MXPA06006820A - Methods for treating retinoid responsive disorders using selective inhibitors of cyp26a and cyp26b - Google Patents
Methods for treating retinoid responsive disorders using selective inhibitors of cyp26a and cyp26bInfo
- Publication number
- MXPA06006820A MXPA06006820A MXPA/A/2006/006820A MXPA06006820A MXPA06006820A MX PA06006820 A MXPA06006820 A MX PA06006820A MX PA06006820 A MXPA06006820 A MX PA06006820A MX PA06006820 A MXPA06006820 A MX PA06006820A
- Authority
- MX
- Mexico
- Prior art keywords
- disorder
- mmol
- carbons
- selective
- lower alkyl
- Prior art date
Links
- 230000002401 inhibitory effect Effects 0.000 title claims abstract description 195
- 239000003112 inhibitor Substances 0.000 title claims abstract description 190
- 201000010099 disease Diseases 0.000 title claims abstract description 139
- 150000004492 retinoid derivatives Chemical class 0.000 title claims abstract description 96
- 125000000217 alkyl group Chemical group 0.000 claims description 302
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 95
- 125000003545 alkoxy group Chemical group 0.000 claims description 68
- 125000003342 alkenyl group Chemical group 0.000 claims description 59
- 125000000304 alkynyl group Chemical group 0.000 claims description 45
- 206010000496 Acne Diseases 0.000 claims description 42
- 125000004005 formimidoyl group Chemical group [H]\N=C(/[H])* 0.000 claims description 42
- 239000000203 mixture Substances 0.000 claims description 40
- 239000011780 sodium chloride Substances 0.000 claims description 36
- 208000006641 Skin Disease Diseases 0.000 claims description 35
- 150000003839 salts Chemical class 0.000 claims description 34
- 150000002148 esters Chemical class 0.000 claims description 29
- 125000001188 haloalkyl group Chemical group 0.000 claims description 26
- 208000009856 Lung Disease Diseases 0.000 claims description 24
- 206010003816 Autoimmune disease Diseases 0.000 claims description 23
- 208000009025 Nervous System Disease Diseases 0.000 claims description 22
- 206010029305 Neurological disorder Diseases 0.000 claims description 22
- 200000000018 inflammatory disease Diseases 0.000 claims description 22
- 230000002062 proliferating Effects 0.000 claims description 22
- 229910052760 oxygen Inorganic materials 0.000 claims description 21
- 229910052717 sulfur Inorganic materials 0.000 claims description 21
- 150000001408 amides Chemical class 0.000 claims description 20
- 208000009745 Eye Disease Diseases 0.000 claims description 18
- 241000282414 Homo sapiens Species 0.000 claims description 9
- 125000001316 cycloalkyl alkyl group Chemical group 0.000 claims description 4
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 4
- 229940004296 Formula 21 Drugs 0.000 claims description 2
- 125000001475 halogen functional group Chemical group 0.000 claims 8
- 239000000126 substance Substances 0.000 abstract description 11
- XEKOWRVHYACXOJ-UHFFFAOYSA-N acetic acid ethyl ester Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 495
- 239000000543 intermediate Substances 0.000 description 310
- 150000001875 compounds Chemical class 0.000 description 234
- VLKZOEOYAKHREP-UHFFFAOYSA-N hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 188
- 239000000243 solution Substances 0.000 description 183
- 239000011541 reaction mixture Substances 0.000 description 171
- WYURNTSHIVDZCO-UHFFFAOYSA-N tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 128
- -1 retinoic acid (RA) Chemical class 0.000 description 125
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 120
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 116
- OKKJLVBELUTLKV-UHFFFAOYSA-N methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 114
- ZMANZCXQSJIPKH-UHFFFAOYSA-N triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 108
- 239000000741 silica gel Substances 0.000 description 106
- 229910002027 silica gel Inorganic materials 0.000 description 106
- 239000008079 hexane Substances 0.000 description 99
- 239000003921 oil Substances 0.000 description 96
- 235000019198 oils Nutrition 0.000 description 96
- YNHIGQDRGKUECZ-UHFFFAOYSA-L Bis(triphenylphosphine)palladium(II) dichloride Chemical compound [Cl-].[Cl-].[Pd+2].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 YNHIGQDRGKUECZ-UHFFFAOYSA-L 0.000 description 86
- 239000003480 eluent Substances 0.000 description 86
- YMWUJEATGCHHMB-UHFFFAOYSA-N methylene dichloride Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 86
- 239000012074 organic phase Substances 0.000 description 85
- 229910052731 fluorine Inorganic materials 0.000 description 83
- 101700067048 CDC13 Proteins 0.000 description 81
- RTZKZFJDLAIYFH-UHFFFAOYSA-N diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 78
- CSNNHWWHGAXBCP-UHFFFAOYSA-L mgso4 Chemical compound [Mg+2].[O-][S+2]([O-])([O-])[O-] CSNNHWWHGAXBCP-UHFFFAOYSA-L 0.000 description 76
- HEMHJVSKTPXQMS-UHFFFAOYSA-M sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 76
- 238000003818 flash chromatography Methods 0.000 description 73
- VEXZGXHMUGYJMC-UHFFFAOYSA-N HCl Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 69
- 239000007787 solid Substances 0.000 description 68
- 239000012267 brine Substances 0.000 description 63
- 238000000034 method Methods 0.000 description 63
- KWYUFKZDYYNOTN-UHFFFAOYSA-M potassium hydroxide Chemical compound [OH-].[K+] KWYUFKZDYYNOTN-UHFFFAOYSA-M 0.000 description 59
- 239000002253 acid Chemical class 0.000 description 58
- 239000000047 product Substances 0.000 description 58
- HEDRZPFGACZZDS-MICDWDOJSA-N deuterated chloroform Substances [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 53
- 125000001153 fluoro group Chemical group F* 0.000 description 53
- 239000011737 fluorine Substances 0.000 description 52
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 51
- 239000003153 chemical reaction reagent Substances 0.000 description 49
- 229910052794 bromium Inorganic materials 0.000 description 44
- 229910052801 chlorine Inorganic materials 0.000 description 44
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 44
- 238000004440 column chromatography Methods 0.000 description 42
- 229910052740 iodine Inorganic materials 0.000 description 41
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 40
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 39
- 239000003039 volatile agent Substances 0.000 description 38
- 125000001072 heteroaryl group Chemical group 0.000 description 36
- 210000004027 cells Anatomy 0.000 description 35
- 239000007864 aqueous solution Substances 0.000 description 34
- 125000004414 alkyl thio group Chemical group 0.000 description 33
- SHGAZHPCJJPHSC-YCNIQYBTSA-N Retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 description 32
- QTBSBXVTEAMEQO-UHFFFAOYSA-N acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 32
- WEVYAHXRMPXWCK-UHFFFAOYSA-N acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 32
- 230000000694 effects Effects 0.000 description 32
- 150000004702 methyl esters Chemical class 0.000 description 32
- 238000006243 chemical reaction Methods 0.000 description 31
- WMFOQBRAJBCJND-UHFFFAOYSA-M lithium hydroxide Chemical compound [Li+].[OH-] WMFOQBRAJBCJND-UHFFFAOYSA-M 0.000 description 31
- 229960001727 Tretinoin Drugs 0.000 description 30
- 229910052736 halogen Inorganic materials 0.000 description 28
- 150000002367 halogens Chemical group 0.000 description 28
- PMZURENOXWZQFD-UHFFFAOYSA-L na2so4 Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 28
- 229930002330 retinoic acid Natural products 0.000 description 28
- 229910052938 sodium sulfate Inorganic materials 0.000 description 28
- 235000011152 sodium sulphate Nutrition 0.000 description 28
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonium chloride Substances [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 26
- 235000019270 ammonium chloride Nutrition 0.000 description 25
- QDHHCQZDFGDHMP-UHFFFAOYSA-N monochloramine Chemical compound ClN QDHHCQZDFGDHMP-UHFFFAOYSA-N 0.000 description 25
- 229910000027 potassium carbonate Inorganic materials 0.000 description 25
- 235000002639 sodium chloride Nutrition 0.000 description 25
- LSXDOTMGLUJQCM-UHFFFAOYSA-M Copper(I) iodide Chemical compound I[Cu] LSXDOTMGLUJQCM-UHFFFAOYSA-M 0.000 description 22
- XKRFYHLGVUSROY-UHFFFAOYSA-N argon Chemical compound [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 description 22
- GBRBMTNGQBKBQE-UHFFFAOYSA-L copper;diiodide Chemical compound I[Cu]I GBRBMTNGQBKBQE-UHFFFAOYSA-L 0.000 description 22
- 239000001184 potassium carbonate Substances 0.000 description 22
- CSCPPACGZOOCGX-UHFFFAOYSA-N acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 21
- 238000010171 animal model Methods 0.000 description 20
- 201000011510 cancer Diseases 0.000 description 20
- 125000004076 pyridyl group Chemical group 0.000 description 20
- DIOHEXPTUTVCNX-UHFFFAOYSA-N 1,1,1-trifluoro-N-phenyl-N-(trifluoromethylsulfonyl)methanesulfonamide Chemical compound FC(F)(F)S(=O)(=O)N(S(=O)(=O)C(F)(F)F)C1=CC=CC=C1 DIOHEXPTUTVCNX-UHFFFAOYSA-N 0.000 description 18
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 18
- 150000001768 cations Chemical class 0.000 description 18
- 125000002541 furyl group Chemical group 0.000 description 18
- 230000014509 gene expression Effects 0.000 description 18
- 125000001624 naphthyl group Chemical group 0.000 description 18
- 125000001544 thienyl group Chemical group 0.000 description 18
- 239000002585 base Substances 0.000 description 17
- 125000002971 oxazolyl group Chemical group 0.000 description 17
- 125000003373 pyrazinyl group Chemical group 0.000 description 17
- 125000003226 pyrazolyl group Chemical group 0.000 description 17
- 125000002098 pyridazinyl group Chemical group 0.000 description 17
- 125000000714 pyrimidinyl group Chemical group 0.000 description 17
- 125000000335 thiazolyl group Chemical group 0.000 description 17
- 125000004494 ethyl ester group Chemical group 0.000 description 16
- 125000002883 imidazolyl group Chemical group 0.000 description 16
- 239000002904 solvent Substances 0.000 description 15
- 210000001519 tissues Anatomy 0.000 description 15
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 14
- CWMFRHBXRUITQE-UHFFFAOYSA-N Trimethylsilylacetylene Chemical group C[Si](C)(C)C#C CWMFRHBXRUITQE-UHFFFAOYSA-N 0.000 description 14
- 239000000284 extract Substances 0.000 description 14
- 239000008194 pharmaceutical composition Substances 0.000 description 14
- VZWXIQHBIQLMPN-UHFFFAOYSA-N Chromane Chemical class C1=CC=C2CCCOC2=C1 VZWXIQHBIQLMPN-UHFFFAOYSA-N 0.000 description 13
- 125000002534 ethynyl group Chemical group [H]C#C* 0.000 description 13
- 239000000706 filtrate Substances 0.000 description 13
- 230000001603 reducing Effects 0.000 description 13
- WPWNEKFMGCWNPR-UHFFFAOYSA-N 3,4-dihydro-2H-thiochromene Chemical class C1=CC=C2CCCSC2=C1 WPWNEKFMGCWNPR-UHFFFAOYSA-N 0.000 description 12
- CXWXQJXEFPUFDZ-UHFFFAOYSA-N Tetralin Chemical class C1=CC=C2CCCCC2=C1 CXWXQJXEFPUFDZ-UHFFFAOYSA-N 0.000 description 12
- 239000012071 phase Substances 0.000 description 12
- CDBYLPFSWZWCQE-UHFFFAOYSA-L sodium carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 12
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 11
- 229960000583 Acetic Acid Drugs 0.000 description 11
- KEIFWROAQVVDBN-UHFFFAOYSA-N Dialin Chemical class C1=CC=C2C=CCCC2=C1 KEIFWROAQVVDBN-UHFFFAOYSA-N 0.000 description 11
- 229910052786 argon Inorganic materials 0.000 description 11
- 210000003491 Skin Anatomy 0.000 description 10
- 230000004069 differentiation Effects 0.000 description 10
- 125000005647 linker group Chemical group 0.000 description 10
- 230000004048 modification Effects 0.000 description 10
- 238000006011 modification reaction Methods 0.000 description 10
- MZRVEZGGRBJDDB-UHFFFAOYSA-N n-butyllithium Chemical compound [Li]CCCC MZRVEZGGRBJDDB-UHFFFAOYSA-N 0.000 description 10
- 238000002560 therapeutic procedure Methods 0.000 description 10
- YXFVVABEGXRONW-UHFFFAOYSA-N toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 10
- 235000011054 acetic acid Nutrition 0.000 description 9
- 150000003530 tetrahydroquinolines Chemical class 0.000 description 9
- LVEYOSJUKRVCCF-UHFFFAOYSA-N 1,3-Bis(diphenylphosphino)propane Chemical compound C=1C=CC=CC=1P(C=1C=CC=CC=1)CCCP(C=1C=CC=CC=1)C1=CC=CC=C1 LVEYOSJUKRVCCF-UHFFFAOYSA-N 0.000 description 8
- VSCWAEJMTAWNJL-UHFFFAOYSA-K Aluminium chloride Chemical compound Cl[Al](Cl)Cl VSCWAEJMTAWNJL-UHFFFAOYSA-K 0.000 description 8
- 206010022114 Injury Diseases 0.000 description 8
- INQOMBQAUSQDDS-UHFFFAOYSA-N Methyl iodide Chemical compound IC INQOMBQAUSQDDS-UHFFFAOYSA-N 0.000 description 8
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 8
- UHOVQNZJYSORNB-UHFFFAOYSA-N benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 8
- 238000005859 coupling reaction Methods 0.000 description 8
- HTJDQJBWANPRPF-UHFFFAOYSA-N cyclopropylamine Chemical compound NC1CC1 HTJDQJBWANPRPF-UHFFFAOYSA-N 0.000 description 8
- 125000005843 halogen group Chemical group 0.000 description 8
- ZYVQBFJEQWSNFU-UHFFFAOYSA-N 6-bromo-2,2,4,4-tetramethyl-3H-chromene-8-carboxylic acid Chemical compound BrC1=CC(C(O)=O)=C2OC(C)(C)CC(C)(C)C2=C1 ZYVQBFJEQWSNFU-UHFFFAOYSA-N 0.000 description 7
- SKRDXYBATCVEMS-UHFFFAOYSA-N Isopropyl nitrite Chemical compound CC(C)ON=O SKRDXYBATCVEMS-UHFFFAOYSA-N 0.000 description 7
- 241000124008 Mammalia Species 0.000 description 7
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 7
- 238000004305 normal phase HPLC Methods 0.000 description 7
- 210000000056 organs Anatomy 0.000 description 7
- 230000000699 topical Effects 0.000 description 7
- YHOLDFRZCVNIHN-UHFFFAOYSA-N 2-[4-[2-(8-hydroxy-2,2,4,4-tetramethyl-3H-chromen-6-yl)ethynyl]phenyl]propanoic acid Chemical compound C1=CC(C(C(O)=O)C)=CC=C1C#CC1=CC(O)=C(OC(C)(C)CC2(C)C)C2=C1 YHOLDFRZCVNIHN-UHFFFAOYSA-N 0.000 description 6
- IMLOMOSKLWEUJD-UHFFFAOYSA-N 2-[4-[2-[8-[[cyclopropyl(methyl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromen-6-yl]ethynyl]phenyl]acetic acid Chemical compound C1CC1N(C)CC(C1=C(C(CC(C)(C)O1)(C)C)C=1)=CC=1C#CC1=CC=C(CC(O)=O)C=C1 IMLOMOSKLWEUJD-UHFFFAOYSA-N 0.000 description 6
- JRDUNTDPOVKFND-UHFFFAOYSA-N 6-[2-[4-(carboxymethyl)phenyl]ethynyl]-2,2,4,4-tetramethyl-3H-chromene-8-carboxylic acid Chemical compound C=1C(C(O)=O)=C2OC(C)(C)CC(C)(C)C2=CC=1C#CC1=CC=C(CC(O)=O)C=C1 JRDUNTDPOVKFND-UHFFFAOYSA-N 0.000 description 6
- 241000699800 Cricetinae Species 0.000 description 6
- 206010014561 Emphysema Diseases 0.000 description 6
- SHGAZHPCJJPHSC-NUEINMDLSA-N Isotretinoin Chemical compound OC(=O)C=C(C)/C=C/C=C(C)C=CC1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-NUEINMDLSA-N 0.000 description 6
- 229960005280 Isotretinoin Drugs 0.000 description 6
- GQYCWGJURVELKM-UHFFFAOYSA-N N-cyclopropyl-6-ethynyl-8-methoxy-N,4,4-trimethyl-2,3-dihydro-1H-naphthalen-1-amine Chemical compound COC1=CC(C#C)=CC(C(CC2)(C)C)=C1C2N(C)C1CC1 GQYCWGJURVELKM-UHFFFAOYSA-N 0.000 description 6
- 206010028980 Neoplasm Diseases 0.000 description 6
- BEOOHQFXGBMRKU-UHFFFAOYSA-N Sodium cyanoborohydride Chemical compound [Na+].[B-]C#N BEOOHQFXGBMRKU-UHFFFAOYSA-N 0.000 description 6
- 230000001808 coupling Effects 0.000 description 6
- 238000010168 coupling process Methods 0.000 description 6
- 238000001914 filtration Methods 0.000 description 6
- 239000000499 gel Substances 0.000 description 6
- AMJDEFLNSBJNAT-UHFFFAOYSA-N methyl 2-(4-iodophenyl)-2-methylpropanoate Chemical compound COC(=O)C(C)(C)C1=CC=C(I)C=C1 AMJDEFLNSBJNAT-UHFFFAOYSA-N 0.000 description 6
- 230000002093 peripheral Effects 0.000 description 6
- 235000017557 sodium bicarbonate Nutrition 0.000 description 6
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 6
- 239000001187 sodium carbonate Substances 0.000 description 6
- 229910000029 sodium carbonate Inorganic materials 0.000 description 6
- 238000003756 stirring Methods 0.000 description 6
- 125000001424 substituent group Chemical group 0.000 description 6
- 230000001225 therapeutic Effects 0.000 description 6
- AQRLNPVMDITEJU-UHFFFAOYSA-N triethylsilane Chemical compound CC[SiH](CC)CC AQRLNPVMDITEJU-UHFFFAOYSA-N 0.000 description 6
- DTQVDTLACAAQTR-UHFFFAOYSA-N trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 6
- AFABGHUZZDYHJO-UHFFFAOYSA-N 2-Methylpentane Chemical class CCCC(C)C AFABGHUZZDYHJO-UHFFFAOYSA-N 0.000 description 5
- NIEBHDXUIJSHSL-UHFFFAOYSA-N 4-iodobenzaldehyde Chemical compound IC1=CC=C(C=O)C=C1 NIEBHDXUIJSHSL-UHFFFAOYSA-N 0.000 description 5
- 210000004556 Brain Anatomy 0.000 description 5
- 206010030032 Ocular disorder Diseases 0.000 description 5
- YJVFFLUZDVXJQI-UHFFFAOYSA-L Palladium(II) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 5
- 206010037888 Rash pustular Diseases 0.000 description 5
- 210000001525 Retina Anatomy 0.000 description 5
- 210000001732 Sebaceous Glands Anatomy 0.000 description 5
- ITMCEJHCFYSIIV-UHFFFAOYSA-N Trifluoromethanesulfonic acid Chemical compound OS(=O)(=O)C(F)(F)F ITMCEJHCFYSIIV-UHFFFAOYSA-N 0.000 description 5
- 102000004965 antibodies Human genes 0.000 description 5
- 108090001123 antibodies Proteins 0.000 description 5
- 125000003118 aryl group Chemical group 0.000 description 5
- 238000004166 bioassay Methods 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 229910052799 carbon Inorganic materials 0.000 description 5
- 239000000969 carrier Substances 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 239000003814 drug Substances 0.000 description 5
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 5
- 230000002757 inflammatory Effects 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- IYIVBGQBEMSRLO-UHFFFAOYSA-N methyl 2-(4-iodophenyl)acetate Chemical compound COC(=O)CC1=CC=C(I)C=C1 IYIVBGQBEMSRLO-UHFFFAOYSA-N 0.000 description 5
- SLZYKGNMDRJRFP-UHFFFAOYSA-N methyl 2-(4-iodophenyl)propanoate Chemical compound COC(=O)C(C)C1=CC=C(I)C=C1 SLZYKGNMDRJRFP-UHFFFAOYSA-N 0.000 description 5
- KDLHZDBZIXYQEI-UHFFFAOYSA-N palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 5
- 239000000843 powder Substances 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 108090000623 proteins and genes Proteins 0.000 description 5
- 201000004681 psoriasis Diseases 0.000 description 5
- JUJWROOIHBZHMG-UHFFFAOYSA-N pyridine Chemical group C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 5
- 150000003254 radicals Chemical class 0.000 description 5
- 102000003702 retinoic acid receptors Human genes 0.000 description 5
- 108090000064 retinoic acid receptors Proteins 0.000 description 5
- 238000004007 reversed phase HPLC Methods 0.000 description 5
- 238000005292 vacuum distillation Methods 0.000 description 5
- 238000005160 1H NMR spectroscopy Methods 0.000 description 4
- JOZLGBMTXUXMMP-UHFFFAOYSA-N 2-[4-[2-(2,2,4,4-tetramethyl-8-propan-2-yloxy-3H-chromen-6-yl)ethynyl]phenyl]acetic acid Chemical compound C=1C(C(CC(C)(C)O2)(C)C)=C2C(OC(C)C)=CC=1C#CC1=CC=C(CC(O)=O)C=C1 JOZLGBMTXUXMMP-UHFFFAOYSA-N 0.000 description 4
- NRDVEUQIXCWYJE-UHFFFAOYSA-N 2-[4-[2-(4-ethenyl-8,8-dimethyl-5-oxo-6,7-dihydronaphthalen-2-yl)ethynyl]phenyl]acetic acid Chemical compound C1=C2C(C)(C)CCC(=O)C2=C(C=C)C=C1C#CC1=CC=C(CC(O)=O)C=C1 NRDVEUQIXCWYJE-UHFFFAOYSA-N 0.000 description 4
- VMSDXFKFZCLHJJ-UHFFFAOYSA-N 2-[4-[2-(8-hydroxy-2,2,4,4-tetramethyl-3H-chromen-6-yl)ethynyl]phenyl]-2-methylpropanoic acid Chemical compound C1=CC(C(C)(C(O)=O)C)=CC=C1C#CC1=CC(O)=C(OC(C)(C)CC2(C)C)C2=C1 VMSDXFKFZCLHJJ-UHFFFAOYSA-N 0.000 description 4
- MDPPEEDVCIGYNV-UHFFFAOYSA-N 2-[4-[2-[8-[[cyclopropyl(methyl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromen-6-yl]ethynyl]phenyl]propanoic acid Chemical compound C1=CC(C(C(O)=O)C)=CC=C1C#CC1=CC(CN(C)C2CC2)=C(OC(C)(C)CC2(C)C)C2=C1 MDPPEEDVCIGYNV-UHFFFAOYSA-N 0.000 description 4
- REPMJKNNNTXORZ-UHFFFAOYSA-N 3-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]phenyl]ethynyl]phenyl]but-2-enoic acid Chemical compound C1CC1N(C)CC(C=1)=CC=CC=1C#CC1=CC=C(C(C)=CC(O)=O)C=C1 REPMJKNNNTXORZ-UHFFFAOYSA-N 0.000 description 4
- JNRBAEZEKCIZHW-UHFFFAOYSA-N 6-ethynyl-2,2,4,4-tetramethyl-3H-chromen-8-ol Chemical compound C#CC1=CC(O)=C2OC(C)(C)CC(C)(C)C2=C1 JNRBAEZEKCIZHW-UHFFFAOYSA-N 0.000 description 4
- JFAORCRAYWRZSP-UHFFFAOYSA-N 8-[[cyclopropyl(propan-2-yl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromene-6-carboxylic acid Chemical compound C=1C(C(O)=O)=CC(C(CC(C)(C)O2)(C)C)=C2C=1CN(C(C)C)C1CC1 JFAORCRAYWRZSP-UHFFFAOYSA-N 0.000 description 4
- 206010001897 Alzheimer's disease Diseases 0.000 description 4
- 210000004369 Blood Anatomy 0.000 description 4
- 101710036491 CYP26A1 Proteins 0.000 description 4
- 102100007077 CYP26A1 Human genes 0.000 description 4
- 206010011732 Cyst Diseases 0.000 description 4
- ZCSHNCUQKCANBX-UHFFFAOYSA-N Lithium diisopropylamide Chemical compound [Li+].CC(C)[N-]C(C)C ZCSHNCUQKCANBX-UHFFFAOYSA-N 0.000 description 4
- OOUDEGCJDUFTFT-UHFFFAOYSA-N N-[(3-ethynylphenyl)methyl]-N-methylcyclopropanamine Chemical compound C1CC1N(C)CC1=CC=CC(C#C)=C1 OOUDEGCJDUFTFT-UHFFFAOYSA-N 0.000 description 4
- HEFFZLCRKUBYRC-UHFFFAOYSA-N N-[(6-ethynyl-2,2,4,4-tetramethyl-3H-chromen-8-yl)methyl]-N-methylcyclopropanamine Chemical compound C=1C(C#C)=CC(C(CC(C)(C)O2)(C)C)=C2C=1CN(C)C1CC1 HEFFZLCRKUBYRC-UHFFFAOYSA-N 0.000 description 4
- 206010054107 Nodule Diseases 0.000 description 4
- 208000008760 Optic Nerve Disease Diseases 0.000 description 4
- 241000700159 Rattus Species 0.000 description 4
- 239000004480 active ingredient Substances 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 239000008346 aqueous phase Substances 0.000 description 4
- 239000012298 atmosphere Substances 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- CPELXLSAUQHCOX-UHFFFAOYSA-M bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- NUCWDCRUEIUKPN-UHFFFAOYSA-J ethane-1,2-diamine;palladium(2+);phosphonato phosphate Chemical compound [Pd+2].[Pd+2].NCCN.NCCN.[O-]P([O-])(=O)OP([O-])([O-])=O NUCWDCRUEIUKPN-UHFFFAOYSA-J 0.000 description 4
- PJZIVTJXRUYXQX-UHFFFAOYSA-N ethyl 3-(4-iodophenyl)prop-2-enoate Chemical compound CCOC(=O)C=CC1=CC=C(I)C=C1 PJZIVTJXRUYXQX-UHFFFAOYSA-N 0.000 description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-N phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 4
- 239000011604 retinal Substances 0.000 description 4
- 239000000377 silicon dioxide Substances 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- BZKBCQXYZZXSCO-UHFFFAOYSA-N sodium hydride Inorganic materials [H-].[Na+] BZKBCQXYZZXSCO-UHFFFAOYSA-N 0.000 description 4
- 238000006467 substitution reaction Methods 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- GFZVDVREQICFPM-SDNWHVSQSA-N (E)-3-[4-[2-[3-[cyclopropyl(methyl)amino]phenyl]ethynyl]phenyl]prop-2-enoic acid Chemical compound C=1C=CC(C#CC=2C=CC(\C=C\C(O)=O)=CC=2)=CC=1N(C)C1CC1 GFZVDVREQICFPM-SDNWHVSQSA-N 0.000 description 3
- LBUJPTNKIBCYBY-UHFFFAOYSA-N 1,2,3,4-tetrahydroquinoline Chemical compound C1=CC=C2CCCNC2=C1 LBUJPTNKIBCYBY-UHFFFAOYSA-N 0.000 description 3
- YVACGDJNLJRXIZ-UHFFFAOYSA-N 2,2,4,4-tetramethyl-6-(2-trimethylsilylethynyl)-3H-chromene-8-carboxylic acid Chemical compound C[Si](C)(C)C#CC1=CC(C(O)=O)=C2OC(C)(C)CC(C)(C)C2=C1 YVACGDJNLJRXIZ-UHFFFAOYSA-N 0.000 description 3
- IJAMLSCTYIXRNM-UHFFFAOYSA-N 2,2,4,4-tetramethyl-8-propan-2-yl-3H-chromene-6-carboxylic acid Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(C(C)C)=CC(C(O)=O)=C2 IJAMLSCTYIXRNM-UHFFFAOYSA-N 0.000 description 3
- QXJWKKXKURVOMU-UHFFFAOYSA-N 2-(2-fluoro-4-iodophenyl)acetic acid Chemical compound OC(=O)CC1=CC=C(I)C=C1F QXJWKKXKURVOMU-UHFFFAOYSA-N 0.000 description 3
- PAHNYFLRMNPGLH-UHFFFAOYSA-N 2-[4-[2-(2,2-diethyl-4,4-dimethyl-8-propan-2-yl-3H-chromen-6-yl)ethynyl]phenyl]-2-methylpropanoic acid Chemical compound C=1C(C(C)C)=C2OC(CC)(CC)CC(C)(C)C2=CC=1C#CC1=CC=C(C(C)(C)C(O)=O)C=C1 PAHNYFLRMNPGLH-UHFFFAOYSA-N 0.000 description 3
- LLAFNYMEEQAISJ-UHFFFAOYSA-N 2-[4-[2-[8-[[cyclopropyl(methyl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromen-6-yl]ethynyl]phenyl]-2-methylpropanoic acid Chemical compound C1CC1N(C)CC(C1=C(C(CC(C)(C)O1)(C)C)C=1)=CC=1C#CC1=CC=C(C(C)(C)C(O)=O)C=C1 LLAFNYMEEQAISJ-UHFFFAOYSA-N 0.000 description 3
- FPQQSJJWHUJYPU-UHFFFAOYSA-N 3-(dimethylamino)propyliminomethylidene-ethylazanium;chloride Chemical compound Cl.CCN=C=NCCCN(C)C FPQQSJJWHUJYPU-UHFFFAOYSA-N 0.000 description 3
- XWNPVMBIPNGEHX-UHFFFAOYSA-N 3-[3-[2-(2,2,4,4-tetramethyl-8-propan-2-yl-3H-chromen-6-yl)ethynyl]phenyl]prop-2-enoic acid Chemical compound C=1C(C(CC(C)(C)O2)(C)C)=C2C(C(C)C)=CC=1C#CC1=CC=CC(C=CC(O)=O)=C1 XWNPVMBIPNGEHX-UHFFFAOYSA-N 0.000 description 3
- XSYGPDVKSKZQKC-UHFFFAOYSA-N 3-[4-(2,2,4,4-tetramethyl-8-propan-2-yl-3H-chromene-6-carbonyl)oxyphenyl]prop-2-enoic acid Chemical compound C=1C(C(CC(C)(C)O2)(C)C)=C2C(C(C)C)=CC=1C(=O)OC1=CC=C(C=CC(O)=O)C=C1 XSYGPDVKSKZQKC-UHFFFAOYSA-N 0.000 description 3
- JVIZCLMGFIFCPH-UHFFFAOYSA-N 3-[4-[2-(5-tert-butyl-2-methylphenyl)ethynyl]phenyl]prop-2-enoic acid Chemical compound CC1=CC=C(C(C)(C)C)C=C1C#CC1=CC=C(C=CC(O)=O)C=C1 JVIZCLMGFIFCPH-UHFFFAOYSA-N 0.000 description 3
- QZINVBMMQKSHKZ-UHFFFAOYSA-N 3-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]phenyl]ethynyl]phenyl]-2-methylprop-2-enoic acid Chemical compound C1CC1N(C)CC(C=1)=CC=CC=1C#CC1=CC=C(C=C(C)C(O)=O)C=C1 QZINVBMMQKSHKZ-UHFFFAOYSA-N 0.000 description 3
- UYRSLWPKZKASRB-UHFFFAOYSA-N 4-tert-butyl-2-hydroxybenzaldehyde Chemical compound CC(C)(C)C1=CC=C(C=O)C(O)=C1 UYRSLWPKZKASRB-UHFFFAOYSA-N 0.000 description 3
- YWTITWZHVYDTNO-UHFFFAOYSA-N 6-bromo-8-methoxy-4,4-dimethyl-2,3-dihydronaphthalen-1-one Chemical compound O=C1CCC(C)(C)C2=C1C(OC)=CC(Br)=C2 YWTITWZHVYDTNO-UHFFFAOYSA-N 0.000 description 3
- FYKUMCVXUUWHDN-UHFFFAOYSA-N 8-cyclopropyl-6-ethynyl-4,4-dimethylspiro[3H-chromene-2,1'-cyclopropane] Chemical compound O1C2=C(C3CC3)C=C(C#C)C=C2C(C)(C)CC21CC2 FYKUMCVXUUWHDN-UHFFFAOYSA-N 0.000 description 3
- RNTVAPJYIHGXEA-UHFFFAOYSA-N 8-hydroxy-4,4-dimethyl-6-(2-trimethylsilylethynyl)-2,3-dihydronaphthalen-1-one Chemical compound C1=C(C#C[Si](C)(C)C)C=C2C(C)(C)CCC(=O)C2=C1O RNTVAPJYIHGXEA-UHFFFAOYSA-N 0.000 description 3
- 206010001019 Acute promyelocytic leukaemia Diseases 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 208000007766 Kaposi Sarcoma Diseases 0.000 description 3
- 208000005749 Leukemia, Promyelocytic, Acute Diseases 0.000 description 3
- 210000004072 Lung Anatomy 0.000 description 3
- KFXDRVPERFGQNZ-UHFFFAOYSA-N N-[(6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-yl)methyl]-N-cyclopropylformamide Chemical compound C=12OC(C)(C)CC(C)(C)C2=CC(Br)=CC=1CN(C=O)C1CC1 KFXDRVPERFGQNZ-UHFFFAOYSA-N 0.000 description 3
- 229920001850 Nucleic acid sequence Polymers 0.000 description 3
- 210000004940 Nucleus Anatomy 0.000 description 3
- FVAUCKIRQBBSSJ-UHFFFAOYSA-M Sodium iodide Chemical compound [Na+].[I-] FVAUCKIRQBBSSJ-UHFFFAOYSA-M 0.000 description 3
- AVFUHBJCUUTGCD-UHFFFAOYSA-M [Br-].[Mg+]C Chemical compound [Br-].[Mg+]C AVFUHBJCUUTGCD-UHFFFAOYSA-M 0.000 description 3
- 230000027455 binding Effects 0.000 description 3
- OKTJSMMVPCPJKN-UHFFFAOYSA-N carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 3
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 3
- 238000004587 chromatography analysis Methods 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 239000012230 colorless oil Substances 0.000 description 3
- 239000006071 cream Substances 0.000 description 3
- 229940079593 drugs Drugs 0.000 description 3
- 230000002255 enzymatic Effects 0.000 description 3
- 238000005886 esterification reaction Methods 0.000 description 3
- BDAGIHXWWSANSR-UHFFFAOYSA-N formic acid Chemical compound OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 3
- 239000001257 hydrogen Substances 0.000 description 3
- 229910052739 hydrogen Inorganic materials 0.000 description 3
- UFHFLCQGNIYNRP-UHFFFAOYSA-N hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 3
- 230000003902 lesions Effects 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 150000007523 nucleic acids Chemical class 0.000 description 3
- 239000002773 nucleotide Substances 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 230000002035 prolonged Effects 0.000 description 3
- 230000002829 reduced Effects 0.000 description 3
- 235000020945 retinal Nutrition 0.000 description 3
- 239000012047 saturated solution Substances 0.000 description 3
- YOQDYZUWIQVZSF-UHFFFAOYSA-N sodium borohydride Chemical compound [BH4-].[Na+] YOQDYZUWIQVZSF-UHFFFAOYSA-N 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 239000007858 starting material Substances 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000002194 synthesizing Effects 0.000 description 3
- 125000005951 trifluoromethanesulfonyloxy group Chemical group 0.000 description 3
- SHGAZHPCJJPHSC-ZVCIMWCZSA-N (2E,4E,6Z,8E)-3,7-dimethyl-9-(2,6,6-trimethylcyclohex-1-en-1-yl)nona-2,4,6,8-tetraenoic acid Chemical compound OC(=O)/C=C(\C)/C=C/C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-ZVCIMWCZSA-N 0.000 description 2
- QNAXXQCFQHHYTO-UHFFFAOYSA-N (3-tert-butylphenyl) trifluoromethanesulfonate Chemical compound CC(C)(C)C1=CC=CC(OS(=O)(=O)C(F)(F)F)=C1 QNAXXQCFQHHYTO-UHFFFAOYSA-N 0.000 description 2
- CNQRHSZYVFYOIE-UHFFFAOYSA-N (4-iodophenyl)methanol Chemical compound OCC1=CC=C(I)C=C1 CNQRHSZYVFYOIE-UHFFFAOYSA-N 0.000 description 2
- AYDTUSBVGIWIOU-VOTSOKGWSA-N (E)-3-(4-iodophenyl)-2-methylprop-2-enoic acid Chemical compound OC(=O)C(/C)=C/C1=CC=C(I)C=C1 AYDTUSBVGIWIOU-VOTSOKGWSA-N 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N (E)-but-2-enedioate;hydron Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 2
- UARIISQGIYUZRB-UHFFFAOYSA-N 1-(6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-yl)ethanone Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(C(=O)C)=CC(Br)=C2 UARIISQGIYUZRB-UHFFFAOYSA-N 0.000 description 2
- IDQPSSOQUMFGJS-UHFFFAOYSA-N 1-(6-bromo-2,2-diethyl-4,4-dimethyl-3H-chromen-8-yl)ethanone Chemical compound BrC1=CC(C(C)=O)=C2OC(CC)(CC)CC(C)(C)C2=C1 IDQPSSOQUMFGJS-UHFFFAOYSA-N 0.000 description 2
- XYVXHLRVEKZBQT-UHFFFAOYSA-N 1-[5-[cyclopropyl(methyl)amino]-8,8-dimethyl-6,7-dihydro-5H-naphthalen-2-yl]-3-(4-iodophenyl)urea Chemical compound C1CC(C)(C)C2=CC(NC(=O)NC=3C=CC(I)=CC=3)=CC=C2C1N(C)C1CC1 XYVXHLRVEKZBQT-UHFFFAOYSA-N 0.000 description 2
- RVDYAXKZVHTFEZ-UHFFFAOYSA-N 2,2-diethyl-6-ethynyl-4,4-dimethyl-8-propan-2-yl-3H-chromene Chemical compound C#CC1=CC(C(C)C)=C2OC(CC)(CC)CC(C)(C)C2=C1 RVDYAXKZVHTFEZ-UHFFFAOYSA-N 0.000 description 2
- NXIFQNMMRNWCIY-UHFFFAOYSA-N 2-(2,2-diethyl-4,4-dimethyl-8-propan-2-yl-3H-chromen-6-yl)ethynyl-trimethylsilane Chemical compound C[Si](C)(C)C#CC1=CC(C(C)C)=C2OC(CC)(CC)CC(C)(C)C2=C1 NXIFQNMMRNWCIY-UHFFFAOYSA-N 0.000 description 2
- AKXADQUATOKZAG-UHFFFAOYSA-N 2-(3-tert-butylphenyl)ethynyl-trimethylsilane Chemical compound CC(C)(C)C1=CC=CC(C#C[Si](C)(C)C)=C1 AKXADQUATOKZAG-UHFFFAOYSA-N 0.000 description 2
- CHXHXUKWDCIJMW-UHFFFAOYSA-N 2-(5-tert-butyl-2-methylphenyl)ethynyl-trimethylsilane Chemical compound CC1=CC=C(C(C)(C)C)C=C1C#C[Si](C)(C)C CHXHXUKWDCIJMW-UHFFFAOYSA-N 0.000 description 2
- DGRZLQJGLDCFJH-UHFFFAOYSA-N 2-[2-fluoro-4-[2-(8-hydroxy-2,2,4,4-tetramethyl-3H-chromen-6-yl)ethynyl]phenyl]acetic acid Chemical compound C=1C(O)=C2OC(C)(C)CC(C)(C)C2=CC=1C#CC1=CC=C(CC(O)=O)C(F)=C1 DGRZLQJGLDCFJH-UHFFFAOYSA-N 0.000 description 2
- XTJZFWFPGYJWAS-UHFFFAOYSA-N 3-bromo-N-cyclopropyl-4-methylbenzamide Chemical compound C1=C(Br)C(C)=CC=C1C(=O)NC1CC1 XTJZFWFPGYJWAS-UHFFFAOYSA-N 0.000 description 2
- KDHPJBGNLIYNBE-UHFFFAOYSA-N 3-bromo-N-cyclopropyl-N,4-dimethylbenzamide Chemical compound C=1C=C(C)C(Br)=CC=1C(=O)N(C)C1CC1 KDHPJBGNLIYNBE-UHFFFAOYSA-N 0.000 description 2
- FGIDJHIWCTVCCK-UHFFFAOYSA-N 3-bromo-N-cyclopropyl-N-methylaniline Chemical compound C=1C=CC(Br)=CC=1N(C)C1CC1 FGIDJHIWCTVCCK-UHFFFAOYSA-N 0.000 description 2
- CYEKUDPFXBLGHH-UHFFFAOYSA-N 3-tert-Butylphenol Chemical compound CC(C)(C)C1=CC=CC(O)=C1 CYEKUDPFXBLGHH-UHFFFAOYSA-N 0.000 description 2
- GQWDSAYCWKRULC-UHFFFAOYSA-N 4,4-dimethyl-8-(2-trimethylsilylethoxymethoxy)-6-(2-trimethylsilylethynyl)-2,3-dihydronaphthalen-1-one Chemical compound C1=C(C#C[Si](C)(C)C)C=C2C(C)(C)CCC(=O)C2=C1OCOCC[Si](C)(C)C GQWDSAYCWKRULC-UHFFFAOYSA-N 0.000 description 2
- BXZQSWBVFJURAC-UHFFFAOYSA-N 4-tert-butyl-2-ethynyl-1-methylbenzene Chemical compound CC1=CC=C(C(C)(C)C)C=C1C#C BXZQSWBVFJURAC-UHFFFAOYSA-N 0.000 description 2
- KAOOIKAJVPJRMK-UHFFFAOYSA-N 5-[cyclopropyl(methyl)amino]-8,8-dimethyl-6,7-dihydro-5H-naphthalene-2-carboxylic acid;azide Chemical compound [N-]=[N+]=[N-].C1CC(C)(C)C2=CC(C(O)=O)=CC=C2C1N(C)C1CC1 KAOOIKAJVPJRMK-UHFFFAOYSA-N 0.000 description 2
- VTHADGVLPFVCHS-UHFFFAOYSA-N 6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-ol Chemical compound BrC1=CC(O)=C2OC(C)(C)CC(C)(C)C2=C1 VTHADGVLPFVCHS-UHFFFAOYSA-N 0.000 description 2
- DUPAHSBMMWEIAT-UHFFFAOYSA-N 6-bromo-2,2,4,4-tetramethyl-8-propan-2-yl-3H-chromene Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(C(C)C)=CC(Br)=C2 DUPAHSBMMWEIAT-UHFFFAOYSA-N 0.000 description 2
- ISBQPSVCARVCNV-UHFFFAOYSA-N 6-bromo-2,2-diethyl-4,4-dimethyl-3H-chromene Chemical compound BrC1=CC=C2OC(CC)(CC)CC(C)(C)C2=C1 ISBQPSVCARVCNV-UHFFFAOYSA-N 0.000 description 2
- KSFOPWPXJIKGBB-UHFFFAOYSA-N 6-bromo-2,2-diethyl-4,4-dimethyl-8-propan-2-yl-3H-chromene Chemical compound BrC1=CC(C(C)C)=C2OC(CC)(CC)CC(C)(C)C2=C1 KSFOPWPXJIKGBB-UHFFFAOYSA-N 0.000 description 2
- LCTKBYBCVVBKLG-UHFFFAOYSA-N 6-bromo-8-hydroxy-4,4-dimethyl-2,3-dihydronaphthalen-1-one Chemical compound C1=C(Br)C=C2C(C)(C)CCC(=O)C2=C1O LCTKBYBCVVBKLG-UHFFFAOYSA-N 0.000 description 2
- HYQFVEANTZWIKH-UHFFFAOYSA-N 6-ethynyl-2,2,4,4-tetramethyl-3H-chromene-8-carboxylic acid Chemical compound C#CC1=CC(C(O)=O)=C2OC(C)(C)CC(C)(C)C2=C1 HYQFVEANTZWIKH-UHFFFAOYSA-N 0.000 description 2
- FXMCIRDKRWSPBT-UHFFFAOYSA-N 6-ethynyl-2,2,4,4-tetramethyl-8-propan-2-yl-3H-chromene Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(C(C)C)=CC(C#C)=C2 FXMCIRDKRWSPBT-UHFFFAOYSA-N 0.000 description 2
- ZWPMPZQCSNFFBH-UHFFFAOYSA-N 8-cyclopropyl-4,4-dimethylspiro[3H-chromene-2,1'-cyclopropane]-6-amine Chemical compound O1C2=C(C3CC3)C=C(N)C=C2C(C)(C)CC21CC2 ZWPMPZQCSNFFBH-UHFFFAOYSA-N 0.000 description 2
- 229940022663 Acetate Drugs 0.000 description 2
- WETWJCDKMRHUPV-UHFFFAOYSA-N Acetyl chloride Chemical compound CC(Cl)=O WETWJCDKMRHUPV-UHFFFAOYSA-N 0.000 description 2
- 208000002874 Acne Vulgaris Diseases 0.000 description 2
- 206010000501 Acne conglobata Diseases 0.000 description 2
- 206010003246 Arthritis Diseases 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N Benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 239000005711 Benzoic acid Substances 0.000 description 2
- GZUXJHMPEANEGY-UHFFFAOYSA-N Bromomethane Chemical compound BrC GZUXJHMPEANEGY-UHFFFAOYSA-N 0.000 description 2
- 210000003169 Central Nervous System Anatomy 0.000 description 2
- 206010012601 Diabetes mellitus Diseases 0.000 description 2
- 206010012689 Diabetic retinopathy Diseases 0.000 description 2
- 208000005679 Eczema Diseases 0.000 description 2
- 229940088598 Enzyme Drugs 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 210000001508 Eye Anatomy 0.000 description 2
- 230000036499 Half live Effects 0.000 description 2
- 206010020649 Hyperkeratosis Diseases 0.000 description 2
- RAXXELZNTBOGNW-UHFFFAOYSA-N Imidazole Chemical compound C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 2
- 208000001126 Keratosis Diseases 0.000 description 2
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M Lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 2
- 229920002521 Macromolecule Polymers 0.000 description 2
- 208000002780 Macular Degeneration Diseases 0.000 description 2
- 208000001344 Macular Edema Diseases 0.000 description 2
- 206010025415 Macular oedema Diseases 0.000 description 2
- FEWJPZIEWOKRBE-XIXRPRMCSA-N Mesotartaric acid Chemical compound OC(=O)[C@@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-XIXRPRMCSA-N 0.000 description 2
- 108020004999 Messenger RNA Proteins 0.000 description 2
- 230000036740 Metabolism Effects 0.000 description 2
- 210000000214 Mouth Anatomy 0.000 description 2
- 210000004400 Mucous Membrane Anatomy 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 2
- NENZXMJTTFCYIZ-UHFFFAOYSA-N N-(3-bromophenyl)-N-methylformamide Chemical compound O=CN(C)C1=CC=CC(Br)=C1 NENZXMJTTFCYIZ-UHFFFAOYSA-N 0.000 description 2
- LFTLOKWAGJYHHR-UHFFFAOYSA-N N-Methylmorpholine N-oxide Chemical compound CN1(=O)CCOCC1 LFTLOKWAGJYHHR-UHFFFAOYSA-N 0.000 description 2
- PQHVWEORJMVMNA-UHFFFAOYSA-N N-[(3-bromo-4-methylphenyl)methyl]-N-methylcyclopropanamine Chemical compound C1CC1N(C)CC1=CC=C(C)C(Br)=C1 PQHVWEORJMVMNA-UHFFFAOYSA-N 0.000 description 2
- PGCANNLAVOGIMS-UHFFFAOYSA-N N-[(3-ethynyl-4-methylphenyl)methyl]-N-methylcyclopropanamine Chemical compound C1CC1N(C)CC1=CC=C(C)C(C#C)=C1 PGCANNLAVOGIMS-UHFFFAOYSA-N 0.000 description 2
- XAQFPIGRQDSIAZ-UHFFFAOYSA-N N-[(3-iodophenyl)methyl]-N-methylcyclopropanamine Chemical compound C1CC1N(C)CC1=CC=CC(I)=C1 XAQFPIGRQDSIAZ-UHFFFAOYSA-N 0.000 description 2
- RNRNQFLQZDDEHK-UHFFFAOYSA-N N-[(3-iodophenyl)methyl]cyclopropanamine Chemical compound IC1=CC=CC(CNC2CC2)=C1 RNRNQFLQZDDEHK-UHFFFAOYSA-N 0.000 description 2
- DFFDHCCKLQIKPN-UHFFFAOYSA-N N-[(6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-yl)methyl]cyclopropanamine Chemical compound C=12OC(C)(C)CC(C)(C)C2=CC(Br)=CC=1CNC1CC1 DFFDHCCKLQIKPN-UHFFFAOYSA-N 0.000 description 2
- HFPQUMNSNFVKSD-UHFFFAOYSA-N N-cyclopropyl-3-ethynyl-N-methylaniline Chemical compound C=1C=CC(C#C)=CC=1N(C)C1CC1 HFPQUMNSNFVKSD-UHFFFAOYSA-N 0.000 description 2
- LLYXMWFYXBWSJV-UHFFFAOYSA-N N-cyclopropyl-N-methyl-3-(2-trimethylsilylethynyl)aniline Chemical compound C=1C=CC(C#C[Si](C)(C)C)=CC=1N(C)C1CC1 LLYXMWFYXBWSJV-UHFFFAOYSA-N 0.000 description 2
- PJZDYMKKYJNZKY-UHFFFAOYSA-N N-methyl-N-[[2,2,4,4-tetramethyl-6-(2-trimethylsilylethynyl)-3H-chromen-8-yl]methyl]cyclopropanamine Chemical compound C=1C(C#C[Si](C)(C)C)=CC(C(CC(C)(C)O2)(C)C)=C2C=1CN(C)C1CC1 PJZDYMKKYJNZKY-UHFFFAOYSA-N 0.000 description 2
- LFCVDWMFZBWGMP-UHFFFAOYSA-N N-methyl-N-[[3-(2-trimethylsilylethynyl)phenyl]methyl]cyclopropanamine Chemical compound C1CC1N(C)CC1=CC=CC(C#C[Si](C)(C)C)=C1 LFCVDWMFZBWGMP-UHFFFAOYSA-N 0.000 description 2
- JZKMFLGHPAMWRH-UHFFFAOYSA-N N-methyl-N-[[4-methyl-3-(2-trimethylsilylethynyl)phenyl]methyl]cyclopropanamine Chemical compound C1CC1N(C)CC1=CC=C(C)C(C#C[Si](C)(C)C)=C1 JZKMFLGHPAMWRH-UHFFFAOYSA-N 0.000 description 2
- KPSSIOMAKSHJJG-UHFFFAOYSA-N Neopentyl alcohol Chemical compound CC(C)(C)CO KPSSIOMAKSHJJG-UHFFFAOYSA-N 0.000 description 2
- CTSLXHKWHWQRSH-UHFFFAOYSA-N Oxalyl chloride Chemical compound ClC(=O)C(Cl)=O CTSLXHKWHWQRSH-UHFFFAOYSA-N 0.000 description 2
- JOXIMZWYDAKGHI-UHFFFAOYSA-N P-Toluenesulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 2
- 206010033733 Papule Diseases 0.000 description 2
- 210000002826 Placenta Anatomy 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 208000002158 Proliferative Vitreoretinopathy Diseases 0.000 description 2
- 210000004915 Pus Anatomy 0.000 description 2
- 206010038848 Retinal detachment Diseases 0.000 description 2
- 206010038932 Retinopathy Diseases 0.000 description 2
- 206010038923 Retinopathy Diseases 0.000 description 2
- 206010038934 Retinopathy proliferative Diseases 0.000 description 2
- MFRIHAYPQRLWNB-UHFFFAOYSA-N Sodium tert-butoxide Chemical compound [Na+].CC(C)(C)[O-] MFRIHAYPQRLWNB-UHFFFAOYSA-N 0.000 description 2
- AKHNMLFCWUSKQB-UHFFFAOYSA-L Sodium thiosulphate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=S AKHNMLFCWUSKQB-UHFFFAOYSA-L 0.000 description 2
- 210000000952 Spleen Anatomy 0.000 description 2
- 210000002784 Stomach Anatomy 0.000 description 2
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Chemical compound OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 2
- 241000282898 Sus scrofa Species 0.000 description 2
- FYSNRJHAOHDILO-UHFFFAOYSA-N Thionyl chloride Chemical compound ClS(Cl)=O FYSNRJHAOHDILO-UHFFFAOYSA-N 0.000 description 2
- VXUYXOFXAQZZMF-UHFFFAOYSA-N Titanium isopropoxide Chemical compound CC(C)O[Ti](OC(C)C)(OC(C)C)OC(C)C VXUYXOFXAQZZMF-UHFFFAOYSA-N 0.000 description 2
- XJDNKRIXUMDJCW-UHFFFAOYSA-J Titanium tetrachloride Chemical compound Cl[Ti](Cl)(Cl)Cl XJDNKRIXUMDJCW-UHFFFAOYSA-J 0.000 description 2
- GGUBFICZYGKNTD-UHFFFAOYSA-N Triethyl phosphonoacetate Chemical compound CCOC(=O)CP(=O)(OCC)OCC GGUBFICZYGKNTD-UHFFFAOYSA-N 0.000 description 2
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N Triphenylphosphine Chemical compound C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 2
- 239000007983 Tris buffer Substances 0.000 description 2
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 2
- OGQICQVSFDPSEI-UHFFFAOYSA-N Zorac Chemical compound N1=CC(C(=O)OCC)=CC=C1C#CC1=CC=C(SCCC2(C)C)C2=C1 OGQICQVSFDPSEI-UHFFFAOYSA-N 0.000 description 2
- 230000001594 aberrant Effects 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-M acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 2
- 239000012346 acetyl chloride Substances 0.000 description 2
- 238000007792 addition Methods 0.000 description 2
- 229960001445 alitretinoin Drugs 0.000 description 2
- 125000005907 alkyl ester group Chemical group 0.000 description 2
- 229930002945 all-trans-retinaldehyde Natural products 0.000 description 2
- 125000003275 alpha amino acid group Chemical group 0.000 description 2
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 2
- 150000001412 amines Chemical class 0.000 description 2
- 150000003863 ammonium salts Chemical class 0.000 description 2
- 230000002547 anomalous Effects 0.000 description 2
- 230000000111 anti-oxidant Effects 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 230000000903 blocking Effects 0.000 description 2
- 229910000090 borane Inorganic materials 0.000 description 2
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N borate Chemical compound [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- UGFAIRIUMAVXCW-UHFFFAOYSA-N carbon monoxide Chemical compound [O+]#[C-] UGFAIRIUMAVXCW-UHFFFAOYSA-N 0.000 description 2
- 229910002091 carbon monoxide Inorganic materials 0.000 description 2
- 230000003197 catalytic Effects 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 238000001816 cooling Methods 0.000 description 2
- 230000000875 corresponding Effects 0.000 description 2
- XDTMQSROBMDMFD-UHFFFAOYSA-N cyclohexane Chemical compound C1CCCCC1 XDTMQSROBMDMFD-UHFFFAOYSA-N 0.000 description 2
- 230000003111 delayed Effects 0.000 description 2
- 201000004624 dermatitis Diseases 0.000 description 2
- HKYGSMOFSFOEIP-UHFFFAOYSA-N dichloro(dichloromethoxy)methane Chemical compound ClC(Cl)OC(Cl)Cl HKYGSMOFSFOEIP-UHFFFAOYSA-N 0.000 description 2
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 2
- 239000006196 drop Substances 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 231100001003 eczema Toxicity 0.000 description 2
- 229940009662 edetate Drugs 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N edta Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 230000002708 enhancing Effects 0.000 description 2
- ZPCFOKAQPFCNRR-UHFFFAOYSA-N ethyl 2,2,4,4-tetramethyl-3H-chromene-6-carboxylate Chemical compound O1C(C)(C)CC(C)(C)C2=CC(C(=O)OCC)=CC=C21 ZPCFOKAQPFCNRR-UHFFFAOYSA-N 0.000 description 2
- MNPPUDJVKHVLKX-UHFFFAOYSA-N ethyl 4-(4-bromo-2-methoxyphenyl)-4-oxobutanoate Chemical compound CCOC(=O)CCC(=O)C1=CC=C(Br)C=C1OC MNPPUDJVKHVLKX-UHFFFAOYSA-N 0.000 description 2
- 238000005755 formation reaction Methods 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N fumaric acid Chemical compound OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 239000012362 glacial acetic acid Substances 0.000 description 2
- UKJFVOWPUXSBOM-UHFFFAOYSA-N hexane;oxolane Chemical compound C1CCOC1.CCCCCC UKJFVOWPUXSBOM-UHFFFAOYSA-N 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- 238000010255 intramuscular injection Methods 0.000 description 2
- 239000007927 intramuscular injection Substances 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 230000002427 irreversible Effects 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- GLXDVVHUTZTUQK-UHFFFAOYSA-M lithium;hydroxide;hydrate Chemical compound [Li+].O.[OH-] GLXDVVHUTZTUQK-UHFFFAOYSA-M 0.000 description 2
- 201000010230 macular retinal edema Diseases 0.000 description 2
- 229920002106 messenger RNA Polymers 0.000 description 2
- 230000004060 metabolic process Effects 0.000 description 2
- 230000035786 metabolism Effects 0.000 description 2
- DZYKILFYVRVLFU-UHFFFAOYSA-N methyl 2-(2-fluoro-4-iodophenyl)acetate Chemical compound COC(=O)CC1=CC=C(I)C=C1F DZYKILFYVRVLFU-UHFFFAOYSA-N 0.000 description 2
- MSEMDBDKZOBLAK-UHFFFAOYSA-N methyl 2-[4-[2-(2,2-diethyl-4,4-dimethyl-8-propan-2-yl-3H-chromen-6-yl)ethynyl]phenyl]-2-methylpropanoate Chemical compound C=1C(C(C)C)=C2OC(CC)(CC)CC(C)(C)C2=CC=1C#CC1=CC=C(C(C)(C)C(=O)OC)C=C1 MSEMDBDKZOBLAK-UHFFFAOYSA-N 0.000 description 2
- GHJDBMAZFIBARP-UHFFFAOYSA-N methyl 2-[4-[2-(4-ethenyl-8,8-dimethyl-5-oxo-6,7-dihydronaphthalen-2-yl)ethynyl]phenyl]acetate Chemical compound C1=CC(CC(=O)OC)=CC=C1C#CC1=CC(C=C)=C(C(=O)CCC2(C)C)C2=C1 GHJDBMAZFIBARP-UHFFFAOYSA-N 0.000 description 2
- YNQBARNBABOAMJ-UHFFFAOYSA-N methyl 2-[4-[2-(8-hydroxy-2,2,4,4-tetramethyl-3H-chromen-6-yl)ethynyl]phenyl]acetate Chemical compound C1=CC(CC(=O)OC)=CC=C1C#CC1=CC(O)=C(OC(C)(C)CC2(C)C)C2=C1 YNQBARNBABOAMJ-UHFFFAOYSA-N 0.000 description 2
- MQPWYJVSDUHUMM-UHFFFAOYSA-N methyl 2-[4-[2-(8-hydroxy-2,2,4,4-tetramethyl-3H-chromen-6-yl)ethynyl]phenyl]propanoate Chemical compound C1=CC(C(C)C(=O)OC)=CC=C1C#CC1=CC(O)=C(OC(C)(C)CC2(C)C)C2=C1 MQPWYJVSDUHUMM-UHFFFAOYSA-N 0.000 description 2
- IDATZIWFZZIKNZ-UHFFFAOYSA-N methyl 3-(4-iodophenyl)prop-2-enoate Chemical compound COC(=O)C=CC1=CC=C(I)C=C1 IDATZIWFZZIKNZ-UHFFFAOYSA-N 0.000 description 2
- LKRXDBCPBCYYTE-UHFFFAOYSA-N methyl 5-[cyclopropyl(methyl)amino]-8,8-dimethyl-6,7-dihydro-5H-naphthalene-2-carboxylate Chemical compound C1CC(C)(C)C2=CC(C(=O)OC)=CC=C2C1N(C)C1CC1 LKRXDBCPBCYYTE-UHFFFAOYSA-N 0.000 description 2
- 150000007522 mineralic acids Chemical class 0.000 description 2
- 230000000051 modifying Effects 0.000 description 2
- 239000002808 molecular sieve Substances 0.000 description 2
- SECXISVLQFMRJM-UHFFFAOYSA-N n-methylpyrrolidone Chemical compound CN1CCCC1=O SECXISVLQFMRJM-UHFFFAOYSA-N 0.000 description 2
- 230000001613 neoplastic Effects 0.000 description 2
- 230000001537 neural Effects 0.000 description 2
- 230000000926 neurological Effects 0.000 description 2
- PVNIIMVLHYAWGP-UHFFFAOYSA-N nicotinic acid Chemical compound OC(=O)C1=CC=CN=C1 PVNIIMVLHYAWGP-UHFFFAOYSA-N 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 230000001936 parietal Effects 0.000 description 2
- 239000000816 peptidomimetic Substances 0.000 description 2
- 229920001184 polypeptide Polymers 0.000 description 2
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000002335 preservative Effects 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- GRTZEZZMAXRYDM-UHFFFAOYSA-N propan-2-yl 6-[2-[4-(2-methoxy-2-oxoethyl)phenyl]ethynyl]-2,2,4,4-tetramethyl-3H-chromene-8-carboxylate Chemical compound C1=CC(CC(=O)OC)=CC=C1C#CC1=CC(C(=O)OC(C)C)=C(OC(C)(C)CC2(C)C)C2=C1 GRTZEZZMAXRYDM-UHFFFAOYSA-N 0.000 description 2
- 239000000376 reactant Substances 0.000 description 2
- 238000010992 reflux Methods 0.000 description 2
- 230000002207 retinal Effects 0.000 description 2
- 230000004264 retinal detachment Effects 0.000 description 2
- 230000002441 reversible Effects 0.000 description 2
- 238000007127 saponification reaction Methods 0.000 description 2
- 210000004378 sebocyte Anatomy 0.000 description 2
- 231100000486 side effect Toxicity 0.000 description 2
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 2
- 229910000033 sodium borohydride Inorganic materials 0.000 description 2
- 235000019345 sodium thiosulphate Nutrition 0.000 description 2
- 239000012453 solvate Substances 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- DKGAVHZHDRPRBM-UHFFFAOYSA-N t-BuOH Chemical compound CC(C)(C)O DKGAVHZHDRPRBM-UHFFFAOYSA-N 0.000 description 2
- 229960000565 tazarotene Drugs 0.000 description 2
- WGLPBDUCMAPZCE-UHFFFAOYSA-N trioxochromium Chemical compound O=[Cr](=O)=O WGLPBDUCMAPZCE-UHFFFAOYSA-N 0.000 description 2
- 230000002792 vascular Effects 0.000 description 2
- IBRARMBNPDUARK-UHFFFAOYSA-N (5-tert-butyl-2-formylphenyl) trifluoromethanesulfonate Chemical compound CC(C)(C)C1=CC=C(C=O)C(OS(=O)(=O)C(F)(F)F)=C1 IBRARMBNPDUARK-UHFFFAOYSA-N 0.000 description 1
- CGEXLKQYLYYDDU-UHFFFAOYSA-N (6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-yl) acetate Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(OC(=O)C)=CC(Br)=C2 CGEXLKQYLYYDDU-UHFFFAOYSA-N 0.000 description 1
- QKKRFXIZQYZKDM-GXDHUFHOSA-N (E)-3-[4-[2-[5-[[cyclopropyl(methyl)amino]methyl]-2-methylphenyl]ethynyl]phenyl]prop-2-enoic acid Chemical compound C1CC1N(C)CC(C=1)=CC=C(C)C=1C#CC1=CC=C(\C=C\C(O)=O)C=C1 QKKRFXIZQYZKDM-GXDHUFHOSA-N 0.000 description 1
- DXLQEJHUQKKSRB-UHFFFAOYSA-N 1,1,1-trifluoro-N-pyridin-2-yl-N-(trifluoromethylsulfonyl)methanesulfonamide Chemical compound FC(F)(F)S(=O)(=O)N(S(=O)(=O)C(F)(F)F)C1=CC=CC=N1 DXLQEJHUQKKSRB-UHFFFAOYSA-N 0.000 description 1
- JZJWCDQGIPQBAO-UHFFFAOYSA-N 1-(4-iodophenyl)ethanone Chemical compound CC(=O)C1=CC=C(I)C=C1 JZJWCDQGIPQBAO-UHFFFAOYSA-N 0.000 description 1
- BACZSVQZBSCWIG-UHFFFAOYSA-N 1-(bromomethyl)-3-iodobenzene Chemical compound BrCC1=CC=CC(I)=C1 BACZSVQZBSCWIG-UHFFFAOYSA-N 0.000 description 1
- PLDWAJLZAAHOGG-UHFFFAOYSA-N 1-bromo-3-methoxybenzene Chemical compound COC1=CC=CC(Br)=C1 PLDWAJLZAAHOGG-UHFFFAOYSA-N 0.000 description 1
- UPHOPMSGKZNELG-UHFFFAOYSA-M 1-carboxynaphthalen-2-olate Chemical compound C1=CC=CC2=C(C([O-])=O)C(O)=CC=C21 UPHOPMSGKZNELG-UHFFFAOYSA-M 0.000 description 1
- ZIYVJICPWLXLIQ-UHFFFAOYSA-N 1-tert-butyl-3-ethynylbenzene Chemical compound CC(C)(C)C1=CC=CC(C#C)=C1 ZIYVJICPWLXLIQ-UHFFFAOYSA-N 0.000 description 1
- OZGSEIVTQLXWRO-UHFFFAOYSA-N 2,4,6-trichlorobenzoyl chloride Chemical compound ClC(=O)C1=C(Cl)C=C(Cl)C=C1Cl OZGSEIVTQLXWRO-UHFFFAOYSA-N 0.000 description 1
- FJSHTWVDFAUNCO-UHFFFAOYSA-N 2-(4-iodophenyl)acetic acid Chemical compound OC(=O)CC1=CC=C(I)C=C1 FJSHTWVDFAUNCO-UHFFFAOYSA-N 0.000 description 1
- RMPXUPUAZGKDBN-UHFFFAOYSA-N 2-(4-iodophenyl)propanoic acid Chemical compound OC(=O)C(C)C1=CC=C(I)C=C1 RMPXUPUAZGKDBN-UHFFFAOYSA-N 0.000 description 1
- AXSOJPZXFCYFMM-UHFFFAOYSA-N 2-(6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-yl)ethynyl-trimethylsilane Chemical compound BrC1=CC(C#C[Si](C)(C)C)=C2OC(C)(C)CC(C)(C)C2=C1 AXSOJPZXFCYFMM-UHFFFAOYSA-N 0.000 description 1
- BPXKZEMBEZGUAH-UHFFFAOYSA-N 2-(chloromethoxy)ethyl-trimethylsilane Chemical compound C[Si](C)(C)CCOCCl BPXKZEMBEZGUAH-UHFFFAOYSA-N 0.000 description 1
- BKOOMYPCSUNDGP-UHFFFAOYSA-N 2-Methyl-2-butene Chemical compound CC=C(C)C BKOOMYPCSUNDGP-UHFFFAOYSA-N 0.000 description 1
- OABAMFBEZKBFBO-UHFFFAOYSA-N 2-[2-fluoro-4-[2-(3-propan-2-yloxycarbonylphenyl)ethynyl]phenyl]acetic acid Chemical compound CC(C)OC(=O)C1=CC=CC(C#CC=2C=C(F)C(CC(O)=O)=CC=2)=C1 OABAMFBEZKBFBO-UHFFFAOYSA-N 0.000 description 1
- LYDNPINFUVYCMA-UHFFFAOYSA-N 2-[3-[2-[3-fluoro-4-(2-methoxy-2-oxoethyl)phenyl]ethynyl]phenyl]-2-methylpropanoic acid Chemical compound C1=C(F)C(CC(=O)OC)=CC=C1C#CC1=CC=CC(C(C)(C)C(O)=O)=C1 LYDNPINFUVYCMA-UHFFFAOYSA-N 0.000 description 1
- ZZVUUPXGJDSLAB-UHFFFAOYSA-N 2-[3-[2-[4-(2-methoxy-2-oxoethyl)phenyl]ethynyl]phenyl]-2-methylpropanoic acid Chemical compound C1=CC(CC(=O)OC)=CC=C1C#CC1=CC=CC(C(C)(C)C(O)=O)=C1 ZZVUUPXGJDSLAB-UHFFFAOYSA-N 0.000 description 1
- GPGJLMRWFDWQOT-UHFFFAOYSA-N 2-[4-[2-(3-ethynyl-5-propan-2-yloxycarbonylphenyl)ethynyl]-2-fluorophenyl]acetic acid Chemical compound CC(C)OC(=O)C1=CC(C#C)=CC(C#CC=2C=C(F)C(CC(O)=O)=CC=2)=C1 GPGJLMRWFDWQOT-UHFFFAOYSA-N 0.000 description 1
- NAKYIGABCRAEQY-UHFFFAOYSA-N 2-[4-[2-(5-tert-butyl-2-methyl-4-propan-2-yloxy-3-prop-2-ynylphenyl)ethynyl]phenyl]acetic acid Chemical compound C1=C(C(C)(C)C)C(OC(C)C)=C(CC#C)C(C)=C1C#CC1=CC=C(CC(O)=O)C=C1 NAKYIGABCRAEQY-UHFFFAOYSA-N 0.000 description 1
- CAEATINCYOTIER-UHFFFAOYSA-N 2-[4-[2-(5-tert-butyl-3-ethenyl-2-methyl-4-propan-2-yloxyphenyl)ethynyl]phenyl]acetic acid Chemical compound C1=C(C(C)(C)C)C(OC(C)C)=C(C=C)C(C)=C1C#CC1=CC=C(CC(O)=O)C=C1 CAEATINCYOTIER-UHFFFAOYSA-N 0.000 description 1
- NOHSMPCYSWGMRX-UHFFFAOYSA-N 2-[4-[2-(5-tert-butyl-3-ethynyl-2-methyl-4-propan-2-yloxyphenyl)ethynyl]phenyl]acetic acid Chemical compound CC1=C(C#C)C(OC(C)C)=C(C(C)(C)C)C=C1C#CC1=CC=C(CC(O)=O)C=C1 NOHSMPCYSWGMRX-UHFFFAOYSA-N 0.000 description 1
- ONTOMOJZTPGSMP-UHFFFAOYSA-N 2-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]-4-propan-2-yloxycarbonylphenyl]ethynyl]-2-fluorophenyl]acetic acid Chemical compound CC(C)OC(=O)C1=CC=C(C#CC=2C=C(F)C(CC(O)=O)=CC=2)C=C1CN(C)C1CC1 ONTOMOJZTPGSMP-UHFFFAOYSA-N 0.000 description 1
- QCMDEWDMHVYWMT-UHFFFAOYSA-N 2-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]-4-propan-2-yloxycarbonylphenyl]ethynyl]phenyl]acetic acid Chemical compound CC(C)OC(=O)C1=CC=C(C#CC=2C=CC(CC(O)=O)=CC=2)C=C1CN(C)C1CC1 QCMDEWDMHVYWMT-UHFFFAOYSA-N 0.000 description 1
- JYUOXNIBDIHICP-UHFFFAOYSA-N 2-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]-4-propan-2-yloxycarbonylphenyl]ethynyl]phenyl]propanoic acid Chemical compound CC(C)OC(=O)C1=CC=C(C#CC=2C=CC(=CC=2)C(C)C(O)=O)C=C1CN(C)C1CC1 JYUOXNIBDIHICP-UHFFFAOYSA-N 0.000 description 1
- PWFUXIVZUHAKLO-UHFFFAOYSA-N 2-[4-[2-[4-cyclopropyl-5-[cyclopropyl(methyl)amino]-8,8-dimethyl-6,7-dihydro-5H-naphthalen-2-yl]ethynyl]phenyl]propanoic acid Chemical compound C1=CC(C(C(O)=O)C)=CC=C1C#CC1=CC(C2CC2)=C(C(CCC2(C)C)N(C)C3CC3)C2=C1 PWFUXIVZUHAKLO-UHFFFAOYSA-N 0.000 description 1
- YQWYCWCBNXSUBH-UHFFFAOYSA-N 2-[4-[2-[5-[cyclopropyl(methyl)amino]-4-methoxy-8,8-dimethyl-6,7-dihydro-5H-naphthalen-2-yl]ethynyl]phenyl]propanoic acid Chemical compound COC1=CC(C#CC=2C=CC(=CC=2)C(C)C(O)=O)=CC(C(CC2)(C)C)=C1C2N(C)C1CC1 YQWYCWCBNXSUBH-UHFFFAOYSA-N 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- XFKCVVFQGHCLIP-UHFFFAOYSA-N 2-ethylbutanedioyl dichloride Chemical compound CCC(C(Cl)=O)CC(Cl)=O XFKCVVFQGHCLIP-UHFFFAOYSA-N 0.000 description 1
- DHXBCVZQFYNLRC-UHFFFAOYSA-N 2-iodo-3-phenylprop-2-enoic acid Chemical compound OC(=O)C(I)=CC1=CC=CC=C1 DHXBCVZQFYNLRC-UHFFFAOYSA-N 0.000 description 1
- FMKOJHQHASLBPH-OUBTZVSYSA-N 2-iodopropane Chemical compound CC([13CH3])I FMKOJHQHASLBPH-OUBTZVSYSA-N 0.000 description 1
- XUYJLQHKOGNDPB-UHFFFAOYSA-M 2-phosphonoacetate Chemical compound OP(O)(=O)CC([O-])=O XUYJLQHKOGNDPB-UHFFFAOYSA-M 0.000 description 1
- ZNGINKJHQQQORD-UHFFFAOYSA-N 2-trimethylsilylethanol Chemical compound C[Si](C)(C)CCO ZNGINKJHQQQORD-UHFFFAOYSA-N 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K 2qpq Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- QGCCNWSXJHGUNL-UHFFFAOYSA-N 3-Iodo-Benzyl Alcohol Chemical compound OCC1=CC=CC(I)=C1 QGCCNWSXJHGUNL-UHFFFAOYSA-N 0.000 description 1
- IUXJDSVNLUIRKZ-UHFFFAOYSA-N 3-[4-[2-[4-cyclopropyl-5-[cyclopropyl(methyl)amino]-8,8-dimethyl-6,7-dihydro-5H-naphthalen-2-yl]ethynyl]phenyl]prop-2-enoic acid Chemical compound C1CC(C)(C)C2=CC(C#CC=3C=CC(C=CC(O)=O)=CC=3)=CC(C3CC3)=C2C1N(C)C1CC1 IUXJDSVNLUIRKZ-UHFFFAOYSA-N 0.000 description 1
- GHOKWGTUZJEAQD-ZETCQYMHSA-M 3-[[(2R)-2,4-dihydroxy-3,3-dimethylbutanoyl]amino]propanoate Chemical compound OCC(C)(C)[C@@H](O)C(=O)NCCC([O-])=O GHOKWGTUZJEAQD-ZETCQYMHSA-M 0.000 description 1
- ZFJOMUKPDWNRFI-UHFFFAOYSA-N 3-bromo-4-methylbenzoic acid Chemical compound CC1=CC=C(C(O)=O)C=C1Br ZFJOMUKPDWNRFI-UHFFFAOYSA-N 0.000 description 1
- HKOSFZXROYRVJT-UHFFFAOYSA-N 3-bromo-N-methylaniline Chemical compound CNC1=CC=CC(Br)=C1 HKOSFZXROYRVJT-UHFFFAOYSA-N 0.000 description 1
- DHXNZYCXMFBMHE-UHFFFAOYSA-M 3-bromopropanoate Chemical compound [O-]C(=O)CCBr DHXNZYCXMFBMHE-UHFFFAOYSA-M 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-M 3-carboxy-2,3-dihydroxypropanoate Chemical compound OC(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-M 0.000 description 1
