NZ726027B2 - Cyclohexyl pyridine derivative - Google Patents

Cyclohexyl pyridine derivative Download PDF

Info

Publication number
NZ726027B2
NZ726027B2 NZ726027A NZ72602715A NZ726027B2 NZ 726027 B2 NZ726027 B2 NZ 726027B2 NZ 726027 A NZ726027 A NZ 726027A NZ 72602715 A NZ72602715 A NZ 72602715A NZ 726027 B2 NZ726027 B2 NZ 726027B2
Authority
NZ
New Zealand
Prior art keywords
mixture
added
bistrifluoromethylphenyl
methylamino
ethyl acetate
Prior art date
Application number
NZ726027A
Other versions
NZ726027A (en
Inventor
Takashi Miyagi
Kohsuke Ohno
Kazuo Shimizu
Hikaru Suzuki
Yasunori Ueno
Original Assignee
Kissei Pharmaceutical Co 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 Kissei Pharmaceutical Co Ltd filed Critical Kissei Pharmaceutical Co Ltd
Priority claimed from PCT/JP2015/063154 external-priority patent/WO2015170693A1/en
Publication of NZ726027A publication Critical patent/NZ726027A/en
Publication of NZ726027B2 publication Critical patent/NZ726027B2/en

Links

Classifications

    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/76Nitrogen atoms to which a second hetero atom is attached
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/84Nitriles
    • Y10S514/872

Abstract

Provided is a novel compound which has an NK1 receptor antagonistic action and which is useful in the prevention and treatment of nausea and vomiting accompanying the administration of antineoplastic drugs, which have a diminished CYP3A4 inhibitory action compared to aprepitant. That is to say, the present invention pertains to a cyclohexyl-pyridine derivative represented by formula (I), or a pharmacologically acceptable salt thereof. In the formula, ring A is 4-fluoro-2-methyl phenyl or the like, X is a hydrogen atom or the like, R1 is a carboxymethyl or the like, R2 is an alkyl or the like, Y is from 0-2 or the like, U is -N(CH3)COC(CH3)2-3,5-bis(trifluoromethyl)phenyl or the like. present invention pertains to a cyclohexyl-pyridine derivative represented by formula (I), or a pharmacologically acceptable salt thereof. In the formula, ring A is 4-fluoro-2-methyl phenyl or the like, X is a hydrogen atom or the like, R1 is a carboxymethyl or the like, R2 is an alkyl or the like, Y is from 0-2 or the like, U is -N(CH3)COC(CH3)2-3,5-bis(trifluoromethyl)phenyl or the like.

