NZ616465B2 - Combination of anti-clusterin oligonucleotide with androgen receptor antagonist for the treatment of prostate cancer - Google Patents
Combination of anti-clusterin oligonucleotide with androgen receptor antagonist for the treatment of prostate cancer Download PDFInfo
- Publication number
- NZ616465B2 NZ616465B2 NZ616465A NZ61646512A NZ616465B2 NZ 616465 B2 NZ616465 B2 NZ 616465B2 NZ 616465 A NZ616465 A NZ 616465A NZ 61646512 A NZ61646512 A NZ 61646512A NZ 616465 B2 NZ616465 B2 NZ 616465B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- arl
- oligonucleotide
- androgen
- custirsen
- prostate cancer
- Prior art date
Links
- 229920000272 Oligonucleotide Polymers 0.000 title claims abstract description 107
- 206010060862 Prostate cancer Diseases 0.000 title claims abstract description 95
- 239000003936 androgen receptor antagonist Substances 0.000 title claims abstract description 42
- 102000003780 Clusterin Human genes 0.000 claims abstract description 288
- 108090000197 Clusterin Proteins 0.000 claims abstract description 288
- 230000014509 gene expression Effects 0.000 claims abstract description 131
- 239000003098 androgen Substances 0.000 claims abstract description 66
- 239000003814 drug Substances 0.000 claims abstract description 42
- 239000011780 sodium chloride Substances 0.000 claims abstract description 19
- 150000003839 salts Chemical class 0.000 claims abstract description 17
- 238000004519 manufacturing process Methods 0.000 claims abstract description 10
- 108010080146 androgen receptors Proteins 0.000 claims description 271
- 102000001307 androgen receptors Human genes 0.000 claims description 271
- 210000004027 cells Anatomy 0.000 claims description 161
- RCFZILUHCNXXFY-DEDWCYLFSA-N Custirsen Chemical group N1([C@@H]2O[C@H](COP(O)(=S)O[C@H]3[C@H]([C@@H](O[C@@H]3COP(S)(=O)O[C@H]3[C@H]([C@@H](O[C@@H]3COP(O)(=S)O[C@H]3[C@H]([C@@H](O[C@@H]3COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=NC=NC(N)=C4N=C3)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C(N=C(N)C=C3)=O)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C(NC(=O)C(C)=C3)=O)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C(NC(=O)C(C)=C3)=O)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C(N=C(N)C=C3)=O)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C(NC(=O)C(C)=C3)=O)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=C(C(NC(N)=N4)=O)N=C3)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=NC=NC(N)=C4N=C3)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=C(C(NC(N)=N4)=O)N=C3)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=NC=NC(N)=C4N=C3)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C(N=C(N)C=C3)=O)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=C(C(NC(N)=N4)=O)N=C3)COP(O)(=S)O[C@@H]3[C@H](O[C@H](C3)N3C4=NC=NC(N)=C4N=C3)COP(O)(=S)O[C@H]3[C@H]([C@@H](O[C@@H]3COP(O)(=S)O[C@H]3[C@H]([C@@H](O[C@@H]3COP(O)(=S)O[C@H]3[C@H]([C@@H](O[C@@H]3COP(O)(=S)O[C@H]3[C@H]([C@@H](O[C@@H]3CO)N3C(N=C(N)C(C)=C3)=O)OCCOC)N3C4=NC=NC(N)=C4N=C3)OCCOC)N3C4=C(C(NC(N)=N4)=O)N=C3)OCCOC)N3C(N=C(N)C(C)=C3)=O)OCCOC)N3C(NC(=O)C(C)=C3)=O)OCCOC)N3C(N=C(N)C(C)=C3)=O)OCCOC)N3C4=NC=NC(N)=C4N=C3)OCCOC)[C@@H](O)[C@H]2OCCOC)C=C(C)C(=O)NC1=O RCFZILUHCNXXFY-DEDWCYLFSA-N 0.000 claims description 150
- 229950001605 CUSTIRSEN Drugs 0.000 claims description 149
- 239000000203 mixture Substances 0.000 claims description 139
- 230000001965 increased Effects 0.000 claims description 60
- 230000003042 antagnostic Effects 0.000 claims description 45
- 239000005557 antagonist Substances 0.000 claims description 42
- 229920002106 messenger RNA Polymers 0.000 claims description 39
- 108020004999 Messenger RNA Proteins 0.000 claims description 38
- 201000011510 cancer Diseases 0.000 claims description 35
- 108020000948 Antisense Oligonucleotides Proteins 0.000 claims description 25
- 239000000074 antisense oligonucleotide Substances 0.000 claims description 25
- 230000002708 enhancing Effects 0.000 claims description 22
- 241000282414 Homo sapiens Species 0.000 claims description 19
- 230000002103 transcriptional Effects 0.000 claims description 18
- 210000004881 tumor cells Anatomy 0.000 claims description 17
- 239000003795 chemical substances by application Substances 0.000 claims description 14
- 210000000805 Cytoplasm Anatomy 0.000 claims description 12
- 239000002773 nucleotide Substances 0.000 claims description 12
- 125000003729 nucleotide group Chemical group 0.000 claims description 12
- 241000229754 Iva xanthiifolia Species 0.000 claims description 8
- 239000002464 receptor antagonist Substances 0.000 claims description 5
- 238000002679 ablation Methods 0.000 claims description 4
- 230000035755 proliferation Effects 0.000 claims description 4
- 108009000611 Proteasome Degradation Proteins 0.000 claims description 3
- 239000003956 nonsteroidal anti androgen Substances 0.000 claims description 3
- 108091007153 p-Akt Proteins 0.000 claims description 2
- 230000014621 translational initiation Effects 0.000 claims description 2
- 101700079254 ARL1 Proteins 0.000 description 301
- 230000000694 effects Effects 0.000 description 76
- 230000002401 inhibitory effect Effects 0.000 description 65
- 238000003197 gene knockdown Methods 0.000 description 53
- 102100011339 KLK3 Human genes 0.000 description 46
- 101700058542 KLK3 Proteins 0.000 description 46
- 101710027788 NPEPPS Proteins 0.000 description 46
- 101710025567 PSAT1 Proteins 0.000 description 46
- 206010028980 Neoplasm Diseases 0.000 description 42
- 101700006234 AKT1 Proteins 0.000 description 40
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 32
- 229940079593 drugs Drugs 0.000 description 31
- 102000004169 proteins and genes Human genes 0.000 description 31
- 108090000623 proteins and genes Proteins 0.000 description 31
- 230000037361 pathway Effects 0.000 description 28
- 238000009097 single-agent therapy Methods 0.000 description 28
- 230000004083 survival Effects 0.000 description 25
- 238000001262 western blot Methods 0.000 description 25
- 230000015556 catabolic process Effects 0.000 description 24
- 230000004059 degradation Effects 0.000 description 24
- 238000006731 degradation reaction Methods 0.000 description 24
- 102100013207 ARIH1 Human genes 0.000 description 23
- 239000002609 media Substances 0.000 description 23
- 101700086988 AR1 Proteins 0.000 description 22
- 101700044276 ATR1 Proteins 0.000 description 22
- 101710043203 P23p89 Proteins 0.000 description 22
- 102100014187 TCF20 Human genes 0.000 description 22
- 101700038188 TCF20 Proteins 0.000 description 22
- 230000006907 apoptotic process Effects 0.000 description 22
- 230000002354 daily Effects 0.000 description 22
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 22
- 101710039033 pkbA Proteins 0.000 description 22
- 238000004166 bioassay Methods 0.000 description 20
- 230000010261 cell growth Effects 0.000 description 20
- 102100001248 AKT1 Human genes 0.000 description 18
- 206010059512 Apoptosis Diseases 0.000 description 18
- 230000025458 RNA interference Effects 0.000 description 18
- 230000000692 anti-sense Effects 0.000 description 18
- 238000002648 combination therapy Methods 0.000 description 18
- 239000012091 fetal bovine serum Substances 0.000 description 18
- 102100007457 FKBP4 Human genes 0.000 description 17
- 210000002966 Serum Anatomy 0.000 description 17
- 230000004913 activation Effects 0.000 description 17
- 238000006366 phosphorylation reaction Methods 0.000 description 17
- 230000000865 phosphorylative Effects 0.000 description 17
- 230000002829 reduced Effects 0.000 description 17
- 108010067247 tacrolimus binding protein 4 Proteins 0.000 description 17
- 238000002560 therapeutic procedure Methods 0.000 description 17
- 230000003247 decreasing Effects 0.000 description 16
- 230000001105 regulatory Effects 0.000 description 16
- 102000005431 Molecular Chaperones Human genes 0.000 description 15
- 108010006519 Molecular Chaperones Proteins 0.000 description 15
- 239000003112 inhibitor Substances 0.000 description 14
- 230000001404 mediated Effects 0.000 description 14
- -1 excipients Substances 0.000 description 13
- 101700024891 EPHB2 Proteins 0.000 description 13
- 101700083887 MAPK1 Proteins 0.000 description 13
- 102100016823 MAPK1 Human genes 0.000 description 13
- 102000004965 antibodies Human genes 0.000 description 13
- 108090001123 antibodies Proteins 0.000 description 13
- 101700042119 HSP83 Proteins 0.000 description 12
- 102100017052 HSP90AA1 Human genes 0.000 description 12
- 101710033238 HSP90AA1 Proteins 0.000 description 12
- 230000001419 dependent Effects 0.000 description 12
- 230000001939 inductive effect Effects 0.000 description 12
- 230000011664 signaling Effects 0.000 description 12
- 230000037327 stress response Effects 0.000 description 12
- CCCIJQPRIXGQOE-XWSJACJDSA-N Metribolone Chemical compound C1CC2=CC(=O)CCC2=C2[C@@H]1[C@@H]1CC[C@](C)(O)[C@@]1(C)C=C2 CCCIJQPRIXGQOE-XWSJACJDSA-N 0.000 description 11
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 11
- 238000004458 analytical method Methods 0.000 description 11
- 229960000997 bicalutamide Drugs 0.000 description 11
- LKJPYSCBVHEWIU-KRWDZBQOSA-N 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 11
- 230000003389 potentiating Effects 0.000 description 11
- 150000002500 ions Chemical class 0.000 description 10
- 210000004940 Nucleus Anatomy 0.000 description 9
- 210000002307 Prostate Anatomy 0.000 description 9
- 230000004663 cell proliferation Effects 0.000 description 9
- 230000023603 positive regulation of transcription initiation, DNA-dependent Effects 0.000 description 9
- 230000004044 response Effects 0.000 description 9
- 230000035916 transactivation Effects 0.000 description 9
- 108009000409 Autophagy Proteins 0.000 description 8
- 241000282412 Homo Species 0.000 description 8
- 239000005089 Luciferase Substances 0.000 description 8
- 101700067074 MAPK Proteins 0.000 description 8
- 101710041325 MAPKAPK2 Proteins 0.000 description 8
- 241000699660 Mus musculus Species 0.000 description 8
- 229920000776 Poly(Adenosine diphosphate-ribose) polymerase Polymers 0.000 description 8
- 238000010317 ablation therapy Methods 0.000 description 8
- 230000006877 autophagy Effects 0.000 description 8
- 238000002512 chemotherapy Methods 0.000 description 8
- 238000003776 cleavage reaction Methods 0.000 description 8
- 230000003993 interaction Effects 0.000 description 8
- 238000001990 intravenous administration Methods 0.000 description 8
- 101710024887 rl Proteins 0.000 description 8
- 230000019491 signal transduction Effects 0.000 description 8
- 101700045897 spk-1 Proteins 0.000 description 8
- 229940088597 Hormone Drugs 0.000 description 7
- 102000013530 TOR Serine-Threonine Kinases Human genes 0.000 description 7
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 7
- 230000027455 binding Effects 0.000 description 7
- 239000002552 dosage form Substances 0.000 description 7
- 231100000673 dose–response relationship Toxicity 0.000 description 7
- 239000005556 hormone Substances 0.000 description 7
- 230000001743 silencing Effects 0.000 description 7
- 230000001629 suppression Effects 0.000 description 7
- 230000001225 therapeutic Effects 0.000 description 7
- 230000003827 upregulation Effects 0.000 description 7
- 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 6
- 108009000009 Unfolded protein response Proteins 0.000 description 6
- 238000007792 addition Methods 0.000 description 6
- 239000000969 carrier Substances 0.000 description 6
- 239000000725 suspension Substances 0.000 description 6
- 238000010798 ubiquitination Methods 0.000 description 6
- 230000004906 unfolded protein response Effects 0.000 description 6
- YPHMISFOHDHNIV-FSZOTQKASA-N Cycloheximide Chemical compound C1[C@@H](C)C[C@H](C)C(=O)[C@@H]1[C@H](O)CC1CC(=O)NC(=O)C1 YPHMISFOHDHNIV-FSZOTQKASA-N 0.000 description 5
- 101700062887 HSPB1 Proteins 0.000 description 5
- 101700029228 Hsp27 Proteins 0.000 description 5
- 230000003044 adaptive Effects 0.000 description 5
- 230000001093 anti-cancer Effects 0.000 description 5
- 230000001640 apoptogenic Effects 0.000 description 5
- 239000003610 charcoal Substances 0.000 description 5
- 150000001875 compounds Chemical class 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 238000000684 flow cytometry Methods 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 230000002018 overexpression Effects 0.000 description 5
- 230000002987 rna-interference Effects 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 230000004614 tumor growth Effects 0.000 description 5
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N Docetaxel Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 4
- MKXKFYHWDHIYRV-UHFFFAOYSA-N Flutamide Chemical compound CC(C)C(=O)NC1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 MKXKFYHWDHIYRV-UHFFFAOYSA-N 0.000 description 4
- 108060001084 Luciferase family Proteins 0.000 description 4
- 229940035295 Ting Drugs 0.000 description 4
- 108090000848 Ubiquitin Proteins 0.000 description 4
- 102400000757 Ubiquitin Human genes 0.000 description 4
- 102000003970 Vinculin Human genes 0.000 description 4
- 108090000384 Vinculin Proteins 0.000 description 4
- CSCPPACGZOOCGX-UHFFFAOYSA-N acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 4
- 230000000996 additive Effects 0.000 description 4
- 230000033228 biological regulation Effects 0.000 description 4
- 230000022131 cell cycle Effects 0.000 description 4
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 4
- 201000010099 disease Diseases 0.000 description 4
- 238000009826 distribution Methods 0.000 description 4
- 229960003668 docetaxel Drugs 0.000 description 4
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 4
- 229960002074 flutamide Drugs 0.000 description 4
- 239000008187 granular material Substances 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 239000008297 liquid dosage form Substances 0.000 description 4
- 108010082117 matrigel Proteins 0.000 description 4
- OKKJLVBELUTLKV-UHFFFAOYSA-N methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 4
- 238000000034 method Methods 0.000 description 4
- 230000003285 pharmacodynamic Effects 0.000 description 4
- 230000017854 proteolysis Effects 0.000 description 4
- 230000035945 sensitivity Effects 0.000 description 4
- 231100000486 side effect Toxicity 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 239000002904 solvent Substances 0.000 description 4
- 230000014616 translation Effects 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- GZOSMCIZMLWJML-VJLLXTKPSA-N Abiraterone Chemical compound C([C@H]1[C@H]2[C@@H]([C@]3(CC[C@H](O)CC3=CC2)C)CC[C@@]11C)C=C1C1=CC=CN=C1 GZOSMCIZMLWJML-VJLLXTKPSA-N 0.000 description 3
- 102000010640 Androgen-Binding Protein Human genes 0.000 description 3
- 108010077825 Androgen-Binding Protein Proteins 0.000 description 3
- 206010061818 Disease progression Diseases 0.000 description 3
- 206010013710 Drug interaction Diseases 0.000 description 3
- 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 3
- 241000124008 Mammalia Species 0.000 description 3
- 206010063834 Oversensing Diseases 0.000 description 3
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 3
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 3
- 229960003604 Testosterone Drugs 0.000 description 3
- 229960000853 abiraterone Drugs 0.000 description 3
- 230000000903 blocking Effects 0.000 description 3
- 230000030833 cell death Effects 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 230000001086 cytosolic Effects 0.000 description 3
- 238000007405 data analysis Methods 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 231100000517 death Toxicity 0.000 description 3
- 239000003085 diluting agent Substances 0.000 description 3
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 230000013632 homeostatic process Effects 0.000 description 3
- 238000001114 immunoprecipitation Methods 0.000 description 3
- 238000001802 infusion Methods 0.000 description 3
- 238000002372 labelling Methods 0.000 description 3
- 231100000225 lethality Toxicity 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 238000002844 melting Methods 0.000 description 3
- 230000025308 nuclear transport Effects 0.000 description 3
- 239000008194 pharmaceutical composition Substances 0.000 description 3
- 230000000275 pharmacokinetic Effects 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 230000002035 prolonged Effects 0.000 description 3
- 108091007934 protein kinase B family Proteins 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 230000004654 survival pathway Effects 0.000 description 3
- 239000000375 suspending agent Substances 0.000 description 3
- 239000006188 syrup Substances 0.000 description 3
- 235000020357 syrup Nutrition 0.000 description 3
- 230000032258 transport Effects 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- HVYWMOMLDIMFJA-DPAQBDIFSA-N (3β)-Cholest-5-en-3-ol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 101700042601 ATF4 Proteins 0.000 description 2
- 102100000637 ATF4 Human genes 0.000 description 2
- 229920002676 Complementary DNA Polymers 0.000 description 2
- 108020004635 Complementary DNA Proteins 0.000 description 2
- BKRDNYHYNZJMPG-UHFFFAOYSA-A Custirsen sodium Chemical compound [Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].COCCOC1C(O)C(COP([O-])(=S)OC2C(C(OC2COP([O-])(=S)OC2C(C(OC2COP([O-])(=S)OC2C(C(OC2COP([O-])(=S)OC2C(OC(C2)N2C3=NC=NC(N)=C3N=C2)COP([O-])(=S)OC2C(OC(C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)OC2C(OC(C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)OC2C(OC(C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)OC2C(OC(C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)OC2C(OC(C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)OC2C(OC(C2)N2C3=C(C(NC(N)=N3)=O)N=C2)COP([O-])(=S)OC2C(OC(C2)N2C3=NC=NC(N)=C3N=C2)COP([O-])(=S)OC2C(OC(C2)N2C3=C(C(NC(N)=N3)=O)N=C2)COP([O-])(=S)OC2C(OC(C2)N2C3=NC=NC(N)=C3N=C2)COP([O-])(=S)OC2C(OC(C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)OC2C(OC(C2)N2C3=C(C(NC(N)=N3)=O)N=C2)COP([O-])(=S)OC2C(OC(C2)N2C3=NC=NC(N)=C3N=C2)COP([O-])(=S)OC2C(C(OC2COP([O-])(=S)OC2C(C(OC2COP([O-])(=S)OC2C(C(OC2COP([O-])(=S)OC2C(C(OC2CO)N2C(N=C(N)C(C)=C2)=O)OCCOC)N2C3=NC=NC(N)=C3N=C2)OCCOC)N2C3=C(C(NC(N)=N3)=O)N=C2)OCCOC)N2C(N=C(N)C(C)=C2)=O)OCCOC)N2C(NC(=O)C(C)=C2)=O)OCCOC)N2C(N=C(N)C(C)=C2)=O)OCCOC)N2C3=NC=NC(N)=C3N=C2)OCCOC)OC1N1C=C(C)C(=O)NC1=O BKRDNYHYNZJMPG-UHFFFAOYSA-A 0.000 description 2
- 241000668709 Dipterocarpus costatus Species 0.000 description 2
- 206010067671 Disease complication Diseases 0.000 description 2
- 238000001135 Friedman test Methods 0.000 description 2
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 2
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 2
- 101710006547 Hsp70Ab Proteins 0.000 description 2
- 241000764238 Isis Species 0.000 description 2
- 206010061289 Metastatic neoplasm Diseases 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- 206010061309 Neoplasm progression Diseases 0.000 description 2
- XJMOSONTPMZWPB-UHFFFAOYSA-M Propidium iodide Chemical compound [I-].[I-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CCC[N+](C)(CC)CC)=C1C1=CC=CC=C1 XJMOSONTPMZWPB-UHFFFAOYSA-M 0.000 description 2
- 239000012083 RIPA buffer Substances 0.000 description 2
- 101710010021 RPS6KA1 Proteins 0.000 description 2
- 102100012085 RPS6KA1 Human genes 0.000 description 2
- 241000242739 Renilla Species 0.000 description 2
- 101710009384 SRC Proteins 0.000 description 2
- 229920002684 Sepharose Polymers 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 150000001298 alcohols Chemical class 0.000 description 2
- 230000000172 allergic Effects 0.000 description 2
- 230000004075 alteration Effects 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 230000002280 anti-androgenic Effects 0.000 description 2
- 239000000051 antiandrogen Substances 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 201000008937 atopic dermatitis Diseases 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 238000010804 cDNA synthesis Methods 0.000 description 2
- 230000004611 cancer cell death Effects 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 231100000504 carcinogenesis Toxicity 0.000 description 2
- 101700018328 ccdB Proteins 0.000 description 2
- 230000001413 cellular Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 230000000295 complement Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 238000010192 crystallographic characterization Methods 0.000 description 2
- 210000004748 cultured cells Anatomy 0.000 description 2
- 230000001120 cytoprotect Effects 0.000 description 2
- 230000003013 cytotoxicity Effects 0.000 description 2
- 231100000135 cytotoxicity Toxicity 0.000 description 2
- 230000003111 delayed Effects 0.000 description 2
- 239000005547 deoxyribonucleotide Substances 0.000 description 2
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 239000000839 emulsion Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 229960004671 enzalutamide Drugs 0.000 description 2
- 150000002148 esters Chemical class 0.000 description 2
- 239000003925 fat Substances 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 235000003599 food sweetener Nutrition 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 238000005417 image-selected in vivo spectroscopy Methods 0.000 description 2
- 230000000977 initiatory Effects 0.000 description 2
- 230000031146 intracellular signal transduction Effects 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 238000010253 intravenous injection Methods 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000011068 load Methods 0.000 description 2
- 230000003340 mental Effects 0.000 description 2
- 230000002503 metabolic Effects 0.000 description 2
- 230000001394 metastastic Effects 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000006011 modification reaction Methods 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 239000006186 oral dosage form Substances 0.000 description 2
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 2
- 239000000825 pharmaceutical preparation Substances 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 230000002335 preservative Effects 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 230000001686 pro-survival Effects 0.000 description 2
