NZ740737B2 - Isoindolinone inhibitors of the mdm2-p53 interaction having anticancer activity - Google Patents
Isoindolinone inhibitors of the mdm2-p53 interaction having anticancer activity Download PDFInfo
- Publication number
- NZ740737B2 NZ740737B2 NZ740737A NZ74073716A NZ740737B2 NZ 740737 B2 NZ740737 B2 NZ 740737B2 NZ 740737 A NZ740737 A NZ 740737A NZ 74073716 A NZ74073716 A NZ 74073716A NZ 740737 B2 NZ740737 B2 NZ 740737B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- chlorophenyl
- methyl
- dihydro
- compound
- fluoro
- Prior art date
Links
- 239000003112 inhibitor Substances 0.000 title claims description 31
- PXZQEOJJUGGUIB-UHFFFAOYSA-N isoindolin-1-one Chemical compound C1=CC=C2C(=O)NCC2=C1 PXZQEOJJUGGUIB-UHFFFAOYSA-N 0.000 title description 11
- 230000003993 interaction Effects 0.000 title description 9
- 230000001093 anti-cancer Effects 0.000 title description 4
- 150000001875 compounds Chemical class 0.000 claims abstract description 531
- 150000003839 salts Chemical class 0.000 claims abstract description 148
- 239000012453 solvate Substances 0.000 claims abstract description 116
- 238000000034 method Methods 0.000 claims abstract description 85
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 29
- 230000008569 process Effects 0.000 claims abstract description 14
- -1 haloC1-6alkyl Chemical group 0.000 claims description 279
- 125000000623 heterocyclic group Chemical group 0.000 claims description 273
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 269
- 125000003854 p-chlorophenyl group Chemical group [H]C1=C([H])C(*)=C([H])C([H])=C1Cl 0.000 claims description 185
- 229910052739 hydrogen Inorganic materials 0.000 claims description 167
- 239000001257 hydrogen Substances 0.000 claims description 165
- 206010028980 Neoplasm Diseases 0.000 claims description 121
- 229910052736 halogen Inorganic materials 0.000 claims description 117
- 150000002367 halogens Chemical class 0.000 claims description 111
- 125000001153 fluoro group Chemical group F* 0.000 claims description 90
- 150000002825 nitriles Chemical class 0.000 claims description 86
- 125000005842 heteroatom Chemical group 0.000 claims description 77
- 150000002431 hydrogen Chemical group 0.000 claims description 74
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 70
- 229910052760 oxygen Inorganic materials 0.000 claims description 70
- 238000011282 treatment Methods 0.000 claims description 66
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 65
- 125000001309 chloro group Chemical group Cl* 0.000 claims description 61
- 229910052717 sulfur Inorganic materials 0.000 claims description 61
- 239000003795 chemical substances by application Substances 0.000 claims description 57
- 201000011510 cancer Diseases 0.000 claims description 54
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 53
- 125000003386 piperidinyl group Chemical group 0.000 claims description 50
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 44
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 44
- 201000010099 disease Diseases 0.000 claims description 44
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 44
- 125000004122 cyclic group Chemical group 0.000 claims description 43
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 43
- 125000001412 tetrahydropyranyl group Chemical group 0.000 claims description 43
- 229910052731 fluorine Inorganic materials 0.000 claims description 42
- 239000003814 drug Substances 0.000 claims description 40
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 claims description 38
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 claims description 35
- 239000011737 fluorine Substances 0.000 claims description 33
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 32
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 claims description 30
- 125000000882 C2-C6 alkenyl group Chemical group 0.000 claims description 30
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 claims description 30
- 235000019260 propionic acid Nutrition 0.000 claims description 30
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 29
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 claims description 28
- DUWWHGPELOTTOE-UHFFFAOYSA-N n-(5-chloro-2,4-dimethoxyphenyl)-3-oxobutanamide Chemical compound COC1=CC(OC)=C(NC(=O)CC(C)=O)C=C1Cl DUWWHGPELOTTOE-UHFFFAOYSA-N 0.000 claims description 28
- 150000002500 ions Chemical class 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
- 229910052799 carbon Inorganic materials 0.000 claims description 26
- 125000001424 substituent group Chemical group 0.000 claims description 26
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Chemical compound CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 claims description 25
- 125000003118 aryl group Chemical group 0.000 claims description 25
- JFDZBHWFFUWGJE-UHFFFAOYSA-N benzenecarbonitrile Natural products N#CC1=CC=CC=C1 JFDZBHWFFUWGJE-UHFFFAOYSA-N 0.000 claims description 25
- 229910052757 nitrogen Chemical group 0.000 claims description 23
- 229940079593 drug Drugs 0.000 claims description 22
- 238000011321 prophylaxis Methods 0.000 claims description 22
- 125000003226 pyrazolyl group Chemical group 0.000 claims description 22
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 claims description 19
- 125000002883 imidazolyl group Chemical group 0.000 claims description 19
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 claims description 18
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 18
- 125000005843 halogen group Chemical group 0.000 claims description 17
- 230000001225 therapeutic effect Effects 0.000 claims description 17
- 125000003601 C2-C6 alkynyl group Chemical group 0.000 claims description 16
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 15
- 150000001721 carbon Chemical group 0.000 claims description 15
- 239000003153 chemical reaction reagent Substances 0.000 claims description 14
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 claims description 13
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 claims description 12
- 102000005962 receptors Human genes 0.000 claims description 12
- 108020003175 receptors Proteins 0.000 claims description 12
- WPYMKLBDIGXBTP-UHFFFAOYSA-N Benzoic acid Natural products OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 claims description 11
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical group [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 claims description 11
- 125000000392 cycloalkenyl group Chemical group 0.000 claims description 11
- 230000001404 mediated effect Effects 0.000 claims description 11
- 238000002560 therapeutic procedure Methods 0.000 claims description 11
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims description 10
- 239000002246 antineoplastic agent Substances 0.000 claims description 10
- 229910052801 chlorine Inorganic materials 0.000 claims description 10
- 125000006552 (C3-C8) cycloalkyl group Chemical group 0.000 claims description 9
- 239000005711 Benzoic acid Substances 0.000 claims description 9
- 206010066476 Haematological malignancy Diseases 0.000 claims description 9
- DHXVGJBLRPWPCS-UHFFFAOYSA-N Tetrahydropyran Chemical compound C1CCOCC1 DHXVGJBLRPWPCS-UHFFFAOYSA-N 0.000 claims description 9
- 235000010233 benzoic acid Nutrition 0.000 claims description 9
- 230000015572 biosynthetic process Effects 0.000 claims description 9
- 208000035475 disorder Diseases 0.000 claims description 9
- 210000001035 gastrointestinal tract Anatomy 0.000 claims description 9
- 238000004519 manufacturing process Methods 0.000 claims description 9
- 238000003786 synthesis reaction Methods 0.000 claims description 9
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 9
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 claims description 8
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 claims description 8
- GAWIXWVDTYZWAW-UHFFFAOYSA-N C[CH]O Chemical group C[CH]O GAWIXWVDTYZWAW-UHFFFAOYSA-N 0.000 claims description 8
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 claims description 8
- 206010025323 Lymphomas Diseases 0.000 claims description 8
- 239000005557 antagonist Substances 0.000 claims description 8
- KQNPFQTWMSNSAP-UHFFFAOYSA-N isobutyric acid Chemical compound CC(C)C(O)=O KQNPFQTWMSNSAP-UHFFFAOYSA-N 0.000 claims description 8
- 230000036210 malignancy Effects 0.000 claims description 8
- 201000008968 osteosarcoma Diseases 0.000 claims description 8
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 claims description 8
- KISFEBPWFCGRGN-UHFFFAOYSA-M sodium;2-(2,4-dichlorophenoxy)ethyl sulfate Chemical compound [Na+].[O-]S(=O)(=O)OCCOC1=CC=C(Cl)C=C1Cl KISFEBPWFCGRGN-UHFFFAOYSA-M 0.000 claims description 8
- 229940124597 therapeutic agent Drugs 0.000 claims description 8
- 206010027406 Mesothelioma Diseases 0.000 claims description 7
- 206010039491 Sarcoma Diseases 0.000 claims description 7
- 208000032839 leukemia Diseases 0.000 claims description 7
- 125000000325 methylidene group Chemical group [H]C([H])=* 0.000 claims description 7
- 125000000229 (C1-C4)alkoxy group Chemical group 0.000 claims description 6
- 239000011230 binding agent Substances 0.000 claims description 6
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 claims description 6
- 238000010511 deprotection reaction Methods 0.000 claims description 6
- 125000004785 fluoromethoxy group Chemical group [H]C([H])(F)O* 0.000 claims description 6
- 210000004072 lung Anatomy 0.000 claims description 6
- 230000037361 pathway Effects 0.000 claims description 6
- 229940100198 alkylating agent Drugs 0.000 claims description 5
- 239000002168 alkylating agent Substances 0.000 claims description 5
- 230000006907 apoptotic process Effects 0.000 claims description 5
- 210000004556 brain Anatomy 0.000 claims description 5
- 238000013329 compounding Methods 0.000 claims description 5
- 238000002360 preparation method Methods 0.000 claims description 5
- 210000002307 prostate Anatomy 0.000 claims description 5
- 125000004191 (C1-C6) alkoxy group Chemical group 0.000 claims description 4
- 201000009030 Carcinoma Diseases 0.000 claims description 4
- 208000017604 Hodgkin disease Diseases 0.000 claims description 4
- 208000021519 Hodgkin lymphoma Diseases 0.000 claims description 4
- 208000010747 Hodgkins lymphoma Diseases 0.000 claims description 4
- 230000001919 adrenal effect Effects 0.000 claims description 4
- 230000001684 chronic effect Effects 0.000 claims description 4
- 230000000069 prophylactic effect Effects 0.000 claims description 4
- 238000001959 radiotherapy Methods 0.000 claims description 4
- QUXVLHIPFRLERT-UHFFFAOYSA-N 4-fluorooxane Chemical compound FC1CCOCC1 QUXVLHIPFRLERT-UHFFFAOYSA-N 0.000 claims description 3
- 208000011691 Burkitt lymphomas Diseases 0.000 claims description 3
- 230000033616 DNA repair Effects 0.000 claims description 3
- 102000004190 Enzymes Human genes 0.000 claims description 3
- 108090000790 Enzymes Proteins 0.000 claims description 3
- 206010018338 Glioma Diseases 0.000 claims description 3
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 claims description 3
- 206010029260 Neuroblastoma Diseases 0.000 claims description 3
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 3
- 102000004243 Tubulin Human genes 0.000 claims description 3
- 108090000704 Tubulin Proteins 0.000 claims description 3
- 239000000556 agonist Substances 0.000 claims description 3
- 230000003432 anti-folate effect Effects 0.000 claims description 3
- 229940127074 antifolate Drugs 0.000 claims description 3
- 210000000845 cartilage Anatomy 0.000 claims description 3
- 230000030833 cell death Effects 0.000 claims description 3
- 210000001072 colon Anatomy 0.000 claims description 3
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 claims description 3
- 239000003968 dna methyltransferase inhibitor Substances 0.000 claims description 3
- HESCAJZNRMSMJG-KKQRBIROSA-N epothilone A Chemical class C/C([C@@H]1C[C@@H]2O[C@@H]2CCC[C@@H]([C@@H]([C@@H](C)C(=O)C(C)(C)[C@@H](O)CC(=O)O1)O)C)=C\C1=CSC(C)=N1 HESCAJZNRMSMJG-KKQRBIROSA-N 0.000 claims description 3
- 239000004052 folic acid antagonist Substances 0.000 claims description 3
- 230000002496 gastric effect Effects 0.000 claims description 3
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 claims description 3
- 206010073071 hepatocellular carcinoma Diseases 0.000 claims description 3
- 231100000844 hepatocellular carcinoma Toxicity 0.000 claims description 3
- 206010024627 liposarcoma Diseases 0.000 claims description 3
- 230000003211 malignant effect Effects 0.000 claims description 3
- 201000001441 melanoma Diseases 0.000 claims description 3
- 125000002524 organometallic group Chemical group 0.000 claims description 3
- 230000019491 signal transduction Effects 0.000 claims description 3
- 210000004872 soft tissue Anatomy 0.000 claims description 3
- 208000011580 syndromic disease Diseases 0.000 claims description 3
- 230000008685 targeting Effects 0.000 claims description 3
- 238000012360 testing method Methods 0.000 claims description 3
- 125000004172 4-methoxyphenyl group Chemical group [H]C1=C([H])C(OC([H])([H])[H])=C([H])C([H])=C1* 0.000 claims description 2
- 206010003571 Astrocytoma Diseases 0.000 claims description 2
- 208000006332 Choriocarcinoma Diseases 0.000 claims description 2
- 206010010356 Congenital anomaly Diseases 0.000 claims description 2
- 229940124087 DNA topoisomerase II inhibitor Drugs 0.000 claims description 2
- 208000007033 Dysgerminoma Diseases 0.000 claims description 2
- 206010014967 Ependymoma Diseases 0.000 claims description 2
- 208000006168 Ewing Sarcoma Diseases 0.000 claims description 2
- 201000008808 Fibrosarcoma Diseases 0.000 claims description 2
- 206010017993 Gastrointestinal neoplasms Diseases 0.000 claims description 2
- 208000032612 Glial tumor Diseases 0.000 claims description 2
- 208000023661 Haematological disease Diseases 0.000 claims description 2
- 208000006937 Hydatidiform mole Diseases 0.000 claims description 2
- 208000000172 Medulloblastoma Diseases 0.000 claims description 2
- 101100400378 Mus musculus Marveld2 gene Proteins 0.000 claims description 2
- 208000005890 Neuroma Diseases 0.000 claims description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 claims description 2
- 206010035104 Pituitary tumour Diseases 0.000 claims description 2
- 206010057846 Primitive neuroectodermal tumour Diseases 0.000 claims description 2
- 229940079156 Proteasome inhibitor Drugs 0.000 claims description 2
- 201000000582 Retinoblastoma Diseases 0.000 claims description 2
- 201000010208 Seminoma Diseases 0.000 claims description 2
- 208000000453 Skin Neoplasms Diseases 0.000 claims description 2
- 208000021712 Soft tissue sarcoma Diseases 0.000 claims description 2
- 239000000317 Topoisomerase II Inhibitor Substances 0.000 claims description 2
- 201000006083 Xeroderma Pigmentosum Diseases 0.000 claims description 2
- RGHILYZRVFRRNK-UHFFFAOYSA-N anthracene-1,2-dione Chemical class C1=CC=C2C=C(C(C(=O)C=C3)=O)C3=CC2=C1 RGHILYZRVFRRNK-UHFFFAOYSA-N 0.000 claims description 2
- 229940045799 anthracyclines and related substance Drugs 0.000 claims description 2
- 230000002280 anti-androgenic effect Effects 0.000 claims description 2
- 230000000340 anti-metabolite Effects 0.000 claims description 2
- 239000000051 antiandrogen Substances 0.000 claims description 2
- 229940100197 antimetabolite Drugs 0.000 claims description 2
- 239000002256 antimetabolite Substances 0.000 claims description 2
- 239000003719 aurora kinase inhibitor Substances 0.000 claims description 2
- 210000003445 biliary tract Anatomy 0.000 claims description 2
- 210000000988 bone and bone Anatomy 0.000 claims description 2
- 210000003679 cervix uteri Anatomy 0.000 claims description 2
- 239000003937 drug carrier Substances 0.000 claims description 2
- 230000002124 endocrine Effects 0.000 claims description 2
- 210000004696 endometrium Anatomy 0.000 claims description 2
- 229930013356 epothilone Natural products 0.000 claims description 2
- 210000003020 exocrine pancreas Anatomy 0.000 claims description 2
- 210000000232 gallbladder Anatomy 0.000 claims description 2
- 210000004602 germ cell Anatomy 0.000 claims description 2
- 208000005017 glioblastoma Diseases 0.000 claims description 2
- 210000003128 head Anatomy 0.000 claims description 2
- 239000003481 heat shock protein 90 inhibitor Substances 0.000 claims description 2
- 201000002529 islet cell tumor Diseases 0.000 claims description 2
- 210000003734 kidney Anatomy 0.000 claims description 2
- 210000004185 liver Anatomy 0.000 claims description 2
- 208000023356 medullary thyroid gland carcinoma Diseases 0.000 claims description 2
- 206010027191 meningioma Diseases 0.000 claims description 2
- 210000000754 myometrium Anatomy 0.000 claims description 2
- 201000008026 nephroblastoma Diseases 0.000 claims description 2
- 208000007538 neurilemmoma Diseases 0.000 claims description 2
- 150000003833 nucleoside derivatives Chemical class 0.000 claims description 2
- ZWLPBLYKEWSWPD-UHFFFAOYSA-N o-toluenecarboxylic acid Natural products CC1=CC=CC=C1C(O)=O ZWLPBLYKEWSWPD-UHFFFAOYSA-N 0.000 claims description 2
- ILUJQPXNXACGAN-UHFFFAOYSA-N ortho-methoxybenzoic acid Natural products COC1=CC=CC=C1C(O)=O ILUJQPXNXACGAN-UHFFFAOYSA-N 0.000 claims description 2
- 210000001672 ovary Anatomy 0.000 claims description 2
- 208000022102 pancreatic neuroendocrine neoplasm Diseases 0.000 claims description 2
- 210000003899 penis Anatomy 0.000 claims description 2
- 230000002093 peripheral effect Effects 0.000 claims description 2
- 210000004303 peritoneum Anatomy 0.000 claims description 2
- 239000003207 proteasome inhibitor Substances 0.000 claims description 2
- 238000002626 targeted therapy Methods 0.000 claims description 2
- DKPFODGZWDEEBT-QFIAKTPHSA-N taxane Chemical class C([C@]1(C)CCC[C@@H](C)[C@H]1C1)C[C@H]2[C@H](C)CC[C@@H]1C2(C)C DKPFODGZWDEEBT-QFIAKTPHSA-N 0.000 claims description 2
- 210000001550 testis Anatomy 0.000 claims description 2
- 210000001685 thyroid gland Anatomy 0.000 claims description 2
- 239000003053 toxin Substances 0.000 claims description 2
- 231100000765 toxin Toxicity 0.000 claims description 2
- 108700012359 toxins Proteins 0.000 claims description 2
- 210000001635 urinary tract Anatomy 0.000 claims description 2
- 210000001215 vagina Anatomy 0.000 claims description 2
- 210000003905 vulva Anatomy 0.000 claims description 2
- 125000004356 hydroxy functional group Chemical group O* 0.000 claims 21
- 208000020816 lung neoplasm Diseases 0.000 claims 3
- XTBAPWCYTNCZTO-UHFFFAOYSA-N 1H-isoindolone Natural products C1=CC=C2C(=O)N=CC2=C1 XTBAPWCYTNCZTO-UHFFFAOYSA-N 0.000 claims 2
- CVICEEPAFUYBJG-UHFFFAOYSA-N 5-chloro-2,2-difluoro-1,3-benzodioxole Chemical group C1=C(Cl)C=C2OC(F)(F)OC2=C1 CVICEEPAFUYBJG-UHFFFAOYSA-N 0.000 claims 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims 2
- 201000005202 lung cancer Diseases 0.000 claims 2
- 239000000333 selective estrogen receptor modulator Substances 0.000 claims 2
- 229940095743 selective estrogen receptor modulator Drugs 0.000 claims 2
- HJTAZXHBEBIQQX-UHFFFAOYSA-N 1,5-bis(chloromethyl)naphthalene Chemical compound C1=CC=C2C(CCl)=CC=CC2=C1CCl HJTAZXHBEBIQQX-UHFFFAOYSA-N 0.000 claims 1
- MCTWTZJPVLRJOU-UHFFFAOYSA-N 1-methyl-1H-imidazole Chemical compound CN1C=CN=C1 MCTWTZJPVLRJOU-UHFFFAOYSA-N 0.000 claims 1
- PAMIQIKDUOTOBW-UHFFFAOYSA-N 1-methylpiperidine Chemical compound CN1CCCCC1 PAMIQIKDUOTOBW-UHFFFAOYSA-N 0.000 claims 1
- 229940122124 17,20 lyase inhibitor Drugs 0.000 claims 1
- 102100032187 Androgen receptor Human genes 0.000 claims 1
- 208000003174 Brain Neoplasms Diseases 0.000 claims 1
- 206010006187 Breast cancer Diseases 0.000 claims 1
- 208000026310 Breast neoplasm Diseases 0.000 claims 1
- 206010009944 Colon cancer Diseases 0.000 claims 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims 1
- 102000002004 Cytochrome P-450 Enzyme System Human genes 0.000 claims 1
- 108010015742 Cytochrome P-450 Enzyme System Proteins 0.000 claims 1
- 229940123780 DNA topoisomerase I inhibitor Drugs 0.000 claims 1
- 102000009058 Death Domain Receptors Human genes 0.000 claims 1
- 108010049207 Death Domain Receptors Proteins 0.000 claims 1
- 229940102550 Estrogen receptor antagonist Drugs 0.000 claims 1
- 108091006027 G proteins Proteins 0.000 claims 1
- 102000030782 GTP binding Human genes 0.000 claims 1
- 108091000058 GTP-Binding Proteins 0.000 claims 1
- 239000000579 Gonadotropin-Releasing Hormone Substances 0.000 claims 1
- 102000014150 Interferons Human genes 0.000 claims 1
- 108010050904 Interferons Proteins 0.000 claims 1
- 102000015696 Interleukins Human genes 0.000 claims 1
- 108010063738 Interleukins Proteins 0.000 claims 1
- 208000015850 Malignant peritoneal mesothelioma Diseases 0.000 claims 1
- 206010033963 Parathyroid tumour Diseases 0.000 claims 1
- 101000857870 Squalus acanthias Gonadoliberin Proteins 0.000 claims 1
- 102000001854 Steroid 17-alpha-Hydroxylase Human genes 0.000 claims 1
- 108010015330 Steroid 17-alpha-Hydroxylase Proteins 0.000 claims 1
- 108010017842 Telomerase Proteins 0.000 claims 1
- 206010043276 Teratoma Diseases 0.000 claims 1
- ATJFFYVFTNAWJD-UHFFFAOYSA-N Tin Chemical compound [Sn] ATJFFYVFTNAWJD-UHFFFAOYSA-N 0.000 claims 1
- 239000000365 Topoisomerase I Inhibitor Substances 0.000 claims 1
- 241000863480 Vinca Species 0.000 claims 1
- 125000003342 alkenyl group Chemical group 0.000 claims 1
- 108010080146 androgen receptors Proteins 0.000 claims 1
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 claims 1
- 239000003972 antineoplastic antibiotic Substances 0.000 claims 1
- 239000003886 aromatase inhibitor Substances 0.000 claims 1
- 229940046844 aromatase inhibitors Drugs 0.000 claims 1
- GOLCXWYRSKYTSP-UHFFFAOYSA-N arsenic trioxide Inorganic materials O1[As]2O[As]1O2 GOLCXWYRSKYTSP-UHFFFAOYSA-N 0.000 claims 1
- 208000002458 carcinoid tumor Diseases 0.000 claims 1
- 229940011871 estrogen Drugs 0.000 claims 1
- 239000000262 estrogen Substances 0.000 claims 1
- 239000003862 glucocorticoid Substances 0.000 claims 1
- XLXSAKCOAKORKW-AQJXLSMYSA-N gonadorelin Chemical compound C([C@@H](C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)NCC(N)=O)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)C1=CC=C(O)C=C1 XLXSAKCOAKORKW-AQJXLSMYSA-N 0.000 claims 1
- 229940035638 gonadotropin-releasing hormone Drugs 0.000 claims 1
- 239000005556 hormone Substances 0.000 claims 1
- 229940088597 hormone Drugs 0.000 claims 1
- 238000009169 immunotherapy Methods 0.000 claims 1
- 229940047124 interferons Drugs 0.000 claims 1
- 229940047122 interleukins Drugs 0.000 claims 1
- 239000003771 matrix metalloproteinase inhibitor Substances 0.000 claims 1
- 229940121386 matrix metalloproteinase inhibitor Drugs 0.000 claims 1
- 210000003739 neck Anatomy 0.000 claims 1
- 239000003528 protein farnesyltransferase inhibitor Substances 0.000 claims 1
- 238000011363 radioimmunotherapy Methods 0.000 claims 1
- 230000003637 steroidlike Effects 0.000 claims 1
- 150000003431 steroids Chemical class 0.000 claims 1
- 229940037128 systemic glucocorticoids Drugs 0.000 claims 1
- 229940021747 therapeutic vaccine Drugs 0.000 claims 1
- 230000006663 ubiquitin-proteasome pathway Effects 0.000 claims 1
- 230000002401 inhibitory effect Effects 0.000 abstract 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 92
- 108050002772 E3 ubiquitin-protein ligase Mdm2 Proteins 0.000 description 82
- 102000012199 E3 ubiquitin-protein ligase Mdm2 Human genes 0.000 description 82
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 72
- 101000721661 Homo sapiens Cellular tumor antigen p53 Proteins 0.000 description 70
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 54
- 125000000217 alkyl group Chemical group 0.000 description 52
- 239000000203 mixture Substances 0.000 description 52
- 108090000623 proteins and genes Proteins 0.000 description 28
- 210000004027 cell Anatomy 0.000 description 23
- 230000000694 effects Effects 0.000 description 22
- 238000009472 formulation Methods 0.000 description 22
- 102000004169 proteins and genes Human genes 0.000 description 22
- 239000000460 chlorine Substances 0.000 description 21
- 235000018102 proteins Nutrition 0.000 description 21
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 20
- 150000001204 N-oxides Chemical class 0.000 description 19
- 150000002148 esters Chemical class 0.000 description 19
- 239000002904 solvent Substances 0.000 description 19
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 18
- 125000004429 atom Chemical group 0.000 description 18
- 125000001072 heteroaryl group Chemical group 0.000 description 18
- 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 17
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 16
- 239000002253 acid Substances 0.000 description 16
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 16
- 239000000543 intermediate Substances 0.000 description 15
- 239000000546 pharmaceutical excipient Substances 0.000 description 14
- 238000000926 separation method Methods 0.000 description 14
- 239000000126 substance Substances 0.000 description 14
- 230000002354 daily effect Effects 0.000 description 13
- 230000001965 increasing effect Effects 0.000 description 13
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 13
- 239000007787 solid Substances 0.000 description 13
- 239000000243 solution Substances 0.000 description 13
- 230000003321 amplification Effects 0.000 description 12
- 238000001802 infusion Methods 0.000 description 12
- 239000000463 material Substances 0.000 description 12
- 238000003199 nucleic acid amplification method Methods 0.000 description 12
- 230000004044 response Effects 0.000 description 12
- 239000003826 tablet Substances 0.000 description 12
- 210000001519 tissue Anatomy 0.000 description 12
- MHCVCKDNQYMGEX-UHFFFAOYSA-N 1,1'-biphenyl;phenoxybenzene Chemical group C1=CC=CC=C1C1=CC=CC=C1.C=1C=CC=CC=1OC1=CC=CC=C1 MHCVCKDNQYMGEX-UHFFFAOYSA-N 0.000 description 11
- 125000004432 carbon atom Chemical group C* 0.000 description 11
- 238000006243 chemical reaction Methods 0.000 description 11
- 230000003287 optical effect Effects 0.000 description 11
- 239000001301 oxygen Substances 0.000 description 11
- KDLHZDBZIXYQEI-UHFFFAOYSA-N palladium Substances [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 11
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 10
- 239000004480 active ingredient Substances 0.000 description 10
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 10
- 239000002775 capsule Substances 0.000 description 10
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 10
- 229910052805 deuterium Inorganic materials 0.000 description 10
- 230000002018 overexpression Effects 0.000 description 10
- 125000004193 piperazinyl group Chemical group 0.000 description 10
- 229920000642 polymer Polymers 0.000 description 10
- 229920006395 saturated elastomer Polymers 0.000 description 10
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 10
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 9
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 9
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 9
- 125000002619 bicyclic group Chemical group 0.000 description 9
- 239000011248 coating agent Substances 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 9
- 125000002757 morpholinyl group Chemical group 0.000 description 9
- 230000002829 reductive effect Effects 0.000 description 9
- 230000003827 upregulation Effects 0.000 description 9
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 9
- 150000001412 amines Chemical class 0.000 description 8
- 230000027455 binding Effects 0.000 description 8
- 230000010261 cell growth Effects 0.000 description 8
- 238000000576 coating method Methods 0.000 description 8
- 238000002425 crystallisation Methods 0.000 description 8
- 239000003085 diluting agent Substances 0.000 description 8
- 230000001976 improved effect Effects 0.000 description 8
- 238000002955 isolation Methods 0.000 description 8
- 239000000314 lubricant Substances 0.000 description 8
- 125000002950 monocyclic group Chemical group 0.000 description 8
- 150000007523 nucleic acids Chemical class 0.000 description 8
- 125000003566 oxetanyl group Chemical group 0.000 description 8
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 7
- SIKJAQJRHWYJAI-UHFFFAOYSA-N Indole Chemical compound C1=CC=C2NC=CC2=C1 SIKJAQJRHWYJAI-UHFFFAOYSA-N 0.000 description 7
- 230000004913 activation Effects 0.000 description 7
- 230000033115 angiogenesis Effects 0.000 description 7
- 125000006615 aromatic heterocyclic group Chemical group 0.000 description 7
- 125000002393 azetidinyl group Chemical group 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 238000001514 detection method Methods 0.000 description 7
- 239000007884 disintegrant Substances 0.000 description 7
- 238000009396 hybridization Methods 0.000 description 7
- 239000007788 liquid Substances 0.000 description 7
- 229940002612 prodrug Drugs 0.000 description 7
- 239000000651 prodrug Substances 0.000 description 7
- 239000000047 product Substances 0.000 description 7
- 238000000746 purification Methods 0.000 description 7
- 239000000523 sample Substances 0.000 description 7
- 239000006104 solid solution Substances 0.000 description 7
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 7
- 125000006555 (C3-C5) cycloalkyl group Chemical group 0.000 description 6
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 6
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 6
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 6
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 6
- 108010079351 Tumor Suppressor Protein p14ARF Proteins 0.000 description 6
- 102100033254 Tumor suppressor ARF Human genes 0.000 description 6
- 230000002159 abnormal effect Effects 0.000 description 6
- 125000003277 amino group Chemical group 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- 239000003054 catalyst Substances 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 239000002552 dosage form Substances 0.000 description 6
- 125000004185 ester group Chemical group 0.000 description 6
- 230000014509 gene expression Effects 0.000 description 6
- 125000002636 imidazolinyl group Chemical group 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 230000007246 mechanism Effects 0.000 description 6
- 108020004999 messenger RNA Proteins 0.000 description 6
- 108020004707 nucleic acids Proteins 0.000 description 6
- 102000039446 nucleic acids Human genes 0.000 description 6
- 238000003752 polymerase chain reaction Methods 0.000 description 6
- 125000006239 protecting group Chemical group 0.000 description 6
- 125000002755 pyrazolinyl group Chemical group 0.000 description 6
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 6
- 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 5
- 230000004544 DNA amplification Effects 0.000 description 5
- YLQBMQCUIZJEEH-UHFFFAOYSA-N Furan Chemical compound C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 5
- 206010027476 Metastases Diseases 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 5
- 150000001299 aldehydes Chemical class 0.000 description 5
- 229940041181 antineoplastic drug Drugs 0.000 description 5
- 125000002843 carboxylic acid group Chemical group 0.000 description 5
- 238000004296 chiral HPLC Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 125000000524 functional group Chemical group 0.000 description 5
- 230000002068 genetic effect Effects 0.000 description 5
- 230000005764 inhibitory process Effects 0.000 description 5
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 5
- 239000003446 ligand Substances 0.000 description 5
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 5
- 230000009401 metastasis Effects 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 239000002105 nanoparticle Substances 0.000 description 5
- 125000003367 polycyclic group Chemical group 0.000 description 5
- 239000000843 powder Substances 0.000 description 5
- 239000002243 precursor Substances 0.000 description 5
- 230000002062 proliferating effect Effects 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 235000000346 sugar Nutrition 0.000 description 5
- 239000011593 sulfur Substances 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 4
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 4
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 4
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 4
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 4
- 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 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
- 108010010803 Gelatin Proteins 0.000 description 4
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 4
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 4
- 208000034578 Multiple myelomas Diseases 0.000 description 4
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 4
- KYRVNWMVYQXFEU-UHFFFAOYSA-N Nocodazole Chemical compound C1=C2NC(NC(=O)OC)=NC2=CC=C1C(=O)C1=CC=CS1 KYRVNWMVYQXFEU-UHFFFAOYSA-N 0.000 description 4
- 206010035226 Plasma cell myeloma Diseases 0.000 description 4
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 4
- 238000010240 RT-PCR analysis Methods 0.000 description 4
- 101710183280 Topoisomerase Proteins 0.000 description 4
- 238000010521 absorption reaction Methods 0.000 description 4
- 150000007513 acids Chemical class 0.000 description 4
- 230000009471 action Effects 0.000 description 4
- 150000001408 amides Chemical class 0.000 description 4
- HUMNYLRZRPPJDN-UHFFFAOYSA-N benzaldehyde Chemical compound O=CC1=CC=CC=C1 HUMNYLRZRPPJDN-UHFFFAOYSA-N 0.000 description 4
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 4
- WGQKYBSKWIADBV-UHFFFAOYSA-N benzylamine Chemical compound NCC1=CC=CC=C1 WGQKYBSKWIADBV-UHFFFAOYSA-N 0.000 description 4
- 210000004369 blood Anatomy 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 239000004067 bulking agent Substances 0.000 description 4
- 150000001768 cations Chemical class 0.000 description 4
- 238000004587 chromatography analysis Methods 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 239000013078 crystal Substances 0.000 description 4
- 230000008025 crystallization Effects 0.000 description 4
- 125000002147 dimethylamino group Chemical group [H]C([H])([H])N(*)C([H])([H])[H] 0.000 description 4
- 238000003821 enantio-separation Methods 0.000 description 4
- XOZIUKBZLSUILX-GIQCAXHBSA-N epothilone D Chemical compound O1C(=O)C[C@H](O)C(C)(C)C(=O)[C@H](C)[C@@H](O)[C@@H](C)CCC\C(C)=C/C[C@H]1C(\C)=C\C1=CSC(C)=N1 XOZIUKBZLSUILX-GIQCAXHBSA-N 0.000 description 4
- 238000004108 freeze drying Methods 0.000 description 4
- 239000008273 gelatin Substances 0.000 description 4
- 229920000159 gelatin Polymers 0.000 description 4
- 235000019322 gelatine Nutrition 0.000 description 4
- 235000011852 gelatine desserts Nutrition 0.000 description 4
- 238000010914 gene-directed enzyme pro-drug therapy Methods 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 150000002430 hydrocarbons Chemical group 0.000 description 4
- 238000007901 in situ hybridization Methods 0.000 description 4
- 125000000468 ketone group Chemical group 0.000 description 4
- 238000004949 mass spectrometry Methods 0.000 description 4
- 230000002503 metabolic effect Effects 0.000 description 4
- 229910052751 metal Inorganic materials 0.000 description 4
- 239000002184 metal Substances 0.000 description 4
- 125000004184 methoxymethyl group Chemical group [H]C([H])([H])OC([H])([H])* 0.000 description 4
- 125000005968 oxazolinyl group Chemical group 0.000 description 4
- 239000012071 phase Substances 0.000 description 4
- 239000003755 preservative agent Substances 0.000 description 4
- 125000003072 pyrazolidinyl group Chemical group 0.000 description 4
- 238000001953 recrystallisation Methods 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 238000012216 screening Methods 0.000 description 4
- 239000004094 surface-active agent Substances 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- 238000011269 treatment regimen Methods 0.000 description 4
- 230000004614 tumor growth Effects 0.000 description 4
- 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 4
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 3
- MRUWJENAYHTDQG-UHFFFAOYSA-N 4H-pyran Chemical compound C1C=COC=C1 MRUWJENAYHTDQG-UHFFFAOYSA-N 0.000 description 3
- OGWKCGZFUXNPDA-CFWMRBGOSA-N 5j49q6b70f 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=O)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 OGWKCGZFUXNPDA-CFWMRBGOSA-N 0.000 description 3
- 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 description 3
- 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 3
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 3
- CWHUFRVAEUJCEF-UHFFFAOYSA-N BKM120 Chemical compound C1=NC(N)=CC(C(F)(F)F)=C1C1=CC(N2CCOCC2)=NC(N2CCOCC2)=N1 CWHUFRVAEUJCEF-UHFFFAOYSA-N 0.000 description 3
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 3
- UGFAIRIUMAVXCW-UHFFFAOYSA-N Carbon monoxide Chemical compound [O+]#[C-] UGFAIRIUMAVXCW-UHFFFAOYSA-N 0.000 description 3
- 231100000277 DNA damage Toxicity 0.000 description 3
- 230000005778 DNA damage Effects 0.000 description 3
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 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 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 3
- MZRVEZGGRBJDDB-UHFFFAOYSA-N N-Butyllithium Chemical compound [Li]CCCC MZRVEZGGRBJDDB-UHFFFAOYSA-N 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- YTPLMLYBLZKORZ-UHFFFAOYSA-N Thiophene Chemical compound C=1C=CSC=1 YTPLMLYBLZKORZ-UHFFFAOYSA-N 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 125000004423 acyloxy group Chemical group 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 235000001014 amino acid Nutrition 0.000 description 3
- 150000001413 amino acids Chemical group 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 230000003466 anti-cipated effect Effects 0.000 description 3
- 239000003963 antioxidant agent Substances 0.000 description 3
- 238000003782 apoptosis assay Methods 0.000 description 3
- 239000002585 base Substances 0.000 description 3
- 230000009286 beneficial effect Effects 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- ZADPBFCGQRWHPN-UHFFFAOYSA-N boronic acid Chemical compound OBO ZADPBFCGQRWHPN-UHFFFAOYSA-N 0.000 description 3
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 3
- 229910002091 carbon monoxide Inorganic materials 0.000 description 3
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 3
- 238000002512 chemotherapy Methods 0.000 description 3
- 238000002648 combination therapy Methods 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000003745 diagnosis Methods 0.000 description 3
- 125000005043 dihydropyranyl group Chemical group O1C(CCC=C1)* 0.000 description 3
- 125000005056 dihydrothiazolyl group Chemical group S1C(NC=C1)* 0.000 description 3
- 125000000532 dioxanyl group Chemical group 0.000 description 3
- 238000012377 drug delivery Methods 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 239000000945 filler Substances 0.000 description 3
- 239000000796 flavoring agent Substances 0.000 description 3
- 239000003979 granulating agent Substances 0.000 description 3
- PZOUSPYUWWUPPK-UHFFFAOYSA-N indole Natural products CC1=CC=CC2=C1C=CN2 PZOUSPYUWWUPPK-UHFFFAOYSA-N 0.000 description 3
- RKJUIXBNRJVNHR-UHFFFAOYSA-N indolenine Natural products C1=CC=C2CC=NC2=C1 RKJUIXBNRJVNHR-UHFFFAOYSA-N 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 125000000842 isoxazolyl group Chemical group 0.000 description 3
- 239000008101 lactose Substances 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 3
- 239000008108 microcrystalline cellulose Substances 0.000 description 3
- 229940016286 microcrystalline cellulose Drugs 0.000 description 3
- 125000006574 non-aromatic ring group Chemical group 0.000 description 3
- 239000002773 nucleotide Substances 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 231100000590 oncogenic Toxicity 0.000 description 3
- 230000002246 oncogenic effect Effects 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- 125000004430 oxygen atom Chemical group O* 0.000 description 3
- 229910052763 palladium Inorganic materials 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- LHNIIDJUOCFXAP-UHFFFAOYSA-N pictrelisib Chemical compound C1CN(S(=O)(=O)C)CCN1CC1=CC2=NC(C=3C=4C=NNC=4C=CC=3)=NC(N3CCOCC3)=C2S1 LHNIIDJUOCFXAP-UHFFFAOYSA-N 0.000 description 3
- 239000000049 pigment Substances 0.000 description 3
- 210000002381 plasma Anatomy 0.000 description 3
- 229910000027 potassium carbonate Inorganic materials 0.000 description 3
- 230000003389 potentiating effect Effects 0.000 description 3
- 230000005522 programmed cell death Effects 0.000 description 3
- 230000002035 prolonged effect Effects 0.000 description 3
- 125000004309 pyranyl group Chemical group O1C(C=CC=C1)* 0.000 description 3
- 125000000714 pyrimidinyl group Chemical group 0.000 description 3
- 125000004929 pyrrolidonyl group Chemical group N1(C(CCC1)=O)* 0.000 description 3
- 125000000168 pyrrolyl group Chemical group 0.000 description 3
- XSCHRSMBECNVNS-UHFFFAOYSA-N quinoxaline Chemical compound N1=CC=NC2=CC=CC=C21 XSCHRSMBECNVNS-UHFFFAOYSA-N 0.000 description 3
- 239000007962 solid dispersion Substances 0.000 description 3
- 239000007909 solid dosage form Substances 0.000 description 3
- 238000010561 standard procedure Methods 0.000 description 3
- 210000002784 stomach Anatomy 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 125000003831 tetrazolyl group Chemical group 0.000 description 3
- 125000002769 thiazolinyl group Chemical group 0.000 description 3
- 231100000331 toxic Toxicity 0.000 description 3
- 230000002588 toxic effect Effects 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 238000013518 transcription Methods 0.000 description 3
- 230000035897 transcription Effects 0.000 description 3
- 230000002103 transcriptional effect Effects 0.000 description 3
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 3
- 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 3
- 239000008215 water for injection Substances 0.000 description 3
- KSOVGRCOLZZTPF-QMKUDKLTSA-N (1s,2s,3r,4r)-3-[[5-fluoro-2-[3-methyl-4-(4-methylpiperazin-1-yl)anilino]pyrimidin-4-yl]amino]bicyclo[2.2.1]hept-5-ene-2-carboxamide Chemical compound N([C@H]1[C@H]([C@@]2([H])C[C@@]1(C=C2)[H])C(N)=O)C(C(=CN=1)F)=NC=1NC(C=C1C)=CC=C1N1CCN(C)CC1 KSOVGRCOLZZTPF-QMKUDKLTSA-N 0.000 description 2
- GRZXWCHAXNAUHY-NSISKUIASA-N (2S)-2-(4-chlorophenyl)-1-[4-[(5R,7R)-7-hydroxy-5-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl]-1-piperazinyl]-3-(propan-2-ylamino)-1-propanone Chemical compound C1([C@H](C(=O)N2CCN(CC2)C=2C=3[C@H](C)C[C@@H](O)C=3N=CN=2)CNC(C)C)=CC=C(Cl)C=C1 GRZXWCHAXNAUHY-NSISKUIASA-N 0.000 description 2
- SVNJBEMPMKWDCO-KCHLEUMXSA-N (2s)-2-[[(2s)-3-carboxy-2-[[2-[[(2s)-5-(diaminomethylideneamino)-2-[[4-oxo-4-[[4-(4-oxo-8-phenylchromen-2-yl)morpholin-4-ium-4-yl]methoxy]butanoyl]amino]pentanoyl]amino]acetyl]amino]propanoyl]amino]-3-hydroxypropanoate Chemical compound C=1C(=O)C2=CC=CC(C=3C=CC=CC=3)=C2OC=1[N+]1(COC(=O)CCC(=O)N[C@@H](CCCNC(=N)N)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CO)C([O-])=O)CCOCC1 SVNJBEMPMKWDCO-KCHLEUMXSA-N 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- UWYZHKAOTLEWKK-UHFFFAOYSA-N 1,2,3,4-tetrahydroisoquinoline Chemical compound C1=CC=C2CNCCC2=C1 UWYZHKAOTLEWKK-UHFFFAOYSA-N 0.000 description 2
- LBUJPTNKIBCYBY-UHFFFAOYSA-N 1,2,3,4-tetrahydroquinoline Chemical compound C1=CC=C2CCCNC2=C1 LBUJPTNKIBCYBY-UHFFFAOYSA-N 0.000 description 2
- CSNIZNHTOVFARY-UHFFFAOYSA-N 1,2-benzothiazole Chemical compound C1=CC=C2C=NSC2=C1 CSNIZNHTOVFARY-UHFFFAOYSA-N 0.000 description 2
- KTZQTRPPVKQPFO-UHFFFAOYSA-N 1,2-benzoxazole Chemical compound C1=CC=C2C=NOC2=C1 KTZQTRPPVKQPFO-UHFFFAOYSA-N 0.000 description 2
- FTNJQNQLEGKTGD-UHFFFAOYSA-N 1,3-benzodioxole Chemical compound C1=CC=C2OCOC2=C1 FTNJQNQLEGKTGD-UHFFFAOYSA-N 0.000 description 2
- FCEHBMOGCRZNNI-UHFFFAOYSA-N 1-benzothiophene Chemical compound C1=CC=C2SC=CC2=C1 FCEHBMOGCRZNNI-UHFFFAOYSA-N 0.000 description 2
- MFJCPDOGFAYSTF-UHFFFAOYSA-N 1H-isochromene Chemical compound C1=CC=C2COC=CC2=C1 MFJCPDOGFAYSTF-UHFFFAOYSA-N 0.000 description 2
- HBEDSQVIWPRPAY-UHFFFAOYSA-N 2,3-dihydrobenzofuran Chemical compound C1=CC=C2OCCC2=C1 HBEDSQVIWPRPAY-UHFFFAOYSA-N 0.000 description 2
- MGADZUXDNSDTHW-UHFFFAOYSA-N 2H-pyran Chemical compound C1OC=CC=C1 MGADZUXDNSDTHW-UHFFFAOYSA-N 0.000 description 2
- CMLFRMDBDNHMRA-UHFFFAOYSA-N 2h-1,2-benzoxazine Chemical compound C1=CC=C2C=CNOC2=C1 CMLFRMDBDNHMRA-UHFFFAOYSA-N 0.000 description 2
- NHQDETIJWKXCTC-UHFFFAOYSA-N 3-chloroperbenzoic acid Chemical compound OOC(=O)C1=CC=CC(Cl)=C1 NHQDETIJWKXCTC-UHFFFAOYSA-N 0.000 description 2
- KAUMDYDNLPQESC-UHFFFAOYSA-N 3-hydroxy-2,3-dihydroisoindol-1-one Chemical compound C1=CC=C2C(O)NC(=O)C2=C1 KAUMDYDNLPQESC-UHFFFAOYSA-N 0.000 description 2
- YEJRWHAVMIAJKC-UHFFFAOYSA-N 4-Butyrolactone Chemical compound O=C1CCCO1 YEJRWHAVMIAJKC-UHFFFAOYSA-N 0.000 description 2
- GYLDXIAOMVERTK-UHFFFAOYSA-N 5-(4-amino-1-propan-2-yl-3-pyrazolo[3,4-d]pyrimidinyl)-1,3-benzoxazol-2-amine Chemical compound C12=C(N)N=CN=C2N(C(C)C)N=C1C1=CC=C(OC(N)=N2)C2=C1 GYLDXIAOMVERTK-UHFFFAOYSA-N 0.000 description 2
- 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 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
- PBCZSGKMGDDXIJ-HQCWYSJUSA-N 7-hydroxystaurosporine Chemical compound N([C@H](O)C1=C2C3=CC=CC=C3N3C2=C24)C(=O)C1=C2C1=CC=CC=C1N4[C@H]1C[C@@H](NC)[C@@H](OC)[C@]3(C)O1 PBCZSGKMGDDXIJ-HQCWYSJUSA-N 0.000 description 2
- PBCZSGKMGDDXIJ-UHFFFAOYSA-N 7beta-hydroxystaurosporine Natural products C12=C3N4C5=CC=CC=C5C3=C3C(O)NC(=O)C3=C2C2=CC=CC=C2N1C1CC(NC)C(OC)C4(C)O1 PBCZSGKMGDDXIJ-UHFFFAOYSA-N 0.000 description 2
- ACCFLVVUVBJNGT-AWEZNQCLSA-N 8-[5-(2-hydroxypropan-2-yl)pyridin-3-yl]-1-[(2s)-2-methoxypropyl]-3-methylimidazo[4,5-c]quinolin-2-one Chemical compound CN1C(=O)N(C[C@H](C)OC)C(C2=C3)=C1C=NC2=CC=C3C1=CN=CC(C(C)(C)O)=C1 ACCFLVVUVBJNGT-AWEZNQCLSA-N 0.000 description 2
- GBJVVSCPOBPEIT-UHFFFAOYSA-N AZT-1152 Chemical compound N=1C=NC2=CC(OCCCN(CC)CCOP(O)(O)=O)=CC=C2C=1NC(=NN1)C=C1CC(=O)NC1=CC=CC(F)=C1 GBJVVSCPOBPEIT-UHFFFAOYSA-N 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 2
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 2
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 2
- PAYRUJLWNCNPSJ-UHFFFAOYSA-N Aniline Chemical compound NC1=CC=CC=C1 PAYRUJLWNCNPSJ-UHFFFAOYSA-N 0.000 description 2
- 101100221077 Arabidopsis thaliana CML12 gene Proteins 0.000 description 2
- 201000001320 Atherosclerosis Diseases 0.000 description 2
- 208000003950 B-cell lymphoma Diseases 0.000 description 2
- QULDDKSCVCJTPV-UHFFFAOYSA-N BIIB021 Chemical compound COC1=C(C)C=NC(CN2C3=NC(N)=NC(Cl)=C3N=C2)=C1C QULDDKSCVCJTPV-UHFFFAOYSA-N 0.000 description 2
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 2
- KXDHJXZQYSOELW-UHFFFAOYSA-M Carbamate Chemical compound NC([O-])=O KXDHJXZQYSOELW-UHFFFAOYSA-M 0.000 description 2
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 2
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 2
- 208000031404 Chromosome Aberrations Diseases 0.000 description 2
- 229920000858 Cyclodextrin Polymers 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- HKVAMNSJSFKALM-GKUWKFKPSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[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 HKVAMNSJSFKALM-GKUWKFKPSA-N 0.000 description 2
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 2
- 239000007821 HATU Substances 0.000 description 2
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 2
- 229930194542 Keto Natural products 0.000 description 2
- 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 2
- 238000005004 MAS NMR spectroscopy Methods 0.000 description 2
- 229940083338 MDM2 inhibitor Drugs 0.000 description 2
- BAVYZALUXZFZLV-UHFFFAOYSA-N Methylamine Chemical compound NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 2
- 238000005481 NMR spectroscopy Methods 0.000 description 2
- 208000022873 Ocular disease Diseases 0.000 description 2
- 108091008606 PDGF receptors Proteins 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 Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- GLUUGHFHXGJENI-UHFFFAOYSA-N Piperazine Chemical compound C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 description 2
- 102000011653 Platelet-Derived Growth Factor Receptors Human genes 0.000 description 2
- KYQCOXFCLRTKLS-UHFFFAOYSA-N Pyrazine Chemical compound C1=CN=CC=N1 KYQCOXFCLRTKLS-UHFFFAOYSA-N 0.000 description 2
- KAESVJOAVNADME-UHFFFAOYSA-N Pyrrole Chemical compound C=1C=CNC=1 KAESVJOAVNADME-UHFFFAOYSA-N 0.000 description 2
- SMWDFEZZVXVKRB-UHFFFAOYSA-N Quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 2
- 238000003559 RNA-seq method Methods 0.000 description 2
- 206010038933 Retinopathy of prematurity Diseases 0.000 description 2
- 206010070834 Sensitisation Diseases 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- 101100054666 Streptomyces halstedii sch3 gene Proteins 0.000 description 2
- FZWLAAWBMGSTSO-UHFFFAOYSA-N Thiazole Chemical compound C1=CSC=N1 FZWLAAWBMGSTSO-UHFFFAOYSA-N 0.000 description 2
- 108010078814 Tumor Suppressor Protein p53 Proteins 0.000 description 2
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 description 2
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 description 2
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 2
- 108091008605 VEGF receptors Proteins 0.000 description 2
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 2
- 230000005856 abnormality Effects 0.000 description 2
- 230000002378 acidificating effect Effects 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
- 230000001154 acute effect Effects 0.000 description 2
- ORILYTVJVMAKLC-UHFFFAOYSA-N adamantane Chemical compound C1C(C2)CC3CC1CC2C3 ORILYTVJVMAKLC-UHFFFAOYSA-N 0.000 description 2
- 208000009956 adenocarcinoma Diseases 0.000 description 2
- 206010064930 age-related macular degeneration Diseases 0.000 description 2
- 125000005907 alkyl ester group Chemical group 0.000 description 2
- 208000026935 allergic disease Diseases 0.000 description 2
- 229950010817 alvocidib Drugs 0.000 description 2
- BIIVYFLTOXDAOV-YVEFUNNKSA-N alvocidib Chemical compound O[C@@H]1CN(C)CC[C@@H]1C1=C(O)C=C(O)C2=C1OC(C=1C(=CC=CC=1)Cl)=CC2=O BIIVYFLTOXDAOV-YVEFUNNKSA-N 0.000 description 2
- 150000003863 ammonium salts Chemical class 0.000 description 2
- 125000000129 anionic group Chemical group 0.000 description 2
- RDOXTESZEPMUJZ-UHFFFAOYSA-N anisole Chemical compound COC1=CC=CC=C1 RDOXTESZEPMUJZ-UHFFFAOYSA-N 0.000 description 2
- 230000008485 antagonism Effects 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 238000010913 antigen-directed enzyme pro-drug therapy Methods 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 238000003491 array Methods 0.000 description 2
- 206010003246 arthritis Diseases 0.000 description 2
- 230000001363 autoimmune Effects 0.000 description 2
- 230000001042 autoregulative effect Effects 0.000 description 2
- 229960002756 azacitidine Drugs 0.000 description 2
- AAMATCKFMHVIDO-UHFFFAOYSA-N azane;1h-pyrrole Chemical compound N.C=1C=CNC=1 AAMATCKFMHVIDO-UHFFFAOYSA-N 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- IOJUPLGTWVMSFF-UHFFFAOYSA-N benzothiazole Chemical compound C1=CC=C2SC=NC2=C1 IOJUPLGTWVMSFF-UHFFFAOYSA-N 0.000 description 2
- LUFPJJNWMYZRQE-UHFFFAOYSA-N benzylsulfanylmethylbenzene Chemical compound C=1C=CC=CC=1CSCC1=CC=CC=C1 LUFPJJNWMYZRQE-UHFFFAOYSA-N 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 239000012472 biological sample Substances 0.000 description 2
- 238000001574 biopsy Methods 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 229950003628 buparlisib Drugs 0.000 description 2
- HQABUPZFAYXKJW-UHFFFAOYSA-N butan-1-amine Chemical compound CCCCN HQABUPZFAYXKJW-UHFFFAOYSA-N 0.000 description 2
- 238000010804 cDNA synthesis Methods 0.000 description 2
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical class C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 2
- KBPLFHHGFOOTCA-UHFFFAOYSA-N caprylic alcohol Natural products CCCCCCCCO KBPLFHHGFOOTCA-UHFFFAOYSA-N 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 230000025084 cell cycle arrest Effects 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 239000007910 chewable tablet Substances 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- VZWXIQHBIQLMPN-UHFFFAOYSA-N chromane Chemical compound C1=CC=C2CCCOC2=C1 VZWXIQHBIQLMPN-UHFFFAOYSA-N 0.000 description 2
- QZHPTGXQGDFGEN-UHFFFAOYSA-N chromene Chemical compound C1=CC=C2C=C[CH]OC2=C1 QZHPTGXQGDFGEN-UHFFFAOYSA-N 0.000 description 2
- 208000020832 chronic kidney disease Diseases 0.000 description 2
- 201000010902 chronic myelomonocytic leukemia Diseases 0.000 description 2
- 229960004316 cisplatin Drugs 0.000 description 2
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 2
- WDDPHFBMKLOVOX-AYQXTPAHSA-N clofarabine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@@H]1F WDDPHFBMKLOVOX-AYQXTPAHSA-N 0.000 description 2
- 229960000928 clofarabine Drugs 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 238000001816 cooling Methods 0.000 description 2
- 239000006184 cosolvent Substances 0.000 description 2
- 150000003950 cyclic amides Chemical group 0.000 description 2
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 2
- 229960000684 cytarabine Drugs 0.000 description 2
- 230000006378 damage Effects 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
- XOZIUKBZLSUILX-UHFFFAOYSA-N desoxyepothilone B Natural products O1C(=O)CC(O)C(C)(C)C(=O)C(C)C(O)C(C)CCCC(C)=CCC1C(C)=CC1=CSC(C)=N1 XOZIUKBZLSUILX-UHFFFAOYSA-N 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 238000002405 diagnostic procedure Methods 0.000 description 2
- 125000004852 dihydrofuranyl group Chemical group O1C(CC=C1)* 0.000 description 2
- 125000005057 dihydrothienyl group Chemical group S1C(CC=C1)* 0.000 description 2
- 239000006185 dispersion Substances 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 229960004679 doxorubicin Drugs 0.000 description 2
- 239000008298 dragée Substances 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 150000002085 enols Chemical class 0.000 description 2
- 229940088598 enzyme Drugs 0.000 description 2
- HESCAJZNRMSMJG-HGYUPSKWSA-N epothilone A Natural products O=C1[C@H](C)[C@H](O)[C@H](C)CCC[C@H]2O[C@H]2C[C@@H](/C(=C\c2nc(C)sc2)/C)OC(=O)C[C@H](O)C1(C)C HESCAJZNRMSMJG-HGYUPSKWSA-N 0.000 description 2
- 230000032050 esterification Effects 0.000 description 2
- 238000005886 esterification reaction Methods 0.000 description 2
- 235000019439 ethyl acetate Nutrition 0.000 description 2
- 229960002949 fluorouracil Drugs 0.000 description 2
- 235000013355 food flavoring agent Nutrition 0.000 description 2
- 235000003599 food sweetener Nutrition 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 2
- 150000004677 hydrates Chemical class 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
- 229960001330 hydroxycarbamide Drugs 0.000 description 2
- 239000012535 impurity Substances 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 125000003387 indolinyl group Chemical group N1(CCC2=CC=CC=C12)* 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 150000007529 inorganic bases Chemical class 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 230000009545 invasion Effects 0.000 description 2
- PNDPGZBMCMUPRI-UHFFFAOYSA-N iodine Chemical compound II PNDPGZBMCMUPRI-UHFFFAOYSA-N 0.000 description 2
- HEBMCVBCEDMUOF-UHFFFAOYSA-N isochromane Chemical compound C1=CC=C2COCCC2=C1 HEBMCVBCEDMUOF-UHFFFAOYSA-N 0.000 description 2
- AWJUIBRHMBBTKR-UHFFFAOYSA-N isoquinoline Chemical compound C1=NC=CC2=CC=CC=C21 AWJUIBRHMBBTKR-UHFFFAOYSA-N 0.000 description 2
- ZLTPDFXIESTBQG-UHFFFAOYSA-N isothiazole Chemical compound C=1C=NSC=1 ZLTPDFXIESTBQG-UHFFFAOYSA-N 0.000 description 2
- 125000001786 isothiazolyl group Chemical group 0.000 description 2
- 208000017169 kidney disease Diseases 0.000 description 2
- 229940043355 kinase inhibitor Drugs 0.000 description 2
- 231100000518 lethal Toxicity 0.000 description 2
- 230000001665 lethal effect Effects 0.000 description 2
- 238000004811 liquid chromatography Methods 0.000 description 2
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 2
- QBZXOWQOWPHHRA-UHFFFAOYSA-N lithium;ethane Chemical compound [Li+].[CH2-]C QBZXOWQOWPHHRA-UHFFFAOYSA-N 0.000 description 2
- 229940124302 mTOR inhibitor Drugs 0.000 description 2
- 208000002780 macular degeneration Diseases 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 239000003628 mammalian target of rapamycin inhibitor Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- 229960000485 methotrexate Drugs 0.000 description 2
- CFCUWKMKBJTWLW-BKHRDMLASA-N mithramycin Chemical compound 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=1C)O[C@@H]1O[C@H](C)[C@@H](O)[C@H](O[C@@H]2O[C@H](C)[C@H](O)[C@H](O[C@@H]3O[C@H](C)[C@@H](O)[C@@](C)(O)C3)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@@H](O)[C@H](O)[C@@H](C)O1 CFCUWKMKBJTWLW-BKHRDMLASA-N 0.000 description 2
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 description 2
- 239000012457 nonaqueous media Substances 0.000 description 2
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 2
- 230000005853 oncogenic activation Effects 0.000 description 2
- 231100000822 oral exposure Toxicity 0.000 description 2
- 150000007530 organic bases Chemical class 0.000 description 2
- 239000003960 organic solvent Substances 0.000 description 2
- 230000003204 osmotic effect Effects 0.000 description 2
- 125000001715 oxadiazolyl group Chemical group 0.000 description 2
- 125000002971 oxazolyl group Chemical group 0.000 description 2
- 150000002923 oximes Chemical class 0.000 description 2
- 229940118537 p53 inhibitor Drugs 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- SZFPYBIJACMNJV-UHFFFAOYSA-N perifosine Chemical compound CCCCCCCCCCCCCCCCCCOP([O-])(=O)OC1CC[N+](C)(C)CC1 SZFPYBIJACMNJV-UHFFFAOYSA-N 0.000 description 2
- 230000002085 persistent effect Effects 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 2
- 230000035790 physiological processes and functions Effects 0.000 description 2
- 239000004014 plasticizer Substances 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- 229960003171 plicamycin Drugs 0.000 description 2
- 238000012877 positron emission topography Methods 0.000 description 2
- 238000004237 preparative chromatography Methods 0.000 description 2
- 238000004262 preparative liquid chromatography Methods 0.000 description 2
- WGYKZJWCGVVSQN-UHFFFAOYSA-N propylamine Chemical compound CCCN WGYKZJWCGVVSQN-UHFFFAOYSA-N 0.000 description 2
- 230000001681 protective effect Effects 0.000 description 2
- 125000001042 pteridinyl group Chemical group N1=C(N=CC2=NC=CN=C12)* 0.000 description 2
- 125000003373 pyrazinyl group Chemical group 0.000 description 2
- 125000002098 pyridazinyl group Chemical group 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 230000000979 retarding effect Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 229950009216 sapanisertib Drugs 0.000 description 2
- BTIHMVBBUGXLCJ-OAHLLOKOSA-N seliciclib Chemical compound C=12N=CN(C(C)C)C2=NC(N[C@@H](CO)CC)=NC=1NCC1=CC=CC=C1 BTIHMVBBUGXLCJ-OAHLLOKOSA-N 0.000 description 2
- 230000001235 sensitizing effect Effects 0.000 description 2
- BLGWHBSBBJNKJO-UHFFFAOYSA-N serabelisib Chemical compound C=1C=C2OC(N)=NC2=CC=1C(=CN12)C=CC1=NC=C2C(=O)N1CCOCC1 BLGWHBSBBJNKJO-UHFFFAOYSA-N 0.000 description 2
- 238000000371 solid-state nuclear magnetic resonance spectroscopy Methods 0.000 description 2
- 230000035882 stress Effects 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 229940124530 sulfonamide Drugs 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 239000003765 sweetening agent Substances 0.000 description 2
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 2
- 239000006068 taste-masking agent Substances 0.000 description 2
- URLYINUFLXOMHP-HTVVRFAVSA-N tcn-p Chemical compound C=12C3=NC=NC=1N(C)N=C(N)C2=CN3[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H]1O URLYINUFLXOMHP-HTVVRFAVSA-N 0.000 description 2
- 150000003509 tertiary alcohols Chemical class 0.000 description 2
- 125000005958 tetrahydrothienyl group Chemical group 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
- 125000004568 thiomorpholinyl group Chemical group 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 125000004306 triazinyl group Chemical group 0.000 description 2
- 125000001425 triazolyl group Chemical group 0.000 description 2
- JKNHZOAONLKYQL-UHFFFAOYSA-K tribromoindigane Chemical compound Br[In](Br)Br JKNHZOAONLKYQL-UHFFFAOYSA-K 0.000 description 2
- 229950003873 triciribine Drugs 0.000 description 2
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 229940124676 vascular endothelial growth factor receptor Drugs 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 229960003862 vemurafenib Drugs 0.000 description 2
- 229960004528 vincristine Drugs 0.000 description 2
- 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 2
- 239000001993 wax Substances 0.000 description 2
- BMKDZUISNHGIBY-ZETCQYMHSA-N (+)-dexrazoxane Chemical compound C([C@H](C)N1CC(=O)NC(=O)C1)N1CC(=O)NC(=O)C1 BMKDZUISNHGIBY-ZETCQYMHSA-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
- YOVVNQKCSKSHKT-HNNXBMFYSA-N (2s)-1-[4-[[2-(2-aminopyrimidin-5-yl)-7-methyl-4-morpholin-4-ylthieno[3,2-d]pyrimidin-6-yl]methyl]piperazin-1-yl]-2-hydroxypropan-1-one Chemical compound C1CN(C(=O)[C@@H](O)C)CCN1CC1=C(C)C2=NC(C=3C=NC(N)=NC=3)=NC(N3CCOCC3)=C2S1 YOVVNQKCSKSHKT-HNNXBMFYSA-N 0.000 description 1
- MHFUWOIXNMZFIW-WNQIDUERSA-N (2s)-2-hydroxypropanoic acid;n-[4-[4-(4-methylpiperazin-1-yl)-6-[(5-methyl-1h-pyrazol-3-yl)amino]pyrimidin-2-yl]sulfanylphenyl]cyclopropanecarboxamide Chemical compound C[C@H](O)C(O)=O.C1CN(C)CCN1C1=CC(NC2=NNC(C)=C2)=NC(SC=2C=CC(NC(=O)C3CC3)=CC=2)=N1 MHFUWOIXNMZFIW-WNQIDUERSA-N 0.000 description 1
- AWOKSNNHYRGYIA-UHFFFAOYSA-N (4,5-dimethoxy-2-nitrophenyl)methyl carbamate Chemical compound COC1=CC(COC(N)=O)=C([N+]([O-])=O)C=C1OC AWOKSNNHYRGYIA-UHFFFAOYSA-N 0.000 description 1
- DSSYKIVIOFKYAU-OMNKOJBGSA-N (4s)-4,7,7-trimethylbicyclo[2.2.1]heptan-3-one Chemical compound C1C[C@]2(C)C(=O)CC1C2(C)C DSSYKIVIOFKYAU-OMNKOJBGSA-N 0.000 description 1
- NWXMGUDVXFXRIG-WESIUVDSSA-N (4s,4as,5as,6s,12ar)-4-(dimethylamino)-1,6,10,11,12a-pentahydroxy-6-methyl-3,12-dioxo-4,4a,5,5a-tetrahydrotetracene-2-carboxamide Chemical compound C1=CC=C2[C@](O)(C)[C@H]3C[C@H]4[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]4(O)C(=O)C3=C(O)C2=C1O NWXMGUDVXFXRIG-WESIUVDSSA-N 0.000 description 1
- SWDZPNJZKUGIIH-QQTULTPQSA-N (5z)-n-ethyl-5-(4-hydroxy-6-oxo-3-propan-2-ylcyclohexa-2,4-dien-1-ylidene)-4-[4-(morpholin-4-ylmethyl)phenyl]-2h-1,2-oxazole-3-carboxamide Chemical compound O1NC(C(=O)NCC)=C(C=2C=CC(CN3CCOCC3)=CC=2)\C1=C1/C=C(C(C)C)C(O)=CC1=O SWDZPNJZKUGIIH-QQTULTPQSA-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
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 1
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- JYEUMXHLPRZUAT-UHFFFAOYSA-N 1,2,3-triazine Chemical compound C1=CN=NN=C1 JYEUMXHLPRZUAT-UHFFFAOYSA-N 0.000 description 1
- 125000001399 1,2,3-triazolyl group Chemical group N1N=NC(=C1)* 0.000 description 1
- 125000001376 1,2,4-triazolyl group Chemical group N1N=C(N=C1)* 0.000 description 1
- BCMCBBGGLRIHSE-UHFFFAOYSA-N 1,3-benzoxazole Chemical compound C1=CC=C2OC=NC2=C1 BCMCBBGGLRIHSE-UHFFFAOYSA-N 0.000 description 1
- FLBAYUMRQUHISI-UHFFFAOYSA-N 1,8-naphthyridine Chemical compound N1=CC=CC2=CC=CN=C21 FLBAYUMRQUHISI-UHFFFAOYSA-N 0.000 description 1
- 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 1
- 125000006017 1-propenyl group Chemical group 0.000 description 1
- HYZJCKYKOHLVJF-UHFFFAOYSA-N 1H-benzimidazole Chemical compound C1=CC=C2NC=NC2=C1 HYZJCKYKOHLVJF-UHFFFAOYSA-N 0.000 description 1
- BAXOFTOLAUCFNW-UHFFFAOYSA-N 1H-indazole Chemical compound C1=CC=C2C=NNC2=C1 BAXOFTOLAUCFNW-UHFFFAOYSA-N 0.000 description 1
- SVUOLADPCWQTTE-UHFFFAOYSA-N 1h-1,2-benzodiazepine Chemical compound N1N=CC=CC2=CC=CC=C12 SVUOLADPCWQTTE-UHFFFAOYSA-N 0.000 description 1
- AMFYRKOUWBAGHV-UHFFFAOYSA-N 1h-pyrazolo[4,3-b]pyridine Chemical compound C1=CN=C2C=NNC2=C1 AMFYRKOUWBAGHV-UHFFFAOYSA-N 0.000 description 1
- QPLJYAKLSCXZSF-UHFFFAOYSA-N 2,2,2-trichloroethyl carbamate Chemical compound NC(=O)OCC(Cl)(Cl)Cl QPLJYAKLSCXZSF-UHFFFAOYSA-N 0.000 description 1
- MXILHVKNAYIRPJ-UHFFFAOYSA-N 2,3,7,7a-tetrahydro-1h-triazolo[4,5-b]pyrazine Chemical compound N1C=CN=C2NNNC21 MXILHVKNAYIRPJ-UHFFFAOYSA-N 0.000 description 1
- KEQTWHPMSVAFDA-UHFFFAOYSA-N 2,3-dihydro-1h-pyrazole Chemical compound C1NNC=C1 KEQTWHPMSVAFDA-UHFFFAOYSA-N 0.000 description 1
- TXQPXJKRNHJWAX-UHFFFAOYSA-N 2-(3-aminopropylamino)ethylsulfanylphosphonic acid;trihydrate Chemical compound O.O.O.NCCCNCCSP(O)(O)=O TXQPXJKRNHJWAX-UHFFFAOYSA-N 0.000 description 1
- PIMQWRZWLQKKBJ-SFHVURJKSA-N 2-[(2S)-1-[3-ethyl-7-[(1-oxido-3-pyridin-1-iumyl)methylamino]-5-pyrazolo[1,5-a]pyrimidinyl]-2-piperidinyl]ethanol Chemical compound C=1C(N2[C@@H](CCCC2)CCO)=NC2=C(CC)C=NN2C=1NCC1=CC=C[N+]([O-])=C1 PIMQWRZWLQKKBJ-SFHVURJKSA-N 0.000 description 1
- RTQWWZBSTRGEAV-PKHIMPSTSA-N 2-[[(2s)-2-[bis(carboxymethyl)amino]-3-[4-(methylcarbamoylamino)phenyl]propyl]-[2-[bis(carboxymethyl)amino]propyl]amino]acetic acid Chemical compound CNC(=O)NC1=CC=C(C[C@@H](CN(CC(C)N(CC(O)=O)CC(O)=O)CC(O)=O)N(CC(O)=O)CC(O)=O)C=C1 RTQWWZBSTRGEAV-PKHIMPSTSA-N 0.000 description 1
- RFIAWPRFJJTTJI-UHFFFAOYSA-N 2-[[1-(hydroxymethyl)cyclopropyl]methoxy]-3H-isoindol-1-one Chemical compound OCC1(CC1)CON1C(C2=CC=CC=C2C1)=O RFIAWPRFJJTTJI-UHFFFAOYSA-N 0.000 description 1
- WJUNQSYQHHIVFX-UHFFFAOYSA-N 2-amino-4-[2,4-dichloro-5-(2-pyrrolidin-1-ylethoxy)phenyl]-n-ethylthieno[2,3-d]pyrimidine-6-carboxamide Chemical compound N1=C(N)N=C2SC(C(=O)NCC)=CC2=C1C(C(=CC=1Cl)Cl)=CC=1OCCN1CCCC1 WJUNQSYQHHIVFX-UHFFFAOYSA-N 0.000 description 1
- MWYDSXOGIBMAET-UHFFFAOYSA-N 2-amino-N-[7-methoxy-8-(3-morpholin-4-ylpropoxy)-2,3-dihydro-1H-imidazo[1,2-c]quinazolin-5-ylidene]pyrimidine-5-carboxamide Chemical compound NC1=NC=C(C=N1)C(=O)N=C1N=C2C(=C(C=CC2=C2N1CCN2)OCCCN1CCOCC1)OC MWYDSXOGIBMAET-UHFFFAOYSA-N 0.000 description 1
- 125000000022 2-aminoethyl group Chemical group [H]C([*])([H])C([H])([H])N([H])[H] 0.000 description 1
- UXGVMFHEKMGWMA-UHFFFAOYSA-N 2-benzofuran Chemical compound C1=CC=CC2=COC=C21 UXGVMFHEKMGWMA-UHFFFAOYSA-N 0.000 description 1
- BEUQXVWXFDOSAQ-UHFFFAOYSA-N 2-methyl-2-[4-[2-(5-methyl-2-propan-2-yl-1,2,4-triazol-3-yl)-5,6-dihydroimidazo[1,2-d][1,4]benzoxazepin-9-yl]pyrazol-1-yl]propanamide Chemical compound CC(C)N1N=C(C)N=C1C1=CN(CCOC=2C3=CC=C(C=2)C2=CN(N=C2)C(C)(C)C(N)=O)C3=N1 BEUQXVWXFDOSAQ-UHFFFAOYSA-N 0.000 description 1
- CMRFJAMFEWYMKE-UHFFFAOYSA-N 2-oxaadamantane Chemical compound C1C(O2)CC3CC1CC2C3 CMRFJAMFEWYMKE-UHFFFAOYSA-N 0.000 description 1
- 125000001494 2-propynyl group Chemical group [H]C#CC([H])([H])* 0.000 description 1
- VHMICKWLTGFITH-UHFFFAOYSA-N 2H-isoindole Chemical compound C1=CC=CC2=CNC=C21 VHMICKWLTGFITH-UHFFFAOYSA-N 0.000 description 1
- UMZCLZPXPCNKML-UHFFFAOYSA-N 2h-imidazo[4,5-d][1,3]thiazole Chemical compound C1=NC2=NCSC2=N1 UMZCLZPXPCNKML-UHFFFAOYSA-N 0.000 description 1
- WPWNEKFMGCWNPR-UHFFFAOYSA-N 3,4-dihydro-2h-thiochromene Chemical compound C1=CC=C2CCCSC2=C1 WPWNEKFMGCWNPR-UHFFFAOYSA-N 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
- HDBQZGJWHMCXIL-UHFFFAOYSA-N 3,7-dihydropurine-2-thione Chemical compound SC1=NC=C2NC=NC2=N1 HDBQZGJWHMCXIL-UHFFFAOYSA-N 0.000 description 1
- AXRCEOKUDYDWLF-UHFFFAOYSA-N 3-(1-methyl-3-indolyl)-4-[1-[1-(2-pyridinylmethyl)-4-piperidinyl]-3-indolyl]pyrrole-2,5-dione Chemical compound C12=CC=CC=C2N(C)C=C1C(C(NC1=O)=O)=C1C(C1=CC=CC=C11)=CN1C(CC1)CCN1CC1=CC=CC=N1 AXRCEOKUDYDWLF-UHFFFAOYSA-N 0.000 description 1
- XDADYJOYIMVXTH-UHFFFAOYSA-N 3h-imidazo[1,2-a]imidazole Chemical compound C1=CN2CC=NC2=N1 XDADYJOYIMVXTH-UHFFFAOYSA-N 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
- YCVAGNVTZICLNM-UHFFFAOYSA-N 4,5,6,7-tetrahydro-1-benzofuran Chemical compound C1CCCC2=C1C=CO2 YCVAGNVTZICLNM-UHFFFAOYSA-N 0.000 description 1
- WJRRGYBTGDJBFX-UHFFFAOYSA-N 4-(2-methyl-3-propan-2-yl-4-imidazolyl)-N-(4-methylsulfonylphenyl)-2-pyrimidinamine Chemical compound CC(C)N1C(C)=NC=C1C1=CC=NC(NC=2C=CC(=CC=2)S(C)(=O)=O)=N1 WJRRGYBTGDJBFX-UHFFFAOYSA-N 0.000 description 1
- HCFRWBBJISAZNK-UHFFFAOYSA-N 4-Hydroxycyclohexylcarboxylic acid Chemical compound OC1CCC(C(O)=O)CC1 HCFRWBBJISAZNK-UHFFFAOYSA-N 0.000 description 1
- QCQCHGYLTSGIGX-GHXANHINSA-N 4-[[(3ar,5ar,5br,7ar,9s,11ar,11br,13as)-5a,5b,8,8,11a-pentamethyl-3a-[(5-methylpyridine-3-carbonyl)amino]-2-oxo-1-propan-2-yl-4,5,6,7,7a,9,10,11,11b,12,13,13a-dodecahydro-3h-cyclopenta[a]chrysen-9-yl]oxy]-2,2-dimethyl-4-oxobutanoic acid Chemical compound N([C@@]12CC[C@@]3(C)[C@]4(C)CC[C@H]5C(C)(C)[C@@H](OC(=O)CC(C)(C)C(O)=O)CC[C@]5(C)[C@H]4CC[C@@H]3C1=C(C(C2)=O)C(C)C)C(=O)C1=CN=CC(C)=C1 QCQCHGYLTSGIGX-GHXANHINSA-N 0.000 description 1
- KDKUVYLMPJIGKA-UHFFFAOYSA-N 4-[[5-amino-1-[(2,6-difluorophenyl)-oxomethyl]-1,2,4-triazol-3-yl]amino]benzenesulfonamide Chemical compound N=1N(C(=O)C=2C(=CC=CC=2F)F)C(N)=NC=1NC1=CC=C(S(N)(=O)=O)C=C1 KDKUVYLMPJIGKA-UHFFFAOYSA-N 0.000 description 1
- MJIALGDLOLWBRQ-MRVPVSSYSA-N 4-[[5-bromo-4-[[(2r)-1-hydroxypropan-2-yl]amino]pyrimidin-2-yl]amino]benzenesulfonamide Chemical compound C1=C(Br)C(N[C@@H](CO)C)=NC(NC=2C=CC(=CC=2)S(N)(=O)=O)=N1 MJIALGDLOLWBRQ-MRVPVSSYSA-N 0.000 description 1
- ZLHFILGSQDJULK-UHFFFAOYSA-N 4-[[9-chloro-7-(2-fluoro-6-methoxyphenyl)-5H-pyrimido[5,4-d][2]benzazepin-2-yl]amino]-2-methoxybenzoic acid Chemical compound C1=C(C(O)=O)C(OC)=CC(NC=2N=C3C4=CC=C(Cl)C=C4C(=NCC3=CN=2)C=2C(=CC=CC=2F)OC)=C1 ZLHFILGSQDJULK-UHFFFAOYSA-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
- FJKROLUGYXJWQN-UHFFFAOYSA-N 4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- GDRVFDDBLLKWRI-UHFFFAOYSA-N 4H-quinolizine Chemical compound C1=CC=CN2CC=CC=C21 GDRVFDDBLLKWRI-UHFFFAOYSA-N 0.000 description 1
- BVUBLIHUYSRSFE-LBPRGKRZSA-N 5-fluoro-3-phenyl-2-[(1S)-1-(7H-purin-6-ylamino)ethyl]quinazolin-4-one Chemical compound N1=CN=C2NC=NC2=C1N[C@@H](C)C1=NC2=CC=CC(=C2C(N1C1=CC=CC=C1)=O)F BVUBLIHUYSRSFE-LBPRGKRZSA-N 0.000 description 1
- WYWHKKSPHMUBEB-UHFFFAOYSA-N 6-Mercaptoguanine Natural products N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 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
- RHXHGRAEPCAFML-UHFFFAOYSA-N 7-cyclopentyl-n,n-dimethyl-2-[(5-piperazin-1-ylpyridin-2-yl)amino]pyrrolo[2,3-d]pyrimidine-6-carboxamide Chemical compound N1=C2N(C3CCCC3)C(C(=O)N(C)C)=CC2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 RHXHGRAEPCAFML-UHFFFAOYSA-N 0.000 description 1
- SJVQHLPISAIATJ-ZDUSSCGKSA-N 8-chloro-2-phenyl-3-[(1S)-1-(7H-purin-6-ylamino)ethyl]-1-isoquinolinone Chemical compound C1([C@@H](NC=2C=3N=CNC=3N=CN=2)C)=CC2=CC=CC(Cl)=C2C(=O)N1C1=CC=CC=C1 SJVQHLPISAIATJ-ZDUSSCGKSA-N 0.000 description 1
- ZZOKVYOCRSMTSS-UHFFFAOYSA-N 9h-fluoren-9-ylmethyl carbamate Chemical compound C1=CC=C2C(COC(=O)N)C3=CC=CC=C3C2=C1 ZZOKVYOCRSMTSS-UHFFFAOYSA-N 0.000 description 1
- 230000035495 ADMET Effects 0.000 description 1
- BUROJSBIWGDYCN-GAUTUEMISA-N AP 23573 Chemical compound C1C[C@@H](OP(C)(C)=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 BUROJSBIWGDYCN-GAUTUEMISA-N 0.000 description 1
- 101150034533 ATIC gene Proteins 0.000 description 1
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 1
- 206010000830 Acute leukaemia Diseases 0.000 description 1
- 208000036762 Acute promyelocytic leukaemia Diseases 0.000 description 1
- 208000003200 Adenoma Diseases 0.000 description 1
- 108010000239 Aequorin Proteins 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 229930195573 Amycin Natural products 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 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 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical compound OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 1
- 201000004569 Blindness Diseases 0.000 description 1
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 1
- 206010058354 Bronchioloalveolar carcinoma Diseases 0.000 description 1
- AREGYBNPHOFGAD-UHFFFAOYSA-N CC(C(N(CC1=CC=CC=C11)C1=O)=O)O Chemical compound CC(C(N(CC1=CC=CC=C11)C1=O)=O)O AREGYBNPHOFGAD-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 102000000584 Calmodulin Human genes 0.000 description 1
- 108010041952 Calmodulin Proteins 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
- 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 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
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 206010007275 Carcinoid tumour Diseases 0.000 description 1
- 206010007572 Cardiac hypertrophy Diseases 0.000 description 1
- 208000006029 Cardiomegaly Diseases 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- KZBUYRJDOAKODT-UHFFFAOYSA-N Chlorine Chemical compound ClCl KZBUYRJDOAKODT-UHFFFAOYSA-N 0.000 description 1
- 208000005243 Chondrosarcoma Diseases 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- PTOAARAWEBMLNO-KVQBGUIXSA-N Cladribine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@H]1C[C@H](O)[C@@H](CO)O1 PTOAARAWEBMLNO-KVQBGUIXSA-N 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 229920002785 Croscarmellose sodium Polymers 0.000 description 1
- 102000013701 Cyclin-Dependent Kinase 4 Human genes 0.000 description 1
- 108010025464 Cyclin-Dependent Kinase 4 Proteins 0.000 description 1
- 108010009392 Cyclin-Dependent Kinase Inhibitor p16 Proteins 0.000 description 1
- 102100021906 Cyclin-O Human genes 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-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
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- 238000000018 DNA microarray Methods 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- ZBNZXTGUTAYRHI-UHFFFAOYSA-N Dasatinib Chemical compound C=1C(N2CCN(CCO)CC2)=NC(C)=NC=1NC(S1)=NC=C1C(=O)NC1=C(C)C=CC=C1Cl ZBNZXTGUTAYRHI-UHFFFAOYSA-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
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 1
- 201000004624 Dermatitis Diseases 0.000 description 1
- 208000008334 Dermatofibrosarcoma Diseases 0.000 description 1
- 206010057070 Dermatofibrosarcoma protuberans Diseases 0.000 description 1
- 206010012689 Diabetic retinopathy Diseases 0.000 description 1
- XBPCUCUWBYBCDP-UHFFFAOYSA-N Dicyclohexylamine Chemical compound C1CCCCC1NC1CCCCC1 XBPCUCUWBYBCDP-UHFFFAOYSA-N 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 206010062805 Dysplastic naevus Diseases 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 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
- XOZIUKBZLSUILX-SDMHVBBESA-N Epothilone D Natural products O=C1[C@H](C)[C@@H](O)[C@@H](C)CCC/C(/C)=C/C[C@@H](/C(=C\c2nc(C)sc2)/C)OC(=O)C[C@H](O)C1(C)C XOZIUKBZLSUILX-SDMHVBBESA-N 0.000 description 1
- 208000032027 Essential Thrombocythemia Diseases 0.000 description 1
- 239000001856 Ethyl cellulose Substances 0.000 description 1
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 1
- JOYRKODLDBILNP-UHFFFAOYSA-N Ethyl urethane Chemical compound CCOC(N)=O JOYRKODLDBILNP-UHFFFAOYSA-N 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- 229920003134 Eudragit® polymer Polymers 0.000 description 1
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 1
- 102000004315 Forkhead Transcription Factors Human genes 0.000 description 1
- 108090000852 Forkhead Transcription Factors Proteins 0.000 description 1
- RVAQIUULWULRNW-UHFFFAOYSA-N Ganetespib Chemical compound C1=C(O)C(C(C)C)=CC(C=2N(C(O)=NN=2)C=2C=C3C=CN(C)C3=CC=2)=C1O RVAQIUULWULRNW-UHFFFAOYSA-N 0.000 description 1
- 206010051066 Gastrointestinal stromal tumour Diseases 0.000 description 1
- 206010018364 Glomerulonephritis Diseases 0.000 description 1
- 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 1
- 238000003747 Grignard reaction Methods 0.000 description 1
- 102000009465 Growth Factor Receptors Human genes 0.000 description 1
- 108010009202 Growth Factor Receptors Proteins 0.000 description 1
- 208000001258 Hemangiosarcoma Diseases 0.000 description 1
- 101000897441 Homo sapiens Cyclin-O Proteins 0.000 description 1
- 101000777293 Homo sapiens Serine/threonine-protein kinase Chk1 Proteins 0.000 description 1
- 244000025221 Humulus lupulus Species 0.000 description 1
- 235000008694 Humulus lupulus Nutrition 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 1
- 206010048643 Hypereosinophilic syndrome Diseases 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010021143 Hypoxia Diseases 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
- 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 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 208000007766 Kaposi sarcoma Diseases 0.000 description 1
- 206010023347 Keratoacanthoma Diseases 0.000 description 1
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 1
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 1
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 1
- 239000005511 L01XE05 - Sorafenib Substances 0.000 description 1
- 239000002067 L01XE06 - Dasatinib Substances 0.000 description 1
- 239000002136 L01XE07 - Lapatinib Substances 0.000 description 1
- 239000005536 L01XE08 - Nilotinib Substances 0.000 description 1
- 239000003798 L01XE11 - Pazopanib Substances 0.000 description 1
- 239000002118 L01XE12 - Vandetanib Substances 0.000 description 1
- CZQHHVNHHHRRDU-UHFFFAOYSA-N LY294002 Chemical compound C1=CC=C2C(=O)C=C(N3CCOCC3)OC2=C1C1=CC=CC=C1 CZQHHVNHHHRRDU-UHFFFAOYSA-N 0.000 description 1
- XNRVGTHNYCNCFF-UHFFFAOYSA-N Lapatinib ditosylate monohydrate Chemical compound O.CC1=CC=C(S(O)(=O)=O)C=C1.CC1=CC=C(S(O)(=O)=O)C=C1.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 XNRVGTHNYCNCFF-UHFFFAOYSA-N 0.000 description 1
- HLFSDGLLUJUHTE-SNVBAGLBSA-N Levamisole Chemical compound C1([C@H]2CN3CCSC3=N2)=CC=CC=C1 HLFSDGLLUJUHTE-SNVBAGLBSA-N 0.000 description 1
- 239000012448 Lithium borohydride Substances 0.000 description 1
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 1
- 208000005777 Lupus Nephritis Diseases 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 229940124640 MK-2206 Drugs 0.000 description 1
- ULDXWLCXEDXJGE-UHFFFAOYSA-N MK-2206 Chemical compound C=1C=C(C=2C(=CC=3C=4N(C(NN=4)=O)C=CC=3N=2)C=2C=CC=CC=2)C=CC=1C1(N)CCC1 ULDXWLCXEDXJGE-UHFFFAOYSA-N 0.000 description 1
- 206010025421 Macule Diseases 0.000 description 1
- 206010064912 Malignant transformation Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 208000025205 Mantle-Cell Lymphoma Diseases 0.000 description 1
- XOGTZOOQQBDUSI-UHFFFAOYSA-M Mesna Chemical compound [Na+].[O-]S(=O)(=O)CCS XOGTZOOQQBDUSI-UHFFFAOYSA-M 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 1
- VVQNEPGJFQJSBK-UHFFFAOYSA-N Methyl methacrylate Chemical compound COC(=O)C(C)=C VVQNEPGJFQJSBK-UHFFFAOYSA-N 0.000 description 1
- 238000006751 Mitsunobu reaction Methods 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 241000238367 Mya arenaria Species 0.000 description 1
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 1
- 208000033833 Myelomonocytic Chronic Leukemia 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
- FOFDIMHVKGYHRU-UHFFFAOYSA-N N-(1,3-benzodioxol-5-ylmethyl)-4-(4-benzofuro[3,2-d]pyrimidinyl)-1-piperazinecarbothioamide Chemical compound C12=CC=CC=C2OC2=C1N=CN=C2N(CC1)CCN1C(=S)NCC1=CC=C(OCO2)C2=C1 FOFDIMHVKGYHRU-UHFFFAOYSA-N 0.000 description 1
- OUSFTKFNBAZUKL-UHFFFAOYSA-N N-(5-{[(5-tert-butyl-1,3-oxazol-2-yl)methyl]sulfanyl}-1,3-thiazol-2-yl)piperidine-4-carboxamide Chemical compound O1C(C(C)(C)C)=CN=C1CSC(S1)=CN=C1NC(=O)C1CCNCC1 OUSFTKFNBAZUKL-UHFFFAOYSA-N 0.000 description 1
- AFJRDFWMXUECEW-LBPRGKRZSA-N N-[(2S)-1-amino-3-(3-fluorophenyl)propan-2-yl]-5-chloro-4-(4-chloro-2-methyl-3-pyrazolyl)-2-thiophenecarboxamide Chemical compound CN1N=CC(Cl)=C1C1=C(Cl)SC(C(=O)N[C@H](CN)CC=2C=C(F)C=CC=2)=C1 AFJRDFWMXUECEW-LBPRGKRZSA-N 0.000 description 1
- XKFTZKGMDDZMJI-HSZRJFAPSA-N N-[5-[(2R)-2-methoxy-1-oxo-2-phenylethyl]-4,6-dihydro-1H-pyrrolo[3,4-c]pyrazol-3-yl]-4-(4-methyl-1-piperazinyl)benzamide Chemical compound O=C([C@H](OC)C=1C=CC=CC=1)N(CC=12)CC=1NN=C2NC(=O)C(C=C1)=CC=C1N1CCN(C)CC1 XKFTZKGMDDZMJI-HSZRJFAPSA-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
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 1
- 206010029113 Neovascularisation Diseases 0.000 description 1
- 208000013901 Nephropathies and tubular disease Diseases 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- ZCQWOFVYLHDMMC-UHFFFAOYSA-N Oxazole Chemical compound C1=COC=N1 ZCQWOFVYLHDMMC-UHFFFAOYSA-N 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- UAOIPNOTWOYAMU-LBPRGKRZSA-N PHA-533533 Chemical compound O=C([C@@H](C)C=1C=CC(=CC=1)N1C(CCC1)=O)NC(=NN1)C=C1C1CC1 UAOIPNOTWOYAMU-LBPRGKRZSA-N 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- QIUASFSNWYMDFS-NILGECQDSA-N PX-866 Chemical compound CC(=O)O[C@@H]1C[C@]2(C)C(=O)CC[C@H]2C2=C1[C@@]1(C)[C@@H](COC)OC(=O)\C(=C\N(CC=C)CC=C)C1=C(O)C2=O QIUASFSNWYMDFS-NILGECQDSA-N 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- SCKXCAADGDQQCS-UHFFFAOYSA-N Performic acid Chemical compound OOC=O SCKXCAADGDQQCS-UHFFFAOYSA-N 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
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 208000007452 Plasmacytoma Diseases 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 206010036790 Productive cough Diseases 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- WTKZEGDFNFYCGP-UHFFFAOYSA-N Pyrazole Chemical compound C=1C=NNC=1 WTKZEGDFNFYCGP-UHFFFAOYSA-N 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 229940127361 Receptor Tyrosine Kinase Inhibitors Drugs 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 206010063837 Reperfusion injury Diseases 0.000 description 1
- 206010038934 Retinopathy proliferative Diseases 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 235000019485 Safflower oil Nutrition 0.000 description 1
- BFZKMNSQCNVFGM-UCEYFQQTSA-N Sagopilone Chemical compound O1C(=O)C[C@H](O)C(C)(C)C(=O)[C@H](CC=C)[C@@H](O)[C@@H](C)CCC[C@@]2(C)O[C@H]2C[C@H]1C1=CC=C(SC(C)=N2)C2=C1 BFZKMNSQCNVFGM-UCEYFQQTSA-N 0.000 description 1
- 102100031081 Serine/threonine-protein kinase Chk1 Human genes 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 101150113537 Spib gene Proteins 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 238000006619 Stille reaction Methods 0.000 description 1
- 208000006011 Stroke Diseases 0.000 description 1
- 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 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 235000019486 Sunflower oil Nutrition 0.000 description 1
- 206010042971 T-cell lymphoma Diseases 0.000 description 1
- 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 1
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 1
- CBPNZQVSJQDFBE-FUXHJELOSA-N Temsirolimus Chemical compound C1C[C@@H](OC(=O)C(C)(CO)CO)[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 CBPNZQVSJQDFBE-FUXHJELOSA-N 0.000 description 1
- IVTVGDXNLFLDRM-HNNXBMFYSA-N Tomudex Chemical compound C=1C=C2NC(C)=NC(=O)C2=CC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)S1 IVTVGDXNLFLDRM-HNNXBMFYSA-N 0.000 description 1
- YCPOZVAOBBQLRI-WDSKDSINSA-N Treosulfan Chemical compound CS(=O)(=O)OC[C@H](O)[C@@H](O)COS(C)(=O)=O YCPOZVAOBBQLRI-WDSKDSINSA-N 0.000 description 1
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- JNPMYSILHRFUPH-UHFFFAOYSA-N UNPD133681 Natural products OC1C(O)CC(=C)CC(C)CC(O2)CC=CC2CC=CC(=O)OC2CC1OC2C=CC1CC(C)=CCO1 JNPMYSILHRFUPH-UHFFFAOYSA-N 0.000 description 1
- 101800003447 VP53 Proteins 0.000 description 1
- 208000024248 Vascular System injury Diseases 0.000 description 1
- 208000012339 Vascular injury 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
- 229940122803 Vinca alkaloid Drugs 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- HGVNLRPZOWWDKD-UHFFFAOYSA-N ZSTK-474 Chemical compound FC(F)C1=NC2=CC=CC=C2N1C(N=1)=NC(N2CCOCC2)=NC=1N1CCOCC1 HGVNLRPZOWWDKD-UHFFFAOYSA-N 0.000 description 1
- GJZGRYXGQBWBEB-AVMFAVRISA-N [(2r)-2-methoxy-3-octadecoxypropyl] [(1r,2r,3s,4r,6r)-2,3,4,6-tetrahydroxycyclohexyl] hydrogen phosphate Chemical compound CCCCCCCCCCCCCCCCCCOC[C@@H](OC)COP(O)(=O)O[C@@H]1[C@H](O)C[C@@H](O)[C@H](O)[C@H]1O GJZGRYXGQBWBEB-AVMFAVRISA-N 0.000 description 1
- XXFXTBNFFMQVKJ-UHFFFAOYSA-N [diphenyl(trityloxy)methyl]benzene Chemical compound C=1C=CC=CC=1C(C=1C=CC=CC=1)(C=1C=CC=CC=1)OC(C=1C=CC=CC=1)(C=1C=CC=CC=1)C1=CC=CC=C1 XXFXTBNFFMQVKJ-UHFFFAOYSA-N 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- 229960000853 abiraterone Drugs 0.000 description 1
- GZOSMCIZMLWJML-VJLLXTKPSA-N abiraterone Chemical compound C([C@H]1[C@H]2[C@@H]([C@]3(CC[C@H](O)CC3=CC2)C)CC[C@@]11C)C=C1C1=CC=CN=C1 GZOSMCIZMLWJML-VJLLXTKPSA-N 0.000 description 1
- DHKHKXVYLBGOIT-UHFFFAOYSA-N acetaldehyde Diethyl Acetal Natural products CCOC(C)OCC DHKHKXVYLBGOIT-UHFFFAOYSA-N 0.000 description 1
- 150000001241 acetals Chemical class 0.000 description 1
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 125000003668 acetyloxy group Chemical group [H]C([H])([H])C(=O)O[*] 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
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 238000010535 acyclic diene metathesis reaction Methods 0.000 description 1
- 230000010933 acylation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 125000005042 acyloxymethyl group Chemical group 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 229950000079 afuresertib Drugs 0.000 description 1
- 238000005054 agglomeration Methods 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 230000001270 agonistic effect Effects 0.000 description 1
- 229910001413 alkali metal ion Inorganic materials 0.000 description 1
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 1
- 125000003545 alkoxy group Chemical group 0.000 description 1
- 230000003281 allosteric effect Effects 0.000 description 1
- HSFWRNGVRCDJHI-UHFFFAOYSA-N alpha-acetylene Natural products C#C HSFWRNGVRCDJHI-UHFFFAOYSA-N 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 229960000473 altretamine Drugs 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- KUFRQPKVAWMTJO-LMZWQJSESA-N alvespimycin Chemical compound N1C(=O)\C(C)=C\C=C/[C@H](OC)[C@@H](OC(N)=O)\C(C)=C\[C@H](C)[C@@H](O)[C@@H](OC)C[C@H](C)CC2=C(NCCN(C)C)C(=O)C=C1C2=O KUFRQPKVAWMTJO-LMZWQJSESA-N 0.000 description 1
- 150000001409 amidines Chemical class 0.000 description 1
- 229960001097 amifostine Drugs 0.000 description 1
- 238000005576 amination reaction Methods 0.000 description 1
- 229960003896 aminopterin 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
- 230000001772 anti-angiogenic effect Effects 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000003388 anti-hormonal effect Effects 0.000 description 1
- 239000012296 anti-solvent Substances 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000002257 antimetastatic agent Substances 0.000 description 1
- 229940045696 antineoplastic drug podophyllotoxin derivative Drugs 0.000 description 1
- 229940045985 antineoplastic platinum compound Drugs 0.000 description 1
- 210000000436 anus Anatomy 0.000 description 1
- 229950004111 apitolisib Drugs 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 230000006793 arrhythmia Effects 0.000 description 1
- 206010003119 arrhythmia Diseases 0.000 description 1
- 239000008122 artificial sweetener Substances 0.000 description 1
- 235000021311 artificial sweeteners Nutrition 0.000 description 1
- 238000006254 arylation reaction Methods 0.000 description 1
- 210000003567 ascitic fluid Anatomy 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 235000006533 astragalus Nutrition 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 230000004900 autophagic degradation Effects 0.000 description 1
- 230000010455 autoregulation Effects 0.000 description 1
- RITAVMQDGBJQJZ-FMIVXFBMSA-N axitinib Chemical compound CNC(=O)C1=CC=CC=C1SC1=CC=C(C(\C=C\C=2N=CC=CC=2)=NN2)C2=C1 RITAVMQDGBJQJZ-FMIVXFBMSA-N 0.000 description 1
- 229960003005 axitinib Drugs 0.000 description 1
- JPNZKPRONVOMLL-UHFFFAOYSA-N azane;octadecanoic acid Chemical class [NH4+].CCCCCCCCCCCCCCCCCC([O-])=O JPNZKPRONVOMLL-UHFFFAOYSA-N 0.000 description 1
- 125000003943 azolyl group Chemical group 0.000 description 1
- 229950005645 barasertib Drugs 0.000 description 1
- 210000000270 basal cell Anatomy 0.000 description 1
- 229960002707 bendamustine Drugs 0.000 description 1
- YTKUWDBFDASYHO-UHFFFAOYSA-N bendamustine Chemical compound ClCCN(CCCl)C1=CC=C2N(C)C(CCCC(O)=O)=NC2=C1 YTKUWDBFDASYHO-UHFFFAOYSA-N 0.000 description 1
- RFRXIWQYSOIBDI-UHFFFAOYSA-N benzarone Chemical compound CCC=1OC2=CC=CC=C2C=1C(=O)C1=CC=C(O)C=C1 RFRXIWQYSOIBDI-UHFFFAOYSA-N 0.000 description 1
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 1
- 229940049706 benzodiazepine Drugs 0.000 description 1
- 125000003310 benzodiazepinyl group Chemical group N1N=C(C=CC2=C1C=CC=C2)* 0.000 description 1
- BNBQRQQYDMDJAH-UHFFFAOYSA-N benzodioxan Chemical compound C1=CC=C2OCCOC2=C1 BNBQRQQYDMDJAH-UHFFFAOYSA-N 0.000 description 1
- WARCRYXKINZHGQ-UHFFFAOYSA-N benzohydrazide Chemical compound NNC(=O)C1=CC=CC=C1 WARCRYXKINZHGQ-UHFFFAOYSA-N 0.000 description 1
- 125000001164 benzothiazolyl group Chemical group S1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 125000004622 benzoxazinyl group Chemical group O1NC(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 description 1
- PUJDIJCNWFYVJX-UHFFFAOYSA-N benzyl carbamate Chemical compound NC(=O)OCC1=CC=CC=C1 PUJDIJCNWFYVJX-UHFFFAOYSA-N 0.000 description 1
- 230000008238 biochemical pathway Effects 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- 239000000090 biomarker Substances 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
- JFIOVJDNOJYLKP-UHFFFAOYSA-N bithionol Chemical compound OC1=C(Cl)C=C(Cl)C=C1SC1=CC(Cl)=CC(Cl)=C1O JFIOVJDNOJYLKP-UHFFFAOYSA-N 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 239000010839 body fluid Substances 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 1
- 229910052794 bromium Inorganic materials 0.000 description 1
- 125000001246 bromo group Chemical group Br* 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 230000003139 buffering effect Effects 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- HGXJOXHYPGNVNK-UHFFFAOYSA-N butane;ethenoxyethane;tin Chemical compound CCCC[Sn](CCCC)(CCCC)C(=C)OCC HGXJOXHYPGNVNK-UHFFFAOYSA-N 0.000 description 1
- 125000004369 butenyl group Chemical group C(=CCC)* 0.000 description 1
- 229930188620 butyrolactone Natural products 0.000 description 1
- BMQGVNUXMIRLCK-OAGWZNDDSA-N cabazitaxel Chemical compound O([C@H]1[C@@H]2[C@]3(OC(C)=O)CO[C@@H]3C[C@@H]([C@]2(C(=O)[C@H](OC)C2=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=3C=CC=CC=3)C[C@]1(O)C2(C)C)C)OC)C(=O)C1=CC=CC=C1 BMQGVNUXMIRLCK-OAGWZNDDSA-N 0.000 description 1
- 229960001573 cabazitaxel Drugs 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 229910000019 calcium carbonate Inorganic materials 0.000 description 1
- MIOPJNTWMNEORI-UHFFFAOYSA-N camphorsulfonic acid Chemical compound C1CC2(CS(O)(=O)=O)C(=O)CC1C2(C)C MIOPJNTWMNEORI-UHFFFAOYSA-N 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- 239000003560 cancer drug Substances 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 238000005251 capillar electrophoresis Methods 0.000 description 1
- 239000007894 caplet Substances 0.000 description 1
- 239000007963 capsule composition Substances 0.000 description 1
- 235000013877 carbamide Nutrition 0.000 description 1
- 125000004452 carbocyclyl group Chemical group 0.000 description 1
- SKOLWUPSYHWYAM-UHFFFAOYSA-N carbonodithioic O,S-acid Chemical compound SC(S)=O SKOLWUPSYHWYAM-UHFFFAOYSA-N 0.000 description 1
- YAYRGNWWLMLWJE-UHFFFAOYSA-L carboplatin Chemical compound O=C1O[Pt](N)(N)OC(=O)C11CCC1 YAYRGNWWLMLWJE-UHFFFAOYSA-L 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 150000007942 carboxylates Chemical class 0.000 description 1
- 150000001733 carboxylic acid esters Chemical class 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 231100000504 carcinogenesis Toxicity 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
- 229960005243 carmustine Drugs 0.000 description 1
- 229950001725 carubicin Drugs 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 239000003729 cation exchange resin Substances 0.000 description 1
- 229940023913 cation exchange resins Drugs 0.000 description 1
- 150000001767 cationic compounds Chemical class 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 230000021164 cell adhesion Effects 0.000 description 1
- 230000032677 cell aging Effects 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000012820 cell cycle checkpoint Effects 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000019522 cellular metabolic process Effects 0.000 description 1
- 230000004637 cellular stress Effects 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229950002352 cenisertib Drugs 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 239000003610 charcoal Substances 0.000 description 1
- OGEBRHQLRGFBNV-RZDIXWSQSA-N chembl2036808 Chemical compound C12=NC(NCCCC)=NC=C2C(C=2C=CC(F)=CC=2)=NN1C[C@H]1CC[C@H](N)CC1 OGEBRHQLRGFBNV-RZDIXWSQSA-N 0.000 description 1
- 229940068682 chewable tablet Drugs 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
- 229960004926 chlorobutanol Drugs 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 208000037976 chronic inflammation Diseases 0.000 description 1
- 208000037893 chronic inflammatory disorder Diseases 0.000 description 1
- 208000024207 chronic leukemia Diseases 0.000 description 1
- WCZVZNOTHYJIEI-UHFFFAOYSA-N cinnoline Chemical compound N1=NC=CC2=CC=CC=C21 WCZVZNOTHYJIEI-UHFFFAOYSA-N 0.000 description 1
- 125000000259 cinnolinyl group Chemical group N1=NC(=CC2=CC=CC=C12)* 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 229960002436 cladribine Drugs 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000000549 coloured material Substances 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 230000002301 combined effect Effects 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 229950002550 copanlisib Drugs 0.000 description 1
- 235000005687 corn oil Nutrition 0.000 description 1
- 239000002285 corn oil Substances 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 229920006037 cross link polymer Polymers 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 150000004292 cyclic ethers Chemical class 0.000 description 1
- 150000004294 cyclic thioethers Chemical group 0.000 description 1
- 229940043378 cyclin-dependent kinase inhibitor Drugs 0.000 description 1
- 238000006352 cycloaddition reaction Methods 0.000 description 1
- 229940097362 cyclodextrins Drugs 0.000 description 1
- 125000003678 cyclohexadienyl group Chemical group C1(=CC=CCC1)* 0.000 description 1
- 125000000596 cyclohexenyl group Chemical group C1(=CCCCC1)* 0.000 description 1
- 125000002433 cyclopentenyl group Chemical group C1(=CCCC1)* 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- AUQDITHEDVOTCU-UHFFFAOYSA-N cyclopropyl cyanide Chemical compound N#CC1CC1 AUQDITHEDVOTCU-UHFFFAOYSA-N 0.000 description 1
- 230000002559 cytogenic effect Effects 0.000 description 1
- 229960002465 dabrafenib Drugs 0.000 description 1
- BFSMGDJOXZAERB-UHFFFAOYSA-N dabrafenib Chemical compound S1C(C(C)(C)C)=NC(C=2C(=C(NS(=O)(=O)C=3C(=CC=CC=3F)F)C=CC=2)F)=C1C1=CC=NC(N)=N1 BFSMGDJOXZAERB-UHFFFAOYSA-N 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 229950006418 dactolisib Drugs 0.000 description 1
- JOGKUKXHTYWRGZ-UHFFFAOYSA-N dactolisib Chemical compound O=C1N(C)C2=CN=C3C=CC(C=4C=C5C=CC=CC5=NC=4)=CC3=C2N1C1=CC=C(C(C)(C)C#N)C=C1 JOGKUKXHTYWRGZ-UHFFFAOYSA-N 0.000 description 1
- 229960002448 dasatinib Drugs 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000020335 dealkylation Effects 0.000 description 1
- 238000006900 dealkylation reaction Methods 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 238000005695 dehalogenation reaction Methods 0.000 description 1
- CFCUWKMKBJTWLW-UHFFFAOYSA-N deoliosyl-3C-alpha-L-digitoxosyl-MTM Natural products CC=1C(O)=C2C(O)=C3C(=O)C(OC4OC(C)C(O)C(OC5OC(C)C(O)C(OC6OC(C)C(O)C(C)(O)C6)C5)C4)C(C(OC)C(=O)C(O)C(C)O)CC3=CC2=CC=1OC(OC(C)C1O)CC1OC1CC(O)C(O)C(C)O1 CFCUWKMKBJTWLW-UHFFFAOYSA-N 0.000 description 1
- 229960000605 dexrazoxane Drugs 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- AQEFLFZSWDEAIP-UHFFFAOYSA-N di-tert-butyl ether Chemical compound CC(C)(C)OC(C)(C)C AQEFLFZSWDEAIP-UHFFFAOYSA-N 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- BMTPVPNVQOYGAP-UHFFFAOYSA-N diethyl 6-methoxy-5,7-dihydroindolo[2,3-b]carbazole-2,10-dicarboxylate Chemical compound N1C2=CC=C(C(=O)OCC)C=C2C2=C1C(OC)=C1NC3=CC=C(C(=O)OCC)C=C3C1=C2 BMTPVPNVQOYGAP-UHFFFAOYSA-N 0.000 description 1
- HPNMFZURTQLUMO-UHFFFAOYSA-N diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 1
- 238000000113 differential scanning calorimetry Methods 0.000 description 1
- 125000004786 difluoromethoxy group Chemical group [H]C(F)(F)O* 0.000 description 1
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 description 1
- 229950009859 dinaciclib Drugs 0.000 description 1
- ZZVUWRFHKOJYTH-UHFFFAOYSA-N diphenhydramine Chemical group C=1C=CC=CC=1C(OCCN(C)C)C1=CC=CC=C1 ZZVUWRFHKOJYTH-UHFFFAOYSA-N 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- NYDXNILOWQXUOF-UHFFFAOYSA-L disodium;2-[[4-[2-(2-amino-4-oxo-1,7-dihydropyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]amino]pentanedioate Chemical compound [Na+].[Na+].C=1NC=2NC(N)=NC(=O)C=2C=1CCC1=CC=C(C(=O)NC(CCC([O-])=O)C([O-])=O)C=C1 NYDXNILOWQXUOF-UHFFFAOYSA-L 0.000 description 1
- 239000007919 dispersible tablet Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 238000004090 dissolution Methods 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
- 229960003668 docetaxel Drugs 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 230000036267 drug metabolism Effects 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 239000007938 effervescent tablet Substances 0.000 description 1
- 239000003480 eluent Substances 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 150000002081 enamines Chemical class 0.000 description 1
- 229950001969 encorafenib Drugs 0.000 description 1
- 239000006274 endogenous ligand Substances 0.000 description 1
- 229960004671 enzalutamide Drugs 0.000 description 1
- WXCXUHSOUPDCQV-UHFFFAOYSA-N enzalutamide Chemical compound C1=C(F)C(C(=O)NC)=CC=C1N1C(C)(C)C(=O)N(C=2C=C(C(C#N)=CC=2)C(F)(F)F)C1=S WXCXUHSOUPDCQV-UHFFFAOYSA-N 0.000 description 1
- 239000002532 enzyme inhibitor Substances 0.000 description 1
- 229960001904 epirubicin Drugs 0.000 description 1
- 230000008472 epithelial growth Effects 0.000 description 1
- QXRSDHAAWVKZLJ-TYFQHMATSA-N epothilone b Chemical compound C/C([C@@H]1C[C@@H]2O[C@@]2(C)CCC[C@@H]([C@H]([C@H](C)C(=O)C(C)(C)[C@H](O)CC(=O)O1)O)C)=C\C1=CSC(C)=N1 QXRSDHAAWVKZLJ-TYFQHMATSA-N 0.000 description 1
- 230000008029 eradication Effects 0.000 description 1
- 229960001433 erlotinib Drugs 0.000 description 1
