WO2009038411A2 - Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety - Google Patents

Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety Download PDF

Info

Publication number
WO2009038411A2
WO2009038411A2 PCT/KR2008/005592 KR2008005592W WO2009038411A2 WO 2009038411 A2 WO2009038411 A2 WO 2009038411A2 KR 2008005592 W KR2008005592 W KR 2008005592W WO 2009038411 A2 WO2009038411 A2 WO 2009038411A2
Authority
WO
WIPO (PCT)
Prior art keywords
oxo
phenyl
pyrazole
carboxamide
hydroxy
Prior art date
Application number
PCT/KR2008/005592
Other languages
French (fr)
Other versions
WO2009038411A3 (en
Inventor
Hyo-Shin Kwak
Hyejin Jung
Kyeongsik Min
Heuisul Park
Deog-Young Choi
Dongchul Lim
Jae-Ung Choi
Mijeong Park
Hyunjung Park
Original Assignee
Lg Life Sciences, Ltd.
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 Lg Life Sciences, Ltd. filed Critical Lg Life Sciences, Ltd.
Publication of WO2009038411A2 publication Critical patent/WO2009038411A2/en
Publication of WO2009038411A3 publication Critical patent/WO2009038411A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/18One oxygen or sulfur atom
    • C07D231/20One oxygen atom attached in position 3 or 5
    • C07D231/22One oxygen atom attached in position 3 or 5 with aryl radicals attached to ring nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to a novel compound for inhibiting beta-secretase activity or a pharmaceutically acceptable salt or isomer thereof, preparation thereof, and a pharmaceutical composition comprising a therapeutically effective amount of the same.
  • AD Alzheimer's disease
  • senile dementia is a neurodegenerative disease which gradually progresses with age and accounts for 50 to 70% of dementia patients.
  • the major symptoms of AD include memory loss, decline in cognitive-reasoning abilities and the like.
  • Most Alzheimer's cases occur around age 65 and progress for about 9 years to result in the death of Ihe patients.
  • the number of AD patients is increasing as society develops and ages, so that it is estimated that there will be approximately 6 million patients within 10 years in the USA and this number will further increase beyond that.
  • acetylcholine esterase inhibitors have been exclusively used as a general therapeutic agent. AriceptTM (Pfizer), ExelonTM (Novartis), and ReminylTM (Janssen) are known as representative examples of the acetylcholine esterase inhibitors.
  • these drugs cannot be defined as therapeutic agents of Alzheimer's disease, because they do not address the root cause of the disease and result in only partial recovery from the disease in some patients (about 40-50%), and their therapeutic effects are seen for a limited time period only.
  • the intrinsic nature of the disease requires long-term administration of the drugs, but administration of these therapeutic drugs is accompanied by various adverse side effects including liver toxicity.
  • Ab42 beta amyloid consisting of 42 amino acid residues
  • Beta-amyloid proteins are produced from a high-molecular weight amyloid precursor protein (APP) found in neuronal cells through serial cleavage events by 3 types of proteases (secretases). This process takes place at the Golgj apparatus of neuronal cells, wherein APP and secretases are anchored in the Golgi membrane.
  • the N-terminus of beta-amyloid (Ab) corresponds to the 99th amino acid from the C-terminus of APP, and this site is cleaved by beta-secretase (hereinafter, sometimes referred to as "beta-site APP cleaving enzyme" or "BACE").
  • the C- terminus of the membrane-bound beta-amyloid (Ab) is cleaved by gamma-secretase to generate a beta-amyloid (Ab) protein which is then secreted extracell ⁇ laiy from neuronal cells.
  • APP may also be cleaved at different sites via an alternative pathway; for example, where the middle site of Ab (between 16 th and 17 th amino acid residues from the N-terminus) is cleaved by alpha- secretase, sAPP alpha having a high molecular weight is produced and secreted. This alternate pathway precludes the formation of beta-amyloid.
  • Ab beta amyloid
  • AMO 40-amino acid fragment
  • Ab42 42-amino acid fragment
  • Ab40 tends to easily aggregate and accelerates the formation of amyloid plaques in the brains of diseased patients, thereby resulting in gradual necrosis of the surrounding neuronal cells. This is assumed to be a major pathogenic mechanism of Alzheimer's disease.
  • Ab40 and Ab42 are produced in a ratio of about 9: 1 under normal conditions.
  • levels of two amyloid proteins AMO and AM2 are increased or where levels of AM2 are selectively increased by mutations of Presenilin 1 and 2 genes, the onset of Alzheimer's disease is further accelerated and the symptoms of the disease are more severe. Therefore, it can be said that lowering of AM2 production is the most important factor for development of anti- Alzheimer drugs. For this purpose, there is a need for development of beta- or gamma-secretase inhibitors.
  • beta- and gamma-secretases are known as aspartic proteases and found in the membrane-bound form. However, no gene coding for gamma-secretase has yet been identified. Further, it is known that substrates for gamma-secretase are not limited to APP, but that the enzyme also participates in the cleavage of Notch proteins, which are known to play crucial roles in regulating cell fate decision during differentiation processes. In particular, gene-knockout animals from which a gamma-secretase gene has been deleted died in utero, and recent clinical tests on gamma-secretase inhibitors showed significant drug toxicity. For these reasons, gamma-secretase inhibitors are not likely to be promising drug candidates. As a result, it has not yet been confirmed whether gamma-secretase inhibitors can be developed as safe anti-Alzheimer medications.
  • BACE-specific inhibitors As discussed above, upon considering that conventional commercially available drugs merely exhibit palliative effects of disease symptoms, thus providing substantially no effects on the progression of the concerned disease, it is certain that development of the BACE-specific inhibitors will lead to the development of a novel and remarkable drug against Alzheimer's disease.
  • many pharmaceutical companies have published study results on BACE inhibitors, for example by Merck (WO 2006/078577, WO 2006/060109 and WO 2006/057983), Elan (WO 2004/022523 and WO 2005/095326), Schering-Plough (WO 2006/014762 and WO 2006/014944), BMS (WO 2005/182105 and WO 2005/030758), and Eli-Lilly (WO 2005/108358 and WO 2006/034093).
  • BACE inhibitor compounds which were compounds having a small molecular weight and low blood-brain barrier (BBB) permeability.
  • BBB blood-brain barrier
  • these compounds exhibited various limitations due to selectivity for cathepsin D (Cat. D) or in vivo cytotoxicity.
  • Cat. D blood-brain barrier
  • a great deal of attention has been recently focused on Merck compounds. Through early introduction of dicarbonyl compounds and subsequently sulfonamide compounds from fellow pharma giant Sunesis, Merck reported excellent inhibitory effects and selectivity of the drug compounds. Since then, numerous drug companies issued various compounds in the form of Merck compounds.
  • the present invention is intended to improve blood-brain barrier (BBB) permeability of a drug compound which is a technical problem suffered by conventional sulfonamide compounds.
  • BBB blood-brain barrier
  • an object of the present invention is to provide a novel compound having beta- secretase inhibitory activity through five-membered heterocyclic compounds apart from sulfonamide in terms of a chemical structure, and pharmaceutically acceptable salts and isomers thereof.
  • R 1 is selected from the group consisting of hydrogen, alkyl, alkene, and -(CH 2 )P-A-R 7 wherein p is 0 to 2, A is aryl or heteroaryl, and R 7 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, -NHC(O)-alkyl and aryloxy;
  • R 2 is selected from the group consisting of hydrogen, alkyl, alkoxy and -(CR 8 R ⁇ )P-R 10 wherein p is 0 to 2, R 8 and R 9 are each independently selected from the group consisting of hydrogen, alkyl and alkoxy, and R 10 is selected from the group consisting of cycloalkyl, aryl, heterocycle and heteroaryl; and
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, alkyl and -(O)Q-(CH 2 )P-R 11 wherein p is 0 to 2, q is 0 or 1 , and R 11 is aryl or heteroaryl,
  • R 5 is selected from the group consisting of hydrogen, alkyl, alkoxy and -(0 ⁇ -(CH 2 )P-R 1 ' wherein p is 0 to 2, q is 0 or 1 , and R 1 ' is aryl or heteroaryl
  • R 6 is selected from the group consisting of hydrogen, alkyl, alkoxy, and -(CH 2 )p-A'-R 12 wherein p is 0 to 2
  • A' is phenyl, heteroaryl, or a bicyclic compound containing phenyl
  • R is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, alkoxy, NHR 13 and NR 13 R 14 wherein R I3 and R 14 are each independently hydrogen or alkyl, or R 13 and R 14 are cyclized to form alkylene; or a pharmaceutically acceptable salt or isomer thereof.
  • the alkyl, alkoxy, aryl, cycloalkyl, heterocycle, and heteroaryl may be substituted or unsubstituted.
  • the substituent may be at least one selected from the group consisting of halogen, amino, alkylamino, dialkylamino, alkylacylamino, Ci-C 4 alkyl, hydroxy, Q- C 4 alkyl alkoxy, aryl alkoxy and oxo. Where appropriate, these substituents may also be substituted.
  • the substituents are as exemplified above and the substituents may be taken together to form a cyclic structure.
  • heteroaryl and heterocycle are each independently a 4 to 8-membered ring containing 1 to 3 hetero atoms selected from the group consisting of O, N and S, and having 0 to 3 double bonds, and preferably a 5 or 6-membered ring having 1 to 2 double bonds.
  • Compounds of Formula (I) in accordance with the present invention have a chemical structure that is distinctly different from that of conventional known sulfonamide- or peptide-based beta-secretase inhibitor compounds.
  • the compounds of the present invention exhibit improved BBB permeability to thereby exert excellent inhibitory effects on human beta-secretase which is correlated with improvement of cognitive functions or prevention and treatment of neurodegenerative diseases such as Alzheimer's disease.
  • the term “isomer” means a compound of the present invention or a salt thereof that has the same chemical formula or molecular formula but is optically or sterically different therefrom.
  • compound of Formula (T) is intended to encompass compounds of Formula (1) per se as well as pharmaceutically acceptable salts and isomers thereof.
  • alkyl means an aliphatic hydrocarbon group.
  • the alkyl moiety may be a "saturated alkyl” group, which means that it does not contain any alkene or alkyne moiety.
  • the alkyl moiety may also be an "unsaturated alkyl” group, which means that it contains at least one alkene or alkyne moiety.
  • An “alkene” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond
  • an “alkyne” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
  • the alkyl moiety may be branched, straight chain, or cyclic.
  • the alkyl group may have 1 to 20 carbon atoms.
  • the alkyl group may also be a medium- sized alkyl having 1 to 10 carbon atoms.
  • the alkyl group could also be a lower alkyl having 1 to 6 carbon atoms.
  • “Ci-C 4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, Ie., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • the alkyl group When the alkyl group is used alone or in combination with alkyloxy, it may be a linear or branched hydrocarbon radical.
  • alkyl group may include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, cyclopropyl, cycloburyl, cyclopentyl, and cyclohexyl.
  • alkoxy refers to oxo alkyl having 1 to 10 carbon atoms.
  • cycloalkyl refers to an unsaturated aliphatic 4 to 10-membered ring, including cyclohexyl.
  • aryl refers to an aromatic group which has at least one ring having a conjugated pi ( ⁇ ) electron system and includes both carbocyclic aryl (for example, phenyl) and heterocyclic aryl (for example, pyridine) groups. This term is intended to include monocyclic or fused-ring polycyclic (Ie., rings which share adjacent pairs of carbon atoms) groups. Specifically, aryl means a 4 to 10-membered, preferably 6 to 10-membered aromatic monocyclic or multicyclic group, including phenyl, naphthyl, and the like.
  • heteroaryl refers to a 4 to 8-membered ring having 1 to 3 hetero atoms selected from the group consisting of N, O and S and 0 to 3 double bonds.
  • the heteroaryl may be preferably an aromatic 4 to 8-membered ring and more preferably a 5 or 6-membered ring, which may be fused with benzo or C 3 -C 8 cycloalkyl.
  • Examples of monocyclic heteroaryl may include, but are not limited to, thiazole, oxazole, thiophene, furan, pyrrole, imidazole, is ⁇ xazole, pyrazole, triazole, thiadiazole, tetrazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, and the like.
  • tricyclic heteroaryl may include, but are not limited to, indole, indoline, benzothiophene, benzofuran, benzimidazole, benzoxazole, benzisoxazole, benzothiazole, benzothiadiazole, benzotriazole, quinoline, isoquinoline, purine, furopyridine and the like.
  • heterocycle refers to a 3 to 10-membered ring which has 1 to 3 hetero atoms selected from the group consisting of N, O and S, may be fused with benzo or C 3 -Cg cycloalkyl, and contains 1 or 2 double bonds or may be saturated.
  • the heterocycle may preferably be a 4 to 8- membered ring, and more preferably a 5 or 6-membered ring, which may include, but are not limited to, piperidine, morpholine, thiomorpholine, pyrrolidine, imidazolidine, tetrahydrofuran, piperazine, and the like.
  • Other terms used herein can be interpreted as having their usual meanings in the art to which the present invention pertains.
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkene and - (CH 2 )P-A-R 7 wherein p is 0 to 2, A is 6 to 10-membered aryl or 5 or 6-membered heteroaryl, and R 7 is selected from the group consisting of hydrogen, halogen, C 1 -C 3 alkyl, C]-C 3 alkoxy, NHC(O)- (C)-C 6 alkyl) and 5 or 6-membered aryloxy;
  • R 2 is selected from the group consisting OfCi-C 6 alkyl, Ci-C 6 alkoxy and -(CR 8 RV-R 10 wherein p is 0 to 2, R 8 and R 9 are each independently hydrogen or Ci-C 4 alkyl, and R 10 is selected from the group consisting of 5 or 6-membered cycloalkyl, 6 to 10-membered aryl, and 5 or 6- membered heteroaryl; and
  • X is C or N
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, C 1 -C 6 alkyl and -(O)q-(CH 2 )p-R u wherein p is 0 to 2, q is 0 or 1, and R 11 is phenyl or 5 or 6-membered heteroaryl, R 5 is selected from the group consisting of hydrogen, Ci-C 6 alkyl, Ci-C 6 alkoxy and -(O)q-
  • R 6 is selected from the group consisting of hydrogen, Cj-C 6 alkyl and -(QHyp-A'-R 12 wherein p is 0 to 2, A' is phenyl, 5 or 6-membered heteroaryl, or a bicyclic compound containing phenyl, and R 12 is selected from the group consisting of hydrogen, halogen, hydroxy, C 1 -C 6 alkyl, Q-C ⁇ alkoxy, NHR 13 and NR 13 R 14 wherein R 13 and R 14 are each independently hydrogen or Q-C 4 alkyl, or R 13 and R 14 may be cyclized to form alkylene.
  • the number of R 1 may be in a range of 1 to 3, and R 1 may be positioned at carbon and/or nitrogen of the pyrazole ring.
  • R 1 in the compound of Formula (T) may consist of R ⁇ substituted at N on position 2 and R ⁇ substituted at C on position 4 of the pyrazole ring.
  • R 1 is selected from the group consisting of hydrogen, Cj-C 4 alkyl, C 2 -C 4 alkene and -(CH 2 )p-A-R 7 wherein p is 0 to 2, A is phenyl or pyridine, and R 7 is selected from the group consisting of hydrogen, halogen, trifluoromethyl, trifluoromethoxy, acetylamino and 5 or 6-membered aryloxy, particularly preferably hydrogen, methyl, hydroxyethyl, vinyl, phenyl, 4-chlorophenyl, 4-trifluoromethyloxyphenyl, 3- trffluoromethylphenyl, 3-trifluorophenyl, 4-trifluoromethylphenyl, 2-trifluoromethylphenyl, benzyl, pyridin-2-yl, 3-phenoxyphenyl and 2-acetylaminophenyl.
  • R 2 is Q-Qalkoxy or -(CR 8 RV-R 10 wherein p is 0 to 2, R 8 and R 9 are each independently hydrogen or Ci-C 4 alkyl, and R 10 is selected from the group consisting of 5 or 6- membered cycloalkyl, 6 to 10-membered aryl and 5 or 6-membered heteroaryl. More preferably, R 2 is selected from the group consisting of cyclohexyl, phenyl, 3,5-difluorophenyl and naphthyl.
  • R 3 is preferably selected from the group consisting of hydrogen, Ci-C 6 alkyl and [-(O)q- (CH 2 )P-R 11 ] wherein R 11 may be phenyl or 5 or 6-membered heteroaryl.
  • R 3 is more preferably hydrogen, benzyloxy or benzyl.
  • R 4 is preferably selected from the group consisting of hydrogen, C 1 -C 4 alkyl and O- (CH 2 )P-R 11 wherein R 11 may be phenyl or 5 or 6-membered heteroaryl.
  • R 4 is more preferably selected from the group consisting of hydrogen, methyl, isobutyl and benzyloxy.
  • R 5 is preferably selected from the group consisting of hydrogen, C 1 -C 6 alkyl and -O-R 11 wherein R 11 may be phenyl or 5 or 6-membered heteroaryl. R 5 is more preferably hydrogen or phenoxy.
  • R 6 is preferably selected from the group consisting of hydrogen, C 1 -C 4 alkyl and -A' -R 12 wherein A' is phenyl, 5 or 6-membered heteroaryl, or a bicyclic compound containing phenyl, and R 12 is selected from the group consisting of hydrogen, halogen, hydroxy, Ci-C 6 alkyl, Ci-C 6 alkoxy, NHR 13 and NR 13 R 14 wherein R 13 and R 14 are each independently hydrogen or Ci-C 4 alkyl, or R 13 and R 14 are cyclized to form alkylene.
  • R 6 is preferably selected from the group consisting of hydrogen, methyl, 3-dimethylaminophenyl, 3-(t-butyl)-phenyl, (3-isopropyl-[l,2,4]- oxadiazol-5-yl), 3-(pyrrolidin-l-yl)phenyl, and (l-hydroxy-indan-5-yl).
  • the compound in accordance with the present invention may form a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salt means acid addition salts of the compound with acids capable of forming a non-toxic acid addition salt containing pharmaceutically acceptable anions, for example, inorganic acids such as sulfuric acid, hydrochloric acid, nitric acid, phosphoric acid, hydrobromic acid and hydroiodic acid; organic carboxylic acids such as tartaric acid, formic acid, citric acid, acetic acid, trichloroacetic acid, trifluoroacetic acid, gluconic acid, benzoic acid, lactic acid, fiimaric acid, and maleic acid; or sulfonic acids such as methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid and naphthalenesulfonic acid.
  • the term “isomer” means a compound of the present invention or a salt thereof, that has the same chemical formula or molecular formula but is optically or stereochemically different therefrom.
  • the compounds of Formula (I) in accordance with the present invention may have an asymmetric carbon center, and therefore can be present in the form of optical isomers (R or S isomeric forms), racemates, diastereomeric mixtures, and individual diasteromers.
  • optical isomers R or S isomeric forms
  • racemates racemates
  • diastereomeric mixtures and individual diasteromers.
  • geometrical isomers trans and cis isomeric forms.
  • the present invention encompasses all these isomeric forms and mixtures.
  • Representative examples of the compound of Formula (T) in accordance with the present invention include the following compounds:
  • a method for preparing a compound of Formula (I) for a better understanding of the present invention, preparation of the compound of Formula (T) will be illustrated with reference to exemplary synthetic Reaction Schemes.
  • skilled persons having knowledge about synthesis of compounds in the art to which the present invention pertains could readily prepare the compound of Formula (I) according to various synthetic methods, based on the chemical structure of Formula (I). Therefore, these methods also fall within the scope of the present invention. That is, the compound of Formula (I) may be prepared by any combination of various synthetic methods disclosed herein or in prior art. Therefore, the below description associated with preparation processes is provided only to disclose exemplary ones, and Hie scope of the present invention is not limited to only such preparation processes as described herein.
  • the compound of Formula (I) can be synthesized by amide-coupling reaction of carboxylic acid of Formula 2 with an amine compound of Formula 3.
  • R 1 , R 2 and B are as defined above.
  • known coupling agents usable in the amide coupling may include, but are not limited to, carbodiimides such as dicyclohexylcarbodiimide (DCC), 1 -(3-dimethylaminopropyl)- 3-ethylcarbodiimide (EDC), 1 ,1 '-dicarbonyldiimidazolc (CDl), etc.
  • HATU hydroxybenzDtriazole
  • IIOBT 1-hydroxybenzDtriazole
  • HOAT l-hydroxy-7-azabenzotriazo]e
  • BOP-Cl bis-(2-oxo-3- oxazolidinyl)-phosphinic acid chloride
  • DPPA diphenylphosphorylazide
  • solvents that can be used in amide coupling may include DCM, DMF, and DCE.
  • bases for activation of the reaction may include triethylamine, diisopropylethylamine, etc.
  • the carboxylic acid of Formula 2 is commercially available or can be synthesized by cyclization and hydrolysis from a compound of Formula 4 or 5 and a hydrazine compound of Formula 6, according to the synthetic procedure of Reaction Scheme 2 below.
  • Cyclization may be carried out by heating reactants in the presence of a base, for example triethyl amine, N-methylmorpholine, diisopropyl ethyl amine, or K 2 CO 3 , in a solvent such as DMF,
  • a base for example triethyl amine, N-methylmorpholine, diisopropyl ethyl amine, or K 2 CO 3
  • a solvent such as DMF
  • reaction is conventionally carried out at a temperature of 60 to 120 ° C for 4 to 24 hours.
  • Hydrolysis to obtain the compound of Formula 2 is carried out in a mixed solution of water and an organic solvent, using a base.
  • organic solvent usable in hydrolysis may include THF, methanol, and dioxane.
  • base may include LiOH, KOH, and
  • the hydrazine compound of Formula 6 is commercially available or may be obtained by conversion of an amine group of an amine compound of Formula 7 into a hydrazine group, according to a known method disclosed in Journal of the American Chemical Society, 198(48),
  • Nitrosation may be carried out in the presence of hydrochloric acid, using NaNO 2 .
  • Reduction of the nitroso compound may be carried out in the presence of hydrochloric acid, using SnCl 2 .
  • a compound where B is in the form of a chain can be synthesized from a commercially available epoxide, according to a synthetic procedure as disclosed in Reaction Scheme 4 below.
  • an azide compound of Formula 9 can be obtained by ring-opening of an epoxide compound using NaN 3
  • an amine compound of Formula 10 can be obtained by reduction of the azide compound through hydrogenation using a
  • the reaction is carried out in a solvent such as methanol, ethanol, and dioxane, at 1 to
  • Reductive animation to obtain a compound of Formula 3-1 is carried out using a compound (ketone or aldehyde) containing a carbonyl group.
  • an available reducing agent may include sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, etc.
  • acid may be used as a catalyst.
  • an available acid catalyst may include inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, and phosphoric acid; organic carbonic acids such as acetic acid, and trifluoroacetic acid; and amine salts such as ammonium chloride. Particularly preferred is hydrochloric acid or acetic acid.
  • Epoxide that is not readily commercially available can be prepared by stereoselective synthesis as shown in Reaction Scheme 5 below.
  • R 2 is -CH 2 -R 2
  • X is a leaving group, such as bromine, iodine, or methanesulfonyl.
  • Stereoselective alkylation is carried out using a chiral auxiliary of Formula 11.
  • hydrolysis of the compound of Formula 12 is carried out to prepare an amino acid derivative of Formula 13.
  • the hydrolysis reaction to obtain the compound of Formula 13 was previously described.
  • the reaction is carried out using iodomethane in the presence of a base to obtain an iodocarbonyl compound of Formula 15.
  • Reduction of the compound of Formula 15 is then carried out to obtain an iodohydrin compound of Formula 16.
  • Reduction is carried out using NaBH 4 , NaB(CN)H 3 , or the like.
  • an epoxide compound of Formula 17 can be obtained by cyclization of the iodohydrin compound in the presence of a base.
  • a heterocyclic compound such as oxadiazole can be prepared according to a synthetic procedure in Reaction Scheme 6 below:
  • an amidoxime compound of Formula 18 is reacted with oxalate to synthesize an oxadiazole compound of Formula 20, and reduction of the reaction product is then carried out using diisobutylaluminum (DIBAL) to obtain an aldehyde compound of Formula 21.
  • DIBAL diisobutylaluminum
  • R 2 is as defined above, and n is 0 or 1.
  • a nitro compound of Formula 21 and an aldehyde of Formula 22 are reacted under basic conditions to prepare a compound of Formula 23, and a nitro group of Formula 23 is reduced in the presence of hydrogen to obtain a compound of Formula 24.
  • the nitro compound of Formula 21 can also be synthesized by a known method disclosed in WO2006/034093, according to a synthetic procedure of Reaction Scheme 8.
  • R is as defined above.
  • Aldehyde of Formula 25 and nitromethane are subjected to coupling reaction under basic conditions to obtain a compound of Formula 26 which is then acetylated to prepare a compound of Formula 27.
  • a compound of Formula 28 is reduced to synthesize a compound of Formula 21.
  • Aldehyde of Formula 22 can be synthesized from an amino acid.
  • An alcohol compound of Formula 30 can be obtained from a compound of Formula 29 using a reducing agent, for example NaBH 4 or LAH, in methanol or THF. Then, the alcohol compound is subjected to oxidation to thereby obtain a compound of Formula 22.