Description

DESCRIPTION TITLE OF THE INVENTION CYCLOHEXYL PYRIDINE DERIVATIVE TECHNICAL FIELD The present invention relates to cyclohexyl pyridine derivatives useful as medicaments.
[0002] More particularly, the present ion relates to cyclohexyl pyridine derivatives or pharmaceutically able salts thereof which have substance P/neurokinin 1 (NK1) receptor antagonist activity, and which are useful as agents for the tion or treatment of cancer-chemotherapy-induced nausea and vomiting (CINV) and so on.
BACKGROUND ART CINV occurs when the vomiting center located in the lateral reticular formation ofthe medulla oblongata receives a us. The area postrema and the solitary nucleus of the medulla oblongata contain NK1 receptors, and the NK1 receptors are believed to be closely ed in vomiting.
Administration of an oplastic agent facilitates the serotonin ion from the enterochromaffin (EC) cells in the digestive tract, and serotonin directly stimulates the vomiting center through 5-hydroxytryptarnine3 (5-HT3) receptors in the digestive tract. Also, when serotonin stimulates the vomiting center through the chemoreceptor trigger zone (CTZ) located in the area postrema of the fourth ventricle, nausea and vomiting occur. Substance P, like serotonin, is found in the EC cells in the digestive tract, and its secretion is promoted by stration of an antineoplastic agent.
Recently, it has been revealed that substance P induces vomiting through the NK1 receptors in the CTZ or by binding to the NK1 receptors in the central nervous , and therefore NKI receptors have been attracting attention as the target for developing etic agents atent literature 1).
Aprepitant is the first selective NK1 receptor antagonist in the world which was approved as a preventive agent for nausea and vomiting associated with administration of antineoplastic agents. Regarding the mechanism of action of aprepitant, it is believed that aprepitant selectively ts the binding of substance P and the NK1 receptors in the central nervous system, which is one of the pathways that induce CINV, and thus prevents CINV. Aprepitant has been launched as a preventive agent for CINV (Non- patent ture 2).
[0005] It is known that aprepitant is metabolized by cytochrome P450 (CYP) 3A4.
Also, aprepitant is known to have a dose—dependent inhibitory effect on CYP3A4, a CYP3A4-inducing effect and a CYP2C9-inducing effect. Accordingly, aprepitant may cause the drug-drug interactions with drugs that inhibit or induce CYP3A4 or with drugs that are metabolized by CYP3A4 or CYP2C9. For example, it is reported that the inhibitory effect of aprepitant on CYP3A4 sometimes inhibits the metabolism of dexamethasone and that the dose should be thus adjusted when dexamethasone is combined with tant (Non-patent literature 3).
Therefore, when tant is used, sufficient care should be ed to the drug—drug interactions based on the inhibitory effect of aprepitant on CYP3A4.
For the above reasons, a novel NK1 receptor antagonist with fewer rug interactions is required in the prevention or treatment of CINV.
Compounds with an NK1 receptor antagonist activity such as casopitant, netupitant, ezlopitant, rolapitant, vestipitant, vofopitant and so on, are known.
However, casopitant is reported to have an inhibitory effect on CYP3A4 and cause the drug-drug interactions due to the effect Won-patent literature 4). Clinical trials on casopitant, as a preventive agent for cancer—chemotherapy-induced nausea and vomiting, had been conducted in the U.S. and Europe; however, its development was discontinued after the application. Netupitant is currently under development as a preventive agent for -chemotherapy-induced nausea and vomiting; however, netupitant is reported to have an inhibitory effect on CYP3A4 and cause the drug-drug interactions due to the effect (Non-patent literature 5). Clinical trials on tant, as a tive agent for cancer-chemotherapy-induced nausea and vomiting, had been conducted in the U.S.; however, its development was discontinued. Clinical trials on vofopitant, as a preventive agent for cancer-chemotherapy-induced nausea and ng, had been conducted in Europe; however, its development was discontinued.
Many of the above compounds resulted in the discontinuance. All the above compounds have not yet on the market.
[0008] ne derivatives claiming to be having NK1 receptor antagonist activity are described in Patent literature 1 to ‘16. And, prodrugs of ne derivatives are described in Patent literature 17 and 18.
However, cyclohexyl pyridine derivatives of the present invention are not described in the above literatures.
Citation List Patent literature Patent ture 1: US. Patent No. 6,479,483 Patent literature 2: US. Patent No. 637 Patent literature 3: US. Patent No. 7,939,533 Patent literature 4: European Patent No. 1,103,545 Patent literature 5: US. Patent No. 7,211,579 Patent literature 6: US Patent Publication No.2006/0030600 Patent literature 7: US. Patent No. 6,576,762 Patent ture 8: US. Patent No. 316 Patent literature 9: US. Patent No. 7,683,056 Patent literature 10: US. Patent No. 8,344,005 Patent literature 11: International ation No. W02011/ 054773 Patent literature 12: US. Patent Publication No. 2007/0071813 Patent literature 13: US. Patent Publication No. 2003/0083345 Patent literature 14: US. Patent Publication No. 2003/0004157 Patent literature 15: US Patent No. 6,849,624 Patent literature 16: US. Patent No. 6,297,375 Patent literature 17: US. Patent No. 6,593,472 Patent literature 18: US. Patent No. 8,426,450 Non-Patent literature Non-patent literature 1: P. J. Hesketh et al., European Journal of Cancer, 2003, Vol. 39, pp. 1074-1080 Non-patent literature 2: Toni M. Dando et al., Drugs, 2004, Vol. 64, No. 7, pp. 777-794 tent literature 3: line B. McCrea et al., CLINICAL PHARMACOLOGY & THERAPEUTICS, 2003, Vol. 74, No. 1, pp. 17-24 Non-patent literature 4: Stefano Zamuner et al., British Journal of Clinical Pharmacology, 2010, Vol. 70, No. 4, pp. 537-546 Non-patent literature 5: Corinna Lanzarotti et al., Support Care Cancer, 2013, Vol. 21, No. 10, pp. 2783-2791 SUMMARY OF THE INVENTION Problem to be solved by the Invention
[0012] A problem of the present invention is to provide a new nd which has NK1 or nist activity, whose CYP3A4 inhibitory activity is reduced compared to aprepitant, and which are useful for the prevention or ent of cancer- chemotherapy-induced nausea and vomiting. A problem ofthe present invention is preferably to provide the above compound whose central transportation property and long-acting medicinal effect is excellent.
Means for solving the Problem The present invention relates to a compound represented by the following formula (I) or a pharmaceutically acceptable salt thereof.
That is, the t invention relates to the following [1] to [12] and the like.
A compound represented by the formula (1): [Chem 1] N X (1) R‘ (R1 wherein ring A is a group represented by the following formula: [Chem.2] X is a hydrogen atom, cyano, halogen, C1-6 alkyl or hydroxymethyl; R1 is a group represented by the following formula: [Chem.3] R1b 0 m WM R111 0r wherein R1a and R1b are each ndently any one of a hydrogen atom, a fluorine atom or C1-6 alkyl; m is 0, l or 2; when m is 2, these Rla and Rlb are optionally different from each other ; R2 is C1-6 alkyl, a hydroxy group or CH, alkoxy; U is a group ented by the following formula: wherein R3a and R313 are each independently a hydrogen atom, C1_6 alkyl, hydroxy C1-6 alkyl or C1_6 alkoxlyC1_6 alkyl; Y is 0, l or 2; when Y is 2, two R2 are optionally different from each other; or a pharmaceutically acceptable salt thereof.
The compound represented by the formula (Ia) according to the above [1]: [Chem.S] wherein ring A and X have the same meaning as described in the above [1]; R” and R1d are each independently a hydrogen atom or methyl; Ul is a group ented by the ing formula: [Chem.6] lec R3d F X” F F F in which R3c and R3d are each independently a hydrogen atom, methyl or hydroxymethyl; n is 0, l or 2; when n is 2, these R10 and R1d are optionally different from each other; or a pharmaceutically acceptable salt thereof.
The compound represented by the formula (lb) according to the above [2]: [Chem.7] wherein Rlc and R1“l have the same meaning as bed in the above [2]; or a pharmaceutically acceptable salt thereof.
The compound represented by the following a according to the above [1]: [Chem. 8] HO F F or a pharmaceutically acceptable salt thereof.
The compound represented by the following a according to the above [1]: [Chem.9] l FF |\ F N/ 0 HO FFF or a ceutically acceptable salt thereof.
The compound represented by the following formula according to the above [1]: [Chem. 1 0] J. FF |\ F o N’ O HO FFF or a pharmaceutically acceptable salt thereof.
The compound represented by the following formula according to the above [1]: [Chem.l 1] or a pharmaceutically acceptable salt thereof.
The compound represented by the following a according to the above [1]: [Chem. 12] .1 FF |\ F N/ O HO F F or a pharmaceutically acceptable salt thereof.
The compound represented by the following formula according to the above [1]: [Chem.13] I F \ F o N’ CNO Hok‘“ F F . or a pharmaceutically acceptable salt f.
The compound represented by the following formula according to the above [1]: [Chem.14] I FF |\N F o N’ CN HO FF or a pharmaceutically acceptable salt thereof.
A ceutical composition comprising as an active ingredient a compound according to any one of the above [1] to [10], or a pharmaceutically acceptable salt The pharmaceutical composition according to the above [1 l], for use in the prevention of cancer—chemotherapy—induced nausea and ng.
Effect of the Invention The compounds of the present invention have an excellent NK1 receptor antagonist activity. And, CYP3A4 inhibitory activity of the compounds of the present invention is reduced compared to aprepitant. The preferable compounds of the present ion excel in central ortation property. The more preferable compounds of the present invention excel in central transportation property and long-acting medicinal effect.
Therefore, the compounds of the t invention or pharmaceutically acceptable salts f are useful as an agent for the prevention or treatment of cancer- chemotherapy—induced nausea and vomiting.
Brief ption of the Drawings [Figure 1] Figure 1 shows the effect on cisplatin—induced acute and delayed emetic response in test example 6. In the figure, each bar chart shows a value of control group (Control), the group intravenously administered with 0.01 mg/kg of the compound of Example 13 (Ex. No 13, 0.01 mg/kg, iv) and the group intravenously administered with 0.1 mg/kg of the compound of Example 13 (Ex. No 13, 0.1 mg/kg, iv) in the acute phase, and a value of control group, the group intravenously administered with 0.01 mg/kg of the compound of Example 13 (Ex. No 13, 0.01 mg/kg, iv) and the group intravenously administered with 0.1 mg/kg of the compound of e 13 (Ex. No 13, 0.1 mg/kg, iv) in the delayed phase from the left respectively.
The vertical axes show the number of retching and vomiting (Retches + Vomits) (the mean+standard error of 3 examples of control group, the mean+standard error of 3 examples of the group intravenously administered with 0.01 mg/kg, and the mean+standard error of 3 examples of the group enously administered with 0.1mg/kg).
Mode for Carrying out the Invention Hereinafter, embodiments ofthe present invention will be discribed in further detail.
In the present ion, each term has the following meaning unless otherwise specified. [001 9] The term “C16 alkyl” means a straight-chained or a branched alkyl group having 1 to 6 carbon atoms, and for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tyl, tert—butyl and the like can be illustrated.
The term “C1_6 alkoxy” means a straight-chained or a branched alkoxy group having 1 to 6 carbon atoms, and for example, methoxy, ethoxy, propoxy, isopropoxy and the like can be illustrated.
The term “hydroxyl C1-6 alkyl” means an C1-6 alkyl group tuted with a hydroxy group such as a hydroxymethyl group, a l—hydroxyethyl group, a l-hydroxy- 1,1-dimethylmethyl group, a 2-hydroxyethyl group, a 2-hydroxymethylpropyl group, a 3—hydroxypropyl group and the like.
The term “C1-6 alkoxyC1-6 alkyl” means the above C1-6 alkyl substituted by the above C1_6 alkoxy. [002 1 ] In the case Where the compounds represented by the a (I) of the present invention contain one or more asymmetric carbon atoms, all stereoisomers in the R- or S—configuration at each of asymmetric carbons and their mixtures are included in the present invention. In such cases, racemic compounds, c mixtures, individual enantiomers and mixtures of diastereomers are included in the scope of the present invention. In the case where the compounds represented by the formula (I) of the present invention have the cis-trans isomers, all cis-trans isomers are included in the t invention.
In the present invention, chemical determination can also be determined according to well—known methods in the art. For example, see also "Tokuron NMR rittai kagaku", Kodansha, 2012, p. 59.
A compound represented by the a (I) of the present invention can also be converted into phannaceutically acceptable salts thereof ing to a general method.
As such salts, acid additive salts and salts with a base can be illustrated.
As the acid additive salt, an acid additive salt with a mineral acid such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and an acid additive salt with an organic acid such as formic acid, acetic acid, roacetic acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, propionic acid, citric acid, succinic acid, tartaric acid, fumaric acid, butyric acid, oxalic acid, malonic acid, maleic acid, lactic acid, malic acid, ic acid, benzoic acid, glutamic acid, aspartic acid and the like can be illustrated.
As the salt with a base, a salt formed with nic base such as a lithium salt, a sodium salt, a potassium salt, a calcium salt, a magnesium salt and the like, and a salt formed with organic base such as N—methyl-D—glucamine, N,N'- dibenzylethylenediamine, triethylamine, piperidine, morpholine, idine, arginine, lysine, choline and the like.
In the present invention, a pharmaceutically acceptable salt also includes a solvate thereof with a pharmaceutically acceptable solvent such as water, ethanol or the like.
In the compounds represented by the formula (I) of the present invention, the symbol R1 and R2 means a substituent of the cyclohexane ring.
In an embodiment of the compound represented by the formula (I) of the present invention, as cyclohexane ring having a substituent on the ring, a group represented by the following formula can be rated.
[Chem.15] ,0/* ”>0* * * R1 R1 R1 OR: * R2 * * R1 R1 R2 R2 R2 R2 U R2 wherein, bonds with (*) are bonding site to the pyridine ring, and R1 and R2 have the same g as bed in the above [1].
A compound represented by the formula (I) of the t invention can also be prepared, for example, by a method described below or a similar method o, or a method described in literatures or a similar method thereto.
Scheme 1 [Chem.16] 7 8 (I) In the formula, L1 and L2 are each ndently a leaving group such as a chlorine atom, a bromine atom, an iodine atom, a trifluoromethanesulfonyloxy group or the like and ring A, X, R], R2, R33, R3b and Y have the same meanings as defined above.
Process 1 Compound (4) can also be prepared by conducting coupling reaction of Compound (2) with Compound (3) in an inert solvent in the presence of a base and a palladium catalyst.
Process 2 Compound (6) can also be prepared by conducting sation reaction of nd (4) with Compound (5) in an inert solvent in the presence of a base.
Process 3 Compound (8) can also be prepared by conducting coupling reaction of Compound (6) with Compound (7) in an inert solvent in the presence of a base and a palladium catalyst.
As the inert solvent, for e, N, N—dimethylformamide, N- methylpyrrolidone, dimethylsulfoxide, diethyl ether, tetrahydrofuran, 1, 4-dioxane, 1, 2- dimethoxyethane, benzene, toluene, xylene, ethanol, water and a mixed solvent thereof can be illustrated. As the base, for example, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, lithium hydroxide, potassium fluoride, cesium fluoride, triethylamine, pyridine, N, N- diisopropylethylamine, 2, 6-lutidine, and 1, abicyclo[5, 4, 0]undecene can be illustrated. As the palladium catalyst, [1,1'—bis (diphenylphosphino) ferrocene]- palladium (II) dichloride oromethane complex (1:1) , tetrakis(triphenylphosphine)palladium(0) and the like can be rated. The reaction temperature is usually at 0°C to reflux temperature. The reaction time is usually from 30 minutes to 7 days, varying based on a used starting al, solvent and reaction temperature or the like.
The above coupling reaction can also be conducted by using a ave reactor (Biotage). When a microwave r is used, the reaction is ted at pressure range: 1 to 30 bar, power range: 1 to 400 W, reaction temperature: room temperature to 300°C, and reaction time: a minute to 1 day, varying based on a used starting material, t and model.
In addition, when a protective group is required for functional group of R1 or R2, the above coupling reaction can also be conducted after introduction of protective group.
