WO2013078254A1 - Bicyclic heteroaryl derivatives as kinase inhibitors - Google Patents
Bicyclic heteroaryl derivatives as kinase inhibitors Download PDFInfo
- Publication number
- WO2013078254A1 WO2013078254A1 PCT/US2012/066153 US2012066153W WO2013078254A1 WO 2013078254 A1 WO2013078254 A1 WO 2013078254A1 US 2012066153 W US2012066153 W US 2012066153W WO 2013078254 A1 WO2013078254 A1 WO 2013078254A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- phenyl
- purin
- cyclobutanamine
- pyrrolo
- pyridin
- Prior art date
Links
- 229940043355 kinase inhibitor Drugs 0.000 title description 4
- 239000003757 phosphotransferase inhibitor Substances 0.000 title description 4
- 150000001875 compounds Chemical class 0.000 claims abstract description 314
- -1 resolved enantiomers Chemical class 0.000 claims abstract description 150
- 150000003839 salts Chemical class 0.000 claims abstract description 46
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 claims abstract description 16
- 239000012453 solvate Substances 0.000 claims abstract description 15
- KZZKOVLJUKWSKX-UHFFFAOYSA-N cyclobutanamine Chemical compound NC1CCC1 KZZKOVLJUKWSKX-UHFFFAOYSA-N 0.000 claims description 132
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 94
- 238000000034 method Methods 0.000 claims description 92
- LWABFMLTBBNLTA-UHFFFAOYSA-N cyclobutyl carbamate Chemical compound NC(=O)OC1CCC1 LWABFMLTBBNLTA-UHFFFAOYSA-N 0.000 claims description 84
- 238000011282 treatment Methods 0.000 claims description 79
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 59
- 108091008611 Protein Kinase B Proteins 0.000 claims description 54
- 102100033810 RAC-alpha serine/threonine-protein kinase Human genes 0.000 claims description 36
- 125000003118 aryl group Chemical group 0.000 claims description 32
- 201000010099 disease Diseases 0.000 claims description 32
- 239000003814 drug Substances 0.000 claims description 31
- 239000008194 pharmaceutical composition Substances 0.000 claims description 30
- 208000035475 disorder Diseases 0.000 claims description 27
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 27
- 241000124008 Mammalia Species 0.000 claims description 25
- 125000001424 substituent group Chemical group 0.000 claims description 25
- 229940002612 prodrug Drugs 0.000 claims description 24
- 239000000651 prodrug Substances 0.000 claims description 24
- 230000000694 effects Effects 0.000 claims description 23
- 229910052739 hydrogen Inorganic materials 0.000 claims description 22
- 229910052757 nitrogen Inorganic materials 0.000 claims description 22
- 229910052799 carbon Inorganic materials 0.000 claims description 20
- 125000005843 halogen group Chemical group 0.000 claims description 19
- 230000001404 mediated effect Effects 0.000 claims description 19
- 239000002207 metabolite Substances 0.000 claims description 19
- 125000001072 heteroaryl group Chemical group 0.000 claims description 18
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims description 12
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 12
- 125000000246 pyrimidin-2-yl group Chemical group [H]C1=NC(*)=NC([H])=C1[H] 0.000 claims description 12
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 11
- 229910052760 oxygen Inorganic materials 0.000 claims description 10
- HNJBEVLQSNELDL-UHFFFAOYSA-N pyrrolidin-2-one Chemical compound O=C1CCCN1 HNJBEVLQSNELDL-UHFFFAOYSA-N 0.000 claims description 10
- 108091000080 Phosphotransferase Proteins 0.000 claims description 9
- 102000020233 phosphotransferase Human genes 0.000 claims description 9
- 229910052717 sulfur Inorganic materials 0.000 claims description 9
- 238000002560 therapeutic procedure Methods 0.000 claims description 9
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 8
- 125000005842 heteroatom Chemical group 0.000 claims description 8
- 230000003463 hyperproliferative effect Effects 0.000 claims description 8
- 238000004519 manufacturing process Methods 0.000 claims description 8
- 125000004043 oxo group Chemical group O=* 0.000 claims description 8
- 125000006570 (C5-C6) heteroaryl group Chemical group 0.000 claims description 7
- 229910003827 NRaRb Inorganic materials 0.000 claims description 6
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Chemical compound OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 claims description 6
- 230000002401 inhibitory effect Effects 0.000 claims description 6
- JHHZLHWJQPUNKB-UHFFFAOYSA-N pyrrolidin-3-ol Chemical compound OC1CCNC1 JHHZLHWJQPUNKB-UHFFFAOYSA-N 0.000 claims description 5
- ALGKHDUXQICRBG-UHFFFAOYSA-N 1-[4-(1-phenylpyrrolo[3,2-c]pyridin-2-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C=2N(C3=CC=NC=C3C=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 ALGKHDUXQICRBG-UHFFFAOYSA-N 0.000 claims description 4
- LYUWDUOAKPBYLL-UHFFFAOYSA-N 1-[4-(2-phenylpyrrolo[2,3-c]pyridin-1-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=CN=CC=C3C=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 LYUWDUOAKPBYLL-UHFFFAOYSA-N 0.000 claims description 4
- LCYUCWBMENPEQG-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-n-methyl-8-phenylpurin-2-amine Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(NC)=NC=C2N=C1C1=CC=CC=C1 LCYUCWBMENPEQG-UHFFFAOYSA-N 0.000 claims description 4
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 claims description 4
- HNQIVZYLYMDVSB-UHFFFAOYSA-N methanesulfonimidic acid Chemical compound CS(N)(=O)=O HNQIVZYLYMDVSB-UHFFFAOYSA-N 0.000 claims description 4
- MEMZWCGZAAJIQO-UHFFFAOYSA-N 1-[4-(1-aminocyclobutyl)phenyl]-2-phenyl-5h-pyrrolo[3,2-c]pyridin-4-one Chemical compound C=1C=C(N2C3=C(C(NC=C3)=O)C=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 MEMZWCGZAAJIQO-UHFFFAOYSA-N 0.000 claims description 3
- CLNKXCWICABYOS-UHFFFAOYSA-N 1-[4-[2-(2-fluorophenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CN1N=CC=C1C1=NC2=CN=C(C=3C(=CC=CC=3)F)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 CLNKXCWICABYOS-UHFFFAOYSA-N 0.000 claims description 3
- XIFVRSXGXGGWTE-UHFFFAOYSA-N 1-[4-[6-(1-methylpyrazol-4-yl)-8-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C1=NN(C)C=C1C1=NC=NC2=C1N=C(C=1C=CC=CC=1)N2C1=CC=C(C2(N)CCC2)C=C1 XIFVRSXGXGGWTE-UHFFFAOYSA-N 0.000 claims description 3
- GTBYWWACKDCQBF-UHFFFAOYSA-N 1-[9-[4-(1-aminocyclobutyl)phenyl]-8-pyridin-2-ylpurin-2-yl]ethane-1,2-diamine Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(C(N)CN)=NC=C2N=C1C1=CC=CC=N1 GTBYWWACKDCQBF-UHFFFAOYSA-N 0.000 claims description 3
- KYVMBXLEPHOOKK-UHFFFAOYSA-N 2-[4-[9-[4-(1-aminocyclobutyl)phenyl]-8-pyridin-2-ylpurin-2-yl]pyrazol-1-yl]ethanol Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2N=CC=CC=2)C2=CN(CCO)N=C2)C=CC=1C1(N)CCC1 KYVMBXLEPHOOKK-UHFFFAOYSA-N 0.000 claims description 3
- 125000003349 3-pyridyl group Chemical group N1=C([H])C([*])=C([H])C([H])=C1[H] 0.000 claims description 3
- GHVXXXKPBRGDPY-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-n,n-diethyl-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-4-amine Chemical compound C=1C=2C(N(CC)CC)=NC=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=N1 GHVXXXKPBRGDPY-UHFFFAOYSA-N 0.000 claims description 3
- DEKGRKVQNZFBFC-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-n,n-dimethyl-8-phenylpurin-6-amine Chemical compound N=1C=2C(N(C)C)=NC=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=C1 DEKGRKVQNZFBFC-UHFFFAOYSA-N 0.000 claims description 3
- LEGKZPBPACKLOR-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-n-methyl-8-phenylpurine-2-carboxamide Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(C(=O)NC)=NC=C2N=C1C1=CC=CC=C1 LEGKZPBPACKLOR-UHFFFAOYSA-N 0.000 claims description 3
- 230000002526 effect on cardiovascular system Effects 0.000 claims description 3
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 claims description 3
- 230000002757 inflammatory effect Effects 0.000 claims description 3
- XXCTXYJTOWUBTF-UHFFFAOYSA-N n-[9-[4-(1-aminocyclobutyl)phenyl]-8-pyridin-2-ylpurin-2-yl]methanesulfonamide Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(NS(=O)(=O)C)=NC=C2N=C1C1=CC=CC=N1 XXCTXYJTOWUBTF-UHFFFAOYSA-N 0.000 claims description 3
- 230000000626 neurodegenerative effect Effects 0.000 claims description 3
- 125000004528 pyrimidin-5-yl group Chemical group N1=CN=CC(=C1)* 0.000 claims description 3
- 125000004943 pyrimidin-6-yl group Chemical group N1=CN=CC=C1* 0.000 claims description 3
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 2
- BICDDBUYBFJAIN-UHFFFAOYSA-N 1-[4-(2-phenyl-1h-pyrrolo[2,3-b]pyridin-3-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C=2C3=CC=CN=C3NC=2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 BICDDBUYBFJAIN-UHFFFAOYSA-N 0.000 claims description 2
- UVFQFDWBEINECI-UHFFFAOYSA-N 1-[4-(2-phenylpyrrolo[2,3-b]pyridin-1-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC=CC=C3C=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 UVFQFDWBEINECI-UHFFFAOYSA-N 0.000 claims description 2
- ATJXXOZVCJJBHR-UHFFFAOYSA-N 1-[4-(3-phenylimidazo[4,5-c]pyridin-2-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C=2N(C3=CN=CC=C3N=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 ATJXXOZVCJJBHR-UHFFFAOYSA-N 0.000 claims description 2
- JQTFCYPWWAUFIE-UHFFFAOYSA-N 1-[4-(5-methyl-6-phenylpyrrolo[2,3-d]pyrimidin-7-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC=NC=C2C(C)=C1C1=CC=CC=C1 JQTFCYPWWAUFIE-UHFFFAOYSA-N 0.000 claims description 2
- RHXWVWGNPGZJIG-UHFFFAOYSA-N 1-[4-(7-phenyl-2-pyrazol-1-ylpurin-8-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C=2N(C3=CN=C(N=C3N=2)N2N=CC=C2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 RHXWVWGNPGZJIG-UHFFFAOYSA-N 0.000 claims description 2
- RVTCIFKZHKVGBN-UHFFFAOYSA-N 1-[4-(8-phenyl-2-piperazin-1-ylpurin-9-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2C=CC=CC=2)N2CCNCC2)C=CC=1C1(N)CCC1 RVTCIFKZHKVGBN-UHFFFAOYSA-N 0.000 claims description 2
- IVDXYNAUMYGGND-UHFFFAOYSA-N 1-[4-[2-(1h-pyrazol-4-yl)-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-7-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(=NC=C3C=C2C=2N=CC=CC=2)C2=CNN=C2)C=CC=1C1(N)CCC1 IVDXYNAUMYGGND-UHFFFAOYSA-N 0.000 claims description 2
- OKOBPBWEZKVXGZ-UHFFFAOYSA-N 1-[4-[2-(1h-pyrazol-4-yl)-8-pyridin-2-ylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2N=CC=CC=2)C2=CNN=C2)C=CC=1C1(N)CCC1 OKOBPBWEZKVXGZ-UHFFFAOYSA-N 0.000 claims description 2
- SDEONDZLJMFSKX-UHFFFAOYSA-N 1-[4-[2-(2-methoxyphenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound COC1=CC=CC=C1C1=NC=C(N=C(C=2N(N=CC=2)C)N2C=3C=CC(=CC=3)C3(N)CCC3)C2=N1 SDEONDZLJMFSKX-UHFFFAOYSA-N 0.000 claims description 2
- MSCGXXDZKSZPMD-UHFFFAOYSA-N 1-[4-[2-(3-methylphenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CC1=CC=CC(C=2N=C3N(C=4C=CC(=CC=4)C4(N)CCC4)C(C=4N(N=CC=4)C)=NC3=CN=2)=C1 MSCGXXDZKSZPMD-UHFFFAOYSA-N 0.000 claims description 2
- ULHBJPFSDDFVLF-UHFFFAOYSA-N 1-[4-[2-(4-fluorophenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CN1N=CC=C1C1=NC2=CN=C(C=3C=CC(F)=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 ULHBJPFSDDFVLF-UHFFFAOYSA-N 0.000 claims description 2
- LLXOPHXGMBOMRC-UHFFFAOYSA-N 1-[4-[2-(4-methoxyphenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C1=CC(OC)=CC=C1C1=NC=C(N=C(C=2N(N=CC=2)C)N2C=3C=CC(=CC=3)C3(N)CCC3)C2=N1 LLXOPHXGMBOMRC-UHFFFAOYSA-N 0.000 claims description 2
- AEXRCQBPELTIBN-UHFFFAOYSA-N 1-[4-[2-phenyl-8-(1h-pyrazol-5-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2NN=CC=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 AEXRCQBPELTIBN-UHFFFAOYSA-N 0.000 claims description 2
- NPCFXYUOUHFANQ-UHFFFAOYSA-N 1-[4-[8-(1-methylbenzimidazol-2-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound N=1C2=CC=CC=C2N(C)C=1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C(C=C1)=CC=C1C1(N)CCC1 NPCFXYUOUHFANQ-UHFFFAOYSA-N 0.000 claims description 2
- TUYFQKONUPDDAU-UHFFFAOYSA-N 1-[4-[8-(1-methylimidazol-2-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CN1C=CN=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 TUYFQKONUPDDAU-UHFFFAOYSA-N 0.000 claims description 2
- ALIMJMCEHHPTBI-UHFFFAOYSA-N 1-[4-[8-(1-methylpyrazol-4-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C1=NN(C)C=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 ALIMJMCEHHPTBI-UHFFFAOYSA-N 0.000 claims description 2
- QRDXHYTWUWWGDX-UHFFFAOYSA-N 1-[4-[8-(1H-imidazol-2-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound N1C(=NC=C1)C=1N(C2=NC(=NC=C2N1)C1=CC=CC=C1)C1=CC=C(C=C1)C1(CCC1)N QRDXHYTWUWWGDX-UHFFFAOYSA-N 0.000 claims description 2
- COTVIDQIKICACY-UHFFFAOYSA-N 1-[4-[8-(1H-imidazol-5-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound N1C=NC(=C1)C=1N(C2=NC(=NC=C2N1)C1=CC=CC=C1)C1=CC=C(C=C1)C1(CCC1)N COTVIDQIKICACY-UHFFFAOYSA-N 0.000 claims description 2
- ZEMWKKLOMCPBEG-UHFFFAOYSA-N 1-[4-[8-(2-ethylpyrazol-3-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CCN1N=CC=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 ZEMWKKLOMCPBEG-UHFFFAOYSA-N 0.000 claims description 2
- NGGCZZZONPAOQO-UHFFFAOYSA-N 1-[4-[8-(3-methylimidazol-4-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CN1C=NC=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 NGGCZZZONPAOQO-UHFFFAOYSA-N 0.000 claims description 2
- IAXNPMSCQGRGRW-UHFFFAOYSA-N 1-[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-4-yl]pyrrolidin-3-ol Chemical compound C=1C=C(N2C3=NC=NC(=C3C=C2C=2N=CC=CC=2)N2CC(O)CC2)C=CC=1C1(N)CCC1 IAXNPMSCQGRGRW-UHFFFAOYSA-N 0.000 claims description 2
- OSJXPSMTSMBXLG-UHFFFAOYSA-N 1-[9-[4-(1-aminocyclobutyl)phenyl]-8-pyridin-2-ylpurin-2-yl]pyrrolidin-3-ol Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2N=CC=CC=2)N2CC(O)CC2)C=CC=1C1(N)CCC1 OSJXPSMTSMBXLG-UHFFFAOYSA-N 0.000 claims description 2
- AUKLVMSTJUBLSC-UHFFFAOYSA-N 2-[4-(1-aminocyclobutyl)phenyl]-1-phenyl-5h-imidazo[4,5-c]pyridin-6-one Chemical compound C=1C=C(C=2N(C3=CC(=O)NC=C3N=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 AUKLVMSTJUBLSC-UHFFFAOYSA-N 0.000 claims description 2
- HRBBDNSJYWCPSL-UHFFFAOYSA-N 2-[9-[4-(1-aminocyclobutyl)phenyl]-2-phenylpurin-8-yl]benzonitrile Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2C(=CC=CC=2)C#N)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 HRBBDNSJYWCPSL-UHFFFAOYSA-N 0.000 claims description 2
- POIMXGBLMFJBHC-UHFFFAOYSA-N 2-[[5-[4-(1-aminocyclobutyl)phenyl]-6-phenylpyrrolo[3,2-d]pyrimidin-2-yl]amino]ethanol Chemical compound C=1C=C(N2C3=CN=C(NCCO)N=C3C=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 POIMXGBLMFJBHC-UHFFFAOYSA-N 0.000 claims description 2
- UQINKPYDSKEWCV-UHFFFAOYSA-N 2-[[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-2-yl]amino]acetic acid Chemical compound C=1C=C(N2C3=NC(NCC(O)=O)=NC=C3C=C2C=2N=CC=CC=2)C=CC=1C1(N)CCC1 UQINKPYDSKEWCV-UHFFFAOYSA-N 0.000 claims description 2
- QKWARHWGCAEWDV-UHFFFAOYSA-N 3-amino-1-methyl-3-[4-[5-methyl-6-phenyl-2-(1h-pyrazol-4-yl)pyrrolo[3,2-d]pyrimidin-7-yl]phenyl]cyclobutan-1-ol Chemical compound C12=NC(C3=CNN=C3)=NC=C2N(C)C(C=2C=CC=CC=2)=C1C(C=C1)=CC=C1C1(N)CC(C)(O)C1 QKWARHWGCAEWDV-UHFFFAOYSA-N 0.000 claims description 2
- ADSOQROUDALIIJ-UHFFFAOYSA-N 4-[9-[4-(1-aminocyclobutyl)phenyl]-8-(2-methylpyrazol-3-yl)purin-2-yl]-n-methylbenzenesulfonamide Chemical compound C1=CC(S(=O)(=O)NC)=CC=C1C1=NC=C(N=C(C=2N(N=CC=2)C)N2C=3C=CC(=CC=3)C3(N)CCC3)C2=N1 ADSOQROUDALIIJ-UHFFFAOYSA-N 0.000 claims description 2
- VMVZYDPSWMYBJY-UHFFFAOYSA-N 5-[4-(1-aminocyclobutyl)phenyl]-6-phenylpyrrolo[3,2-d]pyrimidine-2-carboxylic acid Chemical compound C=1C=C(N2C3=CN=C(N=C3C=C2C=2C=CC=CC=2)C(O)=O)C=CC=1C1(N)CCC1 VMVZYDPSWMYBJY-UHFFFAOYSA-N 0.000 claims description 2
- JURAZGRVQIGAPP-UHFFFAOYSA-N 5-[4-(1-aminocyclobutyl)phenyl]-n,n-dimethyl-6-phenylpyrrolo[3,2-d]pyrimidin-2-amine Chemical compound C=1C2=NC(N(C)C)=NC=C2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=C1 JURAZGRVQIGAPP-UHFFFAOYSA-N 0.000 claims description 2
- DWVGRHAYCWHGLJ-UHFFFAOYSA-N 5-[4-(1-aminocyclobutyl)phenyl]-n-methyl-6-phenylpyrrolo[3,2-d]pyrimidine-2-carboxamide Chemical compound C=1C2=NC(C(=O)NC)=NC=C2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=C1 DWVGRHAYCWHGLJ-UHFFFAOYSA-N 0.000 claims description 2
- ZJGMRUIANFBRQS-UHFFFAOYSA-N 6-[9-[4-(1-aminocyclobutyl)phenyl]-2-phenylpurin-8-yl]-1h-pyridin-2-one Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2NC(=O)C=CC=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 ZJGMRUIANFBRQS-UHFFFAOYSA-N 0.000 claims description 2
- HLLKLEKLBSKZHL-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidine-2-carbonitrile Chemical compound C=1C=C(N2C3=NC(=NC=C3C=C2C=2N=CC=CC=2)C#N)C=CC=1C1(N)CCC1 HLLKLEKLBSKZHL-UHFFFAOYSA-N 0.000 claims description 2
- MXFVLTCFOBULRI-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidine-4-carboxamide Chemical compound C=1C=2C(C(=O)N)=NC=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=N1 MXFVLTCFOBULRI-UHFFFAOYSA-N 0.000 claims description 2
- ZEXGHLXCZAESHQ-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-n-[2-(dimethylamino)ethyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidine-4-carboxamide Chemical compound C=1C=2C(C(=O)NCCN(C)C)=NC=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=N1 ZEXGHLXCZAESHQ-UHFFFAOYSA-N 0.000 claims description 2
- LGSUWLQFIRJRKR-UHFFFAOYSA-N 8-[4-(1-aminocyclobutyl)phenyl]-7-phenyl-3h-purin-2-one Chemical compound C=1C=C(C=2N(C=3C=NC(=O)NC=3N=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 LGSUWLQFIRJRKR-UHFFFAOYSA-N 0.000 claims description 2
- XWCAJKNHFHBMKH-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-2-phenylpurine-8-carbonitrile Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C#N)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 XWCAJKNHFHBMKH-UHFFFAOYSA-N 0.000 claims description 2
- SDVZUNFICIHRBO-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-8-phenylpurin-2-amine Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(N)=NC=C2N=C1C1=CC=CC=C1 SDVZUNFICIHRBO-UHFFFAOYSA-N 0.000 claims description 2
- CWKPJGKIWCIHBU-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-8-pyridin-2-yl-3h-purin-6-one Chemical compound C=1C=C(N2C3=C(C(N=CN3)=O)N=C2C=2N=CC=CC=2)C=CC=1C1(N)CCC1 CWKPJGKIWCIHBU-UHFFFAOYSA-N 0.000 claims description 2
- CVHXDEFYJGZSGQ-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-n,n-dimethyl-8-phenylpurin-2-amine Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(N(C)C)=NC=C2N=C1C1=CC=CC=C1 CVHXDEFYJGZSGQ-UHFFFAOYSA-N 0.000 claims description 2
- AIMMVWOEOZMVMS-UHFFFAOYSA-N cyclopropanecarboxamide Chemical compound NC(=O)C1CC1 AIMMVWOEOZMVMS-UHFFFAOYSA-N 0.000 claims description 2
- ADDRSVZJVHOYKE-UHFFFAOYSA-N n-[1-[4-(2,8-diphenylpurin-9-yl)phenyl]cyclobutyl]acetamide Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2C=CC=CC=2)C=2C=CC=CC=2)C=CC=1C1(NC(=O)C)CCC1 ADDRSVZJVHOYKE-UHFFFAOYSA-N 0.000 claims description 2
- 229910052705 radium Inorganic materials 0.000 claims description 2
- 229910052701 rubidium Inorganic materials 0.000 claims description 2
- KOBCLJHHKOTREH-UHFFFAOYSA-N 1-[4-[8-(5-methyl-1h-imidazol-4-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound N1C=NC(C=2N(C3=NC(=NC=C3N=2)C=2C=CC=CC=2)C=2C=CC(=CC=2)C2(N)CCC2)=C1C KOBCLJHHKOTREH-UHFFFAOYSA-N 0.000 claims 2
- 125000000896 monocarboxylic acid group Chemical group 0.000 claims 2
- DQNIVVLIDAPNDS-UHFFFAOYSA-N 1-[4-[2-(1-methylpyrazol-4-yl)-8-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C1=NN(C)C=C1C1=NC=C(N=C(C=2C=CC=CC=2)N2C=3C=CC(=CC=3)C3(N)CCC3)C2=N1 DQNIVVLIDAPNDS-UHFFFAOYSA-N 0.000 claims 1
- LDJKAZSZDZQYPJ-UHFFFAOYSA-N 1-[4-[4-(1h-pyrazol-4-yl)-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-7-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC=NC(=C3C=C2C=2N=CC=CC=2)C2=CNN=C2)C=CC=1C1(N)CCC1 LDJKAZSZDZQYPJ-UHFFFAOYSA-N 0.000 claims 1
- DEUWMJOPMQFBLI-UHFFFAOYSA-N 1-[4-[8-(3-methylpyridin-2-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CC1=CC=CN=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 DEUWMJOPMQFBLI-UHFFFAOYSA-N 0.000 claims 1
- CIKSCYQRUQIOOG-UHFFFAOYSA-N 5-[4-(1-aminocyclobutyl)phenyl]-n-ethyl-6-phenylpyrrolo[3,2-d]pyrimidin-2-amine Chemical compound C=1C2=NC(NCC)=NC=C2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=C1 CIKSCYQRUQIOOG-UHFFFAOYSA-N 0.000 claims 1
- XQAYKFMZFXCKAP-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-yl-1h-pyrrolo[2,3-d]pyrimidin-4-one Chemical compound C=1C=C(N2C3=C(C(N=CN3)=O)C=C2C=2N=CC=CC=2)C=CC=1C1(N)CCC1 XQAYKFMZFXCKAP-UHFFFAOYSA-N 0.000 claims 1
- NITSAQJPBJYMFL-UHFFFAOYSA-N pyridin-4-ol Chemical compound O=C1C=[C]NC=C1 NITSAQJPBJYMFL-UHFFFAOYSA-N 0.000 claims 1
- OYRRZWATULMEPF-UHFFFAOYSA-N pyrimidin-4-amine Chemical compound NC1=CC=NC=N1 OYRRZWATULMEPF-UHFFFAOYSA-N 0.000 claims 1
- FUXJMHXHGDAHPD-UHFFFAOYSA-N pyrimidine-2-carboxamide Chemical compound NC(=O)C1=NC=CC=N1 FUXJMHXHGDAHPD-UHFFFAOYSA-N 0.000 claims 1
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 364
- 239000000243 solution Substances 0.000 description 154
- 239000000203 mixture Substances 0.000 description 148
- 235000019439 ethyl acetate Nutrition 0.000 description 126
- 239000011541 reaction mixture Substances 0.000 description 117
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical group C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 116
- 239000002904 solvent Substances 0.000 description 110
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 91
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 72
- 229910001868 water Inorganic materials 0.000 description 71
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 70
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 63
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 58
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 56
- 239000002585 base Substances 0.000 description 56
- 239000003054 catalyst Substances 0.000 description 55
- 239000000047 product Substances 0.000 description 55
- 239000012043 crude product Substances 0.000 description 50
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 44
- 239000012044 organic layer Substances 0.000 description 44
- PAYRUJLWNCNPSJ-UHFFFAOYSA-N Aniline Chemical compound NC1=CC=CC=C1 PAYRUJLWNCNPSJ-UHFFFAOYSA-N 0.000 description 42
- 229910000024 caesium carbonate Inorganic materials 0.000 description 42
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 41
- 150000001345 alkine derivatives Chemical group 0.000 description 39
- 238000006243 chemical reaction Methods 0.000 description 39
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 37
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 36
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 36
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 36
- 230000002829 reductive effect Effects 0.000 description 36
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 35
- 229910052938 sodium sulfate Inorganic materials 0.000 description 35
- 235000011152 sodium sulphate Nutrition 0.000 description 35
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 32
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 30
- 238000001914 filtration Methods 0.000 description 30
- 206010028980 Neoplasm Diseases 0.000 description 28
- 239000003153 chemical reaction reagent Substances 0.000 description 28
- 238000010438 heat treatment Methods 0.000 description 27
- OKKJLVBELUTLKV-VMNATFBRSA-N methanol-d1 Chemical compound [2H]OC OKKJLVBELUTLKV-VMNATFBRSA-N 0.000 description 27
- 239000000654 additive Substances 0.000 description 26
- FJDQFPXHSGXQBY-UHFFFAOYSA-L caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 25
- YNHIGQDRGKUECZ-UHFFFAOYSA-L bis(triphenylphosphine)palladium(ii) dichloride Chemical compound [Cl-].[Cl-].[Pd+2].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 YNHIGQDRGKUECZ-UHFFFAOYSA-L 0.000 description 24
- 238000009472 formulation Methods 0.000 description 24
- 125000000623 heterocyclic group Chemical group 0.000 description 24
- 239000000706 filtrate Substances 0.000 description 23
- 238000000746 purification Methods 0.000 description 23
- 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 22
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 22
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 22
- 238000002953 preparative HPLC Methods 0.000 description 22
- UGOMMVLRQDMAQQ-UHFFFAOYSA-N xphos Chemical group CC(C)C1=CC(C(C)C)=CC(C(C)C)=C1C1=CC=CC=C1P(C1CCCCC1)C1CCCCC1 UGOMMVLRQDMAQQ-UHFFFAOYSA-N 0.000 description 22
- 239000012267 brine Substances 0.000 description 21
- 239000003446 ligand Substances 0.000 description 21
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 21
- 238000005160 1H NMR spectroscopy Methods 0.000 description 20
- 201000011510 cancer Diseases 0.000 description 19
- OPQARKPSCNTWTJ-UHFFFAOYSA-L copper(ii) acetate Chemical compound [Cu+2].CC([O-])=O.CC([O-])=O OPQARKPSCNTWTJ-UHFFFAOYSA-L 0.000 description 19
- 230000008878 coupling Effects 0.000 description 19
- 238000010168 coupling process Methods 0.000 description 19
- 238000005859 coupling reaction Methods 0.000 description 19
- NROKBHXJSPEDAR-UHFFFAOYSA-M potassium fluoride Chemical compound [F-].[K+] NROKBHXJSPEDAR-UHFFFAOYSA-M 0.000 description 18
- 239000000741 silica gel Substances 0.000 description 18
- 229910002027 silica gel Inorganic materials 0.000 description 18
- 239000004480 active ingredient Substances 0.000 description 17
- 229940079593 drug Drugs 0.000 description 17
- BWHDROKFUHTORW-UHFFFAOYSA-N tritert-butylphosphane Chemical compound CC(C)(C)P(C(C)(C)C)C(C)(C)C BWHDROKFUHTORW-UHFFFAOYSA-N 0.000 description 17
- CXNIUSPIQKWYAI-UHFFFAOYSA-N xantphos Chemical compound C=12OC3=C(P(C=4C=CC=CC=4)C=4C=CC=CC=4)C=CC=C3C(C)(C)C2=CC=CC=1P(C=1C=CC=CC=1)C1=CC=CC=C1 CXNIUSPIQKWYAI-UHFFFAOYSA-N 0.000 description 17
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 16
- 230000000996 additive effect Effects 0.000 description 16
- 238000004440 column chromatography Methods 0.000 description 16
- 239000000463 material Substances 0.000 description 16
- 150000003335 secondary amines Chemical class 0.000 description 16
- 238000000926 separation method Methods 0.000 description 16
- NPXOKRUENSOPAO-UHFFFAOYSA-N Raney nickel Chemical compound [Al].[Ni] NPXOKRUENSOPAO-UHFFFAOYSA-N 0.000 description 15
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 15
- 229960000583 acetic acid Drugs 0.000 description 15
- 125000000217 alkyl group Chemical group 0.000 description 15
- 230000015572 biosynthetic process Effects 0.000 description 15
- 239000003208 petroleum Substances 0.000 description 15
- 239000000725 suspension Substances 0.000 description 15
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical group [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 14
- 229920006395 saturated elastomer Polymers 0.000 description 14
- 229910019142 PO4 Inorganic materials 0.000 description 13
- 239000002253 acid Substances 0.000 description 13
- 239000012298 atmosphere Substances 0.000 description 13
- 210000004027 cell Anatomy 0.000 description 13
- 238000006880 cross-coupling reaction Methods 0.000 description 13
- 238000003818 flash chromatography Methods 0.000 description 13
- 239000003112 inhibitor Substances 0.000 description 13
- 235000021317 phosphate Nutrition 0.000 description 13
- 239000011698 potassium fluoride Substances 0.000 description 13
- 125000006239 protecting group Chemical group 0.000 description 13
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N triphenylphosphine Substances C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 13
- 229910021595 Copper(I) iodide Inorganic materials 0.000 description 12
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 12
- UEXCJVNBTNXOEH-UHFFFAOYSA-N Ethynylbenzene Chemical compound C#CC1=CC=CC=C1 UEXCJVNBTNXOEH-UHFFFAOYSA-N 0.000 description 12
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 12
- HUMNYLRZRPPJDN-UHFFFAOYSA-N benzaldehyde Chemical compound O=CC1=CC=CC=C1 HUMNYLRZRPPJDN-UHFFFAOYSA-N 0.000 description 12
- LSXDOTMGLUJQCM-UHFFFAOYSA-M copper(i) iodide Chemical compound I[Cu] LSXDOTMGLUJQCM-UHFFFAOYSA-M 0.000 description 12
- 239000001257 hydrogen Substances 0.000 description 12
- 150000007529 inorganic bases Chemical class 0.000 description 12
- 239000011734 sodium Substances 0.000 description 12
- 239000007787 solid Substances 0.000 description 12
- 230000009466 transformation Effects 0.000 description 12
- 238000000844 transformation Methods 0.000 description 12
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 11
- 238000005481 NMR spectroscopy Methods 0.000 description 11
- 102000001253 Protein Kinase Human genes 0.000 description 11
- 238000004587 chromatography analysis Methods 0.000 description 11
- 239000012299 nitrogen atmosphere Substances 0.000 description 11
- 239000012038 nucleophile Substances 0.000 description 11
- 150000007530 organic bases Chemical class 0.000 description 11
- 239000000546 pharmaceutical excipient Substances 0.000 description 11
- 238000003786 synthesis reaction Methods 0.000 description 11
- SECXISVLQFMRJM-UHFFFAOYSA-N N-Methylpyrrolidone Chemical compound CN1CCCC1=O SECXISVLQFMRJM-UHFFFAOYSA-N 0.000 description 10
- 150000001336 alkenes Chemical class 0.000 description 10
- 125000005621 boronate group Chemical class 0.000 description 10
- 125000005620 boronic acid group Chemical class 0.000 description 10
- 125000000524 functional group Chemical group 0.000 description 10
- 150000002901 organomagnesium compounds Chemical class 0.000 description 10
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 10
- 108060006633 protein kinase Proteins 0.000 description 10
- 238000007363 ring formation reaction Methods 0.000 description 10
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 10
- 230000001225 therapeutic effect Effects 0.000 description 10
- 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 9
- SCYULBFZEHDVBN-UHFFFAOYSA-N 1,1-Dichloroethane Chemical compound CC(Cl)Cl SCYULBFZEHDVBN-UHFFFAOYSA-N 0.000 description 9
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical class OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 9
- XTHFKEDIFFGKHM-UHFFFAOYSA-N Dimethoxyethane Chemical compound COCCOC XTHFKEDIFFGKHM-UHFFFAOYSA-N 0.000 description 9
- 239000007864 aqueous solution Substances 0.000 description 9
- RBFQJDQYXXHULB-UHFFFAOYSA-N arsane Chemical class [AsH3] RBFQJDQYXXHULB-UHFFFAOYSA-N 0.000 description 9
- 238000003556 assay Methods 0.000 description 9
- 150000004649 carbonic acid derivatives Chemical class 0.000 description 9
- 229910052802 copper Inorganic materials 0.000 description 9
- 239000010949 copper Substances 0.000 description 9
- SBZXBUIDTXKZTM-UHFFFAOYSA-N diglyme Chemical compound COCCOCCOC SBZXBUIDTXKZTM-UHFFFAOYSA-N 0.000 description 9
- 150000002148 esters Chemical class 0.000 description 9
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 9
- 150000002222 fluorine compounds Chemical class 0.000 description 9
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 9
- 239000012074 organic phase Substances 0.000 description 9
- 150000002941 palladium compounds Chemical class 0.000 description 9
- 150000003003 phosphines Chemical class 0.000 description 9
- 235000003270 potassium fluoride Nutrition 0.000 description 9
- 229910052709 silver Inorganic materials 0.000 description 9
- 239000004332 silver Substances 0.000 description 9
- 150000003512 tertiary amines Chemical class 0.000 description 9
- 229940124597 therapeutic agent Drugs 0.000 description 9
- 238000004809 thin layer chromatography Methods 0.000 description 9
- 229910052723 transition metal Inorganic materials 0.000 description 9
- 150000003624 transition metals Chemical class 0.000 description 9
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 8
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 8
- KZBUYRJDOAKODT-UHFFFAOYSA-N Chlorine Chemical compound ClCl KZBUYRJDOAKODT-UHFFFAOYSA-N 0.000 description 8
- 208000009956 adenocarcinoma Diseases 0.000 description 8
- 239000003085 diluting agent Substances 0.000 description 8
- 229910052736 halogen Inorganic materials 0.000 description 8
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 8
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 8
- VBEGHXKAFSLLGE-UHFFFAOYSA-N n-phenylnitramide Chemical class [O-][N+](=O)NC1=CC=CC=C1 VBEGHXKAFSLLGE-UHFFFAOYSA-N 0.000 description 8
- 238000002360 preparation method Methods 0.000 description 8
- 239000007858 starting material Substances 0.000 description 8
- 101001059454 Homo sapiens Serine/threonine-protein kinase MARK2 Proteins 0.000 description 7
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 7
- 206010025323 Lymphomas Diseases 0.000 description 7
- 206010039491 Sarcoma Diseases 0.000 description 7
- 102100028904 Serine/threonine-protein kinase MARK2 Human genes 0.000 description 7
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 7
- 150000001412 amines Chemical class 0.000 description 7
- 239000002246 antineoplastic agent Substances 0.000 description 7
- 125000004432 carbon atom Chemical group C* 0.000 description 7
- 239000000460 chlorine Substances 0.000 description 7
- 150000002431 hydrogen Chemical class 0.000 description 7
- 229910052751 metal Inorganic materials 0.000 description 7
- 239000002184 metal Substances 0.000 description 7
- 239000000843 powder Substances 0.000 description 7
- 239000003755 preservative agent Substances 0.000 description 7
- 150000003254 radicals Chemical group 0.000 description 7
- 238000012552 review Methods 0.000 description 7
- 239000000758 substrate Substances 0.000 description 7
- QGMGHALXLXKCBD-UHFFFAOYSA-N 4-amino-n-(2-aminophenyl)benzamide Chemical compound C1=CC(N)=CC=C1C(=O)NC1=CC=CC=C1N QGMGHALXLXKCBD-UHFFFAOYSA-N 0.000 description 6
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 6
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 6
- KAESVJOAVNADME-UHFFFAOYSA-N Pyrrole Chemical compound C=1C=CNC=1 KAESVJOAVNADME-UHFFFAOYSA-N 0.000 description 6
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical group C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 description 6
- 230000002378 acidificating effect Effects 0.000 description 6
- 150000001298 alcohols Chemical class 0.000 description 6
- 150000001299 aldehydes Chemical class 0.000 description 6
- 239000003963 antioxidant agent Substances 0.000 description 6
- 235000006708 antioxidants Nutrition 0.000 description 6
- 239000000969 carrier Substances 0.000 description 6
- 125000000753 cycloalkyl group Chemical group 0.000 description 6
- 229940127089 cytotoxic agent Drugs 0.000 description 6
- 239000000796 flavoring agent Substances 0.000 description 6
- 150000002367 halogens Chemical class 0.000 description 6
- 229910052763 palladium Inorganic materials 0.000 description 6
- QNGNSVIICDLXHT-UHFFFAOYSA-N para-ethylbenzaldehyde Natural products CCC1=CC=C(C=O)C=C1 QNGNSVIICDLXHT-UHFFFAOYSA-N 0.000 description 6
- HXITXNWTGFUOAU-UHFFFAOYSA-N phenylboronic acid Chemical compound OB(O)C1=CC=CC=C1 HXITXNWTGFUOAU-UHFFFAOYSA-N 0.000 description 6
- 230000026731 phosphorylation Effects 0.000 description 6
- 238000006366 phosphorylation reaction Methods 0.000 description 6
- 230000009467 reduction Effects 0.000 description 6
- 238000006722 reduction reaction Methods 0.000 description 6
- 238000010992 reflux Methods 0.000 description 6
- FVAUCKIRQBBSSJ-UHFFFAOYSA-M sodium iodide Chemical compound [Na+].[I-] FVAUCKIRQBBSSJ-UHFFFAOYSA-M 0.000 description 6
- 239000000375 suspending agent Substances 0.000 description 6
- 239000003765 sweetening agent Substances 0.000 description 6
- 150000003573 thiols Chemical class 0.000 description 6
- 102100037263 3-phosphoinositide-dependent protein kinase 1 Human genes 0.000 description 5
- HCTISZQLTGAYOX-UHFFFAOYSA-N 4,6-dichloro-5-nitropyrimidine Chemical compound [O-][N+](=O)C1=C(Cl)N=CN=C1Cl HCTISZQLTGAYOX-UHFFFAOYSA-N 0.000 description 5
- 101100322915 Caenorhabditis elegans akt-1 gene Proteins 0.000 description 5
- 201000009030 Carcinoma Diseases 0.000 description 5
- ROSDSFDQCJNGOL-UHFFFAOYSA-N Dimethylamine Chemical compound CNC ROSDSFDQCJNGOL-UHFFFAOYSA-N 0.000 description 5
- 108030004793 Dual-specificity kinases Proteins 0.000 description 5
- IAYPIBMASNFSPL-UHFFFAOYSA-N Ethylene oxide Chemical compound C1CO1 IAYPIBMASNFSPL-UHFFFAOYSA-N 0.000 description 5
- 101000600756 Homo sapiens 3-phosphoinositide-dependent protein kinase 1 Proteins 0.000 description 5
- 101001117146 Homo sapiens [Pyruvate dehydrogenase (acetyl-transferring)] kinase isozyme 1, mitochondrial Proteins 0.000 description 5
- 239000007868 Raney catalyst Substances 0.000 description 5
- 229910000564 Raney nickel Inorganic materials 0.000 description 5
- 150000007513 acids Chemical class 0.000 description 5
- 229940024606 amino acid Drugs 0.000 description 5
- 235000001014 amino acid Nutrition 0.000 description 5
- 125000004429 atom Chemical group 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 239000007859 condensation product Substances 0.000 description 5
- 235000014113 dietary fatty acids Nutrition 0.000 description 5
- 229930195729 fatty acid Natural products 0.000 description 5
- 239000000194 fatty acid Substances 0.000 description 5
- 150000004665 fatty acids Chemical class 0.000 description 5
- 150000003840 hydrochlorides Chemical class 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000005764 inhibitory process Effects 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 239000007788 liquid Substances 0.000 description 5
- 210000004072 lung Anatomy 0.000 description 5
- 239000011259 mixed solution Substances 0.000 description 5
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 5
- RRHNGIRRWDWWQQ-UHFFFAOYSA-N n-iodoaniline Chemical compound INC1=CC=CC=C1 RRHNGIRRWDWWQQ-UHFFFAOYSA-N 0.000 description 5
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 description 5
- 231100000252 nontoxic Toxicity 0.000 description 5
- 230000003000 nontoxic effect Effects 0.000 description 5
- 239000007800 oxidant agent Substances 0.000 description 5
- 230000001590 oxidative effect Effects 0.000 description 5
- 230000036961 partial effect Effects 0.000 description 5
- 239000012071 phase Substances 0.000 description 5
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Substances [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 5
- 150000003141 primary amines Chemical class 0.000 description 5
- 125000006413 ring segment Chemical group 0.000 description 5
- 206010041823 squamous cell carcinoma Diseases 0.000 description 5
- 238000003756 stirring Methods 0.000 description 5
- 239000000126 substance Substances 0.000 description 5
- 239000000080 wetting agent Substances 0.000 description 5
- LBLYYCQCTBFVLH-UHFFFAOYSA-N 2-Methylbenzenesulfonic acid Chemical compound CC1=CC=CC=C1S(O)(=O)=O LBLYYCQCTBFVLH-UHFFFAOYSA-N 0.000 description 4
- GPFGRYFMMHDHGA-UHFFFAOYSA-N 4-chloro-3-iodopyridine Chemical compound ClC1=CC=NC=C1I GPFGRYFMMHDHGA-UHFFFAOYSA-N 0.000 description 4
- GGXIOSCUHASSOL-UHFFFAOYSA-N 6-chloro-3-iodopyridin-2-amine Chemical class NC1=NC(Cl)=CC=C1I GGXIOSCUHASSOL-UHFFFAOYSA-N 0.000 description 4
- 101150107888 AKT2 gene Proteins 0.000 description 4
- 101150051155 Akt3 gene Proteins 0.000 description 4
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 4
- OKKJLVBELUTLKV-MZCSYVLQSA-N Deuterated methanol Chemical compound [2H]OC([2H])([2H])[2H] OKKJLVBELUTLKV-MZCSYVLQSA-N 0.000 description 4
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 4
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 4
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 4
- YLQBMQCUIZJEEH-UHFFFAOYSA-N Furan Chemical group C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- 206010024612 Lipoma Diseases 0.000 description 4
- MZRVEZGGRBJDDB-UHFFFAOYSA-N N-Butyllithium Chemical compound [Li]CCCC MZRVEZGGRBJDDB-UHFFFAOYSA-N 0.000 description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical class OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 4
- GLUUGHFHXGJENI-UHFFFAOYSA-N Piperazine Chemical group C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 description 4
- 239000004698 Polyethylene Substances 0.000 description 4
- 102000001708 Protein Isoforms Human genes 0.000 description 4
- 108010029485 Protein Isoforms Proteins 0.000 description 4
- 102000009516 Protein Serine-Threonine Kinases Human genes 0.000 description 4
- 108010009341 Protein Serine-Threonine Kinases Proteins 0.000 description 4
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 4
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 4
- 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 4
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- WQDUMFSSJAZKTM-UHFFFAOYSA-N Sodium methoxide Chemical compound [Na+].[O-]C WQDUMFSSJAZKTM-UHFFFAOYSA-N 0.000 description 4
- YTPLMLYBLZKORZ-UHFFFAOYSA-N Thiophene Chemical group C=1C=CSC=1 YTPLMLYBLZKORZ-UHFFFAOYSA-N 0.000 description 4
- 125000004423 acyloxy group Chemical group 0.000 description 4
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 235000010323 ascorbic acid Nutrition 0.000 description 4
- 229960005070 ascorbic acid Drugs 0.000 description 4
- 239000011668 ascorbic acid Substances 0.000 description 4
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 4
- 230000037396 body weight Effects 0.000 description 4
- 239000000872 buffer Substances 0.000 description 4
- 150000001732 carboxylic acid derivatives Chemical group 0.000 description 4
- 239000003086 colorant Substances 0.000 description 4
- 125000003963 dichloro group Chemical group Cl* 0.000 description 4
- 239000002270 dispersing agent Substances 0.000 description 4
- 239000002552 dosage form Substances 0.000 description 4
- 238000000605 extraction Methods 0.000 description 4
- 229960002949 fluorouracil Drugs 0.000 description 4
- 238000011010 flushing procedure Methods 0.000 description 4
- 235000003599 food sweetener Nutrition 0.000 description 4
- 238000007306 functionalization reaction Methods 0.000 description 4
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 4
- 239000000543 intermediate Substances 0.000 description 4
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 4
- HPJKCIUCZWXJDR-UHFFFAOYSA-N letrozole Chemical compound C1=CC(C#N)=CC=C1C(N1N=CN=C1)C1=CC=C(C#N)C=C1 HPJKCIUCZWXJDR-UHFFFAOYSA-N 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 150000002632 lipids Chemical class 0.000 description 4
- 150000002739 metals Chemical class 0.000 description 4
- 239000003094 microcapsule Substances 0.000 description 4
- 238000002156 mixing Methods 0.000 description 4
- 239000003921 oil Substances 0.000 description 4
- 235000019198 oils Nutrition 0.000 description 4
- 230000003287 optical effect Effects 0.000 description 4
- 125000002524 organometallic group Chemical group 0.000 description 4
- 239000001301 oxygen Substances 0.000 description 4
- HKOOXMFOFWEVGF-UHFFFAOYSA-N phenylhydrazine Chemical compound NNC1=CC=CC=C1 HKOOXMFOFWEVGF-UHFFFAOYSA-N 0.000 description 4
- 229920001223 polyethylene glycol Polymers 0.000 description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 4
- 108090000765 processed proteins & peptides Chemical group 0.000 description 4
- 238000011321 prophylaxis Methods 0.000 description 4
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 4
- 210000002307 prostate Anatomy 0.000 description 4
- 239000011535 reaction buffer Substances 0.000 description 4
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 4
- 230000019491 signal transduction Effects 0.000 description 4
- VWDWKYIASSYTQR-UHFFFAOYSA-N sodium nitrate Chemical compound [Na+].[O-][N+]([O-])=O VWDWKYIASSYTQR-UHFFFAOYSA-N 0.000 description 4
- 208000024891 symptom Diseases 0.000 description 4
- 239000003826 tablet Substances 0.000 description 4
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 3
- 125000003088 (fluoren-9-ylmethoxy)carbonyl group Chemical group 0.000 description 3
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 3
- JBMBVWROWJGFMG-UHFFFAOYSA-N 2-chloro-7h-purine Chemical compound ClC1=NC=C2NC=NC2=N1 JBMBVWROWJGFMG-UHFFFAOYSA-N 0.000 description 3
- HJOQGBBHVRYTDX-UHFFFAOYSA-N 2-chloro-7h-pyrrolo[2,3-d]pyrimidine Chemical class ClC1=NC=C2C=CNC2=N1 HJOQGBBHVRYTDX-UHFFFAOYSA-N 0.000 description 3
- UWUBPQVLQOHMKF-UHFFFAOYSA-N 5-chloro-1h-imidazo[4,5-b]pyridine Chemical compound ClC1=CC=C2NC=NC2=N1 UWUBPQVLQOHMKF-UHFFFAOYSA-N 0.000 description 3
- UHOVQNZJYSORNB-UHFFFAOYSA-N Benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- UGFAIRIUMAVXCW-UHFFFAOYSA-N Carbon monoxide Chemical compound [O+]#[C-] UGFAIRIUMAVXCW-UHFFFAOYSA-N 0.000 description 3
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 3
- QMMFVYPAHWMCMS-UHFFFAOYSA-N Dimethyl sulfide Chemical compound CSC QMMFVYPAHWMCMS-UHFFFAOYSA-N 0.000 description 3
- 201000008808 Fibrosarcoma Diseases 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- SIKJAQJRHWYJAI-UHFFFAOYSA-N Indole Chemical group C1=CC=C2NC=CC2=C1 SIKJAQJRHWYJAI-UHFFFAOYSA-N 0.000 description 3
- 102100020944 Integrin-linked protein kinase Human genes 0.000 description 3
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 3
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 3
- 208000008770 Multiple Hamartoma Syndrome Diseases 0.000 description 3
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 3
- 102000038030 PI3Ks Human genes 0.000 description 3
- 108091007960 PI3Ks Proteins 0.000 description 3
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 3
- 201000004681 Psoriasis Diseases 0.000 description 3
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical class [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 3
- CZMRCDWAGMRECN-UGDNZRGBSA-N 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 3
- 229930006000 Sucrose Natural products 0.000 description 3
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 3
- 206010043276 Teratoma Diseases 0.000 description 3
- FOCVUCIESVLUNU-UHFFFAOYSA-N Thiotepa Chemical compound C1CN1P(N1CC1)(=S)N1CC1 FOCVUCIESVLUNU-UHFFFAOYSA-N 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 3
- 150000001342 alkaline earth metals Chemical class 0.000 description 3
- 125000003545 alkoxy group Chemical group 0.000 description 3
- 150000001408 amides Chemical class 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 125000003277 amino group Chemical group 0.000 description 3
- 230000006907 apoptotic process Effects 0.000 description 3
- 239000007900 aqueous suspension Substances 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 3
- 229960000397 bevacizumab Drugs 0.000 description 3
- MXQOYLRVSVOCQT-UHFFFAOYSA-N bis(tri-t-butylphosphine)palladium (0) Substances [Pd].CC(C)(C)P(C(C)(C)C)C(C)(C)C.CC(C)(C)P(C(C)(C)C)C(C)(C)C MXQOYLRVSVOCQT-UHFFFAOYSA-N 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 210000004556 brain Anatomy 0.000 description 3
- HXCHCVDVKSCDHU-LULTVBGHSA-N calicheamicin Chemical compound C1[C@H](OC)[C@@H](NCC)CO[C@H]1O[C@H]1[C@H](O[C@@H]2C\3=C(NC(=O)OC)C(=O)C[C@](C/3=C/CSSSC)(O)C#C\C=C/C#C2)O[C@H](C)[C@@H](NO[C@@H]2O[C@H](C)[C@@H](SC(=O)C=3C(=C(OC)C(O[C@H]4[C@@H]([C@H](OC)[C@@H](O)[C@H](C)O4)O)=C(I)C=3C)OC)[C@@H](O)C2)[C@@H]1O HXCHCVDVKSCDHU-LULTVBGHSA-N 0.000 description 3
- 229930195731 calicheamicin Natural products 0.000 description 3
- 229910002091 carbon monoxide Inorganic materials 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 150000001805 chlorine compounds Chemical class 0.000 description 3
- 125000001309 chloro group Chemical group Cl* 0.000 description 3
- KWTSZCJMWHGPOS-UHFFFAOYSA-M chloro(trimethyl)stannane Chemical compound C[Sn](C)(C)Cl KWTSZCJMWHGPOS-UHFFFAOYSA-M 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 210000001072 colon Anatomy 0.000 description 3
- 238000002648 combination therapy Methods 0.000 description 3
- 238000001816 cooling Methods 0.000 description 3
- 229920001577 copolymer Polymers 0.000 description 3
- 239000006071 cream Substances 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 229960004679 doxorubicin Drugs 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 206010016629 fibroma Diseases 0.000 description 3
- 235000013355 food flavoring agent Nutrition 0.000 description 3
- 238000001640 fractional crystallisation Methods 0.000 description 3
- 239000000499 gel Substances 0.000 description 3
- 229960003297 gemtuzumab ozogamicin Drugs 0.000 description 3
- 239000008187 granular material Substances 0.000 description 3
- 201000011066 hemangioma Diseases 0.000 description 3
- 108010059517 integrin-linked kinase Proteins 0.000 description 3
- GURKHSYORGJETM-WAQYZQTGSA-N irinotecan hydrochloride (anhydrous) Chemical compound Cl.C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 GURKHSYORGJETM-WAQYZQTGSA-N 0.000 description 3
- 125000000842 isoxazolyl group Chemical group 0.000 description 3
- BCFGMOOMADDAQU-UHFFFAOYSA-N lapatinib Chemical compound O1C(CNCCS(=O)(=O)C)=CC=C1C1=CC=C(N=CN=C2NC=3C=C(Cl)C(OCC=4C=C(F)C=CC=4)=CC=3)C2=C1 BCFGMOOMADDAQU-UHFFFAOYSA-N 0.000 description 3
- 239000002502 liposome Substances 0.000 description 3
- 229940057995 liquid paraffin Drugs 0.000 description 3
- 150000002690 malonic acid derivatives Chemical class 0.000 description 3
- 201000001441 melanoma Diseases 0.000 description 3
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 3
- 230000002503 metabolic effect Effects 0.000 description 3
- 229960000485 methotrexate Drugs 0.000 description 3
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- BAVYZALUXZFZLV-UHFFFAOYSA-N mono-methylamine Natural products NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 3
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 3
- 238000000655 nuclear magnetic resonance spectrum Methods 0.000 description 3
- 239000004006 olive oil Substances 0.000 description 3
- 235000008390 olive oil Nutrition 0.000 description 3
- 150000007524 organic acids Chemical class 0.000 description 3
- 125000002971 oxazolyl group Chemical group 0.000 description 3
- MUJIDPITZJWBSW-UHFFFAOYSA-N palladium(2+) Chemical compound [Pd+2] MUJIDPITZJWBSW-UHFFFAOYSA-N 0.000 description 3
- YJVFFLUZDVXJQI-UHFFFAOYSA-L palladium(ii) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 229960002087 pertuzumab Drugs 0.000 description 3
- 239000010452 phosphate Substances 0.000 description 3
- 229920003023 plastic Polymers 0.000 description 3
- 239000004033 plastic Substances 0.000 description 3
- 235000011056 potassium acetate Nutrition 0.000 description 3
- 238000012746 preparative thin layer chromatography Methods 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000000069 prophylactic effect Effects 0.000 description 3
- 239000004405 propyl p-hydroxybenzoate Substances 0.000 description 3
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 3
- 108090000623 proteins and genes Proteins 0.000 description 3
- 150000003212 purines Chemical class 0.000 description 3
- 125000000714 pyrimidinyl group Chemical group 0.000 description 3
- 150000004943 pyrrolo[2,3-d]pyrimidines Chemical class 0.000 description 3
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical class OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 description 3
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 3
- 239000003381 stabilizer Substances 0.000 description 3
- 239000011550 stock solution Substances 0.000 description 3
- 239000005720 sucrose Substances 0.000 description 3
- 239000011593 sulfur Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 230000002195 synergetic effect Effects 0.000 description 3
- KTCWXIDJSFQZCV-UHFFFAOYSA-N tert-butyl n-(2-chloro-3-iodopyridin-4-yl)carbamate Chemical compound CC(C)(C)OC(=O)NC1=CC=NC(Cl)=C1I KTCWXIDJSFQZCV-UHFFFAOYSA-N 0.000 description 3
- ZHLMLNURZUMCAM-UHFFFAOYSA-N tert-butyl n-[1-[4-(2-chloro-8-phenylpurin-9-yl)phenyl]cyclobutyl]carbamate Chemical compound C=1C=C(N2C3=NC(Cl)=NC=C3N=C2C=2C=CC=CC=2)C=CC=1C1(NC(=O)OC(C)(C)C)CCC1 ZHLMLNURZUMCAM-UHFFFAOYSA-N 0.000 description 3
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 3
- 125000004044 trifluoroacetyl group Chemical group FC(C(=O)*)(F)F 0.000 description 3
- HEPHBMPRKIHMSY-UHFFFAOYSA-N trimethyl-[2-[(2-methyl-4-trimethylstannylimidazol-1-yl)methoxy]ethyl]silane Chemical compound CC1=NC([Sn](C)(C)C)=CN1COCC[Si](C)(C)C HEPHBMPRKIHMSY-UHFFFAOYSA-N 0.000 description 3
- PUPZLCDOIYMWBV-UHFFFAOYSA-N (+/-)-1,3-Butanediol Chemical compound CC(O)CCO PUPZLCDOIYMWBV-UHFFFAOYSA-N 0.000 description 2
- KWGRBVOPPLSCSI-WPRPVWTQSA-N (-)-ephedrine Chemical compound CN[C@@H](C)[C@H](O)C1=CC=CC=C1 KWGRBVOPPLSCSI-WPRPVWTQSA-N 0.000 description 2
- QBYIENPQHBMVBV-HFEGYEGKSA-N (2R)-2-hydroxy-2-phenylacetic acid Chemical compound O[C@@H](C(O)=O)c1ccccc1.O[C@@H](C(O)=O)c1ccccc1 QBYIENPQHBMVBV-HFEGYEGKSA-N 0.000 description 2
- XMBSMMCPKFDGEO-ZETCQYMHSA-N (2s)-2-amino-5-[[amino-(2-methoxyethylamino)methylidene]amino]pentanoic acid Chemical compound COCCNC(=N)NCCC[C@H](N)C(O)=O XMBSMMCPKFDGEO-ZETCQYMHSA-N 0.000 description 2
- 125000006582 (C5-C6) heterocycloalkyl group Chemical group 0.000 description 2
- ZORQXIQZAOLNGE-UHFFFAOYSA-N 1,1-difluorocyclohexane Chemical compound FC1(F)CCCCC1 ZORQXIQZAOLNGE-UHFFFAOYSA-N 0.000 description 2
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 2
- 125000001462 1-pyrrolyl group Chemical group [*]N1C([H])=C([H])C([H])=C1[H] 0.000 description 2
- YBYIRNPNPLQARY-UHFFFAOYSA-N 1H-indene Chemical compound C1=CC=C2CC=CC2=C1 YBYIRNPNPLQARY-UHFFFAOYSA-N 0.000 description 2
- MVXVYAKCVDQRLW-UHFFFAOYSA-N 1h-pyrrolo[2,3-b]pyridine Chemical class C1=CN=C2NC=CC2=C1 MVXVYAKCVDQRLW-UHFFFAOYSA-N 0.000 description 2
- MLCNOCRGSBCAGH-UHFFFAOYSA-N 2,3-dichloropyrazine Chemical class ClC1=NC=CN=C1Cl MLCNOCRGSBCAGH-UHFFFAOYSA-N 0.000 description 2
- QGFWFWSBFMRDNP-UHFFFAOYSA-N 2,4,6-trichloro-5-nitropyrimidine Chemical compound [O-][N+](=O)C1=C(Cl)N=C(Cl)N=C1Cl QGFWFWSBFMRDNP-UHFFFAOYSA-N 0.000 description 2
- RGJNPJRAXMSHKN-UHFFFAOYSA-N 2,4-dichloro-5-iodopyrimidine Chemical compound ClC1=NC=C(I)C(Cl)=N1 RGJNPJRAXMSHKN-UHFFFAOYSA-N 0.000 description 2
- SHCWQWRTKPNTEM-UHFFFAOYSA-N 2,6-dichloro-3-nitropyridine Chemical compound [O-][N+](=O)C1=CC=C(Cl)N=C1Cl SHCWQWRTKPNTEM-UHFFFAOYSA-N 0.000 description 2
- UUXXKGYFVQVDDL-UHFFFAOYSA-N 2-bromo-3-chloro-6-methoxypyridine Chemical compound COC1=CC=C(Cl)C(Br)=N1 UUXXKGYFVQVDDL-UHFFFAOYSA-N 0.000 description 2
- YGYICBWHAAGGOB-UHFFFAOYSA-N 2-chloro-3-(2-phenylethynyl)pyridine Chemical compound ClC1=NC=CC=C1C#CC1=CC=CC=C1 YGYICBWHAAGGOB-UHFFFAOYSA-N 0.000 description 2
- BYVMKCHHWASQFN-UHFFFAOYSA-N 2-chloro-3-iodopyridin-4-amine Chemical class NC1=CC=NC(Cl)=C1I BYVMKCHHWASQFN-UHFFFAOYSA-N 0.000 description 2
- OHWSWGXNZDSHLM-UHFFFAOYSA-N 2-chloro-3-iodopyridine Chemical class ClC1=NC=CC=C1I OHWSWGXNZDSHLM-UHFFFAOYSA-N 0.000 description 2
- WFJAKBGXNXBMLL-UHFFFAOYSA-N 2-chloro-3-nitropyrazine Chemical class [O-][N+](=O)C1=NC=CN=C1Cl WFJAKBGXNXBMLL-UHFFFAOYSA-N 0.000 description 2
- XNMWTPWQILTFRI-UHFFFAOYSA-N 2-chloro-N-[[4-[2-(N-cyano-S-methylsulfinimidoyl)phenyl]phenyl]methyl]-N-[(4-methylphenyl)methyl]benzamide Chemical compound Cc1ccc(CN(Cc2ccc(cc2)-c2ccccc2\S(C)=N\C#N)C(=O)c2ccccc2Cl)cc1 XNMWTPWQILTFRI-UHFFFAOYSA-N 0.000 description 2
- 125000004398 2-methyl-2-butyl group Chemical group CC(C)(CC)* 0.000 description 2
- 125000004918 2-methyl-2-pentyl group Chemical group CC(C)(CCC)* 0.000 description 2
- 125000004922 2-methyl-3-pentyl group Chemical group CC(C)C(CC)* 0.000 description 2
- YCPYNHRVVOCFEF-UHFFFAOYSA-N 3-chloro-6-methoxy-2-(2-phenylethynyl)pyridine Chemical compound COC1=CC=C(Cl)C(C#CC=2C=CC=CC=2)=N1 YCPYNHRVVOCFEF-UHFFFAOYSA-N 0.000 description 2
- AEVSSZHXGJAPIE-UHFFFAOYSA-N 3-chloropyrazin-2-amine Chemical compound NC1=NC=CN=C1Cl AEVSSZHXGJAPIE-UHFFFAOYSA-N 0.000 description 2
- ZTZOSAQQMYQVRA-UHFFFAOYSA-N 3-iodo-2-methoxypyridin-4-amine Chemical compound COC1=NC=CC(N)=C1I ZTZOSAQQMYQVRA-UHFFFAOYSA-N 0.000 description 2
- 125000004917 3-methyl-2-butyl group Chemical group CC(C(C)*)C 0.000 description 2
- 125000004919 3-methyl-2-pentyl group Chemical group CC(C(C)*)CC 0.000 description 2
- 125000004921 3-methyl-3-pentyl group Chemical group CC(CC)(CC)* 0.000 description 2
- LZPWAYBEOJRFAX-UHFFFAOYSA-N 4,4,5,5-tetramethyl-1,3,2$l^{2}-dioxaborolane Chemical compound CC1(C)O[B]OC1(C)C LZPWAYBEOJRFAX-UHFFFAOYSA-N 0.000 description 2
- YKSZLCOPANWXES-UHFFFAOYSA-N 4,6-dichloro-5-iodopyrimidine Chemical compound ClC1=NC=NC(Cl)=C1I YKSZLCOPANWXES-UHFFFAOYSA-N 0.000 description 2
- DWWDFIMREPVKNS-UHFFFAOYSA-N 4-chloro-3-(2-phenylethynyl)pyridine Chemical compound ClC1=CC=NC=C1C#CC1=CC=CC=C1 DWWDFIMREPVKNS-UHFFFAOYSA-N 0.000 description 2
- WJCQPBKEZPVKGV-UHFFFAOYSA-N 4-chloro-5-(2-phenylethynyl)pyrimidine Chemical compound ClC1=NC=NC=C1C#CC1=CC=CC=C1 WJCQPBKEZPVKGV-UHFFFAOYSA-N 0.000 description 2
- 150000005741 4-chloro-5-iodopyrimidines Chemical class 0.000 description 2
- BPTCCCTWWAUJRK-UHFFFAOYSA-N 4-chloro-7h-pyrrolo[2,3-d]pyrimidine Chemical class ClC1=NC=NC2=C1C=CN2 BPTCCCTWWAUJRK-UHFFFAOYSA-N 0.000 description 2
- 125000004920 4-methyl-2-pentyl group Chemical group CC(CC(C)*)C 0.000 description 2
- WWQQPSDIIVXFOX-UHFFFAOYSA-N 5-bromo-2-chloro-3-nitropyridine Chemical compound [O-][N+](=O)C1=CC(Br)=CN=C1Cl WWQQPSDIIVXFOX-UHFFFAOYSA-N 0.000 description 2
- OQUISEMWDUNAIH-UHFFFAOYSA-N 6-(2-fluorophenyl)-3-(2-phenylethynyl)pyridin-2-amine Chemical compound NC1=NC(C=2C(=CC=CC=2)F)=CC=C1C#CC1=CC=CC=C1 OQUISEMWDUNAIH-UHFFFAOYSA-N 0.000 description 2
- XPHWCAKVRKUXLK-UHFFFAOYSA-N 6-bromo-1h-imidazo[4,5-b]pyridine Chemical compound BrC1=CN=C2N=CNC2=C1 XPHWCAKVRKUXLK-UHFFFAOYSA-N 0.000 description 2
- DSCOSIUAQUDGJM-UHFFFAOYSA-N 6-chloro-1h-pyrrolo[2,3-b]pyridine Chemical class ClC1=CC=C2C=CNC2=N1 DSCOSIUAQUDGJM-UHFFFAOYSA-N 0.000 description 2
- DZPIZFVSNWQVTG-UHFFFAOYSA-N 6-chloro-3-(2-phenylethynyl)pyridin-2-amine Chemical compound NC1=NC(Cl)=CC=C1C#CC1=CC=CC=C1 DZPIZFVSNWQVTG-UHFFFAOYSA-N 0.000 description 2
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 2
- LZIRCTPGZKOZMQ-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidine-2-carboxamide Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(C(=O)N)=NC=C2C=C1C1=CC=CC=N1 LZIRCTPGZKOZMQ-UHFFFAOYSA-N 0.000 description 2
- DEVFVVLSBJSUNB-UHFFFAOYSA-N 7h-purine;hydrochloride Chemical compound Cl.N1=CNC2=NC=NC2=C1 DEVFVVLSBJSUNB-UHFFFAOYSA-N 0.000 description 2
- UJOBWOGCFQCDNV-UHFFFAOYSA-N 9H-carbazole Chemical compound C1=CC=C2C3=CC=CC=C3NC2=C1 UJOBWOGCFQCDNV-UHFFFAOYSA-N 0.000 description 2
- 244000215068 Acacia senegal Species 0.000 description 2
- 235000006491 Acacia senegal Nutrition 0.000 description 2
- 201000003076 Angiosarcoma Diseases 0.000 description 2
- 235000003911 Arachis Nutrition 0.000 description 2
- 244000105624 Arachis hypogaea Species 0.000 description 2
- 239000004475 Arginine Substances 0.000 description 2
- XKRFYHLGVUSROY-UHFFFAOYSA-N Argon Chemical compound [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 description 2
- BFYIZQONLCFLEV-DAELLWKTSA-N Aromasine Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CC(=C)C2=C1 BFYIZQONLCFLEV-DAELLWKTSA-N 0.000 description 2
- 108010011485 Aspartame Proteins 0.000 description 2
- 241000416162 Astragalus gummifer Species 0.000 description 2
- MLDQJTXFUGDVEO-UHFFFAOYSA-N BAY-43-9006 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 MLDQJTXFUGDVEO-UHFFFAOYSA-N 0.000 description 2
- LSNNMFCWUKXFEE-UHFFFAOYSA-M Bisulfite Chemical compound OS([O-])=O LSNNMFCWUKXFEE-UHFFFAOYSA-M 0.000 description 2
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 2
- KXDHJXZQYSOELW-UHFFFAOYSA-M Carbamate Chemical compound NC([O-])=O KXDHJXZQYSOELW-UHFFFAOYSA-M 0.000 description 2
- 239000004215 Carbon black (E152) Substances 0.000 description 2
- 241000223782 Ciliophora Species 0.000 description 2
- 235000001258 Cinchona calisaya Nutrition 0.000 description 2
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 2
- 201000002847 Cowden syndrome Diseases 0.000 description 2
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 2
- 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 2
- 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 2
- GSNUFIFRDBKVIE-UHFFFAOYSA-N DMF Natural products CC1=CC=C(C)O1 GSNUFIFRDBKVIE-UHFFFAOYSA-N 0.000 description 2
- 108010092160 Dactinomycin Proteins 0.000 description 2
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 2
- 206010061818 Disease progression Diseases 0.000 description 2
- 102100030013 Endoribonuclease Human genes 0.000 description 2
- 101710199605 Endoribonuclease Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- VWUXBMIQPBEWFH-WCCTWKNTSA-N Fulvestrant Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3[C@H](CCCCCCCCCS(=O)CCCC(F)(F)C(F)(F)F)CC2=C1 VWUXBMIQPBEWFH-WCCTWKNTSA-N 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- IAJILQKETJEXLJ-UHFFFAOYSA-N Galacturonsaeure Natural products O=CC(O)C(O)C(O)C(O)C(O)=O IAJILQKETJEXLJ-UHFFFAOYSA-N 0.000 description 2
- 208000032612 Glial tumor Diseases 0.000 description 2
- 206010018338 Glioma Diseases 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- AEMRFAOFKBGASW-UHFFFAOYSA-N Glycolic acid Chemical compound OCC(O)=O AEMRFAOFKBGASW-UHFFFAOYSA-N 0.000 description 2
- 229920000084 Gum arabic Polymers 0.000 description 2
- 208000002927 Hamartoma Diseases 0.000 description 2
- 208000001258 Hemangiosarcoma Diseases 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101001117143 Homo sapiens [Pyruvate dehydrogenase (acetyl-transferring)] kinase isozyme 2, mitochondrial Proteins 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 2
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 2
- ZGUNAGUHMKGQNY-ZETCQYMHSA-N L-alpha-phenylglycine zwitterion Chemical compound OC(=O)[C@@H](N)C1=CC=CC=C1 ZGUNAGUHMKGQNY-ZETCQYMHSA-N 0.000 description 2
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 2
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 2
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 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
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 2
- 239000002136 L01XE07 - Lapatinib Substances 0.000 description 2
- 208000018142 Leiomyosarcoma Diseases 0.000 description 2
- 108010000817 Leuprolide Proteins 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 239000004472 Lysine Substances 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 206010027476 Metastases Diseases 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- YNAVUWVOSKDBBP-UHFFFAOYSA-N Morpholine Chemical group C1COCCN1 YNAVUWVOSKDBBP-UHFFFAOYSA-N 0.000 description 2
- 208000034578 Multiple myelomas Diseases 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- QIAFMBKCNZACKA-UHFFFAOYSA-N N-benzoylglycine Chemical compound OC(=O)CNC(=O)C1=CC=CC=C1 QIAFMBKCNZACKA-UHFFFAOYSA-N 0.000 description 2
- UFWIBTONFRDIAS-UHFFFAOYSA-N Naphthalene Chemical compound C1=CC=CC2=CC=CC=C21 UFWIBTONFRDIAS-UHFFFAOYSA-N 0.000 description 2
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 2
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 2
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 2
- 229930012538 Paclitaxel Natural products 0.000 description 2
- 208000031481 Pathologic Constriction Diseases 0.000 description 2
- NFHFRUOZVGFOOS-UHFFFAOYSA-N Pd(PPh3)4 Substances [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 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Natural products OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical group C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- 102000005765 Proto-Oncogene Proteins c-akt Human genes 0.000 description 2
- 108010045717 Proto-Oncogene Proteins c-akt Proteins 0.000 description 2
- WTKZEGDFNFYCGP-UHFFFAOYSA-N Pyrazole Chemical group C=1C=NNC=1 WTKZEGDFNFYCGP-UHFFFAOYSA-N 0.000 description 2
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical group C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 2
- LCTONWCANYUPML-UHFFFAOYSA-N Pyruvic acid Chemical compound CC(=O)C(O)=O LCTONWCANYUPML-UHFFFAOYSA-N 0.000 description 2
- IWYDHOAUDWTVEP-UHFFFAOYSA-N R-2-phenyl-2-hydroxyacetic acid Natural products OC(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-N 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 101710113029 Serine/threonine-protein kinase Proteins 0.000 description 2
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 2
- 102000013530 TOR Serine-Threonine Kinases Human genes 0.000 description 2
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 2
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 2
- 229920001615 Tragacanth Polymers 0.000 description 2
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 2
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 2
- 208000008383 Wilms tumor Diseases 0.000 description 2
- 102100024150 [Pyruvate dehydrogenase (acetyl-transferring)] kinase isozyme 2, mitochondrial Human genes 0.000 description 2
- 235000010489 acacia gum Nutrition 0.000 description 2
- SPEUIVXLLWOEMJ-UHFFFAOYSA-N acetaldehyde dimethyl acetal Natural products COC(C)OC SPEUIVXLLWOEMJ-UHFFFAOYSA-N 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000008186 active pharmaceutical agent Substances 0.000 description 2
- 125000002252 acyl group Chemical group 0.000 description 2
- 210000004100 adrenal gland Anatomy 0.000 description 2
- 238000012382 advanced drug delivery Methods 0.000 description 2
- 125000003158 alcohol group Chemical group 0.000 description 2
- 229960000548 alemtuzumab Drugs 0.000 description 2
- 125000005907 alkyl ester group Chemical group 0.000 description 2
- 125000004422 alkyl sulphonamide group Chemical group 0.000 description 2
- 125000002947 alkylene group Chemical group 0.000 description 2
- 125000006242 amine protecting group Chemical group 0.000 description 2
- ROBVIMPUHSLWNV-UHFFFAOYSA-N aminoglutethimide Chemical compound C=1C=C(N)C=CC=1C1(CC)CCC(=O)NC1=O ROBVIMPUHSLWNV-UHFFFAOYSA-N 0.000 description 2
- 229960003437 aminoglutethimide Drugs 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- YBBLVLTVTVSKRW-UHFFFAOYSA-N anastrozole Chemical compound N#CC(C)(C)C1=CC(C(C)(C#N)C)=CC(CN2N=CN=C2)=C1 YBBLVLTVTVSKRW-UHFFFAOYSA-N 0.000 description 2
- MWPLVEDNUUSJAV-UHFFFAOYSA-N anthracene Chemical compound C1=CC=CC2=CC3=CC=CC=C3C=C21 MWPLVEDNUUSJAV-UHFFFAOYSA-N 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 239000003125 aqueous solvent Substances 0.000 description 2
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 2
- IAOZJIPTCAWIRG-QWRGUYRKSA-N aspartame Chemical compound OC(=O)C[C@H](N)C(=O)N[C@H](C(=O)OC)CC1=CC=CC=C1 IAOZJIPTCAWIRG-QWRGUYRKSA-N 0.000 description 2
- 239000000605 aspartame Substances 0.000 description 2
- 235000010357 aspartame Nutrition 0.000 description 2
- 229960003438 aspartame Drugs 0.000 description 2
- 229940120638 avastin Drugs 0.000 description 2
- HONIICLYMWZJFZ-UHFFFAOYSA-N azetidine Chemical compound C1CNC1 HONIICLYMWZJFZ-UHFFFAOYSA-N 0.000 description 2
- 150000001541 aziridines Chemical class 0.000 description 2
- 125000004069 aziridinyl group Chemical group 0.000 description 2
- 239000008228 bacteriostatic water for injection Substances 0.000 description 2
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 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
- UCMIRNVEIXFBKS-UHFFFAOYSA-N beta-alanine Chemical compound NCCC(O)=O UCMIRNVEIXFBKS-UHFFFAOYSA-N 0.000 description 2
- 125000002619 bicyclic group Chemical group 0.000 description 2
- 239000012148 binding buffer Substances 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 238000009835 boiling Methods 0.000 description 2
- WTEOIRVLGSZEPR-UHFFFAOYSA-N boron trifluoride Chemical class FB(F)F WTEOIRVLGSZEPR-UHFFFAOYSA-N 0.000 description 2
- GXJABQQUPOEUTA-RDJZCZTQSA-N bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 description 2
- 210000000481 breast Anatomy 0.000 description 2
- 125000001246 bromo group Chemical group Br* 0.000 description 2
- 239000006227 byproduct Substances 0.000 description 2
- 229910000019 calcium carbonate Inorganic materials 0.000 description 2
- 235000010216 calcium carbonate Nutrition 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 125000001589 carboacyl group Chemical group 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 230000006315 carbonylation Effects 0.000 description 2
- 238000005810 carbonylation reaction Methods 0.000 description 2
- 208000002458 carcinoid tumor Diseases 0.000 description 2
- YCIMNLLNPGFGHC-UHFFFAOYSA-N catechol Chemical compound OC1=CC=CC=C1O YCIMNLLNPGFGHC-UHFFFAOYSA-N 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 239000002738 chelating agent Substances 0.000 description 2
- LOUPRKONTZGTKE-UHFFFAOYSA-N cinchonine Natural products C1C(C(C2)C=C)CCN2C1C(O)C1=CC=NC2=CC=C(OC)C=C21 LOUPRKONTZGTKE-UHFFFAOYSA-N 0.000 description 2
- QTMDXZNDVAMKGV-UHFFFAOYSA-L copper(ii) bromide Chemical compound [Cu+2].[Br-].[Br-] QTMDXZNDVAMKGV-UHFFFAOYSA-L 0.000 description 2
- 239000007819 coupling partner Substances 0.000 description 2
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 2
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 2
- 239000003405 delayed action preparation Substances 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 238000006193 diazotization reaction Methods 0.000 description 2
- 125000004852 dihydrofuranyl group Chemical group O1C(CC=C1)* 0.000 description 2
- 125000005043 dihydropyranyl group Chemical group O1C(CCC=C1)* 0.000 description 2
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N diphenyl Chemical compound C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 description 2
- 230000005750 disease progression Effects 0.000 description 2
- 208000037765 diseases and disorders Diseases 0.000 description 2
- 238000004821 distillation Methods 0.000 description 2
- VLCYCQAOQCDTCN-UHFFFAOYSA-N eflornithine Chemical compound NCCCC(N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-UHFFFAOYSA-N 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- AAKJLRGGTJKAMG-UHFFFAOYSA-N erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 2
- 239000004403 ethyl p-hydroxybenzoate Substances 0.000 description 2
- 235000010228 ethyl p-hydroxybenzoate Nutrition 0.000 description 2
- 229940087476 femara Drugs 0.000 description 2
- 125000001153 fluoro group Chemical group F* 0.000 description 2
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 2
- CHPZKNULDCNCBW-UHFFFAOYSA-N gallium nitrate Chemical compound [Ga+3].[O-][N+]([O-])=O.[O-][N+]([O-])=O.[O-][N+]([O-])=O CHPZKNULDCNCBW-UHFFFAOYSA-N 0.000 description 2
- BTCSSZJGUNDROE-UHFFFAOYSA-N gamma-aminobutyric acid Chemical compound NCCCC(O)=O BTCSSZJGUNDROE-UHFFFAOYSA-N 0.000 description 2
- 206010017758 gastric cancer Diseases 0.000 description 2
- 208000010749 gastric carcinoma Diseases 0.000 description 2
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 2
- 239000007903 gelatin capsule Substances 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- 208000005017 glioblastoma Diseases 0.000 description 2
- 229960002989 glutamic acid Drugs 0.000 description 2
