WO2018148721A1 - Highly selective akr1c3 inhibitors and methods of use thereof - Google Patents
Highly selective akr1c3 inhibitors and methods of use thereof Download PDFInfo
- Publication number
- WO2018148721A1 WO2018148721A1 PCT/US2018/017986 US2018017986W WO2018148721A1 WO 2018148721 A1 WO2018148721 A1 WO 2018148721A1 US 2018017986 W US2018017986 W US 2018017986W WO 2018148721 A1 WO2018148721 A1 WO 2018148721A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- mmol
- akr1c3
- inhibitor
- arch
- cells
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 67
- 239000003112 inhibitor Substances 0.000 title claims description 214
- 206010060862 Prostate cancer Diseases 0.000 claims abstract description 103
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims abstract description 102
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims abstract description 37
- 208000029052 T-cell acute lymphoblastic leukemia Diseases 0.000 claims abstract description 28
- 208000032839 leukemia Diseases 0.000 claims abstract description 15
- 206010006187 Breast cancer Diseases 0.000 claims abstract description 10
- 208000026310 Breast neoplasm Diseases 0.000 claims abstract description 10
- 210000004027 cell Anatomy 0.000 claims description 400
- 150000001875 compounds Chemical class 0.000 claims description 213
- 230000002195 synergetic effect Effects 0.000 claims description 55
- 108010029485 Protein Isoforms Proteins 0.000 claims description 47
- 102000001708 Protein Isoforms Human genes 0.000 claims description 47
- 206010028980 Neoplasm Diseases 0.000 claims description 46
- 102000004190 Enzymes Human genes 0.000 claims description 43
- 108090000790 Enzymes Proteins 0.000 claims description 43
- 230000000973 chemotherapeutic effect Effects 0.000 claims description 25
- 230000001225 therapeutic effect Effects 0.000 claims description 23
- 201000011510 cancer Diseases 0.000 claims description 19
- 102000005602 Aldo-Keto Reductases Human genes 0.000 claims description 13
- 108010084469 Aldo-Keto Reductases Proteins 0.000 claims description 13
- 239000002246 antineoplastic agent Substances 0.000 claims description 13
- 229940127089 cytotoxic agent Drugs 0.000 claims description 13
- 230000035755 proliferation Effects 0.000 claims description 13
- 230000001413 cellular effect Effects 0.000 claims description 11
- 210000001519 tissue Anatomy 0.000 claims description 9
- 108010080146 androgen receptors Proteins 0.000 abstract description 66
- 102000001307 androgen receptors Human genes 0.000 abstract description 66
- 239000000203 mixture Substances 0.000 abstract description 51
- 230000014509 gene expression Effects 0.000 abstract description 48
- 102000007066 Prostate-Specific Antigen Human genes 0.000 abstract description 22
- 108010072866 Prostate-Specific Antigen Proteins 0.000 abstract description 22
- 230000002255 enzymatic effect Effects 0.000 abstract description 8
- 230000023603 positive regulation of transcription initiation, DNA-dependent Effects 0.000 abstract description 4
- 108010065942 Prostaglandin-F synthase Proteins 0.000 abstract description 2
- 102000004602 Aldo-Keto Reductase Family 1 Member C3 Human genes 0.000 abstract 1
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 540
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 406
- SMYHHZFAWOLAAX-RVDMUPIBSA-N (E)-3-[4-(3-methylbut-2-enyl)-3-(3-phenylpropanoylamino)phenyl]prop-2-enoic acid Chemical compound CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)O)NC(CCC1=CC=CC=C1)=O)C SMYHHZFAWOLAAX-RVDMUPIBSA-N 0.000 description 211
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 205
- 235000019439 ethyl acetate Nutrition 0.000 description 202
- 101150041968 CDC13 gene Proteins 0.000 description 163
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 163
- 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 154
- 229960004671 enzalutamide Drugs 0.000 description 154
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 150
- 238000006243 chemical reaction Methods 0.000 description 139
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 132
- 238000011282 treatment Methods 0.000 description 129
- 239000000243 solution Substances 0.000 description 124
- 238000005160 1H NMR spectroscopy Methods 0.000 description 116
- 238000001644 13C nuclear magnetic resonance spectroscopy Methods 0.000 description 115
- 238000004440 column chromatography Methods 0.000 description 101
- 230000003833 cell viability Effects 0.000 description 90
- 238000000746 purification Methods 0.000 description 89
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 82
- 230000000694 effects Effects 0.000 description 70
- 230000005764 inhibitory process Effects 0.000 description 70
- 238000002203 pretreatment Methods 0.000 description 69
- 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 68
- 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 68
- 229960000975 daunorubicin Drugs 0.000 description 68
- 239000003921 oil Substances 0.000 description 68
- OKKJLVBELUTLKV-VMNATFBRSA-N methanol-d1 Chemical compound [2H]OC OKKJLVBELUTLKV-VMNATFBRSA-N 0.000 description 67
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 62
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 61
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 60
- DGBITFNXKQHKLI-WXCPUVJDSA-N baccharin Chemical class C([C@@]12[C@]3(C)[C@H]4C[C@H]1O[C@@H]1[C@@H]5O[C@]5(C)CC[C@@]13COC(=O)[C@H]1O[C@@]1(C)[C@@H](O)CO[C@H](\C=C\C=C/C(=O)O4)[C@H](O)C)O2 DGBITFNXKQHKLI-WXCPUVJDSA-N 0.000 description 59
- 230000009467 reduction Effects 0.000 description 59
- 239000002904 solvent Substances 0.000 description 52
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical class CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 description 51
- MUMGGOZAMZWBJJ-DYKIIFRCSA-N Testostosterone Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 MUMGGOZAMZWBJJ-DYKIIFRCSA-N 0.000 description 50
- GTHQOPUWLHFKFZ-NNUXYFOWSA-N Baccharin Natural products CC(O)C1OCC(O)C2(C)OC2C(=O)OCC34CCC5(C)OC5C3OC6CC(OC(=O)C=C/C=C/1)C4C6=O GTHQOPUWLHFKFZ-NNUXYFOWSA-N 0.000 description 45
- 238000011278 co-treatment Methods 0.000 description 44
- 229920006395 saturated elastomer Polymers 0.000 description 43
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 42
- 239000003098 androgen Substances 0.000 description 42
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N triphenylphosphine Chemical compound C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 42
- 230000002401 inhibitory effect Effects 0.000 description 39
- 230000003389 potentiating effect Effects 0.000 description 39
- 230000003013 cytotoxicity Effects 0.000 description 38
- 231100000135 cytotoxicity Toxicity 0.000 description 38
- 238000002474 experimental method Methods 0.000 description 37
- 239000007787 solid Substances 0.000 description 34
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 33
- -1 PGF2a prostanoids Chemical class 0.000 description 33
- 150000001408 amides Chemical class 0.000 description 31
- 108010003133 Aldo-Keto Reductase Family 1 Member C2 Proteins 0.000 description 29
- 102100024089 Aldo-keto reductase family 1 member C2 Human genes 0.000 description 29
- 230000015572 biosynthetic process Effects 0.000 description 29
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 27
- 230000006907 apoptotic process Effects 0.000 description 26
- 230000001965 increasing effect Effects 0.000 description 25
- 229960000905 indomethacin Drugs 0.000 description 25
- 239000000741 silica gel Substances 0.000 description 25
- 229910002027 silica gel Inorganic materials 0.000 description 25
- 229960003604 testosterone Drugs 0.000 description 25
- 230000001472 cytotoxic effect Effects 0.000 description 24
- 150000002148 esters Chemical class 0.000 description 24
- 238000005406 washing Methods 0.000 description 24
- 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 23
- 230000001419 dependent effect Effects 0.000 description 23
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 23
- 229960000549 4-dimethylaminophenol Drugs 0.000 description 21
- 101100030361 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) pph-3 gene Proteins 0.000 description 21
- YJVFFLUZDVXJQI-UHFFFAOYSA-L palladium(ii) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 21
- 238000006467 substitution reaction Methods 0.000 description 21
- 239000003814 drug Substances 0.000 description 20
- 238000011534 incubation Methods 0.000 description 20
- 230000036515 potency Effects 0.000 description 20
- 239000011541 reaction mixture Substances 0.000 description 20
- ISXSCDLOGDJUNJ-UHFFFAOYSA-N tert-butyl prop-2-enoate Chemical compound CC(C)(C)OC(=O)C=C ISXSCDLOGDJUNJ-UHFFFAOYSA-N 0.000 description 19
- 239000012044 organic layer Substances 0.000 description 18
- 238000003786 synthesis reaction Methods 0.000 description 18
- XJLXINKUBYWONI-DQQFMEOOSA-N [[(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2s,3r,4s,5s)-5-(3-carbamoylpyridin-1-ium-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl phosphate Chemical compound NC(=O)C1=CC=C[N+]([C@@H]2[C@H]([C@@H](O)[C@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](OP(O)(O)=O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 XJLXINKUBYWONI-DQQFMEOOSA-N 0.000 description 17
- 230000001640 apoptogenic effect Effects 0.000 description 16
- 229910000024 caesium carbonate Inorganic materials 0.000 description 16
- 229940079593 drug Drugs 0.000 description 15
- OSVMTWJCGUFAOD-KZQROQTASA-N formestane Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CCC2=C1O OSVMTWJCGUFAOD-KZQROQTASA-N 0.000 description 15
- 238000001262 western blot Methods 0.000 description 15
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 14
- 238000001816 cooling Methods 0.000 description 14
- 231100000433 cytotoxic Toxicity 0.000 description 14
- 239000003446 ligand Substances 0.000 description 14
- 238000005556 structure-activity relationship Methods 0.000 description 14
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 13
- 229920000776 Poly(Adenosine diphosphate-ribose) polymerase Polymers 0.000 description 13
- 230000007423 decrease Effects 0.000 description 13
- 125000001844 prenyl group Chemical group [H]C([*])([H])C([H])=C(C([H])([H])[H])C([H])([H])[H] 0.000 description 13
- 238000011002 quantification Methods 0.000 description 13
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 12
- 206010013710 Drug interaction Diseases 0.000 description 12
- 229940030486 androgens Drugs 0.000 description 12
- 230000003247 decreasing effect Effects 0.000 description 12
- 231100000673 dose–response relationship Toxicity 0.000 description 12
- 230000000857 drug effect Effects 0.000 description 12
- 239000000543 intermediate Substances 0.000 description 12
- 230000002829 reductive effect Effects 0.000 description 12
- 239000003610 charcoal Substances 0.000 description 11
- 238000011284 combination treatment Methods 0.000 description 11
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 11
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 11
- 229960005471 androstenedione Drugs 0.000 description 10
- 238000003556 assay Methods 0.000 description 10
- ZADPBFCGQRWHPN-UHFFFAOYSA-N boronic acid Chemical compound OBO ZADPBFCGQRWHPN-UHFFFAOYSA-N 0.000 description 10
- 239000012267 brine Substances 0.000 description 10
- 239000012091 fetal bovine serum Substances 0.000 description 10
- 230000002601 intratumoral effect Effects 0.000 description 10
- 230000007246 mechanism Effects 0.000 description 10
- 230000002018 overexpression Effects 0.000 description 10
- 210000002966 serum Anatomy 0.000 description 10
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 10
- NGNBDVOYPDDBFK-UHFFFAOYSA-N 2-[2,4-di(pentan-2-yl)phenoxy]acetyl chloride Chemical class CCCC(C)C1=CC=C(OCC(Cl)=O)C(C(C)CCC)=C1 NGNBDVOYPDDBFK-UHFFFAOYSA-N 0.000 description 9
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 9
- 102000047561 Aldo-keto reductase family 1 member C Human genes 0.000 description 9
- 108700001449 Aldo-keto reductase family 1 member C Proteins 0.000 description 9
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 9
- 230000003993 interaction Effects 0.000 description 9
- 150000003180 prostaglandins Chemical class 0.000 description 9
- SQICJHHIIRGRSA-ZHACJKMWSA-N (E)-3-[3-(3-methylbut-2-enyl)-5-(2-phenylethylcarbamoyl)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=C(C=1)C(NCCC1=CC=CC=C1)=O)/C=C/C(=O)O)C SQICJHHIIRGRSA-ZHACJKMWSA-N 0.000 description 8
- NVKAWKQGWWIWPM-ABEVXSGRSA-N 17-β-hydroxy-5-α-Androstan-3-one Chemical compound C1C(=O)CC[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@H]21 NVKAWKQGWWIWPM-ABEVXSGRSA-N 0.000 description 8
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 8
- 102100026446 Aldo-keto reductase family 1 member C1 Human genes 0.000 description 8
- 101000718028 Homo sapiens Aldo-keto reductase family 1 member C1 Proteins 0.000 description 8
- 239000001963 growth medium Substances 0.000 description 8
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 8
- 230000004048 modification Effects 0.000 description 8
- 238000012986 modification Methods 0.000 description 8
- 238000010992 reflux Methods 0.000 description 8
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 8
- 230000036962 time dependent Effects 0.000 description 8
- NIXOWILDQLNWCW-UHFFFAOYSA-M Acrylate Chemical compound [O-]C(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-M 0.000 description 7
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 7
- 230000000996 additive effect Effects 0.000 description 7
- 230000003292 diminished effect Effects 0.000 description 7
- 230000007062 hydrolysis Effects 0.000 description 7
- 238000006460 hydrolysis reaction Methods 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 7
- 229910052757 nitrogen Inorganic materials 0.000 description 7
- 230000008506 pathogenesis Effects 0.000 description 7
- 150000003431 steroids Chemical class 0.000 description 7
- JAAJQSRLGAYGKZ-JTQLQIEISA-N (1s)-1,2,3,4-tetrahydronaphthalen-1-ol Chemical compound C1=CC=C2[C@@H](O)CCCC2=C1 JAAJQSRLGAYGKZ-JTQLQIEISA-N 0.000 description 6
- ARDNBQBYCPXKGU-LDADJPATSA-N (E)-3-[4-(3-methylbut-2-enyl)-3-[3-[3-(4-methylphenyl)sulfonyloxyphenyl]propanoylamino]phenyl]prop-2-enoic acid Chemical compound CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)O)NC(CCC1=CC(=CC=C1)OS(=O)(=O)C1=CC=C(C)C=C1)=O)C ARDNBQBYCPXKGU-LDADJPATSA-N 0.000 description 6
- HZKNHDLUFBYIQN-VMPITWQZSA-N (e)-3-[4-hydroxy-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound CC(C)=CCC1=CC(\C=C\C(O)=O)=CC=C1O HZKNHDLUFBYIQN-VMPITWQZSA-N 0.000 description 6
- IATHVBOBDPRFCI-UHFFFAOYSA-N 4-iodo-2-(3-methylbut-2-enyl)phenol Chemical compound CC(C)=CCC1=CC(I)=CC=C1O IATHVBOBDPRFCI-UHFFFAOYSA-N 0.000 description 6
- 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 6
- VPGRYOFKCNULNK-ACXQXYJUSA-N Deoxycorticosterone acetate Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)COC(=O)C)[C@@]1(C)CC2 VPGRYOFKCNULNK-ACXQXYJUSA-N 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- 230000009471 action Effects 0.000 description 6
- 239000004305 biphenyl Substances 0.000 description 6
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 6
- 230000008859 change Effects 0.000 description 6
- 239000003795 chemical substances by application Substances 0.000 description 6
- 229960000684 cytarabine Drugs 0.000 description 6
- ZESRJSPZRDMNHY-UHFFFAOYSA-N de-oxy corticosterone Natural products O=C1CCC2(C)C3CCC(C)(C(CC4)C(=O)CO)C4C3CCC2=C1 ZESRJSPZRDMNHY-UHFFFAOYSA-N 0.000 description 6
- 229960003654 desoxycortone Drugs 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 230000018109 developmental process Effects 0.000 description 6
- 230000001404 mediated effect Effects 0.000 description 6
- 229940127293 prostanoid Drugs 0.000 description 6
- 238000010186 staining Methods 0.000 description 6
- 239000000758 substrate Substances 0.000 description 6
- 238000012360 testing method Methods 0.000 description 6
- AEZZZIYTCUXWPU-UHFFFAOYSA-N 3-bromo-5-iodo-N-(2-phenylethyl)benzamide Chemical compound BrC=1C=C(C(=O)NCCC2=CC=CC=C2)C=C(C=1)I AEZZZIYTCUXWPU-UHFFFAOYSA-N 0.000 description 5
- HHTYEQWCHQEJNV-UHFFFAOYSA-N 4-bromo-2-iodoaniline Chemical compound NC1=CC=C(Br)C=C1I HHTYEQWCHQEJNV-UHFFFAOYSA-N 0.000 description 5
- 239000000654 additive Substances 0.000 description 5
- 238000009167 androgen deprivation therapy Methods 0.000 description 5
- 210000000692 cap cell Anatomy 0.000 description 5
- 150000007942 carboxylates Chemical group 0.000 description 5
- 230000005754 cellular signaling Effects 0.000 description 5
- 238000006880 cross-coupling reaction Methods 0.000 description 5
- HZKNHDLUFBYIQN-UHFFFAOYSA-N drupanin Natural products CC(C)=CCC1=CC(C=CC(O)=O)=CC=C1O HZKNHDLUFBYIQN-UHFFFAOYSA-N 0.000 description 5
- 229910052739 hydrogen Inorganic materials 0.000 description 5
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 5
- 230000003211 malignant effect Effects 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 229930014626 natural product Natural products 0.000 description 5
- 230000037361 pathway Effects 0.000 description 5
- 239000002243 precursor Substances 0.000 description 5
- 208000023958 prostate neoplasm Diseases 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 108090000623 proteins and genes Proteins 0.000 description 5
- 230000004044 response Effects 0.000 description 5
- 125000001424 substituent group Chemical group 0.000 description 5
- ZDAFNOSGSMUQEY-GXDHUFHOSA-N (E)-3-[4-(3-methylbut-2-enyl)-3-(3-phenylpropanoyloxy)phenyl]prop-2-enoic acid Chemical compound CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)O)OC(CCC1=CC=CC=C1)=O)C ZDAFNOSGSMUQEY-GXDHUFHOSA-N 0.000 description 4
- SMZOUWXMTYCWNB-UHFFFAOYSA-N 2-(2-methoxy-5-methylphenyl)ethanamine Chemical compound COC1=CC=C(C)C=C1CCN SMZOUWXMTYCWNB-UHFFFAOYSA-N 0.000 description 4
- NIXOWILDQLNWCW-UHFFFAOYSA-N 2-Propenoic acid Natural products OC(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-N 0.000 description 4
- MKJBJYCBKXPQSY-UHFFFAOYSA-N 3-bromo-5-iodobenzoic acid Chemical compound OC(=O)C1=CC(Br)=CC(I)=C1 MKJBJYCBKXPQSY-UHFFFAOYSA-N 0.000 description 4
- MFEILWXBDBCWKF-UHFFFAOYSA-N 3-phenylpropanoyl chloride Chemical compound ClC(=O)CCC1=CC=CC=C1 MFEILWXBDBCWKF-UHFFFAOYSA-N 0.000 description 4
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 4
- 102000007469 Actins Human genes 0.000 description 4
- 108010085238 Actins Proteins 0.000 description 4
- PAYRUJLWNCNPSJ-UHFFFAOYSA-N Aniline Chemical compound NC1=CC=CC=C1 PAYRUJLWNCNPSJ-UHFFFAOYSA-N 0.000 description 4
- 206010059866 Drug resistance Diseases 0.000 description 4
- IVDFJHOHABJVEH-UHFFFAOYSA-N HOCMe2CMe2OH Natural products CC(C)(O)C(C)(C)O IVDFJHOHABJVEH-UHFFFAOYSA-N 0.000 description 4
- 108010044467 Isoenzymes Proteins 0.000 description 4
- 241001529936 Murinae Species 0.000 description 4
- 241000283973 Oryctolagus cuniculus Species 0.000 description 4
- 229930182555 Penicillin Natural products 0.000 description 4
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 4
- 238000009825 accumulation Methods 0.000 description 4
- 229940048053 acrylate Drugs 0.000 description 4
- 239000002671 adjuvant Substances 0.000 description 4
- 230000000240 adjuvant effect Effects 0.000 description 4
- AEMFNILZOJDQLW-QAGGRKNESA-N androst-4-ene-3,17-dione Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CCC2=C1 AEMFNILZOJDQLW-QAGGRKNESA-N 0.000 description 4
- 230000000118 anti-neoplastic effect Effects 0.000 description 4
- 230000037396 body weight Effects 0.000 description 4
- 229910052799 carbon Inorganic materials 0.000 description 4
- 230000004663 cell proliferation Effects 0.000 description 4
- 238000002512 chemotherapy Methods 0.000 description 4
- 230000008878 coupling Effects 0.000 description 4
- 238000010168 coupling process Methods 0.000 description 4
- 238000005859 coupling reaction Methods 0.000 description 4
- 238000013461 design Methods 0.000 description 4
- 239000000890 drug combination Substances 0.000 description 4
- 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 4
- 229960005420 etoposide Drugs 0.000 description 4
- 238000011156 evaluation Methods 0.000 description 4
- 230000001747 exhibiting effect Effects 0.000 description 4
- 235000019253 formic acid Nutrition 0.000 description 4
- 239000007789 gas Substances 0.000 description 4
- 229940088597 hormone Drugs 0.000 description 4
- 239000005556 hormone Substances 0.000 description 4
- 230000003301 hydrolyzing effect Effects 0.000 description 4
- 210000004185 liver Anatomy 0.000 description 4
- 239000012528 membrane Substances 0.000 description 4
- 230000002503 metabolic effect Effects 0.000 description 4
- 229940049954 penicillin Drugs 0.000 description 4
- 239000012071 phase Substances 0.000 description 4
- BHMBVRSPMRCCGG-OUTUXVNYSA-N prostaglandin D2 Chemical compound CCCCC[C@H](O)\C=C\[C@@H]1[C@@H](C\C=C/CCCC(O)=O)[C@@H](O)CC1=O BHMBVRSPMRCCGG-OUTUXVNYSA-N 0.000 description 4
- BHMBVRSPMRCCGG-UHFFFAOYSA-N prostaglandine D2 Natural products CCCCCC(O)C=CC1C(CC=CCCCC(O)=O)C(O)CC1=O BHMBVRSPMRCCGG-UHFFFAOYSA-N 0.000 description 4
- 210000002307 prostate Anatomy 0.000 description 4
- 230000035945 sensitivity Effects 0.000 description 4
- 230000001235 sensitizing effect Effects 0.000 description 4
- 229960005322 streptomycin Drugs 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- UFFBMXYSUXPKQT-MDZDMXLPSA-N tert-butyl (E)-3-[3-bromo-5-(2-phenylethylcarbamoyl)phenyl]prop-2-enoate Chemical compound BrC=1C=C(C=C(C=1)C(NCCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C UFFBMXYSUXPKQT-MDZDMXLPSA-N 0.000 description 4
- 230000001988 toxicity Effects 0.000 description 4
- 231100000419 toxicity Toxicity 0.000 description 4
- LWIHDJKSTIGBAC-UHFFFAOYSA-K tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 4
- YBXLQOBTRHFZQO-RVDMUPIBSA-N (E)-3-[3-(3-methylbut-2-enyl)-4-(3-phenylpropanoylamino)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=CC=1NC(CCC1=CC=CC=C1)=O)/C=C/C(=O)O)C YBXLQOBTRHFZQO-RVDMUPIBSA-N 0.000 description 3
- JSSDGTGEUJNBDG-ABRJVYRDSA-N (E)-3-[4-(3-methylbut-2-enyl)-3-(3-phenylprop-2-enoylamino)phenyl]prop-2-enoic acid Chemical compound C(C=CC1=CC=CC=C1)(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O JSSDGTGEUJNBDG-ABRJVYRDSA-N 0.000 description 3
- CNYLAOOHUMZJOL-JLHYYAGUSA-N (E)-3-[4-benzoyloxy-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound C(C1=CC=CC=C1)(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C CNYLAOOHUMZJOL-JLHYYAGUSA-N 0.000 description 3
- MYLHYIBMEPENHL-JLHYYAGUSA-N (E)-3-[4-hexanoyloxy-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound C(CCCCC)(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C MYLHYIBMEPENHL-JLHYYAGUSA-N 0.000 description 3
- UUJHQFZWUNZXHM-BRBUPDJYSA-N (E)-3-[5-[(E)-3-methylbut-1-enyl]-2-(3-phenylpropanoylamino)phenyl]prop-2-enoic acid Chemical compound CC(/C=C/C=1C=CC(=C(C=1)/C=C/C(=O)O)NC(CCC1=CC=CC=C1)=O)C UUJHQFZWUNZXHM-BRBUPDJYSA-N 0.000 description 3
- BJIVLGIZOMJQRT-GXDHUFHOSA-N (e)-3-[3-(3-methylbut-2-enyl)-4-(3-phenylpropanoyloxy)phenyl]prop-2-enoic acid Chemical compound CC(C)=CCC1=CC(\C=C\C(O)=O)=CC=C1OC(=O)CCC1=CC=CC=C1 BJIVLGIZOMJQRT-GXDHUFHOSA-N 0.000 description 3
- 125000000094 2-phenylethyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])([H])* 0.000 description 3
- UKMJNLBMILIWFH-UHFFFAOYSA-N 3-(2-fluorophenyl)propanoyl chloride Chemical compound FC1=CC=CC=C1CCC(Cl)=O UKMJNLBMILIWFH-UHFFFAOYSA-N 0.000 description 3
- XMEQDAIDOBVHEK-UHFFFAOYSA-N 3-bromo-4-hydroxybenzoic acid Chemical compound OC(=O)C1=CC=C(O)C(Br)=C1 XMEQDAIDOBVHEK-UHFFFAOYSA-N 0.000 description 3
- VLVCDUSVTXIWGW-UHFFFAOYSA-N 4-iodoaniline Chemical compound NC1=CC=C(I)C=C1 VLVCDUSVTXIWGW-UHFFFAOYSA-N 0.000 description 3
- LCQWCASSXVDSTJ-DUXPYHPUSA-N BrC1=C(C=C(C=C1)/C=C/C(=O)O)O Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)O)O LCQWCASSXVDSTJ-DUXPYHPUSA-N 0.000 description 3
- 206010061818 Disease progression Diseases 0.000 description 3
- 239000000579 Gonadotropin-Releasing Hormone Substances 0.000 description 3
- 229910004373 HOAc Inorganic materials 0.000 description 3
- 102100029100 Hematopoietic prostaglandin D synthase Human genes 0.000 description 3
- 101000988802 Homo sapiens Hematopoietic prostaglandin D synthase Proteins 0.000 description 3
- 241000241413 Propolis Species 0.000 description 3
- 229940124639 Selective inhibitor Drugs 0.000 description 3
- 101000857870 Squalus acanthias Gonadoliberin Proteins 0.000 description 3
- NGCGMRBZPXEPOZ-HBBGHHHDSA-N acetic acid;(2s)-n-[(2s)-1-[[(2s)-1-[[(2s)-1-[[(2s)-1-[[2-[[(2s)-1-[[(2s)-1-[(2s)-2-[(2-amino-2-oxoethyl)carbamoyl]pyrrolidin-1-yl]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-2-oxoethyl]amino]-3-(4-hydroxyphenyl)- Chemical compound CC(O)=O.C([C@@H](C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(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=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 NGCGMRBZPXEPOZ-HBBGHHHDSA-N 0.000 description 3
- 239000012190 activator Substances 0.000 description 3
- 150000001336 alkenes Chemical class 0.000 description 3
- 125000003368 amide group Chemical group 0.000 description 3
- 238000010640 amide synthesis reaction Methods 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 230000003042 antagnostic effect Effects 0.000 description 3
- 239000005557 antagonist Substances 0.000 description 3
- 229940045799 anthracyclines and related substance Drugs 0.000 description 3
- 230000009286 beneficial effect Effects 0.000 description 3
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 3
- 230000006696 biosynthetic metabolic pathway Effects 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 210000000481 breast Anatomy 0.000 description 3
- 229910002092 carbon dioxide Inorganic materials 0.000 description 3
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 3
- 230000030833 cell death Effects 0.000 description 3
- 230000024245 cell differentiation Effects 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 238000011260 co-administration Methods 0.000 description 3
- 230000002860 competitive effect Effects 0.000 description 3
- 230000005750 disease progression Effects 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 230000009977 dual effect Effects 0.000 description 3
- 238000010931 ester hydrolysis Methods 0.000 description 3
- 238000003818 flash chromatography Methods 0.000 description 3
- 229940035638 gonadotropin-releasing hormone Drugs 0.000 description 3
- 238000004128 high performance liquid chromatography Methods 0.000 description 3
- 239000001257 hydrogen Substances 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 3
- 230000014759 maintenance of location Effects 0.000 description 3
- VFSOQXCLNBJPHF-HWKANZROSA-N methyl (e)-3-(4-bromo-3-hydroxyphenyl)prop-2-enoate Chemical compound COC(=O)\C=C\C1=CC=C(Br)C(O)=C1 VFSOQXCLNBJPHF-HWKANZROSA-N 0.000 description 3
- 210000000066 myeloid cell Anatomy 0.000 description 3
- 231100001083 no cytotoxicity Toxicity 0.000 description 3
- 231100000252 nontoxic Toxicity 0.000 description 3
- 230000003000 nontoxic effect Effects 0.000 description 3
- 230000003647 oxidation Effects 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- 229910052698 phosphorus Inorganic materials 0.000 description 3
- 229920001184 polypeptide Polymers 0.000 description 3
- 238000011533 pre-incubation Methods 0.000 description 3
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 3
- 229960004618 prednisone Drugs 0.000 description 3
- 102000004196 processed proteins & peptides Human genes 0.000 description 3
- 108090000765 processed proteins & peptides Proteins 0.000 description 3
- 229940069949 propolis Drugs 0.000 description 3
- 210000001095 prostate stromal cell Anatomy 0.000 description 3
- 230000000717 retained effect Effects 0.000 description 3
- 230000002441 reversible effect Effects 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 239000007858 starting material Substances 0.000 description 3
- 230000000365 steroidogenetic effect Effects 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 230000002194 synthesizing effect Effects 0.000 description 3
- IKYYFIXBKXVVLF-QGOAFFKASA-N tert-butyl (E)-3-[3-[3-(2-methoxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound COC1=C(C=CC=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C IKYYFIXBKXVVLF-QGOAFFKASA-N 0.000 description 3
- IWXKISSGWAOAST-LDADJPATSA-N tert-butyl (E)-3-[3-[3-(4-methoxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound COC1=CC=C(C=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C IWXKISSGWAOAST-LDADJPATSA-N 0.000 description 3
- XDKQTTWJWWYHKC-OBGWFSINSA-N tert-butyl (E)-3-[5-(3-methylbut-2-enyl)-2-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound CC(=CCC=1C=CC(=C(C=1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=CC=C1)=O)C XDKQTTWJWWYHKC-OBGWFSINSA-N 0.000 description 3
- 229940126585 therapeutic drug Drugs 0.000 description 3
- 238000004809 thin layer chromatography Methods 0.000 description 3
- KKSDGJDHHZEWEP-SNAWJCMRSA-N trans-3-coumaric acid Chemical compound OC(=O)\C=C\C1=CC=CC(O)=C1 KKSDGJDHHZEWEP-SNAWJCMRSA-N 0.000 description 3
- 230000004614 tumor growth Effects 0.000 description 3
- PQDDLPYXRPBFHH-NTCAYCPXSA-N (E)-3-[3-(3-methylbut-2-enyl)-4-(2-naphthalen-1-ylacetyl)oxyphenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=CC=1OC(CC1=CC=CC2=CC=CC=C12)=O)/C=C/C(=O)O)C PQDDLPYXRPBFHH-NTCAYCPXSA-N 0.000 description 2
- XBOZIXOUBAFKSS-DTQAZKPQSA-N (E)-3-[3-(3-methylbut-2-enyl)-4-(4-phenylbutanoylamino)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=CC=1NC(CCCC1=CC=CC=C1)=O)/C=C/C(=O)O)C XBOZIXOUBAFKSS-DTQAZKPQSA-N 0.000 description 2
- GHNNGIVFHRQWEI-DTQAZKPQSA-N (E)-3-[3-(3-methylbut-2-enyl)-4-(4-phenylbutanoyloxy)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=CC=1OC(CCCC1=CC=CC=C1)=O)/C=C/C(=O)O)C GHNNGIVFHRQWEI-DTQAZKPQSA-N 0.000 description 2
- CCMZFAUZKKRFQU-NTCAYCPXSA-N (E)-3-[3-(3-methylbut-2-enyl)-4-[(2-naphthalen-1-ylacetyl)amino]phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=CC=1NC(CC1=CC=CC2=CC=CC=C12)=O)/C=C/C(=O)O)C CCMZFAUZKKRFQU-NTCAYCPXSA-N 0.000 description 2
- OLZABESEMNMHRL-ACCUITESSA-N (E)-3-[3-(3-methylbut-2-enyl)-5-(3-phenylpropanoylamino)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=C(C=1)NC(CCC1=CC=CC=C1)=O)/C=C/C(=O)O)C OLZABESEMNMHRL-ACCUITESSA-N 0.000 description 2
- YRMVCBBTNHSTDS-NTEUORMPSA-N (E)-3-[3-[3-(2-fluorophenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound FC1=C(C=CC=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O YRMVCBBTNHSTDS-NTEUORMPSA-N 0.000 description 2
- ITJWNJNADAZNEJ-XNTDXEJSSA-N (E)-3-[3-[3-(2-methoxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound COC1=C(C=CC=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O ITJWNJNADAZNEJ-XNTDXEJSSA-N 0.000 description 2
- GUNJMEYLTHSIQB-UKTHLTGXSA-N (E)-3-[3-[3-(3-hydroxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound OC=1C=C(C=CC=1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O GUNJMEYLTHSIQB-UKTHLTGXSA-N 0.000 description 2
- LMUONPDACJHQEA-RIYZIHGNSA-N (E)-3-[3-[3-(4-fluorophenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound FC1=CC=C(C=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O LMUONPDACJHQEA-RIYZIHGNSA-N 0.000 description 2
- JZIHAEYWABPPTA-RIYZIHGNSA-N (E)-3-[3-[3-(4-hydroxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound OC1=CC=C(C=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O JZIHAEYWABPPTA-RIYZIHGNSA-N 0.000 description 2
- QZCNIPPNUYMMDD-OQLLNIDSSA-N (E)-3-[3-[3-(4-methoxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound COC1=CC=C(C=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)O QZCNIPPNUYMMDD-OQLLNIDSSA-N 0.000 description 2
- MHNIWVZVNNDUAR-ZRDIBKRKSA-N (E)-3-[3-bromo-4-(4-phenylbutanoylamino)phenyl]prop-2-enoic acid Chemical compound BrC=1C=C(C=CC=1NC(CCCC1=CC=CC=C1)=O)/C=C/C(=O)O MHNIWVZVNNDUAR-ZRDIBKRKSA-N 0.000 description 2
- FHTRVTIJXUELJV-ACCUITESSA-N (E)-3-[4-(2-cyclohexylacetyl)oxy-2-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound C1(CCCCC1)CC(=O)OC1=CC(=C(C=C1)/C=C/C(=O)O)CC=C(C)C FHTRVTIJXUELJV-ACCUITESSA-N 0.000 description 2
- JGXBEQLENIWIHM-JLHYYAGUSA-N (E)-3-[4-[(2-cyclohexylacetyl)amino]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound C1(CCCCC1)CC(=O)NC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C JGXBEQLENIWIHM-JLHYYAGUSA-N 0.000 description 2
- RBXZTUMBBNLRIR-UKTHLTGXSA-N (E)-3-[4-[3-(2-fluorophenyl)propanoyloxy]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound FC1=C(C=CC=C1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C RBXZTUMBBNLRIR-UKTHLTGXSA-N 0.000 description 2
- XXWIECAEYLHMIQ-GXDHUFHOSA-N (E)-3-[4-[3-(2-methoxyphenyl)propanoyloxy]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound COC1=C(C=CC=C1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C XXWIECAEYLHMIQ-GXDHUFHOSA-N 0.000 description 2
- LWBCCENYDJMKRN-XYOKQWHBSA-N (E)-3-[4-[3-(3-fluorophenyl)propanoyloxy]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound FC=1C=C(C=CC=1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C LWBCCENYDJMKRN-XYOKQWHBSA-N 0.000 description 2
- IAGWXKGRCDLGHI-RIYZIHGNSA-N (E)-3-[4-[3-(4-methoxyphenyl)propanoyloxy]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound COC1=CC=C(C=C1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C IAGWXKGRCDLGHI-RIYZIHGNSA-N 0.000 description 2
- VHIRBAOXWDEDNS-RMKNXTFCSA-N (E)-3-[4-acetamido-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound C(C)(=O)NC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C VHIRBAOXWDEDNS-RMKNXTFCSA-N 0.000 description 2
- UIHYOUXBKVGOJC-JXMROGBWSA-N (E)-3-[4-bromo-3-(3-phenylpropanoyloxy)phenyl]prop-2-enoic acid Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)O)OC(CCC1=CC=CC=C1)=O UIHYOUXBKVGOJC-JXMROGBWSA-N 0.000 description 2
- LKJPYSCBVHEWIU-KRWDZBQOSA-N (R)-bicalutamide Chemical compound C([C@@](O)(C)C(=O)NC=1C=C(C(C#N)=CC=1)C(F)(F)F)S(=O)(=O)C1=CC=C(F)C=C1 LKJPYSCBVHEWIU-KRWDZBQOSA-N 0.000 description 2
- HZLABQIGSINVSS-RMKNXTFCSA-N (e)-3-[4-acetyloxy-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound CC(C)=CCC1=CC(\C=C\C(O)=O)=CC=C1OC(C)=O HZLABQIGSINVSS-RMKNXTFCSA-N 0.000 description 2
- WBYWAXJHAXSJNI-VOTSOKGWSA-M .beta-Phenylacrylic acid Natural products [O-]C(=O)\C=C\C1=CC=CC=C1 WBYWAXJHAXSJNI-VOTSOKGWSA-M 0.000 description 2
- LOYZVRIHVZEDMW-UHFFFAOYSA-N 1-bromo-3-methylbut-2-ene Chemical compound CC(C)=CCBr LOYZVRIHVZEDMW-UHFFFAOYSA-N 0.000 description 2
- YFTHTJAPODJVSL-UHFFFAOYSA-N 2-(1-benzothiophen-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane Chemical compound O1C(C)(C)C(C)(C)OB1C1=CC=C(SC=C2)C2=C1 YFTHTJAPODJVSL-UHFFFAOYSA-N 0.000 description 2
- CZVNCMHHGGKNKP-UHFFFAOYSA-N 2-[4-iodo-2-(3-methylbut-2-enyl)phenyl]-3-phenylpropanoic acid Chemical compound CC(=CCC1=C(C=CC(=C1)I)C(CC2=CC=CC=C2)C(=O)O)C CZVNCMHHGGKNKP-UHFFFAOYSA-N 0.000 description 2
- LFGNVAOGMIWOEZ-UHFFFAOYSA-N 2-[4-iodo-2-(3-methylbut-2-enyl)phenyl]-4-phenylbutanoic acid Chemical compound CC(=CCC1=C(C=CC(=C1)I)C(CCC2=CC=CC=C2)C(=O)O)C LFGNVAOGMIWOEZ-UHFFFAOYSA-N 0.000 description 2
- HHWOYRKNRZSNQE-UHFFFAOYSA-N 2-bromo-4-iodoaniline Chemical compound NC1=CC=C(I)C=C1Br HHWOYRKNRZSNQE-UHFFFAOYSA-N 0.000 description 2
- LNVKBOZTHZJQLM-UHFFFAOYSA-N 2-bromo-5-iodoaniline Chemical compound NC1=CC(I)=CC=C1Br LNVKBOZTHZJQLM-UHFFFAOYSA-N 0.000 description 2
- VABYVFZVTIDNOA-UHFFFAOYSA-N 2-cyclohexylacetyl chloride Chemical compound ClC(=O)CC1CCCCC1 VABYVFZVTIDNOA-UHFFFAOYSA-N 0.000 description 2
- WRMNZCZEMHIOCP-UHFFFAOYSA-N 2-phenylethanol Chemical compound OCCC1=CC=CC=C1 WRMNZCZEMHIOCP-UHFFFAOYSA-N 0.000 description 2
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 2
- 108010060191 3(17)-hydroxysteroid dehydrogenase Proteins 0.000 description 2
- 108010070743 3(or 17)-beta-hydroxysteroid dehydrogenase Proteins 0.000 description 2
- YEGXZBYGDXGWJU-UHFFFAOYSA-N 3-(2-methoxyphenyl)propanoyl chloride Chemical compound COC1=CC=CC=C1CCC(Cl)=O YEGXZBYGDXGWJU-UHFFFAOYSA-N 0.000 description 2
- DEGMFYNULJKSFA-UHFFFAOYSA-N 3-(3-fluorophenyl)propanoyl chloride Chemical compound FC1=CC=CC(CCC(Cl)=O)=C1 DEGMFYNULJKSFA-UHFFFAOYSA-N 0.000 description 2
- YIBRVNQKMYAOIL-UHFFFAOYSA-N 3-(3-methoxyphenyl)propanoyl chloride Chemical compound COC1=CC=CC(CCC(Cl)=O)=C1 YIBRVNQKMYAOIL-UHFFFAOYSA-N 0.000 description 2
- FQVJPHCAWYRYCK-UHFFFAOYSA-N 3-(4-methoxyphenyl)propanoyl chloride Chemical compound COC1=CC=C(CCC(Cl)=O)C=C1 FQVJPHCAWYRYCK-UHFFFAOYSA-N 0.000 description 2
- HCLQARMRCPEALF-DNQXCXABSA-N 3-[[(2r)-2-[(1r)-2-[[1-(1-benzothiophen-2-yl)-2-methylpropan-2-yl]amino]-1-hydroxyethyl]pyrrolidin-1-yl]methyl]benzonitrile Chemical compound C([C@@H]1[C@H](O)CNC(C)(CC=2SC3=CC=CC=C3C=2)C)CCN1CC1=CC=CC(C#N)=C1 HCLQARMRCPEALF-DNQXCXABSA-N 0.000 description 2
- SCXCUTCIFLAPMW-UHFFFAOYSA-N 3-bromo-5-iodoaniline Chemical compound NC1=CC(Br)=CC(I)=C1 SCXCUTCIFLAPMW-UHFFFAOYSA-N 0.000 description 2
- DPGSPRJLAZGUBQ-UHFFFAOYSA-N 4,4,5,5-tetramethyl-2-vinyl-1,3,2-dioxaborolane Substances CC1(C)OB(C=C)OC1(C)C DPGSPRJLAZGUBQ-UHFFFAOYSA-N 0.000 description 2
- VSMDINRNYYEDRN-UHFFFAOYSA-N 4-iodophenol Chemical compound OC1=CC=C(I)C=C1 VSMDINRNYYEDRN-UHFFFAOYSA-N 0.000 description 2
- 241000531891 Alburnus alburnus Species 0.000 description 2
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 2
- 108010037003 Buserelin Proteins 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 2
- WBYWAXJHAXSJNI-SREVYHEPSA-N Cinnamic acid Chemical compound OC(=O)\C=C/C1=CC=CC=C1 WBYWAXJHAXSJNI-SREVYHEPSA-N 0.000 description 2
- 102100034067 Dehydrogenase/reductase SDR family member 11 Human genes 0.000 description 2
- 208000035859 Drug effect increased Diseases 0.000 description 2
- QGXBDMJGAMFCBF-UHFFFAOYSA-N Etiocholanolone Natural products C1C(O)CCC2(C)C3CCC(C)(C(CC4)=O)C4C3CCC21 QGXBDMJGAMFCBF-UHFFFAOYSA-N 0.000 description 2
- 102000053187 Glucuronidase Human genes 0.000 description 2
- 108010060309 Glucuronidase Proteins 0.000 description 2
- BLCLNMBMMGCOAS-URPVMXJPSA-N Goserelin Chemical compound C([C@@H](C(=O)N[C@H](COC(C)(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NNC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 BLCLNMBMMGCOAS-URPVMXJPSA-N 0.000 description 2
- 108010069236 Goserelin Proteins 0.000 description 2
- 238000007341 Heck reaction Methods 0.000 description 2
- 101000896517 Homo sapiens Steroid 17-alpha-hydroxylase/17,20 lyase Proteins 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 206010020802 Hypertensive crisis Diseases 0.000 description 2
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- FKPDQJDUTSAOAC-UHFFFAOYSA-N N-(3-bromo-5-iodophenyl)-3-phenylpropanamide Chemical compound BrC=1C=C(C=C(C=1)I)NC(CCC1=CC=CC=C1)=O FKPDQJDUTSAOAC-UHFFFAOYSA-N 0.000 description 2
- MHMXEJJHCNOFHP-UHFFFAOYSA-N N-(4-bromo-2-iodophenyl)-3-phenylpropanamide Chemical compound BrC1=CC(=C(C=C1)NC(CCC1=CC=CC=C1)=O)I MHMXEJJHCNOFHP-UHFFFAOYSA-N 0.000 description 2
- BAWFJGJZGIEFAR-NNYOXOHSSA-O NAD(+) Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 BAWFJGJZGIEFAR-NNYOXOHSSA-O 0.000 description 2
- 102000004316 Oxidoreductases Human genes 0.000 description 2
- 108090000854 Oxidoreductases Proteins 0.000 description 2
- BHHGXPLMPWCGHP-UHFFFAOYSA-N Phenethylamine Chemical compound NCCC1=CC=CC=C1 BHHGXPLMPWCGHP-UHFFFAOYSA-N 0.000 description 2
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical group C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 2
- 238000004617 QSAR study Methods 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 108091027981 Response element Proteins 0.000 description 2
- 102100021719 Steroid 17-alpha-hydroxylase/17,20 lyase Human genes 0.000 description 2
- 238000006161 Suzuki-Miyaura coupling reaction Methods 0.000 description 2
- OPHXUUMKVSYQLI-FOWTUZBSSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] 3-(2-fluorophenyl)propanoate Chemical compound C(C)(C)(C)OC(\C=C\C1=CC(=C(C=C1)OC(CCC1=C(C=CC=C1)F)=O)CC=C(C)C)=O OPHXUUMKVSYQLI-FOWTUZBSSA-N 0.000 description 2
- SCJSPBMLKJVKOK-GHRIWEEISA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] 3-(2-methoxyphenyl)propanoate Chemical compound C(C)(C)(C)OC(\C=C\C1=CC(=C(C=C1)OC(CCC1=C(C=CC=C1)OC)=O)CC=C(C)C)=O SCJSPBMLKJVKOK-GHRIWEEISA-N 0.000 description 2
- BYRNJPLWZLTZRS-FOWTUZBSSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] 3-(3-fluorophenyl)propanoate Chemical compound FC=1C=C(C=CC=1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)CC=C(C)C BYRNJPLWZLTZRS-FOWTUZBSSA-N 0.000 description 2
- CQDXZHBIJPWUJL-LDADJPATSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] 3-(4-methoxyphenyl)propanoate Chemical compound C(C)(C)(C)OC(\C=C\C1=CC(=C(C=C1)OC(CCC1=CC=C(C=C1)OC)=O)CC=C(C)C)=O CQDXZHBIJPWUJL-LDADJPATSA-N 0.000 description 2
- ITWFVTHMAJQTDF-NBVRZTHBSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] 3-phenylpropanoate Chemical compound C(C)(C)(C)OC(\C=C\C1=CC(=C(C=C1)OC(CCC1=CC=CC=C1)=O)CC=C(C)C)=O ITWFVTHMAJQTDF-NBVRZTHBSA-N 0.000 description 2
- NWOAXXJHEFHFSQ-DTQAZKPQSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] benzoate Chemical compound C(C1=CC=CC=C1)(=O)OC1=C(C=C(C=C1)\C=C\C(=O)OC(C)(C)C)CC=C(C)C NWOAXXJHEFHFSQ-DTQAZKPQSA-N 0.000 description 2
- AKXRXUAXXKFJLQ-DTQAZKPQSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] hexanoate Chemical compound C(CCCCC)(=O)OC1=C(C=C(C=C1)\C=C\C(=O)OC(C)(C)C)CC=C(C)C AKXRXUAXXKFJLQ-DTQAZKPQSA-N 0.000 description 2
- MGTGVHNDHUZGLD-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 2-cyclohexylacetate Chemical compound C1(CCCCC1)CC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C MGTGVHNDHUZGLD-UHFFFAOYSA-N 0.000 description 2
- PVZPETLPADLQHA-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 2-naphthalen-1-ylacetate Chemical compound C1(=CC=CC2=CC=CC=C12)CC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C PVZPETLPADLQHA-UHFFFAOYSA-N 0.000 description 2
- OKHRRDHRUFDBFS-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 3-(2-fluorophenyl)propanoate Chemical compound FC1=C(C=CC=C1)CCC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C OKHRRDHRUFDBFS-UHFFFAOYSA-N 0.000 description 2
- WZOKZZWNIFIOFR-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 3-(2-methoxyphenyl)propanoate Chemical compound COC1=C(C=CC=C1)CCC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C WZOKZZWNIFIOFR-UHFFFAOYSA-N 0.000 description 2
- ANONOWHEYLETQW-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 3-(3-fluorophenyl)propanoate Chemical compound FC=1C=C(C=CC=1)CCC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C ANONOWHEYLETQW-UHFFFAOYSA-N 0.000 description 2
- ASKORRXBOWWNBL-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 3-(4-methoxyphenyl)propanoate Chemical compound COC1=CC=C(C=C1)CCC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C ASKORRXBOWWNBL-UHFFFAOYSA-N 0.000 description 2
- FNOBCDSEJFZFPA-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] acetate Chemical compound CC(C)=CCC1=CC(I)=CC=C1OC(C)=O FNOBCDSEJFZFPA-UHFFFAOYSA-N 0.000 description 2
- FTSYEEBTTHYPHJ-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] benzoate Chemical compound C(C1=CC=CC=C1)(=O)OC1=C(C=C(C=C1)I)CC=C(C)C FTSYEEBTTHYPHJ-UHFFFAOYSA-N 0.000 description 2
- QYNLWTZJVGVONY-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] hexanoate Chemical compound C(CCCCC)(=O)OC1=C(C=C(C=C1)I)CC=C(C)C QYNLWTZJVGVONY-UHFFFAOYSA-N 0.000 description 2
- UVIQSJCZCSLXRZ-UBUQANBQSA-N abiraterone acetate Chemical compound C([C@@H]1[C@]2(C)CC[C@@H]3[C@@]4(C)CC[C@@H](CC4=CC[C@H]31)OC(=O)C)C=C2C1=CC=CN=C1 UVIQSJCZCSLXRZ-UBUQANBQSA-N 0.000 description 2
- 229960004103 abiraterone acetate Drugs 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 230000006978 adaptation Effects 0.000 description 2
- 210000004100 adrenal gland Anatomy 0.000 description 2
- 238000013019 agitation Methods 0.000 description 2
- 239000000556 agonist Substances 0.000 description 2
- 150000001299 aldehydes Chemical class 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 229940061641 androsterone Drugs 0.000 description 2
- QGXBDMJGAMFCBF-HLUDHZFRSA-N androsterone group Chemical group [C@@H]12CCC(=O)[C@@]1(C)CC[C@H]1[C@H]2CC[C@H]2C[C@H](O)CC[C@]12C QGXBDMJGAMFCBF-HLUDHZFRSA-N 0.000 description 2
- 230000008485 antagonism Effects 0.000 description 2
- 230000002280 anti-androgenic effect Effects 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 239000000051 antiandrogen Substances 0.000 description 2
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 description 2
- 238000003782 apoptosis assay Methods 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 229960000997 bicalutamide Drugs 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 230000001851 biosynthetic effect Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 150000001642 boronic acid derivatives Chemical class 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- CUWODFFVMXJOKD-UVLQAERKSA-N buserelin Chemical compound CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](COC(C)(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 CUWODFFVMXJOKD-UVLQAERKSA-N 0.000 description 2
- 229960002719 buserelin Drugs 0.000 description 2
- 230000009702 cancer cell proliferation Effects 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 230000003197 catalytic effect Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 239000013043 chemical agent Substances 0.000 description 2
- 229940044683 chemotherapy drug Drugs 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 229930016911 cinnamic acid Natural products 0.000 description 2
- 235000013985 cinnamic acid Nutrition 0.000 description 2
- 150000001851 cinnamic acid derivatives Chemical class 0.000 description 2
- 238000001360 collision-induced dissociation Methods 0.000 description 2
- 210000001072 colon Anatomy 0.000 description 2
- 229940000425 combination drug Drugs 0.000 description 2
- 230000006957 competitive inhibition Effects 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 239000012043 crude product Substances 0.000 description 2
- 239000002254 cytotoxic agent Substances 0.000 description 2
- 238000010511 deprotection reaction Methods 0.000 description 2
- 238000007865 diluting Methods 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 230000003828 downregulation Effects 0.000 description 2
- 238000007876 drug discovery Methods 0.000 description 2
- 239000003480 eluent Substances 0.000 description 2
- 230000032050 esterification Effects 0.000 description 2
- 238000005886 esterification reaction Methods 0.000 description 2
- 150000002170 ethers Chemical class 0.000 description 2
- 238000009093 first-line therapy Methods 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 229910052731 fluorine Inorganic materials 0.000 description 2
- 229960002913 goserelin Drugs 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 230000003054 hormonal effect Effects 0.000 description 2
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000011221 initial treatment Methods 0.000 description 2
- 150000002500 ions Chemical class 0.000 description 2
- 150000002576 ketones Chemical class 0.000 description 2
- 150000002611 lead compounds Chemical class 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- KKSDGJDHHZEWEP-UHFFFAOYSA-N m-hydroxycinnamic acid Natural products OC(=O)C=CC1=CC=CC(O)=C1 KKSDGJDHHZEWEP-UHFFFAOYSA-N 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 230000001394 metastastic effect Effects 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- PKALKWFZXXGNJD-AATRIKPKSA-N methyl (2E)-3-(3-hydroxyphenyl)acrylate Chemical compound COC(=O)\C=C\C1=CC=CC(O)=C1 PKALKWFZXXGNJD-AATRIKPKSA-N 0.000 description 2
- WBYWAXJHAXSJNI-UHFFFAOYSA-N methyl p-hydroxycinnamate Natural products OC(=O)C=CC1=CC=CC=C1 WBYWAXJHAXSJNI-UHFFFAOYSA-N 0.000 description 2
- 230000003278 mimic effect Effects 0.000 description 2
- 210000005087 mononuclear cell Anatomy 0.000 description 2
- 239000000041 non-steroidal anti-inflammatory agent Substances 0.000 description 2
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 2
- 231100000956 nontoxicity Toxicity 0.000 description 2
- 230000005937 nuclear translocation Effects 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 238000011474 orchiectomy Methods 0.000 description 2
- 229940094443 oxytocics prostaglandins Drugs 0.000 description 2
- 239000013610 patient sample Substances 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 230000036470 plasma concentration Effects 0.000 description 2
- 229910000160 potassium phosphate Inorganic materials 0.000 description 2
- 235000011009 potassium phosphates Nutrition 0.000 description 2
- 238000004393 prognosis Methods 0.000 description 2
- 210000005267 prostate cell Anatomy 0.000 description 2
- 238000011472 radical prostatectomy Methods 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- 230000007420 reactivation Effects 0.000 description 2
- 230000009257 reactivity Effects 0.000 description 2
- 230000007115 recruitment Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 238000003153 stable transfection Methods 0.000 description 2
- 230000037359 steroid metabolism Effects 0.000 description 2
- 239000011550 stock solution Substances 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 230000009044 synergistic interaction Effects 0.000 description 2
- SJQIVSLLOKRKQO-FNORWQNLSA-N tert-butyl (E)-3-(3-amino-4-bromophenyl)prop-2-enoate Chemical compound NC=1C=C(C=CC=1Br)/C=C/C(=O)OC(C)(C)C SJQIVSLLOKRKQO-FNORWQNLSA-N 0.000 description 2
- LBHBTGVPHQAFAV-SOFGYWHQSA-N tert-butyl (E)-3-(4-acetamido-3-bromophenyl)prop-2-enoate Chemical compound BrC=1C=C(C=CC=1NC(C)=O)/C=C/C(=O)OC(C)(C)C LBHBTGVPHQAFAV-SOFGYWHQSA-N 0.000 description 2
- UFRBVGPTZVUBJZ-FNORWQNLSA-N tert-butyl (E)-3-(4-amino-3-bromophenyl)prop-2-enoate Chemical compound NC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)Br UFRBVGPTZVUBJZ-FNORWQNLSA-N 0.000 description 2
- KJIRHMCMRFWHNQ-NBVRZTHBSA-N tert-butyl (E)-3-[3-(3-methylbut-2-enyl)-4-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound CC(=CCC=1C=C(C=CC=1NC(CCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C)C KJIRHMCMRFWHNQ-NBVRZTHBSA-N 0.000 description 2
- VNWDIGVHVVAXFT-KNTRCKAVSA-N tert-butyl (E)-3-[3-(3-methylbut-2-enyl)-4-(4-phenylbutanoylamino)phenyl]prop-2-enoate Chemical compound CC(=CCC=1C=C(C=CC=1NC(CCCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C)C VNWDIGVHVVAXFT-KNTRCKAVSA-N 0.000 description 2
- CADVQYKMXRRJIK-OBGWFSINSA-N tert-butyl (E)-3-[3-(3-methylbut-2-enyl)-4-[(2-naphthalen-1-ylacetyl)amino]phenyl]prop-2-enoate Chemical compound CC(=CCC=1C=C(C=CC=1NC(CC1=CC=CC2=CC=CC=C12)=O)/C=C/C(=O)OC(C)(C)C)C CADVQYKMXRRJIK-OBGWFSINSA-N 0.000 description 2
- TVAGFFVMOAZPCJ-JQIJEIRASA-N tert-butyl (E)-3-[3-(3-methylbut-2-enyl)-5-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound CC(=CCC=1C=C(C=C(C=1)NC(CCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C)C TVAGFFVMOAZPCJ-JQIJEIRASA-N 0.000 description 2
- YSPHGSDBULOCAU-SFQUDFHCSA-N tert-butyl (E)-3-[3-[3-(2-fluorophenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound FC1=C(C=CC=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C YSPHGSDBULOCAU-SFQUDFHCSA-N 0.000 description 2
- CPXBMFVHSDIITL-FOWTUZBSSA-N tert-butyl (E)-3-[3-[3-(3-fluorophenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound FC=1C=C(C=CC=1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C CPXBMFVHSDIITL-FOWTUZBSSA-N 0.000 description 2
- KPZKUYBEHLIWEU-GZTJUZNOSA-N tert-butyl (E)-3-[3-[3-(4-fluorophenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound FC1=CC=C(C=C1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C KPZKUYBEHLIWEU-GZTJUZNOSA-N 0.000 description 2
- JBDSHFURYXOJMB-SDNWHVSQSA-N tert-butyl (E)-3-[3-bromo-4-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound BrC=1C=C(C=CC=1NC(CCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C JBDSHFURYXOJMB-SDNWHVSQSA-N 0.000 description 2
- WWHYPYAOGLTWTL-ZRDIBKRKSA-N tert-butyl (E)-3-[3-bromo-4-[(2-cyclohexylacetyl)amino]phenyl]prop-2-enoate Chemical compound BrC=1C=C(C=CC=1NC(CC1CCCCC1)=O)/C=C/C(=O)OC(C)(C)C WWHYPYAOGLTWTL-ZRDIBKRKSA-N 0.000 description 2
- UMEOHCMABDZPPD-WYMLVPIESA-N tert-butyl (E)-3-[3-bromo-4-[(2-naphthalen-1-ylacetyl)amino]phenyl]prop-2-enoate Chemical compound BrC=1C=C(C=CC=1NC(CC1=CC=CC2=CC=CC=C12)=O)/C=C/C(=O)OC(C)(C)C UMEOHCMABDZPPD-WYMLVPIESA-N 0.000 description 2
- PPIUBLURXNELDP-ZRDIBKRKSA-N tert-butyl (E)-3-[3-bromo-5-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound BrC=1C=C(C=C(C=1)NC(CCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C PPIUBLURXNELDP-ZRDIBKRKSA-N 0.000 description 2
- OCZIYOUFLPQPBS-DTQAZKPQSA-N tert-butyl (E)-3-[4-(2-cyclohexylacetyl)oxy-3-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound C1(CCCCC1)CC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)CC=C(C)C OCZIYOUFLPQPBS-DTQAZKPQSA-N 0.000 description 2
- VOWPHPJDIMOOIM-DICWZGCASA-N tert-butyl (E)-3-[4-(3-methylbut-2-enyl)-3-(3-phenylprop-2-enoylamino)phenyl]prop-2-enoate Chemical compound C(C=CC1=CC=CC=C1)(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C VOWPHPJDIMOOIM-DICWZGCASA-N 0.000 description 2
- QIFHUQHTIWDYSI-NBVRZTHBSA-N tert-butyl (E)-3-[4-(3-methylbut-2-enyl)-3-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=CC=C1)=O)C QIFHUQHTIWDYSI-NBVRZTHBSA-N 0.000 description 2
- ZVFGIIMXFQFHSM-DTQAZKPQSA-N tert-butyl (E)-3-[4-[(2-cyclohexylacetyl)amino]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound C1(CCCCC1)CC(=O)NC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)CC=C(C)C ZVFGIIMXFQFHSM-DTQAZKPQSA-N 0.000 description 2
- NCZNWFGWRWVXGG-FMIVXFBMSA-N tert-butyl (E)-3-[4-acetamido-3-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound C(C)(=O)NC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)CC=C(C)C NCZNWFGWRWVXGG-FMIVXFBMSA-N 0.000 description 2
- NXVSBRCKNPYGCC-FMIVXFBMSA-N tert-butyl (E)-3-[4-acetyloxy-3-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound C(C)(=O)OC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)CC=C(C)C NXVSBRCKNPYGCC-FMIVXFBMSA-N 0.000 description 2
- LSQVODBVPAWZPW-LMJSHCNKSA-N tert-butyl (E)-3-[4-bromo-3-(3-phenylprop-2-enoylamino)phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(C=CC1=CC=CC=C1)=O LSQVODBVPAWZPW-LMJSHCNKSA-N 0.000 description 2
- XTFOTGXVQZIAOS-SDNWHVSQSA-N tert-butyl (E)-3-[4-bromo-3-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=CC=C1)=O XTFOTGXVQZIAOS-SDNWHVSQSA-N 0.000 description 2
- JDFPBWVSVSJCIS-UKTHLTGXSA-N tert-butyl (E)-3-[4-bromo-3-[3-(2-fluorophenyl)propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=C(C=CC=C1)F)=O JDFPBWVSVSJCIS-UKTHLTGXSA-N 0.000 description 2
- MRZHYBGECUEXAS-GXDHUFHOSA-N tert-butyl (E)-3-[4-bromo-3-[3-(2-methoxyphenyl)propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=C(C=CC=C1)OC)=O MRZHYBGECUEXAS-GXDHUFHOSA-N 0.000 description 2
- GNQVWVBNTOIJDT-FMIVXFBMSA-N tert-butyl (E)-3-[4-bromo-3-[3-(3-fluorophenyl)propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC(=CC=C1)F)=O GNQVWVBNTOIJDT-FMIVXFBMSA-N 0.000 description 2
- MVPGEUHDLQJHPH-MDWZMJQESA-N tert-butyl (E)-3-[4-bromo-3-[3-(4-fluorophenyl)propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=C(C=C1)F)=O MVPGEUHDLQJHPH-MDWZMJQESA-N 0.000 description 2
- PGWVIQWMBAZMDQ-NTEUORMPSA-N tert-butyl (E)-3-[4-bromo-3-[3-(4-methoxyphenyl)propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=C(C=C1)OC)=O PGWVIQWMBAZMDQ-NTEUORMPSA-N 0.000 description 2
- SKPSRWCSGSMRCG-SFQUDFHCSA-N tert-butyl (E)-3-[4-bromo-3-[3-[3-(4-methylphenyl)sulfonyloxyphenyl]propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC(=CC=C1)OS(=O)(=O)C1=CC=C(C)C=C1)=O SKPSRWCSGSMRCG-SFQUDFHCSA-N 0.000 description 2
- MKDLKIJBUMIKOC-GXDHUFHOSA-N tert-butyl (E)-3-[5-bromo-2-(3-phenylpropanoylamino)phenyl]prop-2-enoate Chemical compound BrC=1C=CC(=C(C=1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=CC=C1)=O MKDLKIJBUMIKOC-GXDHUFHOSA-N 0.000 description 2
- 125000002088 tosyl group Chemical group [H]C1=C([H])C(=C([H])C([H])=C1C([H])([H])[H])S(*)(=O)=O 0.000 description 2
- 230000007704 transition Effects 0.000 description 2
- 230000003827 upregulation Effects 0.000 description 2
- 238000011144 upstream manufacturing Methods 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 2
- 229920002554 vinyl polymer Polymers 0.000 description 2
- PJVWKTKQMONHTI-UHFFFAOYSA-N warfarin Chemical compound OC=1C2=CC=CC=C2OC(=O)C=1C(CC(=O)C)C1=CC=CC=C1 PJVWKTKQMONHTI-UHFFFAOYSA-N 0.000 description 2
- 229960005080 warfarin Drugs 0.000 description 2
- 239000012130 whole-cell lysate Substances 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- LAJAFFLJAJMYLK-CVOKMOJFSA-N (1s,2s,3r,4r)-3-[[5-chloro-2-[[(7s)-4-methoxy-7-morpholin-4-yl-6,7,8,9-tetrahydro-5h-benzo[7]annulen-3-yl]amino]pyrimidin-4-yl]amino]bicyclo[2.2.1]hept-5-ene-2-carboxamide Chemical compound N1([C@H]2CCC3=CC=C(C(=C3CC2)OC)NC=2N=C(C(=CN=2)Cl)N[C@H]2[C@H]([C@@]3([H])C[C@@]2(C=C3)[H])C(N)=O)CCOCC1 LAJAFFLJAJMYLK-CVOKMOJFSA-N 0.000 description 1
- MPDDTAJMJCESGV-CTUHWIOQSA-M (3r,5r)-7-[2-(4-fluorophenyl)-5-[methyl-[(1r)-1-phenylethyl]carbamoyl]-4-propan-2-ylpyrazol-3-yl]-3,5-dihydroxyheptanoate Chemical compound C1([C@@H](C)N(C)C(=O)C2=NN(C(CC[C@@H](O)C[C@@H](O)CC([O-])=O)=C2C(C)C)C=2C=CC(F)=CC=2)=CC=CC=C1 MPDDTAJMJCESGV-CTUHWIOQSA-M 0.000 description 1
- BSGIKRCNMBTGEB-UHFFFAOYSA-N (5-bromo-1-methylpyrrolo[2,3-c]pyridin-2-yl)-[6-[(dimethylamino)methyl]-4-methyl-3,4-dihydro-1h-isoquinolin-2-yl]methanone Chemical compound BrC1=NC=C2N(C)C(C(=O)N3CC(C4=CC(CN(C)C)=CC=C4C3)C)=CC2=C1 BSGIKRCNMBTGEB-UHFFFAOYSA-N 0.000 description 1
- JHLIGYPHPBLDDL-UHFFFAOYSA-N (5-pyridin-3-ylthiophen-2-yl)methanamine Chemical compound S1C(CN)=CC=C1C1=CC=CN=C1 JHLIGYPHPBLDDL-UHFFFAOYSA-N 0.000 description 1
- OLWVYKFXHCETPJ-VMPITWQZSA-N (E)-3-[3-(3-methylbut-2-enyl)-4-(pyridine-4-carbonyloxy)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=CC=1OC(=O)C1=CC=NC=C1)/C=C/C(=O)O)C OLWVYKFXHCETPJ-VMPITWQZSA-N 0.000 description 1
- KXRLYTUEAMHMJD-ZHACJKMWSA-N (E)-3-[3-(3-methylbut-2-enyl)-5-(2-phenylethoxycarbonyl)phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=C(C=1)C(=O)OCCC1=CC=CC=C1)/C=C/C(=O)O)C KXRLYTUEAMHMJD-ZHACJKMWSA-N 0.000 description 1
- LHORRWNZWXDXQU-JXMROGBWSA-N (E)-3-[3-(3-methylbut-2-enyl)-5-[[4-(trifluoromethoxy)phenyl]methylcarbamoyl]phenyl]prop-2-enoic acid Chemical compound CC(=CCC=1C=C(C=C(C=1)C(NCC1=CC=C(C=C1)OC(F)(F)F)=O)/C=C/C(=O)O)C LHORRWNZWXDXQU-JXMROGBWSA-N 0.000 description 1
- PTODWLIUCPDCTN-DHZHZOJOSA-N (E)-3-[3-[(4-methoxyphenyl)methylcarbamoyl]-5-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound COC1=CC=C(CNC(=O)C=2C=C(C=C(C=2)CC=C(C)C)/C=C/C(=O)O)C=C1 PTODWLIUCPDCTN-DHZHZOJOSA-N 0.000 description 1
- GNHOJNHOYDTILE-YBFXNURJSA-N (E)-3-[4-(3-methylbut-2-enyl)-3-[3-[4-(4-methylphenyl)sulfonyloxyphenyl]propanoylamino]phenyl]prop-2-enoic acid Chemical compound CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)O)NC(CCC1=CC=C(C=C1)OS(=O)(=O)C1=CC=C(C)C=C1)=O)C GNHOJNHOYDTILE-YBFXNURJSA-N 0.000 description 1
- DFEDSIFLBPLNGY-UKTHLTGXSA-N (E)-3-[4-[3-(3-methoxyphenyl)propanoyloxy]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound COC=1C=C(C=CC=1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C DFEDSIFLBPLNGY-UKTHLTGXSA-N 0.000 description 1
- KPEUOETXWJNVJX-NTUHNPAUSA-N (E)-3-[4-[3-(4-fluorophenyl)propanoyloxy]-3-(3-methylbut-2-enyl)phenyl]prop-2-enoic acid Chemical compound FC1=CC=C(C=C1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)O)CC=C(C)C KPEUOETXWJNVJX-NTUHNPAUSA-N 0.000 description 1
- AZWLJKVQAKQPCK-JXMROGBWSA-N (E)-3-[4-bromo-3-[3-(3-methoxyphenyl)propanoylamino]phenyl]prop-2-enoic acid Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)O)NC(CCC1=CC(=CC=C1)OC)=O AZWLJKVQAKQPCK-JXMROGBWSA-N 0.000 description 1
- PXGPLTODNUVGFL-BRIYLRKRSA-N (E,Z)-(1R,2R,3R,5S)-7-(3,5-Dihydroxy-2-((3S)-(3-hydroxy-1-octenyl))cyclopentyl)-5-heptenoic acid Chemical compound CCCCC[C@H](O)C=C[C@H]1[C@H](O)C[C@H](O)[C@@H]1CC=CCCCC(O)=O PXGPLTODNUVGFL-BRIYLRKRSA-N 0.000 description 1
- WOGITNXCNOTRLK-VOTSOKGWSA-N (e)-3-phenylprop-2-enoyl chloride Chemical compound ClC(=O)\C=C\C1=CC=CC=C1 WOGITNXCNOTRLK-VOTSOKGWSA-N 0.000 description 1
- VICOOSNNZUPVHM-IGPZRPDBSA-M (e,3r,5s)-7-[2-(4-fluorophenyl)-4-(3-phenylpentan-3-yl)phenyl]-3,5-dihydroxyhept-6-enoate Chemical compound C=1C=C(\C=C\[C@@H](O)C[C@@H](O)CC([O-])=O)C(C=2C=CC(F)=CC=2)=CC=1C(CC)(CC)C1=CC=CC=C1 VICOOSNNZUPVHM-IGPZRPDBSA-M 0.000 description 1
- ZKSOJQDNSNJIQW-UHFFFAOYSA-N 1-(bromomethyl)-3-methoxybenzene Chemical compound COC1=CC=CC(CBr)=C1 ZKSOJQDNSNJIQW-UHFFFAOYSA-N 0.000 description 1
- WGABOZPQOOZAOI-UHFFFAOYSA-N 2-[4-[[(3,5-dimethoxy-4-methylbenzoyl)-(3-phenylpropyl)amino]methyl]phenyl]acetic acid Chemical compound COC1=C(C)C(OC)=CC(C(=O)N(CCCC=2C=CC=CC=2)CC=2C=CC(CC(O)=O)=CC=2)=C1 WGABOZPQOOZAOI-UHFFFAOYSA-N 0.000 description 1
- TVTJUIAKQFIXCE-HUKYDQBMSA-N 2-amino-9-[(2R,3S,4S,5R)-4-fluoro-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-7-prop-2-ynyl-1H-purine-6,8-dione Chemical compound NC=1NC(C=2N(C(N(C=2N=1)[C@@H]1O[C@@H]([C@H]([C@H]1O)F)CO)=O)CC#C)=O TVTJUIAKQFIXCE-HUKYDQBMSA-N 0.000 description 1