- 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 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
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 235000019325 ethyl cellulose Nutrition 0.000 description 1
- 229920001249 ethyl cellulose Polymers 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 125000002534 ethynyl group Chemical group [H]C#C* 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
- 229960005420 etoposide Drugs 0.000 description 1
- 229960005167 everolimus Drugs 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 208000030533 eye disease Diseases 0.000 description 1
- 230000035558 fertility Effects 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000003818 flash chromatography Methods 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 238000000684 flow cytometry Methods 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
- 239000012530 fluid Substances 0.000 description 1
- 125000003784 fluoroethyl group Chemical group [H]C([H])(F)C([H])([H])* 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
- 201000003444 follicular lymphoma Diseases 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
- 238000002825 functional assay Methods 0.000 description 1
- JKFAIQOWCVVSKC-UHFFFAOYSA-N furazan Chemical compound C=1C=NON=1 JKFAIQOWCVVSKC-UHFFFAOYSA-N 0.000 description 1
- 125000003838 furazanyl group Chemical group 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 229960002584 gefitinib Drugs 0.000 description 1
- 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 1
- 229960005277 gemcitabine Drugs 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
- 238000011223 gene expression profiling Methods 0.000 description 1
- 238000003209 gene knockout Methods 0.000 description 1
- 206010061989 glomerulosclerosis Diseases 0.000 description 1
- 229930182470 glycoside Natural products 0.000 description 1
- 150000002338 glycosides Chemical class 0.000 description 1
- 150000004795 grignard reagents Chemical class 0.000 description 1
- 230000009036 growth inhibition Effects 0.000 description 1
- 201000009277 hairy cell leukemia Diseases 0.000 description 1
- 230000026030 halogenation Effects 0.000 description 1
- 238000005658 halogenation reaction Methods 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 201000011066 hemangioma Diseases 0.000 description 1
- MCAHMSDENAOJFZ-BVXDHVRPSA-N herbimycin Chemical compound N1C(=O)\C(C)=C\C=C/[C@H](OC)[C@@H](OC(N)=O)\C(C)=C\[C@H](C)[C@@H](OC)[C@@H](OC)C[C@H](C)[C@@H](OC)C2=CC(=O)C=C1C2=O MCAHMSDENAOJFZ-BVXDHVRPSA-N 0.000 description 1
- 229930193320 herbimycin Natural products 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
- 125000006038 hexenyl group Chemical group 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- UUADYKVKJIMIPA-UHFFFAOYSA-N hydron;3-(1-methylindol-3-yl)-4-[1-[1-(pyridin-2-ylmethyl)piperidin-4-yl]indol-3-yl]pyrrole-2,5-dione;chloride Chemical compound Cl.C12=CC=CC=C2N(C)C=C1C(C(NC1=O)=O)=C1C(C1=CC=CC=C11)=CN1C(CC1)CCN1CC1=CC=CC=N1 UUADYKVKJIMIPA-UHFFFAOYSA-N 0.000 description 1
- USZLCYNVCCDPLQ-UHFFFAOYSA-N hydron;n-methoxymethanamine;chloride Chemical compound Cl.CNOC USZLCYNVCCDPLQ-UHFFFAOYSA-N 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- 235000013828 hydroxypropyl starch Nutrition 0.000 description 1
- 230000001329 hyperkeratotic effect Effects 0.000 description 1
- 230000009610 hypersensitivity Effects 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 229960001001 ibritumomab tiuxetan Drugs 0.000 description 1
- 206010021198 ichthyosis Diseases 0.000 description 1
- 229960000908 idarubicin Drugs 0.000 description 1
- 229960003445 idelalisib Drugs 0.000 description 1
- YKLIKGKUANLGSB-HNNXBMFYSA-N idelalisib Chemical compound C1([C@@H](NC=2[C]3N=CN=C3N=CN=2)CC)=NC2=CC=CC(F)=C2C(=O)N1C1=CC=CC=C1 YKLIKGKUANLGSB-HNNXBMFYSA-N 0.000 description 1
- 229960001101 ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 210000003405 ileum Anatomy 0.000 description 1
- 229960003685 imatinib mesylate Drugs 0.000 description 1
- 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 1
- UFBBWLWUIISIPW-UHFFFAOYSA-N imidazo[2,1-b][1,3]thiazole Chemical compound C1=CSC2=NC=CN21 UFBBWLWUIISIPW-UHFFFAOYSA-N 0.000 description 1
- CQQJSOXDEFZGFG-UHFFFAOYSA-N imidazo[4,5-d]imidazole Chemical compound C1=NC2=NC=NC2=N1 CQQJSOXDEFZGFG-UHFFFAOYSA-N 0.000 description 1
- YAMHXTCMCPHKLN-UHFFFAOYSA-N imidazolidin-2-one Chemical compound O=C1NCCN1 YAMHXTCMCPHKLN-UHFFFAOYSA-N 0.000 description 1
- 150000002466 imines Chemical class 0.000 description 1
- 125000001841 imino group Chemical group [H]N=* 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 230000003116 impacting effect Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 description 1
- HOBCFUWDNJPFHB-UHFFFAOYSA-N indolizine Chemical compound C1=CC=CN2C=CC=C21 HOBCFUWDNJPFHB-UHFFFAOYSA-N 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
- 230000006698 induction Effects 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 239000012442 inert solvent Substances 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 229910001411 inorganic cation Inorganic materials 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 229940125425 inverse agonist Drugs 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 238000002665 ion therapy Methods 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- 229950006331 ipatasertib Drugs 0.000 description 1
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound 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 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 208000037906 ischaemic injury Diseases 0.000 description 1
- 230000000302 ischemic effect Effects 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- GWVMLCQWXVFZCN-UHFFFAOYSA-N isoindoline Chemical compound C1=CC=C2CNCC2=C1 GWVMLCQWXVFZCN-UHFFFAOYSA-N 0.000 description 1
- 125000004594 isoindolinyl group Chemical group C1(NCC2=CC=CC=C12)* 0.000 description 1
- 125000000904 isoindolyl group Chemical group C=1(NC=C2C=CC=CC12)* 0.000 description 1
- JNPMYSILHRFUPH-QHENZBBHSA-N isolaulimalide Natural products C[C@H]1C[C@H]2CC=C[C@@H](CC=CC(=O)O[C@H]3C[C@@H](O[C@H]3C=C[C@H]4CC(=CCO4)C)[C@@H](O)[C@H](O)CC(=C)C1)O2 JNPMYSILHRFUPH-QHENZBBHSA-N 0.000 description 1
- 125000000555 isopropenyl group Chemical group [H]\C([H])=C(\*)C([H])([H])[H] 0.000 description 1
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 1
- 125000004628 isothiazolidinyl group Chemical group S1N(CCC1)* 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- FABUFPQFXZVHFB-CFWQTKTJSA-N ixabepilone Chemical compound C/C([C@@H]1C[C@@H]2O[C@]2(C)CCC[C@@H]([C@@H]([C@H](C)C(=O)C(C)(C)[C@H](O)CC(=O)N1)O)C)=C\C1=CSC(C)=N1 FABUFPQFXZVHFB-CFWQTKTJSA-N 0.000 description 1
- 229960002014 ixabepilone Drugs 0.000 description 1
- 150000002576 ketones Chemical class 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 229960004891 lapatinib Drugs 0.000 description 1
- DXOJIXGRFSHVKA-BZVZGCBYSA-N larotaxel Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@@]23[C@H]1[C@@]1(CO[C@@H]1C[C@@H]2C3)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 DXOJIXGRFSHVKA-BZVZGCBYSA-N 0.000 description 1
- 229950005692 larotaxel Drugs 0.000 description 1
- 210000000867 larynx Anatomy 0.000 description 1
- JEHCHYAKAXDFKV-UHFFFAOYSA-J lead tetraacetate Chemical compound CC(=O)O[Pb](OC(C)=O)(OC(C)=O)OC(C)=O JEHCHYAKAXDFKV-UHFFFAOYSA-J 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- 229960001691 leucovorin Drugs 0.000 description 1
- 229960001614 levamisole Drugs 0.000 description 1
- CMJCXYNUCSMDBY-ZDUSSCGKSA-N lgx818 Chemical compound COC(=O)N[C@@H](C)CNC1=NC=CC(C=2C(=NN(C=2)C(C)C)C=2C(=C(NS(C)(=O)=O)C=C(Cl)C=2)F)=N1 CMJCXYNUCSMDBY-ZDUSSCGKSA-N 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- XKPJKVVZOOEMPK-UHFFFAOYSA-M lithium;formate Chemical compound [Li+].[O-]C=O XKPJKVVZOOEMPK-UHFFFAOYSA-M 0.000 description 1
- 210000001853 liver microsome Anatomy 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 229950005069 luminespib Drugs 0.000 description 1
- HWYHZTIRURJOHG-UHFFFAOYSA-N luminol Chemical class O=C1NNC(=O)C2=C1C(N)=CC=C2 HWYHZTIRURJOHG-UHFFFAOYSA-N 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 238000007403 mPCR Methods 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- CCERQOYLJJULMD-UHFFFAOYSA-M magnesium;carbanide;chloride Chemical compound [CH3-].[Mg+2].[Cl-] CCERQOYLJJULMD-UHFFFAOYSA-M 0.000 description 1
- FPYJFEHAWHCUMM-UHFFFAOYSA-N maleic anhydride Chemical compound O=C1OC(=O)C=C1 FPYJFEHAWHCUMM-UHFFFAOYSA-N 0.000 description 1
- 235000011090 malic acid Nutrition 0.000 description 1
- 230000036212 malign transformation Effects 0.000 description 1
- 208000006178 malignant mesothelioma Diseases 0.000 description 1
- 201000005282 malignant pleural mesothelioma Diseases 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-N mandelic acid Chemical compound OC(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-N 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 101150024228 mdm2 gene Proteins 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 1
- 229960004961 mechlorethamine Drugs 0.000 description 1
- 229960003194 meglumine 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
- 210000004914 menses Anatomy 0.000 description 1
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 229960004635 mesna Drugs 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 208000037819 metastatic cancer Diseases 0.000 description 1
- 208000011575 metastatic malignant neoplasm Diseases 0.000 description 1
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 1
- UZKWTJUDCOPSNM-UHFFFAOYSA-N methoxybenzene Substances CCCCOC=C UZKWTJUDCOPSNM-UHFFFAOYSA-N 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 150000004702 methyl esters Chemical class 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 125000001570 methylene group Chemical group [H]C([H])([*:1])[*:2] 0.000 description 1
- 125000002816 methylsulfanyl group Chemical group [H]C([H])([H])S[*] 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 239000002829 mitogen activated protein kinase inhibitor Substances 0.000 description 1
- 229960001156 mitoxantrone Drugs 0.000 description 1
- 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 1
- 239000003607 modifier Substances 0.000 description 1
- 201000005328 monoclonal gammopathy of uncertain significance Diseases 0.000 description 1
- 125000004312 morpholin-2-yl group Chemical group [H]N1C([H])([H])C([H])([H])OC([H])(*)C1([H])[H] 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 238000007838 multiplex ligation-dependent probe amplification Methods 0.000 description 1
- 206010028537 myelofibrosis Diseases 0.000 description 1
- 230000002071 myeloproliferative effect Effects 0.000 description 1
- 208000010125 myocardial infarction Diseases 0.000 description 1
- MRHQBBSKCLIMRH-UHFFFAOYSA-N n-(acetamidomethoxymethyl)acetamide Chemical compound CC(=O)NCOCNC(C)=O MRHQBBSKCLIMRH-UHFFFAOYSA-N 0.000 description 1
- TVMXDCGIABBOFY-UHFFFAOYSA-N n-Octanol Natural products CCCCCCCC TVMXDCGIABBOFY-UHFFFAOYSA-N 0.000 description 1
- GTWJETSWSUWSEJ-UHFFFAOYSA-N n-benzylaniline Chemical compound C=1C=CC=CC=1CNC1=CC=CC=C1 GTWJETSWSUWSEJ-UHFFFAOYSA-N 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- JOWXJLIFIIOYMS-UHFFFAOYSA-N n-hydroxy-2-[[2-(6-methoxypyridin-3-yl)-4-morpholin-4-ylthieno[3,2-d]pyrimidin-6-yl]methyl-methylamino]pyrimidine-5-carboxamide Chemical compound C1=NC(OC)=CC=C1C1=NC(N2CCOCC2)=C(SC(CN(C)C=2N=CC(=CN=2)C(=O)NO)=C2)C2=N1 JOWXJLIFIIOYMS-UHFFFAOYSA-N 0.000 description 1
- 125000004593 naphthyridinyl group Chemical group N1=C(C=CC2=CC=CN=C12)* 0.000 description 1
- 210000003928 nasal cavity Anatomy 0.000 description 1
- 210000001989 nasopharynx Anatomy 0.000 description 1
- 229960000801 nelarabine Drugs 0.000 description 1
- IXOXBSCIXZEQEQ-UHTZMRCNSA-N nelarabine Chemical compound C1=NC=2C(OC)=NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@@H]1O IXOXBSCIXZEQEQ-UHTZMRCNSA-N 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 238000007481 next generation sequencing Methods 0.000 description 1
- HHZIURLSWUIHRB-UHFFFAOYSA-N nilotinib Chemical compound C1=NC(C)=CN1C1=CC(NC(=O)C=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)=CC(C(F)(F)F)=C1 HHZIURLSWUIHRB-UHFFFAOYSA-N 0.000 description 1
- 229960001346 nilotinib Drugs 0.000 description 1
- KPMKNHGAPDCYLP-UHFFFAOYSA-N nimustine hydrochloride Chemical compound Cl.CC1=NC=C(CNC(=O)N(CCCl)N=O)C(N)=N1 KPMKNHGAPDCYLP-UHFFFAOYSA-N 0.000 description 1
- 125000006502 nitrobenzyl group Chemical group 0.000 description 1
- 125000000018 nitroso group Chemical group N(=O)* 0.000 description 1
- OSTGTTZJOCZWJG-UHFFFAOYSA-N nitrosourea Chemical compound NC(=O)N=NO OSTGTTZJOCZWJG-UHFFFAOYSA-N 0.000 description 1
- 229950006344 nocodazole Drugs 0.000 description 1
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 1
- 230000004987 nonapoptotic effect Effects 0.000 description 1
- 231100000804 nongenotoxic Toxicity 0.000 description 1
- 230000009871 nonspecific binding Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 239000012038 nucleophile Substances 0.000 description 1
- 238000010534 nucleophilic substitution reaction Methods 0.000 description 1
- 229960003347 obinutuzumab Drugs 0.000 description 1
- WWZKQHOCKIZLMA-UHFFFAOYSA-N octanoic acid Chemical compound CCCCCCCC(O)=O WWZKQHOCKIZLMA-UHFFFAOYSA-N 0.000 description 1
- 229960002450 ofatumumab Drugs 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- IFRGXKKQHBVPCQ-UHFFFAOYSA-N onalespib Chemical compound C1=C(O)C(C(C)C)=CC(C(=O)N2CC3=CC(CN4CCN(C)CC4)=CC=C3C2)=C1O IFRGXKKQHBVPCQ-UHFFFAOYSA-N 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 150000002892 organic cations Chemical class 0.000 description 1
- 150000002895 organic esters Chemical class 0.000 description 1
- 238000006053 organic reaction Methods 0.000 description 1
- 239000003791 organic solvent mixture Substances 0.000 description 1
- 229940127084 other anti-cancer agent Drugs 0.000 description 1
- 210000003101 oviduct Anatomy 0.000 description 1
- 230000016087 ovulation Effects 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
- CQDAMYNQINDRQC-UHFFFAOYSA-N oxatriazole Chemical compound C1=NN=NO1 CQDAMYNQINDRQC-UHFFFAOYSA-N 0.000 description 1
- 239000007800 oxidant agent Substances 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 150000002926 oxygen Chemical class 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 229960001592 paclitaxel Drugs 0.000 description 1
- 210000002741 palatine tonsil Anatomy 0.000 description 1
- 229960004390 palbociclib Drugs 0.000 description 1
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 description 1
- YJVFFLUZDVXJQI-UHFFFAOYSA-L palladium(ii) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 1
- IPCSVZSSVZVIGE-UHFFFAOYSA-N palmitic acid group Chemical group C(CCCCCCCCCCCCCCC)(=O)O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 description 1
- QNGNSVIICDLXHT-UHFFFAOYSA-N para-ethylbenzaldehyde Natural products CCC1=CC=C(C=O)C=C1 QNGNSVIICDLXHT-UHFFFAOYSA-N 0.000 description 1
- 210000003695 paranasal sinus Anatomy 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 238000005192 partition Methods 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 238000000059 patterning Methods 0.000 description 1
- 229950007460 patupilone Drugs 0.000 description 1
- CUIHSIWYWATEQL-UHFFFAOYSA-N pazopanib Chemical compound C1=CC2=C(C)N(C)N=C2C=C1N(C)C(N=1)=CC=NC=1NC1=CC=C(C)C(S(N)(=O)=O)=C1 CUIHSIWYWATEQL-UHFFFAOYSA-N 0.000 description 1
- 229960000639 pazopanib Drugs 0.000 description 1
- 229960003349 pemetrexed disodium Drugs 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- NQPDZGIKBAWPEJ-UHFFFAOYSA-N pentanoic acid group Chemical class C(CCCC)(=O)O NQPDZGIKBAWPEJ-UHFFFAOYSA-N 0.000 description 1
- 125000002255 pentenyl group Chemical group C(=CCCC)* 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
- 229950010632 perifosine Drugs 0.000 description 1
- 201000002513 peritoneal mesothelioma Diseases 0.000 description 1
- 150000004965 peroxy acids Chemical class 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 210000003800 pharynx Anatomy 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- 125000000951 phenoxy group Chemical group [H]C1=C([H])C([H])=C(O*)C([H])=C1[H] 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 150000003014 phosphoric acid esters Chemical class 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- LFSXCDWNBUNEEM-UHFFFAOYSA-N phthalazine Chemical compound C1=NN=CC2=CC=CC=C21 LFSXCDWNBUNEEM-UHFFFAOYSA-N 0.000 description 1
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 229950004941 pictilisib Drugs 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 150000004885 piperazines Chemical class 0.000 description 1
- 125000004574 piperidin-2-yl group Chemical group N1C(CCCC1)* 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
- 229910052697 platinum Inorganic materials 0.000 description 1
- 150000003058 platinum compounds Chemical class 0.000 description 1
- 210000004910 pleural fluid Anatomy 0.000 description 1
- 210000000557 podocyte Anatomy 0.000 description 1
- YJGVMLPVUAXIQN-XVVDYKMHSA-N podophyllotoxin Chemical class COC1=C(OC)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@H](O)[C@@H]3[C@@H]2C(OC3)=O)=C1 YJGVMLPVUAXIQN-XVVDYKMHSA-N 0.000 description 1
- 239000003600 podophyllotoxin derivative Substances 0.000 description 1
- 208000037244 polycythemia vera Diseases 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920005596 polymer binder Polymers 0.000 description 1
- 239000002491 polymer binding agent Substances 0.000 description 1
- 229920000193 polymethacrylate Polymers 0.000 description 1
- 102000054765 polymorphisms of proteins Human genes 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 230000023603 positive regulation of transcription initiation, DNA-dependent Effects 0.000 description 1
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 1
- 238000000634 powder X-ray diffraction Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 208000003476 primary myelofibrosis Diseases 0.000 description 1
- 230000037452 priming Effects 0.000 description 1
- 229960000624 procarbazine 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
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 201000001474 proteinuria Diseases 0.000 description 1
- 230000004850 protein–protein interaction Effects 0.000 description 1
- 208000029817 pulmonary adenocarcinoma in situ Diseases 0.000 description 1
- 230000000541 pulsatile effect Effects 0.000 description 1
- 239000013014 purified material Substances 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- DVUBDHRTVYLIPA-UHFFFAOYSA-N pyrazolo[1,5-a]pyridine Chemical group C1=CC=CN2N=CC=C21 DVUBDHRTVYLIPA-UHFFFAOYSA-N 0.000 description 1
- LDIJKUBTLZTFRG-UHFFFAOYSA-N pyrazolo[1,5-a]pyrimidine Chemical compound N1=CC=CN2N=CC=C21 LDIJKUBTLZTFRG-UHFFFAOYSA-N 0.000 description 1
- PBMFSQRYOILNGV-UHFFFAOYSA-N pyridazine Chemical compound C1=CC=NN=C1 PBMFSQRYOILNGV-UHFFFAOYSA-N 0.000 description 1
- ILVXOBCQQYKLDS-UHFFFAOYSA-N pyridine N-oxide Chemical compound [O-][N+]1=CC=CC=C1 ILVXOBCQQYKLDS-UHFFFAOYSA-N 0.000 description 1
- 125000004076 pyridyl group Chemical group 0.000 description 1
- HNJBEVLQSNELDL-UHFFFAOYSA-N pyrrolidin-2-one Chemical compound O=C1CCCN1 HNJBEVLQSNELDL-UHFFFAOYSA-N 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 150000003856 quaternary ammonium compounds Chemical class 0.000 description 1
- 150000003242 quaternary ammonium salts Chemical class 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
- 150000003254 radicals Chemical class 0.000 description 1
- 229960004432 raltitrexed Drugs 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000022983 regulation of cell cycle Effects 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 230000001718 repressive effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 208000037803 restenosis Diseases 0.000 description 1
- 210000001525 retina Anatomy 0.000 description 1
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 206010039083 rhinitis Diseases 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 229960001302 ridaforolimus Drugs 0.000 description 1
- 125000006413 ring segment Chemical group 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 239000011435 rock Substances 0.000 description 1
- 235000005713 safflower oil Nutrition 0.000 description 1
- 239000003813 safflower oil Substances 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 210000003079 salivary gland Anatomy 0.000 description 1
- 238000007480 sanger sequencing Methods 0.000 description 1
- 238000007127 saponification reaction Methods 0.000 description 1
- 229930195734 saturated hydrocarbon Natural products 0.000 description 1
- HFHDHCJBZVLPGP-UHFFFAOYSA-N schardinger α-dextrin Chemical compound 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
- 230000007017 scission Effects 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 229950000055 seliciclib Drugs 0.000 description 1
- CYOHGALHFOKKQC-UHFFFAOYSA-N selumetinib Chemical compound OCCONC(=O)C=1C=C2N(C)C=NC2=C(F)C=1NC1=CC=C(Br)C=C1Cl CYOHGALHFOKKQC-UHFFFAOYSA-N 0.000 description 1
- 230000009758 senescence Effects 0.000 description 1
- 230000008313 sensitization Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 238000004467 single crystal X-ray diffraction Methods 0.000 description 1
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical class 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 1
- 238000007390 skin biopsy Methods 0.000 description 1
- 208000000587 small cell lung carcinoma Diseases 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 230000003381 solubilizing effect Effects 0.000 description 1
- 229960003787 sorafenib Drugs 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 150000003413 spiro compounds Chemical class 0.000 description 1
- 125000003003 spiro group Chemical group 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000005507 spraying Methods 0.000 description 1
- 210000003802 sputum Anatomy 0.000 description 1
- 208000024794 sputum Diseases 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 230000003019 stabilising effect Effects 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- KXCAEQNNTZANTK-UHFFFAOYSA-N stannane Chemical compound [SnH4] KXCAEQNNTZANTK-UHFFFAOYSA-N 0.000 description 1
- 229910000080 stannane Inorganic materials 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 238000003860 storage Methods 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
- 238000000859 sublimation Methods 0.000 description 1
- 230000008022 sublimation Effects 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 150000005846 sugar alcohols Chemical class 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- IIACRCGMVDHOTQ-UHFFFAOYSA-N sulfamic acid Chemical compound NS(O)(=O)=O IIACRCGMVDHOTQ-UHFFFAOYSA-N 0.000 description 1
- HXJUTPCZVOIRIF-UHFFFAOYSA-N sulfolane Chemical compound O=S1(=O)CCCC1 HXJUTPCZVOIRIF-UHFFFAOYSA-N 0.000 description 1
- MBDNRNMVTZADMQ-UHFFFAOYSA-N sulfolene Chemical compound O=S1(=O)CC=CC1 MBDNRNMVTZADMQ-UHFFFAOYSA-N 0.000 description 1
- 125000000472 sulfonyl group Chemical group *S(*)(=O)=O 0.000 description 1
- 230000006103 sulfonylation Effects 0.000 description 1
- 238000005694 sulfonylation reaction Methods 0.000 description 1
- 239000002600 sunflower oil Substances 0.000 description 1
- WINHZLLDWRZWRT-ATVHPVEESA-N sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 description 1
- 229960001796 sunitinib Drugs 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 206010042863 synovial sarcoma Diseases 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 239000007916 tablet composition Substances 0.000 description 1
- AYUNIORJHRXIBJ-TXHRRWQRSA-N tanespimycin Chemical compound N1C(=O)\C(C)=C\C=C/[C@H](OC)[C@@H](OC(N)=O)\C(C)=C\[C@H](C)[C@@H](O)[C@@H](OC)C[C@H](C)CC2=C(NCC=C)C(=O)C=C1C2=O AYUNIORJHRXIBJ-TXHRRWQRSA-N 0.000 description 1
- 229950001269 taselisib Drugs 0.000 description 1
- 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 1
- 229960001674 tegafur Drugs 0.000 description 1
- WFWLQNSHRPWKFK-ZCFIWIBFSA-N tegafur Chemical compound O=C1NC(=O)C(F)=CN1[C@@H]1OCCC1 WFWLQNSHRPWKFK-ZCFIWIBFSA-N 0.000 description 1
- 229960004964 temozolomide Drugs 0.000 description 1
- 229960000235 temsirolimus Drugs 0.000 description 1
- QFJCIRLUMZQUOT-UHFFFAOYSA-N temsirolimus Natural products C1CC(O)C(OC)CC1CC(C)C1OC(=O)C2CCCCN2C(=O)C(=O)C(O)(O2)C(C)CCC2CC(OC)C(C)=CC=CC=CC(C)CC(C)C(=O)C(OC)C(O)C(C)=CC(C)C(=O)C1 QFJCIRLUMZQUOT-UHFFFAOYSA-N 0.000 description 1
- FGTJJHCZWOVVNH-UHFFFAOYSA-N tert-butyl-[tert-butyl(dimethyl)silyl]oxy-dimethylsilane Chemical compound CC(C)(C)[Si](C)(C)O[Si](C)(C)C(C)(C)C FGTJJHCZWOVVNH-UHFFFAOYSA-N 0.000 description 1
- XBXCNNQPRYLIDE-UHFFFAOYSA-N tert-butylcarbamic acid Chemical compound CC(C)(C)NC(O)=O XBXCNNQPRYLIDE-UHFFFAOYSA-N 0.000 description 1
- 125000001981 tert-butyldimethylsilyl group Chemical group [H]C([H])([H])[Si]([H])(C([H])([H])[H])[*]C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 125000000037 tert-butyldiphenylsilyl group Chemical group [H]C1=C([H])C([H])=C([H])C([H])=C1[Si]([H])([*]C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 150000003512 tertiary amines Chemical class 0.000 description 1
- FPGGTKZVZWFYPV-UHFFFAOYSA-M tetrabutylammonium fluoride Chemical compound [F-].CCCC[N+](CCCC)(CCCC)CCCC FPGGTKZVZWFYPV-UHFFFAOYSA-M 0.000 description 1
- 125000003039 tetrahydroisoquinolinyl group Chemical group C1(NCCC2=CC=CC=C12)* 0.000 description 1
- 125000000147 tetrahydroquinolinyl group Chemical group N1(CCCC2=CC=CC=C12)* 0.000 description 1
- RAOIDOHSFRTOEL-UHFFFAOYSA-N tetrahydrothiophene Chemical compound C1CCSC1 RAOIDOHSFRTOEL-UHFFFAOYSA-N 0.000 description 1
- 238000002411 thermogravimetry Methods 0.000 description 1
- VLLMWSRANPNYQX-UHFFFAOYSA-N thiadiazole Chemical compound C1=CSN=N1.C1=CSN=N1 VLLMWSRANPNYQX-UHFFFAOYSA-N 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 150000007970 thio esters Chemical class 0.000 description 1
- 150000003568 thioethers Chemical class 0.000 description 1
- 125000003396 thiol group Chemical group [H]S* 0.000 description 1
- 150000003573 thiols Chemical group 0.000 description 1
- BRNULMACUQOKMR-UHFFFAOYSA-N thiomorpholine Chemical compound C1CSCCN1 BRNULMACUQOKMR-UHFFFAOYSA-N 0.000 description 1
- 229930192474 thiophene Natural products 0.000 description 1
- 208000030901 thyroid gland follicular carcinoma Diseases 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- MNRILEROXIRVNJ-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=NC=N[C]21 MNRILEROXIRVNJ-UHFFFAOYSA-N 0.000 description 1
- 125000005147 toluenesulfonyl group Chemical group C=1(C(=CC=CC1)S(=O)(=O)*)C 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- 229960004066 trametinib Drugs 0.000 description 1
- LIRYPHYGHXZJBZ-UHFFFAOYSA-N trametinib Chemical compound CC(=O)NC1=CC=CC(N2C(N(C3CC3)C(=O)C3=C(NC=4C(=CC(I)=CC=4)F)N(C)C(=O)C(C)=C32)=O)=C1 LIRYPHYGHXZJBZ-UHFFFAOYSA-N 0.000 description 1
- UMFCIIBZHQXRCJ-NSCUHMNNSA-N trans-anol Chemical compound C\C=C\C1=CC=C(O)C=C1 UMFCIIBZHQXRCJ-NSCUHMNNSA-N 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 206010044412 transitional cell carcinoma Diseases 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 229960003181 treosulfan Drugs 0.000 description 1
- 150000003852 triazoles Chemical class 0.000 description 1
- 125000004205 trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- 125000000026 trimethylsilyl group Chemical group [H]C([H])([H])[Si]([*])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 229910052722 tritium Inorganic materials 0.000 description 1
- 125000002221 trityl group Chemical group [H]C1=C([H])C([H])=C([H])C([H])=C1C([*])(C1=C(C(=C(C(=C1[H])[H])[H])[H])[H])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 229960000281 trometamol Drugs 0.000 description 1
- 230000001573 trophoblastic effect Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 230000009452 underexpressoin Effects 0.000 description 1
- 208000011479 upper respiratory tract disease Diseases 0.000 description 1
- 229940035893 uracil Drugs 0.000 description 1
- 150000003672 ureas Chemical class 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 210000004291 uterus Anatomy 0.000 description 1
- 238000003828 vacuum filtration Methods 0.000 description 1
- ZOCKGBMQLCSHFP-KQRAQHLDSA-N valrubicin 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)CCCC)[C@H]1C[C@H](NC(=O)C(F)(F)F)[C@H](O)[C@H](C)O1 ZOCKGBMQLCSHFP-KQRAQHLDSA-N 0.000 description 1
- 229960000653 valrubicin Drugs 0.000 description 1
- UHTHHESEBZOYNR-UHFFFAOYSA-N vandetanib Chemical compound COC1=CC(C(/N=CN2)=N/C=3C(=CC(Br)=CC=3)F)=C2C=C1OCC1CCN(C)CC1 UHTHHESEBZOYNR-UHFFFAOYSA-N 0.000 description 1
- 229960000241 vandetanib Drugs 0.000 description 1
- 239000002966 varnish Substances 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 230000008728 vascular permeability Effects 0.000 description 1
- 210000005166 vasculature Anatomy 0.000 description 1
- YCOYDOIWSSHVCK-UHFFFAOYSA-N vatalanib Chemical compound C1=CC(Cl)=CC=C1NC(C1=CC=CC=C11)=NN=C1CC1=CC=NC=C1 YCOYDOIWSSHVCK-UHFFFAOYSA-N 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 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
- HHJUWIANJFBDHT-KOTLKJBCSA-N vindesine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(N)=O)N4C)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 HHJUWIANJFBDHT-KOTLKJBCSA-N 0.000 description 1
- 229960004355 vindesine Drugs 0.000 description 1
- 229920002554 vinyl polymer Polymers 0.000 description 1
- 235000012431 wafers Nutrition 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 238000002424 x-ray crystallography Methods 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/40—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
- A61K31/403—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
- A61K31/4035—Isoindoles, e.g. phthalimide
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- 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
- C07B—GENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
- C07B2200/00—Indexing scheme relating to specific properties of organic compounds
- C07B2200/05—Isotopically modified compounds, e.g. labelled
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D209/00—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
- C07D209/02—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
- C07D209/44—Iso-indoles; Hydrogenated iso-indoles
- C07D209/48—Iso-indoles; Hydrogenated iso-indoles with oxygen atoms in positions 1 and 3, e.g. phthalimide
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
- C07D403/06—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
- C07D403/12—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/02—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
- C07D405/12—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/14—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
- C07D413/06—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
- C07D413/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F9/00—Compounds containing elements of Groups 5 or 15 of the Periodic Table
- C07F9/02—Phosphorus compounds
- C07F9/547—Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
- C07F9/553—Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
- C07F9/572—Five-membered rings
- C07F9/5728—Five-membered rings condensed with carbocyclic rings or carbocyclic ring systems
Abstract
The invention provides a MDM2-p53 inhibiting compound of formula (I): (I) or tautomer or a solvate or a pharmaceutically acceptable salt thereof, wherein the various substituents are as defined in the claims. Also provided are pharmaceutical compositions containing the compounds of formula (I), processes for making the compounds and the medical uses of the compounds. esses for making the compounds and the medical uses of the compounds.
Description
The on provides a MDM2-p53 inhibing compound of formula (I): (I) or tautomer or a
solvate or a pharmaceucally acceptable salt thereof, wherein the various substuents are as
defined in the claims. Also provided are pharmaceucal composions containing the compounds
of formula (I), ses for making the compounds and the medical uses of the compounds.
ISOINDOLINONE INHIBITORS OF THE MDM2-P53 INTERACTION
HAVING ANTICANCER ACTIVITY
FIELD OF THE INVENTION
The invention relates to new isoindolinone derivatives, to pharmaceutical compositions comprising
said compounds and to the use of said nds in the ent of es, e.g. cancer.
RELATED APPLICATIONS
This application is related to United Kingdom patent application number 1517217.4 filed 29 September
2015, the contents ofwhich are incorporated herein by reference in their entirety.
BACKGROUND OF THE INVENTION
The transformation-related n 53 (TP53) gene encodes a 53 KDa protein — p53. The tumour
suppressor protein p53 reacts to cellular stresses, such as hypoxia, DNA damage and oncogenic
activation, via a number of posttranslational modifications including phosphorylation, acetylation and
methylation, and acts as a signalling node in the diverse pathways that become activated. p53 has
additional roles in other physiological processes, including autophagy, cell adhesion, cell metabolism,
fertility, and stem cell aging and development. Phosphorylation of p53, resulting from activation of
kinases including ATM, CHK1 and 2, and DNA-PK, s in a stabilised and riptionally active
form ofthe protein, thus producing a range ofgene products. The responses to p53 activation include
apoptosis, survival, cell-cycle arrest, DNA-repair, angiogenesis, invasion and autoregulation. The
specific combination of which, in concert with the cell’s genetic background, gives rise to the observed
cellular effect i.e. apoptosis, cell-cycle arrest or senescence. For tumour cells, the tic pathway
may be ed due to the loss oftumour suppressor proteins and associated cell cycle checkpoint
controls, coupled with oncogenic .
Under conditions of stress such as a and DNA damage it is known that the cellular level of the
protein p53 increases. p53 is known to initiate ription of a number of genes which govern
ssion through the cell cycle, the initiation of DNA repair and programmed cell death. This
provides a mechanism for the tumour suppressor role of p53 ced through genetic studies.
The activity of p53 is negatively and tightly regulated by a binding interaction with the MDM2 protein,
the transcription ofwhich is itselfdirectly regulated by p53. p53 is inactivated when its ctivation
domain is bound by the MDM2 protein. Once inactivated the functions of p53 are repressed and the
p53-MDM2 complex becomes a target for ubiquitinylation.
In normal cells the balance between active p53 and inactive MDM2-bound p53 is maintained in an
autoregulatory negative feedback loop. That is to say that p53 can activate MDM2 expression, which
in turn leads to the repression of p53.
It has been found that vation of p53 by mutation is common in around half of all common adult
sporadic cancers. Furthermore, in around 10% oftumours, gene amplification and over-expression of
WO 55860
MDM2 results in the loss of functional p53, thereby allowing malignant transformation and rolled
tumour growth.
vation of p53 by a range of mechanisms is a frequent causal event in the development and
progression of . These include inactivation by mutation, targeting by oncogenic viruses and, in a
significant proportion of cases, amplification and/or an elevated rate oftranscription ofthe MDM2 gene
ing in overexpression or increased activation of the MDM2 protein. Gene amplification of MDM2
giving rise to overexpression of MDM2 protein has been observed in tumour samples taken from
common sporadic cancers. Overall, around 10% of tumours had MDM2 amplification, with the highest
incidence found in hepatocellular carcinoma (44%), lung (15%), sarcomas and osteosarcomas (28%),
and Hodgkin disease (67%) (Danovi et al., Mol. Cell. Biol. 2004, 24, 5835-5843, Toledo et al., Nat Rev
Cancer 2006, 6, 909-923, Gembarska et al., Nat Med 2012, 18, 1239-1247). ly, transcriptional
activation of MDM2 by activated p53 results in increased MDM2 protein , forming a negative
feedback loop. The essential nature of p53 regulation by MDM2 and MDMX is demonstrated by gene
knockout mouse models. MDM2-/- knockout mice are embryonically lethal around the time of
implantation. Lethality is rescued in the double knockout for Mdm2 and Trp53. MDM2 inhibits the
activity of p53 directly, by binding to and occluding the p53 transactivation domain, and by promoting
the proteosomal destruction ofthe complex, through its E3-ubiquitin ligase activity. In addition, MDM2
is a transcriptional target of p53, and so the two proteins are linked in an autoregulatory feedback
loop, ensuring that p53 activation is ent.
The induction of the p14ARF protein, the alternate reading frame (ARF) t of the 4a
locus, is also a mechanism of negatively regulating the p53-MDM2 interaction. p14ARF ly
interacts with MDM2 and leads to up-regulation of p53 transcriptional response. Loss of p14ARF by a
homozygous mutation in the CDKN2A (lNK4A) gene will lead to elevated levels in MDM2 and,
therefore, loss of p53 function and cell cycle control.
Although MDMX shows strong amino acid sequence and ural homology to MDM2, neither
protein can substitute for loss of the other; MDMX null mice die in utero, whereas MDM2 knockout is
lethal during early embryogenesis, however both can be rescued by p53 knockout, demonstrating
pendence of the ity. MDMX also binds p53 and inhibits pendent transcription, but
unlike MDM2 it is not transcriptionally activated by p53 and so does not form the same gulatory
loop. Furthermore, MDMX has neither E3 ubiquitin ligase activity nor a nuclear localisation signal,
however it is believed to contribute to p53 degradation by forming heterodimers with MDM2 and
contributing to MDM2 stabilisation.
The therapeutic rationale for MDM2-p53 inhibition is that a potent inhibitor of the protein-protein
interaction will te p53 from the repressive control of MDM2, and activate p53 mediated cell death
in the tumour. ln tumours, selectivity is envisioned to result from p53 sensing preexisting DNA-damage
or oncogenic activation signals that had previously been d by the action of MDM2 at normal or
overexpressed levels. In normal cells, p53 activation is anticipated to result in activation of non-
apoptotic pathways and if anything a protective growth inhibition response. In addition due to the non-
genotoxic mechanism of action for MDM2-p53 inhibitors they are suitable for the treatment of cancer
in particular in the pediatric population.
About 50% of cancers harbour cells in which TP53, the gene that encodes for p53, is mutated
resulting in a loss of the protein's tumour suppressor function and sometimes even in p53 protein
ns that gain novel oncogenic functions.
s where there is a high level of MDM2 amplification include liposarcoma (88%), soft tissue
sarcoma (20%), osteosarcoma (16%) oesophageal cancer (13%), and certain paediatric malignancies
including B-cell malignancies.
The present invention describes a novel series of compounds which ively inhibit the MDM2-p53
interaction and which have anticancer activity.
SUMMARY OF THE INVENTION
In a first aspect, the invention provides a compound of formula (I):
s (R5)m
(R4)a
R6 N
(R1)n
or a er or a e or a pharmaceutically acceptable salt thereof, n:
R1 is independently selected from hydroxy, halogen, nitro, nitrile, C1-4alkyl, haloC1-4alkyl,
hydroxyC1-4alkyl, C2-6alkenyl, C1-4alkoxy, haloC1-4alkoxy, C2-4alkynyl,
-O0,1-(CRxRy)v-CO2H, -(CRxRy)v-CO2C1-4alkyl, -(CRxRy)v-CON(C1-4alkyl)2, -P(=O)(Rx)2, -S(O)d-Rx,
-S(O)d-heterocyclic group with 3 to 6 ring s and -S(O)d-N(R8)2;
R2 is ed from en, C1-4 alkyl, C2-6alkenyl, hydroxyC1-4alkyl, -(CRxRy)u-CO2H,
-(CRxRy)u-CO2C1-4alkyl, and -(CRxRy)u-CONRxRy;
s is selected from 0 and 1;
R3 is hydrogen or -(A)t-(CRxRy)q-X;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)t-(CRxRy)q-X then (i) at least one of s, t and q is other than 0 and (ii) when
t is 0 then s is 1 and q is other than 0;
A is a C3-6cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the
heterocyclic group ses one or more heteroatoms selected from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)v-CO2H, -(CH2)v-CO2C1-4alkyl, -S(O)d-
Rx, -C(=O)-C1-4alkyl, -S(O)d-N(H)e(C1-4alkyl)2-e, -NRxRy , -NHSO2Rx, -NRxCORy, and -C(=O)NRxRy;
R4 and R5 are ndently ed from halogen, nitrile, C1-4 alkyl, haloC1-4alkyl, C1-4alkoxy
and haloC1-4alkoxy;
R6 and R7 are independently selected from hydrogen, kyl, haloC1-6alkyl, C2-6alkenyl, C2-
6alkynyl, hydroxy, hydroxyC1-6alkyl, -COOC1-6alkyl, -(CH2)j-O-C1-6alkyl, -(CH2)j-O-(hydroxyC1-6alkyl), -
kyl-NRxRy, -(CRxRy)p-CONRxRy, -(CRxRy)p-NRxCORy, -(CRxRy)p-O-CH2-CONRxRy, heterocyclic
group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring s, -CH2-O-heterocyclic
group with 3 to 7 ring members, H-heterocyclic group with 3 to 7 ring members, -CH2-N(C1-
6alkyl)-heterocyclic group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring
members, C3-8cycloalkyl, -CH2-C3-8cycloalkyl, -CH2-O-C3-8cycloalkyl, and C3-8cycloalkenyl, wherein
said cycloalkyl, cycloalkenyl or heterocyclic groups may be optionally substituted by one or more Rz
groups, and wherein in each instance the cyclic group comprises one or more heteroatoms
ed from N, O, S and oxidised forms thereof;
or the R6 and R7 groups, together with the carbon atom to which they are attached, can join to
form a C3-6cycloalkyl or heterocyclyl group with 3 to 6 ring members, n the heterocyclic group
comprises one or more heteroatoms selected from N, O, S and oxidised forms thereof, and wherein
said C3-6cycloalkyl and heterocyclyl groups may be optionally tuted by one or more Rz groups;
R8 and R9 are independently selected from en, C1-6alkyl, haloC1-6alkyl, hydroxyC1-6alkyl,
-(CH2)k-O-C1-6alkyl, -(CH2)k-O-(hydroxyC1-6alkyl), hydroxyC1-6alkoxy, -(CH2)k-CO2C1-6alkyl, -(CH2)k-
CO2H, -C1-6 alkyl-N(H)e(C1-4alkyl)2-e, -(CH2)j-C3-8cycloalkyl and -(CH2)j-C3-8cycloalkenyl;
Rx and Ry are independently selected from en, halogen, nitro, nitrile, C1-6alkyl, haloC1-
6alkyl, C2-6alkenyl, C2-6alkynyl, hydroxy, hydroxyC1-6alkyl, C1-6alkoxy, -(CH2)k-O-C1-6alkyl, hydroxyC1-
6alkoxy, -COOC1-6alkyl, -N(H)e(C1-4alkyl)2-e, -C1-6alkyl-N(H)e(C1-4alkyl)2-e, -(CH2)k-C(=O)N(H)e(C1-
4alkyl)2-e, C3-8cycloalkyl and cloalkenyl;
or the Rx and Ry groups, together with the carbon or nitrogen atom to which they are attached,
can join to form a C3-6cycloalkyl or saturated heterocyclyl group with 3 to 6 ring s which may be
optionally fused to an aromatic heterocyclyl group of 3 to 5 ring members;
or when on a carbon atom the Rx and Ry groups can join together to form a =CH2 group; Rz is
independently selected from halogen, nitro, nitrile, C1-6alkyl, haloC1-6alkyl, C2-6alkenyl, C2-6alkynyl, =O,
hydroxy, hydroxyC1-6alkyl, C1-6alkoxy, -(CH2)k-O-C1-6alkyl, hydroxyC1-6alkoxy, -C(=O)C1-6alkyl, -
C(=O)C1-6alkyl-OH, -C(=O)C1-6alkyl-N(H)e(C1-4alkyl)2-e, N(H)e(C1-4alkyl)2-e, -(CH2)r-CO2C1-6alkyl, -
(CH2)r-CO2H, -N(H)e(C1-4alkyl)2-e, -C1-6alkyl-N(H)e(C1-4alkyl)2-e, heterocyclyl group with 3 to 6 ring
members, cyclyl group with 3 to 6 ring members substituted by -C(=O)C1-4alkyl, heterocyclyl
group with 3 to 6 ring members substituted by -C(=O)OC1-4alkyl, heterocyclyl group with 3 to 6 ring
s substituted by N(H)e(C1-4alkyl)2-e, -C(=O)heterocyclyl group with 3 to 6 ring members,
C3-8cycloalkyl and C3-8cycloalkenyl, wherein if R7 is pyridine then Rz is other than -NH2;
a, j, d, e, n, r and p are independently selected from 0, 1 and 2;
k and m are independently selected from 1 and 2;
u is selected from 0, 1, 2 and 3; and
v is selected from 0 and 1.
In a second aspect, the ion provides a compound, or a tautomer, pharmaceutically acceptable
salt or e thereof, wherein the nd is selected from:
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-
6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxyethoxy)(2-hydroxypropan
yl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[3-(hydroxymethyl)oxetanyl]methoxy}-
6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)oxo-2,3-
dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarboxylic acid;
(3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](2,3-dihydroxymethylpropoxy)
(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](1,2-dihydroxypropanyl){[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](2-hydroxymethoxypropanyl)
{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][1-(dimethylamino)hydroxypropanyl]-
3-{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}-
-(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](1,2-dihydroxypropanyl){[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](3-hydroxymethylbutoxy)(2-
hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)[(1H-pyrazol
yl)methoxy]-2,3-dihydro-1H-isoindolone;
1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)oxo-2,3-
dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarbonitrile;
N-{[1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)oxo-2,3-
dihydro-1H-isoindolyl]oxy}methyl)cyclopropyl]methyl}methanesulfonamide;
(3R)(4-chlorophenyl)[(4-ethynylphenyl)methyl]{[1-
xymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-ethynylphenyl)methyl]fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)(1,2-dihydroxypropanyl)[(4-ethynylphenyl)methyl]fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
4-{[(1R)(4-chlorophenyl)fluoro({1-[hydroxy(2H₂)methyl]cyclopropyl}(2H₂)methoxy)(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile;
4-{[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan
yl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)[(3-
methyloxetanyl)methoxy]-2,3-dihydro-1H-isoindolone;
4-{[(1R)(4-chlorophenyl)(1,2-dihydroxypropanyl){[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][(1-hydroxycyclopropyl)methoxy](2-
hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
-(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl){[1-
(methoxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3R)(4-Chlorophenyl)[(4-chlorophenyl)methyl]{[1-(hydroxymethyl)cyclobutyl]methoxy}-
6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
-chloro{[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid;
(3R){[4-chloro(morpholinesulfonyl)phenyl]methyl}(4-chlorophenyl){[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropane
carboxamide;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)({1-
xy(2H2)methyl]cyclopropyl}(2H2)methoxy)(2-hydroxypropanyl)-2,3-dihydro-1H-
isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)(oxolan
yloxy)-2,3-dihydro-1H-isoindolone;
-(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)[(oxolan
yl)methoxy]-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-hydroxy
(oxanyl)ethyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2-hydroxy
(piperazinyl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindol-
1-one;
(3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[(3S,4R)hydroxyoxolanyl]oxy}-
6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[(3R,4S)hydroxyoxolanyl]oxy}-
6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)methoxy-2,3-
dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]({1-
[hydroxy(2H₂)methyl]cyclopropyl}(2H₂)methoxy)(2-hydroxypropanyl)-2,3-dihydro-1H-
isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)(3-
hydroxypropoxy)-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2,2-difluorohydroxypropoxy)(2-
hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[2-(hydroxymethyl)cyclobutyl]methoxy}
(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][2-hydroxyoxo(pyrrolidin
yl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
2-[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]hydroxy-N,N-
dimethylpropanamide;
2-[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]hydroxy-N-
methylpropanamide;
(3R){[4-chloro(methylsulfanyl)phenyl]methyl}(4-chlorophenyl)fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfinylphenyl)methyl](4-chlorophenyl)fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro(2-hydroxy
methoxypropanyl){[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
-[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)(1,2-dihydroxypropan-
2-yl)fluoro{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2-hydroxy
(4-methylpiperazinyl)propanyl][(3R)-oxolanyloxy]-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2-hydroxy
(4-methylpiperazinyl)propanyl][(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolone;
-(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro(2-hydroxypropanyl)oxo
oxolanyloxy]-2,3-dihydro-1H-isoindolyl]propanoic acid;
1-({[(1R){[4-chloro(hydroxymethyl)phenyl]methyl}(4-chlorophenyl)fluoro(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarbonitrile;
1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-
hydroxy(1-methyl-1H-pyrazolyl)ethyl]oxo-2,3-dihydro-1H-isoindol
yl]oxy}methyl)cyclopropanecarboxamide;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-hydroxy
(1-methyl-1H-pyrazolyl)ethyl][(1-hydroxycyclopropyl)methoxy]-2,3-dihydro-1H-isoindol
one;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2-hydroxy
(4-methylpiperazinyl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-
1H-isoindolone;
-chloro{[(1R)(4-chlorophenyl)[(1-cyanocyclopropyl)methoxy]fluoro(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-hydroxy
(1-methylpiperidinyl)ethyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-
isoindolone;
(3R){[4-chloro(dimethylphosphoryl)phenyl]methyl}(4-chlorophenyl)fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone;
(3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro
[hydroxy(oxanyl)methyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindol-
1-one;
1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-
hydroxy(oxanyl)ethyl]oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropane
carboxamide;
-chloro{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methylpiperidinyl)ethyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(oxanyl)ethyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]
oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]hydroxyethyl]benzonitrile;
4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]oxo
[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]methyl}(hydroxymethyl)benzonitrile;
4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile;
4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]oxo
[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluorooxanyl)
hydroxyethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-
pyrazolyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid;
-(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluorooxanyl)
hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)(1-cyclobutylhydroxyethyl)fluoro
methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
-(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid;
(4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid;
(4S)(4-Chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)
hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)
ypropyl]trideuteromethoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)ethoxyfluoro[(1R)(4-fluorooxan
yl)hydroxypropyl]oxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(4S)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl](4-methoxyphenyl)butanoic acid;
(4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro{1-hydroxy[trans
hydroxycyclohexyl]propyl}methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid;
2-(5-chloro{[1-(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenoxy)acetic acid;
-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]methoxy-
3-oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid;
-chloro{[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxanyl)propyl]methoxy-
3-oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid;
-chloro{[(1R)(4-chlorophenyl)ethoxyfluoro[(1S)hydroxy(oxanyl)propyl]
oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid;
2-{[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl]methoxy-
3-oxo-2,3-dihydro-1H-isoindolyl]methyl}methylbenzoic acid;
2-{[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl]methoxy-
3-oxo-2,3-dihydro-1H-isoindolyl]methyl}methoxybenzoic acid;
2-(5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)methylpropanoic acid;
2-(5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]
yoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)acetic acid;
hloro{[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxanyl)propyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)acetic acid;
(2S,3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methylpropanoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(3-fluorooxetanyl)methoxy]
(2-hydroxybutanyl)oxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(pyridinyl)propyl]-
1-methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
-[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-(4-
fluoropiperidinyl)hydroxypropyl]methoxy-2,3-dihydro-1H-isoindolone;
4-{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(1-methylpiperidinyl)propyl]oxo
[cishydroxycyclobutoxy]-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile;
(3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluoromethylpiperidin
yl)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid;
tert-butyl 2-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy
3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetate;
tert-butyl 2-{4-[(1R)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy
oxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetate;
2-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxyoxo-2,3-
dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetic acid;
2-{4-[(1R)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxyoxo-2,3-
o-1H-isoindolyl]hydroxypropyl]piperidinyl}acetic acid;
methyl (1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy
oxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}propanoate; and
3-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxyoxo-2,3-
dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}propanoic acid.
In a third aspect, the invention provides a compound, or a tautomer, pharmaceutically acceptable salt
or solvate f, wherein the compound is 2-{[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]
(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}-N,N-dimethylacetamide.
In a fourth aspect, the invention provides a combination comprising a compound as defined in the first
or second aspects of the invention with one or more other therapeutic agents.
In a fifth aspect, the invention es a pharmaceutical ition comprising a compound as
defined in the first or second aspects of the invention or a combination as defined in the third aspect of
the invention.
In a sixth aspect, the invention provdes a pharmaceutical composition comprising a compound as
defined in the first or second s of the invention, one or more other therapeutic agents, and a
pharmaceutically acceptable carrier.
In a seventh aspect, the invention provides the use of a compound as defined in the first or second
aspects of the invention, a combination according to the third aspect of the invention, or a
pharmaceutical ition ing to the fourth or fifth aspects of the invention for the manufacture
of a medicament for the prophylaxis or treatment of a disease state or condition mediated by MDM2-
p53.
In an eighth aspect, the invention provides the use of a compound as defined in the first or second
aspects of the invention, a combination according to the third aspect of the invention, or a
pharmaceutical composition according to the fourth or fifth aspects of the invention in the manufacture
of a medicament for the prophylaxis or treatment of cancer.
In a ninth aspect, the invention provides a process for the preparation of a compound as d in the
first or second aspects of the invention, or a tautomer, pharmaceutically acceptable salt, or solvate
thereof which comprises:
(a) reacting a compound of the following formula with an metallic reagent:
s R5
R4 a
R1 n
wherein R1, R2, R3, R4, R5, R7, a, s, m and n are as defined in the first aspect of the ion,
and/or
(b) interconversion of a compound of formula (I) or protected tive thereof to a further
compound of formula (I) or protected derivative thereof; and/or
(c) deprotection of a protected derivative of a compound of formula (I); and/or
(d) providing a compound of formula (I) and forming a pharmaceutically able salt of
the compound.
In one aspect, the invention provides a compound of a (I):
s (R5)m
(R4)a
R6 N
(R1)n
or a tautomer or a solvate or a pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected from hydroxy, n, nitro, nitrile, C1-4alkyl, haloC1-4alkyl,
hydroxyC1-4alkyl, C2-6alkenyl, C1-4alkoxy, haloC1-4alkoxy, kynyl,
-O0,1-(CRxRy)v-CO2H, -(CRxRy)v-CO2C1-4alkyl, -(CRxRy)v-CON(C1-4alkyl)2, -P(=O)(Rx)2, -S(O)d-Rx,
-S(O)d-heterocyclic group with 3 to 6 ring members and -S(O)d-N(R8)2;
R2 is selected from hydrogen, C1-4 alkyl, C2-6alkenyl, hydroxyC1-4alkyl, -(CRxRy)u-CO2H,
-(CRxRy)u-CO2C1-4alkyl, and -(CRxRy)u-CONRxRy;
s is selected from 0 and 1;
R3 is hydrogen or -(A)t-(CRxRy)q-X;
t is ed from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)t-(CRxRy)q-X then (i) at least one of s, t and q is other than 0 and (ii) when
t is 0 then s is 1 and q is other than 0;
[Followed by page 4]
A is a ycloalkyl group or a heterocyclic group with 3 to 6 ring members, n the
heterocyclic group comprises one or more (e.g.1, 2, or 3) atoms selected from N, O, S and
oxidised forms thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, V-COZC1_4a|kyl, -S(O)d-
RX, -C(=O)-C1_4a|kyl, -S(O)d-N(H)e(C1_4alkyl)2_e, -NR"Ry and -C(=O)NRXRV;
, -NHSOZRX, -NRXCORy,
R4 and R5 are independently selected from halogen, nitrile, C1_4 alkyl, haloC1_4alkyl, C1_4alkoxy
and haloC1_4alkoxy;
R6 and R7 are independently selected from hydrogen, C1_5alkyl, haloC1_6alkyl, C2_6alkenyl, C2.
6alkynyl, hydroxy, hydroxyC1_6alkyl, -COOC1_6alkyl, -(CH2)j-O-C1_6alkyl, -(CH2)j-O-(hydroxyC1_6alkyl), -
C1_5alkyl-NRXRy, -(CRXRy)p-CONRXRV, -(CRXRy)p-NRXCORV, -(CRXRy)p-O-CH2-CONRXRV, heterocyclic
group with 3 to 7 ring members, eterocyclic group with 3 to 7 ring members, -CH2-O-
heterocyclic group with 3 to 7 ring members, -CH2-NH-heterocyclic group with 3 to 7 ring members, -
CHz-N(C1.6alkyl)-heterocyclic group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7
ring members, Cg_8cycloalkyl, -CH2-Cg_8cycloalkyl, -CH2-O-C3_8cycloalkyl, and Cg_8cycloalkenyl,
wherein said lkyl, lkenyl or heterocyclic groups may be optionally substituted by one or
more RZ groups, and wherein in each instance the heterocyclic group ses one or more , 2,
or 3) heteroatoms selected from N, O, S and oxidised forms thereof;
or the R6 and R7 groups, together with the carbon atom to which they are attached, can join to
form a Cg_6cycloalkyl or heterocyclyl group with 3 to 6 ring members, n the heterocyclic group
ses one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms f,
and wherein said 03.6cycloalkyl and heterocyclyl groups may be optionally substituted by one or more
RZ groups;
R8 and R9 are independently selected from hydrogen, C1_6a|kyl, haloC1_6alkyl, hydroxyC1_6a|kyl,
'(CH2)k-O-C1.sa|ky|, '(CH2)k'O-(hydr0XyC1.ealkyl), hydrOXYC1.6a|k0Xy, -(CH2)k-C02C1.5a|ky|, '(CH2)k-
COZH, -C1_6 N(H)e(C1.4alkyl)2_e, -(CH2),--C3.gcycloalkyl and -(CH2)j-C3.gcycloalkenyl;
RX and Ry are independently selected from hydrogen, halogen, nitro, nitrile, C1_6alkyl, haloC1_
6alkyl, C2_6alkenyl, C2_6alkynyl, hydroxy, hydroxyC1_6alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_
6alkoxy, -COOC1_6alkyI, -N(H)e(C1_4alkyI)2.e, -C1.6alkyI-N(H)e(C1_4aIkyl)2_e, -(CH2)k-C(=O)N(H)e(C1_
4alkyl)2_e, Cg_8cycloalkyl and Cg_8cycloalkenyl;
orthe RX and Ry groups, er with the carbon or nitrogen atom to which they are ed,
can join to form a Cg_6cycloalkyl or saturated heterocyclyl group with 3 to 6 ring members which may
be optionally fused to an aromatic heterocyclyl group of 3 to 5 ring members;
or when on a carbon atom the RX and Ry groups can join togetherto form a =CH2 group;
RZ is independently selected from halogen, nitro, nitrile, C1_6alkyl, haloC1_6alkyl, C2_6alkenyl, C2.
5alkynyl, =0, hydroxy, yC1.6alkyl, C1_6alkoxy, -(CH2)k-O-C1.6alkyl, hydroxyC1_6alkoxy, -C(=O)C1_
6alkyl, -C(=O)C1_6alkyl-OH, -C(=O)C1.6alkyl-N(H)e(C1_4alkyl)2.e, N(H)e(C1.4alkyl)2_e, -(CH2),-COZC1_
salkyl, -(CH2),-COZH, -N(H)e(C1_4alkyl)2—e, -C1_6alkyl-N(H)e(C1_4alkyl)2_e, heterocyclyl group with 3 to 6
WO 55860
ring members, heterocyclyl group with 3 to 6 ring members substituted by C1_4alkyl, heterocyclyl
group with 3 to 6 ring members tuted by -C(=O)OC1_4alkyl, heterocyclyl group with 3 to 6 ring
members substituted by -C(=O)N(H)e(C1_4alkyI)2.e, -C(=O)heterocyclyl group with 3 to 6 ring members,
Cg_gcycloalkyl and 03.8cycloalkenyl, wherein if R7 is pyridine then R2 is other then -NH2;
a, j, d, e, n, rand p are independently ed from 0, 1 and 2;
k and m are independently selected from 1 and 2;
u is selected from 0, 1, 2 and 3; and
v and w are independently selected from 0 and 1.
In a further aspect aspect, the invention provides a compound of formula (I):
— (I)
or a tautomer or a solvate or a pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected from hydroxy, halogen, nitro, nitrile, C1_4alkyl, haloCMalkyl,
hydroxyC1_4a|kyl, C2_6alkeny|, C1_4alkoxy, haloC1_4a|koxy, Cz_4alkynyl,
-(CRXRV)V-COZH, -(CRXRy)V-COZC1_4alkyl, -(CRXRy)v-CON(C1_4alkyl)2, -P(=O)(RX)2, -S(O)d-RX, -S(O)dheterocyclic
group with 3 to 6 ring members and -S(O)d-N(R8)2;
R2 is selected from hydrogen, CM alkyl, C2_6alkenyl, hydroxyC1_4alkyl, -(CRXRy)u-COZH,
-(CRXRy)u-COZC1.4alkyl, and -(CRXRy)u-CONRXRV;
s is selected from 0 and 1;
R3 is hydrogen or -(A),-(CRXRy)q-X;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
n when R3 is -(A)1-(CRXRy)q-X then (i) at least one of s, t and q is other than 0 and (ii)
when t is 0 then s is 1 and q is other than 0;
A is a ycloalkyl group or a cyclic group with 3 to 6 ring s, wherein the
heterocyclic group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and
oxidised forms thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -S(O)d-
RX, -C(=O)-C1_4alkyl, -S(O)d-N(H)e(C1_4alkyI)2_e, -NR"Ry and -C(=O)NRXRy;
, -NHSOZRX, -NRXCORy,
R4 and R5 are independently selected from halogen, nitrile, CM alkyl, haloC1.4a|kyl, C1.4alkoxy
and haloC1_4alkoxy;
R6 and R7 are independently selected from hydrogen, C1,6alkyl, haloC1_6alkyl, 02,6alkenyl, CZ.
6alkynyl, hydroxy, yC1_6alkyl, -COOC1_6alkyl, -(CH2)j-O-C1_6alkyl, -(CH2)j-O-(hydroxyC1_6alkyl), -
C1,6alkyl-NRXRV, y)p-CONRXRV, -(CRXRy)p-NRXCORV, y)p-O-CH2-CONRXRY, heterocyclic
group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring members, -CH2-O-
heterocyclic group with 3 to 7 ring members, -CH2-NH-heterocyclic group with 3 to 7 ring s, -
CHz-N(C1.6alkyl)-heterocyc|ic group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7
ring members, 03-8cycloalkyl, -CH2-Cs.gcycloalkyl, -CH2-O-Cs.3cycloalkyl, and C3.3cycloalkenyl,
wherein said cycloalkyl, cycloalkenyl or heterocyclic groups may be optionally substituted by one or
more RZ groups, and wherein in each instance the heterocyclic group comprises one or more (e.g.1, 2,
or 3) heteroatoms selected from N, O, S and oxidised forms thereof;
or the R6 and R7 , together with the carbon atom to which they are attached, can join to
form a Cg_5cycloalkyl or cyclyl group with 3 to 6 ring members, wherein the heterocyclic group
comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms thereof,
and wherein said C3_Scycloalkyl and heterocyclyl groups may be optionally substituted by one or more
RZ groups;
R8 and R9 are ndently selected from hydrogen, C1_5alkyl, haloC1_6alkyl, yC1_5alkyl,
-(CH2)k-O-C1_6a|ky|, -(CH2)k-O-(hydroxyC1,6alkyl), hydrOXYC1_6a|k0Xy, -(CH2)k-C02C1_6a|ky|, '(CH2)k-
COZH, -C1_5 alkyl-N(H)e(C1.4alkyl)2_e, -(CH2)j-C3.gcycloalkyl and -(CH2)j-C3.8cycloalkenyl;
RX and Ry are independently selected from hydrogen, halogen, nitro, nitrile, C1_6alkyl, haloC1_
salkyl, kenyl, Cz_6alkynyl, hydroxy, hydroxyC1.5alkyl, C1_6alkoxy, -(CH2)k-O-C1_5alkyl, hydroxyC1.
6alkoxy, -COOC1_6alkyl, -N(H)e(C1_4alky|)2.e, -C1.6alkyl-N(H)e(C1_4alkyl)2_e, -(CH2)k-C(=O)N(H)e(C1_
4a|ky|)2.e, C3_8cycloalkyl and Cucycloalkenyl;
orthe RX and Ry groups, together with the carbon or nitrogen atom to which they are attached,
can join to form a 03.6cycloalkyl or ted heterocyclyl group with 3 to 6 ring members which may
be optionally fused to an aromatic heterocyclyl group of 3 to 5 ring members;
or when on a carbon atom the RX and Ry groups can join togetherto form a =CH2 group;
RZ is ndently selected from halogen, nitro, nitrile, C1_6alkyl, haloC1_5alkyl, Cz_6alkenyl, CZ.
6alkynyl, :0, hydroxy, hydroxyC1_6alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_6alkoxy, -C(=O)C1_
salkyl, -C(=O)C1_5a|kyl-OH, -C(=O)C1_6alkyI-N(H)e(C1_4alkyl)2.e, -C(=O)N(H)e(C1_4a|kyI)2_e, -(CH2)r-COZC1_
, -(CH2)r-COZH, (C1_4alkyl)2.e, -C1.6alkyl-N(H)e(C1_4alky|)2.e, cyclyl group with 3 to 6
ring members, heterocyclyl group with 3 to 6 ring members substituted by -C(=O)C1_4alkyl, heterocyclyl
group with 3 to 6 ring s tuted by -C(=O)OC1_4alkyl, heterocyclyl group with 3 to 6 ring
members substituted by N(H)e(C1_4alkyl)2.e, -C(=O)heterocyclyl group with 3 to 6 ring members,
Cg_8cycloalkyl and 03.8cycloalkenyl, wherein if R7 is pyridine then R2 is other then -NH2;
a, j, d, e, n, rand p are independently selected from 0, 1 and 2;
k and m are independently selected from 1 and 2;
u is selected from O, 1, 2 and 3; and
v and w are independently selected from 0 and 1.
In further aspects of the invention there is ed a compound of formula (I) for use in the
prophylaxis or treatment of a disease or condition as described herein, methods for the prophylaxis or
treatment of a e or condition as described herein comprising administering to a patient a
compound of a (I), ceutical compositions comprising a compound of fomula (l) and
processes for the synthesis of a compound of formula (I).
DEFINITIONS
Unless the context indicates otherwise, references to formula (I) in all sections of this document
(including the uses, methods and other aspects of the invention) include references to all other sub-
formula, sub-groups, embodiments and examples as defined herein.
“Potency” is a measure of drug ty expressed in terms of the amount required to e an effect
of given intensity. A highly potent drug evokes a larger response at low concentrations. Potency is
proportional to affinity and efficacy. y is the ability ofthe drug to bind to a receptor. Efficacy is the
relationship between receptor occupancy and the ability to te a response at the molecular,
ar, tissue or system level.
The term “antagonist” refers to a type of receptor ligand or drug that blocks or dampens agonist-
mediated biological responses. Antagonists have affinity but no agonistic efficacy for their cognate
receptors, and binding will disrupt the interaction and inhibit the on of any ligand (e.g.
endogenous ligands or substrates, an agonist or inverse agonist) at receptors. The antagonism may
arise ly or indirectly, and may be mediated by any mechanism and at any logical level. As
a result, antagonism of ligands may under different circumstances manifest itself in functionally
different ways. Antagonists mediate their effects by binding to the active site or to allosteric sites on
receptors, orthey may interact at unique binding sites not normally involved in the biological regulation
of the receptor‘s activity. Antagonist activity may be reversible or irreversible depending on the
longevity of the antagonist-receptor complex, which, in turn, depends on the nature of antagonist
receptor binding.
As used herein, the term “mediated", as used e.g. in conjunction with MDM2/p53 as described herein
(and applied for example to s physiological processes, diseases, states, conditions, therapies,
treatments or interventions) is intended to operate tively so that the various processes, diseases,
states, conditions, treatments and interventions to which the term is applied are those in which the
protein plays a biological role. In cases where the term is applied to a disease, state or condition, the
biological role played by the protein may be direct or indirect and may be ary andlor sufficient
for the manifestation of the symptoms of the disease, state or condition (or its aetiology or
progression). Thus, the n function (and in particular aberrant levels of function, e.g. over- or
under-expression) need not necessarily be the proximal cause of the disease, state or condition:
rather, it is contemplated that the mediated diseases, states or ions e those having
multifactorial ogies and complex progressions in which the protein in question is only lly
involved. In cases where the term is applied to treatment, prophylaxis or intervention, the role played
by the protein may be direct or indirect and may be necessary and/or sufficient forthe operation ofthe
treatment, prophylaxis or outcome ofthe intervention. Thus, a e state or condition ed by
a protein includes the development of resistance to any particular cancer drug ortreatment.
The term "treatment" as used herein in the context of treating a condition i.e. state, disorder or
e, pertains generally to treatment and therapy, whether for a human or an animal (e.g. in
veterinary applications), in which some desired therapeutic effect is achieved, for example, the
inhibition ofthe progress ofthe condition, and includes a reduction in the rate of progress, a halt in the
rate of ss, amelioration of the condition, diminishment or alleviation of at least one symptom
associated or caused by the condition being treated and cure ofthe condition. For example, treatment
can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder.
The term ylaxis” (i.e. use of a compound as prophylactic measure) as used herein in the context
of treating a condition i.e. state, disorder or disease, pertains generally to the prophylaxis or
prevention, whether for a human or an animal (e.g. in veterinary applications), in which some desired
preventative effect is achieved, for example, in preventing occurance of a disease or guarding from a
e. Prophylaxis includes complete and total blocking of all symptoms of a disorder for an
indefinite period of time, the mere slowing of the onset of one or l symptoms of the e, or
making the disease less likely to occur.
References to the prophylaxis or treatment of a disease state or condition such as cancer include
within their scope alleviating or ng the incidence e.g. of cancer.
The combinations of the invention may produce a therapeutically efficacious effect relative to the
therapeutic effect of the individual compounds/agents when administered separately.
The term cious’ includes advantageous effects such as additivity, synergism, reduced side
effects, reduced toxicity, sed time to disease progression, increased time of survival,
sensitization or resensitization of one agent to another, or improved response rate. ageously,
an efficacious effect may allow for lower doses of each or either component to be administered to a
patient, thereby decreasing the toxicity of chemotherapy, whilst producing and!or maintaining the
same therapeutic effect. A gistic” effect in the present context refers to a therapeutic effect
produced by the combination which is larger than the sum of the therapeutic effects of the agents of
the combination when presented individually. An “additive” effect in the present context refers to a
therapeutic effect produced by the combination which is larger than the therapeutic effect of any of the
agents of the combination when presented dually. The term “response rate” as used herein
refers, in the case of a solid tumour, to the extent of reduction in the size ofthe tumour at a given time
point, for e 12 weeks. Thus, for example, a 50% response rate means a reduction in tumour
size of 50%. References herein to a “clinical response” referto se rates of 50% or greater. A
“partial response” is defined herein as being a response rate of less than 50%.
As used herein, the term “combination”, as applied to two or more compounds and/or agents, is
intended to define al in which the two or more agents are associated. The terms “combined”
and “combining” in this context are to be reted accordingly.
The association of the two or more compounds/agents in a combination may be physical or non-
al. Examples of physically associated combined compoundsXagents include:
o compositions (e.g. unitary formulations) comprising the two or more compounds/agents in
admixture (for example within the same unit dose);
0 compositions comprising material in which the two or more compoundsiagents are
chemically/physicochemically linked (for example by crosslinking, molecular agglomeration or
g to a common vehicle moiety);
0 compositions comprising material in which the two or more compounds/agents are
chemically/physicochemically co-packaged (for example, disposed on or within lipid vesicles,
particles (e.g. micro- or nanoparticles) or emulsion droplets);
. pharmaceutical kits, pharmaceutical packs or patient packs in which the two or more
nds/agents are co-packaged or co-presented (e.g. as part of an array of unit doses);
Examples of non-physically associated combined compounds/agents include:
0 material (e.g. a non-unitary formulation) comprising at least one of the two or more
nds/agents together with ctions for the extemporaneous ation of the at
least one compound to form a physical association ofthe two or more compounds/agents;
0 material (e.g. a non-unitary formulation) comprising at least one of the two or more
compounds/agents together with instructions for combination therapy with the two or more
compounds/agents;
. material comprising at least one of the two or more compounds/agents together with
ctions for administration to a patient population in which the other(s) of the two or more
compounds/agents have been (or are being) administered;
0 material comprising at least one of the two or more compounds/agents in an amount or in a
form which is cally d for use in combination with the other(s) of the two or more
compounds/agents.
As used herein, the term nation therapy” is intended to define therapies which comprise the use
of a combination of two or more compounds/agents (as defined above). Thus, references to
“combination therapy”, “combinations” and the use of compounds/agents “in combination” in this
application may refer to compounds/agents that are administered as part of the same l treatment
regimen. As such, the posology of each of the two or more compounds/agents may differ: each may
be administered at the same time or at different times. It will ore be appreciated that the
compounds/agents of the combination may be administered sequentially (e.g. before or after) or
simultaneously, either in the same ceutical formulation (i.e. together), or in different
pharmaceutical formulations (i.e. separately). Simultaneously in the same formulation is as a unitary
formulation whereas simultaneously in different pharmaceutical ations is non-unitary. The
posologies of each of the two or more compounds/agents in a ation therapy may also differ
with respect to the route of administration.
As used herein, the term “pharmaceutical kit” defines an array of one or more unit doses of a
pharmaceutical composition together with dosing means (e.g. measuring device) and/or delivery
means (e.g. inhaler or syringe), optionally all contained within common outer packaging. In
pharmaceutical kits comprising a combination of two or more compounds/agents, the individual
compoundSXagents may unitary or non-unitary formulations. The unit ) may be contained within
a blister pack. The pharmaceutical kit may optionally further comprise ctions for use.
As used herein, the term “pharmaceutical pack” defines an array of one or more unit doses of a
ceutical ition, optionally ned within common outer ing. ln pharmaceutical
packs comprising a combination of two or more nds/agents, the individual compounds/agents
may unitary or non-unitary formulations. The unit dose(s) may be contained within a blister pack. The
pharmaceutical pack may optionally r comprise instructions for use.
The term ‘optionally substituted’ as used herein refers to a group which may be unsubstituted or
substituted by a substituent as herein defined.
The prefix “CH” (where x and y are integers) as used herein refers to the number of carbon atoms in a
given group. Thus, a CH; alkyl group contains from 1 to 6 carbon atoms, a 03.6 cycloalkyl group
contains from 3 to 6 carbon atoms, a CM alkoxy group contains from 1 to 4 carbon atoms, and so on.
The term ‘halo’ or ‘halogen’ as used herein refers to ne, ne, bromine or iodine, in particular
fluorine or chlorine.
Each and every hydrogen in the compound (such as in an alkyl group or where referred to as
hydrogen) includes all isotopes of hydrogen, in particular 1H and 2H (deuterium).
The term ‘oxo’ as used herein refers to the group :0.
The term ‘C1_4alkyl’ as used herein as a group or part of a group refers to a linear or branched
saturated hydrocarbon group containing from 1 to 4 carbon atoms respectively. Examples of such
groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert butyl and the like.
The term lkenyl‘ or ‘Cz_6alkenyl‘ as used herein as a group or part ofa group refers to a linear or
branched hydrocarbon group containing from 2 to 4, or 2 to 6 carbon atoms, respectively, and
containing a carbon carbon double bond. Examples of such groups include Cg_4alkenyl or 03.5alkenyl
groups, such as ethenyl (vinyl), 1-propenyl, 2-propenyl (allyl), isopropenyl, butenyl, buta-1,4-dienyl,
pentenyl, and hexenyl.
The term ‘Cz_4alkynyl’ or ‘Cz_6alkynyl’ as used herein as a group or part ofa group refers to a linear or
branched arbon group having from 2 to 4 or2 to 6 carbon atoms, respectively, and containing a
carbon carbon triple bond. Examples of such groups e Cg_4alkynyl or C3,.6alkynyl groups such as
ethynyl and 2 propynyl (propargyl) groups.
The term ‘C1_4alkoxy’ as used herein as a group or part of a group refers to an -O-C1_4alkyl group
wherein C1_4alkyl is as defined herein. Examples of such groups include methoxy, ethoxy, propoxy,
butoxy, and the like.
The term ‘Cg_6cycloalkyl’ as used herein refers to a ted monocyclic hydrocarbon ring of 3 to 6
carbon atoms. Examples ofsuch groups include cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl and
the like.
The term ‘Cg_6cycloalkenyl’ as used herein refers to a partially saturated monocyclic hydrocarbon ring
of 3 to 6 carbon atoms having one or more ly one) carbon carbon double bond(s). Examples of
such groups include cyclopentenyl, cyclohexenyl, and cyclohexadienyl.
The term ‘hydroxyC1_4alky|’ as used herein as a group or part of a group refers to a C1_4a|kyl group as
defined herein wherein one or more (e.g. 1, 2 or 3) than one en atom is replaced with a
hydroxyl group. The term xyC1_4alkyl’ therefore includes monohydroxyC1_4 alkyl, and also
polyhydroxyCM alkyl. There may be one, two, three or more hydrogen atoms replaced with a hydroxyl
group, so the yC1_4alkyl may have one, two, three or more hydroxyl . Examples of such
groups include hydroxymethyl, hydroxyethyl, hydroxypropyl and the like.
The term ‘haloC1_4a|kyl’ as used herein as a group or part of a group refers to a C1_4a|ky| group as
defined herein wherein one or more (e.g. 1, 2 or 3) than one hydrogen atom is replaced with a
halogen. The term ‘haloC1_4alkyl’ therefore includes monohaloC1_4alkyl and also polyhaloC1_4alkyl.
There may be one, two, three or more hydrogen atoms replaced with a halogen, so the haloC1_4alkyl
may have one, two, three or more ns. Examples of such groups e fluoroethyl,
methyl, difluoromethyl, trifluoromethyl or trifluoroethyl and the like.
The term ‘haloC1_4alkoxy’ as used herein as a group or part of a group refers to a -O-C1.4a|ky| group as
defined herein wherein one or more (e.g. 1, 2 or 3) than one hydrogen atom is replaced with a
halogen. The terms ‘haloC1_4alkoxy’ therefore include monohaloC1.4alkoxy, and also polyhaloC1.
4alkoxy. There may be one, two, three or more hydrogen atoms replaced with a halogen, so the
haloC1.4a|koxy may have one, two, three or more halogens. Examples of such groups include
fluoroethyloxy, difluoromethoxy or trifluoromethoxy and the like.
The term “heterocyclyl group” as used herein shall, unless the context indicates othen/vise, include
both aromatic and non-aromatic ring systems. Thus, for example, the term “heterocyclyl group” include
within their scope aromatic, non-aromatic, unsaturated, partially ted and saturated heterocyclyl
ring systems. .
In l, unless the context indicates othenNise, such groups may be monocyclic or
bicyclic (including fused, spiro and bridged bicyclic groups) and may contain, for example, 3 to 12 ring
s, more usually 5 to 10 ring members. Reference to 4 to 7 ring members es 4, 5, 6 or 7
atoms in the ring and reference to 4 to 6 ring s include 4, 5, or 6 atoms in the ring. Examples
of monocyclic groups are groups containing 3, 4, 5, 6, 7 and 8 ring members, more usually 3 to 7, or 4
to 7 and preferably 5, 6 or 7 ring members, more ably 5 or 6 ring members. Examples of bicyclic
groups are those containing 8, 9, 10, 11 and 12 ring members, and more usually 9 or 10 ring
s. The heterocyclyl groups can be heteroaryl groups having from 5 to 12 ring members, more
usually from 5 to 10 ring members. Where reference is made herein to a cyclyl group, the
heterocyclyl ring can, unless the context tes othen/vise, be optionally substituted i.e.
unsubstituted or substituted, by one or more (e.g. 1, 2, 3, or 4 in particular one ortwo) substituents as
defined herein.
The heterocyclyl group can be, for example, a five ed or six ed monocyclic ring or a
bicyclic ure formed from fused five and six membered rings or two fused six membered rings, or
two fused five membered rings. Each ring may contain up to five atoms particularly selected
from nitrogen, sulfur and oxygen and oxidised forms of nitorgen or sulfur. Particularly the heterocyclyl
ring will contain up to 4 heteroatoms, more particularly up to 3 heteroatoms, more usually up to 2, for
example a single heteroatom. In one embodiment, the heterocyclyl ring will contain one or two
heteroatoms selected from N, O, S and oxidised forms of N or S. In one embodiment, the heterocyclyl
ring contains at least one ring nitrogen atom. The nitrogen atoms in the cyclyl rings can be
basic, as in the case of an imidazole or pyridine, or essentially non-basic as in the case of an indole or
pyrrole nitrogen. In general the number of basic nitrogen atoms present in the heterocyclyl group,
including any amino group substituents of the ring, will be less than five.
The heterocyclyl groups can be attached via a carbon atom or a heteroatom (e.g. nitrogen). Equally
the heterocyclyl groups can be substituted on a carbon atom or on a heteroatom (e.g. nitrogen).
Examples of five membered aromatic cyclyl groups include but are not limited to pyrrolyl,
furanyl, thienyl, imidazolyl, nyl, oxazolyl, oxadiazolyl, oxatriazolyl, isoxazolyl, thiazolyl,
thiadiazolyl, isothiazolyl, pyrazolyl, triazolyl and tetrazolyl groups.
Examples of six membered aromatic heterocyclic groups include but are not limited to nyl,
pyrazinyl, pyridazinyl, pyrimidinyl and triazinyl.
The term “heteroaryl” is used herein to denote a heterocyclyl group having aromatic character. The
term “heteroaryl” embraces polycyclic (e.g. ic) ring systems wherein one or more rings are non-
aromatic, ed that at least one ring is ic. In such polycyclic systems, the group may be
attached by the aromatic ring, or by a non-aromatic ring.
Examples of heteroaryl groups are monocyclic and bicyclic groups ning from five to twelve ring
members, and more usually from five to ten ring members.
Examples of five membered heteroaryl groups include but are not limited to pyrrole, furan, thiophene,
imidazole, furazan, oxazole, zole, oxatriazole, ole, thiazole, thiadiazole, isothiazole,
pyrazole, triazole and tetrazole groups.
Examples of six membered heteroaryl groups include but are not limited to ne, pyrazine,
pyridazine, pyrimidine and triazine.
A bicyclic heteroaryl group may be, for example, a group selected from:
a) a benzene ring fused to a 5- or 6-membered ring containing 1, 2 or 3 ring
heteroatoms;
b) a pyridine ring fused to a 5- or 6-membered ring containing 0, 1, 2 or 3 ring
heteroatoms;
c) a pyrimidine ring fused to a 5- or 6-membered ring ning 0, 1 or 2 ring
heteroatoms;
d) a pyrrole ring fused to a 5- or 6-membered ring ning 0, 1, 2 or 3 ring
heteroatoms;
e) a pyrazole ring fused to a 5- or 6-membered ring containing 0, 1 or 2 ring
heteroatoms;
f) an imidazole ring fused to a 5- or 6-membered ring containing 0, 1 or 2 ring
heteroatoms;
g) an e ring fused to a 5- or 6-membered ring containing 0, 1 or 2 ring
heteroatoms;
h) an isoxazole ring fused to a 5- or 6-membered ring containing 0, 1 or 2 ring
heteroatoms;
i) a thiazole ring fused to a 5- or ered ring containing 0, 1 or2 ring heteroatoms;
j) an isothiazole ring fused to a 5- or 6-membered ring containing 0, 1 or 2 ring
heteroatoms;
k) a thiophene ring fused to a 5- or 6-membered ring containing 0, 1, 2 or 3 ring
heteroatoms;
l) a furan ring fused to a 5- or 6-membered ring containing 0, 1, 2 or3 ring heteroatoms;
m) a cyclohexyl ring fused to a 5- or 6-membered ring containing 1, 2 or 3 ring
heteroatoms; and
n) a cyclopentyl ring fused to a 5- or ered ring containing 1, 2 or 3 ring
heteroatoms.
Particular es of ic heteroaryl groups containing a five membered ring fused to another five
membered ring include but are not limited to imidazothiazole (e.g. imidazo[2,1-b]thiazole) and
imidazoimidazole (e.g. imidazo[1,2-a]imidazole).
Particular examples of ic heteroaryl groups containing a six membered ring fused to a five
membered ring include but are not limited to benzofuran, benzothiophene, benzimidazole,
benzoxazole, isobenzoxazole, benzisoxazole, benzothiazole, benzisothiazole, isobenzofuran, indole,
isoindole, indolizine, indoline, isoindoline, purine (e.g., adenine, guanine), indazole, pyrazolopyrimidine
(e.g. pyrazolo[1,5-a]pyrimidine), lopyrimidine (e.g. [1 ,2,4]triazolo[1 yrimidine), benzodioxole,
imidazopyridine and pyrazolopyridine (e.g. pyrazolo[1,5-a]pyridine) groups.
Particular examples of bicyclic heteroaryl groups containing two fused six membered rings include but
are not limited to quinoline, isoquinoline, chroman, roman, isochroman, chromene, isochromene,
ioxan, quinolizine, benzoxazine, pyridine, quinoxaline, oline, cinnoline,
phthalazine, naphthyridine and pteridine groups.
Examples of polycyclic heteroaryl groups containing an aromatic ring and a non-aromatic ring include,
tetrahydroisoquinoline, tetrahydroquinoline, dihydrobenzthiophene, dihydrobenzofuran, 2,3-dihydrobenzo
[1,4]dioxine, benzo[1,3]dioxole, 4,5,6,7-tetrahydrobenzofuran, tetrahydrotriazolopyrazine (e.g.
,6,7,8—tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine), chroman, thiochroman, isochroman, chromene,
isochromene, ioxan, benzoxazine, benzodiazepine, and indoline groups.
A en-containing heteroaryl ring must contain at least one ring nitrogen atom. The nitrogen-
containing heteroaryl ring can be N-linked or C-linked. Each ring may, in addition, contain up to about
four other heteroatoms particularly selected from nitrogen, sulfur and oxygen. Particularly the
heteroaryl ring will contain up to 3 heteroatoms, for e 1, 2 or 3, more usually up to 2 ens,
for example a single nitrogen. The nitrogen atoms in the heteroaryl rings can be basic, as in the case
ofan imidazole or pyridine, or essentially non-basic as in the case of an indole or pyrrole nitrogen. In
general the number of basic nitrogen atoms present in the heteroaryl group, including any amino
group substituents ofthe ring, will be less than five.
Examples of nitrogen-containing heteroaryl groups include, but are not limited to, monocyclic groups
such as pyridyl, pyrrolyl, imidazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, azolyl, isoxazolyl,
thiazolyl, isothiazolyl, furazanyl, lyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, triazolyl (e.g.,
1,2,3-triazolyl, 1,2,4-triazolyl), tetrazolyl, and bicyclic groups such as inyl, isoquinolinyl,
benzimidazolyl, benzoxazolyl, benzisoxazole, benzothiazolyl and benzisothiazole, indolyl, 3H-indolyl,
isoindolyl, indolizinyl, isoindolinyl, purinyl (e.g., e [6-aminopurine], guanine [2-amino
hydroxypurine]), indazolyl, quinolizinyl, benzoxazinyl, benzodiazepinyl, pyridopyridinyl, quinoxalinyl,
quinazolinyl, cinnolinyl, phthalazinyl, naphthyridinyl and pteridinyl.
Examples of en-containing polycyclic heteroaryl groups containing an aromatic ring and a non-
ic ring include tetrahydroisoquinolinyl, tetrahydroquinolinyl, and indolinyl.
The term “non-aromatic” es, unless the context tes othenNise, unsaturated ring systems
without aromatic character, partially saturated and saturated heterocyclyl ring systems. The terms
“unsaturated” and “partially saturated” refer to rings wherein the ring structure(s) contains atoms
sharing more than one valence bond i.e. the ring contains at least one multiple bond e.g. a C=C, C2C
or N=C bond. The term “saturated" refers to rings where there are no multiple bonds between ring
atoms. Saturated heterocyclyl groups e dinyl, morpholinyl, and thiomorpholinyl. lly
saturated heterocyclyl groups include pyrazolinyl, for e pyrazolin-Z-yl and pyrazolinyl.
Examples of non-aromatic heterocyclyl groups are groups having from 3 to 12 ring members, more
usually 5 to 10 ring members. Such groups can be monocyclic or bicyclic, for example, have 3 to 7
ring members in particular 4 to 6 ring members. Such groups particularly have from 1 to 5 or 1 to 4
heteroatom ring members (more usually 1, 2, or 3 heteroatom ring members), usually selected from
nitrogen, oxygen and sulfur and oxidised forms thereof. The heterocyclyl groups can contain, for
example, cyclic ether es (e.g. as in tetrahydrofuran and dioxane), cyclic thioether moieties (e.g.
as in tetrahydrothiophene and ne), cyclic amine moieties (e.g. as in pyrrolidine), cyclic amide
moieties (e.g. as in pyrrolidone), cyclic thioamides, cyclic thioesters, cyclic ureas (e.g. as in
imidazolidinone) cyclic ester moieties (e.g. as in butyrolactone), cyclic sulfones (e.g. as in sulfolane
and sulfolene), cyclic sulfoxides, cyclic sulfonamides and ations thereof (e.g. thiomorpholine).
Particular examples include morpholinyl, piperidinyl (e.g. piperidinyl, piperidinyl, dinyl
and piperidinyl), piperidinonyl, pyrrolidinyl (e.g. pyrrolidinyl, pyrrolidinyl and idinyl),
pyrrolidonyl, azetidinyl, pyranyl (2H-pyran or 4H-pyran), dihydrothienyl, dihydropyranyl, dihydrofuranyl,
dihydrothiazolyl, tetrahydrofuranyl, tetrahydrothienyl, dioxanyl, oxanyl (also known as
tetrahydropyranyl) (e.g. oxanyl), imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, pyrazolinyl,
pyrazolidinyl, piperazinonyl, piperazinyl, and N-alkyl piperazines such as N-methyl piperazinyl. In
general, typical non-aromatic heterocyclyl groups include saturated groups such as piperidinyl,
pyrrolidinyl, azetidinyl, morpholinyl, piperazinyl and N-alkyl piperazines such as N-methyl piperazinyl.
The terms “oxan” and “oxanyl” as used herein referto the group:
which may also be referred to as “tetrahydropyran” ortetrahydropyranyl”.
In a nitrogen-containing non-aromatic heterocyclyl ring the ring must contain at least one ring nitrogen
atom. The en-containing heterocyclyl ring can be N-linked or C-linked. The heterocylic groups
can contain, for example, cyclic amine moieties (e.g. as in idinyl), cyclic amides (such as a
pyrrolidinonyl, piperidinonyl or caprolactamyl), cyclic sulfonamides (such as an isothiazolidinyl 1,1-
dioxide, [1,2]thiazinanyl oxide or [1 ,2]thiazepanyl 1,1-dioxide) and combinations f.
Particular examples of nitrogen-containing non-aromatic heterocyclyl groups include inyl,
morpholinyl, thiomorpholinyl, piperidinyl (e.g. dinyl, piperidin-2yl, dinyl and piperidin
yl), pyrrolidinyl; (e.g. pyrrolidinyl, pyrrolidinyl and pyrrolidinyl), pyrrolidonyl, othiazolyl,
imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, 6H-1,2,5-thiadiazinyl, pyrazolinyl, pyrazolin
yl, pyrazolidinyl, piperazinyl, and N-alkyl piperazines such as N-methyl piperazinyl.
The heterocyclyl groups can be polycyclic fused ring systems or bridged ring systems such as the oxa-
and aza analogues of bicycloalkanes, tricycloalkanes (e.g. adamantane and oxa-adamantane). For
an explanation of the distinction between fused and d ring systems, see Advanced c
Chemistry, by Jerry March, 4th Edition, Vlflley lnterscience, pages 131-133, 1992.
Where, in a tion of a cyclic group or ring, it is stated that the cyclic group ns a certain
number of heteroatom ring members, e.g. as in the phrase “a 5 or6 membered ring containing 0, 1 or
2 nitrogen ring members”, this is to be taken as meaning that apart from the certain number of
heteroatom ring members specified, the remaining ring members are carbon atoms.
The compound of formula (I) may n ted cyclic groups that can be joined to the rest ofthe
molecule by one or more bonds. When the cyclic group is joined to the rest of the molecule by two or
more bonds, these bonds (or two of these bonds) can be made to the same atom (usually a carbon
atom) of the ring or different atoms of the ring. Where the bonds are made to the same atom of the
ring, this results in a cyclic group with a single atom ly a quaternary carbon) bound to two
groups. In other words, when the compound of formula (I) includes a cyclic group that group may
either be linked to the rest of the molecule by a bond or the cyclic group and the rest of the molecule
can have an atom in common e.g. a spiro compound.
The heterocyclyl group can each be unsubstituted or substituted by one or more (e.g. 1, 2 or 3)
substituent groups. For example, heterocyclyl or carbocyclyl groups can be unsubstituted or
substituted by 1, 2, 3 or 4 substituents and particularly it is tituted or has 1, 2 or 3 substituents
as d herein. Where the cyclic group is saturated there may be 2 substituents joined to the same
carbon (where the substituents are the same so called geminal or ‘gem’ disubstitution).
A combination of tuents is permissible only if such as combination results in a stable or
chemically feasible compound (i.e. one that is not substantially altered when kept at 40°C or less for at
least a week).
The various functional groups and substituents making up the compounds of the invention are
particularly chosen such that the molecular weight of the compound ofthe invention does not exceed
1000. More usually, the molecular weight ofthe compound will be less than 750, for example less than
700, or less than 650, or less than 600, or less than 550. More particularly, the molecular weight is
less than 525 and, for example, is 500 or less.
DETAILED DESCRIPTION OF THE INVENTION
The invention es a compound of formula (I):
— (I)
or a tautomer or a solvate or a pharmaceutically acceptable salt thereof, n Het, R1, R2, R3, R4,
R5, R6, R7, a, m, n and s are as defined herein.
The compounds of the formula (I) have a chiral centre, marked below with a “*"2
The compounds of formula (I) include a stereocentre at the position indicated (referred to herein as
(3)) and are chiral non-racemic. Compounds of a (I) have the stereochemistry shown by the
hashed and solid wedged bonds and this stereoisomer predominates.
Typically, at least 55% (e.g. at least 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%) ofthe compound
ofthe a (I) is present as the shown stereoisomer. In one general embodiment, 97% (e.g. 99%)
or more (e.g. substantially all) ofthe total amount ofthe compound of the formula (I) may be present
as a single stereoisomer.
The compounds may also include one or more further chiral centres (e.g. in the -CR5R7OH group
and/or in the R3 group and/or in the —CHR2 group).
Typically, the compound of formula (I) has an enantiomeric excess of at least 10% (e.g. at least 20%,
40%, 60%, 80%, 85%, 90% or 95%). In one general embodiment, the compound of formula (I) has an
enantiomeric excess of 97% (e.g. 99%) or more.
Forthe es ofthis section the isoindolinone ring is numbered as followed:
nds are named in accordance with protocols utilized by chemical naming software packages.
R1 and n
R1 is the substituent(s) on the phenyl group bonded to .
n is 0, 1, 2 or 3. In other words, the phenyl group bonded to -CHR2- group may have 0, 1, 2 or 3
substituents R1.
In one embodiment n is 1, 2 or 3. In one embodiment n is 1 or 2. In another embodiment n is 1.
When n is 2 or 3 (i.e. the phenyl group bonded to -CHR2- group is substituted with more than one R1)
the substituents R1 may be the same or ent (i.e. are independently selected from the definitions
of R1).
R1 may be attached at the ortho (or 0-), meta (or m-) or para (or p—) position of the phenyl group,
wherein the on is defined relative to the point of attachment of the phenyl group to the group -
CHRi.
R1 is independently selected from hydroxy, halogen, nitro, nitrile, C1_4alkyl, haloC1_4alkyl, hydroxyC1_
4alkyl, C2_6alkenyl, C1_4alkoxy, haloC1_4alkoxy, C2_4alkynyl, -Oo,1-(CRXRV)V-COZH, -(CRXRV)V-COZC1_
4alkyl,-(CH2)V-CON(C1.4alkyl)2, -P(=O)(Rx 2, -S(O)d-RX, -S(O)d-heterocyclic group with 3 to 6 ring
members and -S(O)d-N(R8)2.
R1 is ndently ed from hydroxy, halogen, nitro, nitrile, C1_4alkyl, hanC1_4alkyl, hydroxyC1_
4alkyl, Czaalkenyl, C1_4alkoxy, haloC1_4alkoxy, kynyl, -Oo,1-(CH2)V-COZH, -(CH2)V-COZC1_4alkyl,
-(CH2)V-CON(C1_4aIkyI)2, -P(=O)(RX)2, -S(O)d-Rx, -S(O)d-heterocyclic group with 3 to 6 ring members
and -S(O)d-N(R8)2.
R1 is independently selected from hydroxy, halogen, nitro, nitrile, C1_4alkyl, haloC1_4alkyl, hydroxyC1_
4a|ky|, C2_5alkenyl, koxy, haloC1_4alkoxy, C2_4alkynyl, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -(CH2)V-
CON(C1_4aIkyI)2, -P(=O)(RX)2, -RX, -S(O)d-heterocyclic group with 3 to 6 ring members and -
8(O)d-N(R8)2.
In one embodiment, R1 is independently selected from halogen, hydroxy, nitrile, C1_4alkyl, C2_4alkynyl,
or C1_4alkoxy, for example R1 is ndently selected from chloro, hydroxy, nitrile, methyl or
In one embodiment R1 is ndently selected from halogen (e.g. chloro), C1_4alkyl (e.g. methyl), C1.
4alkoxy (e.g. methoxy), -Oo,1-(CRXRV)V-COZH (e.g. -COZH, -(CH2)-COZH, -(C(CH3)2)-COZH, or -O(CH2)-
COZH) or -S(O)d-Rx(e.g. 802CH3).
In one embodiment R1 is 00,1-(CRXRV)V-COZH in particular -COZH, -(CH2)-COZH, -(C(CH3)2)-COZH, or
-O(CH2)-C02H), such as -(C(CH3)2)-COZH.
In one ment, R1 is chloro or nitrile, in particular chloro.
In one embodiment, R1 is nitro (e.g. p—NOZ).
In one embodiment, R1 is nitro at the ortho or meta position.
