngs that would be gi ven to them by one of skill in the art in light of the disclosure and the context. As used in the spécification, however, unless specifîed to the contrary, the following terms hâve the meaning indicated and the following conventions are adhered to.
When a compound of the présent invention is depicted by name or structure without indicatîng ali 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 indicatîng 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, 25 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 lhe named or depicted stéréo, optical or géométrie isomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% pure by weight. Stereo, optical and géométrie 30 isomeric purity is determined by dividïng the weight of the named or depicted stereo, optical and géométrie Isomer in a mixture by the total weight of ail stereo, 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 35 solvatés, hydrates and anhydrous form of ils pharmaceutically acceptable sait are included in the invention. “Solvatés refer to crystalline forms wherein solvent molécules are incorporated into the crystal lattice during crystallizatîon. Solvaté may include water or nonaqueous solvents such as éthanol,
isopropanol, DMSO, acetic acid, ethanolamine, and EtOAc. Solvatés, wherein water is the solvent molécule incorporated into the crystai lattice, are typically referred to as “hydrates. Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. “Anhydrous form refers to compounds with no solvent or water or substantially no solvent or water incorporated into the 5 crystai structure (e.g., less than 1:10, 1:20; 1:100; or 1:200 molar ratio ofsolvent 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 10 use in contact with the lissues of human beings and animais without excessive toxicity. Irritation, allergie response, or other problem or complication, and commensurate with a reasonable benefît/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 are not limited to, minerai or organic acid salts of basic 15 residues such as amines; alkali or organic salts of acidic residues such as corboxylic acids; and the like.
For example, such salts include salts from ammonia, L-arginine, betaine, be net ha mi ne, benzathine, calcium hydroxidc, choline, deanol, diethanolamine (2,2’-iminobis(ethanol)), diethylamine, 2(dïethylamino)-ethanol, 2-aminoethanol, ethylenediamine, N-ethyl-glucamine, hydrabamine, 1Himidazole, lysine, magnésium hydroxide, 4-(2-hydroxyethyl)-morpholine, piperazine, potassium 20 hydroxide, l-(2-hydroxyethyl)-pyrn)lidine, sodium hydroxide, triethanolamine (2,2* ,2nitrilotris(ethanol)), trame thamine, zinc hydroxide, acetic acid, 2.2-dIchloro-acetic acid, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 2,5-dihydroxybenzoic acid, 4-acetumîdo-benzoic acid, (+)-camphoric acid, (+)-camphor-10-sulfonic acid, carbonîc acid, cinnamic acid, citric acid, cyclamic acid, decanoic acid, dodecylsulfuric acid, elhone-l^-disulfonic acid, 25 ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, ethylenediaminetetraacctic acid, formic acid, fumarie acid, galactaric acid, gentîsic acid, D-glucoheptonîc acid, D-gluconic acid, D-glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycine, glycolic acid, bexanoic acid, hippuric acid, hydrobromic acid, hydrochloric acid, isobutyric acid, DL-lactic acid, laclobionic acid, lauric acid, lysine, maleîc acid, (-)-L-malic acid, malonic acid, DL-mandelic acid, methanesulfonîc acid, 30 galactaric acid, naphthalene- 1,5-disulfonic acid, naphthalene-2-sulfonic acid, l-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, octanoic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid (embonîc acid), phosphoric acid, propionic acid, (-)-L-pyroglutamic acid, salicylic acid, 4-amino-salicylie acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid and undecylenic acid. Further pharmaceutically acceptable salts can be 35 formed with cations from metals like aluminium, calcium, lithium, magnésium, potassium, sodium, zinc and the like (sec also Pharmaceutical salis. Berge, S.M. et al., 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 ncidic moiety by conventîonal chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a sufficîent amount of the appropriate base or ncid in water or in on organic diluent like ether, ethyl acetate, éthanol, isopropanol, or acetonitri le, or a mixture thereof.
Salts of acids other than those mentioned above which for example are useful for purifying or isolating the compounds ofthe présent invention (e.g., trifluoro acetate salts) also comprise a part of the invention.
Blological Dota
BACEI Assay (Assay 1)
The inhibitory activity of compounds was assessed by a fluorescence quench assay of BACE1 activity using commcrcially available substrate HiLyte Fluor™488-Glu-Val-Asn-Leu-Asp-Ala-GIu-Phe-Lys(QXL™ 520)-OH (SEQ ID NO:1) AnnSpec, San José, CA) and truncated human beta-secretase, BACEI 15 (amino acids 1-454) fused to a myc-his tag and secreted from HEK293/BACEert. cells into OptiMEM™ (Invitrogen). The substrate was dissolved at I mg/ml in DMSO.
The assay was performed in the presence of OptiMEM (supematant collected over 24 h and cleared from cellular débris by centrifugation) containing the ectodomain of B ACE 1,25 μΙ water containing the desired 2-fold concentration of test compound and 2% DMSO, 1 μΜ substrate peptide, 20 mM NaOAc, 20 pH 4.4, and 0.04% Triton-X100 in a total assay volume of 50 μ! in a 384 well plate. In general, 25 μΐ of compound dilution were given to the plate followed by the addition of 10 μΙ of BACEI containing OptiMEM™ diiuted ! : 10 in water with 02% Triton X-100. The reaction was started with the addition of 15 μ! substrate in NaOAc buffer. The reaction was incubated at rt (dark) in an Envisîon* multïjabel reader (Perkin El mer) and the cleavage of the substrate was rccorded as kinetic for 60 min at ex: 485 nm, 25 ent 538 nm. Blank wells containing no enzyme were included 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 calculating percent control using an uninhibited control containing 1% DMSO as 100% and blank central performed in the absence of enzyme as 0%. IC» values were calculated by fitting percent control w. test compound concentration using Assay Explorer.
H4-APPwt Ced-Bosed Assay (Assay 2)
Cellular potency of the compounds of the invention was assessed in an assay monitoring production of Abetal-x peptides in the H4 neuroglioma cell line (ATCC, Cat. #HTB-148) stably expressing human APP, using on immunoassay such os AlphuLISA (PerkînElmer, Cat.# AL288). Tested compounds were 35 dissolved in DMSO and pre-diluted in the culture medium (DMEM containing 10% FBS and 1% penicillin/streptomycin) to achieve twice the final concentration of the compounds in the assay. Equal volumes of the 2x solutions of the tested compounds and cell suspension were added to a 96-well culture plate, so that each well contaïned-10,000 celfs in a final volume of 200 μ!. Final concentration of
DMSO in the assay was 0.2%. The plates were incubated for 5 hrs at 37°C, 5% COj to allow cells to attach to the bottom of the wells in the presence of the tested compounds. Then the media was removed and replaced with fresh media containing tested compounds at the same final concentration. The plaies were incubated for 18 hrs at37°C, 5% COj. Concentrations of Abl-x were determined using AlphaLISA îmmunoassay (PerkinElmer, Cat# AL288) following manufacturées protocol. Concentrations of Abetal-x in the wells containingeither DMSO or 10 μΜ beta-secretase inhibitor (BACE ïnhibitor IV, EMD Biosciencc, Cat. #565788) were used as uninhibited and background contrôla, correspondingly, for calculatlng percent inhibition values for each well with the tested compounds, These percent inhibition values were regressed against compound concentrations using four-parameter curve fitting, and the IC50 values (concentration of a compound at which 50% of the inhibltory effect was observed) was calculated as the compound concentration corresponding to the inflection point on the curve.
hERG-Channel Assay (Assay 3)
Cells:
HEK (human embryonic kïdney) 293 cells were stably transfectcd with hERG cDNA.
Pipettes and solutions:
Cells were superfuscd with a bath solution containing (mM): NaCI (137), KCI (4.0), MgClj (1.0), CaClj (1.8), Glucose (10), HEPES (10), pH 7.4 with NaOH. Patch pipettes were made from borosilicate glass tubing using a horizontal pu lier and filled with pipette solution containing (mM): K-aspartate (130), 20 MgClj (5.0), EGTA (5.0), KjATP (4.0), HEPES (10.0), pH 7.2 with KOH. 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 clamp amplifier and PatchMaster software. hERG-mediatcd membrane currents were recorded at 35°C, using the whole<cll configuration of the 25 patch-clamp technique. Transfccted HEK293 cells were clamped at a holding potential of-60 mV and hERG-mediated inactivating tail currents were elicited using a puise pattem with fixed amplitudes (activation/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 întervals. During each inter-pulse interval 4 puises sealed down by a factor of 0.2 were recorded for a P/n leak subtraction procedure. R, 30 compensation was employed up to a level that safcly allowed recording devoid of ringing.
Compound préparation and application:
The different concentrations of the test compound were applied scquentially 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 dissoived în DMSO to yicld a master stock solution which was diluted further in DMSO to stock solutions needed for the lower concentrations. Final dilutions in extraccllular buffer were prepared freshly from lhese stocks by a 1:1000 dilution step each before starting the experiments.
Data analysis:
Peak current amplitudes were measured 3 ms after the ramp to +40 mV. For baseline and each concentration the peakcurrents of the three last sweeps before application ofthe next concentration were averaged. Residual currents (I/W were calculated for each cell as the fraction of actua! average peak 5 current and average baseline peak current. Results are presented as percent (%) Inhibition (l-I/W* 100% at lOpM.
In vitro Phosphollpidosis Assay (Assay 4)
The phospholipldogenic potential of test compounds was assayed using the human hematopoetic U937 cell line. The test principle was to analyze the phospholipid content by staining the cells with the fluorescent dye Nilc red.
U937 cells were seeded into cell culture plates at 0.5 x 10e cells/mL în RPMI medium containing 10 % FBS, l % DMSO, and 0.005 % gentamicîn. The cells were then cultivated with or without different concentrations of test compounds for48 h under standard culture conditions.
For harvesting the cells were centrifuged at 130x g for 4 min and washed once with PBS. Then 2x 0 J mL cell suspension was prepared for non-fixed cell measurement (0.5 mL for propidium iodide (PI) viability measurement and 0.5 mL forNile red measurement).
The remainlng cells were fixed with 3.7 % formaldéhyde for 30 min. After a further centrifugation step cells were resuspended with 13 mL Nile red working solution ( l pg/mL) and incubated for 5 min at it.
The cell suspension was then washed twice with 3 mL PBS and centrifuged with 130x g for 4 min. The supematant was discarded and the cells were resuspended with 0.5 mL PBS and kept for flow cytometry measurement.
ForNileredstainingofthe0.5 mL non-fixedcell samples,50pLofaready touseNile redsolution(10 pg/mL) were added per sample. Samples were kept on ice for 5 min. Thereafter, they were washed once 25 with 4 mL PBS (4°C, 250x g for 8 min) and finally resuspended in 400 pL PBS and kept for flow cytometry measurement.
For the viability measurement, 123 pL ofthe ready to use PI solution (10 pg/mL) were added tothe 0.5 mL non-fixed cell suspension. After 15 min of incubation on ice. the samples were measured by flow cytometry using a CouIter Epies XL/MCL flow cytometer.
The viability of the cells of each sample was determined by flow cytometry measurement of the PI content nt channel 2 (568-590 nm). Cut-off gates for the fluorescence-dependent 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 analyzed for phospholipidosis. For that, each Nile red sample (non-fixed and fixed samples) was measured by flow 35 cytometry at channel l (504-541 nm) and channel 4 (660-680 nm).
For each channel, relative Nile red fluorescence intensity of a test sample was calculated compared to control samples and expressed as percentage of control fluorescence intensity. The assessment of the
phospholipidogenic potential and the first efleci concentration (FEC) of a test compound was donc manually based on the fluorescence intensifies at both wavelengths for the fixed cells os well os for the non-fixed cells.
Rat Brain AflLo wering Assay (Assay 5)
The in vivo efficacy 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 demonstrate the ability of compounds of the invention to reduce brain omyioid peptides Αβί-χ. Compounds were administered via oral gavage in 1% Polysorbate-80 and 0.5% Natrosoi*, at the 10 single dosages indicated in Table 3. The animais were sacrificed 3 hrs after dosing, and brains were excised, dissected into cerebeilum and left and right cerebra and flash-frozen in liquid nitrogen.
Thecerebntm was homogenized (5 volumes per weight) ln 20 mM Tris-HCL, pH 8J, 02% Triton-XlOO suppiemented with protease inhibitors (cOmplete, Roche Applied Science) at 4’C using a gloss Dounce homogenizcr. The homogenate was centrifuged at 120,000xg for 60 min ut 4'C, and the supematant was 15 coliected and analyzed for Abl-x using immunoossay with chemiluminescent détection (Meso-Scale
Discovery, Rock vil le, MD (MSD)).