- FQAWBGAIOYWONH-UHFFFAOYSA-N 3-chloroperoxybenzoic acid Chemical compound OC(=O)C1=CC=CC(OOCl)=C1 FQAWBGAIOYWONH-UHFFFAOYSA-N 0.000 description 1
- RZODAQZAFOBFLS-UHFFFAOYSA-N 3-iodobenzaldehyde Chemical compound IC1=CC=CC(C=O)=C1 RZODAQZAFOBFLS-UHFFFAOYSA-N 0.000 description 1
- QXUUNBGWWCYGAG-UHFFFAOYSA-N 4-[(8-ethynyl-2,2,4,4-tetramethyl-3H-chromen-6-yl)carbamoylamino]-2-fluorobenzoic acid Chemical compound C=1C(C#C)=C2OC(C)(C)CC(C)(C)C2=CC=1NC(=O)NC1=CC=C(C(O)=O)C(F)=C1 QXUUNBGWWCYGAG-UHFFFAOYSA-N 0.000 description 1
- VLVCDUSVTXIWGW-UHFFFAOYSA-N 4-iodoaniline Chemical compound NC1=CC=C(I)C=C1 VLVCDUSVTXIWGW-UHFFFAOYSA-N 0.000 description 1
- 125000006306 4-iodophenyl group Chemical group [H]C1=C([H])C(*)=C([H])C([H])=C1I 0.000 description 1
- TWWYVJYKSMGRBF-UHFFFAOYSA-N 5-[cyclopropyl(methyl)amino]-8,8-dimethyl-6,7-dihydro-5H-naphthalene-2-carboxylic acid Chemical compound C1CC(C)(C)C2=CC(C(O)=O)=CC=C2C1N(C)C1CC1 TWWYVJYKSMGRBF-UHFFFAOYSA-N 0.000 description 1
- VNWBZADADUVOIU-UHFFFAOYSA-N 5-tert-butyl-2-(hydroxymethyl)phenol Chemical compound CC(C)(C)C1=CC=C(CO)C(O)=C1 VNWBZADADUVOIU-UHFFFAOYSA-N 0.000 description 1
- FLSBACIXSFRPLT-UHFFFAOYSA-N 5-tert-butyl-2-methylphenol Chemical compound CC1=CC=C(C(C)(C)C)C=C1O FLSBACIXSFRPLT-UHFFFAOYSA-N 0.000 description 1
- SRHRYVIAUTXLIT-UHFFFAOYSA-N 6-bromo-2,2,4,4-tetramethyl-3,5-dihydrochromene-6-carbaldehyde Chemical compound C1=CC(Br)(C=O)CC2=C1OC(C)(C)CC2(C)C SRHRYVIAUTXLIT-UHFFFAOYSA-N 0.000 description 1
- SZKQWSXLUGDACW-UHFFFAOYSA-N 6-bromo-2,2,4,4-tetramethyl-3H-chromene-8-carbaldehyde Chemical compound BrC1=CC(C=O)=C2OC(C)(C)CC(C)(C)C2=C1 SZKQWSXLUGDACW-UHFFFAOYSA-N 0.000 description 1
- ZMEGAKQYQYQIHK-UHFFFAOYSA-N 6-bromo-4,4-dimethyl-3H-chromen-2-one Chemical compound C1=C(Br)C=C2C(C)(C)CC(=O)OC2=C1 ZMEGAKQYQYQIHK-UHFFFAOYSA-N 0.000 description 1
- WMFWCNYKBAYFGA-UHFFFAOYSA-N 6-bromo-8-cyclopropyl-4,4-dimethylspiro[3H-chromene-2,1'-cyclopropane] Chemical compound O1C2=C(C3CC3)C=C(Br)C=C2C(C)(C)CC21CC2 WMFWCNYKBAYFGA-UHFFFAOYSA-N 0.000 description 1
- WIUKUEBVMRRYCA-UHFFFAOYSA-N 6-bromo-N-cyclopropyl-8-methoxy-N,4,4-trimethyl-2,3-dihydro-1H-naphthalen-1-amine Chemical compound COC1=CC(Br)=CC(C(CC2)(C)C)=C1C2N(C)C1CC1 WIUKUEBVMRRYCA-UHFFFAOYSA-N 0.000 description 1
- RKCWVAWAWDWBRN-UHFFFAOYSA-N 6-ethynyl-8-hydroxy-4,4-dimethyl-2,3-dihydronaphthalen-1-one Chemical compound C1=C(C#C)C=C2C(C)(C)CCC(=O)C2=C1O RKCWVAWAWDWBRN-UHFFFAOYSA-N 0.000 description 1
- HRZFUMHJMZEROT-UHFFFAOYSA-L 7681-57-4 Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])(=O)=O HRZFUMHJMZEROT-UHFFFAOYSA-L 0.000 description 1
- 229960004308 ACETYLCYSTEINE Drugs 0.000 description 1
- 206010000269 Abscess Diseases 0.000 description 1
- 206010000503 Acne cystic Diseases 0.000 description 1
- 206010049141 Acne fulminans Diseases 0.000 description 1
- 206010064930 Age-related macular degeneration Diseases 0.000 description 1
- 208000010002 Alcoholic Liver Cirrhosis Diseases 0.000 description 1
- 241000269328 Amphibia Species 0.000 description 1
- 206010002556 Ankylosing spondylitis Diseases 0.000 description 1
- 206010002855 Anxiety Diseases 0.000 description 1
- 206010057666 Anxiety disease Diseases 0.000 description 1
- 206010059512 Apoptosis Diseases 0.000 description 1
- 206010003210 Arteriosclerosis Diseases 0.000 description 1
- 208000006820 Arthralgia Diseases 0.000 description 1
- 208000006673 Asthma Diseases 0.000 description 1
- 206010003694 Atrophy Diseases 0.000 description 1
- 208000006096 Attention Deficit Disorder with Hyperactivity Diseases 0.000 description 1
- 206010003736 Attention deficit/hyperactivity disease Diseases 0.000 description 1
- 208000001992 Autosomal Dominant Optic Atrophy Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 229960000686 Benzalkonium Chloride Drugs 0.000 description 1
- 229940050390 Benzoate Drugs 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical compound OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 1
- 210000000988 Bone and Bones Anatomy 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 210000000481 Breast Anatomy 0.000 description 1
- 206010006451 Bronchitis Diseases 0.000 description 1
- 206010006458 Bronchitis chronic Diseases 0.000 description 1
- 210000001217 Buttocks Anatomy 0.000 description 1
- 229940043253 Butylated Hydroxyanisole Drugs 0.000 description 1
- 229940095259 Butylated Hydroxytoluene Drugs 0.000 description 1
- 239000004255 Butylated hydroxyanisole Substances 0.000 description 1
- CZBZUDVBLSSABA-UHFFFAOYSA-N Butylated hydroxyanisole Chemical compound COC1=CC=C(O)C(C(C)(C)C)=C1.COC1=CC=C(O)C=C1C(C)(C)C CZBZUDVBLSSABA-UHFFFAOYSA-N 0.000 description 1
- 239000004322 Butylated hydroxytoluene Substances 0.000 description 1
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 1
- 101700040785 CHR5 Proteins 0.000 description 1
- MIOPJNTWMNEORI-UHFFFAOYSA-N Camphorsulfonic acid Chemical compound C1CC2(CS(O)(=O)=O)C(=O)CC1C2(C)C MIOPJNTWMNEORI-UHFFFAOYSA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- QOSSAOTZNIDXMA-UHFFFAOYSA-N Carbodicyclohexylimide Chemical compound C1CCCCC1N=C=NC1CCCCC1 QOSSAOTZNIDXMA-UHFFFAOYSA-N 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate dianion Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-N Carbonic acid Chemical compound OC(O)=O BVKZGUZCCUSVTD-UHFFFAOYSA-N 0.000 description 1
- 208000008787 Cardiovascular Disease Diseases 0.000 description 1
- 210000001715 Carotid Arteries Anatomy 0.000 description 1
- 210000003679 Cervix Uteri Anatomy 0.000 description 1
- OSASVXMJTNOKOY-UHFFFAOYSA-N Chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 1
- 229960004926 Chlorobutanol Drugs 0.000 description 1
- 208000007451 Chronic Bronchitis Diseases 0.000 description 1
- 206010057668 Cognitive disease Diseases 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 210000001072 Colon Anatomy 0.000 description 1
- 210000002808 Connective Tissue Anatomy 0.000 description 1
- 206010011401 Crohn's disease Diseases 0.000 description 1
- 108010015742 Cytochrome P-450 Enzyme System Proteins 0.000 description 1
- 102000003849 Cytochrome P450 Human genes 0.000 description 1
- FBPFZTCFMRRESA-KAZBKCHUSA-N D-Mannitol Natural products OC[C@@H](O)[C@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KAZBKCHUSA-N 0.000 description 1
- RGHNJXZEOKUKBD-SQOUGZDYSA-M D-gluconate Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O RGHNJXZEOKUKBD-SQOUGZDYSA-M 0.000 description 1
- 206010012438 Dermatitis atopic Diseases 0.000 description 1
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 1
- 206010013663 Drug dependence Diseases 0.000 description 1
- 206010013774 Dry eye Diseases 0.000 description 1
- 210000001198 Duodenum Anatomy 0.000 description 1
- 102000008491 Dynamins Human genes 0.000 description 1
- 108010020522 Dynamins Proteins 0.000 description 1
- 206010058108 Dyslipidaemia Diseases 0.000 description 1
- 206010058314 Dysplasia Diseases 0.000 description 1
- 229950008913 Edisilate Drugs 0.000 description 1
- 229950005627 Embonate Drugs 0.000 description 1
- 240000006775 Enicostema verticillatum Species 0.000 description 1
- 210000000981 Epithelium Anatomy 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 210000003238 Esophagus Anatomy 0.000 description 1
- 206010064503 Excessive skin Diseases 0.000 description 1
- 229940012356 Eye Drops Drugs 0.000 description 1
- 230000035693 Fab Effects 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 206010060919 Foetal malformation Diseases 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 229960001731 GLUCEPTATE Drugs 0.000 description 1
- 208000008665 Gastrointestinal Disease Diseases 0.000 description 1
- 210000004907 Glands Anatomy 0.000 description 1
- 229940049906 Glutamate Drugs 0.000 description 1
- 229940093915 Gynecological Organic acids Drugs 0.000 description 1
- 210000003128 Head Anatomy 0.000 description 1
- 206010019233 Headache Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 210000001320 Hippocampus Anatomy 0.000 description 1
- 238000006546 Horner-Wadsworth-Emmons reaction Methods 0.000 description 1
- 201000001971 Huntington's disease Diseases 0.000 description 1
- 208000003532 Hypothyroidism Diseases 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 101710004181 INTS2 Proteins 0.000 description 1
- 206010021198 Ichthyosis Diseases 0.000 description 1
- 206010021197 Ichthyosis Diseases 0.000 description 1
- 208000009326 Ileitis Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 208000002551 Irritable Bowel Syndrome Diseases 0.000 description 1
- FMKOJHQHASLBPH-UHFFFAOYSA-N Isopropyl iodide Chemical compound CC(C)I FMKOJHQHASLBPH-UHFFFAOYSA-N 0.000 description 1
- 208000002260 Keloid Diseases 0.000 description 1
- 210000001117 Keloid Anatomy 0.000 description 1
- 206010023330 Keloid scar Diseases 0.000 description 1
- XMAYWYJOQHXEEK-OZXSUGGESA-N Ketoconazole Chemical compound C1CN(C(=O)C)CCN1C(C=C1)=CC=C1OC[C@@H]1O[C@@](CN2C=NC=C2)(C=2C(=CC(Cl)=CC=2)Cl)OC1 XMAYWYJOQHXEEK-OZXSUGGESA-N 0.000 description 1
- 210000003734 Kidney Anatomy 0.000 description 1
- 241000235058 Komagataella pastoris Species 0.000 description 1
- PWKSKIMOESPYIA-BYPYZUCNSA-N L-N-acetyl-Cysteine Chemical compound CC(=O)N[C@@H](CS)C(O)=O PWKSKIMOESPYIA-BYPYZUCNSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- 229940001447 Lactate Drugs 0.000 description 1
- JYTUSYBCFIZPBE-AMTLMPIISA-N Lactobionic acid Chemical compound OC(=O)[C@H](O)[C@@H](O)[C@@H]([C@H](O)CO)O[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O JYTUSYBCFIZPBE-AMTLMPIISA-N 0.000 description 1
- GUBGYTABKSRVRQ-UUNJERMWSA-N Lactose Natural products O([C@@H]1[C@H](O)[C@H](O)[C@H](O)O[C@@H]1CO)[C@H]1[C@@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1 GUBGYTABKSRVRQ-UUNJERMWSA-N 0.000 description 1
- 210000000867 Larynx Anatomy 0.000 description 1
- 208000009625 Lesch-Nyhan Syndrome Diseases 0.000 description 1
- 210000000265 Leukocytes Anatomy 0.000 description 1
- UGFHIPBXIWJXNA-UHFFFAOYSA-N Liarozole Chemical compound ClC1=CC=CC(C(C=2C=C3NC=NC3=CC=2)N2C=NC=C2)=C1 UGFHIPBXIWJXNA-UHFFFAOYSA-N 0.000 description 1
- 206010061523 Lip and/or oral cavity cancer Diseases 0.000 description 1
- 229940040692 Lithium Hydroxide Monohydrate Drugs 0.000 description 1
- 210000004185 Liver Anatomy 0.000 description 1
- 210000001165 Lymph Nodes Anatomy 0.000 description 1
- 210000004324 Lymphatic System Anatomy 0.000 description 1
- 102100014726 MECP2 Human genes 0.000 description 1
- 101700029603 MECP2 Proteins 0.000 description 1
- 102100004834 MT-ND4 Human genes 0.000 description 1
- 101710028315 MT-ND4 Proteins 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- MBBZMMPHUWSWHV-BDVNFPICSA-N Meglumine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 1
- 206010054949 Metaplasia Diseases 0.000 description 1
- 206010027476 Metastasis Diseases 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- LRMHVVPPGGOAJQ-UHFFFAOYSA-N Methyl nitrate Chemical compound CO[N+]([O-])=O LRMHVVPPGGOAJQ-UHFFFAOYSA-N 0.000 description 1
- 210000003657 Middle Cerebral Artery Anatomy 0.000 description 1
- 206010027626 Milia Diseases 0.000 description 1
- 210000002027 Muscle, Skeletal Anatomy 0.000 description 1
- FAWHJXHQSYBMFW-UHFFFAOYSA-N N-[(6-bromo-2,2,4,4-tetramethyl-3H-chromen-8-yl)methyl]-N-methylcyclopropanamine Chemical compound C=1C(Br)=CC(C(CC(C)(C)O2)(C)C)=C2C=1CN(C)C1CC1 FAWHJXHQSYBMFW-UHFFFAOYSA-N 0.000 description 1
- CNEHJYULFIDLBZ-UHFFFAOYSA-N N-cyclopropyl-8-methoxy-N,4,4-trimethyl-5-(2-trimethylsilylethynyl)-2,3-dihydro-1H-naphthalen-1-amine Chemical compound COC1=CC=C(C#C[Si](C)(C)C)C(C(CC2)(C)C)=C1C2N(C)C1CC1 CNEHJYULFIDLBZ-UHFFFAOYSA-N 0.000 description 1
- 150000001204 N-oxides Chemical class 0.000 description 1
- 210000003739 Neck Anatomy 0.000 description 1
- 102100014017 ODAM Human genes 0.000 description 1
- 108060005663 ODAM Proteins 0.000 description 1
- 229940049964 Oleate Drugs 0.000 description 1
- 229920000272 Oligonucleotide Polymers 0.000 description 1
- 206010061323 Optic neuropathy Diseases 0.000 description 1
- 108009000578 Oxidative Stress Proteins 0.000 description 1
- WLJNZVDCPSBLRP-UHFFFAOYSA-N Pamoic acid Chemical compound C1=CC=C2C(CC=3C4=CC=CC=C4C=C(C=3O)C(=O)O)=C(O)C(C(O)=O)=CC2=C1 WLJNZVDCPSBLRP-UHFFFAOYSA-N 0.000 description 1
- 208000003154 Papilloma Diseases 0.000 description 1
- 206010061536 Parkinson's disease Diseases 0.000 description 1
- 210000001428 Peripheral Nervous System Anatomy 0.000 description 1
- 229940096826 Phenylmercuric Acetate Drugs 0.000 description 1
- 229960002553 Phenylmercuric nitrate Drugs 0.000 description 1
- XEBWQGVWTUSTLN-UHFFFAOYSA-M Phenylmercury acetate Chemical compound CC(=O)O[Hg]C1=CC=CC=C1 XEBWQGVWTUSTLN-UHFFFAOYSA-M 0.000 description 1
- 206010062080 Pigmentation disease Diseases 0.000 description 1
- 210000003635 Pituitary Gland Anatomy 0.000 description 1
- 206010035116 Pityriasis rubra pilaris Diseases 0.000 description 1
- 208000002151 Pleural Effusion Diseases 0.000 description 1
- 206010036590 Premature baby Diseases 0.000 description 1
- 210000002307 Prostate Anatomy 0.000 description 1
- 208000005333 Pulmonary Edema Diseases 0.000 description 1
- 206010037423 Pulmonary oedema Diseases 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- PBMFSQRYOILNGV-UHFFFAOYSA-N Pyridazine Chemical compound C1=CC=NN=C1 PBMFSQRYOILNGV-UHFFFAOYSA-N 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J Pyrophosphate Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 206010068733 Respiratory fatigue Diseases 0.000 description 1
- 210000001927 Retinal Artery Anatomy 0.000 description 1
- 102000018822 Retinal Dehydrogenase Human genes 0.000 description 1
- 108010027691 Retinal Dehydrogenase Proteins 0.000 description 1
- 208000004644 Retinal Vein Occlusion Diseases 0.000 description 1
- 208000007014 Retinitis Pigmentosa Diseases 0.000 description 1
- 208000006289 Rett Syndrome Diseases 0.000 description 1
- 206010039073 Rheumatoid arthritis Diseases 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 241000490025 Schefflera digitata Species 0.000 description 1
- 206010039710 Scleroderma Diseases 0.000 description 1
- 210000002966 Serum Anatomy 0.000 description 1
- 208000007056 Sickle Cell Anemia Diseases 0.000 description 1
- 206010040882 Skin lesion Diseases 0.000 description 1
- 229940083599 Sodium Iodide Drugs 0.000 description 1
- UKLNMMHNWFDKNT-UHFFFAOYSA-M Sodium chlorite Chemical compound [Na+].[O-]Cl=O UKLNMMHNWFDKNT-UHFFFAOYSA-M 0.000 description 1
- 208000008513 Spinal Cord Injury Diseases 0.000 description 1
- 206010041834 Squamous cell carcinoma of skin Diseases 0.000 description 1
- 206010041848 Squamous cell carcinoma of the cervix Diseases 0.000 description 1
- 241000519995 Stachys sylvatica Species 0.000 description 1
- 229920002253 Tannate Polymers 0.000 description 1
- 206010043118 Tardive dyskinesia Diseases 0.000 description 1
- NAVMQTYZDKMPEU-UHFFFAOYSA-N Targretin Chemical compound CC1=CC(C(CCC2(C)C)(C)C)=C2C=C1C(=C)C1=CC=C(C(O)=O)C=C1 NAVMQTYZDKMPEU-UHFFFAOYSA-N 0.000 description 1
- 210000001541 Thymus Gland Anatomy 0.000 description 1
- 208000000323 Tourette Syndrome Diseases 0.000 description 1
- 206010044126 Tourette's disease Diseases 0.000 description 1
- 231100000765 Toxin Toxicity 0.000 description 1
- 229940035504 Tromethamine Drugs 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- 210000003932 Urinary Bladder Anatomy 0.000 description 1
- 210000004291 Uterus Anatomy 0.000 description 1
- 206010053648 Vascular occlusion Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 206010047700 Vomiting Diseases 0.000 description 1
- 238000007239 Wittig reaction Methods 0.000 description 1
- 241000269370 Xenopus <genus> Species 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K [O-]P([O-])([O-])=O Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000008351 acetate buffer Substances 0.000 description 1
- BPGDAMSIGCZZLK-UHFFFAOYSA-N acetyloxymethyl acetate Chemical group CC(=O)OCOC(C)=O BPGDAMSIGCZZLK-UHFFFAOYSA-N 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 229960005339 acitretin Drugs 0.000 description 1
- 230000003213 activating Effects 0.000 description 1
- 229960002916 adapalene Drugs 0.000 description 1
- LZCDAPDGXCYOEH-UHFFFAOYSA-N adapalene Chemical compound C1=C(C(O)=O)C=CC2=CC(C3=CC=C(C(=C3)C34CC5CC(CC(C5)C3)C4)OC)=CC=C21 LZCDAPDGXCYOEH-UHFFFAOYSA-N 0.000 description 1
- 230000001058 adult Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 231100000494 adverse effect Toxicity 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- GGCUJPCCTQNTJF-FRCNGJHJSA-N all-trans-4-oxoretinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)C(=O)CCC1(C)C GGCUJPCCTQNTJF-FRCNGJHJSA-N 0.000 description 1
- IHUNBGSDBOWDMA-AQFIFDHZSA-N all-trans-acitretin Chemical compound COC1=CC(C)=C(\C=C\C(\C)=C\C=C\C(\C)=C\C(O)=O)C(C)=C1C IHUNBGSDBOWDMA-AQFIFDHZSA-N 0.000 description 1
- NCYCYZXNIZJOKI-OVSJKPMPSA-N all-trans-retinaldehyde Chemical compound O=C\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C NCYCYZXNIZJOKI-OVSJKPMPSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-N ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 1
- 238000002399 angioplasty Methods 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007172 antigens Proteins 0.000 description 1
- 102000038129 antigens Human genes 0.000 description 1
- 230000036506 anxiety Effects 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 230000036528 appetite Effects 0.000 description 1
- 235000019789 appetite Nutrition 0.000 description 1
- 150000004982 aromatic amines Chemical class 0.000 description 1
- RQNWIZPPADIBDY-UHFFFAOYSA-N arsenic Chemical compound [As] RQNWIZPPADIBDY-UHFFFAOYSA-N 0.000 description 1
- 229910052785 arsenic Inorganic materials 0.000 description 1
- 125000003710 aryl alkyl group Chemical group 0.000 description 1
- 201000001320 atherosclerosis Diseases 0.000 description 1
- 201000008937 atopic dermatitis Diseases 0.000 description 1
- 201000006287 attention deficit hyperactivity disease Diseases 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-M benzenesulfonate Chemical compound [O-]S(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-M 0.000 description 1
- 229940077388 benzenesulfonate Drugs 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-M benzoate Chemical compound [O-]C(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-M 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 229940007550 benzyl acetate Drugs 0.000 description 1
- 125000001743 benzylic group Chemical group 0.000 description 1
- 229960002938 bexarotene Drugs 0.000 description 1
- 230000001588 bifunctional Effects 0.000 description 1
- AZWXAPCAJCYGIA-UHFFFAOYSA-N bis(2-methylpropyl)alumane Chemical compound CC(C)C[AlH]CC(C)C AZWXAPCAJCYGIA-UHFFFAOYSA-N 0.000 description 1
- 201000002393 blood protein disease Diseases 0.000 description 1
- 230000024279 bone resorption Effects 0.000 description 1
- 229910000085 borane Inorganic materials 0.000 description 1
- MCQRPQCQMGVWIQ-UHFFFAOYSA-N boron;methylsulfanylmethane Chemical compound [B].CSC MCQRPQCQMGVWIQ-UHFFFAOYSA-N 0.000 description 1
- 201000009267 bronchiectasis Diseases 0.000 description 1
- 235000019282 butylated hydroxyanisole Nutrition 0.000 description 1
- 235000010354 butylated hydroxytoluene Nutrition 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- 159000000007 calcium salts Chemical class 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 125000001951 carbamoylamino group Chemical group C(N)(=O)N* 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 235000011089 carbon dioxide Nutrition 0.000 description 1
- 125000006297 carbonyl amino group Chemical group [H]N([*:2])C([*:1])=O 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 150000001735 carboxylic acids Chemical class 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000035569 catabolism Effects 0.000 description 1
- 230000024881 catalytic activity Effects 0.000 description 1
- 238000006555 catalytic reaction Methods 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 230000001413 cellular Effects 0.000 description 1
- 230000019522 cellular metabolic process Effects 0.000 description 1
- 230000002490 cerebral Effects 0.000 description 1
- 210000000038 chest Anatomy 0.000 description 1
- 108091006028 chimera Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 229940117975 chromium trioxide Drugs 0.000 description 1
- 229940114081 cinnamate Drugs 0.000 description 1
- 150000001851 cinnamic acid derivatives Chemical class 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 230000002860 competitive Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 201000003883 cystic fibrosis Diseases 0.000 description 1
- 239000008367 deionised water Substances 0.000 description 1
- 230000002939 deleterious Effects 0.000 description 1
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 1
- 230000001419 dependent Effects 0.000 description 1
- 239000007933 dermal patch Substances 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 125000005265 dialkylamine group Chemical group 0.000 description 1
- YXHKONLOYHBTNS-UHFFFAOYSA-N diazomethane Chemical compound C=[N+]=[N-] YXHKONLOYHBTNS-UHFFFAOYSA-N 0.000 description 1
- DMJZZSLVPSMWCS-UHFFFAOYSA-N diborane Chemical compound B1[H]B[H]1 DMJZZSLVPSMWCS-UHFFFAOYSA-N 0.000 description 1
- 125000004177 diethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- SPCNPOWOBZQWJK-UHFFFAOYSA-N dimethoxy-(2-propan-2-ylsulfanylethylsulfanyl)-sulfanylidene-$l^{5}-phosphane Chemical compound COP(=S)(OC)SCCSC(C)C SPCNPOWOBZQWJK-UHFFFAOYSA-N 0.000 description 1
- LCGLNKUTAGEVQW-UHFFFAOYSA-N dimethyl ether Chemical compound COC LCGLNKUTAGEVQW-UHFFFAOYSA-N 0.000 description 1
- 125000000118 dimethyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 125000002147 dimethylamino group Chemical group [H]C([H])([H])N(*)C([H])([H])[H] 0.000 description 1
- SXZIXHOMFPUIRK-UHFFFAOYSA-N diphenylmethanimine Chemical compound C=1C=CC=CC=1C(=N)C1=CC=CC=C1 SXZIXHOMFPUIRK-UHFFFAOYSA-N 0.000 description 1
- 239000001177 diphosphate Substances 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- 201000009910 diseases by infectious agent Diseases 0.000 description 1
- 235000014632 disordered eating Nutrition 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 230000002222 downregulating Effects 0.000 description 1
- 201000006180 eating disease Diseases 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 210000002257 embryonic structures Anatomy 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 150000002169 ethanolamines Chemical class 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- JBNUOBRBOFMWNY-FOCLMDBBSA-N ethyl (E)-3-[4-[2-(3-tert-butylphenyl)ethynyl]phenyl]prop-2-enoate Chemical compound C1=CC(/C=C/C(=O)OCC)=CC=C1C#CC1=CC=CC(C(C)(C)C)=C1 JBNUOBRBOFMWNY-FOCLMDBBSA-N 0.000 description 1
- BLHSNYOCMZLILT-UHFFFAOYSA-N ethyl 2-chloro-2-dipropylphosphorylacetate Chemical compound CCCP(=O)(CCC)C(Cl)C(=O)OCC BLHSNYOCMZLILT-UHFFFAOYSA-N 0.000 description 1
- KGAIOCPMJIVBAY-UHFFFAOYSA-N ethyl 2-chloro-3-(4-iodophenyl)prop-2-enoate Chemical compound CCOC(=O)C(Cl)=CC1=CC=C(I)C=C1 KGAIOCPMJIVBAY-UHFFFAOYSA-N 0.000 description 1
- BVSRWCMAJISCTD-UHFFFAOYSA-N ethyl 2-diethoxyphosphorylpropanoate Chemical compound CCOC(=O)C(C)P(=O)(OCC)OCC BVSRWCMAJISCTD-UHFFFAOYSA-N 0.000 description 1
- YCISVIBSKPOXRF-UHFFFAOYSA-N ethyl 3-(3-iodophenyl)prop-2-enoate Chemical compound CCOC(=O)C=CC1=CC=CC(I)=C1 YCISVIBSKPOXRF-UHFFFAOYSA-N 0.000 description 1
- XHEQVHPHSNNRMH-UHFFFAOYSA-N ethyl 3-[3-[2-(2,2,4,4-tetramethyl-8-propan-2-yl-3H-chromen-6-yl)ethynyl]phenyl]prop-2-enoate Chemical compound CCOC(=O)C=CC1=CC=CC(C#CC=2C=C3C(C)(C)CC(C)(C)OC3=C(C(C)C)C=2)=C1 XHEQVHPHSNNRMH-UHFFFAOYSA-N 0.000 description 1
- GQMVGSATYCSKQN-UHFFFAOYSA-N ethyl 3-[4-[2-(5-tert-butyl-2-methylphenyl)ethynyl]phenyl]prop-2-enoate Chemical compound C1=CC(C=CC(=O)OCC)=CC=C1C#CC1=CC(C(C)(C)C)=CC=C1C GQMVGSATYCSKQN-UHFFFAOYSA-N 0.000 description 1
- BBNIPWJUXAMRFO-UHFFFAOYSA-N ethyl 3-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]phenyl]ethynyl]phenyl]but-2-enoate Chemical compound C1=CC(C(C)=CC(=O)OCC)=CC=C1C#CC1=CC=CC(CN(C)C2CC2)=C1 BBNIPWJUXAMRFO-UHFFFAOYSA-N 0.000 description 1
- OORHNGIDEZMVSP-UHFFFAOYSA-N ethyl 4-(4-bromo-2-methoxyphenyl)butanoate Chemical compound CCOC(=O)CCCC1=CC=C(Br)C=C1OC OORHNGIDEZMVSP-UHFFFAOYSA-N 0.000 description 1
- YCBJOQUNPLTBGG-UHFFFAOYSA-N ethyl 4-iodobenzoate Chemical compound CCOC(=O)C1=CC=C(I)C=C1 YCBJOQUNPLTBGG-UHFFFAOYSA-N 0.000 description 1
- DVTNTCIRUXFHMO-UHFFFAOYSA-N ethyl 8-[[cyclopropyl(propan-2-yl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromene-6-carboxylate Chemical compound C=12OC(C)(C)CC(C)(C)C2=CC(C(=O)OCC)=CC=1CN(C(C)C)C1CC1 DVTNTCIRUXFHMO-UHFFFAOYSA-N 0.000 description 1
- DMOITFQMOBGHCT-UHFFFAOYSA-N ethyl 8-[cyclopropyl(methyl)amino]-3-[2-[4-(2-methoxy-2-oxoethyl)phenyl]ethynyl]-5,5-dimethyl-7,8-dihydro-6H-naphthalene-1-carboxylate Chemical compound CCOC(=O)C1=CC(C#CC=2C=CC(CC(=O)OC)=CC=2)=CC(C(CC2)(C)C)=C1C2N(C)C1CC1 DMOITFQMOBGHCT-UHFFFAOYSA-N 0.000 description 1
- OAYLNYINCPYISS-UHFFFAOYSA-N ethyl acetate;hexane Chemical compound CCCCCC.CCOC(C)=O OAYLNYINCPYISS-UHFFFAOYSA-N 0.000 description 1
- FFLYUXVZEPLMCL-UHFFFAOYSA-N ethylchloranuidyl formate Chemical compound CC[Cl-]OC=O FFLYUXVZEPLMCL-UHFFFAOYSA-N 0.000 description 1
- 239000003889 eye drop Substances 0.000 description 1
- 239000003885 eye ointment Substances 0.000 description 1
- 230000004438 eyesight Effects 0.000 description 1
- 230000001605 fetal Effects 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 230000003176 fibrotic Effects 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 125000002485 formyl group Chemical group [H]C(*)=O 0.000 description 1
- 229940050411 fumarate Drugs 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- DSLZVSRJTYRBFB-DUHBMQHGSA-L galactarate(2-) Chemical compound [O-]C(=O)[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)C([O-])=O DSLZVSRJTYRBFB-DUHBMQHGSA-L 0.000 description 1
- 230000002068 genetic Effects 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- 229940050410 gluconate Drugs 0.000 description 1
- PEDCQBHIVMGVHV-UHFFFAOYSA-N glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 159000000011 group IA salts Chemical class 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- XGIHQYAWBCFNPY-AZOCGYLKSA-N hydrabamine Chemical compound C([C@@H]12)CC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC[C@@]1(C)CNCCNC[C@@]1(C)[C@@H]2CCC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC1 XGIHQYAWBCFNPY-AZOCGYLKSA-N 0.000 description 1
- 150000004677 hydrates Chemical class 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-M hydrogensulfate Chemical compound OS([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-M 0.000 description 1
- 230000003463 hyperproliferative Effects 0.000 description 1
- 230000001969 hypertrophic Effects 0.000 description 1
- 230000002989 hypothyroidism Effects 0.000 description 1
- 230000001146 hypoxic Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000001771 impaired Effects 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 229910001410 inorganic ion Inorganic materials 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-M iodide Chemical compound [I-] XMBWDFGMSWQBCA-UHFFFAOYSA-M 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- KFZMGEQAYNKOFK-UHFFFAOYSA-N iso-propanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 1
- 230000003780 keratinization Effects 0.000 description 1
- 229960004125 ketoconazole Drugs 0.000 description 1
- 150000002576 ketones Chemical class 0.000 description 1
- 238000011005 laboratory method Methods 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 229940099584 lactobionate Drugs 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- POULHZVOKOAJMA-UHFFFAOYSA-M laurate Chemical compound CCCCCCCCCCCC([O-])=O POULHZVOKOAJMA-UHFFFAOYSA-M 0.000 description 1
- 229940070765 laurate Drugs 0.000 description 1
- 229950007056 liarozole Drugs 0.000 description 1
- 201000011486 lichen planus Diseases 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000004301 light adaptation Effects 0.000 description 1
- 239000004571 lime Substances 0.000 description 1
- 230000000670 limiting Effects 0.000 description 1
- UBJFKNSINUCEAL-UHFFFAOYSA-N lithium;2-methylpropane Chemical compound [Li+].C[C-](C)C UBJFKNSINUCEAL-UHFFFAOYSA-N 0.000 description 1
- 235000015250 liver sausages Nutrition 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 159000000003 magnesium salts Chemical class 0.000 description 1
- 229940049920 malate Drugs 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-L malate(2-) Chemical compound [O-]C(=O)C(O)CC([O-])=O BJEPYKJPYRNKOW-UHFFFAOYSA-L 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-L maleate(2-) Chemical compound [O-]C(=O)\C=C/C([O-])=O VZCYOOQTPOCHFL-UPHRSURJSA-L 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 230000003211 malignant Effects 0.000 description 1
- 210000004962 mammalian cells Anatomy 0.000 description 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-M mandelate Chemical compound [O-]C(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-M 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000015689 metaplastic ossification Effects 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 1
- BWCLTDMGMKAHGZ-NTCAYCPXSA-N methyl (E)-3-[4-[2-[3-[[cyclopropyl(methyl)amino]methyl]phenyl]ethynyl]phenyl]prop-2-enoate Chemical compound C1=CC(/C=C/C(=O)OC)=CC=C1C#CC1=CC=CC(CN(C)C2CC2)=C1 BWCLTDMGMKAHGZ-NTCAYCPXSA-N 0.000 description 1
- LHYLPYONWHGNIH-UHFFFAOYSA-N methyl 2-[4-[2-(2,2,4,4-tetramethyl-8-propan-2-yloxy-3H-chromen-6-yl)ethynyl]phenyl]acetate Chemical compound C1=CC(CC(=O)OC)=CC=C1C#CC1=CC(OC(C)C)=C(OC(C)(C)CC2(C)C)C2=C1 LHYLPYONWHGNIH-UHFFFAOYSA-N 0.000 description 1
- YFUVRUPACLMTHL-UHFFFAOYSA-N methyl 2-[4-[2-(8-hydroxy-2,2,4,4-tetramethyl-3H-chromen-6-yl)ethynyl]phenyl]-2-methylpropanoate Chemical compound C1=CC(C(C)(C)C(=O)OC)=CC=C1C#CC1=CC(O)=C(OC(C)(C)CC2(C)C)C2=C1 YFUVRUPACLMTHL-UHFFFAOYSA-N 0.000 description 1
- XRWIJGXECPJEPE-UHFFFAOYSA-N methyl 2-[4-[2-[8-[[cyclopropyl(methyl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromen-6-yl]ethynyl]phenyl]acetate Chemical compound C1=CC(CC(=O)OC)=CC=C1C#CC1=CC(CN(C)C2CC2)=C(OC(C)(C)CC2(C)C)C2=C1 XRWIJGXECPJEPE-UHFFFAOYSA-N 0.000 description 1
- VGVUWFYKDRYHKH-UHFFFAOYSA-N methyl 2-[4-[2-[8-[[cyclopropyl(methyl)amino]methyl]-2,2,4,4-tetramethyl-3H-chromen-6-yl]ethynyl]phenyl]propanoate Chemical compound C1=CC(C(C)C(=O)OC)=CC=C1C#CC1=CC(CN(C)C2CC2)=C(OC(C)(C)CC2(C)C)C2=C1 VGVUWFYKDRYHKH-UHFFFAOYSA-N 0.000 description 1
- FQDDBYGPHUUTRN-UHFFFAOYSA-N methyl 4-isocyanatobenzoate Chemical compound COC(=O)C1=CC=C(N=C=O)C=C1 FQDDBYGPHUUTRN-UHFFFAOYSA-N 0.000 description 1
- 229940102396 methyl bromide Drugs 0.000 description 1
- QMMFVYPAHWMCMS-UHFFFAOYSA-N methyl sulfide Chemical compound CSC QMMFVYPAHWMCMS-UHFFFAOYSA-N 0.000 description 1
- 230000000813 microbial Effects 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 230000002438 mitochondrial Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 102000005614 monoclonal antibodies Human genes 0.000 description 1
- 108010045030 monoclonal antibodies Proteins 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 239000005445 natural product Substances 0.000 description 1
- 229930014626 natural products Natural products 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 230000001264 neutralization Effects 0.000 description 1
- 229960003512 nicotinic acid Drugs 0.000 description 1
- 235000001968 nicotinic acid Nutrition 0.000 description 1
- 239000011664 nicotinic acid Substances 0.000 description 1
- NHNBFGGVMKEFGY-UHFFFAOYSA-N nitrate Chemical compound [O-][N+]([O-])=O NHNBFGGVMKEFGY-UHFFFAOYSA-N 0.000 description 1
- PDTFCHSETJBPTR-UHFFFAOYSA-N nitrooxy(phenyl)mercury Chemical compound [O-][N+](=O)O[Hg]C1=CC=CC=C1 PDTFCHSETJBPTR-UHFFFAOYSA-N 0.000 description 1
- 230000000414 obstructive Effects 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-M oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC([O-])=O ZQPPMHVWECSIRJ-KTKRTIGZSA-M 0.000 description 1
- 229920001542 oligosaccharide Polymers 0.000 description 1
- 150000002482 oligosaccharides Polymers 0.000 description 1
- 210000000287 oocyte Anatomy 0.000 description 1
- 230000003287 optical Effects 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 description 1
- 230000011164 ossification Effects 0.000 description 1
- 230000036542 oxidative stress Effects 0.000 description 1
- 229910052763 palladium Inorganic materials 0.000 description 1
- LXNAVEXFUKBNMK-UHFFFAOYSA-N palladium(II) acetate Substances [Pd].CC(O)=O.CC(O)=O LXNAVEXFUKBNMK-UHFFFAOYSA-N 0.000 description 1
- IPCSVZSSVZVIGE-UHFFFAOYSA-M palmitate Chemical compound CCCCCCCCCCCCCCCC([O-])=O IPCSVZSSVZVIGE-UHFFFAOYSA-M 0.000 description 1
- 229940014662 pantothenate Drugs 0.000 description 1
- 235000019161 pantothenic acid Nutrition 0.000 description 1
- 239000011713 pantothenic acid Substances 0.000 description 1
- 230000001717 pathogenic Effects 0.000 description 1
- 244000052769 pathogens Species 0.000 description 1
- OFBQJSOFQDEBGM-UHFFFAOYSA-N pentane Chemical compound CCCCC OFBQJSOFQDEBGM-UHFFFAOYSA-N 0.000 description 1
- 238000009527 percussion Methods 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000000649 photocoagulation Effects 0.000 description 1
- 238000002428 photodynamic therapy Methods 0.000 description 1
- 230000019612 pigmentation Effects 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- 235000011164 potassium chloride Nutrition 0.000 description 1
- 159000000001 potassium salts Chemical class 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940002612 prodrugs Drugs 0.000 description 1
- 230000000750 progressive Effects 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- GLEUVCSJPNTZKQ-UHFFFAOYSA-N propan-2-yl 2,2,4,4-tetramethyl-6-(2-trimethylsilylethynyl)-3H-chromene-8-carboxylate Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(C(=O)OC(C)C)=CC(C#C[Si](C)(C)C)=C2 GLEUVCSJPNTZKQ-UHFFFAOYSA-N 0.000 description 1
- DRJYYLPFRVQFKC-UHFFFAOYSA-N propan-2-yl 3-[2-[4-[2-(acetyloxymethoxy)-2-oxoethyl]-3-fluorophenyl]ethynyl]-5-ethynylbenzoate Chemical compound CC(C)OC(=O)C1=CC(C#C)=CC(C#CC=2C=C(F)C(CC(=O)OCOC(C)=O)=CC=2)=C1 DRJYYLPFRVQFKC-UHFFFAOYSA-N 0.000 description 1
- QQZNJBYWGHVRSN-UHFFFAOYSA-N propan-2-yl 6-bromo-2,2,4,4-tetramethyl-3H-chromene-8-carboxylate Chemical compound O1C(C)(C)CC(C)(C)C2=C1C(C(=O)OC(C)C)=CC(Br)=C2 QQZNJBYWGHVRSN-UHFFFAOYSA-N 0.000 description 1
- XBDQKXXYIPTUBI-UHFFFAOYSA-M propionate Chemical compound CCC([O-])=O XBDQKXXYIPTUBI-UHFFFAOYSA-M 0.000 description 1
- KYQCOXFCLRTKLS-UHFFFAOYSA-N pyrazine Chemical compound C1=CN=CC=N1 KYQCOXFCLRTKLS-UHFFFAOYSA-N 0.000 description 1
- 150000003242 quaternary ammonium salts Chemical class 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 230000014493 regulation of gene expression Effects 0.000 description 1
- 230000001105 regulatory Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 200000000008 restenosis Diseases 0.000 description 1
- 201000007737 retinal degeneration Diseases 0.000 description 1
- 230000004258 retinal degeneration Effects 0.000 description 1
- 102000027730 retinoid hormone receptors Human genes 0.000 description 1
- 108091008001 retinoid hormone receptors Proteins 0.000 description 1
- 229920002033 ribozyme Polymers 0.000 description 1
- YGSDEFSMJLZEOE-UHFFFAOYSA-M salicylate Chemical compound OC1=CC=CC=C1C([O-])=O YGSDEFSMJLZEOE-UHFFFAOYSA-M 0.000 description 1
- 229960001860 salicylate Drugs 0.000 description 1
- 230000037390 scarring Effects 0.000 description 1
- 201000000980 schizophrenia Diseases 0.000 description 1
- 150000003335 secondary amines Chemical class 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 231100000444 skin lesion Toxicity 0.000 description 1
- 201000010106 skin squamous cell carcinoma Diseases 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229960002218 sodium chlorite Drugs 0.000 description 1
- 235000009518 sodium iodide Nutrition 0.000 description 1
- 229940001584 sodium metabisulfite Drugs 0.000 description 1
- 235000010262 sodium metabisulphite Nutrition 0.000 description 1
- 159000000000 sodium salts Chemical class 0.000 description 1
- 229940001474 sodium thiosulfate Drugs 0.000 description 1
- ODGROJYWQXFQOZ-UHFFFAOYSA-N sodium;boron(1-) Chemical compound [B-].[Na+] ODGROJYWQXFQOZ-UHFFFAOYSA-N 0.000 description 1
- 210000004872 soft tissue Anatomy 0.000 description 1
- 241000894007 species Species 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-M stearate Chemical compound CCCCCCCCCCCCCCCCCC([O-])=O QIQXTHQIDYTFRH-UHFFFAOYSA-M 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 229940086735 succinate Drugs 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-L succinate(2-) Chemical compound [O-]C(=O)CCC([O-])=O KDYFGRWQOYBRFD-UHFFFAOYSA-L 0.000 description 1
- 239000001384 succinic acid Substances 0.000 description 1
- 235000011044 succinic acid Nutrition 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000002459 sustained Effects 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 229960001367 tartaric acid Drugs 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- 238000005496 tempering Methods 0.000 description 1
- ILMRJRBKQSSXGY-UHFFFAOYSA-N tert-butyl(dimethyl)silicon Chemical compound C[Si](C)C(C)(C)C ILMRJRBKQSSXGY-UHFFFAOYSA-N 0.000 description 1
- JOXIMZWYDAKGHI-UHFFFAOYSA-M toluene-4-sulfonate Chemical compound CC1=CC=C(S([O-])(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-M 0.000 description 1
- 230000002588 toxic Effects 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 108020003112 toxins Proteins 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- 125000005270 trialkylamine group Chemical group 0.000 description 1
- QIWRFOJWQSSRJZ-UHFFFAOYSA-N tributyl(ethenyl)stannane Chemical compound CCCC[Sn](CCCC)(CCCC)C=C QIWRFOJWQSSRJZ-UHFFFAOYSA-N 0.000 description 1
- FWSPXZXVNVQHIF-UHFFFAOYSA-N triethyl(ethynyl)silane Chemical group CC[Si](CC)(CC)C#C FWSPXZXVNVQHIF-UHFFFAOYSA-N 0.000 description 1
- 125000001889 triflyl group Chemical group FC(F)(F)S(*)(=O)=O 0.000 description 1
- UORVGPXVDQYIDP-UHFFFAOYSA-N trihydridoboron Substances B UORVGPXVDQYIDP-UHFFFAOYSA-N 0.000 description 1
- DLQYXUGCCKQSRJ-UHFFFAOYSA-N tris(furan-2-yl)phosphane Chemical compound C1=COC(P(C=2OC=CC=2)C=2OC=CC=2)=C1 DLQYXUGCCKQSRJ-UHFFFAOYSA-N 0.000 description 1
- 229960000281 trometamol Drugs 0.000 description 1
- 229960001322 trypsin Drugs 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 210000004881 tumor cells Anatomy 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 239000000439 tumor marker Substances 0.000 description 1
- 201000006704 ulcerative colitis Diseases 0.000 description 1
- 230000036967 uncompetitive Effects 0.000 description 1
- 230000004222 uncontrolled growth Effects 0.000 description 1
- NQPDZGIKBAWPEJ-UHFFFAOYSA-M valerate Chemical compound CCCCC([O-])=O NQPDZGIKBAWPEJ-UHFFFAOYSA-M 0.000 description 1
- 229940070710 valerate Drugs 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 230000003612 virological Effects 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Abstract
The invention provides methods for treating an individual having a retinoid responsive disorder. In one embodiment, a method involves administering to the individual an effective amount of a selective CYP26B inhibitor, the selective CYP26B inhibitor having at least 10-fold selectivity for CYP26B relative to CYP26A. In another embodiment, a method involves administering to the individual an effective amount of a selective CYP26A inhibitor, the selective CYP26A inhibitor having a chemical formula set forth in the specification. The invention further provides screening methods for identifying a selective CYP26A inhibitor or selective CYP26B inhibitor.
Description
METHODS TO TREAT RETINOID SENSITIVE DISORDERS USING SELECTIVE CYP26A AND CYP26B INHIBITORS
BACKGROUND OF THE INVENTION
FIELD OF THE INVENTION This invention relates generally to the treatment of disorders sensitive to retinoids and, in particular, to the therapeutic use of compounds that selectively inhibit CYP26A (P450RAI-1) or CYP26B (P450RAI-2). Retinoids, such as retinoic acid ("RA"), are important modulators of division and differentiation, immune response and embryonic development. These molecules work in cells binding and activating the retinoic acid (RAR) receptors, which successively bind to certain DNA sequences and regulate the target gene expression. Retinoid drugs can be used to beneficially modulate the expression of certain target genes by improving or avoiding disease conditions. For this reason, more than 30 analogs of natural and synthetic origin of retinoic acid have been developed for their use as therapeutic agents. Current therapies with retinoids include the differentiation of acute promyelocytic leukemia (APL); the treatment of nodulocystic acne, a severe form of inflammatory acne; the treatment of psoriasis; the prevention of secondary head and neck cancers; topical therapy of acne vulgaris; and reversal of photolesion as measured by UV / Thacher et al., Current Pharm. Design 6:25:58 (2000)). Unfortunately, retinoids can be highly toxic at therapeutic doses. Therefore, the dosage of retinoids that can be administered to patients is limited by the significant side effects, which include irritation and inflammation of the skin and mucous membranes, elevation of serum triglycerides, dysregulation of bone formation and resorption, headaches, hypothyroidism, and fetal malformation. Another disadvantage of retinoid therapy is that patients, particularly cancer patients, often become resistant to therapy over time. Thus, there is a need to identify new methods to therapeutically increase or maintain the beneficial levels of retinoic acid and other retinoids in the body to treat the disease. The present invention satisfies this need and also provides the advantages described.
SUMMARY OF THE INVENTION The invention provides methods for treating an individual who has a retinoid responsive disorder. In one embodiment, a method involves administering to the individual an effective amount of an inhibitor of CYP26B (P450RAI-2), the selective CYP26B inhibitor having a selectivity of at least 10-fold for CYP26B relative to CYP26A. A selective CYP26B inhibitor used in a method of the invention can have a selectivity of at least 20 times for CYP26B relative to CYP26A (P450RAI-1), for example, a selectivity of at least 100 times, at least 500 times and at at least 1,000 times for CYP26B in relation to CYP26A. Exemplary selective CYP26B inhibitors which can be used in a method of the invention are shown below in the form of Formulas 5, 15 and 30 to 32. In another embodiment, a method for treating an individual having a sensitive disorder the retinoids involve administering to the individual an effective amount of a selective CYP26A inhibitor, the selective CYP26A inhibitor having a selectivity of at least 10-fold for CYP26A relative to CYP26B and having a formula selected from Formulas 1 to 4,6. to 14, 16, 17 and 18 to 29, as shown here below. A selective CYP26A inhibitor used in a method of the invention can have a selectivity of at least 20 times for CYP26A relative to CYP26B, for example a selectivity of at least 100 times, at least 500 times and at least 1, 000 times for CYP26A in relation to CYP26B. Exemplary retinoid responsive disorders that can be treated using a method of the invention include a skin disorder, such as acne, an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and an eye disorder. pulmonary disorder. In different embodiments, a selective CYP26A inhibitor or a selective CYP26B inhibitor can be administered, for example, peripherally or orally. In one embodiment, a method of the invention can be used to treat a human having a retinoid sensitive disorder. The invention further provides a screening method for identifying a selective CYP26A inhibitor having a selectivity of at least 10-fold for CYP26A relative to CYP26B. The method involves connecting a CYP26A with one or more candidate compounds; selecting from the candidate compounds a compound that inhibits CYP26A activity; determining the ability of the compound selected to inhibit CYP26B: activity and to identify a compound having a selectivity of at least 10-fold for CYP26A relative to CYP26B. The invention also provides screening for identifying a selective CYP26B inhibitor having a selectivity of at least 10-fold for CYP26B relative to CYP26A.
BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 shows that 13-cis retinoic acid is effective in reducing the differentiation of the sebaceous gland into the hamster's side organ. Fig. 2 shows that the activity of 13-cis retinoic acid on differentiation of the sebaceous gland can be blocked by a retinoic acid receptor antagonist. Fig. 3 shows that a synthetic RAR pan-antagonist (TTMBB) is effective in reducing the differentiation of the sebaceous gland into the hamster's side organ. Fig. 4 shows that both the selective CYP26A inhibitor and the selective CYP26B are effective in reducing the differentiation of the sebaceous gland into the hamster's side organ.
DETAILED DESCRIPTION OF THE INVENTION Retinoids play important roles in the regulation of gene expression during embryonic development and in the maintenance of adult epithelial tissues. The amount of a retinoid, such as retinoic acid (RA), present in the body at any given time is regulated, in part, by cellular metabolism. Cytochrome P450 molecules CYP26A and CYP26B, also known as P450RAI-1 and P450RAI-2, respectively, metabolize RA into more polarized hydroxylated and oxidized derivatives, thereby reducing the level of this retinoid in the body. By blocking the RA shredding activity of CYP26A or CYP26B, the amount of natural RA or administered in a cell can be maintained at a beneficial level. Accordingly, an inhibitor of CYP26A or CYP26B can be used to maintain or beneficially increase a retinoid level in an individual, either alone or in conjunction with a retinoid treatment. It has been observed that the administration of certain CYP26A inhibitors to mammals causes a significant increase in the levels of endogenous retinoic acid
(see, for example, U.S. Patent Nos.
6. 531,599 and 6,495,552). For example, the treatment of human patients with the CYP26A inhibitor liarozole produces beneficial effects similar to those observed after treatment with retinoids, such as the improvement of psoriasis (Kuijpers, et al., Bri tish Journa.1 of Dermatology 139: 380-389 (1998)). Below, selective inhibitors of CYP26A having a selectivity of at least 10-fold for CYP26A relative to CYP26B have been identified and are referenced as Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. In Table 4 A variety of selective CYP26A inhibitors and selective CYP26B inhibitors are shown below. The selective CYP26A inhibitors shown in Table 4 have a selectivity ranging from at least 10-fold to at least 888-fold selectivity for CYP26A relative to CYP26B. The selective CYP26B inhibitors shown in Table 4 have a selectivity ranging from at least 10-fold to a selectivity at least 83-fold for CYP26B relative to CYP26A. While the selective CYP26A inhibitors described herein are not structurally related to the above-described CYP26A inhibitors, these compounds can provide similar beneficial effects by reducing the destruction of endogenous or administered retinoids, including, without limitation, retinoic acid and acid analogs. retinoic Based on the identification of the selective inhibitors of CYP26A and CYP26B, the present invention provides therapeutic methods involving the selective inhibition of CYP26A or CYP26B. The methods are useful for beneficially treating an individual having any of a variety of retinoid sensitive disorders described herein below or otherwise known in the art. As described herein in Example I, compounds that selectively inhibit the activity of CYP26A or CYP26B were shown to be effective in reducing sebocyte differentiation in an animal model of acne development. In this animal model, which involves the observation of the differentiation of the sebaceous gland in the organ of the hamster side, retinoid receptor and retinoic acid agonists block differentiation, which is analogous to blocking the development of acne in a mammal such as a human. On the other hand, treatment with a selective CYP26A inhibitor or a selective CYP26B inhibitor reduced sebocyte differentiation as effectively as ACCUTANE, one of the most widely used commercially available acne medications with commercially available retinoic acid. Thus, in one embodiment, the present invention provides a method for treating a retinoid responsive disorder by administering an effective amount of a selective CYP26A inhibitor having any of Formulas 1 through 4, 6 through 14, 16, 17 and 18 a 29 or a selective CYP26B inhibitor having a selectivity of at least 10-fold for CYP26B relative to CYP26A. As used herein, the term "retinoid responsive disorder" means a condition or disease that normally has a symptom that is improved, alleviated, delayed at onset, or prevented following the administration of retinoic acid (RA) or a retinoid synthetic that has RA activity. A variety of disorders responsive to retinoids are known in the art and include, without limitation, cancers such as skin cancer, mouth cancer and Kaposi's sarcoma.; skin disorders such as acne, psoriasis and eczema; and other multiple disorders of diverse etiology, including but not limited to emphysema and Alzheimer's disease. The ability of the RA or a synthetic retinoid having the activity of the RA to improve, alleviate, delay the onset of, or prevent at least one symptom of a condition or disease, if not known, can be determined using well-known methods, including those described here below. A synthetic retinoid used for such a determination can be any of a variety of retinoids used experimentally or clinically. Among the exemplary synthetic retinoids in clinical use today are acitretin, isotretinoin, tretinoin, tazarotene, and adapalene. A variety of retinoid sensitive disorders can be treated according to a method of the invention. Such skin disorders include, without limitation, inflammatory and noninflammatory acne, psoriasis, eczema, atopic dermatitis, pityriasis rubra pilaris, multiple-cell carcinomas-actinic keratosis, arsenic keratosis, ichthyosis, and other disorders of the skin. keratinization and cutaneous hyperproliferatives, Darriers disease, lichen planus, glucocorticoid injury (steroid atrophy), cutaneous microbial affection, excessive skin pigmentation, and cutaneous photo-deterioration. In various embodiments, the invention provides methods for treating acne that involve the administration of a selective CYP26A inhibitor or a selective CYP26B inhibitor. Acne is a common disease of the pilosebaceous glands, and is characterized by comedones, papules, pustules, inflamed nodules, cysts filled with superficial pus, and (in extreme cases) sacs that are channeled and deep, inflamed, sometimes purulent. In the art, multiple categories or types of acne have been defined based on the severity of the condition. For example, "superficial acne" is characterized by black spots (open comedones) or white spots (closed comedones), inflamed papules, pustules, and superficial cysts, occasionally producing large cysts. As another example, "deep acne" is similarly characterized except with deep inflamed nodules and cysts filled with pus, which often rupture and become abscesses. Other acne categories include "acne vulgaris", which is the most common form of acne; the "acne conglobata," which is an acne that covers the back, chest, and buttocks with pustules that often connect under the skin; "acne fulminans", which is an extreme form of acne conglobata that involves the sudden outbreak of pustules and nodules, infected nodules, fever, joint pain, and possible weight loss or appetite; "Medicated acne", which is caused by a drug; "comedonal acne", which is acne characterized by blackheads and whiteheads without other forms of skin lesions, and "cystic acne," which occurs when the infected content of a pustule or pimple bursts beneath the skin, instead of on the surface. It is understood that the method of the invention can be used to beneficially treat an individual having any form of mild, moderate or severe acne including, but not limited to, those discussed above. In a further embodiment, the invention provides a method for treating an individual having a proliferative disorder which involves administering to the individual an effective amount of a selective CYP26B inhibitor having a selectivity of at least 10 times for CYP26B relative to CYP26A, or an effective amount of a CYP26A inhibitor, which is represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. As used herein, the term "proliferative disorder" means a disease or abnormal condition. that produces unwanted or anomalous cell growth, viability or proliferation. Among the proliferative disorders include diseases such as cancer, in which the cells are transformed neoplastic, and diseases resulting from the overgrowth of normal cells. For example, cellular proliferative disorders include diseases related to connective tissue overgrowth, such as various fibrotic diseases, including scleroderma, arthritis, alcoholic liver cirrhosis, keloid, and hypertrophic scarring.; vascular proliferative disorders, such as atherosclerosis; benign tumors, and the abnormal proliferation of cells that mediate autoimmune diseases. As used herein, the term "cancer" means a class of diseases characterized by uncontrolled growth of aberrant cells, including all known cancers, and neoplastic conditions, whether characterized as malignant, benign, soft tissue or solid tumor. Specific cancers that can be treated using the method of the invention include, without limitation, cancers of the skin, breast, eye, prostate, cervix, uterus, colon, bladder, esophagus, stomach, lung, larynx, oral cavity, blood and lymphatic system, metaplasias, dysplasias, neoplasias, leucoplasias, papillomas of the mucous membranes and Kaposi's sarcoma. A compound that maintains or beneficially increases the level of a retinoid in the body can be used by selectively inhibiting CYP26A or CYP26B to treat a variety of cancers. For example, the increase in the amount of retinoid in the body of cancer patients has been a successful strategy to treat a variety of cancers. For example, retinoids have been used to reduce or prevent cancers of the mouth, skin, and head and neck in individuals at risk of these tumors (see, for example, Bollag et al., Ann. Oncol. 3: 513-526 ( 1992), Chiesa et al., Eur. J. Cancer B. Oral Oncol. 28: 97-102 (1992), Costa et al., Cancer Res. 54: Supp. 7, 2032-2037 (1994)). Retinoids have also been used to treat squamous cell carcinoma of the cervix and skin (see, for example, Verma, Cancer Res. 47: 5097-5101 (1987); Lippman et al., "Nati Cancer Inst. : 235-241 (1992), Lippman et al., J. Nati Cancer Inst. 84: 241-245 (1992)) and Kaposi's sarcoma (see, for example, Bonhomme, et al., Ann. Oncol. : 234-235 (1991)), and have found significant use in the therapy of acute promyelocytic leukemia (see, for example, Huang et al., Blood 72: 567-572 (1988); Castaigne et al., Blood 76: 1704-1709 (1990), Lo Coco et al., Blood 77: 1657-1659 (1991), Warrell et al., N. Engl. J. Med 324: 1385-1393 (1991), and Chomienne et al. ., FASEB J. 10:
1025-1030 (1996)). Accordingly, a compound which maintains or beneficially increases the level of a retinoid in the organism can be used by selective inhibition of CYP26A or CYP26B to treat a variety of cancers using a method of the invention.