  • a reducing agent for example NaBH 4 or LAH
  • An oxidant may be SO 3 P y
  • a solvent may be DCM.
  • Aldehyde of Formula 31 and piperazine of Formula 32 are reacted in the presence of a base to obtain a compound of Formula 33.
  • the deprotection of the Boc group from the compound of Formula 33 is then carried out to obtain a compound of Formula 34.
  • R i5 is as defined above, and [RA] means reductive amination.
  • aldehyde of Formula 36 is synthesized from a serine derivative of Formula 35. Then, a compound of Formula 37 is synthesized through reductive amination of Formula 36. By repeating the same procedure, aldehyde of Formula
  • reaction product can be separated and purified by conventional post-treatments, for example, chromatography, recrystallization, and the like.
  • a pharmaceutical composition for inhibiting beta-secretase comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt or isomer thereof as an active ingredient and a pharmaceutically acceptable carrier.
  • the composition may further comprise one or more diluents or excipients.
  • the compound of Formula (I) exhibits excellent inhibitory effects on beta-secretase, so the present invention provides a beta-secretase inhibitor composition comprising a compound of Formula (J) in conjunction with a pharmaceutically acceptable carrier.
  • the composition in accordance with the present invention exhibits excellent effects on improvement of cognitive functions or treatment or prevention of neurodegenerative diseases, particularly Alzheimer's disease without being limited thereto.
  • pharmaceutical composition as used herein means a mixture of a compound of the invention with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but are not limited to oral, injection, aerosol, parenteral, and topical administrations.
  • compositions can also be obtained by reacting compounds with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • terapéuticaally effective amount means that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disease being treated.
  • a therapeutically effective amount refers to that amount which has the effect of (i) reversing the rate of progress of a disease, (ii) inhibiting or slowing to some extent further progress of the disease, and/or, (iii) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with the disease.
  • carrier means a chemical compound that facilitates the incorporation of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • carrier facilitates the uptake of many organic compounds into the cells or tissues of an organism.
  • diot defines chemical compounds diluted in water that will dissolve the compound of interest as well as stabilize the biologically active form of the compound. Salts dissolved in buffered solutions are utilized as diluents in the art.
  • One commonly used buffered solution is phosphate buffered saline because it mimics the salt conditions of human body fluid. Since buffer salts can control the pH of a solution at low concentrations, a buffered diluent rarely modifies the biological activity of a compound.
  • physiologically acceptable defines a carrier or diluent that does not abrogate the biological activity and properties of the compound.
  • the compounds described herein can be administered to a human patient per se, or in pharmaceutical compositions in which they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipients. Techniques for formulation and administration of the compounds may be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, 18th edition, 1990.
  • Compounds of the present invention may be administered via any conventional routes, depending upon desired applications.
  • Preferred routes of administration may, for example, include injection, oral and intranasal administrations.
  • the active compounds may be administered by dermal, intraperitoneal, retroperitoneal and rectal routes.
  • the pharmaceutical composition of the present invention may be manufactured in a manner that is itself known, e.g. , by means of conventional mixing, dissolving, granulating, dragee- making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g. , in Remington's Pharmaceutical Sciences, supra.
  • Injectable preparations for example aqueous or oily suspensions for sterile injection may be prepared by a conventional method known in the art, using suitable dispersants, wetting agents and/or suspending agents.
  • solvents that can be used in the formulation of injectable preparations may include water, Ringer's solution, and isotonic NaCl solution.
  • sterile fixing oil is also conventionally used as a solvent or suspending medium. Any non-irritable fixing oil including monoglyceride and diglyceride may be used for this purpose.
  • fatty acids such as oleic acid may also be used for injectable preparations.
  • solid dosage forms for oral administration may include capsules, tablets, pills, powders and granules. Particularly preferred are capsules and tablets. Tablets and pills may be preferably provided with enteric coatings.
  • the solid dosage form may be prepared by mixing an active compound of Formula (I) in accordance with the present invention with one or more inert diluents (such as sucrose, lactose, and starch) and carriers such as lubricants (such as magnesium stearate), disintegrants, binders, and the like.
  • compositions suitable for use in the present invention include compositions in which the active ingredient is contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • a daily dosage of the compound which may be administered to a subject in a single or divided doses, is preferably in a range of 10 to 100 mg/kg.
  • a specific dose of the active ingredient for individual patients may vary depending on various factors such as kinds of compounds to be used, weight, sex, health conditions, and dietary habits of patients, treatment duration, administration manners, excretion rates, drug mixing and severity of disease.
  • EDC l-(3-dimethylaminopropyl)-3-ethylcarbodiimide
  • hydrochloride EtOAc ethyl acetate
  • Step B S-oxo-l-G-phenoxyphenvlM ⁇ -dihvdiO-l-H-pyrazole-S-carboxvlic acid S-oxo-l-CS-phenoxypheny ⁇ S-dihydro-l-H-pyrazole-S-carboxylic acid methyl ester (4.0 g) obtained in Step A was dissolved in methanol, to which water and LiOH were then added, followed by stirring at room temperature for 3 hours. After the reaction was complete, the reaction mixture was adjusted to pH of 2 to 3 with addition of IN HCl, followed by EtOAc extraction. The organic extract was dried over MgSO 4 and distilled under reduced pressure to afford the title compound.
  • Step B 4-methyl-5-oxo-l-vinyl4,5-dihvdiO-l-H-pyraziole-3-carboxylic acid
  • Step A ((lS2RV3-azido-l-benzyl-2-hvdiOxypropyl)-carbamic acid t-butyl ester
  • Step B (Sy2-amino-3-(3,5-difluorophenyl)propionic acid methyl ester
  • Step D [(SVl-fB ⁇ -dMuorobenzylVS-cMoro ⁇ -oxo-propyl ' lcarbamic acid t-butyl ester
  • Step F [(S)-2-(3,5-difluorophenylVl-(S)-oxiranyl-ethyll-carbamic acid t-butyl ester [(lS ⁇ S ⁇ l ⁇ S ⁇ -difluorober ⁇ l ⁇ -hydroxy-S-chloro-propylJcarbarnic acid t-butyl ester
  • Step G [flS,2R)-3-amino-l-(3,5- ⁇ 4fluoro-rjeri2 ⁇ 1)-2-hy ⁇ Voxy-propyl]-carbarnic acid t-butyl ester
  • Step B [(5-bromo-2,3-dihvdro-lH-inden-yl ' )oxy]ft-butyl)dimethylsilane 5-bromoindan-l-ol (260 mg, 1.22 mmol) obtained in Step A was dissolved in methylene chloride (50 mL) and the mixture was cooled to 0 0 C, followed by addition of trimethyldimethylsilyl trifluoromeihane sulfonate (355 mg, 1.34 mmol). After stirring for 30 min, the reaction temperature was elevated to room temperature. The reaction was terminated with addition of a saturated aqueous solution of ammonium chloride, followed by extraction with a 0.5N HCl aqueous solution. The organic layer was taken, dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. The residue was directly used in subsequent reactions without further purification.
  • Step C l-([t-butyl(dimethyl)silylloxy ⁇ indane-5-carbaldehyde
  • Step B (S>2-[(R>3-benzyloxy-2-t-butoxycarto ⁇ acid methyl ester
  • Step C rS)-2-(((RV2-amino-3-benzyloxy-piOpylaminoV3-methyl-butyric acid methyl ester
  • Step D (3S,6RV6-benzyloxymethyl-3-isopiOpyl-piperazin-2-one (S)-2-(((R)-2-amino-3-ber ⁇ loxy-propylamino)-3-me%l-butyric acid methyl ester (1.0 g,
  • Step F ((2R,5S)-5-isopropyl-piperazin-2-yl * )-methanol
  • Step B 3,5-difluoro-5-[(E)-2-nitrovinyl1ben2ene l-(3,5-difluoro)-2-nitro-ethanol (2.19 g, 10.7 mmol) obtained in Step A was dissolved in DCM, followed by addition of 4-dimethylaminopyridine (139 mg) and then (Ac) 2 O (1.11 mL, 11.8 mmol). The reaction solution was stirred at room temperature for 0.5 hours, diluted with DCM, sequentially washed with a 2% HCl aqueous solution, a NaCl saturated aqueous solution and a
  • Step D (RV2-[(lS,2SV3-f3,5-difluoro-phenylH-hydroxy-2-r ⁇ acid t-butyl ester
  • Step E (RV2-[(lS,2SV2-amino-3-(3,5-dfluoro-phenylVl-hvdroxy-propyll-pyrrolidine-l- carboxylic acid t-butyl ester
  • reaction mixture was cooled to -78 ° C, and a solution of t-butyl 4-benzyl- 3-oxopiperazine-l-carboxylate (B) (2.44 g, 8.40 mmol) in 15 mL of anhydrous tetrahydrofuran was added dropwise thereto, followed by stirring at that temperature for 1.5 hours. Thereafter, a solution of 2(S)-2-(dibenzylamino)-3-phenylpropanal (A) (2.99 g, 9.07 mmol) in anhydrous tetrahydrofuran (15 mL) was added to the reaction mixture which was gradually warmed to room temperature.
  • B t-butyl 4-benzyl- 3-oxopiperazine-l-carboxylate
  • A 2(S)-2-(dibenzylamino)-3-phenylpropanal
  • Step B (SV2-[(lS,2S)-2-aniino-l-hydroxy-3-phenylpropyl]-4-benzyl-3-oxo-piperazine-l- carboxylic acid t-butyl ester
  • Step A rSV2-rmethoxy-methyl-carbamoylVpyrroHdine-l-carboxylic acid t-butyl ester
  • S commercially available (S)-l-BOC-proline (2 g, 9.3 mmol) was placed in DCM (30 mL), followed by sequential addition of triethylamine (5. 22 mL, 37.2 mmol), HOBt (1.89 g, 13.95 mmol), EDC (2.14 g, 13. 95 mmol) and meihoxy methyl amine (0.91 g, 9.3 mmol) and stirring at room temperature for 12 hours.
  • Step B (Sy2-acetyl-pyrrolidine-l-carboxylic acid t-butyl ester (S ⁇ methoxy-me ⁇ yl-carbamoyO-pyrrolidine-l-carboxylic acid t-butyl ester (1.29 g,
  • Step C ( " SVl-pyrrolidin-2-yl-ethanone
  • Step D r(SV2-((SV2-aceM-pyrroUdm-l-ylVl-benzyloxymethyl-2-oxo-ethyl]-carbamic acid t-butyl ester
  • Step E (SVl-methyM-oxo-octahydro-pyrrolofl ⁇ -aipyrazme-S-carboxyaldehyde
  • Step F rSV3-( ⁇ S.2SV2-aniino-l-hvdroxy-3-phenyl-proDylVl-methyl-hexahvdro-pyrrolo
  • Step A (2R,3S)-3-arnino-l-O-dimethylaniino-ben2ylainino)4-phenyl-butan-2-ol
  • Step B N-[r2S3RM-[G-dimethylaminophenyl)methylaminol-3-hydroxy-l -phenylbutan-2-yl1-5- oxo- 1 -phenyHH-pyrazole-S-carboxamide
  • Example 22 N-r(lR.2SV3-r3,5-difluorophenylVl-hvdroxy-l-rr2RVpyrrolidin-2-vnpropan-2-yll-5- oxo-l-phenvMH-pyrazole ⁇ -carboxamide
  • Step A 0lV2- ⁇ riS2SV3-r3,5-difluoro-phenyl)-l-hvdiOxy-2-[(5-oxo-l-phenyl-4,5-dihvdro-lH- pyrazole ⁇ -carbonylVarninoJ-propyll-pyrrolidine-l-carboxylic acid t-butyl ester
  • Step B N- ⁇ lR ⁇ Sl-S-rB ⁇ -dmuorophenvn-l-hvdroxy-l-rQRVpyrrolidin ⁇ -ylipiOpan ⁇ -yll-S-oxo- 1 -phenyl-4H-pyrazple-3-c ⁇ boxamide
  • BACE cDNA (ATCC, Cat. No. 6896840) was purchased which was synthesized based on a human BACE2 gene sequence (Accession No. BC014453) disclosed in the public Genbank data base. Only the ectodomain which corresponds to the region of from the amino acid residues 1 to 466 with exclusion of a transmembrane domain and a cytoplasmic domain in the entire BACE gene was re-cloned, and then the base sequence of Fc region which corresponds to 230 amino acids (from the amino acid residues 1 to 466) of human Immunoglobulin G (hlgG) was linked to the 3' end thereof.
  • hlgG human Immunoglobulin G
  • BACE-Fc The BACE (ectodomain)-IgG Fc (hereinafter, referred to as "BACE-Fc") was ligated between BamHL and Xhol sites of pCDNA3 (Invitrogen) as a mammalian expression vector to construct a BACE2-Fc expression vector, designated as pCDNA3 BACE2-Fc.
  • Step B Construction of mammalian cell line expressing BACE2-Fc fusion protein Chinese hamster ovary (CHO) DHFR- cells (ATCC Accession No. CRL9096) were cultured in an alpha-minimum essential medium ( ⁇ -MEM, GIBCO-BRL) supplemented with 10% fetal bovine serum (FBS, GIBCO-BRL) and then transferred to a 100 mm culture plate. When the cells reached confluence, they were transfected with the BACE2-Fc-expressing vector pCDNA3
  • BACE2-Fc using Lipofectamine Plus (Life Technologies). Selection of transfectant cells was carried out in a medium containing 10% dialyzed fetal bovine serum (dFBS, JRH) supplemented with 1 mg/mL of Geneticin (G418 sulfate, GIBCO-BRL). The culture medium was replaced with a fresh one every 4 days. Then, 100 clones were isolated and cultured on a 24-well culture plate. Among these isolated clones, 20 clones showing acceptable growth rates were sub-cultured on a 24-well culture plates for 3 days at an equal cell density (2 x 10 5 cells/mL/24-well). An amount of BACE2- Fc protein secreted into the medium was quantified by ELISA method using goat anti-human IgG
  • Step C Production and purification of BACE2-Fc fusion protein 2 x 10 5 cells/mL of the CHO DHFR- BACE2-Fc #66 cell line were inoculated into a roller bottle containing 250 mL of ⁇ -MEM supplemented with 10% dFBS, and cultured in a Roll-In cell incubator (Bellco) at 37 ° C and 40 rpm for 4 days. When cells reached confluence, they were washed once with 250 mL of a serum-free medium (SFII, GD3CO-BRL), and 500 mL of a serum- free medium containing insulin (0.5 ⁇ g/mL, SIGMA) was added, followed by culture for 3 days.
  • SFII serum-free medium
  • GD3CO-BRL serum-free medium
  • insulin 0.5 ⁇ g/mL, SIGMA
  • Step D Beta-secretase 2 activity assay using fluorescence-labeled specific substrate
  • FRET Fluorescence Resonance Energy Transfer
  • an amyloid precursor protein known as an intracellular beta secretase 2-specific substrate
  • a peptide corresponding to a 10-amino acid region containing a beta-secretase cleavage site was synthesized with inclusion of EDANS as a fluorophore and DABCYL as a quenching group linked thereto.
  • EDANS as a fluorophore
  • DABCYL DABCYL
  • Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 ° C.
  • a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then nine successive doubling dilutions were carried out with an equal volume of DMSO.
  • reaction product was measured by the magnitude of fluorescence at a 350 nm excitation wavelength and a 510 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device).
  • concentration of a synthetic compound inhibiting 50% of beta- secretase activity, Le., IC 50 and Ki were determined by comparing the measurement value with that of a control group with no addition of the synthetic compound.
  • FRET Fluorescence Resonance Energy Transfer
  • Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 ° C.
  • a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then nine successive doubling dilutions were carried out with an equal volume of DMSO.
  • reaction product was measured by the magnitude of fluorescence at a 328 nm excitation wavelength and a 393 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device).
  • concentration of a synthetic compound inhibiting 50% of cathepsin D activity, Ie., IC 50 and Ki were determined by comparing the measurement value with that of a control group with no addition of the synthetic compound.
  • Experimental Example 3 Secreted alkaline phosphatase (SEAP) activity assay Step A: Establishment of permanent cell line expressing SEAP-APPsw-KK A gene which expresses SEAP and Swedish mutant form of APP (CRE-SEAP- APP695sweKK) under the control of cAMP response element (CRE) was cloned into pcDNA3.1(+)Neo (Invitrogen) which is a mammalian expression vector.
  • Neuro-2a cells ATCC Accession No. CCL-131 were cultured in a Dulbecco's minimum essential medium (DMEM, GIBCO-BRL) supplemented with 10% FBS, and then transferred to a 6-well culture plate.
  • DMEM Dulbecco's minimum essential medium
  • the cells When the cells reached confluence, they were transfected with the CRE-SEAP- APP695sweKK expression vector using Lipofectamine 2000 (Life Technologies). Individual clones were isolated and then cultured again on a 6-well culture plate. Following selection of clones, 100 clones showing acceptable growth rates were cultured on a 24-well culture plate for 3 days, followed by culture in DMSO/10 uM Forskolin medium for 6 hours. 50 / ⁇ /well of the culture was aliquoted and reacted with 50 ⁇ l of AttoPhos (Promega).
  • the magnitude of fluorescence was measured at room temperature for 30 min, at a 450 nm excitation wavelength and a 580 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device).
  • 4 clones showing high SEAP activity in forskolin/DMSO and exhibiting the greatest SEAP activity by forskolin were selected.
  • the selected clones were sub-cultured on a 96- well culture plate for one day at an equal cell density (2 X 10 4 cells/96-well), followed by treatment with DMSO/10 ⁇ M Forskolin. After 6 hours, activity of SEAP secreted into the medium was assayed. Clone #159 showing the highest activity of SEAP was selected.
  • Step B SEAP activity assay
  • the clone N2A SEAP-APPsw-KK #159 expressing CRE-SEAP-APP695sweKK was seeded at a density of 2 X 10 4 cells/80 ⁇ JL to each well of a 96-well culture plate. 1OmM Forskolin and DMSO were diluted 100 times with the culture medium and then 10 ⁇ t/weil of the dilution was added to the 96-well plate. Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 ° C.
  • a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then seven successive 3-fold dilutions were carried out with an equal volume of DMSO. 10 ⁇ i of the serial compound dilution was diluted 10 times with 90 ⁇ i of the culture. DMSO was adjusted to a final concentration of 1.1% and the inhibitor was treated through seven successive doubling dilutions from 100 ⁇ M. After treatments were complete, the cells were cultured in a 6% CO 2 incubator at 37 ° C for 5 hours. In order to measure an amount of SEAP secreted into the medium, the culture was first subjected to heat inactivation at 65 ° C for 30 min to thereby abolish activity of other alkaline phosphatases.
  • mice from interbreeding of transgenic (APP/PSldE9) male and female mice were included in experiments.
  • the brains of 3 or 4-day-old mice were dissected, and the hippocampus tissue and cerebral membrane were removed at 4 ° C and minced, followed by treatment with DNase (Sigma, D5025) and protease (Sigma, P5147) and placement in an incubator at 37 ° C for 20 to 25 min.
  • 4 X 10 5 cells/well were seeded onto a 24- well plate coated with poly-L-lysine and containing a medium (27.6 mL Neurobasal + 1.5 mL FBS + 600 ⁇ i B27 + 300 ⁇ i of 200 mM L-glutamine). The cells were cultured in an incubator at 37 "C for 7 days, and then used for subsequent experiments.
  • Step B AMO activity assay Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 ° C . For determination of activity, a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then six successive 3-fold dilutions were carried out with an equal volume of DMSO. The serial compound dilution was diluted 250 times with a medium (29.1 mL Neurobasal + 600 ⁇ i of B27 + 300 ⁇ i of 200 mM L-glutamine + 7.5 ⁇ i of 100 mM L- glutamate).
  • beta-amyloid peptide secreted into the medium was measured by sandwich ELISA (Biosource, #KHB3482) using two types of antibodies specific for the beta-amyloid peptide, which will be briefly illustrated below.
  • the inhibition degree of intracellular beta-secretase activity by synthetic compounds can be determined by using a cell line producing a beta-amyloid peptide from an amyloid precursor protein (APP).
  • APP amyloid precursor protein
  • Step A Establishment of permanent cell line expressing amyloid precursor protein (APP)
  • a mutant form of APP gene (APP75 INFEV), expression of which is under the control of Tet-response element (TRE), was cloned into a PBI-L vector (ClonTech) as a mammalian expression vector that expresses a luciferase gene under the control of Tet-response element (TRE).
  • Neuro-2a cells (ATCC Accession No. CCL- 131) were cultured in a Dulbecco's minimum essential medium (DMEM, GIBCO-BRL) supplemented with 10% FBS, and then transferred to a 6-well culture plate.
  • DMEM Dulbecco's minimum essential medium
  • the cells When the cells reached confluence, they were transfected with the pBI-L APP751 NFEV expression vector using Lipofectamine 2000 (Life Technologies). Individual clones were isolated and then cultured again on a 6-well culture plate. Following selection of clones, 100 clones showing acceptable growth rates were cultured on a 96-well culture plate for 1 day. The culture medium was replaced with a medium containing 1 ⁇ g/mL of doxycycline, followed by culture for 24 hours. 50 ⁇ of Bright-Glo luciferase reagent (Promega) was added to each well which was then left at room temperature for 15 min. Then, luminescence of each well was measured using a luminometer (Victor).
  • Victor luminometer
  • clones showing the highest expression level of luciferase were sub-cultured on a 24-well culture plate for one day at an equal cell density (3 X 10 5 cells/mL/24-well).
  • the culture medium was replaced with Opti-MEM (GIBCO-BRL) containing 1 ⁇ g/mL of doxycycline, followed by culture for 24 hours.
  • An amount of the beta- amyloid peptide secreted into the medium was measured using an ELISA method using antibodies specific for the beta-amyloid peptide.
  • clone #79 showing the highest growth rate and beta-amyloid peptide expression level was selected.
  • Step B ELISA assay of water-soluble beta-amyloid precursor protein (sAPP)
  • IC 50 concentration of a synthetic compound inhibiting 50% of intracellular beta-secretase activity, i.e., IC 50 was determined by comparing the measurement value with that of a control group treated with 1 % DMSO and with no addition of the synthetic compound.
  • Beta-amyloid was carried out by ELISA using two types of antibodies (Human beta amyloid 1-40 colorimetric immunoassay kit, Biosource, California, USA). Two antibodies used in ELISA were an antibody that specifically recognizes the N-terminus of beta- secretase-cleaved beta amyloid and an antibody that binds to the C-terminus thereof. Two different antibodies and the beta amyloid protein were reacted at room temperature for 3 hours (or one or more days at 4°C). The plate was washed four times with a wash solution, and reacted with HRP- conjugated anti-rabbit IgG antibodies for 30 min.
  • the plate was washed four times with a wash solution, and tetramethylbenzidine as the HRP substrate was added, followed by reaction at room temperature for 30 min.
  • the optical density (absorbance) was measured at 450 nm using a microplate reader (SpectraMax 340, Molecular Device). Reduction of beta amyloid was determined by comparing the measurement value with that of a control group.
  • Assay of sAPPbeta secreted amyloid precursor protein beta was also carried out analogously to Hie procedure described in ELISA of beta-amyloid.
  • beta-secretase In order to investigate whether the activity of beta-secretase was inhibited, the inhibition degree of production of beta amyloid which is a beta-secretase cleavage product was examined in animals.
  • the animals used in the experiments were transgenic mice harboring both a Swedish mutant form of beta-APP (chimeric mouse/human amyloid precursor protein 695swe) and a mutant form of presenilin 1 (presenilin l-dE9) (Jankosky JL et al., Biomolecular engineering, 17(6), 157- 165, 2001).
  • the beta-secretase inhibitor was administered at a dose that is expected to result in a decrease of the amyloid protein via an intraperitoneal or subcutaneous route but does not cause toxicity.
  • the animals were anesthetized at a given time point, and blood and cerebral tissues were isolated.
  • the blood was collected in a heparinized tube by cardiac puncture and centriiuged at 13,000 rpm for 10 min (Eppendorf) to separate the plasma
  • the plasma was stored together with the excised brain tissues (cerebral cortex and hippocampus) at 80 ° C until use.
  • the plasma was diluted 5 times, and inhibitory effects on amyloid formation were examined by ELISA as described above.
  • a 4-fold volume of PBS was added to the excised brain tissues which were then homogenized with a sonicator. The homogenate was reacted with guanidine at room temperature for 4 hours.
  • guanidine was adjusted to a final concentration of 5M using 8.2 M guanidine/82 mM Tris HCl (pH 8.0).
  • the beta amyloid protein was extracted and diluted 1:500 in BSAT-DPBS (Dulbecco's phosphate buffered saline with 5% BSA and 0.03% Tween-20), followed by analysis.
  • BSAT-DPBS Dulbecco's phosphate buffered saline with 5% BSA and 0.03% Tween-20
  • a drug compound was dissolved in 10% hydroxypropyl-beta-cyclodextrin (HPCD) and was then administered to a subject, typically at a dose of 15 to 100 mg/kg/day, once or three to five times.
  • HPCD hydroxypropyl-beta-cyclodextrin
  • the compounds of the present invention had a Ki value of about 10 to 100000 nM, preferably 10 to about 1000 nM, and more preferably 10 to 100 nM.
  • IC 50 for SEAP was in the range of 100 to 100000 nM, and preferably 100 to 10000 nM.
  • the compound of Example 15 had a Ki value of 24 nM, IC 50 of 1500 nM for SEAP, and 83-fold higher selectivity for cat. D.
  • compounds of Formula (I) in accordance with the present invention exhibit excellent inhibitory effects on human beta-secretase. Therefore, these compounds can be used as drugs for improvement of cognitive functions or for prevention and treatment of neurodegenerative diseases such as Alzheimer's disease.