- 230000001737 promoting Effects 0.000 description 2
- 239000003207 proteasome inhibitor Substances 0.000 description 2
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 230000000306 recurrent Effects 0.000 description 2
- 230000016914 response to endoplasmic reticulum stress Effects 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 229910001415 sodium ion Inorganic materials 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 230000004936 stimulating Effects 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 239000003765 sweetening agent Substances 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 230000002194 synthesizing Effects 0.000 description 2
- 239000002562 thickening agent Substances 0.000 description 2
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical class [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 2
- 230000036962 time dependent Effects 0.000 description 2
- 210000001519 tissues Anatomy 0.000 description 2
- 230000035897 transcription Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000001052 transient Effects 0.000 description 2
- DTYZSEFHBLCMTE-HDIZBSAMSA-N (2R)-2-[[(2S)-6-amino-2-[[(4R)-5-amino-4-[[(2S)-2-[2-[(3R,4R,5S,6R)-3-(carboxyamino)-5-[(2S,3R,4R,5S,6R)-4,5-dihydroxy-6-(hydroxymethyl)-3-(2-oxopropyl)oxan-2-yl]oxy-2-hydroxy-6-(hydroxymethyl)oxan-4-yl]oxypropanoylamino]propanoyl]amino]-5-oxopentanoyl]am Chemical compound OC(=O)[C@@H](C)NC(=O)[C@H](CCCCN)NC(=O)CC[C@H](C(N)=O)NC(=O)[C@H](C)NC(=O)C(C)O[C@@H]1[C@@H](NC(O)=O)C(O)O[C@H](CO)[C@H]1O[C@H]1[C@H](CC(C)=O)[C@@H](O)[C@H](O)[C@@H](CO)O1 DTYZSEFHBLCMTE-HDIZBSAMSA-N 0.000 description 1
- CUWODFFVMXJOKD-UVLQAERKSA-N (2S)-N-[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2R)-1-[[(2S)-1-[[(2S)-5-(diaminomethylideneamino)-1-[(2S)-2-(ethylcarbamoyl)pyrrolidin-1-yl]-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-3-[(2-methylpropan-2-yl)oxy]-1-oxopropan-2-yl]amino]-3-(4-hy 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 1
- VUFNLQXQSDUXKB-DOFZRALJSA-N 2-[4-[4-[bis(2-chloroethyl)amino]phenyl]butanoyloxy]ethyl (5Z,8Z,11Z,14Z)-icosa-5,8,11,14-tetraenoate Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(=O)OCCOC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 VUFNLQXQSDUXKB-DOFZRALJSA-N 0.000 description 1
- LRSASMSXMSNRBT-UHFFFAOYSA-N 5-Methylcytosine Chemical class CC1=CNC(=O)N=C1N LRSASMSXMSNRBT-UHFFFAOYSA-N 0.000 description 1
- 101700006583 AKT2 Proteins 0.000 description 1
- 101700004058 AKT3 Proteins 0.000 description 1
- 102100006725 AKT3 Human genes 0.000 description 1
- 229940030486 ANDROGENS Drugs 0.000 description 1
- 102100012826 ATF6 Human genes 0.000 description 1
- 101710031276 ATF6 Proteins 0.000 description 1
- 108060000696 ATG3 Proteins 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 210000004100 Adrenal Glands Anatomy 0.000 description 1
- 241000928106 Alain Species 0.000 description 1
- 240000000800 Allium ursinum Species 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- 102100013894 BCL2 Human genes 0.000 description 1
- 108060000885 BCL2 Proteins 0.000 description 1
- 210000000481 Breast Anatomy 0.000 description 1
- 108010037003 Buserelin Proteins 0.000 description 1
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 1
- 240000002804 Calluna vulgaris Species 0.000 description 1
- 235000007575 Calluna vulgaris Nutrition 0.000 description 1
- 240000005589 Calophyllum inophyllum Species 0.000 description 1
- 102000003952 Caspase 3 Human genes 0.000 description 1
- 108090000397 Caspase 3 Proteins 0.000 description 1
- 229940107161 Cholesterol Drugs 0.000 description 1
- 230000036881 Clu Effects 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N DATI Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- 108020001019 DNA Primers Proteins 0.000 description 1
- 239000003155 DNA primer Substances 0.000 description 1
- 102100013957 DNER Human genes 0.000 description 1
- 229940009976 Deoxycholate Drugs 0.000 description 1
- 101700057458 Drice Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 230000008341 ER-associated protein catabolic process Effects 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 240000003550 Eusideroxylon zwageri Species 0.000 description 1
- 239000004606 Fillers/Extenders Substances 0.000 description 1
- 102100000899 GNRH1 Human genes 0.000 description 1
- 102100016472 GP2 Human genes 0.000 description 1
- 210000001035 Gastrointestinal Tract Anatomy 0.000 description 1
- 210000004907 Glands Anatomy 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 108010084340 Gonadotropin-Releasing Hormone Proteins 0.000 description 1
- 101700061835 HSF1 Proteins 0.000 description 1
- 102100003632 HSF1 Human genes 0.000 description 1
- 101710002907 HSFA1A Proteins 0.000 description 1
- 102000005623 HSP27 Heat-Shock Proteins Human genes 0.000 description 1
- 108010045100 HSP27 Heat-Shock Proteins Proteins 0.000 description 1
- 101710006403 HSPA5 Proteins 0.000 description 1
- 102100015710 HSPA5 Human genes 0.000 description 1
- 102000003964 Histone deacetylases Human genes 0.000 description 1
- 108090000353 Histone deacetylases Proteins 0.000 description 1
- 241000282619 Hylobates lar Species 0.000 description 1
- 241000257303 Hymenoptera Species 0.000 description 1
- 229940102223 Injectable Solution Drugs 0.000 description 1
- RCINICONZNJXQF-MZXODVADSA-N Intaxel Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 1
- 102000004889 Interleukin-6 Human genes 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 101700073419 LC3 Proteins 0.000 description 1
- 102100012759 LRP2 Human genes 0.000 description 1
- 241000254158 Lampyridae Species 0.000 description 1
- 108010015372 Low Density Lipoprotein Receptor-Related Protein-2 Proteins 0.000 description 1
- 210000004072 Lung Anatomy 0.000 description 1
- 108040008097 MAP kinase activity proteins Proteins 0.000 description 1
- 102000019149 MAP kinase activity proteins Human genes 0.000 description 1
- 101710036017 MAP1LC3A Proteins 0.000 description 1
- 101700038382 MARK1 Proteins 0.000 description 1
- 102100000538 MARK1 Human genes 0.000 description 1
- 101710032250 MICAL1 Proteins 0.000 description 1
- 101700003240 MLE3 Proteins 0.000 description 1
- 206010025650 Malignant melanoma Diseases 0.000 description 1
- 229950003695 Metribolone Drugs 0.000 description 1
- XLXSAKCOAKORKW-KPKRHBJMSA-N 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)-1-oxopropan-2-yl] Chemical compound 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)C1NC(=O)CC1)C1=CC=C(O)C=C1 XLXSAKCOAKORKW-KPKRHBJMSA-N 0.000 description 1
- GWFRSQBGIGLMFD-BBKOCAQVSA-N N-[9-[(2R,3R,4S,5R)-5-[[bis(4-methoxyphenyl)-phenylmethoxy]methyl]-3,4-dihydroxyoxolan-2-yl]purin-6-yl]-2-phenoxyacetamide Chemical compound C1=CC(OC)=CC=C1C(C=1C=CC(OC)=CC=1)(C=1C=CC=CC=1)OC[C@@H]1[C@@H](O)[C@@H](O)[C@H](N2C3=NC=NC(NC(=O)COC=4C=CC=CC=4)=C3N=C2)O1 GWFRSQBGIGLMFD-BBKOCAQVSA-N 0.000 description 1
- 101700080605 NUC1 Proteins 0.000 description 1
- XWXYUMMDTVBTOU-UHFFFAOYSA-N Nilutamide Chemical compound O=C1C(C)(C)NC(=O)N1C1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 XWXYUMMDTVBTOU-UHFFFAOYSA-N 0.000 description 1
- 101710030713 PHLPP1 Proteins 0.000 description 1
- 102100002774 PHLPP1 Human genes 0.000 description 1
- 102100004316 PTGES3 Human genes 0.000 description 1
- 101710036259 PTGES3 Proteins 0.000 description 1
- 229960001592 Paclitaxel Drugs 0.000 description 1
- 229940067631 Phospholipids Drugs 0.000 description 1
- 108091000081 Phosphotransferases Proteins 0.000 description 1
- 102000012338 Poly(ADP-ribose) Polymerases Human genes 0.000 description 1
- 108010061844 Poly(ADP-ribose) Polymerases Proteins 0.000 description 1
- 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 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 102000001253 Protein Kinases Human genes 0.000 description 1
- 239000007759 RPMI Media 1640 Substances 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102000004278 Receptor protein-tyrosine kinases Human genes 0.000 description 1
- 108090000873 Receptor protein-tyrosine kinases Proteins 0.000 description 1
- 210000003660 Reticulum Anatomy 0.000 description 1
- 241000220010 Rhode Species 0.000 description 1
- 210000003705 Ribosomes Anatomy 0.000 description 1
- 102000001332 SRC Human genes 0.000 description 1
- 108020004459 Small Interfering RNA Proteins 0.000 description 1
- 229920001891 Small hairpin RNA Polymers 0.000 description 1
- 229920001985 Small interfering RNA Polymers 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 102000001854 Steroid 17-alpha-Hydroxylase Human genes 0.000 description 1
- 108010015330 Steroid 17-alpha-Hydroxylase Proteins 0.000 description 1
- 108010085012 Steroid Receptors Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 102100011827 TMPRSS2 Human genes 0.000 description 1
- 101710044694 TMPRSS2 Proteins 0.000 description 1
- 108010027179 Tacrolimus Binding Proteins Proteins 0.000 description 1
- 102000018679 Tacrolimus Binding Proteins Human genes 0.000 description 1
- 241000153282 Theope Species 0.000 description 1
- 230000036201 Tissue concentration Effects 0.000 description 1
- FPKOPBFLPLFWAD-UHFFFAOYSA-N Trinitrotoluene Chemical compound CC1=CC=C([N+]([O-])=O)C([N+]([O-])=O)=C1[N+]([O-])=O FPKOPBFLPLFWAD-UHFFFAOYSA-N 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Tris Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- HMNZFMSWFCAGGW-XPWSMXQVSA-N [3-[hydroxy(2-hydroxyethoxy)phosphoryl]oxy-2-[(E)-octadec-9-enoyl]oxypropyl] (E)-octadec-9-enoate Chemical compound CCCCCCCC\C=C\CCCCCCCC(=O)OCC(COP(O)(=O)OCCO)OC(=O)CCCCCCC\C=C\CCCCCCCC HMNZFMSWFCAGGW-XPWSMXQVSA-N 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000002424 anti-apoptotic Effects 0.000 description 1
- 230000000259 anti-tumor Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007172 antigens Proteins 0.000 description 1
- 102000038129 antigens Human genes 0.000 description 1
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 1
- 229920002847 antisense RNA Polymers 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 1
- 229960002719 buserelin Drugs 0.000 description 1
- 238000010805 cDNA synthesis kit Methods 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 230000004640 cellular pathway Effects 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 230000002596 correlated Effects 0.000 description 1
- 230000000875 corresponding Effects 0.000 description 1
- 230000001472 cytotoxic Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- KXGVEGMKQFWNSR-LLQZFEROSA-M deoxycholate Chemical compound C([C@H]1CC2)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC([O-])=O)C)[C@@]2(C)[C@@H](O)C1 KXGVEGMKQFWNSR-LLQZFEROSA-M 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000036267 drug metabolism Effects 0.000 description 1
- 102000017256 epidermal growth factor-activated receptor activity proteins Human genes 0.000 description 1
- 108040009258 epidermal growth factor-activated receptor activity proteins Proteins 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 235000019634 flavors 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
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 238000005194 fractionation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000002452 interceptive Effects 0.000 description 1
- 230000003834 intracellular Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 230000002601 intratumoral Effects 0.000 description 1
- 238000009114 investigational therapy Methods 0.000 description 1
- 230000003902 lesions Effects 0.000 description 1
- 230000000670 limiting Effects 0.000 description 1
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 230000002132 lysosomal Effects 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 231100000682 maximum tolerated dose Toxicity 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- XOBKSJJDNFUZPF-UHFFFAOYSA-N methoxyethyl Chemical group CCOC XOBKSJJDNFUZPF-UHFFFAOYSA-N 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 230000000051 modifying Effects 0.000 description 1
- 229960000060 monoclonal antibodies Drugs 0.000 description 1
- 102000005614 monoclonal antibodies Human genes 0.000 description 1
- 108010045030 monoclonal antibodies Proteins 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 230000017066 negative regulation of growth Effects 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 229960002653 nilutamide Drugs 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 230000003000 nontoxic Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 101700006494 nucA Proteins 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000002093 peripheral Effects 0.000 description 1
- 230000002085 persistent Effects 0.000 description 1
- 230000003094 perturbing Effects 0.000 description 1
- 239000008024 pharmaceutical diluent Substances 0.000 description 1
- 238000009522 phase III clinical trial Methods 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 150000008105 phosphatidylcholines Chemical class 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 102000003998 progesterone receptors Human genes 0.000 description 1
- 108090000468 progesterone receptors Proteins 0.000 description 1
- 230000026938 proteasomal ubiquitin-dependent protein catabolic process Effects 0.000 description 1
- 230000004845 protein aggregation Effects 0.000 description 1
- 238000000751 protein extraction Methods 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 230000002685 pulmonary Effects 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 238000003762 quantitative reverse transcription PCR Methods 0.000 description 1
- 239000012132 radioimmunoprecipitation assay buffer Substances 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 230000000268 renotropic Effects 0.000 description 1
- 230000000754 repressing Effects 0.000 description 1
- 230000037195 reproductive physiology Effects 0.000 description 1
- 230000000630 rising Effects 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- REYJJPSVUYRZGE-UHFFFAOYSA-N stearylamine Chemical compound CCCCCCCCCCCCCCCCCCN REYJJPSVUYRZGE-UHFFFAOYSA-N 0.000 description 1
- 102000005969 steroid hormone receptors Human genes 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 125000004434 sulfur atoms Chemical group 0.000 description 1
- 230000000153 supplemental Effects 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 229930003347 taxol Natural products 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 230000000699 topical Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 102000003995 transcription factors Human genes 0.000 description 1
- 108090000464 transcription factors Proteins 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 238000004642 transportation engineering Methods 0.000 description 1
- 230000004565 tumor cell growth Effects 0.000 description 1
- 239000002691 unilamellar liposome Substances 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000003442 weekly Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/41—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
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
-
- 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
- A61P33/00—Antiparasitic agents
- A61P33/14—Ectoparasiticides, e.g. scabicides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/24—Drugs for disorders of the endocrine system of the sex hormones
- A61P5/28—Antiandrogens
Abstract
Discloses use of i) an oligonucleotide which reduces clusterin expression and ii) an androgen receptor antagonist having the structure as disclosed in the complete specification, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a mammalian subject afflicted with prostate cancer. Also discloses an oligonucleotide which reduces clusterin expression and an androgen receptor antagonist, in the manufacture of a medicament for the treatment of a mammalian subject afflicted with androgen-independent prostate cancer. ted with prostate cancer. Also discloses an oligonucleotide which reduces clusterin expression and an androgen receptor antagonist, in the manufacture of a medicament for the treatment of a mammalian subject afflicted with androgen-independent prostate cancer.
Description
COMBINATION OF ANTI—CLUSTERIN OLIGONUCLEOTIDE WITH ANDROGEN
RECEPTOR ANTAGONIST FOR THE TREATMENT OF PROSTATE CANCER
This application claims priority of U.S. ional
Application Nos. 61/452,583, filed March 14, 2011, 61/453,309,
filed March 16, 2011, 61/453,885, filed March 17, 2011, and
61/493,336, filed. June 3, 2011, the contents of which. are
hereby incorporated by reference.
Throughout this application, various publications are
nced, including referenced in parenthesis. Full
citations for publications referenced in parenthesis may be
found listed in alphabetical order at the end of the
specification immediately preceding the claims. The
disclosures of all nced publications in their entireties
are hereby incorporated by reference into this application in
order to more fully describe the state of the art to which
this invention pertains.
Field of the Invention
The subject invention relates to combination therapy for
treating prostate cancer.
Background of the ion
Prostate cancer is the most common cancer that affects men,
and the second leading cause of cancer deaths in men in the
Western world. Because prostate cancer is an androgen
ive tumor, androgen. withdrawal, for example via
castration, is utilized in some eutic regimens for
patients with. advanced. prostate . Androgen. withdrawal
leads to extensive apoptosis in the prostate tumor, and hence
to a regression of the disease. However, castration—induced
apoptosis is not complete, and a progression of surviving
tumor cells to androgen—independence ultimately occurs. This
progression is the main le to improving survival and
quality of life, and therapies e of treating prostate
cancer both before and after the progression to androgen
independence are needed.
It has been observed that numerous proteins are expressed in
increased s by prostate tumor cells ing androgen
withdrawal. At least some of these proteins are assumed to be
associated. with. the apoptotic cell death. which. is observed
upon androgen withdrawal. (Raffo et al., 1995; Krajewska et
al., 1996; McDonnell et al., 1992). The functions of many of
the proteins, however, is not completely understood. Clusterin
(also known as ed glycoprotein—2 (SGP—2) or TRPM—Z) is
within this latter category.
Clusterin
Clusterin is a otective chaperone protein that promotes
cell survival and confers broad-spectrum resistance to cancer
treatments (Chi et al. 2005). in Sensibar et al., Cancer
Research 55: 2431—2437, 1995, the authors reported on LNCaP
cells transfected with a gene encoding Clusterin, and watched
to see if expression of this protein altered the effects of
tumor is factor a (TNFd), to which LNCaP cells are very
sensitive. Treatment of the transfected LNCaP cells with TNFd
was shown to result in a transient se in Clusterin
levels for a period. 0" a 'ew hours, but these levels had
dissipated by the time DNA fragmentation ing cell death
was observed.
As described in U.S. Patent No. 7,534,773, the contents of
which are incorporated by reference, enhancement of
2012/000609
tion—induced tumor cell death and delay of the
progression of androgen-sensitive cancer cells to androgen—
independence may be achieved by inhibiting the expression of
clusterin by the cells.
Custirsen
Custirsen is a second—generation antisense oligonucleotide that
inhibits clusterin expression. Custirsen is designed
specifically to bind to a portion of clusterin mRNA, resulting
in the inhibition of the tion of clusterin protein. The
structure of custirsen is ble, for e, in U.S.
Patent No. 6,900,187, the contents of which are incorporated
herein by reference. A broad range of s have shown that
custirsen potently regulates the expression of clusterin,
facilitates apoptosis, and sensitizes cancerous human prostate,
breast, ovarian, lung, renal, r, and melanoma cells to
chemotherapy (Miyake et al. 2005), see also, U.S. Patent
ation Publication No. 2008/0119425 A1. In a clinical
trial for androgen—dependent prostate cancer, the drugs
flutamide and buserelin were used together in combination with
custirsen, increasing prostate cancer cell apoptosis (Chi et
al. 2004; Chi et al., 2005).
Androgen Receptor Antagonists
Androgen receptor (AR) antagonists reduce the stimulation of
prostate cancer cells by androgens by perturbing or reducing a
function of AR, including androgen-AR binding, AR
transcriptional ty, or cellular transport of AR such as
translocation from the cytoplasm to the s. Custirsen is
not an AR antagonist. Custirsen inhibits the progression of
prostate cancer to androgen independence by reducing the anti—
tic effects of rin and is not thought to affect
androgen signaling pathways.
Combination Therapy
The administration of two drugs to treat a given condition,
such as prostate cancer, raises a number of potential
problems. In vivo interactions between two drugs are x.
The effects of any single drug are related to its absorption,
distribution, and elimination. When two drugs are introduced
into the body, each drug can affect the tion,
distribution, and ation of the other and hence, alter
the effects of the other. For instance, one drug may inhibit,
activate or induce the production of enzymes involved in a
metabolic route of elimination of the other drug (Guidance for
Industry. In vivo drug lism/drug interaction studies —
study design, data analysis, and recommendations for dosing
and labeling). Thus, when two drugs are administered to treat
the same condition, it is unpredictable whether each will
complement, have no effect on, or interfere with, the
therapeutic activity of the other in a human subject.
Not only’ may the ction. between. two drugs affect the
intended. therapeutic activity of each drug, but the
interaction may increase the levels of toxic metabolites
(Guidance for Industry. In vivo drug lism/drug
interaction studies - study design, data analysis, and
recommendations for dosing and labeling). The interaction may
also heighten or lessen the side effects of each drug. Hence,
upon administration of two drugs to treat a disease, it is
unpredictable what change will occur in the profile of each
drug.