- 150000004820 halides Chemical class 0.000 description 2
- 125000004438 haloalkoxy group Chemical group 0.000 description 2
- 125000001188 haloalkyl group Chemical group 0.000 description 2
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 2
- BXWNKGSJHAJOGX-UHFFFAOYSA-N hexadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCO BXWNKGSJHAJOGX-UHFFFAOYSA-N 0.000 description 2
- FBPFZTCFMRRESA-UHFFFAOYSA-N hexane-1,2,3,4,5,6-hexol Chemical compound OCC(O)C(O)C(O)C(O)CO FBPFZTCFMRRESA-UHFFFAOYSA-N 0.000 description 2
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 2
- 229930195733 hydrocarbon Natural products 0.000 description 2
- 230000007062 hydrolysis Effects 0.000 description 2
- 238000006460 hydrolysis reaction Methods 0.000 description 2
- 125000002768 hydroxyalkyl group Chemical group 0.000 description 2
- 229960001101 ifosfamide Drugs 0.000 description 2
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 2
- YLMAHDNUQAMNNX-UHFFFAOYSA-N imatinib methanesulfonate Chemical compound CS(O)(=O)=O.C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 YLMAHDNUQAMNNX-UHFFFAOYSA-N 0.000 description 2
- 125000002883 imidazolyl group Chemical group 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000000099 in vitro assay Methods 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 230000001965 increasing effect Effects 0.000 description 2
- PQNFLJBBNBOBRQ-UHFFFAOYSA-N indane Chemical compound C1=CC=C2CCCC2=C1 PQNFLJBBNBOBRQ-UHFFFAOYSA-N 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 2
- 125000000904 isoindolyl group Chemical group C=1(NC=C2C=CC=CC12)* 0.000 description 2
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 2
- 238000000021 kinase assay Methods 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 235000010445 lecithin Nutrition 0.000 description 2
- 239000000787 lecithin Substances 0.000 description 2
- 229940067606 lecithin Drugs 0.000 description 2
- 201000010260 leiomyoma Diseases 0.000 description 2
- 229960003881 letrozole Drugs 0.000 description 2
- GFIJNRVAKGFPGQ-LIJARHBVSA-N leuprolide Chemical compound CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 GFIJNRVAKGFPGQ-LIJARHBVSA-N 0.000 description 2
- 229960004338 leuprorelin Drugs 0.000 description 2
- UBJFKNSINUCEAL-UHFFFAOYSA-N lithium;2-methylpropane Chemical compound [Li+].C[C-](C)C UBJFKNSINUCEAL-UHFFFAOYSA-N 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- 239000012931 lyophilized formulation Substances 0.000 description 2
- RLSSMJSEOOYNOY-UHFFFAOYSA-N m-cresol Chemical compound CC1=CC=CC(O)=C1 RLSSMJSEOOYNOY-UHFFFAOYSA-N 0.000 description 2
- 208000002780 macular degeneration Diseases 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- IUYHWZFSGMZEOG-UHFFFAOYSA-M magnesium;propane;chloride Chemical compound [Mg+2].[Cl-].C[CH-]C IUYHWZFSGMZEOG-UHFFFAOYSA-M 0.000 description 2
- 229960002510 mandelic acid Drugs 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 206010027191 meningioma Diseases 0.000 description 2
- 229960001428 mercaptopurine Drugs 0.000 description 2
- 230000004060 metabolic process Effects 0.000 description 2
- 238000006263 metalation reaction Methods 0.000 description 2
- 229910052752 metalloid Inorganic materials 0.000 description 2
- 150000002738 metalloids Chemical class 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- MGJXBDMLVWIYOQ-UHFFFAOYSA-N methylazanide Chemical compound [NH-]C MGJXBDMLVWIYOQ-UHFFFAOYSA-N 0.000 description 2
- 125000004170 methylsulfonyl group Chemical group [H]C([H])([H])S(*)(=O)=O 0.000 description 2
- 239000004005 microsphere Substances 0.000 description 2
- 239000002480 mineral oil Substances 0.000 description 2
- 235000010446 mineral oil Nutrition 0.000 description 2
- 150000007522 mineralic acids Chemical class 0.000 description 2
- 229960001156 mitoxantrone Drugs 0.000 description 2
- 210000000214 mouth Anatomy 0.000 description 2
- 201000006417 multiple sclerosis Diseases 0.000 description 2
- LBWFXVZLPYTWQI-IPOVEDGCSA-N n-[2-(diethylamino)ethyl]-5-[(z)-(5-fluoro-2-oxo-1h-indol-3-ylidene)methyl]-2,4-dimethyl-1h-pyrrole-3-carboxamide;(2s)-2-hydroxybutanedioic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O.CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C LBWFXVZLPYTWQI-IPOVEDGCSA-N 0.000 description 2
- VAAFEQYKHZGPJQ-UHFFFAOYSA-N n-[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-2-yl]methanesulfonamide Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(NS(=O)(=O)C)=NC=C2C=C1C1=CC=CC=N1 VAAFEQYKHZGPJQ-UHFFFAOYSA-N 0.000 description 2
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 2
- 239000002105 nanoparticle Substances 0.000 description 2
- QZGIWPZCWHMVQL-UIYAJPBUSA-N neocarzinostatin chromophore Chemical compound O1[C@H](C)[C@H](O)[C@H](O)[C@@H](NC)[C@H]1O[C@@H]1C/2=C/C#C[C@H]3O[C@@]3([C@@H]3OC(=O)OC3)C#CC\2=C[C@H]1OC(=O)C1=C(O)C=CC2=C(C)C=C(OC)C=C12 QZGIWPZCWHMVQL-UIYAJPBUSA-N 0.000 description 2
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 2
- 239000000346 nonvolatile oil Substances 0.000 description 2
- 238000007339 nucleophilic aromatic substitution reaction Methods 0.000 description 2
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 150000002894 organic compounds Chemical class 0.000 description 2
- 125000002734 organomagnesium group Chemical group 0.000 description 2
- 229960003104 ornithine Drugs 0.000 description 2
- 150000002905 orthoesters Chemical class 0.000 description 2
- 201000008968 osteosarcoma Diseases 0.000 description 2
- 230000002611 ovarian Effects 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 229960001592 paclitaxel Drugs 0.000 description 2
- LXNAVEXFUKBNMK-UHFFFAOYSA-N palladium(II) acetate Substances [Pd].CC(O)=O.CC(O)=O LXNAVEXFUKBNMK-UHFFFAOYSA-N 0.000 description 2
- 229960001972 panitumumab Drugs 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- AQIXEPGDORPWBJ-UHFFFAOYSA-N pentan-3-ol Chemical compound CCC(O)CC AQIXEPGDORPWBJ-UHFFFAOYSA-N 0.000 description 2
- 125000003538 pentan-3-yl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])[H] 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 2
- 125000004193 piperazinyl group Chemical group 0.000 description 2
- 125000003367 polycyclic group Chemical group 0.000 description 2
- 229920001184 polypeptide Chemical group 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 102000004196 processed proteins & peptides Human genes 0.000 description 2
- 235000018102 proteins Nutrition 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 125000003373 pyrazinyl group Chemical group 0.000 description 2
- 125000003226 pyrazolyl group Chemical group 0.000 description 2
- 125000004076 pyridyl group Chemical group 0.000 description 2
- 125000004527 pyrimidin-4-yl group Chemical group N1=CN=C(C=C1)* 0.000 description 2
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 2
- 229960000948 quinine Drugs 0.000 description 2
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 description 2
- 229960004622 raloxifene Drugs 0.000 description 2
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 2
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 2
- 230000009257 reactivity Effects 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 230000007115 recruitment Effects 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- GHMLBKRAJCXXBS-UHFFFAOYSA-N resorcinol Chemical compound OC1=CC=CC(O)=C1 GHMLBKRAJCXXBS-UHFFFAOYSA-N 0.000 description 2
- 229960004641 rituximab Drugs 0.000 description 2
- FSYKKLYZXJSNPZ-UHFFFAOYSA-N sarcosine Chemical compound C[NH2+]CC([O-])=O FSYKKLYZXJSNPZ-UHFFFAOYSA-N 0.000 description 2
- 229940095743 selective estrogen receptor modulator Drugs 0.000 description 2
- 239000000333 selective estrogen receptor modulator Substances 0.000 description 2
- 229960002930 sirolimus Drugs 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 235000015424 sodium Nutrition 0.000 description 2
- 229910000029 sodium carbonate Inorganic materials 0.000 description 2
- 239000012312 sodium hydride Substances 0.000 description 2
- 229910000104 sodium hydride Inorganic materials 0.000 description 2
- 235000009518 sodium iodide Nutrition 0.000 description 2
- 239000004317 sodium nitrate Substances 0.000 description 2
- 235000010344 sodium nitrate Nutrition 0.000 description 2
- 229960003787 sorafenib Drugs 0.000 description 2
- 235000011069 sorbitan monooleate Nutrition 0.000 description 2
- 239000001593 sorbitan monooleate Substances 0.000 description 2
- 229940035049 sorbitan monooleate Drugs 0.000 description 2
- 239000000600 sorbitol Substances 0.000 description 2
- 125000003003 spiro group Chemical group 0.000 description 2
- 230000036262 stenosis Effects 0.000 description 2
- 208000037804 stenosis Diseases 0.000 description 2
- 201000000498 stomach carcinoma Diseases 0.000 description 2
- PVYJZLYGTZKPJE-UHFFFAOYSA-N streptonigrin Chemical compound C=1C=C2C(=O)C(OC)=C(N)C(=O)C2=NC=1C(C=1N)=NC(C(O)=O)=C(C)C=1C1=CC=C(OC)C(OC)=C1O PVYJZLYGTZKPJE-UHFFFAOYSA-N 0.000 description 2
- 238000000859 sublimation Methods 0.000 description 2
- 230000008022 sublimation Effects 0.000 description 2
- 150000003871 sulfonates Chemical class 0.000 description 2
- 150000003467 sulfuric acid derivatives Chemical class 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 239000007916 tablet composition Substances 0.000 description 2
- FQZYTYWMLGAPFJ-OQKDUQJOSA-N tamoxifen citrate Chemical compound [H+].[H+].[H+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O.C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 FQZYTYWMLGAPFJ-OQKDUQJOSA-N 0.000 description 2
- 239000011975 tartaric acid Substances 0.000 description 2
- 235000002906 tartaric acid Nutrition 0.000 description 2
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 2
- CDYARKRFFXCNOS-UHFFFAOYSA-N tert-butyl n-[1-[4-[2-(2-oxopyrrolidin-1-yl)-8-phenylpurin-9-yl]phenyl]cyclobutyl]carbamate Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2C=CC=CC=2)N2C(CCC2)=O)C=CC=1C1(NC(=O)OC(C)(C)C)CCC1 CDYARKRFFXCNOS-UHFFFAOYSA-N 0.000 description 2
- YTTHMKVRASRPTJ-UHFFFAOYSA-N tert-butyl n-[1-[4-[2-(3-hydroxypyrrolidin-1-yl)-8-pyridin-2-ylpurin-9-yl]phenyl]cyclobutyl]carbamate Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2N=CC=CC=2)N2CC(O)CC2)C=CC=1C1(NC(=O)OC(C)(C)C)CCC1 YTTHMKVRASRPTJ-UHFFFAOYSA-N 0.000 description 2
- JOSYQFMFPVLMPK-UHFFFAOYSA-N tert-butyl n-[1-[4-[[5-amino-2-[3-(trifluoromethoxy)phenyl]pyrimidin-4-yl]amino]phenyl]cyclobutyl]carbamate Chemical compound C=1C=C(NC=2C(=CN=C(N=2)C=2C=C(OC(F)(F)F)C=CC=2)N)C=CC=1C1(NC(=O)OC(C)(C)C)CCC1 JOSYQFMFPVLMPK-UHFFFAOYSA-N 0.000 description 2
- 125000001113 thiadiazolyl group Chemical group 0.000 description 2
- 125000000335 thiazolyl group Chemical group 0.000 description 2
- 239000002562 thickening agent Substances 0.000 description 2
- 229960001196 thiotepa Drugs 0.000 description 2
- 229960003087 tioguanine Drugs 0.000 description 2
- 229960003989 tocilizumab Drugs 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 229960005267 tositumomab Drugs 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 229960000575 trastuzumab Drugs 0.000 description 2
- 125000001425 triazolyl group Chemical group 0.000 description 2
- YYGNJPDQYSBWPH-UHFFFAOYSA-N tributyl-(3-methyl-1,2-oxazol-5-yl)stannane Chemical compound CCCC[Sn](CCCC)(CCCC)C1=CC(C)=NO1 YYGNJPDQYSBWPH-UHFFFAOYSA-N 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 2
- WJKHJLXJJJATHN-UHFFFAOYSA-N triflic anhydride Chemical compound FC(F)(F)S(=O)(=O)OS(=O)(=O)C(F)(F)F WJKHJLXJJJATHN-UHFFFAOYSA-N 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 2
- 229960005486 vaccine Drugs 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- JIAARYAFYJHUJI-UHFFFAOYSA-L zinc dichloride Chemical compound [Cl-].[Cl-].[Zn+2] JIAARYAFYJHUJI-UHFFFAOYSA-L 0.000 description 2
- AIFRHYZBTHREPW-UHFFFAOYSA-N β-carboline Chemical compound N1=CC=C2C3=CC=CC=C3NC2=C1 AIFRHYZBTHREPW-UHFFFAOYSA-N 0.000 description 2
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-N 0.000 description 1
- OGNSCSPNOLGXSM-UHFFFAOYSA-N (+/-)-DABA Natural products NCCC(N)C(O)=O OGNSCSPNOLGXSM-UHFFFAOYSA-N 0.000 description 1
- NNJPGOLRFBJNIW-HNNXBMFYSA-N (-)-demecolcine Chemical compound C1=C(OC)C(=O)C=C2[C@@H](NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-HNNXBMFYSA-N 0.000 description 1
- QCSLIRFWJPOENV-UHFFFAOYSA-N (2-fluorophenyl)boronic acid Chemical compound OB(O)C1=CC=CC=C1F QCSLIRFWJPOENV-UHFFFAOYSA-N 0.000 description 1
- LDDMACCNBZAMSG-BDVNFPICSA-N (2r,3r,4s,5r)-3,4,5,6-tetrahydroxy-2-(methylamino)hexanal Chemical compound CN[C@@H](C=O)[C@@H](O)[C@H](O)[C@H](O)CO LDDMACCNBZAMSG-BDVNFPICSA-N 0.000 description 1
- WDQLRUYAYXDIFW-RWKIJVEZSA-N (2r,3r,4s,5r,6r)-4-[(2s,3r,4s,5r,6r)-3,5-dihydroxy-4-[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-6-[[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxymethyl]oxan-2-yl]oxy-6-(hydroxymethyl)oxane-2,3,5-triol Chemical compound O[C@@H]1[C@@H](CO)O[C@@H](O)[C@H](O)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)[C@H](O)[C@@H](CO[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)O1 WDQLRUYAYXDIFW-RWKIJVEZSA-N 0.000 description 1
- 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 1
- GZHQVQIJLXZTQQ-NRFANRHFSA-N (2s)-1-[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-2-yl]pyrrolidine-2-carboxylic acid Chemical compound C=1C=C(N2C3=NC(=NC=C3C=C2C=2N=CC=CC=2)N2[C@@H](CCC2)C(O)=O)C=CC=1C1(N)CCC1 GZHQVQIJLXZTQQ-NRFANRHFSA-N 0.000 description 1
- FLWWDYNPWOSLEO-HQVZTVAUSA-N (2s)-2-[[4-[1-(2-amino-4-oxo-1h-pteridin-6-yl)ethyl-methylamino]benzoyl]amino]pentanedioic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1C(C)N(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FLWWDYNPWOSLEO-HQVZTVAUSA-N 0.000 description 1
- XMQUEQJCYRFIQS-YFKPBYRVSA-N (2s)-2-amino-5-ethoxy-5-oxopentanoic acid Chemical compound CCOC(=O)CC[C@H](N)C(O)=O XMQUEQJCYRFIQS-YFKPBYRVSA-N 0.000 description 1
- PAORVUMOXXAMPL-SECBINFHSA-N (2s)-3,3,3-trifluoro-2-methoxy-2-phenylpropanoyl chloride Chemical compound CO[C@](C(Cl)=O)(C(F)(F)F)C1=CC=CC=C1 PAORVUMOXXAMPL-SECBINFHSA-N 0.000 description 1
- CQYBNXGHMBNGCG-FXQIFTODSA-N (2s,3as,7as)-2,3,3a,4,5,6,7,7a-octahydro-1h-indol-1-ium-2-carboxylate Chemical compound C1CCC[C@@H]2[NH2+][C@H](C(=O)[O-])C[C@@H]21 CQYBNXGHMBNGCG-FXQIFTODSA-N 0.000 description 1
- LJRDOKAZOAKLDU-UDXJMMFXSA-N (2s,3s,4r,5r,6r)-5-amino-2-(aminomethyl)-6-[(2r,3s,4r,5s)-5-[(1r,2r,3s,5r,6s)-3,5-diamino-2-[(2s,3r,4r,5s,6r)-3-amino-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-6-hydroxycyclohexyl]oxy-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl]oxyoxane-3,4-diol;sulfuric ac Chemical compound OS(O)(=O)=O.N[C@@H]1[C@@H](O)[C@H](O)[C@H](CN)O[C@@H]1O[C@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](N)C[C@@H](N)[C@@H]2O)O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)N)O[C@@H]1CO LJRDOKAZOAKLDU-UDXJMMFXSA-N 0.000 description 1
- CGMTUJFWROPELF-YPAAEMCBSA-N (3E,5S)-5-[(2S)-butan-2-yl]-3-(1-hydroxyethylidene)pyrrolidine-2,4-dione Chemical compound CC[C@H](C)[C@@H]1NC(=O)\C(=C(/C)O)C1=O CGMTUJFWROPELF-YPAAEMCBSA-N 0.000 description 1
- CWCSQLVDSHQDCK-FQEVSTJZSA-N (3s)-1-[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-2-yl]pyrrolidin-3-ol Chemical compound C=1C=C(N2C3=NC(=NC=C3C=C2C=2N=CC=CC=2)N2C[C@@H](O)CC2)C=CC=1C1(N)CCC1 CWCSQLVDSHQDCK-FQEVSTJZSA-N 0.000 description 1
- TVIRNGFXQVMMGB-OFWIHYRESA-N (3s,6r,10r,13e,16s)-16-[(2r,3r,4s)-4-chloro-3-hydroxy-4-phenylbutan-2-yl]-10-[(3-chloro-4-methoxyphenyl)methyl]-6-methyl-3-(2-methylpropyl)-1,4-dioxa-8,11-diazacyclohexadec-13-ene-2,5,9,12-tetrone Chemical compound C1=C(Cl)C(OC)=CC=C1C[C@@H]1C(=O)NC[C@@H](C)C(=O)O[C@@H](CC(C)C)C(=O)O[C@H]([C@H](C)[C@@H](O)[C@@H](Cl)C=2C=CC=CC=2)C/C=C/C(=O)N1 TVIRNGFXQVMMGB-OFWIHYRESA-N 0.000 description 1
- KJAXEBRGQOHHOY-VXRVIWLSSA-N (4s)-4-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s,3r)-2-[[(2s)-2-[[(2s)-1-[(2s)-2-[(2-aminoacetyl)amino]-5-(diaminomethylideneamino)pentanoyl]pyrrolidine-2-carbonyl]amino]-5-(diaminomethylideneamino)pentanoyl]amino]-3-hydroxybutanoyl]amino]-3-hydroxypropan Chemical compound N([C@@H](CCCN=C(N)N)C(=O)N[C@@H]([C@H](O)C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)NCC(O)=O)C(=O)[C@@H]1CCCN1C(=O)[C@H](CCCN=C(N)N)NC(=O)CN KJAXEBRGQOHHOY-VXRVIWLSSA-N 0.000 description 1
- KIUPCUCGVCGPPA-UHFFFAOYSA-N (5-methyl-2-propan-2-ylcyclohexyl) carbonochloridate Chemical compound CC(C)C1CCC(C)CC1OC(Cl)=O KIUPCUCGVCGPPA-UHFFFAOYSA-N 0.000 description 1
- XRBSKUSTLXISAB-XVVDYKMHSA-N (5r,6r,7r,8r)-8-hydroxy-7-(hydroxymethyl)-5-(3,4,5-trimethoxyphenyl)-5,6,7,8-tetrahydrobenzo[f][1,3]benzodioxole-6-carboxylic acid Chemical compound COC1=C(OC)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@H](O)[C@@H](CO)[C@@H]2C(O)=O)=C1 XRBSKUSTLXISAB-XVVDYKMHSA-N 0.000 description 1
- XRBSKUSTLXISAB-UHFFFAOYSA-N (7R,7'R,8R,8'R)-form-Podophyllic acid Natural products COC1=C(OC)C(OC)=CC(C2C3=CC=4OCOC=4C=C3C(O)C(CO)C2C(O)=O)=C1 XRBSKUSTLXISAB-UHFFFAOYSA-N 0.000 description 1
- AESVUZLWRXEGEX-DKCAWCKPSA-N (7S,9R)-7-[(2S,4R,5R,6R)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7H-tetracene-5,12-dione iron(3+) Chemical compound [Fe+3].COc1cccc2C(=O)c3c(O)c4C[C@@](O)(C[C@H](O[C@@H]5C[C@@H](N)[C@@H](O)[C@@H](C)O5)c4c(O)c3C(=O)c12)C(=O)CO AESVUZLWRXEGEX-DKCAWCKPSA-N 0.000 description 1
- JXVAMODRWBNUSF-KZQKBALLSA-N (7s,9r,10r)-7-[(2r,4s,5s,6s)-5-[[(2s,4as,5as,7s,9s,9ar,10ar)-2,9-dimethyl-3-oxo-4,4a,5a,6,7,9,9a,10a-octahydrodipyrano[4,2-a:4',3'-e][1,4]dioxin-7-yl]oxy]-4-(dimethylamino)-6-methyloxan-2-yl]oxy-10-[(2s,4s,5s,6s)-4-(dimethylamino)-5-hydroxy-6-methyloxan-2 Chemical compound O([C@@H]1C2=C(O)C=3C(=O)C4=CC=CC(O)=C4C(=O)C=3C(O)=C2[C@@H](O[C@@H]2O[C@@H](C)[C@@H](O[C@@H]3O[C@@H](C)[C@H]4O[C@@H]5O[C@@H](C)C(=O)C[C@@H]5O[C@H]4C3)[C@H](C2)N(C)C)C[C@]1(O)CC)[C@H]1C[C@H](N(C)C)[C@H](O)[C@H](C)O1 JXVAMODRWBNUSF-KZQKBALLSA-N 0.000 description 1
- RCFNNLSZHVHCEK-IMHLAKCZSA-N (7s,9s)-7-(4-amino-6-methyloxan-2-yl)oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione;hydrochloride Chemical compound [Cl-].O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)C1CC([NH3+])CC(C)O1 RCFNNLSZHVHCEK-IMHLAKCZSA-N 0.000 description 1
- NOPNWHSMQOXAEI-PUCKCBAPSA-N (7s,9s)-7-[(2r,4s,5s,6s)-4-(2,3-dihydropyrrol-1-yl)-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione Chemical compound N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCC=C1 NOPNWHSMQOXAEI-PUCKCBAPSA-N 0.000 description 1
- FPVKHBSQESCIEP-UHFFFAOYSA-N (8S)-3-(2-deoxy-beta-D-erythro-pentofuranosyl)-3,6,7,8-tetrahydroimidazo[4,5-d][1,3]diazepin-8-ol Natural products C1C(O)C(CO)OC1N1C(NC=NCC2O)=C2N=C1 FPVKHBSQESCIEP-UHFFFAOYSA-N 0.000 description 1
- IEXUMDBQLIVNHZ-YOUGDJEHSA-N (8s,11r,13r,14s,17s)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one Chemical compound C1=CC(N(C)C)=CC=C1[C@@H]1C2=C3CCC(=O)C=C3CC[C@H]2[C@H](CC[C@]2(O)CCCO)[C@@]2(C)C1 IEXUMDBQLIVNHZ-YOUGDJEHSA-N 0.000 description 1
- 125000006272 (C3-C7) cycloalkyl group Chemical group 0.000 description 1
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 1
- LKJPYSCBVHEWIU-KRWDZBQOSA-N (R)-bicalutamide Chemical compound C([C@@](O)(C)C(=O)NC=1C=C(C(C#N)=CC=1)C(F)(F)F)S(=O)(=O)C1=CC=C(F)C=C1 LKJPYSCBVHEWIU-KRWDZBQOSA-N 0.000 description 1
- KWTSXDURSIMDCE-QMMMGPOBSA-N (S)-amphetamine Chemical compound C[C@H](N)CC1=CC=CC=C1 KWTSXDURSIMDCE-QMMMGPOBSA-N 0.000 description 1
- MIOPJNTWMNEORI-GMSGAONNSA-N (S)-camphorsulfonic acid Chemical compound C1C[C@@]2(CS(O)(=O)=O)C(=O)C[C@@H]1C2(C)C MIOPJNTWMNEORI-GMSGAONNSA-N 0.000 description 1
- AGNGYMCLFWQVGX-AGFFZDDWSA-N (e)-1-[(2s)-2-amino-2-carboxyethoxy]-2-diazonioethenolate Chemical compound OC(=O)[C@@H](N)CO\C([O-])=C\[N+]#N AGNGYMCLFWQVGX-AGFFZDDWSA-N 0.000 description 1
- UKAUYVFTDYCKQA-UHFFFAOYSA-N -2-Amino-4-hydroxybutanoic acid Natural products OC(=O)C(N)CCO UKAUYVFTDYCKQA-UHFFFAOYSA-N 0.000 description 1
- WBYWAXJHAXSJNI-VOTSOKGWSA-M .beta-Phenylacrylic acid Natural products [O-]C(=O)\C=C\C1=CC=CC=C1 WBYWAXJHAXSJNI-VOTSOKGWSA-M 0.000 description 1
- 125000005988 1,1-dioxo-thiomorpholinyl group Chemical group 0.000 description 1
- KEIFWROAQVVDBN-UHFFFAOYSA-N 1,2-dihydronaphthalene Chemical compound C1=CC=C2C=CCCC2=C1 KEIFWROAQVVDBN-UHFFFAOYSA-N 0.000 description 1
- FONKWHRXTPJODV-DNQXCXABSA-N 1,3-bis[2-[(8s)-8-(chloromethyl)-4-hydroxy-1-methyl-7,8-dihydro-3h-pyrrolo[3,2-e]indole-6-carbonyl]-1h-indol-5-yl]urea Chemical compound C1([C@H](CCl)CN2C(=O)C=3NC4=CC=C(C=C4C=3)NC(=O)NC=3C=C4C=C(NC4=CC=3)C(=O)N3C4=CC(O)=C5NC=C(C5=C4[C@H](CCl)C3)C)=C2C=C(O)C2=C1C(C)=CN2 FONKWHRXTPJODV-DNQXCXABSA-N 0.000 description 1
- WNXJIVFYUVYPPR-UHFFFAOYSA-N 1,3-dioxolane Chemical compound C1COCO1 WNXJIVFYUVYPPR-UHFFFAOYSA-N 0.000 description 1
- JPRPJUMQRZTTED-UHFFFAOYSA-N 1,3-dioxolanyl Chemical group [CH]1OCCO1 JPRPJUMQRZTTED-UHFFFAOYSA-N 0.000 description 1
- LTYWHYHLEKRPQC-UHFFFAOYSA-N 1-[4-(1-methyl-2-phenylpyrrolo[2,3-b]pyridin-3-yl)phenyl]cyclobutan-1-amine Chemical compound C12=CC=CN=C2N(C)C(C=2C=CC=CC=2)=C1C(C=C1)=CC=C1C1(N)CCC1 LTYWHYHLEKRPQC-UHFFFAOYSA-N 0.000 description 1
- RQAHCXDBYSJXMK-UHFFFAOYSA-N 1-[4-(2-chloro-6-phenylpyrrolo[3,2-d]pyrimidin-5-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=CN=C(Cl)N=C3C=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 RQAHCXDBYSJXMK-UHFFFAOYSA-N 0.000 description 1
- HCUBDQQWDHQDRL-UHFFFAOYSA-N 1-[4-(2-chloro-8-pyridin-2-ylpurin-9-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(Cl)=NC=C3N=C2C=2N=CC=CC=2)C=CC=1C1(N)CCC1 HCUBDQQWDHQDRL-UHFFFAOYSA-N 0.000 description 1
- JUZGVEHXXYOCPL-UHFFFAOYSA-N 1-[4-(3-phenyl-1h-pyrrolo[2,3-b]pyridin-2-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C2=C(C3=CC=CN=C3N2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 JUZGVEHXXYOCPL-UHFFFAOYSA-N 0.000 description 1
- YUZPRQPVTKNVJD-UHFFFAOYSA-N 1-[4-(5-methyl-6-phenyl-2-pyrrolidin-1-ylpyrrolo[2,3-d]pyrimidin-7-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C2(N)CCC2)C=CC=1N1C2=NC(N3CCCC3)=NC=C2C(C)=C1C1=CC=CC=C1 YUZPRQPVTKNVJD-UHFFFAOYSA-N 0.000 description 1
- AVJDUQMRSXEWIF-UHFFFAOYSA-N 1-[4-(6-phenyl-2-pyrrolidin-1-ylpyrrolo[3,2-d]pyrimidin-5-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=CN=C(N=C3C=C2C=2C=CC=CC=2)N2CCCC2)C=CC=1C1(N)CCC1 AVJDUQMRSXEWIF-UHFFFAOYSA-N 0.000 description 1
- KWLVZADASCERSE-UHFFFAOYSA-N 1-[4-(6-phenylpyrrolo[2,3-d]pyrimidin-7-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC=NC=C3C=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 KWLVZADASCERSE-UHFFFAOYSA-N 0.000 description 1
- HRWIYYSNHJLBEJ-UHFFFAOYSA-N 1-[4-(8-phenyl-2-pyrrolidin-1-ylpurin-9-yl)phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2C=CC=CC=2)N2CCCC2)C=CC=1C1(N)CCC1 HRWIYYSNHJLBEJ-UHFFFAOYSA-N 0.000 description 1
- GUKZDLFVTXPPRX-UHFFFAOYSA-N 1-[4-[2-(4-methylphenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C1=CC(C)=CC=C1C1=NC=C(N=C(C=2N(N=CC=2)C)N2C=3C=CC(=CC=3)C3(N)CCC3)C2=N1 GUKZDLFVTXPPRX-UHFFFAOYSA-N 0.000 description 1
- JQLVRUZWHCTNHO-UHFFFAOYSA-N 1-[4-[2-phenyl-8-(1h-pyrazol-4-yl)purin-9-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C2=CNN=C2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 JQLVRUZWHCTNHO-UHFFFAOYSA-N 0.000 description 1
- AQKPYTAKJCZZAK-UHFFFAOYSA-N 1-[4-[4-methoxy-2-(1h-pyrazol-4-yl)-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-7-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=2C(OC)=NC(C3=CNN=C3)=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=N1 AQKPYTAKJCZZAK-UHFFFAOYSA-N 0.000 description 1
- IIEXFGSRYKENEJ-UHFFFAOYSA-N 1-[4-[7-methyl-5-phenyl-2-(1h-pyrazol-4-yl)pyrrolo[2,3-d]pyrimidin-6-yl]phenyl]cyclobutan-1-amine Chemical compound C12=CN=C(C3=CNN=C3)N=C2N(C)C(C=2C=CC(=CC=2)C2(N)CCC2)=C1C1=CC=CC=C1 IIEXFGSRYKENEJ-UHFFFAOYSA-N 0.000 description 1
- JISPJVZPFLSICB-UHFFFAOYSA-N 1-[4-[7-phenyl-2-(1h-pyrazol-4-yl)purin-8-yl]phenyl]cyclobutan-1-amine Chemical compound C=1C=C(C=2N(C3=CN=C(N=C3N=2)C2=CNN=C2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 JISPJVZPFLSICB-UHFFFAOYSA-N 0.000 description 1
- HLJRQYTWVUJZML-UHFFFAOYSA-N 1-[4-[8-(2-methylpyrazol-3-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound CN1N=CC=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 HLJRQYTWVUJZML-UHFFFAOYSA-N 0.000 description 1
- RJUYJOJIRSHEJC-UHFFFAOYSA-N 1-[4-[8-(3-methoxypyridin-2-yl)-2-phenylpurin-9-yl]phenyl]cyclobutan-1-amine Chemical compound COC1=CC=CN=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(N)CCC2)C=C1 RJUYJOJIRSHEJC-UHFFFAOYSA-N 0.000 description 1
- CWCSQLVDSHQDCK-UHFFFAOYSA-N 1-[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-2-yl]pyrrolidin-3-ol Chemical compound C=1C=C(N2C3=NC(=NC=C3C=C2C=2N=CC=CC=2)N2CC(O)CC2)C=CC=1C1(N)CCC1 CWCSQLVDSHQDCK-UHFFFAOYSA-N 0.000 description 1
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 1
- DGHHQBMTXTWTJV-BQAIUKQQSA-N 119413-54-6 Chemical compound Cl.C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 DGHHQBMTXTWTJV-BQAIUKQQSA-N 0.000 description 1
- XWIYUCRMWCHYJR-UHFFFAOYSA-N 1h-pyrrolo[3,2-b]pyridine Chemical class C1=CC=C2NC=CC2=N1 XWIYUCRMWCHYJR-UHFFFAOYSA-N 0.000 description 1
- SRSKXJVMVSSSHB-UHFFFAOYSA-N 1h-pyrrolo[3,2-c]pyridine Chemical compound N1=CC=C2NC=CC2=C1 SRSKXJVMVSSSHB-UHFFFAOYSA-N 0.000 description 1
- CAFROQYMUICGNO-UHFFFAOYSA-N 2,2,2-trifluoroethyl formate Chemical compound FC(F)(F)COC=O CAFROQYMUICGNO-UHFFFAOYSA-N 0.000 description 1
- BTOTXLJHDSNXMW-POYBYMJQSA-N 2,3-dideoxyuridine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(=O)NC(=O)C=C1 BTOTXLJHDSNXMW-POYBYMJQSA-N 0.000 description 1
- KEQTWHPMSVAFDA-UHFFFAOYSA-N 2,3-dihydro-1h-pyrazole Chemical group C1NNC=C1 KEQTWHPMSVAFDA-UHFFFAOYSA-N 0.000 description 1
- HCSBTDBGTNZOAB-UHFFFAOYSA-N 2,3-dinitrobenzoic acid Chemical class OC(=O)C1=CC=CC([N+]([O-])=O)=C1[N+]([O-])=O HCSBTDBGTNZOAB-UHFFFAOYSA-N 0.000 description 1
- MFBKQYDNIUTLRN-UHFFFAOYSA-N 2,4,6-trichloro-5-iodopyrimidine Chemical compound ClC1=NC(Cl)=C(I)C(Cl)=N1 MFBKQYDNIUTLRN-UHFFFAOYSA-N 0.000 description 1
- 150000005738 2,4-dichloro-5-bromopyrimidines Chemical class 0.000 description 1
- 150000005709 2,4-dichloro-5-iodopyrimidines Chemical class 0.000 description 1
- INUSQTPGSHFGHM-UHFFFAOYSA-N 2,4-dichloro-5-nitropyrimidine Chemical compound [O-][N+](=O)C1=CN=C(Cl)N=C1Cl INUSQTPGSHFGHM-UHFFFAOYSA-N 0.000 description 1
- NBCOZXBHPKSFSA-UHFFFAOYSA-N 2,4-dichloro-6-methyl-5-nitropyrimidine Chemical compound CC1=NC(Cl)=NC(Cl)=C1[N+]([O-])=O NBCOZXBHPKSFSA-UHFFFAOYSA-N 0.000 description 1
- BOMZMNZEXMAQQW-UHFFFAOYSA-N 2,5,11-trimethyl-6h-pyrido[4,3-b]carbazol-2-ium-9-ol;acetate Chemical compound CC([O-])=O.C[N+]1=CC=C2C(C)=C(NC=3C4=CC(O)=CC=3)C4=C(C)C2=C1 BOMZMNZEXMAQQW-UHFFFAOYSA-N 0.000 description 1
- FFMBYMANYCDCMK-UHFFFAOYSA-N 2,5-dihydro-1h-imidazole Chemical group C1NCN=C1 FFMBYMANYCDCMK-UHFFFAOYSA-N 0.000 description 1
- XDLJIIILKATPAQ-UHFFFAOYSA-N 2,8-dichloro-7h-purine Chemical compound ClC1=NC=C2NC(Cl)=NC2=N1 XDLJIIILKATPAQ-UHFFFAOYSA-N 0.000 description 1
- TXHAHOVNFDVCCC-UHFFFAOYSA-N 2-(tert-butylazaniumyl)acetate Chemical compound CC(C)(C)NCC(O)=O TXHAHOVNFDVCCC-UHFFFAOYSA-N 0.000 description 1
- UHJQWEJGEJILKA-UHFFFAOYSA-N 2-[(4-iodo-2-methylimidazol-1-yl)methoxy]ethyl-trimethylsilane Chemical compound CC1=NC(I)=CN1COCC[Si](C)(C)C UHJQWEJGEJILKA-UHFFFAOYSA-N 0.000 description 1
- QCXJFISCRQIYID-IAEPZHFASA-N 2-amino-1-n-[(3s,6s,7r,10s,16s)-3-[(2s)-butan-2-yl]-7,11,14-trimethyl-2,5,9,12,15-pentaoxo-10-propan-2-yl-8-oxa-1,4,11,14-tetrazabicyclo[14.3.0]nonadecan-6-yl]-4,6-dimethyl-3-oxo-9-n-[(3s,6s,7r,10s,16s)-7,11,14-trimethyl-2,5,9,12,15-pentaoxo-3,10-di(propa Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N=C2C(C(=O)N[C@@H]3C(=O)N[C@H](C(N4CCC[C@H]4C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]3C)=O)[C@@H](C)CC)=C(N)C(=O)C(C)=C2O2)C2=C(C)C=C1 QCXJFISCRQIYID-IAEPZHFASA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- GRWKNBPOGBTZMN-UHFFFAOYSA-N 2-benzyl-3-phenylpropane-1,2-diamine Chemical compound C=1C=CC=CC=1CC(N)(CN)CC1=CC=CC=C1 GRWKNBPOGBTZMN-UHFFFAOYSA-N 0.000 description 1
- GOHBBINNYAWQGO-UHFFFAOYSA-N 2-bromo-3-chloropyridine Chemical class ClC1=CC=CN=C1Br GOHBBINNYAWQGO-UHFFFAOYSA-N 0.000 description 1
- SBRCZAJEFZISCV-UHFFFAOYSA-N 2-bromo-6-(2-fluorophenyl)-3-(2-phenylethynyl)pyridine Chemical compound FC1=CC=CC=C1C(N=C1Br)=CC=C1C#CC1=CC=CC=C1 SBRCZAJEFZISCV-UHFFFAOYSA-N 0.000 description 1
- KMODISUYWZPVGV-UHFFFAOYSA-N 2-bromo-6-methoxypyridine Chemical compound COC1=CC=CC(Br)=N1 KMODISUYWZPVGV-UHFFFAOYSA-N 0.000 description 1
- SIRRAQQGGCNHQB-UHFFFAOYSA-N 2-chloro-1h-imidazo[4,5-b]pyridine Chemical compound C1=CC=C2NC(Cl)=NC2=N1 SIRRAQQGGCNHQB-UHFFFAOYSA-N 0.000 description 1
- UUOLETYDNTVQDY-UHFFFAOYSA-N 2-chloro-3-nitropyridine Chemical class [O-][N+](=O)C1=CC=CN=C1Cl UUOLETYDNTVQDY-UHFFFAOYSA-N 0.000 description 1
- UIEVSGOVFXWCIK-UHFFFAOYSA-N 2-chloro-6-methyl-3-nitropyridine Chemical compound CC1=CC=C([N+]([O-])=O)C(Cl)=N1 UIEVSGOVFXWCIK-UHFFFAOYSA-N 0.000 description 1
- VNBAOSVONFJBKP-UHFFFAOYSA-N 2-chloro-n,n-bis(2-chloroethyl)propan-1-amine;hydrochloride Chemical compound Cl.CC(Cl)CN(CCCl)CCCl VNBAOSVONFJBKP-UHFFFAOYSA-N 0.000 description 1
- IKCLCGXPQILATA-UHFFFAOYSA-N 2-chlorobenzoic acid Chemical class OC(=O)C1=CC=CC=C1Cl IKCLCGXPQILATA-UHFFFAOYSA-N 0.000 description 1
- FNRMMDCDHWCQTH-UHFFFAOYSA-N 2-chloropyridine;3-chloropyridine;4-chloropyridine Chemical compound ClC1=CC=NC=C1.ClC1=CC=CN=C1.ClC1=CC=CC=N1 FNRMMDCDHWCQTH-UHFFFAOYSA-N 0.000 description 1
- NHUBNHMFXQNNMV-UHFFFAOYSA-N 2-ethynylpyridine Chemical compound C#CC1=CC=CC=N1 NHUBNHMFXQNNMV-UHFFFAOYSA-N 0.000 description 1
- CSDSSGBPEUDDEE-UHFFFAOYSA-N 2-formylpyridine Chemical compound O=CC1=CC=CC=N1 CSDSSGBPEUDDEE-UHFFFAOYSA-N 0.000 description 1
- FWWQNJOBQVPUNE-UHFFFAOYSA-N 2-methoxy-3-(2-phenylethynyl)-1h-pyridin-2-amine Chemical compound COC1(N)NC=CC=C1C#CC1=CC=CC=C1 FWWQNJOBQVPUNE-UHFFFAOYSA-N 0.000 description 1
- RAJRANFZSWDUJZ-UHFFFAOYSA-N 2-methylpyrazole-3-carbaldehyde Chemical compound CN1N=CC=C1C=O RAJRANFZSWDUJZ-UHFFFAOYSA-N 0.000 description 1
- MNURQUMUZCBVPI-UHFFFAOYSA-N 2-nitropyrazine Chemical compound [O-][N+](=O)C1=CN=CC=N1 MNURQUMUZCBVPI-UHFFFAOYSA-N 0.000 description 1
- HLTDBMHJSBSAOM-UHFFFAOYSA-N 2-nitropyridine Chemical class [O-][N+](=O)C1=CC=CC=N1 HLTDBMHJSBSAOM-UHFFFAOYSA-N 0.000 description 1
- 125000004105 2-pyridyl group Chemical group N1=C([*])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- RSEBUVRVKCANEP-UHFFFAOYSA-N 2-pyrroline Chemical group C1CC=CN1 RSEBUVRVKCANEP-UHFFFAOYSA-N 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- 125000001698 2H-pyranyl group Chemical group O1C(C=CC=C1)* 0.000 description 1
- NDMPLJNOPCLANR-UHFFFAOYSA-N 3,4-dihydroxy-15-(4-hydroxy-18-methoxycarbonyl-5,18-seco-ibogamin-18-yl)-16-methoxy-1-methyl-6,7-didehydro-aspidospermidine-3-carboxylic acid methyl ester Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 NDMPLJNOPCLANR-UHFFFAOYSA-N 0.000 description 1
- PWMYMKOUNYTVQN-UHFFFAOYSA-N 3-(8,8-diethyl-2-aza-8-germaspiro[4.5]decan-2-yl)-n,n-dimethylpropan-1-amine Chemical compound C1C[Ge](CC)(CC)CCC11CN(CCCN(C)C)CC1 PWMYMKOUNYTVQN-UHFFFAOYSA-N 0.000 description 1
- BRMWTNUJHUMWMS-UHFFFAOYSA-N 3-Methylhistidine Natural products CN1C=NC(CC(N)C(O)=O)=C1 BRMWTNUJHUMWMS-UHFFFAOYSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- SQXBYRRNGPVSAU-UHFFFAOYSA-N 3-chloro-4-iodopyridine Chemical class ClC1=CN=CC=C1I SQXBYRRNGPVSAU-UHFFFAOYSA-N 0.000 description 1
- NHQDETIJWKXCTC-UHFFFAOYSA-N 3-chloroperbenzoic acid Chemical compound OOC(=O)C1=CC=CC(Cl)=C1 NHQDETIJWKXCTC-UHFFFAOYSA-N 0.000 description 1
- BXRLWGXPSRYJDZ-UHFFFAOYSA-N 3-cyanoalanine Chemical compound OC(=O)C(N)CC#N BXRLWGXPSRYJDZ-UHFFFAOYSA-N 0.000 description 1
- JVQIKJMSUIMUDI-UHFFFAOYSA-N 3-pyrroline Chemical group C1NCC=C1 JVQIKJMSUIMUDI-UHFFFAOYSA-N 0.000 description 1
- 125000004364 3-pyrrolinyl group Chemical group [H]C1=C([H])C([H])([H])N(*)C1([H])[H] 0.000 description 1
- 125000001397 3-pyrrolyl group Chemical group [H]N1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-VTZDEGQISA-N 0.000 description 1
- MCGBIXXDQFWVDW-UHFFFAOYSA-N 4,5-dihydro-1h-pyrazole Chemical group C1CC=NN1 MCGBIXXDQFWVDW-UHFFFAOYSA-N 0.000 description 1
- 150000005743 4,6-dichloro-5-iodopyrimidines Chemical class 0.000 description 1
- CLPFFLWZZBQMAO-UHFFFAOYSA-N 4-(5,6,7,8-tetrahydroimidazo[1,5-a]pyridin-5-yl)benzonitrile Chemical compound C1=CC(C#N)=CC=C1C1N2C=NC=C2CCC1 CLPFFLWZZBQMAO-UHFFFAOYSA-N 0.000 description 1
- DODQJNMQWMSYGS-QPLCGJKRSA-N 4-[(z)-1-[4-[2-(dimethylamino)ethoxy]phenyl]-1-phenylbut-1-en-2-yl]phenol Chemical compound C=1C=C(O)C=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 DODQJNMQWMSYGS-QPLCGJKRSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- GAMYYCRTACQSBR-UHFFFAOYSA-N 4-azabenzimidazole Chemical compound C1=CC=C2NC=NC2=N1 GAMYYCRTACQSBR-UHFFFAOYSA-N 0.000 description 1
- BIWOQXBVRYUITN-UHFFFAOYSA-N 4-chloro-5-iodopyrimidine Chemical compound ClC1=NC=NC=C1I BIWOQXBVRYUITN-UHFFFAOYSA-N 0.000 description 1
- PVMNPAUTCMBOMO-UHFFFAOYSA-N 4-chloropyridine Chemical compound ClC1=CC=NC=C1 PVMNPAUTCMBOMO-UHFFFAOYSA-N 0.000 description 1
- SJZRECIVHVDYJC-UHFFFAOYSA-N 4-hydroxybutyric acid Chemical class OCCCC(O)=O SJZRECIVHVDYJC-UHFFFAOYSA-N 0.000 description 1
- QWUFIMUCFQUBOT-UHFFFAOYSA-N 4-methoxypyridine-3-carbaldehyde Chemical compound COC1=CC=NC=C1C=O QWUFIMUCFQUBOT-UHFFFAOYSA-N 0.000 description 1
- 125000000339 4-pyridyl group Chemical group N1=C([H])C([H])=C([*])C([H])=C1[H] 0.000 description 1
- KDDQRKBRJSGMQE-UHFFFAOYSA-N 4-thiazolyl Chemical group [C]1=CSC=N1 KDDQRKBRJSGMQE-UHFFFAOYSA-N 0.000 description 1
- 125000001826 4H-pyranyl group Chemical group O1C(=CCC=C1)* 0.000 description 1
- IDPUKCWIGUEADI-UHFFFAOYSA-N 5-[bis(2-chloroethyl)amino]uracil Chemical compound ClCCN(CCCl)C1=CNC(=O)NC1=O IDPUKCWIGUEADI-UHFFFAOYSA-N 0.000 description 1
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-N 0.000 description 1
- 125000004938 5-pyridyl group Chemical group N1=CC=CC(=C1)* 0.000 description 1
- CWDWFSXUQODZGW-UHFFFAOYSA-N 5-thiazolyl Chemical group [C]1=CN=CS1 CWDWFSXUQODZGW-UHFFFAOYSA-N 0.000 description 1
- WYXSYVWAUAUWLD-SHUUEZRQSA-N 6-azauridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=N1 WYXSYVWAUAUWLD-SHUUEZRQSA-N 0.000 description 1
- 229960005538 6-diazo-5-oxo-L-norleucine Drugs 0.000 description 1
- YCWQAMGASJSUIP-YFKPBYRVSA-N 6-diazo-5-oxo-L-norleucine Chemical compound OC(=O)[C@@H](N)CCC(=O)C=[N+]=[N-] YCWQAMGASJSUIP-YFKPBYRVSA-N 0.000 description 1
- HOSGXJWQVBHGLT-UHFFFAOYSA-N 6-hydroxy-3,4-dihydro-1h-quinolin-2-one Chemical group N1C(=O)CCC2=CC(O)=CC=C21 HOSGXJWQVBHGLT-UHFFFAOYSA-N 0.000 description 1
- 125000004939 6-pyridyl group Chemical group N1=CC=CC=C1* 0.000 description 1
- VVIAGPKUTFNRDU-UHFFFAOYSA-N 6S-folinic acid Natural products C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-UHFFFAOYSA-N 0.000 description 1
- BGLBQHYLQLQUDE-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-n,n-dimethyl-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-4-amine Chemical compound C=1C=2C(N(C)C)=NC=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=N1 BGLBQHYLQLQUDE-UHFFFAOYSA-N 0.000 description 1
- NSKOLWASNGJVQU-UHFFFAOYSA-N 7-[4-(1-aminocyclobutyl)phenyl]-n-methyl-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-4-amine Chemical compound C=1C=2C(NC)=NC=NC=2N(C=2C=CC(=CC=2)C2(N)CCC2)C=1C1=CC=CC=N1 NSKOLWASNGJVQU-UHFFFAOYSA-N 0.000 description 1
- ZGXJTSGNIOSYLO-UHFFFAOYSA-N 88755TAZ87 Chemical compound NCC(=O)CCC(O)=O ZGXJTSGNIOSYLO-UHFFFAOYSA-N 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- OSRJUQZQGGNIEQ-UHFFFAOYSA-N 9-[4-(1-aminocyclobutyl)phenyl]-8-phenyl-3h-purin-6-one Chemical compound C=1C=C(N2C3=C(C(N=CN3)=O)N=C2C=2C=CC=CC=2)C=CC=1C1(N)CCC1 OSRJUQZQGGNIEQ-UHFFFAOYSA-N 0.000 description 1
- HDZZVAMISRMYHH-UHFFFAOYSA-N 9beta-Ribofuranosyl-7-deazaadenin Natural products C1=CC=2C(N)=NC=NC=2N1C1OC(CO)C(O)C1O HDZZVAMISRMYHH-UHFFFAOYSA-N 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 206010001233 Adenoma benign Diseases 0.000 description 1
- 230000007730 Akt signaling Effects 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- CEIZFXOZIQNICU-UHFFFAOYSA-N Alternaria alternata Crofton-weed toxin Natural products CCC(C)C1NC(=O)C(C(C)=O)=C1O CEIZFXOZIQNICU-UHFFFAOYSA-N 0.000 description 1
- 239000005995 Aluminium silicate Substances 0.000 description 1
- 208000024827 Alzheimer disease Diseases 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 208000003120 Angiofibroma Diseases 0.000 description 1
- 108090000644 Angiozyme Proteins 0.000 description 1
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 1
- 102000014654 Aromatase Human genes 0.000 description 1
- 108010078554 Aromatase Proteins 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 206010003571 Astrocytoma Diseases 0.000 description 1
- 201000001320 Atherosclerosis Diseases 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- NOWKCMXCCJGMRR-UHFFFAOYSA-N Aziridine Chemical class C1CN1 NOWKCMXCCJGMRR-UHFFFAOYSA-N 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 201000007815 Bannayan-Riley-Ruvalcaba syndrome Diseases 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- VGGGPCQERPFHOB-MCIONIFRSA-N Bestatin Chemical compound CC(C)C[C@H](C(O)=O)NC(=O)[C@@H](O)[C@H](N)CC1=CC=CC=C1 VGGGPCQERPFHOB-MCIONIFRSA-N 0.000 description 1
- 229940122361 Bisphosphonate Drugs 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 206010073106 Bone giant cell tumour malignant Diseases 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- MBABCNBNDNGODA-LTGLSHGVSA-N Bullatacin Natural products O=C1C(C[C@H](O)CCCCCCCCCC[C@@H](O)[C@@H]2O[C@@H]([C@@H]3O[C@H]([C@@H](O)CCCCCCCCCC)CC3)CC2)=C[C@H](C)O1 MBABCNBNDNGODA-LTGLSHGVSA-N 0.000 description 1
- KGGVWMAPBXIMEM-ZRTAFWODSA-N Bullatacinone Chemical compound O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@@H]1[C@@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@H]2OC(=O)[C@H](CC(C)=O)C2)CC1 KGGVWMAPBXIMEM-ZRTAFWODSA-N 0.000 description 1
- KGGVWMAPBXIMEM-JQFCFGFHSA-N Bullatacinone Natural products O=C(C[C@H]1C(=O)O[C@H](CCCCCCCCCC[C@H](O)[C@@H]2O[C@@H]([C@@H]3O[C@@H]([C@@H](O)CCCCCCCCCC)CC3)CC2)C1)C KGGVWMAPBXIMEM-JQFCFGFHSA-N 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Natural products CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 description 1
- 125000001313 C5-C10 heteroaryl group Chemical group 0.000 description 1
- 101150041968 CDC13 gene Proteins 0.000 description 1
- 101150006084 CHKB gene Proteins 0.000 description 1
- GAWIXWVDTYZWAW-UHFFFAOYSA-N C[CH]O Chemical group C[CH]O GAWIXWVDTYZWAW-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- SHHKQEUPHAENFK-UHFFFAOYSA-N Carboquone Chemical compound O=C1C(C)=C(N2CC2)C(=O)C(C(COC(N)=O)OC)=C1N1CC1 SHHKQEUPHAENFK-UHFFFAOYSA-N 0.000 description 1
- 208000009458 Carcinoma in Situ Diseases 0.000 description 1
- AOCCBINRVIKJHY-UHFFFAOYSA-N Carmofur Chemical compound CCCCCCNC(=O)N1C=C(F)C(=O)NC1=O AOCCBINRVIKJHY-UHFFFAOYSA-N 0.000 description 1
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 206010008263 Cervical dysplasia Diseases 0.000 description 1
- JWBOIMRXGHLCPP-UHFFFAOYSA-N Chloditan Chemical compound C=1C=CC=C(Cl)C=1C(C(Cl)Cl)C1=CC=C(Cl)C=C1 JWBOIMRXGHLCPP-UHFFFAOYSA-N 0.000 description 1
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 1
- XCDXSSFOJZZGQC-UHFFFAOYSA-N Chlornaphazine Chemical compound C1=CC=CC2=CC(N(CCCl)CCCl)=CC=C21 XCDXSSFOJZZGQC-UHFFFAOYSA-N 0.000 description 1
- RENMDAKOXSCIGH-UHFFFAOYSA-N Chloroacetonitrile Chemical compound ClCC#N RENMDAKOXSCIGH-UHFFFAOYSA-N 0.000 description 1
- MKQWTWSXVILIKJ-LXGUWJNJSA-N Chlorozotocin Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](C=O)NC(=O)N(N=O)CCCl MKQWTWSXVILIKJ-LXGUWJNJSA-N 0.000 description 1
- 201000005262 Chondroma Diseases 0.000 description 1
- 208000005243 Chondrosarcoma Diseases 0.000 description 1
- 201000009047 Chordoma Diseases 0.000 description 1
- 208000006332 Choriocarcinoma Diseases 0.000 description 1
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 1
- WBYWAXJHAXSJNI-SREVYHEPSA-N Cinnamic acid Chemical compound OC(=O)\C=C/C1=CC=CC=C1 WBYWAXJHAXSJNI-SREVYHEPSA-N 0.000 description 1
- 206010048832 Colon adenoma Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 229910021590 Copper(II) bromide Inorganic materials 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 208000012609 Cowden disease Diseases 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 229930188224 Cryptophycin Natural products 0.000 description 1
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- WEAHRLBPCANXCN-UHFFFAOYSA-N Daunomycin Natural products CCC1(O)CC(OC2CC(N)C(O)C(C)O2)c3cc4C(=O)c5c(OC)cccc5C(=O)c4c(O)c3C1 WEAHRLBPCANXCN-UHFFFAOYSA-N 0.000 description 1
- NNJPGOLRFBJNIW-UHFFFAOYSA-N Demecolcine Natural products C1=C(OC)C(=O)C=C2C(NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-UHFFFAOYSA-N 0.000 description 1
- 108010002156 Depsipeptides Proteins 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 1
- 229920001353 Dextrin Polymers 0.000 description 1
- 239000004375 Dextrin Substances 0.000 description 1
- 206010012689 Diabetic retinopathy Diseases 0.000 description 1
- AUGQEEXBDZWUJY-ZLJUKNTDSA-N Diacetoxyscirpenol Chemical compound C([C@]12[C@]3(C)[C@H](OC(C)=O)[C@@H](O)[C@H]1O[C@@H]1C=C(C)CC[C@@]13COC(=O)C)O2 AUGQEEXBDZWUJY-ZLJUKNTDSA-N 0.000 description 1
- AUGQEEXBDZWUJY-UHFFFAOYSA-N Diacetoxyscirpenol Natural products CC(=O)OCC12CCC(C)=CC1OC1C(O)C(OC(C)=O)C2(C)C11CO1 AUGQEEXBDZWUJY-UHFFFAOYSA-N 0.000 description 1
- BWLUMTFWVZZZND-UHFFFAOYSA-N Dibenzylamine Chemical compound C=1C=CC=CC=1CNCC1=CC=CC=C1 BWLUMTFWVZZZND-UHFFFAOYSA-N 0.000 description 1
- ZQZFYGIXNQKOAV-OCEACIFDSA-N Droloxifene Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=C(O)C=CC=1)\C1=CC=C(OCCN(C)C)C=C1 ZQZFYGIXNQKOAV-OCEACIFDSA-N 0.000 description 1
- 229930193152 Dynemicin Natural products 0.000 description 1
- 208000007033 Dysgerminoma Diseases 0.000 description 1
- 208000000471 Dysplastic Nevus Syndrome Diseases 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 201000009051 Embryonal Carcinoma Diseases 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 201000009273 Endometriosis Diseases 0.000 description 1
- AFMYMMXSQGUCBK-UHFFFAOYSA-N Endynamicin A Natural products C1#CC=CC#CC2NC(C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C3)=C3C34OC32C(C)C(C(O)=O)=C(OC)C41 AFMYMMXSQGUCBK-UHFFFAOYSA-N 0.000 description 1
- SAMRUMKYXPVKPA-VFKOLLTISA-N Enocitabine Chemical compound O=C1N=C(NC(=O)CCCCCCCCCCCCCCCCCCCCC)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 SAMRUMKYXPVKPA-VFKOLLTISA-N 0.000 description 1
- 206010014967 Ependymoma Diseases 0.000 description 1
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 description 1
- OBMLHUPNRURLOK-XGRAFVIBSA-N Epitiostanol Chemical compound C1[C@@H]2S[C@@H]2C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@H]21 OBMLHUPNRURLOK-XGRAFVIBSA-N 0.000 description 1
- 229930189413 Esperamicin Natural products 0.000 description 1
- JOYRKODLDBILNP-UHFFFAOYSA-N Ethyl urethane Chemical compound CCOC(N)=O JOYRKODLDBILNP-UHFFFAOYSA-N 0.000 description 1
- 208000006168 Ewing Sarcoma Diseases 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 208000007659 Fibroadenoma Diseases 0.000 description 1
- 206010053717 Fibrous histiocytoma Diseases 0.000 description 1
- KRHYYFGTRYWZRS-UHFFFAOYSA-M Fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 description 1
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- 102000001267 GSK3 Human genes 0.000 description 1
- 108060006662 GSK3 Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 208000000527 Germinoma Diseases 0.000 description 1
- 208000007569 Giant Cell Tumors Diseases 0.000 description 1
- 201000005409 Gliomatosis cerebri Diseases 0.000 description 1
- 206010018404 Glucagonoma Diseases 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 244000068988 Glycine max Species 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 108010069236 Goserelin Proteins 0.000 description 1
- BLCLNMBMMGCOAS-URPVMXJPSA-N Goserelin Chemical compound C([C@@H](C(=O)N[C@H](COC(C)(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NNC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 BLCLNMBMMGCOAS-URPVMXJPSA-N 0.000 description 1
- 206010018691 Granuloma Diseases 0.000 description 1
- 229910004373 HOAc Inorganic materials 0.000 description 1
- 206010019629 Hepatic adenoma Diseases 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 1
- 101000777277 Homo sapiens Serine/threonine-protein kinase Chk2 Proteins 0.000 description 1
- LCWXJXMHJVIJFK-UHFFFAOYSA-N Hydroxylysine Natural products NCC(O)CC(N)CC(O)=O LCWXJXMHJVIJFK-UHFFFAOYSA-N 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- MPBVHIBUJCELCL-UHFFFAOYSA-N Ibandronate Chemical compound CCCCCN(C)CCC(O)(P(O)(O)=O)P(O)(O)=O MPBVHIBUJCELCL-UHFFFAOYSA-N 0.000 description 1
- XDXDZDZNSLXDNA-TZNDIEGXSA-N Idarubicin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XDXDZDZNSLXDNA-TZNDIEGXSA-N 0.000 description 1
- XDXDZDZNSLXDNA-UHFFFAOYSA-N Idarubicin Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XDXDZDZNSLXDNA-UHFFFAOYSA-N 0.000 description 1
- WRYCSMQKUKOKBP-UHFFFAOYSA-N Imidazolidine Chemical group C1CNCN1 WRYCSMQKUKOKBP-UHFFFAOYSA-N 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 208000005045 Interdigitating dendritic cell sarcoma Diseases 0.000 description 1
- 238000012695 Interfacial polymerization Methods 0.000 description 1
- 208000007766 Kaposi sarcoma Diseases 0.000 description 1
- 208000002260 Keloid Diseases 0.000 description 1
- SNDPXSYFESPGGJ-BYPYZUCNSA-N L-2-aminopentanoic acid Chemical compound CCC[C@H](N)C(O)=O SNDPXSYFESPGGJ-BYPYZUCNSA-N 0.000 description 1
- 150000008575 L-amino acids Chemical class 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- FFFHZYDWPBMWHY-VKHMYHEASA-N L-homocysteine Chemical compound OC(=O)[C@@H](N)CCS FFFHZYDWPBMWHY-VKHMYHEASA-N 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical compound OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- UCUNFLYVYCGDHP-BYPYZUCNSA-N L-methionine sulfone Chemical compound CS(=O)(=O)CC[C@H](N)C(O)=O UCUNFLYVYCGDHP-BYPYZUCNSA-N 0.000 description 1
- SNDPXSYFESPGGJ-UHFFFAOYSA-N L-norVal-OH Natural products CCCC(N)C(O)=O SNDPXSYFESPGGJ-UHFFFAOYSA-N 0.000 description 1
- DGYHPLMPMRKMPD-UHFFFAOYSA-N L-propargyl glycine Natural products OC(=O)C(N)CC#C DGYHPLMPMRKMPD-UHFFFAOYSA-N 0.000 description 1
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 1
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 1
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 1
- 239000005511 L01XE05 - Sorafenib Substances 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 229920001491 Lentinan Polymers 0.000 description 1
- 208000022010 Lhermitte-Duclos disease Diseases 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 208000002404 Liver Cell Adenoma Diseases 0.000 description 1
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 1
- 208000019693 Lung disease Diseases 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 239000004907 Macro-emulsion Substances 0.000 description 1
- 208000006644 Malignant Fibrous Histiocytoma Diseases 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- VJRAUFKOOPNFIQ-UHFFFAOYSA-N Marcellomycin Natural products C12=C(O)C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C=C2C(C(=O)OC)C(CC)(O)CC1OC(OC1C)CC(N(C)C)C1OC(OC1C)CC(O)C1OC1CC(O)C(O)C(C)O1 VJRAUFKOOPNFIQ-UHFFFAOYSA-N 0.000 description 1
- 229930126263 Maytansine Natural products 0.000 description 1
- 238000003820 Medium-pressure liquid chromatography Methods 0.000 description 1
- 208000000172 Medulloblastoma Diseases 0.000 description 1
- IVDYZAAPOLNZKG-KWHRADDSSA-N Mepitiostane Chemical compound O([C@@H]1[C@]2(CC[C@@H]3[C@@]4(C)C[C@H]5S[C@H]5C[C@@H]4CC[C@H]3[C@@H]2CC1)C)C1(OC)CCCC1 IVDYZAAPOLNZKG-KWHRADDSSA-N 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- VFKZTMPDYBFSTM-KVTDHHQDSA-N Mitobronitol Chemical compound BrC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CBr VFKZTMPDYBFSTM-KVTDHHQDSA-N 0.000 description 1
- 102100024193 Mitogen-activated protein kinase 1 Human genes 0.000 description 1
- 101710166115 Mitogen-activated protein kinase 2 Proteins 0.000 description 1
- 101710166076 Mitogen-activated protein kinase 5 Proteins 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- HRHKSTOGXBBQCB-UHFFFAOYSA-N Mitomycin E Natural products O=C1C(N)=C(C)C(=O)C2=C1C(COC(N)=O)C1(OC)C3N(C)C3CN12 HRHKSTOGXBBQCB-UHFFFAOYSA-N 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 1
- 208000014767 Myeloproliferative disease Diseases 0.000 description 1
- HSHXDCVZWHOWCS-UHFFFAOYSA-N N'-hexadecylthiophene-2-carbohydrazide Chemical compound CCCCCCCCCCCCCCCCNNC(=O)c1cccs1 HSHXDCVZWHOWCS-UHFFFAOYSA-N 0.000 description 1
- JDHILDINMRGULE-LURJTMIESA-N N(pros)-methyl-L-histidine Chemical compound CN1C=NC=C1C[C@H](N)C(O)=O JDHILDINMRGULE-LURJTMIESA-N 0.000 description 1
- OVBPIULPVIDEAO-UHFFFAOYSA-N N-Pteroyl-L-glutaminsaeure Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-UHFFFAOYSA-N 0.000 description 1
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N N-debenzoyl-N-(tert-butoxycarbonyl)-10-deacetyltaxol Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 1
- GDFAOVXKHJXLEI-VKHMYHEASA-N N-methyl-L-alanine Chemical compound C[NH2+][C@@H](C)C([O-])=O GDFAOVXKHJXLEI-VKHMYHEASA-N 0.000 description 1
- ANOFLIFCBIKVDD-UHFFFAOYSA-N N1C(Cl)=NC=C2N=CC=C21 Chemical class N1C(Cl)=NC=C2N=CC=C21 ANOFLIFCBIKVDD-UHFFFAOYSA-N 0.000 description 1
- LJWZSXDLNMOUIP-UHFFFAOYSA-N N1C=CN=C2N=CC=C21 Chemical class N1C=CN=C2N=CC=C21 LJWZSXDLNMOUIP-UHFFFAOYSA-N 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 201000004404 Neurofibroma Diseases 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- SYNHCENRCUAUNM-UHFFFAOYSA-N Nitrogen mustard N-oxide hydrochloride Chemical compound Cl.ClCC[N+]([O-])(C)CCCl SYNHCENRCUAUNM-UHFFFAOYSA-N 0.000 description 1
- KGTDRFCXGRULNK-UHFFFAOYSA-N Nogalamycin Natural products COC1C(OC)(C)C(OC)C(C)OC1OC1C2=C(O)C(C(=O)C3=C(O)C=C4C5(C)OC(C(C(C5O)N(C)C)O)OC4=C3C3=O)=C3C=C2C(C(=O)OC)C(C)(O)C1 KGTDRFCXGRULNK-UHFFFAOYSA-N 0.000 description 1
- 239000006057 Non-nutritive feed additive Substances 0.000 description 1
- QMGVPVSNSZLJIA-UHFFFAOYSA-N Nux Vomica Natural products C1C2C3C4N(C=5C6=CC=CC=5)C(=O)CC3OCC=C2CN2C1C46CC2 QMGVPVSNSZLJIA-UHFFFAOYSA-N 0.000 description 1
- ILUJQPXNXACGAN-UHFFFAOYSA-N O-methylsalicylic acid Chemical class COC1=CC=CC=C1C(O)=O ILUJQPXNXACGAN-UHFFFAOYSA-N 0.000 description 1
- BZQFBWGGLXLEPQ-UHFFFAOYSA-N O-phosphoryl-L-serine Natural products OC(=O)C(N)COP(O)(O)=O BZQFBWGGLXLEPQ-UHFFFAOYSA-N 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 208000022873 Ocular disease Diseases 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 201000010133 Oligodendroglioma Diseases 0.000 description 1
- 229930187135 Olivomycin Natural products 0.000 description 1
- 108010053291 Oncogene Protein v-akt Proteins 0.000 description 1
- 208000010191 Osteitis Deformans Diseases 0.000 description 1
- 208000000035 Osteochondroma Diseases 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- ZJOKWAWPAPMNIM-UHFFFAOYSA-N PD-153035 hydrochloride Chemical compound Cl.C=12C=C(OC)C(OC)=CC2=NC=NC=1NC1=CC=CC(Br)=C1 ZJOKWAWPAPMNIM-UHFFFAOYSA-N 0.000 description 1
- 239000012828 PI3K inhibitor Substances 0.000 description 1
- 208000027067 Paget disease of bone Diseases 0.000 description 1
- VREZDOWOLGNDPW-ALTGWBOUSA-N Pancratistatin Chemical compound C1=C2[C@H]3[C@@H](O)[C@H](O)[C@@H](O)[C@@H](O)[C@@H]3NC(=O)C2=C(O)C2=C1OCO2 VREZDOWOLGNDPW-ALTGWBOUSA-N 0.000 description 1
- VREZDOWOLGNDPW-MYVCAWNPSA-N Pancratistatin Natural products O=C1N[C@H]2[C@H](O)[C@H](O)[C@H](O)[C@H](O)[C@@H]2c2c1c(O)c1OCOc1c2 VREZDOWOLGNDPW-MYVCAWNPSA-N 0.000 description 1
- 208000030852 Parasitic disease Diseases 0.000 description 1
- 208000018737 Parkinson disease Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 108010057150 Peplomycin Proteins 0.000 description 1
- PCNDJXKNXGMECE-UHFFFAOYSA-N Phenazine Natural products C1=CC=CC2=NC3=CC=CC=C3N=C21 PCNDJXKNXGMECE-UHFFFAOYSA-N 0.000 description 1
- IGVPBCZDHMIOJH-UHFFFAOYSA-N Phenyl butyrate Chemical class CCCC(=O)OC1=CC=CC=C1 IGVPBCZDHMIOJH-UHFFFAOYSA-N 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 208000007641 Pinealoma Diseases 0.000 description 1
- KMSKQZKKOZQFFG-HSUXVGOQSA-N Pirarubicin Chemical compound O([C@H]1[C@@H](N)C[C@@H](O[C@H]1C)O[C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1CCCCO1 KMSKQZKKOZQFFG-HSUXVGOQSA-N 0.000 description 1
- 102000010995 Pleckstrin homology domains Human genes 0.000 description 1
- 108050001185 Pleckstrin homology domains Proteins 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 229920002556 Polyethylene Glycol 300 Polymers 0.000 description 1
- 229920002565 Polyethylene Glycol 400 Polymers 0.000 description 1
- 239000004743 Polypropylene Substances 0.000 description 1
- 229910052777 Praseodymium Inorganic materials 0.000 description 1
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 1
- HFVNWDWLWUCIHC-GUPDPFMOSA-N Prednimustine Chemical compound O=C([C@@]1(O)CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)[C@@H](O)C[C@@]21C)COC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 HFVNWDWLWUCIHC-GUPDPFMOSA-N 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- XBDQKXXYIPTUBI-UHFFFAOYSA-N Propionic acid Chemical class CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 102100024924 Protein kinase C alpha type Human genes 0.000 description 1
- 101710109947 Protein kinase C alpha type Proteins 0.000 description 1
- 102000052575 Proto-Oncogene Human genes 0.000 description 1
- 108700020978 Proto-Oncogene Proteins 0.000 description 1
- KYQCOXFCLRTKLS-UHFFFAOYSA-N Pyrazine Natural products C1=CN=CC=N1 KYQCOXFCLRTKLS-UHFFFAOYSA-N 0.000 description 1
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 1
- 201000000582 Retinoblastoma Diseases 0.000 description 1
- 206010038997 Retroviral infections Diseases 0.000 description 1
- 208000005678 Rhabdomyoma Diseases 0.000 description 1
- OWPCHSCAPHNHAV-UHFFFAOYSA-N Rhizoxin Natural products C1C(O)C2(C)OC2C=CC(C)C(OC(=O)C2)CC2CC2OC2C(=O)OC1C(C)C(OC)C(C)=CC=CC(C)=CC1=COC(C)=N1 OWPCHSCAPHNHAV-UHFFFAOYSA-N 0.000 description 1
- NSFWWJIQIKBZMJ-YKNYLIOZSA-N Roridin A Chemical compound C([C@]12[C@]3(C)[C@H]4C[C@H]1O[C@@H]1C=C(C)CC[C@@]13COC(=O)[C@@H](O)[C@H](C)CCO[C@H](\C=C\C=C/C(=O)O4)[C@H](O)C)O2 NSFWWJIQIKBZMJ-YKNYLIOZSA-N 0.000 description 1
- 108010077895 Sarcosine Proteins 0.000 description 1
- 201000010208 Seminoma Diseases 0.000 description 1
- 206010040070 Septic Shock Diseases 0.000 description 1
- 102100031075 Serine/threonine-protein kinase Chk2 Human genes 0.000 description 1
- 208000000097 Sertoli-Leydig cell tumor Diseases 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 229920000519 Sizofiran Polymers 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- LSNNMFCWUKXFEE-UHFFFAOYSA-N Sulfurous acid Chemical class OS(O)=O LSNNMFCWUKXFEE-UHFFFAOYSA-N 0.000 description 1
- BXFOFFBJRFZBQZ-QYWOHJEZSA-N T-2 toxin Chemical compound C([C@@]12[C@]3(C)[C@H](OC(C)=O)[C@@H](O)[C@H]1O[C@H]1[C@]3(COC(C)=O)C[C@@H](C(=C1)C)OC(=O)CC(C)C)O2 BXFOFFBJRFZBQZ-QYWOHJEZSA-N 0.000 description 1
- 229940126624 Tacatuzumab tetraxetan Drugs 0.000 description 1
- CGMTUJFWROPELF-UHFFFAOYSA-N Tenuazonic acid Natural products CCC(C)C1NC(=O)C(=C(C)/O)C1=O CGMTUJFWROPELF-UHFFFAOYSA-N 0.000 description 1
- FZWLAAWBMGSTSO-UHFFFAOYSA-N Thiazole Chemical group C1=CSC=N1 FZWLAAWBMGSTSO-UHFFFAOYSA-N 0.000 description 1
- 101710183280 Topoisomerase Proteins 0.000 description 1
- IWEQQRMGNVVKQW-OQKDUQJOSA-N Toremifene citrate Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 IWEQQRMGNVVKQW-OQKDUQJOSA-N 0.000 description 1
- 206010052779 Transplant rejections Diseases 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
- GCTFWCDSFPMHHS-UHFFFAOYSA-M Tributyltin chloride Chemical compound CCCC[Sn](Cl)(CCCC)CCCC GCTFWCDSFPMHHS-UHFFFAOYSA-M 0.000 description 1
- UMILHIMHKXVDGH-UHFFFAOYSA-N Triethylene glycol diglycidyl ether Chemical compound C1OC1COCCOCCOCCOCC1CO1 UMILHIMHKXVDGH-UHFFFAOYSA-N 0.000 description 1
- FYAMXEPQQLNQDM-UHFFFAOYSA-N Tris(1-aziridinyl)phosphine oxide Chemical compound C1CN1P(N1CC1)(=O)N1CC1 FYAMXEPQQLNQDM-UHFFFAOYSA-N 0.000 description 1
- 208000015778 Undifferentiated pleomorphic sarcoma Diseases 0.000 description 1
- 208000009311 VIPoma Diseases 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 206010048214 Xanthoma Diseases 0.000 description 1
- 206010048215 Xanthomatosis Diseases 0.000 description 1
- WERKSKAQRVDLDW-ANOHMWSOSA-N [(2s,3r,4r,5r)-2,3,4,5,6-pentahydroxyhexyl] (z)-octadec-9-enoate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO WERKSKAQRVDLDW-ANOHMWSOSA-N 0.000 description 1
- SPJCRMJCFSJKDE-ZWBUGVOYSA-N [(3s,8s,9s,10r,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1h-cyclopenta[a]phenanthren-3-yl] 2-[4-[bis(2-chloroethyl)amino]phenyl]acetate Chemical compound O([C@@H]1CC2=CC[C@H]3[C@@H]4CC[C@@H]([C@]4(CC[C@@H]3[C@@]2(C)CC1)C)[C@H](C)CCCC(C)C)C(=O)CC1=CC=C(N(CCCl)CCCl)C=C1 SPJCRMJCFSJKDE-ZWBUGVOYSA-N 0.000 description 1
- IFJUINDAXYAPTO-UUBSBJJBSA-N [(8r,9s,13s,14s,17s)-17-[2-[4-[4-[bis(2-chloroethyl)amino]phenyl]butanoyloxy]acetyl]oxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-3-yl] benzoate Chemical compound C([C@@H]1[C@@H](C2=CC=3)CC[C@]4([C@H]1CC[C@@H]4OC(=O)COC(=O)CCCC=1C=CC(=CC=1)N(CCCl)CCCl)C)CC2=CC=3OC(=O)C1=CC=CC=C1 IFJUINDAXYAPTO-UUBSBJJBSA-N 0.000 description 1
- IHGLINDYFMDHJG-UHFFFAOYSA-N [2-(4-methoxyphenyl)-3,4-dihydronaphthalen-1-yl]-[4-(2-pyrrolidin-1-ylethoxy)phenyl]methanone Chemical compound C1=CC(OC)=CC=C1C(CCC1=CC=CC=C11)=C1C(=O)C(C=C1)=CC=C1OCCN1CCCC1 IHGLINDYFMDHJG-UHFFFAOYSA-N 0.000 description 1
- XZSRRNFBEIOBDA-CFNBKWCHSA-N [2-[(2s,4s)-4-[(2r,4s,5s,6s)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-2,5,12-trihydroxy-7-methoxy-6,11-dioxo-3,4-dihydro-1h-tetracen-2-yl]-2-oxoethyl] 2,2-diethoxyacetate Chemical compound O([C@H]1C[C@](CC2=C(O)C=3C(=O)C4=CC=CC(OC)=C4C(=O)C=3C(O)=C21)(O)C(=O)COC(=O)C(OCC)OCC)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 XZSRRNFBEIOBDA-CFNBKWCHSA-N 0.000 description 1
- AIWRTTMUVOZGPW-HSPKUQOVSA-N abarelix Chemical compound C([C@@H](C(=O)N[C@H](CC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCCNC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N[C@H](C)C(N)=O)N(C)C(=O)[C@H](CO)NC(=O)[C@@H](CC=1C=NC=CC=1)NC(=O)[C@@H](CC=1C=CC(Cl)=CC=1)NC(=O)[C@@H](CC=1C=C2C=CC=CC2=CC=1)NC(C)=O)C1=CC=C(O)C=C1 AIWRTTMUVOZGPW-HSPKUQOVSA-N 0.000 description 1
- 108010023617 abarelix Proteins 0.000 description 1
- 229960002184 abarelix Drugs 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- 239000002250 absorbent Substances 0.000 description 1
- 230000002745 absorbent Effects 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 239000000370 acceptor Substances 0.000 description 1
- ZOZKYEHVNDEUCO-XUTVFYLZSA-N aceglatone Chemical compound O1C(=O)[C@H](OC(C)=O)[C@@H]2OC(=O)[C@@H](OC(=O)C)[C@@H]21 ZOZKYEHVNDEUCO-XUTVFYLZSA-N 0.000 description 1
- 229950002684 aceglatone Drugs 0.000 description 1
- 239000008351 acetate buffer Substances 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 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
- 150000001252 acrylic acid derivatives Chemical class 0.000 description 1
- 229930183665 actinomycin Natural products 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 125000005042 acyloxymethyl group Chemical group 0.000 description 1
- 208000002718 adenomatoid tumor Diseases 0.000 description 1
- 229950004955 adozelesin Drugs 0.000 description 1
- BYRVKDUQDLJUBX-JJCDCTGGSA-N adozelesin Chemical compound C1=CC=C2OC(C(=O)NC=3C=C4C=C(NC4=CC=3)C(=O)N3C[C@H]4C[C@]44C5=C(C(C=C43)=O)NC=C5C)=CC2=C1 BYRVKDUQDLJUBX-JJCDCTGGSA-N 0.000 description 1
- 229940009456 adriamycin Drugs 0.000 description 1
- 239000003463 adsorbent Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 206010064930 age-related macular degeneration Diseases 0.000 description 1
- IAJILQKETJEXLJ-RSJOWCBRSA-N aldehydo-D-galacturonic acid Chemical compound O=C[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)C(O)=O IAJILQKETJEXLJ-RSJOWCBRSA-N 0.000 description 1
- IAJILQKETJEXLJ-QTBDOELSSA-N aldehydo-D-glucuronic acid Chemical compound O=C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)C(O)=O IAJILQKETJEXLJ-QTBDOELSSA-N 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 150000004703 alkoxides Chemical class 0.000 description 1
- 125000005206 alkoxycarbonyloxymethyl group Chemical group 0.000 description 1
- 125000000278 alkyl amino alkyl group Chemical group 0.000 description 1
- 125000003282 alkyl amino group Chemical group 0.000 description 1
- 229940045714 alkyl sulfonate alkylating agent Drugs 0.000 description 1
- 150000008052 alkyl sulfonates Chemical class 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 125000005466 alkylenyl group Chemical group 0.000 description 1
- 238000005905 alkynylation reaction Methods 0.000 description 1
- SHGAZHPCJJPHSC-YCNIQYBTSA-N all-trans-retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 230000007815 allergy Effects 0.000 description 1
- 229940061720 alpha hydroxy acid Drugs 0.000 description 1
- 150000001280 alpha hydroxy acids Chemical class 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-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
- 229960000473 altretamine Drugs 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 229960002749 aminolevulinic acid Drugs 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- 229940025084 amphetamine Drugs 0.000 description 1
- 229960001220 amsacrine Drugs 0.000 description 1
- XCPGHVQEEXUHNC-UHFFFAOYSA-N amsacrine Chemical compound COC1=CC(NS(C)(=O)=O)=CC=C1NC1=C(C=CC=C2)C2=NC2=CC=CC=C12 XCPGHVQEEXUHNC-UHFFFAOYSA-N 0.000 description 1
- 229960002932 anastrozole Drugs 0.000 description 1
- BBDAGFIXKZCXAH-CCXZUQQUSA-N ancitabine Chemical compound N=C1C=CN2[C@@H]3O[C@H](CO)[C@@H](O)[C@@H]3OC2=N1 BBDAGFIXKZCXAH-CCXZUQQUSA-N 0.000 description 1
- 229950000242 ancitabine Drugs 0.000 description 1
- 239000003098 androgen Substances 0.000 description 1
- 229940030486 androgens Drugs 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 239000004037 angiogenesis inhibitor Substances 0.000 description 1
- 150000008064 anhydrides Chemical class 0.000 description 1
- SMWDFEZZVXVKRB-UHFFFAOYSA-N anhydrous quinoline Natural products N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 1
- 230000002280 anti-androgenic effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 239000000051 antiandrogen Substances 0.000 description 1
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 description 1
- 239000000611 antibody drug conjugate Substances 0.000 description 1
- 229940049595 antibody-drug conjugate Drugs 0.000 description 1
- 239000013059 antihormonal agent Substances 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 229940045687 antimetabolites folic acid analogs Drugs 0.000 description 1
- 229940045988 antineoplastic drug protein kinase inhibitors Drugs 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 239000000074 antisense oligonucleotide Substances 0.000 description 1
- 238000012230 antisense oligonucleotides Methods 0.000 description 1
- 229950003145 apolizumab Drugs 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000008346 aqueous phase Substances 0.000 description 1
- 150000008209 arabinosides Chemical class 0.000 description 1
- 229910052786 argon Inorganic materials 0.000 description 1
- 229940078010 arimidex Drugs 0.000 description 1
- 229940087620 aromasin Drugs 0.000 description 1
- 239000003886 aromatase inhibitor Substances 0.000 description 1
- 229940046844 aromatase inhibitors Drugs 0.000 description 1
- 125000005251 aryl acyl group Chemical group 0.000 description 1
- 229950002882 aselizumab Drugs 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 230000035578 autophosphorylation Effects 0.000 description 1
- 229960002756 azacitidine Drugs 0.000 description 1
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 1
- 229950011321 azaserine Drugs 0.000 description 1
- 125000002393 azetidinyl group Chemical group 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 229950001863 bapineuzumab Drugs 0.000 description 1
- 229910052788 barium Inorganic materials 0.000 description 1
- DSAJWYNOEDNPEQ-UHFFFAOYSA-N barium atom Chemical compound [Ba] DSAJWYNOEDNPEQ-UHFFFAOYSA-N 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 235000013871 bee wax Nutrition 0.000 description 1
- 239000012166 beeswax Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 208000001119 benign fibrous histiocytoma Diseases 0.000 description 1
- 229960000686 benzalkonium chloride Drugs 0.000 description 1
- UREZNYTWGJKWBI-UHFFFAOYSA-M benzethonium chloride Chemical compound [Cl-].C1=CC(C(C)(C)CC(C)(C)C)=CC=C1OCCOCC[N+](C)(C)CC1=CC=CC=C1 UREZNYTWGJKWBI-UHFFFAOYSA-M 0.000 description 1
- 229960001950 benzethonium chloride Drugs 0.000 description 1
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000004601 benzofurazanyl group Chemical group N1=C2C(=NO1)C(=CC=C2)* 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 150000001558 benzoic acid derivatives Chemical class 0.000 description 1
- 125000001164 benzothiazolyl group Chemical group S1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 125000004196 benzothienyl group Chemical group S1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- CADWTSSKOVRVJC-UHFFFAOYSA-N benzyl(dimethyl)azanium;chloride Chemical compound [Cl-].C[NH+](C)CC1=CC=CC=C1 CADWTSSKOVRVJC-UHFFFAOYSA-N 0.000 description 1
- WGQKYBSKWIADBV-UHFFFAOYSA-O benzylaminium Chemical compound [NH3+]CC1=CC=CC=C1 WGQKYBSKWIADBV-UHFFFAOYSA-O 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-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 1
- 229940000635 beta-alanine Drugs 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 150000005347 biaryls Chemical class 0.000 description 1
- 229960000997 bicalutamide Drugs 0.000 description 1
- GPRLTFBKWDERLU-UHFFFAOYSA-N bicyclo[2.2.2]octane Chemical group C1CC2CCC1CC2 GPRLTFBKWDERLU-UHFFFAOYSA-N 0.000 description 1
- GNTFBMAGLFYMMZ-UHFFFAOYSA-N bicyclo[3.2.2]nonane Chemical group C1CC2CCC1CCC2 GNTFBMAGLFYMMZ-UHFFFAOYSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 239000004305 biphenyl Substances 0.000 description 1
- 235000010290 biphenyl Nutrition 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-O bis(2-hydroxyethyl)azanium Chemical compound OCC[NH2+]CCO ZBCBWPMODOFKDW-UHFFFAOYSA-O 0.000 description 1
- IPWKHHSGDUIRAH-UHFFFAOYSA-N bis(pinacolato)diboron Chemical compound O1C(C)(C)C(C)(C)OB1B1OC(C)(C)C(C)(C)O1 IPWKHHSGDUIRAH-UHFFFAOYSA-N 0.000 description 1
- HUTDDBSSHVOYJR-UHFFFAOYSA-H bis[(2-oxo-1,3,2$l^{5},4$l^{2}-dioxaphosphaplumbetan-2-yl)oxy]lead Chemical compound [Pb+2].[Pb+2].[Pb+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O HUTDDBSSHVOYJR-UHFFFAOYSA-H 0.000 description 1
- 229950008548 bisantrene Drugs 0.000 description 1
- 150000004663 bisphosphonates Chemical class 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-M bisulphate group Chemical group S([O-])(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-M 0.000 description 1
- 229960005522 bivatuzumab mertansine Drugs 0.000 description 1
- 229950006844 bizelesin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical class N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 208000016738 bone Paget disease Diseases 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- ZADPBFCGQRWHPN-UHFFFAOYSA-N boronic acid Chemical compound OBO ZADPBFCGQRWHPN-UHFFFAOYSA-N 0.000 description 1
- 150000001642 boronic acid derivatives Chemical class 0.000 description 1
- 229960001467 bortezomib Drugs 0.000 description 1
- 201000009480 botryoid rhabdomyosarcoma Diseases 0.000 description 1
- 201000008274 breast adenocarcinoma Diseases 0.000 description 1
- 201000003149 breast fibroadenoma Diseases 0.000 description 1
- 150000001649 bromium compounds Chemical class 0.000 description 1
- 208000003362 bronchogenic carcinoma Diseases 0.000 description 1
- 201000002143 bronchus adenoma Diseases 0.000 description 1
- RRKTZKIUPZVBMF-IBTVXLQLSA-N brucine Chemical compound O([C@@H]1[C@H]([C@H]2C3)[C@@H]4N(C(C1)=O)C=1C=C(C(=CC=11)OC)OC)CC=C2CN2[C@@H]3[C@]41CC2 RRKTZKIUPZVBMF-IBTVXLQLSA-N 0.000 description 1
- RRKTZKIUPZVBMF-UHFFFAOYSA-N brucine Natural products C1=2C=C(OC)C(OC)=CC=2N(C(C2)=O)C3C(C4C5)C2OCC=C4CN2C5C31CC2 RRKTZKIUPZVBMF-UHFFFAOYSA-N 0.000 description 1
- 229960005520 bryostatin Drugs 0.000 description 1
- MJQUEDHRCUIRLF-TVIXENOKSA-N bryostatin 1 Chemical compound C([C@@H]1CC(/[C@@H]([C@@](C(C)(C)/C=C/2)(O)O1)OC(=O)/C=C/C=C/CCC)=C\C(=O)OC)[C@H]([C@@H](C)O)OC(=O)C[C@H](O)C[C@@H](O1)C[C@H](OC(C)=O)C(C)(C)[C@]1(O)C[C@@H]1C\C(=C\C(=O)OC)C[C@H]\2O1 MJQUEDHRCUIRLF-TVIXENOKSA-N 0.000 description 1
- MUIWQCKLQMOUAT-AKUNNTHJSA-N bryostatin 20 Natural products COC(=O)C=C1C[C@@]2(C)C[C@]3(O)O[C@](C)(C[C@@H](O)CC(=O)O[C@](C)(C[C@@]4(C)O[C@](O)(CC5=CC(=O)O[C@]45C)C(C)(C)C=C[C@@](C)(C1)O2)[C@@H](C)O)C[C@H](OC(=O)C(C)(C)C)C3(C)C MUIWQCKLQMOUAT-AKUNNTHJSA-N 0.000 description 1
- 244000309464 bull Species 0.000 description 1
- MBABCNBNDNGODA-LUVUIASKSA-N bullatacin Chemical compound O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@@H]1[C@@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@@H](O)CC=2C(O[C@@H](C)C=2)=O)CC1 MBABCNBNDNGODA-LUVUIASKSA-N 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- DQXBYHZEEUGOBF-UHFFFAOYSA-N but-3-enoic acid;ethene Chemical compound C=C.OC(=O)CC=C DQXBYHZEEUGOBF-UHFFFAOYSA-N 0.000 description 1
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 description 1
- LRHPLDYGYMQRHN-UHFFFAOYSA-N butyl alcohol Substances CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 description 1
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 108700002839 cactinomycin Proteins 0.000 description 1
- 229950009908 cactinomycin Drugs 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 229950009823 calusterone Drugs 0.000 description 1
- IVFYLRMMHVYGJH-PVPPCFLZSA-N calusterone Chemical compound C1C[C@]2(C)[C@](O)(C)CC[C@H]2[C@@H]2[C@@H](C)CC3=CC(=O)CC[C@]3(C)[C@H]21 IVFYLRMMHVYGJH-PVPPCFLZSA-N 0.000 description 1
- 229940112129 campath Drugs 0.000 description 1
- 229940088954 camptosar Drugs 0.000 description 1
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- 208000035269 cancer or benign tumor Diseases 0.000 description 1
- 229950007296 cantuzumab mertansine Drugs 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 235000013877 carbamide Nutrition 0.000 description 1
- 125000002837 carbocyclic group Chemical group 0.000 description 1
- 125000004452 carbocyclyl group Chemical group 0.000 description 1
- 150000001721 carbon Chemical group 0.000 description 1