In another embodiment, the invention provides a method for treating an individual having a neurological disorder responsive to retinoids which involves administering to an individual an effective amount of a selective CYP26B inhibitor having a selectivity of at least 10 times for CYP26B relative to CYP26A, or an effective amount of a selective CYP26A inhibitor represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. As a selective CYP26A inhibitor or a selective CYP26B inhibitor can beneficially maintain or increase the level of a retinoid in a tissue of the nervous system of an individual, such an inhibitor can be used to treat a variety of neurological disorders that respond beneficially to retinoids. Both CYP26A and CYP26B are highly expressed in the brain, and multiple lines of evidence indicate that retinoids such as RA play important roles in normal neurological function and in neurological diseases. For example, retinaldehyde dehydrogenase, the enzyme that forms retinoic acid from retinaldehyde, has 1.5 to 2 times higher activity in the hippocampus and parietal cortex of brains with Alzheimer's disease than in normal controls.
(Conner and Sidell, Mol Chem. Neuropathol 30 (3): 239-52
(1997)). In addition, it has been indicated that retinoid hypofunction and impaired retinoid transport are factors that contribute to Alzheimer's disease (Goodman and Pardee, Proc. Nati, Acad. Sci. USA 4; 100 (5): 2901 -5 (2003)). Exemplary retinoid sensitive neurological disorders that can be treated with a selective CYP26A inhibitor or a selective CYP26B inhibitor according to a method of the invention include, but are not limited to, Alzheimer's Disease, schizophrenia, disease of Parkinson's disease, anxiety, depression, drug addiction, cognitive disorders, emesis, eating disorders, attention deficit hyperactivity disorder, Tourette's syndrome, Huntington's disease, tardive dyskinesia , Lesch-Nyhan disease, Rett syndrome and any neurological disorder that is sensitive to retinoids. In a further embodiment, the invention provides a method for treating an individual having an inflammatory disorder or autoimmune disorder responsive to retinoids or by administering to the individual an effective amount of a selective CYP26B inhibitor having a selectivity of at least 10 times for CYP26B relative to CYP26A, or an effective amount of a selective CYP26A inhibitor represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. A method of the invention can be used to treat any of a variety of inflammatory disorders, including, without limitation, those resulting from injuries; infection by a bacterium, virus, fungus or other pathogen; autoimmune disorders; and other anomalous conditions. Examples of inflammatory disorders responsive to exemplary retinoids that can be treated using a method of the invention include, for example, without limitation, inflammatory skin disorders, e.g., psoriasis; inflammatory gastrointestinal disorders, for example, ileitis, irritable bowel syndrome, ulcerative colitis and Crohn's disease; autoimmune disorders such as rheumatoid arthritis and other forms of arthritis; the rejection of organ transplantation; and any other inflammatory or autoimmune disorder responsive to retinoids. In another embodiment, the invention provides a method for treating an individual having an ocular disorder responsive to retinoids which involves administering to the individual an effective amount of a selective CYP26B inhibitor having a selectivity of at least 10 times for CYP26B relative to CYP26A, or an effective amount of a selective CYP26A inhibitor represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. A method of the invention can be used to treat a variety of ocular disorders, including , without limitation, diabetic retinopathy; macular edema such as macular edema associated with diabetes mellitus or other conditions; degeneration of the retina such as macular degeneration related to age or retinitis pigmentosa; inflammatory disorders of the retina; vascular occlusive conditions of the retina such as retinal vein occlusions or occlusions of the secondary or central retinal artery; retinopathy of premature babies; retinopathy associated with blood disorders such as sickle cell anemia; the lesion that follows the retinal detachment; the injury or attack due to vitrectomy surgery or retinal surgery; and other retinal lesions including therapeutic lesions such as those resulting from laser treatment of the retina, for example, pan-retinal photocoagulation for diabetic retinopathy or photodynamic therapy of the retina, for example, for age-related macular degeneration.; genetic and acquired optic neuropathies such as optic neuropathies characterized primarily by the loss of central vision, for example, hereditary optic neuropathy of
Leber (LHON), autosomal dominant optic atrophy
(Kjer's disease) and other optic neuropathies such as those involving mitochondrial effects, aberrant dynamin-related proteins or inappropriate apoptosis, proliferative vitreoretinopathy (PVR), retinal detachment, dry eye, as well as any ocular disorder that is sensitive to retinoids. Referring to ocular disorders see, for example, Carelli et al., Neurochem. Intl. 40: 573-584 (202); and Olichon et al. , J. Biol. Chem. 278: 7743-7746 (2003). In still another embodiment, the invention provides a method for treating an individual having a pulmonary disorder involving the administration to the individual of an effective amount of a selective CYP26B inhibitor having a selectivity of at least 10-fold for CYP26B relative to CYP26A, or an effective amount of a selective CYP26A inhibitor represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. Retinoid treatment has been used to successfully treat, for example, emphysema , which is a pulmonary disorder resulting from the progressive destruction of the alveolar septa that was considered irreversible until it was shown that the administration of retinoic acid can reverse the anatomical and physiological signs of emphysema in a rat model (Massaro and Massaro, Nature Medicine 3: 675-7 (1997)). A method of the invention can be used to treat emphysema or another pulmonary disorder that is responsive to retinoids. Non-limiting examples of pulmonary disorders include, but are not limited to, obstructive lung disorders such as emphysema, chronic bronchitis, bronchial asthma, bronchiectasis, and cystic fibrosis; and restrictive pulmonary disorders such as interstitial fibrosis, pulmonary edema, respiratory fatigue syndrome in adults, rheumatoid spondylitis and pleural effusion. It is understood that increasing the level of a retinoid in a tissue of an individual may have beneficial effects in individuals having a variety of other retinoid responsive disorders, including cardiovascular disorders such as, without limitation, diseases associated with metabolism of lipids including dyslipidemias and post-angioplasty restenosis; and diabetes. In that case, the methods may be useful for treating a variety of retinoid sensitive disorders including, but not limited to, skin disorders, autoimmune disorders, inflammatory disorders, proliferative disorders, neurological disorders, eye disorders and pulmonary disorders.
By specifically mentioning the above categories of retinoid sensitive disorders, those skilled in the art will understand that all cases and types of these disorders are included in such terms. For example, it is desired that the term "skin disorder" includes any skin disorder having a symptom that is improved, alleviated, delayed in its onset or prevented after administration of the RA or a synthetic retinoid having the activity of the RA. Accordingly, the methods of the invention are applicable to disorders responsive to known retinoids as well as to disorders that have been determined to be responsive to retinoids, for example, in an animal model corresponding to a particular disorder. As described here below, CYP26A and CYP26B have different expression levels in different tissues in humans and other mammals. Thus, a selective CYP26A inhibitor or a selective CYP26B inhibitor can be used to inhibit the activity of a particular CYP26, such as CYP26A, for example, in a selected tissue without substantially altering CYP26B activity in that tissue, and vice versa. A selective CYP26A inhibitor or a selective CYP26B inhibitor can also be used to direct the inhibition of a CYP26 in a particular tissue, if desired. It is also understood that a selective CYP26A inhibitor or a selective CYP26B inhibitor can be used without targeting specific proteins for the inhibition of CYP26. In humans, CYP26A expression has been observed in the liver, brain and placenta (Ray et al J. Biol. Chem. 272: 18702-18708 (1997)); the expression of CYP26B has been observed in the brain (White et al Proc. Nati.
Acad. Sci. USA 97: 6403-6408 (2000) as well as in the kidney, the lung, the spleen, the fetal spleen, the skeletal muscle, the thymus, the peripheral blood leukocytes, the lymph nodes, the bones, the stomach, the placenta , the duodenum, the small intestine, and the pituitary gland (PCT / CA00 / 01493). It is understood that either CYP26A and CYP26B or both may be present in other tissues in humans or other mammals. For example, as shown in Example 1, both CYP26A and CYP26B are expressed in hamster skin. It is understood that a selective CYP26A or selective CYP26B inhibitor can be used to treat a retinoid sensitive disorder associated with a tissue in which either CYP26A or CYP26B is expressed. For example, an individual having cancer in a tissue in which CYP26A is expressed can be treated using a selective CYP26A inhibitor, while an individual having cancer in a tissue in which CYP26B is expressed can be treated using an inhibitor of CYP26A. Selective CYP26B. Thus, the expression of CYP26A or CYP26B in a particular tissue can be used to evaluate whether a treatment of a particular retinoid sensitive disorder with a selective CYP26A inhibitor or selective CYP26B inhibitor is appropriate. It is understood that a selective CYP26A or selective CYP26B inhibitor may be useful for treating a retinoid sensitive disorder even when the target tissue contains a low level of expression of both CYP26A and CYP26B. Methods for determining the expression levels of CYP26A or CYP26B mRNA or protein are well known to those skilled in the art and are described, for example, by Sambrook et al., In Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory , New York (1992) and by Ausubel et al., In Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, MD (1998).
Selective CYP26A and CYP26B inhibitors
A. Functional characteristics
The methods of the invention involve the administration of a selective CYP26A inhibitor or of selective CYP26B. As used herein, the term "selective CYP26A inhibitor" means a compound that reduces the expression or activity of CYP26A at least 10 times more than any reduction made by the inhibitor on the expression or activity of CYP26B. A selective CYP26A inhibitor can have, for example, a selectivity of at least 10 times for CYP26A relative to CYP26B, a selectivity of at least 20 times, at least 40 times, at least 80 times, at least 100 times, at least 500 times, at least 800 times, or at least 1,000 times for CYP26A in relation to CYP26B. As used herein, a "CYP26B inhibitor" means that it reduces the expression or activity of CYP26B at least 10 times more than any reduction made by the inhibitor on the expression or activity of CYP26A. A selective CYP26B inhibitor can have, for example, a selectivity of at least 10 times for CYP26A relative to CYP26B, a selectivity of at least 20 times, at least 40 times, at least 80 times, at least 100 times, at least 500 times, at least 800 times, or at least 1,000 times for CYP26B in relation to CYP26A. Exemplary small molecule selective CYP26A inhibitors are described herein as Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29; the exemplary small molecule selective CYP26B inhibitors are described herein as Formulas 5, 15 and 30 to 32. A selective CYP26A or selective CYP26B inhibitor may also be, for example, a protein, a peptide, a peptidomimetic, a ribozyme , a nucleic acid molecule or oligonucleotide, an oligosaccharide, or a small molecule or combination thereof. A selective CYP26A inhibitor or a selective CYP26B inhibitor useful in a method of the invention can act by any mechanism, and can, for example, alter the catalytic action of the specified enzyme and consequently reduce, or in some cases, stop the catalysis. A selective CYP26A inhibitor or a selective CYP26B inhibitor may therefore be a competitive, uncompetitive, or concurrent inhibitor of CYP26A or CYP26B, respectively, and may additionally function in a reversible or irreversible manner. It is understood that a selective CYP26A inhibitor or a selective CYP26B inhibitor can also act indirectly, for example by reducing or down-regulating the expression of CYP26A or CYP26B mRNA or protein, respectively. A selective CYP26A or selective CYP26B inhibitor useful in a method of the invention does not bind to a retinoic acid receptor (RAR), or subunit thereof.
B. Structural characteristics
As described herein, a variety of structurally unrelated compounds that have a selectivity of at least 10-fold for CYP26A relative to CYP26B may be selective CYP26A inhibitors; and a variety of structurally unrelated compounds that have a selectivity of at least 10-fold for CYP26B relative to CYP26A, may be selective CYP26B inhibitors and therefore may be useful in a method of the invention. A selective CYP26A inhibitor can be, for example, an organic molecule represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29, or a salt, ester, amide, stereoisomer or racemic mixture of the pharmaceutically acceptable A selective CYP26B can be, for example, an organic molecule represented by any of Formulas 5, 15 and 30 to 32, below, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof. As described herein in Table 4, the identified CYP26A inhibitors have selectivities of about 10 times, about 40 times, about 60 times, about 80 times, about 200 times, about 300 times, and about 900 times; the identified CYP26B inhibitors have selectivities of about 10 times, for example about 25 times, about 50 times and about 80 times. It is understood that these and other selective CYP26A inhibitors and selective CYP26B inhibitors may be useful for treating a retinoid responsive disorder according to a method of the invention. The compounds useful in the methods of the invention are shown in two sections below. In the first section, Formulas 18 to 32 are described, which are encompassed by the generic Formulas 1 to 17. In the second section, Formulas 1 to 17 are described.
I. Formulas 18 to 32
Formula 18:
where,
R3 = H or lower alkyl; Y
R1 = R2 = H, Me or Et; X = O or S; Y = CH2N (Me) (cyc-Pr), H, halo, OH, lower alkoxy, lower alkyl, haloalkyl, cycloalkyl, alkenyl, or alkynyl; or
R? = Me; R2 = H; X = H (C) CH2N (Me) (cyc-Pr);, Y = H, halo, lower alkoxy, lower alkyl, cycloalkyl or haloalkyl; or
R? = Me, R2 = H; X = C = 0, Y = H, lower alkyl, haloalkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl.
Formula 19:
where,
R3 = H or lower alkyl; and Rx = R2 = H, Me or Et; X = O or S; and Y = CH2N (Me) (cyc-Pr), H, halo, OH, lower alkoxy, lower alkyl, haloalkyl, cycloalkyl, alkenyl, or alkynyl; or
R3 = H or lower alkyl; and R? = Me; R2 = H; X = H (C) CH2N (Me) (cyc-Pr); and Y = H, halo, lower alkoxy, lower alkyl, cycloalkyl or haloalkyl; or
R3 = H or lower alkyl; and Rx = Me; R2 = H; X = C = 0; and Y = H, lower alkyl, haloalkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl.
Formula 20
where, R? = R2 = Me; X = O or S; and Y = CH2N (Me) (cyc-Pr), OR or COOR, where R = lower alkyl or cycloalkyl; or
Rx = Me; 2 = H; X = H (C) CH2N (Me) (cyc-Pr); and Y = OR, COOR where R = lower alkyl, or cycloalkyl; or
R? = Me; R2 = H; X = C = 0; and Y = alkenyl.
Formula 21:
where, R? = R2 = Me; X = O or S; and Y = lower alkyl, haloalkyl, cycloalkyl, CH2N (Me) (cyc-Pr), OR, COOR, where R = lower alkyl, cycloalkyl; or
R? = Me; R2 = H; X = H (C) CH2N (Me) (cyc-Pr); and Y = lower alkyl, haloalkyl, cycloalkyl, OR, COOR, where R = lower alkyl, or cycloalkyl; or
R? = Me; R2 = H; ? = C = 0; and Y = alkenyl, lower alkyl, or cycloalkyl.
Formula 22
where, R3 = lower alkyl; Y
R1 = R2 = H or Me; or
R? = Me; and R2 = H.
Formula 23:
where,
R3 = lower alkyl; Y
R? = H and R2 = H; and Y = alkenyl, or alkynyl; or
R? = Me and R2 = H or Me; and Y = lower alkyl, alkenyl, or alkynyl; or
R? = H or Me and R2 = Me; and Y = lower alkyl, alkenyl, or alkynyl.
Formula 24
where,
R3 = H or lower alkyl; Y
R? = H, Me or Et; X = 0 or S; and Y = CH2N (Me) (cyc-Pr), H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl, or alkynyl; or
R? = Me; X = H (C) CH2N (Me) (cyc-Pr); and Y = H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl or alkynyl; or
Ri = Me; X = c = 0; and Y = H, lower alkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl.
Formula 25
where,
R3 = H or lower alkyl; Y
Rx = H, Me or Et; X = O or S; and Y = CH2N (Me) (cyc-Pr), H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl or alkynyl; or
R? = Me; X = H (C) CH2N (Me) (cyc-Pr); and Y = H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl or alkynyl; or
R? = Me; ? = c = O; and Y = H, lower alkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl.
Formula 26
where, Rx = H, Me or Et; X = O or S; and Y = CH2N (Me) (cyc-Pr), OR, or COOR, where R = lower alkyl or cycloalkyl; or
R? = Me, X = H (C) CH2N (Me) (cyc-Pr); and Y = alkenyl, OR, COOR, where R = lower alkyl or cycloalkyl; or
R? = Me; X = C = 0; and Y = alkenyl.
Formula 27:
where, Z = H, Me or Cl; Y
R? = H, Me or Et; X = O or S; and Y = lower alkyl, cycloalkyl, CH2N (Me) (cyc-Pr), OR, or COOR, where R lower alkyl or cycloalkyl; or
R? = Me, X = H (C) CH2N (Me) (cyc-Pr); and Y = lower alkyl, cycloalkyl, OR, or COOR, where R = lower alkyl, or cycloalkyl; or
R? = Me; ? = c = 0; and Y = alkenyl, lower alkyl, or cycloalkyl
Formula 28
where, R = lower alkyl; and R? = H; and R2 = Me; or R1 = R2 = Me or H.
Formula 29;
where, X = COOR, C (CH3) 2COOR, CH2N (CH3) (cyc-Pr), where R = lower alkyl; Y = H, lower alkyl, haloalkyl, alkenyl or alkynyl; Y
Ri = H and R2 = Me; or R1 = R2 = Me or H.
A CYP26A inhibitor having a selectivity of at least 10-fold for CYP26B relative to CYP26A may have a chemical formula shown as any of Formulas 30 to 32 described below. Formula 30:
wherein, R = t-Butyl, CH2N (Me) (cyc-Pr), or N (Me) (cyc-Pr); and X = H or Me.
Formula 31
wherein, R = t-Butyl, CH2N (Me) (cyc-Pr), or N (Me) (cyc-Pr); and X = H or Me
Formula 32:
where, n = 0 or 1; Y
Ri = R2 = Me; X = O, S, where R = lower alkyl; and Y = cycloalkyl, alkenyl, alkynyl, lower alkoxy, halo, or haloalkyl; or
R? = Me; R2 = -CH2CH2-; X = O or S; and Y = cycloalkyl, alkenyl, alkynyl, lower alkoxy, halo, or haloalkyl; or
Ra = Me; R2 = H; X = -CHCH2N (Me) (cyc-Pr); and Y = H, lower alkyl, cycloalkyl, alkenyl, lower alkoxy, halo or haloalkyl; or
R? = Me; R2 = H; X = CMe2; and Y = H, lower alkyl, cycloalkyl, alkenyl, alkynyl, lower alkoxy, halo or haloalkyl. II. Formulas 1 to 17
Formula 1
wherein: A is a phenyl or naphthyl, or heteroaryl group selected from a group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; X is O, S or NR where R is H, alkyl of 1 to 6 carbons or benzyl; Y is H, alkyl of 1 to 10 carbons, benzyl, C 1 -C 6 alkyl or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with alkyl C? _C6, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, or I or alkoxy of 1 to 6 carbons; Z is -C = C-, - (CR? = CR?) N ', where n' is an integer having the value 1-5, -CO-NRi-, NRi-CO-; -CO-O-, -0-CO-, -CS-NR-, NRi-CS-, -CO-S-, -S-CO-, -N = N-; or -NRx-CO-NR? -; Ri is independently H or alkyl of 1 to 6 carbons; p is an integer that has the values of 0 to 4;
R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F,
Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 2; R4 is independently H, alkyl of 1 to 6 carbons, or F, alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 2; W is -C (R5) 2- or -CR5 = CR5-; R5 is independently H, halogen, or alkyl of 1 to
3 carbons with the proviso that when W is -C (R5) 2- a.1 minus one R5 is alkyl of 1 to 3 carbons, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (alkyl Ca-Ce ), CH2OCO (C? -C6 alkyl) or a pharmaceutically acceptable base cation.
Formula 2
Formula 2 where the dashed line represents a link or the absence of a link;
A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R2 groups; X is alkyl of 1 to 6 carbons, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl , Br, or I, OR, SR, NRR7, -CO-OR where R is H, alkyl of 1 to 6 carbons or benzyl; Y is H, alkyl of 1 to 10 carbons, benzyl, C 1 -C 6 alkyl or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with alkyl C? -C6, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I , COOR8 or alkoxy of 1 to 6 carbons; Z is -C = C-, - (CR? = CR?) N ', where n' is an integer having the value 1-5, -CO-NRi-, NRi-CO-; -CO-O-, -0-CO-, -CS-NRX-, NRi-CS-, -CO-S-, -S-CO-, -N = N-; or -NRx-CO-NR? -; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F,
Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 2; R4 is independently H, alkyl of 1 to 6 carbons, or alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 4; W is ~ C (R5) 2- or -CR5 = CR5-; R5 is independently H, halogen, or alkyl of 1 to 3 carbons with the proviso that when W is -C (R5) 2- at least one R5 is alkyl of 1 to 3 carbons; R7 is H, lower alkyl, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with lower alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C6-C6 alkyl), CH2OCO (alkyl) Ci-Cß) or a cation of a pharmaceutically acceptable base.
Formula 3
Formula 3 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; X is O, S or NR where R is H, alkyl of 1 to 6 carbons, trialkyl (C-C6) silyl or benzyl; Y is H, alkyl of 1 to 10 carbons, benzyl, alkyl
Cx-C6 or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with C? -C6 alkyl, alkenyl of 2 to 6 carbons and having 1? or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, or I; Z is -CE = C-, - (CR? = CR?) N '/ where n' is an integer that has the value 1-5, -CO-NRx-, NRi-CO-; -CO-O-, -O-CO-, -CS-NR-, NRi-CS-, -CO-S-, -S-CO-, -N = N-; or -NRx-CO-NRi-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; W is -C (R5) 2- or -CR5 = CR5-; R5 is independently H, halogen, or alkyl of 1 to 3 carbons with the proviso that when W is -C (R5) 2- at least one R5 is alkyl of 1 to 3 carbons;
R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C6 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C alkyl? -C6), CH2OCO (Ca-C6 alkyl) or a pharmaceutically acceptable base cation.
Formula 4
Formula 4 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; X is OR7, SR7 or NRR7 where R is H, alkyl of 1 to 6 carbons or benzyl; Y is H, alkyl of 1 to 10 carbons, benzyl, C 1 -C 6 alkyl, benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with alkyl C6-C6, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I , or -COORi;
Z is -G = C-, - (CR? = CR?) N ', where n1 is an integer having the value 1-5, -CO-NRi-, NRi-CO-; -C0-0-, -0-C0-, -CS-NRi-, NRi-CS-, -CO-S-, -S-CO-, -N = N-; or -NRi-CO-NRi-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; W is -C (R5) 2- or -CR5 = CR5-; R5 is independently H, halogen, or alkyl of 1 to 3 carbons with the proviso that when W is -C (RS) 2- at least one R5 is alkyl of 1 to 3 carbons; R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C3 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C alkyl? -Ce), CH2OCO (Ca-C6 alkyl) or a pharmaceutically acceptable base cation.
Formula 5 COORe
Formula 5 wherein: A is a phenyl or naphthyl group, or heteroaryl selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Y is H, alkyl of 1 to 10 carbons, benzyl, alkyl
C? -C6 or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with C? -C6 alkyl, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I, or -COORi; Z is -C = -C-, - (CR = CR?) N ', where n' is an integer having the value 1-5, -CO-NR-, NRx-C0-; -CO-O-, -O-CO-, -CS-NR-, NRi-CS-, -CO-S-, -S-CO-, -N = N-; or -NRi-CO-NRi-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons;
R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has values from 0 to 3, n is an integer that has the values of 0 or 1, p is an integer that has the values of 0 or 1, W is -C (R5) 2- or - CR5 = CR5-; R5 is independently H, halogen, or alkyl of 1 to 3 carbons with the proviso that when W is -C (R5) 2- at least one R5 is alkyl of 1 to 3 carbons, and R8 independently is H, alkyl of 1 at 6 carbons, -CH20 (C? -C6 alkyl), CH2OCO (C? _6 alkyl) or a pharmaceutically acceptable base cation.
Formula 6
Formula 6 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R groups;
X is O, S, NR or CO where R is H or alkyl of 1 to 6 carbons; Y is H, alkyl of 1 to 10 carbons, benzyl, C 1 -C 6 alkyl or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with alkyl C? -C6, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I , OR7, CH2-NRR7 or -COORi; Z is -C = C-, - (CR? = CR?) N ', where n1 is an integer having the value 1-5, -CO-NRx-, NRi-CO-; -CO-O-, -0-CO-, -CS-NR-, NRi-CS-, -CO-S-, -S-CO-, -N = N-; or -NRi-CO-NRi-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 4; W is -C (R5) 2- or -CR5 = CR5-;
R5 is independently H, halogen, or alkyl of 1 to 3 carbons with the proviso that when W is -C (R5) 2- at least one R5 is alkyl of 1 to 3 carbons, and R is H, alkyl of 1 to 6 carbons, 3 to 6 carbon cycloalkyl or 1 to 6 carbon cycloalkyl substituted with L-C6 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C? -C3 alkyl), CH2OCO (C? Alkyl? -C6) or a cation of a pharmaceutically acceptable base.
Formula 7
Formula 7 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Y is alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, OR7, CH2-NRR7 or -COORi; Z is -C = C-, -CO-0-, or -NR1-CO-NR1-; R is independently H or alkyl of 1 to 6 carbons;
Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R 4 is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 4; R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C3 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C alkyl? -C6), CH2OCO (C? -C3 alkyl) or a pharmaceutically acceptable base cation.
Formula 8
Formula 8 where;
A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R2 groups; Y is alkenyl of 2 to 6 carbons, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons; Z is -C = C-, -CO-O-, or - Ri-CO-NR-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer having the values of 0 to 3, - R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C6 alkyl, and R8 is H , alkyl of 1 to 6 carbons, -CH20 (C?-C6 alkyl), CH 2 OCO (L-CS alkyl) or a pharmaceutically acceptable base cation.
Formula 9 Formula 9 wherein A is a phenyl or naphthyl group, or heteroaryl selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Z is -C = C-, -CO-0-, or -NR1-CO-NR1-; R is H or alkyl of 1 to 6 carbons; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkyltium of 1 to 6 carbons or benzyl; m is an integer having the values of 0 to 3, - R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C6 alkyl, and R8 independently is H, alkyl of 1 to 6 carbons,
-CH20 (C? -C6 alkyl), CH2OCO (C? -C3 alkyl) or a pharmaceutically acceptable base cation.
Formula 10
Formula 10 where the broken line represents a link or the absence of a link; A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R2 groups; X is NRR7, or C00R8; Y is H, alkenyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, OR7 or -COORi; Z is -C = C-, -CO-O-, or -NRi-CO-NRi-; R is independently H or alkyl of 1 to 6 carbons; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons;
R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R 4 is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 4; R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C6 alkyl, and R8 independently is H, alkyl of 1 to 6 carbons, -CH20 (Ci alkyl) -Cg), CH2OCO (C? -C6 alkyl) or a pharmaceutically acceptable base cation.
Formula 11
Formula 11 wherein A is a phenyl or naphthyl group, or heteroaryl selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Y is, alkenyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, or alkynyl-alkenyl of 4 to 6 carbons; Z is -C = C-, -C0-0-, or -NRa-CO-NRi-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer having the values of 0 to 4, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C? -C6 alkyl), CH2OCO (C? -C6 alkyl) or a cation of a pharmaceutically base acceptable.
Formula 12
Formula 12 wherein A is a phenyl or naphthyl group, or heteroaryl selected from a group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazole and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Z is -C = C-, -CO-O-, or -NR1-CO-NR1-; R is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F,
Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer having the values of 0 to 3, and R8 independently is H, is alkyl of 1 to 6 carbons, -CH20 (C? -C6 alkyl), CH2OCO (C? -C6 alkyl) or a cation of a pharmaceutically acceptable base.
Formula 13
Formula 13
wherein A is a phenyl or naphthyl group, or heteroaryl selected from a group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Z is -C = C-, -C0-0-, or -NRi-CO-NRi-; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer having the values of 0 to 3, and R8 independently is H, alkyl of 1 to 6 carbons, -CH20 (C? -C6 alkyl), CH2OCO (C -C6 alkyl) or a pharmaceutically-based cation of a base acceptable
Formula 14 COOR8
Formula 14 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; Y is H, alkyl of 1 to 10 carbons, benzyl, alkyl
C? -C6 or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with C? -C6 alkyl, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I, OR7, CH2-NRR7 or -COORi; R is independently H or alkyl of 1 to 6 carbons; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons;
R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen, -o is an integer having the values of 0 to 4; R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C! -C6 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (alkyl) C6), CH2OCO (C? -C6 alkyl) or a pharmaceutically acceptable base cation.
Formula 15
Formula 15 where; is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R 2 groups;
X is O or S; Y is H, alkyl of 1 to 10 carbons, benzyl, Ci-Cg alkyl or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with alkyl C -C6, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I, 0R7, CH2-NRR7 or -COORi; Ri is independently H or alkyl of 1 to 6 carbons; p is an integer that has the values of 0 to 4; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R 4 is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 4; R is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C6 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C alkyl? -C6), CH2OCO (C? -C6 alkyl) or a pharmaceutically acceptable base cation.
Formula 16
Formula 16 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two groups R2; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons; R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer having the values of 0 to 3, and R8 independently is H, alkyl of 1 to 6 carbons, -CH20 (C? -C6 alkyl), CHOCO (C? -C6 alkyl) or a cation of a base pharmaceutically acceptable.
Formula 17 Formula 17 wherein: A is a phenyl or naphthyl, or heteroaryl group selected from the group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R2 groups; X is O or S; Y is H, alkyl of 1 to 10 carbons, benzyl, C? -6 alkyl or benzyl substituted with halogen, alkyl of 1 to 10 carbons substituted with fluorine, cycloalkyl of 3 to 6 carbons, cycloalkyl of 3 to 6 carbons substituted with alkyl C? -C3, alkenyl of 2 to 6 carbons and having 1 or 2 double bonds, alkynyl of 2 to 6 carbons, alkenyl-alkynyl of 4 to 6 carbons, alkynyl-alkenyl of 4 to 6 carbons, Cl, Br, I , OR7, CH2-NRR7 or -COOR ?; Ri is independently H or alkyl of 1 to 6 carbons; R 2 is independently H, alkyl of 1 to 6 carbons, F, Cl, Br, I, CF 3, alkyl substituted with fluorine of 1 to 6 carbons, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons;
R3 is independently alkyl of 1 to 6 carbons, F, Cl, Br, I, alkyl substituted with fluorine of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, alkylthio of 1 to 6 carbons or benzyl; m is an integer that has the values of 0 to 3; R 4 is independently H, alkyl of 1 to 6 carbons, or F; alkyl substituted with fluorine of 1 to 6 carbons, or halogen; or is an integer that has the values of 0 to 4; R7 is H, alkyl of 1 to 6 carbons, cycloalkyl of 3 to 6 carbons or cycloalkyl of 1 to 6 carbons substituted with C? -C6 alkyl, and R8 is H, alkyl of 1 to 6 carbons, -CH20 (C alkyl? -C6), CH2OCO (C? -C6 alkyl) or a pharmaceutically acceptable base cation.
The term alkyl refers to and covers each and every one of the groups that are known as normal alkyl and branched chain alkyl. Unless otherwise specified, lower alkyl means the defined broad definition before alkyl groups having from 1 to 6 carbons in the case of normal lower alkyl, and from 3 to 6 carbons for the lower branched chain alkyl groups. A pharmaceutically acceptable salt can be prepared for any compound used according to the invention having a functionality capable of forming a salt, for example an acid functionality. A pharmaceutically acceptable salt is any salt that retains the activity of the parent compound and does not confer any adverse or adverse effect on the subject to which it is administered and in the context in which it is administered.
The pharmaceutically acceptable salts can be derived from organic or inorganic bases. The salt can be a mono or polyvalent ion. Inorganic ions are sodium, potassium, calcium, and magnesium. Organic salts can be made with amines, namely ammonium salts such as mono-, di- and trialkylamines or ethanolamines. The salts can be formed with caffeine, tromethamine and similar molecules. When there is a sufficiently alkaline nitrogen to be capable of forming acid addition salts, these can be formed with any inorganic or organic acid or alkylating agent such as methyl iodide. Preferred salts are those formed with inorganic acids such as hydrochloric acid, sulfuric acid or phosphoric acid. Likewise, any number of simple organic acids such as mono-, di- or triacids can be used.
Some compounds used according to the present invention may have trans and cis (E and Z) isomers. Unless the specific orientation of the substituents in relation to a double bond or a ring is indicated in the name of the respective compound, and / or specifically showing in the structural formula the orientation of the substituents in relation to the double bond or ring the invention encompasses the trans isomers as well as cis.
Some of the compounds used according to the present invention may contain one or more chiral centers and therefore may exist in enatiomeric and diastereomeric forms. It is intended that the scope of the present invention encompass all isomers per se, as well as mixtures of cis and trans isomers, mixtures of diastereomers and also racemic mixtures of enantiomers (optical isomers).
The novel compounds used according to the invention are encompassed by the general formulas 1 to 17 given above. In each of these formulas a linker or a linking group designated Z covalently connects an aromatic or heteroaromatic radical designated A (R2) -W-COOR8, A (R2) -CH2-COOR8 or A (R2) -COOR8 and another radical cyclic which according to these formulas is a substituted phenyl radical, substituted tetrahydronaphthalene, substituted dihydronaphthalene, substituted chroman, substituted thiochroman or substituted tetrahydroquinoline. In general terms compounds such as XA (R2) -W-COOR8, X4-A (R2) -CH2-COOR8 and X4-A (R2) -COOR8 are commercially available, or can be made according to the chemical literature, or a modification of known chemical procedures, or chemical procedures described herein that are within the practical knowledge of the organic chemist. The group X4 represents a reactive group, which is suitable for coupling the compounds XA (R2) -W-COOR8, XA (R2) -CH2-COOR8 and X4-A (R2) -COOR8 to a derivative of the substituted phenylene radical, tetrahydronaphthalene substituted, substituted dihydronaphthalene, substituted chroman, substituted thiochroman, or substituted tetrahydroquinoline so that as a result of the coupling the linker or the linking radical is formed. In many cases the X4 group is a removable group such as halogen, or trifluoromethanesulfonyloxy, or a group capable of participating in a Wittig or Horner Emmons reaction. In some cases the group X is an ethynyl group capable of undergoing a coupling reaction with a leaving group (such as a halogen or a trifluoromethanesulfonyloxy group) attached to the substituted phenyl radical, substituted tetrahydronaphthalene, substituted dehydronaphthalene, substituted chroman, substituted thiochroman, or substituted tetrahydroquinoline. The group X may also represent an OH or NH2 group that forms an ester linker (COO) or amide (CONH), respectively, when reactions with an activated carboxyl derivative of the substituted phenyl radical, substituted tetrahydronaphthalene, substituted dihydronaphthalene, substituted chroman, substituted thiochroman , or substituted tetrahydroquinoline. Compounds of formulas X4-A (R2) -W-COOR8, X4-A (R2) -CH2-COOR8 and X4-A (R2) -COOR8 are generally referred to herein as "coupling reagents" or only "reactants" "and the preparation of various examples of these coupling reagents is described below in the specific examples. Additional examples are pyridyl, thienyl furyl, pyridazine, pyrazine and other heteroaryl analogs of the coupling reagents described in the specific examples. These reagents can be obtained according to the chemical literature, or modifications of known chemical methods, or of chemical methods described herein that are within the practical knowledge of the practicing organic chemist.
Still further, according to the general synthetic methodology for providing the compounds of Formulas 1 to 17, a substituted phenyl radical, substituted tetrahydronaphthalene, substituted dihydronaphthalene, substituted chroman, substituted thiochroman, or substituted tetrahydroquinoline having a covalently anchored group X5 is first synthesized.
The group X5 reacts with the group X4 of the reagents X4-A (R2) -W-COOR8, X4-A (R2) -CH2-COOR8 and X4-A (R2) -COOR8 to form the linker designated Z in the formulas The group X5 is that which is capable of participating in a coupling reaction, (such as an ethynyl group when X4 is a deletable group), or a deletable group (such as halogen or trifluoromethanesulfonyloxy when X4 is an ethynyl group) ), or an activated carboxylic acid functionality (when X4 is OH or NH2). The group X5 can also be an OH, SH or NH2 group when the group X4 is an activated carboxylic acid functionality. Specific examples of the intermediates substituted phenyl, substituted tetrahydronaphthalene, substituted dihydronaphthalene, substituted chroman, substituted thiochroman, or substituted tetrahydroquinoline having an X5 functionality are provided below, and are also available in the scientific and patent chemical literature. In general terms, for reactants and reactions that covalently link a substituted tetrahydronaphthalene intermediate, substituted dihydronaphthalene, substituted chroman, substituted thiochroman, or substituted tetrahydroquinoline with an aryl or substituted heteroaryl group, of Formulas A (R2) -W-COOR8, A (R2) -CH2-COOR8 and A (R2) COOR8 to form a compound including the linker designated Z, reference is made to U.S. Patent Nos. 5,648,503; 5,723,666, 5,952,345, 6,252,090 and 6,313,107 each of which memoir is hereby expressly incorporated by reference.
The substituted phenyl radical, substituted tetrahydronaphthalene, substituted dihydronaphthalene, substituted chroman, substituted thiochroman, or substituted tetrahydroquinoline of the novel compounds used according to the invention are transformed so as to include specific substituents (such as for example cycloalkyl substituents) encompassed in the scope of the invention, either before or after the radical A (R2) -W-COOR8, A (R2) -CH2-COOR8 or A (R2) -COOR8 has been anchored and the Z linker has formed, as it is illustrated by the specific examples described below. The radical W-COOR8, CH2-COOR8 or COOR8 of the compounds of Formulas 1 to 17 can be modified in order to obtain even more novel compounds. One such modification is the saponification of the compounds in which R8 is an alkyl group, CH20 (C? -C6 alkyl) or CH2OCO (L-C6 alkyl). Another modification is the esterification of the carboxylic acid functionality when the R8 group is H or a cation. Such saponification and esterification reactions are well known in the art and within the practical knowledge of the practicing organic chemist.
With reference to the symbol A in Formulas 1 to 17, the preferred novel compounds used according to the present invention are those in which A is phenyl, naphthyl, pyridyl, thienyl or furyl. Even more preferred compounds are those compounds wherein A is phenyl. As regards the substitutions of groups A
(phenyl) and A (pyridyl), compounds in which the phenyl group is substituted in the 1,4-position (para) and in which the pyridine ring is substituted in the 2,5-position are usually preferred. (The substitution in positions 2,5 in the nomenclature of "pyridine" correspond to the substitution in position 6 in the nomenclature of "nicotinic acid"). In the currently preferred novel compounds used according to the invention there is no substituent R2 in group A, or alternatively the substituent R2 is preferably a fluoro group which is preferably located in the aromatic carbon adjacent (ortho) to the carbon carrying the group W -COOR8, CH2-COOR8 or COOR8.
As regards the radical W-COOR8, the variable W preferably represents -CH = CH-, -CR5 = CH-, CH = CR5- (cinnamic acid derivatives) C (R5) 2 or CHR5 where R5 is preferably methyl . For the group R8, the groups H, lower alkyl of 1 to 3 carbons, -CH20 (alkyl) are preferred.
C? -C3) and -CH2OCO (CX-C3 alkyl), as well as the pharmaceutically acceptable salts of the free acids when R8 is H. Among the lower alkyl groups, -CH20 (CX-C3 alkyl) and -CH2OCO (C-alkyl) ? -C3), methyl, ethyl, CH2OCH3 and CH2OCOCH3 respectively are very preferred at present.
The linker group Z in all the novel compounds used according to the invention is preferably ethinyl,
(-CC-), ester (CO-O), or ureido (NHCONH). On the other hand for the chroman, thiochroman and tetrahydroquinoline derivatives the linker Z is preferably anchored to position 6 (e.g., see Formula 1). For the tetrahydronaphthalene and dihydronaphthalene derivatives the linker Z is preferably anchored to the 6-position as such positions are numbered in Formulas 2 and 11.
The group Ri is preferably methyl when it serves as a substituent attached to a carbon of the nucleus of chroman, thiochroman, tetrahydroquinoline, tetrahydronaphthalene or dihydronaphthalene and is preferably hydrogen when it forms part of the linker Z.
The aromatic portion of the chroman, thiochroman, tetrahydroquinoline, tetrahydronaphthalene or dihydronaphthalene nuclei of the compounds of the present invention is preferably either not substituted with a group R3 (the variable m is zero (0)), or R3 is alkyl or halogen The non-aromatic portion of the chroman, thiochroman, tetrahydroquinoline, tetrahydronaphthalene or dihydronaphthalene nuclei of the compounds of the present invention is preferably either unsubstituted with a group R4 (the variable is zero (0)), or (R4) or represents methyl groups, even more preferably gemomethyl dimethyl or gemyl diethyl groups attached to the 2-position of the chroman core.
The structures of the most preferred compounds of the invention are shown in Table 1. While most of the compounds shown in Table 1 are carboxylic acids, it should be understood that the Cx-C3 alkyl esters, the esters are also preferred. CH2OCH3 and CH2OCOCH3 and the pharmaceutically acceptable salts of these compounds.
Table 1
NA1 = Not Active; DA2 = Weakly Active
The compounds of the invention can be synthesized by applying the general synthetic methodology described above, and by similar modifications of the specific synthetic routes described below that will be readily apparent to chemists practicing synthetic organic chemistry in light of the description and in view of general knowledge available in the art. The specific reaction schemes described below are directed to the synthesis of exemplary and preferred compounds of the invention. While each of the specific and exemplary synthetic routes shown in these schemes can describe the specific compounds of the invention only within the scope of one or two of the general Formulas 1 to 17, the synthetic procedures and methods used herein are adaptable to the knowledge. practical of the practicing organic chemist and can be used with such an adaptation for the synthesis of the compounds of the invention that are not specifically described here as examples.
Analysis to confirm the activity
The ability of a compound to selectively inhibit CYP26A or CYP26B can be determined using any of a variety of assays. Such assays can be performed, for example, in a cell or tissue expressing endogenous or recombinant CYP26A or CYP26B, and generally involve the determination of the enzymatic activity of CYP26A or CYP26B or a downstream effect of the CYP26A or CYP26B enzyme activity before and after the application of a test compound. A downstream effect of the enzymatic activity of CYP26A or CYP26B may be, without limitation, a change in the level of a retinoid in a cell. Methods for measuring the activity of CYP26A or CYP26B are well known to those skilled in the art, and are described, for example, in U.S. Patent No. 6,495,552; WO 01/44443; White et al., Proc. Nati Acad. Sci. USA 97: 6403-6408 (2000); White et al., J. Biol. Chem. 272: 18538-18541 (1997), and in Examples I and II. One type of analysis useful for evaluating the activity of CYP26A or CYP26B involves determining the catabolism of retinoic acid to more polar derivatives including 4-hydroxyiretinic acid and 4-oxoretinoic acid (White et al., Supra 1997). A variety of cell types, including natural and genetically engineered cells, can be used in an in vitro assay to detect the activity of CYP26A or CYP26B or a downstream effect of the enzymatic activity of CYP26A or CYP26B. Natural cells expressing endogenous CYP26A or CYP26B include, for example, cells obtained from an organ that expresses CYP26A or CYP26B, as described above. Cell lines expressing CYP26A include, but are not limited to, HEK293, LC-T, SK-LC6, MCF, NB4, and U937 (White et al., Supra 1997). Cell lines expressing CYP26B include, but are not limited to, HPKla-ras, HeLa and MCF-7 (White et al., Supra 2000). Other natural cells and cell lines that express CYP26A or CYP26B can be identified by those skilled in the art using the methods described herein and other methods well known in the art. Cells for use in the assay of a compound for its ability to selectively inhibit CYP26A or CYP26B can be obtained from a mammal, such as a mouse, rat, pig, goat, primate or human, or a non-mammal. Cells that express CYP26A or CYP26B can be prepared using a variety of methods. Recombinant expression may be advantageous by providing a level of CYP26A or CYP26B expression superior to that found endogenously, and also allows expression in cells or extracts in which expression is not normally found. A recombinant nucleic acid expression construct generally contains a constitutive or inducible promoter of RNA transcription appropriate for the host cell or the transcription-translation system, operably linked to a nucleotide sequence encoding a polypeptide corresponding to CYP26A or CYP26B or an active fragment of it. The expression construct can be a DNA or an RNA, and optionally it can be contained in a vector, such as a plasmid or viral vector. The nucleotide and amino acid sequences of human CYP26A are available to one skilled in the art, for example, with GenBank Accession Number NM_000783 and NM_057157; the nucleotide and amino acid sequences of human CYP26B are available to one skilled in the art, for example, with Genbank Accession Number NM_019885. Other nucleotide and polypeptide sequences of CYP26A and CYP26B are available from GenBank, as are the otoplog sequences of CYP26A and CYP26B from rat, mouse and other species. Any of these nucleotide sequences of CYP26A or CYP26B can be used to recombinantly express CYP26A or CYP26B in an assay to confirm the activity of a selective CYP26A inhibitor or of selective CYP26B. One skilled in the art can recombinantly express desired levels of CYP26A or CYP26B using routine laboratory methods, described, for example, in the molecular biology technical manuals, such as Sambrook et al., Supra (1992) and Ausubel et al., supra (1998). Exemplary host cells that can be used to express recombinant CYP26A and / or CYP26B include isolated mammalian primary cells; established mammalian cell lines, such as COS, CHO, HeLa, NIH3T3, HEK 293-T and PC12; amphibian cells, such as Xenopus embryos and oocytes; and other vertebrate cells. Exemplary host cells also include insect cells such as Drosophila cells, yeast cells such as S. cerevisiae, S. pombe, or Pichia pastoris and prokaryotic cells such as E. coli.
An exemplary cell-based analysis to confirm the ability of a compound to selectively inhibit CYP26A or CYP26B is described in U.S. Patent No. 6,495,552. In summary, CYP26A or CYP26B can be stably transfected in HeLa cells. The cells that grow exponentially are harvested by incubation in trypsin. The cells are then washed and plated in a 48-well plate at 5 x 105 cells in 0.2 ml of MEM medium containing 10% FBS and 0.05 μCi RA- [H3] in the presence or absence of increasing concentrations of the test compounds. The compounds are diluted in 100% DMSO and then added to wells in triplicate to a final concentration of 10, 1 or 0.1 μM. As a positive control for the inhibition of RA metabolism, the cells are also incubated with ketoconazole at 100, 10 and 1 μM. The cells are incubated for 3 hours at 37 ° C. The retinoids are then extracted using the procedure of Bligh et al., Canadian Journal of Biochemistry 37: 911-917 (1959), modified using methylene chloride instead of chloroform. This publication by Bligh et al., Is specifically incorporated herein by reference. The water-soluble radioactivity is quantified using a scintillation counter, and the IC50 values are determined. An exemplary animal model analysis to confirm the ability of a compound to selectively inhibit CYP26A or CYP26B is also described in U.S. Patent No. 6,531,599. Topical application of a selective CYP26A inhibitor or a selective CYP26B inhibitor may result in an increase in endogenous levels of retinoic acid resulting in an irritation induced by retinoic acid in the skin of mice lacking hair. Thus, a hairless mouse model is an exemplary animal model that can be used to confirm that a selective CYP26A inhibitor or a selective CYP26B inhibitor has in vivo activity.
Confirmation of the efficacy of a selective CYP26A inhibitor or inhibitor of CYP26B The efficacy of a selective CYP26A inhibitor or CYP26B inhibitor, such as a compound represented by any of Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29, or a salt, ester, amide, stereoisomer or racemic mixture thereof, in the treatment of a retinoid sensitive disorder in an individual can be confirmed using any of a variety of well-known methods. For example, well-known animal models may be useful to confirm the efficacy of a selective CYP26A inhibitor or of a selective CYP26B inhibitor in the treatment of a retinoid responsive disorder such as a skin disorder, an inflammatory disorder, a autoimmune disorder, a neurological disorder, a proliferative disorder, an eye disorder or a pulmonary disorder. Animal models predictive of such disorders can be used to confirm the efficacy of treatment by measuring the experimental endpoints or appropriate clinical or physiological indicators, which will depend on the particular animal model selected. Those skilled in the art will know which animal models can be used to determine the efficacy or effective amount of a selective CYP26A inhibitor or a selective CYP26B inhibitor useful in the methods of the invention. An exemplary animal model to confirm the efficacy of a selective CYP26A inhibitor or a selective CYP26B inhibitor in the treatment of acne is described in Example 1. A variety of well-known animal models for acne can be used to confirm the efficacy of a selective CYP26A inhibitor or a selective CYP26B inhibitor. A variety of exemplary animal models of inflammatory or autoimmune disorders responsive to retinoids are well known to those skilled in the art. Some of these models are described, for example, in Progress in Inflamation Research, (.J. Parnham, Ed.) Birkh user Verlag, Basel, Switzerland (1998). Those skilled in the art will be able to select an appropriate animal model depending, in part, on the particular disorder to be treated using the method of the invention. An exemplary animal model to confirm the efficacy of a selective CYP26A inhibitor or a selective CYP26B inhibitor in the treatment of retinoid sensitive proliferative disorders generally involves the inoculation or implantation in a laboratory animal of heterologous tumor cells followed by the simultaneous or subsequent administration of a therapeutic treatment. The effectiveness of the treatment can be determined, for example, by measuring the degree of cell or tumor growth or tumor metastasis. The measurement of clinical or physiological indicators can be evaluated alternatively or additionally as an indicator of the effectiveness of the treatment. Exemplary animal tumor models are described, for example, in Brugge et al., Origins of Human Cancer. Cold Spring Harbor Laboratory Press, Plain View, New York, (1991). Among the animal models to confirm the efficacy of a selective CYP26A inhibitor or a selective CYP26B inhibitor in the treatment of a retinoid responsive neurological disorder include, for example, animal models of trauma due to stroke or neural injury that are known in the art. The technique. An experimental model of stroke involves occluding the right middle cerebral artery and both common rat carotid arteries for a short period, followed by reperfusion
(Moore et al., J. Neurochem. 80: 111-118 (2002)). An experimental model of CNS injury is the fluid percussion injury (FPI) model, in which a moderate impact (1.5-2.0 atm.) Is applied to the cerebral parietal cortex (Akasu et al., Neurosci. Lett 329: 305-308 (2002)). Experimental models of spinal cord injury are also used in the art (Scheifer et al., Neurosci, Lett 323: 117-120 (2002)). Suitable models of neural injury due to oxidative stress, hypoxia, radiation and toxins are also known in the art and are useful for confirming the efficacy of selective CYP26A inhibitor or a selective CYP26B inhibitor. A variety of animal models of eye disorders are also known to those skilled in the art. An exemplary animal model of retinal degeneration is described, for example, Lewis et al., Eye 16: 375-387
(2002). In a similar manner, a variety of animal models of lung disorders are known to those skilled in the art. An exemplary animal model of emphysema is described, for example, in Chen et al., J. Invest Surg 11: 129-137 (1998).
Tracing other selective CYP26A and CYP26B inhibitors
The selective CYP26A inhibitor or the selective CYP26B inhibitor used in the method of the invention may also be a small molecule of natural or unnatural origin, or a macromolecule of natural or non-natural origin such as a peptide, a peptidomimetic, an acid nucleic acid, a carbohydrate or a lipid. A selective CYP26A inhibitor or a selective CYP26A inhibitor may additionally be an antibody, or an antigen-binding fragment thereof such as a monoclonal antibody, a humanized antibody, a chimeric antibody, a minibodi, a bifunctional antibody, an antibody of single chain (scFv), a fragment of the variable region (Fv or Fd), Fab or F (ab) 2. A selective CYP26A or CYP26B inhibitor can also be a partially or fully synthetic derivative, an analog or mimetic of a macromolecule of natural origin, or a small organic or inorganic molecule. A variety of methods can be used to confirm that a selective CYP26 inhibitor has a 10-fold selectivity for CYP26B relative to CYP26A. Analyzes to determine the activity of CYP26A and CYP26B are described herein and are well known to those skilled in the art. Therefore, by screening for known or newly synthesized compounds for their ability to selectively inhibit CYP26B activity, a selective inhibitor of CYP26B can be identified and used to treat a retinoid responsive disorder according to a method of the invention.
Therapeutic administration
The methods of the invention involve administering a selective CYP26A inhibitor or a selective CYP26B inhibitor to an individual to treat a retinoid sensitive disorder. As used herein the term "treatment" represents the reduction, delay or prevention of the onset of one or more clinical symptoms, physiological indicators or biochemical markers of a retinoid sensitive disorder, or reduction of the need for concurrent therapy. . Clinical symptoms include perceptible, external or visible signs of the disease. Physiological indicators include the detection of the presence or absence of physical and chemical factors associated with a body process or function. Biochemical markers include those signs of disease that are observable at the molecular level, such as the presence of a marker of the disease, such as a tumor marker. A person skilled in the art will be able to recognize specific clinical symptoms, physiological indicators and biochemical markers associated with a particular retinoid responsive disorder. The term "treatment" encompasses any significant reduction in the symptoms of a retinoid sensitive disorder such as a reduction of at least 30%, 40%, 60%, 70%, 80%, 90% or 100%. The skilled clinician will be able to determine the clinical symptoms, physiological indicators or other appropriate biochemical markers associated with a particular retinoid responsive disorder, such as a skin disorder, a neurological disorder, an autoimmune disorder, an inflammatory disorder, a disorder ocular or a pulmonary disorder. The skilled clinician will know how to determine whether an individual is a candidate for treatment with a selective CYP26A inhibitor or a selective CYP26B inhibitor, based in part on the type and severity of the disorder, the degree of sensitivity of the disorder to retinoid therapy, the affected tissue, and the medical history and the individual's condition. The appropriate effective amount to be administered for a particular application of the methods can be determined by those skilled in the art, using the guidelines provided herein. For example, an effective amount can be extrapolated from in vitro and in vivo assays as described hereinbefore. One skilled in the art will recognize that the clinical symptoms, physiological indicators and biochemical markers of a disorder in an individual can be controlled throughout the course of therapy and that the effective amount of a selective CYP26A inhibitor or an inhibitor of Selective CYP26B that is administered can be adjusted accordingly. The invention may also be practiced by administering an effective amount of a selective CYP26A inhibitor or a selective CYP26B inhibitor with one or more other agents including, but not limited to, one or more retinoids. In such "combined" therapy, it is understood that a selective CYP26A inhibitor or a selective CYP26B inhibitor can be released independently or simultaneously, in the same or different pharmaceutical compositions, and by the same or different routes of administration as the other or other agents more. A selective CYP26A inhibitor or a selective CYP26B inhibitor can beneficially decrease the destruction of therapeutically administered retinoids and thereby allow a reduced amount of retinoids or a reduced frequency of retinoid administration to be administered.; and may increase the efficacy of a retinoid or prevent the development of retinoid resistance in individuals treated with a retinoid. Exemplary retinoids that may be useful in the combination therapy include, without limitation, tretinoin (all-trans-retinoic acid, vitamin A acid), alitretinoin (9-cis-retinoic acid), bexarotene (Tagretin), isotretinoin ( 13-cis-retinoic acid), and tazarotene.
Modifications and pharmaceutical formulations
Selective CYP26A inhibitors have a selectivity for CYP26A of at least 10-fold relative to CYP26B are described herein in Formulas 1 to 4, 6 to 14, 16, 17 and 18 to 29. The exemplary selective CYP26B inhibitors which have the less a 10-fold selectivity for CYP26B relative to CYP26A are described herein in Formulas 5, 15 and 30 to 32. Also included in the present invention are methods employing pharmaceutically acceptable salts, esters and amides derived from formulas 18 to 32 acceptable Suitable pharmaceutically acceptable salts of the selective CYP26A inhibitors and of the selective CYP26B inhibitors useful in the invention include, without limitation, acid addition salts, which can be formed, for example, by mixing a solution of the inhibitor with a solution of an appropriate acid such as hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid and phosphoric acid. When an inhibitor carries an acid radical, suitable pharmaceutically acceptable salts thereof may include alkaline salts such as sodium or potassium salts; alkaline earth salts such as calcium or magnesium salts; salts formed with suitable organic ligands, for example, quaternary ammonium salts. Representative pharmaceutically acceptable salts include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulonate, citrate, dihydrochloride, edetate, edisilate. , stellate, esylate, fumarate, gluceptate, gluconate, glutamate, glycolylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methyl bromide, methylnitrate, methylisulfate, mucate , napsylate, nitrate, ammonium salt of N-methylglucamine, oleate, pamoate (embonate), palmitate, pantothenate, phosphate / diphosphate, polygalacturonate, salicylate, stearate, sulfate, subacetate, succinate, tannate, tartrate, theoclate, tosylate, triethyodide and valerate It is understood that the functional groups of the selective CYP26A inhibitors and the selective CYP26B inhibitors useful in the invention can be modified to enhance the pharmacological utility of the compounds. Such modifications are within the knowledge of the skilled chemist and include, without limitation, esters, amides, ethers, N-oxides, and pro-drugs of the indicated inhibitor. Examples of modifications that can enhance the activity of an inhibitor include, for example, esterification such as the formation of Cx to C6 alkyl esters, preferably Cx to C4 alkyl esters, where the alkyl group is a straight or branched chain . Other acceptable esters include, for example, C5 to C7 cycloalkyl esters and arylalkyl esters such as benzylic esters. Such esters can be prepared from selective CYP26A or CYP26B inhibitors described herein using conventional methods well known in the art of organic chemistry. Other pharmaceutically acceptable modifications include the formation of amides. Modifications of useful amides include, for example, those derived from ammonia; Cx to C6 primary dialkylamines, where the alkyl groups are straight or branched chain; and arylamines having various substitutions. In the case of the secondary amines, the amine may also be in the form of a 5- or 6-membered ring. Methods for preparing these and other amides are well known in the art. It is understood that, when an inhibitor useful in the invention is a compound having at least one chiral center, the compound can exist in the form of chemically distinct enantiomers. further, when a compound has two or more chiral moieties, the compound exists in the form of diastereoisomers. All these isomers and mixtures thereof are encompassed within the scope of the indicated inhibitor. Similarly, when an inhibitor has a structural arrangement that allows the structure to exist in the form of tautomers, these tautomers are encompassed within the scope of the indicated inhibitor. In addition, in crystalline form, an inhibitor can exist in the form of polymorphs; in the presence of a solvent, an inhibitor can form a solvate, for example, with water or a common organic solvent. Such polymorphs, hydrates and other solvates are also encompassed within the scope of the indicated inhibitors as defined herein. A selective CYP26A inhibitor or a selective CYP26B inhibitor useful in the invention is generally administered in a pharmaceutical composition. Such pharmaceutical compositions include the active inhibitor and additionally may include, if desired, an excipient such as a pharmaceutically acceptable carrier or diluent, which is any carrier or diluent that does not substantially have a long-term or permanent deleterious effect when is administered to an individual. Such an excipient is usually mixed with an active compound, or allowed to dilute or englobe the active compound. A carrier can be a solid, semi-solid, or liquid agent that acts as an excipient or vehicle for the active compound. Examples of the pharmaceutically acceptable carriers and diluents include, without limitation, water, such as distilled or deionized water; Saline solution; and other aqueous media. It is understood that the active ingredients may be soluble or may be released as a suspension in the desired carrier or diluent. A pharmaceutical composition may additionally, if desired, include one or more agents such as emulsifying agents, wetting agents, sweetening or flavoring agents, tonicity adjusters, preservatives, buffers or anti-oxidants. Useful tonicity adjusters useful in a pharmaceutical composition may include salts such as sodium chloride, potassium chloride, mannitol or glycerin and other pharmaceutically acceptable tonicity adjusters. Preservatives useful in the pharmaceutical compositions of the invention include, without limitation, benzalkonium chloride, chlorobutanol, trimerosal, phenylmercuric acetate, and phenylmercuric nitrate. Various buffers and means for adjusting the pH can be used to prepare a pharmaceutical composition, including, but not limited to, acetate buffers, citrate buffers, phosphate buffers and borate buffers. In a similar manner, antioxidants useful in the pharmaceutical compositions of the invention are well known in the art and include, for example, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole and butylated hydroxytoluene. It is understood that these and other substances known in the pharmacology art can be included in a pharmaceutical composition useful in the invention. A selective CYP26A inhibitor or an inhibitor of
Selective CYP26B useful in a method of the invention is administered to an individual in an effective amount.
Such an effective amount is generally the minimum dose necessary to achieve the desired therapeutic effect, which may be, for example, the amount broadly necessary to reduce a symptom of a retinoid responsive disorder to a more comfortable, tolerable, acceptable level, or improved. For example, the term "effective amount" when used with respect to the treatment of a retinoid sensitive disorder may be a sufficient dose to reduce a symptom, for example, at least 30%, 40%, 50%, 60 %, 70%, 80%, 90% or 100%. Such a dose is generally in the range of 0.1-1000 mg / day and may be, for example, in the range of 0.1-500 mg / day, 0.5-500 mg / day, 0.5-100. mg / day, 0.5-50 mg / day, 0.5-20 mg / day, 0.5-10 mg / day or 0.5-5 mg / day, determining the actual amount that will be administered physical that takes into consideration the relevant circumstances including the severity of the disorder, the age and weight of the individual, the general physical condition of the individual, and the route of administration. When repeated administration is used, the frequency of administration depends, in part, on the half-life of the inhibitor. Suppositories and sustained-release formulations may be useful in the invention and include, for example, skin patches, depot formulations on or under the skin and formulations for intramuscular injection. It is understood that slow release formulations may also be useful in the methods of the invention. The individual receiving a selective CYP26A inhibitor or a selective CYP26B inhibitor can be any mammal or other vertebrate susceptible to experiencing a retinoid sensitive disorder, eg, a human, primate, horse, cow, pig, dog, cat, hamster, or bird.
Administration of a selective CYP26A inhibitor or a selective CYP26B inhibitor
Various routes of administration may be useful for treating a retinoid sensitive disorder according to the method of the invention depending, for example, on the organ or tissue to be treated, the selective inhibitor or other compound to be included in the composition. , and of the history, the risk factors and the symptoms of the subject. Suitable administration routes for the methods of the invention include both systemic and local administration. As non-limiting examples, a pharmaceutical composition useful for treating a retinoid sensitive disorder can be administered orally or by a subcutaneous pump or other implanted device; using transdermal patches; by intravenous, subcutaneous or intramuscular injection; by means of topical drops, creams, gels or ointments; through suppositories; in the form of a prolonged release formulation implanted or injected; by mini-pump or intrathecal injection; or by epidural injection. It is understood that the frequency and duration of the dosage will depend, in part, on the desired relief and the half-life of the selective CYP26A inhibitor or the selective CYP26B inhibitor. In particular embodiments, the method of the invention is practiced by peripheral administration of a selective CYP26A inhibitor or the selective CYP26B inhibitor. As used herein, the term "peripheral administration" represents methods of administration in which an agent is introduced into a subject outside the central nervous system. Peripheral administration encompasses any route of administration other than direct administration to the marrow or the brain. As such, it is clear that intrathecal and epidural administration as well as injection or cranial implant are not within the scope of the term "peripheral administration" or "peripherally administered". Peripheral administration can be local or systemic. The Local administration causes significantly more pharmaceutical composition to be released at the site of local administration than at regions remote from the site of administration. Systemic administration results in the release of a pharmaceutical composition essentially to the entire peripheral nervous system of the subject and may also make the release to the central nervous system dependent on the properties of the composition. Peripheral administration routes useful in the methods of the invention encompass, without limitation, oral administration, topical administration, intraocular administration, intravenous or other injection, and implanted minipumps or other prolonged release devices or formulations. A pharmaceutical composition useful in the invention may be administered peripherally, for example, orally in any acceptable form such as a tablet, liquid, capsule, powder, or
, Similary; by intravenous, intraperitoneal, intramuscular, subcutaneous or parenteral injection; by transdermal diffusion or electrophoresis; topically in any acceptable form such as drops, creams, gels or ointments; by inhalation; and by means of minipumps or other devices or prolonged release formulations implanted. Topical ophthalmic administration may be useful in the methods of the invention for treating an ocular disorder responsive to retinoids. Such administration can be achieved using, without limitation, eye drops, eye ointments, ocular gels and ocular creams. Such ophthalmic preparations are easy to apply and release the active ingredient effectively and avoid possible systemic side effects. Topical administration includes administration to the skin or mucosa, including the surfaces of the lung. Compositions for topical administration, including those for inhalations, can be prepared in the form of dry powder which can be pressurized or non-pressurized. In non-pressurized powder compositions, the active ingredients in finely divided form can be used in admixture with a larger pharmaceutically acceptable inert carrier comprising particles having a size, for example, up to 100 μm in diameter. Suitable inert carriers include sugars such as lactose. Desirably, at least 95% by weight of the particles of the active ingredient have an effective particle size in the range of 0.01 to 10 μm.
Synthetic Methods
SPECIFIC EXAMPLES The reaction schemes provided below together with the applicable experimental descriptions describe the synthetic routes currently preferred for preparing the preferred compounds of the invention.
Synthetic Procedures for Preparing Coupling Reagents
Reagent 1 Reagent 2
General Procedure A: Methyl 2- (4-iodophenyl) propionate (Reagent 1)
A stirred, cooled (-78 ° C) solution of methyl 4-iodophenyl acetate (described in US 6,252,090, incorporated herein by reference, 2.77 g, 10 mmol) in anhydrous tetrahydrofuran (20 ml) was treated with a solution 1.5 M lithium diisopropylamide in tetrahydrofuran and cyclohexane (8 ml, 12 mmol). The reaction mixture was allowed to warm to 0 ° C over 40 minutes, cooled again to -78 ° C and treated with methyl iodide (0.75 ml, 12 mmol). The reaction mixture was allowed to warm to room temperature over Ih. It was then quenched with a saturated aqueous solution of ammonium chloride, diluted with water and extracted with diethyl ether. The combined organic phase was washed with brine (xl), dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow oil (2.7 g, 92.7 g. %). H1 NMR (300 MHz, CDC13): d 7.66 (d, 2H, J = 8.5 Hz), 7.06 (d, 2H, J = 8.5 Hz), 3.70-3.66 (m, 1H), 3.67 (s, 3H), 1.49 (d, 3H, J = 7.0Hz).
Methyl-2- (4-iodophenyl) -2-methyl propionate (Reagent 2) Following General Procedure A and using methyl 2- (4-iodophenyl) propionate (1.45 g, 5 mmol), lithium diisopropylamide ( 1.5 M in tetrahydrofuran and cyclohexane, 4 mL, 6 mmol), tetrahydrofuran (15 mL) and methyl iodide (0.5 mL, 8 mmol), the title compound was obtained as an oil (1.5 g, 98%). NMR H1 (300 MHz, CDCl3): d 7.66 (d, 2H, J = 8.7Hz), 7.11 (d, 2H, J = 8.7Hz), 3.66 (s, 3H), 1 , 58 (s, 6H).