Abstract

Disclosed are compounds represented by Formula (I) as defined in the specification, or pharmaceutically acceptable salts or isomers thereof, and a pharmaceutical composition for inhibiting beta-secretase activity comprising a therapeutically effective amount of the same.

Description

BETA-SECRETASE INHIBITING COMPOUNDS HAVING OXO-
DIHYDRO-PYRAZOLE MOIETY
FIELD OF THE INVENTION
The present invention relates to a novel compound for inhibiting beta-secretase activity or a pharmaceutically acceptable salt or isomer thereof, preparation thereof, and a pharmaceutical composition comprising a therapeutically effective amount of the same.
BACKGROUND OF THE INVENTION
Alzheimer's disease (AD), also known as senile dementia, is a neurodegenerative disease which gradually progresses with age and accounts for 50 to 70% of dementia patients. The major symptoms of AD include memory loss, decline in cognitive-reasoning abilities and the like. Most Alzheimer's cases occur around age 65 and progress for about 9 years to result in the death of Ihe patients. The number of AD patients is increasing as society develops and ages, so that it is estimated that there will be approximately 6 million patients within 10 years in the USA and this number will further increase beyond that.
Likewise, the proportion of the elderly population is also increasing in Korea, and thus social problems associated with increased prevalence of senile dementia are becoming more serious. Unfortunately, no therapeutic agent has yet been developed which can treat the underlying pathogenic causes of Alzheimer's disease. Until now, acetylcholine esterase inhibitors have been exclusively used as a general therapeutic agent. Aricept™ (Pfizer), Exelon™ (Novartis), and Reminyl™ (Janssen) are known as representative examples of the acetylcholine esterase inhibitors. Strictly speaking, these drugs cannot be defined as therapeutic agents of Alzheimer's disease, because they do not address the root cause of the disease and result in only partial recovery from the disease in some patients (about 40-50%), and their therapeutic effects are seen for a limited time period only. In addition, the intrinsic nature of the disease requires long-term administration of the drugs, but administration of these therapeutic drugs is accompanied by various adverse side effects including liver toxicity.
Therefore, there is an urgent need for development of a novel therapeutic agent that can halt and reverse the progression of Alzheimer's and related diseases. As an approach to find a pathogenic cause responsible for Alzheimer's disease, long-term studies have been undertaken on familial early-onset Alzheimer's disease patients. As a result, it was revealed that mutations in some genes are primarily responsible for the pathogenesis of Alzheimer's disease.
As the common physiological results of the above-mentioned genetic pathogenic factors, the production of beta amyloid consisting of 42 amino acid residues (hereinafter, sometimes referred to as "Ab42") was found to increase. Based on this finding, it is assumed that the production of Ab42 is a main causative agent of Alzheimer's disease pathogenesis. Therefore, if a method capable of decreasing the production of Ab42 is developed, it will be a direct and effective therapeutic measure capable of blocking a pathogenic mechanism of Alzheimer's disease.
Beta-amyloid proteins are produced from a high-molecular weight amyloid precursor protein (APP) found in neuronal cells through serial cleavage events by 3 types of proteases (secretases). This process takes place at the Golgj apparatus of neuronal cells, wherein APP and secretases are anchored in the Golgi membrane. The N-terminus of beta-amyloid (Ab) corresponds to the 99th amino acid from the C-terminus of APP, and this site is cleaved by beta-secretase (hereinafter, sometimes referred to as "beta-site APP cleaving enzyme" or "BACE"). The C- terminus of the membrane-bound beta-amyloid (Ab) is cleaved by gamma-secretase to generate a beta-amyloid (Ab) protein which is then secreted extracellυlaiy from neuronal cells. Alternatively, APP may also be cleaved at different sites via an alternative pathway; for example, where the middle site of Ab (between 16th and 17th amino acid residues from the N-terminus) is cleaved by alpha- secretase, sAPP alpha having a high molecular weight is produced and secreted. This alternate pathway precludes the formation of beta-amyloid.
Cleavage of APP by the action of BACE and gamma-secretase may result in a variety of beta amyloid (Ab) proteins having different lengths, usually a 40-amino acid fragment (AMO) and a 42-amino acid fragment (Ab42). In comparison with Ab40, Ab42 tends to easily aggregate and accelerates the formation of amyloid plaques in the brains of diseased patients, thereby resulting in gradual necrosis of the surrounding neuronal cells. This is assumed to be a major pathogenic mechanism of Alzheimer's disease.
Ab40 and Ab42 are produced in a ratio of about 9: 1 under normal conditions. However, it is known that where levels of two amyloid proteins AMO and AM2 are increased or where levels of AM2 are selectively increased by mutations of Presenilin 1 and 2 genes, the onset of Alzheimer's disease is further accelerated and the symptoms of the disease are more severe. Therefore, it can be said that lowering of AM2 production is the most important factor for development of anti- Alzheimer drugs. For this purpose, there is a need for development of beta- or gamma-secretase inhibitors.
Therefore, many multinational pharmaceutical companies have been investing heavily in research and development in the secretase inhibitor field, especially in the area of beta- or gamma- secretase inhibitors capable of suppressing the production of 42-amino acid beta-amyloid fragments, which form plaques assumed to be an underlying pathological cause of Alzheimer's disease. Unfortunately, there are no noteworthy results of studies in terms of the development of a therapeutic agent that is capable of arresting a mechanism of beta-amyloid production to directly prevent the progression of the concerned disease.
The beta- and gamma-secretases are known as aspartic proteases and found in the membrane-bound form. However, no gene coding for gamma-secretase has yet been identified. Further, it is known that substrates for gamma-secretase are not limited to APP, but that the enzyme also participates in the cleavage of Notch proteins, which are known to play crucial roles in regulating cell fate decision during differentiation processes. In particular, gene-knockout animals from which a gamma-secretase gene has been deleted died in utero, and recent clinical tests on gamma-secretase inhibitors showed significant drug toxicity. For these reasons, gamma-secretase inhibitors are not likely to be promising drug candidates. As a result, it has not yet been confirmed whether gamma-secretase inhibitors can be developed as safe anti-Alzheimer medications.
On the other hand, in case of BACE, the gene thereof was identified through various methods by many pharmaceutical companies and the in vivo activity of beta-secretase was reported in 1999. Furthermore, the X-ray crystal structure of beta-secretase was determined and a peptide inhibitor having high affinity for beta-secretase was also known in the art. As well, it was reported that a gene-knockout animal with deletion of the BACE gene exhibits a normal phenorype, thus suggesting that a BACE-specific inhibitor can be developed as a safer and efficient anti-dementia drug. Taken altogether, these findings indicate that BACE is a more feasible target for the treatment of Alzheimer's disease, as compared to gamma-secretase. As discussed above, upon considering that conventional commercially available drugs merely exhibit palliative effects of disease symptoms, thus providing substantially no effects on the progression of the concerned disease, it is certain that development of the BACE-specific inhibitors will lead to the development of a novel and remarkable drug against Alzheimer's disease. In recent years, many pharmaceutical companies have published study results on BACE inhibitors, for example by Merck (WO 2006/078577, WO 2006/060109 and WO 2006/057983), Elan (WO 2004/022523 and WO 2005/095326), Schering-Plough (WO 2006/014762 and WO 2006/014944), BMS (WO 2005/182105 and WO 2005/030758), and Eli-Lilly (WO 2005/108358 and WO 2006/034093). In Hie beginning, Elan has reported BACE inhibitor compounds, which were compounds having a small molecular weight and low blood-brain barrier (BBB) permeability. However, these compounds exhibited various limitations due to selectivity for cathepsin D (Cat. D) or in vivo cytotoxicity. A great deal of attention has been recently focused on Merck compounds. Through early introduction of dicarbonyl compounds and subsequently sulfonamide compounds from fellow pharma giant Sunesis, Merck reported excellent inhibitory effects and selectivity of the drug compounds. Since then, numerous drug companies issued various compounds in the form of Merck compounds.
Due to excellent BACE inhibitory effects and high Cat. D selectivity, these compounds, particularly sulfonamide compounds were expected to provide the possibility that they may be developed as an anti-Alzheimer drug. However, their low oral applicability and poor BBB permeability are obstacles to development of the drug. For these reasons, there are very little reports on compounds that lower the production of Ab42 in practical animal models. Apart from sulfonamide compounds, Lilly reported efficacy of drug compounds (lowering of Ab42 levels) in a practical animal model, through S.C administration of peptidomimetic compounds having a low molecular weight. However, these peptidomimetic compounds still suffer from disadvantages such as low oral applicability and poor BBB permeability. Therefore, there is no report yet on compounds exerting excellent effects. Schering-Plough also reported peptidic compounds, but these compounds failed to show desired efficacy in the brain of a practical animal model as they were known to be a Pgp substrate. That is, peptidic beta-secretase inhibitors exhibit limited applicability as oral preparations, due to their intrinsic molecular specificities. In particular, sulfonamide type compounds also have poor BBB permeability which makes it difficult to observe desired efficacy of the drug in an animal model. To this end, there is a strong need for development of a compound having excellent beta- secretase inhibitory activity, distinctively different from sulfonamide compounds.
SUMMARY OF THE INVENTION
Therefore, the present invention has been made to solve the above problems and other technical problems that have yet to be resolved.
More specifically, the present invention is intended to improve blood-brain barrier (BBB) permeability of a drug compound which is a technical problem suffered by conventional sulfonamide compounds.
Therefore, an object of the present invention is to provide a novel compound having beta- secretase inhibitory activity through five-membered heterocyclic compounds apart from sulfonamide in terms of a chemical structure, and pharmaceutically acceptable salts and isomers thereof.
It is another object of the present invention to provide a method for preparing the said compound. It is a further object of the present invention to provide a composition for inhibiting beta- secretase activity, comprising a therapeutically effective amount of the said compound as an active ingredient. It is yet another object of the present invention to provide a use of the said novel compound for improvement of cognitive functions or treatment or prevention of neurodegenerative diseases such as Alzheimer's disease.
In accordance with an aspect of the present invention, the above and other objects can be accomplished by the provision of a compound represented by Formula (T):
Figure imgf000008_0001
ris l to 3; R1 is selected from the group consisting of hydrogen, alkyl, alkene, and -(CH2)P-A-R7 wherein p is 0 to 2, A is aryl or heteroaryl, and R7 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, -NHC(O)-alkyl and aryloxy;
R2 is selected from the group consisting of hydrogen, alkyl, alkoxy and -(CR8R^)P-R10 wherein p is 0 to 2, R8 and R9 are each independently selected from the group consisting of hydrogen, alkyl and alkoxy, and R10 is selected from the group consisting of cycloalkyl, aryl, heterocycle and heteroaryl; and
B is any one of substituents represented by Formulae (i) to (iii):
Figure imgf000008_0002
Figure imgf000009_0001
(ϋi) wherein m and n are each independently 0 or 1 , X is C or N,
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl and -(O)Q-(CH2)P-R11 wherein p is 0 to 2, q is 0 or 1 , and R11 is aryl or heteroaryl,
R5 is selected from the group consisting of hydrogen, alkyl, alkoxy and -(0^-(CH2)P-R1 ' wherein p is 0 to 2, q is 0 or 1 , and R1 ' is aryl or heteroaryl, and R6 is selected from the group consisting of hydrogen, alkyl, alkoxy, and -(CH2)p-A'-R12 wherein p is 0 to 2, A' is phenyl, heteroaryl, or a bicyclic compound containing phenyl, R is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, alkoxy, NHR13 and NR13R14 wherein RI3and R14 are each independently hydrogen or alkyl, or R13and R14 are cyclized to form alkylene; or a pharmaceutically acceptable salt or isomer thereof. The alkyl, alkoxy, aryl, cycloalkyl, heterocycle, and heteroaryl may be substituted or unsubstituted. When substituted, the substituent may be at least one selected from the group consisting of halogen, amino, alkylamino, dialkylamino, alkylacylamino, Ci-C4 alkyl, hydroxy, Q- C4 alkyl alkoxy, aryl alkoxy and oxo. Where appropriate, these substituents may also be substituted. When substituted, the substituents are as exemplified above and the substituents may be taken together to form a cyclic structure.
Further, the heteroaryl and heterocycle are each independently a 4 to 8-membered ring containing 1 to 3 hetero atoms selected from the group consisting of O, N and S, and having 0 to 3 double bonds, and preferably a 5 or 6-membered ring having 1 to 2 double bonds.
Compounds of Formula (I) in accordance with the present invention have a chemical structure that is distinctly different from that of conventional known sulfonamide- or peptide-based beta-secretase inhibitor compounds. As will be illustrated in Experimental Examples which will follow, the compounds of the present invention exhibit improved BBB permeability to thereby exert excellent inhibitory effects on human beta-secretase which is correlated with improvement of cognitive functions or prevention and treatment of neurodegenerative diseases such as Alzheimer's disease.
As used herein, the term "isomer" means a compound of the present invention or a salt thereof that has the same chemical formula or molecular formula but is optically or sterically different therefrom.
Unless otherwise indicated, the term "compound of Formula (T)" is intended to encompass compounds of Formula (1) per se as well as pharmaceutically acceptable salts and isomers thereof.
The term "alkyl" means an aliphatic hydrocarbon group. The alkyl moiety may be a "saturated alkyl" group, which means that it does not contain any alkene or alkyne moiety. The alkyl moiety may also be an "unsaturated alkyl" group, which means that it contains at least one alkene or alkyne moiety. An "alkene" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond, and an "alkyne" moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond. The alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic. The alkyl group may have 1 to 20 carbon atoms. The alkyl group may also be a medium- sized alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. By way of example only, "Ci-C4 alkyl" indicates that there are one to four carbon atoms in the alkyl chain, Ie., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. When the alkyl group is used alone or in combination with alkyloxy, it may be a linear or branched hydrocarbon radical.
Typical examples of the alkyl group may include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, t-butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, cyclopropyl, cycloburyl, cyclopentyl, and cyclohexyl. The term "alkoxy" refers to oxo alkyl having 1 to 10 carbon atoms.
The term "cycloalkyl" refers to an unsaturated aliphatic 4 to 10-membered ring, including cyclohexyl.
The term "aryl" refers to an aromatic group which has at least one ring having a conjugated pi (π) electron system and includes both carbocyclic aryl (for example, phenyl) and heterocyclic aryl (for example, pyridine) groups. This term is intended to include monocyclic or fused-ring polycyclic (Ie., rings which share adjacent pairs of carbon atoms) groups. Specifically, aryl means a 4 to 10-membered, preferably 6 to 10-membered aromatic monocyclic or multicyclic group, including phenyl, naphthyl, and the like.
The term "heteroaryl" refers to a 4 to 8-membered ring having 1 to 3 hetero atoms selected from the group consisting of N, O and S and 0 to 3 double bonds. The heteroaryl may be preferably an aromatic 4 to 8-membered ring and more preferably a 5 or 6-membered ring, which may be fused with benzo or C3-C8 cycloalkyl. Examples of monocyclic heteroaryl may include, but are not limited to, thiazole, oxazole, thiophene, furan, pyrrole, imidazole, isόxazole, pyrazole, triazole, thiadiazole, tetrazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, and the like. Examples of tricyclic heteroaryl may include, but are not limited to, indole, indoline, benzothiophene, benzofuran, benzimidazole, benzoxazole, benzisoxazole, benzothiazole, benzothiadiazole, benzotriazole, quinoline, isoquinoline, purine, furopyridine and the like.
The term "heterocycle" refers to a 3 to 10-membered ring which has 1 to 3 hetero atoms selected from the group consisting of N, O and S, may be fused with benzo or C3-Cg cycloalkyl, and contains 1 or 2 double bonds or may be saturated. The heterocycle may preferably be a 4 to 8- membered ring, and more preferably a 5 or 6-membered ring, which may include, but are not limited to, piperidine, morpholine, thiomorpholine, pyrrolidine, imidazolidine, tetrahydrofuran, piperazine, and the like. Other terms used herein can be interpreted as having their usual meanings in the art to which the present invention pertains.
In one preferred embodiment of the present invention, in the compound of Formula (T), ris 1 to 3;
R1 is selected from the group consisting of hydrogen, C1-C6 alkyl, C2-C6 alkene and - (CH2)P-A-R7 wherein p is 0 to 2, A is 6 to 10-membered aryl or 5 or 6-membered heteroaryl, and R7 is selected from the group consisting of hydrogen, halogen, C1-C3 alkyl, C]-C3 alkoxy, NHC(O)- (C)-C6 alkyl) and 5 or 6-membered aryloxy;
R2 is selected from the group consisting OfCi-C6 alkyl, Ci-C6alkoxy and -(CR8RV-R10 wherein p is 0 to 2, R8 and R9 are each independently hydrogen or Ci-C4 alkyl, and R10 is selected from the group consisting of 5 or 6-membered cycloalkyl, 6 to 10-membered aryl, and 5 or 6- membered heteroaryl; and
B is any one of substituents represented by Formulae (i) to (iii):
Figure imgf000013_0001
(iii) wherein m and n are each independently 0 or 1 ,
X is C or N,
R3 and R4 are each independently selected from the group consisting of hydrogen, C1-C6 alkyl and -(O)q-(CH2)p-Ru wherein p is 0 to 2, q is 0 or 1, and R11 is phenyl or 5 or 6-membered heteroaryl, R5 is selected from the group consisting of hydrogen, Ci-C6 alkyl, Ci-C6alkoxy and -(O)q-
(CH2)P-R1 ' wherein p is 0 to 2, q is 0 or 1 , and R1 ' is phenyl or 5 or 6-membered heteroaryl, and
R6 is selected from the group consisting of hydrogen, Cj-C6 alkyl and -(QHyp-A'-R12 wherein p is 0 to 2, A' is phenyl, 5 or 6-membered heteroaryl, or a bicyclic compound containing phenyl, and R12 is selected from the group consisting of hydrogen, halogen, hydroxy, C1-C6 alkyl, Q-Cόalkoxy, NHR13 and NR13R14 wherein R13and R14 are each independently hydrogen or Q-C4 alkyl, or R13and R14 may be cyclized to form alkylene. In Formula (I), the number of R1 may be in a range of 1 to 3, and R1 may be positioned at carbon and/or nitrogen of the pyrazole ring.
Specifically as in Formula (I'), R1 in the compound of Formula (T) may consist of R\ substituted at N on position 2 and R^ substituted at C on position 4 of the pyrazole ring.
Figure imgf000014_0001