W0 2012/123820
Additionally, it is difficult to accurately predict when the
effects of the interaction between the two drugs will become
manifest. For example, metabolic interactions n drugs
may" become apparent upon the initial stration of the
second drug, after the two have reached a steady-state
concentration or upon discontinuation of one of the drugs
(Guidance for Industry. In vivo drug metabolism/drug
interaction studies — study design, data analysis, and
recommendations for dosing and labeling).
Thus, the success of one drug or each drug alone in an in
vitro model, an animal model, or in humans, may not ate
into efficacy when both drugs are administered to humans.
x of the Invention
The present invention relates to a method for ng a
mammalian subject afflicted. with. prostate cancer comprising
administering to the mammalian subject i) an oligonucleotide
which reduces clusterin expression and ii) an androgen
receptor antagonist having the structure
or‘ a ceutically acceptable salt thereof, each. in an
amount that when in combination with the other is effective to
treat the mammalian subject.
In one aspect of the present invention there is provided a use
of i) an oligonucleotide which reduces clusterin expression
and ii) an en receptor antagonist having the structure
O)__%
or a pharmaceutically acceptable salt thereof, in the
manufacture of a medicament for treating a mammalian subject
ted with prostate cancer.
fibflowedbypage6d
In a r aspect of the present invention, there is
provided a use of i) an oligonucleotide which reduces
clusterin expression and ii) an androgen receptor antagonist,
in the cture of a Inedicament for the treatment of a
mammalian subject afflicted with androgen—independent prostate
cancer.
In a further aspect of the present invention there is provided
a. use of custirsen. in the manufacture of a Inedicament for
increasing the sensitivity of ARI resistant prostate cancer
cells to ARI.
In a further aspect of the present invention there is ed
a composition for treating a mammalian subject ted with
te cancer comprising i) an oligonucleotide which reduces
rin expression and ii) an androgen receptor antagonist
having the structure
F30 e g f
or a pharmaceutically acceptable salt thereof.
Some embodiments of the invention provide a method for
treatment of a mammalian subject afflicted with androgen—
independent prostate cancer, consisting‘ of administering to
the subject i) an oligonucleotide which reduces rin
[followed by page 6b]
expression, and ii) an androgen receptor antagonist, each in
an amount that when in combination with the other is effective
to treat the mammalian subject.
An aspect of the present invention provides a ceutical
composition comprising an amount of an oligonucleotide which
reduces Clusterin expression, and an androgen receptor
antagonist for use in treating a mammalian subject afflicted
with en—independent prostate cancer.
[followed by page 7]
—6b-
An aspect of the present invention provides an oligonucleotide
which s clusterin expression for use in combination with
an androgen receptor nist in treating a mammalian
subject afflicted with androgen—independent prostate cancer.
An aspect of the t invention provides a composition for
treating a mammalian subject afflicted with prostate cancer
sing i) an oligonucleotide which reduces clusterin
expression and ii) an androgen receptor antagonist having the
structure
NC k 12
F3C > 4 i
or a pharmaceutically acceptable salt thereof.
PCT/IBZOIZIOOO609
Brief Description of the Drawings
Figure 1. Inhibition of LNCaP cell eration upon
treatment with luM ARl and lOnM siRNA targeting
clusterin (CLU) or lOnM SCR. SCR. is a scrambled
sequence siRNA control. (A), FBS condition is media
supplemented with FBS. (B), CSS condition is
charcoal serum stripped media.
Figure 2. Inhibition of LNCaP cell proliferation upon
ent with luM ARl and SOOnM custirsen or 500nM
SCRB. SCRB is a scrambled sequence antisense
oligonucleotide control. (A), FBS condition is media
mented with PBS. (B), CSS condition is
charcoal serum stripped media.
Figure 3. Inhibition of C4-2 cell proliferation upon treatment
with luM ARl and SOOnM custirsen or SOOnM SCBR. (A),
FBS condition is media supplemented with FBS. (B),
CSS condition is charcoal serum stripped media.
Figure 4. PC—3 (AR—negative) cell eration upon treatment
with luM ARl and lOnM siRNA targeting rin or
lOnM SCR (A). PC—3 (AR~negative) cell proliferation
upon treatment with luM ARl and 500nM custirsen or
SOOnM SCRB (B).
Figure 5. Cytotoxicity in LNCaP cells upon treatment with ARl
and lOnM siRNA targeting clusterin or 10nM SCR (A).
Cytotoxicity in LNCaP cells upon treatment with luM
ARl and SOOnM custirsen or 500nM SCRB (B). Cells
grown in media supplemented with PBS. X—axis is ARl
concentration.
W0 2012/123820
Figure 6. Potency of ARl and custirsen combination therapy in
LNCaP cells. (A), Cell growth inhibition after
treatment with each drug or a combination thereof by
crystal violet assay. X-axis is [AR1]/[custirsen].
P-value was calculated by the Friedman test. (B),
Dose effect curve for each treatment. (C),
Combination index (CI) at l effective doses.
CI =
i, additive effect; Cl < l, combination effect;
C: > ;, antagonistic effect.
Figure 7. Cell Cycle Distribution upon treatment of ARl, siRNA
targeting clusterin, or a combination f in
LNCaP cells. OTR refers to cells d with
oligofectamine transfection reagent (Invitrogen Life
Technologies, Inc.) in the absence of custirsen or
SiRNA.
Figure 8. FACS Analysis of Cell Cycle Distribution upon
treatment of ARl, custirsen, or a combination
thereof in LNCaP cells.
Figure 9. Effect of ARl stration on .AR and clusterin
protein sion in LNCaP cells. (A), lOuM ARl.
(B), after 48 hours of ARl treatment at indicated
concentrations.
Figure 10. Effect of ARl administration on AKT and ERK
phosphorylation and protein levels in LNCaP cells.
(A), lOuM ARl. (B), after 48 hours of ARl treatment
at indicated concentrations. (C), Dose ent
change of expression level of ART or ERK after
W0 2012/123820
treatment with ARl. (D), Dose dependent change of
expression level of ART or ERK after treatment with
ARl .
Figure 11. Effect of ARl administration on AR and clusterin
mRNA expression in LNCaP cells. (A), AR mRNA
expression 48 hours after adding ARl at each
concentration. (B), AR mRNA expression at indicated
time points following the addition of lOpM ARl. (C),
clusterin mRNA expression 48 hours after adding ARl
at each concentration. (D), Clusterin mRNA
expression at indicated. time points following the
addition of lOnM ARl.
Figure 12. Change of protein expression in LNCAP cells after
treatment of siRNA targeting clusterin, custirsen,
or ted controls (A and C). (B), comparison of
clusterin upregulation by bicalutamide and ARl. (D),
Expression of AR perone by treatment with ARl
and sen (ASO).
Figure 13. Effect of lOuM ARl and lOnM siRNA targeting
clusterin on PSA in LNCaP cells (A), or on AR mRNA
expression (B).
Figure 14. Effect 0: lOuM ARl and SOOnM custirsen combination
therapy on AR (A), or PSA mRNA expression (B).
Figure 15. Western blot analysis of protein expression and
PARP cleavage after ent of LNCaP cells with
ARl and lOnM siRNA. targeting clusterin. FBS
condition is media supplemented with PBS. CSS
condition is charcoal serum stripped media.
Figure 16. Effect of ARl and louM siRNA targeting clusterin on
protein‘ levels upon androgen stimulation in LNCaP
cells. R1881 is a potent androgen that is also known
as metribolone.
Figure 17. n blot analysis of protein expression and
PARP cleavage upon treatment of LNCaP cells with ARl
and custirsen or control. Cells (leO6) were seeded
in 10cm dishes with RPMI medium containing 5% PBS.
The next day, cells were transfected. with SOOnM
custirsen or control for 48h. 10pm ARl was then
added to the cells for 48 hours before harvesting
for Western blot analysis. AR and PSA expression
were highly repressed by custirsen and ARl
combination therapy.
Figure 18. n blot analysis of n expression and
phosphorylation upon treatment of LNCaP cells with
ARl and lonM siRNA targeting clusterin, or l.
Phospho—AKT and phosph—ERK are activated by AR1
treatment; however, ARl and. Clu siRNA. combination
therapy reduces levels of phosphorylated ART and ERK
protein. Combination treatment represses the AKT—
mTOR—p7OS6K pathway more ly than erapy.
Figure 19. Western blot analysis of AR proteasome degradation
upon treatment of LNCaP cells with a combination of
ARl and custirsen or an siRNA targeting clusterin.
M6132 is a proteasome inhibitor, and CHX is
cycloheximide, an tor of protein biosynthesis.
AR protein degradation is potently increased by ARl
and custirsen combination therapy.
Figure 20. Effect of ARl and custirsen combination therapy on
AR transcriptional activity. Dual luciferase assay;
LNCaP cells were transfected for 2 days with SOOnM
custirsen in CSS. ARl (luM) or DMSO was then added
with or without R1881 (1nM) for 24 hours before
ting for is.
Figure 21. Increased inhibition of AR ocation from the
cytoplasm to the nucleus upon combination of lonM
siRNA targeting clusterin with louM ARl compared to
monotherapy. LNCaP cells were used.
Figure 22. Increased inhibition of AR translocation from the
cytoplasm to the nucleus upon combination of lonM
siRNA targeting clusterin with lOuM ARl ed to
monotherapy. LNCaP cells were used.
Figure 23. Increased, association. of AR. with. ubiquitin upon
combination treatment of lOuM ARl and 10nM siRNA
targeting clusterin compared to monotherapy (A).
Association of ARV with. ubiquitin. upon combination
treatment of louM ARl and lOnM siRNA targeting
Clusterin, or control in the presence of MG132 (B).
LNCaP cells were used.
Figure 24. Comparison of clusterin knock—down between
treatment of bicalutamide or ARl, in ation
with custirsen (ASO) or control in LNCaP cells.
PCT/IBZOIZIOOO609
Figure 25. Effect of (FKBP52) over—expression on AR
degradation and clusterin knock-down in LNCaP cells.
Figure 26. Decreased tion-resistant prostate cancer
tumor growth and increased survival upon combination
treatment of ARl and custirsen in mice. Male athymic
nude mice were injected s.c. in two sites with LNCaP
cells in Matrigel. The mice were castrated once
tumors reached 150mm3 or‘ the PSA. level increased
above 50ng/mL. Once tumors progressed to castration
resistance (PSA levels increased to the same level
as pre—castration), 10 mice were randomly assigned
to each of ARl + led antisense oligonucleotide
(SCRB) or ARl + custirsen. Custirsen (lOmg/kg/each
dose) or SCRB (lOmg/kg/each dose) was injected i.p.
once daily for the first week and then three times
per week. ARl (lOmg/kg/each dose) was administered
orally once daily ng) 7 days per week for 8 to
12 weeks.
Figure 27. Increased survival upon combination treatment of
ARl and sen in mice. Male athymic nude mice
were injected s.c. in two sites with LNCaP cells in
el. The mice were castrated once tumors
reached lSOHm§ or the PSA level increased above
50ng/mL. Once tumors progressed to castration
resistance (PSA levels increased to the same level
as pre-castration), 10 mice were randomly assigned
to each of ARI + scrambled antisense oligonucleotide
(SCRB) or ARl + sen. Custirsen (lOmg/kg/each
dose) or SCRB (lOmg/kg/each dose) was injected i.p.
2012/000609
once daily for the first week and then three times
per week. ARl (lOmg/kg/each dose) was administered
orally once daily ng) 7 days per week for 8 to
12 weeks.
Figure 28. Decreased PSA protein expression upon combination
treatment of ARl and custirsen in mice. Male athymic
nude mice were injected s.c. in two sites with LNCaP
cells in Matrigel. The mice were castrated once
tumors reached 150mm3 or the PSA. level increased
above 50ng/mL. Once tumors progressed to castration
resistance (PSA levels increased to the same level
as pre-castration), 10 mice were randomly assigned
to each of ARl + scrambled antisense oligonucleotide
(SCRB) or ARl + custirsen. Custirsen (lOmg/kg/each
dose) or SCRB (lOmg/kg/each dose) was injected i.p.
once daily for the first week and then three times
per week. ARl (lOmg/kg/each dose) was administered
orally once daily (morning) 7 days per week for 8 to
12 weeks.
Figure 29. Decreased PSA protein expression upon combination
treatment of ARl and custirsen in mice. Male athymic
nude mice were injected s.c. in two sites with LNCaP
cells in Matrigel. The mice were castrated once
tumors reached. lSOHm9 or the PSA level increased
above 50ng/mL. Once tumors progressed to castration
resistance (PSA levels increased to the same level
as stration), 10 mice were randomly assigned
to each of ARI + led antisense oligonucleotide
(SCRB) or ARl + custirsen. Custirsen kg/each
dose) or SCRB (lOmg/kg/each dose) was injected i.p.
once daily for the first week and then three times
per week. ARl (lOmg/kg/each dose) was stered
orally once daily (morning) 7 days per week for 8 to
12 weeks.
Figure 30. Clusterin expression is induced in ARl resistant
tumors. (A) Increased clusterin expression ing
ARl treatment. (B) Increased. clusterin expression
following ARl treatment in ARl resistant tumors. (C)
Clusterin expression is up—regulated in a time and
dose dependent manner after ARl treatment, as
determined by Western blot analysis.
Figure 31. Combination treatment of custirsen and ARl is more
effective than custirsen or ARl monotherapy in CRPC
LNCaP xenografts. ARl plus custirsen treatment
decreased AR and PSA expression in CRPC xenografts.
Figure 32 . Clusterin knockdown decreases AR. transcriptional
activity and expression of AR—dependent genes.
Transmembrane protease serine 2 (TMPRSSZ) mRNA
levels sed following ARl treatment, clusterin
knockdown, and ARl treatment plus clusterin
knockdown.
Figure 33 . Clusterin knockdown ses AR. protein levels
when ed. with. ARl. The le interaction
between heat shock protein 27 (Hsp27) and AR
contributing to AR transcriptional activity, PSA
expression, and cell survival is depicted.
Figure 34. clusterin. knockdown. decreases heat shock factor
protein 1 ) transcription. ty and
expression of heat shock proteins.
Figure 35. Clusterin overexpression increases HSF—l activity.
Figure 36. Possible mechanism of action for ARl treatment plus
custirsen treatment in a tumor cell.
Figure 37. clusterin and autophagy may play a role in stress
and cancer. Increased clusterin expression following
asmic reticulum (ER) stress, chemo~stress, and
androgen deprivation is depicted.
Figure 38. ARl treatment induces autophagy in LNCaP cells.
Figure 39. Treatment stressors induce clusterin which co—
localizes with LC3B in aggresomes.
Figure 40. ER stress—induced autophagy is inhibited by
clusterin silencing.
Figure 41. CLU is induced by ARl and highly expressed in ARl
resistant cells. C, Dose and time dependent ARl
induction. of CLUL D, CLU‘ is also induced. in .ARl
resistant cells by AR knock down. AR ASO also
induces CLU in several ARl ant MR49F cells.
CLU is high in ARl resistant cells, e.g. MR49F.
Figure 42. ion of stress response (ER, YB—l), as well as
cross talk of pAKT and MAPK.
(followed by page 16a)
Figure 43. The combination of CLU inhibition and ARI increases
inhibition of LNCaP cell growth compared to CLU
inhibition or AR1 monotherapy.
Figure 44. s of combination treatment on tumor volume
and serum PSA level.
Figure 45. Effect of combination treatment on AR
transactivation and ocation. A, Custirsen
combined with AR1 treatment decreases AR
transactivation more than custirsen or ARl
monotherapy. LNCaP cells were transfected 500 nmol/L
of custirsen or SCRB control for 2 consecutive days,
and at day 2, transiently cotransfected with 1 ug of
PSA—luciferase and Renilla-luciferase. On the next
day, the cells were treated 10 umol/L of AR1, then
added 1 nmol/L of R1881 or vehicle for 24 h. Cells
were harvested, and luciferase activity was
determined. Columns ent means of at least
three independent experiments done in triplicate.
PSA activation was normalized Renilla—luciferase
activity. B, Effect of AR translocation by
ation treatment. 24 hours after transfection
with 10 nmol/L of CLU siRNA or SCR siRNA control,
LNCaP cells were treated with DMSO, 10 pmol/L of ARl
and 1nmol/L of R1881 for 30 minutes and fixed in
methanol/acetone for immunofluorescence staining
with anti—AR antibody. Nuclei were stained with
DAPI. ARl inhibited AR translocation from the
cytoplasm to the nucleus. CLU knockdown combined
with ARl shows increased effects of tion of AR
translocation.
—16a— (followed by page 17)
Figure 46. The effect of CLU own combined with ARl on AR
expression.
Figure 47. Western Blot Analysis following CLU knockdown.
(followed by page 18)
Detailed Description of the Invention
The present ion relates to a method for treating a
mammalian subject tedv with. prostate cancer comprising
stering to the mammalian subject i) an oligonucleotide
which reduces clusterin expression and ii) an androgen
receptor antagonist having the structure
NC /JL\ IZ
QC 9 é
o :’
or‘ a pharmaceutically‘ acceptable salt thereof, each. in an
amount that when in combination with the other is effective to
treat the mammalian subject.
In some embodiments of the invention the cancer is androgen—
independent prostate cancer.
In some embodiments, the amount of the oligonucleotide and the
amount of the androgen receptor nist or a
pharmaceutically acceptable salt thereof when taken together
is more effective to treat the subject than when each agent is
administered alone.
In some embodiments, the amount of the oligonucleotide in
combination with the amount of the androgen receptor
antagonist cm: a pharmaceutically acceptable salt thereof is
less than is ally ive when administered alone.
-18—
2012/000609
In some embodiments, the amount of the androgen receptor
antagonist or a pharmaceutically able salt thereof in
combination with the amount of the oligonucleotide is less
than is clinically effective when administered alone.
In some embodiments, the amount of the oligonucleotide and the
amount of the androgen receptor antagonist or a
ceutically acceptable salt thereof when taken together
is effective to reduce a clinical symptom of prostate cancer
in the subject.
In some embodiments, the mammalian subject is human.
In some embodiments, the oligonucleotide is an antisense
oligonucleotide.
In some embodiments, the antisense oligonucleotide spans
either the translation initiation site or the ation site
of clusterin—encoding mRNA.
In some embodiments, the antisense oligonucleotide is modified
to enhance in vivo stability relative to an unmodified
oligonucleotide of the same sequence.
In some embodiments the antisense oligonucleotide consists
ially of an ucleotide selected from the group
consisting of Seq. ID Nos. 1 to 11.
In some embodiments, the antisense oligonucleotide consists
essentially of an oligonucleotide consisting of Seq. ID No. 3.
In some ments, the oligonucleotide is custirsen.
In some embodiments, the amount of custirsen is less than
640mg.
In some embodiments, the amount of custirsen is less than
480mg.
In some embodiments, the amount of custirsen is stered
intravenously once in a seven day period.
In some embodiments, the amount of the androgen receptor
antagonist is less than 240mg.
In some ments, the amount of the androgen receptor
antagonist is from 150mg to 240mg.
In some embodiments, the amount of the androgen receptor
antagonist is from 30mg to 150mg.
In some embodiments, the amount of the en receptor
antagonist is 80mg.
In some embodiments, the amount of the androgen receptor
antagonist is administered orally once per day.
Some embodiments of the invention provide a method for
treatment of a mammalian subject afflicted. with androgen—
independent prostate cancer, consisting of administering to
the subject i) an androgen receptor antagonist and ii) an
oligonucleotide which s clusterin expression, each in an
amount that when in ation with the other is effective to
treat the mammalian subject.
In some embodiments the androgen receptor antagonist is a non-
steroidal antiandrogen.
In some embodiments, the androgen receptor antagonist is ARl.
In some embodiments, the androgen—independent prostate cancer
is resistant to ARl.
In some ments the combination of the androgen receptor
antagonist and the oligonucleotide is effective to decrease
androgen receptor translocation from the cytoplasm to the
s of the tumor cells.
In some embodiments, the combination of the androgen receptor
nist and the oligonucleotide is ive to increase
the proteasome degradation of the androgen receptor protein in
the tumor cells.
in some embodiments, the combination of the androgen receptor
antagonist and. the oligonucleotide is effective to decrease
androgen receptor riptional activity in the tumor cells.
In some embodiments, the combination of the androgen or
antagonist and the oligonucleotide is effective to decrease
the amount of phosphorylated AKT in the tumor cells.
In some embodiments, the ation of the androgen receptor
antagonist and the oligonucleotide is effective to decrease
the amount of phosphorylated ERK in the tumor cells.
W0 2012/123820
In some embodiments, the combination of the androgen or
antagonist and the ucleotide is ive to inhibit the
proliferation of prostate cancer cells.
Some embodiments of the invention provide a Hethod by which
ARl resistant prostate cancer cells are sensitized to ARl by
concomitant treatment with custirsen.
Some embodiments of the ion provide a method of
increasing the sensitivity of ARl resistant prostate cancer
cells to ARI comprising treating the ARl resistant prostate
cancer cells with custirsen.
Some embodiments of the invention provide a method for
treatment of a mammalian subject afflicted with prostate
cancer that is resistant to ARl, comprising stering to
the subject i) ARl and ii) custirsen, each in an amount that
when in combination with the other is effective to treat the
mammalian subject, wherein the custirsen increases the
sensitivity of the prostate cancer to ARl.
An aspect of the present invention provides a pharmaceutical
composition comprising an amount of an oligonucleotide which
reduces clusterin expression, and an en or
antagonist for use in treating a mammalian subject afflicted
with androgen-independent prostate cancer.
An aspect of the t invention provides an oligonucleotide
which reduces clusterin expression for use in combination with
an androgen receptor antagonist in treating a ian
subject afflicted with androgen-independent prostate cancer.