- 150000001723 carbon free-radicals Chemical class 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 229960002115 carboquone Drugs 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- UHBYWPGGCSDKFX-UHFFFAOYSA-N carboxyglutamic acid Chemical compound OC(=O)C(N)CC(C(O)=O)C(O)=O UHBYWPGGCSDKFX-UHFFFAOYSA-N 0.000 description 1
- 125000002843 carboxylic acid group Chemical group 0.000 description 1
- 150000001735 carboxylic acids Chemical class 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- XREUEWVEMYWFFA-CSKJXFQVSA-N carminomycin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=C(O)C=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XREUEWVEMYWFFA-CSKJXFQVSA-N 0.000 description 1
- 229930188550 carminomycin Natural products 0.000 description 1
- XREUEWVEMYWFFA-UHFFFAOYSA-N carminomycin I Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=C(O)C=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XREUEWVEMYWFFA-UHFFFAOYSA-N 0.000 description 1
- 229960003261 carmofur Drugs 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 229950001725 carubicin Drugs 0.000 description 1
- BBZDXMBRAFTCAA-AREMUKBSSA-N carzelesin Chemical compound C1=2NC=C(C)C=2C([C@H](CCl)CN2C(=O)C=3NC4=CC=C(C=C4C=3)NC(=O)C3=CC4=CC=C(C=C4O3)N(CC)CC)=C2C=C1OC(=O)NC1=CC=CC=C1 BBZDXMBRAFTCAA-AREMUKBSSA-N 0.000 description 1
- 229950007509 carzelesin Drugs 0.000 description 1
- 108010047060 carzinophilin Proteins 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 229950006754 cedelizumab Drugs 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 229960003115 certolizumab pegol Drugs 0.000 description 1
- 208000019065 cervical carcinoma Diseases 0.000 description 1
- 210000003679 cervix uteri Anatomy 0.000 description 1
- 229960005395 cetuximab Drugs 0.000 description 1
- 229960000541 cetyl alcohol Drugs 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 238000011944 chemoselective reduction Methods 0.000 description 1
- 238000004296 chiral HPLC Methods 0.000 description 1
- 229960004630 chlorambucil Drugs 0.000 description 1
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 1
- 229910052801 chlorine Inorganic materials 0.000 description 1
- 229950008249 chlornaphazine Drugs 0.000 description 1
- 229960001480 chlorozotocin Drugs 0.000 description 1
- 208000006990 cholangiocarcinoma Diseases 0.000 description 1
- 201000005217 chondroblastoma Diseases 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 235000013985 cinnamic acid Nutrition 0.000 description 1
- 229930016911 cinnamic acid Natural products 0.000 description 1
- 125000000259 cinnolinyl group Chemical group N1=NC(=CC2=CC=CC=C12)* 0.000 description 1
- 238000002983 circular dichroism Methods 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 150000001860 citric acid derivatives Chemical class 0.000 description 1
- 208000009060 clear cell adenocarcinoma Diseases 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- ACSIXWWBWUQEHA-UHFFFAOYSA-N clodronic acid Chemical compound OP(O)(=O)C(Cl)(Cl)P(O)(O)=O ACSIXWWBWUQEHA-UHFFFAOYSA-N 0.000 description 1
- 229960002286 clodronic acid Drugs 0.000 description 1
- 238000005354 coacervation Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 239000003240 coconut oil Substances 0.000 description 1
- 235000019864 coconut oil Nutrition 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 238000011254 conventional chemotherapy Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 239000012084 conversion product Substances 0.000 description 1
- GBRBMTNGQBKBQE-UHFFFAOYSA-L copper;diiodide Chemical compound I[Cu]I GBRBMTNGQBKBQE-UHFFFAOYSA-L 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 108010024505 crosstide peptide Proteins 0.000 description 1
- 150000003983 crown ethers Chemical class 0.000 description 1
- PSNOPSMXOBPNNV-VVCTWANISA-N cryptophycin 1 Chemical compound C1=C(Cl)C(OC)=CC=C1C[C@@H]1C(=O)NC[C@@H](C)C(=O)O[C@@H](CC(C)C)C(=O)O[C@H]([C@H](C)[C@@H]2[C@H](O2)C=2C=CC=CC=2)C/C=C/C(=O)N1 PSNOPSMXOBPNNV-VVCTWANISA-N 0.000 description 1
- 108010089438 cryptophycin 1 Proteins 0.000 description 1
- 108010090203 cryptophycin 8 Proteins 0.000 description 1
- PSNOPSMXOBPNNV-UHFFFAOYSA-N cryptophycin-327 Natural products C1=C(Cl)C(OC)=CC=C1CC1C(=O)NCC(C)C(=O)OC(CC(C)C)C(=O)OC(C(C)C2C(O2)C=2C=CC=CC=2)CC=CC(=O)N1 PSNOPSMXOBPNNV-UHFFFAOYSA-N 0.000 description 1
- 238000002425 crystallisation Methods 0.000 description 1
- 230000008025 crystallization Effects 0.000 description 1
- 201000010305 cutaneous fibrous histiocytoma Diseases 0.000 description 1
- 208000035250 cutaneous malignant susceptibility to 1 melanoma Diseases 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- HPXRVTGHNJAIIH-UHFFFAOYSA-N cyclohexanol Chemical compound OC1CCCCC1 HPXRVTGHNJAIIH-UHFFFAOYSA-N 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 229940104302 cytosine Drugs 0.000 description 1
- 239000000824 cytostatic agent Substances 0.000 description 1
- 230000001085 cytostatic effect Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- KWGRBVOPPLSCSI-UHFFFAOYSA-N d-ephedrine Natural products CNC(C)C(O)C1=CC=CC=C1 KWGRBVOPPLSCSI-UHFFFAOYSA-N 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 229960002806 daclizumab Drugs 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000006240 deamidation Effects 0.000 description 1
- 125000005534 decanoate group Chemical class 0.000 description 1
- 238000007257 deesterification reaction Methods 0.000 description 1
- YSMODUONRAFBET-UHFFFAOYSA-N delta-DL-hydroxylysine Natural products NCC(O)CCC(N)C(O)=O YSMODUONRAFBET-UHFFFAOYSA-N 0.000 description 1
- 229960005052 demecolcine Drugs 0.000 description 1
- 238000010511 deprotection reaction Methods 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 239000007933 dermal patch Substances 0.000 description 1
- 229950003913 detorubicin Drugs 0.000 description 1
- 229910052805 deuterium Inorganic materials 0.000 description 1
- 229950006137 dexfosfoserine Drugs 0.000 description 1
- 235000019425 dextrin Nutrition 0.000 description 1
- 125000002576 diazepinyl group Chemical group N1N=C(C=CC=C1)* 0.000 description 1
- WVYXNIXAMZOZFK-UHFFFAOYSA-N diaziquone Chemical compound O=C1C(NC(=O)OCC)=C(N2CC2)C(=O)C(NC(=O)OCC)=C1N1CC1 WVYXNIXAMZOZFK-UHFFFAOYSA-N 0.000 description 1
- 229950002389 diaziquone Drugs 0.000 description 1
- 125000005057 dihydrothienyl group Chemical group S1C(CC=C1)* 0.000 description 1
- PSHRANCNVXNITH-UHFFFAOYSA-N dimethylamino acetate Chemical compound CN(C)OC(C)=O PSHRANCNVXNITH-UHFFFAOYSA-N 0.000 description 1
- 125000000532 dioxanyl group Chemical group 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 150000002016 disaccharides Chemical class 0.000 description 1
- CEALXSHFPPCRNM-UHFFFAOYSA-L disodium;carboxylato carbonate Chemical compound [Na+].[Na+].[O-]C(=O)OC([O-])=O CEALXSHFPPCRNM-UHFFFAOYSA-L 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- PXJJSXABGXMUSU-UHFFFAOYSA-N disulfur dichloride Chemical compound ClSSCl PXJJSXABGXMUSU-UHFFFAOYSA-N 0.000 description 1
- 125000005883 dithianyl group Chemical group 0.000 description 1
- 125000005411 dithiolanyl group Chemical group S1SC(CC1)* 0.000 description 1
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 1
- AMRJKAQTDDKMCE-UHFFFAOYSA-N dolastatin Chemical compound CC(C)C(N(C)C)C(=O)NC(C(C)C)C(=O)N(C)C(C(C)C)C(OC)CC(=O)N1CCCC1C(OC)C(C)C(=O)NC(C=1SC=CN=1)CC1=CC=CC=C1 AMRJKAQTDDKMCE-UHFFFAOYSA-N 0.000 description 1
- 229930188854 dolastatin Natural products 0.000 description 1
- ZWAOHEXOSAUJHY-ZIYNGMLESA-N doxifluridine Chemical compound O[C@@H]1[C@H](O)[C@@H](C)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ZWAOHEXOSAUJHY-ZIYNGMLESA-N 0.000 description 1
- 229950005454 doxifluridine Drugs 0.000 description 1
- 229950004203 droloxifene Drugs 0.000 description 1
- NOTIQUSPUUHHEH-UXOVVSIBSA-N dromostanolone propionate Chemical compound C([C@@H]1CC2)C(=O)[C@H](C)C[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H](OC(=O)CC)[C@@]2(C)CC1 NOTIQUSPUUHHEH-UXOVVSIBSA-N 0.000 description 1
- 229950004683 drostanolone propionate Drugs 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 238000009510 drug design Methods 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 230000036267 drug metabolism Effects 0.000 description 1
- 229940088679 drug related substance Drugs 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 229960005501 duocarmycin Drugs 0.000 description 1
- VQNATVDKACXKTF-XELLLNAOSA-N duocarmycin Chemical compound COC1=C(OC)C(OC)=C2NC(C(=O)N3C4=CC(=O)C5=C([C@@]64C[C@@H]6C3)C=C(N5)C(=O)OC)=CC2=C1 VQNATVDKACXKTF-XELLLNAOSA-N 0.000 description 1
- 229930184221 duocarmycin Natural products 0.000 description 1
- AFMYMMXSQGUCBK-AKMKHHNQSA-N dynemicin a Chemical compound C1#C\C=C/C#C[C@@H]2NC(C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C3)=C3[C@@]34O[C@]32[C@@H](C)C(C(O)=O)=C(OC)[C@H]41 AFMYMMXSQGUCBK-AKMKHHNQSA-N 0.000 description 1
- 229960002224 eculizumab Drugs 0.000 description 1
- FSIRXIHZBIXHKT-MHTVFEQDSA-N edatrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CC(CC)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FSIRXIHZBIXHKT-MHTVFEQDSA-N 0.000 description 1
- 229950006700 edatrexate Drugs 0.000 description 1
- 229960000284 efalizumab Drugs 0.000 description 1
- 230000000235 effect on cancer Effects 0.000 description 1
- 229960002759 eflornithine Drugs 0.000 description 1
- 229920001971 elastomer Polymers 0.000 description 1
- XOPYFXBZMVTEJF-PDACKIITSA-N eleutherobin Chemical compound C(/[C@H]1[C@H](C(=CC[C@@H]1C(C)C)C)C[C@@H]([C@@]1(C)O[C@@]2(C=C1)OC)OC(=O)\C=C\C=1N=CN(C)C=1)=C2\CO[C@@H]1OC[C@@H](O)[C@@H](O)[C@@H]1OC(C)=O XOPYFXBZMVTEJF-PDACKIITSA-N 0.000 description 1
- XOPYFXBZMVTEJF-UHFFFAOYSA-N eleutherobin Natural products C1=CC2(OC)OC1(C)C(OC(=O)C=CC=1N=CN(C)C=1)CC(C(=CCC1C(C)C)C)C1C=C2COC1OCC(O)C(O)C1OC(C)=O XOPYFXBZMVTEJF-UHFFFAOYSA-N 0.000 description 1
- 229950000549 elliptinium acetate Drugs 0.000 description 1
- 229940120655 eloxatin Drugs 0.000 description 1
- 201000009409 embryonal rhabdomyosarcoma Diseases 0.000 description 1
- 239000003974 emollient agent Substances 0.000 description 1
- 201000003914 endometrial carcinoma Diseases 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- JOZGNYDSEBIJDH-UHFFFAOYSA-N eniluracil Chemical compound O=C1NC=C(C#C)C(=O)N1 JOZGNYDSEBIJDH-UHFFFAOYSA-N 0.000 description 1
- 229950010213 eniluracil Drugs 0.000 description 1
- 229950011487 enocitabine Drugs 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 229960002179 ephedrine Drugs 0.000 description 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 1
- 229960001904 epirubicin Drugs 0.000 description 1
- 229950002973 epitiostanol Drugs 0.000 description 1
- 229930013356 epothilone Natural products 0.000 description 1
- 150000003883 epothilone derivatives Chemical class 0.000 description 1
- 229950009760 epratuzumab Drugs 0.000 description 1
- 229940082789 erbitux Drugs 0.000 description 1
- 229950004292 erlizumab Drugs 0.000 description 1
- 229960001433 erlotinib Drugs 0.000 description 1
- YSMODUONRAFBET-UHNVWZDZSA-N erythro-5-hydroxy-L-lysine Chemical compound NC[C@H](O)CC[C@H](N)C(O)=O YSMODUONRAFBET-UHNVWZDZSA-N 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- ITSGNOIFAJAQHJ-BMFNZSJVSA-N esorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)C[C@H](C)O1 ITSGNOIFAJAQHJ-BMFNZSJVSA-N 0.000 description 1
- 229950002017 esorubicin Drugs 0.000 description 1
- LJQQFQHBKUKHIS-WJHRIEJJSA-N esperamicin Chemical compound O1CC(NC(C)C)C(OC)CC1OC1C(O)C(NOC2OC(C)C(SC)C(O)C2)C(C)OC1OC1C(\C2=C/CSSSC)=C(NC(=O)OC)C(=O)C(OC3OC(C)C(O)C(OC(=O)C=4C(=CC(OC)=C(OC)C=4)NC(=O)C(=C)OC)C3)C2(O)C#C\C=C/C#C1 LJQQFQHBKUKHIS-WJHRIEJJSA-N 0.000 description 1
- 230000032050 esterification Effects 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- 229960001842 estramustine Drugs 0.000 description 1
- FRPJXPJMRWBBIH-RBRWEJTLSA-N estramustine Chemical compound ClCCN(CCCl)C(=O)OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 FRPJXPJMRWBBIH-RBRWEJTLSA-N 0.000 description 1
- 229940011871 estrogen Drugs 0.000 description 1
- 239000000262 estrogen Substances 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 125000001301 ethoxy group Chemical group [H]C([H])([H])C([H])([H])O* 0.000 description 1
- QSRLNKCNOLVZIR-KRWDZBQOSA-N ethyl (2s)-2-[[2-[4-[bis(2-chloroethyl)amino]phenyl]acetyl]amino]-4-methylsulfanylbutanoate Chemical compound CCOC(=O)[C@H](CCSC)NC(=O)CC1=CC=C(N(CCCl)CCCl)C=C1 QSRLNKCNOLVZIR-KRWDZBQOSA-N 0.000 description 1
- OAYLNYINCPYISS-UHFFFAOYSA-N ethyl acetate;hexane Chemical compound CCCCCC.CCOC(C)=O OAYLNYINCPYISS-UHFFFAOYSA-N 0.000 description 1
- 239000005038 ethylene vinyl acetate Substances 0.000 description 1
- 229960005237 etoglucid Drugs 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 229960000255 exemestane Drugs 0.000 description 1
- 208000021045 exocrine pancreatic carcinoma Diseases 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 229950011548 fadrozole Drugs 0.000 description 1
- 229940043168 fareston Drugs 0.000 description 1
- 229940087861 faslodex Drugs 0.000 description 1
- 229950001563 felvizumab Drugs 0.000 description 1
- ODKNJVUHOIMIIZ-RRKCRQDMSA-N floxuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ODKNJVUHOIMIIZ-RRKCRQDMSA-N 0.000 description 1
- 229960000961 floxuridine Drugs 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 1
- 238000002875 fluorescence polarization Methods 0.000 description 1
- 229910052731 fluorine Inorganic materials 0.000 description 1
- 239000011737 fluorine Substances 0.000 description 1
- 229960002074 flutamide Drugs 0.000 description 1
- MKXKFYHWDHIYRV-UHFFFAOYSA-N flutamide Chemical compound CC(C)C(=O)NC1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 MKXKFYHWDHIYRV-UHFFFAOYSA-N 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 239000011888 foil Substances 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- 229960000304 folic acid Drugs 0.000 description 1
- 150000002224 folic acids Chemical class 0.000 description 1
- VVIAGPKUTFNRDU-ABLWVSNPSA-N folinic acid Chemical compound C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-ABLWVSNPSA-N 0.000 description 1
- 235000008191 folinic acid Nutrition 0.000 description 1
- 239000011672 folinic acid Substances 0.000 description 1
- 229950004923 fontolizumab Drugs 0.000 description 1
- 150000004675 formic acid derivatives Chemical class 0.000 description 1
- 229960004783 fotemustine Drugs 0.000 description 1
- YAKWPXVTIGTRJH-UHFFFAOYSA-N fotemustine Chemical compound CCOP(=O)(OCC)C(C)NC(=O)N(CCCl)N=O YAKWPXVTIGTRJH-UHFFFAOYSA-N 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 229960002258 fulvestrant Drugs 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-L fumarate(2-) Chemical class [O-]C(=O)\C=C\C([O-])=O VZCYOOQTPOCHFL-OWOJBTEDSA-L 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 229960002598 fumaric acid Drugs 0.000 description 1
- 125000003838 furazanyl group Chemical group 0.000 description 1
- 125000004612 furopyridinyl group Chemical group O1C(=CC2=C1C=CC=N2)* 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 229940044658 gallium nitrate Drugs 0.000 description 1
- 229960003692 gamma aminobutyric acid Drugs 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 208000015419 gastrin-producing neuroendocrine tumor Diseases 0.000 description 1
- 201000000052 gastrinoma Diseases 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 229960002584 gefitinib Drugs 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 229940020967 gemzar Drugs 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 235000021474 generally recognized As safe (food) Nutrition 0.000 description 1
- 235000021473 generally recognized as safe (food ingredients) Nutrition 0.000 description 1
- 201000003115 germ cell cancer Diseases 0.000 description 1
- 239000012362 glacial acetic acid Substances 0.000 description 1
- 229940080856 gleevec Drugs 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 229940097043 glucuronic acid Drugs 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 229960004275 glycolic acid Drugs 0.000 description 1
- 229930182470 glycoside Natural products 0.000 description 1
- 125000003147 glycosyl group Chemical group 0.000 description 1
- 229960002913 goserelin Drugs 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 201000009277 hairy cell leukemia Diseases 0.000 description 1
- 125000001475 halogen functional group Chemical group 0.000 description 1
- 239000007902 hard capsule Substances 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- 150000002373 hemiacetals Chemical group 0.000 description 1
- 208000006359 hepatoblastoma Diseases 0.000 description 1
- 201000002735 hepatocellular adenoma Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- MNWFXJYAOYHMED-UHFFFAOYSA-N heptanoic acid Chemical class CCCCCCC(O)=O MNWFXJYAOYHMED-UHFFFAOYSA-N 0.000 description 1
- 229940022353 herceptin Drugs 0.000 description 1
- UUVWYPNAQBNQJQ-UHFFFAOYSA-N hexamethylmelamine Chemical compound CN(C)C1=NC(N(C)C)=NC(N(C)C)=N1 UUVWYPNAQBNQJQ-UHFFFAOYSA-N 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 230000003301 hydrolyzing effect Effects 0.000 description 1
- 229920001477 hydrophilic polymer Polymers 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 229920001600 hydrophobic polymer Polymers 0.000 description 1
- 125000004356 hydroxy functional group Chemical group O* 0.000 description 1
- QJHBJHUKURJDLG-UHFFFAOYSA-N hydroxy-L-lysine Natural products NCCCCC(NO)C(O)=O QJHBJHUKURJDLG-UHFFFAOYSA-N 0.000 description 1
- 229960001330 hydroxycarbamide Drugs 0.000 description 1
- 229920003063 hydroxymethyl cellulose Polymers 0.000 description 1
- 229940031574 hydroxymethyl cellulose Drugs 0.000 description 1
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- 206010020718 hyperplasia Diseases 0.000 description 1
- 229940015872 ibandronate Drugs 0.000 description 1
- 229960000908 idarubicin Drugs 0.000 description 1
- 229960003685 imatinib mesylate Drugs 0.000 description 1
- NVOITFNSLADCKT-UHFFFAOYSA-N imidazo[4,5-b]pyrazine Chemical compound C1=C[N]C2=NC=NC2=N1 NVOITFNSLADCKT-UHFFFAOYSA-N 0.000 description 1
- 125000002632 imidazolidinyl group Chemical group 0.000 description 1
- MTNDZQHUAFNZQY-UHFFFAOYSA-N imidazoline Chemical group C1CN=CN1 MTNDZQHUAFNZQY-UHFFFAOYSA-N 0.000 description 1
- 125000004857 imidazopyridinyl group Chemical group N1C(=NC2=C1C=CC=N2)* 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- DBIGHPPNXATHOF-UHFFFAOYSA-N improsulfan Chemical compound CS(=O)(=O)OCCCNCCCOS(C)(=O)=O DBIGHPPNXATHOF-UHFFFAOYSA-N 0.000 description 1
- 229950008097 improsulfan Drugs 0.000 description 1
- 201000004933 in situ carcinoma Diseases 0.000 description 1
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 description 1
- PZOUSPYUWWUPPK-UHFFFAOYSA-N indole Chemical group CC1=CC=CC2=C1C=CN2 PZOUSPYUWWUPPK-UHFFFAOYSA-N 0.000 description 1
- RKJUIXBNRJVNHR-UHFFFAOYSA-N indolenine Chemical group C1=CC=C2CC=NC2=C1 RKJUIXBNRJVNHR-UHFFFAOYSA-N 0.000 description 1
- 125000003387 indolinyl group Chemical group N1(CCC2=CC=CC=C12)* 0.000 description 1
- 125000003406 indolizinyl group Chemical group C=1(C=CN2C=CC=CC12)* 0.000 description 1
- 125000001041 indolyl group Chemical group 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 229940102223 injectable solution Drugs 0.000 description 1
- 229940102213 injectable suspension Drugs 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 229950004101 inotuzumab ozogamicin Drugs 0.000 description 1
- 206010022498 insulinoma Diseases 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 210000002570 interstitial cell Anatomy 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 201000010985 invasive ductal carcinoma Diseases 0.000 description 1
- 150000004694 iodide salts Chemical class 0.000 description 1
- PNDPGZBMCMUPRI-UHFFFAOYSA-N iodine Chemical compound II PNDPGZBMCMUPRI-UHFFFAOYSA-N 0.000 description 1
- 125000002346 iodo group Chemical group I* 0.000 description 1
- 150000005755 iodopyridines Chemical class 0.000 description 1
- 238000005342 ion exchange Methods 0.000 description 1
- 239000012500 ion exchange media Substances 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 229940084651 iressa Drugs 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- AWJUIBRHMBBTKR-UHFFFAOYSA-N iso-quinoline Natural products C1=NC=CC2=CC=CC=C21 AWJUIBRHMBBTKR-UHFFFAOYSA-N 0.000 description 1
- KQNPFQTWMSNSAP-UHFFFAOYSA-N isobutyric acid Chemical class CC(C)C(O)=O KQNPFQTWMSNSAP-UHFFFAOYSA-N 0.000 description 1
- GWVMLCQWXVFZCN-UHFFFAOYSA-N isoindoline Chemical group C1=CC=C2CNCC2=C1 GWVMLCQWXVFZCN-UHFFFAOYSA-N 0.000 description 1
- 238000006317 isomerization reaction Methods 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- ZLTPDFXIESTBQG-UHFFFAOYSA-N isothiazole Chemical group C=1C=NSC=1 ZLTPDFXIESTBQG-UHFFFAOYSA-N 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 210000001117 keloid Anatomy 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 208000022013 kidney Wilms tumor Diseases 0.000 description 1
- 229950000518 labetuzumab Drugs 0.000 description 1
- 150000003893 lactate salts Chemical class 0.000 description 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 229960004891 lapatinib Drugs 0.000 description 1
- 239000010410 layer Substances 0.000 description 1
- 229940115286 lentinan Drugs 0.000 description 1
- 229960001691 leucovorin Drugs 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- RGLRXNKKBLIBQS-XNHQSDQCSA-N leuprolide acetate Chemical compound CC(O)=O.CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 RGLRXNKKBLIBQS-XNHQSDQCSA-N 0.000 description 1
- 229950002950 lintuzumab Drugs 0.000 description 1
- 206010024627 liposarcoma Diseases 0.000 description 1
- 239000008263 liquid aerosol Substances 0.000 description 1
- 238000004811 liquid chromatography Methods 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 229960002247 lomustine Drugs 0.000 description 1
- DHMTURDWPRKSOA-RUZDIDTESA-N lonafarnib Chemical compound C1CN(C(=O)N)CCC1CC(=O)N1CCC([C@@H]2C3=C(Br)C=C(Cl)C=C3CCC3=CC(Br)=CN=C32)CC1 DHMTURDWPRKSOA-RUZDIDTESA-N 0.000 description 1
- YROQEQPFUCPDCP-UHFFFAOYSA-N losoxantrone Chemical compound OCCNCCN1N=C2C3=CC=CC(O)=C3C(=O)C3=C2C1=CC=C3NCCNCCO YROQEQPFUCPDCP-UHFFFAOYSA-N 0.000 description 1
- 229950008745 losoxantrone Drugs 0.000 description 1
- 238000003819 low-pressure liquid chromatography Methods 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 201000005296 lung carcinoma Diseases 0.000 description 1
- RVFGKBWWUQOIOU-NDEPHWFRSA-N lurtotecan Chemical compound O=C([C@]1(O)CC)OCC(C(N2CC3=4)=O)=C1C=C2C3=NC1=CC=2OCCOC=2C=C1C=4CN1CCN(C)CC1 RVFGKBWWUQOIOU-NDEPHWFRSA-N 0.000 description 1
- 229950002654 lurtotecan Drugs 0.000 description 1
- 239000008176 lyophilized powder Substances 0.000 description 1
- OTCKOJUMXQWKQG-UHFFFAOYSA-L magnesium bromide Chemical compound [Mg+2].[Br-].[Br-] OTCKOJUMXQWKQG-UHFFFAOYSA-L 0.000 description 1
- 229910001623 magnesium bromide Inorganic materials 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 1
- FRIJBUGBVQZNTB-UHFFFAOYSA-M magnesium;ethane;bromide Chemical compound [Mg+2].[Br-].[CH2-]C FRIJBUGBVQZNTB-UHFFFAOYSA-M 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 229940098895 maleic acid Drugs 0.000 description 1
- 150000002688 maleic acid derivatives Chemical class 0.000 description 1
- 201000004593 malignant giant cell tumor Diseases 0.000 description 1
- 201000000289 malignant teratoma Diseases 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- MQXVYODZCMMZEM-ZYUZMQFOSA-N mannomustine Chemical compound ClCCNC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CNCCCl MQXVYODZCMMZEM-ZYUZMQFOSA-N 0.000 description 1
- 229950008612 mannomustine Drugs 0.000 description 1
- 229950008001 matuzumab Drugs 0.000 description 1
- WKPWGQKGSOKKOO-RSFHAFMBSA-N maytansine Chemical compound CO[C@@H]([C@@]1(O)C[C@](OC(=O)N1)([C@H]([C@@H]1O[C@@]1(C)[C@@H](OC(=O)[C@H](C)N(C)C(C)=O)CC(=O)N1C)C)[H])\C=C\C=C(C)\CC2=CC(OC)=C(Cl)C1=C2 WKPWGQKGSOKKOO-RSFHAFMBSA-N 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 229960004961 mechlorethamine Drugs 0.000 description 1
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 1
- RQZAXGRLVPAYTJ-GQFGMJRRSA-N megestrol acetate Chemical compound C1=C(C)C2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@@](C(C)=O)(OC(=O)C)[C@@]1(C)CC2 RQZAXGRLVPAYTJ-GQFGMJRRSA-N 0.000 description 1
- 229960004296 megestrol acetate Drugs 0.000 description 1
- 229960001924 melphalan Drugs 0.000 description 1
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 210000002418 meninge Anatomy 0.000 description 1
- 229950009246 mepitiostane Drugs 0.000 description 1
- 229960005108 mepolizumab Drugs 0.000 description 1
- 150000002736 metal compounds Chemical class 0.000 description 1
- 125000005341 metaphosphate group Chemical group 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-M methanesulfonate group Chemical class CS(=O)(=O)[O-] AFVFQIVMOAPDHO-UHFFFAOYSA-M 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 1
- VJRAUFKOOPNFIQ-TVEKBUMESA-N methyl (1r,2r,4s)-4-[(2r,4s,5s,6s)-5-[(2s,4s,5s,6s)-5-[(2s,4s,5s,6s)-4,5-dihydroxy-6-methyloxan-2-yl]oxy-4-hydroxy-6-methyloxan-2-yl]oxy-4-(dimethylamino)-6-methyloxan-2-yl]oxy-2-ethyl-2,5,7,10-tetrahydroxy-6,11-dioxo-3,4-dihydro-1h-tetracene-1-carboxylat Chemical compound O([C@H]1[C@@H](O)C[C@@H](O[C@H]1C)O[C@H]1[C@H](C[C@@H](O[C@H]1C)O[C@H]1C[C@]([C@@H](C2=CC=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C21)C(=O)OC)(O)CC)N(C)C)[C@H]1C[C@H](O)[C@H](O)[C@H](C)O1 VJRAUFKOOPNFIQ-TVEKBUMESA-N 0.000 description 1
- QRMNENFZDDYDEF-GOSISDBHSA-N methyl (8s)-8-(bromomethyl)-2-methyl-4-(4-methylpiperazine-1-carbonyl)oxy-6-(5,6,7-trimethoxy-1h-indole-2-carbonyl)-7,8-dihydro-3h-pyrrolo[3,2-e]indole-1-carboxylate Chemical compound C1([C@H](CBr)CN(C1=C1)C(=O)C=2NC3=C(OC)C(OC)=C(OC)C=C3C=2)=C2C(C(=O)OC)=C(C)NC2=C1OC(=O)N1CCN(C)CC1 QRMNENFZDDYDEF-GOSISDBHSA-N 0.000 description 1
- QPJVMBTYPHYUOC-UHFFFAOYSA-N methyl benzoate Chemical class COC(=O)C1=CC=CC=C1 QPJVMBTYPHYUOC-UHFFFAOYSA-N 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- WBYWAXJHAXSJNI-UHFFFAOYSA-N methyl p-hydroxycinnamate Natural products OC(=O)C=CC1=CC=CC=C1 WBYWAXJHAXSJNI-UHFFFAOYSA-N 0.000 description 1
- NQMRYBIKMRVZLB-UHFFFAOYSA-N methylamine hydrochloride Chemical compound [Cl-].[NH3+]C NQMRYBIKMRVZLB-UHFFFAOYSA-N 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- HRHKSTOGXBBQCB-VFWICMBZSA-N methylmitomycin Chemical compound O=C1C(N)=C(C)C(=O)C2=C1[C@@H](COC(N)=O)[C@@]1(OC)[C@H]3N(C)[C@H]3CN12 HRHKSTOGXBBQCB-VFWICMBZSA-N 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 239000004530 micro-emulsion Substances 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 235000010755 mineral Nutrition 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 230000000116 mitigating effect Effects 0.000 description 1
- 229960005485 mitobronitol Drugs 0.000 description 1
- 229960003539 mitoguazone Drugs 0.000 description 1
- MXWHMTNPTTVWDM-NXOFHUPFSA-N mitoguazone Chemical compound NC(N)=N\N=C(/C)\C=N\N=C(N)N MXWHMTNPTTVWDM-NXOFHUPFSA-N 0.000 description 1
- VFKZTMPDYBFSTM-GUCUJZIJSA-N mitolactol Chemical compound BrC[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)CBr VFKZTMPDYBFSTM-GUCUJZIJSA-N 0.000 description 1
- 229950010913 mitolactol Drugs 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 229960000350 mitotane Drugs 0.000 description 1
- 239000002808 molecular sieve Substances 0.000 description 1
- 125000002950 monocyclic group Chemical group 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 229960001521 motavizumab Drugs 0.000 description 1
- 208000010492 mucinous cystadenocarcinoma Diseases 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 208000025113 myeloid leukemia Diseases 0.000 description 1
- 208000009091 myxoma Diseases 0.000 description 1
- NJSMWLQOCQIOPE-OCHFTUDZSA-N n-[(e)-[10-[(e)-(4,5-dihydro-1h-imidazol-2-ylhydrazinylidene)methyl]anthracen-9-yl]methylideneamino]-4,5-dihydro-1h-imidazol-2-amine Chemical compound N1CCN=C1N\N=C\C(C1=CC=CC=C11)=C(C=CC=C2)C2=C1\C=N\NC1=NCCN1 NJSMWLQOCQIOPE-OCHFTUDZSA-N 0.000 description 1
- IZHFVZCDYQWDKF-UHFFFAOYSA-N n-[1-[4-(2-chloro-8-phenylpurin-9-yl)phenyl]cyclobutyl]acetamide Chemical compound C=1C=C(N2C3=NC(Cl)=NC=C3N=C2C=2C=CC=CC=2)C=CC=1C1(NC(=O)C)CCC1 IZHFVZCDYQWDKF-UHFFFAOYSA-N 0.000 description 1
- NFVJNJQRWPQVOA-UHFFFAOYSA-N n-[2-chloro-5-(trifluoromethyl)phenyl]-2-[3-(4-ethyl-5-ethylsulfanyl-1,2,4-triazol-3-yl)piperidin-1-yl]acetamide Chemical compound CCN1C(SCC)=NN=C1C1CN(CC(=O)NC=2C(=CC=C(C=2)C(F)(F)F)Cl)CCC1 NFVJNJQRWPQVOA-UHFFFAOYSA-N 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- YJEUSJGCQIHPOP-UHFFFAOYSA-N n-[7-[4-(1-aminocyclobutyl)phenyl]-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-2-yl]cyclopropanecarboxamide Chemical compound C=1C=C(N2C3=NC(NC(=O)C4CC4)=NC=C3C=C2C=2N=CC=CC=2)C=CC=1C1(N)CCC1 YJEUSJGCQIHPOP-UHFFFAOYSA-N 0.000 description 1
- HSZCJVZRHXPCIA-UHFFFAOYSA-N n-benzyl-n-ethylaniline Chemical compound C=1C=CC=CC=1N(CC)CC1=CC=CC=C1 HSZCJVZRHXPCIA-UHFFFAOYSA-N 0.000 description 1
- 239000002088 nanocapsule Substances 0.000 description 1
- PSZYNBSKGUBXEH-UHFFFAOYSA-N naphthalene-1-sulfonic acid Chemical class C1=CC=C2C(S(=O)(=O)O)=CC=CC2=C1 PSZYNBSKGUBXEH-UHFFFAOYSA-N 0.000 description 1
- KVBGVZZKJNLNJU-UHFFFAOYSA-N naphthalene-2-sulfonic acid Chemical class C1=CC=CC2=CC(S(=O)(=O)O)=CC=C21 KVBGVZZKJNLNJU-UHFFFAOYSA-N 0.000 description 1
- 125000004593 naphthyridinyl group Chemical group N1=C(C=CC2=CC=CN=C12)* 0.000 description 1
- 229960005027 natalizumab Drugs 0.000 description 1
- 229930014626 natural product Natural products 0.000 description 1
- 229940086322 navelbine Drugs 0.000 description 1
- 201000008026 nephroblastoma Diseases 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 208000007538 neurilemmoma Diseases 0.000 description 1
- 201000004662 neurofibroma of spinal cord Diseases 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 208000004649 neutrophil actin dysfunction Diseases 0.000 description 1
- 229960002653 nilutamide Drugs 0.000 description 1
- XWXYUMMDTVBTOU-UHFFFAOYSA-N nilutamide Chemical compound O=C1C(C)(C)NC(=O)N1C1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 XWXYUMMDTVBTOU-UHFFFAOYSA-N 0.000 description 1
- 229950010203 nimotuzumab Drugs 0.000 description 1
- 229960001420 nimustine Drugs 0.000 description 1
- VFEDRRNHLBGPNN-UHFFFAOYSA-N nimustine Chemical compound CC1=NC=C(CNC(=O)N(CCCl)N=O)C(N)=N1 VFEDRRNHLBGPNN-UHFFFAOYSA-N 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 150000002825 nitriles Chemical class 0.000 description 1
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 description 1
- MCSAJNNLRCFZED-UHFFFAOYSA-N nitroethane Chemical compound CC[N+]([O-])=O MCSAJNNLRCFZED-UHFFFAOYSA-N 0.000 description 1
- KGTDRFCXGRULNK-JYOBTZKQSA-N nogalamycin Chemical compound CO[C@@H]1[C@@](OC)(C)[C@@H](OC)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=C(O)C=C4[C@@]5(C)O[C@H]([C@H]([C@@H]([C@H]5O)N(C)C)O)OC4=C3C3=O)=C3C=C2[C@@H](C(=O)OC)[C@@](C)(O)C1 KGTDRFCXGRULNK-JYOBTZKQSA-N 0.000 description 1
- 229950009266 nogalamycin Drugs 0.000 description 1
- 229940085033 nolvadex Drugs 0.000 description 1
- 231100000344 non-irritating Toxicity 0.000 description 1
- 239000002736 nonionic surfactant Substances 0.000 description 1
- UMRZSTCPUPJPOJ-KNVOCYPGSA-N norbornane Chemical group C1C[C@H]2CC[C@@H]1C2 UMRZSTCPUPJPOJ-KNVOCYPGSA-N 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- 229950005751 ocrelizumab Drugs 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- WWZKQHOCKIZLMA-UHFFFAOYSA-M octanoate Chemical class CCCCCCCC([O-])=O WWZKQHOCKIZLMA-UHFFFAOYSA-M 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- CZDBNBLGZNWKMC-MWQNXGTOSA-N olivomycin Chemical class O([C@@H]1C[C@@H](O[C@H](C)[C@@H]1O)OC=1C=C2C=C3C[C@H]([C@@H](C(=O)C3=C(O)C2=C(O)C=1)O[C@H]1O[C@@H](C)[C@H](O)[C@@H](OC2O[C@@H](C)[C@H](O)[C@@H](O)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@H](O)[C@H](OC)[C@H](C)O1 CZDBNBLGZNWKMC-MWQNXGTOSA-N 0.000 description 1
- 229960000470 omalizumab Drugs 0.000 description 1
- 229950011093 onapristone Drugs 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 238000005580 one pot reaction Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000008427 organic disulfides Chemical class 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 150000002900 organolithium compounds Chemical class 0.000 description 1
- 125000001979 organolithium group Chemical group 0.000 description 1
- 201000008482 osteoarthritis Diseases 0.000 description 1
- 208000003388 osteoid osteoma Diseases 0.000 description 1
- 208000008798 osteoma Diseases 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 210000003101 oviduct Anatomy 0.000 description 1
- 125000001715 oxadiazolyl group Chemical group 0.000 description 1
- 150000003891 oxalate salts Chemical class 0.000 description 1
- 229940116315 oxalic acid Drugs 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- DWAFYCQODLXJNR-BNTLRKBRSA-L oxaliplatin Chemical compound O1C(=O)C(=O)O[Pt]11N[C@@H]2CCCC[C@H]2N1 DWAFYCQODLXJNR-BNTLRKBRSA-L 0.000 description 1
- 229960001756 oxaliplatin Drugs 0.000 description 1
- 125000003551 oxepanyl group Chemical group 0.000 description 1
- 125000003566 oxetanyl group Chemical group 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 238000007243 oxidative cyclization reaction Methods 0.000 description 1
- 150000002924 oxiranes Chemical class 0.000 description 1
- LXCFILQKKLGQFO-UHFFFAOYSA-N p-hydroxybenzoic acid methyl ester Natural products COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 1
- 229960000402 palivizumab Drugs 0.000 description 1
- UQPUONNXJVWHRM-UHFFFAOYSA-N palladium;triphenylphosphane Chemical compound [Pd].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 UQPUONNXJVWHRM-UHFFFAOYSA-N 0.000 description 1
- 238000002638 palliative care Methods 0.000 description 1
- VREZDOWOLGNDPW-UHFFFAOYSA-N pancratistatine Natural products C1=C2C3C(O)C(O)C(O)C(O)C3NC(=O)C2=C(O)C2=C1OCO2 VREZDOWOLGNDPW-UHFFFAOYSA-N 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 208000021255 pancreatic insulinoma Diseases 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 229950011485 pascolizumab Drugs 0.000 description 1
- 239000006072 paste Substances 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 229960001639 penicillamine Drugs 0.000 description 1
- 229960002340 pentostatin Drugs 0.000 description 1
- FPVKHBSQESCIEP-JQCXWYLXSA-N pentostatin Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC[C@H]2O)=C2N=C1 FPVKHBSQESCIEP-JQCXWYLXSA-N 0.000 description 1
- QIMGFXOHTOXMQP-GFAGFCTOSA-N peplomycin Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCCN[C@@H](C)C=1C=CC=CC=1)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1NC=NC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C QIMGFXOHTOXMQP-GFAGFCTOSA-N 0.000 description 1
- 229950003180 peplomycin Drugs 0.000 description 1
- 239000002304 perfume Substances 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 229950003203 pexelizumab Drugs 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 229940127557 pharmaceutical product Drugs 0.000 description 1
- 239000002831 pharmacologic agent Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- DYUMLJSJISTVPV-UHFFFAOYSA-N phenyl propanoate Chemical class CCC(=O)OC1=CC=CC=C1 DYUMLJSJISTVPV-UHFFFAOYSA-N 0.000 description 1
- WLJVXDMOQOGPHL-UHFFFAOYSA-N phenylacetic acid Chemical class OC(=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-UHFFFAOYSA-N 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 229940043441 phosphoinositide 3-kinase inhibitor Drugs 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 125000005541 phosphonamide group Chemical group 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- USRGIUJOYOXOQJ-GBXIJSLDSA-N phosphothreonine Chemical compound OP(=O)(O)O[C@H](C)[C@H](N)C(O)=O USRGIUJOYOXOQJ-GBXIJSLDSA-N 0.000 description 1
- DCWXELXMIBXGTH-UHFFFAOYSA-N phosphotyrosine Chemical compound OC(=O)C(N)CC1=CC=C(OP(O)(O)=O)C=C1 DCWXELXMIBXGTH-UHFFFAOYSA-N 0.000 description 1
- 125000005498 phthalate group Chemical class 0.000 description 1
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 208000024724 pineal body neoplasm Diseases 0.000 description 1
- 201000004123 pineal gland cancer Diseases 0.000 description 1
- 125000000587 piperidin-1-yl group Chemical group [H]C1([H])N(*)C([H])([H])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- NJBFOOCLYDNZJN-UHFFFAOYSA-N pipobroman Chemical compound BrCCC(=O)N1CCN(C(=O)CCBr)CC1 NJBFOOCLYDNZJN-UHFFFAOYSA-N 0.000 description 1
- 229960000952 pipobroman Drugs 0.000 description 1
- NUKCGLDCWQXYOQ-UHFFFAOYSA-N piposulfan Chemical compound CS(=O)(=O)OCCC(=O)N1CCN(C(=O)CCOS(C)(=O)=O)CC1 NUKCGLDCWQXYOQ-UHFFFAOYSA-N 0.000 description 1
- 229950001100 piposulfan Drugs 0.000 description 1
- 229960001221 pirarubicin Drugs 0.000 description 1
- 150000003057 platinum Chemical class 0.000 description 1
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 1
- 229920000747 poly(lactic acid) Polymers 0.000 description 1
- 229920003229 poly(methyl methacrylate) Polymers 0.000 description 1
- 229920000728 polyester Polymers 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 229920002338 polyhydroxyethylmethacrylate Polymers 0.000 description 1
- 239000004926 polymethyl methacrylate Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 229920001155 polypropylene Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 229950004406 porfiromycin Drugs 0.000 description 1
- 238000002600 positron emission tomography Methods 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229960004694 prednimustine Drugs 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- KCXFHTAICRTXLI-UHFFFAOYSA-N propane-1-sulfonic acid Chemical class CCCS(O)(=O)=O KCXFHTAICRTXLI-UHFFFAOYSA-N 0.000 description 1
- 229960003415 propylparaben Drugs 0.000 description 1
- UORVCLMRJXCDCP-UHFFFAOYSA-N propynoic acid Chemical class OC(=O)C#C UORVCLMRJXCDCP-UHFFFAOYSA-N 0.000 description 1
- 201000001514 prostate carcinoma Diseases 0.000 description 1
- 239000003197 protein kinase B inhibitor Substances 0.000 description 1
- 239000003909 protein kinase inhibitor Substances 0.000 description 1
- 230000009822 protein phosphorylation Effects 0.000 description 1
- 125000001042 pteridinyl group Chemical group N1=C(N=CC2=NC=CN=C12)* 0.000 description 1
- WOLQREOUPKZMEX-UHFFFAOYSA-N pteroyltriglutamic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(=O)NC(CCC(=O)NC(CCC(O)=O)C(O)=O)C(O)=O)C(O)=O)C=C1 WOLQREOUPKZMEX-UHFFFAOYSA-N 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 125000004307 pyrazin-2-yl group Chemical group [H]C1=C([H])N=C(*)C([H])=N1 0.000 description 1
- 125000004944 pyrazin-3-yl group Chemical group [H]C1=C([H])N=C(*)C([H])=N1 0.000 description 1
- DNXIASIHZYFFRO-UHFFFAOYSA-N pyrazoline Chemical group C1CN=NC1 DNXIASIHZYFFRO-UHFFFAOYSA-N 0.000 description 1
- 125000002755 pyrazolinyl group Chemical group 0.000 description 1
- 125000002206 pyridazin-3-yl group Chemical group [H]C1=C([H])C([H])=C(*)N=N1 0.000 description 1
- 125000004940 pyridazin-4-yl group Chemical group N1=NC=C(C=C1)* 0.000 description 1
- 125000004941 pyridazin-5-yl group Chemical group N1=NC=CC(=C1)* 0.000 description 1
- 125000004942 pyridazin-6-yl group Chemical group N1=NC=CC=C1* 0.000 description 1
- PBMFSQRYOILNGV-UHFFFAOYSA-N pyridazine Chemical group C1=CC=NN=C1 PBMFSQRYOILNGV-UHFFFAOYSA-N 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- ICSNLGPSRYBMBD-CDYZYAPPSA-N pyridin-2-amine Chemical compound NC1=CC=CC=[15N]1 ICSNLGPSRYBMBD-CDYZYAPPSA-N 0.000 description 1
- UBQKCCHYAOITMY-UHFFFAOYSA-N pyridin-2-ol Chemical compound OC1=CC=CC=N1 UBQKCCHYAOITMY-UHFFFAOYSA-N 0.000 description 1
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 229940107700 pyruvic acid Drugs 0.000 description 1
- 125000002294 quinazolinyl group Chemical group N1=C(N=CC2=CC=CC=C12)* 0.000 description 1
- 125000001567 quinoxalinyl group Chemical group N1=C(C=NC2=CC=CC=C12)* 0.000 description 1
- UOWVMDUEMSNCAV-WYENRQIDSA-N rachelmycin Chemical compound C1([C@]23C[C@@H]2CN1C(=O)C=1NC=2C(OC)=C(O)C4=C(C=2C=1)CCN4C(=O)C1=CC=2C=4CCN(C=4C(O)=C(C=2N1)OC)C(N)=O)=CC(=O)C1=C3C(C)=CN1 UOWVMDUEMSNCAV-WYENRQIDSA-N 0.000 description 1
- 238000000163 radioactive labelling Methods 0.000 description 1
- 239000002287 radioligand Substances 0.000 description 1
- 229960003876 ranibizumab Drugs 0.000 description 1
- 229940099538 rapamune Drugs 0.000 description 1
- BMKDZUISNHGIBY-UHFFFAOYSA-N razoxane Chemical compound C1C(=O)NC(=O)CN1C(C)CN1CC(=O)NC(=O)C1 BMKDZUISNHGIBY-UHFFFAOYSA-N 0.000 description 1
- 229960000460 razoxane Drugs 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000008521 reorganization Effects 0.000 description 1
- 239000013557 residual solvent Substances 0.000 description 1
- 229960003254 reslizumab Drugs 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 208000037803 restenosis Diseases 0.000 description 1
- 208000029922 reticulum cell sarcoma Diseases 0.000 description 1
- 230000002207 retinal effect Effects 0.000 description 1
- 229930002330 retinoic acid Natural products 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 238000004366 reverse phase liquid chromatography Methods 0.000 description 1
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- OWPCHSCAPHNHAV-LMONGJCWSA-N rhizoxin Chemical compound C/C([C@H](OC)[C@@H](C)[C@@H]1C[C@H](O)[C@]2(C)O[C@@H]2/C=C/[C@@H](C)[C@]2([H])OC(=O)C[C@@](C2)(C[C@@H]2O[C@H]2C(=O)O1)[H])=C\C=C\C(\C)=C\C1=COC(C)=N1 OWPCHSCAPHNHAV-LMONGJCWSA-N 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- 239000011435 rock Substances 0.000 description 1
- 229950004892 rodorubicin Drugs 0.000 description 1
- MBABCNBNDNGODA-WPZDJQSSSA-N rolliniastatin 1 Natural products O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@H]1[C@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@@H](O)CC=2C(O[C@@H](C)C=2)=O)CC1 MBABCNBNDNGODA-WPZDJQSSSA-N 0.000 description 1
- IMUQLZLGWJSVMV-UOBFQKKOSA-N roridin A Natural products CC(O)C1OCCC(C)C(O)C(=O)OCC2CC(=CC3OC4CC(OC(=O)C=C/C=C/1)C(C)(C23)C45CO5)C IMUQLZLGWJSVMV-UOBFQKKOSA-N 0.000 description 1
- 229950009092 rovelizumab Drugs 0.000 description 1
- VHXNKPBCCMUMSW-FQEVSTJZSA-N rubitecan Chemical compound C1=CC([N+]([O-])=O)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VHXNKPBCCMUMSW-FQEVSTJZSA-N 0.000 description 1
- 229950005374 ruplizumab 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
- 229960004889 salicylic acid Drugs 0.000 description 1
- 229930182947 sarcodictyin Natural products 0.000 description 1
- 229940043230 sarcosine Drugs 0.000 description 1
- 239000012047 saturated solution Substances 0.000 description 1
- 231100000241 scar Toxicity 0.000 description 1
- HFHDHCJBZVLPGP-UHFFFAOYSA-N schardinger α-dextrin Chemical class O1C(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(O)C2O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC2C(O)C(O)C1OC2CO HFHDHCJBZVLPGP-UHFFFAOYSA-N 0.000 description 1
- 206010039667 schwannoma Diseases 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- CXMXRPHRNRROMY-UHFFFAOYSA-N sebacic acid Chemical class OC(=O)CCCCCCCCC(O)=O CXMXRPHRNRROMY-UHFFFAOYSA-N 0.000 description 1
- 230000036303 septic shock Effects 0.000 description 1
- 208000004548 serous cystadenocarcinoma Diseases 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 229950008684 sibrotuzumab Drugs 0.000 description 1
- 229950003804 siplizumab Drugs 0.000 description 1
- 229950001403 sizofiran Drugs 0.000 description 1
- 210000003625 skull Anatomy 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- URGAHOPLAPQHLN-UHFFFAOYSA-N sodium aluminosilicate Chemical compound [Na+].[Al+3].[O-][Si]([O-])=O.[O-][Si]([O-])=O URGAHOPLAPQHLN-UHFFFAOYSA-N 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910001467 sodium calcium phosphate Inorganic materials 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000007901 soft capsule Substances 0.000 description 1
- 239000008247 solid mixture Substances 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 239000011877 solvent mixture Substances 0.000 description 1
- 238000003797 solvolysis reaction Methods 0.000 description 1
- 229950006551 sontuzumab Drugs 0.000 description 1
- 230000003595 spectral effect Effects 0.000 description 1
- VNFWTIYUKDMAOP-UHFFFAOYSA-N sphos Chemical compound COC1=CC=CC(OC)=C1C1=CC=CC=C1P(C1CCCCC1)C1CCCCC1 VNFWTIYUKDMAOP-UHFFFAOYSA-N 0.000 description 1
- 229950006315 spirogermanium Drugs 0.000 description 1
- ICXJVZHDZFXYQC-UHFFFAOYSA-N spongistatin 1 Natural products OC1C(O2)(O)CC(O)C(C)C2CCCC=CC(O2)CC(O)CC2(O2)CC(OC)CC2CC(=O)C(C)C(OC(C)=O)C(C)C(=C)CC(O2)CC(C)(O)CC2(O2)CC(OC(C)=O)CC2CC(=O)OC2C(O)C(CC(=C)CC(O)C=CC(Cl)=C)OC1C2C ICXJVZHDZFXYQC-UHFFFAOYSA-N 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- DFVFTMTWCUHJBL-BQBZGAKWSA-N statine Chemical compound CC(C)C[C@H](N)[C@@H](O)CC(O)=O DFVFTMTWCUHJBL-BQBZGAKWSA-N 0.000 description 1
- SFVFIFLLYFPGHH-UHFFFAOYSA-M stearalkonium chloride Chemical compound [Cl-].CCCCCCCCCCCCCCCCCC[N+](C)(C)CC1=CC=CC=C1 SFVFIFLLYFPGHH-UHFFFAOYSA-M 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 238000011146 sterile filtration Methods 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 229960001052 streptozocin Drugs 0.000 description 1
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- TYFQFVWCELRYAO-UHFFFAOYSA-N suberic acid Chemical class OC(=O)CCCCCCC(O)=O TYFQFVWCELRYAO-UHFFFAOYSA-N 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 150000003890 succinate salts Chemical class 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- LSNNMFCWUKXFEE-UHFFFAOYSA-L sulfite Chemical class [O-]S([O-])=O LSNNMFCWUKXFEE-UHFFFAOYSA-L 0.000 description 1
- 229940124530 sulfonamide Drugs 0.000 description 1
- 150000003456 sulfonamides Chemical class 0.000 description 1
- 150000003460 sulfonic acids Chemical class 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 229940034785 sutent Drugs 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 229950001072 tadocizumab Drugs 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 229950004218 talizumab Drugs 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 229960003454 tamoxifen citrate Drugs 0.000 description 1
- 229940120982 tarceva Drugs 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- 150000003892 tartrate salts Chemical class 0.000 description 1
- RCINICONZNJXQF-XAZOAEDWSA-N taxol® Chemical compound O([C@@H]1[C@@]2(CC(C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3(C21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-XAZOAEDWSA-N 0.000 description 1
- 229940063683 taxotere Drugs 0.000 description 1
- 229950001788 tefibazumab Drugs 0.000 description 1
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@@H](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229960001278 teniposide Drugs 0.000 description 1
- 208000001608 teratocarcinoma Diseases 0.000 description 1
- SJCKHLJWAXGPGT-UHFFFAOYSA-N tert-butyl n-(2-chloropyridin-4-yl)carbamate Chemical compound CC(C)(C)OC(=O)NC1=CC=NC(Cl)=C1 SJCKHLJWAXGPGT-UHFFFAOYSA-N 0.000 description 1
- GZACWXCBJIJYJQ-UHFFFAOYSA-N tert-butyl n-[1-[4-[2-(1h-pyrazol-4-yl)-8-pyridin-2-ylpurin-9-yl]phenyl]cyclobutyl]carbamate Chemical compound C=1C=C(N2C3=NC(=NC=C3N=C2C=2N=CC=CC=2)C2=CNN=C2)C=CC=1C1(NC(=O)OC(C)(C)C)CCC1 GZACWXCBJIJYJQ-UHFFFAOYSA-N 0.000 description 1
- GVFRTTQQPOLGSH-UHFFFAOYSA-N tert-butyl n-[1-[4-[2-(2-fluorophenyl)-8-(2-methylpyrazol-3-yl)purin-9-yl]phenyl]cyclobutyl]carbamate Chemical compound CN1N=CC=C1C1=NC2=CN=C(C=3C(=CC=CC=3)F)N=C2N1C1=CC=C(C2(CCC2)NC(=O)OC(C)(C)C)C=C1 GVFRTTQQPOLGSH-UHFFFAOYSA-N 0.000 description 1
- YNNQJEWXPVJHMZ-UHFFFAOYSA-N tert-butyl n-[1-[4-[2-(cyclopropanecarbonylamino)-6-pyridin-2-ylpyrrolo[2,3-d]pyrimidin-7-yl]phenyl]cyclobutyl]carbamate Chemical compound C=1C=C(N2C3=NC(NC(=O)C4CC4)=NC=C3C=C2C=2N=CC=CC=2)C=CC=1C1(NC(=O)OC(C)(C)C)CCC1 YNNQJEWXPVJHMZ-UHFFFAOYSA-N 0.000 description 1
- ZIWYIVBJAMPPPX-UHFFFAOYSA-N tert-butyl n-[1-[4-[8-(2-methylpyrazol-3-yl)-2-[3-(trifluoromethoxy)phenyl]purin-9-yl]phenyl]cyclobutyl]carbamate Chemical compound CN1N=CC=C1C1=NC2=CN=C(C=3C=C(OC(F)(F)F)C=CC=3)N=C2N1C1=CC=C(C2(CCC2)NC(=O)OC(C)(C)C)C=C1 ZIWYIVBJAMPPPX-UHFFFAOYSA-N 0.000 description 1
- NFBCTOAMWSIIAL-UHFFFAOYSA-N tert-butyl n-[1-[4-[8-(4-methoxypyridin-3-yl)-2-phenylpurin-9-yl]phenyl]cyclobutyl]carbamate Chemical compound COC1=CC=NC=C1C1=NC2=CN=C(C=3C=CC=CC=3)N=C2N1C1=CC=C(C2(CCC2)NC(=O)OC(C)(C)C)C=C1 NFBCTOAMWSIIAL-UHFFFAOYSA-N 0.000 description 1
- IOGXOCVLYRDXLW-UHFFFAOYSA-N tert-butyl nitrite Chemical compound CC(C)(C)ON=O IOGXOCVLYRDXLW-UHFFFAOYSA-N 0.000 description 1
- 239000012414 tert-butyl nitrite Substances 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- BPEWUONYVDABNZ-DZBHQSCQSA-N testolactone Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(OC(=O)CC4)[C@@H]4[C@@H]3CCC2=C1 BPEWUONYVDABNZ-DZBHQSCQSA-N 0.000 description 1
- 229960005353 testolactone Drugs 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 1
- 125000005958 tetrahydrothienyl group Chemical group 0.000 description 1
- 125000004632 tetrahydrothiopyranyl group Chemical group S1C(CCCC1)* 0.000 description 1
- CXWXQJXEFPUFDZ-UHFFFAOYSA-N tetralin Chemical compound C1=CC=C2CCCCC2=C1 CXWXQJXEFPUFDZ-UHFFFAOYSA-N 0.000 description 1
- CZDYPVPMEAXLPK-UHFFFAOYSA-N tetramethylsilane Chemical compound C[Si](C)(C)C CZDYPVPMEAXLPK-UHFFFAOYSA-N 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 125000005308 thiazepinyl group Chemical group S1N=C(C=CC=C1)* 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 125000001583 thiepanyl group Chemical group 0.000 description 1
- 125000002053 thietanyl group Chemical group 0.000 description 1
- BRNULMACUQOKMR-UHFFFAOYSA-N thiomorpholine Chemical compound C1CSCCN1 BRNULMACUQOKMR-UHFFFAOYSA-N 0.000 description 1
- 229930192474 thiophene Chemical group 0.000 description 1
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- YFTWHEBLORWGNI-UHFFFAOYSA-N tiamiprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC(N)=NC2=C1NC=N2 YFTWHEBLORWGNI-UHFFFAOYSA-N 0.000 description 1
- 229950011457 tiamiprine Drugs 0.000 description 1
- 238000003354 tissue distribution assay Methods 0.000 description 1
- 230000009772 tissue formation Effects 0.000 description 1
- 230000017423 tissue regeneration Effects 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 239000012049 topical pharmaceutical composition Substances 0.000 description 1
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- 229950001802 toralizumab Drugs 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- 206010044412 transitional cell carcinoma Diseases 0.000 description 1
- 238000006478 transmetalation reaction Methods 0.000 description 1
- IUCJMVBFZDHPDX-UHFFFAOYSA-N tretamine Chemical compound C1CN1C1=NC(N2CC2)=NC(N2CC2)=N1 IUCJMVBFZDHPDX-UHFFFAOYSA-N 0.000 description 1
- 229950001353 tretamine Drugs 0.000 description 1
- 229960001727 tretinoin Drugs 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- PXSOHRWMIRDKMP-UHFFFAOYSA-N triaziquone Chemical compound O=C1C(N2CC2)=C(N2CC2)C(=O)C=C1N1CC1 PXSOHRWMIRDKMP-UHFFFAOYSA-N 0.000 description 1
- 229960004560 triaziquone Drugs 0.000 description 1
- YEMJHNYABQHWHL-UHFFFAOYSA-N tributyl(ethynyl)stannane Chemical compound CCCC[Sn](CCCC)(CCCC)C#C YEMJHNYABQHWHL-UHFFFAOYSA-N 0.000 description 1
- 229930013292 trichothecene Natural products 0.000 description 1
- 150000003327 trichothecene derivatives Chemical class 0.000 description 1
- AJSTXXYNEIHPMD-UHFFFAOYSA-N triethyl borate Chemical compound CCOB(OCC)OCC AJSTXXYNEIHPMD-UHFFFAOYSA-N 0.000 description 1
- 125000004205 trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 1
- KVJXBPDAXMEYOA-CXANFOAXSA-N trilostane Chemical compound OC1=C(C#N)C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@@]32O[C@@H]31 KVJXBPDAXMEYOA-CXANFOAXSA-N 0.000 description 1
- 229960001670 trilostane Drugs 0.000 description 1
- WRECIMRULFAWHA-UHFFFAOYSA-N trimethyl borate Chemical compound COB(OC)OC WRECIMRULFAWHA-UHFFFAOYSA-N 0.000 description 1
- CVDDRQSPINQVFT-UHFFFAOYSA-N trimethyl-[2-[(3-methyl-5-trimethylstannylpyrazol-1-yl)methoxy]ethyl]silane Chemical compound CC=1C=C([Sn](C)(C)C)N(COCC[Si](C)(C)C)N=1 CVDDRQSPINQVFT-UHFFFAOYSA-N 0.000 description 1
- NOYPYLRCIDNJJB-UHFFFAOYSA-N trimetrexate Chemical compound COC1=C(OC)C(OC)=CC(NCC=2C(=C3C(N)=NC(N)=NC3=CC=2)C)=C1 NOYPYLRCIDNJJB-UHFFFAOYSA-N 0.000 description 1
- 229960001099 trimetrexate Drugs 0.000 description 1
- 229950000212 trioxifene Drugs 0.000 description 1
- NHDIQVFFNDKAQU-UHFFFAOYSA-N tripropan-2-yl borate Chemical compound CC(C)OB(OC(C)C)OC(C)C NHDIQVFFNDKAQU-UHFFFAOYSA-N 0.000 description 1
- 229960000875 trofosfamide Drugs 0.000 description 1
- UMKFEPPTGMDVMI-UHFFFAOYSA-N trofosfamide Chemical compound ClCCN(CCCl)P1(=O)OCCCN1CCCl UMKFEPPTGMDVMI-UHFFFAOYSA-N 0.000 description 1
- 229950010147 troxacitabine Drugs 0.000 description 1
- RXRGZNYSEHTMHC-BQBZGAKWSA-N troxacitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1O[C@@H](CO)OC1 RXRGZNYSEHTMHC-BQBZGAKWSA-N 0.000 description 1
- HDZZVAMISRMYHH-LITAXDCLSA-N tubercidin Chemical compound C1=CC=2C(N)=NC=NC=2N1[C@@H]1O[C@@H](CO)[C@H](O)[C@H]1O HDZZVAMISRMYHH-LITAXDCLSA-N 0.000 description 1
- 208000022271 tubular adenoma Diseases 0.000 description 1
- 229950003364 tucotuzumab celmoleukin Drugs 0.000 description 1
- 108700008509 tucotuzumab celmoleukin Proteins 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 229940094060 tykerb Drugs 0.000 description 1
- GFNNBHLJANVSQV-UHFFFAOYSA-N tyrphostin AG 1478 Chemical compound C=12C=C(OC)C(OC)=CC2=NC=NC=1NC1=CC=CC(Cl)=C1 GFNNBHLJANVSQV-UHFFFAOYSA-N 0.000 description 1
- 229950009811 ubenimex Drugs 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 229960001055 uracil mustard Drugs 0.000 description 1
- 210000003708 urethra Anatomy 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 229950004362 urtoxazumab Drugs 0.000 description 1
- 210000004291 uterus Anatomy 0.000 description 1
- 210000001215 vagina Anatomy 0.000 description 1
- LLDWLPRYLVPDTG-UHFFFAOYSA-N vatalanib succinate Chemical compound OC(=O)CCC(O)=O.C1=CC(Cl)=CC=C1NC(C1=CC=CC=C11)=NN=C1CC1=CC=NC=C1 LLDWLPRYLVPDTG-UHFFFAOYSA-N 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 229940099039 velcade Drugs 0.000 description 1
- GPXBXXGIAQBQNI-UHFFFAOYSA-N vemurafenib Chemical compound CCCS(=O)(=O)NC1=CC=C(F)C(C(=O)C=2C3=CC(=CN=C3NC=2)C=2C=CC(Cl)=CC=2)=C1F GPXBXXGIAQBQNI-UHFFFAOYSA-N 0.000 description 1
- 229960003862 vemurafenib Drugs 0.000 description 1
- 208000009540 villous adenoma Diseases 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- UGGWPQSBPIFKDZ-KOTLKJBCSA-N vindesine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(N)=O)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1N=C1[C]2C=CC=C1 UGGWPQSBPIFKDZ-KOTLKJBCSA-N 0.000 description 1
- 229960004355 vindesine Drugs 0.000 description 1
- GBABOYUKABKIAF-IELIFDKJSA-N vinorelbine Chemical compound C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC GBABOYUKABKIAF-IELIFDKJSA-N 0.000 description 1
- 229960002066 vinorelbine Drugs 0.000 description 1
- CILBMBUYJCWATM-PYGJLNRPSA-N vinorelbine ditartrate Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.OC(=O)[C@H](O)[C@@H](O)C(O)=O.C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC CILBMBUYJCWATM-PYGJLNRPSA-N 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 229950004393 visilizumab Drugs 0.000 description 1
- 229960001771 vorozole Drugs 0.000 description 1
- XLMPPFTZALNBFS-INIZCTEOSA-N vorozole Chemical compound C1([C@@H](C2=CC=C3N=NN(C3=C2)C)N2N=CN=C2)=CC=C(Cl)C=C1 XLMPPFTZALNBFS-INIZCTEOSA-N 0.000 description 1
- 210000003905 vulva Anatomy 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000029663 wound healing Effects 0.000 description 1
- 229940053867 xeloda Drugs 0.000 description 1
- GDJZZWYLFXAGFH-UHFFFAOYSA-M xylenesulfonate group Chemical group C1(C(C=CC=C1)C)(C)S(=O)(=O)[O-] GDJZZWYLFXAGFH-UHFFFAOYSA-M 0.000 description 1
- 239000011592 zinc chloride Substances 0.000 description 1
- 235000005074 zinc chloride Nutrition 0.000 description 1
- GTLDTDOJJJZVBW-UHFFFAOYSA-N zinc cyanide Chemical compound [Zn+2].N#[C-].N#[C-] GTLDTDOJJJZVBW-UHFFFAOYSA-N 0.000 description 1
- CITILBVTAYEWKR-UHFFFAOYSA-L zinc trifluoromethanesulfonate Chemical compound [Zn+2].[O-]S(=O)(=O)C(F)(F)F.[O-]S(=O)(=O)C(F)(F)F CITILBVTAYEWKR-UHFFFAOYSA-L 0.000 description 1
- ZQJYTTPJYLKTTI-UHFFFAOYSA-M zinc;2h-pyridin-2-ide;bromide Chemical compound Br[Zn+].C1=CC=N[C-]=C1 ZQJYTTPJYLKTTI-UHFFFAOYSA-M 0.000 description 1
- 229950009268 zinostatin Drugs 0.000 description 1
- FBTUMDXHSRTGRV-ALTNURHMSA-N zorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(\C)=N\NC(=O)C=1C=CC=CC=1)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 FBTUMDXHSRTGRV-ALTNURHMSA-N 0.000 description 1
- 229960000641 zorubicin Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- 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
Definitions
- This invention relates to organic compounds useful for therapy and/or prophylaxis in a mammal, and in particular as inhibitors of serine/threonine protein kinases (e.g., AKT and related kinases), pharmaceutical compositions containing the inhibitors, and methods for preparing these inhibitors.
- the inhibitors are useful, for example, for the treatment of hyperproliferative diseases, such as cancer and inflammation, in mammals.
- Protein kinases are enzymes that catalyze the phosphorylation of hydroxy groups on tyrosine, serine and threonine residues of proteins by transfer of the terminal (gamma) phosphate from ATP. Through signal transduction pathways, these enzymes modulate cell growth, differentiation and proliferation, i.e., virtually all aspects of cell life in one way or another depend on PK activity (Hardie, G. and Hanks, S. (1995) The Protein Kinase Facts Book. I and II, Academic Press, San Diego, CA).
- abnormal PK activity has been related to a host of disorders, ranging from relatively non-life threatening diseases, such as psoriasis, to extremely virulent diseases, such as glioblastoma (brain cancer).
- Protein kinases are an important target class for therapeutic modulation (Cohen, P. (2002) Nature Rev. Drug Discovery 1 :309).
- Protein kinases include two classes; protein tyrosine kinases (PTK) and serine- threonine kinases (STK).
- the Protein Kinase B/Akt enzymes are a group of serine/threonine kinases that are overexpressed in a variety of human tumors.
- One of the best-characterized targets of the PI3K lipid products is the 57 KD serine/threonine protein kinase Akt, downstream of PI3K in the signal transduction pathway (Hemmings, B.A. (1997) Science 275:628; Hay N. (2005) Cancer Cell 8:179-183).
- Akt is the human homologue of the protooncogene v-akt of the acutely transforming retrovirus AKT8. Due to its high sequence homology to protein kinases A and C, Akt is also called Protein Kinase B (PKB) and Related to A and C (RAC). Three isoforms of Akt are known to exist, namely Aktl, Akt2 and Akt3, which exhibit an overall homology of 80% (Staal, S.P. (1987) Proc. Natl. Acad. Sci. 84:5034; Nakatani, K. (1999) Biochem. Biophys. Res. Commun. 257:906; Li et al (2002) Current Topics in Med. Chem.
- the Akt isoforms share a common domain organization that consists of a pleckstrin homology domain at the N-terminus, a kinase catalytic domain, and a short regulatory region at the C-terminus.
- both Akt2 and Akt3 exhibit splice variants.
- Akt is phosphorylated (activated) by PDK1 at T308, T309 and T305 for isoforms Aktl (PKBa), Akt2 ( ⁇ ) and Akt3 ( ⁇ ), respectively, and at S473, S474 and S472 for isoforms Aktl, Akt2 and Akt3, respectively.
- PKTa Aktl
- Akt2 Akt2
- Akt3 Akt3
- S473, S474 and S472 isoforms Aktl, Akt2 and Akt3, respectively.
- Akt activation requires its phosphorylation on residue Ser 473 in the C-terminal hydrophobic motif (Brodbeck et al (1999) J. Biol. Chem. 274:9133-9136; Coffer et al (1991) Eur. J. Biochem. 201 :475-481; Alessi et al (1997) Curr. Biol. 7:261-269). Although monophosphorylation of Akt activates the kinase, bis(phosphorylation) is required for maximal kinase activity.
- Akt is believed to assert its effect on cancer by suppressing apoptosis and enhancing both angiogenesis and proliferation (Toker et al. (2006) Cancer Res. 66(8):3963-3966). Akt is overexpressed in many forms of human cancer including, but not limited to, colon (Zinda et al (2001) Clin. Cancer Res. 7:2475), ovarian (Cheng et al (1992) Proc. Natl. Acad. Sci. USA 89:9267), brain (Haas Kogan et al (1998) Curr. Biol. 8:1195), lung (Brognard et al (2001) Cancer Res.
- pancreatic Bosset et al (1995) Int. J. Cancer 64:280-285; Cheng et al (1996) Proc. Natl. Acad. Sci. 93:3636-3641
- prostate Graff et al (2000) J. Biol. Chem. 275:24500
- gastric carcinomas Staal et al (1987) Proc. Natl. Acad. Sci. USA 84:5034-5037.
- PI3K/Akt/mammalian target of rapamycin (mTOR) pathway has been explored for targeted small molecule inhibitor therapy (Georgakis, G. and Younes, A. (2006) Expert Rev. Anticancer Ther. 6(1): 131-140; Granville et al (2006) Clin. Cancer Res. 12(3):679-689). Inhibition of PI3K/Akt signaling induces apoptosis and inhibits the growth of tumor cells that have elevated Akt levels (Kim et al (2005) Current Opinion in Investig. Drugs 6(12): 1250- 1258; Luo et al (2005) Molecular Cancer Ther. 4(6):977-986).
- a compound that inhibits (1) recruitment of Akt to the cell membrane, (2) activation by PDK1 or PDK2, (3) substrate phosphorylation, or (4) one of the downstream targets of Akt could be a valuable anticancer agent, either as a stand-alone therapy or in conjunction with other accepted procedures.
- This invention provides novel compounds that inhibit AKT protein kinases.
- the compounds of the present invention have utility as therapeutic agents for diseases and conditions that can be treated by the inhibition of AKT protein kinases.
- the present invention includes compounds having the general formula
- the invention also provides pharmaceutical compositions comprising a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
- the present invention provides a method of treating diseases or medical conditions in a mammal mediated by AKT protein kinases, comprising administering to said mammal one or more compounds of Formula 1 , or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in an amount effective to treat or prevent said disorder.
- AKT protein kinase mediated conditions that can be treated according to the methods of this invention include, but are not limited to, inflammatory, hyperproliferative, cardiovascular, neurodegenerative, gynecological, and dermatological diseases and disorders.
- the present invention provides a method of inhibiting the production of AKT protein kinases in a mammal, which comprises administering to said mammal a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof in an amount effective to inhibit production of an AKT protein kinase.
- the present invention provides methods of inhibiting the activity of AKT protein kinases, comprising contacting said kinase with a compound of Formula 1.
- inventive compounds may be used advantageously in combination with other known therapeutic agents. Accordingly, this invention also provides pharmaceutical compositions comprising a compound of Formula 1 or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in combination with a second therapeutic agent.
- This invention also provides compounds of Formula 1 and enantiomers, solvates, metabolites, and pharmaceutically acceptable salts and prodrugs thereof for use as medicaments in the treatment of AKT protein kinase-mediated conditions.
- An additional aspect of the invention is the use of a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, for therapy.
- the therapy comprises the treatment of an AKT protein kinase-mediated condition.
- kits for the treatment of an AKT protein kinase- mediated disease or disorder comprising a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, a container, and optionally a package insert or label indicating a treatment.
- the kits may further comprise a second compound or formulation comprising a second pharmaceutical agent useful for treating said disease or disorder.
- This invention further includes methods of preparing, methods of separating, and methods of purifying of the compounds of this invention.
- alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twelve carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below.
- alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n- Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1 -butyl (n-Bu, n-butyl, - CH 2 CH 2 CH 2 CH 3 ), 2-methyl-l -propyl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, - CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH 3 ) 3 ), 1-pentyl (n-pentyl, - CH 2 CH 2 CH 2 CH 3 ), 2-pentyl
- alkylene or "alkylenyl” as used herein refers to a linear or branched saturated divalent hydrocarbon radical of one to twelve carbon atoms, wherein the alkylene radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, methylene, ethylene, propylene, 2-methylpropylene, pentylene, and the like.
- cycloalkyl “carbocycle,” “carbocyclyl” and “carbocyclic ring” as used herein are used interchangeably and refer to saturated or partially unsaturated cyclic hydrocarbon radical having from three to twelve carbon atoms.
- cycloalkyl includes monocyclic and polycyclic (e.g., bicyclic and tricyclic) cycloalkyl structures, wherein the polycyclic structures optionally include a saturated or partially unsaturated cycloalkyl ring fused to a saturated, partially unsaturated or aromatic cycloalkyl or heterocyclic ring.
- cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like.
- Bicyclic carbocycles include those having 7 to 12 ring atoms arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, and bicyclo[3.2.2]nonane.
- the cycloalkyl may be optionally substituted independently with one or more substituents described herein.
- Aryl as used herein means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
- Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring.
- Exemplary aryl groups include, but are not limited to, radicals derived from benzene, naphthalene, anthracene, biphenyl, indene, indane, 1 ,2-dihydronapthalene, 1,2,3,4-tetrahydronapthalene, and the like.
- Aryl groups may be optionally substituted independently with one or more substituents described herein.
- heterocycle refers to a saturated or partially unsaturated carbocyclic radical of 3 to 8 ring atoms in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen and sulfur, the remaining ring atoms being C, where one or more ring atoms may be optionally substituted independently with one or more substituents described below.
- the radical may be a carbon radical or heteroatom radical.
- heterocycle includes heterocycloalkoxy.
- Heterocyclyl also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated, or aromatic carbocyclic or heterocyclic ring.
- heterocyclic rings include, but are not limited to, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl,
- the heterocycle may be C-attached or N-attached where such is possible.
- a group derived from pyrrole may be pyrrol- 1-yl (N-attached) or pyrrol-3-yl (C-attached).
- a group derived from imidazole may be imidazol-l-yl (N-attached) or imidazol-3-yl (C- attached).
- heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
- Non limiting examples of 5 to 6 membered heterocycloalkyl groups containing one, two or three heteroatoms selected from the group consisting of N, S and O can be selected from the group consisting of optionally substituted pyrrolidinyl, piperazinyl, pyrrolidinyl-2-one, thiomorpholine and 1,1 dioxide.
- heteroaryl refers to a monovalent aromatic radical of a 5-
- 6-, or 7-membered ring and includes fused ring systems (at least one of which is aromatic) of 5-10 atoms containing at least one heteroatom independently selected from nitrogen, oxygen, and sulfur.
- heteroaryl groups include, but are not limited to, pyridinyl, imidazolyl, imidazopyridinyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, pur
- Heteroaryl groups may be optionally substituted independently with one or more substituents described herein.
- 5 or 6 membered heteroaryl can be selected from the group consisting of optionally substituted pyridinyl, pyrimidinyl, thiazolyl, imidazolyl, oxazolyl, isoxazolyl, pyrazolyl, pyridinone and benzimidazolyl.
- carbon bonded heterocycles and heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
- carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5- pyridazinyl, 6-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
- nitrogen bonded heterocycles and heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of an isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
- nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1 -pyrrolyl, 1- imidazolyl, 1 -pyrazolyl, and 1-piperidinyl.
- halogen or "halo" as used herein means fluoro, chloro, bromo or iodo.
- haloalkoxyl refers to an alkoxyl group as defined herein that is substituted by 1, 2, 3, 4, 5 or 6 halogen as defined herein.
- haloalkoxyl groups include, but are not limited to Ci-C 6 -haloalkoxyl groups, including trifluoromethoxyl and trifluoroethoxyl.
- haloalkyl refers to an alkyl group as defined herein that is substituted by 1 , 2, 3, 4, 5 or 6 halogen as defined herein. Examples of haloalkyl groups include, but are not limited to Ci-C 6 -haloalkyl groups, including trifluoromethyl and trifluoroethyl.
- hydroxyalkyl refers to an alkyl group as defined herein that is substituted by 1, 2, 3, 4, 5 or 6 hydroxyl as defined herein.
- hydroxyalkyl groups include, but are not limited to CrC 6 -hydroxyalkyl groups, including hydroxymethyl and hydroxyethyl.
- alkylsulfonamide refers to a group of the following formula: -NR'R"S(0) 2 -alkyl., wherein alkyl is as defined herein and R' and R" are as defined herein.
- alkylsulfonamide group is methanesulfonyl.
- hydroxyl refers to an alcohol group, -OH.
- alkoxyl refers to an -O-alkyl group, wherein alkyl is as defined herein.
- alkoxyl groups include, but are not limited to Ci-C 6 -alkoxyl, including methoxy and ethoxy.
- the terms "compound of this invention,” “compounds of the present invention” and “compounds of Formula 1” includes compounds of Formula 1 and tautomers, resolved enantiomers, resolved diastereomers, racemic mixtures, solvates, metabolites, salts (including pharmaceutically acceptable salts) and pharmaceutically acceptable prodrugs thereof.
- the compounds of Formula 1 include solvates, pharmaceutically acceptable prodrugs and salts (including pharmaceutically acceptable salts) of such compounds.