In one preferred embodiment of the present invention, R1 is selected from the group consisting of hydrogen, Cj-C4 alkyl, C2-C4 alkene and -(CH2)p-A-R7 wherein p is 0 to 2, A is phenyl or pyridine, and R7 is selected from the group consisting of hydrogen, halogen, trifluoromethyl, trifluoromethoxy, acetylamino and 5 or 6-membered aryloxy, particularly preferably hydrogen, methyl, hydroxyethyl, vinyl, phenyl, 4-chlorophenyl, 4-trifluoromethyloxyphenyl, 3- trffluoromethylphenyl, 3-trifluorophenyl, 4-trifluoromethylphenyl, 2-trifluoromethylphenyl, benzyl, pyridin-2-yl, 3-phenoxyphenyl and 2-acetylaminophenyl. Preferably, R2 is Q-Qalkoxy or -(CR8RV-R10 wherein p is 0 to 2, R8 and R9 are each independently hydrogen or Ci-C4 alkyl, and R10 is selected from the group consisting of 5 or 6- membered cycloalkyl, 6 to 10-membered aryl and 5 or 6-membered heteroaryl. More preferably, R2 is selected from the group consisting of cyclohexyl, phenyl, 3,5-difluorophenyl and naphthyl.
R3 is preferably selected from the group consisting of hydrogen, Ci-C6 alkyl and [-(O)q- (CH2)P-R11] wherein R11 may be phenyl or 5 or 6-membered heteroaryl. R3 is more preferably hydrogen, benzyloxy or benzyl. R4 is preferably selected from the group consisting of hydrogen, C1-C4 alkyl and O- (CH2)P-R11 wherein R11 may be phenyl or 5 or 6-membered heteroaryl. R4 is more preferably selected from the group consisting of hydrogen, methyl, isobutyl and benzyloxy.
R5 is preferably selected from the group consisting of hydrogen, C1-C6 alkyl and -O-R11 wherein R11 may be phenyl or 5 or 6-membered heteroaryl. R5 is more preferably hydrogen or phenoxy.
R6 is preferably selected from the group consisting of hydrogen, C1-C4 alkyl and -A' -R12 wherein A' is phenyl, 5 or 6-membered heteroaryl, or a bicyclic compound containing phenyl, and R12 is selected from the group consisting of hydrogen, halogen, hydroxy, Ci-C6 alkyl, Ci-C6 alkoxy, NHR13 and NR13R14 wherein R13and R14 are each independently hydrogen or Ci-C4 alkyl, or R13and R14 are cyclized to form alkylene. More preferably, R6 is preferably selected from the group consisting of hydrogen, methyl, 3-dimethylaminophenyl, 3-(t-butyl)-phenyl, (3-isopropyl-[l,2,4]- oxadiazol-5-yl), 3-(pyrrolidin-l-yl)phenyl, and (l-hydroxy-indan-5-yl).
The compound in accordance with the present invention may form a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salt" means acid addition salts of the compound with acids capable of forming a non-toxic acid addition salt containing pharmaceutically acceptable anions, for example, inorganic acids such as sulfuric acid, hydrochloric acid, nitric acid, phosphoric acid, hydrobromic acid and hydroiodic acid; organic carboxylic acids such as tartaric acid, formic acid, citric acid, acetic acid, trichloroacetic acid, trifluoroacetic acid, gluconic acid, benzoic acid, lactic acid, fiimaric acid, and maleic acid; or sulfonic acids such as methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid and naphthalenesulfonic acid. These acid addition salts may be prepared by conventional techniques known in the art, based on the chemical structure of Formula (I). As used herein, the term "isomer" means a compound of the present invention or a salt thereof, that has the same chemical formula or molecular formula but is optically or stereochemically different therefrom. For examples, the compounds of Formula (I) in accordance with the present invention may have an asymmetric carbon center, and therefore can be present in the form of optical isomers (R or S isomeric forms), racemates, diastereomeric mixtures, and individual diasteromers. When the compound of the present invention has a double bond, there may be present geometrical isomers (trans and cis isomeric forms). The present invention encompasses all these isomeric forms and mixtures.
Unless otherwise specified, it should be understood that the compound of Formula (I) in the context of the present invention is intended to encompass pharmaceutically acceptable salts and isomers thereof, all of which are included within the scope of the present invention. For convenience of illustration, the compound of the present invention is simply expressed as a compound of Formula
O)-
Representative examples of the compound of Formula (T) in accordance with the present invention include the following compounds:
N-[(2S,3R)4-[(3-dime%laminophenyl)me%lamino]-3-hydroxy-l-phenyl butan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide;
N-[(2S,3R)-l-(3,5-difluorophenyl)-4-[(3-dime%laminophenyl)me%lamino]-3- hydroxybutan-2-yl]-5-oxo- 1 -phenyl4H-pyrazole-3-carboxamide; l-(4-chlorophenyl)-N-[(2S,3R-l-(3,5-difluorophenyl)-4-[(3- dimethylaminophenyl)methylamino]-3-hydroxybutan-2-yl]-5-oxo-4H-pyrazole-3-carboxami
N-[(lR,2S)-3-(3,5-difluorophenyl)-l-hydroxy-l-[(2S)-pyrrolidin-2-yl]propan-2-yl]-5- oxo- 1 -phenyMH-pyrazole-S-carboxamide; N-[(2S,3R)4-[(3-dime%laminophenyl)me%lamino]-3-hydτoxy-l-phenylbutan-2-yl]-5- oxo-l-[4-(trifluoromethoxy)phenyl]-4H-pyrazx)le-3-carboxamide;
N-[(2S,3R)-4-[(3-dimethylaminophenyl)me%lamino]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo-l-[3-(trifluoromethyl)phenyl]-4H-pyrazole-3-carboxamide; N-[(lS,2R)-3-(3,5-difluorophenyl)-l-hydroxy-l-[(2S)-pyrrolidin-2-yl]propan-2-yl]-5- oxo- 1 -phenyl4H-pyrazole-3-cark)xarnide;
N-[(2S,3R)-l-(3,5-dMuorophenyl)4-[(3-dimethylaminophenyl)methylaniino]-3- hydroxybutan-2-yl]-5-oxo- 1 -[3-(1iifluorOmethyl)phenyl]-4H-pyrazole-3-carboxamide;
N-[(lR,2R)-l-[(3R,8aS)-U,3,4,6,7,8,8a-octahydropyrrolo[l,2-d]pyrazb-3-yl]-3-(3,5- difluorophenyl)- 1 -hydroxypropan-2-yl]-5-oxo- 1 -phenyMH-pyrazole-S-carboxamide;
N-[(lS,2S)-l-[(3R,8aS)-l,23,4,6,7,8,8a-()ctahydropyπOlo[l,2-d]pyrazin-3-yl]-3-(3,5- dffluorophenyl)-l-hydroxypropan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide;
N-[(2S,3R)-4-[(3-dimethylaminophenyl)methylamino]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo- 1 -[2-(trifluoromethyl)phenyl]-4H-pyrazole-3-carboxamide; l-b)enzyl-N-[(2S,3R)4-[(3-dime%lammophenyl)methylamino]-3-hydroxy-l- phenylbutan-2-yl]-5-oxo-4H-pyrazole-3-carboxamide;
N-[(lR,2S)-3-(3,5-Muorophenyl)-l-hydroxy-l-[(2R,4R)-4-phenylmethoxypyrrolidin-2- yl]propan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide;
N-[(lR,2S)-3-(3,5-dMuorophenyl)-l-hydroxy-l-[(2R)-pyrroHdin-2-yl]propan-2-yl]-5- oxo-l-phenyMH-pyrazole-S-carboxamide;
N-[(2S,3R)-4-[(3-dime%laminophenyl)me1hylamino]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo- 1 -pyridin^-yMH-pyrazole-S-carboxamide;
N-[(2S,3R)-4-[(3-dime1hylarainophenyl)methylamino]-3-hydroxy-l-phenylbι^ oxo-l-(3-phenoxyphenyl)4H-pyra2Dle-3-carboxamide; N-[(lS,2S)-l-[(3S,8aS)-l-methyl-4-oxo-2,3,6,7,8,8a-hexahydro-lH-pyirolo[l,2- d]pyrazin-3-yl]-3-(3,5-difluorøphenyl)- 1 -hydroxypropan-2-yl]-5-oxo- 1 -phenyl-4H-pyrazole-3- carboxamide; l-benzyl-N-[(2S,3R)-4-[(3-t-butylphenyl)me%lan±io]-3-hyctoxy-l-phenylbutan-2-yl]- 5-oxo4H-pyrazole-3-carboxamide;
N-[(lS^S)-3-(3,5-difluorophenyl)-l-hydrøxy-l-[(3R,7S>7-phenoxy-l,2,3Λ6,7,8,8a- octahydropyrrolo[l,2-d]pyrazm-3-yl]pro^
N-[(lS,2R)-l-hydroxy-3-mphiMen-2-yl-l-[(2R)-pyrrølidin-2-yl]piOpan-2-yl]-5-oxo-l- phenyl-4H-pyrazole-3-carboxamide; N-[(lR,2S)-l-hydroxy-3-mphthden-2-yl-l-[(2R>pyπOlidin-2-yl]piOpan-2-yl]-5-oxo-l- phenyl4H-pyrazole-3-carboxamide;
N-[(2S,3R)4-[(3-dime1hylaniinophenyl)methylarnino]-3-hydroxy- 1 -phenylbutan-2-yl]- 1 - methyl-5-oxo-4H-pyrazole-3-carboxamide;
N-[(2S,3RH-[(3-t-butylphenyl)me%lamino]-l-(3,5-difluorophenyl)-3-hydroxybutan-2- yl]-5-oxo-l-[3-(trifluorømethyl)phenyl]4H-pyrazole-3-carboxamide; l-benzyl-N-[(2S,3R)-4-[(3-t-butylphenyl)me%lamino]-l-(3,5-difluorophenyl)-3- hydroxybutan-2-yl]-5-oxo4H-pyrazole-3-carboxamide;
N-[(2S,3R)4-[(3Klimethylaminophenyl)methylamino]-3-hydroxy-l-phenylbuto^ ethenyl-5-oxo4-methyl-pyrazole-3-carboxaniide; N-[(2S,3R)4-[(3-dime%lammophenyl)me%lamino]-3-hydroxy-l-phenylbutan-2-yl]-l-
(2-hydroxyethyl)-5-oxo4-methyl-pyrazole-3-carboxamide;
N-[(lR,2S)-3-cyclohexyl-l-hydroxy-l-[(2R)-pyrrolidin-2-yl]propan-2-yl]-5-oxo-l- phenyl4H-pyrazole-3-carboxamide; N-[(2S,3R)-3-hydroxy-l-phenyl-4-[(3-piOpan-2-yl-lA4-oxadiazol-5- yl)methylamino]butan-2-yl]-5-oxo-l-phenyl4H-pyra2Ole-3-carboxamide;
N-[(2S,3R)-3-hy(k)xy-l-phenyl-4-[(3-^ oxo- 1 -phenyMH-pyrazole-S-carboxarnide; 1 -(2-acetylaminophenyl)-N-[(2S,3R)-4-[(3-dime%laminophmyl)me1hylaniino]-3- hydroxy- 1 -phenylbutan-2-yl]-5-oxopyrrolidine-3-carboxamide;
N-[(2S,3R)-3-hydroxy-4-[(l -hydroxy-2,3-dihydrø-lH-inden-5-yl)methylamino]-l - phenylbutan-2-yl]-5-oxo-l-phenyl4H-pyrazole-3-carboxamide;
N-[(2S,3R)-l-(3,5-difluoroρhenyl)-3-hydroxy-4-[(3-pyrroUdin-l- ylphenyl)methylamino]butan-2-yl]-5-oxo-l -phenyMH-pyrazole-S-carboxamide;
N-[(lR,2R)-3-(3,5-difluorophenyl)-l-hydroxy-l-[(2R,5S)-5-(2-methylpropyl)piperazin-2- yl]propan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide;
N-[(lS,2S)-3-(3,5-dmuorophenyl)-l-hydroxy-l-[(2R,5S)-5-(2-me%lpropyl)piperazin-2- yl]propan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide; N-[(lR^S)-3-(3,5-cMuorophenyl)-l-hydroxy-l-[(2R)-pyiτolidin-2-yl]propan-2-yl]-5- oxo-1 -phenyMH-pyrazole-S-carboxamide;
N-[(lS,2S)-l-[(2S)4-benzyl-3-oxopiperazin-2-yl]-l-hydrøxy-3-phenylpropan-2-yl]-5- oxo-l-phenyl-4H-pyrazole-3-carboxamide; and
N-[(lR,2S)-3-(3,5-Muorophenyl)-l-hydroxy-l-[(2R,4RH-phenylmethoxypyrrolidin-2- ylJpropan^-yy-S-oxo-l-CS-trifluoromethy^phenyMH-pyrazole-S-carboxamide.
In accordance with yet another aspect of the present invention there is provided a method for preparing a compound of Formula (I). For a better understanding of the present invention, preparation of the compound of Formula (T) will be illustrated with reference to exemplary synthetic Reaction Schemes. However, skilled persons having knowledge about synthesis of compounds in the art to which the present invention pertains could readily prepare the compound of Formula (I) according to various synthetic methods, based on the chemical structure of Formula (I). Therefore, these methods also fall within the scope of the present invention. That is, the compound of Formula (I) may be prepared by any combination of various synthetic methods disclosed herein or in prior art. Therefore, the below description associated with preparation processes is provided only to disclose exemplary ones, and Hie scope of the present invention is not limited to only such preparation processes as described herein.
As shown in Reaction Scheme 1, the compound of Formula (I) can be synthesized by amide-coupling reaction of carboxylic acid of Formula 2 with an amine compound of Formula 3.
[Reaction Scheme 1]
Figure imgf000020_0001
In Reaction Scheme 1 , R1, R2 and B are as defined above. Examples of known coupling agents usable in the amide coupling may include, but are not limited to, carbodiimides such as dicyclohexylcarbodiimide (DCC), 1 -(3-dimethylaminopropyl)- 3-ethylcarbodiimide (EDC), 1 ,1 '-dicarbonyldiimidazolc (CDl), etc. which are used in a mixture with 1-hydroxybenzDtriazole (IIOBT) or l-hydroxy-7-azabenzotriazo]e (HOAT); bis-(2-oxo-3- oxazolidinyl)-phosphinic acid chloride (BOP-Cl), diphenylphosphorylazide (DPPA), N- [dimethylainino-lH-l,2,3-triazole[4,5-b]pyridin-l-ylnie%lene]-N-me%lmetriaiieaniiniLm (HATU)5 CtC. In addition, examples of solvents that can be used in amide coupling may include DCM, DMF, and DCE. Examples of bases for activation of the reaction may include triethylamine, diisopropylethylamine, etc.
The carboxylic acid of Formula 2 is commercially available or can be synthesized by cyclization and hydrolysis from a compound of Formula 4 or 5 and a hydrazine compound of Formula 6, according to the synthetic procedure of Reaction Scheme 2 below.
[Reaction Scheme 2] RO2C-^-E — CO2R
o o + RI-N-NH2 l) Cyclizati?n lOSf OH
.oJUl o H IO Hydrolysis Ri/ N-N J
O 2
5 Cyclization may be carried out by heating reactants in the presence of a base, for example triethyl amine, N-methylmorpholine, diisopropyl ethyl amine, or K2CO3, in a solvent such as DMF,
THF, dioxane or DMSO. The reaction is conventionally carried out at a temperature of 60 to 120 °C for 4 to 24 hours.
Hydrolysis to obtain the compound of Formula 2 is carried out in a mixed solution of water and an organic solvent, using a base. Examples of the organic solvent usable in hydrolysis may include THF, methanol, and dioxane. Examples of the base may include LiOH, KOH, and
NaOH.
The hydrazine compound of Formula 6 is commercially available or may be obtained by conversion of an amine group of an amine compound of Formula 7 into a hydrazine group, according to a known method disclosed in Journal of the American Chemical Society, 198(48),
15374-75, 2006. [Reaction Scheme 3]
Ri-NH2 l) Nitr0Sati0n , Ri-N -NH2 ii) [H] H
7 6
Nitrosation may be carried out in the presence of hydrochloric acid, using NaNO2. Reduction of the nitroso compound may be carried out in the presence of hydrochloric acid, using SnCl2.
Among the amine compounds of Formula 3, a compound where B is in the form of a chain can be synthesized from a commercially available epoxide, according to a synthetic procedure as disclosed in Reaction Scheme 4 below.
[Reaction Scheme 4]
Figure imgf000022_0001
In Reaction Scheme 4, R6 is as defined above, an azide compound of Formula 9 can be obtained by ring-opening of an epoxide compound using NaN3, and an amine compound of Formula 10 can be obtained by reduction of the azide compound through hydrogenation using a
Pd/C catalyst. The reaction is carried out in a solvent such as methanol, ethanol, and dioxane, at 1 to
3 atm. Reductive animation to obtain a compound of Formula 3-1 is carried out using a compound (ketone or aldehyde) containing a carbonyl group. Examples of an available reducing agent may include sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, etc. In order to facilitate the reaction, acid may be used as a catalyst. Examples of an available acid catalyst may include inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid, and phosphoric acid; organic carbonic acids such as acetic acid, and trifluoroacetic acid; and amine salts such as ammonium chloride. Particularly preferred is hydrochloric acid or acetic acid.
Epoxide that is not readily commercially available can be prepared by stereoselective synthesis as shown in Reaction Scheme 5 below.
[Reaction Scheme 5]
Figure imgf000023_0001
11 1122 13
Figure imgf000023_0002
14 15 16 17
In Reaction Scheme 5, R2 is -CH2-R2, and X is a leaving group, such as bromine, iodine, or methanesulfonyl. Stereoselective alkylation is carried out using a chiral auxiliary of Formula 11. Then, hydrolysis of the compound of Formula 12 is carried out to prepare an amino acid derivative of Formula 13. The hydrolysis reaction to obtain the compound of Formula 13 was previously described. The reaction is carried out using iodomethane in the presence of a base to obtain an iodocarbonyl compound of Formula 15. Reduction of the compound of Formula 15 is then carried out to obtain an iodohydrin compound of Formula 16. Reduction is carried out using NaBH4, NaB(CN)H3, or the like. Further, an epoxide compound of Formula 17 can be obtained by cyclization of the iodohydrin compound in the presence of a base.
A heterocyclic compound such as oxadiazole can be prepared according to a synthetic procedure in Reaction Scheme 6 below:
[Reaction Scheme 6]
Figure imgf000024_0001
First, an amidoxime compound of Formula 18 is reacted with oxalate to synthesize an oxadiazole compound of Formula 20, and reduction of the reaction product is then carried out using diisobutylaluminum (DIBAL) to obtain an aldehyde compound of Formula 21.
In Formula (I) in accordance with the present invention, a compound where B was cyclized can be synthesized using a known method disclosed in WO 2006/034093, according to a synthetic procedure in Reaction Scheme 7 below.
[Reaction Scheme 7] H
Figure imgf000024_0002
21 22 23 24
In Reaction Scheme 7, R2 is as defined above, and n is 0 or 1. In Reaction Scheme 7, a nitro compound of Formula 21 and an aldehyde of Formula 22 are reacted under basic conditions to prepare a compound of Formula 23, and a nitro group of Formula 23 is reduced in the presence of hydrogen to obtain a compound of Formula 24.
The nitro compound of Formula 21 can also be synthesized by a known method disclosed in WO2006/034093, according to a synthetic procedure of Reaction Scheme 8.
[Reaction Scheme 8]
Figure imgf000025_0001
25 26 27
D
Figure imgf000025_0002
21
In Reaction Scheme 8, R is as defined above. Aldehyde of Formula 25 and nitromethane are subjected to coupling reaction under basic conditions to obtain a compound of Formula 26 which is then acetylated to prepare a compound of Formula 27. Following dehydroxylation, a compound of Formula 28 is reduced to synthesize a compound of Formula 21.
Aldehyde of Formula 22 can be synthesized from an amino acid.
[Reaction Scheme 9]
)n RedUCtiOn , )n OxidatiOrU )n
Figure imgf000025_0005
Figure imgf000025_0004
Figure imgf000025_0003
29 30 22
An alcohol compound of Formula 30 can be obtained from a compound of Formula 29 using a reducing agent, for example NaBH4 or LAH, in methanol or THF. Then, the alcohol compound is subjected to oxidation to thereby obtain a compound of Formula 22. An oxidant may be SO3Py, and a solvent may be DCM.
Analogously to the procedure described in Reaction Scheme 7, a piperazine compound can be synthesized according to a synthetic procedure of Reaction Scheme 10 below.
[Reaction Scheme 10]
Figure imgf000026_0001
31 32 33 34
Aldehyde of Formula 31 and piperazine of Formula 32 are reacted in the presence of a base to obtain a compound of Formula 33. The deprotection of the Boc group from the compound of Formula 33 is then carried out to obtain a compound of Formula 34.
Meanwhile, a bicyclic compound can be synthesized according to a synthetic procedure of Reaction Scheme 11.
[Reaction Scheme 11]
BnO
Figure imgf000026_0002
I, -Reduction =^ A ii) Oxidation
Figure imgf000026_0003
38 39
In Reaction Scheme 11, R i5 is as defined above, and [RA] means reductive amination. Analogously to the procedure described in Reaction Scheme 9, aldehyde of Formula 36 is synthesized from a serine derivative of Formula 35. Then, a compound of Formula 37 is synthesized through reductive amination of Formula 36. By repeating the same procedure, aldehyde of Formula
38 is obtained, followed by deprotection of the Boc group and reductive amination to obtain a compound of Formula 39.
Separation of a conventional mixture is carried out by column chromatography, whereas a final compound may be separated by recrystallization or normal-phase or reverse-phase HPLC
(Waters, Delta Pack, 300 x 50 mmlD., C18 5 μso, 100A). When recrystallization or HPLC is employed, a desired compound can be obtained in the form of a trifluoroacetate. When it is desired to obtain hydrochloride, an ion exchange resin may be employed.
As described above, after the reaction according to the method of the present invention is complete, the reaction product can be separated and purified by conventional post-treatments, for example, chromatography, recrystallization, and the like.
In accordance with yet another aspect of the present invention, there is provided a pharmaceutical composition for inhibiting beta-secretase, comprising a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt or isomer thereof as an active ingredient and a pharmaceutically acceptable carrier. If desired, the composition may further comprise one or more diluents or excipients.
The compound of Formula (I) exhibits excellent inhibitory effects on beta-secretase, so the present invention provides a beta-secretase inhibitor composition comprising a compound of Formula (J) in conjunction with a pharmaceutically acceptable carrier. In particular, the composition in accordance with the present invention exhibits excellent effects on improvement of cognitive functions or treatment or prevention of neurodegenerative diseases, particularly Alzheimer's disease without being limited thereto. The term "pharmaceutical composition" as used herein means a mixture of a compound of the invention with other chemical components, such as diluents or carriers. The pharmaceutical composition facilitates administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but are not limited to oral, injection, aerosol, parenteral, and topical administrations. Pharmaceutical compositions can also be obtained by reacting compounds with acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