In one embodiment, R1 is independently selected from hydroxy, n, nitrile, C1_4alky|, haloC1.
4alkyl, hydroxyC1_4alkyl, 02.6alkenyl, C1_4alkoxy, haloC1_4alkoxy, C2_4alkynyl, -(CH2)V-C02H, -(CH2)V-
COZC1_4a|kyl, V-CON(C1_4a|kyl)2, -P(=O)(RX)2, -S(O)d-C1_6alky|, -S(O)d-heterocyclic group with 3 to
6 ring members and -N(R8)2.
In another embodiment, n is 1 and R1 is chloro or nitrile.
In another embodiment, n is 1 and R1 is chloro.
In another embodiment, n is 1 and R1 is e.
In one embodiment, one of the R1 groups or the R1 group (where n=1) is at the para-position (i.e. para
to the point of attachment ofthe phenyl ring). In one embodiment n is 1 and R1 is p-chloro or p—nitrile.
In one embodiment, n is 1 and R1 is halogen (e.g.Cl or F), nitrile, C1_4alkoxy (e.g. -OCH3) or kyl
(e.g. -CH3).
In one embodiment, R1 is -S(O)d-C1_6alkyl, or -S(O)d-heterocyclic group with 3 to 6 ring members and -
S(O)d-N(R8)2. In one embodiment, R1 is -S-C1_6alkyl, -S(O)-C1_6alkyl, -S(O)2-C1_6alky|, -S(O)dheterocyclic
group with 3 to 6 ring members or -S(O)d-N(C1_6alkyl)2.
In another embodiment, R1 is -S-CH3, -S(O)-CH3, -S(O)2-CH3, or -S(O)2-morpholinyl.ln r
embodiment, one or more R1 is -SOZCH3, or -SOz-heterocyclic group with 6 ring members eg.
(morpholinyl), in particular -SOz-(1-morpholinyl).
In one embodiment, R1 is o-(-S(O)d-C1_4alkyl) or o-(-S(O)d-heterocyclic group with 3 to 6 ring
members). In one embodiment, R1 is o-S-C1_4alkyl, o-(-S(O)d-C1_4alkyl) or o-(-S(O)d-heterocyclic group
with 3 to 6 ring members). In one embodiment, R1 is o-(-S(O)2-CH3)
In one embodiment, R1 is -(CH2)u-COZH. In one embodiment, R1 is -COZH. In one embodiment, R1 is
-(CH2)u-COZH at the meta or para on. In one embodiment, R1 is
-(CH2)u-COZH at the ortho position.
In one embodiment, R1 is independently selected from hydroxy, halogen, nitrile, C1_4alkyl, haloC1_
4a|ky|, hydroxyC1_4alkyl, Cz_5alkenyl, C1_4alkoxy, haloC1_4a|koxy, kynyl, -(CH2)V-COZC1_4alkyl, -
(CH2)V-CON(C1_4alkyl)2, -P(=O)(RX)2, -S(O)d-C1_6alkyl, -heterocyclic group with 3 to 6 ring
members and -S(O)d-N(R8)2.
In one embodiment, n is 2. In one ment when n is 2, the phenyl group is substituted with (i) o-
(-S(O)d-C1.4alkyl) or O)d-heterocyclic group with 3 to 6 ring members) and (ii) halogen (e.g. CI or
F), nitrile, or C1_4 alkyl, in particular chloro, nitrile or methyl.
In one embodiment, n is 2 and R1 is (i) -SOZCH3 and (ii) .
In one embodiment n is 2 and R1 is (i) -SOZCH3 and (ii) chloro, nitrile or methyl.
In one embodiment, n is 2 and R1 is (i) -COzH and (ii) chloro.
In one embodiment n is 2 and R1 is (i) -COZH and (ii) chloro, or nitrile.
In one embodiment, when n is 2, the the phenyl group bonded to -CHR2- is substituted with (i)
hydroxyl and (ii) halogen (e.g. Cl or F), or nitrile, in particular chloro, or e.
In one embodiment, the phenyl group bonded to -CHR2- and R1 form a group:
80sz
wherein in particular, R1 is halogen (for example chloro), nitrile or C1_4alkyl (for example -CH3) and RX
is C1_4alkyl (for example -CH3).
In one embodiment, the phenyl group bonded to -CHR2- and R1 form a group:
50sz
wherein in particular, R1 is C1_4alkyl (for example -CH3) and RX is C1_4alkyl (for example -CH3).
In one embodiment when n is 2, the phenyl group is substituted with (i) o-OH or o-CHZOH and (ii)
halogen (e.g. Cl or F), e, or CH alkyl, in particular chloro, or nitrile. In one embodiment, when n is
2, the phenyl group is substituted with (i) y and (ii) halogen (e.g. CI or F) or e, in particular
chloro or nitrile. In one embodiment, when n is 2, the phenyl group is substituted with (i) o-hydroxy
and (ii) p—Cl or p—CN (e.g. p—Cl).
In one embodiment, n is 2 and R1 is e (e.g. at the ortho and para positions ofthe phenyl group).
In one embodiment, R1 is halogen (e.g.Cl or F), C1_4alkynyl (e.g. -CECH), nitrile, -(CH2)VCOOH (e.g.
-COOH) or SOZC1_4alkyl (e.g. SOZCHg) and n is 1 or 2.
In one embodiment, R1 is halogen (e.g.Cl), C1_4alkynyl (e.g. -CECH), nitrile, , yC1_4alkyl (e.g.
CHZOH), VCOOH (e.g. -COOH), -S(O)d-C1_4alkyl (e.g. SCHg, SOCH3, or SOch3), -SOz-(1-
linyl) or -P(=O)(RX 2, (e.g. -P(=O)(CH3)2)-
In one embodiment, n is 1 and R1 is Cl (e.g. p—Cl), CN (e.g. p—CN), or CZ_4alkynyl (e.g. p— C1alkynyl), or
n is 2 and (i) R1 is p-Cl, o-CHZOH; (ii) p—CN, o-CHZOH; or (iii) p—CI, o-COOH, (iv) p-Cl, o-SCH3, (v) p-
Cl, o—S(O)CH3, (vi) p—Cl, O'SOZCH3, (vii) p-Cl, o-SOz-(1-morpholinyl), or (viii) p—Cl, o—P(O)(CH3)2.
In one embodiment, n is 1 and R1 is Cl (e.g. p—Cl), CN (e.g. p—CN), or CZ_4alkynyl (e.g. p— C1alkynyl).
In one embodiment, n is 2 and (i) R1 is p-Cl, o-CHZOH; (ii) p-CN, o-CHZOH; or (iii) p-CI, o-COOH, (iv)
p—Cl, , (v) p—Cl, o-S(O)CH3, (vi) p—Cl, O'SOchg, (vii) p—Cl, o-SOz-(1-morpholinyl), or (viii) p-Cl,
0-P(O)(CH3)2.
In one embodiment n is 1 and R1 is -Cl, -CN or -OMe (e.g. p—Cl, p—CN or p—OMe). In one embodiment
n is 1 and R1 is -CI or -CN (e.g. p—Cl or p—CN).
In one embodiment, n is 2. When n is 2, typically the phenyl group is substituted at the o- and p-
positions. In particular, n is 2 and R1 is substituted by a p-chloro and either o-(-S(O)d-C1_4alkyl) or o-(-
S(O)d-heterocyclic group with 3 to 6 ring members).
In one embodiment, n is 2 and R1 is o-COZH and p-chloro.
In one embodiment, n is 2 and R1 is o-COZH and p-nitrile.
In one embodiment, n is 2 and R1 is H and p—chloro.
In one embodiment, n is 2 and R1 is o—CHZOH and p—nitrile.
In one embodiment, n is 2 and R1 is o-OH and p-chloro.
In one embodiment, n is 2 and R1 is o—OH and p—nitrile.
In one embodiment, n is 2 and R1 is O'SOchg and p—chloro.
In one embodiment n is 2 and R1 is -SOz-(1-morpholinyl) and p—chloro.
In one embodiment, R1 is CRXRV)VCOOH (e.g. -COOH, -CH2COOH, -OCHZCOOH or
-C(CH3)2COOH
In one embodiment, n is 2 and R1 is p—CI and o-Oo,1(CRXRy)VCOOH (e.g. -COOH, -CHZCOOH,
-OCH2COOH or -C(CH3)2COOH).
In one embodiment, R1 is halogen (e.g. CI), hydroxyalkyl (e.g. -CHZOH), C1_4alkynyl (e.g. -CECH),
nitrile, -Ooy1(CRXRy)VCOOH (e.g. -COOH, -CH2COOH, -OCH2COOH or -C(CH3)2COOH) or -SOZC1_
4alky| (e.g. -SOZCH3) and n is 1 or 2.
In one embodiment, R1 is halogen (e.g. CI), hydroxyalkyl (e.g. -CHZOH), C1_4alkynyl (e.g. -CECH),
nitrile, -(CH2)VCOOH (e.g. -COOH) or-SOZCMaIkyI (e.g. -SOZCH3) and n is 1 or 2.
In one embodiment, R1 is independently selected from y, halogen, e, C1_4alkyl, haloC1_
4alkyl, yC1_4alkyl, Cz_6alkenyl, C1_4a|koxy, hanC1_4alkoxy, kynyl, -(CH2)V-COZH, -Oo,1-
)V-COZC1_4a|kyI (e.g. -(CH2)V-COZC1_4aIkyI), -(CH2)V-CON(C1_4aIkyI)2, -P(=O)(RX)2, -S(O)d-C1_
6alkyl, -S(O)d-heterocyclic group with 3 to 6 ring members and -S(O)d-N(R8)2.
In one embodiment wherein n is 2, and one R1 is -Oo,1-(CRXRY)V-COZC1.4alkyl, o-(-S(O)d-C1_4a|kyl) or o-
(-S(O)d-heterocyclic group with 3 to 6 ring members) and one R1 is halogen (e.g. CI or F), nitrile, or C1.
4 alkyl, in particular chloro, nitrile or methyl.
In one embodiment wherein n is 2, and one R1 is o-(-S(O)d-C1_4alkyl) or O)d-heterocyclic group
with 3 to 6 ring members) and one R1 is halogen (e.g. CI or F), nitrile, or 01.4 alkyl, in particular chloro,
nitrile or methyl.
In one embodiment wherein n is 2, and one R1 is -Ooy1-(CRXRV)V-COZC1_4aIkyl, and one R1 is n
(e.g. CI or F), nitrile, or CH alkyl, in particular chloro, nitrile or , such as chloro.
R2 is selected from hydrogen, CM alkyl, C2_6alkenyl, yC1_4alkyl, -(CRXRy)u-COZH, -(CRXRy)u-
COZCMalkyl, and -(CRXRy)u-CONRXRV.
In one embodiment u is selected from 0, 1, or 2. In one ment u is selected from 0 or 1.
In one ment, R2 is selected from hydrogen, CM alkyl, C2_6alkenyl, hydroxyC1_4alkyl and -
(CRXRy)u-002H. In one embodiment, R2 is selected from hydrogen, C1_4 alkyl, hydroxyC1_4alkyl and -
)u-COZH. In one embodiment, R2 is selected from hydrogen, C1_4 alkyl, C2_6alkenyl, and
hydroxyC1,4alkyl. In another embodiment R2 is selected from hydrogen and -(CH2)u-COZH (e.g. -CH2-
COZH).
In one embodiment, R2 is hydrogen, 01.4 alkyl (e.g. -CH3), hydroxyC1_4alkyl (e.g. CHZOH) or
-(CH2)uCOOH (e.g. -COOH, -CHZCOOH, 2-COZH, -(CH(CH3))-COZH or 3)2-COZH, such
as -COOH, -CH2COOH, -CH2CH2-COZH, or -(CH(CH3))-C02H).
In one ment, R2 is selected from hydrogen, C1_4alkyl, 02.6alkenyl, and hydroxyC1_4alkyl.
In one embodiment, R2 is hydrogen, CM alkyl (e.g. -CH3), hydroxyC1_4alkyl (e.g. CHZOH) or
-(CH2)UCOOH (e.g. -CHZCOOH). In one embodiment, R2 is selected from hydrogen, -CH3, -CH20H,
and -CH2COZH.
In one embodiment, R2 is selected from hydrogen, -CH3, -CHZOH, -CH=CH2 and -CH(OH)CHZOH.
In one embodiment, R2 is selected from hydrogen, -CH3, -CHZOH, and -CH2COZH.
In one embodiment, R2 is en or C1_4 alkyl (e.g. -CH3 or -CH2CH3).
In one ment, R2 is selected from hydrogen, -CH3 and -CH2CH3. In one ment, R2 is
selected from hydrogen and methyl.
In one embodiment, R2 is selected from hydrogen and -(RXRy)u-COZH (e.g. -COOH, -CHZCOOH,
-CH2CH2-COZH, -(CH(CH3))-COZH and -(C(CH3)2-COZH).
In one ment, R2 is -(RXRy)uCOOH (e.g. -CH2COOH, -CH2CH2-COZH, -(CH(CH3))-COZH (e.g.
ACOH) or -(C(CH3)2-COZH).
In one embodiment, R2 is hydrogen, C1_4 alkyl (e.g. -CH3) or-(CH2)uCOOH (e.g. -CH2COOH, -CHZCH2-
COZH or-(CH(CH3))-COZH).
In one embodiment, R2 is hydrogen, C1_4alkyl (e.g. -CH3) 2)uCOOH (e.g. -CH2COOH).
In one embodiment, R2 is -(CRXRy)u-COZH (e.g. -CH2-COZH).
In another embodiment, R2 is selected from -(CH(CH3))-COZH and -(C(CH3)2-002H) (e.g. /\C°2H,
/'\COH or -(C(CH3)2-C02H.
In another embodiment, R2 is hydrogen and the compound of formula (I) is a compound of formula (le)
or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
— ('6)
When R2 is other than hydrogen, the compound of formula (I) can exist as at least two
diastereoisomers:
\/(R5)m
F \WL / \
s“ R2
R6 N
/ \ (R1)n
Diastereoisomer 1A
)8 / \/(R5>m
F \
\s‘ R2
R7 N
/ \ (R1)n
Diastereoisomer 18
For the avoidance of doubt, the general a (I) and all subformulae cover both individual
diastereoisomers and mixtures of the diastereoisomers which are related as epimers at the -CHR2-
group. In one embodiment the compound of formula I is diastereoisomer 1A or a er or a solvate
or a pharmaceutically able salt thereof. In one embodiment the nd of formula I is
diastereoisomer 1B or a tautomer or a solvate or a pharmaceutically able salt thereof.
In one embodiment, the compound is diastereoisomer 1A and R2 is selected from:
i. CM alkyl, CZ_6alkenyl, hydroxyC1_4alkyl, -(RXRy)u-COZH (e.g. -COOH, -CH2COOH, -CH2CH2-
COZH, -(CH(CH3))-COZH and -(C(CH3)2-COZH), -(CH2)u-COZC1_4alkyl, and -(CH2)u-CONRXRV;
ii. 01.4 alkyl, C2.5alkenyl, and hydroxyC1.4alkyl.
In one embodiment, the compound is diastereoisomer 1A and R2 is selected from:
i. C14 alkyl, C2_6alkenyl, hydroxyC1_4alkyl, -(CH2)u-COZH, -(CH2)u-COZC1_4alkyl, and -(CH2)u-
Y; or
ii. C1_4alkyl, 02.6alkenyl, and hydroxyC1_4alkyl.
In another ment R2 is selected from hydrogen and -(RXRy)u-COZH (e.g. -COOH, -CHZCOOH,
-CH2CH2-COZH, -(CH(CH3))-COZH and -(C(CH3)2-COZH),
In r ment R2 is selected from hydrogen and -(CH2)u-COZH (e.g. -CH2-COZH).
In one embodiment, the compound is diastereoisomer 1A and R2 is selected from:
i. -CH3, -CHZOH, -CH=CH2 and -CH(OH)CHZOH; or
ii. 01.4 alkyl (e.g. -CH3 or -CHZCH3); or
iii. -CH3 and -CH2CH3.
In one embodiment, the compound is diastereoisomer1B and R2 is selected from:
i. C14 alkyl, Cz_6alkenyl, hydroxyC1_4a|kyl, -(RXRy)u-COZH (e.g. -COOH, -CH2COOH, -CHZCH2-
COZH, -(CH(CH3))-COZH and -(C(CH3)2-COZH), -(CH2)u-COZC1_4alkyl, and -(CH2)u-CONRXRV;
ii. C14 alkyl, 02.6alkenyl, and hydroxyC1_4alkyl.
In one embodiment, the compound is reoisomer1B and R2 is selected from:
i. C14 alkyl, C2_6alkenyl, hydroxyC1_4alkyl, -(CH2)u-COZH, -(CH2)u-COZC1_4alkyl, and -(CH2)u-
CONRXRV; or
ii. C1_4alkyl, Cz_6alkenyl, and hydroxyC1_4alkyl.
In another embodiment R2 is selected from hydrogen and u-COZH (e.g. -CH2-COZH).
In one embodiment, the compound is diastereoisomer1B and R2 is selected from:
i. -CH3, -CHZOH, -CH=CH2 and -CH(OH)CHZOH; or
ii. CH alkyl (e.g. -CH3 or -CH2CH3); or
iii. -CH3 and -CH2CH3.
In r embodiment R2 is selected from hydrogen and -(RXRy)u-COZH (e.g. -COOH, -CH2COOH,
-CH2CH2-COZH, -(CH(CH3))-COZH and -(C(CH3)2-COZH),
In one embodiment R2 is selected from C1_4 alkyl, yC1_4a|kyl, -(CH2)u-COZH, -(CH2)u-COZC1_
alkyl, and -(CH2)W-CONR"Ry (in particular -CH2-C02H) and the compound is diastereoisomer1A.
In one embodiment R2 is ed from C1_4 alkyl, hydroxyC1_4a|kyl, -(CH2)u-COZH, -(CH2)u-COZC1_
4alkyl, and -(CH2),,-CONRXRy (in particular -CH2-CozH) and the compound is diastereoisomer 1B.
In one embodiment R2 is yC1_4alkyl (e.g.-CHZOH) and the compound is diastereoisomer 1A.
In one embodiment R2 is -(CH2)u-COZH (e.g.-CHz-COZH) and the compound is diastereoisomer 1A.
In one embodiment R2 and the hydrogen on the carbon to which it is attached are 2H (i.e. deuterium).
2016/053042
R3 and s
R3 is hydrogen or -(A)1-(CRXRy)q-X;
s is selected from 0 and 1;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then s is 1 and q is otherthan 0;
A is a Cg_6cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group comprises one or more , 2, or 3) heteroatoms selected from N, O, S and oxidised forms
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -S(O)d-RX,
-C(=O)-C1_4alkyl, -S(O)d-N(H)e(C1_4a|ky|)2.e, -NRXRy and -C(=O)NRXRV;
, -NHSOzRX, -NRXCORy,
R9 is independently selected from hydrogen, C1_5alkyl, haloC1_5alkyl, hydroxyC1_6alkyl, -(CH2)k-O-C1_
6alkyl, -(CH2)k-O-(hydroxyC1_6alkyl), hydroxyC1_6alkoxy, -(CH2)k-COZC1_6alky|, -(CH2)k-COZH, -C1_6 alkyl-
N(H)e(C1_4alkyI)2_e, -(CH2)j-C3_8cycloalkyl and -(CH2)j-Cg.8cycloalkenyl;
RX and Ry are independently selected from hydrogen, halogen, nitro, nitrile, C1_6alky|, haloC1_6alkyl, C2.
yl, C2_5alkynyl, hydroxy, yC1_5alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_6alkoxy, -
COOC1.sa|ky|, (C1.4a|ky|)2.e, -C1.ealkyl-N(H)e(C1.4a|ky|)2.e, -(CH2)k-C(=O)N(H)e(C1.4a|ky|)2.e, C3-
gcycloalkyl and cloa|kenyl;
or the RX and Ry groups, er with the carbon or nitrogen atom to which they are attached, can
join to form a Cg_5cycloalkyl or saturated heterocyclyl group with 3 to 6 ring members which may be
optionally fused to an aromatic heterocyclyl group of 3 to 5 ring members or can join to form a =CH
group;
j, d, and e are independently selected from 0, 1 and 2;
k is ed from 1 and 2; and
v is independently selected from 0 and 1.
In one embodiment when t is 1 the group -(CRXRy)q-X and the rest ofthe molecule are attached to the
same carbon atom in the group A. In one embodiment when t is 1 the group (CRXRy)q-X and the rest of
the molecule are attached to different carbon atoms in the group A.
In one embodiment, R3 is hydrogen or -(A)t-(CRXRy)q-X;
s is selected from 0 and 1;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is otherthan 0 and (ii) when t is 0
then sis 1 and q is han 0;
A is a Cg_6cycloalky| group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group comprises one or more (e.g.1, 2, or 3) atoms selected from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -S(O)d-RX,
-C(=O)-C1_4a|kyl, -S(O)d-N(H)e(C1_4a|kyl)2.e, -NRXRy and -C(=O)NRXRV;
, RX, -NRXCORy,
R9 is independently selected from hydrogen and C1_6alkyl;
RX and Ry are ndently selected from en and C1_6alkyl;
d and e are ndently selected from 0, 1 and 2;
v is independently selected from 0 and 1.
In one embodiment, R3 is en or -(A)1-(CRXRy)q-X;
s is selected from 0 and 1;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then s is 1 and q is otherthan 0;
A is a C3_5cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -C(=O)—C1_
4a|ky|, , -NR"CORy, and -C(=O)NRXRY;
R9 is independently selected from hydrogen and C1_6alkyl;
RX and Ry are ndently selected from hydrogen and C1_6alkyl;
v is independently selected from 0 and 1.
In one embodiment, R3 is hydrogen or -(A)1-(CRXRy)q-X;
s is selected from 0 and 1;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)t-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then s is 1 and q is otherthan 0;
A is a C3_5cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, n, -CN, -OR9, Ry, and -C(=O)NRXRy;
R9 is independently selected from hydrogen and C1_6alkyl;
RX and Ry are independently selected from hydrogen and C1_6alkyl;
v is independently selected from 0 and 1.
In one embodiment, R3 is hydrogen or -(A)t-(CRXRy)q-X;
s is selected from 0 and 1;
t is selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then sis 1 and q is otherthan 0;
WO 55860
A is a 03.5cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group comprises one or more , 2, or 3) heteroatoms selected from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, halogen (e.g. fluoro), -OR9, -NRXCOR"; and -C(=O)NRXRy;
R9 is independently selected from hydrogen and C1_6alkyl;
RX and Ry are independently selected from hydrogen and C1_6alkyl;
v is independently selected from 0 and 1.
In one embodiment, R3 is hydrogen and s is 1 Le. the moiety -(CH2)SR3 is -CH3.
In one embodiment, R3 is hydrogen and s is 0 Le. the moiety -(CH2)SR3 is -H.
In one embodiment, t is 1 and A is a C3_ecycloalkyl group or a cyclic group with 3 to 6 ring
s, wherein the heterocyclic group comprises one or more (e.g.1 or 2) heteroatoms selected
from N, O, S and oxidised forms thereof.
In one embodiment, t is 1 and A is a C3,.6cycloalkyl group. In one embodiment, A is a Cg_5cycloalkyl
group. For example, A is selected from a cyclopropyl group, a cyclobutyl group and a entyl
group. In one ment, A is a cyclopropyl group. In one embodiment, A is a cyclobutyl group.
In particular, t is 1 and A is cyclopropyl.
In one embodiment, t is 1 and A is a heterocyclic group with 3 to 6 ring members, wherein the
heterocyclic group comprises one or more (e.g. 1, 2, or 3) heteroatoms selected from N, O, S and
oxidised forms thereof.
In one embodiment, t is 1 and A is a heterocyclic group with 3 to 5 ring s, wherein the
cyclic group comprises one or more (e.g. 1, 2, or 3) heteroatoms selected from N, O, S and
oxidised forms thereof.
In one embodiment, t is 1 and A is an unsaturated heterocyclic group with 3 to 5 ring s,
wherein the heterocyclic group comprises one or more (e.g. 1, 2, or 3) heteroatoms ed from N,
O, S and oxidised forms thereof, in particular 0.
In one embodiment, t is 1 and A is a saturated heterocyclic group with 3 to 5 ring members, wherein
the heterocyclic group comprises one or more (e.g. 1, 2, or 3) heteroatoms ed from N, O, S and
oxidised forms thereof, in particular 0.
In one embodiment, t is 1 and A is a heterocyclic group which is selected from morpholinyl, piperidinyl
(e.g. piperidinyl, piperidinyl, piperidinyl and piperidinyl), piperidinonyl, pyrrolidinyl (e.g.
idinyl, pyrrolidinyl and pyrrolidinyl), pyrrolidonyl, azetidinyl, oxetanyl, pyranyl (2H-pyran
or 4H-pyran), dihydrothienyl, dihydropyranyl, dihydrofuranyl, dihydrothiazolyl, tetrahydrofuranyl (e.g.
tetrahydrofuranyl), tetrahydrothienyl, dioxanyl, oxanyl (e.g. oxanyl), imidazolinyl, imidazolidinonyl,
oxazolinyl, thiazolinyl, pyrazolinyl, pyrazolidinyl, piperazinonyl, piperazinyl, and N-alkyl piperazines
such as N-methyl piperazinyl.
In one embodiment, t is 1 and A is a heterocyclic group which is selected from morpholinyl, piperidinyl
(e.g. piperidinyl, piperidinyl, piperidinyl and piperidinyl), piperidinonyl, pyrrolidinyl (e.g.
pyrrolidinyl, pyrrolidin-Z-yl and pyrrolidinyl), idonyl, azetidinyl, oxetanyl, pyranyl ran
or 4H-pyran), dihydropyranyl, ofuranyl, dihydrothiazolyl, tetrahydrofuranyl (e.g. tetrahydrofuran-
3-yl), dioxanyl, oxanyl (e.g. oxanyl), imidazolinyl, imidazolidinonyl, oxazolinyl, pyrazolinyl,
pyrazolidinyl, piperazinonyl, piperazinyl, and N-alkyl piperazines such as N-methyl piperazinyl.
In particular, t is 1 and A is a heterocyclic group which is oxetanyl (e.g. oxetanyl).
In ular, t is 1 and A is a heterocyclic group which is tetrahydrofuranyl (e.g. ydrofuranyl).
In one embodiment, X is hydrogen, s is 0 and q is 0, and R3 is a cyclic group with 3 to 6 ring
members, n the heterocyclic group comprises one or more (e.g.1, 2, or 3) heteroatoms selected
from N, O, S and oxidised forms thereof. In particular, R3 is tetrahydrofuranyl (e.g. tetrahydrofuran
yl).
In one ment, s is 0 and t is 1 and A is attached directly to the oxygen atom bound to the
isoindolinone. In one embodiments is 1 and the cycloalkyl group is attached via a methylene group
(i.e. -CH2-) to the oxygen atom bound to the isoindolinone.
In one embodiment, A is tetrahydrofuranyl and X is hydrogen.
In one embodiment A is selected from cyclopropyl, oxetanyl and tetrahydrofuranyl.
In one embodiment, q is 0. In one ment, q is 1. In one embodiment, q is 2.
In one ment, A is oxetanyl and X is fluorine.
When q is not 0, RX and Ry are ed from hydrogen, halogen (e.g. fluorine), hydroxy and methyl
e.g. hydrogen and methyl, in ular hydrogen.
In one embodiment, q is 1 and at least one RX and Ry is hydrogen. In one embodiment, q is 2 and at
least two RX and Ry are hydrogen e.g. three RX and Ry are hydrogen.
In one embodiment, -(CRXRy)q- is selected from -CH2- and -CH2CH2-.
In one embodiment, RX and Ry together form a saturated heterocyclyl group with 3 to 6 ring members.
In one embodimentt is 0 and -(CRXR-‘/)q- is -CH2-. In one embodiment t is 0, s is 0, -(CRXRy)q- is -CH2-
and X is y.
In one embodiment, X is selected from -CN, -OH, -O-C1_4alkyl, -O-hydroxyC1_4alkyl, -S(O)d-C1_4alkyl, -
C(=O)-C1_4a|kyl, -NR"Ry -NR"CORy and -C(=O)NRXRV.
In one embodiment, X is selected from -CN, -OH, -O-CH2CHZOH,-S(O)d-C1_4alkyl and
-C(=O)NR"Ry (e.g. -C(=O)NH2 or -C(=O)NH(CH3)). In one embodiment X is selected from
-CN, -OH, -C(=O)NH2 or-C(=O)NH(CH3).
In one embodiment, X is selected from hydrogen, halogen, -CN, -OR9, and NRXRy. In another
embodiment, X is selected from hydrogen, halogen, -CN, -OH, -OCH3, and -C(=O)NH2. In another
ment, X is selected from hydrogen, fluorine, -CN, -OH, and NH2.
In one embodiment, X is selected from hydrogen, fluorine, -CN, -OH and -C(=O)NH2. In one
embodiment, X is ed from hydrogen, -CN, -OH and -C(=O)NH2. In one embodiment, X is
selected from -CN, -OH and -C(=O)NH2.
In one embodiment X is selected from -OH and -C(=O)NH2 e.g. -OH.
In one embodiment, X is -C(=O)NR"Ry (e.g. -C(=O)NH2 or -C(=O)NH(CH3).
In one embodiment, RX and Ry are hydrogen, n (e.g. fluorine), hydroxy and methyl. In one
embodiment, RX and Ry are hydrogen and methyl. In one embodiment, RX and Ry together form a
saturated cyclyl group with 3 to 6 ring members.
In one embodiment, A is a 03.5cycloalkyl group (i.e. g is 1, 2 or 3) and t is 1 and s is 0 or 1, and the
compound of formula (I) is a compound of formula (If) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
V)q-X
— ('1‘)
In one embodiment, A is a C3,.6cycloalkyl group (i.e. g is 1, 2 or 3) and t is 1 and s is 1, and the
compound of formula (I) is a compound of formula (lg) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
(CRXRy)q-X
g / \(mom
\\“\
In one embodiment, A is a C3,.6cycloalkyl group (i.e. g is 1, 2 or 3) and t is 1 and s is 0, and the
compound of formula (I) is a nd of formula (lg’) or a tautomer or a solvate or a
ceutically acceptable salt thereof:
In one embodiment, the nd of formula (I) is a compound of formula (lg’) and g is 2.
In one embodiment, A is a C3,.6cycloalkyl group (i.e. g is 1, 2 or 3) and t is 1 and s is 1, and the
cycloalkyl group is geminally disubstituted (i.e. the group -(CRXR”)q-X and the -CH2-O-isoindolinone
group are both attached to the same atom ofthe cycloalkyl group), and the compound of formula (I) is
a compound of formula (lh) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
— (lh)
In one embodiment, A is a cyclopropyl group (i.e. g is 1), t is 1 and s is 1. Therefore the cycloalkyl
group is a cyclopropyl group and the compound of formula (I) is a nd of formula (Ii) or a
tautomer or a solvate or a ceutically acceptable salt thereof:
In one embodiment, A is a Cg_6cycloalkyl group (i.e. g is 1, 2 or 3), t is 1, sis 1 and X is hydroxy, and
the compound of formula (I) is a compound of the formula (Ij) or a tautomer or a solvate or a
pharmaceutically acceptable salt f:
(CRXRV)q-OH
— (Ij)-
In one ment, A is a 03.6cycloalkyl group (i.e. g is 1, 2 or 3), t is 1, s is 1 and X is -C(=O)NH2 and
the compound of formula (I) is a compound of the formula (lk) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof:
(CRXRy)q—C(=O)NH2
\/(R5)m
In one embodiment, A is a Cg_6cycloalkyl group (i.e. g is 1, 2 or 3), t is 1, s is 1 and X is —CN and the
compound of formula (I) is a compound of the a (lk’) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof:
(CRXRy)q-CN
In another embodiment, A is a C3_5cycloalkyl group (i.e. g is 1, 2 or 3), t is 1, s is 1 and RX and Ry are
en (including 1H and 2H) and the compound of formula (I) is a compound of formula (IL) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
In one embodiment, A is a cyclopropyl or cyclobutyl group (i.e. g is 1 or 2), t is 1, s is 1 and X is
hydroxy and the compound of formula (IL) is a compound of formula (Im) or a tautomer or a solvate or
a pharmaceutically acceptable salt thereof:
— (Im).
In one embodiment, g is 1 and the compound of formula (Im) is a compound of the a (Im’) or a
tautomer or a solvate or a pharmaceutically able salt thereof:
In one embodiment, A is a Cg-cycloalkyl group (i.e. g is 1), t is 1, s is 1 and X is -C(=O)NH2 and the
compound of formula (I) is a compound of a (In) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
wherein q is 0 or 1. In one embodiment of the compound (In), q is 0.
In one embodiment, A is a C3-cycloalkyl group (i.e. g is 1), t is 1, s is 1 and X is -CN and the
compound of formula (I) is a compound of formula (In’) or a tautomer or a solvate or a
ceutically acceptable salt thereof:
(R5)m
(In’).
wherein q is 0 or 1. In one embodiment of the compound (In), q is 0.
In one embodiment of formula (I) and subformulae thereof, the ens in the -(CRXRy)- group of R3
are 2H (i.e. deuterium, D). In one embodiment, the hydrogens in the group -CH2-O group are 2H (i.e.
deuterium, D). In one embodiment, the hydrogens in the -(CRXRy)- and -CH2-O groups are 2H (i.e.
deuterium, D).
In one embodiment q is 0 or1 and RX and Ry are hydrogen or deuterium.
In one embodiment, A is cyclopropyl (i.e. g is 1), t is 1, s is 1, X is hydroxy and the ens in the -
)- and -CH2-O groups are 2H (or D), and the compound of formula (I) is a compound of a
(Io) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
WO 55860
In one embodiment the compound of formula (I) is a compound of formula (lo’) or (lo”) or a tautomer
or a solvate or a pharmaceutically acceptable salt thereof:
('0”)
In one embodiment, R3 is -(CRXRy)q-X and sis 1, t is 0 and q is 1 or 2, and the compound of formula (I)
is a compound ofthe formula (lp):
In one embodiment, RX and Ry are H, and the compound of formula (Ip) is a compound ofthe formula
(lp’) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
In one embodiment, A is a C3_5cycloalkyl group or saturated heterocyclic group with 3 to 6 ring
members, wherein t is 1, and s is 1,Y is ndently selected from -CH2-, 0, or 802, i is 0 or 1, g is
1, 2, 3 or 4 and i + g is 1, 2, 3 or 4 and the compound of formula (I) is a compound of the a (lq)
or a tautomer or a solvate or a pharmaceutically acceptable salt f:
(oRXan-X
— (M)
In one embodiment the compound of formula (I) is a compound ofthe formula (lq’) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
(CRXRY)q-X
— (W)
In one embodiment of the compound of formula (Iq’), q is 1 and RX, Ry and X are hydrogen.
In one embodiment of the compound of formula (Iq’), q is 1, RX and Ry are hydrogen, and X is y.
In one embodiment of the compound of formula (Iq’), q is 1, RX and Ry are en, and X is fluorine.
In one embodiment of the compound of formula (Iq’), q is 0. In one embodiment of the compound of
formula (Iq’), q is Oand X is fluorine.
In one embodiment q is 0 and X is ne and the compound of formula (Iq’) is a compound of the
formula (Iq”) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
— (“J”)
In one embodiment of the compound of (Iq’) orthe compound of (Iq"), g is 1, i is 1 and Y is O.
In one embodiment g is 1, i is 1, Y is 0, q is 0 and X is F and the compound of formula (Iq’) is a
compound of the formula (Iq’”) or a tautomer or a e or a pharmaceutically acceptable salt
thereof:
— (lqm)
In one embodiment, i is 1 and Y is O or 802, in ular 0. In one embodiment, the compound of
formula (Iq) is a compound of formula (lq“”) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
— (Iq”").
In one embodiment, s is 0, t is 1, A is tetrahydofuranyl, q is 0 and X is en. In one embodiment,
R3 is tetrahydrofuranyl and s is 0.
In one embodiment, -(CH2)SR3 is selected from the following table (point of attachment to the oxygen
represented by dashed bond or bond terminus marked “*”):
i0H f: x”!
ICHS
.-‘\ /\OH
-CHZCH20H
'_-\i~H2 D
-CHZCHZCHZOH r0“
_.\/\N/so CH2 a
-CHzCH3
In one embodiment, -(CH2)SR3 is selected from the following table (point of attachment to the oxygen
represented by dashed bond or bond terminus marked “*”)2
-CH3 “
-CHzCHZOH _,.-\iNH2 \DAOH
" r0”
-CHzCHzCHzOH 0
D D D D
\/\OH
F/\F ,
‘__—
OH \A/\u/SO2CH3 “0’40!)
_-“—\/OH \/
A <> \\\—/O
“__v\/CN \/\OH \/\\\N
In one embodiment A is cyclopropyl, t is 1, s is 1, Rxand Ry are hydrogen and X is -OH.
In one embodiment A is ropyl, t is 1, s is 1, Rxand Ry are hydrogen and X is -CN.
In one embodiment R3 is hydrogen and s is 1. In one embodiment, X is hydrogen and s, t, and q are 0.
R4 and a
WO 55860
a is 0, 1, 2 or 3. In other words, the phenyl group of the isoindolinone may have 0, 1, 2 or 3
substituents R4.
In one embodiment a is 0 or 1. In another embodiment a is 0. In r embodiment a is 1.
When a is 2 or 3 (Le. the phenyl group ofthe isoindolinone is substituted with more than one R4) the
tuents R4 may be the same or different (i.e. are ndently selected from the definitions of
R4).
In one embodiment, a is 1 and the substituent R4 is at the 4-position of the isoindolinone, and the
compound of formula (I) is a compound of formula (Ir) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
R6 N
/ \ (R1)n
— (Ir)
R4 is ndently ed from halogen, nitrile, CM alkyl, haloCMalkyl, C1_4alkoxy and haloC1_
4alkoxy.
In one embodiment, R4 is halogen. In one embodiment, R4 is fluoro or chloro. In another
embodiment, R4 is fluoro.
In one embodiment, a is 1, the substituent R4 is at the 4-position of the isoindolinone, and R4 is F
and the compound of formula (I) is a compound of formula (ls) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof:
\(TL / \/<R5)m
F \
\\‘\\ R2
R6 N
/ X(R1
In one embodiment, a is 0, and the compound of formula (I) is a compound of formula (It) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
\(WL / \{mfom
\\\\
\\‘\\ R2
R6 N
/ \ (R1
In one embodiment, R4 is 01.4 alkyI (e.g. -CH3), or halogen (e.g. F or Cl) and a is 1.
In one embodiment, a is 0 and R4 is absent (i.e. hydrogen).
In one embodiment a is 0 or1 and R4 is halogen (e.g. fluorine).
R5 and m
m is 1 or 2. In other words, the phenyl group may have 1 or 2 substituents R5.
In one embodiment, m is 1 and the phenyl group has one substituent.
R5 may be attached at the ortho (or 0-), meta (or m—) or para (or p—) position of the phenyl group,
wherein the position is defined relative to the point of attachment ofthe phenyl group to the tion
of the isoindolinone ring.
When m is 2 (Le. the phenyl group is substituted with more than one R5) the substituents R5 may be
the same or ent (i.e. are independently selected from the definitions of R5).
In one embodiment, m is 1 and the substituent R4 is at the p-position of the phenyl group, and the
compound of formula (I) is a compound of formula (Iu) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
— (IU)
R5 is independently selected from halogen, nitriIe, CM alkyl, haloC1_4alkyl, C1_4alkoxy and haloC1_
4a|koxy.
In one ment, R5 is halogen, C1_4 alkyl, _4alkyl or C1_4alkoxy. In another embodiment R5 is
halogen (e.g. chloro).
In one ment, R5 is halogen (e.g. Cl or F), 01.4 alkyl (e.g. -CH2CH3), nitrile, haloC1_4alkyl (e.g. -
CF3, or -CF2CH3), or haloC1_4alkoxy (e.g. -OCF3), and m is 1 or 2.
In one ment, m is 1 and R5 is selected from halogen, nitrile, CM alkyl, haloC1.4alkyl, C1.4alkoxy
and haloC1_4alkoxy.
In one embodiment, m = 1 and R5 is -Cl (e.g. p-Cl), -F (e.g. 4-F), -CN (e.g. p-CN), -CF3 (e.g. p-CF3), -
OCF3 (e.g. p—OCFg), CF2CH3 (e.g. p-CF2CH3) or -CHZCH3 (e.g. p-CH2CH3), or m = 2 and R5 is p—F or
m-F.
In one ment, m = 1 and R5 is -Cl (e.g. p—Cl)
R6 and R7
R8 and R7 are independently selected from hydrogen, kyl, haloC1_5alkyl, 02.6alkenyl, Cg_5alkynyl,
hydroxy, hydroxyC1_6alkyl, -COOC1_6alkyl, -(CH2)j-O-C1_6alkyl, -(CH2)j-O-(hydroxyC1_6alkyl), lkyl-
NRXRV, y)p-CONRXR", y)p-NRXCORY, -(CRXRy)p-O-CH2-CONRXRY, heterocyclic group with
3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring members, -CH2-O-heterocyclic group
with 3 to 7 ring members, -CH2-NH-heterocyclic group with 3 to 7 ring s, -CH2-N(C1_6alkyl)-
heterocyclic group with 3 to 7 ring members, -C(=O)NH-heterocyc|ic group with 3 to 7 ring members,
03.3cycloalkyl, -CH2-Cs.3cycloalkyl, -CH2-O-C3.3cycloalkyl, and 03.3cycloalkenyl, wherein said
lkyl, cycloalkenyl or heterocyclic groups may be optionally substituted by one or more RZ
groups, and wherein in each instance the heterocyclic group comprises one or more (e.g.1, 2, or 3)
heteroatoms selected from N, O, S and oxidised forms thereof;
or the R6 and R7 groups, together with the carbon atom to which they are attached, can join to form a
Cg_scycloalkyl or heterocyclyl group with 3 to 6 ring members, wherein the heterocyclic group
comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and ed forms thereof,
and wherein said C3_6cycloalkyl and heterocyclyl groups may be optionally substituted by one or more
RZ groups;
RX and Ry are independently selected from hydrogen, halogen, nitro, nitrile, C1_6alkyl, _6alkyl, C2.
6alkenyl, 02.6alkynyl, hydroxy, hydroxyC1_6alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_6alkoxy,
-COOC1.sa|ky|, -N(H)e(C1.4a|ky|)2.e, -C1.aalkyl-N(H)e(C1.4a|kyl)2.e, '(CH2)k'C(=O)N(H)e(C1-4alkyl)2-e, C3-
gcycloalkyl and 03.8cycloalkenyl;
or the RX and Ry groups, er with the carbon or nitrogen atom to which they are attached, can
join to form a Cg_6cycloalkyl or saturated heterocyclyl group with 3 to 6 ring members which may be
optionally fused to an aromatic heterocyclyl group of 3 to 5 ring members;
or when on a carbon atom the RX and Ry groups can join together to form a =CH2 group;
RZ is independently selected from halogen, nitro, nitrile, C1,6alkyl, haloC1_6alkyl, enyl, C2_6alkynyl,
:0, hydroxy, hydroxyC1_6alkyl, C1_5alkoxy, -(CH2)k-O-C1_5alkyl, hydroxyC1_6alkoxy, -C(=O)C1_6alkyl, -
C(=O)C1_6alkyI-OH, -C(=O)C1.6alkyI-N(H)e(C1_4alkyI)2.e, -C(=O)N(H)e(C1_4alkyI)2_e, -(CH2),-002C1_6alkyl, -
-COZH, -N(H)e(C1_4alky|)2.e, -C1.6alkyl-N(H)e(C1_4alky|)2.e, heterocyclyl group with 3 to 6 ring
members, heterocyclyl group with 3 to 6 ring members substituted by C1_4alkyl, heterocyclyl
group with 3 to 6 ring members substituted by -C(=O)OC1_4alkyl, heterocyclyl group with 3 to 6 ring
members substituted by -C(=O)N(H)e(C1_4alkyl)2_e, -C(=O)heterocyclyl group with 3 to 6 ring s,
Cg_8cycloalkyl and 03.8cycloalkenyl, wherein if R7 is pyridine then RZ is other then -NH2;
j, e, r and p are independently selected from 0, 1 and 2; and
k is selected from 1 and 2.
In one embodiment, R6 and R7 are independently ed from hydrogen, C1_6alkyl, haloC1_6alkyl, C2.
6alkenyl, 02.6alkynyl, hydroxy, hydroxyC1_6alkyl, -COOC1_6alkyl, -(CH2)j-O-C1_6alkyl, -(CH2)j'O-
(hydroxyC1_5alkyl), -C1_6alkyI-NRXRy, -(CRXRy)p-CONRXRY, -(CRXRy)p-NRXCORV, -(CRXRy)p-O-CH2-
CONRXRV, heterocyclic group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring
members, -CH2-O-heterocyclic group with 3 to 7 ring s, -CH2-NH-heterocyclic group with 3 to
7 ring members, (C1.6alkyl)-heterocyclic group with 3 to 7 ring members, -C(=O)NH-
heterocyclic group with 3 to 7 ring members, C3_gcycloalkyl, -CH2-Cg_gcycloalkyl, -CH2-O-Cg_scycloalkyl,
and C3,.8cycloalkenyl, wherein said lkyl, cycloalkenyl or heterocyclic groups may be optionally
substituted by one or more RZ groups, and wherein in each ce the heterocyclic group comprises
one or more (e.g.1, 2, or 3) heteroatoms ed from N, O, S and oxidised forms thereof;
In one embodiment R7 is a cycloalkyl, cycloalkenyl or cyclic group optionally substituted by one
or more RZ selected from C1_6alkyl (e.g. methyl), C1_6alkoxy (e.g. methoxy) and -C(=O)C1_6alkyl (e.g. -
C(=O)CH3).
In one embodiment R7 is a cycloalkyl or cycloalkenyl group optionally substituted by one or more RZ
groups wherein RZ is hydroxy.
R6 and R7 may be the same or different.
When R6 and R7 are ent, the compound of formula (I) can exist as at least two diastereoisomers:
Diastereoisomer 2A
— DiastereoisomerZB
For the avoidance of doubt, the general formula (I) and all subformulae cover both individual
diastereoisomers and mixtures ofthe diastereoisomers which are related as epimers at the -CR6R7OH
group.
In one embodiment of the compound of formula (I) R6 and R7 are different and the compound is
diastereoisomer 2A or a tautomer or a solvate or a pharmaceutically acceptable salt thereof.
In one embodiment of the compound of formula (I) R6 and R7 are ent and the nd is
diastereoisomer ZB or a tautomer or a solvate or a pharmaceutically acceptable salt thereof.
In one embodiment, R6 is methyl and the compound of formula (I) is a compound of formula (Iv) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
\(W )8 / m
R6 \
\\\\
\“‘\ R2
R7 (Rbn
/ \ (Rt)n
— (Iv).
In one embodiment, R6 is ethyl and the compound of formula (I) is a compound of a (Iv’) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
In one embodiment, R7 is selected from C1_6alkyl or _6alkyl. In one embodiment R7 is a C3.
acycloalkyl (e.g. cyclopropyl, cyclobutyl or cyclohexyl) optionally substituted by one or more RZ groups
(e.g. -OH).
In one embodiment, R7 is selected from C1_6alkyl, yC1_6alkyl, -(CH2)J--O-C1_6alkyl, -(CH2)J--O-
(hydroxyC1_6a|ky|), -C1_e,a|kyI-NR"Ry (e.g. -C1.6alkyI-N(H)e(C1_4alkyl)2_e), -(CRXRy)p-NRXCORV,
cyclic group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring members, -CH2-
NH-heterocyclic group with 3 to 7 ring s, -CH2-N(C1.6alkyl)-heterocyclic group with 3 to 7 ring
members, -C(=O)NH-heterocyclic group with 3 to 7 ring members, C3_gcycloalkyl, and -CH2-C3_
acycloalkyl, wherein said cycloalkyl or heterocyclic groups may be optionally substituted by one or
more RZ groups, and n in each instance the heterocyclic group ses one or more (e.g. 1,
2, or 3) heteroatoms ed from N, O, S and oxidised forms thereof.
In one ment, R7 is selected from C1_6alkyl, hydroxyC1_6alkyl, -(CH2)J--O-C1_6alkyl, -(CH2),--O-
(hydroxyC1_6alkyl), -C1_6alkyI-N(H)e(C1_4alkyl)2_e, cyclic group with 3 to 7 ring members, -CH2-
heterocyclic group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring members,
cloalkyl, and -CH2-Cg_8cycloalkyl, wherein said cycloalkyl or heterocyclic groups may be
optionally substituted by one or more RZ groups, and wherein in each instance the heterocyclic group
comprises one or more (e.g. 1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms thereof.
In one embodiment, R7 is selected from heterocyclic group with 3 to 7 ring members,-CH2-heterocyclic
group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring members, Cg_8cycloalkyl,
and g_gcycloalkyl, wherein said cycloalkyl or heterocyclic groups may be optionally substituted
by one or more RZ groups, and wherein in each instance the heterocyclic group comprises one or
more (e.g. 1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms thereof.
In one ment, R7 is selected from heterocyclic group with 3 to 7 ring members and -CH2-
heterocyclic group with 3 to 7 ring members, wherein said heterocyclic groups may be optionally
substituted by one or more RZ groups, and wherein in each instance the heterocyclic group comprises
one or more (e.g. 1, or 2) heteroatoms selected from N, O, S and oxidised forms thereof.
In embodiment, the heterocyclic group is saturated. In one embodiment, R7 is saturated heterocyclic
group with 3 to 6 ring members or saturated heterocyclic group with 3 to 6 ring members) such
as n the heterocyclic group is selected from oxetanyl, oxanyl, piperidinyl, piperazinyl,
morpholinyl, pyrrolidinyl, imidazolinyl, azetidinyl, thiomorpolinyl, such as oxanyl, piperdinyl or
piperazinyl.
In one embodiment, R7 is selected from saturated heterocyclic group with 3 to 6 ring members and -
CHz-saturated heterocyclic group with 3 to 6 ring members, n said heterocyclic groups may be
optionally substituted by one or more RZ groups, and wherein in each instance the cyclic group
comprises one or more (e.g. 1, 2, or 3) heteroatoms selected from N, O, S. In one embodiment, R7 is
selected from a en containing saturated heterocyclic group with 3 to 6 ring member and -CH2-
(nitrogen containing saturated heterocyclic group with 3 to 6 ring members), wherein said heterocyclic
2016/053042
groups may be optionally tuted by one or more RZ groups, and wherein the heterocyclic group
may optionally contain one or more (e.g. 1, 2, or 3) additional heteroatoms selected from N, O, S.
In one embodiment, R7 is nitrogen containing saturated heterocyclic group with 3 to 7 ring members or
nitrogen containing saturated heterocyclic group with 3 to 7 ring members), n said
nitrogen containing saturated heterocyclic groups may be optionally substituted by one or more RZ
groups and wherein the nitrogen containing saturated heterocyclic group may optionally contain one or
more (e.g. 1, 2, or 3) additional heteroatoms selected from N, O, S. In one embodiment the nitrogen
containing saturated heterocyclic group with 3 to 7 ring members (such as 3 to 6 ring members) is
ed from piperidinyl, piperazinyl, morpholinyl, pyrrolidinyl, imidazolinyl, azetidinyl, thiomorpolinyl,
such as piperdinyl or piperazinyl.
In one embodiment, R7 is en containing aromatic heterocyclic group with 3 to 6 ring members or
-CH2-(nitrogen containing aromatic heterocyclic group with 3 to 6 ring members), wherein said
heterocyclic groups may be optionally substituted by one or more RZ groups and wherein the
heterocyclic group may optionally contain one or more (e.g. 1, 2, or 3) additional heteroatoms ed
from N, O, S.
In another embodiment, R7 is nitrogen containing aromatic heterocyclic group with 3 to 6 ring
members, n said heterocyclic group may be unsubsituted or substituted by one or more R2
groups, for e ed from halogen (e.g. fluorine), C1_6a|kyl (e.g. methyl), C1_5alkoxy (e.g.
y),and -C(=O)C1_6alkyl (e.g. -C(=O)CH3).
In one embodiment, R7 is oxygen containing aromatic heterocyclic group with 3 to 6 ring members or -
CHZ-(oxygen containing aromatic heterocyclic group with 3 to 6 ring members), wherein said
heterocyclic groups may be optionally substituted by one or more RZ groups and wherein the
heterocyclic group may optionally contain one or more (e.g. 1, 2, or 3) additional heteroatoms selected
from N, O, S.
In another embodiment, R7 is oxygen containing aromatic heterocyclic group with 3 to 6 ring members,
n said heterocyclic group may be unsubsituted or substituted by one or more RZ groups, for
e RZ groups selected from halogen (e.g. fluorine), C1_6alkyl (e.g. methyl), C1_6alkoxy (e.g.
methoxy),and -C(=O)C1_5alkyl (e.g. -C(=O)CH3).
In one ment R7 is selected from heterocyclyl groups containing 5 or 6 ring members optionally
substituted by one or more RZ.
In one embodiment R7 is ed from aromatic heterocyclyl groups containing 5 ring members
optionally tuted by one or more RZ. In one embodiment R7 is selected from an aromatic nitrogen
containing (e.g. diaza) rocyclyl group containing 5 ring members optionally substituted by one or
more RZ. In one embodiment R7 is pyrazolyl (e.g. pyrazolyl or pyrazolyl).
In one embodiment R7 is selected from a saturated rocyclyl group containing 6 ring members
optionally substituted by one or more RZ. In one embodiment R7 is selected from a saturated oxygen
or nitrogen containing heteterocyclyl group containing 6 ring members optionally substituted by one or
more RZ.
In one embodiment R7 is selected from oxanyl, piperidinyl, pyrazolyl or imidazolyl optionally
substituted by one or more RZ. In one embodiment R7 is selected from oxanyl, piperidinyl, lyl or
olyl optionally substituted by one or more RZ, where RZ is selected from halo (e.g. -F) or C1_4a|kyl
(e.g. methyl).
In one embodiment R7 is selected from oxanyl (also known as tetrahydropyranyl) or piperidinyl
optionally substituted by one or more RZ. In one embodiment R7 is selected from oxanyl or piperidinyl
unsubstituted or substituted by one or more RZ, where RZ is selected from halo (e.g. -F) or C1.4alkyl
(e.g. methyl), in particular halo (e.g. -F).
In one embodiment, R7 is 03.8cycloalkyl such as Cs_6cycloalkyl (e.g. cyclobutyl or cyclohexyl) ally
substituted by one or more RZ, for example where RZ is hydroxy. In one embodiment, R7 is cyclohexyl
optionally substituted by one or more hydroxy. In one embodiment R7 is cyclohexyl optionally
substituted by one or more hydroxyl, in the trans stereochemistry (e.g. 4-hydroxycyclohexane).
In one embodiment R7 is selected from H-heterocyclic group with 3 to 7 ring members (e.g. -
CHz-NH-oxanyl and -CH2-N(C1_6alky|)-heterocyclic group with 3 to 7 ring members (e.g. H3-
(piperidinyl) optionally substituted by one or more RZ groups (e.g. , -COCH3).
In one embodiment, R7 is y)p-CONRXRy or -C(=O)NH-heterocyclic group with 3 to 7 ring
members. In one embodiment, R7 is -C(=O)NH-heterocyclic group with 4 to 6 ring members (e.g.
piperidinyl, pyrazolyl, or azetidinyl).
In one embodiment, R7 is y)p-CONRxR-". In one embodiment R7 is -(CRXRy)p-CONH(C1.4alkyl), in
particular -(CO)NHCH3, -(CO)NHCH2CH3 or-(CO)NH(CH(CH3)2).
In one embodiment R7 is -C(=O)NH-heterocyclic group with 3 to 7 ring members (e.g. -C(=O)NH-
piperidinyl, -C(=O)NH-azetidinyl or -C(=O)NH-pyrazolyl) ally substituted by one or more R
groups (e.g. methyl, -COCH3).
In one embodiment, R7 is -C1_6alkyl-NRXRy (e.g. -C1_6alkyl-N(H)e(C1.4alkyl)2_e). In one embodiment R7 is
-CH2NH2, -CH2NHCH3, or -CH2N(CH3)2, In one embodiment R7 is -C1_e;alkyl-NR"Ry wherein RX is Cg.
acycloalkyl. In one embodiment R7 is -C1.2alkyl-NH-C3_6cycloalkyl (e.g. -CH2-NH-cyclopropy|).
In one ment, R7 is -C1_6alkyl-NRXRy wherein the RX and Ry groups, together with the nitrogen
atom to which they are attached, can join to form a C3,.6cycloalkyl or heterocyclyl group with 3 to 6 ring
members. In one embodiment, RX and Ry together form a ted cyclyl group with 3 to 6 ring
members e.g. piperazinyl.
In one embodiment R7 is lkyl-NRXRy, wherein the RX and Ry groups, together with the nitrogen
atom to which they are attached, join to form a C3_6cycloalkyl or saturated heterocyclyl group with 3 to
6 ring members which may be optionally fused to an aromatic heterocyclyl group of 3 to 5 ring
members. In one embodiment R7 is -C1_6a|kyl-NRXR-‘/, wherein the RX and Ry groups, together with the
nitrogen atom to which they are attached, join to form a ted heterocyclyl group with 3 to 6 ring
members which is fused to an aromatic heterocyclyl group of 3 to 5 ring members. RZ is independently
selected from n, nitro, e, C1_6alkyl, haloC1_6alkyl, C2,6alkenyl, C2_6alkynyl, =O, y,
hydroxyC1_5alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_6alkoxy, -C(=O)C1_6a|kyl, -C(=O)C1_6a|kyl-
OH, -C(=O)C1.6alkyI-N(H)e(C1_4alkyl)2.e, N(H)e(C1_4alkyI)2_e, -(CH2)r-COZC1_6alkyl, -(CH2)r-C02H, -
N(H)e(C1_4alkyl)2.e, lkyl-N(H)e(C1_4alkyl)2.e, heterocyclyl group with 3 to 6 ring members,
heterocyclyl group with 3 to 6 ring members substituted by -C(=O)C1_4alkyl, cyclyl group with 3
to 6 ring members substituted by -C(=O)OC1_4alkyl, heterocyclyl group with 3 to 6 ring members
substituted by N(H)e(C1_4alkyl)2.e, -C(=O)heterocyclyl group with 3 to 6 ring members, C3.
8cycloalkyl and Cg_8cycloalkenyl.
In one embodiment RZ is independently selected from halogen (e.g. fluorine), C1_6alkyl (e.g. methyl),
C1_5alkoxy (e.g. methoxy), and -C(=O)C1_5alkyl (e.g. -C(=O)CH3).
In one embodiment RZ is independently selected from C1_6alkyl (e.g. methyl), C1_6alkoxy (e.g.
methoxy),and -C(=O)C1_6alkyl (e.g. -C(=O)CH3).
In one embodiment, R7 is C1_6alkyl (e.g. methyl or ethyl), haloC1_6alkyl (e.g. trifluoromethyl), C2_6alkenyl
(e.g. Czalkenyl), hydroxyC1_6alkyI (e.g. -CHZOH, -CHZCH20H), -C1_6alkyI-NRXRy (e.g. -CH2NH2, -
CHZNHCHg, -CH2N(CH3)2, or -CH2-NH-cyclopropyl), -(CRXRy)p-CONRXRy (e.g. -(CO)NHCH3,
-(CO)NHCH2CH3, -(CO)NHCH2CH2NH2 or -(CO)NH(CH(CH3)2), -(CH2)j-O-C1_6a|kyl (e.g. H3Y
-CHZOCH2CH3 or -CHZOCD3), -(CRXRy)p-NRXCORy (e.g. -CH2NHCOCH3), y)p-O-CH2-CONRXRy
(e.g. -CH2-O-CHZCON(CH3)2), -(CH2)j-O-(hydroxyC1_6alkyl) (e.g. -CH2CHZOH, ), NH-
heterocyclic group with 3 to 7 ring members, C3_6cycloalkyl, heterocyclic group with 3 to 7 ring
members (e.g. oxanyl), or -CH2-heterocyclic group with 3 to 7 ring members wherein the cycloalkyl or
heterocyclic group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and
oxidised forms and may be optionally substituted by one or more RZ groups (for example ed from
C1_5alkyl (e.g. methyl), C1_6alkoxy (e.g. y) and -C(=O)C1_6alkyl (e.g. -C(=O)CH3)).ln one
embodiment, R6 is methyl or ethyl and R7 is C1_6alkyl (e.g. methyl), hydroxyC1_6alkyl, lkyl-NRXRV, -
(CRXRy)p-CONRXRV, -(CH2),--O-C1.salkyl, -(CRXRy)p-NRXCORV, -(CRXRy)p-O-CH2-CONRXRV, '(CH2)j'O-
(hydroxyC1_6alkyl), heterocyclic group with 3 to 7 ring members (e.g. oxanyl), or -CH2-heterocyclic
group with 3 to 7 ring members wherein the heterocyclic group comprises one or more (e.g.1, 2, or 3)
heteroatoms selected from N, O, S and oxidised forms and may be optionally substituted by one or
more RZ groups selected from C1_6alkyl (e.g. methyl), C1_6alkoxy (e.g. methoxy) and -C(=O)C1_6alkyl
(e.g. -C(=O)CH3).
In one embodiment, R6 is selected from hydrogen, C1_6alkyl (e.g. -CH3, -CHZCH3 or -CHZCH2CH3), C2.
6alkenyl (e.g. -CH=CH2) and _6alkyl (e.g. -CF3).
In one embodiment, R6 is selected from hydrogen or C1_6alkyl (e.g. -CH3 CHg).
In one embodiment, R7 is C1_6alkyl (e.g. -CH3 or -CHZCH3), hydroxyC1_6alkyl (e.g. ), -C1_6alkyl-
NR"Ry (e.g. -CH2N(CH3)2), -(CR"Ry)p-CONR"Ry (e.g. -C(=O)N(CH3)2 or -C(=O)NHCH3 or
), j'O-C1_6alkyl (e.g. -CH20CH3), 03.8cycloalkyl (e.g. cyclobutyl or exyl),
heterocyclic group with 3 to 7 ring members e.g.
(point of attachment represented by dashed bond):
or -CH2-heterocyclic group with 3 to 7 ring members e.g.
(point of attachment represented by dashed bond)
wherein when the moiety R7 comprises a heterocyclic or lkyl group, the heterocyclic group may
be ally tuted by one or more RZ groups selected from C1_6a|kyl (e.g. methyl), hydroxy,
halogen (e.g. fluoro), -C(=O)C1,6alkyl (e.g. C(CH3)3), -(CH2),-COZH (e.g. -CH2COOH or
CHZCHZCOOH or -(CH2),-COZC1_6alkyl (e.g. CHZCHZCOOCHg).
In one embodiment, R7 is C1_6alkyl (e.g. methyl or ethyl), haloC1_6alkyl (e.g. trifluoromethyl), C2_6alkenyl
(e.g. Czalkenyl), hydroxyC1_6alky| (e.g. -CHZOH, -CHZCHZOH), |kyl-NRXRy (e.g. -CH2NH2, -
CHZNHCH3_ -CH2N(CH3)2, or -CH2-NH-cyclopropyl), -(CR"Ry)p-CONRXRy (e.g. -(CO)NHCH3,
-(CO)NHCH2CH3, -(CO)NHCH2CH2NH2 or -(CO)NH(CH(CH3)2), -(CH2)j-O-C1_6a|ky| (e.g. -CHZOCH3,
-CH20CHZCH3 or -CHZOCD3), -(CRXRy)p-NRXCORy (e.g. -CH2NHCOCH3), -(CR"Ry)p-O-CH2-CONRXRy
(e.g. -CH2-O-CHZCON(CH3)2), -(CH2)j-O-(hydroxyC1_6alkyl) (e.g. -CH2-O-CH2CHZOH, ), NH-
heterocyclic group with 3 to 7 ring members, 03.6cycloalkyl, heterocyclic group with 3 to 7 ring
members (e.g. oxanyl), or -CH2-heterocyclic group with 3 to 7 ring members wherein the cycloalkyl or
heterocyclic group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and
oxidised forms and may be optionally substituted by one or more RZ groups (for example selected from
C1,6alkyl (e.g. methyl), koxy (e.g. methoxy) and C1_6alkyl (e.g. -C(=O)CH3)).ln one
embodiment, R6 is methyl or ethyl and R7 is C1_6alky| (e.g. methyl), hydroxyC1_6alkyl, -C1_6alkyl-NRXR-‘/, -
(CRXRy)p-CONRXRV, -(CH2),--O-C1.6a|kyl, -(CRXRy)p-NRXCORV, -(CRXRy)p-O-CH2-CONRXRY, '(CH2)j'O-
(hydroxyC1_6alkyl), heterocyclic group with 3 to 7 ring members (e.g. oxanyl), or -CH2-heterocyclic
group with 3 to 7 ring members wherein the cyclic group comprises one or more (e.g.1, 2, or 3)
heteroatoms selected from N, O, S and oxidised forms and may be optionally substituted by one or
more RZ groups selected from C1_6alkyl (e.g. methyl), C1_6alkoxy (e.g. methoxy) and -C(=O)C1_5alkyl
(e.g. -C(=O)CH3).
In one embodiment, R6 is selected from hydrogen or C1.5a|kyl (e.g. -CH3 or-CHZCHs).
In one embodiment, R7 is C1_6alkyl (e.g. -CH3 or -CH2CH3), hydroxyC1_5alkyl (e.g. -CHZOH 0), -C1_
salkyl-NRXRy (e.g. -CH2N(CH3)2), -(CRXRy)p-CONRXRy (e.g. N(CH3)2 or ), -
(CH2)j'O-C1_6alkyl (e.g. -CHZOCH3), heterocyclic group with 3 to 7 ring members e.g.
(point of attachment represented by dashed bond):
or -CH2-heterocyclic group with 3 to 7 ring s e.g.
(point of attachment ented by dashed bond)
wherein when the moiety R7 comprises a cyclic group, the heterocyclic group may be ally
substituted by one or more RZ groups selected from C1.6a|kyl (e.g. methyl).
In one embodiment of formula (I) R7 is a heterocyclic group with 3 to 7 ring members optionally
substituted by one or more RZ groups e.g.
(point of attachment represented by dashed bond)
/ N/
In one embodiment of formula (I) R7 is a heterocyclic group with 3 to 7 ring members optionally
substituted by one or more RZ groups e.g.
(point of attachment represented by dashed bond)
_..”©~ [::::::TCH3
In one embodiment, R7 is a -CH2-heterocyclic group with 3 to 7 ring members optionally substituted by
by one or more RZ groups e.g.
(point of ment represented by dashed bond)
”ONH
In one embodiment, R7 is selected from:
(point of attachment represented by dashed bond):
”©~ [:::::::TCH3
O NCH3
In one embodiment, R7 is selected from:
(point of attachment represented by dashed bond):
73‘ :TCHS
In one embodiment, R6 is hydrogen or C1_6alkyl. In one embodiment, R6 is C1_6alkyl. In one
embodiment, R6 is methyl or ethyl. In one embodiment, R6 is ethyl.
In one embodiment, R6 is C1_6alkyl (such as methyl or ethyl e.g. methyl) and R7 is selected from
hydroxyC1_6alkyl and -(CH2)-O-C1_6alkyl, In one ment, R6 is methyl and R7 is ed from
methyl, -CH2-OH and -CH2-OCH3. In one embodiment R6 is methyl and R7 is methyl, ethyl, or propyl.
In one embodiment R6 is methyl and R7 is methyl.
In one embodiment, R6 is C1.6alkyl or haloC1.6alkyl (e.g. methyl, —monofluoromethyl, trifluoromethyl or
ethyl).
In one embodiment, R6 is 03.8cycloalkyl such as Cmcycloalkyl (e.g. cyclopropyl).
In one embodiment R6 is C1_6alkyl (such as methyl or ethyl e.g. ethyl) and R7 is selected from:
(point of attachment represented by dashed bond or bond terminus marked “*”):
-cHs ”U /
\ \
"CL Q /2\
N,/ /
o W,
t; O \
D °
-CHzCH3
In one embodiment R6 is C1_6alky| (such as methyl or ethyl e.g. ethyl) and R7 is selected from:
(point of attachment represented by dashed bond or bond us marked “*”):
-CH3 IN
,.—"\HEN> ,.—\
D )3 /N
WO 55860
NH N/
L \ I
/ NCH3
In particular, R7 is:
(point of attachment represented by dashed bond):
Om.“
In one embodiment, R6 is C1_6alkyl (such as methyl or ethyl e.g. ) and R7 is oxanyl, and the
compound of formula (I) is a compound of a (lw):
In one embodiment of formula (lw) RZ is hydrogen or fluorine.
In one embodiment, R7 is imidazolyl and the compound of formula (I) is a compound of formula (Ix) or
a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
— (IX)
In one embodiment, R7 is N-methyl piperidinyl and the nd of formula (I) is a compound of
formula (lx’) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
\qL / \/<R5)m R4 \
\ \
\\ 2
(R1)n
/ /\
R6 OH O
— (IX’)
In one embodiment, R7 is 4-quoromethy|piperidinyl and the compound of formula (I) is a
compound of a (Ix”) or a tautomer or a solvate or a pharmaceutically able salt thereof:
(R5)m
R4 \W )5
In one embodiment, R7 is pyrazolyl optionally substituted by one or more RZ groups (e.g. methyl). In
one embodiment, R7 is N-methylpyrazoIyl or N-methylpyrazoIyl.
In one embodiment, R7 is selected from methyl, oxanyl, pyrazolyl, imidazolyl, piperidinyl, and
cyclohexyl wherein said cycloalkyl and heterocyclic groups are optionally substituted by one or more
RZ groups (e.g. methyl, fluorine, or hydroxyl).
In one embodiment, R7 is ed from piperidinyl optionally substituted by one or more RZ groups
(e.g. methyl, fluorine, or hydroxyl, in particular methyl and fluorine).
In one embodiment, the compound of formula (I) is a compound of formula (Ix) and R6 is C1_4alkyl.
In one embodiment, R6 is C1_6a|kyl (e.g. -CH3, 3 or -CHZCH2CH3 such as methyl or ethyl e.g.
ethyl) and R7 is a heterocyclic group with 3 to 7 ring members optionally substituted by one or more R
groups.
In one embodiment, R6 is C1_6alkyl (e.g. -CH3, -CH2CH3 or -CH2CH2CH3 such as methyl or ethyl e.g.
ethyl) and R7 is imidazolyl optionally substituted by one or more RZ groups (e.g. methyl olyl).
In one embodiment, R6 is C1_6alky| (e.g. -CH3, -CH2CH3 or 2CH3 such as methyl or ethyl e.g.
ethyl) and R7 is dinyl optionally substituted by one or more RZ groups (e.g. methyl piperidinyl).
In one ment R6 is C1_6alky| (e.g. -CH3, -CHZCH3 or -CHZCH2CH3 such as methyl or ethyl e.g.
ethyl) and R7 is C1_4alkyl, hydroxy|C1_4alky|, yC1_4alkyl, a cyclic group with 5 or 6 ring
members or C3_5cycloalkyl, wherein the heterocyclic group or Cg_scycloalky| group is optionally
substituted by one or more RZ (e.g. methyl, halogen (such as fluorine), C(=O)Me, or -OH).
In one embodiment R6 is C1_6alkyl (e.g. -CH3, -CHZCH3 or -CHZCHZCH3 such as methyl or ethyl e.g.
ethyl) and R7 is methyl, ethyl, hydroxylmethyl, hydroxyethyl, methoxymethyl, piperidinyl, oxanyl,
imidazolyl, pyrazolyl, cyclobutyl, cyclohexyl, ally substituted by one or more RZ (e.g. methyl,
halogen (such as fluorine), e, or -OH).
In one embodiment, R6 and R7 are both the same. In one embodiment, R6 and R7 are both methyl,
and the compound of formula (I) is a compound of formula (Iy) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof:
In one embodiment the group -CR5R7OH is other than -C(CH3)ZOH.
In one embodiment, R7 is selected from the group ting of:
(point of attachment represented by dashed bond)
.CH3 D” Hikmwz
-CHZOH ]:/\N\ X—\NfiK/
-CHZOCH3 OW \O\
-CH2N(CH3)2 O KKNQ
[\\ 3
NHCH 3
NH NCH3
F F
.v-“‘“\\ {x x"
OW+ ,,,,.©fif°” 0AA
In one embodiment, R7 is selected from the group consisting of:
(point of attachment represented by dashed bond)
-CH20H xj:/\N\ O”
WO 55860
H3 0% \O\
-CH2N(CH3)2 ’0 .-“\N0
NHCH3
In one embodiment RZ is independently selected from halogen, nitro, nitrile, C1_6alkyl, haloC1_6alkyl, C2.
salkenyl, Czaalkynyl, :0, hydroxy, hydroxyC1_5alkyl, C1.6alkoxy, -(CH2)k-O-C1.5alkyl, hydroxyC1.6alkoxy,
-C(=O)C1_6a|kyl, -C(=O)C1_6alkyl-OH, -C(=O)C1.6alkyl-N(H)e(C1.4a|ky|)2.e, -C(=O)N(H)e(C1_4alkyI)2_e,
k-COZC1_6alkyl, -(CH2)r-COZH, -NH(C1_4alkyl), -N(C1_4alky|)2, -C1.6alkyl-N(H)e(C1.4alkyl)2_e,
heterocyclyl group with 3 to 6 ring s, heterocyclyl group with 3 to 6 ring s substituted
by -C(=O)C1.4alkyl, heterocyclyl group with 3 to 6 ring s substituted by -C(=O)OC1.4alkyl,
heterocyclyl group with 3 to 6 ring members substituted by -C(=O)N(H)e(C1_4alky|)2_e,
-C(=O)heterocycly| group with 3 to 6 ring members, 03.8cycloalkyl and C3.8cycloalkenyl.
In another embodiment RZ is independently selected from halogen, nitro, e, C1_6alkyl, haloC1_6a|kyl,
Czsalkenyl, Czealkynyl, =O, hydroxy, hydroxyC1_6alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_
6alkoxy, -C(=O)C1_6alkyl, C1_6alkyI-OH, -C(=O)C1_6alkyl-N(H)e(C1_4alkyl)2.e, N(H)e(C1_
4alkyl)2_e, -(CH2)r-Cozc1_6alkyl, -(CH2)r-COZH, -C1,6alkyI-N(H)e(C1_4alkyI)2_e, heterocyclyl group with 3 to
6 ring members, heterocyclyl group with 3 to 6 ring members substituted by -C(=O)C1_4a|kyl,
heterocyclyl group with 3 to 6 ring members substituted by -C(=O)OC1_4alkyl, heterocyclyl group with 3
to 6 ring members substituted by -C(=O)N(H)e(C1_4alkyI)2.e, -C(=O)heterocyclyl group with 3 to 6 ring
members, 03.8cycloalkyl and Cs_8cycloalkenyl.
In another embodiment when R7 contains a saturated hetereocyclic group then RZ is independently
selected from halogen, nitro, nitrile, C1_6alkyl, haloC1_6alkyl, kenyl, C2_6alkynyl, =O, hydroxy,
hydroxyC1_5alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_6alkoxy, -C(=O)C1_6a|kyl, -C(=O)C1_6a|kyl-
OH, -C(=O)C1.6alkyI-N(H)e(C1_4alkyl)2.e, -C(=O)N(H)e(C1_4alkyI)2_e, r-COZC1_6alky|, -(CH2)r-C02H, -
C1_4alky|)2.e, -C1.6alkyl-N(H)e(C1_4alkyl)2.e, heterocyclyl group with 3 to 6 ring members,
heterocyclyl group with 3 to 6 ring s substituted by -C(=O)C1_4alkyl, heterocyclyl group with 3
to 6 ring members substituted by -C(=O)OC1_4alkyl, heterocyclyl group with 3 to 6 ring members
substituted by N(H)e(C1_4alkyl)2.e, -C(=O)heterocyclyl group with 3 to 6 ring members, C3.
8cycloalkyl and Cg_8cycloalkenyl.
Subformulae
In one embodiment, the compound of formulae (I) is a compound of formulae (II) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
R1 (II)
wherein R1, R2, R3, R4, R5, R6, R7, a, m and s are as defined herein.
In one embodiment, R1 is chloro, e, methyl or methoxy. In one embodiment, R1 is hydroxy or
hydroxyC1_4alkyl (e.g. hydroxyl).
In one embodiment, R1 is OOJ1(CRXRY)VCOOH (e.g. -COOH, -CHZCOOH, -OCHZCOOH or -
C(CH3)2COOH.
In another embodiment, R1 is chloro or nitrile and the compound of formula (II) is a compound of
a (Ila) or (Ilb) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
CI (Ila)
N—: R1
0N (Ilb)
WO 55860
wherein R2, R3, R4, R5, R7, m and s are as defined herein. In one embodiment, R1 is -SOz-RX. In
ular, RX is -SOz-C1_4alkyl, for example -SOz-CH3 or -SOz-heterocyclic group with 5 to 6 ring
s (e.g. -SOz-morpholinyl, typically -SOz-(1-morpholinyl). In another embodiment In one
embodiment, R1 is hydroxy or hydroxyC1_4alkyl (e.g. -CH20H or —OH).
In one embodiment, R6 is methyl or ethyl, and the compound of formula (I) is a compound of formula
(Illa) or (lllb) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
wherein R1, R2, R3, R4, R5, R7, a, m and s are as defined herein.
In one embodiment, a is 1 and the compound of formula (I) is a compound of a (lVa) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
wherein R1, R2, R3, R4, R5, R7, a, m and s are as defined herein.
In one embodiment, s is 0 and the compound of formula (I) is a compound of formula (IVb) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
wherein R1, R2, R3, R4, R5, R7, a, m and s are as defined herein.
In one embodiment, R4 is F and the compound of a (IVa) is a compound of formula (V) or a
tautomer or a solvate or a pharmaceutically acceptable salt thereof:
s \\/<R5)m
R6 N
(R )n
— (V)
wherein R1, R2, R3, R5, R7, m and s are as defined herein.
In one embodiment, m is 1 and the substituent R4 is at the 4-position of the phenyl group, and the
compound of formula (I) is a compound of a (VI) or a tautomer or a e or a
pharmaceutically acceptable salt thereof:
In one embodiment, R5 is chloro and the compound of a (VI) is a compound of formula (Vla) or a
tautomer or a solvate or a ceutically acceptable salt thereof:
In one embodiment, A is a C3,.6cycloalkyl group (g is 1, 2 or 3) and t is 1, and the compound of formula
(VI) is a nd of formula (VII) or a tautomer or a solvate or a pharmaceutically acceptable salt
thereof:
In one embodiment, A is a 03.6cycloalkyl group (g is 1, 2 or 3) and t is 1, and the lkyl group is
geminally disubstituted (i.e. the group -(CRny)-X and the CH2 group (where s is 1) orthe oxygen atom
(where s is 0) are both attached to the same atom of the cycloalkyl group, and the compound of
formula (VII) is a compound of formula (Vlla) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
m-X R5
l6? \0*\/(R1)n
(Vlla)
In one embodiment, g is 1, and so the cycloalkyl group is a cyclopropyl group and the nd of
formula (Vlla) is a compound of formula (Vllb) or a tautomer or a solvate or a pharmaceutically
acceptable salt thereof:
R6 N
/ \ (R1)n
— (Vllb).
In one embodiment, s is 1, and the compound of formula (Vllb) is a compound offormula (Vllc) or a
tautomer or a e or a pharmaceutically acceptable salt thereof:
f: (-XCRXRy)q R5
— (Vllc).
In one embodiment, RX and Ry are hydrogen (including 1H and 2H) and q is 1 and the compound of
formula (Vllc) is a compound of (Vlld) or a er or a solvate or a pharmaceutically acceptable salt
thereof:
\\\‘
(Vlld).
In one embodiment, the compound of formula (Vlld) is a compound of (Vlld’) or a tautomer or a
solvate or a pharmaceutically acceptable salt f:
— (Vlld‘)
In one embodiment, the compound of formula (Vlld) is a compound of (Vlld’) and X is hydroxy.
In one embodiment, X is hydroxy, and the compound of a (Vlld) is a compound ofthe formula
(Vlle) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
In one embodiment, X is NH2 and the compound of formula (Vlle) is a compound ofthe a
(Vlle’) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
C(=O)NH2
€40 6
wherein q is 0 or 1, and in particularq is 0.
In one embodiment, X is -CN and the compound of formula (Vlld) is a compound ofthe a (Vlle”)
or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
KA V ,_O 30 01
R6 Zx’o:Z
(R1)n
(V||e”)
n q is 0 or 1, and in particularq is 0.
In one embodiment, R3 is methyl, and the compound of formula (VI) is a compound of formula (Vllf) or
a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
2016/053042
In one embodiment of Formula (Vlla-e’) R6 is methyl. In one embodiment of Formula (Vlla-e’) R6 is
ethyl.
In one embodiment of Formula (Vlle”) or (Vllf) R6 is methyl. In one embodiment of Formula (V||e") or
(Vllf)R6 is ethyl.
In one embodiment of Formula (Vlle”) or (Vllf)R5 is methyl. In one embodiment of Formula ) or
(Vllf) R6 is ethyl.
In one embodiment ofthe compound of formula (Vlla-e’), R7 is selected from , oxanyl, pyrazolyl,
imidazolyl, piperidinyl, and cyclohexyl wherein said cycloalkyl and heterocyclic groups are optionally
substituted by one or more RZ groups (e.g. methyl, e, or hydroxy).
In one ment of the compound of formula (Vlla-e’), R7 is selected from oxanyl and methyl.
In one embodiment of the compound of formula (Vlle”) or , R7 is selected from methyl, ,
pyrazolyl, imidazolyl, piperidinyl, and cyclohexyl wherein said cycloalkyl and heterocyclic groups are
optionally substituted by one or more RZ groups (e.g. methyl, fluorine, or hydroxy).
In one embodiment of the compound of formula (Vlle”) or , R7 is selected from oxanyl and methyl.
In one ment of the compound of formula (Vlla-f), R7 is selected from piperidinyl optionally
substituted by one or more RZ groups (e.g. methyl, fluorine, or hydroxy).
In another embodiment, the compound of formula (I) is a compound of formula (a) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
wherein R1 is chloro or nitrile, when s is 1 then X is hydroxyl or when s is 0 then X is -C(=O)NH2.
In another embodiment, the compound of formula (I) is a nd of formula (a’) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
/ \ (R1)n
wherein R1 is chloro or nitrile, when s is 1 then X is yl or when sis 0 then X is -CN.
In another embodiment, the compound of formula (I) is a compound of formula (a”) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
(R1)n
wherein R1 is chloro or e, when sis 1 then X is hydroxyl or when s is 0 then X is -C(=O)NH2.
In another ment, the compound of formula (I) is a compound of formula (a’) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
(R)n
WO 55860
wherein R1 is chloro or nitrile, when s is 1 then X is hydroxyl or when s is 0 then X is -CN.
In one embodiment of the compound of a (a), R7 is selected from methyl, oxanyl, pyrazolyl,
imidazolyl, piperidinyl, and cyclohexyl n said cycloalkyl and heterocyclic groups are optionally
substituted by one or more RZ groups (e.g. methyl, fluorine, or hydroxy).
In one embodiment of the nd of formula (a), R7 is oxanyl or methyl.
In one embodiment of the compound of formula (a), R7 is dinyl, optionally substituted with C1_6
alkyl (e.g. methyl) and/or halo (e.g. flouro).
In one ment of the compound of formula (a‘), (a”) or (a”’) R7 is selected from methyl, oxanyl,
pyrazolyl, imidazolyl, piperidinyl, and cyclohexyl wherein said cycloalkyl and heterocyclic groups are
optionally substituted by one or more RZ groups (e.g. methyl, fluorine, or hydroxy).
In one embodiment of the compound of formula (a’), (a”) or (a”’) R7 is oxanyl or methyl.
In one embodiment of the compound of formula (a’), (a”) or (a”’) R7 is piperidinyl, optionally tuted
with C1_6 alkyl (e.g. methyl) and/or halo (e.g. flouro).
In one embodiment, A is a heterocyclyl group with 3 to 6 ring members, wherein the heterocyclic group
comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms thereof
(t is 1; g is 1, 2, 3 or 4; Z represents N, O, S and oxidised forms thereof; i is 1, 2, or 3; and i + g = 2, 3,
4 or 5), and the compound of formula (VI) is a compound of formula (b) or a tautomer or a solvate or a
pharmaceutically acceptable salt f:
Yy(CR R )q xx v -
R6 N
R1 (b)-
In one ment, Y is O and i is 1 and the compound of formula (b) is a compound of formula (ba)
or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
O—| (CRXRy)q-X
/ R5
R1 (ba).
In one embodiment, s is 0, g is 2, q is 0 and X is hydrogen, and the compound of formula (b) is a
compound of formula (bb) or a tautomer or a e or a pharmaceutically acceptable salt thereof:
R1 (bb).
In one embodiment, s is 0, g is 1, Y is O and i is 1 and the compound of formula (b) is a compound of
formula (bc) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof:
R1 (bc).
In one embodiment, the nd of formula (bc) is where q is 0 and X is fluorine.
In another embodiment, the compound of formula (I) is a compound of formula (c) or a tautomer or a
solvate or a pharmaceutically acceptable salt f:
R1 (C)
n R1 is chloro or nitrile, s is 1 and X is yl or sis 0 and X is -C(=O)NH2.
In another embodiment, the compound of formula (I) is a compound of formula (c’) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
3 CI
R1 (C!)
wherein R1 is chloro or nitrile, s is 1 and X is hydroxyl or s is 0 and X is -CN.
In another embodiment, the compound of formula (I) is a nd of formula (0”) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
R1 (6”)
wherein R1 is chloro or nitrile, s is 1 and X is hydroxyl or sis 0 and X is -C(=O)NH2.
In another embodiment, the compound of formula (I) is a compound of a (c ) or a tautomer or a
solvate or a pharmaceutically acceptable salt thereof:
R1 (c"’>
wherein R1 is chloro or nitrile, s is 1 and X is hydroxyl or s is 0 and X is -CN.
In one embodiment of the compound of formula (c), R7 is selected from methyl, oxanyl, pyrazolyl,
imidazolyl, dinyl, and cyclohexyl wherein said cycloalkyl and heterocyclic groups are optionally
substituted by one or more RZ groups (e.g. methyl, fluorine, or hydroxy).
In one embodiment of the nd of formula (c), R7 is oxanyl or methyl.
In one embodiment of the compound of a (c), R7 is dinyl, optionally substituted with C1_6
alkyl (e.g. methyl) and/or halo (e.g. flouro).
In one embodiment of the compound of formula (0’), (c”) or (c ) R7 is ed from methyl, oxanyl,
pyrazolyl, imidazolyl, piperidinyl, and cyclohexyl wherein said cycloalkyl and heterocyclic groups are
optionally substituted by one or more RZ groups (e.g. methyl, fluorine, or hydroxy).
In one embodiment of the compound of a (c’), (c”) or (c ) R7 is oxanyl or methyl.
In one embodiment of the compound of formula (0’), (c”) or (c ) R7 is dinyl, optionally substituted
with C1_6 alkyl (e.g. methyl) and/or halo (e.g. .
In another embodiment ofthe subsformulae described hereinabove, R2 is selected from hydrogen and
-(RXRy)u-002H (e.g. -COOH, -CHZCOOH, -CHZCH2-COZH, -(CH(CH3))-COZH and -(C(CH3)2-COZH).
In another embodiment of the subsformulae described hereinabove, R2 is selected from -(CH(CH3))-
COZH and -(C(CH3)2-COZH).
In another ment, R2 is ed from H3))-COZH and -(C(CH3)2-COZH) (e.g. /\C°2H,
Amor -(C(CH3)2-COZH.
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof wherein:
R1 is independently selected from hydroxy, halogen, nitro, nitrile, kyl, haloC1_4alkyl, hydroxyC1_
4alkyl, C2_6alkenyl, C1_4alkoxy, haloC1_4alkoxy, and C2_4alkynyl;
R2 is selected from hydrogen, CM alkyl, Czealkenyl, hydroxyCMalkyl and -CH2C02H;
R3 is hydrogen or (CRXRy)q-X;
s and t are independently selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then s is 1 and q is otherthan 0;
A is a C3_5cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group ses one or more , 2, or 3) heteroatoms ed from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -C(=O)—C1_
4alkyl, -NRXRY, -NHSOZRX, -NRXCORY; and -C(=O)NRXRV;
R4 and R5 are independently selected from n, nitrile, CM alkyl, haloC1_4a|kyl, C1_4alkoxy and
haloC1_4alkoxy;
R6 and R7 are independently selected from hydrogen, C1_5alkyl, _5alkyl, C2_5alkenyl, C2_6alkynyl,
hydroxy, hydroxyC1_6alkyl, -COOC1_6alkyl, heterocyclic group with 3 to 7 ring s, -CH2-
heterocyclic group with 3 to 7 ring members, -CH2-O-heterocyclic group with 3 to 7 ring members, -
CHz-NH-heterocyclic group with 3 to 7 ring members, -CH2-N(C1_6alkyI)-heterocyclic group with 3 to 7
ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring members, Cg_8cycloalkyl, -CH2-Cg_
8cycloalkyl, -CH2-O-C3_8cycloalkyl, and Cg_8cycloalkenyl, wherein said cycloalkyl, cycloalkenyl or
heterocyclic groups may be optionally substituted by one or more RZ groups, and wherein in each
instance the heterocyclic group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N,
O, S and oxidised forms thereof;
R9 is selected from hydrogen, C1_6alkyl, _6alkyl, hydroxyC1_6alkyl, -(CH2)k-O-C1_6alkyl, -(CH2)k-O-
(hydroxyC1_6a|ky|), hydroxyC1_6a|koxy, -(CH2)k-COZC1_6a|kyl, -(CH2)k-COZH, -C1_5a|ky|-N(H)e(C1_4a|kyl)2_
e, -(CH2)j-C3_8cycloalkyl and -(CH2)j-C3.gcycloalkenyl;
Rx and Ry are independently selected from en, halogen, nitro, nitrile, C1_5alkyl, haloC1_6alkyl, CZ.
6alkenyl, C2_6alkynyl, hydroxy, hydroxyC1_6alkyl, koxy, -(CH2)k-O-C1_6alkyl, yC1_6alkoxy,
-COOC1.ealkyl. -N(H)e(C1.4a|ky|)2»e, -C1.ealkyI-N(H)e(C1.4alky|)2.e, '(CH2)k'C(=O)N(H)e(C1»4a|kyl)2»e C3-
gcycloalkyl and Cg_8cycloalkenyl;
RZ is independently selected from n, nitro, nitrile, C1,6alkyl, haloC1_6alkyl, Czfialkenyl, C2_6alkynyl,
:0, hydroxy, hydroxyC1_6alkyl, C1_5alkoxy, -(CH2)k-O-C1_5alkyl, hydroxyC1_6alkoxy, -C(=O)C1_6alkyl, -
C(=O)C1_6alkyI-OH, -C(=O)C1.6alkyl-N(H)e(C1_4alkyI)2.e, -C(=O)N(H)e(C1_4alkyl)2_e, -(CH2)r-002C1_6alkyl, -
(CH2),-COZH, -N(H)e(C1_4a|ky|)2.e, -C1.6alkyl-N(H)e(C1_4alky|)2.e, cyclyl group with 3 to 6 ring
members, heterocyclyl group with 3 to 6 ring members substituted by -C(=O)C1_4alkyl, heterocyclyl
group with 3 to 6 ring s substituted by -C(=O)OC1_4alkyl, heterocyclyl group with 3 to 6 ring
members substituted by -C(=O)N(H)e(C1_4alkyl)2.e, -C(=O)heterocycly| group with 3 to 6 ring s,
Cg_8cycloalkyl and Cg_8cycloalkenyl;
n, e, r and j are independently selected from 0, 1 and 2;
k and m are ndently selected from 1 and 2; and
v and a are independently selected from 0 and 1.
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof, wherein:
R1 is ndently selected from hydroxy, halogen, nitro, nitrile and C1_4alkyl;
R2 is selected from hydrogen, C1_4 alkyl, Czealkenyl, hydroxyC1_4alkyl and —CH2C02H;
R3 is hydrogen or (CR"Ry)q-X;
s and t are independently ed from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then sis 1 and q is han 0;
A is a C3.5cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic
group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms
thereof;
X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)V-COZH, -(CH2)V-COZC1_4alkyl, -C(=O)—C1_
, -NR"Ry, -NHSOZRX, -NRXCORV; and -C(=O)NRXRV;
R4 and R5 are independently ed from halogen, nitrile and C14 alkyl;
R6 is selected from hydrogen, C1_6alkyl, haloC1_6alkyl, C2_6alkenyl, and C2_6alkynyl;
R7 is selected from hydrogen, C1_6alkyl, haloC1,6alkyl, Cz_6alkenyl, Czealkynyl, y, hydroxyC1_
6alkyl, -COOC1_6alkyl, heterocyclic group with 3 to 7 ring members,
-CH2-heterocyclic group with 3 to 7 ring members, -CH2-O-heterocyclic group with 3 to 7 ring
s, -CH2-NH-heterocyclic group with 3 to 7 ring members, -CH2-N(C1_6alkyl)-heterocyclic group
with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring members, C3_8cycloalkyl, -CH2-
Cg_8cycloalkyl, -CH2-O-C3_8cycloalkyl, and C3_gcycloalkenyl, wherein said cycloalkyl, cycloalkenyl or
heterocyclic groups may be optionally substituted by one or more RZ groups, and wherein in each
instance the heterocyclic group ses one or more (e.g.1, 2, or 3) heteroatoms selected from N,
O, S and oxidised forms thereof;
R9 is selected from hydrogen, C1_aa|kyl, haloC1_6alkyl, hydroxyC1_ea|kyl, -(CH2)k-O-C1_6alkyl, -(CH2)k-O-
(hydroxyC1_6alkyI), yC1_6a|koxy, -(CH2)k-COZC1_6alkyl, k-COZH, -C1_6 alkyl-N(H)e(C1_4alkyI)2_
e, j-Cg_8cycloalkyl and -(CH2)j-Cg.gcycloalkenyl;
RX and Ry are independently selected from hydrogen, halogen, nitro, nitrile, C1_6alkyl, haloC1_6alkyl, C2.
6alkenyl, Cz_6alkynyl, hydroxy, hydroxyC1_6alkyl, C1_6alkoxy, -(CH2)k-O-C1_6alkyl, hydroxyC1_6alkoxy,
.6alkyl, -N(H)e(C1.4alky|)2.e, -C1.6a|ky|-N(H)e(C1.4a|ky|)2.e, '(CH2)k'C(=O)N(H)e(C1-4a|kyl)2-e, C3-
gcycloalkyl and C3_8cycloalkenyl;
RZ is independently ed from n, nitro, nitrile, C1_6alkyl, haloC1_6alkyl, C2_6alkenyl, C2_6alkynyl,
=O, hydroxy, hydroxyC1_5alky|, C1_6a|koxy, k-O-C1_salkyl, hydroxyC1_5alkoxy, -C(=O)C1.6alky|, -
C(=O)C1_6alkyl-OH, -C(=O)C1.6alkyI-N(H)e(C1_4alkyI)2.e, -C(=O)N(H)e(C1.4alkyl)2_e, -(CH2),-COZC1_6alkyl, -
(CH2),-C02H, -N(H)e(C1_4alky|)2.e, -C1.salkyl-N(H)e(C1.4alky|)2.e, heterocyclyl group with 3 to 6 ring
members, heterocyclyl group with 3 to 6 ring members substituted by -C(=O)C1_4alkyl, heterocyclyl
group with 3 to 6 ring members substituted by -C(=O)OC1.4a|kyl, heterocyclyl group with 3 to 6 ring
members substituted by -C(=O)N(H)e(C1_4alkyI)2.e, -C(=O)heterocyclyl group with 3 to 6 ring members,
C3_8cycloalky| and C3.8cycloa|kenyl;
n, e, r and j are independently selected from 0, 1 and 2;
k and m are ndently selected from 1 and 2; and
v and a are independently selected from 0 and 1.
In one embodiment, the invention es a compound of formula (I) or a er or a solvate or a
pharmaceutically acceptable salt thereof, wherein:
R1 is ndently selected from hydroxy, halogen, nitro, nitrile and C1_4alkyl;
R2 is selected from hydrogen, C1_4 alkyl, C2_6alkenyl, hydroxyC1_4alkyl and -CH2COZH;
R3 is hydrogen or -(A),-(CRXRy)q-X;
s and t are independently selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein when R3 is -(A)1-(CRXRy)q-X then (i) at least one ofs, t and q is other than 0 and (ii) when t is 0
then s is 1 and q is otherthan 0;
A is a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic group ses one or
more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms thereof;
X is ed from hydrogen, halogen, -CN and -OR9;
R4 and R5 are independently selected from halogen, nitrile and C1_4alkyl;
R6 is selected from hydrogen and C1_6alkyl;
R7 is selected from cyclic group with 3 to 7 ring members, -CH2-heterocyc|ic group with 3 to 7
ring members, Cg_gcycloalkyl, and -CH2-Cg_8cycloalkyl, wherein said cycloalkyl or heterocyclic groups
may be optionally substituted by one or more RZ groups, and wherein in each instance the heterocyclic
group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms
R9 is selected from hydrogen and C1_6alkyl;
Rx and Ry are independently selected from hydrogen and C1_6alkyl;
RZ is independently selected from n, nitro, nitrile, C1_6alkyl, haloC1_6alkyl, kenyl, hydroxy,
hydroxyC1,6a|kyl, C1_6alkoxy, -C(=O)C1_6alky|, and -N(H)e(C1_4a|kyI)2_e;
n and e are independently selected from 0, 1 and 2
m is selected from 1 and 2; and
a is selected from 0 and 1.
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a e or a
pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected from halogen, hydroxy and nitrile;
R2 is selected from hydrogen, C1_4 alkyl and -CH2COZH;
R3 is -(A)1-(CRXRy)q-X;
A is a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic group comprises one or
more , 2, or 3) heteroatoms ed from N, O, S and ed forms thereof;
s and t are independently selected from O and 1;
q is selected from 0, 1 and 2;
wherein (i) at least one of s, t and q is otherthan 0 and (ii) when t is 0 then sis 1 and q is otherthan 0;
X is selected from hydrogen, n or -OR9;
R4 and R5 are independently selected from halogen;
WO 55860 2016/053042
R6 is selected from hydrogen and C1_6alkyl;
R7 is ed from heterocyclic group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7
ring members, C3_8cycloalkyl, and -CH2-Cg.gcycloalkyl, wherein said cycloalkyl, cycloalkenyl or
heterocyclic groups may be optionally tuted by one or more RZ groups, and wherein in each
instance the heterocyclic group comprises one or more (e.g.1, 2, or 3) heteroatoms selected from N,
O, S and oxidised forms thereof;
R9 is selected from hydrogen and kyl;
RX and Ry are independently selected from hydrogen and C1_6alkyl;
RZ is ndently selected from halogen, nitro, nitrile, and C1_6alkyl;
n is1 and mis 1; and
a is selected from 0 and 1.
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected from n, y and nitrile;
R2 is selected from hydrogen, CM alkyl and -CHZCOZH;
R3 is -(A),-(CRXRy)q-X;
A is a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic group comprises one or
more (e.g.1, 2, or 3) heteroatoms selected from N, O, S and oxidised forms thereof;
s and t are independently selected from 0 and 1;
q is selected from 0, 1 and 2;
wherein (i) at least one of s, t and q is han O and (ii) when t is 0 then sis 1 and q is otherthan 0;
X is selected from hydrogen, halogen and -OR9;
R4 and R5 are independently selected from halogen;
R6 is selected from hydrogen and C1_6alkyl;
R7 is a heterocyclic group with 3 to 7 ring members optionally substituted by one or more RZ ;
R9 is selected from hydrogen and C1_6alkyl;
Rx and Ry are independently selected from hydrogen and C1_6alkyl;
RZ is independently selected from halogen and C1_5alkyl; and
n is, 1 and m is 1; and
ais1.
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a solvate or a
ceutically acceptable salt thereof, wherein:
R1 is halogen (e.g.Cl), C1_4alkynyl (e.g. -CECH), nitrile, hydroxyC1_4alkyl (e.g. CHZOH), -
00,1(CRXRV)VCOOH (e.g. -COOH, -CH2COOH, -OCH2COOH or -C(CH3)ZCOOH, -S(O)d-C1_4alkyl (e.g.
SCH3, SOCHg, or ), -SOz-(1-morpho|inyl) or-P(=O)(RX)2, (e.g. -P(=O)(CH3)2);
nis1or2;
R2 is hydrogen, 01.4 alkyl (e.g. -CH3), hydroxyC1_4alkyl (e.g. CHZOH) or -(CH2)UCOOH (e.g. -
CHZCOOH, -CH2CH2-COZH or-(CH(CH3))-COZH);
the moiety -(CH2)SR3 is selected from:
(point of ment to the oxygen represented by dashed bond or bond terminus indicated by *):
-CHzCH20H _,-\in \DAOH
zCHzOH r0“ D
'CD3 'CH2CH3 QMMIOH
R4 is halogen (e.g. F);
a is 0 or 1;
R5 is halogen (e.g. Cl);
m is 1;
R6 is hydrogen or C1_6alkyl (e.g. -CH3 or 3);
R7 is C1_6alkyl (e.g. -CH3 or -CHzCH3), hydroxyC1_6alkyl (e.g. -CHZOH), -C1,6alkyl-NR"Ry (e.g. -
CH2N(CH3)2), -(CR"Ry)p-CONRXRy ( e.g. -C(=O)N(CH3)2 or -C(=O)NHCH3 or Q), -(CH2)j-O-
kyl (e.g. -CHZOCH3), C3_8cycloa|kyl (e.g. cyclobutyl or cyclohexyl), heterocyclic group with 3 to 7
ring members e.g.
(point of ment represented by dashed bond)
or -CH2-heterocyclic group with 3 to 7 ring members e.g.
(point of attachment represented by dashed bond)
wherein when the moiety R7 comprises a heterocyclic or cycloalkyl group, the heterocyclic group may
be optionally substituted by one or more RZ groups selected from C1_6a|kyl (e.g. methyl), hydroxy,
halogen (e.g. ), -C(=O)C1_5alkyl (e.g. -C(=O)C(CH3)3), ,-COZH (e.g. -CH2COOH or
CHZCHZCOOH or -(CH2),-COZC1_6alkyl (e.g. CHZCHzCOOCHg).
In one embodiment, the ion provides a compound of formula (I) or a tautomer or a solvate or a
pharmaceutically able salt thereof, wherein:
R1 is halogen (e.g.Cl), C1_4a|kyny| (e.g. -CECH), nitrile, hydroxyC1_4alkyl (e.g. CHZOH), -(CH2)VCOOH
(e.g. -COOH), -S(O)d-C1_4alkyl (e.g. SCH3, SOCH3, or SOchg), -SOz-(1-morpholinyl) or -P(=O)(RX)2,
(e.g. -P(=O)(CH3)2);
nis1or2;
R2 is hydrogen, 01.4 alkyl (e.g. -CH3), yC1_4alkyl (e.g. CHZOH) or -(CH2)UCOOH (e.g. -
CHZCOOH);
the moiety -(CH2)SR3 is selected from:
(point of attachment to the oxygen represented by dashed bond or bond terminus indicated by *):
A "I OH
-CH3 ””2 OH X
-CHzCH20H _.--\ANH2 \DAOH
" r0“
-CHZCHZCHZOH 0
D D D D
\/\OH
F/\F ,
""YMCHW \/\0/
A “‘“vfl.E0)
HOI"’bI,
& \/\N/SO2CH2
H O
OH A \“\\\“‘
...--\/°H \Ao
A 2g
“_.-\/CN —_.-"\/\OH
A <> \pNNa
R4 is halogen (e.g. F);
a is 0 or 1;
R5 is halogen (e.g. CI);
mis1;
R8 is hydrogen or C1_6alky| (e.g. -CH3 or -CHZCH3);
R7 is C1_6alky| (e.g. -CH3), hydroxyC1_6a|kyl (e.g. -CHZOH), -(CH2)j-O-C1_5alky| (e.g. H3), -C1_
6alkyI-NRXR" (e.g. -CH2N(CH3)2), -(CRny)p-CONRXRy ( e.g. -C(=O)N(CH3)2 or -C(=O)NHCH3) or
O), heterocyclic group with 3 to 7 ring s e.g.
(point of attachment represented by dashed bond)
or -CH2-heterocyc|ic group with 3 to 7 ring members e.g.
WO 55860
(point of attachment represented by dashed bond)
wherein when R7 comprises a heterocyclic group, the heterocyclic group may be optionally substituted
by one or more RZ groups selected from C1,6alkyl (e.g. methyl).
In one embodiment of formula (I) R7 is a heterocyclic group with 3 to 7 ring members e.g.
(point of attachment represented by dashed bond)
In one embodiment of formula wherein when R7 comprises a heterocyclic group, the heterocyclic
group may be R7 is a heterocyclic group with 3 to 7 ring members optionally substituted by one or
more RZ groups e.g.
(point of attachment represented by dashed bond)
/ N/
o NCHg
or a -CH2-heterocyclic group with 3 to 7 ring s optionally substituted by one or more RZ groups
e.g.
(point of attachment ented by dashed bond)
In one embodiment of formula n when R7 comprises a heterocyclic group, the heterocyclic
group may be R7 is a heterocyclic group with 3 to 7 ring members optionally substituted by one or
more RZ groups e.g.
(point of attachment represented by dashed bond)
/ N/
or a -CH2-heterocyclic group with 3 to 7 ring members optionally substituted by one or more RZ groups
e.g.
