NZ615557B2 - Pyrrolo [2,3-d] pyrimidine derivatives as inhibitors of tropomyosin- related kinases - Google Patents
Pyrrolo [2,3-d] pyrimidine derivatives as inhibitors of tropomyosin- related kinases Download PDFInfo
- Publication number
- NZ615557B2 NZ615557B2 NZ615557A NZ61555712A NZ615557B2 NZ 615557 B2 NZ615557 B2 NZ 615557B2 NZ 615557 A NZ615557 A NZ 615557A NZ 61555712 A NZ61555712 A NZ 61555712A NZ 615557 B2 NZ615557 B2 NZ 615557B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- pyrrolo
- acetamide
- lcms
- carbonyl
- pyridinyl
- Prior art date
Links
- 230000002401 inhibitory effect Effects 0.000 title abstract description 24
- 239000003112 inhibitor Substances 0.000 title abstract description 21
- 108091000081 Phosphotransferases Proteins 0.000 title abstract description 13
- 150000004943 pyrrolo[2,3-d]pyrimidines Chemical class 0.000 title abstract description 5
- 102000030951 Phosphotransferases Human genes 0.000 title abstract 3
- 150000001875 compounds Chemical class 0.000 claims abstract description 360
- 239000000203 mixture Substances 0.000 claims abstract description 148
- 208000002193 Pain Diseases 0.000 claims abstract description 119
- 239000011780 sodium chloride Substances 0.000 claims abstract description 110
- 150000003839 salts Chemical class 0.000 claims abstract description 107
- 201000010099 disease Diseases 0.000 claims abstract description 29
- 125000001424 substituent group Chemical group 0.000 claims abstract description 26
- -1 methoxy, ethyl Chemical group 0.000 claims description 357
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 claims description 216
- DLFVBJFMPXGRIB-UHFFFAOYSA-N acetamide Chemical compound CC(N)=O DLFVBJFMPXGRIB-UHFFFAOYSA-N 0.000 claims description 189
- 125000004076 pyridyl group Chemical group 0.000 claims description 117
- 125000000714 pyrimidinyl group Chemical group 0.000 claims description 114
- CZPWVGJYEJSRLH-UHFFFAOYSA-N 289-95-2 Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 claims description 111
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 43
- 125000000217 alkyl group Chemical group 0.000 claims description 41
- 125000004122 cyclic group Chemical group 0.000 claims description 40
- 101700043017 NTRK1 Proteins 0.000 claims description 33
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 32
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 claims description 26
- 229910052736 halogen Inorganic materials 0.000 claims description 26
- 150000002367 halogens Chemical class 0.000 claims description 26
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 25
- 125000003545 alkoxy group Chemical group 0.000 claims description 24
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 24
- 125000000623 heterocyclic group Chemical group 0.000 claims description 23
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 22
- 239000003814 drug Substances 0.000 claims description 21
- 239000008194 pharmaceutical composition Substances 0.000 claims description 17
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 16
- 229940079593 drugs Drugs 0.000 claims description 15
- 125000005842 heteroatoms Chemical group 0.000 claims description 12
- 229910052760 oxygen Inorganic materials 0.000 claims description 12
- 229910052717 sulfur Inorganic materials 0.000 claims description 11
- 229910052799 carbon Inorganic materials 0.000 claims description 10
- 125000001475 halogen functional group Chemical group 0.000 claims description 8
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 8
- 229910052757 nitrogen Inorganic materials 0.000 claims description 8
- 201000002674 obstructive nephropathy Diseases 0.000 claims description 8
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 claims description 8
- 125000005843 halogen group Chemical group 0.000 claims description 7
- 229910052739 hydrogen Inorganic materials 0.000 claims description 7
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 7
- 125000000475 sulfinyl group Chemical group [*:2]S([*:1])=O 0.000 claims description 7
- 125000006555 (C3-C5) cycloalkyl group Chemical group 0.000 claims description 6
- CHGYICZMKKIHQI-UHFFFAOYSA-N 2-[4-(trifluoromethyl)phenyl]acetamide Chemical compound NC(=O)CC1=CC=C(C(F)(F)F)C=C1 CHGYICZMKKIHQI-UHFFFAOYSA-N 0.000 claims description 6
- 125000003354 benzotriazolyl group Chemical group N1N=NC2=C1C=CC=C2* 0.000 claims description 6
- 125000002619 bicyclic group Chemical group 0.000 claims description 6
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 claims description 6
- 239000003937 drug carrier Substances 0.000 claims description 6
- 125000003854 p-chlorophenyl group Chemical group [H]C1=C([H])C(*)=C([H])C([H])=C1Cl 0.000 claims description 6
- 125000001399 1,2,3-triazolyl group Chemical group N1N=NC(=C1)* 0.000 claims description 5
- 125000003118 aryl group Chemical group 0.000 claims description 5
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 claims description 5
- 125000003226 pyrazolyl group Chemical group 0.000 claims description 5
- 239000002464 receptor antagonist Substances 0.000 claims description 5
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 5
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 claims description 4
- 238000004519 manufacturing process Methods 0.000 claims description 4
- 125000001064 morpholinomethyl group Chemical group [H]C([H])(*)N1C([H])([H])C([H])([H])OC([H])([H])C1([H])[H] 0.000 claims description 4
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 4
- RWRDLPDLKQPQOW-UHFFFAOYSA-N pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 claims description 4
- 125000004429 atoms Chemical group 0.000 claims description 3
- 125000003236 benzoyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C(*)=O 0.000 claims description 3
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 3
- 125000002147 dimethylamino group Chemical group [H]C([H])([H])N(*)C([H])([H])[H] 0.000 claims description 3
- 125000003566 oxetanyl group Chemical group 0.000 claims description 3
- 125000004215 2,4-difluorophenyl group Chemical group [H]C1=C([H])C(*)=C(F)C([H])=C1F 0.000 claims description 2
- HONIICLYMWZJFZ-UHFFFAOYSA-N Azetidine Chemical compound C1CNC1 HONIICLYMWZJFZ-UHFFFAOYSA-N 0.000 claims description 2
- 125000004603 benzisoxazolyl group Chemical group O1N=C(C2=C1C=CC=C2)* 0.000 claims description 2
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 claims description 2
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 claims description 2
- 235000010290 biphenyl Nutrition 0.000 claims description 2
- 239000004305 biphenyl Substances 0.000 claims description 2
- 125000000259 cinnolinyl group Chemical group N1=NC(=CC2=CC=CC=C12)* 0.000 claims description 2
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N diphenyl Chemical compound C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 claims description 2
- 125000001301 ethoxy group Chemical group [H]C([H])([H])C([H])([H])O* 0.000 claims description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 claims description 2
- 125000004005 formimidoyl group Chemical group [H]\N=C(/[H])* 0.000 claims description 2
- 125000003838 furazanyl group Chemical group 0.000 claims description 2
- 125000004857 imidazopyridinyl group Chemical group N1C(=NC2=C1C=CC=N2)* 0.000 claims description 2
- 125000003392 indanyl group Chemical group C1(CCC2=CC=CC=C12)* 0.000 claims description 2
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 claims description 2
- 125000001041 indolyl group Chemical group 0.000 claims description 2
- 125000000904 isoindolyl group Chemical group C=1(NC=C2C=CC=CC12)* 0.000 claims description 2
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 claims description 2
- 125000000842 isoxazolyl group Chemical group 0.000 claims description 2
- YNAVUWVOSKDBBP-UHFFFAOYSA-N morpholine Chemical group C1COCCN1 YNAVUWVOSKDBBP-UHFFFAOYSA-N 0.000 claims description 2
- 125000004593 naphthyridinyl group Chemical group N1=C(C=CC2=CC=CN=C12)* 0.000 claims description 2
- 125000002971 oxazolyl group Chemical group 0.000 claims description 2
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 claims description 2
- GLUUGHFHXGJENI-UHFFFAOYSA-N piperazine Chemical compound C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 claims description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 claims description 2
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 claims description 2
- 125000002098 pyridazinyl group Chemical group 0.000 claims description 2
- 125000000168 pyrrolyl group Chemical group 0.000 claims description 2
- 125000002294 quinazolinyl group Chemical group N1=C(N=CC2=CC=CC=C12)* 0.000 claims description 2
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 claims description 2
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 claims description 2
- 125000000335 thiazolyl group Chemical group 0.000 claims description 2
- 125000001425 triazolyl group Chemical group 0.000 claims description 2
- 125000006274 (C1-C3)alkoxy group Chemical group 0.000 claims 18
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 claims 3
- 102100004109 HEY1 Human genes 0.000 claims 2
- 108010081348 HRT1 protein Hairy Proteins 0.000 claims 2
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 claims 2
- 125000004216 fluoromethyl group Chemical group [H]C([H])(F)* 0.000 claims 2
- 235000019256 formaldehyde Nutrition 0.000 claims 2
- 125000002883 imidazolyl group Chemical group 0.000 claims 2
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 claims 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims 1
- 125000004309 pyranyl group Chemical group O1C(C=CC=C1)* 0.000 claims 1
- 125000001567 quinoxalinyl group Chemical group N1=C(C=NC2=CC=CC=C12)* 0.000 claims 1
- 125000001889 triflyl group Chemical group FC(F)(F)S(*)(=O)=O 0.000 claims 1
- 238000002360 preparation method Methods 0.000 abstract description 237
- 201000011510 cancer Diseases 0.000 abstract description 28
- 238000000034 method Methods 0.000 abstract description 15
- 230000000694 effects Effects 0.000 abstract description 11
- 102000005937 Tropomyosin Human genes 0.000 abstract description 4
- 108010030743 Tropomyosin Proteins 0.000 abstract description 4
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 598
- 238000002953 preparative HPLC Methods 0.000 description 181
- 239000002253 acid Substances 0.000 description 140
- XEKOWRVHYACXOJ-UHFFFAOYSA-N acetic acid ethyl ester Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 121
- QTBSBXVTEAMEQO-UHFFFAOYSA-N acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 100
- WYURNTSHIVDZCO-UHFFFAOYSA-N tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 86
- JUJWROOIHBZHMG-UHFFFAOYSA-N pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 77
- 239000007787 solid Substances 0.000 description 76
- 150000003230 pyrimidines Chemical class 0.000 description 63
- 239000000243 solution Substances 0.000 description 58
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 55
- 235000019439 ethyl acetate Nutrition 0.000 description 53
- 238000005160 1H NMR spectroscopy Methods 0.000 description 51
- OKKJLVBELUTLKV-UHFFFAOYSA-N methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 49
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 44
- YMWUJEATGCHHMB-UHFFFAOYSA-N methylene dichloride Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 43
- 239000000543 intermediate Substances 0.000 description 35
- WEVYAHXRMPXWCK-UHFFFAOYSA-N acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 34
- BDAGIHXWWSANSR-UHFFFAOYSA-N formic acid Chemical compound OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 31
- UIIMBOGNXHQVGW-UHFFFAOYSA-M sodium bicarbonate Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 30
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 26
- PMZURENOXWZQFD-UHFFFAOYSA-L sodium sulphate Substances [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 26
- PVNIIMVLHYAWGP-UHFFFAOYSA-N nicotinic acid Chemical compound OC(=O)C1=CC=CN=C1 PVNIIMVLHYAWGP-UHFFFAOYSA-N 0.000 description 25
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 24
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 24
- KFZMGEQAYNKOFK-UHFFFAOYSA-N iso-propanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 24
- 239000011541 reaction mixture Substances 0.000 description 24
- 239000012071 phase Substances 0.000 description 23
- JNWBBCNCSMBKNE-UHFFFAOYSA-N HATU Chemical compound F[P-](F)(F)(F)(F)F.C1=CN=C2N(OC(N(C)C)=[N+](C)C)N=NC2=C1 JNWBBCNCSMBKNE-UHFFFAOYSA-N 0.000 description 22
- 101710030983 RNF138 Proteins 0.000 description 22
- 101710029702 TICAM1 Proteins 0.000 description 22
- 101710021425 TRIM69 Proteins 0.000 description 22
- 102100003447 TRIM69 Human genes 0.000 description 22
- CSNNHWWHGAXBCP-UHFFFAOYSA-L magnesium sulphate Substances [Mg+2].[O-][S+2]([O-])([O-])[O-] CSNNHWWHGAXBCP-UHFFFAOYSA-L 0.000 description 22
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 22
- 239000002904 solvent Substances 0.000 description 22
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-dimethylformamide Substances CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 21
- 125000001820 oxy group Chemical group [*:1]O[*:2] 0.000 description 21
- 239000012267 brine Substances 0.000 description 19
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N DMSO-d6 Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 18
- 206010022114 Injury Diseases 0.000 description 18
- 239000000741 silica gel Substances 0.000 description 18
- 229910002027 silica gel Inorganic materials 0.000 description 18
- ZMANZCXQSJIPKH-UHFFFAOYSA-N triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 18
- VEXZGXHMUGYJMC-UHFFFAOYSA-N HCl Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 17
- 150000007513 acids Chemical class 0.000 description 17
- 238000006243 chemical reaction Methods 0.000 description 17
- RTZKZFJDLAIYFH-UHFFFAOYSA-N diethyl ether Substances CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 17
- 125000006239 protecting group Chemical group 0.000 description 17
- 229910052938 sodium sulfate Inorganic materials 0.000 description 17
- 235000011152 sodium sulphate Nutrition 0.000 description 17
- DTQVDTLACAAQTR-UHFFFAOYSA-N trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 16
- JJTNLWSCFYERCK-UHFFFAOYSA-N 7H-pyrrolo[2,3-d]pyrimidine Chemical compound N1=CN=C2NC=CC2=C1 JJTNLWSCFYERCK-UHFFFAOYSA-N 0.000 description 15
- 206010065390 Inflammatory pain Diseases 0.000 description 15
- 150000001412 amines Chemical class 0.000 description 15
- CKJNUZNMWOVDFN-UHFFFAOYSA-N methanone Chemical compound O=[CH-] CKJNUZNMWOVDFN-UHFFFAOYSA-N 0.000 description 15
- 239000003921 oil Substances 0.000 description 15
- 235000019198 oils Nutrition 0.000 description 15
- HEMHJVSKTPXQMS-UHFFFAOYSA-M sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 15
- 229940053128 Nerve Growth Factor Drugs 0.000 description 14
- 102000015336 Nerve Growth Factor Human genes 0.000 description 14
- 108010025020 Nerve Growth Factor Proteins 0.000 description 14
- 239000000463 material Substances 0.000 description 14
- 239000012074 organic phase Substances 0.000 description 14
- MBUYFUMAXPNQHG-UHFFFAOYSA-N 7-propan-2-ylpyrrolo[2,3-d]pyrimidine Chemical compound N1=CN=C2N(C(C)C)C=CC2=C1 MBUYFUMAXPNQHG-UHFFFAOYSA-N 0.000 description 13
- 208000004296 Neuralgia Diseases 0.000 description 13
- 239000007788 liquid Substances 0.000 description 13
- 239000012044 organic layer Substances 0.000 description 13
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 13
- 238000004809 thin layer chromatography Methods 0.000 description 13
- WVCHIGAIXREVNS-UHFFFAOYSA-N 2-HYDROXY-1,4-NAPHTHOQUINONE Chemical compound C1=CC=C2C(O)=CC(=O)C(=O)C2=C1 WVCHIGAIXREVNS-UHFFFAOYSA-N 0.000 description 12
- XDTQCCMRBSYSAO-UHFFFAOYSA-N 7-methylpyrrolo[2,3-d]pyrimidine Chemical compound N1=CN=C2N(C)C=CC2=C1 XDTQCCMRBSYSAO-UHFFFAOYSA-N 0.000 description 12
- 102000001253 Protein Kinases Human genes 0.000 description 12
- 239000004480 active ingredient Substances 0.000 description 12
- 239000000460 chlorine Substances 0.000 description 12
- 238000004128 high performance liquid chromatography Methods 0.000 description 12
- 150000002500 ions Chemical class 0.000 description 12
- 239000003960 organic solvent Substances 0.000 description 12
- 239000002245 particle Substances 0.000 description 12
- 239000000843 powder Substances 0.000 description 12
- DNIAPMSPPWPWGF-UHFFFAOYSA-N propylene glycol Chemical group CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 12
- 235000017557 sodium bicarbonate Nutrition 0.000 description 12
- 239000003826 tablet Substances 0.000 description 12
- 125000001981 tert-butyldimethylsilyl group Chemical group [H]C([H])([H])[Si]([H])(C([H])([H])[H])[*]C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 12
- 210000001519 tissues Anatomy 0.000 description 12
- 230000003042 antagnostic Effects 0.000 description 11
- 239000005557 antagonist Substances 0.000 description 11
- 239000003153 chemical reaction reagent Substances 0.000 description 11
- 239000003795 chemical substances by application Substances 0.000 description 11
- 238000004440 column chromatography Methods 0.000 description 11
- 239000002552 dosage form Substances 0.000 description 11
- 235000019253 formic acid Nutrition 0.000 description 11
- 230000004054 inflammatory process Effects 0.000 description 11
- 238000000746 purification Methods 0.000 description 11
- 230000002829 reduced Effects 0.000 description 11
- 238000003756 stirring Methods 0.000 description 11
- 239000003643 water by type Substances 0.000 description 11
- 208000000094 Chronic Pain Diseases 0.000 description 10
- 208000004454 Hyperalgesia Diseases 0.000 description 10
- 208000007999 Hyperesthesia Diseases 0.000 description 10
- 206010061218 Inflammation Diseases 0.000 description 10
- 230000001684 chronic Effects 0.000 description 10
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 10
- 235000019341 magnesium sulphate Nutrition 0.000 description 10
- 239000000725 suspension Substances 0.000 description 10
- 230000001225 therapeutic Effects 0.000 description 10
- AXPWHHFJEONRKU-UHFFFAOYSA-N 7-tert-butylpyrrolo[2,3-d]pyrimidine Chemical compound N1=CN=C2N(C(C)(C)C)C=CC2=C1 AXPWHHFJEONRKU-UHFFFAOYSA-N 0.000 description 9
- 101700028444 USO1 Proteins 0.000 description 9
- 101700061199 WNT1 Proteins 0.000 description 9
- 238000007792 addition Methods 0.000 description 9
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 9
- 238000011067 equilibration Methods 0.000 description 9
- KDLHZDBZIXYQEI-UHFFFAOYSA-N palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 9
- CMWTZPSULFXXJA-VIFPVBQESA-N Naproxen Chemical compound C1=C([C@H](C)C(O)=O)C=CC2=CC(OC)=CC=C21 CMWTZPSULFXXJA-VIFPVBQESA-N 0.000 description 8
- 230000001154 acute Effects 0.000 description 8
- 150000001408 amides Chemical class 0.000 description 8
- QGZKDVFQNNGYKY-UHFFFAOYSA-N ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 8
- KXDAEFPNCMNJSK-UHFFFAOYSA-N benzamide Chemical compound NC(=O)C1=CC=CC=C1 KXDAEFPNCMNJSK-UHFFFAOYSA-N 0.000 description 8
- OKTJSMMVPCPJKN-UHFFFAOYSA-N carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 8
- 229910052801 chlorine Inorganic materials 0.000 description 8
- ZAMOUSCENKQFHK-UHFFFAOYSA-N chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 8
- 238000009472 formulation Methods 0.000 description 8
- VLKZOEOYAKHREP-UHFFFAOYSA-N hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 8
- MZRVEZGGRBJDDB-UHFFFAOYSA-N n-butyllithium Chemical compound [Li]CCCC MZRVEZGGRBJDDB-UHFFFAOYSA-N 0.000 description 8
- 238000007911 parenteral administration Methods 0.000 description 8
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 8
- 238000003786 synthesis reaction Methods 0.000 description 8
- LMDZBCPBFSXMTL-UHFFFAOYSA-N 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide Chemical compound CCN=C=NCCCN(C)C LMDZBCPBFSXMTL-UHFFFAOYSA-N 0.000 description 7
- 208000005298 Acute Pain Diseases 0.000 description 7
- 206010053552 Allodynia Diseases 0.000 description 7
- 206010020751 Hypersensitivity Diseases 0.000 description 7
- 102000007072 Nerve Growth Factors Human genes 0.000 description 7
- 108010008267 Nerve Growth Factors Proteins 0.000 description 7
- RYGMFSIKBFXOCR-UHFFFAOYSA-N copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 7
- 229960004132 diethyl ether Drugs 0.000 description 7
- 238000000105 evaporative light scattering detection Methods 0.000 description 7
- KWYUFKZDYYNOTN-UHFFFAOYSA-M potassium hydroxide Inorganic materials [OH-].[K+] KWYUFKZDYYNOTN-UHFFFAOYSA-M 0.000 description 7
- 102000005962 receptors Human genes 0.000 description 7
- 108020003175 receptors Proteins 0.000 description 7
- 125000000446 sulfanediyl group Chemical group *S* 0.000 description 7
- 229940086542 triethylamine Drugs 0.000 description 7
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 6
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonium chloride Substances [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 6
- FJDQFPXHSGXQBY-UHFFFAOYSA-L Caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 6
- 206010058019 Cancer pain Diseases 0.000 description 6
- 101710036039 NOTCH4 Proteins 0.000 description 6
- 208000001294 Nociceptive Pain Diseases 0.000 description 6
- DHXVGJBLRPWPCS-UHFFFAOYSA-N THP Chemical compound C1CCOCC1 DHXVGJBLRPWPCS-UHFFFAOYSA-N 0.000 description 6
- LWIHDJKSTIGBAC-UHFFFAOYSA-K Tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 6
- 208000009935 Visceral Pain Diseases 0.000 description 6
- 230000002354 daily Effects 0.000 description 6
- 239000003085 diluting agent Substances 0.000 description 6
- 239000000706 filtrate Substances 0.000 description 6
- 230000009610 hypersensitivity Effects 0.000 description 6
- 230000001965 increased Effects 0.000 description 6
- 230000004968 inflammatory condition Effects 0.000 description 6
- 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 6
- 239000010410 layer Substances 0.000 description 6
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 6
- OFBQJSOFQDEBGM-UHFFFAOYSA-N pentane Chemical compound CCCCC OFBQJSOFQDEBGM-UHFFFAOYSA-N 0.000 description 6
- CYPYTURSJDMMMP-WVCUSYJESA-N (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one;palladium Chemical compound [Pd].[Pd].C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1.C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1.C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1 CYPYTURSJDMMMP-WVCUSYJESA-N 0.000 description 5
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 5
- YEDUAINPPJYDJZ-UHFFFAOYSA-N 2-hydroxybenzothiazole Chemical compound C1=CC=C2SC(O)=NC2=C1 YEDUAINPPJYDJZ-UHFFFAOYSA-N 0.000 description 5
- NRGSIZCSAPERDJ-UHFFFAOYSA-N 2-propan-2-yl-7H-pyrrolo[2,3-d]pyrimidine Chemical compound CC(C)C1=NC=C2C=CNC2=N1 NRGSIZCSAPERDJ-UHFFFAOYSA-N 0.000 description 5
- 208000008035 Back Pain Diseases 0.000 description 5
- 210000000988 Bone and Bones Anatomy 0.000 description 5
- 229920000858 Cyclodextrin Polymers 0.000 description 5
- 206010021972 Inflammatory bowel disease Diseases 0.000 description 5
- 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 5
- 108060005033 NTRK2 Proteins 0.000 description 5
- 210000000929 Nociceptors Anatomy 0.000 description 5
- 229940083542 Sodium Drugs 0.000 description 5
- 229940091252 Sodium supplements Drugs 0.000 description 5
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Natural products NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 5
- 239000000654 additive Substances 0.000 description 5
- 230000000996 additive Effects 0.000 description 5
- 150000001299 aldehydes Chemical class 0.000 description 5
- 238000005804 alkylation reaction Methods 0.000 description 5
- 201000005794 allergic hypersensitivity disease Diseases 0.000 description 5
- VHUUQVKOLVNVRT-UHFFFAOYSA-N ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 5
- 235000011114 ammonium hydroxide Nutrition 0.000 description 5
- 239000008346 aqueous phase Substances 0.000 description 5
- 239000004202 carbamide Substances 0.000 description 5
- 239000000969 carrier Substances 0.000 description 5
- 229910052802 copper Inorganic materials 0.000 description 5
- 239000010949 copper Substances 0.000 description 5
- 239000007884 disintegrant Substances 0.000 description 5
- 239000000284 extract Substances 0.000 description 5
- 229920000591 gum Polymers 0.000 description 5
- NPZTUJOABDZTLV-UHFFFAOYSA-N hydroxybenzotriazole Substances O=C1C=CC=C2NNN=C12 NPZTUJOABDZTLV-UHFFFAOYSA-N 0.000 description 5
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 description 5
- 239000008101 lactose Substances 0.000 description 5
- 230000000051 modifying Effects 0.000 description 5
- 108091008022 nociceptors Proteins 0.000 description 5
- 210000000056 organs Anatomy 0.000 description 5
- QVGXLLKOCUKJST-UHFFFAOYSA-N oxygen atom Chemical group [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 5
- 229920000642 polymer Polymers 0.000 description 5
- 229910000027 potassium carbonate Inorganic materials 0.000 description 5
- 230000004044 response Effects 0.000 description 5
- 230000035945 sensitivity Effects 0.000 description 5
- 239000011734 sodium Substances 0.000 description 5
- 229910052708 sodium Inorganic materials 0.000 description 5
- 235000015424 sodium Nutrition 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 230000002459 sustained Effects 0.000 description 5
- ILMRJRBKQSSXGY-UHFFFAOYSA-N tert-butyl(dimethyl)silicon Chemical compound C[Si](C)C(C)(C)C ILMRJRBKQSSXGY-UHFFFAOYSA-N 0.000 description 5
- CUYKNJBYIJFRCU-UHFFFAOYSA-N 3-Aminopyridine Chemical compound NC1=CC=CN=C1 CUYKNJBYIJFRCU-UHFFFAOYSA-N 0.000 description 4
- CDPKJZJVTHSESZ-UHFFFAOYSA-N 4-chlorophenylacetic acid Chemical compound OC(=O)CC1=CC=C(Cl)C=C1 CDPKJZJVTHSESZ-UHFFFAOYSA-N 0.000 description 4
- 206010005949 Bone cancer Diseases 0.000 description 4
- 206010064012 Central pain syndrome Diseases 0.000 description 4
- 208000002551 Irritable Bowel Syndrome Diseases 0.000 description 4
- 206010024324 Leukaemias Diseases 0.000 description 4
- 239000002202 Polyethylene glycol Substances 0.000 description 4
- 206010039073 Rheumatoid arthritis Diseases 0.000 description 4
- 229920002472 Starch Chemical class 0.000 description 4
- 229940032147 Starch Drugs 0.000 description 4
- FPGGTKZVZWFYPV-UHFFFAOYSA-M Tetra-n-butylammonium fluoride Chemical compound [F-].CCCC[N+](CCCC)(CCCC)CCCC FPGGTKZVZWFYPV-UHFFFAOYSA-M 0.000 description 4
- 229940035295 Ting Drugs 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- XKRFYHLGVUSROY-UHFFFAOYSA-N argon Chemical compound [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 description 4
- 239000011230 binding agent Substances 0.000 description 4
- 238000006065 biodegradation reaction Methods 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 239000003054 catalyst Substances 0.000 description 4
- 210000004027 cells Anatomy 0.000 description 4
- 238000007796 conventional method Methods 0.000 description 4
- 150000002148 esters Chemical class 0.000 description 4
- 230000002496 gastric Effects 0.000 description 4
- PEDCQBHIVMGVHV-UHFFFAOYSA-N glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 4
- 150000004820 halides Chemical class 0.000 description 4
- 239000008079 hexane Substances 0.000 description 4
- 230000002757 inflammatory Effects 0.000 description 4
- 239000004615 ingredient Substances 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 239000000314 lubricant Substances 0.000 description 4
- IUYHWZFSGMZEOG-UHFFFAOYSA-M magnesium;propane;chloride Chemical compound [Mg+2].[Cl-].C[CH-]C IUYHWZFSGMZEOG-UHFFFAOYSA-M 0.000 description 4
- 125000001570 methylene group Chemical group [H]C([H])([*:1])[*:2] 0.000 description 4
- DFPAKSUCGFBDDF-UHFFFAOYSA-N nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 4
- IJGRMHOSHXDMSA-UHFFFAOYSA-N nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 230000001473 noxious Effects 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- XMGMFRIEKMMMSU-UHFFFAOYSA-N phenylmethylbenzene Chemical group C=1C=CC=CC=1[C]C1=CC=CC=C1 XMGMFRIEKMMMSU-UHFFFAOYSA-N 0.000 description 4
- 229920001223 polyethylene glycol Polymers 0.000 description 4
- 239000001184 potassium carbonate Substances 0.000 description 4
- 235000015320 potassium carbonate Nutrition 0.000 description 4
- UBQKCCHYAOITMY-UHFFFAOYSA-N pyridin-2-ol Chemical compound OC1=CC=CC=N1 UBQKCCHYAOITMY-UHFFFAOYSA-N 0.000 description 4
- 238000010992 reflux Methods 0.000 description 4
- 230000001624 sedative Effects 0.000 description 4
- 230000011664 signaling Effects 0.000 description 4
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 4
- BZKBCQXYZZXSCO-UHFFFAOYSA-N sodium hydride Inorganic materials [H-].[Na+] BZKBCQXYZZXSCO-UHFFFAOYSA-N 0.000 description 4
- AGGHKNBCHLWKHY-UHFFFAOYSA-N sodium;triacetyloxyboron(1-) Chemical compound [Na+].CC(=O)O[B-](OC(C)=O)OC(C)=O AGGHKNBCHLWKHY-UHFFFAOYSA-N 0.000 description 4
- 230000002269 spontaneous Effects 0.000 description 4
- 239000007921 spray Substances 0.000 description 4
- 235000019698 starch Nutrition 0.000 description 4
- 239000008107 starch Chemical class 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 230000004083 survival Effects 0.000 description 4
- 239000000375 suspending agent Substances 0.000 description 4
- 201000010874 syndrome Diseases 0.000 description 4
- 230000002194 synthesizing Effects 0.000 description 4
- 235000019798 tripotassium phosphate Nutrition 0.000 description 4
- WEBXRQONNWEETE-UHFFFAOYSA-N 2-(4-cyanophenyl)acetic acid Chemical compound OC(=O)CC1=CC=C(C#N)C=C1 WEBXRQONNWEETE-UHFFFAOYSA-N 0.000 description 3
- ATKULCGQSLCGEK-UHFFFAOYSA-N 2-(5-bromopyridin-2-yl)acetic acid Chemical compound OC(=O)CC1=CC=C(Br)C=N1 ATKULCGQSLCGEK-UHFFFAOYSA-N 0.000 description 3
- CVYNISFVRLITAQ-UHFFFAOYSA-N 5,6-didehydro-4H-pyrimidin-3-ium-4-ide Chemical group [C-]1=[N+]=C=NC#C1 CVYNISFVRLITAQ-UHFFFAOYSA-N 0.000 description 3
- 210000001185 Bone Marrow Anatomy 0.000 description 3
- 229940047583 Cetamide Drugs 0.000 description 3
- UDHXJZHVNHGCEC-UHFFFAOYSA-N Chlorophacinone Chemical compound C1=CC(Cl)=CC=C1C(C=1C=CC=CC=1)C(=O)C1C(=O)C2=CC=CC=C2C1=O UDHXJZHVNHGCEC-UHFFFAOYSA-N 0.000 description 3
- NKLCNNUWBJBICK-UHFFFAOYSA-N Dess–Martin periodinane Chemical compound C1=CC=C2I(OC(=O)C)(OC(C)=O)(OC(C)=O)OC(=O)C2=C1 NKLCNNUWBJBICK-UHFFFAOYSA-N 0.000 description 3
- 206010012680 Diabetic neuropathy Diseases 0.000 description 3
- 208000005171 Dysmenorrhea Diseases 0.000 description 3
- 108010010803 Gelatin Proteins 0.000 description 3
- 206010019233 Headache Diseases 0.000 description 3
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 3
- 206010025323 Lymphomas Diseases 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 206010027476 Metastasis Diseases 0.000 description 3
- INQOMBQAUSQDDS-UHFFFAOYSA-N Methyl iodide Chemical compound IC INQOMBQAUSQDDS-UHFFFAOYSA-N 0.000 description 3
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 3
- 210000003205 Muscles Anatomy 0.000 description 3
- 108060005034 NTRK3 Proteins 0.000 description 3
- 206010029260 Neuroblastoma Diseases 0.000 description 3
- 206010025310 Other lymphomas Diseases 0.000 description 3
- 208000008443 Pancreatic Carcinoma Diseases 0.000 description 3
- 208000004550 Postoperative Pain Diseases 0.000 description 3
- SCVFZCLFOSHCOH-UHFFFAOYSA-M Potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 3
- NLKNQRATVPKPDG-UHFFFAOYSA-M Potassium iodide Chemical compound [K+].[I-] NLKNQRATVPKPDG-UHFFFAOYSA-M 0.000 description 3
- 208000000399 Procedural Pain Diseases 0.000 description 3
- QLNJFJADRCOGBJ-UHFFFAOYSA-N Propanamide Chemical compound CCC(N)=O QLNJFJADRCOGBJ-UHFFFAOYSA-N 0.000 description 3
- 206010039491 Sarcoma Diseases 0.000 description 3
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 3
- 210000001835 Viscera Anatomy 0.000 description 3
- DGEZNRSVGBDHLK-UHFFFAOYSA-N [1,10]phenanthroline Chemical compound C1=CN=C2C3=NC=CC=C3C=CC2=C1 DGEZNRSVGBDHLK-UHFFFAOYSA-N 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 238000010640 amide synthesis reaction Methods 0.000 description 3
- 235000019270 ammonium chloride Nutrition 0.000 description 3
- 230000027455 binding Effects 0.000 description 3
- 239000002775 capsule Substances 0.000 description 3
- 238000002512 chemotherapy Methods 0.000 description 3
- VEXZGXHMUGYJMC-UHFFFAOYSA-M chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 3
- 125000001309 chloro group Chemical group Cl* 0.000 description 3
- 238000004587 chromatography analysis Methods 0.000 description 3
- 239000006071 cream Substances 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- XTHFKEDIFFGKHM-UHFFFAOYSA-N dimethoxyethane Chemical compound COCCOC XTHFKEDIFFGKHM-UHFFFAOYSA-N 0.000 description 3
- ROSDSFDQCJNGOL-UHFFFAOYSA-N dimethylamine Chemical compound CNC ROSDSFDQCJNGOL-UHFFFAOYSA-N 0.000 description 3
- SXZIXHOMFPUIRK-UHFFFAOYSA-N diphenylmethanimine Chemical compound C=1C=CC=CC=1C(=N)C1=CC=CC=C1 SXZIXHOMFPUIRK-UHFFFAOYSA-N 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 239000010408 film Substances 0.000 description 3
- 239000000796 flavoring agent Substances 0.000 description 3
- SPWZDQUIUXNYBL-UHFFFAOYSA-N furo[3,2-c]pyridine-2-carboxylic acid Chemical compound N1=CC=C2OC(C(=O)O)=CC2=C1 SPWZDQUIUXNYBL-UHFFFAOYSA-N 0.000 description 3
- 239000000499 gel Substances 0.000 description 3
- 229920000159 gelatin Polymers 0.000 description 3
- 239000008273 gelatin Substances 0.000 description 3
- 235000019322 gelatine Nutrition 0.000 description 3
- 235000011852 gelatine desserts Nutrition 0.000 description 3
- 230000035876 healing Effects 0.000 description 3
- KLGZELKXQMTEMM-UHFFFAOYSA-N hydride Chemical compound [H-] KLGZELKXQMTEMM-UHFFFAOYSA-N 0.000 description 3
- 239000001257 hydrogen Substances 0.000 description 3
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 3
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 3
- 239000007943 implant Substances 0.000 description 3
- 239000011630 iodine Substances 0.000 description 3
- 229910052740 iodine Inorganic materials 0.000 description 3
- PNDPGZBMCMUPRI-UHFFFAOYSA-N iodine Chemical compound II PNDPGZBMCMUPRI-UHFFFAOYSA-N 0.000 description 3
- 230000000670 limiting Effects 0.000 description 3
- WMFOQBRAJBCJND-UHFFFAOYSA-M lithium hydroxide Chemical compound [Li+].[OH-] WMFOQBRAJBCJND-UHFFFAOYSA-M 0.000 description 3
- 235000019359 magnesium stearate Nutrition 0.000 description 3
- 238000004949 mass spectrometry Methods 0.000 description 3
- 238000001819 mass spectrum Methods 0.000 description 3
- 238000006263 metalation reaction Methods 0.000 description 3
- 229920000609 methyl cellulose Polymers 0.000 description 3
- 235000010981 methylcellulose Nutrition 0.000 description 3
- 239000001923 methylcellulose Substances 0.000 description 3
- 125000004170 methylsulfonyl group Chemical group [H]C([H])([H])S(*)(=O)=O 0.000 description 3
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 3
- 239000008108 microcrystalline cellulose Substances 0.000 description 3
- 229940016286 microcrystalline cellulose Drugs 0.000 description 3
- 150000004682 monohydrates Chemical class 0.000 description 3
- IMNFDUFMRHMDMM-UHFFFAOYSA-N n-heptane Chemical compound CCCCCCC IMNFDUFMRHMDMM-UHFFFAOYSA-N 0.000 description 3
- 210000000653 nervous system Anatomy 0.000 description 3
- 230000001537 neural Effects 0.000 description 3
- 239000011570 nicotinamide Substances 0.000 description 3
- 230000003040 nociceptive Effects 0.000 description 3
- 239000000041 non-steroidal anti-inflammatory agent Substances 0.000 description 3
- 230000003000 nontoxic Effects 0.000 description 3
- 231100000252 nontoxic Toxicity 0.000 description 3
- 239000002674 ointment Substances 0.000 description 3
- 201000008482 osteoarthritis Diseases 0.000 description 3
- 230000003647 oxidation Effects 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- NFHFRUOZVGFOOS-UHFFFAOYSA-N palladium;triphenylphosphane Chemical compound [Pd].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.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 NFHFRUOZVGFOOS-UHFFFAOYSA-N 0.000 description 3
- 201000002528 pancreatic cancer Diseases 0.000 description 3
- 230000002093 peripheral Effects 0.000 description 3
- 125000000951 phenoxy group Chemical group [H]C1=C([H])C([H])=C(O*)C([H])=C1[H] 0.000 description 3
- AHWALFGBDFAJAI-UHFFFAOYSA-N phenyl carbonochloridate Chemical compound ClC(=O)OC1=CC=CC=C1 AHWALFGBDFAJAI-UHFFFAOYSA-N 0.000 description 3
- OZAIFHULBGXAKX-UHFFFAOYSA-N precursor Substances N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 3
- 230000002335 preservative Effects 0.000 description 3
- 239000003755 preservative agent Substances 0.000 description 3
- 235000019260 propionic acid Nutrition 0.000 description 3
- 230000001681 protective Effects 0.000 description 3
- ZFCHNZDUMIOWFV-UHFFFAOYSA-N pyrimidine-2-carboxylic acid Chemical compound OC(=O)C1=NC=CC=N1 ZFCHNZDUMIOWFV-UHFFFAOYSA-N 0.000 description 3
- 238000009938 salting Methods 0.000 description 3
- 239000000932 sedative agent Substances 0.000 description 3
- CDBYLPFSWZWCQE-UHFFFAOYSA-L sodium carbonate Substances [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 3
- 229910000029 sodium carbonate Inorganic materials 0.000 description 3
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 3
- 239000011550 stock solution Substances 0.000 description 3
- QAOWNCQODCNURD-UHFFFAOYSA-L sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 3
- 239000004094 surface-active agent Substances 0.000 description 3
- 230000000699 topical Effects 0.000 description 3
- 125000003258 trimethylene group Chemical group [H]C([H])([*:2])C([H])([H])C([H])([H])[*:1] 0.000 description 3
- 229910000404 tripotassium phosphate Inorganic materials 0.000 description 3
- WQZGKKKJIJFFOK-VFUOTHLCSA-N β-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 3
- NOOLISFMXDJSKH-KXUCPTDWSA-N (-)-(1R,3R,4S)-menthol Chemical compound CC(C)[C@@H]1CC[C@@H](C)C[C@H]1O NOOLISFMXDJSKH-KXUCPTDWSA-N 0.000 description 2
- SFLSHLFXELFNJZ-QMMMGPOBSA-N (-)-norepinephrine Chemical compound NC[C@H](O)C1=CC=C(O)C(O)=C1 SFLSHLFXELFNJZ-QMMMGPOBSA-N 0.000 description 2
- QOWBXWFYRXSBAS-UHFFFAOYSA-N (2,4-dimethoxyphenyl)methanamine Chemical compound COC1=CC=C(CN)C(OC)=C1 QOWBXWFYRXSBAS-UHFFFAOYSA-N 0.000 description 2
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2R,3R,4S,5R,6S)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2S,3R,4S,5R,6R)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2R,3R,4S,5R,6R)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 2
- KZPYGQFFRCFCPP-UHFFFAOYSA-N 1,1'-Bis(diphenylphosphino)ferrocene Chemical compound [Fe+2].C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1 KZPYGQFFRCFCPP-UHFFFAOYSA-N 0.000 description 2
- QWENRTYMTSOGBR-UHFFFAOYSA-N 1,2,3-triazole Chemical compound C=1C=NNN=1 QWENRTYMTSOGBR-UHFFFAOYSA-N 0.000 description 2
- ASOKPJOREAFHNY-UHFFFAOYSA-N 1-hydroxybenzotriazole Chemical group C1=CC=C2N(O)N=NC2=C1 ASOKPJOREAFHNY-UHFFFAOYSA-N 0.000 description 2
- GNZVAIZIJBXHHD-UHFFFAOYSA-N 2-(5-fluoropyridin-2-yl)acetic acid Chemical compound OC(=O)CC1=CC=C(F)C=N1 GNZVAIZIJBXHHD-UHFFFAOYSA-N 0.000 description 2
- QBAYIBZITZBSFO-UHFFFAOYSA-N 2-(trifluoromethyl)benzamide Chemical compound NC(=O)C1=CC=CC=C1C(F)(F)F QBAYIBZITZBSFO-UHFFFAOYSA-N 0.000 description 2
- VUKAUDKDFVSVFT-UHFFFAOYSA-N 2-[6-[4,5-bis(2-hydroxypropoxy)-2-(2-hydroxypropoxymethyl)-6-methoxyoxan-3-yl]oxy-4,5-dimethoxy-2-(methoxymethyl)oxan-3-yl]oxy-6-(hydroxymethyl)-5-methoxyoxane-3,4-diol Chemical compound COC1C(OC)C(OC2C(C(O)C(OC)C(CO)O2)O)C(COC)OC1OC1C(COCC(C)O)OC(OC)C(OCC(C)O)C1OCC(C)O VUKAUDKDFVSVFT-UHFFFAOYSA-N 0.000 description 2
- NHWQMJMIYICNBP-UHFFFAOYSA-N 2-chlorobenzonitrile Chemical compound ClC1=CC=CC=C1C#N NHWQMJMIYICNBP-UHFFFAOYSA-N 0.000 description 2
- WEHFQXPHQWWTGF-UHFFFAOYSA-N 2-quinolin-2-ylacetamide Chemical compound C1=CC=CC2=NC(CC(=O)N)=CC=C21 WEHFQXPHQWWTGF-UHFFFAOYSA-N 0.000 description 2
- YYROPELSRYBVMQ-UHFFFAOYSA-N 4-Toluenesulfonyl chloride Chemical compound CC1=CC=C(S(Cl)(=O)=O)C=C1 YYROPELSRYBVMQ-UHFFFAOYSA-N 0.000 description 2
- LAPGMTOHOQPDGI-UHFFFAOYSA-N 4-amino-2,5-difluorobenzonitrile Chemical group NC1=CC(F)=C(C#N)C=C1F LAPGMTOHOQPDGI-UHFFFAOYSA-N 0.000 description 2
- LOSIUHLAYNGOTD-UHFFFAOYSA-N 4-cyclopropyl-2H-triazole Chemical compound C1CC1C1=CNN=N1 LOSIUHLAYNGOTD-UHFFFAOYSA-N 0.000 description 2
- 125000004863 4-trifluoromethoxyphenyl group Chemical group [H]C1=C([H])C(OC(F)(F)F)=C([H])C([H])=C1* 0.000 description 2
- LSYGTEPJDXJRIO-UHFFFAOYSA-N 5-(benzhydrylideneamino)-N-methoxy-N-methylpyridine-3-carboxamide Chemical compound CON(C)C(=O)C1=CN=CC(N=C(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1 LSYGTEPJDXJRIO-UHFFFAOYSA-N 0.000 description 2
- YWTGVFDGOFXIPM-UHFFFAOYSA-N 5-bromo-N-methoxy-N-methylpyridine-3-carboxamide Chemical compound CON(C)C(=O)C1=CN=CC(Br)=C1 YWTGVFDGOFXIPM-UHFFFAOYSA-N 0.000 description 2
- TXWDVWSJMDFNQY-UHFFFAOYSA-N 5-cyclopropyl-1H-pyrazole Chemical compound C1CC1C1=CC=NN1 TXWDVWSJMDFNQY-UHFFFAOYSA-N 0.000 description 2
- CWDWFSXUQODZGW-UHFFFAOYSA-N 5-thiazolyl Chemical group [C]1=CN=CS1 CWDWFSXUQODZGW-UHFFFAOYSA-N 0.000 description 2
- ILFOUBGVORMQIQ-JTDNENJMSA-N 7-[(2R)-1-(oxan-2-yloxy)propan-2-yl]pyrrolo[2,3-d]pyrimidine Chemical compound C([C@@H](C)N1C2=NC=NC=C2C=C1)OC1CCCCO1 ILFOUBGVORMQIQ-JTDNENJMSA-N 0.000 description 2
- ILFOUBGVORMQIQ-AMGKYWFPSA-N 7-[(2S)-1-(oxan-2-yloxy)propan-2-yl]pyrrolo[2,3-d]pyrimidine Chemical compound C([C@H](C)N1C2=NC=NC=C2C=C1)OC1CCCCO1 ILFOUBGVORMQIQ-AMGKYWFPSA-N 0.000 description 2
- PVQZROXLZRBGKP-UHFFFAOYSA-N 8-methoxyimidazo[1,2-a]pyrazine Chemical compound COC1=NC=CN2C=CN=C12 PVQZROXLZRBGKP-UHFFFAOYSA-N 0.000 description 2
- 208000006820 Arthralgia 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
- 210000004369 Blood Anatomy 0.000 description 2
- 102000004219 Brain-Derived Neurotrophic Factor Human genes 0.000 description 2
- 108090000715 Brain-Derived Neurotrophic Factor Proteins 0.000 description 2
- SNPPWIUOZRMYNY-UHFFFAOYSA-N Bupropion Chemical compound CC(C)(C)NC(C)C(=O)C1=CC=CC(Cl)=C1 SNPPWIUOZRMYNY-UHFFFAOYSA-N 0.000 description 2
- 206010006811 Bursitis Diseases 0.000 description 2
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate dianion Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 2
- AHOUBRCZNHFOSL-YOEHRIQHSA-N Casbol Chemical compound C1=CC(F)=CC=C1[C@H]1[C@H](COC=2C=C3OCOC3=CC=2)CNCC1 AHOUBRCZNHFOSL-YOEHRIQHSA-N 0.000 description 2
- WSEQXVZVJXJVFP-UHFFFAOYSA-N Citalopram Chemical compound O1CC2=CC(C#N)=CC=C2C1(CCCN(C)C)C1=CC=C(F)C=C1 WSEQXVZVJXJVFP-UHFFFAOYSA-N 0.000 description 2
- GDLIGKIOYRNHDA-UHFFFAOYSA-N Clomipramine Chemical compound C1CC2=CC=C(Cl)C=C2N(CCCN(C)C)C2=CC=CC=C21 GDLIGKIOYRNHDA-UHFFFAOYSA-N 0.000 description 2
- OROGSEYTTFOCAN-DNJOTXNNSA-N Codeine Chemical compound C([C@H]1[C@H](N(CC[C@@]112)C)C3)=C[C@H](O)[C@@H]1OC1=C2C3=CC=C1OC OROGSEYTTFOCAN-DNJOTXNNSA-N 0.000 description 2
- 102000008186 Collagen Human genes 0.000 description 2
- 108010035532 Collagen Proteins 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- 210000002808 Connective Tissue Anatomy 0.000 description 2
- 206010011401 Crohn's disease Diseases 0.000 description 2
- 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 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N D-sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- MKXZASYAUGDDCJ-SZMVWBNQSA-N Dextromethorphan Chemical compound C1CCC[C@H]2[C@@]3([H])N(C)CC[C@]21C1=CC(OC)=CC=C1C3 MKXZASYAUGDDCJ-SZMVWBNQSA-N 0.000 description 2
- 229960001985 Dextromethorphan Drugs 0.000 description 2
- JAQUASYNZVUNQP-PVAVHDDUSA-N Dextrorphan Chemical compound C1C2=CC=C(O)C=C2[C@@]23CCN(C)[C@@H]1[C@H]2CCCC3 JAQUASYNZVUNQP-PVAVHDDUSA-N 0.000 description 2
- DYHSDKLCOJIUFX-UHFFFAOYSA-N Di-tert-butyl dicarbonate Chemical compound CC(C)(C)OC(=O)OC(=O)OC(C)(C)C DYHSDKLCOJIUFX-UHFFFAOYSA-N 0.000 description 2
- 208000001636 Diabetic Neuropathy Diseases 0.000 description 2
- HPNMFZURTQLUMO-UHFFFAOYSA-N Diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 2
- 206010013935 Dysmenorrhoea Diseases 0.000 description 2
- 101700063373 EIF3E Proteins 0.000 description 2
- CBQGYUDMJHNJBX-OALUTQOASA-N Esreboxetine Chemical compound CCOC1=CC=CC=C1O[C@@H](C=1C=CC=CC=1)[C@H]1OCCNC1 CBQGYUDMJHNJBX-OALUTQOASA-N 0.000 description 2
- 208000001640 Fibromyalgia Diseases 0.000 description 2
- 229960002464 Fluoxetine Drugs 0.000 description 2
- BJHIKXHVCXFQLS-UYFOZJQFSA-N Fructose Natural products OC[C@@H](O)[C@@H](O)[C@H](O)C(=O)CO BJHIKXHVCXFQLS-UYFOZJQFSA-N 0.000 description 2
- 206010017758 Gastric cancer Diseases 0.000 description 2
- 210000001035 Gastrointestinal Tract Anatomy 0.000 description 2
- 206010020243 Hodgkin's disease Diseases 0.000 description 2
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 2
- 101710004181 INTS2 Proteins 0.000 description 2
- BCGWQEUPMDMJNV-UHFFFAOYSA-N Imipramine Chemical compound C1CC2=CC=CC=C2N(CCCN(C)C)C2=CC=CC=C21 BCGWQEUPMDMJNV-UHFFFAOYSA-N 0.000 description 2
- CGIGDMFJXJATDK-UHFFFAOYSA-N Indometacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 description 2
- 208000005615 Interstitial Cystitis Diseases 0.000 description 2
- RPCVIAXDAUMJJP-PZBABLGHSA-N Ispronicline Chemical compound CN[C@@H](C)C\C=C\C1=CN=CC(OC(C)C)=C1 RPCVIAXDAUMJJP-PZBABLGHSA-N 0.000 description 2
- 239000004201 L-cysteine Substances 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- 229960004873 LEVOMENTHOL Drugs 0.000 description 2
- 208000007046 Leukemia, Myeloid, Acute Diseases 0.000 description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 2
- 239000004472 Lysine Substances 0.000 description 2
- 229940091250 Magnesium supplements Drugs 0.000 description 2
- GUBGYTABKSRVRQ-YOLKTULGSA-N Maltose 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-YOLKTULGSA-N 0.000 description 2
- 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 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
- 230000036650 Metabolic stability Effects 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L MgCl2 Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 229920000881 Modified starch Chemical class 0.000 description 2
- BQJCRHHNABKAKU-KBQPJGBKSA-N Morphine Chemical compound O([C@H]1[C@H](C=C[C@H]23)O)C4=C5[C@@]12CCN(C)[C@@H]3CC5=CC=C4O BQJCRHHNABKAKU-KBQPJGBKSA-N 0.000 description 2
- 210000002027 Muscle, Skeletal Anatomy 0.000 description 2
- GWFRSQBGIGLMFD-BBKOCAQVSA-N N-[9-[(2R,3R,4S,5R)-5-[[bis(4-methoxyphenyl)-phenylmethoxy]methyl]-3,4-dihydroxyoxolan-2-yl]purin-6-yl]-2-phenoxyacetamide Chemical compound C1=CC(OC)=CC=C1C(C=1C=CC(OC)=CC=1)(C=1C=CC=CC=1)OC[C@@H]1[C@@H](O)[C@@H](O)[C@H](N2C3=NC=NC(NC(=O)COC=4C=CC=CC=4)=C3N=C2)O1 GWFRSQBGIGLMFD-BBKOCAQVSA-N 0.000 description 2
- LQZMLBORDGWNPD-UHFFFAOYSA-N N-iodosuccinimide Chemical compound IN1C(=O)CCC1=O LQZMLBORDGWNPD-UHFFFAOYSA-N 0.000 description 2
- RTHCYVBBDHJXIQ-UHFFFAOYSA-N N-methyl-3-phenyl-3-[4-(trifluoromethyl)phenoxy]propan-1-amine Chemical compound C=1C=CC=CC=1C(CCNC)OC1=CC=C(C(F)(F)F)C=C1 RTHCYVBBDHJXIQ-UHFFFAOYSA-N 0.000 description 2
- KCTZOTUQSGYWLV-UHFFFAOYSA-N N1C=NC=C2N=CC=C21 Chemical compound N1C=NC=C2N=CC=C21 KCTZOTUQSGYWLV-UHFFFAOYSA-N 0.000 description 2
- 101700009327 NTF3 Proteins 0.000 description 2
- 102100015697 NTF3 Human genes 0.000 description 2
- 206010052639 Nerve injury Diseases 0.000 description 2
- 102000011779 Nitric Oxide Synthase Type II Human genes 0.000 description 2
- 108010076864 Nitric Oxide Synthase Type II Proteins 0.000 description 2
- 229960002748 Norepinephrine Drugs 0.000 description 2
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N Oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 2
- 235000019502 Orange oil Nutrition 0.000 description 2
- XAPRFLSJBSXESP-UHFFFAOYSA-N Oxycinchophen Chemical class N=1C2=CC=CC=C2C(C(=O)O)=C(O)C=1C1=CC=CC=C1 XAPRFLSJBSXESP-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
- 208000000450 Pelvic Pain Diseases 0.000 description 2
- 241000243142 Porifera Species 0.000 description 2
- 206010036376 Post herpetic neuralgia Diseases 0.000 description 2
- 229960003975 Potassium Drugs 0.000 description 2
- 206010060862 Prostate cancer Diseases 0.000 description 2
- 102000001708 Protein Isoforms Human genes 0.000 description 2
- 108010029485 Protein Isoforms Proteins 0.000 description 2
- 108060006633 Protein Kinases Proteins 0.000 description 2
- LOUPRKONTZGTKE-WZBLMQSHSA-N Quinine Chemical compound C([C@H]([C@H](C1)C=C)C2)C[N@@]1[C@@H]2[C@H](O)C1=CC=NC2=CC=C(OC)C=C21 LOUPRKONTZGTKE-WZBLMQSHSA-N 0.000 description 2
- 241000277284 Salvelinus fontinalis Species 0.000 description 2
- 206010070834 Sensitisation Diseases 0.000 description 2
- VGKDLMBJGBXTGI-SJCJKPOMSA-N Sertraline Chemical compound C1([C@@H]2CC[C@@H](C3=CC=CC=C32)NC)=CC=C(Cl)C(Cl)=C1 VGKDLMBJGBXTGI-SJCJKPOMSA-N 0.000 description 2
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 description 2
- 210000000278 Spinal Cord Anatomy 0.000 description 2
- 208000008513 Spinal Cord Injury Diseases 0.000 description 2
- CZMRCDWAGMRECN-GDQSFJPYSA-N Sucrose Natural products O([C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1)[C@@]1(CO)[C@H](O)[C@@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-GDQSFJPYSA-N 0.000 description 2
- 208000002847 Surgical Wound Diseases 0.000 description 2
- JACRWUWPXAESPB-QMMMGPOBSA-N Tropic acid Natural products OC[C@H](C(O)=O)C1=CC=CC=C1 JACRWUWPXAESPB-QMMMGPOBSA-N 0.000 description 2
- 206010047095 Vascular pain Diseases 0.000 description 2
- 101700058266 WNT3 Proteins 0.000 description 2
- HEBKCHPVOIAQTA-SCDXWVJYSA-N Xylitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)CO HEBKCHPVOIAQTA-SCDXWVJYSA-N 0.000 description 2
- 229960002675 Xylitol Drugs 0.000 description 2
- GTLDTDOJJJZVBW-UHFFFAOYSA-N Zinc cyanide Chemical compound [Zn+2].N#[C-].N#[C-] GTLDTDOJJJZVBW-UHFFFAOYSA-N 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-K [O-]P([O-])([O-])=O Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-M acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 2
- 230000002378 acidificating Effects 0.000 description 2
- 239000000443 aerosol Substances 0.000 description 2
- 150000001298 alcohols Chemical class 0.000 description 2
- 125000004202 aminomethyl group Chemical group [H]N([H])C([H])([H])* 0.000 description 2
- 239000000908 ammonium hydroxide Substances 0.000 description 2
- 230000000202 analgesic Effects 0.000 description 2
- 150000008064 anhydrides Chemical class 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 229910052786 argon Inorganic materials 0.000 description 2
- 230000003190 augmentative Effects 0.000 description 2
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- CPELXLSAUQHCOX-UHFFFAOYSA-M bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 2
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 2
- 150000001649 bromium compounds Chemical class 0.000 description 2
- 125000001246 bromo group Chemical group Br* 0.000 description 2
- 229910000024 caesium carbonate Inorganic materials 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- 125000004432 carbon atoms Chemical group C* 0.000 description 2
- 239000001768 carboxy methyl cellulose Substances 0.000 description 2
- 150000001768 cations Chemical class 0.000 description 2
- 229920001436 collagen Polymers 0.000 description 2
- 229960005188 collagen Drugs 0.000 description 2
- 230000000875 corresponding Effects 0.000 description 2
- 230000001808 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 239000012043 crude product Substances 0.000 description 2
- 125000004802 cyanophenyl group Chemical group 0.000 description 2
- 125000000753 cycloalkyl group Chemical group 0.000 description 2
- 238000010511 deprotection reaction Methods 0.000 description 2
- 230000001809 detectable Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- 230000001079 digestive Effects 0.000 description 2
- 238000001035 drying Methods 0.000 description 2
- 230000004064 dysfunction Effects 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 230000002708 enhancing Effects 0.000 description 2
- FCZCIXQGZOUIDN-UHFFFAOYSA-N ethyl 2-diethoxyphosphinothioyloxyacetate Chemical compound CCOC(=O)COP(=S)(OCC)OCC FCZCIXQGZOUIDN-UHFFFAOYSA-N 0.000 description 2
- 238000003818 flash chromatography Methods 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- KRHYYFGTRYWZRS-UHFFFAOYSA-M fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 description 2
- 235000013355 food flavoring agent Nutrition 0.000 description 2
- 150000004675 formic acid derivatives Chemical class 0.000 description 2
- 238000001640 fractional crystallisation Methods 0.000 description 2
- 238000004108 freeze drying Methods 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 230000014509 gene expression Effects 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- UFHFLCQGNIYNRP-UHFFFAOYSA-N hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 2
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 2
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 2
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 2
- 229960004801 imipramine Drugs 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 200000000018 inflammatory disease Diseases 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 230000003834 intracellular Effects 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- XMBWDFGMSWQBCA-UHFFFAOYSA-M iodide Chemical compound [I-] XMBWDFGMSWQBCA-UHFFFAOYSA-M 0.000 description 2
- 238000010902 jet-milling Methods 0.000 description 2
- 125000000468 ketone group Chemical group 0.000 description 2
- 230000003902 lesions Effects 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- WHXSMMKQMYFTQS-UHFFFAOYSA-N lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 2
- 229910052744 lithium Inorganic materials 0.000 description 2
- 201000005202 lung cancer Diseases 0.000 description 2
- 229960003646 lysine Drugs 0.000 description 2
- FYYHWMGAXLPEAU-UHFFFAOYSA-N magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 2
- 239000011777 magnesium Substances 0.000 description 2
- 229910052749 magnesium Inorganic materials 0.000 description 2
- 239000000594 mannitol Substances 0.000 description 2
- 235000010355 mannitol Nutrition 0.000 description 2
- 239000002207 metabolite Substances 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- ACEONLNNWKIPTM-UHFFFAOYSA-N methyl 2-bromopropanoate Chemical compound COC(=O)C(C)Br ACEONLNNWKIPTM-UHFFFAOYSA-N 0.000 description 2
- 239000003595 mist Substances 0.000 description 2
- QDHHCQZDFGDHMP-UHFFFAOYSA-N monochloramine Chemical compound ClN QDHHCQZDFGDHMP-UHFFFAOYSA-N 0.000 description 2
- 239000000178 monomer Substances 0.000 description 2
- 229960005181 morphine Drugs 0.000 description 2
- 229930014694 morphine Natural products 0.000 description 2
- SJRJJKPEHAURKC-UHFFFAOYSA-N n-methylmorpholine Chemical compound CN1CCOCC1 SJRJJKPEHAURKC-UHFFFAOYSA-N 0.000 description 2
- 235000005152 nicotinamide Nutrition 0.000 description 2
- 229960003966 nicotinamide Drugs 0.000 description 2
- 229940021182 non-steroidal anti-inflammatory drugs Drugs 0.000 description 2
- 239000010502 orange oil Substances 0.000 description 2
- 125000002524 organometallic group Chemical group 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- MYMOFIZGZYHOMD-UHFFFAOYSA-N oxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 2
- 229910052763 palladium Inorganic materials 0.000 description 2
- 230000002085 persistent Effects 0.000 description 2
- 235000019271 petrolatum Nutrition 0.000 description 2
- 239000000546 pharmaceutic aid Substances 0.000 description 2
- 230000000275 pharmacokinetic Effects 0.000 description 2
- 239000010452 phosphate Substances 0.000 description 2
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 239000011698 potassium fluoride Substances 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000002633 protecting Effects 0.000 description 2
- 235000018102 proteins Nutrition 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 125000003373 pyrazinyl group Chemical group 0.000 description 2
- DZPAOAZDQHZRGG-UHFFFAOYSA-N pyrazolo[1,5-a]pyrimidine-2-carboxylic acid Chemical compound N1=CC=CN2N=C(C(=O)O)C=C21 DZPAOAZDQHZRGG-UHFFFAOYSA-N 0.000 description 2
- RUUOPSRRIKJHNH-UHFFFAOYSA-N pyridazine-3-carboxylic acid Chemical compound OC(=O)C1=CC=CN=N1 RUUOPSRRIKJHNH-UHFFFAOYSA-N 0.000 description 2
- 230000002285 radioactive Effects 0.000 description 2
- 239000003638 reducing agent Substances 0.000 description 2
- 238000006722 reduction reaction Methods 0.000 description 2
- 230000035807 sensation Effects 0.000 description 2
- 235000019615 sensations Nutrition 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 239000000377 silicon dioxide Substances 0.000 description 2
- 239000001187 sodium carbonate Substances 0.000 description 2
- 235000017550 sodium carbonate Nutrition 0.000 description 2
- 239000012453 solvate Substances 0.000 description 2
- 230000000392 somatic Effects 0.000 description 2
- 238000001694 spray drying Methods 0.000 description 2
- 239000007858 starting material Substances 0.000 description 2
- QIQXTHQIDYTFRH-UHFFFAOYSA-M stearate Chemical compound CCCCCCCCCCCCCCCCCC([O-])=O QIQXTHQIDYTFRH-UHFFFAOYSA-M 0.000 description 2
- 239000008223 sterile water Substances 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 201000011549 stomach cancer Diseases 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 201000004595 synovitis Diseases 0.000 description 2
- 239000000454 talc Substances 0.000 description 2
- 229910052623 talc Inorganic materials 0.000 description 2
- 235000012222 talc Nutrition 0.000 description 2
- 108010038329 tanezumab Proteins 0.000 description 2
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 2
- 200000000020 tissue injury Diseases 0.000 description 2
- YXFVVABEGXRONW-UHFFFAOYSA-N toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 2
- JOXIMZWYDAKGHI-UHFFFAOYSA-M toluene-4-sulfonate Chemical compound CC1=CC=C(S([O-])(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-M 0.000 description 2
- 238000004704 ultra performance liquid chromatography Methods 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- PNVNVHUZROJLTJ-UHFFFAOYSA-N venlafaxine Chemical compound C1=CC(OC)=CC=C1C(CN(C)C)C1(O)CCCCC1 PNVNVHUZROJLTJ-UHFFFAOYSA-N 0.000 description 2
- 235000012431 wafers Nutrition 0.000 description 2
- 238000010626 work up procedure Methods 0.000 description 2
- 239000000811 xylitol Substances 0.000 description 2
- 235000010447 xylitol Nutrition 0.000 description 2
- PTJADDMMFYXMMG-LJQANCHMSA-N (1R)-1-(4-fluorophenyl)-1-[3-(methylamino)propyl]-3H-2-benzofuran-5-carbonitrile Chemical compound C1([C@@]2(C3=CC=C(C=C3CO2)C#N)CCCNC)=CC=C(F)C=C1 PTJADDMMFYXMMG-LJQANCHMSA-N 0.000 description 1
- BKMMTJMQCTUHRP-GSVOUGTGSA-N (2R)-2-aminopropan-1-ol Chemical compound C[C@@H](N)CO BKMMTJMQCTUHRP-GSVOUGTGSA-N 0.000 description 1
- HIQJNYPOWPXYIC-QGZVFWFLSA-N (2R)-3-(4-benzoylphenyl)-2-[(2-methylpropan-2-yl)oxycarbonylamino]propanoic acid Chemical compound C1=CC(C[C@@H](NC(=O)OC(C)(C)C)C(O)=O)=CC=C1C(=O)C1=CC=CC=C1 HIQJNYPOWPXYIC-QGZVFWFLSA-N 0.000 description 1
- PUSNGFYSTWMJSK-GSZQVNRLSA-N (2R,3R,4S,5R,6R)-2,3,4-trimethoxy-6-(methoxymethyl)-5-[(2S,3R,4S,5R,6R)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxyoxane;1-[[(2R,3R,4S,5R,6S)-3,4,5-tris(2-hydroxypropoxy)-6-[(2R,3R,4S,5R,6R)-4,5,6-tris(2-hydroxypropoxy)-2-(2-hydroxypropoxymethyl)oxan- Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](OC)O[C@@H]1COC.CC(O)CO[C@@H]1[C@@H](OCC(C)O)[C@H](OCC(C)O)[C@@H](COCC(O)C)O[C@H]1O[C@H]1[C@H](OCC(C)O)[C@@H](OCC(C)O)[C@H](OCC(C)O)O[C@@H]1COCC(C)O PUSNGFYSTWMJSK-GSZQVNRLSA-N 0.000 description 1
- CCIWVEMVBWEMCY-RCFOMQFPSA-N (2S)-1-[(3aS,4S,7aS)-4-hydroxy-4-(2-methoxyphenyl)-7,7-diphenyl-1,3,3a,5,6,7a-hexahydroisoindol-2-yl]-2-(2-methoxyphenyl)propan-1-one Chemical compound COC1=CC=CC=C1[C@H](C)C(=O)N1C[C@H](C(CC[C@@]2(O)C=3C(=CC=CC=3)OC)(C=3C=CC=CC=3)C=3C=CC=CC=3)[C@H]2C1 CCIWVEMVBWEMCY-RCFOMQFPSA-N 0.000 description 1
- KSYYTARALLSQRH-QMMMGPOBSA-N (2S)-1-[tert-butyl(dimethyl)silyl]oxypropan-2-amine Chemical compound C[C@H](N)CO[Si](C)(C)C(C)(C)C KSYYTARALLSQRH-QMMMGPOBSA-N 0.000 description 1
- DYJIKHYBKVODAC-ZDUSSCGKSA-N (2S)-2-[(benzylamino)methyl]-2,3,7,9-tetrahydro-[1,4]dioxino[2,3-e]indol-8-one Chemical compound C([C@H]1COC=2C=CC3=C(C=2O1)CC(N3)=O)NCC1=CC=CC=C1 DYJIKHYBKVODAC-ZDUSSCGKSA-N 0.000 description 1
- MOLOJNHYNHBPCW-ZETCQYMHSA-N (2S)-2-amino-4-[2-(1-aminoethylideneamino)ethylsulfanyl]butanoic acid Chemical compound CC(=N)NCCSCC[C@H](N)C(O)=O MOLOJNHYNHBPCW-ZETCQYMHSA-N 0.000 description 1
- OQANPHBRHBJGNZ-FYJGNVAPSA-N (3E)-6-oxo-3-[[4-(pyridin-2-ylsulfamoyl)phenyl]hydrazinylidene]cyclohexa-1,4-diene-1-carboxylic acid Chemical compound C1=CC(=O)C(C(=O)O)=C\C1=N\NC1=CC=C(S(=O)(=O)NC=2N=CC=CC=2)C=C1 OQANPHBRHBJGNZ-FYJGNVAPSA-N 0.000 description 1
- GWLVNOAPYJMHQV-BWZBUEFSSA-N (3R,4R,5R)-3-amino-4,5-dimethylheptanoic acid Chemical compound CC[C@@H](C)[C@@H](C)[C@H](N)CC(O)=O GWLVNOAPYJMHQV-BWZBUEFSSA-N 0.000 description 1
- HLISOYNJVMWYQM-IWSPIJDZSA-N (3R,4R,5R)-3-amino-4,5-dimethyloctanoic acid Chemical compound CCC[C@@H](C)[C@@H](C)[C@H](N)CC(O)=O HLISOYNJVMWYQM-IWSPIJDZSA-N 0.000 description 1
- AYXYPKUFHZROOJ-ZETCQYMHSA-N (3S)-3-(aminomethyl)-5-methylhexanoic acid Chemical compound CC(C)C[C@H](CN)CC(O)=O AYXYPKUFHZROOJ-ZETCQYMHSA-N 0.000 description 1
- UHIWBQIWXWWDKT-MRVPVSSYSA-N (4S)-4-hydroxy-2-(3-methoxypropyl)-1,1-dioxo-3,4-dihydrothieno[3,2-e]thiazine-6-sulfonamide Chemical compound O=S1(=O)N(CCCOC)C[C@@H](O)C2=C1SC(S(N)(=O)=O)=C2 UHIWBQIWXWWDKT-MRVPVSSYSA-N 0.000 description 1
- XEYBRNLFEZDVAW-ARSRFYASSA-N (5Z)-7-[(1R,2R,3R)-3-hydroxy-2-[(1E,3S)-3-hydroxyoct-1-en-1-yl]-5-oxocyclopentyl]hept-5-enoic acid Chemical compound CCCCC[C@H](O)\C=C\[C@H]1[C@H](O)CC(=O)[C@@H]1C\C=C/CCCC(O)=O XEYBRNLFEZDVAW-ARSRFYASSA-N 0.000 description 1
- LDXQLWNPGRANTO-GOSISDBHSA-N (9R)-7-[[3,5-bis(trifluoromethyl)phenyl]methyl]-9-methyl-5-(4-methylphenyl)-8,9,10,11-tetrahydro-[1,4]diazocino[2,1-g][1,7]naphthyridine-6,13-dione Chemical compound C([C@H](CN(CC=1C=C(C=C(C=1)C(F)(F)F)C(F)(F)F)C1=O)C)CN(C(C2=NC=CC=C22)=O)C1=C2C1=CC=C(C)C=C1 LDXQLWNPGRANTO-GOSISDBHSA-N 0.000 description 1
- HXTGXYRHXAGCFP-OAQYLSRUSA-N (R)-(2,3-dimethoxyphenyl)-[1-[2-(4-fluorophenyl)ethyl]piperidin-4-yl]methanol Chemical compound COC1=CC=CC([C@H](O)C2CCN(CCC=3C=CC(F)=CC=3)CC2)=C1OC HXTGXYRHXAGCFP-OAQYLSRUSA-N 0.000 description 1
- JVTAAEKCZFNVCJ-UWTATZPHSA-M (R)-lactate Chemical compound C[C@@H](O)C([O-])=O JVTAAEKCZFNVCJ-UWTATZPHSA-M 0.000 description 1
- BKMMTJMQCTUHRP-VKHMYHEASA-N (S)-2-aminopropan-1-ol Chemical compound C[C@H](N)CO BKMMTJMQCTUHRP-VKHMYHEASA-N 0.000 description 1
- YFMFNYKEUDLDTL-UHFFFAOYSA-N 1,1,1,2,3,3,3-Heptafluoropropane Chemical compound FC(F)(F)C(F)C(F)(F)F YFMFNYKEUDLDTL-UHFFFAOYSA-N 0.000 description 1
- LVGUZGTVOIAKKC-UHFFFAOYSA-N 1,1,1,2-Tetrafluoroethane Chemical compound FCC(F)(F)F LVGUZGTVOIAKKC-UHFFFAOYSA-N 0.000 description 1
- FYADHXFMURLYQI-UHFFFAOYSA-N 1,2,4-triazine Chemical compound C1=CN=NC=N1 FYADHXFMURLYQI-UHFFFAOYSA-N 0.000 description 1
- AZQWKYJCGOJGHM-UHFFFAOYSA-N 1,4-Benzoquinone Chemical compound O=C1C=CC(=O)C=C1 AZQWKYJCGOJGHM-UHFFFAOYSA-N 0.000 description 1
- 229940005561 1,4-benzoquinone Drugs 0.000 description 1
- VMLKTERJLVWEJJ-UHFFFAOYSA-N 1,5-naphthyridine Chemical compound C1=CC=NC2=CC=CN=C21 VMLKTERJLVWEJJ-UHFFFAOYSA-N 0.000 description 1
- SNLMOXFUCILIPL-UHFFFAOYSA-N 1,8-naphthyridine-2-carboxylic acid Chemical compound C1=CC=NC2=NC(C(=O)O)=CC=C21 SNLMOXFUCILIPL-UHFFFAOYSA-N 0.000 description 1
- RQEUFEKYXDPUSK-UHFFFAOYSA-N 1-Phenylethylamine Chemical compound CC(N)C1=CC=CC=C1 RQEUFEKYXDPUSK-UHFFFAOYSA-N 0.000 description 1
- QEMSVZNTSXPFJA-HNAYVOBHSA-N 1-[(1S,2S)-1-hydroxy-1-(4-hydroxyphenyl)propan-2-yl]-4-phenylpiperidin-4-ol Chemical compound C1([C@H](O)[C@H](C)N2CCC(O)(CC2)C=2C=CC=CC=2)=CC=C(O)C=C1 QEMSVZNTSXPFJA-HNAYVOBHSA-N 0.000 description 1
- ZHCJQVABVSLUJB-UHFFFAOYSA-N 1-[tert-butyl(dimethyl)silyl]oxybutan-2-amine Chemical compound CCC(N)CO[Si](C)(C)C(C)(C)C ZHCJQVABVSLUJB-UHFFFAOYSA-N 0.000 description 1
- KSYYTARALLSQRH-UHFFFAOYSA-N 1-[tert-butyl(dimethyl)silyl]oxypropan-2-amine Chemical compound CC(N)CO[Si](C)(C)C(C)(C)C KSYYTARALLSQRH-UHFFFAOYSA-N 0.000 description 1
- CQMJEZQEVXQEJB-UHFFFAOYSA-N 1-hydroxy-1,3-dioxobenziodoxole Chemical compound C1=CC=C2I(O)(=O)OC(=O)C2=C1 CQMJEZQEVXQEJB-UHFFFAOYSA-N 0.000 description 1
- IWWCCNVRNHTGLV-UHFFFAOYSA-N 1-phenylcyclopropane-1-carboxylic acid Chemical compound C=1C=CC=CC=1C1(C(=O)O)CC1 IWWCCNVRNHTGLV-UHFFFAOYSA-N 0.000 description 1
- LLSKXGRDUPMXLC-UHFFFAOYSA-N 1-phenylpiperidine Chemical compound C1CCCCN1C1=CC=CC=C1 LLSKXGRDUPMXLC-UHFFFAOYSA-N 0.000 description 1
- SVUOLADPCWQTTE-UHFFFAOYSA-N 1H-1,2-benzodiazepine Chemical compound N1N=CC=CC2=CC=CC=C12 SVUOLADPCWQTTE-UHFFFAOYSA-N 0.000 description 1
- BIQRPLMAKJWHIH-UHFFFAOYSA-N 1H-imidazo[4,5-b]pyridine-2-carboxylic acid Chemical compound C1=CN=C2NC(C(=O)O)=NC2=C1 BIQRPLMAKJWHIH-UHFFFAOYSA-N 0.000 description 1
- GVLRTOYGRNLSDW-UHFFFAOYSA-N 1H-pyrazolo[3,4-b]pyridine Chemical compound C1=CC=C2C=NNC2=N1 GVLRTOYGRNLSDW-UHFFFAOYSA-N 0.000 description 1
- AMFYRKOUWBAGHV-UHFFFAOYSA-N 1H-pyrazolo[4,3-b]pyridine Chemical compound C1=CN=C2C=NNC2=C1 AMFYRKOUWBAGHV-UHFFFAOYSA-N 0.000 description 1
- MVXVYAKCVDQRLW-UHFFFAOYSA-N 1H-pyrrolo[2,3-b]pyridine Chemical class C1=CN=C2NC=CC2=C1 MVXVYAKCVDQRLW-UHFFFAOYSA-N 0.000 description 1
- QMNWYGTWTXOQTP-UHFFFAOYSA-N 1H-triazin-6-one Chemical compound O=C1C=CN=NN1 QMNWYGTWTXOQTP-UHFFFAOYSA-N 0.000 description 1
- NRKYWOKHZRQRJR-UHFFFAOYSA-N 2,2,2-trifluoroacetamide Chemical compound NC(=O)C(F)(F)F NRKYWOKHZRQRJR-UHFFFAOYSA-N 0.000 description 1
- JBQMFBWTKWOSQX-UHFFFAOYSA-N 2,3-dihydro-1H-indene-1-carboxylic acid Chemical compound C1=CC=C2C(C(=O)O)CCC2=C1 JBQMFBWTKWOSQX-UHFFFAOYSA-N 0.000 description 1
- QWQZJEXJTYAPGE-UHFFFAOYSA-N 2,3-dihydro-[1,4]dioxino[2,3-b]pyridine Chemical compound C1=CC=C2OCCOC2=N1 QWQZJEXJTYAPGE-UHFFFAOYSA-N 0.000 description 1
- JQQFRJLHDFTWAW-UHFFFAOYSA-N 2,3-dihydro-[1,4]dioxino[2,3-c]pyridine Chemical compound N1=CC=C2OCCOC2=C1 JQQFRJLHDFTWAW-UHFFFAOYSA-N 0.000 description 1
- ZHUNCEFRDFCYHG-UHFFFAOYSA-N 2,4-dimethyl-3H-1,3-oxazole-2-carboxylic acid Chemical compound CC1=COC(C)(C(O)=O)N1 ZHUNCEFRDFCYHG-UHFFFAOYSA-N 0.000 description 1
- PBKTZKBXPYOHLE-UHFFFAOYSA-N 2-(1,5-naphthyridin-2-yl)acetic acid Chemical compound N1=CC=CC2=NC(CC(=O)O)=CC=C21 PBKTZKBXPYOHLE-UHFFFAOYSA-N 0.000 description 1
- ALDSXDRDRWDASQ-UHFFFAOYSA-N 2-(3-benzoylphenyl)acetic acid Chemical compound OC(=O)CC1=CC=CC(C(=O)C=2C=CC=CC=2)=C1 ALDSXDRDRWDASQ-UHFFFAOYSA-N 0.000 description 1
- YNTJKQDWYXUTLZ-UHFFFAOYSA-N 2-(3-chlorophenoxy)propanoic acid Chemical compound OC(=O)C(C)OC1=CC=CC(Cl)=C1 YNTJKQDWYXUTLZ-UHFFFAOYSA-N 0.000 description 1
- LMRUJTWIAOQDGG-UHFFFAOYSA-N 2-(3-methylsulfonylphenyl)acetamide Chemical compound CS(=O)(=O)C1=CC=CC(CC(N)=O)=C1 LMRUJTWIAOQDGG-UHFFFAOYSA-N 0.000 description 1
- ILZOEMNCWIVMLT-UHFFFAOYSA-N 2-(4-benzoylphenyl)acetic acid Chemical compound C1=CC(CC(=O)O)=CC=C1C(=O)C1=CC=CC=C1 ILZOEMNCWIVMLT-UHFFFAOYSA-N 0.000 description 1
- CUDDZZCNPJYPBF-UHFFFAOYSA-N 2-(4-chlorophenyl)-2-hydroxypropanoic acid Chemical compound OC(=O)C(O)(C)C1=CC=C(Cl)C=C1 CUDDZZCNPJYPBF-UHFFFAOYSA-N 0.000 description 1
- HUPVBFQYJHFONM-UHFFFAOYSA-N 2-(4-fluorophenyl)acetamide Chemical compound NC(=O)CC1=CC=C(F)C=C1 HUPVBFQYJHFONM-UHFFFAOYSA-N 0.000 description 1
- HQJLSBGJUBPKQC-UHFFFAOYSA-N 2-(5-aminopyridin-2-yl)acetaldehyde Chemical compound NC1=CC=C(CC=O)N=C1 HQJLSBGJUBPKQC-UHFFFAOYSA-N 0.000 description 1
- CRBRKSBTPSXXGY-UHFFFAOYSA-N 2-(5-chloropyridin-2-yl)acetamide Chemical compound NC(=O)CC1=CC=C(Cl)C=N1 CRBRKSBTPSXXGY-UHFFFAOYSA-N 0.000 description 1
- AUCKAGADUXSVKT-UHFFFAOYSA-N 2-(5-chloropyridin-2-yl)acetic acid Chemical compound OC(=O)CC1=CC=C(Cl)C=N1 AUCKAGADUXSVKT-UHFFFAOYSA-N 0.000 description 1
- AIBMLJUMOWQTNN-UHFFFAOYSA-N 2-(oxan-2-yloxy)acetamide Chemical compound NC(=O)COC1CCCCO1 AIBMLJUMOWQTNN-UHFFFAOYSA-N 0.000 description 1
- XXTXDVUAHROLBN-UHFFFAOYSA-N 2-(trifluoromethoxy)benzamide Chemical compound NC(=O)C1=CC=CC=C1OC(F)(F)F XXTXDVUAHROLBN-UHFFFAOYSA-N 0.000 description 1
- HCUARRIEZVDMPT-UHFFFAOYSA-N 2-Indolecarboxylic acid Chemical compound C1=CC=C2NC(C(=O)O)=CC2=C1 HCUARRIEZVDMPT-UHFFFAOYSA-N 0.000 description 1
- IRMTXMJNHRISQH-UHFFFAOYSA-N 2-[2-[2-(diaminomethylideneamino)ethyldisulfanyl]ethyl]guanidine Chemical compound NC(N)=NCCSSCCN=C(N)N IRMTXMJNHRISQH-UHFFFAOYSA-N 0.000 description 1
- YXORDBZLZWAHBB-UHFFFAOYSA-N 2-[3-(trifluoromethyl)phenyl]acetamide Chemical compound NC(=O)CC1=CC=CC(C(F)(F)F)=C1 YXORDBZLZWAHBB-UHFFFAOYSA-N 0.000 description 1
- HNORVZDAANCHAY-UHFFFAOYSA-N 2-[4-(trifluoromethyl)phenyl]acetic acid Chemical compound OC(=O)CC1=CC=C(C(F)(F)F)C=C1 HNORVZDAANCHAY-UHFFFAOYSA-N 0.000 description 1
- LHKKVYVRJWNYNV-UHFFFAOYSA-N 2-[4-(trifluoromethyl)triazol-1-yl]acetic acid Chemical compound OC(=O)CN1C=C(C(F)(F)F)N=N1 LHKKVYVRJWNYNV-UHFFFAOYSA-N 0.000 description 1
- RBHQAIFXLJIFFM-UHFFFAOYSA-N 2-[5-methyl-3-(trifluoromethyl)pyrazol-1-yl]acetic acid Chemical compound CC1=CC(C(F)(F)F)=NN1CC(O)=O RBHQAIFXLJIFFM-UHFFFAOYSA-N 0.000 description 1
- XKSAJZSJKURQRX-UHFFFAOYSA-N 2-acetyloxy-5-(4-fluorophenyl)benzoic acid Chemical compound C1=C(C(O)=O)C(OC(=O)C)=CC=C1C1=CC=C(F)C=C1 XKSAJZSJKURQRX-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
- JUIKUQOUMZUFQT-UHFFFAOYSA-N 2-bromoacetamide Chemical compound NC(=O)CBr JUIKUQOUMZUFQT-UHFFFAOYSA-N 0.000 description 1
- AUHYZQCEIVEMFH-UHFFFAOYSA-N 2-bromopropanamide Chemical compound CC(Br)C(N)=O AUHYZQCEIVEMFH-UHFFFAOYSA-N 0.000 description 1
- MONMFXREYOKQTI-UHFFFAOYSA-M 2-bromopropanoate Chemical compound CC(Br)C([O-])=O MONMFXREYOKQTI-UHFFFAOYSA-M 0.000 description 1
- HJOQGBBHVRYTDX-UHFFFAOYSA-N 2-chloro-7H-pyrrolo[2,3-d]pyrimidine Chemical compound ClC1=NC=C2C=CNC2=N1 HJOQGBBHVRYTDX-UHFFFAOYSA-N 0.000 description 1
- UNCQVRBWJWWJBF-UHFFFAOYSA-N 2-chloropyrimidine Chemical compound ClC1=NC=CC=N1 UNCQVRBWJWWJBF-UHFFFAOYSA-N 0.000 description 1
- YURNCBVQZBJDAJ-UHFFFAOYSA-N 2-heptenoic acid Chemical compound CCCCC=CC(O)=O YURNCBVQZBJDAJ-UHFFFAOYSA-N 0.000 description 1
- 125000004200 2-methoxyethyl group Chemical group [H]C([H])([H])OC([H])([H])C([H])([H])* 0.000 description 1
- IWTFOFMTUOBLHG-UHFFFAOYSA-N 2-methoxypyridine Chemical compound COC1=CC=CC=N1 IWTFOFMTUOBLHG-UHFFFAOYSA-N 0.000 description 1
- YNJXVWIPWJKJJD-UHFFFAOYSA-N 2-methoxypyridine-3-carboxamide Chemical compound COC1=NC=CC=C1C(N)=O YNJXVWIPWJKJJD-UHFFFAOYSA-N 0.000 description 1
- VFUDDKHMYGQRAP-UHFFFAOYSA-N 2-methylbenzenesulfonate;pyridin-1-ium Chemical compound C1=CC=NC=C1.CC1=CC=CC=C1S(O)(=O)=O VFUDDKHMYGQRAP-UHFFFAOYSA-N 0.000 description 1
- JRYYVMDEUJQWRO-UHFFFAOYSA-N 2-methylnicotinamide Chemical compound CC1=NC=CC=C1C(N)=O JRYYVMDEUJQWRO-UHFFFAOYSA-N 0.000 description 1
- YOETUEMZNOLGDB-UHFFFAOYSA-N 2-methylpropyl carbonochloridate Chemical compound CC(C)COC(Cl)=O YOETUEMZNOLGDB-UHFFFAOYSA-N 0.000 description 1
- IJUJVDHUJHWQKP-UHFFFAOYSA-N 2-phenyl-1,3,5-tri(propan-2-yl)benzene Chemical group CC(C)C1=CC(C(C)C)=CC(C(C)C)=C1C1=CC=CC=C1 IJUJVDHUJHWQKP-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
- BUDQDWGNQVEFAC-UHFFFAOYSA-N 3,4-dihydro-2H-pyran Chemical compound C1COC=CC1 BUDQDWGNQVEFAC-UHFFFAOYSA-N 0.000 description 1
- UQADNFKCBLPCTM-UHFFFAOYSA-N 3-(2,2,2-trifluoroacetyl)benzoic acid Chemical compound OC(=O)C1=CC=CC(C(=O)C(F)(F)F)=C1 UQADNFKCBLPCTM-UHFFFAOYSA-N 0.000 description 1
- CELVQYDOLLNPMB-UHFFFAOYSA-N 3-[3-(trifluoromethyl)diazirin-3-yl]benzoic acid Chemical compound OC(=O)C1=CC=CC(C2(N=N2)C(F)(F)F)=C1 CELVQYDOLLNPMB-UHFFFAOYSA-N 0.000 description 1
- ZLAZSJJPVYXOGY-UHFFFAOYSA-N 3-bromo-N-methoxy-N-methylpyridine-4-carboxamide Chemical compound CON(C)C(=O)C1=CC=NC=C1Br ZLAZSJJPVYXOGY-UHFFFAOYSA-N 0.000 description 1
- YDPLFBIGFQFIDB-UHFFFAOYSA-N 3-chloropyridine-2-carbonitrile Chemical compound ClC1=CC=CN=C1C#N YDPLFBIGFQFIDB-UHFFFAOYSA-N 0.000 description 1
- XTMUXJBJCMRWPG-UHFFFAOYSA-N 3-chloropyridine-2-carboxylic acid Chemical compound OC(=O)C1=NC=CC=C1Cl XTMUXJBJCMRWPG-UHFFFAOYSA-N 0.000 description 1
- WQYAZBFZFIUIPL-UHFFFAOYSA-N 3-fluoroazetidine Chemical compound FC1CNC1 WQYAZBFZFIUIPL-UHFFFAOYSA-N 0.000 description 1
- CELKOWQJPVJKIL-UHFFFAOYSA-N 3-fluoropyridine Chemical compound FC1=CC=CN=C1 CELKOWQJPVJKIL-UHFFFAOYSA-N 0.000 description 1
- QMDMUIQZTHZWGH-UHFFFAOYSA-N 3-methylpyridine-4-carboxamide Chemical compound CC1=CN=CC=C1C(N)=O QMDMUIQZTHZWGH-UHFFFAOYSA-N 0.000 description 1
- 125000003349 3-pyridyl group Chemical group N1=C([H])C([*])=C([H])C([H])=C1[H] 0.000 description 1
- JVVRCYWZTJLJSG-UHFFFAOYSA-N 4-Dimethylaminophenol Substances CN(C)C1=CC=C(O)C=C1 JVVRCYWZTJLJSG-UHFFFAOYSA-N 0.000 description 1
- CZPAJVBVULSLGG-UHFFFAOYSA-N 4-[(3R)-3-(trifluoromethyl)diazirin-1-ium-3-yl]benzoate Chemical compound C1=CC(C(=O)O)=CC=C1C1(C(F)(F)F)N=N1 CZPAJVBVULSLGG-UHFFFAOYSA-N 0.000 description 1
- NUILQFNJMVDDJG-UHFFFAOYSA-N 4-aminopyrimidine-2-carboxylic acid Chemical compound NC1=CC=NC(C(O)=O)=N1 NUILQFNJMVDDJG-UHFFFAOYSA-N 0.000 description 1
- 125000004801 4-cyanophenyl group Chemical group [H]C1=C([H])C(C#N)=C([H])C([H])=C1* 0.000 description 1
- CMKZSIGRFONKKC-UHFFFAOYSA-N 4-cyanopyridine-2-carboxylic acid Chemical compound OC(=O)C1=CC(C#N)=CC=N1 CMKZSIGRFONKKC-UHFFFAOYSA-N 0.000 description 1
- 229960000549 4-dimethylaminophenol Drugs 0.000 description 1
- UELRAKDBDJRXST-UHFFFAOYSA-N 4-methoxypyridine-2-carboxylic acid Chemical compound COC1=CC=NC(C(O)=O)=C1 UELRAKDBDJRXST-UHFFFAOYSA-N 0.000 description 1
- SAJWBBTYRMBHDQ-UHFFFAOYSA-N 4-methoxypyrimidine-2-carboxylic acid Chemical compound COC1=CC=NC(C(O)=O)=N1 SAJWBBTYRMBHDQ-UHFFFAOYSA-N 0.000 description 1
- BNNMDMGPZUOOOE-UHFFFAOYSA-N 4-methylbenzenesulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1.CC1=CC=C(S(O)(=O)=O)C=C1 BNNMDMGPZUOOOE-UHFFFAOYSA-N 0.000 description 1
- LVEIJLGSXRYDEY-UHFFFAOYSA-N 4-propan-2-yl-2H-triazole Chemical compound CC(C)C1=CNN=N1 LVEIJLGSXRYDEY-UHFFFAOYSA-N 0.000 description 1
- 125000004199 4-trifluoromethylphenyl group Chemical group [H]C1=C([H])C(=C([H])C([H])=C1*)C(F)(F)F 0.000 description 1
- PQUGCKBLVKJMNT-UHFFFAOYSA-N 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-(trifluoromethyl)pyrazole Chemical compound C1=CC(OC)=CC=C1N1C(C=2C=CC(Cl)=CC=2)=CC(C(F)(F)F)=N1 PQUGCKBLVKJMNT-UHFFFAOYSA-N 0.000 description 1
- ULGVHUUBIHTFAM-LZYBPNLTSA-N 5-[(2E)-2-(3-carboxy-4-oxocyclohexa-2,5-dien-1-ylidene)hydrazinyl]-2-hydroxybenzoic acid Chemical compound C1=CC(=O)C(C(=O)O)=C\C1=N\NC1=CC=C(O)C(C(O)=O)=C1 ULGVHUUBIHTFAM-LZYBPNLTSA-N 0.000 description 1
- KQPKPCNLIDLUMF-MRVPVSSYSA-N 5-[(2R)-pentan-2-yl]-5-prop-2-enyl-1,3-diazinane-2,4,6-trione Chemical compound CCC[C@@H](C)C1(CC=C)C(=O)NC(=O)NC1=O KQPKPCNLIDLUMF-MRVPVSSYSA-N 0.000 description 1
- JOPSSWGWLCLPPF-RUDMXATFSA-N 5-[2-(2-carboxyethyl)-3-[(E)-6-(4-methoxyphenyl)hex-5-enoxy]phenoxy]pentanoic acid Chemical compound C1=CC(OC)=CC=C1\C=C\CCCCOC1=CC=CC(OCCCCC(O)=O)=C1CCC(O)=O JOPSSWGWLCLPPF-RUDMXATFSA-N 0.000 description 1
- CWSZBVAUYPTXTG-UHFFFAOYSA-N 5-[6-[[3,4-dihydroxy-6-(hydroxymethyl)-5-methoxyoxan-2-yl]oxymethyl]-3,4-dihydroxy-5-[4-hydroxy-3-(2-hydroxyethoxy)-6-(hydroxymethyl)-5-methoxyoxan-2-yl]oxyoxan-2-yl]oxy-6-(hydroxymethyl)-2-methyloxane-3,4-diol Chemical compound O1C(CO)C(OC)C(O)C(O)C1OCC1C(OC2C(C(O)C(OC)C(CO)O2)OCCO)C(O)C(O)C(OC2C(OC(C)C(O)C2O)CO)O1 CWSZBVAUYPTXTG-UHFFFAOYSA-N 0.000 description 1
- ZQVLPMNLLKGGIU-UHFFFAOYSA-N 5-bromopyridine-2-carbaldehyde Chemical compound BrC1=CC=C(C=O)N=C1 ZQVLPMNLLKGGIU-UHFFFAOYSA-N 0.000 description 1
- MPZDNIJHHXRTIQ-UHFFFAOYSA-N 6-chloroimidazo[1,2-b]pyridazine Chemical compound N1=C(Cl)C=CC2=NC=CN21 MPZDNIJHHXRTIQ-UHFFFAOYSA-N 0.000 description 1
- YXPYIZUQEDQMPS-UHFFFAOYSA-N 6-methoxyquinoline-2-carboxylic acid Chemical compound N1=C(C(O)=O)C=CC2=CC(OC)=CC=C21 YXPYIZUQEDQMPS-UHFFFAOYSA-N 0.000 description 1
- VIQHWADMQYGCLE-UHFFFAOYSA-N 7-(difluoromethyl)-5-methyl-[1,2,4]triazolo[1,5-a]pyrimidine-2-carboxylic acid Chemical compound N1=C(C)C=C(C(F)F)N2N=C(C(O)=O)N=C21 VIQHWADMQYGCLE-UHFFFAOYSA-N 0.000 description 1
- IQNYWAJVTZJHLI-UHFFFAOYSA-N 7-tert-butyl-2-chloropyrrolo[2,3-d]pyrimidine Chemical compound N1=C(Cl)N=C2N(C(C)(C)C)C=CC2=C1 IQNYWAJVTZJHLI-UHFFFAOYSA-N 0.000 description 1
- ACSNBCFUYLEILS-UHFFFAOYSA-N 7-tert-butyl-4-chloropyrrolo[2,3-d]pyrimidine Chemical compound N1=CN=C2N(C(C)(C)C)C=CC2=C1Cl ACSNBCFUYLEILS-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
- YVVMIGRXQRPSIY-UHFFFAOYSA-N 7H-pyrrolo[2,3-d]pyrimidin-2-amine Chemical compound N1C(N)=NC=C2C=CN=C21 YVVMIGRXQRPSIY-UHFFFAOYSA-N 0.000 description 1
- PEHVGBZKEYRQSX-UHFFFAOYSA-N 7H-pyrrolo[2,3-d]pyrimidin-4-amine Chemical class NC1=NC=NC2=C1C=CN2 PEHVGBZKEYRQSX-UHFFFAOYSA-N 0.000 description 1
- 208000002008 AIDS-Related Lymphoma Diseases 0.000 description 1
- 101700069440 AIFM2 Proteins 0.000 description 1
- 101700061329 ARTN Proteins 0.000 description 1
- 101710034857 ATIC Proteins 0.000 description 1
- 210000001015 Abdomen Anatomy 0.000 description 1
- 210000000683 Abdominal Cavity Anatomy 0.000 description 1
- 208000004998 Abdominal Pain Diseases 0.000 description 1
- YBCVMFKXIKNREZ-UHFFFAOYSA-N AcOH acetic acid Chemical compound CC(O)=O.CC(O)=O YBCVMFKXIKNREZ-UHFFFAOYSA-N 0.000 description 1
- 206010000565 Acquired immunodeficiency syndrome Diseases 0.000 description 1
- 208000009956 Adenocarcinoma Diseases 0.000 description 1
- 208000007848 Alcoholism Diseases 0.000 description 1
- XJKJWTWGDGIQRH-BFIDDRIFSA-N Alginic acid Chemical compound O1[C@@H](C(O)=O)[C@@H](OC)[C@H](O)[C@H](O)[C@@H]1O[C@@H]1[C@@H](C(O)=O)O[C@@H](C)[C@@H](O)[C@H]1O XJKJWTWGDGIQRH-BFIDDRIFSA-N 0.000 description 1
- NTJOBXMMWNYJFB-UHFFFAOYSA-N Amisulpride Chemical compound CCN1CCCC1CNC(=O)C1=CC(S(=O)(=O)CC)=C(N)C=C1OC NTJOBXMMWNYJFB-UHFFFAOYSA-N 0.000 description 1
- 239000005695 Ammonium acetate Substances 0.000 description 1
- 229910000013 Ammonium bicarbonate Inorganic materials 0.000 description 1
- PRKQVKDSMLBJBJ-UHFFFAOYSA-N Ammonium carbonate Chemical compound N.N.OC(O)=O PRKQVKDSMLBJBJ-UHFFFAOYSA-N 0.000 description 1
- 229960001301 Amobarbital Drugs 0.000 description 1
- VIROVYVQCGLCII-UHFFFAOYSA-N Amobarbital Chemical compound CC(C)CCC1(CC)C(=O)NC(=O)NC1=O VIROVYVQCGLCII-UHFFFAOYSA-N 0.000 description 1
- 208000007502 Anemia Diseases 0.000 description 1
- 206010002383 Angina pectoris Diseases 0.000 description 1
- 206010002512 Anhidrosis Diseases 0.000 description 1
- 206010002556 Ankylosing spondylitis Diseases 0.000 description 1
- UIQMVEYFGZJHCZ-SSTWWWIQSA-N Antorphin Chemical compound C([C@@H](N(CC1)CC=C)[C@@H]2C=C[C@@H]3O)C4=CC=C(O)C5=C4[C@@]21[C@H]3O5 UIQMVEYFGZJHCZ-SSTWWWIQSA-N 0.000 description 1
- ATALOFNDEOCMKK-OITMNORJSA-N Aprepitant Chemical compound O([C@@H]([C@@H]1C=2C=CC(F)=CC=2)O[C@H](C)C=2C=C(C=C(C=2)C(F)(F)F)C(F)(F)F)CCN1CC1=NNC(=O)N1 ATALOFNDEOCMKK-OITMNORJSA-N 0.000 description 1
- UORJNBVJVRLXMQ-UHFFFAOYSA-N Aprobarbital Chemical compound C=CCC1(C(C)C)C(=O)NC(=O)NC1=O UORJNBVJVRLXMQ-UHFFFAOYSA-N 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 206010003246 Arthritis Diseases 0.000 description 1
- VSWBSWWIRNCQIJ-GJZGRUSLSA-N Asenapine Chemical compound O1C2=CC=CC=C2[C@@H]2CN(C)C[C@H]2C2=CC(Cl)=CC=C21 VSWBSWWIRNCQIJ-GJZGRUSLSA-N 0.000 description 1
- 229960005245 Asenapine Drugs 0.000 description 1
- 229940009098 Aspartate Drugs 0.000 description 1
- IUVMAUQEZFTTFB-YUMQZZPRSA-N Atagabalin Chemical compound C[C@H]1CC(CN)(CC(O)=O)C[C@@H]1C IUVMAUQEZFTTFB-YUMQZZPRSA-N 0.000 description 1
- 206010003816 Autoimmune disease Diseases 0.000 description 1
- XYOVOXDWRFGKEX-UHFFFAOYSA-N Azepine Chemical compound N1C=CC=CC=C1 XYOVOXDWRFGKEX-UHFFFAOYSA-N 0.000 description 1
- 206010003885 Azotaemia Diseases 0.000 description 1
- SECKRCOLJRRGGV-UHFFFAOYSA-N BAY 38-9456 Chemical compound CCCC1=NC(C)=C(C(N=2)=O)N1NC=2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(CC)CC1 SECKRCOLJRRGGV-UHFFFAOYSA-N 0.000 description 1
- MUALRAIOVNYAIW-UHFFFAOYSA-N BINAP Chemical compound C1=CC=CC=C1P(C=1C(=C2C=CC=CC2=CC=1)C=1C2=CC=CC=C2C=CC=1P(C=1C=CC=CC=1)C=1C=CC=CC=1)C1=CC=CC=C1 MUALRAIOVNYAIW-UHFFFAOYSA-N 0.000 description 1
- KPYSYYIEGFHWSV-UHFFFAOYSA-N Baclofen Chemical compound OC(=O)CC(CN)C1=CC=C(Cl)C=C1 KPYSYYIEGFHWSV-UHFFFAOYSA-N 0.000 description 1
- 229960000794 Baclofen Drugs 0.000 description 1
- JUHORIMYRDESRB-UHFFFAOYSA-N Benzathine Chemical compound C=1C=CC=CC=1CNCCNCC1=CC=CC=C1 JUHORIMYRDESRB-UHFFFAOYSA-N 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-N Benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- CYGODHVAJQTCBG-UHFFFAOYSA-N Bifeprunox Chemical compound C=12OC(=O)NC2=CC=CC=1N(CC1)CCN1CC(C=1)=CC=CC=1C1=CC=CC=C1 CYGODHVAJQTCBG-UHFFFAOYSA-N 0.000 description 1
- 229950009087 Bifeprunox Drugs 0.000 description 1
- 230000036912 Bioavailability Effects 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- XVGOZDAJGBALKS-UHFFFAOYSA-N Blonanserin Chemical compound C1CN(CC)CCN1C1=CC(C=2C=CC(F)=CC=2)=C(CCCCCC2)C2=N1 XVGOZDAJGBALKS-UHFFFAOYSA-N 0.000 description 1
- 229950002871 Blonanserin Drugs 0.000 description 1
- 210000000601 Blood Cells Anatomy 0.000 description 1
- 206010006002 Bone pain Diseases 0.000 description 1
- 239000004135 Bone phosphate Substances 0.000 description 1
- 210000004556 Brain Anatomy 0.000 description 1
- 208000000003 Breakthrough Pain Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- QIHLUZAFSSMXHQ-UHFFFAOYSA-N Budipine Chemical compound C1CN(C(C)(C)C)CCC1(C=1C=CC=CC=1)C1=CC=CC=C1 QIHLUZAFSSMXHQ-UHFFFAOYSA-N 0.000 description 1