- BMIBJCFFZPYJHF-UHFFFAOYSA-N 2-methoxy-5-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine Chemical compound COC1=NC=C(C)C=C1B1OC(C)(C)C(C)(C)O1 BMIBJCFFZPYJHF-UHFFFAOYSA-N 0.000 description 1
- KAGDRELXVLNWDO-UHFFFAOYSA-N 3-(4-fluorophenyl)propanoyl chloride Chemical compound FC1=CC=C(CCC(Cl)=O)C=C1 KAGDRELXVLNWDO-UHFFFAOYSA-N 0.000 description 1
- UMCMPZBLKLEWAF-BCTGSCMUSA-N 3-[(3-cholamidopropyl)dimethylammonio]propane-1-sulfonate Chemical compound C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(=O)NCCC[N+](C)(C)CCCS([O-])(=O)=O)C)[C@@]2(C)[C@@H](O)C1 UMCMPZBLKLEWAF-BCTGSCMUSA-N 0.000 description 1
- CURASYMMNYLBQN-UHFFFAOYSA-N 3-bromo-5-iodobenzoyl chloride Chemical compound ClC(=O)C1=CC(Br)=CC(I)=C1 CURASYMMNYLBQN-UHFFFAOYSA-N 0.000 description 1
- XMIIGOLPHOKFCH-UHFFFAOYSA-M 3-phenylpropionate Chemical compound [O-]C(=O)CCC1=CC=CC=C1 XMIIGOLPHOKFCH-UHFFFAOYSA-M 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- LZPWAYBEOJRFAX-UHFFFAOYSA-N 4,4,5,5-tetramethyl-1,3,2$l^{2}-dioxaborolane Chemical class CC1(C)O[B]OC1(C)C LZPWAYBEOJRFAX-UHFFFAOYSA-N 0.000 description 1
- APRZHQXAAWPYHS-UHFFFAOYSA-N 4-[5-[3-(carboxymethoxy)phenyl]-3-(4,5-dimethyl-1,3-thiazol-2-yl)tetrazol-3-ium-2-yl]benzenesulfonate Chemical compound S1C(C)=C(C)N=C1[N+]1=NC(C=2C=C(OCC(O)=O)C=CC=2)=NN1C1=CC=C(S([O-])(=O)=O)C=C1 APRZHQXAAWPYHS-UHFFFAOYSA-N 0.000 description 1
- VQDQISMDUHBUFF-UHFFFAOYSA-N 4-phenylbutanoyl chloride Chemical compound ClC(=O)CCCC1=CC=CC=C1 VQDQISMDUHBUFF-UHFFFAOYSA-N 0.000 description 1
- 101150005096 AKR1 gene Proteins 0.000 description 1
- 102100024090 Aldo-keto reductase family 1 member C3 Human genes 0.000 description 1
- 102100024092 Aldo-keto reductase family 1 member C4 Human genes 0.000 description 1
- 241000256844 Apis mellifera Species 0.000 description 1
- 108010039627 Aprotinin Proteins 0.000 description 1
- OYVQAFCQQOHOJZ-HYARGMPZSA-N CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=C(C=C1)OS(=O)(=O)C1=CC=C(C)C=C1)=O)C Chemical compound CC(=CCC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=C(C=C1)OS(=O)(=O)C1=CC=C(C)C=C1)=O)C OYVQAFCQQOHOJZ-HYARGMPZSA-N 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 1
- 102000003952 Caspase 3 Human genes 0.000 description 1
- 108090000397 Caspase 3 Proteins 0.000 description 1
- 238000006969 Curtius rearrangement reaction Methods 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- YCKRFDGAMUMZLT-UHFFFAOYSA-N Fluorine atom Chemical compound [F] YCKRFDGAMUMZLT-UHFFFAOYSA-N 0.000 description 1
- 101000690301 Homo sapiens Aldo-keto reductase family 1 member C4 Proteins 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 208000037848 Metastatic bone disease Diseases 0.000 description 1
- 101100347578 Mus musculus Mb gene Proteins 0.000 description 1
- 238000011789 NOD SCID mouse Methods 0.000 description 1
- 206010061309 Neoplasm progression Diseases 0.000 description 1
- 101100215778 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) ptr-1 gene Proteins 0.000 description 1
- DFPAKSUCGFBDDF-UHFFFAOYSA-N Nicotinamide Chemical group NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- BELBBZDIHDAJOR-UHFFFAOYSA-N Phenolsulfonephthalein Chemical compound C1=CC(O)=CC=C1C1(C=2C=CC(O)=CC=2)C2=CC=CC=C2S(=O)(=O)O1 BELBBZDIHDAJOR-UHFFFAOYSA-N 0.000 description 1
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 1
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 1
- 229920002565 Polyethylene Glycol 400 Polymers 0.000 description 1
- 102000000471 Prostaglandin F receptors Human genes 0.000 description 1
- 108050008995 Prostaglandin F receptors Proteins 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 239000012083 RIPA buffer Substances 0.000 description 1
- 108091027967 Small hairpin RNA Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- OYNKEWVJRBGLTF-GZTJUZNOSA-N [2-(3-methylbut-2-enyl)-4-[(E)-3-[(2-methylpropan-2-yl)oxy]-3-oxoprop-1-enyl]phenyl] 3-(4-fluorophenyl)propanoate Chemical compound FC1=CC=C(C=C1)CCC(=O)OC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)CC=C(C)C OYNKEWVJRBGLTF-GZTJUZNOSA-N 0.000 description 1
- DNPPKOVPMIEIDH-UHFFFAOYSA-N [3-(3-chloro-3-oxopropyl)phenyl] 4-methylbenzenesulfonate Chemical compound C1=CC(C)=CC=C1S(=O)(=O)OC1=CC=CC(CCC(Cl)=O)=C1 DNPPKOVPMIEIDH-UHFFFAOYSA-N 0.000 description 1
- PDDKFCDCAGDBFO-UHFFFAOYSA-N [4-(3-chloro-3-oxopropyl)phenyl] 4-methylbenzenesulfonate Chemical compound CC1=CC=C(C=C1)S(=O)(=O)OC1=CC=C(C=C1)CCC(=O)Cl PDDKFCDCAGDBFO-UHFFFAOYSA-N 0.000 description 1
- ABIDLSVVZHHWPH-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 3-(3-methoxyphenyl)propanoate Chemical compound COC=1C=C(C=CC=1)CCC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C ABIDLSVVZHHWPH-UHFFFAOYSA-N 0.000 description 1
- BYAPUAXYCMCQLI-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] 3-(4-fluorophenyl)propanoate Chemical compound FC1=CC=C(C=C1)CCC(=O)OC1=C(C=C(C=C1)I)CC=C(C)C BYAPUAXYCMCQLI-UHFFFAOYSA-N 0.000 description 1
- ZWYCWUJCTHAZRQ-UHFFFAOYSA-N [4-iodo-2-(3-methylbut-2-enyl)phenyl] pyridine-4-carboxylate Chemical compound N1=CC=C(C=C1)C(=O)OC1=C(C=C(C=C1)I)CC=C(C)C ZWYCWUJCTHAZRQ-UHFFFAOYSA-N 0.000 description 1
- 229960000583 acetic acid Drugs 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 125000000218 acetic acid group Chemical group C(C)(=O)* 0.000 description 1
- WETWJCDKMRHUPV-UHFFFAOYSA-N acetyl chloride Chemical compound CC(Cl)=O WETWJCDKMRHUPV-UHFFFAOYSA-N 0.000 description 1
- 239000012346 acetyl chloride Substances 0.000 description 1
- 125000003668 acetyloxy group Chemical group [H]C([H])([H])C(=O)O[*] 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 229960003473 androstanolone Drugs 0.000 description 1
- AEMFNILZOJDQLW-UHFFFAOYSA-N androstenedione Natural products O=C1CCC2(C)C3CCC(C)(C(CC4)=O)C4C3CCC2=C1 AEMFNILZOJDQLW-UHFFFAOYSA-N 0.000 description 1
- 239000003817 anthracycline antibiotic agent Substances 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 229960004405 aprotinin Drugs 0.000 description 1
- 239000012223 aqueous fraction Substances 0.000 description 1
- 239000008346 aqueous phase Substances 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 150000001503 aryl iodides Chemical class 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- PASDCCFISLVPSO-UHFFFAOYSA-N benzoyl chloride Chemical compound ClC(=O)C1=CC=CC=C1 PASDCCFISLVPSO-UHFFFAOYSA-N 0.000 description 1
- 125000001231 benzoyloxy group Chemical group C(C1=CC=CC=C1)(=O)O* 0.000 description 1
- AGEZXYOZHKGVCM-UHFFFAOYSA-N benzyl bromide Chemical compound BrCC1=CC=CC=C1 AGEZXYOZHKGVCM-UHFFFAOYSA-N 0.000 description 1
- AJRQIXBBIDPNGK-BVSLBCMMSA-N benzyl n-[(2s)-1-[[(2s)-1-(1,3-benzothiazol-2-yl)-1-oxo-3-[(3s)-2-oxopyrrolidin-3-yl]propan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]carbamate Chemical compound N([C@@H](CC(C)C)C(=O)N[C@@H](C[C@H]1C(NCC1)=O)C(=O)C=1SC2=CC=CC=C2N=1)C(=O)OCC1=CC=CC=C1 AJRQIXBBIDPNGK-BVSLBCMMSA-N 0.000 description 1
- AFYNADDZULBEJA-UHFFFAOYSA-N bicinchoninic acid Chemical compound C1=CC=CC2=NC(C=3C=C(C4=CC=CC=C4N=3)C(=O)O)=CC(C(O)=O)=C21 AFYNADDZULBEJA-UHFFFAOYSA-N 0.000 description 1
- 238000012925 biological evaluation Methods 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 239000005388 borosilicate glass Substances 0.000 description 1
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 125000002843 carboxylic acid group Chemical group 0.000 description 1
- 150000001735 carboxylic acids Chemical class 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000003570 cell viability assay Methods 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- BJDCWCLMFKKGEE-CMDXXVQNSA-N chembl252518 Chemical compound C([C@@](OO1)(C)O2)C[C@H]3[C@H](C)CC[C@@H]4[C@@]31[C@@H]2O[C@H](O)[C@@H]4C BJDCWCLMFKKGEE-CMDXXVQNSA-N 0.000 description 1
- 229940121657 clinical drug Drugs 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 229940126212 compound 17a Drugs 0.000 description 1
- 229940125851 compound 27 Drugs 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 238000005100 correlation spectroscopy Methods 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 230000007123 defense Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 238000006073 displacement reaction Methods 0.000 description 1
- 238000002518 distortionless enhancement with polarization transfer Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000007750 drug combination effect Effects 0.000 description 1
- 238000012912 drug discovery process Methods 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 239000002532 enzyme inhibitor Substances 0.000 description 1
- 125000001033 ether group Chemical group 0.000 description 1
- 125000000816 ethylene group Chemical group [H]C([H])([*:1])C([H])([H])[*:2] 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 229930003944 flavone Natural products 0.000 description 1
- 150000002213 flavones Chemical class 0.000 description 1
- 235000011949 flavones Nutrition 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 239000011737 fluorine Substances 0.000 description 1
- 125000001153 fluoro group Chemical group F* 0.000 description 1
- PTCGDEVVHUXTMP-UHFFFAOYSA-N flutolanil Chemical compound CC(C)OC1=CC=CC(NC(=O)C=2C(=CC=CC=2)C(F)(F)F)=C1 PTCGDEVVHUXTMP-UHFFFAOYSA-N 0.000 description 1
- 239000012520 frozen sample Substances 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000012362 glacial acetic acid Substances 0.000 description 1
- 238000003929 heteronuclear multiple quantum coherence Methods 0.000 description 1
- YWGHUJQYGPDNKT-UHFFFAOYSA-N hexanoyl chloride Chemical compound CCCCCC(Cl)=O YWGHUJQYGPDNKT-UHFFFAOYSA-N 0.000 description 1
- 238000004896 high resolution mass spectrometry Methods 0.000 description 1
- 238000011905 homologation Methods 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- ZPNFWUPYTFPOJU-LPYSRVMUSA-N iniprol Chemical compound C([C@H]1C(=O)NCC(=O)NCC(=O)N[C@H]2CSSC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC=4C=CC=CC=4)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC2=O)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC=2C=CC=CC=2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]2N(CCC2)C(=O)[C@@H](N)CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N2[C@@H](CCC2)C(=O)N2[C@@H](CCC2)C(=O)N[C@@H](CC=2C=CC(O)=CC=2)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N2[C@@H](CCC2)C(=O)N3)C(=O)NCC(=O)NCC(=O)N[C@@H](C)C(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@H](C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H](C(=O)N1)C(C)C)[C@@H](C)O)[C@@H](C)CC)=O)[C@@H](C)CC)C1=CC=C(O)C=C1 ZPNFWUPYTFPOJU-LPYSRVMUSA-N 0.000 description 1
- 238000009434 installation Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000010410 layer Substances 0.000 description 1
- 108020001756 ligand binding domains Proteins 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 238000003468 luciferase reporter gene assay Methods 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 150000004702 methyl esters Chemical class 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 239000002808 molecular sieve Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 230000004660 morphological change Effects 0.000 description 1
- 229940126619 mouse monoclonal antibody Drugs 0.000 description 1
- 210000001167 myeloblast Anatomy 0.000 description 1
- 210000003887 myelocyte Anatomy 0.000 description 1
- BOPGDPNILDQYTO-NNYOXOHSSA-N nicotinamide-adenine dinucleotide Chemical compound C1=CCC(C(=O)N)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C3=NC=NC(N)=C3N=C2)O)O1 BOPGDPNILDQYTO-NNYOXOHSSA-N 0.000 description 1
- 238000010534 nucleophilic substitution reaction Methods 0.000 description 1
- 230000030648 nucleus localization Effects 0.000 description 1
- 239000012074 organic phase Substances 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 201000004228 ovarian endometrial cancer Diseases 0.000 description 1
- 238000012856 packing Methods 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- JLFNLZLINWHATN-UHFFFAOYSA-N pentaethylene glycol Chemical compound OCCOCCOCCOCCOCCO JLFNLZLINWHATN-UHFFFAOYSA-N 0.000 description 1
- 229960003531 phenolsulfonphthalein Drugs 0.000 description 1
- XZCCEQCTCJOBTK-UHFFFAOYSA-N phenyl 4-phenylbutanoate Chemical compound C=1C=CC=CC=1OC(=O)CCCC1=CC=CC=C1 XZCCEQCTCJOBTK-UHFFFAOYSA-N 0.000 description 1
- RGQQGHGIUCRECH-UHFFFAOYSA-N phenyl pyridine-4-carboxylate Chemical compound C=1C=NC=CC=1C(=O)OC1=CC=CC=C1 RGQQGHGIUCRECH-UHFFFAOYSA-N 0.000 description 1
- 229940067107 phenylethyl alcohol Drugs 0.000 description 1
- DHRLEVQXOMLTIM-UHFFFAOYSA-N phosphoric acid;trioxomolybdenum Chemical compound O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.OP(O)(O)=O DHRLEVQXOMLTIM-UHFFFAOYSA-N 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 239000012286 potassium permanganate Substances 0.000 description 1
- 238000012910 preclinical development Methods 0.000 description 1
- 150000003141 primary amines Chemical class 0.000 description 1
- 230000000861 pro-apoptotic effect Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- QGLVEAGMVUQOJP-UHFFFAOYSA-N prop-2-enylboronic acid Chemical compound OB(O)CC=C QGLVEAGMVUQOJP-UHFFFAOYSA-N 0.000 description 1
- 150000003814 prostanoids Chemical class 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- RVQZKNOMKUSGCI-UHFFFAOYSA-N pyridine-4-carbonyl chloride Chemical compound ClC(=O)C1=CC=NC=C1 RVQZKNOMKUSGCI-UHFFFAOYSA-N 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 208000016691 refractory malignant neoplasm Diseases 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 239000012047 saturated solution Substances 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 230000009834 selective interaction Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 239000004055 small Interfering RNA Substances 0.000 description 1
- URGAHOPLAPQHLN-UHFFFAOYSA-N sodium aluminosilicate Chemical compound [Na+].[Al+3].[O-][Si]([O-])=O.[O-][Si]([O-])=O URGAHOPLAPQHLN-UHFFFAOYSA-N 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 238000012430 stability testing Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 230000010009 steroidogenesis Effects 0.000 description 1
- 239000001117 sulphuric acid Substances 0.000 description 1
- 235000011149 sulphuric acid Nutrition 0.000 description 1
- 230000009469 supplementation Effects 0.000 description 1
- 230000001502 supplementing effect Effects 0.000 description 1
- 238000004885 tandem mass spectrometry Methods 0.000 description 1
- RRVSZSWABJNRRF-CCEZHUSRSA-N tert-butyl (E)-3-[3-(2-phenylethylcarbamoyl)-5-phenylmethoxyphenyl]prop-2-enoate Chemical compound C(C1=CC=CC=C1)OC=1C=C(C=C(C=1)C(NCCC1=CC=CC=C1)=O)/C=C/C(=O)OC(C)(C)C RRVSZSWABJNRRF-CCEZHUSRSA-N 0.000 description 1
- OGKHRDJWEZQDBD-GHRIWEEISA-N tert-butyl (E)-3-[3-[3-(3-methoxyphenyl)propanoylamino]-4-(3-methylbut-2-enyl)phenyl]prop-2-enoate Chemical compound COC=1C=C(C=CC=1)CCC(=O)NC=1C=C(C=CC=1CC=C(C)C)/C=C/C(=O)OC(C)(C)C OGKHRDJWEZQDBD-GHRIWEEISA-N 0.000 description 1
- CFRUSJBNMJOJAI-WOJGMQOQSA-N tert-butyl (E)-3-[4-bromo-3-[3-[4-(4-methylphenyl)sulfonyloxyphenyl]propanoylamino]phenyl]prop-2-enoate Chemical compound BrC1=C(C=C(C=C1)/C=C/C(=O)OC(C)(C)C)NC(CCC1=CC=C(C=C1)OS(=O)(=O)C1=CC=C(C)C=C1)=O CFRUSJBNMJOJAI-WOJGMQOQSA-N 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 231100001274 therapeutic index Toxicity 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 description 1
- 230000005751 tumor progression Effects 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/185—Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
- A61K31/19—Carboxylic acids, e.g. valproic acid
- A61K31/192—Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid
-
- 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
- A61K31/21—Esters, e.g. nitroglycerine, selenocyanates
- A61K31/215—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
- A61K31/216—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
-
- 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
- A61K31/21—Esters, e.g. nitroglycerine, selenocyanates
- A61K31/215—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
- A61K31/22—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
- A61K31/222—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin with compounds having aromatic groups, e.g. dipivefrine, ibopamine
-
- 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
- A61K31/21—Esters, e.g. nitroglycerine, selenocyanates
- A61K31/215—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
- A61K31/235—Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
-
- 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
- A61K31/404—Indoles, e.g. pindolol
- A61K31/405—Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
-
- 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
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
- A61K31/4164—1,3-Diazoles
- A61K31/4166—1,3-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. phenytoin
-
- 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
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/4409—Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 4, e.g. isoniazid, iproniazid
-
- 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
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7028—Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
- A61K31/7034—Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
- A61K31/704—Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
-
- 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
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7048—Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
-
- 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
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
- A61K31/7064—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
- A61K31/7068—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
-
- 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
- A61P35/02—Antineoplastic agents specific for leukemia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C233/00—Carboxylic acid amides
- C07C233/01—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
- C07C233/45—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
- C07C233/53—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring
- C07C233/54—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of a saturated carbon skeleton
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C233/00—Carboxylic acid amides
- C07C233/01—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
- C07C233/45—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
- C07C233/53—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring
- C07C233/55—Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring having the carbon atom of the carboxamide group bound to a carbon atom of an unsaturated carbon skeleton
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C235/00—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
- C07C235/02—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
- C07C235/32—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
- C07C235/38—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C235/00—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
- C07C235/70—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton
- C07C235/84—Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C69/00—Esters of carboxylic acids; Esters of carbonic or haloformic acids
- C07C69/608—Esters of carboxylic acids having a carboxyl group bound to an acyclic carbon atom and having a ring other than a six-membered aromatic ring in the acid moiety
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C69/00—Esters of carboxylic acids; Esters of carbonic or haloformic acids
- C07C69/612—Esters of carboxylic acids having a carboxyl group bound to an acyclic carbon atom and having a six-membered aromatic ring in the acid moiety
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C69/00—Esters of carboxylic acids; Esters of carbonic or haloformic acids
- C07C69/612—Esters of carboxylic acids having a carboxyl group bound to an acyclic carbon atom and having a six-membered aromatic ring in the acid moiety
- C07C69/616—Esters of carboxylic acids having a carboxyl group bound to an acyclic carbon atom and having a six-membered aromatic ring in the acid moiety polycyclic
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C69/00—Esters of carboxylic acids; Esters of carbonic or haloformic acids
- C07C69/76—Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
- C07C69/94—Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of polycyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of six-membered aromatic rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D213/00—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
- C07D213/02—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
- C07D213/04—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
- C07D213/60—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D213/78—Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
- C07D213/79—Acids; Esters
-
- 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
- C07F5/00—Compounds containing elements of Groups 3 or 13 of the Periodic Table
- C07F5/02—Boron compounds
- C07F5/025—Boronic and borinic acid compounds
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07C—ACYCLIC OR CARBOCYCLIC COMPOUNDS
- C07C2601/00—Systems containing only non-condensed rings
- C07C2601/12—Systems containing only non-condensed rings with a six-membered ring
- C07C2601/14—The ring being saturated
Definitions
- the present invention relates generally to methods and compositions used for inhibiting the biological activity of an aldo-keto reductase family 1, member C3 (AKR1C3) polypeptide.
- Aldo-keto reductase 1C3 (AKR1C3) specific inhibitors to modulate cellular or tissue proliferation, e.g., prostate cancer and/or castrate-resistant prostate cancer.
- Prostate cancer is the second most common cancer in men, and twenty -percent of all cases develop into castrate- resistant prostate cancer, which often presents with metastatic bone disease and is always fatal. These tumors are synthesized androgens, independently of the testes and there exists a great need in the art for such treatments. All prior AKR1C3 inhibitors have had poor selectivity between AKR1C isozymes resulting in undesirable side effects. As a result no AKR1C3 inhibitor has reached clinical trial, in part, due to lack of selectivity.
- compositions for inhibiting a biological activity of an aldo-keto reductase family 1, member C3 (AKR1C3) polypeptide discloses compositions for inhibiting a biological activity of an aldo-keto reductase family 1, member C3 (AKR1C3) polypeptide.
- the compositions are indomethacin (2-[l-(4-chlorobenzoyl)-5-methoxy-2-methylindol-3-yl]acetic acid) derivatives that are AKR1 C3- specific inhibitors.
- the compositions showed a selective interaction with AKR1C3, they also interacted with AKR1C1 and AKR1C2.
- AKR1C1 and AKR1C2 are involved in hormonal levels and balance and blocking of these affects those levels.
- Aldo-keto reductase 1C3 also known as type 5 17 ⁇ -hydroxy steroid dehydrogenase, is responsible for intratumoral androgen biosynthesis and contributes to the development of castration resistant prostate cancer (CRPC) phenotype and an eventual therapeutic failure.
- Significant upregulation of AKR1C3 is observed in CRPC patient samples and derived cell lines. Being a downstream catalytic enzyme for the synthesis of testosterone and 5-dihydrotestosterone (DHT), AKR1C3 represents a potential therapeutic target to manage CRPC and combat the emergence of resistance to clinically employed first line therapeutics.
- DHT 5-dihydrotestosterone
- the present invention provides novel, potent, isoform selective and hydrolytically stable small-molecule AKR1C3 inhibitor compounds that reduces prostate cancer cell growth in vitro and sensitizes CRPC cell lines towards enzalutamide cytotoxicity.
- the present invention demonstrates that treatment with these inhibitors selectively induce cytotoxicity in a wide variety of cancer cells. Moreover, these inhibitors cause inhibition of AKR1C3 enzymatic activity and consequently reduces androgen receptor (AR) transactivation, AR and prostate specific antigen (PSA) expression levels in CRPC cell lines.
- AR androgen receptor
- PSA prostate specific antigen
- the results demonstrate a novel therapeutic strategy for the treatment of, e.g., prostate cancer, castration-resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
- AML acute myeloid leukemia
- T-ALL T-cell acute lymphoblastic leukemia
- a leukemia e.g., prostate cancer, castration-resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
- AML acute myeloid leukemia
- T-ALL T-cell acute lymphoblastic leukemia
- a leukemia e.g., a castration-resistant prostate cancer disease phenotype, it invariably develops in prostate cancer patients following initial treatment with AR antagonists such as enzalutamide.
- the present invention includes an aldo-keto reductase family 1, member C3 (AKR1C3) inhibitor having one of the following structures:
- the inhibitor is Class I or IA and
- Rl is:
- the inhibitor is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidethyl
- the inhibitor is:
- the inhibitor is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidethyl
- R2 is:
- the inhibitor is:
- R is:
- the inhibitor is:
- R3 is ;
- R2 is:
- the inhibitor is:
- the inhibitor is formulated into a therapeutic effective amount sufficient to reduce or eliminate cancer cells.
- the inhibitor further comprises a chemotherapeutic agent, wherein the inhibitor and the chemotherapeutic have a synergistic effect against cancer cells.
- the inhibitor further comprises providing the inhibitor in an amount that is therapeutically effective to inhibit or reduce cellular or tissue proliferation selected from prostate cancer, castration- resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
- the present invention includes a method of modulating cellular or tissue proliferation comprising the steps of providing a therapeutic effective amount of an inhibitor that is selective for the enzyme AKR1C3 over its isoforms wherein the compound has the formula:
- the inhibitor is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-oxidethyl
- the inhibitor is:
- the inhibitor is: , wherein R2 is:
- the inhibitor is:
- R is:
- the inhibitor is:
- R3 is ;
- the inhibitor is:
- the inhibitor is formulated into a therapeutic effective amount sufficient to reduce or eliminate cancer cells.
- the inhibitor further comprises a chemotherapeutic agent, wherein the inhibitor and the chemotherapeutic have a synergistic effect against cancer cells.
- the inhibitor further comprises providing the inhibitor in an amount that is therapeutically effective to inhibit or reduce cellular or tissue proliferation selected from prostate cancer, castration- resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
- AML acute myeloid leukemia
- T-ALL T-cell acute lymphoblastic leukemia
- the prostate cancer and/or castrate-resistant prostate cancer, breast cancer, leukemia is resistant to chemotherapeutic agents.
- the AKR1C3 inhibitor is synergistic with a chemotherapeutic.
- the present invention provides a composition that selectively inhibits an aldo-keto reductase family 1 enzyme family in a selective manner that selects the AKR1C3 isoform comprising a therapeutic effective amount of a compound or derivatives thereof.
- the present invention provides a method of modulating cellular or tissue proliferation comprising the steps of: providing a therapeutic effective amount of an inhibitor that is selective for the enzyme AKR1C3 over its isoforms wherein the compound is one or more Class I, II, III, or IV baccharin derivatives.
- the cellular or tissue proliferation comprises prostate cancer and/or castrate-resistant prostate cancer, breast cancer, leukemia, ovarian cancer, endometrial cancer, or a combination thereof.
- the present invention provides a method of preventing the development of chemotherapeutic resistant cancers comprising the steps of: providing a therapeutic effective amount of a compound that possess selectivity for the enzyme AKR1C3 over its isoforms wherein the compound is one or more Class I, II, III, or IV baccharin derivatives.
- a method of treating, ameliorating or preventing cancer in a subject in need thereof comprising administering to said subject a composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one Class I, II, III, or IV baccharin derivative.
- FIGS. 1A-1D show images of some embodiments of the compounds of the present invention.
- FIGS. 2A-2C are tables showing prostate cancer cell viability was observed upon treatment with baccahrin.
- FIGS. 3A-3F are graphs showing KV-37 induces potent cytotoxic activity in multiple CaP cell lines.
- FIGS. 4A-4F show pre-treatment with KV-37 sensitizes LNCap and 22Rvl cells to ENZ cytotoxicity.
- FIGS. 5A-5E are images showing KV-37 induces apoptosis in 22Rvl cells.
- FIG. 6 is an image showing downstream molecular changes after KV-37 treatment in 22Rvl cells.
- FIG. 7 is an image showing KV-37 treatment of 22Rvl cells downregulated AKR1C3.
- FIGS. 8A-8B are images showing molecular changes following a combination treatment of KV-37 and ENZ in 22Rvl cells after 72 hour incubation.
- FIG. 9 is a graph of the compound KV-60-5(l) showing no cytotoxicity to LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hour exposure at 1 ⁇ concentration. IC50 value of KV-60-5(l) was determined to be 100 ⁇ .
- FIG. 10 is a graph of the compound KV-60-5(l) showing significant potentiation effect of the clinical antineoplastic enzalutamide in LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hours.
- FIG. 11 in an image of the modelling of KV-60-5a docked into the AKR1C3 enzyme.
- FIG. 12 shows a synthetic scheme for the inhibitor.
- FIG. 13 shows representative derivative compounds with a modification of phenyl (6).
- FIG. 14 shows representative derivative compounds with a modification of Dihydrocinnamyloxy (6).
- FIG. 15 shows representative derivative compounds with a Conversion of Ester to Amide Group (5).
- FIG. 16 shows representative derivative compounds with a Positional Switch of Prenyl & Dihydrocinnamyloxy Group (15).
- FIG. 17 shows representative derivative compounds with a Positional Switch of Dihydrocinnamoyloxy Group Analogs (3).
- FIG. 18 shows representative derivative compounds a Switch of Amide Bond Direction-Reverse Amides (10).
- FIG. 19 shows representative derivative compounds with a Modification of Carboxylate Group (2).
- FIGS. 20A to 20K show that KV-37 induces potent cytotoxic activity in multiple prostate cancer cell lines.
- FIG. 20 A Structure of KV-37. Percentage cell viability of 22Rvl cells cultured in (FIG. 20B) normal media (FIG. 20C) CSS media (FIG. 20D) normal media supplemented with 10 nM ⁇ 4- androstenedione and (FIG. 20E) CSS media supplemented with 10 nM A4-androstenedione. Percentage cell viability of LNCaP cells cultured in (FIG. 20F) normal media and (FIG. 20G) CSS media (FIG. 20H).
- KV-37 acts as a competitive Inhibitor in the conversion of [3H]A4-androstene-3,17-dione to testosterone.
- FIGS. 20J to 220K show that 22Rvl and LNCaPlC3 cells overexpress AKR1C3 whereas LNCaP and WPMY-1 exhibit low AKR1C3 levels.
- FIG. 20J Relative expression of AKR1C3 across prostate cancer cell lines.
- FIG. 20K Culture of 22Rvl cells in CSS media upregulates AKR1C3 expression levels. (NM, normal media; CSS, charcoal stripped serum media)
- FIGS. 21A to 21C shows that KV-37 inhibits conversion of [3H]A4-androstene-3,17-dione to testosterone in LNCaP-lC3 cells.
- LNCaP-lC3 cells (1.5 x 106 cells per well) platted in phenol red-free RPMI, 5% CSS-FBS, 2 mM L-glutamine, 1% P/S and were with DMSO or 10 ⁇ KV-37 for 30 min at 37 °C. After the preincubation, 100 nM [3H]A4-androstene-3,17-dione final concentration was added to the wells and cells were incubated for 24 h.
- FIG. 21A is solvent control; (FIG.
- FIG. 21B contains 10 ⁇ KV- 37 and (FIG. 21C) are LNCaP control cells treated with DMSO.
- the peak at 90 mm is androsterone and the peak at 120 mm is testosterone liberated after ⁇ -glucronidase treatment.
- the data reveal almost complete inhibition of AKR1C3 mediated testosterone production.
- FIGS. 22A to 22C shows the metabolic instability of (FIG. 22A) Baccharin and (FIG. 22B) ester derivative KV-32 on incubation with mouse S9 fractions. (FIG. 22C) Half-life of baccharin and derivative AKR1C3 inhibitors upon incubation with human S9 liver fractions.
- FIGS. 25A to 251 show that pre-treatment with KV-37 sensitizes prostate cancer cell lines to ENZ cytotoxicity.
- FIG. 25G LNCaP cells cultured in normal media and (FIG. 25H) LNCaP cells cultured in CSS media.
- FIG. 251 Quantification of the degree of synergism.
- FIGS. 26A to 26H show that co-treatments of KV-37 with ENZ do not exert synergistic cytotoxic effects in LNCaP and 22Rvl cells.
- FIGS. 27A to 27D shows that AKR1C3 overexpression in LNCaP cells (LNCaPlC3) increases the cytotoxic activity of KV-37 and induces resistance to ENZ that is overcome by pre-treatment with KV- 37.
- Percentage cell viability of LNCaPlC3 cultured in CSS media after (FIG. 27A) KV-37 treatment at indicated time points and concentrations (table insert showing the IC50 values)
- FIG. 27B ENZ treatment at indicated time points and concentrations
- FIG. 27C co-treatment with KV-37 and ENZ for 72 h
- FIG. 27D pre-treatment with KV-37 followed by ENZ exposure for 72 h.
- FIGS. 28A to 28D show that KV-37 induces apoptosis in prostate cancer cells alone and potentiates the degree of apoptosis when combined with ENZ.
- FIG. 28A Dose and time-dependent increase in apoptotic cell population after treatment with KV-37 in indicated cell lines as analyzed by AnnexinV/PI co-staining.
- FIG. 28B Increase in apoptotic cell population after treatment with indicated concentrations of KV-37, ENZ and a 24 h pre-treatment combination of KV-37 and ENZ in indicated cell lines analyzed 72 h post ENZ exposure by AnnexinV/PI co- staining.
- FIG. 28C Western blot showing an increase in C-caspase3 and C-PARP levels after treatment with KV-37 at indicated concentration and time points in prostate cancer cells.
- FIGS. 29 A to 29D show that Molecular changes following KV-37 treatment.
- FIGS. 30A to 30E show that KV-37 induces tumor growth inhibition in prostate cancer xenografts.
- FIG. 30C Representative images showing a reduction in tumor size after treatment with 20 mg/Kg KV-37.
- FIG. 30D shows the mice body weight versus days post implantation.
- FIG. 30E shows the pharmacokinetic parameters determined in plasma after treatment with 20 mg/Kg KV-37.
- FIGS. 31A to 3 ID Indomethacin exerts a weak synergistic drug effect in LNCaPlC3 cells and KV-37 does not induce cytotoxicity alone or in combination with ENZ in WPMY-1 cells.
- FIG. 32 shows the SAR strategy map for the design of Baccharin derivatives.
- FIGS. 33A to 33D show co-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (33A) 1 (33B) 26a (33C) 49a (33D) 49g at indicated concentrations.
- FIGS. 34A to 34D show Co-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (34A) 1 (34B) 26a (34C) 49a (34D) 49g at indicated concentrations.
- FIGS. 35A to 35D show Co-treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (35A) 1 (35B) 26a (35C) 49a (35D) 49g at indicated concentrations.
- FIGS. 36A to 36D show 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (36A) 1 (36B) 26a (36C) 49a (36D) 49g at indicated concentrations.
- FIGS. 37A to 37D show 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (37 A) 1 (37B) 26a (37C) 49a (37D) 49g at indicated concentrations.
- FIGS. 38A to 38D show 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (38A) 1 (38B) 26a (38C) 49a (38D) 49g at indicated concentrations.
- FIGS. 38E to 38F show Treatment of AML cells with chemotherapeutics. Percentage cell viability after 72 h treatment of AML cells with (38E) daunorubicin and (38F) AraC at indicated concentrations.
- FIGS. 39A to 39D show that co-treatment of AKR1C3 inhibitors with AraC in HL-60 cells. Percentage cell viability after 72 h co-treatment of AraC with (39A) 1 (39B) 26a (39C) 49a (39D) 49g at indicated concentrations.
- FIGS. 40A to 40D show that co-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h co-treatment of AraC with (40A) 1 (40B) 4 (40C) 6 (40D) 7 at indicated concentrations.
- FIGS. 41A to 41D show that co-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h co-treatment of AraC with (41 A) 1 (4 IB) 4 (41C) 6 (4 ID) 7 at indicated concentrations.
- FIGS. 42A to 42D show the effect of 24 h Pre-treatment of AKR1C3 inhibitors with AraC in HL-60 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (42A) 1 (42B) 26a (42C) 49a (42D) 49g at indicated concentrations.
- FIGS. 43 A to 43D show the effect of 24 h Pre-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (43A) 1 (43B) 26a (43C) 49a (43D) 49g at indicated concentrations.
- FIGS. 44A to 44D show the effect of 24 h Pre-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (44A) 1 (44B) 26a (44C) 49a (44D) 49g at indicated concentrations.
- FIGS. 45A to 45D show the synergistic effect of compound 49a with daunorubicin in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and daunorubicin in (45 A) COG-317 (45B) COG-329 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of 49a in (45C) COG-317 (45D) COG-329 cells.
- FIGS. 45E to 45G shows treatment of COG T-ALL cells with 49a and chemotherapeutics. Percentage cell viability after 72 h treatment of AML cells with (45E) 49a (45 G) daunorubicin and (45H) AraC at indicated concentration and time points.
- FIGS. 46A to 46D show the synergistic effect of compound 49a with AraC in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and AraC in (46 A) COG-317 (46B) COG-329 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of 49a in (46C) COG- 317 (46D) COG-329 cells.
- FIGS. 46E and 46F Combination treatment of BMMNC cells with 49a and chemotherapeutics. Percentage cell viability of BMMNC cells after pre-treatment with 49a followed by 72 h incubation with (46E) AraC and (46F) daunorubicin at indicated concentrations.
- FIG. 47 shows a Baccharin SAR map
- FIGS. 48A to 48D show the treatment of HL-60 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of HL-60 cells with (48A) 1 (48B) 26a (48C) 49a (48D) 49g at indicated concentrations and time points.
- FIGS. 49A to 49D show the treatment of KGla cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of KGla cells with (49 A) 1 (49B) 26a (49C) 49a (49D) 49g at indicated concentrations and time points.
- FIGS. 50A to 50D shows the treatment of THP-1 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of THP-1 cells with (50A) 1 (50B) 26a (50C) 49a (50D) 49g at indicated concentrations and time points. DETAILED DESCRIPTION OF THE INVENTION
- AKR1C3 is an acronym for aldo-keto reductase family 1, member C3 polypeptide, Aldo-keto reductase 1C3, and is also referred to as type 5 17 -hydroxysteroid dehydrogenase.
- Prostate cancer is the second leading cause of mortality among American men with the highest incidence rate of all cancers reported in the United States. Surgical removal of prostate by radical prostatectomy and radiation therapy are applied to resect localized tumor. However, the treatment of advanced and metastatic forms of prostate cancer relies heavily on androgen deprivation therapy (ADT) that involves surgical (orchiectomy) and/or chemical castration. Chemical agents such as GnRH agonists (Lupreolide, goserelin, buserelin) and antiandrogens (bicalutamide, enzalutamide) reduce the levels of circulating androgens that consequently retard prostate cancer cell proliferation.
- GnRH agonists Loreolide, goserelin, buserelin
- antiandrogens bicalutamide, enzalutamide
- CRPC Castration Resistant Prostate Cancer
- AKR1C3 plays a role in cancer development and chemotherapy resistance, and that this enzyme is a target for treating castrate resistant prostate cancer.
- inhibitors are available that can block AKRlC3's enzymatic activity, they also block AKR1C1 and AKRlC2's activities, which affects healthy hormonal levels and balance.
- the aldo-keto reductase family 1-member C (AKR1C) enzymes are oxidoreductases that catalyze NADPH-dependent reductions of aldehydes and ketones in a range of steroids, carbohydrates, and prostaglandins.
- the isoform AKR1C3 specifically catalyzes the conversion of androgen precursors to potent androgen receptor (AR) ligands, testosterone and a-dihydrotestosterone.
- AKR1C3 is also known as prostaglandin F synthase and as it catalyzes the conversion of prostaglandin D2 to 11 ⁇ -prostaglandin F2 a and its prostanoids, which affects the growth and spread of myeloblasts and myelocytes, it is therefore an important regulator of myeloid cell proliferation and differentiation.
- AKR1C3 makes it responsible for both the pathogenesis and progression of hormone dependent and independent cancers, including castration resistant prostate cancer (CRPC) and acute myeloid leukemia (AML). Additionally, AKR1C3 can lead to clinical chemotherapeutic resistance, as it reduces the effectiveness of first line of defense drugs for CRPC and AML.
- CRPC castration resistant prostate cancer
- AML acute myeloid leukemia
- AKR1C3 inhibition research has found compounds with some degree of selectivity, but further improvements can be made to reduce the inhibition of the other AKR1C isoforms.
- the strategy behind these mechanisms has been taking known AKR1C3 inhibitors, and altering their functional groups through meta and para-directing to increase the inhibitor's level of selectivity. Due to the lack of high selectivity in previous research, no AKR1C3 inhibitor has been approved for clinical testing.
- the present invention provides a series of compounds that possess the greatest selectivity for the enzyme AKR1C3 over its isoforms ever reported.
- the AKR1C3 enzyme is a therapeutic target for prostate and breast cancer and leukemia.
- Abiraterone acetate acts by inhibiting CYP17A1, an upstream enzyme in the steroid biosynthetic pathway, which was approved by the FDA for CRPC treatment in 2011 and is effective in treating state 2 of CRPC.
- CYP17A1 an upstream enzyme in the steroid biosynthetic pathway
- DOC desoxycorticosterone
- ENZ Enzalutamide
- AR potent androgen receptor
- Aldo-keto reductase 1C3 is a downstream enzyme in the steroid biosynthetic pathway and plays a pivotal role in the pathogenesis and progression of CRPC by catalyzing the conversion of weak androgen precursors to the potent androgen receptor (AR) ligands: testosterone (T) and 5a- dihydrotestosterone (5a-DHT). Also known as prostaglandin (PG) F synthase, AKR1C3 catalyzes the conversion of PGD2 to l ip-PGF2a and PGF2a prostanoids and hence acts as an important regulator of myeloid cell proliferation and differentiation.
- PG prostaglandin
- AKR1C3 This dual enzymatic action makes AKR1C3 responsible for the pathogenesis and progression of both hormone dependent and independent cancers.
- a considerable interest has been developed in investigating the role of AKR1C3 in a variety of cancers of the breast, lungs and colon.
- Prior studies have already validated the role of AKR1C3 in the pathogenesis and progression of prostate cancer.
- Clinically, AKR1C3 has been shown to be the most upregulated enzyme isoform among CRPC patients. Apart from inducing a CRPC phenotype, AKR1C3 is also responsible to mediate resistance to ENZ by providing a source of intratumoral androgens.
- AKR1C3 As the enzyme acts at the final steps of androgen synthesis pathway, it eliminates the risk of DOC accumulation and eventually development of life threatening hypertension. Such activities make AKR1C3 an attractive target for managing CRPC disease progression as well as therapeutic resistance.
- Related isoforms AKR1C1 and AKR1C2 share high homology with AKR1C3 and are responsible for normal steroid metabolism. Hence, design of a potent yet isoform selective AKR1C3 inhibitor is a major challenge in the drug discovery process.
- EXAMPLE 1 Novel compounds with higher selectivity for the enzyme AKR1C3 using a structure- activity relationship (SAR) optimization approach.
- SAR structure- activity relationship
- SAR structure-activity relationship
- derivative AKR1C3 inhibitors display a very potent synergistic effect in combination with clinical chemotherapeutic agents in a panel of leukemia cell lines.
- the inventors demonstrate the activity of a hydrolytically stable amide analogue of baccharin to exert a cytotoxic effect in a panel of prostate cancer cell lines and a very high degree of synergistic drug interaction with ENZ.
- Mechanistic studies demonstrate apoptotic cell death in sensitive and ENZ resistant prostate cancer cell lines with a consequent reduction in AR and PSA levels.
- FIGS. 1A-1D show examples of AKR1C3 inhibitors synthesized based on baccharin structural scaffold.
- KV-37 was chosen for further studies as it demonstrated a very potent AKR1C3 inhibitory IC50 of 66 nM.
- Androgen dependent 22Rvl and LNCaP cell lines were chosen to evaluate the activity of KV-37.
- 22Rvl cells were found to be high expressors of AKR1C3 as compared to LNCaP.
- 22Rvl cell line, being intrinsically resistant to ENZ was an apt model for conducting CRPC studies.
- baccharin and the ester analogue were readily hydrolyzed.
- the amide inhibitor KV-37 displayed a remarkable stability and half-life of >240 min.
- the AKR1C3 inhibitory activity of baccharin was completely abrogated when, upon hydrolysis it yielded the known phenol drupanin, exhibiting AKR1C3 inhibition at IC50 of 15 ⁇ and 7 fold selectivity over AKR1C2.
- FIGS. 2A-2C are tables showing prostate cancer cell viability observed upon treatment with baccahrin. No reduction in prostate cancer cell viability was observed upon treatment with baccahrin.
- FIG. 2A, 2B, 2C very low levels of cytotoxicity were observed in DU-145, LNCaP and VCaP cells cultured in normal media when treated with baccharin at concentrations up to 100 ⁇ .
- Effect of AKR1C3 inhibitors on CaP cell viability Baccharin and derivative AKR1C3 inhibitors were screened for cytotoxic effects in a panel of prostate cancer cell lines (DU-145, LNCaP, 22Rvl, VCaP).
- FIGS. 3A-3F are graphs showing KV-37 induces potent cytotoxic activity in multiple CaP cell lines.
- Dose and time dependent reduction in cell viability after inhibitor treatment LNCaP cells cultured in normal media and CSS media respectively.
- FIG. 3C VCaP cells cultured in normal media.
- FIGS. 3D, 3E, 3F 22Rvl cells cultured in normal media, CSS media and CSS media supplemented with 10 nM AD respectively.
- a further enhancement in cytotoxic activity was observed when the cells were cultured in charcoal stripped serum (CSS) media.
- CCS charcoal stripped serum
- the IC50 of KV-37 decreased from 45 ⁇ to 25 ⁇ in LNCaP cells and from 50 ⁇ to 25 ⁇ in 22Rvl cells upon incubation in CSS media as against hormone replete media.
- KV-37 was incubated with 22Rvl cells in CSS media supplemented with 10 nM A4-androstenedione (AD). Similar dose and time dependent reduction in cell viability was observed: IC50 being 25 ⁇ at 72 hours. At higher inhibitor concentrations of 75 ⁇ complete abrogation of cell viability was observed in LNCaP and 22Rvl cells when cultured in CSS media.
- A4-AD is a weak AR ligand and a substrate for AKR1C3. The enzyme converts A4-AD to more potent AR ligand testosterone that drives cell proliferation.
- 10 nM A4-AD to CSS culture further mimics the clinical CRPC phenotype when only castrate levels of circulating androgens are available.
- Compound KV-37 sensitizes CaP cells to enzalutamide cytotoxicity and exerts a synergistic drug effect:
- the dose response curve of ENZ in LNCaP and 22Rvl cells displays an increase in resistance upon culture in CSS media.
- LNCaP and 22Rvl cells were pre-treated with KV-37 for 24 hours followed by exposure to ENZ.
- the combination demonstrated a remarkable synergistic effect at both the time points of 48 and 72 hours post ENZ exposure in normal as well as CSS media.
- concentrations as low as 1 ⁇ of KV-37 were capable of sensitizing both 22Rvl and LNCaP cell lines to reduce cell viability by 50% upon treatment with only 1 ⁇ ENZ when either agent alone did not exert any toxic behavior.
- FIGS. 4A-4F show pre-treatment with KV-37 sensitizes LNCap and 22Rvl cells to ENZ cytotoxicity.
- FIG. 4A, 4B 24 hour pre-treatment of LNCaP cells in normal and CSS media followed by treatment with various concentrations of ENZ for 72 hours
- FIG. 4C, 4D, 4E 24 hour pre-treatment of 22Rvl cells in normal and CSS media followed by treatment with various concentrations of ENZ at indicated time points.
- FIG. 4F Quantification of the degree of synergism. Combination and dose reduction indices (CI, DRI) generated using cognitive system. However, co-treatment experiments did not show any adjuvant effect between both cell lines. Quantification of degree of synergism was made using the Chou- Talalay method. Up to a 100-fold reduction in ENZ dosing was achieved in 22Rvl cells while in LNCaP cells 40 fold reduction was observed.
- FIGS. 5A-5D are images showing KV-37 induces apoptosis in 22Rvl cells.
- FIG. 5A, 5B Dose dependent Increase in apoptotic cell population in 22Rvl cells at indicated time points.
- FIG. 5C Graph quantifying the percentage of apoptotic cells after KV-37 treatment.
- FIG. 5E Western blots showing a dose dependent increase in c-caspase and C-PARP after treatment with KV-37 at indicated time points in CSS media.
- KV-37 induces cell death in 22Rvl cells via apoptosis: 22Rvl cells cultured in CSS media were treated with increasing doses KV-37 for 48 and 72 hours and stained with AnnexinV/PI to measure the apoptotic cell percentage following treatment.
- FIG. 5 outlines the increase in the percentage of apoptotic cells with increase in concentration of KV-37 as analyzed by flow cytometry. Compound incubation for 72 hours (FIG. 5B) demonstrates a further increase in apoptotic cell percentage as compared to the 48 hour time point (FIG. 5A).
- FIGS. 5C and 5D outline the percent increase in apoptosis as compared to the control.
- FIG. 6 is an image showing downstream molecular changes after KV-37 treatment in 22Rvl cells. Western blot showing a decline in AKR1C3, AR and PSA levels.
- PSA is widely used as a biomarker for prostate cancer and governs the disease progression.
- the inventors probed PSA levels after inhibitor treatment that shows a dose dependent reduction in PSA expression signifying a beneficial therapeutic outcome.
- 22Rvl cells were treated with KV-37 at 1 and 10 ⁇ concentration for 48 hours followed by immunostaining against AKR1C3.
- FIG. 7 is an image showing KV-37 treatment of 22Rvl cells downregulated AKR1C3. Immunocytochemical analysis showing a concentration dependent decline in AKR1C3 levels after 48 hours incubation. The data clearly indicates a reduction in AKR1C3 levels as denoted by the green stain. Counterstaining with DAPI shows no morphological changes in the nucleus. Combination of KV-37 with ENZ synergistically downregulates AKR1C3, AR and PSA expression and upregulates c-caspase and c-PARP levels:
- FIGS. 8A-8B are images showing molecular changes following a combination treatment of KV-37 and ENZ in 22Rvl cells after 72 hours incubation.
- western blot analysis was carried out to measure molecular changes in the combination treatments.
- the combination of 25 ⁇ KV-37 with 25 ⁇ ENZ completely abrogates PSA expression with a concomitant decrease in AKR1C3 and AR levels at 72 hours (FIG. 8B).
- a predominant increase in c-caspase and c-PARP levels was also observed at both time points. Taken together the data indicate apoptotic cell death as a primary mechanism of synergistic cytotoxic effect as a consequence of reduction in the activity of AKR1C3 and AR.
- compositions are AKR1C3 inhibitors with >1800-fold selectivity for AKR1C3 over its other isoforms. It provides a composition for the treatment of cancer and contributes to the effectiveness of chemotherapeutic drugs that are otherwise reduced in effect by AKR1C3.
- the aldo-keto reductase family 1 member C (AKR1C) enzymes are oxidoreductases, which catalyze the NADPH-dependent reduction of aldehyde and ketone functionalities on a range of steroids, carbohydrates, and prostaglandins.
- the AKR1C3 enzyme isoform catalyzes the downstream conversion of androgen precursors to the potent androgen receptor (AR) ligands: testosterone and 5a- dihydrotestosterone (5a-DHT).
- AKR1C3 catalyzes the conversion of PGD2 to l ip-PGF2a and PGF2a prostanoids and hence acts as an important regulator of myeloid cell proliferation and differentiation.
- This dual enzymatic action makes AKR1C3 responsible for the pathogenesis and progression of both hormone dependent and independent cancers such as Castration Resistant Prostate Cancer (CRPC) and Acute Myeloid Leukemia (AML).
- CRPC Castration Resistant Prostate Cancer
- AML Acute Myeloid Leukemia
- AKR1C3 is not only responsible for the pathogenesis of the aforementioned malignancies but also mediates resistance to clinical chemotherapeutics.
- KV-60-5a the most selective AKR1C3 inhibitor yet identified, is shown to be non-toxic to LNCaP prostate cancer cells (when grown in charcoal stripped serum (CSS), which serves to upregulate AKR1C3 expression) at 1 ⁇ concentration.
- FIG. 9 is a graph of the compound KV-60-5a showing no cytotoxicity to LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hours exposure at 1 ⁇ concentration. IC50 value of KV-60-5a was determined to be 100 ⁇ . Note that although AKR1C3 expression is enhanced in LNCaP cells grown in CSS media the relative expression of the enzyme target is still low. Clinical samples of castration resistant prostate cancer and other cell lines express much greater levels of AKR1C3. In such cell lines both direct toxicity and potentiation effect are expected to be much greater. These experiments are currently ongoing.
- FIG. 10 is a graph of the compound KV-60-5a showing significant potentiation effect of the clinical antineoplastic enzalutamide in LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hours.
- Synergistic treatment of 1 ⁇ of KV-60-5a with 1 ⁇ of the clinical antineoplastic enzalutamide results in 42% cell viability compared with 1 ⁇ of enzalutamide alone which results in 82% cell viability (FIG. 10).
- FIG. 11 in an image of the modelling of KV-60-5a docked into the AKR1C3 enzyme.
- the inventors Enabled by the synthesized library of AKR1C3 inhibitors the inventors have conducted quantitative structure -activity relationship (QSAR) computer modelling that has identified compounds with retained selectivity and predicted picomolar affinity. Modelling predictions show that the structure of the highly selective inhibitor KV-60-5a changes the pose of the compound when bound with AKR1C3. This results in pi-pi stacking interactions between aromatic amino acids TYR216, TRP227 and PHE311, and aromatic constituents of the inhibitors, which accounts for the enhanced affinity.
- QSAR quantitative structure -activity relationship
- FIG. 12 shows a synthetic scheme for the inhibitor.
- 3-bromo-5-iodo-N-(2- phenylethyl)benzamide (2) To a solution of 3-bromo-5-iodo benzoic acid (500 mg, 1.5 mmol) in dry toluene (10 mL) was added SOC12 (290 ⁇ , 4 mmol) and the mixture was refluxed overnight. The reaction vessel was cooled to room temperature and the solvent was evaporated in vacuo. The resultant brown oil was used further without purification.
- FIG. 13 shows representative derivative compounds with a modification of phenyl (6).
- FIG. 14 shows representative derivative compounds with a modification of Dihydrocinnamyloxy (6).
- FIG. 15 shows representative derivative compounds with a Conversion of Ester to Amide Group (5).
- FIG. 16 shows representative derivative compounds with a Positional Switch of Prenyl & Dihydrocinnamyloxy Group (15).
- FIG. 17 shows representative derivative compounds with a Positional Switch of Dihydrocinnamoyloxy Group Analogs (3).
- FIG. 18 shows representative derivative compounds a Switch of Amide Bond Direction-Reverse Amides (10).
- FIG. 19 shows representative derivative compounds with a Modification of Carboxylate Group (2).
- Aldo-keto reductase 1C3 also known as type 5 17 ⁇ -hydroxysteroid dehydrogenase is responsible for intratumoral androgen biosynthesis, contributing to the development of castration- resistant prostate cancer (CRPC) and eventual chemotherapeutic failure.
- Significant upregulation of AKR1C3 is observed in CRPC patient samples and derived CRPC cell lines.
- AKR1C3 is a downstream steroidogenic enzyme synthesizing intratumoral testosterone (T) and 5a-dihydrotestosterone (DHT), the enzyme represents a promising therapeutic target to manage CRPC and combat the emergence of resistance to clinically employed androgen deprivation therapy.
- KV-37 a potent, isoform selective and hydrolytically stable AKR1C3 inhibitor (E)-3-(4-(3-methylbut-2-en-l-yl)-3-(3- phenylpropanamido)phenyl)acrylic acid (KV-37) which reduces prostate cancer cell growth in vitro and in vivo, and sensitizes CRPC cell lines (22Rvl and LNCaPlC3) towards enzalutamide cytotoxicity.
- KV-37 does not induce cytotoxicity in non- malignant WPMY-1 prostate cells nor does it induce weight loss in mouse xenografts.
- KV-37 reduces androgen receptor (AR) transactivation and prostate specific antigen (PSA) expression levels in CRPC cells lines indicating a therapeutic drug effect in prostate cancer.
- AR androgen receptor
- PSA prostate specific antigen
- Prostate cancer is the third (1) leading cause of mortality among American men and has the highest incidence of all cancers reported in the US (2).
- Surgical removal of the prostate by radical prostatectomy and radiation therapy to resect the localized tumor are standard therapeutic interventions (3).
- the treatment of advanced and metastatic forms of prostate cancer relies heavily on androgen deprivation therapy (ADT) that involves surgical (orchiectomy) and/or chemical castration (4).
- Chemical agents such as gonadotropin releasing hormone (GnRH) agonists (Lupreolide, (5) goserelin, (6) buserelin (7)) and antiandrogens (bicalutamide (8)) retard prostate cancer cell proliferation and lead to remission.
- GnRH gonadotropin releasing hormone
- CRPC castration-resistant prostate cancer
- Abiraterone acetate was approved by the US Food and Drug Administration for CRPC treatment in 2011 and acts by inhibiting P450cl7 (13), an upstream enzyme in the steroid biosynthetic pathway and is effective in CRPC where tumor progression is dependent on intracrine androgen synthesis that consequently activates AR signaling (14,15).
- P450cl7 13
- DOC desoxycorticosterone
- prednisone co-administration with prednisone is imperative
- Enzalutamide ENZ
- ENZ is another clinically used therapeutic that exerts potent androgen receptor (AR) antagonistic activity by inhibiting AR nuclear translocation, co-activator recruitment and AR binding to androgen response elements (ARE's) (17).
- the chemotherapeutic is effective up to stage 3 CRPC where proliferation is dependent on AR activation that occurs even in the absence of an androgen ligand (15).
- stage 3 CRPC where proliferation is dependent on AR activation that occurs even in the absence of an androgen ligand (15).
- resistance develops rapidly due to increase in intratumoral androgen biosynthesis (18), AR overexpression, AR mutation, that makes the receptor ligand promiscuous, or the appearance of AR splice variants that make the AR constitutively active in the absence of the ligand binding domain (15,19,20).
- Type 5 17 -hydroxy steroid dehydrogenase also known as aldo-keto reductase 1C3 (AKR1C3) acts downstream in the steroidogenesis pathway and plays a pivotal role in the pathogenesis and progression of CRPC by catalyzing the conversion of weak androgen precursors to the potent AR ligands: testosterone (T) and 5a-dihydrotestosterone (5a-DHT) (23).
- PG prostaglandin
- AKR1C3 catalyzes the conversion of PGD2 to l ip-PGF2a and PGF2a prostanoids (24).
- An increase in PGF2a leads to activation of the FP receptor and consequently induces proliferation and radiation resistance in prostate cancer cells (25).
- AKR1C3 has been shown to be the most upregulated steroidogenic enzyme in CRPC patients (26). Apart from inducing a CRPC phenotype, AKR1C3 also mediates resistance to ENZ and AA by providing a source of intratumoral androgens and this resistance can be surmounted by indomethacin (INDO) a specific AKR1C3 inhibitor that also inhibits COX isozymes (27,28). As the enzyme acts at the final steps of the androgen synthesis pathway in the prostate (16), AKR1C3 inhibitors would not cause the accumulation of DOC in the adrenal gland and would not have to be co-administered with prednisone.