An aspect of the present ion provides a composition for
treating a mammalian subject ted with te cancer
comprising i) an oligonucleotide which reduces clusterin
expression and ii) an androgen receptor antagonist having the
structure
go > g ,
or a pharmaceutically able salt thereof.
Aspects of the invention involve the increased potency of the
combination of an oligonucleotide which. decreases clusterin
expression and an AR antagonist in the treatment of prostate
cancer compared to ucleotide or AR antagonist
monotherapy. Increased potency es but is not limited to
reduced proliferation of prostate cancer cells, increased
apoptosis of cancer cells, reduced ocation of AR from
the cytoplasm to the nucleus, reduced transcriptional activity
of AR, increased PARP cleavage, reduced AKT phosphorylation,
reduced. ERK phosphorylation, and increased. AR protein
degradation. in embodiments in which AKT and/or IERK
phosphorylation is reduced, all isoforms of AKT and ERK are
envisioned. This includes but is not limited to AKTl, AKTZ,
AKT3, ERK1, and ERKZ. In some embodiments, the increased
some degradation of AR involves the increased
association of AR with ubiquitin.
Each embodiment disclosed herein is contemplated as being
applicable to each of the other disclosed embodiments. Thus,
all combinations of the s elements described herein are
within the scope of the invention.
It is understood that where a parameter range is provided, all
integers within that range, and tenths thereof, are also
provided by the invention. For example, “0.2—5 mg/kg/day”
includes 0.2 mg/kg/day, 0.3 mg/kg/day, 0.4 mg/kg/day, 0.5
day, 0.6 mg/kg/day etc. up to 5.0 mg/kg/day.
Terms
As used, herein, and unless stated otherwise, each. of the
following terms shall have the tion set forth below.
As used herein, “about” in the context of a numerical value or
range means i10% of the numerical value or range recited or
claimed.
As used in the specification and claims of this application,
the term "clusterin" refers to a glycoprotein t in
mammals, including , and denominated as such in the
humans. The sequences of numerous clusterin species are known.
For example, the sequence of human clusterin is described by
Wong et al., Eur. J. Biochem. 221 (3),9l7—925 (1994), and in
NCBI sequence accession number NM_001831 (SEQ ID NO: 43). In
this human sequence, the coding ce spans bases 48 to
1397.
As used herein, “oligonucleotide which reduces clusterin
sion” is an oligonucleotide with a sequence which is
effective to reduce clusterin expression in a cell. The
oligonucleotide which reduces clusterin expression may be, for
example, an antisense oligonucleotide or an RNA interference
inducing molecule.
As used , “antisense oligonucleotide” refers to a non-
RNAi oligonucleotide that reduces clusterin sion and that
has a sequence complementary to rin mRNA. Antisense
oligonucleotides may be antisense oligodeoxynucleotides (ODN).
Exemplary sequences which can be employed as antisense
molecules in the invention are disclosed in PCT Patent
Publication WO 37, U.S. Patent Publication No. US 2002—
0128220 Al, and U.S. Patent No. 6,383,808, all of which are
incorporated herein by reference. Specific antisense sequences
are set forth in the present application as SEQ ID NOS: 1 to
11, and may be found in Table l.
Table 1. Sequence Identification s for Antisense
oligonucleotides
SEQ ID NO: Sequence
1 GCACAGCAGG AGAATCTTCA T
2 TGGAGTCTTT GCACGCCTCG G
3 CAGCAGCAGA GTCTTCATCA T
4 ATTGTCTGAG ACCGTCTGGT C
CCTTCAGCTT TGTCTCTGAT T
6 AGCAGGGAGT CGATGCGGTC A
7 ATCAAGCTGC GGACGATGCG G
8 AGCC CGTGGAGTTG T
9 TTCAGCTGCT CCAGCAAGGA G
AATTTAGGGT TCTTCCTGGA G
CGGA GTTGGGGGCC T
The ODNs employed may be modified to increase the stability of
the ODN’ in ViVO. For example, the ODNs may‘ be employed. as
phosphorothioate derivatives (replacement of a non—bridging
phosphoryl oxygen atom with a sulfur atom) which have increased
ance to nuclease digestion. MOE (2’—O—(2—methoxyethyl))
modification (ISIS backbone) is also effective. The
construction of such modified ODNs is described in detail in
U.S. Patent No. 6,900,187 B2, the contents of which are
incorporated. by reference. In some embodiments, the ODN is
custirsen.
As used herein, “custirsen” refers to an antisense
ucleotide that reduces clusterin. expression. having the
sequence CAGCAGCAGAGTCTTCATCAT (Seq. ID No.: 3), wherein the
anti—clusterin oligonucleotide has a orothioate backbone
hout, has sugar‘ moieties of nucleotides 1—4 and 18—21
bearing 2’-O-methoxyethyl cations, has nucleotides 5—17
which are 2’deoxynucleotides, and has 5—methylcytosines at
nucleotides 1, 4, and 19. sen is also known as TV—lOll,
OGX—Oll, ISIS 112989 and Custirsen Sodium.
As used herein, “RNA interference inducing molecule” refers to
a molecule capable of inducing RNA interference or “RNAi” of
clusterin expression. RNAi involves mRNA degradation, but many
of the biochemical mechanisms underlying this interference are
unknown. The use of RNAi has been described in Fire et al.,
~26-
W0 2012/123820
1998, CartheW' et al., 2001, and. Elbashir‘ et a1., 2001, the
contents of which are incorporated herein by reference.
Isolated RNA molecules can mediate RNAi. That is, the isolated
RNA molecules of the present invention mediate degradation or
block expression of mRNA that is the transcriptional product
of the gene, which is also referred to as a target gene. For
convenience, such mRNA may also be referred to herein as mRNA
to be degraded. The terms RNA, RNA le(s), RNA segment(s)
and RNA fragment(s) may be used interchangeably to refer to
RNA that mediates RNA interference. These terms include
double-stranded. RNA, small interfering‘ RNA (siRNA), hairpin
RNA, single—stranded. RNA, isolated. RNA (partially' ed
RNA, essentially pure RNA, tic RNA, recombinantly
produced RNA), as well as altered RNA that s from
naturally occurring RNA by the addition, deletion,
substitution and/or alteration of one or more nucleotides.
Such alterations can include addition of non—nucleotide
material, such as to the end(s) of the RNA or internally (at
one or more nucleotides of the RNA). Nucleotides in the RNA
les of the present invention can also comprise
nonstandard nucleotides, including non-naturally occurring
tides or deoxyribonucleotides. Collectively, all such
altered RNAi molecules are referred to as analogs or analogs
of naturally-occurring RNA. RNA of the present invention need
only be sufficiently similar to l RNA that it has the
ability to mediate RNAi.
As used herein the phrase "mediate RNAi" refers to and
indicates the ability to distinguish which mRNA molecules are
to be afflicted with the RNAi machinery or process. RNA that
mediates RNAi interacts with the RNAi machinery such that it
WO 23820
directs the machinery to degrade particular mRNAs or to
otherwise reduce the expression of the target protein. In one
embodiment, the t invention relates to RNA. les
that direct cleavage of specific mRNA to which their sequence
corresponds. It is not ary that there be perfect
pondence of the sequences, but the correspondence must
be sufficient to enable the RNA to direct RNAi inhibition by
cleavage or blocking expression of the target mRNA.
As noted above, the RNA molecules of the present invention in
general comprise an RNA portion and some additional portion,
for example a deoxyribonucleotide portion. The total number of
nucleotides in the RNA le is suitably less than in order
to be effective mediators of RNAi. In preferred RNA les,
the number of nucleotides is 16 to 29, more preferably 18 to
23, and most preferably 21-23. Suitable sequences are set
forth in the present application as SEQ ID N08: 19 to 42
(Table 2).
Table 2. Sequence Identification Numbers for RNA Interference
Inducing Molecules
SEQ ID NO: Sequence
NNNNNNNNNH GSU'IhUJNi—‘OKO CCAGAGCUCG CCCUUCUACT T
GUAGAAGGGC GAGCUCUGGT T
CAAC ACCUCCUCCT T
GGAGGAGGUG UUGAGCAUCT T
UAAUUCAACA AAACUGUTT
GACAGUUUUA UUGAAUUAGT T
UAAUUCAACA AAACUGUTT
ACAGUUUUGU UGAAUUATT
7 AUGAUGAAGA CUCUGCUGCT T
8 GCAGCAGAGU CUUCAUCAUT T
_ GUAGAAGGGC GAGCUCUGGT T
GUCCCGCAUC GUCCGCAGCT T
— GCUGCGGACG AUGCGGGACT T
GACAGUUUUA UUGAAUUAGT T
AUGAUGAAGA CUCUGCUGC
GCAGCAGAGU CUUCAUCAU
The siRNA molecules of the invention are used in therapy to
treat patients, including human patients, that have cancers or
other diseases of a type where a therapeutic benefit is
obtained by the inhibition of expression of the targeted
protein. siRNA molecules of the invention are administered to
patients by one or more daily ions (intravenous,
subcutaneous or hecal) or by continuous intravenous or
intrathecal stration for one or more treatment cycles to
reach plasma and tissue concentrations suitable for the
regulation of the ed mRNA and protein.
As used herein, a “mammalian subject afflicted with prostate
cancer” means a mammalian t who was been affirmatively
diagnosed to have prostate cancer.
As used herein, “androgen-independent prostate cancer”
encompasses cells, and tumors predominantly containing cells,
that are not androgen—dependent (not androgen sensitive). Often
-29_
W0 2012/123820
androgen—dependent cells progress from being androgen-dependent
to being androgen-independent. Additionally, in some
ments androgen—independent prostate cancer may encompass
a tumor that overall is not androgen—dependent (not androgen
sensitive) for growth. In some embodiments, androgen
independent prostate cancer has progressed since the
administration of e ablation y and/or the
administration of an AR antagonist (as in hormone blockade
therapY). In some embodiments, there is increased AR expression
in the androgen—independent prostate cancer ed to
prostate cancer that is not androgen—independent.
As used herein, “castration-resistant prostate cancer”
encompasses any androgen—independent ‘prostate cancer that is
resistant to hormone ablation therapy or hormone blockade
therapy. In some embodiments, castration-resistant prostate
cancer has progressed since the administration of hormone
ablation and/or hormone blockade therapy. In some embodiments,
there is increased th expression ill the castration—resistant
te cancer compared to prostate cancer that is not
castration resistant.
As used herein, “androgen—withdrawal” encompasses a reduction
in the level of an androgen in a patient afflicted with
prostate cancer.
As used herein, “hormone blockade therapy” means a reduction in
the function of receptors or cellular pathways that respond to
an en. A. non—limiting' example of a e de
therapy is an AR antagonist.
As used herein “androgen ablation therapy” is any therapy that
is capable of causing androgen-withdrawal in a mammalian
subject afflicted with prostate cancer. Terms used herein that
are synonymous with en ablation therapy, are “androgen
withdrawal” and “hormone ablation therapy”. Non-limiting
es of androgen ablation therapies include both surgical
(removal of both testicals) and l (drug induced
ssion of testosterone or testosterone induced signaling)
castration. Medical castration can be achieved. by various
regimens, ing but not limited to LHRH agents, and agents
that reduce androgen expression from. a gland such as the
adrenal glands (Gleave et al., 1999; Gleave et al., 1998).
As used herein, “AR antagonist” refers to an agent that
perturbs or reduces a function of AR, including androgen
binding, AR ing, cellular transport of AR such as
translocation front the cytoplasnl to the nucleus, AR. protein
levels, or AR protein expression. AR antagonists include but
are not limited to AR-specific monoclonal antibodies,
oligonucleotides that target AR expression (such as AR—
targeting antisense oligonucleotides or RNA inducing
molecules), peptide agents specific for AR, and small molecule
inhibitors specific for AR. An AR antagonist may be ea non—
steroidal antiandrogen such. as ARl, bicalutamide, ide,
nilutamide, RDl62, and ZD4054.
ARl is an AR antagonist of the ion having the structure:
F3C 2 g ,-
2012/000609
Methods of synthesis for ARl are described in U.S. Patent No.
7,709,517 B2, the contents of which are incorporated herein by
reference. Alternatively, AR1 may be ed from Medivation,
Inc. (San sco, California, USA). The CAS Registry No.
for ARl is 915087—33—1, and its PubChem No. is 15951529. AR1
has the chemical formula. C2fih5F4N4028, and. is also known as
MDV3100 and 4-(3—(4—cyano—3—(trifluoromethyl)phenyl)—5,5—
dimethyl—4~oxo—2~thioxoimidazolidinyl)~2~fluoro-N-
methylbenzamide. ARl is a second generation orally available
AR nist that works by blocking androgen binding to AR,
impeding the r ocation of AR from the cytoplasm,
and inhibiting AR-DNA. binding (Tran et al. 2009). ARl is
currently being evaluated in clinical trials for the treatment
of advanced prostate cancer (Scher et al., 2010).
As used herein, “transcriptional activity” refers to a
protein’s ability to bind or otherwise become directly or
indirectly ated with a portion of DNA in a cell
resulting in an influence on the level of expression of one or
more genes.
The inhibition of clusterin expression may be transient, and
may occur in combination with androgen ablation therapy or
administration of an AR antagonist. In humans with. prostate
cancer that is not androgen—independent, this means that
inhibition of expression should be effective starting within a
day or two of androgen withdrawal or administration of an AR
antagonist, and ing for about 3 to 6 months thereafter.
This may require Inultiple doses to accomplish. It will be
appreciated, however, that the period of time may be more
prolonged, starting before castration and extending for
W0 2012/123820
ntial time afterwards t departing from the scope of
the invention.
Aspects of the invention can be applied to the treatment of
en-independent prostate cancer, or to prevent prostate
cancer from becoming androgen-independent.
Aspects of the invention can be applied to the treatment of
castration—resistant ate cancer, or 11) prevent prostate
cancer from becoming castration—resistant.
nation” means either at the same time and frequency, or
more usually, at different times and frequencies than an
oligonucleotide which reduces clusterin expression, as part of
a single treatment plan. Aspects of the invention include the
administration of the oligonucleotide before, after, and/or
during the administration of an AR antagonist. An AR antagonist
may therefore be used, in combination with the oligonucleotide
according to the invention, but yet be administered at
different times, different dosages, and at a different
frequency, than a oligonucleotide which reduces clusterin
expression.
As used herein, an “amount” or “dose” of an oligonucleotide
measured in rams refers to the milligrams of
oligonucleotide t in a drug product, regardless of the
form of the drug product.
As used herein, “effective” when referring to an amount of
oligonucleotide which reduces clusterin expression, an AR
antagonist, or any combination thereof refers to the ty
of oligonucleotide, AR antagonist, or any combination thereof
that is sufficient to yield. a desired. therapeutic response
without undue adverse side s (such as toxicity,
irritation, or allergic response) commensurate with a
reasonable benefit/risk ratio when used in the manner of this
invention.
As used herein, “treating” encompasses, e.g., inhibition,
regression, or stasis of the progression of prostate cancer.
Treating also encompasses the prevention or amelioration of
any symptom or symptoms of te cancer.
As used herein, “inhibition” of disease progression or disease
complication in a subject means preventing' or reducing' the
disease progression and/or disease complication in the subject.
As used herein, a “symptom” associated with prostate cancer
es any al or tory manifestation associated
with prostate cancer, and is not limited to what the subject
can feel or observe.
As used herein, “pharmaceutically acceptable r” refers
to a r or excipient that is suitable for use with humans
and/or animals without undue adverse side effects (such as
ty, irritation, and allergic response) commensurate with
a reasonable benefit/risk ratio. It can be a pharmaceutically
acceptable solvent, suspending agent or vehicle, for
delivering the instant compounds and/or combinations to the
subject.
Dosage Units
Administration of an oligonucleotide that targets rin
expression can be carried out using the various mechanisms
known in the art, including naked administration and
administration in pharmaceutically acceptable lipid carriers.
For example, lipid carriers for antisense delivery are
sed in U.S. Patent Nos. 5,855,911 and 5,417,978, which
are incorporated herein by nce. In general, the
oligonucleotide is administered by intravenous (i.v.),
intraperitoneal (i.p.), subcutaneous (8.0.), or oral routes, or
direct local tumor injection. In red, embodiments, an
oligonucleotide targeting rin expression is administered
by i.v. injection. In some embodiments, the amount or
ucleotide administered is 640mg.
The amount of oligonucleotide administered is one effective to
inhibit the expression of rin in prostate cells. It will
be appreciated that this amount will vary both with the
effectiveness of the oligonucleotide employed, and. with the
nature of any carrier used.
The amount of antisense oligonucleotide targeting rin
expression administered may be from 40 to 640 mg, or 300—640
mg. Administration of the antisense oligonucleotide may be once
in a seven day period, 3 times a week, or more specifically on
days 1, 3 and 5, or 3, 5 and 7 of a seven day period. In some
embodiments, administration of the antisense oligonucleotide is
less frequent than once in a seven day period. Dosages may be
calculated by patient weight, and therefore a dose range of
about 1—20 mg/kg, or about 2-10 mg/kg, or about 3—7 mg/kg, or
about 3—4 mg/kg' could. be used. This dosage is repeated. at
intervals as needed. One al concept is dosing once per
week with 3 loading doses during week one of treatment. The
amount of antisense oligonucleotide administered is one that
WO 23820
has been demonstrated. to be effective in human patients to
inhibit the expression of clusterin in cancer cells.
In some embodiments of the invention, the amount of
oligonucleotide targeting the expression of clusterin required
for treatment of prostate cancer is less in ation with an
AR antagonist, than would. be required. with oligonucleotide
monotherapy.
Custirsen may be formulated at a concentration of 20 mg/mL as
an isotonic, phosphate-buffered saline on for IV
administration and can be supplied as an 8 mL solution
containing 160 mg custirsen sodium in a single vial.
Custirsen may be added to 250 mL 0.9% sodium chloride (normal
). The dose may be administered using either a peripheral
or central indwelling catheter enously as an infusion
over 2 hours. Additionally, an infusion pump may be used.
Administration of an AR antagonist may be oral, nasal,
pulmonary, parenteral, i.v., i.p., intra—articular,
transdermal, intradermal, s.c., l, intramuscular, rectal,
intrathecal, intraocular, and buccal. A, preferred route of
administration for ARl is oral. One of skill in the art will
recognize that higher doses may be required for oral
stration of an AR antagonist than for i.v. injection.
The dose of an AR antagonist may be 30mg, 35mg, 40mg, 45mg,
50mg, 55mg, 60mg, 65mg, 70mg, 75mg, 80mg, 85mg, 90mg, 95mg,
100mg, 150mg, 240mg, 360mg, 480mg, or 600mg. In some
embodiments, the dose of an AR antagonist is less than 30mg. In
these embodiments the dose may be as low as 25mg, 20mg, 15mg,
—36-
10mg, 5mg, or less. In some ments, the dose of an AR
antagonist is administered daily. In some embodiments the dose
is administered orally.
A dosage unit of the oligonucleotide which reduces clusterin
expression and an AR antagonist may comprise one of each singly
or mixtures thereof. A combination of an oligonucleotide which
reduces clusterin. expression and. ARl can be administered. in
oral dosage forms as tablets, capsules, pills, powders,
granules, elixirs, tinctures, suspensions, syrups, and
ons. An oligonucleotide which reduces clusterin
expression and/or an AR antagonist may also be administered in
intravenous (bolus or infusion), intraperitoneal, subcutaneous,
or uscular form, or introduced directly, e.g. by
injection or other methods, into or onto a jprostate cancer
, all using dosage forms well known to those of ordinary
skill in the pharmaceutical arts.
An oligonucleotide which reduces clusterin expression and/or an
AR antagonist of the invention can be administered in admixture
with le pharmaceutical diluents, extenders, excipients,
or carriers (collectively referred to herein as a
pharmaceutically acceptable r) suitably selected with
respect to the intended form of administration and as
consistent with tional pharmaceutical practices. The unit
will be in a form le for oral, rectal, topical,
intravenous or direct injection or parenteral administration.
An oligonucleotide which reduces rin expression and/or an
AR antagonist can be administered alone or mixed with a
pharmaceutically acceptable carrier. This carrier can be a
solid or , and the type of carrier is generally chosen
based on the type of administration being used. Capsule or
tablets can be easily formulated and can be made easy to
w or chew; other solid forms include granules, and bulk
powders. Tablets may contain suitable binders, ants,
diluents, disintegrating agents, coloring , flavoring
agents, flow-inducing agents, and melting agents. Examples of
suitable liquid dosage forms include solutions or sions
in water, pharmaceutically able fats and oils, alcohols
or other organic solvents, including esters, emulsions, syrups
or elixirs, sions, solutions and/or suspensions
reconstituted from non—effervescent es and effervescent
preparations reconstituted from effervescent granules. Such
liquid dosage forms may contain, for e, suitable
solvents, preservatives, emulsifying agents, suspending agents,
diluents, sweeteners, thickeners, and melting agents. Oral
dosage forms optionally n flavorants and coloring agents.
Parenteral and intravenous forms may also include minerals and
other materials to make them compatible with the type of
injection or delivery system chosen.
An oligonucleotide which reduces clusterin expression and/or an
AR antagonist can also be administered in the form of liposome
delivery systems, such. as small unilamellar vesicles, large
allar vesicles, and multilamellar vesicles. Liposomes can
be formed from a variety of phospholipids, such as cholesterol,
stearylamine, or phosphatidylcholines. The compounds may be
administered as components of tissue-targeted emulsions.
For oral administration in liquid, dosage form, ARl may be
combined with any oral, non—toxic, pharmaceutically acceptable
inert r such as ethanol, glycerol, water, and the like.