- 229960002452 Budipine Drugs 0.000 description 1
- ZRIHAIZYIMGOAB-UHFFFAOYSA-N Butabarbital Chemical compound CCC(C)C1(CC)C(=O)NC(=O)NC1=O ZRIHAIZYIMGOAB-UHFFFAOYSA-N 0.000 description 1
- IFKLAQQSCNILHL-QHAWAJNXSA-N Butorphanol Chemical compound N1([C@@H]2CC3=CC=C(C=C3[C@@]3([C@]2(CCCC3)O)CC1)O)CC1CCC1 IFKLAQQSCNILHL-QHAWAJNXSA-N 0.000 description 1
- 229960001113 Butorphanol Drugs 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
- 240000001546 Byrsonima crassifolia Species 0.000 description 1
- 235000003197 Byrsonima crassifolia Nutrition 0.000 description 1
- PSCLGUIRVJRYJG-UHFFFAOYSA-N C1=C2OCOC2=CC(C2=NC(C3=CC4=C5C=CC=CC5=NC4=CN3C2=O)=O)=C1 Chemical compound C1=C2OCOC2=CC(C2=NC(C3=CC4=C5C=CC=CC5=NC4=CN3C2=O)=O)=C1 PSCLGUIRVJRYJG-UHFFFAOYSA-N 0.000 description 1
- SLHCMPVPVQSTBV-UHFFFAOYSA-N C1=CNC2=C3C=CN=C3C=CC2=C1 Chemical compound C1=CNC2=C3C=CN=C3C=CC2=C1 SLHCMPVPVQSTBV-UHFFFAOYSA-N 0.000 description 1
- JVCXXTVXDMJSIE-UXLLHSPISA-N C1C2=C(O)C=CC=C2[C@]23CCN(C)[C@H]1[C@@H]2CCCC3 Chemical compound C1C2=C(O)C=CC=C2[C@]23CCN(C)[C@H]1[C@@H]2CCCC3 JVCXXTVXDMJSIE-UXLLHSPISA-N 0.000 description 1
- 101710009963 CCDC85B Proteins 0.000 description 1
- 210000003996 CFU-GM Anatomy 0.000 description 1
- DTBJMHUKRKATAS-RWANSRKNSA-N C[C@H](COC1OCCCC1)C=1N=CC2=C(N=1)NC=C2 Chemical compound C[C@H](COC1OCCCC1)C=1N=CC2=C(N=1)NC=C2 DTBJMHUKRKATAS-RWANSRKNSA-N 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N C[N+](C)(C)CCO Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- CJZGTCYPCWQAJB-UHFFFAOYSA-L Calcium stearate Chemical compound [Ca+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O CJZGTCYPCWQAJB-UHFFFAOYSA-L 0.000 description 1
- YQEZLKZALYSWHR-UHFFFAOYSA-N Calypsol Chemical compound C=1C=CC=C(Cl)C=1C1(NC)CCCCC1=O YQEZLKZALYSWHR-UHFFFAOYSA-N 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
- DRCMAZOSEIMCHM-UHFFFAOYSA-N Capsazepine Chemical compound C1C=2C=C(O)C(O)=CC=2CCCN1C(=S)NCCC1=CC=C(Cl)C=C1 DRCMAZOSEIMCHM-UHFFFAOYSA-N 0.000 description 1
- 239000004215 Carbon black (E152) Substances 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 229940105329 Carboxymethylcellulose Drugs 0.000 description 1
- 206010007275 Carcinoid tumour Diseases 0.000 description 1
- OFZCIYFFPZCNJE-UHFFFAOYSA-N Carisoprodol Chemical compound NC(=O)OCC(C)(CCC)COC(=O)NC(C)C OFZCIYFFPZCNJE-UHFFFAOYSA-N 0.000 description 1
- 229960004587 Carisoprodol Drugs 0.000 description 1
- 208000003295 Carpal Tunnel Syndrome Diseases 0.000 description 1
- RZEKVGVHFLEQIL-UHFFFAOYSA-N Celecoxib Chemical compound C1=CC(C)=CC=C1C1=CC(C(F)(F)F)=NN1C1=CC=C(S(N)(=O)=O)C=C1 RZEKVGVHFLEQIL-UHFFFAOYSA-N 0.000 description 1
- 210000000170 Cell Membrane Anatomy 0.000 description 1
- PTHCMJGKKRQCBF-UHFFFAOYSA-N Cellulose, microcrystalline Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC)C(CO)O1 PTHCMJGKKRQCBF-UHFFFAOYSA-N 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 229940112822 Chewing Gum Drugs 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- WFNAKBGANONZEQ-UHFFFAOYSA-N Chlorcyclizine Chemical compound C1CN(C)CCN1C(C=1C=CC(Cl)=CC=1)C1=CC=CC=C1 WFNAKBGANONZEQ-UHFFFAOYSA-N 0.000 description 1
- 229960004782 Chlordiazepoxide Drugs 0.000 description 1
- ANTSCNMPPGJYLG-UHFFFAOYSA-N Chlordiazepoxide Chemical compound O=N=1CC(NC)=NC2=CC=C(Cl)C=C2C=1C1=CC=CC=C1 ANTSCNMPPGJYLG-UHFFFAOYSA-N 0.000 description 1
- 241000088885 Chlorops Species 0.000 description 1
- 229940107080 Chlorpheniramine Drugs 0.000 description 1
- ZPEIMTDSQAKGNT-UHFFFAOYSA-N Chlorpromazine Chemical compound C1=C(Cl)C=C2N(CCCN(C)C)C3=CC=CC=C3SC2=C1 ZPEIMTDSQAKGNT-UHFFFAOYSA-N 0.000 description 1
- 229960001076 Chlorpromazine Drugs 0.000 description 1
- TZFWDZFKRBELIQ-UHFFFAOYSA-N Chlorzoxazone Chemical compound ClC1=CC=C2OC(O)=NC2=C1 TZFWDZFKRBELIQ-UHFFFAOYSA-N 0.000 description 1
- 229960003633 Chlorzoxazone Drugs 0.000 description 1
- 229960001231 Choline Drugs 0.000 description 1
- 235000001258 Cinchona calisaya Nutrition 0.000 description 1
- 241000434299 Cinchona officinalis Species 0.000 description 1
- QZUDBNBUXVUHMW-UHFFFAOYSA-N Clozapine Chemical compound C1CN(C)CCN1C1=NC2=CC(Cl)=CC=C2NC2=CC=CC=C12 QZUDBNBUXVUHMW-UHFFFAOYSA-N 0.000 description 1
- 208000006561 Cluster Headache Diseases 0.000 description 1
- 229940108066 Coal Tar Drugs 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 208000006509 Congenital Pain Insensitivity Diseases 0.000 description 1
- 208000002573 Connective Tissue Disease Diseases 0.000 description 1
- OPQARKPSCNTWTJ-UHFFFAOYSA-L Copper(II) acetate Chemical compound [Cu+2].CC([O-])=O.CC([O-])=O OPQARKPSCNTWTJ-UHFFFAOYSA-L 0.000 description 1
- 229960001681 Croscarmellose Sodium Drugs 0.000 description 1
- 229920002785 Croscarmellose sodium Polymers 0.000 description 1
- 229960000913 Crospovidone Drugs 0.000 description 1
- FSIRGTNPQFDCCD-QGMBQPNBSA-N Cyanodothiepin Chemical compound C1SC2=CC=C(C#N)C=C2C(=C/CCN(C)C)/C2=CC=CC=C21 FSIRGTNPQFDCCD-QGMBQPNBSA-N 0.000 description 1
- JURKNVYFZMSNLP-UHFFFAOYSA-N Cyclobenzaprine Chemical compound C1=CC2=CC=CC=C2C(=CCCN(C)C)C2=CC=CC=C21 JURKNVYFZMSNLP-UHFFFAOYSA-N 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N D-Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-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
- AEMOLEFTQBMNLQ-AQKNRBDQSA-N D-glucopyranuronic acid Chemical compound OC1O[C@H](C(O)=O)[C@@H](O)[C@H](O)[C@H]1O AEMOLEFTQBMNLQ-AQKNRBDQSA-N 0.000 description 1
- DEZRYPDIMOWBDS-UHFFFAOYSA-N DCM Dichloromethane Chemical compound ClCCl.ClCCl DEZRYPDIMOWBDS-UHFFFAOYSA-N 0.000 description 1
- CKLJMWTZIZZHCS-UHFFFAOYSA-N DL-aspartic acid Chemical compound OC(=O)C(N)CC(O)=O CKLJMWTZIZZHCS-UHFFFAOYSA-N 0.000 description 1
- UZZWBUYVTBPQIV-UHFFFAOYSA-N DME dimethoxyethane Chemical compound COCCOC.COCCOC UZZWBUYVTBPQIV-UHFFFAOYSA-N 0.000 description 1
- CETRZFQIITUQQL-UHFFFAOYSA-N DMSO dimethylsulfoxide Chemical compound CS(C)=O.CS(C)=O CETRZFQIITUQQL-UHFFFAOYSA-N 0.000 description 1
- 229950007605 Dapitant Drugs 0.000 description 1
- USRHYDPUVLEVMC-FQEVSTJZSA-N Dapoxetine Chemical compound C1([C@H](CCOC=2C3=CC=CC=C3C=CC=2)N(C)C)=CC=CC=C1 USRHYDPUVLEVMC-FQEVSTJZSA-N 0.000 description 1
- HXGBXQDTNZMWGS-RUZDIDTESA-N Darifenacin Chemical compound C=1C=CC=CC=1C([C@H]1CN(CCC=2C=C3CCOC3=CC=2)CC1)(C(=O)N)C1=CC=CC=C1 HXGBXQDTNZMWGS-RUZDIDTESA-N 0.000 description 1
- WAZQAZKAZLXFMK-UHFFFAOYSA-N Deracoxib Chemical compound C1=C(F)C(OC)=CC=C1C1=CC(C(F)F)=NN1C1=CC=C(S(N)(=O)=O)C=C1 WAZQAZKAZLXFMK-UHFFFAOYSA-N 0.000 description 1
- HCYAFALTSJYZDH-UHFFFAOYSA-N Desimpramine Chemical compound C1CC2=CC=CC=C2N(CCCNC)C2=CC=CC=C21 HCYAFALTSJYZDH-UHFFFAOYSA-N 0.000 description 1
- KYYIDSXMWOZKMP-UHFFFAOYSA-N Desvenlafaxine Chemical compound C1CCCCC1(O)C(CN(C)C)C1=CC=C(O)C=C1 KYYIDSXMWOZKMP-UHFFFAOYSA-N 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N Dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 229960003957 Dexamethasone Drugs 0.000 description 1
- SOYKEARSMXGVTM-HNNXBMFYSA-N Dexchlorpheniramine Chemical compound C1([C@H](CCN(C)C)C=2N=CC=CC=2)=CC=C(Cl)C=C1 SOYKEARSMXGVTM-HNNXBMFYSA-N 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- XLMALTXPSGQGBX-GCJKJVERSA-N Dextropropoxyphene Chemical compound C([C@](OC(=O)CC)([C@H](C)CN(C)C)C=1C=CC=CC=1)C1=CC=CC=C1 XLMALTXPSGQGBX-GCJKJVERSA-N 0.000 description 1
- 229950006878 Dextrorphan Drugs 0.000 description 1
- AAOVKJBEBIDNHE-UHFFFAOYSA-N Diazepam Chemical compound N=1CC(=O)N(C)C2=CC=C(Cl)C=C2C=1C1=CC=CC=C1 AAOVKJBEBIDNHE-UHFFFAOYSA-N 0.000 description 1
- WMKGGPCROCCUDY-PHEQNACWSA-N Dibenzylideneacetone Chemical compound C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1 WMKGGPCROCCUDY-PHEQNACWSA-N 0.000 description 1
- ATKXDQOHNICLQW-UHFFFAOYSA-N Dichloralphenazone Chemical compound OC(O)C(Cl)(Cl)Cl.OC(O)C(Cl)(Cl)Cl.CN1C(C)=CC(=O)N1C1=CC=CC=C1 ATKXDQOHNICLQW-UHFFFAOYSA-N 0.000 description 1
- 208000005407 Digestive System Disease Diseases 0.000 description 1
- RBOXVHNMENFORY-DNJOTXNNSA-N Dihydrocodeine Chemical compound C([C@H]1[C@H](N(CC[C@@]112)C)C3)C[C@H](O)[C@@H]1OC1=C2C3=CC=C1OC RBOXVHNMENFORY-DNJOTXNNSA-N 0.000 description 1
- ZZVUWRFHKOJYTH-UHFFFAOYSA-N Diphenhydramine Chemical compound C=1C=CC=CC=1C(OCCN(C)C)C1=CC=CC=C1 ZZVUWRFHKOJYTH-UHFFFAOYSA-N 0.000 description 1
- 206010013142 Disinhibition Diseases 0.000 description 1
- XIQVNETUBQGFHX-UHFFFAOYSA-N Ditropan Chemical compound C=1C=CC=CC=1C(O)(C(=O)OCC#CCN(CC)CC)C1CCCCC1 XIQVNETUBQGFHX-UHFFFAOYSA-N 0.000 description 1
- ADEBPBSSDYVVLD-UHFFFAOYSA-N Donepezil Chemical compound O=C1C=2C=C(OC)C(OC)=CC=2CC1CC(CC1)CCN1CC1=CC=CC=C1 ADEBPBSSDYVVLD-UHFFFAOYSA-N 0.000 description 1
- RUZYUOTYCVRMRZ-UHFFFAOYSA-N Doxazosin Chemical compound C1OC2=CC=CC=C2OC1C(=O)N(CC1)CCN1C1=NC(N)=C(C=C(C(OC)=C2)OC)C2=N1 RUZYUOTYCVRMRZ-UHFFFAOYSA-N 0.000 description 1
- 229960001389 Doxazosin Drugs 0.000 description 1
- RMEDXOLNCUSCGS-UHFFFAOYSA-N Droperidol Chemical compound C1=CC(F)=CC=C1C(=O)CCCN1CC=C(N2C(NC3=CC=CC=C32)=O)CC1 RMEDXOLNCUSCGS-UHFFFAOYSA-N 0.000 description 1
- 229960000394 Droperidol Drugs 0.000 description 1
- 229940112141 Dry Powder Inhaler Drugs 0.000 description 1
- ZEUITGRIYCTCEM-KRWDZBQOSA-N Duloxetine Chemical compound C1([C@@H](OC=2C3=CC=CC=C3C=CC=2)CCNC)=CC=CS1 ZEUITGRIYCTCEM-KRWDZBQOSA-N 0.000 description 1
- 101700053672 ETV6 Proteins 0.000 description 1
- 102100002089 ETV6 Human genes 0.000 description 1
- JAZBEHYOTPTENJ-JLNKQSITSA-N Eicosapentaenoic acid Chemical compound CC\C=C/C\C=C/C\C=C/C\C=C/C\C=C/CCCC(O)=O JAZBEHYOTPTENJ-JLNKQSITSA-N 0.000 description 1
- OTLDLQZJRFYOJR-LJQANCHMSA-N Eletriptan Chemical compound CN1CCC[C@@H]1CC1=CN=C2[C]1C=C(CCS(=O)(=O)C=1C=CC=CC=1)C=C2 OTLDLQZJRFYOJR-LJQANCHMSA-N 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 229940095399 Enema Drugs 0.000 description 1
- 241000792859 Enema Species 0.000 description 1
- 206010015037 Epilepsy Diseases 0.000 description 1
- VAIOZOCLKVMIMN-PRJWTAEASA-N Eplivanserin Chemical compound C=1C=CC=C(F)C=1\C(=N/OCCN(C)C)\C=C\C1=CC=C(O)C=C1 VAIOZOCLKVMIMN-PRJWTAEASA-N 0.000 description 1
- 229950000789 Eplivanserin Drugs 0.000 description 1
- WSEQXVZVJXJVFP-FQEVSTJZSA-N Escitalopram Chemical compound C1([C@]2(C3=CC=C(C=C3CO2)C#N)CCCN(C)C)=CC=C(F)C=C1 WSEQXVZVJXJVFP-FQEVSTJZSA-N 0.000 description 1
- AFAXGSQYZLGZPG-UHFFFAOYSA-N Ethanedisulfonic acid Chemical compound OS(=O)(=O)CCS(O)(=O)=O AFAXGSQYZLGZPG-UHFFFAOYSA-N 0.000 description 1
- 229960005293 Etodolac Drugs 0.000 description 1
- MNJVRJDLRVPLFE-UHFFFAOYSA-N Etoricoxib Chemical compound C1=NC(C)=CC=C1C1=NC=C(Cl)C=C1C1=CC=C(S(C)(=O)=O)C=C1 MNJVRJDLRVPLFE-UHFFFAOYSA-N 0.000 description 1
- 241000400611 Eucalyptus deanei Species 0.000 description 1
- 101700026954 FA2H Proteins 0.000 description 1
- 102100018264 FAAH Human genes 0.000 description 1
- 101700065602 FAAH Proteins 0.000 description 1
- 229950007979 FLUFENISAL Drugs 0.000 description 1
- 206010016059 Facial pain Diseases 0.000 description 1
- IUJDSEJGGMCXSG-UHFFFAOYSA-N Farmotal Chemical compound CCCC(C)C1(CC)C(=O)NC(=S)NC1=O IUJDSEJGGMCXSG-UHFFFAOYSA-N 0.000 description 1
- OJSFTALXCYKKFQ-YLJYHZDGSA-N Femoxetine Chemical compound C1=CC(OC)=CC=C1OC[C@@H]1[C@@H](C=2C=CC=CC=2)CCN(C)C1 OJSFTALXCYKKFQ-YLJYHZDGSA-N 0.000 description 1
- PJMPHNIQZUBGLI-UHFFFAOYSA-N Fentanyl Chemical compound C=1C=CC=CC=1N(C(=O)CC)C(CC1)CCN1CCC1=CC=CC=C1 PJMPHNIQZUBGLI-UHFFFAOYSA-N 0.000 description 1
- 229960002428 Fentanyl Drugs 0.000 description 1
- KTWOOEGAPBSYNW-UHFFFAOYSA-N Ferrocene Chemical compound [Fe+2].C=1C=C[CH-]C=1.C=1C=C[CH-]C=1 KTWOOEGAPBSYNW-UHFFFAOYSA-N 0.000 description 1
- NELSQLPTEWCHQW-UHFFFAOYSA-N Fezolamine Chemical compound N=1N(CCCN(C)C)C=C(C=2C=CC=CC=2)C=1C1=CC=CC=C1 NELSQLPTEWCHQW-UHFFFAOYSA-N 0.000 description 1
- 229950000761 Fezolamine Drugs 0.000 description 1
- 229960002690 Fluphenazine Drugs 0.000 description 1
- PLDUPXSUYLZYBN-UHFFFAOYSA-N Fluphenazine dihydrochloride Chemical compound C1CN(CCO)CCN1CCCN1C2=CC(C(F)(F)F)=CC=C2SC2=CC=CC=C21 PLDUPXSUYLZYBN-UHFFFAOYSA-N 0.000 description 1
- SAADBVWGJQAEFS-UHFFFAOYSA-N Flurazepam Chemical compound N=1CC(=O)N(CCN(CC)CC)C2=CC=C(Cl)C=C2C=1C1=CC=CC=C1F SAADBVWGJQAEFS-UHFFFAOYSA-N 0.000 description 1
- 229960003528 Flurazepam Drugs 0.000 description 1
- SYTBZMRGLBWNTM-UHFFFAOYSA-N Flurbiprofen Chemical compound FC1=CC(C(C(O)=O)C)=CC=C1C1=CC=CC=C1 SYTBZMRGLBWNTM-UHFFFAOYSA-N 0.000 description 1
- 229960004038 Fluvoxamine Drugs 0.000 description 1
- 239000005715 Fructose Substances 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- 101710037135 GAPC2 Proteins 0.000 description 1
- 229960001731 GLUCEPTATE Drugs 0.000 description 1
- 101710042159 GYPC Proteins 0.000 description 1
- UGJMXCAKCUNAIE-UHFFFAOYSA-N Gabapen Chemical compound OC(=O)CC1(CN)CCCCC1 UGJMXCAKCUNAIE-UHFFFAOYSA-N 0.000 description 1
- 208000009471 Gastroesophageal Reflux Diseases 0.000 description 1
- 208000008665 Gastrointestinal Disease Diseases 0.000 description 1
- 206010017885 Gastrooesophageal reflux disease Diseases 0.000 description 1
- JMBQKKAJIKAWKF-UHFFFAOYSA-N Glutethimide Chemical compound C=1C=CC=CC=1C1(CC)CCC(=O)NC1=O JMBQKKAJIKAWKF-UHFFFAOYSA-N 0.000 description 1
- 229960002972 Glutethimide Drugs 0.000 description 1
- 229960002449 Glycine Drugs 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- INJOMKTZOLKMBF-UHFFFAOYSA-N Guanfacine Chemical compound NC(=N)NC(=O)CC1=C(Cl)C=CC=C1Cl INJOMKTZOLKMBF-UHFFFAOYSA-N 0.000 description 1
- 229960002048 Guanfacine Drugs 0.000 description 1
- RQFCJASXJCIDSX-UUOKFMHZSA-N Guanosine monophosphate Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H]1O RQFCJASXJCIDSX-UUOKFMHZSA-N 0.000 description 1
- 101700018402 HTR1B Proteins 0.000 description 1
- 102100018050 HTR1B Human genes 0.000 description 1
- 101710045396 HTR2B Proteins 0.000 description 1
- LNEPOXFFQSENCJ-UHFFFAOYSA-N Haloperidol Chemical compound C1CC(O)(C=2C=CC(Cl)=CC=2)CCN1CCCC(=O)C1=CC=C(F)C=C1 LNEPOXFFQSENCJ-UHFFFAOYSA-N 0.000 description 1
- 210000003128 Head Anatomy 0.000 description 1
- 206010073073 Hepatobiliary cancer Diseases 0.000 description 1
- 210000003494 Hepatocytes Anatomy 0.000 description 1
- GVGLGOZIDCSQPN-PVHGPHFFSA-N Heroin Chemical compound O([C@H]1[C@H](C=C[C@H]23)OC(C)=O)C4=C5[C@@]12CCN(C)[C@@H]3CC5=CC=C4OC(C)=O GVGLGOZIDCSQPN-PVHGPHFFSA-N 0.000 description 1
- 229940120060 Heroin Drugs 0.000 description 1
- LJQLCJWAZJINEB-UHFFFAOYSA-N Hexafluorophosphate Chemical compound F[P-](F)(F)(F)(F)F LJQLCJWAZJINEB-UHFFFAOYSA-N 0.000 description 1
- 229950000177 Hibenzate Drugs 0.000 description 1
- 239000004705 High-molecular-weight polyethylene Substances 0.000 description 1
- 241001539176 Hime Species 0.000 description 1
- 206010060800 Hot flush Diseases 0.000 description 1
- LLPOLZWFYMWNKH-CMKMFDCUSA-N Hydrocodone Chemical compound C([C@H]1[C@H](N(CC[C@@]112)C)C3)CC(=O)[C@@H]1OC1=C2C3=CC=C1OC LLPOLZWFYMWNKH-CMKMFDCUSA-N 0.000 description 1
- 102000004157 Hydrolases Human genes 0.000 description 1
- 108090000604 Hydrolases Proteins 0.000 description 1
- WVLOADHCBXTIJK-YNHQPCIGSA-N Hydromorphone Chemical compound O([C@H]1C(CC[C@H]23)=O)C4=C5[C@@]12CCN(C)[C@@H]3CC5=CC=C4O WVLOADHCBXTIJK-YNHQPCIGSA-N 0.000 description 1
- 239000004354 Hydroxyethyl cellulose Substances 0.000 description 1
- 229920000663 Hydroxyethyl cellulose Polymers 0.000 description 1
- 241000257303 Hymenoptera Species 0.000 description 1
- 208000003532 Hypothyroidism Diseases 0.000 description 1
- HEFNNWSXXWATRW-UHFFFAOYSA-N Ibuprofen Chemical compound CC(C)CC1=CC=C(C(C)C(O)=O)C=C1 HEFNNWSXXWATRW-UHFFFAOYSA-N 0.000 description 1
- ZRVUJXDFFKFLMG-UHFFFAOYSA-N Ilacox Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC1=NC=C(C)S1 ZRVUJXDFFKFLMG-UHFFFAOYSA-N 0.000 description 1
- 208000009326 Ileitis Diseases 0.000 description 1
- XMXHEBAFVSFQEX-UHFFFAOYSA-N Iloperidone Chemical compound COC1=CC(C(C)=O)=CC=C1OCCCN1CCC(C=2C3=CC=C(F)C=C3ON=2)CC1 XMXHEBAFVSFQEX-UHFFFAOYSA-N 0.000 description 1
- 229960000905 Indomethacin Drugs 0.000 description 1
- 206010061216 Infarction Diseases 0.000 description 1
- 229960000367 Inositol Drugs 0.000 description 1
- CDAISMWEOUEBRE-GPIVLXJGSA-N Inositol Chemical compound O[C@H]1[C@H](O)[C@@H](O)[C@H](O)[C@H](O)[C@@H]1O CDAISMWEOUEBRE-GPIVLXJGSA-N 0.000 description 1
- 229960001888 Ipratropium Drugs 0.000 description 1
- 206010061255 Ischaemia Diseases 0.000 description 1
- VFQXVTODMYMSMJ-UHFFFAOYSA-N Isonicotinamide Chemical compound NC(=O)C1=CC=NC=C1 VFQXVTODMYMSMJ-UHFFFAOYSA-N 0.000 description 1
- PWWVAXIEGOYWEE-UHFFFAOYSA-N Isophenergan Chemical compound C1=CC=C2N(CC(C)N(C)C)C3=CC=CC=C3SC2=C1 PWWVAXIEGOYWEE-UHFFFAOYSA-N 0.000 description 1
- 229950001646 Ispronicline Drugs 0.000 description 1
- 241000229754 Iva xanthiifolia Species 0.000 description 1
- DKYWVDODHFEZIM-UHFFFAOYSA-N Ketoprofen Chemical compound OC(=O)C(C)C1=CC=CC(C(=O)C=2C=CC=CC=2)=C1 DKYWVDODHFEZIM-UHFFFAOYSA-N 0.000 description 1
- OZWKMVRBQXNZKK-UHFFFAOYSA-N Ketorolac Chemical compound OC(=O)C1CCN2C1=CC=C2C(=O)C1=CC=CC=C1 OZWKMVRBQXNZKK-UHFFFAOYSA-N 0.000 description 1
- 210000003734 Kidney Anatomy 0.000 description 1
- 235000019766 L-Lysine Nutrition 0.000 description 1
- SFLSHLFXELFNJZ-MRVPVSSYSA-N L-Noradrenaline Natural products NC[C@@H](O)C1=CC=C(O)C(O)=C1 SFLSHLFXELFNJZ-MRVPVSSYSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 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
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- 229950005286 LANEPITANT Drugs 0.000 description 1
- PYZRQGJRPPTADH-UHFFFAOYSA-N Lamotrigine Chemical compound NC1=NC(N)=NN=C1C1=CC=CC(Cl)=C1Cl PYZRQGJRPPTADH-UHFFFAOYSA-N 0.000 description 1
- 210000002414 Leg Anatomy 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- OZYUPQUCAUTOBP-QXAKKESOSA-N Levallorphan Chemical compound C([C@H]12)CCC[C@@]11CCN(CC=C)[C@@H]2CC2=CC=C(O)C=C21 OZYUPQUCAUTOBP-QXAKKESOSA-N 0.000 description 1
- 206010024453 Ligament sprain Diseases 0.000 description 1
- 239000000867 Lipoxygenase Inhibitor Substances 0.000 description 1
- YNESATAKKCNGOF-UHFFFAOYSA-N Lithium bis(trimethylsilyl)amide Chemical compound [Li+].C[Si](C)(C)[N-][Si](C)(C)C YNESATAKKCNGOF-UHFFFAOYSA-N 0.000 description 1
- 239000012448 Lithium borohydride Substances 0.000 description 1
- MJJDYOLPMGIWND-UHFFFAOYSA-N Litoxetine Chemical compound C=1C=C2C=CC=CC2=CC=1COC1CCNCC1 MJJDYOLPMGIWND-UHFFFAOYSA-N 0.000 description 1
- 229950004138 Litoxetine Drugs 0.000 description 1
- 210000004446 Longitudinal Ligaments Anatomy 0.000 description 1
- DIWRORZWFLOCLC-UHFFFAOYSA-N Lorazepam Chemical compound C12=CC(Cl)=CC=C2NC(=O)C(O)N=C1C1=CC=CC=C1Cl DIWRORZWFLOCLC-UHFFFAOYSA-N 0.000 description 1
- 208000008930 Low Back Pain Diseases 0.000 description 1
- PQXKDMSYBGKCJA-CVTJIBDQSA-N Lurasidone Chemical compound C1=CC=C2C(N3CCN(CC3)C[C@@H]3CCCC[C@H]3CN3C(=O)[C@@H]4[C@H]5CC[C@H](C5)[C@@H]4C3=O)=NSC2=C1 PQXKDMSYBGKCJA-CVTJIBDQSA-N 0.000 description 1
- 210000003563 Lymphoid Tissue Anatomy 0.000 description 1
- 206010025312 Lymphoma AIDS related Diseases 0.000 description 1
- 206010064912 Malignant transformation Diseases 0.000 description 1
- 208000003393 Mammary Paget's Disease Diseases 0.000 description 1
- QSLMDECMDJKHMQ-GSXCWMCISA-N Maprotiline Chemical compound C12=CC=CC=C2[C@@]2(CCCNC)C3=CC=CC=C3[C@@H]1CC2 QSLMDECMDJKHMQ-GSXCWMCISA-N 0.000 description 1
- 230000036676 Maximum tolerable dose Effects 0.000 description 1
- BCVXHSPFUWZLGQ-UHFFFAOYSA-N MeCN acetonitrile Chemical compound CC#N.CC#N BCVXHSPFUWZLGQ-UHFFFAOYSA-N 0.000 description 1
- COTNUBDHGSIOTA-UHFFFAOYSA-N MeOH methanol Chemical compound OC.OC COTNUBDHGSIOTA-UHFFFAOYSA-N 0.000 description 1
- BZLVMXJERCGZMT-UHFFFAOYSA-N MeOtBu Chemical compound COC(C)(C)C BZLVMXJERCGZMT-UHFFFAOYSA-N 0.000 description 1
- DYLJVOXRWLXDIG-UHFFFAOYSA-N Meclinertant Chemical compound COC1=CC=CC(OC)=C1C1=CC(C(=O)NC2(C3CC4CC(C3)CC2C4)C(O)=O)=NN1C1=CC=NC2=CC(Cl)=CC=C12 DYLJVOXRWLXDIG-UHFFFAOYSA-N 0.000 description 1
- 229950002178 Meclinertant Drugs 0.000 description 1
- SBDNJUWAMKYJOX-UHFFFAOYSA-N Meclofenamic Acid Chemical compound CC1=CC=C(Cl)C(NC=2C(=CC=CC=2)C(O)=O)=C1Cl SBDNJUWAMKYJOX-UHFFFAOYSA-N 0.000 description 1
- HYYBABOKPJLUIN-UHFFFAOYSA-N Mefenamic acid Chemical compound CC1=CC=CC(NC=2C(=CC=CC=2)C(O)=O)=C1C HYYBABOKPJLUIN-UHFFFAOYSA-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
- 229960003194 Meglumine Drugs 0.000 description 1
- BUGYDGFZZOZRHP-UHFFFAOYSA-N Memantine Chemical compound C1C(C2)CC3(C)CC1(C)CC2(N)C3 BUGYDGFZZOZRHP-UHFFFAOYSA-N 0.000 description 1
- 229940041616 Menthol Drugs 0.000 description 1
- 229940041655 Meperidine Drugs 0.000 description 1
- 229940041659 Mephobarbital Drugs 0.000 description 1
- NPPQSCRMBWNHMW-UHFFFAOYSA-N Meprobamate Chemical compound NC(=O)OCC(C)(CCC)COC(N)=O NPPQSCRMBWNHMW-UHFFFAOYSA-N 0.000 description 1
- 229960004815 Meprobamate Drugs 0.000 description 1
- YECBIJXISLIIDS-UHFFFAOYSA-N Mepyramine Chemical compound C1=CC(OC)=CC=C1CN(CCN(C)C)C1=CC=CC=N1 YECBIJXISLIIDS-UHFFFAOYSA-N 0.000 description 1
- 206010061289 Metastatic neoplasm Diseases 0.000 description 1
- USSIQXCVUWKGNF-UHFFFAOYSA-N Methadone Chemical compound C=1C=CC=CC=1C(CC(C)N(C)C)(C(=O)CC)C1=CC=CC=C1 USSIQXCVUWKGNF-UHFFFAOYSA-N 0.000 description 1
- 229960002803 Methaqualone Drugs 0.000 description 1
- 229960002057 Metharbital Drugs 0.000 description 1
- FWJKNZONDWOGMI-UHFFFAOYSA-N Metharbital Chemical compound CCC1(CC)C(=O)NC(=O)N(C)C1=O FWJKNZONDWOGMI-UHFFFAOYSA-N 0.000 description 1
- GNXFOGHNGIVQEH-UHFFFAOYSA-N Methocarbamol Chemical compound COC1=CC=CC=C1OCC(O)COC(N)=O GNXFOGHNGIVQEH-UHFFFAOYSA-N 0.000 description 1
- ALARQZQTBTVLJV-UHFFFAOYSA-N Methylphenobarbital Chemical compound C=1C=CC=CC=1C1(CC)C(=O)NC(=O)N(C)C1=O ALARQZQTBTVLJV-UHFFFAOYSA-N 0.000 description 1
- NZXKDOXHBHYTKP-UHFFFAOYSA-N Metohexital Chemical compound CCC#CC(C)C1(CC=C)C(=O)NC(=O)N(C)C1=O NZXKDOXHBHYTKP-UHFFFAOYSA-N 0.000 description 1
- VLPIATFUUWWMKC-UHFFFAOYSA-N Mexiletine Chemical compound CC(N)COC1=C(C)C=CC=C1C VLPIATFUUWWMKC-UHFFFAOYSA-N 0.000 description 1
- 229960003955 Mianserin Drugs 0.000 description 1
- 206010027599 Migraine Diseases 0.000 description 1
- 208000008085 Migraine Disorders Diseases 0.000 description 1
- 208000000060 Migraine with Aura Diseases 0.000 description 1
- 206010052787 Migraine without aura Diseases 0.000 description 1
- 229960000600 Milnacipran Drugs 0.000 description 1
- GJJFMKBJSRMPLA-HIFRSBDPSA-N Milnacipran Chemical compound C=1C=CC=CC=1[C@@]1(C(=O)N(CC)CC)C[C@@H]1CN GJJFMKBJSRMPLA-HIFRSBDPSA-N 0.000 description 1
- RONZAEMNMFQXRA-UHFFFAOYSA-N Mirtazapine Chemical compound C1C2=CC=CN=C2N2CCN(C)CC2C2=CC=CC=C21 RONZAEMNMFQXRA-UHFFFAOYSA-N 0.000 description 1
- 229960001785 Mirtazapine Drugs 0.000 description 1
- YFGHCGITMMYXAQ-UHFFFAOYSA-N Modafinil Chemical compound C=1C=CC=CC=1C(S(=O)CC(=O)N)C1=CC=CC=C1 YFGHCGITMMYXAQ-UHFFFAOYSA-N 0.000 description 1
- 241000907681 Morpho Species 0.000 description 1
- 210000000214 Mouth Anatomy 0.000 description 1
- 206010048592 Musculoskeletal disease Diseases 0.000 description 1
- 206010028391 Musculoskeletal pain Diseases 0.000 description 1
- 208000010125 Myocardial Infarction Diseases 0.000 description 1
- ZSXGLVDWWRXATF-UHFFFAOYSA-N N,N-dimethylformamide dimethyl acetal Chemical compound COC(OC)N(C)C ZSXGLVDWWRXATF-UHFFFAOYSA-N 0.000 description 1
- PEECTLLHENGOKU-UHFFFAOYSA-N N,N-dimethylpyridin-4-amine Chemical compound CN(C)C1=CC=NC=C1.CN(C)C1=CC=NC=C1 PEECTLLHENGOKU-UHFFFAOYSA-N 0.000 description 1
- ZZVGLDBDDYESAB-UHFFFAOYSA-N N-(4-(2-((3-Chlorophenylmethyl)Amino)Ethyl)Phenyl)-2-Thiophecarboxamidine Chemical compound ClC1=CC=CC(CNCCC=2C=CC(NC(=N)C=3SC=CC=3)=CC=2)=C1 ZZVGLDBDDYESAB-UHFFFAOYSA-N 0.000 description 1
- WHNWPMSKXPGLAX-UHFFFAOYSA-N N-Vinylpyrrolidone Chemical compound C=CN1CCCC1=O WHNWPMSKXPGLAX-UHFFFAOYSA-N 0.000 description 1
- CVXJAPZTZWLRBP-MUUNZHRXSA-N N-[(2R)-1-[acetyl-[(2-methoxyphenyl)methyl]amino]-3-(1H-indol-3-yl)propan-2-yl]-2-(4-piperidin-1-ylpiperidin-1-yl)acetamide Chemical compound COC1=CC=CC=C1CN(C(C)=O)C[C@H](NC(=O)CN1CCC(CC1)N1CCCCC1)CC1=CNC2=CC=CC=C12 CVXJAPZTZWLRBP-MUUNZHRXSA-N 0.000 description 1
- HVUYHTDPDMCSGV-UHFFFAOYSA-N N-[1-(4-amino-6,7-dimethoxy-5-pyridin-2-ylquinazolin-2-yl)-1,2,3,4-tetrahydroisoquinolin-5-yl]methanesulfonamide Chemical compound COC=1C(OC)=CC2=NC(C3C4=CC=CC(NS(C)(=O)=O)=C4CCN3)=NC(N)=C2C=1C1=CC=CC=N1 HVUYHTDPDMCSGV-UHFFFAOYSA-N 0.000 description 1
- FTNFEHXDETWERN-UHFFFAOYSA-N N-[bis(aziridin-1-yl)phosphoryl]-2,6-dimethyl-5-[(4-propan-2-yloxyphenyl)methyl]pyrimidin-4-amine Chemical compound C1=CC(OC(C)C)=CC=C1CC1=C(C)N=C(C)N=C1NP(=O)(N1CC1)N1CC1 FTNFEHXDETWERN-UHFFFAOYSA-N 0.000 description 1
- XHXVAJHZTIXQQD-UHFFFAOYSA-N N-[bis(aziridin-1-yl)phosphoryl]-5-[(4-butoxyphenyl)methyl]-2,6-dimethylpyrimidin-4-amine Chemical compound C1=CC(OCCCC)=CC=C1CC1=C(C)N=C(C)N=C1NP(=O)(N1CC1)N1CC1 XHXVAJHZTIXQQD-UHFFFAOYSA-N 0.000 description 1
- 150000001200 N-acyl ethanolamides Chemical class 0.000 description 1
- PSACHCMMPFMFAJ-UHFFFAOYSA-N NMM N-methylmorpholine Chemical compound CN1CCOCC1.CN1CCOCC1 PSACHCMMPFMFAJ-UHFFFAOYSA-N 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 101700079360 NTF4 Proteins 0.000 description 1
- 102100016138 NTF4 Human genes 0.000 description 1
- NETZHAKZCGBWSS-CEDHKZHLSA-N Nalbuphine Chemical compound C([C@]12[C@H]3OC=4C(O)=CC=C(C2=4)C[C@@H]2[C@]1(O)CC[C@@H]3O)CN2CC1CCC1 NETZHAKZCGBWSS-CEDHKZHLSA-N 0.000 description 1
- 229960000805 Nalbuphine Drugs 0.000 description 1
- WJBLNOPPDWQMCH-MBPVOVBZSA-N Nalmefene Chemical compound N1([C@@H]2CC3=CC=C(C=4O[C@@H]5[C@](C3=4)([C@]2(CCC5=C)O)CC1)O)CC1CC1 WJBLNOPPDWQMCH-MBPVOVBZSA-N 0.000 description 1
- UZHSEJADLWPNLE-GRGSLBFTSA-N Naloxone Chemical compound O=C([C@@H]1O2)CC[C@@]3(O)[C@H]4CC5=CC=C(O)C2=C5[C@@]13CCN4CC=C UZHSEJADLWPNLE-GRGSLBFTSA-N 0.000 description 1
- 229960004127 Naloxone Drugs 0.000 description 1
- DQCKKXVULJGBQN-XFWGSAIBSA-N Naltrexone Chemical compound N1([C@@H]2CC3=CC=C(C=4O[C@@H]5[C@](C3=4)([C@]2(CCC5=O)O)CC1)O)CC1CC1 DQCKKXVULJGBQN-XFWGSAIBSA-N 0.000 description 1
- 229960003086 Naltrexone Drugs 0.000 description 1
- UNHGSHHVDNGCFN-UHFFFAOYSA-N Naratriptan Chemical compound C=12[CH]C(CCS(=O)(=O)NC)=CC=C2N=CC=1C1CCN(C)CC1 UNHGSHHVDNGCFN-UHFFFAOYSA-N 0.000 description 1
- VRBKIVRKKCLPHA-UHFFFAOYSA-N Nefazodone Chemical compound O=C1N(CCOC=2C=CC=CC=2)C(CC)=NN1CCCN(CC1)CCN1C1=CC=CC(Cl)=C1 VRBKIVRKKCLPHA-UHFFFAOYSA-N 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- OGZQTTHDGQBLBT-UHFFFAOYSA-N Neramexane Chemical compound CC1(C)CC(C)(C)CC(C)(N)C1 OGZQTTHDGQBLBT-UHFFFAOYSA-N 0.000 description 1
- 229950004543 Neramexane Drugs 0.000 description 1
- 208000001738 Nervous System Trauma Diseases 0.000 description 1
- 206010029331 Neuropathy peripheral Diseases 0.000 description 1
- SNICXCGAKADSCV-JTQLQIEISA-N Nicotine Chemical compound CN1CCC[C@H]1C1=CC=CN=C1 SNICXCGAKADSCV-JTQLQIEISA-N 0.000 description 1
- 229960002715 Nicotine Drugs 0.000 description 1
- HYWYRSMBCFDLJT-UHFFFAOYSA-N Nimesulide Chemical compound CS(=O)(=O)NC1=CC=C([N+]([O-])=O)C=C1OC1=CC=CC=C1 HYWYRSMBCFDLJT-UHFFFAOYSA-N 0.000 description 1
- XITQUSLLOSKDTB-UHFFFAOYSA-N Nitrofen Chemical compound C1=CC([N+](=O)[O-])=CC=C1OC1=CC=C(Cl)C=C1Cl XITQUSLLOSKDTB-UHFFFAOYSA-N 0.000 description 1
- ODUCDPQEXGNKDN-UHFFFAOYSA-N Nitroxyl Chemical compound O=N ODUCDPQEXGNKDN-UHFFFAOYSA-N 0.000 description 1
- 229960001073 Nomifensine Drugs 0.000 description 1
- XXPANQJNYNUNES-UHFFFAOYSA-N Nomifensine Chemical compound C12=CC=CC(N)=C2CN(C)CC1C1=CC=CC=C1 XXPANQJNYNUNES-UHFFFAOYSA-N 0.000 description 1
- 229960001158 Nortriptyline Drugs 0.000 description 1
- PHVGLTMQBUFIQQ-UHFFFAOYSA-N Nortryptiline Chemical compound C1CC2=CC=CC=C2C(=CCCNC)C2=CC=CC=C21 PHVGLTMQBUFIQQ-UHFFFAOYSA-N 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N OC(=O)C(N)CCCNC(N)=N Chemical compound OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- DBGIVFWFUFKIQN-UHFFFAOYSA-N Obedrex Chemical compound CCNC(C)CC1=CC=CC(C(F)(F)F)=C1 DBGIVFWFUFKIQN-UHFFFAOYSA-N 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 229950004864 Olamine Drugs 0.000 description 1
- KVWDHTXUZHCGIO-UHFFFAOYSA-N Olanzapine Chemical compound C1CN(C)CCN1C1=NC2=CC=CC=C2NC2=C1C=C(C)S2 KVWDHTXUZHCGIO-UHFFFAOYSA-N 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- FELGMEQIXOGIFQ-UHFFFAOYSA-N Ondansetron Chemical compound CC1=NC=CN1CC1C(=O)C(C=2C(=CC=CC=2)N2C)=C2CC1 FELGMEQIXOGIFQ-UHFFFAOYSA-N 0.000 description 1
- 229960005343 Ondansetron Drugs 0.000 description 1
- 230000035536 Oral bioavailability Effects 0.000 description 1
- DZOJBGLFWINFBF-UMSFTDKQSA-N Osanetant Chemical compound C([C@](C1)(CCCN2CCC(CC2)(N(C(C)=O)C)C=2C=CC=CC=2)C=2C=C(Cl)C(Cl)=CC=2)CCN1C(=O)C1=CC=CC=C1 DZOJBGLFWINFBF-UMSFTDKQSA-N 0.000 description 1
- 229950009875 Osanetant Drugs 0.000 description 1
- 208000010191 Osteitis Deformans Diseases 0.000 description 1
- FDXQKWSTUZCCTM-UHFFFAOYSA-N Oxaprotiline Chemical compound C12=CC=CC=C2C2(CC(O)CNC)C3=CC=CC=C3C1CC2 FDXQKWSTUZCCTM-UHFFFAOYSA-N 0.000 description 1
- OFPXSFXSNFPTHF-UHFFFAOYSA-N Oxaprozin Chemical compound O1C(CCC(=O)O)=NC(C=2C=CC=CC=2)=C1C1=CC=CC=C1 OFPXSFXSNFPTHF-UHFFFAOYSA-N 0.000 description 1
- ADIMAYPTOBDMTL-UHFFFAOYSA-N Oxazepam Chemical compound C12=CC(Cl)=CC=C2NC(=O)C(O)N=C1C1=CC=CC=C1 ADIMAYPTOBDMTL-UHFFFAOYSA-N 0.000 description 1
- UQCNKQCJZOAFTQ-ISWURRPUSA-N Oxymorphone Chemical compound O([C@H]1C(CC[C@]23O)=O)C4=C5[C@@]12CCN(C)[C@@H]3CC5=CC=C4O UQCNKQCJZOAFTQ-ISWURRPUSA-N 0.000 description 1
- 101710040930 PTGS2 Proteins 0.000 description 1
- 102100015381 PTGS2 Human genes 0.000 description 1
- 206010033645 Pancreatitis Diseases 0.000 description 1
- 206010033647 Pancreatitis acute Diseases 0.000 description 1
- 206010033661 Pancytopenia Diseases 0.000 description 1
- 206010033701 Papillary thyroid cancer Diseases 0.000 description 1
- 206010061536 Parkinson's disease Diseases 0.000 description 1
- 229910002666 PdCl2 Inorganic materials 0.000 description 1
- IZUPBVBPLAPZRR-UHFFFAOYSA-N Pentachlorophenol Chemical compound OC1=C(Cl)C(Cl)=C(Cl)C(Cl)=C1Cl IZUPBVBPLAPZRR-UHFFFAOYSA-N 0.000 description 1
- 229960001412 Pentobarbital Drugs 0.000 description 1
- WEXRUCMBJFQVBZ-UHFFFAOYSA-N Pentobarbital Chemical compound CCCC(C)C1(CC)C(=O)NC(=O)NC1=O WEXRUCMBJFQVBZ-UHFFFAOYSA-N 0.000 description 1
- 206010048745 Periarticular disease Diseases 0.000 description 1
- 208000008494 Pericarditis Diseases 0.000 description 1
- 210000000578 Peripheral Nerves Anatomy 0.000 description 1
- GTAIPSDXDDTGBZ-OYRHEFFESA-N Perospirone Chemical compound C1=CC=C2C(N3CCN(CC3)CCCCN3C(=O)[C@@H]4CCCC[C@@H]4C3=O)=NSCC2=C1 GTAIPSDXDDTGBZ-OYRHEFFESA-N 0.000 description 1
- 229950004193 Perospirone Drugs 0.000 description 1
- 229960000762 Perphenazine Drugs 0.000 description 1
- BDABGOLMYNHHTR-UHFFFAOYSA-N Perzinfotel Chemical compound OP(O)(=O)CCN1CCCNC2=C1C(=O)C2=O BDABGOLMYNHHTR-UHFFFAOYSA-N 0.000 description 1
- 229950006454 Perzinfotel Drugs 0.000 description 1
- XADCESSVHJOZHK-UHFFFAOYSA-N Petidina Chemical compound C=1C=CC=CC=1C1(C(=O)OCC)CCN(C)CC1 XADCESSVHJOZHK-UHFFFAOYSA-N 0.000 description 1
- 239000004264 Petrolatum Substances 0.000 description 1
- 229940066842 Petrolatum Drugs 0.000 description 1
- 206010056238 Phantom pain Diseases 0.000 description 1
- 229960002895 Phenylbutazone Drugs 0.000 description 1
- 229940067631 Phospholipids Drugs 0.000 description 1
- QYSPLQLAKJAUJT-UHFFFAOYSA-N Piroxicam Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC1=CC=CC=N1 QYSPLQLAKJAUJT-UHFFFAOYSA-N 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 229920002732 Polyanhydride Polymers 0.000 description 1
- 229920000954 Polyglycolide Polymers 0.000 description 1
- 208000005987 Polymyositis Diseases 0.000 description 1
- 229920001710 Polyorthoester Polymers 0.000 description 1
- 229940068968 Polysorbate 80 Drugs 0.000 description 1
- 239000004372 Polyvinyl alcohol Substances 0.000 description 1
- 206010065016 Post-traumatic pain Diseases 0.000 description 1
- CHKVPAROMQMJNQ-UHFFFAOYSA-M Potassium bisulfate Chemical compound [K+].OS([O-])(=O)=O CHKVPAROMQMJNQ-UHFFFAOYSA-M 0.000 description 1
- OKBMCNHOEMXPTM-UHFFFAOYSA-M Potassium peroxymonosulfate Chemical compound [K+].OOS([O-])(=O)=O OKBMCNHOEMXPTM-UHFFFAOYSA-M 0.000 description 1
- QPCVHQBVMYCJOM-UHFFFAOYSA-N Propiverine Chemical compound C=1C=CC=CC=1C(C=1C=CC=CC=1)(OCCC)C(=O)OC1CCN(C)CC1 QPCVHQBVMYCJOM-UHFFFAOYSA-N 0.000 description 1
- 229940069956 Propoxyphene Drugs 0.000 description 1
- AQHHHDLHHXJYJD-UHFFFAOYSA-N Proprasylyt Chemical compound C1=CC=C2C(OCC(O)CNC(C)C)=CC=CC2=C1 AQHHHDLHHXJYJD-UHFFFAOYSA-N 0.000 description 1
- 102000004226 Prostaglandin-E Synthases Human genes 0.000 description 1
- 108090000748 Prostaglandin-E Synthases Proteins 0.000 description 1
- 102000014961 Protein Precursors Human genes 0.000 description 1
- 108010078762 Protein Precursors Proteins 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 229940079863 Pyrilamine Drugs 0.000 description 1
- URKOMYMAXPYINW-UHFFFAOYSA-N Quetiapine Chemical compound C1CN(CCOCCO)CCN1C1=NC2=CC=CC=C2SC2=CC=CC=C12 URKOMYMAXPYINW-UHFFFAOYSA-N 0.000 description 1
- 229960000948 Quinine Drugs 0.000 description 1
- 239000007868 Raney catalyst Substances 0.000 description 1
- 229910000564 Raney nickel Inorganic materials 0.000 description 1
- 206010037912 Raynaud's phenomenon Diseases 0.000 description 1
- 108091005674 Receptor kinase Proteins 0.000 description 1
- 206010038038 Rectal cancer Diseases 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 206010038419 Renal colic Diseases 0.000 description 1
- 206010072736 Rheumatic disease Diseases 0.000 description 1
- JZCPYUJPEARBJL-UHFFFAOYSA-N Rimonabant Chemical compound CC=1C(C(=O)NN2CCCCC2)=NN(C=2C(=CC(Cl)=CC=2)Cl)C=1C1=CC=C(Cl)C=C1 JZCPYUJPEARBJL-UHFFFAOYSA-N 0.000 description 1
- 229960003015 Rimonabant Drugs 0.000 description 1
- JUOSGGQXEBBCJB-GORDUTHDSA-N Rivanicline Chemical compound CNCC\C=C\C1=CC=CN=C1 JUOSGGQXEBBCJB-GORDUTHDSA-N 0.000 description 1
- TXHZXHICDBAVJW-UHFFFAOYSA-N Rizatriptan Chemical compound C=1[C]2C(CCN(C)C)=CN=C2C=CC=1CN1C=NC=N1 TXHZXHICDBAVJW-UHFFFAOYSA-N 0.000 description 1
- RZJQGNCSTQAWON-UHFFFAOYSA-N Rofecoxib Chemical compound C1=CC(S(=O)(=O)C)=CC=C1C1=C(C=2C=CC=CC=2)C(=O)OC1 RZJQGNCSTQAWON-UHFFFAOYSA-N 0.000 description 1
- 102100010564 SCN10A Human genes 0.000 description 1
- 101710032460 SCN10A Proteins 0.000 description 1
- 102100020052 SCN3A Human genes 0.000 description 1
- 101700035796 SCN3A Proteins 0.000 description 1
- 102100010563 SCN9A Human genes 0.000 description 1
- 101700051996 SCN9A Proteins 0.000 description 1
- 101710042981 SHMT1 Proteins 0.000 description 1
- 229940081974 Saccharin Drugs 0.000 description 1
- CVHZOJJKTDOEJC-UHFFFAOYSA-N Saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 1
- 229940085605 Saccharin Sodium Drugs 0.000 description 1
- WINXNKPZLFISPD-UHFFFAOYSA-M Saccharin sodium Chemical compound [Na+].C1=CC=C2C(=O)[N-]S(=O)(=O)C2=C1 WINXNKPZLFISPD-UHFFFAOYSA-M 0.000 description 1
- SKZKKFZAGNVIMN-UHFFFAOYSA-N Salicilamide Chemical compound NC(=O)C1=CC=CC=C1O SKZKKFZAGNVIMN-UHFFFAOYSA-N 0.000 description 1
- HKFMQJUJWSFOLY-OAQYLSRUSA-N Sarizotan Chemical compound C1=CC(F)=CC=C1C1=CN=CC(CNC[C@@H]2OC3=CC=CC=C3CC2)=C1 HKFMQJUJWSFOLY-OAQYLSRUSA-N 0.000 description 1
- 229950007903 Sarizotan Drugs 0.000 description 1
- 229950008243 Secbutabarbital Drugs 0.000 description 1
- 208000005154 Secretory breast carcinoma Diseases 0.000 description 1
- GZKLJWGUPQBVJQ-UHFFFAOYSA-N Sertindole Chemical compound C1=CC(F)=CC=C1N1C2=CC=C(Cl)C=C2C(C2CCN(CCN3C(NCC3)=O)CC2)=C1 GZKLJWGUPQBVJQ-UHFFFAOYSA-N 0.000 description 1
- 229960002073 Sertraline Drugs 0.000 description 1
- BNRNXUUZRGQAQC-UHFFFAOYSA-N Sildenafil Chemical compound CCCC1=NN(C)C(C(N2)=O)=C1N=C2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(C)CC1 BNRNXUUZRGQAQC-UHFFFAOYSA-N 0.000 description 1
- 210000003491 Skin Anatomy 0.000 description 1
- 102000018674 Sodium Channels Human genes 0.000 description 1
- 108010052164 Sodium Channels Proteins 0.000 description 1
- 229940005550 Sodium alginate Drugs 0.000 description 1
- WBHQBSYUUJJSRZ-UHFFFAOYSA-M Sodium bisulfate Chemical compound [Na+].OS([O-])(=O)=O WBHQBSYUUJJSRZ-UHFFFAOYSA-M 0.000 description 1
- 239000004141 Sodium laurylsulphate Substances 0.000 description 1
- AKHNMLFCWUSKQB-UHFFFAOYSA-L Sodium thiosulphate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=S AKHNMLFCWUSKQB-UHFFFAOYSA-L 0.000 description 1
- FBOUYBDGKBSUES-VXKWHMMOSA-N Solifenacin Chemical compound C1([C@H]2C3=CC=CC=C3CCN2C(O[C@@H]2C3CCN(CC3)C2)=O)=CC=CC=C1 FBOUYBDGKBSUES-VXKWHMMOSA-N 0.000 description 1
- JEYCTXHKTXCGPB-UHFFFAOYSA-N Somnomed Chemical compound CC1=CC=CC=C1N1C(=O)C2=CC=CC=C2N=C1C JEYCTXHKTXCGPB-UHFFFAOYSA-N 0.000 description 1
- WNUQCGWXPNGORO-NRFANRHFSA-N Sonepiprazole Chemical compound C1=CC(S(=O)(=O)N)=CC=C1N1CCN(CC[C@H]2C3=CC=CC=C3CCO2)CC1 WNUQCGWXPNGORO-NRFANRHFSA-N 0.000 description 1
- 229950001013 Sonepiprazole Drugs 0.000 description 1
- 239000004147 Sorbitan trioleate Substances 0.000 description 1
- 229960000391 Sorbitan trioleate Drugs 0.000 description 1
- 210000000952 Spleen Anatomy 0.000 description 1
- 208000006045 Spondylarthropathy Diseases 0.000 description 1
- 206010052775 Spondyloarthropathy Diseases 0.000 description 1
- 229960001940 Sulfasalazine Drugs 0.000 description 1
- MLKXDPUZXIRXEP-MFOYZWKCSA-N Sulindac Chemical compound CC1=C(CC(O)=O)C2=CC(F)=CC=C2\C1=C/C1=CC=C(S(C)=O)C=C1 MLKXDPUZXIRXEP-MFOYZWKCSA-N 0.000 description 1
- 229960000894 Sulindac Drugs 0.000 description 1
- 239000005864 Sulphur Substances 0.000 description 1
- 210000004243 Sweat Anatomy 0.000 description 1
- 210000000106 Sweat Glands Anatomy 0.000 description 1
- 108060008443 TPPP Proteins 0.000 description 1
- IEHKWSGCTWLXFU-IIBYNOLFSA-N Tadalafil Chemical compound C1=C2OCOC2=CC([C@@H]2C3=C([C]4C=CC=CC4=N3)C[C@H]3N2C(=O)CN(C3=O)C)=C1 IEHKWSGCTWLXFU-IIBYNOLFSA-N 0.000 description 1
- DRHKJLXJIQTDTD-OAHLLOKOSA-N Tamsulosine Chemical compound CCOC1=CC=CC=C1OCCN[C@H](C)CC1=CC=C(OC)C(S(N)(=O)=O)=C1 DRHKJLXJIQTDTD-OAHLLOKOSA-N 0.000 description 1
- 229950008160 Tanezumab Drugs 0.000 description 1
- MKTAGSRKQIGEBH-SSDOTTSWSA-N Tebanicline Chemical compound C1=NC(Cl)=CC=C1OC[C@@H]1NCC1 MKTAGSRKQIGEBH-SSDOTTSWSA-N 0.000 description 1
- 229960003188 Temazepam Drugs 0.000 description 1
- SEQDDYPDSLOBDC-UHFFFAOYSA-N Temazepam Chemical compound N=1C(O)C(=O)N(C)C2=CC=C(Cl)C=C2C=1C1=CC=CC=C1 SEQDDYPDSLOBDC-UHFFFAOYSA-N 0.000 description 1
- 229950000334 Temiverine Drugs 0.000 description 1
- 208000008548 Tension-Type Headache Diseases 0.000 description 1
- YBRBMKDOPFTVDT-UHFFFAOYSA-N Tert-Butylamine Chemical compound CC(C)(C)N YBRBMKDOPFTVDT-UHFFFAOYSA-N 0.000 description 1
- 229960003279 Thiopental Drugs 0.000 description 1
- 208000009205 Tinnitus Diseases 0.000 description 1
- 230000036335 Tissue distribution Effects 0.000 description 1
- UPSPUYADGBWSHF-UHFFFAOYSA-N Tolmetin Chemical compound C1=CC(C)=CC=C1C(=O)C1=CC=C(CC(O)=O)N1C UPSPUYADGBWSHF-UHFFFAOYSA-N 0.000 description 1
- UEQUQVLFIPOEMF-UHFFFAOYSA-N Tolvon Chemical compound C1C2=CC=CC=C2N2CCN(C)CC2C2=CC=CC=C21 UEQUQVLFIPOEMF-UHFFFAOYSA-N 0.000 description 1
- KJADKKWYZYXHBB-XBWDGYHZSA-N Topiramic acid Chemical compound C1O[C@@]2(COS(N)(=O)=O)OC(C)(C)O[C@H]2[C@@H]2OC(C)(C)O[C@@H]21 KJADKKWYZYXHBB-XBWDGYHZSA-N 0.000 description 1
- TVYLLZQTGLZFBW-ZBFHGGJFSA-N Tramadol Chemical compound COC1=CC=CC([C@]2(O)[C@H](CCCC2)CN(C)C)=C1 TVYLLZQTGLZFBW-ZBFHGGJFSA-N 0.000 description 1
- 229960004380 Tramadol Drugs 0.000 description 1
- RGCVKNLCSQQDEP-UHFFFAOYSA-N Tranquisan Chemical compound C1CN(CCO)CCN1CCCN1C2=CC(Cl)=CC=C2SC2=CC=CC=C21 RGCVKNLCSQQDEP-UHFFFAOYSA-N 0.000 description 1
- 229950005135 Traxoprodil Drugs 0.000 description 1
- PHLBKPHSAVXXEF-UHFFFAOYSA-N Trazodone Chemical compound ClC1=CC=CC(N2CCN(CCCN3C(N4C=CC=CC4=N3)=O)CC2)=C1 PHLBKPHSAVXXEF-UHFFFAOYSA-N 0.000 description 1
- 229960003991 Trazodone Drugs 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N Trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 1
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-N 0.000 description 1
- ZEWQUBUPAILYHI-UHFFFAOYSA-N Trifluoperazine Chemical compound C1CN(C)CCN1CCCN1C2=CC(C(F)(F)F)=CC=C2SC2=CC=CC=C21 ZEWQUBUPAILYHI-UHFFFAOYSA-N 0.000 description 1
- 229960002324 Trifluoperazine Drugs 0.000 description 1
- DTQVDTLACAAQTR-UHFFFAOYSA-M Trifluoroacetate Chemical class [O-]C(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-M 0.000 description 1
- ITMCEJHCFYSIIV-UHFFFAOYSA-N Trifluoromethanesulfonic acid Chemical compound OS(=O)(=O)C(F)(F)F ITMCEJHCFYSIIV-UHFFFAOYSA-N 0.000 description 1
- 206010044652 Trigeminal neuralgia Diseases 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 229940035504 Tromethamine Drugs 0.000 description 1
- 208000009852 Uremia Diseases 0.000 description 1
- 206010046431 Urethral cancer Diseases 0.000 description 1
- 229940102566 Valproate Drugs 0.000 description 1
- YWPHCCPCQOJSGZ-UHFFFAOYSA-N Viloxazine Chemical compound CCOC1=CC=CC=C1OCC1OCCNC1 YWPHCCPCQOJSGZ-UHFFFAOYSA-N 0.000 description 1
- 206010047627 Vitamin deficiency Diseases 0.000 description 1
- MVWVFYHBGMAFLY-UHFFFAOYSA-N Ziprasidone Chemical compound C1=CC=C2C(N3CCN(CC3)CCC3=CC=4CC(=O)NC=4C=C3Cl)=NSC2=C1 MVWVFYHBGMAFLY-UHFFFAOYSA-N 0.000 description 1
- ZXVNMYWKKDOREA-UHFFFAOYSA-N Zomepirac Chemical compound C1=C(CC(O)=O)N(C)C(C(=O)C=2C=CC(Cl)=CC=2)=C1C ZXVNMYWKKDOREA-UHFFFAOYSA-N 0.000 description 1
- HDOZVRUNCMBHFH-UHFFFAOYSA-N Zotepine Chemical compound CN(C)CCOC1=CC2=CC=CC=C2SC2=CC=C(Cl)C=C12 HDOZVRUNCMBHFH-UHFFFAOYSA-N 0.000 description 1
- 210000002517 Zygapophyseal Joint Anatomy 0.000 description 1
- RVCSYOQWLPPAOA-CVPHZBIISA-M [(5S)-spiro[8-azoniabicyclo[3.2.1]octane-8,1'-azolidin-1-ium]-3-yl] 2-hydroxy-2,2-diphenylacetate;chloride Chemical compound [Cl-].[N+]12([C@H]3CCC2CC(C3)OC(=O)C(O)(C=2C=CC=CC=2)C=2C=CC=CC=2)CCCC1 RVCSYOQWLPPAOA-CVPHZBIISA-M 0.000 description 1
- PRXRUNOAOLTIEF-XDTJCZEISA-N [2-[(2R,3S,4R)-4-hydroxy-3-[(Z)-octadec-9-enoyl]oxyoxolan-2-yl]-2-[(Z)-octadec-9-enoyl]oxyethyl] (Z)-octadec-9-enoate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(OC(=O)CCCCCCC\C=C/CCCCCCCC)[C@H]1OC[C@@H](O)[C@@H]1OC(=O)CCCCCCC\C=C/CCCCCCCC PRXRUNOAOLTIEF-XDTJCZEISA-N 0.000 description 1
- SRUIBJVUBBOHHY-UHFFFAOYSA-N [2H-benzotriazol-4-yloxy(dimethylamino)methylidene]-dimethylazanium;hexafluorophosphate Chemical group F[P-](F)(F)(F)(F)F.CN(C)C(=[N+](C)C)OC1=CC=CC2=C1N=NN2 SRUIBJVUBBOHHY-UHFFFAOYSA-N 0.000 description 1
- DMEPDNFRHUGNPT-UHFFFAOYSA-N [5-(diethylamino)-2-methylpent-3-yn-2-yl] 2-cyclohexyl-2-hydroxy-2-phenylacetate Chemical compound C=1C=CC=CC=1C(O)(C(=O)OC(C)(C)C#CCN(CC)CC)C1CCCCC1 DMEPDNFRHUGNPT-UHFFFAOYSA-N 0.000 description 1
- UQYZFNUUOSSNKT-UHFFFAOYSA-N [benzotriazol-1-yloxy(dimethylamino)methylidene]-dimethylazanium;hexafluorophosphate Chemical compound F[P-](F)(F)(F)(F)F.C1=CC=C2N(OC(N(C)C)=[N+](C)C)N=NC2=C1 UQYZFNUUOSSNKT-UHFFFAOYSA-N 0.000 description 1
- 230000035507 absorption Effects 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- LXNAVEXFUKBNMK-UHFFFAOYSA-N acetic acid;palladium Chemical compound [Pd].CC(O)=O.CC(O)=O LXNAVEXFUKBNMK-UHFFFAOYSA-N 0.000 description 1
- 229960001138 acetylsalicylic acid Drugs 0.000 description 1
- 230000003213 activating Effects 0.000 description 1
- 201000003229 acute pancreatitis Diseases 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000000240 adjuvant Effects 0.000 description 1
- 201000005188 adrenal gland cancer Diseases 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 201000007930 alcohol dependence Diseases 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000003973 alkyl amines Chemical class 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 238000010976 amide bond formation reaction Methods 0.000 description 1
- 238000005576 amination reaction Methods 0.000 description 1
- 229960003036 amisulpride Drugs 0.000 description 1
- USFZMSVCRYTOJT-UHFFFAOYSA-N ammonium acetate Chemical compound N.CC(O)=O USFZMSVCRYTOJT-UHFFFAOYSA-N 0.000 description 1
- 235000019257 ammonium acetate Nutrition 0.000 description 1
- 229940043376 ammonium acetate Drugs 0.000 description 1
- 235000012538 ammonium bicarbonate Nutrition 0.000 description 1
- 239000001099 ammonium carbonate Substances 0.000 description 1
- 230000003444 anaesthetic Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 230000037001 anhydrosis Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000001773 anti-convulsant Effects 0.000 description 1
- 230000001430 anti-depressive Effects 0.000 description 1
- 230000003110 anti-inflammatory Effects 0.000 description 1
- 230000000111 anti-oxidant Effects 0.000 description 1
- 108090001123 antibodies Proteins 0.000 description 1
- 102000004965 antibodies Human genes 0.000 description 1
- 239000001961 anticonvulsive agent Substances 0.000 description 1
- 239000000935 antidepressant agent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 239000003420 antiserotonin agent Substances 0.000 description 1
- 229960001372 aprepitant Drugs 0.000 description 1
- 229960003153 aprobarbital Drugs 0.000 description 1
- 239000011260 aqueous acid Substances 0.000 description 1
- 239000007900 aqueous suspension Substances 0.000 description 1
- 239000008135 aqueous vehicle Substances 0.000 description 1
- 230000002917 arthritic Effects 0.000 description 1
- BSYNRYMUTXBXSQ-UHFFFAOYSA-N aspirin Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O BSYNRYMUTXBXSQ-UHFFFAOYSA-N 0.000 description 1
- 238000000065 atmospheric pressure chemical ionisation Methods 0.000 description 1
- 230000003305 autocrine Effects 0.000 description 1
- 125000000751 azo group Chemical group [*]N=N[*] 0.000 description 1
- 125000003943 azolyl group Chemical group 0.000 description 1
- GVLZYMDNTPNTLV-UHFFFAOYSA-N barbiturate(1-) Chemical compound O=C1[CH-]C(=O)NC(=O)N1 GVLZYMDNTPNTLV-UHFFFAOYSA-N 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 229960001716 benzalkonium 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
- CYDRXTMLKJDRQH-UHFFFAOYSA-N benzyl-dodecyl-dimethylazanium Chemical compound CCCCCCCCCCCC[N+](C)(C)CC1=CC=CC=C1 CYDRXTMLKJDRQH-UHFFFAOYSA-N 0.000 description 1
- 125000000051 benzyloxy group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])O* 0.000 description 1
- 239000000227 bioadhesive Substances 0.000 description 1
- 230000035514 bioavailability Effects 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- SIPUZPBQZHNSDW-UHFFFAOYSA-N bis(2-methylpropyl)aluminum Chemical compound CC(C)C[Al]CC(C)C SIPUZPBQZHNSDW-UHFFFAOYSA-N 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-M bisulphate group Chemical group S([O-])(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-M 0.000 description 1
- 201000000053 blastoma Diseases 0.000 description 1
- 230000000903 blocking Effects 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N borate Chemical compound [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 1
- 201000005216 brain cancer Diseases 0.000 description 1
- WKBOTKDWSSQWDR-UHFFFAOYSA-N bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- RMRJXGBAOAMLHD-CTAPUXPBSA-N buprenorphine Chemical compound C([C@]12[C@H]3OC=4C(O)=CC=C(C2=4)C[C@@H]2[C@]11CC[C@@]3([C@H](C1)[C@](C)(O)C(C)(C)C)OC)CN2CC1CC1 RMRJXGBAOAMLHD-CTAPUXPBSA-N 0.000 description 1
- 229960001736 buprenorphine Drugs 0.000 description 1
- 229940015694 butabarbital Drugs 0.000 description 1
- 235000019437 butane-1,3-diol Nutrition 0.000 description 1
- 229920005549 butyl rubber Polymers 0.000 description 1
- PUPZLCDOIYMWBV-UHFFFAOYSA-N butylene glycol Chemical compound CC(O)CCO PUPZLCDOIYMWBV-UHFFFAOYSA-N 0.000 description 1
- XAAHAAMILDNBPS-UHFFFAOYSA-L calcium hydrogenphosphate dihydrate Chemical compound O.O.[Ca+2].OP([O-])([O-])=O XAAHAAMILDNBPS-UHFFFAOYSA-L 0.000 description 1
- 235000013539 calcium stearate Nutrition 0.000 description 1
- 239000008116 calcium stearate Substances 0.000 description 1
- 150000001718 carbodiimides Chemical class 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 238000005810 carbonylation reaction Methods 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 229960000590 celecoxib Drugs 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000001413 cellular Effects 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 230000035605 chemotaxis Effects 0.000 description 1
- 235000015218 chewing gum Nutrition 0.000 description 1
- 235000015111 chews Nutrition 0.000 description 1
- 229960004831 chlorcyclizine Drugs 0.000 description 1
- 125000006390 chloropyrimidinyl group Chemical group 0.000 description 1
- 229960003291 chlorphenamine Drugs 0.000 description 1
- 239000000544 cholinesterase inhibitor Substances 0.000 description 1
- BLFJCJKTSCOLEY-UHFFFAOYSA-N cinnoline-3-carboxylic acid Chemical compound C1=CC=C2N=NC(C(=O)O)=CC2=C1 BLFJCJKTSCOLEY-UHFFFAOYSA-N 0.000 description 1
- 229960001653 citalopram Drugs 0.000 description 1
- 229960004606 clomipramine Drugs 0.000 description 1
- 229960004362 clorazepate Drugs 0.000 description 1
- XDDJGVMJFWAHJX-UHFFFAOYSA-N clorazepic acid Chemical compound C12=CC(Cl)=CC=C2NC(=O)C(C(=O)O)N=C1C1=CC=CC=C1 XDDJGVMJFWAHJX-UHFFFAOYSA-N 0.000 description 1
- 229960004170 clozapine Drugs 0.000 description 1
- 239000011280 coal tar Substances 0.000 description 1
- 229960003920 cocaine Drugs 0.000 description 1
- ZPUCINDJVBIVPJ-LJISPDSOSA-N cocaine Chemical compound O([C@H]1C[C@@H]2CC[C@@H](N2C)[C@H]1C(=O)OC)C(=O)C1=CC=CC=C1 ZPUCINDJVBIVPJ-LJISPDSOSA-N 0.000 description 1
- ZPUCINDJVBIVPJ-BARDWOONSA-N cocaine Natural products O([C@@H]1C[C@H]2CC[C@H](N2C)[C@@H]1C(=O)OC)C(=O)C1=CC=CC=C1 ZPUCINDJVBIVPJ-BARDWOONSA-N 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 229960004126 codeine Drugs 0.000 description 1
- 201000003963 colon carcinoma Diseases 0.000 description 1
- 201000011231 colorectal cancer Diseases 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 229940000425 combination drugs Drugs 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 239000012059 conventional drug carrier Substances 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- JZCCFEFSEZPSOG-UHFFFAOYSA-L copper(II) sulfate pentahydrate Chemical compound O.O.O.O.O.[Cu+2].[O-]S([O-])(=O)=O JZCCFEFSEZPSOG-UHFFFAOYSA-L 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 235000010947 crosslinked sodium carboxy methyl cellulose Nutrition 0.000 description 1
- 238000002425 crystallisation Methods 0.000 description 1
- 230000005712 crystallization Effects 0.000 description 1
- 238000007333 cyanation reaction Methods 0.000 description 1
- 229960003572 cyclobenzaprine Drugs 0.000 description 1
- VOLSCWDWGMWXGO-UHFFFAOYSA-N cyclobuten-1-yl acetate Chemical compound CC(=O)OC1=CCC1 VOLSCWDWGMWXGO-UHFFFAOYSA-N 0.000 description 1
- 125000000131 cyclopropyloxy group Chemical group C1(CC1)O* 0.000 description 1
- 201000003146 cystitis Diseases 0.000 description 1
- 229960005217 dapoxetine Drugs 0.000 description 1
- 229960002677 darifenacin Drugs 0.000 description 1
- 238000006298 dechlorination reaction Methods 0.000 description 1
- 230000003247 decreasing Effects 0.000 description 1
- 229960003314 deracoxib Drugs 0.000 description 1
- 239000007933 dermal patch Substances 0.000 description 1
- 229960003914 desipramine Drugs 0.000 description 1
- VPIXQGUBUKFLRF-UHFFFAOYSA-N desmethylclomipramine Chemical compound C1CC2=CC=C(Cl)C=C2N(CCCNC)C2=CC=CC=C21 VPIXQGUBUKFLRF-UHFFFAOYSA-N 0.000 description 1
- HEDRZPFGACZZDS-MICDWDOJSA-N deuterated chloroform Substances [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 1
- 229910052805 deuterium Inorganic materials 0.000 description 1
- 229960004193 dextropropoxyphene Drugs 0.000 description 1
- DNUQIKJYTKKUTB-UHFFFAOYSA-N di(propan-2-yl)alumanylium;hydride Chemical compound [H-].CC(C)[Al+]C(C)C DNUQIKJYTKKUTB-UHFFFAOYSA-N 0.000 description 1
- 229960002069 diamorphine Drugs 0.000 description 1
- 229960003529 diazepam Drugs 0.000 description 1
- 229960005422 dichloralphenazone Drugs 0.000 description 1
- 229960001259 diclofenac Drugs 0.000 description 1
- DCOPUUMXTXDBNB-UHFFFAOYSA-N diclofenac Chemical compound OC(=O)CC1=CC=CC=C1NC1=C(Cl)C=CC=C1Cl DCOPUUMXTXDBNB-UHFFFAOYSA-N 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 125000004212 difluorophenyl group Chemical group 0.000 description 1
- 210000002249 digestive system Anatomy 0.000 description 1
- 229960000920 dihydrocodeine Drugs 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-M dihydrogenphosphate Chemical compound OP(O)([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-M 0.000 description 1
- UXGNZZKBCMGWAZ-UHFFFAOYSA-N dimethylformamide DMF Chemical compound CN(C)C=O.CN(C)C=O UXGNZZKBCMGWAZ-UHFFFAOYSA-N 0.000 description 1
- 229960002986 dinoprostone Drugs 0.000 description 1
- 229960000520 diphenhydramine Drugs 0.000 description 1
- JZBWUTVDIDNCMW-UHFFFAOYSA-L dipotassium;oxido sulfate Chemical compound [K+].[K+].[O-]OS([O-])(=O)=O JZBWUTVDIDNCMW-UHFFFAOYSA-L 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 238000006073 displacement reaction Methods 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- SACNIGZYDTUHKB-UHFFFAOYSA-N ditert-butyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane Chemical group CC(C)C1=CC(C(C)C)=CC(C(C)C)=C1C1=CC=CC=C1P(C(C)(C)C)C(C)(C)C SACNIGZYDTUHKB-UHFFFAOYSA-N 0.000 description 1
- KLBQZWRITKRQQV-UHFFFAOYSA-N dl-Thioridazine Chemical compound C12=CC(SC)=CC=C2SC2=CC=CC=C2N1CCC1CCCCN1C KLBQZWRITKRQQV-UHFFFAOYSA-N 0.000 description 1
- IXTMWRCNAAVVAI-UHFFFAOYSA-N dofetilide Chemical compound C=1C=C(NS(C)(=O)=O)C=CC=1CCN(C)CCOC1=CC=C(NS(C)(=O)=O)C=C1 IXTMWRCNAAVVAI-UHFFFAOYSA-N 0.000 description 1
- 229960002994 dofetilide Drugs 0.000 description 1
- 229960003530 donepezil Drugs 0.000 description 1
- 229940000406 drug candidates Drugs 0.000 description 1
- 229960002866 duloxetine Drugs 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 201000006549 dyspepsia Diseases 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 238000000132 electrospray ionisation Methods 0.000 description 1
- 229960002472 eletriptan Drugs 0.000 description 1
- 201000008184 embryoma Diseases 0.000 description 1
- 239000002621 endocannabinoid Substances 0.000 description 1
- 201000009273 endometriosis Diseases 0.000 description 1
- 239000007920 enema Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 125000002587 enol group Chemical group 0.000 description 1
- 229960004341 escitalopram Drugs 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 125000004185 ester group Chemical group 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- HZAXFHJVJLSVMW-UHFFFAOYSA-N ethanolamine Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 1
- DLLJVQNYBYOKGS-UHFFFAOYSA-N ethoxyethane;pentane Chemical compound CCCCC.CCOCC DLLJVQNYBYOKGS-UHFFFAOYSA-N 0.000 description 1
- 239000005038 ethylene vinyl acetate Substances 0.000 description 1
- 125000004705 ethylthio group Chemical group C(C)S* 0.000 description 1
- NNYBQONXHNTVIJ-UHFFFAOYSA-N etodolac Chemical compound C1COC(CC)(CC(O)=O)C2=C1C(C=CC=C1CC)=C1N2 NNYBQONXHNTVIJ-UHFFFAOYSA-N 0.000 description 1
- 229960004945 etoricoxib Drugs 0.000 description 1
- 238000001704 evaporation Methods 0.000 description 1
- 230000000763 evoked Effects 0.000 description 1
- 230000001747 exhibiting Effects 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 230000002349 favourable Effects 0.000 description 1
- 229950003930 femoxetine Drugs 0.000 description 1
- 231100000592 few side effect Toxicity 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 229910052731 fluorine Inorganic materials 0.000 description 1
- 239000011737 fluorine Substances 0.000 description 1
- YCKRFDGAMUMZLT-UHFFFAOYSA-N fluorine atom Chemical compound [F] YCKRFDGAMUMZLT-UHFFFAOYSA-N 0.000 description 1
- 125000001153 fluoro group Chemical group F* 0.000 description 1
- 125000001207 fluorophenyl group Chemical group 0.000 description 1
- 229960002390 flurbiprofen Drugs 0.000 description 1
- CJOFXWAVKWHTFT-XSFVSMFZSA-N fluvoxamine Chemical compound COCCCC\C(=N/OCCN)C1=CC=C(C(F)(F)F)C=C1 CJOFXWAVKWHTFT-XSFVSMFZSA-N 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 229940044170 formate Drugs 0.000 description 1
- 229940050411 fumarate Drugs 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N fumaric acid Chemical compound OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- IZLZCXOWFDCZLD-UHFFFAOYSA-N furo[2,3-b]pyridine-2-carboxylic acid Chemical compound C1=CN=C2OC(C(=O)O)=CC2=C1 IZLZCXOWFDCZLD-UHFFFAOYSA-N 0.000 description 1
- 125000004612 furopyridinyl group Chemical group O1C(=CC2=C1C=CC=N2)* 0.000 description 1
- 102000037240 fusion proteins Human genes 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 229960002870 gabapentin Drugs 0.000 description 1
- 201000006860 gastroesophageal reflux disease Diseases 0.000 description 1
- 239000007903 gelatin capsule Substances 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 230000000762 glandular Effects 0.000 description 1
- 229940050410 gluconate Drugs 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 229940097042 glucuronate Drugs 0.000 description 1
- 230000004116 glycogenolysis Effects 0.000 description 1
- 201000005569 gout Diseases 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- ZRALSGWEFCBTJO-UHFFFAOYSA-N guanidine group Chemical group NC(=N)N ZRALSGWEFCBTJO-UHFFFAOYSA-N 0.000 description 1
- 235000013928 guanylic acid Nutrition 0.000 description 1
- 229960003878 haloperidol Drugs 0.000 description 1
- 239000007902 hard capsule Substances 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 125000003187 heptyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 239000000938 histamine H1 antagonist Substances 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 229920002674 hyaluronan Polymers 0.000 description 1
- 229960003160 hyaluronic acid Drugs 0.000 description 1
- 150000002430 hydrocarbons Chemical class 0.000 description 1
- 229960000240 hydrocodone Drugs 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 150000002431 hydrogen Chemical class 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- IXCSERBJSXMMFS-UHFFFAOYSA-N hydrogen chloride Substances Cl.Cl IXCSERBJSXMMFS-UHFFFAOYSA-N 0.000 description 1
- 229910000041 hydrogen chloride Inorganic materials 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- 238000005984 hydrogenation reaction Methods 0.000 description 1
- 230000003301 hydrolyzing Effects 0.000 description 1
- 229960001410 hydromorphone Drugs 0.000 description 1
- 230000002209 hydrophobic Effects 0.000 description 1
- 235000019447 hydroxyethyl cellulose Nutrition 0.000 description 1
- 230000002989 hypothyroidism Effects 0.000 description 1
- 229960001680 ibuprofen Drugs 0.000 description 1
- 229960003162 iloperidone Drugs 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 239000005414 inactive ingredient Substances 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 230000001524 infective Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000007915 intraurethral administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 238000006192 iodination reaction Methods 0.000 description 1
- 125000002346 iodo group Chemical group I* 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- OEXHQOGQTVQTAT-JRNQLAHRSA-N ipratropium Chemical compound O([C@H]1C[C@H]2CC[C@@H](C1)[N@@+]2(C)C(C)C)C(=O)C(CO)C1=CC=CC=C1 OEXHQOGQTVQTAT-JRNQLAHRSA-N 0.000 description 1
- SUMDYPCJJOFFON-UHFFFAOYSA-M isethionate Chemical compound OCCS([O-])(=O)=O SUMDYPCJJOFFON-UHFFFAOYSA-M 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 150000002513 isocyanates Chemical class 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- FZWBNHMXJMCXLU-BLAUPYHCSA-N isomaltotriose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1OC[C@@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@@H](OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C=O)O1 FZWBNHMXJMCXLU-BLAUPYHCSA-N 0.000 description 1
- 229960003299 ketamine Drugs 0.000 description 1
- 150000002576 ketones Chemical class 0.000 description 1
- 229960000991 ketoprofen Drugs 0.000 description 1
- 229960004752 ketorolac Drugs 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 229960001848 lamotrigine Drugs 0.000 description 1
- 230000002045 lasting Effects 0.000 description 1
- 239000003913 leukotriene B4 receptor antagonist Substances 0.000 description 1
- 229960000263 levallorphan Drugs 0.000 description 1
- 230000004301 light adaptation Effects 0.000 description 1
- NZTNZPDOBQDOSO-UHFFFAOYSA-N lithium;boron(1-) Chemical compound [Li+].[B-] NZTNZPDOBQDOSO-UHFFFAOYSA-N 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 229960004391 lorazepam Drugs 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 229960000994 lumiracoxib Drugs 0.000 description 1
- KHPKQFYUPIUARC-UHFFFAOYSA-N lumiracoxib Chemical compound OC(=O)CC1=CC(C)=CC=C1NC1=C(F)C=CC=C1Cl KHPKQFYUPIUARC-UHFFFAOYSA-N 0.000 description 1
- 229960001432 lurasidone Drugs 0.000 description 1
- 235000018977 lysine Nutrition 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 230000014759 maintenance of location Effects 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
- 230000036210 malignancy Effects 0.000 description 1
- 230000003211 malignant Effects 0.000 description 1
- NUJOXMJBOLGQSY-UHFFFAOYSA-N manganese dioxide Inorganic materials O=[Mn]=O NUJOXMJBOLGQSY-UHFFFAOYSA-N 0.000 description 1
- 229960004090 maprotiline Drugs 0.000 description 1
- 229960003803 meclofenamic acid Drugs 0.000 description 1
- 230000001404 mediated Effects 0.000 description 1
- 229960003464 mefenamic acid Drugs 0.000 description 1
- 229960001929 meloxicam Drugs 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 229960004640 memantine Drugs 0.000 description 1
- 229960000582 mepyramine Drugs 0.000 description 1
- QSHDDOUJBYECFT-UHFFFAOYSA-N mercury Chemical compound [Hg] QSHDDOUJBYECFT-UHFFFAOYSA-N 0.000 description 1
- 229910052753 mercury Inorganic materials 0.000 description 1
- 230000001394 metastastic Effects 0.000 description 1
- 229960001797 methadone Drugs 0.000 description 1
- LSDPWZHWYPCBBB-UHFFFAOYSA-N methanethiol Chemical compound SC LSDPWZHWYPCBBB-UHFFFAOYSA-N 0.000 description 1
- BDAGIHXWWSANSR-UHFFFAOYSA-M methanoate Chemical compound [O-]C=O BDAGIHXWWSANSR-UHFFFAOYSA-M 0.000 description 1
- 229960002330 methocarbamol Drugs 0.000 description 1
- 229960002683 methohexital Drugs 0.000 description 1
- JZMJDSHXVKJFKW-UHFFFAOYSA-M methyl sulfate(1-) Chemical compound COS([O-])(=O)=O JZMJDSHXVKJFKW-UHFFFAOYSA-M 0.000 description 1
- QMMFVYPAHWMCMS-UHFFFAOYSA-N methyl sulfide Chemical compound CSC QMMFVYPAHWMCMS-UHFFFAOYSA-N 0.000 description 1
- 125000006217 methyl sulfide group Chemical group [H]C([H])([H])S* 0.000 description 1
- BAVYZALUXZFZLV-UHFFFAOYSA-N methylamine Chemical compound NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 1
- NQMRYBIKMRVZLB-UHFFFAOYSA-N methylamine hydrochloride Chemical compound [Cl-].[NH3+]C NQMRYBIKMRVZLB-UHFFFAOYSA-N 0.000 description 1
- 229960001703 methylphenobarbital Drugs 0.000 description 1
- 229960003404 mexiletine Drugs 0.000 description 1
- 239000004530 micro-emulsion Substances 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 230000003228 microsomal Effects 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 229960001165 modafinil Drugs 0.000 description 1
- MNNJUOUZAXINJW-UHFFFAOYSA-N molecular hydrogen;hydrobromide Chemical compound Br.[H][H] MNNJUOUZAXINJW-UHFFFAOYSA-N 0.000 description 1
- BDRTVPCFKSUHCJ-UHFFFAOYSA-N molecular hydrogen;potassium Chemical compound [K].[H][H] BDRTVPCFKSUHCJ-UHFFFAOYSA-N 0.000 description 1
- 125000002950 monocyclic group Chemical group 0.000 description 1
- 229940113083 morpholine Drugs 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- 230000003232 mucoadhesive Effects 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 201000009251 multiple myeloma Diseases 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 230000000869 mutational Effects 0.000 description 1
- 239000003158 myorelaxant agent Substances 0.000 description 1
- 201000002481 myositis Diseases 0.000 description 1
- 239000003703 n methyl dextro aspartic acid receptor blocking agent Substances 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 229960005297 nalmefene Drugs 0.000 description 1
- 229960000938 nalorphine Drugs 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 229960002009 naproxen Drugs 0.000 description 1
- 229960005254 naratriptan Drugs 0.000 description 1
- 229960001800 nefazodone Drugs 0.000 description 1
- 239000002742 neurokinin 1 receptor antagonist Substances 0.000 description 1
- 239000002746 neurokinin 2 receptor antagonist Substances 0.000 description 1
- 239000002740 neurokinin 3 receptor antagonist Substances 0.000 description 1
- 210000002569 neurons Anatomy 0.000 description 1
- 230000002981 neuropathic Effects 0.000 description 1
- 201000001119 neuropathy Diseases 0.000 description 1
- 239000003900 neurotrophic factor Substances 0.000 description 1
- PXHVJJICTQNCMI-UHFFFAOYSA-N nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 1
- 229930015196 nicotine Natural products 0.000 description 1
- 235000001968 nicotinic acid Nutrition 0.000 description 1
- 239000011664 nicotinic acid Substances 0.000 description 1
- 229960000965 nimesulide Drugs 0.000 description 1
- 239000002353 niosome Substances 0.000 description 1
- DLWSRGHNJVLJAH-UHFFFAOYSA-N nitroflurbiprofen Chemical compound FC1=CC(C(C(=O)OCCCCO[N+]([O-])=O)C)=CC=C1C1=CC=CC=C1 DLWSRGHNJVLJAH-UHFFFAOYSA-N 0.000 description 1
- 239000000346 nonvolatile oil Substances 0.000 description 1
- 238000000655 nuclear magnetic resonance spectrum Methods 0.000 description 1
- KREXGRSOTUKPLX-UHFFFAOYSA-N octadecanoic acid;zinc Chemical compound [Zn].CCCCCCCCCCCCCCCCCC(O)=O.CCCCCCCCCCCCCCCCCC(O)=O KREXGRSOTUKPLX-UHFFFAOYSA-N 0.000 description 1
- 229960005017 olanzapine Drugs 0.000 description 1
- 229960004110 olsalazine Drugs 0.000 description 1
- 231100000590 oncogenic Toxicity 0.000 description 1
- 230000002246 oncogenic Effects 0.000 description 1
- 239000000014 opioid analgesic Substances 0.000 description 1
- 230000003287 optical Effects 0.000 description 1
- 230000036220 oral bioavailability Effects 0.000 description 1
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 description 1
- PXQPEWDEAKTCGB-UHFFFAOYSA-M orotate Chemical compound [O-]C(=O)C1=CC(=O)NC(=O)N1 PXQPEWDEAKTCGB-UHFFFAOYSA-M 0.000 description 1
- 230000003204 osmotic Effects 0.000 description 1
- 230000003349 osteoarthritic Effects 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- MUBZPKHOEPUJKR-UHFFFAOYSA-L oxalate Chemical compound [O-]C(=O)C([O-])=O MUBZPKHOEPUJKR-UHFFFAOYSA-L 0.000 description 1
- 229960002739 oxaprozin Drugs 0.000 description 1
- 229960004535 oxazepam Drugs 0.000 description 1
- 229960005434 oxybutynin Drugs 0.000 description 1
- 229960005118 oxymorphone Drugs 0.000 description 1
- RZVAJINKPMORJF-UHFFFAOYSA-N p-acetaminophenol Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 1
- 201000008175 pain disease Diseases 0.000 description 1
- PIBWKRNGBLPSSY-UHFFFAOYSA-L palladium(II) chloride Chemical compound Cl[Pd]Cl PIBWKRNGBLPSSY-UHFFFAOYSA-L 0.000 description 1
- IPCSVZSSVZVIGE-UHFFFAOYSA-M palmitate Chemical compound CCCCCCCCCCCCCCCC([O-])=O IPCSVZSSVZVIGE-UHFFFAOYSA-M 0.000 description 1
- 229960005489 paracetamol Drugs 0.000 description 1
- 230000003076 paracrine Effects 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 229960004662 parecoxib Drugs 0.000 description 1
- TZRHLKRLEZJVIJ-UHFFFAOYSA-N parecoxib Chemical compound C1=CC(S(=O)(=O)NC(=O)CC)=CC=C1C1=C(C)ON=C1C1=CC=CC=C1 TZRHLKRLEZJVIJ-UHFFFAOYSA-N 0.000 description 1
- 239000003182 parenteral nutrition solution Substances 0.000 description 1
- 229960002296 paroxetine Drugs 0.000 description 1
- 230000001314 paroxysmal Effects 0.000 description 1
- 230000001575 pathological Effects 0.000 description 1
- 230000035778 pathophysiological process Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 235000019371 penicillin G benzathine Nutrition 0.000 description 1
- 229960000482 pethidine Drugs 0.000 description 1
- 230000003285 pharmacodynamic Effects 0.000 description 1
- VYMDGNCVAMGZFE-UHFFFAOYSA-N phenylbutazonum Chemical compound O=C1C(CCCC)C(=O)N(C=2C=CC=CC=2)N1C1=CC=CC=C1 VYMDGNCVAMGZFE-UHFFFAOYSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-L phosphate Chemical compound OP([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-L 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- RJUAEBLXGFKZMS-UHFFFAOYSA-N piperidin-1-ylmethanol Chemical compound OCN1CCCCC1 RJUAEBLXGFKZMS-UHFFFAOYSA-N 0.000 description 1
- DNUTZBZXLPWRJG-UHFFFAOYSA-N piperidine-1-carboxylic acid Chemical compound OC(=O)N1CCCCC1 DNUTZBZXLPWRJG-UHFFFAOYSA-N 0.000 description 1
- 229960002702 piroxicam Drugs 0.000 description 1
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 1
- 229920000747 poly(lactic acid) polymer Polymers 0.000 description 1
- 229920001888 polyacrylic acid Polymers 0.000 description 1
- 239000004584 polyacrylic acid Substances 0.000 description 1
- 239000004633 polyglycolic acid Substances 0.000 description 1
- 239000004626 polylactic acid Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 229920001296 polysiloxane Polymers 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 235000019422 polyvinyl alcohol Nutrition 0.000 description 1
- 229920000523 polyvinylpolypyrrolidone Polymers 0.000 description 1
- 235000013809 polyvinylpolypyrrolidone Nutrition 0.000 description 1
- 230000002980 postoperative Effects 0.000 description 1
- 235000011056 potassium acetate Nutrition 0.000 description 1
- 229910000343 potassium bisulfate Inorganic materials 0.000 description 1
- 235000003270 potassium fluoride Nutrition 0.000 description 1
- NROKBHXJSPEDAR-UHFFFAOYSA-M potassium fluoride Inorganic materials [F-].[K+] NROKBHXJSPEDAR-UHFFFAOYSA-M 0.000 description 1
- 229910000160 potassium phosphate Inorganic materials 0.000 description 1
- 230000003389 potentiating Effects 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 229960001233 pregabalin Drugs 0.000 description 1
- 150000003138 primary alcohols Chemical class 0.000 description 1
- 229960003910 promethazine Drugs 0.000 description 1
- ATUOYWHBWRKTHZ-UHFFFAOYSA-N propane Chemical compound CCC ATUOYWHBWRKTHZ-UHFFFAOYSA-N 0.000 description 1
- BDERNNFJNOPAEC-UHFFFAOYSA-N propanol Chemical compound CCCO BDERNNFJNOPAEC-UHFFFAOYSA-N 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- XBDQKXXYIPTUBI-UHFFFAOYSA-N propionic acid Chemical compound CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 1
- 229960003510 propiverine Drugs 0.000 description 1
- 229960003712 propranolol Drugs 0.000 description 1
- ZXDUPDQEFOYLOM-UHFFFAOYSA-O propylideneazanium Chemical group [CH2-]CC=[NH2+] ZXDUPDQEFOYLOM-UHFFFAOYSA-O 0.000 description 1
- 210000000064 prostate epithelial cell Anatomy 0.000 description 1
- 201000007094 prostatitis Diseases 0.000 description 1
- 230000000506 psychotropic Effects 0.000 description 1
- WRHZVMBBRYBTKZ-UHFFFAOYSA-N pyrrole-2-carboxylic acid Chemical compound OC(=O)C1=CC=CN1 WRHZVMBBRYBTKZ-UHFFFAOYSA-N 0.000 description 1
- 150000003233 pyrroles Chemical class 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 229960004431 quetiapine Drugs 0.000 description 1
- LOAUVZALPPNFOQ-UHFFFAOYSA-N quinaldic acid Chemical compound C1=CC=CC2=NC(C(=O)O)=CC=C21 LOAUVZALPPNFOQ-UHFFFAOYSA-N 0.000 description 1
- SMWDFEZZVXVKRB-UHFFFAOYSA-N quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 230000035484 reaction time Effects 0.000 description 1
- 229960003770 reboxetine Drugs 0.000 description 1
- 201000001275 rectum cancer Diseases 0.000 description 1
- 238000006268 reductive amination reaction Methods 0.000 description 1
- 230000000268 renotropic Effects 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 229960000425 rizatriptan Drugs 0.000 description 1
- 229960000371 rofecoxib Drugs 0.000 description 1
- 235000019204 saccharin Nutrition 0.000 description 1
- 239000000901 saccharin and its Na,K and Ca salt Substances 0.000 description 1
- 210000003131 sacroiliac joint Anatomy 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 229960002060 secobarbital Drugs 0.000 description 1
- 230000008313 sensitization Effects 0.000 description 1
- 230000001235 sensitizing Effects 0.000 description 1
- 239000002400 serotonin 2A antagonist Substances 0.000 description 1
- 239000003369 serotonin 5-HT3 receptor antagonist Substances 0.000 description 1
- 239000000952 serotonin receptor agonist Substances 0.000 description 1
- 239000003772 serotonin uptake inhibitor Substances 0.000 description 1
- 229960000652 sertindole Drugs 0.000 description 1
- 231100000486 side effect Toxicity 0.000 description 1
- 229960003310 sildenafil Drugs 0.000 description 1
- 150000004760 silicates Chemical class 0.000 description 1
- 235000012239 silicon dioxide Nutrition 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 150000003385 sodium Chemical class 0.000 description 1
- MSXHSNHNTORCAW-UHFFFAOYSA-M sodium 3,4,5,6-tetrahydroxyoxane-2-carboxylate Chemical class [Na+].OC1OC(C([O-])=O)C(O)C(O)C1O MSXHSNHNTORCAW-UHFFFAOYSA-M 0.000 description 1
- 239000000661 sodium alginate Chemical class 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 229910000342 sodium bisulfate Inorganic materials 0.000 description 1
- YOQDYZUWIQVZSF-UHFFFAOYSA-N sodium borohydride Substances [BH4-].[Na+] YOQDYZUWIQVZSF-UHFFFAOYSA-N 0.000 description 1
- 229910000033 sodium borohydride Inorganic materials 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 229960002218 sodium chlorite Drugs 0.000 description 1
- 239000004289 sodium hydrogen sulphite Substances 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 229940001584 sodium metabisulfite Drugs 0.000 description 1
- 235000010262 sodium metabisulphite Nutrition 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 235000011008 sodium phosphates Nutrition 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 229940045902 sodium stearyl fumarate Drugs 0.000 description 1
- 235000019345 sodium thiosulphate Nutrition 0.000 description 1
- MAKUBRYLFHZREJ-JWBQXVCJSA-M sodium;(2S,3S,4R,5R,6R)-3-[(2S,3R,5S,6R)-3-acetamido-5-hydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-4,5,6-trihydroxyoxane-2-carboxylate Chemical compound [Na+].CC(=O)N[C@@H]1C[C@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](C([O-])=O)O[C@@H](O)[C@H](O)[C@H]1O MAKUBRYLFHZREJ-JWBQXVCJSA-M 0.000 description 1
- STFSJTPVIIDAQX-LTRPLHCISA-M sodium;(E)-4-octadecoxy-4-oxobut-2-enoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCCOC(=O)\C=C\C([O-])=O STFSJTPVIIDAQX-LTRPLHCISA-M 0.000 description 1
- ODGROJYWQXFQOZ-UHFFFAOYSA-N sodium;boron(1-) Chemical compound [B-].[Na+] ODGROJYWQXFQOZ-UHFFFAOYSA-N 0.000 description 1
- 239000007901 soft capsule Substances 0.000 description 1
- 239000008247 solid mixture Substances 0.000 description 1
- 239000006104 solid solution Substances 0.000 description 1
- 229960003855 solifenacin Drugs 0.000 description 1
- 235000019337 sorbitan trioleate Nutrition 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 229960002920 sorbitol Drugs 0.000 description 1
- 235000010356 sorbitol Nutrition 0.000 description 1
- 201000005671 spondyloarthropathy Diseases 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 229940114926 stearate Drugs 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 238000003860 storage Methods 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
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 230000003319 supportive Effects 0.000 description 1
- 239000002511 suppository base Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000000152 swallowing Effects 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 239000007916 tablet composition Substances 0.000 description 1
- 229960000835 tadalafil Drugs 0.000 description 1
- 229960002613 tamsulosin Drugs 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- 229960001367 tartaric acid Drugs 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- 239000006068 taste-masking agent Substances 0.000 description 1
- 238000003419 tautomerization reaction Methods 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- FGTJJHCZWOVVNH-UHFFFAOYSA-N tert-butyl-[tert-butyl(dimethyl)silyl]oxy-dimethylsilane Chemical compound CC(C)(C)[Si](C)(C)O[Si](C)(C)C(C)(C)C FGTJJHCZWOVVNH-UHFFFAOYSA-N 0.000 description 1
- YTSOZDAUXGYEHP-UHFFFAOYSA-N tert-butyl-dimethyl-(2-methyl-2-pyrrolo[2,3-d]pyrimidin-7-ylpropoxy)silane Chemical compound N1=CN=C2N(C(C)(CO[Si](C)(C)C(C)(C)C)C)C=CC2=C1 YTSOZDAUXGYEHP-UHFFFAOYSA-N 0.000 description 1
- 125000005931 tert-butyloxycarbonyl group Chemical group [H]C([H])([H])C(OC(*)=O)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- WHRNULOCNSKMGB-UHFFFAOYSA-N tetrahydrofuran THF Chemical compound C1CCOC1.C1CCOC1 WHRNULOCNSKMGB-UHFFFAOYSA-N 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- WVWCTPQHJWMLKI-UHFFFAOYSA-N thieno[3,2-b]pyridine-2-carboxylic acid Chemical compound C1=CC=C2SC(C(=O)O)=CC2=N1 WVWCTPQHJWMLKI-UHFFFAOYSA-N 0.000 description 1
- VKOINCMQZKBKHH-UHFFFAOYSA-N thieno[3,2-c]pyridine-2-carboxylic acid Chemical compound N1=CC=C2SC(C(=O)O)=CC2=C1 VKOINCMQZKBKHH-UHFFFAOYSA-N 0.000 description 1
- 229960002784 thioridazine Drugs 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 231100000886 tinnitus Toxicity 0.000 description 1
- 235000010384 tocopherol Nutrition 0.000 description 1
- 229960001017 tolmetin Drugs 0.000 description 1
- 229960004045 tolterodine Drugs 0.000 description 1
- OOGJQPCLVADCPB-HXUWFJFHSA-N tolterodine Chemical compound C1([C@@H](CCN(C(C)C)C(C)C)C=2C(=CC=C(C)C=2)O)=CC=CC=C1 OOGJQPCLVADCPB-HXUWFJFHSA-N 0.000 description 1
- 229960004394 topiramate Drugs 0.000 description 1
- 230000002588 toxic Effects 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000000844 transformation Methods 0.000 description 1
- 230000001131 transforming Effects 0.000 description 1
- 235000019731 tricalcium phosphate Nutrition 0.000 description 1
- 125000004044 trifluoroacetyl group Chemical group FC(C(=O)*)(F)F 0.000 description 1
- 102000015533 trkA Receptor Human genes 0.000 description 1
- 108010064884 trkA Receptor Proteins 0.000 description 1
- 229960000281 trometamol Drugs 0.000 description 1
- 210000004881 tumor cells Anatomy 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 238000007039 two-step reaction Methods 0.000 description 1
- 125000001493 tyrosinyl group Chemical group [H]OC1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 201000006704 ulcerative colitis Diseases 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 229960002004 valdecoxib Drugs 0.000 description 1
- LNPDTQAFDNKSHK-UHFFFAOYSA-N valdecoxib Chemical compound CC=1ON=C(C=2C=CC=CC=2)C=1C1=CC=C(S(N)(=O)=O)C=C1 LNPDTQAFDNKSHK-UHFFFAOYSA-N 0.000 description 1
- NIJJYAXOARWZEE-UHFFFAOYSA-M valproate Chemical compound CCCC(C([O-])=O)CCC NIJJYAXOARWZEE-UHFFFAOYSA-M 0.000 description 1
- 239000000105 vanilloid receptor agonist Substances 0.000 description 1
- 229960002381 vardenafil Drugs 0.000 description 1
- 229960004688 venlafaxine Drugs 0.000 description 1
- 201000006744 villonodular synovitis Diseases 0.000 description 1
- QYKIQEUNHZKYBP-UHFFFAOYSA-N vinyl ether Chemical compound C=COC=C QYKIQEUNHZKYBP-UHFFFAOYSA-N 0.000 description 1
- 239000003039 volatile agent Substances 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 239000003871 white petrolatum Substances 0.000 description 1
- MWLSOWXNZPKENC-SSDOTTSWSA-N zileuton Chemical compound C1=CC=C2SC([C@H](N(O)C(N)=O)C)=CC2=C1 MWLSOWXNZPKENC-SSDOTTSWSA-N 0.000 description 1
- 229960005332 zileuton Drugs 0.000 description 1
- 150000003751 zinc Chemical class 0.000 description 1
- 229960000607 ziprasidone Drugs 0.000 description 1
- 229960001360 zolmitriptan Drugs 0.000 description 1
- ULSDMUVEXKOYBU-ZDUSSCGKSA-N zolmitriptan Chemical compound C1=C2C(CCN(C)C)=CNC2=CC=C1C[C@H]1COC(=O)N1 ULSDMUVEXKOYBU-ZDUSSCGKSA-N 0.000 description 1
- 229960003414 zomepirac Drugs 0.000 description 1
- 229960004496 zotepine Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/04—Centrally acting analgesics, e.g. opioids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D513/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
- C07D513/02—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
- C07D513/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D519/00—Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
Abstract
Disclosed are pyrrolo[2,3-d]pyrimidine compounds of Formula (I) wherein the substituents are as described in the specification, which includes the compound N-(5-{[2-amino-7-(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-dlpyrimidin-5-yllcarbonyl}pyridin-3-yl)-2-[4-(trifluoromethyl)phenyllacetamide. Also disclosed are processes for the preparation of the compounds, compositions containing the compounds, and the uses of such compounds and salts in treating diseases or conditions associated with tropomyosin-related kinase (Trk), and activity. More specifically the invention relates to the compounds and their salts useful as inhibitors of Trk for the treatment of diseases or conditions such as pain and cancer. so disclosed are processes for the preparation of the compounds, compositions containing the compounds, and the uses of such compounds and salts in treating diseases or conditions associated with tropomyosin-related kinase (Trk), and activity. More specifically the invention relates to the compounds and their salts useful as inhibitors of Trk for the treatment of diseases or conditions such as pain and cancer.