4-Iodo-benzyl alcohol
A stirred, cooled solution (-78 ° C) of ethyl 4-iodobenzoate (available from Lancaster, 12.9 g, 45 mmol) in anhydrous dichloromethane (100 ml) under argon was treated with a 1 M solution of diisobutylaluminum hydride. in dichloromethane (100 ml, 100 mmol). The reaction mixture was allowed to warm to 0 ° C for 1.5 h, quenched with a saturated aqueous solution of ammonium chloride and the resulting emulsion was filtered on a pad of celite. The phases in the filtrate were separated and the aqueous phase was extracted with dichloromethane (xl). The combined organic phase was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to give the title product as a white solid (9 g, 85%). NMR H1 (300 MHz, CDCl3): d 7.65 (d, 2H, J = 7.6 Hz), 7.05
(d, 2H, J "= 7.6Hz), 4.57 (s, 2H), 2.40 (broad s, 1H).
4-Iodo-benzaldehyde
A solution of 4-iodobenzyl alcohol (9 g, 38.29 mmol) in dichloromethane (90 ml) and acetonitrile (10 ml) was treated sequentially with 4 A molecular sieve powder (9 g), tetra-n-propylammonium perruthenate. (0.13 g) and N-methylmorpholine N-oxide (9 g, 76.6 mmol). After stirring at room temperature for 2 h, the reaction mixture was diluted with hexane and subjected to column chromatography on silica gel (230-400 mesh) using 6-10% ethyl acetate in hexane as eluent to provide the title compound (2.5 g pure and 4 g -95% pure, 73%). H1 NMR (300 MHz, CDC13): d 9.96 (s, 1H), 7.92 (d, 2H, J = 8.5Hz), 7.59 (d, 2H, J = 8.5Hz).
4-Ethyl iodo-cinnamate (Reagent 3)
A stirred, cooled (-78 ° C) solution of triethyl phosphonoacetate (11.1 ml, 56 mmol) in anhydrous tetrahydrofuran (100 ml) was treated with a 1.6 M solution of n-butyl lithium in hexanes (27 ml. , 43.75 mmoles). After 10 min, the reaction mixture was introduced via a cannula into a refrigerated (-78 ° C) solution of 4-iodo-benzaldehyde (6.5 g, 28 mmol) in tetrahydrofuran (20 ml). The reaction mixture was allowed to warm to 0 ° C over 1h. It was quenched with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 6-8% ethyl acetate in hexane as eluent to provide the title compound (2.7 g pure, 3.2 g -95% pure, 69%).
NMR H1 (300 MHz, CDCl3): d 7.70 (d, 2H, J = 8.5 Hz), 7.57 (d, 1H, J = 15.8Hz), 7.21 (d, 2H, J = 8.5Hz), 6.43 (d, 1H, "7 = 15.8Hz), 4.25 (c, 2H, J = 7.1Hz), 1.33 (t, 3H, J = 7.1Hz) .
4-iodine-cinnamic acid
A solution of ethyl 4-iodo cinnamate (3.2 g, 10.5 mmol) in methanol (25 ml), tetrahydrofuran (25 ml) and water (15 ml) was treated with lithium hydroxide monohydrate (4, 2 g, 100 mmol) and the resulting reaction mixture was stirred at room temperature for 2 days. The volatiles were evaporated in vacuo and the residue was neutralized with a saturated aqueous solution of ammonium chloride. The precipitated solid was filtered, washed with water and hexane and dried to give the title product as a white solid (2.9 g, 91%). This was used as is for the next stage.
4-Methyl iodo-cinnamate (Reagent 4)
A stirred, cooled solution (ice bath) of iodo-cinnamic acid in methanol was treated with a solution of diazomethane in diethyl ether. The reaction mixture was allowed to warm to room temperature, the volatiles were evaporated in vacuo to provide the title compound.
Ethyl 3- (4-iodo-phenyl) -but-2-enoic acid ester (Reagent 5)
A stirred, cooled (-78 ° C) solution of triethyl 2-phosphonoacetate (4.55 g, 20 mmol) in anhydrous tetrahydrofuran (10 ml) was treated with a 1.6 M solution of n-butyl lithium in hexanes ( 12.8 ml, 20.5 mmol). After 30 min, a solution of 4-iodoacetophenone (2.5 g, 10 mmol) in tetrahydrofuran (5 ml) was introduced via a cannula into the reaction mixture. After 4 h, quenched with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent, followed by preparative normal phase HPLC to give the title compound (0.53 g, 15%). NMR H1 (300 MHz, CDCl3): d 7.67 (d, J = 8.2 Hz, 2H), 6.94 (d, J = 8.2 Hz, 2H), 5.91 (s, 1H), 4 , 01 (c, J = 7, lHz, 2H), 2.14 (s, 6H), 1.12 (t, J "= 7, lHz, 3H).
3-Iodo-benzaldehyde A solution of 3-iodobenzyl alcohol (Aldrich, 4.72 g, 20 mmol) in dichloromethane (50 ml) and acetonitrile (5 ml) was treated sequentially with molecular sieve powder 4 A (5 g), tetra-n-propylammonium perruthenate (0.1 g) and N-Methylmorpholine N-oxide (2.34 g, 40 mmol). After stirring at room temperature for 3 h, the reaction mixture was diluted with hexane and subjected to column chromatography on silica gel (230-400 mesh) using 6-10% ethyl acetate in hexane as eluent to provide the title compound (3.7 g, 80%). This was used as is for the next stage.
Reagent 6
3-Ethyl iodo cinnamate (Reagent 6)
A stirred, cooled (-78 ° C) solution of triethyl phosphonoacetate (11.44 g, 51 mmol) in anhydrous tetrahydrofuran (100 mL) was treated with a 1.6 M solution of n-butyl lithium in hexanes (30 mL). ml, 48 mmol). After 10 min, the reaction mixture was introduced via a cannula into a refrigerated (-78 ° C) solution of 4-iodo-benzaldehyde (3.7 g, 16 mmol) in tetrahydrofuran (20 ml). The reaction mixture was allowed to warm to 0 ° C over 1h. It was quenched with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 8-10% ethyl acetate in hexane as eluent to provide the title compound (4.6 g, 95%). H1 NMR (300 MHz, CDC13): d 7.83 (s, 1H), 7.65 (dd, 1H, J = 7.9, 2Hz), 7.53 (d, 1H, J = 15.8Hz) , 7.43 (dd, 1H, J = 7.6, 2Hz), 7.07 (dd, 1H, J = 7.6, 7.9Hz), 6.38 (d, 1H, «7 = 15, 8Hz), 4.24 (c, 2H, «7 = 6.9Hz), 1.34 (t, 3H, J =
6.9Hz).
Ethyl ester of (E) -3- (4-iodo-phenyl) -2-methyl-acrylic acid (Reagent 7)
A stirred, cooled (-78 ° C) solution of triethyl 2-phosphonopropionate (10 g, 41.9 mmol) in anhydrous tetrahydrofuran (100 ml) was treated with a 1.6 M solution of n-butyl lithium in hexanes ( 25 ml, 40 mmol). After 10 min, the reaction mixture was introduced via a cannula into a refrigerated (-78 ° C) solution of 4-iodo-benzaldehyde (4.66 g, 20 mmol) in tetrahydrofuran (25 ml). After 30 minutes, it was quenched with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 9-10% ethyl acetate in hexane as eluent to provide the title compound (6.3 g, 99%). NMR H1 (300 MHz, CDCl3): d 7.71 (d, 2H, J = 8.4Hz), 7.58 (s, 1H), 7.12 (d, 2H, J = 8.4Hz), 4 , 27 (c, 2H, J = 7.2Hz), 2.08 (d, 3H, J = 1.5Hz), 1.35 (t, 3H, J = 7.2Hz).
2-Chloro-3- (4-iodo-phenyl) -acrylic acid ethyl ester (Reagent 8)
A stirred, cooled (-78 ° C) solution of chloro- (dipropyl-phosphinoyl) -acetic acid ethyl ester (6.1 g, 23.5 mmol) in anhydrous tetrahydrofuran (70 ml) was treated with a solution 1, 6 M n-butyl lithium in hexanes
(14 ml, 22 mmol). After 10 min, the reaction mixture was introduced via a cannula into a refrigerated (-78 ° C) solution of 4-iodo-benzaldehyde (2.61 g, 11.2 mmol) in tetrahydrofuran (25 mL). After 30 minutes, it was quenched with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 4-5% ethyl acetate in hexane as eluent to provide the title compound as a 1: 1 mixture of E and Z isomers (3.6 g, 95%)
4-Iodo-butylphenyl acetate (Reagent 10) A solution of 4-iodophenylacetic acid (Lancaster, 1.31 g, 5 mmol) in anhydrous toluene (10 ml) was heated to 80 ° C and treated with a solution of di-t-butylacetal of N, N-dimethylformamide. After 2 h the reaction mixture was cooled to room temperature and subjected to flash chromatography on silica gel (23-400 mesh) using 10% ethyl acetate in hexane as eluent to provide the title compound (0.degree. , 7 g, 44%). NMR H1 (300 MHz, CDC13): d 7.62 (d, 2H, J = 8.2Hz), 7.01 (d, 2H, J = 8.2Hz), 3.45 (s, 2H), 1 , 43 (s, 9H).
Reagent 11
Acid (2-fluoro-iodo-phenyl) -acetic acid acetoxymethyl ester (Reagent 11)
A solution of 2-fluoro-4-iodophenylacetic acid
(described in US 6,252,090, incorporated herein by reference, 0.82 g, 2.93 mmol) in anhydrous acetonitrile (10 ml) was treated with N, N-diisopropylethylamine (1.27 ml, 7.32 mmol) followed by bromide of acetoxymethyl / bromomethyl acetate (0.896 g, 5.86 mmol) and the resulting reaction mixture was stirred overnight at room temperature. The volatiles were evaporated in vacuo and the residue was diluted with water and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 10-20% ethyl acetate in hexane. as eluent to provide the title compound as an oil (0.75 g, 72%). H1 NMR (300 MHz, CDC13): 7.42 (m, 2H), 6.97 (dd, J = 8.0 and 8.0 Hz, 1H), 5.73 (s, 2H), 3.65 (s, 2H), 2.08 (s, 3H).
2-Trimethylsilane-ethyl ester of (2-fluoro-4-iodo-phenyl) -acetic acid (Reagent 12)
A solution of 2-fluoro-4-iodophenylacetic acid (0.3 g, 1.07 mmol) and 2- (trimethylsilyl) ethanol (0.28 mL, 1.95 mmol) in anhydrous dichloromethane (5 mL) was treated with 4- (dimethylamino) pyridine (0.275 g, 2.3 mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (0.37 g, 1.95 mmol) and the resulting reaction mixture was stirred to the Room temperature during the night. The reaction mixture was then subjected to flash column chromatography using 5% ethyl acetate in hexane as eluent to give the title compound as a white solid (0.37 g, 91%). NMR H1 (300 MHz, CDCl3): 7.44 (m, 2H), 7.02 (dd, J = 8.0, 8.0 Hz, 1H), 4.20 (t, J = 8.5Hz, 2H ), 3.59 (s, 2H), 0.98 (t, J = 8.5 Hz, 2H), 0.02 (s, 9H).
Synthesis of the Preferred Embodiments
Compound 2 Reagent 2 6.252.090 Reaction Scheme 1 General Procedure B: 2- Methyl ester. { 4 - [(8-Cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2,1'-cyclopropane] -6-yl) ethynyl] -phenyl} -propionic (Intermediate 1)
A solution of 8-cyclopropyl-6-ethynyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] (described in U.S. Patent No. 6,252,090) 0.068 g, 0.27 mmol), and methyl-2- (4-iodo phenyl) propionate (Reagent 1, 0.086 g, 0.3 mmol) in triethylamine (3 ml), was treated with copper iodide (I) (0.028 g, 0.15 mmol) and purged with argon for 5 minutes. Dichlorobis (triphenylphosphine) palladium (II) (0.057 g, 0.08 mmol) was added and the reaction mixture was stirred overnight at room temperature. It was diluted with diethyl ether and filtered on a pad of celite. The filtrate was evaporated in vacuo to a brown oil which was subjected to column chromatography on silica gel (230-400 mesh) to give the title compound as an oil (0.072 g, 56%). NMR H1 (300 MHz, CDCl3): d 7.46 (d, 2H, J = 8.4Hz), 7.29 (d, 1H, J = 2.1Hz), 7.25 (d, 2H, J = 8.4Hz), 6.80 (d, 1H, J = 2.1Hz), 3.68 (c, 1H, J = 7.2Hz), 3.66 (s, 3H), 2.02-1, 90 (m, 1H), 1.90 (s, 2H), 1.49 (d, 3H, J "= 7.2Hz), 1.39 (s, 6H), 1.03-0.99 (m , 2H), 0.90-0.83 (m, 2H), 0.68-0.59 (m, 4H).
2- (4- [(8-Cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl) ethynyl] -phenyl} - Propionic (Compound 1)
A solution of 2- methyl acid ester. { 4 - [(8-Cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2,1'-cyclopropane] -6-yl) ethynyl] -phenyl} -propionic (Intermediate 1, 0.072 g, 0.174 mmol) in methanol (5 ml) was treated with a 1 M solution of sodium hydroxide (1 ml, 1 mmol) and the resulting reaction mixture was heated at 55 ° C for 4 h . The reaction mixture was cooled to room temperature and the volatiles were evaporated in vacuo to a residue which was diluted with 10% hydrochloric acid until neutral and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title product as a white solid after flash column chromatography on silica gel (230-400 mesh) (0.04 g). g, 57%). H 1 NMR (300 MHz, CDCl 3): d 7.46 (d, 2 H, J = 8.1 Hz), 7.30-7.25 (m, 3 H), 6.80 (d, 1 H, J = 1, 8Hz), 3.74 (c, 1H, J = 7.2Hz), 1.99-1.96 (m, 1H), 1.91 (s, 2H), 1.51 (d, 3H, J = 7.2 Hz), 1.39 (s, 6H), 1.04-0.99 (m, 2H), 0.90-0.83 (m, 2H), 0.68-0.59 (m, 4H).
Methyl ester of acid 2-. { 4- [(8-cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1 '-cyclopropane] -6-yl) ethynyl] -phenyl} -2-methyl-propionic (Intermediate 2)
Following General Procedure B and using 8-cyclopropyl-6-ethynyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] (0.096 g, 0.38 mmol), Methyl 2- (4-iodophenyl) -2-methyl-propionate (Reagent 2, 0.127 g, 0.41 mmol), triethylamine (3 mL), copper (I) iodide (0.040 g, 0.21 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.080 g, 0.11 mmol) followed by flash column chromatography on silica gel (230-400 mesh), the title compound was obtained as an oil (0.046 g, 47 %). NMR H1 (300 MHz, CDCl3): d 7.39 (d, 2H, J = 8.4Hz), 7.23-7.20 (, 3H), 6.72 (d, 1H, J = 2.1Hz ), 3.58 (s, 3H), 1.92-1.84 (m, 1H), 1.84 (s, 2H), 1.51 (s, 6H), 1.33 (s, 6H) , 0.97-0.92 (m, 2H), 0.83-0.76 (m, 2H), 0.59-0.52 (m, 4H).
2- (4- [(8-Cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl) ethynyl] -phenyl} - 2-methyl-propionic (Compound 2)
A solution of 2- methyl acid ester. { 4 - [(8-Cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2,1'-cyclopropane] -6-yl) ethynyl] -phenyl} -2-methyl-propionic (Intermediate 2, 0.046 g, 0.107 mmol) in methanol (5 mL) was treated with a 1 M solution of sodium hydroxide (1.2 mL, 1.2 mmol) and the resulting reaction mixture it was heated at 55 ° C for 4 h. The reaction mixture was cooled to room temperature and the volatiles were evaporated in vacuo to a residue which was neutralized with 10% hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a white solid after flash column chromatography on silica gel ( 230-400 mesh) (0.067 g, 89%). H1 NMR (300 MHz, CDCl3): d 7.47 (d, 2H, J = 8.1 Hz), 7.36 (d,
2H, J = 8.1 Hz), 7.30 (d, 1H, J = 2.1Hz), 6.80 (d, 1H, J "=
2.1Hz), 1.99-1.91 (m, 1H), 1.91 (s, 2H), 1.60 (s, 6H), 1.40
(s, 6H), 1.04-0.99 (m, 2H), 0.90-0.84 (m, 2H), 0.69-0.59
(m, 4H).
2. NaOH Reagent 7 Compound 3
2. HPLC Reagent 3. NaOH Compound 4
Reaction Scheme 2
Ethyl ester of (E) -3- acid. { 4- [8-cyclopropyl-3,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl] ethynyl-phenyl} -2-methyl-acrylic (Intermediate 3)
Following General Procedure B and using 8-cyclopropyl-6-ethynyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, '-cyclopropane] (0.077 g, 0.3 mmol), ester Ethyl (E) -3- (4-iodo-phenyl) -2-methyl-acrylic acid (Reagent 7, 0.106 g, 0.23 mmol), triethylamine (3 ml), copper (I) iodide (0.029 g) , 0.15 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.064 g, 0.09 mmol) followed by flash column chromatography on silica gel (230-400 mesh), gave the title compound (0, 06 g, 45%). NMR H1 (300 MHz, CDC13): d 7.65 (d, 1H, J = 1.5Hz), 7.52 (d, 2H, J = 8.7Hz), 7.37 (d, 2H, J " = 8.7Hz), 7.32 (d, 1H, J = 1.8Hz), 6.82 (d, 1H, J "= 1.8Hz), 4.27 (c, 2H, J" = 7, 2Hz), 2.14 (d, 3H, J = 1.5Hz), 1.99 (m, 1H), 1.91 (s, 2H), 1.40 (s, 12H), 1.35 (t , 3H, J = 7.2 Hz), 1.04-1.00 (m, 2H), 0.91-0.84 (m, 2H), 0.69-0.59 (m, 4H).
Acid (E) -3-. { 4- [8-Cyclopropyl-3,4-dimethylspiro [2H-l-benzopyran-2, 1'-cyclopropane] -6-yl] ethynyl-phenyl} -2-methyl-acrylic (Compound 3)
A solution of ethyl ester of (E) -3- acid. { 4- [8-cyclopropyl-3,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl] ethynyl-phenyl} -2-methyl-acrylic (Intermediate 3, 0.06 g, 0.13 mmol) in ethanol (2 ml) was treated with a 1 M solution of sodium hydroxide (0.5 ml, 0.5 mmol) and the The resulting reaction mixture was heated at 55 ° C for 4 h. The reaction mixture was cooled to room temperature and the volatiles were evaporated in vacuo to a residue which was neutralized with 5% hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid after flash column chromatography on silica gel (230-400 mesh) (0.044 g, 82%). H1 NMR (300 MHz, CDC13): d 7.81 (d, 1H, J = 1.5Hz), 7.54 (d, 2H, J = 8.4Hz), 7.41 (d, 2H, J = 8.4Hz), 7.33 (d, 1H, J = 2.1Hz), 6.83 (d, 1H, J "= 2.1Hz), 2.17 (d, 3H, J = 1.5Hz) , 2.00 (m, 1H), 1.92 (s, 2H), 1.41 (s, 12H), 1.05-1.00 (m, 2H), 0.91-0.84 (m , 2H), 0.69-0.60 (m, 4H).
Ethyl ester of (Z) -2-chloro-3- (4- [8-cyclopropyl-3,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl] ethynyl}. -Acrylic (Intermediate 4)
Following General Procedure B and using 8-cyclopropyl-6-ethynyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2,1'-cyclopropane] (0.11 g, 0.436 mmol), ethyl ester of (E, Z) -2-chloro-3- (4-iodo-phenyl) -acrylic acid (Reagent 8, 0.162 g, 0.48 mmol), triethylamine (3 ml), copper iodide (I) (0.041 g, 0.21 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.092 g, 0.13 mmol) followed by flash column chromatography on silica gel (230-400 mesh), and preparative normal phase HPLC. using 5% ethyl acetate in hexane as the mobile phase, the title compound was obtained (0.09 g, 45%). H1 NMR (300 MHz, CDC13): d 7.88 (s, 1H), 7.83 (d, 2H, J = 8.1Hz), 7.55 (d, 2H, J = 8.1Hz), 7 , 33 (d, 1H, J = 2.1Hz), 6.82 (d, 1H, J = 2.1Hz), 4.36 (c, 2H, J = 6.9Hz), 1.99 (m, 1H), 1.92 (s, 2H), 1.41 (s, 12H), 1.39 (t, 3H, "7 = 6.9Hz), 1.05-1.00 (m, 2H), 0.91-0.84 (m, 2H), 0.70-0.60 (m, 4H).
Acid (Z) -2-chloro-3-. { 4- [8-cyclopropyl-3,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl] ethynyl} -Acrylic (Compound 4)
A solution of ethyl ester of (Z) -2-chloro-3- acid. { 4- [8-cyclopropyl-3,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] -6-yl] ethynyl} Acrylic (Intermediate 4, 0.09 g, 0.19 mmol) in ethanol (1 ml) and tetrahydrofuran (3 ml) was treated with a 1 M solution of sodium hydroxide (0.7 ml, 0.7 mmol) and the resulting reaction mixture was heated at 55 ° C overnight. The reaction mixture was cooled to room temperature and the volatiles were evaporated in vacuo to a residue which was neutralized with 10% hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid after flash column chromatography on silica gel ( 230-400 mesh) (0.08 g, 95%). H1 NMR (300 MHz, CDC13): d 7.74 (s, 1H), 7.55 (d, 2H, J = 8.1Hz), 7.31 (d, 2H, J = 8.1Hz), 7 , 20 (d, 1H, «7 = 1.8Hz), 6.70 (d, 1H, J = 1.8Hz), 1.86 (, 1H), 1.79 (s, 2H), 1.27 (s, 12H), 0.94-0.81 (m, 2H), 0.77-0.71 (m, 2H), 0.59-0.47 (m, 4H).
United States Patent Intermediate 5 No. 6,252,090
Intermediate 6
Compound 5 Reaction Scheme 3
6-Amino-8-cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] (Intermediate 5)
A solution of 6-bromo-8-cyclopropyl-3,4-dihydro-4,4-dimethylspiro [2JT-1-benzopyran-2, 1'-cyclopropane] (described in US 6,252,090, 0.322 g, 1.049 mmol), benzophenone imine (Fluka 0.093 ml, 1.15 mmol), sodium t-butoxide (0.142 g, 1.47 mmol), tris (dibenzylidene ketone) dipalladium (0) (0.023 g, 0.025 mmol) and (S) - (-) -2, 2'-bis (diphenylphosphino) -l, 1-binaphthyl (Aldrich, 0.047 g, 0.075 mmol) in 7 ml of anhydrous toluene was purged with argon and heated at 95 ° C for 36 h. The reaction mixture was cooled to room temperature, quenched with water and extracted with ethyl acetate. The combined organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give a thick brown oil (0.73 g). The oil was dissolved in tetrahydrofuran (3.5 ml) and treated with 2 M hydrochloric acid (1.7 ml). After stirring at room temperature for 20 minutes, 0.5 ml of 2M hydrochloric acid and 40 ml of water were added and the reaction mixture was extracted with hexane: ethyl acetate (2: 1, 3x60 ml). The aqueous phase was neutralized with potassium hydroxide and extracted with dichloromethane (3 × 50 ml). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide an oil which in column chromatography on silica gel (230-400 mesh) gave the title product as a white solid. Brown color (0.15 g, 58%). H1 NMR (300 MHz, CDC13): d 6.46 (d, 1H, J = 2.7Hz), 6.01 (d, 1H, J = 2.7Hz), 3.28 (s broad, 2H), 2.02-1.93 (m, 1H), 1.87 (s, 2H), 1.34 (s, 6H), 0.97-0.93 (m, 2H), 0.85-0, 78 (m, 2H), 0.61-0.59 (m, 4H).
Methyl ester of 4- acid. { 3 - [8-Cyclopropyl-3, 3-dihydro-4,4-dimethylspiro (2H-1-benzopyran-2, 2'-cyclopropane) -6-yl] -ureido} -benzoic (Intermediate 6)
A solution of 4-isocyanatobenzoic acid methyl ester (Aldrich, 0.17 g, 0.97 mmol) in anhydrous toluene (5 ml) was treated with a solution of 6-amino-8-cyclopropyl-3, 4- dihydro-4,4-dimethylspiro [2H-1-benzopyran-2, 1'-cyclopropane] (Intermediate 5, 0.07 g, 0.28 mmol) in toluene (15 ml). The resulting reaction mixture was stirred at room temperature overnight and at 50-60 ° C for 5h. The volatiles were evaporated in vacuo and the residue was subjected to column chromatography on silica gel (230-400 mesh) to give the title compound as a white solid (0.073 g, 62%). NMR H1 (300 MHz, CDCl3): d 7.93 (d, 2H, J = 9.0Hz), 7.39 (d, 2H, "7 = 9.0Hz), 7.06 (d, 1H, J = 2.4Hz), 6.62 (broad s, 1H), 6.53 (d, 1H, J = 2.4Hz), 3.88 (s, 3H), 2.05-1.97 (m, 1H), 1.89 (s, 2H), 1.35 (s, 6H), 1.01-0.97 (m, 2H), 0.90-0.83 (m, 2H), 0.67. -0.54 (m, 4H).
Acid 4-. { 3- [8-cyclopropyl-3,3-dihydro-4,4-dimethylspiro (2H-l-benzopyran-2, 2'-cyclopropane) -6-yl] -ureido} -benzoic (Compound 5)
A solution of methyl ester of 4- acid. { 3- [8-cyclopropyl-3, 3-dihydro-4,4-dimethylspiro (2H-1-benzopyran-2,2'-cyclopropane) -6-yl] -ureido} -benzoic acid (Intermediate 6, 0.072 g, 0.17 mmol) in methanol (3.4 ml) and tetrahydrofuran (7 ml) was treated with a 0.5 M solution of sodium hydroxide (3.4 ml, 1.7 mmoles) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo to a residue which was diluted with water, neutralized with 10% hydrochloric acid and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a white solid (0.066 g, 95%). NMR H1 (300 MHz, CD3COCD3): d 8.27 (broad s, 1H), 7.82 (d, 2H, J "= 9.0Hz), 7.52 (d, 2H, J" = 9.0Hz ), 7.20 (d, 1H, J = 2.4Hz), 6.66 (d, 1H, J "= 2.4Hz), 1.93-1.90 (m, 1H), 1.80 ( s, 2H), 1.24 (s, 6H), 0.80-0.73 (m, 2H), 0.72-0.67 (m, H), 0.57-0.41 (m, 4H).
(US Patent 6,252,090) R2 = H, R5 = R5 '= H (US Patent 6,252,090) R2 = F, R5 = R5' = H Reagent 1 R2 = H, R5 = H, R5 '= Me
Compound 6 R2 = H, R5 = R5 l '= =? H Compound 7 R2 = F, R5 = R5l = H Compound 8 R2 = H, R5 = H, R5 '= Compound 9 R2 = H, R5 = R5' = Me
Reaction Scheme 4 General Procedure C: 6-Bromo-8- [(cyclopropyl-amino) methyl] -2,2,4, 4-tetramethyl-chroman (Intermediate 7) A stirred, cooled solution (ice bath) of 6 -bromo-2,2,4,4-tetramethylchroman-8-carbaldehyde (U.S. Patent No. 6,252,090, 2.4 g, 8.4 mmol) in dichloromethane (10 ml) and acetonitrile (9 ml) was treated with cyclopropylamine (1.45 ml, 21 mmol). After 5 minutes, acetic acid (1 ml) was added followed by sodium cyanoborohydride (1.33 g, 21 mmol). The reaction mixture was stirred at room temperature for 2 h. The volatiles were removed by vacuum distillation, the residue was diluted with water and extracted with ethyl acetate (x2). The combined organic extract was washed with water, saturated aqueous sodium bicarbonate and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) gave the title compound (1.4 g, 50%) as a clear oil. NMR H1 (300 MHz, CDC13): d 7.27 (d, 1H, J = 2.1Hz), 7.16 (d, 1H, J = 2.1Hz), 3.73 (s, 2H), 2 , 19 (broad s, 1H), 2.09-2.04 (m, 1H), 1.82 (s, 2H), 1.35 (s, 6H), 1.32 (s, 6H), 0 43-0.36 (m, 4H).
6-Bromo-8- [(cyclopropyl-formyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman (Intermediate 8)
A solution of 6-bromo-8 - [(cyclopropyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman (Intermediate 7, 1.4 g, 4.14 mmol) in ethyl formate was subjected to at reflux for 6 h. The solvent was distilled off in vacuo to give the title compound as a clear oil (1.56 g, 100%).
NMR H1 (300 MHz, CDCl3): d 8.37, 8.27 (2s, 1H), 7.35, 7.29
(2d, 1H, J = 2.1Hz), 7.13, 7.11 (2d, 1H, J = 2.1Hz), 4.48
(s, 2H), 2.60-2.50 (m, 1H), 1.81 (s, 2H), 1.34 (s, 6H), 1.32 (s, 6H), 0.74- 0.70 (m, 4H).
6-Bromo-8- [(cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman (Intermediate 9)
A solution of 6-bromo-8 - [(cyclopropyl-formyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman (Intermediate 8, 1.46 g, 4.0 mmol) in anhydrous tetrahydrofuran ( 30 ml) was treated with a 2 M solution of borane complex: methyl sulfide in tetrahydrofuran (5 ml, 10 mmol) and the resulting reaction mixture was refluxed for 2 h. It was then cooled in an ice bath, a saturated aqueous solution of sodium carbonate was carefully quenched and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a white solid (1.55 g, 100%). H1 NMR (300 MHz, CDC13): d 7.26 (d, 1H, J = 2.1Hz), 7.20 (d, 1H, J = 2.1Hz), 3.64 (s, 2H), 2 , 27 (s, 3H), 1.83 (s, 2H), 1.83-1.78 (m, 1H), 1.34 (s, 6H), 1.33 (s, 6H), 0, 48-0.47 (m, 4H).
General Procedure D: 8- [(Cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-6-trimethylsilanylethynylchroman (Intermediate 10)
A solution of 6-bromo-8 - [(cyclopropyl-formyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman (Intermediate 9, 1.5 g, 4.2 mmol) in triethylamine ml) and anhydrous tetrahydrofuran (10 ml) was treated with copper (I) iodide (0.32 g, 1.68 mmol) and purged with argon for 5 minutes. Then trimethylsilylacetylene (2.5 ml, 17.6 mmol) was added followed by dichlorobis (triphenylphosphine) palladium (II) (0.737 g, 1.05 mmol). The resulting reaction mixture was heated at 70 ° C for 17 h. It was then cooled to room temperature, diluted with diethyl ether and filtered on a pad of celite. The filtrate was evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) to give the title compound as a brown oil. NMR H1 (300 MHz, CDC13): d 7.08 (d, 1H, J = 2.1Hz), 6.97 (d, 1H, J = 2.1Hz), 3.40 (s, 2H), 2 , 03 (s, 3H), 1.57 (s, 2H), 1.57-1.53 (m, 1H), 1.09 (2s, 12H), 0.25-0.22 (m, 4H) ), 0.012 (S, 9H).
General Procedure F: 8- [(Cyclopropyl-methyl-amino) -methyl] -6-ethynyl-2, 2,4, 4-tetramethyl-chroman (Intermediate 11)
A solution of 8 - [(cyclopropyl-methyl-amino) -methyl] -2,2,4, 4-tetramethyl-6-trimethylsilanylethynyl-chroman (Intermediate 10, 0.729 g, 1.97 mmol) in methanol (30 ml) it was treated with potassium carbonate (1.4 g, 10.2 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as a brown oil (0.571 g, 98%).
H1 NMR (300 MHz, CDC13): d 7.35 (d, 1H, J = 2.1Hz), 7.25 (d, 1H, J = 2.1Hz), 3.66 (s, 2H), 2 , 98 (s, 1H), 2.28 (s, 3H), 1.83 (s, 2H), 1.83-1.77 (m, 1H), 1.35 (s, 6H), 1, 34 (s, 6H), 0.50-0.47 (m, 4H).
(4 { 8 - [(Cyclopropyl-methyl-amino) -methyl] -2,2,4, 4-tetramethyl-chroman-6-ylethynyl} phenyl) -acetic acid ester (Intermediate 12)
Following General Procedure B and using 8- [(cyclopropyl-methyl-amino) -methyl] -6-ethynyl-2, 2,, -tetramethyl-chroman (Intermediate 11, 0.09 g, 0.3 mmol), ester methyl 4-iodophenylacetic acid (U.S. Patent No. 6,252,090, 0.092 g, 0.33 mmol), triethylamine (3 ml), copper (I) iodide (0.029 g, 0.15 mmol) and dichlorobis (triphenylphosphine) aladium (II) (0.064 g, 0.09 mmol) followed by flash column chromatography on silica gel (230-400 mesh), the title compound was obtained as a yellow oil (0.085 g) , 65%). H1 NMR (300 MHz, CDCl3): d 7.46 (d, 2H, J = 8.4Hz), 7.37 (d, 1H, J = 2.1Hz), 7.27-7.22 (m, 3H), 3.70 (s, 3H), 3.67 (s, 2H), 3.63 (s, 2H), 2.29 (s, 3H), 1.83 (s, 2H), 1, 83-1.81 (m, 1H), 1.35 (2s, 12H), 0.50-0.47 (m, 4H).
(4 { 8 - [(Cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} phenyl) -acetic acid (Compound 6)
A solution of (4- {8 - [(cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} phenyl) -acetic acid methyl ester ( Intermediate 12, 0.057 g, 0.13 mmol) in methanol (1 ml) and tetrahydrofuran (3 ml) was treated with a 1 M solution of sodium hydroxide (0.4 ml, 0.4 mmol) and the reaction mixture The resulting mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo to a residue which was washed with hexane, neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow oil (0.046 g, 84%). H NMR (300 MHz, CDC13): d 7.42-7.26 (m, 6H), 3.94 (s, 2H), 3.57 (s, 2H), 2.48 (s, 3H), 2.04 (m, 1H), 1.82 (s, 2H), 1.35 (s, 6H), 1.33 (s, 6H), 0.55-0.50 (m, 4H).
Methyl ester of 4- ({8- [(cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} -2-fluorophenyl) -acetic acid ester ( Intermediate 13)
Following General Procedure B and using 8- [(cyclopropyl-methyl-amino) -methyl] -6-ethynyl-2, 2,4,4-tetramethyl-chroman (Intermediate 11, 0.084 g, 0.28 mmol), ester methyl 2-fluoro-4-iodophenylacetic acid (US Pat. No. 6,252,090, 0.091 g, 0.3 mmol), triethylamine (3 ml), copper (I) iodide (0.027 g, 0.14 g) mmoles) and dichlorobis (triphenylphosphine) palladium (II) (0.060 g, 0.085 mmol) followed by flash column chromatography on silica gel (230-400 mesh), the title compound was obtained as a yellow oil ( 0.083 g, 64%). NMR H1 (300 MHz, CDCl3): d 7.37 (d, 1H, J = 2.1 Hz), 7.27-7.24 (m, 4H), 3.72 (s, 3H), 3.67 (s, 4H), 2.29 (s, 3H), 1.83 (s, 2H), 1.83-1.81 (m, 1H), 1.35 (s, 12H), 0.50- 0.47 (m, 4H).
Acid (4-. {8- [(cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} -2-fluoro-phenyl) -acetic acid (Compound 7)
A solution of (4- {8 - [(cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} -2-fluoro- methyl ester. phenyl) -acetic (Intermediate 13, 0.060 g, 0.13 mmol) in methanol (1 ml) and tetrahydrofuran (3 ml) was treated with a 1 M solution of sodium hydroxide (0.4 ml, 0.4 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo to a residue which was washed with hexane, neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow oil (0.056 g, 95%). H1 NMR (300 MHz, CDC13): d 7.43 (d, 1H, J "= 2.1Hz), 7.37-7.13 (m, 4H), 3.99 (s, 2H), 3, 61 (s, 2H), 2.52 (s, 3H), 2.10-2.04 (m, 1H), 1.83 (s, 2H), 1.83-1.81 (m, 1H) , 1.36 (s, 6H), 1.35 (s, 6H), 0.90-0.82 (m, 2H), 0.59-0.57 (m, 2H).
2- (4- { 8 - [(Cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} -phenyl) -propionic acid methyl ester ( Intermediate 14)
Following General Procedure B and using 8- [(cyclopropyl-methyl-amino) -methyl] -6-ethynyl-2,2,4,4-tetramethyl-chroman (Intermediate 11, 0.08 g, 0.27 mmol) Methyl 2- (4-iodophenyl) propionate (Reagent 1, 0.086 g, 0.29 mmol), triethylamine (3 ml), copper iodide (I) (0.026 g, 0.14 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.057 g, 0.08 mmol) followed by flash column chromatography on silica gel (230-400 mesh), the title compound was obtained as a brown oil (0.067 g, 54% ). H1 NMR (300 MHz, CDC13): d 7.46 (d, 2H, J = 8.4Hz), 7.37 (d, 1H, J = 2.1Hz), 7.27-7.22 (m, 3H), 3.72 (c, 1H, J = 7.2Hz), 3.67 (s, 5H), 2.29 (s, 3H), 1.83 (s, 2H), 1.83-1 , 79 (m, 1H), 1.50 (d, 3H, J "= 7.2Hz), 1.35 (s, 12H), 0.50-0.47 (m, 4H).
2- (4- (8 - [(Cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} -phenyl) -propionic acid (Compound 8)
A solution of 2- (4- { 8 - [(Cyclopropyl-methyl-amino) -methyl] -2,2,4, 4-tetramethyl-chroman-6-ylethynyl} -phenyl ester) -propionic (Intermediate 14, 0.057 g, 0.12 mmol) in methanol (1 ml) and tetrahydrofuran (3 ml) was treated with a 1 M solution of sodium hydroxide (0.3 ml, 0.3 mmol) and the The resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo to a residue which was washed with hexane, neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid (0.024 g, 45%). H1 NMR (300 MHz, CDC13): d 7.38-7.23 (2m, 6H), 3.85-3.82 (m,
1H), 3.82 (s, 2H), 2.39 (s, 3H), 1.94-1.85 (m, 1H), 1.80
(s, 2H), 1.41 (d, 3H, "7 = 7.2Hz), 1.33 (s, 12H), 0.70-0.60
(m, 2H), 0.50-0.48 (, 2H).
Methyl ester of 2- (4- { 8 - [(cyclopropyl-methyl-amino) -methyl] -2, 2,4,4-tetramethyl-chroman-6-ylethynyl} - phenyl) -2 -methyl-propionic (Intermediate 15)
Following General Procedure B and using 8- [(cyclopropyl-methyl-amino) -methyl] -6-ethynyl-2, 2,4,4-tetramethyl-chroman (Intermediate 11, 0.08 g, 0.27 mmol) Methyl 2- (4-iodophenyl) -2-methyl-propionate (Reagent 2, 0.082 g, 0.27 mmol), triethylamine (2 ml), copper (I) iodide (0.020 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent, was obtained the title compound in the form of a brown oil (0.040 g, 31%). NMR H1 (300 MHz, CDCl3): d 7.50-7.28 (m, 6H), 3.68 (s, 3H), 3.66 (s, 2H), 2.30 (s, 3H), 1.85 (s, 2H), 1.85-1.81 (m, 1H), 1.60 (s, 3H), 1.59 (s, 3H), 1.37 (s, 6H), 1 , 36 (s, 6H), 0.50-0.47 (m, 4H).
2- (4- { 8 - [(Cyclopropyl-methyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-ylethynyl} -phenyl) -2-methyl-propionic acid (Compound 9)
A solution of 2- (4- { 8 - [(cyclopropyl-methyl-amino) -methyl] -2, 2,4,4-tetramethyl-chroman-6-ylethynyl} -phenyl ester) -2-Methyl-propionic (Intermediate 15, 0.040 g, 0.084 mmol) in methanol (2.5 mL) and tetrahydrofuran (2.5 mL) was treated with a 2 M solution of sodium hydroxide (1 mL, 2 mmol) and the resulting reaction mixture was refluxed overnight. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Preparative reverse phase HPLC on a Partisil 10 ODS-3 column using 10% water in acetonitrile as the mobile phase yielded the title compound (0.008 g, 27%). H1 NMR (300 MHz, CDC13): d 7.46-7.32 (m, 6H), 6.90-6.50 (broad s, 1H), 3.84 (s, 2H), 2.41 ( s, 3H), 1.97-1.92 (m, 1H), 1.83 (s, 2H), 1.55 (s, 6H), 1.36 (2s, 12H), 0.73-0 , 68 (, 2H), 0.52-0.46 (m, 2H).
Intermediate 15A Intermediate 17
Compound 13 Compound 11 R5 = H, R5 = Compound 12 R5 = R5, = Me
Intermediate 19 United States No. Compound 10 6,252,090 2. NaOH Reaction Scheme 5-Acetyl-6-bromo-2,2,4,4-tetramethylchroman (Intermediate 5A) A stirred, cooled solution (-78 ° C) of 6- Bromo-2,2,4,4,4-tetramethylchroman (1 g, 3.72 mmol) in anhydrous dichloromethane (10 ml) was treated with aluminum chloride (0.8 g, 6.8 mmol) followed by acetyl chloride ( 0.4 ml, 6.08 mmol). After 10 minutes, the reaction mixture was diluted with water and extracted with diethyl ether. The organic phase was washed with water, and dried over anhydrous sodium sulfate, filtered and evaporated to a residue which was subjected to flash chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane. as eluent to provide the title compound as a solid (0.78 g, 67%).
H1 NMR (300 MHz, CDC13): d 7.64 (d, 1H, J = 2.6Hz), 7.49 (d, 1H, J = 2.6Hz), 2.60 (s, 3H), 1 , 87 (s, 2H), 1.41 (s, 6H), 1.36 (s, 6H). -
8-Acetoxy-6-bromo-2, 2,4, 4-tetramethylchroman (Intermediate 16)
A solution of 8-acetyl-6-bromo-2,2,4,4-tetramethylchroman (Intermediate 15A, 1.3 g, 4.18 mmol) in anhydrous dichloromethane (30 ml) was treated with a 77% aqueous solution. of 3-chloroperoxybenzoic acid (5.75 g, 33.44 mmol) and the resulting reaction mixture was stirred at room temperature for 24 h. The reaction mixture was then cooled in an ice bath and carefully quenched with a saturated solution of sodium thiosulfate. The phases were separated and the organic phase was washed with an aqueous solution of saturated sodium bicarbonate, water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to provide a residue which in column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent afforded the title compound as an oil (1.3 g, 92%). NMR H1 (300 MHz, CDC13): d 7.27 (s, 1H), 7.00 (s, 1H), 2.29 (s, 3H), 1.83 (s, 2H), 1.34 ( s, 6H), 1.32 (s, 6H).
6-Bromo-8-hydroxy-2, 2,4, 4-tetramethylchroman (Intermediate 17)
A solution of 8-acetoxy-6-bromo-2,2,4,4-tetramethylchroman (Intermediate 16, 1.3 g, 3.98 mmol) in methanol was treated with sodium carbonate (0.8 g, 7%). 95 mmoles) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide a residue which in column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent provided the title product in the form of an oil (0.95 g, 84%). NMR H1 (300 MHz, CDCl3): d 6.91 (d, 1H), 6.88 (d, 1H), 5.67 (s, 1H), 1.84 (s, 2H), 1.37 ( s, 6H), 1.32 (s, 6H).
6-Bromo-8-trimethylsilanylethynyl-2,4,4,4-tetramethylchroman (Intermediate 18)
Following General Procedure D and using 6-bromo-8-hydroxy-2,2,4,4-tetramethylchroman (Intermediate 17,
1.0 g, 3.51 mmol), triethylamine (5 ml), copper iodide (I) (0.066 g, 0.351 mmol), trimethylsilylacetylene (2.5 ml, 17.6 mmol) and dichlorobis (triphenylphosphine) palladium ( II) (0.246 g, 0.351 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 0.5% ethyl acetate in hexane as eluent, the title compound was obtained (1.08 g. , -100%) in the form of a brown oil. NMR H1 (300 MHz, CDC13): d 6.97 (d, 1H), 6.86 (d, 1H), 5.61 (s, 1H), 1.84 (s, 2H), 1.37 ( s, 6H), 1.33 (s, 6H), 0.24 (s, 9H).
6-Ethynyl-8-hydroxy-2, 2,4, 4-tetramethyl-chroman (Intermediate 19)
A solution of 6-bromo-8-trimethylsilanylethynyl-2,2,4,4-tetramethylchroman (Intermediate 18, 0.47 g, 1.56 mmol) in methanol (5 ml) was treated with potassium carbonate
(0.2 g, 1.45 mmol) and the resulting reaction mixture was heated at 80 ° C for 3 h. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as a brown oil (0.35 g, 99%). NMR H1 (300 MHz, CDC13): d 6.97 (d, 1H), 6.86 (d, 1H), 5.70 (broad s, 1H), 2.92 (s, 1H), 1.84 (s, 2H), 1.37 (s, 6H), 1.33 (s, 6H).
[4- (8-Hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 20)
Following General Procedure B and using 6-ethynyl-8-hydroxy-2,2,4,4-tetramethyl-chroman (Intermediate 19, 0.035 g, 0.15 mmol), 4-iodophenylacetic acid methyl ester (0.060 g, 0.23 mmol), triethylamine (3 ml), copper iodide (I) (0.020 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by chromatography in an instantaneous column on silica gel (230-400 mesh), and preparative normal phase HPLC using 10% ethyl acetate in hexane as mobile phase, the title compound was obtained (0.015 g, 25%). NMR H1 (300 MHz, CDCl3): d 7.46 (d, 2H, J = 8.1 Hz), 7.24 (d, 2H, J = 8.1 Hz), 7.03 (d, 1H, J = 2.1Hz), 6.91 (d, 1H, J = 2.1Hz), 5.72 (s, 1H), 3.69 (s, 3H), 3.63 (s, 2H), 1.86 (s, 2H), 1.38 (s, 6H), 1.35 (s, 6H).
Methyl ester of acid [2-fluoro-4- (8-hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid (Intermediate 21)
Following General Procedure B and using 6-ethynyl-8-hydroxy-, 2, 4, 4-tetramethyl-chroman (Intermediate
19, 0.05 g, 0.22 mmol), 2-fluoro-4-iodophenylacetic acid methyl ester (0.096 g, 0.33 mmol), triethylamine (3 ml), copper (I) iodide (0.020 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh), and preparative normal phase HPLC using acetate from ethyl to
% in hexane as mobile phase, the title compound was obtained (0.037 g, 43%). NMR H1 (300 MHz, CDCl3): d 7.27-7.18 (m, 3H), 7.03 (d, 1H,
J = 1.8Hz), 6.90 (d, 1H, J = 1.8Hz), 5.68 (s, 1H), 3.72
(s, 3H), 3.67 (s, 2H), 1.87 (s, 2H), 1.39 (s, 6H), 1.36 (s, 6H).
Acid [2-fluoro-4- (8-hydroxy-2,4,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid (Compound 10)
A solution of [2-fluoro-4- (8-hydroxy-2, 2,4, 4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid methyl ether (Intermediate 21, 0.037 g, 0.0493 mmoles) in methanol (2 ml) and tetrahydrofuran (1 ml) was treated with a 2 M solution of potassium hydroxide (2 ml, 4 mmol) and the resulting reaction mixture was stirred at room temperature for 2 h. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product.
(0.024 g, 69%). H NMR (300 MHz, CDCl 3): d 7.26-7.24 (m, 3H), 7.03 (d, 1H, J = 1.8Hz), 6.90 (d, 1H, J = 1, 8Hz), 3.71 (s, 2H), 1.87 (s, 2H), 1.39 (s, 6H), 1.36 (s, 6H).
2- [4- (8-Hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -propionic acid methyl ester (Intermediate 22) 87
Following the general procedure B and using 6-ethynyl-8-hydroxy-2,2,4,4-tetramethyl-chroman (Intermediate 19, 0.04 g, 0.17 mmol), 2- (4-iodophenyl) propionate methyl (Reagent 1, 0.075 g, 0.26 mmol), triethylamine (3 ml), copper iodide (I) (0.020 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh), and preparative normal phase HPLC using 5% ethyl acetate in hexane as mobile phase, the title compound was obtained as a brown oil (0.018 g, 26%). NMR H1 (300 MHz, CDC13): d 7.46 (d, 2H, J = 8.5Hz), 7.26 (d, 2H, J "= 8.5Hz), 7.03 (d, 1H, J = 1.8Hz), 6.91 (d, 1H, J = 1.8Hz), 5.66 (s, 1H), 3.67 (c, 1H, J = 7.5Hz), 1.87 (s) , 2H), 1.50 (d, 3H, "7 = 7.5Hz), 1.39 (s, 6H), 1.36 (s, 6H).
2- [4- (8-Hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -propionic acid (Compound 11)
A solution of 2- [4- (8-hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -propionic acid methyl ester (Intermediate 22, 0.018 g, '0.046 mmol) in methanol (1 ml) and tetrahydrofuran (0.5 ml) was treated with a 2 M solution of potassium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at 80 ° C for 2 h. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid (0.017 g, 100%). NMR H1 (300 MHz, CDC13): d 7.50-7.30 (m, 4H), 7.02 (s, 1H), 6.91 (s, 1H), 3.80-3.70 (m , 1H), 1.86 (s, 2H), 1.52 (d, 3H, J = 7.2Hz), 1.39 (s, 6H), 1.36 (s, 6H).
Methyl ester of 2- [4- (8-hydroxy-2, 2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -2-methyl-propionic acid ester (Intermediate
23) Following General Procedure B and using 6-ethynyl-8-hydroxy-2, 2,4,4-tetramethyl-chroman (Intermediate 19, 0.057 g, 0.25 mmol), methyl 2- (4-iodophenyl) -2-methyl-propionate (Reagent 2, 0.112 g, 0.37 mmol) , triethylamine (3 ml), copper (I) iodide (0.020 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on gel of silica (230-400 mesh) and preparative normal phase HPLC using 5% ethyl acetate in hexane as mobile phase, the title compound was obtained as a brown oil (0.035 g, 35%). H1 NMR (300 MHz, CDCl3): d 7.46 (d, 2H, J = 8.5 Hz), 7.29 (d, 2H, J = 8.5 Hz), 7.03 (d, 1H, J = 1.8Hz), 6.91 (d, 1H, J = 1.8Hz), 5.67 (s, 1H), 3.66 (s, 3H), 1.86 (s, 2H), 1.58 (s, 6H), 1.39 (s, 6H), 1.36 (s, 6H).
2- [4- (8-Hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -2-methyl-propionic acid (Compound 12)
A solution of 2- [4- (8-hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -2-methyl-propionic acid methyl ester (Intermediate 23, 0.035 g, 0.087 mmoles) in methanol (2 ml) and tetrahydrofuran (1 ml) was treated with a 1 M solution of potassium hydroxide (2 ml, 4 mmol) and the resulting reaction mixture was stirred at 80 ° C for 2 h. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid (0.034 g, 100%). NMR H1 (300 MHz, CDCl3): d 7.47 (d, 2H, J = 8.7Hz), 7.35 (d, 2H, J = 8.7Hz), 7.03 (d, 1H, J = 1.8Hz), 6.91 (d, 1H, J = 1.8Hz), 1.86 (s, 2H), 1.60 (s, 6H), 1.39 (s, 6H), 1.36 (s, 6H).
Methyl ester of [4- (8-isopropoxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid (Intermediate 24)
A solution of [4- (8-hydroxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 20, 0.02 g, 0.076 mmol) in acetone (2 ml) was treated with potassium carbonate (0.026 g, 0.19 mmol) and 2-iodopropane (5 ml, large excess) and the resulting reaction mixture was refluxed for 3Oh. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as an oil (0.02 g, 91%). H1 NMR (300 MHz, CDC13): d 7.47 (d, 2H, J = 8.4Hz), 7.24 (d, 2H, J = 8.4Hz), 7.14 (d, 1H, J = 2.1Hz), 6.93 (d, 1H, J "= 2.1Hz), 4.40 (heptet, 1H, J" = 6.3Hz), 3.70 (s, 3H), 3.63 ( s, 2H), 1.83 (s, 2H), 1.38 (s, 6H), 1.35 (s, 6H), 1.33 (d, 3H, J = 6.3Hz).
Acid [4- (8-isopropoxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid (Compound 13)
A solution of [4- (8-isopropoxy-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 24, 0.02 g, 0.05 mmol) in methanol (1 ml) was treated with a 2 M solution of sodium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at room temperature for 3 h. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 5% methanol in ethyl acetate as eluent followed by preparative reverse phase HPLC using 10% water in acetonitrile as mobile phase yielded the title product (0.015 g, 78%). NMR H1 (300 MHz, CDCl3): d 7.47 (d, 2H, J = 8.4Hz), 7.24 (d, 2H, J = 8.4Hz), 7.14 (d, 1H, J = 2.1Hz), 6.92 (d, 1H, J = 2.1Hz), 4.40 (heptet, 1H, «7 = 7.5Hz), 3.65 (s, 2H), 1.83 (s) , 2H), 1.37 (s, 6H), 1.35 (s, 6H), 1.33 (d, 3H, J = 7.5Hz).
A1C13, CH3COCI, CH2C12 United States Intermediate No. 25 6,252,090
Intermediate 26 Intermediate 27
Compound 14 Reagent 2 2. KOH Reaction Scheme 6
6-Bromo-2, 2-diethyl-4, 4-dimethylchroman (Intermediate 25)
A solution of 6-bromo-4,4-dimethyl-chroman-2-one (U.S. Patent No. 6,252,090, 4 g, 15.7 mmol) in anhydrous tetrahydrofuran (20 ml) was treated with a solution 3 M ethylmagesium bromide (10.5 ml, 31.5 mmol) and stirred at room temperature for 2 h. The reaction mixture was poured into cold dilute hydrochloric acid and extracted with ethyl acetate (x2). The combined organic extract was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to give a residue which was dissolved in 50 ml of benzene, was treated with p-toluenesulfonic acid (1 g, 3.92 mmol) and the resulting reaction mixture was refluxed overnight. The reaction mixture was cooled to room temperature, filtered over silica gel and washed with 10% ethyl acetate in hexane. The filtrate and washings were evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent to give the title compound a of a light yellow oil (3.9 g, 84%). H 1 NMR (300 MHz, CDCl 3): d 7.36 (d, 1 H, J = 2.4 Hz), 7.35 (dd, 1 H, J = 2.4, 8.4 Hz), 6.70 (d, 1H, J = 8.4Hz), 1.79 (s, 2H), 1.73-1.55 (m, 4H), 1.34 (s, 6H), 0.90 (t, 6H, J = 7.5Hz).
8-Acetyl-6-bromo-2,2-diethyl-4,4-dimethylchroman (Intermediate 26)
A stirred, cooled suspension (ice bath) of aluminum chloride (1.1 g, 8.38 mmol) in anhydrous dichloromethane (20 ml) was treated with acetyl chloride (0.6 ml, 8.38 mmol). After 5 minutes, a solution of 6-bromo-2,2-diethyl-4,4-dimethylchroman (Intermediate 25,
1.66 g, 5.59 mmol) in dichloromethane. The reaction mixture was stirred for 1 h. The reaction mixture was then poured into water and extracted with diethyl ether
(x2) The combined organic phase was washed with a saturated aqueous solution of sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh). ) using 10% ethyl acetate in hexane as eluent to give the title compound as an oil (1.6 g, 84%). NMR H1 (300 MHz, CDCl3): d 7.64 (d, 1H, J = 2.1Hz), 7.48 (d, 1H, J = 2.1Hz), 2.62 (s, 3H), 1 , 84 (s, 2H), 1.75-1.59 (m, 4H), 1.36 (s, 6H), 0.93 (t, 6H, J = 7.5Hz).
6-Bromo-2, 2-diethyl-8-isopropyl-4, 4-dimethylchroman (Intermediate 27)
A stirred, cooled solution (ice bath) of 8-acetyl-6-bromo-2,2-diethyl-4,4-dimethylchroman (Intermediate 26, 1.57 g, 4.62 mmol) in anhydrous tetrahydrofuran (10 ml. ) was treated with a 3 M solution of methylmagnesium bromide in diethyl ether (3.1 ml, 9.24 mmol). The reaction mixture was allowed to warm to room temperature over 2h. The reaction mixture was poured into cold, diluted aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which in flash column chromatography silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent provided an oil (1.41 g, 86%). A stirred, cooled solution (ice bath) of the oil (1.4 g, 3.93 mmol) in dichloromethane (10 ml) was treated with triethylsilane (5 ml, 31.46 mmol) followed after 30 minutes of acid trifluoroacetic acid (2.4 ml, 31.46 mmol) and the resulting reaction mixture was allowed to warm to room temperature and stirred for 3 h. The volatiles were removed by vacuum distillation and the residue was diluted with water and extracted with ethyl acetate. The combined organic phase was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) to provide the title compound as an oil. clear (0.89 g, 66%) and some starting material recovered (0.23 g, 16.4%). H1 NMR (300 MHz, CDC13): d 7.21 (d, 1H, J = 2.1 Hz), 7.11 (d, 1H, J = 2.1 Hz), 3.40-3.30 (m, 1H), 1.78 (s, 2H), 1.68-1.58 (m, 4H), 1.33 (s, 6H), 1.90 (d, 6H, J = 6.6Hz), 0 , 92 (t, 6H, J = 7.5 Hz).
2, 2-Diethyl-8-isopropyl-6-trimethylsilanylethynyl-4,4-dimethylchroman (Intermediate 28)
Following General Procedure D and using 6-bromo-2, 2-diethyl-8-isopropyl-4,4-dimethylchroman (Intermediate 27, 0.89 g, 2.62 mmol), triethylamine (5 ml), tetrahydrofuran (10 ml), copper iodide (I) (0.050 g, 0.26 mmol), trimethylsilylacetylene (2.5 ml, 17.6 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.184 g, 0.26 mmol) followed of flash column chromatography on silica gel (230-400 mesh) using hexane at 2% ethyl acetate in hexane as eluent, the title compound was obtained (0.73 g, 79%) as a color oil brown. H NMR (300 MHz, CDCl3): d 7.31 (d, 1H), 7.12 (d, 1H), 3.20-3.10 (m, 1H), 1.70 (s, 2H), 1.70-1.45 (m, 4H), 1.34 (s, 6H), 0.95 (d, 6H), 0.68 (t, 6H), 0.00 (s, 9H).
2,2-Diethyl-6-ethynyl-8-isopropyl-4,4-dimethylchroman (Intermediate 29) A solution of 2,2-diethyl-8-isopropyl-6-trimethylsilanylethynyl-4,4-dimethylchroman (Intermediate 28.0 , 73 g, 2.04 mmol) in methanol (40 ml) was treated with potassium carbonate (0.degree., 15 g, 1.08 mmol) and the resulting reaction mixture was heated at 80 ° C for 3 h. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as a brown oil (0.56 g, 96%). NMR H1 (300 MHz, CDCl3): d 7.27 (d, 1H), 7.16 (d, 1H), 3.31-3.06 (m, 1H), 2.96 (s, 1H), 1.81 (s, 2H), 1.81-1.56 (m, 4H), 1.31 (s, 6H), 1.17 (d, 6H), 0.91 (t, 6H).
2- [4- (2, 2-Diethyl-8-isopropyl-4,4-dimethyl-chroman-6-ylethynyl) -phenyl] -2-methyl-propionic acid methyl ester (Intermediate 30)
Following General Procedure B and using 2,2-diethyl-6-ethynyl-8-isopropyl-4,4-dimethylchroman (Intermediate
29, 0.069 g, 0.24 mmol), methyl 2- (4-iodophenyl) -2-methyl-propionate (Reagent 2, 0.146 g, 0.48 mmol), triethylamine (3 ml), copper iodide (I ) (0.025 g, 0.13 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.075 g, 0.107 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent, the title compound was obtained as a yellow oil (0.070 g, 62%). H1 NMR (300 MHz, CDCl3): d 7.47 (d, 2H, J "= 8.2 Hz), 7.31 (d,
2H, J = 8.2Hz), 7.30 (d, 1H, J = 2.1Hz), 7.20 (d, 1H, J =
2.1Hz), 3.65 (s, 3H), 3.40-3.20 (m, 1H), 1.78 (s, 2H), 1.68-1.57 (m, 4H), 1 , 58 (s, 6H), 1.34 (s, 6H), 1.21 (d, 6H, J = 7.0Hz), 0.91 (t, 6H, J = 7.3Hz).
2- [4- (2,2-Diethyl-8-isopropyl-4,4-dimethyl-chroman-6-ylethynyl) -phenyl] -2-methyl-propionic acid (Compound 14)
A solution of 2- [4- (2,2-diethyl-8-isopropyl-4,4-dimethyl-chroman-6-ylethynyl) -phenyl] -2-methyl-propionic acid methyl ester (Intermediate 30, 0.070 g , 0.15 mmol) in methanol (3 ml) and tetrahydrofuran (0.5 ml) was treated with a 5 M solution of potassium hydroxide (2 ml, 10 mmol) and the resulting reaction mixture was stirred at room temperature for 2 days . The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide a residue which in preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase provided the title product in the form of a yellow solid (0.035 g, 51%). NMR H1 (300 MHz, CDC13): d 7.48 (d, 2H, J "= 8.1 Hz), 7.36 (d, 2H, J = 8.2 Hz), 7.31 (d, 1H, J = 2.1Hz), 7.20 (d, 1H, J "= 2.1Hz), 3.40-3.20 (m, 1H), 1.79 (s, 2H), 1.69-1, 60 (m, 4H), 1.61 (s, 6H), 1.35 (s, 6H), 1.21 (d, 6H, J = 7.2Hz), 0.92 (t, 6H, "7 = 7.5Hz).
(US 6,252,090)
Intermediate 15A Intermediate 31 I
Reaction Scheme 7 6-Bromo-8-isopropyl-2, 2,4, 4-tetramethyl-chroman (Intermediate 31)
A stirred, cooled solution (ice bath) of 8-acetyl-6-bromo-2,2,4,4-tetramethylchroman (Intermediate 15A, 3.1 g, 10 mmol) in anhydrous tetrahydrofuran (40 ml) was treated with a 3 M solution of methylmagnesium bromide in diethyl ether (11 ml, 44 mmol). The reaction mixture was allowed to warm to room temperature overnight. The reaction mixture was poured into cold, diluted aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue that in flash column chromatography silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent provided a oil (2.85 g, 87%). The oil (1.67 g, 5.12 mmol) was cooled (ice bath) and treated with triethylsilane (10 ml, 62 mmol) followed after 30 minutes of trifluoroacetic acid (5 ml, 65 mmol) and the The resulting reaction mixture was allowed to warm to room temperature overnight. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic phase was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) to provide the title compound as an oil clear (1 g, 63%). NMR H1 (300 MHz, CDC13): d 7.20 (d, 1H, J = 2.3Hz), 7.09 (d, 1H, J = 2.3Hz), 3.25 (heptet, 1H, J = 7.1 Hz), 1.79 (s, 2H), 1.33 (s, 6H), 1.31 (s, 6H), 1.15 (d, 6H, J = 7.1Hz).
6-Ethynyl-8-isopropyl-2, 2,4, 4-tetramethyl-chroman (Intermediate 32)
Following General Procedure D and using 6-bromo-8-isopropyl-2,2,4,4-tetramethylchroman (Intermediate 31,
1 g, 3.2 mmol), triethylamine (10 ml), copper iodide (I)
(0.04 g, 0.21 mmol), trimethylsilylacetylene (5 mL, 35 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.12 g, 0.17 mmol) followed by flash column chromatography on silica gel (230-400 mesh), intermediate trimethylsilylacetylene was obtained, which was dissolved in methanol and treated with potassium carbonate and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title compound as a brown oil (0.6 g, 73%). %). H NMR (300 MHz, CDCl 3): d 7.34 (d, 1 H, J = 2.1 Hz), 7.21 (d, 1 H, J = 2.1 Hz), 3.50 (heptet, 1 H, J = 6.8Hz), 3.00 (s, 1H), 1.85 (s, 2H), 1.38 (s, 6H), 1.37 (s, 6H), 1.22 (d, 6H, J = 6.8Hz).
Ethyl 3- [3- (8-isopropyl-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acrylic acid ester (Intermediate 33)
Following General Procedure B and using 6-ethynyl-8-isopropyl-2,2,4,4-tetramethylchroman (Intermediate 32, 0.05 g, 0.2 mmol), ethyl 3-iodocinnamate (Reagent 6, 0.118 g , 0.39 mmoles), triethylamine (2 ml), copper (I) iodide (0.025 g, 0.13 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.075 g, 0.107 mmol) followed by flash column chromatography on silica gel (230-400 mesh), the title compound was obtained (0.058 g, 69%). NMR H1 (300 MHz, CDCl3): d 7.62-7.22 (m, 6H), 7.14 (d, 1H, J = 1.8Hz), 6.39 (d, 1H, J = 16, 1Hz), 4.19 (c, 2H, J = 7.0Hz), 3.21 (heptet, 1H, "7 = 6.7Hz), 1.76 (s, 2H), 1.29 (s, 12H). ), 1.27 (t, 3H, J = 7.0Hz) .l, 13 (d, 6H, J = 6.7Hz).
3- [3- (8-Isopropyl-2,2,4,4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acrylic acid (Compound 15)
A solution of 3- [3- (8-isopropyl-2, 2,4, 4-tetramethyl-chroman-6-ylethynyl) -phenyl] -acrylic acid ethyl ester (Intermediate 33, 0.058 g, 0.13 mmol) in ethanol (2 ml) and tetrahydrofuran (2 ml) was treated with a 5 N solution of potassium hydroxide (2 ml, 10 mmol) and the reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo, the residue was neutralized with dilute hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate and evaporated to give the title compound
(0.036 g, 66%). NMR H1 (300 MHz, CDCl3): d 7.69 (d, 1H, J = 15.8Hz), 7.65
(s, 1H), 7.47 (d, 1H, J = 7.6Hz), 7.39 (d, 1H, «7 = 7.9Hz), 7.32-7.17 (m, 2H), 7.14 (d, 1H, "7 = 1.8Hz), 6.41 (d, 1H, J = 15.8Hz), 3.21 (heptet, 1H, J = 6.7Hz), 1.76 ( s, 2H), 1.29 (s, 12H), 1.13 (d, 6H, J = 6.7Hz).
Intermediate 31 Intermediate 34
Intermediate 35 Compound 16 Reaction Scheme 8 8-Isopropyl-2,2,4,4-tetramethyl-chroman-6-carboxylic acid (Intermediate 34) A stirred, cooled solution (-78 ° C) of 6-bromo-8- isopropyl-2,2,4,4,4-tetramethyl-chroman (Intermediate 31, 0.39 g, 1.26 mmol) in anhydrous diethyl ether (10 ml) was treated with a 1.7 M solution of t-butyl lithium in pentane (1.48 ml, 2.516 mmol) and the reaction mixture was stirred for 20 minutes. Carbon dioxide (generated from dry ice) was bubbled into the reaction mixture. The reaction mixture was then quenched with 10% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to a residue which was subjected to flash column chromatography to provide the title compound (0.3, 86%). H1 NMR (300 MHz, CDC13): d 7.87 (d, 1H, J = 2Hz), 7.72 (d, 1H, J = 2Hz), 3.21 (heptet, 1H, J = 7.0Hz) , 1.78 (s, 2H), 1.39 (s, 12H), 1.14 (d, 6H, J = 7.0Hz).