- INDO indomethacin
- AKR1C3 an attractive target for managing CRPC disease progression as well as countering therapeutic drug resistance.
- the related isoforms AKR1C1 and AKR1C2 share high homology with AKR1C3, function to eliminate DHT, and should not be inhibited (29).
- Numerous AKR1C3 inhibitors with diverse scaffolds are known however, none have made their way into the clinic (30-32), Considerable interest has developed in the role of AKR1C3 in a variety of cancers of the breast (33), lungs (34) and colon (35).
- AKR1C3 inhibitor KV-37 (FIG. 20A) (36-38)
- the inventors have previously reported AKR1C3 inhibitors that act synergistically with etoposide and daunorubicin as chemotherapeutic agents in a panel of leukemia cell lines (38).
- the AKR1C3 inhibitor KV-37 exerts high synergistic drug interaction in combination with ENZ, which is superior to that seen with INDO.
- WPMY-1 cells were purchased from ATCC and maintained in DMEM media supplemented with 5% FBS, 100 U/mL penicillin and 0.1 mg/mL streptomycin. Where indicated, cells were also cultured in charcoal stripped (CSS) media prepared by supplementing RPMI 1640 without phenol red with charcoal stripped FBS. All cells were maintained at 37oC in a humidified incubator with 5% carbon dioxide. Enzalutamide (ENZ) or MDV3100 (catalog no. 50-101-3979) and indomethacin (INDO) (catalog no. AAA1991006) were purchased from Fisher Scientific. Stock solutions of ENZ, INDO and KV-37 were prepared in DMSO and were serially diluted for cell culture treatments maintaining the final DMSO concentration at less than 1%.
- Buffer A dH20 + 0.1% formic acid
- Buffer B ACN + 0.1% formic acid
- column Agilent C18 XDB column 5 micron packing 50 X 4.6 mm size; 0 - 1 min 97% A, 1 - 2.5 min gradient to 99% B, 2.5 - 3.5 min 99% B, 4 - 4.1 min gradient to 97% A, 4.1 - 4.5 min 97% A
- IS Warfarin (in MeOH, transition 307.3 to 160.9). Ion transitions followed were 363.1 [M-H] to 186.9 for baccharin and KV-32, and 362.1 [M-H] to 318.1 for KV-37.
- Cell Viability Assays Cells were seeded at a density of 10,000 cells/well in 96-well plates and were incubated in either normal media (24 h) or charcoal stripped serum (CSS) media (48 h). Treatments with ENZ, INDO, KV-37 or combinations of ENZ and KV-37 were made with or without 10 nM A4-AD (AKR1C3 substrate) and incubated at the indicated time points (24, 48, 72 and 96 h). For pretreatment experiments, cells were treated with KV-37 for 24 h followed by the addition of ENZ and incubated for further 72 h. Cell viability was determined by the MTS tetrazolium dye assay as described previously (38).
- LNCaPlC3 cells (1.5 x 106 cells per well) were placed in 2 mL phenol red-free RPMI, 5% CD-FBS, 2 mM L-glu, 1% P/S. Cells were incubated with DMSO, or 10 ⁇ or 30 ⁇ KV-37 for 30 min at 37 °C. After the pre -incubation, 100 nM 3H A4-AD final concentration was added to the wells and cells were incubated for 24 h. Cell media was transferred into labeled borosilicate glass tubes after 48 h and extracted with 2 mL ethyl acetate.
- AnnexinV/FITC apoptosis assay was performed using a kit and according to the manufacturer's instructions (BD Biosciences, catalog no. 556547). Cells were seeded at a density of 0.2 x 106 cells/well in 24- well plates and incubated in CSS media for 48 h followed by treatment with KV-37 for 48 and 72 h. For pretreatment experiments, cells were treated with KV-37 for 24 h followed by the addition of ENZ and incubated for a further 72 h. After appropriate treatments, the samples were analyzed by flow cytometery (Accuri C6, Ann Arbor, MI, USA).
- Tumor volumes were measured twice a week with Vernier calipers and tumor volume was calculated as (L*W2)*3.14)/6.
- mice were humanely euthanized and tumors were collected, weighed and frozen in liquid nitrogen after taking pictures.
- Whole blood was collected in an eppendorf tube with ACD anti-coagulant for plasma separation for pharmacokinetic evaluation of KV-37.
- AKR1C3 inhibitors of AKR1C3 from various structural classes (flavones (30), jasmonates (31) and NSAID's (23,33)) have previously been described.
- IC50 66 nM
- selectivity 109-fold over AKR1C2
- the difference between this Ki value and the IC50 value seen in the preliminary inhibitor screen is consistent with the lower Ki value for the E.NADP+. inhibitor complex which is seen with other AKR1C3 inhibitors when they are screened in the oxidation direction (FIG. 201) (40).
- the 22Rvl cell line shows increased expression of AKR1C3 when grown in CSS media and possesses intrinsic resistance to ENZ and AA (27).
- LNCaP cells stably overexpressing AKR1C3 (LNCaP 1C3) were used to further validate the effect of KV-37 on the target (12).
- WPMY-1 cells, a non-malignant prostate stromal cell line, devoid of AKR1C3 expression were employed to evaluate the selective cytotoxicity of KV-37.
- FIG. 201 shows Competitive inhibition of NADPH dependent reduction of A4-androstene-3,17-dione mediated by AKR1C3 by KV-37.
- KV-37 acts as a competitive Inhibitor in the conversion of [3 ⁇ ] ⁇ 4- androstene-3,17-dione to testosterone.
- Increasing concentrations of KV-37 (0-10 ⁇ ) were used to inhibit the NADPH dependent conversion of fixed concentrations of [3H]A4-androstene-3,17-dione to testosterone catalyzed by recombinant AKR1C3 as measured by radiochromatography.
- FIG. 20J 22Rvl and LNCaPlC3 cells overexpress AKR1C3 whereas LNCaP and WPMY-1 exhibit low AKR1C3 levels.
- FIG. 20J Relative expression of AKR1C3 across prostate cancer cell lines.
- FIG. 20K Culture of 22Rvl cells in CSS media upregulates AKR1C3 expression levels. (NM, normal media; CSS, charcoal stripped serum media).
- KV-37 inhibits AKR1C3 activity in prostate cancer cell lines by inhibiting the conversion of A4-AD to testosterone.
- KV-37 was used to inhibit the production of testosterone in LNCaPlC3 cells following ⁇ - glucuronidase treatment of the aqueous phase. At a concentration of 10 ⁇ , >80% inhibition of testosterone production is blocked and the residual testosterone produced is that seen in untransfected LNCaP cells (FIG. 21A to 21C).
- FIG. 21A to 21C KV-37 inhibits conversion of [3H]A4-androstene-3,17-dione to testosterone in LNCaP- 1C3 cells.
- LNCaP-lC3 cells (1.5 x 106 cells per well) platted in phenol red-free RPMI, 5% CSS-FBS, 2 mM L-glutamine, 1% P/S and were with DMSO or 10 ⁇ KV-37 for 30 min at 37 °C. After the preincubation, 100 nM [3H]A4-androstene-3,17-dione final concentration was added to the wells and cells were incubated for 24 h.
- FIG. 21A is solvent control; FIG.
- FIG. 21B contains 10 ⁇ KV-37 and FIG. 21C are LNCaP control cells treated with DMSO.
- the peak at 90 mm is androsterone and the peak at 120 mm is testosterone liberated after ⁇ -glucronidase treatment.
- the data reveal almost complete inhibition of AKR1C3 mediated testosterone production.
- FIG. 22A-22C Metabolic instability of (FIG. 22 A) Baccharin and (FIG. 22B) ester derivative KV-32 on incubation with mouse S9 fractions.
- FIG. 22C shows the half-life of baccharin and derivative AKR1C3 inhibitors upon incubation with human S9 liver fractions.
- AKR1C3 inhibitor KV-37 exhibits greater cytotoxicity to prostate cancer cell lines compared to baccharin.
- Baccharin and its analog KV-37 were screened for cytotoxic effects in a panel of human prostate cancer cell lines (LNCaP, 22Rvl and LNCaPlC3).
- Baccharin consistently displayed no toxicity among all the cell lines tested, up to concentrations as high as 100 ⁇ (FIG. 23A to 23C). This is in agreement with previous studies (38).
- treatment with KV-37 demonstrated greater cytotoxicity in a dose and time- dependent manner in androgen dependent 22Rvl and LNCaP cell lines.
- the cytotoxic effect was greater in 22Rvl cells due to the higher expression of AKR1C3 as compared to the LNCaP cell line (FIG. 20A-H).
- FIG. 20B, 20C, 20H normal media
- AKR1C3 inhibitor KV-37 sensitizes ENZ resistant prostate cancer cells to ENZ by exerting a synergistic drug effect.
- the 22Rvl cell line is intrinsically resistant to ENZ (27).
- the dose-response curve of ENZ in 22Rvl cells displays increasing resistance upon culture in CSS media (supplementary FIG. S6), which is accompanied by an increase in AKR1C3 expression (FIGS. 20J and 20K).
- LNCaP cells that are sensitive to ENZ-induced cytotoxicity were used as controls to compare the effect of KV-37 in combination with ENZ.
- FIGS. 26A to 26H Co-treatments of KV-37 with ENZ do not exert synergistic cytotoxic effects in LNCaP and 22Rvl cells.
- Pre-treatment with KV-37 sensitizes prostate cancer cell lines to ENZ cytotoxicity.
- Percentage cell viability of prostate cancer cells when pre-treated with KV-37 for 24 h followed by ENZ treatment at indicated concentrations and time points in (FIG. 25 A) 22Rvl cells cultured in normal media (FIG. 25B) and (FIG. 25C) 22Rvl cells cultured in CSS media (FIG. 25D) 22Rvl cells cultured in normal media supplemented with 10 nM A4-androstenedione (FIG. 25E) and (FIG. 25F) 22Rvl cells cultured in CSS media supplemented with 10 nM A4-androstenedione (FIG.
- AKR1C3 overexpression in LNCaP cells confers resistance to ENZ cytotoxicity that is reversed upon KV-37 treatment.
- KV- 37 induced a dose- and time -dependent reduction in cell viability of LNCaPlC3 cells (FIG. 27A and table insert).
- the cytotoxic drug effect increased further as compared to 22Rvl and low AKR1C3 expressing LNCaP cells at all time points tested.
- FIGS. 27A to 27D AKR1C3 overexpression in LNCaP cells (LNCaPlC3) increases the cytotoxic activity of KV-37 and induces resistance to ENZ that is overcome by pre-treatment with KV-37.
- Percentage cell viability of LNCaPlC3 cultured in CSS media after (FIG. 27A) KV-37 treatment at indicated time points and concentrations (table insert showing the IC50 values)
- FIG. 27B ENZ treatment at indicated time points and concentrations
- FIG. 27C co-treatment with KV-37 and ENZ for 72 h
- FIG. 27D pre-treatment with KV-37 followed by ENZ exposure for 72 h.
- KV-37 induces cell death in prostate cancer cells via apoptosis.
- 22Rvl and LNCaPlC3 cells cultured in CSS media were treated with increasing doses of KV- 37.
- the cells were harvested and stained with AnnexinV/PI to measure apoptotic cell percentage.
- the percentage of the apoptotic cell population increased with KV- 37 concentration in a dose- and time -dependent manner (FIG. 28A).
- KV-37 Combination of KV-37 with ENZ potentiates the degree of apoptosis in prostate cancer cells.
- ENZ To determine whether the effect of KV-37 to induce apoptosis in 22Rvl cells persists in the presence of ENZ, cells were pretreated with KV-37 followed by ENZ incubation for 72 h. The cells were subjected to apoptosis analysis by AnnexinV/PI co-staining.
- FIGS. 28A to 28D KV-37 induces apoptosis in prostate cancer cells alone and potentiates the degree of apoptosis when combined with ENZ.
- FIG. 28A Dose and time-dependent increase in apoptotic cell population after treatment with KV-37 in indicated cell lines as analyzed by AnnexinV/PI co-staining.
- FIG. 28B Increase in apoptotic cell population after treatment with indicated concentrations of KV-37, ENZ and a 24 h pre-treatment combination of KV-37 and ENZ in indicated cell lines analyzed 72 h post ENZ exposure by AnnexinV/PI co- staining.
- FIG. 28C Western blot showing an increase in C-caspase3 and C-PARP levels after treatment with KV-37 at indicated concentration and time points in prostate cancer cells.
- AKR1C3 serves as a co-activator for AR (45), it was prudent to measure AR levels after treatment.
- the inventors observed that expression of AR complements AKR1C3 levels and was found to be decreased after 48 h of treatment, while at 72 h, no change in AR levels was observed in both 22Rvl and LNCaPlC3 cell lines. A consistent decline in PSA levels in a concentration- dependent manner was observed in both cell lines at 48 and 72 h treatments.
- Such findings signify a beneficial therapeutic outcome in prostate cancer after treatment with KV-37 (FIG. 29A and 29B).
- FIGS. 29A to 29D Molecular changes following KV-37 treatment.
- KV-37 Treatment of prostate cancer cells with KV-37 does not affect AR transactivation and AR nuclear localization. KV-37 does not displace [3H]-R1881 from the AR in cell binding assays suggesting that it has no affinity for the AR. Similarly, in AR luciferase reporter gene assays no effect was seen with 10 ⁇ KV-37, where 1 nM DHT is the EC50 value.
- the HeLa cell line which contains HeLa- AR3 A-P S A- (ARE)4-Lucl3) also expresses AKR1C3. Using an AKR1C3 substrate, 4- DAD, a blunted response is observed when compared with DHT, likely due to the conversion to T. Unfortunately, inclusion of KV- 37 in the assay resulted in interference, preventing conclusive data from being obtained.
- FIG. 30A to 30E KV-37 induces tumor growth inhibition in prostate cancer xenografts.
- FIG. 30C Representative images showing a reduction in tumor size after treatment with 20 mg/Kg KV-37.
- FIG. 30E Pharmacokinetic parameters determined in plasma after treatment with 20 mg/Kg KV-37.
- FIGS. 31A to 31D Indomethacin exerts a weak synergistic drug effect in LNCaPlC3 cells and KV-37 does not induce cytotoxicity alone or in combination with ENZ in WPMY-1 cells.
- Indomethacin displays diminished cytotoxic effect and weak synergistic drug action compared to the more potent inhibitor KV-37.
- KV-37 exhibits no cytotoxicity alone, or in combination with ENZ, in WPMY-1 prostate cells.
- Prostate tumors resistant to AA and ENZ are characterized by overexpression of steroidogenic enzymes where AKR1C3 is the most overexpressed (26,47,48).
- AKR1C3 is the most overexpressed (26,47,48).
- AR antagonists 13,49.
- Overexpression of AKR1C3 by stable transfection in prostate cancer cells imparted ENZ resistance which was overcome by AKR1C3 knockdown by shRNA.
- Indomethacin a weak AKR1C3 inhibitor, was also able to rescue xenografts from ENZ and AA resistance (27,28).
- KV-37 a nanomolar inhibitor of AKR1C3 with > 100-fold isoform selectivity and metabolic stability (38).
- the inventors demonstrate the synergistic effects of KV-37 with ENZ on prostate cancer cell cytotoxicity is much greater than that seen with INDO, and that the effects of KV-37 are mediated by AKR1C3 inhibition.
- KV-37 inhibits the activity of AKR1C3 in prostate cancer cells by inhibiting the conversion of A4-androstenedione to testosterone.
- KV-37 exerts a preferential cytotoxic effect in AKR1C3 overexpressing 22Rvl and LNCaPlC3 cells as compared to the low expressing LNCaP cell line and non-malignant WPMY-1 cells.
- culture of 22Rvl cells in CSS media that is devoid of androgens, upregulates AKR1C3 expression further and results in greater susceptibility to KV-37-induced cytotoxicity.
- KV-37 displayed an equally robust inhibition of cell viability in such cultures.
- the 22Rvl cell line is inherently resistant to ENZ, whereas LNCaP cells became resistant to ENZ when AKR1C3 was stably expressed in this cell line.
- Pre- treatment with KV-37 restored the sensitivity of 22Rvl cells to ENZ and provided strong synergistic effect resulting in a 200-fold reduction in chemotherapeutic dosing under conditions that mimic the CRPC disease phenotype.
- LNCaP 1C3 cells The strong synergistic effect was maintained in LNCaP 1C3 cells upon pre -treatment with KV-37. A moderate synergistic effect was also observed in these cells if KV-37 was co-administered with ENZ.
- LNCaP cells are low expressers of AKR1C3, only an additive effect was observed, an observation that may be ascribed to the general cytotoxicity of KV-37 towards cancer cells as evidenced previously in leukemic cell lines (38).
- annexinV/PI staining demonstrated a significant increase in the percentage of apoptotic cells compared to either treatment alone, and was confirmed by an increase in C-caspase3 and C-PARP levels.
- the inventors also analyzed the activity of KV-37 alone and in combination with ENZ in the non-malignant human prostate stromal cell line WPMY-1. No reduction in cell viability up to 25 ⁇ was observed, whereas no cell viability reduction in combination experiments was noted, indicating selectivity to cells overexpressing AKR1C3.
- KV-37 can be used as a monotherapy for castration-resistant prostate cancer since it retards ENZ resistant prostate cancer cell growth and induces apoptosis.
- the structurally novel AKR1C3 inhibitor KV-37 synergizes with ENZ and re-sensitizes ENZ-resistant prostate cancer cells to the action of a chemotherapeutic agent.
- Example 3 Potent and Highly Selective AKR1C3 Inhibitors with Chemotherapeutic Potentiation Effect in Acute Myeloid Leukemia (AML) and T-cell Acute Lymphoblastic Leukemia (T-ALL)
- Aldo-keto reductase 1C3 catalyzes the synthesis of 9a,l i -prostaglandin (PG) F2a and PGF2a prostanoids that sustain the growth of myeloid precursors in the bone marrow.
- AKR1C3 confers chemotherapeutic resistance to the anthracyclines: first-line agents for the treatment of leukemias. The enzyme is overexpressed in Acute Myeloid Leukemia (AML) and T-cell acute lymphoblastic leukemia (T ALL). Highly homologous isoforms AKR1C1 and AKR1C2 are required for normal steroid metabolism, inhibition of which is undesirable.
- AML Acute Myeloid Leukemia
- T ALL T-cell acute lymphoblastic leukemia
- the developed compounds exhibited a > 100-fold dose reduction index that resulted in complete resensitization of a daunorubicin-resistant AML cell line to the chemotherapeutic and reduction of the IC50 of cytarabine from 40 nM to ⁇ 1 nM.
- the natural product Baccharin, extracted from honeybee propolis has been shown to potently and selectively inhibit AKR1C3 with an IC50 of 0.11 ⁇ and a fold selectivity of 500 over AKR1C2.23
- the inventors adopted this natural structural scaffold and have previously reported the synthesis and a preliminary structure-activity relationship (SAR) for AKR1C3 inhibition.24
- SAR structure-activity relationship
- the dihydrocinnamoyloxy moiety of the hit scaffold is reported as a structural prerequisite for AKR1C3 inhibition,26 and other groups have synthesized potent AR1C3 inhibitors based on this scaffold.25
- the presence of an ester linkage provides for the hydrolytic lability of baccharin making these analogues unsuitable leads for drug discovery.
- the inventors herein report the discovery of a library of optimized compounds possessing >2800-fold selectivity for AKR1C3 inhibition, with retention of inhibitory potency in the nanomolar range. Further, the inventors demonstrate that the highly isoform selective AKR1C3 inhibitors provide potentiation up to 100-fold of the clinical chemotherapeutics daunorubicin and cytarabine across a panel of AML cell lines and in primary patient-derived T-ALL cells.
- class II-A analogues contained alcohol, methoxy, tosyl or flourine substitution on the B ring.
- class III analogues bearing boronic acid bioisosteres were synthesized. Further, the side chain substitution pattern on the A ring was modified in Class III compounds.
- class IV compounds bearing a 1,3,5-all meta side chain substituent pattern on the A ring were synthesized.
- Class IVA compounds featured changes in spacer length and substituents on the B ring.
- Figure 32 shows a SAR strategy map for the design of Baccharin derivatives of the present invention.
- Baccharin (1), compounds 8b-n (class I and I-A) and 7-7a (class III) were synthesized using a modified literature procedure.35 Nucleophilic substitution of commercially available 4-iodophenol (2) with prenyl bromide yielded the alkylated intermediate 3,36 which was subsequently esterified with an appropriately substituted acid chloride (4a-n) to yield ester intermediates (5a-n).
- Chemoselective hydrolysis of the tert-butyl ester,39 of 6a-n afforded baccharin (1) and derivatives 8b-n, whereas hydrolysis of the pinacol ester afforded boronic acid 7a (Scheme 1).
- Scheme 3 Synthesis of meta-substituted ester derivatives of class II.
- the amide bioisosteres 26a-l, 28 (class II, II-A) and 24 (class III) were accessed through Mizoroki-Heck38 reaction of commercially available 4-bromo-2-iodoaniline (20) with tert-butyl acrylate or vinyl boronic acid pinacol ester to afford 21a and 21b respectively.
- Amide formation with the appropriate acid chloride as previously described yielded amides 23a-23j and 24.
- Subsequent Suzuki-Miyaura41 cross-coupling afforded intermediates 25a-25j and 27 which were chemoselectively hydrolyzed to yield final compounds 26a-26j and 28.
- Scheme 4 Synthesis of meta-substituted amide derivatives of class II, II -A and boronic ester derivative 24 of class III.
- Commercially available 4-bromo-2-iodoaniline (29) was used as a starting material for the synthesis of compounds 33a and 33b of class III analogues.
- 3-bromo-5-iodobenzoic acid (34) was used as the common starting material for the synthesis of amide and retroinverse amide or ester analogues. Conversion of 34 to 3-bromo-5- iodoaniline (35) was achieved by modified Curtius rearrangement, 43 followed by Boc deprotection.
- Scheme 7 Synthesis of 1,3,5-meta-substituted derivatives (58a and 58b) of class IV. Structure-activity relationship.
- Class I and I-A analogues Consistent with the previous findings, baccharin exhibited an IC50 of 0.1 ⁇ for AKR1C3 inhibition with 420-fold selectivity over AKR1C2. Removal of the dihydrocinnamoyloxy group in la (drupanin) resulted in complete abrogation of AKR1C3 inhibition activity and selectivity. Replacement of the dihydrocinnamoyloxy group (compounds 8b-h) resulted in low or sub-micromolar inhibition potency for AKR1C3, without any significant loss in selectivity.
- the amide derivatives, with the exception of 15a were also significantly more promiscuous between AKR1C isoforms with only moderate to minimal selectivity for the AKR1C3 isoform over the AKR1C2 variant.
- Class II and II-A analogues The meta-substituted derivatives 19a, 26a and 26b showed enhanced AKR1C3 inhibitory activity.
- Class III analogues Boronic acid bioisosteres of carboxylic acids may be expected to demonstrate increased binding affinity to their molecular targets on account of their ability to covalently interact with side chain amino acid residues in the binding pocket, as compared to hydrogen bonding interactions provided by their carboxylic acid counterparts.45 Bioisosteric replacement of the carboxylic acid of theparent scaffold with a pinacol boronate (7) or a boronic acid (7a) decreased AKR1C3 inhibition activity by 66- and 23-fold respectively compared to 1, with a concurrent 500-fold decrease in selectivity. Removal of the prenyl moiety (24) abrogated bioactivity in agreement with theprevious findings.
- Class IV and IV-A analogues Arrangement of the side chains on the central ring in a 1,3,5 substitution pattern, greatly enhanced the selectivity for AKR1C3 inhibition over the homologus AKR1C1, 1C2 and 1C4 isoforms, as compared to 1.
- 39b all compounds possessing this substitution pattern showed inhibitory potency in the low sub-micromolar range, with the majority exhibiting activity greater than 1 (39a and 39b, 49a-h) (Tables 6 and 7).
- the meta-amide 39a displayed inhibitory activity similar to 1 with a 1.5-fold increase in selectivity over the closest isoform AKR1C2, while the selectivity over other AKR isoforms was also increased.
- the meta-ester analogue (52) decreased AKR1C3 inhibitory activity by 1.7-fold and selectivity over 1C2 by five-fold from its amide counterpart (49a), while still maintaining an increase in selectivity (1C2) of 1.25-fold from 1 and an equipotent inhibition activity for 1C3.
- the ether analogues (59a-59b) suffered great loss (18-46 fold) in inhibition selectivity over 1C2 and a decrease in inhibition activity (two - four-fold) when compared to 49a (Table 6).
- the 4- fluoro (49b) and 4-methoxy (49c) substitutions on the phenethyl side chain exhibited similar inhibition activity to 1, increasing the selectivity over 1C2 by three-fold.
- Daunorubicin an anthracycline used as front-line treatment for AML, experiences highest susceptibility to be metabolized by AKR1C3 among all of the anthracycline class of antitumor antibiotics.46
- AKR1C3 inhibitors were dosed with daunorubicin in a fixed ratio of 100: 1 (inhibitondaunorubicin).
- Cell viability reduction in combination treatments of AKR1C3 inhibitors 1, 26a, 49a and 49g with daunorubicin was not detected to a level beyond what was observed with daunorubicin treatment alone (Fig.
- FIGS. 33A-33D show the co-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (33A) 1 (33B) 26a (33C) 49a (33D) 49g at indicated concentrations. In KGla cells which are daunorubicin-resistant, none of the combinations with AKR1C3 inhibitors were able to reduce the cell viability by more than 50% (Fig.
- FIGS. 34A-34D shows the co-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (34A) 1 (34B) 26a (34C) 49a (34D) 49g at indicated concentrations.
- FIGS. 34A-34D shows the co-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (34A) 1 (34B) 26a (34C) 49a (34D) 49g at indicated concentrations.
- 35A to 35D shows the co- treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (35A) 1 (35B) 26a (35C) 49a (35D) 49g at indicated concentrations.
- FIGS. 36A to 36D shows the 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (36A) 1 (36B) 26a (36C) 49a (36D) 49g at indicated concentrations. The high AKR1C3 expressing KGla cells displayed a complete abrogation of cell viability at all inhibitor combinations with daunorubicin that reached as low as 0.1 ⁇ inhibitor and 0.001 ⁇ daunorubicin (Fig. 37A-37D).
- FIGS. 37A to 37D shows the 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (37 A) 1 (37B) 26a (37C) 49a (37D) 49g at indicated concentrations. A complete cell viability reduction was also noted in THP-1 cells at 0.01 and 0.1 ⁇ daunorubicin combination with all AKR1C3 inhibitors tested and a cell viability reduction of >50% was seen at 0.001 ⁇ daunorubicin combination as compared to no cell viability reduction at 0.001 ⁇ daunorubicin alone (Fig. 38A-38D).
- FIGS. 38A to 38D shows the 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (38A) 1 (38B) 26a (38C) 49a (38D) 49g at indicated concentrations.
- AKR1C3 inhibition will drive the production of PGJ2 series prostanoids that will eventually activate pro-apoptotic signaling and will be instrumental in providing a synergistic drug effect with the chemotherapeutic AraC.
- 40A to 40D shows the co-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h co-treatment of AraC with (40 A) 1 (40B) 4 (40C) 6 (40D) 7 at indicated concentrations.
- FIGS. 41A to 41D shows the co-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h co-treatment of AraC with (41A) 1 (41B) 4 (41C) 6 (4 ID) 7 at indicated concentrations.
- FIGS. 43A to 43D shows the 24 h Pre-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (43 A) 1 (43B) 26a (43C) 49a (43D) 49g at indicated concentrations.
- FIGS. 44A to 44D shows the 24 h Pre-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (44A) 1 (44B) 26a (44C) 49a (44D) 49g at indicated concentrations.
- the DRI was greater in the relapsed T-ALL cells (COG- 317) which have been shown to overexpress the AKR1C3 target that imparts drug-resistant properties at relapse.
- the degree of drug synergism increased even further among pre-treatment experiments (DRI > 100) and a near complete abrogation of cell viability was noted in COG-317 cells (Fig. 45C, 45D and table insert).
- Both co-treatment and pre-treatments of 49a with AraC showed a very strong synergism in COG-317 cells (DRI > 100) (Fig. 45A, 45C and table insert).
- the effect increased from DRI of 15.7 among co-treatments to >100 in pre-treatment experiments (Fig. 45B, 45D and table insert).
- FIGS. 45A to 45D shows the synergistic effect of compound 49a with daunorubicin in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and daunorubicin in (45A) COG-317 (45B) COG-329 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of 49a in (45C) COG-317 (45D) COG-329 cells.
- FIGS. 45E to 45G shows the treatment of COG T-ALL cells with 49a and chemotherapeutics. Percentage cell viability after 72 h treatment of AML cells with (45E) 49a (45F) daunorubicin and (46G) AraC at indicated concentration and time points.
- FIGS. 46A to 46D shows the synergistic effect of compound 49a with AraC in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and AraC in (A) COG-317 (B) COG-329 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of 49a in (C) COG-317 (D) COG-329 cells.
- FIGS. 46E and 46F shows the combination treatment of BMMNC cells with 49a and chemotherapeutics. Percentage cell viability of BMMNC cells after pre-treatment with 49a followed by 72 h incubation with (46E) AraC and (46F) daunorubicin at indicated concentrations.
- AKR1C3.NADP+- inhibitor complexes Analysis of crystal structures of AKR1C3.NADP+- inhibitor complexes have revealed that the ligand binding site can be dissected into five subsites: oxyanion site (consisting of catalytic residues Tyr55, Hisl l7 and cofactor NADP+), the steroid channel for the binding of steroid ligands, and three sub-pockets (SPs: SP1, SP2 and SP3 to accommodate other ligands).47, 54 In the AKR1C3.NADP+.
- Baccharin complex model (Fig. 2.4), 23 the carboxylate group on cinnamic acid was predicted to be bound in the SP1 pocket and could form hydrogen bonds with the side chain of Serl l8, which contributes to strong binding affinity for 1.
- polar amino acids e.g. Ser308 and Tyr319
- these polar interactions also increase the selectivity of baccharin for AKR1C3 over the other AKR1C isoforms where the amino acids in the corresponding positions are Phel l8, Leu308 and Phe319 in the SP1 pocket.30, 54
- the docked model predicted that the benzyl moiety of the 4-dihydrocinnamoyloxy group was located in the SP3 pocket and could form hydrophobic interactions with the side chain of Gln222 or Phe306 to provide the high binding affinity .23
- polar substitutions e.g.
- the inhibitory potency increased in comparison to 1.
- the presence of either a carbonyl or carboxylate group is required to anchor many ligands to the oxyanion site, because it can form a strong hydrogen-bond with Tyr55 and Hisl 17 and bring the ligands into close proximity of the nicotinamide head group of the cofactor.54
- the carbonyl group on the 4-dihydrocinnamoyloxy group of baccharin was found to be close to the oxyanion site and could form a H-bond interaction with Tyr55 and Hisl 17, which could contribute to the high inhibitory activity of baccharin.
- FIG. 47 shows a Baccharin SAR map.
- FIGS. 48A to 48D show the treatment of HL-60 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of HL-60 cells with (48A) 1 (48B) 26a (48C) 49a (48D) 49g at indicated concentrations and time points.
- FIGS. 49A to 49D show the treatment of KGla cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of KGla cells with (49 A) 1 (49B) 26a (49C) 49a (49D) 49g at indicated concentrations and time points.
- FIGS. 50A to 50D shows the treatment of THP-1 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of THP-1 cells with (50A) 1 (50B) 26a (50C) 49a (50D) 49g at indicated concentrations and time points.
- AKR1C3 inhibitors derived from modification of a natural product have enhanced biological stability and exhibit extremely selective and potent activity. Proof-of-concept that AKR1C3 inhibitors derived from baccharin have a synergistic effect sensitizing AML and T-ALL cells to the chemotherapeutic effects of daunorubicin and AraC is demonstrated.
- AKR1C3 inhibitors Treatment of the non-toxic AKR1C3 inhibitors in in vitro models of acute myeloid leukemia representing various French-American-British (FAB) sub-types of AML (MO, M3 and M5), along with co-administration of a range of clinical chemotherapeutics, results in a synergistic drug action, potentiating cytotoxicity and reducing the IC50 value. This is contrary to recent reports detailing selective AKR1C3 inhibitors do not perform an adjuvant role as compared to pan- AKR1C isoform inhibitors.57 This observation is further extended to the primary patient-derived T-ALL cells where a very strong potentiation of the effects of daunorubicin and AraC was established.
- FAB French-American-British
- AKR1C3 inhibitors may yield powerful adjuvant agents for the synergistic treatment of leukemia with chemotherapeutic drugs, especially since favorable toxicity and pharmacokinetic properties can exist within the baccharin structure.
- potent and selective inhibition of AKR1C3 results in potentiation of the clinical chemotherapeutics: etoposide, daunorubicin and AraC in multiple AML cell lines.
- the identified highly potent and selective derivatives represent valuable lead compounds to understand the structural features required for AKR1C3 inhibitory activity and selectivity, and how these properties interrelate to further theunderstanding of the role which AKR1C3 plays to enable potentiation of chemotherapeutic effect within AML pathophysiology. Further development of the inhibitors describe herein represents a significant drug discovery opportunity for the identification of potent adjuvants to enhance the therapeutic index of chemotherapeutics, in the hope of availing this treatment regime to pediatric and geriatric AML patients.
- Enzyme Inhibition Assay A detailed account of the chemistry procedures and characterizations (1H, 13C NMR, and HRMS) was performed. The enzyme inhibition screen for all compounds was perfbmed in collaboration with the Penning lab at the University of Pennsylvania. (S)-(+)- 1,2,3, 4-tetrahydro-l- naphthol (S-tetralol) was purchased from Sigma-Aldrich (St. Louis, MO). Nicotinamide adenine dinucleotide (NAD+) and nicotinamide adenine dinucleotide phosphate (NADP+) were purchased from Roche Diagnostics (Indianapolis, IN).
- AKR1C3 and AKR1C2 were prepared and purified as previously described.28
- the specific activities of AKR1C3 and AKR1C2 for the oxidation of S-tetralol are 2.0 and 1.5 ⁇ min-1 mg-1, respectively.
- Assay of enzyme activity The dehydrogenase activities of AKR1C3 and AKR1C2 were determined by measuring the UV absorption of NADH formation at 340 nm using Beckman DU640 spectrophotometer.
- a typical assay solution contained 100 mM potassium phosphate pH 7.0, 2.3 mM NAD+, 3.0 mM (S)- (+)-l,2,3,4-tetrahydro-l-naphthol (S-tetralol), 4% acetonitrile (v/v).
- the mixtures were incubated at 37 °C for 3 min followed by adding a serial dilution of AKR1C3 or AKR1C2 solution to a final volume of 1 mL to initiate the reaction. After continuously monitoring for 5 min, the increase in UV absorption using different concentrations of enzyme were recorded to calculate the initial velocity and determine enzyme specific activity.
- IC50 value determination The inhibitory potency for each compound was represented by IC50 value and measured as described before 29-31.
- the IC50 value of baccharin and baccharin analogs was determined by measuring their inhibition on the NADP+ dependent oxidation of S-tetralol catalyzed by AKR1C3 or AKR1C2.
- the concentration of S-tetralol used in this assay for AKR1C3 and AKR1C2 was 165 ⁇ and 15 ⁇ respectively, which was equal to the Km value for each enzyme isoform in order to make a direct comparison of IC50 values.
- HL-60 ATCC® CCL-240TM
- KGla ATCC® CCL-246.1TM
- THP-1 ATCC® TIB-202TM
- IMDM Isocove's Modified Dulbecco's Media
- FBS fetal bovine serum
- penicillin/streptomycin 16% was used to culture HL-60 and KGla cells
- THP-1 cells were cultured using Roswell Park Memorial Institute (RPMI)-1640 medium, supplemented with 20% fetal bovine serum (FBS), 0.05 mM 2-mercaptoethanol and penicillin/streptomycin (1%).
- RPMI Roswell Park Memorial Institute
- Cells were maintained at a density of 0.1-1 x 106 cells/mL under 5% C02 at 37 oC.
- To screen the test compounds cells were seeded at a density of 0.1 x 106 cells/mL in 96- well plates containing 100 ⁇ L cell suspension per well.
- Stock solutions of the test compounds, etoposide, daunorubicin and cytarabine (AraC) were prepared in DMSO. Cells were treated at the indicated concentrations of test compounds with or without the chemotherapeutic agents, limiting the final DMSO concentration to less than 1%.
- MTS reagent CellTiter 96® AQueous One Solution Reagent
- Protein samples (40 ⁇ g ) containing loading dye (7 ⁇ .) were loaded onto 12% SDS polyacrylamide gel and electrophoresed (80 V, 2 hr). Transferred onto a PVDF membrane overnight (25 V, 4 oC). The membrane was blocked with 5 % non-fat milk (1 h) and probed with human anti-AKRlC3 mouse monoclonal antibody (1:500, R&D Systems, MAB7678) and corresponding horseradish peroxidase (HRP) conjugated anti-mouse secondary antibody followed by immunodetection using VersaDocTM (MP 5000). Membrane was stripped and re-probed for ⁇ -actin (1 :5000, Sigma-Aldrich, A5441). Quantity One® software was used to analyze the band intensities and fold change in AKR1C3 enzyme expression was determined based on ⁇ -actin controls.
- HRP horseradish peroxidase
- solvents and reagents and solvents were purchased from Fisher Scientific (Houston, TX), Sigma Aldrich (Milwaukee, WI) or for Prenyl boronic acid pinacol ester, Santa Cruz Biotechnology (Dallas, TX) and were used without further purification except as indicated.
- Anhydrous solvents were purchased from Across Organics and stored under and atmosphere of dry nitrogen over molecular sieves.
- tert-butyl(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-[(3-phenylpropanoyl)oxy]phenyl]prop-2-enoate (6a): To a solution of 5a (490 mg, 1.16 mmol) in anhydrous toluene (7 ml) was added PPh3 (46 mg, 0.175 mmol), Pd(OAc)2 (47 mg, 0.09 mmol) and NEt3 (0.245 ml, 1.75 mmol) the mixture was sitirred and t-Bu acrylate (0.275 ml, 1.75 mmol) was added to the flask which was refluxed overnight.
- PPh3 46 mg, 0.175 mmol
- Pd(OAc)2 47 mg, 0.09 mmol
- NEt3 0.245 ml, 1.75 mmol
- tert-butyl (2E)-3-[4-(acetyloxy)-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoate (6b): To a solution of 5b (350 mg, 1.0 mmol) in dry toluene (6.5 mL) was added PPh3 (42 mg, 0.2 mmol), Pd(OAc)2 (47.5 %, 47 mg, 0.1 mmol) and NEt3 (0.2 mL, 1.6 mmol) the mixture was sitirred for 10 mins and tert-butyl acrylate (0.2 mL, 1.6 mmol) was added to the flask which was refluxed overnight.
- tert-butyl(E)-3-(4-((3-(2-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (6k): To a solution of 5k (430 mg, 1 mmol) in dry toluene (6 mL) was added PPh3 (78 mg, 0.3 mmol), Pd(OAc)2 (47.5 %, 70 mg, 0.15 mmol) and NEt3 (0.4 mL, 3.0 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.4 mL, 2.8 mmol) added and the reaction refluxed overnight.
- tert-butyl(E)-3-(4-((3-(4-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (61): To a solution of 51 (320 mg, 0.7 mmol) in dry toluene (8 mL) was added PPh3 (53 mg, 0.2 mmol), Pd(OAc)2 (23 mg, 0.1 mmol) and NEt3 (0.2 mL, 1.5 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.2 mL, 1.5 mmol) added and the reaction refluxed overnight.
- tert-butyl(E)-3-(4-((3-(3-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acty (6m): To a solution of 5m (280 mg, 0.6 mmol) in dry toluene (8 mL) was added PPh3 (39 mg, 0.15 mmol), Pd(OAc)2 (47.5%, 35 mg, 0.08 mmol) and NEt3 (0.2 mL, 1.5 mmol) the mixture was sitirred for 10 min and tert-butyl acrylate (0.2 mL, 1.5 mmol) added and the reaction refluxed overnight.
- tert-butyl(E)-3-(4-((3-(2-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (6n): To a solution of 5n (320 mg, 0.7 mmol) in dry toluene (8 mL) was added PPh3 (78 mg, 0.3 mmol), Pd(OAc)2 (34 mg, 0.15 mmol) and NEt3 (0.4 mL, 3 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.4 mL, 2 mmol) added and the reaction refluxed overnight.
- tert-butyl (2E)-3-(4-amino-3-bromophenyl)prop-2-enoate (11) To a solution of (10) (570 mg, 1.9 mmol) in dry toluene (8 mL), was added PPh3 (65.5 mg, 0.2 mmol) and Pd(OAc)2 (47.5 %, 60 mg, 0.1 mmol). tert-Butyl Acrylate (370 ⁇ ., 2.5 mmol) and NEt3 (420 ⁇ ., 3.0 mmol) were added and the flask was stirred at reflux overnight.
- tert-butyl (E)-3-(5-bromo-2-(3-phenylpropanamido)phenyl)acrylate (31): To a solution of 30 (300 mg, 0.7 mmol) in dry toluene (10 mL) was added PPh3 (26 mg, 0.1 mmol) and Pd(OAc)2 (12 mg, 0.05 mmol). tert-butyl acrylate (160 ⁇ , 1 mmol) and NEt3 (280 ⁇ , 2 mmol) were added and the reaction was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc gradient 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a white solid (210 mg, 0.5 mmol, 70 %).
- N-(3-bromo-5-iodophenyl)-3-phenylpropanamide (36): To a solution of 35 (380 mg, 1.3 mmol) in dry DCM (12 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-phenyl propanoyl chloride (320 mg, 2 mmol) in DCM (2 mL) and NEt3 (0.4 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a white solid (430 mg, 1 mmol, 78%).
- tert-butyl (E)-3-(3-bromo-5-(3-phenylpropanamido)phenyl)acrylate (37): To a solution of 36 (123 mg, 0.3 mmol) in dry toluene (5 mL) was added PPh3 (8 mg, 0.03 mmol) and Pd(OAc)2 (4 mg, 0.01 mmol). tert-butyl acrylate (60 ⁇ , 0.4 mmol) and NEt3 (70 ⁇ , 0.5 mmol) were added and the reaction was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc gradient 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a brown oil (110 mg, 0.25 mmol, 91%).
- 3-bromo-5-iodo-N-phenethylbenzamide (41a): To a solution of 3-bromo-5-iodo benzoic acid (500 mg, 1.5 mmol) in dry toluene (10 mL) was added SOC12 (290 ⁇ ., 4 mmol) and the mixture was refluxed overnight. The reaction vessel was cooled to room temperature and the solvent was evaporated in vacuo. The resultant brown oil was used further without purification. To a solution of 3-bromo-5-iodo benzoyl chloride in DCM (10 mL) was added DMAP (26 mg, 0.2 mmol) and the flask was purged with nitrogen.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- Emergency Medicine (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Hematology (AREA)
- Oncology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention includes methods and compositions that inhibit AKR1C3 enzymatic activity and consequently reduces androgen receptor (AR) transactivation, AR and prostate specific antigen (PSA) expression levels in, for example, prostate cancer, castration-resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
Description
HIGHLY SELECTIVE AKR1C3 INHIBITORS AND METHODS OF USE THEREOF
TECHNICAL FIELD OF THE INVENTION
The present invention relates generally to methods and compositions used for inhibiting the biological activity of an aldo-keto reductase family 1, member C3 (AKR1C3) polypeptide.
BACKGROUND OF THE INVENTION
Without limiting the scope of the invention, its background is described in connection with compositions and methods of using the same as Aldo-keto reductase 1C3 (AKR1C3) specific inhibitors to modulate cellular or tissue proliferation, e.g., prostate cancer and/or castrate-resistant prostate cancer. Prostate cancer is the second most common cancer in men, and twenty -percent of all cases develop into castrate- resistant prostate cancer, which often presents with metastatic bone disease and is always fatal. These tumors are synthesized androgens, independently of the testes and there exists a great need in the art for such treatments. All prior AKR1C3 inhibitors have had poor selectivity between AKR1C isozymes resulting in undesirable side effects. As a result no AKR1C3 inhibitor has reached clinical trial, in part, due to lack of selectivity.
U.S. Patent Application Publication No. 2014/0371261, entitled, "Indomethacin analogs for the treatment of castrate-resistant prostate cancer," discloses compositions for inhibiting a biological activity of an aldo-keto reductase family 1, member C3 (AKR1C3) polypeptide. The compositions are indomethacin (2-[l-(4-chlorobenzoyl)-5-methoxy-2-methylindol-3-yl]acetic acid) derivatives that are AKR1 C3- specific inhibitors. Although the compositions showed a selective interaction with AKR1C3, they also interacted with AKR1C1 and AKR1C2. AKR1C1 and AKR1C2 are involved in hormonal levels and balance and blocking of these affects those levels.
SUMMARY OF THE INVENTION
Aldo-keto reductase 1C3, also known as type 5 17 β-hydroxy steroid dehydrogenase, is responsible for intratumoral androgen biosynthesis and contributes to the development of castration resistant prostate cancer (CRPC) phenotype and an eventual therapeutic failure. Significant upregulation of AKR1C3 is observed in CRPC patient samples and derived cell lines. Being a downstream catalytic enzyme for the synthesis of testosterone and 5-dihydrotestosterone (DHT), AKR1C3 represents a potential therapeutic target to manage CRPC and combat the emergence of resistance to clinically employed first line therapeutics. The present invention provides novel, potent, isoform selective and hydrolytically stable small-molecule AKR1C3 inhibitor compounds that reduces prostate cancer cell growth in vitro and sensitizes CRPC cell lines towards enzalutamide cytotoxicity. The present invention demonstrates that treatment with these inhibitors selectively induce cytotoxicity in a wide variety of cancer cells. Moreover, these inhibitors cause inhibition of AKR1C3 enzymatic activity and consequently reduces androgen receptor (AR) transactivation, AR and prostate specific antigen (PSA) expression levels in CRPC cell lines. Combination studies of the inhibitors with enzalutamide and other chemotherapies reveal a synergistic drug interaction that induces cytotoxicity of cancer cells via apoptosis. Taken
together, the results demonstrate a novel therapeutic strategy for the treatment of, e.g., prostate cancer, castration-resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia. In the case of a castration-resistant prostate cancer disease phenotype, it invariably develops in prostate cancer patients following initial treatment with AR antagonists such as enzalutamide.
In one embodiment, the present invention includes an aldo-keto reductase family 1, member C3 (AKR1C3) inhibitor having one of the following structures:
derivatives thereof. In one aspect, the inhibitor is Class I or IA and
In another aspect, the inhibitor is:
In another aspect, the inhibitor is
wherein R is:
In another aspect, the inhibitor is:
, wherein Rl is
, and wherein R2 is:
In another aspect, the inhibitor is:
, w ere n Rl s:
In another aspect, the inhibitor is formulated into a therapeutic effective amount sufficient to reduce or eliminate cancer cells. In another aspect, the inhibitor further comprises a chemotherapeutic agent, wherein the inhibitor and the chemotherapeutic have a synergistic effect against cancer cells. In another aspect, the inhibitor further comprises providing the inhibitor in an amount that is therapeutically effective to inhibit or reduce cellular or tissue proliferation selected from prostate cancer, castration- resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
In another embodiment, the present invention includes a method of modulating cellular or tissue proliferation comprising the steps of providing a therapeutic effective amount of an inhibitor that is selective for the enzyme AKR1C3 over its isoforms wherein the compound has the formula:
derivatives thereof.
In another aspect, the inhibitor is
In another aspect, the inhibitor is:
, wherein R is:
In another aspect, the inhibitor is:
, wherein Rl is
In another aspect, the inhibitor is:
, w ere n Rl s:
In another aspect, the inhibitor is formulated into a therapeutic effective amount sufficient to reduce or eliminate cancer cells. In another aspect, the inhibitor further comprises a chemotherapeutic agent, wherein the inhibitor and the chemotherapeutic have a synergistic effect against cancer cells. In another aspect, the inhibitor further comprises providing the inhibitor in an amount that is therapeutically effective to inhibit or reduce cellular or tissue proliferation selected from prostate cancer, castration- resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia. In another aspect, the prostate cancer and/or castrate-resistant prostate cancer, breast cancer, leukemia is resistant to chemotherapeutic agents. In another aspect, the AKR1C3 inhibitor is synergistic with a chemotherapeutic.
The present invention provides a composition that selectively inhibits an aldo-keto reductase family 1 enzyme family in a selective manner that selects the AKR1C3 isoform comprising a therapeutic effective amount of a compound or derivatives thereof.
The present invention provides a method of modulating cellular or tissue proliferation comprising the steps of: providing a therapeutic effective amount of an inhibitor that is selective for the enzyme AKR1C3 over its isoforms wherein the compound is one or more Class I, II, III, or IV baccharin derivatives. The cellular or tissue proliferation comprises prostate cancer and/or castrate-resistant prostate cancer, breast cancer, leukemia, ovarian cancer, endometrial cancer, or a combination thereof.
The present invention provides a method of preventing the development of chemotherapeutic resistant cancers comprising the steps of: providing a therapeutic effective amount of a compound that possess selectivity for the enzyme AKR1C3 over its isoforms wherein the compound is one or more Class I, II, III, or IV baccharin derivatives.
A method of treating, ameliorating or preventing cancer in a subject in need thereof, said method comprising administering to said subject a composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one Class I, II, III, or IV baccharin derivative.
BRIEF DESCRIPTION OF THE DRAWINGS
For a more complete understanding of the features and advantages of the present invention, reference is now made to the detailed description of the invention along with the accompanying figures and in which:
FIGS. 1A-1D show images of some embodiments of the compounds of the present invention.
FIGS. 2A-2C are tables showing prostate cancer cell viability was observed upon treatment with baccahrin.
FIGS. 3A-3F are graphs showing KV-37 induces potent cytotoxic activity in multiple CaP cell lines.
FIGS. 4A-4F show pre-treatment with KV-37 sensitizes LNCap and 22Rvl cells to ENZ cytotoxicity.
FIGS. 5A-5E are images showing KV-37 induces apoptosis in 22Rvl cells.
FIG. 6 is an image showing downstream molecular changes after KV-37 treatment in 22Rvl cells.
FIG. 7 is an image showing KV-37 treatment of 22Rvl cells downregulated AKR1C3.
FIGS. 8A-8B are images showing molecular changes following a combination treatment of KV-37 and ENZ in 22Rvl cells after 72 hour incubation.
FIG. 9 is a graph of the compound KV-60-5(l) showing no cytotoxicity to LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hour exposure at 1 μΜ concentration. IC50 value of KV-60-5(l) was determined to be 100 μΜ.
FIG. 10 is a graph of the compound KV-60-5(l) showing significant potentiation effect of the clinical antineoplastic enzalutamide in LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hours.
FIG. 11 in an image of the modelling of KV-60-5a docked into the AKR1C3 enzyme.
FIG. 12 shows a synthetic scheme for the inhibitor.
FIG. 13 shows representative derivative compounds with a modification of phenyl (6).
FIG. 14 shows representative derivative compounds with a modification of Dihydrocinnamyloxy (6).
FIG. 15 shows representative derivative compounds with a Conversion of Ester to Amide Group (5).
FIG. 16 shows representative derivative compounds with a Positional Switch of Prenyl & Dihydrocinnamyloxy Group (15).
FIG. 17 shows representative derivative compounds with a Positional Switch of Dihydrocinnamoyloxy Group Analogs (3).
FIG. 18 shows representative derivative compounds a Switch of Amide Bond Direction-Reverse Amides (10).
FIG. 19 shows representative derivative compounds with a Modification of Carboxylate Group (2). FIGS. 20A to 20K show that KV-37 induces potent cytotoxic activity in multiple prostate cancer cell lines. (FIG. 20 A) Structure of KV-37. Percentage cell viability of 22Rvl cells cultured in (FIG. 20B) normal media (FIG. 20C) CSS media (FIG. 20D) normal media supplemented with 10 nM Δ4- androstenedione and (FIG. 20E) CSS media supplemented with 10 nM A4-androstenedione. Percentage cell viability of LNCaP cells cultured in (FIG. 20F) normal media and (FIG. 20G) CSS media (FIG. 20H). FIG. 201 shows the IC50 values of KV-37 determined in 22Rvl and LNaCP cells. Data shown as mean ± SD of at least three independent experiments, n=6. The competitive inhibition of NADPH dependent reduction of A4-androstene-3,17-dione mediated by AKR1C3 by KV-37. (A) KV-37 acts as a competitive Inhibitor in the conversion of [3H]A4-androstene-3,17-dione to testosterone. Increasing concentrations of KV-37 (0-10 μΜ) were used to inhibit the NADPH dependent conversion of fixed concentrations of [3H]A4-androstene-3,17-dione to testosterone catalyzed by recombinant AKR1C3 as measured by radiochromatography.
FIGS. 20J to 220K show that 22Rvl and LNCaPlC3 cells overexpress AKR1C3 whereas LNCaP and WPMY-1 exhibit low AKR1C3 levels. (FIG. 20J) Relative expression of AKR1C3 across prostate cancer cell lines. (FIG. 20K) Culture of 22Rvl cells in CSS media upregulates AKR1C3 expression levels. (NM, normal media; CSS, charcoal stripped serum media)
FIGS. 21A to 21C shows that KV-37 inhibits conversion of [3H]A4-androstene-3,17-dione to testosterone in LNCaP-lC3 cells. LNCaP-lC3 cells (1.5 x 106 cells per well) platted in phenol red-free RPMI, 5% CSS-FBS, 2 mM L-glutamine, 1% P/S and were with DMSO or 10 μΜ KV-37 for 30 min at
37 °C. After the preincubation, 100 nM [3H]A4-androstene-3,17-dione final concentration was added to the wells and cells were incubated for 24 h. FIG. 21A is solvent control; (FIG. 21B) contains 10 μΜ KV- 37 and (FIG. 21C) are LNCaP control cells treated with DMSO. The peak at 90 mm is androsterone and the peak at 120 mm is testosterone liberated after β-glucronidase treatment. The data reveal almost complete inhibition of AKR1C3 mediated testosterone production.
FIGS. 22A to 22C shows the metabolic instability of (FIG. 22A) Baccharin and (FIG. 22B) ester derivative KV-32 on incubation with mouse S9 fractions. (FIG. 22C) Half-life of baccharin and derivative AKR1C3 inhibitors upon incubation with human S9 liver fractions.
FIGS. 23A to 23C show that Baccharin does not exert cytotoxicity on prostate cancer cell lines. Percentage cell viability of (FIG. 23A) LNCaP, (FIG. 23B) 22Rvl and (FIG. 23C) LNCaPlC3 cells cultured in CSS media after baccharin treatment at indicated concentration and time points. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 24A to 24D Percentage cell viability of ENZ at indicated time points in (A) LNCaP cells cultured in indicated growth media (B) 22Rvl cells cultured in CSS media (C) 22Rvl cells cultured in CSS media supplemented with 10 nM A4-androstene-3,17-dione (D) 22Rvl cells cultured in normal media supplemented with 10 nM A4-androstene-3,17-dione. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 25A to 251 show that pre-treatment with KV-37 sensitizes prostate cancer cell lines to ENZ cytotoxicity. Percentage cell viability of prostate cancer cells when pre-treated with KV-37 for 24 h followed by ENZ treatment at indicated concentrations and time points in (FIG. 25A) 22Rvl cells cultured in normal media (FIG. 25B) and (FIG. 25C) 22Rvl cells cultured in CSS media (FIG. 25D) 22Rvl cells cultured in normal media supplemented with 10 nM A4-androstenedione (FIG. 25E) and (FIG. 25F) 22Rvl cells cultured in CSS media supplemented with 10 nM A4-androstenedione (FIG. 25G) LNCaP cells cultured in normal media and (FIG. 25H) LNCaP cells cultured in CSS media. (FIG. 251) Quantification of the degree of synergism. Combination index (CI) and dose reduction index (DRI) values for KV-37 and ENZ combination treatments in prostate cancer cells. CI and DRI indices generated using CompuSyn software. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 26A to 26H show that co-treatments of KV-37 with ENZ do not exert synergistic cytotoxic effects in LNCaP and 22Rvl cells. Percentage cell viability of prostate cancer cells after combination treatment with KV-37 and ENZ in (FIG. 26A- FIG. 26C) LNCaP cells at indicated time points cultured in indicated growth media (FIG. 26D- FIG. 26E) 22Rvl cells cultured in indicated growth media at 72 h and (FIG. 26F- FIG. 26H) 22Rvl cells at indicated time points cultured in indicated growth media supplemented with 10 nM A4-androstene-3,17-dione. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 27A to 27D shows that AKR1C3 overexpression in LNCaP cells (LNCaPlC3) increases the cytotoxic activity of KV-37 and induces resistance to ENZ that is overcome by pre-treatment with KV- 37. Percentage cell viability of LNCaPlC3 cultured in CSS media after (FIG. 27A) KV-37 treatment at indicated time points and concentrations (table insert showing the IC50 values) (FIG. 27B) ENZ treatment at indicated time points and concentrations (FIG. 27C) co-treatment with KV-37 and ENZ for 72 h and (FIG. 27D) pre-treatment with KV-37 followed by ENZ exposure for 72 h. Table insert showing the parameters for quantifying the degree of synergism. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 28A to 28D show that KV-37 induces apoptosis in prostate cancer cells alone and potentiates the degree of apoptosis when combined with ENZ. (FIG. 28A) Dose and time-dependent increase in apoptotic cell population after treatment with KV-37 in indicated cell lines as analyzed by AnnexinV/PI co-staining. (FIG. 28B) Increase in apoptotic cell population after treatment with indicated concentrations of KV-37, ENZ and a 24 h pre-treatment combination of KV-37 and ENZ in indicated cell lines analyzed 72 h post ENZ exposure by AnnexinV/PI co- staining. (FIG. 28C) Western blot showing an increase in C-caspase3 and C-PARP levels after treatment with KV-37 at indicated concentration and time points in prostate cancer cells. (FIG. 28D) Western blot showing a greater increase in C-caspase3 and C-PARP levels with a 24 h pre-treatment combination of KV-37 and ENZ in prostate cancer cells analyzed 72 h post ENZ exposure. Data shown as mean ± SD of at least three independent experiments, n=3. *, p<0.05; **, pO.01; ****, pO.0001; ns, non-significant.
FIGS. 29 A to 29D show that Molecular changes following KV-37 treatment. Treatment with KV-37 alone at indicated concentration and time points in (FIG. 29 A) 22Rvl and (FIG. 29B) LNCaPlC3 cells cultured in CSS media. Treatment with indicated concentrations of KV-37, ENZ and a 24 h pre-treatment combination of KV-37 and ENZ in (FIG. 29C) 22Rvl cells and (FIG. 29D) LNCaPlC3 cells cultured in CSS media analyzed 72 h-post ENZ exposure.
FIGS. 30A to 30E show that KV-37 induces tumor growth inhibition in prostate cancer xenografts. Reduction in (FIG. 30A) prostate tumor volume and (FIG. 30B) prostate tumor weight in 22Rvl murine xenografts after treatment with 20 mg/Kg KV-37. (FIG. 30C) Representative images showing a reduction in tumor size after treatment with 20 mg/Kg KV-37. FIG. 30D shows the mice body weight versus days post implantation. FIG. 30E shows the pharmacokinetic parameters determined in plasma after treatment with 20 mg/Kg KV-37.
FIGS. 31A to 3 ID Indomethacin exerts a weak synergistic drug effect in LNCaPlC3 cells and KV-37 does not induce cytotoxicity alone or in combination with ENZ in WPMY-1 cells. Percentage cell viability of LNCaPlC3 cells after (FIG. 31 A) treatment with indicated concentrations of indomethacin for 96 h and (FIG. 3 IB) 24 h pre-treatment with indomethacin followed by ENZ treatment for 72 h. Percentage cell viability of WPMY-1 cells after (FIG. 31C) treatment with indicated concentrations of KV-37 for 96 h and (FIG. 3 ID) 24 h pre-treatment with KV-37 followed by ENZ treatment for 72 h.
Table insert showing the quantification of the degree of synergism. Data shown as mean ± SD of at least three independent experiments, n=6.
FIG. 32 shows the SAR strategy map for the design of Baccharin derivatives.
FIGS. 33A to 33D show co-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (33A) 1 (33B) 26a (33C) 49a (33D) 49g at indicated concentrations.
FIGS. 34A to 34D show Co-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (34A) 1 (34B) 26a (34C) 49a (34D) 49g at indicated concentrations.
FIGS. 35A to 35D show Co-treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (35A) 1 (35B) 26a (35C) 49a (35D) 49g at indicated concentrations.
FIGS. 36A to 36D show 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (36A) 1 (36B) 26a (36C) 49a (36D) 49g at indicated concentrations.
FIGS. 37A to 37D show 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (37 A) 1 (37B) 26a (37C) 49a (37D) 49g at indicated concentrations.
FIGS. 38A to 38D show 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (38A) 1 (38B) 26a (38C) 49a (38D) 49g at indicated concentrations.
FIGS. 38E to 38F show Treatment of AML cells with chemotherapeutics. Percentage cell viability after 72 h treatment of AML cells with (38E) daunorubicin and (38F) AraC at indicated concentrations.
FIGS. 39A to 39D show that co-treatment of AKR1C3 inhibitors with AraC in HL-60 cells. Percentage cell viability after 72 h co-treatment of AraC with (39A) 1 (39B) 26a (39C) 49a (39D) 49g at indicated concentrations.
FIGS. 40A to 40D show that co-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h co-treatment of AraC with (40A) 1 (40B) 4 (40C) 6 (40D) 7 at indicated concentrations.
FIGS. 41A to 41D show that co-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h co-treatment of AraC with (41 A) 1 (4 IB) 4 (41C) 6 (4 ID) 7 at indicated concentrations.
FIGS. 42A to 42D show the effect of 24 h Pre-treatment of AKR1C3 inhibitors with AraC in HL-60 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (42A) 1 (42B) 26a (42C) 49a (42D) 49g at indicated concentrations.
FIGS. 43 A to 43D show the effect of 24 h Pre-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (43A) 1 (43B) 26a (43C) 49a (43D) 49g at indicated concentrations.
FIGS. 44A to 44D show the effect of 24 h Pre-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (44A) 1 (44B) 26a (44C) 49a (44D) 49g at indicated concentrations.
FIGS. 45A to 45D show the synergistic effect of compound 49a with daunorubicin in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and daunorubicin in (45 A) COG-317 (45B) COG-329 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of 49a in (45C) COG-317 (45D) COG-329 cells.
FIGS. 45E to 45G shows treatment of COG T-ALL cells with 49a and chemotherapeutics. Percentage cell viability after 72 h treatment of AML cells with (45E) 49a (45 G) daunorubicin and (45H) AraC at indicated concentration and time points.
FIGS. 46A to 46D show the synergistic effect of compound 49a with AraC in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and AraC in (46 A) COG-317 (46B) COG-329 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of 49a in (46C) COG- 317 (46D) COG-329 cells.
FIGS. 46E and 46F Combination treatment of BMMNC cells with 49a and chemotherapeutics. Percentage cell viability of BMMNC cells after pre-treatment with 49a followed by 72 h incubation with (46E) AraC and (46F) daunorubicin at indicated concentrations.
FIG. 47 shows a Baccharin SAR map
FIGS. 48A to 48D show the treatment of HL-60 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of HL-60 cells with (48A) 1 (48B) 26a (48C) 49a (48D) 49g at indicated concentrations and time points.
FIGS. 49A to 49D show the treatment of KGla cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of KGla cells with (49 A) 1 (49B) 26a (49C) 49a (49D) 49g at indicated concentrations and time points.
FIGS. 50A to 50D shows the treatment of THP-1 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of THP-1 cells with (50A) 1 (50B) 26a (50C) 49a (50D) 49g at indicated concentrations and time points.
DETAILED DESCRIPTION OF THE INVENTION
While the making and using of various embodiments of the present invention are discussed in detail below, it should be appreciated that the present invention provides many applicable inventive concepts that can be embodied in a wide variety of specific contexts. The specific embodiments discussed herein are merely illustrative of specific ways to make and use the invention and do not delimit the scope of the invention.
To facilitate the understanding of this invention, a number of terms are defined below. Terms defined herein have meanings as commonly understood by a person of ordinary skill in the areas relevant to the present invention. Terms such as "a", "an" and "the" are not intended to refer to only a singular entity, but include the general class of which a specific example may be used for illustration. The terminology herein is used to describe specific embodiments of the invention, but their usage does not delimit the invention, except as outlined in the claims.
As used herein, the term AKR1C3 is an acronym for aldo-keto reductase family 1, member C3 polypeptide, Aldo-keto reductase 1C3, and is also referred to as type 5 17 -hydroxysteroid dehydrogenase.
Prostate cancer is the second leading cause of mortality among American men with the highest incidence rate of all cancers reported in the United States. Surgical removal of prostate by radical prostatectomy and radiation therapy are applied to resect localized tumor. However, the treatment of advanced and metastatic forms of prostate cancer relies heavily on androgen deprivation therapy (ADT) that involves surgical (orchiectomy) and/or chemical castration. Chemical agents such as GnRH agonists (Lupreolide, goserelin, buserelin) and antiandrogens (bicalutamide, enzalutamide) reduce the levels of circulating androgens that consequently retard prostate cancer cell proliferation. After an initial response to ADT via castration, invariably in all prostate cancer patients the tumor adapts, giving rise to a more aggressive and fatal phenotype known as Castration Resistant Prostate Cancer (CRPC). CRPC is marked by molecular changes that involve an increase in the expression of androgen synthesizing enzymes, androgen receptor mutation and/or its reactivation. Such adaptations can lead to an increase in intratumoral androgen biosynthesis along with an increase in tumor responsiveness to circulating castrate levels of androgens.
Based on the prior art, it is well known that AKR1C3 plays a role in cancer development and chemotherapy resistance, and that this enzyme is a target for treating castrate resistant prostate cancer. Although inhibitors are available that can block AKRlC3's enzymatic activity, they also block AKR1C1 and AKRlC2's activities, which affects healthy hormonal levels and balance.