The term "therapeutically effective amount" means that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disease being treated. Thus, a therapeutically effective amount refers to that amount which has the effect of (i) reversing the rate of progress of a disease, (ii) inhibiting or slowing to some extent further progress of the disease, and/or, (iii) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with the disease.
The term "carrier" means a chemical compound that facilitates the incorporation of a compound into cells or tissues. For example, dimethyl sulfoxide (DMSO) is a commonly utilized carrier as it facilitates the uptake of many organic compounds into the cells or tissues of an organism.
The term "diluent" defines chemical compounds diluted in water that will dissolve the compound of interest as well as stabilize the biologically active form of the compound. Salts dissolved in buffered solutions are utilized as diluents in the art. One commonly used buffered solution is phosphate buffered saline because it mimics the salt conditions of human body fluid. Since buffer salts can control the pH of a solution at low concentrations, a buffered diluent rarely modifies the biological activity of a compound.
The term "physiologically acceptable" defines a carrier or diluent that does not abrogate the biological activity and properties of the compound. The compounds described herein can be administered to a human patient per se, or in pharmaceutical compositions in which they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipients. Techniques for formulation and administration of the compounds may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 18th edition, 1990.
a) Routes of Administration
Compounds of the present invention may be administered via any conventional routes, depending upon desired applications. Preferred routes of administration may, for example, include injection, oral and intranasal administrations. Alternatively, the active compounds may be administered by dermal, intraperitoneal, retroperitoneal and rectal routes.
b) Composition/Formulation
The pharmaceutical composition of the present invention may be manufactured in a manner that is itself known, e.g. , by means of conventional mixing, dissolving, granulating, dragee- making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g. , in Remington's Pharmaceutical Sciences, supra.
Injectable preparations, for example aqueous or oily suspensions for sterile injection may be prepared by a conventional method known in the art, using suitable dispersants, wetting agents and/or suspending agents. Examples of solvents that can be used in the formulation of injectable preparations may include water, Ringer's solution, and isotonic NaCl solution. In addition, sterile fixing oil is also conventionally used as a solvent or suspending medium. Any non-irritable fixing oil including monoglyceride and diglyceride may be used for this purpose. In addition, fatty acids such as oleic acid may also be used for injectable preparations.
Examples of solid dosage forms for oral administration may include capsules, tablets, pills, powders and granules. Particularly preferred are capsules and tablets. Tablets and pills may be preferably provided with enteric coatings. The solid dosage form may be prepared by mixing an active compound of Formula (I) in accordance with the present invention with one or more inert diluents (such as sucrose, lactose, and starch) and carriers such as lubricants (such as magnesium stearate), disintegrants, binders, and the like.
c) Effective Dosage
Pharmaceutical compositions suitable for use in the present invention include compositions in which the active ingredient is contained in an amount effective to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. When the compound of the present invention is administered for clinical purposes, a daily dosage of the compound, which may be administered to a subject in a single or divided doses, is preferably in a range of 10 to 100 mg/kg. As will be apparent to those skilled in the art, a specific dose of the active ingredient for individual patients may vary depending on various factors such as kinds of compounds to be used, weight, sex, health conditions, and dietary habits of patients, treatment duration, administration manners, excretion rates, drug mixing and severity of disease.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS Now, the present invention will be described in more detail with reference to the following
Examples. These examples are provided only for illustrating the present invention and should not be construed as limiting the scope and spirit of the present inventioa
Abbreviations ased in the preceding Reaction Schemes and the following Preparations and Examples are as follows:
Bu: butyl i-Bu: isobutyl
Nap: naphthyl
BOC (boc): t-butoxycarbonyl c-Hex: cyclohexyl
Bn: benzyl
DCM: dichloromethane
DMF: N,N-dimethyrformamide
EDC: l-(3-dimethylaminopropyl)-3-ethylcarbodiimide, hydrochloride EtOAc: ethyl acetate
Hex: normal hexane
HOBT: hydroxybenzotriazole
LDA: lithium dϋsopropyl amide Ph: phenyl
TFA: trifluoroaectic acid
Preparation 1: 3-phenoxy-phenyl-l -hydrazine 3-phenoxy-phenylamine (3.7 g, 20 mmol) was dissolved in HCl (6 mL) at 0°C, and a solution of NaNO2(13.8 g, 20 mmol) in H2O (3 mL) was added thereto. The mixture was stirred for 10 min, and a solution of SnCl2 in HCl was added dropwise thereto, followed by stirring for 3 hours. After the reaction was complete, a solid material was filtered, washed with diethyl ester, and dried to afford the title compound (3.36 g, 84 %). Mass[M+l] =201
Preparation 2: S-oxo-l-rS-phenoxyphenylM^-dihydro-l-H-pyrazole-S-carboxylic acid Step A: S-oxo-l-CS-phenoxyphenyllAS-dihydro-l-H-pyrazole^-carboxylic acid methyl ester
3-phenoxy-phenyl-l -hydrazine (4.2 g, 20 mmol) synthesized in Preparation 1 was added to diethyl acetylene diester (3.2 g, 20 mmol) in THF, to which Et3N (2.8 mL, 20 mL) was then added, followed by stirring at 80 "C for 15 hours. After the reaction was complete, the reaction mixture was cooled and neutralized with addition of IN HCl, followed by EtOAc extraction. The organic extract was dried over MgSO4 and distilled under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/5) to afford the title compound (4.91 g, 89%).
Step B: S-oxo-l-G-phenoxyphenvlM^-dihvdiO-l-H-pyrazole-S-carboxvlic acid S-oxo-l-CS-phenoxypheny^^S-dihydro-l-H-pyrazole-S-carboxylic acid methyl ester (4.0 g) obtained in Step A was dissolved in methanol, to which water and LiOH were then added, followed by stirring at room temperature for 3 hours. After the reaction was complete, the reaction mixture was adjusted to pH of 2 to 3 with addition of IN HCl, followed by EtOAc extraction. The organic extract was dried over MgSO4 and distilled under reduced pressure to afford the title compound.
Mass[M+l] =297
Preparations 3 to 10 Analogously to the procedure described in Preparation 2, compounds of Preparations 3 to
10 as listed in Table 3 below and represented by the following general formula were synthesized using commercially available aniline or hydrazine.
Figure imgf000033_0001
[Table 3]
Figure imgf000033_0002
Figure imgf000034_0001
Preparation 11: 4-me1fayl-5-oxo-l-vinyl-4,5-dihvdro-lH-pyra2iole-3-carboxylic acid Step A: 4-metfayl-5-oxo-l-vinyl4,5-dihvdro-lH-pyrazole-3-carboxylic acid methyl ester
The title compound was obtained as a by-product during preparation of the compound of Preparation 10.
Step B: 4-methyl-5-oxo-l-vinyl4,5-dihvdiO-l-H-pyraziole-3-carboxylic acid
Analogously to the procedure described in Step B of Preparation 2, the title compound was synthesized using 4-methyl-5-oxo-l-vinyl-4,5-dihydro-lH-pyrazole-3-carboxylic acid methyl ester obtained in Step A. Mass[M+l] =155
Preparation 12: (nS,2RV3-amino-l-benzyl-2-hydroxypropylVcarbamic acidt-butyl ester
Step A: ((lS2RV3-azido-l-benzyl-2-hvdiOxypropyl)-carbamic acid t-butyl ester
Commercially available epoxide (2.6 g, 10 mmol) was dissolved in a sat'd NH4Cl aqueous solution, and the mixture was stirred in ethanol at 80 "C for 12 hours. After the reaction was complete, the reaction mixture was concentrated under reduced pressure, diluted with EtOAc, washed with IN HCl, and extracted with EtOAc. The organic extract was purified by column chromatography (EtOAc/n-Hex = 1/4) to afford the title compound (2.9 g, 95%). Step B: ((I S,2R>3-amino-l -benzyl^-hydroxypropylVcarbarnic acid t-butyl ester
((lS,2R)-3-azido-l-benzyl-2-hydroxypropyl)-carbaniic acid t-butyl ester (2.0 g, 0.65 mmol) obtained in Step A was placed in a hydrogen reaction vessel, to which 1 g of Pd/C was then added. The reaction mixture was reacted at 50 psi for 4 hours and filtered through celite to afford the title compound (1.10 g, 94%) which was then directly used without further purification. Mass[M+l] =281
Preparation 13: [(lS2RV3-arnino-l-(315-Muoro-bemyl)-2-hydroxy-propyl]-carbarnic acid t-butyl ester Step A: (2S,5RV2,5-dihvdro-3,6-dimethoxy-5-isopropyl-2-(3,5-dMuoro)benzylpyrazine
(TR.)-2,5-dihydro-3,6-dirnethoxy-2-isopropylpyrazine (9.2 g, 50 mmol) was dissolved in THF (100 mL), to which n-BuLi (4M in THF, 15 mL) was then added at -78 °C, followed by addition of difluorobenzyl chloride (9.72 g, 60 mmol). After the reaction was complete, NH4Cl was added to the reaction mixture which was then extracted with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/1) to afford the title compound (12.4 g, 80%).
Step B: (Sy2-amino-3-(3,5-difluorophenyl)propionic acid methyl ester
(2S,5R)-2,5-dihydro-3,6-dimethoxy-5-isopropyl-2-(3,5-dMuoro)ber^lpyrazine (12 g, 38.7 mmol) obtained in Step A was dissolved in acetonitrile (50 mL) and 2N HCl (20 mL) was added thereto, followed by stirring at room temperature for 4 hours. After the reaction was complete, NaHCO3 was added, followed by EtOAc extraction. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was recrystallized from EtOAc/n-Hex to afford the title compound (7.07 g, 85%). Step C: (SV2-t-butoxycarbonylamino-3-G.5-difluorophenyl')piOpionic acid methyl ester
(S)-2-amino-3-(3,5-difluorøphenyl)propionic acid methyl ester (7 g, 32.5 mmol) obtained in Step B was dissolved in water, to which NaOH was then added, followed by addition of (BOC)2O (7.80 g, 35. 7 mmol). 12 hours later, the reaction was terminated with addition of IN HCl, followed by EtOAc extraction. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/8) to afford the title compound (9.89 g, 88%).
Step D: [(SVl-fB^-dMuorobenzylVS-cMoro^-oxo-propyl'lcarbamic acid t-butyl ester
Chloroiodomethane (9.14 g, 31.4 mmol) was dissolved in THF, to which LDA (synthesized from nBuLi and diisopropyl amine, 33 mmol) was then added, followed by addition of (S)-2-t-butoxycarbonylamino-3-(3,5-difluorophenyl)propionic acid meihyl ester (9 g, 28.5 mmol) obtained in Step C. The mixture was stirred at room temperature for 12 hours. After the reaction was complete, NH4Cl was added, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/5) to afford the title compound (9.56 g, 79%).
Step E: [(I S,2SV1 -(S^-difluorobenzyπ^-hydroxy-S-chloro-propyllcarbamic acid t-butyl ester [(S^l^jS-difluorobenzyl^-chloro^-oxo-propyycarbamic acid t-butyl ester (9 g, 21 mmol) obtained in Step D was dissolved in MeOH (60 mL), to which NaBH4 (1.55 g, 42 mmol) was then added, followed by stirring at room temperature for 12 hours. After the reaction was complete, water was added thereto with subsequent addition of IN HCl and stirring for 30 min, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/3) to afford the title compound (8.0 g, 89%).
Step F: [(S)-2-(3,5-difluorophenylVl-(S)-oxiranyl-ethyll-carbamic acid t-butyl ester [(lS^S^l^S^-difluorober^l^-hydroxy-S-chloro-propylJcarbarnic acid t-butyl ester
(6.5 g, 15.2 mmol) obtained in Step E was dissolved in THF (45 mL), to which NaOH (0.6 g, 15.2 mmol) was then added, followed by stirring at room temperature for 12 hours. After the reaction was complete, IN HCl was added thereto, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/5) to afford the title compound (3.40 g, 75%).
Step G: [flS,2R)-3-amino-l-(3,5-α4fluoro-rjeri2^1)-2-hyαVoxy-propyl]-carbarnic acid t-butyl ester
Analogously to the procedure described in Steps A and B of Preparation 12, the title compound was obtained using [(S)-2-(3,5κMuorophenyl)-l-(S)-oxiranyl-ethyl]-carbarnic acid t- butyl ester obtained in Step F. MS[MH] =217 (M+l)
Preparation 14: 3-fdimethylairiino)berizaldehyde
Commercially available 3-(dimethylamino)benzyl alcohol (750 mg, 5 mmol) was dissolved in DCM and DMSO, to which Et3N (2.8 mL, 20 mmol) and SO3Py (1.59 g, 10 mmol) were then added. The reaction solution was stirred at room temperature for 2 hours. After the reaction was complete, DCM was distilled under reduced pressure and a saturated NH4Cl aqueous solution was added, followed by extraction with ethyl ester. The organic extract was washed with water, dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/2) to afford the title compound (670 mg, 90%). MS[MH-I] = 150 (M+l)
Preparation 15: 3-(t-butyl)benzaldehyde
3-(t-butyl)phenylbromide (1.07 g, 5 mmol) was dissolved in THF, to which n-Buli (4M in
THF, 1.5 mL) was then added, followed by addition of DMF and stirring at 0°C for 3 hours. After the reaction was complete, NH4Cl was added thereto, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 1/3) to afford the title compound (680 mg,
H O).
MS[MH-I] = 163 (M+l)
Preparation 16: 3-isopropyl-5-formyl-oxadiazole Step A: Methyl 3-isopropyl-5-oxadiazole carboxylate
Isopropyl amidoxime (1.03 g, 10 mmol) was dissolved in DMF (30 mL), and Et3N (2. 8 mL, 20 mmol) and HOBT (2.30 g, 15 mmol) were added thereto. Pyruvic acid (1.04 g, 10 mmol) and EDC (2. 30 g, 13 mmol) were added to the mixture which was then stirred at room temperature for 15 hours. After the reaction was complete, DMF was removed by distillation under reduced pressure, followed by dilution with a saturated aqueous solution of NaHCO3 and extraction with EtOAc. The organic extract was washed with brine, dried over MgSO4 and distilled under reduced pressure. The residue was dissolved in DMF and pyridine was added thereto, followed by stirring at 80 °C for 15 hours. After the reaction was complete, a saturated aqueous solution OfNH4Cl was added thereto, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and distilled under reduced pressure. The residue was purified by column chromatography (EtOAc/n- Hex = 1/5) to afford the title compound (12.8 g, 75%).
Step B: 3-isopropyl-5-(hydroxymethyl')oxadia2ple Methyl 3-isopropyl-5-oxadiazole carboxylate (1.0 g, 4.67 mmol) obtained in Step A was dissolved in THF (10 mL) and LiBH4 (186 mg, 9.3 mmol) was added thereto, followed by stirring at room temperature for 15 hours. After the reaction was complete, water was gradually added, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and distilled under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/5) to afford the title compound (431 mg, 65%).
Step C: 3-isopropyl-5-formyl-oxadiazole
Analogously to the procedure described in Preparation 14, the title compound was obtained using 3-isopropyl-5-(hydroxymethyl)oxadiazole obtained in Step B. Mass[M+l] = 140
Preparation 17: 5-formylindan-l-ol Step A: 5-bromoindan-l-ol
5-bromo-l-indanone (300 mg, 1.42 mmol) was dissolved in 50 mL of methanol, and the mixture was cooled to 0°C, followed by addition of sodium borohydride (65 mg, 1.7 mmol). After stirring for 30 min, an ice bath was removed and the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was extracted with ethyl acetate and a saturated ammonium chloride aqueous solution. The organic layer was taken, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography to afford the title compound (260 mg, 86%).
Step B: [(5-bromo-2,3-dihvdro-lH-inden-yl')oxy]ft-butyl)dimethylsilane 5-bromoindan-l-ol (260 mg, 1.22 mmol) obtained in Step A was dissolved in methylene chloride (50 mL) and the mixture was cooled to 00C, followed by addition of trimethyldimethylsilyl trifluoromeihane sulfonate (355 mg, 1.34 mmol). After stirring for 30 min, the reaction temperature was elevated to room temperature. The reaction was terminated with addition of a saturated aqueous solution of ammonium chloride, followed by extraction with a 0.5N HCl aqueous solution. The organic layer was taken, dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. The residue was directly used in subsequent reactions without further purification.
Step C: l-([t-butyl(dimethyl)silylloxy}indane-5-carbaldehyde
Analogously to the procedure described in Preparation 15, the title compound (326 mg, 97%) was obtained using [(5-bromo-2,3-dihydro-lH-inden-yl)oxy](t-butyl)dimethylsilane (399 mg, 1.22 mmol) obtained in Step B. The compound was directly used in subsequent reactions without further purification. Mass[M+l] =163
Preparation 18: 3-(pyrtOlidin-l-yl)ben2aldehvde Step A: N-[3-(hvdroxymethyl)phenyl1pyrrolidine
3-hydroxymethyl aniline (246 mg, 2 mmol) was dissolved in acetonitrile, and Et3N (560 fd, 4 mmol) was added thereto, followed by addition of dibromobutane (430 mg, 2 mmol). The reaction mixture was reacted in a microwave oven for 5 min, and EtOAc was added thereto, followed by washing with water and drying over MgSO4. The residue was purified by column chromatography (EtOAc/n-Hex = 1/4) to afford the title compound.
Step B: 3-(pyrrolidin-l-yl)ben2aldehyde Analogously to the procedure described in Preparation 14, the title compound was obtained using N-[3-(hydroxymethyl)phenyl]pyrrolidine obtained in Step A. Mass[M+l] =176
Preparation 19: [πS,2RH-benzyl-3-G-dimethylamino-benzylan^ acid t-butyl ester
[(lS,2R)-3-arnino-l-(3,5-difluoro-ben2yl)-2-hydroxy-propyl]-carbamic acid t-butyl ester (360 mg, 2 mmol) obtained in Preparation 13 and 3-(dimetiiylamino)benzaldehyde (300 mg, 2 mmol) obtained in Preparation 14 were placed in dichloroethane (15 mL), to which NaBH(OAc)3 (880 mg, 4 mmol) was then added. The reaction mixture was stirred at room temperature for 4 hours, followed by addition of DMC and extraction with sat'd NaHCO3 (aqueous). EtOAc was added to the residual aqueous solution, followed by dilution and extraction. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/1) to afford the title compound (590 mg, 94%). Mass[M+l] =314
Preparations 20 to 27
Analogously to the procedure described in Preparation 19, compounds of Preparations 20 to 27 as listed in Table 4 below were obtained using aldehydes commercially available or synthesized in Preparations 14 to 18 and amines synthesized in Preparations 12 and 13.
Figure imgf000042_0001
(R^= CH2-R6)
[Table 4]
Figure imgf000042_0002
Preparation 28: fSVN-BOC-prolinal
Analogously to the procedure described in Steps C and D of Preparation 16, the title compound was obtained using commercially available (S)-N-BOC-proline methyl ester. Mass[M+l] =200
Preparation 29: (RVN-BOC-prolinal
Analogously to the procedure described in Steps C and D of Preparation 16, the title compound was obtained using commercially available (R)-N-BOC-proline methyl ester. Mass[M+l] =200
Figure imgf000043_0001
Analogously to the procedure described in Steps C and D of Preparation 16, the title compound was obtained using commercially available (R)-3-benzyloxyproline methyl ester. Mass[M+l] =306
Preparation 31 : (2R,5SV2-formyl-5-isopropyl-piperazine- 1 ,4-dicarboxylic acid di-t-butyl ester Step A: r(SV2-benzyloxy-l-formyl-ethyl)-carbamic acid t-butyl ester Analogously to the procedure described in Preparation 14, the title compound was obtained using commercially available ((R)-2-benzyloxy-l-hydroxymethyl-ethyl)-carbamic acid t- butyl ester.
Step B: (S>2-[(R>3-benzyloxy-2-t-butoxycarto^ acid methyl ester
Analogously to the procedure described in Preparation 19, the title compound was obtained using commercially available (S)-isoleucine methyl ester and ((S)-2-benzyloxy-l-formyl- ethyl)-carbamic acid t-butyl ester obtained in Step A.
Step C: rS)-2-(((RV2-amino-3-benzyloxy-piOpylaminoV3-methyl-butyric acid methyl ester
(S)-2-[(R)-3-benzyloxy-2-t-butoxycarbonylamino-propylamino]-3-methyl-butyric acid methyl ester (1.5 g, 3.65 mmol) obtained in Step B was dissolved in DCM, to which TFA was then added, followed by stirring at room temperature for 2 hours. After the reaction was complete, the solvent was evaporated under reduced pressure. The residue was used directly used without further purification.
Step D: (3S,6RV6-benzyloxymethyl-3-isopiOpyl-piperazin-2-one (S)-2-(((R)-2-amino-3-ber^loxy-propylamino)-3-me%l-butyric acid methyl ester (1.0 g,