8-isopropyl-2,4,4,4-tetramethyl-chroman-6-carboxylic acid ester (Intermediate 35) 4- (2-benzyloxycarbonyl-vinyl) -phenyl ester
A solution of 8-isopropyl-2,2,4,4-tetramethyl-chroman-6-carboxylic acid (Intermediate 34, 0.05 g, 0.18 mmol) and 3- (4-hydroxy-phenyl) benzyl ester ) -acrylic
(described in Journal of Natural Products, 1990, 53 (4), pp. 821-824, Bankova V., 0.046 g, 0.18 mmol) in anhydrous dichloromethane (5 ml) was treated with 4- (dimethylamino) pyridine ( 0.052 g, 0.27 mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride
(0.044 g, 0.36 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was then subjected to flash column chromatography using 20% ethyl acetate in hexane as eluent to give the title compound as a white solid (0.076 g, 83%). H1 NMR (300 MHz, CDC13): d 7.93 (d, 1H, J = 2Hz), 7.78 (d, 1H, "7 = 2Hz), 7.66 (d, 1H," 7 = 16, 1Hz), 7.49 (d, 2H, J = 8.5Hz), 7.35-7.25 (m, 5H), 7.15 (d, 2H, J = 8.5Hz), 6.39 ( d, 1H, "7 = 16.1Hz), 5.18 (s, 2H), 3.24 (heptet, 1H, J = 7.1Hz), 1.80 (S, 2H), 1.31 (s) , 12H), 1.16 (d, 6H, "7 = 7.1Hz).
8-Isopropyl-2,2,4,4-tetramethyl-chroman-6-carboxylic acid 4- (2-carboxy-vinyl) -phenyl ester (Compound 16)
A suspension of t-butyldimethylsilane (0.3 ml, 1.85 mmole), palladium (II) acetate (0.013 g, 0.06 mmole) and triethylamine (0.03 ml, 0.2 mmole) in anhydrous dichloromethane
(2 ml) in argon was treated with a solution of 4- (2-benzyloxycarbonyl-vinyl) -phenyl ester of 8-isopropyl-2,2,4,4-tetramethyl-chroman-6-carboxylic acid (Intermediate 35, 0.063 g, 0.123 mmol) in dichloromethane (2 ml) and the resulting reaction mixture was stirred overnight at room temperature. The reaction mixture was quenched with water and extracted with diethyl ether. The organic extract was dried over anhydrous sodium sulfate, filtered and evaporated to a residue which was subjected to flash column chromatography to yield an intermediate which was treated with acetic acid (1 ml) in water (0.3 ml) and tetrahydrofuran (0.3 ml) at room temperature for lh. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic extract was dried over anhydrous sodium sulfate, filtered and evaporated to a residue which was subjected to preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase to give the title compound (0.007 g). NMR R1 (300 MHz, CDCl3): d 7.89 (d, 1H, J = 2Hz), 7.74 (d, 1H, J = 2Hz), 7.67 (d, 1H, «7 = 15.8Hz ), 7.49 (d, 2H, J = 8.8Hz), 7.15 (d, 2H, J = 8.8Hz), 6.32 (d, 1H, J = 15.8Hz), 3.20 (heptet, 1H, J = 6.8Hz), 1.77 (s, 2H), 1.29 (s, 6H), 1.28 (s, 6H), 1.12 (d, 6H, J = 6 , 8Hz).
United States No. 6,252,090
l. > - NH2 5NaCNBH3) CH2C12, CH3CN, CH3COOH 2. z'-Prl, K2C03, CH3COCH3 Intermediate 36 Intermediate 37
Intermediate 38 Intermediate 39
Reaction Scheme 9
Ethyl-2, 2,4,4-tetramethylchroman-6-carboxylate (Intermediate 36)
A solution of 6-bromo-2,2,2,4-tetramethylchroman (U.S. Patent No. 6,252,090, 2.2 g, 8.08 mmol), palladium acetate (0.145 g, 0.65 mmol) and 1,3-bis (diphenylphosphino) propane (0.267 g, 0.65 mmol) in a mixture of N, N-dimethylformamide (25 ml), ethanol (20 ml) and triethylamine (7 ml) was heated at 90 ° C. in an atmosphere of carbon monoxide at night. The volatiles were removed by vacuum distillation and the residue was diluted with water and extracted with ethyl acetate. The combined organic extract was washed with brine (xl), dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate. 5-10% ethyl in hexane as eluent to give the title compound (1.9 g, 90%). 1 H-NMR (300 MHz, CDCl 3): d 8.00 (d, 1H, J = 2.3Hz), 7.76 (dd, 1H, "7 = 2.1.8.5Hz), 6.79 (d , 1H, J "= 8.5Hz), 4.33 (c, 2H, J = 7.1Hz), 1.85 (s, 2H), 1.36 (s, 6H), 1.37 (s, 6H), 1.39-1.33 (m, 3H).
Ethyl ester of 8-formyl-2,2,4,4,4-tetramethylchroman-6-carboxylic acid (Intermediate 37)
A stirred, cooled solution (ice bath) of 2, 2,4,4-tetramethyl-chroman-6-carboxylic acid ethyl ester (Intermediate 36, 0.5 g, 1.92 mmol) in anhydrous dichloromethane (10 ml. ) was treated with titanium tetrachloride (0.4 ml, 3.26 mmol) followed by dichloromethyl ether (0.17 ml, 1.92 mmol). The reaction was allowed to warm to room temperature over 2 days, carefully quenched with ice and water and extracted with dichloromethane. The organic extract was washed with water and brine, dried over sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to flash column chromatography to provide the title compound (0.11 g, 20%) . H NMR (300 MHz, CDCl 3): d 10.46 (s, 1H), 8.33 (d, 1H, "7 = 2Hz), 8.20 (d, 1H, J = 2Hz), 4.36 ( c, 2H, "7 = 6.7Hz), 1.93 (s, 2H), 1.45 (s, 6H), 1.42 (s, 6H), 1.39 (t, 3H, J = 6 , 7Hz).
8- [(Cyclopropyl-isopropyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-carboxylic acid ethyl ester (Intermediate 38)
Following General Procedure C and using ethyl ester of 8-formyl-2, 2,4,4-tetramethyl-chroman-6-carboxylic acid (Intermediate 37, 0.11 g, 0.23 mmol) in dichloromethane (4 ml) and acetonitrile (2 ml), cyclopropylamine (0.08 ml, 1.1 mmol), acetic acid (0.8 ml) and sodium cyanoborohydride (0.072 g, 1.1 mmol) followed by working-up and flash column chromatography provided an intermediate. The intermediate
(0.122 g, 0.22 mmol) was dissolved in acetone (10 ml) and treated with potassium carbonate (0.153 g, 1.1 mmol) and isopropyl iodide (0.04 ml). The resulting reaction mixture was at 60 ° C for 4 h. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated to an oil. Flash column chromatography on silica gel (230-400 mesh) using 15-20% ethyl acetate in hexane as eluent afforded the title compound (0.09 g, 71%) as a clear oil. H1 NMR (300 MHz, CDC13): d 7.87 (d, 1H, J = 2.1 Hz), 7.85 (d,
1H, J = 2.1Hz), 4.35 (c, 2H, J "= 7.0Hz), 3.72 (s, 2H), 2.97
(heptet, 1H, J = 6.7Hz), 1.97 (m, 1H), 1.83 (s, 2H), 1.37 (t, 3H, J = 7.0Hz), 1.37 (s) , 6H), 1.35 (s, 6H), 1.08 (d,
6H, J = 6.7 Hz), 0.38-0.30 (m, 4H).
8- [(Cyclopropyl-isopropyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-carboxylic acid (Intermediate 39) A solution of 8- [(cyclopropyl-isopropyl-amino ) -methyl] -2, 2, 4, 4-tetramethylchroman-6-carboxylic acid (Intermediate 38, 0.09 g, 0.26 mmol) in ethanol (3 ml) and tetrahydrofuran (1 ml) was treated with a solution 1 M sodium hydroxide (3 ml, 3 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo, the residue was neutralized with dilute hydrochloric acid and extracted with ethyl acetate. The organic extract was washed with water and brine and dried over anhydrous sodium sulfate, filtered and evaporated to give the title compound (0.079 g, 96%). This was used as is for the next stage.
8- [(Cyclopropyl-isopropyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-carboxylic acid ester (Intermediate 40) 4-benzyloxycarbonylmethyl-phenyl ester
A solution of 8 - [(cyclopropyl-isopropyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-carboxylic acid (Intermediate 39, 0.079 g, 0.25 mmol) and benzyl acetate 4-hydroxy-phenyl (APIN, 0.06 g, 0.25 mmol) in anhydrous dichloromethane (5 ml) was treated with 4- (dimethylamino) pyridine (0.06 g, 0.5 mmol) and hydrochloride 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide (0.072 g, 0.37 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was then subjected to flash column chromatography using 20% ethyl acetate in hexane as eluent to give the title compound as an oil (0.093 g, 69%). H NMR (300 MHz, CDCl 3): d 7.98 (s, 2H), 7.32-7.21 (m, 7H), 7.13 (d, 2H, "7 = 8.5Hz), 5, 11 (s, 2H), 3.73 (s, 2H), 3.66 (s, '2H), 2.93 (heptet, 1H, J = 6.5Hz), 1.93 (m, 1H), 1.84 (s, 2H), 1.07 (s, 12H), 1.07 (d, 6H, J "= 6.5Hz).
Ester 4-carboxymethyl-phenyl 8- [(cyclopropyl-isopropyl-amino) -methyl] -2,2,4,4-tetramethyl-chroman-6-carboxylic acid ester (Compound 17)
A solution of 4-benzyloxycarbonylmethyl-phenyl ester of 8 - [(cyclopropyl-isopropyl-amino) -methyl] -2, 2,, 4-tetramethyl-chroman-6-carboxylic acid (Intermediate 40, 0.093 g, 0.17 mmol ) in ethyl acetate (3 ml) was treated with a suspension of 10% palladium on carbon (20 mg) in ethyl acetate and the resulting reaction mixture was stirred under a hydrogen atmosphere at room temperature for 2 h. The reaction mixture was filtered on a pad of celite and the filtrate was evaporated to a residue which was purified by flash column chromatography on silica gel to provide the title compound.
Pate 6,303,785 Intermediate 41
Intermediate 41 Intermediate 42
Intermediate 41 Intermediate 46
Intermediate 50 Intermediate 41 Cl2, Cul, NEt3 COOMe
Int 2. LÍOH int Int
Reaction Scheme 10 6-Bromo-2, 2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 41) A stirred, cooled solution (ice bath) of 6-bromo-2,2,4,4-tetramethylchroman -6-carbaldehyde (US Patent No. 6,303,785 incorporated herein by reference, 3.31 g, 11.15 mmol) in 2-methyl-2-propanol (30 ml) was treated with glacial acetic acid (30 ml). ml) followed by 2-methyl-2-butene (12 ml, 111.5 mmol). A solution of sodium chlorite (2.15 g, 18.95 mmol) in water (15 ml) was added dropwise to the reaction mixture. The reaction mixture was then allowed to warm gradually to room temperature and was stirred for 4 h, to which end it was basified with a 2 N solution of sodium hydroxide and then acidified with 2 N hydrochloric acid and extracted with ethyl acetate. . The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid (3.23 g, 93%). H NMR (300 MHz, CDCl3): d 8.14 (d, 1H, J = 2.7Hz), 7.60 (d, 1H, J = 2.7Hz), 1.95 (s, 2H), 1 50 (s, 6H), 1.39 (s, 6H).
6-Bromo-2,2,4,4-tetramethylchroman-8-carboxylic acid isopropyl ester (Intermediate 42)
A solution of 6-bromo-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 41, 0.3 g, 0.96 mmol) in anhydrous dichloromethane (15 ml) was treated with thionyl chloride (0.degree. , 7 mL, 9.6 mmol) and the reaction mixture was refluxed for 18 h. After cooling to room temperature, the volatiles were removed by vacuum distillation and the residue was dissolved in isopropanol (15 ml). 4- (Dimethylamino) pyridine (0.35 g, 9.6 mmol) was added and the reaction mixture was stirred at room temperature for 5 h. It was diluted with ethyl acetate and washed with 2N hydrochloric acid (x2), 2N sodium hydroxide (x2), and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the product of the product. title in the form of a brown oil (0.32 g, 93%). NMR H1 (300 MHz, CDCl3): d 7.60 (d, 1H, J = 2.4Hz), 7.45 (d, 1H, J "= 2.4Hz), 5.23 (heptet, 1H, J = 6.0Hz), 1.84 (s, 2H), 1.37 (s, 6H), 1.36 (s, 6H), 1.33 (d, 6H, J = 6.0Hz).
Isopropyl Ester of 2, 2, 4, 4-tetramethyl-6-trimethylsilanylethynylchroman-8-carboxylic acid (Intermediate
43)
Following General Procedure D and using isopropyl ester of 6-bromo-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 42, 0.32 g, 0.89 mmol), triethylamine (2 ml), iodide copper (I) (0.060 g, 0.33 mmol), trimethylsilylacetylene (0.5 ml, 3.56 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.16 g, 0.22 mmol) followed by chromatography in a flash column on silica gel (230-400 mesh) using 20% ethyl acetate in hexane as eluent, the title compound (0.23 g, 69%) was obtained as a yellow oil. H1 NMR (300 MHz, CDC13): d 7.58 (d, 1H, J = 2.4Hz), 7.44 (d, 1H, J = 2.4Hz), 5.18 (heptet, 1H, J = 6.3 Hz), 1.80 (s, 2H), 1.32 (s, 6H), 1.31 (s, 6H), 1.29 (d, 6H, J = 6.3Hz), 0.00 (s, 9H).
6-Ethynyl-2,4,4,4-tetramethylchromane-8-carboxylic acid isopropyl ester (Intermediate 44)
A solution of 2,2,4,4-tetramethyl-6-trimethylsilanylethynylchroman-8-carboxylic acid isopropyl ester (Intermediate 43, 0.23 g, 0.62 mmol) in methanol (5 ml) was treated with potassium carbonate (0.85 g, 6.2 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a brown oil which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate 20% in hexane as eluent to give the title compound (0.0246 g, 13%). NMR H1 (300 MHz, CDCl3): d 7.66 (d, 1H, J = 2.1 Hz), 7.53 (d, 1H, J "= 2.1Hz), 5.25 (heptet, 1H, J = 6.3 Hz), 3.02 (s, 1H), 1.88 (s, 2H), 1.40 (s, 6H), 1.37 (d, .6H, J = 6.3Hz), 1 , 36 (s, 6H).
6- (4-Methoxycarbonylmethyl-phenylethynyl) -2, 2,4, 4-tetramethyl-chroman-8-carboxylic acid isopropyl ester (Intermediate 45)
Following General Procedure B and using isopropyl ester of 6-ethynyl-2, 2, 4-tetramethylchroman-8-carboxylic acid (Intermediate 44 0.025 g, 0.08 mmol), methyl 4-iodophenylacetic acid ester (0.027 g, 0.1 mmol), triethylamine (2 ml), copper (I) iodide (0.008 g, 0.04 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.017 g, 0.024 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 40% ethyl acetate in hexane as eluent, the title compound was obtained as a yellow oil (0.019 g, 53%). H1 NMR (300 MHz, CDC13): d 7.68 (d, 1H, J = 2.1Hz), 7.55 (d, 1H, J = 2.1Hz), 7.49-7.24 (m, 4H), 5.25 (m, 1H), 3.70 (s, 3H), 3.64 (s, 2H), 1.88 (s, 2H), 1.39 (s, 6H), 1, 37 (s, 6H), 1.39-1.35 (d, 6H).
6- (4-Carboxymethyl-phenylethynyl) -2,2, -tetramethyl-chroman-8-carboxylic acid isopropyl ester (Compound 18)
A solution of 6- (4-methoxycarbonylmethyl-phenylethynyl) -2,2,4,4-tetramethyl-chroman-8-carboxylic acid isopropyl ester (Intermediate 45, 0.019 g, 0.043 mmol) in ethanol (0.3 ml) , tetrahydrofuran (0.3 ml) and water (0.3 ml) was treated with 1 N lithium hydroxide (0.086 ml, 0.086 mmol) and the resulting reaction mixture was stirred at room temperature for 30 minutes. The volatiles were evaporated in vacuo to a residue which was washed with hexane-ethyl acetate (3: 1), neutralized with 2 N hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a brown oil (0.015 g, 80%). NMR H1 (300 MHz, CDC13): d 7.69 (d, 1H, J "= 2.4Hz), 7.55 (d, 1H, J" = 2.4Hz), 7.50-7.26 ( m, 4H), 5.25 (heptet, 1H), 3.67 (s, 2H), 1.88 (s, 2H), 1.39 (s, 6H), 1.37 (s, 6H), 1.39-1.35 (d, 6H).
Ester 2, 2-dimethylpropyl of 6-bromo-2,2,4,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 46)
A cooled stirred solution (ice bath) of acid
6-bromo-2, 2,4, 4-tetramethylchroman-8-carboxylic acid (Intermediate 41, 0.5 g, 1.6 mmol), neopentyl alcohol (0.35 ml, 3.2 mmol) and 4- (dimethylamino) ) pyridine (0.03 g, 0.24 mmol) in anhydrous dichloromethane (5 ml) was treated with 1,3-dicyclohexylcarbodiimide (0.36 g, 1.76 mmol) and the reaction mixture was allowed to warm to room temperature ambient. After 2 h, the reaction mixture was filtered, the filtrate was diluted with ethyl acetate and washed with 2 N hydrochloric acid, 2 N sodium hydroxide, and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title product as a yellow solid (0.537 g, 88%). NMR H1 (300 MHz, CDC13): d 7.60 (d, 1H, J = 2.4Hz), 7.41 (d, 1H, J = 2.4Hz), 3.91 (s, 2H), 1 , 78 (s, 2H), 1.30 (s, 6H), 1.27 (s, 6H), 0.95 (s, 9H).
2, 2, 4,4-tetramethyl-6-trimethylsilanylethynylchroman-8-carboxylic acid ester 2 (2-dimethylpropyl) (Intermediate 47)
Following General Procedure D and using 2, 2-dimethylpropyl ester of 6-bromo-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 46, 0.54 g, 1.4 mmol), triethylamine (3 ml), copper iodide (I) (0.10 g, 0.52 mmol), trimethylsilylacetylene (0.8 ml, 5.6 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.25 g, 0, 35 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent, the title compound (0.396 g, 71%) was obtained as an oil. yellow color . NMR H1 (300 MHz, CDC13): d 7.64 (d, 1H, J = 1.8Hz), 7.46 (d, 1H, "7 = 1.8Hz), 3.94 (s, 2H), 1.81 (s, 2H), 1.33 (s, 6H), 1.30 (s, 6H), 0.98 (s, 9H), 0.002 (s, 9H).
Ester 2, 2-dimethylpropyl 6-ethynyl-2,4,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 48)
A solution of 2,2,4,4-tetramethyl-6-trimethylsilanylethynylchroman-8-carboxylic acid ester (Intermediate 47, 0.396 g, 1 mmol) in methanol (5 ml) was treated with potassium carbonate. (1.4 g, 10 mmol) and the resulting reaction mixture was stirred at room temperature for 30 minutes. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as a brown oil (0.227 g, 70%). NMR H1 (300 MHz, CDCl3): d 7.72 (d, 1H, J = 1.5Hz), 7.55 (d, 1H, "7 = 1.5Hz), 4.00 (s, 2H), 3.02 (s, 1H), 1.88 (s, 2H), 1.40 (s, 6H), 1.36 (s, 6H), 1.04 (s, 9H).
2- (4-Methoxycarbonylmethyl-phenylethynyl) -2,2,4,4-tetramethyl-chroman-8-carboxylic acid ester 2 (2-dimethylpropyl) (Intermediate 49)
Following General Procedure B and using 2, 2-dimethylpropyl ester of 6-ethynyl-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 48, 0.227 g, 0.70 mmol), methyl 4-methyl ester -iodophenylacetic acid (0.23 g, 0.83 mmol), triethylamine (3 ml), copper (I) iodide (0.07 g, 0.39 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.15 g) g, 0.21 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 20% ethyl acetate in hexane as eluent, the title compound was obtained as a yellow oil ( 0.198 g, 60%). NMR H1 (300 MHz, CDC13): d 7.76 (d, 1H, J = 2.4Hz), 7.58 (d, 1H, J = 2.4Hz), 7.49 (d, 2H, J = 8.1Hz), 7.26 (d, 2H, J = 8.1Hz), 4.01 (s, 2H), 3.70 (s, 3H), 3.64 (s, 2H), 1.88 (s, 2H), 1.40 (s, 6H), 1.38 (s, 6H), 1.05 (s, 9H).
Ester 2, 2-dimethylpropyl of 6- (4-carboxymethyl-phenylethynyl) -2,2,4,4-tetramethyl-chroman-8-carboxylic acid (Compound 19)
A solution of 2- (4-methoxycarbonylmethyl-phenylethynyl) -2, 2,4,4-tetramethyl-chroman-8-carboxylic acid ester, 2,4-dimethylpropyl ester (Intermediate 49, 0.198 g, 0.42 mmol) in ethanol (1 mL), tetrahydrofuran (1 mL) and water (1 mL) was treated with 1 N lithium hydroxide (1.5 mL, 1.5 mmol) and the resulting reaction mixture was stirred at room temperature for 30 minutes. . The reaction mixture was neutralized with 2 N hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the product of the txtule as a greenish-yellow solid (0.16 g, 84%). . NMR H1 (300 MHz, CDCl3): d 7.76 (d, 1H, J = 2.1Hz), 7.59 (d, 1H, J = 2.1Hz), 7.50 (d, 2H, J = 8.1Hz), 7.28 (d, 2H, J = 2.1Hz), 4.02 (s, 2H), 3.67 (s, 2H), 1.89 (s, 2H), 1.41 (s, 6H), 1.39 (s, 6H), 1.06 (s, 9H).
6-Bromo-2,2,4,4,4-tetramethylchroman-8-carboxylic acid t-butyl ester (Intermediate 50)
A solution of 6-bromo-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 41, 0.3 g, 0.96 mmol) and triethylamine (0.1 g, 0.96 mmol) in anhydrous tetrahydrofuran (3 ml) was treated with 2,4,6-trichlorobenzoyl chloride (0.23 g, 0.23 g), , 96 mmol) and the reaction mixture was allowed to stir for 20 minutes. The precipitated solid was separated by filtration and the filtrate was evaporated in vacuo to give a residue which was dissolved in benzene (3 ml) in argon and treated with 4- (dimethylamino) pyridine (0.47 g, 3.84 mmol ) and 2-methyl-2-propanol (0.14 g, 1.92 mmol). After 18 h, the reaction mixture was diluted with ethyl acetate and washed with 2 N hydrochloric acid, 2 N sodium hydroxide and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide a residue that was subjected to column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent to give the title product as a white solid (0.14 g, %). NMR H1 (300 MHz, CDCl3): d 7.54 (d, 1H, J = 2.4Hz), 7.43 (d, 1H, J = 2.4Hz), 1.84 (s, 2H), 1 , 58 (s, 9H), 1.37 (s, 6H), 1.33 (s, 6H).
2, 2, 4, 4-tetramethyl-6-trimethylsilanylethynylchroman-8-carboxylic acid t-butyl ester (Intermediate 51)
Following General Procedure D and using t-butyl ester of 6-bromo-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 50, 0.195 g, 0.53 mmol), triethylamine (2 ml), iodide copper (I) (0.040 g, 0.2 mmol), trimethylsilylacetylene (0.3 ml, 2.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.09 g, 0.13 mmol) followed by chromatography on a flash column on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent, the title compound (0.064 g, 32%) was obtained as a brown oil. H1 NMR (300 MHz, CDC13): d 7.57 (d, 1H, J = 2.1Hz), 7.46 (d, 1H, J = 2.1Hz), 1.84 (s, 2H), 1 , 57 (s, 9H), 1.37 (s, 6H), 1.34 (s, 6H), 0.045 (s, 9H).
6-Ethynyl-2,4,4-tetramethylchromane-8-carboxylic acid t-butyl ester (Intermediate 52)
A solution of 2,2,4,4-tetramethyl-6-trimethylsilanylethynylchroman-8-carboxylic acid t-butyl ester (Intermediate 51, 0.064 g, 0.17 mmol) in methanol (5 ml) was treated with potassium carbonate (0.23 g, 1.7 mmol) and the resulting reaction mixture was stirred at room temperature for 30 minutes. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title compound as a brown oil (0.051 g, 97%). NMR H1 (300 MHz, CDCl3): d 7.52 (d, 1H, J = 2.1Hz), 7.42 (d, 1H, "7 = 1.5Hz), 2.93 (s, 1H), 1.79 (s, 2H), 1.51 (s, 9H), 1.31 (s, 6H), 1.27 (s, 6H).
6- (4-Methoxycarbonylmethyl-phenylethynyl) -2,2,4, 4-tetramethyl-chroman-8-carboxylic acid t-butyl ester (Intermediate 53)
Following General Procedure B and using t-butyl ester of 6-ethynyl-2,2,4,4-tetramethylchroman-8-carboxylic acid (Intermediate 52, 0.051 g, 0.16 mmol), methyl 4-methyl ester -iodophenylacetic acid (0.053 g, 0.19 mmol), triethylamine (3 ml), copper (I) iodide (0.02 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.03 g, 0.043 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 20% ethyl acetate in hexane as eluent, the title compound was obtained as a yellow oil (0.014 g, %). NMR H1 (300 MHz, CDC13): d 7.63 (d, 1H, J = 2.1 Hz), 7.53 (d, 1H, J = 2.1 Hz), 7.48 (d, 2H, J = 8.2Hz), 7.26 (d, 2H, J = 8.2Hz), 3.72 (s, 3H), 3.65 (s, 2H), 1.88 (s, 2H), 1.60 (s, 9H), 1.40 (s, 6H), 1.38 (s, 6H).
6- (4-Carboxymethyl-phenylethynyl) -2,2,4,4-tetramethyl-chroman-8-carboxylic acid t-butyl ester (Compound 20)
A solution of 6- (4-methoxycarbonylmethyl-phenylethynyl) -2,2,4, 4-tetramethyl-chroman-8-carboxylic acid t-butyl ester (Intermediate 53, 0.014 g, 0.03 mmol) in ethanol (0 , 3 mL), tetrahydrofuran (0.3 mL) and water (0.3 mL) was treated with 1 N lithium hydroxide (0.12 mL, 0.12 mmol) and the resulting reaction mixture was stirred at the temperature environment for 3 h. The reaction mixture was neutralized with 2 N hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow oil (0.012 g, 88%). NMR H1 (300 MHz, CDCl3): d 7.63 (d, 1H, J = 2.1 Hz), 7.52 (d, 1H, J "= 2.1 Hz), 7.47 (d, 2H, J "= 8.2Hz), 7.26 (d, 2H, J" = 2.1Hz), 3.67 (s, 2H), 1.87 (s, 2H), 1.59 (s, 9H), 1.39 (s, 6H), 1.36 (s, 6H).
Reaction Scheme 11
- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-yl ester of trifluoromethanesulfonic acid (Intermediate 54)
A solution of 4,4-dimethyl-6-trifluoromethylsulfonyl-1, 2,3,4-tetrahydronaphthalen-1-one (U.S. Patent No. 6,252,090, 0.85 g, 2.64 mmoles) in dichloromethane (6 ml) and acetonitrile (3 ml) was treated with cyclopropylamine (3 ml.43.4 mmol). After 5 minutes, acetic acid (3 ml) was added followed by sodium cyanoborohydride (0.66 g, 10.55 mmol). The reaction was stirred overnight at room temperature. It was then diluted with water and a saturated aqueous solution of sodium carbonate and extracted with ethyl acetate. The combined organic extract was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil. The oil was dissolved in acetone (20 ml) and treated with potassium carbonate
(1.08 g, 7.8 mmol) and methyl iodide (1.6 mL, 26 mmol). The resulting reaction mixture was stirred at room temperature overnight. The solids were separated by filtration, the filtrate was evaporated in vacuo and the residue was subjected to column chromatography on silica gel (230-400 mesh) to give the title compound (0.85 g, 87%) in form of a colorless oil. NMR H1 (300 MHz, CDC13): d 7.61 (d, 1H, J ~ = 9.0Hz), 7.11 (d, 1H, J = 2.4Hz), 6.97 (dd, 1H, J = 2.4, 9.0Hz), 3.92 (t, 1H, J = 8.4Hz), 2.14-2.10 (m, 1H), 2.12 (s, 3H), 1.96. -1.89 (m, 2H), 1.79-1.57 (m, 2H), 1.29 (s, 3H), 1.25 (s, 3H), 0.52-0.36 (m , 4H).
General Procedure E: 5- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-carboxylic acid methyl ester (Intermediate 55)
A solution of 5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-yl ester of trifluoromethanesulfonic acid (Intermediate 54, 0.37 g, , 98 mmoles), palladium acetate (0.05 g, 0.22 mmol) and 1,3-bis (diphenylphosphino) propane (0.096 g, 0.23 mmol) in a mixture of dimethylformamide (4 ml), methanol ( 4 ml) and triethylamine (2 ml) was heated at 70 ° C in an atmosphere of carbon monoxide overnight. The volatiles were distilled in vacuo and the residue was diluted with water and extracted with diethyl ether (x3). The combined organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent, to provide the title compound (0.236 g, 85%). NMR H1 (300 MHz, CDCl3): d 7.96 (d, 1H, J = 1.8Hz), 7.73 (dd, 1H, J = 1.8.8.1Hz), 7.59 (d, 1H, J = 8.1Hz), 3.96 (t, 1H, J = 7.5Hz), 3.89 (s, 3H), 2.17-2.10 (m, 1H), 2.12 ( s, 3H), 1.98-1.83 (m, 2H), 1.82-1.60 (m, 2H), 1.34 (s, 3H), 1.28 (s, 3H), 0 , 54-0.39 (m, 4H).
- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydronaphthalene-2-carboxylic acid (Intermediate 56)
A solution of 5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydronaphthalene-2-carboxylic acid methyl ester (Intermediate 55, 0.236 g, 0.83 mmol) in methanol (4 ml) and tetrahydrofuran (4 ml) was treated with a 2 M solution of sodium hydroxide (4 ml, 8 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as a solid (0.22 g, 100%). H1 NMR (300 MHz, CDC13): d 7.98 (d, 1H, J = 1.8Hz), 7.72 (dd, 1H, J = 1.8.8.2Hz), 7.51 (d, 1H, J = 8.2Hz), 3.93 (t, 1H, J = 7.8Hz), 2.15-2.04 (m, 1H), 2.10 (s, 3H), 1.94- 1.85 (m, 2H), 1.79-1.62 (m, 2H), 1.27 (s, 3H), 1.22 (s, 3H), 0.52-0.40 (m, 4H). 5- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydronaphthalene-2-carboxylic acid azide (Intermediate 57)
A stirred, cooled solution (ice bath) of 5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydronaphthalene-2-carboxylic acid (Intermediate 56, 0.22 g, 0.83 mmoles) in anhydrous tetrahydrofuran (4 ml) was treated with triethylamine (0.16 ml, 1.1 mmol) followed by ethyl chloroformate (0.10 ml, 1.08 mmol). After 5 h, sodium azide (0.081 g, 1.24 mmol) was added and the reaction mixture was allowed to warm to room temperature and stirred overnight. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to give the title product which was used as such for the next reaction (0.24 g, 98%).
1- [5- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-yl] -3- (4-iodo-phenyl) -urea (Intermediate 58 )
A solution of 5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydronaphthalene-2-carboxylic acid azide (Intermediate 57, 0.12 g, 0.4 mmol) in Anhydrous toluene (14 ml) was refluxed in argon for 2 h. 4-iodoaniline (0.114 g, 0.52 mmol) was added and the solution was cooled to room temperature and stirred overnight. The volatiles were evaporated in vacuo and the residue was subjected to column chromatography on silica gel (230-400 mesh) using 20-25% ethyl acetate in hexane as eluent to give the title compound (0.13 g). , 67%). H1 NMR (300 MHz, CDC13): d 7.58 (d, 2H, J = 8.7Hz), 7.51 (d, 1H, J = 8.1Hz), 7.23 (d, 1H, J = 1.8Hz), 7.14 (d, 2H, J = 8.7Hz), 6.99 (dd, 1H, "7 = 1.8.8.1Hz), 6.99 (s wide, 1H), 6.57 (broad s, 1H), 3.92 (t, 1H, "= 7.2Hz), 2.13-2.05 (m, 1H), 2.13 (s, 3H), 1.93 -1.88 (m, 2H), 1.78-1.62 (m, 2H), 1.29 (s, 3H), 1.26 (s, 3H), 0.52-0.39 (m , 4H).
Methyl ester of 4- acid. { 3- [5- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-yl] -ureido} -benzoic (Intermediate 59)
Following General Procedure E and using 1- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-yl] -3- (4-iodo- phenyl) -urea (Intermediate 58, 0.13 g, 0.267 mmole), palladium acetate (0.02 g, 0.09 mmole), 1,3-bis (diphenylphosphino) propane (0.042 g, 0.101 mmole), N , N-dimethylformamide (3 ml), methanol (3 ml) and triethylamine (1 ml) followed by flash column chromatography on silica gel (230-400 mesh) using 30-40% ethyl acetate in hexane as eluent obtained the title compound (0.045 g, 40%). H1 NMR (300 MHz, CDC13): d 7.91 (d, 2H, J = 8.4Hz), 7.51 (d, 1H, J = 8.1Hz), 7.42 (s, 1H), 7 , 37 (d, 2H, J = 8.4Hz), 7.26 (d, 1H, «7 = 1.8Hz), 7.09 (s, 1H), 6.97 (dd, 1H, J" = 2.1.8.1Hz), 3.89 (s, 3H), 3.90-3.84 (m, 1H), 2.11-2.06 (m, 1H), 2.09 (s, 3H), 1.89-1.80 (m, 2H), 1.80-1.64 (m, 2H), 1.24 (s, 3H), 1.21 (s, 3H), 0.50 -0.36 (m, 4H).
Acid 4-. { 3- [5- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-yl] -ureido} -benzoic (Compound 21)
A solution of methyl ester of 4- acid. { 3- [5- (Cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-yl] -ureido} -benzoic (Intermediate 59, 0.045 g,
0.106 mmol) in methanol (2 ml) and tetrahydrofuran (3 ml) was treated with a 2 M solution of sodium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give a solid which was recrystallized from hot acetonitrile to give the title product as a white solid (0.012 g, 28%).
NMR R1 (300 MHz, CD30D): d 7.95 (d, 2H, J = 9.0 Hz), 7.53 (d,
2H, "7 = 9.0Hz), 7.46 (d, 1H, J = 2.1Hz), 7.40 (d, 1H, J =
8.7Hz), 7.09 (s, 1H), 7.19 (dd, 1H, J = 2.1.8.7Hz), 4.06
(t, 1H, J = 6.0Hz), 2.30-2.25 (m, 1H), 2.28 (s, 3H), 2.05- 1.98 (m, 2H), 1, 82-1.68 (m, 2H), 1.32 (s, 3H), 1.30 (s, 3H), 0.60-0.48 (m, 4H).
Intermediate 60
Intermediate 60 Intermediate 62 Intermediate 64
A1C13, CH2C12 Intermediate 65 -Irn * t.ermed Ji-o r6-7¡
Intermediate Intermediate 69
Compound 22 Reaction Scheme 12 4- (4-Bromo-2-methoxy-phenyl) -4-oxo-butyric acid ethyl ester (Intermediate 60)
A stirred, cooled (-30 ° C) solution of 3-bromoanisole (Aldrich, 18.7 g, 100 mmol) and ethylsuccinyl chloride (21 mL, 150 mmol) in anhydrous dichloromethane
(200 ml) was treated with aluminum chloride (26.6 g, 200 mmol) and the reaction mixture was allowed to warm to room temperature and stirred overnight. The reaction mixture was poured into water and extracted with dichloromethane (x2). The combined organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a brown oil. A solid separated upon standing. The liquid supernatant was decanted and the solid was washed with dichloromethane: hexane 1: 3 and dried to provide the title compound. The combined mother liquors and washings were evaporated to a brown oil which was subjected to column chromatography on silica gel (230-400 mesh) using 15% ethyl acetate in hexane as eluent to provide the title compound ( overall 12 g, 38%), and its isomer ethyl ester of 4- (2-chromosome-4-methoxyphenyl) -4-oxo-butyric acid (11.4 g, 36%) and a 1: 1 mixture of both ( 2 g, 6.3%). NMR H1 (300 MHz, CDCl3): d 7.56 (d, 1H, J = 8.7Hz), 7.07-7.03 (m, 2H), 4.07 (c, 2H, J = 7, 2Hz), 3.84 (s, 3H), 3.20 (t, 2H, J = 6.3Hz), 2.61 (t, 2H, J = 6.3Hz), 1.19 (t, 3H, J = 7.2 Hz).
Ethyl 4- (4-bromo-2-methoxy-phenyl) -butyric acid ester (Intermediate 61) A solution of 4- (4-bromo-2-methoxy-phenyl) -4-oxo-butyric acid ethyl ester ( Intermediate 60, 14.73 g, 46.8 mmol) in trifluoroacetic acid (72 ml, 935 mmol) was treated with triethylsilane (30 ml, 187 mmol) and the resulting reaction mixture was heated at 55 ° C for 4 h. The reaction mixture was then cooled to room temperature, neutralized with solid sodium bicarbonate, diluted with water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give a residue which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate to 8% in hexane as eluent to provide the title compound (7.4 g, 53%) as a colorless oil. H1NMR (300 MHz, CDCl3): d 7.02-6.94 (m, 3H), 4.11 (c, 2H, J = 7.2Hz), 3.79 (s, 3H), 2.60 (t, 2H, "7 = 7.2Hz), 2.29 (t, 2H, J = 7.2Hz), 1.88 (quinter, 2H, J = 7.2Hz), 1.25 (t, 3H) , J = 7.2 Hz).
- (4-Bromo-2-methoxy-phenyl) -2-methyl-pentan-2-ol (Intermediate 62)
A stirred, cooled solution (-10 ° C) of 4- (4-bromo-2-methoxy-phenyl) -butyric acid ethyl ester
(Intermediate 61, 7.4 g, 24.6 mmol) in anhydrous tetrahydrofuran (50 ml) was treated with a 3 M solution of methylmagnesium bromide (20.5 ml, 61.5 mmol) and the resulting reaction mixture was left tempering at room temperature for 3 hours. It was quenched with an aqueous solution of saturated ammonium chloride, diluted with water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title product (7.3 g, 100%). H1 NMR (300 MHz, CDCl3): d 6.92-6.87 (m, 3H), 3.71 (s, 3H), 2.48 (t, 2H, J = 7.2Hz), 1.55 -1.38 (m, 4H), 1.11 (s, 6H).
7-Bromo-5-methoxy-1,1-dimethyl-1,2,4,4-tetrahydro-naphthalene (Intermediate 63)
- (4-bromo-2-methoxy-phenyl) -2-methyl-pentan-2-ol
(Intermediate 62, 7.3 g, 24.6 mmol) was treated with 85% sulfuric acid (25 ml) at room temperature. After 30 minutes, the reaction mixture was diluted with cold water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product (5.6 g, 83%). NMR H1 (300 MHz, CDCl3): d 7.01 (d, 1H, "7 = 1.8Hz), 6.68 (d, 1H, J = 'l, 8Hz), 3.71 (s, 3H) , 2.49 (t, 2H, "7 = 6.3Hz), 1.71-1.65 (m, 2H), 1.55-1.51 (m, 2H), 1.18 (s, 6H) ).
6-Bromo-8-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 64)
A solution of 7-bromo-5-methoxy-1,1-dimethyl-1,2,3-tetrahydro-naphthalene (Intermediate 63, 5.6 g, 20.81 mmol) in glacial acetic acid (20 ml) was cooled to 0 ° C and treated with a solution of chromium trioxide (6.16 g, 61.6 mmol) in acetic acid and water (25 ml). The reaction mixture was then allowed to cool to room temperature and stirred for 48 h. It was diluted with water and extracted with diethyl ether (x2). The combined organic phase was washed with water (x3), saturated aqueous sodium bicarbonate (xl) and brine
(xl), dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide an oil. Flash column chromatography on silica gel (230-400 mesh) using 10-20-100% ethyl acetate in hexane as eluent afforded the title compound
(2 g, 33%) in the form of a yellow oil and recovered starting material (2.2 g, 39%). NMR H1 (300 MHz, CDCl3): d 7.12 (d, 1H, J = 1.8Hz), 6.97 (d, 1H, J = 1.8Hz), 3.87 (s, 3H), 2 , 66 (t, 2H, J = 6.6Hz), 1.92
(t, 2H, J - = 6.6Hz), 1.33 (s, 6H).
6-Bromo-8-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-l-one (Intermediate 65)
A stirred, cooled solution (ice bath) of 6-bromo-8-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 64, 0.24 g, 0.83 mmol) in anhydrous dichloromethane (4 ml) was treated with aluminum chloride (0.4 g, 3 mmol). The reaction mixture was allowed to warm to room temperature and stirred overnight. It was poured into water and extracted with dichloromethane and ethyl acetate. The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a brown oil which was subjected to column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent to provide the title product as a light yellow solid (0.13 g, 56%). H1 NMR (300 MHz, CDC13): d 12.85 (s, 1H), 7.00 (d, 1H, J = 1.5Hz), 6.98 (d, 1H, J = 1.5Hz), 2 , 74 (t, 2H, "7 = 6.9Hz), 1.96 (t, 2H," 7 = 6.9Hz), 1.36 (s, 6H).
8-Hydroxy-4,4-dimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalene-l-one (Intermediate 66)
Following General Procedure D and using 6-bromo-8-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 65, 1.56 g, 5.8 mmol), triethylamine (20 ml), copper iodide (I) (0.088 g, 0.46 mmol), trimethylsilylacetylene (3 ml, 21.22 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.325 g, 0.46 mmol) followed of flash column chromatography on silica gel (230-400 mesh) using hexane to 2-5% ethyl acetate in hexane as eluent, the title compound (1.67 g, 100%) was obtained in the form of a Solid orange. H NMR (300 MHz, CDC13): d 12.72 (s, 1H), 6.93 (d, 1H, J = 1.5Hz), 6.88 (d, 1H, J = 1.5Hz), 2 , 74 (t, 2H, J = 6.6Hz), 1.96 (t, 2H, J = 6.6Hz), 1.36 (s, 6H), 0.27 (s, 9H).
6-Ethynyl-8-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalene-lime (Intermediate 67)
A solution of 8-hydroxy-4,4-dimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 66, 2.2 g, 7.4 mmol) in methanol (20 ml) it was treated with potassium carbonate (2.04 g, 14.8 mmol) and the resulting reaction mixture was stirred at room temperature for 5 h. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as an oil (1.58 g, -100%).
NMR H1 (300 MHz, CDCl3): d 12.76 (s, 1H), 6.97 (d, 1H, J = 1.5Hz), 6.88 (d, 1H, J = 1.5Hz), 3 , 28 (s, 1H), 2.73 (t, 2H, "7 = 6.6Hz), 1.94 (t, 2H, J = 6.6Hz), 1.34 (s, 6H).
Methyl ester of acid. { 4- [8, 8-dimethyl-4-hydroxy-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate 68)
Following General Procedure B and using 6-ethynyl-8-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one
(Intermediate 67, 1.58 g, 7.4 mmol), 4-iodophenylacetic acid methyl ester (2.2 g, 7.94 mmol), triethylamine
(12 ml), copper iodide (I) (0.38 g, 1.99 mmol) and dichlorobis (triphenylphosphine) palladium (II) (1.2 g, 1.71 mmol) followed by flash column chromatography on gel of silica (230-400 mesh) using 16% ethyl acetate in hexane as eluent, the title compound was obtained as a yellow oil (2.1 g, 78%). NMR H1 (300 MHz, CDCl3): d 12.79 (s, 1H), 7.52 (d, 2H, "7 =
8.7Hz), 7.29 (d, 2H, J = 8.7Hz), 7.01 (d, 1H, J = 1.5Hz),
6.94 (d, 1H, J = 1.5Hz), 3.71 (m, 3H), 3.65 (s, 2H), 2.76
(t, 2H, J = 6.6Hz), 1.97 (t, 2H, J = 6.6Hz), 1.38 (s, 6H).
Methyl ester of acid. { 4- [8, 8-Dimethyl-5-oxo-4-trifluoromethanesulfonyloxy-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate 69)
A stirred, cooled (0 ° C) solution of (4- [8,8-dimethyl-4-hydroxy-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl}. -acetic (Intermediate 68, 2.1 g, 5.8 mmol) in anhydrous dichloromethane (20 ml) was treated with 4- (dimethylamino) pyridine (1.21 g, 9.9 mmol) followed by N-phenyltrifluoromethanesulfonimide (2). , 2 g, 6.16 mmol) After stirring at room temperature overnight, the reaction mixture was subjected to column chromatography on silica gel (230-400 mesh) using 20% ethyl acetate in hexane. as eluent to provide the title compound (2.6 g, 91%). H NMR (300 MHz, CDC13): d 7.57 (d, 1H, J = 1.2 Hz), 7.49 (d, 2H) , J = 8.4Hz), 7.27 (d, 2H, J = 8.4Hz), 7.19 (d, 1H, J = 1.2Hz), 3.66 (m, 3H), 3.62 (s, 2H), 2.72 (t, 2H, "7 = 6.9Hz), 1.99 (t, 2H, J = 6.9Hz), 1.38 (s, 6H).
Methyl ester of [4- (8,8-dimethyl-5-oxo-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -acetic acid (Intermediate 70)
A solution of acid methyl ester. { 4- [8,8-dimethyl-5-oxo-4-trifluoromethanesulfonyloxy-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate
69, 0.233 g, 0.47 mmol) in anhydrous 1-methyl-2-pyrrolidinone (3 ml) was purged with argon, and treated with lithium chloride (0.061 g, 1.45 mmol), tri-2-furylphosphine (0.0071 g, 0.031 mmol) and tris (dibenzylidene) dipalladium (0)
(0.007 g, 0.015 mmol). After 5 minutes, tributyl (vinyl) tin (0.175 g, 0.55 mmol) was added and the resulting reaction mixture was stirred at room temperature for 2.5 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The organic phase was washed with brine and water, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate 10-15% in hexane as eluent to give the title compound (0.15 g, 86%) as a white solid. NMR H1 (300 MHz, CDCl3): d 7.53 (d, 2H, J = 7.8Hz), 7.51 (d, 1H, J "= 1.8Hz), 7.50 (d, 1H, J = 1.8Hz), 7.43 (dd, 1H, J = 10.5, 17.1Hz), 7.29 (d, 2H, J = 7.8Hz), 5.57 (dd, 1H, J = 1.5, 17.1 Hz), 5.33 (dd, 1H, J = 1.5, 10.5Hz), 3.71 (s, 3H), 3.66 (s, 2H), 2.74 ( t, 2H, J = 6.9Hz), 2.00 (t, 2H, J = 6.9Hz), 1.40 (s, 6H).
[4- (8,8-Dimethyl-5-oxo-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -acetic acid (Compound 22)
A solution of [4- (8,8-dimethyl-5-oxo-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 70, 0.082 g, 0.22 mmol) in methanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 2M solution of lithium hydroxide (1.5 ml, 3 mmol) and the resulting reaction mixture was stirred to the room temperature during l, 5h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid (0.065 g, 82%). NMR H1 (300 MHz, CDCl3): d 7.53 (d, 2H, J = 8.1 Hz), 7.50 (s, 2H), 7.43 (dd, 1H, J = 10.8, 17, 4Hz), 7.31 (d, 2H, J = 8.1Hz), 5.57 (dd, 1H, J = 1.5, 17.4Hz), 5.33 (dd, 1H, J "= 1, 5, 10.8 Hz), 3.68 (s, 2 H), 2.74 (t, 2 H, J = 6.3 Hz), 1.99 (t, 2 H, J = 6.3 Hz), 1.39 ( s, 6H).
Methyl ester of acid. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acetic (Intermediate 71)
A solution of [4- (8,8-dimethyl-5-oxo-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 70, 0.205 g, 0.55 mmol) in dichloromethane (4 ml) and acetonitrile (2 ml) was treated with cyclopropylamine (1 ml, 14.45 mmol). After 5 minutes, acetic acid (1 ml) was added followed by sodium cyanoborohydride (0.138 g, 2.2 mmol). The reaction mixture was stirred overnight at room temperature. It was then diluted with water and a saturated aqueous solution of sodium bicarbonate and extracted with ethyl acetate. The combined organic extract was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil. The oil was dissolved in acetone (10 ml) and treated with potassium carbonate (0.227 g, 1.65 mmol) followed by methyl iodide (0.54 ml, 8.7 mmol) and the resulting reaction mixture was stirred overnight at room temperature. Diethyl ether was added to the reaction mixture and the precipitated solids were removed by filtration, the filtrate was evaporated in vacuo to a residue. Flash column chromatography on silica gel (230-400 mesh) using 4-5% ethyl acetate in hexane as eluent afforded the title compound (0.14 g, 60%). H NMR (300 MHz, CDCl 3): d 7.50 (d, 2H, J = 8.4Hz), 7.47 (s, 1H), 7.45 (s, 1H), 7.26 (d, 2H) , J = 8.4Hz), 7.13 (dd, 1H, J = 10.8, 17.7Hz), 5.47 (dd, 1H, J = 1.5, 17.7Hz), 5.11 ( dd, 1H, J = 1.5, 10.8Hz), 4.04 (t, 1H, J = 5.4Hz), 3.69 (s, 3H), 3.63 (s, 2H), 2, 18 (s, 3H), 2.18-2.14 (m, 1H), 2.02 (, 1H), 1.90-1.75 (m, 2H), 1.58-1.51 (m , 1H), 1.35 (s, 3H), 1.24 (s, 3H), 0.39-0.31 (m, 3H), 0.21-0.17 (m, 1H).
Acid { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acetic (Compound 23)
A solution of acid methyl ester. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-vinyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acetic (Intermediate 71, 0.14 g, 0.327 mmol) in methanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 2M solution of lithium hydroxide (1.5 ml, 3 mmol) and the mixture of The resulting reaction was stirred at room temperature for 2 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title product as a white solid (0.135 g, 96%). NMR H1 (300 MHz, CDC13): d 9.99 (broad s, 1H), 7.47 (d, 2H, J = 8.1Hz), 7.44 (s, 1H), 7.43 (s, 1H), 7.22 (d, 2H, J = 8.1Hz), 7.11 (dd, 1H, J = 10.8, 17.1Hz), 5.47 (dd, 1H, J = 0.9 , 17.1 Hz), 5.11 (dd, 1H, J = 0.9, 10.8Hz), 4.06 (t, 1H, J = 6.0Hz), 3.55 (s, 2H), 2 , 18 (s, 3H), 2.18-2.15 (, 1H), 2.04 (m, 1H), 1.91-1.77 (m, 2H), 1.56-1.50 ( m, 1H), 1.34 (s, 3H), 1.22 (s, 3H), 0.42-0.29 (m, 3H), 0.28-0.21 (m, 1H).
Intermediate 66 Intermediate 73
SEM = CH2OCH2CH2SiMe3
Intermediate 76 Compound 24
Reaction Scheme 13 4, 4-Dimethyl-8- (2-trimethylsilanyl-ethoxymethoxy) -6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 72)
A solution of 8-hydroxy-4,4-dimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 66, 1.67 g, 5.8 mmol) in anhydrous benzene was treated with triethylamine (1.41 g, 11.6 mmol) and a catalytic amount of 4- (dimethylamino) pyridine followed by 2- (trimethylsilyl) ethoxymethyl chloride (1.93 g, 11.6 mmol) and the reaction mixture The resultant was refluxed for 3 days. This was cooled to room temperature, diluted with water and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate 2-6% in hexane as eluent to provide the title product as a yellow oil (1.58 g, 66%). H NMR (300 MHz, CDCl 3): d 7.16 (d, 1H, J = 1.2 Hz), 7.12 (d, 1 H, J = 1.2 Hz), 5.28 (s, 2 H), 3 , 81 (m, 2H), 2.68 (t, 2H, J = 6.9Hz), 1.94 (t, 2H, J = 6.9Hz), 1.34 (s, 6H), 0.96 (m, 2H), 0.27 (s, 9H), 0.00 (s, 9H).
6-Ethinyl-4,4-dimethyl-8- (2-trimethylsilanyl-ethoxymethoxy) -3,4-dihydro-2H-naphthalene-1-one (Intermediate 73)
A solution of 4,4-dimethyl-8- (2-trimethylsilanyl-ethoxymethoxy) -6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 72, 1.58 g, 3.79 mmol) in methanol (20 ml) was treated with potassium carbonate (0.43 g, 3.11 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (1.28 g, 98%). NMR H1 (300 MHz, CDC13): d 7.19 (d, 1H, "7 = 1.2Hz), 7.15 (d, 1H, J = 1.2Hz), 5.26 (s, 2H), 3.79 (m, 2H), 3.19 (s, 1H), 2.67 (t, 2H, J = 6.6Hz), 1.94 (t, 2H, J = 6.6Hz), 1, 33 (s, 6H), 0.95 (m, 2H), -0.016 (s, 9H).
Methyl ester of (4- [8,8-dimethyl-5-oxo-4- (2-trimethylsilanyl-ethoxymethoxy) -5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} - acetic (Intermediate 74)
Following General Procedure B and using 6-ethynyl-4, -dimethyl-8- (2-trimethylsilanyl-ethoxymethoxy) -3,4-dihydro-2H-naphthalen-1-one (Intermediate 73, 1.28 g, 3, 7 mmoles), 4-iodophenylacetic acid methyl ester (1.02 g, 3.7 mmol), triethylamine (30 ml), copper (I) iodide (0.095 g, 0.5 mmol) and dichlorobis (triphenylphosphine) palladium
(II) (0.35 g, 0.5 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-15% ethyl acetate in hexane as eluent, the title compound was obtained in the form of a yellow oil (1.61 g, 88%). NMR H1 (300 MHz, CDC13): d 7.51 (d, 2H, J = 8.1 Hz), 7.28 (d, 2H, J = 8.1 Hz), 7.24 (d, 1H, J = 1.5Hz), 7.19 (d, 1H, J = 1.5Hz), 5.31 (s, 2H), 3.82 (m, 2H), 3.70 (s, 3H), 3.65 (s, 2H), 2.69 (t, 2H, J = 6.6Hz), 1.96 (t, 2H, J "= 6.6Hz), 1.37 (s, 6H), 0.97 ( m, 2H), 0.00 (s, 9H).
Methyl ester of (4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4- (2-trimethylsilanyl-ethoxymethoxy) -5,6,7,8-tetrahydro-naphthalene-2-ethynyl] -phenyl.}. -acetic (Intermediate 75)
A solution of acid methyl ester. { 4- [8, 8-dimethyl-5-oxo-4- (2-trimethylsilanyl-ethoxymethoxy) -5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate 74, 0.905 g, 1.84 mmol) in dichloromethane (8 ml) and acetonitrile (4 ml) was treated with cyclopropylamine (4 ml, 57.8 mmol). After 5 minutes, acetic acid (4 ml) was added followed by sodium cyanoborohydride (0.46 g, 7.32 mmol). The reaction mixture was stirred overnight at room temperature. It was then diluted with water and a saturated aqueous solution of sodium carbonate and extracted with dichloromethane (x2). The combined organic extract was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil. The oil dissolved in acetone
(15 ml) and treated with potassium carbonate (0.745 g, 5.4 mmol) followed by methyl iodide (1.2 ml, 19 mmol) and the resulting reaction mixture was stirred overnight at room temperature. The precipitated solids were separated by filtration, the filtrate was evaporated in vacuo to a residue. Flash column chromatography on silica gel (230-400 mesh) using 2-20% ethyl acetate in hexane as eluent afforded the title compound (0.6 g, 63%). NMR H1 (300 MHz, CDCl3): d 7.49 (d, 2H, J = 8.4Hz), 7.23 (d, 2H, J = 8.4Hz), 7.18 (d, 1H, J = 1.5Hz), 7.06 (d, 1H, J = 1.5Hz), 5.21 (s, 2H), 4.03 (m, 1H), 3.76 (m, 2H), 3.68 (s, 3H), 3.62 (s, 2H), 2.30 (s, 3H), 2.04-1.40 (m, 5H), 1.33 (s, 3H), 1.18 ( s, 3H), 0.97 (m, 2H), 0.26-0.01 (m, 4H), 0.00 (s, 9H).
Methyl ester of 4- [5- (cyclopropyl-methyl-amino) -4-hydroxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acetic (Intermediate 76) A solution of acid methyl ester. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4- (2-trimethylsilanyl-ethoxymethoxy) -5,6,7,8-tetrahydro-naphthalene-2-ylethynyl] -phenyl ester} -acetic (Intermediate 75, 0.37 g, 0.73 mmol) in tetrahydrofuran (12 ml) was treated with 2% sulfuric acid in methanol (14 ml) and the resulting reaction mixture was stirred at room temperature during the night. It was neutralized with a saturated solution of sodium bicarbonate and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which after flash column chromatography on silica gel (230-400 mesh) using 5-20% ethyl acetate in hexane as The eluent afforded the title product as a white solid (0.295 g, 87%). H1 NMR (300 MHz, CDC13): d 12.26 (s, 1H), 7.45 (d, 2H, J = 8.4Hz), 7.22 (d, 2H, J = 8.4Hz), 6 , 96 (d, 1H, J = 1.5Hz), 6.69 (d, 1H, J = 1.5Hz), 4.31 (m, 1H), 3.67 (s, 3H), 3.61 (s, 2H), 2.23 (s, 3H), 2.23-2.17 (m, 1H), 2.05-1.97 (m, 2H), 1.71-1.65 (m , 2H), 1.28 (s, 3H), 1.24 (s, 3H), 0.80-0.45 (m, 4H).
Methyl ester of 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-trifluoromethanesulfonyloxy-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl) -acetic acid ester (Intermediate 77)
A stirred, cooled (0 ° C) solution of 4- [5- (cyclopropyl-methyl-amino) -4-hydroxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene- methyl ester 2-iletinyl] -phenyl} -acetic (Intermediate 76, 0.15 g, 0.275 mmol) in anhydrous dichloromethane was treated with 4- (dimethylamino) pyridine (0.067 g, 0.55 mmol) followed by N-phenyltrifluoromethanesulfonimide (0.147 g, 0.413 mmol). After stirring at room temperature overnight, the reaction mixture was subjected to column chromatography on silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent to give the title compound (0.14 g, 93%). H NMR (300 MHz, CDCl 3): d 7.51 (d, 2H, J = 8.4Hz), 7.30-7.26 (m, 3H), 7.17 (d, 1H, J = 1, 5Hz), 4.04 (m, 1H), 3.72 (s, 3H), 3.66 (s, 2H), 2.37 (s, 3H), 2.25-2.17 (m, 1H) ), 2.09-1.74 (m, 3H), 1.59-1.52 (m, 1H), 1.40 (s, 3H), 1.23 (s, 3H), 0.28- 0.10 (m, 3H), 0.09-0.005 (m, 1H).
Ethyl ester of 8- (cyclopropyl-methyl-amino) -3- (4-methoxycarbonylmethyl-phenylethynyl) -5,5-dimethyl-5,6,7,8-tetrahydro-naphthalene-1-carboxylic acid (Intermediate 78)
Following the General Procedure? and using 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-trifluoromethanesulfonyloxy-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acetic (Intermediate 77, 0.14 g, 0.26 mmol), palladium acetate (0.013 g, 0.06 mmol), 1,3-bis (diphenylphosphino) propane (0.025 g, 0.061 mmol), N, N dimethylformamide (4 ml), ethanol (1.5 ml) and triethylamine (1.5 ml) followed by flash column chromatography on silica gel (230-400 mesh) using 7-10% ethyl acetate in hexane as eluent, the title compound was obtained (0.09 g, 75%). H NMR (300 MHz, CDCl 3): d 7.49 (d, 1 H, J = 1.8 Hz), 7.47 (d, 2 H, J = 8.1 Hz), 7.30 (d, 1 H, J = 1.8Hz), 7.25 (d, 2H, J = 8.1Hz), 4.33 (m, 1H), 4.28-4.13 (m, 2H), 3.70 (s, 3H) , 3.63 (s, 2H), 2.06-1.93 (2m, 6H), 1.72-1.66 (m, 2H), 1.36 (t, 3H, «7 = 7.2Hz ), 1.31 (s, 3H), 1.29 (s, 3H), 0.60-0.40 (m, 1H), 0.40-0.25 (m, 2H), 0.15- 0.00 (m, 1H).
Ethyl 3- (4-carboxymethyl-phenylethynyl) -8- (cyclopropyl-methyl-amino) -5,5-dimethyl-5,6,7,8-tetrahydro-naphthalene-1-carboxylic acid ester (Compound 24)
A solution of 8- (cyclopropyl-methyl-amino) -3- (4-methoxycarbonylmethyl-phenylethynyl) -5,5-dimethyl-5,6,7,8-tetrahydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 78, 0.09 g, 0.19 mmol) in ethanol (2 ml), tetrahydrofuran (3 ml) and water (1.5 ml) was treated with lithium hydroxide (0.11 g, 2.62 mmol) and The resulting reaction mixture was stirred at room temperature for 2 h. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the product of the tittle in the form of a white solid (0.085 g, 94%). NMR H1 (300 MHz, CDCl3): d 7.49 (d, 1H, J = 1.8Hz), 7.46 (d, 2H, "7 = 8.1Hz), 7.30 (d, 1H, J = 1.8Hz), 7.22 (d, 2H, J = 8.1Hz), 4.32 (, 1H), 4.30-4.10 (m, 2H), 3.58 (s, 2H) , 2.06-1.93 (2m, 6H), 1.72-1.65 (m, 2H), 1.35 (t, 3H, J = 7.0Hz), 1.34 (s, 3H) , 1.29 (s, 3H), 0.60-0.40 (m, 1H), 0.40-0.25 (m, 2H), 0.15-0.00 (m, 1H).
I,
Int Intermediate 79
I
and
Diagram of Reaction 14
2 - . 2-Bromo-5 - (cyclopropyl-methyl-amino) -8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphne (Intermediate 79)
A solution of 6-bromo-8-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 64, 1.08 g, 3.81 mmol) in dichloromethane (8 ml) and acetonitrile (4 ml) was treated with cyclopropylamine (5 ml, 72.3 mmol). After 5 minutes, acetic acid (5 ml) was added followed by sodium cyanoborohydride (0.96 g, 15.26 mmol). The reaction mixture was stirred for 2 days at room temperature. It was then diluted with water and a saturated aqueous solution of sodium carbonate and extracted with ethyl acetate. The combined organic extract was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil. The oil was dissolved in acetone (20 ml) and treated with potassium carbonate (1.58 g, 11.43 mmol) followed by methyl iodide (2.1 ml, 33 mmol) and the resulting reaction mixture was stirred overnight at room temperature. Diethyl ether was added and the precipitated solids were removed by filtration, the filtrate was evaporated in vacuo to a residue. Flash column chromatography on silica gel
(230-400 mesh) using 2.5-10% ethyl acetate in hexane as eluent afforded the title compound
(1.08 g, 84%). NMR H1 (300 MHz, CDC13): d 7.08 (d, 1H, J = 1.8Hz), 6.78 (d, 1H, J = 1.8Hz), 3.97 (m, 1H), 3 , 79 (s, 3H), 2.30 (s, 3H),
2.04-1.82 (m, 3H), 1.65-1.27 (m, 2H), 1.30 (s, 3H), 1.16
(s, 3H), 0.30-0.22 (m, 2H), 0.07-0.00 (m, 2H).
- (Cyclopropyl-methyl-amino) -8,8-dimethyl-4-methoxy-2-trimethylsilanylethynyl-5,6,7,8-tetrahydro-naphthalene (Intermediate 80)
Following General Procedure D and using 2-bromo-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 79, 1.08 g , 3.2 mmol), triethylamine (5 ml), copper iodide (I) (0.061 g, 0.32 mmol), tri-ethylsilylacetylene (3 ml, 21.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) ( 0.225 g, 0.32 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using hexane-10% ethyl acetate in hexane as eluent, the title compound was obtained (0.87 g, 80%).
H1 NMR (300 MHz, CDC13): d 7.09 (d, 1H, J = 1.5Hz), 6.73 (d,
1H, J = 1.5Hz), 3.99 (m, 1H), 3.79 (s, 3H), 2.28 (s, 3H),
2.02-1.80 (m, 3H), 1.65-1.26 (2m, 2H), 1.31 (s, 3H), 1.16
(s, 3H), 0.26 (s, 9H), 0.26-0.00 (m, 2H), 0.00- -0.01 (, 2H).
- (Cyclopropyl-methyl-amino) -2-ethynyl-8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 81)
A solution of 5- (cyclopropyl-methyl-amino) -8,8-dimethyl-4-methoxy-2-trimethylsilanylethynyl-5,6,7,8-tetrahydro-naphthalene (Intermediate 80, 0.87 g, 2.45 mmoles) in methanol (20 ml) was treated with potassium carbonate (0.4 g, 2.89 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (0.635 g, 92%). H1 NMR (300 MHz, CDC13): d 7.16 (d, 1H, J = 1.4Hz), 6.79 (d, 1H, J = 1.4Hz), 4.04 (, 1H), 3, 82 (s, 3H), 2.32 (s, 3H), 2.03-1.95 (m, 2H), 1.90-1.80 (m, 1H), 1.70-1.55 (s, m, 1H), 1.45-1.35 (m, 1H), 1.34 (s, 3H), 1.19 (s, 3H), 0.40-0.20 (m, 2H), 0 , 07-0.00 (m, 2H).
Methyl ester of (4- [5- (Cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} - acetic (Intermediate 82)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethynyl-8, 8-dimethyl-4-methoxy-5, 6, 7, 8-tetrahydro-naphthalene (Intermediate 81, 0.065 g, , 23 mmoles), methyl 4-iodophenylacetate (0.063 g, 0.23 mmol), triethylamine (8 ml), copper (I) iodide (0.018 g, 0.093 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.065) g, 0.093 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-20% ethyl acetate in hexane as eluent, the title compound was obtained as a yellow solid ( 0.09 g, 90%). H1 NMR (300 MHz, CDC13): 7.50 (d, J = 8.2 Hz, 2 H), 7.26 (d, J = 8.2 Hz, 2 H), 7.17 (d, J "= 1, 2Hz, 1H), 6.81 (d, J = 1, 2Hz, 1H), 4.04 (s broad, 1H), 3.82 (s, 3H), 3.70 (s, 3H), 3, 64 (s, 2H), 2.32 (s, 3H), 2.05-1.94 (m, 2H), 1.90-1.80 (m 1H), 1.70-1.58 (m , 1H), 1.45-1.35 (m, 1H), 1.38 (s, 3H), 1.20 (s, 3H), 0.38-0.20 (m, 2H), 0, 18-0.02 (m, 2H).
Acid { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Compound 25)
A solution of acid methyl ester. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acetic (Intermediate 82, 0.090 g, 0.208 mmol) in methanol (3 ml) and tetrahydrofuran (2 ml) was treated with a 1.9 M lithium hydroxide solution (1.5 ml, 2.8 mmol) and the The resulting reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated, neutralized with ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using methanol at room temperature. -10% in ethyl acetate as eluent to give the title product as a white amorphous solid (0.062 g, 60%). H1 NMR (300 MHz, CDC13): 7.46 (d, J = 8.2 Hz, 2 H), 7.28 (d, J = 8.2 Hz, 2 H), 7.18 (d, J = 1, 2 Hz) , 1H), 6.81 (d, "7 = 1, 2Hz, 1H), 4.27 (s, 2H), 3.81 (s, 3H), 3.58 (s, 2H), 2, 42 (s, 3H), 2.28-2.18 (m, 1H), 2.15-1.88 (m 2H), 1.75-1.65 (m, 1H), 1.45-1 , 38 (m, 1H), 1.32 (s, 3H), 1.17 (s, 3H), 0.75-0.65 (m, 1H), 0.55-0.42 (m, 2H) ), 0.25-0.15 (m, 1H).
Methyl ester of acid. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-ylethynyl] -2-fluoro-phenyl} -acetic (Intermediate 83)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethynyl-8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 81, 0.085 g, , 3 mmoles), methyl 2-fluoro-4-iodophenylacetate (0.088 g, 0.3 mmol), triethylamine (8 ml), copper iodide (I)
(0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using ethyl acetate 5-20% in hexane as eluent, the title compound was obtained as a yellow solid (0.12 g, 89%). NMR H1 (300 MHz, CDCl3): 7.36-7.17 (m, 4H), 6.81 (d, J = 1, 2Hz, 1H), 4.12 (broad s, 1H), 3.83 (s, 3H), 3.72 (s, 3H), 3.69 (s, 2H), 2.33 (s, 3H), 2.08-1.98 (m 2H), 1.98-1 , 88 (m, 1H), 1.75-1.60 (m, 1H), 1.45-1.35 (m, 1H), 1.35 (s, 3H), 1.19 (s, 3H) ), 0.35-0.25 (m, 2H), 0.15-0.05 (m, 1H).