- phrases "pharmaceutically acceptable” indicates that the substance or composition is compatible chemically and/or toxicologically with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
- a “solvate” refers to an association or complex of one or more solvent molecules and a compound of the invention.
- solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
- hydrate can also be used to refer to a complex wherein the solvent molecule is water.
- a "prodrug” is a compound that may be converted under physiological conditions or by solvolysis to the specified compound or to a salt of such compound.
- Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues, is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of a compound of the present invention.
- the amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes phosphoserine, phosphothreonine, phosphotyrosine, 4- hydroxyproline, hydroxylysine, demosine, isodemosine, gamma-carboxyglutamate, hippuric acid, octahydroindole-2-carboxylic acid, statine, l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, penicillamine, ornithine, 3-methylhistidine, norvaline, beta-alanine, gamma-aminobutyric acid, cirtulline, homocysteine, homoserine, methyl-alanine, para-benzoylphenylalanine, phenylglycine, propargylglycine, sarcosine, methionine sulfone and tert-butylglycine.
- prodrugs are also encompassed.
- a free carboxyl group of a compound of Formula 1 can be derivatized as an amide or alkyl ester.
- compounds of this invention comprising free hydroxy groups may be derivatized as prodrugs by converting the hydroxy group into a group such as, but not limited to, a phosphate ester, hemisuccinate, dimethylaminoacetate, or phosphoryloxymethyloxycarbonyl group, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 115.
- Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups.
- More specific examples include replacement of the hydrogen atom of the alcohol group with a group such as (Ci-C 6 )alkanoyloxymethyl, l-((Ci-C 6 )alkanoyloxy)ethyl, 1- methyl-l-((Ci-C6)alkanoyloxy)ethyl, (Ci-C 6 )alkoxycarbonyloxymethyl, N-(C C 6 )alkoxy- carbonylaminomethyl, succinoyl, (Ci-C 6 )alkanoyl, a-amino(Ci-C 4 )alkanoyl, arylacyl and a- aminoacyl, or ⁇ -aminoacyl-a-aminoacyl, where each a-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(0)(OH) 2 , -P(0)(0(Ci-C 6 )alkyl) 2 or glycosyl (the radical resulting from
- Free amines of compounds of Formula 1 can also be derivatized as amides, sulfonamides or phosphonamides. All of these moieties may incorporate groups including, but not limited to, ether, amine and carboxylic acid functionalities.
- a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as R-carbonyl, RO- carbonyl, NRR'-carbonyl, wherein R and R' are each independently (Ci-Cio)alkyl, (C 3 - C 7 )cycloalkyl, or benzyl, or R-carbonyl is a natural a-aminoacyl or natural oc-aminoacyl-natural a- aminoacyl, -C(OH)C(0)OY wherein Y is H, (d-C 6 )alkyl or benzyl, -C(OY 0 )Y !
- Y 0 is (C C 4 ) alkyl and Yi is (Ci-C 6 )alkyl, carboxy(Ci-C6)alkyl, amino(Ci-C 4 )alkyl or mono-N- or di-N,N- (Ci-C 6 )alkylaminoalkyl, or -C(Y 2 )Y 3 wherein Y 2 is H or methyl and Y 3 is mono-N- or di-N,N-(Cr C 6 )alkylamino, mo ⁇ holino, piperidin-l-yl or pyrrolidin-l-yl.
- compound of the invention may possess a sufficiently acidic group, a sufficiently basic group, or both functional groups, and accordingly react with any of a number of inorganic or organic bases or acids to form a salt.
- salts include those salts prepared by reaction of the compounds of the present invention with a mineral or organic acid or an inorganic base, such salts including, but not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyn-
- the desired salt may be prepared by any suitable method available in the art, for example, by treatment of the free base with an acidic compound, for example, an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p- toluenesulfonic acid or ethanesulfonic acid,
- an acidic compound for example, an inorgan
- the desired salt may be prepared by any suitable method, for example, by treatment of the free acid with an inorganic or organic base.
- suitable inorganic salts include those formed with alkali and alkaline earth metals, such as lithium, sodium, potassium, barium and calcium.
- suitable organic base salts include, for example, ammonium, dibenzylammonium, benzylammonium, 2- hydroxyethylammonium, bis(2-hydroxyethyl)ammonium, phenylethylbenzylamine, dibenzylethylenediamine, and the like salts.
- salts of acidic moieties may include, for example, those salts formed with procaine, quinine and N-methylglucosamine, plus salts formed with basic amino acids such as glycine, ornithine, histidine, phenylglycine, lysine and arginine.
- the salt is a "pharmaceutically acceptable salt" which, unless otherwise indicated, includes salts that retain the biological effectiveness of the corresponding free acid or base of the specified compound and are not biologically or otherwise undesirable.
- the compounds of Formula 1 also include other salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula 1 and/or for separating enantiomers of compounds of Formula 1.
- the present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention, and their uses. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen,
- isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
- the phrase "effective amount” means an amount of compound that, when administered to a mammal in need of such treatment, is sufficient to (i) treat or prevent a particular disease, condition, or disorder mediated by the activity of one or more AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) prevent or delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
- an effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
- the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
- efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
- the amount of a compound of Formula 1 that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
- Treating is intended to mean at least the mitigation of a disease condition in a mammal, such as a human, that is affected, at least in part, by the activity of one or more AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases.
- the terms “treat” and “treatment” refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder.
- beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
- Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
- Those in need of treatment include those already with the condition or disorder as well as those found to be predisposed to having the disease condition but have not yet been diagnosed as having it; modulating and/or inhibiting the disease condition.
- the terms “treating”, “treat”, or “treatment” embrace both preventative, i.e., prophylactic, and palliative treatment.
- the term "mammal” refers to a warm-blooded animal that has or is at risk of developing a disease described herein and includes, but is not limited to, guinea pigs, dogs, cats, rats, mice, hamsters, and primates, including humans.
- inventive compounds of Formula 1 are useful for inhibiting AKT protein kinases. Such compounds have utility as therapeutic agents for diseases that can be treated by the inhibition of the AKT protein kinase signaling pathway and tyrosine and serine/threonine kinase receptor pathways.
- the compounds of Formula 1 may also be useful as inhibitors of tyrosine kinases as well as serine and threonine kinases in addition to AKT.
- one aspect of the invention includes compounds of the Formula 1 :
- X is N or CR 3 ;
- Het is a roup selected from the grou consisting of:
- R 1', R2", and R 3 J is a group of formula (L):
- n 0, 1 or 2;
- R' and R' are independently selected from the group consisting of H, Ci-C6-alkyl and
- R"' is H, OH or C,-C 6 -alkyl; and the rest of R 1 , R 2 , and R 3 are independently selected from the group consisting of: H, cyano, Ci- C 6 -alkyl, aryl or heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, oxo, C]-C6-alkyl, Ci-C6-alkoxyl and C i -C6-hydroxyalkyl;
- R a and R b are independently selected from the group consisting of H, Ci-C6-alkyl, Ci-
- the compounds of formula 1 and tautomers, resolved enantiomers, resolved diastereomers and pharmaceutically acceptable salts thereof is provided.
- the compounds of formula 1 are those wherein R 1 is a group of formula (L):
- the compounds of formula 1 are those wherein R' and R" are H, n is 0 and the remaining substituents are as defined herein.
- the compounds of formula 1 are those wherein R 2 is a group of formula (L):
- the compounds of formula 1 are those wherein R 4 is selected from the group consisting of: halo, , aryl, 5 or 6 membered heteroaryl or heterocycloalkyl, which, aryl, heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one halo and the remaining substituents are as defined herein.
- the compounds of formula 1 are those wherein R 4 is aryl, 5 to 10 membered heterocycloalkyl containing one, two or three heteroatoms selected from the group consisting of N, S and O or 5 to 10 membered heteroaryl containing one, two or three heteroatoms selected from the group consisting of N, S and O, which aryl heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, oxo, hydroxyl, C C 6 -alkyl, C C 6 -alkoxyl, C C 6 -haloalkyl, Ci-C 6 -haloalkoxyl, COOH, C C 6 - hydroxyalkyl and C]-C6-alkylsulfonamide.
- the compounds of formula 1 are those wherein R 4 is aryl, 5 to 6 membered heterocycloalkyl containing one, two or three heteroatoms selected from the group consisting of N, S and O or 5 to 6 membered heteroaryl containing one, two or three heteroatoms selected from the group consisting of N, S and O, which aryl heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, oxo, hydroxyl, C C 6 -alkyl, C C 6 -alkoxyl, C r C 6 -haloalkyl, C r C 6 -haloalkoxyl, COOH, C C 6 - hydroxyalkyl and CrCe-alkylsulfonamide.
- the compounds of formula 1 are those wherein X is N and the remaining substituents are as defined herein.
- the compounds of formula 1 are those wherein X is CR 3 and the remaining substituents are as defined herein.
- the compounds of formula 1 are those wherein R 3 is H or
- Ci-C6-alkyl and the remaining substituents are as defined herein.
- the compounds of formula 1 are those wherein R 1 is phenyl and the remaining substituents are as defined herein.
- R is selected from cyano, Ci-C 6 -alkyl, aryl or 5 or 6 membered heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, Ci-Ce-alkyl, Ci-C6-alkoxyl and C ⁇ - C6-hydroxyalkyl; or is a group of formula (L):
- R 1 is selected from cyano, Ci-Ce-alkyl, aryl or 5 or 6 membered heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, Ci-C6-alkyl and Ci-C6-alkoxyl; or is a group of formula (L):
- R 1 is selected from cyano, Ci-Ce-alkyl, aryl or 5 or 6 membered heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, Ci-C6-alkyl and Ci-C6-alkoxyl.
- R is selected from H, Ci-C 6 -alkyl and aryl; or or is a group of formula (L):
- R 2 is selected from H, Ci-C6-alkyl and aryl.
- R 2 is selected from the group consisting of:
- R 3 is selected from H, C]-C6-alkyl and aryl; or or is a group of formula (L):
- R 3 is selected from H, Ci-C 6 -alkyl and aryl.
- R 3 is selected from the group consisting of:
- the compounds of formula 2 have the following formula 2-A:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 2-A are:
- Non-limiting examples of compounds of formula 2-B are:
- the compounds of formula 2 have the following formula 2-C:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 2-C are:
- the compounds of formula 2 have the following formula 2-D:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- a non limiting example of compound of formula 2-D is l-(4-(l- aminocyclobutyl)phenyl)-2-phenyl-lH-pyrrolo[3,2-b]pyridin-5-ol.
- the compounds of formula 2 have the following formula 2-E:
- R ! , R 2 , R 3 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 2-E are: l-(4-(6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl) cyclobutanamine,
- the compounds of formula 2 have the following formula 2-F:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 2-F are:
- the compounds of formula 2 have the following formula 2-G:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- a non-limiting example of a compound of formula 2-G is l-[4-(6-Phenyl- pyrrolo[2,3-b]pyrazin-5-yl)-phenyl]-cyclobutylamine.
- the compounds of formula 2 have the following formula 2-H:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- the compounds of formula 2 have the following formula 2-1:
- R 1 , R 2 , R 3 , R 4 and m are as defined herein.
- the compounds of formula 2 have the following formula 2-J:
- the compounds of formula 1 have the following formula 3:
- R 1 and R 2 are as defined herein.
- the compounds of formula 3 have the following formula 3-A:
- Non-limiting examples of compounds of formula 3-A are:
- the compounds of formula 3 have the following formula 3-B:
- R 1 , R 2 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 3-B are:
- R 1 , R 2 , R 4 and m are as defined herein.
- the compounds of formula 3 have the following formula 3-D:
- the compounds of formula 3 have the following formula 3-E:
- R 1 , R 2 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 3-E are:
- R 1 , R 2 , R 4 and m are as defined herein.
- the compounds of formula 3 have the following formula 3-G:
- R 1 , R 2 , R 4 and m are as defined herein.
- Non-limiting examples of compounds of formula 3-G are:
- R 1 , R 2 , R 4 and m are as defined herein.
- the compounds of formula 3 have the following formula 3-J:
- R 1 , R 2 , R 4 and m are as defined herein.
- the compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)-stereoisomers or as mixtures thereof.
- the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and diastereomers, and mixtures, racemic or otherwise, thereof. Accordingly, this invention also includes all such isomers, including diastereomeric mixtures, pure diastereomers and pure enantiomers of the compounds of this invention.
- enantiomer refers to two stereoisomers of a compound which are non- superimposable mirror images of one another.
- diastereomer refers to a pair of optical isomers which are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties, and reactivities.
- tautomer or tautomeric form refers to structural isomers of different energies which are interconvertible via a low energy barrier.
- proton tautomers also known as prototropic tautomers
- Valence tautomers include interconversions by reorganization of some of the bonding electrons.
- stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed wedge representing a particular configuration, then that stereoisomer is so specified and defined.
- a "metabolite” is a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof. Such products may result, for example, from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds of Formula 1, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
- Metabolites are identified, for example, by preparing a radiolabelled (e.g., I4 C or
- H) isotope of a compound of the invention administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples.
- a detectable dose e.g., greater than about 0.5 mg/kg
- an animal such as rat, mouse, guinea pig, monkey, or to a human
- sufficient time for metabolism to occur typically about 30 seconds to 30 hours
- isolating its conversion products from the urine, blood or other biological samples typically about 30 seconds to 30 hours
- the metabolite structures are determined in conventional fashion, e.g., by MS, LC/MS or NMR analysis.
- metabolites In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art.
- the metabolites so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.
- Compounds of this invention may be synthesized by synthetic routes that include processes analogous to those well known in the chemical arts, particularly in light of the description contained herein.
- the starting materials are generally available from commercial sources, such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements).
- Compounds of Formula 1 may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds.
- Libraries of compounds of Formula 1 may be prepared by a combinatorial 'split and mix' approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art.
- a compound library comprising at least 2 compounds of Formula 1, or salts thereof.
- Schemes 1-26 show a general method for preparing the compounds of the present invention, as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
- a palladium source such as tris(dibenzylideneacetone) dipalladium
- a ligand such as 9,9-dimethyl-4,5- bis(diphenylphosphino)xanthene (Xantphos)
- a base such as cesium carbonate
- a suitable solvent such as dioxane
- X may be fluoro, chloro, or methanesulfonyl
- a suitably protected aniline 1.2 (see T.W. Greene, supra) with or without a suitable base, such as diisopropylethylamine, with or without a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C to produce nitro-anilines 1.3.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- Reduction of the nitro group can be achieved by reacting nitro aniline 3 with hydrogen gas in the presence of a catalyst, such as Raney-nickel, in a suitable solvent, such as TFEF, at temperatures ranging from 25 °C to 200 °C, and at pressures ranging from 1 atm to 50 atm to produce amino-aniline 1.4.
- a catalyst such as Raney-nickel
- TFEF a suitable solvent
- a chemoselective reduction method such as sulfided platinum on carbon in a suitable solvent, such as ethanol, at temperatures ranging from 25 °C to 200 °C, and at pressures ranging from 1 atm to 50 atm to produce amino-aniline 1.4.
- Cyclization to the fused imidazo-heteroarene 1.6 can be achieved by reacting amino-aniline 1.4 with an aldehyde 1.5a in the presence of an oxidant, such as copper(II)acetate, in a suitable solvent, such as acetic acid, at temperatures ranging from 25 °C to 200 °C.
- imidazo-heteroarenes 1.6 may be synthesized by treating amino-anilines 1.4 with an orthoester 1.5b in the presence of an acid, such as 4-methylbenzenesulfonic acid, in a solvent, such as toluene, at temperatures ranging from 25 °C to 150 °C.
- the reduction of nitro anilines 1.3 can be performed in the presence of an orthoester 1.5b to affect the reduction and cyclization to imidazo-heteroarenes 1.6 in one step.
- R 4 may be a halogen to yield compounds 1.6 which may be reacted further. It may be advantageous to mask such functional groups using a protecting group (for a review of protecting groups see T.W. Greene, P.G.M. Wuts - Protective groups in Organic Synthesis, 3rd ed., John Wiley & Sons, 1999).
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- These cross coupling reactions may be carried out in suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- organometallic derivative e.g., a boronic acid or ester, trifluoroborate salt, organomagnesium, organozinc or organotin compound.
- organometallic derivative e.g., a boronic acid or ester, trifluoroborate salt, organomagnesium, organozinc or organotin compound.
- Any functional groups present, may need to be protected by a suitable protecting group.
- metals or metalloids can be achieved by generally well-known methods, such as metallation using metals or a metal-halogen exchange reaction.
- Useful metals for metallation include alkaline or alkaline earth metals or activated forms of such metals.
- Suitable reagents for use in metal-halogen exchange reactions include organolithium or organomagnesium compounds (e.g., N-butyllithium, tert-butyllithium or iso-propylmagnesium chloride or bromide).
- Subsequent transmetalation reactions of the organometallic intermediate may be performed as needed with a suitable soluble and reactive metal compound, such as magnesium chloride, magnesium bromide, tri-N-butyltin chloride, trimethyltin chloride, trimethyl borate, triethyl borate, tri-iso-propyl borate, zinc triflate or zinc chloride.
- a suitable soluble and reactive metal compound such as magnesium chloride, magnesium bromide, tri-N-butyltin chloride, trimethyltin chloride, trimethyl borate, triethyl borate, tri-iso-propyl borate, zinc triflate or zinc chloride.
- boronic acid pinacol ester can be conveniently achieved by reacting the halogen derivative directly with bis(pinacolato)diboron in the presence of dichloro[l,l'- bis(diphenylphosphino)ferrocene]palladium(II) and suitable bases (e.g., potassium or sodium acetate) in solvents such as DMSO, DMF, DMA or N- methylpyrrolidone at temperatures ranging from 80-160 °C. Conventional heating or microwave irradiation may be employed to maintain the appropriate temperature (for literature precedent of similar transformations, see Ishiyama T. et al., J. Org. Chem., 60.750S (2003)).
- organometallic derivatives may be utilized in cross- coupling reactions similar to those described above. Such couplings can be effected utilizing suitable coupling partners, such as aromatic, heteroaromatic halides or olefinic reagents under conditions identical or evidently similar and/or related to the methods described above.
- suitable coupling partners such as aromatic, heteroaromatic halides or olefinic reagents under conditions identical or evidently similar and/or related to the methods described above.
- Other methods may utilize the reactivity of organometallic derivatives generated from 3.2 by any of the methods described above.
- alkaline or alkaline earth metals e.g., organolithium, organomagnesium or organozinc compounds
- other electrophilic coupling partners such as, for example, activated olefins (MICHAEL-acceptors), aldehydes, nitriles, aromatic nitro compounds, carboxylic acid derivatives, oxiranes, aziridines, organic disulfides or organic halides.
- MICHAEL-acceptors activated olefins
- aldehydes aldehydes
- nitriles aromatic nitro compounds
- carboxylic acid derivatives e.g., oxiranes
- aziridines organic disulfides or organic halides
- This methodology may be extended to the incorporation of non-carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) that may optionally contain suitable protecting groups of alcohols, thiols or amines. Examples of such groups can be found in Greene T., supra. Exemplary methods of such utilization of non-carbon nucleophiles in related cross-coupling reactions may be found in Ley S. et al., Angew. Chem., /75:5558 (2003); Wolfe J. et al., Ace. Chem. Res., 31:805 (1998); Hartwig, Ace. Chem.
- Some of the compounds in the present invention may be synthesized by treatment of chloro purine 3.2 with a nucleophile, such as a primary or secondary amine, or an alcohol, in the presence of carbon monoxide, a suitable catalyst, such as dichloro[l,l'- bis(diphenylphosphino)ferrocene]palladium(II), a base, such as triethylamine, and a solvent, such as dioxane, at temperatures ranging from 80-160 °C. Conventional heating or microwave irradiation may be employed to maintain the appropriate temperature (exemplary methods of carbonylation may be found in Brennbower, A. et al., Angew. Chem., 48(23):4114 (2009)).
- a nucleophile such as a primary or secondary amine, or an alcohol
- a suitable catalyst such as dichloro[l,l'- bis(diphenylphosphino)ferrocene]palladium(II)
- compounds described in the invention may be synthesized by treating chloro-purine 3.2 with a suitably protected carbon-based nucleophiles (e.g., malonates) and non- carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) via nucleophilic aromatic substitution.
- a suitably protected carbon-based nucleophiles e.g., malonates
- non- carbon based nucleophiles e.g., alcohols, thiols, primary or secondary amines
- the reactions may be accelerated using an appropriate acid, such as toluenesulfonic acid, or base, such as triethylamine, diisopropylethylamine, sodium hydride, or an alkaline carbonate base, in the presence or absence of a suitable solvent, such as, but not limited to, THF, dioxane, DMF, DMA, DMSO, or acetonitrile.
- an appropriate acid such as toluenesulfonic acid
- base such as triethylamine, diisopropylethylamine, sodium hydride, or an alkaline carbonate base
- a suitable solvent such as, but not limited to, THF, dioxane, DMF, DMA, DMSO, or acetonitrile.
- 6-Substituted-4-chloro-5-nitro-pyrimidine 4.1 can be treated with an aniline, such as 1.2, in a suitable solvent, such as THF.
- the resulting nitro aniline 4.2 can be reduced with hydrogen gas in the presence of a suitable catalyst, such as raney nickel, in a suitable solvent, such as THF.
- Dianiline 4.3 can be cyclized to purine 4.4 using aldehyde 1.5a in the presence of an oxidant, such as copper(II) acetate in acetic acid.
- R 1 , R 2 , or R 4 may contain functional groups which have to be protected with a suitable protecting group.
- R 1 , R 2 , or R 4 may also be functional handles which allow further elaboration of 4.4.
- Purine 5.2 can by synthesized from 4,6-dichloro-5-nitro- pyrimidine 5.1 according to Scheme 4.
- suitable reagents such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds).
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- cross coupling reactions may be carried out in suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- Tetrasubstituted purines from the invention described herein can be synthesized from dichloropurine 6.2 (Scheme 6) which may be synthesized from 2,4,6-trichloro-5- nitropyrimidine according to Scheme 4. Functionalization of 6.2 according to the conditions described above followed by functionalization of the resulting chloropurine 6.3 according to the conditions described above generates tetrasubstituted purines 6.4.
- R 1 , R 2 , or R 4 may contain functional groups which have to be protected with a suitable protecting group.
- R 1 , R 2 , or R 4 may also be functional handles which allow further elaboration of 7.4.
- 6-Bromo-imidazo[4,5-b]pyridine 8.2 can be synthesized according to Scheme 7.
- suitable reagents such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds).
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- 5-Chloro-imidazo[4,5-b]pyridine 9.2 can be synthesized according to Scheme 7.
- suitable reagents such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds).
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- Dianiline 10.3 can be cyclized to imidazo[4,5- b]pyrazine 10.4 using aldehyde 1.5a in the presence of an oxidant, such as copper(II) acetate in acetic acid.
- an oxidant such as copper(II) acetate in acetic acid.
- R 1 , R 2 , or R 4 may contain functional groups which have to be protected with a suitable protecting group.
- R 1 , R 2 , or R 4 may also be functional handles which allow further elaboration of 10.4.
- Pg is an amine protecting group, e.g., acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc).
- amine protecting group e.g., acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc).
- alkynes 11.3 Treatment of alkynes 11.3 with a suitably protected aniline 1.2 (for a review of protecting groups see T.W. Greene, supra) in the presence of a palladium source, such as tris(dibenzylideneacetone) dipalladium, a ligand, such as 9,9-dimethyl-4,5- bis(diphenylphosphino)xanthene (Xantphos), a base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C produces anilino-alkynes 1 1.4.
- a palladium source such as tris(dibenzylideneacetone) dipalladium
- a ligand such as 9,9-dimethyl-4,5- bis(diphenylphosphino)xanthene (Xantphos)
- a base such as cesium carbonate
- a suitable solvent such as dioxane
- anilino-alkynes 11.4 Treatment of anilino-alkynes 11.4 with a suitable catalyst, such as bis- tributylphosphine palladium, a suitable additive, such as potassium fluoride, a base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C produces aryl-pyrroles 11.5.
- a suitable catalyst such as bis- tributylphosphine palladium
- a suitable additive such as potassium fluoride
- a base such as cesium carbonate
- a suitable solvent such as dioxane
- reaction to form aryl-pyrroles 11.5 from alkynes 11.3 may be performed as a one pot reaction.
- an optionally substituted amino-heteroarene (selected from Figure 2, with X 1 being a bromide, iodide, or trifluoromethanesulfonato, and X 2 being a fluoride or chloride) can be used as compound 11.1 and may be treated with a suitably protected aniline 1.2 (for a review of protecting groups see T.W. Greene, P.G.M.
- selected examples from the invention described herein can be synthesized from 4-chloro-3-iodopyridine 12.1.
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2-dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- 2-chloro-3-iodo-4-amino-pyridines (Scheme 13).
- Treatment of an optionally protected 2-chloro-3-iodo-4-amino-pyridine 13.1 can be treated with nucleophiles such as, but not limited to, amines, alkoxides, or thioalkoxides, in the presence of a suitable solvent, such as dioxane, to yield iodopyridines 13.2.
- iodide 13.2 Treatment of iodide 13.2 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro-bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 13.3.
- a suitable catalyst such as dichloro-bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- iodide 13.4 Treatment of iodide 13.4 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2-dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 13.5.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2-dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- Cyclization of amino alkyne 13.5 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert- butylphosphine)palladium(O), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[3,2-c]pyridine 13.6.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert- butylphosphine)palladium(O)
- a suitable solvent such as dioxane
- selected examples from the invention described herein can be synthesized from optionally substituted 2-bromo-3-chloropyridines 14.1 (Scheme 14).
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- chloride 14.2 Treatment of chloride 14.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 14.3.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- Cyclization of amino alkyne 14.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[3,2-b]pyridines 14.4.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- selected examples from the invention described herein can be synthesized from optionally substituted 2-chloro-3-iodo-pyridines 15.1 (Scheme 15).
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- chloride 15.2 Treatment of chloride 15.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 15.3.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- Cyclization of amino alkyne 15.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[2,3-b]pyridines 15.4.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- chloro alkyne 16.2 can be functionalized in a variety of ways. It may be advantageous to protect reactive functional groups, such as the amino group.
- Treatment of chloro-pyridine 16.2 with suitable reagents such, as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds).
- a suitable transition metal catalyst e.g., palladium compounds.
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- These cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- This methodology may be extended to the incorporation of non-carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) that may optionally contain suitable protecting groups of alcohols, thiols or amines. Examples of such groups can be found in Greene T. et al., supra. Exemplary methods of such utilization of non-carbon nucleophiles in related cross-coupling reactions may be found in Ley S. et al., Angew. Chem., /75:5558 (2003); Wolfe J. et al., Ace. Chem. Res., 31 :805 (1998); Hartwig, Ace. Chem.
- non-carbon based nucleophiles e.g., alcohols, thiols, primary or secondary amines
- compounds described in the invention may be synthesized by treating chloro-imidazo[4,5-b]pyridine 16.2 with a suitably protected carbon-based nucleophiles (e.g., malonates) and non-carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) via nucleophilic aromatic substitution.
- a suitably protected carbon-based nucleophiles e.g., malonates
- non-carbon based nucleophiles e.g., alcohols, thiols, primary or secondary amines
- the reactions may be accelerated using an appropriate acid, such as toluenesulfonic acid, or base, such as triethylamine, diisopropylethylamine, sodium hydride, or an alkaline carbonate base, in the presence or absence of a suitable solvent, such as, but not limited to, THF, dioxane, DMF, DMA, DMSO, or acetonitrile.
- a suitable solvent such as, but not limited to, THF, dioxane, DMF, DMA, DMSO, or acetonitrile.
- Amino-alkyne 16.3 can be converted to bromo-alkyne 16.4 under diazotization conditions, such as sodium nitrate and toluenesulfonic acid, in a suitable solvent, such as acetonitrile, followed by sodium iodide.
- bromide 16.4 Treatment of bromide 16.4 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 16.5.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- Cyclization of amino alkyne 16.5 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[2,3-b]pyridines 16.6.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- a suitable transition metal catalyst e.g., palladium compounds.
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- THF trifluorofuran
- dioxane dimethoxyethane
- diglyme diglyme
- dichloromethane dichloroethane
- acetonitrile acetonitrile
- DMF N-methylpyrrolidone
- water or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- Selected examples from the invention described herein can be synthesized from optionally substituted 4-chloro-5-iodo-pyrimidine 18.1 (Scheme 18).
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- chloride 18.2 Treatment of chloride 18.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields pyrrolo[2,3-d]pyrimidines 18.3.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- a suitable catalyst such as dichloro-bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- Cyclization of amino alkyne 19.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield 2-chloro pyrrolo[2,3-d]pyrimidines 19.4.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- a suitable transition metal catalyst e.g., palladium compounds.
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- the Pd catalyzed coupling or SnAr reaction from 19.4 to 19.5 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 19 as well.
- Selected examples from the invention described herein can be synthesized from optionally substituted 4,6-dichloro-5-iodo-pyrimidine 20.1 (Org. Lett.; 11(8), 2009; 1837 - 1840) as seen in Scheme 20.
- Treatment of dichloride 20.1 with an aniline, such as 1.2, in the presence of a base, such as triethylamine, in a suitable solvent, such as THF results in the formation of iodoaniline 20.2.
- Cyclization of amino alkyne 20.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield 4-chloro pyrrolo[2,3-d]pyrimidines 20.4.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- a suitable transition metal catalyst e.g., palladium compounds.
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- THF trifluorofuran
- dioxane dimethoxyethane
- diglyme diglyme
- dichloromethane dichloroethane
- acetonitrile acetonitrile
- DMF N-methylpyrrolidone
- water or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C.
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- 2,4.6-trichloro-5-iodo-pyrimidine 21.1 (Scheme 21).
- a suitable catalyst such as dichloro-bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- Cyclization of amino alkyne 21.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield 2,4-dichloro pyrrolo[2,3- djpyrimidines 21.4.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- a suitable solvent such as dioxane
- Scheme 20 can lead to 2-chloro-4-substituted-pyrrolo[2,3-d]pyrimidines 21.5, which can be functionalized according to Scheme 19 to lead to tetrasubstituted pyrrolo[2,3-d]pyrimidines 21.6.
- Scheme 22
- Selected examples from the invention described herein can be synthesized from optionally substituted 3-chloro-4-iodo-pyridine 22.1 (Scheme 22).
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- chloride 22.2 Treatment of chloride 22.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields pyrrolo[2,3-d]pyrimidines 22.3.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- Selected examples from the invention described herein can be synthesized from optionally substituted 2,3-dichloro-pyrazines 23.1 (Scheme 23).
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- chloride 23.2 Treatment of chloride 23.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 23.3.
- a catalyst such as tris(dibenzylideneacetone)dipalladium(0)
- a suitable ligand such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos)
- a suitable base such as cesium carbonate
- a suitable solvent such as dioxane
- Cyclization of amino alkyne 23.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[3,2-b]pyrazines 23.4.
- a base such as cesium carbonate and potassium fluoride
- a catalyst such as di(tri-tert-butylphosphine)palladium(0)
- Selected examples from the invention described herein can be synthesized from optionally substituted 2,4-dichloro-5-bromo-pyrimidine 24.1 (Scheme 24).
- a suitable catalyst such as dichloro- bis(triphenylphosphine)palladium (II)
- a suitable additive such as copper(I)iodide
- a base such as triethylamine
- a suitable transition metal catalyst e.g., palladium compounds.
- the coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines.
- Organic or inorganic bases e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates
- additives e.g., lithium chloride, copper halides or silver salts
- THF trifluorofuran
- dioxane dimethoxyethane
- diglyme diglyme
- dichloromethane dichloroethane
- acetonitrile acetonitrile
- DMF N-methylpyrrolidone
- water or mixtures of thereof
- the temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
- the Pd catalyzed coupling or SnAr reaction from 24.3 to 24.4 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 24 as well.
- Selected examples from the invention described herein can be synthesized from optionally substituted 4-chloro-5-iodo-pyrimidine 18.1 (Scheme 25).
- Selected examples from the invention described herein can be synthesized from optionally substituted iodoaniline 19.2 (Scheme 26).
- a suitable catalyst such as palladium(II) acetate
- a suitable additive such as lithium chloride
- a base such as potassium acetate
- a suitable solvent such as DMF
- Another aspect of this invention provides a method of preparing a compound of Formula 1, comprising the steps of reducing a compound having the formula:
- the reaction between the two compounds is an oxidative cyclization.
- Another aspect of this invention provides a method of preparing a compound of Formula 1, comprising the steps of reacting a compound having the formula:
- Het, R 1 and R 2 are as defined herein and X 2 is halo, e.g., chloride or bromide.
- Another aspect of this invention provides a method of preparing a compound of
- R 2 wherein Het, R 1 and R 2 are as defined herein and X 3 is a halogen atom, Pg is an amine protecting group, e.g., acetyl, trifluoroacetyl, BOC, CBz and Fmoc.
- Pg is an amine protecting group, e.g., acetyl, trifluoroacetyl, BOC, CBz and Fmoc.
- Suitable amino-protecting groups include acetyl, trifluoroacetyl, BOC, CBz and Fmoc. The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, supra.
- any of the synthetic methods for preparing compounds of Formula 1 it may be advantageous to separate reaction products from one another and/or from starting materials.
- the desired products of each step or series of steps is separated and/or purified to the desired degree of homogeneity by the techniques common in the art.
- separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography.
- Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
- SMB simulated moving bed
- Another class of separation methods involves treatment of a reaction mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like.
- reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like.
- the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.
- Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization.
- Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers.
- an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
- converting e.g., hydrolyzing
- some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
- Enantiomers can also be
- a single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. "Stereochemistry of Organic Compounds,” John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., (1975) 113(3):283-302).
- Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: “Drug Stereochemistry, Analytical Methods and Pharmacology,” Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
- diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl- ⁇ - phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
- the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
- the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (E. and Wilen, S., supra at p. 322).
- Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer.
- a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-)menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob III. J. Org. Chem., (1982) 47:4165), of the racemic mixture, and analyzing the l H NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers.
- chiral esters such as a menthyl ester, e.g., (-)menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob III. J. Org. Chem., (1982) 47:4165), of the racemic mixture, and analyzing the l H NMR spectrum for the presence of the
- Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse- phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 96/15111).
- a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase ("Chiral Liquid Chromatography” (1989) W. J. Lough, Ed., Chapman and Hall, New York; Okamoto, J. of Chromatogr., (1990) 513:375-378).
- Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
- the compounds of the present invention can be used as prophylactics or therapeutic agents for treating diseases or disorders mediated by modulation or regulation of AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases.
- AKT protein kinase mediated conditions that can be treated according to the methods of this invention include, but are not limited to, inflammatory, hyperproliferative cardiovascular, neurodegenerative, gynecological, and dermatological diseases and disorders.
- said pharmaceutical composition is for the treatment of hyperproliferative disorders, including cancers of the following categories: (1) Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; (2) Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma, non-small cell lung, small cell lung; (3) Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma,
- the AKT protein kinase-mediated condition is a hyperproliferative disease.
- the hyperproliferative disease is cancer.
- the cancer is selected from colon, ovarian, brain, lung, pancreatic, prostate, and gastric carcinomas.
- Compounds and methods of this invention can be also used to treat diseases and conditions such as rheumatoid arthritis, osteoarthritis, Chron's disease, angiofibroma, ocular diseases (e.g., retinal vascularisation, diabetic retinopathy, age-related macular degeneration, macular degeneration, etc.), multiple sclerosis, obesity, Alzheimer's disease, restenosis, autoimmune diseases, allergy, asthma, endometriosis, atherosclerosis, vein graft stenosis, peri- anastomatic prothetic graft stenosis, prostate hyperplasia, chronic obstructive pulmonary disease, psoriasis, inhibition of neurological damage due to tissue repair, scar tissue formation (and can aid in wound healing), multiple sclerosis, inflammatory bowel disease, infections, particularly bacterial, viral, retroviral or parasitic infections (by increasing apoptosis), pulmonary disease, neoplasm, Parkinson's disease, transplant rejection (as angiofibrom
- another aspect of this invention provides a method of treating diseases or medical conditions in a mammal mediated by AKT protein kinases, comprising administering to said mammal one or more compounds of Formula 1 or a pharmaceutically acceptable salt or prodrug thereof in an amount effective to treat or prevent said disorder.
- This invention also provides compounds of Formula 1 for use in the treatment of
- AKT protein kinase-mediated conditions AKT protein kinase-mediated conditions.
- An additional aspect of the invention is the use of a compound of Formula 1 in the preparation of a medicament for therapy, such as for the treatment or prevention of AKT protein kinase-mediated conditions.
- the compounds of the present invention can be used in combination with one or more additional drugs such as described below.
- the dose of the second drug can be appropriately selected based on a clinically employed dose.
- the proportion of the compound of the present invention and the second drug can be appropriately determined according to the administration subject, the administration route, the target disease, the clinical condition, the combination, and other factors.
- the second drug may be used in an amount of 0.01 to 100 parts by weight per part by weight of the compound of the present invention.
- the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of this invention such that they do not adversely affect each other.
- Such drugs are suitably present in combination in amounts that are effective for the purpose intended.
- another aspect of the present invention provides a composition comprising a compound of this invention in combination with a second drug, such as described herein.
- a compound of this invention and the additional pharmaceutically active drug(s) may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. Such sequential administration may be close in time or remote in time.
- the amounts of the compound of this invention and the second drug(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
- the combination therapy may provide "synergy” and prove “synergistic", i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
- a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
- a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes.
- an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
- a “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer, regardless of mechanism of action. Chemotherapeutic agents include compounds used in "targeted therapy” and conventional chemotherapy.
- chemotherapeutic agents include PLX4032 (Roche/Plexxikon),
- dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), mo holino-doxorubicin, cyanomo holino-doxorubicin, 2- pyrrolino-doxorubicin and deoxydoxorubicin), epigallocate, axoxo-L-norleucine
- chemotherapeutic agent also included in the definition of "chemotherapeutic agent” are: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LYl 17018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMA
- chemotherapeutic agent therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen pie), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
- therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RIT
- Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the PI3K inhibitors of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab,
- the compounds of the invention may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal. It will be appreciated that the preferred route may vary with for example the condition of the recipient. Where the compound is administered orally, it may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier or excipient. Where the compound is administered parenterally, it may be formulated with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form, as detailed below.
- a compound of this invention for the therapeutic treatment (including prophylactic treatment) of mammals including humans, it is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition.
- a pharmaceutical composition that comprises a compound of this invention.
- the pharmaceutical composition comprises a compound of Formula 1 in association with a pharmaceutically acceptable diluent or carrier.
- compositions of the invention are formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice.
- Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
- the therapeutically effective amount of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the disorder.
- the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
- composition for use herein is preferably sterile.
- formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished, for example, by filtration through sterile filtration membranes.
- the compound ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
- compositions of the compounds of the present invention may be prepared for various routes and types of administration.
- a compound of this invention having the desired degree of purity may optionally be mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation, a milled powder, or an aqueous solution.
- Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed.
- the pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8.
- Formulation in an acetate buffer at pH 5 is a suitable embodiment.
- the formulations may be prepared using conventional dissolution and mixing procedures.
- the bull- drug substance i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent) is dissolved in a suitable solvent in the presence of one or more excipients.
- solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
- safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water.
- Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof.
- Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine,
- the formulations may also include one or more stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
- stabilizing agents i.e., surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
- the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions.
- colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules
- a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as a compound of Formula 1 and, optionally, an additional therapeutic agent) to a mammal.
- a drug such as a compound of Formula 1 and, optionally, an additional therapeutic agent
- the components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes. See also, Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
- Sustained-release preparations of compounds of this invention may be prepared.
- sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula 1, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
- sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Patent No.
- copolymers of L-glutamic acid and gamma-ethyl-L- glutamate non-degradable ethylene-vinyl acetate
- degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate) and poly-D-(-)-3-hydroxybutyric acid.
- compositions of compounds of this invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
- a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
- This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
- the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared as a lyophilized powder.
- a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butanediol or prepared as a lyophilized powder.
- acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
- sterile fixed oils may conventionally be employed as a solvent or suspending medium.
- any bland fixed oil may be employed including synthetic mono- or diglycerides.
- fatty acids such as oleic acid may likewise be used in the preparation of injectables.
- Formulations suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
- compositions of the invention may also be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder)
- oral use for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs
- topical use for example as creams, ointments, gels, or aqueous or oily solutions or suspensions
- inhalation for example as a finely divided powder or a liquid aerosol
- Suitable pharmaceutically-acceptable excipients for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate, granulating and disintegrating agents such as corn starch or algenic acid; binding agents such as starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl p-hydroxybenzoate, and anti-oxidants, such as ascorbic acid. Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents and procedures well known in the art.
- inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate
- granulating and disintegrating agents such as corn starch or algenic acid
- binding agents such as starch
- compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin, or olive oil.
- an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
- soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin, or olive oil.
- Aqueous suspensions generally contain the active ingredient in finely powdered form together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate.
- suspending agents such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium al
- the aqueous suspensions may also contain one or more preservatives (such as ethyl or propyl p-hydroxybenzoate, anti-oxidants (such as ascorbic acid), coloring agents, flavoring agents, and/or sweetening agents (such as sucrose, saccharine or aspartame).
- preservatives such as ethyl or propyl p-hydroxybenzoate, anti-oxidants (such as ascorbic acid), coloring agents, flavoring agents, and/or sweetening agents (such as sucrose, saccharine or aspartame).
- Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil (such as arachis oil, olive oil, sesame oil or coconut oil) or in a mineral oil (such as liquid paraffin).
- the oily suspensions may also contain a thickening agent such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set out above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
- Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients such as sweetening, flavoring and coloring agents, may also be present.
- the pharmaceutical compositions of the invention may also be in the form of oil-in- water emulsions.
- the oily phase may be a vegetable oil, such as olive oil or arachis oil, or a mineral oil, such as for example liquid paraffin or a mixture of any of these.
- Suitable emulsifying agents may be, for example, naturally-occurring gums such as gum acacia or gum tragacanth, naturally-occurring phosphatides such as soya bean, lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides (for example sorbitan monooleate) and condensation products of the said partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate.
- the emulsions may also contain sweetening, flavoring and preservative agents.
- Syrups and elixirs may be formulated with sweetening agents such as glycerol, propylene glycol, sorbitol, aspartame or sucrose, and may also contain a demulcent, preservative, flavoring and/or coloring agent.
- sweetening agents such as glycerol, propylene glycol, sorbitol, aspartame or sucrose, and may also contain a demulcent, preservative, flavoring and/or coloring agent.
- Suppository formulations may be prepared by mixing the active ingredient with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
- suitable excipients include, for example, cocoa butter and polyethylene glycols.
- Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
- Topical formulations such as creams, ointments, gels and aqueous or oily solutions or suspensions, may generally be obtained by formulating an active ingredient with a conventional, topically acceptable, vehicle or diluent using conventional procedures well known in the art.
- compositions for transdermal administration may be in the form of those transdermal skin patches that are well known to those of ordinary skill in the art.
- Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs.
- Suitable formulations include aqueous or oily solutions of the active ingredient.
- Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis disorders as described below.
- the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
- an article for distribution can include a container having deposited therein the pharmaceutical formulation in an appropriate form.
- suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
- the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
- the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
- the formulations may also be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use.
- sterile liquid carrier for example water
- Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
- Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
- the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore.
- Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
- the amount of a compound of this invention that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the subject treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician.
- a suitable amount of a compound of this invention is administered to a mammal in need thereof.
- Administration occurs in an amount between about 0.001 mg/kg of body weight to about 60 mg/kg of body weight per day. In another embodiment, administration occurs in an amount between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
- dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several small doses for administration throughout the day.
- kits containing materials useful for the treatment of the disorders described above.
- the kit comprises a container comprising a compound of this invention.
- Suitable containers include, for example, bottles, vials, syringes, blister pack, etc.
- the container may be formed from a variety of materials such as glass or plastic.
- the container may hold a compound of this invention or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
- the kit may further comprise a label or package insert on or associated with the container.
- package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
- the label or package inserts indicates that the composition comprising a compound of this invention can be used to treat a disorder mediated, for example, by AKT kinase.
- the label or package insert may also indicate that the composition can be used to treat other disorders.
- kits are suitable for the delivery of solid oral forms of a compound of this invention, such as tablets or capsules.
- a kit preferably includes a number of unit dosages.
- Such kits can include a card having the dosages oriented in the order of their intended use.
- An example of such a kit is a "blister pack".
- Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms.
- a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered.
- a kit may comprise (a) a first container with a compound of this invention contained therein; and (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound useful for treating a disorder mediated by AKT kinase.
- the kit may further comprise a third container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
- BWFI bacteriostatic water for injection
- the kit may further comprise directions for the administration of the compound of this invention and, if present, the second pharmaceutical formulation.
- the kit may further comprise directions for the simultaneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof.
- the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet, however, the separate compositions may also be contained within a single, undivided container.
- the kit comprises directions for the administration of the separate components.
- the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
- kits for treating a disorder or disease mediated by Akt kinase comprising a) a first pharmaceutical composition comprising a compound of this invention or a pharmaceutically acceptable salt thereof; and b) instructions for use.
- the kit further comprises (c) a second pharmaceutical composition, wherein the second pharmaceutical composition comprises a second compound suitable for treating a disorder or disease mediated by Akt kinase.
- the kit further comprises instructions for the simultaneous, sequential or separate administration of said first and second pharmaceutical compositions to a patient in need thereof.
- said first and second pharmaceutical compositions are contained in separate containers. In other embodiments, said first and second pharmaceutical compositions are contained in the same container.
- the compounds of Formula 1 are primarily of value as therapeutic agents for use in mammals, they are also useful whenever it is required to control AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases. Thus, they are useful as pharmacological standards for use in the development of new biological tests and in the search for new pharmacological agents.
- the activity of the compounds of this invention may be assayed for AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases in vitro, in vivo, or in a cell line.
- In vitro assays include assays that determine inhibition of the kinase activity. Alternate in vitro assays quantitate the ability of the inhibitor to bind to kinases and may be measured either by radiolabelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where new inhibitors are incubated with known radioligands.
- the activity of the compounds described in the present invention may be determined by the following kinase assay, which measures the phosphorylation of a fluorescently- labeled peptide by full-length human recombinant active AKT-1 by fluorescent polarization using a commercially available IMAP kit.
- the assay materials are obtained from an IMAP AKT Assay Bulk Kit, product
- the kit materials include an IMAP Reaction Buffer (5x).
- the diluted lx IMAP Reaction Buffer contained 10 raM Tris-HCl, pH 7.2, 10 raM MgCl 2i 0.1% BSA. DTT is routinely added to a final concentration of 1 raM immediately prior to use.
- IMAP Binding Buffer (5x) is also included.
- the Binding Solution is prepared as a 1 :400 dilution of IMAP Binding Reagent into lx IMAP Binding Buffer.
- the fluorescein-labeled AKT Substrate has the sequence (Fl)-
- GRPRTSSFAEG A stock solution of 25 ⁇ is made up in lx IMAP Reaction Buffer.
- the plates used include a Costar 3657 (382-well made of polypropylene and having a white, v-bottom) that is used for compound dilution and for preparing the compound-ATP mixture.
- the assay plate is a Packard ProxiPlateTM-384 F.
- the AKT-1 used is made from full-length, human recombinant AKT-1 that is activated with PDK1 and MAP kinase 2.
- the assay is initiated by the addition of 2.5- ⁇ . aliquots of a solution containing 200 nM of fluorescently-labeled peptide substrate and 20 nM AKT-1. The plate is centrifuged for 1 minute at 1000 g and incubated for 60 minute at ambient temperature. The reaction is then quenched by the addition of 15 of Binding Solution, centrifuged again and incubated for an additional 30 minutes at ambient temperature prior to reading on a Victor 1420 Multilabel HTS Counter configured to measure fluorescence polarization.
- NMR spectra were obtained as CDC1 3 , CD 3 OD, D 2 0 or d6-DMSO solutions (reported in ppm), using tetramethylsilane (0.00 ppm) or residual solvent (CDC1 3 : 7.25 ppm; CD 3 OD: 3.31 ppm; D 2 0: 4.79 ppm; d6-DMSO: 2.50 ppm) as the reference standard.
- peak multiplicities the following abbreviations are used: s (singlet), d (doublet), t (triplet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, are reported in Hertz (Hz).
- Step A To a solution of 2,4-Dichloro-5-nitro-pyrimidine (5.00 g, 26.00mmol) in
- Step B ⁇ l-[4-(2-Chloro-5-nitro-pyrimidin-4-ylamino)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (4.10 g, 9.78 mmol) was dissolved in THF (200 mL) and Raney- Ni (1.00 g) was added. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 4 h. The solvent was removed under reduced pressure and the resulting residue was dissolved in ethyl acetate (200 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated.
- Step C To a solution of ⁇ l-[4-(5-Amino-2-chloro-pyrimidin-4- ylamino)-phenyl]- cyclo-butyl ⁇ -carbamic acid tert-butyl ester (2.5 g, 6.42 mmol) in acetic acid (30 mL) was added benzaldehyde (0.89 g, 8.35 mmol) and Cu(OAc) 2 (0.58 g, 3.21 mmol). The reaction mixture was stirred at 100 °C for 2 h.. Then the solvent was removed under reduced pressure. The resulting residue was dissolved in ethyl acetate (150 mL).
- Step A ⁇ l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (500 mg, 1.05 mmol), phenylboronic acid (256 mg, 2.10 mmol), Pd(dppf)Cl 2 (77 mg, 0.11 mmol), and Cs 2 C0 3 (684 mg, 2.10 mmol) were added into a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was stirred at 100 °C under nitrogen atmosphere for 4 h.
- Step B ⁇ l-[4-(2,8-Diphenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert- butyl ester (200 mg, 0.39 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Examples 2-4 shown in Table 1 can also be made according to the above-described methods.
- Step A To a solution of ⁇ l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclo- butylj-carbamic acid tert-butyl ester (70 mg, 0.147 mmol) in dioxane (20 mL) was added piperazine (63 mg, 0.737 mmol). Then the reaction mixture was stirred at 80 °C for 3h. The solvent was removed under reduced pressure, the resulting residue was dissolved in ethyl acetate (100 mL), washed with water (50 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated.
- Step B ⁇ l-[4-(8-Phenyl-2-piperazin-l-yl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (-65 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Example 6 shown in Table 2 can also be made according to the above-described methods.
- Step A To a solution of ⁇ l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclo- butyl ⁇ -carbamic acid tert-butyl ester (70 mg, 0.147 mmol) in methanol (20 mL) was added sodium methanolate (24 mg, 0.441 mmol). The reaction mixture was stirred at 80 °C for 3 h. Then the solvent was removed under reduced pressure, the resulting residue was dissolved in ethyl acetate (80 mL), washed with water (50 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated.
- Step B tert-butyl l-(4-(2-methoxy-8-phenyl-9H-purin-9-yl)phenyl)cyclo- butylcarbamate (-65 mg) was dissolved in ethyl acetate (20 mL), the solution was cooled to 0 °C and a solution of HCl in ethyl acetate (4 M, 5 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Example 8 shown in Table 3 can also be made according to the above-described methods.
- Step A ⁇ l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (50 mg, 0.11 mmol), pyrrolidin-2-one (14 mg, 0.16 mmol), Pd 2 (dba) 3 (19 mg, 0.02 mmol), Xantphos (15 mg, 0.03 mmol) and Cs 2 C0 3 (68 mg, 0.21 mmol) were dissolved in dioxane (10 mL). The reaction mixture was stirred at 90 °C under nitrogen atmosphere for 6 h.
- Step B (l- ⁇ 4-[2-(2-Oxo-pyrrolidin-l-yl)-8-phenyl-purin-9-yl]-phenyl ⁇ -cyclobutyl)- carbamic acid tert-butyl ester (30 mg, 0.057 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Step A To a solution of ⁇ 1 -[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (150 mg, 0.32 mmol) in dioxane (4 mL) was added the aqueous methylamine amine solution (10 mL, 28% wt) at a sealed vessel. The reaction mixture was stirred at 100 °C overnight. The reaction mixture was diluted with ethyl acetate (80 mL), and washed with water (40 mL). The separated organic layer was dried over sodium sulfate, filtered and
- Step B ⁇ l-[4-(2-Methylamino-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (-120 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- reaction mixture was allowed to be warmed to ambient temperature and stirred for another 2 h.
- the reaction mixture was poured into an aqueous saturated NH 4 CI solution, and extracted with ethyl acetate. The separated organic layer was washed with water twice and dried over sodium sulfate, filtered and concentrated to afford 2-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-4- (trimethylstannyl)-lH-imidazole (-420 mg), which was used in next step without further purification.
- Step A 3-methyl- l-((2-(trimethylsilyl)ethoxy)methyl)-l H-pyrazole (500 mg, 2.35 mmol) was dissolved in THF (18 mL), and the solution was cooled to -78°C, followed by adding butyllithium (2.5 M, 0.94 mL, 2.35 mmol) slowly. The reaction mixture was stirred at this temperature for 1 h. Then, chlorotrimethylstannane (1 M in THF, 2.4 mL, 2.35 mmol) was added into the reaction mixture. It was allowed to be warmed to ambient temperature and stirred overnight. The reaction mixture was poured into aqueous saturated NH 4 CI solution, and extracted with ethyl acetate.
- Step A To a solution of nitroethane (0.15 g, 2.00 mmol) in toluene (6 mL) was added l-chloro-3-isocyanatobenzene (0.61 g, 4.00 mmol). The reaction mixture was stirred at 50 °C for 10 minutes followed by addition of triethylamine (10 mg, 0.1 mmol) and tributyl(ethynyl)stannane (0.60 g, 1.91 mmol). The reaction mixture was stirred at 50 °C overnight. After cooling, the solution was diluted with water and filtered through a celite pad.
- Step A To a solution of ⁇ l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclo- butyl ⁇ -carbamic acid tert-butyl ester (150 mg, 0.316 mmol) in dioxane (4 mL) was added an 2- Methyl-l-(2-trimethylsilanyl-ethoxymethyl)-4-trimethylstannanyl-lH- imidazole (-0.632 mmol, in THF) and Pd(PPh 3 ) 4 (36 mg, 0.032 mmol). The reaction mixture was irradiated by microwave at 120 °C for 50 minutes.
- Step B [l-(4- ⁇ 2-[2-Methyl-3-(2-trimethylsilanyl-ethoxymethyl)-3H-imidazol-4- yl]-8-phenyl-purin-9-yl ⁇ -phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (-150 mg) was dissolved in Methanol (10 mL), and aqueous HC1 (36%, 5 mL) was added into the solution. It was heated at 80 °C for 2 h. The reaction mixture was concentrated under reduced pressure.
- Step A To a solution of ⁇ 1 -[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (250 mg, 0.526 mmol) in DMF (20 mL) were added methylamine hydrochloride (107 mg, 1.578 mmol), Pd(OAc) 2 (35 mg, 0.158 mmol), Xantphos (107 mg, 0.184 mmol) and sodium carbonate (178 mg, 1.683 mmol).
- Step B ⁇ l-[4-(2-Methylcarbamoyl-8-phenyl-purin-9-yl)-phenyl]-cyclo-butyl ⁇ - carbamic acid tert-butyl ester (-200 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h..
- Step A To a solution of ⁇ l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (250 mg, 0.526 mmol) in methanol (30 mL) was added Pd/C (200 mg, 10%wt). The mixture was stirred at 50 °C under the atmosphere of hydrogen (50 psi) for 12 h. The catalyst was removed by filtration, the cake was washed with methanol.
- Step B ⁇ l-[4-(8-Phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (-120 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Step A To a. solution of ⁇ 1 -[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (250 mg, 0.526 mmol) in THF (20 mL) was added pyridin-2- ylzinc(II) bromide (1.1 mL, 1M in THF) and Pd(PPh 3 ) 4 (36 mg, 0.032 mmol). The reaction mixture was stirred at 80 °C under the atmosphere of nitrogen for 4 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate, washed with water twice.
- Step B ⁇ l-[4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (-200 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Step B Compound ⁇ l-[4-(5-Nitro-2-phenyl-pyrimidin-4-ylamino)-phenyl]- cyclobutylj-carbamic acid tert-butyl ester (7.0 g, 15.18 mmol) was dissolved in THF (200 mL) and Raney- Ni (1.00 g) was added carefully. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 4 h.
- Step A ⁇ l-[4-(5-Amino-2-phenyl-pyrimidin-4-ylamino)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (0.25 g, 0.58 mmol) was dissolved in acetic acid (30 mL), then 4- methoxy-pyridine-3-carbaldehyde (87.7 mg, 0.64 mmol) and Cu(OAc) 2 (105 mg, 0.58 mmol) were added separately.
- Step B (l- ⁇ 4-[8-(4-Methoxy-pyridin-3-yl)-2-phenyl-purin-9-yl]-phenyl ⁇ - cyclobutyl)-carbamic acid tert-butyl ester (-150 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Examples 20 to 48 shown in Table 6 can also be made according to the above- described methods.
- Step A Disulfurdichloride (6.6 mL, 101 mmol) was added to a solution of 2- chloroacetonitrile (3.0 g, 40.2 mmol) in CH 2 C1 2 (10 mL) at room temperature, under an insert atomosphere of nitrogen, the resulting mixture was stirred at room temperature for 18 hours.the reaction mixture was concentrated in vacuo to get crude product 4,5-Dichloro-[l,2,3]dithiazol-l- ylium chloride (8 g crude) which was used without purification.
- Step B To a solution of ⁇ l-[4-(5-Amino-2-phenyl-pyrimidin-4-ylamino)-phenyl]- cyclobutyl ⁇ -carbamic acid tert-butyl ester (180mg, 0.414mmol) in THF (20ml) was added 4,5- Dichloro-[l,2,3]dithiazol-l-ylium chloride (99 mg, 0.45 mmol), then the reaction mixture was stirred at room temperature for 1 hour.
- Step C (l- ⁇ 4-[5-(4-Chloro-[l,2,3]dithiazol-5-ylideneamino)-2-phenyl-pyrimidin-4- ylamino]-phenyl ⁇ -cyclobutyl)-carbamic acid tert-butyl ester (150 mg, 0.27 mmol) was dissolved in toluene (15 mL), the reaction mixture was stirred at reflux for 4 hours. The reaction mixture was concentrated in vacuo to afford ⁇ l-[4-(8-Cyano-2-phenyl-purin-9-yl)-phenyI]-cyclobutyl ⁇ -carbamic acid tert-butyl ester.
- Step D The compound ⁇ l-[4-(8-Cyano-2-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (80-160 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, dried in vacuo, to give target compounds as HCl salt. (15.4 mg)
- Step A To a solution of compound ⁇ l-[4-(5-Amino-2-chloro-pyrimidin-4- ylamino)-phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (0.3 g, 0.77 mmol) in dioxane (20 mL)/H 2 0 (4 ml) were added PhB(OH) 2 (2.31 mmol), Pd(dppf)Cl 2 (56.29 mg, 0.077 mmol), and Cs 2 C0 3 (501mg, 1.54mmol). The reaction mixture was stirred at 90 °C under nitrogen atmosphere for 3 h.
- Step B (l- ⁇ 4-[5-Amino-2-(3-trifluoromethoxy-phenyl)-pyrimidin-4-yl-amino]- phenyl ⁇ -cyclobutyl)-carbamic acid tert-butyl ester (200 mg 0.77 mmol) was dissolved in acetic acid (30 mL), then 2-Methyl-2H-pyrazole-3-carbaldehyde (93.5 mg, 0.85 mmol) and Cu(OAc) 2 (140 mg, 0.77 mmol) were added separately. The reaction mixture was stirred at 100 °C for 2 h.
- Step C (l- ⁇ 4-[2-(2-Fluoro-phenyl)-8-(2-methyl-2H-pyrazol-3-yl)-purin-9-yl]- phenyl ⁇ -cyclobutyl)-carbamic acid tert-butyl ester (150 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.
- Step A The tert-butyl l-(4-(5-amino-2-chloropyrimidin-4-ylamino)- phenyl)cyclobutylcarbamate (3.00 g, 7.71 mmol) was dissolved in acetic acid (50 mL), then picolinaldehyde (1.23 g, 11.5 mmol) and Cu(OAc) 2 (1.4 g, 7.71 mmol) were added separately. The reaction mixture was stirred at 80 °C for 2 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with aqueous sat.
- Step A tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)- cyclobutylcarbamate (100 mg, ) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HCl in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h..
- Step A Tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)- phenyl)cyclobutylcarbamate (700 mg, 1.47 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- lH-pyrazole (342 mg, 1.76 mmol), Pd(dppf)Cl 2 (109.5 mg, 0.15 mmol), and Cs 2 C0 3 (1.4 g, 4.41 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL).
- Step B The tert-butyl l-(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9- yl)phenyl)cyclobutylcarbamate (700 mg crude) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HCl in ethyl acetate (4 M, 10 mL) was added slowly. The resulting solution was stirred for 2 h.
- Examples 59 and 60 shown in Table 8 can also be made according to the above- described methods.
- Step A A solution of tert-butyl 1 -(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H- purin-9-yl)phenyl)cyclobutylcarbamate (400 mg, 0.787 mmol) in dioxane(10 mL) and oxirane (10 mL) was stirred at 10 °C for 3 days..
- Step B tert-butyl 1 -(4-(2-( 1 -(2-hydroxyethyl)- 1 H-pyrazol-4-yl)-8-(pyridine-2-yl)-
- Step A A mixture of tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9- yl)phenyl) cyclobutylcarbamate (100 mg, 0.21 mmol) in ethane- 1,2-diamine (1 mL) was put in a sealed tube and stirred at 100 °C for 2 h. The reaction mixture was concentrated and the resulting residue was dissolved in ethyl acetate (50 mL), washed with water. The separated organic layer was dried over sodium sulfate, filtered and concentrated.
- Step A The compound ⁇ l-[4-(2-Chloro-8-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutyl ⁇ - carbamic acid tert-butyl ester (200 mg, 0.42 mmol), pyrrolidin-3-ol (103 mg, 0.84 mmol) and K 2 C0 3 (176 mg, 0.84 mmol) were dissolved in l-methyl-pyrrolidin-2-one (10 mL). The reaction mixture was stirred at 120 °C under nitrogen atmosphere overnight. The mixture was diluted with ethyl acetate (50 mL) and washed with brine (20 mL).
- Step B (l- ⁇ 4-[2-(3-Hydroxy-pyrrolidin-l-yl)-8-pyridin-2-yl-purin-9-yl]-phenyl ⁇ - cyclobutyl)-carbamic acid tert-butyl ester was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4M, 10 mL) was added slowly. The resulting solution was stirred for 2 hrs.
- Step A The compound ⁇ l-[4-(2-Chloro-8-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutyl ⁇ -carbamic acid tert-butyl ester (150 mg, 0.315 mmol), Methanesulfonamide (60 mg, 0.63 mmol), Pd 2 (dba) 3 (29 mg, 0.032 mmol), Xantphos (18 mg, 0.032 mmol) and Cs 2 C0 3 (308 mg, 0.945 mmol) were dissolved in dioxane (10 mL). The reaction mixture was stirred at 100 °C under nitrogen atmosphere for overnight. LC-MS indicated the reaction worked well.
- Step B ⁇ l-[4-(2-Methanesulfonylamino-8-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutyl ⁇ -carbamic acid tert-butyl ester (107 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4M, 10 mL) was added slowly. The resulting solution was stirred for 2 hrs.
- Step C To a solution of tert-butyl l-(4-(5-amino-6- chloropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (200mg, 0.5mmol) in HOAc (20 mL) was added Cu(OAc) 2 46.7mg, 0.26mmol and benzaldehyde (109mg, lmmol). The reaction mixture was stirred at 80°C for lh.
- Step D The solution of tert-butyl l-(4-(6-chloro-8-phenyl-9H-purin-9-yl) phenyl)cyclobutylcarbamate (40mg, 0.08mmol) in HC1 (2 mL, 10% wt) was stirred at 80 °C for lh. The reaction solution was concentrated to give title compound 9-(4-(l-aminocyclobutyl)phenyl)-8- phenyl-lH-purin-6(9H)-one (30mg) as HC1 salt.
- Example 66 shown in Table 9 can also be made according to the above-described methods.
- Step A To the solution of 4, 6-Dichloro-5-nitro-pyrimidine (1 g, 5.2 mmol) in THF
- Step B To a solution of ⁇ l-[4-(6-Chloro-5-nitro-pyrimidin-4-ylamino)- phenyl]- cyclobutyl ⁇ -carbamic acid tert-butyl ester (2 g, 4.77 mmol) and dimethyl-amine (0.77 g, 9.54 mmol) in THF (50 mL) was added DIPEA (3.1 g, 23.9 mmol). The mixture was stirred 20 °C for 2 h. LC-MS showed the starting material was consumed completely. The reaction mixture was diluted with ethyl acetate (30 mL) and washed with brine (2 X 20 mL).
- Step C The compound ⁇ l-[4-(6-Dimethylamino-5-nitro-pyrimidin-4-ylamino)- phenyl]-cyclobutyl ⁇ -carbamic acid tert-butyl ester (2.0 g, 4.67 mmol) was dissolved in THF (60 mL) and Raney- Ni (2.00 g) was carefully added. The reaction mixture was stirred at ambient temperature under the atmosphere of H 2 (balloon) for 1 h..
- Step D The ⁇ l-[4-(5-Amino-6-dimethylamino-pyrimidin-4-ylamino)-phenyl]- cyclo-butyl ⁇ -carbamic acid tert-butyl ester (500 mg, 1.29 mmol) was dissolved in acetic acid (20 mL), and benzaldehyde (204 mg, 1.93 mmol) and Cu(OAc) 2 (233 mg, 1.29 mmol) were added separately. The reaction mixture was heated at 80°C for 2 hrs.
- Step E ⁇ l-[4-(6-Dimethylamino-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester (300 mg, 0.62 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0°C and the solution of HC1 in ethyl acetate (10 mL, 4M) was slowly added. The resulting mixture was stirred for 2 hrs.
- Step A To a solution of 4,6-dichloro-5-nitropyrimidine (2.00 g, 10.3 mmol) in THF
- Step B The tert-butyl l-(4-(6-chloro-5-nitropyrimidin-4-ylamino)phenyl)- cyclobutylcarbamate (1.5 g, 3.58 mmol) was dissolved in THF (50 mL) and Raney- Ni (1.50 g) was carefully added. The reaction mixture was stirred at 20°C under the atmosphere of H 2 (balloon) for 1 h.. After filtration ciliate pad, the filtrate was concentrated to give tert-butyl l-(4-(5-amino-6- chloropyrimidin-4-ylamino)phenyl)cyclobutylcarbamate. (1.3 g ,93% yield:). MS (ESI + ): 412.1 [M+Na] + , 414.1 [M+Na+2] +
- Step C The tert-butyl l-(4-(5-amino-6-chloropyrimidin-4-ylamino)phenyl)- cyclobutylcarbamate (200 mg, 0.573 mmol), l-Methylpyrazole-4-boronic acid pinacol ester (143 mg, 0.688 mmol), Pd(dppf)Cl 2 (40 mg, 0.057 mmol), and Cs 2 C0 3 (560 mg, 1.72 mmol) were dissolved in a mixed solution (dioxane: 10 mL, water: 2 mL). The reaction mixture was charged with nitrogen thrice and heated at 80°C for 2 h.
- Step D The compound tert-butyl l-(4-(5-amino-6-(l -methyl- lH-pyrazol-4- yl)pyrimidin-4-ylamino)phenyl)cyclobutylcarbamate (100 mg, 0.23 mmol) was dissolved in acetic acid (10 mL), benzaldehyde (36 mg, 0.34 mmol) and Cu(OAc) 2 (42 mg, 0.23 mmol) were added separately. The reaction mixture was heated at 80°C for 2 h.
- Step E The tert-butyl 1 -(4-(6-( 1 -methyl- 1 H-pyrazol-4-yl)-8-phenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate (115 mg, 0.22 mmol) was dissolved in ethyl acetate (20 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (10 mL, 4M) was slowly added. The resulting solution was stirred for 2 h.
- Step A To the solution of 2,4-Dichloro-6-methyl-5-nitro-pyrimidine (2.08 g, lO.OOmmol) in THF (50 mL) at -78 °C was slowly added the solution of [l-(4-Amino-phenyl)- cyclobutyl]-carbamic acid tert-butyl ester (2.62 g, 10.00 mmol) in THF (20 mL). The reaction mixture was stirred at ambient temperature for 30 min.. Then it was concentrated to remove the solvent. The obtained residue was dissolved in ethyl acetate (250 mL) and washed by saturated. NaHC0 3 solution, followed by brine.
- Step B ⁇ l-[4-(2-Chloro-6-methyl-5-nitro-pyrimidin-4- ylamino)-phenyl]- cyclobutyl ⁇ -carbamic acid tert-butyl ester (4.3 g, 9.78 mmol) was dissolved in MeOH (100 mL) and Pd/C (1.00 g) was added. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 12 h.. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (200 mL).
- Step C The ⁇ l-[4-(5-Amino-6-methyl-pyrimidin-4-ylamino)-phenyl]- cyclobutyl ⁇ - carbamic acid tert-butyl ester (2.0 g, 5.4 mmol) was dissolved in acetic acid (20 mL), and benzaldehyde (0.74 g, 7 mmol) and Cu(OAc) 2 (1.0 g, 5.4 mmol) were added separately. The reaction mixture was heated at 80 °C for 2 h.
- Step D ⁇ l-[4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl ⁇ - carbamic acid tert-butyl ester ( 0.3 g ) was dissolved in MeOH (30 mL), then added HC1 (aq) (4M, 8 mL) in drops. The mixture was heated to 80 °C, and stirred for 50 min. The product was purified by preparative HPLC to give (160 mg).
- Step A To a suspension of 2,4,6-trichloro-5-nitropyrimidine (0.8g, 3.5mmol) in
- Step B To a solution of tert-butyl l-(4-(2,6-dichloro-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (0.45 g, 1 mmol) in THF (20ml) was added MeONa (53 mg, 1 mmol) in MeOH(l mL). The mixture was stirred at 55 °C for 30mins. The reaction mixture was purified by preparative TLC to tert-butyl l-(4-(2-chloro-6-methoxy-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (120 mg, 26.6% yield). LC/MS: (ESI + ) e/z: 472 [M+Na] + , 474 [M+Na+2] +
- Step C To a solution of tert-butyl l-(4-(2-chloro-6-methoxy-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (100 mg, 0.222 mmol) in THF (10 mL) was added Raney Ni (50 mg) under nitrogen. The suspension was degassed under vacuum and purged with H 2 (balloon). The reaction mixture was stirred at RT for 10 mins.
- Step D To a solution of tert-butyl l-(4-(5-amino-2-chloro-6-methoxypyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (84 mg, 0.2 mmol) in AcOH(5 mL), Benzaldehyde(42.45 mg, 0.4 mmol) and Cu(OAc)2(18.2 mg, 0.1 mmol) was added. The reaction mixture was stirred at 70C for 30 mins.
- Step E To a solution of tert-butyl l-(4-(2-chloro-6-methoxy-8-phenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate(15 mg, 0.03 mmol) in a mixture of Dioxane(5 mL) and H 2 O(0.5 mL), Pd(PPh 3 ) 4 (3 mg,0.0026 mmol), phenylboronic acid (54.87 mg,0.45 mmol) and Cs 2 C0 3 (29.3 mg, 0.09 mmol) was added. The reaction mixture was stirred under nitrogen at 80 °C for lh.
- Step F A solution of tert-butyl l-(4-(6-methoxy-2,8-diphenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate (10 mg) in HCl (5 mL, conc.).was stirred at 80 °C for 30 mins. The mixture was purified by preparative TLC to give 9-(4-(l-aminocyclobutyl)phenyl)-2,8-diphenyl- lH-purin-6(9H)-one (3 mg).
- Step A To a solution of 2-chloro-6-methyl-3-nitropyridine (600 mg, 3.5 mmol) and tert-butyl l-(4-aminophenyl)-cyclobutylcarbamate in THF (20 mL) was added DIPEA (900 mg, 7 mmol). The reaction mixture was stirred at 70°C for 5 h.
- Step B The tert-butyl l-(4-(6-methyl-3-nitropyridin-2-ylamino)phenyl)- cyclobutylcarbamate (550 mg, 1.38 mmol) was dissolved in THF (50 mL) and Raney- Ni (1.00 g) was added. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 1 h. The suspension was filtered through a pad of Celite and the pad was withed with THF (20 mL).
- Step C The tert-butyl l-(4-(3-amino-6-methylpyridin-2-ylamino)phenyl)- cyclobutylcarbamate (300 mg, 0.81 mmol) was dissolved in acetic acid (50 mL), and benzaldehyde (129 mg, 1.22 mmol) and Cu(OAc) 2 (76 mg, 0.42 mmol) were added separately. The reaction mixture was stirred at 100 °C for 2 h. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (50 mL). The organic layer was washed by saturated.
- Step D The tert-butyl l-(4-(5-methyl-2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutylcarbamate (238 mg, 0.52 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0°C and the solution of HC1 in ethyl acetate (10 mL, 4M) was slowly added. The resulting solution was stirred at 20 °C for 20 min.
- Step A A mixture of 5-bromo-2-chloro-3-nitropyridine (1 g, 4 mmol) and tert- butyl l-(4-aminophenyl)cyclobutylcarbamate (1.04 g, 4 mmol) in dioxane (30 mL) was added TEA (2ml) in one portion. Then the reaction mixture was warmed to 70 °C and stirred overnight.
- Step B tert-butyl l-(4-(5-bromo-3-nitropyridin-2-ylamino)phenyl)cycIobutyl carbamate (1 g, 2 mmol) was dissolved in THF (50mL) and treated with the catalytical amount of Raney Ni(0.2 g), then the mixture was stirred under hydrogen (balloon) at room temperature for 30 minutes.
- Step C benzaldehyde (0. 6 g, 1.3 mmol) and Cu(OAc) 2 (158 mg, 0.13 mmol) was added to a solution of tert-butyl l-(4-(3-amino-5-bromopyridin-2- ylamino)phenyl)cyclobutylcarbamate (1 g, 2.87 mmol) in glacial acetic acid (20 mL). The reaction mixture was stirred at 70 °C for lh. then the mixture was diluted with water and basified with saturate sodium bicarbonate and then extracted with ethyl acetate for three times.
- Step D To a solution of tert-butyl l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (0.3 g, 0.5 mmol) in EtOAc was added the solution of HC1 in EtOAc (4 mol/L , 10 ml) at 0 °C dropwise. The reaction mixture was stirred at room temperature for 1 h.
- Step A A mixture of l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutanamine (0.8 mmol), 2-fluorophenyl boronic acid (0.96 mmol), Cs 2 C0 3 (521 mg, 2.4 mmol) and Pd(dppf)Cl 2 (116 mg, 0.16 mmol) in dioxane and H 2 0 (10 ml, 5:1) was stirred at 80 °C for 2 hours under nitrogen atmosphere.
- Step A A solution of tert-butyl l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (80 mg) in MeOH (10 mL) was stirred under hydrogen (balloon) atmosphere in presence of Raney Ni (100 mg) at room temperature for 10 min. The reaction mixture was filtered through celite pad and the filtrate was concentrated to give 60 mg of crude desired compound tert-butyl l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutylcarbamate, which was used directly without further purification.
- Step B To a solution of t tert-butyl l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyI)cyclobutylcarbamate (60 mg crude) in EtOAc was added the solution of HCl in EtOAc (4 mol/L , 10 ml) at 0 °C dropwise. The reaction mixture was stirred at room temperature for 1 h.
- Step A 2,3-Dichloro-pyrazine (14.8 g, 0.1 mol) was added in a 100 mL autoclave which contained 50 mL of the solution of N3 ⁇ 4 aqueous with stirring, and then the reaction mixture was warmed to 100°C and stirred overnight, Cooled to room temperature. Filtrated, the solid was washed with water and further with dichloromethane to afford a solid, which dried on vacuum to afford 3-Chloro-pyrazin-2-ylamine. (10 g, 80% yield).
- Step B The solution of dimethyl sulfoxide (4 g, 0.05 mmol) in 30mL of dry dichloromethane was cooled to -78°C, then (13 g, 0.046 mmol) trifluoromethanesulfonic anhydride was added dropwise under nitrogen with stirring.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention provides compounds, including resolved enantiomers, resolved diastereomers, solvates and pharmaceutically acceptable salts thereof, comprising the Formula 1: wherein Het, X, R1 and R2 are as defined herein.
Description
BICYCLIC HETEROARYL DERIVATIVES AS KINASE INHIBITORS
BACKGROUND OF THE INVENTION
Field of the Invention
[0001] This invention relates to organic compounds useful for therapy and/or prophylaxis in a mammal, and in particular as inhibitors of serine/threonine protein kinases (e.g., AKT and related kinases), pharmaceutical compositions containing the inhibitors, and methods for preparing these inhibitors. The inhibitors are useful, for example, for the treatment of hyperproliferative diseases, such as cancer and inflammation, in mammals.
Description of the State of the Art
[0002] Protein kinases (PK) are enzymes that catalyze the phosphorylation of hydroxy groups on tyrosine, serine and threonine residues of proteins by transfer of the terminal (gamma) phosphate from ATP. Through signal transduction pathways, these enzymes modulate cell growth, differentiation and proliferation, i.e., virtually all aspects of cell life in one way or another depend on PK activity (Hardie, G. and Hanks, S. (1995) The Protein Kinase Facts Book. I and II, Academic Press, San Diego, CA). Furthermore, abnormal PK activity has been related to a host of disorders, ranging from relatively non-life threatening diseases, such as psoriasis, to extremely virulent diseases, such as glioblastoma (brain cancer). Protein kinases are an important target class for therapeutic modulation (Cohen, P. (2002) Nature Rev. Drug Discovery 1 :309).
[0003] Significantly, atypical protein phosphorylation and/or expression is often reported to be one of the causative effects of abnormal cellular proliferation, metastasis and cell survival in cancer. The abnormal regulation and/or expression of various kinases, including Akt, VEGF, ILK, ROCK, p70S6K, Bel, PKA, PKC, Raf, Src, PDK1, ErbB2, MEK, IKK, Cdk, EGFR, BAD, CHKl, CHK2 and GSK3 amongst numerous others, has been specifically implicated in cancer.
[0004] Protein kinases include two classes; protein tyrosine kinases (PTK) and serine- threonine kinases (STK). The Protein Kinase B/Akt enzymes are a group of serine/threonine kinases that are overexpressed in a variety of human tumors. One of the best-characterized targets of the PI3K lipid products is the 57 KD serine/threonine protein kinase Akt, downstream of PI3K in the signal transduction pathway (Hemmings, B.A. (1997) Science 275:628; Hay N. (2005) Cancer Cell 8:179-183). Akt is the human homologue of the protooncogene v-akt of the acutely transforming retrovirus AKT8. Due to its high sequence homology to protein kinases A and C, Akt is also called Protein Kinase B (PKB) and Related to A and C (RAC). Three isoforms of Akt are known to exist, namely Aktl, Akt2 and Akt3, which exhibit an overall homology of 80% (Staal,
S.P. (1987) Proc. Natl. Acad. Sci. 84:5034; Nakatani, K. (1999) Biochem. Biophys. Res. Commun. 257:906; Li et al (2002) Current Topics in Med. Chem. 2:939-971 ; WO 2005/1 13762). The Akt isoforms share a common domain organization that consists of a pleckstrin homology domain at the N-terminus, a kinase catalytic domain, and a short regulatory region at the C-terminus. In addition, both Akt2 and Akt3 exhibit splice variants. Upon recruitment to the cell membrane by PtdInd(3,4,5)P3, Akt is phosphorylated (activated) by PDK1 at T308, T309 and T305 for isoforms Aktl (PKBa), Akt2 (ΡΚΒβ) and Akt3 (ΡΚΒγ), respectively, and at S473, S474 and S472 for isoforms Aktl, Akt2 and Akt3, respectively. Such phosphorylation occurs by an as yet unknown kinase (putatively named PDK2), although PDK1 (Balendran, A., (1999) Curr. Biol. 9:393), autophosphorylation (Toker, A. (2000) J. Biol. Chem. 275:8271) and integrin-linked kinase (ILK) (Delcommenne, M. (1998) Proc. Natl. Acad. Sci. USA, 95:11211) have been implicated in this process. Akt activation requires its phosphorylation on residue Ser 473 in the C-terminal hydrophobic motif (Brodbeck et al (1999) J. Biol. Chem. 274:9133-9136; Coffer et al (1991) Eur. J. Biochem. 201 :475-481; Alessi et al (1997) Curr. Biol. 7:261-269). Although monophosphorylation of Akt activates the kinase, bis(phosphorylation) is required for maximal kinase activity.
[0005] Akt is believed to assert its effect on cancer by suppressing apoptosis and enhancing both angiogenesis and proliferation (Toker et al. (2006) Cancer Res. 66(8):3963-3966). Akt is overexpressed in many forms of human cancer including, but not limited to, colon (Zinda et al (2001) Clin. Cancer Res. 7:2475), ovarian (Cheng et al (1992) Proc. Natl. Acad. Sci. USA 89:9267), brain (Haas Kogan et al (1998) Curr. Biol. 8:1195), lung (Brognard et al (2001) Cancer Res. 61 :3986), pancreatic (Bellacosa et al (1995) Int. J. Cancer 64:280-285; Cheng et al (1996) Proc. Natl. Acad. Sci. 93:3636-3641), prostate (Graff et al (2000) J. Biol. Chem. 275:24500) and gastric carcinomas (Staal et al (1987) Proc. Natl. Acad. Sci. USA 84:5034-5037).
[0006] The PI3K/Akt/mammalian target of rapamycin (mTOR) pathway has been explored for targeted small molecule inhibitor therapy (Georgakis, G. and Younes, A. (2006) Expert Rev. Anticancer Ther. 6(1): 131-140; Granville et al (2006) Clin. Cancer Res. 12(3):679-689). Inhibition of PI3K/Akt signaling induces apoptosis and inhibits the growth of tumor cells that have elevated Akt levels (Kim et al (2005) Current Opinion in Investig. Drugs 6(12): 1250- 1258; Luo et al (2005) Molecular Cancer Ther. 4(6):977-986).
[0007] The development of kinase inhibitors that target abnormally regulated pathways and ultimately result in disease is of enormous ethical and commercial interest to the medical and pharmaceutical community. A compound that inhibits (1) recruitment of Akt to the cell membrane, (2) activation by PDK1 or PDK2, (3) substrate phosphorylation, or (4) one of the downstream
targets of Akt could be a valuable anticancer agent, either as a stand-alone therapy or in conjunction with other accepted procedures.
SUMMARY OF THE INVENTION
[0008] This invention provides novel compounds that inhibit AKT protein kinases. The compounds of the present invention have utility as therapeutic agents for diseases and conditions that can be treated by the inhibition of AKT protein kinases.
[0009] More specifically, the present invention includes compounds having the general
Formula 1 :
and tautomers, resolved enantiomers, resolved diastereomers, solvates, metabolites, salts and
1 2
pharmaceutically acceptable prodrugs thereof, wherein Het, X, R and R are as defined herein.
[0010] The invention also provides pharmaceutical compositions comprising a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof.
[0011] In a further aspect, the present invention provides a method of treating diseases or medical conditions in a mammal mediated by AKT protein kinases, comprising administering to said mammal one or more compounds of Formula 1 , or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in an amount effective to treat or prevent said disorder. AKT protein kinase mediated conditions that can be treated according to the methods of this invention include, but are not limited to, inflammatory, hyperproliferative, cardiovascular, neurodegenerative, gynecological, and dermatological diseases and disorders.
[0012] In a further aspect, the present invention provides a method of inhibiting the production of AKT protein kinases in a mammal, which comprises administering to said mammal a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof in an amount effective to inhibit production of an AKT protein kinase.
[0013] In a further aspect, the present invention provides methods of inhibiting the activity of AKT protein kinases, comprising contacting said kinase with a compound of Formula 1.
[0014] The inventive compounds may be used advantageously in combination with other known therapeutic agents. Accordingly, this invention also provides pharmaceutical compositions
comprising a compound of Formula 1 or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, in combination with a second therapeutic agent.
[0015] This invention also provides compounds of Formula 1 and enantiomers, solvates, metabolites, and pharmaceutically acceptable salts and prodrugs thereof for use as medicaments in the treatment of AKT protein kinase-mediated conditions.
[0016] An additional aspect of the invention is the use of a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, for therapy. In one embodiment, the therapy comprises the treatment of an AKT protein kinase-mediated condition.
[0017] This invention further provides kits for the treatment of an AKT protein kinase- mediated disease or disorder, said kit comprising a compound of Formula 1, or an enantiomer, solvate, metabolite, or pharmaceutically acceptable salt or prodrug thereof, a container, and optionally a package insert or label indicating a treatment. The kits may further comprise a second compound or formulation comprising a second pharmaceutical agent useful for treating said disease or disorder.
[0018] This invention further includes methods of preparing, methods of separating, and methods of purifying of the compounds of this invention.