3.23 mmol) obtained in Step C was dissolved in DMF, to which Et3N (1.4 mL) was then added, followed by stirring at room temperature for 12 hours. After the reaction was complete, the reaction mixture was diluted with a saturated aqueous solution OfNaHCO3, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/4) to afford the title compound (748mg, 84%).
Step E: (3S,6R)-6-hvdroxymethyl-3-isopropyl-piperazin-2-one
Analogously to the procedure described in Step B of Preparation 12, the title compound was obtained using (3 S,6R)-6-ben2yloxymethyl-3-isopropyl-piperazin-2-one obtained in Step D.
Step F: ((2R,5S)-5-isopropyl-piperazin-2-yl*)-methanol
((3S,6R)-6-hydroxymethyl-3-isopropyl-piperazin-2-one (500 mg, 2.67 mmol) obtained in Step E was dissolved in THF (10 mL), to which LAH (180 mg) was then added, followed by stirring at room temperature for 6 hours. After the reaction was complete, H2O was added thereto, followed by addition of IN HCl and stirring at room temperature for 30 min. When a white emulsion was precipitated, extraction with EtOAc was carried out. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 2/3) to afford the title compound (390 mg, 85%). After removal of the solvent under reduced pressure, the product compound was directly used in subsequent reactions without further purification.
Step G: (2R.5SV2-hvdroxymethyl-5-isopropyl-piperazine-L4-dicarboxylic acid di-t-butyl ester ((2R,5S)-5-isopropyl-piperazm-2-yl)-methanol (340 mg, 2 mmol) obtained in Step F was dissolved in DCM, to which Et3N (560 μJL, 4 mmol) was then added, followed by addition of (BOC)2O (571 mg, 2.4 mmol) and stirring at room temperature for 4 hours. After the reaction was complete, IN HCl was added thereto, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/8) to afford the title compound (706 mg, 95%).
Step H: (2R,5S)-2-formyl-5-isopropyl-piperazine-l,4-dicarboxylic acid di-t-butyl ester
Analogously to the procedure described in Preparation 14, the title compound was obtained using (2R,5S)-2-hydroxyrnethyl-5-isopropyl-piperazine-l,4-dicarboxylic acid di-t-butyl ester obtained in Step G. Mass[M+l] = 370
Figure imgf000045_0001
Analogously to the procedure described in Preparation 31, the title compound was obtained using commercially available ((R)-2-ben2yloxy-l-hydroxymethyl-ethyl)-carbamic acid t- butyl ester and (S)-proline methyl ester. Mass|M+l] = 355 Preparation 33 : GR,5S,8SV7-phenoxy-3-fomiyl-hexahvdropyrrolo[l 2-a]pyrazine-2-carboxylic acid t-butyl ester
Analogously to the procedure described in Preparation 31, the title compound was obtained using commercially available ((R)-2-benzyloxy-l-hydroxymethyl-ethyl)-carbamic acid t- butyl ester and (2S,4S)-4-phenoxy pyrrolidine-2-carboxylic acid methyl ester. Mass[M+l] = 446
Preparation 34: (RV2-[nS2S)-2-amino-3-O,5-difluorophenylVl-hvdroxypropyl]pyrrolidine-l- carboxylic acid t-butyl ester Step A: l-G.5-difluoroV2-nitro-ethanol
3,5-difluorobenzaldehyde (2 g, 14.1 mmol) was dissolved in THF, to which K2CO3 (0.3 g, 2.26 mmol) was then added, followed by addition of nitromethane (2.0 rnL, 36.6 mmol) and stirring for 12 hours. After the reaction was complete, a solid material was filtered through celite, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/4) to afford the title compound (2.19 g, 76%).
Step B: 3,5-difluoro-5-[(E)-2-nitrovinyl1ben2ene l-(3,5-difluoro)-2-nitro-ethanol (2.19 g, 10.7 mmol) obtained in Step A was dissolved in DCM, followed by addition of 4-dimethylaminopyridine (139 mg) and then (Ac)2O (1.11 mL, 11.8 mmol). The reaction solution was stirred at room temperature for 0.5 hours, diluted with DCM, sequentially washed with a 2% HCl aqueous solution, a NaCl saturated aqueous solution and a
NaHCO3 saturated aqueous solution, dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/10) to afford the title compound (1.13 g, 56%).
Step C: 3,5-difluoro-5-(2-nitro-ethyl)benzene 3,5-difluoro-5-[(E)-2-nitrovinyl]benzene (1.1 g, 6.02 mmol) obtained in Step B was placed in DMSO and acetic acid solution, and NaBH4 (91 mg, 24.08 mmol) was added thereto. The reaction solution was stirred below 30 °C for 1.5 hours. After the reaction was complete, the reaction solution was diluted with EtOAc, washed with H2O and NaHCO3, dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (2- propanol/chloroform = 1/4) to afford the title compound (1.08 g, 95%).
Step D: (RV2-[(lS,2SV3-f3,5-difluoro-phenylH-hydroxy-2-r^^ acid t-butyl ester
3,5-difluoro-5-(2-nitro-ethyl)benzene (260 mg, 1.38 mmol) obtained in Step C was dissolved in THF, to which (Bu)4NF (1.26 mL, 1. 26 mmol) was then added, followed by addition of (R)-I -BOC-prolinal and stirring at room temperature for 30 min. After the reaction was complete, the reaction mixture was diluted with EtOAc, washed with H2O, dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/5) to afford the title compound (360 mg, 67%).
Step E: (RV2-[(lS,2SV2-amino-3-(3,5-dfluoro-phenylVl-hvdroxy-propyll-pyrrolidine-l- carboxylic acid t-butyl ester
(R)-2-[(lS,2S)-3<3,5-dMuoro-phenyl)-l-hydroxy-2-mtro-propyl]-pyrrolidine-l- carboxylic acid t-butyl ester (560 mg, 1.13 mmol) obtained in Step D was dissolved in THF, to which Raney nickel (Ni) was then added, followed by stirring at room temperature for 4 hours. After the reaction was complete, the reaction mixture was filtered through celite and extracted with EtOAc. The organic extract was washed with H2O and NaHCO3, dried over MgSO4, and concentrated under reduced pressure. The residue was purified by column chromatography (DCM/MeOH = 10/1) to afford the title compound (250 mg, 48%). Mass[M+l] = 357
Preparation 35: (R)-2-[(lS2R)-2-amino-3-(3,5-difluorophenylVl-hvdroxypropyl]pyrrolidine-l- carboxylic acid t-butyl ester Analogously to the procedure described in Step E of Preparation 34, Ihe title compound was obtained using an isomer obtained during Step D of Preparation 34 (isomer ratio = 4:1). Mass[M+l] = 357
Preparations 36 and 37: (SV2-rriS2SV2-amino-3-(3,5-difluoro-phenylVl-hvdroxy-propyl1- pyrrolidine- 1-carboχylic acid t-butyl ester and (S)-2-("(lSj2R)-2-amino-3-(3,5-difluoro-phenyl)-l- hydroxy-propyl]-pyrrolidine-l-carboxylic acid t-butyl ester
Analogously to the procedure described in Steps D and E of Preparation 34, the title compound was obtained using 3,5-difluoro-5-(2-nitro-ethyl)benzene obtained in Step C of Preparation 34 and (S)- 1 -BOC-proline aldehyde (isomer ratio = 4:1). Mass[M+l] = 357
Preparations 38 and 39: (RV2-rriS,2SV2-amino-3-rnaphthyl-l-ylVl-hvdroxy-propyη-pyrrolidine-l- carboxylic acid t-butyl ester and (R)-2-[(lS,2S)-2-amino-3-(naphthyl-l-yl)-l-hydroxy-propyll- pyrrolidine-1-carboxylic acid t-butyl ester Analogously to the procedure described in Preparation 34, the title compound was obtained using 2-(naphthyl-l-yl)-nitroethane and (R)-l-BOC-proline aldehyde (isomer ratio = 4: 1). Mass[M+l] = 371
Preparation 40: (K)-2-|Yl S2SV2-aniino-3-cvclohexyl-l-hydroxypropyl1pyrrolidine-l-carboxylic acid t-butyl ester
Analogously to the procedure described in Preparation 34, the title compound was obtained using 2-cyclohexyl-nitroethane and (R)-I -BOC-proline aldehyde.
Mass[M+l] = 327
Preparation 41: gR,5SV2-|"αS2S)-2-amino-3-G.5-difluoro-phenylVl-hvdroxypropyl1-5-isobutyl- piperazine-l,4-dicarboxylic acid di-t-butyl ester
Analogously to the procedure described in Steps D and E of Preparation 34, the title compound was obtained using (2R,5S)-2-formyl-5-isopropyl-piperazine-l,4-dicarboxylic acid di-t- butyl ester obtained in Preparation 31 and 3,5-difluoro-5-(2-nitro-ethyl)benzene obtained in Step C of Preparation 34.
Mass[M+l] = 528
Preparations 42 and 43: (3R,8aSV3-rπS.2SV2-amino-3-(3,5-diiluoro-phenvn-l-hvdroxypiOPyl1- hexahydro-pyrrolo[l^alpyrazine-2-carboxylic acid t-butyl ester and (3R,8aS)-3-[(lS,2R)-2-amino- 3-(3,5-difluoro-phenyl)- 1 -hydroxypropyli-hexahydro-pyrrolof 12a]pyrazine-2-carboxylic acid t- butyl ester Analogously to the procedure described in Steps D and E of Preparation 34, the title compound was obtained using (3R,5S)-3-fonOyl-hexahydropyrrolo[l,2-a]pyrazine-2-carboxylic acid t-butyl ester obtained in Preparation 32 and 3,5-difluoro-5-(2-nitro-ethyl)benzene obtained in Step C of Preparation 34 (isomer ratio = 4: 1). Mass[M+l] = 511
Preparation 44: (3R.7S.8aSV3-rπS^SV2-arnino-3-(3.5-difluoro-phenylVl-hvdroxypropyll-7- phenoxy-hexahydro-pyrrolo[l,2a]pyrazine-2-carboxylic acid t-butyl ester
Analogously to the procedure described in Steps D and E of Preparation 34, the title compound was obtained using (3R,5S,8S)-7-phenoxy-3-fomiyl-hexahyαVopyπOlo[l,2-a]ρyrazine- 2-carboxylic acid t-butyl ester obtained in Preparation 33 and 3,5-difluoro-5-(2-nitro-ethyl)benzene obtained in Step C of Preparation 34. Mass[M+l] = 504
Preparation 45: (2R.4RV2-[π S^SV2-amino-3-r3,5-difluoro-phenylVl -hydroxypropylH- benzyloxy-pyrroliαϋne-1-carboxylic acid t-butyl ester
Analogously to the procedure described in Steps D and E of Preparation 34, the title compound was obtained using (R)-3-benzyloxy prolinal obtained in Preparation 30 and 3,5- difluoro-5-(2-nitro-ethyl)benzene obtained in Step C of Preparation 34. Mass[M+l] = 463
Preparation 46: ('S)-2-[πS2S)-2-amino-l-hvdroxy-3-phenylpropyl"|-4-benzyl-3-oxo-piperazine-l- carboxylic acid t-butyl ester Step A: (S)-4-benzyl-2-|Tl S,2SV2-(dibenzylamino)-l-hvdroxy-3-phenylpropyl]-3-oxo-piperazine- 1-carboxylic acid t-butyl ester
Dϋsopropylamine (1.42 mL, 10.08 mmol) was dissolved in anhydrous tetrahydrofuran (10 mL) and the mixture was cooled to -78 °C. n-butyl Miium (2.5 M n-hexane solution, 3.9 mL) was added dropwise to the resulting solution. The mixture was stirred for 5 min and then stirred on an ice bath for 30 min. The reaction mixture was cooled to -78 °C, and a solution of t-butyl 4-benzyl- 3-oxopiperazine-l-carboxylate (B) (2.44 g, 8.40 mmol) in 15 mL of anhydrous tetrahydrofuran was added dropwise thereto, followed by stirring at that temperature for 1.5 hours. Thereafter, a solution of 2(S)-2-(dibenzylamino)-3-phenylpropanal (A) (2.99 g, 9.07 mmol) in anhydrous tetrahydrofuran (15 mL) was added to the reaction mixture which was gradually warmed to room temperature. After stirring at room temperature for 16 hours, the reaction was terminated with addition of water, followed by extraction with saturated ammonium chloride (aqueous) and diethyl ether. The organic layer was taken, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (EtOAc/n-Hex = 1/5) to afford the title compound (1.92 g, 24%).
Step B: (SV2-[(lS,2S)-2-aniino-l-hydroxy-3-phenylpropyl]-4-benzyl-3-oxo-piperazine-l- carboxylic acid t-butyl ester
(S)4-ber^l-2-[(lS,2S)-2-(dibenzylarnino>l-hydroxy-3-phenylpropyl]-3-oxo- piperazine-1-carboxylic acid t-butyl ester (550 mg, 0.887 mmol) obtained in Step A was dissolved in 20 mL of ethanol, to which one drop of acetic acid was then added, followed by addition of 500 mg of palladium hydroxide and stirring under 1 atm hydrogen (balloon) for 18 hours. The reaction mixture was filtered through celite, followed by extraction with IN hydrochloric acid (aqueous) and diethyl ether. The aqueous layer was adjusted to a pH of about 8 with addition of an aqueous solution of 6N sodium hydroxide, followed by extraction with methylene chloride. The organic extract was dried over anhydrous MgSO4, filtered and concentrated under reduced pressure to afford the title compound (117 mg, 30%). Mass[M+l] = 440
Preparation 47: (SV3-((lS,2SV2-amino-l-hydroxy-3-phenyl-propyl)-l-methyl-hexahydro- pyrrolofl 2-a1pyrazin-4-one
Step A: rSV2-rmethoxy-methyl-carbamoylVpyrroHdine-l-carboxylic acid t-butyl ester Commercially available (S)-l-BOC-proline (2 g, 9.3 mmol) was placed in DCM (30 mL), followed by sequential addition of triethylamine (5. 22 mL, 37.2 mmol), HOBt (1.89 g, 13.95 mmol), EDC (2.14 g, 13. 95 mmol) and meihoxy methyl amine (0.91 g, 9.3 mmol) and stirring at room temperature for 12 hours. After the reaction was complete, a saturated aqueous solution of NH4Cl was added to the reaction mixture which was then extracted with EtOAc. The organic extract was washed with a saturated aqueous solution OfNaHCO3 and dried over anhydrous MgSO4. The solvent was removed under reduced pressure. The residue was purified by column chromatography (eluent: EtOAc/n-Hex = 1/1) to afford the title compound (1.29 g, 54%).
Step B: (Sy2-acetyl-pyrrolidine-l-carboxylic acid t-butyl ester (S^^methoxy-meΛyl-carbamoyO-pyrrolidine-l-carboxylic acid t-butyl ester (1.29 g,
4.99 mmol) obtained in Step A was placed in THF (10 mL). The reaction mixture was cooled to - 78 "C, and methyl magnesium grignard (12 mL, 15 mmol) was added thereto. The reaction mixture was then warmed to room temperature and stirred for 1 hour. The reaction mixture was cooled to - 78 °C and a saturated aqueous solution OfNH4Cl was added to terminate the reaction, followed by extraction with EtOAc. The organic extract was dried over anhydrous MgSO4 and the solvent was removed under reduced pressure. The residue was purified by column chromatography (eluent: EtOAc/n-Hex = 1/1) to afford the title compound (1.03 g, 97 %).
Step C: ("SVl-pyrrolidin-2-yl-ethanone
Analogously to the procedure described in Step C of Preparation 31, the title compound was obtained using (S^-acetyl-pyrrolidine-l-carboxylic acid t-butyl ester obtained in Step B.
Step D: r(SV2-((SV2-aceM-pyrroUdm-l-ylVl-benzyloxymethyl-2-oxo-ethyl]-carbamic acid t-butyl ester
Analogously to the procedure described in Step A, the title compound was obtained using (S)-l-pyrrolidin-2-yl-ethanone obtained in Step C and commercially available (S)-3-benzyloxy-2-t- butoxycarbonylamino-propionic acid.
Step E: (SVl-methyM-oxo-octahydro-pyrrolofl^-aipyrazme-S-carboxyaldehyde
Analogously to the procedure described in Step C of Preparation 31, Preparation 19, and Steps E, G and H of Preparation 31, the title compound was obtained using [(S)-2-((S)-2-acetyl- pvrroHdin-l-yl)-l-benzyloxymethyl-2-oxo-ethyl]-carbamic acid t-butyl ester obtained in Step D.
Step F: rSV3-(αS.2SV2-aniino-l-hvdroxy-3-phenyl-proDylVl-methyl-hexahvdro-pyrrolo|'1.2- a]pyrazin-4-one
Analogously to the procedure described in Steps D and E of Preparation 34, the title compound was synthesized using (S)-l-me%14-oxo-octahydro-pyirolo[l^-a]pyrazine-3- carboxyaldehyde obtained in Step E and 3,5-difluoiO-5-(2-nitro-ethyl)benzene obtained in Step C of Preparation 34. Mass[M+l] = 440
Preparation 48: l^-acetylamino-phenvD-S-oxo^S-dihydro-lH-pyrazole-S-carboxylic acid
Analogously to the procedure described in Preparations 1 and 2, the title compound was synthesized using N-(2-amino-phenyl)-acetamide. Mass[M+l] = 262
Example 1 : N-[f2S3RH-[(3-dimethylarcmophenyl^
5-oxo-l-phenyl-4H-pyrazole-3-carboxamide
Step A: (2R,3S)-3-arnino-l-O-dimethylaniino-ben2ylainino)4-phenyl-butan-2-ol
[(lS^R)-l-benzyl-3-(3-dime1hylatnino-benzylarnmo)-2-hydroxy-propyl]-carbarm acid t- butyl ester (314 mg, 1 mmol) obtained in Preparation 19 was dissolved in DCM (5 mL), and TFA (2 mL) was added thereto, followed by stirring at room temperature for 1 hour. After the reaction was complete, the solvent was removed under reduced pressure. The residue was directly used in subsequent reactions without further purification.
Step B: N-[r2S3RM-[G-dimethylaminophenyl)methylaminol-3-hydroxy-l -phenylbutan-2-yl1-5- oxo- 1 -phenyHH-pyrazole-S-carboxamide
(2R,3S)-3-amino-l-(3-dimethylamino-benzylamino)^l-phenyl-butan-2-ol (100 mg, 0.32 mmol) obtained in Step A was dissolved in DMF, and the reaction solution was adjusted to a pH of 8 to 9 with addition OfEt3N (280 ≠, 2 mmol), followed by addition of EDC (74.4 mg, 0.41 mmol) and HOBT (973.4 mg, 0.48 mmol) and stirring at room temperature for 12 hours. After the reaction was complete, a saturated aqueous solution Of NaHCO3 was added, followed by extraction with EtOAc. The organic extract was dried over MgSO4 and distilled under reduced pressure. The residue was purified by column chromatography (EtOAc: n-Hex = 2: 1) to afford the title compound (133 mg, 84%).
1H NMR (500 MHz, CDCl3): δ 7.69(d, IH), 7.33 (t, IH), 7.18 (t, IH), 7.06 (t, IH), 6.70 (d, 2H), 6.63 (s, IH), 6.60-6.52 (m, 3H), 4.10(brs, IH), 3.85 (t, 2H), 3.82-3.77 (m, 2H), 2.99-2.90 (m, 2H), 2.87-2.75 (m, 2H), 2.77 (s, 6H)
Examples 2 to 21
Analogously to the procedure described in Example 1 , compounds of Examples 2 to 21 as listed in Table 6 below and represented by the following general formula were synthesized using acids synthesized in Preparations 2 to 11 and amine compounds obtained in Preparations 19 to 27 and 48.
Figure imgf000055_0001
[Table 6]
Figure imgf000055_0002
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
(Me: methyl, Pπpropyl, and *: isomeric form)
Example 22: N-r(lR.2SV3-r3,5-difluorophenylVl-hvdroxy-l-rr2RVpyrrolidin-2-vnpropan-2-yll-5- oxo-l-phenvMH-pyrazole^-carboxamide Step A: 0lV2-{riS2SV3-r3,5-difluoro-phenyl)-l-hvdiOxy-2-[(5-oxo-l-phenyl-4,5-dihvdro-lH- pyrazole^-carbonylVarninoJ-propyll-pyrrolidine-l-carboxylic acid t-butyl ester
Analogously to the procedure described in Step B of Example 1, the title compound was obtained using (R)-2-[(l S^S)-2-arnmo-3-(3,5-diiluoro-phenyl)-l-hydroxy-propyl]pym)lidine-l- carboxylic acid t-butyl ester obtained in Preparation 34 and commercially available 5-oxo-l-phenyl- 4,5-dihydro-lH-pyrazole-3-(^boxyric acid. Step B: N-^lR^Sl-S-rB^-dmuorophenvn-l-hvdroxy-l-rQRVpyrrolidin^-ylipiOpan^-yll-S-oxo- 1 -phenyl-4H-pyrazple-3-c^boxamide
Analogously to the procedure described in Step A of Example 1, the title compound was obtained using (R)-2-{(lS,2S)-3-(3,5-difluoro-phenyl)-l-hydroxy-2-[(5-oxo-l-phenyl-4,5-dihydro- lH-pyrazole-S-carbony^-aniinoJ-propylJ-pyπOlidine-l-carboxylic acid t-butyl ester obtained in Step A.
1H NMR (500MHz, MeOD): δ 8.02-8.00 (m, IH), 7.73 (d, 2H), 7.50 (t, 2H), 7.34 (t, IH), 6.94 (d, 2H), 6.76 (t, IH), 5.96 (s, IH), 4.38-4.35 (m, IH), 3.96-3.94 (m, IH), 3.72-3.70 (m, IH), 3.64- 3.61(m, IH), 3.56-3.52(m,lH), 3.29-3.26 (m, 2H), 3.02(d, 2H), 2.12-1.92 (m, 2H)
Examples 23 to 33
Analogously to the procedure described in Example 22, compounds of Examples 23 to 33 as listed in Table 7 below and represented by the following general formula were synthesized using acid synthesized in Preparation 5, commercially available 5-oxo-l-phenyl4,5-dihydro-lH-pyrazole- 3-carboxylic acid, and amine compounds obtained in Preparations 34 to 41 and 45 to 46.
Figure imgf000059_0001
[Table η
Figure imgf000059_0002
Figure imgf000060_0001
Figure imgf000061_0002
(*: isomeric form)
Examples 34 to 37
Analogously to the procedure described in Example 21, compounds of Examples 34 to 37 as listed in Table 8 below and represented by the following general formula were synthesized using commercially available 5-oxo-l-phenyl-4,5-dihydro-lH-pyrazole-3-carboxylic acid and amine compounds obtained in Preparations 42 to 44 and 47.
Figure imgf000061_0001
[Table 8]
Figure imgf000062_0001
( * and **: isomeric form)
Experimental Example 1 : Enzymatic activity of recombinant beta-secretase 2 Step A: Construction of vector expressing recombinant beta-secretase 2
BACE cDNA (ATCC, Cat. No. 6896840) was purchased which was synthesized based on a human BACE2 gene sequence (Accession No. BC014453) disclosed in the public Genbank data base. Only the ectodomain which corresponds to the region of from the amino acid residues 1 to 466 with exclusion of a transmembrane domain and a cytoplasmic domain in the entire BACE gene was re-cloned, and then the base sequence of Fc region which corresponds to 230 amino acids (from the amino acid residues 1 to 466) of human Immunoglobulin G (hlgG) was linked to the 3' end thereof. The BACE (ectodomain)-IgG Fc (hereinafter, referred to as "BACE-Fc") was ligated between BamHL and Xhol sites of pCDNA3 (Invitrogen) as a mammalian expression vector to construct a BACE2-Fc expression vector, designated as pCDNA3 BACE2-Fc.
Step B: Construction of mammalian cell line expressing BACE2-Fc fusion protein Chinese hamster ovary (CHO) DHFR- cells (ATCC Accession No. CRL9096) were cultured in an alpha-minimum essential medium (α-MEM, GIBCO-BRL) supplemented with 10% fetal bovine serum (FBS, GIBCO-BRL) and then transferred to a 100 mm culture plate. When the cells reached confluence, they were transfected with the BACE2-Fc-expressing vector pCDNA3
BACE2-Fc using Lipofectamine Plus (Life Technologies). Selection of transfectant cells was carried out in a medium containing 10% dialyzed fetal bovine serum (dFBS, JRH) supplemented with 1 mg/mL of Geneticin (G418 sulfate, GIBCO-BRL). The culture medium was replaced with a fresh one every 4 days. Then, 100 clones were isolated and cultured on a 24-well culture plate. Among these isolated clones, 20 clones showing acceptable growth rates were sub-cultured on a 24-well culture plates for 3 days at an equal cell density (2 x 105 cells/mL/24-well). An amount of BACE2- Fc protein secreted into the medium was quantified by ELISA method using goat anti-human IgG