The aldo-keto reductase family 1-member C (AKR1C) enzymes are oxidoreductases that catalyze NADPH-dependent reductions of aldehydes and ketones in a range of steroids, carbohydrates, and prostaglandins. The isoform AKR1C3 specifically catalyzes the conversion of androgen precursors to
potent androgen receptor (AR) ligands, testosterone and a-dihydrotestosterone. AKR1C3 is also known as prostaglandin F synthase and as it catalyzes the conversion of prostaglandin D2 to 11 ^-prostaglandin F2a and its prostanoids, which affects the growth and spread of myeloblasts and myelocytes, it is therefore an important regulator of myeloid cell proliferation and differentiation.
The dual capabilities of AKR1C3 make it responsible for both the pathogenesis and progression of hormone dependent and independent cancers, including castration resistant prostate cancer (CRPC) and acute myeloid leukemia (AML). Additionally, AKR1C3 can lead to clinical chemotherapeutic resistance, as it reduces the effectiveness of first line of defense drugs for CRPC and AML.
Previous AKR1C3 inhibition research has found compounds with some degree of selectivity, but further improvements can be made to reduce the inhibition of the other AKR1C isoforms. The strategy behind these mechanisms has been taking known AKR1C3 inhibitors, and altering their functional groups through meta and para-directing to increase the inhibitor's level of selectivity. Due to the lack of high selectivity in previous research, no AKR1C3 inhibitor has been approved for clinical testing.
The present invention provides a series of compounds that possess the greatest selectivity for the enzyme AKR1C3 over its isoforms ever reported. The AKR1C3 enzyme is a therapeutic target for prostate and breast cancer and leukemia.
Abiraterone acetate (AA) acts by inhibiting CYP17A1, an upstream enzyme in the steroid biosynthetic pathway, which was approved by the FDA for CRPC treatment in 2011 and is effective in treating state 2 of CRPC. However, a majority of patients experience hypertensive crisis due to accumulation of desoxycorticosterone (DOC) and hence, co-administration with prednisone is imperative. Enzalutamide (ENZ) is another clinically used therapeutic that exerts potent androgen receptor (AR) antagonistic activity by inhibiting AR nuclear translocation, co-activator recruitment and AR binding to androgen response elements (ARE's) and henceforth, it could be effective up to state 3 CRPC. Albeit initial response, resistance to both AA and ENZ still develops by either an increase in intratumoral CYP17A1 expression or AR overexpression along with AR mutation that impairs inhibitor binding. Furthermore, due to the tumor heterogeneity and complex dynamics of androgen biosynthesis via various compensating pathways along with frequent mutations in AR and androgen biosynthetic enzymes the prognosis for patients that have advanced to CRPC still remains bleak. A need for novel therapeutics that can delay or reverse the emergence of resistance is inevitable.
Aldo-keto reductase 1C3 (AKR1C3) is a downstream enzyme in the steroid biosynthetic pathway and plays a pivotal role in the pathogenesis and progression of CRPC by catalyzing the conversion of weak androgen precursors to the potent androgen receptor (AR) ligands: testosterone (T) and 5a- dihydrotestosterone (5a-DHT). Also known as prostaglandin (PG) F synthase, AKR1C3 catalyzes the conversion of PGD2 to l ip-PGF2a and PGF2a prostanoids and hence acts as an important regulator of myeloid cell proliferation and differentiation. This dual enzymatic action makes AKR1C3 responsible
for the pathogenesis and progression of both hormone dependent and independent cancers. A considerable interest has been developed in investigating the role of AKR1C3 in a variety of cancers of the breast, lungs and colon. Prior studies have already validated the role of AKR1C3 in the pathogenesis and progression of prostate cancer. Clinically, AKR1C3 has been shown to be the most upregulated enzyme isoform among CRPC patients. Apart from inducing a CRPC phenotype, AKR1C3 is also responsible to mediate resistance to ENZ by providing a source of intratumoral androgens. As the enzyme acts at the final steps of androgen synthesis pathway, it eliminates the risk of DOC accumulation and eventually development of life threatening hypertension. Such activities make AKR1C3 an attractive target for managing CRPC disease progression as well as therapeutic resistance. Related isoforms AKR1C1 and AKR1C2 share high homology with AKR1C3 and are responsible for normal steroid metabolism. Hence, design of a potent yet isoform selective AKR1C3 inhibitor is a major challenge in the drug discovery process.
EXAMPLE 1. Novel compounds with higher selectivity for the enzyme AKR1C3 using a structure- activity relationship (SAR) optimization approach.
The present invention provides a series of compounds that possess the greatest selectivity for the enzyme AKR1C3 over its isoforms ever reported by adopting a structure-activity relationship (SAR) optimization approach based on a structurally novel natural product scaffold 'baccharin' isolated from Brazilian green propolis exhibiting potent AKR1C3 inhibition activity (IC50 = 100 nM) and crucially, exquisite selectivity for the AKR1C3 isoform. Because of the metabolic instability of the ester side chain hydrolytically stable analogues were synthesized that displayed superior AKR1C3 inhibitory activity than baccharin with retention of isoform selectivity. In our previous studies the inventors have shown that derivative AKR1C3 inhibitors display a very potent synergistic effect in combination with clinical chemotherapeutic agents in a panel of leukemia cell lines. In this report, the inventors demonstrate the activity of a hydrolytically stable amide analogue of baccharin to exert a cytotoxic effect in a panel of prostate cancer cell lines and a very high degree of synergistic drug interaction with ENZ. Mechanistic studies demonstrate apoptotic cell death in sensitive and ENZ resistant prostate cancer cell lines with a consequent reduction in AR and PSA levels.
FIGS. 1A-1D show examples of AKR1C3 inhibitors synthesized based on baccharin structural scaffold. KV-37 was chosen for further studies as it demonstrated a very potent AKR1C3 inhibitory IC50 of 66 nM. Androgen dependent 22Rvl and LNCaP cell lines were chosen to evaluate the activity of KV-37. 22Rvl cells were found to be high expressors of AKR1C3 as compared to LNCaP. Moreover, 22Rvl cell line, being intrinsically resistant to ENZ was an apt model for conducting CRPC studies.
Human liver stability of Baccharin and derivative AKR1C3 inhibitors: Baccharin and derivative AKR1C3 inhibitors were subjected to incubation with human S9 fractions in the presence of phase I co- factors from 0-240 minutes. Aliquots were withdrawn and analyzed by LC-MS method at various time points. The half-life of the inhibitor compounds is summarized in table 1.
Table 1
As expected, baccharin and the ester analogue were readily hydrolyzed. The amide inhibitor KV-37 displayed a remarkable stability and half-life of >240 min. The AKR1C3 inhibitory activity of baccharin was completely abrogated when, upon hydrolysis it yielded the known phenol drupanin, exhibiting AKR1C3 inhibition at IC50 of 15 μΜ and 7 fold selectivity over AKR1C2.
FIGS. 2A-2C are tables showing prostate cancer cell viability observed upon treatment with baccahrin. No reduction in prostate cancer cell viability was observed upon treatment with baccahrin. (FIG. 2A, 2B, 2C) very low levels of cytotoxicity were observed in DU-145, LNCaP and VCaP cells cultured in normal media when treated with baccharin at concentrations up to 100 μΜ. Effect of AKR1C3 inhibitors on CaP cell viability: Baccharin and derivative AKR1C3 inhibitors were screened for cytotoxic effects in a panel of prostate cancer cell lines (DU-145, LNCaP, 22Rvl, VCaP). Baccharin consistently displayed a lack of toxicity among all the cell lines tested up to concentrations as high as 100 μΜ (FIG. 2A, 2B, 2C). This is in agreement with previous studies demonstrating little or no toxicity for baccharin in various cancer cell lines, as assayed in the NCI-60 panel. KV-37 however, demonstrated a potent cytotoxic effect, which was time dependent in androgen dependent LNCaP, VCaP and 22Rvl cell lines.
FIGS. 3A-3F are graphs showing KV-37 induces potent cytotoxic activity in multiple CaP cell lines. Dose and time dependent reduction in cell viability after inhibitor treatment: (FIGS. 3A and 3B) LNCaP cells cultured in normal media and CSS media respectively. (FIG. 3C) VCaP cells cultured in normal media. (FIGS. 3D, 3E, 3F) 22Rvl cells cultured in normal media, CSS media and CSS media supplemented with 10 nM AD respectively. A further enhancement in cytotoxic activity was observed when the cells were cultured in charcoal stripped serum (CSS) media. The IC50 of KV-37 decreased from 45 μΜ to 25 μΜ in LNCaP cells and from 50 μΜ to 25 μΜ in 22Rvl cells upon incubation in CSS media as against hormone replete media. In the next set of experiments KV-37 was incubated with 22Rvl cells in CSS media supplemented with 10 nM A4-androstenedione (AD). Similar dose and time dependent reduction in cell viability was observed: IC50 being 25 μΜ at 72 hours. At higher inhibitor concentrations of 75 μΜ complete abrogation of cell viability was observed in LNCaP and 22Rvl cells when cultured in CSS media.
An increase in cytotoxic activity of KV-37 among CaP cells cultured in CSS media is attributable to the overexpression of AKR1C3 when cells are grown in media devoid of androgens. This drives the cellular phenotype to more closely resemble clinical castrate conditions where the cancer cells are dependent on AKR1C3 for survival and proliferation. A4-AD is a weak AR ligand and a substrate for AKR1C3. The
enzyme converts A4-AD to more potent AR ligand testosterone that drives cell proliferation. The addition of 10 nM A4-AD to CSS culture further mimics the clinical CRPC phenotype when only castrate levels of circulating androgens are available.
Compound KV-37 sensitizes CaP cells to enzalutamide cytotoxicity and exerts a synergistic drug effect: The dose response curve of ENZ in LNCaP and 22Rvl cells displays an increase in resistance upon culture in CSS media. In order to restore the sensitivity of these cell lines to ENZ cytotoxicity, LNCaP and 22Rvl cells were pre-treated with KV-37 for 24 hours followed by exposure to ENZ. The combination demonstrated a remarkable synergistic effect at both the time points of 48 and 72 hours post ENZ exposure in normal as well as CSS media. Among CSS cultures, concentrations as low as 1 μΜ of KV-37 were capable of sensitizing both 22Rvl and LNCaP cell lines to reduce cell viability by 50% upon treatment with only 1 μΜ ENZ when either agent alone did not exert any toxic behavior.
FIGS. 4A-4F show pre-treatment with KV-37 sensitizes LNCap and 22Rvl cells to ENZ cytotoxicity. (FIG. 4A, 4B) 24 hour pre-treatment of LNCaP cells in normal and CSS media followed by treatment with various concentrations of ENZ for 72 hours (FIG. 4C, 4D, 4E) 24 hour pre-treatment of 22Rvl cells in normal and CSS media followed by treatment with various concentrations of ENZ at indicated time points. (FIG. 4F) Quantification of the degree of synergism. Combination and dose reduction indices (CI, DRI) generated using compusyn software. However, co-treatment experiments did not show any adjuvant effect between both cell lines. Quantification of degree of synergism was made using the Chou- Talalay method. Up to a 100-fold reduction in ENZ dosing was achieved in 22Rvl cells while in LNCaP cells 40 fold reduction was observed.
FIGS. 5A-5D are images showing KV-37 induces apoptosis in 22Rvl cells. (FIG. 5A, 5B) Dose dependent Increase in apoptotic cell population in 22Rvl cells at indicated time points. (FIG. 5C) Graph quantifying the percentage of apoptotic cells after KV-37 treatment. (FIG. 5E) Western blots showing a dose dependent increase in c-caspase and C-PARP after treatment with KV-37 at indicated time points in CSS media. KV-37 induces cell death in 22Rvl cells via apoptosis: 22Rvl cells cultured in CSS media were treated with increasing doses KV-37 for 48 and 72 hours and stained with AnnexinV/PI to measure the apoptotic cell percentage following treatment. FIG. 5 outlines the increase in the percentage of apoptotic cells with increase in concentration of KV-37 as analyzed by flow cytometry. Compound incubation for 72 hours (FIG. 5B) demonstrates a further increase in apoptotic cell percentage as compared to the 48 hour time point (FIG. 5A). FIGS. 5C and 5D outline the percent increase in apoptosis as compared to the control. In order to further corroborate these findings, western blot analysis was conducted after inhibitor treatment following a similar dosing schedule. The results, in agreement with flow cytometry data consistently show a dose dependent increase in the levels of c-caspase and c- PARP with an even higher expression at 72 hours as compared to 48 hours incubation time (FIGS. 5E and 5F). Taken together, the data strongly suggests apoptosis as the primary mechanism of cytotoxicity for compound KV-37.
FIG. 6 is an image showing downstream molecular changes after KV-37 treatment in 22Rvl cells. Western blot showing a decline in AKR1C3, AR and PSA levels. Treatment of 22Rvl cells with KV-37 downregulates AKR1C3, AR and PSA expression: To evaluate molecular changes following inhibitor treatment in 22Rvl cells, western blot analysis was conducted to measure the expression levels of AKR1C3, AR and PSA. As expected, AKR1C3 expression levels decline after treatment with KV-37 and the downregulation is even stronger at a longer incubation time of 72 hours (FIG. 6). Since, a functional correlation exists between AKR1C3 and AR, it was prudent to measure AR levels after treatment. The inventors observed that the decline in AR levels complement AKR1C3 downregulation and could be the reason for sensitizing the cells to ENZ cytotoxicity to exert a synergistic drug interaction. PSA is widely used as a biomarker for prostate cancer and governs the disease progression. The inventors probed PSA levels after inhibitor treatment that shows a dose dependent reduction in PSA expression signifying a beneficial therapeutic outcome. To further confirm a reduction in the expression of AKR1C3, 22Rvl cells were treated with KV-37 at 1 and 10 μΜ concentration for 48 hours followed by immunostaining against AKR1C3.
FIG. 7 is an image showing KV-37 treatment of 22Rvl cells downregulated AKR1C3. Immunocytochemical analysis showing a concentration dependent decline in AKR1C3 levels after 48 hours incubation. The data clearly indicates a reduction in AKR1C3 levels as denoted by the green stain. Counterstaining with DAPI shows no morphological changes in the nucleus. Combination of KV-37 with ENZ synergistically downregulates AKR1C3, AR and PSA expression and upregulates c-caspase and c-PARP levels:
FIGS. 8A-8B are images showing molecular changes following a combination treatment of KV-37 and ENZ in 22Rvl cells after 72 hours incubation. To determine the mechanism of synergistic interaction between KV-37 and ENZ, western blot analysis was carried out to measure molecular changes in the combination treatments. As outlined in FIG. 8A, the combination of 25 μΜ KV-37 with 25 μΜ ENZ completely abrogates PSA expression with a concomitant decrease in AKR1C3 and AR levels at 72 hours (FIG. 8B). A predominant increase in c-caspase and c-PARP levels was also observed at both time points. Taken together the data indicate apoptotic cell death as a primary mechanism of synergistic cytotoxic effect as a consequence of reduction in the activity of AKR1C3 and AR.
The disclosed compositions are AKR1C3 inhibitors with >1800-fold selectivity for AKR1C3 over its other isoforms. It provides a composition for the treatment of cancer and contributes to the effectiveness of chemotherapeutic drugs that are otherwise reduced in effect by AKR1C3.
The aldo-keto reductase family 1 member C (AKR1C) enzymes are oxidoreductases, which catalyze the NADPH-dependent reduction of aldehyde and ketone functionalities on a range of steroids, carbohydrates, and prostaglandins. The AKR1C3 enzyme isoform catalyzes the downstream conversion of androgen precursors to the potent androgen receptor (AR) ligands: testosterone and 5a- dihydrotestosterone (5a-DHT). Also known as prostaglandin (PG) F synthase, AKR1C3 catalyzes the conversion of PGD2 to l ip-PGF2a and PGF2a prostanoids and hence acts as an important regulator of myeloid cell proliferation and differentiation. This dual enzymatic action makes AKR1C3 responsible for the pathogenesis and progression of both hormone dependent and independent cancers such as Castration Resistant Prostate Cancer (CRPC) and Acute Myeloid Leukemia (AML). AKR1C3 is not only responsible for the pathogenesis of the aforementioned malignancies but also mediates resistance to clinical chemotherapeutics. It reduces the effectiveness of enzalutamide (first line therapy for CRPC) by providing a source of intratumoral androgens and transforms anthracycline antibiotics (first line therapy for AML) to inactive hydroxy metabolites. Related isoform AKR1C2 share high homology with AKR1C3 and transforms potent androgen 5a-DHT to an inactive metabolite. Hence, inhibition of 1C2 isoform is undesirable and the discovery of a potent yet isoform selective AKR1C3 inhibitor is paramount to manage cancer progression and obliterate the therapeutic resistance. Based on a natural product scaffold the inventors have synthesized and characterized a novel, potent and extremely selective AKR1C3 inhibitor that exhibits >1800- fold selectivity towards the AKR1C3 isoform.
Based on a natural product scaffold we have synthesized and characterized a novel, potent and extremely selective AKR1C3 inhibitors (KV-60-5a) and derivatives, that exhibits >1800-fold selectivity towards the AKR1C3 isoform. This is the most selective AKR1C3 inhibitor that has ever been discovered.
KV-60-5a, the most selective AKR1C3 inhibitor yet identified, is shown to be non-toxic to LNCaP prostate cancer cells (when grown in charcoal stripped serum (CSS), which serves to upregulate AKR1C3 expression) at 1 μΜ concentration.
FIG. 9 is a graph of the compound KV-60-5a showing no cytotoxicity to LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hours exposure at 1 μΜ concentration. IC50 value of KV-60-5a was determined to be 100 μΜ. Note that although AKR1C3 expression is enhanced in LNCaP cells grown in CSS media the relative expression of the enzyme target is still low. Clinical samples of castration resistant prostate cancer and other cell lines express much greater levels of AKR1C3. In such cell lines both direct toxicity and potentiation effect are expected to be much greater. These experiments are currently ongoing.
FIG. 10 is a graph of the compound KV-60-5a showing significant potentiation effect of the clinical antineoplastic enzalutamide in LNCaP cells cultured in charcoal stripped serum (CSS) after 72 hours. Synergistic treatment of 1 μΜ of KV-60-5a with 1 μΜ of the clinical antineoplastic enzalutamide results in 42% cell viability compared with 1 μΜ of enzalutamide alone which results in 82% cell viability (FIG. 10). A potentiation effect of approximately 100% for LNCaP cells grown in CSS.
FIG. 11 in an image of the modelling of KV-60-5a docked into the AKR1C3 enzyme. Enabled by the synthesized library of AKR1C3 inhibitors the inventors have conducted quantitative structure -activity relationship (QSAR) computer modelling that has identified compounds with retained selectivity and predicted picomolar affinity. Modelling predictions show that the structure of the highly selective inhibitor KV-60-5a changes the pose of the compound when bound with AKR1C3. This results in pi-pi stacking interactions between aromatic amino acids TYR216, TRP227 and PHE311, and aromatic constituents of the inhibitors, which accounts for the enhanced affinity.
FIG. 12 shows a synthetic scheme for the inhibitor. As seen in 1, 3-bromo-5-iodo-N-(2- phenylethyl)benzamide (2): To a solution of 3-bromo-5-iodo benzoic acid (500 mg, 1.5 mmol) in dry toluene (10 mL) was added SOC12 (290 μΕ, 4 mmol) and the mixture was refluxed overnight. The reaction vessel was cooled to room temperature and the solvent was evaporated in vacuo. The resultant brown oil was used further without purification. To a solution of 3-bromo-5-iodo benzoyl chloride in DCM (10 mL) was added DMAP (26 mg, 0.2 mmol) and the flask was purged with nitrogen. 2- phenylethylamine (250 μΕ, 2 mmol) and NEt3 (420 μΕ, 3 mmol) were added to the flask and the mixture was stirred at 70° C overnight. The reaction mixture was cooled to room temperature, diluted with DCM,
washed with a saturated aqueous NaHC03, water and extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1) provided the title compound as a white solid ( 645 mg, 1.5 mmol, 98%). tert-butyl (2E)-3-{3-bromo-5-[(2- phenylethyl)carbamoyl]phenyl}prop-2-enoate (3): To a solution of (2) (645 mg, 1.5 mmol) in dry toluene (15 mL), was added PPh3 (40 mg, 0.15 mmol), Pd(OAc)2 (17 mg, 0.07 mmol) and the flask was purged with nitrogen. tert-Butylacrylate (300 μΐ., 2.0 mmol) and NEt3 (560 μΐ., 4.0 mmol) were added and the mixture was stirred at reflux overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried ( a2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a brown oil (288 mg, 0.67 mmol, 45%). tert-butyl (2E)-3-[3-(3-methylbut-2-en-l-yl)-5-[(2- phenylethyl)carbamoyl]phenyl] prop-2-enoate (4): To a solution of (3) (80 mg, 0.18 mmol) in dry DMF (2 mL) was added Cs2C03 (130 mg, 0.4 mmol), Pd(dppf)C12 (8.5 mg, 0.01 mmol) and the flask was purged with nitrogen. Prenyl boronic acid pinacol ester (70 μΐ., 0.3 mmol) was added and the mixture was stirred at 90° C overnight. The reaction was allowed to cool and was filtered through a CELITE® pad with EtOAc. The solvent was evaporated in vacuo, re-dissolved in DCM and the residual DMF removed by washing with copious amounts of water in DCM, dried ( a2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1, 2: 1) provided the title compound as a transparent oil (50 mg, 0.12 mmol, 66%). (2E)-3-[3-(3-methylbut-2-en-l-yl)-5-[(2- phenylethyl)carbamoyl]phenyl]prop-2-enoic acid (5): To a solution of (4) (50 mg, 0.12 mmol) in dry toluene (10 mL) was added silica gel (5 mL) and the suspension was stirred at reflux overnight. The reaction was allowed to cool and the mixture was filtered after diluting with 20% MeOH in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (DCM:MeOH = 20: 1) provided the title compound as a white solid (21.7 mg, 0.06 mmol, 50%).
FIG. 13 shows representative derivative compounds with a modification of phenyl (6). FIG. 14 shows representative derivative compounds with a modification of Dihydrocinnamyloxy (6). FIG. 15 shows representative derivative compounds with a Conversion of Ester to Amide Group (5). FIG. 16 shows representative derivative compounds with a Positional Switch of Prenyl & Dihydrocinnamyloxy Group (15). FIG. 17 shows representative derivative compounds with a Positional Switch of Dihydrocinnamoyloxy Group Analogs (3). FIG. 18 shows representative derivative compounds a Switch of Amide Bond Direction-Reverse Amides (10). FIG. 19 shows representative derivative compounds with a Modification of Carboxylate Group (2).
EXAMPLE 2. A Highly Potent AKR1C3 Inhibitor Exhibits Single Agent Cytotoxicity and Significantly Potentiates Enzalutamide in Combination Therapy Across Prostate Cancer Models.
Aldo-keto reductase 1C3 (AKR1C3) also known as type 5 17 β-hydroxysteroid dehydrogenase is responsible for intratumoral androgen biosynthesis, contributing to the development of castration- resistant prostate cancer (CRPC) and eventual chemotherapeutic failure. Significant upregulation of
AKR1C3 is observed in CRPC patient samples and derived CRPC cell lines. As AKR1C3 is a downstream steroidogenic enzyme synthesizing intratumoral testosterone (T) and 5a-dihydrotestosterone (DHT), the enzyme represents a promising therapeutic target to manage CRPC and combat the emergence of resistance to clinically employed androgen deprivation therapy. Herein, the inventors demonstrate the antineoplastic activity of a potent, isoform selective and hydrolytically stable AKR1C3 inhibitor (E)-3-(4-(3-methylbut-2-en-l-yl)-3-(3- phenylpropanamido)phenyl)acrylic acid (KV-37) which reduces prostate cancer cell growth in vitro and in vivo, and sensitizes CRPC cell lines (22Rvl and LNCaPlC3) towards enzalutamide cytotoxicity. Crucially, KV-37 does not induce cytotoxicity in non- malignant WPMY-1 prostate cells nor does it induce weight loss in mouse xenografts. Moreover, KV-37 reduces androgen receptor (AR) transactivation and prostate specific antigen (PSA) expression levels in CRPC cells lines indicating a therapeutic drug effect in prostate cancer. Combination studies of KV-37 with enzalutamide reveal a very high degree of synergistic drug interaction that induces significant cytotoxicity in prostate cancer cells via apoptosis, resulting in >200-fold potentiation of enzalutamide cytotoxicity in drug resistant 22Rvl cells. These results demonstrate a promising therapeutic strategy for the treatment of drug resistant CRPC that invariably develops in prostate cancer patients following initial treatment with AR antagonists such as enzalutamide.
Prostate cancer is the third (1) leading cause of mortality among American men and has the highest incidence of all cancers reported in the US (2). Surgical removal of the prostate by radical prostatectomy and radiation therapy to resect the localized tumor are standard therapeutic interventions (3). The treatment of advanced and metastatic forms of prostate cancer relies heavily on androgen deprivation therapy (ADT) that involves surgical (orchiectomy) and/or chemical castration (4). Chemical agents such as gonadotropin releasing hormone (GnRH) agonists (Lupreolide, (5) goserelin, (6) buserelin (7)) and antiandrogens (bicalutamide (8)) retard prostate cancer cell proliferation and lead to remission. After an initial response to ADT that results in castrate levels of circulating androgens, the tumor adapts, giving rise to a more aggressive and fatal disease known as castration-resistant prostate cancer (CRPC) which is characterized by molecular changes that include an increase in the expression of androgen synthesizing enzymes and reactivation of androgen signaling (9,10). Such adaptations can lead to relapse despite the presence of circulating castrate levels of androgens (11,12).
Abiraterone acetate (AA) was approved by the US Food and Drug Administration for CRPC treatment in 2011 and acts by inhibiting P450cl7 (13), an upstream enzyme in the steroid biosynthetic pathway and is effective in CRPC where tumor progression is dependent on intracrine androgen synthesis that consequently activates AR signaling (14,15). However, to prevent hypertensive crisis due to accumulation of desoxycorticosterone (DOC) in the adrenal gland, co-administration with prednisone is imperative (16). Enzalutamide (ENZ) is another clinically used therapeutic that exerts potent androgen receptor (AR) antagonistic activity by inhibiting AR nuclear translocation, co-activator recruitment and AR binding to androgen response elements (ARE's) (17). The chemotherapeutic is effective up to stage 3
CRPC where proliferation is dependent on AR activation that occurs even in the absence of an androgen ligand (15). After an initial response to AA or ENZ, resistance develops rapidly due to increase in intratumoral androgen biosynthesis (18), AR overexpression, AR mutation, that makes the receptor ligand promiscuous, or the appearance of AR splice variants that make the AR constitutively active in the absence of the ligand binding domain (15,19,20). Due to tumor heterogeneity and the emergence of adaptive pathways of androgen biosynthesis (21), combined with frequent mutations in AR and androgen biosynthetic enzymes (10,22), the prognosis for patients that have advanced to CRPC remains bleak. Thus, there is an urgent need for novel therapeutics that can delay or reverse the inevitable emergence of resistance.
Type 5 17 -hydroxy steroid dehydrogenase, also known as aldo-keto reductase 1C3 (AKR1C3) acts downstream in the steroidogenesis pathway and plays a pivotal role in the pathogenesis and progression of CRPC by catalyzing the conversion of weak androgen precursors to the potent AR ligands: testosterone (T) and 5a-dihydrotestosterone (5a-DHT) (23). Also known as prostaglandin (PG) F synthase, AKR1C3 catalyzes the conversion of PGD2 to l ip-PGF2a and PGF2a prostanoids (24). An increase in PGF2a leads to activation of the FP receptor and consequently induces proliferation and radiation resistance in prostate cancer cells (25).
Clinically, AKR1C3 has been shown to be the most upregulated steroidogenic enzyme in CRPC patients (26). Apart from inducing a CRPC phenotype, AKR1C3 also mediates resistance to ENZ and AA by providing a source of intratumoral androgens and this resistance can be surmounted by indomethacin (INDO) a specific AKR1C3 inhibitor that also inhibits COX isozymes (27,28). As the enzyme acts at the final steps of the androgen synthesis pathway in the prostate (16), AKR1C3 inhibitors would not cause the accumulation of DOC in the adrenal gland and would not have to be co-administered with prednisone. These properties make AKR1C3 an attractive target for managing CRPC disease progression as well as countering therapeutic drug resistance. The related isoforms AKR1C1 and AKR1C2 share high homology with AKR1C3, function to eliminate DHT, and should not be inhibited (29). Numerous AKR1C3 inhibitors with diverse scaffolds are known however, none have made their way into the clinic (30-32), Considerable interest has developed in the role of AKR1C3 in a variety of cancers of the breast (33), lungs (34) and colon (35).
Herein, the inventors report on the actions of a novel, potent, isoform selective and metabolically stable AKR1C3 inhibitor, KV-37 (FIG. 20A) (36-38), in a range of prostate cancer cell lines and a xenograft model. The inventors have previously reported AKR1C3 inhibitors that act synergistically with etoposide and daunorubicin as chemotherapeutic agents in a panel of leukemia cell lines (38). The AKR1C3 inhibitor KV-37 exerts high synergistic drug interaction in combination with ENZ, which is superior to that seen with INDO. Mechanistic studies reveal that KV-37 causes apoptotic cell death in ENZ-resistant prostate cancer cell lines with a consequent reduction in PSA levels and in vivo studies demonstrate a >50% reduction in tumor growth without observable toxicity.
Cell Culture and Reagents. 22Rvl and LNCaP cells were purchased from American Type Culture Collection (ATCC, Manassas, VA) and cultured in RPMI 1640 supplemented with 10% FBS, 100 U/mL penicillin and 0.1 mg/mL streptomycin (39). LNCaPlC3 cells overexpressing AKR1C3 were generated by stable transfection of AKR1C3 plasmid as previously described (12). WPMY-1 cells were purchased from ATCC and maintained in DMEM media supplemented with 5% FBS, 100 U/mL penicillin and 0.1 mg/mL streptomycin. Where indicated, cells were also cultured in charcoal stripped (CSS) media prepared by supplementing RPMI 1640 without phenol red with charcoal stripped FBS. All cells were maintained at 37oC in a humidified incubator with 5% carbon dioxide. Enzalutamide (ENZ) or MDV3100 (catalog no. 50-101-3979) and indomethacin (INDO) (catalog no. AAA1991006) were purchased from Fisher Scientific. Stock solutions of ENZ, INDO and KV-37 were prepared in DMSO and were serially diluted for cell culture treatments maintaining the final DMSO concentration at less than 1%.
Inhibition of Testosterone Production by AKR1C3 Inhibitors. Systems (200 μΕ) containing 100 mM potassium phosphate pH 7.0, 0.9 mM NADPH, 1.2 - 40 μΜ 4-androstene-3,17-dione (A4-AD) (containing 2.6 nM [3H]-4-androstene-3,17-dione), KV-37 (0 -10 μΜ), and 0.3 - 10 μΜ inhibitor plus 1.65 μΜ AKR1C3 were incubated at 37 oC, and samples were prepared as previously reported (40).
Stability Testing of AKR1C3 Inhibitors. Baccharin, KV-37 and a related ester analogue KV-32 (2 μΜ solution in DMSO) were incubated with male CD-I murine liver S9 fractions containing a NADPH generating system for 0 - 240 minutes. Reactions were quenched with 0.5 mL (1 : 1) of acetonitrile (ACN) containing 0.2% formic acid and 100 ng/mL Internal Standard (IS) (final cone. = 50 ng/mL). Samples were vortexed for 15 seconds, incubated at RT for 10 minutes and spun for 5 minutes at 2400 rpm. Supernatant (0.9 mL) was then transferred to an eppendorf tube, chilled and centrifuged for 5 minutes at 13,200 rpm. Supernatant (800 μί) was transferred to a HPLC vial and was then analyzed by Qtrap 4000 mass spectrometer using the following parameters: Ion Source/Gas Parameters: curtain gas (CUR) = 35, collisionally activated dissociation (CAD) = Medium, ionspray voltage (IS) = 4500, temperature (TEM) = 600, nebulizing gas GS1 = 70, drying gas GS2 = 70. Buffer A: dH20 + 0.1% formic acid; Buffer B: ACN + 0.1% formic acid; flow rate 1.5 ml/min; column Agilent C18 XDB column, 5 micron packing 50 X 4.6 mm size; 0 - 1 min 97% A, 1 - 2.5 min gradient to 99% B, 2.5 - 3.5 min 99% B, 4 - 4.1 min gradient to 97% A, 4.1 - 4.5 min 97% A; IS: Warfarin (in MeOH, transition 307.3 to 160.9). Ion transitions followed were 363.1 [M-H] to 186.9 for baccharin and KV-32, and 362.1 [M-H] to 318.1 for KV-37.
Cell Viability Assays. Cells were seeded at a density of 10,000 cells/well in 96-well plates and were incubated in either normal media (24 h) or charcoal stripped serum (CSS) media (48 h). Treatments with ENZ, INDO, KV-37 or combinations of ENZ and KV-37 were made with or without 10 nM A4-AD (AKR1C3 substrate) and incubated at the indicated time points (24, 48, 72 and 96 h). For pretreatment experiments, cells were treated with KV-37 for 24 h followed by the addition of ENZ and incubated for
further 72 h. Cell viability was determined by the MTS tetrazolium dye assay as described previously (38).
Inhibition of Testosterone Production in LNCaP-AKRlC3 Cells. LNCaPlC3 cells (1.5 x 106 cells per well) were placed in 2 mL phenol red-free RPMI, 5% CD-FBS, 2 mM L-glu, 1% P/S. Cells were incubated with DMSO, or 10 μΜ or 30 μΜ KV-37 for 30 min at 37 °C. After the pre -incubation, 100 nM 3H A4-AD final concentration was added to the wells and cells were incubated for 24 h. Cell media was transferred into labeled borosilicate glass tubes after 48 h and extracted with 2 mL ethyl acetate. Samples were vortexed for 30 s and placed in a -20 °C freezer for 1 h to separate the phases. The frozen samples were allowed to thaw at room temperature for 10 min and centrifuged for 20 min before the organic phase was extracted and the process repeated. The 4 mL organic layer extract was then dried under vacuum for radiochromatography. The aqueous fractions were dried under vacuum for 30 min to evaporate any leftover organic solvent residue. 1% glacial acetic acid aqueous solution (25 μΚ) was added to the samples to adjust the pH to 6.5, followed by addition of 18 μL of 25 U^L E.Coli β- glucuronidase. Samples were placed in a water bath at 37 °C for 24 hr. Samples were then extracted as before. All samples were resuspended in 100 μΐ. of ethyl acetate and vortexed for 30 s to mix. Using 10 μΐ. of capillary tubes, each sample was spotted onto a multi-channel UNIPLATE 20 x 20 cm TLC plate and analyzed as described before.
Apoptosis Assay. AnnexinV/FITC apoptosis assay was performed using a kit and according to the manufacturer's instructions (BD Biosciences, catalog no. 556547). Cells were seeded at a density of 0.2 x 106 cells/well in 24- well plates and incubated in CSS media for 48 h followed by treatment with KV-37 for 48 and 72 h. For pretreatment experiments, cells were treated with KV-37 for 24 h followed by the addition of ENZ and incubated for a further 72 h. After appropriate treatments, the samples were analyzed by flow cytometery (Accuri C6, Ann Arbor, MI, USA).
Western Blotting. Whole cell lysates were prepared using 4% (w/v) CHAPS in urea-tris buffer. Protein was quantified and electrophoresed as described previously (41). The membranes were probed with primary antibodies against AR (Cell Signaling #5153P, rabbit mAb), PSA (Cell Signaling #5877S, rabbit mAb), AKR1C3 (Sigma #A6229, mouse mAb), C-PARP (Cell Signaling #5625S, Rabbit mAb), C- caspase3 (Cell Signaling #9661S, rabbit mAb) and Actin (Sigma #A5441, mouse mb). All the antibodies from Cell Signaling were diluted 1 : 1000, AKR1C3 (1:500) and actin (1:2000).
Tumor Xenograft and Pharmacokinetic Study. All animal experiments were carried out according to approved Institutional Animal Care and Use Committee protocols at the University of Texas Southwestern Medical Center (Dallas, TX). Eighteen 6-8 week old female NOD-SCID mice (low circulating testosterone) were implanted with 4 x 106 22Rvl cells in a volume of 0.1 mL in the left flank. When tumor volume reached -100 mm3 (day 13), mice were randomly divided into two groups. One group was administered 20 mg/kg KV-37 QD intra peritoneal and the other group was given vehicle control (10% DMSO, 20% PEG400, 0,5% tween 80, 69.5% carbonate buffer pH = 9.2). Tumor volumes
were measured twice a week with Vernier calipers and tumor volume was calculated as (L*W2)*3.14)/6. On day 34, mice were humanely euthanized and tumors were collected, weighed and frozen in liquid nitrogen after taking pictures. Whole blood was collected in an eppendorf tube with ACD anti-coagulant for plasma separation for pharmacokinetic evaluation of KV-37.
Pharmacokinetic Evaluation of KV-37: For Standards, 98 μΐ. of blank plasma was added to an eppendorf and spiked with 2 μΐ. of IS (Warfarin). For QCs, 98.8 μΐ. blank plasma was added to an eppendorf and spiked with 1.2 μΐ. of IS. 100 μΐ. of plasma was mixed with 200 μΐ. of methanol containing 0.15% formic acid and 37.5 ng/mL IS (IS final cone. = 25 ng/mL). The samples were vortexed for 15 s, incubated at room temperature for 10 min and spun at 13,200 rpm in a standard microcentrifuge. The supernatant was then analyzed by LC- MS/MS as described earlier.
Statistical Analyses and Quantification of Degree of Synergism. Experiments were repeated at least thrice and the statistical significance was calculated using the Student's t test. A p value of < 0.05 was considered statistically significant. IC50 values were calculated by GraphPad prism software. Combination of KV-37 with ENZ was analyzed by CompuSyn software (Biosoft) based on the Chou- Talalay method. Combination Index (CI) and Dose Reduction Index (DRI) values were generated to evaluate the degree of synergistic drug interaction.
It was found that inhibitors of AKR1C3 from various structural classes (flavones (30), jasmonates (31) and NSAID's (23,33)) have previously been described. The inventors adopted a cinnamic acid derivative 'baccharin' and conducted structure-activity relationship (SAR) studies to identify lead AKR1C3 inhibitors (37,38). The cinnamic acid derivative KV-37 (FIG. 1A) was chosen for further studies as it demonstrated potent AKR1C3 inhibition (IC50 = 66 nM) and selectivity (109-fold over AKR1C2) (38). In order to delineate the preliminary mechanism of action, the inventors tested its mode of AKR1C3 inhibition. The inventors found that KV-37 displayed competitive enzyme inhibition versus AKR1C3 when μ4-Αϋ was used as substrate, yielding a Ki value of 3.0 μΜ for the E.NADPH.KV-37 complex. The difference between this Ki value and the IC50 value seen in the preliminary inhibitor screen is consistent with the lower Ki value for the E.NADP+. inhibitor complex which is seen with other AKR1C3 inhibitors when they are screened in the oxidation direction (FIG. 201) (40). The inventors also examined the mode-of-action of KV-37 in androgen-dependent 22Rvl cells (high AKR1C3 expression) and LNCaP cells (low AKR1C3 expression). The 22Rvl cell line shows increased expression of AKR1C3 when grown in CSS media and possesses intrinsic resistance to ENZ and AA (27). LNCaP cells stably overexpressing AKR1C3 (LNCaP 1C3) were used to further validate the effect of KV-37 on the target (12). Further, WPMY-1 cells, a non-malignant prostate stromal cell line, devoid of AKR1C3 expression (FIG. 20J, 20K), were employed to evaluate the selective cytotoxicity of KV-37.
FIG. 201 shows Competitive inhibition of NADPH dependent reduction of A4-androstene-3,17-dione mediated by AKR1C3 by KV-37. KV-37 acts as a competitive Inhibitor in the conversion of [3Η]Δ4- androstene-3,17-dione to testosterone. Increasing concentrations of KV-37 (0-10 μΜ) were used to
inhibit the NADPH dependent conversion of fixed concentrations of [3H]A4-androstene-3,17-dione to testosterone catalyzed by recombinant AKR1C3 as measured by radiochromatography.
FIG. 20J. 22Rvl and LNCaPlC3 cells overexpress AKR1C3 whereas LNCaP and WPMY-1 exhibit low AKR1C3 levels. (FIG. 20J) Relative expression of AKR1C3 across prostate cancer cell lines. (FIG. 20K) Culture of 22Rvl cells in CSS media upregulates AKR1C3 expression levels. (NM, normal media; CSS, charcoal stripped serum media).
KV-37 inhibits AKR1C3 activity in prostate cancer cell lines by inhibiting the conversion of A4-AD to testosterone. KV-37 was used to inhibit the production of testosterone in LNCaPlC3 cells following β- glucuronidase treatment of the aqueous phase. At a concentration of 10 μΜ, >80% inhibition of testosterone production is blocked and the residual testosterone produced is that seen in untransfected LNCaP cells (FIG. 21A to 21C).
FIG. 21A to 21C. KV-37 inhibits conversion of [3H]A4-androstene-3,17-dione to testosterone in LNCaP- 1C3 cells. LNCaP-lC3 cells (1.5 x 106 cells per well) platted in phenol red-free RPMI, 5% CSS-FBS, 2 mM L-glutamine, 1% P/S and were with DMSO or 10 μΜ KV-37 for 30 min at 37 °C. After the preincubation, 100 nM [3H]A4-androstene-3,17-dione final concentration was added to the wells and cells were incubated for 24 h. FIG. 21A is solvent control; FIG. 21B contains 10 μΜ KV-37 and FIG. 21C are LNCaP control cells treated with DMSO. The peak at 90 mm is androsterone and the peak at 120 mm is testosterone liberated after β-glucronidase treatment. The data reveal almost complete inhibition of AKR1C3 mediated testosterone production.
KV-37 exhibits excellent plasma stability above that of the parent and other derivatives. Baccharin undergoes rapid hydrolysis to yield the inactive phenol drupanin (AKR1C3 IC50 = 15 μΜ) (37). In order to compare the hydrolytic stability of lead compounds, baccharin and its derivatives were subjected to incubation with mouse S9 fractions in the presence of phase I co-factors from 0-240 min. Aliquots were withdrawn and analyzed by LC-MS at various time points. As expected, baccharin and a related ester analogue (KV-32) were readily hydrolyzed (FIG. 22A to 22C). The amide-based inhibitor KV-37 displayed remarkable stability (tl/2 = >240 min).
FIG. 22A-22C: Metabolic instability of (FIG. 22 A) Baccharin and (FIG. 22B) ester derivative KV-32 on incubation with mouse S9 fractions. FIG. 22C shows the half-life of baccharin and derivative AKR1C3 inhibitors upon incubation with human S9 liver fractions.
AKR1C3 inhibitor KV-37 exhibits greater cytotoxicity to prostate cancer cell lines compared to baccharin. Baccharin and its analog KV-37 were screened for cytotoxic effects in a panel of human prostate cancer cell lines (LNCaP, 22Rvl and LNCaPlC3). Baccharin consistently displayed no toxicity among all the cell lines tested, up to concentrations as high as 100 μΜ (FIG. 23A to 23C). This is in agreement with previous studies (38). However, treatment with KV-37 demonstrated greater cytotoxicity in a dose and time- dependent manner in androgen dependent 22Rvl and LNCaP cell lines. The cytotoxic
effect was greater in 22Rvl cells due to the higher expression of AKR1C3 as compared to the LNCaP cell line (FIG. 20A-H). For 22Rvl cells, a significant decline in IC50 values (p = 0.0015, O.0001, <0.0001 at 24, 48 and 72 h respectively) was observed after KV-37 treatment when the cells were cultured in CSS media as compared to normal media (FIG. 20B, 20C, 20H). This is attributable to the overexpression of AKR1C3 when cells are grown in media devoid of androgens (supplementary FIG. S2). This drives the cellular phenotype to more closely resemble clinical castrate conditions, where the cancer cells are dependent on AKR1C3 for survival and proliferation. Culture in CSS media supplemented with an AKR1C3 substrate 10 nM A4-AD lead to a slight reduction in cytotoxic effect and a significant increase in IC50 values (p = O.0001, <0.0009, <0.0001 at 24, 48 and 72 h respectively) as compared to those observed in non-supplemented CSS media (FIG. 20C, 20E, 20H). Addition of 10 nM A4-AD to CSS culture mimics the serum level of this androgen in CRPC patients (12,42,43). As A4-AD is a substrate for AKR1C3, this observation suggests that the reduced cytotoxic activity of KV-37 is due to the enzymatic formation of T and DHT and corroborates AKR1C3 as the inhibitor target. Cytotoxic activity of KV-37 was also evaluated in normal media supplemented with 10 nM A4-AD as a control experiment to directly measure the difference in dose responses in normal and CSS media (FIG. 20D, 20H).
FIGS. 23A to 23C: Baccharin does not exert cytotoxicity on prostate cancer cell lines. Percentage cell viability of (FIG. 23A) LNCaP, (FIG. 23B) 22Rvl and (FIG. 23C) LNCaPlC3 cells cultured in CSS media after baccharin treatment at indicated concentration and time points. Data shown as mean ± SD of at least three independent experiments, n=6.
AKR1C3 inhibitor KV-37 sensitizes ENZ resistant prostate cancer cells to ENZ by exerting a synergistic drug effect. The 22Rvl cell line is intrinsically resistant to ENZ (27). The dose-response curve of ENZ in 22Rvl cells displays increasing resistance upon culture in CSS media (supplementary FIG. S6), which is accompanied by an increase in AKR1C3 expression (FIGS. 20J and 20K). LNCaP cells that are sensitive to ENZ-induced cytotoxicity were used as controls to compare the effect of KV-37 in combination with ENZ. To test whether the sensitivity of 22Rvl cells to ENZ could be restored with KV- 37, 22Rvl and LNCaP cells were pre-treated with KV-37 for 24 h followed by exposure to ENZ. The combination demonstrated a remarkable synergistic effect in 22Rvl cells at both 48 and 72 h post ENZ exposure in normal, as well as CSS media (CI = 0.19 and 0.07 respectively) (FIG. 25A-25C, I). Concentrations as low as 1 μΜ of KV-37 were capable of sensitizing 22Rvl cells to only 1 μΜ of ENZ and reduced viability by more than 50%. Either agent employed alone, did not exert any cytotoxic effect. Among cultures supplemented with 10 nM A4-AD in normal and CSS media, the synergistic drug interaction was maintained (CI = 0.33 and 0.10 respectively) (FIG. 25D-25F, 251). Quantification of degree of synergism was made using the Chou-Talalay method (44). Up to a 200-fold reduction in ENZ dosing was achieved in 22Rvl cells (FIG. 21), LNCaP cells however, due to their meager expression of AKR1C3, displayed a lack of synergistic drug effect at the concentrations tested, among both normal and
CSS media, and the interaction was merely additive (CI = 1.12 and 1.14 respectively) (FIG. 2G-I). On the contrary, co-treatment experiments did not show any adjuvant effect in either cell line cultured in normal or CSS media, with or without A4-AD supplementation (FIGS. 26A-26H). This observation suggests that the activity of AKR1C3 needs to be inhibited prior to ENZ treatment in 22Rvl cells for the therapeutic drug effect.
FIGS. 24A to 24D: Percentage cell viability of ENZ at indicated time points in (FIG. 24A) LNCaP cells cultured in indicated growth media (FIG. 24B) 22Rvl cells cultured in CSS media (FIG. 24C) 22Rvl cells cultured in CSS media supplemented with 10 nM A4-androstene-3,17-dione (FIG. 24D) 22Rvl cells cultured in normal media supplemented with 10 nM A4-androstene-3,17-dione. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 26A to 26H: Co-treatments of KV-37 with ENZ do not exert synergistic cytotoxic effects in LNCaP and 22Rvl cells. Percentage cell viability of prostate cancer cells after combination treatment with KV-37 and ENZ in (FIGS. 26A-26C) LNCaP cells at indicated time points cultured in indicated growth media (FIGS. 26D-26E) 22Rvl cells cultured in indicated growth media at 72 h and (FIGS. 26F- 26H) 22Rvl cells at indicated time points cultured in indicated growth media supplemented with 10 nM A4-androstene-3,17-dione. Data shown as mean ± SD of at least three independent experiments, n=6.
FIGS. 25A to 251. Pre-treatment with KV-37 sensitizes prostate cancer cell lines to ENZ cytotoxicity. Percentage cell viability of prostate cancer cells when pre-treated with KV-37 for 24 h followed by ENZ treatment at indicated concentrations and time points in (FIG. 25 A) 22Rvl cells cultured in normal media (FIG. 25B) and (FIG. 25C) 22Rvl cells cultured in CSS media (FIG. 25D) 22Rvl cells cultured in normal media supplemented with 10 nM A4-androstenedione (FIG. 25E) and (FIG. 25F) 22Rvl cells cultured in CSS media supplemented with 10 nM A4-androstenedione (FIG. 25G) LNCaP cells cultured in normal media and (FIG. 25H) LNCaP cells cultured in CSS media. (I) Quantification of the degree of synergism. Combination index (CI) and dose reduction index (DRI) values for KV-37 and ENZ combination treatments in prostate cancer cells. CI and DRI indices generated using CompuSyn software. Data shown as mean ± SD of at least three independent experiments, n=6.
AKR1C3 overexpression in LNCaP cells confers resistance to ENZ cytotoxicity that is reversed upon KV-37 treatment. The LNCaP 1C3 cell line, stably overexpressing AKR1C3, was employed to evaluate the activity of KV- 37 alone and in combination with ENZ to further validate AKR1C3 as the target. KV- 37 induced a dose- and time -dependent reduction in cell viability of LNCaPlC3 cells (FIG. 27A and table insert). The cytotoxic drug effect increased further as compared to 22Rvl and low AKR1C3 expressing LNCaP cells at all time points tested. These observations demonstrate that the bioactivity of KV-37 is AKRlC3-dependent and further corroborates AKR1C3 as the compound's target. Next, a dose- response curve of ENZ was evaluated in LNCaP 1C3 cells that revealed the generation of a resistant phenotype as observed in previous findings.(27) The 72 h IC50 of ENZ increased to 150 μΜ in LNCaPlC3 cells as compared to 50 μΜ in sensitive LNCaP cells (FIG. 27B and FIGS. 24A to 24D). To
overcome this drug resistance, cells were treated with a combination of KV-37 and ENZ as co-treatments for 72 h or as 24 h pre-treatments with KV-37 followed by ENZ exposure for a further 72 h. Co- treatment experiments showed a moderate degree of drug synergism (CI = 0.54) (FIG. 27C and table insert). Among the pre-treatment experiments, a very high degree of synergistic dug interaction was observed (CI = 0.14) and concentrations as low as 1 μΜ of KV-37 were able to sensitize the cells to ENZ- induced cytotoxicity. As a result, the effective ENZ concentration to induce cytotoxicity was reduced by 25-fold and yielding an IC50 of 8.19 μΜ for the drug combination (FIG. 27D and table insert). These findings strongly suggest that KV-37, by virtue of its AKR1C3 inhibiting properties, is able to re-sensitize ENZ-resistant prostate cancer cells that are high expressers of AKR1C3 towards ENZ cytotoxicity.
FIGS. 27A to 27D. AKR1C3 overexpression in LNCaP cells (LNCaPlC3) increases the cytotoxic activity of KV-37 and induces resistance to ENZ that is overcome by pre-treatment with KV-37. Percentage cell viability of LNCaPlC3 cultured in CSS media after (FIG. 27A) KV-37 treatment at indicated time points and concentrations (table insert showing the IC50 values) (FIG. 27B) ENZ treatment at indicated time points and concentrations (FIG. 27C) co-treatment with KV-37 and ENZ for 72 h and (FIG. 27D) pre-treatment with KV-37 followed by ENZ exposure for 72 h. Table insert showing the parameters for quantifying the degree of synergism. Data shown as mean ± SD of at least three independent experiments, n=6.
KV-37 induces cell death in prostate cancer cells via apoptosis. To elucidate the primary mechanism of cell death, 22Rvl and LNCaPlC3 cells cultured in CSS media were treated with increasing doses of KV- 37. After 48 and 72 h time periods post treatment, the cells were harvested and stained with AnnexinV/PI to measure apoptotic cell percentage. The percentage of the apoptotic cell population increased with KV- 37 concentration in a dose- and time -dependent manner (FIG. 28A). Consistent with the results from the cytotoxicity screening, the effect was significantly greater in the LNCaPlC3 cell line as compared to the 22Rvl cells at both time points (p = <0.05 at 25 μΜ KV-37 and <0.003 at 50 μΜ KV-37), underscoring the significance of AKR1C3 overexpression in prostate cancer. In order to further corroborate these findings, Western blot analysis was conducted after inhibitor treatment following an identical dosing schedule. The results, in agreement with AnnexinV/PI assay consistently show a dose-dependent increase in the levels of C-caspase3 and C-PARP with higher expression at 72 h as compared to 48 h incubation time, among both 22Rvl and LNCaPlC3 cell lines (FIG. 28C). Taken together, this data strongly suggests apoptosis as the primary mechanism of cytotoxicity for KV-37.
Combination of KV-37 with ENZ potentiates the degree of apoptosis in prostate cancer cells. To determine whether the effect of KV-37 to induce apoptosis in 22Rvl cells persists in the presence of ENZ, cells were pretreated with KV-37 followed by ENZ incubation for 72 h. The cells were subjected to apoptosis analysis by AnnexinV/PI co-staining. In 22Rvl cells, a combination of 10 μΜ KV-37 with 25 μΜ ENZ displayed a significant increase (p = <0.05) in apoptotic cell percentage as compared to 25 μΜ
ENZ treatment alone, whereas a combination of 25 μΜ KV-37 with 25 μΜ ENZ displayed a highly significant (p = <0.0001) increase in apoptotic cell percentage as compared to either treatments alone (FIG. 28B, left panel). Percent apoptosis was also increased in LNCaPlC3 cells wherein both 10 μΜ and 25 μΜ KV-37 plus 25 μΜ ENZ demonstrated a significant (p = <0.05 and <0.01 respectively) increase in percent apoptosis levels than with either treatment alone (FIG. 28B, right panel). These findings were further confirmed by Western blot analysis showing a greater increase in C-caspase3 and C-PARP levels in drug combination experiments performed with 10 μΜ KV-37 and 25 μΜ ENZ in 22Rvl (FIG. 28D, left panel) and LNCaPlC3 cells (FIG. 28D, right panel). These observations complement the cytotoxicity studies of the drug combinations and further strengthen the hypothesis of apoptotic cell death induced by KV-37 in prostate cancer cells.
FIGS. 28A to 28D. KV-37 induces apoptosis in prostate cancer cells alone and potentiates the degree of apoptosis when combined with ENZ. (FIG. 28A) Dose and time-dependent increase in apoptotic cell population after treatment with KV-37 in indicated cell lines as analyzed by AnnexinV/PI co-staining. (FIG. 28B) Increase in apoptotic cell population after treatment with indicated concentrations of KV-37, ENZ and a 24 h pre-treatment combination of KV-37 and ENZ in indicated cell lines analyzed 72 h post ENZ exposure by AnnexinV/PI co- staining. (FIG. 28C) Western blot showing an increase in C-caspase3 and C-PARP levels after treatment with KV-37 at indicated concentration and time points in prostate cancer cells. (FIG. 28D) Western blot showing a greater increase in C-caspase3 and C-PARP levels with a 24 h pre-treatment combination of KV-37 and ENZ in prostate cancer cells analyzed 72 h post ENZ exposure. Data shown as mean ± SD of at least three independent experiments, n=3. *, p<0.05; **, p<0.01; ****, p<0.0001; ns, non-significant.
Molecular changes following KV-37 treatment in prostate cancer cells. Western blot analysis was conducted to measure the expression levels of AKR1C3, AR and PSA. A slight decrease in the expression level of AKR1C3 was noted in 22Rvl cells at 48 h, whereas, 72 h treatment increased AKR1C3 expression, both changes were observed at 25 μΜ and 50 μΜ KV-37 concentrations (FIG. 29A). In LNCaPlC3 cells a decrease in AKR1C3 expression was noted at only 50 μΜ concentration at 48 h. On the contrary, no change in AKR1C3 expression was observed in the LNCaPlC3 cell line at all concentrations tested at 72 h post KV-37 exposure (FIG. 29B). Since, AKR1C3 serves as a co-activator for AR (45), it was prudent to measure AR levels after treatment. The inventors observed that expression of AR complements AKR1C3 levels and was found to be decreased after 48 h of treatment, while at 72 h, no change in AR levels was observed in both 22Rvl and LNCaPlC3 cell lines. A consistent decline in PSA levels in a concentration- dependent manner was observed in both cell lines at 48 and 72 h treatments. Such findings signify a beneficial therapeutic outcome in prostate cancer after treatment with KV-37 (FIG. 29A and 29B).
Combination of KV-37 with ENZ downregulates AR expression that increases after ENZ treatment. To determine the mechanism of synergistic interaction between KV-37 and ENZ, Western blot analysis was
performed. In contrast to previous reports that indicate AR degradation in 22Rvl cells (46), treatment of 25 μΜ ENZ alone increased AR expression in both 22Rvl and LNCaPlC3 cells signifying drug resistance, which in combination with 10 μΜ KV-37 reduced the expression to less than control levels (FIG. 29C and 29D). Consistent with inhibitor treatment alone, the combination produced no change in AKR1C3 expression levels. However, the combination of 10 μΜ KV-37 with 25 μΜ ENZ completely abrogates PSA expression in 22Rvl cells and reduced PSA to control levels in LNCaPlC3 cells. This data suggests apoptotic cell death as a primary mechanism of synergistic cytotoxic effect as a consequence of reduction in the activity of AKR1C3 and expression of AR.
FIGS. 29A to 29D. Molecular changes following KV-37 treatment. Treatment with KV-37 alone at indicated concentration and time points in (FIG. 29 A) 22Rvl and (FIG. 29B) LNCaPlC3 cells cultured in CSS media. Treatment with indicated concentrations of KV-37, ENZ and a 24 h pre-treatment combination of KV-37 and ENZ in (FIG. 29C) 22Rvl cells and (FIG. 29D) LNCaPlC3 cells cultured in CSS media analyzed 72 h-post ENZ exposure.
Treatment of prostate cancer cells with KV-37 does not affect AR transactivation and AR nuclear localization. KV-37 does not displace [3H]-R1881 from the AR in cell binding assays suggesting that it has no affinity for the AR. Similarly, in AR luciferase reporter gene assays no effect was seen with 10 μΜ KV-37, where 1 nM DHT is the EC50 value. The HeLa cell line which contains HeLa- AR3 A-P S A- (ARE)4-Lucl3) also expresses AKR1C3. Using an AKR1C3 substrate, 4- DAD, a blunted response is observed when compared with DHT, likely due to the conversion to T. Unfortunately, inclusion of KV- 37 in the assay resulted in interference, preventing conclusive data from being obtained.
Antitumor effect and pharmacokinetics of KV-37 in a 22Rvl xenograft model. Treatment with KV-37 at 20 mg/Kg per day in mice harboring 22Rvl xenografts significantly (p =0.0003) inhibited tumor volume by greater than 50% and tumor weight by 35% (p = 0.0179) (FIGS. 30A-30C) as compared to vehicle treated controls. No significant change in body weight was observed (FIG. 30D), indicative of a non- toxic effect. On day 34, mice were euthanized and blood collected at 10 min, 120 min and 360 min post KV-37 administration at 20 mg/Kg and pharmacokinetic parameters analyzed. The peak plasma concentration (Cmax) was determined to be 86.5 μg/mL, and Tmax, AUC, Vz/F and CL/F calculated (FIG. 30E).
FIG. 30A to 30E. KV-37 induces tumor growth inhibition in prostate cancer xenografts. Reduction in (FIG. 30A) prostate tumor volume and (FIG. 30B) prostate tumor weight in 22Rvl murine xenografts after treatment with 20 mg/Kg KV-37. (FIG. 30C) Representative images showing a reduction in tumor size after treatment with 20 mg/Kg KV-37. (FIG. 30E) Pharmacokinetic parameters determined in plasma after treatment with 20 mg/Kg KV-37.
FIGS. 31A to 31D: Indomethacin exerts a weak synergistic drug effect in LNCaPlC3 cells and KV-37 does not induce cytotoxicity alone or in combination with ENZ in WPMY-1 cells. Percentage cell
viability of LNCaPlC3 cells after (FIG. 31A) treatment with indicated concentrations of indomethacin for 96 h and (FIG. 3 IB) 24 h pre-treatment with indomethacin followed by ENZ treatment for 72 h. Percentage cell viability of WPMY-1 cells after (FIG. 31C) treatment with indicated concentrations of KV-37 for 96 h and (FIG. 3 ID) 24 h pre-treatment with KV-37 followed by ENZ treatment for 72 h. Table insert showing the quantification of the degree of synergism. Data shown as mean ± SD of at least three independent experiments, n=6.
Indomethacin displays diminished cytotoxic effect and weak synergistic drug action compared to the more potent inhibitor KV-37. Indomethacin (INDO) (AKR1C3 Ki = 8 μΜ for the E.NADPH.Indomethacin complex) has been shown to reverse ENZ resistance in prostate cancer models (27). The dose-response curve of INDO showed a weak inhibition of cell viability at 96 h incubation, IC50 = 112.8 μΜ (FIG. 31 A). Further, the inventors also administered INDO as a 24 h pre-treatment to LNCaPlC3 cells followed by ENZ exposure for 72 h. A moderate synergistic drug action was observed (CI = 0.32, DRI = 6.4) (FIG. 3 IB) as compared to the very high degree of synergism with KV-37 under identical conditions (CI = 0.14, DRI = 24.8). These findings further bolster the concept that AKR1C3 inhibition by KV-37 sensitizes ENZ-resistant prostate cancer cells to the chemotherapeutic, and is superior to indomethacin
KV-37 exhibits no cytotoxicity alone, or in combination with ENZ, in WPMY-1 prostate cells. The non- malignant prostate stromal cell line WPMY-1 was chosen to evaluate the cytotoxic activity of KV-37. Consistent with the observation that WPMY-1 cells are low expressers of AKR1C3, the reduction of cell viability induced by KV-37 was minimal with 96 h IC50 = 80 μΜ. At concentrations of 1, 10 and 25 μΜ of KV-37, those used in the study of prostate cancer cells, no reduction in WPMY-1 cell viability was observed. An observation consistent with no loss of body weight in mice xenografts upon 20 mg/Kg daily dosing. Further, pre-treatment with KV-37 for 24 h followed by ENZ exposure for 72 h resulted in no loss of cell viability (FIG. 31C and 3 ID).
Prostate tumors resistant to AA and ENZ, are characterized by overexpression of steroidogenic enzymes where AKR1C3 is the most overexpressed (26,47,48). Thus, in situ or intratumoral androgen biosynthesis is a predominant factor of therapeutic failure with AR antagonists (13,49). Overexpression of AKR1C3 by stable transfection in prostate cancer cells imparted ENZ resistance which was overcome by AKR1C3 knockdown by shRNA. Indomethacin, a weak AKR1C3 inhibitor, was also able to rescue xenografts from ENZ and AA resistance (27,28). Continuing the efforts to identify potent and selective AKR1C3 inhibitors the inventors synthesized and characterized KV-37, a nanomolar inhibitor of AKR1C3 with > 100-fold isoform selectivity and metabolic stability (38). The inventors demonstrate the synergistic effects of KV-37 with ENZ on prostate cancer cell cytotoxicity is much greater than that seen with INDO, and that the effects of KV-37 are mediated by AKR1C3 inhibition.
The inventors demonstrate that KV-37 inhibits the activity of AKR1C3 in prostate cancer cells by inhibiting the conversion of A4-androstenedione to testosterone. Crucially, KV-37 exerts a preferential
cytotoxic effect in AKR1C3 overexpressing 22Rvl and LNCaPlC3 cells as compared to the low expressing LNCaP cell line and non-malignant WPMY-1 cells. Furthermore, culture of 22Rvl cells in CSS media, that is devoid of androgens, upregulates AKR1C3 expression further and results in greater susceptibility to KV-37-induced cytotoxicity. Since the physiological androgen level in clinical castrate conditions is of the order of 1-10 nM, CSS was supplemented with 10 nM A4-androstenedione to further mimic the CRPC phenotype (43). KV-37 displayed an equally robust inhibition of cell viability in such cultures. The 22Rvl cell line is inherently resistant to ENZ, whereas LNCaP cells became resistant to ENZ when AKR1C3 was stably expressed in this cell line. Pre- treatment with KV-37 restored the sensitivity of 22Rvl cells to ENZ and provided strong synergistic effect resulting in a 200-fold reduction in chemotherapeutic dosing under conditions that mimic the CRPC disease phenotype. The strong synergistic effect was maintained in LNCaP 1C3 cells upon pre -treatment with KV-37. A moderate synergistic effect was also observed in these cells if KV-37 was co-administered with ENZ. By contrast, as LNCaP cells are low expressers of AKR1C3, only an additive effect was observed, an observation that may be ascribed to the general cytotoxicity of KV-37 towards cancer cells as evidenced previously in leukemic cell lines (38).
Insights into the mechanism of apoptosis mediated by KV-37 were obtained by measuring an increase in the apoptotic markers C-caspase3 and C-PARP, as well as a dose-dependent increase in annexinV/PI after KV-37 treatment. The observation that percent apoptotic cells were greater in LNCaPlC3 cells at any given concentration, compared to 22Rvl cells, corroborates AKR1C3 as the target of KV-37. In combination drug treatments, 25 μΜ ENZ was employed, as this corresponds to the minimum steady state plasma concentration of 12 μg/mL of the drug; the physiological concentration achieved clinically in prostate cancer patients at 150 mg/day dosing regimen (17). A sub-therapeutic concentration of KV-37 (10 μΜ), that exerts no more than 20% reduction in prostate cancer cell viability, was chosen for the drug combination treatments. In combination drug treatments, annexinV/PI staining demonstrated a significant increase in the percentage of apoptotic cells compared to either treatment alone, and was confirmed by an increase in C-caspase3 and C-PARP levels.