[0019] Additional advantages and novel features of this invention shall be set forth in part in the description that follows, and in part will become apparent to those skilled in the art upon examination of the following specification, or may be learned by the practice of the invention. The advantages of the invention may be realized and attained by means of the instrumentalities, combinations, compositions, and methods particularly pointed out in the appended claims.
DETAILED DESCRIPTION OF THE INVENTION
[0020] Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents which may be included within the scope of the present invention as defined by the claims. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described. In the event that one or more of the incorporated literature and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls.
DEFINITIONS
[0021] The term "alkyl" as used herein refers to a saturated linear or branched-chain monovalent hydrocarbon radical of one to twelve carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1 -propyl (n- Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CH3)2), 1 -butyl (n-Bu, n-butyl, - CH2CH2CH2CH3), 2-methyl-l -propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, - CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, - CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-methyl-2- butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2), 3-methyl-l -butyl (-CH2CH2 CH(CH3)2), 2-methyl-l -butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2- hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (- C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3),4-methyl-2-pentyl (- CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (-C(CH3)(CH2CH3)2),2-methyl-3-pentyl (- CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (-C(CH3)2CH(CH3)2),3,3-dimethyl-2-butyl (- CH(CH3)C(CH3)3, 1-heptyl, 1-octyl, and the like.
[0022] The term "alkylene" or "alkylenyl" as used herein refers to a linear or branched saturated divalent hydrocarbon radical of one to twelve carbon atoms, wherein the alkylene radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, methylene, ethylene, propylene, 2-methylpropylene, pentylene, and the like.
[0023] The terms "cycloalkyl," "carbocycle," "carbocyclyl" and "carbocyclic ring" as used herein are used interchangeably and refer to saturated or partially unsaturated cyclic hydrocarbon radical having from three to twelve carbon atoms. The term "cycloalkyl" includes monocyclic and polycyclic (e.g., bicyclic and tricyclic) cycloalkyl structures, wherein the polycyclic structures optionally include a saturated or partially unsaturated cycloalkyl ring fused to a saturated, partially unsaturated or aromatic cycloalkyl or heterocyclic ring. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like. Bicyclic carbocycles include those having 7 to 12 ring atoms arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, and bicyclo[3.2.2]nonane. The cycloalkyl may be optionally substituted independently with one or more substituents described herein.
[0024] "Aryl" as used herein means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent
aromatic ring system. Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Exemplary aryl groups include, but are not limited to, radicals derived from benzene, naphthalene, anthracene, biphenyl, indene, indane, 1 ,2-dihydronapthalene, 1,2,3,4-tetrahydronapthalene, and the like. Aryl groups may be optionally substituted independently with one or more substituents described herein.
[0025] The terms "heterocycle", "hetercyclyl", "heterocycloalkyl" and "heterocyclic ring" as used herein are used interchangeably and refer to a saturated or partially unsaturated carbocyclic radical of 3 to 8 ring atoms in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen and sulfur, the remaining ring atoms being C, where one or more ring atoms may be optionally substituted independently with one or more substituents described below. The radical may be a carbon radical or heteroatom radical. The term "heterocycle" includes heterocycloalkoxy. "Heterocyclyl" also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated, or aromatic carbocyclic or heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinylimidazolinyl, imidazolidinyl, 3-azabicyco[3.1.OJhexanyl, 3-azabicyclo[4.1.Ojheptanyl, azabicyclo[2.2.2]hexanyl, 3H-indolyl quinolizinyl and N-pyridyl ureas. Spiro moieties are also included within the scope of this definition. The heterocycle may be C-attached or N-attached where such is possible. For instance, a group derived from pyrrole may be pyrrol- 1-yl (N-attached) or pyrrol-3-yl (C-attached). Further, a group derived from imidazole may be imidazol-l-yl (N-attached) or imidazol-3-yl (C- attached). Examples of heterocyclic groups wherein 2 ring carbon atoms are substituted with oxo (=0) moieties are isoindoline-l,3-dionyl and 1,1-dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described herein. Non limiting examples of 5 to 6 membered heterocycloalkyl groups containing one, two or three heteroatoms selected from the group consisting of N, S and O can be selected from the group consisting of optionally substituted pyrrolidinyl, piperazinyl, pyrrolidinyl-2-one, thiomorpholine and 1,1 dioxide.
[0026] The term "heteroaryl" as used herein refers to a monovalent aromatic radical of a 5-,
6-, or 7-membered ring and includes fused ring systems (at least one of which is aromatic) of 5-10 atoms containing at least one heteroatom independently selected from nitrogen, oxygen, and sulfur.
Examples of heteroaryl groups include, but are not limited to, pyridinyl, imidazolyl, imidazopyridinyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. Spiro moieties are also included within the scope of this definition. Heteroaryl groups may be optionally substituted independently with one or more substituents described herein. 5 or 6 membered heteroaryl can be selected from the group consisting of optionally substituted pyridinyl, pyrimidinyl, thiazolyl, imidazolyl, oxazolyl, isoxazolyl, pyrazolyl, pyridinone and benzimidazolyl.
[0027] By way of example and not limitation, carbon bonded heterocycles and heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline. Further examples of carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5- pyridazinyl, 6-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl.
[0028] By way of example and not limitation, nitrogen bonded heterocycles and heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of an isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline. Still more typically, nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, 1 -pyrrolyl, 1- imidazolyl, 1 -pyrazolyl, and 1-piperidinyl.
[0029] The term "halogen" or "halo" as used herein means fluoro, chloro, bromo or iodo.
[0030] The term "haloalkoxyl" as used herein refers to an alkoxyl group as defined herein that is substituted by 1, 2, 3, 4, 5 or 6 halogen as defined herein. Examples of haloalkoxyl groups include, but are not limited to Ci-C6-haloalkoxyl groups, including trifluoromethoxyl and trifluoroethoxyl.
[0031] The term "haloalkyl" as used herein refers to an alkyl group as defined herein that is substituted by 1 , 2, 3, 4, 5 or 6 halogen as defined herein. Examples of haloalkyl groups include, but are not limited to Ci-C6-haloalkyl groups, including trifluoromethyl and trifluoroethyl.
[0032] The term "hydroxyalkyl" as used herein refers to an alkyl group as defined herein that is substituted by 1, 2, 3, 4, 5 or 6 hydroxyl as defined herein. Examples of hydroxyalkyl groups include, but are not limited to CrC6-hydroxyalkyl groups, including hydroxymethyl and hydroxyethyl.
[0033] The term "alkylsulfonamide" as used herein refers to a group of the following formula: -NR'R"S(0)2-alkyl., wherein alkyl is as defined herein and R' and R" are as defined herein. A non limiting example of alkylsulfonamide group is methanesulfonyl.
[0034] The term "hydroxyl" as used herein refers to an alcohol group, -OH.
[0035] The term "alkoxyl" as used herein refers to an -O-alkyl group, wherein alkyl is as defined herein. Examples of alkoxyl groups include, but are not limited to Ci-C6-alkoxyl, including methoxy and ethoxy.
[0036] The term "oxo" refers to refers to a double-bonded oxygen (i.e., =0).
[0037] The term "a" as used herein means one or more.
[0038] As used herein, the terms "compound of this invention," "compounds of the present invention" and "compounds of Formula 1" includes compounds of Formula 1 and tautomers, resolved enantiomers, resolved diastereomers, racemic mixtures, solvates, metabolites, salts (including pharmaceutically acceptable salts) and pharmaceutically acceptable prodrugs thereof.
[0039] The compounds of Formula 1 include solvates, pharmaceutically acceptable prodrugs and salts (including pharmaceutically acceptable salts) of such compounds.
[0040] The phrase "pharmaceutically acceptable" indicates that the substance or composition is compatible chemically and/or toxicologically with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
[0041] A "solvate" refers to an association or complex of one or more solvent molecules and a compound of the invention. Examples of solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" can also be used to refer to a complex wherein the solvent molecule is water.
[0042] A "prodrug" is a compound that may be converted under physiological conditions or by solvolysis to the specified compound or to a salt of such compound. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three
or four) amino acid residues, is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of a compound of the present invention. The amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes phosphoserine, phosphothreonine, phosphotyrosine, 4- hydroxyproline, hydroxylysine, demosine, isodemosine, gamma-carboxyglutamate, hippuric acid, octahydroindole-2-carboxylic acid, statine, l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, penicillamine, ornithine, 3-methylhistidine, norvaline, beta-alanine, gamma-aminobutyric acid, cirtulline, homocysteine, homoserine, methyl-alanine, para-benzoylphenylalanine, phenylglycine, propargylglycine, sarcosine, methionine sulfone and tert-butylglycine.
[0043] Additional types of prodrugs are also encompassed. For instance, a free carboxyl group of a compound of Formula 1 can be derivatized as an amide or alkyl ester. As another example, compounds of this invention comprising free hydroxy groups may be derivatized as prodrugs by converting the hydroxy group into a group such as, but not limited to, a phosphate ester, hemisuccinate, dimethylaminoacetate, or phosphoryloxymethyloxycarbonyl group, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 115. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers, wherein the acyl group may be an alkyl ester optionally substituted with groups including, but not limited to, ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem., 1996, 39, 10. More specific examples include replacement of the hydrogen atom of the alcohol group with a group such as (Ci-C6)alkanoyloxymethyl, l-((Ci-C6)alkanoyloxy)ethyl, 1- methyl-l-((Ci-C6)alkanoyloxy)ethyl, (Ci-C6)alkoxycarbonyloxymethyl, N-(C C6)alkoxy- carbonylaminomethyl, succinoyl, (Ci-C6)alkanoyl, a-amino(Ci-C4)alkanoyl, arylacyl and a- aminoacyl, or α-aminoacyl-a-aminoacyl, where each a-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(0)(OH)2, -P(0)(0(Ci-C6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).
[0044] Free amines of compounds of Formula 1 can also be derivatized as amides, sulfonamides or phosphonamides. All of these moieties may incorporate groups including, but not limited to, ether, amine and carboxylic acid functionalities. For example, a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as R-carbonyl, RO- carbonyl, NRR'-carbonyl, wherein R and R' are each independently (Ci-Cio)alkyl, (C3- C7)cycloalkyl, or benzyl, or R-carbonyl is a natural a-aminoacyl or natural oc-aminoacyl-natural a- aminoacyl, -C(OH)C(0)OY wherein Y is H, (d-C6)alkyl or benzyl, -C(OY0)Y! wherein Y0 is (C
C4) alkyl and Yi is (Ci-C6)alkyl, carboxy(Ci-C6)alkyl, amino(Ci-C4)alkyl or mono-N- or di-N,N- (Ci-C6)alkylaminoalkyl, or -C(Y2)Y3 wherein Y2 is H or methyl and Y3 is mono-N- or di-N,N-(Cr C6)alkylamino, moφholino, piperidin-l-yl or pyrrolidin-l-yl.
[0045] For additional examples of prodrug derivatives, see, for example, a) Design of
Prodrugs, edited by H. Bundgaard, (Elsevier, 1985) and Methods in Enzymology, Vol. 42, p. 309- 396, edited by K. Widder, et al. (Academic Press, 1985); b) A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H. Bundgaard, Chapter 5 "Design and Application of Prodrugs," by H. Bundgaard p. 113-191 (1991); c) H. Bundgaard, Advanced Drug Delivery Reviews, 8:1-38 (1992); d) H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77:285 (1988); and e) N. Kakeya, et al., Chem. Pharm. Bull., 32:692 (1984), each of which is specifically incorporated herein by reference.
[0046] Alternatively or additionally, compound of the invention may possess a sufficiently acidic group, a sufficiently basic group, or both functional groups, and accordingly react with any of a number of inorganic or organic bases or acids to form a salt. Examples of salts include those salts prepared by reaction of the compounds of the present invention with a mineral or organic acid or an inorganic base, such salts including, but not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyn-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ-hydroxybutyrates, glycollates, tartrates, methanesulfonates, propanesulfonates, naphthalene- 1 -sulfonates, naphthalene-2-sulfonates, and mandelates. Since a single compound of the present invention may include more than one acidic or basic moiety, the compounds of the present invention may include mono, di or tri-salts in a single compound.
[0047] If the inventive compound is a base, the desired salt may be prepared by any suitable method available in the art, for example, by treatment of the free base with an acidic compound, for example, an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic
acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p- toluenesulfonic acid or ethanesulfonic acid, or the like.
[0048] If the inventive compound is an acid, the desired salt may be prepared by any suitable method, for example, by treatment of the free acid with an inorganic or organic base. Examples of suitable inorganic salts include those formed with alkali and alkaline earth metals, such as lithium, sodium, potassium, barium and calcium. Examples of suitable organic base salts include, for example, ammonium, dibenzylammonium, benzylammonium, 2- hydroxyethylammonium, bis(2-hydroxyethyl)ammonium, phenylethylbenzylamine, dibenzylethylenediamine, and the like salts. Other salts of acidic moieties may include, for example, those salts formed with procaine, quinine and N-methylglucosamine, plus salts formed with basic amino acids such as glycine, ornithine, histidine, phenylglycine, lysine and arginine.
[0049] In certain embodiments, the salt is a "pharmaceutically acceptable salt" which, unless otherwise indicated, includes salts that retain the biological effectiveness of the corresponding free acid or base of the specified compound and are not biologically or otherwise undesirable.
[0050] The compounds of Formula 1 also include other salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula 1 and/or for separating enantiomers of compounds of Formula 1.
[0051] The present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention, and their uses. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen,
2 3 1 1 13 carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as H, H, C, C, ,4C, 13N, 15N, 150, I70, , 80, 32P, 33P, 35S, , 8F, 36C1, 123I and 125I. Certain isotopically-labeled compounds of the present invention (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon- 14 (i.e., 14C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 15 O, 13 N, 11 C and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
[0052] The phrase "effective amount" means an amount of compound that, when administered to a mammal in need of such treatment, is sufficient to (i) treat or prevent a particular disease, condition, or disorder mediated by the activity of one or more AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) prevent or delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein. In the case of cancer, an effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. For cancer therapy, efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
[0053] The amount of a compound of Formula 1 that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
[0054] "Treating" is intended to mean at least the mitigation of a disease condition in a mammal, such as a human, that is affected, at least in part, by the activity of one or more AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases. The terms "treat" and "treatment" refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those found to be predisposed to having the disease condition but have not yet been diagnosed as having it; modulating and/or inhibiting the disease condition.
The terms "treating", "treat", or "treatment" embrace both preventative, i.e., prophylactic, and palliative treatment.
[0055] As used herein, the term "mammal" refers to a warm-blooded animal that has or is at risk of developing a disease described herein and includes, but is not limited to, guinea pigs, dogs, cats, rats, mice, hamsters, and primates, including humans.
[0056] It is to be understood that in instances where two or more radicals are used in succession to define a substituent attached to a structure, the first named radical is considered to be terminal and the last named radical is considered to be attached to the structure in question. Thus, for example, an arylalkyl radical is attached to the structure in question by the alkyl group.
AKT INHIBITORS
[0057] The inventive compounds of Formula 1 are useful for inhibiting AKT protein kinases. Such compounds have utility as therapeutic agents for diseases that can be treated by the inhibition of the AKT protein kinase signaling pathway and tyrosine and serine/threonine kinase receptor pathways.
[0058] The compounds of Formula 1 may also be useful as inhibitors of tyrosine kinases as well as serine and threonine kinases in addition to AKT.
[0059] In general, one aspect of the invention includes compounds of the Formula 1 :
(1)
and tautomers, resolved enantiomers, resolved diastereomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs thereof, wherein:
X is N or CR3;
Het is a roup selected from the grou consisting of:
(J)
is O, 1, 2, 3 or 4;
least one ofR 1', R2", and R 3J is a group of formula (L):
(L)
wherein:
n is 0, 1 or 2;
R' and R' are independently selected from the group consisting of H, Ci-C6-alkyl and
(C=0)-Ci-C6-alkyl;
R"' is H, OH or C,-C6-alkyl; and the rest of R1, R2, and R3 are independently selected from the group consisting of: H, cyano, Ci- C6-alkyl, aryl or heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, oxo, C]-C6-alkyl, Ci-C6-alkoxyl and
C i -C6-hydroxyalkyl;
R4 is selected from the group consisting of: H, halo, cyano, hydroxyl, oxo, Ci-C6-alkyl, Ci-C6- alkoxyl, COOH, C,-C6-alkylsulfonamide, NRaRb, NRa(C=0)Ra, (C=0)NRaRb, (C=0)NRa- CrC6-alkylenyl-NRaRb, NRa-C C6-alkylenyl-COOH, NRa-Ci-C6-alkylenyl-NReRb, aryl, heteroaryl or heterocycloalkyl, which aryl heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, oxo, hydroxyl, Ci-Ce-alkyl, C C6-alkoxyl, Ci-C6-haloalkyl, CrC6-haloalkoxyl, COOH, C C6- hydroxyalkyl and CrCe-alkylsulfonamide; and
Ra and Rb are independently selected from the group consisting of H, Ci-C6-alkyl, Ci-
C6-hydroxylalkyl or C3-C6-cycloalkyl.
[0060] In an embodiment, the compounds of formula 1 and tautomers, resolved enantiomers, resolved diastereomers and pharmaceutically acceptable salts thereof is provided.
[0061] In an embodiment, the compounds of formula 1, are those wherein R1 is a group of formula (L):
and the remaining substituents are as defined herein.
[0062] In another embodiment, the compounds of formula 1, are those wherein R' and R" are H, n is 0 and the remaining substituents are as defined herein.
[0063] In another embodiment, the compounds of formula 1, are those wherein R2 is a group of formula (L):
[0064] In another embodiment, the compounds of formula 1 are those wherein R4 is selected from the group consisting of: halo, , aryl, 5 or 6 membered heteroaryl or heterocycloalkyl, which, aryl, heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one halo and the remaining substituents are as defined herein.
[0065] In another embodiment, the compounds of formula 1 are those wherein R4 is aryl, 5 to 10 membered heterocycloalkyl containing one, two or three heteroatoms selected from the group consisting of N, S and O or 5 to 10 membered heteroaryl containing one, two or three heteroatoms selected from the group consisting of N, S and O, which aryl heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, oxo, hydroxyl, C C6-alkyl, C C6-alkoxyl, C C6-haloalkyl, Ci-C6-haloalkoxyl, COOH, C C6- hydroxyalkyl and C]-C6-alkylsulfonamide.
[0066] In another embodiment, the compounds of formula 1 are those wherein R4 is aryl, 5 to 6 membered heterocycloalkyl containing one, two or three heteroatoms selected from the group consisting of N, S and O or 5 to 6 membered heteroaryl containing one, two or three heteroatoms selected from the group consisting of N, S and O, which aryl heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, oxo, hydroxyl, C C6-alkyl, C C6-alkoxyl, CrC6-haloalkyl, CrC6-haloalkoxyl, COOH, C C6- hydroxyalkyl and CrCe-alkylsulfonamide.
[0067] In another embodiment, the compounds of formula 1 are those wherein X is N and the remaining substituents are as defined herein.
[0068] In another embodiment, the compounds of formula 1 are those wherein X is CR3 and the remaining substituents are as defined herein.
[0069] In another embodiment, the compounds of formula 1 are those wherein R3 is H or
Ci-C6-alkyl and the remaining substituents are as defined herein.
[0070] In another embodiment, the compounds of formula 1 are those wherein R1 is phenyl and the remaining substituents are as defined herein.
[0071] In another emboidment, R is selected from cyano, Ci-C6-alkyl, aryl or 5 or 6 membered heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, Ci-Ce-alkyl, Ci-C6-alkoxyl and C\- C6-hydroxyalkyl; or is a group of formula (L):
[0072] In another emboidment, R1 is selected from cyano, Ci-Ce-alkyl, aryl or 5 or 6 membered heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, Ci-C6-alkyl and Ci-C6-alkoxyl; or is a group of formula (L):
[0073] In another emboidment, R1 is selected from cyano, Ci-Ce-alkyl, aryl or 5 or 6 membered heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, Ci-C6-alkyl and Ci-C6-alkoxyl.
1 is selected from the group consisting of:
[0075] In a certain embodiment, R is selected from H, Ci-C6-alkyl and aryl; or or is a group of formula (L):
(L)
[0076] In a certain embodiment, R2 is selected from H, Ci-C6-alkyl and aryl.
[0077] In a certain embodiment, R2 is selected from the group consisting of:
[0078] In a certain embodiment, R3 is selected from H, C]-C6-alkyl and aryl; or or is a group of formula (L):
(L)
[0079] In a certain embodiment, R3 is selected from H, Ci-C6-alkyl and aryl.
[0080] In a certain embodiment, R3 is selected from the group consisting of:
and
[0082] In another embodiment, the compounds of formula 1 have the following formula 2
wherein Het, R 1 , R2 and R 3 are as defined herein.
(2-A)
wherein R1, R2, R3, R4 and m are as defined herein.
[0084] Non-limiting examples of compounds of formula 2-A are:
1 -(4-(2 -phenyl- 1 H-pyrrolo[2,3-b]pyridin- 1 -yl)phenyl)cyclobutanamine,
l-{4-[6-(2-Fluoro-phenyl)-2-phenyl-pyrrolo[2,3-b]pyridin-l-yl]-phenyl}-yclobutylamine, 1 -(4-(3 -phenyl- 1 H-pyrrolo[2,3-b]pyridin-2-yl)phenyl)cyclobutanamine,
1 -(4-(2 -phenyl- 1 H-pyrrolo[2,3-b]pyridin-3-yl)phenyl)cyclobutanamine, and
1 -(4-( 1 -methyl-2 -phenyl- 1 H-pyrrolo[2,3-b]pyridin-3- yl)phenyl)cyclobutanamine.
[0085] In an embodiment the compounds of formula 2 have the following formula 2-B:
(2-B) wherein R1, R2, R3, R4 and m are as defined herein.
[0086] Non-limiting examples of compounds of formula 2-B are:
1 -(4-(2 -phenyl- 1 H-pyrrolo[2,3-c]pyridin- 1 -yl)phenyl)cyclobutanamine, and
1 -(4-(2 -phenyl- 1 H-pyrrolo[2,3-c]pyridin- 1 -yl)phenyl)cyclobutanamine.
[0087] In an embodiment, the compounds of formula 2 have the following formula 2-C:
(2-C)
wherein R1, R2, R3, R4 and m are as defined herein.
[0088] Non-limiting examples of compounds of formula 2-C are:
1 -(4-( 1 -phenyl- lH-pyrrolo[3,2-c]pyridin-2-yl)phenyl)cyclobutanamine and
1 -(4-( 1 -aminocyclobutyl)phenyl)-2 -phenyl- 1 H-pyrrolo[3,2-c]pyridin-4-ol.
[0089] In an embodiment, the compounds of formula 2 have the following formula 2-D:
(2-D)
wherein R1, R2, R3, R4 and m are as defined herein.
[0090] A non limiting example of compound of formula 2-D is l-(4-(l- aminocyclobutyl)phenyl)-2-phenyl-lH-pyrrolo[3,2-b]pyridin-5-ol.
[0091] In an embodiment, the compounds of formula 2 have the following formula 2-E:
(2-E)
wherein R!, R2, R3, R4 and m are as defined herein.
[0092] Non-limiting examples of compounds of formula 2-E are:
l-(4-(6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl) cyclobutanamine,
1 -(4-( 1 -phenyl- 1 H-pyrrolo [3 ,2-c]pyridin-2-yl)phenyl)cyclobutanamine,
1 -(4-( 1 -aminocyclobutyl)phenyl)-2-phenyl- 1 H-pyrrolo[3 ,2-c]pyridin-4-ol,
l-(4-(6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
l-(4-(2-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
7-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- amine,
l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine, 1 -(7-(4-( 1 -aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)pyrrolidin-3-ol,
l-(4-(6-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
1- (4-(5-methyl-6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
(S)-l-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)pyrrolidin-3-ol,
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-2-carbonitrile, 7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-2- carboxamide,
(S)-l-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)pyrrolidine-2-carboxylic acid,
Nl-(7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)ethane- 1 ,2-diamine,
2- (7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- ylamino)acetic acid,
N-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)cyclopropanecarboxamide,
7-(4-(l-aminocyclobutyl)phenyl)-N,N-dimethyl-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin- 4-amine,
4-{7-[4-(l-aminocyclobutyl)phenyl]-6-(pyridin-2- yl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl}-
1λ6,4- thiomorpholine-l,l-dione,
l-(4-(4-methoxy-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine, l-(7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- yl)pyrrolidin-2-one,
l-[4-(5-Ethyl-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutylamine,
l-(4-(4-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
7-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- amine,
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-4- carboxamide,
7-(4-(l-aminocyclobu1yl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-4-carboxylic acid,
1 - { 4- [4-Methoxy-2-( 1 H-pyrazol-4-yl)-6-pyridin-2-yl-pyrrolo [2,3 -d]pyrimidin-7-yl] -phenyl } - cyclobutylamine,
7-(4-(l-aminocyclobutyl)phenyl)-N,N-diethyl-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- amine,
7-[4-(l-Amino-cyclobutyl)-phenyl]-6-pyridin-2-yl-7H-pyrrolo[2,3-d]pyrimidine-4-carboxylic acid (2-dimethylamino-ethyl)-amide,
1 -(7-(4-( 1 -aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- yl)pyrrolidin-3-ol,
l-{4-[5-Methyl-6-phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}- cyclobutylamine,
l-{4-[2-(2-Fluoro-phenyl)-5-methyl-6-phenyl-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}- cyclobutylamine,
7-(4-(l-aminocyclobutyl)phenyl)-N,N-diethyl-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- amine,
l-[4-(5-Methyl-2,6-diphenyl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutylamine, 1 -(4-(5-methyl-6-phenyl-2-(pyrrolidin- 1 -yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
1 -(4-(7-methyl-5-phenyl-2-( 1 H-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-6- yl)phenyl)cyclobutanamine,
l-[4-(7-Methyl-2,5-diphenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-phenyl]-cyclobutylamine, l-(4-(2-(2-fluorophenyl)-7-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-6- yl)phenyl)cyclobutanamine,
N-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)methanesulfonamide,
7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-ol, l-{4-[2-(2-Fluoro-phenyl)-7-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl]-phenyl}-
cyclobutylamine, and
l-[4-(7-Methyl-5-phenyl-2-pyrimidin-5-yl-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-phenyl]- cyclobutylamine.
[0093] In an embodiment, the compounds of formula 2 have the following formula 2-F:
[0094] Non-limiting examples of compounds of formula 2-F are:
1 - [4-(2-Chloro-6-phenyl-pyrrolo [3 ,2-d]pyrimidin-5 -y l)-phenyl] -cyclobutylamine,
{5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-dimethyl- amine,
l-(4-(6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
l-{4-[6-Phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[3,2-d]pyrimidin-5-yl]-phenyl}-cyclobutylamine, l-[4-(6-Phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutylamine,
l-[4-(5-Methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-phenyl]- cyclobutylamine, Nl-{5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-ethane- 1,2-diamine,
1- {5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-pyrrolidin-
3-ol,
5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxylic acid, tert-butyl l-(4-(2-(ethylamino)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-5- yl)phenyl)cyclobutylcarbamate,
2- {5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-ylamino}- ethanol,
l-(5-(4-(l-aminocyclobutyl)phenyl)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl)pyrrolidin-2- one,
l-[4-(2-Methoxy-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutylamine,
5-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-
carboxamide,
l-(4-(5-methyl-6-phenyl-2-(lH-pyrazol-4-yl)-5H-pyrrolo[3,2-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
l-(4-(5-methyl-2,6-diphenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine, l-(4-(2-chloro-5-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine, and
3-amino-l-methyl-3-(4-(5-methyl-6-phenyl-2-(lH-pyrazol-4-yl)-5H-pyrrolo[3,2-d]pyrimidin- 7-yl)phenyl)cyclobutanol .
[0095] In an embodiment, the compounds of formula 2 have the following formula 2-G:
(2-G)
wherein R1, R2, R3, R4 and m are as defined herein.
[0096] A non-limiting example of a compound of formula 2-G is l-[4-(6-Phenyl- pyrrolo[2,3-b]pyrazin-5-yl)-phenyl]-cyclobutylamine.
[0097] In an embodiment, the compounds of formula 2 have the following formula 2-H:
(2-H)
wherein R1, R2, R3, R4 and m are as defined herein.
[0098] embodiment, the compounds of formula 2 have the following formula 2-1:
(2-1)
wherein R1, R2, R3, R4 and m are as defined herein.
[0099] In an embodiment, the compounds of formula 2 have the following formula 2-J:
(2-J) wherein R , R , R , R and m are as defined herein.
[00100] In an embodiment, the compounds of formula 1 have the following formula 3:
(3)
wherein R 1 and R 2 are as defined herein.
[00101] In an embodiment, the compounds of formula 3 have the following formula 3-A:
(3-A)
wherein R1, R2, R4 and m are as defined herein.
[00102] Non-limiting examples of compounds of formula 3-A are:
l-(4-(5-methyl-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine,
l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine,
1 -(4-(6-(2-fluoropheny l)-2-pheny 1-3 H-imidazo [4, 5 -b]pyridin-3 -y l)phenyl)cyclobutanamine, l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine, and
l-(4-(5-phenyl-2-( yridin-2-yl)-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine.
[00103] In an embodiment, the compounds of formula 3 have the following formula 3-B:
(3-B)
wherein R1, R2, R4 and m are as defined herein.
[00104] Non-limiting examples of compounds of formula 3-B are:
1- (4-(3-phenyl-3H-imidazo[4,5-c]pyridin-2-yl)phenyl)cyclobutanamine and
2- (4-(l -aminocyclobutyl)phenyl)- 1 -phenyl- 1 H-imidazo[4,5-c]pyridin-6-ol.
[00105] In an embodiment the compounds of formula 3 have the following formula 3-C:
(3-C)
wherein R1, R2, R4 and m are as defined herein.
[00106] In an embodiment, the compounds of formula 3 have the following formula 3-D:
(3-D)
wherein R1, R2, R4 and m are as defined herein.
[00107] In an embodiment, the compounds of formula 3 have the following formula 3-E:
(3-E)
wherein R1, R2, R4 and m are as defined herein.
[00108] Non-limiting examples of compounds of formula 3-E are:
l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
7: 1 -(4-(2,8-diphenyl-9H-purin-9-yl)phenyI)cyclobutanamine;
10: N-(l-(4-(2,8-diphenyl-9H-purin-9-yl)phenyl)cyclobutyl)acetamide;
l-(4-(2-phenyl-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-(2-fluorophenyl)-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -[4-(8-Phenyl-2-piperazin- 1 -yl-purin-9-yl)-phenyl]-cyclobutylamine;
l-{4-[2-(2-Fluoro-phenyl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine;
{9-[4-(l-Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl}-methyl-amine;
{ 9-[4-( 1 -Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl } -dimethyl-amine;
28: 1 -(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
9-[4-(l -Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purine-2-carboxylic acid methylamide; l-{9-[4-(l-Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl}-pyrrolidin-2-one;
l-[4-(8-Phenyl-purin-9-yl)-phenyl]-cyclobutylamine;
l-[4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl]-cyclobutylamine;
9-(4-(l-aminocyclobutyl)phenyl)-8-phenyl-9H-purin-2-amine;
l-(4-(2-chloro-8-(thiazol-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-methoxy-8-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-{4-[2-(3 -Methyl-isoxazol-5 -yl)- 8-phenyl-purin-9-yl] -phenyl } -cyclobuty lamine ;
l-{4-[2-(5-Methyl-2H-pyrazol-3-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine;
l-{4-[2-(2-Methyl-3H-imidazol-4-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine; l-[4-(2-Phenyl-8-pyridin-3-yl-purin-9-yl)-phenyl]-cyclobutylamine;
l-(4-(2-phenyl-8-(pyridin-4-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-[4-(2-Ethoxy-8-phenyl-purin-9-yl)-phenyl]-cyclobutylamine;
9-(4-(l-aminocyclobutyl)phenyl)-N,N-dimethyl-8-phenyl-9H-purin-6-amine;
l-{4-[8-Phenyl-2-(lH-pyrazol-4-yl)-purin-9-yl]-phenyl}-cyclobutylamine; l-{4-[2-(l-Methyl-lH-pyrazol-4-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine; l-(4-(8-(4-methoxypyridin-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 H-imidazol-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-phenyl-8-(thiazol-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(9-phenyl-9H-purin-8-yl)phenyl)cyclobutanamine;
1 -(4-(8-phenyl-2-(pyrrolidin- 1 -yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-(3 -methoxypyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine ;
l-(4-(8-(5-methoxypyridin-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(6-( 1 -methyl- 1 H-pyrazol-4-yl)-8-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(2-phenyl-8-(pyrimidin-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-[4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutylamine;
l-(4-(8-tert-butyl-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-( 1 H-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine;
l-(4-(8-(4-methyl-lH-imidazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 H-imidazol-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(4-methylthiazol-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-(l -methyl- 1 H-benzo[d]imidazol-2-yl)-2-phenyl-9H-purin-9- yl)phenyl)cyclobutanamine ;
1 -(4-(8-( 1 -methyl- 1 H-imidazol-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(8-(5-methylisoxazol-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(oxazol-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-pyrazol-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-imidazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
N 1 -(9-(4-(l -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)ethane- 1 ,2-diamine;
N-(9-(4-(l-aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)methanesulfonamide; l-(4-(2-chloro-7-phenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-pyrazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-phenyl-8-( 1 H-pyrazol-4-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-phenyl-8-(thiazol-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(6-methoxypyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(7-phenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
1- (4-(2-phenyl-8-(lH-pyrazol-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)pyrrolidin-3-ol;
2- (4-(9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)- 1 H-pyrazol- 1 - yl)ethanol;
6-(9-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-9H-purin-8-yl)pyridin-2(lH)-one; l-(4-(2-methoxy-7-phenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-6-ol;
8- (4-( 1 -aminocyclobutyl)phenyl)-7-phenyl-7H-purin-2-ol ;
l-(4-(7-phenyl-2-(pyridin-3-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(7-phenyl-2-(pyrimidin-5-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
9- [4-(l-Amino-cyclobutyl)-phenyl]-2-phenyl-9H-purine-8-carbonitrile;
1 -(4-(8-(l -ethyl- lH-pyrazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(7-phenyl-2-( 1 H-pyrazol-4-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
1 -(4-(2-(2-fluorophenyl)-8-(l -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine;
l-(4-(2-phenyl-8-o-tolyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2,7-diphenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
1 -(4-(7-phenyl-2-( 1 H-pyrazol- 1 -yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(8-(3-bromopyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(8-(4-methylthiazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-(3-(trifluoromethoxy)phenyl)-9H-purin-9- yl)phenyl)cyclobutanamine;
l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-(3-(trifluoromethyl)phenyl)-9H-purin-9- yl)phenyl)cyclobutanamine;
l-(4-(7-phenyl-2-(thiazol-4-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(8-(3-methylpyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1- (4-(2-(4-methoxyphenyl)-8-(l -methyl- lH-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine;
2- (9-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-9H-purin-8-yl)benzonitrile;
1 -(4-(2-(2-methoxyphenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine;
4-(9-(4-( 1 -aminocyclobutyl)phenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-2-yl)-N- methylbenzenesulfonamide ;
1 -(4-(8-( 1 -methyl- 1 H-pyrazol-5-yl)-2-p-tolyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-(2-fluorophenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-pyrazol-5-yl)-2-m-tolyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-(4-fluorophenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine ;
l-(4-(8-(3-methoxypyridin-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; and l-(4-(8-(4-methyl-lH-imidazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine.
[00109] In an embodiment the compounds of formula 3 have the following formula 3-F:
(3-F)
wherein R1, R2, R4 and m are as defined herein.
[00110] In an embodiment, the compounds of formula 3 have the following formula 3-G:
(3-G)
wherein R1, R2, R4 and m are as defined herein.
[00111] Non-limiting examples of compounds of formula 3-G are:
l-[4-(2-Phenyl-imidazo[4,5-b]pyrazin-l-yl)-phenyl]-cyclobutylamine; and
1 -(4-(2-phenyl-5-(l H-pyrazol-4-yl)- 1 H-imidazo[4,5-b]pyrazin- 1 -yl)phenyl)cyclobutanamine.
[00112] In an embodiment, the com ounds of formula 3 have the following formula 3-H:
(3-H)
wherein R1, R2, R4 and m are as defined herein.
[00113] embodiment the compounds of formula 3 have the following formula 3-1:
(3-1)
wherein R1, R2, R4 and m are as defined herein.
[00114] In an embodiment, the compounds of formula 3 have the following formula 3-J:
(3-J)
wherein R1, R2, R4 and m are as defined herein.
[00115] The compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)-stereoisomers or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and diastereomers, and mixtures, racemic or otherwise, thereof. Accordingly, this invention also includes all such isomers, including diastereomeric mixtures, pure diastereomers and pure enantiomers of the compounds of this invention. The term "enantiomer" refers to two stereoisomers of a compound which are non- superimposable mirror images of one another. The term "diastereomer" refers to a pair of optical isomers which are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties, and reactivities.
[00116] The compounds of the present invention may also exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons.
[00117] In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed wedge representing a particular configuration, then that stereoisomer is so specified and defined.
METABOLITES OF COMPOUNDS OF FORMULA 1
[00118] Also falling within the scope of this invention are the in vivo metabolic products of compounds of Formula 1 described herein. A "metabolite" is a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof. Such products may result, for example, from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds of Formula 1, including compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
[00119] Metabolites are identified, for example, by preparing a radiolabelled (e.g., I4C or
H) isotope of a compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS, LC/MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art. The metabolites, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.
SYNTHESIS OF COMPOUNDS OF FORMULA 1
[00120] Compounds of this invention may be synthesized by synthetic routes that include processes analogous to those well known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources, such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, N.Y. (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements).
[00121] Compounds of Formula 1 may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds. Libraries of
compounds of Formula 1 may be prepared by a combinatorial 'split and mix' approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art. Thus according to a further aspect of the invention there is provided a compound library comprising at least 2 compounds of Formula 1, or salts thereof.
[00122] For illustrative purposes, Schemes 1-26 show a general method for preparing the compounds of the present invention, as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
[00123] Synthesis of substituted diaryl-imidazoles:
Scheme 1
[00124] The synthesis of certain compounds of the present invention is outlined in Scheme
1. These compounds can be synthesized from ortho-halo nitro-heteroarenes 1.1. Treatment of an optionally substituted nitro-heteroarene 1.1 (selected from Figure 1, where X is may be chloro, bromo, iodo, or trifluoromethanesulfonato) with a suitably protected aniline 1.2 (for a review of protecting groups see T.W. Greene, P.G.M. Wuts - Protective groups in Organic Synthesis, 3rd ed., John Wiley & Sons, 1999) in the presence of a palladium source, such as tris(dibenzylideneacetone) dipalladium, a ligand, such as 9,9-dimethyl-4,5- bis(diphenylphosphino)xanthene (Xantphos), a base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C produces nitro-anilines 1.3.
The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
Figure 1
[00125] Alternatively an optionally substituted nitro-heteroarene 1.1 (selected from Figure
1 , where X is may be fluoro, chloro, or methanesulfonyl) may be treated with a suitably protected aniline 1.2 (see T.W. Greene, supra) with or without a suitable base, such as diisopropylethylamine, with or without a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C to produce nitro-anilines 1.3. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00126] Reduction of the nitro group can be achieved by reacting nitro aniline 3 with hydrogen gas in the presence of a catalyst, such as Raney-nickel, in a suitable solvent, such as TFEF, at temperatures ranging from 25 °C to 200 °C, and at pressures ranging from 1 atm to 50 atm to produce amino-aniline 1.4. Depending on the choice of R1 and R2 it may be advantageous to use a chemoselective reduction method, such as sulfided platinum on carbon in a suitable solvent, such as ethanol, at temperatures ranging from 25 °C to 200 °C, and at pressures ranging from 1 atm to 50 atm to produce amino-aniline 1.4.
[00127] Cyclization to the fused imidazo-heteroarene 1.6 can be achieved by reacting amino-aniline 1.4 with an aldehyde 1.5a in the presence of an oxidant, such as copper(II)acetate, in a suitable solvent, such as acetic acid, at temperatures ranging from 25 °C to 200 °C. Alternatively imidazo-heteroarenes 1.6 may be synthesized by treating amino-anilines 1.4 with an orthoester 1.5b in the presence of an acid, such as 4-methylbenzenesulfonic acid, in a solvent, such as toluene, at temperatures ranging from 25 °C to 150 °C. The reduction of nitro anilines 1.3 can be performed in the presence of an orthoester 1.5b to affect the reduction and cyclization to imidazo-heteroarenes 1.6 in one step.
[00128] Someone skilled in the art will recognize that it may be advantageous to use building blocks 1.1 and 1.2 which will incorporate a functional group for further synthetic
transformations. For example R4 may be a halogen to yield compounds 1.6 which may be reacted further. It may be advantageous to mask such functional groups using a protecting group (for a review of protecting groups see T.W. Greene, P.G.M. Wuts - Protective groups in Organic Synthesis, 3rd ed., John Wiley & Sons, 1999).
Scheme 2
2.1 2.2 2.3 2.4
[00129] Selected examples from the invention described herein can be synthesized according to Scheme 2. 2-Substituted-4-chloro-5-nitro-pyrimidine 2.1 can be treated with aniline, such as 1.2, in a suitable solvent, such as THF. The resulting nitro aniline 2.2 can be reduced with hydrogen gas in the presence of a suitable catalyst, such as raney nickel, in a suitable solvent, such as THF. Dianiline 2.3 can be cyclized to purine 2.4 using aldehyde 1.5a in the presence of an oxidant, such as copper (II) acetate in acetic acid. It will appear to someone skilled in the art that R1, R2, or R4 may contain functional groups which have to be protected with a suitable protecting group. R!, R2, or R4 may also contain functional handles which allow further elaboration of 2.4.
Scheme 3
3.1 3.2 2.4
[00130] Specifically, selected examples from the invention described herein can be synthesized if R4 is chloride (
[00131] Scheme 3). Treatment of purine chloride 3.2 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols) can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases
(e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out in suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00132] In some cases it may be advantageous to achieve cross-couplings to carbon or non- carbon atoms by first converting the respective halogen derivative into the corresponding organometallic derivative (e.g., a boronic acid or ester, trifluoroborate salt, organomagnesium, organozinc or organotin compound). Such compounds are accessible by means of substituting the halide moiety with an appropriate metal or metalloid. Any functional groups present, may need to be protected by a suitable protecting group.
[00133] Introduction of such metals or metalloids can be achieved by generally well-known methods, such as metallation using metals or a metal-halogen exchange reaction. Useful metals for metallation include alkaline or alkaline earth metals or activated forms of such metals. Suitable reagents for use in metal-halogen exchange reactions include organolithium or organomagnesium compounds (e.g., N-butyllithium, tert-butyllithium or iso-propylmagnesium chloride or bromide). Subsequent transmetalation reactions of the organometallic intermediate may be performed as needed with a suitable soluble and reactive metal compound, such as magnesium chloride, magnesium bromide, tri-N-butyltin chloride, trimethyltin chloride, trimethyl borate, triethyl borate, tri-iso-propyl borate, zinc triflate or zinc chloride. Introduction of a boronic acid pinacol ester can be conveniently achieved by reacting the halogen derivative directly with bis(pinacolato)diboron in the presence of dichloro[l,l'- bis(diphenylphosphino)ferrocene]palladium(II) and suitable bases (e.g., potassium or sodium acetate) in solvents such as DMSO, DMF, DMA or N- methylpyrrolidone at temperatures ranging from 80-160 °C. Conventional heating or microwave irradiation may be employed to maintain the appropriate temperature (for literature precedent of similar transformations, see Ishiyama T. et al., J. Org. Chem., 60.750S (2003)). Methods for conversion of the boronic acid pinacol ester obtained by this method into other boronic acid derivatives such as boronic acids, boronates, or trifluoroborate salts are generally well known. As will be apparent to the skilled artisan, such organometallic derivatives may be utilized in cross- coupling reactions similar to those described above. Such couplings can be effected utilizing suitable coupling partners, such as aromatic, heteroaromatic halides or olefinic reagents under conditions identical or evidently similar and/or related to the methods described above.
[00134] Other methods may utilize the reactivity of organometallic derivatives generated from 3.2 by any of the methods described above. For example, derivatives containing alkaline or alkaline earth metals (e.g., organolithium, organomagnesium or organozinc compounds) may be employed in direct couplings to a range of other electrophilic coupling partners such as, for example, activated olefins (MICHAEL-acceptors), aldehydes, nitriles, aromatic nitro compounds, carboxylic acid derivatives, oxiranes, aziridines, organic disulfides or organic halides. Such transformations are generally well known in the art (for reactions with aromatic nitro compounds, see, e.g., Sapountzis L, et al., J. Am. Chem. Soc, J 24:9390 (2002)).
[00135] This methodology may be extended to the incorporation of non-carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) that may optionally contain suitable protecting groups of alcohols, thiols or amines. Examples of such groups can be found in Greene T., supra. Exemplary methods of such utilization of non-carbon nucleophiles in related cross-coupling reactions may be found in Ley S. et al., Angew. Chem., /75:5558 (2003); Wolfe J. et al., Ace. Chem. Res., 31:805 (1998); Hartwig, Ace. Chem. Res., 31 :852 (1998); Navarro O., et al., J. Org. Chem., 69:3173 (2004), Ji J. et al., Org. Lett, 5:461 1 (2003). The skilled artisan will recognize that the compounds obtained by such methods can be further elaborated by generally well known methods to obtain other compounds of the present invention.
[00136] Some of the compounds in the present invention may be synthesized by treatment of chloro purine 3.2 with a nucleophile, such as a primary or secondary amine, or an alcohol, in the presence of carbon monoxide, a suitable catalyst, such as dichloro[l,l'- bis(diphenylphosphino)ferrocene]palladium(II), a base, such as triethylamine, and a solvent, such as dioxane, at temperatures ranging from 80-160 °C. Conventional heating or microwave irradiation may be employed to maintain the appropriate temperature (exemplary methods of carbonylation may be found in Brennfuhrer, A. et al., Angew. Chem., 48(23):4114 (2009)).
[00137] Alternatively, compounds described in the invention may be synthesized by treating chloro-purine 3.2 with a suitably protected carbon-based nucleophiles (e.g., malonates) and non- carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) via nucleophilic aromatic substitution. The reactions may be accelerated using an appropriate acid, such as toluenesulfonic acid, or base, such as triethylamine, diisopropylethylamine, sodium hydride, or an alkaline carbonate base, in the presence or absence of a suitable solvent, such as, but not limited to, THF, dioxane, DMF, DMA, DMSO, or acetonitrile.
Scheme 4
4.2 4.3 4.4
[00138] Selected examples from the invention described herein can be synthesized according to Scheme 4. 6-Substituted-4-chloro-5-nitro-pyrimidine 4.1 can be treated with an aniline, such as 1.2, in a suitable solvent, such as THF. The resulting nitro aniline 4.2 can be reduced with hydrogen gas in the presence of a suitable catalyst, such as raney nickel, in a suitable solvent, such as THF. Dianiline 4.3 can be cyclized to purine 4.4 using aldehyde 1.5a in the presence of an oxidant, such as copper(II) acetate in acetic acid. It will be evident to someone skilled in the art that R1, R2, or R4 may contain functional groups which have to be protected with a suitable protecting group. R1, R2, or R4 may also be functional handles which allow further elaboration of 4.4.
Scheme 5
[00139] Specifically, selected examples from the invention described herein can be synthesized if R4 is chloride (Scheme 5). Purine 5.2 can by synthesized from 4,6-dichloro-5-nitro- pyrimidine 5.1 according to Scheme 4. Treatment of purine chloride 5.2 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling
reactions may be carried out in suitable solvents such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00140] The Pd catalyzed coupling or SnAr reaction from 5.2 to 4.4 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 5 as well.
Scheme 6
6.1 6.2 6.3 6.4
[00141] Tetrasubstituted purines from the invention described herein can be synthesized from dichloropurine 6.2 (Scheme 6) which may be synthesized from 2,4,6-trichloro-5- nitropyrimidine according to Scheme 4. Functionalization of 6.2 according to the conditions described above followed by functionalization of the resulting chloropurine 6.3 according to the conditions described above generates tetrasubstituted purines 6.4.
7.1 7.2 7.3 7.4
[00142] Selected examples from the invention described herein can be synthesized according to Scheme 7. Optionally substituted 2-chloro-3-nitro-pyridine 7.1 may be treated with an aniline 1.2 in the presence of base, such as diisopropylethylamine, and a suitable solvent, such as THF, to yield nitropyridines 7.2. Reduction of nitroaniline 7.2 using hydrogen and a suitable catalyst, such as Raney-nickel, in a solvent, such as TFiF, generates dianiline 7.3. Dianiline 7.3 can be cyclized to imidazo[4,5-b]pyridine 7.4 using aldehyde 1.5a in the presence of an oxidant, such as copper(II) acetate in acetic acid. It will be evident to someone skilled in the art that R1, R2, or R4
may contain functional groups which have to be protected with a suitable protecting group. R1, R2, or R4 may also be functional handles which allow further elaboration of 7.4.
8.1 8.2 8.3
[00143] Selected examples from the invention described herein can be synthesized starting from 5-bromo-2-chloro-3-nitropyridine 8.1. 6-Bromo-imidazo[4,5-b]pyridine 8.2 can be synthesized according to Scheme 7. Treatment of 6-bromo-imidazo[4,5-b]pyridine 8.2 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00144] The Pd catalyzed coupling or SnAr reaction from 8.2 to 8.3 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 8 as well.
9.1 9.2 9.3
[00145] Other selected examples from the invention described herein can be synthesized from 2,6-dichloro-3-nitro-pyridine 9.1 (Scheme 9). 5-Chloro-imidazo[4,5-b]pyridine 9.2 can be synthesized according to Scheme 7. Treatment of 5-chloro-imidazo[4,5-b]pyridine 9.2 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00146] The Pd catalyzed coupling or SnAr reaction from 9.2 to 9.3 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 9 as well.
Scheme 10
10.1 10.2 10.3 10.4
[00147] Selected examples from the invention described herein can be synthesized according to Scheme 10. Optionally substituted 2-chloro-3-nitro-pyrazine 10.1 (Hartman, G.D., et al., J. Het. Chem. 20, 947-950 (1983)) may be treated with an aniline 1.2 in the presence of base, such as triethylamine, and a suitable solvent, such as dioxane, to yield nitropyrazine 10.2. Reduction of nitroaniline 10.2 using hydrogen and a suitable catalyst, such as Raney-nickel, in a solvent, such as THF, generates dianiline 10.3. Dianiline 10.3 can be cyclized to imidazo[4,5- b]pyrazine 10.4 using aldehyde 1.5a in the presence of an oxidant, such as copper(II) acetate in acetic acid. It will be evident to someone skilled in the art that R1, R2, or R4 may contain functional
groups which have to be protected with a suitable protecting group. R1, R2, or R4 may also be functional handles which allow further elaboration of 10.4.
Synthesis of substituted aryl-pyrroles:
Scheme 11
11.6 11.7
[00148] The synthesis of certain compounds of the present invention is outlined in Scheme
11. Many of these compounds may be synthesized using a suitably protected dihalide 11.1 (X1 = bromo, iodo) and X being chloro or bromo. Treatment of a suitably protected dihalide, such as compounds shown in Figure 2 (with X1 being a bromide, iodide, or trifluoromethanesulfonato, and X being a chloride or bromide) with a terminal alkyne 11.2, in the presence of a palladium catalyst, such as dichloro(bis-triphenylphosphine)palladium, an additive, such as copper iodide, and a base, such as triethylamine, yields alkynes 11.3 (For a review on alkynylations, see Negishi et al., Chem. Rev. 103(5), 1979-2013 (2003)). Pg is an amine protecting group, e.g., acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc).
[00149] Treatment of alkynes 11.3 with a suitably protected aniline 1.2 (for a review of protecting groups see T.W. Greene, supra) in the presence of a palladium source, such as tris(dibenzylideneacetone) dipalladium, a ligand, such as 9,9-dimethyl-4,5- bis(diphenylphosphino)xanthene (Xantphos), a base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C produces anilino-alkynes 1 1.4.
[00150] Treatment of anilino-alkynes 11.4 with a suitable catalyst, such as bis- tributylphosphine palladium, a suitable additive, such as potassium fluoride, a base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C produces aryl-pyrroles 11.5.
[00151] Alternatively, the reaction to form aryl-pyrroles 11.5 from alkynes 11.3 may be performed as a one pot reaction.
Figure 2
[00152] Alternatively an optionally substituted amino-heteroarene (selected from Figure 2, with X 1 being a bromide, iodide, or trifluoromethanesulfonato, and X 2 being a fluoride or chloride) can be used as compound 11.1 and may be treated with a suitably protected aniline 1.2 (for a review of protecting groups see T.W. Greene, P.G.M. Wuts - Protective groups in Organic Synthesis, 3rd ed., John Wiley & Sons, 1999) in the presence of a base, such as diisopropylethylamine, with or without a suitable solvent, such as dioxane at temperatures ranging from 25 °C to 200 °C produces anilino-alkynes.
Scheme 12
[00153] Specifically, selected examples from the invention described herein can be synthesized from 4-chloro-3-iodopyridine 12.1. Treatment of optionally substituted 4-chloro-3-
iodopyridine 12.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 12.2. Treatment of 12.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2-dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields pyrrolo[3,2- cjpyridine 12.3.
Scheme 13
13.2 13.3 13.4
^IH2
13.5 13.6
[00154] Specifically, selected examples from the invention described herein can be synthesized starting from 2-chloro-3-iodo-4-amino-pyridines (Scheme 13). Treatment of an optionally protected 2-chloro-3-iodo-4-amino-pyridine 13.1 (such as tert-butyl 2-chloro-3- iodopyridin-4-ylcarbamate, R=Boc) can be treated with nucleophiles such as, but not limited to, amines, alkoxides, or thioalkoxides, in the presence of a suitable solvent, such as dioxane, to yield iodopyridines 13.2. Treatment of iodide 13.2 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro-bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 13.3. Treatment of amino alkyne 13.3 (R=H, following a suitable deprotection) under diazotization conditions, such as sodium nitrate and toluenesulfonic acid, in a suitable solvent, such as acetonitrile, followed by sodium iodide, yields iodide 13.4. Treatment of iodide 13.4 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2-dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable
solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 13.5. Cyclization of amino alkyne 13.5 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert- butylphosphine)palladium(O), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[3,2-c]pyridine 13.6.
Scheme 14
[00155] Specifically, selected examples from the invention described herein can be synthesized from optionally substituted 2-bromo-3-chloropyridines 14.1 (Scheme 14). Treatment of bromide 14.1 with an alkyne 1 1.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 14.2. Treatment of chloride 14.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 14.3. Cyclization of amino alkyne 14.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[3,2-b]pyridines 14.4.
Scheme 15
YH2
[00156] Specifically, selected examples from the invention described herein can be synthesized from optionally substituted 2-chloro-3-iodo-pyridines 15.1 (Scheme 15). Treatment of
iodide 15.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 15.2. Treatment of chloride 15.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 15.3. Cyclization of amino alkyne 15.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[2,3-b]pyridines 15.4.
Scheme 16
16.5 16.6
[00157] Specifically, selected examples from the invention described herein can be synthesized starting from 6-chloro-3-iodo-2 -amino-pyridines (Scheme 16). Treatment of an optionally protected 2-amino-6-chloro-3-iodo-pyridine 16.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro-bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 16.2. Someone skilled in the art will recognize that chloro alkyne 16.2 can be functionalized in a variety of ways. It may be advantageous to protect reactive functional groups, such as the amino group. Treatment of chloro-pyridine 16.2 with suitable reagents such, as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a
suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00158] This methodology may be extended to the incorporation of non-carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) that may optionally contain suitable protecting groups of alcohols, thiols or amines. Examples of such groups can be found in Greene T. et al., supra. Exemplary methods of such utilization of non-carbon nucleophiles in related cross-coupling reactions may be found in Ley S. et al., Angew. Chem., /75:5558 (2003); Wolfe J. et al., Ace. Chem. Res., 31 :805 (1998); Hartwig, Ace. Chem. Res., 31 :852 (1998); Navarro O., et al., J. Org. Chem., 69:3173 (2004), Ji J. et al., Org. Lett, 5:4611 (2003). The skilled artisan will recognize that the compounds obtained by such methods can be further elaborated by generally well known methods to obtain other compounds of the present invention.
[00159] Some of the compounds in the present invention may be synthesized by treatment of
5-chloro-imidazo[4,5-b]pyridine 16.2 with a nucleophile, such as a primary or secondary amine, or an alcohol, in the presence of carbon monoxide, a suitable catalyst, such as dichloro[l,l'- bis(diphenylphosphino)ferrocene]palladium(II), a base, such as triethylamine, and a solvent, such as dioxane, at temperatures ranging from 80-160 °C. Conventional heating or microwave irradiation may be employed to maintain the appropriate temperature (Exemplary methods of carbonylation may be found in Brennfuhrer, A. et al., Angew. Chem., 48(23):4114 (2009)).
[00160] Alternatively, compounds described in the invention may be synthesized by treating chloro-imidazo[4,5-b]pyridine 16.2 with a suitably protected carbon-based nucleophiles (e.g., malonates) and non-carbon based nucleophiles (e.g., alcohols, thiols, primary or secondary amines) via nucleophilic aromatic substitution. The reactions may be accelerated using an appropriate acid, such as toluenesulfonic acid, or base, such as triethylamine, diisopropylethylamine, sodium hydride, or an alkaline carbonate base, in the presence or absence of a suitable solvent, such as, but not limited to, THF, dioxane, DMF, DMA, DMSO, or acetonitrile.
[00161] Amino-alkyne 16.3 can be converted to bromo-alkyne 16.4 under diazotization conditions, such as sodium nitrate and toluenesulfonic acid, in a suitable solvent, such as acetonitrile, followed by sodium iodide. Treatment of bromide 16.4 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 16.5. Cyclization of amino alkyne 16.5 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[2,3-b]pyridines 16.6.
Scheme 17
[00162] An alternative method to synthesize selected examples from the invention described herein is shown in Scheme 17. Treatment of amino alkyne 16.2 with a suitable halide (such as bromide 11.8a) in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 9,9-dimethyl-4,5-bis(diphenylphosphino)xanthene (Xantphos), a base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 25 °C to 200 °C produces 6-chloro pyrrolo[2,3-b]pyridines 17.1. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00163] Treatment of 6-chloro pyrrolo[2,3-b]pyridines 17.1 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling
reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00164] The Pd catalyzed coupling or SnAr reaction from 17.1 to 16.6 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 17 as well.
Scheme 18
18.1 18.2 1 B J
[00165] Selected examples from the invention described herein can be synthesized from optionally substituted 4-chloro-5-iodo-pyrimidine 18.1 (Scheme 18). Treatment of iodide 18.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 18.2. Treatment of chloride 18.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields pyrrolo[2,3-d]pyrimidines 18.3.
Scheme 19
^IH2
19.5
[00166] Selected examples from the invention described herein can be synthesized from optionally substituted 2,4-dichloro-5-iodo-pyrimidine 19.1 (Scheme 19). Treatment of dichloride
19.1 with an aniline, such as 1.2, in the presence of a base, such as triethylamine, in a suitable solvent, such as THF results in the formation of iodoaniline 19.2. Treatment of 19.2 with an alkyne
11.2 in the presence of a suitable catalyst, such as dichloro-bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 19.3. Cyclization of amino alkyne 19.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield 2-chloro pyrrolo[2,3-d]pyrimidines 19.4. Treatment of 2-chloro pyrrolo[2,3-d]pyrimidines 19.4 with suitable reagents such, as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N- methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00167] The Pd catalyzed coupling or SnAr reaction from 19.4 to 19.5 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 19 as well.
Scheme 20
^H2
20.5
[00168] Selected examples from the invention described herein can be synthesized from optionally substituted 4,6-dichloro-5-iodo-pyrimidine 20.1 (Org. Lett.; 11(8), 2009; 1837 - 1840) as seen in Scheme 20. Treatment of dichloride 20.1 with an aniline, such as 1.2, in the presence of a base, such as triethylamine, in a suitable solvent, such as THF results in the formation of iodoaniline 20.2. Treatment of 20.2 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro-bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 20.3. Cyclization of amino alkyne 20.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield 4-chloro pyrrolo[2,3-d]pyrimidines 20.4. Treatment of 4- chloro pyrrolo[2,3-d]pyrimidines 20.4 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out
in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00169] The Pd catalyzed coupling or SnAr reaction from 20.4 to 20.5 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 20 as well.
Scheme 21
}JH2
21.5 21.6
[00170] Selected examples from the invention described herein can be synthesized from
2,4.6-trichloro-5-iodo-pyrimidine 21.1 (Scheme 21). Treatment of trichloride 21.1 with an aniline, such as 1.2, in the presence of a base, such as triethylamine, in a suitable solvent, such as THF, results in the formation of iodoaniline 21.2. Treatment of 21.2 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro-bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 21.3. Cyclization of amino alkyne 21.3 can be achieved by treatment with a base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield 2,4-dichloro pyrrolo[2,3- djpyrimidines 21.4. In scheme 21, the two R4 groups can be the same of can be different.
[00171] Someone skilled in the art will recognize that further functionalization according to
Scheme 20 can lead to 2-chloro-4-substituted-pyrrolo[2,3-d]pyrimidines 21.5, which can be functionalized according to Scheme 19 to lead to tetrasubstituted pyrrolo[2,3-d]pyrimidines 21.6.
Scheme 22
^H2
22.1 22.2
[00172] Selected examples from the invention described herein can be synthesized from optionally substituted 3-chloro-4-iodo-pyridine 22.1 (Scheme 22). Treatment of iodide 22.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 22.2. Treatment of chloride 22.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields pyrrolo[2,3-d]pyrimidines 22.3.
Scheme 23
23.1 23.2 23 3 23.4
[00173] Selected examples from the invention described herein can be synthesized from optionally substituted 2,3-dichloro-pyrazines 23.1 (Scheme 23). Treatment of dichloride 23.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 23.2. Treatment of chloride 23.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields amino alkyne 23.3. Cyclization of amino alkyne 23.3 can be achieved by treatment with a
base, such as cesium carbonate and potassium fluoride, with or without the use of a catalyst, such as di(tri-tert-butylphosphine)palladium(0), in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C to yield pyrrolo[3,2-b]pyrazines 23.4.
Scheme 24
[00174] Selected examples from the invention described herein can be synthesized from optionally substituted 2,4-dichloro-5-bromo-pyrimidine 24.1 (Scheme 24). Treatment of dichloride 24.1 with an alkyne 11.2 in the presence of a suitable catalyst, such as dichloro- bis(triphenylphosphine)palladium (II), a suitable additive, such as copper(I)iodide, a base, such as triethylamine, with or without a suitable solvent, at temperatures ranging from 20 °C to 150 °C yields alkyne 24.2. Treatment of 24.2 with a suitable aniline 1.2 in the presence of a catalyst, such as tris(dibenzylideneacetone)dipalladium(0), a suitable ligand, such as 2-dicyclohexylphosphino- 2',4',6'-triisopropylbiphenyl (X-phos), a suitable base, such as cesium carbonate, in a suitable solvent, such as dioxane, at temperatures ranging from 50 °C to 200 °C yields 2-chloro pyrrolo[3,2- djpyrimidines 24.3.
[00175] Treatment of 2-chloro pyrrolo[3,2-d]pyrimidines 24.3 with suitable reagents, such as boronic acids and boronates, organoboranes, organostannanes, organozinc compounds, organomagnesium compounds, olefins or terminal alkynes (either purchased or obtained via generally well known protocols), can be carried out in the presence of a suitable transition metal catalyst (e.g., palladium compounds). The coupling may optionally be performed in the presence of ligands such as, but not limited to, phosphines, diphosphines, heterocyclic carbenes, or arsines. Organic or inorganic bases (e.g., tertiary or secondary amines, alkaline carbonates, bicarbonates, fluorides or phosphates) and/or other well known additives (e.g., lithium chloride, copper halides or silver salts) maybe utilized to effect, assist or accelerate such transformations. These cross coupling reactions may be carried out in suitable solvents, such as THF, dioxane, dimethoxyethane, diglyme, dichloromethane, dichloroethane, acetonitrile, DMF, N-methylpyrrolidone, water, or mixtures of thereof, at temperatures ranging from 25 °C to 200 °C. The temperature may optionally be maintained with heating, conventional heating or microwave irradiation.
[00176] The Pd catalyzed coupling or SnAr reaction from 24.3 to 24.4 may be performed similarly as in Scheme 3 (compound 3.2 to compound 2.4), and as such, the description of those potential reactions (see above) apply to Scheme 24 as well.
Scheme 25
25.1 25.3
[00177] Selected examples from the invention described herein can be synthesized from optionally substituted 4-chloro-5-iodo-pyrimidine 18.1 (Scheme 25). Treatment of chloride 18.1 with an aniline, such as 1.2, in the presence of a base, such as triethylamine, in a suitable solvent, such as THF results in the formation of iodoaniline 25.1. Treatment of 25.1 with a disubstituted alkyne 25.2 in the presence of a suitable catalyst, such as palladium(II) acetate, a suitable additive, such as lithium chloride, a base, such as potassium acetate, in a suitable solvent, such as DMF, at temperatures ranging from 50 °C to 200 °C yields 5,6-disubstituted pyrrolo[2,3-d]pyrimidines 20.4.
Scheme 26
[00178] Selected examples from the invention described herein can be synthesized from optionally substituted iodoaniline 19.2 (Scheme 26). Treatment of 19.2 with a disubstituted alkyne 25.2 in the presence of a suitable catalyst, such as palladium(II) acetate, a suitable additive, such as lithium chloride, a base, such as potassium acetate, in a suitable solvent, such as DMF, at temperatures ranging from 50 °C to 200 °C yields 5,6-disubstituted 2-chloro-pyrrolo[2,3- d]pyrimidines 26.1. Functionalization of 2-chloro-pyrrolo[2,3-d]pyrimidines 26.1 can be achieved under conditions similar to those outlined for Scheme 19.
[00179] Another aspect of this invention provides a method of preparing a compound of Formula 1, comprising the steps of reducing a compound having the formula:
and reacting it with a compound of formula:
R1 O
wherein Het, R1 and R2 are as defined herein.
[00180] In an embodiment of the method of preparing a compound of formula 1 above, the reaction between the two compounds is an oxidative cyclization.
[00181] Another aspect of this invention provides a method of preparing a compound of Formula 1, comprising the steps of reacting a compound having the formula:
f}JH2
R2
wherein Het, R1 and R2 are as defined herein and X2 is halo, e.g., chloride or bromide.
[00182] Another aspect of this invention provides a method of preparing a compound of
Formula 1 , comprising the steps of having the formula:
R2
wherein Het, R1 and R2 are as defined herein and X3 is a halogen atom, Pg is an amine protecting group, e.g., acetyl, trifluoroacetyl, BOC, CBz and Fmoc.
[00183] In preparing compounds of Formula 1, protection of remote functionalities (e.g., primary or secondary amines, etc.) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, BOC, CBz and Fmoc. The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, supra.
METHODS OF SEPARATION
[00184] In any of the synthetic methods for preparing compounds of Formula 1, it may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
[00185] Another class of separation methods involves treatment of a reaction mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like. Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like. Alternatively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.
[00186] Selection of appropriate methods of separation depends on the nature of the materials involved. For example, boiling point and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply techniques most likely to achieve the desired separation.
[00187] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by
converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC column.
[00188] A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. "Stereochemistry of Organic Compounds," John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., (1975) 113(3):283-302). Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: "Drug Stereochemistry, Analytical Methods and Pharmacology," Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993).
[00189] Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl-β- phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.
[00190] Alternatively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (E. and Wilen, S., supra at p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-)menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob III. J. Org. Chem., (1982) 47:4165), of the racemic mixture, and analyzing the lH
NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse- phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase ("Chiral Liquid Chromatography" (1989) W. J. Lough, Ed., Chapman and Hall, New York; Okamoto, J. of Chromatogr., (1990) 513:375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
METHODS OF TREATMENT WITH COMPOUNDS OF FORMULA 1
[00191] The compounds of the present invention can be used as prophylactics or therapeutic agents for treating diseases or disorders mediated by modulation or regulation of AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases. AKT protein kinase mediated conditions that can be treated according to the methods of this invention include, but are not limited to, inflammatory, hyperproliferative cardiovascular, neurodegenerative, gynecological, and dermatological diseases and disorders.
[00192] In one embodiment, said pharmaceutical composition is for the treatment of hyperproliferative disorders, including cancers of the following categories: (1) Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; (2) Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma, non-small cell lung, small cell lung; (3) Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); (4) Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); (5) Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; (6) Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant
lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; (7) Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform. oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); (8) Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma); (9) Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma]; (10) Skin: advanced melanoma, malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; (11) Adrenal glands: neuroblastoma; (12) Breast: metastatic breast; breast adenocarcinoma; (13) Colon; (14) Oral cavity; (15) Hairy cell leukemia; (16) Head and neck; (17) and others including refractory metastatic disease; Kaposi's sarcoma; Bannayan-Zonana syndrome; and Cowden disease or Lhermitte-Duclos disease, among other kinds of hyperproliferative disorders.
[00193] In certain embodiments, the AKT protein kinase-mediated condition is a hyperproliferative disease.
[00194] In certain embodiments, the hyperproliferative disease is cancer.
[00195] In certain embodiments, the cancer is selected from colon, ovarian, brain, lung, pancreatic, prostate, and gastric carcinomas.
[00196] Compounds and methods of this invention can be also used to treat diseases and conditions such as rheumatoid arthritis, osteoarthritis, Chron's disease, angiofibroma, ocular diseases (e.g., retinal vascularisation, diabetic retinopathy, age-related macular degeneration, macular degeneration, etc.), multiple sclerosis, obesity, Alzheimer's disease, restenosis, autoimmune diseases, allergy, asthma, endometriosis, atherosclerosis, vein graft stenosis, peri- anastomatic prothetic graft stenosis, prostate hyperplasia, chronic obstructive pulmonary disease, psoriasis, inhibition of neurological damage due to tissue repair, scar tissue formation (and can aid
in wound healing), multiple sclerosis, inflammatory bowel disease, infections, particularly bacterial, viral, retroviral or parasitic infections (by increasing apoptosis), pulmonary disease, neoplasm, Parkinson's disease, transplant rejection (as an immunosupressant), septic shock, etc.
[00197] Accordingly, another aspect of this invention provides a method of treating diseases or medical conditions in a mammal mediated by AKT protein kinases, comprising administering to said mammal one or more compounds of Formula 1 or a pharmaceutically acceptable salt or prodrug thereof in an amount effective to treat or prevent said disorder.
[00198] This invention also provides compounds of Formula 1 for use in the treatment of
AKT protein kinase-mediated conditions.
[00199] An additional aspect of the invention is the use of a compound of Formula 1 in the preparation of a medicament for therapy, such as for the treatment or prevention of AKT protein kinase-mediated conditions.
COMBINATION THERAPY
[00200] The compounds of the present invention can be used in combination with one or more additional drugs such as described below. The dose of the second drug can be appropriately selected based on a clinically employed dose. The proportion of the compound of the present invention and the second drug can be appropriately determined according to the administration subject, the administration route, the target disease, the clinical condition, the combination, and other factors. In cases where the administration subject is a human, for instance, the second drug may be used in an amount of 0.01 to 100 parts by weight per part by weight of the compound of the present invention.
[00201] The second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of this invention such that they do not adversely affect each other. Such drugs are suitably present in combination in amounts that are effective for the purpose intended. Accordingly, another aspect of the present invention provides a composition comprising a compound of this invention in combination with a second drug, such as described herein.
[00202] A compound of this invention and the additional pharmaceutically active drug(s) may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. Such sequential administration may be close in time or remote in time. The amounts of the compound of this invention and the second drug(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
[00203] The combination therapy may provide "synergy" and prove "synergistic", i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
[00204] A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer, regardless of mechanism of action. Chemotherapeutic agents include compounds used in "targeted therapy" and conventional chemotherapy.
[00205] Examples of chemotherapeutic agents include PLX4032 (Roche/Plexxikon),
Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sutent (SU 11248, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), Lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline), Lonafarnib (SCH 66336), Sorafenib (BAY43-9006, Bayer Labs), Irinotecan (CAMPTOSAR®, Pfizer) and Gefitinib (IRESSA®, AstraZeneca), AG1478, AG1571 (SU 5271; Sugen), alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin
omegall (Angew Chem. Intl. Ed. Engl. (1994) 33:183-186); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), mo holino-doxorubicin, cyanomo holino-doxorubicin, 2- pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara- C"); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL® (paclitaxel; Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANE™ (Cremophor-free), albumin-engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, Illinois), and TAXOTERE® (doxetaxel; Rhone-Poulenc Rorer, Antony, France); chloranmbucil; GEMZAR® (gemcitabine); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® (vinorelbine); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA®); ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.
[00206] Also included in the definition of "chemotherapeutic agent" are: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LYl 17018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca); (iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3- dioxolane nucleoside cytosine analog); (iv) protein kinase inhibitors; (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Ralf and H- Ras; (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®; PROLEUKTN® rIL-2; a topoisomerase 1 inhibitor such as LURTOTECAN®; ABARELIX® rmRH; (ix) anti-angiogenic agents such as bevacizumab (AVASTIN®, Genentech); and (x) pharmaceutically acceptable salts, acids and derivatives of any of the above.
[00207] Also included in the definition of "chemotherapeutic agent" are therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idee), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
[00208] Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the PI3K inhibitors of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, pascolizumab, pecfusituzumab, pectuzumab, pertuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab,
resyvizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab, tacatuzumab tetraxetan, tadocizumab, talizumab, tefibazumab, tocilizumab, toralizumab, trastuzumab, tucotuzumab celmoleukin, tucusituzumab, umavizumab, urtoxazumab, and visilizumab.
ROUTES OF ADMINISTRATION
[00209] The compounds of the invention may be administered by any route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal. It will be appreciated that the preferred route may vary with for example the condition of the recipient. Where the compound is administered orally, it may be formulated as a pill, capsule, tablet, etc. with a pharmaceutically acceptable carrier or excipient. Where the compound is administered parenterally, it may be formulated with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form, as detailed below.
PHARMACEUTICAL FORMULATIONS
[00210] In order to use a compound of this invention for the therapeutic treatment (including prophylactic treatment) of mammals including humans, it is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition. According to this aspect of the invention there is provided a pharmaceutical composition that comprises a compound of this invention. In certain embodiments, the pharmaceutical composition comprises a compound of Formula 1 in association with a pharmaceutically acceptable diluent or carrier.
[00211] The pharmaceutical compositions of the invention are formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The therapeutically effective amount of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the disorder. The compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
[00212] The composition for use herein is preferably sterile. In particular, formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished, for
example, by filtration through sterile filtration membranes. The compound ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
[00213] Pharmaceutical formulations of the compounds of the present invention may be prepared for various routes and types of administration. For example, a compound of this invention having the desired degree of purity may optionally be mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation, a milled powder, or an aqueous solution. Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed. The pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8. Formulation in an acetate buffer at pH 5 is a suitable embodiment. The formulations may be prepared using conventional dissolution and mixing procedures. For example, the bull- drug substance (i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent) is dissolved in a suitable solvent in the presence of one or more excipients.
[00214] The particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal. In general, safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof. Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™,
PLURONICS™ or polyethylene glycol (PEG). The formulations may also include one or more stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). The active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Osol, A., supra. A "liposome" is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as a compound of Formula 1 and, optionally, an additional therapeutic agent) to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes. See also, Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
[00215] Sustained-release preparations of compounds of this invention may be prepared.
Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula 1, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Patent No. 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L- glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate) and poly-D-(-)-3-hydroxybutyric acid.
[00216] The pharmaceutical compositions of compounds of this invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may
conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
[00217] Formulations suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
[00218] The compositions of the invention may also be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder)
[00219] Suitable pharmaceutically-acceptable excipients for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate, granulating and disintegrating agents such as corn starch or algenic acid; binding agents such as starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl p-hydroxybenzoate, and anti-oxidants, such as ascorbic acid. Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents and procedures well known in the art.
[00220] Compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin, or olive oil.
[00221] Aqueous suspensions generally contain the active ingredient in finely powdered form together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol
anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives (such as ethyl or propyl p-hydroxybenzoate, anti-oxidants (such as ascorbic acid), coloring agents, flavoring agents, and/or sweetening agents (such as sucrose, saccharine or aspartame).
[00222] Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil (such as arachis oil, olive oil, sesame oil or coconut oil) or in a mineral oil (such as liquid paraffin). The oily suspensions may also contain a thickening agent such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set out above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
[00223] Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients such as sweetening, flavoring and coloring agents, may also be present.
[00224] The pharmaceutical compositions of the invention may also be in the form of oil-in- water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, or a mineral oil, such as for example liquid paraffin or a mixture of any of these. Suitable emulsifying agents may be, for example, naturally-occurring gums such as gum acacia or gum tragacanth, naturally-occurring phosphatides such as soya bean, lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides (for example sorbitan monooleate) and condensation products of the said partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening, flavoring and preservative agents.
[00225] Syrups and elixirs may be formulated with sweetening agents such as glycerol, propylene glycol, sorbitol, aspartame or sucrose, and may also contain a demulcent, preservative, flavoring and/or coloring agent.
[00226] Suppository formulations may be prepared by mixing the active ingredient with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Suitable excipients include, for example, cocoa butter and polyethylene glycols. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
[00227] Topical formulations, such as creams, ointments, gels and aqueous or oily solutions or suspensions, may generally be obtained by formulating an active ingredient with a conventional, topically acceptable, vehicle or diluent using conventional procedures well known in the art.
[00228] Compositions for transdermal administration may be in the form of those transdermal skin patches that are well known to those of ordinary skill in the art.
[00229] Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis disorders as described below.
[00230] The pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug. For example, an article for distribution can include a container having deposited therein the pharmaceutical formulation in an appropriate form. Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package. In addition, the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings. The formulations may also be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.
[00231] The invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore. Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
[00232] The amount of a compound of this invention that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the subject treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. In one embodiment, a suitable amount of a compound of this invention is administered to a mammal in need thereof. Administration in one embodiment occurs in an amount between about 0.001 mg/kg of body weight to about 60 mg/kg of body weight per day. In another embodiment, administration occurs in an amount between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several small doses for administration throughout the day. For further information on routes of administration and dosage regimes, see Chapter 25.3 in Volume 5 of Comprehensive Medicinal Chemistry (Corwin Hansch; Chairman of Editorial Board), Pergamon Press 1990, which is specifically incorporated herein by reference.
ARTICLES OF MANUFACTURE
[00233] In another embodiment of the invention, an article of manufacture, or "kit", containing materials useful for the treatment of the disorders described above is provided. In one embodiment, the kit comprises a container comprising a compound of this invention. Suitable containers include, for example, bottles, vials, syringes, blister pack, etc. The container may be formed from a variety of materials such as glass or plastic. The container may hold a compound of this invention or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
[00234] The kit may further comprise a label or package insert on or associated with the container. The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products. In one embodiment, the label or package inserts indicates that the composition comprising a compound of this invention can be used to treat a disorder mediated, for example, by AKT kinase. The label or package insert may also indicate that the composition can be used to treat other disorders.
[00235] In certain embodiments, the kits are suitable for the delivery of solid oral forms of a compound of this invention, such as tablets or capsules. Such a kit preferably includes a number of unit dosages. Such kits can include a card having the dosages oriented in the order of their
intended use. An example of such a kit is a "blister pack". Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms. If desired, a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered.
[00236] According to another embodiment, a kit may comprise (a) a first container with a compound of this invention contained therein; and (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound useful for treating a disorder mediated by AKT kinase. Alternatively, or additionally, the kit may further comprise a third container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
[00237] The kit may further comprise directions for the administration of the compound of this invention and, if present, the second pharmaceutical formulation. For example, if the kit comprises a first composition comprising a compound of this invention and a second pharmaceutical formulation, the kit may further comprise directions for the simultaneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof.
[00238] In certain other embodiments wherein the kit comprises a composition of this invention and a second therapeutic agent, the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet, however, the separate compositions may also be contained within a single, undivided container. In certain embodiments, the kit comprises directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
[00239] Accordingly, a further aspect of this invention provides a kit for treating a disorder or disease mediated by Akt kinase, wherein said kit comprises a) a first pharmaceutical composition comprising a compound of this invention or a pharmaceutically acceptable salt thereof; and b) instructions for use.
[00240] In certain embodiments, the kit further comprises (c) a second pharmaceutical composition, wherein the second pharmaceutical composition comprises a second compound
suitable for treating a disorder or disease mediated by Akt kinase. In certain embodiment comprising a second pharmaceutical composition, the kit further comprises instructions for the simultaneous, sequential or separate administration of said first and second pharmaceutical compositions to a patient in need thereof. In certain embodiments, said first and second pharmaceutical compositions are contained in separate containers. In other embodiments, said first and second pharmaceutical compositions are contained in the same container.
[00241] Although the compounds of Formula 1 are primarily of value as therapeutic agents for use in mammals, they are also useful whenever it is required to control AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases. Thus, they are useful as pharmacological standards for use in the development of new biological tests and in the search for new pharmacological agents.
[00242] The activity of the compounds of this invention may be assayed for AKT protein kinases, tyrosine kinases, additional serine/threonine kinases, and/or dual specificity kinases in vitro, in vivo, or in a cell line. In vitro assays include assays that determine inhibition of the kinase activity. Alternate in vitro assays quantitate the ability of the inhibitor to bind to kinases and may be measured either by radiolabelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where new inhibitors are incubated with known radioligands. These and other useful in vitro and cell culture assays are well known to those of skill in the art.
[00243] Although the invention has been described and illustrated with a certain degree of particularity, it is understood that the present disclosure has been made only by way of example, and that numerous changes in the combination and arrangement of parts can be resorted to by those skilled in the art without departing from the spirit and scope of the invention, as hereinafter claimed.
BIOLOGICAL EXAMPLES
AKT-1 Kinase Assay
[00244] The activity of the compounds described in the present invention may be determined by the following kinase assay, which measures the phosphorylation of a fluorescently- labeled peptide by full-length human recombinant active AKT-1 by fluorescent polarization using a commercially available IMAP kit.
[00245] The assay materials are obtained from an IMAP AKT Assay Bulk Kit, product
#R8059, from Molecular Devices, Sunnyvale, CA. The kit materials include an IMAP Reaction Buffer (5x). The diluted lx IMAP Reaction Buffer contained 10 raM Tris-HCl, pH 7.2, 10 raM MgCl2i 0.1% BSA. DTT is routinely added to a final concentration of 1 raM immediately prior to
use. Also included is IMAP Binding Buffer (5x), and IMAP Binding Reagent. The Binding Solution is prepared as a 1 :400 dilution of IMAP Binding Reagent into lx IMAP Binding Buffer.
[00246] The fluorescein-labeled AKT Substrate (Crosstide) has the sequence (Fl)-
GRPRTSSFAEG. A stock solution of 25 μΜ is made up in lx IMAP Reaction Buffer.
[00247] The plates used include a Costar 3657 (382-well made of polypropylene and having a white, v-bottom) that is used for compound dilution and for preparing the compound-ATP mixture. The assay plate is a Packard ProxiPlate™-384 F.
[00248] The AKT-1 used is made from full-length, human recombinant AKT-1 that is activated with PDK1 and MAP kinase 2.
[00249] To perform the assay, stock solutions of compounds at 10 mM in DMSO are prepared. The stock solutions and the control compound are serially diluted 1 :3 ten times into DMSO (5 of compound + 10 μΐ, of DMSO) to give 50x dilution series over the desired dosing range. Next, l-μΐ., aliquots of the compounds in DMSO are transferred to a Costar 3657 plate containing 49 \iL of 10.4 μΜ ATP in lx IMAP Reaction Buffer containing 1 mM DTT. After thorough mixing, 2.5-μΙ_- aliquots are transferred to a ProxyPlate™-384 F plate.
[00250] The assay is initiated by the addition of 2.5-μΙ. aliquots of a solution containing 200 nM of fluorescently-labeled peptide substrate and 20 nM AKT-1. The plate is centrifuged for 1 minute at 1000 g and incubated for 60 minute at ambient temperature. The reaction is then quenched by the addition of 15
of Binding Solution, centrifuged again and incubated for an additional 30 minutes at ambient temperature prior to reading on a Victor 1420 Multilabel HTS Counter configured to measure fluorescence polarization.
[00251] The compounds of Examples 1-175 were tested in the above assay and found to have an IC5o of less than <25 μΜ.