(point of attachment represented by dashed bond)
”O “NO\N
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a e or a
pharmaceutically acceptable salt thereof, wherein:
R1 is -Cl, -CN, -OH or-OCH3;
n is 1;
R2 is hydrogen;
R3 is hydrogen or (CRXRy)q-X;
sis00r1,andtis1;
A is selected from cyclopropyl, oxetanyl and tetrahydrofuranyl;
X is selected from hydrogen, fluorine, -CN, -OH and -C(=O)NH2;
q is 0 or 1 and RX and Ry are hydrogen or deuterium;
a is 0 or 1 and R4 is halogen (e.g. fluorine);
R5 is halogen (e.g. Cl);
m is 1;
R6 is C1_4a|ky| (e.g. methyl or ethyl);
R7 is C1_4alkyl (e.g. methyl or ethyl), hydroxle1_4alkyl (e.g. hydroxylmethyl or hydroxyethyl),
methoxyC1_4alkyl (e.g. ymethyl), a heterocyclic group with 5 or 6 ring members (e.g. piperidinyl,
oxanyl, olyl or pyrazolyl) or Cg_6cycloalkyl (e.g. utyl or cyclohexyl) wherein said
heterocyclic group with 5 or 6 ring members and 03.5cycloalkyl groups may be optionally substituted
with one ortwo RZ groups independently selected from methyl, halogen (such as fluorine), -C(=O)Me,
and -OH.
WO 55860
In one embodiment, the invention provides a nd of formula (I) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof, wherein:
R1 is -Cl, -CN, -OH or-OCH3;
n is 1;
R2 is hydrogen or -(CH2)u-COZH wherein u is independently selected from 0 and 1;
R3 is hydrogen and s is 1 or R3 is (CRXRy)q-X and t is 1 and q is 1;
A is selected from ropyl;
X is -OH;
RX and Ry are hydrogen or deuterium;
a is 0 or 1 and R4 is halogen (e.g. fluorine);
R5 is halogen (e.g. Cl);
m is 1;
R6 is C1_4alkyl (e.g. methyl or ethyl);
R7 is C1_4a|kyl (e.g. methyl or ethyl), hydroxy|C1_4alkyl (e.g. hydroxylmethyl or hydroxyethyl),
methoxyC1_4alkyl (e.g. ymethyl), a heterocyclic group with 5 or 6 ring members (e.g. piperidinyl,
oxanyl, imidazolyl or pyrazolyl) or 03.6cycloalkyl (e.g. cyclobutyl or cyclohexyl);
wherein said heterocyclic group with 5 or 6 ring s and C3_5cycloalky| groups may be optionally
substituted with one ortwo RZ groups independently selected from methyl, halogen (such as fluorine),
-C(=O)Me, and -OH.
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof, wherein:
R1 is halogen (e.g.Cl) or nitrile;
n is 1;
R2 is hydrogen or -(CH2)uCOOH (e.g. -CHZCOOH);
R3 is hydrogen and s is 1 or R3 is (CRXRy)q-X and t is 1 and q is 1;
A is selected from cyclopropyl;
X is -OH;
RX and Ry are hydrogen or deuterium (e.g. hydrogen);
R4 is halogen (e.g. F);
a is 0 or 1;
R5 is halogen (e.g. Cl);
m is 1;
R6 is hydrogen or C1_6alkyl (e.g. -CH3 or -CH2CH3);
R7 is C1.4alkyl (e.g. methyl), hydroxle1_4alkyl (e.g. hydroxylmethyl), yC1_4alkyl (e.g.
methoxymethyl), a heterocyclic group with 5 or 6 ring members (e.g. dinyl, oxanyl, imidazolyl or
pyrazolyl));
wherein said heterocyclic group with 5 or 6 ring s may be optionally substituted with one or
two RZ groups independently selected from kyl (e.g. methyl).
In one embodiment, the invention provides a compound of formula (I) or a tautomer or a solvate or a
pharmaceutically acceptable salt thereof, wherein:
R1 is halogen (e.g.Cl), nitrile, RXRY)VCOOH (e.g. -COOH, -CHZCOOH, -OCHZCOOH or -
C(CH3)2COOH;
nis1or2;
R2 is ed from hydrogen and -(RXRy)u-COZH (e.g. -COOH, -CH2COOH, -CHZCH2-COZH, -
(CH(CH3))-COZH and -(C(CH3)2-C02H).
R3 is hydrogen and s is 1;
R4 is halogen (e.g. F);
R5 is halogen (e.g. Cl);
m is 1;
R6 is hydrogen or C1_6alkyl (e.g. -CH3 or -CH2CH3);
R7 is C1.4alkyl (e.g. methyl), hydroxle1_4alkyl (e.g. hydroxylmethyl), methoxyC1_4alkyl (e.g.
methoxymethyl), a heterocyclic group with 5 or 6 ring members (e.g. dinyl, oxanyl, imidazolyl or
pyrazolyl»;
wherein said heterocyclic group with 5 or 6 ring members may be optionally substituted with one or
two RZ groups ndently selected from C1_4alkyl (e.g. methyl).
In one embodiment, the invention provides a compound of a (I) which is one ofthe Examples 1-
137 or is selected from the Examples 1-137 or a tautomer, N-oxide, pharmaceutically acceptable salt
or e thereof.
In one embodiment, the invention provides a compound of formula (I) which is one ofthe Examples 1-
97 or is selected from the Examples 1-97 or a tautomer, N-oxide, pharmaceutically acceptable salt or
solvate thereof.
In one embodiment, the invention provides a compound of formula (I) which is selected from the
following compounds, or a er, N-oxide, pharmaceutically acceptable salt or solvate thereof:
4-{[(1R)(4-chIoropheny|)fluoro[1-hydroxy(1-methyI-1H-imidazolyl)propyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile; and
(38)(4-chlorophenyl)[(1R)(4-chIorophenyl)fluoro[1-hydroxy(oxanyl)ethyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid.
In one embodiment, the ion provides a nd of formula (I) which is diastereoisomer 2A and
is selected from the following compounds, or a tautomer, N-oxide, pharmaceutically acceptable salt or
solvate thereof:
4-{[(1R)(4-chIorophenyl)fluoro[1-hydroxy(1-methyI-1H-imidazolyl)propyl]{[1-
(hydroxymethyl)cyclopropy|]methoxy}oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile; and
(3S)—3-(4-chlorophenyl)[(1 4-chlorophenyl)fluoro[1-hydroxy(oxanyl)ethyl]
methoxyoxo-2,3-dihydro-1H-isoindol-Z-yl]propanoic acid.
In one embodiment, the invention provides a compound of formula (I) which is diastereoisomer 28 and
is selected from the ing nds, or a tautomer, N-oxide, ceutically acceptable salt or
solvate thereof:
R)(4-chIorophenyl)fluoro[1-hydroxy(1-methyI-1H-imidazolyl)propyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile; and
(3S)—3-(4-chlorophenyl)[(1R)(4-chIoropheny|)fluoro[1-hydroxy(oxany|)ethyl]
methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid.
In one embodiment, the invention provides a compound of formula (I) which is 2-(5-chloro{[(1R)
(4-chlorophenyl)f|uoro[(1S)hydroxy(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-
isoindolyl]methyl}phenyl)methylpropanoic acid, or a tautomer, N-oxide, pharmaceutically
acceptable salt or solvate thereof.
In one embodiment, the invention provides a compound of formula (I) which is (28,3S)(4-
chlorophenyl)[(1R)(4-ch|orophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]methoxy
oxo-2,3-dihydro-1H-isoindolyl]methylpropanoic acid, or a tautomer, N-oxide, pharmaceutically
acceptable salt or solvate thereof.
For the avoidance of doubt, it is to be understood that each general and specific embodiment and
example for one substituent may be combined with each general and specific embodiment and
example for one or more, in particular all, other substituents as defined herein and that all such
ments are embraced by this application.
SALTS, SOLVATES, ERS, ISOMERS, N-OXIDES, ESTERS, PRODRUGS AND
ISOTOPES
A reference to a compound of the formula (I), sub-groups thereof (e.g. formulae l(c), l(f), l(g), l(g’), l(h),
l(i), l(i), l(k), l(L), l(m), l(m’). l(n), l(O), |(0’), |(0”), l(p), l(p’), l(q), l(q’), l(q”). , l(q’”), l(r), l(S), l(t),
l(u), l(v), l(v’), l(w), l(x), l(x‘), l(y), (ll), (Ila), (llb), (Illa), (lllb), (lVa), (lVb), (V), (VI), (Via), (VII), ,
(VIIb), (VIIc), , (Vlld’), (Vlle), (We), (a), (b), (ba), (bb), (bc) or (c)) and any example also includes
ionic forms, salts, solvates, isomers (including geometric and stereochemical isomers unless
specified), tautomers, N-oxides, esters, gs, isotopes and protected forms thereof, for example,
as sed below; in particular, the salts or tautomers or isomers or es or solvates thereof;
and more particularly the salts or tautomers or N-oxides or solvates thereof. In one embodiment
reference to a compound of the formula (I), sub-groups thereof (e.g. formulae l(c), l(f), l(g), l(g’), l(h),
l(i), l(i), l(k), l(L), l(m), l(m’). l(n), l(O), |(0’), KG”), KP), l(p’), l(q), l(q’), l(q”). l(q’”), l(q'”), l(r), l(S), l(t),
l(u), l(v), l(v’), l(w), l(x), l(x’), l(y), (ll), (Ila), (llb), (Illa), (lllb), (lVa), (lVb), (V), (VI), (Via), (VII), (Vlla),
(VIIb), (VIIc), (VIId), (Vlld’), (Vlle), (Vlle’), (a), (b), (ba), (bb), (bc) or (c)) and any e also includes
the salts ortautomers or solvates thereof.
Salts
Many compounds of the formula (I) can exist in the form of salts, for example acid addition salts or, in
certain cases salts of organic and inorganic bases such as carboxylate, ate and phosphate
salts. All such salts are within the scope ofthis invention, and references to compounds ofthe formula
(I) include the salt forms of the compounds.
The salts ofthe present invention can be synthesized from the parent compound that contains a basic
or acidic moiety by conventional chemical methods such as methods described in Pharmaceutical
Salts: Properties, Selection, and Use, P. Heinrich Stahl (Editor), Camille G. Wermuth (Editor), ISBN: 3-
026-8, ver, 388 pages, August 2002. Generally, such salts can be prepared by reacting
the free acid or base forms of these compounds with the appropriate base or acid in water or in an
organic solvent, or in a mixture ofthe two; generally, nonaqueous media such as ether, ethyl acetate,
ethanol, panol, or acetonitrile are used.
Acid addition salts (mono- or di-salts) may be formed with a wide variety of acids, both inorganic and
organic. Examples of acid addition salts include mono- or di-salts formed with an acid selected from
acetic, 2,2-dichloroacetic, adipic, alginic, ic (e.g. L-ascorbic), L-aspartic, benzenesulfonic,
benzoic, 4—acetamidobenzoic, ic, (+) ric, camphor—sulfonic, (+)-(1S)-camphor
sulfonic, capric, caproic, caprylic, cinnamic, citric, cyclamic, dodecylsulfuric, ethane-1,2-disulfonic,
ethanesulfonic, oxyethanesulfonic, formic, fumaric, galactaric, gentisic, glucoheptonic, D-
gluconic, glucuronic (e.g. D-glucuronic), glutamic (e.g. L-glutamic), or-oxoglutaric, ic, hippuric,
hydrohalic acids (e.g. hydrobromic, hydrochloric, hydriodic), onic, lactic (e.g. (+)-L-lactic, (i)—DL-
lactic), lactobionic, maleic, malic, malic, malonic, (i)-DL-mandelic, methanesulfonic, alene-
2-sulfonic, naphthalene-1,5-disulfonic, 1-hydroxynaphthoic, nicotinic, nitric, oleic, orotic, oxalic,
palmitic, pamoic, phosphoric, propionic, pyruvic, L-pyroglutamic, salicylic, 4-amino-salicylic, sebacic,
stearic, succinic, sulfuric, , (+)-L-tartaric, thiocyanic, p—toluenesulfonic, undecylenic and valeric
acids, as well as acylated amino acids and cation exchange resins.
2016/053042
One particular group of salts consists of salts formed from acetic, hloric, hydriodic, phosphoric,
nitric, sulfuric, citric, lactic, succinic, , malic, isethionic, fumaric, benzenesulfonic,
toluenesulfonic, methanesulfonic (mesylate), ethanesulfonic, naphthalenesulfonic, valeric, acetic,
propanoic, butanoic, malonic, glucuronic and lactobionic acids. One particular salt is the hydrochloride
salt.
In one ment the compound is the tris(hydroxymethyl)aminomethane (TRIS) salt.
If the compound is anionic, or has a functional group which may be anionic (e.g., -COOH may
be -COO'), then a salt may be formed with an organic or inorganic base, generating a suitable cation.
es of suitable inorganic cations include, but are not limited to, alkali metal ions such as Li+, Na+
and K", alkaline earth metal cations such as Ca2+ and Mg”, and other cations such as Al3+ or Zn".
Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e., NH4+) and
substituted ammonium ions (e.g., NH3R+, NHZRZ", NHRJ, NR4+). Examples of some suitable
substituted ammonium ions are those derived from: methylamine, mine, diethylamine,
propylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine,
diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine,
as well as amino acids, such as lysine and arginine. An example of a common quaternary um
ion is N(CH3)4+.
Where the compounds of the formula (I) n an amine function, these may form quaternary
ammonium salts, for example by on with an alkylating agent according to methods well known to
the skilled person. Such quaternary ammonium compounds are within the scope of a (I).
The compounds of the invention may exist as mono- or di-salts depending upon the pKa of the acid
from which the salt is formed.
The salt forms of the compounds of the invention are typically pharmaceutically acceptable salts, and
examples of pharmaceutically acceptable salts are sed in Berge et al., 1977, aceutically
Acceptable Salts," J. Pharm. Sci., Vol. 66, pp. 1-19. However, salts that are not pharmaceutically
acceptable may also be prepared as ediate forms which may then be converted into
pharmaceutically acceptable salts. Such non-pharmaceutically acceptable salt forms, which may be
useful, for example, in the purification or separation of the compounds of the ion, also form part
of the invention.
In one embodiment of the invention, there is provided a pharmaceutical composition comprising a
solution (e.g. an aqueous solution) containing a compound of the formula (I) and sub-groups and
examples thereof as described herein in the form ofa salt in a concentration of greater than 10 mg/ml,
lly greaterthan 15 mg/ml and typically greater than 20 mg/ml.
N-Oxides
nds of the formula (I) containing an amine function may also form N-oxides. A reference
herein to a compound ofthe formula (I) that ns an amine function also includes the N-oxide.
Where a compound contains several amine functions one, or more than one, nitrogen atom may be
oxidised to form an N-oxide. Particular examples of N-oxides are the N-oxides of a tertiary amine or a
en atom of a nitrogen-containing heterocyclylic group.
N-Oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as
hydrogen peroxide or a per-acid (e.g. a peroxycarboxylic acid), see for e Advanced Organic
Chemistry, by Jerry March, 41h Edition, Wiley Interscience, pages. More particularly, N-oxides can be
made by the procedure of L. W. Deady (Syn. Comm. 1977, 7, 509-514) in which the amine nd
is reacted with m—chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as
dichloromethane.
In one embodiment ofthe ion, the compound is an N-oxide, eg. from a nitrogen atom on the R6
or R7 group, for example a pyridine N-oxide.
Geometric isomers and ers
Compounds of the formula (I) may exist in a number of different geometric isomeric, and tautomeric
forms and nces to compounds of the formula (I) include all such forms. For the nce of
doubt, where a compound can exist in one of several geometric isomeric or tautomeric forms and only
one is specifically described or shown, all others are nevertheless embraced by formula (I).
For example, certain heteroaryl rings can exist in the two tautomeric forms such as A and B shown
below. For simplicity, a formula may illustrate one form but the formula is to be taken as embracing
both tautomeric forms.
0 \
.—_- | N—N N—N
\ NH \ N H H
A B A B
Other examples of tautomeric forms e, for example, keto-, enoI-, and enolate-forms, as in, for
example, the following tautomeric pairs: keto/enol (illustrated below), imine/enamine, amide/imino
alcohol, amidine/enediamines, nitroso/oxime, thioketone/enethiol, and nitro/aci-nitro.
T 00 \ [OH H+
\ 0'
—C—C : C=C : czc’
\ / \ H+ / \
keto enol enolate
Stereoisomers
Unless otherwise mentioned or indicated, the al designation of compounds denotes the e
of all possible chemically isomeric forms.
Stereocentres are illustrated in the usual fashion, using ‘hashed’ or ‘solid’ wedged lines. e.g.
Where a compound is described as a mixture of two reoisomers / epimers, the configuration of
the stereocentre is not specified and is represented by straight lines.
Where compounds of the formula (I) contain one or more chiral centres, and can exist in the form of
two or more optical isomers, references to compounds of the formula (I) include all optical isomeric
forms thereof (e.g. enantiomers, epimers and diastereoisomers), either as individual optical isomers,
or mixtures (e.g. racemic or\ es) or two or more optical isomers, unless the context
requires othenNise.
The optical isomers may be terised and identified by their optical activity (i.e. as + and —
s, or d and I isomers) or they may be characterised in terms of their te stereochemistry
using the “R and S” nomenclature developed by Cahn, lngold and Prelog, see Advanced Organic
Chemistry by Jerry March, 41h Edition, John Vlfiley & Sons, New York, 1992, pages 109-114, and see
also Cahn, lngold & Prelog, Angew. Chem. Int. Ed. Engl., 1966, 5, 385-415.
Optical isomers can be separated by a number of techniques including chiral chromatography
(chromatography on a chiral support) and such techniques are well known to the person skilled in the
art.
As an alternative to chiral chromatography, optical isomers can be separated by forming
diastereoisomeric salts with chiral acids such as (+)-tartaric acid, (-)-pyrog|utamic acid, (-)-di-to|uoy|-L-
tartaric acid, (+)-mandelic acid, (-)-malic acid, and (-)-camphorsulfonic acid, separating the
reoisomers by ential crystallisation, and then dissociating the salts to give the individual
enantiomer of the free base.
Additionally enantiomeric separation can be achieved by covalently g a enantiomerically pure
chiral auxiliary onto the nd and then performing diastereisomer separation using conventional
2016/053042
methods such as chromatography. This is then followed by cleavage of the aforementioned covalent
linkage to generate the riate enantiomerically pure product.
Where compounds of the formula (I) exist as two or more optical isomeric forms, one omer in a
pair of omers may exhibit ages over the other enantiomer, for example, in terms of
biological activity. Thus, in certain circumstances, it may be ble to use as a therapeutic agent
only one ofa pair of enantiomers, or only one of a plurality of diastereoisomers.
Accordingly, the invention es compositions containing a compound ofthe formula (I) having one
or more chiral centres, wherein at least 55% (e.g. at least 60%, 65%, 70%, 75%, 80%, 85%, 90% or
95%) of the compound of the formula (I) is present as a single optical isomer (e.g. enantiomer or
diastereoisomer). In one general ment, 99% or more (e.g. substantially all) of the total amount
of the compound of the formula (I) may be present as a single optical isomer (e.g. enantiomer or
diastereoisomer).
Compounds encompassing double bonds can have an E (entgegen) or Z (zusammen)
stereochemistry at said double bond. Substituents on bivalent cyclic or (partially) saturated radicals
may have either the cis- or trans-configuration. The terms cis and trans when used herein are in
accordance with Chemical Abstracts lature (J. Org. Chem. 1970, 35 (9), 2849-2867), and refer
to the position ofthe substituents on a ring moiety.
Of l interest are those compounds of formula (I) which are stereochemically pure. When a
compound of formula (I) is for ce specified as R, this means that the compound is substantially
free of the S isomer. If a compound of formula (I) is for instance specified as E, this means that the
nd is substantially free of the Z isomer. The terms cis, trans, R, S, E and Z are well known to a
person skilled in the art.
Isotopic variations
The present invention includes all pharmaceutically acceptable isotopically—labeled compounds ofthe
invention, i.e. compounds of formula (I), wherein one or more atoms are replaced by atoms having the
same atomic number, but an atomic mass or mass number different from the atomic mass or mass
number usually found in nature.
Examples of isotopes suitable for inclusion in the compounds of the ion comprise isotopes of
hydrogen, such as 2H (D) and 3H (T), carbon, such as “C, 13C and 14C, chlorine, such as 36Cl, fluorine,
123' 125 131
such as 18F, iodine, such as l and
, l, nitrogen, such as 13N and 15N, oxygen, such as 15O, 17O
and 18O, phosphorus, such as ”P, and sulfur, such as 358.
Certain isotopically-labelled compounds of formula (I), for example, those incorporating a radioactive
isotope, are useful in drug and/or substrate tissue distribution studies. The compounds of formula (I)
can also have valuable stic ties in that they can be used for detecting or identifying the
formation of a complex between a labelled compound and other molecules, peptides, proteins,
enzymes or receptors. The detecting or identifying methods can use compounds that are labelled with
labelling agents such as radioisotopes, s, fluorescent substances, luminous substances (for
example, l, luminol derivatives, luciferin, aequorin and luciferase), etc. The radioactive isotopes
tritium, i.e. 3H (T), and -14, i.e. 14C, are particularly useful forthis purpose in view of their ease
of incorporation and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H (D), may afford certain therapeutic
advantages resulting from greater metabolic stability, for example, increased in vivo half-life or
reduced dosage requirements, and hence may be used in some stances.
In particular, every reference to hydrogen in the ation should be construted to cover 1H and 2H,
whether hydrogen is defined explicitly, or hydrogen is present implicitly to satisfy the nt atom’s
(in particular carbon’s) y.
Substitution with positron emitting isotopes, such as 11C, 18F, 15O and ”N, can be useful in Positron
Emission Topography (PET) studies for examining target occupancy.
IsotopicaIIy-Iabeled compounds of formula (I) can generally be prepared by conventional techniques
known to those skilled in the art or by ses analogous to those bed in the accompanying
Examples and Preparations using an appropriate isotopicaIIy-Iabeled reagents in place of the non-
Iabeled reagent previously employed.
Esters
Esters such as carboxylic acid esters, acyloxy esters and phosphate esters of the compounds of
formula (I) bearing a carboxylic acid group or a hydroxyl group are also embraced by Formula (I).
Examples of esters are compounds containing the group -C(=O)OR, wherein R is an ester substituent,
for example, a CH alkyl group, a C342 heterocyclyl group, or a 05.12 aryl group, typically a CH; alkyl
group. ular examples of ester groups include, but are not limited to, -C(=O)OCH3,
-C(=O)OCH2CH3, -C(=O)OC(CH3)3, and -C(=O)OPh. Examples of acyloxy (reverse ester) groups are
represented by -OC(=O)R, wherein R is an acyloxy substituent, for example, a C15 alkyl group, a 03.12
heterocyclyl group, or a 0542 aryl group, typically a C1_6 alkyl group. Particular examples of acyloxy
groups include, but are not d to, -OC(=O)CH3 (acetoxy), -OC(=O)CHZCH3,
-OC(=O)C(CH3)3, -OC(=O)Ph, and -OC(=O)CH2Ph. Examples of ate esters are those derived
from phosphoric acid.
In one embodiment of the invention, formula (I) includes within its scope esters of compounds of the
formula (I) bearing a carboxylic acid group or a hydroxyl group. In r ment of the
invention, a (I) does not include within its scope esters of compounds ofthe formula (I) bearing a
carboxylic acid group or a yl group.
Solvates and Cmstalline forms
Also encompassed by formula (I) are any polymorphic forms of the compounds, and solvates such as
hydrates, alcoholates and the like.
The compounds of the invention may form solvates, for example with water (i.e., hydrates) or common
organic solvents. As used herein, the term “solvate” means a physical association of the compounds
of the present ion with one or more solvent molecules. This physical association es varying
degrees of ionic and nt bonding, including hydrogen bonding. In certain instances the solvate
will be capable of isolation, for example when one or more solvent molecules are orated in the
crystal lattice of the crystalline solid. The term “solvate” is intended to encompass both solution-phase
and isolatable solvates. Non-limiting examples of suitable es include compounds of the invention
in combination with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid or
ethanolamine and the like. The compounds of the invention may exert their ical effects whilst
they are in solution.
Solvates are well known in pharmaceutical chemistry. They can be important to the processes forthe
ation of a substance (e.g. in relation to their purification, the storage of the substance (e.g. its
stability) and the ease of handling of the substance and are often formed as part of the isolation or
purification stages of a chemical synthesis. A person skilled in the art can determine by means of
standard and long used techniques whether a hydrate or other solvate has formed by the ion
conditions or purification conditions used to prepare a given compound. Examples of such ques
include thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), X-ray
crystallography (e.g. single crystal X-ray crystallography or X-ray powder diffraction) and Solid State
NMR (SS-NMR, also known as Magic Angle Spinning NMR or MAS-NMR). Such techniques are as
much a part of the standard analytical toolkit of the skilled chemist as NMR, IR, HPLC and MS.
Alternatively the skilled person can deliberately form a solvate using crystallisation conditions that
e an amount of the solvent required for the particular solvate. Thereafter the standard s
described , can be used to establish whether solvates had formed.
Furthermore, the compounds ofthe present invention may have one or more rph or ous
crystalline forms and as such are intended to be included in the scope ofthe invention.
Complexes
Formula (I) also includes within its scope complexes (e.g. inclusion xes or clathrates with
compounds such as cyclodextrins, or complexes with metals) of the compounds. Inclusion complexes,
clathrates and metal complexes can be formed by means of methods well known to the d person.
Prodrugs
Also encompassed by formula (I) are any pro-drugs ofthe compounds of the formula (I). By “prodrugs”
is meant for example any compound that is converted in vivo into a biologically active compound ofthe
formula (I).
For example, some prodrugs are esters of the active compound (e.g., a physiologically acceptable
metabolically labile ester). During lism, the ester group (-C(=O)OR) is cleaved to yield the
active drug. Such esters may be formed by esterification, for example, of any of the carboxylic acid
groups (-C(=O)OH) in the parent nd, with, where appropriate, prior protection of any other
reactive groups present in the parent compound, followed by deprotection if ed.
Examples of such metabolically labile esters e those ofthe a -C(=O)OR wherein R is:
kyl (e.g., -Me, -Et, -nPr, -iPr, -nBu, -sBu, -iBu, -tBu);
inoalkyl (e.g., aminoethyl; 2-(N,N-diethylamino)ethyl; 2-(4-morpholino)ethyl); and acyloxy-
C1_7alkyl (e.g., acyloxymethyl; acyloxyethyl; pivaloyloxymethyl; acetoxymethyl; 1-acetoxyethyl; 1-(1-
methoxymethy|)ethyI-carbonxonxyethyI; 1-(benzoyloxy)ethyl; isopro poxy-carbonyloxymethyl;
1-isopro poxy-carbonyloxyethyl; cyclohexyI-carbonyloxymethyl; 1-cyclohexyI-carbonyloxyethyl;
cyclohexonxy-carbonyloxymethyl; 1-cyc|ohexonxy-carbonyloxyethyl; (4-oxanyloxy)
carbonyloxymethyl; 1-(4-oxany|oxy)carbonyloxyethyl; (4-oxanyl)carbonyloxymethyl; and
1-(4-tetrahyd ro pyranyl)carbonyloxyethyl).
Also, some prodrugs are activated enzymatically to yield the active compound, or a compound which,
upon further chemical reaction, yields the active compound (for example, as in antigen-directed
enzyme pro-drug therapy (ADEPT), gene-directed enzyme pro-drug therapy (GDEPT), and ligand-
directed enzyme pro-drug therapy (LIDEPT), etc.). For example, the g may be a sugar
derivative or other glycoside conjugate, or may be an amino acid ester derivative. In one embodiment
formula (I) does not include pro-drugs ofthe compounds ofthe formula (I) within its scope.
METHODS FOR THE ATION OF COMPOUNDS OF A (I)
In this section, as in all other sections of this application unless the context indicates othenNise,
references to a (I) also include all other subformula (e.g. formulae I(c), I(f), I(g), I(g’), I(h), I(i), I(j),
|(k), I(L), I(m), I(m’), I(n). I(O), I(O’), I(O”). KP), |(I0’), I(CI), , , I(CI’”). I(Q’”), I(F), I(S). I(t), I(U), I(V),
I(v’), I(w), I(x), I(x’), I(y), (II), (Ila), (llb), (Illa), (IIIb), (IVa), (IVb), (V), (VI), (Via), (VII), (Vlla), (Vllb),
(Vllc), (Vlld), (Vlld’), (Vlle), (Vlle’), (a), (b), (ba), (bb), (bc) or (c)) and es thereof as defined
herein, unless the context indicates othenNise.
Compounds of the formula (I) can be ed in accordance with synthetic methods well known to
the skilled person.
According to a further aspect ofthe invention there is provided a process for preparing a compound of
a (I), or a tautomer, stereoisomer, N-oxide, pharmaceutically acceptable salt, or solvate thereof
which comprises:
According to a further aspect of the invention there is provided a process for preparing a compound of
formula (I) as hereinbefore defined which comprises:
(a) reacting a compound of a (XV) with an organometallic reagent of the fomula R7M
(where M is a metal) for example a Grignard t ofthe a R7MgBr:
(XV)
wherein R1, R2, R3, R4, R5, R6, a, s m and n are as defined herein;
(b) interconversion of a compound of formula (I) or protected derivative f to a further
compound of formula (I) or protected derivative thereof; and/or
(0) deprotection of a protected derivative of a compound of formula (I); and/or
(d) providing a compound of formula (I) and forming a pharmaceutically acceptable salt of the
compound.
The required intermediates are either commercially available, known in the literature, ed by
methods analogous to those in the literature or prepared by methods ous to those described in
the example experimental procedures below. Other compounds may be prepared by functional group
interconversion of the groups using methods well known in the art.
The general synthetic route for the preparation of compounds of formula XV, a key intermediate is set
out in the Schemes below.
NH2 H
w \ N o
\ N/
A / / B
OH \
(R4 E) I < R5)".
XI /
XII XIV
Scheme 1
Example reagents and conditions for Scheme 1: a) NaOH, H20, CHCI3, 85 °C; A) AcOH, rt; B)
Pb(OAC)4, THF, 0 °C; C) NaClOz, H2NSO3H, CchN, H20, rt; D) i) SOCIZ, DMF, THF, ii) amine, i-
PrzEtN, THF; E) i) SOCIZ, DMF, THF, ii) R3(CH2)S-OH, K2003, THF; F) InBrg, R3(CH2)S-OH, DCE, 85
°C; separation and isolation ofthe 3(R) enantiomer can be achieved at this stage by chiral HPLC.
In Scheme 1, R1, R2, R3, R4 and R5 are as described herein and W represents a leaving group, such
as for example halo, e.g. bromo, or a yl group, such as for example acetyl.
N-aroylhydrazone (XI) can be prepared by condensing benzaldehyde (IX) with benzhydrazide (X).
Reaction with Pb(OAc)4 yields aldehyde (XII), from which a Pinnick oxidation provides acid (XIII). The
appropriate benzylamine can then be used to e 3-hydroxyisoindolinone (XIV), and the R3 -
containing side chain added using l de or lnBr3 and the appropriate alcohol.
Intermediates of formula (XV) can be used as a starting point for the synthesis of compounds of the
present invention having varying functionality in the R3, R6 and R7 positions of Formula |.
Scheme 2 below sets out example procedures for introducing various R6 moieties starting from
intermediates of a (XVI) (which is the compound of a (XV) wherein W is Br).
2016/053042
XVII
XVI H
Scheme 2
Example reagents and conditions for Scheme 2: G) (i) toluene, 1,4-dioxane, LiCI, tributyl(1-
ethoxyvinyl)tin, Pd(PPh3)4, (ii) HCI, HZO/THF. H) MeMgCl, in the presence of ZnClz and/or LaCl3-2LiCl,
THF. Separation and isolation ofthe 3(R) enantiomer can be achieved at any stage by chiral HPLC.
Bromide (XVI) can be converted to methyl ketone (XVII) for example using 1,4-dioxane, LiCl, tributyl(1-
ethoxyviny|)tin, Pd(PPh3)4, and further converted to the alcohol XVlll by reaction with a methyl
Grignard t.
Compounds wherein R6 and R7 are hydrogen, can also be prepared ing to the general tic
Scheme 3. Where R3 contains a hydroxyl group, this can be protected during the synthesis by using
standard protecting groups (e.g. TBDMS, TBDPS). Deprotection can be performed using standard
conditions (e.g. TBAF).
WO 55860 2016/053042
Scheme 3
Example reagents and conditions for Scheme 3: I) HCOOLi.HZO, Aczo, Eth, 4,5-
Bis(diphenylphosphino)-9,Q-dimethylxanthene, Pd(OAc)2, DMF; J) LiBH4, THF.
Compounds of formula (XVIII), first shown in Scheme 2, wherein R6 and R7 are methyl, can also be
prepared according to the general synthetic Scheme 4.
In Scheme 4, an intermediate of formula (XXIV) is prepared from an intermediate of formula (XXIII)
according to procedure F (InBr3 with R3(CH2)S-OH),. The intermediate of formula (XXIV) is then
converted to the compound of formula (XVIII) by a Grignard reaction.
XXIII
XXIV
Scheme 4
Example reagents and conditions: F) lnBr3 R3(CH2)S-OH, DCE, 85 °C; H) MeMgCI, ZnClz, THF, 0 °C.
tion and isolation of the 3(R) enantiomer can be achieved at stage F or H by chiral HPLC.
Compounds of general formula XXX can also be prepared according to Schemes 5 and 6.
Scheme 5
Example reagents and conditions: L) nBuLi, Het-CHO, THF, -78°C ; M) MnOz, MeCN, or l2, Kl, K2C03;
D) i) SOCIZ, DMF, THF, ii) amine, i-PrzEtN, THF or HATU, amine, DIPEA, DMF; E) i) SOCIZ, DMF,
THF, ii) R3(CH2)S-OH, K2C03, THF; F) lnBr3, R3(CH2)s-OH, DCE, 85 °C; N) R7ng in the presence of
ZnClz and/or LaCI3-2LiCI, THF or AI(R7)3, THF or EtLi, ZnEtz, THF. Separation of omers and/or
diastereoisomers at Stages E, F and N can be ed by either chiral and/or achiral HPLC.
Intermediate XIII (where W is Br) is reacted with nBuLi and an appropriate aldehyde to provide alcohol
XXVI which is oxidised to the corresponding ketone (XXVII) either using MnOz or IZ/Kl. ediate
XXVII is then converted into the 3-hydroxyisoindolinone XXIX following procedures D and E (of F)
described above.
Intermediates of formula XXIX can be used as a starting point for the synthesis of compounds of the
present ion having varying functionality in the R7 position of Formula |
Alternatively the R7 substituents are introduced earlier in the synthesis as showen in Scheme 6.
Intermediates of formula XXVII can react with organometallic reagents to e tertiary alcohol
(XXXI) which is then converted to final compounds of Formula I ing procedures D and E (or F)
(Scheme 6).
WO 55860 2016/053042
Scheme 6
Example reagents and ions: N) R7ng in the presence of ZnClz and/or LaCl3-2LiCl, THF or
AI(R7)3, THF or EtLi, ZnEtz, THF; D) i) SOCIZ, DMF, THF, ii) amine, tN, THF or HATU, amine,
DIPEA, DMF; E) i) SOCIZ, DMF, THF, ii) R3(CH2)S-OH, K2C03, THF; F) lan, R3(CH2)S-OH, DCE, 85
°C. Separation of enantiomers andior diastereoisomers at Stages N and E/F can be achieved by either
chiral and/or achiral HPLC.
Compounds of formula XVI (first shown in Scheme 2) can also be used to make compounds of
formula XXIX using methods outlined in Scheme 7. In this case, XVI can be converted into a suitable
boronate using, for example, Miyaura conditions. The boronate is then treated with an appropriate
heterocyclic iodide (or heterocyclic bromide) in the presence of carbon monoxide, a suitable catalyst
(such as Pd(dppf)Cl2.) and a solvent (such as toluene or ansole).
Alternatively, compounds of formula XVI can be treated with an appropriate heterocyclic stannane in
the presence of carbon monoxide, a suitable catalyst [such as Pd(dppf)CI2] and a solvent (such as
DMF) to give compounds of formula XXIX (Scheme 7). tion and isolation of the 3(R)
intermediate can be achieved at any stage using chiral HPLC. Compounds of formula XXIX can then
be progressed to compounds of formula XXX (as shown in Scheme 5),
Pd(dppf)C|2.DCM KOAc
, I (R5)m
/Xm W/X
bis(pinacolato)diboron O
1,4-dioxane, 2 hours (R4Ia
2 R2
R \
CU (R1)
Br n I / "
O o
Boronate
Pd(dppf)C|2.DCM
anisole or toluene |
100°C, 18 hours
SnBua
Pd(dppf)CI2.DCM
110°C, 18 hours
/(R‘)n
XXIX
Scheme 7
Compounds of formula XVI can also be used to make compounds of formula XXIX using methods
outlined in Scheme 8. In this case, compounds of formula XVI can be used to make a Weinreb amide
derivative using N,O-dimethylhydroxylamine hydrochloride in the presence of carbon monoxide and a
suitable palladium catalyst (e.g. Xantphos G3 catalyst) . The Weinreb amide can then be reacted with
an appropriate metallated heterocycle (e.g. the t of 4-bromomethyI-1H-pyrazole and nBuLi in
THF) to give compounds of formula XXIX e 8). Separation and ion of the 3(R)
intermediate can be achieved at any stage using chiral HPLC. Compounds of formula XXIX can then
be progressed to compounds of formula XXX (as shown in Scheme 5).
Xantphos G3 catalyst
CO Na2003
Scheme 8
It will be appreciated that certain compounds can exist in different diastereomeric and/or enantiomeric
forms and that processes for their preparation may make use of enantiomerically pure synthetic
precursors.
Alternatively racemic precursors may be used and the mixtures of diastereoisomers generated in
these process may be separated by methods well known to the person skilled in the art, for e
using iral or chiral preparative chromatography or resolution using diastereomeric derivatives:
for example crystallisation of a salt formed with an enantiomerically pure acid such as L-tartaric acid
(or omerically pure base such as (1 R)phenylethanamine); or enantiomer tion of a
diastereomeric derivative formed by covalently linking a enantiomerically pure chiral ary onto the
compound, followed by separation using conventional methods such as chiral or non-chiral
chromatography. The entioned covalent linkage is then cleaved to generate the appropriate
enantiomerically pure product.
A wide range of well known onal group interconversions are known by a person skilled in the art
for converting a precursor compound to a compound of formula I and are described in Advanced
Organic Chemistry by Jerry March, 41h Edition, John Vlfiley & Sons, 1992. For example possible metal
catalysed onalisations such as using organo-tin reagents (the Stille reaction), Grignard reagents
and reactions with nitrogen nucleophiles are described in ‘Palladium Reagents and Catalysts’ [Jiro
Tsuji, Wiley, ISBN 085032-9] and Handbook of Palladium Chemistry for Organic
Synthesis [Volume 1, Edited by Ei-ichi Negishi, Vlfiley, ISBN 031506-0].
If appropriate, the reactions previously described below are followed or preceded by one or more
reactions known to the skilled of the art and are performed in an appropriate order to achieve the
ite substitutions defined above to afford other compounds of formula (I). Non-limiting examples
of such reactions whose conditions can be found in the ture include:
protection of reactive functions,
deprotection of reactive functions,
halogenation,
dehalogenation,
dealkylation,
tion or arylation of amine, aniline, alcohol and ,
Mitsunobu reaction on hydroxyl groups,
cycloaddition reactions on appropriate groups,
reduction of nitro, esters, cyano, aldehydes,
transition metal-catalyzed ng reactions,
acylation,
sulfonylation/introduction of sulfonyl groups,
saponification/hyd rolysis of ester groups,
amidification ortransesterification of ester groups,
esterification or amidification of carboxylic groups,
halogen exchange,
nucleophilic substitution with amine, thiol or alcohol,
ive amination,
oxime formation on carbonyl and ylamine groups,
S-oxidation,
N-oxidation,
salification.
It will be appreciated that certain compounds e.g. compounds of formulae (I), l(c), l(f), l(g), l(g’), l(h),
l(i), l(i), l(k), l(L), l(m). l(m’). l(n), l(O). l(O’), KG”), KP). l(p’), l(q), l(q’). l(q”). l(q’"), l(q””), l(r). l(S), l(t),
l(u), l(v), l(v'), l(w), l(x), l(x’), l(y), (II), (Ila), (Ilb), (Illa), , (IVa), (IVb), (V), (VI), (Via), (VII), ,
(Vllb), (VIIc), (Vlld), (Vlld’), (Vlle), (We), (a), (b), (ba), (bb), (bc) or (c) can exist in different
diastereomeric and/or enantiomeric forms and that processes for their ation may make use of
enantiomerically pure synthetic precursors.
Alternatively racemic precursors may be used and the mixtures of diastereoisomers generated in
these process may be separated by methods well known to the person skilled in the art, for example
using non-chiral or chiral preparative chromatography or resolution using diastereomeric derivatives:
for example crystallisation of a salt formed with an enantiomerically pure acid such as L-tartaric acid;
or enantiomer separation of a diastereomeric derivative formed by covalently linking a enantiomerically
pure chiral auxiliary onto the compound, followed by separation using conventional s such as
chiral chromatography. The aforementioned covalent linkage is then cleaved to generate the
appropriate enantiomerically pure product.
Certain of the ed intermediates, are either commercially available, known in the literature,
ed by methods analogous to those in the ture or prepared by methods analogous to those
described in the example experimental procedures below. Other compounds may be prepared by
functional group interconversion using methods well known in the art.
In a further embodiment the ion provides a novel intermediate. In one embodiment the invention
es a novel intermediate of (VII), (VIII), (IX), (X), (XI), (XII), (XIII), (XIV), (XV), (XVI), , (XIX),
(XX), (XXI), ) and (XXIV).
Protecting Groups
In many of the reactions described herein, it may be necessary to protect one or more groups to
prevent reaction from taking place at an undesirable location on the molecule. Examples of protecting
groups, and methods of protecting and deprotecting functional groups, can be found in Protective
Groups in Organic Synthesis (T. Green and P. Wuts; 3rd Edition; John Wiley and Sons, 1999).
In particular the compound may be synthesised in ted forms and the protecting groups removed
to generate a compound of formula (I).
A hydroxy group may be protected, for example, as an ether (-OR) or an ester (-OC(=O)R), for
example, as: a t-butyl ether; a tetrahydropyranyl (THP) ether; a benzyl, benzhydryl nylmethyl),
or trityl (triphenylmethyl) ether; a trimethylsilyl or t-butyldimethylsilyl ether; or an acetyl ester (-
CH3).
An aldehyde or ketone group may be protected, for example, as an acetal (R—CH(OR)2) or ketal
(R20(OR)2), respectively, in which the carbonyl group (>C=O) is treated with, for example, a y
alcohol. The aldehyde or ketone group is readily regenerated by hydrolysis using a large excess of
water in the presence of acid.
An amine group may be protected, for example, as an amide (-NRCO-R) or a carbamate (-NRCO-
OR), for example, as: a methyl amide -CH3); a benzyl carbamate (-NHCO-OCH2C6H5, -NH-
Cbz or NH-Z); as a t-butyl carbamate (-NHCO-OC(CH3)3, -NH-Boc); a 2-biphenyIpropyl carbamate
-OC(CH3)2C6H4C6H5, -NH-Bpoc), as a 9-fluorenylmethyl carbamate (-NH-Fmoc), as a 6-
nitroveratryl carbamate (-NH-Nvoc), as a ethylsilylethyl ate (-NH-Teoc), as a 2,2,2-
trichloroethyl carbamate (-NH-Troc), as an allyl ate (-NH-Alloc), or as a 2(-phenylsulfonyl)ethyl
carbamate (-NH-Psec).
2016/053042
Other protecting groups for , such as cyclic amines and heterocyclic N-H groups, include
toluenesulfonyl ) and methanesulfonyl (mesyl) groups, benzyl groups such as a para-
methoxybenzyl (PMB) group and tetrahydropyranyl (THP) groups.
A carboxylic acid group may be protected as an ester for example, as: an C1_7 alkyl ester (e.g., a
methyl ester; a t-butyl ester); a CH haloalkyl ester (e.g., a 01.7 oalkyl ester); a 7 alkylsilyl-
C1_7alkyl ester; or a C5_20 aryl-C1_7 alkyl ester (e.g., a benzyl ester; a nitrobenzyl ester; para-
methoxybenzyl ester. A thiol group may be protected, for example, as a thioether (-SR), for example,
as: a benzyl thioether; an acetamidomethyl ether (-S-CH2NHC(=O)CH3).
Isolation and purification ofthe compounds ofthe invention
The compounds of the ion can be isolated and purified according to rd techniques well
known to the person skilled in the art and examples of such methods include chromatographic
techniques such as column chromatography (e.g. flash chromatography) and HPLC. One technique
of particular usefulness in purifying the compounds is preparative liquid chromatography using mass
spectrometry as a means of detecting the purified compounds emerging from the chromatography
Preparative LC-MS is a standard and effective method used for the purification of small organic
molecules such as the nds described herein. The methods forthe liquid tography (LC)
and mass spectrometry (MS) can be varied to provide better separation of the crude materials and
improved detection of the samples by MS. Optimisation of the preparative gradient LC method will
involve varying columns, volatile eluents and modifiers, and gradients. Methods are well known in the
art for optimising preparative LC-MS methods and then using them to purify compounds. Such
methods are described in Rosentreter U, Huber U.; Optimal fraction collecting in preparative LC/MS; J
Comb Chem; 2004; 6(2), 159-64 and Leister W, Strauss K, Wisnoski D, Zhao Z, Lindsley C.,
Development of a custom high-throughput preparative liquid chromatography/mass spectrometer
platform for the preparative cation and analytical analysis of compound libraries; J Comb Chem;
2003; 5(3); An example of such a system for purifying compounds via preparative LC-MS is
bed below in the Examples section of this application (under the heading “Mass ed
Purification LC-MS System”).
Methods of recrystallisation of nds of formula (I) and salt f can be carried out by
methods well known to the skilled person — see for example (P. Heinrich Stahl r), Camille G.
h (Editor), ISBN: 3026-8, Handbook of Pharmaceutical Salts: Properties, Selection,
and Use, Chapter 8, Publisher Wiley-VCH). Products obtained from an organic reaction are seldom
pure when isolated directly from the reaction mixture. If the compound (or a salt thereof) is solid, it
may be purified and/or crystallized by recrystallisation from a suitable solvent. A good recrystallisation
solvent should dissolve a moderate ty of the substance to be purified at elevated temperatures
but only a small quantity of the substance at lowertemperature. It should dissolve impurities readily at
low temperatures or not at all. Finally, the solvent should be readily removed from the purified
WO 55860
product. This usually means that it has a relatively low boiling point and a person skilled in the art will
know recrystallising solvents for a particular substance, or if that information is not available, test
several solvents. To get a good yield of purified material, the minimum amount of hot solvent to
dissolve all the impure material is used. In practice, 3-5% more solvent than necessary is used so the
solution is not saturated. If the impure compound contains an impurity which is insoluble in the
solvent it may then be removed by tion and then allowing the solution to crystallize. In on, if
the impure compound ns traces of coloured material that are not native to the compound, it may
be removed by adding a small amount of decolorizing agent e.g. ting charcoal to the hot
solution, filtering it and then allowing it to crystallize. y crystallization spontaneously occurs upon
cooling the solution. If it is not, crystallization may be induced by cooling the solution below room
ature or by adding a single crystal of pure material (a seed crystal). Recrystallisation can also
be carried out and/or the yield optimized by the use of an anti-solvent or co-solvent. In this case, the
compound is dissolved in a suitable solvent at elevated temperature, ed and then an additional
solvent in which the required compound has low solubility is added to aid crystallization. The crystals
are then typically isolated using vacuum filtration, washed and then dried, for example, in an oven or
via desiccation.
Other examples of methods for cation include sublimation, which includes an heating step under
vacuum for e using a cold finger, and llization from melt (Crystallization Technology
Handbook 2nd Edition, edited by A. Mersmann, 2001).
BIOLOGICAL EFFECTS
It is envisaged that the compound of the invention will be useful in medicine or therapy. The
compounds of the invention, subgroups and examples thereof, have been shown to t the
interaction of p53 with MDM2. Such inhibition leads to cell erative arrest and apoptosis, which
may be useful in preventing ortreating disease states or conditions described herein, for example the
diseases and conditions discussed below and the diseases and conditions described in the
“Background ofthe Invention” section above in which p53 and MDM2 play a role. Thus, for example, it
is envisaged that the compounds of the invention may be useful in alleviating or reducing the
incidence of cancer.
The compounds ofthe present invention may be useful for the treatment of the adult population. The
compounds of the t invention may be useful forthe treatment ofthe pediatric population.
The compounds of the present ion have been shown to be good inhibitors of the formation of
MDM2-p53 complex. The antagonist compounds of formula (I) are e of binding to MDM2 and
exhibiting potency for MDM2. The efficacies of the compounds of the present invention have been
determined against MDM2/p53 using the assay protocol described herein and other methods known in
the art. More ularly, the nds of the formula (I) and sub-groups thereof have affinity for
MDM2/p53.
Certain nds ofthe invention are those having IC50 values of less than 0.1 pM in particular less
than 0.01 or 0.001 uM.
MDM2/p53 on has been implicated in many es due to its role in a variety of process for
example ar remodelling and antiangiogenic processes and regulation of metabolic pathways, as
well as in oncogenesis. As a consequence of their affinity for MDM2 it is anticipated that the
compounds may prove useful in treating or preventing a range of diseases or conditions ing
autoimmune conditions; diabetes mellitus; chronic inflammatory diseases, for e lupus nephritis,
systemic lupus erythematosus (SLE), autoimmune mediated glomerulonephritis, rheumatoid arthritis,
psoriasis, inflammatory bowel disease, autoimmune diabetes mellitus, Eczema hypersensitivity
ons, asthma, COPD, rhinitis, and upper respiratory tract disease; hyperkeratotic diseases such
as autosomal recessive ital ichthyosis (ARCI); kidney diseases including glomerular disorders,
chronic kidney disease (CKD) renal inflammation, podocyte loss, glomerulosclerosis, proteinuria, and
progressive kidney disease; cardiovascular diseases for example cardiac hypertrophy, restenosis,
arrhythmia, atherosclerosis; ischemic injury associated myocardial infarctions, vascular injury, stroke
and reperfusion injury; vascular proliferative diseases; ocular diseases such as age-related macular
degeneration in particular wet form of age-related macular degeneration, ischemic proliferative
retinopathies such as retinopathy of prematurity (ROP) and diabetic retinopathy, and ioma.
As a consequence of their affinity for MDM2 it is anticipated that the compounds may prove useful in
treating or preventing proliferative disorders such as cancers.
Examples of cancers (and their benign counterparts) which may be treated (or inhibited) include, but
are not limited to tumours of epithelial origin (adenomas and carcinomas of various types including
adenocarcinomas, squamous carcinomas, transitional cell carcinomas and other carcinomas) such as
carcinomas of the bladder and urinary tract, breast, gastrointestinal tract (including the gus,
stomach (gastric), small ine, colon, bowel, colorectal, rectum and anus), liver ocellular
oma), gall bladder and biliary system, exocrine pancreas, kidney (for example renal cell
carcinoma), lung (for example adenocarcinomas, small cell lung carcinomas, non-small cell lung
carcinomas, bronchioalveolar carcinomas and mesotheliomas), head and neck (for example cancers
of the tongue, buccal cavity, larynx, pharynx, nasopharynx, tonsil, salivary glands, nasal cavity and
paranasal sinuses), ovary, fallopian tubes, peritoneum, vagina, vulva, penis, testes, cervix,
myometrium, endometrium, thyroid (for example thyroid follicular carcinoma), brain, adrenal, te,
skin and adnexae (for example melanoma, basal cell oma, squamous cell oma,
keratoacanthoma, dysplastic naevus); haematological malignancies (i.e. leukemias, mas) and
premalignant haematological disorders and disorders of borderline malignancy including
haematological malignancies and related conditions of id lineage (for e acute
lymphocytic leukemia [ALL], chronic lymphocytic leukemia [CLL], B-cell lymphomas such as diffuse
large B-cell lymphoma [DLBCL], follicular lymphoma, Burkitt‘s lymphoma, mantle cell lymphoma, T-cell
lymphomas and mias, natural killer [NK] cell lymphomas, Hodgkin’s lymphomas, hairy cell
leukaemia, monoclonal gammopathy of uncertain significance, plasmacytoma, multiple myeloma, and
post-transplant proliferative disorders), and haematological malignancies and related
conditions of myeloid lineage (for example acute myelogenous leukemia [AML], chronic enous
leukemia [CML], chronic myelomonocytic leukemia [CMML], hypereosinophilic syndrome,
myeloproliferative disorders such as polycythaemia vera, essential thrombocythaemia and primary
myelofibrosis, myeloproliferative syndrome, myelodysplastic syndrome, and promyelocytic leukemia);
tumours of mesenchymal origin, for example sarcomas of soft tissue, bone or cartilage such as
osteosarcomas, fibrosarcomas, chondrosarcomas, rhabdomyosarcomas, osarcomas,
liposarcomas, angiosarcomas, Kaposi’s sarcoma, Ewing’s sarcoma, synovial sarcomas, epithelioid
as, gastrointestinal stromal tumours, benign and malignant cytomas, and
dermatofibrosarcoma protuberans; tumours of the central or peripheral s system (for example
astrocytomas (e.g. gliomas), neuromas and glioblastomas, meningiomas, ependymomas, pineal
tumours and schwannomas); endocrine tumours (for example pituitary tumours, adrenal tumours, islet
cell tumours, yroid tumours, carcinoid tumours and medullary carcinoma of the thyroid); ocular
and adnexal tumours (for example retinoblastoma); germ cell and trophoblastic tumours (for example
mas, seminomas, dysgerminomas, hydatidiform moles and choriocarcinomas); and paediatric
and embryonal tumours (for example medulloblastoma, neuroblastoma, Wilms , and primitive
neuroectodermal tumours); or syndromes, congenital or othenNise, which leave the t susceptible
to malignancy (for example Xeroderma Pigmentosum).
Growth of cells is a closely controlled function. Cancer, a condition of abnormal cell , results
when cells replicate in an uncontrolled manner (increasing in number), uncontrollably grow (getting
larger) andlor experience reduced cell death by apoptosis (programmed cell death), is, or
annoikis. In one embodiment abnormal cell growth is selected from uncontrolled cell proliferation,
excessive cell growth or reduced programmed cell death. In particular, the condition or disease of
abnormal cell growth is a cancer.
Thus, in the pharmaceutical compositions, uses or methods of this ion for treating a disease or
condition comprising abnormal cell growth (i.e. uncontrolled and/or rapid cell growth), the disease or
condition comprising al cell growth in one embodiment is a cancer.
Many diseases are characterized by persistent and unregulated angiogenesis. Chronic proliferative
diseases are often accompanied by profound angiogenesis, which can contribute to or maintain an
inflammatory and/0r proliferative state, or which leads to tissue destruction through the invasive
proliferation of blood vessels. Tumour growth and metastasis have been found to be angiogenesis-
dependent. Compounds ofthe invention may ore be useful in preventing and disrupting initiation
of tumour angiogenesis.
enesis is generally used to describe the development of new or replacement blood s, or
neovascularisation. It is a ary and physiological normal process by which vasculature is
established in the embryo. enesis does not occur, in general, in most normal adult tissues,
exceptions being sites of ovulation, menses and wound g. Many diseases, however, are
characterized by persistent and lated angiogenesis. For instance, in arthritis, new capillary
blood vessels invade the joint and destroy cartilage. In diabetes (and in many different eye diseases),
new vessels invade the macula or retina or other ocular structures, and may cause blindness. The
process of atherosclerosis has been linked to angiogenesis. Tumor growth and asis have been
found to be angiogenesis—dependent. The compounds may be beneficial in the treatment of diseases
such as cancer and metastasis, ocular diseases, arthritis and hemangioma.
Therefore, the compounds of the invention may be useful in the treatment of metastasis and
atic cancers. Metastasis or metastatic disease is the spread of a e from one organ or part
to another jacent organ or part. The cancers which can be d by the compounds of the
invention include primary tumours (i.e. cancer cells at the originating site), local invasion (cancer cells
which penetrate and infiltrate surrounding normal tissues in the local area), and metastatic (or
secondary) tumours ie. s that have formed from ant cells which have circulated through
the bloodstream (haematogenous spread) or via tics or across body cavities (trans-coelomic) to
other sites and tissues in the body. In particular, the compounds of the invention may be useful in the
treatment of metastasis and metastatic cancers.
In one embodiment the haematological malignancies is a leukaemia. In another embodiment the
haematological malignancies is a lymphoma. In one embodiment the cancer is AML. In another
embodiment the cancer is CLL.
In one embodiment the compound of the invention is for use in the prophylaxis or treatment of
leukemia, such as acute or chronic leukaemia, in particular acute myeloid leukaemia (AML), acute
lymphocytic leukaemia (ALL), chronic lymphocytic leukaemia (CLL), or chronic myeloid leukemia
(CML). In one embodiment the nd ofthe invention is for use in the prophylaxis or treatment of
lymphoma, such as acute or chronic lymphoma, in ular Burkitt lymphoma, Hodgkin lymphoma,
dgkin lymphoma or difuse large B-cell lymphoma.
In one ment the compound of the invention is for use in the prophylaxis or treatment of acute
myeloid leukaemia (AML) or acute lymphocytic leukaemia (ALL).
One embodiment includes a compound of the invention for use in the prophylaxis or treatment of
cancer in a patient selected from a sub-population possessing s which are p53 wild-type or
have an MDM2 amplification
The cancers may be cancers which are sensitive to treatment with MDM2 tors. The cancers may
be cancers which overexpress MDM2. The cancer may be cancers which are p53 wild-type.
Particular cancers include those with an MDM2 amplification and/or MDM2 overexpression, for
example, hepatocellular carcinoma, lung, as, osteosarcomas, and Hodgkin e.
Particular cancers include those with wild-type p53. Particulars cancers include those cancer cells with
wild-type p53, particularly but not exclusively, if MDM2 is highly expressed.
WO 55860
In one embodiment the cancer is a p53 functional tumours. In one embodiment this e to be
treated is p53 functional solid and haematological malignancies. In another emboidment the patient to
be treated has p53 mutant tumour for example AML patients with p53 mutant tumour.
In one embodiment the cancer is a tumour of the brain, for example glioma, or neuroblastoma.
In one ment the cancer is a cancer of the skin, for example melanoma.
In one embodiment the cancer is a cancer ofthe lung, for example mesothelioma. In one embodiment
the mesothelioma is ant peritoneal mesothelioma or malignant pleural mesothelioma.
In one embodiment the cancer is a cancer of the gastrointestinal tract, for example GIST, gastric,
colorectal or bowel.
In one embodiment the cancer is osteosarcoma.
In one embodiment the cancer is rcoma.
In one embodiment the cancer is s sarcoma.
In one embodiment, the cancer is Iiposarcoma, soft tissue a, osteosarcoma, oesophageal
cancer, and certain tric ancies including B-cell malignancies.
In one embodiment, the cancer is colorectal, , lung and brain
In one embodiment, the cancer is a paediatric cancer.
Whether a particular cancer is one which is sensitive to MDMZ inhibitors, may be determined by a
method as set out in the section headed “Methods of sis”.
A further aspect provides the use of a compound for the manufacture of a medicament for the
treatment ofa disease or condition as described herein, in particular cancer.
Certain cancers are resistant to treatment with particular drugs. This can be due to the type of the
tumour (most common epithelial malignancies are inherently chemoresistant and prostate is relatively
resistant to currently available regimens of herapy or radiation therapy) or resistance can arise
spontaneously as the disease progresses or as a result of treatment. In this regard, references to
prostate includes prostate with ance towards anti-androgen therapy, in particular abiraterone or
enzalutamide, or castrate-resistant prostate. rly references to multiple myeloma includes
bortezomib-insensitive multiple myeloma or refractory multiple myeloma and references to chronic
myelogenous leukemia includes imitanib-insensitive chronic myelogenous leukemia and tory
chronic myelogenous leukemia. In this regard, references to mesothelioma includes mesothelioma
with resistance s topoisomerase s, alkylating agents, antitubulines, antifolates, platinum
compounds and ion therapy, in particular cisplatin-resistant mesothelioma.
The compounds may also be useful in the treatment of tumour growth, pathogenesis, resistance to
chemo- and radio-therapy by sensitising cells to chemotherapy and as an anti-metastatic agent.
Therapeutic anticancer interventions of all types necessarily increase the stresses imposed on the
target tumour cells. Inhibitors of MDM2/p53 represent a class of chemotherapeutics with the potential
for: (i) sensitizing malignant cells to anticancer drugs and/ortreatments; (ii) alleviating or reducing the
incidence of resistance to anticancer drugs and/or treatments; (iii) ing resistance to anticancer
drugs and/or treatments; (iv) potentiating the activity of anticancer drugs and/or treatments; (v)
delaying or ting the onset of resistance to anticancer drugs and/ortreatments.
In one embodiment the invention provides a compound for use in the treatment of a disease or
condition which is ed by MDM2. In a further embodiment the disease or condition which is
mediated by MDM2 is a cancer which is characterised by overexpression and/or increased ty of
MDM2, or high copy number MDM2 and/or wildtype p53.
A r aspect provides the use of a compound for the manufacture of a medicament for the
treatment ofa disease or condition as ed herein, in particular cancer.
In one embodiment there is provided a nd for use in the prophylaxis ortreatment of a disease
or condition ed by MDM2/p53. In one embodiment there is provided a compound for ting
the interaction between of MDM2 protein with p53.
In one embodiment there is provided a pharmaceutical composition comprising an effective amount of
at least one compound as defined. In a further aspect of the present invention, there is provided a
compound as defined in the present
In one embodiment there is ed a method for the prophylaxis or treatment of cancer comprising
the steps of administering to a mammal a medicament comprising at least one compound as defined.
METHODS OF SIS
Prior to administration of a compound of the formula (I), a patient may be screened to determine
r a disease or ion from which the patient is or may be suffering is one which would be
susceptible to treatment with a compound which inhibits 53. The term ‘patient’ includes human
and veterinary subjects such as primates, in particular human patients.
For example, a biological sample taken from a patient may be analysed to determine whether a
condition or e, such as cancer, that the patient is or may be suffering from is one which is
terised by a genetic abnormality or abnormal protein expression which leads to up-regulation of
the levels of MDM2 or to upregulation of a biochemical pathway ream of MDM2/p53.
Examples of such abnormalities that result in activation or sensitisation of MDM2, loss of, or inhibition
of regulatory pathways impacting on MDM2 expression, up-regulation of receptors or their ligands,
cytogenetic aberrations or presence of mutant variants of the receptors or ligands. Tumours with up-
regulation of MDM2/p53, in particular over-expression of MDM2 or exhibit wild-type p53, may be
particularly sensitive to inhibitors of MDMZIp53. For example, amplification of MDM2 and/or deletion
of its negative regulator such as p14ARF has been identified in a range of cancers as discussion in
the uction section.
The term up-regulation includes elevated expression or over-expression, ing gene amplification
(i.e. multiple gene copies), cytogenetic aberration and sed expression by a transcriptional or
ranslational . Thus, the patient may be subjected to a diagnostic test to detect a marker
characteristic of ulation of MDM2. The term diagnosis includes screening. By marker we
include genetic markers including, for example, the measurement of DNA composition to identify
ce of mutations in p53 or amplification MDM2 or deletion (loss) of p14ARF. The term marker
also includes s which are characteristic of up regulation of MDM2/p53, including protein levels,
protein state and mRNA levels of the aforementioned proteins. Gene amplification includes greater
than 7 copies, as well as gains of n 2 and 7 copies.
The diagnostic tests and screens are typically ted on a biological sample (i.e. body tissue or
body fluids) selected from tumour biopsy s, blood samples (isolation and enrichment of shed
tumour , cerebrospinal fluid, plasma, serum, saliva, stool es, sputum, chromosome
analysis, pleural fluid, peritoneal fluid, buccal smears, skin biopsy or urine.
Methods of identification and analysis of netic aberration, c amplification, mutations and
up-regulation of proteins are known to a person skilled in the art. Screening methods could e,
but are not limited to, standard methods such as DNA sequence is by conventional Sanger or
next-generation sequencing s, reverse-transcriptase polymerase chain reaction (RT-PCR),
RNA sequencing (RNAseq), nanostring hybridisation proximity RNA nCounter assays, or in-situ
hybridization such as fluorescence in situ hybridization (FISH) or allele-specific polymerase chain
reaction (PCR),.
In screening by RT-PCR, the level of mRNA in the tumour is assessed by creating a cDNA copy ofthe
mRNA followed by amplification ofthe cDNA by PCR. Methods of PCR amplification, the selection of
primers, and conditions for amplification, are known to a person skilled in the art. Nucleic acid
manipulations and PCR are carried out by standard methods, as described for example in Ausubel,
PM. et al., eds. (2004) Current Protocols in Molecular y, John Vlfiley & Sons Inc., or Innis, MA.
et al., eds. (1990) PCR Protocols: a guide to methods and applications, Academic Press, San Diego.
Reactions and manipulations involving nucleic acid techniques are also described in Sambrook ef al.,
(2001), 3rd Ed, lar Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press.
Alternatively a commercially available kit for RT-PCR (for example Roche Molecular Biochemicals)
may be used, or methodology as set forth in United States patents 4,666,828; 4,683,202; 4,801,531;
,192,659, 5,272,057, 864, and 6,218,529 and incorporated herein by reference. An example of
an in-situ hybridisation technique for assessing mRNA expression would be fluorescence in-situ
hybridisation (FISH) (see Angerer (1987) Meth. Enzymol., 152: 649).
Generally, in situ hybridization comprises the following major steps: (1) fixation of tissue to be
analyzed; (2) prehybridization treatment of the sample to increase accessibility of target nucleic acid,
and to reduce nonspecific binding; (3) hybridization of the mixture of nucleic acids to the nucleic acid in
the biological structure or tissue; (4) post-hybridization washes to remove nucleic acid fragments not
bound in the hybridization, and (5) detection of the hybridized nucleic acid fragments. The probes
used in such applications are typically labelled, for example, with radioisotopes or fluorescent
reporters. Certain probes are sufficiently long, for example, from about 50, 100, or 200 nucleotides to
about 1000 or more nucleotides, to enable specific hybridization with the target nucleic acid(s) under
stringent conditions. Standard methods for carrying out FISH are described in Ausubel, PM et al.,
eds. (2004) t Protocols in Molecular Biology, John Wiley & Sons Inc and scence In Situ
Hybridization: Technical Overview by John M. S. Bartlett in Molecular Diagnosis of Cancer, Methods
and Protocols, 2nd ed.; ISBN: 1760-2; March 2004, pps. 077-088; Series: Methods in
Molecular Medicine.
Methods for gene expression profiling are described by (DePrimo et al. (2003), BMC Cancer, 3:3).
Briefly, the protocol is as follows: double-stranded cDNA is synthesized from total RNA using a (dT)24
oligomer for priming first-strand cDNA synthesis from polyadenylated mRNA, followed by second
strand cDNA synthesis with random hexamer primers. The double-stranded cDNA is used as a
template for in vitro transcription of cRNA using biotinylated ribonucleotides. cRNA is chemically
fragmented according to protocols described by Affymetrix (Santa Clara, CA, USA), and then
hybridized overnight to pecific ucleotide probes on Human Genome Arrays. Alternatively,
single nucleotide polymorphism (SNP) arrays, a type of DNA microarray, can be used to detect
polymorphisms within a population.
Alternatively, the protein products expressed from the mRNAs may be assayed by
histochemistry of tumour samples, solid phase immunoassay with microtitre plates, Western
ng, 2-dimensional SDS-polyacrylamide gel electrophoresis, ELISA, flow cytometry and other
methods known in the art for detection of specific proteins e.g. capillary electrophoresis. Detection
methods would include the use of site specific dies. The d person will ize that all
such well-known techniques can be used for detection of upregulation of MDM2 and p53, detection of
MDM2 or p53 variants or mutants, or loss of negative regulators of MDM2 in the present case.
Abnormal levels of proteins such as MDM2 or p53 can be measured using standard n assays,
for example, those assays described herein. Elevated levels or overexpression could also be detected
in a tissue sample, for example, a tumourtissue by measuring the protein levels with an assay such as
that from on ational. The protein of interest would be immunoprecipitated from the
sample lysate and its levels measured. Assay s also include the use of markers.
In other words, p53 and MDM2 overexpression can be measured by tumour biopsy.
Methods for ing gene copy changes include techniques commoly used in cytogenetic
laboratories such as MLPA plex Ligation-dependent Probe Amplification) a multiplex PCR
method detecting al copy s, or other PCR techniques which can detect gene
amplification, gain and deletion.
Ex-functional assays could also be utilised where appropriate, for example measurement of circulating
leukemia cells in a cancer patient, to assess the response to challenge with an MDM2/p53 inhibitor.
Therefore all of these techniques could also be used to fy tumours particularly suitable for
treatment with the compounds of the invention.
11’]
2016/053042
Therefore in a further aspect of the invention includes use of a compound ing to the invention
for the manufacture of a medicament forthe treatment or prophylaxis of a disease state or condition in
a patient who has been screened and has been determined as suffering from, or being at risk of
suffering from, a disease or condition which would be susceptible to treatment with an 53
inhibitor.
Another aspect of the invention includes a compound of the invention for use in the prophylaxis or
treatment of cancer in a patient selected from a sub-population possessing amplification of MDM2.
Another aspect of the ion includes a compound of the invention for use in the prophylaxis or
treatment of cancer in a patient selected from a sub-population possessing p53 wild-type.
Another aspect of the invention includes a compound of the invention for use in the prophylaxis or
ent of cancer in a patient possessing loss ofa MDM2 negative regulator such as p14ARF.
MRI determination of vessel normalization (eg. using MRI gradient echo, spin echo, and contrast
enhancement to measure blood volume, relative vessel size, and vascular permeability) in
combination with circulating biomarkers may also be used to identify patients suitable for treatment
with a compound ofthe invention.
Thus a further aspect ofthe ion is a method forthe diagnosis and ent of a disease state or
condition mediated by MDM2/p53, which method comprises (i) screening a patient to determine
whether a disease or condition from which the patient is or may be suffering is one which would be
susceptible to treatment with MDM2/p53 inhibitor; and (ii) where it is indicated that the disease or
condition from which the patient is thus susceptible, thereafter stering to the patient a
compound of formula (I) and oups or examples thereof as defined herein.
Advantages of Compounds of the Invention
The compounds of the formula (I) have a number of advantages over prior art nds.
Compounds ofthe invention may have particular advantage in one or more ofthe following s:
(i) or y;
(ii) Superior in vivo efficacy
(iii) Superior PK;
(iv) Superior metabolic stability;
(v) Superior oral bioavailabilty; and
(vi) Superior chemical properties.
Superior potency and in vivo efficacy
The compounds ofthe formula (I) have increased y for MDM2 and in particular increased cell
potency against cell lines known to be sensitive to MDM2 antagonists.
Enhanced target engagement is a highly ble property in a pharmaceutical compound as it allows
for a reduced dosage ofdrug and a good separation (‘therapeutic window’) between MDM2 activity
and toxic effects.
The nds ofthe a (I) have improved cell potency and/or improved ivity for p53 WT
vs mutant p53 cell lines. As a result of increased potency against MDM2 compounds ofthe invention
may have increased in vivo efficacy in cancer cell lines and in vivo models. In addition the compounds
show selectivity for MDM2 over MDMX, despite the close sequence, structural and functional similarity
between these genetic paralogues.
or PK and metabolic stability
The compounds of the formula (I) may have advantageous ADMET properties for example better
metabolic stability (for example as determined with mouse liver microsomes), a better P450 profile,
short half-life and/or beneficial nce (e.g. low or high clearance). It has also been found that
many compounds ofthe formula (I) have an improved PK profile.
These features could conferthe advantage of having more drug available in the systemic circulation to
reach the appropriate site of action to exert its eutic effect. Increased drug concentrations to
exert pharmacological action in tumours potentially leads to improved efficacy which thereby allows
reduced dosages to be administered. Thus, the compounds of formula (I) should exhibit reduced
dosage requirements and should be more readily formulated and administered.
This results in a good separation (‘therapeutic window’) between MDM2 activity and toxic effects.
Many compounds ofthe formula (I) have a reduction in Cmax required for efficacy (due to better
MDM2 potency and/or PK).
Superior oral bioavailability
Potentially the compounds ofthe ion have physiochemical properties suitable for oral exposure
(oral exposure or AUC). In particular, compounds of the formula (I) may exhibit improved oral
bioavailability or improved ucibility of oral absorption. Oral bioavailability can be defined as the
ratio (F) of the plasma exposure ofa compound when closed by the oral route to the plasma re
of the compound when closed by the intravenous (iv) route, expressed as a percentage.
Compounds having an oral bioavailability (F value) ofgreaterthan 10%, 20% or 30%, more
particularly greater than 40%, are particularly advantageous in that they may be administered orally
rather than, or as well as, by eral administration.
Superior physiochemical properties
The nds of the formula (I) may have advantageous chemical properties in particular
chemical stability in acidic conditions and reduced lipophilicity.
Lipophilicity can be measured using a partition-coefficient (logP) or a distribution-coefficient .
The partition coefficient is a ratio of concentrations of un-ionized compound between two immiscible
phases (n-octanol and water) at equilibrium s the distribution coefficient is the ratio ofthe sum
of the concentrations of all forms ofthe compound (ionized plus un-ionized) in each ofthe two phases.
High lipophilicity is associated with poor drug like ties such us low aqueous solubility, poor
pharmacokinetics properties (low oral bioavailability), undesired drug metabolism and high
promiscuity. Compounds with optimal lipophilicity might have greater chances of success in drug
development. However redued logP (or calculated logP, clogP) can be challenging to achieve whilst
retaining an acceptable level of potency for inhibition of n-protein interactions (PPls) due to the
ilic nature ofthe targets involved.
CEUTICAL FORMULATIONS
While it is possible for the active compound to be administered alone, it is generally presented as a
pharmaceutical composition (e.g. formulation).
Thus, the present invention further provides pharmaceutical compositions, as defined above, and
methods of making a pharmaceutical composition comprising (e.g admixing) at least one compound of
formula (I) (and sub-groups thereof as defined herein), together with one or more pharmaceutically
acceptable excipients and optionally other therapeutic or prophylactic agents as described herein.
The pharmaceutically acceptable excipient(s) can be selected from, for example, carriers (e.g. a solid,
liquid or semi-solid carrier), nts, diluents, fillers or bulking agents, granulating agents, coating
agents, release-controlling agents, binding agents, disintegrants, lubricating agents, preservatives,
antioxidants, buffering agents, suspending agents, thickening agents, flavouring agents, sweeteners,
taste masking agents, stabilisers or any other excipients conventionally used in pharmaceutical
compositions. Examples of excipients for various types of pharmaceutical compositions are set out in
more detail below.
The term “pharmaceutically acceptable” as used herein ns to nds, materials,
compositions, and/or dosage forms which are, within the scope of sound l judgment, suitable
for use in contact with the tissues of a subject (e.g. a human t) t excessive toxicity,
irritation, allergic response, or other problem or complication, commensurate with a reasonable
benefit/risk ratio. Each excipient must also be “acceptable” in the sense of being ible with the
other ingredients ofthe formulation.
Pharmaceutical itions ning compounds of the formula (I) can be formulated in
accordance with known techniques, see for example, Remington’s Pharmaceutical Sciences, Mack
Publishing Company, Easton, PA, USA.
The pharmaceutical compositions can be in any form suitable for oral, parenteral, topical, asal,
intrabronchial, sublingual, ophthalmic, otic, rectal, intra-vaginal, or transdermal stration. Where
the compositions are intended for parenteral administration, they can be formulated for enous,
intramuscular, intraperitoneal, subcutaneous stration or for direct ry into a target organ or
tissue by injection, infusion or other means of delivery. The delivery can be by bolus injection, short-
2016/053042
term infusion or longer term infusion and can be via passive delivery or through the utilisation of a
suitable infusion pump or syringe .
Pharmaceutical formulations adapted for eral administration include aqueous and non-aqueous
e injection solutions which may contain xidants, buffers, bacteriostats, co-solvents, surface
active agents, organic solvent mixtures, cyclodextrin complexation agents, emulsifying agents (for forming
and stabilizing emulsion formulations), me components for forming liposomes, gellable polymers for
forming polymeric gels, lyophilisation tants and combinations of agents for, inter alia, stabilising the
active ingredient in a soluble form and rendering the formulation isotonic with the blood ofthe ed
recipient. Pharmaceutical formulations for parenteral administration may also take the form of aqueous
and non-aqueous sterile suspensions which may include suspending agents and thickening agents (R.
G. Strickly, Solubilizing Excipients in oral and injectable ations, Pharmaceutical Research, Vol
21 (2) 2004, p 201-230).
The ations may be presented in unit-dose or multi-dose containers, for example sealed
ampoules, vials and prefilled syringes, and may be stored in a -dried (lyophilised) condition
requiring only the addition ofthe sterile liquid carrier, for example water for injections, immediately
prior to use. In one embodiment, the formulation is provided as an active pharmaceutical ingredient in
a bottle for subsequent reconstitution using an appropriate diluent.
The pharmaceutical formulation can be prepared by lyophilising a compound of a (I), or sub-
groups thereof. Lyophilisation refers to the procedure of -drying a composition. Freeze-drying
and lyophilisation are therefore used herein as synonyms.
Extemporaneous injection ons and suspensions may be prepared from sterile s, granules
and tablets.
Pharmaceutical compositions of the present ion for eral injection can also comprise
pharmaceutically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or
emulsions as well as sterile powders for titution into sterile injectable solutions or dispersions
just priorto use. Examples of suitable aqueous and nonaqueous rs, diluents, solvents or vehicles
include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like),
carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as sunflower oil, safflower
oil, corn oil or olive oil), and injectable organic esters such as ethyl oleate. Proper fluidity can be
maintained, for example, by the use ofthickening materials such as lecithin, by the maintenance ofthe
required particle size in the case ofdispersions, and by the use of surfactants.
The compositions ofthe present invention may also contain adjuvants such as preservatives, wetting
agents, emulsifying agents, and dispersing agents. tion ofthe action of microorganisms may be
ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben,
chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include agents to adjust
tonicity such as sugars, sodium de, and the like. Prolonged absorption of the injectable
pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as
aluminum earate and gelatin.
In one typical embodiment ofthe invention, the pharmaceutical composition is in a form suitable for i.v.
administration, for e by injection or infusion. For intravenous administration, the solution can be
dosed as is, or can be injected into an infusion bag (containing a pharmaceutically acceptable
excipient, such as 0.9% saline or 5% dextrose), before administration.
In another l embodiment, the ceutical composition is in a form suitable for sub-cutaneous
(s.c.) administration.
Pharmaceutical dosage forms suitable for oral administration include tablets (coated or uncoated),
es (hard or soft shell), caplets, pills, lozenges, , solutions, powders, granules, elixirs and
suspensions, sublingual s, wafers or patches such as buccal patches.
Thus, tablet compositions can contain a unit dosage of active compound together with an inert diluent
or r such as a sugar or sugar alcohol, eg; lactose, sucrose, sorbitol or mannitol; and/or a non-
sugar derived diluent such as sodium carbonate, calcium ate, calcium carbonate, or a cellulose
or derivative thereof such as microcrystalline cellulose (MCC), methyl cellulose, ethyl cellulose,
hydroxypropyl methyl cellulose, and starches such as corn starch. Tablets may also contain such
standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants
(e.g. swellable crosslinked polymers such as crosslinked carboxymethylcellulose), lubricating agents
(e.g. stearates), preservatives (e.g. parabens), antioxidants (e.g. BHT), ing agents (for e
phosphate or citrate buffers), and effervescent agents such as citrate/bicarbonate mixtures. Such
excipients are well known and do not need to be discussed in detail here.
Tablets may be designed to release the drug either upon contact with stomach fluids (immediate
e tablets) or to release in a controlled manner (controlled release s) over a prolonged
period of time or with a specific region of the GI tract.
Capsule formulations may be of the hard gelatin or soft gelatin variety and can contain the active
component in solid, olid, or liquid form. n capsules can be formed from animal gelatin or
synthetic or plant derived equivalents thereof.
The solid dosage forms (eg; tablets, capsules etc.) can be coated or un-coated. Coatings may act
either as a protective film (e.g. a r, wax or varnish) or as a mechanism for controlling drug
release or for aesthetic or fication purposes.. The coating (e.g. a Eudragit TM type polymer) can
be designed to release the active component at a desired location within the gastro-intestinal tract.
Thus, the coating can be selected so as to degrade under certain pH conditions within the
gastrointestinal tract, thereby selectively release the compound in the stomach or in the ileum,
duodenum, jejenum or colon.
Instead of, or in addition to, a coating, the drug can be presented in a solid matrix comprising a release
controlling agent, for example a release delaying agent which may be adapted to release the
compound in a controlled manner in the gastrointestinal tract. Alternatively the drug can be presented
in a polymer coating e.g. a polymethacrylate polymer coating, which may be adapted to selectively
release the compound under ions of varying acidity or alkalinity in the gastrointestinal tract.
Alternatively, the matrix material or release retarding coating can take the form of an erodible polymer
(e.g. a maleic anhydride polymer) which is substantially continuously eroded as the dosage form
passes through the gastrointestinal tract. In another alternative, the coating can be designed to
disintegrate under microbial action in the gut. As a further alternative, the active compound can be
formulated in a delivery system that provides osmotic control of the release ofthe compound. Osmotic
e and other d e or sustained release formulations (for example ations based
on ion exchange resins) may be prepared in accordance with methods well known to those skilled in
the art.
The compound of formula (I) may be ated with a carrier and administered in the form of
nanoparticles, the increased surface area of the nanoparticles assisting their absorption. In addition,
rticles offerthe possibility ofdirect penetration into the cell. rticle drug delivery systems
are described in “Nanoparticle Technology for Drug Delivery”, edited by Ram B Gupta and Uday B.
Kompella, Informa Healthcare, ISBN 9781574448573, published 13th March 2006. Nanoparticles for
drug delivery are also described in J. Control. Release, 2003, 91 (1-2), 2, and in Sinha et 3].,
Mol. Cancer Ther. August 1, (2006) 5, 1909.
The pharmaceutical compositions typically comprise from approximately 1% (w/w) to approximately
95% active ingredient and from 99% (w/w) to 5% (w/w) ofa pharmaceutically acceptable ent or
ation of excipients. Typically, the itions comprise from approximately 20% (w/w) to
approximately 90%,% (w/w) active ingredient and from 80% (w!w) to 10% of a pharmaceutically
acceptable excipient or combination of excipients. The pharmaceutical compositions comprise from
approximately 1% to approximately 95%, typically from approximately 20% to approximately 90%,
active ingredient. Pharmaceutical compositions according to the invention may be, for example, in unit
dose form, such as in the form of ampoules, vials, suppositories, pre-filled syringes, dragées, tablets
or capsules.
The pharmaceutically acceptable excipient(s) can be selected according to the desired physical form
of the formulation and can, for example, be selected from diluents (e.g solid diluents such as fillers or
bulking agents; and liquid diluents such as solvents and co-solvents), disintegrants, buffering ,
lubricants, flow aids, release controlling (e.g. release retarding or delaying polymers or waxes) agents,
binders, granulating agents, pigments, plasticizers, idants, preservatives, flavouring agents,
taste masking agents, tonicity adjusting agents and coating agents.
The skilled person will have the expertise to select the appropriate amounts of ients for use in
the formulations. For example s and capsules typically contain 0-20% disintegrants, 0—5%
lubricants, 0-5% flow aids and/or 0-99% (w/w) fillers/ or g agents (depending on drug dose).
They may also contain 0-10% (w/w) polymer binders, 0-5% (w/w) antioxidants, 0-5% (w/w) pigments.
Slow release tablets would in addition contain 0-99% (w/w) rs (depending on dose). The film
coats of the tablet or capsule typically contain 0-10% (w/w) e-controlling (e.g. delaying)
polymers, 0-3% (w/w) pigments, and/or 0-2% (w/w) plasticizers.
eral formulations lly contain O-20% (w/w) buffers, 0-50% (w/w) cosolvents, and/or O-99%
(w/w) Water for Injection (WFI) (depending on dose and if freeze dried). Formulations for intramuscular
depots may also contain O-99% (w/w) oils.
Pharmaceutical compositions for oral stration can be obtained by combining the active
ingredient with solid rs, if desired ating a ing mixture, and processing the mixture, if
desired or necessary, after the addition of appropriate excipients, into s, dragee cores or
capsules. It is also possible for them to be incorporated into a polymer or waxy matrix that allow the
active ingredients to diffuse or be ed in measured amounts.
The compounds of the invention can also be formulated as solid dispersions. Solid dispersions are
homogeneous extremely fine disperse phases of two or more solids. Solid solutions (molecularly
se systems), one type of solid dispersion, are well known for use in pharmaceutical technology
(see (Chiou and Riegelman, J. Pharm. Sci., 60, 1281-1300 (1971)) and are useful in increasing
dissolution rates and sing the ilability of poorly water-soluble drugs.
This invention also provides solid dosage forms comprising the solid solution described herein. Solid
dosage forms include tablets, capsules, chewable tablets and dispersible or effervescent tablets.
Known excipients can be blended with the solid solution to provide the desired dosage form. For
example, a capsule can contain the solid solution blended with (a) a disintegrant and a lubricant, or (b)
a disintegrant, a lubricant and a surfactant. In addition a capsule can contain a bulking agent, such as
lactose or microcrystalline cellulose. A tablet can contain the solid solution blended with at least one
disintegrant, a lubricant, a surfactant, a g agent and a glidant. A chewable tablet can contain the
solid solution blended with a bulking agent, a lubricant, and ifdesired an additional sweetening agent
(such as an artificial sweetener), and suitable flavours. Solid solutions may also be formed by spraying
solutions ofdrug and a suitable polymer onto the surface of inert carriers such as sugar beads (‘non-
pareils’). These beads can subsequently be filled into capsules or compressed into tablets.
The pharmaceutical formulations may be presented to a patient in “patient packs” containing an entire
course of treatment in a single package, y a blister pack. Patient packs have an age over
traditional prescriptions, where a pharmacist divides a patient's supply of a pharmaceutical from a bulk
, in that the patient always has access to the package insert contained in the patient pack,
normally missing in patient prescriptions. The inclusion of a package insert has been shown to
improve patient compliance with the physician’s instructions.
2016/053042
Compositions for topical use and nasal delivery include ointments, creams, sprays, patches, gels,
liquid drops and inserts (for example intraocular inserts). Such compositions can be formulated in
accordance with known methods.
Examples of formulations for rectal or vaginal administration include pessaries and suppositories
which may be, for example, formed from a shaped moldable or waxy material containing the active
compound. Solutions of the active compound may also be used for rectal administration.
Compositions for administration by inhalation may take the form of inhalable powder compositions or
liquid or powder , and can be administrated in standard form using powder inhaler devices or
aerosol dispensing devices. Such devices are well known. For administration by inhalation, the
powdered formulations typically comprise the active compound together with an inert solid powdered
diluent such as lactose.
The compounds of the formula (I) will generally be presented in unit dosage form and, as such, will
typically contain sufficient compound to provide a d level of biological ty. For e, a
formulation may contain from 1 nanogram to 2 grams of active ingredient, e.g. from 1 nanogram to 2
milligrams of active ingredient. Within these ranges, ular sub-ranges of compound are 0.1
milligrams to 2 grams of active ingredient (more usually from 10 milligrams to 1 gram, e.g. 50
milligrams to 500 milligrams), or 1 microgram to 20 milligrams (for example 1 microgram to 10
milligrams, e.g. 0.1 milligrams to 2 milligrams of active ingredient).
For oral compositions, a unit dosage form may contain from 1 milligram to 2 grams, more typically 10
milligrams to 1 gram, for example 50 milligrams to 1 gram, e.g. 100 miligrams to 1 gram, of active
compound.
The active compound will be stered to a patient in need thereof (for example a human or animal
patient) in an amount sufficient to achieve the desired therapeutic effect.
S OF TREATMENT
The compounds of the formula (I) and sub-groups as defined herein may be useful in the prophylaxis
or treatment of a range of disease states or conditions mediated by MDM2/p53. Examples of such
disease states and conditions are set out above.
The compounds are generally administered to a subject in need of such administration, for example a
human or animal patient, lly a human.
The nds will typically be stered in amounts that are therapeutically or prophylactically
useful and which generally are xic. r, in certain situations (for example in the case of life
threatening diseases), the benefits of administering a compound of the formula (I) may gh the
disadvantages of any toxic effects or side effects, in which case it may be considered desirable to
administer compounds in amounts that are associated with a degree oftoxicity.
The compounds may be administered over a prolonged term to maintain beneficial eutic effects
or may be administered for a short period only. Alternatively they may be administered in a continuous
manner or in a mannerthat provides intermittent dosing (e.g. a pulsatile manner).
A typical daily dose of the compound of formula (I) can be in the range from 100 picograms to 100
milligrams per kilogram of body weight, more typically 5 nanograms to 25 milligrams per kilogram of
bodyweight, and more usually 10 nanograms to 15 milligrams per kilogram (e.g. 10 nanograms to 10
milligrams, and more typically 1 microgram per am to 20 milligrams per kilogram, for example 1
microgram to 10 milligrams per kilogram) per kilogram of bodyweight although higher or lower doses
may be administered where required. The compound ofthe a (I) can be administered on a daily
basis or on a repeat basis every 2, or 3, or 4, or 5, or 6, or 7, or 10 or 14, or 21, or 28 days for
example.
Dosages may also be expressed as the amount ofdrug administered relative to the body surface area
of the patient (mg/m2). A l daily dose of the compound of formula (I) can be in the range from
3700 pg/m2 to 3700 mg/mz, more lly 185 ng;’m2 to 925 mg/mz, and more usually 370 ng/m2 to
555 mg/m2 (e.g. 370 ng/m2 to 370 mg/mz, and more typically 37 mg/m2 to 740 mgfmz, for example 37
mgim2 to 370 mg/mz) although higher or lower doses may be administered where ed. The
nd of the formula (I) can be stered on a daily basis or on a repeat basis every 2, or 3, or
4, or 5, or 6, or 7, or 10 or 14, or 21, or 28 days for example.
The compounds of the invention may be administered orally in a range of doses, for example 0.1 to
5000 mg, or 1 to 1500 mg, 2 to 800 mg, or5 to 500 mg, e.g. 2 to 200 mg or 10 to 1000 mg, particular
examples of doses including 10, 20, 50 and 80 mg. The compound may be administered once or more
than once each day. The compound can be administered continuously (i.e. taken every day without a
break for the duration of the treatment regimen). Alternatively, the compound can be administered
intermittently (i.e. taken continuously for a given period such as a week, then discontinued for a period
such as a week and then taken uously for another period such as a week and so on throughout
the duration of the treatment regimen). Examples of treatment regimens ing intermittent
administration include regimens n administration is in cycles of one week on, one week off; or
two weeks on, one week off; or three weeks on, one week off; ortwo weeks on, two weeks off; or four
weeks on two weeks off; or one week on three weeks off - for one or more cycles, e.g. 2, 3, 4, 5, 6, 7,
8, 9 or 10 or more cycles. This tinuous treatment can also be based upon numbers of days
rather than a full week. For example, the treatment can comprise daily dosing for 1 to 6 days, no
dosing for 1 to 6 days with this pattern repeating during the ent protocol. The number ofdays (or
weeks) wherein the nds of the invention are not dosed do not necessarily have to equal the
number of days (or weeks) wherein the compounds ofthe invention are dosed.
In one embodiment, the compounds ofthe invention can be administered in amounts from 3mg/m2 to
125mg/m2 daily. Treatment can be by continuous daily dosing or more usually consist of multiple
cycles of treatment separated by treatment breaks. One example of a single treatment cycle is 5
consecutive daily doses ed by 3 weeks without treatment.
One particular dosing regimen is once a day (e.g. orally) for a week (e.g. 5 days of treatment),
followed by a treatment break of 1, 2, or 3 weeks. An alternative closing regimen is once a week (e.g.
orally), for 1, 2, 3 or4 weeks.
In one ular dosing schedule, a patient will be given an infusion of a compound of the formula (I)
for periods of one hour daily for up to ten days in particular up to five days for one week, and the
treatment ed at a desired interval such as two to four weeks, in particular every three weeks.
More particularly, a patient may be given an infusion of a compound of the formula (I) for periods of
one hour daily for 5 days and the treatment repeated every three weeks.
In another particular dosing schedule, a t is given an infusion over 30 minutes to 1 hour followed
by maintenance infusions of variable duration, for example 1 to 5 hours, e.g. 3 hours.
The compounds of the invention can also be administered by bolus or continuous on. The
compound ofthe invention can be given daily to once every week, or once every two weeks, or once
every three weeks, or once every four weeks during the treatment cycle. If stered daily during a
treatment cycle, this daily dosing can be tinuous over the number of weeks of the treatment
cycle: for example, dosed for a week (or a number of days), no dosing for a week (or a number of
days, with the pattern ing during the treatment cycle.
In a further particular dosing schedule, a patient is given a continuous infusion for a period of 12 hours
to 5 days, and in particulara continuous infusion of 24 hours to 72 hours.
Ultimately, however, the quantity of compound administered and the type of composition used will be
commensurate with the nature of the disease or physiological condition being treated and will be at the
discretion ofthe ian.
It may be cial to use a compound of the invention as a single agent or to combine the compound
of the invention with another agent which acts via a different mechanism to regulate cell growth thus
treating two of the characteristic features of cancer development. Combination experiments can be
med, for example, as described in Chou TC, Talalay P. Quantitative analysis of dose-effect
relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regulat
198422: 27—55.
The nds as defined herein can be administered as the sole therapeutic agent or they can be
administered in combination therapy with one of more other compounds (or therapies) for treatment of
a ular disease state, for e a neoplastic disease such as a cancer as hereinbefore defined.
For the treatment of the above conditions, the compounds of the invention may be advantageously
employed in ation with one or more other medicinal agents, more particularly, with other anti-
cancer agents or adjuvants (supporting agents in the therapy) in cancer therapy. Examples of other
therapeutic agents or treatments that may be administered together (whether concurrently or at
different time intervals) with the compounds of the formula (I) include but are not limited to:
Topoisomerase l inhibitors
Antimetabolites
Tubulin targeting agents
DNA binder and topoisomerase II inhibitors
Alkylating Agents
Monoclonal Antibodies.
Anti-Hormones
Signal Transduction Inhibitors
Proteasome Inhibitors
DNA methyl transferase inhibitors
Cytokines and ids
Chromatin targeted therapies
Radiotherapy, and,
Other therapeutic or prophylactic agents.
Particular examples of anti-cancer agents or adjuvants (or salts thereof), include but are not limited to
any ofthe agents selected from groups (i)-(x|viii), and optionally group , below:
(i) Platinum nds, for example cisplatin (optionally combined with amifostine),
carboplatin or oxaliplatin;
(ii) Taxane compounds, for example paclitaxel, axel protein bound particles (AbraxaneTM),
docetaxel, cabazitaxel or larotaxel;
(iii) Topoisomerase l inhibitors, for example camptothecin compounds, for e
camptothecin, irinotecan(CPT11), SN-38, or topotecan;
(W) Topoisomerase ll inhibitors, for example anti-tumour epipodophyllotoxins or podophyllotoxin
derivatives for example etoposide, orteniposide;
(V) Vinca alkaloids, for example vinblastine, vincristine, liposomal vincristine (Onco-TCS),
lbine, vindesine, nine or vinvesir;
(Vi) Nucleoside derivatives, for example 5-fluorouracil (5-FU, optionally in combination with
leucovorin), gemcitabine, capecitabine, tegafur, UFT, S1, cladribine, cytarabine (Ara-C,
cytosine arabinoside), fludarabine, clofarabine, or nelarabine;
(vii) tabolites, for example clofarabine, aminopterin, or methotrexate, azacitidine,
cytarabine, floxuridine, pentostatin, thioguanine, thiopurine, 6-mercaptopurine, or
hydroxyurea (hydroxycarbamide);
(viii) Alkylating agents, such as nitrogen ds or nitrosourea, for example cyclophosphamide,
chlorambucil, carmustine (BCNU), bendamustine, pa, melphalan, treosulfan, ine
(CCNU), altretamine, busulfan, dacarbazine, estramustine, fotemustine, ifosfamide
nally in combination with mesna), pipobroman, procarbazine, streptozocin,
lomide, uracil, mechlorethamine, methylcyclohexylchloroethylnitrosurea, or ine
(ACNU);
(W Anthracyclines, anthracenediones and related drugs, for example daunorubicin, doxorubicin
(optionally in combination with dexrazoxane), liposomal formulations of doxorubicin (eg.
CaelyxTM, MyocetTM, DoxilTM), idarubicin, mitoxantrone, epirubicin, amsacrine, or valrubicin;
(X) Epothilones, for example ilone, patupilone, EMS-310705, KOS-862 and ZK-EPO,
epothilone A, lone B, desoxyepothilone B (also known as epothilone D or KOS-862),
aza-epothilone B (also known as EMS-247550), aulimalide, isolaulimalide, or luetherobin;
(Xi) DNA methyl transferase inhibitors, for example temozolomide, azacytidine, or bine;
(xii) Antifolates, for example methotrexate, pemetrexed disodium, or raltitrexed;
(xiii) xic antibiotics, for example antinomycin D, cin, cin C, dactinomycin,
carminomycin, daunomycin, levamisole, plicamycin, or mithramycin;
(xiv) Tubulin-binding agents, for example combrestatin, colchicines or nocodazole;
(XV) Signal Transduction inhibitors such as Kinase inhibitors for example receptortyrosine kinase
inhibitors (e.g. EGFR (epithelial growth factor receptor) inhibitors, VEGFR (vascular
endothelial growth factor receptor) inhibitors, PDGFR (platelet-derived growth factor
receptor) inhibitors, Axl inhibitors, MTKI (multi target kinase inhibitors), Raf inhibitors, ROCK
inhibitors, mTOR inhibitors, MEK inhibitors or P|3K Inhibitors) for e imatinib mesylate,
erlotinib, gefitinib, dasatinib, lapatinib, dovotinib, axitinib, nilotinib, vandetanib, vatalinib,
pazopanib, sorafenib, sunitinib, everolimus
, temsirolimus, (RAD 001), vemurafenib
{PLX4032 or RG7204), dabrafenib, encorafenib, selumetinib (AZD6244), trametinib
(GSK121120212), dactolisib (BEZZ35), buparlisib (BKM-120; NVP-BKM-120), ,
copanlisib (BAY6946), ZSTK-474, CUDC-907, apitolisib 980; 2), pictilisib
(pictrelisib, GDC-0941, 1), GDC-0032, GDC-0068, 36771, idelalisib (formerly
CAL-101, GS 1101, 1), MLN1117 (INK1117), MLN0128 (INK128), lPl-145
(INK1197), LY-3023414, ipatasertib, afuresertib, MK-2206, MK—8156, LY-3023414,
LY294002, SF1126 or , sonolisib (PX-866), or AT13148.
(xvi) Aurora kinase inhibitors for e AT9283, barasertib (AZD1152), 1, MK0457
(VX680), cenisertib (R-763), danusertib (PHA-739358), tib (MLN-8237), or MP-470;
(xvii) CDK inhibitors for example AT7519, roscovitine, seliciclib, alvocidib (flavopiridol), clib
(SCH-727965), 7-hydroxy-staurosporine (UCN-01), JNJ-7706621, EMS-387032 . SNS-
032), PHA533533, ZK-304709, or AZD-5438 and including CDK4 inhibitors such as
palbociclib (PD332991) and ribociclib (LEE-011);
(xviii) PKA/B inhibitors and PKB (akt) pathway inhibitors for example 8, AZ-5363,
Semaphore, SF1126 and MTOR inhibitors such as rapamycin analogues, AP23841 and
AP23573, calmodulin inhibitors (forkhead translocation inhibitors), APl-2/TCN (triciribine),
RX—0201, enzastaurin HCl (LY317615), NL101, SR-13668, PX—316, or KRX—0401
(perifosine/ NSC );
(xix) Hsp90 inhibitors for example onalespib (AT13387), herbimycin, amycin (GA), 17-
allylaminodesmethoxygeldanamycin (17-AAG) e.g. NSC-330507, Kos-953 and CNF-
1010, 17-dimethylaminoethylaminodemethoxygeldanamycin hydrochloride (17-DMAG)
WO 55860 2016/053042
e.g. NSC-707545 and Kos-1022, NVP-AUY922 2296), NVP-BEP800, CNF-2024
(BIIB-021 an oral purine), spib (STA-9090), 22 (SC-102112) or lPl-504;
(XX) Monoclonal Antibodies (unconjugated or conjugated to radioisotopes, toxins or other
agents), antibody derivatives and related agents, such as D, anti-VEGFR, anti-HER2
or anti-EGFR antibodies, for example rituximab , ofatumumab (CD20), ibritumomab
tiuxetan (CD20), GA101 (CD20), tositumomab (CD20), epratuzumab (CD22), lintuzumab
(CD33), gemtuzumab ozogamicin (CD33), alemtuzumab (CD52), galiximab (CD80),
trastuzumab (HER2 antibody), pertuzumab , trastuzumab-DM1 (HER2),
ertumaxomab (HER2 and CD3), cetuximab (EGFR), panitumumab (EGFR), necitumumab
(EGFR), nimotuzumab (EGFR), bevacizumab (VEGF), catumaxumab (EpCAM and CD3),
abagovomab (CA125), farletuzumab (folate receptor), elotuzumab (CS1), denosumab
(RANK ligand), figitumumab (IGF1 R), 871 (IGF1R), mapatumumab (TRAIL receptor),
metMAB (met), mitumomab (GD3 ganglioside), naptumomab estafenatox (5T4), or
siltuximab (IL6) or immunomodulating agents such as CTLA-4 blocking antibodies and/or
antibodies against PD-1 and PD-L1 and/or PD-L2 for example ipilimumab (CTLA4), MK-
3475 (pembrolizumab, formerly lambrolizumab, anti-PD-1), nivolumab (a anti-PD-1), BMS-
936559 (anti- PD-L1), MPDL320A, AMP-514 or MEDI4736 (anti-PD-L1), or tremelimumab
(formerly ticilimumab, CP-675,206, anti-CTLA-4);
(XXi) Estrogen receptor antagonists or selective estrogen receptor tors ) or
inhibitors of estrogen synthesis, for example tamoxifen, fulvestrant, fene, droloxifene,
faslodex, or raloxifene;
(xxi i) Aromatase inhibitors and related drugs, such as exemestane, anastrozole, letrazole,
testolactone aminoglutethimide, mitotane or vorozole;
(xxiii) Antiandrogens (i.e. androgen receptor antagonists) and related agents for example
bicalutamide, mide, flutamide, erone, or nazole;
(xxiv) es and analogues f such as medroxyprogesterone, diethylstilbestrol .
diethylstilboestrol) or octreotide;
(XXV) Steroids for example dromostanolone propionate, megestrol acetate, nandrolone
(decanoate, phenpropionate), fluoxymestrone or gossypol,
(xxvi) Steroidal rome P450 17alpha-hydroxylase-17,20-lyase inhibitor (CYP17), e.g.
abiraterone;
(xxvi i) Gonadotropin ing e agonists or antagonists (GnRAs) for example abarelix,
goserelin acetate, histrelin acetate, leuprolide acetate, triptorelin, buserelin, or elin;
(xxviii) Glucocorticoids, for example prednisone, prednisolone, dexamethasone;
(xxix) Differentiating agents, such as retinoids, rexinoids, vitamin D or retinoic acid and retinoic
acid metabolism blocking agents (RAMBA) for example accutane, alitretinoin, bexarotene, or
tretinoin;
Farnesyltransferase inhibitors for example tipifarnib;
(XXXi) Chromatin targeted therapies such as histone deacetylase (HDAC) inhibitors for example
sodium butyrate, suberoylanilide amide acid (SAHA), depsipeptide (FR 901228),
dacinostat (NVP-LAQ824), R306465/ JNJ-16241199, JNJ-26481585, trichostatin A,
vorinostat, chlamydocin, A-173, JNJ-MGCD-0103, 1, or apicidin;
(xxxi i) Drugs ing the ubiquitin-proteasome pathway including proteasome Inhibitors for
example bortezomib, carfilzomib, 770, MLN-9708, or ONX—0912; NEDD8 inhibitors;
HDM2 nist and deubiquitinases (DUBs);
(xxxi i i) Photodynamic drugs for example porfimer sodium or temoporfin;
(xxxiv) Marine organism-derived anticancer agents such as trabectidin;
(XXXV) Radiolabelled drugs for radioimmunotherapy for example with a beta particle-emitting
isotope (e.g. Iodine -131, Yittrium -90) or an alpha le-emitting isotope
, (e.g., Bismuth-
213 or Actinium-225) for example ibritumomab or Iodine tositumomab or alpha radium 223;
(xxxvi) Telomerase tors for example telomestatin;
(xxxvi i) Matrix metalloproteinase tors for example batimastat, marimastat, prinostat or metastat;
(xxxvi i i) Recombinant interferons (such as eron-y and interferon or) and interleukins (e.g.
interleukin 2), for e aldesleukin, denileukin diftitox, eron alfa 2a, eron alfa
2b, or peginterferon alfa 2b;
(xxxix) Selective immunoresponse modulators for example thalidomide, or lenalidomide;
(xl) Therapeutic Vaccines such as sipuleuceI-T (Provenge) or OncoVex;
(xl i) Cytokine-activating agents include Picibanil, Romurtide, Sizofiran, Virulizin, or in;
(xl i i) Arsenic de;
(xl i ii) Inhibitors of G-protein coupled receptors (GPCR) for example atrasentan ;
(xl iv) Enzymes such as L-asparaginase, pegaspargase, rasburicase, or pegademase;
(xlv) DNA repair inhibitors such as PARP inhibitors for example, olaparib, velaparib, iniparib, INO-
1001, AG-014699, or 31;
(xlvi) Agonists of Death receptor (e.g. TNF-related sis inducing ligand (TRAIL) receptor),
such as mapatumumab (formerly HGS-ETR1), conatumumab rly AMG 655),
PR095780, lexatumumab, dulanermin, CS-1008 or recombinant TRAIL ligands
, apomab
such as recombinant Human TRAIL/Ap02 Ligand;
(xlvii) Immunotherapies such as immune checkpoint inhibitors; cancer vaccines and CAR-T cell
therapy;
(xlviii) Regulators of Cell death (apoptosis) including BcI-2 (B-cell lymphoma 2) antagonists such as
venetoclax (ABT-199 or GDC-0199), ABT—737, ABT-263, TW—37, sabutoclax, obatoclax, and
M|M1 and IAP antagonists including LCL-161 (Novartis), Debio-1143 (Debiopharma /
a), 2, Birinapant / TL-32711 (TetraLogic), CUDC-427 / GDC-0917 / RG-7459
(Genentech), JP1201 (Joyant), T-3256336 (Takeda), GDC-0152 (Genentech) or HGS-1029/
AEG-40826 (HGS/ Aegera);
(xl ix) Prophylactic agents (adjuncts); i.e. agents that reduce or alleviate some of the side effects
associated with chemotherapy agents, for example
anti-emetic agents,
agents that prevent or decrease the duration of chemotherapy-associated neutropenia and
prevent complications that arise from d levels of platelets, red blood cells or white
blood cells, for example interleukin-11 (e.g. ekin), erythropoietin (EPO) and analogues
thereof (e.g. darbepoetin alfa), colony-stimulating factor analogs such as granulocyte
macrophage-colony stimulating factor (GM-CSF) (e.g. sargramostim), and granulocyte-
colony stimulating factor (G-CSF) and analogues thereof (e.g. filgrastim, pegfilgrastim),
— agents that inhibit bone tion such as denosumab or bisphosphonates e.g. zoledronate,
zoledronic acid, pamidronate and ibandronate,
— agents that suppress inflammatory responses such as dexamethasone, prednisone, and
prednisolone,
— agents used to reduce blood levels of growth hormone and IGF-I (and other hormones) in
ts with acromegaly or other rare hormone-producing s, such as synthetic forms
ofthe hormone somatostatin e.g. octreotide acetate,
- te to drugs that decrease levels of folic acid such as leucovorin, or c acid,
— agents for pain e.g. opiates such as morphine, diamorphine and yl,
- non-steroidal anti-inflammatory drugs (NSAID) such as COX-2 inhibitors for example
celecoxib, etoricoxib and lumiracoxib,
- agents for mucositis e.g. palifermin,
— agents for the treatment of side-effects ing anorexia, cachexia, oedema or
thromoembolic episodes, such as megestrol acetate.