These data indicate that apoptotic cell death is a primary mechanism of the synergistic cytotoxic drug effect. Androgen-dependent gene expression was analyzed post KV-37 treatment and a dose-dependent reduction in PSA levels was observed. Although the activity of AKR1C3 was inhibited by KV-37, the protein expression levels either remained unchanged, or slightly increased in 22Rvl cells, post treatment, indicating a feed-back increase in AKR1C3 expression. Interestingly, the inventors observed an increase in the AR expression levels after treatment with ENZ alone in both 22Rvl and LNCaP 1C3 cell lines, which is one of the signatures of drug resistance. Combination of ENZ with KV-37 reduced AR expression to control levels or less. Further, treatment with KV-37 alone in 22Rvl murine xenografts reduced tumor volume by more than 50%, whereas no change in mouse body weight was observed. In order to compare the degree of synergistic drug interaction exhibited by the weak AKR1C3 inhibitor
indomethacin (Ki = 8 μΜ) with KV-37 (Ki = 3.0 uM), pre-treatment experiments with indomethacin were conducted in LNCaPlC3 cells followed by ENZ treatment that revealed a moderate degree of drug synergism, much lower than that achieved with KV-37. This study serves as proof-of-concept that AKR1C3 inhibition has the potential to overcome ENZ resistance and corroborates previous findings (27). Further, the inventors also analyzed the activity of KV-37 alone and in combination with ENZ in the non-malignant human prostate stromal cell line WPMY-1. No reduction in cell viability up to 25 μΜ was observed, whereas no cell viability reduction in combination experiments was noted, indicating selectivity to cells overexpressing AKR1C3.
In example 2, the inventors show that KV-37 can be used as a monotherapy for castration-resistant prostate cancer since it retards ENZ resistant prostate cancer cell growth and induces apoptosis. The structurally novel AKR1C3 inhibitor KV-37 synergizes with ENZ and re-sensitizes ENZ-resistant prostate cancer cells to the action of a chemotherapeutic agent.
Example 3. Potent and Highly Selective AKR1C3 Inhibitors with Chemotherapeutic Potentiation Effect in Acute Myeloid Leukemia (AML) and T-cell Acute Lymphoblastic Leukemia (T-ALL)
Aldo-keto reductase 1C3 (AKR1C3) catalyzes the synthesis of 9a,l i -prostaglandin (PG) F2a and PGF2a prostanoids that sustain the growth of myeloid precursors in the bone marrow. Moreover, AKR1C3 confers chemotherapeutic resistance to the anthracyclines: first-line agents for the treatment of leukemias. The enzyme is overexpressed in Acute Myeloid Leukemia (AML) and T-cell acute lymphoblastic leukemia (T ALL). Highly homologous isoforms AKR1C1 and AKR1C2 are required for normal steroid metabolism, inhibition of which is undesirable. The inventors report herein, the identification of novel AKR1C3 inhibitors that demonstrate exquisite isoform selectivity for AKR1C3 over the other closely related isoforms in the order of >2800. Biological evaluation of the inventors' isoform selective inhibitors revealed a high degree of synergistic drug action in combination with the clinical leukemia therapeutics daunorubicin and cytarabine in in vitro cell models of AML and primary patient-derived T-ALL. The developed compounds exhibited a > 100-fold dose reduction index that resulted in complete resensitization of a daunorubicin-resistant AML cell line to the chemotherapeutic and reduction of the IC50 of cytarabine from 40 nM to <1 nM.
The natural product Baccharin, extracted from honeybee propolis has been shown to potently and selectively inhibit AKR1C3 with an IC50 of 0.11 μΜ and a fold selectivity of 500 over AKR1C2.23 The inventors adopted this natural structural scaffold and have previously reported the synthesis and a preliminary structure-activity relationship (SAR) for AKR1C3 inhibition.24 The dihydrocinnamoyloxy moiety of the hit scaffold is reported as a structural prerequisite for AKR1C3 inhibition,26 and other groups have synthesized potent AR1C3 inhibitors based on this scaffold.25 However, the presence of an ester linkage provides for the hydrolytic lability of baccharin making these analogues unsuitable leads for drug discovery. Hydrolysis of the ester results in formation of the known phenol drupanin, with complete abrogation of AKR1C3 inhibitory activity (unpublished data). Theprior studies have reported the design,
synthesis and evaluation of potent AKR1C3 inhibitors bearing a more stable amide bioisostere. These AKR1C3 inhibitors exhibit a six-fold potentiation of etoposide and a ten-fold potentiation of daunorubicin in AML cell lines.27 However, the selectivity, while significantly improved above that of MPA (0.66-fold), remained relatively low (109-fold). Continuing theefforts to identify highly isoform selective AKR1C3 inhibitors the inventors herein report the discovery of a library of optimized compounds possessing >2800-fold selectivity for AKR1C3 inhibition, with retention of inhibitory potency in the nanomolar range. Further, the inventors demonstrate that the highly isoform selective AKR1C3 inhibitors provide potentiation up to 100-fold of the clinical chemotherapeutics daunorubicin and cytarabine across a panel of AML cell lines and in primary patient-derived T-ALL cells.
Chemistry. Based on the baccharin structural scaffold, four different classes of analogues were synthesized and evaluated for AKR1C3 inhibition activity and fold selectivity towards the other highly homologous AKR isoforms (Figure 1). To explore modifications of the dihydrocinnamoyloxy moiety, Class I compounds, bearing an ester or amide link to the phenyl ring with changes in the carbon spacer length were synthesized. Class I-A compounds contained methoxy or flourine substituents on the B ring. The substitution pattern of the carboxylic acid containing side chain was changed to the meta position relative to the side chain amide in Class II analogues. Similar to Class I-A compounds, class II-A analogues contained alcohol, methoxy, tosyl or flourine substitution on the B ring. In order to explore the significance of the carboxylic acid side chain, class III analogues bearing boronic acid bioisosteres were synthesized. Further, the side chain substitution pattern on the A ring was modified in Class III compounds. Finally, class IV compounds bearing a 1,3,5-all meta side chain substituent pattern on the A ring were synthesized. Class IVA compounds featured changes in spacer length and substituents on the B ring.
Figure 32 shows a SAR strategy map for the design of Baccharin derivatives of the present invention. Baccharin (1), compounds 8b-n (class I and I-A) and 7-7a (class III) were synthesized using a modified literature procedure.35 Nucleophilic substitution of commercially available 4-iodophenol (2) with prenyl bromide yielded the alkylated intermediate 3,36 which was subsequently esterified with an appropriately substituted acid chloride (4a-n) to yield ester intermediates (5a-n). Mizoroki-Heck37, 38 coupling of the aryl iodide intermediates (5a-n) with tert-butyl acrylate or vinyl boronic acid pinacol ester using Pd(OAc)2 and PPh3 as a catalyst-ligand complex produced α,β-unsaturated olefins 6a-n and 7 respectively. Chemoselective hydrolysis of the tert-butyl ester,39 of 6a-n afforded baccharin (1) and derivatives 8b-n, whereas hydrolysis of the pinacol ester afforded boronic acid 7a (Scheme 1).
Scheme 1: Synthesis of baccharin (1), substituted ester derivatives of class I, I-A and boronic acid derivatives of class III. The reaction sequence was then modified where p-iodoaniline (9) was regioselectviely brominated using a modified literature procedure that afforded 10.40 Exploiting the greater reactivity of iodide over bromide in Mizoroki-Heck38 couplings, 10 was selectively coupled with tert-butylacrylate yielding the intermediate aniline 11. Coupling with the corresponding acid chloride (12a-e) gave amide intermediates (13a-e). A palladium-catalyzed Suzuki-Miyaura41 cross-coupling reaction installed the prenyl side chain that yielded intermediates42 (14a-e) which were underwent chemoselective hydrolysis of the tert-butyl ester to afford the final compounds (15a-e) of class III (Scheme 2).
Scheme 2: Synthesis of amide bioisosteres of class I ester derivatives. The substitution pattern of the side chains on the baccharin A ring was modified in class II compounds in which the ester chain was substituted to the meta-position relative to the carboxylic acid side chain. Commercially available m- coumaric acid (16) was protected as the methyl ester and subsequently brominated, with column chromatography providing access to the desired regioselectivity in 35% yield. The brominated intermediate (17) was deprotected under basic conditions to yield compound 17a which was followed by esterification of the phenol moiety using phenylpropionyl acid chloride to yield ester derivative 18. Suzuki-Miyaura41 reaction with the appropriately substituted boronic acid pinacol ester afforded prenyl and allyl derivatives (19a, b) of class II (Scheme 3).
Scheme 3: Synthesis of meta-substituted ester derivatives of class II. The amide bioisosteres 26a-l, 28 (class II, II-A) and 24 (class III) were accessed through Mizoroki-Heck38 reaction of commercially available 4-bromo-2-iodoaniline (20) with tert-butyl acrylate or vinyl boronic acid pinacol ester to afford 21a and 21b respectively. Amide formation with the appropriate acid chloride as previously described yielded amides 23a-23j and 24. Subsequent Suzuki-Miyaura41 cross-coupling afforded intermediates 25a-25j and 27 which were chemoselectively hydrolyzed to yield final compounds 26a-26j and 28. Further, hydrolysis of the tosyl moieties in 26i and 26j, afforded 26k and 261 respectively (Scheme 4).
Scheme 4: Synthesis of meta-substituted amide derivatives of class II, II -A and boronic ester derivative 24 of class III. Commercially available 4-bromo-2-iodoaniline (29) was used as a starting material for the synthesis of compounds 33a and 33b of class III analogues. After installation of the amide chain by reaction with acid chloride as previously described, Mizoroki-Heck cross-coupling regioselectively afforded intermediate 31 due to the increased reactivity of iodide over bromide for the reaction.38 Suzuki-Miyaura41 cross coupling with substituted pinacol boronate followed by tert-butyl ester deprotection yielded compounds 33a and 33b (Scheme 5).
Scheme 5: Synthesis of 33a and 33b as amide derivatives of class III.
Further, to modify the side chain substitution pattern of the baccharin scaffold A ring to the 1,3,5 all meta-substituted pattern, 3-bromo-5-iodobenzoic acid (34) was used as the common starting material for the synthesis of amide and retroinverse amide or ester analogues. Conversion of 34 to 3-bromo-5- iodoaniline (35) was achieved by modified Curtius rearrangement, 43 followed by Boc deprotection. A reaction sequence of amide synthesis using acid chloride, followed by Pd-catalyzed cross-couplings in the order of Mizoroki-Heck reaction followed by Suzuki-Miyaura reaction afforded tert-butyl protected intermediates that were subsequently hydrolyzed to afford compounds 39a and 39b. Conversion of 34 to its acid chloride followed by esterification or amide formation using phenylethyl alcohol, or substituted primary amine, yielded the ester and amide intermediates (42, 41a-h) respectively. The reaction sequence of Mizoroki-Heck, Suzuki-Miyaura and tert-butyl ester hydrolysis was used to obtain compounds 49a-h and 50-52 of class IV and IV-A (Scheme 6).
Scheme 6: Synthesis of 1,3,5-meta-substituted derivatives (39a and 39b, 49a-h and 50-52) of class IV and IV-A. Ether analogues 58a and 58b were synthesized using 3-bromo-4-hydroxybenzoic acid (53) as the starting material which was reacted with benzyl bromide or 3-methoxy benzyl bromide to install the ether side chain. After activating the carboxylic acid to an acid chloride, a reaction sequence of amide formation followed by Mizoroki-Heck reaction and tert-butyl ester hydrolysis afforded final compounds (58a and 58b) of class IV (Scheme 7).
Scheme 7: Synthesis of 1,3,5-meta-substituted derivatives (58a and 58b) of class IV. Structure-activity relationship. Class I and I-A analogues: Consistent with the previous findings, baccharin exhibited an IC50 of 0.1 μΜ for AKR1C3 inhibition with 420-fold selectivity over AKR1C2. Removal of the dihydrocinnamoyloxy group in la (drupanin) resulted in complete abrogation of AKR1C3 inhibition activity and selectivity. Replacement of the dihydrocinnamoyloxy group (compounds 8b-h) resulted in low or sub-micromolar inhibition potency for AKR1C3, without any significant loss in selectivity. Introduction of an acetoxy group (8b) and a cyclohexylacetoxy group (8d), decreased AKR1C3 inhibitory activity and selectivity by four-fold. As compared to 1 an increase in the carbon spacer in 8c decreased AKR1C3 inhibitory activity and selectivity by two-fold and four-fold respectively. Excision of the ethyl linker to afford the benzoyloxy moiety (8f) reduced inhibition activity by three-fold and isoform selectivity to AKR1C3 by seven-fold. Replacement of the side chain ester phenyl moiety of 1 with a bulky napthyl (8e) or straight chain hexanoyloxy substituent (8h) exhibited similar inhibition profiles; decreasing the inhibition activity by three-fold with eight-fold loss of selectivity as compared to 1. Replacement of the phenethyl ring in 1 with a pyridine ring (8g) significantly reduced activity and selectivity. Comparison with the phenyl derivative (8f) indicates the pyridine ring is solely responsible, potentially due to its ability to ionize under the assay conditions, resulting in repulsive interactions in the enzyme active site. Interestingly, the corresponding amide derivatives (15a-e) were less active inhibitors than their bioisosteric counterparts (1, 8b-e). Activity ranges varied from three-fold reduction of AKR1C3 inhibitory activity (compare 1 to 15a and 8c to 15c) to eight-fold reduction (compare 8b to 15b) between ester and amide bond derivatives. Likewise, the amide derivatives, with the exception of 15a were also significantly more promiscuous between AKR1C isoforms with only moderate to minimal selectivity for the AKR1C3 isoform over the AKR1C2 variant. Fortuitously the amide derivative of
baccharin (15a) retained potency and high selectivity, IC50 = 0.326 μΜ with 172-fold selectivity for AKR1C3 over the closely related AKR1C2 isozyme (Table 1).
Table 1. Inhibitory properties of class I analogues on AKR1C3 and AKR1C2.
Introduction of electron-withdrawing fluorine substituents on the B-ring at any position (8i-j) exhibited very similar inhibition profiles to 1 and only marginally decreased the fold-selectivity. When an electron-
donating methoxy group is installed on the B-ring inhibition activity is equipotent with the parent scaffold across all substitution positions. However, a clear correlation between selectivity and substitution position is observed. When substitution occurs at the para (81) or ortho (8n) positions a twofold loss of selectivity is apparent. When substitution occurs at the meta position (8m) an inhibitor of equal selectivity to the parent compound is obtained (Table 2).
Table 2. Inhibitory properties of class I-A analogues on AKR1C3 and AKR1C2.
Class II and II-A analogues: The meta-substituted derivatives 19a, 26a and 26b showed enhanced AKR1C3 inhibitory activity. The meta-ester derivative (19a) represents the best combination of potent inhibitor (IC50 = 0.088 μΜ) with selectivity from all of the derivatives discussed thus far, with 261-fold selectivity for AKR1C3. The meta-amide derivative (26a) is the most potent AKR1C3 inhibitor identified among all the baccharin derivatives synthesized to date (IC50 = 0.066 μΜ), equipotent with the most active reported AKR1C3 enzyme inhibitors.44 Selectivity is diminished over the parent scaffold, but the compound still retains a 109-fold affinity for AKR1C3. Introduction of structural rigidity in the side chain amide via formation of a trans alkene (26b) decreased the inhibition activity for AKR1C3 (IC50 = 0.098 μΜ) as compared to 26a but resulted in increased selectivity by 1.5 fold. In comparison with the parent scaffold 26b retained equipotency with 1 but suffered a three-fold loss of selectivity. This data suggests that free rotation on this side chain is essential for high selectivity. Replacement of the prenyl chain with a 3-hydroymethylphenyl group (27) took a toll on both inhibition activity (5-fold reduction over 1) and selectivity (50-fold loss over to 1). Removal of the two methyls of the prenyl chain, to provide terminal alkene 19b, also diminished activity and selectivity suggesting that the methyls extend into an open pocket in the active site of AKR1C3 acting to confer selectivity and inhibition activity . Removal of the prenyl and ester side chains (17a) completely abrogated the bioactivity (Table 3).
Table 3. Inhibitory properties of class II analogues on AKR1C3 and AKR1C2.
Based on the enhanced potency of 26a the inventors next sought to investigate the effect of substituents on the B-ring of the parent scaffold. Such derivatives exhibited a range of activities from compounds more active than 1 (26c-h) to a reduction of approximately 1.5-fold in AKR1C3 inhibition potency (26i- 1). Introduction of fluorine displayed fairly similar potencies, comparable to 26a, with the meta substitution (26d) showing a slight advantage over ortho and para. However, the inhibition selectivity decreased by approximately 1.5-fold (26c-e). Interestingly, methoxy substition at the para positions (26f) increased isoform selectivity as compared to 26a and all substitution patterns conserved the inhibition activity (26f-26h). The inhibition profiles of tosyl (26i and 26j) and hydroxyl (26k and 261) substituted analogues were comparable to 1 with only a marginal decrease in activity, however the selectivity decreased by 5-10 fold (Table 4).
Class III analogues: Boronic acid bioisosteres of carboxylic acids may be expected to demonstrate increased binding affinity to their molecular targets on account of their ability to covalently interact with side chain amino acid residues in the binding pocket, as compared to hydrogen bonding interactions provided by their carboxylic acid counterparts.45 Bioisosteric replacement of the carboxylic acid of theparent scaffold with a pinacol boronate (7) or a boronic acid (7a) decreased AKR1C3 inhibition activity by 66- and 23-fold respectively compared to 1, with a concurrent 500-fold decrease in selectivity. Removal of the prenyl moiety (24) abrogated bioactivity in agreement with theprevious findings. Surprisingly, when the amide chain was placed ortho to the carboxylic acid group (33a) a 1.4-fold increase in activity was observed compared to 15a with comparable selectivity. Replacement of the prenyl moiety in 33a with a 3-hydroymethylphenyl moiety (33b) diminished activity and selectivity by five-fold and 12-fold respectively in comparison to 33a (Table 5).
Class IV and IV-A analogues: Arrangement of the side chains on the central ring in a 1,3,5 substitution pattern, greatly enhanced the selectivity for AKR1C3 inhibition over the homologus AKR1C1, 1C2 and 1C4 isoforms, as compared to 1. With the exception of 39b all compounds possessing this substitution pattern showed inhibitory potency in the low sub-micromolar range, with the majority exhibiting activity greater than 1 (39a and 39b, 49a-h) (Tables 6 and 7). The meta-amide 39a displayed inhibitory activity similar to 1 with a 1.5-fold increase in selectivity over the closest isoform AKR1C2, while the selectivity over other AKR isoforms was also increased. Consistent with previous observations, replacement of the prenyl moiety with 3-hydroymethylphenyl (39b) diminished activity and selectivity over 1C2 by 100-fold and 21-fold respectively. When the amide was reteroinverted (49a), the inventors gratifyingly observed an isoform selectivity >2857-fold over AKR1C2, an increase of seven-fold as compared to 1. Selectivity over the other isozymes AKR1C1 and AKR1C4 increased by 1.5 and 7-fold respectively when compared to 1. Moreover, 49a demonstrated an IC50 of 0.07 μΜ for AKR1C3, which is a 1.5-fold increase from 1. Replacement of the prenyl moiety with an allyl group (50) resulted in a seven-fold reduction in isoform selectivity and four-fold reduction in selectivity in agreement with prior data (19b). Although compound 50 did exhibit similar selectivity over AKR1C2 with a two-fold reduction in potency as compared to thehit scaffold. Replacement of the prenyl chain with a 3-hydroymethylphenyl moiety (51) again reduced AKR1C3 inhibitory activity (three-fold from 1) as well as selectivity (1.3-fold from 1), although to a lesser extent than 39b. The meta-ester analogue (52) decreased AKR1C3 inhibitory activity by 1.7-fold and selectivity over 1C2 by five-fold from its amide counterpart (49a), while still maintaining an increase in selectivity (1C2) of 1.25-fold from 1 and an equipotent inhibition activity for 1C3. The ether analogues (59a-59b) suffered great loss (18-46 fold) in inhibition selectivity over 1C2 and a decrease in inhibition activity (two - four-fold) when compared to 49a (Table 6).
Table 6. Inhibitory properties of class IV analogues on AKR1C1-AKR1C4.
AKR1C3 IC50 and fold selectivity over AKR1C2 highlighted in red. Substitutions on the B-ring were coupled with homologation of the amide chain in class IV-A analogues. Generally, all the substitutions were very well tolerated increasing the selectivity for AKR1C3 inhibition by three - seven-fold in comparison to 1 and maintaining the inhibition potency equivalent to or greater than 1 (49b-f). The 4- fluoro (49b) and 4-methoxy (49c) substitutions on the phenethyl side chain exhibited similar inhibition activity to 1, increasing the selectivity over 1C2 by three-fold. Reduction of the carbon spacer length, along with substitution with N,N-dimethyl (49h), methoxy (49d) or methyl (49g) groups at the 4-position
on the benzyl side chain further increased the inhibitory activity as compared to 1. Compounds 49h and 49d were three-fold more selective than 1, whereas 49g exhibited the best combination of potency (IC50 = 0.071 μΜ) as well as selectivity (>2800, a seven-fold increase from 1). The 4-trifuoromethoxy benzyl containing analogue (49e) exhibited similar inhibition and selectivity profile as 49b. However, movement of the -OCF3 group to the meta position (49f) diminished inhibition activity by 1.3-fold and selectivity to AKR1C2 by 1.5-fold compared to 1 (Table 7).
Table 7. Inhibitory properties of class IV-A analogues on AKR1C1-AKR1C4.
Synergistic effect of AKR1C3 inhibition with daunorubicin in AML cell lines. The hit compound baccharin (1), m-amide analogue (26a) and 1,3,5 trisubstituted analogues 49a and 49g were selected for further studies on AML cell models as they represent a collection of the most active and selective AKR1C3 inhibitors. Consistent with the inventors' previous findings27 the inhibitors did not induce any cytotoxicity in AML cell lines up to concentrations as high as 100 uM (Figs. 48A-48D, 49A-49D, and 50A-50D). Low inhibitor concentrations of 0.1, 1 and 10 μΜ were chosen to evaluate the adjuvant effect with chemotherapeutic agents.
Daunorubicin, an anthracycline used as front-line treatment for AML, experiences highest susceptibility to be metabolized by AKR1C3 among all of the anthracycline class of antitumor antibiotics.46 In order to evaluate if combination of AKR1C3 inhibitors with daunorubicin provides a synergistic effect, AKR1C3 inhibitors were dosed with daunorubicin in a fixed ratio of 100: 1 (inhibitondaunorubicin). Cell viability reduction in combination treatments of AKR1C3 inhibitors 1, 26a, 49a and 49g with daunorubicin was not detected to a level beyond what was observed with daunorubicin treatment alone (Fig. 33A-33D). The combination, when analyzed by the Chou-Talalay method, showed slight antagonistic, to merely an additive effect, in HL-60 cells with all the inhibitors tested (CI = 1.2 - 2.3), except 49a which was weakly synergistic (CI = 0.8) (Fig. 33C, Table 8).
FIGS. 33A-33D show the co-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (33A) 1 (33B) 26a (33C) 49a (33D) 49g at indicated concentrations. In KGla cells which are daunorubicin-resistant, none of the combinations with AKR1C3 inhibitors were able to reduce the cell viability by more than 50% (Fig. 34A-34D) whereas only the concentrations of 0.1-10 μΜ AKR1C3 inhibitor with 0.01 μΜ daunorubicin were able to reduce the cell viability to >50 % in THP-1 cells which are derived from a pediatric patient with M5 sub-type AML (Fig. 35A-35D). FIGS. 34A-34D shows the co-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (34A) 1 (34B) 26a (34C) 49a (34D) 49g at indicated concentrations. FIGS. 35A to 35D shows the co- treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h co-treatment of daunorubicin with (35A) 1 (35B) 26a (35C) 49a (35D) 49g at indicated concentrations.
Combination experiments as co-treatments in KGla (daunorubicin-resistant) and THP-1 cells demonstrated a weak synergistic effect with all tested inhibitors (CI = 0.2-0.8) with the exception of 1 which was additive (Table 8). Such discrepancies can be rationalized to a hydrolytic instability of 1.
Table 8. Quantification of drug interactions in AML cells after co-treatment with AKR1C3 inhibitors and daunorubicin.
Pre-treatment of AKR1C3 inhibitors for 24 h followed by daunorubicin exposure for a further 72 h enhanced cytotoxicity in HL-60 cells. A near complete cell viability reduction was seen in combinations of AKR1C3 inhibitor compounds with 0.1 μΜ daunorubicin (Fig. 5 A-D). When the effect was quantified, a weak to moderate drug synergism was noted (CI = 0.2 - 0.7) (Table 9).
FIGS. 36A to 36D shows the 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in HL-60 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (36A) 1 (36B) 26a (36C) 49a (36D) 49g at indicated concentrations. The high AKR1C3 expressing KGla cells displayed a complete abrogation of cell viability at all inhibitor combinations with daunorubicin that reached as low as 0.1 μΜ inhibitor and 0.001 μΜ daunorubicin (Fig. 37A-37D). A very strong degree of synergistic drug effect with all inhibitors was seen (CI = >0.01), providing a chemotherapeutic dose reduction of > 100-fold (DRI >100) and reducing the combination IC50 to <0.01 μΜ, indicating complete reversal of duanorubcin-resistance (Table 9).
FIGS. 37A to 37D shows the 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in KGla cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (37 A) 1 (37B) 26a (37C) 49a (37D) 49g at indicated concentrations. A complete cell viability reduction was also noted in THP-1 cells at 0.01 and 0.1 μΜ daunorubicin combination with all AKR1C3 inhibitors tested and a cell viability reduction of >50% was seen at 0.001 μΜ daunorubicin combination as compared to no cell viability reduction at 0.001 μΜ daunorubicin alone (Fig. 38A-38D). A strong synergism was established in THP-1 cells (CI = 0.06 - 0.1) where the reduction in IC50 value of daunorubicin was greater than seven-fold. The more potent and selective compounds 49a and 49g provided a greater reduction (DRI = 16) in chemotherapeutic dosing as compared to 1 and 26a (DRI = 6.6 - 8.3) (Table 9). Such high degree of drug synergism can be attributed to the inhibition of AKR1C3 activity using compounds 1, 26a, 49a and 49g for 24 h prior to daunorubicin exposure in pre-treatment experiments as compared to the co- treatments where only an additive to moderate synergism was observed.
FIGS. 38A to 38D shows the 24 h Pre-treatment of AKR1C3 inhibitors with daunorubicin in THP-1 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of (38A) 1 (38B) 26a (38C) 49a (38D) 49g at indicated concentrations.
Table 9. Quantification of drug interactions in AML cells after 24 h pre-treatment with AKR1C3 inhibitors followed by daunorubicin exposure.
Synergistic effect of AKR1C3 inhibition with cytarabine in AML cell lines. Further, the first line AML chemotherapeutic cytarabine (AraC) was combined with AKR1C3 inhibitors under a similar dosing regimen as previously described. Until now, no relationship has been established between the enzymatic activity of AKR1C3 and sensitivity of leukemic cells to AraC. The inventors hypothesized that inhibition of AKR1C3 enzymatic activity will drive the production of PGJ2 series prostanoids that will eventually activate pro-apoptotic signaling and will be instrumental in providing a synergistic drug effect with the chemotherapeutic AraC.Upon co-treatment of AKR1C3 inhibitors with AraC in HL-60 cells the trend of cell viability reduction closely matched to that of AraC alone (Fig. 39A-39D). Effects ranged from moderate synergism (1, 7) (CI = 0.5) to slight antagonism (4) (CI = 3.4) and addition (6) (CI = 1) (Table 10).
FIGS. 39A to 39D shows the Co-treatment of AKR1C3 inhibitors with AraC in HL-60 cells. Percentage cell viability after 72 h co-treatment of AraC with (39A) 1 (39B) 26a (39C) 49a (39D) 49g at indicated concentrations. Similarly, KGla and THP-1 cell lines displayed nearly identical dose inhibition profiles for all AKR1C3 inhibitors that were similar to AraC treatment alone (Fig. 40A-40D and Fig. 41 A-41D). Only an additive drug combination effect was noted that was consistent among all tested inhibitors (CI = 0.9 - 1.4) (Table 10). FIGS. 40A to 40D shows the co-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h co-treatment of AraC with (40 A) 1 (40B) 4 (40C) 6 (40D) 7 at indicated concentrations. FIGS. 41A to 41D shows the co-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h co-treatment of AraC with (41A) 1 (41B) 4 (41C) 6 (4 ID) 7 at indicated concentrations.
Table 10. Quantification of drug interactions in AML cells after co-treatment with AKR1C3 inhibitors and AraC.
The 24 h pre-treatment experiments with AKR1C3 inhibitors and AraC provided strong synergistic effects in HL-60 cells (Fig. 42A^t2D). Combination index values ranged from 0.02-0.09 that provided a dose reduction in AraC ranging from 10-33-fold (Table 11).
FIGS. 42A to 42D shows the 24 h Pre-treatment of AKR1C3 inhibitors with AraC in HL-60 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (42A) 1 (42B) 26a (42C) 49a (42D) 49g at indicated concentrations. Similar to the observations made with pre-treatment of daunorubicin, AKR1C3 inhibitor pre-treatments followed by AraC exposure, also abrogated the cell viability at all compound concentrations, displaying a very strong degree of synergistic drug effect with all inhibitors (CI = <0.01) in the high AKR1C3 expressing KGla cells. A chemotherapeutic DRI of >100-fold was noted that reduced the combination IC50 to <0.01 μΜ (Fig. 43A-43D and Table 11). FIGS. 43A to 43D shows the 24 h Pre-treatment of AKR1C3 inhibitors with AraC in KGla cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (43 A) 1 (43B) 26a (43C) 49a (43D) 49g at indicated concentrations.
Similarly, a strong synergism was observed in THP-1 cells (CI = 0.05-0.07) where the reduction in IC50 value of AraC was greater than 13-fold (Fig. 44A- 4D and Table 11). FIGS. 44A to 44D shows the 24 h Pre-treatment of AKR1C3 inhibitors with AraC in THP-1 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of (44A) 1 (44B) 26a (44C) 49a (44D) 49g at indicated concentrations.
Table 11. Quantification of drug interactions in AML cells after 24 h pre-treatment with AKR1C3 inhibitors followed by AraC exposure.
Synergistic effect of AKR1C3 inhibition in primary patient-derived T-ALL cells. For further evaluation of the adjuvant effects of AKR1C3 inhibitors, primary patient-derived T-ALL cells were chosen. The children's oncology group COG-317 cell line represents a cell line derived from samples of pediatric patients who relapsed after chemotherapy, whereas COG-329 cells were derived from samples of pediatric patients prior to chemotherapy treatment. Compound 49a, with the best combination of AKR1C3 inhibition activity and isoform selectivity, demonstrated a very strong synergistic drug effect in both COG-317 and COG-329 cell lines when co-treated with daunorubicin (DRI = 19.8-13.0 respectively) (Figs. 45 A, 45B and table insert). The DRI was greater in the relapsed T-ALL cells (COG- 317) which have been shown to overexpress the AKR1C3 target that imparts drug-resistant properties at relapse. The degree of drug synergism increased even further among pre-treatment experiments (DRI > 100) and a near complete abrogation of cell viability was noted in COG-317 cells (Fig. 45C, 45D and table insert). Both co-treatment and pre-treatments of 49a with AraC showed a very strong synergism in COG-317 cells (DRI > 100) (Fig. 45A, 45C and table insert). Among COG-329 cells the effect increased from DRI of 15.7 among co-treatments to >100 in pre-treatment experiments (Fig. 45B, 45D and table insert).
FIGS. 45A to 45D shows the synergistic effect of compound 49a with daunorubicin in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and daunorubicin in (45A) COG-317 (45B) COG-329 cells. Percentage cell viability after 72 h exposure of daunorubicin with 24 h pre-treatment of 49a in (45C) COG-317 (45D) COG-329 cells. FIGS. 45E to 45G shows the treatment of COG T-ALL cells with 49a and chemotherapeutics. Percentage cell viability after 72 h treatment of AML cells with (45E) 49a (45F) daunorubicin and (46G) AraC at indicated concentration and time points.
FIGS. 46A to 46D shows the synergistic effect of compound 49a with AraC in primary T-ALL cells. Percentage cell viability after 72 h co-treatment of 49a and AraC in (A) COG-317 (B) COG-329 cells. Percentage cell viability after 72 h exposure of AraC with 24 h pre-treatment of 49a in (C) COG-317 (D) COG-329 cells. FIGS. 46E and 46F shows the combination treatment of BMMNC cells with 49a and chemotherapeutics. Percentage cell viability of BMMNC cells after pre-treatment with 49a followed by 72 h incubation with (46E) AraC and (46F) daunorubicin at indicated concentrations.
Synergistic effect of AKR1C3 inhibition in primary bone marrow mononuclear cells (BMMNC). In order to evaluate the selective chemotherapeutic potentiation of AKR1C3 inhibitor 49a towards leukemic cells, 24 h pre-treatment experiments with 49a followed by incubation with AraC and daunorubicin were performed in primary bone marrow mononuclear cells. No chemotherapeutic potentiation was noted with both AraC and daunorubicin treatments (FIGS. 46E and 46F).
Other groups have studied different classes of inhibitors for AKR1C3 in order to develop new therapeutics for the treatment of CRPC based on NSAID scaffolds. 23, 29-31, 47-53 Although inhibitors with nanomolar potency and high selectivity to AKR1C3 have been identified from these studies, the discovery of new compounds for preclinical development is still required. Among these compounds, baccharin, a natural component extracted from Brazilian propolis, displays a high potency and inhibitory selectivity against AKR1C3.23 This discovery has made baccharin a promising hit to develop a new series of potent and specific inhibitors against AKR1C3.
In this study, the inhibitory potency and selectivity of several baccharin analogs against AKR1C3 were investigated and compared to unmodified baccharin (1). Compound 26a showed the most potent inhibition (IC50: 66 nM) whereas compound 49a displayed the highest selectivity for AKR1C3 over AKR1C2 (IC50 ratio: >2857). Analysis of crystal structures of AKR1C3.NADP+- inhibitor complexes have revealed that the ligand binding site can be dissected into five subsites: oxyanion site (consisting of catalytic residues Tyr55, Hisl l7 and cofactor NADP+), the steroid channel for the binding of steroid ligands, and three sub-pockets (SPs: SP1, SP2 and SP3 to accommodate other ligands).47, 54 In the AKR1C3.NADP+. Baccharin complex model (Fig. 2.4), 23 the carboxylate group on cinnamic acid was predicted to be bound in the SP1 pocket and could form hydrogen bonds with the side chain of Serl l8, which contributes to strong binding affinity for 1. In addition, several polar amino acids (e.g. Ser308 and Tyr319) also reside in the SP1 pocket of AKR1C3 which provide hydroxyl groups that can interact with the polar groups of these inhibitors. As such, these polar interactions also increase the selectivity of baccharin for AKR1C3 over the other AKR1C isoforms where the amino acids in the corresponding positions are Phel l8, Leu308 and Phe319 in the SP1 pocket.30, 54 The docked model predicted that the benzyl moiety of the 4-dihydrocinnamoyloxy group was located in the SP3 pocket and could form hydrophobic interactions with the side chain of Gln222 or Phe306 to provide the high binding affinity .23 In addition, recent studies have shown that polar substitutions (e.g. hydroxyl and carboxylate group) on the phenyl ring of cinnamic acid or the phenyl ring of the 4-dihydroxycinnamoyloxy group decreased the inhibition for AKR1C3 23, 55, 56. For example, the displacement of 4-dihydrocinnamoyloxy group by a hydroxyl group to form drupanin (la) resulted in a loss of inhibitory potency (IC50: 15 μΜ) and only a 7-fold selectivity for AKR1C3 versus AKR1C2. Increase or decrease of carbon spacer along with a replacement of the B-ring in class I analogues exhibited low or sub-micromolar inhibition activities (8b- 8h) but still exhibited >50-fold isoform selectivity. In order to impart hydrolytic stability to the compounds, replacement of the ester functional group with an amide was performed. The corresponding
amide bioisosteres displayed diminished activity and selectivity (15a-15e) suggesting that the rigidity of the side chain amide is not beneficial at the para-position (class I analogues). The B-ring substitutions on the dihydrocinnamoyloxy moiety were very well tolerated and only a marginal reduction of activity and selectivity was observed (8i-8n, class I-A analogues).
When the substitution pattern of the dihydrocinnamoyloxy side chain was changed to the meta-position (class II analogues 19a and 26a and 26b) the inhibitory potency increased in comparison to 1. The presence of either a carbonyl or carboxylate group is required to anchor many ligands to the oxyanion site, because it can form a strong hydrogen-bond with Tyr55 and Hisl 17 and bring the ligands into close proximity of the nicotinamide head group of the cofactor.54 Based on the model of baccharin-docked into the AKR1C3-NADP+ complex structure, the carbonyl group on the 4-dihydrocinnamoyloxy group of baccharin was found to be close to the oxyanion site and could form a H-bond interaction with Tyr55 and Hisl 17, which could contribute to the high inhibitory activity of baccharin. Thus, it seems plausible that in class II analogues the carbonyl group was now positioned in close proximity to the oxyanion site imparting enhanced potency. The presence of a side chain amide increased activity even further (compound 26a) along with exhibiting a good tolerability of B-ring substituents (class II-A analogues). Further modifying the side chain substitution pattern did not increase activity or selectivity (33a-33b). Contrary to expectation, the boronic acid analogue (7a) lost inhibition potency by more than ten-fold and abrogated isoform-selectivity (class III analogues). Similar to class II analogues, the retroinverted amide group of compounds of class IV (49a-f, 51 and 52) could maintain a stronger Hydrogen bond interaction in comparison to the straight amide analogues (39a and 39b) in the oxyanion site to provide an even superior inhibitory potency. In addition, substituting the side chains in a 1,3,5 arrangement at the A-ring places the prenyl ring in close proximity to the hydrophobic amino acid residues of the SP2 subpocket that imparts exquisite selectivity (FIG. 47). FIG. 47 shows a Baccharin SAR map.
FIGS. 48A to 48D show the treatment of HL-60 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of HL-60 cells with (48A) 1 (48B) 26a (48C) 49a (48D) 49g at indicated concentrations and time points.
FIGS. 49A to 49D show the treatment of KGla cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of KGla cells with (49 A) 1 (49B) 26a (49C) 49a (49D) 49g at indicated concentrations and time points.
FIGS. 50A to 50D shows the treatment of THP-1 cells with AKR1C3 inhibitors. Percentage cell viability after 72 h treatment of THP-1 cells with (50A) 1 (50B) 26a (50C) 49a (50D) 49g at indicated concentrations and time points.
In conclusion, the current study revealed a detailed SAR map of baccharin derivatives for inhibition of AKR1C3 and identified new classes of inhibitors (FIG. 47). The 1,3,5 meta-substituted retro-inversion amides of class IV and IV-A are disclosed as the most selective inhibitors of AKR1C3 across all known
inhibitor classes reported thus far. A first of their kind to exhibit >2857-fold selectivity for AKR1C3 inhibition with a retention of inhibitory potency in the nanomolar range. These agents carry a potential to be further developed into clinical drug candidates and span applicability across a diverse variety of malignancies that are AKR1C3 dependent.
AKR1C3 inhibitors derived from modification of a natural product have enhanced biological stability and exhibit extremely selective and potent activity. Proof-of-concept that AKR1C3 inhibitors derived from baccharin have a synergistic effect sensitizing AML and T-ALL cells to the chemotherapeutic effects of daunorubicin and AraC is demonstrated. Treatment of the non-toxic AKR1C3 inhibitors in in vitro models of acute myeloid leukemia representing various French-American-British (FAB) sub-types of AML (MO, M3 and M5), along with co-administration of a range of clinical chemotherapeutics, results in a synergistic drug action, potentiating cytotoxicity and reducing the IC50 value. This is contrary to recent reports detailing selective AKR1C3 inhibitors do not perform an adjuvant role as compared to pan- AKR1C isoform inhibitors.57 This observation is further extended to the primary patient-derived T-ALL cells where a very strong potentiation of the effects of daunorubicin and AraC was established.
These results demonstrate that a strategy developing small molecule AKR1C3 inhibitors may yield powerful adjuvant agents for the synergistic treatment of leukemia with chemotherapeutic drugs, especially since favorable toxicity and pharmacokinetic properties can exist within the baccharin structure. In contrast to previously reported studies, potent and selective inhibition of AKR1C3 results in potentiation of the clinical chemotherapeutics: etoposide, daunorubicin and AraC in multiple AML cell lines. The identified highly potent and selective derivatives represent valuable lead compounds to understand the structural features required for AKR1C3 inhibitory activity and selectivity, and how these properties interrelate to further theunderstanding of the role which AKR1C3 plays to enable potentiation of chemotherapeutic effect within AML pathophysiology. Further development of the inhibitors describe herein represents a significant drug discovery opportunity for the identification of potent adjuvants to enhance the therapeutic index of chemotherapeutics, in the hope of availing this treatment regime to pediatric and geriatric AML patients.
Enzyme Inhibition Assay: A detailed account of the chemistry procedures and characterizations (1H, 13C NMR, and HRMS) was performed. The enzyme inhibition screen for all compounds was perfbmed in collaboration with the Penning lab at the University of Pennsylvania. (S)-(+)- 1,2,3, 4-tetrahydro-l- naphthol (S-tetralol) was purchased from Sigma-Aldrich (St. Louis, MO). Nicotinamide adenine dinucleotide (NAD+) and nicotinamide adenine dinucleotide phosphate (NADP+) were purchased from Roche Diagnostics (Indianapolis, IN). Homogeneous recombinant enzymes AKR1C3 and AKR1C2 were prepared and purified as previously described.28 The specific activities of AKR1C3 and AKR1C2 for the oxidation of S-tetralol are 2.0 and 1.5 μιηοΐ min-1 mg-1, respectively.
Assay of enzyme activity: The dehydrogenase activities of AKR1C3 and AKR1C2 were determined by measuring the UV absorption of NADH formation at 340 nm using Beckman DU640 spectrophotometer.
A typical assay solution contained 100 mM potassium phosphate pH 7.0, 2.3 mM NAD+, 3.0 mM (S)- (+)-l,2,3,4-tetrahydro-l-naphthol (S-tetralol), 4% acetonitrile (v/v). The mixtures were incubated at 37 °C for 3 min followed by adding a serial dilution of AKR1C3 or AKR1C2 solution to a final volume of 1 mL to initiate the reaction. After continuously monitoring for 5 min, the increase in UV absorption using different concentrations of enzyme were recorded to calculate the initial velocity and determine enzyme specific activity.
IC50 value determination: The inhibitory potency for each compound was represented by IC50 value and measured as described before 29-31. The IC50 value of baccharin and baccharin analogs was determined by measuring their inhibition on the NADP+ dependent oxidation of S-tetralol catalyzed by AKR1C3 or AKR1C2. The concentration of S-tetralol used in this assay for AKR1C3 and AKR1C2 was 165 μΜ and 15 μΜ respectively, which was equal to the Km value for each enzyme isoform in order to make a direct comparison of IC50 values. The IC50 value of each compound was acquired from a single experiment with each inhibitor concentration run in quadruplicate and directly calculated by fitting the inhibition data to an equation [y = (range) / [1 + (I/IC50)S] + background] using Grafit 5.0 software. In this equation, "range" is the fitted uninhibited value minus the "background", and "S" is a slope factor. "I" is the concentration of inhibitor. The equation assumes that y falls with increasing "I".
Adjuvant assay: HL-60 (ATCC® CCL-240TM), KGla (ATCC® CCL-246.1TM) and THP-1 (ATCC® TIB-202™) cells were procured from ATCC. Isocove's Modified Dulbecco's Media (IMDM) supplemented with 20% fetal bovine serum (FBS) and penicillin/streptomycin (1%) was used to culture HL-60 and KGla cells whereas THP-1 cells were cultured using Roswell Park Memorial Institute (RPMI)-1640 medium, supplemented with 20% fetal bovine serum (FBS), 0.05 mM 2-mercaptoethanol and penicillin/streptomycin (1%). Cells were maintained at a density of 0.1-1 x 106 cells/mL under 5% C02 at 37 oC. To screen the test compounds, cells were seeded at a density of 0.1 x 106 cells/mL in 96- well plates containing 100 μL cell suspension per well. Stock solutions of the test compounds, etoposide, daunorubicin and cytarabine (AraC) were prepared in DMSO. Cells were treated at the indicated concentrations of test compounds with or without the chemotherapeutic agents, limiting the final DMSO concentration to less than 1%. After incubation at 37 OC, 5% C02 for 24, 48, 72 or 96 h, 20 μL of MTS reagent (CellTiter 96® AQueous One Solution Reagent) was added to each well and incubated at the above mentioned conditions for 2-3 h. Plates were read at OD 490 nm on a plate reader and the viability of cells were plotted as percentage of controls.
Quantification of the degree of synergism: To quantify the degree of synergism, the results of the co- treatment and pre-treatment experiments were analyzed by CompuSyn software (Paramus, NJ) based on the median effect principle or 'Chou-Talalay' method.32 The method is based on the median-effect equation that encompasses the Michaelis-Menten, Hill, Henderson-Hasselbach, and Scatchard equations to provide combination and dose reduction indices.33 The combination index (CI) and dose reduction index (DRI) values were calculated at a constant ratio of chemotherapeutic to AKR1C3 inhibitor at 50%
cytotoxic effect (Fa = 0.5). CompuSyn software was used to generate the CI and DRI values. CK1, synergism; CI> 1.1, antagonism; CI=1.1, additive.34
Western blotting: HL-60 and KGla cells were treated with compound (4) over a period of 48 and 72 h at indicated concentrations after which they were harvested and pelleted. The whole cell lysates were prepared in radioimmunoprecipitation assay RIPA buffer containing protease and phosphatase inhibitors (1 mM PMSF, 38 μg/mL aprotinin, 2.5 mM Na3V04). Samples were incubated on ice for 30 min after which they were sonicated, centrifuged (16000 x g) and supernatant collected. Protein concentration in each sample was estimated following bicinchoninic acid BCA assay protocol by comparing with the BSA standards (PierceTM BCA protein kit). Protein samples (40 μg ) containing loading dye (7 μΐ.) were loaded onto 12% SDS polyacrylamide gel and electrophoresed (80 V, 2 hr). Transferred onto a PVDF membrane overnight (25 V, 4 oC). The membrane was blocked with 5 % non-fat milk (1 h) and probed with human anti-AKRlC3 mouse monoclonal antibody (1:500, R&D Systems, MAB7678) and corresponding horseradish peroxidase (HRP) conjugated anti-mouse secondary antibody followed by immunodetection using VersaDocTM (MP 5000). Membrane was stripped and re-probed for β-actin (1 :5000, Sigma-Aldrich, A5441). Quantity One® software was used to analyze the band intensities and fold change in AKR1C3 enzyme expression was determined based on β-actin controls.
General Chemistry Procedures.
All reactions were carried out in oven- or flame-dried glassware under positive nitrogen pressure unless otherwise noted. Reaction progress was monitored by thin-layer chromatography (TLC) carried out on silica gel plates (2.5 cm x 7.5 cm, 200 μιη thick, 60 F254) and visualized by using UV (254 nm) or by potassium permanganate and/or phosphomolybdic acid solution as indictor. Flash column chromatography was performed with silica gel (40-63 μιη, 60 A) using the mobile phase indicated or on a Teledyne Isco (CombiFlash Rf 200 UV/Vis). Commercial grade solvents and reagents and solvents were purchased from Fisher Scientific (Houston, TX), Sigma Aldrich (Milwaukee, WI) or for Prenyl boronic acid pinacol ester, Santa Cruz Biotechnology (Dallas, TX) and were used without further purification except as indicated. Anhydrous solvents were purchased from Across Organics and stored under and atmosphere of dry nitrogen over molecular sieves.
1H, 13C, COSY, HMQC and DEPT NMR spectra were recorded in the indicated solvent on a Bruker 400 MHz Advance III HD spectrometer at 400 and 100 MHz for 1H and 13C respectively with TMS as an internal standard. Multiplicities are indicated by s (single), d (doublet), dd (doublet of doublets), t (triplet), q (quartet), m (multiplet), br (broad). Chemical shifts (δ) are reported in parts per million (ppm), and coupling constants (J), in hertz. High resolution mass spectroscopy was performed on a LC/MS IT- TOF (Shimadzu) using an ESI source conducted at the University of Texas at Arlington, Shimadzu Center for Advanced Analytical Chemistry. High-pressure liquid chromatography was performed on a Dynamax HPLC system installed with a Varian pro star UV detector with a Phenomenex® Luna (CI 8 100 A, 250x4.6 mm) column. All samples were assessed to be of >96% purity.
General procedure A: Synthesis of ester intermediates:
To a solution of 4-iodo-2-(3-methylbut-2-en-l-yl)phenol (7) (1 equiv) in DCM (5 mL) was added DMAP (0.1 equiv) followed by addition of substituted acid chloride and NEt3 (1.5 equiv). The mixture was stirred overnight at RT. Saturated NaHC03 was added and the layers seperated. The organic layer was washed with H20, dried (Na2S04), filtered and concentrated. The crude product was purified by column chromatography using Hexane/EtOAc gradient (10: 1, 4: 1, 2: 1) and solvent evaporated in vacuo to provide pure esterified compounds.
General procedure B: t-Bu ester hydrolysis:
To a solution of substituted tert-butyl intermediates in toluene was added chromatography grade silica gel and the mixture was refluxed with vigorous agitation overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over celite pad using 10% methanol in DCM as the solvent. The crude product was purified by column chromatography using DCM/MeOH gradient (20: 1, 10: 1) and solvent evaporated in vacuo to provide the final componds.
4-iodo-2-(3-methylbut-2-en-l-yl)phenol (3): To a solution of p-iodophenol (2 g, 9.08 mmol) in toluene (20 ml) was added NaH (60% disopersion in mineral oil, 540 mg, 13.62 mmol) when the effervescence ceased 3,3-dimethylallyl bromide (1.1 ml, 9.5 mmol) was added and the reaction was stirred at room temperature overnight. The reaction mixture was acidified to pH 1 with AcOH, washed with H20, extracted with DCM, dried ( a2S04), filtered and concentrated. Purified by column chromatography (5- 10% Et20:hexane) and solvent evaporated in vacuo to provide the title compound as a yellow oil (1.43 g, 4.96 mmol, 55%).
Rf: 0.5 (Hexane:EtOAc = 4: 1). 1H NMR (400 MHz; CDC13): δ 1.8 (6H, d, J = 8.0 Hz, CH3), 3.33 (2H, d, J = 6.88 Hz, CH2), 5.31 (1H, t, J = 6.6 Hz, CH), 5.46 (1H, s, -OH), 6.59 (1H, d, J = 8.2 Hz, Ar-H), 7.41 (2H, s, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.8, 29.3, 82.7, 117.9, 120.69, 129.7, 135.4, 136.1. 138.3, 154.2.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl-3-phenylpropanoate (5a): Title compound was obtained following genral procedure A using 3-phenyl propionyl chloride (495 mg, 2.93 mmol) as a white solid (490 mg, 1.16 mmol, 60%).
1H NMR (400 MHz; CDC13): δ 1.66 (3H, s, CH3), δ 1.76 (3H, s, CH3), δ 2.91 (2H, t, J = 7.5 Hz, CH2) δ 3.09 (4H, t, J = 6.0 Hz, CH2CH2), δ 5.16 (1H, t, J = 7.1 Hz, CH), δ 6.69 (1H, d, J = 8.3 Hz, ArCH), δ 7.21-7.36 (6H, m, ArCH), δ 7.53 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 17.76, 25.85, 28.30, 30.24, 30.93, 35.68, 36.38, 90.49, 120.70, 124.40, 126.68, 128.44, 128.78, 133.86, 135.94, 136.32, 138.80, 140.03, 148.64, 170.95
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl acetate (5b): Title compound was obtained following genral procedure A using acetyl chloride (0.122 ml, 1.7 mmol) ) as a white solid (350 mg, 1.06 mmol, 92%).
1H NMR (400 MHz; CDC13): δ 1.70 (3H, s, CH3), δ 1.76 (3H, s, CH3), δ 2.31 (3H, s, OCH3), δ 3.19 (2H, d, J = 7.1 Hz, CH2), δ 5.19 (1H, t, J = 7.1 Hz, CH), δ 6.79 (1H, d, J = 8.0 Hz, ArCH), δ 7.55 (2H, d, J = 9.4 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 18.6, 20.9, 25.8, 28.4, 90.5, 120.6, 123.8, 124.3, 133.8, 135.9, 139.0, 148.8, 168.4
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl-4-phenylbutanoate (5c): Title compound was obtained following genral procedure A using 4-phenyl butanoyl chloride (190 mg, 1.1 mmol) as a yellow oil (591 mg, 1.3 mmol, 85%).
1H NMR (400 MHz; CDC13): δ 1.67 (3H, s, CH3), 1.73 (3H, s, CH3), 2.05 - 2.14 (2H, m, CH2), 2.59 (2H, t, J = 7.5 Hz, CH2), 2.77 (2H, t, J = 7.6 Hz, CH2), 3.18 (2H, d, J = 7.1 Hz, CH2), 5.18 (1H, t, J = 7.1 Hz, CH), 6.76 (1H, d, J = 7.9 Hz, Ar-H), 7.22 - 7.25 (3H, m, Ar-H), 7.31 - 7.35 (2H, m, Ar-H), 7.52 - 7.54 (2H, m, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 26.4, 28.4, 33.4, 35.0, 90.4, 120.6, 124.2,125.2, 126.1, 128.2, 128.4, 129.0, 134.0, 135.9, 136.1, 138.8, 141.0, 148.7, 171.5.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 2-cyclohexylacetate (5d): Title compound was obtained following genral procedure A using cyclohexylacetyl chloride (240 mg, 1.5 mmol) as a yellow oil (390 mg, 0.9 mmol, 99%).
1H NMR (400 MHz; CDC13): δ 0.97-1.36 (7H, m, cyc-H), 1.69 (3H, s, CH3), δ 1.74 (3H, s, CH3), 1.81- 1.95 (4H, m, cyc-H), 2.47 (2H, d, J = 7 Hz, CH2), 3.24 (2H, d, J = 7.1 Hz, CH2), 5.23 (1H, t, J = 7.1 Hz, CH), 6.77 (1H, d J = 8.2 Hz, Ar-H), 7.51 - 7.54 (2H, m, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 25.9, 26.0, 28.3, 33.0, 34.9, 41.9, 90.3, 120.7, 124.3, 134.0, 135.8, 136.1, 138.7, 148.8, 171.1.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 2-(naphthalen-l-yl)acetate (5e): Title compound was obtained following genral procedure A using naphthylacetyl chloride (210 mg, 1.1 mmol) as a yellow oil (256 mg, 0.6 mmol, 78%).
Rf: 0.84 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (3H, s, CH3), 1.68 (3H, s, CH3), 2.92 (2H, d, J = 7.2 Hz, CH2), 4.33 (2H, s, CH2), 4.98 (1H, t, J = 7.2 Hz, CH), 6.73 (1H, d, J = 11.3 Hz, Ar-H), 7.35 - 7.60 (6H, m, Ar-H), 7.83 - 7.92 (2H, m, Ar-H), 8.12 (1H, d, J = 11.7 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.1, 39.3, 90.9, 120.5, 123.6, 124.1, 125.5, 126.0, 126.6, 128.3, 128.5, 128.9, 129.7, 132.0, 133.8, 133.9, 135.8, 136.2, 138.7, 148.7, 169.6.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl benzoate (5f): Title compound was obtained following genral procedure A using benzoyl chloride as a yellow oil (270 mg, 0.6 mmol, 71%).
Rf: 0.71 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.56 (3H, s, CH3), 1.69 (3H, s, CH3), 3.25 (2H, d, J = 7.2 Hz, CH2), 5.20 (1H, t, J = 7.2 Hz, CH), 6.92 (1H, d, J = 8.0 Hz, ArCH), 7.52 - 7.69 (6H, m, ArCH), 8.21 (1H, d, J = 8.2 Hz, ArCH).
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl pyridine-4-carboxylate (5g): Title compound was obtained following genral procedure A using THF (7 mL) and isonicotinic acid chloride (425 mg, 3 mmol) as a brown oil (650 mg, 1.6 mmol, 73 %).
1H NMR (400 MHz; CDC13): δ 1.53 (3H, s, CH3), 1.64 (3H, s, CH3), 3.22 (2H, d, J = 6.6 Hz, CH2), 5.18 (1H, t, J = 6.5 Hz, CH), 6.88 (1H, d, J = 8.3 Hz, Ar-H), 7.55 (1H, d, J = 8.4 Hz, Ar-H), 7.59 (1H, s, Ar-H), 7.96 (2H, d, J = 4.0 Hz, Ar-H), 8.84 (2H, d, J = 3.4 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.6, 28.8, 67.9, 65.8, 91.1, 120.5, 123.1, 124.1, 134.0, 136.0, 136.2, 136.3, 139.1, 148.6, 150.8, 163.2.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl hexanoate (5h): Title compound was obtained following general procedure A using hexanoyl chloride (135 mg, 1.0 mmol) as a yellow oil.
1H NMR (400 MHz; CDC13): δ 1.36 - 1.43 (7H, m, CH3 and CH2), 1.50 - 1.62 (2H, m, CH2), 1.69 (3H, s, CH3), 1.76 (3H, s, CH3), 2.57 (2H, d, J = 7.4 Hz, CH2), 3.18 (2H, d, J = 7.0 Hz, CH2), 5.19 (1H, t, J = 7.2 Hz, CH), 6.77 (1H, d, J = 8.3 Hz, ArCH), 7.52 - 7.54 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 13.9, 22.3, 23.5, 24.6, 28.3, 31.3, 3.1, 90.3, 120.7, 124.2, 133.9, 135.9, 136.1, 138.7, 148.8, 171.8
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 3-(4-fluorophenyl)propanoate (5i): Title compound was obtained following genral procedure A using 3-(2-fluorophenyl)propanoyl chloride (260 mg, 1.4 mmol) as a yellow oil (280 mg, 0.63 mmol, 65 %).
Rf: 0.69 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.66 (3H, s, CH3), 1.75 (3H, s, CH3), 2.89 (2H, t, J = 7.5 Hz, CH2), 3.06 (4H, t, J = 7.2 Hz, CH2), 5.14 (1H, t, J = 7.2 Hz, CH), 6.69 (1H, d, J = 8.6 Hz, ArCH), 7.02 (2H, d, J = 8.7 Hz, ArCH), 7.22-7.26 (2H, m, ArCH), 7.50 - 7.53 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.2, 30.0, 35.8, 90.6, 115.2, 115.5, 120.5, 124.2, 129.8, 135.9, 138.8, 138.9, 148.6, 160.4, 162.8, 170.8.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 3-(3-fluorophenyl)propanoate (5j): Title compound was obtained following genral procedure A using 3-(3-fluorophenyl)propanoyl chloride (260 mg, 1.4 mmol) as a yellow oil (430 mg, 1 mmol, 99 %).
1H NMR (400 MHz; CDC13): δ 1.66 (3H, s, CH3), 1.75 (3H, s, CH3), 2.90 (2H, t, J = 7.7 Hz, CH2), 3.0 (4H, t, J = 7.4 Hz, CH2), 5.15 (1H, t, J = 7.2 Hz, CH), 6.70 (1H, d, J = 7.6 Hz, ArCH), 6.92 - 7.0 (2H, m, ArCH), 7.05 (1H, d, J = 7.6 Hz, ArCH), 7.27 - 7.32 (1H, m, ArCH), 7.51 - 7.53 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.2, 30.5, 35.3, 90.6, 115.2, 115.4, 120.5, 124.1, 130.0, 134.0, 135.0, 138.8, 142.4, 148.6, 160.4, 161.7, 170.7.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 3-(2-fluorophenyl)propanoate (5k): Title compound was obtained following genral procedure A using 3-(2-fluorophenyl)propanoyl chloride (260 mg, 1.4 mmol) as a yellow oil (430 mg, 1 mmol, 99 %).
1H NMR (400 MHz; CDC13): δ 1.70 (3H, s, CH3), 1.79 (3H, s, CH3), 2.94 (2H, t, J = 7.9 Hz, CH2), 3.08 - 3.14 (4H, m, CH2), 5.19 (1H, t, J = 7.2 Hz, CH), 6.74 (1H, d, J = 8.3 Hz, ArCH), 6.81 - 6.91 (3H, m, ArCH), 7.28 (1H, t, J = 7.8 Hz, ArCH), 7.53 - 7.57 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.8, 28.3, 30.9, 35.7, 90.6, 111.8, 114.2, 120.7, 124.2, 129.6, 133.9, 136.0, 136.2, 138.8, 141.5, 148.7, 159.8, 170.9.
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 3-(4-methoxyphenyl)propanoate (51): Title compound was obtained following genral procedure A using 3-(4-methoxyphenyl)propanoyl chloride (200 mg, 1 mmol) as a yellow oil (420 mg, 0.9 mmol, 93 %).
Rf: 0.75 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.69 (3H, s, CH3), 1.78 (3H, s, CH3), 2.90 (2H, t, J = 7.5 Hz, CH2), 3.03 - 3.11 (4H, m, CH2), 3.82 (3H, s, CH3), 5.18 (1H, t, J = 7.2 Hz, CH), 6.72 (1H, d, J = 8.3 Hz, ArCH), 6.90 (2H, d, J = 8.6 Hz, ArCH), 7.22 (2H, d, J = 8.6 Hz, ArCH), 7.52 - 7.56 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.8, 28.2, 30.1, 36.0, 55.2, 90.6, 114.0, 120.7, 124.3, 129.3, 132.0, 133.9, 135.9, 136.2, 138.7, 148.7, 158.2, 171.0
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 3-(3-methoxyphenyl)propanoate (5m): Title compound was obtained following genral procedure A using 3-(3-methoxyphenyl)propanoyl chloride (300 mg, 1.5 mmol) as a yellow oil (280 mg, 0.62 mmol, 62 %).
Rf: 0.71 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.70 (3H, s, CH3), 1.79 (3H, s, CH3), 2.94 (2H, t, J = 7.5 Hz, CH2), 3.00 - 3.14 (4H, m, CH2), 3.84 (3H, s, CH3), 5.20 (1H, t, J = 7.2 Hz, CH), 6.74 (1H, d, J = 8.3 Hz, ArCH), 6.81 - 6.91 (3H, m, ArCH), 7.28 (1H, d, J = 8.0 Hz, ArCH), 7.53 - 7.57 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.8, 28.3, 30.9, 35.7, 55.1, 90.6, 111.8, 114.2, 120.7, 124.2, 129.6, 133.9, 136.0, 136.2, 138.8, 141.5, 148.7, 159.8, 170.9
4-iodo-2-(3-methylbut-2-en-l-yl)phenyl 3-(2-methoxyphenyl)propanoate (5n): Title compound was obtained following genral procedure A using 3-(2-methoxyphenyl)propanoyl chloride (300 mg, 1.5 mmol) as a yellow oil (320 mg, 0.71 mmol, 71%).
1H NMR (400 MHz; CDC13): δ 1.73 (3H, s, CH3), 1.80 (3H, s, CH3), 2.94 (2H, t, J = 7.5 Hz, CH2), 3.11 - 3.18 (4H, m, CH2), 3.90 (3H, s, CH3), 5.22 (1H, t, J = 7.2 Hz, CH), 6.76 (1H, d, J = 8.3 Hz, ArCH), 6.92 - 7.00 (3H, m, ArCH), 7.29 - 7.31 (1H, m, ArCH), 7.54 - 7.58 (2H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.8, 26.2, 28.4, 34.1 55.2, 90.4, 110.3, 120.5, 120.8, 124.4, 127.9, 128.2, 133.8, 135.9, 136.2, 138.8, 148.9, 157.5, 171.4
tert-butyl(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-[(3-phenylpropanoyl)oxy]phenyl]prop-2-enoate (6a): To a solution of 5a (490 mg, 1.16 mmol) in anhydrous toluene (7 ml) was added PPh3 (46 mg, 0.175 mmol), Pd(OAc)2 (47 mg, 0.09 mmol) and NEt3 (0.245 ml, 1.75 mmol) the mixture was sitirred and t-Bu acrylate (0.275 ml, 1.75 mmol) was added to the flask which was refluxed overnight. The reaction mixture was washed with a saturated solution of NH4C1, water and extracted over DCM, dried over Na2S04 and filtered. Purified with silica gel flash chromatography using eluent system as Hexane:EtOAc = 60: 1, 30: 1, 20: 1 and solvent evaporated in vacuo to provide the title compound (280 mg, 0.66 mmol, 57%).
Rf : 0.6 (Hexane:EtOAc = 4: 1). 1H NMR (400 MHz, CDC13): δ 1.55 (9H, s, C02t-Bu), 1.69 (3H, s, CH3), 1.76 (3H, s, CH3), 2.93 (2H, t, J = 7.4 Hz, CH2), 3.10 (2H, t, J = 7.6 Hz, CH2), 3.16 (2H, d, J = 7 Hz, CH2), 5.20 (1H, t, J = 7 Hz, CH), 6.31 (1H, d, J = 15.9 Hz, CH), 6.96 (1H, d, J = 8.7 Hz, ArCH) 7.24-7.36 (7H, m, ArCH) 7.55(1H, d, J = 16 Hz, CH). 13C NMR (100 MHz, CDC13): δ 17.8, 25.7, 28.2, 28.5, 29.7, 30.9, 35.8, 80.5, 120.0, 120.35, 121.0, 121.9, 122.6, 126.5, 128.3, 128.6, 129.0, 129.7, 132.6, 133.7, 134.0, 139.9, 142.3, 142.8, 150.0, 166.2, 171.0. M/Z ESI: 443.2 [M+Na]+ (100%), 444.3(35%)
tert-butyl (2E)-3-[4-(acetyloxy)-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoate (6b): To a solution of 5b (350 mg, 1.0 mmol) in dry toluene (6.5 mL) was added PPh3 (42 mg, 0.2 mmol), Pd(OAc)2 (47.5 %, 47 mg, 0.1 mmol) and NEt3 (0.2 mL, 1.6 mmol) the mixture was sitirred for 10 mins and tert-butyl acrylate (0.2 mL, 1.6 mmol) was added to the flask which was refluxed overnight. The reaction was allowed to cool and was washed with saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 30: 1, 20: 1) provided the title compound as a transparent oil (100 mg, 0.3 mmol, 29 %).