160 2-D 1.2789 1 3-E 0.069
130 2-E 0.0044 6 3-E 0.0737
129 2-E 0.0086 60 3-E 0.088
131 2-E 0.0239 59 3-E 0.1072
158 2-E 0.0419 3 3-E 0.1367
109 2-E 0.1436 21 3-E 0.1809
126 2-E 0.2186 32 3-E 0.1847
157 2-E 0.5117 20 3-E 0.2066
139 2-F 0.0077 51 3-E 0.2202
140 2-F 0.0195 11 3-E 0.2259
138 2-F 0.0611 35 3-E 0.2364
137 2-F 0.1 153 4 3-E 0.2367
136 2-F 0.4076 36 3-E 0.2419
116 2-F 0.6512 154 3-G 0.6607
PREPARATIVE EXAMPLES
[00252] In order to illustrate the invention, the following examples are included. However, it is to be understood that these examples do not limit the invention and are only meant to suggest a method of practicing the invention. Persons skilled in the art will recognize that the chemical reactions described may be readily adapted to prepare a number of other compounds of Formula 1, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention. For example, the synthesis of non-exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
[00253] In the examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Lancaster, TCI or Maybridge, and were used without further purification unless otherwise indicated. Tetrahydrofuran (TF1F), dichloromethane (DCM), toluene, and dioxane were purchased from Aldrich in Sure seal bottles and used as received.
[00254] The reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.
[00255] Ή NMR spectra were recorded on a Varian instrument operating at 400 MHz. 1H-
NMR spectra were obtained as CDC13, CD3OD, D20 or d6-DMSO solutions (reported in ppm), using tetramethylsilane (0.00 ppm) or residual solvent (CDC13: 7.25 ppm; CD3OD: 3.31 ppm; D20: 4.79 ppm; d6-DMSO: 2.50 ppm) as the reference standard. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, are reported in Hertz (Hz).
Examples
Example A
{ l-f4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl1-cyclobutyl}-carbamic acid tert-butyl ester
[00256] Step A: To a solution of 2,4-Dichloro-5-nitro-pyrimidine (5.00 g, 26.00mmol) in
THF (100 mL) was added the solution of [l-(4-Amino-phenyl)-cyclo-butyl]-carbamic acid tert- butyl ester (6.82 g, 26.00 mmol) in THF (20 mL) slowly at 0 °C. After addition, the reaction mixture was stirred at ambient temperature for 3 h. Then the reaction was concentrated under reduced pressure. The resulting residue was dissolved in ethyl acetate (250 mL) and washed with saturated sodium bicarbonate solution and brine. The organic layer was separated, dried over sodium sulfate, filtered and concentrated to afford { l-[4-(2-Chloro-5-nitro-pyrimidin-4-ylamino)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (9.50 g, 87% yield). 1H NMR (DMSO, 400 MHz), δ ppm: 10.41 (s, 1H), 9.14 (s, 1H), 7.61 (s, 1H), 7.48-7.46 (m, 2H), 7.43-7.41 (m, 2H), 2.41- 2.35 (m, 4H), 2.08-1.93 (m, 1H), 1.85-1.73 (m, 1H), 1.34 (s, 9H). MS (ESI+) e/z: 420.1/422.1 [M+l] +.
[00257] Step B: {l-[4-(2-Chloro-5-nitro-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-
carbamic acid tert-butyl ester (4.10 g, 9.78 mmol) was dissolved in THF (200 mL) and Raney- Ni (1.00 g) was added. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 4 h. The solvent was removed under reduced pressure and the resulting residue was dissolved in ethyl acetate (200 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography on silica gel (eluted by ethyl acetate: hexane=l :l to 3:1) to give the desired product: { l-[4-(5-Amino-2-chloro-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert- butyl ester. (3.5 g, 92% yield), MS (ESI+) e/z: 390.1/392.1 [M+l]+
[00258] Step C: To a solution of { l-[4-(5-Amino-2-chloro-pyrimidin-4- ylamino)-phenyl]- cyclo-butyl} -carbamic acid tert-butyl ester (2.5 g, 6.42 mmol) in acetic acid (30 mL) was added benzaldehyde (0.89 g, 8.35 mmol) and Cu(OAc)2 (0.58 g, 3.21 mmol). The reaction mixture was stirred at 100 °C for 2 h.. Then the solvent was removed under reduced pressure. The resulting residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with sat. NaHC03 aqueous solution, followed by brine, dried over sodium sulfate, filtered and concentrated to yield 2.50 g of { l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl] -cyclobutyl} -carbamic acid tert-butyl ester.
[00259] Ή NMR (DMSO, 400 MHz), δ ppm: 9.21 (s, 1H), 7.84-7.56 (m, 5H), 7.51-7.40 (m,
2H), 7.38-7.30 (m, 3 H), 2.47-2.40 (m, 4H), 2.08-1.97 (m, 1H), 1.87-1.75 (m, 1H), 1.34 (s, 9H).MS (ESI+) e/z: 476.2/478.2 [M+l]+. (Containing 15% by-product N-{ 1 -[4-(2-Chloro-8-phenyl-purin-9- yl)-phenyl]-cyclobutyl}-acetamide. MS (ESI+) e/z: 418.1/420.1 [M+l]+).
Example 1
1 - [4-(2,8-Diphenyl-purin-9-yl)-phenvH-cvclobutylamine
[00260] Step A: { l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (500 mg, 1.05 mmol), phenylboronic acid (256 mg, 2.10 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were added into a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was stirred at 100 °C under nitrogen atmosphere for 4 h. The solvents were removed under reduced pressure and the resulting residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by preparative TLC (ethyl acetate: Hexane=l :2) to
give desired product: { l-[4-(2,8-Diphenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (200 mg, 37% yield), MS (ESI+) e/z: 518.3 [M+l]+.
[00261] Step B: { l-[4-(2,8-Diphenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert- butyl ester (200 mg, 0.39 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford l-[4-(2,8-Diphenyl-purin-9-yl)-phenyl]-cyclobutylamine (52 mg, 32% yield). 1H NMR (MeOD, 400 MHz), δ ppm: 9.19 (s, 1H), 8.36-8.35 (m, 2H), 7.71- 7.69 (m, 2H), 7.63-7.60 (m, 2H), 7.57-7.48 (m, 2H), 7.43-7.40 (m, 6H), 2.80-2.75 (m, 2H), 2.59- 2.52 (m, 2H), 2.29-2.20 (m, 1H), 2.05-1.95 (m, 1H).
[00262] Examples 2-4 shown in Table 1 can also be made according to the above-described methods.
Table 1
Example 5
1 -r4-(8-Phenyl-2-piperazin- 1 -yl-purin-9-yl)-phenyl]-cvclobutylamine
[00263] Step A: To a solution of { l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclo- butylj-carbamic acid tert-butyl ester (70 mg, 0.147 mmol) in dioxane (20 mL) was added piperazine (63 mg, 0.737 mmol). Then the reaction mixture was stirred at 80 °C for 3h. The solvent was removed under reduced pressure, the resulting residue was dissolved in ethyl acetate (100 mL), washed with water (50 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product { l-[4-(8-Phenyl-2-piperazin-l-yl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (-65 mg) was used without further purification. MS (ESI+) e/z: 526.2 [M+l]+).
[00264] Step B: {l-[4-(8-Phenyl-2-piperazin-l-yl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-65 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford l-[4-(8-Phenyl-2~ piperazin-l-yl-purin-9-yl)-phenyl]- cyclobutylamine (30 mg, 48% yield over two steps). MS (ESI+) e/z: 426.2 [M+1] +. 'HNMR
(MeOD, 400MHz) δ ppm: 8.81 (s, 1H), 7.64 (d, J=8.4 Hz, 2H), 7.52-7.43 (m, 5H), 7.38-7.34 (m, 2H), 4.00 (t, J=4.8 Hz, 4H), 3.18 (t, J=5.0 Hz, 4H), 2.82-2.73 (m, 2H), 2.61-2.50 (m, 2H), 2.30-2.20 (m, 1H), 2.03-1.93 (m, 1H).
[00265] Example 6 shown in Table 2 can also be made according to the above-described methods.
Table 2
Example 7
l-(4-(2-methoxy-8-phenyl-9H-purin-9-vnphenyl)cyclobutanamine
[00266] Step A: To a solution of {l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclo- butyl}-carbamic acid tert-butyl ester (70 mg, 0.147 mmol) in methanol (20 mL) was added sodium methanolate (24 mg, 0.441 mmol). The reaction mixture was stirred at 80 °C for 3 h. Then the solvent was removed under reduced pressure, the resulting residue was dissolved in ethyl acetate (80 mL), washed with water (50 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product tert-butyl l-(4-(2-methoxy-8-phenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate (-65 mg) was used without further purification. MS (ESI+) e/z: 472.2 [M+l]+.
[00267] Step B: tert-butyl l-(4-(2-methoxy-8-phenyl-9H-purin-9-yl)phenyl)cyclo- butylcarbamate (-65 mg) was dissolved in ethyl acetate (20 mL), the solution was cooled to 0 °C and a solution of HCl in ethyl acetate (4 M, 5 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford l-(4-(2-methoxy-8-phenyl-9H-purin— 9-
yl)phenyl)cyclobutanamine (25 mg, 45% yield over two steps). MS (ESI+) e/z: 372.2 [M+1] +. 1 HNMR(MeOD,400MHz), δ ppm: 8.91 (s, 1H), 7.66-7.54 (m, 2H), 7.56-7.54 (m, 2H), 7.49-7.44 (m, 3H), 7.39-7.35 (m, 2H), 3.97 (s, 3H), 2.75-2.65 (m, 2H), 2.50-2.43 (m, 2H), 2.25-2.16 (m, 1H), 1.96-1.87 (m, 1H).
[00268] Example 8 shown in Table 3 can also be made according to the above-described methods.
Table 3
Example 9
1 - ( 9-[4-( 1 -Amino-cyclobutyl Vphenyl]-8-phenyl-9H-purin-2-yl } -pyrrolidin-2-one
[00269] Step A: { l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (50 mg, 0.11 mmol), pyrrolidin-2-one (14 mg, 0.16 mmol), Pd2(dba)3 (19 mg, 0.02 mmol), Xantphos (15 mg, 0.03 mmol) and Cs2C03 (68 mg, 0.21 mmol) were dissolved in dioxane (10 mL). The reaction mixture was stirred at 90 °C under nitrogen atmosphere for 6 h. Then the solvent was removed by under reduce pressure, the resulting residue was dissolved in ethyl acetate (30 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and
concentrated. The crude product was purified by prep-TLC (EtOAc:Hexane=l :l) to give (l-{4-[2- (2-Oxo-pyrrolidin-l-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (30 mg, 52% yield). MS (ESI+) e/z: 525.5 [M+l]+).
[00270] Step B: (l-{4-[2-(2-Oxo-pyrrolidin-l-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutyl)- carbamic acid tert-butyl ester (30 mg, 0.057 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford l-{9-[4-(l-Amino-cyclobutyl)- phenyl]-8-phenyl-9H-purin-2-yl}-pyrrolidin-2-one (20 mg, 83% yield). MS (ESI+) e/z: 425.5 [M+l]+. 1H NMR (MeOD, 400 MHz), δ ppm: 9.26 (s, 1H), 7.69-7.62 (m, 4H), 7.57-7.49 (m, 3H), 7.40 (t, J=7.8 Hz, 2H), 2.98 (s, 3H), 2.82-2.75 (m, 2H), 2.58-2.53 (m, 2H), 2.26-2.21 (m, 1H), 2.05- 1.92 (m, 1H).
Example 10
{ 9-[4-( 1 -Amino-cyclobutyl)-phenyll-8-phenyl-9H-purin-2-yl -methyl-amine
[00271] Step A: To a solution of { 1 -[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (150 mg, 0.32 mmol) in dioxane (4 mL) was added the aqueous methylamine amine solution (10 mL, 28% wt) at a sealed vessel. The reaction mixture was stirred at 100 °C overnight. The reaction mixture was diluted with ethyl acetate (80 mL), and washed with water (40 mL). The separated organic layer was dried over sodium sulfate, filtered and
concentrated. The crude product {9-[4-(l-Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl}- methyl-amine (-120 mg) was used in the step without purification. MS (ESI+) e/z: 471.4 [M+l]+).
[00272] Step B: { l-[4-(2-Methylamino-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-120 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford {9-[4-(l-Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2- yl}-methyl-amine (50 mg, 42% yield). MS (ESI+) e/z: 371.4 [M+lf. 1HNMR(MeOD,400MHz), δ ppm: 8.66 (s, 1H), 7.64-7.61 (m, 2H), 7.51-7.41 (m, 4H), 7.37-7.36 (m, 1H), 7.35-7.33 (m, 2H), 2.87 (s, 3H), 2.82-2.74 (m, 2H), 2.59-2.52 (m, 2H), 2.25-2.22 (m, 1H), 2.00-1.93 (m, 1H).
[00273] Examples 1 1 and 12 shown in Table 4 can also be made according to the above- described methods.
Table 4
Example B
2-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-4-(trimethylstannyl)-lH-imidazole
[00274] 4-iodo-2-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazole (600 mg, 1.77 mmol) was dissolved dichloromethane (25 mL), and the solution was cooled to -78 °C, followed by adding ethylmagnesium bromide (3 M in THF, 0.71 mL, 2.13 mmol) slowly. After addition, the
reaction mixture was stirred at -78 °C for 1 h. Then chlorotrimethylstannane (1 in THF, 2.1 mL, 2.13 mmol) was added slowly into the reaction mixture at the same temperature. After addition, the reaction mixture was allowed to be warmed to ambient temperature and stirred for another 2 h. The reaction mixture was poured into an aqueous saturated NH4CI solution, and extracted with ethyl acetate. The separated organic layer was washed with water twice and dried over sodium sulfate, filtered and concentrated to afford 2-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-4- (trimethylstannyl)-lH-imidazole (-420 mg), which was used in next step without further purification. MS (ESI+) e/z: 377.1 [M+l]+.
3-methyl- 1 -((2-(trimethylsilyl)ethoxy)methyl)-5-(trimethylstannyl)- 1 H-pyrazole
[00275] Step A: 3-methyl- l-((2-(trimethylsilyl)ethoxy)methyl)-l H-pyrazole (500 mg, 2.35 mmol) was dissolved in THF (18 mL), and the solution was cooled to -78°C, followed by adding butyllithium (2.5 M, 0.94 mL, 2.35 mmol) slowly. The reaction mixture was stirred at this temperature for 1 h. Then, chlorotrimethylstannane (1 M in THF, 2.4 mL, 2.35 mmol) was added into the reaction mixture. It was allowed to be warmed to ambient temperature and stirred overnight. The reaction mixture was poured into aqueous saturated NH4CI solution, and extracted with ethyl acetate. The separated organic layer was washed by water twice and dried over sodium sulfate, filtered and concentrated to afford 3-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-5- (trimethylstannyl)-l H-pyrazole (-300 mg), which was used without further purification. MS (ESI+) e/z: 377.2 [M+l]+.
3-methyl-5-(tributylstannyl)isoxazole
[00276] Step A: To a solution of nitroethane (0.15 g, 2.00 mmol) in toluene (6 mL) was added l-chloro-3-isocyanatobenzene (0.61 g, 4.00 mmol). The reaction mixture was stirred at 50 °C for 10 minutes followed by addition of triethylamine (10 mg, 0.1 mmol) and tributyl(ethynyl)stannane (0.60 g, 1.91 mmol). The reaction mixture was stirred at 50 °C overnight. After cooling, the solution was diluted with water and filtered through a celite pad. The resulting filtrate was extracted with toluene, the organic layer was dried over sodium sulfate, filtered and concentrated to afford 3-methyl-5-(tributylstannyl)isoxazole as a yellow oil (-300 mg), which was
used without further purification. MS (ESI+) e/z: 374.2 [M+l]+.
Example 13
l-(4-[2-(2-Methyl-3H-imidazol-4-yl -8-phenyl-purin-9-yll-pheny -cyclobutylamine
[00277] Step A: To a solution of {l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclo- butyl}-carbamic acid tert-butyl ester (150 mg, 0.316 mmol) in dioxane (4 mL) was added an 2- Methyl-l-(2-trimethylsilanyl-ethoxymethyl)-4-trimethylstannanyl-lH- imidazole (-0.632 mmol, in THF) and Pd(PPh3)4 (36 mg, 0.032 mmol). The reaction mixture was irradiated by microwave at 120 °C for 50 minutes. The reaction mixture was diluted with ethyl acetate, and washed with water. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product [l-(4-{2-[2-Methyl-3-(2-trimethylsilanyl-ethoxymethyl)-3H-imidazol-4-yl]-8-phenyl- purin-9-yl}-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (-150 mg) was used without further purification.
[00278] Step B: [l-(4-{2-[2-Methyl-3-(2-trimethylsilanyl-ethoxymethyl)-3H-imidazol-4- yl]-8-phenyl-purin-9-yl}-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (-150 mg) was dissolved in Methanol (10 mL), and aqueous HC1 (36%, 5 mL) was added into the solution. It was heated at 80 °C for 2 h. The reaction mixture was concentrated under reduced pressure. The resulting crude product was purified by preparative HPLC to afford l-{4-[2-(2-Methyl-3H- imidazol-4-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine (25 mg, 19% yield, two steps). 1 HNMR(MeOD,400 MHz), δ ppm:9.12 (s, 1H), 7.68 (d, J=8.4 Hz, 2H), 7.62-7.57 (m, 5H), 7.48 (t, J=7.4 Hz, 1H), 7.38 (t, J=7.6 Hz, 2H), 2.88-2.81 (m, 2H), 2.68-2.60 (m, 2H), 2.43 (s, 3H), 2.32- 2.20 (m, 1H), 2.10-2.00 (m, 1H).
[00279] Examples 14 and 15 shown in Table 5 can also be made according to the above- described methods.
Example 16
- 4-( 1 -Amino-cyclobutyl)-phenvn-8-phenyl-9H-purine-2-carboxylic acid methylamide
[00280] Step A: To a solution of { 1 -[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (250 mg, 0.526 mmol) in DMF (20 mL) were added methylamine hydrochloride (107 mg, 1.578 mmol), Pd(OAc)2 (35 mg, 0.158 mmol), Xantphos (107 mg, 0.184 mmol) and sodium carbonate (178 mg, 1.683 mmol). The reaction mixture was stirred at 100 °C under the atmosphere of carbon monoxide (balloon) overnight. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate, washed with water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product { l-[4-(2-Methylcarbamoyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-200 mg) was used directly in next step without further purification. MS (ESI+) e/z: 499.3 [M+l]+.
[00281] Step B: { l-[4-(2-Methylcarbamoyl-8-phenyl-purin-9-yl)-phenyl]-cyclo-butyl}- carbamic acid tert-butyl ester (-200 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The crude product was collected by filtration, which was purified by preparative HPLC separation to afford 9-[4-(l-Amino-cyclobutyl)-phenyl]-8- phenyl-9H-purine-2-carboxylic acid methyl-amide (83 mg, 40% yield, two steps). MS (ESI+) e/z: 399.3 [M+l]+. 1 HNMR(MeOD,400MHz), δ ppm:9.26 (s, 1H), 7.69-7.62 (m, 4H), 7.57-7.49 (m, 3H), 7.40 (t, J=7.8 Hz, 2H), 2.98 (s, 3H), 2.82-2.75 (m, 2H), 2.58-2.53 (m, 2H), 2.26-2.21 (m, 1H), 2.05-1.92 (m, 1H)
Example 17
l-[4-(8-Phenyl-purin-9-yl)-phenyl -cvclobutylamine
[00282] Step A: To a solution of {l-[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (250 mg, 0.526 mmol) in methanol (30 mL) was added Pd/C (200 mg, 10%wt). The mixture was stirred at 50 °C under the atmosphere of hydrogen (50 psi) for 12 h. The catalyst was removed by filtration, the cake was washed with methanol. The filtrate was concentrated to afford { l-[4-(8-Phenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-120 mg), which was used in next steps without further purification. MS (ESI+) e/z: 442.1 [M+l]+.
[00283] Step B: { l-[4-(8-Phenyl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-120 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was purified by preparative HPLC to afford l-[4-(8-Phenyl-purin-9-yl)-phenyl]-cyclobutylamine (50 mg, 28 % yield, two steps). MS (ESI+) e/z: 342.1 [M+l]+. 1 HNMR(MeOD,400 MHz), δ ppm:9.19 (s, 1H), 8.89 (s, 1H), 7.70-7.64 (m, 2H), 7.64-7.61 (m, 2H), 7.53-7.48 (m, 3H), 7.42-7.38 (m, 2H), 2.80- 2.73 (m, 2H), 2.65-2.50 (m, 2H), 2.27-2.19 (m, 1H), 1.99-1.91 (m, 1H).
Example 18
l-r4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl1-cvclobutylamine
[00284] Step A: To a. solution of { 1 -[4-(2-Chloro-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (250 mg, 0.526 mmol) in THF (20 mL) was added pyridin-2- ylzinc(II) bromide (1.1 mL, 1M in THF) and Pd(PPh3)4 (36 mg, 0.032 mmol). The reaction mixture was stirred at 80 °C under the atmosphere of nitrogen for 4 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate, washed with water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product { l-[4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-200 mg) was used directly without further purification. MS (ESI+) e/z: 519.5 [M+l]+.
[00285] Step B: { l-[4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-200 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford l-[4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutylamine (48 mg, 22%, two steps). MS (ESI+) e/z: 419.5 [M+l]+ 'HNMR MeOD^OO MHz), δ ppm:9.32 (s, 1H), 8.70-8.68 (m, 1H), 8.48-8.45 (m, 1H), 7.95 (t, J=4.0 Hz, 1H), 7.69-7.63 (m, 4H), 7.56-7.49 (m, 4H), 7.43-7.39 (m, 2H), 2.77-2.71 (m, 2H), 2.49-2.42 (m, 2H), 2.38-2.17 (m, 1H), 1.98-1.88 (m, 1H).
tert-butyl l-(4-(5-amino-2-phenylpyrimidin-4-ylamino)phenyl)cyclobutylcarbamate
[00286] I Step A: To a solution of {l-[4-(2-Chloro-5-nitro-pyrimidin-4-ylamino)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (8.40 g, 20.04 mmol) in dioxane (200 mL)/H2O(40ml) were added PhB(OH)2 (3.43g, 28.2mmol), Pd(dppf)Cl2 (0.74g, 0.91mmol), and Cs2CO3(13.02g, 39.98mmol). The reaction mixture was stirred at 90 °C under Nitrogen atmospher for 3 hours. Then the mixture was concentrated under reduced pressure, the resulting residue was dissolved in Ethyl acetate (200 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography (eluted by EtOAc:PE=l :l to 3:1), which gave 7.0 g of desired product { l-[4-(5-Nitro-2-phenyl-pyrimidin-4- ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. (7.0 g, 76% yield).1!-! NMR (CDC13, 400 MHz), δ ppm: 10.16 (s, 1H), 9.32 (s, 1H), 8.36-8.34 (t, J=4.0 Hz, 2H), 7.64 (d, J=8.0 Hz, 2H), 7.51-7.40 (m, 5H), 5.13 (s, 1H), 2.53-2.49 (t, J=8.0 Hz, 4H), 2.09-2.06 (t, J=6.0 Hz, 1H), 1.87-1.83 (m, 1H), 1.33 (s, 9H).
[00287] Step B: Compound { l-[4-(5-Nitro-2-phenyl-pyrimidin-4-ylamino)-phenyl]- cyclobutylj-carbamic acid tert-butyl ester (7.0 g, 15.18 mmol) was dissolved in THF (200 mL) and Raney- Ni (1.00 g) was added carefully. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 4 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to afford {l-[4-(5-Amino-2-phenyl-pyrimidin-4- ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. (6.54 g, 95% yield). LC/MS (ESI+): e/z: 432.2 [M+l]+.
Example 19
l-(4-(8-(4-methoxypyridin-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine
[00288] Step A: {l-[4-(5-Amino-2-phenyl-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (0.25 g, 0.58 mmol) was dissolved in acetic acid (30 mL), then 4- methoxy-pyridine-3-carbaldehyde (87.7 mg, 0.64 mmol) and Cu(OAc)2 (105 mg, 0.58 mmol) were added separately. The reaction mixture was stirred at 100 °C for 2 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with aqueous sat. NaHC03 solution, followed by brine, dried over sodium sulfate, filtered and concentrated to give crude desired product (-150 mg), which was used directly without further purification. LC/MS (ESI+): e/z: 549.2 [M+l]+.
[00289] Step B: (l-{4-[8-(4-Methoxy-pyridin-3-yl)-2-phenyl-purin-9-yl]-phenyl}- cyclobutyl)-carbamic acid tert-butyl ester (-150 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, which was further purified by preparative HPLC to give l-(4-(8-(4-methoxypyridin-3-yl)-2-phenyl-9H-purin- 9-yl)phenyl)cyclobutanamine (60 mg, 30% yield). LC/MS (ESI+): e/z: 449.2 [M+l]+, 451.2 [M+3]+. 1HNMR(MeOD,400 MHz), δ ppm:9.26 (s, 1H), 8.76 (s, 1H), 7.58 (d, J=6.0 Hz, 1H), 8.42- 8.39 (m, 2H), 7.71-7.68 (m, 2H), 7.59-7.57 (t, J=4.4 Hz, 2H),7.46-7.43 (m, 3H), 7.10 (d, J=6.0 Hz, 1H), 3.56 (s, 3H), 2.82-2.77 (m, 2H), 2.71-2.63 (m, 2H), 2.30-1.99 (m, 2H).
[00290] Examples 20 to 48 shown in Table 6 can also be made according to the above- described methods.
Table 6
9-r4-(l-Amino-cvclobutyl -phenyn-2-phenyl-9H-purine-8-carbonitrile
] Step A: Disulfurdichloride (6.6 mL, 101 mmol) was added to a solution of 2-
chloroacetonitrile (3.0 g, 40.2 mmol) in CH2C12 (10 mL) at room temperature, under an insert atomosphere of nitrogen, the resulting mixture was stirred at room temperature for 18 hours.the reaction mixture was concentrated in vacuo to get crude product 4,5-Dichloro-[l,2,3]dithiazol-l- ylium chloride (8 g crude) which was used without purification.
[00292] Step B: To a solution of { l-[4-(5-Amino-2-phenyl-pyrimidin-4-ylamino)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (180mg, 0.414mmol) in THF (20ml) was added 4,5- Dichloro-[l,2,3]dithiazol-l-ylium chloride (99 mg, 0.45 mmol), then the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated in vacuo to give crude product, which was purified by flash chromatography to give (l-{4-[5-(4-Chloro- [l,2,3]dithiazol-5-ylideneamino)-2-phenyl-pyrimidin-4-ylamino]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester. (150mg 65% yield). LC/MS (ESI+): 567.1 [M+H]+
[00293] Step C: (l-{4-[5-(4-Chloro-[l,2,3]dithiazol-5-ylideneamino)-2-phenyl-pyrimidin-4- ylamino]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (150 mg, 0.27 mmol) was dissolved in toluene (15 mL), the reaction mixture was stirred at reflux for 4 hours. The reaction mixture was concentrated in vacuo to afford {l-[4-(8-Cyano-2-phenyl-purin-9-yl)-phenyI]-cyclobutyl}-carbamic acid tert-butyl ester. LC/MS (ESI+): 467.2 [M+l]+
[00294] Step D: The compound { l-[4-(8-Cyano-2-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (80-160 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, dried in vacuo, to give target compounds as HCl salt. (15.4 mg)
Example 50
l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-(3-(trifluoromethoxy)phenyl)-9H-purin-9- vDphenvDcvclobutanamine
[00295] Step A: To a solution of compound { l-[4-(5-Amino-2-chloro-pyrimidin-4- ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (0.3 g, 0.77 mmol) in dioxane (20 mL)/H20 (4 ml) were added PhB(OH)2 (2.31 mmol), Pd(dppf)Cl2 (56.29 mg, 0.077 mmol), and Cs2C03 (501mg, 1.54mmol). The reaction mixture was stirred at 90 °C under nitrogen atmosphere
for 3 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate (40 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography (eluted by EtOAc:hexane=l :l to 3:1) to give the desired product (l-{4-[5-Amino-2-(3- trifluoromethoxy-phenyl)-pyrimidin-4-ylamino]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (200 mg, 20% yield). LC/MS (ESI+): (M+Na: 516.0).
[00296] Step B: (l-{4-[5-Amino-2-(3-trifluoromethoxy-phenyl)-pyrimidin-4-yl-amino]- phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (200 mg 0.77 mmol) was dissolved in acetic acid (30 mL), then 2-Methyl-2H-pyrazole-3-carbaldehyde (93.5 mg, 0.85 mmol) and Cu(OAc)2 (140 mg, 0.77 mmol) were added separately. The reaction mixture was stirred at 100 °C for 2 h. LC/MS showed there was about 40% desired product (l-{4-[8-(2-Methyl-2H-pyrazol-3-yl)-2-(3- trifluoromethoxy-phenyl)-purin-9-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester in the reaction mixture. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with sat. NaHC03 aqueous solution, followed by brine, dried over sodium sulfate, filtered and concentrated to give 150 mg of crude desired product with 80% purity, which was used directly without further purification. LC/MS (ESI+): e/z: 606.2 [M+l]+.
[00297] Step C: (l-{4-[2-(2-Fluoro-phenyl)-8-(2-methyl-2H-pyrazol-3-yl)-purin-9-yl]- phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (150 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, which was further purified by preparative HPLC to give l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-(3- (trifluoromethoxy)phenyl)- 9H-purin-9-yl)phenyl)cyclobutanamine (26.9 mg, 34% yield). LC/MS (ESI+): e/z: 506.2 [M+l]+. Ή MR (MeOD, 400 MHz), δ ppm: 9.32 (s, 1H), 8.41 (d, J=6.8 Hz, 1H), 8.26 (s, 1H), 7.77 (d, J=8.4 Hz, 2H), 7.64 (d, J=8.0 Hz, 2H), 7.57-7.53 (t, J=7.8 Hz, 1H), 7.42- 7.37 (t, J=10.0 Hz, 2H), 6.02 (d, J=1.2 Hz, 1H), 4.27 (s, 3H), 2.88-2.81 (m, 2H), 2.62-2.55 (m, 2H), 2.32-2.27 (m, 1H), 2.05-2.00 (m, 1H).
[00298] Examples 51 to 56 shown in Table 7 can also be made according to the above- described methods.
anamine 7.64 (d, J=8.4 Hz, 2H),
7.42 (s, 1H),
7.26 (d, J=8.4 Hz, 2H),
6.03 (s, 1H), 4.25 (s, 3H),
2.91-2.84 (m, 2H),
2.64-2.57 (m, 2H),
2.39 (s, 3H),
2.31-2.16 (m, 1H),
2.08-2.03 (m, 1H).
Exam le F
tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl -9H-purin-9-yl phenyl)cyclobutylcarbamate
[00299] Step A: The tert-butyl l-(4-(5-amino-2-chloropyrimidin-4-ylamino)- phenyl)cyclobutylcarbamate (3.00 g, 7.71 mmol) was dissolved in acetic acid (50 mL), then picolinaldehyde (1.23 g, 11.5 mmol) and Cu(OAc)2 (1.4 g, 7.71 mmol) were added separately. The reaction mixture was stirred at 80 °C for 2 h. The reaction mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with aqueous sat. NaHC03 solution, followed by brine, dried over sodium sulfate, filtered and concentrated to give desired title compound: tert-butyl l-(4-(2-chIoro-8-(pyridin-2-yl)-9H-purin-9- yl)phenyl)cyclobutylcarbamate (1.8 g, 49% yield). LC/MS (ESI+): 477.0/479.1 [M+l]+.
Example 57
l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine
[00300] Step A: tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)- cyclobutylcarbamate (100 mg, ) was dissolved in ethyl acetate (30 mL), the solution was cooled to
0 °C and HCl in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The crude product was collected by filtration, which was further purified by preparative HPLC to afford the desired product l-(4-(2-chloro-8-(pyridin-2-yl)- 9H-purin-9- yl)phenyl)cyclo-butanamine.(24.5 mg, 31% yield). Ή NMR (MeOD, 400 MHz), δ ppm: 9.10 (s, 1H), 8.38 (d, 1H, J=3.6 Hz), 8.15 (d, 1H, J=8.0 Hz), 7.98-7.94 (m, 1H), 7.30 (d, 2H, J=8.4 Hz), 7.47-7.45 (m, 3H), 2.77-2.72 (m, 2H), 2.53-2.46 (m, 2H), 2.21-2.14 (m, 1H), 1.97 -1.89 (m, 1H). MS (ESI+): 377.1/379.1 [M+l]+.
Exam le 58
1 -(4-(2-( 1 H-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine
[00301] Step A: Tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)- phenyl)cyclobutylcarbamate (700 mg, 1.47 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- lH-pyrazole (342 mg, 1.76 mmol), Pd(dppf)Cl2 (109.5 mg, 0.15 mmol), and Cs2C03 (1.4 g, 4.41 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The resulting mixture was charged with nitrogen thrice and stirred at 100 °C under nitrogen atmosphere for 4 h. TLC showed the stating material was consumed completely. Then the solvents were removed under reduced pressure, the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated to give720 mg of crude desired product tert-butyl l-(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)- cyclobutylcarbamate with purity about 50%, which was used directly without further purification. LC/MS (ESI+): 509.3 [M+l]+.
[00302] Step B: The tert-butyl l-(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9- yl)phenyl)cyclobutylcarbamate (700 mg crude) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and HCl in ethyl acetate (4 M, 10 mL) was added slowly. The resulting solution was stirred for 2 h. The product was collected by filtration, which was further purified by preparative HPLC to give l-(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9- yl)phenyl)cyclobutanamine (40 mg, 12.5 % yield, two steps). 1H NMR (MeOD, 400 MHz), δ ppm: 9.16 (s, 1H), 8.39-8.38 (m, 1H), 8.19 (s, 2H), 8.13-8.11 (m, 1H), 7.99-7.94 (m, 1H), 7.65 (d, 2H, J=8.4Hz), 7.53 (d, 2H, J=8.4 Hz), 7.47-7.40 (m, 1H), 2.86-2.78 (m, 2H), 2.60-2.53 (m, 2H), 2.29-
2.22(m, IH), 2.02-1.97 (m, IH). MS (ESf ): 409.2 [M+l]+.
[00303] Examples 59 and 60 shown in Table 8 can also be made according to the above- described methods.
Table 8
Example 61
2-(4-(9-(4-( 1 -aminocvclobutyl)phenyl)-8-(pyridin-2-yl -9H-purin-2-vn- 1 H-pyrazol- 1 -yDethanol
[00304] Step A: A solution of tert-butyl 1 -(4-(2-(lH-pyrazol-4-yl)-8-(pyridin-2-yl)-9H- purin-9-yl)phenyl)cyclobutylcarbamate (400 mg, 0.787 mmol) in dioxane(10 mL) and oxirane (10 mL) was stirred at 10 °C for 3 days.. The reaction mixture was concentrated to dryness to give the crude desired product: tert-butyl l-(4-(2-(l-(2-hydroxyethyl)-lH-pyrazol-4-yl)-8-(pyridin~2-yl)- 9H-purin-9- yl)phenyl)cyclobutylcarbamate with 70% purity. LC/MS (ESI+): 553.3 [M+l]+.
[00305] Step B: tert-butyl 1 -(4-(2-( 1 -(2-hydroxyethyl)- 1 H-pyrazol-4-yl)-8-(pyridine-2-yl)-
9H-purin-9-yl)phenyl)cyclobutylcarbamate (200 mg, 70% purity) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C, then HC1 in ethyl acetate (4M, 10 mL) was added slowly. The resulting solution was stirred for 2 hrs. The product was collected by filtration, which was further purified by preparative HPLC to give the title compound: 2-(4-(9-(4-(l-aminocyclobutyl)phenyl)-8- (pyridin-2-yl)-9H-purin-2-yl)-lH-pyrazol-l-yl)ethanol (35 mg, 10% yield, two steps). 1H NMR (MeOD, 400 MHz), <5 ppm: 9.17 (s, 1H), 8.39-8.38 (m, lH), 8.27 (s, 1H), 8.15-8.13 (m, 1H), 8.06 (s, 1H), 7.99-7.94 (m, 1H), 7.65 (d, 2H, J=8.4Hz), 7.53 (d, 2H, J=8.4 Hz), 7.46-7.43 (m, 1H), 4.27- 4.25 (m, 2H), 3.92-3.89 (m, 2H), 2.89-2.82 (m, 2H), 2.67-2.60 (m, 2H), 2.30-2.27(m, lH), 2.04- 2.02 (m, 1H). MS (ESI+): 453.2 [M+l]+.
Example 62
N 1 -(9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)ethane- 1 ,2-diamine
[00306] Step A: A mixture of tert-butyl l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9- yl)phenyl) cyclobutylcarbamate (100 mg, 0.21 mmol) in ethane- 1,2-diamine (1 mL) was put in a sealed tube and stirred at 100 °C for 2 h. The reaction mixture was concentrated and the resulting residue was dissolved in ethyl acetate (50 mL), washed with water. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product was purified by preparative
HPLC to give Nl-(9-(4-(l-aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)ethane-l,2- diamine.(45 mg 54% yield).1H NMR (MeOD, 400 MHz), δ ppm: 8.80 (s, IH), 8.33-8.32 (m, IH), 8.00-7.99 (m, IH), 7.93-7.89 (m, IH), 7.57-7.48 (m, IH), 7.58 (d, 2H, J=8.4 Hz), 7.42-7.38 (m, 3H), 3.65-3.64 (m, 2H), 3.16-3.13 (m, 2H), 2.77-2.70 (m, 2H), 2.57-2.50 (m, 2H), 2.22-2.21(m, IH), 1.96-1.91 (m, IH). MS (ESI+): 401.5 [M+l]+
Example 63
l-(9-(4-(l-aminocvclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)pyrrolidin-3-ol
[00307] Step A: The compound { l-[4-(2-Chloro-8-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutyl}- carbamic acid tert-butyl ester (200 mg, 0.42 mmol), pyrrolidin-3-ol (103 mg, 0.84 mmol) and K2C03 (176 mg, 0.84 mmol) were dissolved in l-methyl-pyrrolidin-2-one (10 mL). The reaction mixture was stirred at 120 °C under nitrogen atmosphere overnight. The mixture was diluted with ethyl acetate (50 mL) and washed with brine (20 mL). The organic layer was dried over sodium sulfate, filtered and concentrated to give the (l-{4-[2-(3-Hydroxy-pyrrolidin-l-yl)-8- pyridin-2-yl-purin-9-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester. (210 mg, 95%,yield). LC/MS (ESI+): 528.2, [M+l]+ 528.2 [M+3]+.
[00308] Step B: (l-{4-[2-(3-Hydroxy-pyrrolidin-l-yl)-8-pyridin-2-yl-purin-9-yl]-phenyl}- cyclobutyl)-carbamic acid tert-butyl ester was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4M, 10 mL) was added slowly. The resulting solution was stirred for 2 hrs. Then the solvent was removed by concentration, and the residue was purified by preparative HPLC to give l-(9-(4-(l-aminocyclobutyl)phenyl)-8-(pyridin- 2-yl)-9H-purin-2-yl)pyrrolidin-3-ol. 1H NMR (MeOD, 400 MHz), «5 ppm: 8.82 (s, IH), 8.33-8.32 (m, IH), 8.10-8.02 (m, IH), 7.93-7.89 (m, IH), 7.63 (d, 2H, J=8.4 Hz), 7.50 (d, 2H, J=8.4 Hz), 7.44-7.41 (m, IH), 4.52 (s, IH), 3.70-3.59 (m, 4H), 2.88-2.80 (m, 2H), 2.67-2.60 (m, 2H), 2.30- 2.28(m, IH), 2.05-2.03 (m, IH), 1.96-1.94 (m, 2H). MS (ESI+): 428.5 [M+l]+, 429.5 [M+l]+.
N-(9-(4-n-aminocvclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)methanesulfonamide
[00309] Step A: The compound { l-[4-(2-Chloro-8-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (150 mg, 0.315 mmol), Methanesulfonamide (60 mg, 0.63 mmol), Pd2(dba)3 (29 mg, 0.032 mmol), Xantphos (18 mg, 0.032 mmol) and Cs2C03 (308 mg, 0.945 mmol) were dissolved in dioxane (10 mL). The reaction mixture was stirred at 100 °C under nitrogen atmosphere for overnight. LC-MS indicated the reaction worked well. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (30 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated to give { l-[4-(2- Methanesulfonylamino-8-pyridin-2-yl-purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (107 mg, yield: 63.4%). LC/MS (ESI+): 536.1 [M+l]+,538.1 [M+3]+
[00310] Step B: { l-[4-(2-Methanesulfonylamino-8-pyridin-2-yl-purin-9-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (107 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4M, 10 mL) was added slowly. The resulting solution was stirred for 2 hrs. Then the solvent was removed by concentration, and the residue was purified by preparative HPLC to give N-(9-(4-(l-aminocyclobutyl)phenyl)-8- (pyridin-2-yl)-9H-purin-2-yl)methanesulfonamide (37 mg, 42.5% yield).1!! NMR (MeOD, 400 MHz), δ ppm: 8.98 (s, 1H), 8.35 (d, 1H, J=4.8 Hz), 8.09 (d, 1H, J=8.0 Hz), 7.96-7.92 (m, 1H), 7.60 (d, 1H, J=8.4Hz), 7.8 (d, 2H, J=8.8 Hz), 7.44-7.41 (m, 1H), 3.28 (s, 3H), 2.80-2.73 (m, 2H), 2.60- 2.55 (m, 2H), 2.24-2.22(m, 1H), 1.96-1.94 (m, 1H). MS (ESI+): 436.1 [M+l]+ ,438.1 [M+3]+
Example 65
9-(4-( 1 -aminocyclobutyl)phenyl)-8-phenyl- 1 H-purin-6(9H)-one
[00311] Step A: The solution of 4,6-dichloro-5-nitropyrimidine 500mg, 2.6mmol and tert-
butyl l-(4-aminophenyl)cyclobutylcarbamate 609mg, 2.32mmol in THF (20 mL) was stirred at R.T. for 2hrs until TLC (Hexane/EtOAc=3:l) indicated the consumption of 4,6-dichloro-5- nitropyrimidine. The reaction mixture was concentrated to afford the crude residue, which was purified by flushing chromatography on silica gel (eluting with Hexane/EtOAc= 100/ 1-3/1) to give the product tert-butyl l-(4-(6-chloro-5-nitropyrimidin-4-ylamino)phenyl)cyclobutylcarbamate (350mg, 30.8%). MS (ESI+): 442.1 [M+Naf
[00312] Step B: The solution of tert-butyl l-(4-(6-chloro-5-nitropyrimidin-4-ylamino) phenyl)cyclobutylcarbamate (250mg, 0.6mmol) and Raney Ni (50 mg) in THF (20 mL) was stirred under H2 (balloon) atmosphere for 20 min until TLC (Hexane/EtOAc=l:l) showed the consumption of STM. The reaction was filtered through celite and the filtrate was concentrated under reduced pressure to give tert-butyl l-(4-(5-amino-6- chloropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (180 mg, 77.6%, yield). MS (ESI+): 390.1 [M+lf, 392.1 [M+3]+
[00313] Step C: To a solution of tert-butyl l-(4-(5-amino-6- chloropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (200mg, 0.5mmol) in HOAc (20 mL) was added Cu(OAc)2 46.7mg, 0.26mmol and benzaldehyde (109mg, lmmol). The reaction mixture was stirred at 80°C for lh. The reaction mixture was concentrated and purified by flushing chromatography on silica gel (eluting with Hexane/EtOAc=100/l~3/l) to give tert-butyl l-(4-(6-chloro-8-phenyl-9H- purin-9- yl)phenyl)cyclobutylcarbamate (80mg, 32.8% yield). MS (ESf ): 477.1 [M+H]+ ; 479.1 [M+3]+
[00314] Step D: The solution of tert-butyl l-(4-(6-chloro-8-phenyl-9H-purin-9-yl) phenyl)cyclobutylcarbamate (40mg, 0.08mmol) in HC1 (2 mL, 10% wt) was stirred at 80 °C for lh. The reaction solution was concentrated to give title compound 9-(4-(l-aminocyclobutyl)phenyl)-8- phenyl-lH-purin-6(9H)-one (30mg) as HC1 salt. 1H NMR (MeOD, 400 MHz), δ ppm: 8.04 (s, 1H), 7.67-7.65 (d, J=8.0 Hz, 2H), 7.53-7.50 (m, 3H), 7.43-7.41 (m, 1H), 7.36-7.32 (m, 2H), 2.84- 2.79(m, 2H), 2.64-2.63 (m, 2H), 2.29-2.2.20 (m, 1H), 2.05-1.95 (m, 1H), MS (ESI+): 358.2 [M+l]+, 360.2 [M+3]+
[00315] Example 66 shown in Table 9 can also be made according to the above-described methods.
Example 67
9-(4-(l-aminocvclobutyl)phenyl')-N.N-dimethyl-8-phenyl-9H-purin-6-amine
[00316] Step A: To the solution of 4, 6-Dichloro-5-nitro-pyrimidine (1 g, 5.2 mmol) in THF
(20 mL) was added a solution of [l-(4-amino-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (1.36 g, 5.2 mmol) in THF (20 mL) dropwise at 0 °C. After addition, the reaction mixture was stirred at 0 °C for another 2 h. TLC showed the starting material was consumed completely. The reaction mixture was diluted with ethyl acetate (30 mL) and washed with brine (20 mL) twice. The organic phase was concentrated to give 2 g of crude {l-[4-(6-Chloro-5-nitro-pyrimi-din-4- ylamino)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. ( 95% yield)
[00317] Step B: To a solution of { l-[4-(6-Chloro-5-nitro-pyrimidin-4-ylamino)- phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (2 g, 4.77 mmol) and dimethyl-amine (0.77 g, 9.54 mmol) in THF (50 mL) was added DIPEA (3.1 g, 23.9 mmol). The mixture was stirred 20 °C for 2 h. LC-MS showed the starting material was consumed completely. The reaction mixture was diluted with ethyl acetate (30 mL) and washed with brine (2 X 20 mL). The organic phase was concentrated to give 2 g of the crude product { l-[4-(6-Dimethylamino-5-nitro-pyrimidin-4- ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. (98%, yield). MS (ESI+): 451.1 [M+Na]+ 453.1 [M+Na+2]+
[00318] Step C: The compound {l-[4-(6-Dimethylamino-5-nitro-pyrimidin-4-ylamino)-
phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (2.0 g, 4.67 mmol) was dissolved in THF (60 mL) and Raney- Ni (2.00 g) was carefully added. The reaction mixture was stirred at ambient temperature under the atmosphere of H2 (balloon) for 1 h.. After filtration over ciliate pad, the fitrate was concentrated to give 1.8 g of desired product { l-[4-(5-Amino-6-dimethylamino- pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. ( 96.8%, yield). MS (ESI+): 399.3 [M+l]+, 401.3 [M+l]+
[00319] Step D: The {l-[4-(5-Amino-6-dimethylamino-pyrimidin-4-ylamino)-phenyl]- cyclo-butyl}-carbamic acid tert-butyl ester (500 mg, 1.29 mmol) was dissolved in acetic acid (20 mL), and benzaldehyde (204 mg, 1.93 mmol) and Cu(OAc)2 (233 mg, 1.29 mmol) were added separately. The reaction mixture was heated at 80°C for 2 hrs. The solvent was removed by concentration, and the residue was dissolved in ethyl acetate (50 mL), washed with H3.H2O (20 mL) and brine (20 mL). The organic phase was concentrated to give { l-[4-(6-Dimethylamino-8- phenyl-purin-9-yl)-phenyl]-cyclobutyl} -carba-mic acid tert-butyl ester. (600 mg, 96% yield). LC MS (ESI+): 485.2 [M+l]+ , 487.2 [M+3]+
[00320] Step E: { l-[4-(6-Dimethylamino-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (300 mg, 0.62 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0°C and the solution of HC1 in ethyl acetate (10 mL, 4M) was slowly added. The resulting mixture was stirred for 2 hrs. After filtration, the solid was purified by preparative HPLC to give (9-[4-(l -Amino- cyclobutyl)-phenyl]-8-phenyl-9H-purin-6-yl}-dimethyl-amine. (82 mg, 34.4% yield). Ή NMR (DMSO-d6, 400 MHz), δ ppm: 8.21 (s, 1H), 7.45 (d, 2H, J=6.8 Hz), 7.43 (d, 2H, J=6.8 Hz), 7.45-7.38 (m, 5H), 2.51 (s, 6H), 2.65-2.51 (m, 2H), 2.36-2.39 (m, 2H), 2.12-2.08 (m, 1H), 1.81 -1.76 (m, 1H). MS (ESI+): 385.1 [M+l]+ , 387.1 [M+3]+
Example 68
1 -(4-(6-( 1 -methyl- 1 H-pyrazol-4-yl)-8-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine
[00321] Step A: To a solution of 4,6-dichloro-5-nitropyrimidine (2.00 g, 10.3 mmol) in THF
(40 mL) was slowly added a solution of tert-butyl l-(4-aminophenyl)cyclo- butylcarbamate (2.7 g, 10.3 mmol) in THF (60 mL) drop wise at 0°C. After addition, the reaction mixture was stirred at 0°C for 3 h. TLC showed the starting material was consumed completely. The mixture was washed
brine (30 mL) twice and concentrated. The crude product was purified by column (eluted by Ethyl acetate : Hexane =1 :20) to give tert-butyl l-(4-(6-chloro-5-nitropyrimidin-4- ylamino)phenyl)cycIobutylcarbamate.( 1.5 g, 37.5% yield).
[00322] Step B: The tert-butyl l-(4-(6-chloro-5-nitropyrimidin-4-ylamino)phenyl)- cyclobutylcarbamate (1.5 g, 3.58 mmol) was dissolved in THF (50 mL) and Raney- Ni (1.50 g) was carefully added. The reaction mixture was stirred at 20°C under the atmosphere of H2 (balloon) for 1 h.. After filtration ciliate pad, the filtrate was concentrated to give tert-butyl l-(4-(5-amino-6- chloropyrimidin-4-ylamino)phenyl)cyclobutylcarbamate. (1.3 g ,93% yield:). MS (ESI+): 412.1 [M+Na]+, 414.1 [M+Na+2]+
[00323] Step C: The tert-butyl l-(4-(5-amino-6-chloropyrimidin-4-ylamino)phenyl)- cyclobutylcarbamate (200 mg, 0.573 mmol), l-Methylpyrazole-4-boronic acid pinacol ester (143 mg, 0.688 mmol), Pd(dppf)Cl2 (40 mg, 0.057 mmol), and Cs2C03 (560 mg, 1.72 mmol) were dissolved in a mixed solution (dioxane: 10 mL, water: 2 mL). The reaction mixture was charged with nitrogen thrice and heated at 80°C for 2 h. The reaction mixture was diluted with ethyl acetate (20 mL), washed with brine (10 mL), concentrated and purified by column separation (eluted by EA:PE=1 :1), to give tert-butyl l-(4-(5-amino-6-(l -methyl- lH-pyrazol- 4-yl)pyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (100 mg 40%, yield:). MS (ESf ): 436.2 [M+l]+, 438.2
[M+l]+
[00324] Step D: The compound tert-butyl l-(4-(5-amino-6-(l -methyl- lH-pyrazol-4- yl)pyrimidin-4-ylamino)phenyl)cyclobutylcarbamate (100 mg, 0.23 mmol) was dissolved in acetic acid (10 mL), benzaldehyde (36 mg, 0.34 mmol) and Cu(OAc)2 (42 mg, 0.23 mmol) were added separately. The reaction mixture was heated at 80°C for 2 h. The solvent was removed under reduced pressure, the residue was diluted in ethyl acetate (30 mL), washed with NH3.H2O (10 mL, 10%) and brine (2 xl5 mL). The organic layer was concentrated to give tert-butyl l-(4-(6-(l- methyl-lH-pyrazol-4-yl)-8-phenyl-9H- purin-9-yl)phenyl)cyclobutylcarbamate (115 mg, 95.8% yield). MS (ESI+): 522.3 [M+lf, 524.3 [M+3]+
[00325] Step E : The tert-butyl 1 -(4-(6-( 1 -methyl- 1 H-pyrazol-4-yl)-8-phenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate (115 mg, 0.22 mmol) was dissolved in ethyl acetate (20 mL), the solution was cooled to 0 °C and a solution of HC1 in ethyl acetate (10 mL, 4M) was slowly added. The resulting solution was stirred for 2 h. The solvent was removed by concentration, and the residue was purified by preparative HPLC to give l-(4-(6-(l -methyl- lH-pyrazol-4-yl)-8-phenyl- 9H-purin-9-yl)phenyl) cyclobutanamine. (60 mg, 64% yiekf/H NMR (MeOD, 400 MHz), δ ppm: 8.83 (s, IH), 8.72 (s, IH), 8.58 (s, IH), 7.71-7.66 (m, 4H), 7.55 (d, 2H, J=8.4 Hz), 7.40-7.36 (m, IH), 2.85-2.81 (m, 2H), 2.66-2.61 (m, 2H), 2.29-2.26 (m, IH), 2.01-1.96 (m, IH). MS (ESI+): 422.3
[M+l]+, 424.3 [M+3]+
Example 69
l- 4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl1-cvclobutylamine
[00326] Step A: To the solution of 2,4-Dichloro-6-methyl-5-nitro-pyrimidine (2.08 g, lO.OOmmol) in THF (50 mL) at -78 °C was slowly added the solution of [l-(4-Amino-phenyl)- cyclobutyl]-carbamic acid tert-butyl ester (2.62 g, 10.00 mmol) in THF (20 mL). The reaction mixture was stirred at ambient temperature for 30 min.. Then it was concentrated to remove the solvent. The obtained residue was dissolved in ethyl acetate (250 mL) and washed by saturated. NaHC03 solution, followed by brine. The separated organic layer was dried over sodium sulfate, filtered and concentrated to give { l-[4-(2-Chloro-6-methyl-5-nitro-pyrimidin-4-ylamino)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester. (4.3 g, 98%yield). MS (ESI+) e/z: 370.9 / 372.9 [M+l]+.
[00327] Step B: { l-[4-(2-Chloro-6-methyl-5-nitro-pyrimidin-4- ylamino)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (4.3 g, 9.78 mmol) was dissolved in MeOH (100 mL) and Pd/C (1.00 g) was added. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 12 h.. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (200 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated to give { l-[4-(5-Amino-6-methyl-pyrimidin-4- ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. (3.7 g, 97% yield) MS (ESI+) e/z: 370.9 / 372.9 [M+l]+.
[00328] Step C: The { l-[4-(5-Amino-6-methyl-pyrimidin-4-ylamino)-phenyl]- cyclobutyl}- carbamic acid tert-butyl ester (2.0 g, 5.4 mmol) was dissolved in acetic acid (20 mL), and benzaldehyde (0.74 g, 7 mmol) and Cu(OAc)2 (1.0 g, 5.4 mmol) were added separately. The reaction mixture was heated at 80 °C for 2 h. LC/MS showed there were { l-[4-(6-Methyl-8-phenyl- purin-9-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by saturated. NaHC03 solution, followed by brine, dried over sodium sulfate, filtered and concentrated, purified by TLC to give { l-[4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester. 0.3 g, MS (ESI+) e/z: 370.9 [M+l]+, 372.9 [M+l]+.
[00329] Step D: { l-[4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester ( 0.3 g ) was dissolved in MeOH (30 mL), then added HC1 (aq) (4M, 8 mL) in drops. The mixture was heated to 80 °C, and stirred for 50 min. The product was purified by preparative HPLC to give (160 mg). l-[4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutylamine. Ή-NMR (400 MHz, CDC13) δ: 9.18 (s, 1H), 7.79 (d, J = 8.8Hz, 2H), 7.71-7.45 (m, 7H), 2.91 (s, 3H), 2.80- 2.75 (m, 2H), 2.59-2.52 (m, 2H), 2.29-2.20 (m, 1H), 2.05-1.99 (m, 1H). MS (ESI+) e/z: 355.8 [M+l]+, 357.8 [M+3]+
Example 70
9-(4-(l-aminocvclobutyl)phenyl)-2.8-diphenyl-lH-purin-6(9H)-one
[00330] Step A: To a suspension of 2,4,6-trichloro-5-nitropyrimidine (0.8g, 3.5mmol) in
THF (30ml) was added [l-(4-Amino-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (0.827g, 3.15mmol) at 15°C. The mixture was stirred for 30min at 15°C. The reaction mixture was concentrated and was purified by flushing chromatography on silica gel (eluting with Hexane EtOAc=100/l~3/l) to give tert-butyl l-(4-(2,6-dichloro-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamateas a yellow solid (0.45g, 31.5% yield). 'HNMR (CDCI3, 400 MHz): S9AS(s, 1H), 7.49 (s, 4H), 5.12 (s, ΙΗ,), 2.55-2.51(m, 4Η), 2.13-2.04(m, 1H),1.89-I.86(m, lH),1.42(s, 9H); LC/MS: (ESI+) e/z: 454.3 [M+l]+, 456.3 [M+3]+
[00331] Step B: To a solution of tert-butyl l-(4-(2,6-dichloro-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (0.45 g, 1 mmol) in THF (20ml) was added MeONa (53 mg, 1 mmol) in MeOH(l mL). The mixture was stirred at 55 °C for 30mins.The reaction mixture was purified by preparative TLC to tert-butyl l-(4-(2-chloro-6-methoxy-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (120 mg, 26.6% yield). LC/MS: (ESI+) e/z: 472 [M+Na]+, 474 [M+Na+2]+
[00332] Step C: To a solution of tert-butyl l-(4-(2-chloro-6-methoxy-5-nitropyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (100 mg, 0.222 mmol) in THF (10 mL) was added Raney Ni (50 mg) under nitrogen. The suspension was degassed under vacuum and purged with H2 (balloon). The reaction mixture was stirred at RT for 10 mins. The reaction mixture was purified by
preparative TLC to give tert-butyl l-(4-(5-amino-2-chloro-6-methoxypyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (84 mg, 90%). LC-MS: (ESI+) e/z: 442 [M+Na]+, 444 [M+Na+2]+
[00333] Step D: To a solution of tert-butyl l-(4-(5-amino-2-chloro-6-methoxypyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (84 mg, 0.2 mmol) in AcOH(5 mL), Benzaldehyde(42.45 mg, 0.4 mmol) and Cu(OAc)2(18.2 mg, 0.1 mmol) was added. The reaction mixture was stirred at 70C for 30 mins. The mixture was purified by preparative TLC directly to give tert-butyl l-(4-(2-chloro- 6-methoxy-8-phenyl-9H-purin-9-yl)phenyl)cyclobutylcarbamate (25 mg, 25%). LC/MS: (ESI+) e/z: 506 [M+l]+ 508 [M+3]+;
[00334] Step E: To a solution of tert-butyl l-(4-(2-chloro-6-methoxy-8-phenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate(15 mg, 0.03 mmol) in a mixture of Dioxane(5 mL) and H2O(0.5 mL), Pd(PPh3)4(3 mg,0.0026 mmol), phenylboronic acid (54.87 mg,0.45 mmol) and Cs2C03(29.3 mg, 0.09 mmol) was added. The reaction mixture was stirred under nitrogen at 80 °C for lh. The mixture purified by preparative TLC directly to give tert-butyl l-(4-(6-methoxy-2,8-diphenyl-9H- purin-9-yl)phenyl)cyclobutylcarbamate (10 mg, 61% yield ). LC/MS: (ESI+) e/z: 548 [M+l]+; 550 [M+3]+.
[00335] Step F: A solution of tert-butyl l-(4-(6-methoxy-2,8-diphenyl-9H-purin-9- yl)phenyl)cyclobutylcarbamate (10 mg) in HCl (5 mL, conc.).was stirred at 80 °C for 30 mins. The mixture was purified by preparative TLC to give 9-(4-(l-aminocyclobutyl)phenyl)-2,8-diphenyl- lH-purin-6(9H)-one (3 mg). 'HNMR (MeOD, 400 MHz): <57.97-7.95(d, 2H), 7.70- 7.68 (d, 2H), 7.6-7.46 (m, 8H), 7.39-7.37(m, 2H), 2.83(m, 2H,), 2.68-2.64(m, 2H), 2.35(m, 1H), 1.95(m, 1H). LC/MS: (ESI+) e/z: 434 [M+l]+, 436 [M+3]+.
Example 71
1 -(4-(5 -methyl-2-phenyl-3H-imidazo [4,5 -blpyridin-3 -vDphenyDcyclobutanamine
[00336] Step A: To a solution of 2-chloro-6-methyl-3-nitropyridine (600 mg, 3.5 mmol) and tert-butyl l-(4-aminophenyl)-cyclobutylcarbamate in THF (20 mL) was added DIPEA (900 mg, 7 mmol). The reaction mixture was stirred at 70°C for 5 h. The reaction mixture was concentrated
and the residue was purified by flushing chromatography on silica gel (eluted with Hexane:Ehyl acetate = 20:1) to give tert-butyl l-(4-(6-methyl-3-nitropyridin-2- ylamino)phenyl)cyclobutylcarbamate (550 mg, 39% yield). LC/MS (ESI+): 399.2 [M+l]+, 401.2 [M+3]+.
[00337] Step B: The tert-butyl l-(4-(6-methyl-3-nitropyridin-2-ylamino)phenyl)- cyclobutylcarbamate (550 mg, 1.38 mmol) was dissolved in THF (50 mL) and Raney- Ni (1.00 g) was added. The reaction mixture was stirred at ambient temperature under an atmosphere of hydrogen (balloon) for 1 h. The suspension was filtered through a pad of Celite and the pad was withed with THF (20 mL). The combined filtrates were concentrated to give tert-butyl l-(4-(3- amino-6-methylpyridin-2-ylamino)phenyl)cyclobutylcarbamate (480 mg, 94.7% yield). LC/MS (ESI+): 368.9 [M+l]+, 370.9 [M+3]+.
[00338] Step C: The tert-butyl l-(4-(3-amino-6-methylpyridin-2-ylamino)phenyl)- cyclobutylcarbamate (300 mg, 0.81 mmol) was dissolved in acetic acid (50 mL), and benzaldehyde (129 mg, 1.22 mmol) and Cu(OAc)2 (76 mg, 0.42 mmol) were added separately. The reaction mixture was stirred at 100 °C for 2 h. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (50 mL). The organic layer was washed by saturated. NaHC03 solution, followed by brine, dried over sodium sulfate, filtered and concentrated to give tert-butyl l-(4-(5-methyl-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutyl-carbamate (238 mg, 64.5% yield). LC/MS (ESI+): 455.2 [M+l]+, 457.2 [M+3]+
[00339] Step D: The tert-butyl l-(4-(5-methyl-2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutylcarbamate (238 mg, 0.52 mmol) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0°C and the solution of HC1 in ethyl acetate (10 mL, 4M) was slowly added. The resulting solution was stirred at 20 °C for 20 min. The mixture was poured into 30 mL of aqueous NaHC03, the separated organic layer was washed with brine, dried over Na2S04 and concentrated to give l-(4-(5-methyl-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)- cyclobutanamine (71 mg, 38.4% yield).'H NMR (DMSO-d6, 400 MHz), S ppm: 8.08 (d, 1H, J=8.0 Hz), 7.79 (d, 2H, J=8.4 Hz), 7.52 (d, 2H, J=8.4 HZ), 7.46 (d, 2H, J=8.4 Hz), 7.48-7.37 (m, 3H), 7.26-7.25 (d, 1H, J=8.0 Hz), 2.60-2.54 (m, 4H), 2.51 (s, 3H), 2.23-2.16 (m, 1H), 1.92-1.87 (m, 1H). MS (ESI+): 355.1 [M+lf, 357.1 [M+l]+.
Example 72
l-(4-(6-bromo-2-phenyl-3H-imidazof4.5-b1pyridin-3-yl')phenyl)cvclobutanamine
[00340] Step A: A mixture of 5-bromo-2-chloro-3-nitropyridine (1 g, 4 mmol) and tert- butyl l-(4-aminophenyl)cyclobutylcarbamate (1.04 g, 4 mmol) in dioxane (30 mL) was added TEA (2ml) in one portion. Then the reaction mixture was warmed to 70 °C and stirred overnight. The reaction mixture was cooled to room temperature and concentrated to dryness to give the residue, which was further purified by flash chromatography to afford tert-butyl l-(4-(5-bromo-3- nitropyridin-2-ylamino)phenyl) cyclobutylcarbamate. (1 g, 54% yield).LC/MS: (ESI+):463 [M+1J+, 465 [M+l]+.
[00341] Step B: tert-butyl l-(4-(5-bromo-3-nitropyridin-2-ylamino)phenyl)cycIobutyl carbamate (1 g, 2 mmol) was dissolved in THF (50mL) and treated with the catalytical amount of Raney Ni(0.2 g), then the mixture was stirred under hydrogen (balloon) at room temperature for 30 minutes. The reaction mixture was filtrated and the filtrate was concentrated to afford tert-butyl 1- (4-(3-amino-5-bromopyridin-2-ylamino)phenyl)cyclobutyl carb -amate, which was used for the next step without further purification (0.6 g, 64% yield). LC/MS (ESf ): 433 [M+l]+, 435 [M+3]+,
[00342] Step C: benzaldehyde (0. 6 g, 1.3 mmol) and Cu(OAc)2 (158 mg, 0.13 mmol) was added to a solution of tert-butyl l-(4-(3-amino-5-bromopyridin-2- ylamino)phenyl)cyclobutylcarbamate (1 g, 2.87 mmol) in glacial acetic acid (20 mL). The reaction mixture was stirred at 70 °C for lh. then the mixture was diluted with water and basified with saturate sodium bicarbonate and then extracted with ethyl acetate for three times. The combined organic layers were dried over Na2S04 and concentrated to give the crude product, which was purified by flash chromatography to afford tert-butyl l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (0.4 g, 59 % yield). LC/MS (ESI+): 519 [M+l]+'521 [M+3]+.
[00343] Step D: To a solution of tert-butyl l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (0.3 g, 0.5 mmol) in EtOAc was added the solution of HC1 in EtOAc (4 mol/L , 10 ml) at 0 °C dropwise. The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated under reduced pressure to give l-(4-(6- bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl) cyclobutanamine as HC1 salt was obtained
(120 mg, 49.5% yield).'HNMR (DMSO-i/6, 400 MHz): δ 9.04 (s, 3H), 8.51 (s, 1H), 8.43 (s, 1H), 7.73-7.57 (m, 2H), 7.55-7.37 (m, 7H), 2.64-2.59 (m, 4H), 2.20 (m, 1H), 1.80 (m, 1H). LC/MS (ESI+): 419 [M+l]+'421 [M+3]+.
Example 73
l-(4-(6-(2-fluorophenyl)-2-phenyl-3H-imidazo[4,5-blpyridin-3-yl)phenyl)cyclobutanamine
[00344] Step A: A mixture of l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutanamine (0.8 mmol), 2-fluorophenyl boronic acid (0.96 mmol), Cs2C03(521 mg, 2.4 mmol) and Pd(dppf)Cl2 (116 mg, 0.16 mmol) in dioxane and H20 (10 ml, 5:1) was stirred at 80 °C for 2 hours under nitrogen atmosphere. The cooled reaction mixture was poured into water and extracted with EtOAc for three times, then the combined organic layers were washed with water and brine, dried and concentrated to give the residue, which was further purified by flash chromatography to afford l-(4-(6-(2-fluorophenyl)-2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobut anamine (80 mg, 33 % yield .'HNMR (DMSO-</6, 400 MHz): δ 8.46(s, 1H), δ 8.34(s. 1H), 7.66-7.58(m, 5H), 7.47-7.33(m, 8H), 2.44-2.17(m, 4H), 2.14(m, 1H), 1.80(m, 1H). LC/MS (ESf ): 434.2 [M+l]+'436.2 [M+3]+
Exam le 74
l-(4-(2-phenyl-3H-imidazor4,5-b1pyridin-3-yl)phenyl)cyclobutanamine
[00345] Step A: A solution of tert-butyl l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (80 mg) in MeOH (10 mL) was stirred under hydrogen (balloon) atmosphere in presence of Raney Ni (100 mg) at room temperature for 10 min. The reaction mixture was filtered through celite pad and the filtrate was concentrated to give 60 mg of
crude desired compound tert-butyl l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutylcarbamate, which was used directly without further purification. LC/MS (ESI+): 441.3 [M+l]+, 443.3 [M+3]+
[00346] Step B: To a solution of t tert-butyl l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3- yl)phenyI)cyclobutylcarbamate (60 mg crude) in EtOAc was added the solution of HCl in EtOAc (4 mol/L , 10 ml) at 0 °C dropwise. The reaction mixture was stirred at room temperature for 1 h. The mixture was concentrated under reduced pressure, the crude residue was purified by preparative HPLC to give l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine (20 mg) 1HNMR (CD30D-^, 400 MHz): 68.35(s, 1H), 8.21(s, 1H), 7.66(s, 1H), 7.60-7.30(m, 8H), 2.80(m, 2H), 2.64(m, 2H), 2.20(m, 1H), 1.99(m, 1H). LC/MS (ESI+): 341.3 [M+l]+,343.3 [M+3]+.
Example 75
1 -[4-(2-Phenyl-imidazo 4.5-b1pyrazin- 1 -yl Vphenyl -cvclobutylamine
[00347] Step A: 2,3-Dichloro-pyrazine (14.8 g, 0.1 mol) was added in a 100 mL autoclave which contained 50 mL of the solution of N¾ aqueous with stirring, and then the reaction mixture was warmed to 100°C and stirred overnight, Cooled to room temperature. Filtrated, the solid was washed with water and further with dichloromethane to afford a solid, which dried on vacuum to afford 3-Chloro-pyrazin-2-ylamine. (10 g, 80% yield). LC/MS (ESI+): 130[M+1]+, 132[M+3]+
[00348] Step B: The solution of dimethyl sulfoxide (4 g, 0.05 mmol) in 30mL of dry dichloromethane was cooled to -78°C, then (13 g, 0.046 mmol) trifluoromethanesulfonic anhydride was added dropwise under nitrogen with stirring. After addition the solution was stirred for 2 hours, then was added a solution of 3-Chloro-pyrazin-2-ylamine (5.0 g, 0.039 mmol) in 30mL of dichloromethane and dimethyl sulfoxide (5:1), the resulting reaction mixture was continued stirred at -78°C for 3 hours, quenched by saturate NH4CI solution then diluted with dichloromethane (200 mL) and the organic phase was separated, the water phase was further extracted with dichloromethane twice. The combined organic phase was washed with brine. The organic phase was concentrated to afford S,S-dimethyl-N-(2-pyrazinyl)sulfilimine, which was use directly for next step. (7.5 g, 80% yield) LC/MS (ESI+): 190[M+1]+, 192[M+3]+.
[00349] Step C: To m-chloroperbenzoic acid (8.0g, 0.46 mmol 85%) in dichloromethane
(100 mL)(cooled to -5°C) was added a solution of S,S-dimethyl-N-(2-pyrazinyl)sulfilimine (5.36 g, 0.028 mmol) in 30ml dichloromethane dorpwise at such a rate for 1 hour with the temperature not exceed 0°C, the reaction mixture was stirred for another 40min at 0°C, The solution of dimethyl sulfide in dichloromethane was added at 0°C. Filltered quickly to afford a orange clean solution .The solution was cooled to -78°C, then 03 was bubbled into the mixture until the orange solution was turned to be colorless. The resulting suspension was quenched with sodium dicarbonate solution and then extracted with dichloromethane for three times. The combined organic phase was concentrated to afford the crude product, which was further purified by flash chromatography to afford 2-Chloro-3-nitro-pyrazine. (0.5 g, yield: 7%).1HNMR (DMS0-< 400 MHz): δ 8.60 (s, 1H), δ 8.83 (s, 1H).
[00350] Step D: A mixture of 2-Chloro-3-nitro-pyrazine (0.5 g, 3 mmol) and [l-(4-Amino- phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (0.98 g, 3.6 mmol) in dioxane (20mL) was added Et3N (2ml) in one portion. Then the reaction mixture was warmed to 70°C for overnight. Cooled to room temperature and Concentrated to dryness to afford the residue, which was further purified by flash chromatography to afford { l-[4-(3-Nitro-pyrazin-2-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. (0.57 g, 50% yield). LC/MS (ESI+): 386[M+1]+, 388[M+3]+
[00351] Step E: { l-[4-(3-Nitro-pyrazin-2-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert- butyl ester (0.57 g, 1.5 mmol) was dissolved in THF (30mL) and treated with the catalyst amount of Raney Ni (0.2g ), then the reaction mixture was stirred under hydrogen (balloon) for 30 minutes at room temperature. Filtrated and the filtrate was concentrated to give { l-[4-(3-Amino-pyrazin-2- ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester, which was used for next step without further purification (0.26g, 51% yield). LC/MS (ESI+): 356[M+1]+, 358[M+3]+
[00352] Step F: { l-[4-(3-Amino-pyrazin-2-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (0.26 g, 0.75 mmol) and Cu(OAc)2 (158 mg, 0.13 mmol)was added to a solution of Benzaldehyde (1 g, 2.87 mmol) in glacial acetic acid (20 mL), the reaction mixture was stirred at 70°C for lh. the reaction mixture was diluted with water and bacified with saturates sodium bicarbonate. Extracted with ethyl acetate for three times, the combined organic layer was concentrated to give the crude product ,which was purified by flash chromatography to afford the{l-[4-(2-Phenyl-imidazo[4,5-b]pyrazin-l-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (92 mg, 80 % yield). LC/MS (ESI+): 442[M+1]+, 444[M+4]+
[00353] Step G: To a solution { l-[4-(2-Phenyl-imidazo[4,5-b]pyrazin-l-yl)-phenyl]- cyclobutylj-carbamic acid tert-butyl ester (90mg, 0.2mmol)in ethyl acetate (10 mL) was added a solution of HC1 in ethyl acetate (4mol/L HC1, 10ml) at 0°C dropwise , the reaction mixture was
stirred at room temperature for 1 h. the reaction mixture was concentrated under reduced pressure to afford l-[4-(2-Phenyl-imidazo[4,5-b]pyrazin-l-yl)-phenyl]-cyclobutylamine as HC1 salt. (25 mg, yield: 49.5%). 1HNMR (CD30D-< 400 MHz): δ 8.59(m, 1H), δ 8.40(m, 1H), 7.65-7.35(m, 9H), 2.76(m, 2H), 2.58(m, 2H), 2.20(m, 1H), 1.91(m, 1H). LC/MS (ESI+): 342[M+l , 344[M+3]+
Exam le 76
l-(4-(5-phenyl-2-(pyridin-2-yl)-3H-imidazor4,5-blpyridin-3-yl)phenyl)cvclobutanamine
[00354] Step A: To a solution of 2,6-dichloro-3-nitropyridine (0.20 g, 1.036 mmol) in dioxane (20 mL) were added tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (0.27 g, 1.036 mmol), DIPEA (0.134 g, 1.036 mmol). The reaction mixture was stirred at 70 °C overnight. Then it was concentrated to remove the solvent, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated to afford tert-butyl l-(4-(6-chloro-3-nitropyridin-2-ylamino)phenyl)cyclobutylcarbamate (0.41 g, 94%yiedl ), which was pure enough to be used in next step without further purification. MS (ESI+) e/z: 419 [M+l]+, 421 [M+3]+
[00355] Step B: To a solution of tert-butyl l-(4-(6-chloro-3-nitropyridin-2- ylamino)phenyl)cyclobutylcarbamate (0.41 g, 0.98 mmol) in a mixed solution (dioxane:H20=15:3 mL) were added phenylboronic acid (0.17 g, 1.37 mmol), Pd(dppf)Cl2 (36 mg, 0.049 mmol), and Cs2C03 (0.64 g, 1.96 mmol). The reaction mixture was stirred at 100 C for 3 h.. Then it was concentrated to remove the solvent, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product was purified by column separation (EtOAc:hexane=3:l) to afford tert-butyl l-(4-(3- nitro-6-phenylpyridin-2-ylamino)phenyl)cyclobutylcarbamate (0.40 g, 89% yield). MS (ESI+) e/z: 461 [M+l]+, 463 [M+3]+
[00356] Step C: To a solution of tert-butyl l-(4-(3-nitro-6-phenylpyridin-2- ylamino)phenyl)cyclobutylcarbamate (0.40 g, 0.87 mmol) in THF (20 mL) was added Raney-Ni (0.20 g). The reaction mixture was stirred at ambient temperature under the atmosphere of hydrogen (balloon) for 1 h. The catalyst was removed by filtration, and the filtrate was
concentrated. The residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated to afford tert-butyl l-(4-(3- amino-6-phenylpyridin-2-ylamino)phenyl)cyclobutylcarbamate (0.35 g, 93% yield), which was used in next step without further pufiication. MS (ESI+) e/z: 431 [M+l]+, 432 [M+3]+
[00357] Step D: To a solution of tert-butyl l-(4-(3-amino-6-phenylpyridin-2- ylamino)phenyl)cyclobutylcarbamate (0.40 g, 0.929 mmol) in AcOH (20 mL) were added picolinaldehyde (0.149 g, 1.394 mmol), and Cu(OAc)2 (0.118 g, 0.650 mmol). The reaction mixture was stirred at 100 C for 1 h. It was concentrated to remove the solvent and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated to afford fert-butyl l-(4-(5-phenyl-2-(pyridin-2-yl)-3H- imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutylcarbamate (-0.40 g), which was used in next step without further purification. MS (ESI+) e/z: 518 [M+l]+, 520 [M+3]+.
[00358] Step E: To a solution of tert-butyl l-(4-(5-phenyl-2-(pyridin-2-yl)-3H-imidazo[4,5- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (-0.4 g) in ethyl acetate (20 mL) was added the solution of HC1 in ethyl acetate (5 mL) at 0 C. Then it was allowed to reach ambient temperature for 3 h. The reaction mixture was concentrated to remove the solvent. The crude product was purified by preparative HPLC to afford l-(4-(5-phenyl-2-(pyridin-2-yl)-3H-imidazo[4,5-b]pyridin-3- yl)phenyl)cyclobutanamine (90 mg, 23% yield, two steps). 1H NMR (MeOD, 400 MHz), δ ppm: 8.42-8.40 (m, 1H), 8.25 (d, J=8.4 Hz, 1H), 8.10-7.95 (m, 5H), 7.66-7.64 (m, 2H), 7.59-7.56 (m, 2H), 7.47-7.38 (m, 4H), 2.87-2.82 (m, 2H), 2.65-2.57 (m, 2H), 2.31-2.20 (m, 1H), 2.08-1.96 (m, 1H). MS (ESI+) e/z: 418 [M+l]+ 420 [M+3]+
Example 77
1 -(4-(2 -phenyl- 1 H-pyrrolo[3,2-c1pyridin- 1 -yl)phenyl)cyclobutanamine
[00359] Step A: To a suspension of 4-chloropyridine (10 g, 0.067 mol) in THF (150 mL, dry) was dropping LDA (134 ml, 1 M in hexane) while keeping the temperature below -70°C. The mixture was stirred for 30min at was -78°C and then treated over 10 min with a solution of I2 (17 g, 0.067 mol) in dry THF keeping the internal temperature below -65 °C. The reaction mixture was
allowed to warm to r.t. over 2h, followed by stirring at the same temperature for 30min. The reaction mixture was poured into Na2S03 (20%, 100 mL) aqueous solution and extracted with EtOAc (100 mL, three times). The organic layer was washed with NaHC03 aqueous solution (50 mL), saturated NaCl aqueous solution (50 mL) and dried. The solvent was evaporated and purified by washing with petroleum ether (20mL) to yield the desired product 4-chloro-3-iodopyridine (6.2g, 38% yield). 'HNMR (400MHZ, MeOD) δ 8.92(s, 1H), 8.43-8.41 (d, 1H, J=8), 7.62-7.60 (d, 1H, J=8).
[00360] Step B: To a solution of 4-chloro-3-iodopyridine (239 mg, 1 mmol), 1- ethynylbenzene (102 mg, 1 mmol) and Pd(PPh3)2Cl2 (70 mg, 0.1 mmol) in TEA (15 mL) was added Cul (19 mg, 0.1 mmol). The mixture degassed and charged with N2 for three times. The mixture was stirred at reflux for 60min.The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=100/l~5/l), to give the title product 4-chloro-3-(2-phenylethynyl)pyridine (200 mg, 94% yield). LC/MS (ESI+): 214 [M+l]+. 216 [M+3]+
[00361] Step C: To a solution of 4-chloro-3-(2-phenylethynyl)pyridine (160 mg, 0.748 mmol), Pd2(dba)3 (68 mg, 0.0748 mmol), X-phos (172 mg, 0.299 mmol) and Cs2C03 (487 mg, 1.496 mmol) in Dioxane (8mL) was added tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (196 mg, 0.748 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate =100/1-1/1) to yield the desired product tert-butyl l-(4-(2-phenyl-lH- pyrrolo[3,2-c]pyridin-l-yl)phenyl)cyclobutylcarbamate (44 mg, 13% yield) as a yellow solid. 'HNMR (400 MHz MeOD) δ 8.87(s, 1H), 8.15(s, 1H), 7.55~7.53(m, 2H), 7.26-7.21 (m, 8H), 6.95 (s, 1H), 2.59-2.44 (m, 4H), 2.15-2.00 (m, 1H), 1.95-1.82 (m, 1H), 1.36 (s, 9H).
[00362] Step D: To a solution of HC1 in EtOAc (20 mL, 2 M) was added tert-butyl 1 - ( 4- (
2 -phenyl- lH-pyrrolo [ 3,2-c ] pyridin-l-yl ) phenyl ) cyclobutylcarbamate (44 mg, 0.10 mmol ). The mixture was stirred at r.t. for 60 min. The reaction mixture was filtered by filter paper to give l-(4-(2-phenyl-lH-pyrrolo[3,2-c]pyridin-l-yl)phenyl) cyclobutanamine (23 mg, 56% ).!HNMR (400MHz MeOD) δ 9.25(s, 1H), 8.41~8.39(d, 1H, J=8), 7.72~7.69(m, 3H), 7.54~7.52(m, 2H), 7.38~7.33(m, 6H), 2.85~2.75(m, 2H), 2.65~2.55(m, 2H), 2.30~2.20(m, 1H), 2.05-1.95(m, 1H). LC/MS (ESI+): 340.1 [M+l]+. 342.1 [M+3]+
1 -(4-( 1 -aminocvclobutyl)phenyl)-2-phenyl- 1 H-pyrrolo [3 ,2-c1pyridin-4(5IT)-one
[00363] Step A: t-BuLi (280 mL, 364 mmol) was added dropwise to a solution of the tert- butyl 2-chloropyridin-4-ylcarbamate (20 g, 87.7 mmol) in THF (500 mL) under N2. The mixture is stirred at -78°C for 1 h. Then a solution of I2 in THF (lOOmL) was added drop wise to the mixture. The mixture was stirred at -78 °C for 1.5 h.. The mixture reaction is quenched with saturated NH4CI (lOOmL), the aqueous phase was extracted with EA (2*50mL). The combined organic layers were concentrated and purified by column separation (eluting with petroleum ether/ethyl acetate =1 :20) to give tert-butyl 2-chloro-3-iodopyridin-4-ylcarbamate. (12 g 37.5% yield) MS (ESI) m/z : 354.9 [M+l]+ 356.9 [M+3]+
[00364] Step B: A solution of the tert-butyl 2-chloro-3-iodopyridin-4-ylcarbamate (3 g, 0.85 mol) and NaOMe (4 g, 7.4 mmol) in MeOH (80 mL) was stirred at 100 °C for overnight in a 100 mL of autoclave.. After the solvent was removed by concentration, the residue was partitioned between ethyl acetate (100 mL) and water (50 mL). The separated organic layer was concentrated and purified by column separation (eluted by EA:PE=1 :20), which gave 1.2 g of desired product 3- iodo-2-methoxypyridin-4-amine (Yield: 65.6%). MS (ESI) m/z : 250.9 [M+l]+. 252.9 [M+3]+.
[00365] Step C: To a solution of 3-iodo-2-methoxypyridin-4-amine (5 g, 22.6 mmol), 1- ethynylbenzene (2.3 g, 22.6 mmol), and Pd(PPh3)2Cl2(1.58 g, 2.26 mmol) in TEA (40ml) was added Cul (430 mg, 2.26 mmol). The mixture degassed and charged with N for three times. The mixture was stirred at reflux for 60 min. The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=100/l~5/l), to give the title product 2-methoxy-3-(2-phenylethynyl) pyridine- amine (3g, 58% yield). 'HNMR (400MHz, CDC13) 6 7.78~7.76(d, 1H, J=8), 7.76-7.50 (m, 2H), 7.40-7.30 (m, 3H), 6.29-6.27 (d, 1H, J=8), 4.76 (s, 2H), 3.98(s, 3H) .
[00366] Step D: To a solution of 2-methoxy-3-(2-phenylethynyl)pyridin-4-amine (1.8 g, 8.04 mmol) in CH3CN (10 mL) was added toluene sulfonic acid (3.05g, 24mmol), the mixture was stirred at OoC for 10 min. A solution of NaN02 (830 mg, 16 mmol) and Nal (1.8 g, 16 mmol) in
water (2 mL) was dropwise in the pre-solution, and stirred at 0°C for another lh. The reaction mixture was quenched with Na2S03 solution, and concentrated under reduced pressure to get crude product, which was purified by silica gel chromatography to give the title product 4-iodo-2- methoxy-3-(2-phenylethynyl)pyridine (1.1 g 42%). 'HNMR (400MHZ, MeOD) δ 8.15~7.95(d, 1H, J=8), 7.59~7.57(m, 2H), 7.40-7.30 (m, 3H), 6.59-6.57 (d, 1H, J=8), 3.91(s, 3H) .