Description
PYRROLO [2,3-D] PYRIMIDINE DERIVATIVES AS INHIBITORS OF TROPOMYSIN-RELATED
KINASES
The invention described herein relates to n o[2,3-d}pyrimidine compounds and the
pharmaceutically acceptable salts of such nds. The invention also relates to the processes for the
preparation of the compounds, itions containing the compounds, and the uses of such compounds
and salts in treating diseases or conditions associated with tropomyosin-related kinase (Trk), activity.
More specifically the invention relates to the compounds and their salts useful as inhibitors of Trk .
BACKGROUND
Tropomyosin-related kinases (Trks) are a family of or tyrosine kinases activated by neurotrophins.
Trks play important roles in pain sensation as well as tumour cell growth and survival signaling. Thus,
inhibitors of Trk receptor kinases might provide targeted treatments for conditions such as pain and
cancer. Recent developments in this field have been reviewed by Wang et al in Expert Opin. Ther.
Patents (2009) 19(3): 305-319 and an extract is reproduced below.
“1.1 Trk receptors
As one of the largest family of proteins encoded by the human genome, protein kinases are the central
regulators of signal transduction as well as control of various complex cell processes. Receptor ne
kinases (RTKs) are a ily of protein kinases (up to 100 members) bound to the cell membrane that
ically act on the tyrosine residues of proteins. One small group within this subfamily is the Trk
kinases, with three highly homologous isoforms: TrkA, TrkB, and TrkC. All three isoforms are activated by
high affinity growth factors named neurotrophins (NT): i) nerve growth factor (NGF), which activates TrkA;
ii) derived neurotrophic factor (BDNF) and NT-4/5, which te TrkB; and iii) NT-3, which
activates TrkC. The binding of neurotrophins to the extracellular domain of Trks causes the Trk kinase to
autophosphorylate at several intracellular tyrosine sites and rs downstream signal transduction
pathways. Trks and neurotrophins are well known for their effects on neuronal growth and survival.
1.2 Trks and cancer
Originally isolated from neuronal tissues, Trks were thought to mainly affect the nance and survival
of neuronal cells. However, in the past 20 years, increasing ce has suggested that Trks play key
roles in malignant transformation, chemotaxis, metastasis, and survival signaling in human tumors. The
association n Trks and cancer focused on prostate cancer in r years and the topic has been
reviewed. For example, it was reported that malignant prostate epithelial cells secrete a series of
neurotrophins and at least one Trks. In pancreatic cancer, it was proposed that paracrine and/or autocrine
rophin-Trk interactions may influence the invasive behavior of the cancer. TrkB was also reported to
be overexpressed in metastatic human atic cancer cells. Recently, there have been a number of
new findings in other cancer settings. For example, a translocation leads to expression of a fusion protein
derived from the N-terminus of the ETV6 ription factor and the C-terminal kinase domain of TrkC.
40 The resulting ETV6-TrkC fusions are oncogenic in vitro and appear causative in secretory breast
carcinoma and some acute myelogenous leukemias (AML). Constitutively active TrkA fusions occurred in
a subset of papillary thyroid cancers and colon carcinomas. In neuroblastoma, TrkB expression was
reported to be a strong predictor of aggressive tumor growth and poor sis, and TrkB
overexpression was also associated with increased resistance to chemotherapy in neuroblastoma tumor
cells in vitro. One report showed that a novel splice variant of TrkA called l signaled in the e
of neurotrophins through the inositol phosphate—AKT y in a subset of neuroblastoma. Also,
mutational analysis of the tyrosine kinome revealed that Trk mutations occurred in colorectal and lung
cancers. In summary, Trks have been linked to a y of human cancers, and discovering a Trk inhibitor
and testing it clinically might provide further insight to the biological and l hypothesis of treating
cancer with targeted therapies.
1.3 Trks and pain
Besides the newly developed ation with cancer, Trks are also being recognized as an important
mediator of pain sensation. Congenital insensitivity to pain with anhidrosis (CIPA) is a disorder of the
peripheral nerves (and normally innervated sweat glands) that prevents the patient from either being able
to adequately perceive painful stimuli or to sweat. TrkA s have been shown to cause CIPA in
various ethnic groups.
Currently, eroidal anti-inflammatory drugs (NSAIDs) and opiates have low efficacy and/or side
effects (e.g., gastrointestinal/renal and psychotropic side s, respectively) against neuropathic pain
and therefore development of novel pain treatments is highly desired. It has been recognized that NGF
levels are elevated in response to chronic pain, injury and inflammation and the administration of
ous NGF increases pain hypersensitivity. In addition, inhibition of NGF function with either anti-
NGF antibodies or non-selective small molecule Trk inhibitors has been shown to have effects on pain in
animal models. It appears that a selective Trk inhibitor (inhibiting at least NGF’s target, the TrkA receptor)
might provide clinical benefit for the treatment of pain. ent earlier reviews have covered targeting
NGF/BDNF for the treatment of pain so this review will only focus on small le Trk kinase inhibitors
claimed against cancer and pain. However, it is notable that the NGF antibody tanezumab was very
recently reported to show good efficacy in a Phase II trial against osteoarthritic knee pain.”
International Patent Application ation number W02009/012283 refers to various fluorophenyl
compounds as Trk inhibitors; International Patent Application publication numbers W02009/152087,
W02008/080015 and W02008/08001 and W02009/152083 refer to various fused pyrroles as kinase
modulators; International Patent Application publication numbers /143024 and W02009/143018
refer to various pyrrolo[2,3-d]pyrimidines substituted as Trk inhibitors; International Patent Application
publication s W02004/056830 and W02005/116035 describe various 4-amino-pyrrolo[2,3-
d]pyrimidines as Trk inhibitors. International Patent Application publication number W02011/133637
bes various pyrrolo[2,3-d]pyrimidines and pyrrolo[2,3-b]pyridines as inhibitors of various kinases.
US ional application U861/471758 was filed 5th April 2012 and the whole contents of that application
in it’s entirety are herewith included by reference thereto.
Thus Trk tors have a wide variety of potential medical uses. There is a need to provide new Trk
40 inhibitors that are good drug ates. In particular, compounds should preferably bind potently to the
Trk receptors in a ive manner compared to other receptors, whilst showing little affinity for other
receptors, including other kinase and / or GPC receptors, and show functional ty as Trk receptor
antagonists. They should be non-toxic and demonstrate few side-effects. Furthermore, the ideal drug
candidate will exist in a physical form that is stable, groscopic and easily formulated. They should
preferably be e.g. well absorbed from the gastrointestinal tract, and / or be injectable directly into the
bloodstream, muscle, or subcutaneously, and / or be metabolically stable and possess favourable
pharmacokinetic properties.
Among the aims of this invention are to provide orally-active, efficacious, compounds and salts which can
be used as active drug nces, particularly Trk antagonists, i.e. that block the intracellular kinase
activity of the Trk, e.g. TrkA (NGF) receptor. Other desirable features include good HLM/hepatocyte
stability, oral bioavailability, metabolic stability, absorption, selectivity over other types of kinase, dofetilide
selectivity. Preferable nds and salts will show a lack of CYP inhibition/induction, and be CNS-
spanng.
W
The present invention provides compounds of Formula I:
and pharmaceutically acceptable salts thereof wherein the subsituents are defined below.
The ion also comprises pharmaceutical compositions comprising a therapeutically effective amount
of a compound of formula I as defined herein, or a pharmaceutically acceptable salt f, and a
pharmaceutically able carrier.
The invention is also directed to a method of treating a disease or condition indicated for ent with a
Trk antagonist, in a subject, by administering to a subject in need thereof a therapeutically effective
amount of one or more of the compounds herein, or a pharmaceutically acceptable salt thereof.
Other aspects of the invention will be nt from the remaining description and claims.
Preferably, the compounds of the present invention are potent antagonists at Trk receptors, and have a
suitable PK profile to enable once daily dosing.
The compounds of the present invention are potentially useful in the treatment of a range of disorders
where a Trk antagonist is indicated, particularly pain indications. Depending on the disease and ion
of the t, the term “treatment” as used herein may include one or more of curative, palliative and
prophylactic treatment.
According to the invention a compound of the t invention may be useful to treat any physiological
pain such as inflammatory pain, nociceptive pain, neuropathic pain, acute pain, chronic pain, musculo-
skeletal pain, on-going pain, central pain, heart and vascular pain, head pain, orofacial pain. Other pain
conditions which may be treated include intense acute pain and chronic pain conditions which may involve
the same pain pathways driven by pathophysiological processes and as such cease to provide a
protective mechanism and instead contribute to debilitating symptoms associated with a wide range of
e states.
Pain is a feature of many trauma and e states. When a substantial injury, via disease or trauma, to
body tissue occurs the characteristics of nociceptor activation are altered, this leads to hypersensitivity at
the site of damage and in nearby normal tissue. In acute pain the sensitivity returns to normal once the
injury has healed. However, in many chronic pain states, the ensitivity far outlasts the healing
process and is normally due to s system injury due to maladaptation of the afferent fibres (Woolf &
Salter 2000 Science 288: 1765-1768). Clinical pain is present when discomfort and abnormal sensitivity
feature among the patient’s symptoms. There are a number of typical pain subtypes: 1) spontaneous pain
which may be dull, burning, or stabbing; 2) pain responses to noxious stimuli are exaggerated
(hyperalgesia); 3) pain is ed by normally innocuous stimuli (allodynia) (Meyer et al., 1994 Textbook
of Pain . Pain can be d into a number of different areas because of differing hysiology,
these include nociceptive, inflammatory, neuropathic pain among others. It should be noted that some
types of pain have multiple aetiologies and thus can be classified in more than one area, e.g. Back pain,
Cancer pain have both nociceptive and neuropathic components.
NOCICEPTIVE PAIN
Nociceptive pain is induced by tissue injury or by intense stimuli with the potential to cause injury. Pain
afferents are ted by transduction of stimuli by nociceptors at the site of injury and sensitise the
spinal cord at the level of their termination. This is then d up the spinal tracts to the brain where
pain is perceived (Meyer et al., 1994 Textbook of Pain 13-44). The activation of nociceptors activates two
types of nt nerve fibres. Myelinated A—delta fibres transmit rapidly and are responsible for the sharp
and stabbing pain ions, whilst unmyelinated C fibres transmit at a slower rate and convey the dull
or aching pain. Moderate to severe acute nociceptive pain is a prominent feature of, but is not limited to
40 pain from strains/sprains, post-operative pain (pain following any type of surgical procedure),
posttraumatic pain, burns, myocardial infarction, acute pancreatitis, and renal colic. Also cancer related
acute pain syndromes commonly due to therapeutic interactions such as chemotherapy toxicity,
immunotherapy, hormonal therapy and radiotherapy. Moderate to severe acute nociceptive pain is a
prominent feature of, but is not limited to, cancer pain which may be tumour related pain, (e.g. bone pain,
headache and facial pain, viscera pain) or associated with cancer therapy (e.g. postchemotherapy
syndromes, chronic postsurgical pain syndromes, post radiation syndromes), back pain which may be due
to herniated or ruptured intervertabral discs or abnormalities of the lumbar facet joints, sacroiliac joints,
paraspinal muscles or the posterior longitudinal ligament.
NEUROPATHIC PAIN
According to the invention a compound of the present invention can potentially be used to treat
neuropathic pain and the symptoms of neuropathic pain including hyperalgesia, allodynia and ongoing
pain. Neuropathic pain is defined as pain initiated or caused by a primary lesion or dysfunction in the
nervous system (IASP definition). Nerve damage can be caused by trauma and disease and thus the
term ‘neuropathic pain’ encompasses many disorders with diverse aetiologies. These include but are not
limited to, Diabetic neuropathy, Post herpetic neuralgia, Back pain, Cancer neuropathy, HIV athy,
Phantom limb pain, Carpal Tunnel Syndrome, c alcoholism, hypothyroidism, trigeminal gia,
uremia, or vitamin deficiencies. Neuropathic pain is pathological as it has no protective role. It is often
t well after the original cause has ated, commonly lasting for years, significantly decreasing a
patients y of life (Woolf and Mannion 1999 Lancet 353: 1959-1964). The ms of neuropathic
pain are difficult to treat, as they are often heterogeneous even n patients with the same disease
(Woolf & Decosterd 1999 Pain Supp. 6: 8141-8147; Woolf and Mannion 1999 Lancet 353: 1959-1964).
They include spontaneous pain, which can be continuous, or paroxysmal and abnormal evoked pain, such
as hyperalgesia (increased sensitivity to a noxious stimulus) and allodynia tivity to a normally
ous stimulus).
E ACUTE PAIN AND CHRONIC PAIN
Intense acute pain and chronic pain may involve the same pathways driven by hysiological
processes and as such cease to provide a tive mechanism and instead contribute to debilitating
symptoms associated with a wide range of disease states. Pain is a feature of many trauma and disease
. When a substantial injury, via disease or , to body tissue occurs the characteristics of
nociceptor activation are altered. There is isation in the periphery, locally around the injury and
centrally where the nociceptors terminate. This leads to hypersensitivity at the site of damage and in
nearby normal tissue. In acute pain these mechanisms can be useful and allow for the repair ses
to take place and the hypersensitivity returns to normal once the injury has healed. However, in many
chronic pain states, the hypersensitivity far outlasts the healing process and is normally due to nervous
system injury. This injury often leads to maladaptation of the afferent fibres (Woolf & Salter 2000 Science
288: 1765-1768). Clinical pain is present when discomfort and abnormal sensitivity feature among the
t’s symptoms. Patients tend to be quite heterogeneous and may present with s pain
40 symptoms. There are a number of typical pain subtypes: 1) spontaneous pain which may be dull, burning,
or stabbing; 2) exaggerated pain ses to noxious stimuli (hyperalgesia); 3) pain is produced by
normally innocuous stimuli (allodynia) (Meyer et al., 1994 Textbook of Pain 13-44). Although patients with
back pain, arthritis pain, CNS trauma, or athic pain may have r symptoms, the underlying
mechanisms are different and, therefore, may require different treatment strategies.
CHRONIC PAIN
Chronic pain comprises one or more of, chronic nociceptive pain, chronic athic pain, chronic
inflammatory pain, breakthrough pain, persistent pain hyperalgesia, allodynia, central isation,
peripheral sensitisation, disinhibition and augmented facilitation.
Chronic pain includes cancer pain, e.g. cancer pain arising from malignancy, adenocarcinoma in glandular
tissue, blastoma in embryonic tissue of organs, oma in epithelial , leukemia in tissues that
form blood cells, lymphoma in lymphatic tissue, a in bone marrow, sarcoma in connective or
supportive tissue, adrenal cancer, AIDS-related lymphoma, anemia, bladder cancer, bone , brain
cancer, breast cancer, carcinoid tumour s, cervical cancer, chemotherapy, colon cancer, cytopenia, ,
endometrial cancer, esophageal cancer, gastric cancer, head cancer, neck cancer, hepatobiliary cancer,
kidney cancer, leukemia, liver cancer, lung cancer, lymphoma, Hodgkin's disease, lymphoma, non-
Hodgkin's, nervous system tumours, oral cancer, n cancer, pancreatic cancer, prostate cancer,
rectal cancer, skin cancer, stomach cancer, ular cancer, thyroid cancer, urethral cancer, bone
cancer, sarcomas cancer of the connective tissue, cancer of bone tissue, cancer of blood-forming cells,
cancer of bone marrow, multiple a, leukaemia, primary or secondary bone cancer, tumours that
metastasize to the bone, tumours infiltrating the nerve and hollow viscus, tumours near neural structures.
Cancer pain also comprises visceral pain, e.g. al pain which arises from pancreatic cancer and/or
metastases in the abdomen, somatic pain, e.g. somatic pain due to one or more of bone cancer,
metastasis in the bone, postsurgical pain, sarcomas cancer of the connective tissue, cancer of bone
tissue, cancer of blood-forming cells of the bone marrow, multiple myeloma, leukaemia, primary or
secondary bone cancer.
INFLAMMATORY PAIN
Inflammatory conditions include acute inflammation, persistent acute inflammation, chronic inflammation,
and combined acute and chronic inflammation.
Inflammatory pain includes acute inflammatory pain and/or chronic inflammatory pain wherein the chronic
inflammatory pain can be pain involving both eral and central isation and/or mixed etiology
pain involving both inflammatory pain and athic pain or ptive pain components.
matory pain also comprises hyperalgesia, e.g. primary and/or secondary hyperalgesia. Additionally
or atively the inflammatory pain can e allodynia. Inflammatory pain also comprises pain that
persists beyond tion of an underlying disorder or inflammatory condition or healing of an injury.
Inflammatory pain is pain resulting an inflammatory condition. e.g. in response to acute tissue injury due
40 to trauma, disease e.g. an inflammatory disease, immune on, the ce of foreign substances,
chemicals or infective particles for example micro-organisms. Inflammatory conditions can be either acute
or chronic inflammation or both.
Inflammatory pain can result from an inflammatory condition due to an inflammatory e such as
matoryjoint diseases, inflammatory connective tissue diseases, inflammatory autoimmune
diseases, inflammatory myopathies, matory digestive system diseases, matory air way
es, cellular immune inflammation diseases, hypersensitivities and allergies, vasular inflammation
diseases, non-immune inflammatory disease, synovitis, villonodular synovitis, arthralgias, ankylosing
spondylitis, spondyloarthritis, spondyloarthropathy, gout, Pagets disease, periarticular disorders such as
bursitis, rheumatoid disease, rheumatoid arthritis and osteoarthritis, rheumatoid arthritis or osteoarthritis.
Rheumatoid arthritis in particular, represents ongoing inflammation associated with severe pain. Arthritic
pain is a form of inflammatory pain and arises from inflammation in a joint which causes both peripheral
sensitization and central sensitization. Under inflammatory conditions the nociceptive system is activated
by normally innocuous and nful mechanical stimuli. Additionally when the joint is at rest pain is
present and appears as spontaneous pain and hyperalgesia (augmented pain response on noxious
stimulation and pain on normally nonpainful stimulation). Inflammatory processes in peripheral tissues
lead to central sensitization in the spinal cord, which contributes to hyperalgesia and allodynia lly
associated with inflammatory pain. Other types of inflammatory pain include inflammatory bowel diseases
(IBD).
OTHER TYPES OF PAIN
Other types of pain include but are not limited to:
- Musculo-skeletal disorders including but not limited to a, fibromyalgia, litis, sero-negative
(non-rheumatoid) pathies, non-articular rheumatism, dystrophinopathy, Glycogenolysis,
polymyositis, sitis;
- Central pain or ‘thalamic pain’ as defined by pain caused by lesion or dysfunction of the nervous system
including but not limited to central post-stroke pain, multiple sclerosis, spinal cord injury, Parkinson’s
disease and epilepsy;
- Heart and vascular pain including but not limited to angina, myocardical infarction, mitral is,
pericarditis, Raynaud’s phenomenon, doma, scleredoma, skeletal muscle ischemia;
- al pain, and gastrointestinal disorders. The viscera encompasses the organs of the abdominal
cavity. These organs include the sex organs, spleen and part of the digestive system. Pain associated
with the viscera can be divided into digestive visceral pain and non-digestive visceral pain. Commonly
tered gastrointestinal (GI) disorders include the functional bowel disorders (FBD) and the
inflammatory bowel diseases (IBD). These GI disorders e a wide range of e states that are
tly only moderately controlled, including — for FBD, gastro-esophageal reflux, dyspepsia, the
irritable bowel syndrome (IBS) and onal abdominal pain syndrome (FAPS), and — for IBD, Crohn’s
disease, ileitis, and ulcerative colitis, which all regularly produce visceral pain. Other types of visceral pain
include the pain ated with dysmenorrhea, pelvic pain, cystitis and pancreatitis;
Head pain including but not limited to ne, migraine with aura, migraine without aura cluster
headache, tension-type headache. Orofacial pain including but not limited to dental pain,
temporomandibular myofascial pain, tinnitus, hot flushes, ss leg syndrome and blocking
development of abuse potential. r pain conditions may include, back pain (e.g. chronic lower back
pain), cancer pain, complex regional syndrome, HIV-related neuropathic pain, post-operative induced
neuropathic pain, post-stroke pain, spinal cord injury pain, tic nerve injury pain, diabetic peripheral
neuropathy, moderate / severe interstitial cystitis pain, irritable bowel syndrome pain, moderate / severe
endometriosis pain, moderate / severe pelvic pain, moderate / severe prostatitis pain, moderate / severe
osteoarthritis ost-herpetic neuralgia, rheumatoid arthritis pain, dysmenorrhea pain, pre-emptive
post-operative pain, trigeminal neuralgia, bursitis, dental pain, fibromyalgia or myofacial pain, menstrual
pain, migraine, athic pain (including painful diabetic neuropathy), pain associated with post-
herpetic neuralgia, post-operative pain, ed pain, trigeminal gia, visceral pain (including
interstitial cystitis and IBS) and pain associated with AIDS, nia, burns, cancer, hyperalgesia,
hypersensitisation, spinal trauma and/or degeneration and stroke.
DETAILED DESCRIPTION
Emodiment 1 of the invention is a compound of Formula I:
(l)
or a pharmaceutically acceptable salt f, wherein
R1 is
H, or
C1_5 alkyl optionally substituted by up to 3 substituents ndently selected from OH, CON(R5R6),
SOZR7, SR7, 0R7, CHZOH, COZR5, SONR7R7, NR7SOZR5, CN, N02 and R8 or
a ring system selected from C3_5 cycloalkyl, propellanyl, or a 4-6 membered saturated heterocyclyl
ring, which ring system has up to 3 ring hetero-atoms selected from N, O and S, and which ring
system is optionally substituted by up to 3 substituents independently selected from methyl, OH,
CON(R5R6), sozR7, 0R7, CHZOH, COZR5, SONR7R7, R5, CN, N02 and R8;
R2 is H or methyl;
R3 is H, NH2 or NH(C1_3 alkyl optionally substituted with up to 3 substituents independently selected from
OH and O(C1_3 alkyl));
R101 is H, OH, methyl, cyclopropyl, methoxy, ethyl, ethoxy or CN,
x is a bond, 0, (CH-R4)n, NR1“, OCH2 or CHZO;
R4 is ndently H, CH3, CHZOH, CHZOCHg, OH, NH2, NHCH3, N(CH3)2, CHZNHZ, CHZNHCH3,Or
CH2N(CH3)2,
R104 is H, C1_3 alkyl or a C4_6 ted carbocycle, each of which is optionally substituted by up to 3
substituents independently selected from C1_3 alkyl, CHZOH and NH2;
nis1or2;
R102 is a ring system which is a 3-7 membered monocyclic carbocyclic or heterocyclic system, or an 8
ed bicyclic system, which ring system may be saturated or partially or fully unsaturated, wherein
the heterocyclic ring system may have up to 5 ring hetero-atoms selected from N, S, and 0,
wherein the ic ring system can be 2 rings (carbocyclic-carbocyclic, carbocyclic-heterocyclic,
heterocyclic-carbocyclic or heterocyclic-heterocyclic) fused or linked by a single bond,
which ring system is optionally substituted by up to 3 substituents independently selected from, where
le -
halo, CN, NR5R6, SOZR7, SR7, C1_4 alkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy groups,
C3_6 cycloalkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy groups, C1_3 alkyl substituted by up
to 3 halogen, OH, O(C1_3 alkyl), O(C3_6 cycloalkyl ally substituted by up to 3 OH and/or C1_3 alkoxy
groups, O(C1_3 alkyl substituted by up to 3 halogen), O(C1_3 alkyl substituted by up to 3 OH and/or C1_3
alkoxy groups), NR5SOZR7, :0, R8, C(O)R8, N02, NR5COZR7, NR5COR7,OR8, S(O)R7, and CH2R8;
R5and R6 are each independently
H, or
C1_5 alkyl optionally tuted by up to 3 substituents independently selected from OH, CONR7R7,
sozR7, 0R7, CHZOH, COZR7, SONR7R7, NR7SOZR7, CN, N02 and R9 ,
a ring system selected from 03.5 cycloalkyl, propellanyl, or a 4-6 membered saturated heterocyclyl ring,
which ring system is optionally substituted by up to 3 substituents independently selected from OH,
CON(R7R7), SOZR7, COZR7, SONR7R7, R7, CN, N02, halo, NR7R7,SR7,C1_4 alkyl ally
substituted by up to 3 OH and/or C1_3 alkoxy groups, C3_6 cycloalkyl optionally substituted by up to 3 OH
and/or C1_3 alkoxy , C1_3 alkyl substituted by 1 to 3 halogen, O(C3_6 cycloalkyl optionally substituted
W0 2012/137089 10
by up to 3 OH and/or C1_3 alkoxy groups, O(C1_3 alkyl substituted by up to 3 halogen, O(C1_3 alkyl
substituted by up to 3 OH and/or c1.3 alkoxy, NR7SOZR7,=O,NOZ,NR7COZR7, and S(O)R7
or R5 and R6 together with the N to which they are attached can be a 4-7 membered ring ally
including up to 2 further ring hetero-atoms independently selected from N, O, S, which ring is optionally
substituted by C1_3 alkoxy and / or C1_3 alkyl;
R7 is H, c,_5 alkyl or c,_5 alkoxy,
which C1_5 alkyl or C1_5 alkoxy is optionally substituted by up to 3 substituents independently selected from
halogen;
R8 is a is a ring system which is a 3-7 membered monocyclic carbocyclic or heterocyclic system, or an 8-
14-membered bicyclic system, which ring system may be saturated or lly or fully unsaturated,
wherein the heterocyclic ring system may have up to 5 ring hetero-atoms selected from N, S, and 0,
wherein the bicyclic ring system can be 2 rings (carbocyclic-carbocyclic, carbocyclic-heterocyclic,
cyclic-carbocyclic or heterocyclic-heterocyclic) fused or linked by a single bond,
which ring system is optionally substituted by up to 3 substituents independently selected from, where
possible -
halo, CN, NR5R6, SOZR7, SR7, C1_4 alkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy groups,
C3_6 cycloalkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy groups, C1_3 alkyl substituted by 1
to 3 halogen, OH, O(C1_3 alkyl), O(C3_6 cycloalkyl ally substituted by up to 3 OH and/or C1_3 alkoxy
groups, O(C1_3 alkyl substituted by up to 3 halogen, O(C1_3 alkyl tuted by up to 3 OH and/or C1_3
alkoxy, NR5SOZR7, :0, N02, NR7COR7,NR5COZR7, and S(O)R7;
R9 is a is a ring system which is a 3-7 membered monocyclic yclic or heterocyclic system, or an 8-
14-membered bicyclic system, which ring system may be saturated or lly or fully unsaturated,
n the heterocyclic ring system may have up to 5 ring hetero-atoms selected from N, S, and 0,
wherein the bicyclic ring system can be 2 rings (carbocyclic-carbocyclic, carbocyclic-heterocyclic,
heterocyclic-carbocyclic or heterocyclic-heterocyclic) fused or linked by a single bond,
which ring system is optionally tuted by up to 3 substituents independently selected from, where
possible -
halo, CN, NR7R7, SOZR7, SR7, C1_4 alkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy ,
C3_6 cycloalkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy groups, C1_3 alkyl substituted by 1
to 3 halogen, OH, O(C1_3 alkyl), O(C3_6 cycloalkyl optionally substituted by up to 3 OH and/or C1_3 alkoxy
, O(C1_3 alkyl substituted by up to 3 n, O(C1_3 alkyl substituted by up to 3 OH and/or C1_3
alkoxy, NR7SOZR7, :0, N02, NR7COZR7, NR7COR7,and S(O)R7;
wherein each CH moiety can be replaced by a CF moiety.
Embodiment 2 of the ion is a compound or salt according to embodiment 1 wherein R1 is H, C1_5
40 alkyl optionally substituted by up to 2 OH,
W0 2012/137089 11
or R1 is 01.5 alkyl substituted by CONHZ, CONHCH3, CON(CH3)2, COZH, COZCH3, OCH3, SCH3, SOZCH3,
or R1 is a ring system selected from 03.5 cycloalkyl, propellanyl, or oxetanyl, which ring system is
ally substituted by methyl, OH or CHZOH.
Embodiment 3 of the invention is a nd or salt according to any one of embodiments 1 or 2 wherein
R1 is t-butyl, hydroxy—t-butyl, dihdyroxy—t-butyl, 1-hydroxypropyl or 1,3-dihydroxypropyl.
Embodiemnt 4 of the invention is a nd or salt according to any one of ments 1 to 3
wherein R2 is H.
Embodiemt 5 of the invention is a compound or salt according to any one of embodiments 1 to 4 wherein
R3 is H or NHZ.
ment 6 of the invention is a compound or salt ing to any one of embodiments 1 to 5 wherein
R3 is NH2.
Embodiment 7 ofthc invention is a compound or salt according to any one of embodiments 1 to 5
wherein R3 is H.
Embodiment 8 of the invention is a compound or salt according to any one of ments 1 to 7 wherein
R101 is H.
Embodiment 9 of the invention is a compound or salt ing to any one of embodiments 1 to 7 wherein
R101 is OH.
Embodiment 10 of the invention is a compound or salt according to any one of embodiments 1 to 9
wherein X is a bond, 0, CH2, C2H4, CH(CH3)CH2, CH(CH3), CH(CHZOH), CHZO, CH(NHZ), CH(OH) or NH.
Embodiment 11 of the invention is a compound or salt ing to any one of embodiments 1 to 10
wherein X is CH2.
Embodiment 12 of the invention is a compound or salt according to any one of embodiments 1 to 11
wherein R -
is an optionally substituted nitrogen-containing ring system which is linked to the X moiety
via a nitrogen ring atom.
Embodiment 13 of the invention is a compound or salt according to any one of embodiments 1 to 11
wherein R is an optionally substituted ring system where the ring system is selected from -
benzimidazolyl, benzisoxazolyl, benzofuranyl, azolyl, benzotriazolyl, biphenyl, bipyrazolyl,
cinnolinyl, cyclobutylimidazolyl, cyclobutylpyrazolyl, cyclobutylthiazolyl, cyclopentyltriazolyl,
40 cyclopropylisoxazolyl, cyclopropyloxazolyl, cyclopropylpyrazolyl, cyclopropyltriazolyl, diazirenylphenyl,
dihydronaphthyridinyl, dihydropyrrolopyrazolyl, dioxinopyridinyl, furazanyl, furopyridinyl, furopyrrolyl,
W0 2012/137089 12
olyl, imidazopyrazinyl, imidazopyridazinyl, imidazopyridinyl, imidazopyrimidinyl, imidazothiadiazolyl,
imidazothiazolyl, indanyl, indazolyl, indolyl, isoindolyl, isoxazolopyridinyl, isoxazolyl, isoquinolinyl,
naphthyridinyl, oxazolyl, , phenylcyclopropyl, phenylimidazolyl, phenylpyrazolyl, phenylpyrrolyl,
phenyltetrazolyl, phthalazinyl, purinyl, pyrazinyl, pyrazolyl, pyrazolopyridinyl, pyrazolopyrimidinyl,
lotriazinyl, pyridinyl, pyridazinyl, pyridinyltriazolyl, pyrimidinyl, pyrroloimidazolyl, pyrrolopyrazinyl,
pyrrolopyrimidinyl, pyrrolopyridinyl, pyrrolyl, quinolinyl, quinazolyl, alinyl, tetrahydrobenzisoxazolyl,
ydrocyclopentapyrazolyl, tetrahydrotriazolopyridinyl, tetrazolopyridazinyl, tetrazolopyridinyl,
thiazolyl, thiazolopyridinyl, thiazolopyrimidinyl, thienylpyrazolyl, thienopyridinyl, lopyridinyl and
triazolyl,
Embodiment 14 of the invention is a compound or salt according to embodiment 13 where the optional
substituents are independently selected from, where possible -
halo, methyl, ethyl, propyl, isopropyl, cyclopropyl, CF3, CHFZ, CHZF, CHZOCH3, CN, CHZOH, OCH3, =0,
NH2, SCH3, SOZCH3, phenoxy, fluorophenoxy, benzyl, SCF3, OCF3, SOZCF3, NHSOZCH3, NH802CF3,
C(O)CF3, C(O)CH3, benzoyl, azetidinylmethyl, fluoroazetidinylmethyl and morpholinomethyl.
ment 15 of the invention is a compound or salt according to any one of embodiments 1 to 11, 13
102-
or 14, wherein R IS selected from phenyl, pyrazolyl, 1,2,3-triazolyl, benzotriazolyl, nyl,
pyridinyl and pyridinyl,
each of which is optionally substituted by halo, methyl, ethyl, propyl, isopropyl, cyclopropyl, CF3, CHFZ,
CHZF, CH20CH3, CN, CHZOH, OCH3, =0, NH2, SCH3, SOZCH3, phenoxy, fluorophenoxy, benzyl, SCF3,
OCF3, 802CF3, NH802CH3, NHSOZCF3, C(O)CF3, C(O)CH3, benzoyl, azetidinylmethyl,
fluoroazetidinylmethyl and/or morpholinomethyl.
Embodiment 16 of the invention is a nd or salt according to any one of embodiments 1 to 15 with
R5 and R6 groups present, wherein R5 and R6 are each independently H, C1_3 alkyl optionally substituted
by C1_3 alkoxy, C3_5 cycloalkyl, propellanyl, yl, tetrahydrofuranyl or l,
or R5 and R6 together with the N to which they are attached can be an azetidine, pyrrolidine, piperidine,
piperazine or line ring, which ring is optionally substituted by C1_3 alkoxy and / or C1_3 alkyl.
Embodiment 17 of the invention is a compound according to embodiment 1 of the Formula IA:
W0 2012/137089 13
(IA)
or a pharmaceutically acceptable salt thereof, wherein
R3 is H or NHZ;
R1 is C2_4 alkyl optionally substituted by 1 or 2 OH groups;
R101 is H or OH;
and R102 is phenyl or an aromatic or partially unsaturated 5- or ered heterocycle, which
cycle is optionally fused to a further phenyl or 5-7 membered aromatic or partially unsaturated
heterocyclic ring, wherein each heterocycle has from 1 to 3 ring heteroatoms selected from N, O and S,
and which ring system is ally substituted by up to 3 substituents independently selected from
halo, CF3, C1_4 alkyl and 03.5 lkyl.
Embodiment 18 of the invention is a compound or salt according to embodiment 17 n R101 is H.
Embodiment 19 of the invention is a nd or salt according to embodiment 18 wherein
R1 is t-butyl, hydroxy—t-butyl or 1-hydroxypropyl;
and R102 is 4-trifluromethylphenyl, 4-chlorophenyl, 2,4-difluorophenyl, 5-chloropyridinyl, 5-fluoropyridin-
2-yl, 3-trifluromethylpyrazolylyl, 4-trifluromethylpyrazolyl, 3-trifluromethylmethylpyrazoly|,
3-cyclopropylpyrazolyl, opropylpyrazolyl, 4-trifluromethyl (1 ,2,3-triazolyl), 4-cyclopropyl-
(1 ,2,3-triazolyl), or benzotriazolyl.
Embodiment 20 of the invention is a compound according to embodiment 1, selected from:
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridinyl)—2-[4-
(trifluoromethyl)phenyl]acetamide;
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin
yl)(4-chlorophenyl)acetamide;
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin
yl)(5-fluoropyridinyl)acetamide;
W0 2012/137089 14
N-(5-{[2-amino—7-(2-hydroxy—1,1-dimethylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridin
yl)[3-(trif|uoromethy|)-1H-pyrazoIy|]acetamide;
N-(5-{[2-amino—7-(2-hydroxy—1,1-dimethylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridin
y|)(3-cyc|opropyI-1H-pyrazoIy|)acetamide;
N-{5-[(2-amino—7-tert-butyI-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(4-cyc|opropy|—
1H-1,2,3-triazoly|)acetamide;
N-{5-[(2-amino—7-tert-butyI-7H-pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridinyI}[4-
(trifluoromethyI)-1H-pyrazoIy|]acetamide;
N-{5-[(2-amino—7-tert-butyI-7H-pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridinyI}[4-
(trifluoromethy|)-1H-1,2,3-triazo|—1-y|]acetamide;
N-{5-[(2-amino—7-tert-butyl-7H-pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridiny|}(5-ch|oropyridin-
cetamide;
N-(5-{[2-Amino—7-(2-hydroxy—1,1-dimethylethyl)—7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridin
yl )(5-ch|oropyrid inyl)acetamide;
N-{5-[(7-tert-butyI-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridiny|}[4-(trif|uoromethy|)-1H-
1,2,3-triazo|—1-y|]acetamide;
2-(4-chlorophenyl)-N-[5-({7-[(1S)—2-hydroxy—1-methylethyI]-7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyrid inyl]acetamide
(7-tert-butyI-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridiny|}[4-(trif|uoromethy|)-1H-
pyrazoIyl]acetamide;
N-[5-({7-[(1S)—2-Hydroxy—1-methy|ethyl]—7H-pyrro|o[2,3-d]pyrimidinyl}carbonyl)pyridinyl]—2-[4-
(trifluoromethyl)phenyl]acetamide;
N-[5-({7-[(1R)—2-hydroxy—1-methylethyI]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyI][4-
(trifluoromethyl)phenyl]acetamide;
2-(4-chlorophenyl)-N-[5-({7-[(1R)—2-hydroxy—1-methylethyI]-7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyrid inyl]acetamide;
N-(5-{[7-(2-hydroxy—1,1-dimethylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridinyl)—2-[5-
methyl(trifluoromethyl)-1H-pyrazoIy|]acetamide;
2-(5-chloropyridinyl)-N-(5-{[7-(2-hydroxy—1,1-dimethylethyl)-7H-pyrro|o[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)acetam ide;
[2-Amino—7-(2-hydroxy—1-methy|ethy|)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridiny|)-
2-(4-chlorophenyl)acetamide;
N-(5-{[2-amino—7-(2-hydroxy—1-methylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridinyl)—2-
[4-(trifluoromethyl)phenyl]acetamide;
N-(5-{[2-amino—7-(2-hydroxy—1-methylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridinyl)—2-
(4-chlorophenyl)acetamide;
N-(5-{[2-amino—7-(2-hydroxy—1-methylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridinyl)—2-
[4-(trifluoromethyl)phenyl]acetamide;
N-(5-{[2-Amino—7-(2-hydroxy—1,1-dimethylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]carbonyl}pyridin
40 yl)[5-methyl(trifluoromethyl)-1H-pyrazoIy|]acetamide;
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(4-cyclopropyl-1H-1,2,3-
triazolyl)acetamide;
or a pharmaceutically acceptable salt thereof.
Embodiment 21 of the invention is a pharmaceutical ition comprising a compound of the
formula (I) or a pharmaceutically acceptable salt thereof, as defined in any one of the preceding
embodiments 1 to 20, and a pharmaceutically acceptable carrier.
Embodiment 22 of the invention is a compound of the formula (I) or a pharmaceutically acceptable
salt thereof, as defined in any one of embodiments 1 to 20, for use as a medicament.
Embodiment 23 of the invention is a compound of formula (I) or a pharmaceutically acceptable salt
thereof, as defined in any one of embodiments 1 to 20 for use in the treatment of a disease for which
an Trk receptor antagonist is indicated
ment 24 of the invention is a compound of formula (I) or a ceutically acceptable salt
thereof, as defined in any one of embodiments 1 to 20 for use in the treatment of pain.
Embodiment 25 of the invention is the use of a nd of the formula (I) or a pharmaceutically
acceptable salt or composition thereof, as defined in any one of ments 1 to 20, for the
manufacture of a medicament to treat a e for which a Trk receptor antagonist is indicated
Embodiment 26 of the invention is the use of a compound of the formula (I) or a ceutically
acceptable salt or ition thereof, as d in any one of embodiments 1 to 20, for the
manufacture of a medicament to treat pain.
Embodiment 27 of the invention is a method of treatment of a mammal, to treat a disease for which an
Trk receptor antagonist is indicated, comprising treating said mammal with an effective amount of a
compound of the formula (I) or a pharmaceutically able salt thereof, as defined in any one of
embodiments 1 to 20.
Embodiment 28 of the invention is a method of treatment of pain in a , comprising treating
said mammal with an effective amount of a compound of the formula (I) or a pharmaceutically
acceptable salt thereof, as defined in any one of embodiments 1 to 20.
Embodiment 29 of the invention is nd or salt according to any one of embodiments 1 to 20 for
use in a medical treatment in combination with a r drug susbtance.
Further embodiments of the invention include:
W0 2012/137089 16
Compounds or salts of a (I ) where R1 has a value as exemplified in the Examples below;
Compounds or salts of formula (I ) where X has a value as exemplified in the Examples below;
Compounds or salts of formula (I ) where R has a value as exemplified in the Examples below;
Compounds or salts of formula (I) where R1, R2, R3, R101, X and R102 have a value as exemplified in
the Examples below;
A compound selected from any one of the es below or a pharmaceutically acceptable salt
thereof; and
any novel intermediate compound herein disclosed.
Other embodiments may be envisaged based on the ption below.
“Halogen” means a fluoro, chloro, bromo or iodo group.
“Alkyl” groups, containing the requisite number of carbon atoms, can be unbranched or branched.
es of alkyl include methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, sec-butyl and t-butyl.
“Pharmaceutically acceptable salts” of the nds of formula I include the acid addition and base
addition salts (including disalts, hemisalts, etc.) thereof.
Suitable acid addition salts are formed from acids which form xic salts. Examples include the
acetate, aspartate, benzoate, besylate, onate/carbonate, bisulphate/sulphate, borate, camsylate,
citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate,
hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate,
e, te, mesylate, methylsulphate, ylate, 2-napsylate, nicotinate, e, orotate,
oxalate, palmitate, e, phosphate/hydrogen phosphate/dihydrogen phosphate, rate,
stearate, succinate, tartrate, tosylate and trifluoroacetate salts.
Suitable base addition salts are formed from bases which form non-toxic salts. Examples include the
ium, arginine, benzathine, calcium, choline, diethylamine, ine, glycine, lysine, magnesium,
meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
For a review on suitable salts, see “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by
Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
The compounds of the invention include compounds of formula I and salts thereof as hereinbefore
defined, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as
hereinafter d and isotopically-labelled compounds of formula I.
Unless otherwise specified, compounds of formula (I) containing one or more asymmetric carbon atoms
can exist as two or more stereoisomers. Where a compound of formula (I) contains for example, a keto or
guanidine group or an aromatic , tautomeric isomerism omerism’) can occur. It follows that a
single compound may exhibit more than one type of isomerism.
Included within the scope of the claimed compounds of the present invention are all stereoisomers,
geometric isomers and tautomeric forms of the compounds of formula (I), including compounds exhibiting
17 2012/051363
more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or
base addition salts wherein the counterion is optically active, for e, D-lactate or L-lysine, or
racemic, for e, DL-tartrate or DL-arginine.
es of types of ial tautomerisms shown by the compounds of the invention include
hydroxypyridine <:> pyridone; amide <:> hydroxyl-imine and keto <:> enol tautomersims:
H O OH
o OH
HO N\ o )L A
I NH__ N Ai __
‘— \
/ | l
Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for
example, chromatography and fractional crystallisation.
Conventional techniques for the preparation/isolation of individual enantiomers include chiral sis
from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or other
derivative) using, for e, chiral high pressure liquid chromatography (HPLC).
Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable lly active
compound, for example, an alcohol, or, in the case where the compound of formula (I) contains an acidic
or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine. The resulting diastereomeric
mixture may be separated by chromatography and/or fractional crystallization and one or both of the
diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled
person.
Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically—
enriched form using chromatography, typically HPLC, on a resin with an asymmetric stationary phase and
with a mobile phase ting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50%
isopropanol, typically from 2 to 20%, and from 0 to 5% of an alkylamine, typically 0.1% diethylamine.
Concentration of the eluate affords the enriched mixture.
Mixtures of stereoisomers may be separated by conventional techniques known to those skilled in the art.
[see, for example, ochemistry of c nds” by E L Eliel , New York, 1994).]
The present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula
(I) wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic
mass or mass number different from the atomic mass or mass number usually found in nature.
W0 2012/137089 18
Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of
hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 14C, chlorine, such as 36Cl, fluorine, such as
18F, 123
iodine, such as l and 125' nitrogen, such as 13N and 15N, oxygen, such as 15O, 17O and
, 18O,
phosphorus, such as 32P, and sulphur, such as 35S.
Certain isotopically-labelled compounds of formula (I), for example, those incorporating a radioactive
isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes m, i.e.
3H, and -14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation
and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages
resulting from greater metabolic stability, for example, increased in vivo ife or reduced dosage
requirements, and hence may be preferred in some circumstances.
Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron
on Topography (PET) studies for examining substrate receptor occupancy.
ically-labelled compounds of formula (I) can generally be prepared by conventional techniques
known to those skilled in the art or by processes analogous to those described in the accompanying
Examples and Preparations using an appropriate isotopically-labelled reagents in place of the non-
labelled reagent previously employed.
The routes below, including those mentioned in the Examples and Preparations, illustrate methods of
synthesising compounds of formula (I). The skilled person will appreciate that the compounds of the
ion, and ediates thereto, could be made by methods other than those ically described
, for example by adaptation of the methods described herein, for example by methods known in the
art. le guides to synthesis, functional group interconversions, use of protecting groups, etc., are for
example:“Comprehensive Organic Transformations” by RC , VCH Publishers Inc. (1989);
Advanced c Chemistry” by J. March, Wiley lnterscience (1985); “Designing Organic Synthesis” by
S Warren, Wiley lnterscience (1978); “Organic Synthesis — The Disconnection Approach” by S Warren,
Wiley lnterscience ; book to c Synthesis” by RK Mackie and DM Smith, Longman
(1982); “Protective Groups in Organic Synthesis” by TW Greene and PGM Wuts, John Wiley and Sons,
Inc. (1999); and “Protecting Groups” by PJ, Kocienski, Georg Thieme Verlag ; and any updated
versions of said standard works.
In the general synthetic methods below, unless otherwise specified, the substituents are as defined above
1011 101
with reference to the compounds of formula (I) above. R is the same as R or a suitably protected
version thereof.
W0 2012/137089 19
Scheme 1 illustrates the preparation of the intermediates of general formula (Int 1), where they can be
made from amine (lnt3) where, in those cases where R1 contains an l, a protected form of R1
where a suitable hydroxyl protecting group (PG) is used. Any suitable oxygen protecting group may be
used (as described in “Protecting Groups in c Synthesis” 3rd edition T.W. Greene and PG. Wuts,
Wiley-lnterscience, 1999). Common oxygen protecting groups suitable for use herein e tert-
butyldimethylsilyl (TBDMS), tetrahydropyranyl (THP) and tertbutylsilyl (TBS).
Compounds of formula (Int 1) can be prepared from compounds of formula (Int 2) as illustrated in Scheme
Scheme 1
I N \
+ R1—NH —> \ N \
)L _> \
/ H 2
R N CI /
R301 N/ N R301)LN N
‘R1 ‘R1
|nt4 |nt3 |nt2 |nt1
n R301 can be H or halogen, lly chlorine.
Compounds of formula (Int 1) may be prepared from amine (Int 3) and (lnt4) in a cyclisation step followed
by a dechlorination step. Typical conditions employed involve stirring the amine of general formula (Int 3)
and the aldehyde (Int 4) er, preferably in ethanol at a temperature from room temperature up to
80°C.
e.g. as exemplified in Preps. 1-5
The intermediate chloride (Int 2) is reduced using rd literature conditions, for example
hydrogenation using a suitable catalyst such as palladium on carbon and an additive such as ammonia in
a suitable solvent such as l. Alternatively the chloride may be d by displacing the chloro
with methane thiol followed by Raney Nickel l of the SMe intermediate.
e.g. as exemplified in Preps. 8—13
In those cases where R1 contains one or more alcohols, a protected form of R1 with a suitable hydroxyl
protecting group (PG) can be used. Any suitable oxygen protecting group protection/deprotection system
may be used (as described in cting Groups in Organic Synthesis” 3rd edition T.W. Greene and PG.
Wuts, Wiley-lnterscience, 1999). Common oxygen protecting groups suitable for use herein include tert-
butyldimethylsilyl (TBDMS) and tetrahydropyranyl (THP).
Intermediates of general formula (Int 3) and (Int 4) are either commercially available or will be nown
to those skilled in the art with reference to literature precedents and/or the preparations herein.
Compounds of formula (Int 1) can be prepared from compounds of formula (Int 7) as illustrated in
Scheme 2.
Scheme 2
W0 37089 20 2012/051363
Br Br
N \ 1
301i / + R—NH _.
301A“ll/j —> NI?0\Me /
301A
R N CI R N lTJH R N lTJH
R1 R1
|nt7 |nt3 |nt6 |nt5
A /NI \
R301 N N\
|nt1
Compounds of formula (Int 1) wherein R3 can be H or halogen, typically chlorine, may also be prepared
from compounds of formula (Int 7) through displacement of a halogen, typically chlorine, with amines of
formula (Int 3), in a palladium catalysed Suzuki on followed by an acidic cyclisation.
Typical conditions se stirring the amine of general formula (Int 3) and the intermediate of l
formula (Int 7) together with a suitable base, such as triethylamine, in a t such as acetonitrile or
dichloromethane, to provide compounds of general formula (Int 6).
The vinyl ether can be introduced by reacting intermediate (Int 6) with a suitable c ester and a
suitable base, such as sodium hydroxide and a suitable catalyst such as
tetrakis(triphenylphosphine)palladium (0) in a solvent such as THF at a temperature from room
temperature up to 70°C.
Intermediates of formula (Int 1) can be made by treatment of intermediate (Int 5) with an acid such as
hydrogen de in an organic solvent such as isopropanol at a temperature from room temperature up
to 70°C.
e.g. as exemplified in Preps. 60-62
Intermediates of general formula (Int 3) and (Int 7) are either commercially available or will be well-known
to those skilled in the art with reference to literature precedents and/or the preparations .
Compounds of formula (Int 8) can be prepared from compounds of formula (Int 1) as illustrated in
Scheme 3.
Scheme 3
N \ \ \
)L / 301i /
301 N R N N\
(Int 1) (Int 8)
Wherein R301 is H or halogen, typically chlorine;
W0 37089 21
nds of formula (Int 8) are lly prepared by iodination of the pyrrolopyrimidine intermediates
(Int 1).Typical conditions ed involve stirring the intermediate of general formula (XI) with an
ting reagent such as N-iodosuccinimide in a suitable solvent, such as DMF or acetonitrile.
e.g. as exemplified in Preps. 14-19, 40, 63.
Compounds of formula (Int 8) can also be prepared from compounds of formula (Int 9) as illustrated in
Scheme 4.
Scheme4
R1—LG '
\ (lnt10) N \
\ 301i /
Ram)NL N/ N R N N,
H R1
(lnt9) (lnt8)
Wherein R301 is H or halogen, typically chlorine; LG is halogen or tosylate, triflate or mesylate;
Alternatively ediates of general formula (Int 8) can be prepared by alkylation of the
pyrrolopyrimidine intermediates (Int 9), with compounds of formula (Int 10) using a suitable base such as
m carbonate or potassium carbonate in an organic solvent. A suitable alternative is to use an
additive (such as potassium iodide) as well as a base. Preferred conditions comprise cesium carbonate in
DMF at room temperature.
In those cases where R1 contains one or more alcohols, a protected form of R1 can be used as described
in Scheme 1.
E.g. as exemplified in Preparation 20.
Compounds of formula (Int 12) can be prepared from compounds of formula (Int 11) as illustrated in
Scheme 5.
Scheme 5
W0 2012/137089 22
I I I
r \ ~ R3011\
/ /
R301 N/H N R301 N N
MeHon/I9 Me9\\
(lnt9) (lnt11) R2°°
O (lnt12)
) (lnt14)
Wherein R301 is H or n, typically chlorine;
Intermediates of general formula (Int 9) are reacted in an alkylation reaction to provide an ester
intermediate (Int 11) or (Int 13), from which the ester group can be reduced and protected to furnish a
nd of general formula (Int 14), where R200 is a H or methyl group. As previously mentioned in
Scheme 1 the hydroxy group can be protected with a suitable oxygen protecting group (PG), where the
preferred protecting groups are TBDMS, TBS and THP.
Typical conditions employed for the alkylation involve stirring the compound of l formula (Int 9) with
the appropriate halide together with a suitable base, as described in Scheme 4. Compounds of general
a (Int 11) where R200 is H can be converted to intermediates (Int 13) where R is methyl by a
further alkylation, typically involving a suitable alkylating agent such as methyl iodide and a suitable base
such as potassium xide in an organic solvent such as THF.
e.g. as exemplified in Preps. 20, 21, 41, 53
Reduction of the ester intermediates (Int 11) and (Int 13) can be done by using a le reducing
reagent such as lithium dride, lithium nium hydride or diisobutylalumnium hydride in a
suitable solvent such as ethanol or THF. Alternatively intermediates of general formula (Int 12) can be
made in a two step reaction by hydrolysing the ester of formula (Int 11) or (Int 13) to the appropriate acid
using a suitable base such as aqueous lithium hydroxide in a suitable organic solvent such as THF then
activating the acid using a suitable reagent such as isobutyl chloroformate and using a suitable reducing
agent such as sodium borohydride.
e.g. as exemplified in Preps. 22, 42, 43, 54
Compounds of general formula (Int 14) can be made by protection of the hydroxy group of intermediates
(Int 12) with a suitable oxygen protecting group (PG), where the red protecting groups are TBDMS
and THP,as bed in Scheme 1.
e.g. as exemplified in Preps. 44, 45, 55.
W0 2012/137089 23
Compounds of formula (Int 16) can be prepared from compounds of formula (Int 8) as illustrated in
Scheme 6.
Scheme 6
O N|\ \ O R1o11
N/ \/N/ MeO\N / N R301 N\
“I/Ie I R
1011
R (mm) 301* /Nl \
R N N
X1 \R1
(Int 15)
(Int 16)
) l
Nil/W/N I HO R1011
O Dd \ /
HJKQ R301 N/ NR1
R1011 301A /
(Int 8) R N N
X1 R1
(Int 17) (Int 18)
Wherein R301 is H or n, typically ne; and X1 is a suitable halogen, typically bromine or iodine;
Compounds of formula (Int 16) can be prepared from compounds of formula (Int 8) and (Int 15) through a
metallation of intermediate (Int 8) (using a suitable organometallic reagent such as butyllithium or
isopropylmagnesium chloride) and reacting with the Weinreb amide intermediate (Int 15) at a temperature
from -78°C up to room temperature.
e.g. as exemplified in Preps. 26, 27, 46, 47, 56, 58, 64
Alternatively compounds of formula (Int 15) may be converted into aldehydes of formula (Int 17) by
reduction of the Weinreb amide ediate using a suitable reducing agent. Preferred conditions
comprise diisopropylaluminium hydride in THF at -78°C, exemplified in Preparation 106.
Compounds of formula (Int 17) may then be reacted with compounds of formula (Int 8) according to the
same ation procedure described above. The intermediate alcohol (Int 18) may then be oxidised to
the ketone (Int 16). l oxidation conditions involve using an oxidising reagent such as the Dess-
Martin reagent in DCM or 2-iodoxybenzoic acid in a suitable solvent such as ethyl acetate at a
ature from room ature to reflux temperature.
e.g. as exemplified in Preps. 29, 30
Intermediates of general a (Int 15) and (Int 17) are either commercially ble or will be well-
known to those skilled in the art with reference to literature precedents and/or the preparations herein.
W0 2012/137089 24
Corresponding intermediates and compounds of formula (I) where R is OH are considered as
tautormers of pyridones and can be made using an analogous methodology using a benzyl protecting
1011
group for the Weinreb amide step, viz R is benzyloxy (OBn), e.g. as illustrated below:
N \ /N
0 \
31/ o OBn
MeO\ R N ”‘1 \ /
N / N R
| I \\
Me \\ \\
OBn 3//l\\W ’/
R N N
H N
O O
o /
/ o
Pd-C; hydrogen Bromine, AcOH /
N \\ Br
\ A /
/ N
3 R N
R N N \1
\R1 R
O / O
/ R102-X-COZH
NH4OH
W \ NH2 —> ((I),R101=OH)
u R3AN/N|\ N
R
Compounds of formula (Int 20) can be prepared from compounds of formula (Int 8) as illustrated in
Scheme7
l O
ND MeO
\ + W
/ /|N
301 N Me
R N \
PhYN
('"t 8)
(Int 19) (Int 20)
Wherein R301 is H or halogen, lly chlorine;
Compounds of formula (Int 20) may be ed from compounds of formulae (Int 8) and (Int 19)
ing to a metallation procedure as described in Scheme 6 above.
Typical ions employed involve metallation of the intermediate halide (Int 8) (using a suitable
organometallic reagent such as butyllithium or isopropylmagnesium chloride) and reacting with the
Weinreb amide intermediate (Int 19) at a temperature from -78°C up to room temperature in a suitable
solvent such as THF.
e.g. as exemplified in Preps. 24, 25, 28, 50
Intermediates (Int 19) will be nown to those skilled in the art with reference to literature precedents
and/or the preparations herein.
e.g. as exemplified in Prep. 23
nds of formula (Int 21) can be prepared from compounds of formula (Int 16) as rated in
Scheme 8.
Scheme 8
R Unt16) unt21)
1011
untzz)
Wherein X1 is bromine or iodine;
Compounds of formula (Int 21) may be prepared from compounds of a (Int 16) through direct
amination of the halide using standard literature conditions. For example, amine (Int 21) is typically
prepared using ammonia with a suitable copper catalyst such as copper (ll) sulphate or copper (l) oxide in
suitable solvent such as NMP in a sealed vessel at a temperature n room temperature and 140°C.
Where R301 is Cl this is also displaced by ammonia under the same conditions to provide amines of
general formula (Int 21) where R3 is NH2.
e.g. as exemplified in Preps. 31, 32, 36, 48, 49, 57, 59, 65
De-protection of a hydroxyl ting group on R1 (if present) can also occur under these conditions. In
these cases, either the protecting group can be reapplied as previously described in Scheme 5 or the
amine of l formula (Int 21) can be used directly.
Alternatively compounds of general formula (Int 21) where R3 is H, can be made by converting
intermediates of general formula (Int 16) where R is H, via compounds (Int 22). Typical conditions
W0 2012/137089 26
ed involve stirring the halide of general formula (Int 16), where R301 is H, with benzophenone
imine, a suitable base such as potassium phosphate, a le ligand such as 2-di-tert—butylphosphino—
6'—triisopropylbiphenyl and a suitable catalyst such as tris(dibenzylideneacetone)dipalladium in an
organic solvent such as 1,2-dimethoxyethane at a temperature from room temperature up to the boiling
point of the solvent.
Intermediate (Int 22) can be deprotected to furnish the amines of general formula (Int 21). Typical
conditions employ treatment with an aqueous acid such as hydrogen chloride or citric acid in an organic
solvent such as THF.
e.g. as exemplified in Preps. 33, 37-39
Compounds of formula (Int 20) can be prepared from compounds of formula (Int 22 where R301 is Cl) as
illustrated in Scheme 9.
Scheme 9
(Int 22)
Where R301 is Cl) ('"t 20)
nds of formula (Int 20) may be prepared from compounds of formula (Int 22) where R is chloro
h an ion reaction as ed above. Wherein the ne is is reacted with 2,4-
dimethoxybenzylamine and the amine can be deprotected as previously.
Typical conditions employed involve stirring the chloro-pyrimidine of general formula (Int 22), where R301
is Cl, with 2,4-dimethoxybenzylamine and a suitable additive such as 4-dimethylaminopyridine in a
suitable solvent such as 1,4 dioxane at a temperature from room temperature up to reflux temperature.
e.g. as exemplified in Prep. 51
Compounds of formula (I) can be prepared from compounds of formula (Int 21) and (Int 23) as illustrated
in Scheme 10.
Scheme 10
/N /N
O R1011 O R1011
\ / o \ / O
NI \ NH
\ sz—XJLOH —>
)NL \ \ ”4x
RBAN/ N\ R3 N/ [102
R1 R
(Int 21) (Int 23) (I)
Compounds of formula (I) may be prepared from compounds of formula (Int 21) and (Int 23) via amide
formation, if necessary adding a suitable base (such as DIPEA) and/or ve (such as DMAP), and a
suitable solvent (such as ne).
Typical conditions employed involve stirring the amine of general formula (Int 21) and the acid of general
formula (Int 23) together with a suitable coupling reagent such as HATU or 1-propylphosphonic acid cyclic
anhydride, if necessary adding a suitable base such as NMM, DIPEA or TEA in a suitable solvent such as
pyridine, THF, DMF or DMA at a temperature from room temperature up to 50°C. A suitable alternative is
to use an additive (such as 4-dimethylaminopyridine) as well as a base. Any suitable solvent may be used
in place of those mentioned above. At least one equivalent of the acid (Int 23) and at least one equivalent
of the coupling t should be used and an excess of one or both may be used if desired.
e.g. as exemplified in Examples 1-8, 34-45,48—53, 57-64, Preps. 34, 35, 52, 66-78
Where R1 contains a suitable yl protecting group in intermediate (Int 21), removal of the ting
group (PG) can be done in situ or as an additional step, adding a suitable acid and organic solvent to the
crude residue after the amide formation has taken place. Common protecting groups to use include
TBDMS, which is readily removed by treatment with an acid such as aqueous hydrogen chloride or
aqueous citric acid in an organic solvent such as THF or by treatment with a fluoride source such as
tetrabutylammonium fluoride in an organic solvent such as THF, and THP, which is also readily removed
by treatment with an acid such as aqueous hydrogen chloride in an c solvent such as THF.
e.g. as exemplified in Examples 9-33, 54-56,
Intermediates of general formula (Int 23) are either commercially available or will be nown to those
skilled in the art with reference to ture precedents and/or the preparations herein.
Compounds of formula (I) where R3 is NH2 can be ed from compounds of a (Int 20) as
illustrated in Scheme 11.
Scheme11
/N /N
O R1011 O R1011
\ / \ / 0
NH \ N4
1\ \ 2 J1 e i \ .
/ / i
N N N\ + x OH R3 N N\ R
H R1 IIRWZ R1
MeO OMe
(Int 24) (Int 23) (I)
Compounds of formula (I) can be prepared from compounds of formula (Int 24) via amide bond formation
as previously described in Scheme 10 ed by removal of the dimethoxbenzylamine group in situ, by
adding a suitable acid and organic solvent to the crude residue after the amide formation has taken place.
Suitable acids for this de-protection include hydrogen choride or oroacetic acid in an organic solvent
such as THF.
e.g. as exemplified in Examples 46-47
W0 2012/137089 28
Compounds of formula (I) where R2 is methyl can be prepared from compounds of formula (I) where R2 is
H as illustrated in Scheme 12.
Scheme 12
R (Int 21 )
Compounds of a (I) where R2 is methyl can be prepared from compounds of a (I) where R2 is
H according to an alkylation reaction with methyl iodide as described in Scheme 4.
When XR102 is boc, this can be deprotected using standard protecting group conditions to provide
intermediate (Int 21).
Compounds of formula (I) where X is NR104 can be prepared from compounds of formula (Int 21) as
illustrated in Scheme 13.
Scheme13
/N /N
O R1011 O R1011
\ / \
R104\ R102
ND + —> i\
\ H/ \ m X
R3)\N/ N\ R3 N/ N\1 R102/ \R104
R1 R
(Int25)
(Int 21) (I)
Compounds of a (I) where X is NR104 may be prepared from compounds of formula (Int 21), (Int 25)
and phenylchloroformate. Typical conditions comprise phenyl chloroformate and compounds of a
(Int 24) with pyridine in THF from 0 to 100°C, as exemplified in Example 526.
W0 2012/137089 29
Compounds of formula (I) where a substituent on the ) of R102 is an aminomethyl CHZNRZ group may
be prepared from nds of formula (1) as illustrated in Scheme 14.
Scheme14
/N R1011
O R1011
\ / o
R3)\N/ N|\ \ NW
N OH R3)\
R1011
—, N\ ”WW5N
Compounds of formula (I) where a primary alcohol exists may be oxidised to the aldehyde using Dess
Martin periodinane in DCM at room temperature followed by a reductive amination with a le amine
HNR2 using sodium triacetoxyborohydride and acetic acid in DCM.
Compounds of a (Int 26) can be prepared from compounds of formula (Int 27) as illustrated in
Scheme 15.
Scheme 15
Int 27 Int 26
Wherein R103 is Me or CHZOH;
Compounds of formula (Int 26) can be prepared from compounds of formula (Int 27) through conversion
of an alcohol into a suitable leaving group followed by cyclisation under basic conditions. Preferred
conditions comprise tosyl chloride with n-butyl lithium in THF.
According to a further embodiment the present invention provides novel intermediate compounds.
W0 2012/137089 30
ceutically acceptable salts of a compound of formula (I) may be readily prepared by mixing
er solutions of the compound of a (I) and the desired acid or base, as appropriate. The salt
may precipitate from solution and be collected by tion or may be recovered by ation of the
solvent. The degree of tion in the salt may vary from completely ionised to almost non-ionised.
The compounds of the invention intended for pharmaceutical use may be administered alone or in
combination with one or more other compounds of the invention or in combination with one or more other
drug agent (or as any combination thereof). Generally, they will be administered as a formulation in
association with one or more pharmaceutically acceptable excipients. The term “excipient” is used herein
to describe any biologically inactive ingredient other than the compounds and salts of the ion. The
choice of excipient will to a large extent depend on factors such as the particular mode of administration,
the effect of the excipient on solubility and stability, and the nature of the dosage form. For example, a
compound of the formula I, or a pharmaceutically able salt or solvate thereof, as defined above,
may be stered simultaneously (e.g. as a fixed dose combination), sequentially or separately in
combination with one or more other drug agent.
Exemplary additional agents could be selected from one or more of:
o a Nav1.7 channel modulator, such as a compound disclosed in WO 2009/012242 or
W02010/079443;
c an alternative sodium channel modulator, such as a Nav1.3 modulator (e.g. as disclosed in
W02008/118758); or a Nav1.8 modulator (e.g. as disclosed in , more particularly N-
[6-Amino(2-chloromethoxyphenyl)pyridinyl]—1-methyl-1H-pyrazolecarboxamide);
c an inhibitor of nerve growth factor signaling, such as: an agent that binds to NGF and inhibits NGF
biological activity and/or downstream pathway(s) mediated by NGF signaling (e.g. tanezumab), a
TrkA antagonist or a p75 antagoinsist;
o a compound which ses the levels of endocannabinoid, such as a compound with fatty acid amid
hydrolase inhibitory (FAAH) activity, in particular those disclosed in (e.g. N-
pyridazinyl(3-{[5-(trifluoromethyl)pyridineyl]oxy}benzylidene)piperidenecarboxamide);
c an opioid analgesic, e.g. morphine, heroin, hydromorphone, oxymorphone, hanol, levallorphan,
methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, one, hydrocodone,
propoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine
or ocine;
o a nonsteroidal antiinflammatory drug (NSAID), e.g. aspirin, diclofenac, inal, etodolac, en,
ofen, flufenisal, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofenamic acid,
mefenamic acid, meloxicam, tone, naproxen, nimesulide, nitroflurbiprofen, olsalazine,
oxaprozin, phenylbutazone, piroxicam, sulfasalazine, sulindac, tolmetin or zomepirac;
o a barbiturate sedative, e.g. amobarbital, aprobarbital, butabarbital, butabital, mephobarbital,
metharbital, methohexital, pentobarbital, phenobartital, secobarbital, al, theamylal or thiopental;
W0 2012/137089 31
a benzodiazepine having a sedative action, e.g. chlordiazepoxide, clorazepate, diazepam,
flurazepam, lorazepam, oxazepam, temazepam or lam;
an H1 antagonist having a sedative , e.g. diphenhydramine, pyrilamine, promethazine,
chlorpheniramine or chlorcyclizine;
a sedative such as glutethimide, meprobamate, methaqualone or dichloralphenazone;
a skeletal muscle relaxant, e.g. baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine,
methocarbamol or orphrenadine;
an NMDA receptor antagonist, e.g. dextromethorphan ((+)—3-hydroxy-N-methylmorphinan) or its
metabolite dextrorphan ((+)hydroxy—N-methylmorphinan), ketamine, memantine, pyrroloquinoline
quinine, cis(phosphonomethyl)piperidinecarboxylic acid, budipine, EN-3231 (MorphiDex®, a
combination formulation of morphine and dextromethorphan), topiramate, neramexane or perzinfotel
including an NRZB antagonist, e.g. odil, traxoprodil or (—)-(R)—6-{2-[4-(3-fluorophenyl)
hydroxypiperidinyl]hydroxyethyl-3,4-dihydro-2(1H)-quinolinone;
an alpha-adrenergic, e.g. doxazosin, tamsulosin, ine, guanfacine, dexmetatomidine, modafinil,
or 4-amino-6,7-dimethoxy—2-(5-methane-sulfonamido-1,2,3,4-tetrahydroisoquinolyl)—5-(2-pyridyl)
quinazoline;
a lic antidepressant, e.g. desipramine, imipramine, ptyline or nortriptyline;
an anticonvulsant, e.g. azepine, lamotrigine, topiratmate or valproate;
a inin (NK) antagonist, particularly an NK-3, NK-2 or NK-1 antagonist, e.g. (dR,9R)—7-[3,5-
bis(trifluoromethyl)benzyl]-8,9,10,11-tetrahydromethyl(4-methylphenyl)—7H-[1,4]diazocino[2,1-
g][1,7]-naphthyridine13-dione (TAK-637), 5-[[(2R,3S)—2-[(1R)—1-[3,5-
bis(trifluoromethyl)phenyl]ethoxy—3-(4-fluorophenyl)morpholinyl]-methyl]—1,2-dihydro-3H-1,2,4-
triazolone (MK-869), aprepitant, lanepitant, dapitant or methoxy—5-(trifluoromethoxy)phenyl]-
methylamino]phenylpiperidine (28,38);
a inic antagonist, e.g oxybutynin, tolterodine, propiverine, tropsium chloride, darifenacin,
solifenacin, temiverine and ipratropium;
a COX-2 selective inhibitor, e.g. celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib, or
lumiracoxib;
a coal-tar analgesic, in particular paracetamol;
a eptic such as droperidol, chlorpromazine, haloperidol, perphenazine, thioridazine,
dazine, trifluoperazine, fluphenazine, clozapine, olanzapine, idone, ziprasidone,
quetiapine, sertindole, razole, sonepiprazole, blonanserin, iloperidone, perospirone, ride,
zotepine, bifeprunox, asenapine, lurasidone, amisulpride, ridone, palindore, eplivanserin,
osanetant, rimonabant, meclinertant, Miraxion® or sarizotan;
a vanilloid receptor agonist (e.g. resinferatoxin) or antagonist (e.g. capsazepine);
a beta-adrenergic such as propranolol;
a local anaesthetic such as mexiletine;
a corticosteroid such as dexamethasone;
W0 2012/137089 32
a 5-HT receptor agonist or antagonist, particularly a 5-HT1B/1D agonist such as eletriptan, iptan,
naratriptan, zolmitriptan or rizatriptan;
a 5-HT2A receptor antagonist such as R(+)—alpha-(2,3-dimethoxy-phenyl)—1-[2-(4-fluorophenylethyl)]
piperidinemethanol (MDL-100907);
a 5-HT3 antagonist, such as ondansetron
a cho|inergic (nicotinic) analgesic, such as ispronicline (TC-1734), (E)—N-methyl(3-pyridinyl)
butenamine (RJR-2403), (R)—5-(2-azetidinylmethoxy)—2-chloropyridine (ABT-594) or nicotine;
Tramadol®;
a PDEV inhibitor, such as 5-[2-ethoxy(4-methylpiperazinyl-sulphonyl)phenyl]methyln-
propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidinone (sildenafil), aR)—2,3,6,7,12,12ahexahydromethyl
(3,4-methylenedioxyphenyl)—pyrazino[2',1':6,1]-pyrido[3,4-b]indole-1,4-dione
(lC-351 or tadalafil), thoxy(4-ethyl-piperazinylsu|phonyl)-phenyl]methylpropyl-3H-
o[5,1-f][1,2,4]triazinone (vardenafil), 5-(5-acetylbutoxypyridinyl)ethyl(1-ethyl
azetidinyl)-2,6-dihydro-7H—pyrazolo[4,3-d]pyrimidinone, 5-(5-acetylpropoxypyridinyl)ethyl
(1-isopropylazetidinyl)-2,6-dihydro-7H—pyrazolo[4,3-d]pyrimidinone, 5-[2-ethoxy(4-
ethylpiperaziny|sulphonyl)pyridinyl]ethyl[2-methoxyethyl]—2,6-dihydro-7H-pyrazolo[4,3-
d]pyrimidinone, 4-[(3-chloromethoxybenzyl)amino][(28)—2-(hydroxymethyl)pyrrolidinyl]-N-
(pyrimidinylmethyl)pyrimidinecarboxamide, 3-(1-methyloxopropyl-6,7-dihydro-1H-
pyrazolo[4,3-d]pyrimidinyl)—N-[2-(1-methylpyrrolidinyl)ethyl]propoxybenzenesulfonamide;
an 2-delta ligand such as gabapentin, pregabalin, ylgabapentin, (1d,3d,5d)(3-aminomethyl-bicyclo
[3.2.0]heptyl)—acetic acid, (3S,5R)—3-aminomethylmethyl-heptanoic acid, (3S,5R)—
3-aminomethyl-heptanoic acid, (3S,5R)—3-aminomethyl-octanoic acid, (28,4S)(3-
chlorophenoxy)proline, (28,4S)—4-(3-fluorobenzyl)-proline, [(1R,5R,68)—6-
(aminomethyl)bicyclo[3.2.0]heptyl]acetic acid, 3-(1-aminomethyl-cyclohexylmethyl)—4H-
[1 ,2,4]oxadiazolone, C-[1-(1H-tetrazolylmethyl)-cycloheptyl]-methylamine, (3S,4S)—(1-
aminomethyl-3,4-dimethyl-cyclopentyl)-acetic acid, (3S,5R)—3-aminomethylmethyl-octanoic acid,
(3S,5R)—3-aminomethyl-nonanoic acid, (3S,5R)—3-aminomethyl-octanoic acid, (3R,4R,5R)—3-
amino-4,5-dimethyl-heptanoic acid and (3R,4R,5R)—3-amino-4,5-dimethyl-octanoic acid;
metabotropic glutamate subtype 1 receptor (mGluR1) antagonist;
a serotonin reuptake inhibitor such as sertraline, sertraline metabolite demethylsertraline, fluoxetine,
oxetine (fluoxetine desmethyl metabolite), fluvoxamine, paroxetine, citalopram, citalopram
metabolite desmethylcitalopram, escitalopram, d,l-fenfluramine, femoxetine, ine, cyanodothiepin,
litoxetine, dapoxetine, nefazodone, amine and trazodone;
a noradrenaline (norepinephrine) reuptake inhibitor, such as maprotiline, amine, mirtazepine,
oxaprotiline, fezolamine, tine, mianserin, buproprion, buproprion lite
hydroxybuproprion, nomifensine and zine (Vivalan®), especially a selective enaline
reuptake inhibitor such as reboxetine, in particular (S,S)—reboxetine;
W0 2012/137089 33
o a dual serotonin-noradrenaline reuptake tor, such as venlafaxine, venlafaxine metabolite O-
desmethylvenlafaxine, clomipramine, clomipramine metabolite desmethylclomipramine, duloxetine,
milnacipran and imipramine;
c an inducible nitric oxide synthase (iNOS) inhibitor such as S—[2-[(1-iminoethyl)amino]ethyl]-L-
homocysteine, S—[2-[(1-iminoethyl)-amino]ethyl]-4,4-dioxo-L-cysteine, (1-iminoethyl)amino]ethyl]—
2-methyl-L-cysteine, (28,5Z)—2-aminomethyl[(1-iminoethyl)amino]heptenoic acid, 2-[[(1R,3S)—
3-amino hydroxy(5-thiazolyl)-butyl]thio]chloropyridinecarbonitrile; 2-[[(1R,3S)—3-amino
y(5-thiazolyl)butyl]thio]chlorobenzonitrile, (28,4R)—2-amino[[2-chloro
(trifluoromethyl)phenyl]thio]thiazolebutanol,
2-[[(1R,3S)—3-aminohydroxy(5-thiazolyl) thio](trifluoromethyl)-3 pyridinecarbonitrile, 2-
[[(1R,3S)—3- aminohydroxy— 1 iazolyl)butyl]thio]chlorobenzonitrile, N-[4-[2-(3-
chlorobenzylamino)ethyl]phenyl]thiophenecarboxamidine, or guanidinoethyldisulfide;
c an acetylcholinesterase inhibitor such as donepezil;
o a prostaglandin E2 subtype 4 (EP4) antagonist such as N-[({2-[4-(2-ethyl-4,6-dimethyl-1H-imidazo[4,5-
c]pyridinyl)phenyl]ethyl}amino)-carbonyl]methylbenzenesulfonamide or )—1-({[5-chloro
(3-fluorophenoxy)pyridinyl]carbonyl}amino)ethyl]benzoic acid;
0 a microsomal prostaglandin E synthase type 1 (mPGES—1) inhibitor;
0 a leukotriene B4 antagonist; such as 1-(3-biphenylylmethylhydroxy—chromanyl)—
cyclopentanecarboxylic acid 5696), 5-[2-(2-Carboxyethyl)[6-(4-methoxyphenyl)-5E-
hexenyl]oxyphenoxy]—valeric acid (ONO-4057) or DPC-11870,
a 5-lipoxygenase inhibitor, such as zileuton, 6-[(3-fluoro[4-methoxy-3,4,5,6-tetrahydro-2H-pyran
yl])phenoxy—methyl]methylquinolone (ZD-2138), or 2,3,5-trimethyl(3-pyridylmethyl),1,4-
benzoquinone (CV-6504).
Pharmaceutical compositions suitable for the delivery of compounds and salts of the present ion
and methods for their preparation will be readily apparent to those skilled in the art. Such compositions
and methods for their preparation may be found, for example, in ‘Remington’s ceutical Sciences’,
19th Edition (Mack Publishing Company, 1995).
Compounds and salts of the invention intended for pharmaceutical use may be ed and
administered as crystalline or ous products. They may be obtained, for example, as solid plugs,
powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or
evaporative drying. Microwave or radio frequency drying may be used for this purpose.
Oral Administration
The compounds of the invention may be administered orally. Oral administration may involve swallowing,
so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be
employed by which the compound enters the blood stream directly from the mouth.
W0 2012/137089 34
Formulations suitable for oral administration include solid formulations, such as s, capsules
containing particulates, liquids, or s; lozenges (including -filled), chews; multi- and nano-
particulates; gels, solid solution, me, films (including muco-adhesive), , sprays and liquid
formulations.
Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be
employed as s in soft or hard capsules and typically comprise a carrier, for example, water, ethanol,
hylene glycol, propylene glycol, methylcellulose, or a le oil, and one or more emulsifying
agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a
solid, for example, from a sachet.
The compounds of the invention may also be used in fast-dissolving, isintegrating dosage forms
such as those bed in Expert Opinion in Therapeutic Patents, fl (6), 6 by Liang and Chen
(2001).
For tablet dosage forms, depending on dose, the drug may make up from 1 weight% to 80 weight% of the
dosage form, more typically from 5 weight% to 60 weight% of the dosage form. In addition to the drug,
tablets generally contain a disintegrant. Examples of disintegrants include sodium starch glycolate,
sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone,
polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl
cellulose, starch, pregelatinised starch and sodium alginate. Generally, the disintegrant will comprise from
1 weight% to 25 weight%, preferably from 5 weight% to 20 weight% of the dosage form.
Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include
microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums,
polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose.
Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous
and the like), mannitol, xylitol, dextrose, e, sorbitol, microcrystalline cellulose, starch and dibasic
calcium phosphate dihydrate.
s may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate
80, and glidants such as silicon dioxide and talc. When present, surface active agents may comprise from
0.2 weight % to 5 weight% of the tablet, and glidants may comprise from 0.2 weight% to 1 weight% of the
.
Tablets also generally contain lubricants such as ium stearate, calcium stearate, zinc stearate,
sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate. Lubricants
generally comprise from 0.25 weight% to 10 weight%, preferably from 0.5 weight% to 3 % of the
40 tablet.
Other possible ingredients include anti-oxidants, ants, flavoring agents, preservatives and taste-
masking agents.
Exemplary tablets contain up to about 80% drug, from about 10 weight% to about 90 weight% binder,
from about 0 weight% to about 85 % diluent, from about 2 weight% to about 10 weight%
disintegrant, and from about 0.25 weight% to about 10 weight% lubricant. [Make sure these specific
ranges are relevant]
Tablet blends may be compressed directly or by roller to form s. Tablet blends or portions of blends
may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tableting. The final
ation may comprise one or more layers and may be coated or uncoated; it may even be
encapsulated.
The formulation of tablets is discussed in aceutical Dosage Forms: Tablets, Vol. 1”, by H.
Lieberman and L. Lachman, Marcel Dekker, N.Y., N.Y., 1980 (ISBN 06918—X).
The ing formulations for the various types of administration discussed above may be formulated to
be immediate and/or modified release. Modified release formulations include delayed-, sustained-, -
and programmed release.
, controlled-, ed
Suitable modified release formulations for the purposes of the ion are described in US Patent No.
6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic
and coated particles are to be found in Verma et al, Pharmaceutical Technology e, 25(2), 1-14
(2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
eral Administration
The compounds and salts of the invention may be administered directly into the blood stream, into
muscle, or into an internal organ. Suitable means for parenteral stration include intravenous,
intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial,
intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including
microneedle) injectors, -free injectors and infusion techniques.
Parenteral formulations are typically aqueous solutions which may n excipients such as salts,
carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they
may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in
conjunction with a suitable vehicle such as sterile, pyrogen-free water.
W0 2012/137089 36
The preparation of parenteral formulations under sterile conditions, for example, by lyophilisation, may
readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.
The solubility of compounds of formula (I) and salts used in the preparation of parenteral solutions may be
increased by the use of appropriate formulation techniques, such as the incorporation of solubility-
enhancing agents.
Formulations for parenteral administration may be formulated to be immediate and/or modified release.
Thus, compounds and salts of the invention may be formulated as a solid, semi-solid, or thixotropic liquid
for administration as an implanted depot providing modified e of the active compound. An example
of such ations include drug-coated .
Topical Administration
The compounds and salts of the invention may also be administered topically to the skin or mucosa, that
is, dermally or transdermally. Typical formulations for this purpose include gels, hydrogels, lotions,
ons, creams, ointments, g powders, dressings, foams, films, skin patches, wafers, implants,
sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include
l, water, mineral oil, liquid petrolatum, white petrolatum, in, polyethylene glycol and propylene
glycol. Penetration enhancers may be incorporated [see, for example, Finnin and Morgan, J Pharm Sci,
Q (10), 955-958 (October 1999).] Other means of topical administration include delivery by
electroporation, iontophoresis, horesis, sonophoresis and microneedle or needle-free (e.g.
PowderjectTM, BiojectTM, etc.) injection.
d/lntranasal Administration
The compounds and salts of the invention may also be administered intranasally or by inhalation, typically
in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a
mixed component particle, for example, mixed with phospholipids, such as atidylcholine) from a
dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser rably
an er using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a
le lant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. For intranasal
use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
A pressurised container, pump, spray, atomizer, or nebuliser may contain a solution or suspension of the
compound(s) or salt(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable
alternative agent for sing, solubilising, or extending release of the active, a propellant(s) as solvent
and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
Prior to use in a dry powder or suspension formulation, the drug t is ised to a size suitable
for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate
comminuting , such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form
nanoparticles, high pressure nisation, or spray drying.
Capsules (made, for example, from gelatin or HPMC), blisters and cartridges for use in an inhaler or
insufflator may be formulated to contain a powder mix of the compound or salt of the invention, a le
powder base such as lactose or starch and a performance er such as l-leucine, mannitol, or
magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate, preferably the
. Other suitable excipients include dextran, glucose, maltose, ol, xylitol, fructose, sucrose and
trehalose.
A suitable solution formulation for use in an er using electrohydrodynamics to produce a fine mist
may contain from 1ug to 20mg of the compound or salt of the invention per actuation and the actuation
volume may vary from 1pl to 100ul. A typical formulation may comprise a compound of formula (I) or salt
thereof, propylene glycol, sterile water, ethanol and sodium chloride. Alternative solvents which may be
used instead of propylene glycol include glycerol and polyethylene glycol.
Suitable flavours, such as menthol and levomenthol, or ners, such as saccharin or saccharin
sodium, may be added to those ations of the invention intended for inhaled/intranasal
administration.
Formulations for inhaled/intranasal administration may be formulated to be immediate and/or modified
release using, for example, poly(DL-lactic-coglycolic acid (PGLA). Modified release formulations e
delayed-, sustained-, pulsed-, controlled-, ed and programmed release.
In the case of dry powder inhalers and aerosols, the dosage unit is determined by a prefilled capsule,
blister or pocket or by a system that utilises a gravimetrically fed dosing chamber . Units in accordance
with the invention are typically arranged to administer a metered dose or "puff" containing from 1 to 5000
pg of the compound or salt. The overall daily dose will typically be in the range 1 pg to 20 mg which may
be administered in a single dose or, more y, as divided doses throughout the day.
Rectal/lntravaginal Administration
The compounds and salts of the invention may be administered rectally or vaginally, for example, in the
form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various well
known alternatives may be used as appropriate.
Ocular and Aural Administration
W0 2012/137089 38
The compounds and salts of the invention may also be administered directly to the eye or ear, typically in
the form of drops of a micronised suspension or on in isotonic, pH-adjusted, sterile . Other
formulations suitable for ocular and aural administration include ointments, biodegradable (e.g.
absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and
particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed-linked
polyacrylic acid, polyvinylalcohol, hyaluronic acid; a cellulosic r, for example,
hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose; or a heteropolysaccharide
polymer, for
example, gelan gum, may be incorporated together with a preservative, such as benzalkonium de.
Such formulations may also be delivered by iontophoresis.
Other Technologies
The compounds and salts of the invention may be combined with soluble macromolecular entities, such
as cyclodextrin and suitable derivatives thereof or polyethylene glycol-containing polymers, in order to
improve their lity, dissolution rate, taste-masking, bioavailability and/or ity for use in any of the
aforementioned modes of administration.
Drug-cyclodextrin complexes, for example, are found to be lly useful for most dosage forms and
administration routes. Both inclusion and non-inclusion complexes may be used. As an alternative to
direct complexation with the drug, the extrin may be used as an auxiliary additive, i.e. as a r,
diluent, or solubiliser. Most commonly used for these purposes are , beta- and gamma-
cyclodextrins, es of which may be found in International Patent Applications Nos. WO 72,
WO 94/02518 and WO 98/55148.
For stration to human patients, the total daily dose of the nds and salts of the invention is
typically in the range 0.1 mg to 200 mg depending, of course, on the mode of administration, preferred in
the range 1 mg to 100 mg and more preferred in the range 1 mg to 50 mg. The total daily dose may be
administered in single or divided doses.
These dosages are based on an average human subject having a weight of about 65kg to 70kg. The
physician will readily be able to determine doses for subjects whose weight falls outside this range, such
as infants and the elderly.
For the above-mentioned therapeutic uses, the dosage administered will, of course, vary with the
compound or salt employed, the mode of administration, the treatment desired and the disorder indicated.
The total daily dosage of the compound of formula (|)/salt/solvate (active ingredient) will, generally, be in
the range from 1 mg to 1 gram, preferably 1 mg to 250 mg, more preferably 10 mg to 100 mg. The total
W0 2012/137089 39
daily dose may be administered in single or divided doses. The present invention also asses
sustained release compositions.
The pharmaceutical composition may, for example, be in a form suitable for parenteral injection as a sterile
solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal
administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for
single administration of e dosages. The pharmaceutical composition will include a conventional
pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In
addition, it may include other medicinal or ceutical , carriers, adjuvants, etc.
Exemplary parenteral administration forms include solutions or suspensions of active compounds in e
aqueous solutions, for e, aqueous propylene glycol or dextrose solutions. Such dosage forms can be
suitably buffered, if desired.
Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents. The
pharmaceutical compositions may, if desired, contain additional ingredients such as flavorings, s,
excipients and the like. Thus for oral administration, tablets containing various excipients, such as citric acid
may be employed together with various disintegrants such as starch, alginic acid and certain complex
silicates and with binding agents such as sucrose, gelatin and acacia. Additionally, lubricating agents such
as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes. Solid
compositions of a similar type may also be employed in soft and hard filled gelatin capsules. Preferred
materials, or, include lactose or milk sugar and high molecular weight polyethylene s. When
aqueous suspensions or elixirs are desired for oral administration the active compound n may be
combined with various ning or flavoring agents, coloring matters or dyes and, if desired, emulsifying
agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or
combinations thereof.
Dosage regimens may be adjusted to provide the optimum desired response. For e, a single bolus
may be administered, several divided doses may be administered over time or the dose may be
proportionally d or increased as indicated by the cies of the therapeutic situation. It is
especially advantageous to ate parenteral itions in dosage unit form for ease of administration
and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrete units suited as
unitary dosages for the mammalian subjects to be treated; each unit ning a predetermined quantity of
active nd calculated to e the desired therapeutic effect in association with the required
pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and
directly ent on (a) the unique characteristics of the chemotherapeutic agent and the particular
therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding
such an active compound for the treatment of sensitivity in individuals.
W0 2012/137089 40
Thus, the skilled artisan would appreciate, based upon the disclosure ed herein, that the dose and
dosing regimen is adjusted in ance with methods well-known in the therapeutic arts. That is, the
maximum tolerable dose can be y established, and the effective amount providing a detectable
therapeutic benefit to a patient may also be determined, as can the temporal requirements for
administering each agent to e a detectable therapeutic benefit to the patient. Accordingly, while
n dose and administration regimens are exemplified herein, these examples in no way limit the dose
and administration n that may be provided to a patient in practicing the present ion.
It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated, and
may include single or multiple doses. It is to be further understood that for any particular subject, specific
dosage regimens should be adjusted over time according to the individual need and the professional
judgment of the person stering or supervising the administration of the compositions, and that dosage
ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed
composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamic
parameters, which may e clinical effects such as toxic effects and/or laboratory values. Thus, the
present invention encompasses intra-patient dose-escalation as determined by the skilled artisan.
Determining appropriate dosages and regiments for administration of the chemotherapeutic agent are well-
known in the relevant art and would be understood to be encompassed by the skilled artisan once provided
the teachings disclosed herein.
A pharmaceutical composition of the ion may be prepared, packaged, or sold in bulk, as a single
unit dose, or as a plurality of single unit doses. As used herein, a "unit dose" is discrete amount of the
pharmaceutical composition comprising a ermined amount of the active ingredient. The amount of
the active ingredient is generally equal to the dosage of the active ingredient which would be stered
to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such
a dosage.
For parenteral dosages, this may conveniently be ed as a solution or as a dry powder requiring
ution by a pharmacist, medical practitioner or the patient. It may be provided in a bottle or sterile
syringe. For e it may be provided as a powder in a multicompartment syringe which allows the dry
powder and solvent to be mixed just prior to administration (to aid long-term stability and storage).
Syringes could be used which allow le doses to be administered from a single device.
The relative amounts of the active ient, the pharmaceutically acceptable carrier, and any additional
ients in a pharmaceutical composition of the invention will vary, depending upon the identity, size,
and condition of the subject treated and further depending upon the route by which the composition is to
be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w)
active ingredient.
W0 2012/137089 41
In on to the active ingredient, a pharmaceutical composition of the invention may further comprise
one or more additional pharmaceutically active agents.
Controlled- or sustained-release formulations of a pharmaceutical composition of the invention may be
made using tional technology.
As used herein, "parenteral administration" of a pharmaceutical composition includes any route of
administration characterized by physical breaching of a tissue of a subject and administration of the
pharmaceutical composition through the breach in the tissue. Parenteral administration thus includes, but
is not limited to, stration of a pharmaceutical composition by injection of the composition, by
application of the composition through a surgical incision, by ation of the composition through a
tissue-penetrating non-surgical wound, and the like. In ular, parenteral stration is
contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal
injection, and kidney dialytic infusion techniques.
Formulations of a pharmaceutical composition suitable for eral administration comprise the active
ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic
saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration
or for uous stration. lnjectable ations may be prepared, packaged, or sold in unit
dosage form, such as in s or in multi-dose containers containing a preservative. ations for
parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or
aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations as discussed
below. Such formulations may further comprise one or more additional ingredients including, but not
d to, suspending, stabilizing, or dispersing agents. In one embodiment of a formulation for
parenteral administration, the active ingredient is provided in dry (i.e. powder or granular) form for
reconstitution with a suitable vehicle (e.g. sterile pyrogen-free water) prior to parenteral administration of
the reconstituted composition.
A composition of the present invention can be administered by a y of methods known in the art. The
route and/or mode of administration vary depending upon the desired results. The active compounds can
be prepared with carriers that protect the compound against rapid release, such as a controlled release
ation, ing implants, transdermal patches, and microencapsulated delivery systems.
Biodegradable, patible polymers can be used, such as ethylene vinyl acetate, polyanhydrides,
polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such
formulations are bed by e.g., Sustained and Controlled Release Drug Delivery Systems, J. R.
Robinson, ed., Marcel Dekker, Inc, New York, (1978). Pharmaceutical compositions are preferably
manufactured under GMP conditions.