Each of the compounds present in the combinations of the invention may be given in individually
varying dose schedules and via different routes. As such, the posology of each of the two or more
agents may differ: each may be administered at the same time or at different times. A person skilled in
the art would know through his or her common general knowledge the dosing regimes and
ation therapies to use. For example, the compound of the invention may be using in
combination with one or more other agents which are administered ing to their existing
combination regimen. Examples of standard combination regimens are provided below.
The taxane compound is advantageously administered in a dosage of 50 to 400 mg per square meter
(mg!m2) of body surface area, for example 75 to 250 mg/m2, particularly for paclitaxel in a dosage of
about 175 to 250 mg/m2 and for docetaxel in about 75 to 150 mg/m2 per course of treatment.
The thecin compound is advantageously administered in a dosage of 0.1 to 400 mg per square
meter (mg/m2) of body surface area, for example 1 to 300 mgfmz, particularly for irinotecan in a
dosage of about 100 to 350 mg/m2 and fortopotecan in about 1 to 2 mg/m2 per course tment.
The umour podophyllotoxin derivative is advantageously stered in a dosage of 30 to 300
mg per square meter (mgfmz) of body surface area, for example 50 to 250mg!m2, ularly for
etoposide in a dosage of about 35 to 100 mg/m2 and for teniposide in about 50 to 250 mg/m2 per
course of treatment.
The anti-tumour vinca alkaloid is advantageously administered in a dosage of 2 to 30 mg per square
meter (mg/m2) of body surface area, particularly for vinblastine in a dosage of about 3 to 12 mg/m2 ,
for vincristine in a dosage of about 1 to 2 mg/m2 and for vinorelbine in dosage of about 10 to 30
mglm2 per course of treatment.
The anti-tumour nucleoside derivative is advantageously stered in a dosage of 200 to 2500 mg
per square meter (mg/m2) of body surface area, for example 700 to
1500 mg/mz, particularly for 5-FU in a dosage of 200 to 500mg/m2, for gemcitabine in a dosage of
about 800 to 1200 mg/m2 and for capecitabine in about 1000 to
2500 mg/m2 per course oftreatment.
The alkylating agents such as en mustard or nitrosourea is advantageously administered in a
dosage of 100 to 500 mg per square meter (mg/m2) of body surface area, for example 120 to 200
mglmz, ularly for cyclophosphamide in a dosage of about 100 to 500 mg/m2 for chlorambucil in
a dosage of about 0.1 to 0.2 mg/kg, for tine in a dosage of about 150 to 200 mg/m2 and for
lomustine in a dosage of about 100 to 150 mg/m2 per course oftreatment.
The anti-tumour anthracycline derivative is advantageously administered in a dosage of 10 to 75 mg
per square meter (mg/m2) of body e area, for example 15 to
60 mg/m2, particularly for doxorubicin in a dosage of about 40 to 75 mg/m2, for daunorubicin in a
dosage ofabout 25 to 45mg/m2 and for idarubicin in a dosage ofabout 10 to 15 mg/m2 per course of
treatment.
The antiestrogen agent is advantageously administered in a dosage of about 1 to 100 mg daily
depending on the particular agent and the condition being treated. Tamoxifen is advantageously
administered orally in a dosage of5 to 50 mg, typically 10 to 20 mg twice a day, continuing the therapy
for sufficient time to achieve and maintain a therapeutic effect. Toremifene is advantageously
administered orally in a dosage of about 60mg once a day, continuing the y for sufficient time to
achieve and maintain a therapeutic effect. Anastrozole is advantageously administered orally in a
dosage of about 1mg once a day. Droloxifene is advantageously administered orally in a dosage of
about 20-100mg once a day. Raloxifene is advantageously stered orally in a dosage of about
60mg once a day. Exemestane is advantageously administered orally in a dosage of about 25mg once
a day.
Antibodies are advantageously administered in a dosage of about 1 to 5 mg per square meter (mgfmz)
of body surface area, or as known in the art, ifdifferent. Trastuzumab is advantageously administered
in a dosage of 1 to 5 mg per square meter (mg/m2) of body surface area, particularly 2 to 4mg/m2 per
course of ent.
Where the compound of the formula (I) is administered in combination therapy with one, two, three,
four or more other therapeutic agents (typically one ortwo, more typically one), the compounds can be
stered simultaneously or sequentially. In the latter case, the two or more compounds will be
administered within a period and in an amount and manner that is sufficient to ensure that an
advantageous or synergistic effect is ed. When stered sequentially, they can be
administered at closely spaced intervals (for example over a period of 5-10 minutes) or at longer
intervals (for example 1, 2, 3, 4 or more hours apart, or even longer periods apart where required), the
precise dosage regimen being commensurate with the properties of the therapeutic s). These
dosages may be administered for example once, twice or more per course oftreatment, which may be
repeated for example every 7, 14, 21 or 28 days.
It will be appreciated that the typical method and order of administration and the respective dosage
s and regimes for each component of the combination will depend on the particular other
medicinal agent and compound of the present invention being stered, their route of
administration, the particulartumour being treated and the particular host being d. The optimum
method and order of administration and the dosage amounts and regime can be readily determined by
those skilled in the art using conventional methods and in view of the information set out herein.
The weight ratio of the compound according to the present invention and the one or more other
ncer agent(s) when given as a combination may be determined by the person skilled in the art.
Said ratio and the exact dosage and frequency of administration depends on the particular compound
according to the invention and the other anticancer agent(s) used, the particular condition being
d, the severity ofthe condition being treated, the age, weight, gender, diet, time of administration
and general physical condition of the particular patient, the mode of stration as well as other
medication the individual may be taking, as is well known to those skilled in the art. Furthermore, it is
evident that the effective daily amount may be d or increased depending on the response ofthe
treated subject and/or ing on the evaluation ofthe physician prescribing the compounds ofthe
t invention. A particular weight ratio for the present compound of formula (I) and another
anticancer agent may range from 1/10 to 10/1, more in particular from 1/5 to 5/1, even more in
particular from 1/3 to 3/1.
The compounds of the invention may also be administered in conjunction with non-chemotherapeutic
treatments such as radiotherapy, photodynamic therapy, gene therapy; surgery and controlled diets.
herapy may be for radical, palliative, adjuvant, neoadjuvant or prophylactic purposes.
The compounds ofthe present invention also have therapeutic applications in sensitising tumour cells
for radiotherapy and chemotherapy. Hence the compounds of the present invention can be used as
"radiosensitizer" and/or “chemosensitizer” or can be given in combination with another
"radiosensitizer" and/or “chemosensitizer”. In one embodiment the compound of the invention is for
use as chemosensitiser.
The term "radiosensitizer" is defined as a molecule stered to patients in eutically effective
amounts to se the sensitivity ofthe cells to ionizing ion and/orto promote the treatment of
diseases which are treatable with ionizing radiation.
The term “chemosensitizer” is defined as a molecule administered to patients in therapeutically
ive amounts to increase the sensitivity of cells to chemotherapy and/or promote the treatment of
diseases which are treatable with chemotherapeutics.
Many cancer treatment protocols currently employ radiosensitizers in conjunction with radiation of x-
rays. Examples of x-ray activated radiosensitizers include, but are not limited to, the following:
metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, azole, nimorazole,
mitomycin C, RSU 1069, SR 4233, E09, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5-
iododeoxyuridine (lUdR), bromodeoxycytidine, fluorodeoxyuridine (FudR), hydroxyurea, cisplatin, and
therapeutically effective analogs and derivatives of the same.
Photodynamic therapy (PDT) of cancers employs visible light as the radiation activator of the
sensitizing agent. Examples of photodynamic radiosensitizers include the following, but are not limited
to: hematoporphyrin derivatives, Photofrin, benzoporphyrin derivatives, tin etioporphyrin, pheoborbide-
a, bacteriochlorophyll-a, naphthalocyanines, ocyanines, zinc ocyanine, and therapeutically
effective analogs and derivatives ofthe same.
ensitizers may be stered in conjunction with a eutically effective amount of one or
more other compounds, including but not limited to: compounds which promote the incorporation of
radiosensitizers to the target cells; compounds which control the flow oftherapeutics, nutrients, and/or
oxygen to the target cells; chemotherapeutic agents which act on the tumourwith or without additional
radiation; or other therapeutically effective nds for treating cancer or other diseases.
ensitizers may be administered in ction with a therapeutically effective amount of one or
more other compounds, including but not limited to: compounds which promote the incorporation of
Chemosensitizers to the target cells; nds which control the flow of therapeutics, nutrients,
andIor oxygen to the target cells; chemotherapeutic agents which act on the tumour or other
eutically effective compounds for treating cancer or other disease. Calcium nists, for
example verapamil, are found useful in combination with oplastic agents to establish
chemosensitivity in tumor cells resistant to accepted chemotherapeutic agents and to potentiate the
efficacy of such compounds in drug-sensitive malignancies.
For use in ation therapy with another chemotherapeutic agent, the compound ofthe formula (I)
and one, two, three, four or more other therapeutic agents can be, for example, formulated together in
a dosage form containing two, three, four or more therapeutic agents i.e. in a y pharmaceutical
composition containing all components. In an alternative, the individual eutic agents may be
formulated separately and presented together in the form of a kit, optionally with instructions for their
use.
In one embodiment the pharmaceutical composition comprises a compound of formula I together with
a pharmaceutically acceptable carrier and optionally one or more therapeutic agent(s)
In another embodiment the invention relates to the use of a combination according to the invention in
the manufacture of a pharmaceutical composition for inhibiting the growth of tumour cells.
In a further embodiment the invention relates to a product containing a compound of a I and one
or more anticancer agent, as a combined ation for simultaneous, separate or tial use in
the treatment of patients ing from cancer.
EXAMPLES
The invention will now be illustrated, but not limited, by reference to the specific ments
described in the ing examples. Compounds are named using an automated naming package
such as AutoNom (MDL) or ChemAxon ure to Name or are as named by the chemical supplier.
In the examples, the ing abbreviations are used:
AcOH acetic acid
Boc tert—butyloxycarbonyl
Boc-Abu-OH (S)(Boc-amino)butyric acid
BuLi butyllithium
CDI 1,1-carbonyldiimidazole
DAST Diethylaminosulfur trifluoride
DCM dichloromethane
DCMA Dicyclohexyylmethylamine
DIPEA N-ethyl-N-(1-methylethyl)- 2-propylamine
DMC dimethyl carbonate
DMF N,N-dimethylformamide
DMSO dimethyl sulfoxide
EDC 1-ethyl(3‘-dimethylaminopropyl)—carbodiimide hydrochloride
Et3N triethylamine
EtOAc ethyl acetate
EtOH ethanol
EtZO diethyl ether
2-(7-aza-1H-benzotriazoleyI)-1,1,3,3-tetramethyluronium
HATU
hexafluorophosphate)
HBTU O-benzotriazole-N,N,N’, ’-tetramethyl-uronium-hexafluoro-phosphate
HCI hydrochloric acid
HOAc acetic acid
HOAt 1-hyd roxyaza benzotriazole
HOBt 1-hyd roxybenzotriazole
HPLC high pressure liquid chromatography
IPA isopropyl alcohol
KHMDS potassium hexamethyldisilazide
LiHMDS lithium bis(trimethy|silyl)amide
MeCN acetonitrile
MeOH methanol
mins. s
MS mass spectrometry
MW microwave
NaBH(OAc)3 sodium triacetoxyborohydride
NaOtBu potassium tert—butoxide
NMP N-methyl-Z-pyrrolidinone
NMR nuclear magnetic resonance spectroscopy
Pd2(dba)3 tris(dibenzylideneacetone)dipalladium (o)
Pd(OAc)2 palladium (2) acetate
3)4 tetrakis(triphenylphosphine)palladium (0)
petrol petroleum ether fraction with boiling point range 40 - 60 °C
PyBrop bromo-tris-pyrrolidino-phosphonium hexafluorophosphate
RT room temperature
SiOz silica
TBTU
N,N,N',N'—tetramethyl-O-(benzotriazolyl)uronium tetrafluoroborate
TEA triethylamine
TFA trifluoroacetic acid
THF tetrahydrofuran
UV Ultraviolet
Column chromatography
Purification using column chromatography can be achieved, for example using a Biotage automated
flash purification system with UV monitoring at 298 nm and collection at 254 nm. Biotage automated
chromatography cked silica cartridges were used in most cases. Where stated, the purification
of some compounds was performed using Biotage C18 reversed phase silica columns, which have
octadecyl (end-capped) functionalised silica or Biotage KP-NH cartridges were used for the separation
of highly polar compounds, which uses y amine bonded silica.
Where ary, semi-preparative HPLC can be carried out, for example using one of the following
es: (i) Varian Prostar Modular HPLC system with a binary g system, UV or and
fraction collector and controlled by Varian Star software. (ii) Agilent 1200 HPLC system with a binary
pump, autosampler, on collector and diode array detector and controlled by Agilent ChemStation
Analytical LC-MS system description
In the following examples, many of the compounds prepared were characterised by mass
spectroscopy using the systems and suitable operating conditions set out below. Where atoms with
different isotopes are present and a single mass quoted, the mass quoted for the compound is the
monoisotopic mass (i.e. 35Cl; 79
Br etc.). Several systems can be used, as described below, and these
can be equipped with, and can be set up to run under, closely similar operating conditions. Possible
operating conditions are also bed below.
Agilent 12008L-6140 LC-MS system - RAPID:
HPLC System: Agilent 1200 series SL
Mass Spec Detector: Agilent 6140 single quadrupole
Second Detector: Agilent 1200 MWD SL
Agilent MS running conditions:
Capillary voltage: 3000V on ES pos (2700V on ES Neg)
Fragmentor/Gain: 190 on ES pos (160 on ES neg)
Gain: 1
Drying gas flow: 12.0 L/min
Gas Temperature: 345 °C
Nebuliser Pressure: 60 psig
Scan Range: 0 amu
lonisation Mode: oSpray Positive-Negative switching
Shimadzu Nexera LC-MS system
HPLC System: Shimadzu SlL-30AC autosampler/2x Shimadzu LC-30AD pumps
Mass Spec Detector: Shimadzu 020 single quadrupole MS
Second Detector: Shimadzu SPD-M20A diode array or
Shimadzu MS running conditions:
Qarray DC voltage: 20V on ES Pos (-20V on ES Neg)
Drying gas flow: 20.0 L/min
DL Temperature: 300 °C
Heat Block ature: 350 °C
Nebulising Gas Flow: 1.5 L/min
Scan Range: 100-750 amu
lonisation Mode: ElectroSpray Positive-Negative switching
Mass Directed Purification LC-MS System
Preparative LC-MS is a standard and effective method used for the purification of small organic
molecules such as the compounds described herein. The methods forthe liquid chromatography (LC)
and mass spectrometry (MS) can be varied to provide better tion of the crude materials and
ed detection of the samples by MS. Optimisation of the preparative gradient LC method will
involve varying columns, volatile s and modifiers, and gradients. Methods are well known in the
art for optimising preparative LC-MS methods and then using them to purify compounds. Such
methods are described in Rosentreter U, Huber U.; Optimal fraction collecting in preparative LC/MS; J
Comb Chem; 2004; 6(2), 159-64 and r W, Strauss K, Wisnoski D, Zhao Z, ey C.,
Development of a custom high-throughput preparative liquid chromatography/mass spectrometer
platform for the preparative purification and analytical analysis of compound libraries; J Comb Chem;
2003; 5(3); 322-9.
Several systems for purifying nds via preparative LC-MS are described below although a
person skilled in the art will appreciate that alternative systems and methods to those bed could
be used. In particular, normal phase ative LC based methods might be used in place of the
reverse phase methods described here. Most preparative LC-MS systems utilise reverse phase LC
and volatile acidic modifiers, since the approach is very effective for the purification of small molecules
and because the eluents are compatible with positive ion electrospray mass spectrometry. Employing
other chromatographic ons e.g. normal phase LC, alternatively buffered mobile phase, basic
modifiers etc as outlined in the analytical methods described above could alternatively be used to
purify the compounds.
Preparative LC-MS system description:
Waters Fractionlynx system:
0 Hardware:
2767 Dual Loop Autosampler/Fraction Collector
2525 preparative pump
CFO n fluidic organiser) for column selection
RMA (Waters reagent manager) as make up pump
Waters 20 Mass Spectrometer
Waters 2996 Photo Diode Array detector
Waters 20 Mass Spectrometer
0 Software:
Masslynx 4.1
0 Waters MS g conditions:
Capillary e: 3.5 W (3.2 W on ES Negative)
Cone voltage: 25 V
Source Temperature: 120 °C
Multiplier: 500 V
Scan Range: 0 amu
lonisation Mode: ElectroSpray Positive o_r
ElectroSpray Negative
Agilent 1100 LC-MS preparative system:
0 Hardware:
Autosampler: 1100 series “prepALS”
Pump: 1100 series “PrepPump” for preparative flow gradient and 1100 series ump” for pumping
modifier in prep flow
UV detector: 1100 series “MWD” Multi Wavelength or
MS detector: 1100 series “LC-MSD VL”
Fraction Collector: 2 x “Prep-FC”
Make Up pump: “Waters RMA”
2016/053042
Agilent Active Splitter
0 Software:
ation: Chem32
o Agilent MS running conditions:
ary voltage: 4000 V (3500 V on ES Negative)
FragmentorIGain: 150/1
Drying gas flow: 13.0 L/min
Gas Temperature: 350 °C
Nebuliser Pressure: 50 psig
Scan Range: 125-800 amu
lonisation Mode: ElectroSpray ve o_r
ElectroSpray Negative
Columns: A range of commercially available columns — both achiral and chiral - may be used such
that, in conjunction with the changes in mobile phase, organic modifier and pH, they enabled the
greatest cover in terms ofa broad range of selectivity. All columns were used in accordance with the
manufacturers ended operating conditions. Typically 5 micron particle sized columns were
used where available. For example, columns from Waters (including but not limited to XBridge Prep
Phenyl 5p OBD 100x19mm, XBridge Prep C18 5p OBD 100x19mm, Waters Atlantis Prep T3 OBD 5p
100x19mm and SunFire Prep C18 OBD 5p 100x19mm), Phenomenex ding but not limited to
Synergy MAX-RP and LUXTM Cellulose-2), Astec (ChirobioticTM columns including but not limited to V,
V2 and T2) and Diacel® (including but not limited to Chiralpak® AD-H) were available for screening.
Eluents: Mobile phase eluent was chosen in conjunction with column manufacturers recommended
stationary phase limitations in orderto optimise a columns tion performance.
Methods: According to the analytical trace the most appropriate preparative chromatography type was
chosen. A typical routine was to run an analytical LC-MS using the type of chromatography (low or
high pH) most suited for nd ure. Once the analytical trace showed good chromatography
a suitable preparative method ofthe same type was chosen.
t: All compounds were usually dissolved in 100% MeOH or 100% DMSO or 90:10
Methanol:Water + 0.2% Formic Acid.
ritical Fluid Chromatography (SFC)
In some cases, final compounds were purified by Supercritcal Fluid Chromatography (SFC) using a
Waters Thar Prep100 preparative SFC system (P200 C02 pump, 2545 r pump, 2998 UVfVIS
detector, 2767 liquid handler with Stacked ion Module). The Waters 2767 liquid handler acted as
both auto-sampler and fraction collector.
The column used for the preparative purification of the compounds was a Diacel Chiralpak lA/lB/IC,
YMC Amylose / Cellulose C or Phenomenex Lux Cellulose-4 at 5um 20 - 21.2 x 250 mm unless
otherwise stated.
WO 55860
Appropriate isocratic methods were selected based on methanol, ethanol or isopropanol solvent
systems under un-modified or basic conditions. The standard method used was typically 5-55%
modifier / COZ, 100ml/min, 120 Bar backpressure, 400C column temperature.
All compounds were screened analytically prior to the purification step. Each sample was run under
both un-modified and basic conditions (5.0ul injection, 5/95 gradient for 5 minutes) across ethanol,
methanol and isopropanol. If necessary, secondary screen across extended solvents such as
acetonitrile, ethyl acetate and THF may also be considered. A decision was then made by the analyst
as to what pH and which isocratic condition to use depending on where the desired product elutes and
the separation ed.
The modifier used under basic conditions was diethyl amine (0.1% V/\/). Occasionally formic acid
(0.1% V/V) may be used as an acidic modifier.
The purification was controlled by Waters Fractionlynx software h monitoring at 210-400nm and
triggered a threshold collection value at 260nm unless othen/vise started. Collected fractions were
analysed by SFC (Waters/Thar SFC systems with Waters SQD). The fractions that contained the
desired product were trated by vacuum centrifugation.
From the information provided someone skilled in the art could purify the compounds described herein
by ative LC-MS.
Synthetic Methods
By following methods r and/or analogous to general procedures below, the compounds set out
below were prepared.
The following synthetic procedures are provided for illustration of the s used; for a given
preparation or step the precursor used may not arily derive from the individual batch
synthesised according to the step in the ption given.
Where a compound is described as a mixture of two diastereoisomers / s, the configuration of
the centre is not ied and is represented by straight lines.
As understood by a person skilled in the art, compounds synthesised using the protocols as indicated
may exist as a solvate e.g. hydrate, and/or contain residual solvent or minor ties. nds
isolated as a salt form, may be integer stoichiometric i.e. mono- or di-salts, or of intermediate
stoichiometry.
Some ofthe compounds below are isolated as the salt, for example depending on the acid used in the
purification method. Some compounds are isolated as the free base.
Compounds containing a single stereocentre (R-configuration) at the 3-position are typically isolated
as a single isomer using preparative chiral HPLC (as described in l methods); at (or towards)
the final stage of the synthetic sequence. In these cases the stereochemistry at the 3-position is
designated in accordance with IUPAC, using ‘hashed’ or ‘solid’ wedged lines. Unless stated othen/vise,
a straight line at a stereocentre indicates the compound exists as a mixture of both isomers.
An e (3R)(4-chloropheny|)[(4-chIorophenyl)methyI]{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyI)-2,3-dihydro—1H-isoindoIone is
shown in Figure A.
Purification by
chiral HPLC
racemic on product Example e omer with R—configuration)
Figure A: Example showing purification of 3R-isomer by chiral HPLC
Compounds containing a second stereocentre (e.g. adjacent to the tion) are typically isolated as
a single isomer by preparative achiral and/or chiral HPLC. In these cases, the stereochemistry at the 3
position is ated in the usual fashion, using ‘hashed’ or ‘solid’ wedged lines. An asterisk (*) at
the second stereocentre indicates one (or both) of the diasteroisomers associated with this position
was/were isolated separately. For example, the 2 isomers of (3R)[(4-chloro
methanesulfonylpheny|)methyI](4-chIorophenyI)fluoro[1-hydroxy(1-methyI-1H-pyrazoI
yl)ethyI][(1-hydroxycyclopropyl)methoxy]-2,3-dihydro—1H-isoindoIone were ted by
preparative achiral and/or chiral HPLC to give two separate Examples (Figure B).
Note: Depending on the specific substitution pattern, the numbering system in some analogues may
differ, according to the formal convention of nomenclature.
R, er ,
isomer
Figure B: Asterisk ( * ) means the two isomers were separated and isolated to give the two
diasteroisomeric examples (Example 75 and 76)
In other cases, isomers were separated at an intermediate stage in the synthesis and only one isomer
progressed to the final Example. The relevant isomers can be characterised by either optical rotation
of linearly polarized light and/or or relative retention time on a chiral HPLC coloumn. In these cases,
an asterisk (* ) indicates that the compound was isolated as a single isomer. This is illustrated by
e 80 (Figure C)
0 l
Steps F Preparative
chiral HPLC
—> BOCN O
O 4»
OH separation
OH 0
(1) (2) [a]D20 = '27.8 (C (3)
1.8, MeOH).
'slower' g enatiomer
by chiral HPLC
l j
0 Preparative
,s‘ chiral HPLC
N 802Me
: separation
(5) Cl
Mixture of 2 isomers
Single dlatereOIsmer derIved. . . .
at 3-position
from single enantiomer (3)
Figure C: Synthesis of Example 80, (3R)—2-[(4-ch|oromethanesulfonylphenyl)methyl](4-
chlorophenyl)fluoro[1-hydroxy(1-methylpiperidinyl)ethyl]—3-{[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone. Example is derived from the
levorotary enantiomer of compound (3), followed by a preparative chiral HPLC at the final stage.
The l isomers may be characterised by their optical ty (i.e. as + and — isomers, or d and I
isomers). The centre can also assigned as “R or 8” according to the nomenclature developed by
Cahn, lngold and Prelog, see Advanced Organic Chemistry by Jerry March, 41h Edition, John Wiley &
Sons, New York, 1992, pages 4, and see also Cahn, lngold & Prelog, Angew. Chem. Int. Ed.
Engl., 1966, 5, 385-415.
Optical isomers can be separated by a number of techniques including chiral tography
(chromatography on a chiral support) and such techniques are well known to the person skilled in the
art.
As an alternative to chiral chromatography, optical isomers of basic compounds can be separated by
forming diastereoisomeric salts with chiral acids such as (+)-tartaric acid, roglutamic acid, (-)-ditoluoyl-L-tartaric
acid, (+)-mandelic acid, lic acid, and (-)-camphorsulfonic acid, separating the
diastereoisomeric salts by preferential crystallisation, and then dissociating the salts to give the
individual enantiomer of the free base. Likewise, optical iomers of acidic compounds can be
separated by forming diastereoisomeric salts with chiral amines such as Brucine, Cinchonidine,
quinine etc.
2016/053042
Additionally enantiomeric separation can be achieved by covalently linking a enantiomerically pure
chiral auxiliary onto the compound and then performing diastereisomer separation using conventional
methods such as chromatography. This is then followed by cleavage of the aforementioned covalent
linkage to generate the appropriate enantiomerically pure product. Examples could e making
menthol esters of an acidic compound.
Where compounds of the formula (I) exist as two or more optical isomeric forms, one enantiomer in a
pair of enantiomers may exhibit ages over the other enantiomer, for example, in terms of
biological activity. Thus, in certain circumstances, it may be desirable to use as a therapeutic agent
only one of a pair of enantiomers, or only one of a plurality of diastereoisomers.
Accordingly, the invention provides compositions containing a nd ofthe formula (I) having one
or more chiral centres, wherein at least 55% (e.g. at least 60%, 65%, 70%, 75%, 80%, 85%, 90% or
95%) of the compound of the formula (I) is present as a single optical isomer (e.g. enantiomer or
diastereoisomer). In one general embodiment, 99% or more (e.g. substantially all) of the total amount
of the compound of the formula (I) may be present as a single optical isomer (e.g. enantiomer or
diastereoisomer).
Compounds encompassing double bonds can have an E (entgegen) or Z (zusammen)
stereochemistry at said double bond. Substituents on bivalent cyclic or (partially) saturated radicals
may have either the cis- or trans-configuration. The terms cis and trans when used herein are in
accordance with Chemical Abstracts nomenclature (J. Org. Chem. 1970, 35 (9), 2849-2867), and refer
to the position ofthe substituents on a ring moiety.
Of special interest are those compounds of a (I) which are stereochemically pure. When a
compound of formula (I) is for instance specified as R, this means that the compound is substantially
free of the S isomer. If a compound of a (I) is for instance specified as E, this means that the
compound is substantially free of the Z . The terms cis, trans, R, S, E and Z are well known to a
person d in the art.
ES 1-137
Preparation 1. 4-Fluorohydroxybenzaldehyde
To a mixture of 3-fluorophenol (415 mg, 3.70 mmol) and anhydrous MgClz powder (1.06 g, 11.1 mmol)
in anhydrous acetonitrile (20 mL) was added anhydrous triethylamine (1.94 mL) and
rmaldehyde (811 mg, 27.0 mmol). The mixture was heated to reflux for 4.25 h, during which
time there was a colour change from white to pink to yellow. The reaction e was cooled to room
ature and 5% aqueous HCI was added (20 mL). The product was extracted with EtOAc (2 x 50
mL) and the combined organic extracts washed with H20 (3 x 50 mL), brine (50 mL), dried over
anhydrous M9804 and the solvent removed in vacuo. FCC [dichloromethane-methanol
WO 55860
(100:0)—>(97:3)] of the crude residue afforded the title Preparation 1 (367 mg, 71%) as a white solid;
Rf 0.83 (10% MeOH:CHZC|2); 1H NMR (500 MHz, CDCI3) 6 11.35 (1H, d, J = 1.6 Hz, CH0), 9.82 (1H,
s), 7.55 (1H, dd, J = 6.3 and 8.6 Hz), 6.71 (1H, dt, J = 2.4 and 8.3 Hz) and 6.66 (1H, dd, J = 2.4 and
.4 Hz), 19F NMR (470.7 MHz, CDCI3) 6 -97.53 (m).
Preparation 2: N'-(5’-Bromo-2’-hydroxybenzylidene)—4-chlorobenzohydrazide —
By following a similar procedure to Preparation 3, 5-bromohydroxybenzaldehyde (10 g, 49.7 mmol)
and 4-chlorobenzhydrazide (8.5 g, 49.7 mmol) gave Preparation 2 as an off white solid which was
used in the next step without further purification (16.5g, 94%), 6 max/cm1 1014, 1266, 1354, 1476,
1642, 3069, 3218; 6H(500 MHz; DMSO) 6.91 (1H, d, J = 8.6, 3’-H), 7.44 (1H, dd, J = 2.3, 8.7, 4’-H),
7.61-7.67 (2H, m, Ar—H), 7.81 (1H, d, J = 2.3, 6’-H), 7.94-8.01 (2H, m, Ar—H), 8.62 (1H, s, 1’-CH), 11.2
(1H, br. 5, NH), 12.24 (1H, br. s, OH).
Preparation 3: (E)-N'-(5-Bromofluorohydroxybenzylidene)chlorobenzohydrazide
To a solution of 5-bromofluoro-2—hydroxybenzaldehyde (1.06 g, 4.83 mmol) in acetic acid (23 mL)
was added 4-chlorobenzhydrazide (824 mg, 4.83 mmol) at room temperature and the resulting mixture
stirred for 15 min. The suspension was poured onto water (20 mL) at 0 °C and the resulting
precipitate collected by filtration. The solid was washed with water (3 x 20 mL), then petrol (3 x 20 mL)
and the product dried overnight in the vacuum oven to afford (E)-N'-(5-bromofluoro
hydroxybenzylidene)chlorobenzohydrazide ation 3 (1.69 g, 94%) as a pale yellow solid,
which was used without further purification; Rf 0.84 (10% H2C|2); tOH)/nm 237.8,
292.6, 303.6 and 333.0; IR (cm'1) 1098, 1158, 1242, 1463,1521, 1591, 1660, 2360 and 3261; mp 245
°C (decomp); 1H NMR (500 MHz, DMSO) 6 12.39 (1H, br.s), 11.64 (1H, br.s), 8.63 (1H, s, HC=N),
7.98 (1H, d, J = 8.5 Hz, 2 xArH), 7.69 (1H, s, ArH), 7.65 (2H, d, J = 8.5 Hz, 2 xArH) and 7.60 (1 H, dd,
J = 2.2 and 10.4 Hz, ArH). LRMS (ESI+) m/z 371.2 [M]+.
Preparation 4: 5-Bromo(4-chlorobenzoyl)benzaldehyde
WO 55860
By following a similar procedure to preparation 5, N'-(5’-bromo-2’-hydroxybenzylidene)
chlorobenzohydrazide (Preparation 2) (16.53 g, 46.7 mmol), Pb(OAc)4 (20.7 g, 46.7 mmol) and THF
(492 mL). ed on silica gel eluting with 20% —> 85% EtOAc/Hexane to give the Preparation 4 as
an orange solid (13.41, 88%). 6 max/cm1 764, 927, 1189, 1277, 1585, 1663, 1698, 2354, 2840, 3086;
6H(500 MHz; CDCI3) 7.37 (1H, d, J = 8.2, 3-H), 7.43-7.46 (1H, m, Ar—H), 7.70-7.73 (2H, m, Ar—H), 7.82
(1H, dd, J = 2.0, 7.9, 4-H), 8.14 (1H, d, J = 2.0, 6-H). m/z (ESI+) 323 (M+80%) 325 (100%).
Preparation 5: 5-Bromo(4-chIorobenzoyl)f|uorobenzaldehyd
F 0
Br CH0 Cl
To a suspension of (E)-N'—(5-bromofluorohydroxybenzylidene)chlorobenzohydrazide
(Preparation 3) (1.67 g, 4.49 mmol) in THF (45 mL) was added Pb(OAc)4 (1.99 g, 4.49 mmol)
portionwise. The resulting orange solution was stirred at room ature for 2 h and then filtered
h Celite®, eluting with EtOAc (30 mL). The organics were washed with saturated aqueous
NaHC03 (50 mL), brine (50 mL) dried over anhydrous NaZSO4, and the solvent d in vacuo.
FCC [petrol-ethyl acetate (100:0)—>(95:5)] of the crude residue afforded 5-Bromo(4-chlorobenzoyl)-
3-fluorobenzaldehyde Preparation 5 (1.29 g, 84%) as a yellow solid; Rf 0.87 (40% EtOAc:Petrol);
Amax(EtOH)inm 201.8 and 258.4; IR (cm'1) 1095, 1249, 1271, 1586, 1649, 1710, 2920 and 3072; mp
120.6-122.0 °C; 1H NMR (500 MHz, DMSO) 6 9.87 (1H, s, CH0), 8.27 (1H, d, J = 1.6 Hz, HCCBr),
8.15 (1H, dd, J =1.6 and 8.8 Hz, HCCF) 7.75 (2H, d, J = 8.5 Hz, 2 xAr) and 7.59 (2H, d, J = 8.5 Hz, 2
xArH). LRMS (ESI+) m/z 341.2 [M]+.
Preparation 6: 5-Bromo(4-chlorobenzoyl)benzoic acid
Following procedures similar to thise described in Preparation 7; 5-bromo-2—(4-
chlorobenzoyl)benzaldehyde (Preparation 4) (7.4 g, 22.9 mmol), sodium chlorite (2.9 g, 29.7 mmol),
ic acid (2.77 g, 30.6 mmol), acetonitrile (213 mL) and water (72 mL) gave Preparation 6 as a
biege solid and was used in the next step without further purification (7.4g, 95%). 6 max/cm'1 1014,
1090, 1252,1270, 1288, 1305, 1423, 1481, 1582, 1671, 2551, 2658, 2800; 6H(500 MHz; DMSO) 7.44
(1H, d, J = 8.0, 4-H), .60 (2H, m, Ar-H), 7.63-7.67 (2H, m, Ar—H), 7.96 (1H, dd, J = 2.0, 8.0, 4-H),
8.11 (1H, d, J = 2.0, 6-H), 13.64 (1H, br s, COOH). m/z (ES|-) 337(75%) 339 (M+ 100%) 341 (25%).
ation 7: 5-Bromo(4-chIorobenzoyl)f|uorobenzoic acid
F 0
I I
Br CI
COOH
To a solution of 5-Bromo(4-chIorobenzoyl)fluorobenzaldehyde (Preparation 5) (1.27 g, 3.71
mmol) in acetonitrile (48 mL) was added a solution of sodium te (436 mg, 4.82 mmol) in water
(5.4 mL), followed by a solution of sulfamic acid (468 mg, 4.82 mmol) in water (5.4 mL). The resulting
yellow solution was stirred at room temperature for 3 h and then the solvent removed in vacuo. The
resulting yellow solid was dissolved in ethyl acetate (30 mL) and washed with water (30 mL), brine (30
mL), dried over anhydrous NaZSO4 and the solvent removed to give 5-bromo(4-chlorobenzoyl)
fluorobenzoic acid Preparation 7 (1.33 g, 100%) as a pale yellow solid, which was used without any
further purification; Rf 0.23 (10% MeOH:CH2C|2); tOH)/nm 207.0 and 258.0; IR (cm'1) 1087,
1269, 1397, 1590, 1678, 1711 and 3072; mp 155 °C (decomp.); 1H NMR (500 MHz, DMSO) 6 13.93
(1H, br. 5, COOH), 8.07 (1H, dd, J = 1.5 and 9.0 Hz, HCCF), 8.00 (1H, d, J = 1.5 Hz, HCCBr), 7.74
(2H, d, J = 8.5 Hz, 2 xAr) and 7.59 (2H, d, J = 8.5 Hz, 2 xArH). LRMS (ESl-) m/z 357.0 [M]'.
Preparation 8: 6-Bromo(4-chlorobenzyl)(4-chlorophenyl)hydroxyisoindolinone
The title compound was prepared from 5-bromo-2—(4-chlorobenzoyl)benzoic acid (7.4 g, 21.8 mmol)
using a procedure similar to that bed for Preparation 9. The product was obtained as a yellow
solid (6.78 g, 66%) m/z (ES|-) 462(100%) (M+H")+
Preparation 9: 6-Bromo(4-chlorobenzyl)(4-chlorophenyl)fluorohydroxyisoindolin
WO 55860
To a solution of 5-bromo(4-chlorobenzoyl)fluorobenzoic acid (Preparation 7) (509 mg, 1.42
mmol) in anhydrous THF (7.1 mL) was added thionyl de (0.21 mL, 2.85 mmol) and a catalytic
quantity of DMF (1 drop) and the yellow mixture stirred at room temperature for4 h, then concentrated
under reduced pressure. The residue was dissolve in anhydrous THF (7.1 mL) and 4-
chlorobenzylamine (0.19 mL, 1.56 mmol) and Hiinigs base (0.27 mL, 1.56 mmol) were added and the
mixture stirred at room temperature for 16 h. The reaction was diluted with EtOAc (20 mL) and the
solution washed with water (3 x 20 mL) and brine (20 mL), dried over ous MgSO4 and
concentrated in vacuo to give a dark orange oil. FCC [petrol-ethyl acetate (100:0)—>(95:5)—>(80:20)] of
the crude residue afforded 6-bromo-2—(4-chlorobenzyl)—3-(4-chlorophenyl)fluoro
yisoindolinone Preparation 9 (624 mg, 91%) as a pale yellow solid; LRIVIS (ES|-) m/z 480.1
[M-H]'.
Preparation 10: 6-Bromo(4-chlorobenzyI)(4-chlorophenyl)((1-(hydroxymethyl)
cyclopropyl)methoxy)isoindolinone .
To a on of 6-bromo(4-chlorobenzyl)(4-chlorophenyl)hydroxyisoindolinone
(Preparation 8) (1 g, 2.1 mmol) and 1,1-Bis(hydroxymethyl)cyclopropane (0.604 mL, 6.3 mmol) in
DOE (21 mL) was added lan, (76 mgs, 0.21 mmol) and the resulting mixture was heated to 95 CC for
3 hours. The reaction was cooled to room temperature before being washed with water, brine, and the
c phase dried (MgSO4), filtered and conc. in vacuo. Purification on silica gel (Biotage SP4)
eluting with 20% —> 95% EtOAc/pet gave Preparation 10 as a brown gum (921 mgs; 80%),. (500 MHz,
CDCI3) 0.10-0.19 (2H, m), 0.37-0.46 (2H, m), 1.53 (1H, t), 2.65 (1H, d), 2.81 (1H, d), 3.36 (1H, dd),
3.50 (1H, dd), 4.17 (1H, d), 4.50 (1H, d), 7.01 (1H, d), 7.08-7.22 (8H, m), 7.63 (1H,dd), 8.02 (1H, d).
Preparation 11 : 6-Bromo((1-(((tert-buty|dimethylsiIyl)oxy)methy|)cyclopropyl)methoxy)(4-
chlorobenzy|)(4-chloropheny|)isoindolinone
TBDMSO
$00.
Preparation 10 (0.8 g, 1.46 mmol), ole (0.370 g, 5.44 mmol), TBDMSCI (0.496 g, 3.29 mmol) in
THF (10 mL/0.7 mmol) was heated at 85 °C for 7 hours. After work-up, the crude material was purified
using chromatography on silica (Pet2EtOAc 1:0 to 4:1) to give the d product Preparation 11 as a
colorless oil (0.928 g, 1.403 mol) in 96 1H NMR (500 MHz, CDClg) 5 (ppm) -0.02 (s, 3H), 0.00 (s, 3H),
0.06-0.15 (m, 2H), 0.30-0.43 (m, 2H), 0.83 (s, 9H), 2.61 (d, 1H), 2.87 (d, 1H), 3.35 (d, 1H), 3.65 (d,
1H), 4.30 (d, 1H), 4.37 (d, 1H), 6.97 (d, 1H), 7.04 (d, 2H), 7.10 (d, 2H, J = 8.5 Hz, Ar—H), 7.12-7.20 (m,
4H, Ar—H), 7.60 (dd, 1H), 8.02 (d, 1H); MS(ES+) m/z 456.4 [M+H]+.
Preparation 12: 6-Bromo(4-chlorobenzyI)(4-chlorophenyl)fluoro((1-
(hydroxymethyl)cyclopropyl)methoxy)isoindolinone
To a solution of o(4-chlorobenzyl)(4-chlorophenyl)—4-fluorohydroxyisoindolinone
(Preparation 9) (603 mg, 1.25 mmol) in anhydrous THF (7 mL) was added thionyl chloride (0.18 mL,
2.50 mmol) and a tic quantity of DMF (2 drops) and the orange mixture stirred at room
temperature for 4 h, then concentrated under reduced pressure. The residue was ved in
anhydrous THF and then a solution of 1,1-bis(hydroxymethyl)cyclopropane (0.24 mL, 2.50 mmol) and
K2C03 (345 mg, 2.50 mmol) were added and the reaction mixture stirred at room ature for 18 h.
The reaction mixture was diluted with EtOAc (10 mL) and washed with water (3 x 20 mL), brine (20
mL), dried over NaZSO4, filtered and the solvent removed in vacuo. FCC [petrol-ethyl acetate
(100:0)—>(80:20)] of the crude residue afforded 6-bromo(4-chlorobenzyl)(4-chloropheny|)
fluoro((1-(hydroxymethyl)cyclopropyl)methoxy)isoindo|inone Preparation 12 (389 mg, 55%) as a
yellow gum; 1H NMR (500 MHz, DMSO) 6 0.13 (2H, m), 0.34 (2H, m), 2.81 (1H, d), 2.86 (1H, d), 3.28
(1H, dd), 3.35 (1H, dd), 4.28 (1H, d), 4.34 (1H, d), 7.05 (2H, d), 7.19 (2H, d, J = 8.5 Hz, 2 xArH), 7.25
(2H, d, J = 8.4 Hz, 2 x ArH), 7.30 (2H, d), 7.82 (1H, dd) and 7.89 (1H, dLRMS (ESI+) m/z 464.2 [M-
CsH902]+
Preparation 13: 2-(4-Ch|orobenzyl)(4-chlorophenyl)((1-(hydroxymethyl) cyclopropyl)
methoxy)(propenyl)isoindolinone
WO 55860
A suspension of isopropenylboronic acid ester (239 pL, 1.27 mmol), 6-bromo(4-chlorobenzyl)(4-
chlorophenyl)((1-(hydroxymethyl) cyclopropyl)methoxy)isoindolinone ration 10) (465 g,
0.85 mmol), NaOH (35.7 mgs, 0.89 mmol), and N,N-dicyclohexylmethyl amine (0.191 mL, 0.894
mmol) in THF (4.63 mL) was degassed for 10 minutes before the addition of Pd(dppf)C|2 (62 mgs,
0.09 mmol). The resulting mixture was heated to a rapid reflux for3 hours. The reaction was cooled to
room temperature, diluted with DCM and washed with HCl (1M), water, brine, dried (MgSO4), filtered
and conc. in vacuo. Purification on silica gel eluting with 30% Hexanes gave Preparation 13 as
a white foam (313 mgs; 73%).; m/z (ESI+) 508.4 (10%), 406.3, 408.3 (70%).
Preparation 14: 2-(4-Chlorobenzy|)(4-chlorophenyl)fluoro((1-
(hydroxymethyl)cyclopropyl)methoxy)(prop-1 -enyl)isoindolinone
6-bromo(4-chlorobenzyl)(4-chlo rophenyl)fluoro((1 -
(hydroxymethyl)cyclopropyl)methoxy)isoindo|inone (Preparation 12) (194 mg, 0.343 mmol) was
dissolved in anhydrous THF (4.6 mL) and powdered NaOH (14 mg, 0.343 mmol), DCMA (0.07 mL,
0.343 mmol) and iso-propenylboronic acid pinacol ester (0.1 mL, 0.514 mmol) were added
sequentially at room temperature under a N2 atmosphere. The solution was degassed with N2 for 20
min, then Pd(dppf)C|2-CHZC|2 (28 mg, 0.100 mmol) was added and the solution heated at reflux for 3.5
h. After cooling to room temperature, the on mixture was filtered through Celite®, rinsed with
EtOAc, then transferred to a separating funnel and washed with 1M aq. HCl (20 mL), brine (20 mL),
dried over NaZSO4, filtered and the solvent removed in vacuo. FCC [petrol-ethyl acetate
(100:0)—>(85:15) )—>(70:30)] of the crude residue, followed by preparative HPLC, ed 2—(4-
chlorobenzyl)(4-chlorophenyl)fluoro((1-(hydroxymethyl)cyclopropyl)methoxy)(propen
yl)isoindolinone Preparation 14 (150 mg, 83%) as a pale yellow gum; LRMS (ESI+) m/z 426.3 [M-
C5H902]+.
Preparation 15: 6-Acetyl(4-chlorobenzyl)(4-chIorophenyl)—3-((3’-(hydroxymethyl)
cyclopropyl) methoxy)isoindolinone
To a solution of 2-(4-chlorobenzyl)—3-(4-chlorophenyl)((1-(hydr0xymethy|) cyclopropyl) methoxy)
(propenyl)isoindolinone ration 13) (64 mgs, 0.125 mmol) in dioxane/HZO (3:1, 1.23 mL)
at room temperature was added 2,6-Lutidine (30 uL, 0.251 mmol), OsO4 (~1 mg, 0.0025mmol) and
NalO4 (106 mg, 0.5 mmol) and the resulting mixture stirred until TLC indicted the complete
consumption of the g material. The reaction was diluted with water and DCM and the organic
phase seperated. The aqueous layer was extracted DCM (x3) and the combined organic phases
washed with brine, dried (MgSO4) and conc. in vacu. cation on silica gel ge SP4) eluting
with 20% —> 85% Pet gave Preparation 15 as white foam (37mgs; 60%). HRMS (ESI+)
510.121 (MH+).
Preparation 16: 3-(4-chlorophenyl)((S)(4-chlorophenyl)ethy|)hydroxy(propen
yI)isoindoIinone
The title compound was prepared using a mixture of Preparations 25a and 25b, by following a
procedure similar to that described for Preparation 13. The crude material was purified using
chromatography on silica (Pet:EtOAc 1:0 to 2:1) to give the desired reoisomers as a beige solid
(2.255 g, 5.14 mmol).
Preparation 16a R. = 0.30 (Pet:EtOAc/9:1); MS(ES+) m/z 484.3 [M+H
Preparation 16b: R = 0.15 (Pet:EtOAc/9:1);
Preparation 17: (E)-N'-(5-acety|hydroxybenzylidene)chlorobenzohydrazide
The title compound was prepared from ylhydroxybenzaldehyde (500 mg, 3.1 mmol) using a
procedure similar to that described for Preparation 3. The product was obtained as a yellow coloured
solid (923 mg, 96. LRMS (ES+) m/z 317.2 [M+H
Preparation 18: 5-Acetyl(4-chlorobenzoyl)benzaldehyde
I I
0 O
The title compound was prepared from (E)-N'—(5-acetylhydroxybenzylidene)
chlorobenzohydrazide, (Preparation 17), (0.90 g, 2.8 mmol) and Pb(OAc)4 (2.5 g, 5.7 mmol) using a
procedure similar to that described for Preparation 5. LRMS (ES+) m/z 287.3 [M+H]?
Preparation 19: 5-Acetyl(4-chlorobenzoyl)benzoic acid
O O
The title compound was ed from 5-acetyl(4-chlorobenzoyl)benzaldehyde, Preparation 18,
(690 mg, 2.4 mmol) using a ure similar to that described for Preparation 7. The product was
obtained as an off-white coloured solid (753 mg, 100%). LRMS (ES‘) m/z 301.2 [M-H]';
Preparation 20: yl(4-chlorobenzyl)(4-chlorophenyl)hydroxyisoindolinone
HO O
O 0
To a solution of 5-acetyl(4-chlorobenzoy|)benzoic acid, Preparation 19, (2.0 g, 6.6 mmol) in THF (8
mL) was added thionyl chloride (0.96 mL, 13.2 mmol) and stirred at rt for 2 h before being
concentrated in vacuo. The residue was dissolved in THF (8 mL) and 4-chlorobenzylamine (0.89 mL,
7.3 mmol) and Hiinig’s base (1.3 mL, 7.3 mmol) were added and stirred at rt for 2 h before being
d with EtOAc (8 mL). Washed with water (8 mL), brine (8 mL), dried (MgSO4) and concentrated
in vacuo. Purification (SP4, silica, EtOAc/petrol, 40%) gave Preparation 20 as a yellow solid (1.52 g,
54%). 1H NMR (500 MHz, CDCI3) 6 2.61 (3H, s, CH3), 3.21 (1H, br s, OH), 4.18 (1H, d, J = 15.0 Hz,
NCHH), 4.68 (1H, d, J =15.0 Hz, NCHH), 7.14-7.26 (8H, m, H-Ar), 7.31 (1H, d, J = 8.0 Hz, H-4), 8.07
(1H, dd, J =1.6,8.0 Hz, H-5), 8.26 (1H, d, J = 1.6 Hz, H-7). LRMS (ES') mfz 424.2 [M-H]';
Preparation 21: 6-Acetyl(4-chlorobenzy|)(4-chloropheny|)(2-hydroxyethoxy)isoindolin
8 Cl
O 0
The title compound was prepared in a similar fashion to ation 10, but using 0.8 mol. eq of lnBrs
and 20 mol. eq of ethylene . 1H NMR (500 MHz, CDCI3) 6 (ppm) 1.48-1.55 (m, 1H,), 2.67 (s, 3H),
1.62 (s, 6H), 2.80-2.92 (m, 2H), .52 (m, 2H), 4.11 (d, 1H), 4.67 (d, 1H), 7.14-7.19 (m, 4H), 7.20-
7.27 (m), 8.14 (dd, 1H, ), 8.43-8.46 (m). HMS(ES+) m/z 486.3 [M+H]+;
Preparation 22: 6-Acetyl(3-brom0-2,2-bis(hydroxymethyl)propoxy)(4-chlorobenzyl)(4-
chIorophenyl)isoindolinone
6-Acetyl-isoindolinone derivative (Preparation 20) (0.3 g, 0.704 mmol) in THF (5 mL) under a dried
atmosphere of N2 was added SOCIZ (0.103 mL, 1.41 mmol). The mixture was stirred for 2 hours at
room temperature before to be concentrated in vacuo. To the residue were added 2-(bromomethyl)
(hydroxymethyl)-1,3-propanediol (0.42 g, 2.11 mmol) and anhydrous K2C03 (0.194 g, 1.41 mmol),
followed by THF (5 mL) and the reaction e was stirred overnight. After work-up, crude material
was purified using chromatography on silica (Pet:EtOAc 1:0 to 0:1) to give a yellow solid Preparation
22 (0.186 g, 0.306 mmol. 1H NMR (500 MHz, CDCI3) 6 (ppm) 2.67 (s, 3H, COCH3), 2.78 (d, 1H, J =
9.0 Hz, OCCHHC), 3.00 (d, 1H, J = 9.0 Hz, CCHHC), 3.45-3.65 (m, 6H, CCHzBr, C(CHZOH)2), 4.41 (d,
1H, J = 15.1 Hz, NCHH),, 4.45 (d, 1H, J =15.1 Hz, NCHH), 7.02-7.11 (m, 4H, Ar-H), 7.13 (d, 2H, J =
8.5 Hz, Ar-H), 7.18 (d, 2H, J = 8.7 Hz, Ar-H), 7.27 (d, 1H, J = 8.0 Hz, isoindolinone-H), 8.15 (dd, 1H, J
= 7.9, 1.6 Hz, isoindolinone-H), 8.45 (d, 1H, J = 1.2 Hz, olinone-H; MS(ES+) m/z 456.4 [M+H]+;
Preparation 23: 6-Acetyl(4-chlorobenzy|)(4-chlorophenyl)((3-(hydroxymethyl)oxetan
yl)methoxy)isoindolinone
Bromo-diol (Preparation 22) (0.215 g, 0.354 mmol) was ved in EtOH (10 mL). KOH (0.023 g,
0.407 mmol) was added and the mixture was heated to 90 0C and stirred for 6 hours. Once back to
room temperature, H20 (10 mL) was added followed by introduction of an aqueous 1M solution of HCL
untill pH ~ 2-3. The mixture was ted with EtOAc (3 x 20 mL) and the combined organic phases
were washed with brine, dried over MgSO4, ed and evaporated in vacuo. Crude material purified
using chromatography on silica (PetzEtOAc 1:0 to 1:3) to give the desired product as a white foamy
solid Preparation 23 (0.104 g, 0198 mmol) in 56 1H NMR (500 MHz, CDCI3) 6 (ppm) 1.73 (bs, 1H,
OH), 2.67 (s, 3H, ), 2.92 (d, 1H), 3.05 (d, 1H), 3.67-3.77 (m, 2H), 4.22 (d, 1H), 4.24-4.29 (m, 2H, ),
4.31 (d, 1H), 4.36 (d, 1H), 4.57 (d, 1H), 7.10 (d, 1H,), 7.12-7.18 (m, 4H, H), 7.19-7.24 (m, 3H), 8.15
(dd, 1H), 8.46 (d, 1H, .);; MS(ES+) m/z [M+H]+; 456.1624.
Preparation 24: (R)(4-Chlorophenyl)((S)(4-chloropheny|)ethyl)((2-
(hydroxymethyl)allyl)oxy)(propenyl)isoindolinone
3-(4—Chlorophenyl)((S)(4-chlorophenyl)ethyl)hydroxy(propenyl)isoindolinone,
Preparation 16, (250 mg, 0.57 mmol), 2-methylene-1,3-propandiol (0.23 mL, 2.85 mmol), lnBr3 (305
mg, 0.86 mmol) and DOE (5 mL). Purification (SP4, , EtOAc/petrol, 25%) gave Preparation 24
as a white glassy solid (88 mg, 30%). R = 0.64 (silica, EtOAc/petrol. 1H NMR (500 MHz, CDCI3) 6
1.85 (3H, d, J = 7.3 Hz, ic CH3), 2.17 (3H, s, pene CH3), 3.59 (1H, d, J = 12.2 Hz, -iso-
OCHH), 3.81 (1H, d, J =12.2 Hz, -iso-OCHH), 4.16-4.23 (2H, m, CHZOH), 4.40 (1H, q, J = 7.3 Hz, H-
benzylic), 5.17-5.18 (1H, m, H-isopropene), 5.21-5.24 (2H, m, side-chain alkene CH2), 5.44-5.45 (1H,
m, H-isopropene), 6.95-7.10 (9H, m, H-Ar), 7.59 (1H, dd, J = 1.7, 7.9 Hz, H-5), 7.92 (1H, d, J = 1.7 Hz,
H-7). LRMS (ES+) m/z 508.4 [M+H]+.
Preparation 25a and 25b: 6-Bromo(4-ch|oropheny|)((S)(4-chIorophenyl)ethyl)—3-
hydroxyisoindolinone
CI CI
25a 25b
Starting from 5-bromo(4-chlorobenzoy|)benzoic acid (Preparation 6) and (1 S)(4-
phenyl)ethanamine the title compounds, Preparation 25a and 25b were prepared using
procedures similarto those described for Preparation 9. Productes obtained as off white solids .
25a (8,8): IVIS (ES+) 477.3 [M+H]+. R. = 0.73 (1 :1 EtOAc/petrol);
25b (R,S): MS (ES+) 477.3 [M+H]+. : R. = 0.64 (1 :1 EtOAc/petrol
Preparations 26a and 26b: 6-Bromo(4-chlorophenyl)((S)(4-chlorophenyl)ethyl)((1-
ymethyl)cyclopropyl) methoxy)isoindo|inone
Cl Cl
26a 26b
Preparations 26a and 26b were prepared using procedures similar to those described for
Preparation 12.
26a (S,S):; MS (ES+) 460.2 [M-HOCH2(c-Pr)CHZO]+. ): Rf = 0.51 (2:3 EtOAc/petrol);
26b (R,S):; MS (ES+) 460.2 [M-HOCH2(c-Pr)CH20, Rf: 0.42 (2:3 petrol);
Preparation 27: (R)(4-Chloropheny|)((S)(4-ch|oropheny|)ethyl)((1-
(hydroxymethyl)cyc|opropyl)methoxy)(propenyl)isoindo|inone
Starting from intermediate 26b (500 mg, 0.89 mmol), Preparation 27 was prepared using similar
procedures to those described for Preparation 13. MS (ES+) 522.5 [M+H]+.
Preparation 28: (R)Acetyl(4-chlorophenyl)((S)(4-chlorophenyl)ethyl)((1-
(hydroxymethyl)cyc|opropyl)methoxy)isoindolinone
Starting from Preparation 27, Preparation 28 was prepared using similar procedures to those
bed for Preparation 15. MS (ES+) 524.5 [M+H]+.
Preparation 29: ((1-(((tert-Butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorophenyl)((S)(4-chlorophenyl)ethyl)(propenyl)isoindolinone
TBDMSO
V3.0
o :5
To a solution of (R)(4-chlorophenyl)-2—((S)(4-chlorophenyl)ethyl)((1-
(hydroxymethyl)cyclopropyl)methoxy)(propenyl)isoindolinone, Preparation 27 (2.20 g, 4.21
mmol) in THF (50 mL) was added TBDMSCI (1.27 g, 8.42 mmol) and imidazole (860 mg, 12.6 mmol)
and the e heated at 85 °C for 5 h. The reaction was cooled to RT, extracted into EtOAc (100
mL), washed with 0.3 M aqueous HCI (150 mL), water (150 mL), brine (150 mL), dried over MgSO4
and concentrated under vacuum. MPLC (95:5 petrol to EtOAc then 99:1 petrol/EtOAc) gave the title
compound, Preparation 29 as a colourless oil (1.83 g, 68%); MS (ES+) 422.3 [M-(TBDMSOCH2(c-
Pr)CHZO)]+.
Preparation 30: (3R)—3-((1-(((tert-Butyldimethylsi|yl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorophenyl)((S)(4-chlorophenyl)ethyl)(2-methyloxiranyl)isoindo|inone
At 0 °C, to a solution of (R)((1-(((tert-buty|dimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorophenyl)((S)(4-chlorophenyl)ethyI)(propenyl)isoindolinone, Preparation 29 (500
mg, 0.79 mmol) in DCM (43 mL) was added portionwise mCPBA (271 mg, 1.57 mmol) and the
resulting solution stirred at RT for 18 h. The on was quenched by addition of ted aqueous
NaHC03 (25 mL) and stirred at RT for 30 min. The c layer was ted, washed with brine
(40 mL), dried over MgSO4 and concentrated under vacuum. The crude product, Preparation 30 was
obtained as a colourless oil and carried fon/vard to the next step without purification (708 mg); MS
(ES+) 436.3 [M-(TBDMSOCH2(c-Pr)CHZO)]+.
Preparation 31: (3R)—3-((1-(((tert-Butyldimethylsi|yl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorophenyl)((S)(4-chlorophenyl)ethyl)(2-hydroxymethoxypropanyl)isoindolin
To a solution of sodium (249 mg, 10.8 mmol) in MeOH (1.5 mL) was added dropwise a solution of
(3R)—3-((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chlorophenyI)((S)(4—
chlorophenyl)ethyl)(2-methyloxiranyl)isoindolinone, Preparation 30 (708 mg, 1.08 mmol) in
MeOH (1.5 mL) and the ing solution stirred at RT for 18 h then at 65 °C for 5 h. The on was
cooled to RT, NaOMe (292 mg, 5.4 mmol) was added and the mixture heated at 65 °C for 2h then
cooled to RT. The reaction was quenched by addition ofwater (100 mL), neutralised with aqueous 1.0
M HCI solution, extracted into EtOAc (2 x 100 mL), washed with brine (100 mL), dried over M9804
and concentrated under vacuum. IVIPLC (3:2 petrol/EtOAc) gave the title compound, Preparation 31
as a less oil as a diastereoisomeric mixture (151 mg, 20; MS (ES+) 468.3 [M-(TBDMSOCH2(c-
Pr)CHZO)]+.
ation 32: 3-((1-(((tert-Butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorobenzyl)(4-chlorophenyI)(propenyl)isoindo|inone
TBDMSO
PE C.
To a solution of 2-(4-chlorobenzyI)(4-chlorophenyl)((1-(hydroxymethyl)cyclopropyl)methoxy)
(propenyl)isoindolinone, Preparation 13 (2.73 g, 5.37 mmol) in THF (68 mL) was added
TBDMSCI (1.62 g, 10.7 mmol) and imidazole (1.10 g, 16.1 mmol) and the resulting suspension heated
at 85 °C for 5 h then cooled to RT. The mixture was diluted with EtOAc (150 mL), washed with
aqueous 0.3 M HCI (150 mL), water (150 mL), brine (150 mL) and dried over MgSO4. Purification by
MPLC (100% petrol to 95:5 petrol/EtOAc) gave the title compound, Preparation 32 as a colourless oil
(1.12 g, 34%); Amax (EtOH/nm) 214; IR (cm'1) 2928, 2855, 1704 (C=O), 1433; 1H NMR (500 MHz,
CDCI3) 6 -0.11 (3H, s, SiCHg), -0.09 (3H, s, SiCHs), 0.00-0.03 (2H, m, 2 x c-PrH), 0.25-0.27 (2H, m, 2
x , 0.73 (9H, s, 3)3), 2.08 (3H, m, CH3), 2.52 (1H, d, J = 9.2 Hz, CHH’), 2.80 (1H, d, J =
9.2 Hz, CHH’), 3.27 (1H, d, J =10.3 Hz, C’HH'), 3.56 (1H, d, J = 10.3 Hz, C’HH’), 4.22 (1H, d, J = 14.7
Hz, NCHH’), 4.30 (1H, d, J = 14.7 Hz, NCHH’), 5.08 (1H, m, alkene-CH), 5.35 (1H, s, alkene-CH’),
6.94 (1H, d, J = 8.0 Hz, ArH), 6.95-7.01 (4H, m, 4 x ArH), 7.05-7.09 (4H, m, 4 x ArH), 7.48 (1H, dd, J =
1.7 and 8.0 Hz, ArH), 7.86 (1H, d, J =1.7 Hz, ArH); 13’C (125 MHz, CDCI3) 6 -5.2,-5.3,7.9,8.1, 18.3,
12.9, 25.9 , 42.2, 65.9, 66.1
, 94.3, 114.3, 120.4, 122.8, 128.0, 128.2, 128.4, 130.1, 130.5, 131.6,
133.0, 134.3, 136.0, 137.4, 142.1, 168.3; MS (ES+) 408.3 [M-(TBDMSOCH2(c-Pr)CHZO)]+.
Preparation 33: 3-((1-(((tert-Butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chIorobenzy|)(4-chlorophenyI)(2-methyloxiranyl)isoindolinone
TBDMSO
At 0 °C, to a solution of 3-((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorobenzyl)(4-chlorophenyl)(propenyl)isoindolinone, Preparation 32 (640 mg, 1.03
mmol) in DCM (50 mL) was added mCPBA (355 mg, 2.05 mmol) and the resulting solution stirred at
RT for 18 h. The reaction was quenched by addition of saturated aqueous NaHC03 (50 mL) and
stirred vigorously for 4 h. The organic layer was separated, washed with brine (50 mL), dried over
MgSO4 and trated under vacuum. The crude product, Preparation 33 was carried fonNard to
the next step t purification (650 mg). MS (ES+) 422.3 [M-(TBDMSOCH2(c-Pr)CHZO)]+.
Preparation 34: 3-((1-(((tert-Butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorobenzyl)(4-chlorophenyl)—6-(1-(dimethylamino)hydroxypropan-Z-yl)isoindolinone
TBDMSO
In a sealed microwave vial, to a solution of 3-((1-(((tertbutyldimethylsilyl
)oxy)methyl)cyclopropyl)methoxy)(4-chlorobenzyl)(4-chlorophenyl)—6-(2-
methyloxiranyl)isoindolinone, Preparation 33 (1.08 g, 1.69 mmol) in lVleOH (3.04 mL) was added
dimethylamine (8.45 mL, 16.9 mmol, 2.0 M in MeOH) and the resulting solution heated at 60 °C for4 h
then cooled to RT. The reaction was diluted with water (50 mL), ted into EtOAc (2 x 50 mL),
washed with brine (100 mL), dried over MgSO4 and concentrated under vacuum. Purified by Biotage
using 0-30% MeOH in EtOAc as the eluent gave the title compound as a colourless oil (407 mg,
%).1H NMR (500 MHz, CDCIS) -0.02 (3H, m), 0.00 (3H, s), 0.03-0.12 (2H, m), 0.34-0.35 (2H, m),
0.83-0.84 (9H, m), 1.51 (3H, s), 2.19 (6H, s), .64 (1H, m), 2.76-2.83 (3H, m), .39 (1H, m),
3.63-3.67 (1H, m), 4.30-4.40 (2H, m), 7.05-7.10 (5H, m), 7.15 (4H, s), 7.68-7.71 (1H, m), 7.91-7.92
(1H, m).
Preparation 35: (S)-Ethy| 3-amino(4-chlorophenyl)propanoate hydrochloride
HCI H2N
At 0 °C, to a solution of s-beta-(p-ch|orophenyl)alanine (1.00 g, 5.0 mmol) in EtOH (10 mL) was added
dropwise SOCIZ and the resulting solution heated at 78 °C for 1.5 h. The solution was cooled to RT
and concentrated under vacuum to give the title compound, Preparation 35 as a white solid (1.31 g,
99%);;1H NMR (500 MHz, DMSO) 6 1.08 (3H, t, J = 7.1 Hz, CH3), 3.02 (1H, dd, J = 9.5 and 16.1 Hz,
CHH’C=O), 3.24 (1H, dd, J = 5.4 and 16.1 Hz, O), 3.96-4.01 (2H, m, OCHZ), 4.59 (1H, dd, J =
.4 and 9.5 Hz, NHZCH), 7.49 (2H, d, J = 8.5 Hz, 2 x ArH), 7.62 (2H, d, J = 8.5 Hz, 2 x ArH), 8.91 (3H,
s br,)
Preparation 36; (3S)-Ethyl 3-(5-bromo(4-chlorophenyl)hydroxyoxoisoindolinyl)(4-
chlorophenyl)propanoate
HO O
O COZEt
Starting from (S)-Ethyl 3-amino(4-chlorophenyl)propanoate hydrochloride, preparation 35 was
prepared using similar procedures to those described for Preparation 9. Product was obtained as a
diastereoisomeric e. MS (ES+) 546.2 [M-H]'.
Preparation 37a and 37b: (3S)-Ethy| romo(4-chlorophenyl)((1-
(hydroxymethyl)cyc|opropyl)methoxy)oxoisoindolinyl)(4-chlorophenyl)propanoate
CI CI
37a 37b
Starting from 3-(5-bromo(4—chlorophenyI)hydroxyoxoisoindo|inyl)(4-
chlorophenyl)propanoate ation 37a and 37b were prepared using similar ures to those
described for Preparation 12. The two products were isolated by SiOz chromatography.
37a (8,8): Rf = 0.67 (1 :1 EtOAc/petrol);; MS (ES+) 532.2 [M-HOCH2(c-Pr)CHZO]+.
37b (S,R): Rf: 0.52 (1 :1 EtOAc/petrol); MS (ES+) 530.3 [M-HOCH2(c-Pr)CH20]+.
Preparation 38: (S)—Ethy| 3-(4-chlorophenyl)((R)(4-chlorophenyl)((1-
(hydroxymethyl)cyclopropyl)methoxy)oxo(propenyl)isoindolin-Z-yl)propanoate
ng from preparation 37b, Preparation 38 was prepared using similar procedures to those
described for ation 13. MS (ES+) 492.4 [M-HOCH2(c-Pr)CHZO]+.
Preparation 39: (S)-Ethyl 3-((R)acetyl(4-ch|orophenyl)((1-
(hydroxymethyl)cyc|opropyl)methoxy)oxoisoindolinyl)(4-chlorophenyl)propanoate
Starting from Preparation 38, Preparation 39 was prepared using similar procedures to those
described for Preparation 15. MS (ES+) 494.3 [M-HOCH2(c-Pr)CH20]+.
Preparation 40: (S)—3-((R)Acetyl(4-ch|orophenyl)((1-
(hydroxymethyl)cyclopropyl)methoxy)oxoisoindolinyl)(4-chlorophenyl)propanoic acid
To a solution of (S)-ethyl 3-((R)acetyl(4-chloropheny|)((1-
(hydroxymethy|)cyc|opropyl)methoxy)oxoisoindolin-2—yl)(4-ch|oropheny|)propanoate,
Preparation 39 (130 mg, 0.22 mmol) in THF/water (1.9 mL/3.0 mL) was added LiOH.HZO ( 183 mg,
4.36 mmol) and the resulting yellow solution heated at 60 °C for 3 h then cooled to RT. The solution
was acidified to pH 5 with aqueous 1.0 M HCI, ted with EtOAc (3 X 25 mL), washed with brine
(50 mL), dried over MgSO4 and concentrated under vacuum. MPLC (1 :1 petrol/EtOAc(0.1% AcOH) to
100% EtOAc(0.1% AcOH)) gave the title compound, Preparation 40 as a white solid (85 mg, 68%);
MS (ES+) 566.2 [M-H]'.
Preparation 41: yl(4-chlorobenzyl)—3-(4-chlorophenyl)(3-hydroxy
methylbutoxy)isoindolinone
Starting from 6-acetyl(4-chlorobenzyl)(4-chloropheny|)hydroxyisoindolinone, Preparation
41 was prepared using similar procedure to those described in ation 12.
Preparation 42: 4-[(Triisopropylsilanyl)-ethyny|]-benzylamine
NHBopc
2 step 1 NHBOC
—> —> —>
Cl | | I I
T'PS
TIPS
Step 1: (4-Chloro-benzyI)-carbamic acid tert-butyl ester
4-Chlorobenzylamine (10.5 mL, 86 mmol) was added to a mixture of Amberlyst 15 resin (1.8 g) and ditert-butyl
dicarbonate (18.5 g, 84.7 mmol) and the reaction mixture was stirred at room temperature for
3 hours. The crude mixture was diluted with DCM (200 mL) and the catalyst was removed by filtration.
The t was removed in vacuo to give the desired product as a white solid (18.9 9). MS: [M —
C4H9]+ = 186. 1H NMR (400 MHz, CDCI3): 7.38-7.30 (2H, m), 7.23 (2H, d), 4.87 (1H, s), 4.29 (2H, s),
1.48 (9H, s).
Step 2: {4-[(TriisopropyIsilany|)-ethynyI]-benzyI}-carbamic acid tert-butyl ester
Ethynyl-triisopropyl-silane (13.4 mL, 94.1 mmol) was added to a suspension of (4-chloro-benzyl)—
carbamic acid tert-butyl ester (18.9 g, 78.4 mmol), PdC|2(CH3CN)2 (202 mg, 0.78 mmol), XPhos (1.1 g,
2.3 mmol) and Cs2C03 (53.6 g, 164.6 mmol) in MeCN (170 mL) and the reaction mixture was stirred
under N2 at 110°C for 20 hours. The reaction was then cooled to room temperature, quenched with
water (300 mL) and extracted with EtOAc (2x300 mL). The organic phases were collected, dried over
NaZSO4. The reaction was then cooled to room temperature, quenched with water (300 mL) and
extracted with EtOAc (2x300 mL). The organic phases were collected, dried over NaZSO4, filtered and
concentrated in vacuo. The crude material was columned ent 0-20% EtOAc in Petrol) to give
.7 g ofa yellow oil. 1H NMR (400 MHz, CDCI3): 7.45 (2H, d), 7.23 (2H, d), 4.83 (1H, s), 4.32 (2H,
s), 1.48 (9H, s), .11 (21H, m).
Step 3: 4-[(TriisopropylsiIanyl)-ethynyl]-benzylamine
TFA (25 mL, 326 mmol) was added to a solution of {4-[(Triisopropylsilanyl)-ethynyl]-benzyl}-carbamic
acid utyl ester (15.6 g, 40.3 mmol) in DCM (50 mL). The reaction was stirred for 16 hours at room
temperature and then quenched with water (30 mL) and 2N NaOH until the solution reached pH = 11.
The product was extracted with DCM (3x). The organic phases were collected, dried over NaZSO4,
filtered and concentrated in vacuo to give the desired product (12.5 g) as an orange oil. 1H NMR (400
MHz, CDCI3): 7.46 (2H, d), .24 (2H, m), 3.87 (2H, s), 1.94 (2H, s), 1.15 (21H, s).
Preparation 43: ro(methylthio)phenyl)methanamine
Cl CI
To a solution of 4-chloro(methylthio)benzonitrile (500 mg, 2.72 mmol) in dry THF (10 mL) was
added slowly -THF complex (1 M in THF, 13.6 mL, 13.6 mmol) at 0 °C before ing for 1 h.
After cooling, 1 M HCl in MeOH (10 mL) was added slowly with ice cooling. The solvent was removed
by concentration in vacuo before water (0.61 mmol/mL to benzonitrile) was added, then washed by
EtZO (0.61 L to benzonitrile) before basifying with 2 M NaOH solution to pH 12. EtZO (0.61
L to benzonitrile) was added and the mixture was washed with water (3 x 0.61 mmol/mL to
benzonitrile) and brine (0.4 mol/mL to benzonitrile). The organic layer was dried (MgSO4) and
trated in vacuo to give the product as a yellow oil (400 mg, 78%). LCMS (ES|+) m/z = 171.1 [M-
NHJ’.
Preparation 44: (3R)Acetyl[(4-chIoromethanesulfonylphenyl)methyI](4-chIorophenyl)—
ro[(3R)-oxo|anyloxy]-2,3-dihydro-1H-isoindolone
Tm}:90“OC—l802Me
The title compound was prepared using a similar method to that described in Example 41, Step 1 and
2; using (3R)-hydroxy-tetrahydrofuran instead of cyclopropane-1 ,1-dimethanol. The 3(R) isomer was
isolated as the slower running fraction from Step 1, using preparative chiral HPLC. MS(ES+) m/z 630
[M+H]+.
Preparation 45: Ethyl (3S)[(1R)acetyl(4-chlorophenyl)fluorooxo[(3S)-oxolan
yloxy]-2,3-dihydro-1H-isoindolyl](4-chloropheny|)propanoate
The title compound was prepared in a similar way to Preparation 44, but using ethyl (3S)—3-amino
(4-ch|orophenyl)propanoate hloride (Preparation 35) instead of (4-chloro
(methylsulfonyl)phenyl)methanamine and (3S)-hydroxy-tetrahydrofuran d of (3R)-hydroxy-
tetrahydrofuran. ) m/z 598 [M-H]'
Preparation 46: 2-(4-Chlorobenzoyl)fluoro(1-hydroxymethy|—ethyl)benzoic acid
The title compound was prepared in a similar fashion to Example 73 and 74, Step 1; but using
acetone instead of 1-methyI-1H-pyrazolecarboxaldehyde. MS: [M + H]+ = 337.
Preparation 47: (2-(Aminomethyl)—5-chlorophenyl)dimethy|phosphine oxide
Step 1 Step 2
Cl Cl 0|
Step 1: 4-Chloro(dimethylphosphoryl)benzonitrile
To a round bottomed flask was added 4-chloroiodobenzonitrile (2.5 g, 9.50 mmol),
dimethylphosphine oxide (1.12 g, 14.30 mmol), Pd2(dba)3 (435 mg, 0.48 mmol), Xantphos (550 mg,
0.95 mmol) and the flask was flushed with N2. The solids were taken up in dioxane (25 mL),
triethylamine added (2.10 g, 20.9 mmol) and the reaction was stirred at room temperature for 2h. To
the reaction was added H20 (50 mL) and the aqueous was extracted with EtOAc (2 x 100 mL). The
combined organics were dried (MgSO4), filtered and concentrated in vacuo. The residue was d
on a 50 g SNAP column eluting with MeOH in DCM (0 to 10 %) to give the title compound (1.07 g at
80 % ). 1H NMR (400 MHz, CDCI3) 8.33 - 8.29 (1H, m), 7.76 - 7.73 (1H, m), 7.65 - 7.62 (1H, m),
1.96 (6H, d);
Step 2: (2-(Aminomethyl)ch|orophenyl)dimethylphosphine oxide
To a THF on (10 mL) of 4-chloro(dimethylphosphoryl)benzonitrile (960 mg, 4.50 mmol), under
N2, was added BH3.THF (23 mL, 23.00 mmol, 1M THF) and the reaction was stirred for 1h. The
reaction was quenched by the us addition of MeOH (10 mL). The solution was concentrated in
, re-dissolved in MeOH and loaded on to a 10g SCXII cartridge. The cartridge was sequentially
flushed with MeOH (3 column volumes) and then 2M NH3 MeOH (3 column volumes). The ammonia
wash was concentrated in vacuo. to give the title compound (770 mg). 1H NMR (400 MHz, CDCI3) 7.46
- 7.41 (3H, m), 4.14 (2H, s), 1.83 (6H, d).
Preparation 48: 2-(4-Chlorobenzoy|)f|uoro(tetrahydro-2H-pyrancarbonyl)benzoic acid
o OH O O 0
Steps 1 was performed using a procedure similarto that described in Examples 73 and 74; step 1,
but using tetrahydro-2H-pyrancarbaldehyde instead of yl-1H-pyrazolecarboxaldehyde.
Also, BuzMg was used in place of methylmagnseium chloride. MS: [M+H]+ = 393.
Step 2: 2-(4-Ch|orobenzoyl)—3-fluoro(tetrahydro-2H-pyrancarbonyl)benzoic acid
2-(4—Chlorobenzoyl)—3-fluoro(hydroxy(tetrahydro-2H-pyranyl)methyl)benzoic acid (17.4 g, 44.4
mmol) was stirred in DCM (400 mL) at RT then TEMPO (0.69 g, 4.44 mmol) and tetra-n-
butylammonium de (5.72 g, 17.8 mmol) were added followed by OXONE®, monopersulfate
compound (30 g, 97,7 mmol). The on was allowed to stir at RT for 18 h. TEMPO (0.69 g, 4.44
mmol) was added and the reaction was allowed to stir at RT for an additional 48 h. The solids were
removed by filtration and the filter cake was washed with DCM (2 x 100 mL). The combined filtrates
were concentrated under d pressure and the resulting residue dissolved in EtOAc (500 mL).
The combined organic portions were washed with 2M HCI aqueous solution (2 x 500 mL) and brine
(200 mL), dried (MgSO4), filtered and concentrated to afford the title nd as a pale yellow foam
(16 g, 92% yield). MS: [M-H]' = 389.
Preparation 49: 4-[(1R)Amino(propenyloxy)ethyl]benzonitrile
H N2
F H
OH FNNF OH F>KWN 0V HZN 0V
|| ||
N N | I II
N N Step 1:
N-[(1R)(4-Cyanophenyl)hydroxyethyl]-2,2,2-trifluoroacetamide
TFAA (3.6 mL, 25.2 mmol) was added to a solution of 4-[(1 R)aminohydroxyethyl]benzonitrile (5.0
g, 25.2 mmol) in DCM (100 mL) containing TEA (10.9 mL, 75,6 mmol) and the solution was stirred for
min at room temperature. The reaction was partitioned between DCM and 2N HCI. The aqueous
phase was extracted with DCM (2x) and EtOAc (2x). The organic phases were collected, dried over
MgSO4, filtered and concentrated in vacuo to give the desired product as a white solid (4.8 g, 74%
yield). LCMS: [M-H]'= 257.
Step 2: )(4-Cyanophenyl)—2-(propeny|oxy)ethyl]-2,2,2-trifluoroacetamide
NaH (60% disp. in oil, 1.5 g, 36.9 mmol) was added in portions to a solution of N-[(1R)(4-
cyanophenyl)hydroxyethyl]-2,2,2—trifluoroacetamide (4.76 g, 18.4 mmoL) in DMF (15 mL) at 0°C
under inert atmosphere and the resulting mixture was then stirred for 10 min at room temperature. 3-
iodopropene (1.7 mL, 18.4 mmol) was then added dropwise, the reaction was stirred for 10 min at
room temperature and then quenched with water. The product was extracted with EtOAc (3x), the
c phase was washed 3x with brine and the solvent was removed in vacuo. The crude material
was purified by flash tography on silica gel (gradient 0-30% EtOAc in petrol) to give the title
compound as a colourless liquid (3.5 g, 64% yield). LCMS: = 299.
Step 3: 4-[(1R)Amino(propenyloxy)ethyl]benzonitrile
2N NaOH (10 mL) was added to a solution of N-[(1R)(4-cyanophenyl)(propenyloxy)ethyl]-
trifluoroacetamide (3.5 g, 11.7 mmol) in MeOH (10 mL). The reaction was stirred for 16 hours at
room temperature after which time 4 mL of2N NaOH were added to the mixture and ng was
ued for further 2 hours at 45°C. The solvent was removed in vacuo and the residue was
partitioned between EtOAc and NaHCOs. The organic phase was dried over MgSO4, filtered and
concentrated in vacuo to give 2.3 g (96% yield) ofthe desired compound as a pale yellow oil. LCMS:
[M+H]+ = 203.
Preparation 50: 2-(4-Chlorobenzoy|)f|uoro(1-methyl-1H-imidazoIecarbonyl)benzoic acid
0 (i)MeMgC|;THF;0°C Mnoz
F (ii) nBuLi ; -78°C
\ \
O 0 (iii) EX <N o
\ \
-78°C to RT Step 2 O
Step1 Step 1:
2-(4-Chlorobenzoyl)fluoro(hydroxy(1-methyl-1H-imidazoIy|)methyl)benzoic acid
A 10 litre round bottomed flask fitted with an overhead stirrer with a large paddle was charged with 5-
bromo(4-chlorobenzoyl)fluorobenzoic acid 5 g; 0.5 mol) and ous THF (2610 mL)
added. This solution was cooled to -5°C<T<0°C and a solution of 3 M methyl magnesium chloride in
THF (183 mL; 0.55 mol) was added dropwise at such a rate that the internal ature remained
below 0°C. On complete addition the mixture was stirred at 0°C for 15 minutes and then cooled to -
78°C. A solution of2.5 M n-butyllithium in hexanes (259 mL; 0.647 mol) was added ise at such
a rate so that the internal temperature remained below -70°C. The reaction deepened in colour and
thickened considerably ending up with a sludge like consistency. On complete on the mixture
was stirred at -78°C for 30 minutes prior to the addition of a solution of 1-methyl-1H-imidazole
carbaldehyde (71.3 g; 0.648 mol) in anhydrous THF ( 500 mL) se at such a rate so that the
internal temperature remained below -70°C. On complete addition the mixture was stirred at -78°C for
minutes, the cooling bath removed and the e allowed to reach room temperature. The
mixture was quenched with 1 M HCI, the pH adjusted to 7 and the whole evaporated under reduced
pressure The residue was divided into 4 equal portions and each portion chromatographed on silica
gel (300g) eluting with 0 — 30% MeOH in DCM gradient to afford the title compound as a yellow solid
(151 g; 78%). MS [M + H]+= 389
Step 2: 2-(4-Chlorobenzoyl)fluoro(1-methyl-1H-imidazolecarbonyl)benzoic acid
To a stirred mixture of 2-(4-chlorobenzoyl)fluoro(hydroxy(1-methyl-1H-imidazol
yl)methyl)benzoic acid (24.6 g; 63.4 mmol) in 1,4-dioxane (600 mL) was added activated ese
dioxide (55 g; 634 mmol) and the e heated at 110°C for1 h. LCMS indicated complete reaction.
The reaction was cooled, filtered through celite and GFA filter paper, washed with MeOH. The filtrate
and washings were combined and evaporated under d pressure to afford a dark solid (24g). MS
[M + H]+ = 387.
Preparation 51: 3-((A||y|oxy)methy|)(aminomethy|)benzonitrile hydrochloride (4)
NH(Boc)2, 0Y0
Br K2003, O N
THF, DMF \n/
0 Br
KFsBV
RuPhos CsZCO3\,
Pd(OAc)2, Dioxane H20
.HCl 0 0
H N2
4N HCI T
OM Dioxane ‘—
O <
Step 1: tert-Butyl N-[(2-bromocyano-phenyl)methyl]-N-tert-butoxycarbonyl-carbamate (2)
o(bromomethyl)benzonitrile (1) (8 g, 29.1 mmol), was dissolved in THF (80 mL) and DMF
(80 mL) and the solution was stirred at room temperature under a nitrogen atmosphere. Di-tert-
butylimino carboxylate (9.48 g, 43.6 mmol) and potassium ate (6.0 g, 43.6 mmol) were added
and the reaction was heated at 100 °C overnight. The mixture was diluted with EtOAc (250 mL) and
water (250 mL). The organic phase was collected and the aqueous phase was ted with EtOAc
(250 mL). The combined organic extracts were washed with brine (120 mL), dried (MgSO4), filtered,
and evaporated to dryness. The crude product was purified by trituration with the minimal amount of
methanol (~20 mL) and the solid was collected via filtration to give the desired product (10.3 g, 86%)
as a white solid. 1H NMR (400 MHz, CDCI3): 7.84 (1H, d), 7.59 (1H, dd), 7.23 (1H, d), 4.88 (2H, s),
1.46 (18H, 3)
Step 2: tert-Butyl N-[[2-(Allyloxymethy|)cyano-phenyl]methyl]-N-tert-butoxycarbonyl-
carbamate (3)
tert-Butyl N-[(2-bromocyano-phenyl)methyl]-N-tert-butoxycarbonyl-carbamate (2) (5 g, 12.1 mmol),
RuPhos (0.569 g, 1.21 mmol), potassium [(allyloxy)methyl]trifluoroborate 2.6 g, 14.6 mmol) and
cesium ate (11.9 g, 36.1 mmoL) were dissolved in dioxane (90 mL) and water (10 mL). The
mixture was purged with nitrogen for 5 minutes and then palladium acetate (136 mg, 0.61 mmol) was
added. The mixture was purged with nitrogen for a r5 minutes and then heated at 100 °C for 24
hours. The mixture was concentrated under d pressure and the ing residue was
partitioned between EtOAc (150 mL) and water (150 mL). The organic layer was collected and the
aqueous phase was extracted with more EtOAc (150 mL). The combined organic ts were
passed through a hydrophobic frit, and evaporated under reduced pressure to give a crude product
WO 55860
which was purified by silica column chromatography (gradient elution 0 to 20% EtOAc in iso-Hex), to
give the pure product (3.0 g, 53%) as a yellow oil. 1H NMR (400 MHz, CDCI3):P 7.68 (1H, d), 7.59 -
7.56 (1H, m), 7.29 (1H, d), 5.99 - 5.89 (1H, m), 5.35 - 5.22 (2H, m), 4.86 (2H, s), 4.56 (2H, s), 4.07 -
4.04 (2H, m), 1.44 (18H, s), 0.95 - 0.82 (1H, m)
Step 3: 3-((Allyloxy)methyl)(aminomethyl)benzonitrile hydrochloride (4)
tert-Butyl N-[[2-(allyloxymethyl)cyano-phenyl]methyl]-N-tert-butoxycarbonyl-carbamate (3) (2.6 g,
6.46 mmol) was dissolved in DCM (83 mL) and 4N HCI in dioxane (28.6 mL) was added. The mixture
was stirred at room temperature for 4 hours. The solvents were removed under vacuum and any
remaining solvent traces were removed via co-evaporation from chloroform. The desired product (1.6
g) was isolated as a white solid and was deemed to be sufficiently pure for subsequent steps. 1H NMR
(400 MHz, DMSO): 8.51 - 8.51 (3H, m), 7.93 (1H, dd), 7.88 (1H, d), 7.71 (1H, d), 6.02 - 5.92 (1H, m),
.33 (1H, ddd), 5.22 (1H, dd), 4.65 (2H, s), 4.16 (2H, s), 4.06 (2H, d);
Preparation 52: (S)(4-chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyran
y|)propyl)benzoic acid
Step 1: 2-(4-Chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyrany|)propy|)benzoic
acid
To 50 mL of THF at —50°C under nitrogen atmosphere was added lzinc (62 mL, 1M solution in
s, 62.0 mmol) and ethyl lithium (36 mL, 1.72 M solution in dibutyl ether, 62.0 mmol). The
mixture was d at —50°C for 1 h and then 2-(4-chlorobenzoy|)fluoro(tetrahydro-2H-pyran
carbonyl)benzoic acid (Preparation 48, 9.7 g, 24.0 mmol) was added as a THF (100 mL) solution. The
mixture turned dark orange immediately and the al ature reached —22°C. The mixture was
stirred at —50°C for 20 min before being quenched by slow addition of 2N HCI (500 mL) (Caution).
After stirring for 1 h, the pH was adjusted to 1-2 with 2M HCI and the aqueous was extracted with ethyl
e (200 mL), washed with 2M HCI (75 mL), dried over magnesium sulfate, filtered and
concentrated. The crude product was purified by silica column chromatography (gradient elution 0 to
100% EtOAc in iso-hexane) to give the title compound (9.03 g, 90%) as a colourless foam. MS:
[M+H]+ = 421
Step 2: Methyl (S)(4-chlorobenzoyl)—3-fluoro(1-hydroxy(tetrahydro-2H-pyran
yl)propyl)benzoate
To a round bottom flask containing crude 2-(4-ch|orobenzoy|)f|uoro(1-hydroxy(tetrahydro-2H-
pyranyl)propyl)benzoic acid (6.32 g, 15 mmol), K2C03 (2.69 g, 19 mmol) and DMF (50 mL) was
added methyl iodide (0.934 mL, 16 mmol). The reaction was stirred for 1.5 h at room temperature,
after which point LCMS showed complete sion to the desired product. The mixture was
concentrated under reduced re and the residue dissolved in ethyl acetate (150 mL) and washed
with water (100 mL), then a 4% aqueous LiCl solution (2 x 100 mL). The organic layer was dried over
magnesium sulfate, filtered and concentrated to give a pale yellow foam. The enantiomers were
separated using chiral SFC to give the title compound (8.1 g) as a colourless solid.
Methyl (S)—2-(4-chlorobenzoyl)—3-f|uoro(1-hydroxy(tetrahydro-2H-pyran
yl)propyl)benzoate: Fast running isomer MS: [M+H]+ = 435. ° = -1.83 (c 1.0, MeOH).
Methyl (4-chlorobenzoyl)f|uoro(1-hydroxy(tetrahydro-2H-pyran
yl)propyl)benzoate: Slow running isomer MS: [M+H]+= 435. [or]:,20 = +1.48 (0 1.0, MeOH).
Step 3: (S)(4-Chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyran
yl)propyl)benzoic acid
Methyl (S)(4-chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyranyl)propyl)benzoate (8.2
g, 18.86 mmol) was dissolved in THF (250 mL), methanol (30 mL) and water (50 mL). Anhydrous
LiOH (2.26 g, 94.3 mmol) was added and the mixture was stirred at room temperature for 2 h. The
resultant solution was concentrated to approximately 60 mL volume, diluted with water (500 mL) and
washed with l ether (400 mL). The aqueous layer was then ied with 2N HCI and extracted
with DCM (3 x 200 mL). Combined extracts were dried (MgSO4) and evaporated to afford the title
nd (8.1 g, quant.)as a colourless foam. 1H NMR (400 MHz, CDCI3) 7.86 (1H, s), 7.71 (2H, d),
7.49 - 7.41 (3H, m), 4.05 (1H, dd), 3.98 - 3.93 (1H, m), 3.43 - 3.28 (2H, m), 1.97 - 1.89 (2H, m), 1.77 -
1.74 (1H, m), 1.52 - 1.40 (2H, m), 1.20 - 1.13 (1H, m), 0.75 (3H, dd), OH and COOH not observed.
MS: [M-H+]'= 419. [011020 = -2.3 (c 1.0, MeOH).
Preparation 52b: (R)(4-chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyran
yl)propyl)benzoic acid
The title compound was prepared in a similar fashion to Example 52, but using (R)-2—(4-
chlorobenzoyI)fluoro(1-hydroxy(tetrahydro-2H-pyranyl)propyl)benzoate in Step 3. MS: [M-
H+]'= 4191011020 = +1.8 «:10, MeOH).
Preparation 53: 2-(4-chlorobenzoyl)fluoro(4-fluorotetrahydro-2H-pyrancarbonyl)benzoic
acid
Step 1: Ethyl hlorobenzoy|)f|uoro(tetrahydro-2H-pyrancarbonyl)benzoate
To a stirred solution of 2-(4-chlorobenzoyl)fluoro(tetrahydro-2H-pyrancarbonyl)benzoic acid
(Preparation 48, 100 g, 258 mmol) in DMF (800 mL) was added potassium carbonate (54.8 g, 396
mmol) followed by iodoethane (26.8 mL, 334 mmol). The mixture was stirred for 18 h at RT and then
evaporated to dryness under d pressure. The e was triturated with water (1 L) and the
solid was collected by filtration, washing with more water (3 x 500 mL). The solid was dissolved in
DCM (750 mL), dried (MgSO4), filtered and evaporated under reduced re to give the title
compound (107 g, 99%) as a pale yellow solid. 1H NMR (400 MHz, CDCIS) 8.43 (1H, d), 7.90 (1H, dd),
7.75 - 7.72 (2H, m), 7.47 - 7.44 (2H, m), 4.21 (2H, q), 4.11 - 4.05 (2H, m), 3.63 - 3.48 (3H, m), 1.97 -
1.79 (4H, m), 1.13 (3H, dd).
Step 2: Ethyl 2-(4-chlorobenzoyl)—3-fluoro(4-fluorotetrahydro-2H-pyrancarbonyl)benzoate
A three neck flask was fitted with a nitrogen inlet, a thermometer, and a pressure equalising dropping
funnel. The flask was charged with ethyl hlorobenzoyl)fluoro(tetrahydro-2H-pyran
yl)benzoate (54.0 g, 129 mmol) and dry THF (560 mL). The mixture was then cooled to —78 °C
and LHMDS (170 mL. 170 mmol, 1M in THF) was added at a steady rate such that the internal
reaction temperature did not exceed —60 °C. The mixture was stirred at —78 °C for 20 min, and then a
solution of N-fluorobenzenesulfonimide (53.14 g, 169 mmol) in THF (560 mL) was added steadily
ensuring the internal temperature did not exceed —60 °C. The mixture was stirred at —78 °C for 20
min, and was then allowed to warm to room ature (~1 h). The reaction was quenched with
water (500 mL) and then extracted with ethyl acetate (3 x 250 mL). The combined organic extracts
were dried (MgSO4), filtered, and evaporated under reduced pressure to give a crude residue which
was purified by silica column tography (340 g cartridge, gradient elution 0 to 40% EtOAc in iso-
hexane) to give the title compound (46.2 g, 82%) as a colourless solid. MS: [M+H]+ = 437.
Step 3: 2-(4-Chlorobenzoyl)fluoro(4-fluorotetrahydro-2H-pyrancarbonyl)benzoic acid
2-(4—chlorobenzoyl)—3-fluoro(4-fluorotetrahydro-2H-pyrancarbonyl)benzoate (46.2 g, 105 mmol)
in THF (260 mL) and methanol (260 mL) was added 2M NaOH solution (530 mL). The resulting
orange solution was stirred at room ature for 1 h. Diethyl ether (500 mL) was added and the
layers were separated. The aqueous phase was adjusted to pH 1 using concentrated HCI, and the
resulting mixture was extracted with ethyl acetate (2 x 500 mL). The combined organic extracts were
dried (M9804), ed and evaporated under reduced re to give the title compound (37.4 g,
87%) as a colourless solid. The product was deemed sufficiently pure to be used in the subsequent
step. 1H NMR (400 MHz, CDCI3) 8.62 (1H, s), 8.08 (1H, dd), 7.71 (2H, d), 7.46 - 7.43 (2H, m), 4.01 -
3.84 (4H, m), 2.41 - 2.22 (2H, m), 2.04 - 1.96 (2H, m).
Preparation 54: (R)(4-chlorobenzoyl)fluoro(1-(4-fluorotetrahydro-2H-pyranyl)
ypropyl)benzoic acid
ng from 2-(4-chlorobenzoy|)fluoro(4-fluorotetrahydro-2H-pyrancarbonyl)benzoic acid
(Preparation 53).The title compound was prepared using procedures similar to those described in
Preparation 52. 1H NMR (400 MHz, CDCI3) 7.97 (1H, s), 7.71 (2H, d), 7.57 (1H, d), 7.43 (2H, d), 3.86
(2H, ddd), 3.71 - 3.59 (3H, m), 2.28 - 2.18 (1H, m), 2.03 - 1.60 (5H, m), 0.76 (3H, t). [G]D20 = +1606 (0
1.04, MeOH).
Preparation 55: 2-(4-Chlorobenzoyl)(cyclobutanecarbonyl)—3-fluorobenzoic acid
Starting from cyclobutylaldehyde, the title compound was prepared using procedures similar to those
described in Example 73, steps 1 and 2. MS: [M-H]' = 359
Preparation 56: trans((tert-Butyldiphenylsi|y|)oxy)cyclohexanecarbaldehyde
H0“. H0, TBDPSO,“
O|—>Step 2 Step 3
o Step1 o
Step 1: transHydroxy-N-methoxy-N-methylcyclohexanecarboxamide
To a solution of 4-hydroxycyclohexanecarboxylic acid (25 g, 173 mmol), EDCI (32 g, 208 mmol) and
N,O-dimethylhydroxylamine hydrochloride (19 g, 191 mmol) in DCM (500 mL) under nitrogen at room
temperature was added DIPEA (91 mL, 520 mmol) and the mixture stirred for 20 h. The reaction was
quenched with 2N aqueous HCI (50 mL), partitioned with water (400 mL), layers shaken and
separated, the aqueous re-extracted with DCM (2 x 150 mL). The combined organic extracts were
dried (MgSO4), filtered, and concentrated under reduced pressure to yield the desired product (21 g)
as a thick pale yellow oil. 1H NMR (400 MHz, CDCI3) 3.70 (3H, s), 3.68 - 3.59 (1H, m), 3.18 (3H, s),
2.70 - 2.55 (1H, m), 2.10 - 2.02 (2H, m), 1.88 - 1.80 (2H, m), 1.63 - 1.53 (2H, m), 1.38 - 1.26 (2H, m),
OH g.
Step 2: trans((tert-Butyldiphenylsilyl)oxy)-N-methoxy-N-methylcyclohexanecarboxamide
transHydroxy-N-methoxy—N-methylcyclohexanecarboxamide (12.2 g, 65 mmol), was dissolved in
DMF (200 mL) and stirred at room temperature under a nitrogen atmosphere. tert-
butyl(chloro)diphenylsilane (19.7 g, 71 mmol) was added, followed by imidazole (4.88 g, 71 mmol).
The reaction was d for 18h. The DMF was evaporated under reduced pressure, and the resulting
residue was re-dissolved in EtOAc (250 mL). The organic layer was washed with 4% aqueous LiCl
solution (2 x 150 mL), and then dried ), filtered, and evaporated under reduced pressure. The
crude residue was purified by silica column tography (gradient n 0 to 60% EtOAc in iso-
Hex), to give the pure product as a colourless oil which crystallises upon standing (19.0 g, 69% yield).
MS: [M+H]+ = 426.
Step 3: 4-((tert-Butyldiphenylsi|y|)oxy)cycIohexanecarbaldehyde
trans((tert-Butyldiphenylsilyl)oxy)-N-methoxy-N-methylcyclohexanecarboxamide (0.5 g, 1.17 mmol)
was dissolved in dry THF (7.5 mL) under a nitrogen atmosphere. The solution was cooled to —78°C,
and then DIBAL (1M in hexane, 2.11 mL, 2.11 mmol) was added dropwise. The mixture was stirred at
—78°C for 1.5 h and then quenched with 10% aqueous Rochelle salt solution (10 mL). The mixture
was allowed to warm to room temperature and was then diluted further with EtOAc (40 mL) and more
le salt solution (15 mL). The e was stirred for 20 mins before being transferred to a
separating funnel. The organic phase was collected, and the aqueous phase was extracted with
EtOAc (2 x 30 mL). The ed organic extracts were dried (MgSO4), filtered, and evaporated
under reduced pressure to give a crude residue which was used in next step without further
purification. 1H NMR (400 MHz, CDCI3) 9.56 (1H, s), 7.67 - 7.65 (4H, m), 7.43 - 7.34 (6H, m), 3.64 -
3.55 (1H, m), 2.20 - 2.13 (1H, m), 1.95 - 1.80 (4H, m), 1.48 - 1.37 (2H, m), 1.28 - 1.20 (2H, m), 1.05
(9H, s).
Preparation 57: (-)(1-(1-(tert-butoxycarbonyl)f|uoropiperidiny|)hydroxypropyl)—2-(4-
chIorobenzoyl)fluorobenzoic acid (*both isomers separated and ed)
Step 1: 5-(1-(tert-Butoxycarbonyl)fluoropiperidinecarbonyl)(4-chlorobenzoyl)
fluorobenzoic acid
A mixture of 5-(1-(fert—butoxycarbonyl)piperidine-4—carbonyI)(4-chlorobenzoyl)fluorobenzoic acid
(Example 80 step 2, 50 g, 0.102 mol) and NaOH (4.32g, 0.108 mol) was stirred in anhydrous THF
(250 mL) and anhydrous MeOH (90 mL) until all the NaOH dissolved. The solution was evaporated
under reduced pressure and the residue dissolved in anhydrous THF (400 mL) and added over 1
minute to a stirred solution of 1 M LHMDS in hexanes (125 mL) in ous THF (100 mL) at —40 °C
under nitrogen. The e was stirred for 20 minutes at —40 °C prior to the on of a solution of N-
fluorobenzenesulfonimide (48.6 g, 0.154 mol) in anhydrous THF (400 mL) in a steady stream over 1
minute. On complete on the mixture was stirred with cooling in a bath at —40 °C for 20 minutes.
The mixture was quenched with water (500 mL), stirred at room temperature for 30 minutes, the pH
adjusted to pH2 with 2N HCl and then the aqueous was ted with EtOAc (2 x 750 mL). The
combined cs were dried (MgSO4) and the solvent evaporated. The residue was triturated with
DCM (500 mL) and the solid filtered, washed with DCM and dried to afford the title compound as a
colourless solid (31.3 g, 60%). MS [M-H]'= 506.
Step 2: (-)(1-(1-(tert-butoxycarbonyl)fluoropiperidiny|)hydroxypropyl)—2-(4-
chIorobenzoyl)fluorobenzoic acid
To anhydrous THF (130 mL) at —50 °C under nitrogen was added a 1.72 M solution of EtLi in dibutyl
ether (38.4 mL, 65.96 mmol) followed by 1 M diethylzinc in hexanes (66.4 mL). This was stirred at -50
°C for 70 minutes prior to addition of a solution of tert—butoxycarbonyl)fluoropiperidine
carbonyl)(4-chlorobenzoyl)fluorobenzoic acid (13.4 g, 26.38 mmol) in anhydrous THF (130 mL)
in a gentle stream over 1 minute. On complete addition the mixture was stirred at —50 °C for 20
minutes, quenched by careful addition of water (200 mL), warmed to room temperature, acidified with
1M HCI and extracted into EtOAc (2 x 500 mL). Combined extracts were dried ) and the
solvent evaporated under reduced re. The residue was triturated with isohexane (500 mL), the
solvent decanted and the colourless solid dried to afford the title compound as the racemate. (13.9 g,
99%). MS [M-H]'= 536. The racemate (11.2 g) was separated by SFC to afford the title compound as
the slow running isomer (5.11 g, 45% yield).