[00367] Step E: To a solution of 4-iodo-2-methoxy-3-(2-phenylethynyl)pyridine (335 g, 1 mmol), Pd2(dba)3 (90 mg, 0.1 mmol), X-phos (190 mg, 0.4 mmol) and Cs2C03 (652 mg, 2 mmol) in Dioxane (lOmL) was added 4-(l-aminocyclobutyl) benzenamine (262mg, lmmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60 min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=50/l~l/l) to yield the desired product tert-butyl l-(4-(2-methoxy-3-(2-phenylethynyl) pyridin-4-ylamino)phenyl) cyclobutylcarbamate (350mg, 74.6%). 'HNMR (400MHz, MeOD) δ 7.71~7.69(d, 1H, J=8), 7.55~7.49(m, 4H), 7.36-7.23 (m, 5H), 6.59-6.57 (d, 1H, J=8), 3.96(s, 3H), 2.56~2.48(m, 4H), 2.11~2.04(m, 1H), 1.89~1.80(m, 1H), 1.36(s, 9H) .
[00368] Step F: To a solution of Pd(t-Bu3P)2 (33 mg, 0.064 mmol), KF(150 mg, 1.92 mmol) and Cs2C03 (417 mg, 1.28 mmol) in Dioxane (8 mL) was added tert-butyl l-(4-(2-methoxy-3-(2- phenylethynyl) pyridin-4-ylamino)phenyl) cyclobutylcarbamate (300 mg, 0.64 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=50/l~l/l) to yield the desired product tert-butyl l-(4-(4-methoxy-2-phenyl-lH-pyrrolo [3,2-c]pyridin-l-yl)phenyl) cyclobutylcarbamate (300mg, 85%). 'HNMR (400MHz, MeOD) δ 7.73~7.71(d, 1H, J=8), 7.53-7.5 l(m, 2H), 7.25-7.18 (m, 7H), 6.83(s, 2H), 4.06(s, 3H), 2.66~2.48(m, 4H), 2.18~2.04(m, 1H), 1.89~1.80(m, 1H), 1.36(s, 9H) .
[00369] Step G: Tert-butyl l-(4-(4-methoxy-2-phenyl-lH-pyrrolo [3,2-c]pyridin-l- yl)phenyl) cyclobutylcarbamate (150 mg) and PyHCl C 2 g) were added into a microwave vessel, which was stirred at 120 °C for 20min by microwave irradiation. The reaction mixture was purified by preparative HPLC, to yield the desired product l-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-lH- pyrrolo[3,2-c]pyridin-4-ol (80 mg, 71%). !HNMR (400MHz, DMSO) δ 11.05(s,lH), 7.58~7.50(m, 2H), 7.38~7.20(m, 7H), 7.07-7.05 (d, 1H, J=8), 6.85(s, 1H), 6.05~6.03(d, 1H, J=8), 2.50~2.38(m, 2H), 2.30~2.14(m, 2H), 2.18~2.04(m, 1H), 1.69~1.60(m, 1H). MS (ESI) m/z : 356 [M+l]+. 358
[M+3]+
Example 79
l-(4-(l-aminocyclobutvnphenyl)-2-phenyl-lH-pyrrolo[3.2-blpyridin-5(4H)-one
[00370] Step A: To a solution of 2-bromo-6-methoxypyridine (3 g, 16 mmol) in DMF
(20mL) was added NCS (2.3 g, 17.6 mmol). The mixture was stirred at r.t. for 4 days. The reaction mixture was poured into 150ml water, followed by extraction with ethyl ether. The organic layer was washed with water, NaHC03 aqueous solution, NaCl saturated solution, dried over MgS04 and the solvent was distilled off under reduced pressure to give crude product 2-bromo-3-chloro-6- methoxypyridine (2 g, 57%), which was used to next step without further purification.. 1HNMR (400MHz, CDC13) δ 7.56~7.54(d, 1H, J=8), 6.67-6.65 (d, 1H, J=8), 3.92(s, 3H) .
[00371] Step B: To a solution of 2-bromo-3-chloro-6-methoxypyridine (1.5 g, 6.78 mmol),
1-ethynylbenzene (692 mg, 6.78 mmol), and Pd(PPh3)2Cl2(475 mg, 0.678 mmol) in TEA (40 mL) was added Cul (129 mg, 0.678 mmol). The mixture degassed and charged with N2 for three times. The mixture was stirred at reflux for 60 min. The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=100/l~5/l), to give the title product 3-chloro-6-methoxy-2-(2-phenylethynyl ) pyridine (1.5 g, 91%). MS (ESI) m/z : 243.8 [M+l]+. 245.8 [M+3]+
[00372] Step C: To a solution of 3-chloro-6-methoxy-2-(2-phenylethynyl)pyridine (1.94 g,
8 mmol), Pd2(dba)3 (732 mg, 0.8 mmol), X-phos (1.5 g, 3.2 mmol) and Cs2C03 (5.2 g, 16 mmol) in Dioxane (10 mL) was added tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (2.1 g, 8 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60 min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=50/l~l/l) to yield the desired product tert-butyl l-(4-(6-methoxy-2-(2-phenylethynyl) pyridin-3-ylamino) phenyl) cyclobutylcarbamate (250mg, 7%). MS (ESI) m/z : 470.2 [M+l]+ ,472.2[M+3]+.
[00373] Step D: To a solution of Pd(t-Bu3P)2 (25 mg, 0.05 mmol), KF(117 mg, 1.5 mmol) and Cs2C03 (326 mg, 1 mmol) in Dioxane (8 mL) was added tert-butyl l-(4-(6-methoxy-2-(2- phenylethynyl) pyridin-3-ylamino) phenyl) cyclobutylcarbamate (234 mg, 0.5 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60 min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=50/l~l/l) to yield the desired product tert-butyl l-(4-(5-methoxy-2 -phenyl- lH-pyrrolo[3,2-b] pyridin-l-yl) phenyl) cyclobutylcarbamate (200mg, 36%). MS (ESI) m/z : 470.2 [M+l]+. 472.2 [M+3]+
[00374] Step E: Tert-butyl l-(4-(5-methoxy-2-phenyl-lH-pyrrolo[3,2-b] pyridin-l-yl) phenyl) cyclobutylcarbamate (200 mg) and PyHCl(l g) were added into a microwave vessel, which was stirred at 120 °C for 20 min by microwave irradiation. The reaction mixture was purified by preparative HPLC, to yield the desired product l-(4-(l-aminocyclobutyl)phenyl) -2 -phenyl- 1H- pyrrolo[3,2-b]pyridin-5-ol (60 mg, 40%). 1HNMR (400MHz, MeOD) δ 7.62~7.60(m, 2H), 7.60~7.58(d, 1H, J=8), 7.38-7.36 (m, 2H), 7.26~7.24(m, 5H), 6.51(s,lH) 6.31~6.29(d, 1H, J=8), 2.50~2.68(m, 2H), 2.60~2.54(m, 2H), 2.28-2.14(m, 1H), 1.99~2.00(m, 1H). MS (ESI) m/z : 356 [M+l]+. 358 [M+3]+
Example 80
1 -(4-(2-phenyl- 1 H-pyrrolo[2.3-b1pyridin- 1 -vDphenyDcvclobutanamine
[00375] Step A: To a solution of 2-chloro-3-iodopyridine (239 mg, 1 mmol), ethynyl- benzene (102 mg, 1 mmol), and Pd(PPh3)2Cl2(70 mg, 0.1 mmol) in TEA (15 mLl) was added Cul (19 mg, 0.1 mmol). The mixture degassed and charged with N2 for three times. The mixture was stirred at reflux for 60min. The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=100/l~5/l), to give the title product 2-chloro-3-(2-phenylethynyl)pyridine (130mg, 61%). 1HNMR: CDC13 400MHz δ 8.31~8.30(d, 1H, J=8), 7.81~7.82(d, 1H, J=8), 7.57~7.55(m, 2H), 7.37~7.35(m, 3H), 7.23~7.21(m, 1H)
[00376] Step B: To a solution of 2-chloro-3-(2-phenylethynyl)pyridine (426 mg, 2 mmol),
Pd2(dba)3 (90 mg, 0.1 mmol), X-phos (190 mg, 0.4 mmol) and Cs2C03 (978 mg, 3 mmol) in Dioxane (8mL) was added tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (524 mg, 2 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=l 00/1 -1/1) to yield the desired product tert-butyl l-(4-(3-(2-phenylethynyl)pyridin-2- ylamino)phenyl)cyclobutylcarbamate (220 mg, 25% yield^HNMR: MeOD 400MHz δ 8.08(s, lH), 7.71~7.69(d, 1H, J=8), 7.57~7.53(m, 4H), 7.38-7.36 (m, 5H), 6.75 (s, lH), 2.80-2.76 (m, 4H), 2.15-2.00 (m, 1H), 1.95-1.82 (m, 1H), 1.36 (s, 9H).
[00377] Step C: To a solution of Pd(t-Bu3P)2(25 mg, 0.05 mmol), KF(117 mg, 1.5 mmol) and Cs2C03 (326 mg, 1 mmol) in Dioxane (8mL) was added tert-butyl l-(4-(3-(2- phenylethynyl)pyridin-2-ylamino)phenyl)cyclobutylcarbamate (220 mg, 0.5 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60 min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate= 100/1 -1/1) to yield the desired product tert-butyl l-(4-(2-phenyl-lH-pyrrolo[2,3-b]pyridin-l- yl)phenyl)cyclobutylcarbamate (80mg, 36%). MS (ESI) m/z: 440 [M+l]+. 442 [M+3]+
[00378] Step D: To a solution of HC1 in EtOAc (30 mL, 2 M) was added tert-butyl l-(4-(2- phenyl-lH-pyrrolo[2,3-b]pyridin-l-yl)phenyl) cyclobutylcarbamate (80 mg, 0.18 mmol). The mixture was stirred at r.t. for 60 min. The reaction mixture was filtered to give l-(4-(2-phenyl-lH- pyrrolo[2,3-b]pyridin-l-yl)phenyl)cyclobutanamine (50mg). 'HNMR: MeOD 400MHz δ 8.63~8.61(d, 1H, J=8), 8.32~8.30(d,lH,J=8), 7.71~7.69(m,2H), 7.58~7.56(m,3H), 7.35~7.29(m,5H), 7.11(s,lH), 2.80~2.765(m,2H), 2.68~2.64(m,2H), 2.240~2.20(m,lH), 2.00-1.96(m,lH). MS (ESI) m/z: 340 [M+lf. 342 [M+3]+
Example 81
[00379] Step A: 6-Chloro-3-iodo-pyridin-2-ylamine (2.54g, 0.01 mol) and Ethynyl-benzene
(1.53 g, 0.015 mol) was added in a 100 mL of flask which contained 30mL of Et3N with stirring, and then the catalyst of Cul (0.38 g, 0.002 mol) and Pd(PPh3)2C12 (2 g, 0.002 mol) was added into the mixture. The reaction mixture was warmed to 80 °C for 30 minutes. When TLC indicated 6- chloro-3-iodo-pyridin-2-ylamine was consumed, the reaction mixture was cooled to room temperature. After filtrated, the filtrate was concentrated to give the crude product, which was purified by flash chromatography to afford 6-chloro-3-phenylethynyl-pyridin-2-ylamine (2 g, 71% yield:). LC/MS (ESI+): 229 [M+l]+, 231 [M+3]+
[00380] Step B: A mixture of 6-chloro-3-phenylethynyl-pyridin-2-ylamine (1 g, 4 mmol), 2- flurobenzene boronic (0.7 g, 4.8 mmol), Cs2C03 (3.9 g, 12 mmol) and Pd(dppf)C12 (0.3 g, 0.4 mmol) in the mixture of dioxane and water(10 ml, 5:1) was degassed and charged with nitrogen three times. The reaction mixture was stirred at 80 oC under nitrogen atmosphere for 2h. The cooled solution was poured into water and extracted with EtOAc. The organic layer was washed with water and brine, dried over Na2S04 and concentrated to afford the crude product, which was further purified by flash chromatography to afford 6-(2-fluoro-phenyl)-3-phenylethynyl-pyridin-2- ylamine (1 g, 65 % yield).LC/MS (ESI+): 289 [M+l]+ ,291 [M+3]+
[00381] Step C: To a solution of tert-butyl nitrite (0.6 g, 6 mmol) in 20 mL of CH3CN was added CuBr2 (1.3 g, 6 mmol) at 0 oC. Then the mixture was stirred at this temperature for one hour, which further treated with the solution of 6-(2-fluorophenyI)-3-(phenylethynyl)pyridin-2- amine(lg, 3mmol) in CH3CN. After 6-(2-Fluoro-phenyl)-3-phenylethynyl-pyridin-2-ylamine was consumed, the mixture was treated with 6mol/L HC1 and stirred for 15 minutes. Then the mixture was diluted with water and extracted with ethyl acetate for three times. The organic phase was washed with water and brine, dried over Na2S04 and concentrated to afford the crude product, which was further purified by flash chromatography to afford 2-bromo-6-(2-fluoro-phenyl)-3- phenylethynyl-pyridine ( 0.5 g, 50 % yield) LC/MS (ESI+): 352 [M+l]+ ,354 [M+3]+
[00382] Step D: A mixture of 2-bromo-6-(2-fluoro-phenyl)-3-phenylethynyl-pyridine (0.5 g,
1.5 mmol), [l-(4-Amino-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (0.4 g, 1.5 mmol), Cs2C03 (0.98 g, 3 mmol) ,X-phos(0.14 g, 0.03 mmol) and Pd2(dba)3 (0.29 g, 0.03 mmol) in dioxane (10 ml) was degassed and then charged with nitrogen. The reaction mixture was stirred at 120 oC in microwave for lh. The cooled reaction mixture was poured into water and extracted with ethyl acetate, then the organic layer was washed with water, dried over Na2S04 and concentrated to afford the crude product, which was further purified by flash chromatography to afford (l-{4-[6- (2-Fluoro-phenyl)-3-phenylethynyl-pyridin-2-ylamino]-phenyl}-cyclobutyl)-carbamic acid tert-
butyl ester (0. 3g, 37 % yield). LC/MS: (ESI+): 534 [M+l]+ ,536 [M+3]+
[00383] Step E: A mixture of (l-{4-[6-(2-Fluoro-phenyl)-3-phenylethynyl-pyridin-2- ylamino] -phenyl }-cyclobutyl)-carbamic acid tert-butyl ester (0.3 g, 0.5 mmol), KF (0.09 g, 1.5 mmol), Cs2C03 (0.33 g, 1.5 mmol), and Pd(t-Bu3P)2 (0.15 g, 0.01 mmol) in dioxane (10ml) was stirred at 130oC in microwave for lh. The cooled reaction mixture was poured into water and extracted with ethyl acetate for three times, then the organic layer was washed with water and brine, dried and concentrated to give the residue, which was further purified by flash chromatography to afford (l-{4-[6-(2-Fluoro-phenyl)-2-phenyl-pyrrolo[2,3-b]pyridin-l-yl]- phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (0.3 g, 100 % yield). LC/MS: (ESI+): 534 [M+l]+ ,536 [M+3]+.
[00384] Step F: The solution of (l-{4-[6-(2-Fluoro-phenyl)-2-phenyl-pyrrolo[2,3- b]pyridin-l-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (0.3 g, 0.7 mmol) in EtOAc was added the solution of HC1 in ethyl acetate (4mol/L HC1). The reaction mixture was stirred at room temperature for 1 h. After removal of the solvents under the reduced pressure, the residue was further purified by preparative FfPLC to affordl-{4-[6-(2-Fluoro-phenyl)-2-phenyl-pyrroIo[2,3- b]pyridin-l-yl]-phenyl}-cyclobutylamine. (120 mg, 49.5% yield).lH MR (DMSO-d6, 400 MHz): δ 8.13(d, J =8.0Hz lH), 7.87-7.67(m, 1H), 7.87-7.52(d, J =8.0Hz 1H), 7.51-7.18(m, 12H), 6.86(s, 1H), 2.8(m, 2H), 2.6(m, 2H), 2.3(m, 1H), 2.0(m, 1H). LC/MS: (ESI+): 434.2 [M+l]+ ,436.2 [M+3]+.
Example 82
8-(4-( 1 -aminocvclobutyl)phenyl)-7-phenyl-7H-purin-2-ol
[00385] This compounds can also be made according to the above-described methods.
Example 83
l-(4-(6-phenyl-7H-pyrrolo 2.3-d1pyrimidin-7-yl)phenyl) cvclobutanamine
[00386] Step A: To a solution of 4-chloro-5-iodopyrimidine (1.2 g, 5mmol), 1- ethynylbenzene (510 mg, 5 mmol), and Pd(PPh3)2Cl2 (350 mg, 0.5 mmol) in TEA (15mL) was added Cul (95mg, 0.5mmol). The mixture degassed and charged with N2 for three times. The mixture was stirred at reflux for 60min. The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=100/l~5/l), to give the title product 4-chloro-5-(2-phenylethynyl) pyrimidine (1 g, 93%). 'H MR: CDCI3 400MHz 68.91(s, 1H), 8.80(s, 1H), 7.60~7.57(m, 2H), 7.43~7.25(m, 3H).
[00387] Step B: To a solution of 4-chloro-5-(2-phenylethynyl)pyrimidine (214 mg, 1 mmol), Pd2(dba)3 (45 mg, 0.05 mmol), X-phos (85 mg, 0.2 mmol) and Cs2C03 (439 mg, 1.5 mmol) in Dioxane (8mL) was added tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (262 mg, 1 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60 min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate =100/1-1/1) to yield tert-butyl l-(4-(6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (20 mg, 4.5% yield). 1HNMR: MeOD 400MHz 59 .03(s, 1H), 8.70(s, 1H), 7.54~7.52(m 2H), 7.29~7.25(m, 7H), 6.94(s, 1H), 2.80~2.64(m, 4H), 2.24~2.20(m, 1H), 2.00-1.96(m, 1H), 1.36(s, 9H)
[00388] Step C: To a solution of HC1 in EtOAc (30mL, 2 M) was added tert-butyl l-(4-(6- phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl) cyclobutylcarbamate (20 mg, 0.045 mmol). The mixture was stirred at r.t. for 60 min. The reaction mixture was filtered to give the title product 1- (4-(6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl) cyclobutanamine (3.5 mg). 'HNMR: MeOD 400MHz , δ 9.07(s, 1H), 8.73(s, 1H), 7.62~7.60(m, 2H), 7.46~7.44(m, 2H), 7.37~7.30(m, 5H), 6.98(s, 1H), 2.80~2.76(m, 2H), 2.68~2.64(m, 2H), 2.24~2.20(m,lH), 2.00-1.96(m,lH). MS (ESI) m/z: 341.2 [M+l]+. 343.2 [M+3]+
tert-butyl l-(4-(2-chloro-6-(,pyridin-2-yn-7H-pyrrolo 2.3-dlpyrimidin-7- vDphenvDcvclobutylcarbamate
[00389] Step A: To a solution of 2,4-dichloro-5-iodopyrimidine (5.50 g, 20.00 mmol) in dioxane (100 mL) was slowly added a solution of tert-butyl l-(4-aminophenyl) cyclobutylcarbamate (5.2 g, 20.00 mmol) in dioxane (20 mL) and triethylamine (5 ml). The reaction mixture was stirred at 80 °C overnight. Lc-ms indicated 2,4-dichloro-5-iodopyrimidine was completed consumed. The solvents were removed under reduced pressure to produce a residue, which was dissolved in ethyl acetate (250 mL). The mixture was washed with brine. The combined organic layer was dried over Na2S04 and then filtered. The filtrate was concentrated to give the crude product, which was purified by flash chromatography to afford tert-butyl l-(4-(2-chloro-5- iodopyrimidin-4-ylamino)phenyl)cyclobutylcarbamate. (5.00 g, 50% yield). LC/MS: (ESI+): 501 [M+l]+ , 503 [M+l]+ .
[00390] Step B: tert-butyl l-(4-(2-chloro-5-iodopyrimidin-4-ylamino)phenyl)cyclobutyl carbamate (5.00 g, 0.01 mol), and 2-ethynylpyridine (1.55 g, 0.015 mol) was added in a lOOmL of flask which contained 30 mL of Et3N with stirring, and then the catalyst of Cul(0.38 g, 0.002 mol)and Pd(PPh3)2Cl2(2 g, 0.002 mol) was added into the mixture. The reaction mixture was warmed to 80 °C for 30 minutes. The reaction mixture was cooled to room temperature. After Filtration, the filtrate was concentrated to give the crude product, which was further purified by flash chromatography to afford tert-butyl l-(4-(2-chloro-5-(pyridin-2-ylethynyl)pyrimidin-4- ylamino)phenyl)cyclobutylcarbamate. (3.8 g, 80% yield). LCMS (ESI+): 476.2 [M+l]+ 478.2 [M+3]+
[00391] Step C: A mixture of tert-butyl l-(4-(2-chloro-5-(pyridin-2-ylethynyl)pyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (3.8 g, 8 mmol), KF (0.058 g, 1 mmol), Cs2CO3(0.652 g, 2 mmol) in dioxane (10ml) was stirred at 130°C in microwave for 1 hours. After removed the excess solvent , the residue was washed with water to get a solid, which was dried on vacumn to afford title compound tert-butyl l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (3.4 g, 90% yield). . LC MS (ESI+): 476.2 [M+l]+ 478.2 [M+3]+
Example 84
l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2 -d1pyrirnidin-7-yl')phenyl)cvclobutanamine
[00392] Step A: l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. LC/MS indicated tert-butyl l-(4-(2-(2- aminoethylamino)-6-(pyridin-2-yl)-7H -pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate was consumed. The product was collected by filtration, dried in vacuo to afford l-(4-(2-chloro-6- (pyridin-2-yl)-7H-pyrrolo [2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine as HCl salt. (80mg). Ή MR (MeOD-d4, 400 MHz), δ ppm: 8.99 (s, 1H), 8.42 (s, 1H), 7.87-7.85 (m, 1H), 7.60-7.56 (m, 3H), 7.43-7.40 (m, 2H ), 7.22-7.20 (m, 1H ), 7.18 (S, 1H), 2.78-2.70 (m, 2H), 2.59-2.55 (m, 2H), 2.25-2.20 (m, 1H), 2.02-1.95 (m, 1H). LC/MS: (ESI+): 376.2 [M+l]+, 378.2 [M+3]+
Example 85
Nl-(7-(4-(l-aminocyclobutynphenyl)-6-(pyridin-2-yl)-7H-pyrrolo 2.3-d1pyrimidin-2-yl)ethane-
1 ,2-diamine
[00393] Step A: To the solution of tert-butyl l-(4-(2-chloro-6-(pyridin-2-yl)-7H- pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (238 mg, 0.50 mmol) in dioxane (20 mL) was added ethane- 1,2-diamine (1 mmol). Then the reaction mixture was stirred at 120 °C overnight. The reaction mixture was diluted with water and extracted with ethyl acetate for three times, the combined organic phase was washed water and brine. The combined organic layer was dried over Na2S04 and then filtered, the filtrate was concentrated to give the crude product, which was further purified by flash chromatography to afford tert-butyl l-(4-(2-(2-aminoethylamino)-6-(pyridin-2-yl)- 7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl) cyclobutyl carbamate (120 mg, 49.5% yield) LC/MS
(ESI+): 500.2 [M+l]+ 502.2 [M+3]+
[00394] Step B: The tert-butyl l-(4-(2-(2-aminoethylamino)-6-(pyridin-2-yl)-7H- pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo, to afford Nl-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2- yl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl)ethane-l,2-diamine as HCl salt. (80mg).1H NMR (MeOD-d4, 400 MHz), δ ppm: 8.69(s, lH), 8.39 (s, 1H), 7.74-7.70 (m, lH), 7.58-7.54 (m, 2H), 7.41-7.39 (m, 3H,), 7.39-7.37(m, 1H), 6.99 (s, 1H), 3.63-3.60(m, 2H), 3.17-3.14 (m, 2H), 2.80-2.73 (m, 2H,), 2.65-2.58 (m, 2H), 2.26-2.23(m, 1H), 1.97-1.94(m, 1H). LC/MS: (ESI+): 400.2 [M+l]+, 402.2 [M+3]+
[00395] Examples 86 and 87 shown in Table 10 can also be made according to the above- described methods.
Table 10
Example 88
7-(4-n-aminocvclobutyl)phenyl)-N-methyl-6-(pyri
[00396] Step A: To a solution of l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (238 mg, 0.50 mmol) in dioxane (4 mL) was added the aqueous solution of methylamine (10 mL) at a sealed vessel. The reaction mixture was stirred at 100 °C overnight. The reaction mixture was diluted with ethyl acetate (80 mL), and washed by water (40 mL) twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product was used for step without purification. LC/MS (ESI+): 471.2 [M+l]+,473.2 [M+3]+
[00397] Step B: The tert-butyl l-(4-(2-(methylamino)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 °C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo, to afford 7-(4-(l-aminocyclobutyl) phenyl)-N-methyl-6-(pyridin-2-yl)- 7H-pyrrolo[2,3-d]pyrimidin-2-amineas as HCl salt. Ή NMR (MeOD-d4, 400 MHz), δ ppm: 8.60(s, 1H), 8.44-8.40 (s, 1H), 7.68-7.64(m, 1H), 7.51-7.48 (m, 2H), 7.29-7.21(m, 4H ), 6.94 (s, 1H), 2.88(s, 3H), 2.59-2.54 (m, 2H), 2.29-2.26 (m, 2H), 2.15-2.10(m, 1H), 1.91-1.79(m, 1H). LC/MS: (ESI+): 371.4.2 [M+l]+, 373.4.2 [M+3]+
Exam le 89
(S)-l-(7-(4-(l-aminocvclobutyl)phenyl)-6-(p idin-2-vn-7H-pyrrolo[2,3-d1pyrimidin-2- yl pyrrolidine-2-carboxylic acid
[00398] Step A: To a solution of { l-[4-(2-Chloro-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7- yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (0.20 g, 0.42 mmol) in MP(5 mL) were added Pyrrolidine-2-carboxylic acid (290 mg, 2.53 mmol) and K2C03 (232 mg, 1.684 mmol), the reaction mixture was heated at 120 °C for 12 hours. After cooling to room temperature, the mixture was concentrated in vacuo. Diluted with water and dried over sodium sulfate, then concentrated to give 160 mg of crude desired product which was used without purification.LC/MS (ESI+): 555.2 [M+l]+. 557.2 [M+l]+.
[00399] Step B: The l-{7-[4-(l-tert-Butoxycarbonylamino-cyclobutyl)-phenyl]-6-pyridin-2- yl-7H-pyrrolo[2,3-d]pyrimidin-2-yl}-pyrrolidine-2-carboxylic acid (160 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, dried in vacuo, to give target compounds as HCl salt. (44.4 mg, 33.9% yield). 1H NMR (MeOD, 400 MHz), δ ppm: 8.66 (s, 1H), 8.43 (s, 1H), 7.75-7.26 (m, 7H), 6.96 (s, 1H), 4.28 (s, 2H), 3.84 (s, 2H), 3.71 (d, J=4.4 Hz, 2H), 2.84-2.76 (m, 2H), 2.63-2.56 (m, 2H), 2:34- 2.09 (m, 4H), 2.04-1.95 (m, 2H). LC/MS (ESI+): 455.2 [M+lf
[00400] Examples 90 and 91 shown in Table 11 can also be made according to the above- described methods.
Table 11
N-(7-(4-(l-aminocvclobutyl)phenyl)-6-(p idin-2-ylV7H-p rolo[2,3-d1pyriniidin-2- vDcyclopropanecarboxamide
01] Step A: To a solution of compound { l-[4-(2-Chloro-6-pyridin-2-yl-pyrrolo[2,3-
d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (0.2g, 0.42mmol)in dioxane 10ml were added cyclopropanecarboxamide (98mg, 1.26mmol), Cs2C03 (258mg, 0.84mmol) and [(t-Bu)3P]2Pd (21.6mg, 0.042mmol) in a microwave tube, bubbled eith nitrogen for 5 minutes and heated to 140 °C for 30 minutes. After cooling to room temperature, filtered and concentrated it in vacuo, to give crude product (140 ,mg), which was used without further purification. LC/MS (ESI+): 525.2[M+1]+.
[00402] Step B: The (l-{4-[2-(Cyclopropanecarbonyl-amino)-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl] -phenyl }-cyclobutyl)-carbamic acid tert-butyl ester (140 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, dried in vacuo, to give target compounds as HCl salt. (51.5 mg). 1H NMR (MeOD, 400 MHz), δ ppm: 8.97 (s, 1H), 8.43 (d, J=4.0 Hz, 1H), 7.81-7.31 (m, 7H), 7.16 (m, 1H), 2.28-2.22 (m, 1H), 2.04-1.94 (m, 2H), 1.02-0.99 (m, 2H), 0.90-0.86 (m, 2H).
Example 93
l-(4-(6-(pyridin-2-yl)-7H-pyrrolo|"2.3-d]pyrimidin-7-yl)phenyl)cyclobutanamine
[00403] Step A: To a solution of l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutanamine (250 mg, 0.664 mmol) in methanol (30 mL) was added Pd/C (200 mg). The mixture was stirred at 50 C under the atmosphere of hydrogen (50 psi) for 12 h. The catalyst was moved by filtration, washed by methanol. The filtrate was concentrated to give l-(4-(6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine lH NMR (MeOD- (U, 400 MHz), δ ppm: 9.46 (s, 1H, ArH), 9.07 (s, 1H, ArH), 8.45 (s, 1H, ArH), 7.87-7.85 (m, 1H, ArH), 7.63-7.57 (m, 4H, ArH), 7.47-7.40 (m, 3H, ArH ), 2.75-2.70 (m, 2H, CH2), 2.60-2.53 (m, 2H, CH2), 2.25-2.20 (m, 1H, CH2), 2.02-1.95 (m, 1H, CH2). LC/MS: (ESI+): 342[M+1]+.
Example 94
1 -(4-(2-( 1 H-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolor2,3-d1pyrimidin-7- yDphenyPcyclobutanamine
[00404] Step A: The compound l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl) phenyl)cyclobutyl carbamate (500 mg, 1.05 mmol), lH-pyrazol-4-ylboronic acid (2.10 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was heated at 100 °C under nitrogen atmosphere for 4 h. Then the solvents were removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatograhpy to give tert-butyl l-(4-(2-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (160 mg) .LC/MS: (ESI+): 508.2 [M+l]+
[00405] Step B: The tert-butyl l-(4-(2-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (160 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 °C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo to afford l-(4-(2-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin -7-yl)phenyl) cyclobutanamine. LC/MS: (ESI+):408.2 [M+H]+. 1H NMR (MeOD-d4, 400 MHz), δ ppm: 9.07 (s, 1H), 8.45 (s, 1H), 8.18 (m, 2H), 7.81 (m, 1H), 7.62-7.31 (m, 6H ), 7.18 (s, 1H,), 2.85-2.80 (m, 2H), 2.65-2.58 (m, 2H), 2.25-2.20 (m, 1H), 2.02-1.95 (m, 1H). LC/MS: (ESI+):408.2 [M+H]+
Example 95
[00406] Step A: The compound l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo [2,3 -d]pyrimidin -7-yl)phenyl)cyclobutylcarbamate (238 mg, 0.5 mmol), methanesulfonamide (1 mmol), Pd2(dba)3 (19 mg, 0.02 mmol), Xantphos (15 mg, 0.03 mmol) and Cs2C03 (326 mg, 1 mmol) were dissolved in dioxane (10 mL). The reaction mixture was stirred at 90 °C under nitrogen atmosphere for 6 h. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (30 mL). The organic layer was washed by water dried over sodium sulfate, filtered and concentrated. The crude product was used for de-Boc step without purification. LC/MS: (ESI+): 536.2 [M+H]+.
[00407] Step B: The tert-butyl l-(4-(2-(methylsulfonamido)-6-(pyridin-2-yl)-7H- pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (150 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuoto afford N-(7-(4-(l -aminocyclobutyl)phenyl)-6-(pyridin-2- yl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl)methanesulfonamide as HC1 salt.. Ή NMR (MeOD-d4, 400 MHz), δ ppm: 8.91(s, 1H, ArH), 8.45 (s, 1H, ArH), 7.83-7.81(m, 1H, ArH), 7.59-7.53 (m, 3H, ArH), 7.48-7.45(m, 2H, ArH ), 7.38-7.36(m, 1H, ArH), 7.16 (s, 1H, ArH), 3.30(s, 3H, CH3), 2.80- 2.77 (m, 2H, CH2), 2.65-2.61 (m, 2H, CH2), 2.26-2.23(m, 1H, CH), 1.97-1.94(m, 1H, CH). LC/MS: (ESI+): 436.2 [M+l]+.
Example 96
7-(4-(l-aminocyclobutyl)phenyn-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d1pyrimidine-2-carbonitrile
[00408] Step A: l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (476 mg, 1 mmol), dicyanozinc (2 mmol), Pd2(dba)3 (190 mg, 0.2 mmol) and S-phos (82 mg, 0.2 mmol) were dissolved in l-methylpyrrolidin-2-one (10 mL). The reaction mixture was stirred at 150 °C under nitrogen atmosphere for 30 minutes. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (30 mL). The organic layer was washed by water dried over sodium sulfate, filtered and concentrated. The crude product
was purified by flash chromatography give 200 mg tert-butyl l-(4-(2-cyano-6-(pyridin-2-yl)-7H- pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate. LC/MS: (ESI+): 467.2 [M+l]+, 489.2 [M+23]+.
[00409] Step B: The tert-butyl l-(4-(2-cyano-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-
7-yl)phenyl)cyclobutylcarbamate. (100 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 2 h. The product was collected by filtration, dried in vacuo to afford 7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-2- carbonitrile as HC1 salt.'H NMR (DMSO-de, 400 MHz), δ ppm: 9.34(s, IH, ArH), 8.23 (s, IH, ArH), 7.85-7.81(m, IH, ArH), 7.59-7.52 (m, 4H, ArH, H2), 7.45(S, IH, ArH ), 7.38-7.30(m, 4H, ArH), 2.26-2.19 (m, 3H, CH2, CH), 2.06-2.04 (m, 2H, CH2), 1.74-1.70(m, IH, CH). LC/MS: (ESI+): 367.2 [M+l]+.
Example 97
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo 2,3-dlpyrimidine-2-carboxamide
[00410] Step A: The tert-butyl l-(4-(2-cyano-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-
7-yl)phenyl)cyclobutylcarbamate. (100 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for overnight. The product was collected by filtration, dried in vacuo to afford 7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidine-2-carboxamide as HC1 salt.. 1H NMR (MeOD-d4, 400 MHz), δ ppm: 8.60(s, IH, ArH), 8.44-8.40 (s, IH, ArH), 7.68-7.64(m, IH, ArH), 7.51-7.48 (m, 2H, ArH), 7.29-7.21(m, 4H, ArH ), 6.94 (s, IH, ArH), 2.88(s, 3H, CH3), 2.59-2.54 (m, 2H, CH2), 2.29-2.26 (m, 2H, CH2), 2.15- 2.10(m, IH, CH), 1.91-1.79(m, IH, CH). LC/MS: (ESI+): 385.2 [M+l]+
tert-butyl l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolor2,3-d1pyrimidin-7- yPphenyDcyclobutylcarbamate
[00411] Step A: To the solution of 4,6-dichloro-5-iodopyrimidine (5.50 g, 20.00 mmol)
(prepared according to the ref of Organic Letters; English; 11; 8; 2009; 1837 - 1840; ) in dioxane (100 mL) was slowly added the solution of tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (5.2 g, 20.00 mmol) in dioxane (20 mL) and Et3N(5 ml). The reaction mixture was stirred at 80C for overnight, lc-ms indicated 4,6-dichloro-5-iodopyrimidine was completed consumed. After removed the excess solvents under the reduced pressure to get a residue, which was dissolved in Ethyl acetate (250 mL) and washed water and brine. The combined organic layer was dried over Na2S04 and then filtered, the filtrate was concentrated to give the crude product, which was further purified by flash chromatography to afford tert-butyl l-(4-(6-chloro-5-iodopyrimidin-4- ylamino)phenyl)cyclobutylcarbamate. (5.00 g 50% yield).LC/MS: (ESI+): 501 [M+l], 523 [M+Na].
[00412] Step B: tert-butyl l-(4-(6-chloro-5-iodopyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (5.00 g, 0.01 mol),and 2-ethynylpyridine (1.55 g, 0.015 mol) was added in a lOOmL flask which contained 30 mL of Et3N with stirring, and then the catalyst of Cul(0.38 g, 0.002 mol)and Pd(PPh3)2Cl2(2 g, 0.002 mol) was added into the mixture. The reaction mixtute was warmed to 80 °C for 30 minutes, The reaction mixture was cooled to room
temperature. Filltered and the filtrate was concentrated to give the curde product, which was further purified by flash chromatography to afford tert-butyl l-(4-(6-chloro-5-(pyridin-2- ylethynyl)pyrimidin-4-ylamino)phenyl)cyclobutylcarbamate. (3.8 g, 80% yield). LC/MS (ESI+): 476.2 [M+l]+
[00413] Step C: A mixture of tert-butyl l-(4-(6-chloro-5-(pyridin-2-ylethynyl)pyrimidin-4- ylamino)phenyl)cyclobutylcarbamate (3.8 g, 8 mmol), KF(0.058 g, 1 mmol), Cs2CO3(0.652 g, 2 mmol) in dioxane(lOml) was stirred at 130°C in microwave for 1 hours. A residue was afford after removed the excess solvent , the residue was washed with water to get a solid, then the solid was dried on vacumn to afford tert-butyl l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (3.4 g, 90% yield). LC/MS (ESf ): 476.2 [M+lf.
Example 98
[00414] Step A: To a solution of tert-butyl l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d] pyrimidin-7-yl)phenyl)cyclobutylcarbamate (476mg, lmmol) in methanol (10 mL) was slowly added CH3ONa (108mg, 2.00 mmol) in portions. The reaction mixture was stirred at room temperature overnight. Then it was concentrated to remove the solvent. The obtained residue was dissolved in Ethyl acetate (250 mL) and washed by water, followed by brine. The separated organic layer was dried in sodium sulfate, filtered and concentrated to give tert-butyl l-(4-(4-methoxy-6- (pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate. (450 mg 50% yield) .LC/MS (ESI+): 472.2 [M+l]+
[00415] Step B: The tert-butyl l-(4-(4-methoxy-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 2 h. The product was collected by filtration, dried in vacuo, to afford l-(4-(4-methoxy-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl) phenyl) cyclobutanamine as HC1 salt. (20mg). [M+l]+ nH NMR (MeOD-cU, 400 MHz), δ ppm: 8.41-8.39(m, 2H, ArH), 7.77-7.73 (m, 1H, ArH), 7.59-7.57(m, 2H, ArH), 7.48-7.40 (m, 3H, ArH), 7.29-7.27(m, 1H, ArH ), 7.1 l(s, 1H, ArH), 4.17(s, 3H, CH3), 2.81-2.75 (m, 2H, CH2), 2.64-2.59 (m, 2H, CH2), 2.15-2.10(m, 1H, CH), 1.91-1.79(m, 1H, CH). LC/MS (ESI+): 372.2
Example 99
7-(4-(l-aminocvclobutyl)phenyl)-N-methyl-6-(pyridin^
[00416] Step A: To a solution of l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (238 mg, 0.50 mmol) in dioxane (4 mL) was added the aqueous solution of methylamine (10 mL) at a sealed vessel. The reaction mixture was stirred at 100 °C overnight. The mixture was diluted with ethyl acetate (80 mL), and washed by water (40 mL) twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product was used without purification. LC/MS (ESI ): 471.2 [M+l]
[00417] Step B: The tert-butyl l-(4-(4-(methylamino)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 °C and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo, to afford 7-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-(pyridin-2-yl)-7H- pyrrolo [2,3-d] pyri midin-4-amine as HCl salt.: (M+H:371.4.2). 1H NMR (MeOD-d,, 400 MHz), δ ppm: 8.3 l(s, 1H, ArH), 8.07 (s, 1H, ArH), 7.77-7.67(m, 1H, ArH), 7.49-7.46 (m, 2H, ArH), 7.31- 7.17(m, 4H, ArH ), 7.03 (s, 1H, ArH), 3.03(s, 3H, CH3), 2.71-2.69 (m, 2H, CH2), 2.53-2.50 (m, 2H, CH2), 2.15-2.10(m, 1H, CH), 1.91-1.79(m, 1H, CH). LC/MS: (ESI+)
Example 100
7-(4-(l-aminocyclobutyl)phenyn-N,N-diethyl-6-(pyridin-2-yl)-7H-pwolo[2,3-d]pyrimidin-4-
amine
[00418] Example 101 shown in Table 12 can also be made according to the above-described methods.
Table 12
Example 102
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo 2,3-d1pyrimidin-4-ol
[00419] The tert-butyl l-(4-(4-methoxy-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in water (30 mL), the solution was cooled to 0 °C and the solution of HCl in water (12M, 8 mL) was added slowly. The mixture was stirred at 100 °C for 2 h. The mixture was concentrated, dried in vacuo to afford 7-(4-(l- aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-ol as HCl salt.. !H NMR
(MeOD-dU, 400 MHz), δ ppm: 8.36(s, 1H, ArH), 7.91 (s, 1H, ArH), 7.74-7.70(m, 1H, ArH), 7.57- 7.54 (m, 2H, ArH), 7.43-7.23(m, 4H, ArH ), 7.16 (s, 1H, ArH), 2.79-2.74 (m, 2H, CH2), 2.63-2.58 (m, 2H, CH2), 2.35-2.30(m, 1H, CH), 1.91-1.79(m, 1H, CH). LC/MS: (ESI+): :358.2[M+H]+
Example 103
l-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-ylV7H-pyre^
one
[00420] Step A: The compound l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (238 mg, 0.5 mmol), pyrrolidin-2-one (1 mmol), Pd2(dba)3 (19 mg, 0.02 mmol), Xantphos (15 mg, 0.03 mmol) and Cs2C03 (326 mg, 1 mmol) were dissolved in dioxane (10 mL). The reaction mixture was stirred at 90 °C under nitrogen atmosphere for 6 h.. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (30 mL). The organic layer was washed by water dried over sodium sulfate, filtered and concentrated. The crude product was used without purification. LC/MS: (ESI+): 525.2 [M+l]+.
[00421] Step B: The tert-butyl l-(4-(4-(2-oxopyrrolidin-l-yl)-6-(pyridin-2-yl)-7H-pyrrolo
[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (150 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo, to afford l-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl)pyrrolidin-2-one as HC1 salt. 1H NMR (MeOD-d4, 400 MHz), δ ppm: 8.52(s, 1H, ArH), 8.41 (s, 1H, ArH), 7.81-7.77(m, 1H, ArH), 7.59-7.29 (m, 7H, ArH), 4.26-4.22(m, 2H, CH2 ), 2.83-2.61 (m, 6H, 3CH2), 2.30-2.00 (m, 3H, CH2, CH), 1.91-1.79(m, 1H, CH). LC/MS: (ESI+): 425.2 [M+H]+
Example 104
1 -(4-(4-( 1 H-p azol-4-yl)-6-(pyridin-2-yl)-7H-pyrroloi2,3-d1pyrimidin-7- vDphenvPcyclobutanamine
[00422] Step A: The compound tert-butyl l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyI)cyclobutylcarbamate (500 mg, 1.05 mmol), lH-pyrazol-4-ylboronic acid (2.10 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was heated at 100 °C under nitrogen atmosphere for 4 h. Then the solvents were removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatograhpy to give 160 mg of tert-butyl l-(4-(4-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate.LC/MS: (ESI+): 508.2 [M+l]
[00423] Step B: The tert-butyl l-(4-(4-(lH-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (160 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo to afford l-(4-(4-(lH-pyrazol-4-yl) -6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutanamine,.. Ή NMR (MeOD-dt, 400 MHz), δ ppm: 8.73 (s, 1H, AxH), 8.61 (s, 1H, ArH), 8.40 (s, 1H, ArH), 7.80-7.76 (m, 1H, ArH), 7.66-7.56 (m, 4H, ArH), 7.45- 7.43 (m, 2H, ArH ), 7.29 (s, 1H, ArH), 2.80-2.74 (m, 2H, CH2), 2.62-2.55 (m, 2H, CH2), 2.25-2.20 (m, 1H, CH2), 2.02-1.95 (m, 1H, CH2). LC/MS: (ESI+): 408.2 [M+H]+
Example 105
l-(7-(4-(l-aminocyclobutyl)phenyl)-6-(p idin-2-yl)-^
ol
[00424] Step A: To a solution of tert-butyl l-(4-(4-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (238 mg, 0.50 mmol) in dioxane (20 mL) was added pyrrolidin-3-ol hydrochloride (1 mmol) and K2C03 (2 mmol). Then the reaction mixture was stirred at 120 °C overnight. The reaction mixture was diluted with water and extracted with ethyl acetate for several times, the combined organic phase was washed water and brine. The combined organic layer was dried over Na2S04 and then filtered, the filtrate was concentrated to give the crude product, which was further purified by flash chromatography to afford tert-butyl l-(4-(4-(3- hydroxypyrrolidin-l-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutylcarbamate (120 mg, 49.5% yield) LC-MS (ESI+): 527.2 [M+l]+
[00425] Step B: The tert-butyl l-(4-(4-(3-hydroxypyrrolidin-l-yl)-6-(pyridin-2-yl)-7H- pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (120 mg) was dissolved in ethyl acetate (30 mL). The solution was cooled to 0 degree and the solution of HCl in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, dried in vacuo to afford l-(7-(4-(l-aminocyclobutyl)phenyl) -6-(pyridin-2- yl)-7H-pyrrolo [2,3-d]pyrimidin-4-yl)pyrrolidin-3-ol as HCl salt. (80 mg). Ή NMR (MeOD- U, 400 MHz), δ ppm: 8.28(s, 1H, ArH), 8.02 (s, 1H, ArH), 7.62-7.58(m, 1H, ArH), 7.49-7.46 (m, 2H, ArH), 7.31-7.26(m, 3H, ArH ), 7.21-7.14(m, 2H, ArH ), 4.5 l(s, 1H, CH), 4.01-3.82(m, 4H, 2CH2), 2.72-2.57 (m, 2H, CH2), 2.54-2.49 (m, 2H, CH2), 2.17-2.1 l(m, 3H, CH, CH2), 1.91-1.79(m, 1H, CH). LC/MS: (ESI+): 427.2 [M+l]+.
Example H
7-[4-(l-Amino-cvclobutyl)-phenyl]-6-pyridin-2-yl-7H-pmolo[2J-d1pyrimidine-4-carboxylic acid methyl ester
[00426] Step A: To the solution of l-[4-(4-Chloro-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-
7-yl)-phenyl]-cyclobutylamine (2.0 g, 4.2mmol) in a mixed solution (DMF: 35 ml and MeOH: 15ml) were added Pd(OcA)2 (lOOmg, 0.45mmol), dppf (234mg, 0.42mmol) and TEA (5ml). it was stirred at 80°C under the atmosphere of CO (50 psi) for 14 hours. It was concentrated to remove the solution, the residue was dissolved in EA and washed by water twice, the separated organic layer was dried over sodium sulfate, filtered and concentrated to get crude product, which was purified by flash chromatography, to give 7-[4-(l-Amino-cyclobutyl)-phenyl]-6-pyridin-2-yl-7H- pyrrolo[2,3-d]pyrimidine-4-carboxylic acid methyl ester (1.3g 62% yield) LC/MS: 500.2 [M+l]+
Example I
7-(4-(l-(tert-butoxycarbonylamino)cvclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3- dlpyrimidine-4-carboxylic acid
[00427] 7-[4-( 1 -tert-Butoxycarbonylamino-cyclobutyl)-phenyl]-6-pyridin-2-yl-7H- pyrrolo[2,3-d]pyrimidine-4-carboxylic acid methyl ester (150 mg, 0.3 mmol) was dissolved in IN LiOH (10ml) and MeOH (10 mL) and then the reaction mixture was stirred at 30 °C for 2 hours. Methanol was removed under reduced pressure and the mixture was acidified with HC1 (6 N) carefully till PH= 5, the resulting solid was collected by filtration, dried over vaccum to give 7-(4- (l-(tert-butoxycarbonylamino)cyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-4- carboxylic acid.(120 mg, 82.3% yield).
Example 106
7-(4-n-aminocvclobutyl phenyl)-6-(p ^ acid
[00428] To a solution of 7-[4-(l-tert-Butoxycarbonylamino-cyclobutyl)-phenyl]-6-pyridin-
2-yl-7H-pyrrolo[2,3-d]pyrimidine-4-carboxylic acid (80 mg) in EtOAc (5ml) was added HC1/EA (15ml) at 0 °C. The reaction mixture was stirred at ambient temperature for 3 hours. It was concentrated to give crude product, which was purified by preparative HPLC to give 7-(4-(l- aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-4-carboxylic acid, (37 mg, 58% yield) LC/MS (ESI+): 385.9[M+1]+.
Exam le 107
7-(4-(l-aminocvclobutynphenyl)-6-(pyridin-2-yl)-7H-pyrrolor2,3-d pyrimidine-4-carboxamide
[00429] Step A: To a solution of NH3 in THF (10 M, 20 mL) was added 7-[4-(l-tert-
Butoxycarbonylamino-cyclobutyl)-phenyl]-6-pyridin-2-yl-7H-pyrrolo[2,3-d]pyrimidine-4- carboxylic acid methyl ester (0.10 g, 0.2 mmol) in was dissolved in THF (20 mL) in at a sealed vessel , the reaction mixture was stirred at 120 °C for 6 hours. The reaction mixture was concentrated in vacuo, and pured into next reaction without purification. LC/MS (ESI+): 485.1 [M+H]+
[00430] Step B: { l-[4-(4-Carbamoyl-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (150 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, dried in vacuo to give 7-[4-(l-Amino-cyclobutyl)-phenyl]-6-pyridin-2-yl-7H-pyrrolo[2,3-
d]pyrimidine-4-carboxylic acid amide as HCl salt (16.8mg). H NMR (MeOD, 400 MHz), δ ppm: 8.88(s, 1H), 8.46-8.44(m, 1H), 7.85-7.58(m, 5H), 7.46-7.43(m, 2H), 7.38-7.34(m, 1H), 2.82- 2.75(m, 2H), 2.60-2.53(m, 2H), 2.25-2.22(m,lH), 1.98-1.92(m, 1H). LC/MS (ESI+): 407.1 [M+Na]+
[00431] Example 108 shown in Table 13 can also be made according to the above-described methods.
Table 13
Examples J and K
ferf-butyl 1 -(4-(2-chloro-4-(diethylamino)-6- ferf-butyl 1 -(4-(2-chloro-4-methoxy-6-(pyridin-2-yl)-7H- (pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl) pyrrolo[2,3-cf|pyrimidin-7-yl)phenyl)cyclobutylcarbamate -phenyl)cyclobutylcarbamate
[00432] Step A: To a solution of 2,4,6-Trichloro-5-iodo-pyrimidine (6.16 g, 20.00 mmol) in dioxane (100 mL) was slowly added the solution of tert-butyl l-(4- aminophenyl)cyclobutylcarbamate (5.2 g, 20.00 mmol) in dioxane (20 mL) and triethylamine (5 ml). The reaction mixture was stirred at 80 C for overnight, lc-ms indicated 2,4,6-Trichloro-5-iodo- pyrimidine was completed consumed. After removed the excess solvents under the reduced pressure to get a residue, which was dissolved in Ethyl acetate (250 mL) and washed water and
brine. The combined organic layer was dried over Na2S04 and then filtered, the filtrate was concentrated to give the crude product, which was further purified by flash chromatography to afford { l-[4-(2,6-Dichloro-5-iodo-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert- butyl ester (5.50 g, yield: 50%).LC/MS: (ESI+): 535 [M+H]+.
[00433] Step B: { l-[4-(2,6-Dichloro-5-iodo-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (5.30 g, 0.01 mol),and 2-ethynylpyridine (1.55 g, 0.015 mol) was added in a lOOmL flask which contained 30 mL of Et3N with stirring, and then the catalyst of Cul(0.38 g, 0.002 mol)and Pd(PPh3)2C12(2 g, 0.002 mol) was added into the mixture. The reaction mixtute was warmed to 80 oC for 90 minutes, lc-ms indicated {l-[4-(2,6-Dichloro-5-iodo- pyrimidin-4-ylamino)-phenyl]-cyclobutyl} -carbamic acid tert-butyl ester was consumed. The reaction mixture was cooled to room temperature. Filtrated, the filtrate was concentrated to give the curde product, which was further purified by flash chromatography to afford (4.1 g, yield:80%){l-[4-(2,6-Dichloro-5-pyridin-2-ylethynyl-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester and(2.1 g, yield:40%) LC/MS (ESI+): 510.2 [M+l]+ (M+Na:522).{l- [4-(2-Chloro-6-diethylamino-5-pyridin -2-ylethynyl -pyrimidin-4-ylamino)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester . (2.0 g, 40% yield). LC/MS (ESI+): 547.2 [M+l]+ .
[00434] Step C: A mixture of {l-[4-(2,6-Dichloro-5-pyridin-2-ylethynyl-pyrimidin-4- ylamino)-phenyl]-cyclobutyl} -carbamic acid tert-butyl ester (2.1 g, 8 mmol)and , KF(0.058 g, 1 mmol), Cs2CO3(0.652 g, 2 mmol) in dioxane(lOml) was stirred at 130°C in microwave for 1 hours. A residue was afford after removed the excess solvent , the residue was washed with water to get a solid, then the solid was dried on vacumn to afford {l-[4-(2,4-Dichloro-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl} -carbamic acid tert-butyl ester (3.4 g, yield: 90%). . LC/MS (ESI+): 510.2 [M+l]+ 522 [M+Na].
[00435] Step D: A mixture of { l-[4-(2-Chloro-6-diethylamino-5-pyridin-2-ylethynyl- pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (2.0 g, 7 mmol) and, KF (0.058 g, 1 mmol), Cs2C03 (0.652 g, 2 mmol) in dioxane (10ml) was stirred at 130°C in microwave for 1 hours. A residue was afford after removed the excess solvent , the residue was washed with water to get a solid, then the solid was dried on vacumn to afford { l-[4-(2-Chloro-4-diethylamino- 6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (3.6 g, yield: 90%). LC/MS (ESI+): 547.2 [M+l]+
7- 4-(l-Amino-cyclobutvD-phenyl]-2-(2-fluoro-phenyl)-6-p idin-2-yl-3J-dihydro-pyrrolo[2^ dlpyrimidin-4-one
[00436] Step A: To the solution of {l-[4-(2,4-Dichloro-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (510 mg, 10 mmol) in methanol (10 mL) was slowly added CH3ONa (108 mg, 20.00 mmol) in portions. The reaction mixture was stirred at room temperature overnight. Then it was concentrated to remove the solvent. The obtained residue was dissolved in Ethyl acetate (250 mL) and washed by water, followed by brine. The separated organic layer was dried in sodium sulfate, filtered and concentrated to give { 1- [4-(2-Chloro-4-methoxy-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester. (450 mg, 50%yield) LC/MS: (ESI+): 506.3 [M+l]+
[00437] Step B: The { l-[4-(2-Chloro-4-methoxy-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7- yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (253 mg, 0.50 mmol), boronic acid (0.75 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was heated at 100 °C under nitrogen atmosphere for 4 h. Then the solvents were removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatogrphy to afford of (l-{4-[2-(2-Fluoro-phenyl)-4-methoxy-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}- cyclobutyl)-carbamic acid tert-butyl ester (160 mg). LC/MS: (ESI+): 566.3 [M+l]+
[00438] Step C: The (l-{4-[2-(2-Fluoro-phenyl)-4-methoxy-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (120 mg) was dissolved in water (30 mL), the solution was cooled to 0 °C and the solution of HCl in water (12M, 10 mL) was added slowly. The mixture was stirred at 100 °C for 1 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford 7-[4-(l-Amino-cyclobutyl)- phenyl]-2-(2-fluoro-phenyl)-6-pyridin-2-yl-3,7-dihydro-pyrrolo[2,3-d]pyrimidin-4-one. !H MR (MeOD-d4, 400 MHz), δ ppm: 8.31 (s, 1H, ArH), 7.67-7.65 (m, 2H, ArH), 7.64-7.49 (m, 3H, ArH), 7.47-7.36 (m, 3H, ArH), 7.22-7.16 (m, 3H, ArH ), 7.14 (s, 1H, ArH), 2.75-2.65 (m, 2H, CH2), 2.61- 2.50 (m, 2H, CH2), 2.15-2.10 (m, 1H, CH), 1.91-1.79 (m, 1H, CH). LC/MS (ESI+): 453.3 [M+l]+
Example 110
r7-r4-(l-Amino-cvclobutyl -phenyll-2- H-p azol-4-yl)-6-pyridin-2-yl-7H-pyrrolo[2,3- d]pyrimidin-4-yll-diethyl-amine
[00439] Step A: The { l-[4-(2-Chloro-4-diethylamino-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (273 mg, 0.50 mmol), boronic acid (0.75 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was heated at 100 °C under nitrogen atmosphere for 4 h. Then the solvents were removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatogrphy to afford of (l-{4-[4-Diethylamino-2-(lH-pyrazol-4-yl)-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester(160 mg) . LC MS: (ESI+): 579.3 [M+l
[00440] Step B: The (l-{4-[4-Diethylamino-2-(lH-pyrazol-4-yl)-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl] -phenyl }-cyclobutyl)-carbamic acid tert-butyl ester (120 mg) was dissolved in water (30 mL), the solution was cooled to 0 °C and the solution of HCl in ethyl acetate(4M, 10 mL) was added slowly. The mixture was stirred at room temperature for 1 h. The crude product was collected by filtration, which was further purified by preparative HPLC to afford [7-[4-(l-Amino- cyclobutyl)-phenyl]-2-(lH-pyrazol-4-yl)-6-pyridin-2-yl-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-diethyl- amine. 1H NMR (MeOO-< , 400 MHz), δ ppm: 8.50(s, 1H, ArH), 8.38 (s, 1H, ArH), 7.72-7.70 (m, 1H, ArH), 7.54-7.52 (m, 2H, ArH), 7.45-7.41 (m, 3H, ArH), 7.29 (s, 1H, ArH), 7.26-7.23 (m, 1H, ArH), 3.70-3.65 (m, 4H, 2CH2), 2.86-2.78 (m, 2H, CH2), 2.67-2.59 (m, 2H, CH2), 2.23-2.06 (m, 1H, CH), 2.04-1.98 (m, 1H, CH),1.21-1.18 (m, 6H, 2CH3). LC/MS (ESI+): 479.3 [M+l]+
Example 111
l-{4-r4-Memoxy-2-(lH-pyrazol-4-yl)-6-pyridin-2-yl-pyro
cyclobutylamine
[00441] Step A: The { l-[4-(2-Chloro-4-methoxy-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7- yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (253 mg, 0.50 mmol), boronic acid (0.75 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was heated at 100 °C under nitrogen atmosphere for 4 h. Then the solvents were removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by flash chromatogrphy to afford ( 1 - {4- [4-Methoxy-2-( 1 H-pyrazol-4-yl)-6-pyridin-2-yl-pyrrolo [2,3 -d]pyrimidin-7-yl] -phenyl } - cyclobuty -carbamic acid tert-butyl ester (160 mg). LC/MS: (ESI+): 538.3 [M+l]+
[00442] Step B: The (l-{4-[4-Methoxy-2-(lH-pyrazol-4-yl)-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (120 mg) was dissolved in water (30 mL), the solution was cooled to 0 C and the solution of HC1 in water (12M, 10 mL) was added slowly. The mixture was stirred at 100 °C for 1 h.. The crude product was collected by filtration, which was further purified by preparative HPLC to afford l-{4-[4-Methoxy-2-(lH- pyrazol-4-yl)-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}-cyclobutylamine. Ή NMR (MeOD-d4, 400 MHz), δ ppm: 8.50 (s, 1H, ArH), 8.41 (s, 1H, ArH), 8.29 (s, 1H, ArH), 7.76-7.72 (m, 2H, ArH), 7.59-7.53 (m, 3H, ArH), 7.30-7.26 (m, 1H, ArH), 7.00 (s, 1H, ArH), 4.21 (s, 3H, CH3), 2.86-2.78 (m, 2H, CH2), 2.67-2.59 (m, 2H, CH2), 2.23-2.06 (m, 1H, CH), 2.04-1.98 (m, 1H, CH). LC/MS (ESI+): 438.3 [M+l]+
Example 112
[00443] Step A: To a solution of 3-chloro-4-iodopyridine (1 g, 4.2 mmol), 1-ethynylbenzene
(430 mg, 4.2 mmol), and Pd(PPh3)2Cl2(147 mg, 0.21 mmol) in TEA (30 mL) was added Cul (40 mg, 0.21 mmol). The mixture was stirred at reflux for 60min.The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate =100/1-5/1), to give the title product 3-chloro-4-(2- phenylethynyl)pyridine (0.85 g, 95%). 'HNMR: CDC13 400MHZ δ 8.67(s, 1H), 8.47~8.45(d, 1H, J=8), 7.70~7.68(m, 2H), 7.44~7.26(m, 4H).
[00444] Step B: To a solution of 3-chloro-4-(2-phenylethynyl)pyridine (426 mg, 2 mmol),
Pd2(dba)3 (90 mg, 0.1 mmol), X-phos (190 mg, 0.4 mmol) and Cs2C03 (978 mg, 3 mmol) in Dioxane (8 mL) was added tert-butyl l-(4-aminophenyl)cyclobutylcarbamate (524 mg, 2 mmol). The mixture degassed and charged with N2 for three times. And then the resulting mixture was stirred at 120 °C for 60 min by microwave irradiation. The reaction mixture was filtered by filter paper. The filtrate was concentrated under reduced pressure to get crude product, which was purified flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate =100/1-1/1) to yield the desired product fert-butyl l-(4-(2-phenyl-lH-pyrrolo[2,3-c ] pyridin-1- yl)phenyl ) cyclobutylcarbamate (100 mg, 11%). 'HNMR: MeOD 400MHz δ 8.55(s,lH), 8.25~8.23(d, 1H, J=8), 7.84~7.82(d, 1H, J=8), 7.58-7.56 (m, 2H), 7.36-7.28 (m, 7H), 7.02 (s, 1H), 2.52-2.46 (m,4H), 2.15-2.00 (m,lH), 1.95-1.82 (m,lH), 1.36 (s,9H)
[00445] Step C: To a solution of HC1 in EtOAc (30 mL, 2 M) was added tert-butyl l-(4-(2- phenyl-lH-pyrrolo[2,3-c] pyridin-l-yl)phenyl) cyclobutylcarbamate (100 mg, 0.23 mmol). The mixture was stirred at r.t. for 60min. The reaction mixture was filtered to give l-(4-(2-phenyl-lH- pyrrolo[2,3-c]pyridin-l-yl)phenyl)cyclobutanamine (51 mg).1HNMR: MeOD 400MHz 6 8.79(s, 1H), 8.35~8.33(d, 1H, J=8), 8.23~8.21(d, 1H, J=8), 7.74~7.72(m, 2H), 7.59~7.57(m, 2H), 7.46~7.44(m, 3H), 7.39~7.37(m, 2H), 7.34(s, 1H), 2.80~2.76(m, 2H), 2.68~2.64(m, 2H), 2.24~2.20(m, 1H), 2.00-1.96(m, 1H). MS (ESI) m/z: 340.2 [M+l]+. 342.2 [M+3]+
Example 113
l-[4-(6-Phenyl-pyrrolo[2,3-b1pyrazin-5-yl)-phenyl1-cvclobutylamine
[00446] Step A: 2,3-Dichloro-pyrazine (1.48 g, 0.01 mol),and Ethynyl-benzene (1.53 g, 0.015 mol) was added in a lOOmL flask which contained 30mL of Et3N with stirring, and then the catalyst of Cul(0.38 g, 0.002 mol)and Pd(PPh3) 2C12(2 g, 0.002 mol) was added into the mixture. The reaction mixture was warmed to 80°C for 0.5h, lc-ms indicated 2,3-Dichloro-pyrazine was consumed. The reaction mixture was cooled to room temperature. Filtrated, the filtrate was concentrated to give the crude product, the crude product was further purified by flash chromatography to afford 2-Chloro-3-phenylethynyl-pyrazine. (1.5 g, yield: 71%). LC/MS (ESI+): 215 [M+l]+ 217 [ +3]+
[00447] Step B: A mixture of 2-Chloro-3-phenylethynyl-pyrazine (1.5 g, 7 mmol), [l-(4-
Amino-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (2.2 g, 8.4 mmol), Cs2C03(4.5 g, 14 mmol) ,X-phos(0.65 g, 1.4 mmol) and Pd2(dba)3 (1.3 g, 1.4 mmol) in dioxane(10 ml) was stirred at 120°C in microwave for 1 hours. The reaction mixture was cooled to room temperature , then poured into water and extracted with ethyl acetate for three times, the combined organic layer was washed with water and brine, dried and concentrated to afford the which was further purified by flash chromatography to afford { l-[4-(3-PhenylethynyI-pyrazin-2-ylamino)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (1.2 g, 40 % yield). LC/MS (ESI+): 441 [M+l]+ ,443 [M+3]+
[00448] Step C: A mixture of { l-[4-(3-Phenylethynyl-pyrazin-2-ylamino)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (0.44 g, 1 mmol), KF(0.058 g, 1 mmol), Cs2CO3(0.652 g, 2 mmol) , and Pd(t-Bu3P)2 (0.15 g, 0.01 mmol) in dioxane(lOml) was stirred at 130°C in microwave for lh. After cooled to room temperature ,the reaction mixture was poured into water and extracted with ethyl acetate for three times, then the combined organic layer was washed with water and brine, dried and concentrated to give a residue, which was further purified by flash chromatography to afford {l-[4-(6-Phenyl-pyrrolo[2,3-b]pyrazin-5-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (0.3 g, yield: 68%). LC/MS: (ESI+): 441 [M+l]+ ,443 [M+3]+
[00449] Step D: The solution of l-[4-(6-Phenyl-pyrrolo[2,3-b]pyrazin-5-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (0.3 g, 0.7 mmol) in ethyl acetate was added the solution of HC1 in ethyl acetate (4mol/L HC1, 10 ml), the reaction mixture was stirred at room temperature for 1 h. After remove the solvents to afford a residue,which was further purified by prep-hplc to afford l-[4-(6-Phenyl-pyrrolo[2,3-b]pyrazin-5-yl)-phenyl]-cyclobutylamine. (120 mg, yield: 49.5%). 'HNM (DMSO-< , 400 MHz): δ 8.49 (d, J = 2.8Hz 1H,), δ 8.22 (d, J = 2.8Hz 1H,), δ 7.51 (d, J = 8.4Hz 2H,), δ 7.25(d, J=8.4Hz 2H,), 7.39 - 7.33 (m, 5H),7.07(s, 1H), 2.48 (m, 2H), 2.38 (m, 2H), 2.0 (m, 1H), 1.7 (m, 1H). LC/MS: (ESI+): 341 [M+l]+ ,343 [M+3]+
{ l-f4-(2-Chloro-6-phenyl-pyrrolof3,2-dlpyrimidin-5-yl)-phenyl1-cvclobutyll-carbamic acid tert- butyl ester
[00450] Step A: To a solution of 5-bromo-2,4-dichloropyrimidine (10.00 g, 43.88 mmol) in
TEA (100 mL) were added ethynylbenzene (4.48 g, 43.88 mmol), Pd(PPh3)2Cl2 (1.00 g, 1.42 mmol), and Cul (1.67 g, 8.76 mmol). The reaction mixture was stirred at 100 °C under the atmosphere of nitrogen for 2 h. After cooling, the solvent was removed by concentration, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered, and concentrated. The crude product was purified by column separation (eluted by: hexane / ethyl acetate = 20: 1) to afford 5-bromo-2-chloro-4- (phenylethynyl)pyrimidine. (7.10 g, 55% yield). 1H MR (DMSO, 400 MHz), δ ppm: 9.11 (s, 1H), 7.73-7.71 (m, 2H), 7.62-7.52 (m, 3H). MS (ESI+) e/z: 295 [M+l]+.
[00451] Step B: To a solution of 5-bromo-2-chloro-4-(phenylethynyl)pyrimidine (1.80 g,
6.13 mmol) in dioxane (20 mL) were added [l-(4-Amino-phenyl)-cyclobutyl]-carbamic acid tert- butyl ester (1.45 g, 5.53 mmol), Pd2(dba)3 (0.28 g, 0.31 mmol), X-Phos (0.29 g, 0.61 mmol), and Cs2C03 (3.99 g, 12.25 mmol). The reaction mixture was stirred at 100 °C under the atmosphere of nitrogen for 4 h.. After cooling, the solvent was removed by concentration, and the residue was dissolved in ethyl acetate. The solution was washed by water twice. The separated organic layer was dried over sodium sulfate, filtered, and concentrated. The crude product was purified by column separation (eluted by: hexane / ethyl acetate = 10 : l to 3 : l) to afford tert-butyl l-(4-(2- chloro-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-5-yl)phenyl)cyclobutylcarbamate (2.10 g, 72% yield). Ή NMR (MeOD, 400 MHz), δ ppm: 8.52 (s, 1H), 7.59-7.56 (m, 2H), 7.40-7.29 (m, 7H), 6.92 (s, 1H), 2.63-2.45 (m, 4H), 2.18-2.01 (m, 1H), 1.99-1.82 (m, 1H). MS (ESI+) e/z: 475 [M+l]+.
Example 114
l-[4-(2-Chloro-6-phenyl-pyrrolor3.2-d]pyrimidin-5-yl)-phenyl1-cyclobutylamine
[00452] Step A: { l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (150 mg, 0.316 mmol) was dissolved in ethyl acetate (20 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4 M, 5 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, and the crude product was purified by HPLC separation to afford l-[4-(2-Chloro-6- phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutylamine (70 mg, mg, 59 % yield). Ή NMR (MeOD, 400 MHz), δ ppm: 8.52 (s, 1H), 7.64 (d, J=8.4 Hz, 2H), 7.46 (d, J=8.4 Hz, 2H), 7.41-7.32 (m, 5H), 6.94 (s, 1H), 2.81-2.74 (m, 2H), 2.60-2.53 (m, 2H), 2.24-2.21 (m, 1H), 2.02-1.95 (m, 1H). MS (ESI+) e/z: 375 [M+l]+.
Example 115
j5-r4-n-Amino-cvclobutyl)-phenyl1-6-phenyl-^
[00453] Step A: To the solution of { 1 -[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (200 mg, 0.42 mmol) in dioxane (3 mL) was added the aqueous dimethylamine solution (10 mL) at a sealed vessel. The reaction mixture was stirred at 100 C overnight. LC/MS indicated the reactions worked well. It was diluted with ethyl acetate (80 mL), and washed by water (40 mL) twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product { l-[4-(2-Dimethylamino-6-phenyl- pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester was (-180 mg) used without purification. MS (ESI+) e/z: 484 [M+l]+.
[00454] Step B: { l-[4-(2-Dimethylamino-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-
cyclobutyl}-carbamic acid tert-butyl ester (-180 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The crude product was collected by filtration, and purified by HPLC separation to afford {5-[4-(l-Amino-cyclobutyl)-phenyl]-6- phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-dimethyl-amine (53 mg, 33 %, two steps). 1H NMR (MeOD, 400 MHz), δ ppm: 8.30 (s, 1H), 7.59 (d, J=8.4 Hz, 2H), 7.37-7.28 (m, 7H), 6.63 (s, 3H), 3.21 (s, 6H), 2.79-2.72 (m, 2H), 2.57-2.50 (m, 2H), 2.25-2.18 (m, 1H), 1.98-1.90 ( m, 1H).MS (ESI+) e/z: 384 [M+l]+.
Example 116
1 - { 4- [6-Phenyl-2-( 1 H-pyrazol-4-yl)-pyrrolo [3 ,2-d]pyrimidin-5 -yl] -phenyl I -cyclobutylamine
[00455] Step A: { l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (250 mg, 0.53 mmol), 4-(4,4,5,5-Tetramethyl- [l,3,2]dioxaborolan-2-yl)-lH-pyrazole (153 mg, 0.79 mmol), Pd(dppf)Cl2 (31 mg, 0.04 mmol), and Cs2C03 (343 mg, 1.05 mmol) were dissolved in a mixed solution (dioxane: 15 mL, water: 3 mL). The reaction mixture was heated at 100 C under nitrogen atmosphere for 4 h. Then the solvents were removed by concentration, and the residue was dissolved in ethyl acetate (150 mL). The organic layer was washed by water, dried over sodium sulfate, filtered and concentrated. The crude product ( 1 - {4- [6-Phenyl-2-( 1 H-pyrazol-4-yl)-pyrrolo [3 ,2-d]pyrimidin-5-yl] -phenyl } -cyclobuty 1)- carbamic acid tert-butyl ester (-200 mg) was used in next step without further purification. MS (ESI+) e/z: 507 [M+l]+.
[00456] Step B: (l-{4-[6-Phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[3,2-d]pyrimidin-5-yl]- phenyl}-cycIobutyl)-carbamic acid tert-butyl ester (-200 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, and the crude product was purified by HPLC separation to afford l-{4-[6-Phenyl-2- (lH-pyrazol-4-yl)-pyrrolo [3, 2-d]pyrimidin-5-yl] -phenyl} -cyclobutylamine (80 mg, 37 % yield). 1H NMR (MeOD, 400 MHz), δ ppm: 8.62 (s, 1H), 8.30 (s, 2H), 7.66-7.63 (m, 2H), 7.48-7.32 (m, 7H), 6.94 (s, 1H), 2.81-2.74 (m, 2H), 2.59-2.51 (m, 2H), 2.26-2.22 (m, 1H), 1.99-1.93 (m, 1H). MS
(ESf ) e/z: 407 [M+l]+.
Example
2- { 5-f 4-( 1 -Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyiTolo[3,2-d1pyrimidin-2-ylamino } -ethanol
[00457] Step A: {l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (200 mg, 0.42 mmol) and 2-Amino-ethanol (10 mL) were put into a sealed vessel. And it was stirred at 130 °C for 6 h. The reaction mixture was diluted with ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product (l-{4-[2-(2-Hydroxy-ethylamino)-6-phenyl- pyrrolo[3,2-d]pyrimidin-5-yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (-160 mg) was used without further purification. MS (ESI+) e/z: 500 [M+l .
[00458] Step B: (l-{4-[2-(2-Hydroxy-ethylamino)-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl]- phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (-160 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h. The product was collected by filtration, and the crude product was purified by HPLC separation to afford 2-{5-[4-(l-Amino- cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-ylamino} -ethanol (75 mg, 45% yield, two steps). 1H MR (MeOD, 400 MHz), δ ppm: 8.24 (s, 1H), 7.60 (d, J-8.4 Hz, 2H), 7.39 (d, J=8.4 Hz, 2H), 7.35-7.28 (m, 5H), 6.61 (s, 1H), 3.75 (t, J=5.6 Hz, 2H), 3.54 (t, J=5.6 Hz, 2H), 2.83-2.76 (m, 2H), 2.63-2.56 (m, 2H), 2.27-2.22 (m, 1H), 1.99-1.96 (m, 1H). MS (ESI+) e/z: 400 [M+l]+.
[00459] Example 118 shown in Table 13 can also be made according to the above-described methods.
The following compound can be made according to methods described herein:
Example 119
5-(4-(l-aminocvclobutyl)phenyl)-N-ethyl-6-phenyl-5H-pyrrolo[3,2-d1pyrimidin-2-amine
l-{5-[4-(l-Amino-cvclobutyl)-phenyll-6-phenyl-5H-pyrrolo 3,2-d1pyrimidin-2-yll-py^
[00460] Step A: To the solution of { 1 -[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (250 mg, 0.53 mmol) in MP (30 mL) was added Pyrrolidin-3-ol (183 mg, 2.10 mmol), and K2C03 (363 mg, 2.63 mmol). Then the reaction mixture was stirred at 130 °C overnight. The solvent was removed by concentration, and the resulting residue was diluted with ethyl acetate (100 mL), washed by water (50 mL) twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product ( 1 - {4-[2-(3-Hydroxy-pyrrolidin- 1 -yl)-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl]-phenyl } -cyclobutyl)- carbamic acid tert-butyl ester (-180 mg) was used without further purification. MS (ESI+) e/z: 526 [M+l]+.
[00461] Step B: (l-{4-[2-(3-Hydroxy-pyrrolidin-l-yl)-6-phenyl-pyrrolo[3,2-d]pyrimidin-5- yl]-phenyl}-cyclobutyl)-carbamic acid tert-butyl ester (-180 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The crude product was collected by filtration, and purified by HPLC separation to afford l-{5-[4-(l-Amino-cyclobutyl)- phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-pyrrolidin-3-ol (39 mg, 17 % yield, two steps). 1H NMR (MeOD, 400 MHz), δ ppm: 8.26 (s, 1H), 7.58 (d, J=8.4 Hz, 2H ), 7.33-7.27 (m, 7H), 6.61 (s, 1H), 4.58-4.51 (m, 1H), 3.73-3.69 (m, 3H), 3.62-3.59 (m, 1H), 2.74-2.70 (m, 2H), 2.52-2.45 (m, 2H), 2.20-2.11 (m, 2H), 2.08-2.00 (m, 1H), 1.98-1.92 (m, 1H). MS (ESI+) e/z: 426 [M+l]+.
Example 121
1 - r4-(6-Phenyl-pyrrolo [3 ,2-d1pyrimidin-5 -yP-phenyl] -cyclobutylamine
[00462] Step A: To a solution of { l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (250 mg, 0.53 mmol) in methanol (30 mL) were added Pd/C (200 mg) and KOAc(103 mg, 1.05 mmol). The mixture was stirred at ambient temperature under the atmosphere of hydrogen (balloon) for 12 h.. The catalyst was moved by filtration, washed by methanol. The filtrate was concentrated to give desired product {l-[4-(6- Phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (-100 mg), which was used in de-Boc reaction without purification. MS (ESI+) e/z: 441 [M+l]+.
[00463] Step B: {l-[4-(6-Phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester (-100 mg) was dissolved in ethyl acetate (20 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4 M, 5 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, and the crude product was purified by HPLC separation to afford l-[4-(6-Phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenylj-cyclobutylamine (20 mg, 11 % yield). 1H NMR (MeOD, 400 MHz), δ ppm: 8.90 (s, 1H), 8.66 (s, 1H), 7.62 (d, J=8.8 Hz, 2H), 7.41-7.33 (m, 7H), 7.00 (s, 1H), 2.72-2.65 (m, 2H), 2.45-2.39 (m, 2H), 2.20-2.13 (m, 1H), 1.98-1.82 (m, 1H). MS (ESI+) e/z: 341 [M+l]+.
Example N
5-(4-(l-(tert-butoxycarbonylamino)cvclobutyl)phenyl)-6-phenyl-5H-pyrrolo[3,2-d1pyrimidine-2- carboxylic acid
[00464] Step A: To the solution of { l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (450 mg, 0.95 mmol) in a mixed solution (DMF: 15 mL, MeOH: 5 mL) were added Pd(OAc)2 (21 mg, 0.10 mmol), dppf (53 mg, 0.10 mmol) and TEA (lmL). The reaction mixture was stirred at 80 C under the atmosphere of carbon monoxide (50 psi) overnight.. It was concentrated to remove the solvent, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product purified by column separation (EtOAc:Hexane =1 :3) to afford 5-[4-(l -tert-Butoxycarbonylamino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2- d]pyrimidine-2-carboxylic acid methyl ester (250 mg, 53% yield). MS (ESI+) e/z: 499[M+1]+.
[00465] Step B: To a solution of 5-[4-(l-tert-Butoxycarbonylamino-cyclobutyl)-phenyl]-6- phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxylic acid methyl ester (250 mg, 0.502 mmol) in THF (10 mL) was added a solution of lithium hydroxide hydrate (84 mg, 2.00 mmol) in H20 (10 mL). The reaction mixture was stirred at ambient temperature for 3 h.. Then the solution was acidified by IN HC1 solution to pH 7-8. The solution was concentrated to give crude product 5-[4-(l-tert- Butoxycarbonylamino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxylic acid (-220 mg), which was used without further purification. MS (ESI+) e/z: 485[M+1]+.
Example 122
5-[4-(l-Amino-cvclobutylVphenyll-6-phenyl-5H-pyrrolo[3.2-d1pyrimidine-2-carboxylic acid
[00466] Step A: 5-[4-(l-tert-Butoxycarbonylamino-cyclobutyl)-phenyl]-6-phenyl-5H- pyrrolo[3,2-d]pyrimidine-2-carboxylic acid (-220 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, and the crude product was purified by HPLC separation to afford 5-[4-(l-Amino-cyclobutyl)- phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxylic acid (150 mg, 78 %, two steps). Ή NMR (MeOD, 400 MHz), δ ppm: 8.17 (s, IH), 7.66-7.59 (m, 2H), 7.49-7.34 (m, 7H), 7.08 (s, IH), 2.89-2.75 (m, 2H), 2.72-2.55 (m, 2H), 2.31-2.20 (m, IH), 2.05-1.95 (m, IH). MS (ESI+) e/z: 385 [M+l]+.
Example 123
5-(4-(l-aminocyclobutynphenyl)-N-methyl-6-phenyl-5H-pyrrolor3,2-d1pyrimidine-2-carboxamide
[00467] Step A: DIPEA (0.5 mL) was added a solution of 5-(4-(l-(tert-butoxy-
carbonylamino)cyclobutyl)phenyl)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxylic acid (170 mg, 0.35 mmol), methanaminium chloride (118 mg, 1.75 mmol) and HATU (700 mg, 1.8 mmol) in DMF (5 mL). The mixture was stirred at 10°C for overnight. LC-MS showed the reaction worked well. The reaction mixture was diluted with Ethyl acetate (30 mL), washed with Na2C03 (20 mL, a.q.) and brine (2 x 20 mL). The organic phase was concentrated to dryness to give the product to give the product tert-butyl l-(4-(2-(methylcarbamoyl)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-5- yl)phenyl)cyclobutylcarbamate (75 mg, 43% yield). LC/MS (ESI+): 498.2 [M+H]+
[00468] Step B: tert-butyl l-(4-(2-(methylcarbamoyl)-6-phenyl-5H-pyrrolo[3,2- d]pyrimidin-5-yl)phenyl)cyclobutylcarbamate (75 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 °C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, and the crude product was purified by preparative HPLC to afford 5-(4-(l- aminocyclobutyl)phenyl)-N-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxamide. 1H NMR (MeOD, 400 MHz), δ ppm: 8.25 (s, 1H), 7.62 (d, 2H, J=6.8 Hz), 7.41 (d, 2H, J=6.8 Hz), 7.36-7.31 (m, 5H), 6.63 (s, 1H), 3.18 (s, 3H), 2.82-2.79 (m, 2H), 2.66-2.61 (m, 2H), 2.24-2.22(m, 1H), 1.96-1.94 (m, 1H). MS (ESI+) e/z: 398.2 [M+l]+
Example 124
1 -(5-(4-( 1 -aminocyclobutyl)phenyl)-6-phenyl-5H-pyrrolo|"3,2-d1pyrimidin-2-yl)pyrrolidin-2-one
[00469] Step A: The compound tert-butyl l-(4-(2-chloro-6-phenyl-5H-pyrrolo[3,2- d]pyrimidin-5-yl)phenyl)cyclobutylcarbamate (300 mg, 0.63 mmol), Pyrrolidin-2-one (161 mg, 1.89 mmol), Pd2(dba)3 (57.6 mg, 0.0063 mmol), Xantphos (36 mg, 0.0.63 mmol) and Cs2C03 (616 mg, 1.89 mmol) were dissolved in dioxane (20 mL). The reaction mixture was stirred at 100 °C under nitrogen atmosphere overnight. Then the solvent was removed by concentration, and the residue was dissolved in ethyl acetate (30 mL). The organic layer was washed by water dried over sodium sulfate, filtered and concentrated to give tert-butyl l-(4-(2-(2-oxopyrrolidin-l-yl)-6-phenyl- 5H-pyrrolo[3,2-d]pyrimidin-5-yl)phenyl)cyclobutylcarbamate (186 mg, yield: 56%). LC/MS (ESI+): 524.2 [M+H]+
[00470] Step B: tert-butyl l-(4-(2-(2-oxopyrrolidin-l-yl)-6-phenyl-5H-pyrrolo[3,2- d]pyrimidin-5-yl)phenyl)cyclobutylcarbamate(186 mg) was dissolved in ethyl acetate (30 mL), the solution was cooled to 0 C and the solution of HC1 in ethyl acetate (4 M, 8 mL) was added slowly. The mixture was stirred at ambient temperature for 4 h.. The product was collected by filtration, and the crude product was purified by preparative HPLC to afford title compound: l-(5-(4-(l- aminocyclobutyl)phenyl)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl)pyrrolidin-2-one (54 mg). 1H NMR (MeOD, 400 MHz), δ ppm: 8.56 (s, IH), 7.52 (d, 2H, J= 6.8 Hz), 7.46 (m, 2H, J= 6.8 Hz), 7.41-7.32 (m, 5H), 6.93 (s, IH), 4.21-4.17 (t, 2H), 2.82-2.79 (m, 2H), 2.7 1-2.68 (t, 2H), 2.64-2.60 (m, 2H), 2.24-2.22(m, IH), 2.21-2.18 (m, 2H),2.06-1.96 (m, IH). LC/MS (ESI+): 424.3 [M+H]+
Exam le 125
l-f4-(2-Methoxy-6-phenyl-pyrrolo 3,2-d1pyrimidin-5-yl)-phenyll-cyclobutylamine
[00471] Step A: To the solution of { l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (190 mg, 0.4 mmol) and NaOMe ( 432 mg, 8 mmol) in MeOH (2 mL) was heated at 120°C by MW for 50 min.. The reaction mixture was concentrated to dryness. The residue was diluted with EA (20 mL), wished with brine (2*10 mL). The organic phase was concentrated to dryness to give the product { l-[4-(2-Methoxy-6-phenyl- pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutyl}-carbamic acid methyl ester (137 mg, 85% yield). LC/MS (ESI+): 484.2 [M+H]+
[00472] Step B: LiOH (178 mg, 7.4 mmol) was added a solution of { l-[4-(2-Methoxy-6- phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutyl}-carbamic acid methyl ester (130 mg, 0.304 mmol) in a mixture solution (dioxane:10 mL, H20: 5 mL). The mixture was heated at 120°C for overnight in a 30 mL of sealed rude.. The cooled mixture was diluted with Ethyl acetate (20 mL) and washed brine (2* 10 mL). The organic phase was concentrated to dryness, which was purified by preparative HPLC to give l-[4-(2-Methoxy-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)- phenyl]-cyclobutylamine (26 mg, 19% yield:). 1H NMR (MeOD, 400 MHz), δ ppm: 8.41 (s, IH), 7.62 (d, 2H, J=8.4 Hz), 7.42 (m, 2H, J=8.4 Hz), 7.39-7.30 (m, 5H), 6.80 (s, IH), 4.04 (s, 3H), 2.80- 2.73 (m, 2H), 2.59-2.52 (m, 2H), 2.47-2.22(m, IH), 1.97-1.94 (m, IH). MS (ESI+): 371.2 [M+H]+.