Rf: 0.90 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.77 (3H, s, CH3), 2.33 (3H, s, C02CH3), 3.25 (2H, d, J = 7.1 Hz, CH2), 5.23 (1H, t, J = 7 Hz, CH), 6.31 (1H, d, J = 15.9 Hz, CH), 7.05 (1H, d, J = 8.8 Hz, Ar-H), 7.37 (2H, s, Ar-H), 7.56 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 20.8, 25.7, 28.7, 60.3, 80.5, 120.1, 121.0, 122.7,
126.5, 129.7, 132.6, 133.7, 134.0, 142.8, 150.1, 166.2, 169.1. M/Z ESI: 215 (100%), 353.1 0 [M+Na]+ (50%)
4-[(lE)-3-(tert-butoxy)-3-oxoprop-l-en-l-yl]-2-(3-methylbut-2-en-l-yl)phenyl 4-phenylbutanoate (6c): To a solution of 5c (591 mg, 1.3 mmol) in dry toluene (6.5 mL) was added PPh3 (42 mg, 0.2 mmol), Pd(OAc)2 (47.5 %, 47 mg, 0.1 mmol) and NEt3 (0.2 mL, 1.6 mmol) the mixture was sitirred for 10 mins and tert-butyl acrylate (0.3 mL, 2.0 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 20: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (260 mg, 0.6 mmol, 44 %).
Rf: 0.88 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.67 (3H, s, CH3), 1.76 (3H, s, CH3), 2.05 - 2.14 (2H, m, CH2), 2.59 (2H, t, J = 7.5 Hz, CH2), 2.77 (2H, t, J = 7.6 Hz, CH2), 3.24 (2H, d, J = 7.1 Hz, CH2), 5.22 (1H, t, J = 7.1 Hz, CH), 6.31 (1H, d, J = 15.9 Hz, CH), 7.02 (1H, d, J = 8.9 Hz, Ar-H), 7.22 - 7.25 (3H, m, Ar-H), 7.31 - 7.39 (4H, m, Ar-H), 7.56 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 26.4, 28.2, 28.6, 33.4, 35.0, 80.4, 120.0, 121.0, 122.7, 126.1, 126.5, 128.4, 129.7, 132.5, 133.8, 134.0, 141.0, 142.8, 150.1, 166.3, 171.6.
tert-butyl (2E)-3-{4-[(2-cyclohexylacetyl)oxy]-3-(3-methylbut-2-en-l-yl)phenyl}prop-2-enoate (6d): To a solution of 5d (390 mg, 0.9 mmol) in dry toluene (4 mL) was added PPh3 (26 mg, 0.1 mmol), Pd(OAc)2 (47.5 %, 25 mg, 0.05 mmol) and NEt3 (0.21 mL, 1.5 mmol) the mixture was sitirred for 10 mins and tert-Butyl acrylate (0.21 mL, 1.4 mmol) was added and the reaction refluxed overnight. The reaction was allowed to cool, washed with saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a transpaent oil (370 mg, 0.85 mmol, 95 %).
1H NMR (400 MHz; CDC13): δ 0.90-1.46 (7H, m, cyc-H), 1.55 (9H, s, C(CH3)3), 1.84 (3H, s, CH3), 1.96 (3H, s, CH3), 1.84-1.96 (4H, m, cyc-H), 2.47 (2H, d, J = 7 Hz, CH), 3.24 (2H, d, J = 7.1 Hz, CH2), 5.23 (1H, t, J = 7.1 Hz, CH), 6.31 (1H, d, J = 15.9, CH), 7.04 (1H, d J = 8.8 Hz, Ar-H), 7.39-7.36 (2H, m, Ar-H), 7.55 (1H, d, J = 15.9, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 26.0, 26.1, 28.2, 28.5, 33.0, 34.9, 42.0, 80.5, 119.9, 121.0, 122.7, 126.4, 129.7, 132.4, 133.7, 134.0, 142.9, 150.1, 166.3, 171.2.
tert-but l(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-{[2-(naphthalen-l-yl)acetyl]oxy}phenyl^
(6e): To a solution of 5e (256 mg, 0.6 mmol) in dry toluene (6.5 mL) was added PPh3 (42 mg, 0.2 mmol), Pd(OAc)2 (47.5 %, 47 mg, 0.1 mmol) and NEt3 (0.2 mL, 1.6 mmol) the mixture was sitirred for 10 mins and tert-Butyl aery late (0.1 mL, 0.8 mmol) was added to the flask and the reaction refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, water, extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a transparent oil (66.1 mg, 0.1 mmol, 17
%).
1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.56 (3H, s, CH3), 1.69 (3H, s, CH3), 2.99 (2H, d, J = 7.1 Hz, CH2), 4.35 (2H, s, CH2), 5.02 (1H, t, J = 7.2 Hz, CH), 6.29 (1H, d, J = 15.9 Hz, CH), 6.99 (1H, d, J = 8.1 Hz, Ar-H), 7.31 - 7.33 (2H, m, Ar-H), 7.47 - 7.61 (5H, m, Ar-H and CH), 7.85 - 7.92 (2H, m, Ar-H), 8.14 (1H, d, J = 8.6 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.7, 25.6, 28.2, 28.3, 39.3, 80.5, 120.0, 120.7, 122.5, 123.6, 125.5, 125.9, 126.3, 126.5, 128.3, 128.4, 128.8, 129.5, 129.8, 132.0, 132.5, 133.5, 133.9, 134.1, 142.8, 150.0, 166.2, 169.6.
(E)-4-(3-(tert-butoxy)-3-oxoprop-l-en-l-yl)-2-(3-methylbut-2-en-l-yl)phenyl benzoate (6f): To a sitirred solution of 5f (270 mg, 0.6 mmol) in dry toluene (12 mL) was added PPh3 (20 mg, 0.07 mmol), Pd(OAc)2 (47.5 %, 17 mg, 0.03 mmol), NEt3 (0.1 mL, 1 mmol) and tert-Butyl acrylate (0.2 mL, 1.3 mmol) and the mixture was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, water, extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 20: 1, 10: 1, 5: 1) provided the title compound as a transparent oil (72 mg, 0.2 mmol, 16 %).
1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, C(CH3)3), 1.60 (3H, s, CH3), 1.70 (3H, s, CH3), 3.32 (2H, d, J = 7.0 Hz, CH2), 5.25 (1H, t, J = 7.5 Hz, CH), 6.35 (1H, d, J = 15.9 Hz, CH), 7.18 (1H, d, J = 8.7 Hz, ArCH), 7.43 (2H, s, ArCH), 7.52 - 7.69 (4H, m, ArCH and CH), 8.22 (2H, d, J = 7.7 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.6, 28.2, 28.9, 80.5, 120.1, 121.1, 122.9, 126.6, 128.6, 129.2, 129.8, 130.2, 133.7, 134.3, 138.9, 142.8, 150.4, 164.8, 166.2.
4-[(lE)-3-(tert-butoxy)-3-oxoprop-l-en-l-yl]-2-(3-methylbut-2-en-l-yl)phenyl pyridine-4-carboxylate (6g): To a solution of 5g (650 mg, 1.6 mmol) in dry toluene (10 mL) was added PPh3 (42 mg, 0.2 mmol), Pd(OAc)2 (47.5 %, 40 mg, 0.1 mmol) and NEt3 (0.4 mL, 2.7 mmol) the mixture was sitirred for 10 min and tert-butyl acrylate (0.4 mL, 2.8 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 4: 1, 2: 1, 0: 1) provided the title compound as a yellow oil (350 mg, 0.88 mmol, 54 %).
Rf: 0.31 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.51 (9H, s, C(CH3)3), 1.51 (3H, s,
CH3), 1.64 (3H, s, CH3), 3.26 (2H, d, J = 6.6 Hz, CH2), 5.17 (1H, t, J = 6.2 Hz, CH), 6.32 (1H, d, J = 15.9 Hz, CH), 7.14 (1H, d, J = 8.5 Hz, Ar-H), 7.40 (2H, s Ar-H), 7.55 (1H, d, J = 15.9 Hz, CH), 7.96 (2H, d, J = 4.2 Hz, Ar-H), 8.84 (2H, d, J = 4.2 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.6, 28.1, 29.0, 60.3, 80.5, 120.5, 120.8, 122.5, 123.1, 126.7, 130.0, 133.1, 133.9, 134.0, 136.4, 142.5, 149.8, 150.8, 163.3, 166.1.
(E)-4-(3-(tert-butoxy)-3-oxoprop-l-en-l-yl)-2-(3-methylbut-2-en-l-yl)phenyl hexanoate (6h): Following the procedure as described for 6a the title compound was obtained as a white soild (150 mg, 0.4 mmol, 40%).
1H NMR (400 MHz; CDC13): δ 0.89 - 0.96 (3H, m, CH3), 1.28 - 1.41 (6H, m, CH2), 1.55 (9H, s, (CH3)3), 1.71 (3H, s, CH3), 1.77 (3H, s, CH3), 2.58 (2H, t, J = 7.2 Hz, CH2), 3.24 (2H, d, J = 6.8 Hz, CH2), 5.23 (1H, t, J = 7.2 Hz, CH), 6.31 (1H, d, J = 15.9 Hz, CH), 7.04 (1H, d, J = 8.9 Hz, ArCH), 7.37 - 7.40 (2H, m, ArCH), 7.56 (1H, d, J = 16.0 Hz, CH). 13C NMR (100 MHz; CDC13): δ 13.9, 17.8, 22.3, 24.6, 25.7, 28.2, 28.6, 31.3, 34.2, 80.4, 120.0, 121.0, 122.7, 126.4, 129.7, 132.5, 133.7, 134.1, 142.8, 150.2, 166.2, 171.9
tert-butyl (E)-3-(4-((3-(4-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (6i): To a solution of 5i (250 mg, 0.6 mmol) in dry toluene (6 mL) was added PPh3 (52 mg, 0.2 mmol), Pd(OAc)2 (47.5 %, 48 mg, 0.1 mmol) and NEt3 (0.4 mL, 3.0 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.2 mL, 1.4 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 4: 1, 2: 1, 0: 1) provided the title compound as a yellow oil (160 mg, 0.36 mmol, 60%).
Rf: 0.85 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, CH3), 1.68 (3H, s, CH3), 1.76 (3H, s, CH3), 2.91 (2H, t, J = 7.3 Hz, CH2), 3.06 (2H, t, J = 7.4 Hz, CH2), 3.13 (2H, d, J = 7.1 Hz, CH2), 5.18 (1H, t, J = 7.2 Hz, CH), 6.33 (1H, d, J = 15.6 Hz, CH), 6.95 - 7.04 (3H, m, ArCH), 7.23 - 7.28 (2H, m, ArCH), 7.35 - 7.37 (2H, m, ArCH), 7.57 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.2, 28.5, 30.0, 35.8, 80.5, 115.2, 115.4, 120.1, 120.9, 122.6, 126.5, 129.7, 129.8, 129.9, 132.6, 133.7, 134.0, 142.8, 150.0, 166.2, 170.9
tert-butyl (E)-3-(4-((3-(3-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (6j): To a solution of 5j (430 mg, 1.3 mmol) in dry toluene (6 mL) was added PPh3 (78 mg, 0.3 mmol), Pd(OAc)2 (47.5 %, 70 mg, 0.15 mmol) and NEt3 (0.4 mL, 3.0 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.4 mL, 2.8 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (260 mg, 0.6 mmol, 45%).
Rf: 0.51 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, CH3), 1.69 (3H, s, CH3), 1.76 (3H, s, CH3), 2.93 (2H, t, J = 7.5 Hz, CH2), 3.09 (2H, t, J = 7.5 Hz, CH2), 3.15 (2H, d, J = 7.1 Hz, CH2), 5.19 (1H, t, J = 7.2 Hz, CH), 6.32 (1H, d, J = 15.9 Hz, CH), 6.93 - 7.01 (3H, m, ArCH), 7.07 (1H, d, J = 7.6 Hz, ArCH), 7.27 - 7.32 (1H, m, ArCH), 7.35 - 7.38 (2H, m, ArCH), 7.56 (1H, d, J = 15.9 Hz,
CH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.2, 28.5, 35.4, 80.5, 113.3, 113.5, 115.2, 115.4, 120.1, 120.9, 122.6, 124.0, 126.5, 129.7, 132.6, 133.7, 134.0, 142.5 142.8, 150.0, 161.7, 166.2, 170.8.
tert-butyl(E)-3-(4-((3-(2-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (6k): To a solution of 5k (430 mg, 1 mmol) in dry toluene (6 mL) was added PPh3 (78 mg, 0.3 mmol), Pd(OAc)2 (47.5 %, 70 mg, 0.15 mmol) and NEt3 (0.4 mL, 3.0 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.4 mL, 2.8 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (250 mg, 0.6 mmol, 57%).
Rf: 0.73 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, CH3), 1.69 (3H, s, CH3), 1.77 (3H, s, CH3), 2.94 (2H, t, J = 7.5 Hz, CH2), 3.13 (2H, t, J = 7.5 Hz, CH2), 3.17 (2H, d, J = 7.1 Hz, CH2), 5.19 (1H, t, J = 7.2 Hz, CH), 6.32 (1H, d, J = 15.9 Hz, CH), 6.99 (1H, d, J = 8.8 Hz, ArCH), 7.03 - 7.12 (2H, m, ArCH), 7.19 - 7.32 (2H, m, ArCH), 7.35 - 7.37 (2H, m, ArCH), 7.56 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 24.6, 25.7, 28.2, 28.6, 34.2, 80.5, 115.3, 120.0, 121.0, 122.6, 124.2, 126.5, 128.3, 129.7, 130.7, 132.6, 133.7, 134.0, 142.8, 150.0, 162.4, 166.2, 170.8.
tert-butyl(E)-3-(4-((3-(4-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (61): To a solution of 51 (320 mg, 0.7 mmol) in dry toluene (8 mL) was added PPh3 (53 mg, 0.2 mmol), Pd(OAc)2 (23 mg, 0.1 mmol) and NEt3 (0.2 mL, 1.5 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.2 mL, 1.5 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (200 mg, 0.4 mmol, 47%).
Rf: 0.69 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, CH3), 1.68 (3H, s, CH3), 1.76 (3H, s, CH3), 2.89 (2H, t, J = 7.8 Hz, CH2), 3.04 (2H, t, J = 7.6 Hz, CH2), 3.14 (2H, d, J = 7.2 Hz, CH2), 3.80 (3H, s, CH3), 5.19 (1H, t, J = 7.2 Hz, CH), 6.31 (1H, d, J = 15.9 Hz, CH), 6.88 (2H, d, J = 8.6
Hz, ArCH), 6.97 (1H, d, J = 8.9 Hz, ArCH), 7.21 (2H, d, J = 8.6 Hz, ArCH), 7.35 - 7.37 (2H, m, ArCH), 7.55 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.2, 28.4, 30.0, 36.1, 55.2, 80.5, 114.0, 120.0, 121.0, 122.6, 126.4, 129.3, 129.6, 132.0, 132.6, 133.7, 134.0, 142.8, 150.1, 158.2, 166.2, 171.1.
tert-butyl(E)-3-(4-((3-(3-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acty (6m): To a solution of 5m (280 mg, 0.6 mmol) in dry toluene (8 mL) was added PPh3 (39 mg, 0.15 mmol), Pd(OAc)2 (47.5%, 35 mg, 0.08 mmol) and NEt3 (0.2 mL, 1.5 mmol) the mixture was sitirred for 10 min and tert-butyl acrylate (0.2 mL, 1.5 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2:1) provided the title compound as a yellow oil (220 mg, 0.4 mmol, 77%).
Rf: 0.55 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, CH3), 1.70 (3H, s, CH3), 1.77 (3H, s, CH3), 2.93 (2H, t, J = 7.8 Hz, CH2), 3.08 (2H, t, J = 7.6 Hz, CH2), 3.17 (2H, d, J = 7.2 Hz, CH2), 3.82 (3H, s, CH3), 5.20 (1H, t, J = 7.2 Hz, CH), 6.33 (1H, d, J = 15.9 Hz, CH), 6.81 (1H, dd, Jl = 2.4 Hz, J2 = 8.6 Hz, ArCH), 6.84 (1H, t, J = 1.8 Hz, ArCH), 6.88 (1H, d, J = 7.6 Hz, ArCH), 6.99 (1H, d, J = 8.8 Hz, ArCH), 7.26 (1H, t, J = 7.8 Hz, ArCH), 7.36 - 7.38 (2H, m, ArCH), 7.57 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 28.2, 30.9, 35.7, 55.1, 80.5, 111.8, 114.2, 120.0, 120.6, 121.0, 122.7, 126.5, 129.6, 129.7, 133.7, 134.1, 141.5, 142.8, 150.1, 150.8, 166.2, 171.0.
tert-butyl(E)-3-(4-((3-(2-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylate (6n): To a solution of 5n (320 mg, 0.7 mmol) in dry toluene (8 mL) was added PPh3 (78 mg, 0.3 mmol), Pd(OAc)2 (34 mg, 0.15 mmol) and NEt3 (0.4 mL, 3 mmol) the mixture was sitirred for 10 min and tert- butyl acrylate (0.4 mL, 2 mmol) added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (150 mg, 0.3 mmol, 47%).
Rf: 0.70 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.57 (9H, s, CH3), 1.71 (3H, s, CH3), 1.78 (3H, s, CH3), 2.91 (2H, t, J = 7.8 Hz, CH2), 3.10 (2H, t, J = 7.6 Hz, CH2), 3.19 (2H, d, J = 7.2 Hz, CH2), 3.88 (3H, s, CH3), 5.22 (1H, t, J = 7.2 Hz, CH), 6.33 (1H, d, J = 15.9 Hz, CH), 6.92 (2H, q, J = 7.4 Hz, ArCH), 7.00 (1H, d, J = 8.8 Hz, ArCH), 7.23 - 7.26 (2H, m, ArCH), 7.36 - 7.38 (2H, m, ArCH), 7.57 (1H, d, J = 15.9 Hz, CH).
(E)-2-(3-methylbut-2-en-l-yl)-4-(2-(4,4^ 3- phenylpropanoate (7): To a solution of 5a (350 mg, 0.8 mmol) in dry toluene (8 mL) was added PPh3 (27 mg, 0.1 mmol), Pd(OAc)2 (12 mg, 0.05 mmol) and NEt3 (0.3 mL, 2 mmol) the mixture was sitirred for 10 min and 4,4,5, 5-tetramethyl-2-vinyl-l,3,2-dioxaborolane (0.2 mL, 1 mmol) was added and the reaction refluxed overnight. The reaction was allowed to cool and was washed with a saturated aqueous NH4C1, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1) provided the title compound as a yellow oil (70 mg, 0.15 mmol, 20%).
Rf: 0.74 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.34 (12H, s, CH3), 1.68 (3H, s, CH3), 1.76 (3H, s, CH3), 2.92 (2H, t, J = 7.1 Hz, CH2), 3.11 (2H, t, J = 7.8 Hz, CH2), 3.14 (2H, d, J = 7.8 Hz, CH2), 5.20 (1H, t, J = 7.8 Hz, CH), 6.12 (1H, d, J = 18.4 Hz, CH), 6.93 (1H, d, J = 8.6 Hz, ArCH), 7.23 - 7.39 (8H, m, ArCH and CH). 13C NMR (100 MHz; CDC13): δ 17.8, 24.8, 25.7, 28.4, 30.9, 35.8, 83.3, 121.1, 122.2, 125.5, 128.3, 128.3, 128.5, 128.6, 128.7, 133.5, 133.5, 135.4, 140.0, 148.7, 149.2, 171.1.
(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-[(3-phenylpropanoyl)oxy]phenyl]prop-2-enoic acid (1): To a solution of 6a in toluene (10 ml) was added chromatography grade silica gel (3.3 g) and the mixture was reflued with vigorous agitation overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over celite pad using 10% methanol in DCM as the solvent. Purified with silica gel flash chromatography using eluent system as Hexane:EtOAc = 2: 1, 1: 1, 0: 1 and solvent evaporated in vacuo to provide the title compound (139.5 mg, 0.38 mmol, 58%).
Rf: 0.1 (Hexane:EtOAc = 4: 1). 1H NMR (400 MHz, CDC13): δ 1.69 (3H, s, CH3), 1.77 (3H, s, CH3), 2.94 (2H, t, J = 7.7 Hz, CH2), 3.11 (2H, t, J = 7.6 Hz, CH2), 3.17 (2H, d, J=7.4Hz, H-l"), 5.20 (1H, t, J = 7.3 Hz, CH), 6.40 (1H, d, J = 16 Hz, CH), 7.00 (1H, d, J = 8.3 Hz, ArCH) 7.24-7.42 (7H, m, ArCH), 7.75 (1H, d, J = 16 Hz, CH). 13C NMR (100 MHz, CDC13): δ 17.8, 25.7, 28.4, 30.8, 35.8, 116.7, 120.8, 122.8, 126.5, 126.8, 128.6, 130.1, 131.9, 133.9, 134.3, 139.9, 146.3, 150.6, 170.6, 171.0 M/Z ESI: 363.1 [M-, 100%], 364.2(25%)
(2E)-3-[4-(acetyloxy)-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoic acid (8b): To a solution of 6b (100 mg, 0.3 mmol) in dry toluene (5 mL) was added silica gel (3 mL) and the mixture was refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad and the solution concentrated. Purification by column chromatography (Hexane:EtOAc = 4: 1, 2: 1, 1: 1, 0: 1) provided the title compound as a white solid (86 mg, 0.3 mmol, 99 %).
Rf: 0.11 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.73 (3H, s, CH3), 1.78 (3H, s, CH3), 2.34 (3H, s, C02CH3), 3.27 (2H, d, J = 7.7 Hz, CH2), 5.24 (1H, t, J = 7.1 Hz, CH), 6.41 (1H, d, J = 15.9 Hz, CH), 7.09 (1H, d, J = 8.4 Hz, Ar-H), 7.43 (2H, s, Ar-H), 7.75 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.8, 20.8, 25.7, 28.6, 117.1 120.8, 122.9, 126.9, 130.2, 132.0, 134.0, 146.3, 150.7, 169.0, 172.1. m/z (ESI): 273.0 [M - H]- (100%).
(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-[(4-phenylbutanoyl)oxy]phenyl]prop-2-enoic acid (8c): To a solution of 6c (230 mg, 0.5 mmol) in dry toluene (5 mL) was added silica gel (3 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad using 10% methanol in DCM as the solvent. The solvent was evaporated in vacuo and purified by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1 : 1) to provide the title compound as a white solid (12.2 mg, 0.03 mmol, 6 %).
Rf: 0.10 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.68 (3H, s, CH3), 1.76 (3H, s, CH3), 2.12 (2H, q, J = 7.5 Hz, CH2), 2.62 (2H, t, J = 7.5 Hz, CH2), 2.78 (2H, t, J = 7.5 Hz, CH2), 3.25 (2H, d, J =7.1 Hz, CH2), 5.23 (1H, t, J = 7.2 Hz, CH), 6.40 (1H, d, J = 15.0, CH), 7.06 (1H, d, J = 8.5 Hz, Ar-H), 7.23 - 7.26 (3H, m, Ar-H), 7.31 - 7.35 (2H, m, Ar-H), 7.42 - 7.44 (2H, m, Ar-H), 7.76 (1H, d, J = 15.95 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.8, 25.7, 26.2, 26.4, 28.6, 33.4, 35.1, 117.0, 120.8, 122.9,
126.1, 126.9, 128.5, 130.2, 131.9, 134.1, 134.3, 141.0, 146.3, 150.7, 171.6, 171.9. m/z (ESI): 377.1 [M - H]- (100%).
(2E)-3-{4-[(2-cyclohexylacetyl)oxy]--(3-methylbut-2-en-l-yl)phenyl}prop-2-enoic acid (8d): To a solution of 6d (370 mg, 0.85 mmol) in dry toluene (10 mL) was added silica gel (5 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad using 10% methanol in DCM as the solvent which was concentrated in vacuo. Purification by column chromatography (Hexane:EtOAc = 4: 1, 1: 1) provided the title compound as a white solid (130 mg, 0.3 mmol, 40 %).
Rf: 0.14 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.05 - 1.39 (7H, m, cyc-H), 1.72 (3H, s, CH3), 1.79 (3H, s, CH3), 1.84-1.96 (3H, m, cyc-H), 2.47 (2H, d, J = 7 Hz, CH), 3.24 (2H, d, J = 7.1 Hz, CH2), 5.23 (1H, t, J = 7.1 Hz, CH), 6.39 (1H, d J = 15.9, CH), 7.04 (1H, d, J = 8.8 Hz, Ar-H), 7.39-7.36 (2H, m, Ar-H), 7.75 (1H, d, J = 15.9, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 26.0, 28.5, 33.0, 34.9, 41.9, 117.0, 120.8, 122.9, 126.8, 128.3, 128.5, 128.9, 130.1, 131.8, 134.0, 134.3, 146.4, 150.8, 171.1, 172.3. m/z (ESI): 355.1 [M - H]- (100%)
(2E)-3- [3-(3-methylbut-2-en- 1 -yl)-4- { [2-(naphthalen- 1 -yl)acetyl]oxy }phenyl]prop-2-enoic acid (8e) : To a solution of 6e (66.1 mg, 0.15 mmol) in toluene (5 mL) was added silica gel (3 mL) and the mixture was refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite pad® using 10% methanol in DCM as the solvent. The solvent was evaporated in vacuo to provide the title compound as a white solid (34 mg, 0.1 mmol, 60 %).
Rf: 0.20 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.57 (3H, s, CH3), 1.70 (3H, s, CH3), 3.00 (2H, d, J = 7.3 Hz, CH2), 4.36 (2H, s, CH2), 5.03 (1H, t, J = 7.2 Hz, CH), 6.37 (1H, d, J = 15.9 Hz, CH), 7.03 (1H, d, J = 8.2 Hz, Ar-H), 7.34 - 7.38 (2H, m, Ar-H), 7.48 - 7.62 (4H, m, Ar-H) 7.72 (1H, d, J = 15.9 Hz, CH), 7.87 (1H, d, J = 8.1 Hz, Ar-H), 7.89 (1H, dd, Jl = 8.7 Hz, J2 = 21.7 Hz, Ar-H), 8.14 (1H, d, J = 8.5 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.7, 25.6, 28.2, 29.7, 39.3, 117.0, 120.5, 122.7, 123.6, 125.5, 125.9, 126.6, 126.8, 128.3, 128.4, 128.8, 129.7, 130.0, 131.9, 132.0, 133.8, 133.9, 134.3, 146.3, 150.6, 169.6. m/z: (ESI): 399.0 [M - H]- (75 %).
(E)-3-(4-(benzoyloxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8f): To a solution of 6f (72 mg, 0.2 mmol) in toluene (6 mL) was added silica gel (2 mL) and the mixture was refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite pad® using 10% methanol in DCM as the solvent. The solvent was evaporated in vacuo to provide the title compound as a white solid (32 mg, 0.09 mmol, 50 %).
1H NMR (400 MHz; MeOD): δ 1.61 (3H, s, CH3), 1.72 (3H, s, CH3), 3.35 (2H, d, J = 7.0 Hz, CH2), 5.26 (1H, t, J = 7.5 Hz, CH), 6.45 (1H, d, J = 16.0 Hz, CH), 7.23 (1H, d, J = 8.2 Hz, ArCH), 7.49 - 7.57 (4H, m, ArCH), 7.68 (1H, t, J = 7.0 Hz, ArCH), 7.81 (1H, d, J = 15.9 Hz, CH), 8.23 (2H, d, J = 7.3 Hz, ArCH). 13C NMR (100 MHz; MeOD): δ 17.8, 25.6, 28.8, 117.0, 120.8, 123.0, 127.0, 128.6, 130.2, 130.3, 132.0, 133.7, 134.0, 134.6, 146.3, 151.0, 164.7, 171.1, 171.5.
(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-(pyridine-4-carbonyloxy)phenyl]prop-2-enoic acid (8g): To a solution of 6g (350 mg, 0.9 mmol) in dry toluene (25 mL) was added silica gel (20 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a yellow solid (150 mg, 0.4 mmol, 50 %)
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.55 (3H, s, CH3), 1.62 (3H, s, CH3), 3.36 (2H, d, J = 7.0 Hz, CH2), 5.19 (1H, t, J = 7.0 Hz, CH), 6.50 (1H, d, J = 16.0 Hz, CH), 7.26 (1H, d, J = 8.0 Hz, Ar-H), 7.57 (1H, s Ar-H), 7.59 (1H, d, J = 2.1 Hz, Ar-H), 7.69 (1H, d, J = 15.9 Hz, CH), 8.12 (2H, d, J = 6.1 Hz, Ar-H), 8.86 (2H, d, J = 5.1 Hz, Ar-H). 13C NMR (100 MHz; MeOD): δ 16.5, 24.3, 28.9, 60.1, 118.4, 121.2, 122.8, 123.3, 126.6, 129.9, 133.0, 134.2, 143.7, 150.1, 150.3, 163.1, 168.8. m/z (ESI): 338.1 [M + H]+ (100 %)
(E)-3-(4-(hexanoyloxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8h): Following the procedure as described for 1 the title compound was obtained as a white soild (50 mg, 0.15 mmol, 37%).
1H NMR (400 MHz; MeOD): δ 1.36 - 1.43 (7H, m, CH3 and CH2), 1.50 - 1.62 (2H, m, CH2), 1.69 (3H, s, CH3), 1.76 (3H, s, CH3), 2.57 (2H, t, J = 7.4 Hz, CH2), 3.18 (2H, d, J = 7.0 Hz, CH2), 5.19 (1H, t, J = 7.2 Hz, CH), 6.45 (1H, d, J = 15.7 Hz, CH), 7.01 (1H, d, J = 8.3 Hz, ArCH), 7.52 - 7.54 (2H, m, ArCH), 7.46 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; MeOD): δ 12.8, 16.5, 22.0, 24.2, 24.4, 28.3, 31.0, 33.5, 121.2, 122.6, 126.1, 129.3, 132.8, 132.9, 134.1, 142.3, 150.2, 172.2
(E)-3-(4-((3-(4-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8i): To a solution of 8i (160 mg, 0.36 mmol) in dry toluene (8 mL) was added silica gel (3 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a white solid (72 mg, 0.2 mmol, 52%).
Rf: 0.16 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz, MeOD): δ 1.65 (3H, s, CH3), 1.72 (3H, s, CH3), 2.92 (2H, t, J = 7.2 Hz, CH2), 3.08 (2H, t, J = 7.0 Hz, CH2), 3.17 (2H, d, J=7.4Hz, CH2), 5.13 (1H, t, J = 7.0 Hz, CH), 6.40 (1H, d, J = 15.9 Hz, CH), 7.02 (2H, t, J = 8.6 Hz, ArCH), 7.29 (2H, t, J = 8.1 Hz, ArCH), 7.40-7.44 (2H, m, ArCH), 7.62 (1H, d, J = 15.9 Hz, CH) 13C NMR (100 MHz, MeOD): δ 16.5, 24.4, 28.1, 29.5, 35.1, 114.6, 117.9, 121.0, 122.6, 129.7, 132.3, 133.0, 134.2, 136.1, 144.0, 150.4, 160.4, 162.8, 168.8, 171.1,
(E)-3-(4-((3-(3-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8j): To a solution of 8j (260 mg, 0.6 mmol) in dry toluene (8 mL) was added silica gel (5 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a white solid (103 mg, 0.3 mmol, 45%).
Rf: 0.18 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz, MeOD): δ 1.67 (3H, s, CH3), 1.73 (3H, s, CH3), 2.98 (2H, t, J = 7.0 Hz, CH2), 3.07 (2H, t, J = 7.7 Hz, CH2), 3.12 (2H, d, J=7.1 Hz, CH2), 5.15 (1H, t, J = 7.2 Hz, CH), 6.42 (1H, d, J = 15.9 Hz, CH), 6.93 - 6.99 (2H, m, ArCH), 7.06 (1H, d, J = 8.4 Hz, ArCH), 7.12 (1H, d, J = 7.6 Hz, ArCH), 7.32 (1H, q, Jl = 6.1 Hz, J2 = 7.9 Hz, ArCH), 7.43 (1H, s, ArCH), 7.46 (1H, dd, Jl = 2.1 Hz, J2 = 8.3 Hz, ArCH), 7.63 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz, MeOD): δ 16.5, 24.4, 28.1, 29.9, 30.0, 34.7, 112.8, 114.9, 117.9, 121.0, 122.6, 123.9, 126.3, 129.5, 129.8, 132.3, 133.0, 134.2, 143.0, 144.0, 150.4, 161.7, 168.8, 171.1.
(E)-3-(4-((3-(2-fluorophenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8k): To a solution of 8k (250 mg, 0.6 mmol) in dry toluene (6 mL) was added silica gel (3 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a white solid (78 mg, 0.2 mmol, 35%).
Rf: 0.16 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz, MeOD): δ 1.67 (3H, s, CH3), 1.72 (3H, s, CH3), 2.93 (2H, t, J = 7.3 Hz, CH2), 3.13 (2H, t, J = 7.1 Hz, CH2), 3.12 (2H, d, J = 7.1 Hz, CH2), 5.14 (1H, t, J = 7.1 Hz, CH), 6.41 (1H, d, J = 15.9 Hz, CH), 6.98 (1H, d, J = 8.2 Hz, ArCH), 7.05 - 7.13 (2H, m, ArCH), 7.23 - 7.28 (1H, m, ArCH), 7.33 (1H, t, J = 7.6 Hz, ArCH), 7.41 - 7.45 (2H, m, ArCH), 7.62 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz, MeOD): δ 16.5, 23.9, 24.4, 28.2, 33.6, 114.7, 114.9, 117.9, 121.1, 122.7, 124.0, 126.3, 128.1, 129.5, 130.5, 132.2, 133.0, 134.2, 144.0, 150.4, 168.8, 171.0.
(E)-3-(4-((3-(4-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (81): To a solution of 81 (200 mg, 0.4 mmol) in dry toluene (12 mL) was added silica gel (6 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a white solid (73 mg, 0.2 mmol, 42%).
Rf: 0.07 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz, MeOD): δ 1.66 (3H, s, CH3), 1.74 (3H, s, CH3), 2.90 (2H, t, J = 7.1 Hz, CH2), 2.98 (2H, t, J = 7.0 Hz, CH2), 3.07 (2H, d, J = 7.2 Hz, CH2), 3.77 (3H, s,
CH3), 5.14 (1H, t, J = 7.2 Hz, CH), 6.41 (1H, d, J = 15.9 Hz, CH), 6.86 (2H, d, J = 8.7 Hz, ArCH), 6.96 (1H, d, J = 8.2 Hz, ArCH), 7.20 (2H, d, J = 8.7 Hz, ArCH), 7.41 (1H, s, ArCH), 7.45 (1H, dd, Jl = 2.1 Hz, J2 = 8.3 Hz, ArCH), 7.62 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz, MeOD): δ 16.5, 24.4, 28.0, 29.6, 35.4, 54.2, 133.5, 117.9, 121.0, 122.7, 126.2, 129.0, 129.4, 132.0, 132.3, 133.0, 134.2, 144.0, 150.5, 158.3, 168.8, 171.4.
(E)-3-(4-((3-(3-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8m): To a solution of 8m (220 mg, 0.5 mmol) in dry toluene (8 mL) was added silica gel (5 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a white solid (60 mg, 0.15 mmol, 32%).
Rf: 0.09 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz, MeOD): δ 1.65 (3H, s, CH3), 1.72 (3H, s, CH3), 2.91 (2H, t, J = 7.1 Hz, CH2), 3.00 (2H, t, J = 7.2 Hz, CH2), 3.09 (2H, d, J = 7.1 Hz, CH2), 3.76 (3H, s, CH3), 5.14 (1H, t, J = 7.2 Hz, CH), 6.40 (1H, d, J = 15.9 Hz, CH), 6.76 - 6.78 (1H, m, ArCH), 6.84 (2H, s, ArCH), 6.95 (1H, d, J = 8.2 Hz, ArCH), 7.21 (1H, t, J = 8.2 Hz, ArCH), 7.39 - 7.43 (2H, m, ArCH), 7.62 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz, MeOD): δ 16.6, 24.5, 28.1, 30.4, 35.1, 54.1, 111.4, 113.8, 117.9, 120.3, 121.1, 122.7, 126.3, 129.1, 129.5, 132.3, 133.0, 134.2, 141.7, 144.0, 150.5, 159.9, 168.8, 171.3.
(E)-3-(4-((3-(2-methoxyphenyl)propanoyl)oxy)-3-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (8n): To a solution of 8n (150 mg, 0.3 mmol) in dry toluene (8 mL) was added silica gel (5 mL) and the mixture refluxed overnight. Upon cooling the reaction mixture was diluted with 10% methanol in DCM and filtered over a celite® pad. Purified by column chromatography (DCM:Mehanol = 10: 1, 5: 1) and solvent was evaporated in vacuo to provide the title compound as a white solid (73 mg, 0.2 mmol, 56%).
Rf: 0.1 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz, MeOD): δ 1.71 (3H, s, CH3), 1.78 (3H, s, CH3), 2.93 (2H, t, J = 7.1 Hz, CH2), 3.10 (2H, t, J = 7.5 Hz, CH2), 3.20 (2H, d, J = 7.2 Hz, CH2), 3.88 (3H, s, CH3), 5.23 (1H, t, J = 7.2 Hz, CH), 6.41 (1H, d, J = 15.9 Hz, CH), 6.92 (2H, q, J = 6.4 Hz ArCH), 7.03
(1H, d, J = 8.9 Hz, ArCH), 7.23 - 7.28 (2H, m, ArCH), 7.42 (2H, s, ArCH), 7.678 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz, MeOD): δ 17.8, 25.7, 26.2, 28.5, 34.1, 55.2, 110.2, 117.0, 120.5, 120.9, 122.9, 126.9, 127.9, 128.2, 130.1, 130.2, 131.8, 133.9, 134.3, 146.4, 150.8, 157.5, 171.5, 172.3.
(E)-(3-(3-methylbut-2-en-l-yl)-4-((3-phenylpropanoyl)oxy)st ryl)boronic acid (7a): The solution of 7 (20 mg, 0.05 mmol) in 1: 1 MeOH/H20 was stirred at 80oC overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a white solid (5 mg, 0.01 mmol, 20%).
Rf: 0.1 (Hexane:EtOAc, 4: 1) 1H NMR (400 MHz; CDC13): δ 1.68 (3H, s, CH3), 1.73 (3H, s, CH3), 2.94 (2H, t, J = 7.2 Hz, CH2), 3.06 (2H, t, J = 7.5 Hz, CH2), 3.11 (2H, d, J = 7.4 Hz, CH2), 5.16 (1H, t, J = 7.2 Hz, CH), 6.31 (1H, d, J = 18.0 Hz, CH), 6.90 (1H, d, J = 8.2 Hz, ArCH), 7.20 - 7.34 (7H, m, ArCH and CH), 7.39 (1H, dd, Jl = 2.1 Hz, J2 = 8.3 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 16.5, 24.4, 28.2, 30.4, 35.2, 121.4, 122.2, 124.9, 126.0, 128.0, 128.1, 128.3, 132.6, 133.6, 135.8, 140.2, 147.1, 149.2,
171.5.
2-bromo-4-iodoaniline (10): To a solution of 4-iodo aniline (1.1 g, 5.0 mmol), in 6 mL of HO Ac, was added a solution of Br2 (250 μΐ., 4.8 mmol) and the mixture stirred overnight at room temperature. The reaction mixture was washed with brine, saturated aqueous NaHC03 and extracted with Et20, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a brown oil (570 mg, 1.9 mmol, 44 %)
Rf: 0.46 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 6.54 (1H, d, J = 8.2 Hz, ArCH), 7.37 (1H, d, J = 8.4 Hz, ArCH), 7.70 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 110.0, 117.3, 131.1, 134.4, 136.9, 139.9, 143.8.
tert-butyl (2E)-3-(4-amino-3-bromophenyl)prop-2-enoate (11): To a solution of (10) (570 mg, 1.9 mmol) in dry toluene (8 mL), was added PPh3 (65.5 mg, 0.2 mmol) and Pd(OAc)2 (47.5 %, 60 mg, 0.1 mmol). tert-Butyl Acrylate (370 μΐ., 2.5 mmol) and NEt3 (420 μΐ., 3.0 mmol) were added and the flask was stirred at reflux overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1) provided the title compound as a brown oil (238.5 mg, 0.8 mmol, 64 %)
Rf : 0.23 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13,): δ 1.51 (9H, s, C(CH3)3), 6.15 (1H, d, J = 15.8 Hz, CH), 6.69 (1H, d, J = 8.1 Hz, ArCH), 7.23 (1H, d, J = 8.2 Hz, ArCH), 7.40 (1H, d, J = 15.8 Hz, CH), 7.55 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.2, 80.2, 109.0, 115.3, 117.0, 126.0, 128.4, 132.4, 142.3, 145.7, 166.6.
tert-butyl (2E)-3-[3-bromo-4-(3-phenylpropanamido)phenyl]prop-2-enoate (13a): To a solution of (11) (290 mg, 1.0 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol). A solution of PhCH2CH2COCl (255 mg, 1.5 mmol) in DCM (2 mL) was added to the mixture followed by addition NEt3 (420 μΕ, 3.0 mmol). The solution was heated to 700C and stirred overnight. The reaction was allowed to cool and washed with a saturated aqueous NaHC03, water and extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a brown oil (40 mg, 0.1 mmol, 10 %).
Rf: 0.51 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 2.78 (2H, t, J = 7.6 Hz, CH2), 3.09 (2H, t, J = 7.6 Hz, CH2), 6.31 (1H, d, J = 15.9 Hz, CH), 7.24 - 7.34 (5H, m, ArCH), 7.44 - 7.48 (2H, m, CH and ArCH), 7.67 (1H, d, J = 1.8 Hz, ArCH), 8.41 (1H, d, J = 8.5 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 31.3, 39.6, 80.6, 120.4, 126.5, 128.0, 128.3, 128.7, 131.4, 136.7, 140.1, 141.2, 165.9, 170.3.
tert-butyl (2E)-3-(3-bromo-4-acetamidophenyl)prop-2-enoate (13b): To a solution of 11 (181 mg, 0.6 mmol) in dry DCM (3 mL) was added DMAP (13 mg, 0.1 mmol) and a solution of CH3COC1 (80 mg, 1 mmol) in DCM (1 mL) followed by the addition of NEt3 (210 μΐ., 1.5 mmol). The solution was heated to 400C and stirred overnight. The reaction was allowed to cool and washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 4: 1, 2: 1, 1: 1) provided the title compound as a yellow oil (140 mg, 0.4 mmol, 68 %)
Rf: 0.20 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 2.26 (3H, s, CH3), 6.30 (1H, d, J = 15.9 Hz, CH), 7.44 (1H, s, NH), 7.47 (1H, d, J = 5.7 Hz, ArCH), 7.62 (2H, d, ArCH and CH), 8.39 (1H, d, J = 7.9 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 24.9, 28.1, 80.6, 113.1, 120.5, 121.4, 128.0, 131.4, 131.6, 136.8, 141.2, 165.9, 168.2.
(2E)-3-[3-bromo-4-(4-phenylbutanamido)phenyl]prop-2-enoic acid (13c): To a solution of 11 (150 mg, 0.5 mmol ) in dry DCM (3 mL) was added DMAP (13 mg, 0.1 mmol) and a solution of PhCH2CH2CH2COCl (185 mg, 1 mmol) in DCM (1 mL) followed by the addition of NEt3 (140 μΕ, 1 mmol). The reaction was heated to 400C and stirred overnight. The reaction was allowed to cool and washed with saturated aqueous NaHC03 and water, extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1, 1 : 1) provided the title compound as a yellow oil (20 mg, 0.05 mmol, 10 %).
Rf: 0.4 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 2.11 (2H, d, J = 7.4 Hz, CH2), 2.46 (2H, d, J = 7.1 Hz, CH2), 2.75 (2H, d, J = 7.4 Hz, CH2), 6.32 (1H, d, J = 15.8 Hz, CH), 7.23 - 7.34 (5H, m, ArCH), 7.46 - 7.49 (2H, m, ArCH and CH), 7.68 (1H, s, ArCH), 8.24 (1H, d, J = 8.5 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 26.7, 28.2, 34.9, 37.0, 80.7, 113.2, 120.4, 121.4, 125.3, 126.1, 128.0, 128.2, 128.3, 128.5, 128.5, 129.0, 131.4, 131.6, 136.8, 141.1, 141.2, 165.9, 170.9.
tert-butyl (2E)-3-[3-bromo-4-(2-cyclohexylacetamido)phenyl]prop-2-enoate (13d): To a solution of 11 (150 mg, 0.5 mmol) in dry DCM (3 mL) was added DMAP (13 mg, 0.1 mmol) and a solution of cyclohexylacetyl chloride (160 mg, 1 mmol) in DCM (1 mL), followed by the addition of NEt3 (140 μΐ^, 1 mmol). The reaction was heated to 400C and stirred overnight. The reaction was allowed to cool and washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a yellow oil (100 mg, 0.2 mmol, 46 %).
Rf: 0.54 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 0.9-1.37 (7H, m, CH2), 1.53 (9H, s, C(CH3)3), 1.67-1.90 (4H, m, CH2), 2.31 (2H, d, J = 6.8 Hz, CH2), 6.30 (1H, d, J = 15.8 Hz, CH), 7.44 - 7.47 (2H, m, ArCH and CH), 7.69 (1H, s, ArCH), 8.42 (1H, d, J = 8.2 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 26.0, 26.1, 26.2, 28.1, 33.1, 33.1, 35.5, 46.1, 80.6, 113.2, 120.3, 121.4, 128.0, 131.4, 131.5, 136.8, 141.2, 165.9, 170.7, 174.2.
tert-butyl (2E)-3-{3-bromo-4-[2-(naphthalen-l-yl)acetamido]phenyl}prop-2-enoate (13e): To a solution of 11 (183.1 mg, 0.6 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol) and a solution of naphthacetyl chloride (205 mg, 1 mmol) in DCM (1 mL), followed by addition of NEt3 (210 μΕ, 1.5 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a brown oil (210 mg, 0.4 mmol, 75 %).
Rf: 0.52 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 4.24 (2H, s, CH2), 6.24 (1H, d, J = 15.9 Hz, CH), 7.37 - 7.58 (7H, m, Ar-H and CH), 7.72 (1H, s, Ar-H), 7.90 - 7.92 (1H, m, Ar-H), 8.02 (1H, d, J = 7.7 Hz, Ar-H), 8.40 (1H, d, J = 8.4 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 28.2, 43.0, 80.6, 113.1, 120.3, 120.8, 123.6, 125.7, 126.4, 127.2, 127.8, 128.8, 128.9, 129.1, 130.0, 131.2, 131.6, 132.0, 134.1, 136.6, 141.2, 165.9, 169.1
tert-butyl (2E)-3-[3-(3-methylbut-2-en-l-yl)-4-(3-phenylpropanamido)phenyl]prop-2-enoate (14a): To a solution of 13a (40 mg, 0.1 mmol) in dry DMF (2 mL) was added CsC03 (65 mg, 0.2 mmol) and Pd(dppf)C12 (16 mg, 0.02 mmol). Prenyl boronic acid pinacol ester (44 μΐ., 0.2 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was filtered through a celite® pad with EtOAc. The solvent was evaporated in vaccuo, re-dissolved in DCM and the residual DMF removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (20 mg, 0.04 mmol, 44 %)
Rf: 0.30 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.74 (3H, s, CH3), 1.77 (3H, s, CH3), 2.63 (2H, t, J = 7.3 Hz, CH2), 3.06 (2H, t, J = 7.1 Hz, CH2), 3.20 (2H, d, J = 6.2 Hz, CH2), 5.14 (1H, t, J = 6.4 Hz, CH), 6.31 (1H, d, J = 16.1 Hz, CH), 7.23 - 7.33 (6H, m, Ar-H), 7.39 (1H, d, J = 8.3 Hz, Ar-H), 7.53 (1H, d, J = 16.0 Hz, CH), 8.04 (1H, d, J = 7.6 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 18.0, 25.7, 28.2, 31.3, 31.6, 39.7, 80.4, 119.2, 121.3, 122.4, 126.4, 126.8, 128.3, 128.6, 129.4, 134.8, 137.6, 140.5, 143.0, 166.4, 170.1.
tert-butyl (2E)-3-[4-acetamido-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoate (14b): To a solution of 13b (140 mg, 0.4 mmol) in dry DMF (2 mL) was added CsC03 (230 mg, 0.7 mmol) and Pd(dppf)C12 (40 mg, 0.05 mmol) and the flask was flushed with nitrogen again. Prenyl boronic acid pinacol ester (140 μΐ., 0.6 mmol) was added and the reaction was heated at 900C overnight. The reaction was allowed to cool and was extracted over a celite® pad with EtOAc. The solvent was evaporated in vaccuo and residual DMF was removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1, 1 :2, 1:5, 0: 1) to provide the title compound as a transparent oil (51 mg, 0.5 mmol, 38 %)
Rf: 0.09 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.81 (3H, s, CH3), 1.83 (3H, s, CH3), 2.10 (3H, s, CH3), 3.33 (2H, d, J = 6.3 Hz, CH2), 5.21 (1H, t, J = 5.2 Hz, CH), 6.30 (1H, d, J = 15.8 Hz, CH), 7.29 (1H, d, J = 8.2 Hz, Ar-H), 7.41 (1H, s, Ar-H), 7.52 (1H, d, J = 15.9
Hz, CH), 8.01 (1H, d, J = 7.8 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 24.8, 25.7, 28.2, 31.6, 80.4, 119.2, 121.4, 122.5, 126.8, 128.8, 129.4, 130.9, 134.7, 137.8, 143.0, 166.4, 168.0.
tert-butyl (2E)-3-[3-(3-methylbut-2-en-l-yl)-4-(4-phenylbutanamido)phenyl]prop-2-enoate (14c): To a solution of 13c (20 mg, 0.05 mmol) in dry DMF (3 mL) was added Cs2C03 (230 mg, 0.8 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (140 μΐ., 0.6 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a celite® pad with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4:1, 2: 1, 1: 1, 0: 1) provided the title compound as a transparent oil (17 mg, 0.04 mmol, 80 %).
1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.73 (3H, s, CH3), 1.74 (3H, s, CH3), 2.06 (2H, d, J = 7.4 Hz, CH2), 2.32 (2H, d, J = 7.1 Hz, CH2), 2.73 (2H, d, J = 7.4 Hz, CH2), 3.31 (2H, d, J = 6.5 Hz, CH2), 5.18 (1H, t, J = 7.2 Hz, CH), 6.31 (1H, d, J = 16.8 Hz, CH), 7.20 - 7.42 (7H, m, Ar-H), 7.53 (1H, d, J = 16.2 Hz, CH), 8.10 (1H, d, J = 8.2 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 17.9, 25.6, 26.9, 28.2, 31.7, 35.1, 36.9, 37.0, 80.3, 119.1, 121.4, 122.1, 126.1, 126.9, 128.4, 128.5, 129.5, 130.7, 134.9, 137.9, 141.2, 143.0, 166.5, 170.7.
tert-butyl (2E)-3-[4-(2-cyclohexylacetamido)-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoate (14d): To a solution of 13d (100 mg, 0.2 mmol) in dry DMF (3 mL) under was added CsC03 (230 mg, 0.8 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (140 μΐ., 0.6 mmol) was added and the flask heated at 900C overnight. The reaction was allowed to cool and was extracted over a celite® pad with EtOAc and concentrated in vacuo. Residual DMF was removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a transparent oil (20 mg, 0.05 mmol, 21 %).
Rf: 0.42 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 0.99 - 1.32 (7H, m, CH2), 1.54 (9H, s, C(CH3)3), 1.71 (4H, m, CH2), 1.82 (3H, s, CH3), 1.84 (3H, s, CH3), 2.18 (2H, d, J = 6.4 Hz, CH2), 3.34
(2H, d, J = 6.3 Hz, CH2), 5.22 (1H, t, J = 6.8 Hz, CH), 6.31 (1H, d, J = 15.8 Hz, CH), 7.31 (1H, d, J = 14.0 Hz, CH), 7.40 (1H, s, CH), 7.53 (1H, d, J = 16.5 Hz, CH), 8.14 (1H, d, J = 8.7 Hz, Ar-H). 13C NMR (100 MHz; CDC13): δ 18.1, 25.7, 26.0, 26.1, 28.2, 31.7, 33.2, 35.6, 46.2, 80.3, 119.0, 121.5, 126.9, 128.2, 129.0, 129.4, 130.3, 130.6, 134.8, 138.0, 143.1, 166.5, 170.5.
tert-butyl (2E)-3-[3-(3-methylbut-2-en -yl)-4-[2-(naphthalen -yl)acetamido]phenyl]prop-2-enoate (14e): To a solution of 13e (210 mg, 0.4 mmol) in dry DMF (2 mL) was added CsC03 (230 mg, 0.8 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (140 μΐ., 0.6 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and extracted over a celite® pad with EtOAc. The solvent was evaporated and residual DMF was removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1, 0: 1) to provide the title compound as a transparent oil (50 mg, 0.1 mmol, 24.5 %).
Rf: 0.23 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 1.58 (3H, s, CH3), 1.65 (3H, s, CH3), 3.08 (2H, d, J = 5.0 Hz, CH2), 4.09 - 4.17 (2H, m, CH2), 4.94 (1H, t, J = 7.1 Hz, CH), 6.28 (1H, d, J = 15.9 Hz, CH), 7.25 - 7.57 (7H, m, Ar-H and CH), 7.88 (2H, t, J = 7.9 Hz, Ar- H), 8.09 (1H, d, J = 8.4 Hz, Ar-H), 8.20 (1H, d, J = 7.6 Hz, Ar-H), 8.34 (lH,s, NH). 13C NMR (100 MHz; CDC13): δ 25.6, 28.2, 30.8, 59.3, 60.4, 73.2, 80.4, 119.2, 120.0, 121.9, 123.7, 125.2, 126.1, 126.4, 126.7, 126.9, 128.9, 129.3, 129.8, 131.0, 131.5, 134.2, 134.5, 135.6, 136.8, 143.0, 166.5, 170.7
(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-(3-phenylpropanamido)phenyl]prop-2-enoic acid (15a): To a solution of 14a (20 mg, 0.04 mmol) in dry toluene (10 mL) was added silica gel (3 mL) and the suspension was stirred at reflux overnight. The reaction was allowed to cool and the mixture was filtered after diluting with 20 % MeOH in DCM, dried (Na2S04), filtered and the solvent evaporated in vacuo to provide the title compound as a white solid (9.3 mg, 0.02 mmol, 50 %)
Rf: 0.00 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.70 (3H, s, CH3), 1.77 (3H, s, CH3), 2.72 (2H, t, J = 7.0 Hz, CH2), 3.03 (2H, t, J = 7.3 Hz, CH2), 3.21 (2H, d, J = 6.9 Hz, CH2), 5.19 (1H, t, J
= 7.0 Hz, CH), 6.42 (1H, d, J = 15.8 Hz, CH), 7.14 - 7.30 (5H, m, Ar-H), 7.34 - 7.46 (3H, m, Ar-H), 7.61 (1H, d, J = 16.5 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 29.4, 31.3, 37.8, 117.7, 121.3, 125.6, 125.9, 126.1, 127.8, 128.1, 128.1, 128.8, 132.3, 133.2, 136.5, 137.0, 140.6, 144.2, 169.0, 172.6. m/z (ESI): 364.2 [M + H] + (47.5 %), 386.3 [M + Na]+ (45 %). HRMS-ESI: (m/z) calculated for C23H25N03, 386.1727; found, 386.1729
(2E)-3-[4-acetamido-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoic acid (15b): To a solution of 14b (51.0 mg, 0.5 mmol) in dry toluene (6 mL) was added silica gel (3 mL) and heated at reflux overnight. The reaction was allowed to cool and was filtered with 20 % MeOH in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (DCM:MeOH = 1 :0, 9: 1, 4: 1) provided the title compound as a white solid (20.3 mg, 0.07 mmol, 50 %)
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.75 (3H, s, CH3), 1.78 (3H, s, CH3), 2.16 (3H, s, CH3), 3.37 (2H, d, J = 6.9 Hz, CH2), 5.25 (1H, t, J = 6.4 Hz, CH), 6.43 (1H, d, J = 16.0 Hz, CH), 7.43 - 7.58 (3H, m, Ar-H), 7.63 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz; MeOD): δ 21.7, 24.4, 29.3, 29.6, 31.6, 117.7, 121,3, 125.7, 126.1, 132.3, 133.2, 136.4, 137.2, 144.2, 170.7. m/z (ESI): 296.1 [M + Na] + (100 %), 274.2 [M + H]+ (95 %).
(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-(4-phenylbutanamido)phenyl]prop-2-enoic acid (15c): To a solution of 14c (17 mg, 0.04 mmol) in dry toluene (6 mL) was added silica gel (3 mL) and the reaction refluxed overnight. The reaction was allowed to cool and was filtered with 20% MeOH in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 4: 1, 2: 1, 1: 1) provided the title compound as a white solid (7.3 mg, 0.02 mmol, 50 %).
Rf: 0.00 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.74 (3H, s, CH3), 1.75 (3H, s, CH3), 2.07 (2H, d, J = 7.5 Hz, CH2), 2.33 (2H, d, J = 7.0 Hz, CH2), 2.73 (2H, d, J = 7.1 Hz, CH2), 3.33 (2H, d, J = 6.5 Hz, CH2), 5.19 (1H, t, J = 6.3 Hz, CH), 6.39 (1H, d, J = 16.9 Hz, CH), 7.20 - 7.46 (7H, m, Ar-H), 7.73 (1H, d, J = 16.3 Hz, CH), 8.17 (1H, d, J = 8.6 Hz, Ar-H). 13C NMR (100 MHz; MeOD): δ 13.0, 16.6, 19.4, 24.4, 27.3, 29.8, 34.9, 35.4, 60.1, 119.3, 121.5, 125.6, 126.1, 127.8, 128.1, 128.8, 132.6, 133.2, 136.5, 141.5, 143.0, 173.3. m/z (ESI): 378.3 [M + H]+ (100 %), 400.3 [M + Na]+ (100 %).
(2E)-3-[4-(2-cyclohexylacetamido)-3-(3-methylbut-2-en-l-yl)phenyl]prop-2-enoic acid (15d): To a solution of 14d (20 mg, 0.05 mmol) in dry toluene (6 mL) was added silica gel (3 mL) and heated at reflux overnight. The reaction was allowed to cool and was filtered with 20% MeOH in DCM, dried (Na2S04) filtered and concentrated in vacuo. Purification by column chromatography (Hexane:EtOAC = 4:1, 2: 1, 1 : 1) Provided the title compound as a white solid (13.2 mg, 0.03 mmol, 93 %).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.03 - 1.38 (7H, m, CH2), 1.73 (3H, s, CH3), 1.78 (3H, s, CH3), 1.78 - 1.90 (4H, m, CH2), 2.29 (2H, d, J = 6.9 Hz, CH2), 3.37 (2H, d, J = 6.9 Hz, CH2), 5.27 (1H, t, J = 7.7 Hz, CH), 6.43 (1H, d, J = 15.9 Hz, CH), 7.43 - 7.49 (3H, m, Ar-H), 7.63 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 25.8, 25.9, 29.3, 29.6, 32.8, 35.5, 44.0, 117.9, 121.4, 125.7, 126.2, 129.0, 132.3, 133.3, 136.5, 137.1, 144.1, 173.0. m/z (ESI): 356.3 [M + H]+ (100 %), 378.2 [M + Na]+ (75 %).
(2E)-3-[3-(3-methylbut-2-en-l-yl)-4-[2-(naphthalen-l-yl)acetamido]phenyl]prop-2-enoic acid (15e): To a solution of 14e (50 mg, 0.1 mmol) in dry toluene (6 mL) was added silica gel (3 mL) and stirred at reflux overnight. The reaction was allowed to cool and was filtered with 20 % MeOH in DCM, dried (Na2S04) filtered and concentrated in vacuo. Purification by column chromatography (Hexane:EtOAc = 4: 1, 2: 1, 1 : 1) provided the title compound as a white solid (20.7 mg, 0.05 mmol, 51.8 %).
Rf: 0.01 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.80 (6H, s, 2x CH3), 3.36 (2H, s, CH2), 3.45 (2H, d, J = 5.8 Hz, CH2), 5.23 (1H, t, J = 7.7 Hz, CH), 6.43 (1H, d, J = 16.4 Hz, CH), 7.43 - 7.64 (7H, m, Ar-H and CH), 7.88 - 7.95 (3H, m, Ar-H), 8.37 (1H, d, J = 7.9 Hz, Ar-H). 13C NMR (100 MHz; MeOD): δ 24.5, 30.5, 72.7, 120.7, 122.7, 124.0, 124.8, 125.4, 125.7, 125.8, 126.3, 128.3, 128.6, 129.2, 131.3, 131.3, 133.5, 134.2, 134.8, 136.0, 137.1, 144.0, 172.4. m/z (ESI): 397.9 [M - H] - (20 %)
methyl (E)-3-(3-hydroxyphenyl)acrylate (16a): To a solution of m-coumaric acid (1 g, 6 mmol) in methanol (10 mL), was added sulphuric acid (0.6 mL) and the mixture refluxed overnight. The reaction mixture was allowed to cool and washed with brine, water, extracted in DCM, dried (Na2S04), filtered and concentrated to provide the title compound as a white solid. (1 g, 5.6 mmol, 93 %).
Rf: 0.29 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz, CDC13): δ 3.82 (3H, s, -OCH3), 6.42 (1H, d, J = 17 Hz, CH) 6.89 (1H, d, J = 8.13 Hz, Ar-H), 7.02 (1H, s, Ar-H), 7.11 (1H, d J = 7.5 Hz, Ar-H), 7.65 (1H, d, J = 17.0 Hz, CH). 13C NMR (100MHz, CDC13): δ 51.9, 60.5, 114.3, 119.1, 120.3, 134.0, 134.9, 143.4, 167.0, 171.4.
methyl (E)-3-(4-bromo-3-hydroxyphenyl)acrylate (17): To a solution of 16a (1 g, 5.6 mmol), in HOAc (8 mL) was added a solution of Br2 (310 μΐ., 6 mmol) in HOAc (2 mL) and the solution stirred at room temperature for 5 hr. A further solution of Br2 (85 μΕ, 1.6 mmol) in HOAc (0.9 mL) and the mixture stirred overnight at room temperature. The reaction mixture was washed with brine, saturated aqueous NaHC03 and extracted in Et20, dried (Na2S04), filtered and concentrated. Purification by column chromatography (16 % DCM in EtOAc:Hexane = 1 : 1) provided the title compound as a brown oil (610 mg, 2.3 mmol, 35 %).
Rf: 0.23 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 3.83 (3H, s, -OCH3), 6.33 (1H, d, J = 15.0 Hz, CH), 6.79 (1H, d, J = 8.6 Hz, ArCH), 7.12 (1H, s, Ar-H), 7.42 (1H, d, J = 8.5 Hz, Ar-H), 7.99 (1 H, d, J = 15.9 Hz, CH). 13C NMR (100MHz; CDC13): δ 51.9, 114.3, 115.3, 119.2, 120.2, 134.0, 143.5, 155.9, 167.2, 171.7.
(E)-3-(4-bromo-3-hydroxyphenyl)acrylic acid (17a): To a solution of 17 (330 mg, 1.3 mmol) in water (8 mL) and THF (2 mL) was added NaOH (100 mg, 2.5 mmol) and the reaction refluxed overnight. The solution was allowed to cool and was acidified with AcOH, washed with water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purifcation by column chromatography (Hexane:EtOAc = 9:1, 4: 1, 2: 1, 1: 1, 0: 1) provided the title compound as a white solid (150 mg, 0.6 mmol, 45%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 6.39 (1H, d, J = 15.8 Hz, CH), 6.78 (1H, dd, Jl = 2.9 Hz, J2 = 8.7 Hz, ArCH), 7.15 (1H, d, J = 2.9 Hz, ArCH), 7.44 (1H, d, J = 8.7 Hz, ArCH),
7.97 (1 H, d, J = 15.9 Hz, CH). 13C NMR (100MHz; MeOD): δ 113.6, 113.7, 118.9, 120.5, 133.6, 134.6, 142.9, 157.7, 168.3.
(E)-3-(4-bromo-3-((3-phenylpropanoyl)oxy)phenyl)acrylic acid (18): To a solution of (17a) (280 mg, 0.8 mmol) in DCM (8 mL) was added DMAP (12 mg, 0.1 mmol), NEt3 (210 μΐ., 1.5 mmol) and PhCH2CH2COCl (400 mg, 2.5 mmol) in DCM (2 mL) and the reaction stirred overnight at room temperature. The reaction was washed with saturated aqueous NaHC03 and water, dried (Na2S04), filtered and concentrated. Purifcation by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1 : 1, 0:1) provided the title compound as a white solid (110 mg, 0.3mmol, 36%).
Rf: 0.1 (Hex:EtOAc = 4: 1). 1H NMR (CDC13, 400 MHz): δ 2.94 (2H, t, J = 7.5 Hz, CH2), 3.11 (2H, t, J = 7.5 Hz, CH2), 6.37 (1H, d, J = 15.8 Hz, CH), 6.96 (1H, m, ArCH), 7.31 (6H, m, ArCH), 7.62 (1H, d, J = 8.6 Hz, ArCH), 8.12 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz, CDC13): δ 30.9, 35.8, 120.8, 120.9, 121.9, 125.0, 126.6, 128.4, 128.6, 134.2, 135.1, 139.7, 144.4, 150.0, 171.0, 171.3.