(+)(1-(1-(tert-butoxycarbony|)fluoropiperidiny|)hydroxypropyl)(4-chlorobenzoyl)
fluorobenzoic acid: Fast running isomer* 1H NMR (400 MHz, CDCI3) 7.97 (1H, s), 7.72 (2H, d), 7.54
(1H, d), 7.43 (2H, d), 4.01 - 4.01 (2H, m), 3.00 - 2.89 (2H, m), 2.28 - 2.19 (1H, m), 2.08 - 1.98 (2H, m),
1.81 - 1.50 (3H, m), 1.43 (9H, s), 0.75 (3H, dd), exchangeable protons not observed. [db20 = +31410
(0 1, MeOH).
(-)(1-(1-(tert-butoxycarbonyl)fluoropiperidinyl)-1 -hydroxypropyl)(4-chlorobenzoyl)
fluorobenzoic acid: Slow running isomer* 1H NMR (400 MHz, CDCIg) 7.97 (1 H, s), 7.72 (2H, d), 7.54
(1H, d), 7.43 (2H, d), 4.01 - 4.01 (2H, m), 3.00 - 2.89 (2H, m), 2.28 - 2.19 (1H, m), 2.08 - 1.98 (2H, m),
1.81 - 1.50 (3H, m), 1.43 (9H, s), 0.75 (3H, dd), exchangeable s not observed. [or]DZ° = °(c
1, MeOH).
Preparation 58: (2-Bromomethylphenyl)methanamine
Prepared in a r manner to that described in e 33, Step 1; from 2-bromo
methylbenzonitrile. 1H NMR (400 MHz, CDCI3) 7.37 (1H, s), 7.25 (1H, d), 7.08 (1H, d), 3.86 (2H, s),
2.31 (3H, s).
Preparation 59: (2-Bromomethoxyphenyl)methanamine
Prepared in a similar manner to that described Example 33, Step 1; from 2-bromo
benzonitrile. 1H NMR (400 MHz, CDCI3) 7.26(1H, d), 7.11 (1H, d), 6.84 (1H, dd), 3.85 (2H, s),
3.79 (3H, s), 1.5 (2H, br s).
Preparation 60: (S)(1-(1-(tert-butoxycarbonyl)piperidinyl)hydroxypropyI)(4-
chlorobenzoyl)fluorobenzoic acid
BocN
Steps 1-2. Starting from the piperidine ketone (Examples 80 and 81, step 2, 11.0 g, 23 mmol), Steps
1 and 2 were performed using a procedure similar to that described for Preparation 52, Step 1-2, The
racemic mixture was separated by chiral SFC to give
(-)-tert-buty|-(S)(1-(4-(4-ch|orobenzoy|)f|uoro(methoxycarbony|)pheny|)
hydroxypropyl)piperidinecarboxylate (3g). MS: [|\¢’|+H]+ = 534, [01],;20 = -34.15 (c 1.18, MeOH).
rt-butyI-(S)(1-(4-(4-ch|orobenzoy|)f|uoro(methoxycarbonyl)pheny|)
hydroxypropyl)piperidinecarboxy|ate (3.6 g). MS: [M+H]+= 534, [or]D20 = +24.46 (c 1.02, MeOH).
Step 3: (S)(1-(1-(tert-butoxycarbonyl)piperidinyl)hydroxypropyl)(4-chlorobenzoyl)—3-
fluorobenzoic acid
Using procedures similar to those bed in Preparation 52 Step3, (-)-tert-buty| (S)(1-(4—(4-
chIorobenzoyI)fluoro(methoxycarbonyl)phenyl)hydroxypropy|)piperidinecarboxylate (3.0 g,
.6 mmol) gave 3.1 g ofthe title compound. MS: [M+H]+ = 518, [db20 = -37.51 (0.97 g/100 mL),
Preparation 61: 2-(4-Chlorobenzoyl)fluoro(pyridinecarbonyl)benzoic acid
The title compound was prepared using procedures similarto those bed in Example 73 (step 1-
2), but using pyridinecarboxaldehyde in step 1. Iodine (2 mol. Eq.), K2C03 (2 mol eq.), Ki (0.25 mol
eq) in water (90°C) was used as alternative oxidation conditions in Step 2. MS: [M + H]+ = 384.
ation 62: Propenyl (28,3S)amino(4-chlorophenyl)methylpropanoate
2016/053042
o M N, H ? E
\ S’N\ '
'S» N 0\ >4. ’H :
OH PMB
St 1 ('3' Step 2 " :3
9P o
0 Step 3 o'
CI Cl
Step 4
HZN - l4
O\/\ :
Step 5 "S—N OW
Step 1: N-[(1Z)-(4-Chlorophenyl)methylidene]methylpropane(R)sulfinamide. (R)
methylpropanesulfinamide (18.1 g, 150 mmol), robenzaldehyde (20.0 g, 143 mmol) and
052003 (51.0 g, 157 mmol) were suspended in DCM (150 mL) and stirred at room temperature for
16h. The mixture was then filtered through Celite and the volatiles were removed in vacuo to give the
desired product as a white solid (26.0 g, 75% yield). LCMS: [M+H]+ = 244.
Step 2: (4-Methoxyphenyl)methyl (28,3S)(4-chloropheny|)methy|[(2-methylpropane
(R)-sulfinyl)amino]propanoate. n-BuLi (2.5 M in hexane, 16.8 mL, 45 mml) was slowly added to a
solution ofdiisopropylamine (6.3 mL, 45 mmol) in THF (20 mL) at 0°C and the reaction stirred for 30
min. at the same temperature. The mixture was then cooled to -78°C and (4-methoxyphenyl)methyl
propanoate (7.2 mL, 40 mmol) was slowly added, keeping the temperature below -70°C. After 30min.
triisopropoxytitanium(|V) (1M in , 80 mL, 84 mmol) was added to the reaction mixture
and stirring was maintained for further 30min at -78°C. y, a solution of N-[(1Z)-(4-
chlorophenyl)methylidene]methylpropane(R)sulfinamide (5.0 g, 20 mmol) in THF (15 mL) was
slowly added and the reaction was stirred for further 2h at -78°C. NH4CI (aq), water and EtOAc were
added to the on, the mixture was wormed up to room temperature and vigorously stirred for 30
min. to dissolve most of the solid. The organic phase was separated and the aqueous phase was
extracted with EtOAc (3x). The organic phases were collected, dried over NaZSO4 and concentrated in
vacuo. The residue was columned on silica gel (gradient 0-100% EtOAc in Petrol) to give the desired
product as a yellow oil (5.1g, 58% yield). 1H NMR (400 MHz, DMSO-d6): .27 (2H, m), 7.24 (2H,
d), 7.00 (2H, t), 6.91-6.80 (2H, m), 5.72-5.63 (1H, m), 4.88-4.73 (2H, m), 4.24 (1H, t), 3.75 (3H, s),
2.99-2.87 (1H, m), 1.28 (3H, d), 1.02-0.90 (9H, m).
Step 3: (ZS,3S)(4-Chloropheny|)methyl[(2-methy|propane(R)-
sulfinyl)amino]propanoic acid. TFA (12 mL) was added to a solution of (4-methoxyphenyl)methyl
(2S,3S)—3-(4-chlorophenyl)methyl[(2-methylpropane-2— (R)-sulfinyl)amino]propanoate (6.0 g, 13.7
mmol) in DCM (20 mL) and the reaction was stirred at room temperature for 1h. The volatiles were
removed in vacuo and the residue was partitioned between 1M HCI and EtOAc. The aqueous phase
was extracted with EtOAc (3x), the organic phases were collected, dried over NaZSO4 and
concentrated in vacuo to give the give the desired product ad a light brown solid which was used
without further purifications in the next step. LCMS: [M+H]+ = 318.
Step 4: Propenyl (ZS,3S)(4-chloropheny|)methyI[(2-methy|propane(R)-
su|finy|)amino]propanoate. Allyl bromide (2.3 mL, 26 mmol) was added to a sion of )-
3-(4-chloropheny|)methyl[(2-methylpropane-2—(R)—sulfinyl)amino]propanoic acid (crude from
previous step, ca. 13 mmol) and K2C03 (5.4 g, 39 mmol) in DMF. The reaction mixture was stirred at
room temperature for 2h, quenched with water and extracted with EtOAc. The organic was dried over
NaZSO4 and trated in vacuo. The residue was columned on silica gel (gradient 0-100% EtOAc
in Petrol) to give the desired product as a yellow oil (2.8g, 57% yield over 2 steps). 1H NMR (400 MHz,
DMSO-d6): 7.40-7.32 (2H, m), 7.29 (2H, d), 5.74-5.58 (2H, m), 5.15-5.02 (2H, m), 4.41-4.31 (2H, m),
4.27 (1H, t), 3.00-2.88 (1H, m), 1.28 (3H, d), 1.01 (9H, s).
Step 5: Propenyl (ZS,3S)amino(4-chlorophenyl)methylpropanoate. HCI (4M in
dioxane, 15 mL) was added to a solution of propenyl )(4-chlorophenyl)methyl[(2-
methylpropane(R)-sulfinyl)amino]propanoate (2.8 g, 7.8 mmol) in EtOH (20 mL) and the suspension
was stirred at room temperature for 30 min. The volatiles were removed in vacuo and the residue was
partitioned between NaHC03 and EtOAc. The organic was collected, dried over NaZSO4 and
concentrated in vacuo to give the desired product as a yellow oil (1.7g, 86% yield). 1H NMR (400
MHz, DMSO-d6): 7.34 (4H, s), 5.81-5.68 (1H, m), 5.22-5.06 (2H, m), 4.46-4.33 (2H, m), .94 (1H,
m), 2.76-2.60 (1H, m), 1.96 (2H, s), 1.09 (3H, d).
Preparation 63: Ethyl 2-[2-(aminomethyl)—5-chlorophenoxy]acetate hydrochloride
N EISOC $00 HCI
| | HN HN H2N
Step 1 OH EP/OH Step 2 fij/OVCOZEI‘ O
Step 3 VCOZEt
CI CI Cl
Step 1: tert-Butyl N-[(4-chlorohydroxyphenyl)methy|]carbamate
To a stirred solution of 4-chlorohydroxybenzonitrile (1.57 g, 10.0 mmol) in dry ol (70 mL),
cooled to 0°C, were added Boczo (4.36 g, 20.0 mmol) and NiCl2-6HZO (0.24 g, 1.0 mmol). NaBH4
(2.65 g, 70.0 mmol) was then added in small portions over 30 min. The reaction was exothermic and
effervescent. The resulting reaction mixture ning a finely divided black precipitate was allowed to
warm to room ature and left to stir for a further 1 h, at which point diethylenetriamine (1.1mL,
.0 mmol) was added. The mixture was allowed to stir for 30 min before solvent evaporation. The
residue was dissolved in DCM (50 mL), dine (2 mL) was added and stirred for 30 min, then the
organic phase was washed with water and saturated NaHC03. The organic layer was dried (MgSO4)
and the solvent d in vacuo and the crude product was d by column chromatography to
afford the title compound (1.47 g, 57%). MS:[M + H]+ = 256.
Step 2: Ethyl 2-[2-({[(tert-butoxy)carbonyl]amino}methyl)chIorophenoxy]acetate
To a solution of tert-butyl N-[(4-chlorohydroxyphenyl)methyl]carbamate (1.47 g, 5.7 mmol) in DMF
(30 mL) was added KZCO3 (0.95 g, 6.9 mmol) and the mixture was stirred for 30 min. Ethyl
bromoacetate (1.4 mL, 8.55 mmol) was added and the stirring was continued for 2 hr. Water was
added, the on mixture was ted with EtOAc, the organic phase was washed with brine (3X),
dried and the solvent was evaporated. The crude material was purified by column chromatography,
eluted with petroleum ether — EtOAc 0-40% to afford the title compound (1.64 g, 84%). MS:[M + H]+ =
243 (M-Boc).
Step 3: Ethyl aminomethyl)chlorophenoxy]acetate hydrochloride
To a solution of ethyl 2-[2-({[(tert-butoxy)carbonyl]amino}methyl)chlorophenoxy]acetate (1.8 g, 5.25
mmol) in dioxane (20 mL) was added 4M dioxane —HC| and the mixture was stirred for 16 hr. The
solvent was evaporated to afford white solid (1.3 g, 89%). MS:[M + H]+ = 244.
Preparation 64: 2-(4-chlorobenzoyl)fluoro(1-hydroxytrans
hydroxycyclohexyl)propyl)benzoic acid (*both isomers separated and isolated)
O Step 1 TBDPSO,“
OH Step2
Step 1: 5-(trans((tert-Butyldiphenylsilyl)oxy)cyclohexanecarbonyl)(4-chlorobenzoyI)
fluorobenzoic acid
Starting from trans((tert-butyldiphenylsi|y|)oxy)cyclohexanecarbaldehyde (Preparation 56), the title
compound was prepared using procedures similar to those described in Example 73, steps 1 and 2.
MS: [M - H]‘ = 641.
Step 2: 5-(1-(trans((tert-butyldiphenylsilyl)oxy)cyclohexyl)hydroxypropyl)(4-
chlorobenzoyl)fluorobenzoic acid
The title compound was prepared using the procedure described in ation 52, step 1, and the
enantiomers were ted by chiral SFC.
(+)(1-(trans((tert-butyldiphenylsilyl)oxy)cyclohexyl)hydroxypropyl)—2-(4-chlorobenzoyl)-
3-fluorobenzoic acid: *fast running isomer 1H NMR (400 MHz, CDCIg) 7.8 (1H, s), 7.70 - 7.63 (6H,
m), 7.43 - 7.34 (9H, m), 3.55 - 3.46 (1H, m), 1.94 - 1.78 (5H, m), 1.43 - 1.24 (3H, m), 1.03 (9H, s), 0.96
- 0.83 (3H, m), 0.69 (3H, t), Exchangeable not observed. MS: [M - H]- = 671. [db20 = +2765 (0 1.0
MeOH).
(-)(1-(trans((tert-butyldiphenylsi|y|)oxy)cyclohexyl)hydroxypropy|)(4-ch|orobenzoyl)-
3-fluorobenzoic acid: *slow g isomer 1H NMR (400 MHz, CDCI3) 7.79 (1H, s), 7.70 - 7.63 (6H,
m), 7.44 - 7.33 (9H, m), 3.54-3.48 (1H, m), 1.96 - 1.75 (5H, m), 1.46 - 1.16 (3H, m), 1.03 (9H, s), 0.96 -
0.85 (3H, m), 0.69 (3H, t), Exchangeable not observed. MS: [M - H]- = 671. [or]D20 = -24.62 (c 1.0,
MeOH).
Step 3: 2-(4-chlorobenzoy|)fluoro(1-hydroxytranshydroxycyclohexy|)propy|)benzoic
acid
(-)1-(trans((tert-Butyldiphenylsily|)oxy)cyclohexy|)hydroxypropy|)(4-chlorobenzoy|)
fluorobenzoic acid (3.5 g, 5.2 mmol) was dissolved in THF (70 mL) and the mixture was treated with
TBAF (1M in THF, 20.7 mL, 20.7 mmol) and heated overnight at 60 °C. The on was quenched
with ted aqueous NaHCOs solution and extracted with ethyl acetate (2 x 75 mL). The combined
organic layers were dried (MgSO4), filtered and evaporated to dryness under reduced pressure to give
a crude product. The residue was purified by column chromatography (gradient elution, 20 to 100%
ethyl e in iso-hexane (with 0.1% formic acid)) to give the title compound (1.92 g, 85%) as a
colourless oil. MS: [M - H]- = 433.
Preparation 65: 2-(4-chlorobenzoyl)fluoro(1-methyl-1H-pyrazo|ecarbony|)benzoic acid
The title compound was prepared using ures similarto those described in Example 72, Step 1
and Step 2, but using 1-methyl-1H-pyrazolecarbaldehyde instead of 1-methyl-1H—pyrazole
aldehyde in Step 1; and manganese dioxide in 1,4-dioxane at 100°C instead of TEMPO/sodium
hypochlorite in Step 2 + H]+= 387
. MS [M
Example 1: (3R)(4-ch|oropheny|)[(4-chlorophenyl)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone
To a solution of MeMgBr (3M THF, 0.97 mL, 2.91 mmol) in THF (4.0 mL) at room temperature was
added Zn(ll)Cl2 (30 mgs, 0.22 mmol) and the ing on stirred at this temperature for 1 hour.
The solution was cooled to 0 CC before the addition of a cool solution of yl-2—(4-chlorobenzy|)
(4-chlorophenyl)((3’-(hydroxymethyl) cyclopropyl)methoxy)isoindolinone (Preparation 15) (573
mgs, 1.12 mmol) in THF (4.0 mL) and the resulting e stirred at 0 0C for 2 hours. The reaction
was quenched by the addition of a saturated solution of NH4C| and the resulting mixture extracted
with EtOAc (x3). The combined organic phases were washed with brine, dried ), filtered and
concentrated in vacuo. Purification on silica gel (Biotage SP4) eluting with 20% —> 80% EtOAcfPet
gave product as a clear gum (320 mg; 54%). Separation by chiral HPLC (Chrialpak IA 250 x 10 mm
id, 75% heptane, 2-propanol, 4.5 mLfmin) gave the title compound (Example 1) (R)(4-
Chlorobenzyl)(4-chlorophenyl)((3’-(hydroxymethyl)cyclopropyl) methoxy)(2-hydroxypropan
indo|inone. 6H (500 MHz, CD30D) 0.12-0.23 (2H, m, 3’-CH2), 0.37-0.45 (2H, m, 3’-CH2’), 1.56
(6H, s, (CH3)2), 2.76 (1H, d, J = 9.0, 2’-H), 2.88 (1H, d, J = 9.0, 2’-H’), 3.42 (1H, d, J = 11.2, 4’-H), 3.58
(1H, d, J = 11.2, 4’-H‘), 4.40 (1H, cl, J = 15.1, N-CH), 4.45 (1H, d, J = 15.1, N-CH’), 7.07-7.14 (4H, m,
Ar-H), 7.15-7.22 (5H, m, Ar—H & 4-H), 7.76 (1H, dd, J = 1.2, 8.1, 5-H), 8.02 (1H, d, J = 1.2, 7-H);
HRMS, Found; M+ 526.1540/..
Example 2: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindolone
OH 0
In a microwave vial was added Hg(OAc)2 (145 mg, 0.456 mmol) and water (0.3 mL). After stirring for
min, THF (0.25 mL) was added, followed by a solution of hlorobenzyl)(4-chlorophenyl)
((1-(hydroxymethyl)cyclopropyl)methoxy)(propeny|)isoindolinone (Preparation 14)
(150 mg, 0.285 mmol) in THF (0.5 mL). The orange solution was stirred at room temperature for 1.5
h, after which time perchloric acid (60%, 7.5 pL) was added, with the orange colour fading to yellow
after 2.5 h. 1M aq. NaOH (0.8 mL) was added and the solution turned brown, then NaBH4 (22 mg,
0.570 mmol) was introduced and the colour changed to metallic grey. The on mixture was left to
stir for 16 h, then filtered through Celite®, followed by a thiol dge and the solvent removed in
vacuo. FCC [petrol-ethyl acetate (100:0)—>(80:20)—>(50:50)—>(20:80)] of the crude residue, followed
by preparative HPLC, afforded 28 mg, 20%, of a white solid. Separation of the two enantiomers was
carried out by preparative chiral HPLC to give (R)-2—(4-Chlorobenzyl)(4-chloropheny|)fluoro
((1-(hydroxymethyl)cyclopropyl)methoxy)(2-hydroxypropanyl)isoindolinone (Example 2). 1H
NMR (500 MHz, CDCI3): 7.77 (1H, d, 7-H), 7.37 (1H, dd, ArH), 7.20-7.26 (4H, m, 4 xArH), 7.10-7.16
(4H, m, 4 xArH), 4.55 (1H, d, NC-H’), 4.11 (1H, d, NC-H), 3.53 (1H, d, 4’-H’), 3.34 (1H, d, 4’-H), 2.96
(1 H, d, 2’-H’), 2.69 (1 H, d, 2’-H), 1.55-1.64 (6H, 2 x s, 2 x CH3), 0.37-0.44 (2H, m, Cy-Py-Hz) and 0.08-
0.23 (2H, m, Cy-Py-H). MS: [M- cngoz]+ 442
Example 3: (3R)(4-chIorophenyl)—2-[(4—chlorophenyl)methyI](2-hydroxyethoxy)—6-(2-
hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindolone
The title compound was ed from (Preparation 21) (0.062 g, 0.128 mmol), ZnClz (0.005 g,
0.035 mmol) and of MeMgCl (3M in THF) (0.15 mL, 0.46 mmol) using a procedure similar to that
bed for e 1. The product was obtained as a white foamy solid (0.030 g, 0.062 mmol, 47
%). Separation by preparative chiral prep HPLC gave Example 3. 1H NMR (500 MHz, CDCI3) 6 (ppm)
1.50 (s, 1H), 1.62 (s, 6H), 1.82 (s, 1H), 2.77-2.83 (m, 1H), 2.87-2.95 (m, 1H), 3.33-3.47 (m, 2H), 4.08
(d, 1H), 4.64 (d, 1H), 7.09 (dd, 1H), .20 (m, 4H), 7.20-7.25 (m, 4H), 7.71 (dd, 1H), 8.0 (d, 1H);
MS(ES+) m/z 486.3 [M+H]+;
Example 4: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[3-(hydroxymethyl)oxetan
yl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindolone
Starting from Preparation 23. The title compound was prepared in a similar fashion to Example 1, but
using 4.5 mol eq. of MeMgCl. 1H NMR (500 MHz, CDCI3) 6 (ppm) 1.60 (s, 1H, OH), 1.62 (s, 3H,
C(CH3)2), 1.63 (s, 3H, C(CH3)2), 1.92 (s, 1H, OH), 2.99 (d, 1H, J = 9.2 Hz, OCHHC), 3.03 (d, 1H, J =
9.2 Hz, , 3.67-3.75 (m, 2H, CCHZOH), 4.18 (d, 1H, J = 15.2 Hz, NCHH), 4.25 (d, 1H, J = 6.3
Hz, oxetane CHH), 4.30 (d, 1H, J = 6.2 Hz, oxetane CHH), 4.33 (d, 1H, J = 6.3 Hz, oxetane CHH),
4.36 (d, 1H, J = 6.2 Hz, oxetane CHH), 4.56 (d, 1H, J = 15.2 Hz, NCHH), 7.07 (d, 1H, J = 7.9 Hz,
isoindolinone-H), 7.09-7.19 (m, 6H, Ar—H), 7.22 (d, 2H, J = 8.9 Hz, Ar—H), 7.73 (dd, 1H, J = 8.0, 1.7 Hz,
isoindolinone-H), 8.03 (d, 1H, J = 1.7 Hz, isoindolinone-H);; MS(ES+) m/z 586.3 [M+HCOO‘]';
Exam pie 5: 1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropan-Z-yl)
oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarboxylic acid
>8 Cl
CH 0
To a solution of 2—(4-chlorobenzyl)(4-chlorophenyl)((1-(hydroxymethyl)cyclopropyl)methoxy)
(2-hydroxypropanyl)isoindolinone (Example 1) (110 mg, 0.21 mmol), RuCI3.H20 (10 mg, 0.02
mmol) in a mixture of EtOAc (0.7 L alcohol), MeCN (0.7 L alcohol), and water (0.4
mmol/mL alcohol), was added sodium periodate (4.1 mol eq.) and stirred at rt for 15 min before being
diluted with EtOAc (0.04 mmol/mL alcohol) and filtered through a Celite plug. The filtrates were
washed with water (0.04 mmol/mL alcohol) and the aqueous was extracted with EtOAc (2 x 0.04
mmol/mL l). The combined organic phases were washed with brine (0.02 mmol/mL alcohol),
dried (MgSO4) and concentrated in vacuo., sodium periodate (185 mg, 0.86 mmol), MeCN (0.6 mL),
EtOAc (0.6 mL) and water (1.0 mL). Purification (SP4, , EtOAc/petrol (0.1% AcOH), 60%)
followed by semi-preparative chiral HPLC (C-18 silica, MeCN/0.1% aq. formic acid, 40%) gave
Example 5 as a white solid (54 mg, 48%). 1H NMR (500 MHz, CDCI3) 6 0.48-0.52 (1H, m, H-
ropane), 0.57-0.61 (1H, m, H-cyclopropane), 1.18-1.21 (1H, m, H-cyclopropane), 1.25-1.29 (1H,
m, H-cyclopropane), 1.60 (3H, s, CH3), 1.62 (3H, s, CH3), 2.71 (1H, d, J = 9.6 Hz, -iso-OCHH), 3.16
(1H, d, J = 9.6 Hz, -iso-OCHH), 4.19 (1H, d, J = 15.1 Hz, NCHH), 4.58 (1H, d, J = 15.1 Hz, NCHH),
7.09-7.19 (9H, m, H-Ar), 7.71 (1H, dd, J = 1.8, 8.0 Hz, H-5), 7.98 (1H, d, J = 1.8 Hz, H-7);; LRMS (ES'
) m/z 538.3 [M-H]';
Example 6: (3R)(4-chlorophenyl)[(1S)(4-chloropheny|)ethyl](2,3-dihydroxy
methylpropoxy)(2-hydroxypropanyl)-2,3-dihyd ro-1 H-isoindolone
(*as a mixture of isomers at the position shown)
.Ҥ 0.
(S)—3-(4-Chlorophenyl)((S)(4-chlorophenyl)ethyl)((2-(hydroxymethyl)allyl)oxy)(propen
indolinone, Preparation 24, (90 mg, 0.18 mmol), Hg(OAc)2 (140 mg, 0.44 mmol), water (0.5
mL), THF (0.5 mL), 6 M NaOH (0.2 mL) and NaBH4 (269 mg, 7.1 mmol). Purification (SP4, silica,
EtOAc/petrol, 80%) gave Example 6 as a mixture of 2 isomers (27 mg, 28%). 1H NMR (500 MHz,
CDCIg) 6 mixture of reoisiomers 0.95 (3H, s, -iso-OCH2C(OHCH3)CHZOH), 1.00 (3H, s, -iso-
OCH2C(OHCH3)CH20H), 1.64 (6H, s, C(CH3)ZOH), 1.65 (6H, s, C(CH3)ZOH), 1.67 (3H, d, J = 7.3 Hz,
NCHCH3), 1.71 (3H, d, J = 7.5 Hz, NCHCH3), 1.87 (2H, brs, OH x2), 2.00 (2H, brs, OH x2), 2.65-2.73
(4H, m, -iso-OCH2 x2), 3.23-3.36 (4H, m, CHZOH x2), 4.24 (1H, q, J :75 Hz, NCHCH3), 4.28 (1H, q, J
= 7.3 Hz, NCHCH3), 7.01 (1H, d, J = 7.9 Hz, H-4), 7.02 (1H, d, J = 7.9 Hz, H-4), 7.28-7.36 (6H, m, HAr
), 7.55-7.57 (2H, m, H-Ar), 7.71-7.74 (2H, m, H-5), 7.98-7.99 (2H, m, H-7). LRMS (ES+) m/z 566.4
[M+Na]+.
Example 7: -(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone
Example 7 was prepared from Preparation 28 using similar procedure to those described in Example
1. NMR (500 MHz, CDCIS) 6 0.33-0.36 (1H, m), 0.42-0.45 (1H, m), 0.47-0.54 (2H, m), 1.53 (3H, s),
1.55 (3H, s, ), 1.84 (3H, d, ), 2.89 (1H, d, ), 3.21 (1H, d, ), 3.56 (2H, ), 4.28 (1H, q, )), .98 (9H, m,
), 7.63 (1H, dd, ), 7.85 (1H, d, ); MS (ES+) 540.5 [M+H]+
Examples 8 and 9: (3R)(4-chIorophenyl)[(4-chIorophenyl)methyl](1,2-dihydroxypropan-
2-yl){[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone
*The two separate epimers at the position shown
To a solution of 2-(4-chIorobenzyI)(4-chIorophenyI)((1-(hydroxymethyl)cyclopropyl)methoxy)
(propeny|)isoindo|inone, Preparation 13 (1.20 g, 2.36 mmol) in acetone/water (18 mL/7.1 mL)
was added methyl morpholine-N-oxide (318 mg, 2.71 mmol) and OsO4 (2.5wt% in tBuOH, 0.90 mL,
0.09 mmol) and the resulting yellow/orange solution stirred at RT for 3.5 h. The on was diluted
with DCM (125 mL), washed with ted aqueous sodium sulphite (125 mL), dried over MgSO4 and
concentrated under vacuum. MPLC (1:1 petrol/EtOAc to 100% EtOAc) gave the product as a white
solid as a mixture of 4 diastereoisomers (592 mg, 46%). Purification by preprative chiral HPLC (324
mg, Chiralpak IA 250 x 10 mm, 92.5:7.5 Heptane/Ethanol) gave-
Example 8 (isomer 1): 6H (500 MHz, CDCI3) 0.13-0.16 (2H, m, 2 x c-PrH), 0.40-0.42 (2H, m, 2 x c-
PrH), 1.57 (3H, s, CH3), 2.64 (1 H, d, J = 9.4 Hz, CHH’), 2.64 (1H, d, J = 9.4 Hz, CHH’), 3.35 (1H, d, J =
11.3 Hz, CHH’), 3.49 (1H, d, J =11.3 Hz, CHH’), 3.69 (1H, d, J =11.0 Hz, CHH’), 3.79 (1H, d, J =11.0
Hz, CHH’), 4.19 (1H, d, J = 14.9 Hz, CHH’), 4.54 (1H, d, J = 14.9 Hz, CHH’), 7.12--7.21 (9H, m, 9 x
ArH), 7.70 (1 H, dd, J = 1.7 and 7.9 Hz, ArH), 7.97 (1H, d, J :17 Hz, ArH).
Example 9 (isomer 2): 6H (500 MHz, CDCI3) 0.14-0.15 (2H, m, 2 x c-PrH), 0.40-0.41 (2H, m, 2 x c-
PrH), 1.57 (3H, s, CH3), 2.63 (1H, d, J = 9.4 Hz, CHH’), 2.65 (1H, d, J = 9.4 Hz, CHH’), 3.34 (1H, d, J
=11.3 Hz, CHH’), 3.49 (1H, d, J =11.3 Hz, CHH’), 3.69 (1H, d, J =11.0 Hz, CHH’), 3.61 (1H, d, J =
11.0 Hz, CHH’), 4.19 (1H, d, J =14.9 Hz, CHH’), 4.54 (1H, d, J =14.9 Hz, CHH’), 7.21 (9H, m, 9
x ArH), 7.69 (1 H, d, J = 7.6 Hz, ArH), 7.96 (1H, s, ArH).
Examples 10 and 11 : (3R)(4-ch|oropheny|)[(1S)(4-chloropheny|)ethyl](2-hydroxy
methoxypropany|){[1-(hydroxymethyl)cycIopropyl]methoxy}-2,3-dihydro-1H-isoindol
*The two separate epimers at the position shown
At 0 °C, to a solution of (3R)((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl) methoxy) (4-
chlorophenyI)((S)(4-chlorophenyl)ethyI)(2-hydroxymethoxy propanyl)isoindolinone,
Preparation 31 (80 mg, 0.12 mmol), in THF (4 mL) was added TBAF (1.0 M in THF, 0.13 mL, 0.13
mmol) and the reaction stirred at 0 °C for 1 h then at RT for 18 h. The reaction was diluted with water
(20 mL), neutralised with 1.0 M aqueous HCI, extracted into EtOAc (2 x 25 mL), washed with brine (50
mL) and dried over MgSO4. MPLC (1:1 petrol/EtOAc to 100% EtOAc) and reparative HPLC
(ACE 5 C18-AR 150 x 4.6 mm id, 5um, 55:45 AcetonitrileNVater + 0.1 %v/v Formic Acid) gave
t as a white solid as a diastereoisomeric mixture (36 mg, 53%); MS (ES+) 570.4 [M+H]+;
Purification by preparative chiral HPLC (82 mg, Chiralpak IC 250 x 10 mm Mobile Phase:
e/Ethanol 87.5:12.5) gave :-
Example 11 (*isomer1): 6H (500 MHz, CDCI3) 0.32-0.36 (1H, m, c-PrH), 0.42-0.46 (1H, m, c-PrH),
0.47-0.53 (2H, m, 2 x c-PrH), 1.44 (3H, s, CH3), 1.63 (3H, d, J = 7.3 Hz, CHCH3), 2.69 (1H, d, J = 9.5
Hz, CHH’), 3.21 (1H, d, J = 9.5 Hz, CHH’), 3.32 (3H, s, OCH3), 3.43 (1H, d, J = 9.2 Hz, CHH’), 3.54
(1H, d, J = 9.2 Hz, CHH’), 3.56 (2H, s, CH2), 4.26 (1H, q, J = 7.3 Hz, , 6.92-6.99 (9H, m, 9 x
ArH), 7.62 (1H, dd, J = 1.7 and 8.0 Hz, ArH), 7.81 (1H, d, J = 1.7 HZ, ArH). m/Z 468.4 [M-
OCH2C(CH2-CH2)CH20H]+
Example 10 (*isomer 2): 1H-NMR 5H (500 MHz, CDCI3) 0.32-0.38 (1H, m, c-PrH), 0.42-0.48 (1H, m,
c-PrH), 0.47-0.53 (2H, m, 2 x c-PrH), 1.48 (3H, s, CH3), 1.83 (3H, d, J = 7.3 Hz, , 2.87 (1H, d,
J = 9.5 Hz, CHH’), 3.22 (1H, d, J = 9.5 Hz, CHH’), 3.32 (3H, s, OCH3), 3.41 (1 H, d, J = 9.2 Hz, CHH’),
3.51 (1H, d, J = 9.2 Hz, CHH’), 3.58 (2H, s, CH2), 4.28 (1H, q, J = 7.3 Hz, CHCH3), 8.92-8.99 (9H, m,
9 x ArH), 7.84 (1H, dd, J = 1.7 and 7.9 Hz, ArH), 7.79 (1H, d, J = 1.7 Hz, ArH). : m/z 488.4 [M-
OCH2C(CH2-CH2)CH20H]+
Examples 12 and 13: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][1-(dimethylamino)-
2-hydroxypropanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindol
(*Two isomers at the on shown)
ection of Preparation 34 using a similar procedure to that described for Example 10 followed
by purification by preparative chiral HPLC gave the two diastereoisomers:
Example 12 (*isomer1): 1H NMR (500 MHz, CDClg) 0.12-0.20 (2H, m), .45 (2H, m), 1.83 (3H,
s), 2.58 (1H, d), 2.88-2.29 (3H, m), 2.89 (1H, d), 2.99-3.00 (3H, m), 3.29 (1H, d), 3.39-3.42 (1H, m),
3.55 (1H, d), 3.82-3.88 (1H, m), 4.18 (1H, cl), 4.54 (1H, d), 7.11-7.22 (9H, m), 7.88 (1H, d), 7.94 (1H,
s). Ms:[M+H]*=589.5.
Example 13 (* isomer 2): 1H NMR (500 MHz, c0013) 0.12-0.23 (2H, m), 0.36-0.42 (2H, m), 1.80 (3H,
s), 2.42 (1H, d), 2.84 (3H, s), 2.92 (1H, d), 2.99-3.00 (3H, m), 3.15 (1H, d), 3.40-3.45 (1H, m), 3.58-
3.83 (2H, m), 4.17 (1H, d), 4.53 (1H, cl), 7.11-7.15 (4H, m), 7.19-7.23 (5H, m), 7.85 (1H, s), 7.98 (1H,
d). H]+=569.5.
Example 14: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl){[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindol-
2-yl]propanoic acid
Starting from Preparation 40, Example 14 was prepared using similar ures to those described
for e 1. 1H NMR (500 MHz, MeOD) 6 0.40-0.44 (4H, m, 4 x c-PrH), 1.47 (6H, s, 2 x CH3), 2.91
(1H, d, J = 9.2 Hz, CHH’), 3.09-3.21 (2H, m, CHH’ and CHCHH’), 3.51 (2H, s, CH2), 3.73 (1H, dd, J =
11.1 and 16.2 Hz, CHCHH’), 4.64 (1H, dd, J = 3.6 and 11.1 Hz, CHCHH’), 6.89-6.97 (8H, m, 8 x ArH),
7.02 (1H, d, J = 8.0 Hz, ArH), 7.65 (1H, dd, J = 1.7 and 8.0 Hz, ArH), 7.89 (1 H, d). MS. 582.1 (M-H+)'
Example 15: (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](1,2-dihydroxypropan
yl){[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone
Starting from (R)(4-ch|oropheny|)-2—((S)(4-ch|oropheny|)ethy|)((1-
(hydroxymethy|)cyclopropyl)methoxy)(propeny|)isoindo|inone (Preparation 27) ; Example
was prepared by using procedures similarto those described for Example 8. Product was obtained
as a white solid as a diastereoisomeric e (65 mg, 31 (500 MHz, CDCI3) 6 0.34-0.36 (1H, m, c-
PrH), 0.43-0.52 (3H, m, 3 x c-PrH), 1.49-1.50 (3H, m, CH3), 1.82-1.84 (3H, m, NCHCH3), 2.89-2.92
(1H, m, alkyI-CH), 3.18-3.21 (1H, m, CH), 3.51-3.56 (2H, m, 2 x CH), 3.58-3.63 (1H, m,
alkyl-CH), 3.71-3.75 (1 H, m, alkyI-CH), 4.28 (1 H, q, J = 7.3 Hz, NCHCH3), 6.92-6.97 (8H, m, 8 x ArH),
7.00 (1H, d, J = 7.9 Hz, ArH), 7.60-7.62 (1H, m, ArH), 7.84-7.85 (1H, m, ArH); MS (ES+) 454.3 [M-
HOCH2(c-Pr)CH20]';
Example 16: (3R)(4-chlorophenyl)—2-[(4-chlorophenyl)methyl](3-hydroxymethylbutoxy)-
6-(2-hydroxypropan-Z-yl)—2,3-dihydro-1H-isoindolone
VCQ“
OH ob
Starting from Preparation 41, Example 16 was prepared using similar ure to those described
in Example 1. Purification by chrial HPLC (Chiralpak IC 250 x 10 mm, Mobile Phase: Heptane/Ethanol
80:20): gave Example 16: (R)(4-Chlorobenzy|)(4-chlorophenyl)(3—hydroxy—3-methylbutoxy)
(2-hydroxypropan-2—yl)isoindolinone (39 mg) . 1H NMR (500 MHz, CDCI3) 6 0.98 (3H, s, CH3), 1.02
(3H, s, CH3), 1.13-1.19 (1H, m, CH), 1.35-1.41 (1H, m, CH), 1.55 (3H, s, CH3), 1.56 (3H, s, CH3), 2.75-
2.80 (1H, m, CH), 2.86-2.91 (1H, m, CH), 3.91 (1H, d, J = 14.9 Hz, NCHH’), 4.65 (1H, d, J =14.9 Hz,
NCHH’), 7.03 (1H, d, J = 7.9 Hz, ArH), 7.11-7.18 (8H, m, 8 x ArH), 7.65 (1H, dd, J = 1.7 and 7.9 Hz,
ArH), 7.93 (1H, d, J = 1.7 Hz, ArH); 13C (125 MHz, CDCIS) 6 29.1, 29.9, 31.9, 32.0, 41.5, 42.4, 60.0,
70.0, 72.6, 95.2, 120.0, 122.7, 127.8, 128.4, 128.8, 129.5, 130.7, 131.4, 133.3, 134.7, 136.2, 137.0,
143.4, 151.7, 168.3; MS (ES+) 424.3 [M-(OH)C(CH3)2CH2CHZO]';
Example 17: (3R)(4-chlorophenyI)[(4-chlorophenyl)methyI](2-hydroxypropanyl)
yrazolyl)methoxy]-2,3-dihydro-1H-isoindolone
HN:/>—N / Step 1 Step 2 0H
N / Step 3
I co Et Nj>_I 2 CO2Et
‘N / l5 /
SEM CI
SEM'
Example 17, Step 1: Ethyl 1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolecarboxylate
NaH (80% in oil, 130 mg, 4.3 mmol, 1.5 eq.) was added to 4-ethoxycarbonyl-1H-pyrazole (400 mg,
2.86 mmol, 1 eq.) in THF (8 mL), and the mixture stirred at r.t. for 30 min. SEMCI (556 uL, 3.14 mmol,
1.1 eq.) was added and the on d at r.t. for 18 h. The mixture was partitioned between EtOAc
(2 X 30 mL) and water (20 mL), the organic layers dried (MgSO4) and the solvent removed in vacuo.
The residue was purified by MPLC on silica with gradient elution from 5-20% EtOAc/petrol to give the
title compound as a clear oil (657 mg, 85%); HRMS found 271.1468 MH+
Example 17, Step 2: (1-((2-(Trimethylsilyl)ethoxy)methyl)-1H-pyrazolyl)methanol
LiAIH4 (1 M in THF, 1.94 mL, 1.94 mmol, 1.5 eq.) was added to ethyl 1-((2-
thylsilyl)ethoxy)methyl)-1H-pyrazole-4—carboxylate (350 mg, 1.29 mmol) in THF (4 mL) at 0 °C,
and the e was stirred at r.t. for 4 h. Water (74 uL) was added, followed by NaOH (15% aq, 220
uL) and water (74 uL). The e was d at r.t. for 1 h, filtered, the solids washed with EtOAc,
and the filtrate evaporated in vacuo to give the title compound as a clear oil (330 mg, ; HRMS
calc for C10H2102N28i 229.1367, found 229.1361. MH+
Example 17, Step 3: 6-Acetyl(4-chlorobenzyl)(4-chlorophenyl)((1-((2-
(trimethylsilyl)ethoxy)methyl)-1H-pyrazolyl)methoxy)isoindolinone
Prepared in a similar manner to Preparation 12 using 6-acetyl(4-chlorobenzy|)(4-chlorophenyl)—
3-hydroxyisoindolinone (Preparation 20) (150 mg, 0.35 mmol, 1 eq.), SOCIZ (51 uL, 0.70 mmol, 2
eq.) and (1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolyl)methanol (161 mg, 0.70 mmol, 2 eq.) The
title compound was obtained as a clear glass (105 mg, 47%); HRMS found 636.1830. MH+
Example 17, Step 4: 2-(4-ChIorobenzyl)(4-chlorophenyI)(2-hydroxypropanyl)((1-((2-
(trimethylsilyl)ethoxy) methyl)—1H-pyrazolyl)methoxy)isoindolinone
Prepared in a similar manner to e 1 using MeMgCl (151 uL, 3 M in THF, 0.45 mmol, 1.5 eq.)
and ZnClz (8 mg, 0.06 mmol, 0.2 eq.) in THF (1 mL), followed by 6-acetyl(4—chlorobenzyl)(4-
chlorophenyl)((1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolyl)methoxy)isoindo|inone (192
mg, 0.30 mmol, 1 eq). The title compound was obtained
as a clear gum (78 mg, 40%); HRMS 652.2145. MH+
Example 17, Step 5: (3R)—3-(4-chlorophenyl)[(4-chIorophenyl)methyl](2-hydroxypropan-Z-
yl)[(1H-pyrazolyl)methoxy]-2,3-dihydro-1H-isoindolone
Et4NF.H20 (149 mg, 1.0 mmol, 10 eq.) and 4 A molecular sieves (50 mg) were added to 2-(4-
ch|orobenzy|)(4-ch|orophenyl)(2-hydroxypropanyl)((1-((2-(trimethylsilyl)ethoxy) methy|)-1H-
lyl)methoxy)isoindolinone (65 mg, 0.1 mmol) in THF (2 mL) and the mixture was heated to
65 °C for 3 h, allowed to cool to r.t., and partitioned between EtOAc (25 mL) and water (3 x 20 mL).
The organic layer was dried (MgSO4) and the solvent removed in vacuo to give a white solid (25 mg,
48%). The enantiomers were separated by chiral HPLC (Daicel Chiralpak IA, 250 x 10mm i.d., 5pm, n-
heptane: 2-propanol 5:1; 4.7 mL/min) to give the title compound (6 mg). 1H NMR (500 MHz; CDCI3) LH
1.63 (6H, 2 x s), 3.66 (1H, d), 3.76 Hz (1H, d), 4.00 (1H, d), 4.72 (1H, d), 7.08-7.27 (9H, m), 7.72 (1H,
dd), 8.03 (1H, dd); HRMS found 522.1334. MH+
Exam ple 18: 1-({[(1R)(4-chlorophenyl)—2-[(4-chlorophenyl)methyl](2-hydroxypropanyl)
oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarbonitrile
NC‘e Cl
Z Stei, Hex Ste, 2 00
EtOZC CN CN
Example 18, Step 1: 1-(Hydroxymethyl)cyclopropanecarbonitrile
LiBH4 (573 mg, 26.3 mmol, 2 eq) was added to ethylcyanocyclopropane carboxylate (1.7 mL, 13.15
mmol, 1 eq.) in THF (18 mL) and the mixture was heated to 50 °C for 18 h. The reaction was cooled to
0 °C and NaHC03 (sat’d aq) was added until gas evolution ceased. The mixture was diluted with brine
(20 mL) and extracted with EtOAc (1 x 60 mL), DCM (3 x 60 mL) and 10% MeOH/DCM (2 x 60 mL).
The organic extracts were combined, dried (MgSO4) and the solvent removed in vacuo to give the title
compound as a clear oil (820 mg, 65%); 1H NMR (500 MHz; CDCI3): 0.96-1.01 (2H, m), .31 (2H,
m), 3.64 (2H, s).
Example 18, Step 2: 1-(((5-Bromo(4-chlorobenzyl)(4-chlorophenyl)oxoisoindolin
yl)oxy)methyl)cyclopropanecarbonitrile
Thionyl de (120 uL, 1.64 mmol, 2 eq.) was added to 6-bromo(4—chlorobenzyl)(4-
chlorophenyl)hydroxyisoindolinone (Preparation 8) (382 mg, 0.82 mmol, 1 eq.) in THF (4 mL)
and the e was stirred at r.t. for 4 h. The solvent was removed in vacuo, the residue re-dissolved
in THF (2 mL), cyanoalcohol (Example 18, Step 1; 160 mg, 1.64 mmol, 2 eq.) in THF (2 mL) was
added and the mixture was stirred at r.t. for 18 h. The mixture was diluted with EtOAc (2 x 30 mL) and
washed with water. The organic layers were combined, dried over MgSO4, and the t removed in
vacuo. The crude mixture was ed by MPLC on SiOz with gradient elution from 0-2% EtZO/DCM to
give the title compound as a clear gum (390 mg, 87%); MS (ES+) 543.3, 545.3 [M+H]+.
Example 18, Step 3 and 4: 1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropane
carbonitrile
Using the product from e 18, Step 2, The title compound was prepared by following
procedures similarto those described in Preparation 13 and Example 2: 1H NMR (500 MHz; CDCI3):
0.35-0.42 (1H, m), 0.42-0.49 (1H, m), .20 (2H, m), 1.62 (2 x 3H, 2 x s), 2.31 (1H, d), 2.91 (1H,
d), 4.02 (1H, d), 4.72 (1H, d), 7.15-7.22 (5H, m), 7.25-7.30 (4H, m), 7.74 (1H, dd), 8.00 (1H, d); MS
(ES+) 521.4, 523.4 [M+H]+.
Example 19: N-{[1-({[(1R)(4-chlorophenyl)[(4-chIorophenyl)methyl](2-hydroxypropan
yl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropyl]methyl}methanesulfonamide
2 NHSOzMe
No NHSOzMe
24 Step1 Step 3 Step 2 Step 4
—> )4 —> —> —>
OH OTBDPS OTBDPS OTBDPS
Ho O
B, \\ b Step5
MesozNH34
OH 0
Example 19, Step 1: 1-(((tert-Butyldiphenylsilyl)oxy)methyl) cyclopropanecarbonitrile
1-(Hydroxymethyl)cyclopropanecarbonitrile (1.1 g, 11.3 mmol), I (2.66 mL, 11.3 mmol, 1 eq.),
imidazole (926 mg, 13.6 mmol, 1.2 eq.) and DMAP (69 mg, 0.57 mmol, 0.05 eq.) were combined in
DCM (15 mL) and stirred at r.t. for 18 h. The e was partitioned between EtOAc (2 X 20 mL) and
water (20 mL), washed with brine, dried (MgSO4) and solvent removed in vacuo. The residue was
purified by MPLC on silica with gradient elution from 2-40% EtOAc/petrol to give the title nd as
a clear oil (2.05 g, 54%); HRMS calc for 01N28i 353.2044, found 353.2034.
Example 19, Step 2: (1-(((tert-Butyldiphenylsilyl)oxy)methyl)cyclopropyl) methanamine
ert-Buty|diphenylsily|)oxy)methy|)cyc|opropanecarbonitrile (220 mg, 0.66 mmol) was dissolved in
MeOH (10 mL) and hydrogenated through a Raney nickel catcart on Thales H-cube at 20 °C, 20 bar
for 3 h, with constant recycling ofthe reaction mixture (1 mL/min flow rate). The solvent was removed
in vacuo and the residue purified by MPLC on silica with gradient elution from 0-20% MeOH/EtOAc to
give the title compound as a clear gum (78 mg, 46%); 1H NMR (500 MHz; CDCI3) 0.30-0.41 (4H, m),
1.06 (9H, s), 2.38 (2H, brs), 2.72 (2H, s), 3.57 (2H, s), .46 (6H, m), 7.62-7.69 (4H, m).
Example 19, Step 3: N-((1-(((tert-ButyldiphenylsiIyl)oxy)methyl)cyclopropyl)methyl)
esulfonamide
MsCl (120 uL, 1.54 mmol, 1.1 eq.) was added to a mixture of (1-(((tertbutyldiphenylsily
|)oxy)methy|)cyc|opropyl) methanamine (480 mg, 1.4 mmol, 1 eq.) and Et3N (236 uL,
1.69 mmol, 1.2 eq.) in DCM (4 mL) at 0 °C and the mixture was stirred at r.t. for 18 h, partitioned
between DCM (2 x 30 mL) and water (20 mL), washed with brine, dried over MgSO4, and the solvent
removed in vacuo. The residue was purified by MPLC on SiOz with a gradient from 10-35%
EtOAc/petrol to give the title compound as a white solid (520 mg, 88%); 1H NMR(500 MHz; CDCI3)
0.36-0.40 (2H, m), 0.49-0.54 (2H, m), 1.08 (9H, s), 2.90 (3H, s), 3.15 (2H, d), 3.54 (2H, s), 4.93 (1H,
m), 7.36-7.51 (6H, m), 7.62-7.68 (4H, m).
Example 19, Step 4: N-((1-(Hydroxymethyl)cyclopropyl)methyl) methanesulfonamide
N-((1-(((tert-Butyldiphenylsilyl)oxy)methyl)cyclopropyl)methyl) esulfonamide (500 mg, 1.2
mmol, 1 eq.) and Et4NF (197 mg, 1.32 mmol, 1.1 eq.) were combined in THF (10 mL) and stirred at r.t.
for 2 h. The mixture was diluted with water (10 mL) and brine (10 mL) and extracted with EtOAc (2 x
mL) and DCM (4 x 20 mL). The organic extracts were combined, dried over MgSO4, and the
solvent removed in vacuo. Purification by MPLC on SiOz with a gradient from 70-100% EtOAc/petrol
gave the title compound as a clear oil (188 mg, 88%); MS ES- 178.1 [M-H]’.
Example 19, Step 5: (((5-Bromo(4-chlorobenzyl)(4-chlorophenyl)oxoisoindolin
yl)oxy)methyl)cyclopropyl)methyl)methanesulfonamide
Thionyl chloride (117 uL, 1.62 mmol, 2 eq.) was added to 6-bromo(4—chlorobenzyl)(4-
chlorophenyl)hydroxyisoindolinone (Preparation 8) (375 mg, 0.81 mmol, 1 eq.) and DMF (1
drop, cat.) in THF (4 mL) and stirred at r.t. for 4 h. The solvent was removed in vacuo, the residue
redissolved in THF (2 mL), and the alcohol in THF (2 mL) was added. The mixture was stirred at r.t. for
96 h, partitioned between EtOAc (2 x 30 mL) and water (20 mL). The organic extracts were combined,
dried over MgSO4, and the solvent removed in vacuo. The crude product was purified by MPLC on
SiOz with a gradient from 25-50% EtOAc/petrol to give the title compound as a clear gum (385 mg,
76%); MS ES- 523.2 [M-H]'.
Example 19, Step 6 and 7: N-{[1-({[(1R)(4-chloropheny|)[(4-chlorophenyl)methyl](2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindol
}methyl)cyclopropyl]methyl}methanesulfonamide
Starting from the product obtained in Step 5; Example 19 was prepared using ures similar to
those described for Preparation 13 and e 2.1H NMR(500 MHz; CDCI3) 0.08-0.17 (2H, m),
0.38-0.47 (2H, m), 1.61 (3H, s), 1.63 (3H, s), 1.83 (1H, s), 2.53 (1H, d), 2.80 (1H, d), 2.84-2.93 (4H,
m), 3.16 (1H, dd), 4.15 (1H, d), 4.30 (1H, t), 4.59 (1H, d), 7.13-7.19 (5H, m), 7.19-7.27 (4H, m), 7.75
(1H, dd), 8.00 (1H, d); MS ES+ 603.4, 605.3 [M+H]+.
Example 20: (3R)(4-chlorophenyl)[(4-ethynylphenyl)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone
2016/053042
Example 20, Step 1: 6-Acetyl(4-chlorophenyl)hydroxy(4—
((triisopropylsilyl)ethynyl)benzyl)isoindolinone
TIPS
Prepared in a similar manner to that bed in Preparation 8 from: 5-acetyI(4-
chlorobenzoy|)benzoic acid (Preparation 19) (1 g, 3.303 mmol), SOCIZ (0.48 mL, 6.607 mmol), (4-
((triisopropylsilyl)ethynyl)phenyl)methanamine (Preparation 42) (1.14 g, 3.964 mmol) and DIPEA
(1.27 mL, 7.27 mmol) in THF (20 mL). Purified by e using 0-30% EtOAc in petrol as the eluent
gave the title compound as a pale yellow solid (1.283 g, 68%). MS:[M+H]+=456.4.
Example 20, Step 2: 6-Acetyl(4-chlorophenyl)((1-(hydroxymethyl)cyclopropyl)methoxy)
(4-((triisopropylsilyl)ethynyl)benzyl)isoindolinone
VA Cl
TIPS
Prepared in a similar manner to that described in Preparation 22 from: 6-acetyl-3—(4-chlorophenyl)
hydroxy(4-((triisopropylsi|y|)ethyny|)benzy|)isoindo|inone (400 mg, 0.70 mmol), SOCIZ (166 mg,
0.10 mL, 1.40 mmol), 1,1-bis(hydroxymethyl)cyc|opropane (104 mg, 0.14 mL, 1.40 mmol) and K2C03
(194 mg, 1.40 mmol) in THF (1.5 mL). Purified by Biotage using 0-40% EtOAc in petrol as the eluent
gave the title compound as a colourless oil (205 mg, 45%); 1H NMR (500 MHz, CDCI3) 0.13-0.17 (2H,
m) 0.39-0.43 (2H, m), 1.11 (21H, 5), 2.67-2.69 (4H, m), 2.77 (1H, d), 3.35 (1H, d), 3.50 (1H, d), 4.21
(1H, d), 4.59 (1H, d), 7.12-7.13 (2H, m), 7.17-7.26 (5H, m), 7.28 (2H, m), 8.14-8.15 (1H, m), 8.43-8.44
(1H, m).
Example 20, Step 3: 3-(4-ChlorophenyI)((1-(hydroxymethyl)cyclopropyl)methoxy)(2-
hydroxypropany|)(4-((triisopropylsilyl)ethyny|)benzyl)isoindo|inone
wk Cl
HO O
TIPS
Prepared in a similar manner to that described in Example 1 from: 6-acetyl(4-chlorophenyl)((1-
(hydroxymethyl)cyc|opropyl)methoxy)(4-((triisopropylsilyl)ethynyl)benzyl)isoindolinone (180 mg,
0.27 mmol), Zn(ll)C|2 (7.5 mg, 0.055 mmol) and MeMgCl (0.23 mL, 3.0 M in THF, 0.17 mmol) in THF
(1.94 mL). The title product was obtained as a white solid (148 mg, 82%); 1H NMR (500 MHz, CDCI3)
0.12-0.18 (2H, m), .43 (2H, m), 1.11 (21H, 5), 1.61 (3H, s), 1.62 (3H, s), 2.66 (1H, d), 2.82 (1H,
d), 3.35 (1H, d), 3.48 (1H, d), 4.19 (1H, d), 4.57 (1H, d), 7.09-7.13 (3H, m),7.17-7.21 (4H, m), 7.26-
7.28 (2H, m), 7.71 (1H, dd), 7.99 (1H, d).
Example 20, Step 4: (3R)(4-chlorophenyl)[(4-ethyny|pheny|)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihyd ro-1 H-isoindolone
ng from 3-(4-chlorophenyl)((1-(hydroxymethyl)cyclopropyl)methoxy)—6-(2-hydroxypropan-2—yl)-
2-(4-((triisopropylsilyl)ethynyl)benzyl)isoindolinone, deprotection using a r procedure to that
described in Example 10 gave Example 20. 1H NMR (500 MHz, CDCI3) 0.09-0.18 (2H, m), 0.37-0.42
(2H, m), 1.61 (3H, s), 1.62 (3H, s), 2.65 (1H, d), 2.82 (1H, d), 3.04 (1H, s), 3.34 (1H, d), 3.47 (1H, d),
4.17 (1H, d), 4.60 (1H, d), 7.11 (1H, d), 7.16-7.22 (6H, m), 7.31 (2H, d), 7.72 (1H, dd), 7.99 (1H, d).
MS:[M+H]+=516.4.
Example 21: -(4-chlorophenyl)[(4-ethyny|phenyl)methyl]fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindolone
Example 21. Step 1: 6-Bromo(4-chloro-phenyl)fluorohydroxy{4-[(triisopropylsilanyl)-
l]-benzyI}-2,3-dihydro-isoindolone
The title compound was prepared from 5-bromo(4-chloro-benzoyl)fluoro-benzoic acid (7.9 g, 22.2
mmol) and 4-[(triisopropylsilanyl)-ethynyl]-benzylamine (7.0 g, 24.4 mmol) in a similar manner to that
described for Preparation 9. 1H NMR (400 MHz, DMSO-d6): 7.84-7.72 (2H, m), 7.59 (1H, s), 7.33-
7.20 (6H, m), 7.12 (2H, d), .37 (1H, m), 4.29 (1H, d), 1.13-1.05 (21H, m).
Example 21. Step 2: 6-Bromo(4-chIoro-phenyl)fluoro(1-hydroxymethyl-
cyclopropylmethoxy){4-[(triisopropylsilanyl)-ethynyl]-benzyl}-2,3-dihydro-isoindolone
The title compound was prepared from 6-bromo(4-chloro-pheny|)f|uorohydroxy{4-
[(triisopropylsilanyl)-ethyny|]-benzy|}-2,3-dihydro-isoindolone (5.0 g, 7.9 mmol) and (1-
hydroxymethyl-cyclopropyl)—methano| (4.1 g, 39.9 mmol) in a similar manner to that described for
Preparation 10. 1H NMR (400 MHz, DMSO-d6): 7.91 (1H, d), 7.83 (1H, dd), 7.35-7.12 (6H, m), 7.03
(2H, d), 4.51-4.27 (3H, m), 3.44-3.33 (2H, m), 2.86 (2H, s), 1.09 (21H, s), 0.41-0.26 (2H, m), 0.26-0.10
(2H, m).
Example 21, Step 3: 6-Acetyl(4-chloro-phenyl)[4-(3,3-diisopropylmethyl-pentynyl)-
benzyl]fluoro(1-hydroxymethyl-cyclopropylmethoxy)-2,3-dihydro-isoindolone
3)4 (84 mg, 0.07 mmol) and LiCl (178 mg, 4.24 mmol) were added to a solution of 6-bromo
(4-chloro-phenyl)-4—fluoro(1-hydroxymethyl-cyclopropylmethoxy){4-[(triisopropylsilany|)-ethyny|]-
benzy|}-2,3-dihydro-isoindolone (1.0 g, 1.4 mmol) in dioxane/toluene (1:1, 20 mL) under N2 and the
resulting solution was ed for 10 minutes. Tributyl(1-ethoxyvinyl)tin (475 pL, 1.41 mmol) was
added and the reaction was stirred at 110°C for 1.5 hours. The reaction e was cooled to room
tempearature, quenched with NaHC03 and extracted with EtOAc. The organic phase was dried over
NaZSO4, filtered and concentrated in vacuo. The crude material was columned (gradient 0-50% EtOAc
in Petrol) to give 900 mg ofa pale yellow solid which was dissolved in e (10 mL) followed by 2M
HCI (6 mL). The solution was stirred at room temperature for 1 hour, quenched with NaHCOs and
extracted with DCIVI. The organic phase was dried over NaZSO4, filtered and concentrated in vacuo to
give the desired t as a yellow solid. 1H NMR (400 MHz, DMSO-d6): 8.21 (1H, d), 7.95 (1H, dd),
7.37-7.10 (6H, m), 7.06 (2H, d), 4.51-4.28 (3H, m), .33 (2H, m), 2.94-2.77 (2H, m), 2.69 (3H, s),
1.08 (21H, s), 0.41-0.25 (2H, m), 0.23-0.10 (2H, m).
Example 21. Step 4: 3-(4-Chloro-phenyI)fluoro(1-hydroxymethyl-cyclopropylmethoxy)(1-
hydroxymethyl-ethyl){4-[(triisopropylsilanyl)-ethynyl]-benzyl}-2,3-dihydro-isoindolone
The title compound was prepared from yl(4-chloro-phenyl)[4-(3,3-diisopropylmethylpentynyl
)—benzyl]fluoro(1-hydroxymethyl-cyclopropylmethoxy)-2,3-dihydro-isoindolone (871
mg, 1.29 mmol) in a similar manner to that described for Example 1. 1H NMR (400 MHz, 6):
7.80 (1H, d), 7.49 (1H, d), 7.36-7.10 (6H, m), 7.05 (2H, d), 5.36 (1H, s), 4.50-4.25 (3H, m), 3.42-3.32
(2H, m), 2.91-2.75 (2H, m), 1.48 (6H, s), 1.09 (21H, s), 0.40-0.25 (2H, m), 0.15 (2H, s).
Example 21. Step 5: (3R)(4-chlorophenyl)[(4-ethynylphenyl)methyl]fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindolone
The title compound was prepared by following pocedures similar to those described for Example 11.
Purification by ative chiral HPLC gave the title compound (173 mg). 1H NMR (400 MHz, DMSO-
d5): 7.80 (1H, d), 7.50 (1H, d), 7.37-7.17 (6H, m), 7.09 (2H, d), 5.36 (1H, s), 4.50-4.35 (2H, m), 4.28
(1H, d), 4.11 (1H, s), 3.38 (1H, dd), 3.28 (1H, dd), 2.89 (1H, d), 2.77 (1H, d), 1.48 (6H, s), 0.39-0.24
(2H, m), 0.18-0.00 (2H, m). MS:[M+H]+=534.
Examples 22 and 23: (3R)(4-chlorophenyl)(1,2-dihydroxypropan-Z-yl)[(4-
ethynylphenyl)methyl]fluoro{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-
isoindolone
(*Two isomers at the position shown)
Example 22 and Example 23. Step 1: 3-(4-ChIoro-phenyl)fluoro(1-hydroxymethylcyclopropylmethoxy
)isopropenyl{4-[(triisopropylsilanyl)-ethynyl]-benzyl}-2,3-dihydro-
isoindolone
The title compound was prepared in a similar fashion to Preparation 13. 1H NMR (400 MHz, DMSO-
d6): 7.79 (1H, d), 7.63-7.55 (1H, m), 7.34-7.09 (6H, m), 7.05 (2H, d), 5.68 (1H, s), 5.30 (1H, s), 4.49-
4.28 (3H, m), 3.36 (2H, d), 2.87 (2H, s), 2.17 (3H, s), 1.16-1.03 (21H, m), 0.42-0.26 (2H, m), 0.17 (2H,
Example 22 and Example 23. Step 2: 3-(4-Chloro-phenyl)(1,2-dihydroxymethyl-ethyl)
(1-hydroxymethyl-cyclopropylmethoxy){4-[(triisopropylsilanyl)-ethynyl]-benzyl}-2,3-
dihydro-isoindolone
The title compound was prepared by using a similar procedure to that described in Example 27 Step
4. 1H NMR (400 MHz, DMSO-d6): 7.79 (1H, dd), 7.50-7.42 (1H, m), 7.35-7.10 (6H, m), 7.05 (2H, d),
.28 (1H, s), 4.86-4.79 (1H, m), 4.42 (1H, t), 4.35 (2H, s), 3.54-3.40 (2H, m), 3.36 (2H, dd), 2.93-2.78
(2H, m), .40 (25H, m), 1.43 (3H, s), 1.14-1.03 (21H, m), 0.40-0.27 (2H, m), 0.21-0.09 (2H, m).
Example 22 and Example 23. Step 3: (3R)(4-chlorophenyl)(1,2-dihydroxypropanyl)[(4-
ethynylphenyl)methyl]fluoro{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-
isoindolone r A and B)
The title compounds were prepared from 3-(4-chloro-phenyI)(1,2-dihydroxymethyI-ethyI)fluoro-
3-(1-hydroxymethyI-cyclopropylmethoxy){4-[(triisopropylsilanyI)-ethynyI]-benzy|}-2,3-dihydro-
oIone (1.0 g, 1.4 mmol) in a similar manner to that described in Example 10. Purification by
preparative chiral HPLC gave:-
e 22 (*isomer1): 1H NMR (400 MHz, DMSO-d6): 7.78 (1H, s), 7.47 (1H, d), .16 (6H,
m), 7.09 (2H, d), 5.29 (1 H, s), 4.82 (1H, t), 4.50-4.32 (2H, m), 4.28 (1H, d), 4.12 (1H, s), 3.52-3.34 (3H,
m), 3.29 (1H, dd), 2.89 (1H, d), 2.78 (1H, d), 1.43 (3H, s), 0.38-0.25 (2H, m), 0.17-0.00 (2H, m). MS:
[M-H]'=548.
Example 23 )*isomer 2): 1H NMR (400 MHz, DMSO-d6): 7.79 (1H, d), 7.46 (1H, d), .13 (6H,
m), 7.09 (2H, d), 5.42-5.12 (1H, m), 4.98-4.60 (1H, m), 4.41 (1H, d), 4.29 (1H, d), 4.12 (1H, s), 3.50
(1H, d), 3.44 (1H, d), 3.38 (2H, d), 3.29 (1H, d), 2.89 (1H, d), 2.79 (1H, d), 1.43 (3H, s), 0.39-0.29 (2H,
m), 0.18-0.00 (2H, m)
Exam ple 24: 4-{[(1R)(4-chlorophenyl)fluoro({1-
[hydroxy(2Hz)methyl]cycIopropyl}(2H2)methoxy)(2-hydroxypropanyl)oxo-2,3-dihydro-1H-
isoindolyl]methyl}benzonitrile
2016/053042
Example 24. Step 1: 4-[5-Bromo(4-chloro-phenyl)fluorohydroxyoxo-1,3-dihydro-
isoindo|ylmethyl]-benzonitrile
The title compound was prepared from 5-bromo(4-ch|oro-benzoy|)fluoro-benzoic acid (4.0 g, 11.2
mmol) and 4-aminomethyl-benzonitrile hydrochloride (2.0 g, 11.2 mmol) in a similar manner to that
described for ation 9 MS: [M-H]'=470.
Exam ple 24. Step 2: 4-{[5-Bromo(4-chlorophenyl)fluoro({1-
[hydroxy(2H2)methyl]cyclopropyl}(2H2)methoxy)oxo-2,3-dihydro-1H-isoindol-Z-
yl]methyl}benzonitrile
The title compound was prepared from 4—[5-bromo(4-ch|oro-pheny|)f|uorohydroxyoxo-1,3-
dihydro-isoindolylmethyI]-benzonitrile (1.0 g, 2.1 mmol) and {1 -
[hydroxy(2H2)methyl]cyclopropy|}(2H2)methanol (890 mg, 8.4 mmol) in a similar manner to that
described for Preparation 10 MS: [M-H]’=558.
Exam ple 24. Step 3: 4-{[1-(4-Chlorophenyl)fluoro({1-
[hydroxy(2H2)methyl]cyclopropyl}(2H2)methoxy)oxo(propenyl)-2,3-dihydro-1H-
isoindolyl]methyl}benzonitrile
The title compound was prepared from bromo(4-ch|oropheny|)f|uoro({1-
[hydroxy(2H2)methyl]cyclopropy|}(2H2)methoxy)oxo-2,3-dihydro-1H-isoindolyl]methy|}benzonitrile
(750 mg, 1.33 mmol) in a r mannerto that described in Preparation 13. MS: [M-H]'=519.
Exam ple 24. Step 4: 4-{[(1R)(4-chlorophenyl)—7-fluoro({1-
[hydroxy(2H2)methyl]cycIopropyl}(2H2)methoxy)(2-hydroxypropany|)oxo-2,3-dihydro-1H-
isoindolyl]methyl}benzonitrile
The title compound was prepared from 4—{[1-(4-ch|oropheny|)f|uoro({1-
[hydroxy(2H2)methyl]cyclopropy|}(2H2)methoxy)oxo(propenyl)-2,3-dihydro-1H-isoindoI
yl]methyl}benzonitrile (660 mg, 1.27 mmol) in a similar manner to that described for Example 2. 1H
NMR (400 MHz, DMSO-d6): 7.81 (1H, d), 7.61 (2H, d), 7.57-7.46 (1H, m), 7.34-7.15 (6H, m), 5.37 (1H,
s), 4.44 (2H, s), 4.40-4.30 (1H, m), 1.48 (6H, s), 0.39-0.26 (2H, m), 0.21-0.03 (2H, m). MS: [M-H]'
=537.
Exam ple 25: R)(4-chlorophenyI){[1-(hydroxymethyl)cyclopropyl]methoxy}(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile
Example 25, Step 1: 6-Acetyl(4-bromobenzyl)(4-chlorophenyl)hydroxyisoindolinone
Prepared in a similar manner to that described in Preparation 9 from: 5-acetyl(4-
chlorobenzoyl)benzoic acid. MS:[M-H]'=470.2.
Exam ple 25, Step 2: 6-Acetyl(4-bromobenzyl)(4-chlorophenyl)((1-
(hydroxymethyl)cyclopropyl)methoxy)isoindolinone
DOgOC.
0 5b
Prepared in a similar mannerto that described in Preparation 22 from: 6-acetyl(4-bromobenzy|)
(4-chlorophenyl)hydroxyisoindolinone. 1H NMR (500 MHz, CDCI3) 0.09-0.18 (2H, m), 0.40-0.44
(2H, m), 2.67-2.69 (4H, m), 2.78 (1H, d), 3.36 (1H, d), 3.50 (1H, d), 4.19 (1H, d), 4.55 (1H, d), 7.07-
7.08 (2H, m), 7.17-7.18 (2H, m), .26 (3H, m), 7.30-7.32 (2H, m), 8.14 (1H, dd), 8.43 (1H, d).
2016/053042
Example 25, Step 3: 2-(4-Bromobenzyl)(4-chlorophenyl)—3-((1-
xymethyl)cyclopropyl)methoxy)(2-hydroxypropanyl)isoindolinone
Prepared in a similar manner to that described in Example 1 from: 6-acetyl(4—bromobenzyI)(4-
chlorophenyI)((1-(hydroxymethyl)cyclopropyl)methoxy)isoindolinon. 1H NMR (500 MHz, CDCI3)
0.12-0.20 (2H, m), 0.39-0.44 (2H, m), 1.61 (3H, s), 1.62 (3H, s), 2.66 (1H, d), 2.83 (1H, d), 3.37 (1H,
d), 3.48 (1H, d), 4.17 (1H, d), 4.52 (1H, d), 7.07-7.11 (3H, m), 7.17-7.22 (4H, m), 7.29-2.31 (2H, m),
7.72 (1H, dd), 7.99 (1H, d).
Example 25, Step 4: 4-{[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}
(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindoIyl]methyl}benzonitrile
In a microwave vial, a mixture of 2-(4-bromobenzyI)(4-ch|orophenyl)-3—((1-
(hydroxymethyl)cyclopropyl)methoxy)(2-hydroxypropanyl)isoindolinone (140 mg, 0.25 mmol),
zinc powder (3.3 mg, 0.05 mmol), Pd(OAc)2 (11.2 mg, 0.05 mmol), rac(di-tert-butylphosphino)-1,1’-
binapthyl (39.8 mg, 0.10 mmol) and Zn(CN)2 (32.3 mg, 0.275 mmol) in MeCN (1.5 mL) was ed
under N2 for 20 min then heated to 120 °C for 1h. The reaction was cooled to RT, diluted with EtOAc
(25 mL) and filtered through Celite. The organic layer was washed with water (100 mL), brine (100
mL), dried over NaZSO4 and concentrated under vacuum. Purified by Biotage 0-50% EtOAc in petrol
as the eluent then semi-preparative HPLC gave the racemic mixture as a white solid (55 mg, 43%).
Purification by preparative chiral HPLC gave the title compound as a white solid (24.4 mg). 1H NMR
(500 MHz, CDgoD-d4) 0.16-0.29 (2H, m), .46 (2H, m), 1.58 (6H, s), 2.84-2.90 (2H, m), 3.48-3.56
(2H, m), 4.44 (1H, d), 4.64 (1H, d), 7.15-7.21 (5H, m), 7.26-7.28 (2H, m), 7.47-7.49 (2H, m), 7.78 (1H,
dd), 8.04 (1H, d). MS:[M+H]+=517.4.
e 26: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyI](2-hydroxypropanyl)[(3-
methyloxetanyl)methoxy]-2,3-dihydro-1H-isoindolone
0%001O
Example 26, Step 1: 2-(Bromomethyl)methyIpropane-1,3-diol
OH Br
H054
At -10 °C, to a solution of 3-methyloxetanemethano| (1.00 g, 9.8 mmol) in THF (12.3 mL) was
added aqueous HBr (48 wt%, 3.9 mL) and the resulting yellow/brown solution was stirred for 4 h at -10
°C then at RT for 12 h. The mixture was diluted with brine (100 mL) and extracted into EtZO (4 x 100
mL). The combined organic layers were dried over NaZSO4 and concentrated under vacuum to give
the title compound as a white solid (1.36 g, 76%); 1H NMR (500 MHz, CDClg) 0.91 (3H, s), 2.73 (2H,
s), 3.53 (2H, s), 3.65 (4H, s).
Example 26, Step 2: 6-Bromo(3-bromo(hydroxymethyl)methylpropoxy)—2-(4-
chlorobenzyl)(4-chlorophenyl)isoindolinone
Prepared in a similar manner to that described in Preparation 22 from: 6-bromo-2—(4-ch|orobenzyl)
(4-chloropheny|)hydroxyisoindolinone (Preparation 9) (635 mg, 1.37 mmol), SOCIZ (325 mg,
0.20 mL, 2.73 mmol), momethyl)-2—methy|propane-1,3-diol (500 mg, 2.73 mmol) and K2C03
(377 mg, 2.73 mmol) in THF (2.9 mL). ed by Biotage using 0-20% EtOAc in petrol as the eluent
gave the title compound as a white solid (302 mg, 35%). H]+=628.3.
Example 26, Step 3: 6-Bromo(4-chlorobenzyl)(4-chlorophenyl)((3-methyloxetan
yl)methoxy)isoindolinone
0% c.
To a on of 6-bromo(3-bromo-2—(hydroxymethyl)methy|propoxy)(4-chlorobenzyl)(4-
chlorophenyl)isoindolinone (820 mg, 1.31 mmol) in EtOH (28 mL) was added KOH (88 mg, 1.57
mmol) and the mixture heated at reflux for6 h then cooled to RT. The reaction was diluted with water
(50 mL) and acidified to pH 6 with 1.0 M aqueous HCI solution. The reaction was extracted into EtOAc
(3 X 40 mL), washed with brine, dried over M9804 and concentrated under vacuum. Purified by
Biotage using 0-20% EtOAc in petrol as the eluent gave the title compound as a white solid (516 mg,
72%). 1H NMR (500 MHz, CDCI3) 1.13 (3H, s), 2.75-2.81 (2H, m), 4.16-4.22 (2H, m), 4.26-4.27 (1H,
m), 4.32-4.34 (2H, m), 4.51 (1H, d), 6.97 (1H, d), 7.07-7.09 (2H, m), 7.13-7.14 (2H, m), 7.17-7.23 (4H,
m), 7.65 (1H, dd), 8.06 (1H, d).
Example 26, Step 4: 2-(4-Chlorobenzy|)(4-chloropheny|)((3-methy|oxetany|)methoxy)
(propeny|)isoindo|inone
0% CI
Prepared in a similar to that described in Preparation 13 from: 6-bromo(4-ch|orobenzyl)
orophenyl)((3-methyloxetanyl)methoxy)isoindo|inone. 1H NMR (500 MHz, CDCI3) 1.13
(3H, s), 2.20 (3H, s), 2.79 (2H, s), 4.16-4.27 (3H, m), 4.32-4.35 (2H, m), 4.52-4.55 (1H, m), 5.21 (1H,
s), 5.48 (1H, s), 7.04 (1H, d), 7.09-7.14 (4H, m), 7.17-7.23 (4H, m), 7.63 (1H, dd), 8.00 (1H, d).
Example 26, Step 5: yl(4-chlorobenzyl)(4-chIoropheny|)((3-methy|oxetan
yI)methoxy)isoindolinone
0% CI
Prepared in a r manner to that described in Preparation 15 from: 2-(4-chlorobenzyl)(4-
chlorophenyl)((3-methyloxetanyl)methoxy)(propenyl)isoindo|inone. MS:[M+H]+=510.4.
Example 26, Step 6: (3R)(4-ch|oropheny|)[(4-chloropheny|)methy|](2-hydroxypropan
yI)[(3-methy|oxetanyl)methoxy]-2,3-dihydro-1H-isoindoIone
Prepared in a similar manner to that described in Example 1 from: 6-acetyl(4—chlorobenzyl)(4-
chlorophenyl)((3-methyloxetanyl)methoxy)isoindolinone. 1H NMR (500 MHz, CDCI3) 1.13 (3H,
s), 1.63 (3H, s), 1.64 (3H, s), 2.78 (2H, s), 4.16-4.22 (2H, m), .27 (1H, m), 4.34-4.36 (2H, m),
4.54 (1H, d), 7.60 (1H, d), 7.10-7.14 (4H, m), 7.19-7.22 (4H, m), 7.73 (1H, dd), 8.02 (1H, d).
MS:[M+H]+=526.4.
Examples 27 and 28: 4-{[(1R)(4-chlorophenyl)(1,2-dihydroxypropanyl){[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile
(*both isomers at position shown)
Example 27 and e 28, Step 1: 4-((5-Bromo(4-chlorophenyl)hydroxy
oxoisoindolinyl)methyl)benzonitrile
Prepared in a similar manner to that described in Preparation 9 from: 5-bromo-2—(4-
chlorobenzoy|)benzoic acid and 4-(aminomethyl)benzonitrile. MS:[M-H]'=453.1.
Example 27 and Example 28, Step 2 and 3: 4-((1-(4-Chlorophenyl)((1-
(hydroxymethyl)cyclopropyl)methoxy)oxo(propenyl)isoindolin
yl)methyl)benzonitrile
9» or
Starting from : 4-((5-bromo(4-ch|orophenyI)hydroxyoxoisoindo|iny|)methy|)benzonitrile, the
title nd was prepared in a similar mannerto that described in ation 12 and 13. 1H NMR
(500 MHz, CD30D) 0.19-0.27 (2H, m), 0.41-0.46 (2H, m), 2.21 (3H, s), 2.85-2.91 (2H, m), 3.49-3.56
(2H, m), 4.45 (1H, d), 4.64 (1H, d), 5.23 (1H, s), 5.51 (1H, s), 7.15-7.22 (5H, m), 7.26-7.28 (2H, m),
7.47-7.48 (2H, m), 7.76 (1H, dd), 7.99 (1 H, d).
e 27 and Example 28, Step 4: 4-{[(1R)(4-chlorophenyl)(1,2-dihydroxypropan-Z-yl)
{[1-(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindol-Z-
yl]methyl}benzonitrile
At 0 °C, to a solution of tert-butanol/water (8.91 mL/8.91 mL) was added AD-mix-B (2.49 g) followed by
portion wise addition of 4-((1-(4-ch|oropheny|)((1-(hydroxymethy|)cyc|opropyl)methoxy)oxo
(propeny|)isoindo|iny|)methy|)benzonitrile (900 mg, 1.80 mmol) and the resulting mixture was
stirred at 0 °C for 48 h. Nazsos (0.98 g, 7.78 mmol) was added and the reaction warmed to RT and
diluted with water (50 mL). The reaction was extracted with EtOAc (2 x 50 mL), dried over MgSO4 and
concentrated under . Purified by Biotage using 0-100% EtOAc in petrol as the eluent gave a
diastereoisomeric mixture of product (177 mg, 18%). Chiral HPLC gave the title compounds:.
Example 27 er1): 1H NMR (500 MHz, 00013) 0.13-0.21 (2H, m), 0.42-0.46 (2H, m), 1.58 (3H,
s), 2.64 (1H, s), 2.72 (1H, d), 2.88 (1H, d), 3.40 (1H, d), 3.51 (1H, d), 3.71 (1H, d), 3.82 (1H, d), 4.41
(1H, d), 4.50 (1H, d), 7.14-7.18 (5H, m), 7.26-7.28 (2H, m), 7.44-7.46 (2H, m), 7.72 (1H, dd), 7.99 (1H,
d). MS:[M+H]*=533.4.
Example 23 (*iomers 2): 1H NMR (500 MHz, CDCI3) .21 (2H, m), 0.42-0.47 (2H, m), 1.59 (3H,
s), 2.64 (1H, s), 2.73 (1H, d), 2.86 (1H, d), 3.41 (1H, d), 3.51 (1H, d), 3.70 (1H, d), 3.81 (1H, d), 4.41
(1H, d), 4.50 (1H, d), 7.14-7.18 (5H, m), 7.27-7.23 (2H, m), 7.44-7.46 (2H, m), 7.72 (1H, dd), 7.93 (1H,
d). MS:[M+H]*=533.4.
Example 29: -(4-chlorophenyl)[(4-chlorophenyl)methyl][(1-
hydroxycyclopropyl)methoxy](2-hydroxypropanyl)—2,3-dihydro-1H-isoindolone
Example 29, Step 1: 6-Acetyl((1-((tert-butyldiphenylsilyl)oxy)cyclopropyl)methoxy)(4-
chlorobenzyl)(4-chlorophenyl)isoindolinone
OTBDPS
Prepared in a similar manner to that described in Preparation 12 from: 6-acetyl-2—(4-chlorobenzyl)
(4-chlorophenyl)hydroxyisoindolinone (Preparation 20) and (1-((tert-
butyldiphenylsilyl)oxy)cyclopropyl)methanol). 1H NMR (500 MHz, CDCI3) 0.07-0.12 (2H, m), 0.66-0.75
(2H, m), 0.99 (9H, s), 2.57 (1H, d), 2.67 (3H, s), 2.76 (1H, d), 4.24 (2H, s), 6.83 (1H, d), 6.97-6.98 (2H,
m), 7.06-7.14 (5H, m), 7.25-7.31 (5H, m), 7.37-7.40 (2H, m), 7.56-7.58 (2H, m), 7.65-7.66 (2H, m),
7.99 (1H, dd), 8.34 (1 H, d).
Example 29, Step 2: 3-((1-((tert-Buty|diphenylsiIyl)oxy)cyc|opropy|)methoxy)(4-chIorobenzyl)-
3-(4-chlorophenyl)(2-hydroxypropanyl)isoindolinone
OTBDPS
HO 0
Prepared in a similar manner to that bed in Example 1. 1H NMR (500 MHz, CDCI3) 0.05-0.11
(2H, m), 0.62-0.72 (2H, m), 1.00 (9H, s), 1.61 (6H, s), 2.56 (1H, d), 2.84-2.86 (1H, m), 4.20-4.27 (2H,
m), .77 (1H, m), .98 (2H, m), 7.04-7.06 (2H, m), .13 (4H, m), 7.28-7.32 (4H, m),
7.37-7.41 (2H, m), 7.57-7.61 (3H, m), 7.66-7.68 (2H, m), 7.91 (1H, d).
Example 29, Step 3: (3R)—3-(4-chlorophenyI)[(4-chlorophenyl)methyI][(1 -
hydroxycyc|opropyl)methoxy](2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone
Deprotection in a similar mannerto that described for Example 10 gave Example 29.. 1H NMR (500
MHz, CDCI3) 0.19-0.26 (2H, m), 0.70-0.76 (2H, m), 1.61-0.62 (6H, m), 2.71 (1H, d), 2.94 (1H, d), 4.20
(1H, d), 4.54 (1H, d), 7.10 (1H, d), 7.13 (4H, s), 7.20-7.24 (4H, m), 7.71 (1H, dd), 7.99 (1H, d).
MS:[M+H]+=512.4.
Example 30: R)(4-Ch|orophenyl)[(4-chlorophenyl)methyI](2-hydroxypropanyI)
oxo-2,3-dihydro-1H-isoindoly|]oxy}-N,N-dimethylacetamide
Example 30, Step 1: Methyl 2-((5-bromo(4-chlorobenzyl)(4-chlorophenyl)oxoisoindolinyl)oxy)acetate
Using methyl glycolate, the title compound was prepared using a procedure similar to that described
for Preparation 10. MS: [M-C3,H503,]+ 446.
Example 30, Step 2: Bromo(4-chlorobenzyl)(4-chlorophenyl)oxoisoindolin
yl)oxy)-N,N-dimethylacetamide
Methyl 2-((5-bromo(4-ch|orobenzy|)(4-chIorophenyI)oxoisoindoliny|)oxy)acetate (756 mg,
1.41 mmol) and 40% aqueous in water dimethylamine (7.6 mL) were mixed and stirred at room
temperature for 6.5 h. The solvent was d in vacuo and FCC [dichloromethane-methanol
(100:0)—>(94:6)] of the crude residue afforded 2—((5-bromo(4-ch|orobenzy|)(4-ch|oropheny|)
oxoisoindoliny|)oxy)-N,N-dimethylacetamide (524 mg, 68%) as a white foam. MS: [M-C4H8N02]+
446.
Example 30, Step 3: (R)((2-(4-chlorobenzyl)(4-chlorophenyl)(2-hydroxypropan-Z-yl)
oxoisoindoliny|)oxy)-N,N-dimethylacetamide
The title compound was prepared using similar procedures to those described in ation 13 and
Example 2. 1H NMR (500 MHz, : 7.94 (1H, d, 7-H), 7.62 (1H, dd, ArH), 7.35-7.30 (2H, m, 2 x
ArH), 7.24-7.18 (4H, m, 4 xArH), 7.12-7.07 (3H, m, 3 xArH), 4.74 (1H, d, NC-H’), 3.87 (1H, d, NC-H),
3.20 (1H, d, 4'-H’), 3.08 (1H, d, 4’-H), 2.76 (3H, s, NCH3), 2.37 (3H, s, NCH3) and 156-152 (6H, m, 2
x CH3). MS: [M-C4H8NOZ]+ 424.
Example 31: (3R)(4-Chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropan-Z-yl){[1-
(methoxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone
Example 31, Step 1: 2-(4-Chlorobenzyl)(4-chlorophenyl)—3-((1-
(methoxymethyl)cyc|opropyl)methoxy)(propenyl)isoindolinone
To a solution of 2-(4-chlorobenzyI)(4-chlorophenyI)((1-(hydroxymethyl)cyclopropyl)methoxy)
(propenyl)isoindolinone, Preparation 13 (318 mg, 0.625 mmol) in anhydrous THF (3 mL)
under nitrogen, was added tBuOK (140 mg, 1.25 mmol) at room ature and the mixture stirred
for 1 h before cooling to 0 °C. Mel (0.08 mL, 1.25 mol) in anhydrous THF (1 mL) was added dropwise
and stirred for 3.5 h. Diluted with EtOAc (10 mL) and washed with water (2 x 20 mL), brine (20 mL),
dried over anhydrous NaZSO4, filtered and the t removed n vacuo. FCC [petrol-ethyl e
(100:0)—>(80:20)] of the crude residue afforded 2-(4-chlorobenzyI)(4-chlorophenyI)((1-
(methoxymethyl)cyclopropyl)methoxy)(propenyl)isoindolinone (278 mg, 85%) as a
colourless gum. MS: [C6H11OZ]+ 406.
Example 31, Step 2: (R)—2-(4-Chlorobenzyl)(4-chIorophenyl)(2-hydroxypropan-Z-yI)((1-
(methoxymethyl)cyclopropyl)methoxy)isoindolinone
The title compound was ed using similar ures to those described for Example 2.1H NMR
(500 MHz, CDCI3) 8.00 (1H, m, 7-H), 7.73 (1H, dd, ArH), 7.24-7.17 (4H, m, 4 xArH) 7.16-7.10 (4H, m,
4 xArH), 7.08 (1H, d, ArH), 4.46 (1H, d, NC-H’), 4.30 (1H, d, NC-H), 3.43 (1H, d, 4’-H’), 3.31 (3H, s,
OCH3), 3.16 (1H, d, 4'-H), 2.88 (1H, d, 2’-H’), 2.61 (1H, d, 2’-H), .62 (6H, m, 2 x CH3), 0.45-0.38
(2H, m, Cy-Py—Hz) and 0.25-0.15 (2H, m, Cy-Py-H). MS: [M-C6H1102]+ 424.
Example 32: (3R)(4-Chlorophenyl)[(4-chlorophenyl)methyl]{[1-(hydroxymethyl)
cyclobutyl]methoxy}(2-hydroxypropanyI)-2,3-dihydro-1H-isoindolone
Example 32, Step 1: Cyclobutane-1,1-diyldimethanol
HO, Q ,OH
To a solution ofdiethyl-1,1-cyclobutanedicarboxylate (1.00 g, 4.99 mmol) in anhydrous THF (13 mL) at
0 °C was added LiBH4 (239 mg, 11.0 mmol) portionwise over 10 min and then stirred for further 10 min
before heating to 60 °C for 2 h. The reaction mixture was cooled to room temperature and then to 0
°C. Water (2.5 mL) was added cautiously, followed by 1 M HCI until gas evolution ceased, and then
neutralised with sat. sol. NaOH. Extracted with EtOAc (3 x 10 mL) and then combined. To the
aqueous layer was added brine (5 mL) and then extracted with EtOAc (2 x 10 mL). The combined
c phases were washed with brine (15 mL), dried (NaZSO4), filtered and concentrated in vacuo to
give the title compound as a colourless thick oil (523 mg, 90%), which was used without purification.
1H NMR (500 MHz, CDCI3) 3.75 (4H, s, 2 x HCZOH), 2.32 (2H, br s, 2 x OH), .90 (2H, m, H-3)
and 1.80-1.77 (4H, m, H-2, H-4).
Example 32, Step 2: (R)(4-chlorobenzyl)(4-chlorophenyl)((1-
(hydroxymethyl)cyclobutyl)methoxy)—6-(2-hydroxypropanyl)isoindolinone
Starting from Preparation 8, the title compound was prepared using ures similar to those
described in Preparation 10, Preparation 13 and Example 2. 1H NMR (500 MHz, CDCI3) 8.01 (1H, d,
7-H), 7.72 (1H, dd, ArH) .11 (8H, m, 8 xArH), 7.08 (1H, d, ArH), 4.52 (1H, d, NC-H’), 4.23 (1H,
d, NC-H), 3.53 (2H, d, OCHz), 2.88 (1H, d, CHZOH), 2.79 (1H, d, CHZOH) and 1.86-1.51 (12H, m, 2 x
CH3 and 3 x CH2). MS: [M- C6H11OZ]+ 424.
Example 33: 5-chloro{[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}
(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindoIyl]methyl}benzoic acid
2016/053042
0: (I.‘ O
O i 2
Example 33, Step 1; (2-Bromochlorophenyl)methanamine
To a solution of 2-bromochlorobenzonitrile (500 mg, 2.3 mmol), in dry THF (50 mL) was added
slowly borane-THF complex (1 M, 12 mL, 11.5 mmol) at 0 °C before ing for 1 h. After cooling
down, 1 M HCI in lVleOH (20 mL) was d slowly with ice cooling. The solvent was removed by
concentration in vacuo before water (0.61 mmol/mL to benzonitrile) was charged, then washed by
EtZO (0.61 mmol/mL to benzonitrile) before basifying with 2 M NaOH solution to pH 12. EtZO (15 mL)
was added and the mixture was washed with water (3 x 15 mL) and brine (1 mL). The organic phase
was dried (MgSO4) and concentrated in vacuo to give a yellow oil (345 mg, 68%). LCMS (ESP) m/z =
220.1 [M+H]+.
Example 33, Step 2; 6-Acetyl(2-bromochlorobenzyl)(4-chlorophenyl)—3-
hydroxyisoindolinone
0 N Br
0 0
The title compound was prepared using procedures similar to those described for Preparation 9.
LCMS (ESI‘) m/z = 502.1 [M-H]'
Example 33, Step 3; 6-Acetyl(2-bromochlorobenzyl)(4-chlorophenyl)((1-
(hydroxymethyl)cyclopropyl)methoxy)isoindolinone .
The title compound was prepared using procedures similar to those described for ation 10.
LCMS (ES|+) m/z = 588.3 [M+H]+.
Example 33, Step 4; 6-Acetyl(2-bromochlorobenzyl)((1-(((tert-
butyldimethylsilyI)oxy)methy|)cyc|opropy|)methoxy)(4-chlorophenyl)isoindolinone
TBDMSO
0 ~ .
O 0
The title compound was prepared using procedures similar to those described for Preparation 29.
LCMS (ESP) m/z = 486.2 [M]+.
Example 33, Step 5, 2-((5-Acety|((1-(((tertbutyldimethylsi
|y|)oxy)methy|)cyclopropyl)methoxy)(4-chlorophenyl)oxoisoindolin
y|)methy|)chlorobenzoic acid
TBDMSO
To a solution of the us compound (306 mg, 0.44 mmol) in dry DMF (4.0 mL) was added
os (104 mg, 0.18 mmol), Pd(OAc)2 (20 mg, 0.09 mmol), HCOOLi-HZO (122 mg, 1.74 mmol)
and Eth (0.25 mL, 1.74 mmol) before degassing for 15 mins, and then A020 (0.17 mL, 1.74 mmol)
was introduced before microwave for 30 mins at 140 °C. The mixture was gone through a plug of
Celite, EtOAc (30 mL) was added and the mixture was washed with water (3 x 30 mL) and brine (20
mL). The organic phase was dried (M9804) and concentrated in vacuo. Chromatography (silica;
EtOAc with 0.1% acetic acid, petrol 20 — 80%) gave a greasy solid (78 mg, 43%). LCMS (ESI') m!z =
666.3 [M-H]‘.
Example 33, Step 6; 2-((1-((1-(((tert-Buty|dimethylsi|y|)oxy)methy|)cyclopropy|)methoxy)(4-
chIorophenyl)(2-hydroxypropan-Z-yI)oxoisoindolinyl)methyI)chlorobenzoic acid
TBDMSO
The title compound was prepared using procedures similarto those described for Example 1:. LCMS
(ESI‘) m/z = 682.4 [M-H]'.
Example 33, Step 7: 5-chloro{[(1R)(4-chloropheny|){[1-
(hydroxymethyl)cyc|opropyl]methoxy}(2-hydroxypropany|)oxo-2,3-dihydro-1H-isoindol-
ethyl}benzoic acid
The product from Example 33, Step 6 was deprotected using procedures similar to those described
for Example 10,to give Example 33. 1H-NMR (500 MHz, CDCI3) 6 0.26-0.30 (1H, m, cyclopropane
CHHCHZ), 0.36-0.40 (1H, m, cyclopropane CHHCHZ), 0.50-0.59 (2H, m, cyclopropane CHZCHZ), 1.63
(6H, s, CH3), 2.50 (1H, d, C-O-CHH), 3.29 (1H, d, C-O-CHH), 3.45 (1H, d, CHHOH), 3.86 (1H, d,
CHHOH), 5.01 (1H, d, N-CHH), 5.19 (1H, d, N-CHH), 7.08 (2H, ,d, H-Ar), 7.12 (1H, d, H-Ar), 7.16 (2H,
d, H-Ar), .26 (2H, m, H-4 and H-Ar), .77 (2H, m, H-5 and H-Ar), 8.04 (1H, s, H-7). LCMS
(ESI‘) m/z = 568.3[M-H]'.
e 34: (3R){[4-chloro(morpholinesulfonyl)phenyl]methyl}(4-chlorophenyl)
{[1-(hydroxymethyl)cyc|opropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindol
VA CI
o“ O
Example 34, Step 1, 4-Chloromercaptobenzonitrile
WO 55860
4-Chlorofluorobenzonitrile (200 mg, 1.29 mmol) and NaZS (110 mg, 1.41 mmol) were added into a
microwave vial, DMF (1 mL) was charged into the mixture before stirring for 1 h at room ature.
1 M NaOH solution was charged to pH 12 then washed by EtZO (3 x 10 mL, acedified mixture with 1
M HCI to pH 1-2 and extracted with EtZO (3 x 10 mL), the combined extracts were washed by water
(30 mL) and brine (20 mL), dried by MgSO4, concentrate in vacuo to give a yellow solid (156 mg,
72%).1H NMR (500 MHz, CDCI3) 6 4.14 (1H, 5, SH), 7.21 (1H, d, J = 8.4 Hz, H-Ar), 7.42 (1H, s, H-Ar),
7.52 (1H, d, J = 8.3 Hz, H-Ar).
Example 34, Step 2, 4-Chloro(morpholinosulfonyl)benzonitrile
To a stirring mixture of romercaptobenzonitrile (50 mg, 0.30 mmol), t-BuNCl (333 mg, 1.20
mmol) and H20 (0.02 mL) in MeCN (3 mL) was added NCS (120 mg, 0.90 mmol) at 0 °C, after 30
mins, morpholine (0.03 mL, 0.30 mmol) was d into reaction mixture before stirring overnight at
room temperature, extracted with EtOAc (3 x 10 mL), the combined organic extracts were washed with
water (30 mL) and brine (20 mL), dried by MgSO4 and concentrated in vacuo. Chromatography (silica;
EtOAc, petrol 20 — 50%) gave a white solid (59 mg, 70%). LCMS (ESP) m/z = 287.2 [M+H]+.
e 34, Step 3, (4-Chloro(morpholinosu|fony|)pheny|)methanamine
(,3. 0
Prepared by the same method as described in Example 33, Step 1 (195 mg, 0.68 mmol), THF (4 mL),
1 mol/L borane-THF complex (3.4 mL, 3.40 mmol) and 1 M HCI in MeOH (4 mL). Concentration in
vacuo gave yellow oil (90 mg, 46%). LCMS (ES|+) m/z = 291.2 [M+H]+.
Example 34, Step 4, 6-Acetyl(4-chloro(morpholinosulfonyl)benzyl)(4-chlorophenyI)
hydroxyisoindolinone
0 N :él\N/\\o
O 0
The title compound was prepared using procedures similar to those described for ation 9.
LCMS (ESI') m/z = 573.3 [M-H]'.
Example 34, Step 5, 6-Acetyl(4-chloro(morpholinosulfonyl)benzyl)(4-chlorophenyl)
ydroxymethyl)cyclopropyl)methoxy)isoindolinone
The title compound was prepared using procedures similar to those described in Preparation 22.
LCMS (ESI+) m/z = 681.4 [M+Na]+.
Example 34, Step 6, (3R){[4-chIoro(morpholinesuIfony|)phenyl]methyI}(4-
chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-
dihydro-1H-isoindolone
The title nd was prepared using procedures similarto those described for Example 1. 1H-NMR
(500 MHz, CDCI3) 6 .23 (1H, m, cyclopropane CHHCHZ), 0.30-0.32 (1H, m, cyclopropane
), 0.44-0.49 (2H, m, cyclopropane CHZCHZ), 1.65 (6H, s, CH3), 1.85 (2H, bs, 2H), 2.66 (1H, d,
C-O-CHH), 2.96-3.01 (2H, m, H-morpholine), 3.07-3.11 (2H, m, H-morpholine), 3.18 (1H, d, C-OCHH
), 3.33 (1H, d, CHHOH), 3.64-3.71 (4H, m, H-morpholine), 3.82 (1H, d, CHHOH), 4.92 (1H, d, N-
CHH), 5.01 (1H, d, N-CHH), 7.11 (2H, d, H-Ar), 7.18-7.24 (5H, m, H-Ar), 7.72 (1H, s, H-Ar), 7.79 (1H,
d, H-5), 8.04 (1H, s, H-7). LCMS (ESI') m/z = 673.4 [M-H]'.
Example 35: 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)
fluoro(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindoIy|]oxy}methyl)cyclopropane
carboxamide
MeOZC Step1 HzNOC Step2 H2NOC
Example 35, Step 1: Methyl 1-carbamoylcyclopropanecarboxylate
A solution ofdimethyl cyclopropane-1,1-dicarboxylate (57.8 g, 0.365 mol) in 7M ammonia in methanol
was stirred at room temperature for 90 h, then evaporated under reduced pressure to afford the title
compound as a colourless solid (52.2g, 99%). 1H NMR (400 MHz; DMSO-da): 7.82 (1H, s), 7.35 (1H,
s), 3.63 (3H, s), 1.34 (4H, 3).
Example 35, Step 2: 1-(Hydroxymethyl)cyclopropanecarboxamide
Lithium aluminium hydride (27.2 g, 0.719 mol) was added portionwise over 0.75 h. to a stirred
suspension of methyl 1-carbamoylcyclopropanecarboxylate (51.2 g, 0.358 mol) in THF (1.5 L) at 0°C
under nitrogen. On complete addition the mixture was stirred at 0°C for a further 1.25 h. prior to
dropwise addition of water (27 mL), 15% NaOH (27 mL) and water (82 mL) sequentially. The mixture
was d to warm to room temperature, stirred for 0.5 h. and filtered through celite. The celite was
washed with EtOAc and the combined filtrate and washings evaporated to give the title nd as
a colourless solid (33.4 g, 71%). 1H NMR (400 MHz; a): 7.10-6.85 (2H, m), 4.97 (1H, t), 3.48
(2H, d), .85 (2H, m), .58 (2H, m).
Example 35, Step 3: oro(methylsulfonyl)phenyl)methanamine
Hexamethylenetetramine (40.8 g, 0.291 mol) was added in one portion to a stirred, room temperature
solution of 1-(bromomethyl)chloro(methylsulfonyl)benzene (75 g, 0.264 mol) in EtOAc (1 L). After
1.5 h. the mixture was chilled in ice and the precipitate filtered, washed with cold EtOAc and air dried.
This material was suspended in MeOH (375 mL), concentrated HCI (150 mL) added dropwise over
0.33 h. and the mixture heated at 40°C for 2 h. The mixture was cooled, concentrated under reduced
pressure and the residue basified with 5M NaOH, with cooling, and extracted with dichloromethane (3
x 500 mL). ed extracts were dried (MgSO4) and evaporated and the e chromatographed
on silica gel eluting with 20 - 100 % EtOAc in isohexane gradient followed by 0 - 10 % MeOH
ning 7N NH3 in EtOAc gradient to afford the title compound as a pale brown solid (27.8 g, 48 %).
1H NMR (400 MHz, 00013): 8.02 (1H, d), 7.58 (1H, dd), 7.51 (1H, cl), 4.19 (2H, s), 3.23 , 1.64
(2H, s).
Example 35, Step 4: (4-Chloro(methylsuIfonyl)phenyl)methanamine6-bromo(4-chloro
(methylsulfonyl)benzyl)(4-chlorophenyl)fluorohydroxyisoindolinone
HATU (45.6 g, 0.12 mol) was added to a stirred solution of o(4-chlorobenzoyl)
fluorobenzoic acid (35.79 g, 0.1 mol) and (4-chloro(methylsulfonyl)phenyl)methanamine (21.95 g,
0.1 mol) in anhydrous DMF (100 mL) at room temperature. The mixture was stirred at room
temperature for 18 h, at 60°C for 18 h, then cooled and concentrated under reduced re. The
residual oil was poured into water (1.5 L), extracted with dichloromethane (4 x 500 mL) and the
combined extracts washed with water (2 x 1 L), dried (M9804) and evaporated and the residue
chromatographed on silica gel eluting with 20 — 100 % EtOAc in ane gradient to give the title
compound as a pale yellow solid (42.7 g, 76 %). 1H NMR (400 MHz, CDCI3): 7.88 (1H, d), 7.70 (1H,
d), 7.57 (1H, d), 7.49 - 7.45 (1H, m), 7.36 - 7.33 (1H, m), 7.25 (4H, s), 5.13 (1H, d), 4.73 (1H, d), 5.00 -
4.20 (1H, br s), 3.07 (3H, s).
Example 35, Step 5: 1-(((5-Bromo(4-chloro-2(methylsulfonyl)benzyl)(4-chlorophenyl)
oxoisoindolin-1yl)oxy)methyl)cyclopropanecarboxamide
The title compound was prepared using procedures similarto those described for Preparation 12. 1H
NMR (400 MHz, CDCI3): 7.99 (1H, d), 7.93 (1H, dd), 7.75 (1H, d), .47 (1H, m), 7.30-7.19 (5H,
m), 6.98 (1H, s), 6.76 (1H, s), .88 (2H, m), 3.36-3.20 (5H, m), 1.05-0.90 (2H, m), 0.63-0.53 (2H,
Example 35, Step 6: -(4-Chloro(methylsulfonyl)benzyl)(4-chlorophenyl)fluoro
oxo(propenyl)isoindolinyl)oxy)methyl)cycIopropanecarboxamide
The title compound was prepared using procedures similar to those described for Preparation 13., H
NMR(400 MHz; a): 7.87 (1H, d), 7.76 (1H, d), 7.70 (1H, dd), 7.50(1H, dd), 7.30 - 7.20 (5H, m),
6.98 (1H, s), 6.70 (1H, s), 5.73 (1H, s), 5.33 (1H, s), 5.00 - 4.85 (2H, m), 3.36 - 3.23 (5H, m), 2.20 (3H,
s), 0.97 - 0.85 (2H, m), 0.65 - 0.45 (2H, m).