The pharmaceutical itions may be prepared, packaged, or sold in the form of a sterile injectable
aqueous or oily suspension or solution. This suspension or solution may be formulated according to the
40 known art, and may comprise, in addition to the active ingredient, additional ingredients such as the
W0 2012/137089 42
sing agents, wetting agents, or suspending agents described herein. Such sterile able
ations may be prepared using a non-toxic parenterally-acceptable diluent or t, such as water
or 1,3-butane diol, for example. Other acceptable diluents and ts include, but are not d to,
Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-
glycerides. Other parentally-administrable formulations which are useful e those which comprise
the active ingredient in microcrystalline form, in a liposomal ation, or as a component of a
biodegradable polymer system. Compositions for sustained release or tation may comprise
pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange
resin, a sparingly soluble polymer, or a sparingly e salt.
The e dosage administered of each active ingredient will vary depending upon any number of
factors, including but not limited to, the type of animal and type of disease state being treated, the age of
the , and the route(s) of administration.
The following non-limiting Preparations and Examples illustrate the preparation of compounds and salts of
the present invention.
GENERAL EXPERIMENTAL
The Preparations and Examples that follow illustrate the invention but do not limit the invention in any
way. All starting materials are available commercially or described in the literature. All temperatures are
in OC. Flash column chromatography was carried out using Merck silica gel 60 (9385). Thin layer
chromatography (TLC) was carried out on Merck silica gel 60 plates (5729). “R” ents the distance
travelled by a compound divided by the distance travelled by the t front on a TLC plate. Melting
points were determined using a Gallenkamp MPD350 apparatus and are uncorrected. NMR was carried
out using a Varian-Unity lnova 400MHz NMR spectrometer or a Varian Mercury 400MHz NMR
spectrometer. Mass spectroscopy was carried out using a Finnigan Navigator single quadrupole
electrospray mass spectrometer or a Finnigan aQa APCl mass spectrometer.
Where it is stated that compounds were prepared in the manner described for an earlier Preparation or
Example, the skilled person will appreciate that reaction times, number of equivalents of reagents and
reaction temperatures may be modified for each specific reaction, and that it may nevertheless be
necessary or desirable to employ different work-up or purification conditions.
The invention is illustrated by the ing non-limiting examples in which the following abbreviations and
definitions are used:
The Preparations and Examples that follow illustrate the invention but do not limit the invention in any
way. All starting materials are available commercially or described in the literature. All temperature are in
°C. Flash column chromatography was carried out using Merck silica gel 60 (9385) or Redisep silica.
NMR was carried out using a Varian y 400MHz NMR spectrometer or a Jeol ECX 400MHz NMR.
W0 2012/137089 43
The mass spectra were obtained using:
Waters ZQ ESCI
Applied tem's 00 5 min LC-MS
Waters Alliance 2795 with ZQ2000 (ESI)
Aglient 110 HPLC 5 min (System 5)
Waters ZQ ESCI 8min LC-MS
Waters ce 2695 with ZQ2000 (ESI) 25 min
HP 1100 HPLC with Waters Micromass ZQ mass detector 12.5 min LC-MS
UPLC mass spectra were obtained using a Waters Acquity ZQD (ESI) 1.5 min LC-MS
WATERS ACQUITY UPLC/WATERS 3100 MSD/PL-ELS 2100 ICE ELSD
Where singleton compounds have been analysed by LCMS, there are six methods used. These are
illustrated below.
System 1
6 minute LC-MS gradient and instrument conditions
A: 0.1 % formic acid in water
B: 0.1 % formic acid in acetonitrile
Column: C18 phase Waters Sunfire 50 x 4.6 mm with 5 micron particle size
nt: 95-5% A over 3 min, 1 min hold, 2 min re-equilibration, 1.5mL/min flow rate
UV: 210nm - 450nm DAD
Temperature: 50°C
System 2
2 minute LC-MS gradient and instrument conditions
A: 0.1 % formic acid in water
B: 0.1 % formic acid in acetonitrile
Column: C18 phase Phenomenex 20 x 4.0 mm with 3 micron particle size
nt: 70-2% A over 1.5min, 0.3 min hold, 0.2 re-equilbration, 1.8mL/min flow rate
UV: 210nm - 450nm DAD
Temperature: 75°C
System 3
minute LC-MS gradient and instrument conditions
A: 0.1 % formic acid in water
B: 0.1 % formic acid in acetonitrile
Column: C18 phase Waters Sunfire 50 x 4.6 mm with 5 micron particle size
Gradient: 95-5% oner 3 min, 1 min hold, 1 min re-equilibration, min flow rate
UV: 225nm — ELSD - MS
Temperature: ambient
System 4
minute LC-MS gradient and instrument conditions
40 A: 0.1 % ammonium hydroxide in water
WO 37089 44
B: 0.1 % ammonium hydroxide in acetonitrile
Column: C18 phase XTerra 50 x 4.6 mm with 5 micron particle size
Gradient: 95-5% oner 3 min, 1 min hold, 1 min re-equilibration, 1.5mL/min flow rate
UV: 225nm — ELSD - MS
Temperature: ambient
System 5
minute LC-MS gradient and instrument conditions
A: 0.0375 % TFA in water
B: 0.01875 % TFA in acetonitrile
Column: C18 phase Welch XB 50 x 2.1 mm with 5 micron particle size
Gradient: 99-0% A over 4 min, 0.70 min ilibration, 0.8 mL/min flow rate
UV: 225nm — ELSD - MS
Temperature: 50°C
System 6
5 minute LC-MS gradient and instrument conditions
A: 0.0375 % TFA in water
B: 0.01875 % TFA in acetonitrile
Column: C18 phase Welch XB 50 x 2.1 mm with 5 micron particle size
Gradient: 90-0% A over 4 min, 0.70 min re-equilibration, 0.8 mL/min flow rate
UV: 225nm — ELSD - MS
Temperature: 50°C
System 7
minute LC-MS nt and instrument conditions
A: 10 mmol ammonium bicarbonate in water
B: acetonitrile
Column: C18 phase XBridge 150 x 3.0 mm with 5 micron le size
Gradient: 95-5% A over 15 min, 10 min hold, 2 min re-equilibration, 0.5mL/min flow rate
UV: 200nm - 350nm DAD
Temperature: 30°C
System 8
3 minute LC-MS gradient and instrument conditions
A: 0.05% formic acid in water
B: acetonitrile
Column: C18 phase Restek 30 x 2.1 mm with 3 micron particle size
Gradient: 98—2% A over 2 min, 0.25 min hold, 0.75 min re-equilibration, min flow rate
UV: 200nm - 350nm DAD
Temperature: 50°C
System 9
minute LC-MS gradient and instrument conditions
40 A: 0.05% formic acid in water
WO 37089 45
B: acetonitrile
Column: C18 phase XBridge 50 x 4.6 mm with 5 micron particle size
Gradient: 90-10% A over 3 min, 1 min hold, 1min re-equilibration, min flow rate
UV: 200nm - 260nm DAD
Temperature: 25°C
System 10
minute LC-MS gradient and instrument conditions
A: 10 mM ammonium acetate in water
B: acetonitrile
Column: C18 phase Gemini NX 50 x 4.6 mm with 5 micron particle size
Gradient: 90-10% A over 3 min, 1 min hold, 1min ilibration, 1.2mL/min flow rate
UV: 200nm - 260nm DAD
Temperature: 25°C
Where singleton compounds have been purified by High Performance Liquid Chromatography, unless
otherwise stated, one of four s were used, and these are shown below.
Waters Purification Systems with mass spec or UV detection
Prep system 1
minute prep LC-MS gradient and instrument conditions
A: 0.1% formic acid in water
B: 0.1% formic acid in acetonitrile
Column: C18 phase Sunfire 100 x 19.0 mm
Gradient: 95-2% A over 7 min, 2 min hold, 1 min re-equilibration, 18 mL/min flow rate
Temperature: ambient
Prep system 2
minute prep LC-MS gradient and instrument conditions
A: 0.1% DEA in water
B: 0.1% DEA in acetonitrile
Column: C18 phase Xterra 100 X190 mm
Gradient: 95-2% A over 7 min, 2 min hold, 1 min re-equilibration, 18 mL/min flow rate
Temperature: ambient
Prep system 3
7 minute prep LC-MS gradient and instrument conditions
A: 0.05% ammonia in water
B: acetonitrile
: C18 phase e 50 x19.0 mm
Gradient: 90-20% A over 7 min, 20 mL/min flow rate
Temperature: ambient
Prep system 4
40 8 minute prep LC-MS gradient and instrument conditions
W0 2012/137089 46
A: 0.1% TFA in water
B: acetonitrile
: C18 phase Sepax BR 100 x 21.2 mm
nt: 96-33% A over 8 min, 30 mL/min flow rate
Temperature: ambient
Where it is stated that compounds were ed in the manner described for an earlier Preparation or
Example, the skilled person will appreciate that on times, number of equivalents of reagents and
reaction temperatures may have been modified for each specific reaction, and that it may nevertheless be
necessary, or ble, to employ different work-up or cation conditions. The invention is illustrated
by the following non-limiting Examples in which the following abbreviations and definitions are used:
AcOH — acetic acid; APCI - atmospheric pressure chemical ionization; Arbocel is a filter agent; br s —
broad singlet; BINAP — 2,2’-bis(diphenylphosphino)-1,1’-binapthyl; nBuLi — n-Butyllithium; CDCI3—
deuterated chloroform; Cs2C03 is caesium carbonate; Cul is copper (l) iodide; Cu(OAc)2 is copper (ll)
acetate; 6 — chemical shift; d — doublet; DAD — diode array detector; DCE — 1,2-dichloroethane
DCM — dichloromethane; DEA — lamine; DIBAL — utylaluminium hydride; DlPEA —
diisopropylethylamine; DMAP — 4-dimethylaminopyridine; DME — dimethoxyethane; DMF — N,N-
dimethylformamide; DMF-DMA - methylformamide-dimethylacetal; DMSO — dimethylsulphoxide
DPPF — 1,1’-bis(diphenylphosphino)ferrocene; ELSD — evaporative light scattering detector; ESI -
electrospray ionization; EtZO — diethylether; EtOAc/EA — ethyl acetate; EtOH — ethanol; g — gram; HATU -
zabenzotriazolyl)-1,1,3,3-tetramethyluronium hexafluorophosphate; HBTU is O-benzotriazolyl-
N,N,N’,N’-tetramethyluronium hexafluorophosphate; HCI is hydrochloric acid;HOBT is N-
hydroxybenzotriazole hydrate; HPLC — high pressure liquid chromatography; IPA — isopropyl alcohol;
K2C03 is potassium carbonate; KHSO4 is potassium hydrogen te; KOAc is potassium acetate; KOH
is potassium hydroxide; K3PO4 is potassium phosphate tribasic; KF - potassium fluoride; L is litre; LCMS
— liquid tography mass spectrometry; LiHMDS — Lithium hexamethyldisilazide; m — multiplet; mg —
milligram; mL — itre; M/Z — Mass Spectrum Peak; MeCN — acetonitrile; MeOH — methanol; 2-MeTHF —
2-methyltetrahydrofuran; MgSO4 is magnesium sulphate; MnOZ — manganese dioxide; NaClOZ — sodium
chlorite; NaH - sodium hydride; NaHC03- sodium hydrogencarbonate; Na2C03 - sodium carbonate;
NaH2PO4- sodium phosphate; NaH803 - sodium bisulphite; NaHSO4 - sodium hydrogensulphate; NaOH -
sodium ide; NaZSO4 - sodium sulphate; NH3 — ammonia; NH4C| — ammonium chloride; NMM — N-
MethylMorpholine; NMR — nuclear magnetic resonance; Pd/C — palladium on carbon; PdCl2 — palladium
dichloride; Pd2(dba)3 is tris(dibenzylideneacetone)dipalladium(0); Pd(PPh3)4 - palladium
tetrakis(triphenylphosphine); Pd(OAc)2 — palladium acetate; PTSA — para-toluenesulfonic acid; Prep —
preparation; Rt — retention time; q — quartet; s — singlet; TBDMS — tertbutyldimethylsilyl; TBME —
tertbutyldimethylether; TCP — 1-propylphosphonic acid cyclic anhydride; TEA — triethylamine; TFA —
oroacetic acid; THF — tetrahydrofuran; TLC — thin layer tography; (R, S) — racemic mixture;
WSCDI - 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride.
For the avoidance of doubt, named compounds used herein have been named using IUAPC, Chemdraw
and/or Name Pro ACD Labs Name Software v7.11TM or using other standard nomenclature. NMR spectra
were measured in deuterated solvents and were consistent with the names/structures given below.
Example 1: N-{5-[(7-tert—Butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(5-fluoropyridin
yl)acetam ide
\/ O
/ F
N/|\ ” ‘N/
KN N
Me Me
2-(5-Fluoropyridinyl)acetic acid (23.1 mg, 0.149 mmol) (see Preparation 92) was added to (5-
aminopyridinyl)(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (40 mg, 0.135 mmol ) (see
Preparation 31), 1-propylphosphonic acid cyclic anhydride (0.2 mL, 0.338 mmol, 50% in EtOAc) and
triethylamine (0.65 mL, 0.474 mmol) in THF (3 mL). The mixture was stirred at 25°C for 18 hours,
ated in vacuo and ioned between saturated aqueous sodium bicarbonate (5 mL) and ethyl
acetate (5 mL). The organic phase was dried over sodium sulfate, evaporated in vacuo and the residue
was triturated with pentane:diethyl ether (3:1, 1 mL) to afford the title compound as an off white solid in
65% yield, 38 mg.
1H NMR (400 MHz, DMSO) 6: 1.79 (s, 9H), 3.95 (s, 2H), 7.50 (m, 1H), 7.72 (m, 1H), 8.21 (s, 1H), 8.50 (d,
1H), 8.76 (d, 1H), 8.95 (d, 1H), 9.00 (s, 1H), 9.48 (s, 1H), 10.72 (s, 1H); LCMS (System 4): R = 2.86 min;
m/z 433 [M+H]+.
Examples 2 to 8 were prepared according to the method bed above for Example 1, starting from (5-
aminopyridinyl)(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (see Preparation 31) and the
appropriate acids.
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 4): R = 3.26
yl)carbonyl]pyridinyl}[4-(trifluoromethyl)-1H-1,2,3-triazol- min; m/z 473 [M+H]+
cetamide
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 4): R = 3.04
yl)carbonyl]pyridinyl}(3-cyclopropyl-1H-pyrazol min; m/z 444 [M+H]+
yl)acetamide
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 4): R = 2.75
yl)carbonyl]pyridinyl}(4-cyclopropyl-1H-1,2,3-triazol min; m/z 445 [M+H]+
yl)acetamide
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 4): R = 3.04
bonyl]pyridinyl}[3-(trifluoromethyl)-1H-pyrazol min; m/z 472 [M+H]+
yl]acetamide
W0 37089 48
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS m 4): R = 2.92
y|)carbonyl]pyridinyl}(5-ch|oropyridinyl)acetamide min; m/z 449 [M+H]+
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 4): R = 2.96
y|)carbonyl]pyridinyl}(4-cyc|opropyl-1H-pyrazol min; m/z 443 [M+H]+
y| )acetamide
N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS(system 4)
y|)carbonyl]pyridinyl}[4-(trif|uoromethy|)-1H-pyrazol : R = 3.00 min; m/z 472
yl]acetamide [M+H]+
Example 9: N-[5-({7-[(1S)—2-Hydroxymethylethy|]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridin
y|][4-(trif|uoromethyl)phenyl]acetamide
KN' N
HofMe
4-(Trifluoromethyl)phenylacetic acid (33.6 g, 165 mmol) was added to (5-aminopyridinyl){7-[(1S)—2-
—butyl(dimethy|)si|y|]oxy}methy|ethy|]-7H-pyrro|o[2,3-d]pyrimidinyl}methanone (45.2 g, 110
mmol) (see Preparation 37), ylphosphonic acid cyclic anhydride (194 mL, 329 mmol, 50% solution
in EtOAc) and triethylamine (53.6 mL, 384 mmol ) in THF (400 mL). The mixture was d at 25°C for 2
hours then saturated aqueous sodium bicarbonate (250 mL) was added and the organic layer was
separated. The aqueous phase was extracted with EtOAc (2 x 200 mL) and all organic phases were
ed and dried over sodium sulfate then evaporated in vacuo.
The residue brown solid was dissolved in THF (400 mL) and aqueous HCI (200 mL, 2M) was added. The
mixture was stirred at room temperature for 2 hours then cooled to 0°C and sodium hydroxide (28 g) was
added. The mixture was stirred for 3 hours then water (100 mL) was added. The organic layer was
separated and the aqueous phase was extracted with EtOAc (2 x 300 mL) and all organic phases were
combined and dried over sodium sulfate then evaporated in vacuo. The crude solid was recrystallised
using ethyl acetate (150 mL) to afford the title compound as a white solid in 63% yield, 33.4 g.
1H NMR (400 MHz, DMSO) 6: 1.51 (d, 3H), 3.68-3.79 (m, 1H), 3.81-3.93 (m, 3H), 4.93-5.06 (m, 2H), 7.55-
7.63 (m, 2H), 7.67-7.75 (m, 2H), 8.41-8.49 (m, 2H), 8.73 (d, 1H), 8.98 (s, 1H), 9.00 (d, 1H), 9.44 (s, 1H),
.72 (s, 1H); LCMS (System 1): Rt = 4.53 min; m/z 484 [M+H]+.
W0 2012/137089 49
Example 10: N-[5-({7-[(1S)—2-Hydroxy—1-methy|ethy|]-7H-pyrro|o[2,3-d]pyrimidinyl}carbonyl)pyridin
y|][5-methyl(trifluoromethy|)-1H-pyrazolyl]acetamide
K|N\ FF
% Hydrochloric acid in 1,4- dioxane (0.2 mL) was added to Preparation 66 (59 mg, 0.098 mmol) in
THF (2 mL) and the mixture was stirred at room temperature for 18 hours. The mixture was evaporated in
vacuo and triturated with pentane:diethyl ether (3:1, 1 mL) to afford the title compound as an off white
solid in 86% yield, 41 mg.
LCMS (system 4): Rt = 2.85 min; m/z 488.2 [M+H]+.
Examples 11 to 16 were prepared according to the method described above for Example 10, starting
from the appropriate protected alcohol.
2-(4-chlorophenyl)-N-[5-({7-[(1S)—2-hydroxy—1-methy|ethy|]- LCMS m 4): Rt = 2.89
7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridin min; m/z 450 [M+H]+
N--[5-({7-[(18)--h-ydroxy-1methylethy|]--7H--pyrro|o[2, 3- LCMS (system 4): Rt = 2.88
d]pyrimidin--y|}carbonyl)pyridin--y-|]-[3-(trifluoromethyl)— min; m/z 474 [M+H]+
1H--p razol- 1-
N--[5-({7-[(18)--h-ydroxy-1methylethy|]-H-pyrro|o[2, 3- LCMS (system 4): Rt = 2.68
d]pyrimidin--y|}carbonyl)pyridin--y-|]-[4-(trifluoromethyl)— min; m/z 475 [M+H]+
1H- 1 ,2 ,3-triazol- 1-
2--(-5chloropyridin--y-|)-N--[5-({7-[(18)--hydroxy LCMS (system 4): Rt = 2.72
ethy|]-H-p,yrro|o[23--d]pyrimidin--y|}carbony|)pyridin- min; m/z 451 [M+H]+
2--(-2H-benzotriazol-y-|)-N--[5-({7-[(18)--hydroxy LCMS m 4): Rt = 2.64
methylethy|]-H-p,yrro|o[23--d]pyrimidin--y|}carbony|)pyridin- min; m/z 457 [M+H]+
2-(,24--difluorophenyl)—-N--[5-({7-[(18)--hydroxy LCMS m 4) Rt = 2.74
methylethy|]-H-p,yrro|o[23--d]pyrimidin--y|}carbony|)pyridin- min; m/z 452 [M+H]+
Example 17: N-[5-({7-[(1R)—2-Hydroxy—1-methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridin
y|][5-methyl(trifluoromethy|)-1H-pyrazolyl]acetamide
HOJqu Me
(5-Methy|trif|uoromethyl-pyrazoly|)acetic acid (46.8 mg, 0.225 mmol) was added to (R,S) (5-
aminopyridinyl){7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethy|]-7H-pyrro|o[2,3-d]pyrimidin
yl}methanone (66 mg, 0.173 mmol ) (see ation 36), 1-propylphosphonic acid cyclic anhydride
(0.31 mL, 0.519 mmol ) and DIPEA (0.09mL, 0.606 mmol) in THF (5 mL). The mixture was heated at
reflux for 48 hours, evaporated in vacuo and partitioned between saturated aqueous sodium onate
(5 mL) and ethyl acetate (5 mL). The organic phase was dried over sodium sulfate, evaporated in vacuo
and the residue was purified by column chromatography on silica gel (gradient of EtOAc: Hexane 85: 15 )
to afford the intermediate as an off white solid in 53% yield, 52 mg.
% Hydrochloric acid in 1,4- dioxane (0.4 mL) was added to the intermediate (52 mg, 0.091 mmol) in
THF (2 mL) and the mixture was stirred at room temperature for 1.5 hours. The mixture was evaporated in
vacuo and triturated with e:diethyl ether (3:1, 1 mL) to afford the title compound as an off white
solid in 94% yield, 42 mg.
1H NMR (400 MHz, DMSO) 6: 1.49 (d, 3H), 2.32 (d, 3H), 3.56 (m, 1H), 5.00 (m, 1H), 5.20 (s, 2H), 6.56 (s,
H), 8.45 (s, 1H), 8.54 (s, 1H), 8.79 (s, 1H), 9.02 (s, 2H), 9.48 (s, 1H), 11.05 (s, 1H);
LCMS(system 4): R = 2.86 min; m/z 488 [M+H]+.
Examples 18 to 24 were prepared according to the method described above for Example 17, starting
from (5-aminopyridinyl){7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}methanone (see Preparation 36).
2--(-4cyclopropyl- 1H- 1 ,2 ,3-triazol- 1--y-|)-N--[5-({7-[(1R)— LCMS (system 4): Rt = 2.21
2--h-ydroxy-1methy-lethyl]-H-pyrrolo[2, 3--d]pyrimidin- min; m/z 474 [M+H]+
2--(5-f|uoropyridin--y|)--N--[5-({7-[(1R)—-hydroxy LCMS (system 4): Rt = 2.24
methylethyl]--7H-p—yrrolo[2, 3--d]pyrimidin- min; m/z 435 [M+H]+
N--[5-({7-[(1R)—-hydroxymethylethyl]-7H- LCMS m 4): Rt = 2.98
pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyl] min; m/z 484 [M+H]+
N-[5-({7-[(1R)—2-hydroxymethylethyl]-7H- LCMS (system 4): Rt = 2.70
pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyl] min; m/z 475 [M+H]+
N--[5-({7-[(1R)—hydroxy1-1i-methylethyl]--7H- LCMS (system 4): Rt = 2.80
pyrrolo[2, 3--d]pyrimidin--yl}carbonyl)pyridin--y-|] min; m/z 474 [M+H]+
2--(5-chloropyridin--y|)--N--[5-({7-[(1R)—-hydroxy LCMS (system 4): R = 2.49
methylethyl]--7H-p—yrrolo[2, yrimidin- min; m/z 451 [M+H]+
chlorophenyl)--N--[5-({7-[(1R)—-hydroxy LCMS (System 2): R = 1.01
methylethyl]--7H-p—yrrolo[2, 3--d]pyrimidin- min; m/z 450 [M+H]+
Example 25: (R,S) 2-(4-CyclopropyI-1H-pyrazoIy|)-N-(5-{[7-(2-hydroxymethy|ethy|)-7H-pyrro|o[2,3-
d]pyrim idiny|]carbony|}pyridiny|)acetamide
The title compound was prepared according to the method bed for Example 9 using (R,S) (5-
aminopyridiny|){7-[2-{[tert-buty|(dimethy|)si|y|]oxy}methy|ethy|]-7H-pyrro|o[2,3-d]pyrimidin
yl}methanone (see Preparation 39) and (4-cyc|opropy|-1H-pyrazoIy|)acetic acid (see Preparation 88)
to afford the title compound as a white solid in 14% yield, 20 mg.
1H NMR (400 MHz, DMSO) 6: 0.46 (d, 2H), 0.79 (d, 2H), 1.49 (d, 3H), 1.70 (m, 1H), 3.73 (m, 1H), 3.87
(m, 1H), 5.00 (s, 4H), 7.26 (s, 1H), 7.54 (s, 1H), 8.40 (s, 1H), 8.47 (s, 1H), 8.75 (d, 1H), 8.98 (s, 1H), 9.00
(d, 1H), 9.44 (s, 1H), 10.75 (s, 1H); LCMS (system 4): R = 2.53 min; m/z 445 .
Examples 26 to 33 were prepared according to the method described above for Example 10, starting
from the appropriate ted a|coho| TBDMS ether.
N--(-5{[7-(2-hydroxy—1, 1--d-imethy|ethy|)Hp-y,rro|o[2 3- LCMS (system 4): 2.89
d]pyrimidin--y|]carbony|}pyridin--y-|)-[5-methyl- min; m/z 502 [M+H]+t
N--(-5{[7-(2-hydroxy—1, 1--dimethy|ethy|)-Hp-yrro|o[2,3- LCMS (system 4): R = 3.03
d]pyrimidiny|]carbony|}pyridiny|)[4- min; m/z 498 [M+H]+
2-(4-ch|oropheny|)-N-(5-{[7-(2-hydroxy-1,1-dimethy|ethy|)- H NMR (400 MHz, DMSO) 6.
7H-pyrro|o[2,3-d]pyrim idiny|]carbony|}pyridin 1.73 (s, 6H), 3.75 (s, 2H), 3.96
y| )acetamide (s, 2H), 7.39 (s, 4H), 8.22 (s,
H), 8.51 (s, LCMS (system 4):
t: 3.02 min; m/z 464.1
[M+H]+
2-(5-ch|oropyridiny|)-N-(5-{[7-(2-hydroxy-1,1-
dimethylethy|)-7H-pyrro|o[2,3-d]pyrimidin LCMS m 4): R = 2.72
y|]carbony|}pyridiny|)acetamide min; m/z 465 [M+H]+
N--(-5{[7-(2-hydroxy—1, 1--d-imethy|ethy|)H-pyrro|o[2, 3- LCMS (system 4): R = 2.80
d]pyrimidin--y|]carbony|}pyridin--y-|)-[3-(trifluoromethy|)- min; m/z 488 [M+H]+
1H--p razol- 1-
2--(-4cyc|opropy|- 1H- 1 ,2 ,3-triazol- 1--y-|)-N--(-5{[7-(2-hydroxy- LCMS m 4): R: 2.47
1 ,1--dimethy|ethy|)-H-p,yrro|o[23--d]pyrimidin- min; m/z 461 [M+H]
N--(-5{[7-(2-hydroxy—1, 1--dimethy|ethy|)-H-p,yrro|o[2 3- LCMS (system 4): R = 2.78
d]pyrimidin--y|]carbony|}pyridin--y-|)-[4-(trifluoromethy|)- min; m/z 489 [M+H]+
1H- 1 ,2 ,-3triazol- 1-
2--(-4cyc|opropy|- yrazo| 1--y-|)-N--(-5{[7-(2-hydroxy-1,1- LCMS(system 4): R = 2.68
dimethylethy|)--7H--p,yrro|o[23--d]pyrimidin- min; m/z 460 [M+H]+
Example 34: N-(5-{[2-Amino(2-hydroxymethy|ethy|)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin-
3-yl)(4-ch|oropheny|)acetamide iomer 1)
4-Chlorophenylacetic acid (25 mg, 0.14 mmol) was added to [2-amino(2-hydroxymethy|ethy|)-7H-
pyrrolo[2,3-d]pyrimidinyl](5-aminopyridinyl)methanone (50 mg, 0.16 mmol) (see Preparation 57)
and HATU (91 mg, 0.24 mmol) in pyridine (2 mL). The mixture was stirred at room temperature for 16
hours. ted aqueous sodium bicarbonate (5 mL) was added then extracted with ethyl acetate (3 x 5
mL). The combined organic phases were washed with brine (5 mL) then dried over sodium sulfate and
evaporated in vacuo. The residue was ed by preparative TLC (95:5 DCM:MeOH) to afford the title
nd as a yellow solid in 48% yield, 32 mg.
LCMS (system 5): Rt = 2.90 min; m/z 465 [M+H]+.
Examples 35 to 45 were prepared according to the method described above for Example 34, starting
from [2-amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl](5-aminopyridin
y|)methanone (see Preparation 57, omer1 or Preparation 59, enantiomer 2) and the appropriate
acids.
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 1
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)(5- LCMS (system 5): Rt = 2.63
min; m/z 466 [M+H]+
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 1
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)[4- LCMS (system 5): Rt = 2.88
min; m/z 499 [M+H]+
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 1
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)(5- LCMS (system 5): Rt = 2.25
min; m/z 450 [M+H]+
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 1
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)[3- LCMS (system 4): Rt = 2.39
trifluorometh | -1H-p razol |]acetamide min; m/z 489 [M+H]+
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 1
o[2,3-d]pyrimidinyl]carbonyl}pyridiny|)[4- LCMS (system 5): Rt = 2.61
trifluorometh | -1H-1,2,3-triazol | acetamide min; m/z 490 M+H
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 2
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)(5- LCMS (system 4): Rt = 1.89
fluorop ridin | acetamide min; m/z 450 [M+H]+
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 2
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)(5- LCMS (system 4): Rt = 2.16
chlorop ridin | acetamide min; m/z 466 [M+H]+
N-(5-{[2-amino(2-hydroxymethylethyl)—7H- Enantiomer 2
o[2,3-d]pyrimidinyl]carbonyl}pyridiny|)(4- LCMS (system 4): Rt = 2.63
chlorophen | acetamide min; m/z 465 [M+H]+
N-(5-{[2-amino—7-(2-hydroxymethy|ethy|)-7H- omer 2
o[2,3-d]pyrimidinyl]carbonyl}pyridiny|)[4- LCMS (system 4): Rt = 2.73
min; m/z 499 [M+H]+
N-(5-{[2-amino—7-(2-hydroxymethy|ethy|)-7H- Enantiomer 2
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)[3- LCMS (system 4): Rt = 2.38
orometh | -1H-p razol ' min; m/z 489 [M+H]+
N-(5-{[2-amino—7-(2-hydroxymethy|ethy|)-7H- Enantiomer 2
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|)[4- LCMS (system 5): Rt = 2.60
trifluorometh | -1H-1,2,3-triazol ' min; m/z 490 [M+H]+
e 46: N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)(5-ch|oropyridinyl )acetam ide
\ /N o
N/ \ H \ /
)\\ I
H2N N
Me)$Me
oropyridinyl)acetic acid (26.1 g, 152 mmol) (see Preparation 90) was added to (5-aminopyridin-
3-y|){7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethy|ethy|)[(2,4-dimethoxybenzyl)amino]—7H-pyrrolo[2,3-
d]pyrimidinyl}methanone (75.0 g, 130 mmol ) (see Preparation 51), ylphosphonic acid cyclic
anhydride (187 mL, 317 mmol, 50% solution in EtOAc) and triethylamine (61.9 mL, 444 mmol ) in THF
(423 mL). The mixture was stirred at 25°C for 2 hours then ted aqueous sodium bicarbonate (400
mL) was added and the organic layer was separated. The aqueous phase was extracted with EtOAc (400
mL) and all organic phases were combined and dried over sodium sulfate then evaporated in vacuo.
The residue brown solid was dissolved in trifluoroacetic acid (300 mL) and the solution was stirred at 50°C
for 3 hours then evaporated in vacuo. Methanol (1800 mL) was added to the residue and the mixture was
filtered. The filtrate was evaporated in vacuo and azeotroped with ethanol (3 x 200 mL).
Potassium carbonate (87.7 9, mmol) was added to the crude trifluoroacetamide in methanol (300 mL) and
the mixture was stirred at room temperature for 16 hours. The mixture was poured into water (2000 mL)
and filtered. The solid was washed with water (200 mL) then triturated with ethanol (2 x 200 mL at room
temperature then 380 mL at 50°C) to afford the title compound as a yellow solid in 48% yield, 29.9 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.64 (s, 6H), 3.90 (d, 2H), 3.95 (s, 2H), 5.05 (t, 1H), 6.54 (br s, 2H), 7.49
(d, 1H), 7.69 (s, 1H), 7.92 (dd, 1H), 8.40 (m, 1H), 8.56 (m, 1H), 8.64 (d, 1H), 8.94 (d, 1H), 8.96 (s, 1H),
.71 (s, 1H); LCMS (System 3): R: 9.92 min; m/z 480 [M+H]+.
Example 47: N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)[5-methyl(trifluoromethy|)-1H-pyrazolyl]acetamide
\ / OMe
N/ N4, — F
I \ ” N‘ /
x N
N F
H2N N F
Me>§Me
The title compound was prepared according to the method described for Example 46 using (5-
yridinyl){7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)[(2,4-dimethoxybenzyl)amino]—
7H-pyrrolo[2,3-d]pyrimidinyl}methanone (see ation 51) and (5-methyltrifluoromethyl-pyrazol-
1-yl)acetic acid (46.8 mg, 0.225 mmol) to afford the title compound as a brown solid in 79% yield, 82 mg.
LCMS (System 1): R = 2.83 min; m/z 517 [M+H]+.
Examples 48 to 53 were prepared according to the method described above for Example 34, starting
from [2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl](5-aminopyridin
yl)methanone (see Preparation 48) and the appropriate acids.
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)—7H- LCMS (system 5):
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|) R: 3.12 min; m/z
513 M+H
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)—7H- LCMS (system 5):
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|) R: 2.89 min; m/z
479 [M+H]+
[2-amino(2-hydroxy-1,1-dimethylethyl)—7H- MS (ESCI): m/z
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|) 481 [M+H]+
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)—7H- LCMS (system 5):
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|) R: 2.42 min; m/z
-fluorop ridin l acetamide 464 [M+H]+
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)—7H- LCMS m 4):
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|) R: 2.54 min; m/z
[3- trifluorometh l-1H-p 1- l]acetamide 503 [M+H]+
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)—7H- LCMS (system 4)
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridiny|) R = 2.23 min; m/z
3-c cloprop l-1H-p razol lacetamide 475 [M+H]+
Example 54: N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)[4-(trifluoromethyl)-1H-1,2,3-triazolyl]acetamide
\ / o F
Jk/ — F
I \ N N‘N’zN
)\\ N
HO
The title compound was ed according to the method described for Example 9 using [7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)amino-7H-pyrrolo[2,3-d]pyrimidinyl]{5-aminopyridin
hanone (see Preparation 48a) and [4-(trifluoromethyl)-1H-1,2,3-triazolyl]acetic acid (see
Preparation 81) to afford the title compound as a brown solid in 85% yield, 62 mg.
1H NMR (400 MHz, DMSO-d6) 6: 1.63 (s, 6H), 3.89 (d, 2H), 5.01 (br, 1H), 5.56 (s, 2H), 6.67 (br s, 2H),
7.72 (s, 1H), 8.36 (s, 1H), 8.70 (s, 1H), 8.94-8.96 (m, 3H), 11.05 (s, 1H); LCMS (system 5): R: 2.71 min;
m/z 502 .
Example 55: N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)(2H-benzotriazolyl)acetamide
\ / O
/ N NI
1 | \ H ‘N/
\N N
Me>$Me
H0
The title compound was ed according to the method described for Example 9 using {2-amino
[1 ,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}(5-aminopyridin
yl)methanone (see Preparation 49) to afford the title compound as a brown solid in 46% yield, 20 mg.
1H NMR (400 MHz, DMSO-d6) 6: 1.63 (s, 6H), 3.88 (d, 2H), 5.04 (t, 1H), 5.80 (s, 2H), 6.54 (s, 2H), 7.46-
7.49 (m, 2H), 7.69 (s, 1H), 7.95-7.97 (m, 2H), 8.35 (s, 1H), 8.69 (s, 1H), 8.96 (s, 1H), 8.98 (s, 1H), 11.06
(s, 1H); MS (ESCI): m/z 486 [M+H]+.
Example 56: N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)(4-cyclopropyl-1H-1,2,3-triazolyl)acetamide
The title nd was prepared according to the method described for Example 10 using N-[5-({2-
amino[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyridinyl](4-cyclopropyl-1H-1,2,3-triazolyl)acetamide (see Preparation 52) to afford
the title compound as a yellow solid in 24% yield, 14 mg.
1H NMR (400 MHz, DMSO-D6) 6: 0.71-0.72 (m, 2H), 0.89-0.91 (m, 2H), 1.64 (s, 6H), 1.94-1.99 (m, 1H),
3.89 (d, 2H), 5.07 (t, 1H), 5.31 (s, 2H), 6.53 (s, 2H), 7.69 (s, 1H), 7.86 (s, 1H), 8.35 (s, 1H), 8.65 (s, 1H),
8.95 (m, 2H), 11.04 (s, 1H); LCMS (system 5): R: 2.42 min; m/z 476 [M+H]+.
Examples 57 to 64 were prepared according to the method described above for Example 1, starting from
(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)(5-aminopyridinyl)methanone (see Preparation
65) and the appropriate acids.
N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5): R:
yl)carbonyl]pyridinyl}(3-cyclopropyl-1H-pyrazol 2.90 min; m/z 459 [M+H]+
N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5, 12 min
yl)carbonyl]pyridinyl}(4-cyclopropyl-1H-1 ,2,3-triazol run): R = 6.13 min; m/z 460
[M+H]+
LCMS (system 4): R = 2.60
min; m/z 448 [M+H]+
N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5): R = 2.99
yl)carbonyl]pyridinyl}[4-(trifluoromethyl)-1H-pyrazol-1 min; m/z 487 [M+H]+
|]acetamIde
(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5): R = 3.04
yl)carbonyl]pyridinyl}[4-(trifluoromethyl)-1H-1,2,3- min; m/z 488 [M+H]+
triazol |]acetamide
N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5): R = 3.09
yl)carbonyl]pyridinyl}[3-(trifluoromethyl)-1H-pyrazol-1 min; m/z 487 [M+H]+
amide
N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5): R = 2.95
lcarbon |]p ridin l 5-chlorop ridin lacetamide min; m/z 464 [M+H]+
N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 5): R = 3.09
yl)carbonyl]pyridinyl}[5-methyl(trifluoromethyl)-1H- min; m/z 501 [M+H]+
amide
The following Examples were prepared according to Method a le 34 at 50°C) or Method b
(Example 1 using DIPEA) as described above starting from (5-Aminopyridinyl)(7-isopropyl-7H-
pyrrolo[2,3-d]pyrimidinyl)methanone ration 95) and the riate acid.
65 2-(4-fluorophenyl)-N-{5-[(7-isopropyl-7H- LCMS (system 1): R = 2.73 min; m/z
pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin 418[M+H]+.
yl}acetamide
‘N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 9): R = 3.22 min; m/z
yl)carbonyl]pyridinyl}[3- 468 [M+H]+.
(trifluoromethyl)phenyl]acetamide
67 N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 9): R = 3.22 min; m/z
yl)carbonyl]pyridinyl}[4- 468 [M+H]+.
(trifluoromethyl)phenyl]acetamide
2-(3,4-dichlorophenyl)-N-{5-[(7-isopropyl-7H- LCMS (system 7): R = 11.34 min; m/z
pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin 468 [M+H]+
yl}acetamide
57 2012/051363
2-(4-chlorophenyl)-N-{5-[(7-isopropy|-7H- m/z 434 [M+H]+
pyrro|o[2,3-d]pyrim idiny|)carbony|]pyridin
yl}acetam ide
\lO 2-[2-(cyclopropyloxy)phenyl]-N-{5-[(7-isopropy|- LCMS m 4): R = 3.13 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 456 [M+H]+
y|)carbony|]pyridiny|}acetamide Prep HPLC (method 2)
Using [2-(cyclopropyloxy)pheny|]acetic
acid
Preparation 160 .
\l _\ 2-(4-cyanophenyl)-N-{5-[(7-isopropy|-7H- LCMS (system 2): R = 1.16 min; m/z
pyrro|o[2,3-d]pyrim idiny|)carbony|]pyridin 425 [M+H]+
yl}acetam ide
\lN 2-[4-cyano(trifluoromethyl)pheny|]-N-{5-[(7- LCMS (system 1): R = 2.82 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 493 [M+H]+
y|)carbony|]pyridiny|}acetamide Using [4-cyano
(trifluoromethy|)pheny|]acetic acid
(Preparation 164).
\l (JO 2-[4-(cyclopropyloxy)phenyl]-N-{5-[(7-isopropy|- LCMS (System 3) R = 3.05 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 456 [M+H]+
y|)carbony|]pyridiny|}acetamide Using [4-(cyclopropyloxy)pheny|]acetic
acid (Preparation 161).
Prep method 2
N4; cyclopropyloxy)phenyl]-N-{5-[(7-isopropy|- LCMS (system 1) R = 2.52 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 456 [M+H]
y|)carbony|]pyridiny|}acetamide Using [3-(cyclopropyloxy)pheny|]acetic
acid (Preparation 162).
2-[3-(hydroxymethyl)pheny|]-N-{5-[(7-isopropy|- LCMS (System 2): R = 1.3 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 430 [M+H]+
bony|]pyridiny|}acetamide
\l O) 2-(4-cyanofluorophenyl)-N-{5-[(7-isopropy|- LCMS (System 2): R = 1.5 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 443 [M+H]+
y|)carbony|]pyridiny|}acetamide
\l \l 2-(4-cyanomethoxyphenyl )-N-{5-[(7- LCMS m 2): R = 1.3 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 455 [M+H]+
y|)carbony|]pyridiny|}acetamide
\l (D 2-(6-fluorooxo-1,3-dihydro—2H-isoindoIy|)- LCMS (System 2): R = 1.5 min; m/z
(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin 473 [M+H]+
y|)carbony|]pyridiny|}acetamide
\l (O N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 2): R = 1.2 min; m/z
y|)carbony|]pyridiny|}(3-pyridiny|-1H- 467 [M+H]+.
pyrazoIy|)acetamide
2-(1H-benzimidazoIyl)-N-{5-[(7-isopropyI-7H- LCMS (System 2): R = 1.0 min; m/z
pyrro|o[2,3-d]pyrim idiny|)carbony|]pyridin 440 [M+H]+
tam ide
CO _\ N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 2): R = 1.1 min; m/z
y|)carbony|]pyridiny|}quinoxa|in 452 [M+H]+
ylacetam ide
00N N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 9): R: 2.63 min; m/z
y|)carbony|]pyridiny|}(1,5-naphthyridin 452 [M+H]+
yl)acetamide Using 1,5-naphthyridinylacetic acid
(Preparation 188).
WO 37089 58
2-(3-amino-1,2-benzisoxazoIyl)-N-{5-[(7- LCMS (System 9): R: 2.76 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 456 [M+H]+ using (3-amino-
y|)carbony|]pyridiny|}acetamide benzo[d]isoxazolyl)-acetic acid ethyl
ester (Preparation 129)
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 9): R: 2.90 min; m/z
y|)carbony|]pyridiny|}[2-(trif|uorom ethyl)- 508 [M+H]+
1H-benzimidazoIy|]acetamide
CO 01 2-imidazo[1,2-a]pyridiny|-N-{5-[(7-isopropyl- LCMS (System 10): R: 2.57 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 440 [M+H]+ using imidazo[1,2-
y|)carbony|]pyridiny|}acetamide a]pyridiny|-acetic acid (Preparation
132)
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 9): R: 2.54 min; m/z
y|)carbony|]pyridiny|}(1H-pyrazolo[4,3- 441 [M+H]+
diny|)acetamide Using 1H-pyrazolo[4,3-b]pyridin
y|)acetic acid (Preparation 190).
CD \I N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 9): R = 2.74 min; m/z
y|)carbony|]pyridiny|}[3- 478 [M+H]+
(methylsulfonyl)pheny|]acetamide
2-(4-cyclopropyI-1H-1,2,3-triazoly|)-N-{5-[(7- LCMS (System 9): R = 2.68 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 431 .
y|)carbony|]pyridiny|}acetamide Using 4-cyclopropyI-1H-1,2,3-triazol
ic acid (Preparation 83).
2-(1,3-benzoxazoIyl)-N-{5-[(7-isopropy|-7H- LCMS (System 9): R = 2.79 min; m/z
pyrrolo[2,3-d]pyrim idiny|)carbonyl]pyridin 441.1 [M+H]+
yl}acetam ide
2-(3-cyclopropyI-1H-pyrazoIy|)-N-{5-[(7- LCMS (System 9): R = 2.75 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 430 [M+H]+
y|)carbony|]pyridiny|}acetamide (3-cyclopropyI-1H-pyrazoIy|)acetic
acid (Preparation 80).
(D _\ 2-[5-(cyclopropyloxy)pyrid |]-N-{5-[(7- LCMS (system 3) R = 2.36 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 457 [M+H]+
y|)carbony|]pyridiny|}acetamide Prep method 2
Using [5-(cyc|opropy|oxy)pyridin
|]acetic acid Preparation 163 .
(DN 2-(2,3-dihydrobenzofuranyl)—N-{5-[(7— LCMS (system 3): R = 2.91 min; m/z
pyI-7H-pyrrolo[2,3-d]pyrimidin 442 [M+H]+
bony|]pyridiny|}acetamide Prep HPLC (method 1)
(9 0O 2-(2H-indazoIyl)-N-{5-[(7-isopropy|-7H- LCMS (system 3): R = 2.74 min; m/z
pyrrolo[2,3-d]pyrim idiny|)carbonyl]pyridin 440 [M+H]+
yl}acetam ide Prep HPLC (method 1)
(9 4s 2-(5-f|uoro-2H-indazoIyl)-N-{5-[(7-isopropy|- LCMS (system 9): R: 2.96 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 458 [M+H]+
y|)carbony|]pyridiny|}acetamide Using -(5-f|uoro-2H-indazoIy|)acetic
acid
Preparation 174 .
WO 37089 59
(9 U1 2-(5-fluoro-1H-indazoIy|)-N-{5-[(7-isopropy|- LCMS (system 9): R: 2.97 min; m/z
rolo[2,3-d]pyrimidin 458 [M+H]+
y|)carbony|]pyridiny|}acetamide Using (5-fluoro-1H-indazoIy|)acetic
acid (Preparation 172).
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.62 min; m/z
y|)carbony|]pyridiny|}[5-methy| 472 [M+H]+
(trifluorom ethyl )-1 H-pyrazoI-1 -y|]acetam ide
(9 \l (7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 3): R = 2.97 min; m/z
y|)carbony|]pyridiny|}[3-(trif|uorom ethyl)- 458 [M+H]+
1H-pyrazoIy|]acetamide Prep HPLC (method 1)
2-(5-chloropyridiny|)-N-{5-[(7-isopropy|-7H- LCMS (system 9): R = 2.86 min; m/z
pyrrolo[2,3-d]pyrim idiny|)carbony|]pyridin 435 [M+H]+
yl}acetam ide Using (5-ch|oropyridiny|)acetic acid
Preparation 90 .
2-(1H-indazoIyl)-N-{5-[(7-isopropy|-7H- m/z 440 [M+H]+
pyrrolo[2,3-d]pyrim idiny|)carbony|]pyridin Using 1H-indazoIylacetic acid
yl}acetam ide (Preparation 182).
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 7): R = 8.73 min; m/z
y|)carbony|]pyridiny|}quino|in 451 [M+H]+
am ide
2-(7-f|uoro-2H-indazoIyl)-N-{5-[(7-isopropy|- LCMS m 9): R: 2.95 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 458 [M+H]+
y|)carbony|]pyridiny|}acetamide Using (7-fluoro-2H-indazoIyl)acetic
acid (Preparation 178).
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 9): R: 2.72 min; m/z
y|)carbony|]pyridiny|}(1H-pyrazolo[3,4- 441 [M+H]+
b]pyridiny|)acetamide Using 1 H-pyrazolo[3,4-b]pyridin
y|)acetic acid (Preparation 180).
2-(1H-indazoIy|)-N-{5-[(7-isopropy|-7H- LCMS (system 3): R = 2.82 min; m/z
o[2,3-d]pyrimidiny|)carbony|]pyridin 440 [M+H]+
yl}acetamide Prep HPLC (method 1)
2-(3-isopropyImethyl-1H-pyrazoIy|)-N-{5- LCMS (system 8): R = 1.65 min; m/z
[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin 446 [M+H]+
y|)carbony|]pyridiny|}acetamide Using (3-isopropyImethyI-1H-
pyrazoIy|)acetic acid (Preparation
167).
(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.52 min; m/z
y|)carbony|]pyridiny|}(4-isopropy|-1H- 433 [M+H]+
1,2,3-triazo|y|)acetamide
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 9): R = 2.98 min; m/z
y|)carbony|]pyridiny|}[4-(trif|uorom ethyl)- 459 [M+H]+
1H-1,2,3-triazo|y|]acetamide Using [4-(trifluoromethy|)-1H-1,2,3-
triazoIy|]acetic acid ration
81).
2-(7-fluoro-1H-indazoIy|)-N-{5-[(7-isopropy|- LCMS (system 9): R = 2.96 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 458 [M+H]+
y|)carbony|]pyridiny|}acetamide Using (7-fluoro-1H-indazoIy|)acetic
acid
Preparation 176 .
2-(2H-benzotriazoIyl)-N-{5-[(7-isopropy|-7H- LCMS (system 8): R = 1.57 min; m/z
pyrrolo[2,3-d]pyrim idiny|)carbony|]pyridin 441 [M+H]+
yl}acetam ide
W0 2012/137089 60
2-(7-fluoro—2-methyI-1H-indoIy|)-N-{5-[(7- LCMS (system 8): R 1.64 min; m/z
isopropyI-7H-pyrro|o[2,3-d]pyrimidin 471 [M+H]+
y|)carbonyl]pyridinyl}acetamide
2-(5-chloro—2-methyI-1H-indoIy|)-N-{5-[(7- LCMS (system 8): R 1.69 min; m/z
isopropyI-7H-pyrro|o[2,3-d]pyrimidin 487 [M+H]+
y|)carbonyl]pyridinyl}acetamide
2-(1H-indoIy|)-N-{5-[(7-isopropy|—7H- LCMS (system 8): R 1.66 min; m/z
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin 439 [M+H]+
y|}acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS m 9): R = 3.13 min; m/z
y|)carbonyl]pyridiny|}[1-isopropy| 500 [M+H]+
(trifluoromethy|)-1H-pyrazoIy|]acetamide Using 1-isopropyI(trifluoromethyl)-
azoIy|]acetic acid
(Preparation 185).
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.50 min; m/z
y|)carbonyl]pyridiny|}(3-methy|-1H- 404 [M+H]+
pyrazoIy|)acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.45 min; m/z
y|)carbonyl]pyridiny|}(1H-pyrazoI 390 [M+H]+
y|)acetamide
-dimethy|-1H-pyrazoIy|)-N-{5-[(7- LCMS (system 8): R = 1.55 min; m/z
isopropyI-7H-pyrro|o[2,3-d]pyrimidin 418 [M+H]+
y|)carbonyl]pyridinyl}acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.62 min; m/z
y|)carbonyl]pyridiny|}(2-methy|-1H-indoI 453 [M+H]+
y|)acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.57 min; m/z
yl)carbonyl]pyridiny|}(5-methoxy-1H-indol- 469 [M+H]+
3-y|)acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.35 min; m/z
y|)carbonyl]pyridinyl}pyridin 401 [M+H]+
ylacetamide
2-(5-chloro—1H-benzotriazoIy|)-N-{5-[(7- LCMS m 8): R = 1.61 min; m/z
isopropyI-7H-pyrro|o[2,3-d]pyrimidin 475 [M+H]+
y|)carbonyl]pyridinyl}acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.61 min; m/z
y|)carbonyl]pyridiny|}[3-(2-thieny|)-1H- 472 [M+H]+
pyrazoIy|]acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS m 8): R = 1.37 min; m/z
bonyl]pyridiny|}(7- 455 [M+H]+
methylimidazo[1,2-a]pyrimidinyl)acetamide
2-(2,6-dimethyl-9H-puriny|)-N-{5-[(7- LCMS (system 8): R = 1.43 min; m/z
isopropyI-7H-pyrro|o[2,3-d]pyrimidin 470 [M+H]+
y|)carbonyl]pyridinyl}acetamide
WO 37089 61 2012/051363
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.60 min; m/z
y|)carbonyl]pyridiny|}(6-nitro-1H-indazol- 485 [M+H]+
1-y|)acetam ide
2-(5-chloro—1-methyI-1H-indazoIy|)-N-{5-[(7- LCMS (system 8): R = 1.73 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 488 [M+H]+ J. Med Chem.
y|)carbonyl]pyridinyl}acetamide 1992, 35, 2155-2165
2-(5-fluoro—2-methyI-1H-indoIy|)-N-{5-[(7- LCMS (system 8): R = 1.63 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 471 [M+H]+
y|)carbonyl]pyridinyl}acetamide
2-(6-chloroimidazo[1,2—a]pyridinyl)—N-{5-[(7- LCMS (system 8): R = 1.45 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 474 [M+H]+
y|)carbonyl]pyridinyl}acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.36 min; m/z
y|)carbonyl]pyridiny|}(2-methy|-1H- 454 [M+H]+
benzimidazoIy|)acetamide
2-imidazo[1,2-a]pyridinyI-N-{5-[(7-isopropyl- LCMS (system 8): R = 1.31 min; m/z
7H-pyrro|o[2,3-d]pyrimidin 440 [M+H]+
y|)carbonyl]pyridinyl}acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS m 8): R = 1.49 min; m/z
y|)carbonyl]pyridiny|}(5-methoxy-1H- 470 [M+H]+
pyrro|o[3,2-b]pyridiny|)acetamide
(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.49 min; m/z
y|)carbonyl]pyridiny|}(5-methy|-1H- 404 [M+H]+
pyrazoIy|)acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.43 min; m/z
y|)carbonyl]pyridiny|}(7-methy|-1H- 454 [M+H]+
benzimidazoIy|)acetamide
2-(1H-indoIyl)-N-{5-[(7-isopropy|—7H- LCMS (system 8): R = 1.59 min; m/z
pyrro|o[2,3-d]pyrim idinyl)carbonyl]pyridin 439 [M+H]+
yl}acetam ide
2-(1H-benzimidazoIy|)-N-{5-[(7-isopropy|—7H- LCMS (system 8): R = 1.40 min; m/z
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin 440 [M+H]+
yl}acetamide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.45 min; m/z
y|)carbonyl]pyridiny|}(3-methy|-1H- 404 [M+H]+
pyrazoIy|)acetamide
2-(1H-benzotriazoIy|)-N-{5-[(7-isopropyI-7H- LCMS (system 8): R = 1.52 min; m/z
pyrro|o[2,3-d]pyrim idinyl)carbonyl]pyridin 441 [M+H]+
yl}acetam ide
N-{5-[(7-isopropy|—7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.68 min; m/z
y|)carbonyl]pyridiny|}(1-methy|-1H-indoI 453 [M+H]+
yl)acetamide
(7-i80propyI-7H-pyrrolo[2,3-d1pyrimidin—5— LCMS (system 8): R = 1.66 min; m/z
y|)carbonyl]pyridiny|}(4-pheny|-1H-pyrro|_ 465 [M+H]+
2-y|)acetam ide
2-(2-ethyI-3H-imidazo[4,5-b]pyridiny|)-N-{5- LCMS (system 8): R = 1.49 min; m/z
[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin 469 [M+H]+
bony|]pyridiny|}acetamide
2-(5-chloro-1H-indoIyl)-N-{5-[(7-isopropy|- LCMS (system 8): R = 1.67 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 473 [M+H]+
y|)carbony|]pyridiny|}acetamide
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.71 min; m/z
y|)carbony|]pyridiny|}(1-pheny|-1H-pyrro|_ 465 [M+H]+ Can be prepared by a
3-y|)acetam ide similar method to Harrak, Y. et aI.
Bioorganic & Medicinal Chemistry
14 890.
, ,
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.63 min; m/z
y|)carbony|]pyridiny|}(5-methy|-1H-indoI 453 [M+H]+
yl)acetamide
thyI-1H-benzimidazoIy|)-N-{5-[(7- LCMS (system 8): R = 1.37 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 468 [M+H]+
y|)carbony|]pyridiny|}acetamide
2-imidazo[1,2-a]pyrimidinyI-N-{5-[(7- LCMS (system 8): R = 1.38 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 441 [M+H]+
y|)carbony|]pyridiny|}acetamide
2-(1H-indazoIyl)-N-{5-[(7-isopropy|-7H- LCMS (system 8): R = 1.55 min; m/z
pyrro|o[2,3-d]pyrim idiny|)carbony|]pyridin 440 [M+H]+
yl}acetam ide
2-(5-fluoro—7-methoxy—1H-indoIy|)-N-{5-[(7- LCMS (system 8): R = 1.67 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 487 [M+H]+
y|)carbony|]pyridiny|}acetamide
2-(1-benzyI-1H-1,2,3-triazoIyl)-N-{5-[(7- LCMS (system 8): R = 1.57 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 481 [M+H]+
y|)carbony|]pyridiny|}acetamide
2-(6-chloromethyI-1H-indazoIy|)-N-{5-[(7- LCMS (system 8): R = 1.72 min; m/z
isopropyI-7H-pyrrolo[2,3-d]pyrimidin 488 [M+H]+
y|)carbony|]pyridiny|}acetamide Using -(6-ch|oromethy|-1H-indazol-
3- | acetic acid
Preparation 170 .
2-(7-chloro-1H-indoIyl)-N-{5-[(7-isopropy|- LCMS m 8): R = 1.67 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 473 [M+H]+
y|)carbony|]pyridiny|}acetamide
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.68 min; m/z
y|)carbony|]pyridiny|}(5-methylpheny|- 480 [M+H]+
1 zoIy|)acetamide
romopyridiny|)-N-{5-[(7-isopropy|-7H- LCMS (system 8): R = 1.60 min; m/z
pyrro|o[2,3-d]pyrim idiny|)carbony|]pyridin 479 [M+H]+
yl}acetam ide
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.41 min; m/z
y|)carbony|]pyridiny|}pyridin 401 [M+H]+
ylacetamide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.55 min; m/z
yl)carbonyl]pyridinyl}(6-methoxypyridin 431 [M+H]+
yl)acetam ide
2-(2,5-dimethy|-1H-indoIy|)-N-{5-[(7- LCMS (system 8): R = 1.67 min; m/z
isopropyl-7H-pyrrolo[2,3-d]pyrimidin 467 [M+H]+
yl)carbonyl]pyridinyl}acetamide
(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.33 min; m/z
yl)carbonyl]pyridinyl}(6-methylpyrid in 415 [M+H]+
yl)acetam ide
(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.40 min; m/z
yl)carbonyl]pyridinyl}(2-methquu inol in 465 [M+H]+
yl)acetam ide Using (2-methquuinolinyl)acetic
acid (Preparation 165).
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.60 min; m/z
bonyl]pyridinyl}(5-methoxy 483 [M+H]+
methyl-1H-indolyl)acetamide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.46 min; m/z
yl)carbonyl]pyridinyl}tetrazolo[1,5- 443 [M+H]+. (can be ed via a
b]pyridazinylacetam ide similar method to W02010/129379, 11
Nov 2010
2-(5-hydroxy-1H-indolyl)-N-{5-[(7-isopropyl- LCMS (system 8): R = 1.45 min; m/z
7H-pyrrolo[2,3-d]pyrimidin 455 [M+H]+
yl)carbonyl]pyridinyl}acetamide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.47 min; m/z
yl)carbonyl]pyridinyl}(5H-pyrrolo[2,3- 441 [M+H]+
b]pyrazinyl)acetam ide
2-(5,6-dimethyl-1H-benzimidazoly|)-N-{5-[(7- LCMS (system 8): R = 1.43 min; m/z
pyl-7H-pyrrolo[2,3-d]pyrimidin 468 [M+H]+
yl)carbonyl]pyridinyl}acetamide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.48 min; m/z
yl)carbonyl]pyridinyl}(4-pyridinyl-1H- 468 [M+H]+
1,2,3-triazolyl)acetamide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.43 min; m/z
yl)carbonyl]pyridinyl}(4-methyl-1H-1,2,3- 405 [M+H]+
lyl)acetamide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.55 min; m/z
yl)carbonyl]pyridinyl}(5-methylphenyl- 480 [M+H]+
1H-pyrazolyl)acetam ide
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (system 8): R = 1.55 min; m/z
yl)carbonyl]pyridinyl}[2- 448 [M+H]+ can be prepared via a
(m ethylthio)pyrim idinyl]acetamide r method to Smrz, R. et al.
Collection of Czechoslovak Chemical
Communications (1976), 41(9),
2771-87
W0 2012/137089 64
N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 8): R = 1.44 min; m/z
y|)carbony|]pyridiny|}pyrimidin 402 [M+H]+
ylacetamide
Example 166: 2-(5-Cyanopyridiny|)-N-{5-[(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridin-
cetam ide
\ / O
/ ;N
N“ \ H \N/ KN | N
)‘Me
Zinc cyanide (28 mg, 0.23 mmol) was added to romo-pyridiny|)-N-[5-(7-(propany|)-7H-
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridiny|]-acetamide (Example 317, 75 mg, 0.16 mmol) in DMF (2
mL) and the mixture was degassed with argon for 10 minutes. Then tris(dibenzylideneacetone)dipa||adium
(3 mg, 0.003 mmol) and 1,1’-bis(diphenylphosphino)ferrocene (7 mg, 0.012 mmol) were added and the
mixture was heated at 100°C for 40 mins under microwave irradiation. The e was diluted with
EtOAc (5 mL) and washed with water (2 mL), brine (2 mL) and dried over sodium sulphate. The filtrate
was evaporated in vacuo and purified by preparative TLC (3% MeOH in DCM) to afford the title nd
as a light brown solid in 18% yield, 12 mg.
1H NMR (400 MHz, DMSO) 6: 1.55 (d, 6H), 4.07 (s, 2H), 5.09 (m, 1H), 7.67 (d, 1H), 8.30 (m, 1H), 8.44 (s,
1H), 8.52 (s, 1H), 8.74 (d, 1H), 8.98-8.99 (m, 3H), 9.44 (s, 1H), 10.79 (s, 1H);
LCMS (System 9): R = 2.75 min; m/z 426 [M+H]+.
Example 167: 2-[5-FIuoro(trif|uoromethy|)pheny|]-N-{5-[(7-methy|-7H-pyrro|o[2,3-d]pyrimidin
y|)carbony|]pyridiny|}acetamide
1-(3-Dimethy|aminopropyl)ethy|carbodiimide HCI (150 uL, 0.5M in DMF) was added to 5-f|uoro
(trif|uoromethy|)pheny|acetic acid (90 umol), N-methylmorpholine (25 uL, 150 umol), 1-
hydroxybenzotriazole (15 umol, 0.05M in DMF) and (5-aminopyridiny|)(7-methy|-7H-pyrro|o[2,3-
d]pyrimidiny|)methanone (Preparation 110, 75 umol, 0.25 M in DMF). The mixture was d at 50°C
for 2 hours and then evaporated in vacuo and purified by prep-HPLC (method 4) to afford the title
compound.
LCMS (system 5): R = 2.66 min; m/z 458 [M+H]+
The following es were prepared according to the method described above for Example 167
starting from (5-aminopyridiny|)(7-methy|-7H-pyrro|o[2,3-d]pyrimidiny|)methanone (Preparation 110)
and the appropriate acid.
168 2-(3-methylphenyl)-N-{5-[(7-methy|-7H- LCMS (system 5): R = 2.53 min;
pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridin m/z 386 [M+H]+
yl}acetamide
(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R 2.61 min;
y| )carbonyl]pyridiny|}[2- m/z 440 [M+H]+
uoromethyl)pheny|]acetamide
2-(3,5-difluorophenyl)-N-{5-[(7-methy|-7H- LCMS (system 5): R
pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridin m/z 408 [M+H]+
yl}acetamide
(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R
y| )carbonyl]pyridinyI}(2,4,6- m/z 426 [M+H]+
trifluoropheny|)acetamide
(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R
y| )carbonyl]pyridinyI}(2,3,6- m/z 426 [M+H]+
trifluoropheny|)acetamide
N-{5-[(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R
y| nyl]pyridiny|}[4- m/z 450 [M+H]+
(methylsulfonyl)phenyl]acetamide
2-[3-f|uoro(trifluoromethyl)phenyI]-N-{5-[(7- LCMS (system 5): R
methyl-7H-pyrro|o[2,3-d]pyrimidin m/z 458 [M+H]+
y|)carbonyl]pyridinyl}acetamide
2-(3-methoxyphenyl)-N-{5-[(7-methy|-7H- LCMS:Rt= 2.34 min; m/z 402
pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridin [|V|+H]+
yl}acetamide
N-{5-[(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R 2.76 min;
y| )carbonyl]pyridiny|}[3- m/z 456 [M+H]+
(trifluoromethoxy)phenyl]acetamide
N-{5-[(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R 2.54 min;
y| )carbonyl]pyridinyI}(2,3,5- m/z 426 [M+H]+
trifluoropheny|)acetamide
N-{5-[(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R = 1.95 min;
y| )carbonyl]pyridiny|}(2-methquuinolin m/z 437 [M+H]+
y| )acetamide Using (2-methquuinoliny|)acetic
acid (Preparation 165).
N-{5-[(7-m ethyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R = 2.37 min;
y| )carbonyl]pyridiny|}pheny|acetamide m/z 372 [M+H]+
2-(2-chIorofluorophenyl)-N-{5-[(7-m ethyl-7H- LCMS m 5): R = 2.51 min;
pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridin m/z 424 [M+H]+
yl}acetamide
2-(4-methoxyphenyl)-N-{5-[(7-m ethyl-7H- LCMS:Rt= 2.22 min; m/z 402
pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridin [|V|+H]+
yl}acetamide
W0 2012/137089 66
N-{5-[(7-methyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5): R
y|)carbonyl]pyridinyI}{4- m/z 472 [M+H]+
[(trifluoromethyl)thio]pheny|}acetamide
2-biphenyIyI-N-{5-[(7-methyl-7H-pyrro|o[2,3- LCMS (system 6):
d]pyrimidinyl)carbonyl]pyridinyl}acetamide m/z 448 [M+H]+
N-{5-[(7-methyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5):
y|)carbonyl]pyridiny|}(2,4,5- m/z 426 [M+H]+
trifluoropheny|)acetamide
N-{5-[(7-methyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5):
y|)carbonyl]pyridinyI}{3- m/z 472 [M+H]+
luoromethyl)thio]pheny|}acetamide
2-(4-methylphenyl)-N-{5-[(7-methy|-7H- LCMS (system 5):
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin m/z 386 [M+H]+
y|}acetamide
N-{5-[(7-methyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (system 5):
y|)carbonyl]pyridiny|}(3,4,5- m/z 426 [M+H]+
trifluoropheny|)acetamide
2-(2,4-dimethylphenyl)-N-{5-[(7-methy|-7H- LCMS (system 5):
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin m/z 400 [M+H]+
y|}acetamide
2-(2,3-dihydro—1-benzofurany|)-N-{5-[(7- LCMS (system 5):
methyl-7H-pyrro|o[2,3-d]pyrimidin m/z 414 [M+H]+
bonyl]pyridinyl}acetamide
-difluorophenyl)-N-{5-[(7-methy|-7H- LCMS (system 5):
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin m/z 408 [M+H]+
y|}acetamide
2-(2,4-difluorophenyl)-N-{5-[(7-methy|-7H- LCMS (system 5):
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin m/z 408 [M+H]+
y|}acetamide
2-[2-fluoro(trifluoromethyl)phenyI]-N-{5-[(7- LCMS (system 5):
methyl-7H-pyrro|o[2,3-d]pyrimidin m/z 458 [M+H]+
y|)carbonyl]pyridinyl}acetamide
-difluorophenyl)-N-{5-[(7-methy|-7H- LCMS (system 5):
pyrro|o[2,3-d]pyrimidinyl)carbonyl]pyridin m/z 408 [M+H]+
y|}acetamide
N-{5-[(7-methyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 2): Rt
y|)carbonyl]pyridinyI}[3- m/z 440 [M+H]+
(trifluoromethyl)phenyl]acetamide
2-(3,4-difluorophenyl)-N-{5-[(7-methy|-7H- LCMS m 2): Rt
pyrro|o[2,3-d]pyrimidinyl)carbonyl]yridine m/z 408 [M+H]+
y|}acetamide
N-{5-[(7-methyl-7H-pyrro|o[2,3-d]pyrimidin LCMS (System 2): Rt
bonyl]yridiney|}[4- m/z 456 [M+H]+
(trifluoromethoxy)phenyl]acetamide
hlorophenyl)-N-{5-[(7-methy|-7H- LCMS m 2): R: 1.5 min; m/z
pyrro|o[2,3-d]pyrimidiny|)carbony|]yridine 406 [M+H]+
yl}acetamide
Example 198: 3-(2-chlorophenyl)-N-{5-[(7-methy|-7H-pyrro|o[2,3-d]pyrimidiny|)carbonyI]pyridin
y|}propanamide
The title compound was prepared according to the method described above for Example 167 starting
from (5-aminopyridiny|)(7-methy|-7H-pyrro|o[2,3-d]pyrimidiny|)methanone (Preparation 110) and 3-
(2-ch|oropheny|)propanoic acid.
LCMS (system 5): R = 2.66 min; m/z 420 [M+H]+
The following Examples were prepared according to Method b (Example 1 using DIPEA) as described
above starting from (5-aminopyridinyl)(7-tert-buty|-7H-pyrro|o[2,3-d]pyrimidiny|)methanone
(Preparation 31) and the appropriate acids.
N-{5-[(7-tert-buty|-7H-pyrro|o[2,3- LCMS (system 9): R = 3.16 min; m/z 445 [M+H]+
d]pyrimidiny|)carbony|]pyridin- using (2-cyclopropyI-1,3-oxazoIy|)acetic acid
3-y|}(2-cyc|opropy|-1 ,3-oxazol- (Preparation 155).
4- | acetamide
N-{5-[(7-tert-buty|-7H-pyrro|o[2,3- LCMS m 9): R = 2.87 min; m/z 492 [M+H]+
d]pyrimidiny|)carbony|]pyridin-
3-y|}[3-
meth Isulfon | phen |]acetamide
N-{5-[(7-tert-buty|-7H-pyrro|o[2,3- LCMS m 9): R = 2.76 min; m/z 432[M+H]+
d]pyrimidiny|)carbony|]pyridin-
2-(1,3-dimethyI-1H-pyrazol-
4- | acetamide
N-{5-[(7-tert-buty|-7H-pyrro|o[2,3- LCMS (system 9): R = 2.73 min; m/z 418[M+H]+
d]pyrimidiny|)carbony|]pyridin-
3-y|}(1-methy|-1H-pyrazoI
y|)acetamide
N-{5-[(7-tert-buty|-7H-pyrro|o[2,3- LCMS (system 9): R = 2.80 min; m/z 458 [M+H]+ using
d]pyrimidiny|)carbony|]pyridin- lopropyImethy|-1H-pyrazoIy|)-acetic acid
3-y|}(3-cyc|opropy|methy|- (Preparation 125)
1H-pyrazoIyl )acetamide
Example 204: {7-[(1S)—2-hydroxymethy|ethy|]-7H-pyrro|o[2,3-d]pyrimidiny|}carbony|)pyridin
y|][3-(trif|uoromethyl)phenyl]acetamide
10% Hydrochloric acid in 1,4- dioxane (0.2 mL) was added to N-(5-{7-[(S)(tert-Buty|-dimethyl-
si|any|oxy)methy|-ethy|]-7H-pyr
ro|o[2,3-d]pyrimidinecarbonyI}-pyridinyl)(3-trif|uoromethyl-phenyl)—acetamide (Preparation 105,
59 mg, 0.098 mmol) in THF (2 mL) and the mixture was stirred at room temperature for 18 hours. The
mixture was evaporated in vacuo and triturated with ether-pentane to afford the title nd as an off
white solid in 86% yield, 41 mg.
LCMS (system 9): Rt = 2.97 min; m/z 484 [M+H]+.
The following es were prepared according to the method described above for Example 204 using
the appropriate preparations as described.
205 N-[5-({7-[(1R)—2-hydroxy—1-methy|ethy|]-7H- LCMS (system 9): Rt =
pyrrolo[2,3-d]pyrim -y|}carbony|)pyrid iny|] 2.89 min; m/z 484 [M+H]+
[3-(trifluoromethy|)pheny|]acetam ide Using (Preparation 142).
206 N-[5-({7-[(1R)—2-hydroxy—1-methy|ethy|]-7H- LCMS (system 9): Rt =
pyrrolo[2,3-d]pyrim idiny|}carbony|)pyrid iny|] 2.52 min; m/z 449 [M+H]+
(4-isopropyI-1H-1,2,3-triazoly|)acetamide Using (Preparation 144).
207 N-[5-({7-[(1R)—2-hydroxy—1-methy|ethy|]-7H- LCMS (system 9): Rt =
pyrrolo[2,3-d]pyrim idiny|}carbony|)pyrid iny|] 2.87 min; m/z 516 [M+H]+
[1 -isopropyI(trifluoromethyl)-1 H-pyrazoI Using (Preparation 150).
y|]acetamide
208 2-(4-cyanophenyl )-N-[5-({7-[(1 R)—2-hyd roxy-1 - LCMS (system 9): Rt =
ethy|]-7H-pyrro|o[2,3-d]pyrimidin 2.68 min; m/z 441 [M+H]+
y|}carbony|)pyridinyl]acetamide Using (Preparation 151).
209 N-[5-({7-[(1R)—2-hydroxy—1-methy|ethy|]-7H- LCMS (system 9): Rt =
pyrrolo[2,3-d]pyrim idiny|}carbony|)pyrid iny|] 2.57 min; m/z 494 [M+H]+
[3-(methylsulfonyl)phenyl]acetamide Using (Preparation 152).
N-[5-({7-[(1R)—2-hydroxy—1-methy|ethy|]-7H- LCMS (system 9): Rt =
o[2,3-d]pyrim idiny|}carbony|)pyrid |] 1.96 min; m/z 467 [M+H]+
quinolinylacetamide Using (Preparation 153).
W0 2012/137089 69
The following Examples were prepared according to the method described above for Example 34 at
50°C, starting from [2-amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl](5-aminopyridin-
3-yl)methanone (enantiomer 1, Preparation 57) and the appropriate acids.
N-(5-{[2-amino(2-hydroxymethylethyl)- LCMS (system 10): Rt = 2.48
7H-pyrrolo[2,3-d]pyrimidin . +
min, m/z 482 [M+H]_
yl]carbonyl}pyridinyl)—2-quinolin
ylacetamide
212 N-(5-{[2-amino(2-hydroxymethylethyl)- LCMS m 10): Rt = 2.57
7H-pyrrolo[2,3-d]pyrimidin . +
mm’ ””2 456 [Mm]_
yl]carbonyl}pyridinyl)—2-(4-
cyanophenyl)acetamide
N-(5-{[2-amino(2-hydroxymethylethyl)- LCMS (system 10): Rt = 2.87
7H-pyrrolo[2,3-d]pyrimidin
min; m/z 499 [M+H]+
yl]carbonyl}pyridinyl)—2-[3-
(trifluoromethyl)phenyl]acetamide
The following Examples were prepared according to the method described above for Example 34 at
50°C, starting from [2-amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl](5-aminopyridin-
ethanone (enantiomer 2, Preparation 59) and the appropriate acids.
N-(5-{[2-amino(2-hydroxymethylethyl)-7H- _ _
pyrrolo[2,3-d]pyrimidinyl]carb0ny|}Pyridiny|)(4- LCMmSin(-sry1s/tzta;n5g)th/IIR:I:I]+2.26
cyanophenyl)acetamide ’
[2-amino(2-hydroxymethylethyl)-7H-
LCMS (system 9): Rt = 2.72
pyrrolo[2,3-d]pyrim -yl]carbonyl}pyridinyl)—2-[3-
min; m/z 499 [M+H]+
(trifluoromethyl)phenyl]acetamide
N-(5-{[2-amino(2-hydroxymethylethyl)-7H-
LCMS (system 9): Rt = 1.62
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridinyl)—2-
min; m/z 482 [M+H]+
quinolinylacetamide
The following es were prepared according to the method described above for Example 34 at 50°C
ng from (5-aminopyridinyl)(7-oxetan-3—yl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation
100) and the appropriate acid.
2-(3,4-dichlorophenyl)-N-{5-[(7-oxetanyl-7H-pyrrolo[2,3- LCMS (system 3): R = 3.04
d]pyrimidinyl)carbonyl]pyridinyl}acetam ide
min; m/z 482 [M+H]+
2-(4-cyanophenyl)-N-{5-[(7-oxetanyl-7H-pyrrolo[2,3- LCMS (System 2): Rt = 1.3
d]pyrImIdInyl)carbonyl]pyrIdInyl}acetamIde
min; m/z 439 [M+H]+
2-(4-chlorophenyl)-N-{5-[(7-oxetanyl-7H-pyrrolo[2,3- LCMS m 2): Rt = 1.5
d]pyrImIdInyl)carbonyl]pyrIdInyl}acetamIde
min; m/z 448 [M+H]+
2-(3-chlorophenyl)-N-{5-[(7-oxetanyl-7H-pyrrolo[2,3- LCMS (System 2): Rt = 1.5
d]pyrimidinyl)carbonyl]pyridinyl}acetam ide
min; m/z 448 [M+H]+
yanophenyl)-N-{5-[(7-oxetanyl-7H-pyrrolo[2,3- LCMS (System 2): Rt = 1.3
d]pyrImIdInyl)carbonyl]pyrIdInyl}acetamIde
min; m/z 439 [M+H]+
N-{5-[(7-oxetanyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (System 2): Rt = 1.0
yl)carbonyl]pyrIdInyl}qumolInylacetamIde
min; m/z 465 [M+H]+
2-(6-chloroimidazo[1,2-a]pyridinyl)-N-{5-[(7-oxetanyl- LCMS m 2): Rt = 1.0
rolo[2,3-d]pyrimidinyl)carbonyl]pyridin _ +
ml“, m/z 488 [M+H]_
y|}acetamide
N-{5-[(7-oxetanyl-7H-pyrrolo[2,3-d]pyrimidin LCMS (System 2): Rt = 1.3
§I;acetamflgyl carbon l ridiny}l 1-oxo-1,3-dih( Vdro-2H-isoindol . +
min; m/z 469 [M+H]
The following Examples were prepared according to the method described above for Example 204, using
the preparations as described.
225 2-(4-cyanophenyl)-N-(5-{[7-(2-hydroxy-1,1- LCMS (System 9): R = 2.74 min; m/z
dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin 455.4 [M+H]+
yl]carbonyl}pyridinyl)acetamide Using Preparation 131
226 N-(5-{[7-(2-hydroxy-1,1-dimethylethyl)-7H- LCMS (System 9): R = 2.99 min; m/z 498
o[2,3-d]pyrimidinyl]carbonyl}pyridin [M+H]+
yl)[3-(trifluoromethyl)phenyl]acetamide Using Preparation 136
227 N-(5-{[7-(2-hydroxy-1,1-dimethylethy|)-7H- H NMR (400 MHz, DMSO) 6: 1.22 (d,
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin 6H), 1.71 (s, 6H), 2.96 (m, 2H), 3.94 (s,
y|)(4-isopropyl-1H-1,2,3-triazol 2H), 5.35 (s, 2H), 7.87 (s, 1H), 8.20 (s,
yl)acetamide 1H), 8.44 (s, 1H), 8.75 (s, 1H), 8.98 (m,
2H), 9.46 (s, 1H), 11 (s,1H)
Using Preparation 144
228 N-(5-{[7-(2-hydroxy-1,1-dimethylethyl)-7H- LCMS (System 9): R = 2.61 min; m/z 481
pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin [M+H]+
yl)quinolinylacetamide Using Preparation 140
Example 229: racemic 2-{5-[(5-{[(4-Ch|oropheny|)acety|]amino}pyridiny|)carbonyI]-7H-pyrro|o[2,3-
d]pyrimidinyl}propanamide
LN/ “Hz/Q”.
O¥Me
2-bromopropionamide (46.5 mg, 0.31 mmol) was added to a e of 2-(4-ch|oropheny|)-N-[5-(7H-
pyrro|o[2,3-d]pyrimidiny|carbony|)pyridiny|]acetamide (Example 308, 100 mg, 0.26 mmol) and
cesium carbonate (150 mg, 0.46 mmol) in DMF (1.3 mL). The mixture was d at 60°C for 3 hours. The
reaction e was cooled and water (10 mL) was added. The mixture was extracted with EtOAc (3 x 10
mL) and the combined c phases were passed through a phase separator and evaporated in vacuo.
Purification by preparative HPLC gave the title compound as a white solid in 34% yield, 15 mg.
LCMS (System 3): R = 2.13 min; m/z 463 [M+H]+.
Example 230: c 2-{5-[(5-{[(4-Ch|oropheny|)acety|]amino}pyridiny|)carbonyI]-7H-pyrro|o[2,3-
d]pyrimidiny|}-N,N-dimethylpropanamide
The title compound was prepared according to the method described for Example 34 at 50°C using
racemic 2-{5-[(5-{[(4-ch|oropheny|)acety|]amino}pyridiny|)carbonyI]-7H-pyrro|o[2,3-d]pyrimidin
y|}propanoic acid (Example 354) and dimethylamine. Purification was accomplished by preparative
HPLC (method 1) to afford the title compound.
LCMS (system 3): R = 2.39 min; m/z 491 [M+H]+.
Example 231: racemic 2-{5-[(5-{[(4-Ch|oropheny|)acety|]amino}pyridiny|)carbonyI]-7H-pyrro|o[2,3-
d]pyrimidiny|}-N-methy|propanamide
The title compound was prepared according to the method described for Example 34 at 50°C using 2-{5-
[(5-{[(4-ch|oropheny|)acety|]amino}pyridiny|)carbonyI]-7H-pyrro|o[2,3-d]pyrimidiny|}propanoic acid
(Example 354) and methylamine. Purification was accomplished by preparative HPLC (method 1) to
afford the title compound.
LCMS (system 3): R = 2.31 min; m/z 477 [M+H]+.
e 232: 2-(4-Cyanophenyl)-N-[5-({7-[2-hydroxy(hydroxymethy|)methylethyI]-7H-pyrrolo[2,3-
d]pyrimidiny|}carbony|)pyridiny|]acetamide
\ 0W“
Nk/l \ H
\N N
4-Cyanophenylacetic acid (19 mg, 0.10 mmol) was added to nopyridinyl){7-[2-{[tert—
butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)methylethyl]-7H-pyrrolo[2,3-d]pyrimidin-
-yl}methanone (Preparation 116, 54 mg, 0.97 mmol), 1-propylphosphonic acid cyclic ide (0.17
mL, 0.291 mmol) and triethylamine (0.047 mL, 0.291 mmol) in tetrahydrofuran (3 mL). The mixture was
stirred at 50°C for 16 hours. Saturated aqueous sodium bicarbonate (5 mL) was added then extracted
with dichloromethane (3 x 7 mL). The combined organic phases were dried over magnesium sulphate and
ated in vacuo.
The residue was dissolved in tetrahydrofuran (3 mL) and a utylammonium fluoride on in THF (1
mL of a1M solution, 1 mmol) was added and the solution was stirred for 1 hour. Saturated aqueous
sodium bicarbonate (5 mL) was added then extracted with dichloromethane (3 x 5 mL). The combined
organic phases were dried over ium sulphate, evaporated in vacuo and purified by preparative
HPLC (method 2).
LCMS m 4): R = 2.52 min; m/z 471 [M+H]+
The following Examples were prepared according to the method described above for Example 232
starting from (5-aminopyridinyl){7-[2-{[tert—butyl(dimethyl)silyl]oxy}({[tert-
butyl(dimethyl)silyl]oxy}methyl)—1-methylethyl]—7H-pyrrolo[2,3-d]pyrimidinyl}methanone (Preparation
116) and the appropriate acids.
233 N-[5-({7-[2-hydroxy(hydroxymethyl) LCMS (system 4) Rt = 2.52 min; m/z
methylethyl]—7H-pyrrolo[2,3-d]pyrimidin 497 [M+H]+
yl}carbonyl)pyridinyl]quinolinylacetamide Prep method 1
234 N-[5-({7-[2-hydroxy(hydroxymethyl) LCMS m 4) Rt = 2.69 min; m/z
methylethyl]—7H-pyrrolo[2,3-d]pyrimidin 518 [M+H]+
yl}carbonyl)pyridinyl][5-methyl Prep method 1
(trifluoromethyl)—1H-pyrazolyl]acetamide
235 2-(4-chlorophenyl)-N-[5-({7-[2-hydroxy LCMS (system 4) Rt = 2.83 min; m/z
(hydroxymethyl)—1-methylethyl]—7H-pyrrolo[2,3- 480 [M+H]+
d]pyrimidinyl}carbonyl)pyridinyl]acetamide Prep method 1
236 N-[5-({7-[2-hydroxy(hydroxymethyl) LCMS (system 4) Rt = 2.81 min; m/z
methylethyl]—7H-pyrrolo[2,3-d]pyrimidin 514 [M+H]+
yl}carbonyl)pyridinyl][4- Prep method 1
(trifluoromethyl)phenyl]acetamide
237 2-(5-chloropyridinyl)-N-[5-({7-[2-hydroxy LCMS (system 3) Rt = 2.37 min; m/z
h drox meth lmeth leth l]-7H-p rrolo[2,3- 481 [M+H]+
W0 2012/137089 73
d]pyrimidinyl}carbonyl)pyridinyl]acetamide Prep method 1
Using (5-chloropyridinyl)acetic
acid (Preparation 90).
N-[5-({7-[2-hydroxy(hydroxymethyl) LCMS m 4) Rt = 2.84 min; m/z
methylethyl]-7H-pyrrolo[2,3-d]pyrim idin 514 [M+H]+
bonyl)pyrid inyl]—2-[3- Prep method 1
(trifluoromethyl)phenyl]acetamide
N-[5-({7-[2-hydroxy(hydroxymethyl) LCMS (system 3) Rt = 2.59 min; m/z
methylethyl]-7H-pyrrolo[2,3-d]pyrim idin 504 [M+H]+
yl}carbonyl)pyrid inyl]—2-[3-(trifl uorom ethy|)-1 H- Prep method 2
pyrazolyl]acetamide
The following Examples were prepared ing to the method bed above for Example 1 using
DIPEA and purification by preparative HPLC, starting from (2-aminoisopropyl-7H-pyrrolo[2,3-
d]pyrimidinyl)(5-aminopyridinyl)methanone (Preparation 122) and the appropriate acids.
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R = 2.57 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}[4- 474 [M+H]+
(trifluoromethyl)—1H-1,2,3-triazolyl]acetamide Using 4-(trifluoromethyl)-1H-1,2,3-
triazolyl]acetic acid (Preparation
81 .
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R = 2.56 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}(4- 440 [M+H]+
cyanophenyl)acetamide
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R: 2.46 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}(5- 450 [M+H]+
chloropyridinyl)acetamide Using -(5-ch|oropyridinyl)acetic acid
Preparation 90 .
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R = 2.20 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}(5- 434 [M+H]+
fluoropyrid inyl )acetamide Using (5-fluoropyridinyl)acetic acid
(Preparation 92).
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 10): R = 3.22 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}[4- 483 [M+H]+
(trifluoromethyl)phenyl]acetamide
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 10): R = 3.22 min; m/z
midinyl)carbonyl]pyridinyl}[3- 483 [M+H]+
(trifluoromethyl)phenyl]acetamide
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R = 2.22 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}(1,3- 456 [M+H]+
benzoxazolyl)acetamide
(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R = 2.15 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}[3- 493 [M+H]+
lsulfonyl)phenyl]acetamide
N-{5-[(2-aminoisopropyl-7H-pyrrolo[2,3- LCMS (System 9): R = 2.16 min; m/z
d]pyrimidinyl)carbonyl]pyridinyl}(4- 448 [M+H]+
isopropyl-1H-1,2,3-triazolyl)acetamide
N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3- LCMS m 9): R = 2.01 min; m/z
d]pyrimidiny|)carbony|]pyridinyI}(4- 446 [M+H]+
ropyI-1H-1,2,3-triazo|y|)acetamide Using Preparation 83
N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3- LCMS (System 10): R = 3.03 min; m/z
d]pyrimidiny|)carbony|]pyridinyI}(4- 449 [|\/|+|—|]+
ch|oropheny|)acetamide
N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3- LCMS (System 10): R = 3.03 min; m/z
d]pyrimidiny|)carbonyl]pyridinyI}[1- 515 [M+H]+
isopropyI(trifluoromethy|)-1H-pyrazoI Using Prep 185
y|]acetamide
N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3- LCMS (System 10): R = 2.92 min; m/z
d]pyrimidiny|)carbonyl]pyridinyI}[3- 473 [M+H]+
(trifluoromethy|)-1H-pyrazoIyl]acetamide
N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3- LCMS (System 9) R 2.46 min; m/z 445
midiny|)carbony|]pyridinyI}(3- [m+H]+
cyclopropyI-1H-pyrazoIy|)acetamide
N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3- LCMS (System 9): R = 2.77 min; m/z
d]pyrimidiny|)carbonyl]pyridinyI}[1- 515 [|\/|+|—|]+
isopropyI(trifluoromethy|)-1H-pyrazoI
y|]acetamide
Example 255: N-{5-[(2-aminoisopropyI-7H-pyrro|o[2,3-d]pyrimidiny|)carbonyl]pyridinyI}(5-
cyanopyridiny|)acetam ide
I \ H \N /
H2N)\\N N
)‘Me
The title nd was prepared according to the method described for Example 166 using N-{5-[(2-
aminoisopropyI-7H-pyrro|o[2,3-d]pyrimidiny|)carbonyl]pyridiny|}(5-bromopyridiny|)acetamide
(Example 318) to afford the title compound in 45% yield, 30 mg. 1H NMR (400 MHz, DMSO-d6) 6: 1.45
(d, 6H), 4.06 (s, 2H), 4.84 (m, 1H), 6.62 (s, 2H), 7.66 (d, 1H), 7.97 (s, 1H), 8.29 (dd, 1H), 8.37 (t, 1H), 8.66
(d, 1H), 8.92 (s, 1H), 8.95-8.98 (m, 2H), 10.75 (s, 1H);LCMS (System 10): R: 2.49 min; m/z 441 [M+H]+.
The following Examples were prepared according to the method described above for Example 34 at 50°C
starting from [2-amino(2-hydroxy-1,1-dimethy|ethy|)-7H-pyrro|o[2,3-d]pyrimidiny|](5-aminopyridin
y|)methanone (Preparation 48) and the riate acids.
Data
[2-amino(2-hydroxy-1,1-dimethy|ethy|)-7H- LCMS (System 10): R = 2.91
pyrrolo[2,3-d]pyrimidiny|]carbony|}pyridiny|)[3- min; m/z 513 [M+H]+
trifluorometh Iphen |]acetamide
N- 5- [2-amino 2-h drox -1,1-dimeth |eth |-7H- LCMS S stem 10 : R = 2.48
pyrro|o[2,3-d]pyrimidiny|]carbony|}pyridiny|)(1,3- min; m/z 486 [M+H]
benzoxazoI | acetamide
N-(5-{[2-amino(2-hydroxy-1,1-dimethy|ethy|)-7H- LCMS (System 10). =2.50
pyrrolo[2,3-d]pyrimidiny|]carbony|}pyridiny|)[3- min; m/z 523 [M+H])
meth Isulfon | phen |]acetamide
N-(5-{[2-amino(2-hydroxy-1,1-dimethy|ethy|)-7H- LCMS m 10): R = 2.62
o[2,3-d]pyrimidiny|]carbony|}pyridiny|)(4- min; m/z 470 [M+H]+
c n | acetamide
N-(5-{[2-amino(2-hydroxy-1,1-dimethy|ethy|)-7H- LCMS (System 10): R = 2.89
pyrrolo[2,3-d]pyrimidiny|]carbony|}pyridiny|)[1- min; m/z 545 [M+H]+
isoprop | trifluorometh | -1H-p razoI |]acetamide Usino Prep 185
Example 261: 4-cyano(trifluoromethyl)phenyl)-N-(5-(7-(1,3-dihydroxymethylpropanyl)—7H-
pyrro|o[2,3-d]pyrimidinecarbonyl)pyridiny|)acetamide
\ / o
le \ N
\ H
N CF3
2-(4-cyano(trifluoromethyl)phenyl)acetic acid (Preparation 164, 29.8 mg, 0.13 mmol) was added to a
stirred on of (5-aminopyridiny|){7-[2-{[tert-buty|(dimethy|)si|y|]oxy}({[tert-
dimethy|)si|y|]oxy}methy|)methy|ethy|]-7H-pyrro|o[2,3-d]pyrimidiney|)methanone (Preparation
116, 55.6 mg, 0.10 mmol) and 2—(7—aza—‘Ei-f-benzotriazoi-i—yi)—1,1,3,3—tetramethyiurcnium
hexaftuorophcsphate (57 mg, 0.15 mmol) in pyridine (0.5 mL). The mixture was stirred at 50°C for 16
hours, then cooled to room temperature and partitioned between saturated aqueous sodium bicarbonate
(5 mL) and DCM (5 mL). The organic phase was separated and concentrated in vacuo to provide a
residue that was dissolved in 3 mL of THF and then treated with 3 mL of a 1N s solution of HCI.
The mixture was stirred rapidly for 4 hours at room temperature and then basified by the addition of 4 mL
of a 1N aqueous NaOH solution. The mixture was extracted with three te 5 mL portions of a 95/5
DCM/MeOH mixture. The combined organic extracts were dried by passage through a phase separator
and then concentrated in vacuo to obtain a crude residue. This residue was dissolved in 1 mL DMSO
and purified by preparative HPLC to afford the title compound as an off white solid in 56% yield, 30 mg. 1H
NMR (400 MHz, DMSO) 6: 1.68 (s, 3H), 3.83 (m, 2H), 4.01 (s, 2H), 4.19 (m, 2H), 5.00 (m, 2H), 7.82 (m,
H), 8.01 (d, 1H), 8.18 (m, 1H), 8.21 (s, 1H), 8.66 (s, 1H), 8.97 (m, 2H), 9.40 (s, 1H), 10.78 (s, 1H).
LCMS m 2): R = 0.90 min; m/z 539 [M+H]+.
The following Examples were ed according to the method described above for Example 261,
starting from ((5-aminopyridiny|){7-[2-{[tert-buty|(dimethy|)si|y|]oxy}({[tert-
buty|(dimethy|)si|y|]oxy}methy|)methy|ethy|]-7H-pyrro|o[2,3-d]pyrimidiney|)methanone (Preparation
116) and the appropriate acids followed by si|y| group deprotection.
N-(5-(7-(1,3-dihydroxymethylpropanyl)—7H— LCMS (system 2): R = 0.96
pyrro|o[2,3-d]pyrimidinecarbony|)pyridiny|)(3- min; m/z 516 [M+H]+
oromethoxy)pheny|)acetamide
N-(5-(7-(1,3-dihydroxymethylpropanyl)—7H— LCMS (system 2): R = 1.05
pyrro|o[2,3-d]pyrimidinecarbony|)pyridiny|)(4- min; m/z 532 [M+H]+
f|uoro(trif|uoromethy|)pheny|)acetamide
N-(5-(7-(1,3-dihydroxymethylpropanyl)—7H— LCMS (system 2): R = 1.04
pyrro|o[2,3-d]pyrimidinecarbonyl)pyridiny|) min; m/z 500 [M+H]+
(trifluoromethyl)benzamide
7-(1,3-dihydroxymethylpropanyl)—7H— LCMS (system 2): R = 1.09
pyrro|o[2,3-d]pyrimidinecarbonyl)pyridiny|)f|uoro- min; m/z 532 [M+H]+
4-(trif|uoromethy|)benzamide
Example 266: 2-[3-(azetidiny|methy|)pheny|]-N-{5-[(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidin
y|)carbony|]pyridiny|}acetamide formate salt.
N\ 0 .HCOOH
<\ H
/ \
N N
NI |
/ W;
Me"<
ine hydrochloride (10 mg, 0.11 mmol) was added to a stirred solution of 2-(3-formylpheny|)-N-{5-
[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridiny|}acetamide (Preparation 192, 23 mg,
0.05 mmol) in DCM (1 mL) and acetic acid (0.04 mL, 0.60 mmol). The reaction mixture was d for 30
min at room temperature before addition of sodium triacetoxyborohydride (29 mg, 0.14 mmol) and stirring
continued for 3 hours. Water (1 mL) was added and the mixture concentrated in vacuo (with toluene
azeotroping). The residue was purified by preparative HPLC to afford the title compound as a formate salt
in 12% yield, 3.4 mg.
LCMS: R = 2.95 min; m/z 469 [M+H]+.
Example 267: 2-{3-[(3-f|uoroazetidiny|)methyl]pheny|}-N-{5-[(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidin
y|)carbony|]pyridiny|}acetamide
The title compound was prepared according to the method described for Example 266 using 2-(3-
phenyl)-N-{5-[(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridiny|}acetamide
(Preparation 192, 60 mg, 0.14 mmol) and 3-fluoroazetidine to afford the title compound in 16% yield, 11
LCMS: R = 2.75 min; m/z 487 [M+H]+.
Example 268: N-{5-[(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)carbonyl]pyridiny|}[3-(morpho|in
y|methy|)pheny|]acetamide formate salt
The title compound was prepared ing to the method bed for Example 266 using 2-(3-
formy|phenyl)-N-{5-[(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)carbony|]pyridiny|}acetamide
(Preparation 192, 60 mg, 0.14 mmol) and morpholine to afford the title compound as a formate salt in
% yield, 21 mg.
LCMS: R = 2.70 min; m/z 499 [M+H]+
es 269-283 General method:
The mixture of (5-aminopyridinyl)(7-(2,2,3,3,9,9,10,10-octamethyl-4,8-dioxa-3,9-disilaundecanyl)—7H-
pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 196, 54.2 mg, 0.10 mmol), HATU (57 mg, 0.15
mmol) and the requisite carboxylic acid (0.13 mmol) in pyridine (0.5 mL) were stirred at 50 °C for 16
hours. After cooling to room temperature, saturated sodium bicarbonate solution (5 mL) was added and
the mixture was extracted with DCM (5 mL). The organic layer was passed through a phase separator
and concentrated in vacuo. The ing residue was dissolved in THF (2 mL) and 1 N aqueous HCI (2
mL) added to the solution. The reaction mixture was stirred for 2 hours at room temperature and
quenched with 1 N aqueous NaOH (3 mL). The mixture was then treated according to one of the following
methods:
Method A
The e was extracted with a mixture of DCM/MeOH (95/5, 5 mL x 3), the ed organic layers
passed through a phase tor and concentrated in vacuo to obtain a crude residue. This residue was
dissolved in DMSO (1 mL) and ed via HPLC to yield the desired compound.
Method B
The mixture was suspended in DCM/MeOH (95/5, 5 mL), the solid ed and washed with water (5 mL)
and DCM (5 mL) and dried in vacuo to yield the desired compound.
Method C
The mixture was extracted with a mixture of EtOAc/MeOH (95:5, 5 mL x 3), the combined organic layers
passed through a phase separator and concentrated in vacuo to obtain a crude residue. This residue was
dissolved in DMSO (1 mL) and purified via preparative HPLC to yield the desired compound.
N-(5-(7-(1,3-dihydroxypropanyl)-7H-pyrrolo[2,3-d]pyrimidine LCMS: R = 2.69
carbonyl)pyridinyl)(trif|uoromethyl)benzamide min; m/z 486 [M+H]+
2-(4-chlorophenyl)-N-(5-(7-(1,3-dihydroxypropanyl)—7H- LCMS: R = 2.63
pyrrolo[2,3-d]pyrimidinecarbonyl)pyridinyl)acetamide min; m/z 466 [M+H]+
N- 5- 7- 1,3-dih drox propan l-7H-p 2,3-d]p rimidine LCMS: R: 2.68
carbonyl)pyrid inyl)(trif|uoromethoxy)benzam ide min; m/z 502 [M+H]+
N-(5-(7-(1,3-dihydroxypropan-Z-yl)-7H-pyrro|o[2,3-d]pyrimidine—5- LCMS: R = 1.27 min;
carbonyl)pyrid inyl)—2-(quino| l)acetamide m/z 483 [M+H]+
N-(5-(7-(1,3-dihydroxypropan-Z-yl)-7H-pyrro|o[2,3-d]pyrimidine—5- LCMSI Rt = 2.69
carbonyl)pyrid inyl)(4-(trif|uoromethyl)phenyl)acetamide min; m/z 500 [M+H]+
N-(5-(7-(1,3-dihydroxypropan-Z-yl)-7H-pyrro|o[2,3-d]pyrimidine—5- m/Z 500 [M+H]+
carbon | o ridin I 3- trifluorometh | ohen | acetamide
N-[5-({7-[2-hydroxy—1-(hydroxymethyl)ethyl]—7H-pyrro|o[2,3- m/z 497 [M+H]+
d]pyrimidinyl}carbonyl)pyridinyl]—2-(2-methy|quinolin Preparation 165
yl)acetamide
2-(3,4-dich|oropheny|)-N-[5-({7-[2-hydroxy—1-(hydroxymethy|)ethyl]- m/Z 500 [M+H]+
7H-pyrro|o[2,3-d]pyrimidinyl}carbonyl)pyridinyl]acetamide
2-[3-fluoro(trifluoromethyl)pheny|]-N-[5-({7-[2-hydroxy m/z 518 [M+H]+
xymethyl)ethyl]—7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyrid inyl]acetam ide
2-(2,4-dichIorophenyl)-N-[5-({7-[2-hydroxy—1-(hydroxymethy|)ethyl]- m/Z 500 [M+H]+
7H-pyrro|o[2,3-d]pyrimidinyl}carbonyl)pyridinyl]acetam ide
N-[5-({7-[2-hydroxy—1-(hydroxymethyl)ethyl]—7H-pyrro|o[2,3- m/z 516 [M+H]+
d]pyrimidinyl}carbonyl )pyridinyl]—2-[3-
(trifluoromethoxy)phenyl]acetamide
N-[5-({7-[2-hydroxy—1-(hydroxymethyl)ethyl]—7H-pyrro|o[2,3- m/z 516 [M+H]+
d]pyrimidinyl}carbonyl inyl]—2-[4-
(trifluoromethoxy)phenyl]acetamide
ro—N-[5-({7-[2-hyd roxy-1 -(hyd roxym ethyl )ethy|]-7 H-pyrro|o[2,3- m/z 504 [M+H]+
d]pyrimidinyl}carbonyl)pyridiny|](trif|uoromethyl)benzamide
2-(5-chloropyridinyl)-N-[5-({7-[2-hydroxy—1-(hydroxymethy|)ethy|]— LCMS: Rt = 0.90 min,
7H-pyrro|o[2,3-d]pyrimidinyl}carbonyl)pyridinyl]acetam ide m/z 467 [M+H]+
N-[5-({7-[2-hydroxy—1-(hydroxymethyl)ethyl]—7H-pyrro|o[2,3- LCMSI Rt = 1.81
d]pyrimidinyl}carbonyl)pyridinyl]—2-[3-(trif|uoromethyl)-1H- min, m/z 490 [M+H]+
pyrazoIyl]acetamide
Example 284: 4-benzoyl-N-alpha-(tert—butoxycarbonyl)-N-{5-[(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidin
yl)carbonyl]pyridinyI}-L-phenylalaninamide
k| /
N N 0
)‘Me HNTo Me
O EMe
(5-aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (20 mg, 0.07 mmol) (see
Preparation 95) was dissolved in pyridine (1mL) and 4-benzoyl-N-(tert-butoxycarbonyl)-D-phenylalanine
was added (26 mg, 0.07 mmol) followed by HATU (27 mg, 0.07 mmol). The mixture was stirred at 50 0C
for 5 hours and then cooled, evaporated in vacuo and the crude material was ed by column
tography on silica gel ent of DCM:Methanol 100:0 to 95:5) to afford the title compound as a
yellow solid in 52% yield, 23 mg.