Process 4 A compound represented by the formula (I) can also be prepared by conducting reduction such as tic ion method of the olefin of Compound (8). The catalytic reduction method can be conducted, for example, by allowing Compound (8) to react by using a catalyst under a hydrogen gas atmosphere in an inert solvent. As the inert solvent, for example, methanol, ethanol, ethyl acetate, tetrahydrofuran and acetic acid can be rated. As the catalyst, for example, palladium-carbon powder, m-carbon powder, um-carbon powder, platinum-carbon powder doped with vanadium can be illustrated. The reaction temperature is usually at room temperature to reflux temperature. The on time is usually from 30 minutes to 7 days, varying based on a used starting material, solvent and reaction temperature or the like.
In on, when a protective group was introduced into functional group in the above step 3, a compound represented by the formula (I) can also be prepared by conducting deprotection reaction after the above reduction reaction.
Scheme 2 [003 7] [Chem 17] 1 1 O o 0 0° é, process 5 process 6 g 0 —. O‘S\|<F —>F ‘ (R2), R1 (sz F R1 (R2)Y 9 10 7 In the formula, R1, R2 and Y have the same meanings as defined above. [003 8] Compound (10) can also be prepared by conducting reaction of nd (9) with romethanesulfonic anhydride in an inert solvent in the presence of a base. As the inert solvent, for example, N, N-dimethylformamide, N-methylpyrrolidone, dimethylsulfoxide, diethyl ether, tetrahydrofuran, 1, 4-dioxane, l, 2-dimethoxyethar1e, benzene, toluene, xylene, and a mixed solvent thereof can be illustrated. As the base, for example, ium carbonate, sodium ate, cesium carbonate, sodium hydroxide, ium hydroxide, lithium hydroxide, potassium e, cesium fluoride, triethylamine, pyridine, N, N-diisopropylethylamine, 2, 6-lutidine, 2,6-Di-tert-butyl methylpyridine and l, 8-diazabicyclo[5, 4, 0]undecene can be illustrated. The reaction temperature is usually at 0°C to reflux temperature. The reaction time is usually from 30 minutes to 7 days, varying based on a used starting material, solvent and reaction ature or the like.
[0039] Process 6 Compound (7) can also be prepared by conducting coupling on of Compound (10) with Compound (1 1) in an inert solvent in the presence of a base and a palladium catalyst. As the inert solvent, for example, N, N-dimethylformamide, N- methylpyrrolidone, dimethylsulfoxide, diethyl ether, tetrahydrofuran, l, 4-dioxane, l, 2- oxyethane, benzene, toluene, xylene, and a mixed solvent thereof can be illustrated. As the base, for example, potassium ate, potassium acetate, sodium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide, lithium hydroxide, potassium fluoride, cesium fluoride, triethylamine, pyridine, N, N- diisopropylethylamine, 2, 6—lutidine, and l, 8-diazabicyclo[5, 4, 0]-7—undecene can be illustrated. As the palladium catalyst, for example, [1,1'-bis (diphenylphosphino) ferrocene]—palladium (II) dichloride -dich10romethane complex (1 :1), bis(triphenylphosphine)palladium(ll) dichloride can be illustrated. The reaction temperature is usually at room temperature to reflux temperature. The reaction time is usually from 30 minutes to 7 days, varying based on a used starting al, solvent and reaction ature or the like.
The above-mentioned schemes are exemplary for preparing compounds ented by the formula (I) of the present invention and synthetic intermediates thereof. The above schemes can be changed or modified into schemes which a person ordinarily skilled in the art can easily understand.
[0041] In the above schemes, when a protective group is necessary based on ion of functional group, operations of introduction and remove can also be conducted optionally in combination according to a general method.
Compounds represented by the formula (I) of the present invention and intermediates thereof can also be ed and purified, if required, according to conventional isolation and purification techniques well known to a person ordinarily skilled in the art in the relevant field, such as solvent extraction, crystallization, recrystallization, chromatography, ative high performance liquid chromatography or the like.
The compounds of the present invention have an excellent NK1 receptor nist activity, and thus can also be used as an agent for the prevention or treatment of various diseases mediated by NK1 receptor. For example, the compounds of the present invention are useful as antiemetic agent, especially useful as preventive agent of cancer—chemotherapy (for example, cisplatin)—induced gastrointestinal symptom (for example, nausea and vomiting). Preferable compounds of the t invention are not only useful for acute cancer—chemotherapy-induced nausea and vomiting but also d cancer—chemotherapy-induced-nausea and vomiting.
In an embodiment, the compounds of the present invention have an excellent NK1 receptor antagonist activity, and thus can also be used as an agent for the prevention of postoperative nausea and vomiting (PONV), nausea and vomiting associated with radiotherapy, morphine-induced vomiting or motion sickness, and the ent of schizophrenia, social phobia, anxiety and depression, alcoholism, irritable bowel syndrome, ulcerative colitis, coughing, , atopic dermatitis, psoriasis, pruritus, pain, migraine, tinnitus, benign prostatic hyperplasia, overactive bladder or urinary incontinence.
Pharmaceutical compositions of the present invention can be administered in s dosage forms depending on their usages. As such dosage forms, for example, powders, granules, fine granules, dry syrups, tablets, capsules, injections, liquids, ointments, suppositories and poultices can be illustrated, which are stered orally or parenterally.
Pharmaceutical compositions of the present invention can be prepared by using a compound represented by the formula (I) or a pharmaceutically able salt thereof and at least one of a pharmaceutical additive. These pharmaceutical itions can be ated by ng, diluting 0r dissolving with appropriate pharmaceutical additives such as excipients, disintegrants, binders, lubricants, diluents, buffers, tonicity agents, preservatives, wetting agents, emulsifying agents, dispersing agents, stabilizing , lizing agents and the like, according to a conventional formulation procedure depending upon their dosage forms.
When a pharmaceutical composition of the present invention is used in the prevention or treatment, the dosage of a compound represented by the formula (I) or a pharmaceutically able salt thereof as the active ingredient is appropriately decided to depend on the age, sex, body , degree of disorders and treatment of each patient and the like. The dosage for an adult can be decided within the range of, for example, 0.1 to 1000 mg per day, 0.1 to 500 mg per day, 0.1 to 100 mg per day, or 0.1 to 50 mg per day in the case of oral administration, and the daily dose can be divided into one, two, three or four times per day and administered. And, the dosage for an adult can be decided within the range of, for example, 0.1 to 1000 mg per day, 0.1 to 500 mg per day, 0.1 to 100 mg per day, or 0.1 to 50 mg per day in the case of parenteral administration, and the daily dose can be divided into one, two, three or four times per day and administered.
When a pharmaceutical composition of the present invention is used in the ‘ prevention of cancer-chemotherapy-induced nausea and vomiting, this pharmaceutical can also be stered before administration of antineoplastic agents. For example, the pharmaceutical can be administered immediately before administration to before an hour and a half of the administration in chemotherapy, and after the second day, the pharmaceutical can also be administered in the morning.
[0049] In an embodiment, a compound represented by the formula (I) of the present ion or a ceutically acceptable salt thereof can also be used in combination with any other medicament other than NK1 receptor antagonists. As such other medicaments used in combination, for example, corticosteroid and 5-HT3 or nist antiemetic agent can be illustrated.
When a compound represented by the formula (I) of the present invention or a pharrnaceutically acceptable salt thereof are used in combination with the other medicament, it can be administered as a formulation sing together with their active ients or as formulations each of which is separately formulated from each active ingredient. When separately formulated, these ations can be administered separately or concurrently. [005 1] Furthermore, the dosage of the compound represented by the formula (I) of the present invention or a pharmaceutically able salt thereof can be reduced depending on the dosage of the other medicaments used in combination.
EXAMPLES The present invention is r illustrated in more detail by way of the following Reference Examples, Examples and Test Examples. However, the present invention is not d o.
Reference Example 1 4-(4,4,5,5-Tetramethyl-[1,3,2]-dioxaboro1anyl)cycloheXenecarboxy1ic acid ethyl ester To a solution of 4-oxocyclohexanecarboxylic acid ethyl ester (1.00 g) and 2,6- di-tert-butylmethylpyridine (1.39 g) in romethane (40 mL) was added trifluoromethanesulfonic anhydride (1.74 g) at room temperature, and the mixture was stirred at room temperature for 16 hours. The insoluble material was removed by filtration, and then washed with dichloromethane (5 mL). The filtrate was concentrated under reduced pressure, and to the residue was added dichloromethane (5 mL). The insoluble material was removed by filtration, and then washed with dichloromethane (3 mL). The filtrate was concentrated under reduced pressure, and to the residue was added dichloromethane (3 mL). The insoluble material was removed by filtration, and then washed with dichloromethane (2 mL). The filtrate was trated under reduced pressure to give 4-trifluoromethanesulfonyloxy—cyclohexenecarboxylic acid ethyl ester (1.57 g). Under an argon gas atmosphere, a suspension of 4- trifluoromethanesulfonyloxy-cyclohexenecarboxylic acid ethyl ester (1.57 g), bis(pinacolato)diboran (1.39 g), [l,1'-bis(diphenylphosphino)ferrocene]palladium(II) ride dichloromethane complex (1:1) (0.13 g) and potassium e (1.53 g) in dimethyl sulfoxide (26 mL) was stirred at 50°C for 4.5 hours. The reaction mixture was cooled to room temperature and water was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous ium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel (eluent: ne/ethyl acetate=100/0-85/15) to give the title compound (0.84 g).
Reference es 2 to 7 The compounds of Reference Examples 2 to 7 were prepared in a similar manner to that described in Reference Example 1 using the corresponding starting materials. [005 5] Reference Example 8 (6-Chloroiodopyridinyl)carbamic acid tert—butyl ester Under an argon gas atmosphere, to a solution of oropyridin yl)carbamic acid tert—butyl ester (5.0 g) and N, N, N', N'—tetramethylethane-l,2-diamine (7.7 g) in diethyl ether (120 mL) was added se n-butyllithium (2.65 mol/L tetrahydrofuran solution, 25 mL) at —78°C. After the mixture was stirred at -10°C for 2 hours, to the mixture was added dropwise a solution of iodine (11.4 g) in diethyl ether (40 mL) at -78°C, and the resulting mixture was stirred at room temperature for 1 day.
To the reaction e was added a ted aqueous ammonium chloride solution, and the resulting e was extracted with diethyl ether. The organic layer was washed with 10% aqueous sodium pyrosulphite solution and brine, and dried over anhydrous ium sulfate, and the solvent was removed under reduced pressure.
The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=100/0-60/40) to give the title compound (2.59 g).
Reference Example 9 (6-Chloroiodopyridin—3—yl)methylcarbamic acid tert—butyl ester To a solution of (6-chloroiodopyridinyl)carbamic acid tert—butyl ester (2.59 g) in N,N-dimethylformamide (30 mL) was added sodium hydride (60%, 0.32 g) under ice-cooling, and the mixture was d at room temperature for 30 minutes. To the mixture was added iodomethane (2.60 g) under ice-cooling and the mixture was stirred at room temperature overnight. To the reaction mixture was added a saturated aqueous sodium hydrogen carbonate solution, and the resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure.
The obtained crude product was d by column chromatography on aminopropylsilylated silica gel (eluent: n-hexane/ethyl acetate=1 00/0-70/3 0) to give the title compound (2.66 g).
Reference Example 10 (6-Chloroiodopyridin—3-y1)methylamine To a on of oroiodopyridinyl)methylcarbamic acid tert-butyl ester (2.66 g) in dichloromethane (10 mL) was added trifluoroacetic acid (8.23 g) under ice-cooling, and the mixture was stirred at room ature for 2 hours. The reaction mixture was concentrated under reduced pressure, and to the residue was added a saturated aqueous sodium carbonate solution, and the mixture was extracted with ethyl acetate. The c layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was concentrated under reduced pressure to give the title compound (1.89 g). nce Example 11 [6-Chloro(4-fluoro—2—methylphenyl)pyridin—3 -yl]methylamine To a mixture of (6-chloroiodopyridinyl)methylamine (1.89 g), 4—fluoro methylphenyl boronic acid (1.30 g), 1,2-dimethoxyethane (20 mL) and water (20 mL) were added palladium (II) acetate (0.16 g), triphenylphosphine (0.37 g) and sodium carbonate (3.73 g) at room temperature, and the mixture was d at 90°C overnight.
The on mixture was cooled to room temperature and water was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium e, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel (eluent: n—hexane/ethyl acetate=100/0—50/50) to give the title compound (1.56 g). [005 9] nce Example 12 (6-Chloro0rth0—tolylpyridin—3—yl)methylamine To a e of (6-chlor0-4—iodopyridin—3—yl)methylamine (0.70 g), 2— methylphenyl boronic acid (0.42 g), 1,2-dimethoxyethane (10 mL) and water (10 mL) were added palladium (II) acetate (0.058 g), triphenylphosphine (0.14 g) and sodium carbonate (1.38 g) at room temperature, and the mixture was stirred at 90°C overnight.
The reaction mixture was cooled to room temperature and water was added. The resulting mixture was ted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under d pressure. The ed crude product was purified by colunm chromatography on silica gel (eluent: n-hexane/ethyl acetate=100/O-70/30) to give the title compound (0.54 g).
Reference Example 13 6—Chloronitropyridinecarbonitrile To a solution of 2, 6-dichloronitropyridine (2.50 g) in N—methylpyrrolidone (25 mL) was added copper (I) cyanide (2.32 g) at room temperature, and the mixture was stirred at 180°C for 1 hour. The reaction mixture was cooled to room temperature, and to the mixture were added ethyl e and water. The ble material was removed by filtration. The filtrate was washed with brine, and the separated aqueous layer was re-extracted with ethyl acetate. The combined organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under d pressure. The ed crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=90/10-70/3 0) to give the title compound (0.90 g).
Reference Example 14 3-Aminochloropyridine—2-carbonitrile To a solution of 6—chloronitropyridinecarbonitrile (0.32 g) and concentrated hydrochloric acid (1.2 mL) in ethanol (3 .6 mL) was added iron powder (0.34 g) at room ature, and the mixture was heated at reflux for 30 minutes. The reaction mixture was cooled to room temperature, and basified by the addition of saturated aqueous sodium hydrogen carbonate solution. To the reaction mixture was added ethyl acetate, and the resulting mixture was filtered through a Celite (registered trademark) pad. The e was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was concentrated under reduced pressure to give the title compound (0.24 g).
Reference Example 15 3—Aminobromochloropyridine—Z-carbonitrile To a solution of 3-amino-6—chloropyridinecarbonitrile (0.24 g) in N,N- dimethylformamide (8 mL) was added N-bromosuccinimide (0.37 g) at room temperature, and the mixture was stirred at the same temperature overnight. To the reaction mixture was added saturated aqueous sodium thiosulfate solution, and the resulting e was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel (eluent: ne/ethyl acetate=75/25-50/50) to give the title nd (0.30 g).
[0063] Reference Example 16 ochloro(4—fluoro-2—methylphenyl)pyridinecarbonitrile A mixture of 3-amino—4-bromo-6—chloropyridine—2-carbonitrile (0.15 g), 4— fluoromethylphenylboronic acid (0.08 g), tetrakis(triphenylphosphine)palladium(0) (0.07 g), sodium carbonate (0.20 g), 1, 2-dimethoxyethane (3.2 mL) and water (0.8 mL) was d at 100°C under microwave irradiation for 1 hour. The reaction mixture was cooled to room temperature and water was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with saturated s sodium hydrogen ate solution and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel t: n- hexane/ethyl acetate=50/50-0/100) to give the title compound (0.14 g).
Reference Example 17 3-Benzyloxy(3,5-bistrifluoromethylphenyl)—2-methylpropionic acid Under an argon gas atmosphere, to a solution of 2-(3,5- bistrifluoromethylphenyl)propionic acid methyl ester (0.60 g) in tetrahydrofuran (5 mL) was added dropwise lithium diisopropylamide (1.09 mol/L tetrahydrofuran/n—hexane solution, 2 mL) at —78°C, and the e was stirred at the same temperature for 15 minutes. To the reaction mixture was added a solution of benzyl methyl ether (0.34 g) in tetrahydrofuran (2 mL) at -78°C, and the mixture was stirred at room temperature for 1 hour. To the reaction mixture was added a saturated aqueous ammonium chloride solution, and the resulting mixture was extracted with ethyl acetate.