Examples of suitable liquid dosage forms include solutions or
suspensions in water, pharmaceutically acceptable fats and
PCT/11320122000609
oils, alcohols or other c solvents, including esters,
ons, syrups or elixirs, suspensions, solutions and/or
suspensions reconstituted from non-effervescent granules and
effervescent preparations reconstituted. from. effervescent
es. Such liquid dosage forms may contain, for example,
suitable solvents, preservatives, emulsifying agents,
suspending agents, diluents, sweeteners, thickeners, and
melting agents.
in some embodiments of the invention, the amount of AR
antagonist required for ent of prostate cancer is less in
combination with an oligonucleotide targeting the expression of
clusterin, than would. be required. with AR antagonist
monotherapy.
A dosage unit may se a single compound or Huxtures of
compounds. A dosage unit can be prepared for oral or injection
dosage forms.
According to an aspect of the invention, there is provided an
oligonucleotide which reduces clusterin expression—containing
pharmaceutical composition packaged in dosage unit form,
wherein the amount of the oligonucleotide in each dosage unit
is 640mg or less. Said pharmaceutical composition may include
an AR. antagonist, and. may be in an injectable on or
suspension, which may further n sodium ions.
According to another aspect of the invention, there is provided
the use of an oligonucleotide targeting clusterin expression
and an AR antagonist in the manufacture of a ment for the
treatment of cancer, where the medicament is ated to
deliver a dosage of 640mg or less of oligonucleotide to a
patient. The medicament may contain sodium ions, and/or be in
the form of an injectable solution.
General techniques and compositions for making dosage forms
useful in the present invention are bed in the following
references: 7 Modern Pharmaceutics, Chapters 9 and 10 (Banker &
Rhodes, Editors, 1979); Pharmaceutical Dosage Forms: s
(Lieberman et al., 1981); Ansel, Introduction to Pharmaceutical
Dosage Forms 2nd. Edition (1976); Remington's Pharmaceutical
Sciences, 17th ed. (Mack Publishing Company, , Pa.,
1985); Advances in Pharmaceutical Sciences (David. Ganderton,
Trevor Jones, Eds., 1992); Advances in Pharmaceutical es
Vol 7. (David Ganderton, Trevor Jones, James McGinity, Eds.,
1995); Aqueous Polymeric Coatings for Pharmaceutical Dosage
Forms (Drugs and the Pharmaceutical es, Series 36 (James
McGinity, Ed., 1989); Pharmaceutical Particulate rs:
Therapeutic Applications: Drugs and the Pharmaceutical
Sciences, Vol 61 (Alain d, Ed., 1993); Drug Delivery to
the Gastrointestinal Tract (Ellis Horwood Books in the
Biological Sciences. Series in Pharmaceutical Technology; J. G.
Hardy, S. S. Davis, Clive G. Wilson, Eds.); Modern
Pharmaceutics Drugs and the ceutical Sciences, Vol. 40
(Gilbert S. Banker, Christopher T. Rhodes, Eds.). These
references in their entireties are hereby incorporated by
reference into this application.
This invention will be better understood by reference to the
mental Details which follow, but those skilled in the art
will readily appreciate that the specific experiments detailed
are only illustrative of the invention as described more fully
in the claims which follow thereafter.
W0 2012/123820
Experimental Details
Example 1. Clusterin inhibitor Custirsen together with AR
nist ARI is a potent combination therapy in castrationresistant
te cancer models.
Introduction and Objective
AR and intra-tumoral androgen synthesis are implicated in
promoting tumor cell survival and development of castration—
resistant prostate cancer (CRPC). ARl, has shown ty in
preclinical and clinical studies. Previous studies link
androgen ablation therapy with clusterin upregulation and
castration resistance. The antisense inhibitor, custirsen,
increases cell death when ed with castration or
chemotherapy in prostate cancer (CaP) models. Herein below,
the y of custirsen and ARl combination therapy to delay
progression in a castration—resistant LNCaP model was tested.
Methods
Effects of dual vs ation ARl and custirsen
regimens on AR—positive LNCaP cell proliferation (Figures 1—4,
and 6-7) and al (Figure 5) as well as protein (Figures
9, 11, and 13-15), and gene expression (Figures 13 and 14)
were analyzed using a crystal violet assay, flow cytometry,
western blotting and RT-PCR, respectively. AR transcriptional
activity was measured. by a ciferase reporter assay,
while AR, degradation was assessed. by' a cycloheximide chase
assay. The LNCaP cell line used for experiments was AR
positive.
The effects of combination treatment on castration—resistant
LNCaP tumor' growth. were assessed. in ted. male athymic
nude mice. Male athymic nude mice were inoculated with LNCaP
cells in Matrigel in two sites of mouse flank lesion. The mice
were castrated once tumors reached 150mm3 or the PSA level
increased above SOng/mL. Once tumors ssed to castration
resistance (PSA levels increased to the same level as pre-
tion), 10 mice were randomly assigned to each of ARl +
led antisense oligonucleotide (SCRB) or ARl + custirsen
treatment groups. Custirsen (lOmg/kg/each dose) or SCRB
(10mg/kg/each dose) was injected i.p. once daily for the first
week and then three times per week. AR1 (lOmg/kg/each dose)
was administered orally once daily (morning) 7 days per week
for 8 to 12 weeks. Tumor volume was measured once per week.
Serum PSA was determined weekly. PSA doubling time (PSAdt) and
velocity were calculated by the ope method (PSAt ~—
PSAHHUKLX em). All animal procedures were performed according
to the guidelines of the Canadian Council on Animal Care and
appropriate institutional certification.
The combination of custirsen and ARl more potently suppressed
LNCaP cell growth rates in a dose and time dependent manner
compared to custirsen or AR1 monotherapy (Figures 1—8).
Surprisingly, PARP cleavage (Figure 15), sub GO/Gl apoptotic
fraction (Figures 7 and 8) and repressed AKT phosphorylation
(Figures 10 and 18) was most increased with combined therapy.
Additionally, sen acceleratedv AR degradation (Figures
-19 and 22) and repressed AR transcriptional activity
(Figures 20—22) in combination with_ ARl. In vivo, combined
custirsen and ARl icantly delayed tion—resistant
LNCaP tumor progression and PSA progression (Figures 26—29)
compared to scrambled oligonucleotide control and ARl (p<0.05
and p<0.05 at 12 weeks, respectively).
Conclusions
Custirsen combined with ARl down—regulated AR levels and
ty and suppressed castration—resistant LNCaP cell growth
in vitro and in Vivo, providing pre-clinical proof-of-
principle as a promising approach for AR-targeting therapy in
CRPC.
Example 2. Materials and Methods
Prostate Cancer Cell Lines and Reagents
LNCaP cells were kindly provided. by Dr. Leland. W.K. Chung
(1992, MDACC, Houston Tx) and tested. and. authenticated. by
whole—genome and. whole—transcriptome cing on na
Genome Analyzer IIX platform in July 2009. LNCaP cells were
maintained RPMI 1640 (Invitrogen Life logies, Inc.)
mented with 5% fetal bovine serum and 2mmol/L L—
glutamine. Cells were cultured. in a humidified. 5% COg/air
atmosphere at 370C. Cycloheximide and MG—132 were purchased
from Calbiochem, R1881 (Perkin-Elmer), AR1 (MDV-3100; Haoyuan
Chemexpress Co., Limited). Antibodies: anti-GRP78, anti—CREBZ
(ATF4), CLU C-l8, AR N-ZO, AR 441, PSA C-l9, Ubiquitin, pERK,
B—tubulin and vinculin from Santa Cruz Biotechnology; anti—
phospho-elFZa from Invitrogen Life Technologies; anti—ATF6
from Imgenex Corp; Atg3, LC3, pAkt/Akt, meOR/mTOR,
pp7OS6K/p7OS6K, poly(ADP ribose)polymerase (PARP)form from
Cell Signaling Technology; and anti—Vinculin and —Actin
from Aldrich.
CLU siRNA and antisense treatment
siRNAs were purchased from Dharmacon Research, Inc., using the
siRNA sequence corresponding to the human CLU initiation site
in exon 2 and a le control as previously described
(Lamoureux et al., 2011). —generation antisense
(custirsen) and scrambled (Sch) oligonucleotides with a 2’—O—
(2-methoxy)ethyl modification were supplied by OncoGenex
-43_
WO 23820
Pharmaceuticals. custirsen sequence (5'—CAGCAGCAGAGTCTTCATCAT—
3'; SEQ ID NO: 3) corresponds to the initiation site in exon
II of human CLU. The Sch control sequence was 5'—
CAGCGCTGACAACAGTTTCAT—3’ (SEQ ID NO: 44). Prostate cells were
treated with siRNA or oligonucleotides, using protocols
described previously (Lamoureux et a1., 2011).
Western blotting analysis and immunoprecipitation
Total proteins were extracted using RIPA buffer (50mM Tris, pH
7.2, 1% NP-40, 0.1% deoxycholate, 0.1% SDS, 100mM NaCl, Roche
complete protease inhibitor cocktail) and submitted to n
blot as we described previously (Zoubeidi et al., 2007). For
immunoprecipitation, total ns (500 ug) were pre—cleared
with protein—G sepharose (Invitrogen Life Technologies) for 1
h at 4°C and immunoprecipitated with 2 ug anti—AR, or
globulin G (IgG) as a control overnight at 4°C. The
immune xes were recovered with protein—G sepharose for 2
h and then washed with radioimmunoprecipitation assay buffer
(RIPA) at least , centrifuged, and submitted. to SDS—
PAGE, followed by Western blotting.
Quantitative Reverse Transcription—PCR
Total RNA was extracted from cultured cells after 48 hours of
treatment using TRIzol reagent (Invitrogen Life Technologies,
Inc.). Two ug of total RNA was reversed transcribed using the
Transcriptor First Strand cDNA Synthesis Kit (Roche Applied
e). Real time monitoring of PCR amplification of
complementary DNA (cDNA) was performed using DNA primers
(supplemental table) on ABI PRISM 7900 HT Sequence Detection
System (applied Biosystems) with SYBR PCR Master Mix (Applied
Biosystems). Target gene expression 5'-TACCAGCTCACCAAGCTCCT—3'
(forward; SEQ ID NO: 45) or 5’-GCTTCACTGGGTGTGGAAAT-3'
(reverse; SEQ ID NO: 46) targeting human AR, 5'—
CACAGCCTGTTTCATCCTGA-S' rd; SEQ ID NO: 47) or 5'—
AGGTCCATGACCTTCACAGC—B' (reverse; SEQ ID NO: 48) targeting
human PSA, were normalized to b- actin levels using 5'—
GGCACCACACCTTC-B' (forward; SEQ ID NO: 49) or 5'-
AGCACTGTGTTGGCGTACAG—3' (reverse; SEQ ID NO: 50) as an
internal rd, and the comparative cycle threshold (Ct)
method was used to calculated relative quantification of
target mRNAs. Each assay was performed in triplicate.
Immunofluorescence
LNCaP cells were grown on coverslips and transfected with CLU
siRNA or control. 48 hours post ection cells were
treated, with lOuM. of ARI i 3. nM iRl88I for 6 'hours. After
treatment, cells were fixed in ice-cold. methanol completed
with 3% acetone for 10 min at —200C. Cells were the washed
thrice with PBS and incubated with 0.2% Triton/PBS for 10 min,
followed. by washing' and. 30 min blocking“ in 3% nonfat Inilk
before the addition of antibody overnight to detect AR
(1:250). Antigens were visualized, using anti-mouse antibody
coupled with FITC (1:500; 30 min). Photomicrographs were taken
at 20X magnification using Zeiss Axioplan II fluorescence
microscope, followed by analysis with imaging software
ern Eclipse, Empix Imaging, Inc.).
AR transcription activity
LNCaP cells were seeded. at a density of 5XI04 in 12—well
plates and. transfected. the following day‘ with. custirsen or
SCRB. The next day cells were transfected with sen or
SCRB together with ciferase (PSA-Luc) reporter (-6,100
to +12) and Renilla—luciferase plasmid using Lipofectin
t (1.5uL per well; Invitrogen), as described previously
(Sowery et al., 2008). After 24 h, the medium was replaced
with RPMI (Invitrogen) containing 5% charcoal-stripped serum
(CSS), mented with l nmol/L R1881 or ethanol vehicle and
umol/L of ARl or DMSO for 48 h. Cells were harvested, and
luciferase activity measured, as before (Sowery et al., 2008).
Reporter assays were normalized. to Renilla and luciferase
activity expressed by Firefly to Renilla ratio in ary
light units. All experiments were carried out in triplicate
wells and ed five times using different preparations of
plasmids.
Cell proliferation and cell cycle assays
Cultured cells were transfected with CLU or SCR siRNA,
custirsen or SCRB, and then treated with ARl or DMSO l
24h. after ection. After a time course exposure, cell
growth was measured by crystal Violet assay as previously
bed (Gleave et al., 2005). Detection and quantitation of
apoptotic cell cycle population were analyzed by flow—
cytometry (Beckman Coulter Epics Elite; Beckman, Inc.) based
on 2N and 4N DNA content as previously described (Lamoureux et
al., 2011). For CSS condition, LNCaP cells were plated in RPMI
with 5% FBS switched to C88 at the next day, and treatment was
started same as FBS condition. Each assay was done in
triplicate three times.
Protein stability and degradation
To assess the effect of combination treatment on AR protein
stability, LNCaP cells treated. with custirsen or SCRB were
changed 48 h later to RPMI + 5% serum containing 10 umol/L of
cycloheximide and 10 umol/L of ARl incubated at 37 °C for 2 to
6 or 16 h and western blot was done using AR and vinculin
WO 23820
antibodies. Degradation was tested in LNCaP cells by a 6—hour
incubation with RPMI+5% FBS media containing 10 umol/L of
MGl32 and 10 umol/L ARl 24 hours after siRNA or ABC
transfection. Western blot was done using AR. and. vinculin
antibodies.
Determination of increased efficacy of combination therapy
Crystal violet assay was d to analyze cell growth
inhibition for each single drug or their combination. LNCaP
cells were treated. with. 10 nmol/L CLU siRNA. or SCR siRNA
combined with tion dose of ARl and 500 nmol/L custirsen
or SCRB as well. Next, cells were d for two consecutive
days with dose escalating custirsen or oligofectamime only,
and one day later treated with indicated concentration of ARl
or DMSO for 48h. The data was inputted in CalcuSyn® software,
and the dose effect curve drawn for each treatment to
ate the ation index (CI) at several effective
doses (CI=1; additive effect, CI<1; combination effect, CI>l;
antagonistic effect).
Animal treatment
Male athymic mice (Harlan Sprague—Dawley, Inc.) were injected
s.c. with leO6 LNCaP cells. When tumors grow in 150mm3 and
serum PSA was >50ng/ml, mice are castrated. Once tumors
progressed to castrate resistance, mice were randomly assigned
to ARl plus either 10 mg/kg custirsen or SCRB i.p. once daily
for 7 days and. then three times per‘ week thereafter. Each
mental group consisted of 13 mice. Simultaneously, mice
were treated with ARl once daily p.o., lOmg/kg/each dose for 7
days per week. Tumor volume and serum PSA was Heasured as
previously described (Sowery et al., 2008). All animal
procedures were performed according to the guidelines of the
Canadian Council on Animal Care. Each three mice xenografts
were sacrificed at 7 days after start treatment and the rest
were harvested the end of the study and snap—frozen in liquid
nitrogen. Protein extraction was done by soliciting tumors in
RIPA buffer with protease inhibitor and total cell lysate was
used to assess AR and clusterin expression within the
xenografts and referenced for B—tubulin as described in the
section on Western blotting.
Statistical analysis
All results are expressed as the average i SE. Two-tailed t—
tests, y ANOVA or Wilcoxon matched—pairs tests were used
for statistical is. Combination effects were calculated
by CalcuSyn software. The differences between single treatment
and combination treatment was analysis by Freidman test and
done with. JMP version 4; *P<0.05, **P<0.0I, and. ***P<0.00I
were considered significant.
Example 3. CLU is highly expressed in ARI ant cells and
xenografts
ARI is a novel anti-androgen which binds the AR LED and
ts the growth of castration—resistant afts (Tran
et al., 2009). Data from phase II and III trials show that
ARI is active in both pre- and post—chemotherapy—treated
patients and. decreases levels of PSA. and. ating" tumor
cells (Scher et al., 2010) (Sher, GU—ASCO, 2012).
Unfortunately, like first line hormone therapies, CRPC—LNCaP
afts evolved mechanisms of resistance after the addition
of ARI to tion. CLU was found to be up—regulated in ARI
resistant tumors compared to vehicle treated tumors by western
blot (Fig. 41A, left panel) and immunohistochemistry (Fig. 41A
right panel, Fig. 30A), suggesting that ARI treatment induces
stress activated molecular chaperone CLU in CRPC tumors
2012/000609
similar‘ to that seen with. castration in castrate sensitive
tumors. To facilitate study of mechanisms of ARl recurrence,
different cell lines were created from xenograft tumors
maintained under ARl treatment that were resistant to ARl, and
found. that these ARl resistant cells also expressed. higher
levels of CLU compared to CRPC tumors (Fig. 30B). These data
indicate that increased CLU is associated with pment of
the ARl recurrence phenotype.
Example 4. AR pathway inhibition induces and CLU
Both ARl antisense ches were used to confirm whether CLU
is induced by AR pathway inhibition. Compared to bicalutamide
and androgen deprivation using CSS, ARl induces CLU in both a
time— and dose~dependent , in parallel with reduced AR
activation indicated by decreased PSA expression. To further
te whether ARl ion of CLU is AR dependent, 2
different nse sequences ing the first exon in AR
potently down-regulated AR in a dose-dependent and sequence—
specific manner in LNCaP cells, in parallel with induction of
CLU (Fig. 41C). Together these data suggest that AR pathway
inhibition by androgen deprivation, AR LBD antagonism, or
nse knockdown induces CLU, possibly as part of an
adaptive stress response.
Clusterin expression is up—regulated by ARl treatment.
Clusterin expression is up-regulated in a time and dose
dependent manner after ARl treatment. LNCaP Cells are treated
for different durations and with different concentrations of
ARl in RPMIl64O media with 5% FBS. Cells are harvested and
performed for western blot analysis. Clusterin protein
sion is up regulated in a time and dose dependent
manner. Androgen depleted treatment enhances Clusterin
expression, ally in AR1. LNCaP Cells are treated with 10
umol/L of Bicalutamide or AR1 for 48hrs in RPMIl64O media with
% FBS or 5% CSS (charcoal striped serum; terone
depleted media). Clusterin sion is strongly induced by
anti-androgen treatment or CSS condition. Additionally,
clusterin protein expression strongly increases in AR1
treatment compared to bicalutamide treatment in western blot
analysis. The combination of ARl and custirsen is more
effective at reducing te cancer cell proliferation than
the combination of tamide and custirsen.
Example 5. ARI induces ER stress
Whether AR1 induces ER stress with increase of CLU was
evaluated since molecular chaperones like CLU are ant in
regulating Inisfolded. protein. and. endoplasmic reticular (ER)
stress responses d et al., 2007), and many anti-cancer
agents are known to induce ER stress. ER stress activates a
complex intracellular signaling pathway, called the unfolded
protein response (UPR), which is ed to re—establish
protein homeostasis (proteostasis) by inhibiting protein
translation and. promoting' ER—associated. protein degradation
via the ubiquitin-proteasome system (UPS). ARl is found to
induce CLU expression itant with. up—regulation of ER
stress markers such as GRP78, ATF4, IREl, CHOP and cleaved-
ATF6, tent with ER stress and UPR activation.
Example 6. ARl—induced CLU is mediated by p90Rsk—YB—1
signalling pathway
YB-l binds to CLU promoter leading to increased CLU expression
after ER stress (Shiota et al., 2011). Since ARl can activate
Akt and Erk signalling (Carver et al., 2011), it was
postulated that AR1 mediated activation of Akt (Evdokimova et
al., 2006) and Erk ford et al., 2008) pathways leads to
o-activation and nuclear translocation of YB—l
(Evdilomova et al., 2006), with up-regulation of CLU and
inhibition of stress—induced apoptosis. Figure 42B confirms
that ARl treatment ses Akt and p9ORsk phosphorylation,
which was accompanied with increased phospho—YB—l levels (Fig.
42B). YB—l knockdown using siRNA in ation with ARl
treatment abrogates ARl—induced CLU both at the n and
mRNA levels (Fig. 42C) suggesting' that ARl—induced. CLU is
mediated by YB-l.
Since YB—l can be phosphorylated by both Akt and p9ORsk
(Evdilomova et al., 2006) (Stratford et al., 2008), LY294002
was used to inhibit Akt and SL0101 was used to inhibit p90Rsk
to further define the predominant pathway mediating ARl
induced up—regulation of CLU. Inhibition of Akt did not affect
ARl induced up—regulation of CLU (Fig. 42D); in contrast,
inhibition of p90Rsk using SL101 abrogates ARl-induced. CLU
(Fig. 42E). Without wishing to be bound by any scientific
theory, collectively these data indicate that the p90Rsk—YB—1
pathway is required for ARl induced CLU expression.
Example 7. The combination of CLU inhibition and ARI ses
inhibition of LNCaP cell growth compared to CLU inhibition or
ARl monotherapy.
Whether CLU knockdown potentiates the anti—cancer activity of
ARl was evaluated, because R drugs (July et al., 2002)
like ARl (Figs. 30 and 41) induce up-regulation of CLU and CLU
functions as a or in treatment resistance (Zoubeidi et
al., 2010b; Gleave et al., 2005). LNCaP cells were treated
with custirsen and subsequently treated with indicated
concentrations of ARl. Custirsen significantly enhanced ARl
activity, reducing cell ity compared with control Sch
plus ARl in both time- (Fig. 43A left panel) and dose— (Fig.