(E)-3-(4-(3-methylbut-2-en-l-yl)-3-((3-phenylpropanoyl)oxy)phenyl)acrylic acid (19a): To a solution of (18) (430 mg, 1.1 mmol) in dry DMF (8 mL) was added CsC03 (555 mg, 1.7 mmol) and Pd(dppf)C12 (50 mg, 0.06 mmol). Prenyl boronic acid pinacol ester (330 μΕ, 1.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was filtered through a celite® pad with EtOAc, the solvent was evaporated and the residue re-dissolved in DCM. The residual DMF was removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAC = 9: 1, 4: 1, 2: 1, 1: 1, 0: 1) provided the title compound as a yellow oil (120 mg, 0.3 mmol, 27%).
Rf: 0.10 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.72 (3H, s, CH3), 1.77 (3H, s, CH3), 2.90 (2H, t, J = 7.2 Hz, CH2), 3.04 (2H, t, J = 7.2 Hz, CH2), 3.43 (2H, d, J = 6.7 Hz, CH2), 5.15 (1H, t, J = 5.9 Hz, CH), 6.29 (2H, d, J = 15.7 Hz, CH), 6.94 (1H, d, J = 7.2 Hz, Ar-H), 7.16 - 7.33 (7H, m, Ar-H), 7.93 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 30.5, 31.3, 35.3, 119.0, 119.9, 122.3, 122.9, 126.0, 128.14, 128.18, 130.4, 132.3, 134.0, 138.6, 140.2, 141.5, 149.3, 168.5, 171.7. m/z
(ESI): 387.2 [M + Na]+ (100 %), 365.2 [M + H]+ (70 %). HRMS-ESI: (m/z) calculated for C23H2404, 363.1602; found, 363.1609.
(E)-3-(4-allyl-3-((3-phenylpropanoyl)oxy)phenyl)acrylic acid (19b): To a solution of (18) (110 mg, 0.3 mmol) in dry DMF (4 mL) was added CsC03 (75 mg, 0.4 mmol) and Pd(dppf)C12 (13 mg, 0.01 mmol). Allyl boronic acid pinacol ester (70 μΐ., 0.4 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was filtered through a celite® pad with EtOAc, the solvent was evaporated and the residue re-dissolved in DCM. The residual DMF was removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAC = 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a yellow oil (60 mg, 0.2 mmol, 60%).
Rf: 0.10 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 2.93 (2H, t, J = 7.5 Hz, CH2), 3.11 (2H, t, J = 7.5 Hz, CH2), 3.53 (2H, d, J = 6.1 Hz, CH2), 5.01 (1H, dd, Jl = 1.5 Hz, J2 = 17.0 Hz, CH2), 5.12 (1H, dd, Jl = 1.5 Hz, J2 = 10.1 Hz, CH2), 5.90 - 6.00 (1H, m, CH), 6.33 (2H, d, J = 15.7 Hz, CH), 7.02 (1H, dd, Jl = 2.4 Hz, J2 = 8.3 Hz, ArCH), 7.20 - 7.38 (7H, m, ArCH), 8.03 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 30.9, 35.9, 36.9, 116.7, 119.4, 119.5, 123.6, 126.5, 128.4, 128.6, 131.3, 134.1, 136.1, 137.0, 139.9, 143.4, 149.3 171.3, 171.5.
tert-butyl (2E)-3-(3-amino-4-bromophenyl)prop-2-enoate (21a): To a solution of 2-bromo-5-iodo aniline (250 mg, 0.8 mmol) in dry toluene (6 mL) was added PPh3 (26 mg, 0.1 mmol) and Pd(OAc)2 (47.5 %, 24 mg, 0.05 mmol). tert-butyl acrylate (160 μΐ., 1.1 mmol) and NEt3 (210 μΐ., 1.5 mmol) were added and the reaction was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc gradient = 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a white solid (160 mg, 0.5 mmol, 64 %).
Rf: 0.19 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 6.25 (1H, d, J = 15.9 Hz, CH), 6.60 (1H, s, Ar-H), 6.92 (1H, s, Ar-H), 7.34 (1H, d, J = 8.4 Hz, Ar-H), 7.89 (1H, d J = 15.8
Hz, CH). 13C NMR (100 MHz; CDC13): δ 28.18, 80.7, 113.5, 118.3, 122.4, 131.9, 133.7, 134.9, 142.1, 145.8, 165.8.
(E)-2-bromo-5-(2-(4,4,5,5 etramethyl-l,3,2-dioxaborolan-2-yl)vinyl)aniline (21b): To a solution of 2- bromo-5-iodo aniline (300 mg, 1 mmol) in dry toluene (10 mL) was added PPh3 (26 mg, 0.1 mmol) and Pd(OAc)2 (11 mg, 0.05 mmol). 4,4,5,5-tetramethyl-2-vinyl-l,3,2-dioxaborolane (170 μΐ., 1 mmol) and NEt3 (225 μΐ., 1.5 mmol) were added and the reaction was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc gradient = 9: 1, 4: 1, 2: 1, 1: 1, 0: 1) provided the title compound as a brown oil (64 mg, 0.15 mmol, 15%).
Rf: 0.13 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.33 (12H, s, CH3), 6.06 (1H, d, J = 18.2 Hz, CH), 6.56 (1H, dd, Jl = 2.6 Hz, J2 = 8.4 Hz, ArCH), 6.96 (1H, d, J = 2.6 Hz, ArCH), 7.32 (1H, d, J = 8.4 Hz, ArCH), 7.65 (1H, d J = 18.2 Hz, CH). 13C NMR (100 MHz; CDC13): δ 24.8, 83.4, 113.1, 113.6, 117.6, 133.4, 137.7, 145.0, 147.7.
tert-butyl (E)-3-(4-bromo-3-(3-phenylpropanamido)phenyl)acrylate (23a): To a solution of 21a (160 mg, 0.5 mmol) in dry DCM (9 mL) was added DMAP (8 mg, 0.6 mmol), a solution of PhCH2CH2COCl (170 mg, 1 mmol) in DCM (2 mL) and NEt3 (0.1 mL, 1 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1, 1 : 1) provided the title compound as a white solid (200 mg, 0.4 mmol, 88 %).
Rf: 0.32 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 2.68 (2H, t, J = 7.4 Hz, CH2), 3.03 (2H, t, J = 7.4 Hz, CH2), 6.29 (1H, d, J = 15.8 Hz, CH), 7.18 - 7.29 (5H, m, Ar-H), 7.43 (1H, d, J = 8.3, Ar-H), 7.76 (1H, s, Ar-H), 7.84 (1H, s, Ar-H), 7.89 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 28.1, 31.4, 39.2, 80.9, 118.7, 119.5, 123.2, 126.4, 128.3, 128.6, 133.5, 134.9, 137.5, 140.4, 141.6, 165.8, 170.8
tert-butyl (E)-3-(4-bromo-3-cinnamamidophenyl)acrylate (23b): To a solution of 21a (190 mg, 0.6 mmol) in dry DCM (9 mL) was added DMAP (13 mg, 0.1 mmol), a solution of cinnamoyl chloride (166 mg, 1 mmol) in DCM (2 mL) and NEt3 (0.1 mL, 1 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow solid (100 mg, 0.2 mmol, 36%).
Rf: 0.33 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 6.34 (1H, d, J = 15.8 Hz, CH), 6.62 (1H, d, J = 15.5 Hz, CH), 7.31 - 7.37 (3H, m, ArCH), 7.46 - 7.49 (4H, m, ArCH), 7.76 (1H, d, J = 15.9 Hz, CH) 7.91 (1H, d, J = 15.9 Hz, CH), 8.04 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 14.2, 21.1, 28.1, 60.4, 81.0, 123.2, 127.9, 128.9, 130.1, 133.6, 134.3, 134.9, 137.8, 141.6, 142.9, 164.4, 165.8, 171.3.
tert-butyl (E)-3-(4-bromo-3-(3-(4-fluorophenyl)propanamido)phenyl)acrylate (23c): To a solution of 21a (140 mg, 0.5 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-(4- fluorophenyl)propanoyl chloride (223 mg, 1.2 mmol) in DCM (2 mL) and NEt3 (0.2 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow solid (150 mg, 0.3 mmol, 70%).
Rf: 0.13 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 2.65 (2H, t, J = 7.4 Hz, CH2), 3.00 (2H, t, J = 7.6 Hz, CH2), 6.29 (1H, d, J = 15.8 Hz, CH), 6.94 (2H, t, J = 8.6 Hz, ArCH), 7.14 (2H, t, J = 8.5 Hz, ArCH), 7.25 - 7.28 (1H, m, ArCH), 7.45 (1H, d, J = 8.6, ArCH), 7.86 - 7.90 (2H, m, ArCH and CH).
tert-butyl (E)-3-(4-bromo-3-(3-(3-fluorophenyl)propanamido)phenyl)acrylate (23d): To a solution of 21a (250 mg, 0.8 mmol) in dry DCM (8 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-(3- fluorophenyl)propanoyl chloride (250 mg, 1.5 mmol) in DCM (2 mL) and NEt3 (0.2 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow solid (400 mg, 0.8 mmol, 94%).
Rf: 0.5 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 2.67 (2H, t, J = 7.4 Hz, CH2), 3.02 (2H, t, J = 7.7 Hz, CH2), 6.29 (1H, d, J = 15.8 Hz, CH), 6.85 - 6.91 (2H, m, ArCH), 6.98 (1H, d, J = 7.6 Hz, ArCH), 7.20 - 7.29 (2H, m, ArCH), 7.46 (1H, d, J = 8.6, ArCH), 7.87 (2H, d, J = 16.0 Hz, ArCH and CH). 13C NMR (100 MHz; CDC13): δ 28.1, 30.9, 38.7, 81.0, 113.2, 113.4, 115.0, 115.2, 118.7, 122.4, 123.2, 124.0, 130.1, 133.5, 134.9, 137.5, 141.6, 143.0, 165.8, 170.4.
tert-butyl (E)-3-(4-bromo-3-(3-(2-fluorophenyl)propanamido)phenyl)acrylate (23e): To a solution of 21a (180 mg, 0.6 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-(2- fluorophenyl)propanoyl chloride (223 mg, 1.2 mmol) in DCM (2 mL) and NEt3 (0.2 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow solid (300 mg, 0.6 mmol, 90%).
Rf: 0.4 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 2.69 (2H, t, J = 7.4 Hz, CH2), 3.07 (2H, t, J = 7.7 Hz, CH2), 6.30 (1H, d, J = 15.8 Hz, CH), 6.99 - 7.07 (2H, m, ArCH), 7.16 - 7.30 (3H, m, ArCH), 7.47 (1H, d, J = 8.6 Hz, ArCH), 7.85 (1H, d, J = 2.4, ArCH), 7.88 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 25.0, 28.1, 37.6, 80.9, 115.2, 115.4, 118.7, 122.4, 123.3, 124.2, 124.2, 128.2, 128.3, 130.8, 130.8, 133.5, 134.9, 141.5, 165.8, 170.5.
tert-butyl (E)-3-(4-bromo-3-(3-(4-methoxyphenyl)propanamido)phenyl)acrylate (23f): To a solution of 21a (100 mg, 0.3 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-(4- methoxyphenyl)propanoyl chloride (200 mg, 1 mmol) in DCM (2 mL) and NEt3 (0.1 mL, 1 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (120 mg, 0.26 mmol, 76%).
Rf: 0.25 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 2.64 (2H, t, J = 7.3 Hz, CH2), 3.00 (2H, t, J = 7.4 Hz, CH2), 3.79 (3H, s, CH3), 6.31 (1H, d, J = 15.8 Hz, CH), 6.84 (2H, d, J = 8.6 Hz, ArCH), 7.14 (2H, d, J = 8.6 Hz, ArCH), 7.25 - 7.28 (1H, m, ArCH), 7.48 (1H, d, J = 8.6 Hz, ArCH), 7.82 (1H, d, J = 2.4, ArCH), 7.80 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 28.1, 30.5, 39.6, 55.2, 80.9, 114.0, 118.5, 119.4, 122.3, 123.3, 129.3, 132.3, 133.5, 134.9, 137.4, 141.5, 158.1, 165.7, 170.7.
(E)-3-(4-bromo-3-(3-(3-methoxyphenyl)propanamido)phenyl)acrylic acid (23g): To a solution of 21a (170 mg, 0.6 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-(3- methoxyphenyl)propanoyl chloride (300 mg, 1.5 mmol) in DCM (2 mL) and NEt3 (0.2 mL, 2 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (260 mg, 0.5 mmol, 90%).
Rf: 0.24 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, C(CH3)3), 2.66 (2H, t, J = 7.4 Hz, CH2), 2.98 (2H, t, J = 7.7 Hz, CH2), 3.73 (3H, s, CH3), 6.27 (1H, d, J = 15.8 Hz, CH), 6.71 - 6.78 (3H, m, ArCH), 7.16 (1H, t, J = 8.4 Hz, ArCH), 7.28 (1H, d, J = 8.6 Hz, ArCH), 7.42 (1H, d, J = 8.6 Hz, ArCH), 7.84 (1H, s, ArCH), 7.86 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 28.1,
31.4, 38.9, 55.0, 80.9, 111.5, 114.1, 118.7, 119.4, 120.6, 122.5, 123.1, 129.6, 133.4, 134.7, 137.7, 141.6, 142.0, 159.7, 165.8, 171.0.
tert-butyl (E)-3-(4-bromo-3-(3-(2-methoxyphenyl)propanamido)phenyl)acrylate (23h): To a solution of 21a (230 mg, 0.8 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-(2- methoxyphenyl)propanoyl chloride (300 mg, 1.5 mmol) in DCM (2 mL) and NEt3 (0.2 mL, 2 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (330 mg, 0.7 mmol, 93%).
Rf: 0.22 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 2.67 (2H, t, J = 7.3 Hz, CH2), 3.04 (2H, t, J = 7.7 Hz, CH2), 3.80 (3H, s, CH3), 6.32 (1H, d, J = 15.8 Hz, CH), 6.85 - 6.92 (2H, m, ArCH), 7.16 - 7.28 (3H, m, ArCH), 7.47 (1H, d, J = 8.6 Hz, ArCH), 7.87 (1H, d, J = 2.4 Hz, ArCH), 7.90 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz; CDC13): δ 26.4, 28.1, 37.7, 55.3, 80.8, 110.3, 118.6, 119.2, 120.7, 122.3, 123.2, 127.8, 128.5, 130.1, 133.4, 134.9, 137.7, 141.6, 157.2, 165.7, 171.3.
tert-butyl (E)-3-(4-bromo-3-(3-(4-(tosyloxy)phenyl)propanamido)phenyl)acrylate (23i): To a solution of 21a (516 mg, 1.7 mmol) in dry DCM (10 mL) was added DMAP (26 mg, 0.2 mmol), a solution of 4-(3- chloro-3-oxopropyl)phenyl 4-methylbenzenesulfonate (630 mg, 2 mmol) in DCM (2 mL) and NEt3 (0.6 mL, 4.5 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a brown oil (600 mg, 1 mmol, 58%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.51 (9H, s, C(CH3)3), 2.41 (3H, s, CH3), 2.63 (2H, t, J = 7.6 Hz, CH2), 2.96 (2H, t, J = 7.6 Hz, CH2), 6.29 (1H, d, J = 15.8 Hz, CH), 6.80 (2H, d, J = 8.5 Hz, ArCH), 7.07 (2H, d, J = 8.5 Hz, ArCH), 7.24 - 7.29 (2H, m, ArCH), 7.42 (2H, d, J = 8.6 Hz,
ArCH), 7.63 (2H, d, J = 8.3 Hz, ArCH), 7.86 (1H, d, J = 15.8, CH), 7.93 (1H, d, J = 2.4 Hz, ArCH), 8.17 (1H, br-s, NH). 13C NMR (100MHz, CDC13): 5 21.7, 28.1, 30.5, 38.6, 81.0, 115.5, 118.6, 119.3, 122.3, 122.4, 123.1, 128.3, 129.5, 129.8, 132.0, 133.4, 134.7, 137.7, 139.8, 141.6, 145.6, 147.8, 165.9, 170.6.
tert-butyl (E)-3-(4-bromo-3-(3-(3-(tosyloxy)phenyl)propanamido)phenyl)acrylate (23j): To a solution of 21a (490 mg, 1.6 mmol) in dry DCM (10 mL) was added DMAP (26 mg, 0.2 mmol), a solution of 3-(3- chloro-3-oxopropyl)phenyl 4-methylbenzenesulfonate (575 mg, 1.7 mmol) in DCM (2 mL) and NEt3 (0.4 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a brown oil (514 mg, 0.9 mmol, 56%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, C(CH3)3), 2.42 (3H, s, CH3), 2.60 (2H, t, J = 7.5 Hz, CH2), 2.97 (2H, t, J = 7.6 Hz, CH2), 6.30 (1H, d, J = 15.8 Hz, CH), 6.70 (1H, d, J = 8.9 Hz, ArCH), 6.94 (1H, s, ArCH), 7.08 - 7.16 (2H, m, ArCH), 7.25 - 7.32 (3H, m, ArCH), 7.45 (1H, d, J = 8.6 Hz, ArCH), 7.63 (2H, d, J = 8.3 Hz, ArCH), 7.64 (1H, d, J = 8.3, CH), 7.87 (1H, d, J = 15.7 Hz, CH), 7.89 (1H, s, ArCH), 8.17 (1H, br-s, NH). 13C NMR (100MHz, CDC13): δ 21.7, 28.1, 30.8, 38.4, 80.9, 118.6, 119.4, 120.0, 122.1, 122.4, 123.2, 127.3, 128.3, 129.6, 129.8, 132.1, 133.5, 134.8, 137.6, 141.5, 142.6, 145.5, 149.6, 165.7, 170.3, 171.2.
(E)-N-(2-bromo-5-(2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)vinyl)phenyl)-3-phenylpropanamide (24): To a solution of 21a (64 mg, 0.1 mmol) in dry DCM (5 mL) was added DMAP (2 mg, 0.01 mmol), a solution of 3-phenyl propanoyl chloride (50 mg, 0.3 mmol) in DCM (2 mL) and NEt3 (0.1 mL, 0.5 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (40 mg, 0.08 mmol, 87%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.33 (12H, s, CH3), 2.67 (2H, t, J = 7.4 Hz, CH2), 3.05 (2H, t, J = 7.6 Hz, CH2), 6.10 (1H, d, J = 18.2 Hz, CH), 7.22 - 7.34 (6H, m, ArCH), 7.47 (1H, d, J = 8.6 Hz, ArCH), 7.59 (1H, d, J = 2.2 Hz, ArCH), 7.65 (1H, d, J = 18.0 Hz, CH). 13C NMR (100 MHz; CDC13): δ 24.8, 31.4, 39.4, 83.5, 118.2, 118.7, 121.6, 126.4, 128.3, 128.6, 133.3, 137.2, 137.8, 140.4, 147.0, 170.3.
tert-butyl (E)-3-(4-(3-methylbut-2-en-l-yl)-3-(3-phenylpropanamido)phenyl)acrylate (25a): To a solution of 23a (200 mg, 0.4 mmol) in dry DMF (2 mL) was added CsC03 (230 mg, 0.7 mmol) and Pd(dppf)C12 (40 mg, 0.05 mmol). Prenyl boronic acid pinacol ester (140 μΐ., 0.62 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (80 mg, 0.2 mmol, 42 %).
Rf: 0.33 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.73 (3H, s, CH3), 1.74 (3H, s, CH3), 2.67 (2H, t, J = 7.4 Hz, CH2), 3.05 (2H, t, J = 7.4 Hz, CH2), 3.38 (2H, d, J = 6.4 Hz, CH2), 5.16 (1H, t, J = 5.6 Hz, CH), 6.27 (1H, d, J = 15.7 Hz, CH), 7.11 (1H, d, J = 8.0 Hz, Ar-H), 7.22 - 7.35 (5H, m, Ar-H), 7.57 (1H, s, Ar-H), 7.71 (1H, s, Ar-H), 7.84 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 28.2, 31.5, 31.5 39.2, 80.5, 117.9, 121.8, 122.4, 126.3, 128.3, 128.6, 130.1, 132.7, 133.8, 136.1, 137.1, 140.6, 140.8, 166.3, 170.6
tert-butyl (E)-3-(3-cinnamamido-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25b): To a solution of 23b (100 mg, 0.2 mmol) in dry DMF (4 mL) was added Cs2C03 (163 mg, 0.5 mmol) and Pd(dppf)C12 (25 mg, 0.03 mmol). Prenyl boronic acid pinacol ester (111 μΐ., 0.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated.
Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (80 mg, 0.2 mmol, 83%).
Rf: 0.5 (Hexane:EtOAc, 4:1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.74 (3H, s, CH3), 3.39 (2H, d, J = 6.9 Hz, CH2), 5.16 (1H, t, J = 7.1 Hz, CH), 6.31 (1H, d, J = 15.7 Hz, CH), 6.64 (1H, d, J = 15.5 Hz, CH), 7.15 (1H, d, J = 8.2 Hz, ArCH), 7.31 - 7.38 (3H, m, ArCH), 7.47 - 7.58 (3H, m, ArCH), 7.76 (1H, d, J = 15.5 Hz, CH), 7.86 (1H, d, J = 15.8 Hz, CH), 7.91 (1H, s, ArCH). 13C NMR (100MHz , CDC13): δ 17.9, 25.7, 28.2, 31.6, 80.5, 117.9, 120.8, 121.6, 122.4, 127.9, 128.8, 129.9, 130.2, 132.7, 133.8, 134.5, 136.5, 137.2, 140.8, 142.3, 164.2, 166.4
tert-butyl (E)-3-(3-(3-(4-fluorophenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25c): To a solution of 23c (150 mg, 0.3 mmol) in dry DMF (2 mL) was added Cs2C03 (230 mg, 0.7 mmol) and Pd(dppf)C12 (40 mg, 0.05 mmol). Prenyl boronic acid pinacol ester (100 uL, 0.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (66 mg, 0.15 mmol, 50%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.50 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.74 (3H, s, CH3), 2.63 (2H, t, J = 7.4 Hz, CH2), 3.02 (2H, t, J = 7.4 Hz, CH2), 3.38 (2H, d, J = 7.0 Hz, CH2), 5.14 (1H, t, J = 7.0 Hz, CH), 6.27 (1H, d, J = 15.7 Hz, CH), 6.96 (2H, t, J = 8.6 Hz, ArCH), 7.12 (1H, d, J = 8.1 Hz, ArCH), 7.18 (2H, t, J = 8.4 Hz, ArCH), 7.34 (1H, dd, Jl = 2.1 Hz, J2 = 8.2 Hz, ArCH), 7.45 (1H, s, NH), 7.72 (1H, d, J = 2.0, ArCH), 7.84 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 28.2, 30.6, 31.5, 39.3, 80.5, 115.2, 115.4, 117.9, 121.5, 121.9, 122.4, 129.7, 129.8, 130.1, 132.8, 133.8, 136.0, 136.2, 137.2, 140.7, 166.2, 170.2.
tert-butyl (E)-3-(3-(3-(3-fluorophenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25d): To a solution of 23d (400 mg, 0.9 mmol) in dry DMF (10 mL) was added Cs2C03 (530 mg, 1.8 mmol) and
Pd(dppf)C12 (104 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (260 uL, 1.1 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (150 mg, 0.4 mmol, 44%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.74 (3H, s, CH3), 2.66 (2H, t, J = 7.2 Hz, CH2), 3.04 (2H, t, J = 7.4 Hz, CH2), 3.37 (2H, d, J = 6.9 Hz, CH2), 5.15 (1H, t, J = 7.0 Hz, CH), 6.26 (1H, d, J = 15.7 Hz, CH), 6.87 - 6.94 (2H, m, ArCH), 6.99 (1H, d, J = 7.6 Hz, ArCH), 7.21 - 7.28 (1H, m, ArCH), 7.35 (1H, dd, Jl = 1.9 Hz, J2 = 8.2 Hz, ArCH), 7.58 (1H, s, NH), 7.71 (1H, d, J = 1.8, ArCH), 7.83 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 28.2, 31.0, 31.5, 38.7, 80.5, 113.1, 113.3, 115.1, 115.3, 117.9, 121.5, 121.9, 122.4, 124.0, 124.0, 130.0, 130.1, 140.7, 143.1, 161.6, 164.1, 166.3, 170.2.
tert-butyl (E)-3-(3-(3-(2-fluorophenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25e): To a solution of 23e (300 mg, 0.6 mmol) in dry DMF (10 mL) was added Cs2C03 (460 mg, 1.4 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (200 uL, 0.8 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (200 mg, 0.4 mmol, 69%).
Rf: 0.4 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 1.71 (3H, s, CH3), 1.73 (3H, s, CH3), 2.67 (2H, t, J = 7.4 Hz, CH2), 3.06 (2H, t, J = 7.6 Hz, CH2), 3.36 (2H, d, J = 6.9 Hz, CH2), 5.14 (1H, t, J = 7.0 Hz, CH), 6.25 (1H, d, J = 15.7 Hz, CH), 6.98 - 7.05 (2H, m, ArCH), 7.09 (1H, d, J = 8.9 Hz, ArCH), 7.15 - 7.24 (2H, m, ArCH), 7.35 (1H, dd, Jl = 1.8 Hz, J2 = 8.2 Hz, ArCH), 7.71 (1H, d, J = 1.8, ArCH), 7.79 (1H, s, NH), 7.82 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.1, 25.7, 28.2, 31.5, 37.5, 80.5, 115.1, 115.3, 118.2, 121.8, 122.5, 124.1, 127.3, 128.1, 130.1, 130.8, 132.2, 133.7, 136.2, 137.1, 140.8, 159.8, 162.3, 166.3, 170.4.
tert-butyl (E)-3-(3-(3-(4-methoxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25f): To a solution of 23f (120 mg, 0.3 mmol) in dry DMF (3 mL) was added Cs2C03 (166 mg, 0.5 mmol) and Pd(dppf)C12 (22 mg, 0.03 mmol). Prenyl boronic acid pinacol ester (110 μΐ., 0.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (50 mg, 0.1 mmol, 37%).
Rf: 0.25 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.74 (3H, s, CH3), 2.63 (2H, t, J = 7.4 Hz, CH2), 2.99 (2H, t, J = 7.6 Hz, CH2), 3.38 (2H, d, J = 7.0 Hz, CH2), 3.78 (3H, s, CH3), 5.15 (1H, t, J = 7.0 Hz, CH), 6.26 (1H, d, J = 15.7 Hz, CH), 6.83 (2H, d, J = 8.5 Hz, ArCH), 7.10 - 7.15 (3H, m, ArCH), 7.35 (1H, d, J = 9.4 Hz, ArCH), 7.49 (1H, s, NH), 7.69 (1H, s, ArCH), 7.84 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 28.2, 30.7, 31.5, 39.6, 55.2, 80.5, 114.0, 117.9, 121.5, 121.8, 122.4, 129.3, 130.1, 132.6, 132.7, 133.7, 136.1, 137.1, 140.8, 158.1, 166.3, 170.7.
tert-butyl (E)-3-(3-(3-(3-methoxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25g): To a solution of 23g (200 mg, 0.6 mmol) in dry DMF (10 mL) was added Cs2C03 (460 mg, 1.4 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (200 μΐ., 0.8 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (240 mg, 0.5 mmol, 81%).
Rf: 0.43 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.74 (3H, s, CH3), 2.66 (2H, t, J = 7.4 Hz, CH2), 3.02 (2H, t, J = 7.6 Hz, CH2), 3.37 (2H, d, J =
6.2 Hz, CH2), 3.76 (3H, s, CH3), 5.15 (1H, t, J = 7.0 Hz, CH), 6.26 (1H, d, J = 15.7 Hz, CH), 6.75 - 6.82 (3H, m, ArCH), 7.11 (1H, d, J = 8.3 Hz, ArCH), 7.20 (1H, t, J = 7.7 Hz, ArCH), 7.36 (1H, dd, Jl = 2.0 Hz, J2 = 8.2 Hz, ArCH), 7.60 (1H, s, NH), 7.69 (1H, d, J = 2.0 Hz, ArCH), 7.83 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 28.2, 31.5, 31.6, 55.1, 80.5, 111.6, 114.1, 117.9, 120.7, 121.6, 121.8, 122.5, 129.6, 130.1, 132.7, 133.7, 136.1, 137.1, 140.8, 142.2, 159.7, 166.3, 170.6.
tert-butyl (E)-3-(3-(3-(2-methoxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylate (25h): To a solution of 23h (330 mg, 0.7 mmol) in dry DMF (10 mL) was added Cs2C03 (460 mg, 1.4 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (200 μΐ., 0.8 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (290 mg, 0.6 mmol, 89%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.74 (3H, s, CH3), 2.66 (2H, t, J = 7.5 Hz, CH2), 3.04 (2H, t, J = 7.6 Hz, CH2), 3.37 (2H, d, J = 6.9 Hz, CH2), 3.80 (3H, s, CH3), 5.15 (1H, t, J = 7.0 Hz, CH), 6.27 (1H, d, J = 15.8 Hz, CH), 6.86 (2H, q, J = 6.5 Hz, ArCH), 7.09 (1H, d, J = 8.2 Hz, ArCH), 7.15 - 7.22 (2H, m, ArCH), 7.36 (1H, dd, Jl = 1.9 Hz, J2 = 8.2 Hz, ArCH), 7.74 (1H, d, J = 1.8 Hz, ArCH), 7.79 (1H, s, NH), 7.84 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 25.7, 26.5, 28.2, 31.5, 37.6, 55.2, 80.4, 110.3, 117.9, 120.6, 121.5, 121.7, 122.5, 127.6, 128.8, 130.0, 130.0, 132.6, 133.6, 136.4, 136.9, 140.8, 157.3, 166.3, 171.3.
tert-butyl (E)-3 -(4-(3-methy lbut-2-en- 1 -y l)-3 -(3 -(4-(tosy loxy)pheny l)propanamido)pheny l)acry late (25i) : To a solution of 23i (600 mg, 1 mmol) in dry DMF (5 mL) was added Cs2C03 (490 mg, 1.5 mmol) and Pd(dppf)C12 (80 mg, 0.1 mmol). Prenyl boronic acid pinacol ester (340 μΐ., 1.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by
washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (400 mg, 0.7 mmol, 68%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, C(CH3)3), 1.70 (3H, s, CH3), 1.72 (3H, s, CH3), 2.41 (3H, s, CH3), 2.62 (2H, t, J = 7.5 Hz, CH2), 2.98 (2H, t, J = 7.5 Hz, CH2), 3.36 (2H, d, J = 6.9 Hz, CH2), 5.13 (1H, t, J = 7.0 Hz, CH), 6.26 (1H, d, J = 15.7 Hz, CH), 6.83 (2H, d, J = 8.5 Hz, ArCH), 7.10 (2H, d, J = 8.4 Hz, ArCH), 7.25 (2H, d, J = 8.0 Hz, ArCH), 7.33 (1H, dd, Jl = 8.2 Hz, J2 = 2.1 Hz, ArCH), 7.64 (2H, d, J = 8.3 Hz, ArCH), 7.77 (1H, d, J = 2.0 Hz, ArCH), 7.82 (1H, s, ArCH), 7.83 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; CDC13): δ 17.9, 24.8, 25.6, 28.1, 30.7, 31.5, 38.7, 80.5, 117.8, 121.4, 121.8, 122.3, 122.4, 128.4, 129.5, 129.7, 130.1, 132.2, 133.7, 136.2, 137.1, 139.8, 140.8, 145.4, 147.9, 166.3, 170.3.
tert-buty 1 (E)-3 -(4-(3-methy lbut-2-en- 1 -y l)-3 -(3 -(3-(tosy loxy)pheny l)propanamido)pheny l)acry late (25j) : To a solution of 23j (270 mg, 0.4 mmol) in dry DMF (4 mL) was added Cs2C03 (211 mg, 0.6 mmol) and Pd(dppf)C12 (32 mg, 0.04 mmol). Prenyl boronic acid pinacol ester (110 μΐ., 0.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (290 mg, 0.7 mmol, 50%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.51 (9H, s, C(CH3)3), 1.69 (3H, s, CH3), 1.72 (3H, s, CH3), 2.40 (3H, s, CH3), 2.58 (2H, t, J = 7.5 Hz, CH2), 2.95 (2H, t, J = 7.5 Hz, CH2), 3.37 (2H, d, J = 6.9 Hz, CH2), 5.13 (1H, t, J = 7.0 Hz, CH), 6.24 (1H, d, J = 15.7 Hz, CH), 6.72 (1H, d, J = 7.8 Hz, ArCH), 6.91 (1H, s, ArCH), 7.07 - 7.15 (3H, m, ArCH), 7.24 (2H, d, J = 8.0 Hz, ArCH), 7.36 (1H, dd, Jl = 8.2 Hz, J2 = 2.1 Hz, ArCH), 7.64 (2H, d, J = 8.3 Hz, ArCH), 7.75 (1H, d, J = 2.0 Hz, ArCH), 7.82 (1H, d, J = 15.8 Hz, CH), 7.92 (1H, s, NH). 13C NMR (100 MHz; CDC13): δ 17.9, 21.6, 25.6, 28.1, 30.9, 31.5, 38.4, 80.4, 117.8, 119.9, 121.5, 121.8, 122.1, 122.5, 127.3, 128.3, 129.6, 129.7, 130.1, 132.6, 136.3, 137.0, 140.7, 142.8, 145.4, 149.6, 166.2, 170.2.
(E)-3-(4-(3-methylbut-2-en-l-yl)-3-(3-phenylpropanamido)phenyl)acrylic acid (26a): To a solution of 25a (80 mg, 0.2 mmol) in dry toluene (6 mL) was added silica gel (3 mL) and the suspension stirred at reflux overnight. The reaction was allowed to cool and the mixture filtered, washing with 20 % MeOH in DCM, and the solvent evaporated in vacuo to provide the title compound as a white solid (18.7 mg, 0.05 mmol, 27 %).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz, MeOD): δ 1.72 (3H, s, CH3), 1.79 (3H, s, CH3), 2.66 (2H, t, J = 7.4 Hz, CH2), 3.01 (2H, t, J = 7.4 Hz, CH2), 3.43 (2H, d, J = 6.6 Hz, CH2), 5.16 (1H, t, J = 5.9 Hz, CH), 6.33 (1H, d, J = 15.7 Hz, CH), 7.16 - 7.30 (6H, m, Ar-H), 7.46 (1H, d, J = 8.0 Hz, Ar-H), 7.80 (1H, s, Ar-H), 7.96 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz, MeOD): δ 16.6, 24.4, 31.3, 31.3, 38.4, 117.5, 119.2, 121.7, 122.6, 125.8, 128.1, 129.9, 132.0, 133.2, 136.9, 137.0, 140.7, 142.3, 168.8, 172.2. m/z (ESI): 364.2 [M + H]+ (100%), 386.3 [M + Na]+ (17.5%). HRMS-ESI: (m/z) calculated for C23H25N03, 362.1762; found, 362.1761.
(E)-3-(3-cinnamamido-4-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (26b): Compound 25b (80 mg, 0.2 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1 : 1) provided the title compound as a white soild (20 mg, 0.05 mmol, 30%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz, MeOD): δ 1.74 (3H, s, CH3), 1.80 (3H, s, CH3), 3.47 (2H, d, J = 7.0 Hz, CH2), 5.19 (1H, t, J = 7.1 Hz, CH), 6.39 (1H, d, J = 15.8 Hz, CH), 6.81 (1H, d, J = 15.6 Hz, CH), 7.23 (1H, d, J = 8.3 Hz, ArCH), 7.39 - 7.46 (3H, m, ArCH), 7.61 - 7.64 (3H, m, ArCH), 7.69 (1H, d, J = 15.6 Hz, CH), 8.0 (1H, d, J = 15.7 Hz, CH), 8.0 (1H, d, J = 2.1 Hz, ArCH). 13C NMR (100MHz, MeOD): δ 16.6, 24.5, 31.4, 99.9, 117.4, 119.5, 120.7, 121.6, 122.6, 127.5, 128.6, 129.6, 130.0, 132.1, 133.4, 134.8, 137.1, 137.1, 141.5, 142.2, 165.2
(E)-3-(3-(3-(4-fluorophenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (26c): Compound 25c (66 mg, 0.15 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a white soild (50 mg, 0.13 mmol, 87%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.73 (3H, s, CH3), 1.79 (3H, s, CH3), 2.66 (2H, t, J = 7.4 Hz, CH2), 3.00 (2H, t, J = 7.4 Hz, CH2), 3.4 (2H, d, J = 7.4 Hz, CH2), 5.16 (1H, t, J = 7.8 Hz, CH), 6.33 (1H, d, J = 17.5 Hz, CH), 7.00 (2H, t, J = 8.6 Hz, ArCH), 7.18 (1H, d, J = 8.3 Hz, ArCH), 7.27 (2H, dd, Jl = 3.2 Hz, J2 = 5.5 Hz, ArCH), 7.46 (1H, dd, Jl = 2.1 Hz, J2 = 8.2 Hz, ArCH), 7.81 (1H, d, J = 2.0, ArCH), 7.96 (1H, d, J = 17.3 Hz, CH). 13C NMR (100 MHz; MeOD): δ 24.4, 29.2, 30.4, 31.3, 38.4, 114.5, 114.7, 117.5, 121.6, 122.6, 129.6, 129.7, 129.9, 132.2, 133.3, 136.6, 136.9, 136.9, 142.1, 162.7, 172.0.
(E)-3 -(3 -(3-(3 -fluoropheny l)propanamido)-4-(3 -methy lbut-2-en- 1 -y l)pheny l)acry lie acid (26d) : Compound 25d (150 mg, 0.4 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a white soild (120 mg, 0.32 mmol, 80%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.72 (3H, s, CH3), 1.78 (3H, s, CH3), 2.68 (2H, t, J = 7.3 Hz, CH2), 3.02 (2H, t, J = 7.4 Hz, CH2), 3.41 (2H, d, J = 6.9 Hz, CH2), 5.15 (1H, t, J = 7.0 Hz, CH), 6.33 (1H, d, J = 15.7 Hz, CH), 6.92 (1H, t, J = 8.2 Hz, ArCH), 7.02 (1H, d, J = 9.8 Hz, ArCH), 7.08 (1H, d, J = 7.6 Hz, ArCH), 7.17 (1H, d, J = 8.3 Hz, ArCH), 7.29 (1H, q, J = 7.9 Hz, ArCH), 7.46 (1H, dd, Jl = 2.1 Hz, J2 = 8.2 Hz, ArCH), 7.79 (1H, d, J = 2.1, ArCH), 7.95 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 30.9, 31.3, 37.9, 112.4, 112.6, 114.6, 114.8, 117.5, 121.7, 122.6, 123.9, 129.7, 129.8, 129.9, 132.0, 133.3, 136.8, 137.0, 161.7, 164.1, 171.9.
(E)-3-(3-(3-(2-fluorophenyl)propanamido)-4-(3-methylbut-2-en- 1 -yl)phenyl)acrylic acid (26e) : Compound 25e (200 mg, 0.4 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a white soild (130 mg, 0.34 mmol, 85%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.72 (3H, s, CH3), 1.78 (3H, s, CH3), 2.68 (2H, t, J = 7.4 Hz, CH2), 3.05 (2H, t, J = 7.6 Hz, CH2), 3.41 (2H, d, J = 6.9 Hz, CH2), 5.14 (1H, t, J = 7.0 Hz, CH), 6.32 (1H, d, J = 15.7 Hz, CH), 7.03 - 7.11 (2H, m, ArCH), 7.16 (1H, d, J = 8.3 Hz, ArCH), 7.20 - 7.25 (1H, m, ArCH), 7.30 (1H, t, J = 7.6 Hz, ArCH), 7.45 (1H, dd, Jl = 1.8 Hz, J2 = 8.2 Hz, ArCH), 7.80 (1H, d, J = 2.1, ArCH), 7.96 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 24.6, 31.3, 36.7, 114.6, 114.8, 117.6, 119.2, 121.8, 122.6, 123.9, 127.9, 129.9, 130.5, 132.0, 133.2, 137.0, 142.3, 159.8, 1652.3, 168.9, 171.9.
(E)-3-(3-(3-(4-methoxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (26f): Compound 25f (50 mg, 0.1 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a white soild (25 mg, 0.06 mmol, 53%).
Rf: 0.25 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.73 (3H, s, CH3), 1.79 (3H, s, CH3), 2.64 (2H, t, J = 7.4 Hz, CH2), 2.95 (2H, t, J = 7.6 Hz, CH2), 3.44 (2H, d, J = 7.2 Hz, CH2), 3.76 (3H, s, CH3), 5.15 (1H, t, J = 8.4 Hz, CH), 6.33 (1H, d, J = 15.7 Hz, CH), 6.85 (2H, d, J = 8.6 Hz, ArCH), 7.18 (3H, d, J = 8.4 Hz, ArCH), 7.46 (1H, d, Jl = 2.3 Hz, J2 = 8.2 Hz, ArCH), 7.79 (1H, d, J = 2.1 Hz, ArCH), 7.96 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.5, 24.4, 30.5, 31.3, 38.7, 54.2, 113.4, 117.6, 119.4, 121.7, 122.6, 128.9, 129.9, 132.0, 132.6, 133.2, 136.9, 136.9, 142.2, 157.8, 158.2, 172.4.
(E)-3-(3-(3-(3-methoxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acry acid (26g): Compound 25g (240 mg, 0.5 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a white soild (62 mg, 0.16 mmol, 31%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.73 (3H, s, CH3), 1.79 (3H, s, CH3), 2.67 (2H, t, J = 7.4 Hz, CH2), 2.99 (2H, t, J = 7.6 Hz, CH2), 3.43 (2H, d, J = 6.7 Hz, CH2), 3.76 (3H, s, CH3), 5.16 (1H, t, J = 7.0 Hz, CH), 6.33 (1H, d, J = 15.7 Hz, CH), 6.76 (1H, dd, Jl = 2.5 Hz, J2 = 7.9 Hz, ArCH), 6.83 - 6.85 (2H, m, ArCH), 7.16 - 7.22 (2H, m, ArCH), 7.46 (1H, dd, Jl = 2.2 Hz, J2 = 8.2 Hz, ArCH), 7.80 (1H, d, J = 2.1 Hz, ArCH), 7.97 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 31.3, 31.4, 38.3, 5.1, 111.3, 113.6, 117.6, 129.3, 120.3, 121.8, 122.6, 129.0, 129.9, 132.0, 133.2, 136.9, 137.0, 142.2, 142.3, 159.8, 168.8, 172.3.
(E)-3-(3-(3-(2-methoxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (26h): Compound 25h (290 mg, 0.6 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a white soild (110 mg, 0.3 mmol, 50%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.73 (3H, s, CH3), 1.79 (3H, s, CH3), 2.64 (2H, t, J = 7.5 Hz, CH2), 3.00 (2H, t, J = 7.6 Hz, CH2), 3.43 (2H, d, J = 6.9 Hz, CH2), 3.84 (3H, s, CH3), 5.15 (1H, t, J = 7.0 Hz, CH), 6.33 (1H, d, J = 15.8 Hz, CH), 6.86 (1H, t, J = 7.5 Hz, ArCH), 6.93 (1H, d, J = 7.9 Hz, ArCH), 7.16 - 7.21 (3H, m, ArCH), 7.46 (1H, dd, Jl = 2.2 Hz, J2 = 8.3 Hz, ArCH), 7.81 (1H, d, J = 2.1 Hz, ArCH), 7.97 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 24.4, 26.3, 31.3, 36.8, 54.2, 109.9, 117.7, 119.1, 120.0, 121.8, 122.6, 127.3, 128.6, 129.5, 129.9, 132.0, 133.1, 136.9, 137.0, 142.4, 157.4, 168.7, 172.7.
(E)-3-(4-(3-methylbut-2-en-l-yl)-3-(3-(4-(tosyloxy)phenyl)propanamido)phenyl)acrylic acid (26i): Compound 25i (400 mg, 0.7 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a white soild (104 mg, 0.2 mmol, 28%).
Rf: 0.4 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 1.72 (3H, s, CH3), 1.79 (3H, s, CH3), 2.39 (3H, s, CH3), 2.65 (2H, t, J = 7.5 Hz, CH2), 2.99 (2H, t, J = 7.5 Hz, CH2), 3.45 (2H, d, J = 6.8 Hz, CH2), 5.16 (1H, t, J = 7.0 Hz, CH), 6.34 (1H, d, J = 15.7 Hz, CH), 6.88 (2H, d, J = 8.5 Hz, ArCH), 7.18 - 7.31 (5H, m, ArCH), 7.44 (1H, d, J = 8.3 Hz, ArCH), 7.59 (2H, d, J = 8.2 Hz, ArCH), 7.82 (1H, d, J = 1.8 Hz, ArCH), 7.97 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.5, 20.1, 24.4, 30.5, 31.3 38.0, 117.4, 121.6, 122.0, 122.6, 128.2, 129.3, 129.4, 129.9, 132.0, 132.1, 133.3, 136.9, 137.0, 140.0, 142.0, 145.6, 148.1.
(E)-3-(4-(3-methylbut-2-en-l-yl)-3-(3-(3-(tosyloxy)phenyl)propanamido)phenyl)acrylic acid (26j): Compound 25j (290 mg, 0.5 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (DCM:MeOH = 20: 1, 10: 1) provided the title compound as a white soild (110 mg, 0.2 mmol, 41%).
Rf: 0.2 (EtOAC). 1H NMR (400 MHz; MeOD): δ 1.70 (3H, s, CH3), 1.76 (3H, s, CH3), 2.39 (3H, s, CH3), 2.61 (2H, t, J = 7.5 Hz, CH2), 2.94 (2H, t, J = 7.5 Hz, CH2), 3.40 (2H, d, J = 6.9 Hz, CH2), 5.13 (1H, t, J = 7.0 Hz, CH), 6.32 (1H, d, J = 15.7 Hz, CH), 6.75 (1H, d, J = 7.3 Hz, ArCH), 6.95 (1H, s, ArCH), 7.14 - 7.22 (3H, m, ArCH), 7.30 (2H, d, J = 8.0 Hz, ArCH), 7.47 (1H, dd, Jl = 8.2 Hz, J2 = 2.1 Hz, ArCH), 7.61 (2H, d, J = 8.3 Hz, ArCH), 7.81 (1H, d, J = 2.1 Hz, ArCH), 7.95 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.6, 20.2, 24.5, 30.7, 31.3, 37.7, 117.6, 119.4, 119.6, 121.7, 121.9, 122.6, 127.0, 128.2, 129.2, 129.5, 129.9, 132.0, 132.1, 133.2, 136.9, 137.0, 142.2, 143.0, 145.6, 149.7, 168.8, 171.6.
(E)-3-(3-(3-(4-hydroxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (26k): Compound 26i (80 mg, 0.15 mmol) was dissolved in MeOH (4 mL) and refluxed with 1M NaOH (2 mL) overnight. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a white soild (50 mg, 0.12 mmol, 84%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; MeOD): δ 1.73 (3H, s, CH3), 1.79 (3H, s, CH3), 2.62 (2H, t, J = 7.5 Hz, CH2), 2.92 (2H, t, J = 7.5 Hz, CH2), 3.44 (2H, d, J = 7.4 Hz, CH2), 5.16 (1H, t, J = 7.0 Hz, CH), 6.33 (1H, d, J = 15.7 Hz, CH), 6.71 (2H, d, J = 8.5 Hz, ArCH), 7.08 (2H, d, J = 8.5 Hz, ArCH), 7.17 (1H, d, J = 8.3 Hz, ArCH), 7.46 (1H, d, Jl = 2.1 Hz, J2 = 8.2 Hz, ArCH), 7.81 (1H, d, J = 2.1 Hz, ArCH), 7.97 (1H, d, J = 15.8 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.5, 24.4, 30.6, 31.3, 38.8, 114.8, 117.6, 121.7, 122.6, 128.9, 129.8, 131.4, 132.2, 133.2, 136.9, 142.2, 155.3, 168.9, 172.5.
(E)-3-(3-(3-(3-hydroxyphenyl)propanamido)-4-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (261): Compound 26j (15 mg, 0.02 mmol) was dissolved in MeOH (2 mL) and refluxed with 1M NaOH (2 mL) overnight. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a white soild (8 mg, 0.02 mmol, 90%).
Rf: 0.2 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 1.73 (3H, s, CH3), 1.79 (3H, s, CH3), 2.65 (2H, t, J = 7.5 Hz, CH2), 2.94 (2H, t, J = 7.5 Hz, CH2), 3.44 (2H, d, J = 7.0 Hz, CH2), 5.17 (1H, t, J = 7.0 Hz, CH), 6.35 (1H, d, J = 15.6 Hz, CH), 6.63 (1H, d, Jl = 2.1 Hz, J2 = 8.2 Hz, ArCH), 6.71 - 6.74 (2H, m, ArCH), 7.10 (1H, t, J = 7.4 Hz, ArCH), 7.17 (1H, d, J = 8.3 Hz, ArCH), 7.47 (1H, d, Jl = 2.1 Hz, J2 = 8.4 Hz, ArCH), 7.81 (1H, d, J = 2.4 Hz, ArCH), 7.95 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz; MeOD): δ 16.5, 24.4, 31.3, 31.3, 99.9, 122.7, 114.8, 117.6, 119.1, 121.7, 122.6, 129.0, 129.8, 132.0, 133.3, 136.9, 142.2, 157.1, 172.3.
tert-butyl (E)-3-(3'-(¾ydroxymethyl)-2-(3-phenylpropanamido)-[l,r-biphenyl]-4-yl)acrylate (27): To a solution of (23a) (100 mg, 0.2 mmol) in dry DMF (4 mL) was added Cs2C03 (166 mg, 0.5 mmol) and Pd(dppf)C12 (22 mg, 0.03 mmol). (3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)methanol (110 μΐ., 0.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9:1, 4: 1) provided the title compound as a transparent oil (70 mg, 0.15 mmol, 66%).
Rf: 0.5 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.47 (9H, s, C(CH3)3), 2.69 (2H, t, J = 7.6 Hz, CH2), 3.07 (2H, t, J = 7.6 Hz, CH2), 4.67 (2H, s, CH2), 6.29 (1H, d, J = 15.8 Hz, CH), 7.13 - 7.39 (12H, m, ArCH), 7.56 (1H, d, J = 15.9 Hz, CH), 7.79 (1H, s, NH), 7.85 (1H, s, OH). 13C NMR (100 MHz , CDC13): δ 28.1, 31.5, 39.2, 65.0, 80.6, 117.6, 121.1, 121.4, 126.1, 126.4, 128.3, 128.4, 128.6, 129.0, 130.8, 132.9, 137.2, 138.7, 139.5, 140.5, 141.0, 142.1, 166.5, 170.9.
(E)-3-(3'-(hydroxymethyl)-2-(3-phenylpropanamido)-[l,l'-biphenyl]-4-yl)acrylic acid (28): Compound 27 (70 mg, 0.15 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (DCM:MeOH = 20: 1, 10: 1) provided the title compound as a white soild (27 mg, 0.06 mmol, 45%).
Rf: 0.3 (DCM:MeOH, 10: 1).1H NMR (400MHz , MeOD): δ 2.72 (2H, t, J = 7.6 Hz, CH2), 3.04 (2H, t, J = 7.6 Hz, CH2), 4.68 (2H, s, CH2), 6.41 (1H, d, J = 15.8 Hz, CH), 7.18 - 7.23 (2H, m, ArCH), 7.28 - 7.32 (6H, m, ArCH), 7.39 - 7.45 (2H, m, ArCH), 7.62 - 7.66 (2H, m, ArCH and CH), 7.99 (1H, d, J = 2.0 Hz, ArCH). 13C NMR (100 MHz , MeOD): δ 31.3, 38.5, 63.6, 117.3, 119.5, 121.3, 125.6, 125.8, 127.9, 128.0, 128.1, 128.5, 130.5, 132.6, 138.1, 138.6, 139.7, 140.7, 141.6, 143.2, 172.4.
N-(4-bromo-2-iodophenyl)-3-phenylpropanamide (30): To a solution of 4-bromo-2-iodo aniline (300 mg, 1 mmol) in dry DCM (5 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-phenylpropanoyl chloride (400 mg, 2 mmol) in DCM (2 mL) and NEt3 (0.4 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (300 mg, 0.7 mmol, 69%).
Rf: 0.5 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz, CDC13): δ 2.76 (2H, t, J = 7.4 Hz, CH2), 3.09 (2H, t, J = 7.8 Hz, CH2), 7.21 - 7.34 (5H, m, ArCH), 7.45 (1H, dd, Jl = 8.6 Hz, J2 = 2.2 Hz ArCH), 7.89 (1H, d, J = 2.2 Hz, ArCH), 8.14 (1H, d, J = 8.8 Hz, ArCH). 13C NMR (100 MHz, CDC13): δ 31.3, 39.6, 126.5, 128.3, 128.7, 132.2, 137.3 140.1, 140.4.
tert-butyl (E)-3-(5-bromo-2-(3-phenylpropanamido)phenyl)acrylate (31): To a solution of 30 (300 mg, 0.7 mmol) in dry toluene (10 mL) was added PPh3 (26 mg, 0.1 mmol) and Pd(OAc)2 (12 mg, 0.05 mmol). tert-butyl acrylate (160 μΕ, 1 mmol) and NEt3 (280 μΕ, 2 mmol) were added and the reaction was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc gradient = 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a white solid (210 mg, 0.5 mmol, 70 %).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz , CDC13): δ 1.53 (9H, s, C(CH3)3), 2.71 (2H, t, J = 7.6 Hz, CH2), 3.05 (2H, t, J = 7.3 Hz, CH2), 6.27 (1H, d, J = 15.7 Hz, CH), 7.20 - 7.24 (3H, m, ArCH), 7.30 - 7.35 (2H, m, ArCH), 7.40 (1H, dd, Jl = 8.6 Hz, J2 = 2.2 Hz ArCH), 7.51 - 7.56 (2H, m, ArCH and CH), 7.60 (1H, s, ArCH). 13C NMR (100 MHz, CDC13): δ 28.1, 31.4, 38.8, 81.1, 121.4, 123.7, 126.3, 126.4, 128.3, 128.3, 128.6, 128.6, 129.6, 131.8, 133.1, 134.6, 136.6, 140.3, 165.6, 171.1.
tert-butyl (E)-3-(5-(3-methylbut-2-en-l-yl)-2-(3-phenylpropanamido)phenyl)acrylate (32a): To a solution of 31 (100 mg, 0.2 mmol) in dry DMF (3 mL) was added Cs2C03 (160 mg, 0.5 mmol) and Pd(dppf)C12 (25 mg, 0.03 mmol). Prenyl boronic acid pinacol ester (100 μΐ., 0.4 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (30 mg, 0.07 mmol, 68%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400MHz , CDC13): δ 1.52 (9H, s, C(CH3)3), 1.73 (3H, s, CH3), 1.77 (3H, s, CH3), 2.73 (2H, t, J = 7.6 Hz, CH2), 3.08 (2H, t, J = 7.4 Hz, CH2), 3.32 (2H, d, J = 6.8 Hz, CH2), 5.29 (1H, t, J = 7.3 Hz, CH), 6.31 (1H, d, J = 15.7 Hz, CH), 7.16 - 7.26 (4H, m, ArCH), 7.30 - 7.34 (3H, m, ArCH), 7.54 (1H, d, J = 8.2 Hz, ArCH), 7.64 (1H, d, J = 15.7 Hz, CH). 13C NMR (100 MHz, CDC13): δ 17.8, 25.7, 28.2, 33.8, 80.8, 119.7, 120.1, 122.2, 122.5, 125.3, 126.6, 128.3, 128.4, 128.6, 130.6, 133.3, 138.3, 140.5, 166.0, 170.9.
tert-butyl (E)-3-(3'-(hydroxymethyl)-4-(3-phenylpropanamido)-[l,l'-biphenyl]-3-yl)acrylate (32b): To a solution of 31 (70 mg, 0.1 mmol) in dry DMF (3 mL) was added Cs2C03 (70 mg, 0.2 mmol) and Pd(dppf)C12 (10 mg, 0.01 mmol). (3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)methanol (50 μΐ., 0.2 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9:1, 4: 1) provided the title compound as a transparent oil (20 mg, 0.04 mmol, 27%).
Rf: 0.5 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz , CDC13): δ 1.55 (9H, s, C(CH3)3), 2.75 (2H, t, J = 7.6 Hz, CH2), 3.09 (2H, t, J = 7.4 Hz, CH2), 4.75 (2H, s, CH2), 6.32 (1H, d, J = 15.8 Hz, CH), 7.23 - 7.50 (10H, m, ArCH and CH), 7.63 - 7.67 (2H, m, ArCH), 7.77 (1H, d, J = 8.2 Hz, ArCH). 13C NMR (100 MHz, CDC13): δ 28.1, 31.5, 38.9, 65.1, 81.0, 122.6, 125.0, 125.3, 125.5, 126.1, 126.3, 127.5, 127.5, 128.3, 128.6, 129.0, 129.1, 134.9, 138.1, 140.1. 140.5, 141.5, 166.1, 171.0.
(E)-3-(5-((E)-3-methylbut-l-en-l-yl)-2-(3-phenylpropanamido)phenyl)acrylic acid (33a): Compound 32a (30 mg, 0.07 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1, 2: 1, 1: 1) provided the title compound as a white soild (18 mg, 0.05 mmol, 71%).
Rf: 0.4 (DCM:MeOH, 10: 1). 1H NMR (400 MHz , MeOD): δ 1.75 (3H, s, CH3), 1.74 (3H, s, CH3), 2.74 (2H, t, J = 7.6 Hz, CH2), 3.04 (2H, t, J = 7.4 Hz, CH2), 3.37 (2H, d, J = 8.7 Hz, CH2), 5.33 (1H, t, J = 8.8 Hz, CH), 6.42 (1H, d, J = 15.7 Hz, CH), 7.15 - 7.23 (3H, m, ArCH), 7.29 - 7.33 (4H, m, ArCH), 7.50 (1H, s, ArCH), 7.70 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz, MeOD): δ 24.4, 31.5, 33.2, 37.7, 122.5, 125.8, 126.8, 128.0, 128.1, 129.9, 130.1, 132.6, 132.8, 133.6, 133.7, 139.1, 140.4, 140.6, 173.2.
(E)-3-(3'-(hydroxymethyl)-4-(3-phenylpropanamido)-[l,l'-biphenyl]-3-yl)acrylic acid (33b): Compound 32b (20 mg, 0.05 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (DCM:MeOH = 20: 1, 10: 1) provided the title compound as a white soild (13 mg, 0.03 mmol, 73%).
Rf: 0.5 (Hexane:EtOAc, 1 : 1). 1H NMR (400 MHz , MeOD): δ 2.79 (2H, t, J = 7.3 Hz, CH2), 3.07 (2H, t, J = 7.8 Hz, CH2), 4.70 (2H, s, CH2), 6.57 (1H, d, J = 15.8 Hz, CH), 7.21 (1H, q, J = 4.4 Hz, ArCH), 7.31 - 7.47 (7H, m, ArCH), 7.57 (1H, d, J = 7.6 Hz, ArCH), 7.65 - 7.67 (2H, m, ArCH), 7.79 (1H, d, J = 15.8
Hz, CH), 7.94 (1H, s, ArCH). 13C NMR (100 MHz, MeOD): δ 31.4, 37.7, 63.7, 124.7, 125.0, 125.4, 125.8, 125.8, 126.9, 128.0, 128.1, 128.4, 128.6, 130.2, 135.1, 139.2, 139.4, 139.9, 140.6, 142.1 173.2.
N-(3-bromo-5-iodophenyl)-3-phenylpropanamide (36): To a solution of 35 (380 mg, 1.3 mmol) in dry DCM (12 mL) was added DMAP (13 mg, 0.1 mmol), a solution of 3-phenyl propanoyl chloride (320 mg, 2 mmol) in DCM (2 mL) and NEt3 (0.4 mL, 3 mmol). The reaction was heated to 700C and stirred overnight. The reaction was allowed to cool and was washed with saturated aqueous NaHC03, water and extracted in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 12: 1, 9: 1, 4: 1, 2: 1) provided the title compound as a white solid (430 mg, 1 mmol, 78%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.67 (2H, t, J = 7.4 Hz, CH2), 3.03 (2H, t, J = 7.5 Hz, CH2), 7.21 - 7.34 (5H, m, ArCH), 7.57 (1H, s, ArCH), 7.65 (1H, s, ArCH), 7.73 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 31.3, 39.3, 94.0, 122.1, 122.9, 126.5, 127.1, 128.3, 128.7, 135.3, 139.6, 140.2, 170.6.
tert-butyl (E)-3-(3-bromo-5-(3-phenylpropanamido)phenyl)acrylate (37): To a solution of 36 (123 mg, 0.3 mmol) in dry toluene (5 mL) was added PPh3 (8 mg, 0.03 mmol) and Pd(OAc)2 (4 mg, 0.01 mmol). tert-butyl acrylate (60 μΕ, 0.4 mmol) and NEt3 (70 μΕ, 0.5 mmol) were added and the reaction was refluxed overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc gradient = 9: 1, 4: 1, 2: 1, 1 : 1, 0: 1) provided the title compound as a brown oil (110 mg, 0.25 mmol, 91%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 2.67 (2H, t, J = 7.4 Hz, CH2), 3.03 (2H, t, J = 7.5 Hz, CH2), 6.31 (1H, d, J = 15.9 Hz, ArCH), 7.20 - 7.24 (5H, m, ArCH), 7.40 (1H, d, J = 15.9 Hz, ArCH), 7.52 (1H, s, ArCH), 7.68 (1H, s, ArCH), 7.71 (1H, s, ArCH).
13C NMR (100 MHz; CDC13): δ 28.1, 31.3, 39.1, 80.9, 117.8, 118.6, 122.0, 122.9, 126.4, 128.3, 128.6, 128.6, 140.3, 165.9, 171.1.
tert-butyl (E)-3-(3-(3-methylbut-2-en-l-yl)-5-(3-phenylpropanamido)phenyl)acrylate (38a): To a solution of 37 (110 mg, 0.2 mmol) in dry DMF (3 mL) was added Cs2C03 (130 mg, 0.4 mmol) and Pd(dppf)C12 (20 mg, 0.02 mmol). Prenyl boronic acid pinacol ester (0.1 mL, 0.4 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1) provided the title compound as a transparent oil (100 mg, 0.2 mmol, 95%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.52 (3H, s, CH3), 1.57 (3H, s, CH3), 2.67 (2H, t, J = 7.4 Hz, CH2), 3.07 (2H, t, J = 7.5 Hz, CH2), 3.31 (2H, d, J = 7.2 Hz, CH2), 5.29 (1H, t, J = 7.3 Hz, CH2), 6.36 (1H, d, J = 15.9 Hz, ArCH), 7.20 - 7.34 (8H, m, ArCH), 7.52 (1H, d, J = 15.9 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 25.7, 28.2, 31.5, 34.1, 39.4, 41.1, 80.5, 110.9, 111.3, 116.7, 120.6, 122.3, 126.3, 126.4, 128.3, 128.6, 128.6, 140.5, 143.2, 147.2, 147.6, 166.2, 170.4.
tert-butyl (E)-3-(3'-(hydroxymethyl)-5-(3-phenylpropanamido)-[l,l'-biphenyl]-3-yl)acrylate (38b): To a solution of 37 (130 mg, 0.3 mmol) in dry DMF (3 mL) was added Cs2C03 (195 mg, 0.6 mmol) and Pd(dppf)C12 (20 mg, 0.02 mmol). (3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)methanol (120 mg, 0.5 mmol) was added and the flask was heated at 900C overnight. The reaction was allowed to cool and was extracted over a pad of celite® with EtOAc, the solvent evaporated and re-dissolved in DCM. Residual DMF was removed by washing with copious amounts of water in DCM, the organic layer was dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1, 1 : 1) provided the title compound as a transparent oil (100 mg, 0.2 mmol, 73%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). δ 1.55 (9H, s, C(CH3)3), 2.64 (2H, t, J = 7.4 Hz, CH2), 3.09 (2H, t, J = 7.7 Hz, CH2), 4.68 (2H, s, CH2), 6.35 (1H, d, J = 15.9 Hz, ArCH), 7.19 - 7.50 (10H, m, ArCH and CH), 7.56 (1H, s, ArCH), 7.59 (1H, s, ArCH), 8.08 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 28.2, 31.4, 39.0, 64.9, 80.8, 120.3, 120.9, 122.5, 125.6, 126.1, 126.3, 128.3, 128.6, 128.6, 128.9, 129.2, 135.6, 138.7, 140.1, 140.5, 141.5, 142.1, 143.1, 166.3, 171.2.
(E)-3-(3-(3-methylbut-2-en-l-yl)-5-(3-phenylpropanamido)phenyl)acrylic acid (39a): Compound 38a (100 mg, 0.2 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (DCM:MeOH = 20: 1, 10: 1) provided the title compound as a white soild (55 mg, 0.15 mmol, 75%).
Rf: 0.2 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 1.75 (3H, s, CH3), 1.78 (3H, s, CH3), 2.68 (2H, t, J = 7.6 Hz, CH2), 3.01 (2H, d, J = 7.6 Hz, CH2), 3.35 (2H, d, J = 7.4 Hz, CH2), 5.33 (1H, t, J = 7.3 Hz, CH), 6.44 (1H, d, J = 15.9 Hz, CH), 7.13 (1H, s, ArCH), 7.17 - 7.21 (1H, m, ArCH), 7.29 - 7.32 (2H, m, ArCH), 7.38 (1H, s, ArCH), 7.55 - 7.70 (3H, m, ArCH). 13C NMR (100 MHz; MeOD): 27.2, 31.3, 33.5, 38.4, 110.3, 116.4, 118.6, 121.5, 122.3, 123.5, 125.8, 128.0, 128.0, 132.7, 139.0, 140.7, 143.2, 144.4, 172.3. HRMS-ESI: (m/z) calculated for C23H25N03, 386.1727 [M+Na]+; found, 386.1739.
(E)-3-(3'-(hydroxymethyl)-5-(3-phenylpropanamido)-[l,r-biphenyl]-3-yl)acrylic acid (39b): Compound 38b (100 mg, 0.2 mmol) was hydrolyzed following the general procedure B. Purification by column chromatography (DCM:MeOH = 20: 1, 10: 1) provided the title compound as a white soild (50 mg, 0.1 mmol, 60%).