The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n- hexane/ethyl acetate=100/0-70/30) to give 3-benzyloxy-2—(3,5- bistrifluoromethylphenyl)methylpropionic acid methyl ester (0.78 g). To a solution of the obtained nd (0.78 g) in l (3 mL) was added 5.0 mol/L s sodium hydroxide solution (1 mL) at room temperature, and the mixture was stirred at room temperature for 3 hours. To the reaction mixture was added 2.0 mol/L hydrochloric acid (3 ml), and the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium e.
The solvent was d under reduced pressure to give the title compound (0.77 g).
Reference Example 18 —Bistrifluoromethylphenyl)-N-[6-chloro(4-fluoromethylphenyl)pyridin yl]-N-methylisobutylamide To a solution of -bistrifluoromethylphenyl)methylpr0pionic acid (0.66 g) in dichloromethane (10 mL) were added oxalyl chloride (0.56 g) and N,N- dimethylformamide (2 drops) at room temperature, and the mixture was stirred at the same temperature for 1 hour. The reaction e was concentrated under reduced pressure to give the residue. Under an argon gas atmosphere, to a solution of [6-chloro- 4-(4-fluoromethylphenyl)pyridin-3 -yl]methylamine (0.50 g) in tetrahydrofuran (10 mL) was added dropwise potassium bis(trimethylsily1)amide (0.5 mol/L toluene solution, 5.0 mL) under ice-cooling, and the mixture was stirred at room temperature for s. To the reaction mixture was added dropwise a solution of the above residue in tetrahydrofuran (5 mL) under ice-cooling, and the e was stirred at room temperature for 2 hours. To the reaction mixture was added 1.0 mol/L aqueous sodium hydrogen carbonate solution, and the mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was d by column chromatography on aminopropylsilylated silica gel (eluent: n-hexane/ethyl acetate=100/0—60/40) to give the title compound (1.03 g). nce Examples 19 and 20 The nds of Reference Examples 19 and 20 were prepared in a r manner to that described in Reference Example 18 using the corresponding starting materials.
Reference Example 21 2—(3,5—Bistrifluoromethylphenyl)-N-[6-chlorocyano(4-fluor0 methylphenyl)pyridin—3—y1]isobutylamide To a solution of 2-(3,5—bistrifluoromethylphenyl)methylpropionic acid (0.31 g) in dichloromethane (2.6 mL) were added oxalyl chloride (0.26 g) and N,N— dimethylformamide (2 drops) at room temperature, and the mixture was stirred at the same ature for 1 hour. The reaction mixture was concentrated under reduced pressure to give the residue. To a solution of 3-aminochloro(4-fluoro methylphenyl)pyridinecarbonitrile (0.14 g) in tetrahydrofuran (5 mL) was added sodium bis(trimethylsi1yl)amide (1.0 mol/L tetrahydrofuran solution, 1.1 mL) under ice- cooling, and the mixture was stirred at the same temperature for 30 minutes. To the reaction mixture was added dropwise a solution of the above residue in tetrahydrofuran (2.0 mL) under ice~cooling, and the mixture was stirred at room temperature for 30 minutes. To the reaction mixture was added a saturated aqueous sodium hydrogen carbonate solution, and the resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the t was removed under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel t: n-hexane/ethyl acetate=8 5/ 1 0) to give the title compound (0.21 g).
[0068] Reference Example 22 2-(3,5-Bistrifluoromethylphenyl)-N—[6-chlorocyano(4-fluoro methylphenyl)pyridinyl]-N—methylisobutylamide To a solution of -bistrifluoromethylphenyl)-N-[6-chlorocyano(4- fluoromethy1phenyl)pyridinyl]isobutylamide (0.21 g) in N,N-dimethylformamide (2.4 mL) was added sodium hydride (60%, 0.018 g) under ice-cooling, and the mixture was d at the same temperature for 5 minutes. To the reaction mixture was added iodomethane (0.11 g) under ice-cooling, and the mixture was stirred at room temperature overnight. To the reaction mixture was added water, and the resulting mixture was extracted with ethyl e. The organic layer was washed with water and brine, and dried over ous magnesium sulfate, and the solvent was d under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: ne/ethyl acetate=90/ 10—50/50) to give the title compound (0.09 g).
Reference Example 23 4- [5- { [2-(3 ,5—Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4-fluoro- 2-methylphenyl)pyridin—2—yl]cyclohex—3—enecarboxy1ic acid ethyl ester A mixture of 2-(3,5—bistrifluoromethylphenyl)-N-[6-chloro(4-fluoro methylphenyl)pyridin-3 -y1]-N-methylisobutylamide (0.08 g), 4-(4,4,5,5-tetramethyl- [1,3,2]-dioxaborolanyl)cyclohexenecarboxylic acid ethyl ester (0.08 g), sodium carbonate (0.05 g), tetrakis(triphenylphosphine)palladium(0) (0.02 g), 1, 2- dimethoxyethane (1.0 mL), water (0.2 mL) and ethanol (0.2 mL) was stirred at 120°C under microwave ation for 1 hour. The reaction mixture was cooled to room temperature and water was added. The ing mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel (eluent: n-hexane/ethyl e=100/O-80/20) to give the title compound (0.03 g).
Reference Examples 24 to 26 The compounds of Reference Examples 24 to 26 were prepared in a similar manner to that described in nce Example 23 using the corresponding starting als. [007 l ] Reference Example 27 2-(3,5-Bistrifluoromethylphenyl)-N-[6-(1,4-dioxaspiro[4.5]decenyl)—4—(4-fluoro- 2-methylpheny1)pyridin—3-yl]-N—methy1isobutylamide A mixture of 2-(3,5-bistrifluoromethylphenyl)—N-[6—chloro-4—(4-fluoro methylphenyl)pyridinyl]-N—methylisobutylamide (0.53 g), 8-(4,4,5,5-tetramethyl- [l,3,2]-dioxaborolanyl)-l,4—dioxaspiro[4.5]decene (0.29 g), sodium ate (0.32 g), tetrakis(triphenylphosphine)palladium(0) (0.12 g), l, 2-dimethoxyethane (7.5 mL), water (1.5 mL) and ethanol (1.5 mL) was stirred at 120°C under microwave irradiation for 30 minutes. The reaction mixture was cooled to room ature and water was added. The ing mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under d pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=90/10- /90) to give the title compound (0.52 g).
Reference Examples 28 to 30 The compounds of Reference Examples 28 to 30 were prepared in a similar - manner to that described in Reference Example 23 using the corresponding starting materials.
Reference Example 31 2-{4-[5- { [2-(3 ,5-Bistrifluoromethylphenyl)—2-methylpropionyl]methylamino} (4- 2-methylphenyl)pyridin—2—yl]cyclohexenyl}methy1pr0pionic acid ethyl ester A mixture of -bistrifluoromethylphenyl)-N-[6-chloro(4-fluoro-2— methylphenyl)pyridin-3 -yl]-N-methylisobutylamide (0.11 g), 2-methyl[4-(4,4,5,5- tetramethyl[l,3,2]dioxaborolanyl)cyclohexenyl]propionic acid ethyl ester (0.12 g), sodium carbonate (0.06 g), tetrakis(triphenylphosphine)palladium(0) (0.02 g), 1, 2- dimethoxyethane (1.5 mL), water (0.3 mL) and ethanol (0.3 mL) was stirred at 120°C under microwave irradiation for 30 minutes. The on mixture was cooled to room temperature and water was added. The ing mixture was extracted with ethyl acetate. The c layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was d under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n- hexane/ethyl acetate=100/0-60/40) to give the title compound (0.12 g).
Reference Example 32 {4- [5— { [2-(3 ,5 -Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- fluoromethylphenyl)pyridin—2-yl]cyclohexeny1}acetic acid methyl ester A mixture of 2-(3,5-bistrifluoromethylphenyl)-N-[6-chloro—4-(4-fluoro-2— methylphenyl)pyridiny1]-N-methylisobutylamide (2.00 g), [4-(4,4,5,5- tetramethyl[1,3,2]dioxaborolanyl)cyclohexenyl]acetic acid methyl ester (1.26 g), tetrakis(triphenylphosphine)palladium(0) (0.22 g), 2.0 mol/L aqueous sodium carbonate solution (5.6 mL), 1, 2-dimethoxyethane (22.5 mL) and ethanol (5 .6 mL) was stirred at 120°C under microwave irradiation for 30 minutes. The reaction mixture was cooled to room temperature and water was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under d pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel t: n—hexane/ethyl acetate=1 0/50) to give the title compound (2.14 g).
Reference Example 33 2-(3 ,5—Bistrifluoromethylphenyl)-N-[4-(4-fluoromethylphenyl)(l -methyl-3 - oxocyclohexyl)pyridin—3—yl]-N-methylisobutylamide To a suspension of copper (I) iodide (0.05 g) in diethyl ether (2 mL) was added methyllthium (1.13 mol/L diethyl ether solution, 0.45 mL) under ice-cooling, and the e was stirred at the same temperature for 15 minutes. To the reaction mixture was added dropwise 2-(3,5-bistrifluoromethylphenyl)-N-[4-(4-fluoromethylphenyl)— 6-(3 -oxocyclohex-l—enyl)pyridinyl]—N—methylisobutylamide (0.10 g) in diethyl ether (1 mL) under ice-cooling, and the mixture was stirred at room temperature for 1 hour.
To the reaction mixture was added a saturated s ammonium chloride solution, and the resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was d under reduced pressure. The ed crude product was purified by column chromatography on silica gel t: n-hexane/ethyl acetate=85/ l 5-50/5 0) to give the title compound (0.50 g).
[0076] Reference Example 34 {3 - [5 -{ [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- fluoromethylphenyl)pyridinyl]cyclohex—2-enylidene}acetic acid ethyl ester To a suspension of sodium hydride (60%, 0.012 g) in tetrahydrofuran (2 mL) was added diethyl phosphonoacetate (0.08 g) under ice-cooling, and the mixture was stirred at the same ature for 30 minutes. To the reaction mixture was added 2- (3 ,5 -bistrifluoromethylphenyl)—N—[4-(4-fluoromethylphenyl)-6—(3-oxocyclohex enyl)pyridinyl]-N—methy1isobutylamide (0.10 g) in tetrahydrofuran(1 mL) at room temperature, and the mixture was stirred at the same temperature overnight and at 50°C for 24 hours. The reaction mixture was cooled to room temperature and water was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium e, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=8 5/ 1 5-40/60) to give the title compound (0.05 g).
Reference Example 35 The compound of Reference Example 35 was prepared in a r manner to that described in Reference Example 34 using the corresponding ng material.
Reference Example 36 ~Bistrifluoromethylphenyl)-N—[6-(1,4-dioxaspiro[4.5]decyl)(4-fluoro-2— methylphenyl)pyridinyl]-N—methy1isobutylamide Under a hydrogen gas atmosphere, a suspension of - bistrifluoromethylphenyl)-N-[6-(1,4-dioxaspiro[4.5]decen—8-yl)(4-fluoro methylphenyl)pyridin-3—yl]-N-methylisobutylamide (0.52 g) and 10% palladium on carbon (0.10 g, wet) in methanol (10 mL) was stirred at room temperature ght.
The reaction mixture was filtered through a Celite (registered ark) pad, and the filtrate was concentrated under reduced pressure to give the title compound (0.50 g).
Reference Example 37 2-(3,5-Bistrifluoromethylphenyl)-N-[4-(4-fluoromethylphenyl)(4- lohexyl)pyridin—3-yl]-N-methylisobutylamide To a solution of 2-(3,5-bistrifluoromethylphenyl)—N-[6-(l,4- dioxaspiro[4.5]decyl)(4—fluoro-2—methy1phenyl)pyridinyl]-N— methylisobutylamide (0.50 g) in acetone was added 1.0 mol/L hydrochloric acid (3.0 mL) at room temperature, and the mixture was stirred at 50°C for 2 hours. The reaction mixture was cooled to room temperature and water was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=90/10-10/90) to give the title compound (0.41 g).
Reference Example 38 { [2-(3 ,5—Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- fluoromethylphenyl)pyridin—2—yl]— l -hydroxycyclohexyl}acetic acid ethyl ester Under an argon gas phere, to a solution of ethyl acetate (0.021 g) in tetrahydrofuran (1 mL) was added dropwise lithium diisopropylamide solution (1.09 mol/L tetrahydrofuran/n-hexane solution, 0.20 mL) at -78°C, and the e was stirred at the same temperature for 20 minutes. To the reaction mixture was added a solution of 2-(3,5—bistrifluoromethylphenyl)-N-[4-(4-fluoromethylphenyl)—6-(4— oxocyclohexyl)pyridinyl]-N-methylisobutylamide (0.10 g) in tetrahydrofuran (1 mL) at -78°C, and the mixture was stirred at room ature for 2 hours. To the reaction mixture was added a saturated aqueous ammonium de solution, and the resulting e was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column tography on silica gel (eluent: n-hexane/ethyl e=90/10-10/90) to give the title compound (0.10 g). [008 1] Reference Example 39 2—{4-[5-{ [2-(3,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino}(4- fluoromethylphenyl)pyridinyl]cyclohexylidene}propionic acid ethyl ester To a suspension of sodium hydride (60%, 0.017 g) in tetrahydrofuran (2 mL) was added 2-phosphonopropionic acid triethyl ester (0.11 g) under ice—cooling, and the mixture was stirred at the same ature for 30 minutes. To the reaction mixture was added a solution of 2—(3,5-bistrifluoromethylphenyl)—N-[4-(4-fluoromethylphenyl) (4-oxocyclohexyl)pyridin-3—yl]-N-methylisobutylamide (0.14 g) in tetrahydrofuran (1 mL) at room temperature, and the mixture was stirred at 50°C overnight. The reaction mixture was cooled to room temperature and a saturated aqueous ammonium chloride solution was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous ium sulfate, and the solvent was removed under reduced pressure. The ed crude t was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=100/0- 50/50) to give the title compound (0.13 g).
Reference e 40 {3 -[5-{ [2—(3 ,5 ifluoromethylphenyl)—2-methylpropionyl]methylamino } (4- fluoromethylphenyl)pyridinyl]cyclohexyl}acetic acid ethyl ester Under a hydrogen gas atmosphere, a suspension of {3-[5-{ 5— '10 bistrifluoromethylphenyl)—2-methylpropionyl]methylamino} fluoro methylphenyl)pyridin—2-yl]cyclohexenylidene}acetic acid ethyl ester (0.03 g) and % palladium on carbon (0.01 g, wet) in methanol (1 mL) was stirred at room temperature overnight. The reaction mixture was filtered through a Celite (registered trademark) pad, and the filtrate was concentrated under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n- hexane/ethyl acetate=85/15-40/60) to give the title compound (0.02 g).
Reference Example 41 The compound of Reference Example 41 was prepared in a similar manner to that described in Reference Example 40 using the corresponding starting material.
Reference Example 42 2-{4-[5—{ [2-(3,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino}(4- fluoromethylphenyl)pyridin—2-yl]cyclohexyl}propionic acid ethyl ester Under a hydrogen gas atmosphere, a suspension of 2-{4-[5-{[2-(3,5- bistrifluoromethylphenyl)methylpropionyl]methylamino}—4—(4-fluor0 methylphenyl)pyridinyl]cyclohexylidene}propionic acid ethyl ester (0.13 g) and 10% palladium on carbon (0.025 g, wet) in methanol (5 mL) was stirred at room temperature overnight. The reaction mixture was filtered through a Celite (registered trademark) pad, and the filtrate was trated under d pressure to give the title compound (0.11 g).
Reference Example 43 4~ [5 -{ [2-(3 ,5-Bistrifluoromethylphenyl)-2—methylpropionyl]methylamino} (4-fluoro- ylphenyl)pyridinyl]cyclohexanecarboxylic acid ethyl ester Under a hydrogen gas atmosphere, a suspension of 4-[5-{ [2—(3,5- bistrifluoromethylphenyl)methylpropionyl]methylamino} (4—fluoro methylphenyl)pyridin—2-yl]cyclohexenecarboxylic acid ethyl ester (0.03 g) and 10% palladium on carbon (0.010 g, wet) in l (1 mL) was stirred at room ature for 14 hours. The reaction mixture was filtered through a Celite (registered trademark) pad, and the filtrate was concentrated under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n—hexane/ethyl acetate=70/30-50/5 0) to give the title compound (0.02 g).
Reference Examples 44 to 47 The nds of Reference Examples 44 to 47 were prepared in a similar manner to that described in Reference Example 43 using the corresponding starting materials.
Reference Example 48 {4-[5-{ [3-Benzyloxy(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethylphenyl)pyridinyl]cyclohex enyl}acetic acid To a e of {4-[5-{[3-benzyloxy(3,5—bistrifluoromethylphenyl)—2- methylpropionyl]methylamino} (4-fluoromethylphenyl)pyridin—2—yl]cyclohex-3 - enyl}acetic acid ethyl ester (0.11 g), tetrahydrofuran (1 mL), methanol (0.5 mL) and water (0.5 mL) was added lithium hydroxide monohydrate (0.03 g) at room temperature, and the mixture was stirred at the same temperature overnight. To the reaction mixture was added 2.0 mol/L hydrochloric acid (0.4 mL), and the solvent was removed under reduced re. To the residue was added water, and extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: ethyl e/methanol=1 00/0-90/10) to give the title compound (0.05 g).
Reference Examples 49 and 50 trans{4-[5-{ [2-(3,5-Bistrifluoromethylphenyl)-2—methylpropionyl]methylamino} (4-fluoromethylphenyl)pyridinyl]cyclohexyl}methylpropionic acid ethyl ester (Reference Example 49), and cis{4-[5-{[2-(3,5-bistrifluor0methylphenyl) methylpropionyl]methylamino}(4—fluoromethylphenyl)pyridinyl]cyclohexyl}- 2-methy1propionic acid ethyl ester (Reference Example 50) Under a hydrogen gas atmosphere, a mixture of 2-{4-[5—{[2-(3,5- bistrifluoromethylphenyl)-2—methylpropionyl]methylamino}(4-fluoro methylphenyl)pyridinyl]cyclohexenyl}methylpropionic acid ethyl ester (0.11 g), 10% palladium on carbon (0.03 g, wet), methanol (2 mL) and tetrahydrofuran (1 mL) was stirred at room temperature ght. The reaction mixture was filtered through a Celite (registered trademark) pad, and the filtrate was concentrated under reduced pressure. The obtained crude product was d by column chromatography on silica gel (eluent: n—hexane/ethyl acetate=100/0-60/40) to give nce es 49 (0.05 g) and Reference Examples 50 (0.04 g). In the above chromatography, the compound of Reference Examples 49 was in the high polarity side, and the compound of Reference Examples 50 was in the low polarity side.