43A right panel) dependent s. To determine whether this
effect was additive or a ation effect, the dose—
dependent effects with constant ratio design and the CI values
were calculated according to the Chou and Talalay median
effect pal (Chou et al., 1984). Figure 6B shows the
dosesresponse curve (combination or monotherapy with custirsen
or ARl) along side the CI plots, indicating that the
combination of sen with, ARl has enhanced. effects on
tumor cell growth (Fig. 6C, right panel). The ation of
custirsen and. ARl also had increased efficacy’ at reducing
viability of AR positive castrate resistant C4~2 and
custirsen—resistant cells compared to ARl or custirsen
monotherapy, but not in AR negative PC3 cells.
Flow cytometric analysis indicates sen significantly
increases (P < 0.001) ARl induced apoptosis (sub—Gl fraction)
when ed with ARl (30%) compared with control Sch
(15.2%), custirsen (20%), control Sch or ARl (18.3%) (Fig.
43C). In addition, combination sen plus ARl increases
caspase—dependent apoptosis compared. with ARl or custirsen
monotherapy, as shown by cleaved PARP and caspase—3 activity.
Collectively, these data indicate that the combination of
custirsen and ARl induced apoptosis more than custirsen or ARl
monotherapy.
Example 8. Clusterin knock down combined with ARl treatment
mostly enhances cell growth inhibition and apoptosis in AR
positive LNCaP cells.
LNCaP cells are seeded in 12—well culture plates in 5x104
cells per well with 5% FBS or 5% CSS containing RPMI medium.
The next day, cells are transfected with lOnmol/L of CLU siRNA
WO 23820
or SCR siRNA control at once and also daily with 500 nmol/L of
custirsen or SCRB control for 2 days. The next day post
ect with siRNA or antisense oligo, LNCaP cells are
treated with /L of ARl and cell growth assays are
med on day 0, 1, 2, 3, 4 by crystal violet assay. (day
of ART treatment defined as 100%). CLU knockdown + ARl
ation treatment most significantly repress cell growth.
pmol/L of ARl is combined with 10 nmol/L of CLU siRNA; lO
umol/L of ART combined with 500 nmol/L of custirsen.
Combination ent enhances LNCaP apoptosis in flow
cytometry analysis. Cells are treated with lOnmol/l; of CLU
siRNA or SCR siRNA control at once and also daily with 500
nmol/L of custirsen or SCRB control for 2 days in 5% PBS
containing RPMI medium. The next day post transfect with siRNA
or antisense oligo, LNCaP cells are treated with lOumol/L of
ARl and FACS analysis are performed after 48hrs treatment. The
proportion of cells in sub—GO, GO-Gl, S, G2—M is ined by
propidium iodide staining. Combination treatment increases
Sub—GO/l apoptotic fraction apoptosis in LNCaP cells. p<0.01
in combination CLU siRNA with ART and p<0.001 in combination
custirsen with ARl relative to oligofectamime and DMSO
control. P value represents between treatment arms and their
respective controls *p<0.05, **p<0.01, ***p<0.001 (Wilcoxon
matched~pairs test).
Combination ent enhances apoptosis. LNCaP cells are
pretreated with 10 umol/L of ARl for 48 h before treatment
with CLU’ or SCR‘ siRNA. and. custirsen. or SCRB control. PARP
cleavage expression levels are measured by Western blot. All
experiments are repeated at least thrice.
Example 9. The combination, of custirsen ‘with. ARl ent
shows increased efficacy compared to custirsen or ARl
monotherapy.
Inhibition of growth is observed in LNCaP cells treated with
CLU siRNA or sen combined with ARl in Vitro. Cells are
transfected with lOnmol/L of CLU siRNA or SCR siRNA control at
once and. also daily' withv 500 nmol/L of sen or SCRB
control for 2 days in 5% FBS RPMI medium. The next day post
transfect with siRNA. or antisense oligo, LNCaP cells are
treated with various concentrations of ARl. Three days after
treatment, cell viability is determined by crystal violet
assay. Viable cell density' is normalized. to that of cells
treated at DMSO control (ARl compound is dissolved in DMSO and
adjusted. indicated. concentrations). The ation of
sen with ARl shows increased efficacy at reducing cell
viability compared to custirsen or ARl monotherapy. Data
points are means of triplicate is. P value represents
between treatment arms and their respective controls *p<0.05,
**p<0.01 (Student’s t-test).
Cell growth inhibition is evaluated for each single drug or
their combination by crystal violet assay. LNCaP cells are
treated with variable concentration of ARl or custirsen. There
is a significant difference between each single treatment and
those combinations. P value is ated by Friedman test.
The data are input and calculated by CalcuSyn software®.
r Combination index (CI) at several effective dose.
CI=1; additive effect, CI<l; combination effect, CI>l;
nistic effect. These data indicate the combination
effect at combination treatment.
W0 2012/123820
Example 10. AR and PSA sion in combination treatment.
AR protein expression decreases after CLU knockdown using
custirsen combined with ARl. LNCaP Cells are ected with
lOnmol/L of CLU siRNA or SCR siRNA control at once and also
daily with 500 nmol/L of custirsen or SCRB l for 2 days
in 5% FBS RPMI medium. The next day post transfect with siRNA
or antisense oligo, LNCaP cells are d with /L of
ARl. 48hrs later, cells are ted for protein and mRNA.
The n expression is analyzed by western blot. CLU
knockdown combined with ART treatment has enhanced potency in
decreasing' AR expression compared to monotherapy. AR
expression is strongly repressed by CLU knockdown with ARl.
Cells are treated. with. lOnmol/l: of CLU siRNA or SCR siRNA
combined with lOpmol/L of ARl or bicalutamide. AR expression
is detected by western blot. Combination treatment does not
affect AR mRNA level. mRNA expression is analyzed by
quantitative RT—PCR, AR and PSA levels are normalized to
levels of B-actin mRNA and expressed as mean i SE. **P<0.0l
***p<0.001 (Wilcoxon matched—pairs test). “OTR” means cells
treated with oligofectamime only. OTR and DMSO d cells
were defined as 1009.
Example 11. Combination ARI plus custirsen has increased
efficacy in delaying CRPC LNCaP tumor growth
The in vivo effects of co—targeting‘ the AR and. the stress
response using‘ ed custirsen with ARl were evaluated.
Male nude mice bearing LNCaP xenografts were castrated when
serum PSA reached 75ng/ml and followed until serum PSA and
tumor growth rates increased. back to pre—castrate levels,
indicating progression to castration resistance. Mice were
then randomly assigned for treatment with ARl plus either
control Sch (n 10) or custirsen (n=lO). At baseline, mean
LNCaP tumor volume and serum PSA levels were similar in both
groups. Custirsen significantly enhanced the mor effect
of ARl, reducing mean tumor volume from 1600 mm3 to 650 mm3 by
12 weeks (**; pS0.05), compared to control Sch (Fig. 44A).
Overall survival (defined as asia for tumour volume
exceeding 10% of body weight) was significantly prolonged with
combined ARl + custirsen compared with ARl + Sch control (90%
vs 30% at week 16, tively; *; pS0.05). Serum PSA levels
were also significantly lower (~4—folds) (Fig. 44C), and PSA
doubling time is significantly prolonged (*, p<0.05) in the
custirsen + ARl group (***, p<0.001) ed with ARl control
group. To evaluate the pharmacodynamics effects of combination
treatment on target n levels, western blot analysis from
tumour tissue (3 animals each) was performed for AR, PSA and
CLU. Fig. 44D illustrates that AR and CLU expression levels
were reduced in combination-treatment tumour tissue compared
to ARl controls. Collectively, these data demonstrate that
co—targeting the AR and the resultant CLU-regulated stress
response potentiates the effects of ARl in a human CRPC
xenograft model.
The efficacy of sen and ARl combination y is
enhanced compared to ARl or custirsen erapy in a CRPC
xenograft model. Figure 44A~B shows the effect on tumor volume
and serum PSA level by combination treatment.
Example 12. Combination ARl plus custirsen has increased
efficacy in CRPC xenograft model.
LNCaP cells are inoculated s.c. into athymic nude mice. When
xenografts grow to ~500 mm3, or PSA >50 ng/ml mice are
castrated. Treatment is started when PSA levels increased to
pre—castration . Custirsen or SCRB are injected i.p.
—56-
PCT/11320121000609
once daily for 1 week and then 3 times/week fter. ARl is
administrated once daily. Total LNCaP xenograft proteins are
extracted in. RIPA, buffer after custirsen. or SCRB treatment
combined with ARl. (three mice per group) and Western blots
are done with AR, PSA, and CLU antibodies; vinculin is used as
a loading control. ulin ratio is calculated. ation
ARl plus sen has increased efficacy at prolonging
survival in the CRPC xenograft model.
Example 13. Combination .ARl plus CLU silencing reduces AR
nuclear translocation and transcriptional ty more
effectively than ARl or CLU silencing monotherapy.
The in vivo study in Figure 44 shows that custirsen in
combination with. ARl induces rapid. decrease in PSA, before
changes in tumor volume became nt; in addition, AR
protein levels appeared lower in the combination treated
tumors compared to AR1 alone treated tumors, suggesting that
CLU own might iate AR targeting and modulate AR
signalling pathway. The effects of combination treatment on
androgen—induced, AR—mediated gene activation were evaluated.
LNCaP cells were treated with ARl or custirsen alone or in
combination and evaluated for changes in R1881 stimulated PSA
transactivation (Fig. 45A). As expected, ARl reduced R1881
d AR transcriptional activity, as measured by PSA
luciferase transactivation assay, by 95%; interestingly
custirsen also reduced AR activity by 90%, and this effect was
enhanced in combination with ARl suggesting that CLU knockdown
potentiates ARl inhibition of AR activity. To define how CLU
can affect AR riptional activity, the effect of CLU
knockdown i ARl on ligand—induced AR nuclear translocation was
evaluated. As expected, while AR nuclear translocation was
decreased by AR1, CLU knockdown in combination with ARl
maximally inhibited R1881—induced AR nuclear translocation
(Fig 45B). This co-targeting inhibitory effect was also
confirmed by fractionation assay showing that targeting CLU in
combination with ARl inhibits Rl88l-induced nuclear AR levels
(Fig. 45C).
Example 14. CLU knockdown combined with ARl treatment
rates AR degradation via the proteasome pathway.
To investigate the fate of AR after combination treatment, the
effect of CLU knockdown combined with ARl on AR expression was
evaluated both at the protein and mRNA levels. CLU ing
using siRNA (Fig. 46 A right upper panel) or custirsen (Fig.
46 A left upper panel) resulted in decreased AR protein, but
not mRNA levels (Fig. 46 lower panel) only in combination with
ARl, suggesting that CLU knockdown in combination with ARl may
affect AR stability. AR n stability was then evaluated
using eximide, which inhibits protein synthesis. AR
protein levels decreased significantly with rapid degradation
after custirsen—induced CLU knockdown ed with ARl (Fig.
46B), suggesting that CLU knockdown leads to AR instability
when complexed with ARl.
AR forms a dimer x with Hsp90 to provide stability
for ligand—unbound. AR. Indeed, without Hsp90 g, the
unfolded n will be recognized and degraded by the
ubiquitin-proteasome system (Solit et al., 2003; Zoubeidi et
al., 2010c). Whether ARl affects AR binding- to Hsp90, and
subsequent effects if combined with CLU silencing was first
evaluated. ARl treatment actually increases AR-Hsp90
interactions, consistent with prior reports that ARl
sequesters AR in the cytoplasm. Interestingly, CLU knockdown
in combination with ARl decreases the ation between AR
~58—
W0 23820
and Hsp90, as shown in Figure 46C. Without wishing to be bound
by any scientific theory, these data are consistent with a
view that ARl—AR—Hsp90 complex becomes more vulnerable
to degradation under conditions of CLU silencing. To assess
whether~ AR degradation. under these conditions involves the
ubiquitin—proteasome system, levels of ubiquitinated AR were
measured under conditions of mono- or after combination
therapy, and as shown in Figure 46D, AR ubiquitination levels
were highest under co—targeted combination conditions. AR
protein levels were next evaluated in the presence or e
of proteasome inhibitor (MG132) to characterize role of
proteasome and MG132 was found. to te AR. degradation
under conditions of CLU knockdown plus ARl, implicating* AR
degradation via the proteasome (Fig. 46E). t wishing to
be bound by any scientific theory, taken together these data
suggest CLU knockdown accelerates AR ation via a
some mediated pathway preferentially when the AR is
bound to ARl.
Example 15. AR degradation rates are accelerated by
ation treatment.
Combination treatment rapidly decreases AR expression. LNCaP
cells are treated with 500 nmol/L of custirsen or SCRB control
and then treated, with. 10 umol/L of ARl and. 10 umol/L of
cycloheximide various time periods. DMSO is used as control.
AR protein levels are measured by Western blot analysis. CLU
knockdown combined with ARl accelerates proteasomal
degradation of AR. LNCaP cells are treated with CLU siRNA or
SCR siRNA and custirsen or SCRB control, and then treated with
pmol/L of ARl and 10 umol/L MG-132 for 6h. DMSO is used as
control. AR protein levels are measured by n blot
analysis.
W0 2012/123820
Example 16. Combination treatment effects AR ubiquitination.
LNCaP cells are treated with 10 nmol/L of CLU siRNA or SCR
siRNA control in the presence of FBS and then d with 10
umol/L of ARl and 10 umol/L of . Immunoprecipitation is
done using anti—AR antibody , and Western blot analysis
is done using anti—AR antibodies (441) or anti—Ubiquitin
antibodies. Input is blotted with AR (N-ZO) antibody. Without
wishing to be bound. by any scientific , combination
treatment facilitates proteasomal degradation of AR via
ubiquitination of AR.
Example 17. CLU knockdown decreases levels of molecular co—
chaperones involved in AR stability
Without wishing to be bound by any scientific theory, the data
herein tes that ARl monotherapy sequesters AR—Hsp90
complexes in the cytoplasm; however when combined. with CLU
knockdown the AR—Hsp90~ARl heterocomplex becomes destabilized,
leading to AR ubiquitination and ation, and reduced AR
nuclear transport and. activity. One explanation is the CLU
inhibition may lower Hsp90 levels through its affects on HSF-l
regulation (Lamoureax et al. 2011); however ation
therapy' did. not significantly* lower Hsp90 levels, and. data
illustrated in Figure 42 implicates YB-l as the key stress-
activated transcription factor mediating ARl increases in CLU.
Since Hsp90 functions in cooperation with co—chaperones to
confer ity of client proteins, an unbiased approach was
initially used to identify Hsp9O perone affect by CLU
expression. The gene profiling analysis from LNCaP cells and
PC—B treated with l and CLU siRNA disclosed herein shows
that CLU expression correlated. with the Hsp90 co-chaperone
FKBP52 (Hsp56). Western blotting was used to confirm that CLU
knockdown. reduces FKBP52, but not FKBPSl or‘ Hsp90, protein
levels (Fig. 47A). To ascertain. the role of FKBP52 in. AR
ity under conditions of ARl treatment and CLU silencing,
FKBP52 was overexpressed after CLU knockdown and AR expression
was evaluated. Figure 47B shows that FKBP52 rescues AR from
degradation induced by CLU knockdown and ARl treatment. FKBP52
overexpression also partially restores PSA expression,
indicating increased AR activity when FKBP52 levels are
ed under conditions of CLU knockdown. These data suggest
that CLU knockdown in combination with ARl induces AR
degradation by affecting FKBP52 levels and the stability of
the AR-co-chaperone complex.
To further define how CLU regulates FKBP52 levels under
conditions of ARl induced stress, public databases were mined
and provided information ting that YB—l binds to FKBP52
with high stringency of 12 in ChIP on ChIP is. Western
blotting confirmed that YB—l knockdown ses FKBP52
expression levels (Fig. 47C). ARl treatment activates a stress
response involving' YB—l transactivation. of CLU} as well as
increased Akt and p90rsk activity (Fig 42). Since YB~l
knockdown decreases both CLU and FKBP52 levels, and CLU can
also enhance p—AKT activity, how CLU knockdown affects
interactivity between YB-l, AKT and p90rsk to affect FKBP52
levels under conditions of ARl treatment was igated
next. Interestingly, similar to previous s that CLU can
enhance AKT phosphorylation, CLU knockdown was found to also
abrogate ARl induced phosphorylation of YB—l and p9ORsk (Fig.
47D). Without wishing to be bound by any scientific theory,
since the p9ORsk—YB-l pathway is the key regulator for ARl
induced CLU sion (Figure 42), collectively these data
identify an ARl treatment—induced feed forward loop involving
pYB—l, p90rsk, CLU, and the AR co—chaperone FKBPSZ for AR
stability, nuclear translocation and activation.
Example 18. ation treatment inhibits Akt/mTOR signalling
pathway.
ARl activates phosphorylatior of Akt and ERK. LNCaP cells are
treated with ARl in media with 5% PBS at various time periods
and doses. Western blot analysis is done using phospho Akt,
phospho ERK, Akt and ERK antibodies. CLU knockdown attenuates
Akt/mTOR signalling pathway through inhibition of phospho Akt
activation by ARl treatment. LNCaP cells are treated with 10
nmol/L CLU siRNA or SCR siRhA l in the ce of PBS
and then treated with 10 umol/L of ARl. After 48h treatment,
n blot analysis is done using phospho Akt, phospho ERK,
phospho mTOR, phospho P7OS6K, Akt, ERK, mTOR and P7OS6K
antibodies.
Example 19. Possible explanation of combination effect n
clusterin knockdown and ARl for AR ve state.
Androgen binding to AR leads to rapid translocation from
cytoplasm to nucleus and it leads to enhance tion of AR—
regulated genes. Clusterin up-regulates the AKT/mTOR pathway
and it leads to cell survival, cell proliferation and cell
growth. Custirsen induced. clusterin knockdown represses AKT
phosphorylation and. attenuates androgen transportation from
cell surface via repressing megalin expression. ARl ly
binds to AR and inhibits its translocation to nucleus. Without
wishing to be bound by any scientific theory, these results
lead to accelerate AR proteasomal degradation and down—
regulated mTOR signalling pathway.
—62-
Example 20. Clusterin knock down combined with ARl treatment
mostly enhances cell growth inhibition and apoptosis in C4-2
cells, but does not have a combination effect in AR negative
PC—3 cells.
C4—2 cell growth is evaluated upon combination treatment. C4-2
cells are seeded in 12~well culture plates in 3x104 cells per
well with 5% PBS or 5% CSS containing RPMI medium. The next
day, cells are transfected with lOnmol/L of CLU siRNA or SCR
siRNA control. The next day post ect with siRNA, C4-2
cells are treated with 10 umol/L of ARl and cell growth assays
were performed on day 0, l, 2, 3, 4 by crystal violet assay.
(day of ARl treatment defined as 100%). CLU own and ARl
combination treatment represses cell growth most
significantly. PC—3 cell growth is evaluated upon combination
treatment. PC—3 cells are seeded in 12—well culture plates in
3x104 cells per well with 5% PBS containing DMEM medium. The
next day, cells are transfected with lOnmol/L of CLU siRNA or
SCR siRNA l at once and also daily with 500 nmol/L of
custirsen or SCRB control for 2 days. The next day‘ post
transfect with siRNA or antisense oligo, PC—3 cells are
treated. with. 10 nmol/L of ARl and cell growth assays are
performed on day 0, l, 2, 3 by crystal violet assay. (day of
ARl treatment defined as 100%). Combination treatment enhances
LNCaP apoptosis in flow cytometry analysis. Cells are treated
with /L of CLU siRNA or SCR siRNA control at once and
also daily with 500 nmol/L of custirsen or SCRB l for 2
days in 5% FBS containing RPM; . The next day' post
transfect with siRNA or antisense oligo, LNCaP cells are
treated with lOumol/L of ARl and FACS analysis were performed
after 48hrs treatment. Proportion of cells in sub—GO, GO—Gl,
S, GZ—M was determined by propidium iodide staining.
Combination ent increases Sub-GO/l tic fraction
~63-
apoptosis in LNCaP cells. lCa: ed with CLU siRNA, le:
combined with custirsen.
Example 21. Clusterin and AR mRNA expression is up—regulated
in a time and dose dependent manner after ARI treatment.
LNCaP Cells are treated with deferent time and different
concentration of AR1 in RPM1164O media with 5% FBS. AR1 is
treated at various concentrations and exposure times. Cells
are harvested and ed for mRNA level by quantitative RT—
PCR. AR and CLU levels are normalized to levels of B—actin
mRNA and expressed as mean f SD. ARl exposure time of O h and
dose of Onmol/L defined as 100%. *P<0.05 **P<0.01 ***p<0.001
(Wilcoxon matched-pairs test).
AR protein expression decreases after CLU knockdown combined
with, ARl in both androgen depleted. and. androgen stimulated
cells. LNCaP Cells are transfected with 10nmol/L of CLU siRNA
or SCR siRNA control in 5% CSS with or without 1 nmol/L of
R1881 ning RPMI . The next day post transfect with
siRNA, LNCaP cells are treated with /L of AR1. 48hrs
later, cells are harvested for protein. The protein sion
is analyzed by Western blot. CLU knockdown combined with AR1
treatment decreases AR expression with greater efficacy than
CLU knockdown alone or ARl monotherapy.