Example 285: 4-benzoyl-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}-D-
phenylalaninamide
o \/O
N\\ NH
kI /
N N 0
)‘Me NH2
4-benzoyl-Nal pha-(tert-butoxycarbonyl)-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin-
3-yl}-D-phenylalaninamide (Example 284, 20 mg, 0.03 mmol) was stirred with 10% hydrochloric acid in
oxane (5 mL) at room temperature for 1 hour. The mixture was evaporated in vacuo and purified by
preparative HPLC to afford the title compound as a gum in 56% yield, 9 mg.
LCMS (system 5): R = 2.89 min; m/z 533 [M+H]+.
Example 286 :4-benzoyl-N-alpha—(tert-butoxycarbonyl)-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin
yl)carbonyl]pyridinyl}-L-phenylalaninamide
The title compound was prepared according to Example 284 using nopyridinyl)(7-isopropyl-7H-
pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and 4-benzoyl-N-(tert-butoxycarbonyl)-L-
phenylalanine
Example 287: 4-benzoyl-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}-L-
phenylalaninamide
W0 2012/137089 80
The title compound was prepared according to the method described for Example 285 using 4-benzoyl-
Nalpha-(tert-butoxycarbonyl)-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}-L-
phenylalaninamide le 286) to afford the title compound in 36% yield, 5 mg.
LCMS (system 4): R = 2.30 min; m/z 533 [M+H]+.
Example 288: 2-(3-benzoylphenyl)-N-(5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)pyridin
yl)acetam ide
The title compound was prepared according to the method described for Example 34 ng from (5-
aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone ration 95) and 2-(3-
benzoylphenyl)acetic acid to afford the title compound in 86% yield, 31 mg.
LCMS (system 3): R = 3.05 min; m/z 504 [M+H]+.
Example 289 enzoylphenyl)-N-(5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)pyridin
yl)acetamide
The title compound was prepared according to the method described for Example 34 starting from (5-
aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and 2-(4-
benzoylphenyl)acetic acid (JOC, 1961, 1635) to afford the title nd in 94% yield, 34 mg. LCMS
(system 3): R = 3.12 min; m/z 504 [M+H]+.
Example 290: N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}[3-
(trifluoromethyl)-3H-diazirenyl]benzam ide
\/ FF
N \ \ I2
kN/| 0
Me)\Me
The title compound was prepared according to the method described for Example 34 ng from (5-
aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and 3-[3-
(trifluoromethyl)-3H-diazirenyl]benzoic acid (Preparation 197) to afford the title compound as a white
solid in 36% yield, 23 mg.
1H NMR (400 MHz, d4-MeOH) 6:1.65 (s, 6H), 5.20 (m, 1H), 7.52-7.61 (m, 4H), 7.79 (s, 1H), 8.07 (m, 1H),
8.29 (s, 1H), 8.94 (s, 1H), 8.81 (m, 1H), 9.55 (s, 1H).
Example 291: N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}[3-
(trifluoromethyl)-3H-diazirenyl]benzam ide
W0 2012/137089 81
The title compound was prepared according to the method described for Example 34 starting from (5-
yridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and 4-[3-
(trifluoromethyl)-3H-diazirenyl]benzoic acid to afford the title compound as a white solid in 4% yield, 2
mg. LCMS (system 3): R = 3.64 min; m/z 494 [M+H]+.
Example 292: N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}
(trifluoroacetyl)benzam ide
N\\ M
K/NN b/Z;
Me}Me
The title compound was prepared according to the method described for Example 34 ng from (5-
aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and 3-
(trifluoroacetyl)benzoic acid (Preparation 200) to afford the title compound as a white solid in 80% yield,
41 mg. LCMS (system 3): R = 2.52 min; m/z 482 [M+H]+.
Example 293: 1-(4-chlorophenyl){5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin
yl}urea
\ / 0
KN | \ H H
1-ch|oroisocyanatobenzene (24 mg, 0.15 mmol) was added to (5-aminopyridinyl)(7-methyl-7H-
pyrrolo[2,3-d]pyrimidinyl)methanone (30 mg, 0.12 mmol) (Preparation 232) in pyridine (1.0 mL). The
mixture was stirred at room temperature overnight then evaporated in vacuo to yield the product as a
crude residue. The crude residue was purified by ative HPLC to afford the title nd in 49%
yield, 24 mg.
LCMS: R = 1.58 min; m/z 407 [M+H]+
The following Examples were prepared according to the method bed above for Example 293,
starting from (5-aminopyridinyl)(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 232)
and the appropriate isocyanates.
1-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 1.64 min; m/z 441
yl)carbonyl]pyridinyl}[4-(trifluoromethyl)phenyl]urea [M+H]+
1-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: Rt = 1.64 min; m/z 441
l carbon l]p ridin - trifluorometh l phen l]urea
W0 37089 82
(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: Rt = 1.67 min; m/z 457
yl)carbonyl]pyridiny|}[4-(trif|uoromethoxy)phenyl]urea [M+H]+
Example 297: 2-(4-chlorophenyl)-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}-
ylacetamide
\ / 0
/ NW
L | \ M;
>‘Me
(4-chlorophenyl)acetic acid (22 mg, 0.13 mmol) was added to a stirring mixture of (7-isopropyl-7H-
pyrrolo[2,3-d]pyrimidinyl)[5-(methylamino)pyridinyl]methanone (30 mg, 0.10 mmol) (Preparation
149) andN-[(dimethylamino)(3H-[1,2,3]triazolo[4,5-b]pyridinyloxy)methylene]-N-methylmethanaminium
hexafluorophosphate (57 mg, 0.15 mmol) in pyridine (1.0 mL). The mixture was heated to 50 °C for 16
hours, evaporated in vacuo and partitioned n saturated aqueous sodium bicarbonate (10 mL) and
EtOAc (10 mL). The aqueous layer was washed with further EtOAc (2 x 10 mL). The combined organics
were dried over NaZSO4, filtered and concentrated in vacuo and purified by preparative HPLC to afford the
title compound 43% yield, 19 mg,
1H NMR (400 MHz, DMSO) 6: 1.52 (d, 6H), 3.40 (br. s, 3H), 3.59 (s, 2H), 5.00-5.18 (m, 1H), 7.00-7.42
(m, 4H), 8.21 (s, 1H), 8.50 (s, 1H), 8.86 (s, 1H), 8.90-9.12 (m, 2H), 9.48 (s, 1H);
LCMS: R = 2.99 min; m/z 448 [M+H]+
The following Example was prepared according to the method described above for Example 297, starting
from (7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)[5-(methylamino)pyridinyl]methanone (Preparation
149) and the appropriate acid.
2-(4-cyanophenyl)-N-{5-[(7-isopropyl-7H-pyrrolo[2,3- LCMS: R = 2.86 min;
d]p n | carbon l]p ridin l-N-meth lacetamide m/z 439 [M+H]+
Example 299: 2-(4-Chloro-phenyl)-N-{5-[7-(3-hydroxymethyl-oxetanyl)-7H-pyrrolo[2,3-d]pyrimidine
carbonyl]—pyridinyl}-acetamide
W0 2012/137089 83
N \ N
k /W
| H
N N
OH CI
N-(5-{7-[3-(tert-Butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidinecarbonyl}-
pyridinyl)(4-chloro-phenyl)-acetamide (Preparation 203 35 mg, 0.059 mmol) was dissolved in dry
THF (0.5 mL) and tetrabutylammonium fluoride (0.065 mL of a 1M solution in THF, 0.065 mmoL) was
added. The reaction was stirred for 30 min at room temperature. The mixture was partitioned between
water and EtOAc. The layers were separated and the aqueous layer was extracted twice with EtOAc.
The combined organic layers were concentrated and purified by ative HPLC to give the title
compound in quantitative yield 28 mg.
LCMS (System 4): R = 3.03 min; m/z 478 [M+H]+
Example 300: 7—(3-Hydroxymethyl-oxetanyl)-7H-pyrrolo[2,3-d]pyrimidinecarbonyl]-pyridin
(4-trifluoromethyl-phenyl)—acetamide:
The title compound was prepared according to the method described for Example 299 starting from N-(5-
{7-[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidinecarbonyl}-pyridin
yl)(4-trifluoromethyl-phenyl)—acetamide (Preparation 204) to afford the title compound as a white solid
in quantitative yield, 30 mg. LCMS: R = 2.74 min; m/z 512 [M+H]+.
Example 301: hloropyridinyl)-N-[5-({7-[3-(hydroxymethyl)oxetanyl]—7H-pyrrolo[2,3-d]pyrimidin-
-yl}carbonyl)pyridinyl]acetamide
The title compound was prepared according to the method bed for Example 299 starting from N-[5-
({7-[3-({[tert-butyl(dimethyl)silyl]oxy}methyl)oxetanyl]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridin
yl](5-chloropyridinyl)acetamide (Preparation 223) to afford the title nd as a pale yellow liquid
in 87% yield, 23 mg. LCMS (System 4): R = 1.57 min; m/z 479 [M+H]+.
Example 302: N-[5-({7-[3-(hydroxymethyl)oxetan—3-yl]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridin
yl][3-(trifluoromethyl)-1H-pyrazolyl]acetamide
\ / O CF3
N/I \ ”4’“;
KN N
The title compound was prepared according to the method described for Example 299 starting from
N-[5-({7-[3-({[tert—butyl(dimethyl)silyl]oxy}methyl)oxetanyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyridinyl][3-(trifluoromethyl)-1H-pyrazolyl]acetamide (34 mg, 0.055 mmol)
(Preparation 224) to give the the title compound in 63% yield, 17.7mg.
LCMS m 5) R = 1.57 min; m/z 502 [M+H]+.
W0 2012/137089 84
Example 303: [7-(2-aminooxoethyl)-7H-pyrro|o[2,3-d]pyrimidiny|]carbony|}pyridiny|)(4-
chlorophenyl)acetamide
To a stirred solution of 2-(4-ch|oropheny|)-N-[5-(7H-pyrro|o[2,3-d]pyrimidiny|carbony|)pyridin
tamide (Example 308, 50 mg, 0.13 mmol) in DMF ( 1 ml) was added Cszcog (75mg, 0.23 mmol)
followed by 2-bromoacetamide (21 .3mg, 0.154 mmol). The reaction was then stirred at room temperature
overnight. The reaction was quenched with water (5 mL) and extracted with EtOAc (3 x 5 mL).
The organics were combined, washed with water (5 mL) then brine (5 mL), dried over anhydrous
magnesium sulfate and concentrated in vacuo. Purification by preparative HPLC gave the title compound
in 59% yield, 34mg.
LCMS m 4): R = 1.56 min; m/z 449 [M+H]+.
Example 304: 2-{5-[(5-{[(4-ch|oropheny|)acety|]amino}pyridiny|)carbony|]-7H-pyrro|o[2,3-d]pyrimidin
y|}-N,N-dimethylacetamide
{5-[(5-{[(4-ch|oropheny|)acety|]amino}pyridiny|)carbony|]-7H-pyrro|o[2,3-d]pyrimidiny|}acetic acid
potassium salt (Example 307, 50 mg, 0.102 mmol) was added to a stirred solution of dimethyl amine HCI
(12.5 mg, 0.153 mmol) and HATU (58.2mg, 0.153 mmol) in pyridine (2 mL) and the resultant solution was
stirred at 50°C (reactivial) for 14 hours. The reaction was cooled to 25°C, diluted with DCM (5 mL) then
quenched with saturated NaHCO3 (aq) (5 mL) and extracted with further DCM (3 x 5 mL). The organics
were ed, washed with saturated brine (5 mL), dried over anhydrous magnesium sulfate and
concentrated in vacuo. Purification by preparative HPLC gave the title compound in 55% yield, .
LCMS: R = 1.86 min; m/z 477 [M+H]+.
Example 305: 2-(4-ch|orophenyl)-N-[5-({7-[2-(methy|amino)oxoethy|]-7H-pyrro|o[2,3-d]pyrimidin
y|}carbony|)pyridiny|]acetamide
{5-[(5-{[(4-ch|oropheny|)acety|]amino}pyridiny|)carbony|]-7H-pyrro|o[2,3-d]pyrimidiny|}acetic acid
potassium salt (Example 307, 50 mg, 0.102 mmol) was added to a stirred solution of methyl amine
hydrochloride (10.3 mg, 0.153 mmol) and HATU (58.2mg, 0.153 mmol) in pyridine (2 mL) . The resultant
solution was stirred at 50 °C (reactivial) for 14 hours. The reaction was cooled to 25 oC, diluted with DCM
(5 mL) then ed with saturated NaHC03 (aq) (5 mL) and ted with further DCM (3 x 5 mL).
The organics were combined, washed with saturated brine (5 mL), dried over anhydrous magnesium
sulfate and trated in vacuo. Purification by preparative HPLC gave the title compound in 65% yield,
.5mg.
LCMS: R = 2.63 min; m/z 463 [M+H]+.
Example 306 Methyl {5-[(5-{[(4-ch|oropheny|)acety|]amino}pyridiny|)carbony|]-7H-pyrro|o[2,3-
d]pyrimidinyl}acetate
W0 2012/137089 85
\ / 0
\N N
To a stirred solution of 2-(4-chlorophenyl)-N-[5-(7H-pyrrolo[2,3-d]pyrimidinylcarbonyl)pyridin
y|]acetamide (Example 308, 250mg, 0.638 mmol) in DMF (4 mL) was added Cszcog (374mg, 1.15 mmol)
followed by methyl bromoacetate (73uL, 0.766 mmol). The reaction was stirred at 25 °C for 3 hours and
then quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The organics were combined,
washed with water (10 mL) and saturated brine (10 mL), dried over anhydrous magnesium sulfate and
concentrated in vacuo to give a pale yellow oil (356mg) which solidified on standing. Purification by
column chromatography on silica gel ent of 0-100% 90:10:1 DCM/MeOH/NH3 in DCM) gave the title
compound as a pale yellow solid in 70% yield, 208 mg. 1HNMR (400MHz, CDCI3) 6 3.78 (s, 2H), 3.81 (s,
3H), 5.15 (s, 2H), .41 (m, 4H), 7.51 (s, 1H), 7.88 (s, 1H), 8.46 (m, 1H), 8.78-8.82 (m, 2H), 9.04 (s,
1H), 9.65 (s, 1H). LCMS (System 4): R = 2.07 min; m/z 464 [M+H]+.
e 307 {5-[(5-{[(4-chlorophenyl)acetyl]amino}pyridinyl)carbonyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}acetic acid potassium salt
To a suspension of methyl {5-[(5-{[(4-chlorophenyl)acetyl]amino}pyridinyl)carbonyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}acetate le 306, 197 mg, 0.425 mmol) in MeOH (4 mL) was added an
aqueous solution of KOH (0.425 mL of a 1M solution, 0.425 mmol) and then further MeOH (4 mL) was
added. The reaction was stirred for 2 hours at room temperature. The on was concentrated in vacuo
to give the title compound as a light brown solid in 99% yield, 205 mg.
LCMS (System 4): R = 1.76 min; m/z 450 [M+H]+.
Example 308 : 2-(4-Chlorophenyl)-N-[5-(7H-pyrrolo[2,3-d]pyrimidinylcarbonyl)pyridinyl]acetamide
The title compound was ed according to the method described for Example 46 to afford the title
compound as a white solid in 87% yield, 930 mg.
0 \/ 0
N/ \ ”W0
1H NMR (400 MHz, DMSO) a: 3.74 (s, 2H), 7.38 (m, 4H), 8.34 (s, 1H), 8.46 (s, 1H), 8.71 (s, 1H), 8.94 (s,
1H), 8.97 (s, 1H), 9.45 (s, 1H), 10.66 (s, 1H), 13.14 (s, 1H);LCMS (system 9): R = 2.87 min; m/z 392
[M+H]+.
Example 309: 2-(4-chlorophenyl)-N-(5-{[7-(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidin
y|]carbonyl}pyridinyl)acetamide
W0 2012/137089 86
\ /o
Nl/\\
HNW0
N N
(5-aminopyridinyl)[7-(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidinyl]methanone (Preparation
222, 15.5 mg, 0.05 mmol) was added to a ng mixture of 4-chlorophenylacetic acid (11.1mg, 0.065
mmol) and HATU (28.5 mg, 0.075 mmol) in pyridine (0.25 mL). The mixture was heated to 50 °C and
stirred for 16 hours. The mixture was allowed to cool to room temperature and saturated sodium
bicarbonate on (5 mL) was added. The mixture was extracted with ethyl acetate (3 x 5 mL) and the
combined c fractions were washed with brine, dried (MgSO4) and the solvent was removed under
reduced re to obtain the crude product which was autopurified.
LCMS (system 4): R = 3.08 min; m/z 462[M+H]+.
The following Examples were prepared according to the method described above for Example 309
starting from 5-aminopyridinyl)[7-(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidinyl]methanone
(Preparation 222) and the appropriate acids.
2-(5-chloropyridinyl)-N-(5-{[7-(3-methyloxetan LCMS (System 2): R = 0.88 min;
yl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin m/z 463 [M+H]+
yl)acetamide Using Preparation 90
N-(5-{[7-(3-methyloxetanyl)-7H-pyrrolo[2,3- LCMS (System 2): R = 0.97 min;
d]pyrimidinyl]carbonyl}pyridinyl)[3- m/z 486 [M+H]+
(trifluoromethyl)—1H-pyrazolyl]acetamide
N-(5-{[7-(3-methyloxetanyl)-7H-pyrrolo[2,3- LCMS (System 2): R = 0.92 min;
d]pyrimidinyl]carbonyl}pyridinyl)[4- m/z 487 [M+H]+
(trifluoromethyl)—1H-1,2,3-triazolyl]acetamide Using Preparation 85
Example 313: 2-(4-chlorophenyl)—N-[5-({7-[(methylthio)methyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyridinyl]acetamide
zN CI
“i\ \ Mo
Potassium carbonate (38 mg, 0.275 mmol) was added to a stirring solution of 2-(4-chlorophenyl)-N-[5-
(7H-pyrrolo[2,3-d]pyrimidinylcarbonyl)pyridinyl]acetamide (Example 308, 60.0 mg, 0.153 mmol) in
DMF (1.0 mL) at room temperature. After 10 min, methyl methyl sulfide (19 uL, 0.23 mmol) was
added to the mixture and the reaction was stirred for 24 hours at room temperature. Water (3 mL) was
added to the e and it was extracted with EtOAc (3 x 5 mL). The combined organic fractions were
washed with water (5 mL), brine (5 mL), dried over magnesium e and concentrated under reduced
pressure to give a pale yellow oil. The crude material was purified by column chromatography on silica gel
(gradient of 100% DCM to 90:10:1 OH/NH3) to give the title compound as a pale yellow solid in
36% yield, 25 mg.
1H NMR (400 MHz, CDCI3) 6: 2.13 (s, 3H), 3.79 (s, 2H), 5.39 (s, 2H), 7.30-7.34 (m, 3H), 7.40-7.43 (m,
2H), 8.02 (s, 1H), 8.52 (m, 1H), 8.74-8.75 (d, 1H), 8.82-8.83 (d, 1H), 9.05 (s,1H), 9.65 (s, 1H); LCMS
(system 4): R = 1.97 min; m/z 452; 454 [M+H]+.
e 314 : hlorophenyl)-N-[5-({7-[(methylsulfonyl)methyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}carbonyl)pyridinyl]acetamide
Potassium peroxomonosulfate (Oxone, 67.2 mg, 0.110 mmol) was added to a stirring solution of 2-(4-
chlorophenyl)-N-[5-({7-[(methylthio)methyl]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyl]acetamide
(Example 313, 25.0 mg, 0.055 mmol) in methanol (1.0 mL) and water (0.25 mL) at 0 0C. After 1 hour, the
reaction was allowed to warm to room ature and stirred for 24 hours. The reaction mixture was
cooled to 0 °C and sodium metabisulfite (0.5M, 1ml) was added. The reaction mixture was evaporated
under reduced pressure to remove the methanol. Water (3 mL) was added to the mixture and it was
extracted with EtOAc (3 x 5 mL). The combined organic fractions were washed with water (5 mL), brine (5
mL), dried over magnesium sulfate the solvent was removed under reduced pressure to give the crude
product as an off-white solid. The crude material was ed by column chromatography on silica gel
(gradient of 100% DCM to 90:10:1 OH/NH3) to give the title compound as a pale yellow solid in
17% yield, 27 mg. LCMS (system 4): R = 1.91 min; m/z 484; 486 [M+H]+.
Example 315: yclopropyl-5—trifluoromethyl-1H-pyrazolyl)-N-[5-(7-isopropyl-7H-pyrrolo [2, 3-d]
pyrimidinecarbonyl)-pyridinyl]-acetamide
To a on of (5-Amino-pyridinyl)-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
(Preparation 95, 50 mg, 0.17 mmol), (1-Cyclopropyltrifluoromethyl-1H-pyrazolyl)—acetic acid
(Preparation 141, 47.1 mg, 0.21 mmol) and TEA (0.08 mL, 0.62 mmol) in THF (1 mL), 1-
propylphosphonic acid cyclic anhydride (50% solution in EtOAc, 0.26 mL, 0.44 mmol) was added and the
mixture was stirred at room temperature for 14 hours. The reaction mixture was evaporated under
reduced pressure and the residue partitioned between water and ethyl acetate. The organic layer was
W0 2012/137089 88
washed with saturated sodium onate solution and dried (NaZSO4) and evaporated in vacuo.
cation by column tography on silica gel (gradient of EtOAc:Hexane 0:100 to 80:20) gave the
title compound as a white solid in 76 % yield, 67 mg. 1H NMR (400 MHz, DMSO-D6) 6: 1.05-1.07 (m, 2H),
1.08-1.15 (m, 2H), 1.54 (d, 6H), 3.73-3.76 (m, 3H), 5.06-5.13 (m, 1H), 7.55 (s, 1H), 8.44 (s, 1H), 8.51 (s,
1H), 8.73 (s, 1H), 8.94 (d, 1H), 8.99 (s, 1H), 9.44 (s, 1H), 10.61 (s, 1H).LCMS (System 10): R = 3.03 min
m/z 498 [M+H]+
Example 316: N-(5-{[2-amino(1-hydroxy—2-methylpropanyl)-7H-pyrrolo[2,3-d]pyrimidin
bonyl}pyridinyl)(5-bromopyridinyl)acetamide
The title compound was prepared according to the method described for Example 34 at 50°C using [2-
amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl](5-aminopyridinyl)methanone
(Preparation 48) and (5-bromopyridinyl)acetic acid to afford the title compound as a yellow solid in
60% yield, 75 mg. LCMS (System 10): R: 2.69 min; m/z 524 [M+H]+
Example 317: 2-(5-bromopyridinyl)-N-(5-{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]carbonyl}pyridinyl)acetamide
The title compound was prepared ing to the method described for Example 1 with DIPEA using (5-
aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and (5-
bromopyridinyl)acetic acid to afford the title compound as a yellow solid in 45% yield, 75 mg. LCMS
(System 9): R = 2.97 min; m/z 479 [M+H]+.
Example 318 : N-{5-[(2-Aminoisopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(5-
bromopyridinyl)acetamide
The title compound was prepared according to the method described for Example 1 with DIPEA using (2-
aminoisopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)(5-aminopyridinyl)methanone
(Preparation 122) and (5-bromopyridinyl)acetic acid to afford the title compound as a colourless oil in
57% yield, 75 mg.
LCMS (System 10): R: 2.81 min; m/z 494 [M+H]+.
Library Protocol 1
The compounds below were prepared in el in the ing manner.
2 “4
X_R102
o o
/ /
\ N \ N
N \ \ —> N \ \
k /| EDCI/HOBT/NMM/ k /|
N N\ DMF/50C/2 hrs N N,
Me Me
A 0.25 M stock solution of (5-amino-pyridinyl)-(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
(Preparation 110) in anhydrous DMF was prepared. Stock solutions (0.30 M) of each acid monomer
was prepared in anhydrous DMF. A stock solution of EDCI (0.5 M) and HOBT (0.05 M) in anhydrous DMF
W0 2012/137089 89
were prepared. 300u| (90 umol, ) of each acid monomer solution was dispensed to 8 mL vials, followed by
300 pl (75 umol) of (5-amino-pyridinyl)-(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone solution.
N-methyl line (150 pmol, 2.0 eq), 300 pl EDCI solution (150 umol) and HOBT (15pmol) were
added to each vial. The vials were capped and shaken at 50 °C for 2 hours. The solvent was removed
using a ac, and the final product purified by HPLC under the conditions listed to provide the final
compounds.
2-(4-chlorophenyl)—N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.59 min;
yl)carbonyl]pyridinyl}acetamide m/z 406 [M+H]+
3-(4-chlorophenyl)—N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.70 min;
yl)carbonyl]pyridinyl}propanamide m/z 420 [M+H]+
3-(3-methylphenyl)-N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.69 min;
yl)carbonyl]pyridinyl}propanamide m/z 400 [M+H]+
N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin LCMS: R = 1.80 min;
yl}(pyridinyl)propanamide m/z 387 [M+H]+
3-(3-chlorophenyl)—N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.69 min;
yl)carbonyl]pyridinyl}propanamide m/z 420 [M+H]+
3-(3-fluorophenyl)-N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.56 min;
yl)carbonyl]pyridinyl}propanamide m/z 404 [M+H]+
(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin LCMS: R = 2.80 min;
yl}[2-(trifluoromethoxy)phenoxy]acetamide m/z 472 [M+H]+
ethylphenyl)-N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS:Rt = 2.63 min;
yl)carbonyl]pyridinyl}propanamide m/z 400 [M+H]+
3-(2-fluorophenyl)-N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.53 min;
| carbon l]p ridin l propanamide m/z 404 [M+H]+
3-(4-fluorophenyl)-N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS: R = 2.55 min;
yl)carbonyl]pyridinyl}propanamide m/z 404 [M+H]+
(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin LCMS:Rt = 2.50 min;
yl}phenylpropanamide m/z 386 [M+H]+
3-(4-methylphenyl)-N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin LCMS:Rt = 2.65 min;
yl)carbonyl]pyridinyl}propanamide m/z 400 [M+H]+
N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin LCMS: R = 2.60 min;
yl}(4-{[(trifluoromethyl)sulfonyl]amino}phenyl)acetamide m/z 519 [M+H]+
N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin LCMS: R = 2.53 min;
yl}phenoxyacetamide m/z 388 [M+H]+
2-[4-(methoxymethyl)phenyl]-N-{5-[(7-methyl-7H-pyrrolo[2,3- LCMS: R = 2.31 min;
d]pyrimidinyl)carbonyl]pyridinyl}acetamide m/z 416 [M+H]+
2-(4-fluorophenoxy)—N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidin-5— LCMS: R = 2.44 min;
| carbon l]p ridin l acetamide m/z 406 [M+H]+
W0 2012/137089 90
Example 336: N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}
phenylcyclopropanecarboxamide
The title compound was prepared according to the method described for Library protocol 1 starting from
(5-amino-pyridinyl)-(7-methy|-7H-pyrrolo[2,3-d]pyrimidiny|)-methanone (Preparation 110) and 1-
phenylcyclopropanecarboxylic acid to afford the title compound. LCMS: R = 2.61 min; m/z 398 [M+H]+'
Example 337: N-{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}indanecarboxamide
The title compound was prepared ing to the method described for y protocol 1 starting from
(5-amino-pyridinyl)-(7-methy|-7H-pyrrolo[2,3-d]pyrimidiny|)-methanone ration 110) and
indanecarboxylic acid to afford the title compound. LCMS: R = 2.58 min; m/z 398 [M+H]+'
Example 338: -{5-[(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}
phenylpropanamide
The title compound was prepared according to the method described for Library protocol 1 starting from
(5-amino-pyridinyl)-(7-methy|-7H-pyrrolo[2,3-d]pyrimidiny|)-methanone (Preparation 110) and (2R)-
ylpropanoic acid to afford the title compound. LCMS: R = 2.52 min; m/z 386 [M+H]+'
Example 339: 3-hydroxy—2-phenyl-N-(5-{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin-
3-yl)propanamide
\ / o
N/ \ H
KN I N
)‘Me OH
The title compound was prepared according to the method described for Example 167 using (5-
aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 95) and tropic
acid to afford the title compound as a yellow solid in 7% yield, 15 mg.
1H NMR (400 MHz, DMSO-d6) 6: 1.57 (d, 6H), 3.16-3.22 (m, 1H), 3.37-3.41 (m, 1H), 3.60-3.64 (m, 1H),
.10 (m, 1H), 6.30 (t, 1H), 7.10 (t, 1H), 7.20 (t, 2H), 7.30-7.32 (m, 3H), 8.12 (s, 1H), 8.15 (d, 1H), 8.61 (s,
1H), 8.96 (s, 1H), 9.44 (s, 1H).
LCMS (system 10): R: 2.36 min; m/z 430 [M+H]+.
The following Example was prepared according to es 1 and 34 for s a and b as described
above.
W0 2012/137089 91 2012/051363
N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]- LCMS (system 8): R =
pyridinyl}(5-methoxy-1H-indolyl)acetamide 1.63 min; m/z 469 [M+H]+
The following examples were prepared ing to Example 356 using (5-amino-pyridinyl)-(7-
isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)—methanone (Preparation 95) and the appropriate acid.
LCMS R: 2.95 min; m/z 420
3-chloro-N-(5-{[7-(propanyl)-7H-pyrrolo[2,3-
d]pyrimidinyl]carbonyl}pyridinyl)benzamide
4-chloro-N-(5-{[7-(propanyl)-7H-pyrrolo[2,3- LCMS R: 2.93 min; m/z 420
d]pyrimidinyl]carbonyl}pyridinyl)benzamide [M+H]+.
N-(5-{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrimidin LCMS R: 2.99 min; m/z 454
yl]carbonyl}pyridinyl) [M+H]+.
(trifluoromethyl)benzamide
N-(5-{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrimidin
LCMS R: 3.05 min; m/z 470
yl]carbonyl}pyridinyl) [M+H]+.
(trifluoromethoxy)benzamide
2-(2-cyanophenoxy)-N-(5-{[7-(propanyl)-7H-
LCMS R: 0.67 min; m/z 441
o[2,3-d]pyrimidinyl]carbonyl}pyridin [M+H]+.
yl)acetamide
N-(5-{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrimidin
LCMS R: 3.01 min; m/Z 500
yl]carbonyl}pyridinyl)[4- [M+H]+.
(trifluoromethoxy)phenoxy]acetamide
Example 350 : 2-amino(4-chlorophenyl)-N-(5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidine
carbonyl)pyridinyl)acetamide
To utyl (1-(4-chlorophenyl)((5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)pyridin
yl)amino)oxoethyl)carbamate (Preparation 299, 64 mg, 0.11 mmol) was added 4M HCI/dioxane (5 mL)
and the reaction stirred at room temperature for 18 hours. The mixture was evaporated in vacuo and
purified by preparative reverse phase HPLC to give the title compound as a beige solid in 8% yield, 4 mg.
W0 2012/137089 92
1H NMR (400 MHz, MeOD) 6: 1.59 (d, 6H), 4.63 (s, 1H), 5.16 (m, 1H), 7.35 (d, 2H), 7.47 (d, 2H), 8.33 (s,
1H), 8.62 (s, 1H), 8.69 (s, 1H), 8.88 (s, 1H), 8.92 (s, 1H), 9.47 (s, 1H).
LCMS R = 2.81 min; MS m/z 449 [M+H]+
Example 351: N-{5-[2-Amino(2-hydroxy-1,1-dimethyl-ethy|)-7H-pyrrolo[2,3-d]pyrimidinecarbonyl]-
nyl}(5-cyano-pyridinyl)-acetamide
H N
0 /\
N|\\
HNJ\N/N
To a solution of N-{5-[2-Amino(2-hydroxy-1,1-dimethyl-ethy|)-7H-pyrrolo[2,3-d]pyrimidinecarbonyl]—
pyridinyl}(5-bromo-pyridiny|)-acetamide (Example 316, 75 mg, 0.143 mmol) in DMF (2 mL) was
added Zn(CN)2 (25 mg, 0.215 mmol) and the reaction mixture was degassed with argon for 10 minutes.
Pd2(dba)3 (3 mg, 0.002 mmol) and is(diphenylphosphino)ferrocene 6 mg, 0.011 mmol) were then
added and the resultant reaction mixture was heated at 1000C for 40 minutes under microwave irradiation.
The reaction mixture was diluted with EtOAc (20 mL) and washed with water (2 x 10 mL) and brine (10
mL). The c layer was dried (NaZSO4) and evaporated in vacuo. The crude material was purified by
preparative TLC (7% MeOH in DCM) to afford the title compound as yellow solid in 31% yield, 21 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.63 (s, 6H), 3.89 (d, 2H), 4.06 (s, 2H), 5.04 (t, 1H), 6.53 (s, 2H), 7.65-
7.68 (m, 2H), 8.29 (dd, 1H), 8.40 (s, 1H), 8.64 (d, 1H), 8.93-8.97 (m, 3H), 10.75 (s, 1H).
LCMS (system 10): R = 2.50 min MS m/z 471[M+H]+
Example 352: 2-(1-Cyclopropyltrifluoromethyl-1H-pyrazo|y|)-N-[5-(7-isopropyl-7H-pyrrolo [2, 3-d]
pyrimidinecarbony|)-pyridiny|]-acetamide
To a solution of (5-Amino-pyridinyl)-(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)-methanone
(Preparation 95, 50 mg, 0.17 mmol), lopropyltrifluoromethyl-1H-pyrazolyl)—acetic acid
(Preparation 148, 47.1 mg, 0.21 mmol) and TEA (0.08 mL, 0.62 mmol) in THF (1mL), 1-
phosphonic acid cyclic anhydride (50% solution in EtOAc, 0.26 mL, 0.44 mmol) was added and the
mixture was stirred at room temperature for 18 hours. The reaction mixture was evaporated under
d pressure and the residue partitioned between water and EtOAc. The organic layer was washed
with saturated sodium bicarbonate solution, dried (NaZSO4) and evaporated in vacuo. Purification by
W0 2012/137089 93
column chromatography on silica gel (EtOAc) gave the title nd as a white solid in 77 % yield, 68
1H NMR (400 MHz, DMSO-D6) 6: 1.00-1.01 (m, 2H), 1.06-1.14 (m, 2H), 1.54 (d, 6H), 3.70 (s, 2H), 3.84
(m, 1H), 5.08-5.12 (m, 1H), 7.98 (s, 1H), 8.45 (s, 1H), 8.51 (s, 1H), 8.74 (s, 1H), 8.96 (d, 1H), 8.99 (s, 1H),
9.44 (s, 1H), 10.60 (s, 1H).
LCMS (system 10): R = 3.02 min MS m/z 498 [M+H]+.
Example 353: racemic Methyl 2-{5-[(5-{[(4-chlorophenyl)acetyl]amino}pyridinyl)carbonyl]—7H-
o[2,3-d]pyrimidinyl}propanoate
The title compound was prepared according to the method described for Example 229 using 2-(4-
chlorophenyl)-N-[5-(7H-pyrrolo[2,3-d]pyrimidinylcarbonyl)pyridinyl]acetamide (Example 308), methyl
2-bromopropionate and cesium carbonate. Purified using preparative HPLC (method 1) to afford the title
compound.
LCMS (system 2): R = 1.42 min MS m/z 478 [M+H]+
Example 354: c 2-{5-[(5-{[(4-Chlorophenyl)acetyl]amino}pyridinyl)carbonyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}propanoic acid
The title compound was prepared according to the method described for Preparation 155 using methyl 2-
{5-[(5-{[(4-chlorophenyl)acetyl]amino}pyridinyl)carbonyl]-7H-pyrrolo[2,3-d]pyrimidinyl}propanoate
(Example 353) to afford the title nd as a white solid in 100% yield, 97 mg.
LCMS (system 2): R = 1.40 min; m/z 464 [M+H]+.
Example 355: 2-(4,5-Dichloro-imidazoly|)-N-[5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)-
pyridinyl]-acetamide (method d)
N \ NH
KNwe:o
N?0.
(4,5-Dichloro-imidazoly|)-aceticacid (25.2 mg, 0.130 mmol) was added to (5-amino-pyridinyl)-(7-
isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone (28.1 mg, 0.1 mmol), HATU (49.4 mg, 0.130 mmol)
and DIPEA (51.7 uL, 0.300 mmol) in anhydrous DMF (1 mL). The mixture was stirred at 50°C for 16 hours
and then ated in vacuo and purified by prep-HPLC (method 5) to afford the title compound in 46%
yield, 21.2 mg.
LCMS (system 8): R = 1.55 min; m/z 458 [M+H]
Example 356: 7-Difluoromethylmethyl-[1,2,4]triazolo[1,5-a]pyrimidinecarboxylic acid [5-(7-isopropyl-
7H-pyrrolo[2,3-d]pyrimidinecarbonyl)-pyridinyl]-amide (method e)
W0 2012/137089 94
7-Difluoromethylmethyl-[1,2,4]triazolo[1,5-a]pyrimidinecarboxylic acid (29.64 mg, 0.130 mmol) was
added to (5-amino-pyridinyl)-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone (28.1 mg, 0.1
mmol), HATU (49.42 mg, 0.130 mmol) and DIPEA (22.4 uL, 0.13 mmol) in anhydrous DMF (1 mL). The
mixture was stirred at 50°C for 16 hours and then evaporated in vacuo and purified by prep-HPLC
(method 5) to afford the title compound in 12 % yield, 6.1 mg.
LCMS (system 8): R = 1.56 min; m/z 492 [M+H]+
The following Examples were prepared according to one of the methods for Examples 355 (Method d)
and 356 d e) using (5-aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
(Preparation 95).
LCMS (system 8): R = 1.55
2-(4,5-Dichloro-imidazoly|)-N-[5-(7-
min; m/z 458 [M+H]+.
isopropyl-7H-pyrrolo[2,3-d]pyrimidine
Prep HPLC (method 5)
carbonyl)—pyridinyl]-acetamide
2-(3,5-Dimethyl-1,2,4-triazoly|)-N-[5-(7- LCMS (system 8): R: 1.43
isopropyl-7H-pyrrolo[2,3-d]pyrimidine min; m/z 419 .
carbonyl)-pyridinyl]-acetamide Prep HPLC d 5)
7-lsopropyl-7H-pyrrolo[2,3- LCMS (system 8): R: 1.27
d]pyrimidinecarbonyl)—pyridinyl](2- min; m/z 404 [M+H]+.
methyl-imidazoly|)-acetamide Prep HPLC (method 5)
2-lmidazo[2,1-b]thiazolyl-N-[5-(7- LCMS (system 8): R: 1.42
isopropyl-7H-pyrrolo[2,3-d]pyrimidine min; m/z 446 [M+H]+.
carbonyl)-pyridinyl]-acetamide Prep HPLC (method 5)
2-(4-Hydroxy-phthalaziny|)-N-[5-(7- LCMS m 8): R = 1.46
isopropyl-7H-pyrrolo[2,3-d]pyrimidine min; m/z 468 [M+H]+.
carbonyl)—pyridinyl]-acetamide Prep HPLC (method 5)
2-(2,3-Dimethy|-imidazo[2,1-b]thiazo|—6-y|)-N- LCMS (system 8): R = 1.48
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 474 [M+H]+.
-carbonyl)—pyrid inyl]—acetam ide Prep HPLC (method 5)
2-[4-(1-Hydroxy-cyc|openty|)-1,2,3-triazo| LCMS (system 8): R = 1.44
y|]-N-[5-(7-isopropy|-7H-pyrro|o[2,3- min; m/z 475 [M+H]+.
d]pyrimidinecarbonyl)—pyridiny|]- Prep HPLC d 5)
acetamide (From Prep 294)
N-[5-(7-|sopropy|-7H-pyrro|o[2,3- LCMS (system 8): R = 1.57
d]pyrim idinecarbonyl)—pyridiny|](5-m- min; m/z 482 [M+H]+.
to|y|-tetrazo|y|)-acetamide Prep HPLC (method 5)
2-[4-(4-FIuoro—pheny|)-imidazoIy|]-N-[5-(7- LCMS (system 8): R = 1.45
isopropyI-7H-pyrro|o[2,3-d]pyrimidine
min; m/z 484 [M+H]+.
carbonyl)-pyridinyI]-acetamide
Prep HPLC (method 5)
LCMS (system 8): R = 1.60
min; m/z 432 [M+H]+.
2-(5-|sopropy|-pyrazo|y|)-N-[5-(7- Prep HPLC (method 5)
isopropyI-7H-pyrro|o[2,3-d]pyrimidine (Acid can be prepared in an
carbonyl)-pyridinyI]-acetamide analogus to method given in
WOO3/072572)
2-(2-EthyI-imidazo[2,1-b]—1,3,4-thiadiazol
LCMS (system 8): R = 1.60
y|)-N-[5-(7-isopropy|-7H-pyrro|o[2 ,3-
min; m/z 475 .
d]pyrimidinecarbonyl)—pyridiny|]-
Prep HPLC (method 5)
acetamide
LCMS (system 8): R = 1.45
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 460 [M+H]+.
midinecarbonyl)—pyridiny|](2-
Prep HPLC (method 5)
methy|-imidazo[2,1-b]thiazo|y|)-acetamide
(from Prep 289)
hloromethyI-1,2,4-triazo|y|)-N-[5- LCMS (system 8): R: 1.48
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 439 .
yl)-pyridinyI]-acetamide Prep HPLC (method 5)
N-[5-(7-|sopropy|-7H-pyrro|o[2,3- LCMS (system 8): R = 1.34
d]pyrimidinecarbonyl)—pyridiny|](2- min; m/z 466 [M+H]+.
phenyl-imidazoIy|)-acetamide Prep HPLC (method 5)
2-(2-Chloro-imidazo[2,1-b]thiazo|y|)-N-[5-
LCMS (system 8): R = 1.59
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
min; m/z 480 [M+H]+.
carbonyl)—pyridinyI]-acetamide
Prep HPLC (method 5)
LCMS (system 8): R = 1.26
2-|midazo|y|-N-[5-(7-isopropy|-7H-
min; m/z 390 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbonyl)—pyridin-
Prep HPLC d 5)
3-yI]-acetam ide
LCMS (system 8): R = 1.46
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 460 [M+H]+.
d]pyrimidinecarbonyl)—pyridiny|](3-
Prep HPLC (method 5)
methy|-imidazo[2,1-b]thiazo|y|)-acetamide
LCMS (system 8): R: 1.57
2-[1-(4-Hydroxy-pheny|)-1H-pyrroIy|]-N-[5-
min; m/z 481 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbonyl)-pyridinyI]-acetamide
N-[5-(7-|sopropy|-7H-pyrro|o[2,3- LCMS (system 8): R = 1.41
d]pyrimidinecarbonyl)—pyridiny|] min; m/z 442 [M+H]+.
[1 ,2,4]triazo|o[1 ,5-a]pyrimidinyI-acetamide Prep HPLC (method 5)
LCMS (system 8): R: 1.44
7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 402 .
midinecarbonyl)—pyridiny|]
Prep HPLC (method 5)
pyrazinyI-acetamide
LCMS (system 8): R = 1.48
2-[4-(3-Hydroxy—pheny|)-1,2,3-triazo|y|]-N-
min; m/z 483 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
onyl)—pyrid inyl]—acetam ide
(from Prep 292)
LCMS (system 8): R = 1.63
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 468 [M+H]+.
d]pyrimidinecarbonyl)—pyridiny|](5-
Prep HPLC (method 5)
phenyl-tetrazoIy|)-acetamide
7-DifluoromethyImethyl-[1,2,4]triazo|o[1,5- LCMS (system 8): R = 1.56
midinecarboxylic acid [5-(7- min; m/z 492 .
isopropy|-7H-pyrro|o[2,3-d]pyrimidine Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.73
Furo[3,2-c]pyridinecarboxylic acid [5-(7-
min; m/z 427 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
4-MethyI-furazancarboxylic acid [5-(7- LCMS (system 8): R: 1.61 min;
py|-7H-pyrro|o[2,3-d]pyrimidine m/z 392 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
LCMS (system 8): R = 1.75
1-MethyI-1H-indazoIecarboxylic acid [5-(7-
min; m/z 440 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.67
1H-Indolecarboxylic acid [5-(7-isopropyl-
min; m/z 425 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|)-
Prep HPLC (method 5)
pyridiny|]-amide
yItrifluoromethy|-thiazo|e LCMS (system 8): R = 1.62
carboxylic acid [5-(7-isopropyI-7H- min; m/z 475 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS (system 8): R = 1.67
-CyclopropyI-oxazolecarboxylic acid [5-
min; m/z 417 .
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.52
Pyridazinecarboxylic acid [5-(7-isopropyl-
min; m/z 388 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|)-
Prep HPLC (method 5)
Pyridinyl]—amide
3-MethyItrifluoromethyl-1H-pyrazoIe LCMS (system 8): R = 1.53
carboxylic acid [5-(7-isopropyI-7H- min; m/z 458 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide (Acid from prep 290)
4-MethyI-pyridinecarboxylic -(7- LCMS (system 8): Rt = 1.70
py|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 401 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
3-CyclobutyI-1H-pyrazoIecarboxylic acid LCMS (system 8): Rt = 1.59
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 430 [M+H]+.
ony|)-pyrid iny|]-am ide Prep HPLC (method 5)
OxazoIecarboxylic acid [5-(7-isopropyl- LCMS (system 8): Rt = 1.46
ro|o[2,3-d]pyrim idinecarbony|)- min; m/z 377 [M+H]+.
ny|]-amide Prep HPLC (method 5)
4-MethyI-1H-imidazoIecarboxylic acid [5- LCMS (system 8): Rt = 1.52
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 390 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
LCMS (system 8): Rt = 1.56
1H-Pyrro|o[2,3-c]pyridinecarboxylic acid min; m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide (Acid: Synthesis, 1993, 295-
297)
-MethyI-pyrazinecarboxylic acid[5-(7- LCMS (system 8): Rt = 1.58
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 402 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
1 ,3-DimethyI-1H-pyrazoIecarboxylic acid LCMS (system 8): Rt = 1.53
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 404 [M+H]+.
-carbony|)-pyrid iny|]-am ide Prep HPLC (method 6)
1H-Pyrazo|o[4,3-b]pyridinecarboxylic acid LCMS (system 8): Rt = 1.46
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 427 [M+H]+.
ony|)-pyrid iny|]-am ide Prep HPLC (method 5)
(Acid: see US 20110111046)
-EthyI-isoxazolecarboxylic acid[5-(7- LCMS (system 8): Rt = 1.63
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 405 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
2-Methyl-2H-pyrazoIecarboxylic acid [5- LCMS (system 8): Rt = 1.54
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 390 [M+H]+.
carbony|)-pyrid |]-am ide Prep HPLC (method 5)
Quinolinecarboxylic acid [5-(7-isopropyl- LCMS (system 8): Rt = 1.66
7H-pyrro|o[2,3-d]pyrim idinecarbony|)- min; m/z 437 [M+H]+.
Pyridinyl]—amide Prep HPLC (method 5)
-Fluoro-1H-pyrro|o[2,3-b]pyridine LCMS (system 8): Rt = 1.61
carboxylic acid isopropyI-7H- min; m/z 444 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC d 5)
3-y|]-amide (Acid: W02008/107543)
1 ,5-DimethyI-1H-pyrazoIecarboxylic acid LCMS (system 8): Rt = 1.57
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 404 [M+H]+.
-carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
2-CyclopropyI-oxazolecarboxylic acid [5- LCMS (system 8): Rt = 1.64
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 417 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
4-Trifluoromethy|-thiazoIecarboxylic acid LCMS (system 8): Rt = 1.58
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 461 [M+H]+.
-carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
LCMS (system 8): Rt = 1.51
min; m/z 416 [M+H]+.
6,7-Dihydro-5H-pyrro|o[1,2-c]imidazo|e
Prep HPLC (method 5)
carboxylic acid [5-(7-isopropyI-7H-
(Acid can be preparaed by
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
oxidation of aldehyde bed
3-y|]-amide
in Tetrahedron, 1999, 8111)
Thieno[3,2-c]pyridinecarboxylic acid [5-(7- LCMS (system 8): Rt = 1.77
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 443 .
carbony|)-pyrid iny|]-am ide Prep HPLC d 5)
1-Methy|trif|uoromethyl-1H-pyrazoIe
LCMS (system 8): Rt = 1.60
carboxylic acid [5-(7-isopropyI-7H-
min; m/z 458 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
Prep HPLC (method 5)
3-y|]-amide
WO 37089 100
|midazo[1,5-a]pyridinecarboxylicacid [5- LCMS (system 8): Rt = 1.47
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 426 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
1H-Pyrro|o[2,3-b]pyridinecarboxylic acid LCMS (system 8): Rt = 1.54
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 426 [M+H]+.
-carbony|)-pyrid iny|]-am ide Prep HPLC d 5)
3-EthyI-isoxazolecarboxylic acid[5-(7- LCMS (system 8): Rt = 1.60
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 405 [M+H]+.
y|)-pyrid iny|]-am ide Prep HPLC (method 5)
4-Methoxy—pyridinecarboxylic acid [5-(7- LCMS (system 8): Rt = 1.67
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 417 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
Pyrimidinecarboxylic acid [5-(7-isopropyl- LCMS (system 8): Rt = 1.46
7H-pyrro|o[2,3-d]pyrim idinecarbony|)- min; m/z 388 .
pyridiny|]-amide Prep HPLC (method 5)
3-MethyI-1H-pyrazoIecarboxylic acid [5-
LCMS (system 8): Rt = 1.46
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
min; m/z 390 [M+H]+.
carbony|)-pyrid |]-am ide
Prep HPLC (method 5)
-MethyI-1H-pyrrolo[2,3-b]pyridine LCMS (system 8): Rt = 1.54
carboxylic acid [5-(7-isopropyI-7H-
min; m/z 440 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
Prep HPLC (method 5)
3-y|]-amide
-MethyI-pyrimidinecarboxylic acid [5-(7- LCMS (system 8): Rt = 1.60
py|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 402 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
Thieno[3,2-b]pyridinecarboxylic acid [5-(7- LCMS (system 8): Rt = 1.64
py|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 443 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
Pyrazolo[1,5-a]pyrimidinecarboxylic acid LCMS (system 8): Rt = 1.54
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- min; m/z 427 [M+H]+.
ony|)-pyrid iny|]-am ide Prep HPLC (method 5)
4,5,6,7-Tetrahydro-1,2-benzisoxazoIe
LCMS (system 8): Rt = 1.75
carboxylic acid [5-(7-isopropyI-7H-
min; m/z 431 [M+H]+.
o[2,3-d]pyrimidinecarbony|)-pyridin-
Prep HPLC (method 5)
3-y|]-amide
|midazo[1,5-a]pyridinecarboxylicacid [5- LCMS (system 8): Rt = 1.75
propyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 426 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
LCMS (system 8): Rt = 1.65
1H-Pyrro|o[2,3-b]pyridinecarboxylic acid
min; m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
1,4,5,6-Tetrahydro-cyc|opentapyrazo|e LCMS (system 8): Rt = 1.59
carboxylic acid [5-(7-isopropyI-7H- min; m/z 416 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS (system 8): Rt = 1.41
4-Amino-pyrimidinecarboxylic acid [5-(7-
min; m/z 403 [M+H]+.
py|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): Rt = 1.32
1H-Pyrro|o[2,3-c]pyridinecarboxylic acid
min; m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): Rt = 1.51
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 401 [M+H]+.
d]pyrimidinecarbony|)-pyridiny|]
Prep HPLC (method 5)
methyl-isonicotinamide
LCMS m 8): Rt = 1.33
|midazo[1,2-a]pyridinecarboxylicacid [5-
min; m/z 426 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
1H-1,2,4-TriazoIecarboxylic acid[5-(7- LCMS (system 8): Rt = 1.38
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 377 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 7)
oxy—pyridinecarboxylic acid [5-(7-
LCMS (system 8): Rt = 1.66
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
min; m/z 417 [M+H]+.
carbony|)-pyrid iny|]-am ide
Prep HPLC (method 5)
Cinnolinecarboxylic acid [5-(7-isopropyl- LCMS (system 8): Rt = 1.57
7H-pyrro|o[2,3-d]pyrim idinecarbony|)- min; m/z 438 [M+H]+.
pyridiny|]-amide Prep HPLC (method 5)
[1 ,2,4]Triazo|o[1 ,5-a]pyridinecarboxylic LCMS m 8): Rt = 1.51
acid [5-(7-isopropyI-7H-pyrro|o[2,3- min; m/z 427 [M+H]+.
d]pyrim 5-carbony|)-pyridiny|]-am ide Prep HPLC (method 5)
Pyrazolo[1,5-a]pyrimidinecarboxylic acid
LCMS m 8): Rt = 1.54
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
min; m/z 427 [M+H]+.
-carbony|)-pyrid iny|]-am ide
Prep HPLC (method 5)
Pyridinecarboxylic acid [5-(7-isopropyl- LCMS (system 8): Rt = 1.62
7H-pyrro|o[2,3-d]pyrim idinecarbony|)- min; m/z 387 [M+H]+.
Pyridinyl]—amide Prep HPLC (method 5)
-Ch|oro-N-[5-(7-isopropy|-7H-pyrro|o[2,3- LCMS (system 8): Rt = 1.62
d]pyrimidinecarbony|)-pyridiny|]- min; m/z 421 [M+H]+.
nicotinamide Prep HPLC (method 5)
4H-Furo[3,2-b]pyrro|ecarboxy|ic acid [5-
LCMS (system 8): R = 1.59
(7-isopropyI-7H-pyrrolo1[2,3-d]pyrimidine
min; m/z 415 [M+H]+.
carbony|)-pyrid |]-am ide
Prep HPLC (method 5)
LCMS (system 8): R: 1.34 min;
|midazo[1,2-a]pyridinecarboxylicacid [5-
m/z 426 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.52 min;
1-EthyI-1H-pyrazoIecarboxylic acid [5-(7-
m/z 404 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid |]-am ide
LCMS (system 8): R = 1.58
1-|sopropy|-1H-imidazoIecarboxylic acid min; m/z 418 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine- Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide (Acid :WO2010/009062
LCMS (system 8): R = 1.62
8—Methoxy—imidazo[1 ,2-a]pyrazine min; m/z 457 [M+H]+.
carboxylic acid [5-(7-isopropyI-7H- Prep HPLC (method 5)
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- (Acid can be prepared by
3-y|]-amide carbonylation of the bromide
e.g. /078408)
LCMS m 8): R = 1.55
1H-Pyrro|o[2,3-b]pyridinecarboxylic acid
min; m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
ony|)-pyrid |]-am ide
LCMS (system 8): R: 1.69 min;
2-Methy|propy|-2H-pyrazoIecarboxylic
m/z 432 [M+H]+.
acid [5-(7-isopropyI-7H-pyrro|o[2,3-
Prep HPLC (method 5)
d]pyrim idinecarbony|)-pyridiny|]-am ide
LCMS (system 8): R: 1.79 min;
4-|sopropy|-thiazoIecarboxylic acid [5-(7- m/z 435 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide (Acid: WO2007/017144
LCMS (system 8): R: 1.77 min;
3-MethyI-furo[2,3-c]pyridinecarboxylic acid m/z 441 [M+H]+.
isopropyI-7H-pyrro|o[2,3-d]pyrim idine- Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide (Acid: WOO4052348)
LCMS m 8): R: 1.69 min;
6-MethyI-pyridinecarboxylic acid[5-(7-
m/z 401 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
-Chloromethy|trif|uoromethyl-1H- LCMS (system 8): R = 1.63
pyrazoIecarboxylic acid [5-(7-isopropyl- min; m/z 492 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|)- Prep HPLC (method 5)
pyridiny|]-amide
LCMS (system 8): R: 1.63 min;
oxy—1H-benzimidazole-2—carboxylic m/z 456 [M+H]+.
acid [5-(7-isopropyI-7H-pyrro|o[2,3- Prep HPLC (method 5)
d]pyrim idinecarbony|)-pyridiny|]-am ide (Acid: /017144)
4-Chloro—1,5-dimethyI-1H-pyrazoIe LCMS (system 8): R: 1.65 min;
carboxylic acid [5-(7-isopropyI-7H- m/z 438 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
amide
LCMS (system 8): R, = 1.58
1H-Pyrro|o[3,2-c]pyridinecarboxylic acid
min; m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.76 min;
4-EthyI-pyridinecarboxylic acid [5-(7- m/z 415 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide (Acid: JOC, 1990, 55, 738-741)
2,3-Dihydro-1,4-dioxino[2,3-b]pyridine LCMS (system 8): R, = 1.54
carboxylic acid [5-(7-isopropyI-7H- min; m/z 445 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS (system 8): R: 1.58 min;
ro|o[3,2-b]pyridinecarboxylic acid
m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R, = 1.59
min; m/z 426 [M+H]+.
Pyrazo|o[1,5-a]pyridinecarboxylic acid [5-
Prep HPLC d 5)
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
(Acid: J. Med. Chem. (2007),
carbony|)-pyrid iny|]-am ide
45(21), 4594-4597)
LCMS m 8): R, = 1.68
6-Methoxy—quinolinecarboxylic acid [5-(7-
min; m/z 467 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC d 5)
carbony|)-pyridiny|]-amide
-MethyI(2,2,2-trif|uoro-ethy|)-2H- LCMS (system 8): R: 1.66 min;
pyrazoIecarboxylic acid [5-(7-isopropyl- m/z 472 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|)- Prep HPLC (method 5)
pyridiny|]-amide (Acid: )
LCMS (system 8): R = 1.58
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 455 [M+H]+.
midinecarbony|)-pyridiny|]
Prep HPLC (method 5)
trifluoromethyI-nicotinamide
LCMS (system 8): R: 1.46 min;
1-(2-Cyano-ethy|)-1H-pyrazoIecarboxylic
m/z 429 .
acid [5-(7-isopropyI-7H-pyrro|o[2,3-
Prep HPLC (method 5)
d]pyrim idinecarbony|)-pyridiny|]-am ide
LCMS (system 8): R = 1.45
min; m/z 427 [M+H]+.
Furo[3,2-c]pyridinecarboxylic acid [5-(7-
Prep HPLC (method 5)
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
(Acid: J.Het.Chem.
carbony|)-pyrid iny|]-am ide
,24(2), 373-6)
LCMS (system 8): R: 1.32 min;
1H-Pyrro|o[3,2-c]pyridinecarboxylic acid
m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.70
Furo[3,2-c]pyridinecarboxylic acid [5-(7-
min; m/z 427 .
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.58
2,4-Dimethyl-oxazolecarboxylic acid [5-(7-
min; m/z 405 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.32
yI-1H-pyrro|o[2,3-c]pyridine
min; m/z 440 [M+H]+.
carboxylic acid [5-(7-isopropyI-7H-
Prep HPLC (method 5)
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
(Acid: 7144)
3-y|]-amide
,6-Dihydro-4H-pyrro|o[1,2-b]pyrazo|e LCMS (system 8): R = 1.59
carboxylic acid [5-(7-isopropyI-7H- min; m/z 416 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS (system 8): R: 1.79 min;
2-CyclobutyI-thiazoIecarboxylic acid [5-(7- m/z 447 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide (Acid: WO2009098448)
LCMS (system 8): R: 1.53 min;
3-FIuoro-N-[5-(7-isopropyI-7H-pyrro|o[2,3-
m/z 405 [M+H]+.
d]pyrimidinecarbony|)-pyridiny|]-
Prep HPLC (method 5)
isonicotinamide
yI-imidazo[1,2-a]pyrimidine LCMS (system 8): R: 1.52 min;
carboxylic acid [5-(7-isopropyI-7H- m/z 441 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS m 8): R: 1.57 min;
-Methy|-isoxazoIecarboxylic acid [5-(7-
m/z 391 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.62
methoxy—pyrimidinecarboxylic acid
min; m/z 448 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.58 min;
yI-pyrazinecarboxylic acid[5-(7-
m/z 402 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R = 1.49
yI-1H-imidazoIecarboxylic acid [5- min; m/z 390 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine Prep HPLC
carbony|)-pyrid iny|]-am ide (method 5)
LCMS (system 8): R: 1.71 min;
-Chloro—pyridinecarboxylic acid[5-(7-
m/z 421 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.68 min;
1,8—Naphthyridinecarboxylic acid[5-(7-
m/z 438 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.59 min;
yI-1H-imidazoIecarboxylic acid [5-
m/z 390 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC d 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.56 min;
-Methoxymethyl-isoxazolecarboxylic acid
m/z 421 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.67 min;
azinecarboxylic acid [5-(7-
m/z 438 [M+H]+.
py|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
,6-Dihydro-4H-pyrro|o[1,2-b]pyrazo|e LCMS (system 8): R: 1.55 min;
carboxylic acid [5-(7-isopropyI-7H- m/z 416 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC d 5)
3-y|]-amide
LCMS (system 8): R: 1.58 min;
-Bromo—pyrimidinecarboxylic acid [5-(7-
m/z 467 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.55 min;
6-Cyano-N-[5-(7-isopropy|-7H-pyrro|o[2,3-
m/z 412 .
d]pyrimidinecarbony|)-pyridiny|]-
Prep HPLC (method 5)
nicotinamide
LCMS (system 8): R: 1.58 min;
Pyrazo|o[1,5-a]pyrimidinecarboxylic acid
m/z 427 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC d 5)
-carbony|)-pyrid iny|]-am ide
3-MethyI-1H-pyrazo|o[3,4-b]pyridine LCMS (system 8): R: 1.53 min;
carboxylic acid [5-(7-isopropyI-7H- m/z 441 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS (system 8): R: 1.73 min;
7-MethyI-pyrazo|o[1,5-a]pyridinecarboxylic
m/z 440 [M+H]+.
acid [5-(7-isopropyI-7H-pyrro|o[2,3-
Prep HPLC (method 5)
d]pyrim idinecarbony|)-pyridiny|]-am ide
LCMS (system 8): R: 1.59 min;
-MethyI-oxazolecarboxylic acid [5-(7-
m/z 391 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.74 min;
Thieno[3,4-c]pyridinecarboxylic acid [5-(7- m/z 443 [M+H]+.
py|-7H-pyrro|o[2,3-d]pyrimidine Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide (Acid: WO 2002100857)
LCMS (system 8): R: 1.65 min;
2,6-DimethyI-pyrimidinecarboxylic acid [5-
m/z 416 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.62 min;
4-Cyano—pyridinecarboxylic acid [5-(7-
m/z 412 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5).
carbony|)-pyrid iny|]-am ide
-MethoxymethyImethy|-1H-pyrazoIe LCMS (system 8): R: 1.56 min;
carboxylic acid [5-(7-isopropyI-7H- m/z 434 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC d 5).
3-y|]-amide (Acid: Prep 299)
LCMS (system 8): R: 1.68 min;
m/z 444 [M+H]+.
4,5-Dichloro-1H-imidazoIecarboxylic acid
Prep HPLC (method 5).
isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
(Acid: Angewandte Chemie,
-carbony|)-pyrid iny|]-am ide
417-1418)
LCMS (system 8): R: 1.78 min;
m/z 482 [M+H]+.
4-Methoxy—8-methyI-1,7-naphthyridine
Prep HPLC (method 5)
carboxylic acid [5-(7-isopropyI-7H-
(Acid: can be ed using
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridinchemistry
outlined in
3-y|]-amide
WOO7011811)
LCMS (system 8): R: 1.63 min;
2-EthyImethyl-oxazolecarboxylic acid
m/z 419 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.57 min;
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
m/z 417 [M+H]+.
d]pyrimidinecarbony|)-pyridiny|]
Prep HPLC (method 5)
methoxy—nicotinamide
LCMS m 8): R: 1.49 min;
azo|o[3,4-b]pyridinecarboxylic acid
m/z 427 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
109 2012/051363
LCMS (system 8): R: 1.44 min;
3H-Imidazo[4,5-b]pyridinecarboxylic acid
m/z 427 [M+H]+.
isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.61 min;
m/z 457 [M+H]+.
8—Methoxy—imidazo[1 ,2-a]pyrazine
Prep HPLC (method 5)
carboxylic acid [5-(7-isopropyI-7H-
(Acid: can be made from the
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
appropriate halo derivative e.g.
3-y|]-amide
WOO7028051)
LCMS m 8): R = 1.51
N-[5-(7-|sopropy|-7H-pyrro|o[2,3-
min; m/z 387 [M+H]+.
d]pyrimidinecarbony|)-pyridiny|]-
Prep HPLC (method 5)
otinamide
LCMS (system 8): R: 1.70 min;
3-MethyI-pyridinecarboxylic acid[5-(7-
m/z 401 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.59 min;
Furo[2,3-b]pyridinecarboxylic acid [5-(7- m/z 427 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide (Acid: WO 8)
3-IsopropyI-1,2,4-triazo|o[4,3-a]pyridine LCMS (system 8): R: 1.57 min;
ylic acid [5-(7-isopropyI-7H- m/z 469 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
LCMS (system 8): R: 1.50 min;
OxazoIecarboxylic acid [5-(7-isopropyl-
m/z 377 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|)-
Prep HPLC (method 5)
PyridinyI]-amide
LCMS (system 8): R: 1.62 min;
3-Chloro-pyridinecarboxylic acid[5-(7-
m/z 421 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.44 min;
|midazo[1,2-a]pyrimidinecarboxylic acid
m/z 427 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
ony|)-pyrid iny|]-am ide
(Acid — Prep 300)
LCMS (system 8): R: 1.67 min;
-CyclopropyI-isoxazolecarboxylic acid [5-
m/z 417 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.58 min;
-Cyclopropy|-2H-pyrazoIecarboxylic acid
m/z 416 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC d 5)
-carbony|)-pyrid iny|]-am ide
4,7-DimethyI-pyrazo|o[5,1-c][1,2,4]triazine
LCMS (system 8): R: 1.69 min;
carboxylic acid [5-(7-isopropyI-7H-
m/z 456 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
Prep HPLC (method 5)
3-y|]-amide
N-[5-(7-|sopropy|-7H-pyrro|o[2,3- LCMS (system 8): R: 1.51 min;
d]pyrimidinecarbony|)-pyridiny|] m/z 401 [M+H]+.
methyl-nicotinamide Prep HPLC (method 5)
LCMS m 8): R = 1.64
1-CyclobutyI-1H-imidazoIecarboxylic acid
min; m/z 430 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
(Acid — Prep 288)
Pyrazo|o[1,5-a]pyridinecarboxylic acid [5- LCMS (system 8): R = 1.65
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine min; m/z 426 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC (method 5)
LCMS (system 8): R: 1.51 min;
o|o[1,5-a]pyridinecarboxylic acid [5-
m/z 428 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
Commercial
8-Tetrahydro-1,2,4-triazo|o[4,3-
LCMS (system 8): R: 1.60 min;
a]pyridinecarboxylic acid [5-(7-isopropyl-
m/z 431 [M+H]+.
7H-pyrro|o[2,3-d]pyrim 5-carbony|)-
Prep HPLC (method 5)
pyridiny|]-amide
Commercial
2-MethyItrifluoromethyI-oxazole LCMS (system 8): R: 1.69 min;
ylic acid [5-(7-isopropyI-7H- m/z 459 [M+H]+.
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC d 5)
3-y|]-amide
LCMS (system 8): R: 1.36 min;
1H-Pyrro|o[3,2-b]pyridinecarboxylic acid
m/z 426 [M+H]+.
[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC d 5)
-carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.57 min;
Pyrazo|o[3,4-b]pyridinecarboxylic acid [5-
m/z 427 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
azolecarboxylic acid [5-(7- LCMS (system 8): R = 1.65
isopropy|-7H-pyrro|o[2,3-d]pyrimidine min; m/z 426 [M+H]+.
carbony|)-pyrid iny|]-am ide Prep HPLC d 5)
LCMS (system 8): R: 1.58 min;
3-Methy|-isoxazo|o[5,4-b]pyridine m/z 442 [M+H]+.
carboxylic acid [5-(7-isopropyI-7H-
Prep HPLC (method 5)
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
(Acid - sis, 2009, 1858-
amide
1864)
LCMS (system 8): R: 1.45 min;
Pyridazinecarboxylic acid [5-(7-isopropyl-
m/z 388 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|)-
Prep HPLC (method 5)
Pyridinyl]—amide
LCMS (system 8): R: 1.66 min;
2,3-Dihydro-1,4-dioxino[2,3-c]pyridine
m/z 445 [M+H]+.
carboxylic acid [5-(7-isopropyI-7H-
Prep HPLC (method 5)
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin-
(Acid: )
3-y|]-amide
LCMS (system 8): R: 1.58 min;
1H-Pyrazo|o[3,4-b]pyridinecarboxylic acid m/z 427 [M+H]+.
isopropyI-7H-pyrro|o[2,3-d]pyrim idine- Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide (Acid: )
LCMS (system 8): R: 1.68 min;
-Fluoro-1H-indazoIecarboxylic acid [5-(7-
m/z 444 [M+H]+.
isopropy|-7H-pyrro|o[2,3-d]pyrimidine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
LCMS (system 8): R: 1.59 min;
2,5-Dimethyl-2H-pyrazoIecarboxylic acid
m/z 404 [M+H]+.
isopropyI-7H-pyrro|o[2,3-d]pyrim idine-
Prep HPLC (method 5)
-carbony|)-pyrid iny|]-am ide
W0 2012/137089 112
LCMS m 8): R: 1.56 min;
Thiazo|o[4,5-b]pyridinecarboxylic acid [5-
m/z 444 [M+H]+.
(7-isopropyI-7H-pyrro|o[2,3-d]pyrim idine
Prep HPLC (method 5)
carbony|)-pyrid iny|]-am ide
6-Chloro—imidazo[1,2-b]pyridazine LCMS (system 8): R: 1.68 min;
carboxylic acid [5-(7-isopropyI-7H- m/z 461 .
pyrro|o[2,3-d]pyrimidinecarbony|)-pyridin- Prep HPLC (method 5)
3-y|]-amide
Example 517: 2-(5,8-Dihydro-6H-[1,7]naphthyridiny|)-N-[5-(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidine
carbonyl)-pyridiny|]-acetamide
\/JJ\/NO \
\ N
To a DMF solution of (5,8-Dihydro-6H-[1,7]naphthyridiny|)-acetic acid hydrochloride (Preparation 235)
(34.2 mg, 0.18 mmol) were added (5-Amino-pyridiny|)-(7-isopropy|-7H-pyrroIo[2,3-d]pyrimidiny|)-
methanone (Preparation 95) (50 mg, 0.18 mmo|), HATU (136.4 mg, 0.36 mmol) and Hunig’s base (0.092
ml, 0.54 mmo|). The mixture was heated at 50°C for 20 hours and then diluted with ethyl acetate, washed
with water, brine, dried over sodium sulphate and evaporated to dryness in vacuo. The crude solid was
purified over ative TLC e|uting with 5% MeOH/EtOAc to afford the title compound as off white solid
in 26% yield, 21 mg. 1H NMR (400 MHz, DMSO-D6) 6: 1.55 (d, 6H), 2.85-2.92 (m, 4H), 3.45 (s, 2H), 3.80
(s, 2H), 5.10 (m, 1H), 7.19 (dd, 1H), 7.56 (d, 1H), 8.32 (d, 1H), 8.53 (s, 1H), 8.56 (t, 1H), 8.73 (d, 1H), 8.99
(s, 1H), 9.07 (d, 1H), 9.45 (s, 1H), 10.31 (s, 1H); LCMS m 9): R = 1.77 min; m/z 456 [M+H]+.
Example 518: 2-Hydroxy—N-[5-(7-isopropyI-7H-pyrro|o[2,3-d]pyrimidinecarbony|)-pyridiny|]pheny|-
acetamide
\ / O
N'k\ N
\ HW
N/ K HO
To stirred solution of N-[5-(7-Isopropy|-7H-pyrro|o[2,3-d]pyrimidinecarbony|)-pyridiny|]pheny|
(tetrahydro-pyranyloxy)-acetamide (60 mg, 0.12 mmol) ration 237) in e (1 mL) was
added dioxane-HCI (1 mL of a 4N solution) at 0°C. The mixture was stirred at room temperature for 2
hours. All the volatiles were removed in vacuo and the solid obtained was triturated with diethyl ether to
afford the title compound as a yellowish solid in 78% yield, 42 mg.
W0 2012/137089 113
1H NMR (400 MHz, DMSO-D6) 6: 1.55 (d, 6H), 5.08-5.12 (m, 1H), 5.19 (s, 1H), 7.30 (t, 1H), 7.37 (t, 2H),
7.53 (d, 2H), 8.56 (s, 1H), 8.63 (s, 1H), 8.76 (br s, 1H), 9.05 (s, 1H), 9.17 (br s, 1H), 9.48 (br s, 1H), 10.56
(s, 1H); LCMS (system 10): R = 2.81 min; m/z416 [M+H]+.
Example 519: N-[2-Ethoxy—5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)-pyridiny|](5-
methyltrifluoromethyl-pyrazoly|)-acetamide
NJk/N‘H _
o O FF
\ [(1 \/Me
N \ \
KN/ N
>‘Me
The title compound was prepared according to the method described for Example 1 using (5-Amino—6-
ethoxy—pyridinyl)-(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)-methanone (Preparation 242) and (5-
trifluoromethyl-pyrazoly|)-acetic acid to afford the title compound as off white solid in 50%
yield, 40 mg.
1H NMR (400 MHz, CDCI3) 6: 1.38 (t, 3H), 1.60 (d, 6H), 2.38 (s, 3H), 4.50 (q, 2H), 4.95 (s, 2H), 5.14-5.22
(m, 1H), 6.44 (s, 1H), 7.93 (s, 1H), 8.41 (s, 1H), 8.44 (s, 1H), 8.99 (s, 1H), 9.04 (s, 1H), 9.57 (s, 1H);
LCMS (system 10): R = 3.48 min; m/z 516 [M+H]+.
Example 520: N-[5-(2-Aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridiny|](4-cyano-
pheny|)-acetamide
The title compound was prepared according to the method described for Example 1 using (2-amino
tert-butyl-7H-pyrro|o[2,3-d]pyrimidiny|)-(5-amino-pyridinyl)—methanone (see ation 65) and 4-
cyano phenyl acetic acid to afford the title compound as off white solid in 58%, 23 mg.
LCMS m 10): R = 2.78 min; m/z 454 .
Example 521: 2-(4-Chloro-phenyl)—3-hydroxy—N-[5-(7-isopropyl-7H-pyrro|o[2,3-d]pyrimidinecarbonyl)—
pyridinyl]-propionamide
\ / 0
Z/ IZ
)‘Me OH
W0 2012/137089 114
2-(4-Chloro-phenyl)hydroxy—propionic acid (570 mg, 2.84 mmol) (Preparation 244) was added to a
solution of (5-Amino-pyridinyl)-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)—methanone (200 mg, 0.71
mmol) (Preparation 95) in THF (5 mL). Di-isopropyl ethylamine (0.64 mL, 3.56 mmol), EDC|.HC| (273
mg, 1.42 mmol) and HOBT (193 mg, 1.42 mmol) were added and the mixture was d at 25°C for 48
hours. The reaction mixture was quenched with saturated aqueous sodium bicarbonate solution (2 mL)
and extracted with ethyl acetate (3 x 15 mL). The combined organic layer was washed with water (5mL),
brine (5 mL), dried over sodium te and evaporated in vacuo. The crude material was purified by
preparative TLC (dichloromethane: methanol 93:7) to afford the title compound as a yellow solid in 3%
yield, 10 mg.
1H NMR (400 MHz, 6) 6: 1.55 (d, 6H), 3.58-3.61 (m, 1H), 3.88-3.91 (m, 1H), 4.04 (t, 1H), 5.07-
.13 (m, 2H), 7.41 (s, 4H), 8.47 (s, 1H), 8.50 (s, 1H), 8.72 (s, 1H), 8.99 (s, 2H), 9.43 (s, 1H), 10.65 (s, 1H);
LCMS (system 10): R = 2.99 min; m/z 464 [M+H]+.
e 522: 2-(4-Cyano-phenyl)—N-{5-[7-(1-hydroxymethyl-cyclopropyl)-7H-pyrrolo[2,3-d]pyrimidine
carbonyl]—pyridinyl}-acetam ide
(4-Cyano-phenyl)—acetic acid (6.1 mg, 0.04 mmol) was added to a solution of (5-Amino-pyridinyl)—{7-[1-
(tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-pyrrolo[2,3-d]pyrimidinyl}-methanone (15 mg, 0.04
mmol) ration 251) in THF (1 mL). Then 1-propylphosphonic acid cyclic anhydride (0.07 mL, 0.11
mmol) and triethylamine (0.02 mL, 0.13 mmol) were added and the mixture was stirred at 25°C for 4
hours. The reaction mixture was quenched with saturated aqueous sodium bicarbonate solution (2 mL)
and extracted with ethyl acetate (3 x 5 mL). The combined organic layer was washed with water (5mL),
brine (5 mL), dried over sodium sulphate and evaporated in vacuo. The crude was dissolved in methanol
(1 mL), PTSA (5 mg) was added and stirred at room temperature for 16 hours. The reaction mixture was
ed with saturated aqueous sodium bicarbonate solution (2 mL) and extracted with
dichloromethane (5 x 5 mL). The combined organic layer was washed with brine (5 mL), dried over
sodium sulphate and ated in vacuo. The crude material was purified by preparative TLC
oromethane: methanol 95:5) to afford the title compound as a white solid in 58% yield, 10 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.13-1.15 (m, 2H), 1.27-1.29 (m, 2H), 3.66 (d, 2H), 3.87 (s, 2H), 5.00 (t,
1H), 7.56 (d, 2H), 7.82 (d, 2H), 8.22 (s, 1H), 8.45 (s, 1H), 8.74 (s, 1H), 8.96 (s, 1H), 9.00 (s, 1H), 9.44 (s,
1H), 10.73 (s, 1H); LCMS (system 10): R = 2.57 min; m/z 453 [M+H]+.
The following Examples were prepared ing to the method described above for Example 522
starting from (5-Amino-pyridinyl)-[7-(3-methyl-oxetanyl)-7H-pyrrolo[2,3-d]pyrimidinyl]—methanone
(Preparation 251) and the appropriate acids.
W0 2012/137089 115
2-(5-Chloro-pyridiny|)-N-{5-[7-(1-hydroxymethy|-
cyclopropyl)-7H-pyrro|o[2,3-d]pyrimidinecarbony|]- LCMS m 10): Rt =
pyridiny|}-acetamide 2.53 min; m/z 463 [M+H]+
N-{5-[7-(1-HydroxymethyI-cyclopropyl)-7H-pyrro|o[2,3-
LCMS (system 10): R =
d]pyrimidinecarbony|]-pyridiny|}(4-trif|uoromethyl-
2.89 min; m/z 496 [M+H]+.
pheny|)-acetamide
7-(1-HydroxymethyI-cyclopropyl)-7H-pyrro|o[2,3-
LCMS (system 9): R = 2.71
d]pyrimidinecarbony|]-pyridiny|}(3-trif|uoromethyl-
min; m/z 486 [M+H]+.
pyrazoIy|)-acetamide
Example 526: 1-(3-CyclopropyI-1'-methy|-1'H-[1,4']bipyrazo|y|y|)[5-(7-isopropy|-7H-pyrro|o[2,3-
d]pyrim idinecarbony|)-pyridiny|]-urea
Phenyl chloroformate (0.03 mL, 0.24 mmol) was added slowly to a solution of 3-CyclopropyI-1'-methy|-
1'H-[1,4']bipyrazo|y|y|amine (Preparation 297, 40 mg, 0.19 mmol) and ne (0.03 mL) in THF (2 mL)
at 0°C and the mixture was stirred at room temperature for 4 hours. A on of (5-Amino-pyridiny|)-(7-
isopropyI-7H-pyrro|o[2,3-d]pyrimidiny|)-methanone (Preparation 95) (55.4 mg, 0.19 mmol) in DMF (1
mL) and was then added and the reaction mixture heated at 100°C for 16 hours. The mixture was cooled
and diluted with ethyl e (15 mL), and washed with aqueous saturated NaHCO3 solution (2 x 10 mL),
water (10 mL), brine (10 mL), dried over sodium te and evaporated to dryness in vacuo. The crude
material was purified over preparative TLC plate (e|uting with 5% methanol in DCM) to afford the title
compound as off white solid in 15% yield, 15 mg. 1H NMR (400 MHz, DMSO-d6) 6: 0.64-0.65 (m, 2H),
0.85-0.88 (m, 2H), 1.56 (d, 6H), 1.82-1.85 (m, 1H), 3.89 (s, 3H), 5.09-5.12 (m, 1H), 6.10 (s, 1H), 7.66 (s,
1H), 8.05 (s, 1H), 8.36 (s, 1H), 8.52 (s, 1H), 8.61 (br s, 1H), 8.65 (s, 1H), 8.78 (s, 1H), 8.99 (s, 1H), 9.45
(s, 2H); LCMS (system 10): R = 2.75 min; m/z 511 [M+H]+.
Example 527: 1-(3-tert-ButyI-1'-methy|-1'H-[1,4']bipyrazonIyl)[5-(7-isopropy|-7H-pyrro|o[2,3-
d]pyrimidinecarbony|)-pyridiny|]-urea
The title compound was prepared according to the method described for Example 526 using (5-Amino-
pyridiny|)-(7-isopropy|-7H-pyrro|o[2,3-d]pyrimidiny|)-methanone (Preparation 95) and 3-tert-ButyI-1'-
methyl-1'H-[1,4']bipyrazonIylamine (Preparation 296) to afford the title compound as yellow solid in
% yield, 17 mg.LCMS (system 10): R = 3.01 min; m/z 527 [M+H]+.
W0 2012/137089 116
The following Examples were prepared according to the method described above for Example 1, starting
from (2-aminotert—butyl-7H—pyrrolo[2,3-d]pyrimidinyl)(5-aminopyridinyl)methanone (Preparation
65) and the appropriate acids.
2-Am inotert-butyl-7H-pyrrolo[2,3-d]pyrim idine
LCMS (system 10): Rt = 2.79
carbonyl)-pyridinyl]—2-(1H-benzoimidazolyl)—
min; m/z 469 [M+H]+.
acetamide
LCMS (system 10): R = 2.94
N-[5-(2-Aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidine min; m/z 508 [M+H]+
carbonyl)-pyrid inyl]—2-(5-bromo-pyridinyl)-acetam ide
LCMS (system 10): R = 2.78
min; m/z 455 [M+H]+ (acid
N-[5-(2-Am inotert-butyl-7H-pyrrolo[2,3-d]pyrim idine
can be ed by
carbonyl)-pyrid inyl]—2-(5-cyano-pyridinyl)-acetam ide
cyanation of an appropriate
bromo derivative
The following Examples were prepared according to the method described for Example 522 using (5-
Amino-pyridinyl)—{7-[(S)—1-methyl(tetrahydro-pyranyloxy)—ethyl]—7H-pyrrolo[2,3-d]pyrimidinyl}-
one (Preparation 33) and the appropriate acid.
2-(4-Difluoromethoxy—phenyl)-N-{5-[7-((S)
LCMS (system 9): R = 2.80 min;
hydroxymethyl-ethyl)-7H-pyrrolo[2,3-
m/z 482 [M+H]+
d]pyrim idinecarbonyl]-pyridinyl}-acetam ide
N-{5-[7-((S)Hydroxy—1-methyl-ethyl)—7H-
LCMS (system 9): Rt = 2.58 min;
pyrrolo[2,3-d]pyrim 5-carbonyl]-pyridinyl}
m/z 456 [M+H]+
lyl-acetam ide
2-(1H-Benzoimidazolyl)-N-{5-[7-((S)hydroxy-
LCMS (system 10): Rt = 2.42 min;
1-methyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine
m/z 456 [M+H]+.
carbonyl]-pyridinyl}-acetam ide
N-{5-[7-((S)Hydroxy—1-methyl-ethyl)—7H-
LCMS (system 9): Rt = 2.99 min;
pyrrolo[2,3-d]pyrim idinecarbonyl]-pyridinyl}
m/z 500 [M+H]+
(4-trifluoromethoxy—phenyl)—acetam ide
-Difluoro-phenyl)-N-{5-[7-((S)hydroxy—1 -
LCMS (system 9): Rt = 2.75 min;
methyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine
m/z 452 [M+H]+
carbonyl]-pyridinyl}-acetam ide
The following Examples were prepared according to the Method described for d]pyrimidinyl]methanone
ration 186) and the appropriate acid.
W0 2012/137089 117
LCMS (system 10): R =
2-(5-Chloro-pyridiny|)-N-{5-[7-(2-m ethoxy—1 ,1-
2.92 min; m/z 479
dimethyl-ethyl)-7H-pyrro|o[2,3-d]pyrim idinecarbony|]-
[|V|+H]+
pyridinyI}-acetam ide
LCMS (system 10): R =
2-(5-FIuoro-pyridiny|)-N-{5-[7-(2-methoxy-1 ,1- 2.79 min; m/z 463
dimethyl-ethyl)-7H-pyrro|o[2,3-d]pyrimidinecarbony|]- [M+H]+.
pyridinyI}-acetamide
LCMS m 10): R =
2-(3-CyclopropyI-pyrazoly|)-N-{5-[7-(2-methoxy—1 ,1- 2.89 min; m/z 474
dimethyl-ethyl)-7H-pyrro|o[2,3-d]pyrimidinecarbony|]- [M+H]+.
pyridinyI}-acetamide
LCMS (system 10): R =
2-(4-FIuoro-pheny|)-N-{5-[7-(2-methoxy—1,1-dimethy|- 3.03 min; m/z 462
-7H-pyrro|o[2,3-d]pyrimidinecarbony|]-pyridin [M+H]+.
y|}-acetamide
e 540: N-[5—(7-tert—Buty|-7H-pyrro|o[2,3-d]pyrimidinecarbony|)-pyridiny|](4-f|uoro-pheny|)-
N-methyl-acetamide
N|\\
)PMe
Me Me
To a stirred solution of N-[5—(7-tert—Buty|-7H-pyrro|o[2,3-d]pyrimidinecarbony|)-pyridiny|](4-f|uoro-
pheny|)-acetamide (Example 546, 115 mg, 0.27 mmol) in anhydrous THF (4.5 mL), was added NaH (60%
in paraffin OH, 10.7 mg, 0.27 mmol) at 0°C under en and the resulting mixture stirred for 10 min. Mel
(0.017 mL, 0.27 mmol) was then added and the reaction mixture was stirred at room temperature for 1
hour. Aqueous saturated ammonium chloride (5 mL) was added and the mixture extracted with ethyl
e (2 x 10 mL). The organic phase was washed with water (10 mL), brine (10 mL), dried over sodium
sulphate and evaporated to dryness in vacuo. The crude al was purified via preparative TLC
(e|uting with 5% methanol in DCM) to afford the title compound as off white solid in 46% yield, 55 mg. 1H
NMR (400 MHz, DMSO-D6) 6: 1.79 (s, 9H), 3.28 (s, 3H), 3.58 (br, 2H), 7.08-7.20 (m, 4H), 8.20 (s, 1H),
8.32 (s, 1H), 8.83 (s, 1H), 8.97 (br, 1H), 9.01 (s, 1H), 9.50 (s, 1H); LCMS (system 10): R = 3.08 min; m/z
446 [M+H]+.
Example 541: [5—(7-tert—ButyI-7H-pyrro|o[2,3-d]pyrimidinecarbony|)-pyridiny|]-carbamic acid tert-
butyl ester
W0 2012/137089 118
N N
Me Me
To a d solution of (5-Amino-pyridinyl)—(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)—methanone
(Preparation 31) (200 mg, 0.68 mmol) in DCM (4 mL) was added boc-anhydride (0.155 mL, 0.68 mmol)
and Hunig’s base (0.24 mL, 1.36 mmol) and the on mixture was stirred at room ature for 16
hours. It was diluted with DCM (15 mL) and washed with water (2 x 10 mL), brine (10 mL), dried over
sodium sulphate and evaporated to dryness in vacuo. The crude al was purified by column
chromatography on silica gel (Methanol:DCM 2:98) to afford the title compound as light brown gum in
41% yield, 110 mg. LCMS (system 10): R = 3.16 min; m/z 396 [M+H]+.
Example 542: [5-(7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridinyl]—methyl-carbamic acid
tert-butyl ester
The title compound was prepared according to the method described for Example 540 using [5-(7-tert-
Butyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)-pyridinyl]-carbamic acid tert-butyl ester (Example 541) to
afford the title compound as yellow solid in 79% yield, 90 mg. LCMS (system 10): R = 3.83 min; m/z 410
[M+H]+.
Example 543: N-[5-(7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridinyl]—2-(5-chloro-pyridin-
2-yl)-N-methyl-acetamide
N \
/ CI
o /\
“W \ kN/
N)r Me Mil/'9
The title compound was prepared according to the method described for e 1 using (7-tert-butyl-
7H-pyrrolo[2,3-d]pyrimidinyl)—(5-methylamino-pyridinyl)—methanone (Preparation 187) and (5-chloro—
pyridinyl)-acetic ee Preparation 90) to afford the title compound as off white solid in 21% yield,
mg. 1H NMR (400 MHz, DMSO-D6) 6: 1.78 (s, 9H), 3.32 (s, 3H), 3.77 (brs, 2H), 7.26 (br, 1H), 7.83 (br,
1H), 8.21 (s, 1H), 8.32 (s, 1H), 8.48 (s, 1H), 8.85 (s, 1H), 8.97 (s, 1H), 9.02 (s, 1H), 9.51 (s, 1H); LCMS
(system 10): R = 3.29 min; m/z 463.2 [M+H]+.
Example 544: N-[5-(7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridinyl]—2-(4-chloro-phenyl)-
N-methyl-acetamide
W0 2012/137089 119
The title compound was prepared according to the method described for Example 1 using (7-tert—Butyl-
7H-pyrrolo[2,3-d]pyrimidinyl)-(5-methylamino-pyridinyl)-methanone ration 187) and (4-chlorophenyl
)-acetic acid to afford the title compound as off white solid in 45% yield, 21 mg.
LCMS m 10): R = 3.28 min; m/z 462 [M+H]+.
The following Examples were prepared according to the method described above for Example 1, starting
from (5-aminopyridinyl)(7-tert—butyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 31) and
the appropriate acids.
LCMS (system 10): R = 3.18
N-[5-(7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidine-5—
min; m/z 448 [M+H]+.
carbonyl)-pyridinyl](4-chloro-phenyl)—
acetam ide
LCMS m 10): R = 3.08
N-[5-(7-tert-Butyl-7H-pyrrolo[2,3-d]pyrImIdIne
min; m/z 432 [M+H]+.
carbonyl)-pyridinyl](4-fluoro-phenyl)—
acetam ide
The following Examples were prepared according to the method described for Example 1 starting from [2-
7-(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]-(5-amino-pyridinyl)-methanone
(Preparation 261) and the appropriate acid.
LCMS (system 10): R = 2.85
N-{5—[2-Amino(2-m ethoxy-1 ,1-dimethyl-ethyl)—7H-
min; m/z 494 [M+H]+.
pyrrolo[2,3-d]pyrim idinecarbonyl]-pyridinyl}
(Acid: Prep 90)
(5-chloro-pyridinyl)-acetamide
LCMS (system 10): R = 2.77
N-{5—[2-Amino(2-m ethoxy-1 ,1-dimethyl-ethyl)—7H-
min; m/z 478 .
o[2,3-d]pyrim idinecarbonyl]-pyridinyl}
(Acid Prep 92)
oro-pyridinyl)-acetam ide
LCMS (system 10): R = 3.02
N-{5—[2-Amino(2-m ethoxy-1 ,1-dimethyl-ethyl)—7H-
min; m/z 517 [M+H]+.
pyrrolo[2,3-d]pyrim idinecarbonyl]-pyridinyl}
(Acid Prep 85)
(3-trifluoromethyl-pyrazoly|)-acetamide
LCMS (system 10): R = 2.93
N-{5—[2-Amino(2-m ethoxy-1 ,1-dimethyl-ethyl)—7H-
min; m/z 518 [M+H]+.
o[2,3-d]pyrim idinecarbonyl]-pyridinyl}
(Acid Prep 81)
(4-trifluoromethyl-[1 ,2,3]triazoly|)-acetamide
LCMS (system 10): R = 3.01
N-{5—[2-Amino(2-m -1 ,1-dimethyl-ethyl)—7H-
min; m/z 517 [M+H]+.
pyrrolo[2,3-d]pyrim idinecarbonyl]-pyridinyl}
(Acid Prep 85)
(4-trifluoromethyl-pyrazoly|)-acetamide
LCMS (system 10): R = 2.69
N-{5—[2-Amino(2-m ethoxy-1 ,1-dimethyl-ethyl)—7H-
min; m/z 490 [M+H]+.
pyrrolo[2,3-d]pyrim idinecarbonyl]-pyridinyl}
(Acid Prep 83)
(4-cyclopropyl-[1,2,3]triazoly|)-acetamide
W0 2012/137089 120
The following Examples were prepared according to the method described for Example 522 using (5-
Amino-pyridiny|)-{7-[(S)methy|(tetrahydro-pyrany|oxy)-ethy|]-7H-pyrro|o[2,3-d]pyrimidiny|}-
methanone (Preparation 49) and the appropriate acid.
N-{5-[2-Amino(2-hydroxy-1 ethyI-ethy|)- LCMS (system 10): R = 2.67 min;
7H-pyrro|o[2,3-d]pyrim idinecarbony|]-pyridin m/z 485 [M+H]+.
y|}indazoIyI-acetam ide
LCMS (system 10): R = 2.52 min;
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)-
m/z 513 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|]-pyridin
y|}(4-oxo-4H-q uinazolinyl)—acetam ide
LCMS (system 9): R = 2.07 min;
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- m/z 482 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|]-pyridin
y|}(3,5-difluoro-pyridiny|)-acetamide Acid Prep 265)
LCMS (system 9): R = 1.36 min;
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- m/z 460 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|]-pyridin
y|}(5-methyl-pyridiny|)-acetam ide Acid Prep 274
N-(5-{[2-amino(2-hydroxy-1,1-dimethylethy|)-7H-
LCMS (system 9): R = 2.19 min;
pyrro|o[2,3-d]pyrimidiny|]carbony|}pyrid iny|)-
m/z 552 [M+H]+.
m ethy|(methy|su|fony|)am ino]pheny|}-
Acid Prep 271
acetam ide
LCMS (system 9): R = 1.42 min;
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)-
m/z 485 [M+H]+.
ro|o[2,3-d]pyrim 5-carbony|]-pyridin
y|}(1H-benzoimidazoIy|)-acetamide
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- LCMS (system 9): R = 2.18 min;
7H-pyrro|o[2,3-d]pyrim idinecarbony|]-pyridin m/z 530 [M+H]+.
y|}(2-oxotrif|uorom ethy|-2H-pyridiny|)-
acetam ide
LCMS (system 9): R = 2.23 min;
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- m/z 498 [M+H]+.
7H-pyrro|o[2,3-d]pyrim 5-carbony|]-pyridin
y|}(5-ch|orofluoro-pyridiny|)-acetamide
LCMS (system 10): R = 2.64 min;
m/z 490 [M+H]+ (acid can be
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- prepared from the riate
7H-pyrro|o[2,3-d]pyrim 5-carbony|]-pyridin bromide using the method in
y|}(5-ethoxy-pyridiny|)-acetamide /114271)
LCMS (system 9): R = 2.40 min;
2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)-
m/z 514 [M+H]+.
7H-pyrro|o[2,3-d]pyrim idinecarbony|]-pyridin
y|}(5-trif|uoromethyl-pyrid iny|)-acetam ide
LCMS (system 9): R = 2.39 min;
N-{5-[2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- m/z 503 [M+H]+.
7H-pyrro|o[2,3-d]pyrimidinecarbony|]-pyridin
Acid prep 85
y|}(4-trif|uoromethyl-pyrazoly|)-acetamide
2-Amino(2-hydroxy-1 ,1-dimethyI-ethy|)- LCMS (system 9) R = 4.47 min (12
7H-p rro|o[2,3-d]p rimidinecarbon |]-p ridin min run ; m/z 538 [M+H]+.
W0 2012/137089 121
-yl}(4-methanesulfonylamino-phenyl)—acetamide—
The following Examples were prepared according to the method described for Examples 73-87 using (5-
pyridinyl)-{7-[(S)methyl(tetrahydro-pyranyloxy)—ethyl]—7H-pyrrolo[2,3-d]pyrimidinyl}-
methanone (Preparation 49) and the appropriate acid.
N-{5-[2-Amino(2-hydroxy-1,1-dimethyl-ethyl)-7H- LCMS (system 9): R = 2.56
pyrrolo[2,3-d]pyrimidinecarbonyl]—pyridinyl}(4- min; m/z 511 [M+H]+.
difluoromethoxy-phenyl)-acetamide
LCMS (system 9): R = 2.74
N-{5-[2-Am ino(2-hydroxy-1 ,1-dimethyl-ethyl)-7H-
min; m/z 529 [M+H]+.
pyrrolo[2,3-d]pyrimidinecarbonyl]—pyridinyl}(4-
trifluoromethoxy-phenyl)-acetamide
N-{5-[2-Amino(2-hydroxy-1,1-dimethyl-ethyl)-7H- LCMS (system 9): R = 2.50
pyrrolo[2,3-d]pyrimidinecarbonyl]—pyridinyl}(3,4- min; m/z 481 [m+H]+_
difluoro-phenyl)—acetamide
LCMS (system 10): R =
2.35 min; m/z 481 [M+H]+
2-Amino(2-hydroxy-1,1-dimethyl-ethyl)-7H- (acid can be ed by
pyrrolo[2,3-d]pyrimidinecarbonyl]—pyridinyl}(5- oxidation of the appropriate
chloro-pyrimidinyl)-acetamide aldehyde WOO4110453)
Example 570: 2-(4-Cyano-phenyl)-N-[5-(7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridinyl]-acetamide
\ / o
“t \ N
\ H
N N
The title compound was prepared according to the method described for Example 343 using 2-(4-Cyano-
phenyl)-N-{5-[7-(2-trimethylsilanyl-ethoxymethyl)-7H-pyrrolo[2,3-d]pyrimidinecarbonyl]-pyridinyl}-
acetamide (Preparation 277) to afford the title compound as a white solid in 99% yield, 1H NMR (400
MHz, DMSO-d6) 6: 3.87 (s, 2H), 7.55 (d, 2H), 7.81 (d, 2H), 8.36 (s, 1H), 8.47 (s, 1H), 8.72 (s, 1H), 8.97
(m, 2H), 9.47 (s, 1H), 10.66 (s, 1H), 13.14 (s, 1H); LCMS (System 10): R = 2.48 min; m/z 383 [M+H]+.
The following es were prepared according to the method described above for e 1, starting
from (5-Amino-pyridinyl)-[7-(3-methyl-oxetanyl)—7H-pyrrolo[2,3-d]pyrimidinyl]-methanone
(Preparation 222) and the appropriate acid.
N-{5-[7-(3-Methyl-oxetanyl)—7H-pyrrolo[2,3-d]pyrImIdIne-
LCMS m 10): Rt: 302
-carbonyl]-pyridinyl}(4-trifluoromethyl-pheny|)- . +
mIn, m/z 496 [M+H]_
acetamide
W0 2012/137089 122
N-{5-[7-(3-Methyl-oxetanyl)—7H-pyrrolo[2,3-d]pyrim idine-
LCMS (System 10): Rt = 2.98
-carbonyl]-pyridinyl}(3-trifluoromethyl-pheny|)-
min; m/z 496 [M+H]+.
acetamide
2-(3-F|uorotrifluoromethyl-phenyl)-N-{5-[7-(3-methyl-
LCMS (System 10): Rt = 3.01
oxetanyl)—7H-pyrrolo[2,3-d]pyrimidinecarbonyl]-
min; m/z 514 [M+H]+.
pyridinyl}-acetamide
N-{5-[7-(3-Methyl-oxetanyl)—7H-pyrrolo[2,3-d]pyrimidine- LCMS (System 10): Rt = 3.00
-carbonyl]-pyridinyl}trifluoromethyl-benzamide min; m/z 482 [M+H]+ .
N-{5-[7-(3-Methyl-oxetanyl)—7H-pyrrolo[2,3-d]pyrimidine- LCMS (System 10): Rt = 3.05
-carbonyl]-pyridinyl}trifluoromethoxy—benzamide min; m/z 498 [M+H]+
Example 576: N-{5-[2-Amino(2-hydroxy-1,1-dimethyl-ethyl)—7H-pyrrolo[2,3-d]pyrimidinecarbonyl]-
pyridinyl}(4-cyclopropyl-pyrazoly|)-acetamide
The title nd was prepared according to the method described for Example 1 using {2-amino
[1 ,1-dimethy|(tetrahydro-pyranyloxy)—ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}-(5-amino-pyridinyl)—
methanone (Preparation 49) and lopropyl-pyrazoly|)-acetic acid to afford the title nd as
a white solid in 72 % yield, 16.5 mg.