Reference Example 51 {4- [5-{ [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } —4-(4- fluoromethylphenyl)pyridin—2-yl]cyclohexyl}acetic acid methyl ester Under a hydrogen gas atmosphere, a suspension of {4-[5-{[2-(3,5- bistrifluoromethylphenyl)methylpropionyl]methylamino}-4—(4-fluoro phenyl)pyridinyl]cyclohexeny1}acetic acid methyl ester (2.14 g) and 10% palladium on carbon (E101 NE/W type (EVONIK))(0.21 g) in ol (96 mL) was stirred at room temperature overnight. The reaction mixture was filtered through a Celite (registered trademark) pad, and the filtrate was concentrated under reduced pressure. The obtained crude product was purified by column chromatography on aminopropylsilylated silica gel t: n-hexane/ethyl acetate=100/0-40/60) to give the title compound (2.13 g).
Reference e 52 {4- [5-{ [2-(3 ,5-Bistrifluoromethylpheny1)methy1propionyl]methylamino}(4- fluoromethylphenyl)pyridinyl] methylcyclohexyl } acetic acid methyl ester To a suspension of sodium hydride (60%, 0.020 g) in tetrahydrofuran (2 mL) was added dimethyl phosphonoacetic acid methyl ester (0.09 g) under ice-cooling, and the mixture was stirred at room temperature for 1 hour. To the reaction mixture was added a solution of 2-(3,5-bistrifluoromethylphenyl)-N—[4-(4-fluoromethylphenyl) (4-oxocyclohexyl)pyridin—3-yl]-N-methylisobuty1amide (0.15 g) in tetrahydrofuran (1 mL) at room temperature, and the mixture was stirred at 50°C for 30 minutes. The reactiOn mixture was cooled to room temperature and a saturated s um chloride solution was added. The resulting e was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over ous magnesium sulfate, and the t was d under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n—hexane/ethyl acetate=90/10-30/70) to give {4-[5-{[2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethy1phenyl)pyridin yl]cyclohexy1idene}acetic acid methyl ester (0.15 g).
To a suspension of copper (I) iodide (0.05 g) in diethyl ether (0.40 mL) was added methyllithium (1.13 mol/L diethyl ether solution, 0.50 mL) under ice-cooling, and the mixture was stirred at the same temperature for 10 minutes, and the solvent was concentrated under reduced pressure. Under an argon gas atmosphere, to the residue was added dichloromethane (0.40 mL) under ice-cooling, and the e was stirred for 5 minutes, and the solvent was concentrated under reduced pressure. To the residue was added dichloromethane (0.40 mL), and cooled to —78°C. To the mixture was added trimethylsilyl chloride (0.03 g), and to the resulting mixture was added dropwise a solution of {4-[5-{[2-(3,5-bistrifluoromethylphenyl)methylpropionyl]methylamino}- 4-(4-fluoromethylphenyl)pyridiny1]cyclohexylidene}acetic acid methyl ester (0.09 g) in romethane (1.0 mL). The resulting e was d under ice-cooling for 1 hour and at room temperature overnight. To the reaction mixture was added a saturated aqueous ammonium chloride solution, and the resulting mixture was ted with ethyl acetate. The c layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n— hexane/ethyl acetate=90/10-40/60) to give the title compound (0.08 g). [009 1 ] Reference Example 53 The compound of Reference Example 53 was prepared in a r manner to that described in Reference Example 16 using the corresponding starting material.
Reference Example 54 The compound of nce Example 54 was prepared in a similar manner to that described in Reference Example 21 using the corresponding ng material.
Reference Example 55 The compound of Reference Example 55 was prepared in a similar manner to that described in Reference Example 22 using the ponding starting material.
Reference Examples 56 and 57 The compounds of Reference Examples 56 and 57 were ed in a similar manner to that described in nce Example 23 using the corresponding starting materials.
Reference Examples 58 and 59 The compounds of Reference Examples 58 and 59 were prepared in a similar manner to that described in Reference Example 43 using the corresponding starting materials.
Reference Example 60 {4-[5- { [2-(3 ,5—Bistrifluoromethylphenyl)methylpropionyl]methylamino } —4-(4- fluoro—2-methylphenyl)- l -oxypyridinyl]cyclohexyl}acetic acid ethyl ester To a solution of {4—[5-{[2-(3,5—bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethylphenyl)pyridin yl]cyclohexyl}acetic acid ethyl ester (0.37 g) in dichloromethane was added m- chloroperoxybenzoic acid (purity 70%, 0.55 g) under ice-cooling, and the mixture was stirred at room ature overnight. The reaction mixture was cooled with ice, and to the mixture was added 1.0 mol/L s sodium hydroxide on. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column tography on silica gel (eluent: n-hexane/ethyl acetate=40/60—0/100) to give the title compound (0.35 g).
Reference Example 61 {4-[5 - { [2-(3 trifluoromethylphenyl)—2-methy1propiony1]methylamino}chloro (4-fluoromethylphenyl)pyridinyl]cyclohexyl}acetic acid ethyl ester A mixture of {4-[5-{ [2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino }—4—(4—fluoro-2—methylphenyl)oxypyridin yl]cyclohexyl}acetic acid ethyl ester (0.20 g) and phosphoryl chloride (0.60 mL) was stirred at 120°C for 2 hours. The reaction mixture was cooled with ice, and basified by the addition of water and aqueous ammonia. The resulting mixture was extracted with ethyl e. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was d under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n- hexane/ethyl acetate=90/ 1 0-50/5 0) to give the title compound (0.16 g).
Reference Example 62 {4- [5-{ [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4— 2-methylphenyl)methylpyridinyl]cyclohexyl}acetic acid ethyl ester A mixture of {4-[5-{[2-(3,5-bistrifluoromethylphenyl)~2- methylpropionyl]methylamino} -6—chloro(4—fluoro—2-methylphenyl)pyridin yl]cyclohexyl}acetic acid ethyl ester (0.04 g), 2,4,6—trimethylcyclotriboroxane (0.014 g), tetrakis(triphenylphosphine)palladium(0) (0.007 g), sodium ate (0.016 g) and 1, 2-dimethoxyethane (2.9 mL) was stirred at 120°C under microwave irradiation for 1 hour. The reaction mixture was cooled to room temperature and water was added. The resulting mixture was ted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the t was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=80/20-20/80) to give the title compound (0.019 g).
Reference Example 63 {4-[5- { [2-(3 trifluoromethylphenyl)methylpropionyl]methylamino } (4- 2-methylphenyl)hydroxymethylpyridinyl]cyclohexyl}acetic acid ethyl ester To a solution of {4-[5—{[2-(3,5-bistrifluoromethylphenyl)-2— propionyl]methylamino} (4-fluoro-2—methylphenyl)— 1 -oxypyridin-2— yl]cyclohexyl}acetic acid ethyl ester (0.15 g) in dichloromethane (2.0 mL) was added trimethyloxonium tetrafluoroborate (0.04 g) at room temperature, and the mixture was stirred at the same temperature for 2 hours. The reaction mixture was concentrated under reduced pressure, and to the residue was added methanol (2.0 mL). To the mixture was added a on of ammonium persulfate (0.01 g) in water (0.02 mL) at 65°C, and the mixture was stirred at the same temperature for 1 hour. To the reaction mixture was added a solution of ammonium persulfate (0.01 g) in water (0.02 mL) at 65°C, and the mixture was stirred at the same temperature for 13 hours. After the reaction mixture was cooled to room temperature, the solvent was concentrated under reduced pressure. To the residue was added an aqueous solution of sodium carbonate, and the mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate. The t was concentrated under d pressure to give a mixture of the title compound and {4-[5-{[2-(3,5- bistrifluoromethylphenyl)methylpropionyl]methylamino}(4-fluoro—2- methylphenyl)hydroxymethylpyridiny1]cyclohexyl}acetic acid methyl ester (0.06 [01 00] Example 1 4-[5 -{ [2-(3 ,5-Bistrifluoromethylphenyl)—2-methylpropionyl]methylamino } (4-fluoro— 2-methylphenyl)pyridinyl]cyclohexanecarboxylic acid To a mixture of 4-[5-{ [2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethy1phenyl)pyridin yl]cyclohexanecarboxylic acid ethyl ester (0.022 g), tetrahydrofuran (0.375 mL), methanol (0.375 mL) and water (0.150 mL) was added lithium ide monohydrate (0.014 g) at room ature, and the mixture was stirred at the same temperature for 72 hours. To the reaction mixture were added 2.0 mol/L hydrochloric acid (0.170 mL) and water, and the resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over ous sodium sulfate. The t was concentrated under reduced pressure to give the title compound (0.018 g). [01 01] Example 2 3- { 4- [5 -{ [2-(3 ,5-Bistrifluoromethylphenyl)-2—methylpropionyl]methylamino} (4- fluoromethylphenyl)pyridinyl]cyclohexyl}propi0nic acid To a mixture of 3-{4-[5-{[2-(3,5-bistrifluoromethylphenyl)—2- methylpropionyl]methylamino} —4-(4-fluoromethylphenyl)pyridin y1]cyclohexyl}propionic acid ethyl ester (0.019 g), tetrahydrofuran (0.375 mL), methanol (0.375 mL) and water (0.150 mL) was added lithium hydroxide drate (0.012 g) at room temperature, and the mixture was stirred at the same temperature for 6 hours. To the reaction mixture was added 2.0 mol& hydrochloric acid (0.140 mL) and water, and the resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous sodium sulfate. The solvent was concentrated under reduced pressure to give the title compound (0.017 g). [0 1 02] Example 3 {3 - [5 -{ [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- fluoro—2-methylphenyl)pyridinyl]cyclohexyl} acetic acid To a mixture of {3-[5-{[2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethylphenyl)pyridin-2— lohexyl}acetic acid ethyl ester (0.023 g), tetrahydrofuran (0.50 mL), methanol (0.25 mL) and water (0.25 mL) was added lithium hydroxide monohydrate (0.007 g) at room temperature, and the mixture was stirred at the same temperature overnight. The reaction mixture was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under d pressure. The ed crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate/methanol=5 0/50/0-0/ 1 00/0-0/90/10) to give the title compound (0.003 g). [01 03] Example 4 {3- [5- { [2-(3 ,5—Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- 2-methylphenyl)pyridinyl]methylcyclohexyl}acetic acid To a mixture of {3-[5-{[2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethylphenyl)pyridinyl]—3- cyclohexyl}acetic acid ethyl ester (0.022 g), tetrahydrofuran (0.40 mL), methanol (0.20 mL) and water (0.20 mL) was added lithium hydroxide monohydrate (0.006 g) at room ature, and the mixture was stirred at the same temperature overnight. The reaction mixture was lized by the addition of acetic acid. The resulting e was ted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by colurrm chromatography on silica gel (eluent: n-hexane/ethyl acetate/methanol=50/50/0-0/100/0-0/90/10) to give the title compound (0.007 g). [01 04] Example 5 {4-[5-{ [2-(3 ,5 -Bistrifluoromethylphenyl)hydr0xymethylpropionyl]methylamino } - 4-(4-fluoromethylphenyl)pyridiny1]cyclohexyl}acetic acid Under a hydrogen gas here, a suspension of {4-[5-{ [3-benzyloxy (3,5-bistrifluoromethylphenyl)methylpropionyl]methylamino}-4—(4-fluoro methylphenyl)pyridin-2—yl]cyclohexeny1}acetic acid (0.045 g) and 10% palladium on carbon (0.03 g, wet) in methanol (1.5 mL) was stirred at room temperature for 5 hours.
To the reaction mixture was added 10% palladium on carbon (0.03 g, wet). Under a hydrogen gas atmosphere, the resulting mixture was stirred at room temperature overnight. The reaction mixture was filtered h a Celite (registered trademark) pad, and the filtrate was concentrated under d pressure to give the title compound (0.04 g). [01 05] Example 6 {4- [5-{ [2-(3 ,5-Bistrifiuoromethylphenyl)methylpropionyl]methylamino } (4- fluoromethylphenyl)pyridinyl] hydroxycyclohexyl } acetic acid To a mixture of {4-[5-{ 5-bistrifluoromethylphenyl) methylpropionyl]methylamino } (4-fluoromethylphenyl)pyridin—2—yl] hydroxycyclohexyl}acetic acid ethyl ester (0.05 g), tetrahydrofuran (1.00 mL), methanol (0.50 mL) and water (0.50 mL) was added lithium hydroxide monohydrate (0.015 g) at room temperature, and the mixture was stirred at the same temperature overnight. To the reaction mixture was added 2.0 mol/L hydrochloric acid (0.20 mL), and the solvent was removed under reduced pressure. To the residue was added water, and the resulting mixture was extracted with ethyl e. The organic layer was washed with brine, and dried over anhydrous ium sulfate. The solvent was concentrated under reduced pressure to give the title compound (0.046 g). [0 1 06] Example 7 6—[5- { [2-(3 ,5 -Bistrifluoromethylphenyl)—2-methylpropionyl]methylamino} (4-fluoro- 2-methylphenyl)pyridinyl] spiro [2.5]octane— 1 -carboxylic acid A mixture of 6-[5-{ [2-(3,5—bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethylphenyl)pyridin yl]spiro[2.5]0ctane-1—carboxylic acid ethyl ester (0.020 g), 1.0 mol/L aqueous sodium hydroxide solution (0.09 mL), tetrahydrofuran (0.60 mL) and ol (0.30 mL) was stirred at 140°C under microwave irradiation for 1 hour and a half. The reaction mixture was cooled to room temperature and water was added. The resulting e was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl e/methanol=40/60/0-0/100/0-0/90/10) to give the title compound (0.005 g). [01 07] Example 8 {4- [5 —{ [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino} cyano (4-fluoro—2-methylphenyl)pyridinyl]cyclohexyl}acetic acid To a mixture of {4-[5-{[2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}-6—cyano(4-fluoromethylphenyl)pyridin-2— yl]cyclohexyl}acetic acid methyl ester (0.017 g), ydrofuran (0.30 mL), methanol (0.15 mL) and water (0.15 mL) was added lithium hydroxide monohydrate (0.005 g) at room temperature, and the mixture was stirred at the same ature for 2 days. The reaction mixture was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was d under d pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl acetate=5 0/5 0-0/ 100) to give the title compound (0.008 g). [01 08] Example 9 {4-[5 — { [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } ortho- tolylpyridin-Z-yl]-cyclohexyl}acetic acid To a mixture of {4-[5-{[2—(3,5-bistrifluoromethylphenyl)~2- methylpropionyl]methylamino } -4—0rtho-tolylpyridin—2-yl]-cyclohexyl } acetic acid methyl ester (0.08 g), tetrahydrofuran (1.00 mL), methanol (0.50 mL) and water (0.50 mL) was added lithium hydroxide drate (0.021 g) at room temperature, and the mixture was stirred at the same temperature for 3 hours. To the reaction mixture was added 2.0 mol/L hydrochloric acid (0.28 mL), and the solvent was removed under reduced pressure. To the residue was added water, and the resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate. The t was trated under reduced pressure to give the title compound (0.071 g).
Example 10 2-{4- [5 - { [2—(3 ,5—Bistrifluoromethylphenyl)methylpropionyl]methylamino} (4- fluoromethylphenyl)pyridiny1]cyclohexyl}pr0pionic acid A e of 2-{4—[5—{ [2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}-4—(4-fluoromethylphenyl)pyridin yl]cyclohexyl}propionic acid ethyl ester (0.11 g), 1.0 mol/L aqueous sodium hydroxide solution (0.50 mL), tetrahydrofuran (0.50 mL) and methanol (1.50 mL) was stirred at 140°C under microwave irradiation for 1 hour and a half. The reaction mixture was cooled to room temperature and 1.0 mol/L hydrochloric acid (0.60 mL) was added. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium e. The solvent was concentrated under reduced pressure to give the title compound (0.10 g). [0 1 1 0] Example 11 2- {4- [5 - { [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino} (4-fluoro-2—methylphenyl)pyridin—2-yl]cyclohexyl} —2-methylpropionic acid A mixture of trans{4-[5-{ [2-(3,5—bistrifluoromethylphenyl) methylpropionyl]methylamino} fluoromethy1phenyl)pyridin—2-yl]cyclohexyl} - 2-methylpropionic acid ethyl ester (0.054 g), 1.0 mol/L aqueous sodium hydroxide solution (0.25 mL), tetrahydrofuran (0.25 mL) and methanol (0.75 mL) was stirred at 140°C under microwave irradiation for 1 hour and a half. The reaction mixture was cooled to room temperature and 1.0 mol/L aqueous sodium ide solution (0.25 mL) was added. The ing mixture was stirred at 140°C under microwave irradiation for 1 hour and a half. The reaction mixture was cooled to room temperature and 1.0 mol/L hydrochloric acid (0.60 mL) was added. The ing mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The t was concentrated under reduced pressure to give the title compound (0.022 g). [01 1 1] Example 12 cis—2- {4— [5 - { [2-(3 ,5-Bistrifluoromethylphenyl)—2-methylpropionyl]methylamino} (4- fluoromethylpheny1)pyridin—2-yl]cyclohexyl}methylpropionic acid A mixture of cis{4-[5-{ [2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino } (4—fluoromethylphenyl)pyridinyl]cyclohexyl} - 2-methylpropionic acid ethyl ester (0.044 g), 1.0 mol/L aqueous sodium hydroxide on (0.20 mL), ydrofuran (0.20 mL) and methanol (0.60 mL) was stirred at 140°C under microwave irradiation for 1 hour and a half. The reaction mixture was cooled to room temperature and 1.0 mol/L aqueous sodium hydroxide solution (0.20 mL) was added. The ing mixture was stirred at 140°C under microwave irradiation for 1 hour and a half. The reaction mixture was cooled to room temperature and 1.0 mol/L hydrochloric acid (0.50 mL) was added. The resulting mixture was ted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n-hexane/ethyl e=90/10-10/90) to give the title compound (0.014 g). [0 1 1 2] Example 13 and 14 trans- {4— [5-{ [2—(3 ,5—Bistrifluoromethylphenyl)—2-methy1propiony1]methylamino } (4— 2-methylpheny1)pyridiny1]cyclohexyl}acetic acid (Example 13), and cis-{4- [5 - { [2-(3 ,5-bistrifluoromethylphenyl)methy1propionyl]methylamino} (4-fluoro methylpheny1)pyridinyl]cyclohexyl}acetic acid (Example 14) (1) Synthesis of a mixture of trans and cis s To a mixed solution of {4-[5-{[2-(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino} —4-(4-fluoromethylphenyl)pyridinyl]cyclohexyl} - acetic acid methyl ester (2.13 g) in tetrahydrofuran (32 mL)-methanol (16 mL)-water (16 mL) was added lithium hydroxide monohydrate (0.41 g) at room temperature and the mixture was stirred at the same temperature for 17 hours. To the reaction mixture was added 2.0 mol/L hloric acid (4.9 mL), and the solvent was removed under reduced pressure. To the residue was added water, and the resulting mixture was extracted with ethyl e. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate. The solvent was trated under reduced pressure to give a crude product (a mixture of trans- and cis-{4-[5-{[2-(3,5— bistrifluoromethylphenyl)—2-methy1propionyl]methylamino}(4-fluoro—2- methylphenyl)pyridinyl]cyclohexyl}acetic acid) (2.08 g). (2) Separation of trans and cis isomers A mixture of trans- and cis—{4-[5-{ [2-(3,5-bistrifluoromethylphenyl)~2- methylpropionyl]methylamino} (4-fluoromethy1phenyl)pyridin—2- yl]cyclohexyl}acetic acid (36.6 g) was separated by liquid chromatography under the ing conditions to give Example 13 (17.0 g) and Example 14 (16.2 g) respectively.