Streptavidin 96-well plates (MSD) were pre-blocked with 5% Blocker A solution (MSD) for 1 hr at rt on on orbital shaker and washed 4 times with phosphate bufTered saline (PBS). The wells were pre-coated with 20 ng/weil of biotinylated antibody S10-39155 (Clone M3.2, spécifie for amino acids 10-15 of the 20 rodent Αβ) for 1 hr at rt and washed 4 times with PBS. For Αβί-χ analysis, 25 μΙ of either the cleared brain lysâtes or ΑβΙ-40 standards (8-500 pg/ml, with 2x incrément) were Incubated for 1 hr at rt with constant shaking. The wells were washed 4 times with PBS, and 25 μΐ of the détection antibody (SuifoTAG labeled αηΐϊ-Αβ40 antibody supplied by MSD) was added and incubated for 1 hr at rt. After 4 washes with PBS, 150 μΙ ofthe chemiluminescence détection reagent (Read BufferT, MSD) was added, 25 and the plate was read on an MSD Sector Jmager 6000 instrument. The calibration curve was fit into a non-lincarfour-parameter régression model, and the Αβί-χ concentrations were calculated foreach 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 brains from the animais treated with vehicle alone.
Table I shows the following properties of the compounds of the présent invention: B ACE 1 inhi bit ory potency os measured in assay 1, cellular inhibitory potency as measured in assay 2, hERG inhibition as measured ïn assay 3, and first effect concentration (FEC) of phosphol ipidosis as measured in assay 4.
Table 1. Biologlcal properties of compounds of the présent Invention
Example BACE1 ICjo nM (assay 1) 114-APPw t ccll IC$o nM (assay 2) hERG % inhibition @ 10 μΜ (assay 3) Phospholipldosls FEC ICsoMM (assay 4)
Example BACE1 IC» nM (assay 1) H4-APPwt ccll IC» nM (assay 2) hERG % inhibition @ 10 μΜ (assay 3) Phospholipidosis FEC ICjq μΜ (assay 4)
1 li 0.76 8 200
2 8 0.29 22 200
3 10 0.57 9 > 200
4 5 0.28 0 > 400
5 8 0.90 7 200
6 II 1.24 35 200
7 2 1.42 8 400
8 5 0.49 16 > 200
9 9 1.90 16 400
10 9 1.12 38 200
il 4 0.35 6 > 400
12 3 1.10 II 400
13 9 0.92 18 200
14 5 0.12 20 200
Example BACE1 ICm nM (assay 1) H4-APPwtcell ICjo nM (assay 2) hERG % Inhibition ® 10 μΜ (assay 3) Phospholipidosls FEC lCmgM (assay 4)
15 II 0.11 11 200
16 11 0.20 6 200
17 14 0.89 12 > 200
18 9 1.02 17 > 400
19 6 0.40 8 400
20 0.28 8 > 400
21 5 0.48 24 400
22 8 0.26 9 > 400
23 12 0.18 16 > 200
24 5 0.65 23 200
25 0.99 10 200
26 9 0.20 S > 200
27 6 0.95 19 > 200
28 0.5 9 400
Example BACEI ICm nM (assay 1) H4-APPwtcell IC»nM (assay 2) hERG % Inhibition O 10 μΜ (assny 3) Phospholipldosls FEC IC$o μΜ (assay 4)
29 0.3 7 200
Table 2 provides data showing that certain compounds of the présent invention hâve significantly lower IC» inhibitory values in the BACEi enzymatic assay (Assay i) and ln the cellular Abetaassay (Assay 2) 5 relative to certain comparator compounds described in WO 2010/105179.
Table 2.
Compounds of the présent invention
Comparison examples:
Example 5 Example 14
Example 15 example 281 in
WO 2010/105179
BACE ICjo (assay i ) 35 nM
H4-Cell assay (assay 2) 7.0 nM hERG (assay 3) 16% at 10 μΜ
exampte 2S9 in
WO 2010/105179
BACE IC» (assay 1) 49 nM
H4-Cell assay (assay 2) 16 nM
Example 8 Exampie 11
example 121 in
WO 2010/105179
B ACE IC» (assay 1) IO7nM
H4-Ccll Assay (assay 2) 16 nM
example 320 In
WO 2010/105179
BACEICM (assay 1)415 nM
H4-Cell Assay (assay 2) 16 nM
The abilily of compounds of the invention to reduce brain Αβ was demonstrated in rats, as described in Assay 5, and the in vivo efficacy data are presented in Table 3.
Table 3«
Example Dose (mg/kg) % Αβ Réduction
2 30 42
4 25 75
6 50 60
8 25 37
9 25 37
12 25 39
13 30 47
14 25 67
15 25 62
Example Dose (mg/kg) % Αβ Réduction
17 25 70
18 25 56
20 25 73
21 50 59
22 12.5 45
23 25 68
26 25 71
28 12J 30
29 25 78
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 in a subject wherein the inhibition of the 5 activity of the β-secretase enzyme (BACE1) is of therapeutic benefit, including but not limited to the treatment, amelioration or prévention of neurodegeneratîve disorders, disorders characterized by cognitive décliné, cognitive impairment, dementia, and diseases characterized by production of β-amyloid deposits or neurofibrillary tangles.
Compounds of the présent invention nre useful for treatment of Alzheimcr's disease, Trisomy 21 (Down 10 Syndrome), Heredttary Cérébral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), senile dementia, cérébral amyloid ongtopathy, dégénérative dementia, dementias of mixed vascular and degenerative origin, dementia associated with Parkinson's disease, dementia associated with progressive supronuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimcr’s disease, dry âge related macular degeneration (AMD), and glaucome. The “dry form of 15 AMD, also known as central géographie atrophy, results from atrophy to the rrtinal pigment épithélial layer below the neurosensory retina, which causes vision loss through loss of photoreceptors (rods and cônes) în the central part of the eye. No medical or surglcal treatment is currently availabie for this condition. Treatments availabie so far (e.g., suggested by the National Eye Institute) include the use of vïtamin suppléments with high doses of antioxidants, lutein and zeaxanthin, which may slow the 20 progression of dry macular degeneration. Glaucoma is a disease where fluid pressure inside the eye increases, causing irréversible damage to the optic nerve and loss of vision. Abeta colocalizes with apoptotic retina! ganglion cells in experimental glaucoma and induces significant retinal ganglion cell apoptosis in a dose- and time-dependent manner.
Accordingly, the présent invention relates to a compound or a pharmaceutically acceptable sait thereof for 25 use 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 οΓ therapeutic benefit.
Furthermore, the présent invention relates to the use of a compound in the treatment of neurodegenerative 5 disorders. disorders characterized by cognitive décliné, cognitive impairment, dementia, and diseases characterized by production of β-amyloid deposits or neurofïbrillary tangles.
Therefore, the présent invention relates to the use of a compound of the présent invention in the treatment of Alzheimer’s disease, Trisomy 21 (Down Syndrome), Hereditary Cérébral Hemorrhage with Amyloldosls ofthe Dutch-type (HCHWA-D), sentie dementia, cérébral amyloid angiopathy, dégénérai!ve 10 dementia, dementias of mixed vascular and degeneratîve origin, dementia associated with Parkinson's disease, dementia associated with progressive supranuclear palsy, dementia associated with cortical basal degeneratlon, diffuse Lewy body type of Alzheimer*! discase, dry AMD, and glaucoma.
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 an 15 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 subject in need thereof, comprising administering to a subject and/or contacting a receptor thereof with an effective amount of at 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 20 administering to said subject an effective amount of at least one disclosed compound or a pharmaceutically acceptable sait thereof.
The invention includes a therapeutic method for treating or ameliorating a BACEI mediated disorder in u subject In need thereof comprising administering to a subject in need thereof an effective amount of a compound of the invention described herein, or pharmaceutically acceptable salts thereof or composition 25 thereof.
As used herein, the term “subject and “patient” may be used interchangeably, and means a mammal in need of treatment, e.g., companion animais (e.g., dogs, cals, and the like), 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 'treatment” refers to obtaining desired pharmacological and/or physiological effect. The effect can be prophylactic (l.e., rcducing the likelihood of developing the disorder or disease) or therapeutic, which includes achievîng, partially or substantialiy, one or more of the following results: partially or totally rcducing the extent of the disease, disorder or syndrome; ameliorating or improving a clinical symptom or indicator associated with the disorder; or delaying, inhibiting or decreasing the likelihood of the progression of the disease, disorder or syndrome.
The dose range of the compounds according to the présent invention applicable per day îs 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 conveniently 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 factors known by those skilled in the art such as 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 manner which 5 allows a pharmaceutically effective amount to be delivercd based upon patient's unique condition.
Pharmaceutical Compositions
Suitable préparations for administering the compounds of the présent invention will be apparent to those with ordtnary skill in the art and include for example tablets, pills, capsules, suppositories, lozenges, 10 trochcs, solutions, syrups, élixirs, sachets, injectables, inhalatives and powders, etc. The content of the pharmaceutically active compound(s) should be in lhe range from 0.1 to 95 wt.-%, preferably 5 to 90 wt.% of the composition as a whole.
Suitable tablets may be obtained, for example, by mixing one or more compounds of the invention with known excipients, for example inert diluents, carriers, dîsintegrants, adjuvants, surfactants, binders and/or 15 lubricants. The tablets may also consist of several layers.
Combination Thcrapy
In one embodiment, the présent invention includes combination thcrapy for treating or ameliorating a disease or a disorder described herein. The combination therapy comprises administering a combination 20 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 actîng neuroprotectîve and/or disease-rnodifytng substances; anti-oxidants (e.g., vitamin E or gtnkoli de); untiinflammatory substances (e.g., Cox inhibitors, NSAIDs additiona lly or exclusively having Abeta lowering 25 properties); HMG-CoA reductase inhibitors (statins); acétylcholinestérase inhibitors (e.g., donepezil, rivastigmine, tacrine, galantamine,; tacrine); NMDA receptor antagonists (e.g., memanline); AMPA receptor agonists; AMPA receptor positive modulators, AMPAkines, monoamine receptor reuptake inhibitors, substances modulating the concentration or release of neurotransmittcrs; substances inducing the sécrétion of growth hormone (e.g., ibutamoren mesylale and capromorelin); CB-1 receptor antagonists 30 or Inverse agonists; antibiotics (e.g., minocyclin 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, alpha2-adrenoreceptor 35 antagonists, calcium antagonists, muscarinic receptor ΜI agonists or partial agonists or positive modulators, muscarinic receptor M2 antagonists, muscarinic receptor M4 antagonists. metabotropic glutamate-receptor 5 positive modulators, antidepressants, such as citalopram, fluoxetine, paroxetine.
sertraline nnd trazodone; nnxiolytics, such as lorazépam and oxazepam: antîphychotics, such as aripïprazole, clozapine, haloperidol, olanzapine, quetiapine, rispéridone and ziprasidone, and other substances that modulate receptors or enzymes tn 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 immunisation with Abeta or parts thereof or passive immunisation with humanised anti-Abeta antibodies or nanobodies) for the treatment of the above-mentioned diseases and conditions. Combination therapy indudes co-administration of the compound of the invention with one or more other agent, sequentïal administration of the compound nnd one or more other agent, administration of a 10 composition containing a compound and one or more other agent, or simultaneous administration of separute compositions containing the compound and one or more other agent.
EXPERIMENTAL SECTION
Methods of Préparation of Compounds
Compounds of the invention cnn be prepared employing conventional methods that utilize readily available reagents and starting materials. The reagents used in the préparation of the compounds of this invention can be either commercially obtained or can be prepared by standard procedures described in the literature.
Micro wave reactions were carried out in CEM reactor using discovery SP System.
Where NMR data are presented. spectra were obtained in Varian -400 (400 MHz) and are reported as ppm downfield from tetramethylsilane with number of proton, multiplicities and coupling constants indicated parenthetically along with référence to deuterated solvent.
Compounds were purified by basic préparative HPLC method as described below. Mtthod I;
Mobile phase A: water with 0.05% ammonia solution; 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 ln min %A %B
0.0 68 32
12.00 38 62
12.20 0 100
S 13.5
100
13.7 90 10
Method 2:
Mobile phase A; water with 0.05% ammonia solution: Mobile phase B; ACN: Flow rate: 25 mlVmin; 10 Détection: UV 220 nm/254 nm; Column: Durashell CI8 250*30mm‘5tim; Column température: 30°C
Time in min %A %B
0.0 67 33
12.00 47 53
12.20 0 100
15 13.5 0 100
13.7 90 10
LC-MS data were obtained by utilizing the following chromatographie condition:
HPLC System: Waters ACQUTTY; Column: Waters ACQU1TY CSH™ C18 1.7 μΜ
Guard column: Waters Assy. Frît, 0.2 μ M, 2.1 mm; Column 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 μΕ; Acquisition time; approximately 1.5 min.