Discussion
In prostate cancer, the androgen receptor (AR) continues to
drive te ant progression after castration. While
new AR pathway inhibitors like ARl prolong al in CRPC,
resistance rapidly develops and is often associated with re~
activation of AR signalling and induction of the
cytoprotective one, clusterin (CLU). Since adaptive
stress pathways ted. by treatment can facilitate
development of ed treatment resistance, co-targeting the
stress response activated by AR inhibition, and mediated
through CLU, may create conditional lethality and e
outcomes. The data herein show that co—targeted the AR and
stress—induced CLU by combining ARl with custirsen, and
defined mechanisms of combination activity using the castrate—
resistant LNCaP model.
ARl induced markers of ER stress markers and chaperone
proteins, including CLU, as well as the AKT and MAPK
signalosome. This stress response was coordinated by a feed
forward loop involving p-YB—l, p90rsk, and CLU. Combination
CLU knockdown plus ARl suppressed LNCaP cell growth rates by
enhancing apoptotic rates over that seen with ARI or custirsen
monotherapy. In vivo, combined custirsen + ARl significantly
delayed castration—resistant LNCaP tumor progression and PSA
progression. istically, ARl d AR cross talk
activation of AKT and MAPK pathways was repressed with
combined therapy. Interestingly, CLU knockdown also
accelerated. AR. degradation and repressed. AR transcriptional
activity when combined with ARl, through mechanisms involving
decreased HSF—l and YB—l regulated expression of AR co—
chaperones FKBPSZ.
Co—targeting adaptive stress ys activated by AR y
inhibitors, and mediated through CLU, creates conditional
lethality and es mechanistic and inical proof—of—
principle to guide biologically rational combinatorial
clinical trial design.
Prostate cancer is the most common solid malignancy and second
leading cause of cancer deaths among males in Western
countries (Siegel et al., 2011). While early-stage disease is
treated with curative surgery or radiotherapy, the mainstay of
ent for locally ed, recurrent or metastatic
prostate cancer is androgen ablation therapy, which reduces
serum testosterone to castrate levels and suppresses androgen
receptor (AR) activity. Despite high initial response rates
after androgen on, progression to castrate resistant
prostate cancer (CRPC) occurs within 3 years (Gleave et al.,
2001; vsky et al., 2000; berg et al., 1999;
Goldenberg et al., 1996; Gleave et al., 1998; Bruchovsky et
al., 2006). Over 80% of CRPC specimens express the AR and
androgen—responsive genes (Chen et al., 2004), indicating that
the AR axis remains activate despite castration. Hence, the
AR is a key driver of CRPC, and is supported by treatment—
activated growth factor signalling ys (Miyake et al.,
2000), survival genes (Miyake et al., 1999), and
cytoprotective chaperone networks (Rocchi et al., 2004).
Docetaxel chemotherapy (Petrylak et al., 2004) was the first
therapy to prolong survival in CRPC, stratifying the treatment
landscape into pre— and post—chemotherapy states. More
recently, two new AR pathway tors, the CYP17 inhibitor
abiraterone (de Bono et al., 2011) and the AR antagonist ARl
(Tran et al., 2009), have produced promising survival gains
and are rapidly changing the CRPC landscape. Despite
—66-
W0 2012/123820
significant responses (Tran et al., 2009; Harris et al., 2009;
Scher et al., 2010), abiraterone and ARl activate redundant
survival pathways that vely drive treatment resistance
and recurrent CRPC progression. Realization of the full
potential of these novel AR pathway inhibitors will require
characterization of these stress—activated survival responses,
and rational combinatorial co—targeting strategies ed to
abrogate them.
Molecular chaperones play central roles in stress responses by
maintaining protein homeostasis and playing prominent roles in
signalling and transcriptional regulatory networks. Clusterin
(CLU) is a stress~activated. chaperone originally cloned as
“testosterone—repressed) prostate message 2” (TRPM—Z)
(Montpetit et al., 1986) from post—castration regressing rat
te, but was subsequently defined as a stress-activated
and apoptosis-associated, rather than an androgen—repressed,
gene (Cochrane et al., 2007). CLU is transcriptionally
regulated by HSFl (Lamoureux et al., 2011) and YB—l (Shiota et
al., 2011), ting —induced sis by suppressing
protein ation (Poon et al., 2002), p53—activating stress
signals (Trougakos et al., 2009), and conformationally—altered
Bax (Zhang et al., 2005; Trougakos et al., 2009) while
enhancing Akt phosphorylation (Ammar et al., 2008) and trans—
activation of NF-KB and HSF—l (Lamoureux et al., 2011; Shiota
et al., 2011; Poon et al., 2002; Trougakos et al., 2009; Zhang
et al., 2005; Ammar et al., 2008; Zoubeidi et al., . CLU
is sed in many human cancers (Yom et al., 2009; Kruger
et al., 2007; Zhang et al., 2006), including prostate, where
it increases following castration and to become highly
expressed in CRPC (July‘ et al., 2002). CLU’ over-expression
confers treatment resistance (Miyake et al., 2000), while CLU
-67—
tion potentiates activity of most anti-cancer therapies
in many preclinical models (Miyake et al., 2005; Sowery et
al., 2008; Gleave et al., 2005; Zoubeidi et al., 2010b). The
CLU inhibitor, OGX—Oll (custirsen, OncoGenex Pharmaceuticals),
is currently in Phase III trials after a randomized phase II
study in CRPC reported 7 Hwnth gain in overall survival and
50% reduced death rate (HR=O.50) when combined with docetaxel
chemotherapy (Chi et al., 2010).
Since CLU’ is induced. by treatment stress, including
castration, and functions as an important mediator of the
stress response, the esis herein that ARl treatment
induces the stress response and CLU, and that co—targeting the
AR and. stress—response pathways mediated‘ by‘ CLU’ may create
conditional lethality and improve cancer control was tested.
The data described herein set out to correlate ARI treatment
stress and resistance with CLU induction, identify ys
ting CLU activation, and define mechanisms by which CLU
inhibition iates anti~AR therapy in CRPC.
Many strategies used to kill cancer cells induce stress— and
redundant survival responses that promote survival and
emergence of treatment resistance, which. is the underlying
basis for' most ‘ . This therapeutic resistance
s from. a Darwinian interplay' of innate and adaptive
survival pathways activated by selective pressures of
treatment. In prostate cancer, androgen ablation induces tumor
cell sis and clinical responses in Hmst patients but
also triggers ssion within 2—3 years to castration
resistant prostate cancer (CRPC) (Gleave et al., 2001;
Bruchovsky et al., 2000; Goldenberg et al., 1999; Goldenberg
et al., 1996; Gleave et al., 1998). Experimentally, CRPC
W0 2012/123820
progression is attributedl to re—activation of the AR axis
(Miyake et al., 2000; Miyake et al., 1999) supported by growth
factor (Miyake et al., 2000; Culig et al., 2004; Craft et al.,
1999) and survival gene (Miyake et al., 1999; Gleave et al.,
1999; Miayake et al., 2000; Rocchi et al., 2004; Miyake et
al., 2000) networks. Recently new AR pathway inhibitors like
abiraterone and ARl (Rocchi et al., 2004) have been shown to
prolong survival and ally validate the AR as the main
driver of CRPC (Miyake et al., 2000; Miyake et al., 1999).
Not all patients respond to these inhibitors, and resistance
develops in many initial responders (Petrylak et al., 2004);
moreover, disease progression frequently correlates with a
rising PSA level, indicating continued AR ling and
highlighting need for onal therapies targeting the
lar basis of treatment resistance fill CRPC. Defining
interactions between the AR and redundant survival pathways
will build new combinatorial gies that control
progression and improve outcomes.
Persistent AR signalling in CRPC is ated to occur via AR
amplification and mutations that increase sensitivity to low
levels of BET and other steroids (Miyake et al., 2000; Miyake
et al., 1999; Zoubeidi et al., 2007), or AR splice variants
that drive constitutively active truncated receptors lacking a
LED (Nizard et al., 2007; Carver et al., 2011; Evdokimova et
al., 2006). Other AR-related, mechanisms include altered
levels of AR vators or co chaperones (hsp27), and AR
phosphorylation via activated src or tyrosine kinase receptors
like EGFR (Chi et al., 2010). Another more dynamic mechanism
involves reciprocal ck regulation n AR and PIBK
pathways whereby AR inhibition activates AKT signaling by
reducing levels of the AKT phosphatase PHLPP, and PIBK
inhibition activates AR signaling by relieving feedback
inhibition of HER kinases; inhibition of one activates the
other, thereby enhancing survival. These istic insights
are guiding design of combinatorial regimens co-targeting the
AR. pathway with inhibitors against histone deacetylase (de
Bono et al., 2011), src, ART, and. AR chaperone heat-shock
proteins (Hsp)—90 and Hsp27.
Inhibiting the stress se activated by AR pathway
inhibitors is another combinatorial co—targeting strategy.
Many anti—cancer agents induce ER stress (Rutkowski et al.,
2007), which activates a complex intracellular signaling
pathway, termed the unfolded protein response (UPR), tailored
to reestablish protein homeostasis by inhibiting protein
translation and stimulating the ubiquitin—proteasome system
(UPS) to enhance ER-associated protein degradation (BRAD)
(Harding et al., 2002). Chaperones like CLU are key ors
of ER stress responses. AR. pathway tion is known to
induce ER stress and CLU with reciprocal pathway activation of
AKT, which are all implicated in castration ance.
Consistent with these prior s, the data herein show that
ARl induces ER stress and the UPR, and go on to define feed—
forward. links between stress—induced YB—l activity to CLU
activation in parallel with enhanced AKT and MARK signaling,
which collectively support AR stability and ty under ARl
treatment conditions. YB—l and CLU are both stress—activated
survival chaperone proteins onally associated with anti—
cancer treatment resistance (Poon et al., 2002) (Zoubeidi et
al., 2007). Under stress conditions, YB-l is phospho-activated
by AKT (Evdokimova et al., 2006) and p90RSK e et al.,
2005), stimulating its nuclear translocation and binding to
target promoters. CLU is transcribed by, and acts as, a
W0 2012/123820
critical downstream mediator of stress—induced YB-l activity
and paclitaxel resistance (Shiota). YB-l can also function as
an mRNA chaperone protein to regulate translation of certain
stress-associated transcripts (Law et al., 2010; Evdokimova et
al., 2009). Using YBl RNA—1P ized to macroarrays with
different platform technologies, YB: was found to bind to CLU
mRNA. The data disclosed herein show that YBl binds
preferentially to CLU mRNA after ARl induced ER , and
found that YBl is associated with CLU—mRNA in different
polysomal fraction. Since polysomal fractions represent
translationally active mRNAs that are bound by ribosomes or
other elements of the translational machinery, and post—
polysomal mRNAs are me—depleted and hence
translationally inactive ; Evdokimova et al., 2009; Evdokimova
et al., 2006a), CLU mRNA will be amplified from these
fractions. These data indicate that YB-l mediates not only
transcriptional, but also ational, induction of CLU in
response to ARl induced ER stress.
CLU is a stress—activated molecular chaperone closely linked
to treatment resistance and cancer progression (Miyake et al.,
2000; Gleave and Miyake, 2005; Trougakos and Gonos, 2009b),
where its overexpression confers broad—spectrum treatment
resistance (Tran et al., 2009; Yom et al., 2009). Similar to
castration and other treatment ors, ARl increases CLU
expression ; moreover, CLU levels are higher in A21
ant tumors, as they are in CRPC compared to castrate
naive cancers. CLU is not only transcriptionally regulated by
HSF~1, but also enhances HSF-l-mediated transcriptional
activity in a orward manner (Lamoureax et al., 2011).
CLU is also activated by prosurvival pathways including the AR
and downstream of IL—6 (via JAR/stat) and IGF-lR (via Src—MEK—
W0 2012/123820
ERK—Erg—l) signalling pathways. CLU suppresses stress~induced
apoptosis by inhibiting protein aggregation, p53-activating
stress signals, and conformationally-altered. Bax (Zhang et
al., 2005; Trougakos et al., 2009) while ing Akt
orylation (Sowery et al., 2008; Chou et al., 1984) and
trans—activation of NF—KB and HSF-l.
This —activated anti—apoptotic function for CLU results
in broad-spectrunx resistance to many“ anti-cancer~ therapies,
and identifies it as a potential anti—cancer target. A CLU
antisense inhibitor, custirsen, enhances cancer cell death in
combination with therapeutic stressors in many nical
cancer models. Indeed, combination docetaxel plus custirsen
phase III clinical trials are underway in CRPC after
randomized Phase II studies reported a significant survival
benefit when sen was added to docetaxel (Zoubeidi et
al., 2010b; Culig et al., 2004). While CLU inhibition has been
reported to enhance castration and delay time to CRPC in
androgen—dependent xenografts, the pure AR antagonist ARl now
enables investigation of effects of AR pathway inhibition and
CLU in treatment response in vitro and in vivo in CRPC models.
The data disclosed. herein established. that CLU’ was induced
after ARl and AR knockdown in ARl—sensitive and resistant
LNCaP cells, tively, and that CLU remained highly
expressed. in most ARl ant LNCaP xenografts and cell
lines. Co-targeting the AR and CLU using ARl plus custirsen
enhanced apoptotic rates over monotherapy. istically,
ARl induced cross talk activation of ART and MAPK pathways was
repressed with combined therapy. Unexpectedly, we found that
AR ubiquitination and proteasome-mediated ation rates
were accelerated. when ARl was combined. with. CLU knockdown.
While ARl alone did. not alter AR stability, when CLU was
WO 23820
inhibited, stress—activation. of YB—l and MAPK was blunted,
resulting in decreased. YB-l activated expression of AR co-
chaperones Hsp56 (FKBP52) and Hsp90, which led to
ubiquitination and. proteasomal degradation. of the AR,
sing' AR. transcriptional activity' beyond. that observed
with AR1 monotherapy, and even in ARl resistant cell lines.
These results highlight a role for CLU in supporting' YB—l
mediated sion of other molecular chaperones under
context dependent stress ions, similar to its ability to
enhance HSF-l—mediated transactivation of Hsp70 and Hsp27
after Hsp90 inhibition (Lamoureax et al., 2011). In addition
to CLU, HSF—l and YB—l orchestrate expression of other
molecular chaperones involved in ses of folding,
trafficking, and riptional activation of the AR and
other steroid receptors. In the absence of ligand, AR is
inately cytoplasmic, maintained in an inactive, but
highly responsive state by a large dynamic heterocomplex
composed. of Hsp90 and. Hsp70, and. co-chaperones like Hsp56.
These Hsp AR co—chaperones play important roles in AR
stability and activation. g—Flynn et al., 2005; Yang et
al., 2006). Ligand binding leads to a conformational change in
the AR and dissociation from the large Hsp complex to
associate with Hsp27 for nuclear transport and transcriptional
activation of target genes (Zoubeidi et al., 2006; Abdul et
al., 2001). Compared to the first generation. AR. antagonist
bicalutamide, which does not inhibit AR nuclear transport, we
show that und AR remains cytoplasmic and complexed with
its Hsp chaperones, Hsp90 and FKBP52. This cytoplasmic
confinement of AR. complexed. with. its Hsp co—chaperones may
increase its susceptibility to degradation under conditions of
ER stress and chaperone suppression.
PCT/IBZOlZ/000609
Without g to be bound by any scientific theory, the data
herein identifies another mechanimn by which CLU inhibition
potentiates anti-AR therapy, via suppression of MAPK and Akt
signalling pathways activated after AR y inhibition. The
data herein m. previous reports that ARl induces Akt
phosphorylation, and also show that MAPK and p90rsk are
activated by ARl to mediate YB—l phosphorylation. The results
herein demonstrate that CLU knock down combined with ARl
abrogates both Akt and p90rsk activation.
Without g to be bound by any scientific theory, the data
herein define a stress—induced. feed-forward loop ing
ARl—induced YB—l transactivation of CLU, with CLU facilitating
pro—survival ART and p90rsk signalling, phospho-activation of
YB—l, and expression of AR co-chaperones that stabilize the AR
under conditions of ARl treatment. Co-targeting adaptive
stress pathways ted by AR pathway inhibitors, and
mediated through CLU, potentiate anti—AR activity by
decreasing AR expression levels and activity, as well as
ctivation of Akt and MAPK signaling ys induced by ARl.
These results provide mechanistic and pre-clinical proof—of—
principle to support combinatorial clinical s with ARl
and custirsen.
Aspects of the present invention relate to the unexpected
discovery that an ucleotide targeting clusterin
expression such as custirsen, together with an AR antagonist
as a combination is more potent than a monotherapy of either
agent for ent of prostate cancer. This increased
efficacy is in addition to increased cancer cell death, and
includes reduced proliferation of the cancer cells, reduced
W0 2012/123820
translocation of AR from the cytoplasm to the nucleus, d
riptional activity of AR, increased PARP cleavage,
reduced AKT phosphorylation, reduced ERK phosphorylation, and
increased. AR. protein degradation. Figure 30 shows that ARl
resistant prostate cancer tumors have increased clusterin
expression. Without wishing' to be bound. by' any scientific
theory, the data herein may be reflecting that the resistance
of these tumors to ARl may‘ be due to increased. clusterin
expression, and therefore, decreasing clusterin expression
increases the ivity of ARl ant tumors to ARl
treatment. Thus, ARl resistant prostate cancer cells are
ized to ARl by concomitant treatment with sen.
ARl s autophagy in prostate cancer cells (Fig. 38), and
clusterin silencing can inhibit ER stress—induced autophagy.
Autophagy is ea well conserved lysosomal degradation pathway
for intra—cellular ion that can confer stress tolerance
and sustain cell viability under adverse conditions. Without
g to be bound by any scientific theory, it is possible
that sed autophagy following ARl treatment may enhance
prostate cancer cell survival, and. inhibition of clusterin
expression inhibits this increased autophagy, thereby
resulting in reduced. cancer cell survival and. enhanced. ARl
activity.
Without wishing to be bound by any scientific theory,
decreased clusterin expression may enhance the activity of ARl
by decreasing AR stability via suppression of HSF—l mediated
regulation of AR co-chaperones such as FKBP52 and Hsp27.
Without wishing to be bound by any scientific theory,
decreased clusterin expression may enhance the activity of ARl
320121000609
by sing the induction of AKT levels and/or
phosphorylation ing ARl treatment.
ARl monotherapy is able to treat castration—resistant prostate
cancer in humans; r, custirsen monotherapy has not been
shown to inhibit the progression of prostate cancer after it
has progressed to androgen—independence in any model .
It is therefore surprising that the combination treatment of
ARl and custirsen would be more potent than treatment with AR:
alone. Furthermore, ARl and custirsen combination therapy is
surprisingly potent, and is able to halt prostate cancer cell
growth in vitro, whereas cells receiving either agent alone
erate by over 200% over a period of four days (Fig. 2B).
Surprisingly, also, the combination of custirsen and ARl is
able to reduce AR. protein expression. by over 80%, whereas
sen alone has no effect, and ARl alone reduces
expression by only about 40% (Figure 17). Finally, the
combination of custirsen and ARl increases the survival of
treated mice afflicted with castration—resistant prostate
cancer to about 90% at 16 weeks from the start of treatment,
as compared to about 40% for ARl alone.
Additionally, the combination of ARl and, custirsen. is more
effective at reducing tumor growth in mammals than the
combination of bicalutamide and custirsen. The combination of
ARl and custirsen is more effective at reducing tumor growth
in mammals than the combination of flutamide and custirsen.
The combination of ARl and, custirsen, is more effective at
reducing cancer cell proliferation than the combination of
bicalutamide and custirsen. The combination of ARl and
custirsen is more effective at reducing cancer cell
proliferation than the combination of flutamide and custirsen.
The combination of ARI and custirsen is more effective at
increasing cancer cell apoptosis than the ation of
bicalutamide and custirsen. The combination of ARI and
custirsen is more effective at increasing cancer cell
apoptosis than the combination of flutamide and custirsen.
In addition to increased anti—tumor potency, combination
y' may also allow dose reduction strategies to reduce
ty. For example, ARI is known to induce side effects
such as fatigue and has a maximum tolerated dose of 240mg/day
(Scher et al., 2010). However, the present invention discloses
that doses of AR; as low as g/day in combination with
custirsen are effective to decrease tumor size and. prolong
survival in mice. The NIH provides guidance on the conversion
of doses used in mouse studies to those riate for human
use based on Equivalent Surface Area Dosage Conversion s
(NIH Equivalent Surface Area Dosage Conversion Factors
Guidance, Posted August 2007: Freidreich et al., 1966).
According to the NIH conversion factor table, the lOmg/kg/day
dose described for use in mice herein is equivalent to .83
mg/kg/day in a 60kg human, equaling a dose of about 49.8mg/day
for a 60kg human, or about 83mg/day for a lOOkg human. These
doses are much lower than the dose of 240mg/kg recommended for
phase III trials in humans (Scher et al., 2009). Therefore, an
aspect of~ the invention provides a. combination of gnu anti—
clusterin oligonucleotide and an AR antagonist ive to
treat prostate cancer in which the amount of the AR antagonist
in the combination is less than the ive amount used in
monotherapy. The surprising potency* of combination therapy
comprising an oligonucleotide which reduces Clusterin levels
and an AR nist can be used to decrease doses of one or
WO 23820
both agents in humans, enabling therapeutic benefit with less
side effects.
-78—
References
Abdul, M. and. N. Hoosein, Inhibition. by anticonvulsants of
prostate-specific antigen and interleukin—6 secretion by
human prostate cancer cells. Anticancer Res, 2001.
21(3B): p. .
Ammar, H. and J.L. Closset, Clusterin activates survival
through the phosphatidylinositol 3—kinase/Akt pathway. J
Biol Chem, 2008. 283(19): p. 61.
Bruchovskyy N., et al., Characterization. of 5 a—reducatase
gene expression in stroma and lium of human
prostate. Journal of Steroid Biochemistry and Molecular
Biology, 1996. 59(5/6): p. 397—404.