[Separation ions of the liquid chromatography] (A) Preparative isolation system Device name: K-Prep (KYOTO CHROMATO Co., Ltd.) ; (B) Separation conditions Column: CHIRALPAK (registered trademark) IA ; Size: 5 cm ID. x 25 cmL. ; Particle size: 5 um ; Mobile phase: n-hexane/ethanol/acetic 5/15/0.l<v/v/v> Flow rate: 35 mL/min ; Temperature: 30°C ; Detection wavelength: 254 nm ; Injection method: Loop injection ; Injection volume: 10-20 mL (20 g/L solution) (C) Retention time Example 13: approximately 21 min, Example 14: approximately 17 min Example 13 and 14 were analyzed under the following analytical conditions.
[Analytical ions of the liquid chromatography] (A) Analytical System Pump: LC-20AD (Shimadzu Corporation) ; Detector: SPD-20A (Shimadzu Corporation) ; Auto Sampler: SIL-20A (Shimadzu Corporation) (B) Analytical Conditions Column: CHIRALPAK (registered trademark) IA ; Size:0.46 ch.D. X 25 cmL. ; Mobile phase: n-hexane/ethanol/acetic acid=85/15/0.l<V/V/V> Flow rate: 1.0 mL/min; Temperature: 40°C ; Detection wavelength: 254 nm ; ion volume: 10 uL (C) Retention time e 13 : 6.164 min, Example 14 : 5.016 min [01 14] e 15 {4- [5- { [2-(3 ,5 -Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- 2-methylphenyl)pyridinyl] — 1 -methylcyclohexyl } acetic acid To a mixture of {4-[5-{[2-(3,5-bistrifluoromethylphenyl)—2- methylpropionyl]methylamino } (4-fluoromethylphenyl)pyridin—2-yl] - l - methylcyclohexyl}acetic acid methyl ester (0.078 g), tetrahydrofuran (0.60 mL), methanol (0.30 mL) and water (0.30 mL) was added lithium hydroxide monohydrate (0.022 g) at room ature, and the mixture was stirred at the same temperature for 1 hour and at 50°C for 3 hours. The reaction mixture was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, and dried over anhydrous magnesium sulfate, and the solvent was d under d pressure. The obtained crude product was purified by column chromatography on silica gel (eluent: n—hexane/ethyl acetate/methanol=20/80/0- 0/100/0-0/90/10) to give the title compound (0.069 g). [01 1 5] Example 16 [4—(5-{ [2-(3 ,5—Bistrifluoromethylphenyl)—2-methy1propionyl]methylamino}cyano—4- 0rtho—tolylpyridin—2-yl)cyclohexyl]acetic acid To a mixture of [4-(5-{[2-(3,5-bistrifluoromethylphenyl)—2- methylpropionyl]methylamino}cyanoortho-tolylpyridin-Z-yl)cyclohexyl]acetic acid ethyl ester (0.018 g), tetrahydrofuran (0.40 mL), ol (0.20 mL) and water (0.20 mL) was added lithium hydroxide monohydrate (0.005 g) at room temperature, and the mixture was d at the same temperature overnight. The reaction mixture was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was concentrated under reduced pressure to give the title compound (0.016 g). [01 16] Example 17 {4— [5— { [2-(3 , 5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } chloro (4—fluoro—2—methylphenyl)pyridinyl]cyclohexyl } acetic acid To a mixture of {4-[5-{[2-(3,5—bistrifluoromethylphenyl) methylpropionyl]methylamino} —6—chloro(4-fluoromethylphenyl)pyridin—2- lohexyl}acetic acid ethyl ester (0.020 g), tetrahydrofuran (0.50 mL), methanol (0.25 mL) and water (0.25 mL) was added lithium hydroxide monohydrate (0.005 g) at room ature, and the mixture was stirred at the same temperature for 2 hours. The on mixture was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was concentrated under reduced pressure to give the title compound (0.018 g). [0 1 l 7] Example 18 {4— [5-{ [2-(3 ,5-Bistrifluoromethylphenyl)methylpropionyl]methylamino } (4- fluoromethylphenyl)methylpyridin—2-yl]cyclohexyl}acetic acid To a mixture of {4-[5-{[2-(3,5-bistrifluoromethylphenyl)—2- methylpropionyl]methylamino}(4—fluoromethylphenyl)methylpyridin yl]cyclohexyl}acetic acid ethyl ester (0.019 g), ydrofuran (0.50 mL), methanol (0.25 mL) and water (0.25 mL) was added lithium hydroxide monohydrate (0.005 g) at room temperature, and the mixture was stirred at the same temperature overnight. The reaction e was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was concentrated under d pressure to give the title compound (0.018 g). [01 18] Example 19 {4- [5-{ [2-(3 ,5 ifluoromethylphenyl)methylpropionyl]methylamino } —4-(4- ' 2-methylphenyl)hydroxymethylpyridin-2—yl]cyclohexyl}acetic acid To a e of a mixture of {4-[5-{ [2-(3,5—bistrifluoromethylphenyl) methylpropionyl]methylamino}(4-fluoromethylphenyl)—6-hydroxymethylpyridin- 2-yl]cyclohexyl}acetic acid ethyl ester and {4-[5-{[2—(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino} (4-fluoromethylphenyl)hydroxymethylpyridin- 2-yl]cyclohexyl}acetic acid methyl ester (0.025 g), tetrahydrofuran (0.50 mL), methanol (0.25 mL) and water (0.25 mL) was added lithium hydroxide monohydrate (0.007 g) at room temperature, and the mixture was stirred at same temperature overnight. The reaction mixture was neutralized by the addition of acetic acid. The resulting mixture was extracted with ethyl acetate. The organic layer was washed with water and brine, and dried over anhydrous magnesium sulfate. The solvent was concentrated under reduced pressure to give the title compound (0.023 g). [01' 1 9] Example 20 and 21 trans- {4— [5 — { [2-(3 trifluoromethylphenyl)methylpropionyl]methylamino} cyano(4-fluoromethylphenyl)pyridin—2-yl]cyclohexyl}acetic acid (Example 20) and cis— {4— [5 - { [2-(3 ,5-bistrifluoromethylphenyl)methylpropionyl]methylamino } cyano(4-fluoromethylphenyl)pyridinyl]cyclohexyl}acetic acid le 21) A mixture of trans— and cis—{4-[5—{[2—(3,5-bistrifluoromethylphenyl) methylpropionyl]methylamino}cyano(4-fluoromethylphenyl)pyridin—2- yl]cyclohexyl}acetic acid (Example 8) (0.18 g) was isolated by liquid chromatography under the following conditions to give Example 20 (0.035 g) and e 21 (0.037 g) respectively.
[Separation conditions of the liquid chromatography] (A) Preparative isolation system Device name: Preparative HPLC System (Gilson, Inc.) (B) Separation conditions Column: ustain tered trademark) C18 ; Size: 20 mmI.D. x 50 mmL. ; Particle size: 5 um Mobile phase: acetonitrile/ 10 mM aqueous ammonium acetate solution=45/55<v/v> ; Flow rate: 30 mL/min ; Temperature: room temperature ; Detection wavelength: 220 nm (C) Retention time Example 20: approximately 10.4 min, Example 21: imately 9 min Example 20 and 21 were analyzed under the following analytical conditions.
[Analytical ions of the liquid chromatography] (A) Analytical System PumszC-IOAT dzu Corporation) ; Detector: SPD-lOA (Shimadzu Corporation) ; Auto Sampler: SIL—lOA (Shimadzu Corporation) (B) Analytical Conditions Column: il (registered trademark) ODS-3 ; Size: 4.6 mmI.D. x 250 mL. ; Mobile phase: acetonitrile/ 10 mM s ammonium acetate solution=40/60- 80/20<v/v> Flow rate: 1.0 mL/min; Temperature: 40°C ; Detection wavelength: 225 nm ; Injection volume:5 uL (C) Retention time Example 20: 16.786 min, Example 21: 17.286 min [0 12 1] Tables 1 to 11 show the chemical structures of the above compounds of Reference Examples 1 to 63, and the chemical structures and the physical properties of the above compounds of es 1 to 21. The abbreviations in these Tables: “Ref No.”, “Ex No.”, “Str.”, “Physical data”, “lH-NMR”, “DMSO-d6” and “CDC13” ent Reference Example number, Example , chemical structure, physical property, hydrogen nuclear magnetic resonance spectrum, dimethylsulfoxide-d6 and form-d1, respectively. And, “MS” and “ESI_APCI” represent mass spectrometry and measurement of Electrospray ionization-Atmospheric pressure chemical ionization, respectively.
[Table 1] Ref. Ref.
Str. 11 12 13 14 [Table 2] [Table 3] 8131‘. Str.
[Table 4] [Table 5] [Table 6] [Table 7] Physical data 1H-NMR 5 ppm (DMSO—dfi) 2 1.00- 3.00 (22H, m), 6.80-7.30 (4H, m), 7.50—7.90 (2H, m), 8.04 (1H, s), 8.30 (1H, s), 12.17 (1H, brs) MS PCI, m/z) : 625 (M+H)+ lH—NMR 5 ppm (DMSO-d6) : 0 90— 2.90 (26H, m), 6.80-7.30 (4H, m), 7.55-7 95 (2H, m), 8.04 (1H, s), 8.20-8.40 (1H, m), 12.03 (1H, brs) MS (ESI_APCI, m/Z) : 653 (M+H)+ 1H—NMR 5 ppm (CDC13) Z 0.80- 3.10 (24H, m), 6 75—7 30 (4H, m), 7.65 (2H, brs), 7.77 (1H, brs), 8.37 (1H, brs) MS (ESI_APCI, m/z) 2 639(M+H)+ 1H-NMR 5 ppm (CD013) 3 0.80— 2. 80 , m), 6.80‘7.35 (4H, m), 7.67 (2H, brs), 7.78 (1H, brs), 8.39 (1H, brs) MS (ESI_APCI, 10/2) 3 653 (M+H)+ ‘H-NMR 5 Inm1(DMSO-d6) :1.00— 2.90 (21H, m), 3.20—3.90 (2H, m), 4.40~5.00 (1H, m), 6.90— 7.35 (4H, m), 7 40-8 15 (3H, m), 8.20-8.45 (1H, m), 12.11 (1H, brs) MS (ESI_APCI, m/z) : 655 (M+H)+ [Table 8] Physical data lH-NMR 5 ppm (DMSO-d6) :1.10- 2.90 (24H, m), 6.90—7.35 (4H, m), 7.65—7.85 (2H, m), 8.04 (1H, 8), 8.30 (1H, S) MS (ESI_APCI, m/Z) 3 655 (M+H)+ 1H-NMR 5 ppm (CD013) 1 0.90- 2.95 (24H, m), 6.75—7.35 (4H, m), 7.65 (2H, brs), 7.77 (1H, s), 8.30—8.60 (1H, m) MS (ESI_APC1, m/z) : 651(M+H)+ 1H-NMR 5 ppm (CDC13) I 1.05— 2.95 (24H, m), 6.80-7.35 (4H, m), 7.50-7.80(3H, m) MS (ESI_APCI, m/z) : 664 (M+H)+ 1H—NMR 5 ppm d6) $1.00— 2.90 (24H, m), 7 00—7.40 (5H, m), 7.70—7 90 (2H, m), 8.04 (1H, s), 8 25-8 35 (1H, m), 12.00 (1H, brs) MS (ESI_APCI, m/z) : 621 (M+H>+ 1H-NMR 5 ppm (DMSO-d6) 31.00- 3.00 (26H, m), 6.90—7.30 (4H, m), 7.65—7.85 (2H, m), 8.04 (1H, s), .35 (1H, m), 12.03 (1H, brs) MS (ESI_APCI, m/z) I 653 (M+H)+ [Table 9] EX.NQ Physical data 1H—NMR 6 ppm (DMSO-d6) 21.05 (s, 6H), 1.10—2.80 (22H, m), 6.90—7.30 (4H, m), 7.65—7.85 (2H, m), 8.03 (1H, s), 8.30 r—-‘ ._. (1H, s), 12.07 (1H, brs) MS (ESI_APCI, m/z) : 667 (M+H)+ 1H—NMR 5 ppm (DMso—d6) :0.95 (s, 6H), 1.00~3.20 (22H, m), 6.90-7.30 (4H, m), 7.66—7.85 (2H, m), 8.04 (1H, s), 8.36 1—- 2 (1H, s), 12.01 (1H, brs) 0 MS (ESI_APCI, m/z) : 667 (M+H)+ 1H-NMR 6 ppm (DMSO-d6) I 0.90- 2.80 (24H, m), .30 (4H, m), 7.60-7.90 (2H, m), 8.04 (1H, s), 8.30 (1H, s), 12.01 1—4 3 (1H, brs) MS (ESI_APCI, m/z) : 639 (M+H)+ 1H—NMR 5 ppm (DMSO—d6) Z 0.90- 2.90 (24H, m), 6 80—7 40 (4H, m), 7.60—7.90 (2H, m), 8.04 (1H, s), 8.31 (1H, s), 11.99 >——- 4 (1H, brs) MS (ESI_APCI, m/z) 2 639 (M+H)+ 1H—NMR 5 ppm ) I 1.16 (3H, s), 1 20—2 80 (28H, m), 6.80—7.35 (4H, m), 7.66 (2H, brs), 7.78 (1H, s), 8.46 (1H, brs) MS (ESI_APCI, m/z) : 653 (M+H)+ [Table 10] Physical data 1H—NMR 6 rmm1<CDCIQ : 0.90—2.95 (24H, m), 6 85- 7.40 (4H, no, 7.62 (1H, m), 7.72 (2H, m), 7.75 (1H, brs) MS PCI, m/z) : 646 <M+H)+ lH—NMR 5 ppm (CDClB) I 1.05-2.95 (24H, no, 6.80- 7.25 (4H, m), 7.68 (1H, m), 7.74 (1H, no, 7.78 (1H, brs) MS (ESI_APCI, m/z) : 674 (M+H)+ 1H—NMR 5 ppm (CDClB) I 1 05—2 95 (27H, m), 6.90- 7.25 (4H, m), 7.65 (1H, m), 7.69 (1H, m), 7.78 (1H, brs) MS (ESI_APCI, m/z) : 653 (M+H)+ 1H—NMR 6 ppm (CD013) I 1. 05-2. 95 (25H, m), 4. 40- 4.50(1H, m), 4. 60-4. 75 (1H, m), 6. 90-7. 25 (4H, m), 7.61 (1H, m), 7.66 (1H, m), 7.79 (1H, brs) MS (ESLAPCI, m/z) I 669 (M+H)+ [Table 11] Physical data 1H-NMR 5 ppm (CDClQ 2 1.10—2.90 (24H, m), 6.85— 7.25 (4H, m), 7.61 (1H, m), 7.69 (1H, HO, 7.77 (1H, brs) MS (ESI_APCI, m/Z) I 664 (M+H) + 1H-NMR 5 ppm (CDClQ I 1.20—2.95 (24H, m), 6.85— 7.25 (4H, m), 7.62 (1H, m), 7.69 (1H, m), 7.77 (1H, brs) MS (ESI_APCI, m/Z) I 664 (M+H) + Test Example 1 Affinity for human NK1 receptor (1) Preparation of human NK1 receptor expression vector PCR was performed using human adult normal tissue-derived brain cDNA (BioChain) as the te, with the forward primer of SEQ ID NO:1 and the reverse primer of SEQ ID NO:2, using a PCR enzyme, PrimeSTAR Max DNA Polymerase or PrimeSTAR GXL DNA Polymerase (registered trademark, Takara Bio). The amplified product was inserted into a plasmid luntII-TOPO tered trademark), Life Technologies) using Zero Blunt PCR Cloning Kit (registered trademark, Life Technologies). By a l method, Escherichia coli (One Shot TOP10 competent cells, Life Technologies) was ormed by the plasmid into which the amplified product had been inserted. The Escherichia coli cells were cultured on an LB agar medium containing 50 ug/mL kanamycin for a day. After the culture, a colony was selected and cultured in an LB medium containing 50 ug/mL of kanamycin. After the culture, the d was purified using Quantum Prep Plasmid Miniprep Kit ad).
The plasmid was double digested for about two hours using restriction enzymes, XhoI and HindIII (New England Biolabs). Then, ophoresis using 1% agarose gel was performed, and the fragment that was cleaved was collected and purified using TaKaRa RICOCHIP (Takara Bio). Separately, a plasmid was also purified from Escherichia coli that had been transformed by a vector (pcDNA3.1(—) (registered trademark), Life Technologies), and the plasmid was double digested for about two hours using restriction enzymes, XhoI and HindIII (New England Biolabs). Then, electrophoresis using 1% agarose gel was performed, and the vector that was cleaved was ted and purified using TaKaRa RICOCHIP (Takara Bio). The fragment cut out of unt-II and the pcDNA3.1(-) vector d with the restriction enzymes were ligated using DNA Ligation Kit <Mighty Mix> (Takara Bio). By a general method, Escherichia coli (One Shot TOPl O competent cells, Life Technologies) was transformed by the plasmid obtained by the ligation. The Escherichia coli cells were cultured on an LB agar medium containing 50 ug/mL of ampicillin for a day. After the e, a colony was selected and ed in an LB medium containing 50 ug/mL of ampicillin, and then the plasmid was purified using Quantum Prep Plasmid ep Kit (Bio-Rad). The protein—encoding nucleotide sequence (SEQ ID NO:3) of the obtained plasmid was completely identical to the nucleotide sequence (NM_001 058.3) of human tachykinin receptor 1 (TACRl , NKlR) registered on a known database (NCBI). Therefore, it was confirmed that the cloned gene sequence was the nucleotide sequence ofhuman NK1 receptor and that the amino acid sequence which would be translated was human NK1 or. The .1(-) (registered trademark) into which the nucleotide sequence of SEQ ID NO:3 was inserted was used as the human NK1 receptor expression plasmid. [01 34] (2) ation of human NK1 receptor—expressing cells (2-1) Culture of 293T cells Using a liquid D-MEM (Dulbecco’s Modified Eagle Medium) medium (low glucose, containing L-glutamine, Wako Pure al Industries) mented with an antibiotic penicillin—streptomycin solution (Life Technologies, final penicillin concentration of 100 U/mL and final streptomycin concentration of 100 ug/mL) and fetal bovine serum (final concentration of 10%), 293T cells (RIKEN) were cultured in an tor under the condition of 5% C02 gas at 37°C. ' (2-2) Subculture of 293T cells Almost nt cells were washed with PBS (Phosphate Buffered Saline, Wako Pure Chemical Industries), detached using 0.05% trypsin-EDTA (Life Technologies) and suspended in the liquid medium. The cell suspension was diluted . with the above liquid medium in such a manner that the spread ratio became 1:10, and then the cells were cultured. (2-3) Preparation for human NK1 receptor—expressing cells Confluent cells were washed with PBS, detached using 0.05% trypsin-EDTA (Life Technologies) and suspended in a liquid D-MEM medium (low glucose, containing L—glutamine, Wako Pure Chemical ries) supplemented with fetal bovine serum (final concentration of 10%). The cell suspension was diluted with the liquid medium, and the cells were seeded into the wells of a poly-D-lysine-coated 96- well microplate (BD Biocoat (registered trademark), Nippon Becton Dickinson) at a density of 5x104 cells/well and a liquid medium volume of 100 uL/well. After seeding, the cells were cultured in an tor under the condition of 5% C02 gas at 37°C for about four to five hours, and the cells to be transfected with the human NK1 receptor expression plasmid were thus prepared. (2-4) Transfection of human NK1 receptor expression plasmid into 293T cells For the transfection of the human NK1 receptor expression plasmid, Lipofectamine 2000 (registered trademark, Life logies) was used. The human NK1 receptor expression plasmid was diluted with Opti-MEM (registered trademark) I Reduced-Serum Medium (Life Technologies) to a concentration resulting in 0.2 ug/25 uL/well. At the same time, Lipofectamine 2000 (registered trademark, Life Technologies) was diluted with Opti-MEM (registered ark) I Reduced-Serum Medium (Life Technologies) to a concentration resulting in 0.4 uL/25 uL/well and incubated at room temperature for five minutes. After five minutes, to form a complex ofhuman NK1 receptor expression plasmid/Lipofectamine 2000, the diluted human NK1 receptor expression d and the d Lipofectamine 2000 were mixed and incubated at room temperature for 20 to 25 minutes. After the incubation, 50 uL/well of the complex solution was added to the cells to be transfected with the human NK1 receptor expression plasmid, and the cells were cultured in an incubator under the condition of 5% C02 gas at 37°C for about 48 hours. The cells that were cultured for 48 hours were used for the assays as the human NK1 receptor-expressing cells. [0 1 3 5] (3) Measurement of binding y to human NKI receptor (3-1) Preparation of membrane fraction from human NKI or-expressing cells Human NK1 receptor—expressing cells were prepared in a 1750m2 culture flask (Nippon Becton Dickinson). The formation of a complex of the human NK1 receptor sion plasmid and Lipofectamine 2000 was performed by calculating the culture area ratio and increasing the scale of the method described in the above 2-4 by the ratio.