Gradient Prognun:
Time (min) B%
0 10
0.8 90
1.20 90
121 10
Mass Spectrometer Parameters
Mass Spectrometer Waters SQD; lonization: Positive Electrospray Ionization (ESI); Mode Scan (1004400 m/z in evety 0.2 second); ES Capillary Voltage:3.5 kv; ES Cône Voltage: 25 v Source Température : 120 °C; Disolvation Température; 500 °C; Desolvatîon Gas Flow: Nitrogen Setting 650 (L/h); Cône Gas Flow: Nitrogen Setting 50 (L/h)
For Example 10, step 2, and the alternative synthesis of Intermediate 38, step I and 2, the following chromatographie conditions and instrumentation were used:
LC-MS data were obtained by utilizing the following chromatographie condition:
HPLC System: Agiient i 100 Sériés
Column: Zorax Eclipse XDB-C8,2.1x50mm
Column tem: Mobile Phase: 35 °C A: Formic Acid : Water ( 1:1000, v:v) B: Formic Acid ; ACN (1:1000, v:v) Gradient Program:
Tlmc (min) 0 3 4.5 5.0 B% 5 95 95 5
Flow Rate: 0.60 mL/min
injection Volume: 2pL
Rétention Times: Approximately 1-4 min
Acquisition time: approximately 5 min
Mass Spectrometer Parameters
Mass Spectrometer: Agiient 77
lonization Positive Electrospray lonization (ESi)
Mode ES Capillary Voltage; Scan (100-800 m/z in every 0.2 second) 3.5 kv
ES Cône Voltage: 25 v
Source Température Disolvation Température: 120 °C 500 °C
Desolvation Gas Flow; Nitrogen Setting 650 (L/h)
Cône Gas Flow: Nitrogen Setting 50 (L/h)
For Example 27 the following chromatographie conditions and instrumentation were used:
HPLC System: Waters Alliance Z DA- und MS-Detector
Column: Waters XBridge Cl8,4.6 x 30 mm, 3.5 pm
Gradient Program: Time % Sol %Sol Flow [ml/min] Temp [°C]
[min] [H2O,0.1%TFA] [Methanol]
0.0 95 5 4 60
1.6 0 100 4 60
1.85 0 100 4 60
1.9 95 5 4 60
SFC séparation and charactcrization of compounds were carried out under the following method.
Method A:
Instrument: Thar SFC 80; Column: AD 250mm*30mm, Sum; Mobile phase: A: Supercritîcal CO2, B: IPA 5 (0.05% DEA), A: B =80:20 at 60ml/min; Column Temp: 38 °C; Nozzle Pressure: 100 Bar; Nozzle Temp:
C; EvaporatorTemp: 20 ’C; Trimmer Temp: 25 ’C; Wavelength: 220nm.
Method B:
Instrument: SFC MG2; Column: OJ 250mm*30mm, Sum; Mobile phase: A: Supercritîcal CO2, B:
MeOH (0.05% DEA), A:B =90:10 at 70ml/min; Column Temp: 38 ’C; Nozzle Pressure: 100 Bar Nozzle 10 Temp: 60 ’C; Evaporator Temp: 20 BC; Trimmer Temp: 25 ’C; Wavelength: 220nm
The following techniques, solvents and reagents that may be referred by their following abbreviations:
Abbrevfatlon Mcanlng
ACN acetonitrile
B oc tirt-butoxy carbonyl or f-butoxy carbonyl
brine saturated aqueous NaCl
DCM methyl ene chloride
DIEA diisopropyl ethyl amine
DMF di methyl formamide
DMSO dimethyl sulfoxide
dppf 1, l-bis(diphenylphosphino)feiroccnc
EDCI [ -(3-d i methy laminopropy l)-3-e thy Icarbodi i i mi de hydrochloride
EtI ethyl iodide
Et ethyl
Et2O ethyl ether
EtOAc ethyl acetate
EtOH éthanol
Abbreviation Meaning
HPLC high performance liquid chromatography
LDA lithium diisopropylamide
MeOH methanol
Mel methyl Iodide
Me methyl
MeâS dimethyl sulfide
MsO methane sulfonyl chloride
NaOMe sodium methoxide
PdChdppf [ 1, l-bisidiphenylphosphinoiferrocene] dichloropalladium(II)
Pd2(dba)j tris(dibenzylideneacetone)dipal!adium(0)
PE petroleum ether
ri room température
SFC super crilical fluid chromatography
r-BuOK potassium tert butoxide
/-BuLi tert butyl lithium
r-BuNHrBH3 tert butylamin-borane complex
/-BuOOH tert butyl peroxide
TFA trifluoroacetic acid
TF AA trifluoroocetic acid anhydride
THF tetrahydrofuran
TLC thîn layer chromatography
Ti(OEt)4 titanium tetraethoxide
Exemple 1
140 °C
Step 1; Synthesis of intermediate 3
IBUOK.THF 0 methyl acrylate Br^xX^U DMF, CHjCHar t 2
A mixture of compound 1 (50.0 g, 236 mmol) and methyl acrylate (42.0 g, 472 mmol) in anhydrous THF (900 mL) was pre-cooled at 0°C and z-BuOK (31.8 g, 284 mmol, l. I eq) was added in equal portion over 30 mtn, the mixture was then warmed up to rt over l h and was stirred for 40 min at rt. DMF (200mL) and EtI (74 g, 472 mmol) were added to this reaction mixture, and stirred at rt ovemight. THF was removed under reduced pressure. The residue was diluted with HjO (300 mL) and extracted with EtOAc, concentrated to nfTord the crude compound 2 (120.0 g). This product was used as is for next step.
A mixture of compound 2 (120.0 g, 310 mmol) and LîCI (130.0 g, 3100 mmol) in DMSO (900 mL) was refluxed ovemight. The mixture was quenched with water (3L) and extracted with EtOAc (3 x 400 mL). The separated organic phase was dried nnd concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether: EtOAc = 20:1) to give intermediate 3 (15 g, 20%).
'H NMR: (CDClj): δ 7.91 (s, IH), 7.74 (dd, 8.0 Hz, I H), 7.41 (d, J » 8.0 Hz. I H), 3.80 (s, 2H), 2.482.53 (m, 2H), 2.33-2.49 (m, IH), 2.15-2.23 (m, IH), 1.75-1.95 (m,4H), 1.2l-1.40(m, IH),0.88(t, J = 8.0 Hz, 3H).
To a mixture of THF (20 mL) and MeOH (5 mL) at -78°C was added intermediate 3 (6.0 g, 18.7 mmol), NaBH4 (355 mg, 9.3 mmol) and CeCljJHjO (70 mg, 0.19 mmol). The mixture was stirred at -78C for 20 min, quenched with satd. NHjCl solution (30 mL), and extracted with EtOAc (400 mL X 4). The EtOAc phases were combined nnd concentrated to nfford a crude compound 4 (6.5 g, crude).
To a mixture of compound 4 (6.5 g, 20.0 mmol) and NaH (3.2 g, 80,0 mmol) in DMF (100 mL) at 0°C was added Mel (11.4 g, 80.0 mmol). The mixture was stirred nt rt ovemight. The mixture was quenched with HjO, extracted with EtOAc, concentrated to afford the crude product, which was purified by column on silica gel (eluent: petroleum ether: ethyl acetate from 20: 1 to 15: I ) to afford intermediate 5(33 g, 56%).
LC-MS: tR = 1315 min, MS (ESI) m/z 339.1 [M+Hf.
'H NMR: (CDCh): 5 7.88 (s, I H), 7.69 (dd, 7= 8A 2.0 Hz, IH), 7.31 (d, J = 8.4 Hz, I H), 339 (s, 3H),
2.97 (s.2H),2.88-2.94 (m, 1 H), 221-226 (m, |H), 1.81-1.87 (m, IH), 1.70-1,78 (m, IH), 1.40-139 (m,
4H), 1.12-1.39 (m, 2H), 0.88 (U= 8.0 Hz, 3H).
Step 3t Synthesîs of intermediate 6A & 6B
The mixture of intermediate 5 (33 g, 10.4 mmol) and titanium (IV) ethoxide (23.7 g, 104 mmol) in dry THF (40 mL) was stirred at rt for I h. (S)-AT-rert-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 then cooled and water (400 mL) was added and filtered. The aqueous layer was extracted with EtOAc (3 x 400 mL). The separated organic phases were combined and dried and concentrated under reduced pressure.
The residue was purified by column chromatography on silica gel (pctroleum ether EtOAc = 20:1) and compounds eluted in the foliowing order (o give intermediate 6A (13 g, 33%) and 6D (13 g, 33%).
Step 4: Synthesîs of intermediate 7B
To a mixture of ethoxy-ethene ( 13 g, 17.0 mmol) in anhydrous THF (20 mL) at -78 °C under a Nj atmosphère, r-BuLI (13.0 mL, 17.0 mmol, 13 M ln hexane) was added drop wise and stirred for 20 min. The resulting mixture was then stirred at 0 °C for an additionul 45 min. To this mixture at -78 °C,
Intermediate 6B (13 g, 3.4 mmol) was added drop wise in anhydrous THF (20 mL) and stirred for23 h. The reaction was quenched with sat. NHjCI (50 mL) and extracted with EtOAc (3 x 300 mL). The organic phases were combined and concentrated to give the residue and which was purified by column on silica gel (petroleum ether EtOAc = 20: 1) to afford intermediate 7B ( 1.2 g, 69%).
Step Si Synthesis of intermediate SB
Intermediate 7B ( 1.2 g. 2.4 mmol) was added to DCM: MeOH (5:1.20 mL), the mixture was chilled to 78 “C and ozone was bubbled through the mixture for 20 min. The mixture was then purged with Ni and 5 treated with MejS at -78 °C. The reaction was then allowed to warm to ri and stirred for 3 h. The solvent was removed under vacuum, the residue was purified by préparativeTLC (petroleum ether EtOAc = 3:
1) to give compound 8D (860 mg, 70%).
LC-MS: tR » 1.351 min, MS (ESI) m/z 516.1 [M+H]*Aep 6: Synthesis oflntermediate 9B
To compound 8B(860 mg. 1.7 mmol) in MeOH (10 mL) was added a 4 M HCl solution in dioxane (2 mL). The resulting mixture was stirred for 30 min. Solvent was removed under reduced pressure to afford crude compound 9B (800 mg). The residue was used for next step without further purification.
Step 7: Synthesis oflntermediate 10B
A suspension of intermediate 9B (500 mg. 1.9 mmol). Zn(CN)2 (300 mg. 2.6 mmol), Pd^dbafo (150 mg, 0.16 mmol), dppf ( 160 mg, 0.32 mmol) and Zn dust (60 mg, 0.9 mmol) In DMF (15 mL) was heated under 120 C for 3 h in CEM microwave reactor. The mixture was concentrated under vacuum and the residue was purifiedby column on silica gel (eluent: petroleum ether. EtOAc from 20*. I to 8*. I) to afford 20 compound 10B (150 mg, 40%).
Step 8: Synthesis of intermediate 11B
OEt •'OCHj
NaHCOj
11B
10B
Intermediate 10B (150 mg, 0.42 mmol) was added to DCM (10 mL), HjO (10 mL) and NaHCOj (350 mg, 4.2 mmol). To this mixture was added thiophosgene (100 mg, 0.S4 mmol) under vigorous stirring, and stirred for 50 min at rt and extracted with DCM (3 x 40 mL). The organic layer was washed with brine (2 x 40 mL), dried and solvent was removed under reduced pressure to afford crude compound 11B (150 g, 93%), which was used for next step without further purification.
Step 9: Synthesls of intermediate 12B
11B «B
To a mixture of compound 11B ( 150 mg, 0.39 mmol) in THF (5 mL) was added 2amînomethylpyrimidine (67 mg, 0.78 mmol) and TEA (395 mg, 3.90 mmol). The mixture was stirred ovemight at rt. The réaction was diluted with water and extracted with EtOAc (30 mL). The residue was purified by column chromatography (petroleum ether: ethyl acetate s |0:1) to afford 12B (100 mg, 70%). LC-MS: tR = 1.204 min MS (ES1) m/z 462.2 [M+H]*.