LCMS (System 10): R = 2.62 min; m/z 475 [M+H]+.
Example 577: N-[5-(2-Aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridinyl](4-
cyclopropyl-pyrazoly|)-acetamide
The title compound was prepared ing to the method described for Example 1 using (2-Amino—7-
tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)—(5-amino-pyridinyl)—methanone (see Preparation 65) and (4-
cyclopropyl-pyrazoly|)-acetic acid to afford the title compound as a pale yellow solid in 16 % yield, 12
mg.LCMS m 10): R = 2.80 min; m/z 459 [M+H]+.
Example 578: N-[5-(2-Aminobicyclo[1.1.1]penty|-7H-pyrro|o[2,3-d]pyrimidinecarbonyl)—pyridin
yl](5-fluoro-pyridiny|)-acetamide
\ / O
N \ \ H
JL \ /
H2N N N
W0 2012/137089 123
To a solution of (5-amino-pyridinyl)-[7-bicyclo[1.1.1]penty|(4-methoxy-benzylamino)-7H-
pyrrolo[2,3-d]pyrimidinyl]-methanone ration 287) (50 mg, 0.11 mmol) in THF (5 mL) at room
temperature was added 5-fluoro neyl acetic acid (27 mg, 0.17 mmol), TEA (0.08 mL, 0.56 mmol)
and 1-propylphosphonic acid cyclic ide (50% solution in EtOAc, 0.20 mL, 0.34 mmol). The
resulting mixture was before stirred for 18 hours. The mixture was trated under reduced pressure
and the residue was partitioned between saturated sodium bicarbonate solution (10 mL) and ethyl acetate
(25 mL). The organic phase was dried over sodium sulphate and concentrated under reduced pressure.
TFA (1.5 mL) was added and the resulting mixture stirred at room ature for 18 hours. The mixture
was concentrated under d pressure and the residue partitioned between saturated sodium
bicarbonate (10 mL) and ethyl acetate (30 mL). The organic phase was dried over sodium sulphate,
concentrated under reduced pressure ed by Preparative TLC (MeOH:DCM 5: 95) to afford the title
compound as off white solid in 31% yield, 16 mg. 1H NMR (400 MHz, DMSO-d6) 6: 2.32 (s, 6H), 2.66 (s,
1H), 3.94 (s, 2H), 6.57 (s, 2H), 7.49 (m, 1H), 7.69-7.73 (m, 2H), 8.38 (s, 1H), 8.50 (d, 1H), 8.65 (d, 1H),
8.92 (s, 1H), 8.96 (d, 1H), 10.67 (s, 1H); LCMS (System 10): R = 2.85 min; m/z 458 .
The following Examples were prepared according to the method described above for Example 578
starting from ((5-Amino-pyridinyl)-[7-bicyclo[1.1.1]pent—1-y|(4-methoxy-benzylamino)-7H-pyrrolo[2,3-
d]pyrimidinyl]-methanone (Preparation 287) and the appropriate acid.
N-[5-(2-Aminobicyclo[1.1.1]pentyl-7H-pyrrolo[2,3-
LCMS (System 10): Rt = 2.87
d]pyrimidinecarbonyl)—pyridinyl](3-
min; m/z 517 [M+H]+.
methanesulfonyl-phenyl)-acetamide
LCMS (System 10): Rt = 2.92
N-[5-(2-Aminobicyclo[1.1.1]pentyl-7H-pyrrolo[2,3-
min; m/z 497 [M+H]+.
d]pyrimidinecarbonyl)—pyridinyl](3-
trifluoromethyl-pyrazoly|)-acetamide
N-[5-(2-Aminobicyclo[1.1.1]pentyl-7H-pyrrolo[2,3-
LCMS (System 10): Rt = 2.89
d]pyrimidinecarbonyl)—pyridinyl](4-
min; m/z 498 [M+H]+.
trifluoromethyl-[1,2,3]triazoly|)-acetamide
(Acid : Prep 81)
LCMS (System 10): Rt = 2.94
N-[5-(2-Aminobicyclo[1.1.1]pentyl-7H-pyrrolo[2,3-
min; m/z 464 [M+H]+.
d]pyrimidinecarbonyl)-pyridinyl](4-cyano-
phenyl)-acetamide
LCMS m 9) : Rt = 6.28
2-Aminobicyclo[1.1.1]pentyl-7H-pyrrolo[2,3- min; m/z 474 [M+H]+.
d]pyrimidinecarbonyl)—pyridinyl](5-chloro-pyridin- (ACid Prep 90)
2-yl)-acetamide
Examples 584-5923 illustrate compounds of general formula :
that fall into general formula (I) by virtue of pyridone tautomerism.
584 2-(4-cyanophenyl)-N-(2-oxo{[7-(propanyl)—7H-pyrrolo[2,3- LCMS Rt = 5.25 min;
d]pyrimidinyl]carbonyl}-1,2-dihydropyridinyl)acetamide
m/z 441 [M+H]+
585 N-(2-oxo{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrim idin
y|]carbonyl}-1,2-dihydropyridinyl)—2-[2-
LCMS Rt = 4.04 min;
(trifluoromethoxy)phenoxy]acetamide
m/z 516 [M+H]+
586 2-(2-cyclopropyl-1 ,3-oxazolyl)-N-(2-oxo{[7-(propanyl)—7H- LCMS Rt = 3.39 min;
o[2,3-d]pyrimidinyl]carbonyl}-1,2-dihydropyridin
m/z 447 [M+H]+
l acetamide
587 2-[5-methyl(trifluoromethyl)-1H-pyrazolyl]-N-(2-oxo{[7- LCMS Rt = 3.51 min;
nyl)—7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}-1,2-
m/z 433 [M+H]+
dih drop ridin lacetamide
588 N-(2-oxo{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrim idin LCMS Rt = 3.51 min;
y|]carbonyl}-1,2-dihydropyridinyl)—2-(quinolinyl)acetamide
m/z 435 [M+H]+
589 2-(4-chlorophenyl)-N-(2-oxo{[7-(propanyl)—7H-pyrrolo[2,3-
d]pyrimidinyl]carbonyl}-1,2-dihydropyridinyl)acetamide
LCMS Rt = 3.82 min;
m/z 450 [M+H]+
590 2-(5-chloropyridinyl)-N-(2-oxo{[7-(propanyl)—7H- LCMS Rt = 3.22 min;
o[2,3-d]pyrimidinyl]carbonyl}-1,2-dihydropyridin
m/z 451 [M+H]+
l acetamide
591 N-(2-oxo{[7-(propanyl)-7H-pyrrolo[2,3-d]pyrimidin LCMS Rt = 3.31 min;
y|]carbonyl}-1 ,2-dihydropyridinyl)[4-(trifluoromethyl)—1H-
m/z 475 [M+H]+
1,2,3-triazol |]acetamide
592 [2-Amino(2-hydroxy-1,1-dimethylethyl)—7H-pyrrolo[2,3-
d]pyrimidinyl]carbonyl}oxo—pyridinyl)—2-(5-chloropyridin
l acetamide
593 N-[5-({7-[(1S)—2-Hydroxy—1-methylethyl]—7H-pyrrolo[2,3- m/Z 500, RT 4.94
d]pyrimidinyl}carbonyl)—2-oxo—pyridinyl]—2-[4-
trifluorometh Iphen |]acetamide
Preparation 1: 7-[(1S)—2-{[tert—Butyl(dimethyl)silyl]oxy}methylethyl]—4-chloro-7H-pyrrolo[2,3-
d]pyrimidine
(S)—2-tert—Butyldimethylsilyloxy—1-methylethylamine (120 g, 636 mmol) was added to (4,6-
dichloropyrimidinyl)acetaldehyde (52.8 g, 276 mmol) in ethanol (500 mL). The mixture was heated to
W0 2012/137089 125
reflux for 45 minutes. The reaction mixture was evaporated in vacuo then the residue was diluted with
water (400 mL) and ted with ethyl acetate (600 mL). The organic extract was evaporated in vacuo
and the crude material was purified by column tography on silica gel (gradient of pentane:EtOAc
90:10 to 80:20) to afford the title compound as a yellow liquid in 67% yield, 60.4 g.
1H NMR (400 MHz, CDCI3) 5: -0.92 (s, 6H), 0.80 (s, 9H), 1.58 (d, 3H), 3.86 (m, 2H), 5.04 (m, 1H), 6.59
(d, 1H), 7.40 (d, 1H), 8.60 (s, 1H).
Preparation 2: 7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloro-7H-pyrrolo[2,3-d]pyrimidine
The title compound was prepared ing to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde and 1-{[tert-butyl(dimethyl)silyl]oxy}methylpropanamine to afford
the title nd as a yellow oil in 38% yield, 377 mg.
1H NMR (400 MHz, CDCI3) 6: 0.00 (s, 6H), 0.94 (s, 9H), 1.97 (s, 6H), 4.28 (s, 2H), 6.78 (d, 1H), 7.66 (d,
1H), 8.83 (s, 1H).
Preparation 3: (2R)—2-(4-Chloro-7H-pyrrolo[2,3-d]pyrimidinyl)propano|
”C ' \
\N N
Ho\)'"lVIe
The title compound was prepared according to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde and (R)—2-aminopropanol to afford the title compound as a yellow
solid in 100% yield, 11.12 g.
LCMS (system 2): R = 0.89 min; m/z 212 [M+H]+.
Preparation DL3: (R,S) 7-[2-{[tert-Butyl(dimethyl)silyl]oxy}methylethyl]-4—chloro-7H-pyrrolo[2,3-
d]pyrimidine
W0 2012/137089 126
The title compound was prepared according to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde and 2-tert-butyldimethylsilyloxy—1-methylethylamine to afford the title
compound as an orange oil in 77% yield, 4.08 g.
1H NMR (400 MHz, CDCI3) 6: -0.90 (s, 6H), 0.82 (s, 9H), 1.58 (d, 3H), 3.84 (m, 2H), 5.05 (m, 1H), 6.59
(d, 1H), 7.42 (d, 1H), 8.60 (s, 1H).
Preparation 4: (2S)—2-(4-Chloro-7H-pyrrolo[2,3-d]pyrimidinyl)propano|
”C I \
\N N
Ho\)‘lVIe
The title compound was prepared according to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde and (S)—2-amino—1-propanol to afford the title nd as a cream
solid in 98% yield, 10.9 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.42 (d, 3H), 3.72 (m, 2H), 4.89 (m, 1H), 6.63 (d, 1H), 7.83 (d, 1H),
8.59 (s, 1H).
Preparation 5: 7-tert-Butylchloro-7H-pyrrolo[2,3-d]pyrimidine
k\N |
)VMe
Me Me
The title compound was ed according to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde and tert—butylamine to afford the title compound as a yellow liquid in
77% yield, 1.61 g.
1H NMR (400 MHz, 6) 6: 1.75 (s, 9H), 6.60 (d, 1H), 7.79 (d, 1H), 8.63 (s, 1H).
Preparation 6: 4-Chloro—7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-
midine
”C I \
\N N
2,3-Dihydropyran (25.0 mL, 270 mmol) was added to (2R)—2-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin
yl)propanol (11.00 g, 51.97 mmol) (see Preparation 3) and pyridinium toluenesulphonate (3.92 g,
W0 2012/137089 127
.6 mmol) in DCM (150 mL). The reaction mixture was washed with water (200 mL) and the aqueous
phase was extracted with DCM (2 x 150 mL). The combined c phases were dried over magnesium
sulfate and evaporated in vacuo. The crude material was ed by column chromatography on silica gel
(gradient of heptane:EtOAc 100:0 to 70:30) to afford the title nd as a yellow oil in 100% yield,
.65 g.
LCMS m 2): R = 1.30 min; m/z 296 [M+H]+.
Preparation 7: 4-Chloro[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidine
”C I \
\N N
The title compound was prepared according to the method described for Preparation 6 using (2S)—2-(4-
-7H-pyrrolo[2,3-d]pyrimidinyl)propano| (see Preparation 4) to afford the title compound as a
yellow oil in 87% yield, 11.5 g.
1H NMR (400 MHz, CDCI3) 6: 1.43-1.71 (m, 9H), 3.42 (m, 1H), 3.57 (m, 1H), 3.69 (m, 1H), 4.00 (m, 1H),
4.52 (m, 1H), 5.19 (m, 1H), 6.60 (d, 1H), 7.44 (d, 1H), 8.61 (s, 1H).
Preparation 8: 7-[(1S)—2-{[tert—Butyl(dimethyl)silyl]oxy}methylethyl]-7H-pyrrolo[2,3-d]pyrimidine
Palladium (10% on carbon, 18 g) was added to 7-[(1S)—2-{[tert—butyl(dimethyl)silyl]oxy}methylethyl]
chloro-7H-pyrrolo[2,3-d]pyrimidine (182 g, 558 mmol) (see Preparation 1) in ethanol (900 mL) and
concentrated ammonia solution (100 mL) and hydrogenated (60 psi, 20°C) for 18 hours. The reaction
mixture was filtered through ArbocelTM and the filtrate was evaporated in vacuo. Diethyl ether (300 mL)
was added to the residue and the mixture was filtered. The filtrate was evaporated in vacuo to afford the
title compound as an orange oil in 94% yield, 162.7 g.
1H NMR (400 MHz, CDCI3) 6: -0.90 (s, 3H), 0.80 (s, 9H), 1.58 (d, 3H), 3.86 (m, 2H), 5.06 (m, 1H), 6.53
(d, 1H), 7.40 (d, 1H), 8.83 (s, 1H), 8.96 (s, 1H).
W0 37089 128
Preparation 9: 7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidine
The title nd was prepared according to the method described for Preparation 8 using 7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloro-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 2) to
afford the title compound as a white solid in 97% yield, 327 mg.
1H NMR (400 MHz, CDCI3) 6: 0.01 (s, 6H), 0.94 (s, 9H), 2.02 (s, 6H), 4.30 (s, 2H), 6.92 (d, 1H), 7.84 (d,
1H), 9.13 (s, 1H), 9.21 (s,1H).
Preparation 10: (R,S) 7-(2-{[tert-Butyl(dimethyl)silyl]oxy}methylethyl)-7H-pyrrolo[2,3-d]pyrimidine
The title compound was prepared according to the method described for Preparation 8 using (R,S) 7-(2-
{[tert—butyl(dimethyl)silyl]oxy}methylethyl)chloro-7H-pyrrolo[2,3-d]pyrimidine (see Preparation DL3)
to afford the title compound as a brown liquid in 78% yield, 1.12 g.
1H NMR (400 MHz, DMSO-D6) 6: 0.01 (s, 6H), 0.88 (s, 9H), 1.69 (d, 3H), 4.09 (m, 2H), 5.20 (m, 1H),
6.84 (d, 1H), 7.92 (d, 1H), 8.96 (s, 1H), 9.18 (s, 1H).
Preparation 11: 7-[(1R)—1-Methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidine
”C I \
OJ'HIMe
The title compound was ed according to the method described for Preparation 8 using 4-chloro
[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 6) to
afford the title compound as a yellow oil in 100% yield, 13.78 g.
LCMS (system 2): R = 0.73 min; m/z 262 [M+H]+.
W0 2012/137089 129
Preparation 12: 7-[(1S)—1-Methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidine
' \
KN N
O\)‘Me
The title compound was prepared according to the method described for Preparation 8 using ro
[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidine (see ation 7) to
afford the title compound as a colourless oil in 90% yield, 9.18 g.
1H NMR (400 MHz, coc13) 5: 1.44-1.69 (m, 9H), 3.42 (m, 1H), 3.57 (m, 1H), 3.69 (m, 1H), 4.02 (m, 1H),
4.54 (m, 1H), 5.27 (m, 1H), 6.69 (d, 1H), 7.57 (d, 1H), 8.91 (s, 1H), 8.99 (s, 1H).
Preparation 13: 7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidine
Me Me
The title compound was prepared according to the method described for ation 8 using 7-tert-butyl-
4-chloro-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 5) to afford the title compound as a yellow liquid in
94% yield, 1.27 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.75 (s, 9H), 6.57 (d, 1H), 7.66 (d, 1H), 8.78 (s, 1H), 8.98 (s, 1H).
Preparation 14: 7-[(1S)—2-{[tert—Butyl(dimethyl)silyl]oxy}methylethyl]iodo-7H-pyrrolo[2,3-d]pyrimidine
N-lodosuccinimide (124 g, 553 mmol) was added to )—2-{[tert—butyl(dimethyl)silyl]oxy}
methylethyl]-7H-pyrrolo[2,3-d]pyrimidine (153.4 g, 526 mmol) (see Preparation 8) in acetonitrile (700 mL).
The e was stirred at room temperature for 16 hours then saturated aqueous sodium thiosulfate (700
mL) was added. The mixture was extracted with EtOAc (800 mL) then the organic extract was dried over
magnesium sulfate and evaporated in vacuo. The crude material was purified by column chromatography
on silica gel (gradient of pentane:EtOAc 90:10 to 80:20) to afford the title compound as a yellow solid in
66% yield, 145 g. 1H NMR (400 MHz, CDCI3) 6: -0.90 (d, 6H) 0.80 (s, 9H) 1.58 (d, 3H) 3.84 (m, 2H) 5.07
(m, 1H) 7.48 (s, 1H) 8.73 (s, 1H) 8.86 (s, 1H).
W0 2012/137089 130
Preparation 15: 7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)iodo-7H-pyrrolo[2,3-d]pyrimidine
The title compound was prepared according to the method described for Preparation 14 using 7-(2-{[tert-
dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 9) to afford the
title compound as a brown oil in 59% yield, 270 mg.
1H NMR (400 MHz, CDCI3) 6: 0.01 (s, 6H), 0.92 (s, 9H), 2.00 (s, 6H), 4.20 (s, 2H), 7.95 (s, 1H), 9.02 (s,
1H), 9.16 (s, 1H).
Preparation 16: (R,S) [tert-Butyl(dimethyl)silyl]oxy}methylethyl)iodo-7H-pyrrolo[2,3-
d]pyrimidine
The title compound was prepared according to the method described for Preparation 14 using (R,S) 7-(2-
{[tert—butyl(dimethyl)silyl]oxy}methylethyl)-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 10) to afford
the title compound as a yellow liquid in 74% yield, 1.18 g.
LCMS (system 1): R = 4.03 min; m/z 418 [M+H]+.
Preparation 17: 5-lodo[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidine
The title compound was prepared according to the method described for ation 14 using 7-[(1R)—1-
methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidine (see Preparation 11) to afford
the title compound as a brown oil in 34% yield, 7.50 g.
1H NMR (400 MHz, CDCI3) 6: 1.43-1.74 (m, 9H), 3.42 (m, 1H), 3.54 (m, 1H), 3.67 (m, 1H), 3.99 (m, 1H),
4.54 (m, 1H), 5.23 (m, 1H), 7.51 (s, 1H), 8.74 (s, 1H), 8.88 (s, 1H).
W0 2012/137089 131
Preparation 18: 5-lodo[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidine
The title compound was prepared according to the method described for Preparation 14 using 7-[(1S)—1-
methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidine (see Preparation 12) to afford
the title compound as a brown oil in 71% yield, 9.68 g.
1H NMR (400 MHz, CDCI3) 6: 1.44-1.65 (m, 9H), 3.45 (m, 1H), 3.56 (m, 1H), 3.67 (m, 1H), 3.99 (m, 1H),
4.54 (m, 1H), 5.24 (m, 1H), 7.51 (s, 1H), 8.76 (s, 1H), 8.89 (s, 1H).
ation 19: 7-tert-Butyliodo-7H-pyrrolo[2,3-d]pyrimidine
Me
The title compound was prepared according to the method described for Preparation 14 using 7-tert-
butyl-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 13) to afford the title compound as a yellow solid in
71% yield, 1.55 g.
LCMS (system 1): R = 3.12 min; m/z 302 .
Preparation 20: (R,S) Methyl 2-(5-iodo-7H-pyrrolo[2,3-d]pyrimidinyl)propanoate
KN N
Me>\’(OO
Methylbromopropionate (6.83 mL, 61.2 mmol) was added to a mixture of -7H-pyrrolo[2,3-
d]pyrimidine (15.0 g, 61.0 mmol) and cesium carbonate (35.9 g, 110.0 mmol) in DMF (75 mL). The
mixture was stirred at room temperature for 4 hours. The reaction mixture was d with water (250 mL)
and extracted with diethyl ether (100 mL). The organic layer was washed with brine (70 mL), dried over
magnesium sulfate and ated in vacuo to afford the title compound as an off-white solid in 83%
yield, 16.87 g.
1H NMR (400 MHz, CDCI3) 6: 1.82 (d, 3H), 3.76 (s, 3H), 5.72 (q, 1H), 7.48 (s, 1H), 8.76 (s, 1H), 8.89 (s,
1H).
W0 2012/137089 132
Preparation 21: Methyl 2-(5-iodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropanoate
Potassium t—butoxide (71.3 mL, 71.3 mmol, 1.0 M in THF) was added to (R,S) methyl 2-(5-iodo-7H-
pyrrolo[2,3-d]pyrimidinyl)propanoate (16.9 g, 50.9 mmol) (see Preparation 20) and iodomethane (4.44
mL, 71.3 mmol) in THF (100 mL). The mixture was stirred at room temperature for 15 s then water
(20 mL) and aqueous HCI (0.3 mL, 2M) was added. THF was d by evaporation in vacuo then the
aqueous residue was extracted with EtOAc (250 mL). The organic phase was dried over magnesium
sulfate and evaporated in vacuo. The crude solid was purified by column chromatography on silica gel
(80:20 pentane:EtOAc) to afford the title nd as a white solid in 51% yield, 8.92 g.
1H NMR (400 MHz, CDCI3) 6: 1.93 (s, 6H), 3.68 (s, 3H), 7.43 (s, 1H), 8.75 (s, 1H), 8.85 (s, 1H).
Preparation 22: 2-(5-lodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropanol
Lithium borohydride (32.3 mL, 64.6 mmol, 2.0 M in THF) was added to methyl 2-(5-iodo-7H-pyrrolo[2,3-
d]pyrimidinyl)methylpropanoate (8.92 g, 25.9 mmol) (see ation 21) in ethanol (70 mL). The
mixture was stirred at room temperature for 17 hours then water (70 mL) was added. The mixture was
evaporated in vacuo then the residue was partitioned between DCM (250 mL) and water (50 mL). The
aqueous phase was extracted with DCM:MeOH (90:10, 2 x 250 mL) and the combined organic phases
were dried over ium e and evaporated in vacuo to afford the title compound as an off- white
solid in 100% yield, 8.20 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.65 (s, 6H), 3.16 (d, 2H), 7.77 (s, 1H), 8.67 (s, 1H), 8.80 (s, 1H).
Preparation 23: 5-[(Diphenylmethylene)amino]-N-methoxy—N-methylnicotinamide
N/l \
Me\l?l
Benzophenone imine (205 mL, 1.22 mol) was added to 5-bromo-N-methoxy-N-methylisonicotinamide
(250 g, 1.02 mol), tris(dibenzylideneacetone)dipalladium (28.0 g, 31.0 mmol), 2-di-tert-butylphosphino—
W0 2012/137089 133 2012/051363
2',4',6'-triisopropylbiphenyl (34.7 g, 82.0 mmol) and freshly ground ium phosphate tribasic (541 g,
2.55 mol) in 1,2-dimethoxyethane (2500 mL). The mixture was stirred at 50°C for 17 hours. The reaction
mixture was filtered h ArbocelTM and the pad was washed with EtOAc (500 mL). The filtrate was
evaporated in vacuo and the crude material was purified by column chromatography on silica gel (gradient
of heptane:EtOAc 70:30 to 0:100) to afford the title compound as an orange gum in 51% yield, 180.0 g.
1H NMR (400 MHz, DMSO-D6) 6: 3.19 (s, 3H), 3.37 (s, 3H), .23 (m, 2H), 7.29 (m, 1H), 7.32-7.39
(m, 3H), 7.46-7.53 (m, 2H), 7.57 (m, 1H), 7.67-7.73 (m, 2H), 8.09 (d, 1H), 8.27 (d, 1H).
Preparation 24: {7-[(1S)—2-{[tert—Butyl(dimethyl)silyl]oxy}methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}{5-
[(diphenylmethylene)amino]pyridinyl}methanone
lsopropyl magnesium chloride (68.8 mL, 138 mmol, 2.0 M in THF) was added to 7-[(1S)—2-{[tert—
butyl(dimethyl)silyl]oxy}methylethyl]iodo-7H-pyrrolo[2,3-d]pyrimidine (52.2 g, 125 mmol) (see
Preparation 14) in THF (400 mL) at 0°C, under nitrogen. The mixture was stirred at 0°C for 1 hour then a
solution of 5-[(diphenylmethylene)amino]-N-methoxy-N-methylnicotinamide (47.5 g, 138 mmol) (see
Preparation 23) in THF (100 mL) was added dropwise at 0°C. The mixture was warmed to room
temperature and stirred at this temperature for 16 hours. The reaction mixture was quenched with 10%
aqueous ammonium chloride (250 mL) and extracted with ethyl acetate (2 x 250 mL). The combined
organic extracts were washed with brine (250 mL), dried over sodium sulfate, evaporated in vacuo and
the crude material was purified by column tography on silica gel (gradient of EtOAc:pentane 10:90
to 60:40) to afford the title nd as a less gum in 88% yield, 63.2 g.
R = 7.94 min; m/z 576 [M+H]+.
Preparation 25: [7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]{5-
[(diphenylmethylene)amino]pyridinyl}methanone
W0 2012/137089 134 2012/051363
The title compound was prepared according to the method described for Preparation 24 using 7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)iodo-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 15) and
-[(diphenylmethylene)amino]-N-methoxy—N-methylnicotinamide (see Preparation 23) to afford the title
compound as a colourless gum in 56% yield, 1.49 g.
1H NMR (400 MHz, CDCI3) 6: 0.21 (s, 6H), 0.67 (s, 9H), 1.80 (s, 6H), 4.10 (s, 2H), 7.14 (d, 2H), 7.33 (m,
3H), 7.45 (m, 2H), 7.54 (m, 1H), 7.58 (t, 1H), 7.79 (d, 2H), 7.89 (s, 1H), 8.18 (d, 1H), 8.58 (d, 1H), 9.01 (s,
1H), 9.60 (s, 1H).
Preparation 26: (5-Bromopyridinyl){7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-
pyrrolo[2,3-d]pyrimidinyl}methanone
The title compound was prepared according to the method described for Preparation 24 using 5-iodo
[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 17)
and 5-bromo-N-methoxy—N-methylnicotinamide to afford the title compound as a brown oil in 66% yield,
215 mg.
LCMS (system 2): R = 1.27 min; m/z 447 [M+H]+.
Preparation 27: (5-Bromopyridinyl){7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}methanone
N/ Br
KN I \
of'v'e
The title nd was prepared ing to the method described for Preparation 24 using 5-iodo
[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidine (see ation 18)
and 5-bromo-N-methoxy—N-methylnicotinamide to afford the title compound as a colourless oil in 32%
yield, 181 mg.
LCMS (system 2): R = 1.27 min; m/z 447 [M+H]+.
Preparation 28: (R,S) {7-[2-{[tert—Butyl(dimethyl)silyl]oxy}methylethyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}{5-[(diphenylmethylene)amino]pyridinyl}methanone
W0 2012/137089 135
nButyllithium (0.57 mL, 1.31 mmol, 2.3 M in s) was added to (R,S) 7-(2-{[tert—
butyl(dimethyl)silyl]oxy}methylethyl)iodo-7H-pyrrolo[2,3-d]pyrimidine (500 mg, 1.19 mmol) (see
Preparation 16) in dry ether (20 mL) at -78°C and the reaction mixture was stirred for 30 minutes. Then
-[(diphenylmethylene)amino]-N-methoxy-N-methylnicotinamide (372 mg, 1.07 mmol) (see Preparation
23) in dry ether (25mL) was added drop wise at the same temperature. After 15 s the mixture was
quenched with saturated aqueous ammonium chloride (50 mL) and extracted with ethyl acetate (70 mL).
The organic t was dried over sodium sulfate, evaporated in vacuo and purified by column
chromatography on silica gel (hexane:EtOAc 70: 30) to afford the title compound as an off-white solid in
19% yield, 134 mg.
LCMS (System 4): R = 4.53 min; m/z 576 [M+H]+.
Preparation 29: (R,S) (5-Bromopyridinyl)(7-tert-butyl-7H—pyrrolo[2,3-d]pyrimidinyl)methanol
l \
Me Me
The title compound was prepared according to the method bed for Preparation 28 using 7-tert-
butyliodo-7H-pyrrolo[2,3-d]pyrimidine (see ation 19) and 5-bromo—pyridinecarbaldehyde to
afford the title compound as a colourless oil in 37% yield, 486 mg.
LCMS (System 4): R = 2.94 min; m/z 362 [M+H]+.
Preparation 30: (5-Bromopyridinyl)(7-tert—butyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
W0 2012/137089 136
2-lodoxybenzoic acid (909 mg, 3.25 mmol) was added to (R,S) (5-bromopyridinyl)(7-tert-butyl-7H-
pyrrolo[2,3-d]pyrimidinyl)methanol (405 mg, 1.34 mmol) (see Preparation 29) in ethyl e (30mL)
and the mixture was refluxed for 4 hours. The mixture was filtered and the filtrate was ated in
vacuo to afford the title compound as a white solid in 95% yield, 554 mg.
LCMS (system 4): R, = 3.28 min; m/z 360 [M+H]+.
Preparation 31: (5-Aminopyridinyl)(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
K | N
Me Me
Copper sulfate pentahydrate (55 mg, 0.24 mmol) was added to (5-bromopyridinyl)(7-tert-butyl-7H-
pyrrolo[2,3-d]pyrimidinyl)methanone (292 mg, 0.81 mmol) (see Preparation 30) and concentrated
ammonia solution (20 mL). The mixture was heated in a sealed vessel at 140°C for 17 hours. The reaction
mixture was evaporated in vacuo and the residue was stirred in aqueous hydrochloric acid (10 mL, 2M) at
room temperature for 17 hours. The reaction mixture was basified to pH 9 using ted s
sodium carbonate then extracted with DCM (3 x 20 mL). The combined organic ts were dried over
magnesium sulfate and evaporated in vacuo to afford the title compound as a white solid in 49% yield,
240 mg.
LCMS (System 4): R, = 2.68 min; m/z 296 [M+H]+.
ation 32: (5-Aminopyridinyl){7—[(1S)—2-hydroxymethylethyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}methanone
The title compound was prepared according to the method described for Preparation 31 using (5-
bromopyridinyl){7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidin
yl}methanone (see Preparation 27) to afford the title compound as a white solid in 10% yield, 134 mg.
LCMS (system 2): R, = 0.59 min; m/z 298 [M+H]+.
W0 2012/137089 137
Preparation 33: (5-Aminopyridinyl){7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}methanone
N/ NH2
KN ' \
O\)‘Me
Benzophenone imine (0.40 mL, 2.4 mmol) was added to (5-bromopyridinyl){7-[(1S)—1-methyl
(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone (891 mg, 2.0 mmol) (see
Preparation 27), tris(dibenzylideneacetone)dipalladium (55 mg, 0.06 mmol), 2-di-tert—butylphosphino-
2',4',6'-triisopropylbiphenyl (68 mg, 0.16 mmol) and freshly ground potassium phosphate tribasic (1.06 g,
.0 mmol) in 1,2-dimethoxyethane (4 mL). The mixture was stirred at 50°C for 17 hours. The reaction
mixture diluted with DCM (10 mL), filtered through ArbocelTM and the pad was washed with DCM (5 mL).
The filtrate was evaporated in vacuo and the crude material was dissolved in THF (10 mL). Aqueous citric
acid (5 mL, 2M) was added and the mixture was stirred at room ature for 16 hours. Water (40 mL)
was added then sodium hydroxide was added to basify the mixture. The e was extracted with
EtOAc (3 x 40 mL) and the combined c extracts were dried over magnesium sulfate and evaporated
in vacuo. The e was purified by column chromatography on silica gel ent of EtOAc:MeOH
100:0 to 80:20) to afford the title compound as a white solid in 70% yield, 506 mg.
LCMS (system 1): R = 3.27 min; m/z 382 [M+H]+.
Preparation 34: 2-(2H-Benzotriazolyl)-N-[5-({7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-
7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyl]acetamide
The title compound was prepared according to the method described for Example 1 using (5-
aminopyridinyl){7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidin
yl}methanone (see Preparation 33) and benzotriazolyl-acetic acid to afford the title compound as a
yellow solid in 72% yield, 70 mg.
LCMS (system 5): R = n, m/z 541 [M+H]+.
Preparation 35: 2-(2,4-Difluorophenyl)—N-[5-({7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-
pyrrolo[2,3-d]pyrimidin-5—yl}carbonyl)pyridinyl]acetamide
W0 2012/137089 138
The title compound was prepared according to the method bed for Example 1 using (5-
aminopyridinyl){7-[(1S)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidin
y|}methanone (see Preparation 33) and 2,5-difluorophenylacetic acid to afford the title compound as a
yellow solid in 75% yield, 75 mg.
LCMS (system 5): R = 3.08min, m/z 536 [M+H]+.
Preparation 36: (5-Aminopyridinyl){7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}methanone
Copper (l) oxide (9.2 mg, 0.06 mmol) was added to (5-bromopyridinyl){7-[(1R)—1-methyl(tetrahydro-
2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidinyl}methanone (285 mg, 0.64 mmol) (see
Preparation 26) and trated ammonia solution (2 mL) in 1-methylpyrrolidinone (0.5 mL). The
mixture was heated in a sealed vessel at 80°C for 17 hours. Ethyl acetate (5 mL) and water (5 mL) were
added to the reaction mixture and then filtered through a glass fibre filter. The organic phase was dried
over magnesium e and evaporated in vacuo. The crude solid was purified by column
chromatography on silica gel (gradient of EtOAc:MeOH:cNH3 100:0:0 to 95:5:0.5) to afford the title
compound as a colourless oil in 70% yield, 171 mg.
LCMS m 2): R = 0.78 min; m/z 382 [M+H]+.
Preparation 37: (5-Aminopyridinyl){7-[(1S)—2-{[tert—butyl(dimethyl)silyl]oxy}methylethyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}methanone
W0 2012/137089 139 2012/051363
s citric acid (120 mL, 2.0 M) was added to {7-[(1S)—2-{[tert—butyl(dimethyl)silyl]oxy}methylethyl]-
7H-pyrrolo[2,3-d]pyrimidinyl}{5-[(diphenylmethylene)amino]pyridinyl}methanone (63.2 g, 110 mmol)
(see Preparation 24) in THF (274 mL) and the mixture was stirred at room temperature for 17 hours. The
mixture was cooled to 0°C, water (200 mL) added and the e was basified using sodium hydroxide
(28 g). The mixture was extracted with ethyl acetate (150 mL) then the aqueous phase was extracted with
ethyl acetate (2 x 200 mL). The combined organic phases were washed with brine (600 mL), dried over
sodium sulfate and evaporated in vacuo to afford the title compound as a semi-solid in tative yield,
45.2 g.
LCMS (system 2): R = 1.16 min; m/z 412 [M+H]+.
Preparation 38: (5-Aminopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-
pyrrolo[2,3-d]pyrim idinyl]methanone
N/ NH2
l \
Me“aMeO
Me‘sf—Me
lVIe‘é‘Me
The title compound was prepared according to the method described for Preparation 37 using [7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]{5-
[(diphenylmethylene)amino]pyridinyl}methanone (see Preparation 25) to afford the title compound as a
white solid in 81% yield, 872 mg.
1H NMR (400 MHz, DMSO-d6) 6: 0.22 (s, 6H), 0.63 (s, 9H), 1.66 (s, 6H), 4.12 (s, 2H), 5.65 (s, 2H), 7.27
(dd, 1H), 8.08 (s, 1H), 8.14-8.17 (m, 2H), 8.97 (s, 1H), 9.45 (s, 1H).
W0 2012/137089 140
Preparation 39: (R,S) (5-Aminopyridinyl){7-[2-{[tert-butyl(dimethyl)silyl]oxy}methylethyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}metha none
N/ NH2
I \
MeNH
Me\s{—Me
M9+Me
The title nd was prepared according to the method described for Preparation 37 using (R,S) {7-
ert-butyl(dimethyl)silyl]oxy}methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}{5-
[(diphenylmethylene)amino]pyridinyl}methanone (see Preparation 28) to afford the title compound as a
white solid in 89% yield, 86 mg.
LCMS (System 4): R =1.37 min; m/z 412 .
Preparation 40: 2-Chloroiodo-7H—pyrrolo[2,3-d]pyrimidine
CI N N
H
N-lodosuccinimide (742 g, 3.30 mol) was added to 2-chloro-7H—pyrrolo[2,3-d]pyrimidine (482.5 g, 3.14
mol) in acetonitrile (2500 mL) at 12°C. The mixture was stirred at room temperature for 1 hour then
sodium metabisulphite (650 g in 4500 mL of water) was added. The mixture was stirred for 1 hour then
filtered to afford the title compound as a orange solid in 82% yield, 716.2 g.
1H NMR (400 MHz, DMSO-D6)6: 7.83 (s, 1H), 8.63 (s, 1H), 12.73 (s, 1H).
Preparation 41: Methyl 2-(2-chloro—5-iodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropanoate
Methyl 2-bromomethylpropanoate (663 mL, 5.13 mmol) was added to 2-chloroiodo-7H—pyrrolo[2,3-
d]pyrimidine (358.1 g, 1.28 mol) (see Preparation 40), potassium iodide (21.3 g, 128 mmol) and cesium
carbonate (1670 g, 5.13 mol) in DMF (7162 mL). The e was heated at 60°C for 19 hours. The
reaction mixture was diluted with water (7000 mL) and stirred at room temperature for 42 hours. The
mixture was filtered and the solid was washed with water (500 mL) to afford the title compound as a beige
solid in 92% yield, 445.8 g.
W0 2012/137089 141
1H NMR (400 MHz, coc13) 6: 1.89 (s, 6H), 3.65 (s, 3H), 7.39 (s, 1H), 8.56 (s, 1H).
Preparation 42: 2-(2-Chloro—5-iodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropanoic acid
Lithium hydroxide monohydrate (4.08 g, 97.5 mmol) was added to methyl 2-(2-chloroiodo-7H-
pyrrolo[2,3-d]pyrimidinyl)methylpropanoate (18.5 g, 48.7 mmol) (see Preparation 41) in THF (185
mL) and water (45 mL). The mixture was stirred at 60°C for 3 hours then the reaction mixture volume was
reduced to one third by evaporation in vacuo. The aqueous residue was acidified using aqueous HCI (2.0
M) then extracted with EtOAc (4 x 200 mL). The organic phase was evaporated in vacuo and the crude
material was triturated with hexane (100 mL) to afford the title compound as a white solid in 90% yield,
16.0 g.
LCMS (system 5) R: 2.24 min; m/z 366 [M+H]+.
Preparation 43: 2-(2-Chloroiodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropanol
Cl N
MeMeOH
Route a
lsobutyl formate (6.6 mL, 50.02 mmol) was added to hloro—5-iodo-7H-pyrrolo[2,3-d]pyrimidin-
7-yl)methylpropanoic acid (16.6 g, 45.48 mmol) (see Preparation 42) and triethylamine (12.64 mL,
90.9 mmol) in THF (300 mL) at 0°C under en. The mixture was stirred at room temperature for 3
hours then filtered through a short plug of Celite TM. The filtrate was cooled to 0°C and sodium borohydride
(8.6 g, 227.6 mmol) in water (300 mL) was added. The mixture was stirred for 10 minutes at 0°C,
extracted with ethyl acetate (3 x 150 mL) then the organic extract was washed with brine (150 mL) and
dried over sodium sulfate. The solution was evaporated in vacuo and the residue was ated with
hexane to afford the title nd as a white solid in 63% yield, 10.0 g.
1H NMR (400 MHz, DMSO-Dg) 6: 1.64 (s, 6H), 3.85 (d, 2H), 4.99 (t, 1H), 7.82 (s, 1H), 8.63 (s, 1H).
W0 2012/137089 142
Route b
Diisobutylaluminium hydride (300 mL, 300 mmol, 1M in THF) was added dropwise to methyl 2-(2-chloro-
-iodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropanoate (51.8 g, 136 mmol) (see Preparation 41) in
THF (150 mL) at 0°C. The mixture was stirred for 90 minutes then methanol (27.9 mL) and aqueous HCI
(20 mL, 2M) was added. Water (100 mL), aqueous HCI (280 mL, 2M) and EtOAc (150 mL) were added
and the mixture was stirred at room temperature for 30 minutes. The mixture was filtered and the solid
was washed with water (150 mL) and tertbutylmethyl ether (150 mL) to afford the title compound as a
white solid in 56% yield, 26.8 g.
LCMS (System 1) R: 4.88 min; m/z 352 [M+H]+.
ation 44: 7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloroiodo-7H-pyrrolo[2,3-
d]pyrimidine
t—Butyldimethylsilyl chloride (78.8 g, 518 mmol) was added to 2-(2-chloroiodo-7H-pyrrolo[2,3-
d]pyrimidinyl)methylpropano| (140 g, 398 mmol) (see Preparation 43) and imidazole (67.8 g, 996
mmol) in DMF (996 mL) at 0°C. The mixture was stirred at room temperature for 16 hours. The mixture
was poured into saturated aqueous sodium bicarbonate (1500 mL) and extracted with heptane:EtOAc
(1:1, 1500 mL). The organic extract was washed with brine (2 x 900 mL) then dried over magnesium
e and ated in vacuo to afford the title nd as a brown gum in 96% yield, 178.2 g.
LCMS (System 1) R: 8.32 min; m/z 466 [M+H]+.
Preparation 45: 2-Chloro—7-[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]iodo-7H-pyrrolo[2,3-
d]pyrimidine
CIkN | N
MemeO
The title compound was prepared according to the method described for Preparation 6 using 2-(2-chloro-
5-iodo-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropano| (see ation 43) to afford the title
nd as a yellow oil in 74% yield, 7.3 g.
LCMS (system 5) R: 4.01 min; m/z 436 [M+H]+.
W0 2012/137089 143
Preparation 46: (5-Bromopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)—2-chloro-7H-
pyrrolo[2,3-d]pyrim idinyl] methanone
Cl N
‘ Me
Me/ISI—K
Me Me Me
The title compound was prepared according to the method described for Preparation 28 using 7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloro—5-iodo-7H-pyrrolo[2,3-d]pyrimidine (see Preparation
44) and 5-bromo-N-methoxy—N-methylnicotinamide to afford the title nd as a yellow solid in 42%
yield, 1.40 g.
1H NMR (400 MHz, CDCI3) 6: -0.18 (s, 6 H), 0.63 (s, 9 H), 1.72 (s, 6H), 4.09 (s, 2H), 8.24 (s, 1H), 8.39 (s,
1H), 8.95 (s, 1H), 8.99 (d, 1H), 9.36 (s, 1H).
ation 47: (5-Bromopyridinyl){2-chloro[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-
o[2,3-d]pyrimidin-5—yl}methanone
The title compound was prepared according to the method described for Preparation 28 using 2-chloro
[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]iodo-7H-pyrrolo[2,3-d]pyrimidine (see Preparation
45) and 5-bromo-N-methoxy—N-methylnicotinamide to afford the title compound as a yellow solid in 41%
yield, 3.0 g.
1H NMR (400 MHz, CDCI3) 6: 1.42-1.62 (m, 6H), 1.83 (s, 6H), 3.35-3.39 (m, 1H), 3.53-3.56 (m, 1H), 3.85
(d, 1H), 4.22 (d, 1H), 4.47 (m, 1H), 7.98 (s, 1H), 8.25 (s, 1H), 8.87 (s, 1H), 8.94 (s, 1H), 9.45 (s, 1H).
Preparation 48: [2-Amino—7-(2-hydroxy-1,1-dimethylethyl)—7H-pyrrolo[2,3-d]pyrimidinyl](5-
aminopyridinyl)methanone
N/ NHZ
| \
H2N)\\N N
MeweOH
The title compound was prepared according to the method described for Preparation 36
W0 2012/137089 144
using (5-bromopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloro-7H-pyrrolo[2,3-
midinyl]methanone (see Preparation 46) to afford the title compound as a yellow solid in 48%
yield, 300 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.64 (s, 6H), 3.90 (d, 2H), 5.07 (t, 1H), 5.60 (s, 2H), 6.50 (s, 2H), 7.23
(s, 1H), 7.61 (s, 1H), 8.11 (m, 2H), 8.93 (s, 1H).
[7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1 ,1-dimethylethyl)amino-7H-pyrrolo[2,3-d]pyrimidinyl]{5-
aminopyridinyl}methanone (Preparation 48a) was also ed from the reaction mixture.
1H NMR (400 MHz, DMSO) 6: -0.18 (s, 6H), 0.66 (s, 9H), 1.67 (s, 6H), 4.05 (s, 2H), 5.59 (s, 2H), 6.51 (s,
2H), 7.19 (s, 1H), 7.57 (s, 1H), 8.07 (s, 1H), 8.10 (s, 1H), 8.93 (s, 1H).
Preparation 49: {2-Amino[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}(5-aminopyridinyl)methanone
N/ NHZ
| \
H2NJ§N N
MeweO
The title compound was prepared according to the method described for Preparation 36
using (5-bromopyridinyl){2-chloro[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-
d]pyrimidinyl}methanone (see Preparation 47) to afford the title compound as a yellow solid in 52%
yield, 1.3 g.
LCMS (system 5) R: 2.72 min; m/z 411 .
Preparation 50: [7-(2-{[tert-Butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloro-7H-pyrrolo[2,3-
d]pyrimidinyl]{5-[(diphenylmethylene)amino]pyridinyl}methanone
I \
C'AN\ O
MeN):Me0‘
Me/ISI+
Me Me Me
pyl magnesium chloride (105 mL, 210 mmol, 2.0 M in THF) was added to 7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)chloroiodo-7H-pyrrolo[2,3-d]pyrimidine (89.0 g, 190 mmol)
(see Preparation 44) in THF (450 mL) at 0°C, under nitrogen. The mixture was stirred at 0°C for 1 hour
then a solution of 5-[(diphenylmethylene)amino]-N-methoxy—N-methylnicotinamide (72.6 g, 210 mmol)
W0 2012/137089 145
(see Preparation 23) in THF (200 mL) was added dropwise at 0°C. The mixture was warmed to room
temperature and stirred at this temperature for 16 hours. The on mixture was quenched with 10%
aqueous um chloride (500 mL) and the organic phase was separated. The aqueous phase was
extracted with ethyl acetate (2 x 300 mL). The combined organic extracts were washed with brine (400
mL), dried over sodium e, evaporated in vacuo and the crude al was purified by column
chromatography on silica gel ent of heptane:EtOAc 100:0 to 60:40) to afford the title compound as a
colourless gum in 66% yield, 78.9 g.
LCMS (System 1) R: 8.60 min; m/z 624 [M+H]+.
Preparation 51: (5-Aminopyridinyl){7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)[(2,4-
oxybenzyl)amino]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone
Me\O/©:O\HJ%N/|Me>§——O Me
|e Me/ISi—K
Me Me Me
2,4-Dimethoxybenzylamine (99.4 g, 594 mmol) was added to [7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-
dimethylethyl)chloro-7H-pyrrolo[2,3-d]pyrimidinyl]{5-[(diphenylmethylene)amino]pyridin
yl}methanone (53.0 g, 85 mmol) (see Preparation 50) and 4-dimethylaminopyridine (2.07 g, 17.0 mmol)
in 1,4-dioxane (170 mL). The mixture was heated to reflux for 2 days then cooled to room temperature
and filtered. The filtrate was evaporated in vacuo, the residue was dissolved in EtOAc (300 mL) and
washed with saturated aqueous ammonium chloride (500 mL). The organic phase was dried over
magnesium sulfate and evaporated in vacuo.
The crude e was dissolved in THF (200 mL) and aqueous citric acid (200 mL, 2M) was added. The
mixture was stirred at room temperature for 5 hours then diluted with water (200 mL). The mixture was
extracted with EtOAc (300 mL) and the organic extract was washed with aqueous potassium carbonate
(300 mL, 2M). The c phase was dried over magnesium sulfate, evaporated in vacuo and the
residue was purified by column chromatography on silica (gradient of pentane: EtOAc 100:0 to 0:100,
followed by EtOAc:MeOH 95:5) to afford the title compound as a less oil in 78% yield, 39.0 g.
LCMS (System 1) R: 5.97 min; m/z 591 [M+H]+.
Preparation 52: N-[5-({2-Amino[1,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}carbonyl)pyridinyl]—2-(4-cyclopropyl-1H-1,2,3-triazolyl)acetamide
W0 2012/137089 146 2012/051363
The title compound was prepared according to the method described for Example 1 using {2-amino
[1 ,1-dimethyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}(5-aminopyridin
y|)methanone (see Preparation 49) and (4-cyclopropyl-1H-1,2,3-triazolyl)acetic acid (see Preparation
83) to afford the title compound as a yellow solid in 86% yield, 70 mg.
LCMS (system 5): R: 2.87 min; m/z 560 [M+H]+.
Preparation 53: (R,S) Methyl 2-(2-chloroiodo-7H-pyrrolo[2,3-d]pyrimidinyl)propanoate
CI N H0
Me
The title compound was prepared according to the method described for Preparation 20 using 2-chloro
iodo-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 40) to afford the title compound as a brown solid in
62% yield, 18.0 g.
1H NMR (400 MHz, CDCI3) 6: 1.77 (d, 3H), 3.75 (s, 3H), 5.67 (q, 1H), 7.46 (s, 1H), 8.58 (s, 1H).
Preparation 54: (R,S) 2-(2-Chloroiodo-7H—pyrrolo[2,3-d]pyrimidinyl)propano|
The title nd was prepared according to the method described for Preparation 22 using (R,S)
methyl 2-(2-chloroiodo-7H-pyrrolo[2,3-d]pyrimidinyl)propanoate (see Preparation 53) to afford the
title nd as a yellow solid in 75% yield, 11.0 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.40 (d, 3H), 3.62-3.74 (m, 2H), 4.83 (m, 1H), 4.98 (t, 1H), 8.03 (s, 1H),
8.64 (s, 1H).
W0 2012/137089 147
Preparation 55: 7-(2-{[tert-Butyl(dimethyl)silyl]oxy}methylethyl)chloroiodo—7H-pyrrolo[2,3-
d]pyrimidine
The title compound was prepared ing to the method described for Preparation 44 using 2-(2-
chloro—5—iodo-7H—pyrrolo[2,3-d]pyrimidinyl)propanol (see Preparation 54) to afford the title
compound as a yellow solid in 89% yield, 12.50 g.
1H NMR (400 MHz, CDCI3) 6: -0.09 (s, 6H), 0.80 (s, 9H), 1.53 (d, 3H), 3.80 (d, 2H), 5.04 (m, 1H), 7.46 (s,
1H), 8.55 (s, 1H).
The enantiomers were separated using a Chiralpak IC 20 x 250 mm, 98:2:0.1 heptane:lPA:diethylamine
(flow rate — 18.0 ute).
Enantiomer 1 Yield 5.2 g, 99% e.e. (first eluting peak at 7.10 mins)
Enantiomer 2 Yield 5.0 g, 99% e.e. (second eluting peak at 7.64 mins)
Preparation 56: (5-Bromopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}methylethyl)chloro-7H-
pyrrolo[2,3-d]pyrimidinyl]methanone (enantiomer 1)
>\\ Me
0‘81
The title compound was ed according to the method described for Preparation 28 using [tert-
butyl(dimethyl)silyl]oxy}methylethyl)chloroiodo-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 55,
enantiomer 1) and 5-bromo-N-methoxy—N-methylnicotinamide to afford the title compound as a yellow
solid in 30% yield, 1.0 g.
1H NMR (400 MHz, DMSO-D6) 6: -0.14 (s, 6H), 0.61 (s, 9H), 1.52 (d, 3H), 3.91-3.96 (m, 2H), 5.00 (m, 1H),
8.37 (s, 1H), 8.58 (s, 1H), 8.95 (s, 1H), 9.00 (s, 1H), 9.33 (s, 1H).
W0 2012/137089 148
Preparation 57: [2-Amino—7-(2-hydroxy—1-methylethyl)-7H-pyrrolo[2,3-d]pyrimidin-5—yl](5-aminopyridin
y|)methanone (enantiomer 1)
1" \ 2
H2N \N N)\\
The title compound was ed ing to the method described for Preparation 36
using (5-bromopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}methylethyl)chloro-7H-pyrrolo[2,3-
d]pyrimidinyl]methanone (see Preparation 56) to afford the title compound as a yellow solid in 51%
yield, 450 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.41 (d, 3H), 3.66 (m, 2H), 4.74 (m, 1H), 5.01 (t, 1H), 5.59 (s, 2H), 6.55
(s, 2H), 7.22 (s, 1H), 7.83 (s, 1H), 8.11-8.13 (m, 2H), 8.91 (s, 1H); LCMS (system 5) R = 1.72 min; m/z
313 [M+H]+.
Preparation 58 : (5-Bromopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}methylethyl)chloro-7H-
pyrrolo[2,3-d]pyrimidinyl]methanone (enantiomer 2)
o \ /
I' \
CI \N N)\\
Me ,M9
O‘ISi Me
lVIe>4Me
The title compound was prepared ing to the method described for Preparation 28 using 7-(2-{[tert-
butyl(dimethyl)silyl]oxy}methylethyl)chloroiodo-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 55,
enantiomer 2) and 5-bromo-N-methoxy—N-methylnicotinamide to afford the title compound as a yellow
solid in 30% yield, 1.4 g.
1H NMR (400 MHz, DMSO-D6) 6: -0.14 (s, 6H), 0.61 (s, 9H), 1.52 (d, 3H), 3.91-3.96 (m, 2H), 5.00 (m, 1H),
8.37 (s, 1H), 8.58 (s, 1H), 8.95 (s, 1H), 9.00 (s, 1H), 9.33 (s, 1H).
W0 2012/137089 149
Preparation 59: [2-Amino—7-(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidin-5—yl](5-aminopyridin
hanone (enantiomer 2)
H2N \N N)\\
The title compound was prepared according to the method described for Preparation 36
using (5-bromopyridinyl)[7-(2-{[tert-butyl(dimethyl)silyl]oxy}methylethyl)chloro-7H-pyrrolo[2,3-
d]pyrimidinyl]methanone (see Preparation 58) to afford the title compound as a yellow solid in 51%
yield, 450 mg.
LCMS (system 5) R = 1.70 min; m/z 313 [M+H]+.
Preparation 60: 5-Bromo-N-tert—butylchloropyrimidinamine
kN%/|VI\BFN Me
Cl N
Me)LMe
utylamine (5.28 g, 72 mmol) was added to o-2,4-dichloropyrimidine (15 g, 66 mmol) and
triethylamine (19.9 g, 197 mmol) in acetonitrile (450 mL) at room temperature and the mixture was stirred
at room temperature for 16 hours. Then the mixture was evaporated in vacuo and the crude residue was
partitioned between EtOAc (450 mL) and water (400 mL). The c layer was separated, washed with
brine (400 mL) then dried over sodium sulfate and evaporated in vacuo. The residue was purified by
column chromatography on silica gel (hexane:EtOAc 88:12) to afford the title compound as a yellow oil in
52% yield, 8.8 g.
LCMS (system 5): R = 3.46 min; m/z 265 [M+H]+.
Preparation 61: N-tert—Butylchloro[(E)ethoxyvinyl]pyrimidinamine
ANNf/OV\ NH
Cl Me
Catecholborane (7.8 g, 65.4 mmol) in THF (50 mL) was added dropwise to a solution of 40%
ethoxyacetylene in hexane (12.8 mL, 72.5 mmol) under nitrogen at 0-5°C. The mixture was d for 2
hours at room temperature then heated at 70°C for 2 hours. The e was then cooled to room
temperature and a solution of 5-bromo-N-tert—butylchloropyrimidinamine (10 g, 37.8 mmol) (see
Preparation 60) in THF (50 mL) was added. The solution was degassed with argon for about 25 minutes
W0 2012/137089 150
followed by the addition of Pd(PPh3)4 (1.3 g, 1.13 mmol) and powdered sodium hydroxide (4.53 g, 113
mmol). The mixture was heated at 70°C for 16 hours and then cooled to room temperature. EtOAc (200
mL) was added and the e was filtered through a CeliteTM pad. The filtrate was evaporated in vacuo
and the residue was purified by column chromatography on silica gel (gradient of hexane:EtOAc 93:7 to
90:10) to afford the title compound as a yellow oil in 55% yield, 5.3 g.
LCMS m 5): R = 3.65 min; m/z 256 [M+H]+.
ation 62: 7-tert-Butylchloro-7H-pyrrolo[2,3-d]pyrimidine
C|)\\N N
Me'é‘Me
Concentrated HCI (25 mL) was added to N-tert—butyl-2—chloro[(E)ethoxyvinyl]pyrimidinamine (5.3
g, 20.72 mmol) (see Preparation 61) in panol (210 mL) and the mixture was heated at reflux for 4
hours. The reaction mixture was then evaporated in vacuo and the residue was basified with saturated
aqueous NaHCO3 and extracted with EtOAc (200 mL). The organic extract was dried over sodium sulfate
and evaporated in vacuo. The residue was purified by column chromatography on silica gel (gradient of
hexane:EtOAc 93:7 to 90:10) to afford the title nd as a yellow oil in 85% yield, 3.7 g.
LCMS (system 5): R = 3.42 min; m/z 210 [M+H]+.
Preparation 63: 7-tert-Butylchloroiodo-7H—pyrrolo[2,3-d]pyrimidine
Ax N
CI N
M99\Me
The title compound was ed according to the method described for Preparation 14 using 7-tert-
butylchloro-7H-pyrrolo[2,3-d]pyrimidine (see Preparation 62) to afford the title compound as a brown
solid in 87% yield, 4.7 g.
1H NMR (400 MHz, CDCI3) 6: 1.55 (s, 9H), 7.39 (s, 1H), 8.54 (s, 1H).
Preparation 64: (5-Bromopyridinyl)(7-tert-butylchloro-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
W0 2012/137089 151
The title compound was prepared according to the method described for Preparation 28 using 7-tert-
butylchloroiodo-7H-pyrrolo[2,3-d]pyrimidine (see ation 63) and 5-bromo-N-methoxy—N-
methylnicotinamide to afford the title compound as a brown oil in 36% yield, 2.1 g.
1H NMR (400 MHz, CDCI3) 6: 1.82 (s, 9H), 7.78 (s, 1H), 8.25 (s, 1H), 8.88 (s, 1H), 8.92 (s, 1H), 9.44 (s,
1H).
Preparation 65: (2-Aminotert-butyl-7H—pyrrolo[2,3-d]pyrimidinyl)(5-aminopyridinyl)methanone
H2N N N
lVIe9‘Me
The title compound was ed according to the method described for Preparation 36 using (5-
bromopyridinyl)(7-tert-butylchloro-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (see Preparation 64)
to afford the title compound as a white solid in 55% yield, 870 mg.
LCMS (system 5): R = 2.42 min; m/z 311 [M+H]+.
Preparations 66 to 70 were prepared according to Example 1, ng from (5-aminopyridinyl){7-[(1S)-
2-{[tert-butyl(dimethyl)silyl]oxy}methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone (see
Preparation 37) and the appropriate acids of formula :
W0 2012/137089 152
LCMS (system 4). R: 3.51min, m/z 589 [M+H]
LCMS(system 4): R: 3.55min, m/z 565 [M+H]
Preparations 71 to78 were prepared according to the method described above for Example 1, starting
from (5-aminopyridiny|)[7-(2-{[tert—buty|(dimethy|)si|y|]oxy}-1,1-dimethylethy|)-7H-pyrro|o[2,3-
d]pyrimidiny|]methanone (see Preparation 38) and the appropriate acids of formula :
(system 4): R: 3.88 min; m/z 616 [M+H]
LCMS m 4): R: 4.09 min; m/z 612 [M+H]
:- LCMS(system 4): R = 4.07 min; m/z 578 [M+H]+
LCMS(system 4): R: 3. 81 min; m/z 579 [M+H]
LCMS(system 4): R: 3.79 min; m/z 602 [M+H]
LCMS(system 4): R: 3.58 min; m/z 575 [M+H]
LCMS(system 4): R: 3.74 min; m/z 603 [M+H]
W0 2012/137089 153
LCMS (system 4): R = 3.78 min; m/z 573 [M+H]+
Potassium carbonate (7.67 g, 55.56 mmol) was added to 3-cyclopropyl-1H-pyrazole (2.0 g, 18.52 mmol)
in dry DMF (20 mL) at 25°C and the mixture was stirred for 20 minutes. Ethyl bromoacetate (2.06 mL,
18.52 mmol) was added then the mixture was stirred for 2 days at room ature. The reaction e
was neutralized with aqueous HCI (1.0 M), extracted with ether (40 mL) and the organic extract was
washed with brine (30 mL), dried over sodium sulfate then evaporated in vacuo. The residue was purified
by column chromatography on silica gel (hexane:EtOAc 88:12) to afford the title compound as a yellow oil
in 42% yield, 1.50 g.
1H NMR (400 MHz, DMSO) 6: 0.59 (d, 2H), 0.83 (d, 2H), 1.19 (t, 3H), 1.83 (m, 1H), 4.13 (q, 2H), 4.91 (s,
2H), 5.94 (d, 1H), 7.54 (d, 1H).
Preparation 80: (3-Cyclopropyl-1H-pyrazolyl)acetic acid
The title compound was prepared according to the method described for Preparation 42 using ethyl (3-
cyclopropyl-1H-pyrazolyl) acetate (see Preparation 79) to afford the title compound as a white solid in
83% yield, 4.06 g.
LCMS(system 4): R = 1.16 min; m/z167[M+H]+.
ation 81: [4-(Trifluoromethyl)-1H-1,2,3-triazolyl]acetic acid
F N=N\
F lOH
0
Trifluoromethyl acetylene (22.0 g, 0.234 mol) in THF (210 mL) was added to sodium ascorbate (2.77 g,
14.0 mmol), ethyl azidoacetate (27.1 g, 0.210 mol) and copper sulfate (4.76 mL, 0.3 M in water) in water
W0 2012/137089 154 2012/051363
(105 mL). The mixture was stirred at room temperature for 240 hours then evaporated in vacuo. The
residue was extracted with EtOAc (500 mL) and the organic phase was dried over ium sulfate
then ated in vacuo.
Sodium hydroxide (7.32 g, 0.183 mol) in water (30 mL) was added to the residue (32.7 g, 0.146 mol) in
methanol (50 mL) and the mixture was stirred at room temperature for 17 hours. The methanol was
evaporated in vacuo and the residue was diluted with water (10 mL). Potassium hydrogen sulfate (26.6 g,
0.195 mol) in water (70 mL) was added. The solution was evaporated in vacuo and the crude solid was
ed by crystallisation using water to afford the title compound as a white solid in 75% yield, 25.8 g.
1H NMR (400 MHz, DMSO-d6) 6: 5.40 (s, 2H), 8.85 (s, 1H), 13.50 (br s, 1H).
Preparation 82: Ethyl (4-cyclopropyl-1H—1,2,3-triazolyl)acetate
N=l\{
Vg/NClo
Cyclopropylacetyene (15 g, 0.116 mol), ethyl azidoacetate (11.5 g, 0.174 mol), triethylamine (0.32 mL,
2.33 mmol) and copper iodide (442 mg, 2.33 mmol) in acetonitrile (100 mL) were stirred at 25°C for 18
hours. The mixture was evaporated in vacuo and the residue was partitioned between water (100 mL) and
ethyl acetate (100 mL). The organic phase was dried over sodium sulfate, evaporated in vacuo and
purified by column chromatography on silica gel (EtOAc: Hexane 40: 60) to afford the title compound as a
colorless liquid in 95% yield, 21.6 g.
1H NMR (400 MHz, DMSO) 6: 0.68 (m, 2H), 0.90 (m, 2H), 1.21 (t, 3H), 1.95 (m, 1H), 4.17 (q, 2H), 5.29 (s,
2H), 7.81 (s, 1H).
Preparation 83: (4-Cyclopropyl-1H—1,2,3-triazolyl)acetic acid
N=l\{
The title compound was prepared according to the method described for Preparation 42 using ethyl (4-
cyclopropyl-1H—1,2,3-triazolyl)acetate (see Preparation 82) to afford the title nd as a yellow
solid in 63% yield, 13.0 g.
LCMS m 4): R = 1.86 min; m/z 186[M+H]+.
Preparation 84: tert—Butyl [4-(trifluoromethyl)-1H-pyrazolyl]acetate
W3”...Me)40Me
W0 2012/137089 155
The title compound was prepared according to the method bed for Preparation 79 using 4-
(trifluoromethyl)—1H-pyrazole and tert butyl cetate to afford the title compound as a yellow solid in
24% yield, 1.32 g.
LCMS(system 4): Rt=3.64min; m/z 251 [M+H]+.
Preparation 85: [4-(Trifluoromethyl)-1H-pyrazolyl]acetic acid
01..
Trifluoroacetic acid (10 mL) was added to tert-butyl [4-(trifluoromethyl)-1H-pyrazolyl]acetate (1.3 g, 5.2
mmol) (see Preparation 84) in dry DCM (10 mL) and the mixture was stirred for 18 hours at 25°C. Then
the mixture was evaporated in vacuo and the residue was purified by trituration with diethyl pentane
(1 :9, 2 mL) to afford the title compound as a white solid in 79% yield, 800 mg.
LCMS(system 4): R: n; m/z 193 [M+H]+.
Preparation 86: utyl [4-bromo-1H-pyrazolyl]acetate
BrQNl—N\
o OMe
Me
The title compound was prepared according to the method described for Preparation 79 using 4-bromo-
1H-pyrazole and tert butyl bromoacetate to afford the title compound as a yellow solid in 34% yield, 48.0
1H NMR (400 MHz, CDCI3) 6: 1.42 (s, 9H), 4.70 (s, 2H), 7.40 (s, 2H).
Preparation 87: tert—Butyl (4-cyclopropyl-1H-pyrazolyl)acetate
0 Me
Palladium acetate (215 mg, 0.957 mmol) was added to tert-butyl mo-1H-pyrazolyl]acetate (5 g,
19.14 mmol) (see Preparation 86), cyclopropyl boronic acid (8.22 g, 95.74 mmol), potassium phosphate
(8.12 g, 38.29 mmol) and tricyclohexylphosphine (537 mg, 1.91 mmol) in toluene: water (60mL:15mL).
The mixture was degassed for 20 minutes then refluxed for 18 hours. The reaction mixture was filtered
through CeliteTM, the filtrate was evaporated in vacuo and the residue was purified by column
chromatography on silica gel (gradient of EtOAc: hexane 15: 85) to afford the title cpompound as an off
white solid in 21% yield, 1.3 g.
LCMS (System 4): Rt=3.17min; m/z 223[M+H]+.
W0 2012/137089 156
Preparation 88: (4-Cyclopropyl-1H-pyrazolyl)acetic acid
\NNl0H
The title compound was prepared according to the method described for Preparation 85 using tert-butyl
(4-cyclopropyl-1H-pyrazolyl)acetate (see Preparation 87) to afford the title compound as a yellow solid
in 75% yield, 1.0 g.
LCMS m 4): R min; m/z 165[M+H]+.
Preparation 89: Ethyl (5-chloropyridinyl)acetate
OAMe
Cesium carbonate (71 g, 218 mmol) was added to 2-bromochloropyridine (14 g, 73 mmol) and diethyl
malonate (22 mL, 145 mmol) in dry 1,4-dioxane (280 mL) and the solution was degassed with argon for
minutes. Then copper (l) oxide (2.8 g, 14.55 mmol) and picolinic acid (3.6 g, 29 mmol) were added and
the mixture was stirred in a sealed vessel at 130°C for 24 hours. The mixture was cooled to room
temperature, ed with water (100 mL) and extracted with EtOAc (3 x 100 ml). The organic extracts
were washed with water (200 mL), brine (200 mL), dried over sodium sulfate and evaporated in vacuo.
The residue was purified by column chromatography on silica gel (EtOAc: Hexane 92: 8) to afford the title
compound as a yellow oil in 54% yield, 8.0 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.17 (t, 3H), 3.85 (s, 2H), 4.08 (q, 2H), 7.42 (d, 1H), 7.90 (dd, 1H), 8.54
(d, 1H).
Preparation 90: (5-Chloropyridinyl)acetic acid
The title compound was prepared according to the method described for Preparation 42 using ethyl (5-
pyridinyl)acetate (see Preparation 89) to afford the title compound as a brown solid in 51% yield,
3.5 g.
LCMS (system 4): R: 1.00 min; m/z 172 .
Preparation 91: Ethyl (5-fluoropyridinyl)acetate
OAMe
W0 2012/137089 157
The title nd was prepared according to the method described for Preparation 89 using 2-bromo-
ropyridine to afford the title compound as a yellow oil in 20% yield, 5 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.17 (t, 3H), 3.84 (s, 2H), 4.08 (q, 2H), 7.42-7.45 (m, 1H), 7.67-7.72 (m,
1H), 8.48 (d, 1H).
Preparation 92: (5-Fluoropyridinyl)acetic acid
F /{\j
The title compound was prepared ing to the method described for Preparation 42 using ethyl (5-
fluoropyridinyl)acetate (see Preparation 91) to afford the title nd as a brown solid in 57% yield,
2.4 g.
1H NMR (400 MHz, DMSO-d6) 6: 3.75 (s, 2H), 7.41-7.44 (m, 1H), 7.65-7.70 (m, 1H), 8.47 (d, 1H), 12.50
(br s, 1H).
ation 93: 5-lodoisopropyl-7H-pyrrolo[2,3-d]pyrimidine
“C I \
>‘Me
To a mixture of 5-iodo-7H-pyrrolo[2,3-d]pyrimidine (Preparation 201, 2.90 g, 12.0 mmol) and cesium
carbonate (5.78 g, 17.8 mmol) in DMF (45 mL) was added propane (1.78 mL, 17.8 mmol). The
mixture was stirred at room temperature for 3 hours. The reaction mixture was then poured onto saturated
aqueous ammonium chloride (500 mL) and a solid precipitated. The solid was collected by filtration,
rinsed with water (200 mL) and dried under reduced pressure for 17 hours to afford the title compound as
a brown solid in 77% yield, 2.61 g.
1H NMR (400 MHz, CDCI3) 6: 1.53 (d, 6H), 5.15 (m, 1H), 7.40 (s, 1H), 8.73 (s, 1H), 8.88 (s, 1H); LCMS
(system 2): R = 1.02 min; m/z 288 [M+H]+.
Preparation 94: (5-Bromopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
To a stirred solution of 5-iodoisopropyl-7H-pyrrolo[2,3-d]pyrimidine (Preparation 93, 4.85 g, 16.9
mmol) in THF (90 mL) at 0°C, under nitrogen was added isopropyl magnesium chloride (9.28 mL, 18.6
W0 2012/137089 158
mmol, 2.0 M in diethyl ether). The mixture was stirred at 0 °C for 1 hour then a solution of 5-bromo-N-
methoxy-N-methylnicotinamide (Preparation 227, 4.55 g, 18.6 mmol) in THF (10 mL) was added
dropwise at 0 °C. The mixture was warmed to room temperature and stirred for 16 hours. The reaction
mixture was quenched with saturated aqueous um de (200 mL) and extracted with ethyl
acetate (3 x 200 mL). The combined organics were concentrated under reduced re and purified by
silica gel column chromatography eluting with gradient of DCM 95:5 to 50:50 to afford a light brown
oil. The crude material was recrystallised using EtOAc:heptane (15:200 mL) to afford the title compound
as a white solid in 33% yield, 2.16 g.
1H NMR (400 MHz, CDCI3) 6: 1.60 (d, 6H), 5.20 (m, 1H), 7.79 (s, 1H), 8.28 (dd, 1H), 8.90 (d, 1H), 8.95 (d,
1H), 9.03 (s, 1H), 9.59 (s, 1H); LCMS (system 2): R = 1.36 min; m/z 346 .
Preparation 95: (5-Aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
The title compound was prepared according to the method described for Preparation 31 using (5-
bromopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 94) to afford the
title compound as a white solid in 69% yield, 1.20 g.
1H NMR (400 MHz, CDCI3) 6: 1.58 (d, 6H), 3.98 (br s, 2H), 5.18 (m, 1H), 7.39 (dd, 1H), 7.89 (s, 1H), 8.29
(brs, 1H), 8.43 (br s, 1H), 9.01 (s, 1H), 9.59 (s, 1H); LCMS (system 2): R = 0.50 min; m/z 282 [M+H]+.
Preparation 96: 4-Chlorooxetanyl-7H-pyrrolo[2,3-d]pyrimidine
I \
KN N
The title compound was prepared according to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde (Preparation 208) and oxetanamine to afford the title compound
as a yellow solid in 67% yield, 2.81 g.
LCMS m 1): R = 1.92 min; m/z 210, 212 [M+H]+.
W0 2012/137089 159
Preparation 97: 7-Oxetanyl-7H-pyrrolo[2,3-d]pyrimidine
The title nd was prepared according to the method described for Preparation 8 using 4-chloro
oxetanyl-7H-pyrrolo[2,3-d]pyrimidine (Preparation 96) to afford the title compound as a white solid in
90% yield, 1.20 g.
1H NMR (400 MHz, DMSO-d6) 6: 4.97-5.06 (m, 4H), 5.96 (m, 1H), 6.74 (d, 1H), 8.04 (d, 1H), 8.80 (s, 1H),
9.03 (s, 1H).
Preparation 98: —7-oxetanyl-7H-pyrrolo[2,3-d]pyrimidine
“%RN
The title compound was prepared according to the method described for Preparation 14 using an-
3-yl-7H-pyrrolo[2,3-d]pyrimidine (Preparation 97) to afford the title compound as a white solid in 49%
yield, 999 mg.
1H NMR (400 MHz, DMSO-d6) 6: .08 (m, 4H), 5.94 (m, 1H), 8.28 (s, 1H), 8.75 (s, 1H), 8.85 (s, 1H).
Preparation 99: {5-[(Diphenylmethylene)amino]pyridinyl}(7-oxetanyl-7H-pyrrolo[2,3-d]pyrimidin
yl)methanone
The title compound was prepared according to the method described for Preparation 94 using 5-iodo
oxetanyl-7H-pyrrolo[2,3-d]pyrimidine (Preparation 98) and 5-[(diphenylmethylene)amino]-N-methoxy—
N-methylnicotinamide (Preparation 23) to afford the title compound as a yellow solid in 55% yield, 253
mg.
1H NMR (400 MHz, DMSO-d6) 6: 4.95-5.06 (m, 2H), 5.15-5.24 (m, 2H), 5.96 (m, 1H), 7.21-7.80 (m, 11H),
8.22 (d, 1H), 8.52 (s, 1H), 8.62 (m, 1H), 9.01 (s, 1H), 9.44 (s, 1H).
W0 2012/137089 160
Preparation 100: (5-Aminopyridinyl)(7-oxetanyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
The title compound was prepared according to the method described for Preparation 37 using {5-
[(diphenylmethylene)amino]pyridinyl}(7-oxetanyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
(Preparation 99) to afford the title compound as a yellow solid in 96% yield, 128 mg.
1H NMR (400 MHz, DMSO-d6) 6: 4.95-5.04 (m, 2H), 5.14-5.22 (m, 2H), 5.66 (br s, 2H), 5.97 (m, 1H), 7.33
(m, 1H), 8.19 (d, 1H), 8.25 (d, 1H), 8.62 (s, 1H), 9.00 (s, 1H), 9.46 (s, 1H).
Preparation 101: 5-lodo{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine
| \
K N
Me-jSi~|\/|e
To a stirred solution of 5-iodo-7H-pyrrolo[2,3-d]pyrimidine (Preparation 201, 735 mg, 3.00 mmol) in DMF
(5 mL) at 0 °C was added to sodium hydride (132 mg, 3.30 mmol, 60% in oil). The mixture was stirred at
room temperature for 30 s, cooled to -20 °C and 2-(trimethylsilyl)ethoxymethyl chloride (0.58 mL,
3.30 mmol) added. The reaction e was stirred at -20°C for 3 hours then water (30 mL) was added.
The mixture was ted with EtOAc (2 x 50 mL) and the combined c phases were dried over
magnesium sulphate and evaporated under reduced pressure. The crude solid was purified by column
chromatography on silica gel, eluting with a gradient of heptane:EtOAc 100:0 to 50:50), to afford the title
compound as a white solid in 61% yield, 691 mg.
1H NMR (400 MHz, DMSO-d6) 6: -0.11 (s, 9H), 0.81 (t, 2H), 3.51 (t, 2H), 5.61 (s, 2H), 8.01 (s, 1H), 8.77 (s,
1H), 8.89 (s, 1H).
W0 2012/137089 161
Preparation 102: {5-[(Diphenylmethylene)amino]pyridinyl}(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-
pyrrolo[2,3-d]pyrimidinyl)methanone
I \
KN O
Me‘ZSi‘Me
The title compound was prepared according to the method described for Preparation 28 using 5-iodo
{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidine (Preparation 101) and 5-
[(diphenylmethylene)amino]-N-methoxy—N-methylnicotinamide (Preparation 23) to afford the title
compound as a yellow oil in 32% yield, 460 mg.
1H NMR (400 MHz, DMSO-d6) 6: -0.11 (s, 9H), 0.84 (m, 2H), 3.59 (m, 2H), 5.71 (s, 2H), 7.27 (m, 2H),
7.37 (m, 3H), 7.51 (m, 2H), 7.59 (m, 2H), 7.73 (d, 2H), 8.22 (d, 1H), 8.38 (s, 1H), 8.56 (d, 1H), 9.00 (s,
1H), 9.45 (s, 1H); LCMS (system 9): R = 2.36 min; m/z 534 [M+H]+.
Preparation 103: (5-Aminopyridinyl)(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin
yl)methanone
/\/o\/Z
MeTSi‘Me
The title compound was prepared according to the method described for Preparation 37 using {5-
[(diphenylmethylene)amino]pyridinyl}(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidin
yl)methanone (Preparation 102) to afford the title compound as a less oil in 73% yield, 2.10 g.
1H NMR (400 MHz, 6) 6: 0.09 (s, 9H), 0.84 (t, 2H), 3.60 (t, 2H), 5.66 (m, 4H), 7.30 (s, 1H), 8.18
(s, 2H), 8.55 (s, 1H), 9.02 (s, 1H), 9.47 (s, 1H); ystem 9): R = 3.25 min; m/z 370 [M+H]+.
ation 104: 2-(4-chlorophenyl)-N-(5-(7-((2-(trimethylsilyl)ethoxy)methyl)-7H-pyrrolo[2,3-
d]pyrimidinecarbonyl)pyridinyl)acetamide
W0 2012/137089 162
K | \
N O)
Me—jSi-Me
The title compound was prepared according to the method described for Examples 73-87 using (5-
aminopyridinyl)(7-{[2-(trimethylsilyl)ethoxy]methyl}-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
ration 103) and 4-chlorophenylacetic acid to afford the title compound as a white solid in 66%
yield, 223 mg.
1H NMR (400 MHz, DMSO-d6) 6: -0.11 (s, 9H), 0.82 (t, 3H), 3.60 (t, 2H), 3.74 (s, 2H), 5.70 (s, 2H), 7.37
(t, 4H), 8.47 (s, 1H), 8.60 (s, 2H), 8.72 (s, 1H), 8.98 (s, 1H), 9.03 (s, 1H), 9.48 (s, 1H);
LCMS (system 9): R = 3.50 min; m/z 522 [M+H]+.
Preparation 105: N-[5—({7-[(1S)—2-{[tert—butyl(dimethyl)silyl]oxy}methylethyl]-7H-pyrrolo[2,3-
d]pyrimidin--y)|}carbonylpyridin--y-|]-[3-(trifluoromethyl)phenyl]acetamide
TBSOx)‘élokd
Prepared according to Example 1, using (8) (5-aminopyridinyl){2-{[tert-butyl(dimethyl)silyl]oxy}
methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone (Preparation 37) and 3-
trifluoromethylphenylacetic acid with DIPEA as base.
1H NMR (400 MHz, DMSO) 6: -0.24 (s, 3H), -0.18 (s, 3H), 0.56 (s, 9H), 1.52 (d, 3H), 3.86 (m,3H), 3.94
(m, 1H), 5.06 (m, 1H), 7.58 (t, 1H), 7.63 (t,2H), 7.72 (s, 1H), 8.45 (s, 1H), 8.50 (s, 1H), 8.67 (d, 1H), 8.93
(d, 1H), 8.98 (s,1H), 9.45 (s,1H), 10.73 (s,1H).
LCMS (System 9): R = n; m/z 598 [M+H]+.
Preparation 106: 5-[(Diphenylmethylene)amino]nicotinaldehyde
/ H
To a stirred solution of phenylmethylene)amino]-N-methoxy—N-methylnicotinamide (Preparation 23,
7.50 g, 0.021 mol) in THF (150 mL) at -70°C was added diisopropylaluminium hydride (42 mL, 0.042 mol,
W0 2012/137089 163
1.0 M in THF) and the resulting mixture stirred at -70°C for 2 hours. Water (20 mL) and ethyl e
(100 mL) were added. The organic phase was separated, concentrated under reduced pressure and
purified by silica gel column chromatography eluting with EtOAc: petroleum ether 1:10 to afford the title
compound as a brown solid in 65% yield, 4 g.
The title compound can also be prepared according to the following process:
A mixture of 5-bromonicotinaldehyde (2790 mg, 15.0 mmol), diphenylmethanimine (3.01 mL, 18 mmol),
Pd2(dba)3 (412 mg, 0.45 mmol), di-tert-butyl(2',4',6'-triisopropyl-[1,1'-biphenyl]yl)phosphine (510 mg,
1.2 mmol) and K3PO4 (7960 mg, 37.5 mmol) in DME (30.0 mL) was stirred at 50 °C for 16 hours. After
cooling to room temperature, the reaction was diluted with DCM (50 mL) and the mixture filtered h a
pad of arbocel. The filter cake was washed with DCM (50 mL) and the filtrate concentrated under
reduced pressure. The crude material was tallized from ethyl acetate/heptane to give the desired
compound as a solid in 78% yield, 3341 mg.
1H NMR (400 MHz, DMSO-d6) 6: 7.18-7.26 (m, 2H), .39 (m, 3H), 7.47-7.54 (m, 2H), 7.55-7.62 (m,
2H), 7.68-7.75 (m, 2H), 8.25 (d, 1H), 8.63 (d, 1H), 10.00 (s, 1H).
Preparation 107: 2,2,3,3,9,9,10,10-octamethyl-4,8-dioxa-3,9-disilaundecanol
\/K/OTBDMS
To a stirred solution of glycerol (4.01 mL, 55 mmol) and ole (18.7 g, 275 mmol) in DMF (150 mL) at
0 °C was added tert-butyldimethylsilyl chloride (17.2 g, 113 mmol) in DMF (33 mL). The on e
was allowed to warm to room temperature and stirred for 16 hours. Water (500 mL) was added to the
reaction mixture and the resulting mixture extracted with heptane (500 mL x 3). The ed organic
layers were washed with water (300 mL), dried over MgSO4 and concentrated under reduced pressure.
The crude material was purified by silica gel column chromatography with a gradient elutant of
heptane:EtOAc 100:0 to 80:20 to afford the title compound as a colorless oil in 68% yield, 11.9 g.
1H NMR (400 MHz, DMSO-d6) 6: 0.03 (s, 12H), 0.86 (s, 18H), 3.40-3.59 (m, 5H), 4.58 (d, 1H).
Preparation 108: 2,2,3,3,9,9,10,10-octamethyl-4,8-dioxa-3,9-disilaundecanyl trifluoromethanesulfonate
FTe/oF
(3,ng
TBDMSO\)\/OTBDMS
To a stirred solution of 2,2,3,3,9,9,10,10-octamethyl-4,8-dioxa-3,9-disilaundecanol (Preparation 107,
6410 mg, 20 mmol) and pyridine (2.42 mL, 30 mmol) in DCM (40 mL) at -50 °C was added
trifluoromethanesulfonic anhydride (5.05 mL, 30 mmol) and the reaction stirred at -30 °C for 2 hours.
Aqueous 1 N HCl (40 mL) was added to the on and the mixture was extracted with DCM (40 mL x
3). The combined organic layers were concentrated under reduced pressure to obtain a colourless oil
which was used in the next step (Preparation 196) without r purification.
W0 2012/137089 164
Preparation 109: 4-Chloro-[7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl]{5-
[(diphenylmethylene)amino]pyridinyl}methanone
CI \ /
N/ \ RN I N\ 0
The title compound was prepared according to the method described for Preparation 28 followed by
Preparation 30 using roiodomethyl-7H—pyrrolo[2,3-d]pyrimidine (Preparation 226) and 5-
[(diphenylmethylene)amino]nicotinaldehyde (Preparation 106) to afford the title compound as a yellow
solid (11.0 g, 48%).
1H NMR (400 MHz, c0013) 6: 3.87 (s, 3H), 7.06 (m, 2H), 7.28 (m, 2H), 7.37 (m, 2H), 7.45 (m, 3H), 7.70
(m, 3H), 8.15 (d, 1H), 8.49 (d, 1H), 8.69 (s, 1H).
Preparation 110 :(5-Aminopyridinyl)(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone
“(H 2
N N
Citric acid (2 M, 200 mL) was added to 4-chloro-[7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl]{5-
[(diphenylmethylene)amino]pyridinyl}methanone (Preparation 109, 30 g, 0.066 mol) in THF (200 mL)
and the mixture was stirred at room temperature for 30 minutes. Ether (200 mL) was added and the
phases were separated. The aqueous layer was neutralised with aqueous sodium carbonate then the
solid was collected by filtration and dried under vacuum to give (5-aminopyridinyl)(4-chloro—7-methyl-
7H-pyrrolo[2,3-d]pyrimidinyl)methanone as a brown solid, 18 g, 95%
Methanethiol sodium salt (15.5 g, 0.22 mol) was added to 5-aminopyridinyl)(4-chloromethyl-7H-
pyrrolo[2,3-d]pyrimidinyl)methanone (21 g, 0.073 mol) in methanol (300 mL) and the e was
stirred at room temperature for 7 hours. The reaction e was poured into ice-water (200 mL) and the
precipitate was filtered. The filter cake was washed with water (100 mL) then acetone (20 mL) to afford
the (5-aminopyridinyl)[7-methyl(methylthio)-7H-pyrrolo[2,3-d]pyrimidinyl]methanone as a brown
solid, 15 g, 69%
Raney nickel (10 g) was added to (5-aminopyridinyl)[7-methyl(methylthio)-7H-pyrrolo[2,3-
midinyl]methanone (1.5 g, 5.0 mmol) and conc ammonia (150 mL) in 1,4 dioxane (150 mL). The
mixture was ed for 6 hours then filtered. The te was evaporated in vacuo and purified by prep
HPLC (method 4) to afford the title compound as a brown solid in 100% yield, 1.9 g.
1H NMR (400 MHz, DMSO-DG) 6: 3.88 (s, 3H), 5.63 (s, 2H), 7.27 (m, 1H), 8.14-8.18 (m, 2H), 8.40 (s, 1H),
8.97 (s, 1H), 9.42 (s, 1H).
W0 2012/137089 165
Preparation 111: 2,2,3,3,6,9,9,10,10-Nonamethyl-4,8-dioxa-3,9-disilaundecanamine
Me NH
TBDMSOQQ/OTBDMS2
The title compound was prepared according to the method described for Preparation 44 using 2-amino
methyl-1,3-propanediol to afford the title compound as a colourless oil in 100% yield, 23.0 g.
1H NMR (400 MHz, CDCI3) 6: 0.05 (s, 12H), .99 (m, 21H), 3.36-3.42 (m, 4H).
Preparation 112: 7-[2-{[tert—Butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-
methylethyl]chloro-7H-pyrrolo[2,3-d]pyrimidine
The title compound was prepared ing to the method described for ation 1 using
2,2,3,3,6,9,9,10,10-nonamethyl-4,8-dioxa-3,9-disilaundecanamine (Preparation 111) to afford the title
compound as a colourless gum in 75% yield, 8.91 g.
1H NMR (400 MHz, CDCI3) 6: 0.09-0.11 (m, 12H), .79 (m, 18H), 1.74 (s, 3H), 4.06-4.09 (m, 2H),
4.29-4.31 (m, 2H), 6.52 (m, 1H), 7.44-7.45 (m, 1H), 8.56 (m, 1H).
Preparation 113: 7-[2-{[tert—Butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-
methylethyl]-7H-pyrrolo[2,3-d]pyrimidine
N%\/IE\>K N
8%OTBDMS
OTBDMS
The title compound was prepared according to the method described for Preparation 8 using 7-[2-{[tert-
butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-methylethyl]chloro-7H-pyrrolo[2,3-
d]pyrimidine (Preparation 112) to afford the title compound as a yellow oil in 99% yield, 8.14 g.
1H NMR (400 MHz, CDCI3) 6: -0.13 (s,6H), -0.10 (s,6H), 0.78 ), 1.75 (s, 3H), 4.11 (d, 2H), 4.33 (d,
2H), 6.45 (d, 1H), 7.41 (d, 1H), 8.78 (s,1H), 8.90 (s, 1H).
W0 2012/137089 166
Preparation 114: 7-[2-{[tert—Butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-
methylethyl]iodo-7H-pyrrolo[2,3-d]pyrimidine
TBDMSO
The title compound was prepared according to the method described for Preparation 14 using 7-[2-{[tert-
butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-methylethyl]-7H-pyrrolo[2,3-d]pyrimidine
(Preparation 113) to afford the title compound as a yellow oil in 88% yield, 7.98 g.
1H NMR (400 MHz, CDCI3) 0 (s, 12H), 0.79 (s, 18H), 1.75 (s, 3H), 4.07 (d, 2H), 4.27 (d, 2H), 7.49
(s, 1H), 8.70 (s, 1H), 8.82 (s, 1H).
Preparation 115: {[tert—Butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-
methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}{5-[(diphenylmethylene)amino]pyridinyl}methanone
The title compound was ed according to the method described for Preparation 28 using 7-[2-{[tert-
butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)—1-methylethyl]iodo-7H-pyrrolo[2,3-
d]pyrimidine (Preparation 114) and 5-[(diphenylmethylene)amino]—N-methoxy—N-methylnicotinamide
(Preparation 23) to afford the title nd as a yellow foam in 69% yield, 1.76 g.
1H NMR (400 MHz, CDCI3) 6: 0.10-0.12 (m, 12H), 0.73-0.76 (m, 18H), 1.77 (s, 3H), 4.06-4.09 (m, 2H),
4.34-4.36 (m, 2H), 7.11-7.16 (m, 2H), 7.28-7.33 (m, 3H), 7.42-7.47 (m, 2H), 7.50-7.54 (m, 2H), 7.78-7.80
(m, 2H), 7.93 (s, 1H), 8.15-8.16 (m, 1H), 8.56 (m, 1H), 8.94 (s, 1H), 9.58 (s, 1H).
W0 2012/137089 167
Preparation 116: (5-Aminopyridinyl){7-[2-{[tert-butyl(dimethyl)silyl]oxy}({[tertbutyl
(dimethyl)silyl]oxy}methyl)methylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone
N/ NH2
K | \
8V.OTBDMS
TBDMSO
The title compound was prepared ing to the method described for Preparation 37 using {7-[2-{[tert-
butyl(dimethyl)silyl]oxy}({[tert-butyl(dimethyl)silyl]oxy}methyl)methylethyl]-7H-pyrrolo[2,3-d]pyrimidin-
-yl}{5-[(diphenylmethylene)amino]pyridinyl}methanone ration 115) to afford the title compound
as a yellow foam in 85% yield, 1.15 g.
LCMS (System 1): R = 4.01 min; m/z 556 [M+H]+.
Preparation 117: 1-lsopropyltrifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester
Me/kN
. \
FF F Me
To a suspension of 5-trifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester (13 g, 62.5 mmol) and
cesium carbonate (61.1 g, 187.5 mmol) in DMF (70 mL) was added -propane (6.86 mL, 68.75
mmol) and the resulting mixture allowed to stir at room ature for 16 hours. The crude reaction
mixture was poured onto water (100 mL) and extracted with EtOAc (100 mL x 3). The combined organics
were washed with water (50 mL x 2), brine (50 mL), dried over sodium sulphate, filtered and concentrated
under reduced pressure to afford the title compound as an off-white solid in 65% yield, 10.2 g.
1H NMR (400 MHz, CDCI3) 6: 1.33 (t, 3H), 1.53 (d, 6H), 4.30 (q, 2H), 4.50-4.57 (m, 1H), 8.00 (s, 1H);
LCMS (system 9): R: 3.55 min; m/z 251 [M+H]+.
Preparation 118: propyltrifluoromethyl-1H-pyrazolyl)—methanol
Me/k'?J \
The title compound was prepared according to the method described for Preparation 183 using 1-
isopropyltrifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester (Preparation 117) to afford the title
compound an off-white solid in 97% yield, 8.3 g.
W0 2012/137089 168
1H NMR (400 MHz, CDCI3) 6: 1.50 (d, 6H), 1.72 (t, 1H), 4.47-4.54 (m, 1H), 4.65 (d, 2H), 7.50 (s, 1H);
LCMS (system 9): R: 2.77 min; m/z 209 [M+H]+.
Preparation 119: (1-|sopropyltrifluoromethyl-1H-pyrazolyl)-acetonitrile
Me/k
The title compound was prepared ing to the method described for Preparation 184 using (1-
pyltrifluoromethyl-1H-pyrazolyl)—methanol (Preparation 118) to afford the title compound as
an off-white solid in 58% yield, 5 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.42 (d, 6H), 3.92 (s, 2H), 4.56-4.63 (m, 1H), 8.06 (s, 1H).
Preparation 120: 2-Chloroiodoisopropyl-7H-pyrrolo[2,3-d]pyrimidine
fl?N / \
>‘Me
The title compound was prepared according to the method described for Preparation 93 using ro
iodoisopropyl-7H—pyrrolo[2,3-d]pyrimidine (Preparation 40) to afford the title compound as a white solid
in 87% yield, 5.5 g.
1H NMR (400 MHz, CDCI3) 5: 1.50 (d, 6H), 5.10 (m, 1H), 7.36 (s, 1H), 8.55 (s, 1H); LCMS (System 10) R
= 3.6 min; m/z 322 [M+H]+.
Preparation 121: (5-Bromopyridinyl)(2-chloroisopropyl-7H—pyrrolo[2,3-d]pyrimidinyl)methanone
The title compound was prepared according to the method described for Preparation 28 using 2-chloro-5—
iodoisopropyl-7H—pyrrolo[2,3-d]pyrimidine (Preparation 120) and 5-bromo-N-methoxy—N-
methylnicotinamide to afford the title compound as a yellow solid in 41% yield, 1.2 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.54 (d, 6H), 5.02 (m, 1H), 8.41 (d, 1H), 8.63 (s, 1H), 9.0 (m, 2H), 9.33
(s, 1H).
Preparation 122: (2-Aminoisopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)(5-aminopyridinyl)methanone
W0 2012/137089 169
The title compound was prepared ing to the method described for Preparation 31 using (5-
bromopyridinyl)(2-chloroisopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 121) to
afford the title compound as a yellow solid in 58% yield, 500 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.47 (d, 6H), 4.82-4.86 (m, 1H), 5.60 (s, 2H), 6.58 (s, 2H), 7.23 (s, 1H),
7.88 (s, 1H), 8.12 (m, 2H), 8.91 (s, 1H); LCMS (System 9) R = 0.99 min; m/z 297 [M+H]+.
Preparation123: (1-|sopropyltrifluoromethyl-1H-pyrazolyl)—acetic acid
Me Me
“l/ OH
F F
F O
The title nd was prepared according to the method described for Preparation 185 using (1-
isopropyltrifluoromethyl-1H-pyrazolyl)-acetonitrile (Preparation 119) to afford the title compound as
off-white solid in 82% yield, 4.5 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.41 (d, 6H), 3.50 (s, 2H), 4.52-4.58 (m, 1H), 7.89 (s, 1H), 12.35 (br,
1H); LCMS (system 10): R: 1.56 min; m/z 235 [M-H]'.
Preparation 124: (3-Cyclopropylmethyl-1H-pyrazolyl)-acetonitrile
To a suspension of potassium tert-butoxide (8.95 g, 79.9 mmol) in DME (250 mL) -78 0C under nitrogen
was added a solution of 1-(isocyanomethyl yl)methyl benzene (9.36 g, 47.94 mmol) in DME (50
ml). After stirring for 10 minutes, a solution of 3-cyclopropylmethyl-1H-pyrazolecarbaldehyde (6 g,
39.95 mmol) in DME (100 mL) was added. The resulting mixture was allowed to stir at -78 0C for 1 hour
and then at room temperature for 1hour. ol (50 mL) was added and the resulting mixture refluxed
for 1 hour. The on e was quenched with saturated ammonium chloride solution (200 mL) and
extracted with EtOAc (2 x 200 mL). The combined organic layers were washed with brine (2 x 50 mL),
dried over sodium sulphate, filtered and concentrated under reduced pressure. The crude material was
purified by silica gel column chromatography eluting with hexane:EtOAc 90:10 to afford the title compound
an off-white solid in 67% yield, 4.3 g.
W0 2012/137089 170
1H NMR (400 MHz, DMSO-d6) 6: 0.67-0.7 (m, 2H), 0.79-0.83 (m, 2H), 1.75-1.80 (m, 1H), 3.68 (s, 3H),
3.79 (s, 2H), 7.54 (s, 1H); LCMS (system 9): R: 2.53 min; m/z162[M+H]+.
Preparation 125: (3-Cyclopropylmethyl-1H-pyrazolyl)-acetic acid
l\|/|e
0 \/
The title compound was prepared according to the method described for Preparation 141 using (3-
ropylmethyl-1H-pyrazolyl)—acetonitrile (Preparation 124) to afford the title nd as solid
in 83% yield, 4 g.
1H NMR (400 MHz, DMSO-d6) 6: 0.62-0.66 (m, 2H), 0.73-0.77 (m, 2H), 1.67-1.74 (m, 1H), 3.38 (s, 2H),
3.66 (s, 3H), 7.40 (s, 1H), 12.22 (br, 1H); LCMS (system 9): R: 1.97 min; m/z 181 [M+H]+.
Preparation 126: (3-Cyano—4-fluoro-phenyl)-acetic acid
N\\\/(>/erH0
To a solution of (3-bromo—4-fluoro—phenyl)—acetic acid (10 g, 42.9 mmol) in DMF (65 mL) was added
copper (l) e (7.7 g, 85.8 mmol) and heated at 130 °C for 24 hrs. The reaction mixture was cooled to
room temperature and diluted with ethyl e (250 mL). The organic layer was washed with water (5 x
50 mL), brine (50 mL), dried over sodium filtered and trated under reduced pressure. The crude
material was re-crystallized from diethyl ether and hexane to afford the title compound as a yellow solid in
65% yield, 5 g.
1H NMR (400 MHz, DMSO-d6) 6: 3.68 (s, 2H), 7.48 (t, 1H), 7.66-7.71 (m, 1H), 7.82 (dd, 1H), 12.53 (br s,
1H); LCMS (system 10): R: 1.39 min; m/z178[M-H]'.
Preparation 127: (3-Cyano—4-fluoro-phenyl)-acetic acid ethyl ester
Ora;/N
To a suspension of (3-cyano—4-fluoro-phenyl)—acetic acid (500 mg, 2.79 mmol) (Preparation 126) and
potassium carbonate (770 mg, 5.58 mmol) in DMF (5 mL) was added ethyl iodide (0.89 mL, 11.16 mmol)
and the reaction mixture stirred at room temperature for 4 hours. The crude on mixture was poured
onto water (10 mL) and extracted with ethyl acetate (3 x 15 mL). The combined organic layers were
washed with water (5 x 10 mL), brine (10 mL), dried over sodium sulphate, filtered and concentrated
under reduced pressure to afford the title compound as an off-white solid in 87% yield, 500 mg.
1H NMR (400 MHz, CDCI3) 6: 1.26 (t, 3H), 3.61 (s, 2H), 4.16 (q, 2H), 7.17 (t, 1H), 7.49-7.55 (m, 2H).
Preparation 128: (3-Amino-benzo[d]isoxazol-5—yl)—acetic acid ethyl ester
W0 2012/137089 171
r0 0
To a solution of (3-cyanofluoro-phenyl)—acetic acid ethyl ester (Preparation 127, 400 mg, 1.93 mmol)
and acetohydroxamic acid (362 mg, 4.83 mmol) in DMF (40 mL) and water (15 mL) was added ium
carbonate (1.6 g, 11.58 mmol) and the reaction mixture stirred at room temperature for 12 hours. The
on mixture was diluted with water (100 mL) and the resulting white precipitate and collected by
filtration. The crude material was purified by silica gel column chromatography eluting with a nt of
dichloromethane: methanol 100:0 to 97:3 to afford the title compound as an off-white solid in 59% yield,
250 mg.
LCMS (system 9): R: 2.87 min; m/z 221 [M+H]+.
Preparation 129: (3-Amino-benzo[d]isoxazolyl)—acetic acid
OH /
The title compound was prepared according to the method described for Preparation 141 using (3-amino-
benzo[d]isoxazolyl)-acetic acid ethyl ester ration 128) to afford the title compound as an off-
white solid in 69% yield, 30 mg.
1H NMR (400 MHz, DMSO-de) 6: 3.65 (s, 2H), 6.36 (br s, 2H), 7.36-7.42 (m, 2H), 7.69 (s, 1H), 12.37 (br s,
1 H); LCMS (system 10): R: 1.65 min; m/z193[M+H]+.
Preparation 130: o[1,2-a]pyridinyl-acetic acid ethyl ester
Me/\Ow\ \N
To a stirred solution of 7-bromo-imidazo[1,2-a]pyridine (600 mg, 3.0 mmol) and diethyl malonate (0.93
mL, 6.1 mmol) in dry dioxane (15 mL) was added cesium carbonate (3 gm, 9.1 mmol). Argon was
bubbled through the mixture for 10 minutes and then copper (l) iodide (116 mg, 0.61 mmol) and picolinic
acid (150 mg, 1.22 mmol) were added. The ant mixture was heated in a sealed tube at 130 0C for
24 hours. The reaction mixture was cooled to room temperature, quenched with water (10 mL) and
extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with water (2 x 10 mL),
brine (10 mL), dried over sodium sulphate, filtered and concentrated under reduced pressure. The crude
al was purified by silica gel column chromatography eluting with a gradient of dichloromethane:
methanol 100:0 to 98:2 to afford the title compound as an off-white gum in 29% yield, 180 mg.