Acid { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-ylethynyl] -2-fluoro-phenyl} -acetic (Compound 26)
A solution of acid methyl ester. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-ylethynyl] -2-fluoro-phenyl} -acetic (Intermediate 83, 0.12 g, 0.27 mmol) in methanol (4 ml) and tetrahydrofuran (4 ml) was treated with a 2 M solution of lithium hydroxide (2 ml, 4 mmol) and the mixture of The resulting reaction was stirred at room temperature for 2 h. The reaction mixture was concentrated, neutralized with ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using methanol at room temperature. -8% in ethyl acetate as eluent to give the title product as a white amorphous solid (0.041 g, 35%). H1 NMR (300 MHz, CDC13): 7.35-7.15 (m, 4H), 6.81 (d, "7 = 1.2Hz, 1H), 4.31 (broad s, 1H), 3, 82 (s, 3H), 3.64 (s, 2H), 2.46 (s, 3H), 2.32-2.22 (m, 1H), 2.18-1.88 (m 2H), 1.78-1.65 (m, 1H), 1.50-1.40 (m, 1H), 1.32 (s, 3H), 1.17 (s, 3H), 0.80-0, 70 (m, 1H), 0.58-0.40 (m, 2H), 0.28-0.18 (m, 1H).
Methyl ester of acid 2-. { 4- [5- (Cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl) -propionic acid (Intermediate 84) Following the General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethynyl-8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 81, 0.085 g, 0.30 mmoles), methyl 2- (4-iodophenyl) propionate (Reagent 1, 0.087 g, 0.3 mmol), triethylamine (8 ml), copper iodide (I) (0.019 g, 0.1 mmol) and dichlorobis ( triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-20% ethyl acetate in hexane as eluent, the compound of the title in the form of a yellow solid (0.115 g, 86%). H1 NMR (300 MHz, CDC13): d 7.50 (d, 2H, J = 8.1 Hz), 7.28 (d, 2H, "J = 8.1 Hz), 7.16 (d, 1H," 7 = 1.2Hz), 6.81 (d, 1H, J = 1.2Hz), 4.04 (m, 1H), 3.83 (s, 3H), 3.74 (c, 1H, J = 6.9Hz), 3.67 (s, 3H), 2.31 (s, 3H), 2.03-1.98 (m, 2H), 1.89-1.83 (, 1H), 1, 68-1.59 (m, 1H), 1.51 (d, 3H, J = 6.9Hz), 1.42-1.27 (m, 1H), 1.35 (s, 3H), 1, 20 (s, 3H), 0.31-0.23 (m, 2H), 0.07-0.008 (m, 2H).
Acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -propionic (Compound 27)
A solution of 2- methyl acid ester. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -propionic (Intermediate 84, 0.115 g, 0.26 mmol) in methanol (3 ml) and tetrahydrofuran (2 ml) was treated with a 3 M solution of potassium hydroxide (1 ml)., 3 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using methanol at room temperature. % in ethyl acetate as eluent to give the title product as a yellow solid (0.062 g, 56%). H1 NMR (300 MHz, CDC13): d 7.50 (d, 2H, "7 = 8.1Hz), 7.32 (d,
2H, "7 = 8.1Hz), 7.17 (s, 1H), 6.80 (s, 1H), 4.23 (m, 1H),
3.80 (s, 3H), 3.68 (c, 1H, "7 = 7.2Hz), 2.38 (s, 3H), 2.22-2.18 (m, 1H), 2.07 -1.87 (m, 2H), 1.70-1.57 (m, 1H), 1.47
(d, 3H, "7 = 7.2Hz), 1.38-1.27 (m, 1H), 1.31 (s, 3H), 1.16 (s, 3H), 0.65-0, 62 (m, 1H), 0.41-0.35 (m, 2H), 0.17-0.00 (m, 1H).
Methyl ester of acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -2-methyl-propionic (Intermediate 85)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethynyl-8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 81, 0.090 g, , 32 mmol), methyl 2- (4-iodophenyl) -2-methyl-propionate (Reagent 2, 0.097 g, 0.3 mmol), triethylamine (8 ml), copper (I) iodide (0.019 g, , 1 mmole) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-20% ethyl acetate in hexane as eluent, the title compound was obtained as a solid (0.09 g, 78%). H1 NMR (300 MHz, CDC13): d 7.50 (d, 2H, "7 = 8.1Hz), 7.31 (d, 2H," 7 = 8.1Hz), 7.16 (d, 1H, J = 1.2Hz), 6.80 (d, 1H, «7 = 1.2Hz), 4.03 (m, 1H), 3.83 (s, 3H), 3.66 (s, 3H), 2.31 (s, 3H), 2.01-1.97 (m, 2H), 1.89-1.83 (, 1H), 1.68-1.59 (m, 1H), 1.59 (s, 6H), 1.42-1.27 (m, 1H), 1.34 (s, 3H), 1.20 (s, 3H), 0.31-0.22 (m, 2H), 0.07-0.00 (m, 2H).
Acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -2-methyl-propionic (Compound 28)
A solution of 2- methyl acid ester. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} -2-methyl-propionic (Intermediate 85, 0.09 g, 0.196 mmol) in methanol (3 mL) and tetrahydrofuran (2 mL) was treated with a 3 M solution of potassium hydroxide (1.5 mL, 4.5 mmoles) and the resulting reaction mixture was stirred at room temperature for 3 days. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using methanol at room temperature. % in ethyl acetate as eluent to give the title product as a yellow solid (0.057 g, 65%). H1 NMR (300 MHz, CDC13): d 7.46 (d, 2H, "7 = 8.4Hz), 7.37 (d, 2H," 7 = 8.4Hz), 7.18 (d, 1H, «7 = 1.2Hz), 6.81 (d, 1H,« 7 = 1.2Hz), 4.22 (m, 1H), 3.83 (s, 3H), 2.38 (s, 3H) , 2.19-1.90 (m, 3H), 1.71-1.56 (m, 1H), 1.56 (s, 6H), 1.45-1.33 (m, 1H), 1 , 33 (s, 3H), 1.17 (s, 3H), 0.70-0.50 (m, 1H), 0.38-0.25 (m, 2H), 0.16-0.00 (m, 1H).
Intermediate 81 Reagent 4 R, = H R¡ > = Me ",. p _ -u -, P ^, P ^ Compound 5 - hl Reagent 7 R5 = Me R8 = Et _ ^ v _ Mp, 0 d Compound K-5- e 2. KOH Reaction Scheme 15
Methyl ester of (E) -3- acid. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acrylic (Intermediate 86)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethynyl-8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 81, 0.095 g, 0.336 mmoles), methyl 4-iodocinnamate (Reagent 4, 0.097 g, 0.336 mmol), triethylamine (8 ml), copper (I) iodide (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) ( 0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-15% ethyl acetate in hexane as eluent, the title compound was obtained in the form of a solid white (0.12 g, 80%). H NMR (300 MHz, CDCl 3): d 7.68 (d, 1H, J = 15.9Hz), 7.53 (cAb, 4H, J = 8.4Hz), 7.19 (s, 1H), 6 , 83 (s, 1H), 7.46 (d, 1H, "7 = 15.9Hz), 4.04 (m, 1H), 3.84 (s, 3H), 3.82 (s, 3H) , 2.32 (s, 3H), 2.04-1.97 (m, 2H), 1.90-1.83 (m, 1H), 1.68-1.60 (m, 1H), 1 , 43-1.27 (m, 1H), 1.36 (s, 3H), 1.21 (s, 3H), 0.32-0.23 (m, 2H), 0.08-0.00 (m, 2H).
Acid (E) -3-. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} Acrylic acid (Compound 29)
A solution of methyl ester of (E) -3- acid. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} Acrylic (Intermediate 86, 0.12 g, 0.27 mmol) in methanol (4 ml) and tetrahydrofuran (3 ml) was treated with a 3 M solution of potassium hydroxide (1 ml, 3 mmol) and the mixture of The resulting reaction was stirred at room temperature overnight. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using methanol at room temperature. % in ethyl acetate as eluent to give the title product as a white solid (0.041 g, 35%). NMR H1 (300 MHz, CDC13): d 7.58 (d, 1H, "7 = 16.2Hz), 7.44 (cAb, 4H), 7.13 (s, 1H), 6.77 (s, 1H), 7.45 (d, 1H, J = 16.2Hz), 4.05 (m, 1H), 3.79 (s, 3H), 2.42 (s, 3H), 2.19-1 , 97 (m, 2H), 1.67-1.45 (m, 1H), 1.45-1.37 (m, 1H), 1.37-1.20 (m, 1H), 1.30 (s, 3H), 1.12 (s, 3H), 0.80-0.60 (m, 1H), 0.50-0.30 (m, 2H), 0.20-0.00 (m , 1 HOUR) .
Ethyl ester of (E) -3- acid. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -cyclohexa-2,4-dienyl} -2-methyl-acrylic (Intermediate 87)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethynyl-8,8-dimethyl-4-methoxy-5,6,7,8-tetrahydro-naphthalene (Intermediate 81 0.08 g, 0.28 mmol), ethyl ester of (E) -3- (4-iodo-phenyl) -2-methyl-acrylic acid (Reagent 7, 0.09 g, 0.28 mmol), triethylamine (8 ml), copper (I) iodide (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) ) using 5-10% ethyl acetate in hexane as eluent, the title compound was obtained as a white solid (0.12 g, 90%). NMR H1 (300 MHz, CDC13): d 7.67 (d, 1H, "7 = 1.2Hz), 7.57 (d, 2H," 7 = 8.4Hz), 7.40 (d, 2H, «7 = 8.4Hz), 7.19 (d, 1H,« 7 = 1.5Hz), 6.83 (d, 1H, «7 = 1.5Hz), 4.28 (c, 2H,« 7 = 7.2Hz), 4.04 (m, 1H), 3.84 (s, 3H), 2.32 (s, 3H), 2.15 (d, 3H, "7 = 1.2Hz), 2 , 03-1.83 (m, 3H), 1.68-1.50 (m, 1H), 1.45-1.20 (m, 1H), 1.36 (s, 3H), 1.35 (t, 3H, J = 7.2 Hz), 1.20 (s, 3H), 0.32-0.23 (m, 2H), 0.08-0.00 (m, 2H).
Acid (E) -3-. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -cyclohexa-2,4-dienyl} -2-methyl-acrylic (Compound 30)
A solution of methyl ester of (E) -3- acid. { 4- [5- (cyclopropyl-methyl-amino) -4-methoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene-2-ylethynyl] -cyclohexa-2,4-dienyl} -2-methyl-acrylic acid (Intermediate 87, 0.12 g, 0.25 mmol) in methanol (3 ml) and tetrahydrofuran (2 ml) was treated with a 3 M potassium hydroxide (1 ml, 3 mmol) solution and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was recrystallized from hot acetonitrile to give the title product as a white solid ( 0.055 g, 49%). H1 NMR (300 MHz, CDC13): d 7.78 (d, 1H, "7 = 1.2Hz), 7.57 (d, 2H," 7 = 8.1Hz), 7.43 (d, 2H, J = 8.1 Hz), 7.29 (d, 1H, «7 = 1.5Hz), 6.93 (d, 1H,« 7 = 1.5Hz), 4.93 and 4.70 (2m, 1H ), 3.97 (s, 3H), 2.54 (s, 3H), 2.40-1.60 (m, 4H), 2.16 (d, 3H, "7 = 1.2Hz), 1 , 46-1.23 (m, 1H), 1.46 (s, 3H), 1.23 (s, 3H), 0.90-0.20 (m, 4H).
2. HPLC
Intermediate
Intermediate 90 Intermediate 91
Reagent 4 Compound 31 2. NaOHTLiOH / KOH
2. NaOH / LiOH / KOH
Reagent 5 Compound 32 2. NaOH / LiOH / KOH
Reaction Scheme 16 Cyclopropyl- (3-iodo-benzyl) -amine (Intermediate 88)
A solution of 3-iodobenzyl bromide (Aldrich, 3.2 g, 10.77 mmol) in ethanol (20 ml) was treated with cyclopropylamine (7 ml, 101.5 mmol) and the resulting reaction mixture was stirred at 3 days long at room temperature. The volatiles were evaporated in vacuo, the residue was diluted with ethyl acetate and washed with an aqueous solution of saturated sodium bicarbonate, water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo until an oil that was subjected to column chromatography on silica gel (230-400 mesh) using 10-20% ethyl acetate in hexane as eluent afforded the title product (2.4 g, 81%). H1 NMR (300 MHz, CDC13): d 7.67 (s, 1H), 7.58 (d, 1H, "7 = 9.0Hz), 7.27 (d, 1H," 7 = 6.0Hz) , 7.05 (dd, 1H, "7 = 6.0, 9.0Hz), 3.78 (s, 2H), 2.13 (m, 1H), 1.76 (s broad, 1H), 0 50-0.35 (m, 4H).
Cyclopropyl- (3-iodo-benzyl) -methyl-amine (Intermediate 89)
A solution of cyclopropyl- (3-iodo-benzyl) -amine (Intermediate 88, 4.1 g, 15 mmol) in acetone (20 ml) was treated with potassium carbonate (2.07 g, 15 mmol) and sodium iodide. methyl (1.4 ml, 22.5 mmol) and the resulting reaction mixture was stirred at room temperature for Ih. Diethyl ether was added, the solids were removed by filtration and the filtrate was evaporated to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent to provide the title compound (3.3 g, 77%). NMR H1 (300 MHz, CDCl3): d 7.62 (d, 1H, J = 1.5Hz), 7.55 (dd, 1H, "7 = 1.5, 7.8Hz), 7.21 (dd) , 1H, J "= 1.5, 7.8Hz), 7.01 (t, 1H," 7 = 7.8Hz), 3.61 (s, 2H), 2.22 (s, 3H), 1 69 (m, 1H), 0.50-0.38 (m, 4H) Cyclopropylmethyl- (3-trimethylsilanylethynyl-benzyl) -amine (Intermediate 90)
Following General Procedure D and using cyclopropyl- (3-iodo-benzyl) -methyl-amine (Intermediate 89, 0.97 g, 3.4 mmol), triethylamine (10 ml), copper iodide (I) (0.051 g) , 0.27 mmol), trimethylsilylacetylene (2 ml, 14 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.19 g, 0.27 mmol) followed by flash column chromatography on silica gel (230-400 mesh). ) using hexane-5% ethyl acetate in hexane as eluent, the title compound was obtained (0.695 g, 80%). H NMR (300 MHz, CDCl 3): d 7.37-7.31 (m, 2H), 7.25-7.20 (m, 2H), 3.61 (s, 2H), 2.22 (s) , 3H), 1.69 (m, 1H), 0.50-0.32 (m, 4H), 0.25 (s, 9H).
Cyclopropyl- (3-ethynyl-benzyl) -methyl-amine (Intermediate 91)
A solution of cyclopropylmethyl- (3-trimethylsilanylethynyl-benzyl) -amine (Intermediate 90, 0.355 g, 1.38 mmol) in methanol (10 ml) was treated with potassium carbonate (0.13 g, 0.95 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (0.22 g, 86%). NMR H1 (300 MHz, CDC13): d 7.41-7.35 (m, 2H), 7.26-7.23 (m, 2H), 3.63 (s, 2H), 3.05 (s) , 1H), 2.23 (s, 3H), 1.70 (m, 1H), 0.48-0.40 (m, 4H).
Methyl ester of (E) -3- (4-. {3, 3- [(cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -acrylic acid (Intermediate 92)
Following General Procedure B and using cyclopropyl- (3-ethynyl-benzyl) -methyl-amine (Intermediate 91, 0.060 g, 0.32 mmol), methyl 4-iodo-cinnamate (Reagent 4, 0.093 g, 0.32 mmoles), triethylamine (8 ml), copper (I) iodide (0.015 g, 0.08 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.056 g, 0.08 mmol) followed by flash column chromatography on gel of silica (230-400 mesh) using 5-15% ethyl acetate in hexane as eluent, the title compound was obtained (0.11 g, 100%). NMR H1 (300 MHz, CDC13): d 7.66 (d, 2H, < 7 = 16.2Hz), 7.54-7.39 (m, 2H), 7.31-7.25 (m, 2H), 6.43 (d, 2H, "7 = 16.2Hz), 3.80 (s, 3H), 3.65 (s, 2H), 2.25 (s, 3H), 1.72 ( m, 1H), 0.49-0.42 (m, 4H).
Acid (E) -3- (4- { 3- [(cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -acrylic (Compound 31)
A solution of (E) -3- (4- {3 - [(cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -acrylic acid methyl ester (Intermediate 92, 0.11 g , 0.32 mmol) in methanol (3 ml) and tetrahydrofuran (2 ml) was treated with a 3M solution of potassium hydroxide (1 ml, 3 mmol) and the resulting reaction mixture was stirred at room temperature for 2 hours. days. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using 10% methanol. % in ethyl acetate as eluent to give the title product as a yellow solid (0.038 g, 36%). H1 NMR (300 MHz, CD30D): d 7.61-7.38 (m, 9H), 6.53 (d, 1H, J = 15.9Hz), 3.93 (s, 2H), 2.48. (s, 3H), 2.09 (m, 1H), 0.64-0.61 (m, 4H).
Ethyl 3- (4-. {3- [3- (cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -but-2-enoic acid ester (Intermediate 93)
Following General Procedure B and using cyclopropyl- (3-ethynyl-benzyl) -methyl-amine (Intermediate 91, 0.12 g, 0.64 mmol), 3- (4-iodo-phenyl) -butyl ethyl ester -2Z-enoic (Reagent 5, 0.2 g, 0.64 mmol), triethylamine (8 ml), copper (I) iodide (0.012 g, 0.063 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.045 g) , 0.064 mmol) followed by flash column chromatography on silica gel (230-400 mesh), gave the title compound (0.17 g, 70%). NMR H1 (300 MHz, CDC13): d 7.52-7.40 (m, 4H), 7.31-7.18 (m, 4H), 5.91 (s, 1H), 4.01 (c , "7 = 7, 1Hz, 2H), 3.66 (s, 2H), 2.26 (s, 3H), 2.17 (s, 3H), 1.74-1.70 (m, 1H) , 1.10 (t, "7 = 7, 1Hz, 3H), 0.50-0.43 (m, 4H).
3- (4- (3- [(Cyclopropyl-methyl-amino) -methyl] -phenylethynyl] -phenyl) -but-2-enoic acid (Compound 32)
A solution of 3- (4- (3- [(cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -but-2-enoic acid ethyl ester (Intermediate 93, 0.17 g, , 46 mmole) in ethanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 3.4 M solution of potassium hydroxide (1 ml, 3.4 mmole) and the resulting reaction mixture was stirred at room temperature for 36 h The reaction mixture was extracted with diethyl ether, and the aqueous phase was neutralized with 10% aqueous hydrochloric acid and evaporated to a solid The solid was subjected to flash column chromatography using ethyl acetate as eluent to give the title product as a white solid (0.05 g, 32%). H NMR (300 MHz, CDC13): d 7.49-7.43
(m, 4H), 7.32-7.20 (m, 4H), 5.93 (s, 1H), 3.70 (s, 2H), 2.29 (s, 3H), 2.17 ( s, 3H), 1.76-1.73 (m, 1H), 0.50-0.48 (m, 4H).
Ethyl 3- (4-. {3- [3- (cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -2-methyl-acrylic acid ester (Intermediate 94)
Following General Procedure B and using cyclopropyl- (3-ethynyl-benzyl) -methyl-amine (Intermediate 91, 0.1 g, 0.54 mmol), ethyl ester of (E) -3- (4-iodo- phenyl) -2-methyl-acrylic (Reagent 7, 0.17 g, 0.54 mmol), triethylamine (10 ml), copper iodide (I) (0.019 g, 0.1 mmol) and dichlorobis- (triphenylphosphine) palladium (II)
(0.071 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 2-10% ethyl acetate in hexane as eluent, the title compound was obtained (0.15 g). , 75%). H 1 NMR (300 MHz, CDCl 3): d 7.66-7.25 (m, 9H), 4.27 (c, J = 7.3 Hz, 2H), 3.65 (s, 2H), 2.25 (s, 3H), 2.13 (d, "7 = 1.2 Hz, 3H), 1.75-1.65 (m, 1H), 1.35 (t," 7 = 7, 3Hz, 3H ), 0.50-0.40 (m, 4H).
3- (4- { 3- [(Cyclopropyl-methyl-amino) -methyl] -phenylethynyl} -phenyl) -2-methyl-acrylic acid (Compound 33)
A solution of 3- (4-. {3- [3- (cyclopropylmethyl-amino) -methyl] -phenylethynyl] -phenyl) -2-methyl-acrylic acid ethyl ester (Intermediate 94, 0.15 g, 0.4 mmol) in ethanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 3M solution of potassium hydroxide (1 ml, 3 mmol) and the resulting reaction mixture was stirred at room temperature. the room temperature during the night. The reaction mixture was concentrated, neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to a solid. The solid was subjected to flash column chromatography using 5% methanol in ethyl acetate as eluent to give the title product as an amorphous solid (0.115 g, 83%). NMR H1 (300 MHz, CDC13): 7.71-7.25 (, 9H), 3.81 (s, 2H), 2.44 (s, 3H), 2.13 (d, "7 = 1, 2 Hz, 3H), 1.92-1.80 (m, 1H), 0.76-0.66 (m, 2H), 0.58-0.48 (m, 2H).
1. NaH, THF; Mei, ref lux 2. BH3: Me2S, THF.refiujo Intermediate 95
-G I Reagent 3 2. KOH
Reaction Scheme 17
3 - . 3-Bromo-N-cyclopropyl-4-methyl-benzamide (Intermediate 95)
A stirred, cooled solution (ice bath) of 3-bromo-4-methyl-benzoic acid (Aldrich, 5 g, 23.25 mmol) in benzene (50 ml), dichloromethane (10 ml) and N, N-dimethylformamide (0.5 ml) was treated with oxalyl chloride (4 ml, 46.5 mmol). The reaction mixture was allowed to warm to room temperature over 3 h. The volatiles were then removed by vacuum distillation, the residue was diluted with anhydrous dichloromethane (50 ml) under argon, cooled (ice bath) and treated with 4- (dimethylamino) pyridine (5.67 g, 46, 5 mmoles) followed by cyclopropylamine (1.93 ml, 27.9 mmoles). After 3 h, the reaction mixture was diluted with dichloromethane and washed with water. The organic phase was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to give the title product which was used as such for the next step (6.0 g, -100%).
3-Bromo-N-cyclopropyl-4, N-dimethyl-benzamide (Intermediate 96)
A stirred, cooled solution (ice bath) of 3-bromo-N-cyclopropyl-4-methyl-benzamide (Intermediate 95.6 g, 23.25 mmol) in anhydrous tetrahydrofuran (100 mL) in argon was treated with small portions. of sodium hydride (1.6 g, 40 mmol, 60% dispersion in mineral oil). The reaction mixture was allowed to warm to room temperature and after Ih, methyl iodide (3.11 ml, 50 mmol) was added and the reaction mixture was refluxed for 5 h. It was cooled to room temperature, poured into cold water and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a dirty brown solid which was used as such for the next step (6.3 g, - 100%).
(3-Bromo-4-methyl-benzyl) -cyclopropyl-methyl-amine (Intermediate 97)
A solution of 3-bromo-N-cyclopropyl-4, N-dimethyl-benzamide (Intermediate 96, 5.3 g, 19.77 mmol) in anhydrous tetrahydrofuran (50 ml) was treated with borane-methyl sulfide complex ( 10 ml, 100 mmol) and the resulting reaction mixture was refluxed for 2 h. It was cooled to room temperature and carefully treated with a saturated solution of sodium carbonate, saturated until the effervescence ceased, and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent afforded the title product as an oil (3.2 g, 63%). NMR H1 (300 MHz, CDC13): d 7.47 (s, 1H), 7.17 (d, 1H, "7 = 7.8Hz), 7.12 (d, 1H," 7 = 7.8Hz) , 3.63 (s, 2H), 2.40 (s, 3H), 2.27 (s, 3H), 1.73 (m, 1H), 0.92-0.43 (m, 4H).
Cyclopropyl-methyl- (4-methyl-3-trimethylsilanylethynyl-benzyl) -amine (Intermediate 98)
Following General Procedure D and using cyclopropyl- (3-bromo-4-methyl-benzyl) -methyl-amine (Intermediate 97, 2.24 g, 8.81 mmol), triethylamine (10 ml), tetrahydrofuran (5 ml) , copper iodide (I) (0.4 g, 2.1 mmol), trimethylsilylacetylene (5 ml, 35.4 mmol) and dichlorobis (triphenylphosphine) palladium (II) (1.45 g, 2.06 mmol) followed of flash column chromatography on silica gel (230-400 mesh) using 6-10% ethyl acetate in hexane as eluent, the title compound was obtained (2.25 g, 94%). NMR H1 (300 MHz, CDC13): d 7.08 (s, 1H), 6.84 (2s, 2H), 3.31 (s, 2H), 2.15 (s, 3H), 1.95 ( s, 3H), 1.41 (m, 1H), 0.25-0.00 (m, 4H), 0.00 (s, 9H).
Cyclopropyl- (3-ethynyl-4-methyl-benzyl) -methyl-amine (Intermediate 99)
A solution of cyclopropylmethyl- (4-methyl-3-trimethylsilanylethynyl-benzyl) -amine (Intermediate 98, 0.95 g, 3.5 mmol) in methanol (10 ml) was treated with potassium carbonate (2)., 3 g, 16.6 mmol) and the resulting reaction mixture was stirred at room temperature for lh. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (0.67 g, 96%). H NMR (300 MHz, CDC13): d 7.12 (s, 1H), 6.87 (2s, 2H), 3.33 (S, 2H), 2.98 (s, 1H), 2.16 ( s, 3H), 1.96 (s, 3H), 1.42 (m, 1H), 0.24-0.00 (m, 4H).
Ethyl ester of (E) -3- (4- {5- [(Cyclopropyl-methyl-amino) -methyl] -2-methyl-phenylethynyl} -phenyl) -acrylic acid (Intermediate 100)
Following General Procedure B and using cyclopropyl- (3-ethynyl-4-methyl-benzyl) -methyl-amine (Intermediate 99, 0.095 g, 0.48 mmol), ethyl 4-iodo-cinnamate (Reagent 3, 0.144 g , 0.47 mmol), triethylamine (13 ml), copper iodide (I) (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.071 g, 0.1 mmol) followed by Flash column on silica gel (230-400 mesh) using 5-20% ethyl acetate in hexane as eluent gave the title compound (0.14 g, 82%). H1 NMR (300 MHz, CDCl3): d 7.66 (d, 1H, "7 = 15.9Hz), 7.53 (cAb, 4H," 7 = 6.3Hz), 7.41 (s, 1H) , 7.15 (2s, 2H), 6.44 (d, 1H, "7 = 15.9Hz), 4.26 (c, 2H," 7 = 7.2Hz), 3.62 (s, 2H) 2.48 (s, 3H), 2.24 (s, 3H), 1.68 (m, 1H), 1.33 (t, 3H, "7 = 7.2Hz), 0.49-0, 41 (m, 4H).
Acid (E) -3- (4- { 5- [(Cyclopropyl-methyl-amino) -methyl] -2-methyl-phenylethynyl} -phenyl) -acrylic (Compound 34)
A solution of (E) -3- (4-. {5- [(cyclopropyl-methyl-amino) -methyl] -2-methyl-phenylethynyl} -phenyl) -acrylic acid ethyl ester (Intermediate 100, 0.14 g, 0.37 mmol) in ethanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 3M solution of potassium hydroxide (1 ml, 3 mmol) and the resulting reaction mixture was stirred to the Room temperature during the night. The reaction mixture was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using methanol at room temperature. % in ethyl acetate as eluent to give the title product as an amorphous solid (0.071 g, 55%). NMR H1 (300 MHz, CDCl3): d 7.62 (d, 1H, "7 = 15.9Hz), 7.61 (s, 1H), 7.38 (s, 4H), 7.19 (s, 2H), 6.56 (d, 1H, "7 = 15.9Hz), 3.87 (s, 2H), 2.50 (s, 3H), 2.49 (s, 3H), 1.94 ( m, 1H), 0.89-0.83 (m, 2H), 0.60-0.57 (m, 2H).
Intermediate 101 Intermediate 102
K2C03, MeOH
Intermediate 103 2. NaOH Reaction Scheme 18
Trifluoromethanesulfonic acid 3-tert-butyl-phenyl ester (Intermediate 101)
A stirred, cooled solution (ice bath) of 3-t-butylphenol (Aldrich, 2 g, 13.3 mmol) in anhydrous dichloromethane (15 ml) was treated with 2- [N, N'-bis (trifluoromethylsulfonyl) amino] ] -5-chloropyridine (7.8 g, 20 mmol) followed by 4- (dimethylamino) pyridine (3.2 g, 26.6 mmol). The cooling bath was removed and the reaction mixture was stirred at room temperature for 18 h. It was diluted with ethyl acetate, washed with 2 N hydrochloric acid, 2 N sodium hydroxide, and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent afforded the title product as a clear oil (3.06 g, 82%). NMR H1 (300 MHz, CDCl3): d 7.42-7.32 (m, 2H), 7.24 (d, 1H, "7 = 1.8Hz), 7.10-7.06 (m, 1H ), 1.33 (s, 9H).
(3-t-Butyl-phenylethynyl) -trimethyl-silane (Intermediate 102)
Following General Procedure D and using trifluoromethanesulfonic acid 3-tert-butyl-phenyl ester, (Intermediate 101, 2.54 g, 9.0 mmol), triethylamine (2 ml), copper (I) iodide (0.63) g, 3.33 mmol), trimethylsilylacetylene (5 ml, 36 mmol) and dichlorobis (triphenylphosphine) palladium (II) (1.6 g, 2.25 mmol) followed by flash column chromatography on silica gel (230 mesh). 400) using 10% ethyl acetate in hexane as eluent, the title compound was obtained as a brown oil which was used as such for the next step.
1-t-Butyl-3-ethynyl-benzene (Intermediate 103)
A solution of 3-t-butyltrimethylsilanylethynylbenzene
(Intermediate 102, 0.47 g, 2.04 mmol) in methanol (20 ml) was treated with potassium carbonate (2)., 8 g, 20.2 mmol) and the resulting reaction mixture was stirred at room temperature for 3 days. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent afforded the title compound as a light yellow oil (0.125 g, 39%).
NMR H1 (300 MHz, CDCl3): d 7.40 (d, 1H, J = 1.5Hz), 7.39-7.10 (m, 3H), 2.91 (s, 1H), 1.18 (s, 9H).
(E) -3- [4- (3-Butyl-phenylethynyl) -phenyl] -acrylic acid (Compound 35)
A solution of (E) -3- [4- (3-t-butyl-phenylethynyl) -phenyl] -acrylic acid ethyl ester (Intermediate 103, 0.015 g, 0.047 mmol) in ethanol (2 ml) and tetrahydrofuran (2) ml) was treated with a 2M solution of lithium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was neutralized with 10% aqueous hydrochloric acid and evaporated in vacuo to a solid which was washed with water and hexane and dried to give the title product as a white solid (0.012 g, 85%). %). NMR
H1 (300 MHz, CDC13): d 7.78 (d, 1H, J = 16.2Hz), 7.59-7.26
(m, 8H), 6.47 (d, 1H, "7 = 16.2Hz), 1.34 (s, 9H).
2. NaBH4, Intermediate MeOH 104 * "" 4 'Intermediate 106
2. MeOH, K2C03 Intermediate 107 Intermediate 108
Intermediate 110 2. Compound KOH 36
Reaction Scheme 19
4-t-Butyl-2-hydroxy-benzaldehyde (Intermediate 104)
A stirred, cooled solution (ice bath) of 3-t-butylphenol (1.5 g, 10 mmol) in anhydrous dichloromethane was treated with titanium tetrachloride (1.86 ml, 17 mmol) followed by dichloromethyl ether (0, 9 ml, 20 mmol). The reaction was allowed to warm to room temperature over 1 h, quenched carefully with ice and water and extracted with dichloromethane. The organic extract was washed with water and brine, dried over sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to flash column chromatography using 2-2.5% ethyl acetate in hexane as eluent to provide the title compound (1.37 g, 77%). RM1 H1
(300 MHz, CDC13): d 11.02 (s, 1H), 9.81 (s, 1H), 7.45 (d,
J = 8.2Hz, 1H), 7.03 (dd, "7 = 8.2, 1.7Hz, 1H), 6.99 (d," 7 = 1.7 Hz, 1H), 1.31 ( s, 9H).
Trifluoromethanesulfonic acid 5-t-butyl-2-formyl-phenyl ester (Intermediate 105)
A stirred, cooled solution (ice bath) of
4-t-butyl-2-hydroxy-benzaldehyde (Intermediate 104, 0.75 g, 4.21 mmol) in anhydrous dichloromethane (10 ml) was treated with triethylamine (1.76 ml, 12.64 mmol) followed by 2 - [N, N-bis (trifluoromethylsulfonyl) amino] pyridine (1.81 g, 4.62 mmol). The reaction mixture was allowed to warm to room temperature overnight. The volatiles were evaporated and the residue was subjected to flash column chromatography using 2-2. 5% ethyl acetate in hexane as eluent to provide the title compound (0.16 g) and a 1: 1 mixture of product and starting material (0.47 g). The title compound was used as such for the next step.
-t-Butyl-2-hydroxymethyl-phenol (Intermediate 106)
A stirred, cooled solution (ice bath) of a 1: 1 mixture of 5-t-butyl-2-formyl-phenyl ester of trifluoromethanesulfonic acid and 4-t-butyl-2-hydroxy-benzaldehyde (Intermediate 105, 0, 47 g) in methanol (8 ml) was treated with sodium borohydride (0.1 g, 2.64 mmol). After Ih, the reaction mixture was diluted with water and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to flash chromatography on silica gel (230-400 mesh) to provide the title product. (0.3 g). 1 H-NMR (300 MHz, CDC13): d 6.94-6.84 (m, 3H), 4.72 (s, 2H), 1.26 (s, 9H).
-t-Butyl-2-methyl-phenol (Intermediate 107)
A solution of 5-t-butyl-2-hydroxymethyl-phenol
(Intermediate 106, 0.215 g, 1.19 mmol) in ethyl acetate was treated with 5% palladium on carbon (0.04 g) and the resulting reaction mixture was stirred under a hydrogen atmosphere at room temperature for 2 hours. , 5h. The reaction mixture was then filtered on a pad of celite and the filtrate was evaporated in vacuo to give the title compound as a white solid
(0.19 g, 97%). NMR H1 (300 MHz, CDC13): d 7.03 (d, J = 7.9 Hz, 1H), 6.86 (dd, "7 = 7.9, l, 7Hz, 1H), 6.78 (d) , "7 = 1.7 Hz, 1H), 5.20 (s, 1H), 2.20 (s, 3H), 1.25 (s, 9H).
-t-Butyl-2-methyl-phenyl ester of trifluoromethanesulfonic acid (Intermediate 108)
A solution of 5-t-butyl-2-methyl-phenol (Intermediate 107, 0.19 g, 1.15 mmol) and 4- (dimethylamino) pyridine (0.28 g, 2.3 mmol) in anhydrous dichloromethane ( 8 ml) was treated with N-phenyltrifluoromethanesulfonimide (0.54 g, 1.5 mmol), and the resulting reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuo and the residue was subjected to column chromatography on silica gel (230-400 mesh) to give the title compound as a colorless oil (0.28 g, 82%). 1 H-NMR (300 MHz, CDC13): d 7.30-7.20 (m, 3H), 2.33 (s, 3H), 1.30 (s, 9H).
(5-t-Butyl-2-methyl-phenylethynyl) -trimethyl-silane (Intermediate 109)
Following General Procedure D and using 5-t-butyl-2-methyl-phenyl ester of trifluoro-methanesulfonic acid (Intermediate 108, 0.28 g, 0.94 mmol), triethylamine (3 ml), trimethylsilylacetylene (1 ml, 7 mmoles), N, N-dimethylformamide (6 ml) and dichlorobis (triphenylphosphine) palladium (II) (0.053 g, 0.076 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using hexane as eluent, the title compound was obtained (0.16 g, 69%). H1 NMR (300 MHz, CDC13): d 7.44 (d, "7 = 1.7Hz, 1H), 7.22 (dd," 7 = 8.2, 1, 7Hz, 1H), 7.10 ( d, "7 = 8, 2Hz, 1H), 2.39 (s, 3H), 1.28 (s, 9H), 0.26 (s, 9H).
4-t-Butyl-2-ethynyl-l-methyl-benzene (Intermediate 110)
Following the general procedure F and using (5-t-butyl-2-methyl-phenylethynyl) -trimethyl-silane (Intermediate 109, 0.16 g, 0.66 mmol), methanol (5 ml) and potassium carbonate ( , 05 g, 0.36 mmol), the title compound was obtained (0.08 g, 67%). H1 NMR (300 MHz, CDC13): d 7.49 (d, "7 = 1.7Hz, 1H), 7.30 (dd," 7 = 8.2, 1.7Hz, 1H), 7.15 ( d, "7 = 8, 2Hz, 1H), 3.16 (s, 1H), 2.42 (s, 3H), 1.32 (s, 9H).
Ethyl 3- [4- (5-t-butyl-2-methyl-phenylethynyl) -phenyl] -acrylic acid ester (Intermediate 111)
Following General Procedure B and using 4-t-butyl-2-ethynyl-1-methyl-benzene (Intermediate 110, 0.08 g, 0.47 mmol), ethyl 4-iodocinnamate (0.12 g, 0, 4 mmoles), triethylamine (8 ml), copper iodide (I) (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by column chromatography Flash on silica gel (230-400 mesh) using 2-4% ethyl acetate in hexane as eluent, the title compound was obtained (0.09 g, 55%). 1 H-NMR (300 MHz, CDCl 3): d 7.67 (d, "7 = 16, 1Hz, 1H), 7.56-7.48 (m, 5H), 7.28 (dd," 7 = 8, 2, 1.7Hz, 1H), 7.16 (d, «7 = 8, 2Hz, 1H), 6.44 (d,« 7 = 16, 1Hz, 1H), 4.27 (c, «7 = 7, 1Hz, 2H), 2.48 (s, 3H), 1.33 (t, "7 = 7, 1Hz, 3H), 1.32 (s, 9H).
3- [4- (5-t-Butyl-2-methyl-phenylethynyl) -phenyl] -acrylic acid (Compound 36)
A solution of 3- [4- (5-t-butyl-2-methyl-phenylethynyl) -phenyl] -acrylic acid ethyl ester (Intermediate 111, 0.09 g, 0.26 mmol) in ethanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 3M solution of potassium hydroxide (1 ml, 3 mmol) and the resulting reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated in vacuo slightly, the residue was neutralized with dilute hydrochloric acid, and the solid that formed was filtered and washed with water and acetonitrile and dried to provide the title product (0.064 g, 77%) .
H1 NMR (300 MHz, CDC13): d 7.78 (d, "7 = 16, 1Hz, 1H), 7.58-7.53 (m, 5H), 7.29 (dd," 7 = 7, 9, 1.7Hz, 1H), 7.17 (d, «7 = 7, 9Hz, 1H), 6.47 (d,« 7 = 16, 1Hz, 1H), 2.48 (s, 3H), 1.32 (s, 9H).
Intermediate 112 Intermediate 113
,
2. NaOH
Compound 37 Reaction Scheme 20
N- (3-Bromo-phenyl) -N-methyl-formamide (Intermediate 112)
A solution of 3-bromo-N-methylaniline (prepared as described by Lopez et al in Tet. Lett., 1999, 40, 11, pp. 2071-2074 incorporated herein by reference; 7.4 g, 39.5 mmol) in formic acid (20 ml) was refluxed for 3 h. The reaction mixture was then cooled to room temperature, diluted with water and extracted with diethyl ether. The organic phase was washed with an aqueous solution of saturated sodium bicarbonate, water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a dark brown oil. .
(3-Bromo-phenyl) -cyclopropyl-methyl-amine (Intermediate 113)
A stirred, cooled solution (0 ° C) of N- (3-bromophenyl) -N-methyl-formamide (Intermediate 112, 2.6 g, 9.7 mmol) and titanium tetra-iso-propoxide (3.9 ml, 10.67 mmol) in tetrahydrofuran (40 ml) was treated with a 3 M solution of ethylmagesium bromide in ether (8.08 ml, 24.25 mmol) in argon and the resulting reaction mixture was allowed to warm to the ambient temperature gradually and refluxed at 55 ° C overnight. It was then cooled in an ice bath, quenched with a saturated aqueous solution of ammonium chloride, filtered over celite and the aqueous phase extracted with diethyl ether. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide an oil. Flash column chromatography on silica gel
(230-400 mesh) using 1.5% ethyl acetate in hexane as eluent afforded the title compound
(0.321 g, 15%).
Cyclopropyl-methyl- (3-trimethylsilanylethynyl-phenyl) -amine (Intermediate 114)
Following General Procedure D and using (3-bromo-phenyl) -cyclopropyl-methyl-amine (Intermediate 113, 0.056 g, 0.25 mmol), triethylamine (3 ml), copper iodide (I)
(0.025 g, 0.13 mmol), trimethylsilylacetylene (2.5 ml, 17.6 mmol) and dichlorobiphenyl (triphenylphosphine) -palladium (II) (0.065 g, 0.09 mmol) followed by flash column chromatography on silica gel. silica (230-400 mesh) using 1.5% ethyl acetate in hexane as eluent, the title compound was obtained (0.051 g, 84%).
Cyclopropyl- (3-ethynyl-phenyl) -methyl-amine (Intermediate 115)
A solution of cyclopropyl-methyl- (3-trimethylsilanylethynyl-phenyl) -amine (Intermediate 114, 0.05 g, 0.2 mmol) in methanol (5 ml) was treated with potassium carbonate (0.063 g, 0.46 mmol) ) and the resulting reaction mixture was heated at 80 ° C for 3 h. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (0.035 g, 100%).
Ethyl ester of (E) -3- (4- [3- (cyclopropyl-methyl-amino) -phenylethynyl} -acrylic acid (Intermediate 116)
Following General Procedure B and using cyclopropyl- (3-ethynyl-phenyl) -methyl-amine (Intermediate 115, 0.035 g, 0.2 mmol), ethyl 4-iodo-cinnamate (0.082 g, 0.27 mmol), triethylamine (3 ml), copper iodide (I)
(0.025 g, 0.13 mmol) and dichlorobis (triphenylphosphine) -palladium (II) (0.033 g, 0.047 mmol) followed by flash column chromatography on silica gel
(230-400 mesh), and preparative normal phase HPLC using 10% ethyl acetate in hexane as the mobile phase, gave the title compound (0.020 g, 29%).
(E) -3- (4- [3- (Cyclopropyl-methyl-amino) -phenylethynyl] -phenyl} -acrylic acid (Compound 37)
A solution of ethyl ester of (E) -3- acid. { 4- [3- (cyclopropyl-methyl-amino) -phenylethynyl} -acrylic (Intermediate
116, 0.020 g, 0.057 mmol) in ethanol (1 ml) was treated with a 1 M solution of sodium hydroxide (1 ml, 1 mmol) and the resulting reaction mixture was heated at 80 ° C for
minutes. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide a residue which in preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase gave the title product as a solid yellow (0.006 g, 33%).
Intermediate 118
Intermediate 120 Intermediate 122 R2 = H R5 = H R2 = F, R5 = H Reagent 1 R2 = H, R5 = Me
Compound 38 R2 = H R5 = H Compound 39 R¡ = F, R5 = H Compound 40 R2 = H, R5 = Me
Reaction Scheme 21
4-Bromo-2-methyl-benzoic acid isopropyl ester (Intermediate 117)
A solution of 4-bromo-2-methyl-benzoic acid (Aldrich, 5.4 g, 25 mmol) in benzene (75 ml) and isopropanol (75 ml) was treated with concentrated sulfuric acid (1.5 ml) and heated to reflux for 4 days using a Dean water trap. -Stark. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with water and a saturated aqueous sodium bicarbonate solution, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a clear oil which was used as is. for the next stage (6.12 g, 95%).
Isopropyl ester of 4-bromo-2-bromomethyl-benzoic acid (Intermediate 118)
A solution of 4-bromo-2-methyl-benzoic acid isopropyl ester (Intermediate 117, 6.12 g, 23.8 mmol) in carbon tetrachloride (120 ml) was treated with N-bromosuccinimide (4.6 g, 26.18 mmoles) and 2,2'-azobisisobutyronitrile (0.6 g) and the resulting reaction mixture was refluxed overnight. It was cooled to room temperature, the solids were separated by filtration and washed with hexane: diethyl ether 1: 1, and the filtrate and the washings were evaporated in vacuo to provide an oil (5.1 g, 64%) whIt was used as is for the next stage.
Isopropyl ester of 4-bromo-2-cyclopropylaminomethyl-benzoic acid (Intermediate 119)
A stirred, cooled solution (ice bath) of 4-bromo-2-bromomethyl-benzoic acid isopropyl ester (Intermediate 118, 5.1 g, 15.17 mmol) in acetonitrile (25 ml) was treated with cyclopropylamine (2 ml). ml, 28.9 mmol). The reaction mixture was allowed to warm to room temperature. After 2 h, the volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 4-20% ethyl acetate in hexane as eluent afforded the title product (1.33 g, 28%). NMR H1 (300 MHz, CDCl3): d 7.73 (d, 1H, "7 = 8.4Hz), 7.56 (d, 1H," 7 = 2.1Hz), 7.41 (dd, 1H, "7 = 2.1, 8.4Hz), 5.21 (heptet, 1H, J = 6.3Hz), 4.00 (s, 2H), 2.39 (s broad, 1H), 2.06 ( , 1H), 1.35 (d, 6H, "7 = 6.3Hz), 0.42-0.34 (m, 4H).
Isopropyl ester of 4-bromo-2- [(cyclopropyl-methyl-amino) -methyl] -benzoic acid (Intermediate 120)
A solution of 4-bromo-2-cyclopropylaminomethyl-benzoic acid isopropyl ester (Intermediate 119, 1.33 g, 4.26 mmol) in acetone (8 ml) was treated with potassium carbonate (2.36 g, 17%). 05 mmoles) and methyl iodide (0.53 ml, 8.52 mmol) and the resulting reaction mixture was stirred at room temperature for 3 h. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered over a short pad of silica gel (230-400 mesh) and evaporated in vacuo to give the title product (1.23 g, 70%). NMR H1 (300 MHz, CDCl3): d 7.64 (d, 1H, "7 = 2.1Hz), 7.58 (d, 1H, J = 8.4Hz), 7.39 (dd, 1H," 7 = 2.1, 8.4Hz), 5.20 (heptet, 1H, «7 = 6.0Hz), 3.97 (s, 2H), 2.22 (s, 3H), 1.77 (m , 1H), 1.35 (d, 6H, < 7 = 6.0Hz), 0.46-0.38 (m, 4H).
2- [(Cyclopropyl-methyl-amino-methyl) -methyl] -4-trimethylsilanylethynyl-benzoic acid isopropyl ester (Intermediate 121)
Following General Procedure D and using isopropyl ester of 4-bromo-2- [(cyclopropyl-methyl-amino) -methyl] -benzoic acid (Intermediate 120, 1.23 g, 3.68 mmol), triethylamine (10 ml) , tetrahydrofuran (5 ml), copper (I) iodide (0.21 g, 1.1 mmol), trimethylsilylacetylene (2.1 ml, 14.7 mmol) and dorobis (triphenylphosphine) palladium (II) (0.77 g) g, 1.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 7% ethyl acetate in hexane as eluent, the title compound was obtained as an oil (1.2 g, -100%). NMR H1 (300 MHz, CDCl3): d 7.62 (d, 1H, "7 = 8.1Hz), 7.53 (s, 1H), 7.35 (d, 1H," 7 = 8.4Hz) , 5.20 (heptet, 1H, "7 = 6.3Hz), 3.95 (s, 2H), 2.22 (s, 3H), 1.74 (m, 1H), 1.36 (d, 6H, "7 = 6.3Hz), 0.37-0.28 (m, 4H), 0.27 (s, 9H).
2- [(Cyclopropyl-methyl-amino) -methyl] -4-ethynyl-benzoic acid isopropyl ester (Intermediate 122)
A solution of 2- [(cyclopropyl-methyl-amino) -methyl] -4-trimethylsilanylethynyl-benzoic acid isopropyl ester (Intermediate 121, 0.34 g, 1 mmol) in methanol (2 ml) was treated with potassium carbonate (0.207 g, 1.5 mmol) and the resulting reaction mixture was stirred at room temperature for 4 h. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound as an oil.
(0.21 g, 78%). H1 NMR (300 MHz, CDC13): d 7.67 (d, 1H, "7 = 7.8Hz), 7.64 (d,
1H, "7 = 1.8Hz), 7.38 (dd, 1H, J = 1.8, 7.8Hz), 5.21 (heptet, 1H," 7 = 6.0Hz), 3.96 (s) , 2H), 3.16 (s, 1H), 2.22
(s, 3H), 1.74 (m, 1H), 1.36 (d, 6H, «7 = 6.0Hz), 0.44-0.33
(m, 4H).
2- [(Cyclopropyl-methyl-amino) -methyl] -4- (4-methoxycarbonylmethyl-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 123)
Following General Procedure B and using 2- [(cyclopropyl-methyl-amino) -methyl] -4-ethynyl-benzoic acid isopropyl ester (Intermediate 122, 0.09 g, 0.33 mmol), methyl 4-methyl ester -iodophenylacetic acid (0.09 g, 0.33 mmol), triethylamine (2 ml), copper (I) iodide (0.04 g, 0.21 mmol) and dichlorobis (triphenylphosphine) -palladium (II) (0, 1 g, 0.14 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10-15% ethyl acetate in hexane as eluent, the title compound was obtained as an oil. (0.1 g, 72%). H1 NMR (300 MHz, CDCl3): d 7.70 (d, 1H, "7 = 7.8Hz), 7.62 (d, 1H," 7 = 1.8Hz), 7.52 (d, 2H, «7 = 8.1 Hz), 7.43 (dd, 1H,« 7 = 1.8, 7.8Hz), 7.28 (d, 2H, «7 = 8.1Hz), 5.25 (heptet, 1H, "7 = 6.0Hz), 4.00 (s, 2H), 3.71 (s, 3H), 3.65 (s, 2H), 2.26 (s, 3H), 1.78 ( m, 1H), 1.38 (d, 6H, "7 = 6.0Hz), 0.44-0.40 (m, 4H).
4- (4-Carboxymethyl-phenylethynyl) -2- [(cyclopropyl-methyl-amino) -methyl] -benzoic acid isopropyl ester (Compound 38)
A solution of 2- [(cyclopropyl-methyl-amino) -methyl] -4- (4-methoxycarbonylmethyl-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 123, 0.1 g, 0.23 mmol) in a mixture of methanol (2 ml), tetrahydrofuran (2 ml) and water (1 ml) was treated with lithium hydroxide monohydrate (0.042 g, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 2 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The combined organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a solid. Preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase gave the title product as a white solid
(0.068 g, 72%). H1 NMR (300 MHz, CDC13): d 9.05 (s broad,
1H), 7.73 (d, 1H, "7 = 8.4Hz), 7.66 (s, 1H), 7.44-7.37 (m, 3H), 7.23-7.21 (m , 2H), 5.20 (heptet, 1H, «7 = 6.0Hz), 4.21 (s, 2H), 3.52 (s, 2H), 2.36 (s, 3H), 1.94 (m, 1H), 1.36 (d, 6H, "7 = 6.0Hz), 0.55-0.43 (m, 4H).
2- [(Cyclopropyl-methyl-amino) -methyl] -4- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 124)
Following General Procedure B and using 2- [(cyclopropyl-methyl-amino) -methyl] -4-ethynyl-benzoic acid isopropyl ester (Intermediate 122, 0.05 g, 0.18 mmol), 2-methyl-2-methyl ester -fluoro-4-iodophenylacetic acid (0.07 g, 0.24 mmol), triethylamine (2 ml), copper (I) iodide (0.04 g, 0.21 mmol) and dichlorobis (triphenylphosphine) aladin (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 16% ethyl acetate in hexane as eluent, the title compound was obtained as an oil (0.04 g, 50%). NMR H1 (300 MHz, CDC13): d 7.63 (d, 1H, "7 = 7.8Hz), 7.55 (d, 1H," 7 = 1.2Hz), 7.35 (dd, 1H, "7 = 1.2, 7.8Hz), 7.26-7.17 (m, 3H), 5.16 (heptet, 1H," 7 = 6.3Hz), 3.93 (s, 2H), 3.66 (s, 3H), 3.64 (s, 2H), 2.20 (s, 3H), 1.71 (m, 1H), 1.31 (d, 6H, «7 = 6.3Hz ), 0.40-0.33 (m, 4H).
4- (4-Carboxymethyl-3-fluoro-phenylethynyl) -2- [(cyclopropyl-methyl-amino) -methyl] -benzoic acid isopropyl ester (Compound 39)
A solution of 2- [(cyclopropyl-methyl-amino) -methyl] -4- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 124, 0.04 g, 0.09 mmol) in a mixture of methanol (2 ml), tetrahydrofuran (2 ml) and water (1 ml) was treated with lithium hydroxide monohydrate (0 ml)., 042 g, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 2 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The combined organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a solid. Preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase gave the title product as a white solid
(0.026 g, 54%). H1 NMR (300 MHz, CDC13): d 7.71 (d, 1H, "7 = 8.1Hz), 7.64 (s, 1H), 7.41 (d, 1H," 7 = 8.1Hz) , 7.17-7.09 (m, 3H), 5.20 (heptet, 1H, "7 = 6.3Hz), 4.16 (s, 2H), 3.54 (s, 2H), 2, 34 (s, 3H), 1.91 (m, 1H), 1.36 (d, 6H, "7 = 6.3Hz), 0.50-0.41 (m, 4H).
2- [(Cyclopropyl-methyl-amino) -methyl] -4- (4-methoxycarbonylmethyl-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 125)
Following General Procedure B and using 2- [(cyclopropyl-methyl-amino) -methyl] -4-ethynyl-benzoic acid isopropyl ester (Intermediate 122, 0.07 g, 0.26 mmol), 2- (4- methyl iodophenyl) -propionate (Reagent 1, 0.081 g, 0.29 mmol), triethylamine (2 ml), copper (I) iodide (0.03 g, 0.158 mmol) and dichlorobis (triphenylphosphine) palladium (II) ( 0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10-15% ethyl acetate in hexane as eluent, the title compound was obtained in the form of a oil (0.09 g, 81%).
NMR H1 (300 MHz, CDCl3): d 7.57 (d, 1H, "7 = 8.1 Hz), 7.49 (d, 1H," 7 = 1.8Hz), 7.39 (d, 2H, «7 = 8.4Hz), 7.30 (dd, 1H,« 7 = 1.8, 8.1Hz), 7.18 (d, 2H, «7 = 8.4Hz), 5.10 (heptet, 1H, "7 = 6.0Hz), 3.88 (s, 2H), 3.63 (c, 1H," 7 = 7.2Hz), 3.56 (s, 3H), 2.13 (s, 3H), 1.65 (m, 1H), 1.40 (d, 3H, "7 = 7.2Hz), 1.25 (d, 6H," 7 = 6.0Hz), 0.35-0, 27 (m, 4H).
4- [4- (1-carboxy-ethyl) -phenylethynyl] -2- [(cyclopropyl-methyl-amino) -methyl] -benzoic acid isopropyl ester (Compound 40)
A solution of 2- [(cyclopropyl-methyl-amino) -methyl] -4- (4-methoxycarbonylmethyl-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 125, 0.09 g, 0.21 mmol) in a mixture of methanol (2 ml), tetrahydrofuran (2 ml) and water (1 ml) was treated with lithium hydroxide monohydrate (0.042 g, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 4 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The combined organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a white solid foam (0.053 g, 61%). H1 NMR (300 MHz, CDC13): d 7.68 (d, 1H, "7 = 8.1Hz), 7.58 (d, 1H," 7 = 1.8Hz), 7.44-7.25 ( m, 5H), 5.13 (heptet, 1H, "7 = 6.0Hz), 4.18 (s, 2H), 3.79 (m, 1H), 2.32 (s, 3H), 1, 89 (m, 1H), 1.39 (d, 3H, "7 = 6.6Hz), 1.28 (d, 6H," 7 = 6.3Hz), 0.52-0.21 (m, 4H) ).
I
Intermediate 12 í Intermediate 129
Compound 41 Reaction Scheme 22
4,4-Dimethyl-8- (2-propoxy) -6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalene-1-one (Intermediate 126)
A solution of 8-hydroxy-4,4-dimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 66, 0.32 g, 1.12 mmol) in acetone (20 ml) it was treated with potassium carbonate (0.773 g, 5.6 mmol) and 2-iodopropane (2 g, 11.76 mmol) and the resulting reaction mixture was refluxed for 3 days. Cooled to room temperature, the solids were removed by filtration and the filtrate was evaporated in vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 2-6% ethyl acetate. in hexane as eluent to provide the title product as (0.055 g, 15%). NMR H1 (300 MHz, CDCl3): d 7.04 (d, 1H, "7 = 1.2Hz), 6.89 (d, 1H," 7 = 1.2Hz), 4.57 (heptet, 1H, «7 = 6.3Hz), 2.66 (t, 2H,« 7 = 7.2Hz), 1.92 (t, 2H, «7 = 7.2Hz), 1.38 (d, 6H,« 7 = 6.3 Hz), 1.33 (s, 6H), 0.27 (s, 9H).
4, 4-Dimethyl-6-ethynyl-8- (2-propoxy) -3,4-dihydro-2H-naphthalen-1-one (Intermediate 127)
A solution 4,4-dimethyl-8- (2-propoxy) -6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 126, 0.055 g, 0.167 mmol) in methanol (5 ml) was treated with potassium carbonate (0.03 g, 0.22 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solvent was evaporated in vacuo, the residue was diluted with water and extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound
(0.042 g, 98%). H NMR (300 MHz, CDCl3): d 7.08 (d, 1H, "7 = 1.2Hz), 6.93 (d, 1H," 7 = 1.2Hz), 4.56 (heptet, 1H, «7 = 6.0Hz), 3.19 (s, 1H), 2.67 (t, 2H,« 7 = 6.9Hz), 1.93 (t, 2H, «7 = 6.9Hz), 1 , 39 (d, 6H, "7 = 6.0Hz), 1.34 (s, 6H).
Methyl ester of acid. { 4- [8, 8-dimethyl-5-oxo-4- (2-propoxy) -5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate 128) Following General Procedure B and using 4,4-dimethyl-6-ethynyl-8- (2-propoxy) -3,4-dihydro-2H-naphthalene-lime (Intermediate 127, 0.075 g, 0 , 29 mmol), 4-iodophenylacetic acid methyl ester (0.081 g, 0.29 mmol), triethylamine (8 ml), tetrahydrofuran (3 ml), copper (I) iodide (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-15% ethyl acetate in hexane as eluent, obtained the title compound as a yellow oil (0.07 g, 64%). NMR H1 (300 MHz, CDC13): d 7.52 (d, 2H, "7 = 8.4Hz), 7.29 (d, 2H," 7 = 8.4Hz), 7.12 (d, 1H, «7 = 1.5Hz), 6.97 (d, 1H,« 7 = 1.5Hz), 4.60 (heptet, 1H, «7 = 5.8Hz), 3.71 (s, 3H), 3 , 66 (s, 2H), 2.68 (t, 2H, "7 = 6.6Hz), 1.95 (t, 2H," 7 = 6.6Hz), 1.41 (d, 6H, "7 = 5.8Hz), 1.36 (s, 6H).
Methyl ester of acid. { 4- [5- (cyclopropyl-methyl-amino) -4-isopropoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate 129)
A solution of acid methyl ester. { 4- [8, 8-dimethyl-5-oxo-4- (2-propoxy) -5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Intermediate 128, 0.07 g, 0.187 mmol) in dichloromethane (3 ml) and acetonitrile (1.5 ml) was treated with cyclopropylamine (1 ml, 14.45 mmol). After 5 minutes, acetic acid (1 ml) was added followed by sodium cyanoborohydride (0.12 g, 1.91 mmol). The reaction mixture was stirred overnight at room temperature. It was then diluted with water and a saturated aqueous solution of sodium carbonate and extracted with dichloromethane (x2). The combined organic extract was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to an oil. The oil was dissolved in acetone (15 ml) and treated with potassium carbonate (0.2 g, 1.45 mmol) followed by methyl iodide (1 ml, 15.8 mmol) and the resulting reaction mixture was stirred overnight at room temperature. The precipitated solids were separated by filtration, the filtrate was evaporated in vacuo to a residue. Flash column chromatography on silica gel (230-400 mesh) using 2.5-6% ethyl acetate in hexane as eluent afforded the title compound (0.045 g, 53%). H1 NMR (300 MHz, CDC13): d 7.50 (d, 2H, "7 = 8.4Hz), 7.26 (d, 2H," 7 = 8.4Hz), 7.12 (d, 1H, «7 = 1.5Hz), 6.77 (d, 1H,« 7 = 1.5Hz), 4.58 (heptet, 1H, «7 = 6.3Hz), 4.04 (m, 1H), 3 , 70 (s, 3H), 3.64 (s, 2H), 2.32 (s, 3H), 2.10-1.95 (m, 2H), 1.84-1.78 (m, 1H) ), 1.66-1.60 (m, 1H), 1.40-1.26 (m, 1H), 1.39 and 1.35 (2d, 6H, "7 = 6.3Hz), 1, 34 (s, 3H), 1.19 (s, 3H), 0.29-0.22 (m, 2H), 0.083-0.00 (m, 2H).
Acid { 4- [5- (cyclopropyl-methyl-amino) -4-isopropoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acetic (Compound 41)
A solution of (4- [5- (cyclopropyl-methyl-amino) -4-isopropoxy-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl ester} -acetic (Intermediate 129, 0.045 g, 0.098 mmol) in methanol (2 ml) and tetrahydrofuran (2 ml) was treated with 2M lithium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred to the ambient temperature for 2 h The volatile substances were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate.The organic phase was washed with water and brine, and dried over sulfate of anhydrous magnesium, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using 5% methanol in ethyl acetate as eluent to provide the title product in the form of a white solid (0, 027 g, 61%). NMR H1 (300 MHz, CDCl3): d 7.46 (d, 2H, "7 = 8.1Hz), 7.29 (d, 2H," 7 = 8.1Hz), 7.14 (d, 1H, «7 = 1.2Hz), 6.80 (d, 1H,« 7 = 1.2Hz), 4.62 (, heptet, 1H, J = 6.0Hz), 4.31 (, 1H), 3.58 (s, 2H), 2.46 (s, 3H), 2.46-2.39 (m, 1H), 2.14-1.87 (m, 2H), 1.72-1.67 (m , 1H), 1.42-1.23 (m, 1H), 1.40 and 1.34 (2d, 6H, "7 = 6.0Hz), 1.31 (s, 3H), 1.16 ( s, 3H), 0.80-0.70 (m, 1H), 0.53-0.38 (m, 2H), 0.23-0.18 (m, 1H).
Br Nc to EtOH, conc. H2S04) »- 2. K2C03, EtOH Intermediate 130
Intermediate 132 Compound 42 R2 = H R2 = H Compound 43 R2 = F R2 = F 2. LiOH, EtOH, THF, H20
Intermediate 136
Reaction Scheme 23 Ethyl 2- (3-bromo-phenyl) -2-methyl-propionic acid ester (Intermediate 130)
A solution of 2- (3-bromo-phenyl) -2-methyl-propionitrile (prepared as described by Barlaam et al., "7. Med. Chem., 1999, 42, 23, 4890-4908 incorporated herein by reference; 4 g, 6.24 mmol) was dissolved in ethanol (40 ml), treated with concentrated sulfuric acid (1 ml) and the resulting reaction mixture was refluxed for 36 h. The reaction mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate 5% in hexane as eluent to give the title product as an orange oil (0.77 g, 46%). NMR H1 (300 MHz, CDC13): d 7.48 (s, 1H), 7.36 (dd, 1H, J = 2.8, 7.7Hz), 7.26 (dd, 1H,, 7 = 2 , 8, 8.3Hz), 7.20 (dd, 1H, "7 = 7.8, 8.3Hz), 4.12 (c, 2H," 7 = 7.0Hz), 1.55 (s, 6H), 1.18 (t, 3H, "7 = 7.0Hz).
Ethyl ester of 2-methyl-2- (3-trimethylsilanylethynyl-phenyl) -propionic acid (Intermediate 131)
Following General Procedure D and using 2- (3-bromo-phenyl) -2-methyl-propionic acid ethyl ester
(Intermediate 130, 0.77 g, 2.84 mmol), triethylamine (5 ml), copper iodide (I) (0.044 g, 0.23 mmol), trimethylsilylacetylene (2 ml, 14.1 mmol) and dichlorobis ( triphenylphosphine) palladium (II) (0.159 g, 0.23 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using hexane to 5% ethyl acetate in hexane as eluent, the title (0.74 g, 90%) in the form of a orange-colored oil. H1 NMR (300 MHz, CDC13): d 7.45 (s, 1H), 7.33-7.24 (m, 3H), 4.12 (c, 2H, "7 = 7.0Hz), 1, 56 (s, 6H), 1.17 (t, 3H, "7 7.0Hz), 0.25 (s, 9H).
Ethyl 2- (3-ethynyl-phenyl) -2-methyl-propionic acid ester (Intermediate 132)
A solution of 2-methyl-2- (3-trimethylsilanylethynyl-phenyl) -propionic acid ethyl ester (Intermediate 131, 0.74 g, 2.56 mmol) in ethanol (10 ml) was treated with potassium carbonate (0.degree. , 2 g, 1.45 mmol). The resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo and the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate 1-5% in hexane as eluent to provide the title product (0.4 g, 72%). NMR H1 (300 MHz, CDC13): d 7.56 (s, 1H), 7.45-7.33 (m, 3H), 4.18 (c, 2H, "7 = 7.0Hz), 3, 14 (s, 1H), 1.63 (s, 6H), 1.24 (t, 3H, "7 = 7.0Hz).
Ethyl 2- [3- (4-methoxycarbonylmethyl-phenylethynyl) -phenyl] -2-methyl-propionic acid ester (Intermediate 133)
Following General Procedure B and using 2- (3-ethynyl-phenyl) -2-methyl-propionic acid ethyl ester (Intermediate 132, 0.101 g, 0.47 mmol), methyl 4-iodophenylacetic acid ester (0.129 g, 0.47 mmol), triethylamine (8 ml), copper iodide (I) (0.01 g, 0.05 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.035 g, 0.05 mmol) followed by chromatography in an instantaneous column on silica gel (230-400 mesh) using 10-15% ethyl acetate in hexane as eluent, the title compound was obtained as an oil (0.14 g, 82%). NMR R1 (300 MHz, CDCl3): d 7.52-7.25 (m, 8H), 4.13 (c, 2H, "7 = 7.0Hz), 3.70 (s, 3H), 3, 64 (s, 2H), 1.58 (s, 6H), 1.18 (t, 3H, "7 = 7.0Hz).
2- [3- (4-Methoxycarbonylmethyl-phenylethynyl) -phenyl] -2-methyl-propionic acid (Compound 42)
A solution of 2- [3- (4-methoxycarbonylmethyl-phenylethynyl) -phenyl] -2-methyl-propionic acid ethyl ester
(Intermediate 133, 0.12 g, 0.33 mmol) in ethanol (2 ml) and tetrahydrofuran (2 ml) was treated with 2M lithium hydroxide.