Step I: Synthesls of Example 1
12B
Compound 12B (100 mg, 0.22 mmol) In MeOH (10 mL) and NH»OH (3 mL) was added followed by tBuO2H ( 1 mL). After the addition, the mixture was stirred at rt for 24 h. To lhe mixture was quenched with saturated NaiSiOj (0.5 mL) solution. The residue was partitioned between EtOAc (20 mL) and H2O (10 mL). The organic layer was separaied and wnshed with brine (10 mL), dried, filtered and concentrated under vacuum. The residue was purified by HPLC (method 1) to give compound Example 1( 14.60 mg. 15%).
LC-MS: tn = 0.933 min, MS (ESI) m/z 445.2 [M+H]*.
'HNMR: (CDjOD):58.74(d,Jn5.2Hz,2H), 7.6i (dd,7=7.6, 1.6 Hz, 1 H),7.52 (s, IH), 7.45 (d,7 = 8.0Hz, IH), 7.35 (1,7 = 5.2 Hz, IH), 4.94 (s,2H). 338 (s, 3H), 3.17 (s, 2H), 2.80-2.87 (m, IH),2.08-2.13 (m, IH), 1.90-1.94 (m. IH), 138-1.85 (m,2H). 1.22-139 (m,3H), 1.12-1.18 (m,2H), 0.76(1,7=8.0 Hz, 3 H).
Example2
N «
This compound was synthesized from intermediate 10B from Example 1 os shown In scheme below.
N.
10B
Example 2
Step 1: Synthesis of intermediate 13
To a stirred solution of thiourea (23 g, 302 mmol) in THF (5.0 L) under argon at 0 °C were added sodium hydride (29.9 g, 755 mmol, 60% în minerai oil). After 5 min, the ice bath was removed, and the reaction mixture was stirred at room température for 10 min. The mixture was cooled to 0 °C again, di-tert-butyl dicarbonate (138 g, 635 mmol) was added, and the ice bath was removed after 30 min of stirring at that température. The resulting slurry was stirred for another 2 h at rt. Then the reaction was quenched with an aqueous solution of saturated NaHCOj (500 mL). The reaction mixture was poured into water (5.0 L) and extracted with EtOAc (3 x 2.0 L). The combined organic layer was dried, filtered, and concentrated in vacuo to give intermediate 13 (80 g, 96%) as a white solid, which was used for next step without further purification.
To a mixture of intermediate 13 (4.14 g, 15.0 mmol) and anhydrous THF (300 mL) was added NaH (60% in minerai oil, 720 mg, 18.0 mmol) at 0 C. The reaction mixture was stirred at 0 °C for 1 h, then TFAA (3.47 g/233 mL, 165 mmol) was added and the stirring continued for an additions! I h. Then, 4(aminomethyl)tetrahydropyran (25 g, 16.5 mmol) and EtjN (3.03 g/4.16 mL, 30.0 mmol) in anhydrous THF (130 mL) was added and the resulting réaction was stirred at rt ovemight. HjO (150 mL) was added to quench the reaction and the mixture was extracted with EtOAc (3 x 200 mL). The combined organic layers were dried, and the solvent was removed under reduced pressure. The residue was purified by flash column chromatography to afford compound 13 (354 g, 86%) os a white solid.
LCMS·. CR = 0.973 min; MS (ES!) m/z 219 [M-t-Buf.
Step 2: Synthesis of intermediate 14
To a mixture of compound 10B (25 g, 7.0 mmol) in 30 mL of DMF was added compound 13 (2.3 g, 8.4 mmol), EDCI (2.5 g, 14.0 mmol) and DŒA (1.7 g, 14.0 mmol). The mixture was stirred at rt ovemight. It was extracted with EtOAc (3 x 80 mL), washed with brine (3 x 50 mL), dried and the solvent was
removed under reduced pressure. The residue was purified by column chromatography (petroleum ether. ethyl acetate = 5:1) to afFord 14 (2.7 g, 75%).
LC-MS: tR = 0.972 min, MS (ESI) m/z 495.3 [M-t-Buf.
Step 3: Synthesis of Example 2
To a mixture of intermediate 14 (2.7 g. 4.9 mmol) in DCM (30 mL) was added TFA (6 mL). After the addition, the mixture was stirred at rt for 1 h. The reaction mixture was adjusted by NaHCOj solution to pH 8.0-9.0. The organic layer was concentrated under reduced pressure. The residue was purified by column chromatography (petroleum ether: ethyl acetate = 1:1 ) to give compound Example 2 (1.83 g,
83%) as a white solid.
LC-MS: tR » 0.897 min, MS (ES1) m/z 451.2 [M+H] *.
’H-NMR: (CDjOD): δ 7.66 (dd, J= 8.0,1.6 Hz, I H), 7.51 (d, J = 7.6 Hz, IH), 7.33 (s, i H), 3.92-3.98 (m, 2H), 3.37-3.43 (m, 7H), 3.20 (m, 2H),2.78-2.83(m, IH),2.16-2.20(m, IH), l.87-2.03(m, IH), 1.7115 1.77 (m,lH), 138-1.62 (m, IH), 1.51-1.54 (m,2H), 1.28-1.37 (m, 7H), 1.09-1.10 (m. IH), 0.76 (t, J = 7.6
Hz, 3H).
Exampie 3
Synthesis of intermediate 18 25
fl MeMgBr OH DAST.CHjCIj >L,NHBoc ' ' -70 °C-r.t F ^A^NHBoc HCI-dloxane f ------► ^X^NHjHCI
th” ►
19 18 17 16
Step 1: Synthesis Intermediate 16
Mixture of compound 15 (2.0 g, 10.6 mmol) in anhydrous THF (20 mL) was added to a solution of methyl magnésium bromide (14 mL, 42 mmol, 3.0 M in EhO) at -30 “C undera nitrogen atmosphère. The mixture was stirred at -30 °C for 4 h, and then quenched by addition of water (40 mL) and aq. HCl (50 mL, I M) with stirring at 0 °C. The mixture was separated, and the aqueous layer was extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with brine (2 x 50 mL), dried, fiitered and concentrated under vacuum to give the crade intermediate 16 (2.1 g, 100% crade) os a colorless oil, which was used dircctly in next step without purification.
'H NMR: (CDCIj): δ4.97 (br, 1 H), 3.10 (s, 2H), 2.17 (br, IH), 1.44 (s. 9H), 1.20(s,6H).
Step 2: Synthesis of Intermediate 17
To a mixture of intermediate 16 (3.0 g, 15.9 mmol, crade) In anhydrous DCM (50 mL) was added DAST (2.3 mL, 17.4 mmol) at -78 °C under a nitrogen atmosphère. The mixture was stirred at -78 *C for 1 h, and allowed to warm to rt ovemight The mixture was then cooled to 0 °C, and quenched by addition of saturated aqueous layer NoHCOj (30 mL) with stirring at 0 °C slowly. The mixture was separuted, and 10 the aqueous layer was extracted with DCM (2x 20 mL). The combined organic layers were washed with brine (2 x 30 mL), dried, filtered and concentrated under vacuum to give the crade Intermediate 17 (2.5 g. 76% crade), which was used dircctly in next step without purification.
'HNMR: (CDCIj): 54.82 (br, IH). 3.30-3.35 (d, 6.0 Hz, IH), 324-326 (d, 6.0 Hz, IH), 1.44 (s,
9H). 1.37 (s, 3H), 1.35 (s,3H).
**F NMR: (CDCIj 400 MHz): δ -144.93.
Step 3,· Synthesis of Intermediate 18
To a mixture of intermediate 17 (2.0 g, 10.5 mmol, crade) in anhydrous DCM (10 mL) was added a mixture of HCI-dioxane (10 mL, 40 mmol, 4 M in dioxane) with stirring. The mixture was stirred at rt for 20 2 h after which time the solvent was concentrated under vacuum. The residue was washed with a mixture of DCM: petroleum ether ( 1:1 ) (3 x 10 mL), and the precipitate was collected and dried under vacuum to give the crude compound 18 (1.1 g), which was used dircctly in the next step without purification.
Ή NMR: (CDjOD): δ 3.15-3.25 (d, J = 20.0 Hz, 2H), 1.51 (s, 3H), 1.48 (s, 3H). ”F NMR: (CDCIj 400 M Hz); 5-147.59.
Exampie 3
N.
Example 3 was synthesized from intermediate 11B from Example 1 following the same procedure as in Example 1 and utilizing intermediate 18 in step 9 of Example 1.
LC-MS: tR = 1.12 min, MS (ES1) m/z 427 [M+H]*.
’H-NMR: (CDjOD) 8 7.65 (dd. I H, J = 8.2 Hz). 731 (d, I H, J = 8 Hz). 7.31 (s, 1 H). 3.72 (dd. 2H, J = 22.
Hz). 3.37 (s, 3H), 3.20 (ap q, 2H, J = 16Hz), 2.82 (m. IH), 2.18 (m, IH). l.90(m.lH). |.79-l.70(m. IH), 132-11.22 (m. Ι0Η). 121-1.09 (m. IH). 0.77 (t.3H,J = 7 Hz).
O
CICHjCN
Synthesis of intermediate 25
NaN3 NaHCOa
Pd/C ---> h2
Step 1: Synthesis of intermediate 20
A mixture of dihydro-2H-pyran-4(3H)-one (19,50.0 g, 500 mmol) and 2-chloroacetonitrile (35.0 g, 350 mmol) in tert-butanol (50 mL) was stirred for 30 min. To this mixture was added a solution of f-BuOK (60 g, 550 mmol) in tert-butanol (500 mL) over 40 min. The reaction mixture was stirred at rt for 16 h. It was diluted with water and quenched with 10% HCl. The réaction mixture was concentrated to one-third of its original volume, and extracted with diethyl ether four times. The combined organic layers were washed with brine, dried over MgSQi, filtered, and concentrated to afford intermediate 20 (57 g), which was used directly in next step without purification.
Step 2: Synthesis of intermediate 21
Intermediate 20 (57 g) was mîxed with dichioromethane (200 mL) in a polypropylene bottle. The bottic was cooled to 0 °C and 70% hydrogen fluoride-pyridine (50 mL) was added siowly. The mixture was aliowed to warm to room température ovemight. The reaction mixture was diluted with ethyl acetate (500 mL) and poured into saturated aqueous NaHCOj. Additional soiid NaHCOj was used to neutralize the mixture carefuily untii bubblîng ccased. The organic layer was Isoiated, and the aqueous layer was extracted with ethyl acetate (3 x 500 mL). The combined organic layers were washed with 1% aqueous HCl solution, brine, dried (MgSOJ, filtered and concentrated to give crude intermediate 21 (54 g), which was used directly in the next step without purification.
'H NMR: (CDCIj): δ 4.37 (m, 2H), 3.96-2.70 (m, 4H), 1.97 - 1.81 (m, 4H).
Step 3: Synthesis ofintermediate 22
Το a mixture of întermediate 21 (54 g; 340 mmol) in 2-propanol (1000 mL) and water (250 mL) ut 0 °C was added sodium borohydride (20 g, 509 mmol). The mixture was stirred and allowed to warm to rt □ver 3 h. The reaction was quenched with acetone, and stirred for another l h. The clear liquid was separated from solid by decantîng. Additional EtOAc was used to wash the solid, and was decanted. The combined organic solution was concentrated. The residue was purified with flash column chromatography on silica gel eluting with 5-20% EtOAc în hexancs to give intermediate 22 (22 g, 40% for 3 steps) as a liquid.
'H NMR: (CDC13): 5:3.82-3.77 (m, 4H). 3.72-332 (dd, J = 20.8,6.4 Hz. 2H), 2.69(s, 1 H), 1.82-1.60 (m, 4H).
Step 4: Synthesis of intermediate 23
To a mixture of intermediate 22 (20 g, 150 mmol) and triethylamine (22.7 g, 225 mmol) in DCM (200 mL) was added MsCI (25.8 g, 225 mmol) at 0 °C. The mixture was stirred at rt for 2 h, and then water was added. The aqueous layer was extracted with DCM (2 x 200 mL). The solvent was dried and removed to afford crude intermediate 23 (30 g, 100%). whîch was used for the next step without further purification.
’H NMR: (CDCIj): 5:4.22 (d, J»20.0 Hz, 2H), 3.87-3,82 (m,4H),3.06(s,3H)t 1.88-1.68 (m,4H).
Step S: Synthesis of intermediate 24
To a mixture of intermediate 23 ( 10 g, 47 mmol) with DMF (150 mL) was added NaNj (16 g. 250 mmol) and NaHCOj (93 mg. 100mmol)at 120°C. Themixture wasstirredat 120°Cfor20h,thereaction quenched with water. extracted with EtOAc (2 x 300 mL). The solvent was dried and removed under vacuum to afford crude intermediate 24 (8 g), which was used for the next step without further purification.