LCMS (system 10): R: 2.61 min; m/z 205 [M+H]+.
Preparation 131: N-(5-{[7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin-
5-yl]carbonyl}pyridinyl)(4-cyanophenyl)acetamide
W0 2012/137089 172 2012/051363
Prepared according to the method described for Example 1 using (5-aminopyridinyl)[7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]methanone (Preparation 38),
and ophenylacetic acid with DIPEA.
1H NMR (400 MHz, DMSO) 6: -0.26 (s, 6H), 0.58 (s, 9H), 1.75 (s, 6H), 3.86 (s, 2H), 4.10 (s, 2H), 7.54 (d,
2H), 7.82 (d, 2H), 8.16 (s, 1H), 8.51 (s, 1H), 8.68 (s, 1H), 8.88 (s, 1H), 8.98 (s, 1H), 9.47 (s, 1H), 10.73 (s,
1H);
LCMS (System 9): R = 3.80 min; m/z 569 [M+H]+.
Preparation 132: |midazo[1,2-a]pyridinyl-acetic acid
0 /N/\>
HO \N
To a solution of imidazo[1,2-a]pyridinyl-acetic acid ethyl ester (180 mg, 0.65 mmol) (Preparation 130)
in dioxane (4 mL) at 0 0C was added 2 N aqueous sodium hydroxide solution (4 mL). Then reaction
mixture was heated at 900C for 6 hours. After cooling to 0 0C, the e was acidified to pH 4 with with
2 N aqueous hydrochoric acid and extracted with 20% isopropanol in dichoromethane (8 x 10 mL). The
combined organics were dried over sodium e and concentrated under reduced re to afford the
title compound as an off-white solid in 70% yield, 80 mg.
LCMS (system 10): R: 1.40 min; m/z177[M+H]+.
Preparation 133: Pyrazolo[3,4-b]pyridinyl-acetic acid ethyl ester
N N
To a solution of 7-aza indazole (250 mg, 2.1 mmol) and ethyl bromoacetate (0.47 mL, 4.2 mmol) in DMF
(8 mL) was added K2CO3 (1.16 gm, 8.4 mmol) and the resulting mixture stirred at 70 °C for 16 hours. The
reaction mixture was cooled to room temperature and d with ethyl acetate (20 mL). The organic
layer was washed with water (2 x 5 mL), brine (5 mL), dried over sodium sulphate, filtered and
concentrated under reduced pressure. The crude material was purified using silica gel column
chromatography eluting with EtOAc:hexane 10:90 to afford the title compound as an off-white solid in 49%
yield, 210 mg.
W0 2012/137089 173
1H NMR (400 MHz, DMSO-d6) 6: 1.19 (t, 3H), 4.14 (q, 2H), 5.35 (s, 2H), 7.25-7.28 (m, 1H), 8.22 (s, 1H),
8.29 (d, 1H), 8.55 (d, 1H).
Preparation 134: Pyrazolo[3,4-b]pyridinyl-acetic acid
N N
To a stirred solution of pyrazolo[3,4-b]pyridinyl-acetic acid ethyl ester (210 mg, 1.02 mmol)
(Preparation 133) in THF (4 mL) and water (1 mL) at 0 0C was added LiOH.H20 (129 mg, 3.06 mmol).
The on mixture was stirred at room temperature for 2 hours. The pH was of the mixture was
adjusted to pH 4 with 2 N aqueous hydrochoric acid and extracted with 20% isopropanol in
dichloromethane (8 x 5 mL). The combined organics were dried over sodium sulfate, filtered and
concentrated under reduced pressure to afford the title compound as an off-white solid in 40% yield, 70
1H NMR (400 MHz, DMSO-d6) 6: 5.22 (s, 2H), 7.25 (dd, 1H), 8.19 (s, 1H), 8.27 (dd, 1H), 8.55 (dd, 1H),
13.15 (brs, 1H); LCMS (system 9): R: 1.93 min; m/z178[M+H]+.
Preparation 135: 1-Cyclopropyltrifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester
N/ O
V/ O/\Me
4,4,4-Trifluoro—3-oxo-butyric acid ethyl ester (16 g, 86.4 mmol) was dissolved in acetic ide (33.6 g,
329.6 mmol) and yl orthoformate (38.4 g, 260 mmol) was added to the mixture. The resultant mixture
was refluxed for 18 hours. The e was concentrated under reduced pressure to obtain 20 g of 2-[1-
Ethoxy—meth-(E)-ylidene]-4,4,4-trifluorooxo—butyric acid ethyl ester as crude. This was taken in EtOH
(50 mL) and added to a suspension of cyclopropyl hydrazine hydrochloride (9.95 g, 91.7 mmol) and
DIPEA (28.3 ml, 166.7 mmol) in EtOH (150 mL) at -20°C. The resultant mixture was slowly warmed to
room temperature and stirred for 16 hours. The mixture was concentrated under reduced pressure and
residue formed was partitioned n EtOAc (50 mL) and water (50 mL). The organic layer was
washed with 2N HCl (25 mL), water (25 mL), brine (25 mL), dried 4) and evaporated in vacuo. The
crude al was purified by column chromatography on silica gel (EtOAc:Hexane 5:95) to afford the
title compound as off white sticky solid in 7% yield, 1.4 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.10-1.21 (m, 4H), 1.26 (t, 3H), 3.90 (m, 1H), 4.26 (q, 2H), 7.98 (s, 1H).
Preparation 136: N-(5-{[7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin-
5-yl]carbonyl}pyridinyl)[3-(trifluoromethyl)phenyl]acetamide
W0 2012/137089 174
Prepared according to the method described for Example 1 using (5-aminopyridinyl)[7-(2-{[tert-
buty|(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrro|o[2,3-d]pyrimidinyl]methanone (Preparation 38),
and luoromethylphenylacetic acid with DIPEA.
1H NMR (400 MHz, DMSO) 6: -0.27 (s, 6H), 0.56 (s, 9H), 1.75 (s, 6H), 3.87 (s, 2H), 4.09 (s, 2H), 7.57-
7.64
Preparation 137: lopropyltrifluoromethyl-1H-pyrazolyl)—methanol
V OH
A on of 1-Cyclopropyltrifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester (Preparation 135,
1.4 g, 5.64 mmol) in dry toluene (25 mL) was cooled to -78°C and DIBAL-H (11.8 mL of 1.2 M solution in
toluene, 14.1 mmol) was added dropwise to it. The reaction mixture was stirred at -78°C for 2 hours and
poured into 2N HCI (10 mL). This was d for a further 4 hours at room temperature followed by
extraction with EtOAc (2 x 25 mL) and the combined organic layers were washed with water (2 x 10 mL),
brine (10 mL) dried 4) and evaporated in vacuo to afford the title compound as off white solid in
100% yield, 1.2 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.03-1.17 (m, 4H), 3.68-3.73 (m, 1H), 4.42 (d, 2H), 5.15 (t, 1H), 7.51 (s,
1H); LCMS (system 10): R: 2.68 min; m/z 207 [M+H]+
Preparation 138: N-(5-{[7-(2-{[tert-butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)—7H-pyrrolo[2,3-d]pyrimidin-
5-yl]carbonyl}pyridinyl)—2-(4-isopropyl-1H-1,2,3-triazolyl)acetamide.
Prepared according to the method described for Example 1 using (5-aminopyridinyl)[7-(2-{[tert—
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)—7H-pyrrolo[2,3-d]pyrimidinyl]methanone (Preparation 38),
and [4-(trifluoromethyl)-1H-pyrazolyl]acetic acid (Preparation 85) with DIPEA as base.
LCMS (System 9): R = 3.67 min; m/z 577 [M+H]+.
Preparation 139: (1-Cyclopropyltrifluoromethyl-1H-pyrazolyl)-acetonitrile
W0 2012/137089 175
A solution of lopropyltrifluoromethyl-1H-pyrazolyl)—methanol (Preparation 137, 1.2 g, 5.82
mmol) in DCM (15 mL) was cooled to 0°C and thionyl de (0.85 mL, 11.7 mmol) was added. The
reaction e was stirred at 0°C for 2 hours and diluted with DCM. The organic layer was washed with
water, brine and dried (Na2804) and evaporated in vacuo. The crude residue obtained was dissolved in
dioxane (25 mL) and water (25 mL) and tetrabutyl ammonium bromide (1.38 g, 4.28 mmol) was added.
The reaction mixture was stirred for 10 mins ed by the on of KCN (1.28 g, 19.82 mmol) and
resultant mixture was stirred for a further 16 hours at room temperature. The e was diluted with
EtOAc (50 mL) and washed with water (1 x 10 mL), brine (1 x 10 mL), dried (Na2804) and evaporated in
vacuo. The crude material was purified by column chromatography on silica gel (Hexane:EtOAc 10:90) to
afford the title compound as light yellow solid in 56% yield, 700 mg.
1H NMR (400 MHz, CDCI3) 6: 1.06-1.13 (m, 2H), 1.24-1.29 (m, 2H), 3.61-3.62 (m, 1H), 3.66 (s, 2H), 7.49
(s, 1H).
Preparation 140: N-(5-{[7-(2-{[tert—butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin-
5-yl]carbonyl}pyridinyl)quinolinylacetamide
KIN \N/
Prepared according to the method described for Example 1 using (5-aminopyridinyl)[7-(2-{[tert-
butyl(dimethyl)silyl]oxy}-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]methanone (Preparation 38),
and 2-quinolinylacetic acid with DIPEA as base.
1H NMR (400 MHz, DMSO) 6: -0.28 (s, 6H), 0.55 (s, 9H), 1.74 (s, 6H), 3.98 (s, 2H), 4.09 (s, 2H), 7.51 (m,
1H), 7.60 (m, 1H), 7.94 (m, 1H), 8.00 (s, 1H), 8.16 (s, 1H), 8.33 (m, 1H), 8.54 (m, 1H), 8.67 (s, 1H), 8.84
(m, 1H), 8.91 (s, 1H), 8.98 (s, 1H), 9.47 (s, 1H), 10.77 (s, 1H);
LCMS (System 9): R = 3.72 min; m/z 595 [M+H]+.
Preparation 141: (1-Cyclopropyltrifluoromethyl-1H-pyrazolyl)—acetic acid
K 7 OH
F O
To a solution of (1-Cyclopropyltrifluoromethyl-1H-pyrazolyl)-acetonitrile (Preparation 139, 700 mg,
3.25 mmol) in EtOH (15 mL) was added aqueous 1N NaOH (15 mL). The resulting solution was heated at
60°C for 16 hours. The mixture was concentrated and the residue was dissolved in water (10 mL) and
W0 2012/137089 176
washed with EtOAc. The pH of aqueous layer was adjusted to 5 using 1N HCI and extracted with 10%
IPA in DCM (4 x 30 mL). The organic layer was dried (Na2804) and evaporated in vacuo to afford the title
compound as a solid in 85% yield, 650 mg.
1H NMR (400 MHz, DMSO-DG) 6: 1.04-1.07 (m, 2H), 1.11-1.16 (m, 2H), 3.55 (s, 2H), 3.69-3.73 (m, 1H),
7.47 (s, 1H), 12.45 (br, 1H); LCMS (system 10): R: 1.50 min; m/z 233 [M-H]+.
Preparation 142 N-[5-({7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidin-
-yl}carbonyl)pyridinyl][3-(trifluoromethyl)phenyl]acetamide
O CF3
\ / °
KN' \ H
THPO
Prepared according to the method bed above for Example 1, using nopyridinyl){7-[(1R)—1-
methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone ration 36)
and 3-trifluoromethylphenylacetic acid with DIPEA as base.
1H NMR (400 MHz, DMSO-d6) 6: 1.29-1.42 (m, 5H), 1.54 ( d, 3H), 3.50 (m, 1H), 3.71 (m, 1H), 3.86 (s,
2H), 3.93 (m, 1H), 4.05 (m, 1H), 4.44 (s, 1H), 4.58 (s, 1H), 5.17 (m, 1H), 7.56 (m, 1H), 7.63 (m, 2H), 7.72
(s, 1H), 8.47 (m, 2H), 8.70 (s, 1H), 8.96 (m, 2H), 9.45 (s, 1H), 10.72 (s, 1H);
LCMS (System 9): R = 3.56 min; m/z 568 [M+H]+.
Preparation 143: 5-Trifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester
.N’ 0
H,N /
F Me
To a suspension of hydrazine hydrochloride (10 g, 147 mmol) in EtOH (500 mL), DIPEA (45.3 mL, 267
mmol) was added slowly at -200C and stirred for 10 mins. Then 2-[1-Ethoxy-meth-(E)-ylidene]-4,4,4-
trifluorooxo-butyric acid ethyl ester (Preparation 135, 32 g, 133.33 mmol) was added to above solution
and the resulting mixture was stirred at room temperature for 16 hours. The reaction mixture was
concentrated under reduced pressure and residue was ioned between EtOAc (200 mL) and water
(50 mL). The organic layer was washed with water (25 mL), dried (Na2804) and evaporated in vacuo. The
crude material was purified by column chromatography on silica gel (Hexane:EtOAc 90:10) to afford the
title nd as off white solid in 43% yield, 13 g.
1H NMR (400 MHz, DMSO-DG) 6: 1.26 (t, 3H), 4.25 (q, 2H), 8.57 (s, 1H), 14.10 (br s, 1H).
Preparation 144: N-[5-({7-[(1R)- 1-methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-
d]pyrimidinyl}carbonyl)pyridinyl]—2-(4-isopropyl-1H-1,2,3-triazolyl)acetamide
/N Me
\ / 0 Me
N/ \ ”kw ¢N
k ' N
\N N
THPO
Prepared according to the method described above for e 1, using (5-aminopyridinyl){7-[(1R)—1-
methyl(tetrahydro-2H-pyranyloxy)ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}methanone (Preparation 36)
and (4-isopropyl-1H-1,2,3-triazolyl)acetic acid with DIPEA as base.
1H NMR (400 MHz, DMSO-de) 6: 1.22 (6H, d), 1.23-1.30 (m, 5H), 1.54(d, 3H), 3.4 (1H, m), 3.72 (m, 1H),
3.88 (m, 1H), 3.95 (m, 1H), 4.10 (m, 1H), 4.45 (s, 1H), 4.59 (s, 1H), 5.20 (1H, m), 5.36 (s, 2H), 7.88 (s,
1H), 8.43 (m, 1H), 8.53 (d, 1H), 8.75 (s, 1H), 8.97 (m, 2H), 9.45 (s, 1H), 10.96 (s, 1H);
LCMS (System 9): R = 2.86 min; m/z 533 [M+H]+.
Preparation 145: 1-Cyclopropyltrifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester
it \ O
FF F Me
Cyclopropyl boronic acid (11 g, 127 mmol), Copper e (17.4 g, 95.7 mmol), Pyridine (17.7 g, 223
mmol) and triethylamine (22.4 mL, 160 mmol) were added successively to a solution of 5-Trifluoromethyl-
1H-pyrazole—4-carboxylic acid ethyl ester (Preparation 143, 6.63 g, 31.9 mmol) in THF (70 mL) and the
ing mixture was allowed to stir at 600C for 36 hours. The reaction mixture was filtered over a celite
bed and filtrate was concentrated in vacuo and diluted with EtOAc (200 mL). The organic layer was
washed with 1N HCI (1 x 25 mL), brine (1 x 25 mL) and dried (Na2804) and evaporated in vacuo. The
crude material was purified by column chromatography on silica gel (Hexane:EtOAc 85:15) to afford the
title compound as brown solid in 29% yield, 2.3 g.
1H NMR (400 MHz, CDCI3) 6: 1.08-1.14 (m, 2H), 1.17-1.21 (m, 2H), 1.33 (t, 3H), .67 (m, 1H), 4.30
(q, 2H), 8.01 (s, 1H); LCMS (system 10): R: 3.39 min; m/z 249 [M+H]+.
Preparation 146: lopropyltrifluoromethyl-1H-pyrazolyl)—methanol
A solution of 1-Cyclopropyltrifluoromethyl-1H-pyrazolecarboxylic acid ethyl ester (Preparation 145,
3.5 g, 14.11 mmol) in dry toluene (70 mL) was cooled to -78°C and DIBAL-H (29.4 mL of a 1.2 M solution
in toluene, 35.3 mmol) was added dropwise to it. The reaction mixture was stirred at -78°C for 2 hours and
then poured into 2N HCI (25 mL) followed by r stirring for 2 hours at room temperature. The mixture
W0 2012/137089 178
was extracted with EtOAc (2 x 50 mL) and the combined organic layers were washed with water (2 x 15
mL), brine (15 mL) and dried (NaZSO4) and evaporated in vacuo to afford the title compound as off white
solid in 100% yield, 3 g.
1H NMR (400 MHz, CDCI3) 6: 1.02-1.07 (m, 2H), 1.11-1.16 (m, 2H), 1.68 (t, 1H), 3.57-3.63 (m, 1H), 4.64
(d, 2H), 7.53 (s, 1H); LCMS (system 10): R: 2.57 min; m/z 207 .
Preparation 147: (1-Cyclopropyltrifluoromethyl-1H-pyrazolyl)-acetonitrile
The title compound was prepared according to the method described for Preparation 139 using (1-
Cyclopropyltrifluoromethyl-1H-pyrazolyl)—methanol (Preparation 146) to afford the title compound as
yellow solid in 70% yield, 2.2 g.
1H NMR (400 MHz, DMSO-DG) 6: 0.98-1.03 (m, 2H), 1.06-1.11 (m, 2H), 3.83-3.88 (m, 1H), 3.91 (s, 2H),
8.08 (s, 1H); LCMS (system 10): R: 3.10 min; m/z 216 [M+H]+.
Preparation 148: (1-Cyclopropyltrifluoromethyl-1H-pyrazolyl)—acetic acid
N\ / OH
F F
F O
The title nd was prepared according to the method described for ation 141 using (1-
Cyclopropyltrifluoromethyl-1H-pyrazolyl)-acetonitrile (Preparation 147) to afford the title compound
as a solid in 79% yield, 1.9 g.
1H NMR (400 MHz, DMSO-DG) 6: 0.96-1.07 (m, 4H), 3.49 (s, 2H), 3.76-3.84 (m, 1H), 7.91 (s, 1H), 12.27
(br, 1H); LCMS (system 10): R: 1.41 min; m/z 233 [M-H]+.
Preparation 149: (7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)[5-(methylamino)pyridinyl]methanone
HaMe
W0 2012/137089 179
The title compound was prepared according to the method bed for Preparation 31 using (5-
bromopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 94) and
methylamine (20 mL) in 13% yield, 78 mg.
LCMS (system 2). R: 0.91 min; m/z 296 [M+H]
The following Preparations were prepared according to the method described above for Example 1, using
(5-aminopyridinyl){7-[(1R)—1-methyl(tetrahydro-2H-pyranyloxy)ethyl]—7H-pyrrolo[2,3-d]pyrimidin
yl}methanone ration 36) and the corresponding carboxylic acid with DIPEA. All carboxylic acids
are commercially available unless othenNise mentioned.
O / o
”’K/R102
——-ii-—
({7-[(1R)— 1--m-ethy|2-- LCMS (System 9). R:
3.24 min; m/z 600
(tetrahydro-2H-pyran [M+H]+
yloxy)ethyl]-7H-pyrrolo[2,3- Using (Prep 185).
d]pyrimidinyl}carbonyl)pyridin
y|][1-isopropyl
(trifluoromethyl)—1H-pyrazol
yl]acetamide
2-(4-cyanophenyl)-N-[5-({7-[(1R)- LCMS (System 9): R =
2.94 min; m/z 525
1-methyl(tetrahyd ro-2H-pyran .
yloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}carbonyl)pyridin
yl]acetamide
N-[5-({7-[(1R)- 1-methyI LCMS (System 9): R =
2.82 min; m/z 578
(tetrahydro-2H-pyran [M+H]+
yloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}carbonyl)pyridin
y|][3-
(methylsulfonyl)phenyl]acetamide
{7-[(1R)- 1-methyI LCMS (System 9): R =
2.97 min; m/z 551
hydro-2H-pyran [M+H]+.
yloxy)ethyl]-7H-pyrrolo[2,3-
d]pyrimidinyl}carbonyl)pyridin
y|]quinolinylacetamide
W0 2012/137089 180
Preparation 154: Ethyl (2-cyclopropyl-1,3-oxazolyl)acetate
Ethyl 4-chloroacetoacetate (20.0 g, 122.0 mmol) was added to cyclopropanecarboxamide (3.52 g, 41.5
mmol) in toluene (100 mL) and 1,4-dioxane (100 mL). The mixture was refluxed at 120°C for 17 hours
then evaporated in vacuo. The crude solid was purified by column chromatography on silica gel (80:20
petroleum ether: EtOAc) to afford the title nd as a white solid in 50% yield, 4.00 g.
1H NMR (300 MHz, DMSO-de) 6: 0.80-1.00 (m, 4H), 1.20 (t, 3H), 2.10 (m, 1H), 3.50 (s, 2H), 4.10 (q, 2H),
7.80 (s, 1H).
Preparation 155: lopropyl-1,3-oxazoly|)acetic acid
Lithium hydroxide monohydrate (7.83 g, 186.7 mmol) was added to ethyl (2-cyclopropyl-1,3-oxazol
yl)acetate (Preparation 154, 7.00 g, 35.9 mmol) in THF (200 mL) and water (100 mL). The mixture was
stirred at room temperature for 2 hours then the reaction e volume was reduced to one third by
evaporation in vacuo. The aqueous residue was ied using aqueous HCl (1.0 M) then extracted with
EtOAc (200 mL). The organic phase was evaporated in vacuo and the crude material was ated with
diethyl ether (100 mL) to afford the title compound as a white solid in 66% yield, 4.00 g.
1H NMR (300 MHz, CDCI3) 6: 1.05 (m, 4H), 2.10 (m, 1H), 3.60 (s, 2H), 7.40 (s, 1H), 10.00 (brs, 1H).
Preparation 156: Methyl [2-(vinyloxy)phenyl]acetate
Copper acetate (1.42 g, 7.82 mmol) was added to DCM (6 mL) and stirred for 20 minutes, with a drying
tube attached. Trivinylcycloboroxane (1.24 g, 5.16 mmol), cesium carbonate (2.55 g, 7.82 mmol) and
methyl 2-hydroxyphenyl acetate (1.30 g, 7.82 mmol) were added and the mixture d at room
temperature for 17 hours. Saturated aqueous sodium bicarbonate (25 mL) was added and the mixture
was extracted with DCM (20 mL). The organic phase was filtered, dried over magnesium sulphate and
evaporated in vacuo to afford the title compound as a black oil in 52% yield, 784 mg. This material was
used crude in subsequent steps.
W0 2012/137089 181
1H NMR (400 MHz, CDCI3) 6: 3.67 (s, 2H), 3.39 (s, 3H), 4.40 (dd, 1H), 4.69 (dd, 1H), 6.59 (dd, 1H), 6.97-
7.08 (m, 2H), 7.23-7.28 (m, 2H).
Preparation 157: Methyl [4-(vinyloxy)phenyl]acetate
H2C=\0%
The title compound was prepared according to the method described for ation 156 using methyl 4-
hydroxyphenyl acetate to afford the title compound as a colourless oil in 70% yield, 914 mg.
1H NMR (400 MHz, CDCI3) 6: 3.58 (s, 2H), 3.39 (s, 3H), 4.42 (m, 1H), 4.75 (m, 1H), 6.62 (m, 1H), 6.94-
6.97 (m, 2H), 7.21-7.24 (m, 2H).
Preparation 158: Methyl [3-(vinyloxy)phenyl]acetate
The title compound was prepared according to the method described for Preparation 156 using methyl 3-
hydroxyphenyl acetate to afford the title compound as a colourless oil in 56% yield, 835 mg.
1H NMR (400 MHz, CDCI3) 6: 3.61 (s, 2H), 3.70 (s, 3H), 4.44 (m, 1H), 4.78 (m, 1H), 6.63 (m, 1H), 6.89-
6.94 (m, 2H), 7.00 (m, 1H), 7.27 (m, 1H).
Preparation 159: Methyl [5-(vinyloxy)pyridinyl]acetate
The title compound was prepared ing to the method described for Preparation 156 using methyl
(5-hydroxypyridinyl)acetate to afford the title compound as a yellow oil in 25% yield, 76 mg.
LCMS m 2): R = 0.85 min; m/z 194 [M+H]+.
W0 2012/137089 182
Preparation 160: [2-(Cyclopropyloxy)phenyl]acetic acid
Diethyl zinc (2.34 mL, 2.34 mmol, 1M in e) was cooled to 0°C under nitrogen then
chloroiodomethane (0.35 mL, 4.68 mmol) in DOE (1 mL) was added drop-wise. The mixture was stirred
under nitrogen at 0°C for 20 minutes then methyl [2-(vinyloxy)phenyl]acetate (Preparation 156, 150 mg,
0.78 mmol) in DOE (1 mL) was added. The reaction mixture was stirred at 0°C for 30 minutes then stirred
at room temperature for 17 hours. Saturated aqueous ammonium chloride (10 mL) was added and the
mixture was extracted with DCM (4 x 8 mL). The ed organic phases were dried over magnesium
sulphate and evaporated in vacuo. Sodium hydroxide (3.28 mL, 3.28 mmol, 1 M) was added to the
residue in THF (3 mL). The mixture was heated at 80°C for 17 hours then ated in vacuo.
Hydrochloric acid (10 mL, 1M) was added to the residue then extracted with EtOAc (10 mL). The organic
phase was evaporated in vacuo to afford the title compound as a yellow oil in 53% yield, 83 mg.
1H NMR (400 MHz, CDCI3) 6: 0.65-0.71 (m, 4H), 3.54 (s, 2H), 3.71 (m, 1H), 6.82-6.87 (m, 2H), 7.05-7.23
(m, 2H).
Preparation 161: [4-(Cyclopropyloxy)phenyl]acetic acid
ed according to the method described for Preparation 160 using methyl [4-
(vinyloxy)phenyl]acetate (Preparation 157) to afford the title compound as a colourless oil in 44% yield,
360 mg.
1H NMR (400 MHz, CDCI3) 6: 0.65-0.71 (m, 4H), 3.59 (s, 2H), 3.71 (m, 1H), 6.99-7.02 (m, 2H), 7.16-7.20
(m, 2H).
W0 2012/137089 183
Preparation 162: [3-(Cyclopropyloxy)phenyl]acetic acid
The title compound was prepared according to the method described for ation 160 using methyl
[3-(vinyloxy)phenyl]acetate (Preparation 158) to afford the title compound as a colourless oil in 33%
yield, 50 mg.
1H NMR (400 MHz, CDCI3) 6:0.74—0.81 (m, 4H), 3.57—3.67 (m, 2H), 3.72 (m, 1H), 6.74-7.03 (m, 3H),
7.22 (m, 1H).
Preparation 163: clopropyloxy)pyridinyl]acetic acid
The title compound was prepared according to the method described for Preparation 160 using methyl
nyloxy)pyridinyl]acetate (Preparation 159) to afford the title compound as a yellow oil in 38%
yield, 28 mg.
LCMS (system 2): R = 0.55 min; m/z 194 [M+H]+.
ation 164: [4-Cyano(trifluoromethyl)phenyl]acetic acid
Lithium diisopropylamide (13.8 mL, 24.8 mmol, 1.8M in THF) was added to 4-methyl
(trifluoromethyl)benzonitrile (2.30 g, 12.4 mmol) in THF (20 mL) at -78°C and stirred for 5 minutes at -
78°C. Excess solid carbon dioxide was added then the mixture was stirred at room temperature for 17
hours. Saturated aqueous ammonium chloride (10.5 mL) and EtOAc (20 mL) were added then the
W0 2012/137089 184
aqueous layer was acidified with HCI acid on (1 M). This was extracted with EtOAc (3 x 15 mL) and
the ed organic phases were dried over sodium sulphate and evaporated in vacuo to afford the title
compound as a brown oil in 88% yield, 2.52 g.
1H NMR (400 MHz, CDCI3) 6: 3.81 (s, 2H), 7.62 (d, 1H), 7.73 (s, 1H), 7.83 (d, 1H).
Preparation 165: (2-Methquuinolinyl)acetic acid
N Me
ic aldehyde (33.0 mL, 0.40 mol) was added to 3-aminophenylacetic acid (30.0 g, 0.20 mmol) in
concentrated hydrochloric acid (400 mL) and toluene (100 mL) at 110°C. The mixture was heated at
110°C for 90 minutes. The aqueous layer was separated, washed with diethyl ether (350 mL) then
neutralised with aqueous ammonia. The aqueous solution was washed with chloroform (3 x 500 mL) and
the organic phase was evaporated in vacuo. The solid residue was ed with chloroform (900 mL) and
methanol (100 mL) then the solution was decanted and purified by column chromatography on silica gel
(gradient of chloroform:MeOH 9:1 to 4:1) to afford a mixture of isomeric acids. This was purified by
fractional crystallisation using isopropanol to afford the title compound as a white solid in 12% yield, 4.90
g.
1H NMR (400 MHz, DMSO-d6) 6: 2.64 (s, 3H), 3.78 (s, 2H), 7.37 (d, 1H), 7.44 (dd, 1H), 7.80 (s, 1H), 7.84
(d, 1H), 8.19 (d, 1H), 12.40 (brs, 1H).
Preparation 166: Ethyl (3-isopropylmethyl-1H-pyrazolyl)acetate
Mew\,N N
Me 1
o O\\
Ethyl bromoacetate (1.00 mL, 9.03 mmol) was added to 5-isopropylmethyl-1H-pyrazole (1.07 g, 8.60
mmol) and potassium carbonate (3.57 g, 25.9 mol) in DMF (10 mL). The mixture was d at room
temperature for 17 hours then EtOAc (20 mL) and s HCI (20 mL, 1 M) were added. The organic
phase was dried over magnesium sulphate then evaporated in vacuo. The residue was purified by column
chromatography on silica gel (hexane:EtOAc 4:1) to afford the title compound as a yellow oil in 34% yield,
607 mg.
1H NMR (400 MHz, CDCI3) 6: 1.26 (m, 9H), 2.03 (s, 3H), 2.96 (m, 1H), 4.21 (q, 2H), 4.78 (s, 2H), 7.13 (s,
1H).
W0 2012/137089 185
Preparation 167: (3-lsopropylmethyl-1H-pyrazolyl)acetic acid
Mew\ /NN
Lithium hydroxide (342 mg, 8.15 mmol) in water (4 mL) was added to ethyl (3-isopropylmethyl-1H-
pyrazolyl)acetate (Preparation 166, 571 mg, 2.72 mmol) in methanol (4 mL) and the mixture was
stirred at room temperature for 30 minutes. Aqueous hloric acid (2 M) was added to acidify the
mixture then the solution was extracted with EtOAc (10 mL). The c phase was dried over
magnesium sulphate and evaporated in vacuo to afford the title compound as a cream solid in 66% yield,
328 mg.
m/z 183 [M+H]+.
Preparation 168: Ethyl (6-chloro-1H-indazolyl)acetate
\ f
’N Me
Concentrated sulphuric acid (0.25 mL) was added to a on of 2-(6-chloro-1H-indazolyl)acetic acid
(2.024 g, 9.60 mmol) in EtOH (10 mL). The mixture was heated at 80°C for 4 hours then evaporated in
vacuo. The residue was partitioned between EtOAc (30 mL) and 5% aqueous sodium bicarbonate (30
mL). The c phase was dried over sodium sulphate, evaporated in vacuo and purified by column
chromatography on silica gel (DCM:MeOH 99:1) to afford the compound 3 as a white solid in 83% yield,
1.90 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.17 (t, 3H), 4.00 (s, 2H), 4.10 (q, 2H), 7.11 (d, 1H), 7.56 (s, 1H), 7.74
(d, 1H), 13.00 (s, 1H).
Preparation 169: Ethyl (6-chloromethyl-1H-indazolyl)acetate
N (
NI Me
Sodium hydride (24 mg, 0.602 mmol, 60% in oil) was added to ethyl (6-chloro-1H-indazolyl)acetate
(Preparation 168, 120 mg, 0.502 mmol) in THF (4 mL) at 0°C and the mixture was stirred for 30 minutes.
Then iodomethane (0.09 mL, 1.508 mmol) was added. The mixture was stirred at room temperature for 30
mins then water (4 mL) was added. The e was ated in vacuo and the aqueous residue was
acidified with aqueous HCl (6 M). This was extracted with ethyl acetate (10 mL) and the organic phase
W0 2012/137089 186
was dried over sodium sulphate and evaporated in vacuo to afford the title compound as a white solid in
100% yield, 150 mg.
1H NMR (400 MHz, DMSO-d6) 6: 1.18 (t, 3H), 3.89 (s, 3H), 4.07 (s, 2H), 4.13 (q, 2H), 7.15 (d, 1H), 7.56 (s,
1H), 7.74 (d, 1H);
LCMS (System 9): R =3.48 min; m/z 253 [M+H]+.
Preparation 170: (6-Chloromethyl-1H-indazolyl)acetic acid
CI "l
Aqueous potasium hydroxide (5.93 mL, 10%) was added to ethyl (6-chloromethyl-1H-indazol
yl)acetate (Preparation 169, 1.78 g, 0.704 mol) in MeOH (30 mL). The e was stirred for 1 hour at
25°C then the methanol was evaporated in vacuo. The aqueous residue was washed with l ether
(30 mL) then acidified with aqueous HCl (6 M). The mixture was extracted with ethyl acetate (30 mL) and
the organic phase was dried over sodium sulphate, evaporated in vacuo and purified by column
chromatography on silica gel (DCM:MeOH 95: 5 ) to afford the title compound as a white solid in 56%
yield, 865 mg.
1H NMR (400 MHz, DMSO-d6) 6: 3.79 (s, 2H), 4.14 (s, 3H), 7.12 (d, 1H), 7.73 (d, 1H), 7.83 (s, 1H). 12.51
(s, 1H).
Preparation 171: Ethyl (5-fluoro-1H-indazolyl)acetate
QMN 4..
The title compound was ed according to the method bed for Preparation 93 using 5-fluoro-
1H-indazole and ethyl bromoacetate to afford the title compound as an off- white solid in 53% yield, 260
1H NMR (400 MHz, CDCI3) 6: 1.24 (t, 3H), 4.22 (q, 2H), 5.13 (s, 2H), 7.17 (m, 1H), 7.28 (m, 1H), 7.37 (d,
1H), 7.99 (s, 1H); LCMS (System 9): R: 3.14 min; m/z 223 .
Preparation 172: (5-Fluoro-1H-indazolyl)acetic acid
W0 37089 187
The title compound was prepared according to the method described for Preparation 155 using ethyl (5-
-1H-indazolyl)acetate (Preparation 171) to afford the title compound as a yellow solid in 62%
yield, 140 mg.
1H NMR (400 MHz, DMSO-d6) 6: 5.26 (s, 2H), 7.28 (m, 1H), 7.54 (dd, 1H), 7.68 (dd, 1H), 8.06 (s, 1H),
13.11 (brs, 1H).
LCMS (System 9): R: 1.49 min; m/z193[M-H]'
ation 173: Ethyl (5-fluoro-2H-indazolyl)acetate
F —>/—O
0 \—Me
The title compound was prepared according to the method described for Preparation 93 using 5-fluoro-
1H-indazole and ethyl bromoacetate to afford the title compound as an off- white solid in 27% yield, 130
1H NMR (400 MHz, CDCI3) 6: 1.26 (t, 3H), 4.25 (q, 2H), 5.17 (s, 2H), 7.06-7.11 (m, 1H), 7.21-7.24 (m,
1H), 7.64-7.68 (m, 1H), 7.96 (s, 1H); LCMS (System 9): R: 3.04 min; m/z 223 [M+H]+
Preparation 174:(5-F|uoro-2H-indazolyl)acetic acid
The title compound was prepared according to the method described for Preparation 155 using ethyl (5-
fluoro-2H-indazolyl)acetate (Preparation 173) to afford the title compound as a yellow solid in 100%
yield, 160 mg.
LCMS m 9): R: 1.49 min; m/z193[M-H]'
ation 175: Ethyl (7-fluoro-1H-indazolyl)acetate
The title compound was prepared according to the method described for Preparation 93 using 7-fluoro-
1H-indazole and ethyl bromoacetate to afford the title compound as an off- white solid in 41% yield, 200
1H NMR (400 MHz, CDCI3) 6: 1.29 (t, 3H), 4.22 (q, 2H), 5.28 (s, 2H), 6.98-7.07 (m, 2H), 7.49 (m, 1H), 8.02
(d, 1H).
Preparation 176: (7-Fluoro—1H-indazolyl)acetic acid
W0 2012/137089 188
The title compound was prepared according to the method described for Preparation 155 using ethyl (7-
fluoro-1H-indazolyl)acetate (Preparation 175) to afford the title nd as a yellow solid in 68%
yield, 120 mg.
1H NMR (400 MHz, DMSO-d6) 6: 5.27 (s, 2H), 7.12 (m, 1H), 7.23 (m, 1H), 7.60 (d, 1H), 8.17 (d, 1H),
13.18 (br s, 1H); LCMS (System 9):
R: 1.38 min; m/z195[M+H]+
Preparation 177: Ethyl (7-fluoro-2H-indazolyl)acetate
N_>—O
0 \—Me
The title compound was prepared according to the method described for Preparation 93 using 7-fluoro-
1H-indazole and ethyl bromoacetate to afford the title compound as an off- white solid in 35% yield, 175
1H NMR (400 MHz, CDCI3) 6: 1.28 (t, 3H), 4.25 (q, 2H), 5.28 (s, 2H), 6.93 (m, 1H), 6.99 (m, 1H), 7.43 (d,
1H), 8.06 (d, 1H).
Preparation 178: (7-Fluoro-2H-indazolyl)acetic acid
The title nd was prepared according to the method described for ation 155 using ethyl (7-
fluoro-2H-indazolyl)acetate (Preparation 177) to afford the title compound as a yellow solid in 75%
yield, 110 mg.
1H NMR (400 MHz, DMSO-d6) 6: 5.34 (s, 2H), 6.97-7.05 (m, 2H), 7.56 (d, 1H), 8.50 (d, 1H), 13.30 (br s,
1H); LCMS (System 9): R: 1.40 min; m/z195[M+H]+
ation 179: Ethyl 1H-pyrazolo[3,4-b]pyridinylacetate
W0 37089 189
The title compound was prepared ing to the method described for Preparation 93 using 1H-
pyrazolo[3,4-b]pyridine and ethyl bromoacetate to afford the title compound as an off- white solid in 49%
yield, 210 mg.
1H NMR (400 MHz, DMSO-d6) 6: 1.19 (t, 3H), 4.14 (q, 2H), 5.35 (s, 2H), 7.27 (m, 1H), 8.27 (s, 1H), 8.29
(d, 1H), 8.55 (d, 1H).
Preparation 180: 1H-Pyrazolo[3,4-b]pyridinylacetic acid
///<OH
\ /
The title compound was prepared according to the method described for Preparation 155 using ethyl 1H-
pyrazolo[3,4-b]pyridinylacetate (see Preparation 179) to afford the title compound as a yellow solid in
40% yield, 70 mg.
1H NMR (400 MHz, DMSO-d6) 6: 5.22 (s, 2H), 7.25 (dd, 1H), 8.19 (s, 1H), 8.27 (dd, 1H), 8.55 (dd, 1H),
13.15 (brs, 1H).
Preparation 181: tert—Butyl 1H-indazolylacetate
/ 0 Me
N 0JVMe
H Me
6-Bromo-1H-indazole (1.3 g, 6.6 mmol) and t—butylacetate (1.33 mL, 9.9 mmol) in toluene (20 mL) were
degassed with argon for 15 mins. Then the mixture was cooled to 0°C and LiHMDS (16.5 mL, 16.5 mmol,
1M in hexane) was added dropwise. Bis(dibenzylideneacetone)palladium (380 mg, 0.66 mmol) and tri-t—
butyl ine luoroborate (383 mg, 1.32 mmol) were added and the mixture was stirred at 10°C
for 2 hours. The mixture was quenched with water (10 mL) then extracted with EtOAc (3 x 25 mL). The
combined organic phases were washed with water (2 x 10 mL), brine (10 mL) and dried over sodium
te (Na2804). The filtrate was evaporated in vacuo and purified by column chromatography on silica
gel (hexane:EtOAc 80:20) to afford the title compound as a white solid (65%, 1.00 g).
1H NMR (400 MHz, CDCI3) 6: 1.43 (s, 9H), 3.64 (s, 2H), 7.08 (dd, 1H), 7.39 (s, 1H), 7.69 (d, 1H), 8.03 (s,
1H), 10.05 (brs, 1H).
Preparation 182: 1H-lndazolylacetic acid
/ O
N OH
Hydrochloric acid (10 mL, 4 M in 1.4 dioxane) was added to tert—butyl 1H-indazolylacetate
(Preparation 181, 1.00 g, 4.3 mmol) in 1,4 dioxane (5 mL) at 0°C and the mixture was stirred at room
W0 2012/137089 190
temperature for 16 hours. The mixture was evaporated in vacuo and the residue was triturated with dry
ether to afford the title compound as a white solid in 100% yield, 800 mg.
1H NMR (400 MHz, CDCI3) 6: 3.69 (s, 2H), 7.00 (d, 1H), 7.41 (s, 1H), 7.67 (d, 1H), 8.02 (s, 1H), 12.81 (br
s, 1H).
Preparation 183: [1-lsopropyl(trifluoromethyl)-1H-pyrazolyl]methanol
F N_'{'
Diisobutylaluminium hydride (99 mL, 120 mmol, 1.2 M solution in e) was added to ethyl 1-isopropyl-
-(trifluoromethyl)-1H-pyrazolecarboxylate (WO 2007071900, 12 g, 48 mmol) in toluene (220 mL) at -
78°C . The reaction e was stirred at -78°C for 2 hours then poured into s HCI (100 mL, 2 M).
The mixture was stirred for 4 hours at room temperature then extracted with EtOAc (400 mL). The organic
phase was washed with water (200 mL), brine (200 mL) and dried over sodium te. The filtrate was
ated in vacuo to afford the title compound as a colourless oil in 100% yield, 10.5 g.
1H NMR (400 MHz, CDCI3) 6: 1.51 (d, 6H), 4.57-4.66 (m, 3H), 7.58 (s, 1H).
1H NMR (400 MHz, CDCI3) 6: 1.26 (s, 4H), 3.83 (s, 2H), 6.47 (d, 1H), 7.26 (d, 1H), 8.83 (s, 1H), 8.88 (s,
1H); LCMS (system 10): R = 1.67 min; m/z189.9[M+H]+.
Preparation 184: [1-lsopropyl(trifluoromethyl)-1H-pyrazolyl]acetonitrile
Thionyl chloride (5.26 mL, 72 mmol) was added to [1-isopropyl(trifluoromethyl)-1H-pyrazol
yl]methanol (Preparation 183, 7.5 g, 36 mmol) in DCM (75 mL) at 0 °C and the mixture was stirred for 2
hours. The mixture was d with DCM (30 mL) and the organic phase was washed with water (75 mL),
brine (75 mL) and dried over sodium te. The filtrate was evaporated in vacuo to afford 4-
(chloromethyl)isopropyl(trifluoromethyl)-1H-pyrazole in 86% yield, 7 g.
Tetrabutyl ammonium bromide (7.95 gm, 24.7 mmol) was added to 4-(chloromethyl)isopropyl
(trifluoromethyl)—1H-pyrazole (7 g, 31 mmol) in dioxane (75 mL) and water (75 mL) and the mixture was
stirred for 10 min. Potassium cyanide (7.42 g, 114 mmol) was added and the mixture was stirred for 16
hours at room temperature. The mixture was diluted with EtOAc (100 mL) then the organic phase was
washed with water (100 mL), brine (100 mL) and dried over sodium sulphate. The filtrate was evaporated
W0 2012/137089 191
in vacuo and purified by column chromatography on silica gel (hexane:EtOAc 90:10) to afford the title
compound as a white solid in 100% yield, 7.00 g.
1H NMR (400 MHz, DMSO-d6) 6: 1.43 (d, 6H), 3.99 (s, 2H), 4.61 (m, 1H), 7.71 (s,1H).
ation 185: [1-lsopropyl(trifluoromethyl)-1H-pyrazolyl]acetic acid
F \\
Aqueous sodium hydroxide (150 mL of a 1 M solution) was added to [1-isopropyl(trifluoromethyl)-1H-
pyrazolyl]acetonitrile (Preparation 184, 6.2 g, 28.6 mmol) in EtOH (150 mL) and the e was
heated at 60 °C for 16 hours. The mixture was evaporated in vacuo and the residue was dissolved in
water (50 mL) then washed with EtOAc (100 mL). The aqueous phase was ied to pH 5 using 1N HCI
and extracted with 10% IPA in DCM (4 x 100 mL). The combined organic phases were dried over sodium
sulphate and evaporated in vacuo to afford the title compound as a white solid in 75% yield, 5.0 g.
1H NMR (400 MHz, DMSO-de) 6: 1.42 (d, 6H), 3.56 (s, 2H), 4.58 (m, 1H), 7.57 (s, 1H), 12.28 (br s, 1H).
Preparation 186: no-pyridinyl)-[7-(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]-methanone
N \ \
lN/ N
Me%\/OMe
The title compound was prepared according to the method described for Preparation 65 using (5-Bromo—
pyridinyl)-[7-(2-methoxy-1 ,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]-metha none (Preparation
263) to afford the title compound as off white solid in 41% yield, 650 mg.
1H NMR (400 MHz, DMSO-D6) 6 1.76 (s, 6H), 3.19 (s, 3H), 3.96 (s, 2H), 5.65 (s, 2H), 7.30 (s, 1H), 8.05
(s, 1H), 8.16 (s, 2H), 8.97 (s, 1H), 9.44 (s, 1H); LCMS (system 10): R = 2.56 min; m/z 327 [M+H]+.
Preparation 187: (7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidinyl)—(5-methylamino-pyridinyl)-methanone
W0 2012/137089 192
N—Me
o /\
N|\ \
kN/ N
[5-(7-tert-Butyl-7H-pyrrolo[2,3-d]pyrimidine-5—carbonyl)—pyridiny|]-methy|-carbamic acid tert-butyl ester
(Example 542) was treated with 4N HCI in Dioxane at room temperature for 2 hours. The solvent was
removed in vacuo and the solid obtained was triturated with diethyl ether to afford the title compound as
white solid in 100% yield, 76 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.80 (s, 9H), 2.84 (s, 3H), 7.27 (br s, 1 H), 8.21 (d, 1H), 8.29 (s, 1H),
8.41 (s, 1H), 9.03 (s, 1H), 9.49 (s, 1H); LCMS m 10): R = 2.74 min; m/z 310 .
Preparation 188: 1,5—naphthyridinylacetic acid
N\ \
0 I
/ /
A mixture of 3-bromo-1,5-naphthyridine (540 mg, 2.58 mmol), diethylmalonate (0.8 mL, 5.17 mmol) and
cesium carbonate (2.53 g, 7.75 mmol) in 1,4-dioxane (6 mL) was degassed with argon for 15 min then
picolinic acid (64 mg, 0.517 mmol) and Cul (50 mg, 0.258 mmol) was added. The mixture was heated in a
sealed tube at 110 °C for 24 hours then cooled to room temperature, diluted with ethyl acetate (10 mL),
washed with water (10 mL) and brine (10 mL). The organic phase was dried over sodium sulphate then
evaporated in vacuo and purified by column chromatography on silica gel (hexane:EtOAc 70:30) to afford
450 mg of ethyl phthyridinylacetate.
Sodium hydroxide (10 mL, 2.0 M) was added to ethyl 1,5-naphthyridinylacetate in 1,4-dioxane (10 mL)
and the mixture was heated at 100 °C for 6 hours. The reaction mixture was cooled to room temperature
and acidified to pH 4 using aqueous 2M HCI and then evaporated in vacuo. The residue was azeotroped
with toluene (2 x 15 mL) then dissolved in THF (50 mL) and d at 40 °C for 30 min. The mixture was
filtered, the filtrate was evaporated in vacuo and the residue was triturated with diethyl ether to afford the
title compound as an off white solid, 200 mg.
1H NMR (400 MHz, DMSO-da) 6: 3.94 (s, 2H), 7.76 (m, 1H), 8.31 (s, 1H), 8.42 (d, 1H), 8.91 (d, 1H), 8.99
(dd, 1H), 12.65 (brs, 1H); LCMS m 10): R = 1.49 min; m/z187[M-H]+
Preparation 189: Tert-butyl 1H-pyrazo|o[4,3-b]pyridiny|acetate
W0 2012/137089 193
/ \
N\ |
Me 0V
Tert—butyl bromoacetate (0.55 mL, 3.69 mmol) was added to 1H-pyrazolo[4,3-b]pyridine (220 mg, 1.85
mmol) and Cs2CO3 (723mg, 2.22 mmol) in anhydrous DMF (7 mL). The e was stirred at room
temperature for 1 hour. The reaction mixture was diluted with ethyl acetate (50 mL), washed with water (3
x 30 mL), brine (30 mL), dried over sodium sulphate, evaporated in vacuo and purified by column
chromatography on silica gel (hexane:EtOAc 80:20) to afford a colourless gum in 51% yield, 220mg.
1H NMR (400 MHz, DMSO-Da) 6: 1.39 (s, 9H), 5.32 (s, 2H), 7.42 (dd, 1H), 8.14 (d, 1H), 8.32 (s, 1H), 8.55
(d, 1H).
Preparation 190: 1H-Pyrazolo[4,3-b]pyridinylacetic acid
/ \
N\ I
Tert—butyl azolo[4,3,b]pyridinylacetate (Preparation 189, 220 mg, 0.944 mmol) was dissolved in
HCl (4 mL, 4.0 M in 1,4 dioxane) and stirred under nitrogen at room temperature for 4 hours. The mixture
was evaporated in vacuo and the residue was triturated with anhydrous diethyl ether to afford the title
compound as an off white solid in 60% yield, 120mg.
1H NMR (400 MHz, DMSO-Ds) 6: 5.36 (s, 2H), 7.51 (dd, 1H), 8.30 (d, 1H), 8.36 (s, 1H), 8.62 (d, 1H);
LCMS m 10): R = 1.39 min; m/z176[M-H]+.
Preparation 192: 2-(3-formylphenyl)-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridin
yl}acetamide
Me’<
Dess Martin Periodinane (392 mg, 0.93 mmol) was added to a solution of (2-[3-(hydroxymethyl)phenyl]—N-
-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}acetamide (Preparation 193, 265
mg, 0.62 mmol) in dichloromethane (15 mL) and the mixture was d at room temperature for 18 hours.
Water (10 mL) was added and the aqueous layer extracted with a 95:5 mixture of
dichloromethane/methanol (3 x 10 mL). The combined organic layers were dried over magnesium sulfate
W0 2012/137089 194
and concentrated in vacuo. The residue was purified by column tography using silica gel (gradient
of dichloromethane to dichloromethane/methanol (10:1 to 9:1) to afford the title compound as a brown
solid in 80% yield, 211 mg.
LCMS (System 4): R = 2.11 min; m/z 428 .
Preparation 193: 2-[3-(hydroxymethyl)phenyl]-N-{5-[(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidin-5—
yl)carbonyl]pyridinyl}acetamide
Me’<
A mixture of (5-aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)methanone ration
95, 250 mg, 0.89 mmol), [4-(hydroxymethyl)phenyl]acetic acid (177 mg, 1.06 mmol) and HATU (505 mg,
1.33 mmol) in pyridine (7 mL) was stirred at 50 °C for 3 hours, then at room ature for 18 hours.
The mixture was concentrated in vacuo and azeotroped with toluene. The residue was purified by column
chromatography using silica gel (gradient of dichloromethane to dichloromethane/methanol (10:1 to 9:1)
to afford the title compound as a gum. This material was dissolved in a 9:1 mixture of
dichloromethane/methanol (1.5 mL) and added dropwise to diethyl ether (100 mL), the resulting
itate was filtered off and dried in vacuo to afford the title compound as a white solid, 412 mg, which
was used crude in subsequent reactions.
LCMS (System 2): R = 1.33 min; m/z 430 .
Preparation 194: (5-((Diphenylmethylene)amino)pyridinyl)(7H-pyrrolo[2,3-d]pyrimidinyl)methanol
/N O
N \ N
N N
H O
A mixture of 7H-pyrrolo[2,3-d]pyrimidine (Preparation 202, 953 mg, 8.0 mmol), 5-
[(diphenylmethylene)amino]nicotinaldehyde ration 106, 3340 mg, 11.7 mmol) and KOH (1350 mg,
24 mmol) in MeOH (16 mL) was stirred for 16 hours at room temperature. The reaction was neutralized
with saturated aqueous ammonium chloride solution and the mixture was extracted with EtOAc (3 x 200
mL). The combined organic layers were dried over sodium sulfate and concentrated in vacuo to obtain a
crude residue. The crude material was purified by column chromatography on silica gel (EtOAc:MeOH =
100:0 to 80:20,) to give the desired compound as a solid in 62% yield, 2003 mg.
W0 2012/137089 195
1H NMR (400 MHz, DMSO-d6) 6: 5.88-5.99 (m, 2H), .99 (m, 1H), 7.04-7.14 (m, 3H), .27 (m,
3H), 7.43-7.51 (m, 2H), 7.51-7.58 (m, 1H), .69 (m,2H), 7.89 (d, 1H), 8.21 (d, 1H), 8.72 (s, 1H), 8.73
(s, 1H), 11.92 (brs, 1H)
Preparation 195: (5-((Diphenylmethylene)amino)pyridinyl)(7H-pyrrolo[2,3-d]pyrimidinyl)methanone
N\ N
WuI 0
To a stirred solution of (5-((Diphenylmethylene)amino)pyridinyl)(7H-pyrrolo[2,3-d]pyrimidin
yl)methanol (Preparation 194, 1820 mg, 4.5 mmol) in MeCN (45 mL) was added Mn02 (1960 mg, 22.5
mmol) portionwise and the resulting mixture stirred at 50 °C overnight. Another n of Mn02 (1960 mg,
22.5 mmol) was added to the reaction and the mixture heated to reflux for 5 hr. After cooling to room
temperature, the reaction mixture was filtered through a pad of arbocel, the filter cake rinsed with DCM
(100 mL) and the resulting filtrate concentrated in vacuo. The resulting material was purified by column
tography on silica gel (gradient of EtOAc:MeOH 100:0 to 90:10) to give the desired compound as
a solid in 61% yield, 1120 mg.
1H NMR (400 MHz, DMSO-d6) 6: 7.24-7.33 (m, 2H), 7.37-7.64 (m, 7H), .78 (m, 3H), 8.28 (d, 1H),
8.53 (d, 1H), 8.94 (s, 1H), 9.41 (s, 1H), 13.09 (brs, 1H)
Preparation 196: (5-aminopyridinyl){7-[2-{[tert-butyl(dimethyl)silyl]oxy}({[tert-
dimethyl)silyl]oxy}methyl)ethyl]—7H-pyrrolo[2,3-d]pyrimidinyl}methanone
\\ /
NC\ NH
N/ N
OTBDMS
OTBDMS
A mixture of (5-((Diphenylmethylene)amino)pyridinyl)(7H-pyrrolo[2,3-d]pyrimidinyl)methanone
(Preparation 195, 1120 mg, 2.77 mmol) and Cs2C03 (2710 mg, 8.31 mmol) in DMF (10 mL) was stirred
at room temperature for 30 min. A solution of crude 2,2,3,3,9,9,10,10-octamethyl-4,8-dioxa-3,9-
disilaundecanyl trifluoromethanesulfonate (Preparation 108) in DMF (3.8 mL) was then added to the
reaction and the resulting mixture stirred at room temperature for 16 hours. The reaction was quenched
with saturated aqueous ammonium chloride solution (100 mL) and the mixture extracted with EtOAc (100
W0 2012/137089 196
mLx3). The combined c layers were washed with water (200 mL), dried over sodium sulfate and
concentrated in vacuo. The ing material was purified by column chromatography on silica gel
(gradient of heptane:EtOAc 100:0 to 50:50) to provide (5-((diphenylmethylene)amino)pyridinyl)(7-
(2,2,3,3,9,9,10,10-octamethyl-4,8-dioxa-3,9-disilaundecanyl)-7H-pyrrolo[2,3-d]pyrimidin
yl)methanone.
The above material was dissolved in THF (20 mL) and aqueous 1N citric acid (20 mL) was added to the
solution. The reaction e was stirred for 4 hours at room temperature, diluted with water (100 mL)
and basified with NaOH to pH 7. The resulting mixture was extracted with EtOAc (3 x 150 mL), the
combined organic ons were dried over Na2804 and concentrated in vacuo. The resulting material
was purified by column chromatography on silica gel ne:EtOAc = 40:60 to 0:100) to give the desired
compound as a white solid in 49% yield, 739 mg.
1H NMR (400 MHz, DMSO-d6) 6: -0.11 (s, 6H), -0.07 (s, 6H), 0.68 (s, 18H), 3.99-4.16 (m, 4H), 5.01-5.11
(m, 1H), 5.65 (br s, 2H), 7.21-7.26 (m, 1H), 8.13 (d, 1H), 8.17 (d, 1H), 8.38 (s, 1H), 8.98 (s, 1H), 9.44 (s,
1H).
Preparation 197: 3-(3-(trifluoromethyl)-3H-diazirinyl)benzoic acid
0 N~‘N
F F
Ethyl 3-(3-(trifluoromethyl)-3H-diazirinyl)benzoate (Preparation 198, 60 mg, 0.23 mmol) was stirred in
a 2:1 e of THF:water (2mL). Lithium hydroxide (5 mg. 0.23 mmol) was added and the e stirred
at 25° C for 18 hours. The reaction mixture was acidified to pH 1 using 6M aqueous hydrochloric acid and
then extracted with ethyl acetate (3 x 20 mL). The combined organic extracts were dried over magnesium
sulfate and evaporated in vacuo to afford the title compound as a solid in 75% yield, 40 mg. This material
was used in the next step t further purification.
Preparation 198 Methyl 3-(3-(trifluoromethyl)-3H-diazirinyl)benzoate
MeAO o
Ethyl 3-(3-(trifluoromethyl)diaziridinyl)benzoate (Preparation 199, 226 mg, 0.87 mmol) was stirred in
methanol (10mL) with triethylamine (0.36 mL, 2.61 mmol). lodine (662 mg, 2.61 mmol) was dissolved in 2
mL methanol and added portion-wise until an orange-brown colour persisted. The reaction e was
evaporated in vacuo and the residue was diluted with 1M aq. NaOH (30 mL) and then extracted with
EtOAc (3 x 30 mL). The combined organic extracts were dried (MgSO4) and evaporated in vacuo to afford
the title compound as a gum in 27% yield, 60 mg. This material was used in the next step without further
purification.
1H NMR (400 MHz, CDCI3) 6: 1.40 (t, 3H), 4.38 (q, 2H), 7.46-7.51 (m, 2H), 7.83 (s, 1H), 8.08 (d, 1H).
Preparation 199: Ethyl 3-(3-(trifluoromethyl)diaziridinyl)benzoate
W0 2012/137089 197
To a solution of ethyl 3-(2,2,2-trifluoroacetyl)benzoate (500 mg, 2.15 mmol) in ethanol was added
pyridine (5 mL) and hydroxylamine hloride (500 mg, 7.2 mmol) and the resulting e stirred at
57°C for 3 hours. The reaction mixture was cooled and passed through an ion exhange column g
with methanol (30 mL). The methanol solution was evaporated in vacuo to give (E)-ethyl 3-(2,2,2-trifluoro-
1-(hydroxyimino)ethyl)benzoate as an oil in 71% yield, 401 mg which was used in the next step without
further purification. To a stirred solution of (E)-ethyl 3-(2,2,2-trifluoro(hydroxyimino)ethyl)benzoate (401
mg, 1.54 mmol) in dichloromethane (10 mL) was added DMAP (17 mg, 0.14 mmol) and the mixture
cooled to 0°C. 4-methylbenzenesu|fony| chloride (331 mg, 1.74 mmol) was added portion-wise as a
solution in dichloromethane (5 mL). The reaction mixture was then left to stand at room temperature for
18 hours. The mixture was diluted with water (10 mL) and the organic layer separated, dried over
magnesium sulfate and evaporated in vacuo to afford (E)-ethyl 3-(2,2,2-trifluoro
((tosyloxy)imino)ethyl)benzoate as an oil in 78% yield, 500 mg. This material was used in the next step
without further purification. A e of hyl 3-(2,2,2-trifluoro((tosyloxy)imino)ethyl)benzoate (500
mg, 1.2 mmol) in diethylether (5 mL) in a 3-necked flask equipped with an internal thermometer and
sor was cooled to -78°C. Ammonia gas was introduced for 5 min and then stirring continued for 45
min. The reaction mixture was warmed to -33°C and stirred for 2 hours after which it was allowed to warm
to room temperature overnight with stirring. The mixture was evaporated in vacuo and the crude al
purified by column chromatography on silica gel ent of dichloromethane:ethyl acetate 100:0 to
90:10) to afford the title compound as a gum in 76% yield, 240mg.
1H NMR (400 MHz,CDCI3) 6: 1.31 (t, 3H), 4.31 (q, 2H), 7.40-7.44 (m, 1H), 7.74 (d, 1H), 8.02 (d, 1H), 8.20
(s, 1H).
ation 200: 3-(2,2,2-trifluoroacetyl)benzoic acid
O OH
The title compound was prepared according to the method described for Preparation 42 starting from
ethyl 3-(2,2,2-trifluoroacetyl)benzoate to afford the title compound as a white solid in 26% yield, 23 mg.
1H NMR (400 MHz,CDCI3) 6: 7.71 (t, 1H), 8.32 (d, 1H), 8.45 (d, 1H), 8.80 (s, 1H).
W0 2012/137089 198 2012/051363
Preparation 201: 5-iodo—7H-pyrrolo[2,3-d]pyrimidine
I \
K N
A mixture of 7H-pyrrolo[2,3-d]pyrimidine (Preparation 202, 28.0 g, 235 mmol) and N-iodosuccinimide
(55.4 g, 246 mmol) in acetonitrile (470 mL) was stirred at room temperature for 16 hours. The solids were
filtered, rinsed with acetonitrile (150 mL) and dried in vacuo. The solid was dissolved in 1.5 L of 1N
aqueous sodium hydroxide solution and to it was added 2N aqueous en chloride solution until ~pH
9. The resulting precipitate was filtered, rinsed with water (300 mL), and dried in vacuo for 16 hours at 70
oC, ~10 mbar, to afford the title compound in 81% yield, 46.84 g.
1H NMR (400 MHz, a) 6: 7.82 (s, 1H), 8.73 (s, 1H), 8.80 (s, 1H), 12.56 (br s, 1H); LCMS (system
1): R = 0.87 min; m/z 246 [M+H]+.
Preparation 202: 7H-pyrrolo[2,3-d]pyrimidine
k\N I \
In each of four separate reaction vessels, 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (50.0 g, 260 mmol) was
suspended in ethanol (1.4 L) and concentrated ammonium ide on (140 mL). 10% palladium
on carbon (2.5 g) was added to each vessel and the mixture was pressurized to 20 psi hydrogen and
stirred at room temperature overnight. Those reactions still ning starting material were charged with
another 1 g of 10% palladium on carbon, pressurized to 20 psi hydrogen and stirred until the starting
material had been consumed. The reaction mixture was ed over Arbocel, washed with ethanol, and
the filtrate was evaporated to obtain a white solid. The four crude reaction ts were combined,
suspended in 500 mL water and extracted with ethyl acetate (3 x 500 mL). The organic layers were
ed, dried over magnesium sulfate, filtered and the filtrate was concentrated in vacuo to obtain 122
g of a white solid. The aqueous layer was further extracted with 5% ol in ethyl acetate (3 x 500
mL), the organic layers combined, dried over magnesium sulfate, filtered and the filtrate concentrated in
vacuo to obtain another 30 g of white solid. The solids were combined to obtain the title compound in
98% yield, 152 g.
1H NMR (400 MHz, CDCI3) 6: 6.63 (dd, 1H), 7.43 (dd, 1H), 8.96 (s, 1H), 9.07 (s, 1H), 11.65 (br. s., 1 H).
Preparation 203: N-(5-{7-[3-(tert-Butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-
d]pyrimidinecarbonyl}-pyridinyl)(4-chloro—phenyl) acetamide:
W0 2012/137089 199
(5-Amino-pyridinyl)-{7-[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidin-
methanone (Preparation 216, 30 mg, 0.068 mmol) was ved in dry pyridine (2 mL) under
nitrogen and to this was added (4-chloro-phenyl)—acetic acid (17 mg, 0.102 mmol), ed by HATU (39
mg, 0.102 mmol). The on was heated to 50°C and stirred overnight. The reaction was cooled to
room temperature, diluted with dichloromethane and saturated aqueous sodium bicarbonate was added.
The phases were separated and the aqueous layer was extracted with dichloromethane (3 x 5mL). The
combined organics were trated to dryness to afford N-(5-{7-[3-(tert-butyl-dimethyl-
silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidinecarbonyl}-pyridinyl)(4-chloro-phenyl)-
acetamide as a pale yellow solid 35 mg, 87% yield.
1H NMR (400 MHz, CDCI3) 65-023 (s, 6 H) 0.65 (s, 9 H) 3.77 (s, 2 H) 4.26 (s, 2 H) 4.89 (d, J=7.6 Hz, 2 H)
.27 (d, J=7.8 Hz, 2 H) 7.30 (d, J=8.6 Hz, 2 H) 7.37 - 7.44 (m, 2 H) 7.72 (s, 1 H) 8.49 (s, 1 H) 8.70 (d,
J=2.3 Hz, 1 H) 8.78 (s, 1 H) 8.94 (s, 1 H) 9.64 (s, 1 H).
Preparation 204: N-(5-{7-[3-(tert-Butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-
d]pyrimidinecarbonyl}-pyridinyl)(4-trifluoromethyl-phenyl)-acetamide:
(5-Amino-pyridinyl)-{7-[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidin-
-yl}-methanone (Preparation 216, 30 mg, 0.068 mmol) was dissolved in dry pyridine (2 mL) under
nitrogen and to this was added (4-trifluoromethyl-phenyl)-acetic acid (21 mg, 0.102 mmol), followed by
HATU (39 mg, 0.102 mmol). The reaction mixture was heated to 50°C and d overnight. The reaction
was then cooled to room temperature, diluted with dichloromethane (5 mL) and saturated aqueous
sodium bicarbonate was added (5 mL). The phases were separated and the aqueous layer was extracted
with dichloromethane (3 x 5mL). The combined organics were concentrated to dryness to afford N-(5-{7-
[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidinecarbonyl}-pyridinyl)-
2-(4-trifluoromethyl-phenyl)-acetamide as a pale yellow solid 39 mg, 92% yield.
1H NMR (400 MHz, DMSO-da) 63-029 (s, 6 H) 0.58 (s, 9 H) 3.84 (s, 2 H) 4.16 (s, 2 H) 4.77 (d, J=7.4 Hz, 2
H) 5.21 (d, J=7.4 Hz, 2 H) 7.56 (d, J=7.8 Hz, 2 H) 7.68 (d, J=7.8 Hz, 2 H) 8.28 (s, 1 H) 8.46 (t, J=2.1 Hz, 1
H) 8.69 (d, J=2.0 Hz, 1 H) 8.91 - 8.95 (m, 2 H) 9.46 (s, 1 H) 10.71 (s, 1 H).
W0 37089 200
Preparation 205: 2-Allyl-malonic acid diethyl ester
H2C— o
O >
Malonic acid diethyl ester (100 g, 0.625 mol) was added dropwise at 0°C to a mixture of sodium de
(46.8 g, 0.688 mol) in ethanol (1 L). The reaction mixture was d for 4 hours. 3-Bromo—propene (83.2
g, 0.688 moles) was added dropwise to the mixture at 0°C. After addition, the mixture was warmed to
reflux and stirred overnight. The mixture was cooled to room temperature, filtered and the solvent was
removed in vacuo to afford the title compound in 86% yield, 100 g which was used for the next step
without further purification.
Preparation 206: 5-Allylhydroxy-3H-pyrimidinone
kx I
N OH
l-malonic acid diethyl ester (Preparation 205, 100 g, 0.5 mol) was added dropwise to a mixture of
sodium methoxide (27 g, 0.5 mol) in l (1 L) at 0-5°C and the mixture was stirred at this temperature
for 10 min. Formamidine acetate (51.9 g, 0.5 mol) was added and the e was stirred at room
temperature overnight. The solvent was removed in vacuo, and aqueous hydrochloric acid (36.5%) and
water were added to adjust the pH to approximately 3 at 0-20°C. The resulting mixture was filtered to
afford the title compound as a colorless solid in 57% yield, 87.12 g.
1H NMR (400 MHz, DMSO-da) 6: 2.96 (d, 1H), 4.89 (m, 2H), 5.76 (m, 1H), 7.89 (s, 1H).
Preparation 207: 5-Allyl-4,6-dichloro-pyrimidine
k\N Cl
5-Allylhydroxy—3H-pyrimidinone (Preparation 206, 40 g, 0.263 mol) was added to POCI3 (100 mL) at
room temperature. The solution was stirred and warmed to reflux for 8 hours. The mixture was evaporated
in vacuo to remove most of the POCI3. The residue was poured slowly onto ice-water, which was
extracted with ethyl e (500 mL x 4), washed with brine (300 mL), dried over sodium sulfate and
evaporated in vacuo to afford the title compound as a yellow oil in 59% yield, 31 g.
1H NMR (400 MHz,CDCI3 ) 6: 3.59 (m, 2H), 5.09 (m, 2H), 5.79 (m, 1H), 8.59 (s, 1H).
Preparation 208: (4,6-Dichloro-pyrimidinyl)-acetaldehyde
W0 2012/137089 201
k\N |
To a stirred on of 5-Allyl-4,6-dichloro-pyrimidine (Preparation 207, 30 g, 0.159 mole) in dry
dichloromethane (400 mL) was bubbled ozone at -70°C for 30 min. After excess ozone was purged by
nitrogen gas, dimethyl sulfide (10 mL) was added at -5°C, and the reaction was d for 2 hours. The
mixture was washed with water, brine, dried over sodium sulfate, and evaporated in vacuo. The crude
material was purified by trituration from pentane-diethyl ether to afford the title compound as a colorless
solid in 84% yield, 10 g.
1H NMR (400 CI3) 6: 4.15 (s, 2H), 8.74 (s, 1H), 9.80 (s, 1H).
Preparation 209: 2-(4-Chloro-pyrrolo[2,3-d]pyrimidinyl)hydroxymethyl-propane-1,3-diol
N \ \
KN/ N
ine (1.27 g, 10.5 mmol) was added to 4,6-Dichloro-pyrimidinyl)-acetaldehyde (Preparation 208,
1.0 g, 5.2 mmol) in ethanol (40 mL) and stirred at reflux temperature for 16 hours. The reaction mixture
was evaporated in vacuo and partitioned between dichloromethane and aqueous saturated sodium
bicarbonate. The separated aqueous phase was extracted with dichloromethane twice more and the
combined organics were washed with ted brine and evaporated in vacuo to afford the title
compound as a pale yellow foam in 74% yield, 1.0 g.
1H NMR (400 MHz,CDCI3) 8 ppm 2.97 (br. s, 1H), 3.09 - 3.27 (m, 2H), 3.51 (d, J=9.18 Hz, 1H), 3.83 - 4.00
(m, 4H), 4.06 (d, J=11.13 Hz, 1H), 5.43 (dd, J=6.44, 1.76 Hz, 1H), 6.16 (dd, J=10.74, 4.69 Hz, 1H), 8.40
(s, 1H); LCMS (System 2): R = 1.04 min; m/z 258 [M+H]+.
Preparation 210: esulfonic acid 2-(4-chloro-pyrrolo[2,3-d]pyrimidinyl)hydroxy
hydroxymethyl-propyl ester
NI \ \
kN/ N
HO7‘ ,9
0‘8:
OH 0
Triethylamine (0.879 mL, 6.31 mmol) and trimethylamine hydrochloride (253 mg, 2.65 mmol) were added
to a solution of 2-(4-chloro-pyrrolo[2,3-d]pyrimidinyl)hydroxymethyl-propane-1,3-diol (Preparation
W0 2012/137089 202
209, 650 mg, 2.52 mmol) in dichloromethane (20 mL) at 0°C. The mixture was d portion-wise with
tosyl chloride (505 mg, 2.65 mmol) and stirred at 0°C for 16 hours. The reaction e was treated with
water and stirred for 10 min. The resulting mixture was washed with citric acid, ted aqueous sodium
bicarbonate, and saturated brine, then ated in vacuo. The crude product was purified by column
tography on silica gel ent of EtOAc:DCM 0:100 to 30:70) to afford the title compound as a
colorless solid in 55% yield, 570 mg.
LCMS (system 2): R = 1.28 min; m/z 412 [M+H]+.
Preparation 211: Chloro-pyrrolo[2,3-d]pyrimidinyl)-oxetanyl]-methanol
nButyllithium (12.2 mL, 30.6 mmol, 2.5 M in hexanes) was added to toluenesulfonic acid 2-(4-chloro-
pyrrolo[2,3-d]pyrimidinyl)hydroxyhydroxymethyl-propyl ester (Preparation 210, 5.73 g, 13.9
mmol) in THF (100 mL) at 0°C and stirred for 5 min. The reaction mixture was then warmed to room
temperature and stirred for 16 hours, at which point it was quenched with saturated aqueous ammonium
chloride. The ing mixture was extracted with ethyl acetate (100mL x3) and the combined organic
phases were dried (MgSO4) and concentrated in vacuo. The crude product was triturated with
dichloromethane and filtered to afford the title compound in 36% yield, 1.2 g.
1H NMR (400 MHz, MeOH-d4) 6: 4.15 (s, 2H), 4.92 (d, J=7.22 Hz, 2H), 5.22 (d, J=7.03 Hz, 2 H), 6.69 (d,
J=3.51 Hz, 1H), 7.51 (d, J=3.71 Hz, 1H), 8.51 (s, 1H).
Preparation 212: 7-[3-(tert-Butyl-dimethyl-silanyloxymethyl)-oxetanyl]chloro-7H-pyrrolo[2,3-
d]pyrimidine
NI \ \
kN/ N
TBDMSO i’
lmidazole (277 mg, 4.07 mmol) was added to [3-(4-Chloro-pyrrolo[2,3-d]pyrimidinyl)-oxetanyl]-
methanol (Preparation 211, 650 mg, 2.71 mmol) in dicloromethane (10 mL), and this mixture was treated
with a solution of t—butyldimethylsilyl chloride (495 mg, 3.25 mmol) in dichloromethane (5 mL). The
resulting mixture was stirred at room temperature for 16 hours, then quenched with water (50 mL). The
mixture was extracted with dichloromethane (3 x 50mL), and the combined organic phases were washed
with water and saturated brine, and evaporated in vacuo to afford the title compound as a pale brown oil
in 91% yield, 875 mg.
W0 2012/137089 203
1H NMR (400 MHz, CDCI3) 6: -0.21 (s, 6H), 0.77 (s, 9H), 4.22 (s, 2H), 4.88 (d, J=7.22 Hz, 2H), 5.19 (d,
J=7.03 Hz, 2H), 6.61 (d, J=3.71 Hz, 1H), 7.11 (d, J=3.71 Hz, 1H), 8.55 (s, 1H); LCMS (system 2): R =
1.82 min; m/z 354 [M+H]+.
Preparation 213: 7-[3-(tert-Butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidine
N \ \
m/NN
fiOTBDMS
The title compound was prepared according to the method bed for Preparation 8 using tert-
Butyl-dimethyl-silanyloxymethyl)-oxetanyl]chloro-7H-pyrrolo[2,3-d]pyrimidine (Preparation 212) to
afford the title compound as a brown oil in 82% yield, 650 mg.
1H NMR (400 MHz, CDCI3) 6: -0.24 (s, 6H), 0.76 (s, 9H), 4.24 (s, 2H), 4.91 (d, J=7.03 Hz, 2H), 5.22 (d,
J=7.03 Hz, 2H), 6.55 (d, J=3.51 Hz, 1H), 7.08 (d, J=3.71 Hz, 1H), 8.78 (s, 1H), 8.97 (s, 1H); LCMS
(system 2): R = 0.90 min; m/z 320 [M+H]+.
Preparation 214: 7-[3-(tert-Butyl-dimethyl-silanyloxymethyl)-oxetanyl]—5-iodo-7H-pyrrolo[2,3-
d]pyrimidine
I S OTBDMS
The title compound was prepared according to the method described for Preparation 14 using tert-
Butyl-dimethyl-silanyloxymethyl)-oxetanyl]—7H-pyrrolo[2,3-d]pyrimidine (Preparation 213) and DMF to
afford the title compound as a brown solid in 75% yield, 675 mg.
1H NMR (400 MHz, CDCI3) 6: -0.24 (s, 6H), 0.77 (s, 9H), 4.20 (s, 2H), 4.88 (d, J=7.22 Hz, 2H), 5.21 (d,
J=7.03 Hz, 2H), 7.17 (s, 1H), 8.77 (s, 1H), 8.81 (s, 1H); LCMS (system 2): R = 1.55 min; m/z 446 [M+H]+.
Preparation 215: (7-(3-(((tert-Butyldimethylsilyl)oxy)methyl)oxetanyl)-7H-pyrrolo[2,3-d]pyrimidin
yl)(5-((diphenylmethylene)amino)pyridinyl)methanone
fiOTBDMS
The title compound was prepared according to the method described for Preparation 24 using 7-[3-(tert-
butyl-dimethyl-silanyloxymethyl)-oxetanyl]—5-iodo-7H-pyrrolo[2,3-d]pyrimidine (Preparation 214) and 5-
W0 2012/137089 204
enylmethylene)amino]-N-methoxy-N-methylnicotinamide (Preparation 23) to afford the title
compound as a purple solid in 51% yield, 240 mg.
1H NMR (400 MHz, CDCI3) 6: 0.20 (s, 6H), 0.70 (s, 9H), 4.28 (s, 2H), 4.89 (d, J=7.22 Hz, 2H), 5.20 (d,
J=7.22 Hz, 2H), 7.11 - 7.18 (m, 2H), 7.33 (m, 3H), 7.40 - 7.48 (m, 2H), 7.49 - 7.57 (m, 3H), 7.79 (d, J=7.42
Hz, 2H), 8.18 (d, J=2.54 Hz, 1H), 8.60 (d, J=1.95 Hz, 1H), 8.93 (s, 1H), 9.60 (s, 1H).
Preparation 216: (5-Amino-pyridinyl)-{7-[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}-methanone
fiomoms
The title compound was prepared according to the method bed for ation 37 using (7-(3-
(((tert-butyldimethylsilyl)oxy)methyl)oxetanyl)-7H-pyrrolo[2,3-d]pyrimidinyl)(5-
((diphenylmethylene)amino)pyridinyl)methanone (Preparation 215) to afford the title compound as a
colourless solid in 76% yield, 132 mg.
1H NMR (400 MHz, CDCI3) 6: 0.01 (s, 6H), 0.88 (s, 9H), 4.11 (br. s, 2H), 4.45 (s, 2H), 5.17 (m, 2H), 5.40
(m, 2H), 7.58 (s, 1H), 7.80 (s, 1H), 8.45 (s, 1H), 8.61 (s, 1H), 9.15 (s, 1H), 9.83 (s, 1H).
Preparation 217: 2-(4-chloro-7H-pyrrolo[2,3-d]pyrimidinyl)methylpropane-1,3-diol
N/g”
How9 OH
The title compound was prepared according to the method described for Preparation 1 using (4,6-
dichloropyrimidinyl)acetaldehyde (Preparation 208) and 2-aminomethylpropane-1,3-diol to afford
the title compound as a yellow liquid in 79% yield, 3.88 g.
1H NMR (400 MHz, DMSO-da) 6: 1.66 (s, 3H), 3.86 (dd, 2H), 4.13 (dd, 2H), 4.92 (t, 2H), 6.58 (d, 1H),
7.73 (d, 1H), 8.59 (s, 1H).
Preparation 218: 4-chloro(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidine
N/ \ u | N
N ZSMe
W0 2012/137089 205
n-BuLi (6.6 mL of a 2.5 M solution in hexane, 16.5 mmol) was added to a solution of 2-(4-chloro-7H-
pyrrolo[2,3-d]pyrimidinyl)methylpropane-1,3-diol (Preparation 217, 3.63 g, 15.0 mmol), in THF (80
mL) at -78 °C. The reaction mixture was allowed to warm to -50 °C in 2 hours and TsCl (3.15 mg, 16.5
mmol) in THF (20 mL) was added to the reaction. The reaction was allowed to warm to 0 °C in 3 hours
and additional n-BuLi (6.6 mL of a 2.5 M solution in hexane solution, 16.5 mmol) was slowly added to the
reaction mixture. The mixture was stirred for 1 hour at 0 °C and stirred at 60 °C for 16 hours. After cooling
to room temperature, the on was quenched by 50 mL of ted aqueous NH4C| solution and 100
mL of water, and the e was extracted with EtOAc (3x 100 mL). The combined organic layers were
dried over MgSO4 and concentrated in vacuo. The crude e was purified by column chromatogranhy
on silica gel ent of EtOAc:heptane 20:80 to 70:30) to give a solid in 55% yield, 1.87 mg.
1H NMR (400 MHz, DMSO-da) 6: 1.79 (s, 3H) 4.72 (d, 2H) 5.15 (d, 2H) 6.69 (d, 1H) 7.78 (d, 1H) 8.59 (s,
1H).
Preparation 219: 7-(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidine
NfiKN
NZSMQ
The title compound was prepared according to the method described for Preparation 8 using 4-chloro
(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidine (Preparation 218) to afford the title compound as a
yellow liquid in 22% yield, 0.341 g.
1H NMR (400 MHz, DMSO-da) 6: 1.81 (s, 3H), 4.72-4.77 (m, 2H), 5.16-5.21 (m, 2H), 6.69 (d, 1H), 7.66 (d,
1H), 8.76 (s, 1H), 9.02 (s, 1H).
The title compound may also be prepared ing to the following method:
LiHMDS (14.5 mL of a 1M solution in THF, 14.5 mmol) was added slowly to a solution of 2-Methyl
pyrrolo[2,3-d]pyrimidinyl-propane-1,3-diol (Preparation 278, 3 g, 14.5 mmol) in anhydrous THF (200
mL) over a period of 2 hours (using a syringe pump) under nitrogen at 0°C. After completion of the
addition, the reaction mixture was stirred for an additional 40 min before TsCl (2.76 g, 14.5 mmol) as a
solution in THF (50 mL) was slowly added. The mixture was d for another 1 hour at 0°C. TLC
showed consumption of starting al and another equivalent of LiHMDS (14.5 mL, 14.5 mmol) was
added to the mixture and it was heated at 60° for 16 hours. Sat. aq. NH4C| solution (200 mL) was then
added and the mixture then extracted with EtOAc (3 x 100 mL). The combined organic layers were dried
(Na2804) and evaporated in vacuo. The crude material was purified by column chromatography on silica
gel (gradient of EtOAc:Hexane 3:7 to 2:3) to afford the title compound as a colourless gum in 37% yield, 1
W0 37089 206
Preparation 220: 5-iodo(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidine
I \
K N
N Me
The title compound was prepared according to the method described for Preparation 14 using 7-(3-
methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidine (Preparation 219) to afford the title compound as a yellow
solid in 78% yield, 0.44 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.80 (s, 3H), 4.71 (d, 2H), 5.18 (d, 2H), 7.94 (s, 1H), 8.76 (s, 1H), 8.82
(s, 1H).
Preparation 221: (5-((diphenylmethylene)amino)pyridinyl)(7-(3-methyloxetanyl)-7H-pyrrolo[2,3-
d]pyrimidinyl)methanone
The title compound was prepared according to the method described for Preparation 24 using 5-iodo
(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidine ration 220) to afford the title compound.
LCMS (system 2): R = 1.24 min; m/z 474 [M+H]+.
ation 222: (5-aminopyridinyl)[7-(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidinyl]methanone
Nk/l NH2
\N NMe
The title compound was prepared according to the method described for Preparation 37 using (5-
((diphenylmethylene)amino)pyridinyl)(7-(3-methyloxetanyl)-7H-pyrrolo[2,3-d]pyrimidin
yl)methanone (Preparation 221) to afford the title compound as a white solid in 14% yield over two
steps, 64 mg.
1H NMR (400 MHz, DMSO-da) 6: 1.89 (s, 3H), 4.75 (d, 2H), 5.23 (d, 2H), 5.64 (br s, 2H), 7.29-7.36 (m,
1H), 8.17 (d, 1H), 8.24 (d, 1H), 8.41 (s, 1H), 8.95 (s, 1H), 9.46 (s, 1H); LCMS (System 4): R = 2.30 min;
m/z 310 [M+H]+.
W0 2012/137089 207
Preparation 223: N-[5-({7-[3-({[tert-butyl(dimethyl)silyl]oxy}methyl)oxetanyl]—7H-pyrrolo[2,3-
m idinyl}carbonyl)pyrid inyl]—2-(5-chloropyridinyl)acetamide
\/0/
/ N /
OTBDMS
(5-Amino-pyridinyl)-{7-[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidin-
methanone (Preparation 216, 30 mg, 0.068 mmol) was added to a stirred mixture of (5-
chloropyridinyl)acetic acid (17.5 mg, 0.102 mmol) and HATU (38.8 mg, 0.102 mmol) in pyridine (2 ml).
The on mixture was warmed to 50°C and stirred at this ature for 14 hours. The reaction was
then cooled and a further portion of HATU (38.8 mg, 0.102 mmol) was added. The reaction was warmed
to 50°C and stirred at this temperature for 8 hours and then allowed to cool to room temperature and
stirred for a further 60 hours. The on mixture was then diluted with DCM (20ml) and the ant
solution quenched with saturated NaHCO3 (20 ml). The layers were separated and the aqueous layer
extracted with DCM (3x 20 ml). The combined organic layers were washed with brine (20ml) and then
concentrated in vacuo to give the crude product as a pale yellow oil (35 mg) which was taken forward for
use in the preparation of Example 238.
Preparation 224: N-[5-({7-[3-({[tert-butyl(dimethyl)silyl]oxy}methyl)oxetanyl]—7H-pyrrolo[2,3-
d]pyrimidinyl}carbonyl)pyridinyl]—2-[3-(trifluoromethyl)-1H-pyrazolyl]acetamide
\ / O CF3
N / \ ”kiwi;
OTBDMS
(5-Amino-pyridinyl)-{7-[3-(tert-butyl-dimethyl-silanyloxymethyl)-oxetanyl]-7H-pyrrolo[2,3-d]pyrimidin-
-yl}-methanone (Preparation 216, 30 mg, 0.068 mmol) was added to a stirred mixture of [3-
(trifluoromethyl)—1H-pyrazolyl]acetic acid (19.8 mg, 0.102 mmol) and HATU (38.8 mg, 0.102 mmol) in
pyridine (2 ml). The on mixture was warmed to 50°C and stirred at this temperature for 14 hours.
The reaction mixture was then cooled to room temperature and diluted with DCM (20ml) and the resultant
solution quenched with saturated NaHCO3 (20 ml). The layers were separated and the aqueous layer
extracted with DCM (3x 20 ml). The combined organic layers were washed with saturated brine (20ml)
and then concentrated in vacuo to give the crude product as a pale yellow solid (44 mg) which was taken
forward crude for use in the preparation of Example 239.
Preparation 225: 4-Chloroiodo-7H-pyrrolo[2,3-d]pyrimidine
W0 2012/137089 208
“I”\
To a suspension of 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (70.5 g, 0.45 mol) in DCM (1.8 L) was added N-
iodosuccinimide (120 g, 0.54 mol) in portions. After on, the mixture was stirred at room temperature
ght. The solid was filtered and washed with water (250 mL), MeOH (280 mL) and CHZCIZ (280 mL)
sequentially. The solid was dried under vacuum to afford roiodo-7H-pyrrolo[2,3-d]pyrimidine as a
brown solid in 79% yield, 107g.
Preparation 226: 4-Chloroiodomethyl-7H-pyrrolo[2,3-d]pyrimidine
”C”\
To a mixture of 4-chloroiodo-7H-pyrrolo[2,3-d]pyrimidine (Preparation 225, 30 g, 0.1 mol), Cs2C03 (50
g, 0.15 mol) in DMF (150 mL), Mel (28.4 g, 0.2 mol) was added dropwise at 0°C. After addition, the
mixture was stirred at room temperature for 10 hours. The reaction mixture was cooled to 0°C and
quenched by the addition of water (500 mL). Then the solid was collected by filtration and washed with
EtZO (100 mL) to afford 4-chloroiodomethyl-7H-pyrrolo[2,3-d]pyrimidine as a brown solid in 61%
yield, 20g.
Preparation 227: 5-Bromo-N-methoxy-N-methyl-nicotinamide
Br OMe
\ N/
| |
/ Me
To a solution of 5-bromo-nicotinic acid (100 g, 0.5 mol) in THF (2 L) at 0°C, (COC|)2 (95 g, 0.74 mol) was
added. After stirring for 0.5 h, Et3N (152 g, 1.5 mol) and O, N-dimethyl hydroxylamine.HCl (140 g, 1.5 mol)
were added. The reaction mixture was stirred at room temperature for 2.5 hours and then water (200 mL)
and EtOAc (500 mL) were added. The organic layer was separated, dried (MgSO4) and trated in
vacuo to afford 5-the title compound as a brown oil in 82% yield, 100%.
Preparation 228: (4-Chloromethyl-7H-pyrrolo[2,3-d]pyrimidinyl)(5-
((diphenylmethylene)amino)pyridinyl)methanol
W0 2012/137089 209
To stirred solution of 4-chloroiodomethyl-7H-pyrrolo[2,3-d]pyrimidine ration 226, 6.6 g, 22.7
mmol) in THF (130 mL) was added n-BuLi (18.2 mL of a 2.5M solution in hexane, 45 mmol) at -75 °C
under N2, and the mixture was d for 50 min. A solution of 5-
((diphenylmethylene)amino)nicotinaldehyde (Preparation 106, 6.5 g, 22.7 mmol) in dry THF (50 mL) was
added and the mixture was stirred at -70 °C for 80 min. The mixture was quenched with sat. aq. NH4C|
solution and extracted with EtOAc (300 mL). The organic layer was dried 4) and concentrated in
vacuo to give the title compound as a brown solid in 48% yield, 5.2g.
Preparation 229: (4-chloromethyl-7H-pyrrolo[2,3-d]pyrimidinyl)(5-
((diphenylmethylene)amino)pyridinyl)methanone
CI \/
To a solution of (4-chloromethyl-7H-pyrrolo[2,3-d]pyrimidinyl)(5-((diphenylmethylene)amino)pyridin-
3-yl)methanol (Preparation 228, 11.4 g, 25 mmol) in CHZCIZ (150 mL), artin periodinane (15.9 g,
37 mmol) was added in portions. The reaction mixture was stirred at room temperature for 10 hours and
then aqueous NaOH (30 mL) was added. The mixture was stirred for a further 0.5 hours. The mixture was
then separated and the aqueous layer was exacted by CHZCIZ (100 mL x 2). The combined organic layers
were trated and washed with ether to provide the title compound as a brown solid in 99% yield,
11g.
1H NMR (400 MHz, CDCI3) 6 3.87 (s, 3H), 7.06 (m, 2H), 7.27 (m, 3H), 7.37 (m, 2H), 7.45 (m, 3H), 7.71 (m,
2H), 8.15 (d, J=2.4,1H), 8.49 (d, J=1.2,1H), 8.68 (s, 1H).
Preparation 230: (5-Amino-pyridinyl)-(4-chloromethyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
CI \ /
[\L/ 2
I \
N N\
W0 2012/137089 210
A solution of [5-(benzhydrylidene-amino)pyridinyl]-(4-chloromethyl-7H-pyrrolo[2,3-d]pyrimidiny|)-
methanone (Preparation 229, 30 g, 0.066 mol) in THF (200 mL) was added aq. citric acid (200 mL), the
mixture was stirred for 30 min at room temperature. Ether was added and the layers were separated. The
aqueous layer was ed to pH7 by aqueous Na2C03. Then the mixture was filtered. The filter cake
was evaporated with toluene and the residue was washed with EtOAc (200 mL) to give the title nd
as a brown solid in 95% yield, 18g.
Preparation 231: (5-Amino-pyridinyl)-(7-methylmethylsulfanyl-7H-pyrrolo[2,3-d]pyrimidinyl)—
methanone
To a solution of (5-amino-pyridinyl)-(4-chloromethyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
(Preparation 230, 21 g, 0.073 mol) in MeOH (300 mL) was added CH3SNa (15.5 g, 0.22 mol). The
resulting mixture was stirred for 7 hours at room temperature. The mixture was poured into ice-water (200
mL), the precipitate was filtered, the filter cake was washed with water (100 mL) then acetone (20 mL) to
give no-pyridinyl)-(7-methyl-4—methylsulfanyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone as a
brown solid in 69% yield, 15g.
Preparation 232: (5-Amino-pyridinyl)-(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
[\L/l NH2
N '1
A mixture of (5-amino-pyridinyl)-(7-methylmethylsulfanyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
(Preparation 231, 1.5 g, 5 mmol), Raney Ni (10 g) and NH3H20 (150 mL) in e (150 mL) was
refluxed for 6 hours. The mixture was filtered and the filtrate was concentrated. The residue was purified
via preparative HPLC to give (5-amino-pyridinyl)-(7-methyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
as a brown solid in 15% yield, 0.19g. 1H NMR (400 MHz DMSO-da) 3.88 (s, 3H), 5.63 (s, 2H), .28
(m, 1H), 8.15-8.18 (m, 2H), 8.40 (s, 1H), 8.97 (s, 1H), 9.42 (s, 1H).
Preparation 233: 5,6,7,8-Tetrahydro-[1,7]naphthyridine hloride
HN /
HCI
A methanolic solution (25 mL) of 7-Benzyl-5,6,7,8-tetrahydro-[1,7]naphthyridine (J. Het. Chem. 2001, 38,
535 ) (1.5g, 6.69 mmol) was degassed with argon for 20 min followed by the addition of 4N HCI in
dioxane (2 mL) and Pd/C (300 mg, 20 wt%) and stirred under 50 psi hydrogen pressure at room
W0 2012/137089 211
temperature for 24h. After completion of the reaction the mixture was filtered on a short pad of celite and
washed with methanol (4 x 25 mL). The filtrate was evaporated to s in vacuo and crystallized from
methanol to afford the title compound as yellowish solid in 65% yield (calculated as 2HC| salt), 900 mg.
1H NMR (400 MHz, DMSO-D6) 6: 3.10 (t, 2H), 3.40 (q, 2H), 4.36 (s, 2H), 7.50 (dd, 1H), 7.91 (d, 1H), 8.55
(d, 1H), 9.96 (brs, 2H); LCMS (system 10): R = 1.56 min; m/z135.2 [M+H]+.
Preparation 234: (5,8-Dihydro-6H-[1,7]naphthyridinyl)-acetic acid tert-butyl ester
Me O
MeMe>l\oJJ\/
N(IiN/
A DMF solution (8 mL) of 5,6,7,8-Tetrahydro-[1,7]naphthyridinehydrochloride (Preparation 233, 400 mg,
1.93 mmol), bromo-acetic acid tert-butyl ester (414.1 mg, 2.12 mmol) and triethylamine (1.64 mL, 11.92
mmol) was heated at 80°C for 16 hours. The reaction mixture was diluted with EtOAc (40 mL), washed
with water (3 x 25 mL), brine (20 mL), dried over Na2804 and evaporated to dryness in vacuo to afford the
title compound as colorless sticky solid in 100% yield, 480 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.43 (s, 9H), 2.81 (s, 4H), 3.34 (s, 2H), 3.72 (s, 2H), 7.16 (dd, 1H), 7.51
(d, 1H), 8.29 (d, 1H); LCMS (system 10): R = 3.02 min; m/z 249.4 [M+H]+.
Preparation 235: (5,8-Dihydro-6H-[1,7]naphthyridinyl)-acetic acid hydrochloride
0 \
HoJJ\/N I
(5,8—Dihydro-6H-[1,7]naphthyridinyl)—acetic acid utyl ester (Preparation 234, 1.25 g, 5.03 mmol)
was treated with 4N HCI in e (25 ml) at room temperature for 2 hours. The mixture was evaporated
to dryness in vacuo and the solid residue was triturated with diethyl ether to afford the title compound as
white solid in 78% yield (calculated as HCI salt), 900 mg.
1H NMR (400 MHz, MeOD) 6: 3.38 (t, 2H), 3.79 (t, 2H), 4.38 (s, 2H), 4.84 (s, 2H), 7.79 (dd, 1H), 8.25 (d,
1H), 8.70 (d, 1H).
Preparation 236: Phenyl-[(tetrahydro-pyranyloxy)]—acetic acid
O O
To a stirred solution of hydroxy-phenyl-acetic acid (2 g, 13.4 mmol) in DCM (30 mL) was added PTSA
(51.2 mg, 0.27 mmol) at 0°C followed by the addition of hydro-2H-pyran (1.55 g, 18.4 mmol). The
mixture was stirred at 0°C for another 15 min and then lly warmed up to room temperature and
stirred for a r 1.5 hours. The reaction mixture was d with DCM (100 mL). The c phase
was washed with saturated aq. Na2CO3 (2 x 20 mL), water (20 mL), brine (20 mL), dried (NaZSO4) and
evaporated in vacuo. The crude material was purified by column chromatography on silica gel
W0 2012/137089 212
(EtOAc:petro|eum ether 2:5) to afford the title compound as a yellow sticky solid in 48% yield, 1.5 g.
LCMS (system 10): R = 1.82 min; m/z 237 [M+H]+.
Preparation 237: N-[5-(7-lsopropyl-7H-pyrrolo[2,3-d]pyrimidinecarbonyl)—pyridinyl]phenyl
hydro-pyranyloxy)—acetamide
\ / O
t.\ N
\ WC»
N N O
)‘Me
Me C70
The title nd was prepared according to the method described for Example 1 using phenyl-
[(tetrahydro-pyranyloxy)]-acetic acid (Preparation 236) and no-pyridinyl)-(7-isopropyl-7H-
pyrrolo[2,3-d]pyrimidinyl)—methanone (Preparation 95) to afford the title compound as a yellow solid in
68% yield, 60 mg.
LCMS (system 10): R = 3.56 min; m/z 500.2 [M+H]+.
Preparation 238: 5-Bromohydroxy—nicotinic acid
HO \
N/ OH
Bromine (3.6 mL, 70.5 mmol) was added dropwise to a suspension of oxy—nicotinic acid (7 g, 50.32
mmol) in water (70 mL) at 0°C and the mixture was stirred at room temperature for 4 hours. The
precipitated solid was filtered, washed with cold water and dried to get the title compound as off white
solid in 82% yield, 9 g.
1H NMR (400 MHz, DMSO-D6) 6: 8.03 (d, 1H), 8.14 (d, 1H), 12.57 (brs, 1H).
Preparation 239: 5-Bromohydroxy—N-methoxy—N-methyl-nicotinamide
Me\ Br
N \
. |
,o /
Me N OH
5-Bromohydroxy—nicotinic acid (Preparation 238, 4 g, 18.4 mmol), HATU (13.95 g, 36.71 mmol), O,NDimethyl-hydroxylamine
hydrochloride (2.15 g, 22.02 mmol) and DIPEA (15.82 mL, 91.75 mmol) were
taken in ous DMF (30 mL). The mixture was stirred at room temperature for 16 hours. The reaction
mixture was diluted with EtOAc (150 mL), washed with saturated aq. Na2C03 (2 x 50 mL), water (4 x 30
mL), brine (30 mL), dried (Na2804) and ated in vacuo. The crude mixture was purified by column
chromatography on silica gel (gradient of EtOAc:Hex 4:6 to 6:4) to afford the title compound as light
yellow sticky solid in 38%, 1.8 g.
1H NMR (400 MHz, DMSO-D6) 6: 3.22 (s, 3H), 3.61 (s, 3H), 7.95 (d, 1H), 8.14 (d, 1H), 12.45 (brs, 1H);
LCMS (system 10): R = 1.67 min; m/z 261, 263 [M+H]+.
Preparation 240: 5-Bromoethoxy—N-methoxy—N-methyl-nicotinamide
W0 2012/137089 213
Me\N Br
I I
Me’O N/ OK
A mixture of 5-bromohydroxy-N-methoxy—N-methyl-nicotinamide (Preparation 239, 1.80 g, 6.90 mmol),
ethyl iodide (2.8 mL, 34.5 mmol) and silver carbonate (3.8 g, 13.8 mmol) in DCM (10 mL) was d at
room ature for 40 hours. The reaction mixture was diluted with DCM (50 mL), washed with water (2
x 30 mL), brine (20 mL), dried (Na2804) and evaporated in vacuo. The crude material was purified by
column chromatography on silica gel (EtOAc:petroleum ether 2:5) to afford the title compound as a yellow
sticky solid in 30% yield, 600 mg.
1H NMR (400 MHz, CDCI3) 6: 1.44 (t, 3H), 3.35 (s, 3H), 3.57 (s, 3H), 4.47 (q, 2H), 8.23 (d, 1H), 8.54 (d,
1H).
Preparation 241: (5-Bromoethoxy—pyridinyl)-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)—
methanone
/ O\/Me
NI \ \
kN/ N
>‘Me
A solution of 5-lodoisopropyl-7H-pyrrolo[2,3-d]pyrimidine (Preparation 93, 530 mg, 1.85 mmol) in
anhydrous diethyl ether (8 mL) was cooled to -78°C under nitrogen and n-BuLi (1 mL of a 2.03M solution
in Hexane, 2.03 mmol) was added drop wise over a period of 10 min. Just after the completion of addition
of n-BuLi, a solution of 5-Bromoethoxy-N-methoxy—N-methyl-nicotinamide (Preparation 240) in
anhydrous diethyl ether (7 mL) was added slowly to the mixture and it was d at the same
temperature for r 1 hour. The reaction mixture was warmed to room ature and allowed to stir
for another hour before being quenched with sat. aq. NH4C| (15 mL). The mixture was extracted with
EtOAc (3 x 15 mL), washed with water (20 mL), brine (15 mL), dried (Na2804) and evaporated in vacuo.