Examples 126 and 127
1-(4-(5-methyl-6-phenyl-7 - -pyrrolo[2.3-dlPyrimidin-7- 1-(4-(6-methyl-5-phenyl-7H-pyrro[o[2.3--yi yhphenyltoyclobutanamine pyrimidin-7-yl)phenyl)cyclobutanamine
[00473] Step A: To a solution of 4-Chloro-5-iodo-pyrimidine (200 mg, 0.83 mmol) and [1-
(4-Amino-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (218 mg, 0.83 mmol) in dioxane (10 mL) was added DIPEA (214 mg, 1.66 mmol). The mixture was heated at 100°C for 18 h. After removal of solvent by concentration, the residue was purified by silica gel chromatography eluted with Hexane Ethyl acetate = 10:1 to give { l-[4-(5-Iodo-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}- carbamic acid tert-butyl ester. (230 mg, 59%yield). LCMS (ESI+): 466.8 [M+l]+
[00474] Step B: The { l-[4-(5-Iodo-pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (200 mg, 0.42 mmol) and Prop-l-ynyl-benzene (73 mg, 0.63 mmol) were dissolved in DMF (5 mL), and then LiCl (17.6 mg, 0.42 mmol), Pd(OAc)2 (9.4 mg, 0.04 mmol) and KOAc (82 mg, 0.84 mmol) were added. The resulting mixture was charged with N2 thrice and irradiated at 120°C by MW for 1 h. TLC showed the starting material was consumed completely. The mixture was diluted with water (20 mL), extracted with EtOAc (3*20). The combined extracts were concentrated to give mixture cantaining {l-[4-(5-Methyl-6-phenyl-pyrrolo-[2,3-d]pyrimidin- 7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester and { l-[4-(6-Methyl-5-phenyl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester. (183 mg, 93%yield).
[00475] Step C: To the mixture of { 1 -[4-(5-Methyl-6-phenyl-pyrrolo[2,3-d]pyrimidin-7-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester and { l-[4-(6-methyl-5-phenyl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (183 mg, 0.4 mmol) in ethyl acetate (20 mL) was added HCl/EtOAc (10 mL, 2M) at 0°C. The resulting solution was stirred for 2 hrs. After removal of solvent by concentration, the residue was purified by prep-HPLC to give l-[4- (5-Methyl-6-phenyl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutylamine (12 mg, yield: 8.4%). Ή MR (MeOD, 400 MHz), δ ppm: 9.07 (s, 1H), 8.74 (s, lH), 7.54 (d, 2H, J=8.4 Hz), 7.40-7.36 (m, 2H), 7.35-7.33 (m, 3H), 7.31-7.28 (m, 2H), 2.80-2.73 (m, 2H), 2.53-2.46 (m, 2H), 2.40 (s, 3H),
2.23-2.19(m, 1H), 1.96-1.91 (m, 1H). and l-[4-(6-Methyl-5-phenyl-pyrrolo[2,3-d]pyrimidin-7-yl)- phenyl]-cyclobutylamine (14 mg, yield: 9.8%): 1H NMR (MeOD, 400 MHz), δ ppm: 8.93 (s, 1H), 8.67 (s, 1H), 7.73 (d, 2H, J=8.4 Hz), 7.59-7.51 (m, 6H), 7.41-7.37 (m, 1H), 2.80-2.73 (m, 2H), 2.53-2.46 (m, 2H), 2.40 (s, 3H), 2.23-2.19(m, 1H), 1.96-1.91 (m, 1H).
Example 128
l-[4-(5-Ethyl-6-pyridin-2-yl-pyiTolo[23-d]pyrimidin-7-yl)-phenyl1-cyclobutylamine
[00476] Step A: To a solution of {l-[4-(2-Chloro-6-pyridin-2-yl-pyrrolo [2,3-d]pyrimidin-7- yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (240 mg, 0.5 mmol) in DCM (10 mL) was added NIS (110 mg, 0.5 mmol). The mixture was stirred at r.t. for overnight. The reaction mixture was filtered, and the solvent was concentrated under reduced pressure to get crude product, which was purified by flash chromatography (petroleum ether/ethyl acetate= 100/1-1/1) to yield the desired product { l-[4-(2-Chloro-5-iodo-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (210mg, 69%). MS (ESI) m/z: 602.2[M+1]+ ,624.1 [M+Na]+.
[00477] Step B: To a solution of { l-[4-(2-Chloro-5-iodo-6-pyridin-2-yl-pyrrolo[2,3-d] pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (300 mg, 0.5 mmol), potassium vinyltrifluoroborate (67 mg, 0.5 mmol), and Pd(dppf)Cl2(35 mg, 0.05 mmol) in n-PrOH (20 mL) was added TEA (0.5 mL). The mixture degassed and charged with N2 for three times. The mixture was stirred at 90°C for overnight. The reaction mixture was concentrated under reduced pressure and purified by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate=100/l~l/l), to give the title product { l-[4-(2-Chloro-6-pyridin-2-yl-5-vinyl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (30mg, 12%). MS (ESI) m/z: 502.2[M+1]+ ,524.4[M+Na]+.
[00478] Step C: To a solution of {l-[4-(2-Chloro-6-pyridin-2-yl-5-vinyl-pyrrolo [2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (60 mg, 0.12 mmol) in MeOH(30 mL) was added Pd/C (50 mg). The mixture was stirred under H2 atmosphere at reflux for overnight. The reaction mixture was filtered, and the solvent was concentrated under reduced pressure to give the product {l-[4-(5-Ethyl-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-
cyclobutyl}-carbamic acid tert-butyl ester (25 mg, 45%), which was used to next step without furthermore purification. MS (ESI) m/z : 470.3[M+1]+.
[00479] Step D: To a solution of HC1 in EtOAc (30 mL, 2M) was added { l-[4-(5-Ethyl-6- pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (25 mg). The mixture was stirred at r.t. for lh. The reaction mixture was filtered to give the crude product, which was purified by preparative HPLC to yield l-[4-(5-Ethyl-6-pyridin-2-yl-pyrrolo[2,3- d]pyrimidin-7-yl)-phenyl]-cyclobutylamine (12mg, 63% yield). 'HNMR: MeOD 400MHz δ 9.1 l(s, 1H), 8.93(s, 1H), 8.64~8.62(t, 1H, J=8), 7.62~7.60(t, 1H, J=8), 7.41~7.39(m, 2H), 7.26~7.24(m, 2H), 7.17~7.15(m, 2H), 2.30~2.94(m, 2H), 2.58~2.54(m, 2H), 2.20-2.16(m, 2H), 2.10~1.98(m, 1H), 1.96~1.90(m, lH),1.33~1.24(m,3H). MS (ESI) m/z : 370.3[M+1]+
Example O
{ l-[4-(2-Chloro-5-methyl-6-phenyl-pyrrolo[2,3-d1pyrimidin-7-yl)-phenyl1-cvclobutvU-carbamic acid tert-butyl ester
[00480] Step A: To a solution of 2,4-Dichloro-5-iodo-pyrimidine (7.7 g, 28 mmol) and [1-
(4-Amino-phenyl)-cyclobutyl]-carbamic acid tert-butyl ester (6.2 g, 24 mmol) in dioxane (150mL) was added K2C03 (4.9 g, 36 mol). The mixture was stirred at 100°C overnight. The precipitate was filtered off, the solvent was concentrated under reduced pressure to give crude product, which was purified by silica gel chromatography to give the desired product { l-[4-(2-Chloro-5-iodo- pyrimidin-4-ylamino)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (2.1g, 15% yield). MS (ESI) m/z : 501.1[M+1]+ ,523.0[M+Na]+.
[00481] Step B: To a solution of { l-[4-(2-Chloro-5-iodo-pyrimidin-4-ylamino)-phenyl] - cyclobutyl}-carbamic acid tert-butyl ester (200 mg, 0.4 mmol), Pd(OAc)2(9 mg, 0.04 mmol), PPh3(21 mg, 0.08 mmol), LiCl(17 mg, 0.4 mmol) and K2C03 (276 mg, 2 mmol) in DMF (1 mL) was added Prop-l-ynyl-benzene (0.15mL). The mixture was degassed and charged with N2 for three times. The reaction mixture was stirred at 120°C for 30min by microwave irradiation. The reaction mixture was concentrated, and pardoned with water and DCM. The organic layer was concentrated and purified by silica gel chromatography (eluting with petroleum ether/ethyl acetate
=20/1-0/1), to give the title product { l-[4-(2-Chloro-5-methyl-6-phenyl-pyrrolo[2,3-d]pyrimidin-7- yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (19mg,10% yield). MS (ESI) m/z :
489.2[M+1]+ .
Example 129
l-(4-r5-Methyl-6-phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[2,3-dlpyrimidin-7-yl1-phenvn- cvclobutylamine
[0003] Step A: To a solution of { l-[4-(2-Chloro-5-methyl-6-phenyl-pyrrolo[2,3-d] pyrimidin-7-yl)-phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (150 mg, 0.31 mmol), 4- (4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (120 mg, 0.62 mmol), and Cs2C03 (200 mg, 0.62 mmol) in DME/H20 (5/1, 30 mL) was added Pd(dppf)Cl2 (22 mg, 0.03 mmol). The mixture was degassed and charged with N2 for three times. The mixture was stirred at 100 C for overnight. The reaction mixture was filtrated, the filtrates was concentrated under reduced pressure and partioned with water and DCM. The organic layer was combined and removed by rotary evaporation to give crude product which was purified by silica gel chromatography to give the title product ( 1 - { 4- [5 -Methyl-6-phenyl-2-( 1 H-pyrazol-4-yl)-pyrrolo [2,3-d]pyrimidin-7-yl] -phenyl } - cyclobutyl)-carbamic acid tert-butyl ester (100 mg ). MS (ESI) m/z : 521.3[M+1]+ ,543.1[M+Na]+ .
[0004] Step B: To a solution of HC1 in EtOAc (30 mL, 2M) was added (l-{4-[5-Methyl-6- phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}-cyclobutyl)-carbamic acid tert- butyl ester (lOOmg). The mixture was stirred at r.t. for lh. The reaction mixture was filtered to yield the title product l-{4-[5-Methyl-6-phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[2,3-d]pyrimidin-7-yl]- phenyl}-cyclobutylamine (65.8 mg, 82% yield^HNMR: MeOD 400MHz δ 9.33(s, 1H), 8.39(s, 2H), 7.64~7.62(m, 2H), 7.51~7.49(m, 2H), 7.40~7.39(m, 3H), 7.35~7.33(m, 2H), 2.82~2.76(m, 2H), 2.66~2.59(m, 2H), 2.47(s,3H), 2.10~1.98(m, 1H), 2.01~1.97(m, 1H). MS (ESI) m/z : 421.3[M+1]+
[00482] Examples 130 and 131 shown in Table 13 can also be made according to the above- described methods.
Example 132
l-(4-(2-phenyl-lH-pyrrolo[2,3-blpyridin-3-yl)phenyl)cyclobutanamine
[00483] Step A: To a solution of 3-bromopyridin-2-amine (200 mg, 1.16 mmol) in DMF (3 mL) were added tert-butyl l-(4-(phenylethynyl)phenyl)cyclobutylcarbamate (802 mg, 2.31 mmol), Pd(OAc)2 (26 mg, 0.12 mmol), LiCl (50 mg, 1.16 mmol) and KOAc (228 mg, 2.31 mmol). The
reaction mixture was stirred at 130 °C under microwave for 3 h. LC/MS showed about 18% of desired products (3 and 4). The reaction mixture was diluted with ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude products were purified by pre-TLC (e luted by: ethyl acetate: hexane = 1 :1), which gave two desired products tert-butyl l-(4-(2-phenyl-lH-pyrrolo[2,3-b]pyridin-3- yl)phenyl)cyclobutylcarbamate and tert-butyl l-(4-(3-phenyl-lH-pyrrolo[2,3-b]pyridin-2- yl)phenyl)cyclobutylcarbamate (130 mg, 25% Yield). MS (ESI+) e/z: 440.2 [M+l] +.
[00484] Step B: To a solution of tert-butyl l-(4-(2-phenyl-lH-pyrrolo[2,3-b]pyridin-3- yl)phenyl)cyclobutylcarbamate and tert-butyl l-(4-(3-phenyl-lH-pyrrolo[2,3-b]pyridin-2- yl)phenyl)cyclobutylcarbamate (130 mg, 0.30 mmol) in ethyl acetate (20 mL) was added a solution of HC1 in ethyl acetate (5 mL) at 0 °C. Then it was allowed to reach ambient temperature for 3 h. The reaction mixture was concentrated to remove the solvent. The crude product was purified by HPLC separation, which gave l-(4-(2-phenyl-lH-pyrrolo[2,3-b]pyridin-3- yl)phenyl)cyclobutanamine (20 mg, 20% yield). Ή NMR (MeOD, 400 MHz), δ ppm: 8.63 (d, J=6.8 Hz, IH), 8.47 (d, J=4.8 Hz, IH), 7.68-7.60 (m, 7H), 7.56-7.40 (m, 3H), 2.86-2.79 (m, 2H), 2.68-2.61 (m, 2H), 2.30-2.25 (m, IH), 2.03-1.94 (m, IH). MS (ESI+) e/z: 340.2 [M+l] +, and l-(4- (3-phenyl-lH-pyrrolo[2,3-b]pyridin-2-yl)phenyl)cyclobutanamine (26 mg, 26% yield ). 1H NMR (MeOD, 400 MHz), δ ppm: 8.07 (d, J=8.0 Hz, IH), 7.55-7.49 (m, 7H), 7.37-7.35 (m, 3H), 7.25- 7.22 (m, IH), 2.86-2.74 (m, 2H), 2.68-2.57 (m, 2H), 2.31-2.20 (m, IH), 2.06-1.97 (m, IH). MS (ESI+) e/z: 340.2 [M+l] +.
[00485] Example 133 shown in Table 14 can also be made according to the above-described methods.
Table 14
Example 134
l-(4-(l-methyl-2-phenyl-lH-pyrrolo[2 -blpyridin-3-yDphenyl)cvclobutanamine
[00486] Step A: To a solution of 3-methylpyridin-2-amine (5.0 g, 46.24 mmol) in pyridine (50 mL) was slowly added benzoyl chloride (6.50, 46.24 mmol) at 0 °C. After addition, the reaction mixture was stirred at ambient temperature overnight. Then the solvent was removed by concentration in vacuo, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was concentrated. The obtained crude product was dissolved in methanol (50 mL), and an aqueous sodium hydroxide solution (2 N, 30 mL) was added. The mixture was heated at 80 °C for 2 h. Then the solvents were concentrated to remain about 20% volume, which then diluted with ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product N-(3-methylpyridin-2-yl)benzamide (6.2 g, 63% yield) was pure enough to be used in next step without further purification. MS (ESI+) e/z: 213.1 [M+l] +.
[00487] Step B: To a solution of N-(3-methylpyridin-2-yl)benzamide (2.5 g, 11.78 mmol) in
THF was slowly added w-BuLi (14.1 mL, 35.25 mmol) at -78 °C. After addition, the reaction mixture was stirred at ambient temperature for 2 h. The solvent was removed by concentration in vacuo. The residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel column separation to give 2-phenyl-lH-pyrrolo[2,3-b]pyridine (2.0 g, 87% yield). MS (ESI+) e/z: 195.1 [M+1] +.
[00488] Step C: A solution of iodine (2.67 g, 10.56 mmol) in DMF (5 mL) was slowly added into a solution of 2-phenyl-lH-pyrrolo[2,3-b]pyridine (2.0 g, 10.30 mmol), and potassium hydroxide (82%, 2.11 g, 30.90 mmol) in DMF (35 mL). The reaction mixture was stirred at ambient temperature for 1 h. Then, iodomethane (2.19 g, 15.45 mmol) was slowly added into the mixture, and it was stirred for another 1 h. Then solvent was removed by concentration in vacuo.
The residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product 3-iodo-l-methyl-2-phenyl- lH-pyrrolo[2,3-b]pyridine (3.0 g, 87% yield) was pure enough to be used in next step without further purification. MS (ESI+) e/z: 335.0[M+1] +.
[00489] Step D: To a mixed solution (Dioxane / H20 = 12 / 3 mL) of 3-iodo- 1 -methyl-2- phenyl-lH-pyrrolo[2,3-b]pyridine (400 mg, 1.20 mmol) were added tert-butyl l-(4-(4 ,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)cyclobutylcarbamate (536 mg, 1.44 mmol),
Pd(dppf)Cl2 (70 mg, 0.10 mmol), and Cs2C03 (780 mg, 2.29 mmol). The reaction mixture was stirred at 80 °C under the atmosphere of nitrogen for 4 h. The solvents were removed by
concentration in vacuo, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The obtained crude product was used in de-Boc reaction directly. MS (ESI+) e/z: 454.2[M+1] +.
[00490] Step E: To a solution of ter/-butyl l-(4-(l-methyl-2-phenyl-lH-pyrrolo[2,3- b]pyridin-3-yl)phenyl)cyclobutylcarbamate (350 mg, 0.64 mmol) in ethyl acetate (20 mL) was added a solution of HC1 in ethyl acetate (5 mL) at 0 °C. Then it was allowed to reach ambient temperature for 3 h. The reaction mixture was concentrated to remove the solvent. The crude product was purified by HPLC separation, which gave the desired product l-(4-(l-methyl-2- phenyl-lH-pyrrolo[2,3-b]pyridin-3-yl)phenyl)cyclobutanamine (80mg, 19% yiled, two steps). Ή NMR (MeOD, 400 MHz), δ ppm: 8.68 (d, J=7.6 Hz, 1H), 8.54 (d, J=5.6 Hz, 1H), 7.67-7.61 (m, 1H), 7.52-7.43 (m, 9H), 3.86 (s, 3H), 2.81-2.73 (m, 2H), 2.66-2.56 (m, 2H), 2.22-2.21 (m, 1H), 1.99-1.91 (m, 1H). MS (ESI+) e/z: 354.2[M+1] +.
Example 135
l-(4-(6-phenyl-5H-pyrrolo 3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine
[00491] Step A: To a solution of 2,4-dichloro-6-methyl-5-nitropyrimidine (7 g, 34 mmol) and CH3COOK (9.8 g, 100 mmol) in MeOH (150 mL) was added Pd C (3 g). The mixture was stirred at H2 atmosphere for overnight. The reaction mixture was filtered, and the solvent was concentrated under reduced pressure to give the product 4-methylpyrimidin-5 -amine (1.5 g, 43%), which was used to next step without furthermore purification. 'HNMR: MeOD 400MHz δ 8.28(s,
1H), 8.04(s, 1H), 2.3 l(s, 3H).
[00492] Step B: 4-methylpyrimidin-5-amine (1.09 g, 10 mmol) was dissolved in dry THF
(150 mL) under nitrogen. The solution was cooled to -78°C and a solution of n-BuLi was added dropwise. The solution was kept at the same temperature for 30min and then warm to r.t., stirred for 2h. Then cooled to -78°C, ethyl benzoate (1.5 g, 10 mmol) in THF (30 mL) was added, and the resulting mixture was stirred at -78°C for 30min, and was warmed to r.t. for lh. The reaction mixture was quenched with drop some water, and neutralized with NaHC03. EtOAc and water were added, portioned. The organic layer was concentrated and purified by by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate =100/1-0/1), to give the title product 6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (0.8 g, 41%). MS (ESI) m/z :196.1 [M+1]+.
[00493] Step C: To a solution of 6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (700 mg, 3.5 mmol) in DMF (30 mL) was added KOH (703 mg, 7.2 mmol). The mixture was stirred at r.t. for 15min, then a solution of I2 in DMF was added dropwise and the mixture was stirred for another 15min at the same temperature. To the reaction mixture was added Na2S03 solution, and extracted with EtOAc. The organic layer was washed with brine and concentrated under reduced pressure. The residue was purified by sical gel chromatography (eluting with PE/EA= 1/0-0/1) to give the product 7-iodo-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (400 mg, 35%). MS (ESI) m/z : 321.8[M+1]+.
[00494] Step D: To a solution of 7-iodo-6-phenyl-5H-pyrrolo[3,2-d] pyrimidin.? (320 mg,
1.0 mmol), tert-butyl l-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl) phenyl) cyclobutylcarbamate (560mg, 1.5mmol), and Cs2C03 (652 mg, 2.0 mmol) in CH3CN/H20 (10/1 , 10ml) was added Pd(dppf)Cl2 (70 mg, O.lmmol). The mixture was irradiated by microwave at 80 C for lOmin.The reaction mixture was filtrated, the filtrates was washed was water and the organic layer was combined and removed by rotary evaporation to give crude product tert-butyl 1- (4-(6-phenyl-5H-pyrrolo [3,2-d]pyrimidin-7-yl)phenyl) cyclobutylcarbamate, which was to do next step without further purification. MS (ESI) m/z : 441.1 [M+l]+.
[00495] Step E: To a solution of HC1 in EtOAc (30 mL, 2 M) was added tert-butyl l-(4-(6- phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (200 mg). The mixture was stirred at r.t. for 4h. The reaction mixture was filtered to give the crude product, which was purified by pre-HPLC to yield the title product l-(4-(6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7- yl)phenyl)cyclobutanamine (7.4 mg). 1HNMR: MeOD 400MHz δ 8.91(s, 1H), 8.78(s, 1H), 8.35~8.33(d, 1H, J=8), 8.23~8.21(d, 1H, J=8), 7.74~7.72(m, 2H), 7.59~7.57(m, 2H), 7.62~7.56(m, 2H), 7.54~7.46(m, 4H), 7.45~7.35(m, 2H), 2.80~2.76(m, 2H), 2.68~2.64(m, 2H), 2.24~2.20(m, 1H), 2.00-1.96(m, 1H).
Example 136
l-[4-(5-Methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-phenyl]- cyclobutylamine
[00496] Step A: To a solution of 2,4-dichloro-6-methyl-5-nitropyrimidine (7 g, 34 mmol) and CH3COOK (9.8 g, 100 mmol) in MeOH (150 mL) was added Pd/C (3 g). The mixture was stirred at H2 atmosphere for overnight.The reaction mixture was filtered, and the solvent was concentrated under reduced pressure to give the product 4-methylpyrimidin-5-amine (1.5 g 43%), which was used to next step without furthermore purification. 'HNMR: MeOD 400MHz δ 8.28(s, 1H), 8.04(s, 1H), 2.3 l(s, 3H).
[00497] Step B: 4-methylpyrimidin-5-amine (1.09 g, 10 mmol) was dissolved in dry THF
(150 mL) under nitrogen. The solution was cooled to -78°C and a solution of n-BuLi was added dropwise. The solution was kept at the same temperature for 30min and then warm to r.t., stirred for 2h. Then cooled to -78°C, ethyl benzoate (1.5 g, 10 mmol) in THF (30 mL) was added, and the resulting mixture was stirred at -78°C for 30min, and was warmed to r.t. for lh. The reaction mixture was quenched with drop some water, and neutralized with NaHC03. EtOAc and water were added, portioned. The organic layer was concentrated and purified by by flush column chromatography on silica gel (eluting with petroleum ether/ethyl acetate =100/1-0/1), to give the title product 6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (0.8 g, 41%). MS (ESI) m/z : 196.1[M+1]+.
[00498] Step C: To a solution of 6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (700 mg, 3.5 mmol) in DMF (30 mL) was added KOH (703 mg, 7.2 mmol). The mixture was stirred at r.t. for 15min, then a solution of I2 in DMF was added dropwise and the mixture was stirred for another 15 min at the same temperature. To the reaction mixture was added Na2S03 solution, and extracted with EtOAc. The organic layer was washed with brine and concentrated under reduced pressure. The residue was purified by sical gel chromatography (eluting with petroleum ether/ethyl acetate =1/0-0/1) to give the product 7-iodo-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (400 mg, 35%). MS (ESI) m/z : 321.8[M+1]+.
[00499] Step D: To a solution of 7-Iodo-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (170 mg,
0.81 mmol) and K2C03 (225 mg, 1.62 mmol) in DMF (5 mL) was added Mel (116 mg, 0.81mmol) while keeping an ice-water bath. The mixture was stirred at for 3h. To the reaction mixture was
added water and extracted with EtOAc. The organic layer was washed with brine and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with petroleum ether/ethyl acetate =1/0-0/1) to give the product 7-Iodo-5-methyl-6-phenyl-5H- pyrrolo[3,2-d]pyrimidine (70 mg, 40%). 'HNMR: CDC13 400MHz 69 .l l(s, 1H), 8.78(s, 1H), 7.61~7.57(m,3H), 7.52~7.48(m,2H), 3.80(s, 3H).
[00500] Step E: To a solution of 7-Iodo-5-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine
(150mg, 0.45mmol), {l-[4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]- cyclobutyl }- carbamic acid tert-butyl ester (167 mg, 0.45 mmol), and Cs2C03 (292 mg, 0.9 mmol) in CH3CN/H20 (5/1, 3mL) was added Pd(dppf)Cl2 (32 mg, 0.045 mmol). The mixture was irradiated by microwave at lOOC for 20 min.The reaction mixture was filtrated, the filtrates was washed was water and the organic layer was combined and removed by rotary evaporation to give crude product which was to do next step without further purification. MS (ESI) m/z : 455.1[M+1]+.
[00501] Step F: To a solution of HC1 in EtOAc (30 mL, 2M) was added {l-[4-(5-Methyl-6- phenyl-5H^yrrolo[3,2-dJpyrimidin-7-yl)-phenylJ-cyclobutyl}-carbamic acid tert-butyl ester (200 mg, crude). The mixture was stirred at r.t. for lh. The reaction mixture was filtered to give the crude product, which was purified by pre-HPLC to yield the title product l-[4-(5-Methyl-6-phenyl- 5H-pyrrolo[3,2-d]pyrimidin-7-yl)-phenyl]- cyclobutylamine (32 mg). 'HNMR: MeOD 400MHz δ 9.07(s, 1H), 8.89(s, 1H), 7.54~7.51(m, 5H), 7.45~7.38(m, 4H), 3.80(s, 3H), 2.80~2.76(m, 2H), 2.58~2.54(m, 2H), 2.24~2.20(m, 1H), 2.00-1.96(m, 1H).
Example 137
l-(4-(2-chloro-5-methyl-6-phenyl-5H-pyrrolo 3.2-d1pyrimidin-7-yl)phenyl)cyclobutanamine
[00502] Step A: To a solution of 5-bromo-2,4-dichloropyrimidine (10.00 g, 43.88 mmol ) in
TEA (100 mL) were added ethynylbenzene (4.48 g, 43.88 mmol), Pd(PPh3)2Cl2 (1.00 g, 1.42 mmol), and Cul (1.67 g, 8.76 mmol). The reaction mixture was stirred at 100 °C under the atmosphere of nitrogen for 2 h. After cooling, the solvent was removed by concentration in vacuo, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered, and concentrated. The crude product was purified by
column separation (eluted by: hexane / ethyl acetate = 20 : 1), which gave the desired product 5- bromo-2-chloro-4-(phenylethynyl)pyrimidine (7.10 g, 55% Yield). MS (ESI+) e/z: 292.9[M+1]+.
[00503] Step B: To a solution of 5-bromo-2-chloro-4-(phenylethynyl)pyrimidine (3.10 g,
10.56 mmol) in dioxane (80 mL) were added tert-butyl carbamate (1.61 g, 13.73 mmol), Pd2(dba)3 (484 mg, 0.53 mmol), X-phos (503 mg, 1.06 mmol), and Cs2C03 (6.88 g, 21.12 mmol). The reaction mixture was stirred at 100 °C under the atmosphere of nitrogen for 8 h. The solvent was removed by concentration, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product tert-butyl 2-chloro-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-5-carboxylate was used in next step without further purification. MS (ESI+) e/z: 330.1, 332.1 [M+l]+.
[00504] Step C: To a solution of the crude product tert-butyl 2-chloro-6-phenyl-5H- pyrrolo[3,2-d]pyrimidine-5-carboxylate (3.0 g) in ethyl acetate (60 mL) was added a solution of HC1 in ethyl acetate (10 mL). The reaction mixture was heated to reflux overnight. After cooling, the resulting precipitate was collected by filtration, washed by ethyl acetate, dried in vacuo, which gave the desired product 2-chloro-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (1.7 g, 70% yield, two steps). MS (ESI+) e/z: 230.0, 232.0[M+1]+.
[00505] Step D: A solution of I2 (3.99 g, 15.81 mmol) in DMF (10 mL) was slowly added to a solution of 2-chloro-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (3.30 g, 14.37 mmol) and KOH (82%, 1.97 g, 28.74 mmol) in DMF (80 mL) at ambient temperature. The reaction mixture was stirred for 1 h. Then, iodomethane (3.06 g, 21.65 mmol) was slowly added into the mixture, and it was stirred for another 1 h. The solvent was removed by concentration in vacuo, and the residue was dissolved in ethyl acetate, washed with water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product 2-chloro-7-iodo-5-methyl-6-phenyl-5H- pyrrolo[3,2-d]pyrimidine (3.00 g, 89% yield) was pure enough to be used in next reaction. MS (ESI+) e/z: 369.9, 371.9[M+1]+.
[00506] Step E: To a mixed solution (dioxane / H20 = 25 / 5 mL) of 2-chloro-7-iodo-5- methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine (2.50 g, 6.78 mmol) were added tert-butyl l-(4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)cyclobutylcarbamate (3.82 g, 10.17 mmol), Pd(dppf)Cl2 (496 mg, 0.68 mmol), and Cs2C03 (4.42 g, 13.56 mmol). The reaction mixture was stirred at 100 °C for 3 h. Then the solvents were removed by concentration in vacuo, and the residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude produce was purified by silica gel column separation (eluted by: hexane / ethyl acetate = 1 :1), which gave the desired product fert-butyl l-(4- (2-chloro-5-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (1.1 Og,
33%). MS (ESI+) e/z: 489.2, 491.2[M+1]+.
[00507] Step F: To a solution of tert-butyl l-(4-(2-chloro-5-methyl-6-phenyl-5H- pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (100 mg, 0.20 mmol) in ethyl acetate (10 mL) was added a solution of HC1 in ethyl acetate (5 mL) at 0 °C. Then it was allowed to reach ambient temperature for 3 h. The reaction mixture was concentrated to remove the solvent. The crude product was purified by HPLC separation, which gave the desired product l-(4-(2-chloro-5- methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine (30 mg, 38% yiled). 1H NMR (MeOD, 400 MHz), <5 ppm: 8.95 (s, 1H), 7.55-7.50 (m, 4H), 7.46-7.39 (m, 5H), 3.78 (s, 3H), 2.76-2.71 (m, 2H), 2.57-2.51 (m, 2H), 2.25-2.18 (m, 1H), 1.95-1.89 (m, 1H). MS (ESI+) e/z: 389.2, 391.2[M+1] +.
Example 138
l-(4-(5-methyl-2,6-diphenyl-5H-pyrrolo[3,2-d1pyrimidin-7-yl)phenyl)cyclobutanamine
[00508] Step A: To a mixed solution (dioxane / H20 = 3 : 0.5 mL) of tert-butyl l-(4-(2- chloro-5-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (160 mg, 0.33 mmol) were added phenylboronic acid (80 mg, 0.65 mmol), Pd(dppf)Cl2 (24 mg, 0.03 mmol), and CS2CO3 (213 mg, 0.65 mmol). The reaction mixture was stirred at 110 °C for 0.5 h. The solvent was removed by concentration in vacuo. The residue was dissolved in ethyl acetate, washed by water twice. The separated organic layer was dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel column separation (eluted by: hexane : ethyl acetate = 1 : 3), which gave desired product tert-butyl l-(4-(5-methyl-2,6-diphenyl-5H-pyrrolo[3,2- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (100 mg, 58% yield). MS (ESf) e/z: 531.3[M+1] +.
[00509] Step B: To a solution of tert-butyl l-(4-(5-methyl-2,6-diphenyl-5H-pyrrolo[3,2- d]pyrimidin-7-yl)phenyl)cyclobutylcarbamate (100 mg, 0.19 mmol) in ethyl acetate (10 mL) was added a solution of HC1 in ethyl acetate (4 mL) at 0 °C. Then it was allowed to reach ambient temperature for 3 h. The resulting precipitate was collected by filtration, washed by ethyl acetate, dried in vacuo, which gave the desired product l-(4-(5-methyl-2,6-diphenyl-5H-pyrrolo[3,2- d]pyrimidin-7-yl)phenyl)cyclobutanamine (80 mg, 99% yiled). 1H NMR (DMSO, 400 MHz), δ ppm: 9.38 (s, 1H), 8.69 (s, 3H), 8.50-8.47 (m, 2H), 7.64-7.44 (m, 12H), 3.73 (s, 3H), 2.67-2.52 (m,
4H), 2.22-2.15 (m, 1H), 1.84-1.78 (m, 1H). MS (ESI+) e/z: 431.3[M+1] +.
[00510] Example 140 shown in Table 15 can also be made according to the above-described methods.
Table 15
Example P
{l-f4-(2-Chloro-7-methyl-5-phenyl-7H-pyrrolo 2,3-d]pyrimidin-6-yl)-phenyl]-cvclobutvU- carbamic acid tert-butyl ester
[00511] Step A: Pd(PPh3)2Cl2 (2.00 g, 5.00mmol) and Cul (2.00 g, lO.OOmmol) were combined in a round-bottom flask, then degassed and refilled with N2 three times. To the mixture was added NEt3 (150 mL), Iodo-benzene (10.20 g, 50.00mmol), Ethynyl-trimethyl-silane (9.80 g, lOO.OOmmol), then the mixture was heated to 100 °C for 30 min. The solvent was removed by vacuum evaporator , then added EA, filtered through Celite. The solvent was removed , and purified by column (eluted by Hexane) to get Trimethyl-phenylethynyl-silane (8.20 g, 98% yield).
[00512] Step B: Methylamine (2.0 M in THF, 60 mL) was cooled to -78 °C, was added the solution of 2,4-Dichloro-5-iodo-pyrimidine in THF (200 mL) cooled to -78 °C. After addition, the mixture was stirred at room temperature for 1 h. The solution was removed by vacuum evaporator. To the residue, was added DCM, which made the residue just dissolved, then n-hexane was added
to the solution. The desired product (2-Chloro-5-iodo-pyrimidin-4-yl)-methyl-amine was crystallized out, and collected by filtration. (7.0g, yield: 52%), MS (ESI+) e/z: 269.7 [M+l]+, 271.7 [M+3]+.
[00513] Step C: (2-Chloro-5-iodo-pyrimidin-4-yl)-methyl-amine(2.70 g, 10.00 mmol),
LiCl(0.42 g, lO.OOmmol) and Na2C03 (4.42 g, 40.00 mmol) are dissolved in DMF (100 mL). The mixture was heated to 100 °C for 12 h. The mixture was poured to water and extracted with EA, dried over Na2S04, purified by column (eluted by ethyl acetate: Hexane = 1 :10) to get desired product 2-Chloro-7-methyl-5-phenyl-6-trimethylsilanyl-7H-pyrrolo[2,3-d]pyrimidine (1.0 g, yield: 32%). Ή MR (CDC13, 400 MHz), δ ppm: 8.29 (s, 1H), 7.25-7.19 (m, 3H), 7.13-7.06 (m, 2H), 3.76 (s, 3 H), 0.00 (s, 9H). MS (ESI+) e/z: 315.9 [M+l]+, 317.9 [M+3]+.
[00514] Step D: To a solution of 2-Chloro-7-methyl-5-phenyl-6-trimethylsilanyl-7H- pyrrolo[2,3-d]pyrimidine (315 mg, 1 mmol) and CF3C02Ag (221 mg, 1 mmol) in THF (15 mL) at - 78 °C under N2 was added dropwise a solution of I2 (254 mg, 1 mmol) in THF (10 mL). The reaction mixture was stirred for 2 h at -78 °C. The resulting mixture was diluted with DCM and then filtered through Celite. The filtrate was washed succeccively with 50% Na2S203, H20, and brine. After drying (MgS04), filtration, and evaporation, the residue was chromatographed on silica (eluted by ethyl acetate: Hexane = 1 :10) to afford 2-Chloro-6-iodo-7-methyl-5-phenyl-7H- pyrrolo[2,3-d] pyrimidine (333 mg, 90%) . MS (ESI+) e/z: 369.8 [M+l]+, 371.8 [M+3]+.
[00515] Step E: 2-Chloro-6-iodo-7-methyl-5-phenyl-7H-pyrrolo[2,3-d] pyrimidine (333 mg,
0.90 mmol), {l-[4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan- 2-yl)- phenyl]-cyclobutyl}-carbamic acid tert-butyl ester (560 mg, 1.5 mmol), Pd(dppf)Cl2 (77 mg, 0.11 mmol), and Cs2C03 (684 mg, 2.10 mmol) were dissolved in a mixed solution (dioxane: 20 mL, water: 4 mL). The reaction mixture was stirred at 100 °C under nitrogen atmosphere for 12 h. LC/MS indicated the reaction worked well. Then the solvents were removed under reduced pressure, the resulting residue was dissolved in ethyl acetate (150 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column (ethyl acetate: Hexane - 1 :2) to give { l-[4-(2-Chloro-7-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (391 mg, 80% yield), MS (ESI+) e/z: 489.2 [M+l]+, 491.2 [M+3]+.
l-[4-(7-Methyl-2.5-diphenyl-7H-pyrrolo[2 -d^
[00516] Step A: l-[4-(7-Methyl-2,5-diphenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-phenyl]- cyclobutylamine (80 mg, 0.16 mmol), phenylboronic acid (24 mg, 0.2 mmol), Pd(dppf)Cl2 (22 mg, 0.03 mmol), Cs2C03 (130 mg, 0.4 mmol) and a mixed solution (dioxane: 5 mL, water: 1 mL) were combined in a microwave tube, and heated to 130 °C for 30 min. LC/MS indicated the reaction worked well. Then the solvents were removed under reduced pressure, the resulting residue was dissolved in ethyl acetate (50 mL). The organic layer was washed with water, dried over sodium sulfate, filtered and concentrated. The crude product was purified by pre-TLC (ethyl acetate: Hexane=l :2) to give { l-[4-(7-Methyl-2,5-diphenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl) -phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (70 mg, 82 % yield), MS (ESI+) e/z: 531.2 [M+l]+, 532.2 [M+2]+ .
[00517] Step B: { l-[4-(7-Methyl-2,5-diphenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl) -phenyl]- cyclobutyl}-carbamic acid tert-butyl ester (70 mg, 0.13 mmol) was dissolved in MeOH (30 mL), then added HC1 (aq) (4M, 8 mL) in drops. The mixture was heated to 80 °C, and stirred for 50 min. LC/MS showed the reaction worked well. The product was purified by HPLC separation to give target compound (17 mg, yield: 30%). 1H-NMR (400 MHz, CDC13) δ: 9.18 (s, 1H), 7.79 (d, J = 8.8Hz, 2H), 7.71-7.45 (m, 7H), 2.91 (s, 3H), 2.80-2.75 (m, 2H), 2.59-2.52 (m, 2H), 2.29-2.20 (m, 1H), 2.05-1.99 (m, 1H). MS (ESI+) e/z: 431.1 [M+l]+
[00518] Examples 141 to 174 shown in Table 16 can also be made according to the above- described methods.
Table 16
[00519] The foregoing description is considered as illustrative only of the principles of the invention. Further, since numerous modifications and changes will be readily apparent to those skilled in the art, it is not desired to limit the invention to the exact construction and process shown as described above. Accordingly, all suitable modifications and equivalents may be considered to fall within the scope of the invention as defined by the claims that follow.
[00520] The words "comprise," "comprising," "include," "including," and "includes" when used in this specification and in the following claims are intended to specify the presence of stated features, integers, components, or steps, but they do not preclude the presence or addition of one or more other features, integers, components, steps, or groups.
Claims
What is claimed is:
1. A compound of Formula (1):
and tautomers, resolved enantiomers, resolved diastereomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs thereof, wherein:
X is N or CR3;
is a roup selected from the grou consisting of:
(J)
m is 0, 1 , 2, 3 or 4;
at least one of R', R2, and R3 is a group of formula (L):
wherein:
n is 0, 1 or 2;
R' and R" are independently selected from the group consisting of H, C]-C6-alkyl and -
(C=0)-CrC6-alkyl;
R' " is H, OH or Ci-C6-alkyl; and the rest of R1, R2, and R3 are independently selected from the group consisting of: H, cyano, Ci- C6-alkyl, aryl or heteroaryl, which aryl or heteroaryl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, cyano, oxo, CrCe-alkyl, CrCe-alkoxyl and C i -C6-hydroxyalkyl;
R4 is selected from the group consisting of: H, halo, cyano, hydroxyl,oxo, C Ce-alkyl, Ci-C6- alkoxyl, COOH, d-Ce-alkylsulfonamide, NRaRb, NRa(C=0)Ra, (C=0) RaRb, (C=0) Ra- CrC6-alkylenyl-NRaRb, NRa-Ci-C6-alkylenyl-COOH, NRa-CrC6-alkylenyl-NRaRb, aryl, heteroaryl or heterocycloalkyl, which aryl, heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one substituent selected from the group consisting of: halo, oxo, hydroxyl, C C6-alkyl, C C6-alkoxyl, C C6-haloalkyl, C C6-haloalkoxyl, COOH, C C6- hydroxyalkyl and Ci-C6-alkylsulfonamide;
Ra and Rb are independently selected from the group consisting of H, Ci-C6-alkyl, Q-
C6-hydroxylalkyl or C3-C6-cycloalkyl.
2. The compounds of claim 1 , wherein R1 is a group of formula (L):
4. The compounds of any one of claim 1 to 3, wherein R' and R" are H, n is 0.
5. The compounds of any one of claim 1 to 4, wherein R4 is selected from the group consisting of: halo, aryl, 5 to 6 membered heteroaryl or heterocycloalkyl containing one, two or three heteroatoms selected from the group consisting of N, S and O, aryl and heteroaryl, which aryl, heteroaryl or heterocycloalkyl is unsubstituted or substituted by at least one halo.
6. The compounds of any one of claim 1 to 5, wherein R3 is H or C]-C6-alkyl.
7. The compounds of any one of claim 1 or 3 to 6, wherein R1 is phenyl.
9. The compounds of claim 8, having the formula (2 -A):
(2-A)
10. The compounds of claim 9, wherein they are selected from the group consisting of:
1 -(4-(2 -phenyl- 1 H-pyrrolo[2,3-b]pyridin-l -yl)phenyl)cyclobutanamine,
l-{4-[6-(2-Fluoro-phenyl)-2-phenyl-pyrrolo[2,3-b]pyridin-l-yl]-phenyl}-yclobutylamine, l-(4-(3-phenyl-lH-pyrrolo[2,3-b]pyridin-2-yl)phenyl)cyclobutanamine,
1 -(4-(2-phenyl- 1 H-pyrrolo[2,3-b]pyridin-3-yl)phenyl)cyclobutanamine, and
l-(4-(l-methyl-2-phenyl-lH-pyrrolo[2,3-b]pyridin-3-yl)phenyl)cyclobutanamine.
11. The compounds of claim 8, having the formula (2-B):
12. The compounds of claim 11, wherein they are selected from the group consisting of:
1 -(4-(2-phenyl- lH-pyrrolo[2,3-c]pyridin- 1 -yl)phenyl)cyclobutanamine, and 1 -(4-(2 -phenyl- 1 H-pyrrolo[2,3-c]pyridin- 1 -yl)phenyl)cyclobutanamine.
13. The compounds of claim 8, having the formula (2-C):
14. The compounds of claim 13, wherein they are selected from the group consisting of: 1 -(4-(l -phenyl- lH-pyrrolo[3,2-c]pyridin-2-yl)phenyl)cyclobutanamine and
l-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-lH-pyrrolo[3,2-c]pyridin-4-ol.
(2-D)
16. The compounds of claim 15, wherein it is l-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-lH- pyrrolo[3,2-b]pyridin-5-ol.
17. The compounds of claim 8 having the formula (2-E):
(2-E)
18. The compounds of claim 17, wherein they are selected from the group consisting of:
l-(4-(6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl) cyclobutanamine,
1 -(4-( 1 -phenyl- 1 H-pyrrolo[3 ,2-c]pyridin-2-yl)phenyl)cyclobutanamine,
4-{7-[4-(l-aminocyclobutyl)phenyl]-6-(pyridin-2- yl)-7H-pyrrolo[2,3-d]pyrimidin-2-yl}-
1 λ6,4- thiomorpholine- 1 , 1 -dione,
1 -(4-( 1 -aminocyclobutyl)phenyl)-2-phenyl- 1 H-pyrrolo[3 ,2-c]pyridin-4-ol,
l-(4-(6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
l-(4-(2-(lH-pyrazol-4-yl)-6-(jpyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
7-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- amine,
l-(4-(2-chloro-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
l-(7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)pyrrolidin-3-ol,
l-(4-(6-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
1- (4-(5-methyl-6-phenyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
(S)-l-(7-(4-(l-aminocyclobutyl)phenyl)-6-^yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)pyrrolidin-3-ol,
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-2-carbonitrile, 7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyiTolo[2,3-d]pyrimidine-2- carboxamide,
(S)- 1 -(7-(4-( 1 -aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyriniidin-2- yl)pyrrolidine-2-carboxylic acid,
Nl-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)ethane- 1 ,2-diamine,
2- (7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- ylamino)acetic acid,
N-(7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)cyclopropanecarboxamide,
7-(4-(l-aminocyclobutyl)phenyl)-N,N-dimethyl-6-(pyridin-2-yl)-7H-pyiTolo[2,3-d]pyrimidin- 4-amine,
l-{4-[5-Methyl-6-phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}- cyclobutylamine,
l-(4-(4-methoxy-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)phenyl)cyclobutanamine, l-(7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- yl)pyrrolidin-2-one,
l-[4-(5-Ethyl-6-pyridin-2-yl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutylamine,
1 -(4-(4-( 1 H-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
7-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- amine,
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-4- carboxamide,
7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidine-4-carboxylic acid,
l-{4-[4-Methoxy-2-(lH-pyrazol-4-yl)-6-pyridin-2-yl-pyiTolo[2,3-d]pyrimidin-7-yl]-phenyl}-
cyclobutylamine,
7-(4-(l-aminocyclobutyl)phenyl)-N,N-diethyl-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- amine,
7-[4-(l-Amino-cyclobutyl)-phenyl]-6-pyridin-2-yl-7H-pyrrolo[2,3-d]pyrimidine-4-carboxylic acid (2-dimethylamino-ethyl)-amide,
1 -(7-(4-( 1 -aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4- yl)pyrrolidin-3-ol,
l-{4-[5-Methyl-6-phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}- cyclobutylamine,
l-{4-[2-(2-Fluoro-phenyl)-5-methyl-6-phenyl-pyrrolo[2,3-d]pyrimidin-7-yl]-phenyl}- cyclobutylamine,
7-(4-( 1 -aminocyclobutyl)phenyl)-N,N-diethyl-6-(pyridin-2-yl)-7H-pyrrolo[2,3 -d]pyrimidin-4- amine,
l-[4-(5-Methyl-2,6-diphenyl-pyrrolo[2,3-d]pyrimidin-7-yl)-phenyl]-cyclobutylamine, l-(4-(5-methyl-6-phenyl-2-(pyrrolidin-l-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
l-(4-(7-methyl-5-phenyl-2-(lH-pyrazol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-6- yl)phenyl)cyclobutanamine,
l-[4-(7-Methyl-2,5-diphenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-phenyl]-cyclobutylamine, l-(4-(2-(2-fluorophenyl)-7-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-6- yl)phenyl)cyclobutanamine,
N-(7-(4-(l-aminocyclobutyl)phenyl)-6-( yridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-2- yl)methanesulfonamide,
7-(4-(l-aminocyclobutyl)phenyl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-ol, l-{4-[2-(2-Fluoro-phenyl)-7-methyl-5-phenyl-7H-pyrrolo[2,3-d]pyrimidin-6-yl]-phenyl}- cyclobutylamine, and
l-[4-(7-Methyl-5-phenyl-2-pyrimidin-5-yl-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-phenyl]- cyclobutylamine.
20. The compounds of claim 19, wherein they are selected from the group consisting of:
l-[4-(2-Chloro-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutylamine,
{ 5 - [4-( 1 - Amino-cyclobutyl)-phenyl] -6-phenyl-5H-pyrrolo [3 ,2-d]pyrimidin-2-yl } -dimethyl- amine,
l-(4-(6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
l-{4-[6-Phenyl-2-(lH-pyrazol-4-yl)-pyrrolo[3,2-d]pyrimidin-5-yl]-phenyl}-cyclobutylamine, l-[4-(6-Phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutylamine,
l-[4-(5-Methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)-phenyl]- cyclobutylamine, Nl-{5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-ethane- 1 ,2-diamine,
1- {5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl}-pyrrolidin-
3-ol,
5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2-carboxylic acid, tert-butyl l-(4-(2-(ethylamino)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-5- yl)phenyl)cyclobutylcarbamate,
2- {5-[4-(l-Amino-cyclobutyl)-phenyl]-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-ylamino}- ethanol,
l-(5-(4-(l-aminocyclobu1yl)phenyl)-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-yl)pyrrolidin-2- one,
l-[4-(2-Methoxy-6-phenyl-pyrrolo[3,2-d]pyrimidin-5-yl)-phenyl]-cyclobutylamine,
5-(4-(l-aminocyclobutyl)phenyl)-N-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidine-2- carboxamide,
l-(4-(5-methyl-6-phenyl-2-(lH-pyrazol-4-yl)-5H-pyrrolo[3,2-d]pyrimidin-7- yl)phenyl)cyclobutanamine,
l-(4-(5-methyl-2,6-diphenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine,
l-(4-(2-chloro-5-methyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-7-yl)phenyl)cyclobutanamine, 5-(4-(l-aminocyclobutyl)phenyl)-N-ethyl-6-phenyl-5H-pyrrolo[3,2-d]pyrimidin-2-amine, and 3-amino-l-methyl-3-(4-(5-methyl-6-phenyl-2-(lH-pyrazol-4-yl)-5H-pyrrolo[3,2-d]pyrimidin- 7-yl)phenyl)cyclobutanol.
21. The compounds of claim 8, having the formula (2-G):
(2-G)
22. The compounds of claim 21, wherein it is l-[4-(6-Phenyl-pyrrolo[2,3-b]pyrazin-5-yl)-phenyl]- cyclobutylamine.
23. The compounds of claim 8, having the formula (2-H):
(2-H)
24. The compounds of claim 8, having the formula (2-1):
(2-1)
(2-J)
(3)
(3-A)
28. The compounds of claim 27, wherein they are selected from the group consisting of: l-(4-(5-methyl-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine, l-(4-(6-bromo-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine, l-(4-(6-(2-fluorophenyl)-2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine, l-(4-(2-phenyl-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine, and
l-(4-(5-phenyl-2-( yridin-2-yl)-3H-imidazo[4,5-b]pyridin-3-yl)phenyl)cyclobutanamine.
29. The compounds of claim 26, having the formula (3-B):
(3-B)
30. The compounds of claim 29, wherein they are selected from the group consisting of:
1- (4-(3-phenyl-3H-imidazo[4,5-c]pyridin-2-yl)phenyl)cyclobutanamine and
2- (4-(l-aminocyclobutyl)phenyl)-l-phenyl-lH-imidazo[4,5-c]pyridin-6-ol.
31. The compounds of claim 26, havin the formula (3-C):
(3-C)
32. The compounds of claim 26, having the formula (3-D):
33. The compounds of claim 26, having the formula (3-E)
(3-E)
34. The compounds of claim 33, wherein they are selected from the group consisting of: l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2,8-diphenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
N-(l-(4-(2,8-diphenyl-9H-purin-9-yl)phenyl)cyclobutyl)acetamide;
l-(4-(2-phenyl-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-(2-fluorophenyl)-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -[4-(8-Phenyl-2-piperazin- 1 -yl-purin-9-yl)-phenyl]-cyclobutylamine;
l-{4-[2-(2-Fluoro-phenyl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine;
{9-[4-(l-Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl}-methyl-amine;
{ 9-[4-( 1 -Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl } -dimethyl-amine;
l-(4-(2-chloro-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
9-[4-( 1 -Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purine-2-carboxylic acid methylamide; l-{9-[4-(l-Amino-cyclobutyl)-phenyl]-8-phenyl-9H-purin-2-yl}-pyiTolidin-2-one;
l-[4-(8-Phenyl-purin-9-yl)-phenyl]-cyclobutylamine;
l-[4-(8-Phenyl-2-pyridin-2-yl-purin-9-yl)-phenyl]-cyclobutylamine;
9-(4-(l-aminocyclobutyl)phenyl)-8-phenyl-9H-purin-2-amine;
l-(4-(2-chloro-8-(thiazol-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(2-methoxy-8-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-{4-[2-(3-Methyl-isoxazol-5-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine; l-{4-[2-(5-Methyl-2H-pyrazol-3-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine; l-{4-[2-(2-Methyl-3H-imidazol-4-yl)-8-phenyl-purin-9-yl]-phenyl}-cyclobutylamine; l-[4-(2-Phenyl-8-pyridin-3-yl-purin-9-yl)-phenyl]-cyclobutylamine;
l-(4-(2-phenyl-8-(pyridin-4-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-[4-(2-Ethoxy-8-phenyl-purin-9-yl)-phenyl]-cyclobutylamine;
9-(4-(l-aminocyclobutyl)phenyl)-N,N-dimethyl-8-phenyl-9H-purin-6-amine;
l-{4-[8-Phenyl-2-(lH-pyrazol-4-yl)-purin-9-yl]-phenyl}-cyclobutylamine;
1 - {4-[2-( 1 -Methyl- 1 H-pyrazol-4-yl)-8-phenyl-purin-9-yl]-phenyl} -cyclobutylamine; l-(4-(8-(4-methoxypyridin-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 H-imidazol-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-phenyl-8-(thiazol-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(9-phenyl-9H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(8-phenyl-2-(pyrrolidin-l-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(3-methoxypyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(8-(5-methoxypyridin-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(6-( 1 -methyl- 1 H-pyrazol-4-yl)-8-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(2-phenyl-8-(pyrimidin-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-[4-(6-Methyl-8-phenyl-purin-9-yl)-phenyl]-cyclobutylamine;
l-(4-(8-tert-butyl-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-( 1 H-pyrazol-4-yl)-6-(pyridin-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7- yl)phenyl)cyclobutanamine;
1 -(4-(8-(4-methyl- 1 H-imidazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 H-imidazol-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(4-methylthiazol-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-benzo[d]imidazol-2-yl)-2-phenyl-9H-purin-9- yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-imidazol-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(8-(5-methylisoxazol-3-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(8-(oxazol-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-pyrazol-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-imidazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)ethane- 1 ,2-diamine;
N-(9-(4-(l-aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)methanesulfonamide; l-(4-(2-chloro-7-phenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(2-phenyl-8-(lH-pyrazol-4-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2-phenyl-8-(thiazol-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-( 1 H-pyrazol-4-yl)-8-(pyridin-2-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(6-methoxypyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(7-phenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
1- (4-(2-phenyl-8-(lH-pyrazol-5-yl)-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)pyrrolidin-3-ol;
2- (4-(9-(4-( 1 -aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-2-yl)- 1 H-pyrazol- 1 - yl)ethanol;
6-(9-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-9H-purin-8-yl)pyridin-2(lH)-one;
l-(4-(2-methoxy-7-phenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
9-(4-(l-aminocyclobutyl)phenyl)-8-(pyridin-2-yl)-9H-purin-6-ol;
8- (4-( 1 -aminocyclobutyl)phenyl)-7-phenyl-7H-purin-2-ol;
l-(4-(7-phenyl-2-(pyridin-3-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(7-phenyl-2-(pyrimidin-5-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
9- [4-(l-Amino-cyclobutyl)-phenyl]-2-phenyl-9H-purine-8-carbonitrile;
1 -(4-(8-( 1 -ethyl- 1 H-pyrazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; l-(4-(7-phenyl-2-(lH-pyrazol-4-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
1 -(4-(2-(2-fluorophenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yI)-9H-purin-9- yl)phenyl)cyclobutanamine;
l-(4-(2-phenyl-8-o-tolyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(2,7-diphenyl-7H-purin-8-yl)phenyl)cyclobutanamine;
1 -(4-(7-phenyl-2-( 1 H-pyrazol- 1 -yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
l-(4-(8-(3-bromopyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(4-methylthiazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine;
l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-(3-(trifluoromethoxy)phenyl)-9H-purin-9- yl)phenyl)cyclobutanamine;
l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-(3-(trifluoromethyl)phenyl)-9H-purin-9- yl)phenyl)cyclobutanamine;
l-(4-(7-phenyl-2-(thiazol-4-yl)-7H-purin-8-yl)phenyl)cyclobutanamine;
1- (4-(8-(3-methylpyridin-2-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; 1 -(4-(2-(4-methoxyphenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine ;
2- (9-(4-(l-aminocyclobutyl)phenyl)-2-phenyl-9H-purin-8-yl)benzonitrile;
1 -(4-(2-(2-methoxyphenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine;
4-(9-(4-( 1 -aminocyclobutyl)phenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-2-yl)-N- methylbenzenesulfonamide ;
l-(4-(8-(l-methyl-lH-pyrazol-5-yl)-2-p-tolyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-(2-fluorophenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- yl)phenyl)cyclobutanamine;
1 -(4-(8-( 1 -methyl- 1 H-pyrazol-5-yl)-2-m-tolyl-9H-purin-9-yl)phenyl)cyclobutanamine;
1 -(4-(2-(4-fluorophenyl)-8-( 1 -methyl- 1 H-pyrazol-5-yl)-9H-purin-9- y l)pheny l)cyclobutanamine ;
l-(4-(8-(3-methoxypyridin-4-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine; and
1 -(4-(8-(4-methyl- 1 H-imidazol-5-yl)-2-phenyl-9H-purin-9-yl)phenyl)cyclobutanamine.
. The compounds of claim 26, havin the formula (3-F)
(3-F)
36. The compounds of claim 26, h ing the formula (3-G)
(3-G)
37. The compounds of claim 36, wherein they are selected from the group consisting of:
l-[4-(2-Phenyl-imidazo[4,5-b]pyrazin-l-yl)-phenyl]-cyclobutylamine; and
l-(4-(2-phenyl-5-(lH-pyrazol-4-yl)-lH-imidazo[4,5-b]pyrazin-l-yl)phenyl)cyclobutanamine.
38. The compounds of claim 26, havin the formula (3-H):
(3-H)
39. The compounds of claim 26 having the formula (3-1):
(3-D
40. The compounds of claim 26, having the formula (3-J):
(3-J)
A pharmaceutical composition comprising a compound as claimed in any one of Claims 1
42. A method of treating an AKT-mediated disease or disorder in a mammal, said method comprising administering to said mammal a therapeutically effective amount of a compound as claimed in any one of Claims 1-40.
43. The method of Claim 42, wherein said disease or disorder is an inflammatory, hyperproliferative, cardiovascular, neurodegenerative, gynecological, or dermatological disease or disorder.
44. A method of inhibiting the production of AKT protein kinase in a mammal, which comprises administering to said mammal an effective amount of a compound as claimed in any one of Claims 1-40.
45. A method of inhibiting the activity of AKT protein kinase in a mammal, which comprises contacting said kinase with a compound as claimed in any one of Claims 1-40.
46. A compound as claimed in any one of Claims 1-40 for use as a medicament in the treatment of AKT protein kinase-mediated conditions.
47. The use of a compound as claimed in any one of Claims 1-40 in the manufacture of a medicament for therapy.
48. A kit for treating an AKT protein kinase-mediated condition, wherein said kit comprises: a) a first pharmaceutical composition comprising a compound as claimed in any one of Claims 1-40; and
b) optionally instructions for use.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161562950P | 2011-11-22 | 2011-11-22 | |
US61/562,950 | 2011-11-22 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2013078254A1 true WO2013078254A1 (en) | 2013-05-30 |
Family
ID=47297471
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2012/066153 WO2013078254A1 (en) | 2011-11-22 | 2012-11-20 | Bicyclic heteroaryl derivatives as kinase inhibitors |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2013078254A1 (en) |
Cited By (30)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014140592A1 (en) * | 2013-03-13 | 2014-09-18 | Proximagen Limited | Imidazo[4,5-c]pyridine and pyrrolo[2,3-c]pyridine derivatives as ssao inhibitors |
US20140275013A1 (en) * | 2013-03-15 | 2014-09-18 | University Of Washington Through Its Center For Commercialization | Compounds and compositions for the treatment of parasitic diseases |
US9186361B2 (en) | 2013-03-15 | 2015-11-17 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
CN105130987A (en) * | 2015-10-08 | 2015-12-09 | 张妍 | Synthetic method for medical intermediate pyrrolo-pyridine compound |
US9296754B2 (en) | 2013-03-15 | 2016-03-29 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US9303034B2 (en) | 2013-12-19 | 2016-04-05 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
JP2016124825A (en) * | 2014-12-27 | 2016-07-11 | ヤマサ醤油株式会社 | 2,6-Dichloro-8-iodo-7-deazapurine for the synthesis of polysubstituted 7-deazapurine derivatives |
JP2016539980A (en) * | 2013-12-09 | 2016-12-22 | ユーシービー バイオファルマ エスピーアールエル | Purine derivatives as modulators of TNF activity |
CN106459036A (en) * | 2014-03-24 | 2017-02-22 | 艾科尔公司 | Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine |
US9580415B2 (en) | 2013-03-13 | 2017-02-28 | Proximagen Limited | Compounds |
US9718847B2 (en) | 2013-03-15 | 2017-08-01 | Plexxikon Inc. | Heterocyclic compounds and uses thereof |
US9822109B2 (en) | 2013-03-15 | 2017-11-21 | Plexxikon Inc. | Heterocyclic compounds and uses thereof |
US10065954B2 (en) | 2014-09-17 | 2018-09-04 | Proximagen Limited | Substituted imidazo[4,5-c]pyridines as SSAO inhibitors |
US10239881B2 (en) | 2014-10-27 | 2019-03-26 | University Health Network | RIPK2 inhibitors and method of treating cancer with same |
US10508113B2 (en) | 2018-03-12 | 2019-12-17 | Abbvie Inc. | Inhibitors of tyrosine kinase 2 mediated signaling |
US10717735B2 (en) | 2017-10-13 | 2020-07-21 | Plexxikon Inc. | Solid forms of a compound for modulating kinases |
US11168093B2 (en) | 2018-12-21 | 2021-11-09 | Celgene Corporation | Thienopyridine inhibitors of RIPK2 |
WO2022109577A1 (en) * | 2020-11-18 | 2022-05-27 | Relay Therapeutics, Inc. | Fgfr inhibitors and methods of making and using the same |
US11352328B2 (en) | 2016-07-12 | 2022-06-07 | Arisan Therapeutics Inc. | Heterocyclic compounds for the treatment of arenavirus |
US11471455B2 (en) | 2018-10-05 | 2022-10-18 | Annapurna Bio, Inc. | Compounds and compositions for treating conditions associated with APJ receptor activity |
US11530208B2 (en) | 2014-09-17 | 2022-12-20 | Proximagen, Llc | Imidazo[4,5-C]pyridine derived SSAO inhibitors |
US11618751B1 (en) | 2022-03-25 | 2023-04-04 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 derivatives |
CN116283993A (en) * | 2021-12-20 | 2023-06-23 | 艾立康药业股份有限公司 | A kind of pyrimidine compound and its preparation method and application |
US11767321B2 (en) | 2020-10-05 | 2023-09-26 | Enliven Inc. | 5- and 6-azaindole compounds for inhibition of BCR-ABL tyrosine kinases |
US11780845B2 (en) | 2019-05-13 | 2023-10-10 | Relay Therapeutics, Inc. | FGFR inhibitors and methods of use thereof |
US11814388B1 (en) | 2020-08-28 | 2023-11-14 | Ferris State University | Substituted pyrrolo[2,3-d]pyrimidines and pyrazolo[3,4-d]pyrimidines as inhibitors for multi-resistant cancers |
WO2024095005A1 (en) | 2022-11-02 | 2024-05-10 | Cerevance, Inc. | Diaryl diazole and diaryl triazole derivatives for use in treating a disease associated with kcnk13 activity |
WO2024107565A1 (en) * | 2022-11-14 | 2024-05-23 | Alterome Therapeutics, Inc. | Akt1 modulators |
US12281112B2 (en) | 2021-04-07 | 2025-04-22 | Ventus Therapeutics U.S., Inc. | Compounds for inhibiting NLRP3 and uses thereof |
US12312351B2 (en) | 2022-10-31 | 2025-05-27 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US3773919A (en) | 1969-10-23 | 1973-11-20 | Du Pont | Polylactide-drug mixtures |
WO1996015111A1 (en) | 1994-11-15 | 1996-05-23 | Regents Of The University Of Minnesota | Method and intermediates for the synthesis of korupensamines |
WO2005113762A1 (en) | 2004-05-18 | 2005-12-01 | Pfizer Products Inc. | CRYSTAL STRUCTURE OF PROTEIN KINASE B-α (AKT-1) AND USES THEREOF |
WO2011055115A1 (en) * | 2009-11-04 | 2011-05-12 | Almac Discovery Limited | Akt / pkb inhibitors |
WO2011082270A2 (en) * | 2009-12-30 | 2011-07-07 | Arqule. Inc. | Substituted imidazopyridinyl-aminopyridine compounds |
-
2012
- 2012-11-20 WO PCT/US2012/066153 patent/WO2013078254A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US3773919A (en) | 1969-10-23 | 1973-11-20 | Du Pont | Polylactide-drug mixtures |
WO1996015111A1 (en) | 1994-11-15 | 1996-05-23 | Regents Of The University Of Minnesota | Method and intermediates for the synthesis of korupensamines |
WO2005113762A1 (en) | 2004-05-18 | 2005-12-01 | Pfizer Products Inc. | CRYSTAL STRUCTURE OF PROTEIN KINASE B-α (AKT-1) AND USES THEREOF |
WO2011055115A1 (en) * | 2009-11-04 | 2011-05-12 | Almac Discovery Limited | Akt / pkb inhibitors |
WO2011082270A2 (en) * | 2009-12-30 | 2011-07-07 | Arqule. Inc. | Substituted imidazopyridinyl-aminopyridine compounds |
Non-Patent Citations (59)
Title |
---|
"Beilsteins Handbuch der organischen Chemie", SPRINGER-VERLAG |
"Chiral Liquid Chromatography", 1989, CHAPMAN AND HALL |
"Comprehensive Medicinal Chemistry", vol. 5, 1990, PERGAMON PRESS |
"Design of Prodrugs", 1985, ELSEVIER |
"Drug Stereochemistry, Analytical Methods and Pharmacology", 1993, MARCEL DEKKER, INC. |
"Methods in Enzymology", vol. 42, 1985, ACADEMIC PRESS, pages: 309 - 396 |
"Remington's Pharmaceutical Sciences", 1980 |
ADVANCED DRUG DELIVERY REVIEWS, vol. 19, 1996, pages 115 |
ALESSI ET AL., CURR. BIOL., vol. 7, 1997, pages 261 - 269 |
ANGEW CHEM. INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186 |
BALENDRAN, A., CURR. BIOL., vol. 9, 1999, pages 393 |
BELLACOSA ET AL., INT. J. CANCER, vol. 64, 1995, pages 280 - 285 |
BRENNFIIHRER, A. ET AL., ANGEW. CHEM., vol. 48, no. 23, 2009, pages 4114 |
BRENNFIIHRER, A. ET AL., ANGEW.CBEM., vol. 48, no. 23, 2009, pages 4114 |
BRODBECK ET AL., J. BIOL. CHEM., vol. 274, 1999, pages 9133 - 9136 |
BROGNARD ET AL., CANCER RES., vol. 61, 2001, pages 3986 |
CHENG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 9267 |
CHENG ET AL., PROC. NATL. ACAD. SCI., vol. 93, 1996, pages 3636 - 3641 |
COFFER ET AL., EUR. J. BIOCHEM., vol. 201, 1991, pages 475 - 481 |
COHEN, P., NATURE REV. DRUG DISCOVERY, vol. 1, 2002, pages 309 |
DELCOMMENNE, M., PROC. NATL. ACAD. SCI. USA, vol. 95, 1998, pages 11211 |
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC. |
GEORGAKIS, G.; YOUNES, A., EXPERT REV. ANTICANCER THER., vol. 6, no. 1, 2006, pages 131 - 140 |
GRAFF ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 24500 |
GRANVILLE ET AL., CLIN. CANCER RES., vol. 12, no. 3, 2006, pages 679 - 689 |
H. BUNDGAARD ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 77, 1988, pages 285 |
H. BUNDGAARD, ADVANCED DRUG DELIVERY REVIEWS, vol. 8, 1992, pages 1 - 38 |
H. BUNDGAARD: "A Textbook of Drug Design and Development", 1991, article "Design and Application of Prodrugs", pages: 113 - 191 |
HAAS KOGAN ET AL., CURR. BIOL., vol. 8, 1998, pages 1195 |
HARDIE, G.; HANKS, S.: "The Protein Kinase Facts Book. I and II", 1995, ACADEMIC PRESS |
HARTMAN, G.D. ET AL., J. HET. CHEM., vol. 20, 1983, pages 947 - 950 |
HARTWIG, ACE. CHEM. RES., vol. 31, 1998, pages 852 |
HAY N., CANCER CELL, vol. 8, 2005, pages 179 - 183 |
HEMMINGS, B.A., SCIENCE, vol. 275, 1997, pages 628 |
ISHIYAMA T. ET AL., J. ORG. CHEM., 2003 |
J. MED. CHEM., vol. 39, 1996, pages 10 |
JACOB III., J. ORG. CHEM., vol. 47, 1982, pages 4165 |
JI J. ET AL., ORG. LETT, vol. 5, 2003, pages 4611 |
KIM ET AL.: "Current Opinion in Investig", DRUGS, vol. 6, no. 12, 2005, pages 1250 - 1258 |
LEY S. ET AL., ANGEW. CHEM., vol. 75, 2003, pages 5558 |
LI ET AL., CURRENT TOPICS IN MED. CHEM., vol. 2, 2002, pages 939 - 971 |
LOCHMULLER, C. H., J. CHROMATOGR., vol. 113, no. 3, 1975, pages 283 - 302 |
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-19, 1967, WILEY |
LUO ET AL., MOLECULAR CANCER THER., vol. 4, no. 6, 2005, pages 977 - 986 |
N. KAKEYA ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692 |
NAKATANI, K., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 257, 1999, pages 906 |
NAVARRO O. ET AL., J. ORG. CHEM., vol. 69, 2004, pages 3173 |
NEGISHI ET AL., CHEM. REV., vol. 103, no. 5, 2003, pages 1979 - 2013 |
OKAMOTO, J. OF CHROMATOGR., vol. 513, 1990, pages 375 - 378 |
ORG. LETT., vol. 11, no. 8, 2009, pages 1837 - 1840 |
ROWE, RAYMOND C.: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS |
SAPOUNTZIS L ET AL., J. AM. CHEM. SOC, vol. 24, 2002, pages 9390 |
STAAL ET AL., PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 5034 - 5037 |
STAAL, S.P., PROC. NATL. ACAD. SCI., vol. 84, 1987, pages 5034 |
T.W. GREENE; P.G.M. WUTS: "Protective groups in Organic Synthesis", 1999, JOHN WILEY & SONS |
TOKER ET AL., CANCER RES., vol. 66, no. 8, 2006, pages 3963 - 3966 |
TOKER, A., J. BIOL. CHEM., vol. 275, 2000, pages 8271 |
WOLFE J. ET AL., ACE. CHEM. RES., vol. 31, 1998, pages 805 |
ZINDA ET AL., CLIN. CANCER RES., vol. 7, 2001, pages 2475 |
Cited By (61)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9580415B2 (en) | 2013-03-13 | 2017-02-28 | Proximagen Limited | Compounds |
US9951068B2 (en) | 2013-03-13 | 2018-04-24 | Proximagen Limited | Imidazo[4,5-C]pyridine and pyrrolo[2,3-C]pyridine derivatives as SSAO inhibitors |
EA034066B1 (en) * | 2013-03-13 | 2019-12-24 | Проксимэйджен, Элэлси | IMIDAZO[4,5-c]PYRIDINE DERIVATIVES AS SSAO INHIBITORS |
US11512082B2 (en) | 2013-03-13 | 2022-11-29 | Proximagen, Llc | Substituted imidazo[4,5-c]pyridine compounds and compositions thereof |
US10590125B2 (en) | 2013-03-13 | 2020-03-17 | Proximagen, Llc | Compounds |
US10428066B2 (en) | 2013-03-13 | 2019-10-01 | Proximagen, Llc | Imidazo[4,5-C]pyridine and pyrrolo[2,3-C]pyridine derivatives as SSAO inhibitors |
US10968223B2 (en) | 2013-03-13 | 2021-04-06 | Proximagen, Llc | Imidazo[4,5-c]pyridine and pyrrolo[2,3-c]pyridine derivatives as SSAO inhibitors |
US10766897B2 (en) | 2013-03-13 | 2020-09-08 | Proximagen, Llc | Imidazo[4,5-c]pyridine and pyrrolo[2,3-c]pyridine derivatives as SSAO inhibitors |
US9428498B2 (en) | 2013-03-13 | 2016-08-30 | Proximagen Limited | Substituted imidazo[4,5-c]pyridine compounds and compositions thereof |
US9676769B2 (en) | 2013-03-13 | 2017-06-13 | Proximagen Limited | Imidazo[4,5-C]pyridine and pyrrolo[2,3-C]pyridine derivatives as SSAO inhibitors |
WO2014140592A1 (en) * | 2013-03-13 | 2014-09-18 | Proximagen Limited | Imidazo[4,5-c]pyridine and pyrrolo[2,3-c]pyridine derivatives as ssao inhibitors |
US9296754B2 (en) | 2013-03-15 | 2016-03-29 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US9233961B2 (en) | 2013-03-15 | 2016-01-12 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US10519177B2 (en) | 2013-03-15 | 2019-12-31 | Plexxikon Inc. | Heterocyclic compounds and uses thereof |
US9718847B2 (en) | 2013-03-15 | 2017-08-01 | Plexxikon Inc. | Heterocyclic compounds and uses thereof |
US9186361B2 (en) | 2013-03-15 | 2015-11-17 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US9822109B2 (en) | 2013-03-15 | 2017-11-21 | Plexxikon Inc. | Heterocyclic compounds and uses thereof |
US10501460B2 (en) | 2013-03-15 | 2019-12-10 | Plexxikon Inc. | Heterocyclic compounds and uses thereof |
US20140275013A1 (en) * | 2013-03-15 | 2014-09-18 | University Of Washington Through Its Center For Commercialization | Compounds and compositions for the treatment of parasitic diseases |
JP2016539980A (en) * | 2013-12-09 | 2016-12-22 | ユーシービー バイオファルマ エスピーアールエル | Purine derivatives as modulators of TNF activity |
US10596175B2 (en) | 2013-12-19 | 2020-03-24 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US9700559B2 (en) | 2013-12-19 | 2017-07-11 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US11123348B2 (en) | 2013-12-19 | 2021-09-21 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US9303034B2 (en) | 2013-12-19 | 2016-04-05 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
US10085989B2 (en) | 2013-12-19 | 2018-10-02 | Novartis Ag | Compounds and compositions for the treatment of parasitic diseases |
JP2017516749A (en) * | 2014-03-24 | 2017-06-22 | アークル インコーポレイテッド | Method for preparing 3- (3- (4- (1-aminocyclobutyl) phenyl) -5-phenyl-3H-imidazo [4,5-B] pyridin-2-yl) pyridin-2-amine |
EP3122748A4 (en) * | 2014-03-24 | 2017-09-27 | ArQule, Inc. | Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine |
US9834556B2 (en) | 2014-03-24 | 2017-12-05 | Arqule, Inc. | Process of preparing 3-(3-(4-aminocyclobutyl)phenyl)-5-phenyl-3H-imidazo[4,5-B]pyridin-2-YL)pyridin-2-amine |
CN106459036A (en) * | 2014-03-24 | 2017-02-22 | 艾科尔公司 | Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine |
TWI662033B (en) * | 2014-03-24 | 2019-06-11 | 美商亞闊股份有限公司 | Process of preparing 3-(3-(4-(1-aminocyclobutyl)phenyl)-5-phenyl-3h-imidazo[4,5-b]pyridin-2-yl)pyridin-2-amine |
US11530208B2 (en) | 2014-09-17 | 2022-12-20 | Proximagen, Llc | Imidazo[4,5-C]pyridine derived SSAO inhibitors |
US10065954B2 (en) | 2014-09-17 | 2018-09-04 | Proximagen Limited | Substituted imidazo[4,5-c]pyridines as SSAO inhibitors |
US10875863B2 (en) | 2014-10-27 | 2020-12-29 | University Health Network | RIPK2 inhibitors and method of treating cancer with same |
US10239881B2 (en) | 2014-10-27 | 2019-03-26 | University Health Network | RIPK2 inhibitors and method of treating cancer with same |
JP2016124825A (en) * | 2014-12-27 | 2016-07-11 | ヤマサ醤油株式会社 | 2,6-Dichloro-8-iodo-7-deazapurine for the synthesis of polysubstituted 7-deazapurine derivatives |
CN105130987A (en) * | 2015-10-08 | 2015-12-09 | 张妍 | Synthetic method for medical intermediate pyrrolo-pyridine compound |
US11352328B2 (en) | 2016-07-12 | 2022-06-07 | Arisan Therapeutics Inc. | Heterocyclic compounds for the treatment of arenavirus |
US10717735B2 (en) | 2017-10-13 | 2020-07-21 | Plexxikon Inc. | Solid forms of a compound for modulating kinases |
US10508113B2 (en) | 2018-03-12 | 2019-12-17 | Abbvie Inc. | Inhibitors of tyrosine kinase 2 mediated signaling |
US11471455B2 (en) | 2018-10-05 | 2022-10-18 | Annapurna Bio, Inc. | Compounds and compositions for treating conditions associated with APJ receptor activity |
US11944622B2 (en) | 2018-10-05 | 2024-04-02 | Annapurna Bio, Inc. | Compounds and compositions for treating conditions associated with APJ receptor activity |
US11168093B2 (en) | 2018-12-21 | 2021-11-09 | Celgene Corporation | Thienopyridine inhibitors of RIPK2 |
US11780845B2 (en) | 2019-05-13 | 2023-10-10 | Relay Therapeutics, Inc. | FGFR inhibitors and methods of use thereof |
US11814388B1 (en) | 2020-08-28 | 2023-11-14 | Ferris State University | Substituted pyrrolo[2,3-d]pyrimidines and pyrazolo[3,4-d]pyrimidines as inhibitors for multi-resistant cancers |
US11767321B2 (en) | 2020-10-05 | 2023-09-26 | Enliven Inc. | 5- and 6-azaindole compounds for inhibition of BCR-ABL tyrosine kinases |
US11807638B2 (en) | 2020-10-05 | 2023-11-07 | Enliven Inc. | 5- and 6-azaindole compounds for inhibition of Bcr-Abl tyrosine kinases |
US12240846B2 (en) | 2020-10-05 | 2025-03-04 | Enliven Inc. | 5- and 6-azaindole compounds for inhibition of Bcr-Abl tyrosine kinases |
WO2022109577A1 (en) * | 2020-11-18 | 2022-05-27 | Relay Therapeutics, Inc. | Fgfr inhibitors and methods of making and using the same |
US12152034B2 (en) | 2020-11-18 | 2024-11-26 | Relay Therapeutics, Inc. | FGFR inhibitors and methods of making and using the same |
US12351578B2 (en) | 2021-04-07 | 2025-07-08 | Ventus Therapeutics U.S., Inc. | Compounds for inhibiting NLRP3 and uses thereof |
US12312350B2 (en) | 2021-04-07 | 2025-05-27 | Ventus Therapeutics U.S., Inc. | Compounds for inhibiting NLRP3 and uses thereof |
US12281112B2 (en) | 2021-04-07 | 2025-04-22 | Ventus Therapeutics U.S., Inc. | Compounds for inhibiting NLRP3 and uses thereof |
CN116283993A (en) * | 2021-12-20 | 2023-06-23 | 艾立康药业股份有限公司 | A kind of pyrimidine compound and its preparation method and application |
CN116283993B (en) * | 2021-12-20 | 2024-05-03 | 艾立康药业股份有限公司 | A pyrimidine compound and its preparation method and application |
US12168657B2 (en) | 2022-03-25 | 2024-12-17 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 derivatives |
US12195460B2 (en) | 2022-03-25 | 2025-01-14 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors |
US11618751B1 (en) | 2022-03-25 | 2023-04-04 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 derivatives |
US12312351B2 (en) | 2022-10-31 | 2025-05-27 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors |
US12331048B2 (en) | 2022-10-31 | 2025-06-17 | Ventus Therapeutics U.S., Inc. | Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors |
WO2024095005A1 (en) | 2022-11-02 | 2024-05-10 | Cerevance, Inc. | Diaryl diazole and diaryl triazole derivatives for use in treating a disease associated with kcnk13 activity |
WO2024107565A1 (en) * | 2022-11-14 | 2024-05-23 | Alterome Therapeutics, Inc. | Akt1 modulators |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2013078254A1 (en) | Bicyclic heteroaryl derivatives as kinase inhibitors | |
EP2240483B1 (en) | Pyrimidyl cyclopentanes as akt protein kinase inhibitors | |
CN101981037B (en) | Pyrazolopyrimidine PI3K inhibitor compounds and methods of use | |
TWI441824B (en) | Tricyclic pi3k inhibitor compounds and methods of use | |
CN105452223B (en) | As Raf kinases and/or the fused tricyclic carbamide compounds of Raf kinases dimer inhibitor | |
RU2539568C2 (en) | Pyrazolpyrimidine compounds jak inhibitors and methods | |
JP5808869B2 (en) | Bicyclic piperazine compounds | |
CN102224152B (en) | Pyrazolopyridine PI3K inhibitor compounds and methods of use | |
EP2279188A1 (en) | Purine pi3k inhibitor compounds and methods of use | |
EP2611798A1 (en) | Pyridazinones, method of making, and method of use thereof | |
CN104583215B (en) | Diene also-and piperazine-[2,3-D] pyrimidine PI3K inhibitor compound and using method | |
AU2015268776B2 (en) | Tricyclic PI3k inhibitor compounds and methods of use | |
HK1149270B (en) | Pyrimidyl cyclopentanes as akt protein kinase inhibitors |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 12798115 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 12798115 Country of ref document: EP Kind code of ref document: A1 |