(1 mL, 2 mmol) and the resulting reaction mixture was stirred at room temperature for Ih. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as an oil (0.11 g, 95%). NMR H1 (300 MHz, CDCl3): d 7.48 (s, 1H), 7.35-7.04 (m, 7H), 4.11 (c, 2H, "7 = 7.0Hz), 3, 32 (s, 2H), 1.50 (s, 6H), 1.11 (t, 3H, "7 = 7.0Hz).
Ethyl 2- [3- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -phenyl] -2-methyl-propionic acid ester (Intermediate 134) Following General Procedure B and using 2- (3-ethynyl) ethyl ester phenyl) -2-methyl-propionic (Intermediate 132, 0.10 g, 0.46 mmol), 2-fluoro-4-iodophenylacetic acid methyl ester (0.136 g,
0.46 mmoles), triethylamine (8 ml), copper iodide (l) (0.01 g, 0.05 mmol) and dichlorobis (triphenylphosphine) -palladium (II)
(0.035 g, 0.05 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10-15% ethyl acetate in hexane as eluent, the title compound was obtained as an oil (0.15 g, 85%). NMR H1 (300 MHz, CDCl3): d 7.52 (s, 1H) 7.39-7.21 (m, 6H), 4.13 (c, 2H, "7 = 7.0Hz), 3.71 (s, 3H), 3.68 (s, 2H), 1.58 (s, 6H), 1.18 (t, 3H, "7 = 7.0Hz).
2- [3- (3-Fluoro-4-methoxycarbonylmethyl-phenylethynyl) -phenyl] -2-methyl-propionic acid (Compound 43)
A solution of 2- [3- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -phenyl] -2-methyl-propionic acid ethyl ester (Intermediate 134, 0.13 g, 0.34 mmol) in ethanol (2 ml) and tetrahydrofuran (2 ml) was treated with 2 M lithium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at room temperature for 45 minutes. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product (0.125 g, -100%). NMR H1 (300 MHz, CDCl3): d 7.48 (s, 1H) 7.34-7.06 (m, 6H), 4.10 (c, 2H, "7 = 7.0Hz), 3.41 (s, 2H), 1.52 (s, 6H), 1.13
(t, 3H, "7 = 7.0Hz).
Isopropyl ester of 3-bromo-benzoic acid (Intermediate 135)
A solution of 3-bromobenzoic acid (Aldrich, 2.4 g, 11.9 mmol) in isopropanol (20 ml) was treated with 1 ml of concentrated sulfuric acid and the resulting reaction mixture was refluxed overnight. The reaction mixture was then cooled to room temperature and diluted with water and extracted with diethyl ether. The organic phase was dried over anhydrous sodium sulfate, filtered and evaporated to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent to provide the compound of the title in the form of an oil (2.54 g, 88%). H1 NMR (300 MHz, CDC13): d 8.14 (s, 1H), 7.95 (d, "7 = 7, 6Hz, 1H), 7.64 (d," 7 = 7, 6Hz, 1H) , 7.29 (t, "7 = 7, 6Hz, 1H), 5.24 (hept," 7 = 6, 1Hz, 1H), 1.35 (d, "7 = 6, 1Hz, 6H).
3-Trimethylsilanylethynyl-benzoic acid isopropyl ester (Intermediate 136)
Following General Procedure D and using isopropyl ester of 3-bromo-benzoic acid (Intermediate 135, 1.25 g, 5.14 mmole), triethylamine (12 ml), copper iodide (I) (0.078 g, 0.41 mmoles), trimethylsilylacetylene (4 ml, 28.16 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.288 g, 0.41 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using ethyl acetate. 3% ethyl in hexane as eluent, the title compound (1.25 g, 94%) was obtained as an orange oil. H1 NMR (300 MHz, CDC13): d 8.09 (s, 1H), 7.96 (d, "7 = 7, 6Hz, 1H), 7.59 (d," 7 = 7, 6Hz, 1H) , 7.35 (t, «7 = 7, 6Hz, 1H), 5.24 (hept,« 7 = 6, 1Hz, 1H), 1.35 (d, «7 = 6, 1Hz, 6H), 0 , 25 (s, 9H).
Isopropyl ester of 3-ethynyl-benzoic acid (Intermediate 137)
A solution of 3-trimethylsilanylethynyl benzoic acid isopropyl ester (Intermediate 136, 0.6 g, 2.3 mmol) in anhydrous tetrahydrofuran (3 ml) was treated with a 1 M solution of tetra-n-butylammonium fluoride in tetrahydrofuran. (4.6 mL, 4.6 mmoles) and the resulting reaction mixture was stirred in an ice bath for 5 min. Water was added and the reaction mixture was extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to an oil which was purified by flash column chromatography using 5% -30% ethyl acetate in hexane as eluent to provide the title compound as a solid (0.33 g, 76%). H1 NMR (300 MHz, CDC13): d 8.15 (s, 1H), 8.01 (d, "7 = 7, 6Hz, 1H), 7.64 (d," 7 = 7, 6Hz, 1H) , 7.39 (t, «7 = 7, 6Hz, 1H), 5.25 (hept,« 7 = 6, 1Hz, 1H), 3.13 (s, 1H), 1.37 (d, «7 = 6, 1Hz, 6H).
3- (4-Ethoxycarbonylmethyl-3-fluoro-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 138)
Following General Procedure B and using 3-ethynyl-benzoic acid isopropyl ester (Intermediate 137, 0.099 g, 0.53 mmol), 2-fluoro-4-iodophenylacetic acid ethyl ester (0.164 g, 0.53 mmol), triethylamine (3 ml), copper (I) iodide (0.01 g, 0.05 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.035 g, 0.5 mmol) followed by flash column chromatography on silica gel. silica (230-400 mesh) using 7-10% ethyl acetate in hexane as eluent, the title compound was obtained as a light orange oil (0.08 g, 92%). H1 NMR (300 MHz, CDC13): d 8.17 (s, 1H), 8.01 (d, "7 = 7, 6Hz, 1H), 7.67 (d," 7 = 7, 6Hz, 1H) , 7.42 (t, «7 = 7, 6Hz, 1H), 7.29-7.22 (m, 3H), 5.21 (hept,« 7 = 6, 1Hz, 1H), 4.18 ( c, .7 = 7, 1Hz, 2H), 3.68 (s, 2H), 1.38 (d, «7 = 6, 1Hz, 6H), 1.26 (t,« 7 = 7, 1Hz, 3H).
3- (4-Carboxymethyl-3-fluoro-phenylethynyl) -benzoic acid isopropyl ester (Compound 44)
A solution of 3- (4-ethoxycarbonylmethyl-3-fluoro-phenylethynyl) -benzoic acid isopropyl ester (Intermediate 138, 0.1 g, 0.27 mmol) in isopropanol (2 ml) and tetrahydrofuran (2 ml) was treated with a 2 M solution of lithium hydroxide (1 ml, 2 mmol). After 40 min. at room temperature, the reaction mixture was concentrated in vacuo a little, neutralized with 10% hydrochloric acid and the solid formed was filtered, washed with water and dried to give the title compound (0.09 g, 97%). H NMR (300 MHz, CDC13): d 8.18 (s, 1H), 8.02 (d, J = 7.6Hz, H), 7.68 (d, "7 = 7, 6Hz, 1H), 7.44 (t, "7 = 7, 6Hz, 1H), 7.31-, 24 (m, 3H), 5.27 (hept," 7 = 6, 1Hz, 1H), 3.74 (s, 2H), 1.39 (d, "7 = 6, 1Hz, 6H).
TiCl4, C12CH (0CH3), CH2C12 Intermediate 139
Intermediate 140 Intermediate 141
Intermediate 144 Compound 45
Reaction Scheme 24 4-Bromo-2-t-butyl-5-methyl-phenol (Intermediate 139)
A solution of 4-bromo-3-methylphenol (Aldrich, 5.1 g, 27.3 mmol) in anhydrous dichloromethane (50 ml) was treated with 2-methyl-2-propanol (15 ml) and concentrated sulfuric acid (3 ml). ml) and stirred at room temperature for 3 months. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography using 3-5% ethyl acetate in hexane as eluent afforded the title compound as a dark yellow oil (3.42 g, 51%). This was used as is for the next stage. H NMR (300 MHz, CDC13): d 7.40 (s, 1H), 6.56 (s, 1H), 5.23 (s, 1H), 2.30 (s, 3H), 1.41 ( s, 9H).
3 - . 3-Bromo-5- t-butyl-6-hydroxy-2-methyl-benzaldehyde (Intermediate 140)
A stirred, cooled solution (ice bath) of 4-bromo-2-t-butyl-5-methyl-phenol (Intermediate 139, 0.85 g, 3.5 mmol) in anhydrous dichloromethane (7 ml) was treated with titanium tetrachloride (0, 64 ml, 5.8 mmol) followed by D, rj-dichloromethyl ether (0.3 g, 3.5 mmol). The reaction mixture was allowed to warm to room temperature for 4 h. The reaction mixture was diluted with diethyl ether, washed with brine (xl) and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230 mesh) -400) using 1% ethyl acetate in hexane to provide the title compound as a yellow solid (0.58 g, 61%). NMR H1 (300 MHz, CDCl3): d 12.89 (s, 1H), 10.32 (s, 1H), 7.60 (s, 1H), 2.63 (s, 3H), 1.38 ( s, 9H).
3-Bromo-5-t-butyl-6-isopropoxy-2-methyl-benzaldehyde (Intermediate 141)
A stirred, cooled solution (ice bath) of 3-bromo-5-t-butyl-6-hydroxy-2-methyl-benzaldehyde (Intermediate
140, 0.58 g, 2.14 mmol) in anhydrous N, N-dimethylformamide
(10 ml) was treated with sodium hydride (0.34 g of 60% suspension in mineral oil, 8.56 mmol). After 30 minutes, 2-iodopropane (1.3 ml, 12.84 mmol) was added and the reaction mixture was heated at 75 ° C overnight. The reaction mixture was then cooled and poured into ice water and extracted with diethyl ether. The organic extract was then washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to an oil. Flash column chromatography using 2-4% ethyl acetate in hexane as eluent afforded the title product (0.43 g, 64%). NMR H1 (300 MHz, CDC13): d 10.23 (s, 1H), 7.68 (s, 1H), 4.34 (heptet, 1H, "7 = 6.2Hz), 2.57 (s, 3H), 1.40 (s, 9H), 1.28 (d, 6H, "7 = 6.2Hz).
3- t-Butyl-2-isopropoxy-6-methyl-5-trimethylsilanylethynyl-benzaldehyde (Intermediate 142)
Following General Procedure D and using 3-bromo-5-t-butyl-6-isopropoxy-2-methyl-benzaldehyde (Intermediate 141, 0.43 g, 1.37 mmol), triethylamine, copper iodide (I) ( 0.021 g, 0.11 mmol), trimethylsilylacetylene
(1 ml), and dichlorobis- (triphenylphosphine) palladium (II) (0.077 g, 0.11 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 2% ethyl acetate in hexane as eluent, the title compound was obtained (0.45 g, -100%). NMR H1 (300 MHz, CDCl3): d 10.10 (s, 1H), 7.41 (s, 1H), 4.19
(heptet, 1H, "7 = 6.1Hz), 2.44 (s, 3H), 1.21 (s, 9H), 1.09 (d, 6H," 7 = 6.1Hz), 0.08 (s, 9H).
3- Butyl-5-ethynyl-2-isopropoxy-6-methyl-benzaldehyde (Intermediate 143)
A solution of 3-butyl-2-isopropoxy-6-methyl-5-trimethylsilanylethynyl-benzaldehyde (Intermediate 142, 0.45 g, 1.37 mmol) in methanol (5 ml) and tetrahydrofuran was treated with potassium carbonate ( 0.2 g, 1.45 mmol) and the resulting reaction mixture was stirred at room temperature for 3 h. The reaction mixture was evaporated in vacuo and the residue was extracted with diethyl ether and washed with water and brine. The organic phase was dried, filtered and evaporated in vacuo to give the title compound (0.35 g, 90%). H1 NMR (300 MHz, CDC13): d 10.28 (s, 1H), 7.63 (s, 1H), 4.38 (s, 4.38 (s, 1H, "7 = 6.2Hz), 4.38 (s, 1H), 2.63 (s, 3H), 1.39 (s, 9H), 1.29 (d, 6H, J = 6.2Hz).
[4- (5-t-Butyl-3-formyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 144) Following General Procedure B and using 3-t-butyl- 5-ethynyl-2-isopropoxy-6-methyl-benzaldehyde (Intermediate 143, 0.35 g, 1.35 mmol), 4-iodophenylacetic acid methyl ester (0.374 g, 1.35 mmol), triethylamine (8 ml) , copper iodide (I) (0.02 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.072 g, 0.1 mmol) followed by flash column chromatography on silica gel (230 mesh) 400) using 3-5% ethyl acetate in hexane as eluent, the title compound was obtained as a white solid (0.37 g, 75%). H1 NMR (300 MHz, CDC13): d 10.29 (s, 1H), 7.65 (s, 1H), 7.48 (d, 2H, "7 = 8.2Hz), 7.53 (d, 2H, "7 = 8.2Hz), 4.38 (heptet, 1H," 7 = 6.1Hz), 3.68 (s, 3H), 3.62 (s, 2H), 2.68 (s, 3H), 1.41 (s, 9H), 1.27 (d, 6H, "7 = 6.1Hz).
[4- (5- t-Butyl-3-ethynyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 145)
Anhydrous tetrahydrofuran (3 ml) was added to a 2 M solution of trimethylsilyldiazomethane in hexanes (0.37 ml, 0.74 mmol) and the resulting reaction mixture was cooled to -78 ° C. A solution of 1.6 M zi-butyl lithium in hexanes (0.5 ml, 0.8 mmol) was added followed, after 30 minutes, by a solution of methyl ester of acid [4- (5-t- butyl-3-formyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic (Intermediate 144, 0, 2 g, 0.49 mmol) in anhydrous tetrahydrofuran and the resulting reaction mixture was stirred at -78 ° C for Ih and at 0 ° C for 40 minutes. The reaction mixture was then quenched with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether. The organic phase was washed with brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using ethyl acetate at 2, 5-4% in hexane as eluent followed by preparative normal phase HPLC using 5% ethyl acetate in hexane as the mobile phase to give the title product as a colorless oil (0.023 g, 11.6%). H1-NMR (300 MHz, CDC13): d 7.49 (d, 2H, "7 = 8.0Hz), 7.44 (s, 1H), 7.26 (d, 2H," 7 = 8.0Hz) 5,76
(heptet, 1H, "7 = 6.1Hz), 3.70 (s, 3H), 3.64 (s, 2H), 3.58
(s, 1H), 2.58 (s, 3H), 1.39 (s, 9H), 1.31 (d, 6H, "7 =
6.1Hz).
[4- (5- t-Butyl-3-ethynyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] acetic acid (Compound 45)
A solution of [4- (5-butyl-3-ethynyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 145, 0.023 g, 0.057 mmol) in methanol (1, 5 ml) and tetrahydrofuran (1.5 ml) was treated with 1M lithium hydroxide (0.5 ml, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 45 minutes. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product (0.020 g, 91%). H1 NMR (300 MHz, CDC13): d 7.47 (d, 2H, "7 = 8.0Hz), 7.43 (s, 1H), 7.24 (d, 2H," 7 = 8.0Hz) , 5.75 (heptet, 1H, "7 = 6.1Hz), 3.62 (s, 2H), 3.57 (s, 1H), 2.57 (s, 3H), 1.38 (s, 9H), 1.30 (d, 6H, "7 = 6.1Hz).
[4- (5- t-Butyl-4-isopropoxy-2-methyl-3-vinyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 146)
A solution of methylidene triphenylphosphorane [5 ml of a 0.1 M solution, 0.5 mmol, generated from methyltriphenylphosphonium bromide (2.5 g, 7 mmol) and a 1.6 M solution of n-butyl lithium in hexanes (2.9 ml, 4.7 mmol) in 50 ml of tetrahydrofuran] was added to a solution of [4- (5-butyl-3-formyl-4-isopropoxy-2-methyl-phenylethynyl) methyl ester) phenyl] -acetic (Intermediate 144, 0.052 g, 0.13 mmol) in tetrahydrofuran (1 ml). After lh the reaction mixture was quenched with water and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a clear oil which after flash column chromatography on silica gel (230-400 mesh) using ethyl acetate 5% hexane as eluent afforded the title compound (0.02 g, 39%). NMR H1 (300 MHz, CDCl3): d 7.48 (d, 2H, "7 = 7.9Hz), 7.39 (s, 1H), 7.25 (d, 2H," 7 = 7.9Hz) ", 6.73 (dd, 1H," 7 = 11.4, 17.9Hz), 5.49 (dd, 1H, "7 = 2.0, 11.4Hz), 5.37 (dd, 1H, "7 = 2.1, 17.9Hz), 4.93 (heptet, 1H," 7 = 6.4Hz), 3.70 (s, 3H), 3.63 (s, 2H), 2.44 ( s, 3H), 1.40 (s, 9H), 1.17 (d, 6H, "7 = 6.4Hz).
[4- (5- t-Butyl-4-isopropoxy-2-methyl-3-vinyl-phenylethynyl) -phenyl] -acetic acid (Compound 46) A solution of [4- (5-t-butyl) methyl ester -4-isopropoxy-2-methyl-3-vinyl-phenylethynyl) -phenyl] -acetic acid (Intermediate 146, 0.02 g, 0.049 mmol) in methanol (1.5 ml) and tetrahydrofuran (1.5 ml) was treated with 1M lithium hydroxide (0.5 ml, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 45 minutes. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the product of the txtule (0.020 g, -100%). H1 NMR (300 MHz, CDC13): d 7.48 (d, 2H, "7 = 8.2Hz), 7.39 (s, 1H), 7.24 (d, 2H, 7 = 8.2Hz), 6.72 (dd, 1H, "7 = 11.4, 17.9Hz), 5.49 (dd, 1H," 7 = 2.0, 11.4Hz), 5.37 (dd, 1H, "7 = 2.1, 17.9Hz), 4.92 (heptet, 1H, «7 = 6.2Hz), 3.64 (s, 2H), 2.43 (s, 3H), 1.40 (s, 9H), 1.17 (d, 6H, "7 = 6.2Hz).
Intermediate 142 Intermediate 147
Intermediate 148
Reaction Scheme [4 - (5-t-Butyl-3-hydroxymethyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 147)
A stirred, cooled solution (ice bath) of [4- (5-t-butyl-3-formyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 142, 0.172 g , 0.42 mmol) in methanol (4 ml) was treated with sodium borohydride (0.degree., 02 g, 0.51 mmol) and the mixture. The resulting reaction mixture was stirred for 1.5 h. The reaction mixture was quenched with water and extracted with diethyl ether. The organic phase was washed with water (xl) and brine (xl), dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh). ) using 15-20% ethyl acetate in hexane as eluent to give the title product as a white solid (0.15 g, 88%). NMR H1 (300 MHz, CDC13): d 7.48 (d, 2H, «7 = 8.5Hz), 7.47 (s, 1H), 7.25 (d, 2H, <7 = 8.5 Hz), 4.74 (s broad, 2H), 4.74-4.60 (m, 1H), 3.69 (s, 3H), 3.63 (s, 2H), 2 , 60 (s, 3H), 1.40 (s, 9H), 1.27 (d, 6H, "7 = 6.2Hz).
[4- (3-Bromomethyl-5-t-butyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 148)
A stirred, cooled solution (ice bath) of [4- (5-butyl-3-hydroxymethyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 147, 0.15 g, 0.37 mmol) and triphenylphosphine (0.125 g, 0.48 mmol) in anhydrous dichloromethane (5 ml) was treated with N-bromosuccinimide (0.085 g, 0.48 mmol) in argon and the resulting reaction mixture was left tempering at room temperature and stirred overnight. The reaction mixture was quenched with a dilute aqueous solution of sodium bicarbonate and extracted with diethyl ether. The organic phase was washed with water (xl) and brine (xl), dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which in flash column chromatography silica gel (230-400 mesh) using 4-5% ethyl acetate in hexane as eluent afforded the title compound (0.12 g, 69%) as a colorless oil. This was used as is for the next stage.
Methyl ester of acid. { 4- [5- t -butyl-4-isopropoxy-2-methyl-3- (3-trimethylsilanyl-prop-2-ynyl) -phenylethynyl] -phenyl} -acetic (Intermediate 149)
A solution of [4- (3-bromomethyl-5-t-butyl-4-isopropoxy-2-methyl-phenylethynyl) -phenyl] -acetic acid methyl ester (Intermediate 148, 0.12 g, 0.25 mmol) in triethylamine (1 ml) and N, N-dimethylformamide (4 ml) was purged with argon and treated with trimethylsilylacetylene (0.5 ml, 3.5 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.025 g, 0.036 g). mmoles). The resulting reaction mixture was heated at 85 ° C overnight at the end of which it was cooled to room temperature and subjected to column chromatography on silica gel (230-400 mesh) using 4% ethyl acetate in hexane as eluent followed by preparative normal phase HPLC using 3% ethyl acetate in hexane as mobile phase to provide the title compound as an oil (0.038 g, 31%). H1-NMR (300 MHz, CDC13): d 7.50 (d, 2H, "7 = 7.9Hz), 7.48 (s, 1H), 7.26 (d, 2H," 7 = 7.9Hz) , 4.89 (heptet, 1H, «7 = 6.5Hz), 3.70 (s, 3H), 3.64 (s, 2H), 3.50 (s, 2H), 2.57 (s, 3H), 1.40 (s, 9H), 1.27 (d, 6H, "7 = 6.5Hz), 0.12 (s, 9H).
[4- (5- t-Butyl-4-isopropoxy-2-methyl-3-prop-2-ynyl-phenylethynyl) -phenyl] -acetic acid (Compound 47)
A solution of acid methyl ester. { 4- [5- t -butyl-4-isopropoxy-2-methyl-3- (3-trimethylsilanyl-prop-2-ynyl) -phenylethynyl] -phenyl} -acetic (Intermediate 149, 0.038 g, 0.078 mmol) in methanol (1.5 ml) and tetrahydrofuran (1.5 ml) was treated with 2M lithium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at room temperature for 1.5 h. The volatiles were evaporated in vacuo to a residue which was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product (0.032 g, 98%). H1 NMR (300 MHz, CDC13): d 7.50 (d, 2H, "7 = 8.1Hz), 7.43 (s, 1H), 7.27 (d, 2H," 7 = 8.1Hz) , 4.82 (heptet, 1H, «7 = 6.4Hz), 3.67 (s, 2H), 3.48 (d, 2H, J = 2.5Hz), 2.58 (s, 3H), 1.39 (s, 9H), 1.28 (d, 6H, "7 = 6.4Hz).
Intermediate 150 Intermediate 152 intermediate 154
Intermediate 155 Intermediate 156
Compound 49 R5 = H, R5l = Me Compound 50 R5 = R '= Me
Intermediate 164 THF, H20 Compound 48 Reaction Scheme 26 Ethyl 4- (2-bromo-4-methoxy-phenyl) -4-oxo-butyric acid (Intermediate 150)
A stirred, cooled (-30 ° C) solution of 3-bromoanisole (18.7 g, 100 mmol) and ethylsuccinyl chloride (21 mL, 150 mmol) in anhydrous dichloromethane (200 mL) was treated with aluminum chloride (26 mL). , 6 g, 200 mmol) and the reaction mixture was allowed to warm to room temperature and stirred overnight. The reaction mixture was poured into water and extracted with dichloromethane (x2). The combined organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a brown oil. A solid separated upon standing. The liquid supernatant was decanted and the solid was washed with dichloromethane: hexane 1: 3 and dried to give the ethyl ester isomer of 4- (4-bromo-2-methoxy-phenyl) -4-oxo-butyric acid. The combined mother liquors and washings were evaporated to a brown oil which was subjected to column chromatography on silica gel (230-400 mesh) using 15% ethyl acetate in hexane as eluent to provide the ethyl ester isomer of 4- (4-bromo-2-methoxy-phenyl) -4-oxo-butyric acid (overall 12 g, 38%), and the title compound (11.4 g, 36%) and a 1: 1 mixture of both (2 g, 6.3%). H NMR (300 MHz, CDCl3): d 7.59 (d, 1H, "7 = 8.8Hz), 7.14 (d, 1H," 7 = 2.6Hz), 6.87 (dd, 1H, «7 = 2.6, 8.8Hz), 4.14 (c, 2H,« 7 = 7.0Hz), 3.83 (s, 3H), 3.23 (t, 2H, «7 = 6, 4Hz), 2.74 (t, 2H, "7 = 6.4Hz), 1.25 (t, 3H," 7 = 7.0Hz).
Ethyl 4- (2-bromo-4-methoxy-phenyl) -butyric acid ester (Intermediate 151)
A solution of 4- (2-bromo-4-methoxy-phenyl) -4-oxo-butyric acid ethyl ester (Intermediate 150, 6.45 g, 20.5 mmol) in trifluoroacetic acid (32 ml, 409 mmol) treated with triethylsilane (14.4 ml, 90 mmol) and the resulting reaction mixture was heated at 55 ° C for 3 h. The reaction mixture was then cooled to room temperature, neutralized with solid sodium bicarbonate, diluted with water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (5.4 g, 88%) as a colorless oil. NMR H1 (300 MHz, CDCl3): d 7.11 (d, 1H, "7 = 8.2Hz), 7.08 (d, 1H," 7 = 2.6Hz), 6.79 (dd, 1H, «7 = 2.6, 8.2Hz), 4.13 (c, 2H,« 7 = 7.3Hz), 3.76 (s, 3H), 2.71 (t, 2H, 7 = 7.6Hz ), 2.34 (t, 2H, "7 = 7.6Hz), 1.92 (quint, 2H," 7 = 7.6Hz), 1.26 (t, 3H, J = 7.3Hz).
- (2-Bromo-4-methoxy-phenyl) -2-methyl-pentan-2-ol (Intermediate 152)
A stirred, cooled solution (-10 ° C) of 4- (2-bromo-4-methoxy-phenyl) -butyric acid ethyl ester (Intermediate 151, 5.4 g, 18 mmol) in anhydrous tetrahydrofuran (100 ml) it was treated with a 3 M solution of methylmagnesium bromide (16 ml, 48 mmol) and the resulting reaction mixture was allowed to warm to room temperature over 3 h. It was quenched with an aqueous solution of saturated ammonium chloride, diluted with water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a viscous oil (5.16 g, -100%).
H1NMR (300 MHz, CDCl3): d 7.11 (d, 1H, "7 = 8.5Hz), 7.08 (d,
1H, "7 = 2.6Hz), 6.78 (dd, 1H," 7 = 2.6, 8.5Hz), 3.77 (s,
3H), 2.67 (t, 2H, J = 7.3 Hz), 1.69-1.43 (m, 4H), 1.21 (s, 6H).
-Bromo-2-methoxy-l, 1-dimethyl-l, 2,3, 4-tetrahydro-naphthalene (Intermediate 153)
- (2-Chromium-4-methoxy-phenyl) -2-methyl-pentan-2-ol (Intermediate 152, 5.16 g, 17.9 mmol) was treated with 85% sulfuric acid (50 mL) at room temperature. After 30 minutes, the reaction mixture was diluted with cold water and extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product (4.63 g, 96%) a light yellow oil form. H1 NMR (300 MHz, CDC13): d6, 96 (d, 1H, "7 = 2.6Hz), 6.86 (d, 1H," 7 = 2.6Hz), 3.76 (s, 3H) , 2.68 (t, 2H, "7 = 6.7Hz), 1.83-1.75 (m, 2H), 1.62-1.58 (m, 2H), 1.26 (s, 6H) ).
8-Bromo-6-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-l-one (Intermediate 154)
A solution of 5-bromo-2-methoxy-1,1-dimethyl-1,2,3,4-tetrahydro-naphthalene (Intermediate 153, 4.6 g, 17.1 mmol) in glacial acetic acid (20 ml) it was cooled to 0 ° C and treated with a solution of chromium trioxide (5.5 g, 55 mmol) in acetic acid and water (20 ml each). The reaction mixture was then allowed to cool to room temperature and stirred for 24 h. It was diluted with water and extracted with diethyl ether (x2). The combined organic phase was washed with water (x3), saturated aqueous sodium bicarbonate (xl) and brine
(xl), dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title compound (3.9 g, 81%) as a yellow oil. H1 NMR (300 MHz, CDCl3): d 7.09 (d, 1H, "7 = 2.6Hz), 6.87 (d, 1H," 7 = 2.6Hz), 3.85 (s, 3H) , 2.71 (t, 2H, "7 = 7.0Hz), 1.96 (t, 2H," 7 = 7.0Hz), 1.35 (s, 6H).
6-Methoxy-4,4-dimethyl-8-vinyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 155)
A solution of 8-bromo-6-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 154, 2.83 g, 10 mmol) and tributyl (vinyl) tin (3 ml, 10 mmol) in anhydrous N, N-dimethylformamide (30 ml) was purged with argon and treated with tetrakis (triphenylphosphine) palladium (0) (0.3 g, 0.26 mmol). The resulting reaction mixture was heated at 91 ° C for two days at the end of which it was cooled to room temperature, diluted with water and extracted with diethyl ether (x2). The combined organic phase was washed with water (xl), and brine (xl), dried over anhydrous magnesium sulfate, filtered and evaporated to a pale yellow oil. Flash chromatography using 15% ethyl acetate in hexane as eluent afforded the title product (1.7 g, 73%) a light yellow oil form. NMR H1 (300 MHz, CDCl3): d 7.50 (dd, 1H, "7 = 10.8, 17.3Hz), 6.85 (s, 2H), 5.50 (dd, 1H," 7 = 1.4, 17.3 Hz), 5.28 (dd, 1H, "7 = 1.4, 10.8Hz), 3.88 (s, 3H), 2.68 (t, 2H," 7 = 6). , 7Hz), 1.95 (t, 2H, "7 = 6.7Hz), 1.35 (s, 6H).
8-Cyclopropyl-6-methoxy-4, 4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 156)
A stirred, cooled solution (-40 ° C) of 6-methoxy-4,4-dimethyl-8-vinyl-3,4-dihydro-2H-naphthalene-1-one
(Intermediate 155, 51.7 g, 7.4 mmol) in diethyl ether (10 ml) was treated with a solution of diazomethane in ether (40 mmol in 50 ml of ether) followed by palladium (II) acetate.
(0.08 g) and the resulting reaction mixture was warmed to -25 ° C when effervescence was observed. The reaction mixture was then filtered through a plug of silica and the filtrate was evaporated to give a dark brown residue which was subjected to column chromatography on silica gel (23-400 mesh) using ethyl acetate 20% in hexane as eluent to provide the title product as a light yellow solid (1.5 g, 83%). H NMR (300 MHz, CDCl3): d 6.71 (d, 1H, "7 = 2.6Hz), 6.44 (d, 1H," 7 = 2.6Hz), 3.82 (s, 3H) 2.98 (m, 1H), 2.69 (t, 2H, J = 6.7Hz), 1.94 (t, 2H, "7 = 6.7Hz), 1.34 (s, 6H), 1.02-0.88 (m, 2H), 0.65-0.59 (m, 2H).
8-Cyclopropyl-6-hydroxy-3,4-dihydro-2H-naphthalen-1-one (Intermediate 157)
A solution of 8-cyclopropyl-6-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 156, 1.5 g, 6.14 mmol) and sodium cyanide (2 g, 40.8 mmol) in anhydrous dimethylsulfoxide (25 ml) was heated at 230 ° C overnight in argon. The reaction mixture was then cooled to room temperature, poured into ice and acidified (Caution! Hydrogen cyanide evolution!) With dilute hydrochloric acid and extracted with ethyl acetate (x2). The combined organic extract was washed with brine (xl), dried over anhydrous sodium sulfate, filtered and evaporated to give a dark brown oil. Flash column chromatography on silica gel (230-400 mesh) using 2-5% ethyl acetate in hexane as eluent afforded the title compound as a solid (1.1 g, 78%). NMR H1 (300 MHz, CD3COCD3): d 8.14 (s, 1H), 6.75 (d, 1H, "7 = 2.4Hz), 6.40 (d, 1H," 7 = 2.4Hz) , 3.02 (m, 1H), 2.62 (t, 2H, "7 = 6.8Hz), 1.94 (t, 2H," 7 = 6.8Hz), 1.33 (s, 6H) , 0.93-0.89 (m, 2H), 0.59-0.55 (m, 2H).
Ester 4-cyclopropyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalene-2-trifluoro-methanesulfonic acid (Intermediate 158)
A solution of 8-cyclopropyl-6-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 157, 1.1 g, 4.78 mmol) and 4-dimethylaminopyridine (1 , 22 g, 10 mmol) in anhydrous dichloromethane (20 ml) was treated 2- [N, N-bis (trifluoromethylsulfonyl) amino] -5-chloro-pyridine (2.07 g, 5.26 mmol) in argon to the room temperature. After 3.5 h, the reaction mixture was subjected to flash chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent to give the title compound as a solid
(1.76 g, 100%). H1 NMR (300 MHz, CDC13): d 7.10 (d, 1H, "7 = 2.3Hz), 6.78 (d, 1H," 7 = 2.3Hz), 2.90 (m, 1H) , 2.78 (t, 2H, "7 = 7.0Hz), 2.01 (t, 2H," 7 = 7.0Hz), 1.38 (s, 6H), 1.10-1.04 ( m, 2H), 0.67-0.62 (m, 2H).
8-Cyclopropyl-4,4-dimethyl-6- (trimethylsilanyl) ethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 159)
Following General Procedure B and using 4-cyclopropyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalene-2-trifluoro-methanesulfonic acid ester
(Intermediate 158, 1.09 g, 3 mmol), triethylamine (5 ml), tetrahydrofuran (5 ml), copper (I) iodide (0.12 g, 0.6 mmol), dichlorobis (triphenylphosphine) palladium (II ) (0.42 g, 0.6 mmol) and (trimethylsilyl) acetylene (2.2 ml, 15 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 7% ethyl acetate in hexane as eluent, the title compound was obtained as an orange oil (1.05 g, quantitative). H1 NMR (300 MHz, CDC13): d 7.29 (d, 1H, "7 = 1.2Hz), 6.98 (d, 1H," 7 = 1.2Hz), 2.81 (m, 1H) , 2.72 (t, 2H, «7 = 6.7Hz), 1.95 (t, 2H, J = 6.7Hz), 1.34 (s, 6H), 1.01-0.95 (m , 2H), 0.66-0.61 (m, 2H), 0.26 (s, 9H).
8-Cyclopropyl-4,4-dimethyl-6-ethynyl-tetralone (Intermediate 160)
Following General Procedure F and using 8-cyclopropyl-4,4-dimethyl-6- (trimethylsilanyl) ethynyl-1-tetralone (Intermediate 159, 1.05 g, 3.38 mmol), methanol (20 ml) and carbonate potassium (1 g, 14.5 mmol) followed by flash column chromatography using 7% ethyl acetate in hexane as eluent, the title compound (0.57 g, 80%) was obtained as a solid of color light yellow.
NMR H1 (300 MHz, CDCl3): d 7.34 (d, 1H, "7 = 2.5Hz), 7.02 (d, 1H," 7 = 2.5Hz), 3.19 (s, 1H) , 2.83 (m, 1H), 2.74 (t, 2H, "7 = 6.7Hz), 1.97 (t, 2H," 7 = 6.7Hz), 1.35 (s, 6H) , 1.03-0.86 (m, 2H), 0.66-0.61 (m, 2H).
Ethyl 3- [4- (4-cyclopropyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -acrylic acid ester (Intermediate 161)
Following General Procedure B and using 8-cyclopropyl-4,4-dimethyl-6-ethynyl-1-tetralone (Intermediate 160, 0.1 g, 0.42 mmol), ethyl ester of (E) -3- ( 4-iodo-phenyl) -acrylic (0.13 g, 0.42 mmol), triethylamine (1 ml), copper (I) iodide (0.02 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium ( II) (0.070 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh), the title compound was obtained (0.12 g, 69%). NMR H1 (300 MHz, CDCl3): d 7.65 (d, 1H, J = 15.8Hz), 7.52 (cAB, 4H, "7 = 8.1Hz), 7.37 (d, 2H," 7 = 1.5Hz), 7.05 (d, 1H, J = 1.5Hz), 6.45 (d, 1H, «7 = 15.8Hz), 4.26 (c, 2H,« 7 = 7 , 2Hz), 2.88-2.79 (m, 1H), 2.77-2.71 (m, 2H), 2.00-1.92 (m, 2H), 1.36-1.21 (m, 9H), 1.04-0.97 (m, 2H), 0.69-0.59 (m, 2H).
Ethyl ester of 3- acid. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -acrylic (Intermediate 162)
Following General Procedure C and using ethyl ester of 3- [4- (4-cyclopropyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -acrylic (Intermediate 161, 0.12 g, 0.29 mmol) in dichloromethane (4 ml) and acetonitrile (2 ml), cyclopropylamine (1 ml, 14.5 mmol), acetic acid (1 ml) and sodium cyanoborohydride. (0.16 g, 2.4 mmol) followed by work-up gave an intermediate in the form of an oil, which was used as such for the next step. The residue (crude 0.18 g) was dissolved in acetone (6 ml) and treated with potassium carbonate (0.28 g, 2 mmol) and methyl iodide (1 ml, 16 mmol). The resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated to an oil. Flash column chromatography on silica gel (230-400 mesh) followed by preparative normal phase HPLC using 5% ethyl acetate in hexane as mobile phase gave the title compound (0.08 g) as an oil Of course, it was used as is for the next stage.
3- (4- [4-Cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -acrylic acid (Compound 48)
A solution of ethyl ester of 3- acid. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} Acrylic (Intermediate 164, 0.08 g, 0.17 mmol) in methanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 2 M solution of sodium hydroxide (2 ml, 4 mmol) and the mixture of The resulting reaction was refluxed overnight. The reaction mixture was cooled to room temperature, the volatiles were evaporated in vacuo, the residue was diluted with a saturated aqueous solution of ammonium chloride, and extracted with ethyl acetate (x2). The combined organic extract was dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a solid. Preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase gave the title product as a solid (0.04 g, 50%). H1 NMR (300 MHz, CDC13): d 7.76 (d, 1H, "7 = 15.8Hz), 7.54 (cAb, 4H," 7 = 8.8Hz), 7.38 (d, 1H, J = 1.5Hz), 6.96 (d, 1H, «7 = 1.5Hz), 6.47 (d, 1H,« 7 = 15.8Hz), 4.31 (t, 1H, «7 = 4.7 Hz) 2.27 (s, 3H), 2.40-1.43 (m, 6H), 1.38 (s, 3H), 1.23 (s, 3H), 0.98-0, 78 (m, 4H), 0.39-0.13 (m, 4H).
8-Cyclopropyl-5- (cyclopropyl-methyl-amino) -4,4-dimethyl- (2-trimethylsilanyl) ethynyl-1,2,3,4-tetrahydronaphthalene (Intermediate 163)
Following General Procedure C and using 8-cyclopropyl-4,4-dimethyl-6- (trimethylsilanyl) ethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 159, 0.77 g, 2.5 mmol ) in dichloromethane (6 ml) and acetonitrile (3 ml), cyclopropylamine (3 ml, 45 mmol), acetic acid (1 ml) and sodium cyanoborohydride (0.63 g, 9.5 mmol) followed by working-up was given a intermediate in the form of an oil, which was used as is for the next stage. The residue (2.5 mmol) was dissolved in acetone (20 ml) and treated with potassium carbonate (1.03 g, 7.5 mmol) and methyl iodide (1.55 ml, 25 mmol). The resulting reaction mixture was stirred at room temperature for 2 days. The solids were separated by filtration, the filtrate and the washings were evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 2-4% ethyl acetate in hexane as the mobile phase afforded the title compound (0.58 g, 75%). 1 H-NMR (300 MHz, CDCl 3): d 7.31 (d, "7 = 1, 6Hz, 1H), 6.89 (d," 7 = 1, 6Hz, 1H), 4.27 (broad s, 1H ), 2.40-2.30 (m, 1H), 2.30-2.20 (m, 1H), 2.24 (s, 3H), 2.10-2.00 (m, 1H), 2.00-1.80 (m, 2H), 1.60-1.50 (m, 1H), 1.35 (s, 3H), 1.20 (s, 3H), 0.90-0, 75 (m, 4H), 0.40-0.25 (m, 3H), 0.26 (s, 9H), 0.20-0.10 (m, 1H).
8-Cyclopropyl-5- (cyclopropyl-methyl-amino) -2-ethynyl-4,4-dimethyl-1,2,3,4-tetrahydronaphthalene (Intermediate 164)
A solution of 8-cyclopropyl-5- (cyclopropyl-methyl-amino) -4,4-dimethyl- (2-trimethylsilanyl) ethynyl-1, 2,3,4-tetrahydronaphthalene (Intermediate 163, 0.3 g, 0, 82 mmol) in methanol (10 ml) was treated with potassium carbonate (0.2 g, 1.44 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The solids were separated by filtration, the residue was diluted with water and extracted with diethyl ether. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated to give the title compound (0.22 g, 92%). 1 H-NMR (300 MHz, CDCl 3): d 7.44 (d, "7 = 1, 6Hz, 1H), 7.01 (d," 7 = 1, 6Hz, 1H), 4.38 (broad s, 1H) ), 3.11 (s, 1H), 2.48-2.38 (m, 1H), 2.38-2.28 (m, 1H), 2.34 (s, 3H), 2.18- 2.08 (m, 1H), 2.05-1.85 (m, 2H), 1.70-1.60 (m, 1H), 1.44 (s, 3H), 1.30 (s, 3H), 1.00- 0.85 (m, 4H), 0.50-0.35 (m, 3H), 0.30-0.18 (m, 1H).
Methyl ester of acid 2-. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -propionic (Intermediate 165)
Following General Procedure B and using 8-cyclopropyl-5- (cyclopropyl-methyl-amino) -2-ethynyl-4,4-dimethyl-1,2,3,4-tetrahydronaphthalene (Intermediate 164, 0.11 g, 0 , 37 mmole), methyl 2- (4-iodophenyl) propionate
(Reagent 1, 0.108 g, 0.37 mmol), triethylamine (10 ml), copper (I) iodide (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0 , 1 mmol) followed by work-up and flash column chromatography on silica gel (230-400 mesh) using l -4% ethyl acetate in hexane as eluent, the title compound was obtained as an amorphous solid pale yellow color (0.148 g, 87%). H1 NMR (300 MHz, CDC13): d 7.51 (d, "7 = 8, 5Hz, 2H), 7.39 (d," 7 = 1, 6Hz, 1H), 7.29 (d, "7 = 8, 5Hz, 2H), 6.97 (d, «7 = 1, 6Hz, 1H), 4.32 (broad s, 1H), 3.75 (c, .7 = 7, 0Hz, 1H), 3.70 (s, 3H), 2.40-2.30 (m, 1H), 2.30-2.20 (m, 1H), 2.28 (s, 3H), 2.18-2, 08 (m, 1H), 2.02-1.82 (m, 2H), 1.62-1.52 (m, 1H), 1.52 (d, "7 = 7, 0Hz, 3H), 1 , 39 (s, 3H), 1.25 (s, 3H), 0.98-0.80 (m, 4H), 0.45-0.25 (m, 3H), 0.20-0.15 (m, 1H).
2- (4- [4-Cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -propionic acid (Compound 49)
A solution of 2- methyl acid ester. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5, 6,7, 8 -tetrahydro-naphthalen-2-ethynyl] -phenyl} -propionic (Intermediate 165, 0.075 g, 0.16 mmol) in methanol (2 ml) and tetrahydrofuran (2 ml) was treated with 2 M lithium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture was stirred at the room temperature for 5h. The reaction mixture was neutralized with ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to give the title product as a yellow solid (0.07 g, 96%). H1 NMR (300 MHz, CDC13): d 7.50 (d, «7 = 8, 5Hz, 2H), 7.39 (d,« 7 = 1, 6Hz, 1H), 7.31 (d, «7 = 8, 5Hz, 2H), 6.97 (d, «7 = 1, 6Hz, 1H), 4.34 (broad s, 1H), 3.74 (c,« 7 = 7, 0Hz, 1H), 2.40-2.30 (m, 1H), 2.30-2.20 (m, 1H), 2.29 (s, 3H), 2.18-2.08 (m, 1H), 2, 02-1.82 (m, 2H), 1.62-1.52 (m, 1H), 1.52 (d, "7 = 7, 0Hz, 3H), 1.39 (s, 3H), 1 , 24 (s, 3H), 0.98-0.80 (m, 4H), 0.40-0.30 (m, 3H), 0.20-0.15 (m, 1H).
Methyl ester of acid 2-. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl ester} -2-methyl-propionic (Intermediate 166)
Following General Procedure B and using 8-cyclopropyl-5- (cyclopropyl-methyl-amino) -2-ethynyl-4,4-dimethyl-1, 2,3,4-tetrahydronaphthalene (Intermediate 164, 0.11 g, 0 , 37 mmol), methyl 2- (4-iodophenyl) -2-methyl-propionate (Reagent 2, 0.118 g, 0.39 mmol), triethylamine (10 mL), copper (I) iodide (0.019 g, , 1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by work-up and flash column chromatography on silica gel (230-400 mesh) using ethyl acetate at 1% - 4% in hexane as eluent, the title compound was obtained as a pale yellow amorphous solid (0.125 g, 70%). H1 NMR (300 MHz, CDCl3): d 7.51 (d, "7 = 8, 5Hz, 2H), 7.39 (d," 7 = 1, 6Hz, 1H), 7.33 (d, "7 = 8, 5Hz, 2H), 6.97 (d, «7 = 1, 6Hz, 1H), 4.32 (broad s, 1H), 3.68 (s, 3H), 2.40-2.30 (m, 1H), 2.30-2.20 (m, 1H), 2.28 (s, 3H), 2.15-2.05 (m, 1H), 2.00-1.80 (m , 2H), 1.61 (s, 6H), 1.62-1.52 (m, 1H), 1.39 (s, 3H), 1.25 (s, 3H), 0.95-0, 80 (m, 4H), 0.45-0.30 (m, 3H), 0.20-0.10 (m, 1H).
Acid 2-. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -2-methyl-propionic (Compound 50)
A solution of 2- methyl acid ester. { 4- [4-cyclopropyl-5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -2-methyl-propionic (Intermediate 166, 0.125 g, 0.266 mmol) in methanol (2.5 ml) and tetrahydrofuran (2.5 ml) was treated with 3M potassium hydroxide (1 ml, 3 mmol) and the mixture of The resulting reaction was stirred at room temperature overnight. The reaction mixture was neutralized with ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to give the title product as a pale yellow amorphous solid (0.12 g, 98% ). H1 NMR (300 MHz, CDCl3): d 7.51 (d, "7 = 8.5Hz, 2H), 7.40-7.38 (m, 3H), 6.97 (d," 7 = 1, 6Hz, 1H), 4.33 (s broad, 1H), 2.40-2.30 (, 1H), 2.30-2.20 (m, 1H), 2.28 (s, 3H), 2 , 10-2.00 (m, 1H), 2.00-1.80 (m, 2H), 1.62 (s, 6H), 1.60-1.50 (m, 1H), 1.39 (s, 3H), 1.24 (s, 3H), 0.95-0.80 (m, 4H), 0.45-0.30 (m, 3H), 0.20-0.10 (m , 1 HOUR) .
United States Patent No.
Reaction Scheme 27
Cyclopropyl- (4, -dimethyl-6-trimethylsilanylethynyl-1, 2,3,4-tetrahydro-naphthalen-1-yl) -methyl-amine (Intermediate 167)
Following General Procedure C and using 4,4-dimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (described in U.S. Patent No. 6,252,090, 1.23 g, 4.6 mmol) in dichloromethane (7 ml) and acetonitrile (3 ml), cyclopropylamine (2.5 ml, 36 mmol), acetic acid (2.5 ml) and sodium cyanoborohydride (0.58 g, 8.6 mmoles) followed by work-up and flash column chromatography on silica gel (230-400 mesh) using 8% ethyl acetate in hexane as eluent an intermediate was provided as a golden yellow solid (1.07 g, 76%). The intermediate (0.67 g, 2.62 mmol) was dissolved in acetone (10 ml) and treated with potassium carbonate (2.2 g, 16 mmol) and methyl iodide (0.75 ml, 12 mmol). . The resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated to an oil which was used as such for the next step.
Cyclopropyl- (6-ethynyl-4, 4-dimethyl-l, 2,3,4-tetrahydro-naphthalen-1-yl) -methyl-amine (Intermediate 168)
A solution of cyclopropyl- (4,4-dimethyl-6-trimethylsilanylethynyl-1, 2,3,4-tetrahydro-naphthalen-1-yl) -methyl-amine (Intermediate 167, 0.67 g, 2.62 mmol) in methanol (10 ml) was treated with potassium carbonate (1 g, 7.23 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo and the residue was diluted with water and extracted with diethyl ether. The organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a light yellow oil (0.5 g, 75%). NMR H1 (300 MHz, CDC13): d 7.47 (d, 1H, "7 = 8.2Hz), 7.41 (d, 1H," 7 = 1.4Hz), 6.79 (dd, 1H, «7 = 8.2, 1.4Hz), 3.92 (t, 1H,« 7 = 8.2Hz), 3.01 (s, 3H), 2.11 (s, 3H), 2.15- 2.07 (m, 1H), 1.95-1.57 (m, 4H), 1.29 (s, 3H), 1.24 (s, 3H), 0.53-0.37 (m, 4H).
Methyl ester of acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -propionic (Intermediate 169)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethenyl-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene (Intermediate 168, 0.116 g, 0.46 mmol) Methyl 2- (4-iodophenyl) propionate (Reagent 1, 0.17 g, 0.59 mmol), triethylamine (0.75 ml), copper iodide (I) (0.07 g, 0.37 mmol) ) and tetrakis (triphenylphosphine) palladium (0) (0.022 g, 0.019 mmol) followed by flash column chromatography on silica gel (230-400 mesh) and preparative normal phase HPLC using 5% ethyl acetate in hexane as eluent , the title compound was obtained (0.08 g, 42%). NMR H1 (300 MHz, CDC13): d 7.51-7.43 (, 3H), 7.29-7.22 (m, 4H), 3.94 (t, 1H, "7 = 7.9Hz) , 3.76-3.62 (m, 1H), 3.65 (s, 3H), 2.12 (s, 3H), 2.15-2.08 (m, 1H), 2.00-1 , 54 (2m, 4H), 1.52-1.46 (2d, 3H, "7 = 7.4Hz), 1.31 (s, 3H), 1.27 (s, 3H), 0.53- 0.38 (m, 4H).
Acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} -propionic (Compound 51)
A solution of 2- methyl acid ester. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -propionic (Intermediate 169, 0.022 g, 0.05 mmol) in methanol (2 ml) and tetrahydrofuran (2 ml) was treated with a 2 M solution of sodium hydroxide (1 ml, 2 mmol) and the resulting reaction mixture it was stirred at room temperature overnight. The reaction mixture was neutralized with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to preparative reverse phase HPLC using 10% water in acetonitrile as mobile phase to provide the product of the title (0.008 g, 40%).
H NMR (300 MHz, CDCl 3): d 7.50-7.44 (m, 3H), 7.31-7.27 (m, 3H), 7.20 (dd, 1H, "7 = 8.2. , 1.5Hz), 4.00 (t, 1H, J = 8.2Hz), 3.74 (c, 1H, «7 = 7.1Hz), 1H), 2.15 (s, 3H), 2 , 15-2.10 (m, 1H), 1.98-1.81 (m, 2H), 1.80-1.63 (m, 2H), 1.51 (d, 3H, "7 = 7"). , 1Hz), 1.31 (s, 3H), 1.27 (s, 3H), 0.52-0.49 (m, 4H).
Methyl ester of 2- (4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -2- methyl-propionic (Intermediate 170)
Following General Procedure B and using 5- (cyclopropyl-methyl-amino) -2-ethyl-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalene (Intermediate 168, 0.16 g, 0.63 mmoles), methyl 2- (4-iodophenyl) -2-methyl-propionate
(Reagent 2, 0.18 g, 0.58 mmol), triethylamine (3 ml), copper iodide (I) (0.048 g, 0.25 mmol) and tetrakis (triphenylphosphine) palladium (0) (0.032 g, 0.027) mmoles) followed by flash column chromatography on silica gel (230-400 mesh) and preparative normal phase HPLC using 6% ethyl acetate in hexane as mobile phase, the title compound was obtained (0.14 g, 56 %). NMR H1 (300 MHz, CDCl3): d 7.54-7.47 (m, 4H), 7.34-7.26 (m, 3H), 3.97 (t, 1H, "7 = 7.9Hz ), 3.68 (s, 3H), 2.16 (s, 3H), 2.16-2.00 (m, 1H), 2.00-1.61 (2m, 4H), 1.61 (s). s, 6H), 1.35 (s, 3H), 1.30 (s, 3H), 0.56-0.44 (m, 4H).
Acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -2-methyl-propionic (Compound 52)
A solution of 2- methyl acid ester. { 4- [5- (cyclopropyl-methyl-amino) -8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl ester} -2-methyl-propionic (Intermediate 170, 0.08 g, 0.19 mmol) in methanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 2 M solution of sodium hydroxide (2 ml, 4 mmol) and the resulting reaction mixture was refluxed overnight. The volatiles were evaporated in vacuo and the residue was diluted with a saturated aqueous solution of ammonium chloride and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title product.
(0.07 g, -100%). H1 NMR (300 MHz, CDC13): d 9.47 (broad s, 1H), 7.53-7.49 (m, 4H), 7.39 (d, 2H, "7 = 8.5Hz), 7 , 26 (dd, 1H, "7 = 7.9,
1.5Hz), 3.97 (t, 1H, «7 = 7.9Hz), 2.16 (s, 3H), 2.16-2.00 (m, 1H), 2.00-1.61 (2m, 4H), 1.61 (s, 6H), 1.35 (s, 3H), 1.30 (s, 3H), 0.56-0.44 (m, 4H).
PhNTf2, DMAP, CH2C12
Application from the United States Published Intermediate No. 171 2003/0166932.
Intermed o 172 3. Mei, K2C03, acetone intermediate 173
Reaction Scheme 28
6-Hydroxy-4, 4, 7-trimethyl-3, 4-dihydro-2H-naphthalen-l-one (Intermediate 171)
A solution of 6-methoxy-4,4,7-trimethyl-3,4-dihydro-2H-naphthalen-1-one (described in US 2003/0166932, published September 4, 2003, incorporated herein by reference; , 5 g, 25.6 mmol) and sodium cyanide (6.25 g, 127 mmol) in anhydrous dimethyl sulfoxide (100 ml) was heated at 230 ° C for 48 h in argon. The reaction mixture was then cooled to room temperature, poured into ice and acidified (Caution! Evolution of hydrogen cyanide!) With dilute hydrochloric acid and extracted with ethyl acetate (x2). The combined organic extract was washed with brine (xl), dried over anhydrous sodium sulfate, filtered and evaporated to give the title compound, which was used as is for the next step (5.2 g, -100% ). NMR H1 (300 MHz, CDC13): d 7.86 (s, 1H), 6.87 (s, 1H), 2.70 (t, 2H, "7 = 7.0Hz), 2.24 (s, 3H), 1.97 (t, 2H, "7 = 7.0Hz), 1.32 (s, 6H).
Ester 3.8, 8-trimethyl-5-oxo-5, 6,7, 8-tetrahydro-naphthalene-2-yl of trifluoromethanesulfonic acid (Intermediate 172)
A solution of 6-hydroxy-4,4,7-trimethyl-3,4-dihydro-2H-naphthalen-l-one (Intermediate 171, 5.2 g, 25.6 mmol) and 4-dimethylaminopyridine (6.1 g, 50 mmol) in anhydrous dichloromethane (50 ml) was treated with N-phenyltrifluoromethanesulfonimide (9.54 g, 26.7 mmol) in argon and stirred at room temperature for lh. The reaction mixture was subjected to flash chromatography on silica gel (230-400 mesh) using 6-7% ethyl acetate in hexane as eluent to give the title compound (6.4 g, 75%). NMR H1 (300 MHz, CDC13): d 7.96 (s, 1H), 7.28 (s, 1H), 2.74 (t, 2H, "7 = 7.0Hz), 2.37 (s, 3H), 2.04 (t, 2H, "7 = 7.0Hz), 1.39 (s, 6H).
4,4, 7-Trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 173)
Following General Procedure D and using ester 3,8,8-trimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalene-2-yl of trifluoromethanesulfonic acid (Intermediate 172,
.04 g, 15 mmol), triethylamine (20 ml), copper iodide (I) (0.6 g, 3 mmol), trimethylsilylacetylene (5.3 ml, 37.5 mmol) and dichlorobis (triphenylphosphine) -palladium (II) (2.2 g, 3 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 6% ethyl acetate in hexane as eluent, the title compound was obtained (4 g, 93%) in the form of a light yellow solid. NMR H1 (300 MHz, CDC13): d 7.54 (s, 1H), 7.19 (s, 1H), 2.42 (t, 2H, "7 = 7.0Hz), 2.14 (s, 3H), 1.70 (t, 2H, "7 = 7.0Hz), 1.08 (s, 6H), 0.00 (s, 9H).
Cyclopropyl- (6-ethynyl-4, 4, 7-trimethyl-1,2,4,4-tetrahydro-naphthalen-1-yl) -methyl-amine (Intermediate 174)
Following General Procedure C and using 4,4, 7-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 173, 4 g, 14 mmol) in dichloromethane (30 ml) and acetonitrile (10 ml), cyclopropylamine (3.11 ml, 45 mmol), acetic acid (3.2 ml) and sodium cyanoborohydride (2 g, 30 mmol) followed by work-up and flash column chromatography on silica gel (230 mesh) -400) using 10% ethyl acetate in hexane as eluent an intermediate was provided in the form of a light yellow solid, which was used as such for the next step (4.1 g, 90%). The intermediate (4.1 g, 13 mmol) was dissolved in acetone (40 ml) and treated with potassium carbonate (10 g, 72 mmol) and methyl iodide (2.5 ml, 40 mmol). The resulting reaction mixture was stirred at room temperature overnight. The volatile substances were evaporated in vacuumThe residue was dissolved in methanol (100 ml) and treated with potassium carbonate (10 g, 72 mmol) and the resulting reaction mixture was stirred at room temperature for 1.5 h. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether (x2). The combined organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated to an oil which was filtered over a short plug of silica gel (230-400 mesh) to give the title compound (3.2 g, 92%). %) in the form of a clear oil. H NMR (300 MHz, CDCl3): d 7.42 (s, 1H), 7.38 (s, 1H), 3.49 (t, 1H, "7 = 7.0Hz), 3.23 (s, 1H), 2.40 (s, 3H), 2.15 (s, 3H), 2.15-2.10 (m, 1H), 1.97-1.62 (2m, 4H), 1.30 (s, 3H), 1.26 (s, 3H), 0.56-0.28 (m, 4H).
2- (4- [5- (Cyclopropyl-methyl-amino) -3,8,8-trimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester. 2-methyl-propionic (Intermediate 175)
Following General Procedure B and using cyclopropyl- (6-ethynyl-4,4,7-trimethyl-1, 2,3,4-tetrahydro-naphthalen-1-yl) -methyl-amine (Intermediate 174, 0.1 g 0.29 mmole), methyl 2- (4-iodophenyl) -2-methylpropionate (Reagent 2, 0.09 g, 0.29 mmol), triethylamine (8 ml), copper (I) iodide ( 0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 1-2% acetate of ethyl in hexane as eluent, the title compound was obtained (0.035 g, 26%). H1 NMR (300 MHz, CDC13): d 7.49 (d, J = 8.5 Hz, 2 H), 7.41 (s, 1 H), 7.38 (d, J = 8.5 Hz, 2 H), 7 , 32 (s, 1H), 3.92 (m, 1H), 3.67 (s, 3H), 2.43 (s, 3H), 2.18-2.10 (m, 1H), 2, 14 (s, 3H), 1.98-1.85 (m, 2H), 1.80-1.64 (m, 2H), 1.60 (s, 6H), 1.31 (s, 3H) , 1.26 (s, 3H), 0.58-0.42 (m, 4H).
Acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -3,8,8-trimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} -2-methyl-propionic (Compound 53)
A solution of 2- methyl acid ester. { 4- [5- (cyclopropyl-methyl-amino) -3,8,8-trimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl ester} -2-methyl-propionic (Intermediate 175, 0.035 g, 0.08 mmol) in methanol (2 ml) and tetrahydrofuran (2 ml) was treated with a 2 M solution of sodium hydroxide (2 ml, 4 mmol) and the The resulting reaction mixture was refluxed for 2 days. The volatiles were evaporated in vacuo and the residue was neutralized with 5% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was purified by preparative reverse phase HPLC using 10% water in acetonitrile as mobile phase to provide the product of the title (0.022 g, 64%). NMR H1 (300 MHz, CDCl3): d 7.48 (d, J = 8.5 Hz, 2H), 7.41 (s, 1H), 7.37-7.34 (m, 3H), 3.95 (m, 1H), 2.40 (s, 3H), 2.18-2.10 (m, 1H), 2.14 (s, 3H), 1.98-1.85 (m, 2H), 1.80-1.64 (m, 2H), 1.57 (s, 6H), 1.29 (s, 3H), 1.25 (s, 3H), 0.56-0.42 (m, 4H).
Inte
2. [> -NH2, NaCNBH3, CH2C12, CH3CN, CH3COOH 3. Mei, K2C03, CH3COCH3 4. KOH
Compound 54 Reaction Scheme 29
8-Ethyl -4,4-dimethyl-6-methoxy-3,4-dihydro-2H-naphthalen-1-one (Intermediate 176)
A solution of 8-vinyl-6-methoxy-4,4-dimethyl-3,4-dihydro-2H-naphthaleri-1-one (Intermediate 155, 1.12 g, 4.86 mmol) in ethyl acetate ml) was treated with 10% palladium on carbon (100mg) and the resulting reaction mixture was stirred under a hydrogen atmosphere overnight. The reaction mixture was filtered on a pad of celite and the filtrate was evaporated to give the title product (1.1 g, 98%). NMR H1 (300 MHz, CDCl3): d 6.77 (d, 1H, "7 = 2.6Hz), 6.54 (d, 1H, < 7 = 2.6Hz), 3.87 (s, 3H) ), 3.05 (c, 2H, "7 = 7.3Hz), 2.67 (t, 2H," 7 = 6.7Hz), 1.95 (t, 2H, "7 = 6.7Hz), 1.36 (s, 6H), 1.23 (t, 3H, "7 = 7.3Hz).
8-Ethyl-6-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 177)
A solution of 8-ethyl-4,4-dimethyl-6-methoxy-3,4-dihydro-2H-naphthalen-1-one (Intermediate 176, 1.1 g, 4.73 mmol) and sodium cyanide (1 , 6 g, 33 mmol) in anhydrous dimethyl sulfoxide (20 ml) was heated at 210 ° C overnight in argon. The reaction mixture was then cooled to room temperature, poured into ice and acidified
(Caution! Evolution of hydrogen cyanide!) Using 10% hydrochloric acid and extracted with ethyl acetate. The combined organic extract was washed with brine (xl), dried over anhydrous sodium sulfate, filtered and evaporated to give a dark orange solid. Flash column chromatography on silica gel (230-400 mesh) using 10-20% ethyl acetate in hexane as eluent afforded the title compound as a yellow solid (0.82 g, 82%) . H1 NMR (300 MHz, CD3C0CD3): d 8.99 (s, 1H), 6.81 (d, 1H, "7 = 2.6Hz), 6.64 (d, 1H," 7 = 2.6Hz) , 2.99 (c, 2H, J = 7.3Hz), 2.60 (t, 2H, J = 6.7Hz), 1.93 (t, 2H, «7 = 6.7Hz), 1.34 (s, 6H), 1.17 (t, 3H, "7 = 7.3Hz).
4-Ethyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalene-2-trifluoro-methanesulfonic acid ester
(Intermediate 178)
A solution of 8-ethyl-6-hydroxy-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 177, 0.27 g, 1.24 mmol) and 4-dimethylaminopyridine (0.242) g, 1.98 mmol) in anhydrous dichloromethane (10 ml) was treated with 2- [N, N-bis (trifluoromethylsulfonyl) amino] -5-chloro-pyridine (0.58 g, 1.48 mmol) in argon a the room temperature for 5h. The reaction mixture was subjected to flash chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent to give the title compound (0.43 g, 98%). NMR H1 (300 MHz, CDCl3): d 7.15 (d, 1H, "7 = 2.6Hz), 7.04 (d, 1H," 7 = 2.6Hz), 3.05 (c, 2H, «7 = 7.3Hz), 2.74 (t, 2H,« 7 = 6.7Hz), 2.00 (t, 2H, «7 = 6.7Hz), 1.38 (s, 6H), 1 , 24 (t, 3H, «7 = 7.3Hz).
8-Ethyl-4,4-dimethyl-6- (trimethylsilanyl) ethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 179) Following General Procedure D and using ester 4-ethyl-8, 8- dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalene-2-trifluoro-methanesulfonic acid (Intermediate 178, 0.9 g, 2.57 mmol), triethylamine (6 ml), N, N-dimethylformamide anhydrous (5 ml), dichlorobis (triphenylphosphine) palladium (II) (0.144 g, 0.2 mmol) and (trimethylsilyl) acetylene (2 ml, 13.64 mmol), the reaction was carried out overnight in a tube sealed at 90 ° C. Work-up followed by flash column chromatography on silica gel (230-400 mesh) using 2-3% ethyl acetate in hexane as eluent afforded the title compound as an orange oil (0.82 g). , quantitative). H1 NMR (300 MHz, CDC13): d 7.34 (d, 1H, "7 = 1.5Hz), 7.21 (d, 1H," 7 = 1.5Hz), 2.97 (c, 2H, «7 = 7.6Hz), 2.69 (t, 2H,« 7 = 6.7Hz), 1.95 (t, 2H, .7 = 6.7Hz), 1.35 (s, 6H), 1 , 20 (t, 3H, "7 = 7.6Hz), 0.27 (s, 9H).
8-Ethyl-6-ethynyl-4,4-dimethyl-3,4-dihydro-2H-naphthalene-1-one
(Intermediate 180)
Following General Procedure F and using 8-ethyl-4,4-dimethyl-6- (trimethylsilanyl) ethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 179, 0.66 g, 2.2 mmol ), methanol (10 ml) and potassium carbonate (0.4 g, 2.9 mmol) gave the title compound as an orange oil (0.59 g, 100%). H1 NMR (300 MHz, CDC13): d 7.51 (d, 1H, "7 = 1.5Hz), 7.37 (d, 1H," 7 = 1.5Hz), 3.32 (s, 1H) , 3.10 (c, 2H, "7 = 7.3Hz), 2.84 (t, 2H," 7 = 6.7Hz), 2.08 (t, 2H, "7 = 6.7Hz), 1 , 48 (s, 6H), 1.33 (t, 3H, "7 = 7.3Hz).
Methyl ester of 2- [4- (4-ethyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] -2-methyl-propionic acid ester ( Intermediate 181)
Following General Procedure B and using 8-ethyl-6-ethynyl-4,4-dimethyl-3,4-dihydro-2H-naphthalen-1-one
(Intermediate 180, 0.09 g, 0.39 mmol), methyl 2- (4-iodo phenyl) -2-methyl-propionate (Reagent 2, 0.152 g, 0.5 mmol), triethylamine (8 ml), copper iodide (I) (0.024 g,
0.12 mmol) and dichlorobis (triphenylphosphine) palladium (II)
(0.087 g, 0.12 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 2-10% ethyl acetate in hexane as eluent, the title compound was obtained as an oil (0.095 g, 59%). H1 NMR (300 MHz, CDC13): d 7.53 (d, "7 = 8, 8Hz, 2H), 7.43 (d," 7 = 1, 8Hz, 1H), 7.35 (d, "7 = 8, 8Hz, 2H), 7.30 (d, «7 = 1, 8Hz, 1H), 3.68 (s, 3H), 3.03 (c,« 7 = 7, 3Hz, 2H), 2 , 73 (t, «7 = 6, 9Hz, 2H), 1.99 (t,« 7 = 6, 9Hz, 2H), 1.61 (s, 6H), 1.40 (s, 6H), 1 , 25 (t, «7 = 7, 3Hz, 3H).