The human NK1 receptor-expressing cells were ted in a buffer solution for the membrane on preparation (50 mM Tris (Wako Pure Chemical), 120 mM sodium de (Wako Pure Chemical Industries), 5 mM potassium chloride (Wako Pure Chemical Industries), 1 mM ethylenediaminetetraacetic acid (Sigma), 0.002 mg/rnL tatin (Peptide Institute), 0.04 mg/ bacitracin (Wako Pure Chemical Industries), 0.005 mg/mL phosphoramidon (Peptide Institute) and 0.5 mM phenylmethylsulfonyl fluoride (Wako Pure Chemical Industries), pH7.4) and centrifuged at 1,880 g for 10 s, and the cell sediment was ded in the buffer solution for the membrane fraction ation. After freezing and g the cells once, the cells were homogenized using a Dounce-type homogenizer (cooled on ice, 1000 rpm, 20 times).
The homogenized cell suspension was centrifuged at 20,000 rpm for 10 minutes, and the supernatant was removed to obtain cell sediment. The cell sediment was suspended again in the buffer solution for the membrane fraction preparation and homogenized using a Dounce—type homogenizer (cooled on ice, 1000 rpm, 30 times). The cell sion was centrifuged at 20,000 rpm for 10 minutes, and the supernatant was removed to obtain cell sediment. The same homogenization and centrifugation were repeated again, and final cell sediment was obtained. The final cell sediment was suspended in a buffer solution for the receptor binding test (50 mM Tris (Wako Pure Chemical Industries), 3 mM manganese chloride (Wako Pure Chemical Industries), 0.002 Ing/mL chymostatin (Peptide Institute), 0.04 mg/ bacitracin (Wako Pure Chemical Industries) and 0.02% bovine serum n (Sigma), pH 7.4), and the protein concentration was measured using BCA Protein Assay Kit e). (3 -2) Receptor binding test The buffer on for the receptor binding test was dispensed to the wells of a 96-well assay plate (Greiner) at 22.5 uL/well. DMSO solutions of a test compound, which were prepared at an 80-time higher concentration using 100% dimethyl sulfoxide (DMSO), were added to the wells at 2.5 uL/well (final concentrations of 1 nM to 100 nM), and the solutions were mixed. As a radiolabeled ligand, 125I-substance P ance P, [1251]Tyr8-, PerkinElmer) was used. 125I-substance P was diluted with the buffer solution for the receptor binding test to a concentration ing in 125 pmol/25 uL/well and added to the 96—well assay plate, and the solutions were mixed. The membrane fraction prepared from the human NK1 receptor-expressing cells was diluted with the buffer solution for the receptor binding test to a concentration resulting in 8 to rig/well, suspended until the suspension became in such a homogenous state that the suspension could flow through a 27G injection needle smoothly and then added to the 96-well assay plate at 150 uL/well. Then, the plate was incubated at room temperature for 60 minutes while shaking the plate. The reaction solutions were n-filtered through a multiscreen 96-well filter plate (Millipore) which had been pre—treated with 0.3% polyethyleneimine, and the on was terminated by washing with a washing solution (50 mM Tris and 0.02% bovine serum albumin, pH 7.4) four times. The bottom of the microplate was dried at 60°C, and then 100 uL/well of MicroScint 20 (PerkinElmer) was dispensed to the wells. The top of the plate was sealed with TopSeal A (PerkinElmer), and the plate was shaken for 5 to 10 minutes. Then, the radioactivities were measured with nt NXT (registered ark) (PerkinElmer). The radioactivity of each well was calculated by subtracting the radioactivity of the well to which 10 14M aprepitant was added (non-specific binding).
The g rate (%) of 125I-substance P = (the radioactivity of the group to which the test compound was added) / (the radioactivity of the group to which the vehicle was added) x100 was calculated. Using analysis re, GraphPad Prism (GraphPad Software), the binding rate (%) was plotted against the concentration of the test compound andlinearly approximated, and the tration required for 50% inhibition, ICso, was calculated. These results were shown in Table 12 and 13. In the table, Ex. No. means the Example number, and IC50 (nM) is the tration required for 50% inhibition. (4) Results [01 3 6] [Table 12] [013 7] [Table 13] As shown in Table 12 and 13, it was demonstrated that the compounds of the t invention t a high binding affinity for human NK1 receptor. [0 1 3 8] Test Example 2 Inhibitory effect on human NKl receptor (1) Preparation of human NK1 receptor-expressing cells Human NK1 receptor-expressing cells were prepared by the same methods as those described in 2-3 of Test Example 1. (2) Study on inhibitory effect on increase in intracellular calcium concentration The human NK1 receptor-expressing cells were washed with 300 uL/well of a washing solution (20 mM HEPES/Hank’s Balanced Salt on (HBSS) pH 7.3). A fluorescent calcium indicator (Fluo-4 Direct Calcium Assay Kit, Life logies, containing 0.42 mM probenecid and 0.1% bovine serum albumin, prepared according to the protocol of the product) was added to the wells at 150 uL/well, and the plate was ted at 37°C for 30 s in an incubator. Then, DMSO ons of a test compound, which were prepared at an e higher concentration using 100% dimethyl sulfoxide (DMSO), were added to the wells at 2.5 uL/well (final concentrations of 0.1, 1 and 10 uM), and the solutions were mixed. Then, the plate was further incubated at 37°C for 30 minutes in an incubator. After 30 minutes, the ellular calcium concentrations were measured immediately.
The intracellular calcium concentrations were each measured as a fluorescent signal using FDSS (registered trademark) 7000 (Hamamatsu Photonics). A substance P (Peptide Institute, Inc.) solution which was prepared at 0.4 uM or 4 uM using an assay buffer (20 mM HEPES/Hank’s Balanced Salt Solution (HBSS) pH 7.3, containing 0.1% bovine serum albumin) was added automatically to each well at 50 uL/well (final concentration of 0.1 or 1 uM) 10 seconds after starting reading, and the fluorescent signal was measured up to 120 seconds.
The intracellular calcium concentration (%) of the cells to which a test compound was added was calculated by the equation below, where the fluorescent signal of the group to which the vehicle (DMSO) was added was regarded as 100%, and the fluorescent signal before the addition of substance P was regarded as 0%.
Intracellular calcium concentration (%) = (Fluorescent signal of test compound addition group) / (Fluorescent signal of vehicle addition group) X100 The intracellular calcium tration (%) calculated was regarded as the remaining agonist activity of substance P ance P—Response Remaining: SPRR).
These results were shown in Table 14 and 15. In the table, Ex. No. means the Example number. SPRR (%) is the value obtained when the concentration of substance P was 1 ”M and the concentration of the compound was 0.1 uM. (3) Results [Table 14] [Table '15] As shown in Table 14 and 15, it was demonstrated that the compounds of the present invention exhibit a potent human NK1 or antagonist activity. [0 1 42] Test Example 3 Inhibitory effect on CYP3A4 A dimethyl sulfoxide (DMSO) solution of a test compound with a concentration 1000 times higher than the evaluation concentration was prepared, and a reaction solution was prepared by diluting the solution. Enzyme reaction was performed by incubating in a potassium ate buffer solution (pH 7.4) ning 1 nM to 20 uM test compound, 3.2 mM magnesium chloride, 0.2 pmol human CYP3A4 (BD Biosciences), 0.5 mM reduced nicotinamide adenine dinucleotide phosphate (NADPH) and 3 uM Luciferin—IPA ga) at 37°C for 10 minutes. The volume of the reaction solution was 50 uL/well. The 30-minute pre-incubation group was incubated at 37°C for minutes before adding the substrate, the Luciferin—IPA on (12.5 uL/well). At the end of the enzyme reaction, 50 uL/well of a Luciferin detection reagent (Promega) was added to the wells, and the plate was left at room temperature for 20 minutes. Then, the emission intensities were measured with te M1000 (TECAN). The enzyme activities (%) relative to the value of the group to which the test compound was not added were calculated. A dose-response curve was drawn using analysis software, GraphPad Prism (GraphPad Software), and the concentration of each compound that exhibited 50% inhibition, IC50, was calculated. As a ative example, aprepitant, which is an NK1 or antagonist, was tested in the same .
IC50 values of the 30-minute pre-incubation groups using the test compounds were measured by the above measurement method, and the s are shown in Table 16 and 17. In the table, EX. No. means the Example number, and IC50 (uM) is the concentration required for 50% inhibition.
[Table 16] Aprepitant [Table 17] As shown in Table 16 and 17, it was demonstrated that the CYP3A4-inhibitory activities of the compounds of the present invention are reduced as compared to that of aprepitant. Therefore, it is ed that the compounds of the t invention have fewer drug—drug interactions based on the inhibitory effect on CYP3A4 than aprepitant.
[0145] Test Example 4 Effect on foot-tapping (1) Effect on foot-tapping The test compound solution was prepared by dissolving the test compound in a vehicle (a mixture of 50% N,N—dimethylacetamide (Wako Pure Chemical Industries), % propylene glycol (Wako Pure Chemical Industries), 4% 2-hydroxypropyl-B- cyclodextrin (Wako Pure Chemical Industries) and 16% distilled water).
A male gerbil (Japan SLC) was anesthetized with isoflurane, and 0.1 mg/kg of a test compound was administered from the jugular vein. After four hours, GR73632 (5 pmol/S ul saline), which is an NK1 receptor t, was administered into the cerebral ventricle at the part 1 mm lateral to and 4.5 mm below the bregma in the head, under anesthesia with isoflurane. After the administration, the gerbil was moved to an observation cage, and the foot-tapping period during 30 minutes after the recovery of the righting reflex was ed. The foot-tapping tion rate (%) of each test nd was calculated by the following equation.
Foot-tapping inhibition rate (%) = {1 - (Foot-tapping period when test nd was stered) / (Foot-tapping period when t was administered)} x100
[0146] (2) Measurement of drug concentrations After foot-tapping was finished, laparotomy was performed ately under anesthesia with ether, and a blood sample was taken from the abdominal vena cava. At the same time, the brain was extracted. Through a quantitative analysis using liquid chromatography-mass spectrometry (LC/MS), the concentrations of the test compound in the plasma and the brain were measured. (3) Results The effects on foot-tapping were measured by the above test method, and the results are shown in Table 18 and 19. In the table, Ex. No. means the Example number.
Inhibition (%) is the foot-tapping inhibition rate, and Conc. (nM) is the drug concentration in the brain.
[Table 18] Cone. (nM) [Table 19] Inhibition(/)00 As shown in Table 18 and 19, the compounds of the t invention were penetrated into the central s system and exhibited an excellent NK1 receptor antagonist activity also in vivo. [0 l 49] Test Example 5 Ferret pharmacokinetic test (1) Methods The test compound solution for intravenous administration was prepared by dissolving the test compound in a vehicle (a mixture of 50% N,N-dimethy1acetamide (Wako Pure Chemical Industries), 30% propylene glycol (Wako Pure Chemical Industries), 4% 2-hydroxypropyl-B-cyclodextrin (Wako Pure al Industries) and 16% distilled water). As an oral administration on, the suspension (0.5% methylcellulose ) was used.
Under anesthesia with isoflurane, 0.1 mg/kg of the test compound was intravenously administered to a male ferret (Marshall BioResources Japan) from the l vein. In the case of oral administration, 1 mg/kg of the test compound was orally administered to an awake animal. After the administration of the test compound, blood samples were taken sequentially from the brachial cephalic vein up to 7 days after the administration. Through a quantitative analysis using liquid chromatography-mass spectrometry (LC/MS), the concentrations of the test compound in the plasma were ed. (2) Results The pharmacokinetic test in a ferret was tested by the above test method, and the results are shown in Table 20 and Table 21. In the tables, Ex. No. means the e number. t1/2, CLtot and Vss are the half-life, the total body clearance and the steady-state volume of distribution, based on the plasma concentrations in the case of intravenous administration, respectively. Cmax, AUC and BA are the maximum plasma test compound concentration, the area under the plasma test compound tration- time curve within 7days after the administration and the bioavailability, in the case of oral administration, respectively. [01 5 0] [Table 20] Ex.No. CLtot(mL/min/kg) Vs s (mL/kg) 1 3 2. 4 8 9 [Table 21] Ex. No. Cmax (ng/mL) AUC (ng - mi n/mL) BA (%) 13 1,258 5,294,066 m As shown in Table 20 and Table 21, the compound of the present invention exhibited an excellent oral absorbability with low clearance. [0 1 52] Test Example 6 Effect on Cisplatin-induced acute and delayed emetic response (1) Methods The test compound solution was prepared by dissolving the test compound in a vehicle (a mixture of 50% N,N—dimethylacetamide (Wako Pure Chemical Industries), % propylene glycol (Wako Pure Chemical ries), 4% 2-hydroxypropyl-B- cyclodextrin (Wako Pure Chemical Industries) and 16% distilled water). The vehicle only was administered to the control group.
Under anesthesia with isoflurane, 0.01 mg/kg or 0.1mg/kg of the test compound was intravenously administered to a male ferret (Marshall ources Japan) from the jugular vein. tin in , which was heated to 40-50°C, was intraperitoneally administered at 5 mg/kg one hour after the drug administration. The ferret was observed for 72 hours from immediately after the Cisplatin administration, and the number of retching (periodic abdominal contraction without vomiting of the gastric t) and vomiting was counted. (2) s The results are shown in Figure 1. In the control group, an increase in the number of retching and vomiting was ed in the acute phase (up to 24 hours after the Cisplatin administration) and in the delayed phase (24 hours to 72 hours after the Cisplatin administration). In the group to which the compound of Example 13 was intravenously administered, the inhibition of the number of retching and vomiting was observed in the acute phase and in the delayed phase.
It was demonstrated that the compound of the present invention has a long- acting medicinal effect and an inhibitory effect on the cisplatin—induced acute and delayed emetic responses.
Test Example 7 Evaluation ofhERG current ( l ) Methods A dimethyl sulfoxide (DMSO) solution of the test compound with a concentration 1000 times higher than the evaluation concentration (10 HM) was ed, and a solution with a final ation concentration was prepared by diluting the solution. The hERG current was measured by a whole-cell method using a patch clamp system, where a cover glass on which hERG channel-expressing human embryonic kidney (HEK) 293 cells were seeded was placed on a perfiasion bath and a perfusion solution was caused to flow. The change in the hERG channel-derived current caused by a pulse protocol (holding potential of -80 mV, depolarization pulse of +20 mV for 1.9 seconds, repolarization pulse of —50 mV for 2 seconds, stimulated at intervals of 15 seconds) of data acquisition/analysis re, pCLAMP9 (Axon Instruments, Inc.), was measured. The measurement conditions were a flow rate of about 1.5 mL/min and a temperature of about 33°C. Two wave forms immediately before applying the test compound and two wave forms immediately after the application for 10 minutes were analyzed, and the statistical analysis was performed.
The value before the ation of the test compound was ed as 100%, and the change rate based on the value was ined. (2) Results The effect of the test compound on hERG t was evaluated by the above method (the result is shown in Table 22). In the table, Ex. No. means the Example number, and the change rate is the average at the standard error.
[Table 22] Ex.No. % (n=3) 1 3 8 1. 1 i 5 . 6 l 4 8 O. 3 i" 3 . 9 The compound of the present ion did not cause any change in the hERG current with a statistical significance compared to the vehicle control (0.1% DMSO).
INDUSTRIAL APPLICABILITY The compounds of the present invention or pharmaceutically acceptable salts thereof have an excellent NK1 receptor antagonist activity, and thus are also useful as an agent for the prevention or treatment of cancer-chemotherapy—induced nausea and vomiting.
SEQUENCE LISTING FREE TEXT [0 1 5 6] <Sequence listing 1> SEQ ID NO:1 is the sequence of forward primer which was used for DNA amplification of SEQ ID NO:3. nce listing 2> SEQ ID NO:2 is the ce of reverse primer which was used for DNA amplification of SEQ ID NO:3.