(Pierce). As a result, clone #66 showing the highest growth rate and BACE2-Fc expression (3 mg/1
L of culture) was selected.
Step C: Production and purification of BACE2-Fc fusion protein 2 x 105 cells/mL of the CHO DHFR- BACE2-Fc #66 cell line were inoculated into a roller bottle containing 250 mL of α-MEM supplemented with 10% dFBS, and cultured in a Roll-In cell incubator (Bellco) at 37 °C and 40 rpm for 4 days. When cells reached confluence, they were washed once with 250 mL of a serum-free medium (SFII, GD3CO-BRL), and 500 mL of a serum- free medium containing insulin (0.5 βg/mL, SIGMA) was added, followed by culture for 3 days. After the culture medium was harvested, 500 mL of a serum-free medium was again added thereto and then the cells were cultured for another 3 days. This procedure was repeated two times. All conditioned media harvested were centrifuged at 7000 rpm for 20 min (Beckman, JA 10 rotor) to isolate only the wash solution. The wash solution was filtered through a 0.45 μm filter and was then passed through a protein A sepharose chromatography column (Pharmacia) equilibrated with 20 mM sodium phosphate buffer (pH 7.0), followed by washing with 20 mM sodium phosphate buffer (pH 7.0) to completely remove the non-adsorbed proteins. 100 mM sodium acetate buffer (pH 3.5) was added to elute the adsorbed proteins, so the BACE2-Fc protein (MW 75 kDa) of over 95% purity was obtained.
Step D: Beta-secretase 2 activity assay using fluorescence-labeled specific substrate
To determine enzymatic activity of beta-secretase 2 and inhibition efficiency of synthetic compounds, a Fluorescence Resonance Energy Transfer (FRET) enzyme activity assay was carried out using the purified B ACE2-Fc fusion protein and a fluorescence-labeled beta secretase 2-specific substrate. This will be briefly illustrated in the following.
From the entire amino acid sequence of an amyloid precursor protein (APP) known as an intracellular beta secretase 2-specific substrate, a peptide corresponding to a 10-amino acid region containing a beta-secretase cleavage site was synthesized with inclusion of EDANS as a fluorophore and DABCYL as a quenching group linked thereto. When this fluorescence-labeled substrate is reacted with BACE2-Fc, the BACE2 functional region is cleaved and the quenching group is detached, and EDANS emits fluorescence at 510 nm upon excitation with 350 nm light, whereby the level of fluorescence is measured to accurately and conveniently determine the extent of the cleavage reaction. Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 °C. For determination of activity, a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then nine successive doubling dilutions were carried out with an equal volume of DMSO. 10 μi of the diluted compound solution was added to a 96-well assay plate containing 600 μM of a fluorescence-labeled BACE substrate dissolved in 15 μJL of reaction buffer (50 mM sodium acetate, pH 4.5, 0.05% CHAPS) and 10 βi of 50% DMSO, so that DMSO was adjusted to a final concentration of 10%, and the inhibitor was treated through nine successive doubling dilutions from 500 uM. 65 μl of the purified BACE2-Fc fusion protein solution was added to a final concentration of 0.4 μg/mL and reacted at room temperature for 1 hour. An amount of the reaction product was measured by the magnitude of fluorescence at a 350 nm excitation wavelength and a 510 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device). The concentration of a synthetic compound inhibiting 50% of beta- secretase activity, Le., IC50 and Ki were determined by comparing the measurement value with that of a control group with no addition of the synthetic compound.
Experimental Example 2: Enzymatic activity of recombinant cathepsin D
To determine enzymatic activity of cathepsin D and inhibition efficiency of synthetic compounds, a Fluorescence Resonance Energy Transfer (FRET) enzyme activity assay was carried out using cathepsin D (Calbiochem, #219401) and fluorescence-labeled cathepsin D-specific substrate (Bachem #M-2455). This will be briefly illustrated in the following. As a cathepsin D-specific substrate, a 10-aa peptide was synthesized with inclusion of Mca as a fluorophore and Dnp as a quenching group linked thereto. When this fluorescence-labeled substrate is reacted with cathepsin D, the cafhepsin D functional region is cleaved and the quenching group is detached, and Mca emits fluorescence at 393 nm upon excitation with 328 nm light, whereby the level of fluorescence is measured to accurately and conveniently determine the extent of the cleavage reaction.
Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 °C. For determination of activity, a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then nine successive doubling dilutions were carried out with an equal volume of DMSO. 10 μl of the diluted compound solution was added to a 96-well assay plate containing 7 μM of a fluorescence-labeled cathepsin D substrate dissolved in 50 μl of reaction buffer (50 mM sodium acetate, pH 4.0) and 10 μl of 25% DMSO, so that DMSO was adjusted to a final concentration of 5%, and the inhibitor was treated through nine successive doubling dilutions from 250 μM. 30 μl of the purified cathepsin D fusion protein solution was added to a final concentration of 0.75 ng/mL, followed by reaction at 37 °C for 1 hour. An amount of the reaction product was measured by the magnitude of fluorescence at a 328 nm excitation wavelength and a 393 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device). The concentration of a synthetic compound inhibiting 50% of cathepsin D activity, Ie., IC50 and Ki were determined by comparing the measurement value with that of a control group with no addition of the synthetic compound.
Experimental Example 3: Secreted alkaline phosphatase (SEAP) activity assay Step A: Establishment of permanent cell line expressing SEAP-APPsw-KK A gene which expresses SEAP and Swedish mutant form of APP (CRE-SEAP- APP695sweKK) under the control of cAMP response element (CRE) was cloned into pcDNA3.1(+)Neo (Invitrogen) which is a mammalian expression vector. Neuro-2a cells (ATCC Accession No. CCL-131) were cultured in a Dulbecco's minimum essential medium (DMEM, GIBCO-BRL) supplemented with 10% FBS, and then transferred to a 6-well culture plate. When the cells reached confluence, they were transfected with the CRE-SEAP- APP695sweKK expression vector using Lipofectamine 2000 (Life Technologies). Individual clones were isolated and then cultured again on a 6-well culture plate. Following selection of clones, 100 clones showing acceptable growth rates were cultured on a 24-well culture plate for 3 days, followed by culture in DMSO/10 uM Forskolin medium for 6 hours. 50 /^/well of the culture was aliquoted and reacted with 50 μl of AttoPhos (Promega). The magnitude of fluorescence was measured at room temperature for 30 min, at a 450 nm excitation wavelength and a 580 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device). Among the selected clones, 4 clones showing high SEAP activity in forskolin/DMSO and exhibiting the greatest SEAP activity by forskolin were selected. The selected clones were sub-cultured on a 96- well culture plate for one day at an equal cell density (2 X 104 cells/96-well), followed by treatment with DMSO/10 μM Forskolin. After 6 hours, activity of SEAP secreted into the medium was assayed. Clone #159 showing the highest activity of SEAP was selected.
Step B: SEAP activity assay
The clone N2A SEAP-APPsw-KK #159 expressing CRE-SEAP-APP695sweKK was seeded at a density of 2 X 104 cells/80 μJL to each well of a 96-well culture plate. 1OmM Forskolin and DMSO were diluted 100 times with the culture medium and then 10 μt/weil of the dilution was added to the 96-well plate. Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 °C. For determination of activity, a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then seven successive 3-fold dilutions were carried out with an equal volume of DMSO. 10 μi of the serial compound dilution was diluted 10 times with 90 μi of the culture. DMSO was adjusted to a final concentration of 1.1% and the inhibitor was treated through seven successive doubling dilutions from 100 μM. After treatments were complete, the cells were cultured in a 6% CO2 incubator at 37 °C for 5 hours. In order to measure an amount of SEAP secreted into the medium, the culture was first subjected to heat inactivation at 65 °C for 30 min to thereby abolish activity of other alkaline phosphatases. 50 μi of the heat-inactivated culture and 50 μi of AttoPhos (Promega) were reacted at room temperature for 30 min. The magnitude of fluorescence was measured at a 450 ran excitation wavelength and a 580 nm emission wavelength, using a fluorescent plate reader (SpectraMax Gemini XS, Molecular Device). The concentration of a synthetic compound inhibiting 50% of SEAP activity, Ie., IC50 was determined by comparing the measurement value with that of a control group with no addition of the synthetic compound.
Experimental Example 4: Assay of amyloid beta in neurons of transgenic mice Step A: Culture of primary neurons of transgenic mice (APP/PSldE9)
Offspring mice from interbreeding of transgenic (APP/PSldE9) male and female mice were included in experiments. The brains of 3 or 4-day-old mice were dissected, and the hippocampus tissue and cerebral membrane were removed at 4°C and minced, followed by treatment with DNase (Sigma, D5025) and protease (Sigma, P5147) and placement in an incubator at 37 °C for 20 to 25 min. After being separated into cells, 4 X 105 cells/well were seeded onto a 24- well plate coated with poly-L-lysine and containing a medium (27.6 mL Neurobasal + 1.5 mL FBS + 600 μi B27 + 300 μi of 200 mM L-glutamine). The cells were cultured in an incubator at 37 "C for 7 days, and then used for subsequent experiments.
Step B: AMO activity assay Each of synthetic compounds at a concentration of 10 mM was dissolved in DMSO and then stored at 20 °C . For determination of activity, a 10 mM DMSO solution was first added to the right row of a 96-well plate, and then six successive 3-fold dilutions were carried out with an equal volume of DMSO. The serial compound dilution was diluted 250 times with a medium (29.1 mL Neurobasal + 600 μi of B27 + 300 μi of 200 mM L-glutamine + 7.5 μi of 100 mM L- glutamate). 350 μi of the dilution was added to the cultured primary neurons and the cells were then cultured in an incubator at 37 °C for 8 hours. An expression level of the beta-amyloid peptide secreted into the medium was measured by sandwich ELISA (Biosource, #KHB3482) using two types of antibodies specific for the beta-amyloid peptide, which will be briefly illustrated below.
50 μi of the water-soluble beta-amyloid or cell culture diluted to different concentrations and 50 μi of reporter antibodies were mixed and added to an antibody-coated plate, followed by reaction at room temperature for 3 hours (or one or more days at 4 °C ). The culture plate was washed five times with a 1-fold volume of a wash solution (Biosource, #KHB3482), and 100 μi of horseradish peroxidase (HRP)-coηjugated antibodies diluted 100 times with an antibody dilution solution (Biosource, #KHB3482) was added thereto, followed by reaction at room temperature for 30 min. Then, HRP-conjugated antibodies were removed and the plate was washed five times with a 1-fold volume of a wash solution, to which 100 μi of a chromogene solution (Biosource, #KHB3482) was added, followed by reaction at room temperature for 30 min. Then, 100 μi of a stop solution (Biosource, #KHB3482) was added thereto, followed by reaction at room temperature for 30 min. The optical density (absorbance) was measured at 450 nm using a microplate reader (SpectraMax 340, Molecular Device). The concentration of a synthetic compound inhibiting 50% of intracellular beta-secretase activity, Ie., IC50 was determined by comparing the measurement value with that of a control group treated with 0.4% DMSO and with no addition of the synthetic compound.
Experimental Example 5: In vivo assay of beta-secretase activity
The inhibition degree of intracellular beta-secretase activity by synthetic compounds can be determined by using a cell line producing a beta-amyloid peptide from an amyloid precursor protein (APP).
Step A: Establishment of permanent cell line expressing amyloid precursor protein (APP)
A mutant form of APP gene (APP75 INFEV), expression of which is under the control of Tet-response element (TRE), was cloned into a PBI-L vector (ClonTech) as a mammalian expression vector that expresses a luciferase gene under the control of Tet-response element (TRE). Neuro-2a cells (ATCC Accession No. CCL- 131) were cultured in a Dulbecco's minimum essential medium (DMEM, GIBCO-BRL) supplemented with 10% FBS, and then transferred to a 6-well culture plate. When the cells reached confluence, they were transfected with the pBI-L APP751 NFEV expression vector using Lipofectamine 2000 (Life Technologies). Individual clones were isolated and then cultured again on a 6-well culture plate. Following selection of clones, 100 clones showing acceptable growth rates were cultured on a 96-well culture plate for 1 day. The culture medium was replaced with a medium containing 1 μg/mL of doxycycline, followed by culture for 24 hours. 50 ≠ of Bright-Glo luciferase reagent (Promega) was added to each well which was then left at room temperature for 15 min. Then, luminescence of each well was measured using a luminometer (Victor). Following selection of clones, 4 clones showing the highest expression level of luciferase were sub-cultured on a 24-well culture plate for one day at an equal cell density (3 X 105 cells/mL/24-well). The culture medium was replaced with Opti-MEM (GIBCO-BRL) containing 1 μg/mL of doxycycline, followed by culture for 24 hours. An amount of the beta- amyloid peptide secreted into the medium was measured using an ELISA method using antibodies specific for the beta-amyloid peptide. As a result, clone #79 showing the highest growth rate and beta-amyloid peptide expression level was selected.
Step B: ELISA assay of water-soluble beta-amyloid precursor protein (sAPP)
3 X 105 cells/well of Neuro-2a APP751 NFEV # 79 with permanent expression of a mutant form of APP were seeded onto a 24-well culture plate. The cells were cultured in a 6% CO2 incubator at 37 °C for 24 hours. When the cells reached confluence, the culture medium was replaced with Opti-MEM (GIBCO-BRL) containing 300 μi of 1 μg/mL of doxycycline and the beta-secretase inhibitor (compounds of Examples) diluted to concentrations of individual steps, followed by culture for 24 hours. An expression level of water-soluble beta-amyloid precursor secreted into the medium was measured using a human sAPP assay kit (TBL) according to the manufacturer's instructions. This will be briefly illustrated in the following.
100 μi of the water-soluble beta-amyloid precursor or cell culture diluted to different concentrations was added to an antibody-coated plate, followed by reaction at room temperature for 4 hours (or one or more days at 4 "C). The culture plate was washed seven times with a wash solution (PBS + 0.05% Tween 20), and 100 μi of HRP-conjugated antibodies diluted 30 times with an antibody dilution solution (PBS + 1% BSA + 0.05% Tween 20) was added thereto, followed by reaction at room temperature for 30 min. Then, the plate was washed nine times with a wash solution, to which 100 μi of a TMB solution was added, followed by reaction at room temperature for 30 min. Then, 100 μi of a IN sulfuric acid (H2SO4) solution was added thereto, followed by reaction at room temperature for 30 min. The optical density (absorbance) was measured at 450 nm using a microplate reader (SpectraMax 340, Molecular Device). The concentration of a synthetic compound inhibiting 50% of intracellular beta-secretase activity, i.e., IC50 was determined by comparing the measurement value with that of a control group treated with 1 % DMSO and with no addition of the synthetic compound.
Experimental Example 6: Beta-amyloid assay
Quantitative analysis of beta-amyloid was carried out by ELISA using two types of antibodies (Human beta amyloid 1-40 colorimetric immunoassay kit, Biosource, California, USA). Two antibodies used in ELISA were an antibody that specifically recognizes the N-terminus of beta- secretase-cleaved beta amyloid and an antibody that binds to the C-terminus thereof. Two different antibodies and the beta amyloid protein were reacted at room temperature for 3 hours (or one or more days at 4°C). The plate was washed four times with a wash solution, and reacted with HRP- conjugated anti-rabbit IgG antibodies for 30 min. The plate was washed four times with a wash solution, and tetramethylbenzidine as the HRP substrate was added, followed by reaction at room temperature for 30 min. The optical density (absorbance) was measured at 450 nm using a microplate reader (SpectraMax 340, Molecular Device). Reduction of beta amyloid was determined by comparing the measurement value with that of a control group.
Assay of sAPPbeta (secreted amyloid precursor protein beta) was also carried out analogously to Hie procedure described in ELISA of beta-amyloid.
Experimental Example 7: Animal tests (in vivo assay)
In order to investigate whether the activity of beta-secretase was inhibited, the inhibition degree of production of beta amyloid which is a beta-secretase cleavage product was examined in animals. The animals used in the experiments were transgenic mice harboring both a Swedish mutant form of beta-APP (chimeric mouse/human amyloid precursor protein 695swe) and a mutant form of presenilin 1 (presenilin l-dE9) (Jankosky JL et al., Biomolecular engineering, 17(6), 157- 165, 2001). The beta-secretase inhibitor was administered at a dose that is expected to result in a decrease of the amyloid protein via an intraperitoneal or subcutaneous route but does not cause toxicity. Following administration of the drug, the animals were anesthetized at a given time point, and blood and cerebral tissues were isolated. The blood was collected in a heparinized tube by cardiac puncture and centriiuged at 13,000 rpm for 10 min (Eppendorf) to separate the plasma The plasma was stored together with the excised brain tissues (cerebral cortex and hippocampus) at 80 °C until use. For analysis, the plasma was diluted 5 times, and inhibitory effects on amyloid formation were examined by ELISA as described above. A 4-fold volume of PBS was added to the excised brain tissues which were then homogenized with a sonicator. The homogenate was reacted with guanidine at room temperature for 4 hours. For extraction of the amyloid protein with guanidine- buffer, guanidine was adjusted to a final concentration of 5M using 8.2 M guanidine/82 mM Tris HCl (pH 8.0). The beta amyloid protein was extracted and diluted 1:500 in BSAT-DPBS (Dulbecco's phosphate buffered saline with 5% BSA and 0.03% Tween-20), followed by analysis.
Experimental Example 8: Dosing and formulation
A drug compound was dissolved in 10% hydroxypropyl-beta-cyclodextrin (HPCD) and was then administered to a subject, typically at a dose of 15 to 100 mg/kg/day, once or three to five times.
The compounds of the present invention had a Ki value of about 10 to 100000 nM, preferably 10 to about 1000 nM, and more preferably 10 to 100 nM. In addition, IC50 for SEAP was in the range of 100 to 100000 nM, and preferably 100 to 10000 nM. The compound of Example 15 had a Ki value of 24 nM, IC50 of 1500 nM for SEAP, and 83-fold higher selectivity for cat. D.
INDUSTRIAL APPLICABILITY As apparent from the foregoing, compounds of Formula (I) in accordance with the present invention exhibit excellent inhibitory effects on human beta-secretase. Therefore, these compounds can be used as drugs for improvement of cognitive functions or for prevention and treatment of neurodegenerative diseases such as Alzheimer's disease.
Although the preferred embodiments of the present invention have been disclosed for illustrative purposes, those skilled in the art will appreciate that various modifications, additions and substitutions are possible, without departing from the scope and spirit of the invention as disclosed in the accompanying claims.