Step 6: Synthesis of intermediate 25
To a mixture of intermediate 24 (8 g, 50 mmol) in ethyl acetate (100 mL) was added Pd/C (0.8 g. 10% content) under a nitrogen atmosphère, the mixture was degassed and exchanged with hydrogen for 3 times. The final mixture was stirred at room température under 1 atm. hydrogen atmosphère for 24 h. The catalyst was filtered ofT through a pad of Celite* and washed with EtOAc (2 x 50 mL). The combined filtrate was concentrated under reduced pressure to give intermediate 25 (5.3 g, 80%). *H
NMR: (CDjOD): δ 3.83-3.79 (m, 4H), 2.76-2.71 (d, 8.0 Hz, 2H), 1.83-1.65 (m,4H).
”F NMR: (CDjOD, 400 MHz ) δ: -169.66.
Example 4 was synthesized from intermediate 11B following the same procedure as described for Example 1 utilizing intermediate 25 instead of 2-pyrimidylmethonamine in siep 9.
LC-MS: tR = 0.98 min, MS (ES1) m/z 469 [M+HJ*.
Ή-NMR: (CDjOD) δ 7.64 (d, 1 H, J » 8 Hz), 7250 (d, 1 H, J = 8 Hz). 7.31 (s, 1 H), 3.84-3.65 (m, 6H), 3.36 (s.3H),3.19(apq,2H,J = 16 Hz),2.81 (m, lH),2.l7(m, IH), 1.89-1.66 (m,6H), 130-137 (m,3H), 1.34 (m, 2H), 1.20-1.11 (m. |H).0.76(t,3H,J = 8Hz).
Example 5
Toa mixture of ethoxyethene (1.3 g, 17.0 mmol) in anhydrous THF (20mL)at-78eCunderaNï atmosphère was added dropwise i-BuLi (13.0 mL, 17.0 mmol, 1.3 M in hexane) and the mixture stirred for 20 min. The resulting mixture was then stirred at 0 °C for another 45 min and compound 6A (13 g,
3.4 mmol) in anhydrous THF (20 mL) was added and stirred for 23 h. The reaction was quenched with 25 sat. NH4CI (50 mL) and extracted with EtOAc (3 x 300 mL). The organic phases were combined and concentrated to give a crude product. It was purified by column on silica gel (petroleum ether: EtOAc» 20: I) to afford compound 7A ( 1.2 g, 69%) which was used as is for the next step.
Step 5: Synthesis of Intermediate &4
A mixture of compound 7A (1.2 g, 2.4 mmol) in DCM: MeOH = 5:1 (20 mL), was chilled to -78 °C and ozone was bubbled through the mixture for 20 min. The mixture was purged with Na and treated with Me2S (5 mL) at -78 “C, then allowed to warm to rt and stirred for 3 h. The solvent was removed under vacuum, the residue was purified by préparative TLC (petroleum ether: EtOAc» 3:1 ) to give compound 8A (860 mg, 70%). LC-MS: tR = 1.333 min; MS (ESI) m/z 516.1 [M+HJ*.
Step 6: Synthesis of intermediate 9A
To a mixture of compound 8A (860 mg, 1.7 mmol) in MeOH (iO mL) was added a 4 M HCl solution in dioxane (2 mL). The resulting mixture was stirred for 30 min at rt Solvent was removed under reduced pressure to afford crude compound 9A (800 mg) which was used for the next step without further purification. LC-MS: tR » 0.976 min; MS (ESI) m/z 361.1 [M+H]+.
Step7: Synthesis of intermediate IDA
A mixture of compound 9A(500 mg, 1.9 mmol), Zn(CN)2(3OO mg, 2.6 mmol), Pd2(dba)j (i50 mg, 0.16 mmol), dppf (160 mg, 032 mmol) and Zn dust (60 mg. 0.9 mmol) in DMF (15 mL) was heated to 120 “C for 3 h in CEM microwave reactor. The mixture was concentrated under vacuum and the residue was purified by column on silica gel (eluent: petroleum ether: EtOAcfrom 20: I to 8: I) to afford compound 10A (300 mg, 40%). LC-MS: tR = 0.880; MS (ESI) m/z 308.1 [M+H] t
Step 8î Synthesis of intermediate 11A
csct, NaHCOj
To a mixture of 10A (300 mg, 0.84 mmol) in DCM (10 mL), HjO (10 mL) and NoHCOj (655 mg, 8.4 mmol) was added thiophosgene (180 mg, 1.68 mmol). The mixture was stirred for 50 min, then extracted with DCM (3 x 40 mL), washed with brine (2 x 40 mL), dried and the solvent was removed under reduced pressure to afford crude compound 11A (300 g,), which was used for the next step without further purification.
To compound 11A (200 mg, 030 mmol) in THF (10 mL) was added to Λ(2aminomethyljtetrahydrofuran (61 mg, 0.6 mmol) and triethylamine (2 mL. 5.0 mmol). The mixture was stirred at rt ovemight. The reaction was diluted with water and extracted with EtOAc (30 mL). The residue was purified by column chromatography (petroleum ether: ElOAc= 10; |) to afford 12A ( 180 ntg, 79%).
A mixture of intermediate 12A (250 mg. 0.54 mmol) in MeOH ( 10 mL) and NH4OH (3 mL) was added a S solution of r-BuOjH ( ! mL, 9M in hexane) and stirred at rt for 24 h. The reaction was quenched by saturated NaîSiOj(0.5 mL). The residue was partîtioned between EtOAc (20 mL) and HîO (tO mL). The organic layer was separated and washed with brine (10 mL), dried, filtered and concentrated under vacuum. The residue was purified by HPLC (method 1) to give Example 5 (89.10 mg, 52%).
LC-MS: tR = 0.971 min, MS (ESI) m/z 437.2 [M+H]*.
Ή NMR: (CDjOD): fi 7.60 (dd. 8.0. 1.6 Hz, tH), 746 (d, J= 7.6 Hz, 1 H), 7.28 (s, 1 H), 4.08-4.01 (m, IH), 3.63-3.90 (m, 4H). 3.33 (s, 3H), 3.09-3.20 (m, 2H), 2.74-2.79 (m. IH), 1.80-2.06 (m. 5H), 1.65-1.78 (m, IH). 1.55-1.64 (m, 2H), 1.29-1.35 (m, 3H), 1.07-1.29 (m, IH). 0.89-0.96 (m, IH), 0.85 (t, 7.6 Hz,
3H).
0 tQuOK,THF O O
Λ methyf acrylato Br». Λ UCI. OMSO_ Brx
1 0 J DMF, CHJ I coc 140 °C I coc
1 26 I COîMe 27 \
An oven dried 3 L flask charged with of 6-bromo- !-indanone (100 g, 473.8 mmoi), methyl acryiate (86.4 g, 90 mL, 995 mmol, 2.1 eq) and anhydrous THF (800 mL), the flask was immersed in an ice-water cooling bath and stirred. Initially, tBuOK (0.5 g) was added carefully, after 2 min, second portion of tBuOK (0.5 g) was added. The cooling bath was removed and remaining tBuOK (63 g) was added in even portions over 20 min (total 64 g, 568.6 mmol, 1.2 eq). The mixture was stirred for another 2 h at rt. DMF (240 mL) was added to the reaction mixture, followed by Met (134.6 g, 60 mL, 947.6 mmol, 2.0 eq) and the mixture was stirred for another 2 h. The reaction was quenched with 10% citric acid solution. Then the reaction mixture was concentrated under reduced pressure to remove most solvent before it was filtered. The cake was washed with water, followed by MeOH to give the crude intermediate 26 (200 g) which was used in the next step dircctly.
To a solution of compound 26 (200 g, 547.6 mmol, crude) In THF/HjO (1.8 L/l .8 L) was added LiOH.HjO (92 g, 2190 mmol, 4.0 eq). The mixture was stirred for 16 h at rt and then 12 h at 70°C. The reaction mixture was concentrated under reduced procedure to remove THFand filtered. The cake was washed with HjO, and then it was stirred with MeOH (50 mL) for a few min and filtered again, and washed with additional amount of MeOH (50 mL). The solid was collected to give intermediate 27 (75 g. 51.7%),
Step 2: Synthesis of intermediate 29
A three neck flask was charged with CeC!j.7H2O (1.2 g, 3.3 mmol) and anhydrous MeOH (60 mL) under a nitrogen atmosphère and stirred to yield dear solution. Compound 27 (10.0 g, 32.6 mmoi) and anhydrous THF (240 mL) were added under nitrogen atmosphère, the mixture was cooled down to -78 °C. NaBH» (0.4 g, 13.0 mmol) was added at -78 ’C under a nitrogen atmosphère with vigorous stirring. The mixture was stirred at -78 ’C for 20 min. The reaction mixture was quenched by addition of saturated aqueous NH4CI ( 100 mL) and H2O (200 mL) at -78 ’C with stirring. The mixture was slowly allowed to warm to ambient température. The mixture was extracted with EtOAc (3 x 150 mL). The combined organic layers were washed with HjO (2 x 200 mL), brine (2 x 200 mL), dried, filtered and concentrated under vacuum, the residue was purified by column chromatography on silica gel eluting with petroleum ether EtOAc (20: 1 to 3:1) to give intermediate 28 (7.5 g, 75%). LC-Ms: tR = 3.195 min: MS (ESI) mlz 311.0 [M+HJ*.
‘H NMR: (CDOj): <57.59 (s. IH), 722-7.25 (d, J = 8.4 Hz, IH), 7.08 (s, IH),6.88-6.91 (dd, J= 2.4, 8.4 Hz, iH). 6.80-6.81 (d, J » 2.4 Hz, IH), 5.84 (s, IH), 4.87 (s, 2H), 4.31-4.36 (m, 2H), 3-50-3.55 (q. J = 6.8
Hz, 2H), 3.15-3.25 (m, 1H), 3.09-3.14 (d, J = 15.6 Hz, 1 H), 3.00-3.06 (d, J= 15.2 Hz, 1H), 1.90-2.10 (m, 3H), 1.25-1 JO (m, 5H), 1.15-1.25 (I, J = 6.4Hz,3H).
O 0
Bra OX l NaH, CH3I Br-. y.iOH-------* Ί / DMF 1 CÔC Y'OCHa
28 29
To a mixture of compound 28 (6.18 g, 20 mmol) in DMF (20 mL) was added NaH (60% in minerai oil, 0.96 g, 40 mmol) at 0 °C. Then the mixture was stirred at 0 °C for 2 h, then Mel (3 J mL) was added to the mixture and stirred ovemight. The mixture was diluted with EtOAc (40 mL) and HjO (40 mL), extracted with EtOAc(2 x 60 mL). The combined organic phases were dried and the solvent was removed lo give intermediate 29 (5.0 g).
Step 2: Synthesis of intermediate 30A & 30B
To a solution of intermediate 29 (5.0 g, 15.3 mmol) in THF (100 mL) was added T1(0Et)4 (35.0 g, 153 mmol). After being stirred at rt for 1 h, (SJ-W-ren-butylsulfinamide (7.4 g, 61.2 mmol) was added. The reaction mixture was stirred at reflux ovemight and the mixture was partîtioned between H2O (80 mL) and EtOAc (80 mL), The mixture was filtered and the filtrate was extracted with EtOAc (3 x 80 mL). The combined organic layers were washed with brine (50 mL), dried and concentrated to the residue. The residue was purified by column chromatography on silica gel (petroleum ethen EtOAc = 20:1) eluted In the following order to give intermediate 30A ( 1.6 g, 35%) and 30B ( 1.4 g, 33%).
Synthesis of Example 6
Intermediate 30A was further elaborated as illustrated in steps 4 through 10 in Example 5. In step 9,2aminomethylpyrimïdine was used instead of/f-(2-aminomelhyl)tetnihydrofuran.
LC-MS: tR = 1.05 MS (ESI) m/z431.4 [M+H]*.
lH NMR: (CDjOD): 6 8.78 (d, J» 4.8 Hz, 2H), 7.76 (s, 1 H), 7.75 (dd, J= 6.0,1.6 Hz, 1 H). 736 (d, J=
8.4 Hz, 1 H) 7.44 (t, 5.2 Hz 1H), 5.16 (m. 2H). 3.38 (s, 3H), 3.24 (m, 2H), 2.79 (m, IH). 2.15 (m. IH).
1.74 (m, IH). 1.65 (d, J= 6.8 Hz, IH), 1.39-1.57 (m, 4H), 0.99 (d, J= 6.4 Hz, 3H).