Example 35, Step 7: 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)-
7-fluoro(2-hydroxypropan-Z-yl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropane-
1-carboxamide
The title compound was prepared using procedures similar to those described for Example 2. Chiral
separation using supercritical fluid chromatography gave the title compound as the slower running
isomer (0.245 9). 1H NMR (400 MHz, CDCI3): 7.91-7.87 (2H, m), 7.53 (1H, d), 7.37-7.30 (1H, m), 7.78-
7.15 (5H, m), 6.53 (1H, s), 5.45 (1H, s), 5.05-4.95 (2H, m), 3.36 (1H, d), 3.00 (3H, s), 2.96 (1H, d),
1.96 (1H, s), 1.65 (6H, s), 1.32-1.15 (2H, m), 0.60-0.30 (2H, m). MS(ES+) m/z 633.3\635.3 [M+H]+.
Example 36: (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)({1-
[hydroxy(2H2)methyl]cyclopropyl}(2H2)methoxy)(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-
isoindolone
Example 36 Step 1, 6-Bromo(4-chloro(methylsulfonyl)benzyl)(4-chlorophenyl)—3-
hydroxyisoindolinone
The title compound was prepared using procedures similar to those described for ation 9:
LCMS (ESI') m/z = 538.0 [M-H]'.
Example 36 Step 2, 6-Bromo(4-chloro(methylsulfonyl)benzyl)—3-(4-chlorophenyl)—3-((1-
ratedhydroxymethyl)cyclopropyl)deuteratedmethoxy)isoindolinone
The title compound was prepared using procedures similarto those described for Preparation 22 but
using 1,1-bis(deuteratedhydroxymethy|)cyc|opropane LCMS (ESI+) m/z = 650.2 [M+Na]+.
Example 36 Step 3 : -[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)—3-
({1-[hydroxy(2H2)methyl]cyclopropyl}(2H2)methoxy)(2-hydroxypropanyl)-2,3-dihydro-1H-
isoindolone
D CI
D O
0Q l/
N s\
Starting from e 36, Step 2, the title compound was prepared using procedures similar to those
described in Preparation 13 and Example 2. 1H-NMR (500 MHz, CDCI3) 6 0.22-0.26 (1H, m,
cyclopropane CHHCHZ), 0.32-0.36 (1H, m, cyclopropane CHHCH2), 0.45-0.51 (2H, m, cyclopropane
CHZCHZ), 1.65 (6H, s, CH3), 1.83 (2H, bs, 2H), 3.06 (3H, s, SOchg), 5.02 (2H, s, N-CH3), 7.13-7.20
(4H, m, H-Ar), 7.24-7.26 (2H, m, H-Ar), 7.30 (1H, d, H-Ar), 7.79 (1H, d, H-5), 7.89 (1H, s, H-Ar), 8.02
(1 H, s, H-Ar). LCMS (Esr) m/z = 608.4 [M+H]+
Example 37 and 38: (3R)(4-Chlorophenyl)[(4-chIorophenyl)methyl](2-hydroxypropan-Z-
yl)(oxolanyloxy)-2,3-dihydro-1H-isoindolone
(*both isomers at the position shown)
OH 0
The title nds were prepared using procedures r to those described in Preparations 10
and 13 and Example 2. The two diastereoisomers were isolated by ative chiral HPLC.
Example 37 (isomer 1): 1H-NMR Spectrum: 5H (500 MHz, CDCI3): 8.01 (1 H, d), 7.79 (1H, dd), 7.25-
7.13 (9H, m), 4.52 (1H, d), 4.31 (1H, d), 3.88-3.83 (2H, m), 3.59-3.55 (1H, m), .19 (2H, m),
1.79-1.74 (1H, m), 1.59 (6H, s), .46 (1H, m).
Example 38: (isomer 2): H-NMR Spectrum: 6H (500 MHz, CDCI3): 8.01 (1H, d), 7.79 (1H, dd), 7.21-
7.13 (9H, m), 4.49 (1H, d), 4.43 (1H, d), 3.88-382 (2H, m), 3.63-3.59 (1H, m), 3.49 (1H, dd), 3.14 (1H,
dd), 1.65-1.61 (2H, m), 1.59 (6H, 3).
Example 39 and 40: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropan-Z-
yl)[(oxolanyl)methoxy]-2,3-dihydro-1H-isoindolone
(*both isomers at the position shown)
Step 1; 3-(Hydroxymethyl)tetrahydrofuran,
To a solution of tetrahydrofurancarboxylic acid (0.247 mL, 2.58 mmol) in THF (13 mL) at 0 °C was
added slowly lithium aluminium hydride (1.0 M in THF, 5.2 mL, 5.16 mmol), stirred for 10 minutes then
allowed to attain room temperature and stirred for a further 3 hours. The reaction mixture was cooled
to 0 °C and diluted with diethyl ether (15 mL), then treated sequentially with water (0.2 mL), NaOH
(15% solution, 0.2 mL) and water (0.6 mL) and stirred for 30 minutes. The white suspension was then
treated with sodium sulfate, stirred for a further 20 minutes, filtered over celite, washed with diethyl
ether (2x 20 mL) and concentrated in vacuo to yield 224 mg (85%) of the title compound as a
colourless oil which was carried fon/vard to the next stage t further purification.1H-NMR
Spectrum: 6H (500 MHz, CDCI3): 3.90-3.84 (2H, m), 3.78-3.73 (1H, m), 3.66-3.63 (2H, m), 3.61-3.57
(1H, m), .46 (1H, m), .01 (1H, m), .62 (1H, m)
Step 2: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropany|)[(oxolan-
3-yl)methoxy]-2,3-dihydro-1H-isoindoIone
(*both isomers at the position shown)
Starting from 3-(hydroxymethyl)tetrahydrofuran and Preparation 8, the title compounds were prepared
using procedures r to those described for Preparations 10 and 13 and Example 2. The two
diastereoisomers were isolated by preparative chiral HPLC.
Example 39 (*isomer 1): 1H-NMR Spectrum: 5H (500 MHz, ): 8.06 (1H, d), 7.79 (1H, dd),
7.31-7.22 (8H, m), 7.16 (1H, d), 4.67 (1H, d), 4.15 (1H, d), 3.77-3.75 (1H, m), 3.68-3.59 (2H, m), 3.49-
3.47 (1H, m), 2.79-2.77 (1H, m), 2.69-2.66 (1H, m), 2.10-2.02 (1H, m), 1.83-1.76 (1H, m), 1.59 (6H, s),
1.30-1.24 (1H, m).
Example 40 er 2): 1H-NMR Spectrum: 5H (500 MHz, (00300): 7.94 (1 H, d), 7.68-7.66 (1 H, m),
7.17-7.14 (4H, m), 7.07-7.04 (5H, m), 4.46 (1H, d), 4.13 (1H, CI), 3.59-3.55 (3H, m), 3.18-3.15 (1H, m),
2.68-2.60 (2H, m), 2.12-2.04 (1H, m), 1.86-1.79 (1H, m), 1.47 (6H, s), 1.44-1.39 (1H, m).
Examples 41 and 42: (3R)[(4-chloromethanesuIfonylphenyl)methyl](4-chIorophenyl)
fluoro[1-hydroxy(oxany|)ethyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-
indolone
(*both isomers att the position shown)
HO HO
Cl Cl
HO 0 l>§ F
Step 1 F
o 0 O 0
0 Step 2
N 802Me O —.
N 802Me 302W
0 Br
0b O O5
or Cl
2016/053042
Step 1: 6-Bromo(4-chIoro(methylsulfonyl)benzyI)(4-chlorophenyl)fluoro((1-
(hydroxymethyl)cyclopropy|)methoxy)isoindolinone
Thionyl chloride (3 mL, 42.2 mmol) was added se to a solution of 6-bromo-2—(4-chloro
(methylsulfonyl)benzyl)(4-chlorophenyl)fluorohydroxyisoindolinone (Example 35, Step 4 )
(4.72 g, 8.44 mmol) in THF (50 mL) at 0 °C under a nitrogen atmosphere. DMF (20 drops) were added
and the orange solution was allowed to warm to rt over 1 d. The reaction mixture was concentrated in
vacuo and dissolved in THF (40 mL). Cyclopropane-1,1-diyldimethanol (1.72 g, 16.9 mmol) was
added followed by KZCOS (2.33 g, 16.9 mmol) and the orange e was stirred at rt under an
here of nitrogen for 1 d. DCM and water were added and the layers separated. The aqueous
layer was extracted with DCM and the combined organic layers were dried (phase separator) and
trated in vacuo. The residue was purified by Biotage (30 — 35% EtOAc in iso-hexanes) to give
the title compound (3.33 g, 61%) as a pale yellow solid.
1H NMR (400 MHz, DMSO) 6.01 (1H, d), 7.95 (1H, dd), 7.79 (1H, d), 7.56 (1H, dd), 7.36 (2H, d), 7.30
— 7.26 (3H, m), 5.01 — 4.90 (2H, m), 4.45 (1H, t), 3.46 — 3.31 (2H, m), 3.29 (3H, s), 3.05 - 2.96 (2H, m),
0.42 - 0.40 (2H, m), 0.27 (1H, d), 0.19 (1 H, dd).
Step 2: 6-Acetyl(4-chIoro(methylsulfonyl)benzyl)(4-chlorophenyl)fluoro((1-
(hydroxymethyl)cyclopropyl)methoxy)isoindolinone
The title compound was prepared in a r manner to that described in Example 21 Step 3, using
6-bromo(4-chlor0(methylsulfonyl)benzyl)(4-chlorophenyl)fluoro((1-
(hydroxymethyl)cyclopropy|)methoxy)isoindolinone.
1H NMR (400 MHz, DMSO) 6.30 (1H, d), 6.06 (1H, d), 7.60 (1H, d), 7.56 (1H, dd), 7.36 - 7.26 (5H, m),
.04 — 4.94 (2H, m), 3.46 — 3.30 (2H, m), 3.30 (3H, s), 3.06 — 2.98 (2H, m), 2.75 (3H, s), 0.41 (2H, dd),
0.26 - 0.25 (1H, m), 0.16 (1H, d).
Step 3: -[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-
hydroxy(oxany|)ethyl]{[1-(hydroxymethyl)cyclopropy|]methoxy}-2,3-dihydro-1H-
isoindolone
LaCI3.2LiC| (0.6 M in THF, 1.93 mL, 1.16 mmol) was added to a solution of 6-acetyl(4-chloro
(methylsulfonyl)benzyl)(4-chlorophenyl)fluoro((1-
(hydroxymethyl)cyc|opropyl)methoxy)isoindolinone (703 mg, 1.16 mmol) in THF (7 mL) under at
atmosphere of nitrogen and the yellow solution d at rt for 50 min and cooled to 0 °C. (Tetrahydro-
2H-pyran-4—yl)magnesium chloride (Novel Compound Solutions, 0.5 M in 2-Me-THF, 23.2 mL, 11.6
mmol) was added slowly and the cool bath was removed. The red solution was allowed to warm to rt
over 30 min and quenched with saturated aqueous NH4C| solution. DCM and water were added and
the layers separated. The aqueous layer was extracted with DCM and the combined organic layers
were washed with brine, dried (phase separator) and concentrated in vacuo. The residue was purified
by Biotage (65 — 100% EtOAc in iso-hexanes) and submitted for chiral purification by SFC.
Example 41, Diastereoisomer 1:
1H NMR (400 MHz, c0013) 7.91 (1H, d), 7.76 (1H, d), 7.40 - 7.33 (2H, m), 7.30 - 7.27 (2H, m), 7.24 -
7.17 (3H, m), 5.06 — 4.96 (2H, m), 4.07 - 3.93 (2H, m), 3.60 (1 H, dd), 3.42 - 3.23 (4H, m), 3.04 (3H, s),
2.60 (1H, d), 2.00 (1H, dd), 1.91 - 1.62 (1H, m), 1.79 (1H, s), 1.61 (3H, s), 1.53 - 1.40 (2H, m), 1.29 -
1.22 (2H, m), 0.51 (2H, s), 0.49 - 0.39 (1H, m), 0.22 (1H, d). MS(ES+) m/z 692 [M+H]+
Example 42, Diastereoisomer 2:
1H NMR (400 MHz, c0013) 7.91 (1H, d), 7.72 (1H, s), 7.43 (1H, d), 7.36 (1H, dd), 7.31 — 7.17 (5H, m),
.07 — 4.96 (2H, m), 4.07 - 3.93 (2H, m), 3.61 (1H, dd), 3.40 - 3.22 (4H, m), 3.03 (3H, s), 2.77 (1 H, d),
2.01 (1H, s), 1.66 - 1.60 (2H, m), 1.62 (3H, s), 1.52 - 1.39 (2H, m), 1.31 — 1.25 (2H, m), 0.51 (2H, s),
0.49 - 0.40 (1 H, m), 0.22 (1H, d). MS(ES+) m/z 692 [M+H]+
Examples 43 and 44: (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)—4-
fluoro[2-hydroxy(piperazinyl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-
2,3-dihydro-1H-isoindolone
(*both isomers att the position shown)
TBDMSO
HO TBDMSO
F Step 1
o 0 Q/CI Step 2 Gel
@N *’
SOZMB 302Me
Br \\61%;
0 soMe
Step 3
:30 TBDMSO
Step 4 CI
soMe DSHG
HNQ0N SOZMe
Step 1
The product from Example 41, Step 1 was purified by preparative chiral HPLC to provide
(3R)bromo[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone as a single enantiomer. TBDMS
protection (following a procedure similar to Preparation 11) gave acetyl((1-(((tert-
butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chloro(methylsu|fony|)benzyl)(4-
phenyl)fluoroisoindolinone.
Step 2:
60% Sodium Hydride sed in mineral oil (0.132 g; 3.44 mmol) was added portionwise over 10
minutes to a stirred solution of (R)acetyl((1-(((tertbutyldimethylsilyl
)oxy)methyl)cyclopropyl)methoxy)(4-chloro(methylsu|fony|)benzyl)(4-
chlorophenyl)fluoroisoindolinone (1.56 g; 2.17 mmol) and trimethylsulfoxonium iodide (0.533 g;
2.42 mmol) in anhydrous DMSO (9 mL) and anhydrous THF (9 mL), at room temperature, under
nitrogen. After 20 hours, water (400 mL) was added, the mixture extracted with EtOAc (2 x 250 mL)
and the combined organics dried (MgSO4) and evaporated under reduced pressure. The residue was
purified by chromatography on silica gel (100 g) eluting with 0-50% EtOAc in isohexane gradient to
afford 3-((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chloro
(methylsulfonyl)benzyl)(4—chlorophenyl)fluoro(2—methyloxiranyl)isoindolinone as a pale
yellow foam (0.736 g, 46%). 1H NMR (400 MHz, : 7.90 (1H, d), 7.77 (1H, d), 7.40 - 7.27 (2H,
m), 7.25 - 7.20 (2H, m), 7.22 - 7.10 (3H, m), 5.10 - 4.85 (2H, m), 3.85 - 3.70 (1H, m), 3.35 - 3.20 (2H,
m), 3.10 - 3.00 (1H, m), 2.97 (3H, s), 2.80 - 2.60 (2H, m), 1.55 (3H, s), 0.82 (9H, s), 0.50 - 0.35 (2H,
m), 0.25 - 0.05 (2H, m), 0.01 (6H, s).
Step 3 and 4:
Step 3 was perfomed by following procedures similarto those described in Preparation 34 (but using
zine instead ofdimethylamine). Step 4 was performed by following procedures similar to those
described for Example 10. Chiral separation using supercritical fluid chromatography gave
Example 43 (the faster running isomer) (74 mg, 30%). 1H NMR (400 MHz, : 7.90 (1H, d), 7.77
(1H, d), 7.46 (1H, dd), 7.40 - 7.30 (1H, m), 7.30 - 7.10 (5H, m), 5.10 - 4.98 (2H, m), 3.82 (1H, d), 3.40 -
3.30 (1H, m), 3.30 -3 .20 (1H, m), 3.03 (3H, s), 2.85 - 2.65 (7H, m), 2.55 - 2.45 (2H, m), 2.38 - 2.25
(2H, m), 2.20 - 1.65 (3H, m), 1.51 (3H, s), 0.55 - 0.45 (2H, m), 0.45 - 0.35 (1H, m), 0.30 - 0.15 (1H, m).
MS(ES+) m/z 706 [M+H]+.
Example 44 (the slower running isomer) (76 mg, 31%). 1H NMR (400 MHz, : 7.90 (1H, d), 7.75
(1H, d), 7.48 (1H, dd), 7.40 - 7.30 (1H, m), 7.30 - 7.10 (5H, m), 5.10 - 4.90 (2H, m), 3.82 (1H, d), 3.40 -
3.30 (1H, m), 3.30 - 3.20 (1H, m), 3.03 (3H, s), 2.85 - 2.65 (7H, m), 2.55 - 2.45 (2H, m), 2.38 - 2.25
(2H, m), 2.20 - 1.65 (3H, m), 1.51 (3H, s), 0.55 - 0.45 (2H, m), 0.45 - 0.35 (1H, m), 0.30 - 0.15 (1H, m).
) m/z 706 [M+H]+.
Examples 45 and 46: (3R)(4-ChIorophenyl)[(1S)(4-chlorophenyl)ethyl]{[(3S,4R)
hydroxyoxolany|]oxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindolone and (3R)(4-
chlorophenyl)[(1S)(4-chlorophenyl)ethyI]{[(3R,4S)hydroxyoxolanyl]oxy}(2-
hydroxypropan-Z-y|)-2,3-dihydro-1H-isoindolone
o “0’9o “0"”
Cl Cl
<5 o L0
.....O/
OH 0
Examples 45 and 46, Step 1:
3-(4-ChlorophenyI)[(1S)(4-chlorophenyl)ethyl][(4-hydroxyoxolany|)oxy](propen-
2-yl)-2,3-dihydro-1H-isoindolone
The title compound (as a mixture to 4 isomers) was prepared using a similar procedure as be for
ation 10, using 1,4-anhydroerythritol and Preparation 13 The crude material was purified
using chromatography on silica (Pet:EtOAc 1:0 to 1:2) to give the desired products as foamy
colourless oil. |The product (a mixture of4 isomers) was used directly in the next step.
Example 45 and Example 46, Step 2
Example 45 and Example 46 were prepared using similar procedure to that decribed for Example 2.
The desired ts were isolated was using chromatography on silica (Pet:EtOAc 2:1 to 0:1).
Example 45 r 1): R, = 0.25 (Pet:EtOAc/1:2). 1H NMR (500 MHz, c0013) 6 (ppm) 1.61 (s, 3H,
2), 1.62 (s, 3H, C(CH3)2), 1.85 (s, 1H, C(CH3)ZOH), 1.91 (d, 3H, J = 7.3 Hz, NCHCH3), 2.49 (d,
1H, J = 3.6 Hz, CHOH), 3.27-3.37 (m, 1H, H-b), 3.67 (dd, 1H, J = 10.4, 4.0 Hz, H-a), 3.83 (dd, 1H, J =
.3, 1.2 Hz, H’-a), 3.87-4.01 (m, 3H, H-d, H’-d, H-c), 4.28 (q, 1H, J = 7.3 Hz, NCH), 6.94 (d, 2H, J =
8.5 Hz, Ar-H), 6.97-7.09 (m, 6H, Ar-H); 7.20 (d, 1H, J = 8.0 Hz, isoindolinone-H), 7.73 (d, 1H, J = 7.9,
1.5 Hz, isoindolinone-H), 7.98 (d, 1H, J = 1.3 Hz, isoindolinone-H); MS(ES+) m/z 484.3 [M+H]+;
Example 46 (iomer 2): Rf = 0.57 (Pet:EtOAc/1:2); 1H NMR (500 MHz, CDCI3) 6 (ppm) 1.61 (s, 6H,
C(CH3)2), 1.74 (d, 3H, J = 7.2 Hz, NCHCH3), 1.82 (s, 1H, C(CH3)20H), 2.06 (s, 1H, CHOH), 2.63-2.68
(m, 1H, H-b), 2.71 (dd, 1H, J = 8.5, 7.3 Hz, H-d), 3.28 (dd, 1H, J = 8.5, 8.4 Hz, H’-d), 3.42 (dd, 1H, J =
.3, 4.1 Hz, H-a), .49 (m, 1H, H-c), 3.53 (dd, 1H, J =10.4,1.5 Hz, H’-a), 4.19 (q, 1H, J = 7.3
Hz, NCH), 6.97 (d, 1H, J = 8.0 Hz, isoindolnone-H), 7.28 (d, 2H, J = 8.4 Hz, Ar—H), 7.31-7.44 (m, 4H,
Ar-H), 7.57 (d, 2H, J = 8.4 Hz, Ar—H), 7.68 (dd, 1H, J = 7.9, 1.6 Hz, isoindolinone-H), 7.98 (d, 1H, J =
1.3 Hz, isoindolinone-H); ) m/z 484.3 [M+H]+;
Example 47: (3R)(4-Chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropan-Z-yl)
methoxy-2,3-dihydro-1H-isoindolone
o J: J
OH 0
Starting from Preparation 8 the title compound was prepared using quently) processes similar
to those described in Preparation 12; Example 21, step 3 and Example 1. 1H-NMR Spectrum: (in
CDCI3) 6 (ppm) 1.81 (s, 6H, C(CH3)2), 2.12 (s, 1H, OH), 2.62 (s, 3H, OCH3), 4.07 (d, 1H, J =14.8 Hz,
NCHH), 4.58 (d, 1H, J = 14.8 Hz, NCHH), 7.06 (d, 1H, J = 8.0 Hz, Ar-H), 7.12-7.17 (m, 2H, Ar-H),
7.15-7.25 (m, 6H, Ar-H), 7.89 (dd, 1H, J = 8017 Hz, Ar-H), 8.00 (d, 1H, J = 1.6 Hz, Ar-H. ms (M-H")
m/z = 456.4.
Example 48: (3R)(4-Chlorophenyl)—2-[(4-chlorophenyl)methyl]({1 -
[hydroxy(2H2)methyl]cyclopropyl}(2H2)methoxy)(2-hydroxypropanyl)-2,3-dihydro-1H-
isoindolone
D OH
The title compound was ed using procedures similarto those bed for Example 1. 1H NMR
(500 MHz; CDCI3) LH 0.10-0.18 (2H, m, 2 x H-cyclopropyl), 0.38-0.44 (2H, m, 2 x H-cyclopropyl), 1.52
(1H, br s, OH), 1.61 (3H, s, CH3), 1.63 (3H, s, CH3), 1.75 (1H, br s, OH), 4.18 (1H, d, J = 15.0 Hz,
CHaHbPh), 4.55 (1H, d, J =15.0 Hz, CHaHbPh), 7.10 (1H, d, J = 8.0 Hz, H-4), 7.12-7.23 (8H, m, H-Ar),
7.72 (1H, dd, J = 1.7 and 8.0 Hz, H-5), 7.99 (1H, d, J = 1.7 Hz, H-7). LCMS (ES+) m/z 424.3, 426.3
[(M-c3 sidechain)+H]+
Example 49: (3R)—3-(4-Chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropan-Z-yl)(3-
hydroxypropoxy)-2,3-dihydro-1H-isoindolone
g00.
The title compound was prepared using procedures similar to those described for Example 1. 1H-
NMR Spectrum: 6H (500 MHz, CDCI3) 1.40-1.55 (2H, m, CH2CH2CH20H), .64 (6H, m, 2 x
CH3), 2.79-2.85 (1H, m, CH2CH2CH20H), 2.85-2.93 (1H, m, CH2CH2CH20H), 3.57 (2H, t, J = 6.1
Hz, CH2CH2CH20H), 4.05 (1H, d, J = 14.8 Hz, NC-H), 4.65 (1H, d, J = 14.9 Hz, NC-H’), 7.08 (1H, d,
wo 2017/055860 2016/053042
J = 7.9 Hz, ArH), .25 (8H, m, 8 xArH), 7.70 (1 H, dd, J = 1.7 and 7.9 Hz, ArH) and 7.99 (1 H, d, J
= 1.2 Hz, 7-H). ; (ES+) m/z 424.3 [M-O(CH-'2)3OH]+
Example 50: (3R)—2-[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)—4-fluoro
{[1-(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropan-Z-yl)-2,3-dihydro-1H-isoindol
D Cl
F (j
N SOZMe
OH 0
The title compound was prepared using procedures similar to those described for Example 36. 1H-
NMR Spectrum: 6H (500 MHz, CDCI3) 0.17-0.25 (1H, m, H), 0.38-0.42 (1 H, m, Cy-Py-H), 0.46-
0.53 (2H, m, Cy-Py—H2), 1.60-1.64 (6H, 2 x s, 2 x CH3), 2.77 (1H, d, J = 9.1 Hz, 2’-H), 3.03 (3H, s,
), 3.25 (1H, J = 9.1 Hz, 2’-H’), 3.36 (1H, d, J = 11.2 Hz, 4’-H), 3.81 (1H, d, J =11.1 Hz, 4’-H'),
4.94-5.04 (2H, m, NC-H, NC-H’), 7.14-7.21 (3H, m, 3 xArH), 7.25-7.29 (2H, m, 2 xArH), 7.32 (1H, dd,
J = 2.3 and 8.4 Hz, ArH), 7.45 (1H, dd, J = 1.3 and 10.8 Hz, ArH), 7.81 (1H, d, J = 1.4 Hz, ArH) and
7.89 (1H, d, J = 2.3, 7-H). (ES+) m/z 520.3 [M-C5H902]+
Example 51: (3R)(4-Chlorophenyl)—2-[(4-chlorophenyl)methyl](2,2-difluoro
hydroxypropoxy)(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone
Starting from Preparation 20 and difluoroprpoanediol, the title compound was prepared by following
procedures similarto those described in Preparation 12 and Example 1. 1H-NMR Spectrum: 6H (500
MHz, CDCI3) 1.55 (3H, s, CH3), 1.56 (3H, s, CH3), 2.93-3.02 (2H, m, CH2), 3.63-3.78 (2H, m, CH2),
4.19 (1H, d, J = 15.0 Hz, NCHH’), 4.45 (1H, d, J =15.0 Hz, NCHH’), 7.04-7.11 (7H, m, 7 x ArH), 7.13-
7.15 (2H, m, 2 x ArH), 7.67 (1H, dd, J =1.7 and 8.0 Hz, ArH), 7.94 (1H, d, J =1.7 Hz, ArH). m/z 536.4
lM+Hl+
Examples 52 and 53: (3R)(4-ChlorophenyI)[(4-chlorophenyl)methyl]{[2-
(hydroxymethyl)cyclobutyl]methoxy}(2-hydroxypropanyl)—2,3-dihydro-1H-isoindolone
(both isomers as shown)
HO .D HO
'17 CI
0 0/CI
\\\\g
OH 0
OH O
Example 52 and 53, Step 1: (2-Hydroxymethyl-cyclobutyl)-methanol
A suspension of cyclobutane oxylic acid (649mg, 4.50 mmol) in tetrahydrofuran (45 mL) was
cooled to 0 0C and treated with lithium aluminium hydride (1M in THF, 18.0 mL, 18.01 mmol) and
d for 3 hours. Once complete by TLC (SiOZ; 30% MeOH in DCM) the reaction was diluted with
diethyl ether (45 mL) and treated sequentially with water (0.68 mL), sodium ide (15% aq., 0.68
mL) and water (2.1 mL) then stirred for 30 minutes. Sodium sulphate was added and the solution
mixture stirred for a further 30 minutes. Once ed the c layer was concentrated in vacuo to
yield the title compound (305 mg, 58%) as a colourless oil that was used without the need for further
purification.
Example 52 and 53, Step 2:
Starting from (2-hydroxymethyl-cyclobutyl)-methanol, Example 52 and 53 were made in a similar
mannerto Example 1. The two desired s were isolated by preparative chiral HPLC.
Example 52 (isomer 1): 6H (500 MHz, (c0300): 8.05 (1H, d), 7.79 (1H, dd), 7.25-7.21 (4H, m), 7.19-
7.14 (5H, m), 4.52 (1H, d), 4.33 (1H, d), 354-349 (1H, m), 3.44-3.40 (1H, m), 03 (1H, m), 2.91-
2.87 (1H, m), 2.53-2.49 (1H, m), 2.37-2.35 (1H, m), 2.04-1.96 (2H, m), 1.73-1.63 (2H, m), 1.59 (6H, s).
Example 53 (isomer 2): 5.. (500 MHz, (c0300): 8.06 (1H, d), 7.79 (1H, dd), 7.25-7.21 (4H, m), 7.19-
7.14 (5H, m), 4.52 (1H, d), 4.33 (1H, d), 3.54-3.49 (1H, m), 3.44-3.40 (1H, m), 3.06-3.03 (1H, m), 2.91-
2.87 (1H, m), 2.53-2.48 (1H, m), 2.39-2.35 (1H, m), 2.04-1.96 (2H, m), 1.72-1.63 (2H, m), 1.59 (6H, s).
Examples 54 and 55: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][2-hydroxyoxo
(pyrrolidinyl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-
isoindolone
(*Both isomers at the position shown)
Example 54 and Example 55, Step 1: (R)((1-(((tert-
Butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)—2-(4-chlorobenzyl)—3-(4-chlorophenyl)—6-
(propenyl)isoindolinone
TBSO
TBSCI (2.62 g, 17.4 mmol, 1.5 eq.), imidazole (1.19 g, 1.5 eq.) and hlorobenzy|)(4-
chlorophenyI)((1-(hydroxymethyl) cyclopropyl) methoxy)(propenyl)isoindolinone (R-
isomer) (5.9 g, 11.6 mmol, 1 eq,) were dissolved in THF (100 mL) and heated to 50 oC for6 h. Allowed
to cool to r.t., and partitioned between EtOAc (2 x 70 mL) and H20 (70 mL). Organic extracts were
combined, dried over M9804, and solvent removed in vacuo. The residue was purified by MPLC (1-
% EtOAc/petrol) to give a clear oil (6.045 g, 84%); 1H NMR (500 MHz; CDCI3) 6 -0.02 (3H, s, CHgsi),
0.00 (3H, s, , 0.07-0.12 (2H, m, cPr), 0.33-0.38 (2H, m, cPr), 0.83 (9H, s, tBu), 1.27 (3H, s, Me),
.19 (2H, brs, ), 2.62 (1H, d, CHaHbO), 2.89 (1H, d, CHaHbO), 3.36 (1H, d, CHaHbO), 3.65 (1H, d,
CHaHbO), 4.32 (1H, d, CHaHbN), 4.39 (1H, d, CHaHbN», 5.18 (1H, m, H-alkene), 5.44 (1H, m, H-
alkene), .22 (9H, m, H-Ar), 7.58 (1H, dd, H-5), 7.96 (1H, d, H-7).
Example 54 and Example 55, Step 2:
(3R)—3-((1-(((tert-Butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chlorobenzyl)(4-
chlorophenyl)—6-(1,2-dihydroxypropan-Z-yl)isoindolinone
TBSO
Ho Ob
?????????????????????????????? ????????????????????????????????????
The title compound was prepared using procedures similar to those described for Example ing
product as re of 2 diasteroisomers). 1H NMR (500 MHz; CDC' 3) o -0.02 (3H, s, CH3Si), 0.00
(3H, s, CH3Si), 0.07-0.13 (2H, m, cPr), 0.33-0.39 (2H, m, cPr), 0.83 (9H, s, tBu), 1.56 (3H, s, Me), 1.82
(1H, br s, OH), 2.60 (1H, d, CHaHbO), 2.64 (1H, br s, OH), 2.88 (1H, d, CHaHbO), 3.36 (1H, d,
CHaHbO), 3.62-3.74 (2H, m, CHaHbO and CHaHbOH), 3.81 (1H, d, CHaHbOH), 4.31 (1H, d, CHaHbN),
4.40 (1H, d, CHaHbN)), 7.03-7.13 (SH, m, H-Ar), 7.13-7.20 (4H, m, H-Ar), 7.67 (1H, dd, H-5), 7.94 (1H,
d, J = 1.5 Hz, H-7); MS ES+ 440.3, 442.3 [M-sidechain(
e 54 and Example 55, Step 3:
2-((R)((1-(((tert-Butyldi methylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chlorobenzyl)(4-
phenyl)oxoisoindolinyl)hydroxypropanoic acid
,)- (1
-2,1)(
OH O �
TEMPO (161 mg, 1.03 mmol, 0.25 eq.), NaCIO2 (744 mg, 8.22 mmol, 2 eq.) and NaOCI (30 µl, 0.08
mmol, 0.02 eq.) were added to
(3R)((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chlorobenzyl)(4-
chlorophenyl)(1,2-dihydroxypropanyl)isoindolinone (2.7 g, 4.11 mmol, 1 eq.) in a mixture of
MeCN (20 ml) and sodium phosphate buffer (pH 6.5, 16 ml), and the mixture was stirred at r.t. for 5
days. Water was added, and the pH adjusted to pH 8 with NaOH (1 M aq.). Na2SO3 (aq) was added,
and the pH adjusted to pH 2 with HCI (1 M aq). The mixture was extracted with EtOAc (2 x 50 ml),
dried over MgSO4, and the solvent was removed in vacuo. Purification by MPlC on silica with a
gradient from 0-10% MeOH/DCM gave a white foam (2.27 g, 83%, ca. 3:1 mixture of distereoisomers);
Major diastereoisomer: 1H NMR (500 MHz; CDC' 3) o 0.00 (3H, s, CH3Si), 0.02 (3H, s, CH3Si), 0.10-
0.17 (2H, m, cPr), 0.35-0.42 (2H, m, cPr), 0.85 (9H, s, tBu), 1.94 (3H, s, Me), 2.65 (1H, d, CHaHbO),
2.90 (1H, d, ), 3.39 (1H, d, CHaHbO), 3.65 (1H, m, CHaHbO), 3.81 (1H, d, CHaHbOH), 4.35
(1H, d, ), 4.41 (1H, d, CHaHbN)), 7.00-7.20 (9H, m, H-Ar), 7.96 (1H, dd, H-5), 8.29 (1H, d, H-
7); MS ES- 668.3, 670.3 [M-Hr.
Example 54 and Example 55, Step 4:
N *
OH O
The title compound was prepared from 2-((R)((1-(((tertbutyldimethylsilyl
)oxy)methyl)cyclopropyl)methoxy)(4-chlorobenzyl)(4-chlorophenyl)
oxoisoindolinyl)hydroxypropanoic acid, using PyBrop and pyrrolidine. Deprotection, using similar
procedures to those described for Example 10, followed by preparative HPLC gave the two s.
Example 54 (*isomer 1). 1H-NMR Spectrum: (500 MHz, CDCl3) δ 0.12-0.18 (2H, m, cyclopropane
CH2CH2), 0.40-0.44 (2H, m, cyclopropane CH2CH2), 1.55-1.59 (2H, m, H-pyrrolidine), 1.72-1.77 (3H,
m, H-pyrrolidine and OH), 1.85 (3H, s, CH3), 2.56-2.61 (1H, m, olidine), 2.72 (1H, d, J = 9.4 Hz,
C-O-CHH), 2.76 (1H, d, J = 9.4 Hz, C-O-CHH), 3.09-3.14 (1H, m, H-pyrrolidine), 3.39 (1H, d, J = 11.4
Hz, CHHOH), 3.47 (1H, d, J = 11.4 Hz, CHHOH), 3.51-3.56 (2H, m, H-pyrrolidine), 4.24 (1H, d, J =
14.9 Hz, N-CHH), 4.51 (1H, d, J = 14.9 Hz, N-CHH), 5.44 (1H, s, OH), .21 (9H, m, H-Ar), 7.51
(1H, d, J = 8.0 Hz, H-5), 8.0 (1H, s, H-7). MS (ESI+) m/z = 507.4 [M+H]+
e 55 (*isomer 2): 1H-NMR Spectrum: (500 MHz, CDCl3) δ 0.09-0.13 (2H, m, cyclopropane
CH2CH2), 0.38-0.43 (2H, m, cyclopropane ), 1.51-1.59 (2H, m, H-pyrrolidine), 1.70-1.78 (3H,
m, H-pyrrolidine and OH), 1.85 (3H, s, CH3), 2.59-2.66 (2H, m, H-pyrrolidine and C-O-CHH), 2.74 (1H,
d, J = 9.3 Hz, C-O-CHH), 3.05-3.10 (1H, m, H-pyrrolidine), 3.33 (1H, d, J = 11.3 Hz, CHHOH), 3.48
(1H, d, J = 11.3 Hz, CHHOH), 3.55-3.58 (2H, m, olidine), 4.17 (1H, d, J = 14.8 Hz, , 4.54
(1H, d, J = 14.8 Hz, N-CHH), 5.38 (1H, s, OH), 7.11-7.22 (9H, m, H-Ar), 7.53 (1H, d, J = 8.1 Hz, H-5),
8.0 (1H, s, H-7). MS (ESI+) m/z = 507.4 [M+H]+
Examples 56 and 57: 2-[(1R)(4-Chlorophenyl)[(4-chlorophenyl)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]hydroxy-N,N-
dimethylpropanamide (*both isomers at the position highlighted)
Example 56 and 57, Step 1:
2-((R)((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-chlorobenzyl)(4-
chlorophenyl)oxoisoindolinyl)hydroxy-N,N-dimethylpropanamide
To a on of 2-((R)((1-(((tert-butyldimethylsilyl)oxy)methyl)cyclopropyl)methoxy)(4-
chlorobenzyl)(4-chlorophenyl)oxoisoindolinyl)hydroxypropanoic acid (Example 54 and
Example 55, step 3) (400 mg, 0.60 mmol), PyBroP (420 mg, 0.90 mmol) and pyridine (0.05 mL, 0.60
mmol) in MeCN (4 mL) was added ylamine solution (2M in MeCN, 0.75 mL, 1.5 mmol) before
stirring at room temperature overnight. The reaction was extracted with EtOAc (3 x 10 mL), the
combined organic extracts were washed with water (30 mL) and brine (20 mL), dried by MgSO4 and
concentrated in vacuo.Chromatography (silica; EtOAc, petrol 20 – 90%) gave a white solid (250 mg,
71%). LCMS (ESI+) m/z = 719.5 [M+Na]+.
Example 56 and 57, Step 2
2-((1R)(4-Chlorobenzyl)(4-chlorophenyl)((1-(hydroxymethyl)cyclopropyl)methoxy)
oxo-2,3-dihydro-1H-isoindolyl)hydroxy-N,N-dimethylpropanamide (*both s at the
position highlighted)
N *
OH O
The title compound was prepared using procedures similar to those described for Example 10.
Purification by chiral HPLC gave the title compound:
1H-NMR (500 MHz, CDCl3) δ 0.11-0.15 (2H, m, cyclopropane CH2CH2), 0.39-0.43 (2H, m,
cyclopropane ), 1.63 (1H, brs, OH), 1.86 (3H, s, CH3), 2.59-2.64 (4H, m, H and NCH3
), 2.80 (1H, d, C-O-CHH), 3.02 (3H, brs, N-CH3), 3.34 (1H, d, CHHOH), 3.50 (1H, d, CHHOH),
4.18 (1H, d, N-CHH), 4.54 (1H, d, N-CHH), 5.39 (1H, s, OH), 7.11-7.18 (7H, m, H-Ar), 7.21 (2H, d, HAr
), 7.50 (1H, d, H-5), 8.0 (1H, s, H-7). LCMS (ESI-) m/z = 627.4 [M+Formate]-.
WO 55860
Example 56 (*isomer 1): 1H-NMR Spectrum: (500 MHz, CDCI3) 6 012-018 (2H, m, cyclopropane
CHZCHZ), .44 (2H, m, cyclopropane CHZCHZ), 1.59 (1H, brs, OH), 1.87 (3H, s, CH3), 2.59 (3H,
brs, N-CH3), 2.70 (1H, d, J = 9.4 Hz, C-O-CHH), 2.79 (1 H, d, J = 9.4 Hz, C-O-CHH), 3.00 (3H, brs, N-
CH3), 3.38 (1H, d, J = 11.4 Hz, , 3.48 (1H, d, J =11.4 Hz, CHHOH), 4.24 (1H, d, J = 14.8 Hz,
N-CHH), 4.51 (1H, d, J =14.8 Hz, N-CHH), 5.44 (1H, s, OH), 7.11-7.17 (7H, m, H-Ar), 7.20 (2H, d, J =
8.9 Hz, H-Ar), 7.47 (1H, d, J = 8.0 Hz, H-5), 8.0 (1H, s, H-7). MS (ESI+) m/z = 481.3 [M+H]+
Example 57 (*isomer 2): 1H-NMR Spectrum: (500 MHz, CDCI3) 6 0.11-0.15 (2H, m, cyclopropane
CHZCHZ), 0.39-0.43 (2H, m, cyclopropane CHZCHz), 1.63 (1H, brs, OH), 1.86 (3H, s, CH3), 2.59-2.64
(4H, m, C-O-CHH and N-CH3), 2.80 (1H, d, J = 9.5 Hz, C-O-CHH), 3.02 (3H, brs, N-CH3), 3.34 (1H, d,
J = 11.3 Hz, CHHOH), 3.50 (1H, d, J =11.3 Hz, CHHOH), 4.18 (1H, d, J =14.8 Hz, , 4.54 (1H,
d, J =14.8 Hz, N-CHH), 5.39 (1H, s, OH), 7.11-7.18 (7H, m, H-Ar), 7.21 (2H, d, J = 8.9 Hz, H-Ar), 7.50
(1H, d, J = 8.0 Hz, H-5), 8.0 (1H, s, H-7). MS (ESI+) m/z = 481.3 [M+H]+
Examples 58 and 59: )(4-Chlorophenyl)[(4-chlorophenyl)methyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindoIyl]hydroxy-N-
methylpropanamide
(*both isomers at the position highlighted)
The title compound was prepared from 2-((R)((1-(((tertbutyldimethylsilyl
)oxy)methyl)cyclopropyl)methoxy)(4-chlo robe nzy|)-1 -(4-ch|o rophenyI)
oxoisoindolinyI)hydroxypropanoic acid, using PyBrop and methylamine. Deprotection, using
similar procedures to those bed for Example 10, followed by preparartive HPLC gave the two
isomers.
Example 53 (*isomer1): 1H-NMR : (500 MHz, CDCI3) 5 0.13-0.19 (2H, m), .44 (2H, m), 1.67
(1H, brs,), 1.84 (3H, s), 2.66 (1H, d, ), 2.79 (3H, d), 2.83 (1H, d, ), 3.36 (1H, d), 3.46 (1H, d, ), 4.05
(1H, s), 4.21 (1H, d, ), 4.50 (1H, d), 6.66 (1H, q, ), 7.08-7.18 (9H, m, ), 7.62 (1H, d, ), 6.23 (1H, s).
LCIVIS (ESI') m/z = 567.3 [M-H]'
Example 59 (*isomer 2):1H-NMR: (500 MHz, cool?) 5 0.12-0.16 (2H, m), 0.36-0.43 (2H, m), 1.65
(3H, s), 2.63 (1H, d, ), 2.79 (3H, d, ), 2.65 (1H, d, ), 3.34 (1H, d, J= 11.4 Hz, CHHOH), 3.50 (1H, d, J:
11.4 Hz, CHHOH), 4.19 (1H, d, ), 4.52 (1H, d), 6.90 (1H, q, ), 7.09-7.20 (9H, m, ), 7.61 (1H, d), 8.23
(1 H). LCMS (ESI’) m/z = 567.3 [M-H]‘
Examples 60: (3R){[4-chloro(methylsulfanyl)phenyl]methyl}(4-chlorophenyl)—4-fluoro
{[1-(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindol
Starting from 5-bromo(4-ch|orobenzoy|)f|uorobenzoic acid and Preparation 43 (4-chloro
(methyIthio)phenyl)methanamine), the title nd was prepared by following procedures similarto
those described in Preparation 9, Preparation 10, Example 21, Step 3 and Example 1; in a
sequential manner.
1H NMR (500 MHz, c0013) 67.81 (1H, s, H-7), 7.39 (1H, 05), 7.23 (2H, CI, .19 (3H, m), 7.01 (1H,
s), 6.96 (1H, d), 4.68 (1H, d), 4.43 (1H, d), 3.49 (1H, q), 3.41 (1H, q), 2.95 (1H, d), 2.89 (1H, d), 2.38
(3H, s), 1.90 (1H, s), 1.71 (1H, t), 1.61 (6H, d), 0.41-0.48 (2H, m), 0.28-0.31 (1H, m), 0.14-0.17 (1H,
m). LCMS (ESI') m/z = 634.3 [M+Formate]‘
Examples 61 and 62: (3R)[(4-Chloromethanesulfinylphenyl)methyl](4-chIorophenyl)
fluoro{[1-(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-
isoindolone
(*both isomers at position highlighted)
To a solution of (3R){[4-chIoro(methylsuIfanyl)phenyl]methy|}(4-chlorophenyl)fluoro{[1-
(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropany|)-2,3-dihydro-1H-isoindoI0ne
(Example 60) (140 mg, 0.24 mmol) in MeOH (3 mL) was added sodium ate (56 mg, 0.26 mmol)
in portions over 3 mins at 0 °C. The mixture was stirred overnight, H20 was added, and extracted with
EtOAc . The combined organic extracts were washed with brine, dried (MgSO4) and trated in
vacuo. Chromatography (SP4, silica; MeOH/EtOAc 0-30%) gave the products as white solids (70 mg,
48% and 70 mg, 48%).
Example 61: *fast running isomer: 1H NMR (500 MHz, 00013) 5 7.85 (1H, d’), 7.81 (1H, d), 7.48 (1H,
d), 7.40 (1H, dd), .34 (3H, m), 7.25-7.26 (2H, m), 4.71 (1H, d), 4.28 (1H, d), 3.62 (1H, d), 3.33
(1H, d), 2.82 (1H, d), 2.72 (1H, d), 2.62 (3H, s, CH3), 2.27 (1H, br s), 1.99 (1H, s), 1.62 (3H, s), 1.61
(3H, s), 0.39-0.44 (2H, m), 0.19-0.21 (1 H, m), 0.01-0.03 (1 H, m). LCMS (ESP) m/z = 682.3 [M+Na]+;
e 62: *slow running isomer: 1H NMR (500 MHz, CDCI3) 5 7.82 (1H, d), 7.78 (1H, cl), 7.42 (1 H,
dd), 7.24 (1H, dd), 7.15-7.19 (4H, m), 7.13 (1H, d), 4.60 (1H, d), 4.46 (1H, d), 3.59 (1H, dd, J = 6.1
and 11.5 Hz, CHHOH), 3.49 (1H, dd, J = 6.1 and 11.5 Hz, CHHOH), 3.04 (1H, d,), 2.91 (1H, d), 1.90
(1H, s), 2.72 (3H, s), 1.67 (1H, t), 1.63 (3H, s), 1.62 (3H, s), 0.41-0.49 (2H, m), 0.30-0.33 (1H, m),
0.05-0.09 (1H, m). LCMS (ESP) m/z = 628.3 [M+Na]+
Examples 63 and 64: (3R)[(4-Chloromethanesulfonylpheny|)methyl](4-chlorophenyl)
(2-hydroxymethoxypropan-Z-yl){[1 -(hyd roxymethyl)cyclopropyl]methoxy}-2,3-
dihydro-1H-isoindolone
(*both isomers at the position ghted)
The title compounds were ed using methods similar to those described in Example 43; but
using sodium methoxide d of piperazine.
Example 63 (the faster running isomer) (31mg, 14%). 1H NMR (400 MHz, DMSO-d6): 7.85 (1H, d),
7.77 (1H, d), 7.58-7.50 (2H, m), 7.34-7.23 (5H, m), 5.49 (1H, s), 5.02-4.66 (2H, m), 4.39 (1H, t), 3.53-
3.44 (2H, m), 3.28 (3H, s), 3.24 (3H, s), 3.02 (1H, d), 2.89 (1H, d), 1.47 (3H, s), 0.41-0.32 (2H, m),
0.26-0.17 (1H, m), 0.06 (1H, d). MS(ES+) m/z 652 [M+H]+.
Example 64 (the slower running isomer) (11 mg, 5%). 1H NMR (400 MHz, DMSO-d6): 7.85 (1H, d),
7.77 (1H, d), 7.57-7.50 (2H, m), 7.33-7.22 (5H, m), 5.49 (1H, s), 5.01-4.81 (2H, m), 4.39 (1H, t), 3.54-
3.44 (2H, m), 3.28 (3H, s), 3.24 (3H, s), 3.02 (1H, d), 2.90 (1H, d), 1.47 (3H, s), 0.37 (2H, d), 0.27-0.18
(1H, m), 0.11-0.03 (1H, m). MS(ES+) m/z 652 .
Examples 65 and 66: (3R)[(4-Chloromethanesulfonylpheny|)methy|](4-chlorophenyl)
(1 ,2-dihydroxypropanyl)f|uoro{[1-(hydroxymethyl)cycIopropyl]methoxy}-2,3-dihydro-
1H-isoindolone
(*both isomers at the position shown)
The title compounds were ed using methods similar to those described in Example 41, Step 1
and Example 27; in a sequential manner.
Example 65 (faster running, major isomer, 174 mg, 26%): 1H NMR (400 MHz, DMSO-d6): 7.84 (1H,
d), 7.77 (1H, d), 7.58-7.50 (2H, m), 7.32-7.21 (5H, m), 5.30 (1H, s), .80 (3H, m), 4.39 (1H, t),
3.57-3.40 (3H, m), 3.25 (3H, s), 3.03 (1H, d), 2.90 (1H, d), 1.46 (3H, s), 0.37 (2H, s), 0.27-0.18 (1H,
m), 0.07 (1H, d). MS(ES+) m/z 638 [M+H]+.
Example 66 (slower running, minor isomer, 74 mg, 11%): 1H NMR (400 MHz, DMSO-d6): 7.85 (1H,
dd), 7.77 (1H, d), 7.57-7.48 (2H, m), 7.32-7.23 (5H, m), 5.30 (1H, s), 4.99-4.79 (3H, m), 4.39 (1H, t),
3.57-3.39 (3H, m), 3.25 (3H, s), 3.02 (1H, d), .87 (1H, m), 1.46 (3H, s), 0.37 (2H, s), 0.28-0.17
(1H, m), 0.08 (1H, d). ) m/z 638 [M+H]+.
Examples 67 and 68: (3R)[(4-Chloromethanesulfonylphenyl)methyl](4-chlorophenyl)
fluoro[2-hydroxy(4-methy|piperaziny|)propany|][(3R)-oxolanyloxy]-2,3-dihydro-
1H-isoindolone (*both isomers at the position shown)
\Xm—: $02Me
Starting from Preparation 44, the title compounds were prepared using methods similar to those
described in Example 43, steps 2 and 3 (but using N-methyl-piperazine instead of piperazine).
Example 67 r running isomer, 132 mg, 28%): 1H NMR (400 MHz, DMSO-d6): 7.92 (1 H, d), 7.80
(1H, d), 7.60-7.49 (2H, m), 7.36-7.19 (5H, m), 5.26 (1H, s), 5.09-4.67 (2H, m), 4.02-3.94 (1H, m), 3.78
(1H, q), 3.57-3.48 (1H, m), 3.27-3.15 (4H, m), 2.61 (1H, d), 2.34 (4H, s), 2.16 (4H, s), 2.08 (3H, s),
1.74-1.62 (1H, m), 1.59-1.38 (4H, m). MS(ES+) m/z 706 [M+H]+.
Example 68 (slower running isomer, 131 mg, 28%): 1H NMR (400 MHz, DMSO-d6): 7.85 (1H, d), 7.78
(1H, d), .51 (2H, m), .20 (5H, m), 5.23 (1H, s), 4.92 (2H, s), 3.99-3.92 (1H, m), 3.78 (1H,
q), 3.58-3.49 (1H, m), 3.43-3.36 (1H, m), 3.24 (3H, s), 2.59 (1H, d), 2.35 (5H, d), 2.16 (4H, s), 2.08
(3H, s), 1.74-1.63 (1H, m), 1.57-1.40 (4H, m). MS(ES+) m/z 706 [M+H]+.
Examples 69 and 70: (3R)[(4-Chloromethanesulfonylphenyl)methyl](4-chlorophenyl)
fluoro[2-hydroxy(4-methylpiperaziny|)propany|][(3S)-oxolanyloxy]-2,3-dihydro-
1H-isoindolone (*both isomers at the position shown)
\N/fi
|\ S( )2Ne
The title compounds were prepared using procedures similarto those described in Examples 67 and
68, but using (3S)-hydroxy-tetrahydrofuran.
Example 69 (faster running , 96 mg, 39%): 1H NMR (400 MHz, DMSO-d6): 7.92 (1H, d), 7.78
(1H, d), 7.59-7.50 (2H, m), 7.34-7.19 (5H, m), 5.27 (1H, s), 5.06-4.71 (2H, m), 4.01-3.93 (1H, m), 3.75
(1H, q), 3.60-3.51 (1H, m), 3.45-3.40 (1H, m), 3.24 (3H, s), 3.15 (1H, dd), 2.61 (1H, d), 2.41-2.26 (5H,
m), 2.16 (4H, s), 2.07 (3H, s), 1.81-1.64 (1H, m), 1.59 (1H, d), 1.50 (3H, s). MS(ES+) m/z 706 [M+H]+.
Example 70 (slower running isomer, 97 mg, 39%): 1H NMR (400 MHz, DMSO-d6): 7.84 (1 H, d), 7.77
(1H, d), 7.63-7.50 (2H, m), 7.30-7.19 (5H, m), 5.23 (1H, s), 4.98 (2H, s), 3.99-3.92 (1H, m), 3.77 (1H,
q), .54 (1H, m), 3.46-3.40 (1H, m), 3.25 (3H, s), 3.17 (1H, dd), 2.59 (1H, d), 2.34 (5H, s), 2.16
(4H, s), 2.08 (3H, s), 1.86-1.73 (1H, m), 1.73-1.61 (1H, m), 1.53 (3H, s). MS(ES+) m/z 706 [M+H]+.
Example 71: (3S)(4-Chlorophenyl)[(1R)(4-chloropheny|)f|uoro(2-hydroxypropan-Z-
yl)oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]propanoic acid
Step 1: -[(1R)Acetyl(4-chlorophenyl)fluorooxo[(3S)-oxolanyloxy]-2,3-
dihydro-1H-isoindolyl](4-chlorophenyl)propanoic acid
The title compound was prepared from ethyl (3S)[(1R)acetyl(4-chlorophenyl)fluorooxo
[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl](4-chlorophenyl)propanoate (Preparation 45) in
a r manner as described in Preparation 40. MS(ES+) m/z 570 [M-H]'
Step 2: (3S)(4-Chlorophenyl)[(1R)(4-chloropheny|)quoro(2-hydroxypropan-Z-yl)
oxo[(3S)-oxolanyloxy]-2,3-dihydro-1 H-isoindolyl]propanoic acid
Using the t from Step 1, the title compound was prepared in a similar fashion to Example 1.
1H NMR (400 MHz, DMSO-d6): 12.42 (1H, s), 7.79 (1H, s), 7.51 (1H, d), 7.10 (4H, dd), 6.99 (4H, d),
.36 (1H, s), 4.62 (1H, dd), 4.21-4.13 (1H, m), 3.92 (1H, q), 3.77-3.63 (2H, m), 3.50 (1H, cl), 3.23 (1H,
dd), 3.12 (1H, dd), 2.25-2.09 (1H, m), 2.09-1.94 (1H, m), 1.47 (6H, s); MS(ES+) m/z 570 [M-H]'
e 72: 1-({[(1R){[4-Chloro(hydroxymethyl)phenyl]methyl}(4-chlorophenyl)
fluoro(2-hydroxypropan-Z-yl)oxo-2,3-dihydro-1H-isoindoly|]oxy}methyl)cyclopropane
carbonitrile
To a solution of 5-chloro{[(1R)(4-ch|oropheny|)[(1-cyanocyclopropyl)methoxy]fluoro(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindoIyl]methy|}benzoic acid (Example 79) (233 mg,
0.4 mmol) in THF (10 mL) were added triethylamine (0.17 mL, 1.2 mmol) and isobutyl chloroformate
(0.08 mL, 0.6 mmol) and the reaction e was stirred for 1h. Water (0.1 mL) was added followed
by NaBH4 (0.045 g, 1.2 mmol) in small ns. Stirred for 1 h, water (10 mL) was added and the
product was extracted with EtOAc. The crude product was purified on Silica, eluted with petrol —
EtOAc 0 — 80% to afford the title compound (150 mg, 66%).
1H NMR (400 MHz, DMSO-d6): 7.82 (1H, d), 7.53 (1H, dd), 7.37-7.22 (5H, m), .03 (2H, m), 5.37
(1H, s), 5.21 (1H, t), 4.54-4.36 (3H, m), 4.33 (1H, d), 3.02 (1H, d), 2.80 (1H, d), 1.48 (6H, s), 1.23-1.13
(2H, m), 0.78-0.68 (1H, m), 0.61-0.52 (1H, m). MS(ES+) m/z 472 [3 [M-CN(c—Pr)CHZO)]+.
Exam ples 73 and 74: 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-
chIorophenyl)fluoro[1-hydroxy(1-methyl-1H-pyrazo|y|)ethyl]oxo-2,3-dihydro-1H-
isoindolyl]oxy}methyl)cyclopropane-1 -carboxamide
(*both isomers at position shown)
Step 1: 2-(4-Chlorobenzoyl)fluoro(hydroxy(1-methyl-1H-pyrazoly|)methyl)benzoic acid
A 5 litre round bottom flask fitted with an overhead stirrer was charged with 5-bromo-2—(4-
chlorobenzoyl)fluorobenzoic acid (100 g, 0.28 mol) and anhydrous THF (1.5 L). The solution was
cooled to -3 °C and a solution of methyl magnesium chloride (2.15M in THF, 130 mL, 0.279 mol) was
added dropwise at such a rate that the internal temperature remained below -1 °C (25 min). On
complete addition, the mixture was stirred at 0 °C for 15 min then cooled to -78 °C. A solution of n-
butyllithium (2.2M in hexanes, 152 mL; 0.33 mol) was added dropwise over 30 min at such a rate so
that the internal temperature ed below -70 °C. On te addition the mixture was stirred at
L78 °C for 30 min. A on of 1-methyl-1H-pyrazolecarboxaldehyde (39.7 g, 0.36 mol) in
anhydrous THF (500 mL) was added se over 20 min at such a rate so that the internal
temperature remained below -70 °C. On complete addition the mixture was stirred at -78 °C for 15
min, the cooling bath removed and the mixture allowed to reach RT. The mixture was quenched with 1
M HCI, the pH adjusted to 1-2 and extracted with EtOAc (2 x 500 mL). Combined organics were dried
(M9804) and the solvent evaporated. The residue was divided into four equal portions and each
portion chromatographed on silica gel (300 g) eluting with 0 — 20% MeOH in DCM gradient to afford
the title nd as a colourless solid (48.33 g, 44.5%). lmpure fractions were pooled, ated
and tographed to afford a further quantity of title compound (11.05 g, 10.2%).MS: [M +H]+ =
Step 2: 2-(4-Chlorobenzoyl)fluoro(1-methyl-1H-pyrazolecarbonyl)benzoic acid
To a stirred mixture of 2-(4-chlorobenzoyl)fluoro(hydroxy(1-methyl-1H-pyrazol
yl)methyl)benzoic acid (20 g, 51.48 mmol) in EtOAc (86 mL) at 0 °C was added 10% aqueous KBr
(29.83 mL, 25 mmol) ed by TEMPO (0.82 g, 5.23 mmol). To this stirred mixture was added a
solution of sodium hydrogen carbonate (5.40 g, 64.25 mmol) and sodium hypochlorite (89 mL, 5-20%
aqueous solution ex Fisher, catalogue number /PB17) in water (47 mL) at such a rate so that
the reaction temperature ed below 5 °C. on was stopped upon complete oxidation as
indicated by LCMS (approximately half of the solution was required). The reaction was quenched by
addition ofdilute s sodium sulphite solution and the mixture extracted with EtOAc (4 x 500 mL).
Combined organics were dried (M9804) and the solvent removed under reduced pressure to give the
title compound as a pale orange solid (15.27 g, 76%). The aqueous layer was acidified with 2 M HCI
and ted with EtOAc (500 mL). The organics were dried (MgSO4) and the solvent d under
reduced pressure to give a further ty ofthe title compound (3.44 g, 17%). MS:[M +H]+ = 387.
Step 3: 2-(4-Chloro(methylsulfony|)benzyl)(4-chlorophenyl)fluorohydroxy(1-
methyl-1H-pyrazolecarbonyl)isoindolinone
2-(4-Chlorobenzoyl)—3-fluoro(1-methyl-1H-pyrazole-4—carbonyl)benzoic acid (18.20 g, 46.55 mmol)
and (4-chloro(methylsulfonyl)phenyl)methanamine (Example 35, step 3) (19.4 g, 88.30 mmol) were
stirred in DMF (90 mL) at RT under nitrogen. HATU (26.80 g, 70.53 mmol) was added and the
reaction mixture stirred at RT for 1.25 h. The reaction mixture was d with water, sat. aq. NaHC03
solution and EtOAc. The layers were separated and the aqueous was extracted with EtOAc. The
combined cs were washed with 4% LiCl aq. solution, dried (M9804), filtered and concentrated in
vacuo. The residue was purified using a 340 g SNAP column eluting with EtOAc in iso-hexanes (0 to
100%) to give the title 8.8 g, 32 % yield. MS: [M +H]+ = 587.
Step 4: (R)(((2-(4-Chloro(methylsulfonyl)benzy|)—1-(4-chlorophenyl)fluoro(1-methyl-
1 H-pyrazolecarbonyl)oxoisoindoliny|)oxy)methyl)cyclopropanecarboxamide
The title compound was prepared from 2-(4-chloro(methylsulfonyl)benzyl)(4-chlorophenyl)
fluorohydroxy(1-methyl-1H-pyrazole-4—carbonyl)isoindolinone (2.0 g, 3.40 mmol) in a similar
manner to that described in preparation 10. The product was separated using chiral SFC. Slowest
eluting isomer.MS: [M +H]+ = 685.
Step 5: 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-
y(1-methyl-1H-pyrazoly|)ethy|]oxo-2,3-dihydro-1H-isoindol
yl]oxy}methyl)cyclopropanecarboxamide
(R)(((1-(4-Chlorophenyl)—2-((5-chloropyridinyl)methyl)—7-fluoro(1-methyl-1H-pyrazole
carbonyl)oxoisoindoliny|)oxy)methyl)cyclopropanecarbonitrile (620 mg, 0.90 mmol) was stirred in
THF (5 mL) at -15°C under nitrogen. MeMgCl (2.15M in THF, 0.91 mL, 1.96 mmol) was added
dropwise forming an orange solution and then stirred for 10 mins at this temperature. The on was
quenched with sat. aq. NH4CI solution, water was then added and the reaction was warmed to RT.
The aqueous was extracted with DCM (2 x 50 mL) and the combined organics were dried (phase
separator) and concentrated in vacuo. The residue was purified using a 25 g SNAP column, eluting
with MeOH in DCM (0 to 5 %). Fractions containing the title compound were concentrated in vacuo
and the compound was ted using chiral SFC.
Example 73: *faster eluting . 1H NMR (400 MHz, CDCI3) 7.90 (1 H, d), 7.82 (1 H, d), 7.53 - 7.49
(1H, m), 7.42 (1H, s), 7.37 - 7.33 (2H, m), 7.25 - 7.18 (5H, m), 6.52 - 6.52 (1H, m), 5.42 - 5.42 (1H, m),
.04 - 4.92 (2H, m), 3.90 (3H, s), 3.36 (1H, d), 2.99 (3H, s), 2.91 (1H, d), 2.32 (1H, s), 1.94 (3H, s),
1.32 - 1.20 (2H, m), 0.53 - 0.46 (1H, m), 0.39 - 0.33 (1H, m); MS: [M +H]+ = 701.
Example 74: *slower eluting isomer. 1H NMR (400 MHz, CDCI3) 7.89 (1 H, d), 7.82 (1 H, s), 7.55 - 7.50
(1H, m), 7.41 (1H, s), 7.37 - 7.32 (2H, m), 7.25 - 7.17 (5H, m), 6.52 - 6.52 (1H, m), 5.42 - 5.42 (1 H, m),
.04 - 4.94 (2H, m), 3.91 (3H, s), 3.36 (1H, d), 3.00 (3H, s), 2.93 (1H, d), 2.34 (1H, s), 1.94 (3H, s),
1.32 - 1.21 (2H, m), 0.54 - 0.47 (1H, m), 0.42 - 0.36 (1H, m); MS: [M +H]+ = 701.
Example 75 and 76: (3R)[(4-ch|oromethanesulfonylpheny|)methyl](4-chlorophenyl)
fluoro[1-hydroxy(1-methy|-1H-pyrazo|yl)ethyl][(1-hydroxycyclopropyl)methoxy]-2,3-
dihydro-1H-isoindolone
(*both isomers at position shown)
Step 1: (R)(4-Chloro(methylsulfonyl)benzyl)(4-chlorophenyl)fluoro((1-
hydroxycyclopropyl)methoxy)(1-methyl-1H-pyrazoIecarbonyl)isoindolinone
The title compound was prepared from 2-(4-chloro(methylsulfonyl)benzyl)(4-chloropheny|)
fluorohydroxy(1-methyl-1H-pyrazolecarbonyl)isoindolinone (2.0 g, 3.4 mmol) in a similar
manner to that bed in preparation 12. The product was separated using chiral SFC. Slowest
eluting isomer. MS: [M +H]+ = 658.
Step 2: (3R)[(4-chIoromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-
hydroxy(1-methyl-1H-pyrazoly|)ethyl][(1-hydroxycyclopropyl)methoxy]-2,3-dihydro-1H-
isoindolone
The title nd was prepared from (R)(4-ch|oro(methylsulfonyl)benzyl)(4-chloropheny|)
fluoro((1-hydroxycyclopropyl)methoxy)(1-methyl-1H-pyrazolecarbonyl)isoindolinone (255
mg, 0.39 mmol) in a similar manner to that described in Examples 73 and 74, step 5. The product
was separated using chiral SFC.
Example 75: *slower eluting isomer. 1H NMR (400 MHz, CDCI3) 7.84 (1H, d), 7.80 (1H, d), 7.45 (1H,
dd), 7.40 (1H, s), 7.34 - 7.20 (5H, m), 7.08 (2H, d), 5.22 (1H, d), 5.02 (1H, d), 3.90 (3H, s), 3.34 (1H,
d), 3.24 (1H, s), 3.03 (3H, s), 2.90 (1H, d), 2.27 (1H, s), 1.93 (3H, s), 0.93 - 0.80 (2H, m), 0.83 - 0.58
(1H, m), 0.43 - 0.37 (1 H, m); MS: [M +H]+ = 874.
Example 76: *faster eluting isomer. 1H NMR (400 MHz, CDCI3) 7.84 (1H, d), 7.81 (1H, d), 7.45 (1H,
dd), 7.40 (1H, d), 7.33 - 7.21 (5H, m), 7.08 (2H, d), 5.21 (1H, d), 5.01 (1H, d), 3.90 (3H, s), 3.32 (1H,
d), 3.21 (1H, s), 3.02 (3H, s), 2.91 (1H, d), 2.28 (1H, s), 1.93 (3H, s), 0.93 - 0.80 (2H, m), 0.82 - 0.55
(1H, m), 0.43 - 0.38 (1 H, m); MS: [M +H]+ = 874.
Examples 77 and 78: (3R)[(4-ch|oromethanesuIfonylphenyl)methyl](4-chlorophenyl)
fluoro[2-hydroxy(4-methy|piperaziny|)propanyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone
The title compounds were prepared using methods similar to those described in Example 43.
Example 77: *fast running isomer: 1H NMR (400 MHz, CDCI3) 7.91 (1H, d), 7.77 (1H, d), 7.48 (1H,
dd), 7.35 (1H, dd), 7.29 - 7.28 (2H, m), 7.22 (1H, d), 7.17 (2H, d), 5.00 (2H, d), 4.40 (1H, s) 3.83 (1H,
d), 3.35 (1H, d), 3.27 (1H, d), 3.04 (3H, s), 2.80 (1H, d), 2.71 (2H, dd), 2.53 - 2.37 (8H, m), 2.25 (3H,
s), 2.01 - 2.01 (1H, m), 1.51 (3H, s), 0.50 (2H, dd), 0.49 - 0.37 (1H, m), 0.21 (1H, d). MS [M+H]+=720.
Example 78: *slow running : 1H NMR (400 MHz, CDCI3) 7.91 (1H, d), 7.78 (1H, d), 7.48 (1H,
dd), 7.34 (1H, dd), 7.28 (2H, d), 7.21 - 7.18 (3H, m), 5.00 (2H, s), 4.47 (1H, d), 3.82 (1H, d), 3.37 (1H,
d), 3.28 (1 H, d), 3.04 (3H, s), 2.81 (1 H, d), 2.73 - 2.87 (2H, m), 2.52 (2H, s), 2.37 (8H, s), 2.25 (3H, s),
2.00 - 2.00 (1 H, m), 1.51 (3H, s), 0.52 - 0.37 (3H, m), 0.21 - 0.17 (1H, m). MS [M+H]*=720.
e 79: 5-chloro{[(1R)(4-ch|orophenyl)[(1-cyanocyclopropyl)methoxy]fluoro
(2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid
Br Step 1 HO 0
Step 2
’ D —>
N Br
.0 ob
FO;/_©/.370002HQ Step 4
Step 1: 2-(2-BromochIorobenzyl)—3-(4-chlorophenyI)fluorohydroxy(2-hydroxypropan-
2-yl)isoindolinone
Starting from Preparation 46 and (2-bromochlorophenyl)methanamine (Example 33, step 1) (1.3
g, 6.24 mmol), the title nd was prepared using methods similar to Example 35, step 4 to
afford 2-(2-bromo-4—ch|orobenzyl)-3—(4-chlorophenyl)fluorohydroxy(2-hydroxypropan
yl)isoindolinone as a colourless solid (2.15 g, 64%). MS [M+H]+=538.
Step 2: 1-(((2-(2-Bromochlorobenzyl)(4-chlorophenyl)fluorooxo(propen
yl)isoindolinyl)oxy)methyl)cyclopropanecarbonitrile
The title compound was prepared from 2-(2-bromochlorobenzyl)(4-chlorophenyl)fluoro
hydroxy(2-hydroxypropanyl)isoindolinone (2.1 g, 3.9 mmol) and 1-
(hydroxymethy|)cyclopropanecarbonitrile (3.42 g, 9.6 mmol) using the method of ation 10 to
furnish (1.7 g, 71%). MS [M+H]+=599.
Step 3: (R)(((2-(2-Bromochlorobenzyl)—1-(4-chlorophenyl)fluoro(2-hydroxypropan
yl)oxoisoindolinyl)oxy)methyl)cycIopropanecarbonitrile
Step 3 was performed in an analogous fashion to Example 2 to give the title compound as a racemate
(1.6 g). Purification by SFC gave the title compound as the fast running isomer. MS [M+H]+=617.
Step 4: 5-chloro{[(1R)(4-chlorophenyI)[(1-cyanocyclopropyl)methoxy]fluoro(2-
hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid
Step 4 was med in an analogous n to e 33, Step 5. Purification by preparative
HPLC afforded (R)chloro((1-(4-ch|orophenyl)fluoro((1-
(hydroxymethyl)cyclopropyl)methoxy)(2-hydroxypropanyl)oxoisoindolinyl)methyl)benzoic
acid (305 mg, 62%). 1H NMR (400 MHz, DMSO) 7.84 (1H, s), 7.67 (1H, d), 7.58 (1H, s), 7.56 (1H, s),
7.41 (1H, dd), 7.32 - 7.22 (5H, m), 4.90 (1H, d), 4.79 (1H, d), 3.16 (1H, d), 2.84 (1H, d), 1.48 (6H, s),
1.27 - 1.15 (2H, m), 0.86 - 0.79 (1H, m), 0.69 - 0.62 (1H, m), OH not observed. MS [M+H]“=583.
Examples 80 and 81: -[(4-chloromethanesuIfonylphenyl)methyl](4-chlorophenyl)—4-
fluoro[1-hydroxy(1-methylpiperidinyl)ethyl]{[1-
(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone
(*both isomers at position shown)
CI CI CI
F Step 1 F Step 2 F
—> —>
O o BocN O o BocN O 0 OH OH OH
O OH O O O
BocN
Step 6
Steps 1 and 2 were performed using ures similar to those described in Examples 73 and 74;
but using tert-butyl 4-formylpiperidinecarboxylate instead of 1-methyl-1H-pyrazole
carboxaldehyde. Also, BuzMg was used in place of methylmagnseium chloride.
Step 3: 5-(1-(1-(tert-Butoxycarbonyl)piperidinyl)hydroxyethyl)—2-(4-chlorobenzoyI)
fluorobenzoic acid
To a RB flask containing 5-(1-(tert-butoxycarbony|)piperidine—4-carbony|)(4-chlorobenzoy|)
fluorobenzoic acid (15.57 g, 31.7 mmol), under N2, was added THF (300 mL) and cooled to —10 °C.
MeMgCl (34.5 mL, 79.4 mmol, 2.3M in THF) was added over a period of 5 mins. Immediately after the
completion of the addition, LCMS analysis showed complete conversion of the starting al. The
reaction was quenched with 1 M HCI (200 mL). The reaction was extracted with EtOAc (2 x 200 mL),
dried with MgSO4, ed and cone. in vacuo. The crude material was purified by column
chromatography, Biotage Isolera, 340g cartridge 0% — 60% EtOAc (0.1% formic acid) in DCM (0.1%
formic acid) to afford 5-(1-(1-(tert-butoxycarbonyl)piperidinyI)hydroxyethyI)(4-chIorobenzoy|)
fluorobenzoic acid as an off white foam (15.1 g, 93%). Purification by chiral preparative LCMS.
* Fast running isomer (Isomer A): MS: [M—H]‘ = 504, [or].;,20 = +28.7 (c 1.9, MeOH)
* Slow running isomer r B): MS: [M—H]‘ = 504, [or]:,20 = -27.8 (c 1.8, MeOH)
Step 4: hIorobenzoyI)f|uoro(1-hydroxy(piperidiny|)ethyl)benzoic acid
hydrochloride
(-)(1-(1-(tert-Butoxycarbonyl)piperidinyl)hydroxyethyI)(4-chIorobenzoyI)fluorobenzoic
acid (Isomer B) (6.09 g, 12.0 mmol) was stirred in 4N HCI in dioxane (70 mL) at rt for 10 min. The
orange solution was concentrated in vacuo yielding 2—(4-chIorobenzoyI)fluoro(1-hydroxy
(piperidin-4—yl)ethyl)benzoic acid hydrochloride (Isomer B) as an orange solid (6.88 g) which was used
in the next step without further purification. MS: [M+H]+ = 406.
In a similar manner (1-(1-(tert-Butoxycarbonyl)piperidinyl)hydroxyethyI)(4-
chIorobenzoyI)fluorobenzoic acid (Isomer A) (7.50 g, 14.8 mmol) furnished 2-(4-chlorobenzoy|)
(1-hydroxy(piperidinyl)ethyl)benzoic acid hydrochloride (Isomer A) which was used in the
next step without further cation. MS: [M+H]+ = 406.
Step 5: 2-(4-ChIorobenzoyI)f|uoro(1-hydroxy(1-methylpiperidinyl)ethyl)benzoic acid
(-)(4-ChIorobenzoyI)fluoro(1-hydroxy(piperidin-4—yl)ethyl)benzoic acid hydrochloride
(Isomer B) (6.88 g, ca. 12.0 mmol) was stirred in MeOH (100 mL) at rt under nitrogen and
formaldehyde solution (37% wt in water, 24 mmol, 1.95 mL) was added. The orange solution was
stirred at RT for 5 min and N (14.4 mmol, 905 mg) was added. The yellow solution was d
at RT for 1 d after which time a less solid had precipitated. The mixture was trated in
vacuo to give 2-(4-chIorobenzoyI)fluoro(1-hydroxy(1-methy|piperidinyl)ethyl)benzoic acid
(Isomer B) as a yellow solid (6 g) which was used in the next step without further purification.
In a similar manner (+)(4-chIorobenzoyl)fluoro(1-hydroxy(piperidiny|)ethyl)benzoic acid
hydrochloride (Isomer A) (6.00 g, ca. 14.8 mmol) furnished 2-(4-chIorobenzoyI)f|uoro(1-hydroxy-
1-(1-methy|piperidinyl)ethyl)benzoic acid (Isomer A) which was used in the next step without further
purification. MS: [M+H]+ = 419.9.
Step 6: 2-(4-ChIoro(methylsuIfony|)benzyI)(4-chIorophenyl)f|uorohydroxy(1-
hydroxy(1-methylpiperidinyl)ethyl)isoindolinone
Starting from (-)(4-chIorobenzoyI)fluoro(1-hydroxy(1-methy|piperidiny|)ethyl)benzoic acid
(Isomer B) (2 g, 4 mmol), Step 6 was performed in a similar fashion to Example 35, Step 4 to give 2-
(4-chloro(methylsuIfonyI)benzyI)—3-(4-chlorophenyI)fluorohydroxy(1-hydroxy(1-
methylpiperidinyl)ethy|)isoindolinone (Isomer B), 939 mg, 38% as a pale orange solid. MS:
[M+H]+ = 621
In a similar manner, (+)(4-Ch|orobenzoyI)fluoro(1-hydroxy(1-methy|piperidin
y|)ethyl)benzoic acid (Isomer A) (1.55 g, ca. 3.70 mmol) furnished 2—(4-chloro
(methylsu|fonyl)benzyl)(4—chlorophenyl)fluoro—3-hydroxy(1-hydroxy(1-methylpiperidin
y|)ethy|)isoindo|inone (Isomer A), 1.05 g, 46%. MS: [M+H]+ = 621.
Step 7: (3R)[(4-chIoromethanesulfonylphenyl)methyI](4-chlorophenyl)fluoro[1-
hydroxy(1-methy|piperidiny|)ethyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-
dihydro-1H-isoindolone
Using (-)(4-chl0ro(methylsuIfonyl)benzyl)(4-chlorophenyl)—4-fluor0hydroxy(1-hydroxy
hylpiperidinyl)ethyl)isoindolinone (Isomer B) (847 mg, 1.36 mmol), Step 7 was performed
in a similar fashion to Example 41, step 1 to give 366 mg of 2-(4—chloro(methylsulfonyl)benzyl)
(4-chlorophenyl)fluoro(1-hydroxy(1-methylpiperidinyl)ethyl)((1-
(hydroxymethyl)cyc|opropyl)methoxy)isoindolinone (Isomer B) as a pale yellow solid which was
further purified using preparative HPLC and ted by chiral SFC to yield Example 80 (55 mg, the
later running isomer)
Example 801H NMR (400 MHz, CDCI3) 7.91 (1 H, d), 7.72 (1H, d), 7.46 - 7.42 (1 H, m), 7.36 (1 H, dd),
7.30 (2H, d), 7.24 - 7.17 (3H, m), 5.06 - 4.96 (2H, m), 3.80 (1H, d), 3.38 (1H, d), 3.24 (1H, d), 3.03 (3H,
s), 2.92 (2H, dd), 2.78 (1H, d), 2.35 - 2.35 (3H, m), 2.25 (3H, s), 1.96 - 1.83 (2H, m), 1.74 (1H, d), 1.61
(3H, s), 1.49 - 1.35 (3H, m), 0.50 (2H, s), 0.49 - 0.38 (1H, m), 0.23 (1H, d); MS: [M+H]+ = 705.4.
In a similar manner (+)(4-chloro(methylsulfonyl)benzyl)(4-chlorophenyl)fluorohydroxy
(1-hydroxy(1-methylpiperidinyl)ethyl)isoindolinone (Isomer A) (787 mg, 1.27 mmol) furnished
138 mg 2-(4-chloro(methylsulfonyl)benzyl)(4-chlorophenyl)fluoro(1-hydroxy(1-
methylpiperidinyl)ethyl)((1-(hydroxymethyl)cyclopropyl)methoxy)isoindolin0ne (Isomer A)
which was further purified using preparative HPLC and separated by chiral SFC to yield Example 81
(8 mg, the later running isomer).
Example 81 1H NMR (400 MHz, c0013) 8.40 (1H, s), 7.91 (1H, d), 7.70 (1H, d), 7.45 (1H, dd), 7.35
(1H, dd), 7.30 (2H, d), 7.23 (1H, d), 7.19 (2H, d), 5.00 (2H, s), 3.73 - 3.67 (1H, m), 3.44 (1H, d), 3.35 -
3.23 (2H, m), 3.14 (1H, d), 3.03 (3H, s), 2.88 (1H, d), 2.51 (3H, s), 2.28 - 2.24 (3H, m), 1.94 - 1.77 (4H,
m), 1.82 (3H, s), 1.40 - 1.31 (1H, m), 0.50 (2H, dd), 0.48 - 0.38 (1H, m), 0.28 (1H, d); MS: [M+H]+ =
705.
Example 82: (3R){[4-chloro(dimethylphosphoryl)phenyl]methyl}(4-chlorophenyl)fluoro-
(hydroxymethyl)cyc|opropy|]methoxy}(2-hydroxypropa nyl)-2,3-dihydro-1H-isoindol
K6 Cl
F o O/
O\ /
N \P\
Starting from 5-bromo(4-chlorobenzoyI)fluorobenzoic acid and ation 47: (2-
(aminomethyI)chlorophenyl)dimethylphosphine oxide, the title compound was prepared using
procedures similar to those described in preparation 9, preparation 10., Example 21 Step 3 and
Example 41 Step 3; in a sequential manner.
*fast running isomer: 1H NIVIR (400 MHz, c0013) 7.81 (1H, d), 7.45 (1H, dd), 7.31 (2H, d), 7.19 - 7.13
(2H, m), 7.10 - 7.06 (3H, m), 5.36 (1H, d), 5.09 (2H, d), 4.37 (1H, dd), 3.84 (1H, d), 2.97 (1H, d), 2.18
(1H, d), 1.89 (1H, s), 1.77 (3H, d), 1.74 (3H, d), 1.65 (3H, s), 1.64 (3H, s), 0.58 - 0.42 (3H, m), 0.27 -
0.22 (1H, m). [M + H] + = 620.
Examples 83 and 84: (3R)[(4-chloromethanesulfonylphenyl)methyl](4-ch|orophenyl)
fluoro[hydroxy(oxany|)methy|]{[1-(hydroxymethyl)cyc|opropy|]methoxy}-2,3-dihydro-
1H-isoindolone
(*both isomers at position shown)
802Me Step 2
‘0 Step 3
N SOZMe
O O
Step 1 and 2: Starting from Preparation 48, Steps 1 and 2 were performed using ures similar
to those described in Examples 73 and 74 Step 3 and Step 4, but using 1,1-
bis(hydroxymethyl)cyclopropane d of 1-(hydroxymethyl)cyclopropanecarboxamide. MS: [M
(cyclopropane-1,1-diyldimethanol]+ = 574
Step 3: (3R)[(4-chloromethanesulfonylpheny|)methyl](4-chlorophenyI)f|uoro
[hydroxy(oxanyl)methyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-
isoindolone
The product from Step 2 (300 mg, 0.44 mmol) was dissolved in ol (1 mL) under nitrogen at RT
with stirring. Sodium borohydride (25 mg, 0.66 mmol) was added to the reaction mixture and the
reaction was allowed to stir at room temperature for 10 min then quenched and diluted with water. The
reaction was extracted with DCM. The combined organic portions were dried (MgSO4) and
concentrated under reduced pressure. The crude e was purified on a 10 g SNAP silica cartridge,
eluting with ethyl acetate in isohexane (10 to 100%). Fractions containing pure product were
trated under reduced pressure to yield a white solid (247 mg) which was separated using chiral
SFC.
Example 83 *slower eluting isomer. 1H NMR (400 MHz, CDCI3) 7.91 (1H, d), 7.72 (1H, s), 7.35 (1H,
dd), 7.28 - 7.26 (2H, m), 7.24 - 7.16 (4H, m), 5.06 - 4.96 (2H, m), 4.55 (1H, dd), 4.05 - 3.93 (2H, m),
3.80 (1H, dd), 3.42 - 3.23 (4H, m), 3.04 (3H, s), 2.78 (1H, d), 2.11 (1H, d), 2.01 - 1.96 (1H, m), 1.94 -
1.84 (1H, m), 1.78 (1H, d), 1.54 - 1.37 (2H, m), 1.29 - 1.21 (1H, m), 0.54 - 0.48 (2H, m), 0.46 - 0.38
(1H, m), 0.22 (1H, d); MS: [M + H]+ = 678.
e 84 *faster eluting isomer. 1H NMR (400 MHz, CDCI3) 7.91 (1H, d), 7.67 (1H, s), 7.35 (1H,
dd), 7.30 - 7.26 (3H, m), 7.24 - 7.17 (3H, m), 5.06 - 4.96 (2H, m), 4.56 (1H, dd), 4.06 - 3.94 (2H, m),
3.82 (1H, dd), 3.40 - 3.25 (4H, m), 3.03 (3H, s), 2.75 (1H, d), 2.16 (1H, d), 2.03 - 1.99 (1H, m), 1.93 -
1.83 (1H, m), 1.78 (1H, d), 1.55 - 1.37 (2H, m), 1.28 (1H, d), 0.54 - 0.47 (2H, m), 0.46 - 0.40 (1H, m),
0.23 (1H, d); MS: [M + H]+ = 678.
e 85 and 86: 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-ch|orophenyl)-
7-fluoro[1-hydroxy(oxanyl)ethy|]oxo-2,3-dihydro-1H-isoindol
yl]oxy}methyl)cyclopropanecarboxamide
(*both isomers at on shown)
802Me
Using 2-(4-chloro(methylsulfonyl)benzyl)(4-chlorophenyl)fluorohydroxy(tetrahydro-2H-
pyrancarbonyl)isoindolinone (Examples 83 and 84; Step 1), the title compound was prepared
using methods similar to those described in Preparation 12 (using 1-
(hydroxymethyl)cyclopropanecarboxamide instead of 1,1-bis(hydroxymethyl)cyclopropane) and
Examples 73 and 74 Step 5.
Example 85 *slower g isomer. 1H NMR (400 MHz, CDCI3) 7.90 (1 H, d), 7.77 (1H, d), 7.50 - 7.46
(1H, m), 7.36 (1H, dd), 7.28 - 7.18 (5H, m), 6.52 - 6.52 (1H, m), 5.42 - 5.42 (1H, m), 5.06 - 4.95 (2H,
m), 4.07 - 3.94 (2H, m), 3.41 - 3.28 (3H, m), 3.00 (3H, s), 2.93 (1H, d), 1.91 - 1.84 (1H, m), 1.82 (1H,
s), 1.63 (3H, s), 1.56 (1 H, s), 1.53 - 1.40 (2H, m), 1.33 - 1.22 (3H, m), 0.53 - 0.47 (1H, m), 0.42 - 0.35
(1H, m); MS: [M + H]+ = 705.
Example 86 *fastereluting isomer. 1H NMR (400 MHz, coc13) 7.91 (1H, s), 7.81 (1H, s), 7.44 (1H, cl),
7.36 (1H, d), 7.26 - 7.19 (5H, m), 6.52 - 6.52 (1H, m), 5.42 (1H, s), 5.05 - 4.95 (2H, m), 4.06 - 3.96
(2H, m), 3.39 - 3.35 (3H, m), 3.00 (3H, s), 2.92 (1H, d), 1.91 - 1.84 (2H, m), 1.67 - 1.56 (4H, m), 1.47
(2H, s), 1.26 - 1.24 (3H, m), 0.51 (1H, s), 0.37 — 0.37 (1H, m); ; MS: [M + H]+ = 705
Exam ple 87: 5-chloro{[(1R)(4-chIorophenyl)fluoro[1-hydroxy(1-methy|piperidin
y|)ethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid
(Example isolated as a single isomer at the position shown*)
Step 1: tert-Butyl 4-[1-[2-[(2-bromochloro-phenyl)methyl](4-chlorophenyl)—7-fluoro
hydroxyoxo-isoindolinyl]hydroxy-ethyl]piperidinecarboxylate
Prepared in a r manner to that described for Example 35, step 4 from (-)[1-(1-ten‘-
butoxycarbonylpiperidyl)—1-hydroxy-ethyl](4-ch|orobenzoyl)fluoro-benzoic acid and (2-bromo-
4-chlorophenyl)methanamine. MS: [M - HZO]+= 691.
Step 2: 2-[(2-BromochIoro-phenyl)methyl](4-chlorophenyI)fluoro[1-hydroxy(4-
piperidyl)ethyl]methoxy-isoindolinone
Prepared in a similar manner to that described in Preparation 10 from utyl 4-[1-[2-[(2-bromo
-phenyl)methyl](4-chlorophenyl)fluorohydroxyoxo-isoindolinyl]hydroxy-
ethyl]piperidinecarboxylate and methanolMS: [M + H]+ = 623.
Step 3: 2-[(2-Bromochloro-phenyl)methyl](4-chlorophenyl)fluoro[1-hydroxy(1-
methylpiperidyl)ethyl]methoxy-isoindolinone
The title compound was prepared from 2-[(2-bromoch|oro-phenyl)methyl](4-chlorophenyl)
fluoro[1-hydroxy(4-piperidyl)ethyl]methoxy-isoindolinone in analogous fashion to Example
81, step 5. MS: [M + H]+= 637.
Step 4: 5-Chloro[[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl
piperidyl)ethyl]methoxyoxo-isoindolinyl]methyl]benzoic acid
Prepared in a similar manner to that described for Example 33, step 5 from 2-[(2-bromochlorophenyl
l](4-chlorophenyl)fluoro[1-hydroxy(1-methylpiperidyl)ethyl]methoxy-
isoindolinone. 1H NMR (400 MHz, CDCI3) 7.87 (1H, s), 7.53 (1H, d), 7.49 (1H, d), 7.31 (2H, d), 7.24
(2H, d), 7.19 (1H, d), 7.11 (1H, dd), 4.96 (1H, d), 4.81 (1H, d), 3.47 (1H, d), 3.36-3.32 (1H, m), 2.87
(3H, s), .71 (2H, m), 2.73 (3H, s), 2.11 - 2.02 (1H, m), 1.99 - 1.90 (1H, m), 1.80 - 1.63 (2H, m),
1.62 (3H, s), 1.51 (1H, m). MS: [M + H]+= 601.
Examples 88 and 89: (3S)(4-chlorophenyl)[(1 R)(4-chlorophenyl)fluoro[1-hydroxy
(oxanyl)ethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
(*both isomers separated and isolated)
Step 1: (38)—Ethy| hlorophenyI)(1-(4-chlorophenyl)fluorohydroxyoxo
(tetrahydro-2H-pyrancarbonyl)isoindolinyl)propanoate
2-(4-Chlorobenzoyl)fluoro(tetrahydro-2H-pyrancarbonyl)benzoic acid ration 48) (3.99
g, 10.2 mmol) was reacted with (S)—ethyl 3-amino(4-chlorophenyl)propanoate hydrochloride (3.5 g,
13.25 mmol) in an analogous fashion as described in Example 35, step 4 to afford the title compound
(3.4 g, 50%) as a yellow solid. MS: [M - H]' = 598.
Step 2: (S)-Ethy| hlorophenyl)—3-((R)(4-chlorophenyl)fluoromethoxyoxo
(tetrahydro-2H-pyrancarbonyl)isoindolinyl)propanoate
The title compound was prepared from (3S)—ethyl 3-(4-chlorophenyl)(1-(4-chlorophenyl)fluoro
hydroxyoxo(tetrahydro-2H-pyrancarbonyl)isoindolinyl)propanoate and methanol in a similar
manner as described in Preparation 12. The diastereoisomers were separated chiral SFC. [M + H]+ =
614.
Step 3: (S)(4-Chlorophenyl)((R)(4-chlorophenyl)fluoromethoxyoxo
(tetrahydro-2H-pyrancarbonyl)isoindolinyl)propanoic acid
The title compound was prepared from (S)—ethyl 3-(4-chlorophenyl)((R)(4-chlorophenyI)fluoro-
oxyoxo(tetrahydro-2H-pyrancarbonyl)isoindolinyl)propanoate in an analogous
fashion as described in Preparation 40. [M + H]+ = 586.
Step 4: (3S)(4-chlorophenyl)—3-[(1R)(4-chlorophenyl)—7-fluoro[1-hydroxy(oxan
yI)ethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
The title compound was prepared from (4-chlorophenyl)((R)(4-chloropheny|)fluoro
methoxyoxo(tetrahydro-2H-pyrancarbonyl)isoindolinyl)propanoic acid in a similar manner to
that bed in Example 1 to give the racemate which was separated by chiral SFC.
e 88: *fast running isomer 1H NMR (400 MHz, CDCI3) 7.67 (1H, d), 7.34 - 7.29 (1H, m), 7.03
(4H, s), 7.00 (4H, s), 4.68 (1H, dd), 4.04 - 3.90 (2H, m), 3.68 (1H, dd), 3.36 - 3.24 (3H, m), 3.08 (3H,
s), 1.86 - 1.77 (1H, m), 1.61 — 1.55 (4H, m), 1.47 - 1.36 (2H, m), 1.26 - 1.17 (1H, m), (OH and COZH
not visible). [M + Hr = 602.
Example 89: *slow running isomer‘H NMR (400 MHz, CDCI3) 7.68 (1H, s), 7.29 (1H, d), 7.03 (4H, s),
7.00 (4H, s), 4.69 (1H, dd), 4.05 - 3.90 (2H, m), 3.69 - 3.61 (1 H, m), 3.37 - 3.24 (3H, m), 3.09 (3H, s),
1.87 - 1.77 (1H, m), 1.63 — 1.56 (4H, m), 1.50 - 1.37 (2H, m), 1.20 (1H, d), (OH and COZH not visible).
[M + H]+ = 602.
Examples 90 and 91: 4-[(1R)[(1R)(4-Chloropheny|)fluoro[1-hydroxy(1-methyl-1H-
imidazolyl)propyl]oxo[(3S)-oxo|anyloxy]-2,3-dihydro-1H-isoindolyl]
hydroxyethyl]benzonitrile
(*both isomers separated and isolated)
\ \\
\N N
Step 1, 2 and 3: Starting from Preparations 49 and 50, Steps 1, 2 and 3 were performed using
procedures similarto those described in Example 73 Step 3, Step 4 and Step 5, but using and (38)-
hydroxy-tetrahydrofuran instead of 1-(hydroxymethyl)cyclopropanecarboxamide in Step 2 and
EtMgCl instead of MeMgCl in Step 3.
Step 4: 4-[(1R)[(1R)(4-Chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazol
yl)propyl]oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindol-Z-yl]
hydroxyethyl]benzonitrile
Pd(PPh3)4 (56 mg, 0.05 mmol) and K2C03 (252 mg, 1.88 mmol) were added to a solution of 4-[(1 R)
[(1R)(4-chlorophenyl)fluoro[1-hydr0xy(1-methy|-1H-imidazolyl)propyl]oxo[(3S)-
oonanyloxy]-2,3-dihydro-1H-isoindoly|](propenyloxy)ethyl]benzonitrile (630 mg, 0.94
mmol) in MeOH (10 mL) and the resulting mixture was stirred at 80°C for 2 hours. The solvent was
then d in vacuo and the residue was purified by flash chromatography on silica gel (gradient (0-
100% EtOAc in ). The two reoisomers were then separated by chiral HPLC.
Example 90, isomer 1 (73 mg, 12% yield): 1H NMR (400 MHz, DMSO-d6): 7.90 (1H, s), 7.58-7.50
(2H, m), 7.47 (2H, d), 7.18 (2H, d), 7.07-8.82 (5H, m), 5.58 (1H, s), 5.13 (1H, dd), 4.49-4.40 (1H, m),
4.35-4.24 (2H, m), 4.10-3.98 (1H, m), 3.87 (1H, q), 3.89-3.82 (1H, m), 3.81 (3H, s), 3.48-3.42 (1H, m),
3.20-3.08 (1H, m), 2.28-2.05 (4H, m), 0.70 (3H, t); LCMS: [M+H]+= 831.
Example 91, isomer 2 (97 mg, 18% yield): 1H NMR (400 MHz, DMSO-d6): 7.85 (1H, s), 7.81-7.51
(2H, m), 7.48 (2H, d), 7.18 (2H, d), .83 (5H, m), 5.58 (1H, s), 5.13 (1H, t), 4.49-4.39 (1H, m),
.24 (2H, m), 4.09-3.97 (1H, m), 3.87 (1H, q), 3.70-3.58 (4H, m), 3.43-3.37 (1H, m), 3.11 (1H,
dd), 2.25-2.02 (4H, m), 0.70 (3H, t); LCMS: [M+H]"= 831.
Examples 92 and 93: 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methy|-1H-imidazol-
4-yl)propy|]oxo[(3S)-oxoIanyloxy]-2,3-dihydro-1H-isoindolyl]methyl}
(hydroxymethyl)benzonitrile
(*both isomers separated and isolated)
The title nds were prepared in a similarfashion as in Example 90, but using Preparation 51
instead of 49 in step 1.
Example 92: *fast running isomer 1H NMR (400 MHz, CDCI3) 7.81 (1H, d), 7.57 (1H, d), 7.45 - 7.38
(2H, m), 7.38 (1H, d), 7.28 (1H, d), 7.20 (4H, s), 8.83 (1 H, d), 4.74 - 4.85 (2H, m), 4.58 (1H, dd), 4.42
(1H, d), 3.85 - 3.70 (2H, m), 3.89 (3H, s), 3.82 - 3.53 (3H, m), 3.23 (1 H, dd), 2.75 - 2.88 (1H, m), 2.23 -
2.08 (2H, m), 1.48 - 1.37 (2H, m), 0.84 (3H, dd). [M + H]+ = 831.
Example 93: *slow running isomer‘H NMR (400 MHz, CDCI3) 7.89 (1H, d), 7.58 - 7.51 (2H, m), 7.41 -
7.38 (2H, m), 7.25 - 7.23 (1H, m), 7.19 (4H, s), 8.85 (1H, d), 4.73 - 4.88 (2H, m), 4.58 (1H, d), 4.42
(1H, d), 3.88 - 3.72 (2H, m), 3.70 (3H, s), 3.82 - 3.54 (3H, m), 3.22 (1 H, dd), 2.23 - 2.08 (2H, m), 1.50 -
1.40 (2H, m), 1.27 - 1.08 (1H, m), 0.84 (3H, dd). [M + H]+ = 831.
Examples 94 and 95: 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazol-
4-yl)propyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindol-Z-
yl]methyl}benzonitrile
(*both isomers separated and isolated)
F O HO O
N O Step1 N
(\N‘ O \ O N
OH <\N
O 0
o o
Step 2
Step 3 <
gfiiF
Step 1 and 2: Starting from Preparation 50, Steps 1 and 2 were performed using procedures similar
to those described in Examples 73 Step 3 and Step 4 but using 4-(aminomethyl)benzonitrile in Step 1
and [1-(hydroxymethyl)cyclopropyl]methanol in Step 2.
Step 3: 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]
{[1-(hydroxymethyl)cycIopropyl]methoxy}oxo-2,3-dihydro-1H-isoindol-Z-
yl]methyl}benzonitrile
A solution of (R)((1-(4-chlorophenyl)fluoro((1-(hydroxymethyl)cyclopropyl)methoxy)(1-
methyl-1H-imidazolecarbonyl)oxoisoindolinyl)methyl)benzonitrile) (190 mg, 0.325mmol), In
anhydrous CHZCIZ (10 mL) was stirred at 0 °C under nitrogen . AlEt3 (2.5 mL, 1M in hexanes, 2.5
mmol) was added drop-wise and the mixture was stirred at 0 °C for 1 h. The on was quenched
with sat. aq. NH4Cl and diluted with CHZCIZ (10mL) and water (7 mL). Solids were removed by
filtration and the filtrate was ted with CHZCIZ (2 x 15 mL). Combined organics were dried
(MgSO4) and the solvent evaporated. The crude residue was purified by column chromatography,
him, 12 g KP-sil cartridge 0-5% MeOH in EtOAc to afford the racemate (98 mg, 49%). Chiral
ative HPLC gave the title nds:
Example 94 (faster running isomer) 1H NMR (400 MHz, CDCI3) 7.71 (1H, s), 7.47 (3H, dd), 7.36 (1H,
s), 7.27-7.26 (2H, m), 7.18 (4H, s), 6.84 (1H, s), 4.49 (1H, d), 4.32 (1H, d), 3.69 (3H, s), 3.53 (1H, d),
2016/053042
3.41 (1H, d), 2.97 (1H, d), 2.79 (1H, d), 2.24 - 2.07 (2H, m), 1.25 (1H, s), 0.86 (3H, dd), 0.45 (2H, dd),
0.28 - 0.23 (1H, m), 0.15 - 0.09 (1H, m). MS: [M+H]*=615
Example 95: (slower g isomer) 1H NMR (400 MHz, CDCI3) 7.71 (1H, s), 7.49 (3H, dd), 7.36
(1H, s), 7.28-7.27 (2H, m), 7.19 (4H, s), 6.85 (1H, s), 4.51 (1H, d), 4.31 (1H, d), 3.69 (3H, s), 3.55 (1H,
s), 3.52 - 3.47 (1H, m), 3.41 (1H, dd), 2.96 (1H, d), 2.78 (1H, d), 2.22 - 2.06 (2H, m), 0.85 (3H, dd),
0.44 (2H, dd), 0.29 - 0.23 (1H, m), 0.15 - 0.08 (1 H, m). MS: [M+H]+=615
Examples 96 and 97: 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazol-
4-yl)propyl]oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile
(*both isomers separated and isolated)
The title nds were prepared in a similar manner as in Example 94 and Example 95, but using
(3S)—hydroxy-terahydrofuran in Step 2. Chiral preparative HPLC gave the title compounds.
Example 96: 1H NMR (400 MHz, CDCI3) 7.70 (1H, d), 7.55 - 7.52 (1H, m), 7.46 (2H, d), 7.36 (1H, s),
7.27 - 7.24 (2H, m), 7.16 (4H, s), 6.85 (1H, d), 4.50 (1H, d), 4.32 (1H, d), 3.84 - 3.76 (2H, m), 3.70 (3H,
s), 3.63 - 3.52 (3H, m), 3.23 (1H, dd), 2.24 - 2.07 (2H, m), 1.51 - 1.44 (2H, m), 0.84 (3H, t). MS:
[M+H]*=601
Example 97: 1H NMR (400 MHz, CDCI3) 7.81 (1H, d), 7.48 - 7.42 (3H, m), 7.36 (1H, s), 7.28 - 7.25
(2H, m), 7.17 (4H, s), 6.83 (1H, d), 4.51 (1H, d), 4.32 (1H, d), 3.82 - 3.75 (2H, m), 3.69 (3H, s), 3.62 -
3.54 (3H, m), 3.25 (1H, dd), 2.23 - 2.07 (2H, m), 1.49 - 1.42 (2H, m), 0.84 (3H, t). MS: [M+H]+=601.
Examples 98 and 99: (3S)(4-chloropheny|)[(1R)(4-chlorophenyl)fluoro[1-(4-
fluorooxanyl)hydroxyethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
(*both isomers separated and isolated)
Step 1 ~30
O COZEt
Step 2
Step 3
Step 1 and 2: Ethyl (38)—3-(4-chlorophenyl)—3-(1-(4-chlorophenyl)fluoro(4-fluorotetrahydro-
2H-pyrancarbonyl)hydroxyoxoisoindolinyl)propanoate
Starting from h|0robenzoyl)f|uor0(4-fluorotetrahydro-2H-pyrancarbonyl)benzoic acid
(Preparation 53), hyl 3-amino(4-ch|0r0pheny|)propanoate hydrochloride (Preparation 35)
and MeOH, Steps 1-2 were performed by following procedures similarto those described in Example
, step 4 and Preparation 10 respectively. The diastereoisomers were separated using chiral SFC
to give ethyl (S)(4-ch|or0phenyl)((R)(4-chlor0pheny|)f|uoro(4—fluor0tetrahydro-2H-pyran-
4-carbonyl)—1-methoxyoxoisoindo|iny|)propanoate. MS: [M-MeOH]+ = 600.
Step 3: (S)(4-Chlorophenyl)((R)(4-chlorophenyl)fluoro(4-fluorotetrahydro-2H-
pyrancarbonyI)methoxyoxoisoindolinyl)propanoic acid
Step 3 was performed using procedures rto those described in Preparation 52, Step 3. MS: [M
- H]' = 602.
Step 4: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)—7-fluoro[1-(4-fluorooxanyl)
hydroxyethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
Step 4 was performed using procedures similar to those described in Example 73, Step 5.
Example 98: *fast g isomer: 1H NMR (400 MHz, CDCI3) 7.78 (1H, s), 7.39 (1H, d), 7.04 (4H, s),
7.00 (4H, s), 4.88 (1H, dd), 3.89 - 3.56 (5H, m), 3.31 (1H, dd), 3.10 (3H, s), 1.98 - 1.80 (7H, m), 1.44
(1H, dd); COOH missing. MS: [M+H]+ = 820.
Example 99: * Slow running isomer: 1H NMR (400 MHz, CDCI3) 7.79 (1H, s), 7.39 (1H, d), 7.03 (4H,
s), 7.00 (4H, s), 4.89 (1H, dd); 3.90 - 3.58 (5H, m), 3.30 (1H, dd), 3.11 (3H, s), 1.97 - 1.73 (3H, m),
1.73 - 1.83 (4H, m), 1.47 - 1.39 (1H, m); COOH missing. MS: [M+H1+ = 820.