The crude material was purified by column chromatography on silica gel (EtOAc:petroleum ether 2:10) to
afford the title compound as a yellow gum in 49% yield, 350 mg.
1H NMR (400 MHz, CDCI3) 6: 1.50 (t, 3H), 1.60 (d, 6H), 4.55 (q, 2H), 5.15-5.20 (m, 1H), 7.82 (s, 1H), 8.32
(d, 1H), 8.57 (d, 1H), 9.00 (s, 1H), 9.53 (s, 1H).
Preparation 242: (5-Aminoethoxy—pyridinyl)-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidinyl)-methanone
W0 2012/137089 214
\ [<1 O\/Me
N \ \
lN/ N
Me)‘Me
Copper(|) oxide (38.6 mg, 0.27 mmol) was added to (5-Bromoethoxy—pyridin3-yl)-(7-isopropyl-7H-
pyrrolo[2,3-d]pyrimidinyl)-methanone (Preparation 241, 350 mg, 0.89 mmol) in concentrated a
solution (6 mL) and NMP (1 mL). The mixture was heated in a sealed vessel at 130°C for 17 hours. The
reaction mixture was cooled to room temperature and diluted with water (10 ml). It was extracted with
% i-PrOH in DCM (6 x 50 mL), dried (NaZSO4) and evaporated in vacuo. The crude material was
ed by column chromatography on silica gel (MeOH:DCM 5:95) to afford the title compound as a
yellow solid in 24% yield, 70 mg. LCMS (system 10): R = 2.99 min; m/z 326 .
Preparation 243: 2-(4-Chloro-phenyl)hydroxy-propionic acid methyl ester
l\|/le
O OH
To a stirred solution of (4-Chloro-phenyl)—acetic acid methyl ester (2 g, 10.8 mmol) in DMSO (22 mL) was
added sodium methoxide (29.2 mg, 0.54 mmol) at 0°C. Paraformaldehyde (342 mg, 11.4 mmol) was then
added and the reaction mixture was stirred at room temperature for 4 hours. The reaction mixture was
diluted with EtOAc (100 mL), washed with water (3 x 20 mL), brine (20 mL), dried (Na2804) and
evaporated in vacuo. The crude material was purified by column chromatography on silica gel (gradient of
hexane:EtOAc 100:0 to 84:16) to afford the title compound as a colourless gum in 52% yield, 1.2 g.
1H NMR (400 MHz, CDCI3) 8: 2.23 (t, 1H), 3.70 (s, 3H), 3.79-3.83 (m, 2H), .12 (m, 1H), 7.20 (d,
2H), 7.30 (d, 2H); LCMS (system 10): R = 3.03 min; m/z 215 [M+H]+.
Preparation 244: hloro-phenyl)hydroxy-propionic acid
HO OH
To a stirred solution of 2-(4-Chloro-phenyl)hydroxy-propionic acid methyl ester (Preparation 243, 500
mg, 2.33 mmol) in THF (7 mL) was added a solution of lithium hydroxide monohydrate (244 mg, 5.82
mmol) in water (2 mL) dropwise at 0°C and the resulting mixture stirred at room temperature for 1 hour.
The reaction mixture was acidified (pH~3) with 2N hydrochloric acid and extracted with DCM (3 x 20 mL).
The combined c layers were washed with brine (10 mL), dried 4) and evaporated in vacuo to
afford the title compound as a white solid in 90% yield, 420 mg.
W0 37089 215
1H NMR (400 MHz, 6) 6: 3.58 (dd, 1H), 3.66 (t, 1H), 3.87 (t, 1H), 7.32 (d, 2H), 7.38 (d, 2H), 12.36
(br, 1H).
Preparation 245: 1-(4-Chloro-pyrrolo[2,3-d]pyrimidinyl)-cyclopropanecarboxylic acid methyl ester
To a stirred solution of (4,6-dichloro-pyrimidinyl)acetaldehyde (Preparation 208, 2 g, 10.5 mmol) and
1-Amino-cyclopropane-carboxylicacidmethylester hydrochloride (1.33 g, 11.6 mmol) in ethanol (30 mL)
was added triethylamine (4.4 mL, 31.6 mmol) and the mixture heated in a sealed tube at 100°C for 10
hours. Acetic acid (1.21 mL, 21.1 mmol) was then added and the mixture heated at 100°C for additional
16 hours. The reaction mixture was cooled to room temperature and diluted with DCM (200 mL). The
organic layer was washed with water (2 x 50 mL), brine (50 mL), dried (Na2804) and evaporated in vacuo.
The crude material was ed by column chromatography on silica gel ent of hexane:EtOAc 100:0
to 90:10) to afford the title nd as a solid in 35% yield, 900 mg.
1H NMR (400 MHz, CDCI3) 6: .66 (m, 2H), 1.96-1.99 (m, 2H), 3.63 (s, 3H), 6.60 (d, 1H), 7.23 (d,
1H), 8.65 (s, 1H); LCMS (system 10): R = 2.89 min; m/z 252.1 [M+H]+.
Preparation 246: 1-Pyrrolo[2,3-d]pyrimidinyl-cyclopropanecarboxylic acid methyl ester
A solution of 1-(4-chloro-pyrrolo [2,3-d]pyrimidinyl)-cyclopropanecarboxylic acid methyl ester
(Preparation 245, 920 mg, 3.65 mmol) in ethanol (20 mL) was degassed with argon for 15 min.
Ammonium hydroxide (4 mL) and 10% palladium on charcoal were added and the reaction mixture was
stirred at room temperature under hydrogen (balloon pressure) for 5 hours. The reaction mixture was
filtered on a celite bed, washed with ethanol (2 x 10 mL) and the filtrate was evaporated in vacuo. The
crude material was purified by column chromatography on silica gel (gradient of DCM:methanol 100:0 to
98:2) to afford the title compound as a gum in 59% yield, 470 mg.
1H NMR (400 MHz, CDCI3) 6: 1.63-1.66 (m, 2H), 1.95-1.99 (m, 2H), 3.63 (s, 3H), 6.55 (d, 1H), 7.20 (d,
1H), 8.90 (s, 1H), 8.94 (s, 1H); LCMS m 10): R, = 2.22 min; m/z218.2 [M+H]+.
Preparation 247: (1-Pyrrolo[2,3-d]pyrimidinyl-cyclopropyl)-methanol
W0 2012/137089 216
To a stirred solution of 1-Pyrrolo[2,3-d]pyrimidinyl-cyclopropanecarboxylic acid methyl ester
(Preparation 246, 610 mg, 2.80 mmol) in ethanol (15 mL) was added sodium borohydride (318.7 mg,
8.42 mmol) and the e heated to reflux for 16 hours. The reaction mixture was quenched with water
(5 mL) and ted with EtOAc (3 x 10 mL). The combined organic layers were washed with water (5
mL), brine (5 mL), dried (Na2804) and evaporated in vacuo. The crude material was purified by column
chromatography on silica gel (gradient of DCM:methanol 100:0 to 98:2) to afford the title compound as a
light yellow solid in 47% yield, 250 mg.
1H NMR (400 MHz, CDCI3) 6: 1.26 (s, 4H), 3.83 (s, 2H), 6.47 (d, 1H), 7.26 (d, 1H), 8.83 (s, 1H), 8.88 (s,
1H); LCMS (system 10): R = 1.67 min; m/z189.9[M+H]+.
Preparation 248: Tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-pyrrolo[2,3-d]pyrimidine
“3kN/
To a d solution of (1-Pyrrolo[2,3-d]pyrimidinyl-cyclopropyl)-methanol (Preparation 247, 240 mg,
1.27 mmol) in THF (12 mL) was added 3,4-dihydro-2H-pyran (0.46 mL, 5.07 mmol) followed by addition of
PTSA (24 mg, 0.13 mmol). The reaction mixture was stirred at 60°C for 4 hours. The on mixture was
cooled to room temperature, ed with saturated aqueous sodium bicarbonate solution (15 mL) and
extracted with dichloromethane (3 x 25 mL). The combined organic layer was washed with brine (10 mL),
dried over sodium sulphate and evaporated in vacuo. The crude material was purified by column
chromatography on silica gel (gradient of dichloromethane:methanol 100:0 to 98:2) to afford the title
nd as a light brown gum in 90% yield, 320 mg.
1H NMR (400 MHz, CDCI3) 6: 1.18-1.26 (m, 4H), 1.31-1.55 (m, 5H), 1.67-1.69 (m, 1H), 3.28-3.31 (m, 1H),
3.44 (t, 1H), 3.68 (d, 1H), 3.88 (d, 1H), 4.46 (s, 1H), 6.45 (d, 1H), 7.33 (d, 1H), 8.88-8.90 (m, 2H); LCMS
(system 10): R = 2.82 min; m/z 274.6 [M+H]+.
Preparation 249: 5-lodo[1-(tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-pyrrolo[2,3-d]pyrimidine
To a stirred solution of 7-[1-(Tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-pyrrolo[2,3-d]pyrimidine
(Preparation 248, 390 mg, 1.43 mmol) in DMF (8 mL) was added N-iodosuccinimide (481.5 mg, 2.14
mmol) and stirred at room temperature for 4 hours. The reaction mixture was quenched with water (8 mL)
and extracted with ethyl acetate (3 x 20 mL). The combined c layer was washed with water (5 x 15
mL), brine (10 mL), dried over sodium sulphate and evaporated in vacuo. The crude material was purified
by column chromatography on silica gel (gradient of dichloromethane:methanol 100:0 to 99:1) to afford
the title compound as a solid in 79% yield, 450 mg.
W0 2012/137089 217
1H NMR (400 MHz, coc13) 5: 1.13-1.70 (m, 10H), 3.23-3.32 (m, 1H), 3.42 (t, 1H), 3.65 (d, 1H), 3.37 (d,
1H), 4.47 (s, 1H), 7.44 (s, 1H), 3.70 (s, 1H), 3.90 (s, 1H); LCMS m 10): Rt = 3.23 min; m/z 400.2
[M+H]+.
Preparation 250: (5-Bromo-pyridinyl)-{7-[1-(tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-
pyrrolo[2,3-d]pyrim idinyl}-methanone
\ K]
“C \
N N 0
av 0o
To a stirred solution of 5-lodo[1-(tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-pyrrolo[2,3-
midine (Preparation 249, 450 mg, 1.13 mmol) in diethyl ether (6 mL) was added n-butyl lithium (2M
in hexane, 0.62 mL, 1.24 mmol) drop wise at -70°C. Then a solution of 5-Bromo-N-methoxy-N-methyl-
namide (304 mg, 1.24 mmol) in l ether (2.5 mL) was added drop wise at -70°C and stirred at
same temperature for another 30 minutes. The reaction was allowed to warm to room temperature slowly.
The reaction was quenched with saturated aqueous ammonium chloride solution (10 mL) and extracted
with ethyl acetate (3 x 20 mL). The combined organic layer was washed with water (20 mL), brine (20
mL), dried over sodium sulphate and evaporated in vacuo. The crude material was purified by column
chromatography on silica gel (gradient of dichloromethane:methanol 100:0 to 98:2) to afford the title
nd as a light brown sticky solid in 35% yield, 250 mg.
1H NMR (400 MHz, DMSO-D6) 6: .57 (m, 10H), 3.17-3.28 (m, 2H), 3.64 (d, 1H), 3.95 (d, 1H), 4.57
(s, 1H), 8.36 (s, 1H), 8.40 (s, 1H), 8.96 (s, 1H), 8.99 (s, 1H), 9.02 (s, 1H), 9.45 (s, 1H); LCMS (system 10):
R = 3.17 min; m/z 456.8, 459 [M+H]+.
ation 251: (5-Amino-pyridinyl)-{7-[1-(tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}-methanone
\ K1
“1 \ \
kN/ N
To a stirred solution of (5-Bromo-pyridinyl)-{7-[1-(tetrahydro-pyranyloxymethyl)-cyclopropyl]-7H-
pyrrolo[2,3-d]pyrimidinyl}-methanone (Preparation 250, 200 mg, 0.45 mmol) in 1-Methyl-pyrrolidin
one (1.5 mL) was added ammonium hydroxide (15 mL). Then copper (l) oxide (3 mg, 0.02 mmol) was
added and the reaction mixture was heated in a sealed tube at 130°C for 16 hours. The reaction mixture
was cooled to room temperature, extracted with 10% methanol in dichloromethane (5 x 25 mL). The
combined organic layer was dried over sodium sulphate and evaporated in vacuo. The crude material was
W0 2012/137089 218
purified by column chromatography on silica gel (gradient of dichloromethane:methano| 100:0 to 96:4) to
afford the title compound as a white sticky solid in 45% yield, 80 mg.
1H NMR (400 MHz, 6) 8: 1.17-1.53 (m, 10H), .28 (m, 2H), 3.63 (d, 1H), 3.94 (d, 1H), 4.55
(s, 1H), 5.64 (s, 2H), 7.27 (s, 1H), 8.18-8.20 (m, 3H), 8.99 (s, 1H), 9.42 (s, 1H); LCMS (system 10): R =
2.59 min; m/z 394.1 .
Preparation 252: (S)—2-Pyrrolo[2,3-d]pyrimidinyl-propano|
Me\\\\
The title compound was prepared according to the method described for Preparation 8 using (2S)—2-(4-
Chloro-7H-pyrrolo[2,3-d]pyrimidinyl)propano| (Preparation 4) to afford the title compound as a
white solid in 100% yield, 2.8 g.
1H NMR (400 MHz, DMSO-d6) 8: 1.43 (d, 3H), .79 (m, 2H), 4.89-4.94 (m, 1H), 4.97 (t, 1H), 6.61 (d,
1H), 7.71 (d, 1H), 8.75 (s, 1H), 8.97 (s, 1H).
Preparation 253: (2-Hydroxy-1,1-dimethyl-ethyl)-carbamic acid tert-butyl ester
Me36
0 Me
OANH
Mei/OH
To a mixture of 2-Aminomethyl-propano| (2 g, 22.43 mmol) and triethyl amine (3.12 mL, 22.43 mmol)
in THF was added boc-anhydride (4.89 g, 22.43 mmol) slowly at 0°C and was stirred at room temperature
for 16 hours. The reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over
sodium te and evaporated in vacuo to afford the title compound as colourless oil in 92% yield, 3.9
1H NMR (400 MHz, CDCI3) 8: 1.23 (s, 6H), 1.42 (s, 9H), 3.56-3.58 (d, 2H), 4.00 (brs, 1H), 4.62 (brs, 1H).
Preparation 254: (2-Methoxy—1,1-dimethyl-ethyl)-carbamic acid tert-butyl ester
Me36
0 Me
OANH
MekOMe
KOH (3.47 g, 61.82 mmol) was added to a solution of (2-Hydroxy-1,1-dimethyl-ethyl)-carbamic acid tert-
butyl ester (Preparation 253, 3.9 g, 20.6 mmol) in 1,4-dioxane (30 mL) followed by the slow addition of
dimethyl sulphate at room temperature. The mixture was further allowed to stir at room temperature for 48
hours. Reaction mass was filtered through a short pad of celite, washed with DCM (3 x 50 mL). The
W0 2012/137089 219
combined filtrate was washed with water (2 x 50 mL), brine (30 mL), dried over sodium sulphate and
evaporated in vacuo to afford the title compound as yellow oil in 88% yield, 3.7 g.
1H NMR (400 MHz, CDCI3) 6: 1.27 (s, 6H), 1.41 (s, 9H), 3.29 (s, 2H), 3.35 (s, 3H), 4.73 (brs, 1H).
Preparation 255: oxy-1,1-dimethyl-ethylamine
Me#\/0Me
TFA (5.8 mL, 78.7 mmol) was added slowly to a DCM (25 mL) solution of (2-Methoxy-1,1-dimethyl-ethyl)—
carbamic acid tert-butyl ester (Preparation 254, 3.2 g, 15.74 mmol) at 0°C. The mixture was allowed to
stir at room temperature for another 3 hours and then all the volatiles were removed in vacuo. The residue
was treated with aqueous saturated NaHCO3 solution (50 mL) and extracted with a mixture of lPA/DCM
(1:4) (4 x 50 mL). The combined organics was dried over sodium sulphate and evaporated to dryness in
vacuo to afford the title compound as light brown oil in 100% yield, 1.6 g.
1H NMR (400 MHz, CDCI3) 6: 1.32 (s, 6H), 3.30 (s, 2H), 3.35 (s, 3H), 7.80 (brs, 2H).
ation 256: (5-Bromo—2-chloro-pyrimidinyl)-(2-methoxy-1,1-dimethyl-ethyl)—amine
ClAN
N/jBrI
MeJVOMe
Triethyl amine (73.1 mL, 526.6 mmol) was added slowly to a solution of 5-Bromo-2,4-dichloro-pyrimidine
(40 g, 175.5 mmol) in acetonitrile (400 mL) at 0°C and then 2-Methoxy-1,1-dimethyl-ethylamine
(Preparation 255, 23.4 g, 263.3 mmol) was added to the mixture portion wise. The reaction mixture was
stirred for another 16 hours at room temperature. TLC showed the presence of unreacted starting
pyrimidine, but the reaction was not ued further. All the volatiles were removed in vacuo and the
e was taken in ethyl acetate, washed with water, brine, dried over sodium sulphate and evaporated
to s in vacuo. The crude material was ed by column chromatography on silica gel (100-200
mesh, gradient of ethyl acetate:hexane 1:9 to 2:4) to afford the title compound as white solid in 23% yield
(10 g of starting pyrimidine was recovered), 12 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.40 (s, 6H), 3.32 (s, 3H), 3.48 (s, 2H), 6.19 (s, 1H), 8.29 (s, 1H); LCMS
(system 10): R = 3.56 min; m/z 294, 296 [M+H]+.
Preparation 257: [2-Chloro((E)ethoxy-viny|)-pyrimidinyl]-(2-methoxy-1,1-dimethyl-ethyl)—amine
if\OMe N \
Cl N NH
MeJVOMe
The title compound was prepared according to the method bed for ation 61 using (5-Bromo—
2-chloro-pyrimidinyl)-(2-methoxy-1,1-dimethyl-ethyl)—amine (Preparation 256), catechol borane and
ethoxyacetylene (40% in hexane) to afford the title compound as light brown gum in 44% yield, 2.6 g.
W0 2012/137089 220 2012/051363
1H NMR (400 MHz, DMSO-D6) 6: 1.25 (t, 3H), 1.37 (s, 6H), 3.26 (s, 3H), 3.56 (s, 2H), 3.92 (q, 2 H), 5.74
(d, 1H), 6.12 (s, 1H), 6.94 (d, 1H), 7.88 (s, 1H); LCMS (system 10): R = 3.65 min; m/z 286.3 [M+H]+.
Preparation 258: 2-Chloro—7-(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine
CIAN/m3N
MefiflOMe
The title compound was prepared according to the method described for Preparation 62 using [2-Chloro-
ethoxy-vinyl)-pyrimidinyl]-(2-methoxy-1,1-dimethyl-ethyl)—amine (Preparation 257) to afford
the title compound as off white solid in 91% yield, 2g.
1H NMR (400 MHz, DMSO-D6) 6 1.68 (s, 6H), 3.17 (s, 3H), 3.85 (s, 2H), 6.62 (d, 1H), 7.63 (d, 1H), 8.90
(s, 1H); LCMS (system 10): R = 3.29 min; m/z 240 [M+H]+.
Preparation 259: 2-Chloroiodo—7-(2-methoxy—1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine
The title compound was ed according to the method described for Preparation 63 using ro-
7-(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine (Preparation 258) to afford the title
compound as yellowish solid in 75% yield, 2.3 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.67 (s, 6H), 3.17 (s, 3H), 3.83 (s, 2H), 7.81 (s, 1H), 8.64 (s, 1H); LCMS
(system 10): R = 3.65 min; m/z 365.8 [M+H]+.
Preparation 260: (5-Bromo—pyridinyl)-[2-chloro(2-methoxy—1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-
d]pyrimidinyl]-methanone
CIA/N N
The title compound was prepared according to the method described for Preparation 64 using 2-Chloro-
-iodo—7-(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine (Preparation 259) and 5-Bromo-N-
methoxy—N-methyl-nicotinamide to afford the title compound as yellow gum in 34% yield, 900 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.73 (s, 6H), 3.21 (s, 3H), 3.90 (s, 2H), 8.22 (s, 1H), 8.42 (t, 1H), 8.98 (t,
2H), 9.35 (s, 1H).
Preparation 261: no(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]—(5-aminopyridinyl
)-methanone
W0 2012/137089 221
N \ \
JL /
H2N N N
Me+/OMe
The title compound was prepared according to the method described for Preparation 65 using mo—
pyridinyl)-[2-chloro(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]-methanone
(Preparation 260) to afford the title compound as yellow solid in 41% yield, 300 mg.
1H NMR (400 MHz, 6) 6: 1.68 (s, 6H), 3.19 (s, 3H), 3.89 (s, 2H), 5.61 (s, 2H), 6.54 (s, 2H), 7.23
(s, 1H), 7.55 (s, 1H), 8.11 (dd, 2H), 8.93 (s, 1H); LCMS (system 10): R = 2.40 min; m/z 341.2 [M+H]+.
Preparation 262: (2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidine
An anhydrous THF (100 mL) solution of 2-(5-lodo-pyrrolo[2,3-d]pyrimidinyl)methyl-propano|
(Preparation 22) (32 g, 100.91 mmol) was added slowly to a sion of NaH (60% in in oil, 2.98
g, 121.13 mmol) in anhydrous THF (200 mL) at 0°C under nitrogen. The mixture was warmed to room
temperature and stirred for another 30 minutes and again cooled to 0°C and methyl iodide (19 mL, 302.82
mmol) was added drop wise. The reaction mixture was stirred at room temperature for 2 hours,quenched
with aqueous saturated ammonium chloride, and extracted with ethyl acetate. The organic layer was
washed with brine, dried over sodium sulphate and evaporated to dryness in vacuo. The crude material
was purified by column chromatography on silica (100-200 mesh, gradient of hexane:EtOAc 100:0 to
70:30) to afford the title compound as a yellow sticky solid in 22% yield, 7.5 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.70 (s, 6H), 3.15 (s, 3H), 3.89 (s, 2H), 7.77 (s, 1H), 8.70 (s, 1H), 8.82
(s, 1H); LCMS (system 10): R = 3.42 min; m/z 331.6 [M+H]+.
Preparation 263: (5-Bromo—pyridinyl)-[7-(2-methoxy-1,1-dimethyl-ethyl)-7H-pyrrolo[2,3-d]pyrimidin
yl]-methanone
\ :1
m / N
Me%\/OM9
The title compound was prepared according to the method described for ation 64 using 5-lodo
hoxy—1,1-dimethyl-ethyl)—7H-pyrrolo[2,3-d]pyrimidine (Preparation 262) and 5-Bromo—N-methoxy—
N-methyl-nicotinamide to afford the title compound as light brown gum in 51% yield, 3g.
W0 2012/137089 222
1H NMR (400 MHz, DMSO-d6) 6: 1.76 (s, 6H), 3.19 (s, 3H), 3.96 (s, 2H), 8.17 (s, 1H), 8.42 (t, 1H), 8.98
(brs, 2H), 900 (s 1H), 947 (s 1H); LCMS (system 10): R: 3.13 mm m/z 388.6 390.6 [M+H].
Preparation 264: 2-(3,5-Difluoro-pyridinyl)-malonic acid diethyl ester
To a stirred solution of 2,3,5—trifluoro-pyridine (2 g, 15.02 mmol) in dimethyl sulfoxide (20 mL) was added
diethyl malonate (4.60 g, 28.72 mmol). Then cesium ate (9.35 g, 28.72 mmol) was added and the
reaction mixture was heated to 110°C for 16 hours. The reaction mixture was cooled to room temperature
and diluted with ethyl acetate (100 mL). The organic layer was washed with water (2 x 25 mL), brine (25
mL), dried over sodium sulphate and evaporated in vacuo to afford the title compound as oil in 85% yield,
3.5 g.
1H NMR (400 MHz, CDCI3) 6: .30 (m, 6 H), 4.20-4.29 (m, 4 H), 4.88 (s, 1 H), .81 (m, 1 H),
8.01 (s, 1 H).
Preparation 265: (3,5-Difluoro-pyridinyl)-acetic acid
F F
0 /
HO N
To a stirred solution of 2-(3,5-difluoro-pyridinyl)-malonic acid diethyl ester (1 g, 3.56 mmol)
(Preparation 264) in THF (15 mL) was added a solution of lithium hydroxide monohydrate (462 mg, 10.4
mmol) in water (4 mL) se at 0°C and the mixture heated to reflux for 2 hours. The reaction mixture
was cooled to room temperature, acidified (pH ~3) with 2N hloric acid and extracted with 20%
panol-dichloromethane (5 x 20 mL). The combined organic layers were dried (NaZSO4) and
ated in vacuo. The crude material was purified by column chromatography on silica gel (gradient of
DCM: MeOH 100:0 to 95:5) to afford the title compound as a solid in 53% yield, 336 mg.
1H NMR (400 MHz, DMSO-D6) 6 3.32 (s, 2H), 7.74-7.79 (m, 1H), 7.99 (t, 1H); LCMS (system 10): R =
0.65 min; m/z174[M+H]+.
Preparation 266: (4--Nitro-phenyl))-acetic acid ethyl ester
Me“Wm
To a stirred solution of (4--nitro--phenyl))-acetic acid (3 g, 16.4 mmol) in ethanol (30 mL) was added
sulphuric acid (1 mL) and the reaction mixture was heated to reflux for 16 hours. The reaction mixture was
neutralized with 2N aq. NaOH solution and extracted with EtOAc (3 x 50 mL). The combined organic
layers were dried (Na2804) and evaporated in vacuo to afford the title compound as a light yellow oil in
98% yield, 3.4 g.
1H NMR (400 MHz, CDCI3) 6: 1.25 (t, 3H), 3.71 (s, 2H), 4.16 (q, 2H), 7.45 (d, 2H), 8.18 (d, 2H).
Preparation 267: (4-Amino-phenyl)—acetic acid ethyl ester
W0 2012/137089 223
MeAO/K/Q/NHZ
A solution of (4-n-itro--phenyl))-acetic acid ethyl ester (Preparation 266, 3.4 g, 16.1 mmol)'In methanol (100
mL) was degassed with argon for 15 min, treated with 10% palladium on charcoal (700 mg), and then
stirred at room temperature under hydrogen (balloon pressure) for 16 hours. The reaction mixture was
filtered through celite, washed with methanol (2 x 20 mL) and the filtrate was evaporated in vacuo to
afford the title compound as an oil in 96% yield, 2.8 g.
1H NMR (400 MHz, CDCI3) 8: 1.23 (t, 3H), 3.47 (s, 2H), 3.60 (br s, 2H), 4.11 (q, 2 H), 6.63 (d, 2H), 7.05 (d,
2H).
ation 268: (4-Formylamino-phenyl)—acetic acid ethyl ester
Me/\
O fH
A mixture of acetic anhydride (0.33 mL, 3.57 mmol) and formic acid (0.17 mL, 4.46 mmol) was heated at
60°C for 2 hours. The reaction mixture was cooled to 0°C, a solution of (4-amino-phenyl)—acetic acid ethyl
ester (Preparation 267, 500 mg, 2.79 mmol) in THF (10 mL) was added slowly to the on mixture
and the reaction was allowed to stir at room temperature for 16 hours. The reaction mixture was
lized with sat. aq. Na2CO3 solution and extracted with diethyl ether (2 x 25 mL). The ed
organic layers were dried (Na2804) and evaporated in vacuo. The crude material was purified by column
chromatography on silica gel ent of OH 100:0 to 98:2) to afford the title compound as an oil
in 93% yield, 500 mg.
1H NMR (400 MHz, DMSO-d6) 8: 1.17 (t, 3H), 3.59 (s, 2H), 4.05 (q, 2H), 7.20 (d, 2H), 7.52 (d, 2H), 8.25
(d, 1H), 10.15 (s, 1H).
Preparation 269: (4-Methylamino-phenyl)—acetic acid ethyl ester
Me“O/K/Q/Ezvle
To a stirred solution of (4-formylamino-phenyl))-acetic acid ethyl ester ration 268, 500 mg, 2.41
mmol) in THF (10 mL) was added borane-dimethyl sulphide complex (0.3 mL, 3.13 mmol) at 0°C and the
mixture stirred at room temperature for 1 hour. The reaction mixture was quenched with MeOH (5 mL)
and evaporated in vacuo. The crude al was purified by column chromatography on silica gel
(gradient of hexane:EtOAc 100:0 to 75:25) to afford the title compound as an oil in 91% yield, 460 mg.
1H NMR (400 MHz, CDCI3) 8: 1.23 (t, 3H), 2.81 (s, 3H), 3.48 (s, 2H), 3.66 (br s, 1 H), 4.11 (q, 2H), 6.56 (d,
2H), 7.09 (d, 2 H).
Preparation 270: [4-(Methanesulfonyl-methyl-amino)-phenyl]-acetic acid ethyl ester
W0 2012/137089 224
Me/\O/K/Q/(lfvl‘e
To a stirred solution of (4-methylamino-phenyl))-acetic acid ethyl ester (Preparation 269, 418 mg, 2.16
mmol)'In ne (4 mL) was added methanesulfonyl chloride (0.25 mL, 3.24 mmol) and the mixture
stirred at room temperature for 3 hours. The reaction mixture was quenched with water (10 mL) and
extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with water (10 mL), brine
(10 mL), dried (Na2804) and evaporated in vacuo to afford the title compound as a gum in 78% yield, 460
1H NMR (400 MHz, CDCI3) 6: 1.25 (t, 3H), 2.82 (s, 3H), 3.30 (s, 3H), 3.59 (s, 2H), 4.14 (q, 2H), 7.28-7.33
(m, 4H).
Preparation 271: [4-(Methanesulfonyl-methyl-amino)-phenyl]-acetic acid
0 Me
HOWONIS’Me”\\
The title compound was ed according to the method described for ation 265 using [4-
(methanesulfonyl-methyl-amino)-phenyl]-acetic acid ethyl ester (Preparation 270) to afford the title
nd as a solid in 85% yield, 350 mg.
1H NMR (400 MHz, DMSO-D6) 6: 2.92 (s, 3H), 3.21 (s, 3H), 3.55 (s, 2H), 7.28 (d, 2H), 7.33 (d, 2H);
LCMS (system 10): R = 1.05 min; m/z 244 [M+H]+.
Preparation 272: —5-methyl-pyridine
N\Me
To a stirred solution of 2-bromomethyl-pyridine (2 g, 11.6 mmol) in acetonitrile (25 mL) was added
sodium iodide (6.97 g, 46.5 mmol) and the mixture heated to reflux. Acetyl chloride (1.24 mL, 17.44 mmol)
was added se under reflux conditions and the reaction mixture was heated to reflux for 16 hours.
The reaction mixture was cooled to room temperature, quenched with sat. aq. Na2C03 solution (15 mL)
and extracted with EtOAc (3 x 25 mL). The combined organic layers were washed with brine (10 mL),
dried (NaZSO4) and evaporated in vacuo to afford the title compound as an oil in 55% yield, 1.4 g.
LCMS (system 10): R = 3.10 min; m/z 220 [M+H]+.
Preparation 273: 2-(5-Methyl-pyridinyl)-malonic acid diethyl ester
MeAO /
MeAO 0
To a stirred solution of 2-lodo—5-methyl-pyridine (Preparation 272, 1 g, 4.57 mmol) and diethyl malonate
(2.08 mL, 13.70 mmol) in anhydrous dioxane (12 mL) was added cesium carbonate (4.46 gm, 13.7 mmol)
W0 2012/137089 225
and the solution was degassed with argon for 30 min. Cul (174 mg, 0.91 mmol) and picolinic acid (225
mg, 1.83 mmol) were added and the resultant mixture was heated in a sealed tube at 100°C for 16 hours.
The reaction mixture was cooled to room temperature, quenched with water (25 mL) and extracted with
EtOAc (3 x 25 ml). The combined organic layers were washed with water (2 x 10 mL) and brine (10 mL),
dried (Na2804) and evaporated in vacuo. The crude material was purified by column chromatography on
silica gel (gradient of hexane:EtOAc 100:0 to 90:10) to afford the title compound as an oil in 23% yield,
300 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.15-1.18 (m, 6H), 2.29 (s, 3H), 4.12-4.17 (m, 4H), 5.01 (s, 1H),
7.30 (d, 1H), 7.62 (dd, 1H), 8.34 (s, 1H).
Preparation 274: (5-Methyl-pyridinyl)-acetic acid
O /
HO N
The title compound was prepared according to the method described for Preparation 265 using 2-(5-
-pyridinyl)-malonic acid diethyl ester ration 273) to afford the title compound as a solid in
83% yield, 100 mg.
1H NMR (400 MHz, DMSO-D6) 6: 2.26 (s, 3H), 3.64 (s, 2H), 7.21 (d, 1H), 7.53 (d, 1H), 8.29 (s, 1H); LCMS
(system 10): R, = 0.73 min; m/z 152 [M+H]+.
Preparation 275: [1 ,2,4]triazolo[1 ,5-a]pyridinecarboxylic acid
HO / xN
\ N \ N/>
To a on of methyl 2-aminoisonicotinate (28.8 g, 191 mmol) in DMF (97.5 mL) was added DMF-DMA
(70.6 mL, 496 mmol) and the mixture heated to 130 °C for 12 hours. The mixture was then concentrated to
give a residue. To the residue was added MeOH (381 mL), ed by NHZOHSO4 (31.9 g, 248 mmol)
and the resulting mixture was stirred at room temperature overnight. The reaction mixture was
concentrated to give methyl [1 ,2,4]triazolo[1 ,5-a]pyridinecarboxylate in 18% yield, 6g. To a solution of
methyl [1 ,2,4]triazolo[1 ,5-a]pyridinecarboxylate (3 g, 16 mmol) in methanol was added 1M aq. LiOH (70
mL) and the ing e stirred for 10 hours at room temperature. The pH was adjusted to 5-6 using
aq. HCI and the whole mixture extracted with EtOAc (30 mL><3).The combined organic layers were dried
(Na2804) and concentrated in vacuo to give the title nd as a white solid in 38% yield, 1.05 g
1H NMR (400 MHz, DMSO-d6) 6: 7.56-7.58 (m, 1H), 8.32 (m, 1H), 8.66 (m, 1H), .06 (m, 1H), 13.5-
14.0 (s, 1H).
Preparation 276: 1-methyl-1H-pyrrolo[2,3-c]pyridinecarboxylic acid
W0 2012/137089 226
1-methyl-1H-pyrrolo[2,3-c]pyridinecarbaldehyde (WO 05066132) (6.5 g, 40.6 mmol) was dissolved in a
mixture of THF (120 mL) and tert-butyl alcohol (40 mL) under nitrogen. 2-Methylbutene (120 mL of a
2M solution in THF, 46 mmol) was added followed by a solution of NaClOz (11.0 g, 122 mmol) and
NaH2PO4 (21.9 g, 183 mmol) in water (30 mL). The on mixture was stirred at room temperature
overnight under nitrogen. The reaction mixture was concentrated in vacuo to remove organic solvents,
and the residue was filtered. The itate contained the title compound as a white solid in 57% yield,
4.1 g.
1H NMR (400MHz, DMSO-d6) 6: 3.96 (8, 3H), .89 (d, 1H), 8.23 (s, 1H), 8.28 (d, 1H), 8.91 (s, 1H),
12.3 (s, 1H).
ation 277: 2-(4-Cyano-phenyl)-N-{5-[7-(2-trimethylsilanyl-ethoxymethyl)-7H-pyrrolo[2,3-
midinecarbonyl]-pyridinyl}-acetamide
\ / 0
le \ N
\ HW
N/ N
[SJ-Me
Me Me
The title compound was prepared according to the method described for Example 1 using (5-Aminopyridinyl
)-[7-(2-trimethylsilanyl-ethoxymethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]-methanone (Preparation
103) and 4-cyanophenylacetic acid to afford the title compound as an off-white solid in 71% yield, 235 mg.
1H NMR (400 MHz, DMSO-D6) 6: -0.11 (s, 9H), 0.82 (t, 2H), 3.59 (t, 2H), 3.87 (s, 2H), 5.70 (s, 2H), 7.55
(d, 2H), 7.82 (d, 2H), 8.47 (s, 1H), 8.61 (s, 1H), 8.73 (s, 1H), 8.98 (s, 1H), 9.04 (s, 1H), 9.48 (s, 1H), 10.75
(s, 1H); LCMS (System 10): R = 3.25 min; m/z 513 [M+H]+.
Preparation 278: 2-Methylpyrrolo[2,3-d]pyrimidinyl-propane-1,3-diol
N \ \
kN/1%N
MefiOH
The title compound was prepared according to the method described for Preparation 246 using 2-(4-
Chloro-pyrrolo[2,3-d]pyrimidinyl)methyl-propane-1,3-diol (Preparation 217) to afford the title
compound as an ite solid in 93% yield, 800 mg.
1H NMR (400 MHz, DMSO-D6) 6: 1.65 (s, 3H), 3.87-3.91 (m, 2H), 4.10-4.14 (m, 2H), 4.94 (t, 2H), 6.55 (d,
1H), 7.63 (d, 1H), 8.72 (s, 1H), 8.95 (s, 1H).
Preparation 279: N-(5-{7-[2-(tert-Butyl-dimethyl-silanyloxy)-1,1-dimethyl-ethyl]-7H-pyrrolo[2,3-
d]pyrimidinecarbonyl}-pyridinyl)(4-cyclopropyl-pyrazoly|)-acetamide
W0 2012/137089 227
\ / o
le\ \ H/ZgN
N N Me N.
f | N
TBDMSO Me
The title compound was prepared according to the method described for Example 1 using (5-Aminopyridinyl
)-{7-[2-(tert-butyl-dimethyl-silanyloxy)—1,1-dimethyl-ethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}-
methanone (Preparation 38) and (4-Cyclopropyl-1H-pyrazolyl)acetic acid (Preparation 88) to afford
the title compound as an an off-white solid in 41% yield, 55 mg.
1H NMR (400 MHz, 6) 6:-0.24 (s, 6H), 0.46 (m, 2H), 0.60 (s, 9H), 0.79 (m, 2H), 1.67 (m, 1H),
1.75 (s, 6H), 4.11 (s, 2H), 4.98 (s, 2H), 7.26 (s, 1H), 7.52 (s, 1H), 8.16 (s, 1H), 8.46 (s, 1H), 8.71 (d, 1H),
8.91 (d, 1H), 8.99 (s, 1H), 9.47 (s, 1H), 10.75 (s, 1H); LCMS (System 10): R = 3.78 min; m/z 574 [M+H]+.
Preparation 280: Bicyclo[1.1.1]penty|-(5-bromochloro-pyrimidinyl)-amine
jjBr
CI N NH
To a solution of 5-Bromo-2,4-dichloro-pyrimidine (6 g, 26.3 mmol) and bicyclo[1.1.1]pent—1-ylamine (4.7 g,
39.5 mmol) in acetonitrile (60 mL), was added TEA (16.5 mL, 118 mmol), and the mixture was stirred at
°C for 18 hours. The volatiles were removed in vacuo and the residue partitioned between water and
EtOAc. The c phase was dried (Na2804) and evaporated in vacuo. Purification by column
tography on silica gel (EtOAc:Hexane 1:99) afforded the title compound as a white solid in 82%
yield, 4.8 g.
1H NMR (400 MHz, 6) 6: 2.10 (d, 6H), 2.48-2.50 (m, 1H), 8.26 (d, 2H); LCMS m 10): R =
3.64 min; m/z 276 [M+H]+.
Preparation 281: Bicyclo[1 .1 .1]pentyl-[2-chloro((Z)ethoxy-vinyl)-pyrimidinyl]-amine
A?\OEt N \
CI N NH
To a stirred solution of Bicyclo[1.1.1]pentyl-(5-bromochloro-pyrimidinyl)-amine (Preparation 280,
2 g, 7.29 mmol) in dry toluene (70 mL) was added (Z)ethoxy—2-(tributylstannyl)ethene (2.7 mL, 8.03
mmol). The reaction mixture was purged with N2 for 20 min and then Pd(PPh3)4 (421 mg, 0.36 mmol) was
added, followed by degassing for another 20 min and heating to 1100 C under N2 overnight. The on
was cooled to room temperature, quenched with a 2M solution of KF and filtered through a pad of Celite.
The filtrate was partitioned n water (50 mL) and EtOAc (200 mL). The organic phase was washed
with brine (2 x 25 mL), dried (NaZSO4) and evaporated in vacuo.The crude residue was purified by column
W0 2012/137089 228
chromatography on silica gel : Hexane 18: 82) to afford the title compound as a pale green solid in
77% yield, 1.5 g.
1H NMR (400 MHz, DMSO-D6) 6: 1.24 (t, 3H), 2.08 (d, 6H), 2.47 (m, 1H), 3.99 (q, 2H), 5.17 (d, 1H), 6.55
(d, 1H), 7.90 (s, 1H), 8.39 (s, 1H); LCMS (System 10): R = 3.54 min; m/z 266 .
Preparation 282: 7-Bicyclo[1.1.1]pentylchloro-7H-pyrrolo[2,3-d]pyrimidine
842%
The title compound was prepared according to the method described for Preparation 62 using
bicyclo[1.1.1]penty|-[2-chloro((Z)ethoxy-vinyl)-pyrimidinyl]-amine (Preparation 281) to afford
the title compound as an off-white solid in 89% yield, 1.4 g.
1H NMR (400 MHz, DMSO-D6) 6:2.41 (d, 6H), 2.69 (m, 1H), 6.67 (d, 1H), 7.61 (d, 1H), 8.92 (s, 1H);
LCMS (System 10): R = 3.45 min; m/z 220 [M+H]+.
ation 283: 7-Bicyclo[1.1.1]pent—1-ylchloroiodo-7H-pyrrolo[2,3-d]pyrimidine
CI—(/N \ '
N— |
The title compound was prepared according to the method described for Preparation 63 using 7-
bicyclo[1.1.1]pentylchloro-7H-pyrrolo[2,3-d]pyrimidine (Preparation 282) to afford the title compound
as a brown solid in 72% yield, 1.3g .
1H NMR (400 MHz, DMSO-D6) 6: 2.40 (d, 6H), 2.68 (m, 1H), 7.90 (s, 1H), 8.67 (s, 1H); LCMS (System
): R = 3.89 min; m/z 346 [M+H]+.
Preparation 284: (7-Bicyclo[1.1.1]pentylchloro-7H-pyrrolo[2,3-d]pyrimidinyl)—(5-bromo-pyridin
yl)-methanone
The title compound was prepared according to the method described for Preparation 64 using 7-
bicyclo[1.1.1]pentylchloroiodo-7H-pyrrolo[2,3-d]pyrimidine ration 283) to afford the title
compound as an off-white solid in 58% yield, 1.1 g.
1H NMR (400 MHz, DMSO-D6) 6: 2.47 (d, 6H), 2.72 (m, 1H), 8.40-8.42 (m, 2H), 8.99-9.00 (m, 2H), 9.33
(s, 1H); LCMS (System 10): R = 3.73 min; m/z 405 [M+H]+.
Preparation 285: [7-Bicyclo[1.1.1]pentyl(4-methoxy—benzylamino)-7H-pyrrolo[2,3-d]pyrimidinyl]—
(5-bromo-pyridinyl)-methanone
W0 2012/137089 229
To a solution of (7-Bicyclo[1.1.1]penty|chloro-7H-pyrrolo[2,3-d]pyrimidinyl)-(5-bromo-pyridiny|)-
methanone ration 284, 1.1 g, 2.72 mmol) and 4-methoxy benzyl amine (1.06 mL, 8.16 mmol) in
dioxane (40 mL), DIPEA (1.7 mL, 10.8 mmol) was added and mixture was heated at 110°C under
microwave irradiation for 6 hours. The volatiles were removed in vacuo and the residue was partitioned
between water (50 mL) ethyl acetate (150 mL). Organic phase was dried over sodium sulphate,
evaporated in vacuo and purified by column chromatography on silica gel (gradient of EA: Hexane 25: 75)
to afford the title compound as off white solid in 84 % yield, 1.15 g.
1H NMR (400 MHz, DMSO-D6) 6: 2.35 (d, 6H), 2.64 (m, 1H), 3.70 (s, 3H), 4.45 (d, 2H), 6.85 (d, 2H), 7.27
(d, 2H), 7.75 (s, 2H), 8.32 (m, 1H), 8.91-8.94 (m, 2H); LCMS (System 10): R = 3.81 min; m/z 504.2
[M+H]+.
Preparation 286: [5-(Benzhydrylidene-amino)-pyridinyl]-[7-bicyclo[1.1.1]pentyl(4-methoxy-
benzylamino)-7H-pyrrolo[2,3-d]pyrimidinyl]-methanone
To a stirred solution of [7-Bicyclo[1.1.1]pentyl(4-methoxy—benzylamino)-7H-pyrrolo[2,3-d]pyrimidin
-bromo-pyridinyl)—methanone (Preparation 285, 1.18 g, 2 mmol) and benzophenoneimine (0.50
mL, 3 mmol) in dry e (50 mL), cesium carbonate (3.2 g, 10 mmol) was added and the reaction
mixture was purged under N2 for 20 min and then Pd(OAc)2 (45 mg, 0.2 mmol) and BINAP (125 mg, 0.2
mmol) were added followed by degassing for another 10 min and ing overnight. The reaction mass
was cooled to room temperature and filtered through a pad of Celite. The filtrate was partitioned between
water (25 mL) and EtOAc (100 mL). The c phase was washed with brine (10 mL), dried (NaZSO4)
and evaporated in vacuo. Purification by column tography on silica gel (EtOAc:Hexane 25:75)
afforded the title compound as a yellow solid in 85 % yield, 1.1g .
1H NMR (400 MHz, DMSO-D6) 6: 2.36 (d, 6H), 2.65 (m, 1H), 3.70 (s, 3H), 4.44 (d, 2H), 6.84 (d, 2H),
7.24-7.29 (m, 4H), 7.36-7.37 (m, 3H), 7.50-7.52 (m, 4H), 7.58 (m, 1H), 7.70-7.72 (m, 3H), 8.15 (d, 1H),
8.48 (d, 1H), 8.89 (s, 1H); LCMS (System 9): R = 4.02 min; m/z 605 [M+H]+.
ation 287: (5-Amino-pyridinyl)—[7-bicyclo[1.1.1]pentyl(4-methoxy—benzylamino)-7H-
pyrrolo[2,3-d]pyrimidinyl]-methanone
W0 2012/137089 230
To a solution of [5-(Benzhydrylidene-amino)-pyridinyl]-[7-bicyclo[1.1.1]pent—1-yl(4-methoxy—
benzylamino)-7H-pyrrolo[2,3-d]pyrimidinyl]-methanone (Preparation 286, 475 mg, 0.78 mmol) in THF
(15 mL), citric acid (15 mL of a 1N aq. solution) was added at room temperature and the reaction mixture
stirred for 2 hours. The reaction was then quenched with sat. aq. NaZCO3 solution and extracted with
EtOAc (2 x 25 mL). The ed organic phases were dried (NaZSO4) and ated in vacuo.
Purification by column chromatography on neutral alumina (Methanol:DCM 3: 97) to afford the title
compound as a pale yellow solid in 92 % yield, 320 mg.
1H NMR (400 MHz, DMSO-D6) 6: 2.36 (d, 6H), 2.64 (m, 1H), 3.70 (s, 3H), 4.45 (d, 2H), 5.59 (s, 2H), 6.84
(d, 2H), 7.22 (m, 1H), 7.28 (d, 2H), 7.57 (s, 1H), 7.72 (br s, 1H), 8.10-8.12 (m, 2H), 8.92 (s, 1H); LCMS
(System 10): R = 3.10 min; m/z 441 [M+H]+.
Preparation 288: 1-Cyclobutyl-1H-imidazolecarboxylic acid hydrochloride
COZH
<5 HCI
Ethyl 3-(dimethylamino)isocyanoacrylate (WO 2007042545) (45 g, 0.27 mol) was added to
utylamine (50 g, 0.70 mol) and heated to reflux for 2 hours. The solution was then cooled and
concentrated. The residue was purified by column chromatography over silica gel (3:1 Heptane).
The oily residue was ated with TBME:heptane (1:1) and the resulting solid was collected and dried,
giving 1-cyclobutyl-1H-imidazolecarboxylic acid ethyl ester (35 g, 67%, second crop not harvested). 1-
Cyclobutyl-1H-imidazolecarboxylic acid ethyl ester (35 g, 0.21 mol) was dissolved in 6 N HCI (300 mL)
and refluxed for 1 day. The solution was trated to dryness in vacuo. The solid was azeotroped
with toluene, triturated with toluene and then dried under vacuum, giving the title compound 88% yield,
37.2 g, m/z 167 [M+H]+.
Preparation 289: (2-Methylimidazo[2,1-b][1,3]thiazolyl)acetic Acid Hydrochloride
)1 >¢N OH
8 HCI
W0 2012/137089 231
Ethyl 4-bromooxobutanoate (92 g, 0.35 mol) was added to a solution of 5-methylthiazolamine (40 g,
0.35 mol) in e (400 mL). The e was left to stand overnight and then evaporated to give 2-
amino(4-ethoxy-2,4-dioxobutyl)methylthiazolium bromide in 82% yield, 101 g. 2-amino(4-
ethoxy-2,4-dioxobutyl)methylthiazolium bromide (101g, 0.28 mol) was then refluxed in ethanol
(250 mL) for 2 hours. The solvent was then evaporated to give ethyl 2-(2-methylimidazo[2,1-b]thiazol
yl)acetate hydrobromide as yellow crystals in 97% yield, 85 g. Solid K2003 was added to a solution of
ethyl 2-(2-methylimidazo[2,1-b]thiazolyl)acetate hydrobromide (85 g, 0.28 mol) in water (300 mL) to
pH ~8. The product was extracted with form (3 X 100 mL), and the combined extracts were dried
over NaZSO4 and evaporated to give ethyl 2-(2-methylimidazo[2,1-b]thiazolyl)acetate in 84% yield 52 g.
Ethyl 2-(2-methylimidazo[2,1-b]thiazolyl)acetate (52 g, 0.23 mol) was refluxed in 10% aqueous HCI
(150 mL) for 2 hours. The solution was evaporated to dryness to give the title compound as brown
crystals in 59% yield 32.3 g; m/z 197 [M+H]+.
Preparation 290: 3-Methyl(trifluoromethyl)-1H-pyrazolecarboxylic acid
HO Me
F / \
F H
To a solution of benzyl cetate (57.6 g, 300 mmol) in MeOH (50 mL) in a 350 mL pressure vessel
was added HZNMe (150 mL of a 2M solution in MeOH, 300 mmol) and acetic acid (2 mL). The capped
vessel was placed in an oil bath at 70 oC and the reaction mixture was stirred for 16 hours. After the
mixture had cooled to room temperature, the solvent was evaporated in vacuo leaving a yellow on,
which was dissolved in EtOAc (500 mL) and MgSO4 was added to remove water. The drying agent was
filtered off, and the solvent evaporated in vacuo to give benzyl hylamino)butenoate as a viscous
yellow oil in quantitative yield 60 g which was used without further purification.
To a solution of benzyl 3-(methylamino)butenoate (60 g, 300 mmol) and pyridine (27 mL, 330 mmol)
in THF (500 mL), cooled to —20 0C, was added triflic anhydride (45 mL, 315 mmol) over a 30 min period.
During the addition, the temperature was kept below -10 oC. The reaction mixture was allowed to warm to
room temperature overnight ing in a yellow clear on mixture. The solvent was evaporated in
vacuo and the orange residue was taken up in water (1L) and EtZO (1L). Upon g the mixture well in
a 3-L separatory funnel, all solid material dissolved. The organic layer was separated and washed with
water (3 x 500 mL), brine (500 mL) and dried over MgSO4. Filtration and evaporation of the solvent in
vacuo provided benzyl 3-(Methylamino)(trifluoroacetyl)butenoate as an ite solid in 97% yield,
90 g that was used without additional purification.
To a solution of benzyl 3-(methylamino)(trifluoroacetyl)but—2-enoate (90 g, 300 mmol) in a mixture of
THF (900 mL) and acetic acid (100 mL) was added hydrazine monohydrate (14.6 mL, 300 mmol) over a 5
min period. The on mixture was heated to reflux for 3 hours, allowed to cool to room temperature
and the solvents were evaporated in vacuo to afford a bright yellow mass. The mass was dissolved in
EtOAc (1L) and water (1L) with gentle heating, allowed to cool to room temperature and the aqueous
layer was neutralized to pH 7-8 with NaHCO3. This mixture was transferred to a 3-L separating funnel, the
W0 2012/137089 232
layers were mixed vigorously, separated and the organic layer was washed with water (3 x 500 mL) dried
(MgSO4), filtered and ated in vacuo to give benzyl 3-methyl(trifluoromethyl)-1H-pyrazole
carboxylate as an off-white solid in 89% yield 74 g that was used without r purification.
A mixture of benzyl 3-methyl(trifluoromethyl)-1H-pyrazolecarboxylate (74 g, 285 mmol), Pd/C (45 g,
% on C) and EtOAc (1L) was treated in a Parr apparatus at room temperature with H2 (15 psi) for 5
hours. The st was filtered off over Celite and the solvent was removed in vacuo. EtZO (1 L) and water
(200 mL) were added and the aqueous layer was made slightly basic (pH 8—9) with NaZCO3. The layers
were ted and the aqueous layer was extracted with EtZO (5 x 200 mL). To the aqueous layer was
added dropwise conc. HCI until pH 4-5 and followed by extraction with EtZO (3 x 500 mL). The combined
organic layers were dried (MgSO4), filtered, evaporated in vacuo to give the title compound in 74% yield
7.4 g; mp. 308-310 °C(dec.)m/z195[M+H]+
Preparation 291: Ethyl [4-(3-Hydroxyphenyl)-1H-1,2,3-triazolyl]acetate
Me\/0\(\N\ _\N—N
Ethyl azidoacetate (14.9 g, 115 mmol) was dissolved in tert—butanol (200 mL), and 95% 3-
hydroxybenzonitrile (13.7 g, 110 mmol) was added. A solution of sodium ascorbate (2.18 g, 11 mmol) in
water (100 mL) was added, followed by a 0.3 M solution of copper sulfate under argon. The mixture was
stirred at room temperature for 12 hours. The solution was evaporated to dryness in vacuo, the residue
was dissolved in EtOAc (100 mL), dried (MgSO4), ed, evaporated in vacuo to give the title compound
as brown crystals in ~100% (27.7 g) yield.
MS m/z 246 [M-H]'
Preparation 292 : [4-(3-Hydroxyphenyl)-1H-1,2,3-triazolyl]acetic Acid Hydrate
W0 2012/137089 233
Ethyl [4-(3-Hydroxyphenyl)-1H-1,2,3-triazolyl]acetate (Preparation 291, 27.2 g, 0.110 mol) was
dissolved in methanol (200 mL), and a solution of NaOH (4.84 g, 121 mmol) in water (40 mL) was added.
The solution was kept at room ature for 24 hours. Methanol was evaporated in vacuo, water (120
mL) was added, and the solution was ed with activated charcoal. The mixture was filtered, and
11 M HCI was added to the filtrate. The e was dissolved by addition of water (120 mL) and cooled to
carry out the crystallization. The crystals were filtered, washed with water (2 x 25 mL), and ated to
dryness to afford the title compound as brown crystals (mp 186.3-188.7 °C) in 99.0% yield 23.9 9. MS m/z
218 [M-H]'
Preparation 293: Ethyl [4-(1-hydroxycyclopentyl)-1H-1,2,3-triazolyl]acetate
Me\\
Of’l \ OH
Ethyl azidoacetate (2.58 g, 20 mmol) was ved in tert—butanol (15 mL), and 1-
hydroxycyclopentanecarbonitrile (2.20 g, 20 mmol) was added. A solution of sodium ascorbate (0.792 g, 4
mmol) in water (10 mL) followed by a 0.3 M solution of copper sulfate (0.67 mL) was added to the mixture,
and stirring continued at room temperature for a further 48 hours. The solution was evaporated to dryness
in vacuo and the e was dissolved in EtOAc (50 mL), dried (M9804), filtered and evaporated in
vacuo to afford the title compound as green-yellow crystals in 98% yield, 4.69 9.
MS m/z 238 [M-H]'
Preparation 294: [4-(1-hydroxycyclopentyl)-1H-1,2,3-triazolyl]acetic acid
HO\(\N\ \ OH
To a solution of ethyl [4-(1-hydroxycyclopentyl)-1H-1,2,3-triazolyl]acetate (Preparation 293, 36.4 g, 151
mmol) in water (50 mL), was added a solution of NaOH (7.60 g, 190 mmol) in water (25 mL). The solution
was refluxed with activated charcoal, filtered through Celite, and NaHSO4 (25.8 g, 190 mmol) added.
EtOAc (50 mL) was added to the filtrate. The formed precipitate was separated by filtration, dissolved in
EtOAc (100 mL), and the solution was filtered. The water layer was extracted with EtOAc (10 X 50 mL),
and the combined extracts were evaporated to a volume of 100 mL. The itate was filtered, washed
with ethyl e (2 x 50 mL), and concentrated under reduced pressure to afford the title compound as a
ess crystalline substance in 87% yield, 27.8 9. mp 1240-1260 0C; MS m/z 212 [M+H]+
Preparation 295: Di-tert-butyl 1-(1-methyl-1H-pyrazolyl)hydrazine-1,2-dicarboxylate
W0 2012/137089 234
To a solution of 4-iodomethyl-1H-pyrazole (21 g, 0.12 mol) in dry ether (200 mL) at -78 °C was added
n-BuLi (84.5 mL of a 2.5 M solution in hexane, 0.18 mol) over a period of 30 min and the mixture stirred
for a further 30 min. A solution of t-butyl (Z)-diazene-1,2-dicarboxylate (30.4 g, 0.12 mol) in ether
(100 ml) was added to the reaction mixture over a period of 10 min and the resultant mixture stirred at -
78°C for 1 hour. The reaction was warmed to 0 °C and quenched with ice-water and ted with ether
(3 x 100mL). The organic layer was separated, dried (NaZSO4) and evaporated in vacuo. The crude
material was washed with hexane and then dried under vacuum to give the title compound in 30% yield,
11.2 g.
1H NMR (400 MHz, DMSO-d6): d 9.60 (s, 1 H), 7.62 (s, 1 H), 7.25 (s, 1 H), 3.76 (s, 3 H), 1.44 (m, 18 H).
LCMS: 313 (M+H) +
Preparation 296: 3-tert-butyl-1'-methyl-1'H-1,4'-bipyrazolamine
/ \
N NH
N-N‘
A mixture of di-tert-butyl 1-(1-methyl-1H-pyrazolyl)hydrazine-1,2-dicarboxylate (Preparation 295 ,12.3
g, 0.039 mol) and 4,4-dimethyloxopentanenitrile (5.4 g, 0.043 mol) was dissolved in MeOH (36 mL) and
HCI (12 mL) was added slowly. The reaction mixture was stirred at 65 °C for 16 hours. The reaction was
led to remove MeOH and basified with sat. aq. NaHCO3 solution to pH~8 followed by extraction with
DCM. The organic layer was ted, dried (NaZSO4) followed by concentration in vacuo. The crude
mixture was ed by column tography on silica gel (hexane:EtOAc 50:50) to give the title
compound in 41% yield, 7.2 g.
1H NMR (400 MHz, DMSO-d6) d 7.91 (s, 1 H), 7.58 (s, 1 H), 5.28 (s, 1 H), 5.04 (s, 2 H), 3.81 (s, 3 H), 1.21
(s, 9 H); LCMS: m/z 220 [M+H]+.
Preparation 297: 3-cyclopropyl-1'-methyl-1'H-1,4'-bipyrazolamine
W0 2012/137089 235
The title compound was prepared according to the method described for Preparation 296: using di-tert-
butyl 1-(1-methyl-1H-pyrazolyl)hydrazine-1,2-dicarboxylate (Preparation 295) and opropyl
oxopropanenitrile to afford the title compound in 48% yield, 1.7g.
1HNMR (400 MHZ, DMSO-d6): 0.55-0.57 (m, 2H), .80 (m, 2H), .72 (m, 1H), 3.83(s, 3H), 5.08-
5.09 (m, 3H), 7.57(s, 1H), 7.90(s, 1H).
LCMS: [M+H]+ 204
Preparation 298: N-[5-({7-[3-({[tert-butyl(dimethyl)silyl]oxy}methyl)oxetanyl]-7H-pyrrolo[2,3-d]pyrimidin-
-yl}carbonyl)pyridinyl]—2-(4-chlorophenyl)acetamide
\/ 0
/ NWCI
KN N
OTBDMS
The title compound was prepared according to the method described for Preparation 223 using (5-
aminopyridinyl)(7-(3-methyloxetanyl)—7H-pyrrolo[2,3-d]pyrimidinyl)methanone (Preparation 222)
to afford the title compound as a white solid in 87% yield, 35 mg.
LCMS (System 2): R = 1.49 min; m/z 592 [M+H]+.
Preparation 299: 5-(methoxymethyl)methyl-1H-pyrazolecarboxylic acid
Me
To a on of ethyl 5-(methoxymethyl)methyl-1H-pyrazolecarboxylate (W09743277) (177 mg,
0.893 mmol) in MeOH, a 0.5 M solution of LiOH was added (5.3 mL, 2.68 mmol). The reaction was stirred
at room temperature ght. The on was evaporated to dryness and the residue was adjusted to
pH5 with a 2 N HCI solution. The aqueous solution was extracted with EtOAc and the organic layer was
dried over NaZSO4 and evaporated in vacuo to give the title compound as a white solid 95%, 145mg.
1H NMR (400MHz, methanol-d4) 6.68 (s, 1 H), 4.44 (s, 2 H), 3.84 (s, 3 H), 3.29 (s, 3 H). MS m/z 171
[M+H]+
Preparation 300: lmidazo[1,2-a]pyrimidinecarboxylic acid
N N
HO \ NJ
A mixture of 6-bromoimidazo[1,2-a]pyrimidine (1.17 g, 6 mmol), BINAP (18 mg, 0.06 mmol), PdCl2 (6 mg)
in MeOH (30 mL) and triethylamine (1.8 mL) was heated to 80°C under CO (50 psi) for 12 hours in DMF
W0 2012/137089 236
(97.5 mL). A (70.6 mL, 495.8 mmol) was added and the mixture heated to 130 °C for 12 hours.
The mixture was filtered and concentrated to give methyl imidazo[1,2-a]pyrimidinecarboxylate (365 mg,
%) as a yellow solid which was used in the next step without further purification. To a solution of methyl
imidazo[1,2-a]pyrimidinecarboxylate (365 mg, 2.1 mmol) in methanol was added 1M aq. LiOH (9.0 mL)
and the resulting mixture was stirred for 10 hours at room temperature. The pH was adjusted to 5-6 using
aq. HCI and the whole mixture ted with EtOAc (3x30mL).The ed organic layers were dried
over NaZSO4 and evaporated in vacuo to give the title compound as white solid in 39% yield, 133 mg
1H NMR (400 MHz, DMSO-d6): 69.15 (m, 1H), 8.8 (m, 1H), 7.9 (m, 1H), 7.65 (m, 1H),
Preparation 301: [1 ,2,4]triazolo[1 ,5-a]pyridinecarboxylic acid
HO / /N
\ N \
To a solution of methyl 2-aminoisonicotinate (28.8 g, 191 mmol) in DMF (97.5 mL) was added DMF-DMA
(70.6 mL, 496 mmol) and the mixture heated to 130 °C for 12 hours. The mixture was then concentrated to
give a residue. To the residue was added methanol (381 mL), ed by NH2OHSO4 (31.9 g, 248
mmol) and the resulting e was stirred at room temperature overnight. The reaction mixture was
concentrated to give methyl [1,2,4]triazolo[1,5-a]pyridinecarboxylate in 18% yield, 6g. To a solution of
methyl [1 riazolo[1 ,5-a]pyridinecarboxylate (3 g, 16 mmol) in methanol was added 1M aq. LiOH (70
mL) and the resulting mixture stirred for 10 hours at room temperature. The pH was adjusted to 5-6 using
aq. HCI and the whole mixture extracted with EtOAc (30 mL><3).The combined organic layers were dried
over NaZSO4 and concentrated in vacuo to give the title compound as white solid in 38% yield, 1.05 g
1H NMR (400 MHz, DMSO-d6): 613.5-14.0 (s, 1H), 9.04-9.06 (m, 1H), 8.66 (m, 1H), 8.32 (m, 1H), 67.56-
7.58 (m, 1H).
Preparation 302: 1-methyl-1H-pyrrolo[2,3-c]pyridinecarboxylic acid
| \
1-methyl-1H-pyrrolo[2,3-c]pyridinecarbaldehyde (WO 05066132) (6.5 g, 40.6 mmol) was dissolved in a
mixture solvent of THF (120 mL) and t-butyl alcohol (40 mL) under nitrogen. A 2M 2-methylbutene
solution in THF (120 mL, 46 mmol) was added followed by a solution of NaClOz (11.02 g, 122 mmol) and
4 (21.9 g, 183 mmol) in water (30 mL). The reaction mixture was d at room temperature
overnight under nitrogen. The reaction mixture was concentrated in vacuo to remove organic solvents,
and the e was filtered. The precipitate ned the title compound as white solid in 57% yield, 4.1
g.
1H NMR: DMSO-d6 400MHz: 6 3.96 (S, 3 H), 7.87-7.89 (d, 1 H), 8.23 (s, 1 H), 8.28 (d, 1 H), 8.91 (s, 1 H),
12.3 (s, 1 H).
Preparation 303: 2-((tert-butoxycarbonyl)amino)(4-chlorophenyl)acetic acid
W0 2012/137089 237
To 4-chlorophenylglycine (1.50 g, 8.08 mmol) and sodium hydroxide (0.65 g, 16.2 mmol) in water (20 mL)
was added di-tert—butyl onate (1.76 g, 8.08 mmol) in acetonitrile (15 mL) and the mixture was stirred
at room temperature for 18 hours. The mixture was then washed with DCM (20 mL) and acidified using
2N HCI. The resulting aqueous layer was extracted with DCM (2 x 25 mL) and the combined organic
layers were washed with brine before being dried over MgSO4, filtered and evaporated in vacuo to give
the title compound as a colourless oil in 87% yield, 2.00 g.
Preparation 304: tert-Butyl (1-(4-chlorophenyl)((5-(7-isopropyl-7H-pyrrolo[2,3-d]pyrimidine
carbonyl)pyridinyl)am ino)oxoethyl)carbamate
0 HN
\,( 3‘0
N \ \ Me
kN/| N MeaM/e
)‘Me
Me
To pyridine (4 mL) in a sealed vessel was added 2-((tert-butoxycarbonyl)amino)(4-chlorophenyl)acetic
acid (67 mg, 0.24 mmol) (see Preparation 303), (5-aminopyridinyl)(7-isopropyl-7H-pyrrolo[2,3-
d]pyrimidinyl)methanone (52 mg, 0.18 mmol) (see Preparation 95) and N,N,N',N'-tetramethyl-O-(7-
azabenzotriazolyl)uronium hexafluorophosphate (105 mg, 0.28 mmol). The reaction was heated at
50°C for 18 hours and then evaporated in vacuo and purified by column chromatography (gradient of
100:0 to 88:12 DCM:MeOH) to give the title compound in 49% yield, 64 mg. LCMS (basic): R = 0.81 min;
m/z 549 .
Biological Activity
lsolated TRK Enzyme assays use the HTRF KinEASE-TK kit (Cisbio Cat# 62TKOPEJ) with recombinant
His-tagged cytoplasmic s of each TRK receptor sourced from lnvitrogen (see table . This
activity-assay measures the phosphorylation of ne es within a substrate from the HTRF kit
which has been validated by Cisbio for a variety of tyrosine kinases including the TRK receptors.
Assay details:
W0 37089 238
-Target Invitrogen Cat# FAC enzyme FAC ATP—Assay Reaction Time
TRKA PV3144 aa 441-796 4nM 40uM 35min
(NTRK1)
I-- --PV3616 aa 526-838 1nM 1.4uM
I-----TRKC PV3617 aa 510-825 10nM 15uM 30min
0.5mM stock solutions of test compounds are prepared and serially diluted in 100% DMSO. A standard
curve using the compound of Example 135 disclosed in W02005/116035 of 150uM is also prepared on
each test plate. High percentage effect (HPE) is defined by 150uM PF00 and 0% effect (ZPE)
is defined by 100% DMSO. Greiner low volume black plates containing 0.2ul of serially diluted compound,
standard and HPE/ZPE are d using the Bravo nanolitre dispenser.
1X enzyme buffer is ed from 5X Enzymatic Buffer from the Cisbio KinEASE TK kit using MilliQ
water. The buffer is then mented with 10mM MgCl and 2mM DTT (both from Sigma). In the case
of TRKB, the buffer is also supplemented with 125nM Supplement Enzymatic Buffer (SEB) from the
Cisbio kit.
2X FAC of enzyme and 2X FAC ATP diluted in 1X complete enzyme buffer is incubated at room
temperature for 20minutes to preactivate the enzyme. Following this preactivation step, 5ul/well of
enzyme + ATP mix is added using a Multidrop Micro to the assay plate, spotted with 0.2ul 100% DMSO
compound. This is left for 20mins at room temperature before adding 5ul of 2uM TK—substrate-Biotin
(from the Cisbio kit) diluted in 1X enzyme buffer (1uM FAC) using the Multidrop Micro. The on is
incubated at room temperature for the optimized assay reaction time (see table). The reaction is stopped
by adding 10u|/well HTRF Detection Buffer containing 0.25uM Streptavidin-XL665 (0.125uM FAC) and
1:200 TK Antibody-Cryptate using a Multidrop.
After the Detection Reagent addition, plates are covered and incubated at room temperature for 60
minutes. HTRF signal is read using an Envision reader, measured as a ratio of emissions at two different
wavelengths, 620nm and 665nm. Any compound that inhibits the action of the TRK kinase will have a
lower fluorescence ratio value 665/620nM than compounds which do not inhibit theTRK kinase. Test
compound data are sed as percentage inhibition defined by HPE and ZPE values for each plate.
Percentage inhibition in the ce of test compound is plotted against compound concentration on a
log scale to ine an |C50 from the resultant sigmoid curve.
Cell Based Assays were carried out using Cell lines from DiscoveRx utilising their PathHunter technology
and reagents in an antagonist assay:
DiscoveRx cell line Cat# e rophin
TRKA co sed with p75 93-0529C3 —
W0 2012/137089 239 2012/051363
TRKB 93-0463C3 BDNF
TRKB co expressed with p75 93-0530C3 BDNF
TRKC 93-0464C3 NT3
TRKC co expressed with p75 93-0531C3 NT3
The assays are based upon eRx’s etary Enzyme Fragment Complementation (EFC)
logy. In the case of the TRK cell lines, the enzyme acceptor (EA) n is fused to a 8H2 protein
and the TRK receptor of interest has been tagged with a Prolink tag.
Upon neurotrophin binding, the TRK receptor becomes phosphorylated, and the tagged 8H2 protein
binds. This results in functional complementation and ed B-Galactosidase activity which is can be
measured using the luminescent Galacton Star substrate within the PathHunter reagent kits.
Generally, small molecule inhibitors bind to the kinase domain so are not competing with the neurotrophin
(agonist) which binds to an ellular site. This means that the |C50 is a good e of affinity and
should be unaffected by concentration neurotrophin stimulant.
Cryopreserved PathHunter cells are used from either in-house produced batches or bulk s bought
directly from DiscoveRx. Cryopreserved cells are resuscitated, spun 1000rpm for 4min to remove
ng media, and resuspended in MEM + 0.5% horse serum (both lnvitrogen) to 5e5cells/ml. The cells
are then plated using a Multidrop into Greiner white tissue culture treated plates at 20u|/well and
incubated for 24h at 37°C, 5% C02, high ty. On the day of the assay, the cell plates are allowed to
cool to room temperature for 30min prior to the assay.
4mM stock solutions of test compounds are prepared and serially diluted in 100% DMSO. A standard
curve using the compound of Example 135, WO2005/116035 at a top concentration of 150uM is also
prepared on each test plate. High percentage effect (HPE) is d by 150uM of the compound of
Example 135, WO2005/116035 and 0% effect (ZPE) is defined by 100% DMSO. Plates containing 1u| of
serially diluted compound, standard and HPE/ZPE are diluted 1/66 in assay buffer (PBS minus Ca2+,
minus Mg2+ with 0.05% pluronic F127) using a Wellmate. Using a Platemate Plus, 5ul of 1/66 diluted test
compounds is then transferred to the cell plate and allowed to reach equilibrium by incubating for 30min at
room temperature before addition of agonist stimulus: 10u|/well of 2nM (0.571nM FAC) of the cognate
neurotrophin (Peprotech) diluted in agonist buffer (HBSS with 0.25% BSA). Final assay concentration of
the test compounds is , (the compound of Example 135, WO2005/116035 FAC is 0.325uM). The
plates are left at room temperature for a further 2hours before addition of 10u| of the DiscoveRx
PathHunter detection reagent (made up by adding 1 part Galacton Star, 5 parts Emerald II and 19 parts
Cell Assay Buffer as per the manufacturer’s instructions).
After reagent addition, plates are covered and incubated at room temperature for 60 minutes.
Luminescence signal is read using an Envision. Test compound data are expressed as percentage
inhibition d by HPE and ZPE values for each plate. Percentage inhibition in the presence of test
W0 37089 240
compound is plotted against compound tration on a log scale to determine an |C50 from the
resultant sigmoid curve.
Brain Penetration Assays
In Vitro
MDCK-BCRP: MDCK-BCRP data were collected according to the method described in “A 96-Well Efflux
Assay To Identify ABCG2 Substrates Using a Stably Transfected MDCK ll Cell Line”
http://pubs.acs.org/doi/full/10.1021/mpO50088t
Yongling Xiao, Ralph Davidson, Arthur Smith, Dennis Pereira, Sabrina Zhao, John Soglia, David Gebhard,
Sonia de , and David B. Duignan, Mol. Pharm. 3 (1), pp 45—54.
, 2006,
MDCK-MDR1: MDCK-MDR1 data were collected according to the method described in “Are MDCK Cells
Transfected with the Human MDR1 Gene a Good Model of the Human lntestinal Mucosa? “
http://wwwspringerlink.com/content/gfhglgbr4fnp3khf/fulltext.pdf
Fuxing Tang, Kazutoshi Horie, and Ronald T. rdt, Pharmaceutical Research, Vol. 19, No. 6, June
2002.
In Vivo
Brain penetration was measured according to the method described in ”Assessing brain free fraction in
early drug ery”. Read, K; Braggio, 8., Expert Opinion Drug Metab Toxicol. (2010) 6 (3) 337- 344.
Example TrkA enzyme TrkB enzyme TrkA cell TrkB cell TrkC cell MDCK MDRl MDCK MDRl RRCK BCRP RRCK BCRP
number potency (nM) potency (nM) y (nM) potency (nM) potency (nM) Papp AB (x10— Papp BA (x10— Papp AB (x10—6 Papp BA (x10—
6 cm/sec) 6 cm/sec) cm/sec) 6 cm/sec)
1 3.7 9.5 1.5 5.5 14
2 8.4 113 5.3 3.9 1.1 <1 15
3 4.1 5.1 1.8 8.7 20
4 3.9 162 5.7 6.5 4.4 <1 13 12 39
2.6 73 3.5 3.6 2.0 1.5 14
6 2.5 2.0 1.3 6.9 16
7 11 41 2.1 0.8 5.6 20
8 2.4 15 1.9 2.0 2.3 1.8 15
9 3.7 94 13 6.6 5.9 <1 20 9 43
5.7 78 30 18 18 <1 14
11 3.3 197 17 11 6.6 <1 23
12 9.2 547 68 22 <1 3.1
13 20 449 1130 184 <1 1.0
14 13 773 41 15 4.8 <1 21
17 514 143 40 <1 15
16 16 445 99 15
17 23 103 45 <1 13
18 128 >8660 5300 <1 <1
19 249 5480 1840 206 <1 8.5
14 635 22 19 <1 17
21 71 5740 1770 <1 1.3
22 25 596 190 <1 6.9
23 26 93 52 <1 31
24 125 427 37 27 8.9 <1 10
24 433 65 19 <1 13
26 3.5 30 14 5.9 8.6 <1 16 7.5 32
27 2.8 55 3.9 5.4 3.2 <1 17
28 4.9 75 2.7 1.9 <1 20
29 2.8 68 5.8 4.0 1.3 23
4.0 98 11 7.5 <1 15
31 18 401 198 <1 2.6
32 9.0 423 87 59 18 <1 3.4
33 5.7 96 2.0 1.6 1.0 24
34 3.6 200 23 25 13 <1 11
8.9 223 103 37 <1 5.3
36 4.5 303 11 13 13 <1 12
37 67 2250 571
38 7.2 1060 589 <1 2.1
39 24 >8660 4710 <1 <1
40 67 2580 506 <1 2.4
41 7.6 203 70 <1 6.3
42 3.8 191 44 27 13 <1 9.5
43 5.1 136 17 19 11 <1 13
44 12 996 554 <1 1.6
45 25 >8660 3790 <1 <1
45 3.2 94 6.0 4.2 0.9 <1 18 7 31
47 25 27 35 13 <1 21
48 1.7 40 2.9 3.6 2.5 <1 12
49 2.1 37 3.1 2.1 2.1 <1 17
50 6.6 143 10.0 2.1 <1 24
51 7.8 198 35 7.5 9.9 <1 11
52 3.1 103 22 16 8.1 <1 9.1
53 4.0 163 16 5.0 8.6 <1 13
54 6.7 368 174 <1 1.3
55 7.0 179 12 4.5 <1 20
56 9.5 962 451 517 1.1 1.0
57 1.2 40 4.0 2.5 5.1 26
58 2.1 315 11 11 1.2 54
59 4.6 110 3.4 2.4 5.7 24
60 1.8 13 1.6 0.9 <1 18
61 4.5 100 13 8.4 1.4 <1 39
62 1.8 41 1.9 2.7 5 99
63 1.2 25 2.6 1.8 0.4 5.6 20
54 1.5 19 2.0 1.4 <1 4.2
W0 37089 242
Trka enzyme Trka enzyme Trka enzyme
71-5
94-1
67 49-1
300 174
976 230
1080
283 6270
567 233 1840
133
203 22 5840 166 110
23 44s 3470
392 24 244 702
44s 3400
908 26 3100
355 27 1280
28 206 3840
29 9810
284 240
831 2590
681 3100
330 3100
312 3080
334 3100
496 3100
1850
40 579
4 67-4
42 719
43 2630
we 44 277
101 204 45 836
102 46 573 820
103 147 873
104 148 173
105 149 3570
406 404
107 232 835
108 791
W0 37069 243
Trka enzyme Trka enzyme Trka enzyme
197 430 242 267 4350
198 243 288 43-1
199 244 289 1370
200 245 290 167
201 462 246 291 3230
202 412 247 292 37.7
203 248 293 20-2
204 249 294 61-4
205 250 295 31-9
206 123 25 296 47.7
207 252 297 299
206 129 253 296 1460
209 272 254 299 539
210 255 300 52-2
211 256 301 428
212 257 302 156
256 303 1220
259 304 3900
260 305 2640
26 126 306 N/D
262 307 N/D
263 306 592
264 309 53.9
265 -=__ 98-4
223 268 579
224 269 557
225 343
233 323
234 439 324
235 496 325
236 326
237 327
236 297 326
239 329
240 330
241 331
W0 37089 244
Trka enzyme Trka enzyme Trka enzyme
332 381 426 228
333 382 427 93-4
334 383 428
336 384 429 2740
337 385 430 297
338 386 431 235
339 251 387 432 2790
340 451 388 433 248
343 389 434
344 328 390 435 5490
345 382 39 246 438 205
348 253 392 437 107
347 393 438 312
348 394 439 22-5
350 395 440 676
333 333 443 3333
352 397 442 36-6
353 N/D 398 443 47.7
354 N/D 399 444 498
355 302 400 593 445 5.51
356 40 446 812
357 302 402 447 919
358 403 448 272
359 404 449 155
360 958 405 450 3150
361 406 451 7170
362 407 452 542
383 408 854 453 834
364 409 454 633
365 455 1070
366 354 456 6960
367 412 457 674
368 753 458 213
369 414 459 4-06
370 460 9800
371 461 9800
372 432 3323
373 463 186
374 224 434 3333
375 420 465 2130
376 421 466 400
377 422 451 487 538
378 423 468 5100
379 424 469 523
380 422 425 798 470
W0 37069 245
Trka enzyme Trka enzyme Trka enzyme
471 562 3-75
472 563 3-11
473 564 1-57
474 520 565 17-9
475 643 52 567 20.6
476 522 566 12.1
477 523 569 34.4
476 524 570 6970
479 525 571 2.92
460 526 572 9.5
461 527 573 17.9
482 526 574 14.2
463 529 575 3.09
464 530 576 296
53 577 1-9
466 532 576 6.07
487 533 579 8-11
488 534 580 0-825
489 535 581 3-21
490 536 582 9-79
491 537 583 0-681
492 538 584 581
493 646 539 565 36.7
494 540 586 1000
495 54 N/D 567 20.5
496 542 N/D 566 334
497 543 589 31-3
498 544 590 71-2
499 545 591 142
500 546 —_
501 201 547 —_
502 548 —_
503 963 549 —_
504 550 —_
505 55 —_
506 552 —_
507 553 —_
508 554 270 _
509 555 —_
510 249 556 —_
511 144 557 —_
512 558 —_
559 374 _
514 560 —_
561——
W0 2012/137089 246
All publications cited in this application are each herein incorporated by reference in their entirety.
Although the invention has been bed above with reference to the disclosed embodiments, those
skilled in the art will readily iate that the specific experiments detailed are only illustrative of the
invention. It should be understood that various cations can be made without departing from the
spirit of the invention. Accordingly, the invention is limited only by the following claims.
Claims (30)
1. A compound of a I: N R101 N X R102 R3 N N 5 (I) or a pharmaceutically able salt thereof, wherein R1 is H, or C1-5 alkyl optionally substituted by up to 3 substituents independently selected from OH, 10 CON(R5R6), SO2R7, SR7, OR7, CH2OH, CO2R5, SONR7R7, NR7SO2R5, CN, NO2 and R8 , or a ring system selected from C3-5 cycloalkyl, propellanyl, or a 4-6 membered saturated heterocyclyl ring, which ring system has up to 3 ring hetero-atoms selected from N, O and S, and which ring system is optionally substituted by up to 3 substituents independently selected from , OH, CON(R5R6), SO2R7, OR7, CH2OH, CO2R5, SONR7R7, NR7SO2R5, CN, NO2 and R8; R2 is H or methyl; R3 is H, NH2 or NH(C1-3 alkyl optionally substituted with up to 3 substituents ndently selected from OH and O(C1-3 alkyl)); R101 is H, OH, methyl, cyclopropyl, methoxy, ethyl, ethoxy or CN, X is a bond, O, (CH-R4)n, NR104, OCH2 or CH2O; 25 R4 is independently H, CH3, CH2OH, CH2OCH3, OH, NH2, NHCH3, N(CH3)2, CH2NH2, CH2NHCH3,or CH2N(CH3)2; R104 is H, C1-3 alkyl or a C4-6 saturated carbocycle, each of which is ally substituted by up to 3 substituents independently selected from C1-3 alkyl, CH2OH and NH2; n is 1 or 2; R102 is a ring system which is a 3-7 membered monocyclic carbocyclic or heterocyclic system, or an 8membered bicyclic system, which ring system may be saturated or partially or fully unsaturated, wherein the heterocyclic ring system may have up to 5 ring hetero-atoms selected from N, S, and O, wherein the bicyclic ring system can be 2 rings (carbocyclic-carbocyclic, carbocyclic-heterocyclic, 5 heterocyclic-carbocyclic or heterocyclic-heterocyclic) fused or linked by a single bond, which ring system is optionally substituted by up to 3 substituents independently selected from, where possible - halo, CN, NR5R6, SO 7, SR7, C 2R 1-4 alkyl ally substituted by up to 3 OH and/or C 1-3 alkoxy groups, 10 C3-6 cycloalkyl optionally substituted by up to 3 OH and/or C1-3 alkoxy groups, C1-3 alkyl substituted by up to 3 halogen, OH, O(C1-3 alkyl), O(C3-6 cycloalkyl optionally substituted by up to 3 OH and/or C1-3 alkoxy groups, O(C1-3 alkyl substituted by up to 3 halogen), O(C1-3 alkyl substituted by up to 3 OH and/or C1-3 alkoxy groups), NR5SO 7, =O, R8, C(O)R8, NO 5CO 7, NR5COR 7,OR 8, S(O)R7, 2R 2, NR 2R and CH2R8; R5 and R6 are each independently H, or C1-5 alkyl optionally substituted by up to 3 substituents independently selected from OH, CONR 7R7, SO 7, 7, CN, NO 9 , 2R OR7, CH2OH, CO2R7, SONR7R7, NR7SO 2R 2 and R 20 or a ring system selected from C3-5 cycloalkyl, propellanyl, or a 4-6 ed ted heterocyclyl ring, which ring system is optionally substituted by up to 3 substituents independently selected from OH, CON(R7R7), SO2R7, CO2R7, SONR7R7, NR7SO 7, CN, NO 7R7,SR 7,C 2R 2, halo, NR 1-4 alkyl ally tuted by up to 3 OH and/or C1-3 alkoxy groups, C3-6 cycloalkyl optionally tuted 25 by up to 3 OH and/or C1-3 alkoxy groups, C1-3 alkyl substituted by 1 to 3 halogen, O(C3-6 cycloalkyl optionally substituted by up to 3 OH and/or C1-3 alkoxy groups, O(C1-3 alkyl substituted by up to 3 halogen, O(C1-3 alkyl substituted by up to 3 OH and/or C1-3 alkoxy, NR7SO 7,=O,NO 7CO 7, 2R 2,NR 2R and S(O)R7, or R5 and R6 together with the N to which they are ed can be a 4-7 ed ring optionally 30 including up to 2 further ring hetero-atoms independently selected from N, O, S, which ring is optionally substituted by C1-3 alkoxy and / or C1-3 alkyl; R7 is H, C1-5 alkyl or C1-5 alkoxy, which C1-5 alkyl or C1-5 alkoxy is optionally substituted by up to 3 tuents independently selected 35 from halogen; R8 is a is a ring system which is a 3-7 membered monocyclic carbocyclic or heterocyclic system, or an 8membered bicyclic , which ring system may be saturated or partially or fully unsaturated, wherein the heterocyclic ring system may have up to 5 ring hetero-atoms selected from 40 N, S, and O, wherein the bicyclic ring system can be 2 rings (carbocyclic-carbocyclic, carbocyclic-heterocyclic, heterocyclic-carbocyclic or heterocyclic-heterocyclic) fused or linked by a single bond, which ring system is optionally substituted by up to 3 substituents independently selected from, where possible - halo, CN, NR5R6, SO2R7, SR7, C1-4 alkyl optionally tuted by up to 3 OH and/or C1-3 alkoxy groups, 5 C3-6 cycloalkyl optionally tuted by up to 3 OH and/or C1-3 alkoxy groups, C1-3 alkyl substituted by 1 to 3 halogen, OH, O(C1-3 alkyl), O(C3-6 cycloalkyl optionally substituted by up to 3 OH and/or C1-3 alkoxy groups, O(C1-3 alkyl substituted by up to 3 halogen, O(C1-3 alkyl substituted by up to 3 OH and/or C1-3 alkoxy, NR5SO 7, =O, NO 7COR 7,NR 5CO 7, and ; 2R 2, NR 2R 10 R9 is a is a ring system which is a 3-7 membered monocyclic carbocyclic or heterocyclic system, or an 8membered bicyclic system, which ring system may be saturated or partially or fully unsaturated, wherein the heterocyclic ring system may have up to 5 ring hetero-atoms selected from N, S, and O, n the bicyclic ring system can be 2 rings (carbocyclic-carbocyclic, yclic-heterocyclic, 15 heterocyclic-carbocyclic or cyclic-heterocyclic) fused or linked by a single bond, which ring system is optionally substituted by up to 3 substituents ndently selected from, where possible - halo, CN, NR7R7, SO2R7, SR7, C1-4 alkyl optionally substituted by up to 3 OH and/or C1-3 alkoxy groups, 20 C3-6 cycloalkyl optionally substituted by up to 3 OH and/or C1-3 alkoxy , C1-3 alkyl substituted by 1 to 3 halogen, OH, O(C1-3 alkyl), O(C3-6 cycloalkyl ally substituted by up to 3 OH and/or C1-3 alkoxy groups, O(C1-3 alkyl substituted by up to 3 halogen, O(C1-3 alkyl substituted by up to 3 OH and/or C1-3 alkoxy, NR7SO 7, =O, NO 7CO 7, NR7COR 7,and S(O)R7; 2R 2, NR 2R 25 wherein each CH moiety can be replaced by a CF moiety.
2. A compound or salt according to claim 1 wherein R 1 is H, C 1-5 alkyl optionally substituted by up to 2 OH, or R1 is C1-5 alkyl substituted by CONH2, CONHCH3, CON(CH3)2, CO2H, CO2CH 3, OCH3, SCH3, 30 SO 2CH 3, or R1 is a ring system selected from C3-5 cycloalkyl, propellanyl, or oxetanyl, which ring system is optionally substituted by methyl, OH or CH2OH.
3. A compound or salt according to any one of claims 1 or 2 wherein R1 is t-butyl, hydroxy-t- 35 butyl, dihdyroxy-t-butyl, 1-hydroxypropyl or 1,3-dihydroxypropyl.
4. A nd or salt according to any one of claims 1 to 3 wherein R2 is H.
5. A compound or salt according to any one of claims 1 to 4 wherein R3 is H or NH2.
6. A compound or salt according to any one of claims 1 to 5 wherein R3 is NH2.
7. A compound or salt according to any one of claims 1 to 5 wherein R3 is H.
8. A compound or salt according to any one of claims 1 to 7 n R101 is H. 5
9. A compound or salt according to any one of claims 1 to 7 wherein R101 is OH.
10. A compound or salt according to any one of claims 1 to 9 wherein X is a bond, O, CH 2, C2H4, CH(CH 3)CH 2, CH(CH3), CH(CH2OH), CH2O, CH(NH2), CH(OH) or NH. 10
11. A compound or salt according to any one of claims 1 to 10 wherein X is CH2.
12. A compound or salt according to any one of claims 1 to 11 wherein R102 is an optionally substituted nitrogen-containing ring system which is linked to the X moiety via a en ring atom.
13. A compound or salt according to any one of claims 1 to 11 wherein R102 is an optionally substituted ring system where the ring system is selected from - benzimidazolyl, benzisoxazolyl, benzofuranyl, benzoxazolyl, benzotriazolyl, biphenyl, bipyrazolyl, cinnolinyl, cyclobutylimidazolyl, cyclobutylpyrazolyl, cyclobutylthiazolyl, entyltriazolyl, 20 cyclopropylisoxazolyl, cyclopropyloxazolyl, cyclopropylpyrazolyl, cyclopropyltriazolyl, diazirenylphenyl, dihydronaphthyridinyl, dihydropyrrolopyrazolyl, dioxinopyridinyl, furazanyl, ridinyl, furopyrrolyl, imidazolyl, imidazopyrazinyl, imidazopyridazinyl, imidazopyridinyl, imidazopyrimidinyl, othiadiazolyl, 25 imidazothiazolyl, indanyl, indazolyl, indolyl, isoindolyl, isoxazolopyridinyl, isoxazolyl, isoquinolinyl, naphthyridinyl, oxazolyl, phenyl, phenylcyclopropyl, imidazolyl, phenylpyrazolyl, phenylpyrrolyl, phenyltetrazolyl, phthalazinyl, purinyl, nyl, pyrazolyl, pyrazolopyridinyl, pyrazolopyrimidinyl, pyrazolotriazinyl, pyridinyl, pyridazinyl, pyridinyltriazolyl, pyrimidinyl, pyrroloimidazolyl, pyrrolopyrazinyl, 30 opyrimidinyl, pyrrolopyridinyl, pyrrolyl, quinolinyl, quinazolyl, quinoxalinyl, tetrahydrobenzisoxazolyl, tetrahydrocyclopentapyrazolyl, ydrotriazolopyridinyl, tetrazolopyridazinyl, olopyridinyl, thiazolyl, thiazolopyridinyl, thiazolopyrimidinyl, thienylpyrazolyl, thienopyridinyl, triazolopyridinyl and triazolyl.
14. A compound or salt according to claim 13 where the optional substituents are independently selected from, where possible - halo, methyl, ethyl, propyl, pyl, cyclopropyl, CF3, CHF2, CH2F, CH2OCH 3, CN, CH2OH, OCH3, =O, NH2, SCH3, SO2CH 3, phenoxy, fluorophenoxy, , SCF3, OCF3, SO2CF 3, NHSO2CH 3, 40 NHSO 2CF 3, C(O)CF3, C(O)CH3, benzoyl, azetidinylmethyl, fluoroazetidinylmethyl and morpholinomethyl.
15. A compound or salt according to any one of claims 1 to 11, 13 or 14, wherein R102 is selected from phenyl, pyrazolyl, 1,2,3-triazolyl, benzotriazolyl, pyridinyl, nyl and pyridinyl, each of which is optionally substituted by halo, methyl, ethyl, propyl, isopropyl, cyclopropyl, CF3, 5 CHF2, CH2F, CH2OCH3, CN, CH2OH, OCH3, =O, NH2, SCH3, SO2CH3, phenoxy, fluorophenoxy, benzyl, SCF3, OCF3, SO2CF3, NHSO2CH3, NHSO2CF3, C(O)CF3, C(O)CH3, l, azetidinylmethyl, fluoroazetidinylmethyl and/or morpholinomethyl.
16. A nd or salt according to any one of claims 1 to 15 with R5 and R6 groups present, 10 wherein R5 and R6 are each independently H, C1-3 alkyl ally substituted by C1-3 alkoxy, C3-5 lkyl, propellanyl, oxetanyl, tetrahydrofuranyl or pyranyl, or R5 and R6 together with the N to which they are attached can be an azetidine, pyrrolidine, piperidine, piperazine or morpholine ring, which ring is optionally substituted by C1-3 alkoxy and / or C1-3 alkyl.
17. A compound ing to claim 1 of the Formula IA: N R101 O R102 R3 N N (IA) or a pharmaceutically acceptable salt thereof, wherein R3 is H or NH2; R1 is C2-4 alkyl optionally substituted by 1 or 2 OH groups; R101 is H or OH; and R102 is phenyl or an aromatic or partially unsaturated 5- or 6-membered heterocycle, which heterocycle is optionally fused to a further phenyl or 5-7 membered aromatic or partially unsaturated heterocyclic ring, n each heterocycle has from 1 to 3 ring heteroatoms selected from N, O and 30 and which ring system is ally substituted by up to 3 substituents independently selected from halo, CF3, C1-4 alkyl and C3-5 cycloalkyl.
18. A compound or salt according to claim 17 wherein R101 is H.
19. A compound or salt according to claim 18 wherein R1 is t-butyl, hydroxy-t-butyl or 1-hydroxypropyl; and R102 is 4-trifluromethylphenyl, 4-chlorophenyl, 2,4-difluorophenyl, 5-chloropyridinyl, 5- 5 fluoropyridinyl, 3-trifluromethylpyrazolylyl, 4-trifluromethylpyrazolyl, 3-trifluromethyl methylpyrazolyl, 3-cyclopropylpyrazolyl, opropylpyrazolyl, 4-trifluromethyl (1,2,3-triazolyl), 4-cyclopropyl- (1,2,3-triazolyl), or benzotriazolyl. 10 20. A compound ing to claim 1, selected from: N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin yl)[4-(trifluoromethyl)phenyl]acetamide; N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin yl]carbonyl}pyridinyl)(4-chlorophenyl)acetamide; 15 N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin yl]carbonyl}pyridinyl)(5-fluoropyridinyl)acetamide; N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin bonyl}pyridinyl)[3-(trifluoromethyl)-1H-pyrazolyl]acetamide; N-(5-{[2-amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin 20 yl]carbonyl}pyridinyl)(3-cyclopropyl-1H-pyrazolyl)acetamide; N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(4- cyclopropyl-1H-1,2,3-triazolyl)acetamide; N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}[4- (trifluoromethyl)-1H-pyrazolyl]acetamide; 25 N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}[4- (trifluoromethyl)-1H-1,2,3-triazolyl]acetamide; N-{5-[(2-aminotert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(5- chloropyridinyl)acetamide; N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin 30 yl]carbonyl}pyridinyl)(5-chloropyridinyl)acetamide; N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}[4-(trifluoromethyl)- 1H-1,2,3-triazolyl]acetamide; 2-(4-chlorophenyl)-N-[5-({7-[(1S)hydroxymethylethyl]-7H-pyrrolo[2,3-d]pyrimidin yl}carbonyl)pyridinyl]acetamide 35 N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}[4-(trifluoromethyl)- azolyl]acetamide; N-[5-({7-[(1S)Hydroxymethylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyl]- trifluoromethyl)phenyl]acetamide; N-[5-({7-[(1R)hydroxymethylethyl]-7H-pyrrolo[2,3-d]pyrimidinyl}carbonyl)pyridinyl]- 40 2-[4-(trifluoromethyl)phenyl]acetamide; 2-(4-chlorophenyl)-N-[5-({7-[(1R)hydroxymethylethyl]-7H-pyrrolo[2,3-d]pyrimidin yl}carbonyl)pyridinyl]acetamide; N-(5-{[7-(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridinyl) [5-methyl(trifluoromethyl)-1H-pyrazolyl]acetamide; 2-(5-chloropyridinyl)-N-(5-{[7-(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin yl]carbonyl}pyridinyl)acetamide; 5 N-(5-{[2-Amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin yl)(4-chlorophenyl)acetamide; [2-amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin yl)[4-(trifluoromethyl)phenyl]acetamide; N-(5-{[2-amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin 10 yl)(4-chlorophenyl)acetamide; N-(5-{[2-amino(2-hydroxymethylethyl)-7H-pyrrolo[2,3-d]pyrimidinyl]carbonyl}pyridin yl)[4-(trifluoromethyl)phenyl]acetamide; N-(5-{[2-Amino(2-hydroxy-1,1-dimethylethyl)-7H-pyrrolo[2,3-d]pyrimidin bonyl}pyridinyl)[5-methyl(trifluoromethyl)-1H-pyrazolyl]acetamide; 15 and N-{5-[(7-tert-butyl-7H-pyrrolo[2,3-d]pyrimidinyl)carbonyl]pyridinyl}(4-cyclopropyl-1H- 1,2,3-triazolyl)acetamide; or a pharmaceutically acceptable salt thereof.
20
21. A pharmaceutical ition comprising a compound of the formula (I) or a pharmaceutically acceptable salt thereof, as defined in any one of the preceding claims 1 to 20, and a pharmaceutically acceptable carrier.
22. A compound of the formula (I) or a pharmaceutically acceptable salt thereof, as defined in 25 any one of claims 1 to 20, for use as a medicament.
23. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as d in any one of claims 1 to 20 for use in the treatment of a disease for which an Trk receptor antagonist is ted.
24. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined in any one of claims 1 to 20 for use in the treatment of pain.
25. The use of a compound of the formula (I) or a pharmaceutically acceptable salt or 35 composition thereof, as d in any one of claims 1 to 20, for the manufacture of a medicament to treat a disease for which an Trk receptor nist is indicated.
26. The use of a compound of the formula (I) or a pharmaceutically acceptable salt or composition thereof, as defined in any one of claims 1 to 20, for the manufacture of a 40 medicament to treat pain.
27. A compound or salt according to any one of claims 1 to 20 for use in a medical treatment in ation with a further drug susbtance.
28. A compound according to claim 1, substantially as herein described with reference to any 5 one of the examples.
29. A pharmaceutical composition according to claim 21, substantially as herein described with nce to any one of the examples. 10
30. The use according to claim 25 or claim 26, substantially as herein described with reference to any one of the examples.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161471758P | 2011-04-05 | 2011-04-05 | |
US61/471,758 | 2011-04-05 | ||
PCT/IB2012/051363 WO2012137089A1 (en) | 2011-04-05 | 2012-03-22 | Pyrrolo [2, 3 -d] pyrimidine derivatives as inhibitors of tropomyosin- related kinases |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ615557A NZ615557A (en) | 2014-08-29 |
NZ615557B2 true NZ615557B2 (en) | 2014-12-02 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CA2832291C (en) | Pyrrolo[2,3-d)pyrimidine tropomyosin-related kinase inhibitors | |
ES2525581T3 (en) | N-sulfonylbenzamide derivatives useful as voltage-dependent sodium channel inhibitors | |
JP2013531030A (en) | N-sulfonylbenzamide as an inhibitor of voltage-gated sodium channels | |
JP7566888B2 (en) | Azole-fused pyridazin-3(2H)-one derivatives | |
ES2976515T3 (en) | Compounds and compositions for treating conditions associated with APJ receptor activity | |
CA2885253A1 (en) | Pyrrolo[2,3-d]pyrimidine tropomyosin-related kinase inhibitors | |
US9163021B2 (en) | Pyrrolo[3,2-c]pyridine tropomyosin-related kinase inhibitors | |
TW201546060A (en) | TROPOMYOSIN-RELATED kinase inhibitors | |
JP6837072B2 (en) | 6,7-Dihydro-5H-pyrazolo [5,1-b] [1,3] Oxazine-2-carboxamide compound | |
EP2903986A1 (en) | Tropomyosin-related kinase inhibitors | |
NZ615557B2 (en) | Pyrrolo [2,3-d] pyrimidine derivatives as inhibitors of tropomyosin- related kinases | |
TW201410677A (en) | Condensed ring heterocyclic compound | |
OA16607A (en) | Pyrrolo [2, 3 -D] pyrimidine derivatives as inhibitors of tropomyosin- related kinases. | |
WO2016009296A1 (en) | N-acylpiperidine ether tropomyosin-related kinase inhibitors | |
TW201617334A (en) | N-acylpyrrolidine ether tropomyosin-related kinase inhibitors |