Example 7 .0
This was synthesized by a procedure similar as described in Example 6. Intermediale 30A was further elaborated as described in Example 1 through steps 4-10. (2-meÜioxy) ethylamine was used in step 9 followed by oxidation as described in step 10 to yield Example 7.
LC-MS: tR = 1.08 min, MS(ESI) m/z 397 [M+HJ*.
Ή NMR: (CDjOD) fi 7.74 (d, 1 H, J = 8 Hz), 7.63 (d, 1 H, J = 1 Hz), 737 (d, 1 H. J = 8 Hz), 4.02-3.95 (m, IH), 3.89-3.83 (m, IH), 334 (m. 2H). 3.36 (s, 3H), 335 (s, 3H), 3.24 (apq, 2H, J - 16 Hz)), 2.75 (m, IH), 2.10(m, IH), 1.79(dt, 1 H, J « 13,2 Hz), l.56(m, IH), 1.41 (m,3H), 1.14«,1H,J« 13 Hz), 1.01 (d, 3H,J«6 Hz).
Exnmple8
This was synthesized by a procedure similar as described in Example 6. (3-methyloxetan-3yljmethanamine was used as described in Example 1 in step 9 followed by oxidation as described in step 10 to yield Example 8.
LC-MS: tR » 0.930 min, MS (ESI) m/z 423.0 [M+H]*.
Ή NMR: (CDjOD): S7.66-7,64(d, J·72 Hz, IH), 731-7.49 (d, J = 7.6 Hz, IH), 7.34 (s, IH),4.724.67 (m, 2H),4.29-4.25(m, 2H), 3.74-3.59(m, 2H), 3.37 (s, 3H), 3.25-3.14 (m, 2H),2.74-2.67 (m, IH), 2.08-2.03 (m. !H), 1.80-133 (m, 3H), 1.30(m,5H), l.08(m, IH), 0.90(m. 3H).
This was synthesized by a procedure similar as described In Example 6.4-(aminomethyl)pyrimidine was used as described in Exampie 1 in step 9, followed by oxidation as described in step 10 in Example 6 to yield Example 9.
LCMS: tR = 0.88 min, MS (ESI) m/z 4312 [M+H]*.
'HNMR: (CDiOD):59.05 (s, IH), 8.70-8.71 (d,Jo5.2Hz, 1 H), 7.60-7.62 (d, 7 = 7.6 Hz, 1H),7A47.47 (m, 3H), 4.86 (s, 2H), 3.35 (s, 3H), 3.10-320 (q, 2H), 2.70-2.71 (m, IH), 2.04-2.06 (m, IH), 1.70 (m, 2H), 1.491 (m, IH), 130-133 (m. 2H), 1.15-1.18 (m, IH), 0.95-0.96(d,7 = 6.0 Hz, 3H).
Example 10
Step I: Synthesis of intermediate 32 □
1) KOtBu, THF
C°Ctort
2) methyl methaerylatp
3) methyl aaylata
4) Met
To a mixture of 6-bromo-indan-l -one (100.00 g, 473.8 mmol) in anhydrous THF (1 L)at 0°C was added r-BuOK (583 g, 521.2 mmol, 1.1 eq), 2 min later the mixture was warmed up 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 onc portion. After 2h, methyl acrylate (49.0 g, 512 mL, 568.6 mmol, 12 eq) was added to the reaction mixture. After3hatrt,Mel(IOl g, 443 mL, 710.7 mmol, 13 eq) wasadded to the reaction mixture, and it was stirred for 16 h. H2O (I L) was added followed by LiOH*HjO (793 g, 18952 mmol, 4.0 eq), the mixture was stirred for 28 h at room température. THF was removed under reduced pressure. The residue was diluted with ΗιΟ (I L) and filtered, washed with HjO until the fiitrate was neutral. The product was washed with MeOH to afford 50 g of intermediate 32.
Step 2: Synthesis of intermediate 33
Το a mixture of intermediate 32 (60.0 g, 186.9 mmol) and FeCij (33.0 g, 205.5 mmol, 1.1 eq) in THF (600 mL) was added NaBHjCN (29.4 g, 367.1 mmol, 2.5 eq) at 0 ’C. The mixture was aliowed to warm to room température and stirred for Ih at rt. The reaction was quenched by addition of water and THF was removed under vacuum. It was extracted with DCM (3 x 200 mL). The combined organic phases were washed with HjO and brine, dried, and concentrated under vacuum to give the crude product, which was purified by column chromatography on silica gei to generale compound 33 (25.2 g, 42%) and 33A (12.0 g).
LC-MS: tR « 1.239 min, MS (ESI) m/z 323.1 [M+H] *.
'H-NMR (CDCIj): δ: 7.889-7.894 (s, IH), 7.671-7.696 (d, IH), 7.311-7.332(d, IH), 3.605 (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 2: Alternative synthesis of intermediate 33
t-BuNHj-BHj FeCij
THF,-10 ’ ’C
A mixture of FeCij (6.0 g, 37.0 mmoi) with toluene (60 mL) was cooled to 0 °C. A mixture of compound 32 (11.9 g, 37.0 mmol) in THF (48 mL) was then added to the mixture. The mixture was stirred for 5 min at 0°C and then cooled to -10 °C. A solution of f-BuNHj-BHj (3.5 g, 40.7 mmol) in THF (12 mL) was added dropwise to the reaction mixture at -10 °C. The reaction mixture was stirred at about -10 °C for 30 min, quenched with 6N aq HCl solution (10 mL), stirred at about 0 ’C for 30 min, and then aliowed to warm to room température. 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 ιΛ volume, heated to 50 ’C to obtain a solution, and then cooled to 0 eC over l h and held at 0 ’C for 1 h. The solid was filtered and washed with cold (0 °C) toluene (12 mL), and dried under vacuum to give compound 33 (9.93 g. 83%).
LC-MS: tR «= 236 min, MS (ESI) m/z 323.0/325.0 [M+H]
Step 3: Synthesis of intermediate 34
Το a mixture of compound 33 (20.0 g, 61.9 mmol) with DMF (200 mL) was added NaH (5.0 g, 123.8 mmol, 2.0 eq) at 0°C. Then it was stirred for 15 min at 0°C and Mel (17.6 g, 123.8 mmol, 2.0 eq) was added at 0°C. Then it was warmed to rt and stirred for 15 h at rt. The mixture was quenched with HjO and extracted with EtOAc. The combined organic phases were washed with H2O and brine. dried, concentrated to afford crude product, which was purified by column on slica gel (eluent: petroleum ether: ElOAcfrom 100/1 to 5/1) to afford intermediate 34 (20 g, 96.2%).
The mixture of compound 34 (20.0 g, 59.3 mmol) and titanium (IV) ethoxide ( 108.2 g, 474.4 mmol) ln dry THF (200 ml) was stirred at rt for 1 h. (S)-/V-/erf-butylsuifinamlde (29 g, 237.2 mmol) was added. The resulting mixture was stirred at 80 °C under Ni atmosphère ovemight. The reaction mixture was then cooled and water (400 ml) was added. The mixture was filtered and the aqueous layer was extracted with EtOAc (3 x 400 mL). The separated organic phase was dried and concentrated under reduced pressure to give crude producL The residue was purified by column chromatography on silica gel (petroleum ether: EtOAc= 20:1) to give intermediate 35 (18.4 g, 70.5%).
Step S: Synthesis of intermediate 36
r-BuLi ( 131 mL, 170.3 mmol, 13 M in hexane) was added dropwise to a solution of ethyl vinyl ether (123 g, 170.3 mmol, 5.0 eq) in anhydrous THF (100 mL) at -78 ’C under Nj and stirred for 20 min. The resulting mixture was then stirred at 0 ’C for another 45 min. The solution was re-cooled to -78 ”C and compound 35 (15.0 g, 34.1 mmol) in anhydrous THF (50 mL) was added dropwise and the mixture was stirred for 2 h at -78 °C. The réaction mixture was quenched with sat NHiCI (50 mL) and extracted with EtOAc (3 x 300 mL). The organic phase was concentrated to give the residue, which was purified by silica gel column chromatography to afford intermediate 36 (11 g, 64.7%) and 36A (1.441 g, 100% purity).
LC-MS tR = 5.676 min; MS (ESI) m/z 514.2 [M+Hf.
’H-NMR (CDjOD): δ 7346 (s, IH), 7.454-7.479 (d, IH), 7208-7.228 (d, IH), 4.6204.755 (d, IH), 4373-4381 (m, I H),4.0484.055 (m, IH), 3.844-3.903 (m, 2H), 3.458-3.474 (s. 3H), 2.986-3.000 (m. 2H),2326-2377(m, IH). 1.969-2.001 (m, IH), 1.671 (s. IH), l.457-I320(t, J» 12 Hz,3H), 1373-1.408 15 (m, 2H). 1328 (s, 9H), 1.169-1278 (m, 5H), l.073-l.I06(d, 3H).
Step S: Synthesîs of intermediate 37
A mixture of intermediate 36 (4.8 g, 937 mmol) in DCM:MeOH « 5:1 (40 mL), was chilled to -78 ’C and ozone was bubbled through the mixture for 20 min. The mixture was then purged with Ni and treated with MeiS (10 mL) at -78 °C, then allowed to warm up to rt and stirred for 3 h. The solvent was removed under vacuum, the residue was purified by column chromatography on silica gei (petrolcum ether: EtOAc= 20:1 to 8:1) to give intermediate 37 (33 g, 72.9%).
LC-MS IR = 1297 min; MS (ESI) m/z 516.1 [M+H]+.
Ή NMR (CDCIj): δ 7.84 (s, IH), 7.42-7.44 (d, J « 8.0 Hz. IH), 7.09-7.11 (d, J = 8.0 Hz, 1 H), 4.40 (s, IH), 4.26-4.39 (m, 2H), 3.44 (s, 3 H), 2.93-2.97 (d, J = 15.6 Hz, IH), 2.70-2.74 (d, J = 15.2 Hz, IH), 2.222.30(t, J = 10.0Hz, IH), 1.75-1.79 (m, IH), 1.61-1.66 (m, IH), 134-1.57 (m»2H),
1.32-1.38 (m,4H), 1.14 (s, 9H), 1.06-1.08 (d, J = 6.0Hz, 3H), 0.89-0.91 (d, J = 6.0 Hz, 3H), 0.67-0.74 (m, IH).
Step 6: Synthesis of intermediate 38
To compound 37 (860 mg, 1.7 mmol) in MeOH (10 mL) was added a 4 M HCl solution in dioxane (2 mL). The resulting mixture was stirred for 30 min. Solvent was removed under reduced pressure to afford crude intermediate 38 (800 mg). The residue was used for lhe next step without further purification.
Alternative synthesls of Intermediate 38
Step 1: Synthesis of intermediate 39
A mixture of intermediate 6 (5.00 g. 11.4 mmoi), diethoxyacetonitriie (33 mL, 24.4 mmoi) and THF (50 mL) was cooled to -7 °C and treated dropwise with LDA (25.0 mL, 45.0 mmol, 1.8M in THF/heptane/ethyl benzene). The mixture was stirred at -7 to -2 °C for 2 h, and then quenched with water (50 mL) and saturated aqueous NH4C1 (25 mL). Hexanes (100 mL) was added, and the layers were separated. The organic layer was washed with water, brine, and was concentrated to give crude intermediate 39 (9.00 g, 139%) which was used directly in lhe next step.
LC-MS: tR = 3.74 min, MS (ESI) m/z 523.2/525.2 [M-OEt+H]*
Step 2: Synthesis of intermediate 38
A mixture of above Intermediate 39 (9.00 g, 11.4 mmol) in EtOH (30 mL) was treated with 6N aqueous HCl (20 mL). The réaction mixture was heated at 15 °C for 24 h and cooled to rt The reaction was extracted with toluene (50 mL), and the aqueous phase was then basîfîed to pH = 8 with 2N aqueous
NaOH (-60 mL). Toluene ( 100 mL) was added, and the loyers were stirred and separated. The organic layer was washed with aqueous NaHCOj and brine and concentrated. Hexanes was added and the solution was concentrated again to give crude intermediate 38 (3.47 g, 74%) which was used directly in the next step. LC-MS: tR = 0.86 min, MS (ESI) m/z 410.2/412.2 [M+H]*
Step 7; Synthesis of intermediate 40
Intermediate 40 was synthesized in an analogous fashion as described in step 7 of intermediate 10A. It was used directly in the next step.
Step 8: Synthesis of intermediate 41
OCH3
Intermediate 41 was synthesized in an analogous fashion as described in step 8 of intermediate 11 A. The crude intermediate 41 was used directly in the next step.