Exam ple 100: (4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)—7-fluoro[1-hydroxy(1-
methyl-1H-pyrazolyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindol-Z-yl]butanoic acid
(*prepared as a e of epimers at the position shown)
Starting from Preparation 65 and methyl —amino(4—chlorophenyl)butanoate, the title
compound was prepared using procedures similar to those described in Example 99; but using
EtMgCI/ZnCIz instead of MeMgCI in the final step. 1H NMR (400 MHz, CDCI3): 7.74 (1 H, d), .44
(1H, m), 7.34 (1H, d), 7.04-7.00 (8H, m), 6.21 (1H, t), 4.23-4.15 (1H, m), 3.88 (3H, s), 3.19 (3H, d),
3.11-3.02 (1H, m), 2.55-2.44 (2H, m), 2.32-2.11 (6H, m), 0.90-0.85 (4H, m). MS: [M+H]+ = 626.
Example 101 and 102: (38)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-
fluorooxany|)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic
acid
(*both isomers separated and ed)
Starting from (S)(4-chlorophenyI)((R)(4-chIorophenyl)fluoro(4-fluorotetrahydro-2H-pyran-
4-carbonyl)methoxyoxoisoindolinyl)propanoic acid le 99 step 3), the title compounds
were prepared using procedures similar to those described in Preparation 52, Step 1. The title
compounds, prepared as a mixture, were subsequently separated using chiral SFC.
Example 101: *fast running isomer: 1H NMR (400 MHz, CDCI3) 7.74 (1H, s), 7.39 (1H, d), 7.04 (4H,
s), 7.01 (4H, d), 4.68 (1H, dd), 3.88 - 3.57 (5H, m), 3.30 (1H, dd), 3.10 (3H, s), 2.25 - 2.15 (2H, m),
2.05 - 1.81 (3H, m), 1.66 - 1.42 (2H, m), 0.69 (3H, t); COOH missing. MS: [M+H]+ = 634.
Example102: * Slow running isomer: 1H NMR (400 MHz, CDCI3) 7.75 (1H, s), 7.37 (1H, d), 7.04 (4H,
s), 7.01 (4H, s), 4.70 (1H, dd), 3.87 - 3.58 (5H, m), 3.29 (1H, dd), 3.11 (3H, s), 2.25 - 2.15 (2H, m),
2.02 - 1.80 (3H, m), 1.71 - 1.51 (1 H, m), 1.43 (1H, dd), 0.69 (3H, t); COOH missing. MS: [M+H]+ = 634.
Example 103 and 104: (3S)(4-chIorophenyl)[(1R)(4-chlorophenyI)(1-cyc|obutyl
hydroxyethyl)fluoromethoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
(*both isomers separated and isolated)
Starting from 2-(4-chlorobenzoyI)(cyclobutanecarbonyI)fluorobenzoic acid (Preparation 55) the
title nd was prepared using methods similar to those described for Example 98. Except that
2M aqueous HCI on was used instead of LiOH in Step 3.
Example 103: *fast running isomer: 1H NMR (400 MHz, CDCI3) 7.70 (1 H, s), 7.31 (1 H, d), 7.04 - 6.99
(4H, m), 6.98 (4H, s), 4.67 (1H, dd), 3.61 - 3.60 (1H, m), 3.33 - 3.25 (1H, m), 3.06 (3H, s), 2.74 - 2.65
(1H, m), 2.05 - 1.93 (2H, m), 1.87 - 1.73 (2H, m), 1.66 (1H, d), 1.56 - 1.54 (1H, m), 1.44 (3H, s); OH
and COOH missing; MS: [M+H]+ = 572.
Example 104: * Slow running isomer: 1H NMR (400 MHz, CDCI3) 7.71 (1H, s), 7.29 (1H, cl), 7.05 -
6.93 (8H, m), 4.70 (1H, dd), 3.59 (1H, s), 3.22 - 3.17 (1H, m), 3.06 (3H, s), 2.74 - 2.65 (1H, m), 2.07 -
1.66 (5H, m), 1.61 1H, m), 1.44 (3H, s); OH and COOH missing; MS: [M+H]+ = 572.
Exam ple 105: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy
(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
Step 1: Ethyl (3S)(4-chlorophenyl)(1-(4-chlorophenyl)fluorohydroxy((S)hydroxy-
1-(tetrahydro-2H-pyranyl)propyl)oxoisoindolinyl)propanoate
(S)(4-Chlorobenzoyl)—3-f|uoro(1-hydroxy(tetrahydr0-2H-pyrany|)propy|)benzoic acid
(Preparation 52, 1.43 g, 3.39 mmol) was reacted with (S)-ethy| 3-amino(4-ch|oropheny|)propanoate
hydrochloride (Preparation 35, 1.16 g, 4.41 mmol) in an analogous fashion as described in Example
, step 4 to afford the title compound (1.37 g, 64%) as a colourless foam. MS: [M - H]' = 628.
Step 2: Ethyl (4-chlorophenyl)((R)(4-chlorophenyl)fluoro((S)hydroxy
(tetrahydro-2H-pyranyl)propyl)methoxyoxoisoindolinyl)propanoate
The title compound was prepared from ethyl (3S)(4—chIorophenyI)(1-(4-ch|orophenyl)f|uoro
hydroxy((S)hydroxy(tetrahydro-2H-pyran-4—yl)propyl)oxoisoindolinyl)propanoate (1.3 g,
2.06 mmol) and methanol (0.83 mL, 20 mmol) in a similar manner as described in Preparation 10.
The diastereoisomers were ted using chiral SFC giving the title compound (0.39 g) as a pale
yellow foam. MS: [M - MeOH]+= 612.
Step 3: (3S)(4-chlorophenyI)[(1R)(4-chIorophenyl)f|uoro[(1S)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
The title compound was prepared from ethyl (S)(4-ch|or0phenyl)((R)(4-chloropheny|)f|uoro-
-((S)hydroxy(tetrahydro-2H-pyranyl)propyI)methoxyoxoisoindo|iny|)propanoate
(0.391 g, 0.6 mmol) using procedures similar to those described in Preparation 52, Step 3. The
crude product was purified by preparative HPLC to give pure title compound (80 mg) as a white solid.
1H NMR (400 MHz, CDCI3) 7.62 (1H, d), 7.29 (1H, dd), 7.04 (4H, s), 7.00 (4H, s), 4.68 (1H, dd), 4.03
(1H, dd), 3.90 (1H, dd), 3.74 (1H, dd), 3.40 - 3.25 (3H, m), 3.09 (3H, s), 1.97 - 1.86 (3H, m), 1.73 (1H,
d), 1.49 - 1.35 (2H, m), 1.07 (1H, d), 0.68 (3H, t); OH and COOH missing. MS: [M+H]+ = 616.
Starting from the appropriate chiral acid intermediate (e.g. ation 52, ation 52b or
Preparation 54), the following Examples were prepared using ures similar to those described
in Example 105 steps 1-3. An appropriately protected or- or B-amino acid was used in Step 1 [e.g.
methyl (S)—4-amino(4-chlorophenyl)butanoate or (S)-ethy| 3-amino(4-ch|oropheny|)propanoate
hydrochloride (Preparation 35)] and an appropriate alcohol (e.g. MeOH, EtOH, CD30H) was used in
Step 2.
In some cases the product was isolated as a tris(hydroxymethyl)aminomethane (TRIS) salt (by
dissolving in MeOH, treated with ydroxymethyl)aminomethane and ation).
Example 106: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy
(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
WO 55860
1H NMR (400 MHz, 00013) 7.62 (1H, d), 7.28 (2H, d), 7.04 (4H, s), 7.01 (4H, s), 4.68 (1H, dd), 4.03
(1H, dd), 3.89 (1H, dd), 3.73 (1H, dd), 3.42 - 3.25 (3H, m), 3.10 (3H, s), 1.96 - 1.85 (3H, m), 1.74 (1H,
d), 1.49 - 1.36 (2H, m), 1.06 (1H, d), 0.68 (3H, t); COOH missing. MS: [M+H]+ = 616.
Exam ple 107: (4S)(4-ch|oropheny|)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy
(oxany|)propyI]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid
1H NMR (400 MHz, 00013) 7.61 (s, 1H), 7.06 - 7.01 (m, 4H), 6.99 (d, 2H), 4.19 (dd, 1H), 4.02 (dd, 1H),
3.90 (dd, 1H), 3.40 - 3.24 (m, 2H), 3.19 (s, 3H), 3.13 - 3.03 (m, 1H), 2.54 - 2.44 (m, 1H), 2.32 - 2.11
(m, 2H), 2.07 - 1.96 (m, 1H), 1.96 - 1.84 (m, 3H), 1.73 (d, 3H), 1.47 - 1.36 (m, 3H), 1.27 - 1.16 (m, 1H),
1.07 (d, 1H), 0.69 (dd, 2H). MS: [M+H]+ = 630.
Exam ple 108: (4S)(4-ch|oropheny|)[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy
(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid
1H NMR (400 MHz, CDCI3) 7.62 - 7.59 (3H, m), 7.41 (2H, d), 7.35 (2H, d), 7.31 - 7.26 (3H, m), 4.09 -
4.00 (2H, m), 3.91 (1H, dd), 3.41 - 3.27 (2H, m), 2.88 - 2.77 (1H, m), 2.51 - 2.42 (4H, m), 2.03 - 1.97
(2H, m), 1.97 - 1.85 (3H, m), 1.73 (1H, d), 1.48 - 1.38 (2H, m), 1.08 (1H, d), 0.70 - 0.64 (3H, m); OH
and COOH missing. MS: [M+H]+ = 630.
Exam ple 109: (4S)(4-Chloropheny|)[(1R)(4-chlorophenyl)fluoro[(1R)-1 -(4-
fluorooxany|)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid
(tris(hydroxymethyl)aminomethane salt)
’]
1H NMR (400 MHz, DMSO) 7.76 (1H, s), 7.48 (1H, d), 7.18 - 7.12 (4H, m), 7.10 - 7.02 (4H, m), 4.25
(1H, dd), 3.88 (1H, dd), 3.72 (1H, dd), 3.56 - 3.50 (1H, m), 3.47 - 3.39 (1H, m), 3.37 (6H, s), 3.18 (3H,
s), 2.84 - 2.73 (1H, m), 2.48 - 2.38 (1H, m), 2.26 - 2.17 (1H, m), 2.05 - 1.88 (6H, m), 1.07 - 0.99 (1H,
m), 0.63 (3H, t); 7 protons missing (OH x 4, NH2 and COOH). MS: [M+H]+ = 648.
Example 110: -(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)(4-
fluorooxany|)hydroxypropyl]trideuteromethoxyoxo-2,3-dihydro-1H-isoindol-Z-
yl]propanoic acid
1H NMR (400 MHz, 00013) 7.61 (1H, s), 7.26 (1H, d), 7.07 - 6.99 (8H, m), 4.19 (1H, dd), 3.51 - 3.43
(1H, m), 3.19 (3H, s), 3.14 - 3.04 (1H, m), 2.53 - 2.44 (1H, m), 2.30 - 1.85 (7H, m), 1.67 - 1.60 (1H, m),
1.31 - 1.07 (5H, m), 0.66 (3H, dd). MS: [M+H]+ = 637.
Example 111 : (33)(4-chlorophenyl)[(1R)(4-ch|orophenyl)ethoxyfluoro[(1R)(4-
fluorooxany|)hydroxypropyl]oxo-2,3-dihydro-1H-isoindolyl]propanoic acid
1H NMR (400 MHz, 00013) 7.73 (1H, s), 7.36 (1H, d), 7.06 - 7.00 (8H, m), 4.69 (1H, dd), 3.86 - 3.75
(3H, m), 3.66 - 3.57 (2H, m), 3.30 - 3.12 (3H, m), 2.26 - 2.13 (2H, m), 2.02 - 1.82 (3H, m), 1.67 - 1.40
(3H, m), 1.28 (3H, dd), 0.68 (3H, dd). MS: [M+H]+ = 637.
Exam ple 113: (4S)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)
hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindo|y|](4-methoxyphenyl)butanoic
acid
1H NMR (400 MHz, CDCI3) 7.73 (1H, s), 7.35 (1H, d), 7.05 - 6.93 (6H, m), 6.58 (2H, d), 4.16 (1H, dd),
3.81 (2H, d), 3.73 (3H, s), 3.66 - 3.58 (2H, m), 3.17 (3H, s), 3.11 - 3.10 (1H, m), 2.51 - 2.42 (1H, m),
2.31 - 2.28 (4H, m), 2.27 - 2.13 (3H, m), 1.89 - 1.44 (2H, m), 0.69 (3H, dd); COOH not observed. MS:
[M+H]+ = 644.
Exam ple 114: (4S)(4-chlorophenyl)[(1R)(4-chIorophenyl)f|uoro{1-hydroxy[trans-
oxycyclohexyl]propy|}methoxyoxo-2,3-dihydro-1H-isoindol-Z-yl]butanoic acid
(*Example prepared and isolated as a single isomer at the position )
HO,“
Starting from the single isomer of 2-(4-chIorobenzoyl)fluoro(1-hydroxytrans—4-
hydroxycyclohexyl)propyl)benzoic acid (Preparation 64, Step 3), the title compound was prepared by
following procedures similarto those described in Example 105. 1H NMR (400 MHz, CDCI3) 7.61 (1H,
s), 7.26 (1H, d), 7.07 - 6.99 (8H, m), 4.19 (1H, dd), 3.51 - 3.43 (1H, m), 3.19 (3H, s), 3.14 - 3.04 (1H,
m), 2.53 - 2.44 (1H, m), 2.30 - 1.85 (7H, m), 1.67 - 1.60 (1H, m), 1.31 - 1.07 (5H, m), 0.66 (3H, dd),
exchangeable not observed. MS: [M+H]+= 644.
Exam ple 115: 2-(5-chloro{[1-(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)
hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindo|y|]methy|}phenoxy)acetic acid
(tris(hydroxymethyl)aminomethane salt)
Starting from (4-chlorobenzoyl)fluoro(1-(4-fluorotetrahydro-2H-pyranyl)
hydroxypropyl)benzoic acid (Preparation 54) and ethyl 2—[2-(aminomethyl)—5-chlorophenoxy]acetate
hydrochloride (Preparation 63), the title compound was prepared by following procedures similar to
those described in Example 105.1H NMR (400 MHz, DMSO-d5 —DZO): 7.71 (1H, s), 7.40 (1H, d), 7.26-
7.17 (4H, m), 6.91 (1H, d), 6.67 (1H, dd), 6.50 (1H, d), 4.38 (2H, s), .00 (2H, m), 3.84-3.74 (1H,
m), 3.66 (1H, dd), 3.46 (7H, s), 2.84 (3H, s), .03 (1H, m), 1.99-1.74 (4H, m), 1.14 (1H, d), 1.04-
0.95 (1H, m), 0.56 (3H, t).MS:[M + H]+ = 650.
Exam ple 116: 5-chloro{[(1R)(4-chIorophenyl)f|uoro[(1S)hydroxy(oxan
y|)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid
cr cr
/ F /
F o
\ [: ]
-“ Step3 "
O O
N Br —. COZH
I OH ob OH 0
Step 1 and Step 2: Starting from (-)-(S)(4-chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-
pyranyl)propyl)benzoic acid (Preparation 52), (2-bromochlorophenyl)methanamine (Example
33, Step 1) and MeOH, Step 1 and Step 2 were performed by following procedures similar to those
described in Example 35, step 4 and ation 10 respectively Chiral separation using
supercritical fluid chromatography gave (R)(2-bromochlorobenzyl)(4-chlorophenyl)fluoro
((S)—1-hydroxy(tetrahydro-2H-pyranyl)propyl)methoxyisoindolinone. MS: [M+H]+ = 638
Step 3: 5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]
methoxyoxo-2,3-dihydro-1H-isoindoIyl]methyl}benzoic acid
The title compound was prepared in a similar manner to that described for Example 33, step 5. 1H
NMR (400 MHz, DIVISO) 7.78 (1H, s), 7.69 (1H, s), 7.50 (1H, d), 7.37 - 7.31 (5H, m), 7.24 (1H, s), 4.98
(1H, s), 4.94 - 4.81 (2H, m), 3.98 - 3.93 (1H, m), 3.82 (1H, dd), 3.40 - 3.30 (1H, m), 3.28 - 3.21 (1H,
m), 2.88 (3H, s), 2.05 - 1.90 (3H, m), 1.73 (1H, d), 1.48 - 1.30 (2H, m), 1.02 (1H, d), 0.65 (3H, t),
COOH missing. MS: [M + H]+= 602.
Starting from the appropriate chiral acid intermediate (e.g. Preparation 52, Preparation 54), the
following Examples were prepared using procedures similarto those described in e 116 steps
1-3. The appropriate benzylamine [e.g. (2-bromochlorophenyl)methanamine, Preparation 58 or
Preparation 59] was used in Step 1 and an appropriate l used in Step 2. In some cases the
product was isolated as a tris(hydroxymethyl)aminomethane (TRIS) salt (by dissolving in MeOH,
treated with ydroxymethyl)aminomethane and evaporation). Purification by preparative HPLC
gave the products as single isomers, with the configuration shown.
Exam ple 117: 5-chloro{[(1R)(4-chIorophenyl)f|uoro[(1R)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methy|}benzoic acid
FogCI/
N COZH
Prepared from (+)-(R)—2-(4-chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyran
yl)propyl)benzoic acid (Preparation 52b). 1H NMR (400 MHz, CDCI3) 7.73 (2H, d), 7.30 - 7.18 (6H, m,
overlapping CHCI3), 7.14 (1H, d), 5.12 (1H, d), 4.64 (1H, d), 4.01 (1H, dd), 3.84 (1H, dd), 3.37 - 3.19
(2H, m), 2.81 (3H, s), 1.98-1.71 (4H, m), 1.71 (1H, d), 1.45 - 1.33 (2H, m), 1.05 - 0.99 (1H, m), 0.64
(3H, t). COOH missing MS: [M + H]+= 602.
Exam ple 118: 5-chloro{[(1R)(4-ch|oropheny|)ethoxyf|uoro[(1S)hydroxy(oxan-
4-yl)propyl]oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid -
(tris(hydroxymethyl)aminomethane salt)
1H NMR (400 MHz, CDCI3) 7.79 (1H, s), 7.50 (1H, s), 7.21 (2H, d), 7.08 - 7.00 (2H, m), 6.82 (1H, d),
.02 (2H, s), 4.59 - 4.59 (3H, m), 3.97 - 3.94 (1H, m), 3.85 - 3.81 (1H, m), 3.73 (6H, s), 3.33 - 3.17 (2H,
m), 3.06 - 2.94 (2H, m), 1.84 - 1.59 (4H, m), 1.43 - 1.34 (2H, m), 1.00 (4H, dd), 0.55 (3H, t), one
exchangeable proton not observed. MS: [M+H]+= 616.
Exam ple 119: R)(4-chlorophenyl)f|uoro[(1R)(4-fluorooxanyl)
hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindoly|]methy|}methylbenzoic acid
1H NMR (400 MHz, CDCI3) 7.78 (1H, s), 7.69 (1H, s), 7.40 - 7.31 (4H, m), 7.26 - 7.19 (3H, m), 4.99
(1H, d, J=15.4 Hz), 4.83 (1H, d, J=15.4 Hz), 3.86 - 3.77 (2H, m), 3.68 - 3.57 (2H, m), 2.80 (3H, s), 2.34
(3H, s), 2.25 - 2.14 (1H, m), 2.00 - 1.80 (2H, m), 1.70 - 1.42 (3H, m), 1.26 (1H, s), 0.69 (3H, dd);
COOH not observed. [M+H]+ = 601
Exam ple 120: 2-{[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)
ypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}methoxybenzoic acid -
tris(hydroxymethyl)aminomethane salt
_ HO OH
O \7<:+
H3N OH
1H NMR (400 MHz, CDCI3) 7.83 (1H, s), 7.32 - 7.18 (5H, m), 7.13 - 7.13 (1H, m), 6.95 (1H, d), 6.68
(1H, d), 4.92 (2H, d), 4.65 (1H, d), 3.82 - 3.78 (1H, m), 3.72 (6H, s), 3.61 - 3.48 (4H, m), 2.87 (3H, s),
2.62 (1H, s), 2.08 - 2.00 (1H, m), 1.94 - 1.60 (6H, m), 1.26 - 1.16 (1H, m), 0.55 (3H, s). [M+H]+ = 616
Exam ple 121: 2-(5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxan
pyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)methylpropanoic
acid (tris(hydroxymethyl)aminomethane salt)
Cl F /
/ 0 0
O .-\
\ O'
- o— Step2 o
O N
__ o
o “ OH o
I‘ OH O I
OH CI
+H N3 \g/OH
Step 1: Methyl 2-(5-chloro(((R)(4-chlorophenyl)fluoro((S)hydroxy(tetrahydro-2H-
pyranyl)propyl)methoxyoxoisoindolinyl)methyl)phenyl)methylpropanoate
A diastereomeric mixture at C-3 of 2-(2-bromochIorobenzyI)(4-chIorophenyl)f|uoro((S)
hydroxy(tetrahydro-2H-pyranyl)propy|)methoxyisoindoIinone (1 9, prepared as in Example
116, Step 2 (omitting chiral separation), methyl trimethylsilyl dimethylketene acetal (820 mg, 4.71
mmol), zinc fluoride (486.6 mg, 4.71 mmol) and di-u-bromobis(tri-t-butylphosphonino) adium (I)
{[P(t-Bu)3]PdBr} (STREM) (243 mg, 0.314 mmol) were placed in a round bottomed flask under
nitrogen then degassed DMF (20 mL) was added. The on was degassed with nitrogen for a
further 5 minutes then heated (using a pre-heated stirrer block set at 70 0C) for 18 h. The reaction was
allowed to cool and degassed for 10 min. The reaction was charged with further methyl trimethylsilyl
dimethylketene acetal (858 mg, 4.92 mmol), zinc fluoride (500 mg, 4.84 mmol) and {[P(t-Bu)3]PdBr}
(250 mg, 0.32 mmol) then heated (using a pre-heated stirrer block set at 70 0C) for 4 h. The reaction
was allowed to cool and then the DMF was removed under reduced pressure. Water (20 mL) and
EtOAc (50 mL) was added and the resulting black suspension was filtered. The layers of the filtrate
were separated and the organics retained. The aqueous portion was extracted with EtOAc (20 mL)
then the combined organic extracts were dried ) and concentrated under reduced pressure to
afford a black residue (1.5 g). The residue was dissolved in THF (15 mL) at RT with stirring and 1M
TBAF solution in THF (7.75 ml) was added. The reaction was stirred for 15 min at RT. The reaction
was diluted with EtOAc (60 mL) and washed with water (20 mL). The organic portion was dried
(MgSO4) and concentrated under reduced pressure to afford a black residue (1.4 g). The crude
material was purified by column chromatography using an isochratic gradient of diethyl ether 75% in
isohexane, to afford a foam (540 mg). The two diastereoisomers were separated by chiral SFC to
yield the title compound as the fast running isomer (300 mg). MS [M - C3 Methoxy]+ = 626.1
Step 2: 2-(5-chloro{[(1R)(4-chlorophenyI)fluoro[(1S)hydroxy(oxanyl)propyl]-
1-methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)methylpropanoic acid
Methyl 2-(5-chloro(((R)(4-chlorophenyl)fluoro((S)hydroxy(tetrahydro-2H-pyran
yl)propyl)methoxyoxoisoindolinyl)methyl)phenyl)methylpropanoate (300 mg) was dissolved
in THF (22 mL) then a solution on lithium hydroxide drate (109.1 mg, 4.5 mmol) in water (7.5
mL) and ol (4.5 mL) was added at RT with stirring. The reaction was heated at 65 0C for four
days then at 75 °C for one day. The on was allowed to cool then the volatiles were d
under reduced pressure. The resulting emulsion was diluted with ether (50 mL) and water (20 mL) and
the pH adjusted (by the addition of 2M s HCI) to facilitate extraction of the product into the
c layer. The organic portion was passed h a phase separation cartridge and the volatiles
were removed under reduced pressure to afford a crude foam (200 mg). The crude material was
purified by column chromatography, eluting with a gradient of 0-100% diethyl ether in isohexane
followed by washing with 100% EtOAc with 0.1% formic acid additive to afford a crude e (70
mg). Further purifcation by Prep HPLC afforded the title compound (46 mgs) as a colourless foam.
The product was then isolated as the alt by dissolving in MeOH, treating with TRIS and
evaporating. 1H NMR (400 MHz, CDCI3) 7.74 (1H, s), 7.31 (1H, s), 7.31 - 7.28 (1H, m), 7.24 (3H, s),
7.17 (2H, d), 7.08 (1H, d), 7.01 (1H, dd), 4.50 - 4.37 (2H, m), 4.04 (1H, dd), 3.92 (1H, dd), 3.43 (6H, s),
3.39 - 3.28 (2H, m), 2.96 (3H, s), 2.73 (1H, dd), 2.01 - 1.89 (3H, m), 1.73 (1H, s), 1.52 - 1.50 (6H, m),
1.14 - 1.07 (1H, m), 0.72 (3H, dd) (7 exchangeable protons not observed). MS [M+H]+= 644.
Exam ple 122: 2-(5-chloro{[(1R)(4-chloropheny|)fluoro[(1S)hydroxy(oxan
pyl]methoxyoxo-2,3-dihydro-1 ndolyl]methyl}phenyl)acetic acid
(tris(hydroxymethyl)aminomethane salt)
Step 1: Methyl 2-(5-chloro(((R)(4-chlorophenyl)fluoro((S)hydroxy(tetrahydro-2H-
pyranyl)propyl)methoxyoxoisoindolinyl)methyl)phenyl)acetate
1-(tert-Butyldimethylsilyloxy)methoxyethene (1270.3 mg, 6.75 mmol), zinc fluoride (697.5 mg, 6.75
mmol), tri-t—butylphosphine (136 mg, 0.67 mmol) and benzylideneacetone) palladium(0) (193.9
mg, 0.337 mmol) were divided evenly between two reaction tubes. A degassed solution of a
diastereomeric mixture at C-3 of 2-(2-bromo-4—chlorobenzyl)(4-chlorophenyl)—4-fluoro((S)
hydroxy(tetrahydro-2H-pyranyl)propyl)methoxyisoindolinone (860 mg, 1.35 mmol, prepared
as in Example 116 Step 2 omitting chiral separation) in DMF (20 mL) was divided evenly between the
two reaction tubes. The mixture in each tube was degassed with nitrogen for a further 30 s prior to
being sealed then stirred with heating (using a ated stirrer block set at 70 °C) for 18 h. The
reaction was allowed to cool to RT and DMF was removed under reduced re. Water (20 mL)
and EtOAc (50 mL) was added and the resulting black suspension ed. The layers of the te
were separated and the organics retained. The aqueous portion was extracted with EtOAc (20 mL)
then the combined organic extracts were dried ) and concentrated under reduced pressure to
afford a black residue (1.5 g). The crude material was purified by column chromatography with a
gradient of 50-90% l ether in isohexane to afford a colourless foam (510 mg). The two
diastereoisomers were separated by chiral SFC to yield the title compound as the fast running isomer
(140 mg, 33% yield). MS [M-C3 methoxy]+= 598.1
Step 2: 2-(5-chloro{[(1R)(4-chlorophenyl)—7-fluoro[(1S)hydroxy(oxanyl)propyI]
methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)acetic acid
(tris(hydroxymethyl)aminomethane salt)
Step 2 was performed using conditions smilarto those described in Preparation 52 Step 3 to give the
title compound. 1H NMR (400 MHz, CDCI3) 7.78 (1H, s), 7.25 - 7.18 (4H, m), 7.14 (1H, d), 7.06 - 6.97
(3H, m), 4.70 (1H, d), 4.21 (1H, d), 4.00 (1H, d), 3.86 (1H, d), 3.70 (1H, d), 3.59 (6H, s), 3.54 (1H, d),
3.38 - 3.22 (3H, m), 2.74 (3H, s), 1.89 - 1.78 (2H, m), 1.69 (1H, d), 1.48-1.34 (2H, m), 1.05 (1H, d),
0.64 (3H, dd), seven exchangeable protons not observed. MS [M+H]+= 616
Example 123: 2-(5-chloro{[(1R)(4-chIorophenyl)fluoro[(1R)hydroxy(oxan
yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)acetic acid
The title compound was ed in a similarfashion to Example 122, but starting from (+)-(R)—2-(4-
chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyranyl)propyl)benzoic acid (prepared in a
similarfashion to Preparation 52). 1H NMR (400 MHz, CDCI3) 7.67 (s, 2H), 7.30-7.10 (m, 7H), 4.71 (d,
1H), 4.15 (d, 1H), 4.03 (dd, 1H), 3.88 (dd, 1H), 3.80 (d, 1H), 3.72 (d, 1H), 3.37 (t, 1H), 3.25 (t, 1H),
2.72 (s, 3H), 1.95-1.82 (m), 1.71 (d, 1H), 1.48-1.32 (m, 2H), 1.10-0.98 (m, 1H), 0.91-0.80 (m, 1H), 0.65
(t, 3H). MS [M - OMe']+ = 584
Exam ple 124: (2S,3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy-
1-(oxany|)propyl]methoxyoxo-2,3-dihydro-1H-isoindo|y|]methylpropanoic acid
Step 1: Propenyl (28,3S)(4-chlorophenyl)[1-(4-chlorophenyl)fluorohydroxy
[(1S)hydroxy(oxany|)propyl]oxo-2,3-dihydro-1H-isoindoly|]methy|propanoate
To a solution of (S)—2-(4-chlorobenzoyl)fluoro(1-hydroxy(tetrahydro-2H-pyran
y|)propy|)benzoic acid (Preparation 52) (0.686 g, 1.6 mmol), propenyl )amino(4-
chlorophenyl)methylpropanoate (Preparation 62) (0.54 g, 2.12 mmol) and diisopropylethylamine
(0.83 mL, 4.8 mmol) in DMF (15 mL) was added HATU (0.91 g, 2.4 mmol) and the reaction mixture
was stirred for 2 hrs. Water was added and extracted with ethyl acetate. The organic phase was
washed with saturated , brine, dried and the solvent ated. The crude product was
purified by chromatography to afford the title compound (0.75 g, 72%). MS: [M-H]' =654.
Step 2: Propenyl (ZS,3S)(4-chIorophenyl)[(1R)(4-chlorophenyl)—7-fluoro[(1S)
y(oxany|)propyl]methoxyoxo-2,3-dihydro-1H-isoindol-Z-yl]
methylpropanoate
The title nd was prepared from ethyl )(4-chlorophenyl)[1-(4-chlorophenyl)fluoro-
1-hydroxy[(1S)hydroxy(oxanyl)propyl]oxo-2,3-dihydro-1H-isoindolyl]—2-
methylpropanoate and methanol in a similar manner as described in Preparation 10, but using MeOH
instead of 1,1-bis(hydroxymethyl)cyclopropane. The diastereoisomers were separated by chiral SFC,
the title compound was the faster eluting isomer. MS: [M + H]+ = 670.
Step 3: (2S,3S)(4-Chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy
(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindoly|]methy|propanoic acid
The title compound was prepared from propenyl (28,3S)(4-chlorophenyl)[(1R)(4-
chlorophenyl)fluoro[(1S)hydroxy(oxan-4—yl)propyl]methoxyoxo-2,3-dihydro-1H-
isoindolyl]methylpropanoate in an analogous fashion as described in Example 90, step 4. 1H
NMR (400 MHz, DMSO-d6): 12.56-12.00 (1H, m), 7.71 (1H, s), 7.42 (1H, d), 7.02 (4H, d), 6.88 (3H, d),
4.91 (1H, s), 4.23 (1H, d), 3.99-3.85 (2H, m), 3.75 (1H, dd), 3.25-3.10 (5H, m), 2.02-1.90 (1H, m),
.78 (2H, m), 1.67 (1H, d), 1.43-1.17 (6H, m), 0.95 (1H, d), 0.58 (3H, t). MS:[M + H]+ = 630.
Example 124a: (23,3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy-
1-(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindoIy|]methy|propanoic acid
hydroxymethyl)aminomethane salt)
Example 124 was dissolved in EtOH and 1 mol. eq. oftris(hydroxymethyl)aminomethane was added.
The solvent was removed in vacuo to give a colourless solid. 1H NMR (500 MHz, DMSO-d6) 6 7.69 (s,
1H), 7.39 (d, J = 10.7 Hz, 1H), 7.01 (broad s, 4H), 6.96 — 6.88 (m, 4H), 4.92 (broad s, 1H), 4.34 — 4.22
(m, 1H), 3.88 (dd, J = 10.9, 4.2 Hz, 1H), 3.74 (dd, J =11.1,4.2 Hz, 1H), 3.71 — 3.61 (m, 1H), 3.29 (s,
6H), 3.33 — 3.22 (m, 1H), 3.21 — 3.14 (m, 1H), 3.13 (s, 3H), 1.94 (tt, J = 12.2, 3.6 Hz, 1H), 1.89 — 1.78
(m,2H), 1.66 (d, J = 12.8 Hz, 1H), 1.41 — 1.24 (m, 2H), 1.19 (d, J: 6.8 Hz, 3H), 0.93 (d, J = 13.2 Hz,
1H), 0.57 (t, J = 7.3 Hz, 3H). MS:[M + H]+ = 630.
Example 125 and 126: (3S)(4-chlorophenyI)[(1R)(4-chlorophenyl)fluoro[(3-
fluorooxetanyl)methoxy](2-hydroxybutanyl)—3-oxo-2,3-dihydro-1H-isoindol-Z-
yl]propanoic acid (*both isomers separated and isolated)
Cl O
F HO O ErgfiéF F
F 00 :E
N COZEt
_. —> COzEt
Br N
0 0
Step 3 l
KEF O
F Step 4
~‘ COZH <— 00
OCH2
Step 1: Ethyl (S)((R)bromo(4-chlorophenyl)—7-fluoro((3-fluorooxetanyl)methoxy)
oxoisoindolinyl)(4-chlorophenyl)propanoate
The title compound was prepared from (38)-ethy| 3-(5-bromo(4—ch|oropheny|)hydroxy
oxoisoindolinyI)(4-ch|oropheny|)propanoate ration 36, 7.19 g, 12.7 mmol) and (3-
fluorooxetanyl)methanol (4.00 g, 38 mmol) in a similar manner as bed in Preparation 10.
The diastereoisomers were separated by column chromatography eluting with DCM. The title
nd was obtained as a colourless solid (1.93 g). MS: [M - (3-fluorooxetanyl)methanol]+ =
550.
Step 2: Step 2 was performed by using procedures similarto those bed in Example 21 Step3,
to give ethyl (S)((R)acety|—1-(4-ch|orophenyI)fluoro((3-f|uorooxetanyl)methoxy)
oxoisoindolinyI)(4-chIoropheny|)propanoate (1.29 g) as a yellow solid. MS: [M - (3-fluorooxetan-
3-y|)methano|]+ = 512.
Step 3: (S)((R)Acetyl(4-chlorophenyl)—7-fluoro((3-fluorooxetanyl)methoxy)
oxoisoindolinyI)(4-chlorophenyl)propanoic acid
The title compound was prepared from ethyl (S)—3-((R)acety|—1-(4-ch|oropheny|)f|uoro((3-
fluorooxetanyl)methoxy)—3-oxoisoindo|iny|)(4-chIorophenyl)propanoate (1.19 g, 1.92 mmol)
using procedures similar to those described in Preparation 52, Step 3The crude product was
obtained as a sticky orange solid (1.26 9). MS: [M+H]+ = 590.
Step 4: (3S)—3-(4-chlorophenyl)[(1R)(4-chIorophenyl)f|uoro[(3-fluorooxetan
yl)methoxy](2-hydroxybutanyl)oxo-2,3-dihydro-1H-isoindolyl]propanoic acid
The title compound was prepared by using procedures similar to those described in Example 41, Step
3; except using ethylmagnesium chloride instead of (tetrahydro-2H-pyran-4—yl)magnesium chloride.
26’]
Example 125: *fast running : 1H NMR (400 MHz, CDCI3) 7.73 (1H, d), 7.38 (1H, dd), 7.04 - 6.94
(8H, m), 4.93 - 4.72 (3H, m), 4.68 - 4.59 (2H, m), 3.73 - 3.62 (2H, m), 3.52 - 3.36 (2H, m), 1.89 - 1.80
(2H, m), 1.59 (3H, s), 0.80 (3H, dd); OH and COOH missing. MS: [M+H]+ = 620.
Example 126: * Slow running isomer: 1H NMR (400 MHz, CDCI3) 7.74 (1H, s), 7.37 (1H, d), 7.02 -
6.95 (8H, m), 4.93 - 4.59 (5H, m), 3.72 - 3.63 (2H, m), 3.52 - 3.37 (2H, m), 1.89 - 1.81 (2H, m), 1.58
(3H, s), 0.82 (3H, dd); OH and COOH missing. MS: [M+H]+ = 620.
Examples 127 and 128: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-
hydroxy(pyridinyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
(*both isomers separated and isolated)
CI CI
Step 1: Ethyl (3S)(4-chlorophenyl)[1-(4-chlorophenyl)fluorohydroxyoxo
(pyridinecarbonyl)-2,3-dihydro-1H-isoindolyl]propanoate
To a stirred solution of 2-(4-chIorobenzoyI)fluoro(pyridinecarbonyl)benzoic acid (preparation
61) (0.48 g, 1.25 mmol), (S)-ethy| 3-amino(4-chlorophenyl)propanoate hydrochloride (0.47 g, 1.87
mmol) and diisopropyl ethylamine (0.9 mL, 5.0 mmol) in DMF (15 mL) was added N-propylphosphonic
acid ide, cyclic trimer (50% w/w, 1.211 mL,1.87 mmol) and the reaction mixture was stirred for1
h. Water was added and the t was extracted with ethyl acetate. The crude product was d
by chromatography, eluted with petrol ether — ethyl acetate 0-50% to afford the title compound (0.36 g,
39%). MS: [M + H]+ = 384.
Step 2: Ethyl (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoromethoxyoxo
(pyridinecarbonyl)-2,3-dihydro-1H-isoindolyl]propanoate
The title compound was prepared from ethyl (3S)(4-chloropheny|)[1-(4-ch|orophenyl)f|uoro
hydroxyoxo(pyridine-2—carbonyI)-2,3-dihydro-1H-isoindoIyl]propanoate
and methanol in a similar manner as described in Preparation 10. The diastereoisomers were
separated by column chromatography [M + H]+ = 607.
Step 3: (3S)(4-ChlorophenyI)[(1R)(4-chlorophenyl)fluoromethoxyoxo
(pyridinecarbonyl)-2,3-dihydro-1H-isoindolyl]propanoic acid
The title compound was prepared from ethyl (38)(4-chlorophenyI)[(1R)(4-ch|orophenyl)
fluoromethoxyoxo(pyridinecarbonyl)-2,3-dihydro-1H-isoindolyl]propanoate
in an analogous fashion as described in Preparation 40. [M + H]+ = 579.
Step 4: (38)(4-chlorophenyl)[(1R)(4-ch|oropheny|)fluoro[1-hydroxy(pyridin
pyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid
To a solution of (38)(4-chlorophenyl)[(1R)(4-chlorophenyI)fluoromethoxyoxo
(pyridinecarbonyl)-2,3-dihydro-1H-isoindolyl]propanoic acid (0.5 g, 0.86 mmol) in THF 10 mL)
was added ZnClz (1.7 mL, 0.5 M in THF, 0.85 mmol) and the mixture was stirred for 30 mins, cooled to
-30°C and EtMgCl (1.29 mL, 2 M in THF, 2.58 mmol) was added and stirred for 30 mins. Saturated
NH4C| solution was added, acidified to pH=4.5 and extracted with ethyl acetate. The diastereoisomers
were ted by chiral SFC.
Example 127: fast running isomer
1H NMR (400 MHz, DMSO-ds): 12.38 (1H, s), .55 (1H, m), 7.85 (1H, cl), 7.82-7.70 (2H, m), 7.83-
7.58 (1H, m), .23 (1H, m), 7.17-7.04 (4H, m), 7.04-8.88 (4H, m), 8.08 (1H, s), 4.80 (1H, dd),
3.48 (1H, dd), 3.18 (1H, dd), 3.08-3.00 (3H, m), 2.44-2.34 (1H, m), 2.34-2.24 (1H, m), 0.71 (3H, t). MS:
[M + H]+ = 809.
Example 128: slow running isomer
1H NMR (400 MHz, DMSO-ds): 12.29 (1H, d), 8.59-8.53 (1H, m), 7.88 (1H, d), 7.83-7.70 (2H, m), 7.59
(1H, dd), 7.28-7.22 (1H, m), 7.17-7.05 (4H, m), 7.05-8.92 (4H, m), 8.07 (1H, s), 4.81 (1H, dd), 3.49-
3.41 (1H, m), 3.22-3.09 (1H, m), 3.03 (3H, s), 2.41 (1H, dd), 2.34-2.28 (1H, m), 0.71 (3H, t).
MS: [M + Hr = 809.
Example 129: (3R)[(4-chloromethanesulfonylphenyl)methyI](4-chlorophenyI)fluoro
[1-(4-f|uoropiperidinyl)hydroxypropyl]methoxy-2,3-dihydro-1H-isoindolone (Example
isolated as a single isomer at the position shown*)
Starting from (-)(1-(1-(tert-butoxycarbonyl)fluoropiperidinyl)hydroxypropyl)(4-
chlorobenzoyl)fluorobenzoic acid (preparation 57) and (4-chloro
(methylsu|f0ny|)phenyl)methanamine (Example 35, step 3), the title compound was prepared using
procedures r to those described in Example 87 (Steps 1-2). 1H NMR (400 MHz, CDCI3) 7.93
2016/053042
(1H, d), 7.78 (1H, s), 7.49 (1H, d), 7.45 - 7.38 (2H, m), 7.32 (2H, d), 7.24 (2H, d), 4.96 (2H, q), 3.02
(3H, s), 2.96 (3H, s), 2.95 - 2.83 (4H, m), 2.26 - 2.16 (2H, m), 2.09 - 1.95 (2H, m), 1.86 - 1.62 (3H, m),
1.44 - 1.25 (1 H, m), 0.71 (3H, dd); MS [M+H]+ = 653.
Exam ple 130: 4-{[(1R)(4-ch|oropheny|)f|uoro[(1S)hydroxy(1-methy|piperidin
yI)propyI]oxo[cishydroxycyc|obutoxy]-2,3-dihydro-1H-isoindolyl]methy|}benzonitrile
Starting from (S)—5-(1-(1-(tert-butoxycarbonyl)piperidinyl)hydroxypropyl)(4-ch|orobenzoyl)
fluorobenzoic acid (Preparation 60), 4-aminomethylbenzonitrile and cis((tert-
butyldimethylsily|)oxy)cyc|obutano|, the title compound was prepared using procedures similar to
those described in Example 87 (Steps 1-3). Purification by chiral SFC gave the title compound (slow
running isomer): 1H NMR (400 MHz, CDCI3) 7.62 (1H, d), 7.50 (2H, d), 7.34 (2H, d), 7.30 (1H, dd),
7.28 - 7.24 (4H, m), 4.62 (1H, d), 4.12 (1H, d), 3.61 - 3.53 (1H, m), 3.08 - 3.00 (1H, m), 2.91 (1H, d),
2.80 (1H, d), 2.22 (3H, s), 1.94 - 7.75 (9H, m), 1.73 - 1.66 (3H, m), 1.43 - 1.20 (3H, m), 0.67 (3H, dd).
MS: [M+H]+: 618.
Exam ple 131: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-f|uoro
methylpiperidiny|)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindol
yl]propanoic acid
(*single isomer ted and isolated)
Step 1
Step 2
Step 3
Step 4
2016/053042
Step 1 and 2: Starting from (-)(1-(1-(tert-butoxycarbonyl)f|uoropiperidiny|)hydroxypropy|)
(4-chlorobenzoyI)f|uorobenzoic acid (Preparation 57), Steps 1 and 2 were performed using
s similarto those described in Example 105. MS: [M + H]+= 661.1.
Step 3: ethyl -(4-chlorophenyl)(1-(4-chlorophenyI)fluoro(1-(4-fluoro
methylpiperidinyl)hydroxypropyI)methoxyoxoisoindolinyl)propanoate
Step 3 was performed using procedures similar to those described in Example 81, Step 5. The
desired diastereoisomer was isolated using chiral SFC as the fastest eluting isomer. MS: [M + H]+ =
675.1.
Step 4: (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluoro
methylpiperidinyl)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindol
yl]propanoic acid
Step 4 was performed using methods similar those described in Preparation 52, Step 3 to give the
title compound which precipitated and was collected by filtration during the work up. 1H NMR (400
MHz, DMSO) 7.70 (1H, s), 7.42 (1H, d), 7.16 (2H, d), 7.09 (3H, d), 7.05 (1H, s), 6.98 (2H, d), 5.53 (1H,
s), 4.63 (1H, dd), 3.48 (1H, dd), 3.18 (1H, dd), 3.08 (3H, s), 2.70 - 2.67 (1H, m), 2.13 (3H, s), 2.05 -
1.99 (1H, m), 1.95 - 1.80 (5H, m), 1.78 - 1.69 (1H, m), 1.11 - 1.05 (1H, m), 0.55 (3H, t), OH and COOH
not observed. MS: [M+H]+ = 647.3.
Example 132: utyl 2-{4-[(1S)-1 -[(1R)-1 -(4-chlorophenyl)[(4-chlorophenyl)methyl]
fluoromethoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetate
Example 133: tert-butyl 2-{4-[(1R)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]
fluoromethoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetate
(*both isomers ted and isolated)
BocN O
O 0
Me 0
step 3 Mea/ \n/\N
Steps 1-2. Starting from 5-(1-(tert-butoxycarbony|)piperidinecarbony|)(4-chIorobenzoyl)
fluorobenzoic acid (Examples 80 and 81, step 2), steps 1-2 were performed using methods similar to
those described in Example 73, Steps 3 and 4 respectively, except T3P was used d of HATU in
Steps 1 and MeOH instead of 1-(hydroxymethy|)cyc|opropanecarboxamide in Step 2 to give 3-(4-
chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy(piperidinecarbonyl)-2,3-dihydro-1H-
isoindolone. MS [M+H]+ = 527
Step 3: tert-butyl 1-(4-chIorophenyl)[(4-chlorophenyl)methyI]fluoromethoxyoxo-
2,3-dihydro-1H-isoindolecarbonyl]piperidiny|}acetate
To the reaction flask ning 3-(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy
(piperidinecarbonyl)-2,3-dihydro-1H-isoindolone (1.729 g, 3.28 mmol), K2C03 (1.80 g, 13.0
mmol) and DMF (30 mL) was added terf-butyl bromoacetate (0.53 mL, 3.57 mmol). The reaction was
stirred at room temperature for 90 minutes at which time LCMS analysis showed complete
consumption of starting material. Solvent was then d and the residue was taken up in EtOAc
(10 mL) and sat. NaHC03. The aqueous layer was r extracted with EtOAc (3 x 50 mL). The
combined organics were dried over anhydrous NaZSO4, filtered, and trated to an oily residue.
This material was then purified via SiOz (petroleum ether:EtOAc) to yield 1.505 g (72%) ofthe racemic
product. The racemic material was then subjected to chiral preparatory separation to yield the
enantiomers as white solids.
Fast enantiomer: 1H NMR (400 MHz, 00013): 8.22 (1 H, d), 7.75 (1H, dd), 7.28 (5H, d), 7.26-7.18 (4H,
m), 4.88 (1H, d), 4.07 (1H, Cl), 331-320 (1H, m), 3.19 (2H, s), 3.04 (2H, d), 2.77 (3H, s), 2.50-2.34
(2H, m), 1.99-1.84 (4H, m), 1.49 (9H, s). MS [M+NH4]+= 841.
Slow enantiomer: 1H NMR (400 MHz, CDCI3): 8.22 (1H, s), 7.75 (1H, d), 7.28 (5H, s), 7.28-7.17 (4H,
m), 4.88 (1H, d), 4.07 (1H, d), 3.31-3.21 (1H, m), 3.19 (2H, s), 3.04 (2H, d), 2.77 (3H, s), 4
(2H, m), 2.00-1.84 (4H, m), 1.50 (9H, s). MS [M+NH4]+ = 841.
Step 4: Taking the slow-eluting isomer from Step 3, Step 4 was performed using a method similar to
that bed in Preparation 52, Step 1. Purification by preparative chiral HPLC gave the title
compounds.
Example 132— fast diastereomer: 1H NMR (400 MHz, CDCIg): 7.64 (1 H, s), 7.32-7.15 (9H, m), 4.61
(1H, d), 4.05 (1H, d), 3.05 (2H, s), 3.01 (1H, d), 2.89 (1H, d), 2.72 (3H, s), 2.16-1.96 (2H, m), 1.91 (2H,
q), 1.82 (1H, d), 1.74 (1H, s), 1.71-1.54 (2H, m), 1.44 (10H, 5), 1.41-1.31 (1H, m), 0.68 (3H, t). MS
[M+H]+ = 671.
Example 133— slow diastereomer: 1H NMR (400 MHz, c0013): 7.88 (1 H, s), 7.33-7.17 (9H, m), 4.84
(1H, d), 4.08 (1H, d), 3.07 (2H, s), 3.04 (1H, d), 2.90 (1H, d), 2.75 (3H, s), 2.19-2.02 (2H, m), 2.02-1.89
(2H, m), .77 (2H, m), 1.77-1.59 (2H, m), 1.59-1.45 (9H, m), 1.45-1.38 (2H, m), 0.70 (3H, t). MS
[M+H]+ = 871.
Example 134: 2-{4-[(1S)[(1R)(4-chloropheny|)[(4-chloropheny|)methyl]fluoro
methoxyoxo-2,3-dihydro-1H-isoindoI-S-yl]hydroxypropyl]piperidiny|}acetic acid
Exam ple 135: 2-{4-[(1R)[(1R)(4-chloropheny|)[(4-chloropheny|)methyl]fluoro
methoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetic acid
(*prepared and isolated as a single isomers)
OH 0 ’OH 0
Examples 134: and 135 were prepared from Examples 132 and 133 respectively by using
procedures similarto those bed in Example 121, Step 2.
Example 134: 1H NMR (400 MHz, DMSO-da): 7.68 (1H, s), 7.43-7.32 (3H, m), 7.32-7.21 (4H, m), 7.17
(2H, d), 4.93 (1H, s), 4.45 (1H, d), 4.14 (1H, cl), 3.12 (1H, d), 3.00 (1H, d), 2.92 (2H, s), 2.73 (3H, s),
2.33-2.14 (3H, m), 1.86 (2H, q), .67 (2H, m), .23 (2H, m), 1.16 (1H, d), 0.58 (3H, t). MS
[M+H]+ = 615.
Example 135 1H NMR (400 MHz, DMSO-d6): 7.69 (1H, s), 7.39 (1H, d), 7.34 (2H, CI), 7.31-7.21 (4H,
m), 7.18 (2H, d), 5.15-4.87 (1H, m), 4.46 (1H, d), 4.13 (1H, d), 3.19 (1H, d), 3.03 (3H, s), 2.73 (3H, s),
2.45-2.26 (3H, m), 1.95-1.69 (4H, m), 1.47-1.31 (2H, m), 1.16 (1H, d), 0.59 (3H, t). MS [M-H+]' = 613.
Example 136: Methyl 3-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro
methoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}propanoate
BocN O —>
OH .
_ step 2 step 3
: Et OH 0
Et OH 0
CI b
Starting from (S)(1-(1-(tert-butoxycarbony|)piperidinyl)hydroxypropy|)(4-ch|orobenzoyl)
fluorobenzoic acid (Preparation 60) and 4-chlorobenzylamine, the title compound was prepared using
methods similar to those bed for Example 87 (Steps 1-3), except that methyl methacrylate/DBU
were used instead of NaBH3CN in step 3. 1H NMR (400 MHz, CDCI3): 7.63 (1H, d), 7.31-7.15 (9H, m),
4.61 (1H, d), 4.05 (1H, d), 3.66 (3H, s), 2.96 (1H, d), 2.84 (1H, d), 2.72 (3H, s), 2.69-2.56 (2H, m), 2.47
(2H, t), 2.07-1.79 (5H, m), 1.79-1.55 (3H, m), 1.40-1.28 (2H, m), 0.67 (3H, t). MS [M+H]+= 643.
Example 137: (1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro
methoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}propanoic acid
WO 55860
Starting from Example 136, the title compound was prepared using a procedure similar to that
described in Preparation 52 (Step 3). 1H NMR (400 MHz, DMSO): 7.69 (1H, s), 7.40 (1H, d), 7.35
(2H, d), 7.31-7.21 (3H, m), 7.18 (2H, d), 4.89 (1H, s), 4.46 (1H, d), 4.13 (1H, d), 3.01 (1H, d), 2.87 (1H,
d), 2.74 (3H, s), 2.58-2.52 (3H, m), 2.36-2.25 (2H, m), 2.06-1.73 (5H, m), 1.71 (1H, d), 1.35-1.18 (2H,
m), 1.09 (1H, d), 0.57 (3H, t). MS = 627.
BIOLOGICAL ASSAYS
MDM2-p53 interaction using a l plate binding assay (ELISA)
The ELISA assay was performed in streptavidin coated plates which were preincubated with 200 pl
per well of 1ug ml‘1 biotinylated |P3 peptide. The plates were ready to use for MDM2 binding after
washing the plate with PBS.
Compounds and control ons in DMSO aliquoted in 96-well plates were pre-incubated in a final
2.5-5 % (v/v) DMSO concentration at room temperature (for example 20 °C) for 20 min with 190 pl
aliquots of optimized concentrations of in vitro translated MDM2, before transfer of the MDM2-
compound mixture to the b-IP3 streptavidin plates, and incubation at 4 °C for 90 min. After washing
three times with PBS to remove unbound MDM2, each well was incubated at 20 °C for 1 hour with a
TBS-Tween (50mM Tris pH7.5; 150mM NaCl; 0.05% Tween 20 nonionic detergent) buffered solution
of y mouse monoclonal anti-MDM2 antibody (Ab-5, Calbiochem, used at a 1/10000 or 1/200
on ing on the antibody stock solution used), then washed three times with een
before incubation for 45 mins at 20 °C with a TBS-Tween buffered solution of a goat-anti-mouse
horseradish dase (HRP) conjugated secondary antibody (used at 1/20000 or 1/2000 depending
on the antibody stock solution). The unbound secondary antibody was removed by washing three
times with TBS-Tween. The bound HRP activity was measured by enhanced chemiluminesence
(ECLT'VI Amersham Biosciences) using the oxidation of the diacylhydrazide substrate, luminol, to
te a quantifiable light signal. The percentage of MDM2 inhibition at a given concentration is
calculated as the [1 - (RLU detected in the compound treated sample — RLU negative DMSO control)
+ (RLU of DMSO ve and negative controls)] x 100 or as the (RLU detected in the compound
treated sample + RLU of DMSO controls) x 100. The |C50 was calculated using a plot of % MDM2
inhibition vs concentration and is the average of two orthree independent experiments.
Western blot analysis
SJSA cells were treated for 6 hours with 5, 10 and 20 ulVl of compounds in 0.5% DMSO. The cells
together with 0.5% DMSO only controls were washed with ice-cold phosphate buffered saline (PBS)
and protein extracts prepared by lysing the cells in SDS buffer (62.5mM Tris pH 6.8; 2% sodium
l sulphate(SDS); 10% ol) with sonication for 2x5seconds (Soniprep 150ME) to break
down high molecular weight DNA and reduce the viscosity of the samples. The n concentration
of the samples was ted using the Pierce BCA assay system (Pierce, Rockford, IL) and 50ug
aliquots of protein analysed using standard SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and
Western immunoblotting procedures. B-mercaptoethanol (5%) and bromophenol blue (0.05%) were
added and the samples, which were then boiled for 5 minutes, ed by brief centrifugation, before
loading onto a pre-cast 4-20% nt Tris-Glycine ed SDS-polyacrylamide gel (lnvitrogen).
Molecular weight standards (SeeBlueTM, Invitrogen) were included on every gel and electrophoresis
was carried out in a Novex XL tank (Invitrogen) at 180 volts for 90 s. The ted proteins
were transferred electrophoretically overnight from the gel onto a Hybond C nitrocellulose membrane
(Amersham) using a BioRad ophoresis tank and 25mM Tris, 190mM glycine and 20% methanol
transfer buffer at 30 volts or two hours at 70 volts. Primary antibodies used for immunodetection of the
transferred proteins were: mouse monoclonal NCL-p53DO-7 (Novocastra) at 1:1000; MDM2(Ab-1,
clone IF2) (Oncogene) at 1:500; WAF1 (Ab-1, clone 4D10) (Oncogene) at 1:100; Actin (AC40) (Sigma)
at 1:1000. The secondary antibody used was peroxidase conjugated, affinity purified, goat anti-mouse
(Dako) at 121000. Protein detection and visualisation was performed by enhanced chemiluminescence
(ECLTM, am) with light detection by exposure to blue-sensitive autoradiography film (Super RX,
Fuji).
Protocol A: SJSA-7 and SN40R2 assays
The MDM2 ied cell lines tested were an isogenic matched pair of p53 wild-type and mutated
osteosarcoma (SJSA-1 and , respectively). All cell cultures were grown in RPMI 1640
medium , Paisley, UK) supplemented with 10% fetal calf serum and routinely tested and
confirmed negative for mycoplasma infection. The growth of cells and its inhibition was measured
using the sulphorhodamine B (SRB) method as previously outlined. 100 pl of 3x104/ml and 2x104/ml
SJSA-1 and SN40R2 cells, respectively, were seeded into 96-well tissue culture plates and incubated
at 37 °C in a 5% COZ humidified incubator for 24hrs, after which the medium was replaced with 100 pl
of test medium containing a range of MDM2-p53 antagonist concentrations and incubated for a further
72 hrs to allow cell growth before adding 25 pL of 50% oroacetic acid (TCA) to fix the cells for 1 h
at 4 °C. The TCA was washed off with distilled water and 100 pL of SRB dye (0.4% w/v in 1% acetic
acid) (Sigma-Aldrich, Poole, Dorset) added to each well of the plate. Following incubation with the
SRB dye at room temperature for 30 min, the plates were washed with 1% acetic acid and left to dry.
The SRB stained protein, which is a e of the number of cells in a well, was then resuspended
in 100 pL of10 mM Tris-HCI (pH 10.5) and the absorbance at A = 570 nm ed in each well using
a FluoStar Omega Plate reader. The Glso was calculated by non-linear regression analysis of the data
using Prism v4.0 statistical software.
Protocol B: SJSA-1 and SN40R2 assays
The CellTiter—Glo® Luminescent Cell Viability Assay is a homogeneous method to determine the
number of viable cells in culture based on tation ofthe ATP present, which signals the presence
of metabolically active cells. Both SJSA-1 and SN40R2 were grown in RPMI 1640 (Life Technologies
#61870) supplemented with 10% FBS (PAA #A15-204) and 10 U/ml llin/streptomycin. 2000 cells
in 75 pl were seeded in each well of a 96 well plate and left at 37 °C in a 5% C02 humidified incubator
for 24hrs. A range of MDMZ-p53 antagonist concentrations in DMSO was then added to the cells to a
final DMSO concentration of 0.3%, and incubated for a r 72 hrs to allow cell growth. 100 pl of
CTG reagent (Promega #G7573) was added to all wells and luminescence was measured on the
WO 55860
topcount. The EC50 values were ined from a sigmoidal 4 parameter curve fit using XLfit in
conjunction with Activity Base (IDBS; Guildford, Surrey, UK).
Anti-proliferative Activity
Inhibition of cell growth is ed using the Alamar Blue assay (Nociari, M. M, Shalev, A., Benias,
P., Russo, C. Journal of Immunological Methods 1998, 213, 157-167). The method is based on the
ability of viable cells to reduce resazurin to its fluorescent product resorufin. For each proliferation
assay cells are plated onto 96 well plates and allowed to rfor 16 hours prior to the addition of
inhibitor compounds (in 0.1% DMSO v/v) for a further 72 hours. At the end of the incubation period
% (v/v) Alamar Blue is added and incubated for a further 6 hours prior to determination of
fluorescent product at 535nM ex / 590nM em. The anti-proliferative activities of compounds of the
invention can be determined by measuring the ability ofthe compounds to inhibit growth in cancer cell
lines for example as available from DSMZ ECACC or ATCC.
Results
Table 1 — biological data obtained from assays as described herein
Patent MDM2 |C50 SJSA-1 SJSA1 |C50 SN40R2 SN40R2 |C50
Example (pM) |C50 (pM) |C50 (pM) (pM)
(pM) (Protocol (Protocol col B)
(Protocol B) A)
1 0.012 0.49 0.55 18 10% at 10
2 0.0046 0.33 0.46 17 22% at 10
3 0.093
4 0.043
0.14
6 0.12
7 0.0066
8 0.0047 0.33 18
9 0.011
0.0037 0.14 7.5
11 0.033
12 0.0058 0.51 0.69 5.9
13 0.12 4.6 5.9
14 0.0050 0.83 0.49 10% at 30 9% at 10
0.019
16 0.14 2.1 13
17 0.063 0.95 8.1
18 0.045 0.80 18
19 0.022 0.62 2.0 13 13
0.011 0.33 11
21 0.0078 0.23 0.39 15 51% at 10
22 0.0052 0.21 18
24 0.0075 0.37 0.63 21 19% at 10
0.0072 0.71 1.1 25 14% at 10
26 0.032 1.7 17
27 0.065 2.1 29% at 30
28 0.026 0.93 26% at 30
29 0.11 1.4 17
0.086 2.4 27
31 0.038 1.2 18
32 0.87 15
2016/053042
Patent MDM2 IC50 SJSA-1 SJSA1 IC50 SN40R2 SN40R2 IC50
Example (pM) IC50 (pM) IC50 (pM) (pM)
(pM) (Protocol (Protocol (Protocol B)
(Protocol B) A)
33 0.0019 9.1 7% at 30
34 0.0046 0.093 9.9
0.0018 0.16 0.69 23 13
36 0.0019 0.078 17
37 0.041 1.2 13
38 0.026 0.67 17
39 0.068 2.0 18
40 0.063 1.5 17
41 0.0016 0.14 13
42 34%@0.00030 0.011 0.03 12 10
43 47%@0.0010 0.57 12
44 0.0058 0.83 6.8
45 0.23
46 10.78
47 0.43
48 0.0073 0.46 0.97 17 24% at 10
49 0.082 1.6 18
50 0.00080 0.079 0.032 17 22% at 10
51 0.13
52 0.15 1.8
53 0.12 1.9
54 0.15
55 1.7 11
56 0.12
57 0.061 1.4 16
58 0.018 0.59 15
59 0.0041 0.25 19
60 0.014
61 0.016 0.69 44% at 30
62 0.0023 0.055 55% at 30
63 71%@0.0010 0.096 19% at 10
64 0.0021
65 0.0018 0.26
66 0.0030
67 60%@0.0010 0.53 9.4
68 0.0070 1.8 13
69 0.00070 0.081 0.16 15 6.6
70 0.0057 0.68 4.9
71 0.0020 0.66 0.7 44 3% at 10
72 0.0015 0.14 0.17 16 45% at 10
73 0.012 3.6 39% at 30
74 0.00050 0.28 1.0 28 13
75 73%@0.0010 0.12 0.35 22 12
76 0.0095 1.0 13
77 61%@0.00030 0.46 3.7
78 0.0046 0.41 1.4 5.9 4.2
79 0.0022 8.1 10% at 30
80 73%@0.0010 0.83 13
81 0.0026
82 0.0025 0.21 51% at 30
83 0.0010 0.53 11
84 39%@0.00030 0.065 18
Patent MDM2 IC50 SJSA-1 SJSA1 IC50 SN40R2 SN40R2 IC50
Example (pM) IC50 (pM) IC50 (pM) (pM)
(pM) col col (Protocol B)
(Protocol B) A)
85 0.00049 0.049 13
86 56%@0.10
87 82%@0.0030 37% at 10 1% at 10
88 0.00079 0.15 0.23 39 11% at 10
89 0.012 3.6 3% at 10
90 39%@0.030 97% at 10 6% at 10
91 78%@0.0010 0.080 0.059 26 13% at 10
92 76%@0.0010 0.080 0.084 36 12% at 10
93 49%@0.030 3.3 12% at 10
94 64%@0.10
95 87%@0.0010 0.036 0.022 16 21% at 10
96 0.00064 0.071 0.075 19 17% at 10
97 45%@0.10
98 0.0008 0.081 0.13 33 11% at 10
99 0.012 3.2 4% at 10
100 0.0063 1.7 7% at 10
101 55%@0.00030 0.026 0.026 18 11% at 3
102 0.017 1.4 26% at 10
103 55%@0.030 0.8 18% at 10
104 70%@0.10 42% at 10 5% at 10
105 92%@0.0010 0.022 0.05 33 20% at 10
106 57%@0.030 3.2 8% at 10
107 78%@0.0010 0.021 0.038 24 18% at 10
108 0.0061 27% at 10 29% at 10
109 92%@0.0010 0.012 0.02 26 75% at 10
110 0010 0.026 0.013 17 30% at 10
111 61%@0.0010 0.024 0.037 9 51% at 10
113 57%@0.0010 0.02 10% at 10
114 81%@0.0010 0.029 0.063 20 15% at 10
115 73%@0.0010 0.22 2% at 10
116 88%@0.0010 0.08 0.14 44 12% at 10
117 45% a10.03 30% at10 19% at10
118 0010 0.36 8% at 10
119 54%@0.0010 0.06 0.2 39 7% at 10
120 76%@0.0010 0.063 0.095 40% at 50 4% at 10
121 93%@0.0010 0.015 0.015 26 18% at 10
122 88%@0.0010 0.024 20% at 10
123 42%@0.030 107% at 10 16% at 10
124 80%@0.0010 0.023 0.027 23 55% at 10
125 18%@0.10
126 0.0019 0.6 0.61 30 7% at 10
127 0.0045 1.4 14% at 10
128 39% 0.10
129 90%@0.0010 0.047 0.048 6 112% at 10
130 98%@0.0010 0.23 87% at 10
131 89%@0.0010 0.044 0.093 22 -3% at 10
132 43%@0.030 0.75 34% at 10
133 6%@0.10 37% at 10 89% at 10
134 0.0011 0.78 2% at 10
135 40%@0.10 20% at 10 7% at 10
136 0.0013 0.056 86% at 10
137 0.00057 0.15 12% at 10
WO 55860
Where more than one data point has been ed, the table above shows an average (e.g.
geometric or arithmetic mean) of these data points.
It is of course to be understood that the invention is not intended to be restricted to the s
ofthe above ments which are described by way of example only.
Combination Protocol for Cell Proliferation
The effect of a compound of formula (I) (Compound 1) in combination with an anticancer
agent (Compound II) can be ed using the following technique. Cells from human cells
lines (e.g.SJSA—1) were seeded onto 96-well tissue culture plates at a concentration of
2.5x103, 6.0 x103, or 4.0 x103 cells/well respectively. Cells were allowed to recover for 24-48
hours priorto addition of compound(s) or e control (0.35-0.5% DMSO) as follows:
Compounds were added concurrent for 72-96 hours. Following a total of 72-96 hours
compound incubation, cells were fixed with ice-cold 10% (w/v) trichloroacetic acid for 1 hour
on ice and then washed four times with dH20 using a plate washer (Labsystems Wellwash
) and air-dried. Cells were then stained with 0.4% (w/v) Sulforhodamine B (Sigma) in
1% acetic acid for 20 min at room temperature and then washed four times with 1% (v/v)
acetic acid and air-dried before the addition of 10mM Tris buffer to solubilise the dye.
metric product was quantified by reading at Abs490nm or Abs570nm on a Wallac
Victorz plate reader (1420 multilabel counter, Perkin Elmer Life Sciences). The leo for
Compound II in the ce of varying doses of Compound I was determined. Synergy was
determined when the leo shifted down in the presence of sub-effective doses of Compound I.
Additivity was determined when the response to Compound II and nd I er
resulted in an effect equivalent to the sum of the two compounds individually. Antagonistic
effects were defined as those causing the IC50 to shift upwards, i.e. those where the response
to the two compounds was less than the sum of the effect of the two compounds individually.
PHARMACEUTICAL FORMULATIONS EXAMPLES
(i) Tablet Formulation
A tablet composition containing a compound of the formula (I) is prepared by mixing an
appropriate amount of the compound (for example 50-250 mg) with an appropriate diluent,
disintegrant, compression agent and/or glidant. One possible tablet comprises 50 mg of the
nd with 197 mg of lactose (BP) as diluent, and 3 mg magnesium te as a
lubricant and compressing to form a tablet in known manner. The compressed tablet may be
optionally film coated.
(ii) e Formulation
A capsule formulation is prepared by mixing 100-250 mg of a compound of the formula (I)
with an equivalent amount of lactose and filling the resulting mixture into standard hard gelatin
es. An appropriate disintegrant and/or glidant can be included in appropriate amounts
as required.
(iii) Iniectable Formulation I
A parenteral composition for administration by injection can be prepared by dissolving a
compound of the formula (I) (e.g. in a salt form) in water containing 10% propylene glycol to
give a concentration of active compound of 1.5 % by weight. The solution is then made
isotonic, sterilised by filtration or by terminal sterilisation, filled into an ampoule or vial or pre-
filled syringe, and sealed.
iv In'ectable Formulation II
A eral composition for ion is prepared by dissolving in water a compound of the
formula (I) (e.g. in salt form) (2 mg/ml) and mannitol (50 mg/ml), sterile filtering the solution or
by terminal isation, and filling into sealable 1 ml vials or ampoules or pre-filled syringe.
v In'ectable formulation III
A formulation for i.v. delivery by injection or infusion can be prepared by dissolving the
compound of a (I) (e.g. in a salt form) in water at 20 mg/ml and then adjusted for
isotonicity. The vial is then sealed and sterilised by autoclaving or filled into an ampoule or
vial or pre-filled syringe, sterilised by filtration and sealed.
(vi) Iniectable ation IV
A formulation for i.v. delivery by injection or infusion can be prepared by dissolving the
compound of formula (I) (e.g. in a salt form) in water ning a buffer (e.g. 0.2 M acetate
pH 4.6) at 20mg/ml. The vial, ampoule or lled e is then sealed and ised by
autoclaving or sterilized by filtration and sealed.
(vii) Subcutaneous or Intramuscular Injection Formulation
A composition for sub-cutaneous or uscular stration is prepared by mixing a
compound ofthe formula (I) with pharmaceutical grade corn oil to give a concentration of 5-50
mg/ml. The composition is sterilised and filled into a suitable container.
(viii) Lyophilised formulation I
Aliquots of formulated compound of formula (I) are put into 50 ml vials and Iyophilized. During
lisation, the itions are frozen using a one-step freezing protocol at (—45 °C). The
temperature is raised to -10 °C for annealing, then lowered to freezing at —45 °C, followed by
primary drying at +25 °C for approximately 3400 minutes, followed by a secondary drying with
increased steps if temperature to 50 °C. The pressure during primary and secondary drying is
set at 80 millitor.
ix L o hilised formulation II
WO 55860 2016/053042
Aliquots of formulated compound of formula (I) or a salt thereof as defined herein are put into
50 mL vials and lyophilized. During lyophilisation, the compositions are frozen using a one-
step freezing protocol at (—45 0C). The temperature is raised to —10 °C for annealing, then
lowered to freezing at -45 °C, followed by primary drying at +25 °C for approximately 3400
s, followed by a secondary drying with sed steps if temperature to 50 °C. The
pressure during primary and secondary drying is set at 80 millitor.
x L o hilised Formulation for use in i.v. administration I“
An aqueous buffered solution is prepared by dissolving a compound of formula I in a buffer.
The buffered solution is filled, with filtration to remove particulate matter, into a container
(such as a Type 1 glass vial) which is then lly sealed (e.g. by means of a Fluorotec
r). If the compound and formulation are sufficiently stable, the ation is sterilised
by autoclaving at 121°C for a suitable period of time. If the formulation is not stable to
autoclaving, it can be sterilised using a suitable filter and filled under sterile conditions into
sterile vials. The solution is freeze dried using a suitable cycle. On completion of the freeze
drying cycle the vials are back filled with nitrogen to atmospheric pressure, stoppered and
secured (e.g. with an aluminium crimp). For intravenous administration, the freeze dried solid
can be reconstituted with a pharmaceutically acceptable diluent, such as 0.9% saline or 5%
dextrose. The solution can be closed as is, or can be diluted further into an infusion bag
(containing a pharmaceutically acceptable diluent, such as 0.9% saline or 5% se),
before administration.
(xii) Powder in a bottle
A composition for oral stration is prepared by g a bottle or vial with a compound of
the formula (I). The composition is then tituted with a suitable diluent for example
water, fruit juice, or commercially available vehicle such as OraSweet or Syrspend. The
reconstituted solution may be dispensed into dosing cups or oral syringes for administration.
Claims (52)
1. A compound of formula (I): s (R5)m (R4)a R6 N (R1)n or a er or a solvate or a pharmaceutically acceptable salt thereof, wherein: R1 is ndently selected from hydroxy, halogen, nitro, nitrile, kyl, haloC1-4alkyl, hydroxyC1-4alkyl, C2-6alkenyl, C1-4alkoxy, haloC1-4alkoxy, C2-4alkynyl, -O0,1-(CRxRy)v-CO2H, -(CRxRy)v-CO2C1-4alkyl, -(CRxRy)v-CON(C1-4alkyl)2, -P(=O)(Rx)2, -S(O)d- -S(O)d-heterocyclic group with 3 to 6 ring members and -S(O)d-N(R8)2; R2 is selected from hydrogen, C1-4 alkyl, C2-6alkenyl, hydroxyC1-4alkyl, -(CRxRy)u-CO2H, -(CRxRy)u-CO2C1-4alkyl, and -(CRxRy)u-CONRxRy; s is selected from 0 and 1; R3 is hydrogen or -(A)t-(CRxRy)q-X; t is selected from 0 and 1; q is selected from 0, 1 and 2; wherein when R3 is -(A)t-(CRxRy)q-X then (i) at least one of s, t and q is other than 0 and (ii) when t is 0 then s is 1 and q is other than 0; A is a C3-6cycloalkyl group or a heterocyclic group with 3 to 6 ring members, n the heterocyclic group comprises one or more heteroatoms selected from N, O, S and oxidised forms thereof; X is selected from hydrogen, halogen, -CN, -OR9, -(CH2)v-CO2H, -(CH2)v-CO2C1-4alkyl, - S(O)d-Rx, -C(=O)-C1-4alkyl, -N(H)e(C1-4alkyl)2-e, -NRxRy , -NHSO2Rx, -NRxCORy, and - C(=O)NRxRy; R4 and R5 are ndently selected from halogen, nitrile, C1-4 alkyl, -4alkyl, C1- 4alkoxy and haloC1-4alkoxy; R6 and R7 are independently selected from hydrogen, C1-6alkyl, haloC1-6alkyl, C2- 6alkenyl, C2-6alkynyl, hydroxy, hydroxyC1-6alkyl, -6alkyl, -(CH2)j-O-C1-6alkyl, -(CH2)j-O- (hydroxyC1-6alkyl), -C1-6alkyl-NRxRy, -(CRxRy)p-CONRxRy, -(CRxRy)p-NRxCORy, -(CRxRy)p-OCH2-CONRxRy , heterocyclic group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring members, -CH2-O-heterocyclic group with 3 to 7 ring members, -CH2-NH-heterocyclic group with 3 to 7 ring members, -CH2-N(C1-6alkyl)-heterocyclic group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring members, cloalkyl, -CH2-C3-8cycloalkyl, - CH2-O-C3-8cycloalkyl, and C3-8cycloalkenyl, wherein said lkyl, cycloalkenyl or heterocyclic groups may be optionally tuted by one or more Rz groups, and wherein in each instance the heterocyclic group comprises one or more heteroatoms selected from N, O, S and oxidised forms thereof; or the R6 and R7 groups, together with the carbon atom to which they are attached, can join to form a C3-6cycloalkyl or heterocyclyl group with 3 to 6 ring members, wherein the heterocyclic group ses one or more heteroatoms selected from N, O, S and ed forms thereof, and n said C3-6cycloalkyl and heterocyclyl groups may be optionally substituted by one or more Rz groups; R8 and R9 are independently selected from hydrogen, C1-6alkyl, haloC1-6alkyl, hydroxyC1- 6alkyl, -(CH2)k-O-C1-6alkyl, -(CH2)k-O-(hydroxyC1-6alkyl), hydroxyC1-6alkoxy, -(CH2)k-CO2C1-6alkyl, - (CH2)k-CO2H, -C1-6 alkyl-N(H)e(C1-4alkyl)2-e, -(CH2)j-C3-8cycloalkyl and -(CH2)j-C3-8cycloalkenyl; Rx and Ry are independently selected from hydrogen, halogen, nitro, nitrile, C1-6alkyl, haloC1-6alkyl, C2-6alkenyl, C2-6alkynyl, y, hydroxyC1-6alkyl, C1-6alkoxy, -(CH2)k-O-C1- 6alkyl, hydroxyC1-6alkoxy, -COOC1-6alkyl, -N(H)e(C1-4alkyl)2-e, -C1-6alkyl-N(H)e(C1-4alkyl)2-e, - (CH2)k-C(=O)N(H)e(C1-4alkyl)2-e, C3-8cycloalkyl and C3-8cycloalkenyl; or the Rx and Ry groups, together with the carbon or nitrogen atom to which they are attached, can join to form a C3-6cycloalkyl or ted heterocyclyl group with 3 to 6 ring members which may be optionally fused to an aromatic heterocyclyl group of 3 to 5 ring members; or when on a carbon atom the Rx and Ry groups can join together to form a =CH2 group; Rz is independently selected from halogen, nitro, nitrile, C1-6alkyl, haloC1-6alkyl, C2-6alkenyl, C2- 6alkynyl, =O, hydroxy, hydroxyC1-6alkyl, C1-6alkoxy, -(CH2)k-O-C1-6alkyl, hydroxyC1-6alkoxy, - C(=O)C1-6alkyl, -C(=O)C1-6alkyl-OH, -C(=O)C1-6alkyl-N(H)e(C1-4alkyl)2-e, -C(=O)N(H)e(C1- )2-e, -(CH2)r-CO2C1-6alkyl, -(CH2)r-CO2H, -N(H)e(C1-4alkyl)2-e, lkyl-N(H)e(C1-4alkyl)2-e, heterocyclyl group with 3 to 6 ring members, heterocyclyl group with 3 to 6 ring members substituted by -C(=O)C1-4alkyl, heterocyclyl group with 3 to 6 ring s substituted by - C(=O)OC1-4alkyl, cyclyl group with 3 to 6 ring s substituted by -C(=O)N(H)e(C1- 4alkyl)2-e, -C(=O)heterocyclyl group with 3 to 6 ring members, C3-8cycloalkyl and C3- 7 is pyridine then Rz is other than -NH alkenyl, wherein if R 2; a, j, d, e, n, r and p are independently selected from 0, 1 and 2; k and m are independently selected from 1 and 2; u is selected from 0, 1, 2 and 3; and v is selected from 0 and 1.
2. A compound according to claim 1, wherein: A is a C3-6cycloalkyl group or a heterocyclic group with 3 to 6 ring members, wherein the heterocyclic group comprises 1, 2, or 3 heteroatoms selected from N, O, S and oxidised forms
3. A compound according to claim 1 or 2, n: R6 and R7 are independently selected from hydrogen, C1-6alkyl, haloC1-6alkyl, C2- 6alkenyl, C2-6alkynyl, hydroxy, hydroxyC1-6alkyl, -COOC1-6alkyl, -(CH2)j-O-C1-6alkyl, -(CH2)j-O- (hydroxyC1-6alkyl), -C1-6alkyl-NRxRy, -(CRxRy)p-CONRxRy, -(CRxRy)p-NRxCORy, -(CRxRy)p-OCH2-CONRxRy , heterocyclic group with 3 to 7 ring members, -CH2-heterocyclic group with 3 to 7 ring members, -CH2-O-heterocyclic group with 3 to 7 ring members, -CH2-NH-heterocyclic group with 3 to 7 ring members, (C1-6alkyl)-heterocyclic group with 3 to 7 ring members, -C(=O)NH-heterocyclic group with 3 to 7 ring members, C3-8cycloalkyl, -CH2-C3-8cycloalkyl, - CH2-O-C3-8cycloalkyl, and C3-8cycloalkenyl, wherein said cycloalkyl, cycloalkenyl or heterocyclic groups may be optionally substituted by one or more Rz groups, and n in each instance the heterocyclic group ses 1, 2, or 3 heteroatoms selected from N, O, S and oxidised forms f; or the R6 and R7 groups, together with the carbon atom to which they are attached, can join to form a C3-6cycloalkyl or heterocyclyl group with 3 to 6 ring members, wherein the heterocyclic group comprises 1, 2, or 3 heteroatoms selected from N, O, S and oxidised forms f, and wherein said C3-6cycloalkyl and heterocyclyl groups may be ally substituted by one or more Rz groups.
4. A nd according to any one of claims 1 to 3, wherein R1 is halogen, hydroxy, nitrile, C1-4alkyl, C2-4alkynyl, or C1-4alkoxy.
5. A compound according to any one of claims 1 to 4, wherein n is 1 and R1 is chloro or nitrile.
6. A compound according to claim 5 wherein n is 1 and R1 is chloro.
7. A nd according to any one of claims 1 to 4, wherein n is 2 and the substituents R1 are at the ortho- and para-positions of the phenyl ring.
8. A compound according to claim 1, wherein R1 is independently ed from hydroxy, halogen, nitrile, C1-4alkyl, haloC1-4alkyl, hydroxyC1-4alkyl, kenyl, C1-4alkoxy, haloC1- 4alkoxy, kynyl, -(CH2)v-CO2H, -(CRxRy)v-CO2C1-4alkyl, -(CH2)v-CON(C1-4alkyl)2, - P(=O)(Rx)2, -S(O)d-C1-6alkyl, -S(O)d-heterocyclic group with 3 to 6 ring members and - S(O)d-N(R8)2.
9. A compound according to claim 1, wherein n is 2, and one R1 is -(CRxRy)v-CO2C1-4alkyl and one R1 is halogen, nitrile, or C1-4 alkyl, in particular chloro, nitrile or methyl.
10. A nd ing to claim 1, wherein n is 2 and one R1 is -(CRxRy)v-CO2C1-4alkyl and one R1 is chloro.
11. A compound according to claim 9 or 10 wherein n is 2 and one R1 is Cl or F.
12. A compound according to any one of claims 1 to 11, wherein R2 is selected from hydrogen, C1-4 alkyl, C2-6alkenyl, and hydroxyC1-4alkyl.
13. A compound according to any one of claims 1 to 11, wherein R2 is hydrogen or - )u-CO2H.
14. A compound according to claim 13 wherein R2 is -COOH, -CH2COOH, 2-CO2H, -(CH(CH3))-CO2H or -(C(CH3)2)-CO2H.
15. A compound ing to any one of claims 1 to 14, wherein R3 is -(A)t-(CRxRy)q-X and A is a C3-6cycloalkyl group.
16. A nd according to claim 15 wherein R3 is -(A)t-(CRxRy)q-X and A is a cyclopropyl group.
17. A compound according to any one of claims 1 to 14, wherein R3 is -(A)t-(CRxRy)q-X and A is a heterocyclic group with 3 to 5 ring members.
18. A compound according to claim 17 wherein R3 is (CRxRy)q-X and A is a heterocyclic group with 5 ring members.
19. A compound according to any one of claims 1 to 18, wherein s is 0.
20. A compound according to any one of claims 1 to 18, wherein s is 1.
21. A compound according to any one of claims 1 to 20, wherein X is hydrogen, halogen, - CN, -OR9, or -C(=O)NRxRy.
22. A compound according to any one of claims 1 to 14, wherein the compound of formula (I) is a compound of the formula (R5)m (R4)a R6 N (R1)n or a tautomer or a solvate or a pharmaceutically acceptable salt thereof.
23. A compound according to any one of claims 1 to 22, wherein a is 1.
24. A compound according to any one of claims 1 to 23, wherein the compound of formula (I) is a compound of the formula (Ir): ( )s (R5)m R6 N (R1)n (Ir) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof.
25. A compound according to claim 24, wherein the compound is a compound is of the a (Is): ( )s (R5)m R6 N (R1)n (Is) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof.
26. A compound according to any one of claims 1 to 25, wherein m is 1 and the substituent R5 is at the para-position of the phenyl group.
27. A compound ing to any one of claims 1 to 26, wherein R5 is chloro.
28. A compound according to any one of claims 1 to 27, wherein R7 is selected from a heterocyclic group with 3 to 7 ring s and a -CH2-heterocyclic group with 3 to 7 ring members, wherein said heterocyclic groups may be optionally substituted by one or more Rz groups, and wherein in each instance the cyclic group comprises one or more heteroatoms selected from N, O, S and oxidised forms thereof.
29. A compound according to claim 28, n R7 is selected from , piperidinyl, pyrazolyl or imidazolyl optionally substituted by one or more Rz, wherein Rz is selected from halo or C1-4alkyl.
30. A compound according to any one of claims 1 to 29, wherein R6 is methyl or ethyl.
31. A compound ing to claim 1, wherein the compound is of the formula (VIIf): R6 N (R1)n (VIIf) or a tautomer or a solvate or a pharmaceutically acceptable salt f, wherein: n is 1 and R1 is chloro or nitrile; R2 is hydrogen, C1-4 alkyl, or -CH2COOH, -CH2CH2-CO2H or-(CH(CH3))-CO2H; R4 and R5 are halogen; R6 is methyl or ethyl; and R7 is oxanyl, piperidinyl, pyrazolyl or imidazolyl optionally substituted by one or more Rz, where Rz is halo or C1-4alkyl.
32. A compound according to claim 1, n the compound is of the formula (Iw): (Iw) or a tautomer or a solvate or a pharmaceutically acceptable salt thereof, n: n is 1 and R1 is chloro or nitrile; R2 is hydrogen, C1-4 alkyl, or -CH2COOH, 2-CO2H or-(CH(CH3))-CO2H; s is 1 and R3 is hydrogen; R4 is halogen; m is 1 or 2 and R5 is halogen; and Rz is hydrogen or fluorine.
33. A compound of the formula (Iw) according to claim 32, wherein R4 is fluorine.
34. A compound of formula (I) according to claim 1 or a tautomer or a solvate or a pharmaceutically acceptable salt thereof, wherein: n is 1 or 2 and R1 is halogen, nitrile, or -O0,1(CRxRy)vCOOH n v is 0 or 1; R2 is selected from hydrogen and –(CRxRy)u-CO2H wherein u is 0, 1 or 2; Rx and Ry are en or methyl; R3 is hydrogen and s is 1; a is 1 and R4 is n; m is 1 and R5 is halogen; R6 is hydrogen or kyl; R7 is C1-4alkyl, hydroxylC1-4alkyl, methoxyC1-4alkyl, or a heterocyclic group with 5 or 6 ring members, wherein said heterocyclic group with 5 or 6 ring members may be optionally substituted with one or two Rz groups independently selected from C1-4alkyl.
35. A compound according to claim 1, or a tautomer, pharmaceutically acceptable salt or solvate thereof, wherein the compound is selected from: (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- olone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxyethoxy)(2- hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[3-(hydroxymethyl)oxetan yl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone; 1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl) oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarboxylic acid; (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](2,3-dihydroxy methylpropoxy)(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; -(4-chlorophenyl)[(4-chlorophenyl)methyl](1,2-dihydroxypropanyl){[1- (hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](2-hydroxy methoxypropanyl){[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H- isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][1-(dimethylamino) ypropanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H- isoindolone; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl){[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)oxo-2,3-dihydro-1H- isoindolyl]propanoic acid; (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl](1,2-dihydroxypropanyl)- (hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](3-hydroxymethylbutoxy)(2- hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)[(1H- pyrazolyl)methoxy]-2,3-dihydro-1H-isoindolone; 1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl) oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropanecarbonitrile; N-{[1-({[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl) oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropyl]methyl}methanesulfonamide; (3R)(4-chlorophenyl)[(4-ethynylphenyl)methyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; (3R)(4-chlorophenyl)[(4-ethynylphenyl)methyl]fluoro{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; (3R)(4-chlorophenyl)(1,2-dihydroxypropanyl)[(4-ethynylphenyl)methyl] fluoro{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone; R)(4-chlorophenyl)fluoro({1- [hydroxy(2H₂)methyl]cyclopropyl}(2H₂)methoxy)(2-hydroxypropanyl)oxo-2,3- dihydro-1H-isoindolyl]methyl}benzonitrile; 4-{[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}(2- hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)[(3- methyloxetanyl)methoxy]-2,3-dihydro-1H-isoindolone; 4-{[(1R)(4-chlorophenyl)(1,2-dihydroxypropanyl){[1- (hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindol yl]methyl}benzonitrile; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][(1-hydroxycyclopropyl)methoxy]- 6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl){[1- (methoxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone; (3R)(4-Chlorophenyl)[(4-chlorophenyl)methyl]{[1- (hydroxymethyl)cyclobutyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindol- 1-one; 5-chloro{[(1R)(4-chlorophenyl){[1-(hydroxymethyl)cyclopropyl]methoxy}(2- hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid; (3R){[4-chloro(morpholinesulfonyl)phenyl]methyl}(4-chlorophenyl){[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro (2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropane carboxamide; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)({1- xy(2H2)methyl]cyclopropyl}(2H2)methoxy)(2-hydroxypropanyl)-2,3-dihydro- 1H-isoindolone; -(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl) nyloxy)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl) [(oxolanyl)methoxy]-2,3-dihydro-1H-isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1- hydroxy(oxanyl)ethyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H- isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2- hydroxy(piperazinyl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3- dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[(3S,4R)hydroxyoxolan- 3-yl]oxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(1S)(4-chlorophenyl)ethyl]{[(3R,4S)hydroxyoxolan- 3-yl]oxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl) methoxy-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]({1- xy(2H₂)methyl]cyclopropyl}(2H₂)methoxy)(2-hydroxypropanyl)-2,3-dihydro- 1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2-hydroxypropanyl)(3- hydroxypropoxy)-2,3-dihydro-1H-isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; -(4-chlorophenyl)[(4-chlorophenyl)methyl](2,2-difluorohydroxypropoxy)- 6-(2-hydroxypropanyl)-2,3-dihydro-1H-isoindolone; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[2- (hydroxymethyl)cyclobutyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H-isoindol- 1-one; (3R)(4-chlorophenyl)[(4-chlorophenyl)methyl][2-hydroxyoxo(pyrrolidin yl)propanyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindol one; 2-[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]hydroxy- N,N-dimethylpropanamide; 2-[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindolyl]hydroxy-N- methylpropanamide; (3R){[4-chloro(methylsulfanyl)phenyl]methyl}(4-chlorophenyl)fluoro{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; (3R)[(4-chloromethanesulfinylphenyl)methyl](4-chlorophenyl)fluoro{[1- (hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro(2- hydroxymethoxypropanyl){[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3- o-1H-isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)(1,2- dihydroxypropanyl)fluoro{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro- 1H-isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2- hydroxy(4-methylpiperazinyl)propanyl][(3R)-oxolanyloxy]-2,3-dihydro-1H- olone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2- hydroxy(4-methylpiperazinyl)propanyl][(3S)-oxolanyloxy]-2,3-dihydro-1H- isoindolone; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro(2-hydroxypropanyl)- 3-oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]propanoic acid; 1-({[(1R){[4-chloro(hydroxymethyl)phenyl]methyl}(4-chlorophenyl)fluoro (2-hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}methyl)cyclopropane carbonitrile; 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro [1-hydroxy(1-methyl-1H-pyrazolyl)ethyl]oxo-2,3-dihydro-1H-isoindol yl]oxy}methyl)cyclopropanecarboxamide; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1- hydroxy(1-methyl-1H-pyrazolyl)ethyl][(1-hydroxycyclopropyl)methoxy]-2,3- dihydro-1H-isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[2- hydroxy(4-methylpiperazinyl)propanyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H-isoindolone; 5-chloro{[(1R)(4-chlorophenyl)[(1-cyanocyclopropyl)methoxy]fluoro(2- hydroxypropanyl)oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1- y(1-methylpiperidinyl)ethyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}- 2,3-dihydro-1H-isoindolone; (3R){[4-chloro(dimethylphosphoryl)phenyl]methyl}(4-chlorophenyl)fluoro {[1-(hydroxymethyl)cyclopropyl]methoxy}(2-hydroxypropanyl)-2,3-dihydro-1H- isoindolone; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro [hydroxy(oxanyl)methyl]{[1-(hydroxymethyl)cyclopropyl]methoxy}-2,3-dihydro-1H- olone; 1-({[(1R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro [1-hydroxy(oxanyl)ethyl]oxo-2,3-dihydro-1H-isoindol yl]oxy}methyl)cyclopropanecarboxamide; 5-chloro{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methylpiperidin yl)ethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(oxan yl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; 4-[(1R)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazol yl)propyl]oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl] hydroxyethyl]benzonitrile; 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]- 3-oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]methyl} (hydroxymethyl)benzonitrile; 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]- 1-{[1-(hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindol yl]methyl}benzonitrile; 4-{[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl-1H-imidazolyl)propyl]- 3-oxo[(3S)-oxolanyloxy]-2,3-dihydro-1H-isoindolyl]methyl}benzonitrile; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluorooxanyl) hydroxyethyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(1-methyl- azolyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluorooxanyl) hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)(1-cyclobutylhydroxyethyl) fluoromethoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxan- 4-yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxan- 4-yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxan- 4-yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid; (4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxan- 4-yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid; (4S)(4-Chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxan yl)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid; -(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxan yl)hydroxypropyl]trideuteromethoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)ethoxyfluoro[(1R)(4- fluorooxanyl)hydroxypropyl]oxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (4S)[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl]- 1-methoxyoxo-2,3-dihydro-1H-isoindolyl](4-methoxyphenyl)butanoic acid; (4S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro{1-hydroxy[trans hydroxycyclohexyl]propyl}methoxyoxo-2,3-dihydro-1H-isoindolyl]butanoic acid; 2-(5-chloro{[1-(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl) hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenoxy)acetic acid; 5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl] yoxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid; ro{[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxanyl)propyl] methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid; 5-chloro{[(1R)(4-chlorophenyl)ethoxyfluoro[(1S)hydroxy(oxan yl)propyl]oxo-2,3-dihydro-1H-isoindolyl]methyl}benzoic acid; 2-{[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl] methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}methylbenzoic acid; 2-{[(1R)(4-chlorophenyl)fluoro[(1R)(4-fluorooxanyl)hydroxypropyl] methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}methoxybenzoic acid; 2-(5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]- 1-methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)methylpropanoic acid; 2-(5-chloro{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]- 1-methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)acetic acid; 2-(5-chloro{[(1R)(4-chlorophenyl)fluoro[(1R)hydroxy(oxanyl)propyl]- 1-methoxyoxo-2,3-dihydro-1H-isoindolyl]methyl}phenyl)acetic acid; (2S,3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy (oxanyl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]methylpropanoic acid; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[(3-fluorooxetan yl)methoxy](2-hydroxybutanyl)oxo-2,3-dihydro-1H-isoindolyl]propanoic acid; -(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-hydroxy(pyridin yl)propyl]methoxyoxo-2,3-dihydro-1H-isoindolyl]propanoic acid; (3R)[(4-chloromethanesulfonylphenyl)methyl](4-chlorophenyl)fluoro[1-(4- fluoropiperidinyl)hydroxypropyl]methoxy-2,3-dihydro-1H-isoindolone; 4-{[(1R)(4-chlorophenyl)fluoro[(1S)hydroxy(1-methylpiperidin yl)propyl]oxo[cishydroxycyclobutoxy]-2,3-dihydro-1H-isoindol yl]methyl}benzonitrile; (3S)(4-chlorophenyl)[(1R)(4-chlorophenyl)fluoro[1-(4-fluoro methylpiperidinyl)hydroxypropyl]methoxyoxo-2,3-dihydro-1H-isoindol yl]propanoic acid; tert-butyl 2-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro methoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetate; tert-butyl 2-{4-[(1R)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro methoxyoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetate; 2-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy oxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetic acid; 2-{4-[(1R)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy oxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}acetic acid; methyl 3-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoro yoxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidin yl}propanoate; and 3-{4-[(1S)[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl]fluoromethoxy oxo-2,3-dihydro-1H-isoindolyl]hydroxypropyl]piperidinyl}propanoic acid.
36. A compound, or a er, pharmaceutically acceptable salt or solvate thereof, wherein the compound is 2-{[(1R)(4-chlorophenyl)[(4-chlorophenyl)methyl](2- ypropanyl)oxo-2,3-dihydro-1H-isoindolyl]oxy}-N,N-dimethylacetamide.
37. A compound according to claim 1, or a tautomer, ceutically acceptable salt or solvate thereof, wherein the compound is (2S,3S)(4-chlorophenyl)[(1R)(4- chlorophenyl)fluoro[(1S)hydroxy(oxanyl)propyl]methoxyoxo-2,3- dihydro-1H-isoindolyl]methylpropanoic acid.
38. A compound according to claim 1, or a tautomer, pharmaceutically acceptable salt or solvate thereof, wherein the compound is (3S)(4-chlorophenyl)[(1R)(4- chlorophenyl)fluoro[1-hydroxy(oxanyl)ethyl]methoxyoxo-2,3-dihydro- 1H-isoindolyl]propanoic acid.
39. A compound ing to claim 1, or a tautomer, pharmaceutically acceptable salt or solvate thereof, wherein the compound is 2-(5-chloro{[(1R)(4-chlorophenyl) [(1S)hydroxy(oxanyl)propyl]methoxyoxo-2,3-dihydro-1H- isoindolyl]methyl}phenyl)methylpropanoic acid.
40. A compound according to claim 1, or a tautomer, ceutically acceptable salt or solvate thereof, wherein the compound is 4-{[(1R)(4-chlorophenyl)fluoro[1- hydroxy(1-methyl-1H-imidazolyl)propyl]{[1- (hydroxymethyl)cyclopropyl]methoxy}oxo-2,3-dihydro-1H-isoindol yl]methyl}benzonitrile.
41. A combination comprising a compound as defined in any one of claims 1 to 40 with one or more other therapeutic .
42. A combination according to claim 41 wherein the one or more other therapeutic agents are anticancer agents.
43. A combination according to claim 41 or 42 wherein the one or more other therapeutic agents are anticancer agents selected from groups (i) to (xlix) as d below: i. um compounds; ii. taxane compounds; iii. topoisomerase I inhibitors; iv. topoisomerase II inhibitors; v. vinca ids; vi. nucleoside derivatives; vii. antimetabolites; viii. alkylating agents; ix. anthracyclines and anthracenediones; x. epothilones; xi. DNA methyl transferase inhibitors; xii. antifolates; xiii. cytotoxic antibiotics; xiv. tubulin-binding agents; xv. signal transduction inhibitors; xvi. aurora kinase inhibitors; xvii. CDK tors; xviii. PKA/B inhibitors and PKB (akt) pathway tors; xix. hsp90 inhibitors; xx. monoclonal dies (unconjugated or conjugated to radioisotopes, toxins or other agents) and antibody derivatives; xxi. estrogen receptor antagonists or selective estrogen receptor modulators (SERMs) or inhibitors of estrogen synthesis; xxii. aromatase inhibitors; xxiii. antiandrogens (i.e. androgen receptor nists); xxiv. hormones and analogues thereof; xxv. steroids; xxvi. steroidal cytochrome P450 17alpha-hydroxylase-17,20-lyase inhibitor (CYP17); xxvii. gonadotropin releasing hormone agonists or nists (GnRAs); xxviii. glucocorticoids; xxix. differentiating agents; xxx. farnesyltransferase inhibitors; xxxi. tin targeted therapies; xxxii. drugs targeting the ubiquitin-proteasome pathway including proteasome Inhibitors; xxxiii. photodynamic drugs; xxxiv. marine sm-derived anticancer agents; xxxv. radiolabelled drugs for radioimmunotherapy; xxxvi. telomerase tors; xxxvii. matrix metalloproteinase inhibitors; xxxviii. recombinant interferons and interleukins; xxxix. selective immunoresponse tors; xl. therapeutic vaccines; xli. cytokine-activating agents; xlii. arsenic trioxide; xliii. inhibitors of G-protein d receptors (GPCR); xliv. enzymes; xlv. DNA repair inhibitors; xlvi. agonists of death receptor; xlvii. immunotherapies; xlviii. regulators of cell death (apoptosis) antagonists; xlix. prophylactic agents (adjuncts).
44. A pharmaceutical composition comprising a compound as defined in any one of claims 1 to 40 or a combination as defined in any one of claims 41 to 43.
45. A pharmaceutical composition according to claim 44 comprising a compound as defined in any one of claims 1 to 40, one or more other therapeutic , and a pharmaceutically acceptable carrier.
46. A pharmaceutical composition according to claim 45 wherein the one or more other therapeutic agents are anticancer agents.
47. The use of a nd as defined in any one of claims 1 to 40, a combination according to any one of claims 41 to 43, or a pharmaceutical composition ing to any one of claims 44 to 46 for the manufacture of a medicament for the prophylaxis or treatment of a disease state or ion mediated by MDM2-p53.
48. The use of a compound as defined in any one of claims 1 to 40, a combination according to any one of claims 41 to 43, or a pharmaceutical ition according to any one of claims 44 to 46 in the manufacture of a medicament for the prophylaxis or treatment of cancer.
49. The use as defined in claim 48, wherein the medicament is to be used in combination with one or more other compounds or therapies.
50. The use as d in claim 49, wherein one of the one or more therapies is radiotherapy.
51. The use as defined in any one of claims 48 to 50, wherein the cancer is: - tumours of epithelial origin; haematological malignancies and ignant haematological disorders and disorders of borderline malignancy; tumours of mesenchymal origin; tumours of the central or peripheral s system; endocrine s; ocular and adnexal tumours; germ cell and blastic tumours; paediatric and embryonal tumours; or syndromes, congenital or otherwise, which leave the patient susceptible to malignancy; - carcinomas of the bladder and urinary tract, , gastrointestinal tract, liver, gall bladder and biliary system, exocrine pancreas, kidney, lung, head and neck, ovary, ian tubes, peritoneum, vagina, vulva, penis, testes, cervix, myometrium, endometrium, thyroid, brain, adrenal, prostate, skin or adnexae; - haematological malignancies and related conditions of id lineage and haematological malignancies and related conditions of myeloid lineage; - sarcomas of soft tissue, bone or cartilage; astrocytomas neuromas and glioblastomas, meningiomas, ependymomas, pineal tumours and schwannomas; pituitary tumours, adrenal tumours, islet cell tumours, parathyroid tumours, carcinoid s and medullary carcinoma of the thyroid; retinoblastoma; teratomas, seminomas, dysgerminomas, hydatidiform moles and choriocarcinomas; medulloblastoma, lastoma, Wilms tumour, and primitive neuroectodermal tumours; or Xeroderma Pigmentosum; - leukaemia; - leukaemia which is acute myeloid leukaemia (AML), acute lymphocytic leukaemia (ALL), chronic cytic mia (CLL), or chronic myeloid leukaemia (CML); - Lymphoma; - lymphoma which is Burkitt lymphoma, Hodgkin ma, dgkin lymphoma or diffuse large B-cell lymphoma; - a tumour of the brain, a cancer of the skin, a cancer of the lung, a cancer of the gastrointestinal tract, osteosarcoma, liposarcoma, Ewing’s sarcoma, soft tissue sarcoma, oesophageal cancer, colorectal cancer, breast cancer, lung cancer, brain cancer, or a tric cancer; - hepatocellular carcinoma, lung cancer, sarcomas, osteosarcomas, or Hodgkin disease; - glioma or neuroblastoma; - melanoma; - mesothelioma; - malignant peritoneal mesothelioma or malignant pleural elioma; - GIST, gastric, colorectal, colon or bowel ; or - fibrosarcoma.
52. A process for the preparation of a nd as defined in any one of claims 1 to 35 or 37 to 40, or a tautomer, pharmaceutically acceptable salt, or solvate thereof which comprises: (a) reacting a compound of the following a with an organometallic reagent: s R5 R4 a R1 n wherein R1, R2, R3, R4, R5, R7, a, s, m and n are as defined in any one of claims 1 to 34; and/or (b) interconversion of a compound of formula (I) or protected derivative thereof to a further compound of formula (I) or protected derivative thereof; and/or (c) deprotection of a ted derivative of a compound of formula (I); and/or (d) providing a compound of formula (I) and forming a pharmaceutically acceptable salt of the compound.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GB1517217.4 | 2015-09-29 | ||
GBGB1517217.4A GB201517217D0 (en) | 2015-09-29 | 2015-09-29 | Pharmaceutical compounds |
PCT/GB2016/053042 WO2017055860A1 (en) | 2015-09-29 | 2016-09-29 | Isoindolinone inhibitors of the mdm2-p53 interaction having anticancer activity |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ740737A NZ740737A (en) | 2021-10-29 |
NZ740737B2 true NZ740737B2 (en) | 2022-02-01 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2021202962B2 (en) | Isoindolinone inhibitors of the MDM2-p53 interaction having anticancer activity | |
AU2021202973B2 (en) | Isoindolinone inhibitors of the MDM2-p53 interaction having anticancer activity | |
US12077526B2 (en) | Isoindolinone inhibitors of the MDM2-P53 interaction and process for making them | |
NZ740737B2 (en) | Isoindolinone inhibitors of the mdm2-p53 interaction having anticancer activity | |
RU2794333C1 (en) | Isoindolinone inhibitors of mdm2-p53 interaction with anti-cancer activity | |
NZ740859B2 (en) | Isoindolinone inhibitors of the mdm2-p53 interaction having anticancer activity | |
BR112018005936B1 (en) | ISOINDOLINONE INHIBITORS OF MDM2-P53 INTERACTION, PHARMACEUTICAL COMPOSITION, ITS USES AND PROCESS FOR PREPARING THE COMPOUND |