Methyl ester of acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -4-ethyl-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl] -phenyl} -2-methyl-propionic (Intermediate 182)
Following General Procedure C and using 2- [4- (4-ethyl-8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -phenyl] methyl ester] -2-methyl-propionic (Intermediate 181, 0.095 g, 0.23 mmol) in dichloromethane (3 ml) and acetonitrile (1.5 ml), cyclopropylamine (1 ml, 14.5 mmol), acetic acid (1 ml) and sodium cyanoborohydride (0.12 g, 1.91 mmol) followed by work-up gave an intermediate in the form of an oil, which was used as such for the next step. The intermediate (crude 0.23 mmol, 0.13 g) was dissolved in acetone (6 ml) and treated with potassium carbonate (0.23 g, 1.66 mmol) and methyl iodide (1.5 ml, 25 mmol). The resulting reaction mixture was stirred at room temperature overnight. The solids were separated by filtration, the filtrate and the washings were evaporated in vacuo to an oil. Flash column chromatography on silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent afforded the title compound (0.07, 65%). H1 NMR (300 MHz, CDC13): d 7.55 (d, «7 = 8, 8Hz, 2H), 7.43 (d,« 7 = 1, 7Hz, 1H), 7.37 (d, «7 = 8, 8Hz, 2H), 7.22 (d, «7 = 1, 7Hz, 1H), 4.13 (m, 1H), 3.72 (s, 3H), 2.78-2.68 ( m, 2H), 2.32-2.24 (m, 1H), 2.25 (s, 3H), 2.18-2.08 (m, 1H), 1.99-1.79 (m, 2H), 1.65 (s, 6H), 1.63-1.53 (m, 1H), 1.42 (s, 3H), 1.29 (s, 3H), 1.23 (t, « 7 = 7, 3Hz, 3H), 0.50-0.40 (m, 3H), 0.30-0.20 (m, 1H).
Acid 2-. { 4- [5- (cyclopropyl-methyl-amino) -4-ethyl-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} -2-methyl-propionic (Compound 54)
A solution of 2- methyl acid ester. { 4- [5- (cyclopropyl-methyl-amino) -4-ethyl-8,8-dimethyl-5,6,7,8-tetrahydro-naphthalen-2-ethynyl] -phenyl} -2-methyl-propionic (Intermediate 182, 0.035 g, 0.076 mmol) in methanol (3 mL) and tetrahydrofuran (2 mL) was treated with 3M potassium hydroxide (2 mL, 4 mmol) and the resulting reaction mixture was heated at 80 ° C for 2 days. The reaction mixture was neutralized with ammonium chloride and extracted with ethyl acetate. The organic phase was washed with water and brine, and dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to provide a residue which was purified by preparative reverse phase HPLC to provide the title product (0.023 g, %). NMR H1 (300 MHz, CDCl3): d 7.49 (d, "7 = 8, 4Hz, 2H), 7.36-7.26
(m, 3H), 7.16 (d, "7 = 1, 7Hz, 1H), 4.06 (m, 1H), 2.71-2.63 (m,
2H), 2.25-2.17 (m, 1H), 2.18 (s, 3H), 2.05-2.00 (m, 1H), 1.95-1.78 (m, 2H) , 1.60-1.50 (m, 1H), 1.58 (s, 6H), 1.35 (s, 3H), 1.22 (s, 3H), 1.16 (t, "7 = 7, 3Hz, 3H), 0.4-0.3 (m, 3H), 0.2-0.1 (m, 1H).
Intermediate 184 Intermediate 173 3. LiOH, EtOH, H20
4. HCOOH,], 4-dioxane Reaction Scheme 30
Ester 4,4, 7-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-yl of trifluoromethanesulfonic acid (Intermediate 183)
A stirred, cooled solution (-78 ° C) of 4,4,7-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 173, 0.95 g, 3.33 mmol ) in anhydrous tetrahydrofuran (10 ml) under argon was treated with a 1 M solution of sodium bis (trimethylsilyl) amide in tetrahydrofuran (5 ml, 5 mmol). After lh, N-phenyltrifluoromethanesulfonimide (1.08 g, 3.33 mmol) was added and the reaction mixture was stirred at room temperature for lh. The reaction was quenched with a saturated aqueous solution of ammonium chloride, diluted with water and extracted with diethyl ether (x2). The combined organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to flash chromatography on silica gel using 2-4% ethyl acetate in hexane as eluent to provide the title compound. title (0.73 g, 52%). H NMR (300 MHz, CDC13): d 7.08 (s, 1H), 6.92 (s, 1H), 5.67 (t, 2H, "7 = 5.0Hz), 2.15 (s, 3H), 2.08 (d, 2H, "7 = 5.0Hz), 1.00 (s, 6H), 0.00 (s, 9H).
Ethyl ester of 4,4,7-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 184)
Following General Procedure E and using 4,4, 7-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-yl ester of trifluoromethanesulfonic acid (Intermediate 183, 0.73 g, 1.75 mmol) , palladium acetate
(0.1 g, 0.45 mmol), 1,3-bis (diphenylphosphino) propane (0.1 g, 0.24 mmol), N, N-dimethylformamide (3.5 ml), ethanol (3.5 ml) and triethylamine (3.5 ml) followed by flash column chromatography on silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent was obtained the title compound (0.435 g, 73 %). NMR H1 (300 MHz, CDC13): d 7.42 (s, 1H), 7.10 (s, 1H), 6.76 (t, 2H, "7 = 5.0Hz), 4.04 (c, 2H, "7 = 7.0Hz), 2.15 (s, 3H), 2.02 (d, 2H," 7 = 5.0Hz), 1.09 (t, 3H, "7 = 7.0Hz) , 0.97 (s, 6H), 0.00 (s, 9H).
Ethyl ester of 6-ethynyl-4, 4, 7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 185)
Following General Procedure F and using 4,4,7-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester
(Intermediate 184, 0.43 g, 1.3 mmol), ethanol (4 ml) and potassium carbonate (0.84 g, 6.06 mmol), the title compound was obtained (0.33 g, 95% ). NMR H1 (300 MHz, CDC13): d 7.70 (s, 1H), 7.40 (s, 1H), 7.05 (t, 2H, "7 = 5.0Hz), 4.30 (c, 2H, "7 = 7.0Hz), 2.43 (s, 3H),
2.30 (d, 2H, "7 = 5.0Hz), 1.36 (t, 3H," 7 = 7.0Hz), 1.23 (s,
6H).
Ethyl 6- (4-methoxycarbonylmethyl-phenylethynyl) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Intermediate 186)
Following General Procedure B and using ethyl ester of 6-ethynyl-4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 185, 0.126 g, 0.47 mmol), methyl ester of 4-iodophenylacetic acid (0.13 g, 0.47 mmol), triethylamine (2 ml), copper (I) iodide (0.029 g, 0.15 mmol) and dichlorobis (triphenylphosphine) palladium
(II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10-12% ethyl acetate in hexane as eluent, the title compound was obtained in the form of a viscous oil (0.144 g, 74%). H NMR (300 MHz, CDCl 3): d 7.72 (s, 1 H), 7.47 (d, 2 H, "7 = 8.1 Hz), 7.35 (s, 1 H), 7.27 (d, 2H, "7 = 8.1Hz), 7.05 (t, 2H," 7 = 5.0Hz), 4.34 (c, 2H, "7 = 7.0Hz), 3.70 (s, 3H) , 3.64 (s, 2H), 2.48 (s, 3H), 2.32 (d, 2H, "7 = 5.0Hz), 1.38 (t, 3H," 7 = 7.0Hz) , 1.27 (s, 6H).
Ethyl 6- (4-carboxymethyl-phenylethynyl) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Compound 55)
A solution of 6- (4-carboxymethyl-phenylethynyl) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 186, 0.144 g, 0.35 mmol) in ethanol (2 ml) was treated with a 1 M solution of lithium hydroxide (1 ml, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 3 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was purified by preparative reverse phase HPLC using 10% water in acetonitrile as mobile phase to give the title product (0.071 g, fifty%). NMR H1 (300 MHz, CDCl3): d 7.68 (s, 1H), 7.47 (broad d, 2H, "7 = 8.1Hz), 7.41 (s, 1H), 7.21 (d width, 2H), 7.04 (t, 2H, «7 = 5.0Hz), 4.31 (c, 2H,« 7 = 7.0Hz), 3.65 (s wide, 2H), 2.46 (s, 3H), 2.30 (d, 2H, "7 = 5.0Hz), 1.37 (t, 3H," 7 = 7.0Hz), 1.24 (s, 6H).
Ethyl 6- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Intermediate 187)
Following General Procedure B and using ethyl ester of 6-ethynyl-4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 185, 0.2 g, 0.75 mmol), ester methyl 2-fluoro-4-iodophenylacetic acid (0.22 g, 0.75 mmol), triethylamine (2 ml), copper iodide (I) (0.03 g, 0.16 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.1 g, 0.14 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 10-12% ethyl acetate in hexane as eluent, the title in the form of a viscous oil (0.23 g, 73%). H NMR (300 MHz, CDCl3): d 7.73 (s, 1H), 7.42 (s, 1H), 7.30-7.20 (m, 3H), 7.06 (t, 2H, « 7 = 5.0Hz), 4.32 (c, 2H, «7 = 7.0Hz), 3.71 (s, 3H), 3.68 (s, 2H), 2.47 (s, 3H), 2.32 (d, 2H, "7 = 5.0Hz), 1.37 (t, 3H," 7 = 7.0Hz), 1.26 (s, 6H).
Ethyl 6- (4-carboxymethyl-3-fluoro-phenylethynyl) -4,4,7-trimethyl-3,4-dihydro-naphthalene-l-carboxylic acid ester (Compound 56)
A solution of 6- (4-carboxymethyl-3-fluoro-phenylethynyl) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 187, 0.24 g, 0 , 54 mmoles) in ethanol (2 ml) was treated with a 2 M solution of lithium hydroxide (1 ml, 2 mmoles) and the resulting reaction mixture was stirred at room temperature for 3 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was purified by preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase to provide the title product (0.05. g, 22%). H NMR (300 MHz, CDC13): d 7.69 (s, 1H), 7.41 (s, 1H), 7.27-7.19 (m, 3H), 7.05 (t, 2H, « 7 = 4.7Hz), 4.32 (c, 2H, «7 = 7.0Hz), 3.64 (broad s, 2H), 2.45 (s, 3H), 2.31 (d, 2H, "7 = 4.7Hz), 1.37 (t, 3H," 7 = 7.0Hz), 1.25 (s, 6H).
6- (t-Butyl-dimethyl-silanyloxy) -4,4, 7-trimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 188)
A solution of 6-hydroxy-4,4,7-trimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 171, 2.04 g, 10 mmol) in
N, N-dimethylformamide anhydrous (10 ml) in argon was treated with imidazole (1 g, 14.7 mmol) followed by t-butyldimethylsilyl chloride (1.5 g, 10 mmol). After stirring the reaction mixture at room temperature overnight, it was poured into water and extracted with diethyl ether
(x2) The combined organic phase was dried over anhydrous sodium sulfate, filtered and evaporated to a residue which was purified by flash column chromatography on silica gel (230-400 mesh) using 8-14% ethyl acetate in hexane as eluent to provide the title compound (2.5 g, 79%). NMR H1 (300 MHz, CDCl3): d 7.74 (s, 1H), 6.65 (s, 1H), 2.56 (t, 2H, "7 = 6.8Hz), 2.09 (s, 3H), 1.88 (t, 2H, "7 = 6.8Hz), 1.24 (s, 6H), 0.93 (s, 9H), 0.17 (s, 6H).
Ester 6- (t-butyl-dimethyl-silanyloxy) -4,4, 7-trimethyl-3,4-dihydro-naphthalene-1-yl of trifluoromethanesulfonic acid (Intermediate 189)
A stirred, cooled solution (-78 ° C) of 6- (t-butyl-dimethyl-silanyloxy) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-yl ester of trifluoromethanesulfonic acid
(Intermediate 188, 2.53 g, 8 mmol) in anhydrous tetrahydrofuran (25 ml) in argon was treated with a 1 M solution of sodium bis (trimethylsilyl) amide in tetrahydrofuran (12 ml, 12 mmol). After lh, N-phenyltrifluoromethanesulfonimide (4.28 g, 12 mmol) was added and the reaction mixture was stirred at room temperature for 1 h. The reaction was quenched with a saturated aqueous solution of ammonium chloride, diluted with water and extracted with diethyl ether (x2). The combined organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to flash chromatography on silica gel using 4% ethyl acetate in hexane as eluent to provide the title compound ( 1.4 g, 39%). NMR H1 (300 MHz, CDCl3): d 6.90 (s, 1H), 6.49 (s, 1H), 5.53 (t, 2H, "7 = 5.0Hz), 2.09 (d, 2H, "7 = 5.0Hz), 1.95 (s, 3H), 1. 01 (s, 6H), 0.78 (s, 9H), 0.00 (s, 6H).
Ethyl ester of 6- (t-butyl-dimethyl-silanyloxy) -4,4,7-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 190) Following General Procedure E and using ester 6- ( t-butyl-dimethyl-silanyloxy) -4,4, 7-trimethyl-3,4-dihydro-naphthalene-1-yl of trifluoromethanesulfonic acid (Intermediate 189, 3.4 g, 7.55 mmol), palladium (0.36 g, 1.62 mmol), 1,3-bis (diphenylphosphino) propane (0.36 g, 0.86 mmol), N, N-dimethylformamide (7 mL), ethanol (7 mL) and triethylamine (7 ml) followed by flash column chromatography on silica gel (230-400 mesh) using 7% ethyl acetate in hexane as eluent gave the title compound (1.35 g, 48%). H NMR (300 MHz, CDCl 3): d 7.40 (s, 1H), 6.65 (t, 2H, "7 = 5.0Hz), 6.65 (s, 1H), 4.08 (c, 2H, "7 = 7.0Hz), 2.04 (d, 2H," 7 = 5.0Hz), 1.96 (s, 3H), 1.13 (t, 3H, "7 = 7.0Hz) , 0.99 (s, 6H), 0.79 (s, 9H), 0.00 (s, 6H). Ethyl 6- (t-butyl-dimethyl-silanyloxy) -4,4,7-trimethyl-l, 2,3,4-tetrahydro-naphthalene-1-carboxylic acid ester (Intermediate 191)
A solution of 6- (t-butyl-dimethyl-silanyloxy) -4,4,7-trimethyl-3,4-dihydro-naphthalene-l-carboxylic acid ethyl ester (Intermediate 190, 0.95 g, 2.54 mmoles) in ethanol was treated with a suspension of palladium on carbon at 5%
(0.3 g) in ethyl acetate (0.5 ml) and the resulting reaction mixture was stirred under a hydrogen atmosphere overnight. The solids were filtered on a pad of celite and the filtrate was evaporated in vacuo to give the title compound as a viscous oil (0.95 g, -100%). NMR H1 (300 MHz, CDC13): d 6.66 (s, 1H), 6.51 (s, 1H), 3.95 (c, 2H, "7 = 7.0Hz), 3.46 (m, 1H), 1.92 (s, 3H), 1.93-1.75
(m, 2H), 1.64-1.55 (m, 1H), 1.38-1.30 (m, 1H), 1.06 (s,
3H), 1.01 (t, 3H, "7 = 7.0Hz), 1.01 (s, 3H), 0.80 (s, 9H), 0.00 (s, 6H).
Ethyl ester of 4,4,7-trimethyl-6-trifluoromethanesulfonyloxy-1,2,3,4-tetrahydro-naphthalene-1-carboxylic acid (Intermediate 192)
The ethyl ester of 6- (t-butyl-dimethyl-silanyloxy) -4,4,7-trimethyl-1,2,3,4-tetrahydro-naphthalene-1-carboxylic acid (Intermediate 191, 0.95 g, 2) , 54 mmol) was treated with a 1 M solution of tetra-n-butylammonium fluoride in tetrahydrofuran (4 mL, 2 mmol) in argon and the resulting reaction mixture was stirred at room temperature for 45 min. Water was added and the reaction mixture was extracted with 10% ethyl acetate in diethyl ether. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to an oil which was used for the next step. The oil was dissolved in anhydrous dichloromethane under argon and treated with 4- (dimethylamino) pyridine (0.62 g, 5.1 mmol) and N-phenyltrifluoromethanesulfonimide (0.91 g, 2.54 mmol). After ih at room temperature, the reaction mixture was subjected to flash column chromatography using 8% ethyl acetate in hexane as eluent to give the title compound as an oil (0.86 g, 86% ). NMR H1 (300 MHz, CDC13): d 7.19 (s, 1H), 7.07 (s, 1H), 4.17 (c, 2H, J = 7.0Hz), 3.73 (t, 1H) , "7 = 5.9Hz), 2.30 (s, 3H), 2.18-1.97 (m, 2H), 1.87-1.78 (m, 1H), 1.70-1, 56 (m, 1H), 1.31-1.25 (2s, 3H and It, 3H, overlapping).
Ethyl ester of 4,4,7-trimethyl-6-trimethylsilanylethynyl-1, 2,3,4-tetrahydro-naphthalene-l-carboxylic acid (Intermediate 193)
Following General Procedure D in a sealed tube and using 4,4, 7-trimethyl-6-trifluoromethanesulfonyloxy-1, 2,3,4-tetrahydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 192, 0.86 g , 2.2 mmol), triethylamine (2 ml), copper iodide (I) (0.083 g, 0.44 mmol), trimethylsilylacetylene (2 ml, 14 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.306 g, 0.44 mmole) followed by flash column chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent, and preparative normal phase HPLC using 5% ethyl acetate in hexane as the phase mobile in order to separate the starting material recovered from the product, the title compound (0.26 g) was obtained. H NMR (300 MHz, CDCl 3): d 7.21 (s, 1H), 6.72 (s, 1H), 3.95 (c, 2H, "7 = 7.0Hz), 3.49 (t, 1H, "7 = 5.8Hz), 2.13 (s, 3H), 1.95-1.62 (m, 2H), 1.60-1.48 (m, 1H), 1.42-1 , 31 (m, 1H), 1.10-1.00 (2s, 3H and lt, 3H, overlapping), 0.04 (s, 9H).
Ethyl 6- (4-t-butoxycarbonylmethyl-phenylethynyl) -4,4,7-trimethyl-1,2,3,4-tetrahydro-naphthalene-1-carboxylic acid ester (Intermediate 194)
The ethyl ester of 4,4,7-trimethyl-6-trimethylsilanylethynyl-1,2,3,4-tetrahydro-naphthalene-1-carboxylic acid (Intermediate 193, 0.26 g, 0.76 mmol) was treated with a 1 M solution of tetra-n-butylammonium fluoride in tetrahydrofuran (3 ml, 3 mmol) in argon and the resulting reaction mixture was stirred at room temperature for 1 h. Water was added and the reaction mixture was extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to an oil which was used as such for the next step. Following General Procedure B and using the oil (0.76 mmol), 4-iodo-butylphenyl acetate (Reagent 10, 0.23 g, 0.72 mmol), triethylamine (2 ml), copper iodide (I ) (0.06 g, 0.32 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.14 g, 0.2 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using acetate of 12% ethyl in hexane as eluent, the title compound was obtained as a pale yellow, viscous oil (0.23 g, 66%). H1 NMR (300 MHz, CDC13): d 7.50 (s, 1H), 7.48 (d, 2H, "7 = 8.5Hz), 7.24 (d, 2H," 7 = 8.5Hz) , 6.98 (s, 1H), 4.17 (c, 2H, "7 = 7.0Hz), 3.74 (t, 1H," 7 = 5.8Hz), 3.52 (s, 2H) , 2.42 (s, 3H), 2.27-1.99 (m, 2H), 1.87-1.78 (m, 1H), 1.63-1.44 (m, 1H), 1 , 43 (s, 9H), 1.32 (s, 3H), 1.26 (s, 3H), 1.23 (t, 3H, hidden).
Ethyl 6- (4-carboxymethyl-phenylethynyl) -4,4,7-trimethyl-1,3,4,4-tetrahydro-naphthalene-1-carboxylic acid ester (Compound 57)
A solution of 6- (4-t-butoxycarbonylmethyl-phenylethynyl) -4,4,7-trimethyl-1,2,3,4-tetrahydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 194, 0.23 g , 0.5 mmol) in 1,4-dioxane (1 ml) was treated with formic acid (3 ml) and the resulting reaction mixture was stirred at room temperature for 6 h. Water was added and the reaction mixture was extracted with ethyl acetate (x2). The combined organic phase was washed with water, dried over anhydrous magnesium sulfate, filtered and evaporated to an oil. Preparative reverse phase HPLC using 10% water in acetonitrile as the mobile phase gave the title compound (0.15 g, 74%). NMR H1 (300 MHz, CDC13): d 7.48 (s, 1H), 7.46 (broad d, 2H), 7.23 (broad d, 2H), 6.96 (s, 1H), 4, 17 (c, 2H, "7 = 7.0Hz), 3.73 (t, 1H," 7 = 5.8Hz), 3.54 (broad s, 2H), 2.40 (s, 3H), 2 , 29-1.95 (m, 2H), 1.85-1.77 (m, 1H), 1.62-1.44 (m, 1H), 1.31 (s, 3H), 1.26 (s, 3H), 1.25 (t, 3H, hidden)
Intermediate 195
Intermediate 199
Intermediate 197
R2 = H Compound 58 R2 = H R2 = F Compound 59 R2 = F 3. LiOH, H20, EtOH,
Reaction Scheme 31-Hydroxy-2,4,4-trimethyl-3,4-dihydro-2H-naph talen-1-one (Intermediate 195)
A solution 6-methoxy-2,4,4-trimethyl-3,4-dihydro-2H-naphthalen-1-one (described in the Journal of Pharmaceutical Sciences, 1970, 59 (6), pp. 869-870, Floyd et al, incorporated herein by reference: 1.2 g, 5.5 mmol) and sodium cyanide (2 g, 41 mmol) in anhydrous dimethyl sulfoxide (15 ml) was heated at 230 ° C for 24 h in argon. The reaction mixture was then cooled to room temperature, poured into ice and acidified (Caution! Evolution of hydrogen cyanide!) With dilute hydrochloric acid and extracted with ethyl acetate (x2). The combined organic extract was washed with brine (xl), dried over anhydrous sodium sulfate, filtered and evaporated to give the title compound, which was used as such for the next step (1 g, 89%).
6.8, 8-Trimethyl-5-oxo-5, 6,7,8-tetrahydro-naphthalene-2-yl ester of trifluoromethanesulfonic acid (Intermediate 196)
A solution of 6-hydroxy-2,4,4-trimethyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 195, 1 g, 5 mmol) and 4- (dimethylamino) pyridine (1.22 g) , 10 mmol) in anhydrous dichloromethane (10 ml) was treated with N-phenyltrifluoromethanesulfonimide (1.78 g, 10 mmol), and the resulting reaction mixture was stirred at room temperature for 2 h. Flash column chromatography of the reaction mixture on silica gel
(230-400 mesh) using 5% ethyl acetate in hexane as eluent afforded the title compound as a white solid (1.45 g, 86%). H1 NMR (300 MHz, CDC13): d 8.06 (d, 1H, "7 = 8.5Hz), 7.25 (d, 1H," 7 = 2.0Hz), 6.79 (dd, 1H, "7 = 8.5, 2.0Hz), 2.79 (m, 1H), 1.94 (m, 2H), 1.41 (s, 3H), 1.37 (s, 3H), 1, 22 (d, 3H, "7 = 6.7Hz).
2,4, 4-Trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 197)
Following General Procedure D and using 6,8,8-trimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalene-2-yl ester of trifluoromethanesulfonic acid (Intermediate 196,
1.45 g, 4.3 mmol), triethylamine (5 ml), copper iodide (I) (0.21 g, 0.26 mmol), trimethylsilylacetylene (3 ml, 21 mmol) and dichlorobis (triphenylphosphine) palladium ( II)
(0.75 g, 1.07 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent, the title compound was obtained (1.28 g. , -100%). NMR H1 (300 MHz, CDC13): d 7.64 (d, 1H, "7 = 7.9 Hz), 7.22 (d, 1H," 7 = 2.0Hz), 7.08 (dd, 1H) , "7 = 7.9, 2.0Hz), 2.50 (m, 1H), 1.94 (d, 2H," 7 = 8.8Hz)), 1.13 (s, 3H), 1, 08 (s, 3H), 0.96 (d, 3H, "7 = 6.8Hz), 0.00 (s, 9H).
2,4,5-Trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-yl ester of trifluoro-methanesulfonic acid (Intermediate 198)
A stirred, cooled solution (ice bath) of
2,4,4-Trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-2H-naphthalen-1-one (Intermediate 197, 1.28 g, 4.5 mmol) in anhydrous dichloromethane (10 ml) was treated with 2%. , 6-di-t-butyl-4-methylpyridine (2.04 g, 9.91 mmol) and trifluoromethanesulfonic anhydride (1.52 mL, 9 mmol) and the resulting reaction mixture was stirred at room temperature for 5 days at the end of which it was subjected to flash chromatography on silica gel (230-400 mesh) using 5% ethyl acetate in hexane as eluent to give the title compound as an oil (1.59 g, 85% ). H1 NMR (300 MHz, CDC13): d 7.09 (d, 1H, J = 7.9Hz), 7.07 (d, 1H, "7 = 1.5Hz), 6.98 (dd, 1H, J = 7.9, 1.5Hz), 2.04 (s, 2H), 1.72 (s, 3H), 1.03 (s, 6H), 0.00 (s, 9H).
Ethyl ester of 2,4,4-trimethyl-β-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester
(Intermediate 199)
Following General Procedure E and using 2,4,4-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-1-yl ester of trifluoromethanesulfonic acid
(Intermediate 198, 1.59 g, 3.8 mmol), palladium acetate (0.1 g, 0.45 mmol), 1,3-bis (diphenylphosphino) propane (0.1 g, 0.24 mmol) , N, N-dimethylformamide (2.4 ml), ethanol (2.4 ml) and triethylamine (2.4 ml) followed by flash column chromatography on silica gel (230-400 mesh) using ethyl acetate 5%. % in hexane as eluent was obtained the title compound (0.31 g, 24%) as a yellow oil. H1 NMR (300 MHz, CDC13): d 7.12 (d, 1H, "7 = 1.5Hz), 7.01 (dd, 1H," 7 = 8.2, 1.8Hz), 6.77 ( d, 1H, "7 = 8.2Hz), 4.10 (c, 2H," 7 = 7.0Hz), 1.93 (s, 2H), 1.73 (s, 3H), 1.08 ( t, 3H, "7 = 7.0Hz), 0.99 (s, 6H), 0.00 (s, 9H).
Ethyl ester of 6-ethynyl-2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 200)
Following the general procedure F and using 2, 4, 4-trimethyl-6-trimethylsilanylethynyl-3,4-dihydro-naphthalene-l-carboxylic acid ethyl ester
(Intermediate 199, 0.31 g, 0.92 mmol), ethanol (2 ml) and potassium carbonate (0.3 g, 2.2 mmol), the title compound was obtained (0.26 g, > 100%). H1-NMR (300 MHz, CDC13): d 7.32 (d, 1H, "7 = 1.5Hz), 7.20 (dd, 1H," 7 = 8.2, 1.5Hz), 6.96 ( d, 1H, "7 = 8.2Hz), 4.27 (c,
2H, "7 = 7.0Hz), 3.00 (s, 1H), 2.10 (s, 2H), 1.90 (s, 3H),
1.27 (t, 3H, "7 = 7.0Hz), 1.16 (s, 6H).
Ethyl 6- (4-methoxycarbonylmethyl-phenylethynyl) -2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Intermediate 201)
Following General Procedure B and using ethyl ester of 6-ethynyl-2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (Intermediate 200, 0.106 g, 0.38 mmol), methyl ester of 4-iodophenylacetic acid (0.106 g, 0.38 mmol), triethylamine (2 ml), copper iodide (I)
(0.02 g, 0.105 mmol) and dichlorobis (triphenylphosphine) palladium
(II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 12-15% ethyl acetate in hexane as eluent, the title compound was obtained a light yellow oil form (0.075 g, 47%). H NMR (300 MHz, CDCl3): d 7.49 (d, 2H, "7 = 7.9Hz), 7.45 (d, 1H," 7 = 1.5Hz), 7.32 (dd, 1H, «7 = 7.9, 1.5Hz), 7.26 (d, 2H,« 7 = 7.9Hz), 7.07 (d, 1H, «7 = 7.9Hz), 4.37 (c, 2H, "7 = 7.0Hz), 3.70 (s, 3H), 3.63 (s, 2H), 2.22 (s, 2H), 2.00 (s, 3H), 1.38 ( t, 3H, "7 = 7.0Hz), 1.27 (s, 6H).
Ethyl 6- (4-carboxymethyl-phenylethynyl) -2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Compound 58)
A solution of 6- (4-carboxymethyl-phenylethynyl) -2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester (0.075 g, 0.18 mmol) in ethanol (2 ml) ) was treated with a 1 M solution of lithium hydroxide (1 ml, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 0.5 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to provide the title product (0.055 g, 76%). H NMR (300 MHz, CDCl3): d 7.48 (d, 2H, "7 = 7.9Hz), 7.44 (d, 1H," 7 = 1.5Hz), 7.31 (dd, 1H, «7 = 7.9, 1.7Hz), 7.23 (broad d, 2H,« 7 = 7.7Hz), 7.06 (d, 1H, «7 = 7.9Hz), 4.36 (c , 2H, "7 = 7.0Hz), 3.60 (broad s, 2H), 2.20 (s, 2H), 1.99 (s, 3H), 1.37 (t, 3H," 7 = 7.0Hz), 1.26 (s, 6H).
Ethyl 6- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Intermediate 202)
Following General Procedure B and using ethyl ester of 6-ethynyl-2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid (0.16 g, 0.59 mmol), methyl ester of acid 2-fluoro-4-iodophenylacetic acid (Intermediate 200, 0.16 g, 0.56 mmol), triethylamine (2 ml), copper iodide (I) (0.07 g, 0.37 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.11 g, 0.16 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 15% ethyl acetate-in hexane as eluent, the title in the form of a viscous oil (0.15 g, 58%). H1 NMR (300 MHz, CDCl3): d 7.44 (d, 1H, "7 = 1.5Hz), 7.32 (dd, 1H," 7 = 7.9, 1.5Hz), 7.30- 7.19 (m, 3H), 7.08 (d, 1H, "7 = 7.9Hz), 4.37 (c, 2H," 7 = 7.0Hz), 3.71 (s, 3H), 3.68 (s,
2H), 2.21 (s, 2H), 2.00 (s, 3H), 1.37 (t, 3H, "7 = 7.0Hz),
1.27 (s, 6H).
Ethyl 6- (4-carboxymethyl-3-fluoro-phenylethynyl) -2,4,4-trimethyl-3,4-dihydro-naphthalene-l-carboxylic acid ester (Compound 59)
A solution of 6- (4-carboxymethyl-3-fluoro-phenylethynyl) -2,4,4-trimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 202, 0.15 g, 0 , 35 mmol) in ethanol (2 ml) was treated with a 1 M solution of lithium hydroxide (1 ml, 1 mmol) and the resulting reaction mixture was stirred at room temperature for 0.5 h. The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product (0.1 g, 67%). NMR H1 (300 MHz, CDCl3): d 7.44 (d, 1H, "7 = 1.5Hz), 7.32 (dd, 1H," 7 = 8.2, 1.5Hz), 7.22- 7.18 (m, 3H), 7.07 (d, 1H, "7 = 7.9Hz), 4.36 (c, 2H, 7 = 7.0Hz), 3.66 (s wide, 2H), 2.20
(s, 2H), 1.99 (s, 3H), 1.37 (t, 3H, "7 = 7.0Hz), 1.26 (s, 6H).
Ethyl ester of [4- (8,8-dimethyl-5-trifluoromethanesulfonyloxy-7,8-dihydro-naphthalen-2-ylethynyl) -2-fluoro-phenyl] -acid acid (Intermediate 203)
A solution of [4- (8,8-dimethyl-5-oxo-5,6,7,8-tetrahydro-naphthalen-2-ylethynyl) -2-fluoro-phenyl] -acetic acid methyl ester (Patent of the United States No. 6,252,090; 0.28 g, 0.77 mmol) in anhydrous dichloromethane (5 ml) was treated with 2,6-di-t-butyl-4-methylpyridine (0.189 g, 0.92 mmol) and trifluoromethanesulfonic anhydride (0.136 ml, 0.81 mmol) and the resulting reaction mixture was stirred at room temperature for 4 h at the end of which it was diluted with water and extracted with ethyl acetate. The organic extract was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated in vacuo to a residue which was subjected to flash chromatography on silica gel (230-400 mesh) using ethyl acetate at room temperature. % in hexane as eluent to give the title compound as a pale orange oil (0.32 g, 84%). NMR H1 (300 MHz, CDC13): d 7.46-7.22 (m, 6H), 6.00 (t, «7 = 4, 8Hz, 1H), 3.72 (s, 3H), 3, 70 (s, 2H), 2.41 (d, "7 = 4, 8Hz, 2H), 1.33 (s, 6H).
Ethyl 6- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -4,4-dimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Intermediate 204) Following General Procedure E and using acid methyl ester [4- (8,8-dimethyl-5-trifluoromethanesulfonyloxy-7,8-dihydro-naphthalen-2-ylethynyl) -2-fluoro-phenyl] -acetic (Intermediate 203, 0.32 g, 0.65 mmol), palladium acetate (0.015 g, 0.064 mmol), 1,3-bis (diphenylphosphino) propane (0.027 g, 0.064 mmol), N, N-dimethylformamide (5 ml), ethanol (2 ml) and triethylamine (2 ml) followed of flash column chromatography on silica gel (230-400 mesh) using 5-15% ethyl acetate in hexane as eluent gave the title compound (0.15 g, 55%) as a color oil yellow. 1 H-NMR (300 MHz, CDCl 3): d 7.84 (d, 7 = 8, 2Hz, 1H), 7.47 (d, "7 = 1.7Hz, 1H), 7.37 (dd," 7 = 8.2, 1.7Hz, 1H), 7.30-7.15 (m, 3H), 7.08 (t, «7 = 4, 8Hz, 1H), 4.31 (c,« 7 = 7 , 0Hz, 2H), 3.71 (s, 3H), 3.68 (s, 2H), 2.34 (d, «7 = 4, 8Hz, 2H), 1.37 (t,« 7 = 7 , 0Hz, 3H), 1.28 (s, 6H).
Ethyl 6- (4-carboxymethyl-3-fluoro-phenylethynyl) -4,4-dimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ester (Compound 60)
A solution of 6- (3-fluoro-4-methoxycarbonylmethyl-phenylethynyl) -4,4-dimethyl-3,4-dihydro-naphthalene-1-carboxylic acid ethyl ester (Intermediate 204, 0.15 g, 0.36 mmoles) in ethanol (3 ml) and tetrahydrofuran (3 ml) was treated with a 2M solution of lithium hydroxide (1.5 ml, 3 mmol) and the resulting reaction mixture was stirred at room temperature for 1.5 hrs. . The volatiles were evaporated in vacuo, the residue was neutralized with a saturated aqueous solution of ammonium chloride and extracted with diethyl ether and ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was purified by preparative reverse phase HPLC using 5% water in acetonitrile as the mobile phase to give the title product (0.04 g). g, 27%). H1 NMR (300 MHz, CDC13): d 7.81
(d, «7 = 8, 2Hz, 1H), 7.46 (d,« 7 = 1, 7Hz, 1H), 7.37 (dd, «7 = 8.2 yl, 7Hz, 1H), 7, 27-7.09 (m, 3H), 7.07 (t, «7 = 4, 8Hz, 1H), 4.31 (c,« 7 = 7, 0Hz, 2H), 3.66 (s, 2H) ), 2.33 (d, J = 4.8 Hz, 2H), 1.37
(t, "7 = 7, 0Hz, 3H), 1.27 (s, 6H).
3. TBAF, Intermediate THF 205
Reagent 11
Reaction Scheme 32 Isopropyl Ester 3,5-dibromobenzoic acid (Intermediate 205)
A solution of 3,5-dibromobenzoic acid (Aldrich, 2.4 g, 8.6 mmol) in benzene (150 ml) and isopropanol (50 ml) was treated with concentrated sulfuric acid (2 ml) and heated to reflux for the night using a Dean-Stark water trap. The volatiles were evaporated in vacuo, the residue was diluted with water and extracted with diethyl ether. The organic phase was washed with water and a saturated aqueous sodium bicarbonate solution, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a clear oil which was used as is. for the next stage (2.7 g, -100%).
3, 5-Diethynyl-benzoic acid isopropyl ester (Intermediate 206)
Following General Procedure D and using 3,5-dibromo-benzoic acid isopropyl ester (Intermediate 205, 2.7 g, 8.6 mmol), triethylamine (30 ml), copper (I) iodide (0.45 g) , 2.4 mmol), trimethylsilylacetylene (6.8 ml, 48 mmol) and dichlorobis (triphenylphosphine) palladium (II) (1.75 g, 2.4 mmol) followed by flash column chromatography on silica gel (230 mesh) -400) using 3% ethyl acetate in hexane as eluent, the isopropyl 3,5-bis-trimethylsilanylethynyl-benzoic acid intermediate is obtained. The intermediate (2.8 g, 7.85 mmol) was treated with a 1 M solution of tetra-n-butylammonium fluoride in tetrahydrofuran (25 mL, 25 mmol) and the resulting reaction mixture was stirred in an ice bath. during Ih. Water was added and the reaction mixture was extracted with diethyl ether. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to an oil which was redissolved in diethyl ether (10 ml) and treated with hexane (150 ml). The precipitated solid was filtered and dried to provide the title compound (1.3 g, 78%). NMR H1 (300 MHz, CDC13): d 8.08 (d, 1H, "7 = 1.4Hz), 7.73 (d, 1H," 7 = 1.4Hz), 5.23 (heptet, 1H, "7 = 6.3 Hz), 3.13 (s, 2 H), 1.35 (d, 6 H," 7 = 6.1 Hz).
3-Ethynyl-5- [3-fluoro-4- (3-trimethylsilanyl-propoxycarbonylmethyl) -phenylethynyl] -benzoic acid isopropyl ester (Intermediate 207)
Following General Procedure B and using 3, 5-diethynyl-benzoic acid isopropyl ester (Intermediate 206, 0.36 g, 1.72 mmol), 2-trimethylsilane-ethyl ester of (2-fluoro-4-iodo) acid phenyl) -acetic (0.132 g, 0.86 mmol), triethylamine (8 ml), copper iodide (I) (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent, the title compound was obtained as a colorless oil (0, 15 g, 37%). H1NMR (300 MHz, CDC13): d 8.10 (m, 1H), 8.07 (m, 1H), 7.75 (m, 1H), 7.19-7.25 (m, 3H), 5.24 (hept, "7 = 6.2Hz, 1H), 4.19 (t," 7 = 8, 5Hz, 2H), 3.64 (s, 2H), 3.14 (s, 1H), 1.35 (d, "7 = 6, 2Hz, 6H), 0.97 (t," 7 = 8, 5Hz, 2H), 0.00 (s, 9H).
3- (4-Carboxymethyl-3-fluoro-phenylethynyl) -5-ethynyl-benzoic acid isopropyl ester (Compound 61)
A solution of 3-ethynyl-5- [3-fluoro-4- (3-trimethylsilanyl-propoxycarbonylmethyl) -phenylethynyl] -benzoic acid isopropyl ester (Intermediate 207, 0.15 g, 0.32 mmol) in anhydrous dimethylsulfoxide ( 4 ml) was treated with tetra-n-ethylammonium fluoride (0.19 ml, 1.3 mmol) and the resulting reaction mixture was stirred at room temperature for 5 min. Water was added and the reaction mixture was extracted with ethyl acetate. The organic phase was washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to an oil which was purified by recrystallization from ethyl acetate / hexane to give the title compound as a solid of color white (0.045 g, 38%). H NMR (300 MHz, CDCl 3): d .8.13 (m, 1H), 8.10 (, 1H), 7.78 (m, 1H), 7.23-7.30 (m, 3H), 5.29 (hept, "7 = 6, 4Hz, 1H), 3.74 (s, 2H), 3.15 (s, 1H), 1.38 (d," 7 = 6, 4Hz, 6H).
3- (4-Acetoxymethoxycarbonylmethyl-3-fluoro-phenylethynyl) -5-ethynyl-benzoic acid isopropyl ester (Compound 62)
Following General Procedure B and using 3,5-diethynyl-benzoic acid isopropyl ester (Intermediate
206, 0.27 g, 1.27 mmol), acetoxymethyl acid ester
(2-fluoro-4-iodo-phenyl) -acetic acid (0.224 g, 0.64 mmol), triethylamine (8 ml), copper (I) iodide (0.019 g, 0.1 mmol) and dichlorobis (triphenylphosphine) palladium (II) (0.07 g, 0.1 mmol) followed by flash column chromatography on silica gel (230-400 mesh) using 2.5-20% ethyl acetate in hexane as eluent, compound was obtained of the title in the form of an orange solid
(0.09 g, 32%). H1 NMR (300 MHz, CDC13): d 8.13 (m, 1H), 8.10 (m, 1H), 7.79
(m, 1H), 7.23-7.32 (m, 3H), 5.78 (s, 2H), 5.27 (hept,
J = 6.4Hz, 1H), 3.75 (s, 2H), 3.15 (s, 1H), 2.12 (s, 3H),, 38 (d, «7 = 6, 4Hz, 6H) Intermediate 36 Intermediate 208
Intermediate 210 Intermediate 211 Intermediate 212 Compound 63 Reaction Scheme 33
8-Iodo-2, 2, 4,4-tetramethylchroman-6-carboxylate ethyl (Intermediate 208)
A solution of ethyl 2, 2, 4, 4-tetramethylchroman-6-carboxylate (Intermediate 36, 0.733 g, 2.8 mmol) in anhydrous dichloromethane (10 ml) was treated with silver trifluoromethanesulfonate (I) (0.719 g, 2.8 mmol) and iodine (0.71 g, 2.8 mmol) and the resulting solution was stirred at room temperature for 4 h. The reaction mixture was treated with a saturated, aqueous solution of sodium thiosulfate and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to a residue which was subjected to column chromatography on silica gel (230-400 mesh) using 5-10% ethyl acetate in hexane as eluent to provide the title compound (0.88 g, 81%) a light yellow oil form. H1 NMR (300 MHz, CDC13): d 8.26 (d, 1H, "7 = 2.0Hz), 7.96 (d, 1H," 7 = 2.0Hz), 4.34 (c, 2H, "7 = 7.1 Hz), 1.87 (s, 2H), 1.40 (s, 6H), 1.37 (s, 6H), 1.41-1.35 (m, 3H).
8-Trimethylsilanylethynyl-2, 2,4, 4-tetramethylchroman-6-carboxylic acid ethyl ester (Intermediate 209)
A solution of ethyl 8-iodo-2, 2,4,4-tetramethylchroman-6-carboxylate (Intermediate 208, 0.88 g, 2.26 mmol) in triethylamine (10 ml) was treated with copper iodide (I )
(0.043 g, 0.266 mmol) and purged with argon for 5 minutes. Then trimethylsilylacetylene (3 ml,
21.22 mmole) followed by dichlorobis (triphenylphosphine) palladium
(II) (0.159 g, 0.226 mmol). The resulting reaction mixture was heated at 70 ° C overnight in a sealed tube. It was then cooled to room temperature, diluted with diethyl ether and filtered on a pad of celite. The filtrate was evaporated off vacuo to an oil which was subjected to column chromatography on silica gel (230-400 mesh) using 10% ethyl acetate in hexane as eluent to give the title compound (0.803 g, 99%) . H NMR (300 MHz, CDCl 3): d 7.93 (s, 1H), 7.92 (s, 1H), 4.32 (c, 2H, "7 = 7.0Hz), 1.86 (s, 2H), 1.38 (s, 6H), 1.34 (s, 6H), 1.38-1.34 (m, 3H), 0.24 (s, 9H).
8-Ethinyl-2, 2,4, 4-tetramethylchroman-6-carboxylic acid (Intermediate 210)
A solution of ethyl 8-trimethylsilanylethynyl-2,2,4,4-tetramethylchroman-6-carboxylate (Intermediate 209, 0.525 g, 1.47 mmol) in ethanol (5 ml) was treated with a 2N aqueous solution of sodium hydroxide. sodium (5 ml, 10 mmol) and the resulting solution was adjusted to pH -5 with 10% aqueous hydrochloric acid and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo to give the title product as a brown solid.
(0.316 g, 84%). H NMR (300 MHz, CDCl 3): d 8.02 (s, 2H), 3.23 (s, 1H), 1.89 (s, 2H), 1.42 (s, 6H), 1.38 ( s, 6H).
8-Ethynyl-2,4,4,4-tetramethyl-chroman-6-carboxylic acid azide (Intermediate 211)
A stirred, cooled solution (ice bath) of 8-ethynyl-2,2,4,4-tetramethyl-chroman-6-carboxylic acid
(Intermediate 210, 0.52 g, 2 mmol) in anhydrous tetrahydrofuran (10 ml) in argon, treated with triethylamine (0.86 ml, 6 mmol) followed by ethyl chloroformate (0.25 ml,
2.6 mmol) and the resulting reaction mixture was allowed to warm to room temperature and was stirred for 2 h.
Sodium azide (0.19 g, 3 mmol) was added and the reaction mixture was stirred overnight. The reaction mixture was then diluted with water and extracted with diethyl ether.
The organic extract was dried over anhydrous magnesium sulfate, filtered and evaporated to a residue which was purified by flash column chromatography on silica gel (230-400 mesh) to give the title compound as a solid color yellow (0.32 g, 56%). H NMR (300 MHz, CDCl 3): d 7.96 (cAB, 2H, "7 = 2.1Hz), 3.24 (s, 1H), 1.89 (s, 2H), 1.42 (s, 6H), 1.37 (s, 6H).
Ethyl 4- [3- (8-ethynyl-2,2,4,4-tetramethyl-chroman-6-yl) -ureido] -2-fluoro-benzoic acid ester (Intermediate 212)
A solution of 8-ethynyl-2,2,2,4-tetramethyl-chroman-6-carboxylic acid azide (Intermediate 211, 0.104 g, 0.37 mmol) in anhydrous toluene was refluxed under argon overnight. Ethyl 4-amino-2-fluoro-benzoate (described by Teng et al., In the Journal of Medicinal Chemistry, 1996, 39, pp. 3035-3038, 0.114 g, 0.622 mmol) was added and the reaction mixture was added. refluxed for 5.5 h. The reaction mixture was cooled to room temperature and subjected to column chromatography on silica gel (230-400 mesh) using 20-33% ethyl acetate in hexane as eluent to give the title compound contaminated with some of the same. ethyl-2-fluoro-benzoate. This was used as is for the next stage.
4- [3- (8-Ethynyl-2,2,4,4-tetramethyl-chroman-6-yl) -ureido] -2-fluoro-benzoic acid (Compound 63)
A solution of 4- [3- (8-ethynyl-2, 2,4,4-tetramethyl-chroman-6-yl) -ureido] -2-fluoro-benzoic acid ethyl ester
(Intermediate 212, 0.12 g) in methanol (2 ml), tetrahydrofuran (2 ml) and water (1 ml) was treated with lithium hydroxide (0.177 g, 4.2 mmol) and the resulting reaction mixture was stirred at room temperature overnight. The volatiles were evaporated in vacuo, the residue was diluted with water and neutralized with dilute hydrochloric acid and extracted with ethyl acetate. The organic phase was dried over anhydrous magnesium sulfate, filtered and evaporated to give the title compound as a solid (0.07 g, 46% for two steps). H1 NMR (300 MHz, CD3OD): d 7.86 (dd, 1H, "7 = 8.8, 8.5Hz), 7.53 (dd, 1H," 7 = 13.7, 2.0Hz), 7.42 (d, 1H, «7 = 2.3Hz), 7.28 (d, 1H,« 7 = 2.3Hz), 7.14 (dd, 1H, «7 = 2.0, 8.8Hz ), 3.50 (s, 1H), 1.86 (s, 2H), 1.35 (s, 12H).
Alternatively, the composition can be pressurized and contain a compressed gas, such as nitrogen or a liquefied gas propellant. The liquefied propellant medium and of course the total composition is preferably such that the active inhibitor does not dissolve there to a substantial degree.
It is understood that modifications that do not substantially affect the activity of the different embodiments of this invention are also included in the definition of the invention provided herein. Accordingly, it is desired that the following examples illustrate but not limit the present invention.
EXAMPLE I Inhibition of the Differentiation of Sebaceous Glands in Hamster by Blocking the Signaling of Retinoic Acid Receptors
This example shows that inhibitors of CYP26B block the differentiation of sebaceous glands in hamsters. The differentiation of the sebaceous gland from the side organ of hamsters is a model system for the development of acne. To determine the efficacy of selective CYP26B inhibitors for treating acne, similar inhibitors were tested in this model system. To confirm that this model system can predict the efficacy of a compound in the reduction of acne, 13-cis-retinoic acid (acutan) was used as a positive control. As shown in Figure 1, 13-cis-retinoic acid was effective in reducing the differentiation of the sebaceous gland in the side organ of hamsters. For the model hamster system, Syrian golden male hamsters were fed with LabDiet # 5002 rodent diet and housed individually in a 14/10 hour light / dark cycle. For the treatment, the animals were randomized according to body weight and were also weighed in each treatment, so that the drug doses could be adjusted accordingly. The animals were treated daily by oral gavage, 6 days / week for 4 weeks. At the end of the treatments, the animals were sacrificed by inhalation of carbon dioxide. The organs of the side were then excised and extended on a small piece of record that was placed in a small histology cassette for fixation in 10% buffered formaldehyde. The specimen was divided into 5 portions equally spaced to make paraffin blocks and a H & E section of 5 micrometers was prepared for each block. The two sections that contained more sebaceous glands were used to determine the average area of the acini of the side organ. A video camera anchored to a standard microscope with the objective of 4X was used to capture the images of the sebaceous glands. The computer programs Videoshop and NIH Image 1.63 were used to quantify the areas of the acini. All the recognizable sebaceous glands of the two sections were analyzed and the means were derived for the comparison between different treatments. In all studies, blood samples were also collected to determine serum triglyceride levels using a Ginder Trinder Trinder Kit from Sigma Chemicals (St. Louis, MO).
To determine whether retinoic acid receptor (RAR) signaling is required for the 13-cis-retinoic acid-induced reduction of sebaceous gland differentiation from the hamster side organ, a RAR pan-agonist was assayed. As shown in Figure 2, compound X pan-agonist of RAR blocked the effect of the treatment of 13-cis-retinoic acid on the differentiation of the sebaceous glands.
The ability of the different RAR antagonists to reduce the differentiation of sebaceous glands from the hamster side was determined. As shown in Figure 3, 13-cis-retinoic acid (30 mg / kg) and
TTNPB (0.02 mg / kg and 0.08 mg / kg) were effective in reducing the differentiation of sebaceous glands, while Compound Y (100 mg / kg) and all trans retinoic acid
(3 mg / kg) had substantially the same activity as the control vehicle.
Table 1 shows a summary of the experimental results showing the effect of different retinoid compounds on the differentiation of the sebaceous glands in the organ of the hamster side. These results indicate that the activation of retinoic acid receptor signaling is sufficient to reduce the differentiation of the sebaceous glands. However, toxicity of RAR agonists such as TTNPB was observed at effective doses. Inactive pro-drugs such as 13-cis-retinoic acid and 4-oxo-13-cis-RA, which had lower toxicity levels, appeared to be more effective.
Since CYP26 functions in cells by reducing the signaling of retinoic acid receptors, the effect of CYP26 inhibition to increase signaling of retinoic acid receptors was examined, thereby reducing the differentiation of hamster sebaceous glands . The expression of CYP26A and CYP26B in sebaceous glands of the hamster side organ was determined using Taqman RT-PCR. The hamsters were treated as described above and sacrificed at different times in time (1 day to 3 weeks). The organs of the side were excised and placed in a dissecting microscope. The sebaceous glands were removed and placed in liquid nitrogen immediately. The RNAs were isolated from the glands using Trizol reagents (Invitrogen, San Diego, CA) as described by the manufacturer. The RNAs were treated with DNase to remove the contaminating genomic DNAs and further purified with the DNA-free kit (Ambion;
Austin, TX). Quantitative RT-PCR was performed on an ABI 7700 machine using the Platinum qRT-PCR kit from Invitrogen with primers and probes specific for CYP26.
As shown in Table 2, both CYP26A and CYP26B are expressed in sebaceous glands of the hamster side, and the expression of both is induced by 13-cis-retinoic acid. It seems that the expression of CYP26B is higher in cells in a late state of differentiation.
To determine whether selective CYP26B inhibitors reduce the differentiation of sebaceous glands from the hamster side, two CYP26B inhibitors were tested. As shown in Figure 4, Compound A (50 mg / kg) and Compound Y (12.5 mg / kg) reduced the differentiation of sebaceous glands from the hamster side at least as effectively as 13-cis acid -retinoic (30 mg / kg). Compound A is a selective inhibitor of CYP26A that has a selectivity of at least 10-fold for CYP26A relative to CYP26B. Compound Y is a selective inhibitor of CYP26B that has a selectivity of at least 10-fold for CYP26B relative to CYP26A. The chemical structures of these compounds are shown in Table 3, below. Therefore, it is expected that selective inhibitors of CYP26A or CYP26B effectively reduce or prevent acne.
Table 3
In summary, this example demonstrates that selective inhibitors of CYP26 are effective in reducing the differentiation of sebaceous glands from the side into hamsters, a model system for the development of acne.
EXAMPLE II
Cell-based method for the identification of CYP26 inhibitors
In this example, a cell-based analysis is described to identify compounds that selectively inhibit CYP26A or CYP26B activity.
HeLa cells stably transfected with CYP26A and CYP26B were kept in plates for 100 mm tissue culture in MEM medium containing 10% FBS and 100 μg / ml hygromycin. The cells that grew exponentially were harvested by incubation in trypsin. The cells were then washed with IX PBS and plated in 48-well plates at 5 x 10 5 cells in 0.2 ml of MEM medium containing 10% FBS and RA- [H3] of 0.5 μCi in the presence or absence of concentrations increasing of the test compounds. The compounds were diluted in 100% DMSO and then added to wells in triplicate at final concentrations of 10, 1 or 0.1 μM. As a positive control for the inhibition of RA metabolism, the cells were also incubated with ketoconazole at 100, 10 and 1 μM. The cells were incubated for 3 hours at 37 ° C. The retinoids were then extracted using a modified Bligh and Dyer procedure (Bligh and Dyer, supra (1959)), in which methylene chloride, as opposed to chloroform, was used, and the aqueous soluble radioactivity was quantified using a scintillation counter H.H . The values of the ID50 represent the inhibitor concentration required to inhibit the metabolism of RA by 50 percent and were derived manually from the transformed logarithmic data.
A variety of compounds identified by inhibiting CYP26A or CYP26B selectively are shown in Table 4. It was determined that the compounds listed did not have a substantial RAR inhibitory activity. To determine the selectivity of a CYP26A inhibitor, the ID50 of the inhibitor was divided with respect to CYP26A by the ID50 of the inhibitor with respect to CYP26B. To determine the selectivity of a CYP26B inhibitor, the DI50 of the inhibitor was divided with respect to CYP26B by the DI50 of the inhibitor with respect to CYP26A.
Table 4
Note: DA = Weakly Active; NA = Not Active
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the related claims.
Claims (74)
1. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to such an individual an effective amount of a selective CYP26B inhibitor, wherein said selective CYP26B inhibitor has a selectivity of at least 10 times for CYP26B in relation to CYP26A.
2. The method of claim 1, wherein said retinoid sensitive disorder is a skin disorder.
3. The method of claim 2, wherein said skin disorder is acne.
4. The method of claim 1, wherein said retinoid sensitive disorder is a disorder selected from an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
5. The method of claim 1, wherein said inhibitor is administered peripherally.
6. The method of claim 5, wherein said inhibitor is administered orally.
7. The method of claim 1, wherein said individual is a human.
8. The method of claim 1, wherein said selective CYP26B inhibitor has a selectivity of at least 20-fold for CYP26B relative to CYP26A.
9. The method of claim 8, wherein said selective CYP26B inhibitor has a selectivity of at least 100-fold for CYP26B relative to CYP26A.
.10. The method of claim 9, wherein said selective CYP26B inhibitor has a selectivity of at least 500 times for CYP26B relative to CYP26A.
11. The method of claim 10, wherein said selective CYP26B inhibitor has a selectivity of at least 1000 times for CYP26B relative to CYP26A.
12. The method of claim 1, wherein said selective CYP26B inhibitor has the formula: Formula 30 wherein, R = t-Butyl, CH2N (Me) (cyc-Pr), or N (Me) (cyc-Pr); Y X = H or Me.
13. The method of claim 1, wherein said selective CYP26B inhibitor has the formula: Formula 31 wherein, R = t-Butyl, CH2N (Me) (cyc-Pr), or N (Me) (cyc-Pr); Y X = H or Me.
14. The method of claim 1, wherein said selective CYP26B inhibitor has the formula: Formula 32 where, n = 0 or 1; Y R? = R2 = Me; X = O, S, where R = lower alkyl; and Y = cycloalkyl, alkenyl, alkynyl, lower alkoxy, halo, or haloalkyl; or R? = Me; R2 = -CH2CH2-; X = O or S; and Y = cycloalkyl, alkenyl, alkynyl, lower alkoxy, halo, or haloalkyl; or Ra = Me; R2 = H; X = -CHCH2N (Me) (cyc-Pr); and Y = H, lower alkyl, cycloalkyl, alkenyl, lower alkoxy, halo or haloalkyl; or Rx = Me; R = H; X = CMe2; and Y = H, lower alkyl, cycloalkyl, alkenyl, alkynyl, lower alkoxy, halo or haloalkyl.
15. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 18 where, R3 = H or lower alkyl; and R? = R2 = H, Me or Et; X = 0 or S; and Y = CH2N (Me) (cyc-Pr), H, halo, OH, lower alkoxy, lower alkyl, haloalkyl, cycloalkyl, alkenyl, or alkynyl; or R? = Me; R2 = H; X = H (C) CH2N (Me) (cyc-pr); and Y = H, halo, lower alkoxy, lower alkyl, cycloalkyl or haloalkyl; or R1 = Me, R2 = H; X = C = 0; and Y = H, lower alkyl, haloalkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl.
16. The method of claim 15, wherein said retinoid responsive disorder is a skin disorder.
17. The method of claim 15, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
18. The method of claim 15, wherein said administration is oral administration.
19. The method of claim 16, wherein said skin disorder is acne.
20. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 19 where, R3 = H or lower alkyl; R1 = R2 = H, Me or Et; X = 0 or S; and Y = CH2N (Me) (cyc-Pr), H, halo, OH, lower alkoxy, lower alkyl, haloalkyl, cycloalkyl, alkenyl, or alkynyl; or R3 = H or lower alkyl; and R? = Me; R2 = H; X = H (C) CH2N (Me) (cyc-Pr); and Y = H, halo, lower alkoxy, lower alkyl, cycloalkyl or haloalkyl; or R3 = H or lower alkyl; and R? = Me; R2 = H; X = C = 0; and Y = H, lower alkyl, haloalkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
21. The method of claim 20, wherein said retinoid responsive disorder is a skin disorder.
22. The method of claim 20, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
23. The method of claim 20, wherein said administration is oral administration.
24. The method of claim 21, wherein said skin disorder is acne.
25. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 20 where, R? = R2 = Me; X = O or S; and Y = CH2N (Me) (cyc-Pr), OR, COOR, where R = lower alkyl, cycloalkyl; or R? = Me; R2 = H; X = H (C) CH2N (Me) (cyc-Pr); and Y = OR, COOR where R = lower alkyl, or cycloalkyl; or R? = Me; R2 = H; X = C = 0; and Y = alkenyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
26. The method of claim 25, wherein said retinoid sensitive disorder is a skin disorder.
27. The method of claim 25, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
28. The method of claim 25, wherein said administration is oral administration.
29. The method of claim 26, wherein said skin disorder is acne.
30. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 21 where, R? = R2 = Me; X = O or S; and Y = lower alkyl, haloalkyl, cycloalkyl, CH2N (Me) (cyc-Pr), OR, or COOR, where R = lower alkyl, or cycloalkyl; or R1 = Me; R2 = H; X = H (C) CH2N (Me) (cyc-Pr); and Y = lower alkyl, haloalkyl, cycloalkyl, OR, COOR, where R = lower alkyl or cycloalkyl; or R? = Me; R2 = H; X = C = 0; and Y = alkenyl, lower alkyl, or cycloalkyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
31. The method of claim 30, wherein said retinoid sensitive disorder is a skin disorder.
32. The method of claim 30, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
33. The method of claim 30, wherein said administration is oral administration.
34. The method of claim 31, wherein said skin disorder is acne.
35. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 22 wherein, R3 = lower alkyl; and Rx = R2 = H or Me; or Rx = Me; and R2 = H, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
36. The method of claim 35, wherein said retinoid sensitive disorder is a skin disorder.
37. The method of claim 35, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
38. The method of claim 35, wherein said administration is oral administration.
39. The method of claim 36, wherein said skin disorder is acne.
40. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 23 wherein, R3 = lower alkyl; Y R1 = H and R2 = H; and Y = alkenyl, or alkynyl; or Rx = Me and R2 = H or Me; and Y = lower alkyl, alkenyl, or alkynyl; or R? = H or Me and R2 = Me; and Y = lower alkyl, alkenyl, or alkynyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
41. The method of claim 40, wherein said retinoid sensitive disorder is a skin disorder.
42. The method of claim 40, wherein said retinoid responsive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
43. The method of claim 40, wherein said administration is oral administration.
44. The method of claim 41, wherein said skin disorder is acne.
45. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 24 where, R3 = H or lower alkyl; Y Rx = H, Me or Et; X = O or S; and Y = CH2N (Me) (cyc-Pr), H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl, or alkynyl; or Rx = Me; X = H (C) CH2N (Me) (cyc-Pr); and Y = H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl or alkynyl; or R? = Me; X = C = 0; and Y = H, lower alkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
46. The method of claim 45, wherein said retinoid responsive disorder is a skin disorder.
47. The method of claim 45, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
48. The method of claim 45, wherein said administration is oral administration.
49. The method of claim 46, wherein said skin disorder is acne.
50. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 25 where, R3 = H or lower alkyl; and Rx = H, Me or Et; X = O or S; and Y = CH2N (Me) (cyc-Pr), H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl or alkynyl; or Rx = Me; X = H (C) CH2N (Me) (cyc-Pr); and Y = H, lower alkoxy, lower alkyl, cycloalkyl, alkenyl or alkynyl; or R = Me; X = C = 0; and Y = H, lower alkyl, lower alkoxy, cycloalkyl, alkenyl or alkynyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
51. The method of claim 50, wherein said retinoid sensitive disorder is a skin disorder.
52. The method of claim 50, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
53. The method of claim 50, wherein said administration is oral administration.
54. The method of claim 51, wherein said skin disorder is acne.
55. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 26 where, R? = H, Me or Et; X = 0 or S; and Y = CH2N (Me) (cyc-Pr), OR, or COOR, where R lower alkyl or cycloalkyl; or R? = Me, X = H (C) CH2N (Me) (cyc-Pr); and Y = alkenyl, OR, COOR, where R = lower alkyl or cycloalkyl; or R? = Me; X = C = 0; and Y = alkenyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
56. The method of claim 55, wherein said retinoid sensitive disorder is a skin disorder.
57. The method of claim 55, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
58. The method of claim 55, wherein said administration is oral administration.
59. The method of claim 56, wherein said skin disorder is acne.
60. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 27 where, Z = H, Me or Cl; Y Rx = H, Me or Et; X = O or S; and Y = lower alkyl, cycloalkyl, CH2N (Me) (cyc-Pr), OR, or COOR, where R = lower alkyl or cycloalkyl; or Rs = Me, X = H (C) CH2N (Me) (cyc-Pr); and Y = lower alkyl, cycloalkyl, OR, or COOR, where R = lower alkyl, or cycloalkyl; or R? = Me; X = C = 0; and Y = alkenyl, lower alkyl, or cycloalkyl, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
61. The method of claim 60, wherein said retinoid sensitive disorder is a skin disorder.
62. The method of claim 60, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
63. The method of claim 60, wherein said administration is oral administration.
64. The method of claim 61, wherein said skin disorder is acne.
65. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to such an individual an effective amount of a selective CYP26A inhibitor having the formula: Formula 28 where, R = lower alkyl; Y R? = H; and R2 = Me; OR RX = R2 = Me or H, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
66 The method of claim 65, wherein said retinoid sensitive disorder is a skin disorder.
67. The method of claim 65, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
68. The method of claim 65, wherein said administration is oral administration.
69. The method of claim 66, wherein said skin disorder is acne.
70. A method for the treatment of an individual having a retinoid responsive disorder, comprising administering to an individual such an effective amount of a selective CYP26A inhibitor having the formula: Formula 29 where, X = COOR, C (CH3) 2COOR, CH2N (CH3) (cyc-Pr), where R = lower alkyl; Y = H, lower alkyl, haloalkyl, alkenyl or alkynyl; Y RX = H and R2 = Me; or R1 = R2 = Me or H, or a pharmaceutically acceptable salt, ester, amide, stereoisomer or racemic mixture thereof.
71. The method of claim 70, wherein said retinoid responsive disorder is a skin disorder.
72. The method of claim 70, wherein said retinoid sensitive disorder is a disorder selected from the group consisting of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, a neurological disorder, an eye disorder, and a pulmonary disorder.
73. The method of claim 70, wherein said administration is oral administration.
74. The method of claim 71, wherein said skin disorder is acne.
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US60/530,601 | 2003-12-17 |
Publications (1)
Publication Number | Publication Date |
---|---|
MXPA06006820A true MXPA06006820A (en) | 2006-12-13 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US7468391B2 (en) | Methods for treating retinoid responsive disorders using selective inhibitors of CYP26A and CYP26B | |
US7638648B2 (en) | Compounds having selective cytochrome P450RAI-1 or selective cytochrome P450RAI-2 inhibitory activity and methods of obtaining the same | |
US6855512B2 (en) | Methods for identifying inhibitors of cytochrome P450RAI | |
US6291677B1 (en) | Compounds having activity as inhibitors of cytochrome P450RAI | |
DE60116642T2 (en) | COMPOUNDS WITH CYTOCHROM P450RA1 HARDENING ACTIVITY | |
US6359135B1 (en) | Compounds having activity as inhibitors of cytochrome P450RAI | |
US6303785B1 (en) | Compounds having activity as inhibitors of cytochrome P450RAI | |
US7351737B2 (en) | 4-[(8-substituted)-6-chromanoyl]-and 4-[8-substituted)-chroman-6-YL-ethynyl]-benzoic and phenylacetic acids, their esters and salts having cytochrome P450RAI inhibitory activity | |
US7019034B2 (en) | Compositions and methods for reducing serum glucose and triglyceride levels in diabetic mammals | |
US6387951B1 (en) | Compounds having activity as inhibitors of cytochrome P450RAI | |
US6369261B1 (en) | Compounds having activity as inhibitors of cytochrome P450RAI | |
US6603019B2 (en) | 1-imidazolyl substituted tetrahydronaphthalene derivatives as inhibitors of eytochrome P450RAI | |
MXPA06006820A (en) | Methods for treating retinoid responsive disorders using selective inhibitors of cyp26a and cyp26b | |
JP2018516971A (en) | Beta-naphthoisoflavone, composition containing the same and use thereof | |
US20180185325A1 (en) | 12H-BENZO[b]XANTHEN-12-ONES, COMPOSITIONS CONTAINING, AND USES OF, SAME | |
US20040077721A1 (en) | Compositions and methods using compounds having cytochrome P450RAI inhibitory activity co-administered with vitamin A |