Step 9: Synthesis of intermediate 42
Intermediate 42 was synthesized in an analogous fashion as described in step 9 of intermediate 12Λ. The crude intermediate 42 was used directly in the next step.
Step 10: Synthesis of Example 10
Το a solution of intermediate 42 (400 mg, 0.8 mmol) in EtOH (8 mL) was added NHj-HjO ( l mL) and rerr-butyl bydroperoxide (1 mL). After addition, the mixture was stirred at room température for 12 h. The solvent was removed by évaporation under vacuum. The residue was purified by preparative HPLC method 1 to give Example 6 (65.0 mg, 20% yield).
LC-Ms: tR « 0.945 min, MS (ESl) m/z 463.2 [M+H] *.
’H NMR: (CDjOD) δ 8.65-8.70 (s, 2H), 7.60-7.65 (d, J « 7.2 Hz, I H), 7.45-7.55 (m. 2H), 4.95-5.00 (s, 2H), 3.40-3.45 (s, 3H), 3.10-3.20 (m,2H), 2.30-2.40 (m. IH), 1.70-1.80(m, 3H), 1.45-1.55 (m, 1H), 1.25l .35 (m, 2H), 0.90-1.00 (m, 6H).
”FNMR (400 MHz, MeOD): -141.57
Example 11
Example 11 was synthesized as per procedure described in Example 10. In step 9, (3-methyloxetan-3-yl) methanamine was used instead of (5-fluoropyrimidine)-2-methylamine to yield Example 11 LC-MS: tR = 0.96 min, MS (ESI) m/z 437 [M+Hf.
Ή NMR: (CDjOD): δ 7.75 (dd, J = 7.6,1.6 Hz, 1 H), 7.68 (d, J = 1.6 Hz, I H), 756 (d, J = 7.6 Hz, I H), 4.60 (d, J = 6.4 Hz, 2H), 452 (dd, J = 8.0,6.4 Hz, 2H), 3.97 (d, J = 15.6 Hz, IH), 3,69 (d, J = 15.6 Hz, IH), 3.44 (s. 3H), 325 (m, 2H). 2.44 (t, J = 10.0Hz, IH), 1.81-1.75 (m,2H), 1.67(m, IH), 1.41 (s. 3H), 156 (m, IH), 150-1.21 (m. 2H), 1.07 (d, J « 6.4 Hz, 3 H), 0.98 (d, J = 6.4 Hz, 3 H).
Example 12
Example 12 was synthesized as per procedure described Example 10. In step 9,2-amînooxetane was used instead of (5-fluoropyrimidine)-2-methylamine to yield Examplc 12
LC-MS: tR « 0.91 min, MS (ESI) m/z 409 [M+HJ *.
Ή NMR: (CDjOD): δ 7.75 (dd, J = 7.6,12 Hz, IH). 7.71 (d, J = 12 Hz, I H), 756 (d, J » 7.6 Hz, IH), 528 (m, 1 H), 5.13 (dd, J = 14.0, 6.8 Hz, 2H), 4.87 (dd, J - 8.0, 6.4 Hz, 2H), 3.44 (s, 3H), 326 (m, 2H), 2.43 (t, J = 10.0 Hz, IH), 1.85-1.77 (m, 2H), 1.68 (m, IH), 155-1.18 (m, 3H). 1.03 (d, J = 6.4 Hz,3H), 0.97 (d, J = 6.4 Hz, 3H).
Example 13
Example 13 was synthesized as per procedure described Example 10. In step 9, oxetan-3-ylmethanamine was used instead of (5-fluorpyriminde)-2methylamine to yield Example 13.
LC-MS: tR = 0.904 min; MS (ES!) m/z 4235 [M+HJ*.
’HNMR: (CDjOD): δ7.60-7.62 (d, J = 8.0 Hz, 1 H), 7.45-7.47 (d, J = 8.0 Hz, IH),7.27(s, 1H),4.694.73 (d, J=72 Hz, 2H), 4.44U.49 (d, J= 72 Hz, 2H), 3.85-3.91 (m, IH), 3.74-3.80 (m, IH), 3.42(s, 3H),354-358(m, IH),3.08-322(m,2H),2.32-257(1, J = 10.0 Hz, IH), 1.61-1.71 (m,3H), I58(m, IH), 1.19-1.23 (m. I H), 0.92-0.99 (m,7H).
Example 14 was synthesized as per procedure described Example 10. In step 9, (S)-2-(aminomethyl)tetrahydrofuran was used Instead of (5-fluoropyrimidine)-2-methylamine to yield Example 14. LC-MS; tR = !.02 min, MS (ESD m/z437 [M+H]*.
lH NMR :(CDjOD): δ 7.75 (d, J = 7.6 Hz, 1 H), 733 (d, J » 7.6 Hz, ! H). 7.35 (s, 1 H), 4.16 (m. ! H), 3.96 (m. IH),3.83 (m, !H),3.70(m.2H),330(s,3H),3.30(d, J = 15.6 HzJH), 3.19(d, J » 15.6 Hz. IH), 2.42 (t, J = 10.0 Hz, IH), 2.08-1,95 (m, 3H), 1.88-1.1.63 (m. 4H), 135 (m. IH),l.40-l30 (m,2H), 1.05 (d. J = 6.4 Hz, 3H), 1.01 (d, J = 6.4 Hz, 3H).
Example 15 was synthesized os per procedure described in Example 10. In step 9, (/?)-2-(nmïnomethyl>tetrahydrofuran was used instead of (5-fluoropyrimidine)-2-methylamÎne to yield example 15. LC-MS: tR = 1.02 min, MS (ESD m/z 437 [M+H] *.
Ή NMR: (CDjOD): δ 7.61 (d, J = 8.0 Hz, I H), 7.46 (d, J = 8.0 Hz, l H), 725 (s, 1 H), 4.07 (m lH), 3.88 (m, IH), 3.73 (m, IH), 3.66 (dd, J = 14.8,32 Hz, IH), 337 (dd, J = 14.8,6.8 Hz, 1 H), 342 (s,3H), 3.23 (d, J = 16.0 Hz, IH), 3.10 (d, 16.0 Hz, IH), 2.35 (t, J= 10.4 Hz. IH). 2.01-1.86 (m, 3H), 1.76-130 (m. 4H). 1.44 (t, J » 132 Hz, I H), 124 (m, IH), 1.03 (t, J = 12.8 Hz, 1 H), 0.98 (d, J = 6.4 Hz, 3H), 0.93 (d, J = 6,4 Hz, IH).
Example 16
Example 16 was synlhesîzed as per procedure described in Exampie 10. In step 9,2-(aminomethyi)tetrahydropyran was used instead of (5-fluoropyrimldine)-2-methyiamine to yield Example 16 LC-MS: tR = 0.958 min, MS (ESÎ) m/z 451.3 [M+H]*.
'H NMR (CDjOD): S 7.62-7.65 (d, J = 7.6 Hz, IH). 7.48-7.50 (d, J = 8.0 Hz, IH), 7.30 (s, 1 H), 3.93-3.96 (m, 2H).3.37-3.45 (m,7H).3.23-3.27(d, J = 16.0Hz. lH),3.1l-3.15(d, J= 16.0Hz, IH),235-2.40(t, J = 10.0 Hz, IH). 1.96-2.03 (m, IH), 133-1.80 (m,5H), 133-1.46 (m,4H), 0.92-1.08 (m, 7H).
Exemple 17
Example 17 was synlhesized as per procedure described in Example 10. In step 9, intermediate 18 was used instead of (5-fluoropyrimidine)-2-methylamine to yield Example 17,
LC-MS: tR = 0.969 min, MS (ESÎ) m/z 427.2 [M+H]*.
1H NMR: (CDjOD): δ 7.63-7.65 (d, J = 8.0 Hz. 1 H), 7.49-731 (d, J = 8.0 Hz, 1 H). 730 (s, 1 H). 3.703.76 (m,2H),3.45 (s, 3H). 3.13-3.28 (m,2H), 236-2.41 (t,J= 10.0 Hz, IH), 1.65-1.84 (m,3H), L48131 (m. IH), 137-1.42 (m, 6H), 1.26-133 (m, IH), 1.02-1.09 (m, 1 H).0.92-1.00(m,6H). ”FNMR: (CDjOD): <5-13938.
Example 18
Step 9: Synthesis of intermediate 43
To a solution of compound 41 (I g, 2 JI mmol) in THF (25 mL) was added compound 2-(2aminomethyl)oxetane (262 mg, 3.01 mmol) and triethylamine (760 mg, 733 mmol). The mixture was 5 stirred at rt ovemighL The réaction mixture was diluted with EtOAc (45 mL), followed by saturated aqueous NaHCOj (2 x 35 mL) and brine (2 x 35 mL). Solvent was removed after dried to give crude compound 43 and 43A which was purified under SFC-method A. The diastereomers were separated by a SFC-method B. The desired diastereomer 43 was isolated as a second peak under these conditions. This was further elaboroted as described in step 10 of Exampie 10 to yield Example 18.
LC-MS: tR » 0.863 min, MS (ESI) m/z 423. J [M+HJ*.
'H NMR: (CDjOD):<57.6)-7.63(d,J = 8.0Hz, JH), 7.47-7.49 (d,J = 7.6Hz, JH), 730 (s, IH),4.955.01 (m, JH), 4.65-4.70 (m, IH), 432-438 (m, IH), 3.73-3.85 (m, 2H), 3.43 (s. 3H), 3.22-3.26(d, J = 16.0 Hz, IH), 3.10-3.14 (d, J = 16.4 Hz. 1 H), 2.64-2.72 (m, IH), 233-2.45 (m,2H), 139-1.76 (m,3H), 1.40-1.49 (m, IH), 1.22-127 (m, IH), 0.93-1.07 (m,7H).
Exemple 19
Synthesis of 3-((2-amino)ethyl)oxetane
O
Raney NI ,NHï
CN
MeOH o
Step 1: Synthesis of intermediate 44
To 3-oxanone (0.42 g, 6 mmol) in DCM (20 mL) was added 2-(triphenylphosphorany)idene) acetonitrile ( 1.8 g, 6 mmol) and stirred ovemight at rt. After which time, the solvent was removed under reduced
pressure to afford crude product (260 mg, crude), which was purified by chromatography on silica gel (pctroleum: EtOAc, 3:1) to give intermediate 44 (260 mg, yield 46%) as a white solid.
Step 2: Synthesis of intermediate 45
To a mixture of intermediate 44 (260 mg, 2.74 mmol) in MeOH (10 mL) was added Raney-Ni (100 mg) and stirred at rt under hydrogen atmosphère for 12 h. The mixture was then filtered through a pad of Celite*. The filtrate was concentrated to give intermediate 45 (200 mg, crude).
Example 19
Example 19 was synthesized as per procedure described in Example 10. ln step 9, intermediate 45 was used to yield Example 19.
LCMS; tR = 2.358 min, MS (ESI) m/z 437.3 [M+Hf.
lH NMR; (CDjOD): δ 7.64-7.66 (d, IH), 7.49-751 (d, J = 8.0 Hz, 1H), 731 (s, IH), 4.74-4.79 (d, 2H), 4.40-4.43 (d, 2H), 3.49-352 (m. 2H). 3.45 (s, 3H), 2.95-3.27 (m, 3H), 236-2.41 (m, IH), 1.97-2.05 (m, 2H), 1.21-1.80 (m, 5H), 1.01-1.09 (m,4H), 0.92-0.99 (m, 3H).
O ^X^NHjHCI
Et3N, THF
NC
Το α solution of compound 41 (100 mg, 0.25 mmol) In dry THF (3 mL) was added 2-aminoacetone hydrochloride (41 mg, 0377 mmol) and triethylamine (76 mg, 0.754 mmol). The mixture wns stined ovemight at rt. The reaction was quenched by adding water (3 mL) and extracted with EtOAc (2x5 mL). The combined organic layers were dried and evaporated under vacuum. The crude material was 5 purified by prep-TLC to afford compound 46 (50 mg, 24%).
Step 2: Synthesis of Intermediate 47
To a solution of compound 46 (50 mg, 0.118 mmol) in toluene (3 mL) was added ethylene glycol (0.03 mL) nndp-toluene sulfonic acid (1.1 mg, 0.0068 mmol). The solution was heated to reflux for 2 days.
The reaction mixture wns cooled to rt and brine (3 mL) was added. The mixture was extracted with EtOAc (2x5 mL). The combined organic layers were dried and evaporated under vacuum. The crude material was purified by prep-TLC to afford compound 47 (51 mg, %).