Rf: 0.2 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 2.72 (2H, t, J = 7.3 Hz, CH2), 3.02 (2H, t, J = 7.7 Hz, CH2), 6.54 (1H, d, J = 15.8 Hz, CH), 7.17 - 7.22 (1H, m, ArCH), 7.28 - 7.29 (4H, m, ArCH), 7.37 - 7.39 (1H, m, ArCH), 7.44 (1H, t, J = 7.5 Hz, ArCH), 7.53 - 7.56 (2H, m, ArCH), 7.64 (1H, s, ArCH), 7.70 (1H, d, J = 16.0 Hz CH) 7.78 (1H, s, ArCH), 7.85 (1H, s, ArCH). 13C NMR (100 MHz; MeOD): 31.3, 38.5, 63.7, 117.5, 120.1, 122.2, 125.2, 125.5, 125.8, 126.0, 128.0, 128.1, 128.6, 135.5, 139.5, 140.0, 140.7, 142.1, 142.4, 144.5, 172.5.
3-bromo-5-iodo-N-phenethylbenzamide (41a): To a solution of 3-bromo-5-iodo benzoic acid (500 mg, 1.5 mmol) in dry toluene (10 mL) was added SOC12 (290 μΐ., 4 mmol) and the mixture was refluxed overnight. The reaction vessel was cooled to room temperature and the solvent was evaporated in vacuo. The resultant brown oil was used further without purification. To a solution of 3-bromo-5-iodo benzoyl chloride in DCM (10 mL) was added DMAP (26 mg, 0.2 mmol) and the flask was purged with nitrogen. 2-phenylethylamine (250 μΐ., 2 mmol) and NEt3 (420 μΐ., 3 mmol) were added to the flask and the mixture was stirred at 70oC overnight. The reaction mixture was cooled to room temperature, diluted with DCM, washed with a saturated aqueous NaHC03, water and extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1) provided the title compound as a white solid ( 645 mg, 1.5 mmol, 98%).
Rf: 0.51 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.93 (2H, t, J = 7.0 Hz, CH2), 3.68 (2H, q, J = 7.0 Hz, CH2), 7.22 - 7.36 (5H, m, ArCH), 7.77 (1H, t, J = 1.6 Hz, ArCH), 7.93 - 7.94 (2H, m, ArCH). 13C NMR (100 MHz; CDC13,): δ 35.5, 41.4, 94.4, 123.2, 126.7, 128.7, 129.5, 134.4, 137.9, 138.5, 142.2, 164.7.
3-bromo-N-(4-fluorophenethyl)-5-iodobenzamide (41b): Following the procedure as described for 41a the title compound was synthesized from 2-(4-fluorophenyl)ethan-l -amine (170 μΐ., 1.2 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white soild (300 mg, 0.7 mmol, 90%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.91 (2H, t, J = 7.0 Hz, CH2), 3.67 (2H, q, J = 7.0 Hz, CH2), 7.06 (2H, t, J = 8.0 Hz, ArCH), 7.17 - 7.20 (2H, m, ArCH), 7.78 (1H, t, J = 1.3 Hz, ArCH), 7.93 (1H, t, J = 1.4 Hz, ArCH), 7.96 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 34.7, 41.5, 94.5, 115.6, 123.2, 129.4, 130.2, 134.1, 134.6, 137.8, 142.3, 164.7.
3-bromo-5-iodo-N-(4-methoxyphenethyl)benzamide (41c): Following the procedure as described for 41a the title compound was synthesized from 2-(4-methoxyphenyl)ethan-l -amine (180 μΐ., 1.2 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white soild (420 mg, 0.9 mmol, 95%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.85 (2H, t, J = 7.0 Hz, CH2), 3.61 (2H, q, J = 7.0 Hz, CH2), 3.78 (3H, s, CH3), 6.85 (2H, d, J = 8.6 Hz, ArCH), 7.12 (2H, d, J = 8.6 Hz, ArCH), 7.79 (1H, t, J = 1.3 Hz, ArCH), 7.92 (1H, t, J = 1.4 Hz, ArCH), 7.94 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 34.6, 41.6, 55.2, 94.3, 114.0, 123.1, 129.5, 129.7, 130.5, 134.7, 138.0, 142.1, 158.3, 164.6.
3-bromo-5-iodo-N-(4-methoxybenzyl)benzamide (41d): Following the procedure as described for 41a the title compound was synthesized from (4-methoxyphenyl)methanamine (70 μΐ., 0.5 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white soild (HO mg, 0.2 mmol, 83%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 3.81 (3H, s, CH3), 4.52 (2H, d, J = 5.5 Hz, CH2), 6.88 (2H, d, J = 8.6 Hz, ArCH), 7.25 (2H, d, J = 8.7 Hz, ArCH), 7.85 (1H, t, J = 1.3 Hz, ArCH), 7.95 (1H, t, J = 1.4 Hz, ArCH), 8.01 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 43.8, 55.3, 94.4, 114.2, 123.2, 129.3, 129.6, 129.6, 134.7, 137.7, 142.3, 159.2, 164.4.
3-bromo-5-iodo-N-(4-(trifluoromethoxy)benzyl)benzamide (41e): Following the procedure as described for 41a the title compound was synthesized from (4-(tafluoromethoxy)phenyl)methanamine (230 μΐ., 1.5 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white soild (460 mg, 0.7 mmol, 99%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 4.60 (2H, d, J = 5.8 Hz, CH2), 7.19 (2H, d, J = 8.7 Hz, ArCH), 7.35 (2H, d, J = 8.7 Hz, ArCH), 7.87 (1H, t, J = 1.6 Hz, ArCH), 7.98 (1H, t, J = 1.6 Hz, ArCH), 8.02 (1H, t, J = 1.4 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 43.5, 94.5, 121.3, 123.3, 129.3, 129.6, 134.7, 136.3, 137.3, 142.6, 148.7, 164.6.
3-bromo-5-iodo-N-(3-(trifluoromethoxy)benzyl)benzamide (411): Following the procedure as described for 41a the title compound was synthesized from (3-(trifluoromethoxy)phenyl)methanamine (230 uL, 1.5 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white soild (300 mg, 0.6 mmol, 80%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 4.57 (2H, d, J = 5.8 Hz, CH2), 7.14 (2H, s, ArCH), 7.22 (2H, d, J = 8.7 Hz, ArCH), 7.33 - 7.37 (1H, m, ArCH), 7.84 (1H, t, J = 1.6 Hz, ArCH), 7.95 (1H, t, J = 1.6 Hz, ArCH), 8.00 (1H, t, J = 1.4 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 43.6, 94.5, 120.0, 120.2, 123.2, 126.0, 129.6, 130.1, 134.7, 137.2, 139.9, 142.5, 149.4, 164.8.
3-bromo-5-iodo-N-(4-methylbenzyl)benzamide (41g): Following the procedure as described for 41a the title compound was synthesized from p-tolylmethanamine (200 uL, 1.5 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white soild (490 mg, 0.7 mmol, 99%).
Rf: 0.4 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.37 (3H, s, CH3), 4.58 (2H, d, J = 5.5 Hz, CH2), 7.18 - 7.28 (4H, m, ArCH), 7.87 (1H, t, J = 1.6 Hz, ArCH), 7.98 (1H, t, J = 1.5 Hz, ArCH), 8.02 (1H, t, J = 1.5 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 21.1, 44.2, 94.4, 123.2, 128.8, 129.5, 134.4, 134.6, 137.7, 142.4, 164.3.
3-bromo-N-(4-(dimethylamino)benzyl)-5-iodobenzamide (41h): Following the procedure as described for 41a the title compound was synthesized from 4-(aminomethyl)-N,N-dimethylaniline (75 uL, 0.5 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a orange soild (130 mg, 0.2 mmol, 83%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.97 (6H, s, CH3), 4.51 (2H, d, J = 5.3 Hz, CH2), 6.73 (2H, d, J = 8.7 Hz, ArCH), 7.23 (2H, d, J = 8.7 Hz, ArCH), 7.86 (1H, t, J = 1.6 Hz, ArCH), 7.97 (1H, t, J = 1.5 Hz, ArCH), 8.01 (1H, t, J = 1.5 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 40.1, 40.5, 44.1, 94.4, 110.9, 112.7, 123.2, 124.8, 129.3, 129.5, 134.4, 137.9, 142.2, 150.3, 164.1.
phenethyl 3-bromo-5-iodobenzoate (42): Following the procedure as described for 41a the title compound was synthesized from 2-phenylethan-l-ol (130 μΐ., 1 mmol). Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a yellow oil (170 mg, 0.4 mmol, 80%).
Rf: 0.7 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 3.10 (2H, d, J = 7.0 Hz, CH2), 4.55 (2H, d, J = 7.0 Hz, ArCH), 7.28 - 7.39 (5H, m, ArCH), 8.04 (1H, t, J = 1.7 Hz, ArCH), 8.11 (1H, t, J = 1.6 Hz, ArCH), 8.28 (1H, t, J = 1.5 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 35.1, 66.2, 94.0, 123.0, 126.8, 128.6, 128.9, 131.9, 133.4, 137.1, 137.4, 143.7, 163.7.
tert-butyl (E)-3-(3-bromo-5-(phenethylcarbamoyl)phenyl)acrylate (43a): To a solution of 41a (645 mg, 1.5 mmol) in dry toluene (15 mL), was added PPh3 (40 mg, 0.15 mmol), Pd(OAc)2 (17 mg, 0.07 mmol) and the flask was purged with nitrogen. tert-Butylacrylate (300 μΐ., 2.0 mmol) and NEt3 (560 μΐ., 4.0 mmol) were added and the mixture was stirred at reflux overnight. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a brown oil (288 mg, 0.67 mmol, 45%).
Rf: 0.32 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 2.95 (2H, t, J = 6.9 Hz, CH2), 3.71 (2H, q, J = 6.9 Hz, CH2), 6.37 (1H, d, J = 15.9 Hz, CH), 7.23 - 7.36 (5H, m, ArCH), 7.45 (1H, d, J = 15.9 Hz, CH), 7.70 (2H, s, ArCH), 7.79 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 35.5, 41.3, 81.0, 122.8, 123.1, 124.9, 126.7, 128.7, 128.8, 130.9, 133.1, 137.0, 137.1, 138.6, 140.7, 165.4, 165.5.
tert-but l (E)-3-(3-bromo-5-((4-fluorophenethyl)carbamoyl)phenyl)acrylate (43b): Following the procedure as described for 43a the title compound was obtained as a white soild (70 mg, 0.16 mmol, 24%).
Rf: 0.20 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, (CH3)3), 2.91 (2H, t, J = 7.0 Hz, CH2), 3.69 (2H, q, J = 6.1 Hz, CH2), 6.39 (1H, d, J = 15.9 Hz, CH), 7.03 (2H, t, J = 8.6 Hz, ArCH), 7.18 - 7.21 (2H, m, ArCH), 7.47 (1H, d, J = 15.9 Hz, CH), 7.72 (2H, s, ArCH), 7.79 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 34.8, 41.4, 81.1, 115.4, 122.9, 123.2, 124.9, 130.2, 130.9, 133.1, 134.2, 134.2, 137.0, 137.0, 140.6, 165.4, 165.6.
tert-butyl (E)-3-(3-bromo-5-((4-methoxyphenethyl)carbamoyl)phenyl)acrylate (43c): Following the procedure as described for 43a the title compound was obtained as a white soild (200 mg, 0.4 mmol, 40%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, (CH3)3), 2.87 (2H, t, J = 7.0 Hz, CH2), 3.65 (2H, q, J = 6.1 Hz, CH2), 6.36 (1H, d, J = 15.9 Hz, CH), 6.86 (2H, d, J = 8.9 Hz, ArCH), 7.13 (2H, d, J = 8.6 Hz, ArCH), 7.49 (1H, d, J = 15.9 Hz, CH), 7.69 (1H, t, J = 1.4 Hz, ArCH). 7.72 (1H, t, J = 1.2 Hz, ArCH). 7.79 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 34.6, 41.5, 55.2, 81.0, 114.1, 122.7, 123.1, 125.0, 129.7, 130.5, 131.0, 133.0, 136.9, 137.1, 140.7, 158.3, 158.3, 165.5, 165.6.
tert-but l (E)-3-(3-bromo-5-((4-methoxybenzyl)carbamoyl)phenyl)acrylate (43d): Following the procedure as described for 43a the title compound was obtained as a white soild (40 mg, 0.09 mmol, 36%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, (CH3)3), 3.81 (3H, s, CH3), 4.57 (2H, d, J = 5.5 Hz, CH2), 6.40 (1H, d, J = 15.9 Hz, CH), 6.89 (2H, d, J = 8.9 Hz, ArCH), 7.27 - 7.29 (2H, m, ArCH), 7.48 (1H, d, J = 15.9 Hz, CH), 7.74 (1H, t, J = 1.4 Hz, ArCH). 7.82 (1H, t, J = 1.2 Hz, ArCH). 7.87 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 43.8, 55.3, 81.0, 114.2, 122.9, 123.1, 125.1, 129.4, 129.7, 131.0, 133.1, 136.9, 137.0, 140.7, 159.2, 165.2, 165.4.
tert-butyl (E)-3-(3-bromo-5-((4-(trifluoromethoxy)benzyl)carbamoyl)phenyl)acrylate (43e): Following the procedure as described for 43a the title compound was obtained as a white soild (210 mg, 0.4 mmol,
53' %).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, (CH3)3), 4.61 (2H, d, J = 5.8 Hz, CH2), 6.38 (1H, d, J = 15.9 Hz, CH), 7.18 (2H, d, J = 7.9 Hz, ArCH), 7.35 (2H, d, J = 8.6 Hz, ArCH), 7.44 (1H, t, J = 15.9 Hz, CH), 7.72 (1H, t, J = 1.4 Hz, ArCH). 7.82 (1H, t, J = 1.3 Hz, ArCH). 7.88 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 43.4, 81.1, 119.1, 121.2, 121.6, 122.9, 123.2, 125.1, 129.2, 131.0, 133.3, 136.5, 137.1, 140.6, 148.6, 165.4, 165.6.
tert-butyl (E)-3-(3-bromo-5-((3-(trifluoromethoxy)benzyl)carbamoyl)phenyl)acrylate (43f): Following the procedure as described for 43a the title compound was obtained as a white soild (110 mg, 0.2 mmol, 36%).
Rf: 0.28 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.52 (9H, s, (CH3)3), 4.63 (2H, d, J = 5.8 Hz, CH2), 6.38 (1H, d, J = 15.9 Hz, CH), 7.14 - 7.18 (2H, m, ArCH), 7.26 - 7.28 (1H, m, ArCH), 7.36 (1H, t, J = 7.9 Hz, CH), 7.45 (1H, t, J = 15.9 Hz, CH), 7.73 (1H, t, J = 1.4 Hz, ArCH). 7.82 (1H, t, J = 1.3 Hz, ArCH). 7.88 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 43.6, 81.1, 120.0, 120.3, 122.9, 123.2, 125.1, 126.1, 130.2, 131.0, 133.3, 136.5, 137.1, 140.1, 140.6, 165.4, 165.6.
tert-butyl (E)-3-(3-bromo-5-((4-methylbenzyl)carbamoyl)phenyl)acrylate (43 g): Following the procedure as described for 43a the title compound was obtained as a white soild (100 mg, 0.23 mmol, 30%).
Rf: 0.3 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, (CH3)3), 2.34 (3H, s, CH3), 4.55 (2H, d, J = 5.5 Hz, CH2), 6.37 (1H, d, J = 15.9 Hz, CH), 7.15 (2H, d, J = 7.8 Hz, ArCH), 7.22 (2H, d, J = 8.0 Hz, ArCH), 7.44 (1H, d, J = 15.9 Hz, CH), 7.70 (1H, t, J = 1.4 Hz, ArCH). 7.81 (1H, t, J = 1.2 Hz, ArCH). 7.87 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 21.1, 28.1, 44.0, 81.0, 122.8, 123.1, 125.1, 127.9, 129.4, 131.0, 133.1, 134.6, 136.8, 137.0, 137.4, 140.7, 165.4, 165.4.
tert-butyl (E)-3-(3-bromo-5-((4-(dimethylamino)benzyl)carbamoyl)phenyl)acrylate (43h): Following the procedure as described for 43a the title compound was obtained as a brown soild (90 mg, 0.2 mmol, 71%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, (CH3)3), 2.96 (6H, s, CH3), 4.53 (2H, d, J = 5.3 Hz, CH2), 6.41 (1H, d, J = 15.9 Hz, CH), 7.26 - 7.28 (5H, m, ArCH), 7.52 (1H, d, J = 15.9 Hz, CH), 7.80 (1H, t, J = 1.6 Hz, ArCH). 8.05 (1H, t, J = 1.3 Hz, ArCH). 8.11 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ
phenethyl (E)-3-bromo-5-(3-(tert-butoxy)-3-oxoprop-l-en-l-yl)benzoate (44): Following the procedure as described for 43a the title compound was synthesized from 42 as a yellow soild (125 mg, 0.3 mmol, 72%).
Rf: 0.65 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, C(CH3)3), 3.10 (2H, t, J = 6.9 Hz, CH2), 4.56 (2H, t, J = 7.0 Hz, CH2), 6.43 (1H, d, J = 15.9 Hz, CH), 7.26 - 7.38 (5H, m, ArCH),
7.45 (1H, d, J = 15.9 Hz, CH), 7.70 (2H, s, ArCH), 7.79 (1H, t, J = 1.6 Hz, ArCH). 13C NMR (100 MHz; CDC13,): δ 21.0, 28.1, 31.5, 66.0, 81.0, 122.9, 126.7, 127.3, 128.5, 128.6, 128.9, 132.6, 133.3, 134.6, 136.9, 137.5, 140.6, 164.6, 165.4, 171.0.
tert-butyl (E)-3-(3-(3-methylbut-2-en-l-yl)-5-(phenethylcarbamoyl)phenyl)acrylate (45a): To a solution of (43a) (80 mg, 0.18 mmol) in dry DMF (2 mL) was added Cs2C03 (130 mg, 0.4 mmol), Pd(dppf)C12 (8.5 mg, 0.01 mmol) and the flask was purged with nitrogen. Prenyl boronic acid pinacol ester (70 μΐ., 0.3 mmol) was added and the mixture was stirred at 90oC overnight. The reaction was allowed to cool and was filtered through a celite® pad with EtOAc. The solvent was evaporated in vacuo, re-dissolved in DCM and the residual DMF removed by washing with copious amounts of water in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4:1, 2: 1) provided the title compound as a transparent oil (50 mg, 0.12 mmol, 66%).
Rf: 0.50 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.77 (3H, s, CH3), 2.95 (2H, t, J = 6.9 Hz, CH2), 3.37 (2H, d, J = 7.3 Hz, CH2), 3.73 (2H, q, J = 6.1 Hz, CH2), 5.28 (1H, t, J = 7.3 Hz), 6.39 (1H, d, J = 15.9 Hz, CH), 7.25 - 7.37 (5H, m, ArCH), 7.41 (1H, s, ArCH), 7.51 (1H, s, ArCH), 7.55 (1H, d, J = 16.0 Hz, CH), 7.61 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 34.0, 35.6, 41.1, 80.6, 121.1, 121.9, 123.7, 126.6, 128.3, 128.7, 128.8, 130.6, 133.7, 135.0, 135.4, 138.8, 142.6, 143.1, 166.0, 167.1.
tert-butyl (E)-3-(3-((4-fluorophenethyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylate (45b): Following the procedure as described for 45a the title compound was obtained as a transparent oil (70 mg, 0.16 mmol, 95%).
Rf: 0.22 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.76 (3H, s, CH3), 2.92 (2H, t, J = 6.9 Hz, CH2), 3.37 (2H, d, J = 7.3 Hz, CH2), 3.69 (2H, q, J = 6.1 Hz, CH2), 5.28 (1H, t, J = 7.3 Hz), 6.39 (1H, d, J = 15.9 Hz, CH), 7.02 (2H, t, J = 8.6 Hz, ArCH), 7.20 (2H, t, J = 8.5 Hz, ArCH), 7.41 (1H, s, ArCH), 7.50 (1H, s, ArCH), 7.55 (1H, d, J = 16.0 Hz, CH), 7.62 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 28.1, 34.0, 34.9, 41.2, 80.7,
115.4, 115.6, 121.2, 121.9, 123.6, 128.3, 130.1, 130.2, 130.7, 133.7, 135.1, 135.3, 142.6, 143.1, 166.0,
tert-but l (E)-3-(3-((4-methoxyphenethyl)carbamo (45c): Following the procedure as described for 45a the title compound was obtained as a transparent oil (90 mg, 0.2 mmol, 46%).
Rf: 0.50 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.77 (3H, s, CH3), 2.89 (2H, t, J = 6.9 Hz, CH2), 3.37 (2H, d, J = 7.3 Hz, CH2), 3.68 (2H, q, J = 6.1 Hz, CH2), 3.81 (3H, s, CH3), 5.28 (1H, t, J = 5.9 Hz), 6.39 (1H, d, J = 15.9 Hz, CH), 6.89 (2H, d, J = 8.6 Hz, ArCH), 7.17 (2H, t, J = 8.6 Hz, ArCH), 7.41 (1H, s, ArCH), 7.51 (1H, s, ArCH), 7.56 (1H, d, J = 16.0 Hz, CH), 7.62 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 34.0, 34.7, 41.3, 55.2, 80.7, 114.1, 121.1, 121.9, 123.7, 128.3, 129.7, 130.6, 130.7, 133.7, 135.0, 135.5, 142.6, 143.1, 158.3, 166.0, 167.1.
tert-butyl (E)-3-(3-((4-methoxybenzyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylate (45d): Following the procedure as described for 45a the title compound was obtained as a transparent oil (30 mg, 0.06 mmol, 76%).
Rf: 0.17 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.73 (3H, s, CH3), 1.77 (3H, s, CH3), 3.39 (2H, d, J = 7.2 Hz, CH2), 3.82 (3H, s, CH3), 4.59 (2H, d, J = 5.5 Hz, CH2), 5.29 (1H, t, J = 8.2 Hz), 6.40 (1H, d, J = 15.9 Hz, CH), 6.91 (2H, d, J = 8.6 Hz, ArCH), 7.30 (2H, d, J = 8.5 Hz, ArCH), 7.43 (1H, s, ArCH), 7.57 (1H, d, J = 16.0 Hz, CH), 7.60 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 34.0, 43.7, 55.3, 80.6, 114.2, 121.2, 121.9, 123.7, 128.4, 129.3, 130.1, 130.7, 133.7, 135.1, 135.2, 142.6, 143.1, 159.1, 165.9, 166.9.
tert-butyl (E)-3-(3-(3-methylbut-2-en-l-yl)-5-((4-(trifluoromethoxy)benzyl)carbamoyl)phenyl)acty (45e): Following the procedure as described for 45a the title compound was obtained as a transparent oil (140 mg, 0.3 mmol, 53%).
Rf: 0.25 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.73 (3H, s, CH3), 1.77 (3H, s, CH3), 3.39 (2H, d, J = 7.2 Hz, CH2), 3.82 (3H, s, CH3), 4.59 (2H, d, J = 5.5 Hz, CH2), 5.29 (1H, t, J = 8.2 Hz), 6.40 (1H, d, J = 15.9 Hz, CH), 6.91 (2H, d, J = 8.6 Hz, ArCH), 7.30 (2H, d, J = 8.5 Hz, ArCH), 7.43 (1H, s, ArCH), 7.57 (1H, d, J = 16.0 Hz, CH), 7.60 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 34.0, 43.7, 55.3, 80.6, 114.2, 121.2, 121.9, 123.7, 128.4, 129.3, 130.1, 130.7, 133.7, 135.1, 135.2, 142.6, 143.1, 159.1, 165.9, 166.9.
tert-butyl (E)-3-(3-(3-methylbut-2-en-l-yl)-5-((3-(trifluoromethoxy)benzyl)carbamoyl)phenyl)acrylate (45f): Following the procedure as described for 45a the title compound was obtained as a transparent oil (60 mg, 0.1 mmol, 55%).
Rf: 0.27 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.53 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.76 (3H, s, CH3), 3.32 (2H, d, J = 7.8 Hz, CH2), 4.66 (2H, d, J = 5.8 Hz, CH2), 5.27 (1H, t, J = 8.6 Hz), 6.40 (1H, d, J = 15.9 Hz, CH), 7.14 - 7.21 (2H, m, ArCH), 7.28 - 7.31 (1H, m, ArCH), 7.37 (1H, d, J = 7.9 Hz, ArCH), 7.43 (1H, s, ArCH), 7.55 (1H, d, J = 16.0 Hz, CH), 7.62 (1H, s, ArCH), 7.73 (1H, s, ArCH) 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 34.0, 43.4, 80.7, 119.9, 120.2, 121.3, 121.9, 123.8, 126.0, 128.5, 130.1, 130.9, 133.8, 134.8, 135.2, 140.6, 142.5, 143.2, 149.4, 149.5, 166.0, 167.1.
tert-buty 1 (E)-3-(3 -((4-methy lbenzy l)carbamoy l)-5 -(3 -methy lbut-2-en- 1 -y l)pheny 1) aery late (45g) : Following the procedure as described for 45a the title compound was obtained as a transparent oil (70 mg, 0.16 mmol, 72%).
Rf: 0.35 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.54 (9H, s, C(CH3)3), 1.72 (3H, s, CH3), 1.76 (3H, s, CH3), 2.36 (3H, s, CH3), 3.37 (2H, d, J = 7.2 Hz, CH2), 4.60 (2H, d, J = 5.4 Hz, CH2), 5.28 (1H, t, J = 8.6 Hz), 6.40 (1H, d, J = 15.9 Hz, CH), 7.17 (2H, d, J = 7.8 Hz, ArCH), 7.26 (2H, d, J = 8.0 Hz, ArCH), 7.42 (1H, s, ArCH), 7.56 (1H, d, J = 16.0 Hz, CH), 7.61 (1H, s, ArCH), 7.72 (1H, s, ArCH) 13C NMR (100 MHz; CDC13,): δ 17.9, 21.1, 25.7, 28.1, 34.0, 43.9, 80.6, 121.2, 122.0, 123.8, 128.0, 128.5, 129.4, 130.7, 133.7, 135.0, 135.1, 135.2, 137.3, 142.6, 143.1, 166.0, 166.9.
tert-butyl (E)-3-(3-((4-(dimethylamino)benzyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylate (45h): Following the procedure as described for 45a the title compound was obtained as a transparent oil (40 mg, 0.09 mmol, 44%).
Rf: 0.15 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 1.73 (3H, s, CH3), 1.77 (3H, s, CH3), 2.97 (6H, s, CH3), 3.39 (2H, d, J = 7.2 Hz, CH2), 4.55 (2H, d, J = 5.3 Hz, CH2), 5.29 (1H, t, J = 7.1 Hz), 6.40 (1H, d, J = 15.9 Hz, CH), 6.74 (2H, d, J = 8.6 Hz, ArCH), 7.26 (2H, d, J = 8.6 Hz, ArCH), 7.42 (1H, s, ArCH), 7.57 (1H, d, J = 16.0 Hz, CH), 7.59 (1H, s, ArCH), 7.70 (1H, s, ArCH) 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 34.0, 40.6, 43.9, 80.6, 112.7, 121.2, 122.0, 123.7, 125.4, 128.4, 129.2, 130.6, 133.6, 135.1, 135.4, 142.6, 143.1, 150.2, 166.0, 166.8.
tert-butyl (E)-3-(3-allyl-5-(phenethylcarbamoyl)phenyl)acrylate (46): Following the procedure as described for 45a the title compound was obtained using allyl boronic acid pinacol ester as a transparent oil (50 mg, 0.1 mmol, 49%).
Rf: 0.26 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, C(CH3)3), 2.96 (2H, t, J = 6.9 Hz, CH2), 2.43 (2H, d, J = 6.6 Hz, CH2), 3.73 (2H, q, J = 5.9 Hz, CH2), 5.09 - 5.15 (2H, m, CH2), 5.89 - 5.99 (1H, m, CH), 6.40 (1H, d, J = 15.9 Hz, CH), 7.25 - 7.30 (3H, m, ArCH), 7.34 - 7.38 (2H, m,
ArCH), 7.44 (1H, s, ArCH), 7.52 (1H, s, ArCH), 7.56 (1H, d, J = 16.0 Hz, CH), 7.63 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 35.6, 39.8, 41.7, 80.7, 116.9, 121.3, 126.6, 128.5, 128.7, 128.8, 129.6, 130.8, 135.2, 135.5, 136.2, 138.8, 141.3, 142.4, 165.9, 167.0.
tert-butyl (E)-3-(3'-(hydroxymethyl)-5-( henet^^ (47): Following the procedure as described for 45a the title compound was obtained using hydroxymethyl phenyl boronic acid pinacol ester as a transparent oil (50 mg, 0.1 mmol, 49%).
Rf: 0.65 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.56 (9H, s, C(CH3)3), 2.98 (2H, t, J = 6.8 Hz, CH2), 3.74 (2H, q, J = 6.1 Hz, CH2), 4.77 (2H, s, CH2), 6.43 (1H, d, J = 15.9 Hz, CH), 7.25 - 7.48 (8H, m, ArCH), 7.56 (1H, s, ArCH), 7.58 (1H, d, J = 15.9 Hz, CH), 7.73 (1H, s, ArCH), 7.75 (1H, s, ArCH), 7.87 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 28.1, 35.6, 41.2, 80.8, 121.7, 124.9, 125.6, 126.2, 126.6, 126.6, 127.0, 128.7, 128.8, 129.1, 135.5, 135.9, 138.8, 139.7, 141.8, 142.0, 142.2, 165.9, 167.0.
phenethy 1 (E)-3-(3 -(tert-butoxy)-3-oxoprop- 1 -en- 1 -y l)-5-(3 -methy lbut-2-en- 1 -y l)benzoate (48) : Following the procedure as described for 45a the title compound was synthesized from 44 as a transparent oil (67 mg, 0.1 mmol, 90%).
Rf: 0.57 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.57 (9H, s, C(CH3)3), 1.75 (3H, s, CH3), 1.79 (3H, s, CH3), 3.11 (2H, t, J = 6.9 Hz, CH2), 3.40 (2H, d, J = 7.3 Hz, CH2), 4.55 (2H, t, J = 6.9 Hz, CH2), 5.30 (1H, t, J = 7.3 Hz), 6.43 (1H, d, J = 15.9 Hz, CH), 7.28 - 7.38 (5H, m, ArCH), 7.49 (1H, s, ArCH), 7.60 (1H, d, J = 16.0 Hz, CH), 7.83 (1H, s, ArCH), 7.98 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 17.9, 25.7, 28.1, 28.2, 34.0, 35.2, 80.6, 121.2, 122.0, 126.4, 126.6, 128.5, 129.0, 130.8, 130.9, 132.2, 133.7, 135.0, 137.8, 142.6, 142.9, 166.0, 166.1.
(E)-3-(3-(3-methylbut-2-en-l-yl)-5-(phenethylcarbamoyl)phenyl)acrylic acid (49a): To a solution of (45a) (50 mg, 0.12 mmol) in dry toluene (10 mL) was added silica gel (5 mL) and the suspension was stirred at reflux overnight. The reaction was allowed to cool and the mixture was filtered after diluting with 20% MeOH in DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (DCM:MeOH = 20: 1) provided the title compound as a white solid (21.7 mg, 0.06 mmol, 50%).
Rf: 0.27 (DCM: MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 1.77 (3H, s, CH3), 1.78 (3H, s, CH3), 2.94 (2H, t, J = 7.3 Hz, CH2), 3.43 (2H, d, J = 7.4 Hz, CH2), 3.59 - 3.64 (2H, m, CH2), 5.35 (1H, t, J = 7.4 Hz), 6.53 (1H, d, J = 16.0 Hz, CH), 7.20 - 7.32 (5H, m, ArCH), 7.54 (1H, s, ArCH), 7.63 (1H, s, ArCH), 7.68 (1H, d, J = 16.0 Hz, CH), 7.80 (1H, s, ArCH). 13C NMR (100 MHz; CDC13,): δ 16.5, 24.5, 33.4, 35.0, 41.2, 119.1, 122.0, 123.7, 125.9, 128.1, 128.5, 128.6, 130.6, 133.0, 134.8, 135.2, 139.1, 143.1, 143.9, 168.3, 168.6. HRMS-ESI: (m/z) calculated for C23H25N03 , 386.1727 [M+Na] +; found, 386.1736.
(E)-3-(3-((4-fluorophenethyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (49b): Following the procedure as described for 49a the title compound was obtained as a white solid (20 mg, 0.05 mmol, 41%).
Rf: 0.22 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.77 (3H, s, CH3), 1.79 (3H, s, CH3), 2.92 (2H, t, J = 7.3 Hz, CH2), 3.44 (2H, d, J = 7.2 Hz, CH2), 3.58 - 3.62 (2H, m, CH2), 5.35 (1H, t, J = 7.2 Hz, CH), 6.54 (1H, d, J = 16.0 Hz, CH), 7.03 (2H, t, J = 8.9 Hz, ArCH). 7.28 (2H, q, Jl = 5.4 Hz, J2 = 3.1 Hz, ArCH), 7.56 (1H, s, ArCH), 7.63 (1H, s, ArCH), 7.68 (1H, d, J = 15.9 Hz, CH), 7.81 (1H, s, ArCH). 13C NMR (100 MHz; CD30D): δ 16.5, 24.5, 33.4, 34.2, 41.2, 110.7, 114.5, 114.7, 122.0, 123.7, 128.5, 130.1, 130.1, 130.6, 133.1, 134.8, 135.1, 143.2, 147.0, 160.4, 168.3. HRMS-ESI: (m/z) calculated for C23H24N03F, 382.1813 [M+H] +; found, 382.1804.
(E)-3-(3-((4-methoxyphenethyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (49c): Following the procedure as described for 49a the title compound was obtained as a white solid (26 mg, 0.06 mmol, 30%).
Rf: 0.25 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.77 (3H, s, CH3), 1.79 (3H, s, CH3), 2.87 (2H, t, J = 7.4 Hz, CH2), 3.44 (2H, d, J = 7.2 Hz, CH2), 3.57 (2H, q, Jl = 7.3 Hz, J2 = 6.3 Hz, CH2), 3.77 (3H, s, CH3), 5.35 (1H, t, J = 7.3 Hz, CH), 6.54 (1H, d, J = 15.9 Hz, CH), 6.87 (2H, d, J = 8.6 Hz, ArCH), 7.18 (2H, d, J = 8.6 Hz, ArCH), 7.55 (1H, s, ArCH), 7.63 (1H, s, ArCH), 7.67 (1H, d, J = 16.0 Hz, CH), 7.80 (1H, s, ArCH), 8.61 (1H, t, J = 5.5 Hz, -NH). 13C NMR (100 MHz; CD30D): δ 16.5, 24.5, 33.4, 34.2, 41.4, 54.2, 110.7, 113.4, 122.0, 123.6, 128.5, 129.4, 130.6, 131.0, 133.0, 134.8, 135.2, 135.3, 143.1, 149.9, 158.3, 168.3.
(E)-3-(3-((4-methoxybenzyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (49d): Following the procedure as described for 49a the title compound was obtained as a white solid (12 mg, 0.03 mmol, 45%).
Rf: 0.25 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.76 (3H, s, CH3), 1.78 (3H, s, CH3), 3.44 (2H, d, J = 7.3 Hz, CH2), 3.78 (3H, s, CH3), 4.52 (2H, d, J = 4.1 Hz, CH2), 5.35 (1H, t, J = 7.3 Hz, CH), 6.55 (1H, d, J = 16.0 Hz, CH), 6.90 (2H, d, J = 8.7 Hz, ArCH), 7.29 (2H, d, J = 8.7 Hz, ArCH), 7.55 (1H, s, ArCH), 7.68 (1H, d, J = 16.0 Hz, CH), 7.71 (1H, s, ArCH), 7.89 (1H, s, ArCH), 9.00 (1H, t, J = 5.6 Hz, -NH). 13C NMR (100 MHz; CD30D): δ 16.5, 24.4, 33.4, 42.6, 54.2, 110.7, 113.5, 119.4, 122.0, 123.8, 128.5, 130.6, 130.7, 133.0, 134.9, 135.1, 143.2, 143.7, 159.0, 168.0. HRMS-ESI: (m/z) calculated for C23H25N04, 402.1676 [M+Na] +; found, 402.1669.
(E)-3-(3-(3-methylbut-2-en-l-yl)-5-((4-(trifluoromethoxy)benzyl)carbamoyl)phenyl)acrylic acid (49e): Following the procedure as described for 49a the title compound was obtained as a white solid (39 mg, 0.09 mmol, 75%).
Rf: 0.25 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.77 (3H, s, CH3), 1.78 (3H, s, CH3), 3.45 (2H, d, J = 7.2 Hz, CH2), 4.61 (2H, s, CH2), 5.36 (1H, t, J = 7.3 Hz, CH), 6.56 (1H, d, J = 15.9 Hz, CH), 7.26 (2H, d, J = 7.9 Hz, ArCH), 7.47 (2H, d, J = 8.7 Hz, ArCH), 7.58 (1H, s, ArCH), 7.69 (1H, d, J = 16.0 Hz, CH), 7.73 (1H, s, ArCH), 7.92 (1H, s, ArCH). NMR (100 MHz; CD30D): δ 16.5, 24.4, 33.4, 42.4, 119.3, 119.3, 120.7, 122.0, 123.8, 128.7, 128.8, 130.7, 133.1, 134.8, 134.9, 138.1, 143.3, 143.8, 148.1, 168.2. HRMS-ESI: (m/z) calculated for C23H22N04F3, 456.1393 [M+Na] +; found, 456.1403.
(E)-3-(3-(3-methylbut-2-en-l-yl)-5-((3-(trifluoromethoxy)benzyl)carbamoyl)phenyl)acrylic acid (49f): Following the procedure as described for 49a the title compound was obtained as a white solid (18 mg, 0.04 mmol, 33%).
Rf: 0.21 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.77 (3H, s, CH3), 1.78 (3H, s, CH3), 2.87 (2H, t, J = 7.4 Hz, CH2), 3.45 (2H, d, J = 7.5 Hz, CH2), 4.63 (2H, d, Jl = 4.0 Hz, CH2), 5.36 (1H, t, J = 7.3 Hz, CH), 6.56 (1H, d, J = 16.0 Hz, CH), 7.18 (2H, d, J = 8.1 Hz, ArCH), 7.29 (1H, s, ArCH), 7.37 - 7.47 (2H, m, ArCH), 7.58 (1H, s, ArCH), 7.73 (1H, s, ArCH), 7.70 (1H, d, J = 16.0 Hz, CH), 7.92 (1H, s, ArCH). NMR (100 MHz; CD30D): δ 16.5, 24.4, 33.4, 42.6, 119.2, 119.6, 122.0, 123.8, 125.8, 128.6, 129.8, 130.8, 133.1, 134.8, 134.9, 141.6, 143.3, 143.9, 149.3, 150.1, 168.2, 168.6. HRMS-ESI: (m/z) calculated for C23H22N04F3, 456.1393 [M+Na] +; found, 456.1387.
(E)-3 -(3 -((4-methy lbenzy l)carbamoy l)-5 -(3 -methy lbut-2-en- 1 -y l)pheny l)acry lie acid (49g) : Following the procedure as described for 49a the title compound was obtained as a white solid (18 mg, 0.05 mmol, 30%).
Rf: 0.20 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.76 (3H, s, CH3), 1.77 (3H, s, CH3), 2.31 (3H, s, CH3), 3.43 (2H, d, J = 7.3 Hz, CH2), 4.54 (2H, d, J = 5.4 Hz, CH2), 5.35 (1H, t, J = 7.3 Hz, CH), 6.54 (1H, d, J = 16.0 Hz, CH), 7.15 (2H, d, J = 7.8 Hz, ArCH), 7.24 (2H, d, J = 8.0 Hz, ArCH), 7.55 (1H, s, ArCH), 7.68 (1H, d, J = 16.0 Hz, CH), 7.71 (1H, s, ArCH), 7.90 (1H, s, ArCH), 9.03 (1H, t, J = 5.7 Hz, -NH). NMR (100 MHz; CD30D): δ 16.5, 19.7, 24.5, 33.4, 42.9, 119.1, 122.0, 123.8, 127.1, 128.7, 128.7, 130.6, 133.0, 134.8, 135.0, 135.6, 136.5, 143.2, 143.9, 168.0, 168.7. HRMS-ESI: (m/z) calculated for C23H25N03, 364.1907 [M+H] +; found, 364.1899.
(E)-3-(3-((4-(dimethylamino)benzyl)carbamoyl)-5-(3-methylbut-2-en-l-yl)phenyl)acrylic acid (49h): Following the procedure as described for 49a the title compound was obtained as a white solid (15 mg, 0.04 mmol, 42%).
Rf: 0.27 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CD30D): δ 1.77 (3H, s, CH3), 1.78 (3H, s, CH3), 2.91 (6H, s, N(CH3)2), 3.44 (2H, d, J = 7.2 Hz, CH2), 4.48 (2H, d, J = 5.6 Hz, CH2), 5.36 (1H, t, J = 7.3 Hz, CH), 6.55 (1H, d, J = 16.0 Hz, CH), 6.78 (2H, d, J = 8.7 Hz, ArCH), 7.23 (2H, d, J = 8.7 Hz, ArCH), 7.55 (1H, s, ArCH), 7.67 (1H, d, J = 16.0 Hz, CH), 7.71 (1H, s, ArCH), 7.89 (1H, s, ArCH), 8.92 (1H, t, J = 5.9 Hz, -NH). NMR (100 MHz; CD30D): δ 16.5, 24.4, 33.4, 39.7, 42.8, 112.9, 119.4, 122.1, 123.7, 126.9, 128.2, 128.7, 130.5, 133.0, 134.9, 135.2, 143.1, 143.7, 150.2, 168.0, 168.9. HRMS-ESI: (m/z) calculated for C24H28N203, 393.2173 [M+H] +; found, 393.2169.
(E)-3-(3-allyl-5-(phenethylcarbamoyl)phenyl)acrylic acid (50): Following the procedure as described for 49a the title compound was synthesized using 46 as a white solid (11 mg, 0.03 mmol, 27%).
Rf: 0.26 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 2.94 (2H, t, J = 7.1 Hz, CH2), 3.48 (2H, d, J = 6.7 Hz, CH2), 3.60 (2H, t, J = 7.8 Hz, CH2), 5.11 - 5.17 (2H, m, CH2), 5.96 - 6.06 (1H, m, CH),
6.55 (1H, d, J = 16.0 Hz, CH), 7.20 - 7.32 (5H, m, ArCH), 7.59 (1H, s, ArCH), 7.66 (1H, s, ArCH), 7.70 (1H, s, ArCH), 7.82 (1H, d, J = 16.0 Hz, CH). 13C NMR (100 MHz; CDC13,): δ 35.0, 39.3, 41.2, 115.5, 124.0, 125.9, 127.6, 128.1, 128.5, 128.8, 129.4, 130.8, 134.9, 135.3, 136.5, 139.1, 141.5, 143.6, 168.1. HRMS-ESI: (m/z) calculated for C21H21N03, 336.1594 [M+H] +; found, 336.1595.
(E)-3-(3'-(hydroxymethyl)-5-( henethylcarbamoyl)-[l,r-biphenyl]-3-yl)acrylic acid (51): Following the procedure as described for 49a the title compound was synthesized using 47 as a white solid (14 mg, 0.03 mmol, 35%).
Rf: 0.19 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CDC13): δ 2.97 (2H, t, J = 7.1 Hz, CH2), 3.65 (2H, t, J = 7.5 Hz, CH2), 4.72 (2H, s, CH2), 6.66 (1H, d, J = 16.0 Hz, CH), 7.22 - 7.50 (7H, m, ArCH), 7.62 (1H, d, J = 7.5 Hz, ArCH), 7.71 (1H, s, ArCH), 7.75 (1H, d, J = 15.9 Hz, CH), 7.99 (2H, d, J = 4.8 Hz, ArCH), 8.06 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 35.0, 41.3, 63.6, 124.7, 124.8, 125.2, 125.6, 126.0, 126.3, 126.9, 128.1, 128.5, 128.7, 129.1, 129.6, 135.6, 135.7, 139.1, 139.5, 142.1, 142.2, 168.0.
(E)-3-(3-(3-methylbut-2-en-l-yl)-5-(phenethoxycarbonyl)phenyl)acrylic acid (52): Following the procedure as described for 49a the title compound was synthesized using 48 as a white solid (30 mg, 0.08 mmol, 51%).
1H NMR (400 MHz; CDC13): δ 1.76 (3H, s, CH3), 1.80 (3H, s, CH3), 3.12 (2H, t, J = 7.0 Hz, CH2), 3.42 (2H, d, J = 7.2 Hz, CH2), 4.57 (2H, t, J = 6.9 Hz, CH2), 5.32 (1H, t, J = 7.3 Hz, CH), 6.51 (1H, d, J = 15.9 Hz, CH), 7.29 - 7.38 (5H, m, ArCH), 7.53 (1H, s, ArCH), 7.80 (1H, d, J = 15.9 Hz, CH), 7.87 (1H, s, ArCH), 8.02 (1H, s, ArCH). 13C NMR (100 MHz; CDC13): δ 17.9, 25.7, 33.9, 35.2, 65.7, 118.1, 121.8, 126.7, 128.5, 128.9, 129.0, 131.1, 131.5, 132.5, 133.9, 134.3, 137.7, 143.1, 146.0, 166.0, 171.3. HRMS-ESI: (m/z) calculated for C23H2404, 387.1567 [M+Na] +; found, 387.1573.
benzyl 3-(benzyloxy)-5-bromobenzoate (54a): To a solution of 3-bromo-5-hydroxy benzoic acid (200 mg, 1 mmol) in DIPEA (0.4 mL, 2.4 mmol) was added benzyl bromide (0.2 mL, 2.2 mmol) and the solution was heated at 150 oC overnight. The solution was allowed to cool and extracted with EtOAc, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1, 2: 1) provided the title compound as a transparent oil. (310 mg, 0.8 mmol, 80%).
Rf: 0.6 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): 5 5.11 (2H, s, CH2), 5.41 (2H, s, CH2), 7.34 - 7.51 (11H, m, ArCH), 7.68 (1H, q, J = 1.3 Hz, ArCH), 7.88 (1H, t, J = 1.4 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 67.2, 70.5, 114.6, 122.8, 123.0, 125.2, 127.6, 128.3, 128.4, 128.7, 128.7, 128.8, 132.8, 135.6, 135.9, 159.3, 164.9.
3 -methoxy benzyl 3-bromo-5-((3-methoxybenzyl)oxy)benzoate (54b): Following the procedure as described for 54a the title compound was obtained as a transparent oil. (270 mg, 0.59 mmol, 51%).
1H NMR (400 MHz; CDC13): δ 3.84 (3H, s, CH3), 3.85 (3H, s, CH3), 5.07 (2H, s, CH2), 5.35 (2H, s, CH2), 6.90 - 7.05 (6H, m, ArCH), 7.30 - 7.36 (3H, m, ArCH), 7.63 (1H, q, J = 1.3 Hz, ArCH), 7.83 (1H, t, J = 1.4 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 55.2, 67.0, 70.3, 112.9, 113.8, 113.8, 113.8, 114.6, 119.7, 120.4, 122.7, 123.0, 125.2, 129.7, 129.7, 132.7, 137.1, 137.4, 159.3, 159.8, 159.9, 164.9.
3-(benzyloxy)-5-bromobenzoic acid (55a): To a solution of 54a in MeOH was added 1M KOH (10 mL) and refluxed for 4 h. The pH of the solution was adjusted to 3 and extracted with EtOAc, dried (Na2S04), filtered and concentrated to provide the title compound as a white soild. (250 mg, 0.8 mmol, 99%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 5.07 (2H, s, CH2), 7.29 - 7.43 (6H, m, ArCH), 7.57 (1H, q, J = 1.3 Hz, ArCH), 7.71 (1H, t, J = 1.4 Hz, ArCH). 13C NMR (100 MHz; CDC13): δ 70.0, 114.4, 122.1, 124.6, 127.2, 127.7, 128.2, 133.4, 136.3, 159.5, 166.5.
3-bromo-5-((3-methoxybenzyl)oxy)benzoic acid (55b): Following the procedure as described for 55a the title compound was obtained as a white soild. (160 mg, 0.47 mmol, 78%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 3.79 (3H, s, CH3), 5.09 (2H, s, CH2), 6.79 - 7.00 (4H, m, ArCH), 7.37 (1H, s, CH3), 7.58 (1H, s, CH3), 7.75 (1H, s, CH3). 13C NMR (100 MHz; CDC13): δ 63.7, 69.9, 114.4, 118.6, 119.3, 122.1, 122.1, 124.6, 128.9, 129.2, 133.6, 137.8, 142.9, 159.5, 166.5.
3-(benzyloxy)-5-bromo-N-phenethylbenzamide (57a): To a solution of 55a (250 mg, 0.8 mmol) in dry toluene (7 mL) was added SOC12 (220 μΐ., 3 mmol) and the mixture was refluxed overnight. The reaction vessel was cooled to room temperature and the solvent was evaporated in vacuo. The resultant brown oil was used further without purification. To a solution of the acid chloride in DCM (10 mL) was added DMAP (26 mg, 0.2 mmol) and the flask was purged with nitrogen. 2-phenylethylamine (250 μΐ., 2 mmol) and NEt3 (350 μΐ., 2.5 mmol) were added to the flask and the mixture was stirred at 70oC overnight. The reaction mixture was cooled to room temperature, diluted with DCM, washed with a saturated aqueous NaHC03, water and extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 10: 1, 4: 1) provided the title compound as a white solid ( 180 mg, 0.4 mmol, 50%).
Rf: 0.2 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.92 (2H, t, J = 7.0 Hz, CH2), 3.69 (2H, q, J = 6.0 Hz, CH2), 5.03 (2H, s, CH2), 7.23 - 7.42 (13H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 35.6, 41.3, 70.4, 112.4, 121.2, 122.3, 122.9, 126.6, 127.5, 128.3, 128.7, 128.8, 128.8, 135.9, 137.5, 138.7, 159.5, 166.0.
3-bromo-5-((3-methoxybenzyl)oxy)-N-phenethylbenzamide (57b): Following the procedure as described for 57a the title compound was obtained as a white soild. (130 mg, 0.3 mmol, 60%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 2.94 (2H, t, J = 7.0 Hz, CH2), 3.70 (2H, q, J = 6.0 Hz, CH2), 3.84 (3H, s, CH3), 5.04 (2H, s, CH2), 7.89 - 7.00 (2H, m, ArCH), 7.23 - 7.37 (10H, m, ArCH). 13C NMR (100 MHz; CDC13): δ 35.6, 41.3, 55.2, 70.3, 112.4, 112.9, 113.7, 119.6, 121.2, 122.2, 122.9, 126.6, 128.7, 128.8, 129.7, 137.4, 137.5, 138.7, 159.5, 159.8, 165.9.
tert-butyl (E)-3-(3-(benzyloxy)-5-(phenethylcarbamoyl)phenyl)acrylate (58a): To a solution of 57a (100 mg, 0.3 mmol) in dry DMF (1.5 mL), was added DABCO (4 mg, 0.03 mmol), Pd(OAc)2 (4 mg, 0.015 mmol) and the flask was purged with nitrogen. tert-Butylacrylate (60 μΐ., 0.4 mmol) and K2C03 (55 mg, 0.4 mmol) were added and the mixture was stirred at 120oC for 18 h. The reaction was allowed to cool and washed with saturated aqueous NH4C1, brine, extracted with DCM, dried (Na2S04), filtered and concentrated. Purification by column chromatography (Hexane:EtOAc = 9: 1, 4: 1, 2: 1) provided the title compound as a white solid. (30 mg, 0.06 mmol, 22%).
Rf: 0.21 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, (CH3)3), 2.96 (2H, t, J = 7.0 Hz, CH2), 3.73 (2H, q, J = 6.0 Hz, CH2), 5.11 (2H, s, CH2), 6.37 (1H, d, J = 15.9 Hz, CH), 7.21 - 7.46 (13H, m, ArCH), 7.52 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 28.1, 35.6, 41.1, 70.3, 80.8, 114.6, 117.2, 118.5, 121.7, 126.7, 127.5, 128.2, 128.6, 128.7, 128.8, 136.1, 136.3, 136.7, 142.2, 159.2, 165.8, 166.7.
tert-butyl (E)-3-(3-((3-methoxybenzyl)oxy)-5-(phenethylcarbamoyl)phenyl)acrylate (58b): Following the procedure as described for 58a the title compound was obtained as a white solid. (30 mg, 0.06 mmol, 20%).
Rf: 0.1 (Hexane:EtOAc, 4: 1). 1H NMR (400 MHz; CDC13): δ 1.55 (9H, s, (CH3)3), 2.96 (2H, t, J = 7.0 Hz, CH2), 3.72 (2H, q, J = 6.0 Hz, CH2), 3.84 (3H, s, CH3), 5.08 (2H, s, CH2), 6.37 (1H, d, J = 15.9 Hz, CH), 7.20 - 7.38 (12H, m, ArCH), 7.52 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; CDC13): δ 28.1, 35.6, 41.1, 55.2, 70.1, 80.8, 112.9, 113.7, 114.7, 117.1, 118.5, 119.6, 121.7, 126.7, 128.7, 128.7, 128.8, 129.7, 136.3, 136.7, 137.7, 138.7, 142.2, 159.8, 165.8, 166.7.
(E)-3-(3-(benzyloxy)-5-(phenethylcarbamoyl)phenyl)acrylic acid (59a): Following the general procedure B the title compound was obtained as a white solid (8 mg, 0.02 mmol, 33%).
Rf: 0.3 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; MeOD): δ 2.94 (2H, t, J = 7.0 Hz, CH2), 3.61 (2H, t, J = 6.0 Hz, CH2), 5.18 (2H, s, CH2), 6.55 (1H, d, J = 15.9 Hz, CH), 7.20 - 7.49 (12H, m, ArCH), 7.58 (1H, s, ArCH), 7.65 (1H, d, J = 15.9 Hz, CH). 13C NMR (100 MHz; MeOD): δ 35.0, 41.3, 69.9, 115.0, 116.8, 118.8, 125.9, 127.3, 127.6, 128.1, 128.1, 128.5, 136.2, 136.4, 136.7, 139.1, 143.4, 159.3, 167.8.
(E)-3-(3-((3-methoxybenzyl)oxy)-5-(phenethylcarbamoyl)phenyl)acrylic acid (59b): Following the general procedure B the title compound was obtained as a white solid (10 mg, 0.02 mmol, 38%).
Rf: 0.2 (DCM:MeOH, 20: 1). 1H NMR (400 MHz; CDC13): δ 2.93 (2H, t, J = 7.0 Hz, CH2), 3.61 (2H, t, J = 6.0 Hz, CH2), 3.81 (3H, s, CH3), 5.15 (2H, s, CH2), 6.55 (1H, d, J = 15.9 Hz, CH), 6.89 (1H, dd, Jl = 2.1 Hz, J2 = 8.0 Hz, ArCH), 7.03 - 7.05 (2H, m, ArCH), 7.19 - 7.33 (6H, m, ArCH), 7.37 (1H, s, ArCH), 7.44 (1H, s, ArCH), 7.58 (1H, s, ArCH), 7.61 (1H, d, J = 16.0 Hz, CH). 13C NMR (100 MHz; CDC13): δ 35.0, 41.3, 54.2, 69.7, 112.6, 113.1, 114.9, 116.7, 118.7, 119.3, 125.9, 128.1, 128.5, 129.2, 136.3, 136.4, 138.2, 139.1, 142.7, 159.2, 159.9, 167.8.
It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method, kit, reagent, or composition of the invention, and vice versa. Furthermore, compositions of the invention can be used to achieve methods of the invention.
It will be understood that particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims.
All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.
As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
The term "or combinations thereof as used herein refers to all permutations and combinations of the listed items preceding the term. For example, "A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.
All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
Claims
1. An aldo-keto reductase family 1, member C3 (AKR1C3) inhibitor having one of the following structures:
derivatives thereof.
2. The inhibitor of claim 1, wherein the inhibitor is Class I or IA and
4. The inhibitor of claim 1, wherein the inhibitor is:
5. The inhibitor of claim 1, wherein the inhibitor is
; or , wherein R2 is:
, wherein Rl is
, and wherein R2 is:
8. The inhibitor of claim 1, wherein the inhibitor is:
, w ere n Rl s:
9. The inhibitor of claim 1, wherein the inhibitor is formulated into a therapeutic effective amount sufficient to reduce or eliminate cancer cells.
10. The inhibitor of claim 1, further comprising a chemotherapeutic agent, wherein the inhibitor and the chemotherapeutic have a synergistic effect against cancer cells.
11. The inhibitor of claim 1, further comprising providing the inhibitor in an amount that is therapeutically effective to inhibit or reduce cellular or tissue proliferation selected from prostate cancer, castration-resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
12. A method of modulating cellular or tissue proliferation comprising the steps of
providing a therapeutic effective amount of an inhibitor that is selective for the enzyme AKR1C3 over its isoforms wherein the compound has the formula:
derivatives thereof.
13. The method of claim 12, wherein the inhibitor is Class I or IA and
15. The method of claim 12, wherein the inhibitor i
16. The method of claim 12, wherein the inhibitor i
wherein R is:
18. The method of claim 12, wherein the inhibitor is:
, wherein R3 is ; or
and wherein R2 is:
, wherein Rl is:
20. The method of claim 12, wherein the inhibitor is formulated into a therapeutic effective amount sufficient to reduce or eliminate cancer cells.
21. The method of claim 12, further comprising a chemotherapeutic agent, wherein the inhibitor and the chemotherapeutic have a synergistic effect against cancer cells.
22. The method of claim 12, wherein the cellular or tissue proliferation comprises prostate cancer, castration-resistant prostate cancer, breast cancer, acute myeloid leukemia (AML), T-cell acute lymphoblastic leukemia (T-ALL), or a leukemia.
23. The method of claim 12, wherein the prostate cancer and/or castrate-resistant prostate cancer, breast cancer, leukemia is resistant to chemotherapeutic agents.
24. The method of claim 12, wherein the AKR1C3 inhibitor is synergistic with a chemotherapeutic.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201762458267P | 2017-02-13 | 2017-02-13 | |
US62/458,267 | 2017-02-13 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018148721A1 true WO2018148721A1 (en) | 2018-08-16 |
Family
ID=63107890
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2018/017986 WO2018148721A1 (en) | 2017-02-13 | 2018-02-13 | Highly selective akr1c3 inhibitors and methods of use thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2018148721A1 (en) |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2021110085A1 (en) * | 2019-12-03 | 2021-06-10 | 深圳艾欣达伟医药科技有限公司 | Method for associating with expression level of akr1c3 enzyme via content of prostaglandin, and use of screening for drug administration |
CN112979571A (en) * | 2021-02-24 | 2021-06-18 | 中国药科大学 | AKR1C3 selective inhibitor, preparation method and application thereof |
WO2024030627A1 (en) * | 2022-08-05 | 2024-02-08 | Board Of Regents Of The University Of Nebraska | Inhibitors of akr1c3 and methods of use |
WO2024035780A1 (en) * | 2022-08-09 | 2024-02-15 | Board Of Regents Of The University Of Nebraska | Akr1c3 targeted heterobifunctional small molecule proteolysis targeting chimeras |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4014920A (en) * | 1972-06-08 | 1977-03-29 | Ciba-Geigy Corporation | N-cinnamylcarbamic acid esters |
JP2004026760A (en) * | 2002-06-27 | 2004-01-29 | Api Co Ltd | Apoptosis-inducing agent of cancer cell, method for producing the same, anticancer agent containing the same as active ingredient, food preparation and cosmetic |
-
2018
- 2018-02-13 WO PCT/US2018/017986 patent/WO2018148721A1/en active Application Filing
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4014920A (en) * | 1972-06-08 | 1977-03-29 | Ciba-Geigy Corporation | N-cinnamylcarbamic acid esters |
JP2004026760A (en) * | 2002-06-27 | 2004-01-29 | Api Co Ltd | Apoptosis-inducing agent of cancer cell, method for producing the same, anticancer agent containing the same as active ingredient, food preparation and cosmetic |
Non-Patent Citations (6)
Title |
---|
ENDO, SATOSHI ET AL.: "Selective inhibition of human type-5 17P-hydroxysteroid dehydrogenase (AKR1C3) by baccharin, a component of Brazilian propolis", JOURNAL OF NATURAL PRODUCTS, vol. 75, 2012, pages 716 - 721, XP055560566 * |
ENDO, SATOSHI ET AL.: "Synthesis of non-prenyl analogues of baccharin as selective and potent inhibitors for aldo-keto reductase 1C3", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 22, no. 19, 2014, pages 5220 - 5233, XP029061988 * |
NINA, NELIDA ET AL.: "Chemical profiling and antioxidant activity of Bolivian propolis", JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE, vol. 96, no. 6, 2016, pages 2142 - 2153, XP055560577 * |
VERMA, KSHITIJ ET AL.: "Selective AKR1C3 inhibitors potentiate chemotherapeutic activity in multiple acute myeloid leukemia (AML) cell lines", ACS MEDICINAL CHEMISTRY LETTERS, vol. 7, no. 8, 2016, pages 774 - 779, XP055560571 * |
ZANG, TIANZHU ET AL.: "Screening baccharin analogs as selective inhibitors against type 5 17p-hydroxysteroid dehydrogenase (AKR1C3", CHEMICO-BIOLOGICAL INTERACTIONS, vol. 234, 2015, pages 339 - 348, XP055560575 * |
ZDERO, C. ET AL.: "Diterpene glycosides and other constituents from Argentinian Baccharis species", PHYTOCHEMISTRY, vol. 25, no. 12, 1986, pages 2841 - 2855, XP001100680 * |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2021110085A1 (en) * | 2019-12-03 | 2021-06-10 | 深圳艾欣达伟医药科技有限公司 | Method for associating with expression level of akr1c3 enzyme via content of prostaglandin, and use of screening for drug administration |
CN112979571A (en) * | 2021-02-24 | 2021-06-18 | 中国药科大学 | AKR1C3 selective inhibitor, preparation method and application thereof |
WO2024030627A1 (en) * | 2022-08-05 | 2024-02-08 | Board Of Regents Of The University Of Nebraska | Inhibitors of akr1c3 and methods of use |
WO2024035780A1 (en) * | 2022-08-09 | 2024-02-15 | Board Of Regents Of The University Of Nebraska | Akr1c3 targeted heterobifunctional small molecule proteolysis targeting chimeras |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2018148721A1 (en) | Highly selective akr1c3 inhibitors and methods of use thereof | |
Verma et al. | Potent and Highly Selective Aldo–Keto Reductase 1C3 (AKR1C3) Inhibitors Act as Chemotherapeutic Potentiators in Acute Myeloid Leukemia and T-Cell Acute Lymphoblastic Leukemia | |
Duan et al. | Design, synthesis, and biological evaluation of novel dual inhibitors targeting lysine specific demethylase 1 (LSD1) and histone deacetylases (HDAC) for treatment of gastric cancer | |
Zhou et al. | Design and synthesis of α-naphthoflavone chimera derivatives able to eliminate cytochrome P450 (CYP) 1B1-mediated drug resistance via targeted CYP1B1 degradation | |
US9757350B2 (en) | Small molecule inhibitors of replication protein A that also act synergistically with cisplatin | |
Romagnoli et al. | Design, synthesis and biological evaluation of novel vicinal diaryl-substituted 1H-Pyrazole analogues of combretastatin A-4 as highly potent tubulin polymerization inhibitors | |
EA011932B1 (en) | Substituted propenylpiperazine derivatives as novel inhibitors of histone deacetylase | |
Ghiano et al. | Antitubercular activity of 1, 2, 3-triazolyl fatty acid derivatives | |
Tang et al. | Discovery of a chiral fluorinated azetidin-2-one as a tubulin polymerisation inhibitor with potent antitumour efficacy | |
Kärkkäinen et al. | Structural properties for selective and efficient l-type amino acid transporter 1 (LAT1) mediated cellular uptake | |
WO2016109470A1 (en) | Small molecule stimulators of steroid receptor coactivator proteins and their use in the treatment of cancer | |
WO2018215070A1 (en) | Dual modulators of farnesoid x receptor and soluble epoxide hydrolase | |
Yeong et al. | Sirtuin inhibition and anti-cancer activities of ethyl 2-benzimidazole-5-carboxylate derivatives | |
CN115701991A (en) | Pyrazolylpropionamide compounds and their use for treating prostate cancer | |
Luan et al. | Anti-inflammatory activity of fluorine-substituted benzo [h] quinazoline-2-amine derivatives as NF-κB inhibitors | |
JP2017517521A (en) | Sulfonamide compounds and their use as STAT5 inhibitors | |
Wang et al. | Discovery of 4-methoxy-N-(1-naphthyl) benzenesulfonamide derivatives as small molecule dual-target inhibitors of tubulin and signal transducer and activator of transcription 3 (STAT3) based on ABT-751 | |
He et al. | Development of Biaryl-Containing Aldo-Keto Reductase 1C3 (AKR1C3) Inhibitors for Reversing AKR1C3-Mediated Drug Resistance in Cancer Treatment | |
AU2017341257B2 (en) | Hydroxamate triterpenoid derivatives | |
Duan et al. | Discovery of novel, potent, and orally bioavailable HDACs inhibitors with LSD1 inhibitory activity for the treatment of solid tumors | |
Pippione et al. | Structure-guided optimization of 3-hydroxybenzoisoxazole derivatives as inhibitors of Aldo-keto reductase 1C3 (AKR1C3) to target prostate cancer | |
KR20150041786A (en) | Combination therapy for the treatment of cancer and immunosuppression | |
Wagdy et al. | From EGFR kinase inhibitors to anti-inflammatory drugs: Optimization and biological evaluation of (4-(phenylamino) quinazolinyl)-phenylthiourea derivatives as novel NF-κB inhibitors | |
Xin et al. | Discovery of Novel ERα and Aromatase Dual-Targeting PROTAC Degraders to Overcome Endocrine-Resistant Breast Cancer | |
CN115297860B (en) | Novel N-aryl oxamic acids |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 18751218 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18751218 Country of ref document: EP Kind code of ref document: A1 |