Claims

WHAT IS CLAIMED IS
1. A compound represented by Formula (T):
Figure imgf000075_0001
wherein ris l to 3;
R1 is selected from the group consisting of hydrogen, alkyl, alkene, and -(CH2)p-A-R7 wherein p is 0 to 2, A is aryl or heteroaryl, and R7 is selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, -NHC(O)-alkyl and aryloxy;
R2 is selected from the group consisting of hydrogen, alkyl, alkoxy and -(CR8R9^-R10 wherein p is 0 to 2, R8 and R9 are each independently selected from the group consisting of hydrogen, alkyl and alkoxy, and R10 is selected from the group consisting of cycloalkyl, aryl, heterocycle and heteroaryl; and B is any one of substituents represented by Formulae (i) to (ϋi):
Figure imgf000075_0002
Figure imgf000076_0001
(iii) wherein m and n are each independently 0 or 1 , X is C or N,
R3 and R4 are each independently selected from the group consisting of hydrogen, alkyl and -(OXi-(CH2)P-R1 ' wherein p is O to 2, q is O or 1 , and R1 ' is aryl or heteroaryl,
R5 is selected from the group consisting of hydrogen, alkyl, alkoxy and -(O)q-(CH2)p-Rn wherein p is 0 to 2, q is 0 or 1 , and R11 is aryl or heteroaryl, and R6 is selected from the group consisting of hydrogen, alkyl, alkoxy, and -(CH2)P-A' -R12 wherein p is 0 to 2, A' is phenyl, heteroaryl, or a bicyclic compound containing phenyl, R12 is selected from the group consisting of hydrogen, halogen, hydroxy, alkyl, alkoxy, NHR13 and NR13R14 wherein R13and R14 are each independently hydrogen or alkyl, or R13and R14 are cyclized to formalkylene, wherein the alkyl, alkoxy, aryl, cycloalkyl, heterocycle, and heteroaryl may be unsubstituted or substituted with at least one substituent selected from the group consisting of halogen, amino, alkylamino, dialkylamino, alkylacylamino, Cj-C4 alkyl, hydroxy, C1-C4 alkyl alkoxy, aryl alkoxy and oxo, and the heteroaiyl and heterocycle are each independently a 4 to 8-membered ring containing 1 to 3 hetero atoms selected from the group consisting of O, N and S, and having 0 to 3 double bonds; or a pharmaceutically acceptable salt or isomer thereof.
2. The compound according to claim 1 , wherein ris 1 to 3;
R1 is selected from the group consisting of hydrogen, Ci-C6 alkyl, C2-C6 alkene and - (CH2)P-A-R7 wherein p is 0 to 2, A is 6 to 10-membered aryl or 5 or 6-membered heteroaiyl, and R7 is selected from the group consisting of hydrogen, halogen, Ci-C3 alkyl, Ci-C3 alkoxy, NHC(O)- (C1-C6 alkyl) and 5 or 6-membered aryloxy;
R2 is selected from the group consisting Of Ci-C6 alkyl, Q-Cόalkoxy and -(CR8R^p-R10 wherein p is 0 to 2, R8 and R9 are each independently hydrogen or C1-C4 alkyl, and R10 is selected from the group consisting of 5 or 6-membered cycloalkyl, 6 to 10-membered aryl, and 5 or 6- membered heteroaryl; and B is any one of substituents represented by Formulae (i) to (iii):
Figure imgf000077_0001
(Hi) wherein m and n are each independently 0 or 1 , X is C or N, R3 and R4 are each independently selected from the group consisting of hydrogen, Ci-C6 alkyl and -(0^-(CH2)P-R1 ' wherein p is 0 to 2, q is 0 or 1, and R11 is phenyl or 5 or 6-membered heteroaryl,
R5 is selected from the group consisting of hydrogen, Cj-C6 alkyl, Ci-C6alkoxy and -(O)q- (CH2)P-R1 ' wherein p is 0 to 2, q is 0 or 1 , and Rπ is phenyl or 5 or 6-membered heteroaryl, and R6 is selected from the group consisting of hydrogen, Ci-C6 alkyl and -(CH2)p-A'-R12 wherein p is 0 to 2, A' is phenyl, 5 or 6-membered heteroaryl, or a bicyclic compound containing phenyl, and R12 is selected from the group consisting of hydrogen, halogen, hydroxy, Ci-C6 alkyl, Ci-C6 alkoxy, NHR13 and NR13R14 wherein RI3and R14 are each independently hydrogen or Ci-C4 alkyl, or RI3and R14 are cyclized to form alkylene.
3. The compound according to claim 2, wherein R1 is selected from the group consisting of hydrogen, C1-C4 alkyl, C2-C4 alkene and -(CH2)p-A-R7 wherein p is 0 to 2, A is phenyl or pyridine, and R7 is selected from the group consisting of hydrogen, halogen, trifluoromethyl, trifluoromethoxy, acerylamino and 5 or 6-membered aryloxy.
4. The compound according to claim 3, wherein R1 is selected from the group consisting of hydrogen, methyl, hydroxyethyl, vinyl, phenyl, 4-chlorophenyl, 4-trifluoromethyloxyphenyl, 3- trifluoromethylphenyl, 4-trifluoromethylphenyl, 2-trifluoromethylphenyl, benzyl, pyridin-2-yl, 3- phenoxyphenyl and 2-acetylaminophenyl.
5. The compound according to claim 2, wherein R2 is C1-C6 alkoxy or -(CR^^p-R10 wherein p is 0 to 2, R8 and R9 are each independently hydrogen or Ci-C4 alkyl, and R10 is selected from the group consisting of 5 or 6-membered cycloalkyl, 6 to 10-membered aryl and 5 or 6- membered heteroaryl.
6. The compound according to claim 5, wherein R2 is selected from the group consisting of cyclohexyl, phenyl, 3,5-difluorophenyl and naphthyl.
7. The compound according to claim 2, wherein R3 is selected from the group consisting of hydrogen, C1-C6 alkyl and [-(COq-(CH2)P-R11] wherein R11 is phenyl or 5 or 6-membered heteroaryl.
8. The compound according to claim 7, wherein R3 is hydrogen, benzyloxy or benzyl.
9. The compound according to claim 2, wherein R4 is selected from the group consisting of hydrogen, Ci-C4 alkyl and 0-(CH2)P-R1 ' wherein R1 ' is phenyl or 5 or 6-membered heteroaryl.
10. The compound according to claim 9, wherein R4 is selected from the group consisting of hydrogen, methyl, isobutyl and benzyloxy.
11. The compound according to claim 2, wherein R5 is selected from the group consisting of hydrogen, C1-C6 alkyl and -O-R11 wherein R1 ' may be phenyl or 5 or 6-membered heteroaryl.
12. The compound according to claim 11 , wherein R5 is hydrogen or phenoxy .
13. The compound according to claim 2, wherein R6 is selected from the group consisting of hydrogen, C1-C4 alkyl and -A'-R12 wherein A' is phenyl, 5 or 6-membered heteroaryl, or a bicyclic compound containing phenyl, and R12 is selected from the group consisting of hydrogen, halogen, hydroxy, C1-C6 alkyl, Ci-C6 alkoxy, NHR13 and NR13R14 wherein R13and R14 are each independently hydrogen or Ci-C4 alkyl, or Rl3and R14 are cyclized to form alkylene.
14. The compound according to claim 13, wherein R is selected from the group consisting of hydrogen, methyl, 3-dimethylaminophenyl, 3-(t-butyl)-phenyl, (3-isopropyl-[l,2,4]-oxadiazol-5- yl), 3-(pyrrolidin-l-yl)phenyl, and (l-hydroxy-indan-5-yl).
15. The compound according to claim 1, wherein the compound of Formula (I) is any one selected from:
N-[(2S,3R)4-[(3-dimethylaminophenyl)methylamino]-3-hydroxy-l-phenyl butan-2-yl]-5-oxo- 1 -phenyMH-pyrazole-S-carboxamide; N-[(2S,3R)-l-(3,5-dMuorophenylH-[(3-dime%lammophenyl)me%lamino]-3- hydroxybutan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide; l-(4-chlorophenyl)-N-[(2S,3R-l-(3,5-difluorophenyl)-4-[(3- dimethylaminophenyl)me1hylamino]-3-hydroxybu1an-2-yl]-5-oxo-4H-pyrazole-3-carto^ N-[(lR,2S)-3-(3,5-difluorophenyl)-l-hydroxy-l-[(2S)-pyrrolidin-2-yl]propan-2-yl]-5- oxo-l-phenyl4H-pyraz»le-3-carboxarnide;
N-[(2S,3R)4-[(3-dimelhylaminophenyl)methylamino]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo- 1 -[4-(trifluoromethoxy)phenyl]-4H-pyrazole-3-carboxamide; N-[(2S3R)-4-[(3-dimethylaminophenyl)me%lamko]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo-l-[3-(tiifluoiOmethyl)phenyl]4H-pyrazDle-3-carboxamide;
N-[(lS^R)-3-(3,5-difluorophenyl)-l-hydroxy-l-[(2S)-pyrroHdin-2-yl]propan-2-yl]-5- oxo-l-phenyl4H-pyrazole-3-carboxamide;
N-[(2S,3R)-l-(3,5-difluorophenyl)4-[(3-dimethylaminophenyl)methylamino]-3- hydroxybutan-2-yl]-5-oxo-l-[3-(1rifluoromethyl)phenyl]4H-pyrazole-3-carboxamide;
N-[(lR^R)-l-[(3R,8aS)-l,2,3,4,6,7,8,8a-octahydropyrrolo[l,2-d]pyrazin-3-yl]-3-(3,5- difluorophenyl)-l-hydroxypropan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide;
N-[(lS,2S)-l-[(3R,8aS)-l,2,3,4,6,7,8,8a-octahydropyiτolo[l,2-d]pyrazin-3-yl]-3-(3,5- difluorophenyl)- 1 -hydroxypropan-2-yl]-5-oxo- 1 -phenyl^H-pyrazole-S-carboxamide; N-[(2S,3R)4-[(3-dmethylammophe^^^ oxo- 1 -[2-(trifluorømethyl)phenyl]-4H-pyrazole-3-carboxamide; l-benzyl-N-[(2S,3R)4-[(3-dimethylaminophenyl)methylamino]-3-hydroxy-l- phenylbutan-2-yl]-5-oxo4H-pyrazole-3-carboxamide;
N-[(lR,2S)-3-(3,5-Muorophenyl)-l-hydroxy-l-[(2R,4R)4-ρhenylmethoxypyrrolidin-2- yl]propan-2-yl]-5-oxo-l -phenyMH-pyrazole-S-carboxamide;
N-[(lR^S)-3-(3,5-dffluorophenyl)-l-hydroxy-l-[(2R)-pyrrolidin-2-yl]prøpan-2-yl]-5- oxo-l-phenyl4H-pyrazole-3-carboxamide;
N-[(2S,3R)4-[(3-dime%laminophenyl)me%lainino]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo-1 -pyridin^-yMH-pyrazole-S-carboxamide; N-[(2S,3R)-4-[(3-dime%laminophenyl)me%lamino]-3-hydroxy-l-phenylbutan-2-yl]-5- oxo-l-(3-phenoxyphenyl)-4H-pyrazole-3-carboxamide;
N-[(lS,2S)-l-[(3S,8aS)-l-methyl4-oxo-2,3,6,7,8,8a-hexahydro-lH-pyrrolo[l,2- d]pyrazm-3-yl]-3-(3,5-dMuorophenyl)-l-hydroxypropan-2-yl]-5-oxo-l-phenyl-4H-pyra^ carboxamide; l-benzyl-N-[(2S,3R)4-[(3-t-bιitylphenyl)me%lamino]-3-hydroxy-l-phenylbutan-2-yl]- 5-oxo4H-pyrazole-3-carboxamide;
N-[(lS,2S)-3-(3,5-difluoiOphenyl)-l-hydroxy-l-[(3R,7S)-7-phenoxy-l,2,3,4,6,7,8,8a- octahydropyiTolo[12-d]pyra-dn-3-yl]propan-2-yl]-5-oxo-l-phenyl4H-pyrazole-3-carboxamide; N-[(l S,2R)- 1 -hydroxy-S-naphthalen^-yl- 1 -[(2R)-pyrrolidin-2-yl]propan-2-yl]-5-oxo- 1 - phenyMH-pyrazole-S-carboxamide;
N-[(lR,2S)-l-hydroxy-3-naphthden-2-yl-l-[(2R)-pyrrolidin-2-yl]propan-2-yl]-5-oxo-l- phenyl-4H-pyrazole-3-carboxamide;
N-[(2S3R)-4-[(3-dime1hylaminophenyl)me1hylamino]-3-hydroxy-l-phenylbutan-2-yl]-l- methyl-5-oxo-4H-pyrazole-3-carboxamide;
N-[(2S,3RH-[(3-t-butylphenyl)me%lamino]-l-(3,5-difluoiOphenyl)-3-hydroxybutan-2- yl]-5-oxo-l-[3-(1rifluoromethyl)phenyl]4H-pyrazole-3-carboxarnide; l-benzyl-N-[(2S,3R)-4-[(3-t-butylphenyl)me%lamino]-l-(3,5-difluoroρhenyl)-3- hydroxybutan-2-yl]-5-oxo-4H-pyrazole-3-carboxamide; N-[(2S,3R)4-[(3-dimethylamhophenyl)me%lamino]-3-hydroxy-l-phenylbutan-2-yl]-l- ethenyl-5-oxo-4-methyl-pyrazDle-3-carboxamide;
N-[(2S,3R)4-[(3-dime%laminophenyl)me%lamino]-3-hydroxy-l-phenylbutan-2-yl]-l- (2-hydroxye1hyl)-5-oxo-4-methyl-pyraziole-3-carboxamide; N-KlR^SyS-cyclohexyl-l-hydroxy-l-P^-pyrroHdin^-ylJpropan^-yy-S-oxo-l- phenyMH-pyrazole-3-carboxamide;
N-[(2S,3R)-3-hydrøxy- 1 -phenyl-4-[(3-propan-2-yl- 1 ,2,4-oxadiazol-5- yl)methylamino]butan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide; N-[(2S,3R)-3-hydroxy-l-phenyl4-[(3-pyrrolidin-l-ylphenyl)methylamino]butan-2-yl]-5- oxo-l-phenyl-4H-pyrazole-3-carboxamide; l-(2-acetylaminophenyl)-N-[(2S3RH-[(3-dimethylaminophenyl)melhylatnino]-3- hydroxy- 1 -phenylbutan^-ylJ-S-oxopyrrolidine-S-carboxamide;
N-[(2S3R)-3-hydroxy4-[(l-hydroxy-2,3-dihydro-lH-inden-5-yl)methylamino]-l- phenylbutan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxaniide;
N-[(2S,3R)-l-(3,5-difluorophenyl)-3-hydroxy-4-[(3-pyrroUdin-l- ylphenyl)methylamino]butan-2-yl]-5-oxo- 1 -phenyMH-pyrazole-S-carboxamide;
N-[(lR,2R)-3-(3,5-difluoroρhenyl)-l-hyckoxy-l-[(2R,5S)-5-(2-me%lpropyl)piperazin-2- yl]propan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide; N-[(l S,2S)-3-(3,5-difluoiOphenyl)-l -hydroxy- 1 -[(2R,5S)-5-(2-methylpropyl)piperazin-2- yl]propan-2-yl]-5-oxo-l-phenyl-4H-pyrazole-3-carboxamide;
N-[(lR^S)-3-(3,5-difluorophenyl)-l-hydroxy-l-[(2R)-pyrrolidin-2-yl]propan-2-yl]-5- oxo-l-phenyl4H-pyrazole-3-carboxamide;
N-[(lS,2S)-l-[(2S)4-benzyl-3-oxopiperazin-2-yl]-l-hydroxy-3-phenylpropan-2-yl]-5- oxo- 1 -phenyMH-pyrazole-S-carboxamide; and
N-[(lR^S)-3-(3,5-dmuorophenyl)-l-hydroxy-l-[(2R,4RH-phenylmethoxypyrrolidin-2- yl]prøpan-2-yl]-5-oxo-l-(3-trifluoromethyl)phenyMH-pyrazole-3-carboxamide.
16. A pharmaceutical composition for inhibiting beta-secretase, comprising a therapeutically effective amount of a compound of Formula (T) of claim 1 or a pharmaceutically acceptable salt or isomer thereof as an active ingredient and a pharmaceutically acceptable carrier.
17. The composition according to claim 16, wherein the composition is used for the improvement of cognitive functions or treatment or prevention of neurodegenerative diseases.
18. The composition according to claim 17, wherein the composition is used for the treatment of Alzheimer's disease.
PCT/KR2008/005592 2007-09-21 2008-09-22 Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety WO2009038411A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2007-0096504 2007-09-21
KR20070096504 2007-09-21

Publications (2)

Publication Number Publication Date
WO2009038411A2 true WO2009038411A2 (en) 2009-03-26
WO2009038411A3 WO2009038411A3 (en) 2009-05-07

Family

ID=40468634

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2008/005592 WO2009038411A2 (en) 2007-09-21 2008-09-22 Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety

Country Status (2)

Country Link
KR (1) KR20090031331A (en)
WO (1) WO2009038411A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102471228A (en) * 2010-04-12 2012-05-23 上海源力生物技术有限公司 Intermediates for synthesis of aliskiren and their preparation
US9862708B2 (en) 2014-02-14 2018-01-09 Tempest Therapeutics, Inc. Pyrazolone compounds and uses thereof
CN108997242A (en) * 2018-07-28 2018-12-14 浙江大学 Substituted phenyl pyrazole quinoline ketone derivatives and preparation and application
WO2023176867A1 (en) * 2022-03-18 2023-09-21 日清食品ホールディングス株式会社 Method for producing 2-acetyl-1-pyrroline

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003006021A1 (en) * 2001-07-10 2003-01-23 Elan Pharmaceuticals, Inc. Alpha-hydroxyamide statine derivatives for the treatment of alzh eimer's disease
WO2005087215A1 (en) * 2004-03-09 2005-09-22 Elan Pharmaceuticals, Inc. Substituted urea and carbamate, phenacyl-2-hydroxy-3-diaminoalkane, and benzamide-2-hydroxy-3-diaminoalkane aspartyl-protease inhibitors
WO2007047306A1 (en) * 2005-10-12 2007-04-26 Elan Pharmaceuticals, Inc. Methods of treating amyloidosis using aryl-cyclopropyl derivative aspartyl protease inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003006021A1 (en) * 2001-07-10 2003-01-23 Elan Pharmaceuticals, Inc. Alpha-hydroxyamide statine derivatives for the treatment of alzh eimer's disease
US20030083356A1 (en) * 2001-07-10 2003-05-01 Schostarez Heinrich Josef Statine derivatives for the treatment of Alzheimer's disease
WO2005087215A1 (en) * 2004-03-09 2005-09-22 Elan Pharmaceuticals, Inc. Substituted urea and carbamate, phenacyl-2-hydroxy-3-diaminoalkane, and benzamide-2-hydroxy-3-diaminoalkane aspartyl-protease inhibitors
WO2007047306A1 (en) * 2005-10-12 2007-04-26 Elan Pharmaceuticals, Inc. Methods of treating amyloidosis using aryl-cyclopropyl derivative aspartyl protease inhibitors

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102471228A (en) * 2010-04-12 2012-05-23 上海源力生物技术有限公司 Intermediates for synthesis of aliskiren and their preparation
US9862708B2 (en) 2014-02-14 2018-01-09 Tempest Therapeutics, Inc. Pyrazolone compounds and uses thereof
CN108997242A (en) * 2018-07-28 2018-12-14 浙江大学 Substituted phenyl pyrazole quinoline ketone derivatives and preparation and application
WO2023176867A1 (en) * 2022-03-18 2023-09-21 日清食品ホールディングス株式会社 Method for producing 2-acetyl-1-pyrroline

Also Published As

Publication number Publication date
KR20090031331A (en) 2009-03-25
WO2009038411A3 (en) 2009-05-07

Similar Documents

Publication Publication Date Title
KR101768946B1 (en) Proline derivatives
RU2672582C2 (en) Bicyclic inhibitors
CN107207421B (en) Novel trifluoromethylpropionamide derivatives
WO2009038412A2 (en) Beta-secretase inhibiting compounds
JP2004527548A (en) Heterocyclic dicarbamides as caspase inhibitors
CN107531653B (en) Novel difluoroketoamide derivatives
CA2868321A1 (en) Kynurenine-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof
US9000185B2 (en) Cycloalkyl ether compounds and their use as BACE inhibitors
JP2019503390A (en) Therapeutic compounds
KR20220040480A (en) Difluorohaloallylamine sulfone derivative inhibitor of lysyl oxidase, preparation method and use thereof
US9868731B2 (en) Indole amide compound as inhibitor of necrosis
WO2009038411A2 (en) Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety
AU7086900A (en) Heterocyclic ketone and thioester compounds and uses
KR20090033583A (en) Compounds having glysin aryl amide moiety as inhibitors of beta-secretase
US20080070885A1 (en) Macrocyclic compounds having aspartic protease inhibiting activity and pharmaceutical uses thereof
US10927097B2 (en) Indole compound as inhibitor of necrosis
JP2019526564A (en) Novel trifluoromethylpropanamide derivatives as HTRA1 inhibitors
KR20090033752A (en) Beta-secretase inhibiting compounds having indol thiazoline moiety
US6624313B2 (en) Process for preparing hydroxypyrrolidinyl ethylamine compounds useful as kappa agonists
JP2936324B2 (en) Sulfonamide derivative
KR20090034009A (en) Beta-secretase inhibiting compounds having glysine moiety
WO2020219808A1 (en) Dicarbamate inhibitors of ns5a for treatment of hepatitis c virus infections and related diseases
JPH09278742A (en) Sulfonamide derivative
WO2006115134A1 (en) Novel benzofuran derivative, pharmaceutical composition comprising the same, and use of the derivative or composition
BR112013017987A2 (en) heterocyclic derivatives and their use in the treatment of neurological disorders

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08832052

Country of ref document: EP

Kind code of ref document: A2

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08832052

Country of ref document: EP

Kind code of ref document: A2