Bruchovsky, N., et al., Intermittent androgen suppression for
prostate cancer: Canadian Prospective Trial and related
observations. Mol Urol, 2000. 4(3): p. 191—9; discussion
201.
Carthew, R.W., Gene silencing by -stranded RNA. Current
Opinion in Cell Biology; 2001, 13:244—248.
Carver, B.S., et al., Reciprocal feedback regulation of PI3K
and androgen receptor ing in PTEN—deficient
te cancer. Cancer Cell, 2011. 19(5): p. 575—86.
Chen, C.D., et al., Molecular determinants of resistance to
antiandrogen y. Nat Med, 2004. 10(1): p. 33~9.
Cheung~Flynn, J., et al., Physiological role for the
cochaperone FKBP52 in androgen receptor signaling. Mol
Endocrinol, 2005. 19(6): p. 1654—66.
Chi et al., A Phase I Pharmacokinetic and Pharmacodynamic
Study of custirsen, a 2’—Methoxyethyl antisense
Oligonucleotide to Clusterin, in Patients with zed
Prostate Cancer. Journal of the National Cancer
Institute; 2005, 97(17)1287—1296.
Chi et al., A phase I pharmacokinetic (PK) and pharmacodynamic
(PD) study of custirsen, a 2’methoxyethyl
phosphorothioate nse to clusterin, in patients with
prostate cancer prior to radical prostatectomy. Journal
of Clinical Oncology; 2004 ASCO Annual Meeting
Proceedings, vol. 22, no. 148:3033.
Chi, K.N., et al., Randomized phase II study of xel and
prednisone with or without OGX—Oll in patients with
metastatic castration—resistant prostate cancer. LI Clin
Oncol, 2010. 28(27): p. 4247—54.
Chou, T.C. and. P. Talalay, Quantitative is of dose-
effect relationships: the combined. s of multiple
drugs or enzyme inhibitors. Adv Enzyme Regul, 1984. 22:
p. 27~55.
Cochrane, D.R., et al., ential regulation of clusterin
and its isoforms by androgens in prostate cells. J Biol
Chem, 2007. 282(4): p. 2278—87.
Craft, N., et al., A mechanism for e—independent
prostate cancer h modulation of androgen receptor
signaling by the HER—2/neu tyrosine kinase. Nature
Medicine, 1999. 5(3): p. 280-5.
Culig, Z., Androgen receptor cross-talk with cell signalling
ys. Growth Factors, 2004. 22(3): p. 179—84.
de Bono, J.S., et al., Abiraterone and increased survival in
metastatic prostate cancer. N Engl J Med, 2011. 364(21):
p. 1995—2005.
Elbashir et al., es of 21-nucleotide RNAs mediate RNA
interference in cultured mammalian cells. Nature; 2001,
411:494—498.
Evdokimova, V., et al., Translational activation of snaill and
other developmentally regulated transcription factors by
W0 2012/123820
YB—l promotes an epithelial—mesenchymal transition.
Cancer Cell, 2009. 15(5): p. 402-15.
Evdokimova, V., et al., Akt—mediated YB-l phosphorylation
activates translation of silent mRNA species. Mol Cell
Biol, 2006. 26(1): p. 277—92.
Evdokimova, V., L.P. Ovchinnikov, and P.H. Sorensen, Y~box
binding n 1: providing a new angle on translational
regulation. Cell Cycle, 2006a. 5(11): p. 1143—7.
Fire et al., Potent and specific genetic interference by
double-stranded. RNA. in Caenorhabditis elegans. ;
1998, 391:806—11.
Freireich, et al., Quantitative comparison of toxicity of
ncer agents in mouse, rat, dog, monkey, and man.
Cancer Chemother Rep. 1966; 50(4):219—244.
Gleave, M.E., et al., Randomized comparative study of 3 versus
8-month neoadjuvant hormonal therapy before radical
prostatectomy: biochemical and. pathological effects. J
Urol, 2001. 166(2): p. 500-6; discussion 506-7.
Gleave, M., et al., Intermittent androgen ssion for
prostate cancer: ale and clinical experience.
Prostate Cancer Prostatic Dis, 1998. 1(6): p. 289—296.
Gleave, M. and H. Miyake, Use of antisense oligonucleotides
targeting the cytoprotective gene, clusterin, to enhance
en— and chemo~sensitivity in prostate cancer. World
J Urol, 2005. 23(1): p. 38—46.
Gleave, M. and K.N. Chi, Knock-down of the cytoprotective
gene, clusterin, to enhance e and chemosensitivity
in prostate and other cancers. Ann N Y Acad Sci, 2005.
1058: p. 1—15.
Gleave, M., et al., Progression to androgen ndence is
delayed by adjuvant treatment with nse Bc1—2
oligodeoxynucleotides after castration in the LNCaP
-81—
prostate tumor model. Clin Cancer Res, 1999. 5(10): p.
2891-8.
Goldenberg, S.L., et al., Clinical Experience with
Intermittent Androgen ssion in te Cancer:
Minimum of 3 Years' Follow-Up. Mol Urol, 1999. 3(3): p.
287—292.
Goldenberg, S.L., et al., Low dose cyproterone acetate plus
ose diethylstilbesterol — a protocol for reversible
medical tion. Urology, 1996. 47(6): p. 4.
Harding, H.P., et al., Transcriptional and translational
control in the Mammalian unfolded protein response. Annu
Rev Cell Dev Biol, 2002. 18: p. 575-99.
Harris, W.P., et al., Androgen deprivation therapy: progress
in tanding isms of resistance and optimizing
androgen depletion. Nat Clin Pract Urol, 2009. 6(2): p.
76-85.
July, L.V., et al., Clusterin expression is significantly
enhanced in prostate cancer cells following androgen
withdrawal therapy. Prostate, 2002. 50(3): p. 179—88.
Krajewska et al., Immunohistochemical analysis of bcl—2, baX,
bcl—X, and mcl—l expression in prostate cancers. Am. J.
Pathol; 1996, 148:1567—1576.
Kruger, S., et al., Prognostic icance of rin
immunoreactivity in breast cancer. Neoplasma, 2007.
54(1): p. 46—50.
Lamoureux, F., et al., Clusterin Inhibition using OGX-Oll
Synergistically Enhances Hsp90 inhibitor Activity by
Suppressing the Heat Shock Response in Castrate Resistant
Prostate Cancer. Cancer Res, 2011.
Lassi et al., Update on castrate—resistant prostate cancer:
2010. Current Opinion in Oncology; 2010, 22:263-267.
PCT/IBZOIZ/000609
Law, J.H., et al., Molecular decoy to the Y-box binding
protein-1 suppresses the growth of breast and prostate
cancer cells whilst sparing normal cell viability. PLoS
One. 5(9).
Miyaki et al., Antisense oligodeoxynucleotide therapy
targeting clusterin gene for te cancer: Vancouver
experience from discovery to clinic. International
Journal of Urology; 2005, 12: 785—794.
McDonnell et al., Expression of the proto-oncogene bcl-2 in
the prostate and its association with the emergence of
androgen~independent prostate cancer. Cancer Res; 1992,
52:6940—6944.
Miyaki et al., Antisense oligodeoxynucleotide y
ing clusterin gene for prostate cancer: Vancouver
experience from discovery to clinic. International
Journal of Urology; 2005, 12: 785-794.
Miyake, H., et al., Overexpression of n-like growth
factor binding protein—5 helps accelerate progression to
androgen—independence in the human prostate LNCaP tumor
model through activation of phosphatidylinositol 3'—
kinase pathway. Endocrinology, 2000. 141(6): p. 2257—65.
, H., A. Tolcher, and M.E. Gleave, Antisense Bcl~2
oligodeoxynucleotides inhibit progression to androgen—
independence after castration in the Shionogi tumor
model. Cancer Res, 1999. 59(16): p. 4030—4.
Miyake, H., et al., terone-repressed prostate e-2
is an antiapoptotic gene involved in progression to
androgen independence in prostate . Cancer Res,
2000. 60(1): p. 170-6.
Miyake, H., I. Hara, and M.E. Gleave, Antisense
oligodeoxynucleotide therapy targeting clusterin gene for
te cancer: Vancouver experience from discovery to
clinic. Int J Urol, 2005. 12(9): p. 785-94.
Miayake, H., A. Tolcher, and M.E. Gleave, Chemosensitization
and delayed androgen—independent recurrence of te
cancer with the use of antisense Bel—2
oligodeoxynucleotides. J Natl Cancer Inst, 2000. 92(1):
p. 34—41.
Montpetit, M.L., K.R. s, and M. Tenniswood, Androgen~
sed messages in the rat ventral prostate. Prostate,
1986. 8(1): p. 25—36.
NIH Equivalent e Area Dosage Conversion Factors
Guidance, Posted August 2007. Accessed from
web.ncifcrf.gov/rtp/lasp/intra/acuc/fred/guidelines/ACUC4
2EquivSurfAreaDosageConversion.pdf on January 14, 2011.
Nizard, P., et al., Stress—induced retrotranslocation of
clusterin/ApoJ into the cytosol. Traffic, 2007. 8(5): p.
554—65.
Oh et al., Management of e refractory prostate cancer:
current standards and. future prospects. J Urol; 1998,
160(4):1220—9.
Petrylak, D.P., et al., Docetaxel and estramustine compared
with mitoxantrone and sone for advanced refractory
prostate cancer. N Engl J Med, 2004. 351(15): p. 1513*20.
Poon, S., et al., Mildly acidic pH activates the extracellular
molecular chaperone clusterin. J Biol Chem, 2002.
277(42): p. 39532—40.
Raffo et al., Overexpression of bcl—2 protects prostate cancer
cells from apoptosis in vitro and confers resistance to
en depletion in vivo. Cancer Res; 1995 55(19):
4448—4445.
Rocchi, P., et al., Heat shock protein 27 increases after
androgen ablation and plays a cytoprotective role in
—84-
hormone-refractory prostate cancer. Cancer Res, 2004.
64(18): p. 6595-602.
ski, D.T. and R.J. Kaufman, That which does not kill me
makes me stronger: adapting to chronic ER stress. Trends
Biochem Sci, 2007. : p. 469~76.
Scher, H.I., et al., Antitumour ty of ARl in castration—
resistant prostate : a phase 1—2 study. Lancet,
2010. 375(9724): p. 1437—46.
Scher et. al., Antitumor activity (If MDV3100 ill a phaseI/II
study of castration—resistant te cancer (CRPC).
2009 ASCO Meeting, J Clin Oncol 27:158, 2009 (suppl;
abstr 5011).
Sensibar et al., Prevention of Cell Death Induced by Tumor
Necrosis Factor d in LNCaP Cells by Overexpression of
Sulfated Glycoprotein—2 (Clusterin). Cancer Research;
1995, 55: 2431—2437.
Shiota, M., et al., Clusterin is a critical downstream
mediator of stress~induced YB—l transactivation in
prostate cancer. Mol Cancer Res, 2011.
Siegel, R., et al., Cancer statistics, 2011: the impact of
eliminating socioeconomic and racial disparities on
premature cancer deaths. CA Cancer J Clin, 2011. 61(4):
p. 212—36.
Solit, D.B., H.I. Scher, and N. Rosen, Hsp90 as a therapeutic
target in prostate cancer. Semin Oncol, 2003. 30(5): p.
709*16.
Sowery, R.D., et al., Clusterin knockdown using the antisense
ucleotide OGX—Oll re—sensitizes docetaxel—
refractory prostate cancer PC—3 cells to chemotherapy.
BJU Int, 2008. 102(3): p. .
ord, A.L., et al., Y—box binding protein-1 serine 102 is
a downstream target of p90 ribosomal S6 kinase in basal-
PCT/m20121000609
like breast cancer cells. Breast Cancer Res, 2008. 10(6):
p. R99.
Tran, C., et al., Development of a second—generation
antiandrogen for treatment of advanced prostate cancer.
Science, 2009. 324(5928): p. 787-90.
kos, I.P., et al., Intracellular Clusterin ts
mitochondrial apoptosis by ssing p53—activating
stress signals and stabilizing the cytosolic Ku70-Bax
protein complex. Clin Cancer Res, 2009. 15(1): p. 48—59.
Wong et al., Molecular characterization of human TRPM—
2/clusterin, a gene associated with sperm maturation,
apoptosis and neurodegeneration. Eur. J. Biochem. 1994,
221 (3):917—925.
Yagoda et al., Cytotoxic chemotherapy for advanced hormone-
resistant prostate cancer. ; 1993, ?1 (Supp. 3):
10981109.
Yang, 2., et al., FK506-binding protein 52 is essential to
uterine reproductive physiology controlled by the
progesterone receptor’ A isofornu Mol Endocrinol, 2006.
(11): p. 2682~94.
Yom, C.K., et al., Clusterin overexpression and relapse~free
al in breast cancer. Anticancer Res, 2009. 29(10):
p. 3909—12.
Zhang, H., et al., Clusterin inhibits apoptosis by interacting
with activated Bax. Nat Cell Biol, 2005. 7(9): p. 909—15.
Zhang, S., et al., Clusterin expression and univariate
analysis of overall al in human breast .
Technol Cancer Res Treat, 2006. 5(6): p. 573—8.
di, A., et al., Clusterin facilitates COMMDl and I-
kappaB degradation to enhance NF-kappaB activity in
prostate cancer cells. Mol Cancer Res, 2010a. 8(1): p.
119—30.
~86-
Zoubeidi, A., K. Chi, and M. , Targeting the
cytoprotective chaperone, clusterin, for treatment of
advanced cancer. Clin Cancer Res, 2010b. 16(4): p. 1088—
Zoubeidi, A., et al., Cooperative interactions between
en receptor (AR) and hock protein 27
facilitate AR transcriptional activity. Cancer Res, 2007.
67(21): p. 10455—65.
Zoubeidi, A., et al., Hsp27 promotes insulin-like growth
factor—I survival signaling in prostate cancer via
p9ORsk—dependent phosphorylation and inactivation of BAD.
Cancer Res, 2010c. 70(6): p. 2307-17.
-87—
Claims (35)
1. Use of i) an oligonucleotide which reduces clusterin expression and ii) an androgen receptor antagonist having the structure or a pharmaceutically' acceptable salt thereof, in the manufacture of a medicament for treating a mammalian subject afflicted with prostate cancer.
2. The use of claim 1, wherein the cancer is en— ndent prostate cancer.
3. The use of claim 1, wherein the subject has been previously treated with androgen ablation y.
4. The use of any one of claims 1—3, n the amount of the ucleotide and the amount of the androgen receptor antagonist or a pharmaceutically acceptable salt thereof when taken together is more effective to treat the subject than when each agent is administered alone.
5. The use of any one of claims 1-4, wherein the amount of the oligonucleotide in combination with the amount of the androgen receptor antagonist or a pharmaceutically ~88- acceptable salt thereof is less than is ally effective when administered alone.
The use of any one of claims 1-5, wherein the amount of the androgen. receptor antagonist or' a pharmaceutically able salt thereof in combination with the amount of the oligonucleotide is less than is clinically effective when administered alone.
The use of any one of claims 1—6, wherein the amount of the oligonucleotide and the amount of the en receptor antagonist or a pharmaceutically able salt thereof when taken together is effective to reduce a clinical symptom of prostate cancer in the subject.
The use of any one of claims 1—7, wherein the mammalian subject is human.
The use of any one of claims 1—8, wherein the oligonucleotide is an antisense oligonucleotide.
10. The use of claim 9, wherein the antisense oligonucleotide spans either the translation initiation site or the termination site of clusterin—encoding mRNA.
ll. The use of claim 10, wherein the nse oligonucleotide comprises nucleotides in the sequence set forth in SEQ ID NOS: 1 to ll.
12. The use of claim 10, wherein the antisense oligonucleotide comprises nucleotides in the sequence set forth in SEQ ID NO: 3.
l3. The use of claim 11 or 12, wherein the antisense oligonucleotide is modified to enhance in vivo stability relative to an 'unmodified. oligonucleotide of the same 8equence .
14. The use of claim 13, wherein the ucleotide is custirsen.
15. The use of claim 14, wherein the amount of custirsen is less than 640mg.
l6. The use of claim 15, wherein the amount of custirsen is less than 480mg.
l7. The use of any one of claims 14—16, wherein the amount of custirsen is ated for administration intravenously once in a seven day period.
l8. The use of any one of claims l—l7, wherein the amount of the en or antagonist is less than 240mg.
19. The use of any one of claims 1—17, wherein the amount of the androgen receptor antagonist is from 150mg to 240mg.
20. The use of any one of claims 1—18, wherein the amount of the androgen receptor antagonist is from 30mg to 150mg.
21. The use of any one of claims 1—18, wherein the amount of the androgen receptor antagonist is 80mg.
22. The use of any one of claims l-21, wherein the amount of the androgen receptor antagonist is formulated for administration orally once per day.
23. Use of i) an oligonucleotide which reduces clusterin expression and ii) an en receptor antagonist, in the manufacture of a medicament for the treatment of a mammalian subject afflicted with androgen—independent prostate cancer.
24. The use of claim 23, wherein the androgen receptor antagonist is a non—steroidal antiandrogen.
25. The use of claim 23 or 24, wherein the androgen or antagonist is ARl.
26. The use of any one of claims 1—25, wherein the combination of the oligonucleotide and the androgen receptor antagonist is effective to decrease androgen or translocation from the cytoplasm to the s of the tumor cells.
27. The use of any one of claims 1—25, wherein the combination of the oligonucleotide and the androgen receptor antagonist is ive to increase the proteasome degradation of the androgen receptor protein in the tumor cells.
28. The use of any one of claims 1-25, wherein the combination of the oligonucleotide and the androgen receptor antagonist is effective to decrease androgen receptor transcriptional activity in the tumor cells.
29. The use of any one of claims 1—25, wherein the ation of the oligonucleotide and the androgen receptor nist is ive to decrease the amount of phosphorylated AKT in the tumor cells.
30. The use of any one of claims 1—25, wherein the combination of the oligonucleotide and the androgen receptor antagonist is effective to decrease the amount of phosphorylated ERK in the tumor cells.
31. The use of any one of claims 1—25, wherein the combination of the oligonucleotide and the androgen receptor antagonist is effective to inhibit the proliferation of te cancer cells.
32. Use of custirsen in the manufacture of a medicament for increasing the ivity of ARl resistant prostate cancer cells to ARl.
33. A ition for treating a mammalian subject afflicted with prostate cancer comprising i) an oligonucleotide which reduces clusterin expression and ii) an androgen receptor antagonist having the structure FaC H or a pharmaceutically acceptable salt thereof.
34. A composition according to claim 33, wherein the prostate cancer is androgen independent prostate cancer.
35. The use of any one of claims 1, 23 or 32, ntially as herein. described. with. reference to any one of the Examples and/or
Applications Claiming Priority (9)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161452583P | 2011-03-14 | 2011-03-14 | |
US61/452,583 | 2011-03-14 | ||
US201161453309P | 2011-03-16 | 2011-03-16 | |
US61/453,309 | 2011-03-16 | ||
US201161453885P | 2011-03-17 | 2011-03-17 | |
US61/453,885 | 2011-03-17 | ||
US201161493336P | 2011-06-03 | 2011-06-03 | |
US61/493,336 | 2011-06-03 | ||
PCT/IB2012/000609 WO2012123820A1 (en) | 2011-03-14 | 2012-03-14 | Combination of anti-clusterin oligonucleotide with androgen receptor antagonist for the treatment of prostate cancer |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ616465A NZ616465A (en) | 2015-08-28 |
NZ616465B2 true NZ616465B2 (en) | 2015-12-01 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2012228007B2 (en) | Combination of anti-clusterin oligonucleotide with androgen receptor antagonist for the treatment of prostate cancer | |
EP3062808B1 (en) | Treatment of metastatic prostate cancer | |
Zoubeidi et al. | Targeting the cytoprotective chaperone, clusterin, for treatment of advanced cancer | |
Yamamoto et al. | siRNA lipid nanoparticle potently silences clusterin and delays progression when combined with androgen receptor cotargeting in enzalutamide-resistant prostate cancer | |
Ischia et al. | The promise of heat shock protein inhibitors in the treatment of castration resistant prostate cancer | |
US20170145418A1 (en) | Combination of Anti-Clusterin Oligonucleotide with HSP90 Inhibitor for the Treatment of Prostate Cancer | |
US20240041881A1 (en) | Compositions and methods for treating cancer | |
De Velasco et al. | Targeting castration-resistant prostate cancer with androgen receptor antisense oligonucleotide therapy | |
Olearczyk et al. | Targeting of hepatic angiotensinogen using chemically modified siRNAs results in significant and sustained blood pressure lowering in a rat model of hypertension | |
EP1545561B1 (en) | Oligonucleotides for treatment of prostate and other cancers | |
WO2018052891A2 (en) | Modulation of pcsk9 and ldlr through drp1 inhibition | |
Ha et al. | Targeting GRP78 suppresses oncogenic KRAS protein expression and reduces viability of cancer cells bearing various KRAS mutations | |
US20180153850A1 (en) | Compositions and methods for treatment of cancer | |
US8722872B2 (en) | Compositions and methods for treatment of prostate and other cancers | |
NZ616465B2 (en) | Combination of anti-clusterin oligonucleotide with androgen receptor antagonist for the treatment of prostate cancer | |
Zoubeidi et al. | Clusterin as a Target for Treatment of Castration-Resistant Prostate Cancer | |
NZ616474B2 (en) | Combination of anti-clusterin oligonucleotide with hsp90 inhibitor for the treatment of prostate cancer | |
Graff et al. | 148 Therapeutic targeting of the pro-survival transcription factor CREB sensitizes glioblastomas to temozolomide-based therapy |