Claims (1)

1. A compound represented by the formula (1): [Chem. 1] N X ( I ) R‘ (R1 5 n ring A is a group represented by the following formula: [Chem.2] or)? X is a hydrogen atom, cyano, halogen, C1_6 alkyl or hydroxymethyl; 10 R1 is a group represented by the following formula: [Chem.3] 0 R11: R1a 0!" wherein R1a and R11) are each independently any one of a en atom, a fluorine atom or C1
NZ726027A 2014-05-07 2015-05-07 Cyclohexyl pyridine derivative NZ726027B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2014095776 2014-05-07
JP2014-095776 2014-05-07
PCT/JP2015/063154 WO2015170693A1 (en) 2014-05-07 2015-05-07 Cyclohexyl-pyridine derivative

Publications (2)

Publication Number Publication Date
NZ726027A NZ726027A (en) 2021-11-26
NZ726027B2 true NZ726027B2 (en) 2022-03-01

Family

ID=

Similar Documents

Publication Publication Date Title
US10011568B2 (en) Cyclohexyl pyridine derivative
AU2014347644B2 (en) Carboxymethyl piperidine derivative
TW201028424A (en) 2-pyridin-2-yl-pyrazol-3(2h)-one derivatives, preparation and therapeutic use thereof
CA2937012A1 (en) Fused pyrazole derivative
NZ726027B2 (en) Cyclohexyl pyridine derivative
JP6609253B2 (en) Cyanothiophene derivatives
US10399949B2 (en) NK1 receptor antagonist