To a mixture of compound 47 (51 mg, 0.113 mmol) in MeOH (2.5 mL) was added aqueous ammonia (0.8 mL), t-BuOOH (2.5 mL). The mixture was stined ovemight at rt. Then sat. Na2S2O3 (2.5 mL) was added to quench the reaction. The mixture was extracted with EtOAc (2x5 mL). The combined organic layers were dried and evaporated under vacuum. The crude material was purified by basic prep-HPLC to afford 20 Example 20 ( 12.8 mg, 25%) ns a white solid.
IH-NMR (CD3OD): δ 7.60 (d, J = 8.1 Hz. IH), 7.50 (d, J> 8.1 Hz, IH), 7.22 (s. 1H), 3.864.01 (m, 4H).
3.64-3.75(m, 2H), 3.42(s,3H), 3.03-3.23 (m,2H), 234(t,IH), 1.61-1.84 (m,3H), 1.43 (s, IH), 1.20l.37(m,4H), 0.89-1.08 (m, 7H).
LC-MS tR = 0.891 min, MS (ESI) m/z 4533 [M+H]*
Exampie 21
Example 21 was synthesized as per procedure described in Example 10. In step 9, (R)-( J,4-dioxan-2y|)methanamlne was used to yield Example 21.
LC-MS: (K = 0.928 min, MS (ESI) m/z 4533 [M+H]*.
'H NMR: (CDjOD): δ 7.63-7.6) (dd, J = 1.6 Hz, 4.0 Hz, IH), 7.48 (d. IH), 73 (s. IH), 3.6-3.8 (m, 6H),
3.6-3.5 (m, 2H), 3.45 (s, 3H). 3.3-3.J (m, 3H), 2.4 (m, IH), 1.8-1.6 (m, 3H), 1.6-I.4 (m, IH), 13-1.2 (m, IH), l.I (m, IH), 0.9-1.01 (m,6H).
Example 22
Ρ-Ά
Example 22 was synthesized as per procedure described in Example 10. In siep 9, (S)-( l,4-dioxan-2y])methanamine was used instead of (5-fluoropyrimidine)-2-methy]amine to yield Example 22. LC-MS: tR 0.928 min, MS (ESI) m/z 4533 [M+H]*.
'H NMR: (CDjOD): δ 7.63-7.61 (dd, J = 1.6 Hz, 4.0 Hz. 1 H). 748 (d, IH). 73 (s. IH), 3.6-3.8 (m. 6H).
3.6-3.5 (m, 2H), 3.45 (s, 3H), 33-3.1 (m, 3H). 2.4 (m, IH), 1.8-1.6 (m. 3H), 1.6-1.4 (m. IH), 13-1.2 (m. IH), 1.1 (m, IH), 0.9-1.01 (m,6H).
Example 23
Example 23 was synthesized as per procedure described in Example 10. In step 9, intermediate 25 was used to yield Exemple 23.
LC-MS: tR = 0.918 min, MS (ESI) m/z 469.2[M+H]*.
'H NMR: (CDjOD): B 7.62 (d, J « 8.0 Hz. 1 H), 7.48 (d, J = 8.0 Hz, I H), 7.27 (s, I H), 3.88-3.79 (m, 3H),
3.73-3.62 (m,3H),3.43(s,3H),3.25-3.09(m,2H), 236(t, IH), 1.81-1.60(m, 7H), 1.46(m, IH), 1.23 (m, IH), 1.06 (m, IH), 0.99-0.92 (m, 6H). I9F NMR: (CDÜOD 400 MHz): 6-160.19.
Example 24
Example 24 was synthesized as per procedure described in Example 10. In step 9,3-pyrimidylmethanamine was used instead of (5-fluoropyrimidine)-2-methylamine to yield Example 24. LC-MS: tR = 0.867 min, MS (ESI) mfz 445.1 [M+HJ*.
lH NMR: (CDjOD): S 9.08 (s. IH), 8.74 (d, J = 5.2 Hz, IH), 7.65 (d. J = 7.6 Hz, 1 H). 7.48 (m, 3H), 4.90 (s, 2H), 3.46 (s, 3H), 3.12-3.24 (m, 2H), 2.41 (m, IH), 1.63-1.75 (m, 3H), 1.49 (m. IH), 1.28 (m, 2H), 1.02 (d, J = 6.4 Hz, 3H), 0.96 (d, J = 6.4 Hz, 3H).
Example 25
Example 25 was synthesized in a method similar to Example 18. In step 9, (tctrohydrofuran-3yl)methanamine was utilized and the two diastereomers were separated by SFC-Method B. Further élaboration of intermediate arising out of the second peak from the SFC-Method B yielded Example 26. LCMS: tR = 0.845 min; m/z 430.3[M+H]*.
Ή NMR: (CDjOD): δ 7.63-7.66 (m, IH). 7.49-7.51 (m, IH). 7.31 (s, IH), 3.84-3.93 (m. IH), 3.71-3.81 (m, 2H), 3.48-3.63 (m, 3H), 3.43 (s, 3H), 3.11-3.31 (m, 2H). 231-2.78 (m. 2H), 1.22-2.09 (m, 7H), 1.001.06 (m, 4H), 0.93-0.99 (m, 3H).
Example 26
Example 26 was synthesized in a method similar to Example 1. In the synthesis of Example 28, in step 1 of Example I, methyl méthacrylate was used Instead of methyl acrylate. In step 3, the corresponding polar isomer 6B wns isolated and further elaborated as described in Example I. In step 9, intermediate 17 was utilized to yïeld Exampfe 28.
LC-MS: tR = 1.16 min, MS (ESI) m/z 441 [M+H]*.
'H NMR (CDjOD): δ 7,64 (dd. IH, J = 8„ 2 Hz), 730 (d, I H, J = 8 Hz), 7.30 (s, 1 H). 3.73 (dd, 2H, J =
22,4 Hz). 3.44 (s, 3H), 3.19 (ap q, 2H, J = 16 Hz), 2.49 (t. I H, J « 10 Hz), 1.82-1.72 (m, 2H), 1.71 -1.63 (m, IH), 135 (m, IH). 1.42-1.33 (m. 8H), 122-1.12 (m. IH), 1.08 (t, IH,J = 13Hz). 1.01 (d,3H,J = 6 Hz). 0.79 (t, 3H. J « 7 Hz).
Examplc 27
Example 27 was synthesized by a method asdescribed in Example 6. (l,4-dioxan-2-yl)methanamine was used in step 9 followed by oxîdation os described in step 10 to yield Example 27.
LC-MS: tR » 0.68 min. MS (ESI) m/z 439 [M+H]* lH NMR: (CDjOD, 400 MHz) δ 7.67 (not resolved, IH). 7.48 (d, 1 H). 7.20 (not resolved, IH), 6.66 (s, 2H). 3.78-2.97 (m, 14H),239(m, IH), l.92(m, IH), 1.66(m,2H), l.42(m, IH), 1.28- 1.03(m,2H), 0.89 (d, 3H), 0.88 (m, IH)
Examplc 28
Example 28 was synthesized from intermediate 11B from example 1 following the same procedure as In example 1 and utilizing-S-2-(anünomethyI) dioxane in step 9 of example 1.
LC-MS: tR » 0.894 min, MS (ESI) m/z 453.2 [M+H]+ 'H NMR: (CD3OD): δ 7.63 (dd, J= 7.6, 12 Hz, IH), 7.48 (d, J = 7.6 Hz, IH), 728 (s, IH), 3.61-3.85 (m, 8H), 3.58 (s, 3H), 356 (s, iH). 3.17 (m, 2H), 2.77-2.82 (m, IH), 2.10-2.17 (m, IH), 1.85-1.88 (m,
IH), 1.69-1.75(m, IH), 137-1.45 (m, 3H), 124-1.34 (m,2H), 1.09-1.15(m, IH),0.75(t,J=7.6 Hz, 3H).
Example 29
Starting with 6-Fluoro-3-indanonet Example 29 was synthesized in a method similar to Example 20. LC-MS tR = 1.02 min; MS (ESI) m/z 446 [M+H] *.
'H NMR (CDjOD ): δ 726 (dd, J « 8.4,5.2 Hz, IH), 6.95 (m, I H), 6.62 (dd, J = 8.4,2.4 Hz, 1 H). 4.023.89 (m,4H), 3.70 (d. J = 14.8 Hz, 1 H), 3.65 (d, J = 14.8 Hz, IH), 3.42 (s, 3H),3,12 (d, J = 152 Hz, IH),
2.98 (d. J = 152 Hz, IH), 2.34 (t, J = 10.0 Hz, IH), 1.79-I.60(m, 3H), 1.43 (m, IH), 134 (m, iH), 1.32 (m, IH), 1.30(5, 3H), l.00(m, IH), 0.99 (d, J = 6.8 Hz, 3 H), 0.94 (d, J = 6.0 Hz, 3H).

Claims (14)

  1. VVhat is daimed ls:
    1. A compound represented by a structural formula selected from:
    N=\ ^N. N-y w 1 Wy-rC0 W H-y»Y v-0 H>yY~ N<t Æ ^=° ΥγΑ Ns\ ’vyN'-W H.N Nr-(J>F
    Μς; 1JXazo/ H2N VN. _°
    or a pharmaceutically acceptable sait thereof.
  2. 2. A compound according to claim 1 or a pharmaceutically acceptable sait thereof for use as a médicament.
  3. 3. A pharmaceutical composition comprising at least one compound according to claim l or a pharmaceutically acceptable sait thereof in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier.
    10
  4. 4. A compound according to claim 1 or a pharmaceutically acceptable sait thereof for use in the treatment of a BACE] mediated disorder or disease.
  5. 5. A compound or a pharmaceutically acceptable sait thereof for use according to claim 4, wherein the BACE 1 medtated disorder or disease Is selected from the group consisting of a neurodegenerative disorder, cognitive décliné, cognitive impairment, dementia, and disease characterized by the production of β-omyloid deposits or neurofibrillary tangles.
  6. 6. A compound or a pharmaceutically acceptable sait thereof for use according to claim 5, wherein
    5 the disorder or disease 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 omyloid angiopathy, degenerative dementia, dementias of mixed vascular and degenerative origin, dementia associated with Parkinson’s disease, dementia associated with progressive supranudear palsy, dementia associated with cortical basal degeneratîon, diffuse Lewy body type of
    10 Alzheimer’s disease, dry âge related macular degeneratîon (AMD), and glaucoma.
  7. 7. A compound or a pharmaceutically acceptable sait thereof for use according to claim 6, wherein the disorder or disease Is Alzheimer’s disease.
    15
  8. 8. A compound or a pharmaceutically acceptable sait thereof for use according to claim 6, wherein the disorder or disease is glaucoma,
  9. 9. Use of a compound according to claim 1 or a pharmaceutically acceptable sait thereof for the manufacture of a médicament for the treatment of a BACEI mediated disorder in a subject.
  10. 10. Use of a compound according to claim 9 or a pharmaceutically acceptable sait thereof, wherein the BACE1 mediated disease ordisorder is selected from the group consistingof a neurodegenerative disorder, cognitive décliné, cognitive Impairment, dementia, and disease characterized by the production of β-omyloid deposits or neurofibrillary tangles.
  11. 11. Use of a compound according to claim 10 or a pharmaceutically acceptable sait thereof. wherein the disorder or disease 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
    30 degenerative origin, dementia associated with Parkinson’s discase, dementia associated with progressive supranudear palsy, dementia associated with cortical basal degeneratîon, diffuse Lewy body type of Alzheimer’s disease, dry âge related macular degeneratîon (AMD), and glaucoma.
  12. 12. Use of a compound according to claim 11 or a pharmaceutically acceptable sait thereof, wherein 35 the disease or disorder is Alzheimer’s disease.
  13. 13. Use of a compound according to claim 11 or a pharmaceutically acceptable sait thereof, wherein the disease or disorder is glaucoma.
  14. 14. A compound selected from the group consisting of:
OA1201400390 2012-03-05 2013-03-04 Inhibitors of beta-secretase. OA17087A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US1201400390 2012-03-05

Publications (1)

Publication Number Publication Date
OA17087A true OA17087A (en) 2016-03-23

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017236042B2 (en) Inhibitors of beta-secretase
ES2773439T3 (en) Novel Methyl Piperidine Compounds Useful for Inhibiting Microsomal Prostaglandin E2 Synthase-1
KR101891494B1 (en) New compounds
US10059668B2 (en) LSD1 inhibitors
US9018391B2 (en) Inhibitors of beta-secretase
OA17087A (en) Inhibitors of beta-secretase.
US9290477B2 (en) Inhibitors of β-secretase
OA17202A (en) Inhibitors of beta-secretase.